CN111763215B - Compound with nitrogen-containing heterocyclic structure and preparation method and application thereof - Google Patents

Compound with nitrogen-containing heterocyclic structure and preparation method and application thereof Download PDF

Info

Publication number
CN111763215B
CN111763215B CN202010705735.XA CN202010705735A CN111763215B CN 111763215 B CN111763215 B CN 111763215B CN 202010705735 A CN202010705735 A CN 202010705735A CN 111763215 B CN111763215 B CN 111763215B
Authority
CN
China
Prior art keywords
compound
egfr
dichloro
reaction
fluorophenyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202010705735.XA
Other languages
Chinese (zh)
Other versions
CN111763215A (en
Inventor
刘冠锋
杨茂
李红波
张俊
刘龙飞
拓方彬
原晨光
李英富
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chengdu Haibowei Pharmaceutical Co ltd
Original Assignee
Chengdu Haibowei Pharmaceutical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chengdu Haibowei Pharmaceutical Co ltd filed Critical Chengdu Haibowei Pharmaceutical Co ltd
Priority to CN202010705735.XA priority Critical patent/CN111763215B/en
Publication of CN111763215A publication Critical patent/CN111763215A/en
Application granted granted Critical
Publication of CN111763215B publication Critical patent/CN111763215B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention discloses a compound with a nitrogen-containing heterocyclic structure, a preparation method and application thereof, wherein the compound has an obvious inhibiting effect on an epidermal growth factor receptor inhibitor (EGFR), can be used as the EGFR inhibitor and used for preparing medicaments for treating EGFR-mediated diseases, and the EGFR-mediated diseases comprise but are not limited to malignant tumors and the like, so that the compound has a wide application prospect.

Description

Compound with nitrogen-containing heterocyclic structure and preparation method and application thereof
Technical Field
The invention relates to the technical field of pharmaceutical chemistry, in particular to a compound with a nitrogen-containing heterocyclic structure, and a preparation method and application thereof.
Background
EGFR is one of the epidermal growth factor receptor (HER) family members, and EGFR signaling pathways play important roles in physiological processes such as growth, proliferation, and differentiation of cells. Studies have shown that there is high or abnormal expression of EGFR in many solid tumors. EGFR is involved in the inhibition of tumor cell proliferation, angiogenesis, tumor invasion, metastasis and apoptosis.
Studies have shown that the epidermal growth factor receptor EGFR is capable of activating intracellular downstream signaling pathways to promote cell division and proliferation, mainly in lung adenocarcinoma, asian, non-smoking and female patients, with mutations in the EGFR gene in about 15% of caucasians and 30-50% of asian. The EFGR Activity Mutation (AMs) is a common driver of non-small cell lung cancer, which occurs mainly in the 18/19/20/21 exon of the EGFR gene, resulting in activation of EGFR even in the absence of ligand, so that downstream signaling pathways continue to be hyperactivated, leading to malignant proliferation of cells (tumors), and thus, inhibition of abnormal EGFR signaling pathways is one of the strategies for treating tumors. The mutation rate of people without smoking history is as high as 50-60%, the common mutation sites are exons 19 and 21, account for 90%, and are called typical mutation, and the rest 10% are the mutations of exons 18 and 20. EGFR exon 20 insertion mutation is often resistant to EGFR-TKI and has a poor prognosis. However, there are no approved targeting drugs and the standard therapy remains traditional cytotoxic chemotherapy.
First-generation EGFR inhibitors, such as gefitinib and erlotinib, have clinically significant drug response in about 70% of patients, which is manifested by significant tumor shrinkage, and thus EGFR is a very effective target, but after 10-16 months of treatment, drug resistance occurs, and patients with nearly 2/3 develop secondary acquired variation T790M.
By analyzing the cause of drug resistance, a new generation of EGFR inhibitors, also called second generation EGFR-TKI, is developed on the basis of gefitinib/erlotinib. Unlike the first generation, the second generation compounds mostly adopt irreversible covalent bonding, and compared with successful formulas such as Afatinib and Dacomitinib, the second generation compounds are approved by FDA for the treatment of double-mutation non-small cell lung cancer. Although second generation drugs can overcome the resistance problem, these compounds have serious dose-dependent side effects such as rash and diarrhea, thus greatly limiting their clinical use.
After the third-generation EGFR inhibitor is developed, the EGFR-T790M drug resistance mutation problem is finally solved. The first clinical entry of the international third-generation EGFR is 2013, and by now, a plurality of third-generation EGFR small-molecule inhibitors enter the clinical stage globally. Currently, the global sales of the marketed drug oxitinib mesylate in 2015 to 2019 are respectively 0.18 million dollar, 4.23 million dollars, 9.55 million dollars, 18.6 million dollars and 31.89 million dollars, which are far higher than the first two generations of EGFR inhibitors and have great market prospects.
Although good, the Oxitinib still inevitably causes the drug resistance problem of C797S (namely cis-form or trans-form three mutations including del19/T790M/C797S or L858R/T790M/C797S) during the use process, and has no capability of 20ins mutation. For 20ins mutation, TAK-788 and Bocintinib have stronger activity. (Poziotinib), a targeted drug specifically directed against EGFR/HER2 exon 20 insertion mutation, was hoped that the objective remission rate for treatment of EGFR exon 20 insertion mutation in previous small-scale clinical studies exceeded 40%, but declined as the sample size expanded. Therefore, the development of new EGFR inhibitors is of great interest.
Disclosure of Invention
The invention mainly solves the technical problem of providing a compound which can effectively inhibit an epidermal growth factor receptor inhibitor EGFR.
In order to solve the technical problems, the invention adopts a technical scheme that:
providing a compound having the structure shown in formula I or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, a pharmaceutically acceptable hydrate, solvate, or salt thereof:
Figure BDA0002594682410000021
wherein:
a is selected from C or N;
y is selected from substituted or unsubstituted N-containing monocyclic group, substituted or unsubstituted N-containing spiro group, substituted or unsubstituted N-containing bridged group, substituted or unsubstituted N-containing fused ring group, and N atom is bonded to the carbonyl end;
b is selected from O or NR4,R4Selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl;
R1independently at each occurrence, selected from the group consisting of hydrogen, halogen, hydroxy, cyano, amino, ester, acyl, amide, sulfonyl, phosphoryl, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl;
“R1independently selected for each occurrence "means when defining R1When the number n is greater than 1, different R1May be selected from the same or different groups. For example, n is 2, one R1May be selected from substituted or unsubstituted alkyl, another R1May be selected from halogens; or, n is 2, two R1May each be selected from substituted or unsubstituted alkyl groups; the same applies to the rest of similar cases.
R2Selected from substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl;
R3selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl;
Y、R1、R2、R3、R4wherein said substituted substituent is selected from the group consisting of deuterium, halogen, hydroxy, cyano, amino, carboxy, alkyl, cycloalkyl, heteroalkyl, heterocycloalkyl, aryl, heteroaryl, ester, acyl, amide, sulfonyl, phosphoryl;
n is selected from 0, 1,2,3,4 and 5;
when A is N and R2When it is methyl, Y is not
Figure BDA0002594682410000031
Further, Y is selected from the following substituted or unsubstituted groups:
Figure BDA0002594682410000032
when A is N and R2When it is methyl, Y is not
Figure BDA0002594682410000033
Further, Y is selected from the following substituted or unsubstituted groups:
Figure BDA0002594682410000034
when A is N and R2When it is methyl, Y is not
Figure BDA0002594682410000035
Further, Y is selected from
Figure BDA0002594682410000036
In a particular embodiment of the invention, R1Selected from hydrogen, halogen, substituted or unsubstituted C1-C6 alkyl;
further, R1Is selected from halogen, n is selected from 1,2 and 3; further, R1Is selected from F, Cl, and n is selected from 2 and 3.
Further, the compound has a structure shown in formula II or a tautomer, mesomer, racemate, enantiomer, diastereomer or mixture form, pharmaceutically acceptable hydrate, solvate or salt thereof:
Figure BDA0002594682410000041
in a particular embodiment of the invention, B is selected fromO or NR4,R4Selected from H, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted 2-6-membered N heteroalkyl, wherein the substituent is selected from C1-C3 alkyl and halogen;
further, R4Selected from H, substituted or unsubstituted 2-4-membered N heteroalkyl, wherein the substituent is selected from C1-C3 alkyl;
further, B is selected from O, NH, NCH2CH2N(CH3)2Preferably O, NH.
In a particular embodiment of the invention, R2Is selected from substituted or unsubstituted C1-C3 alkyl, wherein, the substituent is selected from deuterium and halogen;
further, R2Selected from methyl and CD3、CHF2Preferably methyl.
In a particular embodiment of the invention, R3Selected from H, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted 2-6-membered N heteroalkyl, wherein the substituent is selected from C1-C3 alkyl and halogen;
further, R3Selected from H, CH2N(CH3)2Preferably H.
In the present invention, the term "2-to 4-membered N heteroalkyl" means that the number of main chain skeleton atoms of the heteroalkyl is 2 to 4, and the same holds true for the rest of the similar cases.
In a particular embodiment of the invention, the compound structure is selected from one of the following:
Figure BDA0002594682410000042
Figure BDA0002594682410000051
the invention also provides a medicinal composition, and the active ingredients of the medicinal composition are selected from one or the combination of more than two of the compounds or the stereoisomer, the solvate, the hydrate, the pharmaceutically acceptable salt or the eutectic crystal thereof.
The invention also provides the application of the compound or the stereoisomer, the solvate, the hydrate, the pharmaceutically acceptable salt or the eutectic crystal thereof in preparing the epidermal growth factor receptor inhibitor; further, the epidermal growth factor receptor inhibitor is an EGFR inhibitor.
The invention also provides the use of the compound or the stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or eutectic crystal thereof in the preparation of medicaments for treating diseases causing EGFR overexpression.
The invention also provides application of the compound or stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or eutectic crystal thereof in preparing a medicament for treating diseases caused by EGFR overexpression.
The invention also provides the use of the above compound or a stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or co-crystal thereof in the preparation of a medicament for the treatment of any one or more of cancer.
The invention also provides a preparation method of the compound, which comprises the following steps:
Figure BDA0002594682410000061
(1) reacting the compound c with the compound d under the action of alkali to obtain a compound e;
further, the base may be selected from sodium carbonate, potassium carbonate, and the like, and in a specific embodiment of the present invention, the base is potassium carbonate.
(2) Removing the Boc protective group from the compound e to generate a compound f;
the removal of the Boc protective group is a conventional reaction in the field of organic synthesis, and all methods for realizing the removal of the Boc group in the field can be applied to the invention; in a specific embodiment of the invention, the Boc group is removed by trifluoroacetic acid.
(3) And reacting the compound f with the compound g under a basic condition to generate a compound h.
The base added in step (3) may be sodium carbonate, potassium carbonate, etc., and in a specific embodiment of the present invention, the base added in step (3) is sodium carbonate, and the solvent is a mixed solvent of THF/water in a volume ratio of 5/1.
The invention also provides another preparation method of the compound, which comprises the following steps:
Figure BDA0002594682410000062
(1) reacting the compound i with the compound b under an acidic condition to generate a compound j;
in a particular embodiment of the invention, compound i is reacted with compound b by addition of hydrochloric acid.
(2) Carrying out cross coupling reaction on halogen in the compound j and amino in the compound k to generate a compound l;
there are many methods available in the art for reacting halogenated aromatic hydrocarbons with amines and are suitable for use in the present invention, such as Buchwald coupling.
(3) Removing the Boc protective group from the compound l to generate a compound m;
the removal of the Boc protective group is a conventional reaction in the field of organic synthesis, and all methods for realizing the removal of the Boc group in the field can be applied to the invention; in a specific embodiment of the invention, the Boc group is removed by trifluoroacetic acid.
(4) And (3) reacting the compound m with the compound g under a basic condition to generate a compound n.
The base added in step (4) may be sodium carbonate, potassium carbonate, etc., and in a specific embodiment of the present invention, the base added in step (4) is sodium carbonate, and the solvent is a mixed solvent of THF/water in a volume ratio of 5/1.
The invention also provides a preparation method of the compound, which comprises the following steps:
Figure BDA0002594682410000071
R2is not methyl;
(1) removing methyl from the compound c to obtain a compound o;
the removal of the methyl group on the methoxy group is a conventional reaction in the field of organic synthesis, and all methods capable of realizing the removal of the methyl group on the methoxy group in the field can be applied to the method; in the specific embodiment of the invention, the compound c is mixed with pyridine hydrochloride, and the methyl is removed under the condition of raising the temperature to about 170 ℃.
(2) Reacting the compound o with the compound d under the action of alkali to generate a compound p;
further, the base may be selected from sodium carbonate, potassium carbonate, and the like, and in a specific embodiment of the present invention, the base is potassium carbonate.
(3) Reacting the compound p with CD3I or 2-chloro-2, 2-difluoro-1-phenylethane-1-ketone under the action of alkali to generate a compound q;
further, the base may be selected from sodium carbonate, potassium carbonate, and the like, and in a specific embodiment of the present invention, the base is potassium carbonate.
(4) Removing the Boc protecting group from the compound q to generate a compound r;
the removal of the Boc protective group is a conventional reaction in the field of organic synthesis, and all methods for realizing the removal of the Boc group in the field can be applied to the invention; in a specific embodiment of the invention, the Boc group is removed by trifluoroacetic acid.
(5) And (3) reacting the compound r with the compound g under a basic condition to generate a compound s.
The base added in step (5) may be sodium carbonate, potassium carbonate, etc., and in a specific embodiment of the present invention, the base added in step (5) is sodium carbonate, and the solvent is a mixed solvent of THF/water in a volume ratio of 5/1.
The pharmaceutical composition containing the compound of the invention or the stereoisomer, solvate, hydrate, pharmaceutically acceptable salt or cocrystal thereof can contain pharmaceutically acceptable auxiliary materials.
As used herein, "pharmaceutically acceptable" is meant to include any material that does not interfere with the effectiveness of the biological activity of the active ingredient and is not toxic to the host to which it is administered.
The pharmaceutically acceptable auxiliary materials are general names of all the additional materials except the main medicine in the medicine, and the auxiliary materials have the following properties: (1) no toxic effect on human body and few side effects; (2) the chemical property is stable and is not easily influenced by temperature, pH, storage time and the like; (3) has no incompatibility with the main drug, and does not influence the curative effect and quality inspection of the main drug; (4) does not interact with the packaging material. The auxiliary materials in the invention include, but are not limited to, a filler (diluent), a lubricant (glidant or anti-adhesion agent), a dispersing agent, a wetting agent, an adhesive, a regulator, a solubilizer, an antioxidant, a bacteriostatic agent, an emulsifier, a disintegrating agent and the like. The binder comprises syrup, acacia, gelatin, sorbitol, tragacanth, cellulose and its derivatives (such as microcrystalline cellulose, sodium carboxymethylcellulose, ethyl cellulose or hydroxypropyl methylcellulose), gelatin slurry, syrup, starch slurry or polyvinylpyrrolidone; the filler comprises lactose, sugar powder, dextrin, starch and its derivatives, cellulose and its derivatives, inorganic calcium salt (such as calcium sulfate, calcium phosphate, calcium hydrogen phosphate, precipitated calcium carbonate, etc.), sorbitol or glycine, etc.; the lubricant comprises superfine silica gel powder, magnesium stearate, talcum powder, aluminum hydroxide, boric acid, hydrogenated vegetable oil, polyethylene glycol and the like; the disintegrating agent comprises starch and its derivatives (such as sodium carboxymethyl starch, sodium starch glycolate, pregelatinized starch, modified starch, hydroxypropyl starch, corn starch, etc.), polyvinylpyrrolidone or microcrystalline cellulose, etc.; the wetting agent comprises sodium lauryl sulfate, water or alcohol, etc.; the antioxidant comprises sodium sulfite, sodium bisulfite, sodium pyrosulfite, dibutylbenzoic acid, etc.; the bacteriostatic agent comprises 0.5% of phenol, 0.3% of cresol, 0.5% of chlorobutanol and the like; the regulator comprises hydrochloric acid, citric acid, potassium (sodium) hydroxide, sodium citrate, and buffer (including sodium dihydrogen phosphate and disodium hydrogen phosphate); the emulsifier comprises polysorbate-80, sorbitan fatty acid, pluronic F-68, lecithin, soybean lecithin, etc.; the solubilizer comprises Tween-80, bile, glycerol, etc. The term "pharmaceutically acceptable salt" refers to a salt of a compound of the present invention with an acid or base that is suitable for use as a pharmaceutical. The acid base is a generalized Lewis acid base. Suitable acids for forming the salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, etc., organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, phenylmethanesulfonic acid, benzenesulfonic acid, etc.; and acidic amino acids such as aspartic acid and glutamic acid.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, parenteral (intravenous, intramuscular, or subcutaneous), and topical administration.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers, e.g., ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, and oils, especially cottonseed, groundnut, corn germ, olive, castor and sesame oils, or mixtures of such materials, and the like.
In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms for topical administration of the compounds of the present invention include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
The compounds of the invention can likewise be used in injectable preparations. Wherein the injection is selected from liquid injection (water injection), sterile powder for injection (powder injection) or tablet for injection (refers to impression tablet or machine pressing tablet prepared by aseptic operation method of medicine, and is dissolved with water for injection for subcutaneous or intramuscular injection when in use).
Wherein the powder for injection contains at least an excipient in addition to the above compound. The excipients, which are components intentionally added to a drug in the present invention, should not have pharmacological properties in the amounts used, however, the excipients may aid in the processing, dissolution or dissolution of the drug, delivery by a targeted route of administration, or stability.
"substituted" means that a hydrogen atom in a molecule is replaced by a different atom or molecule.
"alkyl" refers to an aliphatic hydrocarbon group and to a saturated hydrocarbon group. The alkyl moiety may be a straight chain or branched chain alkyl. Typical alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, hexyl, and the like.
The C1-Cn used in the invention comprises C1-C2 and C1-C3 … … -C1-Cn, wherein n is an integer more than one; the prefix as a substituent denotes the minimum and maximum number of carbon atoms in the substituent, e.g., "C1-C6 alkyl" means a straight or branched chain alkyl group containing one to 6 carbon atoms.
"heteroalkyl" refers to an alkyl group containing one or more heteroatoms such as N, O, S, P, B.
"amido" is a chemical structure having the formula-C (O) NHR or-NHC (O) R, wherein R can be selected from alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, and the like.
"Sulfonyl" is a compound having the formula-S (═ O)2The chemical structure of R, including sulfonamide, wherein R can be selected from alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, amino, and the like;
"phosphoryl" is a chemical structure having the formula-P (═ O) RR ', where R, R' may each be independently selected from alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, hydroxy, amino, and the like;
"ester group" means a chemical structure having the formula-COOR, wherein R is selected from alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl.
"cycloalkyl" refers to a saturated or unsaturated cyclic hydrocarbon substituent such as "C3-C6 cycloalkyl" refers to a cycloalkyl group having 3-6 carbon atoms in the ring backbone structure.
"Heterocycloalkyl" refers to a cycloalkyl group containing at least one heteroatom in the ring backbone.
Heteroatoms include, but are not limited to O, S, N, P, Si and the like.
"Ring" refers to any covalently closed structure, including, for example, carbocycles (e.g., aryl or cycloalkyl), heterocycles (e.g., heteroaryl or heterocycloalkyl), aryls (e.g., aryl or heteroaryl), nonaromatic (e.g., cycloalkyl or heterocycloalkyl). The "ring" in the present invention may be a monocyclic ring or a polycyclic ring, and may be a fused ring, a spiro ring or a bridged ring.
Typical heterocycloalkyl groups include, but are not limited to:
Figure BDA0002594682410000101
"aryl" means a planar ring having a delocalized pi-electron system and containing 4n +2 pi electrons, where n is an integer. The aryl ring may be composed of five, six, seven, eight, nine or more than nine atoms. Aryl groups include, but are not limited to, phenyl, naphthyl, phenanthryl, anthracyl, fluorenyl, indenyl, and the like.
Typical heteroaryl groups include, but are not limited to:
Figure BDA0002594682410000102
"halogen" or "halo" refers to fluorine, chlorine, bromine or iodine.
The alkyl, heteroalkyl, cyclic, heterocyclic, amino, ester, carbonyl, amide, sulfonyl, phosphoryl, boronic acid, boronic ester, boronic acid ester, guanidino, acyl guanidino, aryl, heteroaryl, and the like, as described herein, may be unsubstituted alkyl, heteroalkyl, cyclic, heterocyclic, amino, ester, carbonyl, amide, sulfonyl, phosphoryl, boronic acid, boronic ester, guanidino, acyl guanidino, aryl, heteroaryl, and may be substituted alkyl, heteroalkyl, cyclic, heterocyclic, amino, ester, carbonyl, amide, sulfonyl, phosphoryl, boronic acid, boronic ester, guanidino, acyl guanidino, aryl, heteroaryl.
Hereinbefore, unless already indicated, the term "substituted" means that the mentioned groups may be substituted by one or more additional groups each and independently selected from alkyl, cycloalkyl, aryl, carboxy, heteroaryl, heterocycloalkyl, hydroxy, alkoxy, alkylthio, aryloxy, nitro, acyl, halogen, haloalkyl, amino and the like.
The invention has the beneficial effects that:
(1) the invention provides a series of compounds with EGFR (epidermal growth factor receptor) inhibiting activity, tests show that the compounds have obvious inhibiting effect on EGFR (epidermal growth factor receptor), provide a new scheme for treating diseases taking EGFR as a treatment target, such as malignant tumor diseases and the like, can be used for preparing medicaments for treating related diseases, and have wide application prospect.
(2) Compared with the bociclib, the compound has better or equivalent activity to a protein kinase inhibitor, better stability of liver microsomes and higher development value.
Detailed Description
The technical solutions of the present invention are described clearly and completely below, and it is obvious that the described embodiments are some, not all embodiments of the present invention. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
In the present invention, the structure of the compound is determined by a Mass Spectrometry (MS) or nuclear magnetic resonance (1HNMR) apparatus. The term "room temperature" means between 10 ℃ and 25 ℃. The abbreviations have the following meanings:
NMP: n-methyl pyrrolidone;
EA: ethyl acetate;
THF: tetrahydrofuran;
DMA: n, N-dimethylacetamide;
DMF: n, N-dimethylformamide;
TLC: thin layer chromatography;
PE refers to petroleum ether (with a boiling point of 60-90 ℃);
DCM: dichloromethane;
H2o: distilled water;
DMSO is dimethyl sulfoxide;
TFA: trifluoroacetic acid;
Pd2(dba)3: 3,3,6, 6-tetramethyl-9- (1,2,3, 4-tetrahydroxybutyl) -4,5,7, 9-tetrahydro-2H-heteroanthracene-1, 8-dione);
x-phos: 2-dicyclohexylphosphonium-2, 4, 6-triisopropylbiphenyl;
1, 4-dioxane: 1, 4-dioxane.
Preparation of intermediates
Intermediate 1: preparation of 4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-ol
Figure BDA0002594682410000121
Step 1: preparation of 4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazoline-6-acetate
The compound 4-chloro-7-methoxyquinazoline-6-acetate (4.00g, 15.87mmol) was dissolved in NMP (60mL), 3, 4-dichloro-2-fluoroaniline (2.84g, 15.87mmol) was added thereto, and the mixture was stirred at 110 ℃ for 4 hours. And (5) detecting by TLC spot plates, and cooling the reaction liquid to room temperature after the raw materials are reacted. Water (600mL) was added to the reaction solution, and extraction was performed 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. Na (Na)2SO4After drying, the crude product is directly used in the next reaction.
EM (calculated): 395.0; MS (ESI) M/z (M + H) +: 396.0
Step 2: preparation of 4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-ol the crude 4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-acetate was dispersed in acetonitrile (100mL) and the system was in an off-white suspension. Ammonia (50mL, 28% in H) was added dropwise to the system2O), stirring at room temperature for 40 minutesA clock. The reaction solution was filtered, and the filter cake was washed with a small amount of water and acetonitrile in this order and dried to obtain the objective compound (4.90g, yield in two steps: 87.5%) as an off-white solid.
EM (calculated): 353.0, respectively; MS (ESI) M/z (M + H)+:354.0
Intermediate 2: preparation of 4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-ol
Figure BDA0002594682410000122
Step 1: preparation of 6- (benzyloxy) -N- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinolin-4-amine
The compound, 6- (benzyloxy) -4-chloro-7-methoxyquinoline (5.00g, 16.72mmol), was dissolved in NMP (50mL), to which was added 3, 4-dichloro-2-fluoroaniline (2.99g, 16.72mmol), dissolved, concentrated hydrochloric acid (10mL) was added, and the mixture was stirred at 110 ℃ overnight. After completion of the reaction, the reaction solution was cooled to room temperature, stirred under an ice-water bath and acetonitrile (50mL) was slowly added thereto. A large amount of solid was precipitated, collected by filtration, rinsed with a small amount of acetonitrile, and dried to give the objective compound (4.46g, yield 60.3%) as a white solid.
EM (calculated): 442.1; MS (ESI) M/z (M + H)+:443.2
Step 2: preparation of 4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-ol
The compound 6- (benzyloxy) -N- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinolin-4-amine (4.40g, 9.95mmol) was dissolved in THF/MeOH (44mL,10/1), to which was added Pd/C (440mg, 10% W/W), and stirred at room temperature for 3 hours. After the reaction, the reaction solution was filtered, and the filter cake was washed with a small amount of methanol. The filtrate was concentrated to dryness to give the objective compound (3.12g, yield 89.2%) as a brown solid.
EM (calculated): 352.0; MS (ESI) M/z (M + H)+:353.1
Intermediate 3: preparation of 6-bromo-N- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinazolin-4-amine
Figure BDA0002594682410000131
The compound 6-bromo-4-chloro-7-methoxyquinazoline (2.50g, 9.19mmol) was dissolved in NMP (20mL), 3, 4-dichloro-2-fluoroaniline (2.84g, 9.19mmol) was added thereto, and the mixture was stirred at 110 ℃ for 6 hours. After completion of the reaction, the reaction mixture was cooled, and then water (200mL) was added thereto, followed by extraction with EA 3 times. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. Anhydrous Na2SO4After drying, the reaction mixture was concentrated to dryness, and the obtained crude product was purified by column chromatography (PE/EA ═ 3/1 to 1/1) to obtain the objective compound (1.68g, yield 44.1%) as a yellow solid.
EM (calculated): 414.9, respectively; MS (ESI) M/z (M + H)+:416.0。
Intermediate 4: preparation of 6-bromo-N- (3-chloro-4-fluorophenyl) -7-methoxyquinazolin-4-amine
Figure BDA0002594682410000132
The compound 6-bromo-4-chloro-7-methoxyquinazoline (1.00g, 3.68mmol) was dissolved in NMP (20mL), 3-chloro-4-fluoroaniline (0.53g, 3.68mmol) was added thereto, and the mixture was stirred at 110 ℃ overnight. After completion of the reaction, the reaction mixture was cooled, and then water (200mL) was added thereto, followed by extraction with EA 3 times. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. Anhydrous Na2SO4After drying, the reaction mixture was concentrated to dryness, and the obtained crude product was purified by column chromatography (PE/EA ═ 3/1 to 1/1) to obtain the target compound (737mg, yield 52.6%) as a yellow solid.
EM (calculated): 381.0, respectively; MS (ESI) M/z (M + H)+:382.0。
Intermediate 5: preparation of 6-bromo-N- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinolin-4-amine
Figure BDA0002594682410000141
The compound 6-bromo-4-chloro-7-methoxyquinoline (3.00g, 11.07mmol) was dissolved in NMP (30mL), 3, 4-dichloro-2-fluoroaniline (1.98g, 11.07mmol) was added thereto, and after dissolution, concentrated hydrochloric acid (5mL) was added thereto, and the mixture was stirred at 110 ℃ overnight. After completion of the reaction, the reaction solution was cooled to room temperature, stirred under an ice-water bath and acetonitrile (30mL) was slowly added thereto. A large amount of solid was precipitated, collected by filtration, rinsed with a small amount of acetonitrile, and dried to give the objective compound (2.60g, yield 56.8%) as a yellow solid.
EM (calculated): 413.9; MS (ESI) M/z (M + H)+:415.0
Intermediate 6: preparation of 4- ((3, 4-dichloro-2-fluorophenyl) amino) quinazoline-6, 7-diol
Figure BDA0002594682410000142
The compound 4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-ol (500mg, 1.42mmol) was mixed with pyridine hydrochloride (2.00g, 17.39mmol) and stirred at 170 ℃ for 4 h under nitrogen. After completion of the reaction, the reaction mixture was cooled to room temperature, and water (10mL) was added thereto with stirring. A large amount of solid precipitated, which was collected by filtration and dried to give the title compound (252mg, yield 52.3%) as an off-white solid.
EM (calculated): 339.0; MS (ESI) M/z (M + H)+:340.0
Example 1
Preparation of 1- (1R,5S) - (3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-yl) oxy) -8-azabicyclo [3.2.1] octan-8-yl) prop-2-en-1-one
Figure BDA0002594682410000143
Step 1: preparation of (1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-yl) oxy) -8-azabicyclo [3.2.1] octane-8-tert-butyl carboxylate
Mixing compound 4- ((3, 4-dichloro)-2-fluorophenyl) amino) -7-methoxyquinazolin-6-ol (200mg, 0.57mmol) and (1R,5S) -3- (p-tolyloxy) -8-azabicyclo [3.2.1]Octane-8-tert-butyl formate (328mg, 0.86mmol) was added to DMF (10mL), potassium carbonate (157mg, 1.14mmol) was added thereto, and the mixture was stirred overnight at 70 ℃. After completion of the reaction, the reaction mixture was cooled to room temperature, and water (100mL) was added thereto. Extract 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. Anhydrous Na2SO4After drying, the crude product was concentrated to dryness, and the crude product was purified by column chromatography (PE/EA 10/1-1/1) to obtain the objective compound (80mg, yield 25.0%) as a white solid.
EM (calculated): 562.2; MS (ESI) M/z (M + H)+:563.2
Step 2: preparation of 6- ((1R,5S) -8-azabicyclo [3.2.1] octan-3-yl) oxy) -N- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinazolin-4-amine trifluoroacetate
Compound (1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-yl) oxy) -8-azabicyclo [3.2.1] octane-8-tert-butyl carboxylate (80mg, 0.14mmol) was dissolved in DCM (5mL), to which TFA (1mL) was added, and stirred at room temperature for 2 hours. After the reaction is finished, the reaction solution is concentrated to be dry, and the obtained crude product is directly used in the next reaction.
EM (calculated): 462.1 of the first group; MS (ESI) M/z (M + H)+:463.2
And step 3: preparation of 1- (1R,5S) - (3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-yl) oxy) -8-azabicyclo [3.2.1] octan-8-yl) prop-2-en-1-one
The compound 6- ((1R,5S) -8-azabicyclo [3.2.1]Octane-3-yl) oxy) -N- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinazolin-4-amine trifluoroacetate (crude) was dissolved in THF/water (5mL, 5/1), sodium carbonate (74mg, 0.70mmol) was added and stirred in an ice-water bath for 10 min. Acryloyl chloride (13mg, 0.14mmol) was added dropwise to the reaction system, the reaction solution was warmed to room temperature, and stirring was continued for 1 hour. After completion of the reaction, water (10mL) was added to the reaction system, and extracted 3 times with EA. The organic phases were combined and washed with anhydrous Na2SO4And (5) drying. ConcentratingTo dryness, the crude product obtained was purified by column chromatography (PE/EA-10/1) to give the title compound (45mg, 62.3% total yield of two steps) as a white solid.
EM (calculated): 516.1; MS (ESI) M/z (M + H)+:517.2
1H NMR(400MHz,DMSO-d6)δ1.86-1.88(1H,m),1.99-2.06(3H,m),2.10-2.19(2H,m),2.25-2.27(2H,m),3.97(3H,s),4.54-4.56(2H,m),4.85-4.88(1H,m),5.68-5.71(1H,m),6.21(1H,d,J=2.0Hz),6.70-6.75(1H,m),7.27(1H,s),7.59-7.62(2H,m),7.74(1H,s),8.40(1H,s),9.61(1H,s).
The compounds of examples 2-5 were synthesized according to the procedure described in example 1:
TABLE 1
Figure BDA0002594682410000151
Figure BDA0002594682410000161
Figure BDA0002594682410000171
Example 6
1- (4- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-yl) oxy) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002594682410000172
Step 1: preparation of tert-butyl 4- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-yl) oxy) piperidine-1-carboxylate
Compound 4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-ol (120mg, 0.34mmol) and tert-butyl 4- (p-oxy) piperidine-1-carboxylate (181mg, 0.51mmol) were added to DMF (4mL), to which was added potassium carbonate (94mg, 0.68mmol), and stirred at 70 ℃ overnight. After the reaction is finishedAfter completion, the reaction mixture was cooled to room temperature, and water (100mL) was added thereto. Extract 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. Anhydrous Na2SO4After drying, the reaction mixture was concentrated to dryness, and the obtained crude product was purified by column chromatography (PE/EA ═ 10/1 to 3/1) to obtain the target compound (60mg, yield 33.0%) as a brown solid.
EM (calculated): 535.1; MS (ESI) M/z (M + H)+:536.2
Step 2: preparation of N- (3, 4-dichloro-2-fluorophenyl) -7-methoxy-6- (piperidin-4-yloxy) quinolin-4-amine trifluoroacetate
Tert-butyl 4- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-yl) oxy) piperidine-1-carboxylate (60mg, 0.11mmol) was dissolved in DCM (5mL), to which TFA (1mL) was added and stirred at room temperature for 2 hours. After the reaction is finished, the reaction solution is concentrated to be dry, and the obtained crude product is directly used in the next reaction.
EM (calculated): 435.1; MS (ESI) M/z (M + H)+:436.1
And step 3: preparation of 1- (4- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-yl) oxy) piperidin-1-yl) prop-2-en-1-one
The compound N- (3, 4-dichloro-2-fluorophenyl) -7-methoxy-6- (piperidin-4-yloxy) quinolin-4-amine trifluoroacetate (crude) was dissolved in THF/water (5mL, 5/1), sodium carbonate (58mg, 0.55mmol) was added thereto and stirred in an ice-water bath for 10 minutes. Acryloyl chloride (10mg, 0.11mmol) was added dropwise to the reaction system, the reaction solution was warmed to room temperature, and stirring was continued for 1 hour. After completion of the reaction, water (10mL) was added to the reaction system, and extracted 3 times with EA. The organic phases were combined and washed with anhydrous Na2SO4And (5) drying. The reaction mixture was concentrated to dryness, and the resulting crude product was purified by column chromatography (PE/EA ═ 10/1) to give the title compound (12mg, total yield in two steps 22.3%) as an off-white solid.
EM (calculated): 489.1, respectively; MS (ESI) M/z (M + H)+:490.1
1H NMR(400MHz,CDCl3)δ1.85-1.95(2H,m),2.06-2.17(2H,m),3.61-3.66(2H,m),3.84-3.92(1H,m),3.90(3H,s),4.03-4.11(1H,m),5.00-5.09(1H,m),5.70-5.73(1H,m),6.27-6.31(1H,m),6.42(1H,s),6.58-6.65(1H,m),7.20-7.22(1H,m),7.33-7.36(2H,m),7.44(1H,s),7.95-8.05(2H,m).
The compound of example 7 was synthesized according to the procedure shown in example 6:
TABLE 2
Figure BDA0002594682410000181
Example preparation of 81- ((1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) prop-2-en-1-one
Figure BDA0002594682410000191
Step 1: preparation of (1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-yl) amino) -8-azabicyclo [3.2.1] octane-8-tert-butyl carboxylate
The compound 6-bromo-N- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinazolin-4-amine (200mg, 0.48mmol), (1R,5S) -3-3-amino-8-azabicyclo [ 3.2.1%]Octane-8-carboxylic acid tert-butyl ester (163mg, 0.72mmol), Pd2(dba)3(46mg, 0.05mmol), x-phos (48mg, 0.10mmol) and sodium tert-butoxide (92mg, 0.96mmol) were dispersed in DMA (8mL) and placed in an oil bath pre-heated to 100 ℃ under nitrogen and stirred for 2 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, and water (80mL) was added thereto and stirred for 5 minutes. Extract 3 times with EA, combine the organic phases and wash 1 time with saturated aqueous NaCl. Na (Na)2SO4After drying, it was concentrated to dryness, and the resulting crude product was purified by column chromatography (DCM/MeOH ═ 50/1) to give the title compound (137mg, yield 51.1%) as a white solid.
EM (calculated): 561.2, respectively; MS (ESI) M/z (M + H)+:562.2
Step 2: n is a radical of6- ((1R,5S) -8-azabicyclo [ 3.2.1)]Octane-3-yl) -N4- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinazoline-4, 6-diamine, trisPreparation of fluoroacetate
Compound (1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-yl) amino) -8-azabicyclo [3.2.1] octane-8-tert-butyl carboxylate (135mg, 0.24mmol) was dissolved in DCM (15mL), to which TFA (2mL) was added, and stirred at room temperature for 2 hours. After the reaction is finished, the reaction solution is concentrated to be dry, and the obtained crude product is directly used in the next reaction.
EM (calculated): 461.1; MS (ESI) M/z (M + H)+:462.1
And step 3: preparation of 1- ((1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinazolin-6-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) prop-2-en-1-one
Compound N6- ((1R,5S) -8-azabicyclo [ 3.2.1)]Octane-3-yl) -N4- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinazoline-4, 6-diamine trifluoroacetate salt (crude) is dissolved in THF/water (10mL, 5/1), sodium carbonate (127mg, 1.20mmol) is added thereto and stirred in an ice-water bath for 10 minutes. Acryloyl chloride (22mg, 0.24mmol) was added dropwise to the reaction system, the reaction solution was warmed to room temperature, and stirring was continued for 1 hour. After completion of the reaction, water (10mL) was added to the reaction system, and extracted 3 times with EA. The organic phases were combined and washed with anhydrous Na2SO4And (5) drying. The reaction mixture was concentrated to dryness, and the resulting crude product was purified by column chromatography (DCM/MeOH 100/1) to give the title compound (47mg, 37.9% in two-step total yield) as a yellow solid.
EM (calculated): 515.1; MS (ESI) M/z (M + H)+:516.1
1H NMR(400MHz,DMSO-d6)δ1.51-1.56(1H,m),1.60-1.66(1H,m),1.88-1.94(1H,m),1.99-2.13(5H,m),3.93(3H,s),4.05-4.10(1H,m),4.57-4.59(2H,m),5.37(1H,d,J=9.2Hz),5.69(1H,dd,J=10.4Hz,2.4Hz),6.16-6.21(1H,m),6.70-6.76(1H,m),7.07(1H,s),7.13(1H,s),7.58(1H,dd,J=8.0Hz,1.2Hz),7.64-7.68(1H,m),8.25(1H,s),9.33(1H,s).
The compounds of examples 9, 10, 15, 16 were synthesized according to the procedure described in example 8:
TABLE 3
Figure BDA0002594682410000201
Figure BDA0002594682410000211
Example 11
1- ((1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-yl) amino) -8-azabicyclo [3.2.1]Preparation of octan-8-yl) prop-2-en-1-one
Figure BDA0002594682410000221
Step 1: preparation of (1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-yl) amino) -8-azabicyclo [3.2.1] octane-8-tert-butyl carboxylate
The compound 6-bromo-N- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinolin-4-amine (300mg, 0.72mmol), (1R,5S) -3-amino-8-azabicyclo [3.2.1]Octane-8-carboxylic acid tert-butyl ester (244mg, 1.08mmol), Pd2(dba)3(64mg, 0.07mmol), x-phos (67mg, 0.14mmol) and sodium tert-butoxide (138mg, 1.44mmol) were dispersed in 1,4-dioxane (20mL) and placed in an oil bath pre-heated to 100 ℃ under nitrogen and stirred for 4 hours. After completion of the reaction, the reaction liquid was cooled to room temperature and concentrated to dryness, and the obtained crude product was purified by column chromatography (DCM/MeOH 50/1) to obtain the objective compound (192mg, yield 47.6%) as a yellow solid.
EM (calculated): 560.2; MS (ESI) M/z (M + H)+:561.2
Step 2: n is a radical of6- ((1R,5S) -8-azabicyclo [ 3.2.1)]Octane-3-yl) -N4Preparation of (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinoline-4, 6-diamine trifluoroacetate
Compound (1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-yl) amino) -8-azabicyclo [3.2.1] octane-8-tert-butyl carboxylate (190mg, 0.34mmol) was dissolved in DCM (15mL), to which TFA (2mL) was added, and stirred at room temperature for 2 hours. After the reaction is finished, the reaction solution is concentrated to be dry, and the obtained crude product is directly used in the next reaction.
EM (calculated): 460.1; MS (ESI) M/z (M + H)+:461.1
And step 3: preparation of 1- ((1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-methoxyquinolin-6-yl) amino) -8-azabicyclo [3.2.1] octan-8-yl) prop-2-en-1-one
Compound N6- ((1R,5S) -8-azabicyclo [ 3.2.1)]Octane-3-yl) -N4- (3, 4-dichloro-2-fluorophenyl) -7-methoxyquinoline-4, 6-diamine trifluoroacetate salt (crude) is dissolved in THF/water (15mL, 5/1), sodium carbonate (180mg, 1.70mmol) is added thereto and stirred in an ice-water bath for 10 minutes. Acryloyl chloride (31mg, 0.34mmol) was added dropwise to the reaction system, the reaction solution was warmed to room temperature, and stirring was continued for 1 hour. After completion of the reaction, water (10mL) was added to the reaction system, and extracted 3 times with EA. The organic phases were combined and washed with anhydrous Na2SO4And (5) drying. The reaction mixture was concentrated to dryness, and the resulting crude product was purified by column chromatography (DCM/MeOH 100/1) to give the title compound (53mg, 30.3% in two-step total yield) as an off-white solid.
EM (calculated): 514.1; MS (ESI) M/z (M + H)+:515.1
1H NMR(400MHz,DMSO-d6)δ1.50-1.62(2H,m),1.83-1.87(2H,m),1.91-2.06(4H,m),3.94(3H,s),3.97-4.04(1H,m),4.52-4.54(2H,m),5.47(1H,d,J=9.2Hz),5.69(1H,dd,J=10.4Hz,2.4Hz),6.18(1H,dd,J=16.8Hz,2.4Hz),6.56(1H,dd,J=6.0Hz,2.0Hz),6.69-6.75(1H,m),7.02(1H,s),7.20(1H,s),7.32-7.36(1H,m),7.58(1H,dd,J=9.0Hz,1.4Hz),8.24(1H,d,J=5.6Hz),9.21(1H,brs).
The compound of example 12 was synthesized according to the procedure shown in example 11:
TABLE 4
Figure BDA0002594682410000231
Example 13
Preparation of 1- (4- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7- (difluoromethoxy) quinazolin-6-yl) oxy) piperidin-1-yl) prop-2-en-1-one
Figure BDA0002594682410000241
Step 1: preparation of 4- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-hydroxyquinazolin-6-yl) oxy) piperidine-1-tert-butyl formate
The compound 4- ((3, 4-dichloro-2-fluorophenyl) amino) quinazoline-6, 7-diol (200mg, 0.59mmol) and tert-butyl 4- (p-oxy) piperidine-1-carboxylate (188mg, 0.53mmol) were added to DMF (10mL), to which was added potassium carbonate (98mg, 0.71mmol), warmed to 50 ℃ and stirred overnight. After completion of the reaction, the reaction mixture was cooled to room temperature, and water (100mL) was added thereto. Extract 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. Anhydrous Na2SO4After drying, it was concentrated to dryness, and the obtained crude product was purified by column chromatography (PE/EA ═ 1/1) to obtain the objective compound (93mg, yield 30.2%) as a white solid.
EM (calculated): 522.1; MS (ESI) M/z (M + H)+:523.2
Step 2: preparation of 4- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7- (difluoromethoxy) quinazolin-6-yl) oxy) piperidine-1-tert-butyl formate
The compound 4- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-hydroxyquinazolin-6-yl) oxy) piperidine-1-tert-butyl carboxylate (70mg, 0.13mmol) and 2-chloro-2, 2-difluoro-1-phenylethane-1-one (49mg, 0.26mmol) were added to acetonitrile/water (10mL, 5/1), to which was added potassium carbonate (36mg, 0.26mmol), warmed to 80 ℃ and stirred for 1 hour. After completion of the reaction, the reaction mixture was cooled to room temperature, and water (50mL) was added thereto. Extracting with EA for 3 times, anhydrous Na2SO4After drying, it was concentrated to dryness, and the resulting crude product was purified by column chromatography (DCM/MeOH ═ 70/1) to give the title compound (30mg, yield 40.5%) as a yellow solid.
EM (calculated): 572.1, respectively; MS (ESI) M/z (M + H)+:573.2
And step 3: preparation of N- (3, 4-dichloro-2-fluorophenyl) -7- (difluoromethoxy) -6- (piperidin-4-yloxy) quinazolin-4-amine trifluoroacetate
Compound 4- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7- (difluoromethoxy) quinazolin-6-yl) oxy) piperidine-1-tert-butylcarboxylate (30mg, 0.05mmol) was dissolved in DCM (5mL), to which TFA (1mL) was added and stirred at room temperature for 2 hours. After the reaction is finished, the reaction solution is concentrated to be dry, and the obtained crude product is directly used in the next reaction.
EM (calculated): 472.1, respectively; MS (ESI) M/z (M + H)+:473.1
And 4, step 4: preparation of 1- (4- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7- (difluoromethoxy) quinazolin-6-yl) oxy) piperidin-1-yl) prop-2-en-1-one
The compound N- (3, 4-dichloro-2-fluorophenyl) -7- (difluoromethoxy) -6- (piperidin-4-yloxy) quinazolin-4-amine trifluoroacetate (crude) was dissolved in THF/water (5mL, 5/1), sodium carbonate (27mg, 0.25mmol) was added and stirred in an ice-water bath for 10 min. Acryloyl chloride (5mg, 0.05mmol) was added dropwise to the reaction system, the reaction solution was warmed to room temperature, and stirring was continued for 30 minutes. After completion of the reaction, water (5mL) was added to the reaction system, and extracted 3 times with EA. The organic phases were combined and washed with anhydrous Na2SO4And (5) drying. Concentrate to dryness and purify the resulting crude by TLC (DCM/MeOH ═ 60/1) to give the title compound (10mg, total yield of two steps 38.0%) as an off-white solid.
EM (calculated): 526.1 of the total weight of the steel; MS (ESI) M/z (M + H)+:527.1
1H NMR(400MHz,DMSO-d6)δ1.67-1.77(2H,m),1.96-2.08(2H,m),3.48-3.57(2H,m),3.78-3.85(2H,m),4.99-5.05(1H,m),5.68-5.71(1H,m),6.10-6.15(1H,m),6.82-6.89(1H,m),7.19(1H,t,J=73.6Hz),7.51-7.53(2H,m),7.55-7.59(2H,m),8.04(1H,brs),8.30(1H,s),8.45(1H,s).
Example 14
1- ((1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7- (methoxy-d)3) Quinazolin-6-yl) oxy) -8-azabicyclo [3.2.1]Preparation of octan-8-yl) prop-2-en-1-one
Figure BDA0002594682410000251
Step 1: preparation of (1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-hydroxyquinazolin-6-yl) oxy) -8-azabicyclo [3.2.1] octane-8-tert-butyl carboxylate
The compound 4- ((3, 4-dichloro-2-fluorophenyl) amino) quinazoline-6, 7-diol (250mg, 0.74mmol) and (1R,5S) -3- (p-tolyloxy) -8-azabicyclo [3.2.1]Tert-butyl octane-8-carboxylate (252mg, 0.66mmol) was added to DMF (10mL), potassium carbonate (153mg, 1.11mmol) was added thereto, and the mixture was heated to 50 ℃ and stirred overnight. After completion of the reaction, the reaction mixture was cooled to room temperature, and water (100mL) was added thereto. Extract 3 times with EA. The organic phases were combined and washed 1 time with saturated aqueous NaCl solution. Anhydrous Na2SO4After drying, it was concentrated to dryness, and the obtained crude product was purified by column chromatography (PE/EA ═ 1/1) to obtain the objective compound (107mg, yield 29.6%) as a white solid.
EM (calculated): 548.1, respectively; MS (ESI) M/z (M + H)+:549.2
Step 2: (1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7- (methoxy-d)3) Quinazolin-6-yl) oxy) -8-azabicyclo [3.2.1]Preparation of octane-8-tert-butyl formate
Mixing compound (1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7-hydroxyquinazolin-6-yl) oxy) -8-azabicyclo [3.2.1]Octane-8-tert-butyl formate (105mg, 0.19mmol) and potassium carbonate (40mg, 0.29mmol) were added to acetonitrile (10mL), and deuterated iodomethane (28mg, 0.19mmol) was added thereto, and the mixture was stirred at room temperature for 3 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, and water (50mL) was added thereto. Extracting with EA for 3 times, anhydrous Na2SO4After drying, it was concentrated to dryness, and the resulting crude product was purified by column chromatography (DCM/MeOH ═ 100/1) to give the title compound (60mg, yield 56.1%) as a yellow solid.
EM (calculated): 565.2; MS (ESI) M/z (M + H)+:566.2
And step 3: 6- ((1R,5S) -8-azabicyclo [3.2.1]Octane-3-yl) oxy) -N- (3, 4-dichloro-2-fluorophenyl) -7- (methoxy-d3) Quinazolin-4-amine trifluoroacetate saltPreparation of
Mixing compound (1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7- (methoxyl-d)3) Quinazolin-6-yl) oxy) -8-azabicyclo [3.2.1]Octane-8-tert-butyl carboxylate (60mg, 0.11mmol) was dissolved in DCM (6mL), and TFA (1mL) was added thereto and stirred at room temperature for 2 h. After the reaction is finished, the reaction solution is concentrated to be dry, and the obtained crude product is directly used in the next reaction.
EM (calculated): 465.1, respectively; MS (ESI) M/z (M + H)+:466.2
And 4, step 4: 1- ((1R,5S) -3- ((4- ((3, 4-dichloro-2-fluorophenyl) amino) -7- (methoxy-d)3) Quinazolin-6-yl) oxy) -8-azabicyclo [3.2.1]Preparation of octan-8-yl) prop-2-en-1-one
The compound 6- ((1R,5S) -8-azabicyclo [3.2.1]Octane-3-yl) oxy) -N- (3, 4-dichloro-2-fluorophenyl) -7- (methoxy-d3) Quinazolin-4-amine trifluoroacetate (crude) was dissolved in THF/water (5mL, 5/1), sodium carbonate (58mg, 0.55mmol) was added and stirred in an ice water bath for 10 minutes. Acryloyl chloride (10mg, 0.11mmol) was added dropwise to the reaction system, the reaction solution was warmed to room temperature, and stirring was continued for 30 minutes. After completion of the reaction, water (5mL) was added to the reaction system, and extracted 3 times with EA. The organic phases were combined and washed with anhydrous Na2SO4And (5) drying. Concentrate to dryness and purify the resulting crude by TLC (DCM/MeOH ═ 60/1) to give the title compound (14mg, total yield of two steps 24.6%) as an off-white solid.
EM (calculated): 519.1, respectively; MS (ESI) M/z (M + H)+:520.1
1H NMR(400MHz,DMSO-d6)δ1.83-1.87(1H,m),1.96-2.08(4H,m),2.12-2.23(3H,m),4.52-4.53(2H,m),4.97(1H,s),5.70(1H,dd,J=10.0Hz,2.4Hz),6.17-6.22(1H,m),6.70-6.77(1H,m),7.14(1H,s),7.56-7.61(2H,m),7.82(1H,s),8.38(1H,s),9.72(1H,s).
Test example 1 inhibitory Effect of Compounds on kinase Activity
1: test materials:
EGFR(Carna,No.13CBS-00055),EGFR T790M(Carna,No.08CBS-0510N),EGFR T790M/L858R(Carna,No.12CBS-0765H),Kinase substrate22(GL,No.P190917-CL11239 3),DMSO(Sigma,No.SHBG3288V),384-well plate(Corning,No.12619003).
2: the experimental method comprises the following steps:
2.1 preparation of Compounds
Compounds were received by the administrator and dissolved in 100% DMSO to make 10mM stock, stored in nitrogen cabinet protected from light.
2.2 kinase reaction Processes
(1) A1 XKinase buffer was prepared.
(2) Preparation of compound concentration gradient: test compounds were tested at 1000nM, diluted to 100-fold final concentration in 100% DMSO solutions in 384source plates, and compounds were diluted 3-fold with Precision, 10 concentrations. Using a dispenser Echo 550 to the target plate OptiPlate-384F transfer 250nL 100 times the final concentration of compounds.
(3) A2.5 fold final concentration of Kinase solution was prepared using a 1 XKinase buffer.
(4) Add 10. mu.L of 2.5 fold final concentration kinase solution to the compound well and positive control well, respectively; mu.L of 1 XKinase buffer was added to the negative control wells.
(5) Centrifuge at 1000rpm for 30 seconds, shake the plate and incubate at room temperature for 10 minutes.
(6) A mixture of ATP and Kinase substrate22 was made up at 5/3 fold final concentration using 1 XKinase buffer.
(7) The reaction was initiated by adding 15. mu.L of a mixed solution of ATP and substrate at 5/3-fold final concentration.
(8) The 384 well plates were centrifuged at 1000rpm for 30 seconds, shaken and mixed and incubated at room temperature for the appropriate time.
(9) Add 30. mu.L of termination detection solution to stop the kinase reaction, centrifuge at 1000rpm for 30 seconds, shake and mix.
(10) The conversion was read using a Caliper EZ Reader.
2.3 data analysis
Formula for calculation
Figure BDA0002594682410000271
Wherein: conversion% _ sample is the Conversion reading for the sample; conversion% _ min: negative control well mean, representing conversion readings without enzyme live wells; conversion% _ max: positive control wells are averaged for conversion readings in wells without compound inhibition.
Fitting a dose-response curve:
the log values of the concentrations were taken as the X-axis and the percent inhibition as the Y-axis, and the log (inhibitor) vs. response-Variable slope of the analytical software GraphPad Prism5 was used to fit the dose-effect curves to obtain IC50 values for the enzyme activities of the respective compounds. The calculation formula is:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))
the test results are shown in table 5:
inhibitory Activity of the Compounds of Table 5 against EGFR WT and mutant T790M, T790M/L858R kinase (IC50)
Example numbering WT(nM) T790M(nM) T790M/L858R(nM)
1 0.26 1.7 2.2
2 0.35 72 192
3 0.57 105 /
4 0.90 43 /
5 2.0 52 /
6 1.0 1.0 1.0
7 1.5 1.2 2.03
8 0.30 0.34 0.52
9 0.32 0.60 1.02
10 20.0 / /
11 0.86 0.70
12 4.5 2.3 /
13 34 883 >1000
14 21 470 /
15 0.63 1.22 2.53
16 0.92 1.39 1.98
Pozitinib 0.28 0.79 0.81
Test example 2 inhibitory Effect of Compounds on cell Activity
1: cell lines
Cell lines Cell type Cell number/well Culture medium
BaF3 EGFR-D770-N771ins_SVD Suspended in water 3000 RPMI-1640+10%FBS
Placing at 37 ℃ and 5% CO2And culturing under 95% humidity.
2: reagent and consumable
Fetal bovine serum FBS (GBICO, Cat #10099-141)
CellTiter-
Figure BDA0002594682410000282
Luminescent Cell Viability Assay(Promega,Cat#G7573)
96-hole transparent flat-bottom black wall plate (
Figure BDA0002594682410000281
Cat#165305)
RPMI-1640(Hyclone,Cat#SH30809.01)
3: cell culture and inoculation:
cells in the logarithmic growth phase were harvested and counted using a platelet counter. Detecting the cell viability by using a trypan blue exclusion method to ensure that the cell viability is over 90 percent;
adjusting the cell concentration; add 90 μ Ι _ of cell suspension to 96-well plates, respectively;
cells in 96-well plates were incubated overnight at 37 ℃ with 5% CO2 and 95% humidity.
4: drug dilution and dosing:
preparing 10 times of drug solution, wherein the highest concentration is 10 mu M, the concentration is 9, the dilution is 3.16 times, 10 mu L of drug solution is added into each hole of a 96-hole plate inoculated with cells, three multiple holes are arranged for each drug concentration, the final acting concentration of the compound is 1 mu M, the concentration is 9, the dilution is 3.16 times, and the final acting concentration of DMSO is 0.1%;
the cells in the dosed 96-well plate were incubated for a further 72 hours at 37 ℃ under 5% CO2 and 95% humidity, after which they were subjected to CTG analysis.
5: reading the plate at the end:
melt CTG reagents and equilibrate cell plates to room temperature for 30 minutes;
adding equal volume of CTG solution into each well;
the cells were lysed by shaking on an orbital shaker for 5 minutes;
the cell plate was left at room temperature for 20 minutes to stabilize the luminescence signal;
and reading the cold light value.
6: data processing
Data were analyzed using GraphPad Prism 7.0 software, fitted to the data using non-linear S-curve regression to derive a dose-effect curve, and IC50 values were calculated therefrom.
The cell survival rate (%) × (Lum test drug-Lum culture solution control)/(Lum cell control-Lum culture solution control) × 100%.
The test results are shown in table 6:
TABLE 6 Compound vs BaF3Inhibitory Activity of EGFR-D770-N771ins _ SVD cells IC50(nm)
Examples of the embodimentsNumber (C) BaF3 EGFR-D770-N771ins_SVD
1 3.07
6 33.53
7 22.83
8 1.50
9 28.78
11 17.15
12 397.10
15 15.38
16 20.94
Pozitinib 3.23
Test example 3 detection of Compound liver microsome stability test
1: materials and methods
Buffer solution:
(1)100mM potassium phosphate buffer, pH 7.4; (2)10mM MgCl 2.
Preparation of compound solution:
(1) preparation of 100. mu.M working solution: mu.L of the stock solution (10mM) of the test or control group was diluted with 495. mu.L of methanol to give a compound concentration of 100. mu.M (99% MeOH).
(2) Preparation of 10 μ M working solution: mu.L of 100. mu.M working solution was diluted with 450. mu.L of 100mM potassium phosphate buffer to give a compound concentration of 10. mu.M (9.9% MeOH).
Composition of NADPH (prototype coenzyme II) regeneration System (final concentration of isocitrate dehydrogenase in culture broth 1.0 unit/mL):
β -nicotinamide adenine dinucleotide phosphate, supplier: chem-impex international shipment number: preparation of a solution of N00616 liver microsomes (final concentration of 0.5mg protein/mL), the liver microsomes were of the type shown in Table 7:
TABLE 7
Figure BDA0002594682410000301
Stopping liquid:
acetonitrile ice-cold solution containing 100ng/mL tolbutamide and 100ng/mL labetalol as internal standards.
The method comprises the following operation steps:
(1) in addition to the blank matrix plate wells, 10 μ L of working solution of test or control drug was added to each of the plate wells (T0, T5, T10, T20, T30, T60, and NCF 60).
(2) 80 μ L/well of the microsome solution was dispensed onto each plate using Apricot and the mixture of microsome solution and compound was incubated at 37 ℃ for about 10 minutes.
(3) To NCF60 was added 10. mu.L of 100mM potassium phosphate buffer/well, incubated at 37 ℃ and timer 1 was started for the time shown in Table 8.
TABLE 8
Figure BDA0002594682410000302
(4) After preheating, 10. mu.L/well of NADPH regeneration system was dispensed to each plate using Apricot to start the reaction.
TABLE 9 Final concentrations of each component in the incubation Medium
Figure BDA0002594682410000303
(5) Incubate at 37 ℃ and start timer 2, see table 10 for data.
Watch 10
Figure BDA0002594682410000311
(6) The reaction was stopped by adding pre-cooled stop solutions (containing 100ng/mL tolbutamide and 100ng/mL albuterol) at 4 ℃ to each well.
(7) The sample plate was then shaken on a shaking trigger for about 10 minutes.
(8) The samples were centrifuged at 4000rpm for 20min at 4 ℃.
(9) And adding 300 mu L of HPLC-grade water into each hole of another 96-hole plate, adding 100 mu L of supernate obtained by centrifugation into the corresponding hole position, and uniformly mixing the two for LC/MS/MS detection.
And (3) data analysis:
calculating t1/2 and Clint (mic) values according to first-order elimination kinetics
The first order elimination kinetics equation is:
Figure BDA0002594682410000312
Figure BDA0002594682410000313
Figure BDA0002594682410000314
Figure BDA0002594682410000315
Figure BDA0002594682410000316
the results of the partial compound liver microsome stability test are shown in table 11:
TABLE 11
Figure BDA0002594682410000317
Figure BDA0002594682410000321
From the research data of the patent drug property, the compound has obvious inhibition effect on the activity of a protein kinase inhibitor, and compared with clinical second-stage Pozitinib, the compound has better or equivalent kinase activity and cell activity and can be used as the protein kinase inhibitor; the compound has longer half-life period, lower clearance rate and more residual quantity after 60min, has more obvious advantage in the aspect of stability of liver microsomes, can reduce the application amount and the use frequency of medicaments, and has wide application prospect in resisting malignant tumor diseases or inflammatory diseases.
Although embodiments of the present invention have been shown and described, it will be appreciated by those skilled in the art that changes, modifications, substitutions and alterations can be made in these embodiments without departing from the principles and spirit of the invention, the scope of which is defined in the appended claims and their equivalents.

Claims (5)

1. A compound characterized by: has the following structure or a pharmaceutically acceptable salt thereof:
Figure DEST_PATH_IMAGE001
compound 11.
2. Use of a compound of claim 1 for the preparation of an EGFR inhibitor.
3. Use according to claim 2, wherein the EGFR inhibitor is selected from a wild-type EGFR inhibitor, an EGFR T790M inhibitor or an EGFR T790M/L858R inhibitor.
4. Use of a compound of claim 1, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a disease that causes overexpression of EGFR.
5. Use of a compound of claim 1 or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of a disease caused by overexpression of EGFR.
CN202010705735.XA 2020-07-21 2020-07-21 Compound with nitrogen-containing heterocyclic structure and preparation method and application thereof Active CN111763215B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010705735.XA CN111763215B (en) 2020-07-21 2020-07-21 Compound with nitrogen-containing heterocyclic structure and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010705735.XA CN111763215B (en) 2020-07-21 2020-07-21 Compound with nitrogen-containing heterocyclic structure and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN111763215A CN111763215A (en) 2020-10-13
CN111763215B true CN111763215B (en) 2021-05-18

Family

ID=72726890

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010705735.XA Active CN111763215B (en) 2020-07-21 2020-07-21 Compound with nitrogen-containing heterocyclic structure and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN111763215B (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113444052A (en) * 2021-07-01 2021-09-28 江苏君若药业有限公司 Synthesis of gefitinib
TW202328105A (en) * 2021-08-24 2023-07-16 美商安索治療公司 Quinazoline compounds for treatment of disease
CN115894486B (en) * 2021-09-30 2024-02-09 北京赛特明强医药科技有限公司 Hydrogenated pyridoquinazoline compound, composition and application thereof
CN117567466B (en) * 2024-01-16 2024-04-16 成都金瑞基业生物科技有限公司 Preparation method of quinazoline derivative

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101679384A (en) * 2007-06-05 2010-03-24 韩美药品株式会社 Novel amide derivative for inhibiting the growth of cancer cells
CN104513229A (en) * 2013-09-28 2015-04-15 正大天晴药业集团股份有限公司 Quinazoline derivatives and preparation method thereof
WO2015154725A1 (en) * 2014-04-11 2015-10-15 四川海思科制药有限公司 Quinazoline derivative and preparation method and use in medicine thereof
WO2019120213A1 (en) * 2017-12-19 2019-06-27 南京明德新药研发股份有限公司 Quinazoline derivative and use thereof
CN110577514A (en) * 2018-06-08 2019-12-17 江苏威凯尔医药科技有限公司 human epidermal growth factor receptor inhibitor and preparation method and application thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20130107479A (en) * 2012-03-22 2013-10-02 한미약품 주식회사 Novel sulfonate derivatives for inhibiting cancer cell growth

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101679384A (en) * 2007-06-05 2010-03-24 韩美药品株式会社 Novel amide derivative for inhibiting the growth of cancer cells
CN104513229A (en) * 2013-09-28 2015-04-15 正大天晴药业集团股份有限公司 Quinazoline derivatives and preparation method thereof
WO2015154725A1 (en) * 2014-04-11 2015-10-15 四川海思科制药有限公司 Quinazoline derivative and preparation method and use in medicine thereof
WO2019120213A1 (en) * 2017-12-19 2019-06-27 南京明德新药研发股份有限公司 Quinazoline derivative and use thereof
CN110577514A (en) * 2018-06-08 2019-12-17 江苏威凯尔医药科技有限公司 human epidermal growth factor receptor inhibitor and preparation method and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
基于喹唑啉结构的EGFR酪氨酸激酶抑制剂的设计与合成;覃雪梅;《中国博士学位论文全文数据库 医药卫生科技辑》;20170215(第2期);E079-12 *

Also Published As

Publication number Publication date
CN111763215A (en) 2020-10-13

Similar Documents

Publication Publication Date Title
CN111763215B (en) Compound with nitrogen-containing heterocyclic structure and preparation method and application thereof
CN110352188B (en) Fluoroallylamine derivatives and use thereof
EP2880035B1 (en) Novel pyrrolopyrimidine compounds as inhibitors of protein kinases
EP3985000A1 (en) 2,4-disubstituted pyrimidine derivative, preparation method for same, and uses thereof
KR102090453B1 (en) Salts of an epidermal growth factor receptor kinase inhibitor
US20220259235A1 (en) EGFR Inhibitor, Composition, and Preparation Method Therefor
EP3327014A1 (en) Egfr inhibitor and pharmaceutically acceptable salt and polymorph thereof, and use thereof
EP3173412A1 (en) 2,4-disubstituted 7h-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medicinal use thereof
JP6948659B1 (en) Pyridadinyl thiaazole carboxamide compound
EP2927232A1 (en) Heteroaryl alkyne compound and application thereof
US11512074B2 (en) Substituted diamino heterocyclic carboxamide compound and a composition containing the compound and use thereof
CN112110938B (en) Compound as protein kinase inhibitor and preparation method and application thereof
CN111377935B (en) Selective CDK4/6 inhibitor and application thereof
US20230095530A1 (en) Compound used as ret kinase inhibitor and application thereof
CN113993873A (en) EGFR inhibitors for the treatment of cancer
CN113416181B (en) Quinazoline derivative and application thereof
WO2021057867A1 (en) Class of cdk inhibitor based on organic arsine, preparation method and application thereof
CN109438279B (en) Small molecule compound for overcoming EGFR drug-resistant mutation and preparation method and application thereof
EP4155304A1 (en) Compound used as ret kinase inhibitor and application thereof
EP3766883B1 (en) Imidaxopyrolone compound and application thereof
CN111233774B (en) Amino pyrimidine compound
US20230133169A1 (en) Egfr inhibitor, composition, and method for preparation thereof
CN113354630B (en) 5,6-dihydrobenzo [ h ] quinazoline compound and application thereof
CN113773273B (en) Benzisothiazole compound, preparation method and application thereof
CN114423750B (en) 2, 4-Disubstituted pyrimidine derivative, preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
PE01 Entry into force of the registration of the contract for pledge of patent right
PE01 Entry into force of the registration of the contract for pledge of patent right

Denomination of invention: A compound with a nitrogen-containing heterocyclic structure and its preparation method and use

Effective date of registration: 20230612

Granted publication date: 20210518

Pledgee: Industrial Bank Limited by Share Ltd. Chengdu branch

Pledgor: Chengdu haibowei Pharmaceutical Co.,Ltd.

Registration number: Y2023510000144