WO2016082713A1 - 2-氨基嘧啶类化合物及其药物组合物和应用 - Google Patents

2-氨基嘧啶类化合物及其药物组合物和应用 Download PDF

Info

Publication number
WO2016082713A1
WO2016082713A1 PCT/CN2015/094954 CN2015094954W WO2016082713A1 WO 2016082713 A1 WO2016082713 A1 WO 2016082713A1 CN 2015094954 W CN2015094954 W CN 2015094954W WO 2016082713 A1 WO2016082713 A1 WO 2016082713A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
pyrimidin
naphthalen
butenamide
alkyl
Prior art date
Application number
PCT/CN2015/094954
Other languages
English (en)
French (fr)
Inventor
丁克
丁健
陈成斌
耿美玉
任小梅
谢华
涂正超
陈奕
Original Assignee
中国科学院上海药物研究所
中国科学院广州生物医药与健康研究院
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
Priority to CA2968633A priority Critical patent/CA2968633C/en
Priority to US15/529,333 priority patent/US10059694B2/en
Priority to EP15863532.6A priority patent/EP3225619B1/en
Priority to AU2015353210A priority patent/AU2015353210B2/en
Priority to CN201580016514.9A priority patent/CN106458930B/zh
Priority to KR1020177017442A priority patent/KR101941929B1/ko
Application filed by 中国科学院上海药物研究所, 中国科学院广州生物医药与健康研究院 filed Critical 中国科学院上海药物研究所
Priority to SG11201704244SA priority patent/SG11201704244SA/en
Priority to NZ733009A priority patent/NZ733009A/en
Priority to RU2017122062A priority patent/RU2704129C2/ru
Priority to JP2017528838A priority patent/JP6474068B2/ja
Publication of WO2016082713A1 publication Critical patent/WO2016082713A1/zh
Priority to IL252477A priority patent/IL252477B/en

Links

Images

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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • 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
    • 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/47One nitrogen atom and one oxygen or sulfur atom, e.g. cytosine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • 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
    • 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

Definitions

  • the invention belongs to the field of chemical medicine, in particular to 2-aminopyrimidine compounds and pharmaceutical compositions and uses thereof.
  • Tumor molecular targeted therapy is a treatment based on the selective killing of tumor cells by chemical or biological means of key molecules closely related to tumor growth.
  • the characteristics of targeted therapy are: high specificity, strong selectivity, and mild side effects; when combined, it can enhance the efficacy of traditional chemotherapy and radiotherapy and reduce postoperative recurrence.
  • Tumor targeted therapy is a hot spot and development trend of cancer treatment.
  • PTKs Protein tyrosine kinases
  • PTKs Protein tyrosine kinases
  • Studies have shown that more than half of all proto-oncogenes and oncogene activation are associated with protein tyrosine kinases.
  • the development of anti-tumor drugs with tyrosine kinase as a target has become a hot spot in the world, and it is also the focus of research and investment in drug development institutions in various countries.
  • EGFR Epidermal growth factor receptor
  • NSCLC non-small cell lung cancer
  • EGFR-positive NSCLC patients have significantly higher response rates to EGFR-TKI (EGFR-tyrosine kinase inhibitors) than EGFR wild-type NSCLC patients, progression-free survival (PFS) And the overall survival (OS) period is also significantly extended.
  • PFS progression-free survival
  • OS overall survival
  • Strategy 1 Continue to use EGFR-TKI, cross-use of gefitinib and erlotinib. In short, the continued use of TKI after TKI progress has certain benefits, but the degree of benefit is very limited.
  • Strategy 2 Development of a new EGFR-TKI.
  • Strategy 3 Treatment for other targets. Since the "bypass activation pathway" plays an important role in EGFR-TKI resistance, targeted drugs for these bypasses continue to emerge. However, the current EGFR-TKI still can not solve the clinical pressure caused by drug resistance, and the existing drugs are mostly EGFR reversible or irreversible inhibitors with quinazoline or quinolinamide as the basic mother nucleus. The toxic side effects caused by the poor selectivity of wild-type cells are also unavoidable.
  • a compound of formula I a pharmaceutically acceptable salt or stereoisomer thereof or a prodrug molecule thereof,
  • R 1 and R 2 are each independently H, halogen, cyano, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 6 cycloalkyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 6 cycloalkoxy;
  • R 3 is H or -(CH 2 ) m NR 8 R 9 ;
  • R 4 and R 5 are independently H, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 1 -C 6 alkane Oxygen, halogen, -(CH 2 ) m NR 8 R 9 , -(CH 2 ) m CR 6 R 8 R 9 ; wherein each m is independently 0, 1, 2 or 3;
  • R 6 is H or C 1 -C 3 alkyl;
  • each R 8 and each R 9 are independently selected from H, substituted or unsubstituted C 1 -C 6 alkyl, or R 8 , R 9 and the attached N or C together form unsubstituted or Substituted 3-8 membered monocyclic or fused ring containing 1, 2 or 3 heteroatoms selected from O, N, S;
  • W is NH, N(C 1 -C 3 alkyl), O or S;
  • X, Y, Z are each independently N or -CR 10 , wherein R 10 is hydrogen, halogen, substituted or unsubstituted C 1 -C 3 alkyl, substituted or unsubstituted C 1 -C 3 alkoxy ;
  • the salt is selected from a mineral acid salt or an organic acid salt such as a hydrochloride, a sulfate, a hydrobromide, a phosphate, a nitrate, an acetate, a maleate, a p-toluene. Sulfonate, methanesulfonate or trifluoroacetic acid.
  • each of the substituents is independently a monosubstituted, disubstituted, trisubstituted or tetrasubstituted.
  • W is NH, O, S or -N(CH 3 ).
  • X is N or -CH-.
  • Y is -CH-.
  • Z is -CH-.
  • R 1 and R 2 are each independently H, halogen, cyano, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 3 -C 6 cycloalkyl, C 3- C 6 cycloalkoxy, halogenated C 1 -C 6 alkyl or halogenated C 1 -C 6 alkoxy.
  • R 1 is H, fluorine, chlorine, bromine, cyano, C 1 -C 4 alkyl, fluoro C 1 -C 3 alkyl, fluoro C 1 -C 3 alkoxy or C 1 -C 3 alkoxy.
  • the fluoro C 1 -C 3 alkyl group is a trifluoromethyl group.
  • It is a phenyl group, a pyrrolyl group or a pyridyl group.
  • the compound of formula I has one or more of the following characteristics:
  • R 3 is H, -(CH 2 ) s N(C 1 -C 3 alkyl)(C 1 -C 3 alkyl), or Where s is 1 or 2 or 3;
  • R 4 is H, C 1 -C 3 alkyl, halogenated C 1 -C 3 alkyl, C 1 -C 3 alkoxy, halogenated C 1 -C 3 alkoxy, halogen, -( CH 2 ) m NR 8 R 9 , or
  • R 5 is H, C 1 -C 3 alkyl, halogenated C 1 -C 3 alkyl, C 1 -C 3 alkoxy, halogenated C 1 -C 3 alkoxy, halogen, -( CH 2 ) m NR 8 R 9 ,
  • each m is independently 0, 1, 2 or 3;
  • Each n 1 , each n 2 , each n 3 is independently 0, 1, 2 or 3;
  • Each V is independently CH, C (C 1 -C 3 alkyl) or N;
  • the compound of formula I has one or more of the following characteristics:
  • R 3 is H
  • R 4 is H, -CH 3 ,
  • R 5 is H, -CH 3 , F,
  • W is NH;
  • X, Y and Z are CH;
  • the structure of the compound of formula I is as shown in formula II:
  • R 1 , R 2 , R 3 , R 4 and R 5 are as defined above.
  • each substituent in the compound of formula I is a substituent at a corresponding position in each particular compound of the examples.
  • the compound of formula I is:
  • a second aspect of the invention provides a process for the preparation of a compound of formula I according to the first aspect, which comprises reacting a compound of formula III or a salt thereof with a compound of formula A or a salt thereof to give a compound of formula I, or a compound of formula B or Its salt reacts to give a compound of formula I,
  • R 7 is halogen, -OCOR 14 or OR 14 ,
  • R 14 is C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, C 6 -C 10 aryl or C 6 -C 10 aryl C 1 -C 6 alkyl, preferably C 1 -C 3 alkyl, halogenated C 1 -C 3 alkyl, phenyl or phenyl C 1 -C 3 alkyl.
  • a pharmaceutical composition comprising:
  • a compound of formula I according to the first aspect of the invention a pharmaceutically acceptable salt or stereoisomer thereof or a prodrug molecule thereof; and a pharmaceutically acceptable carrier.
  • a fourth aspect of the invention provides the use of a compound of formula I according to the first aspect, a pharmaceutically acceptable salt or stereoisomer thereof or a prodrug molecule thereof, or a pharmaceutical composition according to the third aspect, Make:
  • the tumor is selected from the group consisting of: non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cancer, gastrointestinal Stromal tumor, leukemia, histiocytic lymphoma, nasopharyngeal carcinoma, head and neck cancer, colon cancer, rectal cancer, glioma.
  • a method of treating a tumor wherein the subject of the first aspect is administered to the subject a compound, a pharmaceutically acceptable salt or stereoisomer thereof or a prodrug molecule thereof, or a pharmaceutical composition according to the third aspect.
  • a method of inhibiting EGFR protease wherein a compound of formula I, a pharmaceutically acceptable salt or a stereoisomer thereof or a prodrug molecule thereof, according to the first aspect, is administered to a subject in need thereof, or The pharmaceutical composition of the third aspect.
  • a method for inhibiting IGF1R protease which comprises administering a compound of formula I, a pharmaceutically acceptable salt or a stereoisomer thereof or a prodrug molecule thereof, according to the first aspect, or The pharmaceutical composition of the third aspect.
  • the desired subject is a non-human mammal or human, preferably a human, mouse or rat.
  • R 1 , R 2 , R 4 , R 5 , W, X, Y and Z are as defined above, provided that the compound of formula III does not include the following structure:
  • the salt is selected from a mineral acid salt or an organic acid salt such as a hydrochloride, a sulfate, a hydrobromide, a phosphate, a nitrate, an acetate, a maleate, a p-toluene. Sulfonate, methanesulfonate or trifluoroacetic acid.
  • It is a phenyl group, a pyrrolyl group or a pyridyl group.
  • the compound of the formula I of the present invention and a pharmaceutically acceptable salt thereof can inhibit the growth of a plurality of tumor cells and inhibit the EGFR, Her family protease, and particularly can selectively act on EGFR L858R/T790M and EGFR Del E745_A750 lung cancer. cell. Compared to wild-type cancer cells, such compounds have greater than 30-fold selectivity.
  • the compounds can be used for the preparation of anti-tumor drugs, and can overcome the resistance of the existing drugs gefitinib, erlotinib and the like, and are a novel class of drugs capable of overcoming the resistance of existing EGFR tyrosine kinase inhibitors. And a protein kinase inhibitor with selective and good pharmacokinetic properties.
  • the compounds of the present invention, and pharmaceutically acceptable salts thereof are useful in the preparation of hyperproliferative diseases such as tumors in humans and other mammals.
  • Figure 1 is a graph showing the results of the effects of compounds CCB120067 and CO1686 on the phosphorylation of EGFR T790M/L858R kinase and its downstream signaling pathway protein in NCI-H1975 cells containing the EGFR T790M/L858R mutation.
  • Figure 2 is a graph showing the results of the effects of the compounds CCB120067 and CO1686 on the phosphorylation of EGFR kinase and its downstream signaling pathway protein in A431 cells with high expression of wild-type EGFR.
  • Fig. 3 is a graph showing the results of growth inhibition of compound CCB120067 and CO1686 on subcutaneous xenografts of human lung cancer NCI-H1975 nude mice.
  • the inventors of the present application have extensively and intensively studied, and for the first time, unexpectedly developed a novel 2-aminopyrimidine compound, which can solve problems such as drug resistance, poor selectivity, and poor pharmacokinetic properties. On the basis of this, the present invention has been completed.
  • any variable e.g. R 8, R 9, etc.
  • any variable e.g. R 8, R 9, etc.
  • combinations of substituents and variables are allowed as long as such combinations stabilize the compound.
  • a line drawn from a substituent into the ring system means that the bond referred to can be attached to any ring atom that can be substituted. If the ring system is polycyclic, it means that such a bond is only attached to any suitable carbon atom of the adjacent ring. It will be appreciated that one of ordinary skill in the art can select substituents and substitution patterns for the compounds of the present invention to provide compounds which are chemically stable and which are readily synthesized from readily available starting materials by techniques in the art and the methods set forth below.
  • substituent itself is substituted by more than one group, it is understood that these groups may be on the same carbon atom or on different carbon atoms as long as the structure is stabilized.
  • substituted with a substituent selected from the group below is considered equivalent to the phrase “substituted with at least one substituent", and in this case the preferred embodiment will have from 1 to 4 substituents.
  • alkyl as used herein is meant to include branched and straight chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • the definition of "C 1 -C 6 " in “C 1 -C 6 alkyl” includes a group having 1, 2, 3, 4, 5 or 6 carbon atoms arranged in a straight chain or a branched chain.
  • “C 1 -C 6 alkyl” specifically includes methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, pentyl, hexyl.
  • cycloalkyl refers to a monocyclic saturated aliphatic hydrocarbon group having the specified number of carbon atoms.
  • cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl and the like.
  • alkoxy refers to the group -O- having an alkyl structure, such as -OCH 3, -OCH 2 CH 3, -OCH 2 CH 2 CH 3, -O-CH 2 CH (CH 3) 2, - OCH 2 CH 2 CH 2 CH 3 , -O-CH(CH 3 ) 2 and the like.
  • the "5- to 7-membered aromatic ring containing 0, 1, 2 or 3 hetero atoms selected from O, N or S” means a benzene ring or a heteroaryl ring
  • the heteroaryl ring within the scope of the present invention includes It is not limited to: imidazolyl, triazolyl, pyrazolyl, furyl, thienyl, oxazolyl, isoxazolyl, pyrazinyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl.
  • halo or halo as used herein means chloro, fluoro, bromo and iodo.
  • the present invention provides a compound of formula I, a pharmaceutically acceptable salt or stereoisomer thereof or a prodrug molecule thereof,
  • R 1 , R 2 , R 3 , W, R 4 , R 5 , X, Y, Z The definition is the same as before.
  • the invention includes the free forms of the compounds of formula I, as well as the pharmaceutically acceptable salts and stereoisomers thereof.
  • Some specific exemplary compounds herein are protonated salts of amine compounds.
  • the term "free form" refers to an amine compound in a non-salt form.
  • the pharmaceutically acceptable salts included include not only exemplary salts of the particular compounds described herein, but also all typical pharmaceutically acceptable salts of the free forms of the compounds of formula I.
  • the free form of the particular salt of the compound can be isolated using techniques known in the art.
  • the free form can be regenerated by treating the salt with a suitable dilute aqueous base such as a dilute aqueous solution of NaOH, a dilute aqueous solution of potassium carbonate, dilute aqueous ammonia, and a dilute aqueous solution of sodium bicarbonate.
  • a suitable dilute aqueous base such as a dilute aqueous solution of NaOH, a dilute aqueous solution of potassium carbonate, dilute aqueous ammonia, and a dilute aqueous solution of sodium bicarbonate.
  • the free form differs somewhat from its respective salt form in solubility in certain physical properties, such as in polar solvents, but for purposes of the invention such acid and base salts are otherwise pharmaceutically equivalent to their respective free forms.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the compounds of the present invention containing a basic moiety or an acidic moiety by conventional chemical methods.
  • the salt of the basic compound is prepared by ion exchange chromatography or by reaction of the free base with a stoichiometric or excess amount of the desired salt or mixture of the inorganic or organic acid in a suitable solvent or combination of solvents.
  • a salt of an acidic compound is formed by reaction with a suitable inorganic or organic base.
  • pharmaceutically acceptable salts of the compounds of the invention include the conventional non-toxic salts of the compounds of the invention which are formed by the reaction of a basic compound of the invention with an inorganic or organic acid.
  • conventional non-toxic salts include those derived from inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, phosphoric acid, nitric acid, and the like, and also include organic acids such as acetic acid, propionic acid, succinic acid, glycolic acid, and hard.
  • Fatty acid lactic acid, malic acid, tartaric acid, citric acid, ascorbic acid, pamoic acid, maleic acid, hydroxymaleic acid, phenylacetic acid, glutamic acid, benzoic acid, salicylic acid, p-aminobenzenesulfonic acid, 2-acetyl A salt prepared from oxy-benzoic acid, fumaric acid, toluenesulfonic acid, methanesulfonic acid, ethanedisulfonic acid, oxalic acid, isethionic acid, trifluoroacetic acid or the like.
  • a suitable "pharmaceutically acceptable salt” refers to a salt prepared by pharmaceutically acceptable non-toxic bases including inorganic bases and organic bases.
  • Salts derived from inorganic bases include aluminum salts, ammonium salts. Salt, calcium salt, copper salt, iron salt, ferrous salt, lithium salt, magnesium salt, manganese salt, manganese salt, potassium salt, sodium salt, zinc salt and the like. Ammonium salts, calcium salts, magnesium salts, potassium salts and sodium salts are particularly preferred.
  • a salt derived from a pharmaceutically acceptable organic non-toxic base comprising a salt of a primary, secondary and tertiary amine, the substituted amine comprising a naturally occurring substituted amine, a cyclic amine and a basic ion exchange resin such as a fine Amino acid, betaine, caffeine, choline, N,N'-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, aminoethanol, ethanolamine, B Diamine, N-ethylmorpholine, N-ethylpiperidine, glucosamine, glucosamine, histidine, hydroxycobalamin, isopropylamine, lysine, methylglucamine, morpholine, piperazine , piperidine, guanidine, polyamine resin, procaine, guanidine, theobromine, triethylamine, trimethylamine, tripropylamine, t
  • the acidic moiety such as a carboxyl group
  • a cationic moiety such as tetravalent
  • the compounds of the invention are potential internal salts or zwitterions.
  • the compounds of the invention can be prepared using the reactions as shown in the scheme below. Accordingly, the following illustrative schemes are for illustrative purposes and are not limited to the listed compounds or any particular substituents. The number of substituents shown in the schemes does not necessarily have to correspond to the number used in the claims, and for the sake of clarity, it is shown that the mono-substituents are attached to a compound which allows multiple substituents under the definition of formula I above.
  • R 7 is halogen, -OCOR 14 or OR 14 ,
  • R 14 is C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, C 6 -C 10 aryl or C 6 -C 10 aryl C 1 -C 6 alkyl, preferably C 1 -C 3 alkyl, halogenated C 1 -C 3 alkyl, phenyl or phenyl C 1 -C 3 alkyl.
  • the compound of the formula IV is reduced to obtain the compound of the formula III or a salt thereof, and the reduction reaction may be carried out by adding a reducing agent such as iron powder, zinc powder or stannous chloride; or it may be introduced in the presence of a hydrogenation catalyst. Hydrogen is reacted, and the hydrogenation catalyst may be palladium carbon, activated nickel or platinum dioxide; the solvent of the reduction reaction is one of acetic acid, hydrochloric acid, sulfuric acid, methanol, ethanol, water, ethyl acetate, acetonitrile and tetrahydrofuran or Several.
  • a reducing agent such as iron powder, zinc powder or stannous chloride
  • a hydrogenation catalyst may be palladium carbon, activated nickel or platinum dioxide
  • the solvent of the reduction reaction is one of acetic acid, hydrochloric acid, sulfuric acid, methanol, ethanol, water, ethyl acetate, acetonitrile and tetrahydrofuran or
  • the reaction of a compound of formula III or a salt thereof and a compound of formula A or a salt thereof is carried out in the presence of a condensing agent and a suitable base, in the presence or absence of a catalyst, in a suitable solvent.
  • the condensing agent is preferably selected from the group consisting of N,N'-dicyclohexylcarbodiimide (DCC), N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide or a hydrochloride thereof.
  • EDC EDC or EDC.HCl
  • carbonyl diimidazole CDI
  • N N'-diisopropylcarbodiimide
  • DIC O-benzotriazole-N, N, N', N' -tetramethylurea tetrafluoroborate
  • HATU O-(7-azobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • HBTU Benzotriazole-N,N,N',N'-tetramethylurea hexafluorophosphate
  • HBTU Benzotriazole-N,N,N',N'-tetramethylurea hexafluorophosphate
  • BOP phosphate
  • PyBOP benzotriazol-1-yl-oxytripyrrolidinyl
  • the catalyst is 1-hydroxy-benzotriazole (HOBt) Or 4-dimethylaminopyridine
  • the reaction of the compound of the formula III or a salt thereof and the compound of the formula B or a salt thereof is carried out in the presence of a base in a suitable solvent; preferably, the base is selected from the group consisting of pyridine, piperidine, pyrrolidine, imidazole, morpholine, N -methylmorpholine, quinoline, 4-dimethylaminopyridine, triethylamine, diethylamine, tri-n-butylamine, tripropylamine, diisopropylamine, diisopropylethylamine, sodium methoxide, Sodium ethoxide, Potassium tert-butoxide, butyl lithium, 1,8-diazacyclo[5,4,0]undecene-7, N-methylmorpholine, quinoline, 4-dimethylaminopyridine, sodium hydrogen, One or more of sodium hydroxide, potassium hydroxide, lithium hydroxide, sodium hydrogencarbonate, potassium hydrogencarbonate, sodium carbonate and
  • a halogenated hydrocarbon solvent other solvents, and combinations thereof;
  • the aromatic hydrocarbon solvent is one or more selected from the group consisting of benzene, toluene, xylene, and the like; and the ether solvent is selected from the group consisting of tetrahydrofuran and diethyl ether.
  • the halogenated hydrocarbon solvent is selected from the group consisting of dichloromethane and chloroform One or more of carbon tetrachloride and dichloroethane; the other solvent is selected from the group consisting of methanol, ethanol, ethylene glycol, n-hexane, cyclohexane, N,N-dimethylformamide, One or more of N,N-dimethylacetamide, dimethyl sulfoxide, N-methylpyrrolidone, acetone, acetonitrile, ethyl acetate, water; the temperature of the reaction is preferably from -30 ° C to 150 ° C ,better Between -10 °C ⁇ 120 °C; the reaction time is preferably between 10 minutes to 24 hours.
  • the salt of the compound of formula III, the salt of the compound of formula A and the salt of the compound of formula B are selected from mineral acid salts or organic acid salts, such as hydrochlorides, sulfates, hydrobromides, phosphates, nitrates, In acid salt, maleate salt, p-toluenesulfonate, methanesulfonate or trifluoroacetic acid.
  • the application provides a method for treating a proliferative disease or condition, such as a tumor in a human or other mammal, using a compound of Formula I, and a pharmaceutically acceptable salt thereof.
  • the compounds designed herein and pharmaceutically acceptable salts thereof can be used to treat or control non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer , liver cancer, skin cancer, epithelial cell carcinoma, gastrointestinal stromal tumor, leukemia, histiocytic lymphoma, nasopharyngeal carcinoma, head and neck cancer, colon cancer, rectal cancer, glioma and other transitional proliferative diseases.
  • Metabolites of the compounds and pharmaceutically acceptable salts thereof, and prodrugs which can be converted in vivo to the structures of the compounds and pharmaceutically acceptable salts thereof, are also included in the claims of the present application. in.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an active ingredient in a safe and effective amount, together with a pharmaceutically acceptable carrier.
  • active ingredient refers to a compound of formula I as described herein.
  • compositions of the present invention are useful as EGFR and IGF1R protease inhibitors.
  • a medicament for the preparation and prevention of a tumor is prepared.
  • the pharmaceutical compositions contain from 1 to 2000 mg of active ingredient per dose, more preferably from 10 to 200 mg of active ingredient per dose.
  • the "one dose” is a tablet.
  • “Pharmaceutically acceptable carrier” means: one or more compatible solid or liquid fillers or gel materials which are suitable for human use and which must be of sufficient purity and of sufficiently low toxicity.
  • composition capable of intermingling with the active ingredients of the present invention and with respect to each other without significantly reducing the efficacy of the active ingredients.
  • Examples of pharmaceutically acceptable carriers are cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid). , magnesium stearate), calcium sulfate, vegetable oil (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyol (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifier (such as ), a wetting agent (such as sodium lauryl sulfate), a coloring agent, a flavoring agent, a stabilizer, an antioxidant, a preservative, a pyrogen-free water, and the like.
  • cellulose and its derivatives such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.
  • gelatin such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate,
  • the compound of the formula I of the present invention can be formed by non-bonding with a macromolecular compound or a polymer. Compound.
  • the compound of the formula I of the present invention can be attached to a macromolecular compound or a polymer as a small molecule by a chemical bond.
  • the macromolecular compound may be a biological macromolecule such as a polysaccharide, a protein, a nucleic acid, a polypeptide, or the like.
  • the administration form of the active ingredient or the pharmaceutical composition of the present invention is not particularly limited, and representative administration forms include, but are not limited to, oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous) and the like.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active ingredient is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or mixed with the following ingredients:
  • a filler or a compatibilizer for example, starch, lactose, sucrose, glucose, mannitol, and silicic acid;
  • binders such as, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia;
  • a humectant for example, glycerin
  • a disintegrant such as agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate;
  • an absorption accelerator for example, a quaternary amine compound
  • an adsorbent for example, kaolin
  • a lubricant such as talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate, or a mixture thereof.
  • the dosage form may also contain a buffer.
  • the solid dosage forms can also be prepared with coatings and shell materials, such as casings and other materials known in the art. They may contain opacifying agents and the release of the active ingredient in such compositions may be released in a portion of the digestive tract in a delayed manner. Examples of embedding components that can be employed are polymeric and waxy materials.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or elixirs.
  • the liquid dosage form may contain inert diluents conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or a mixture of these substances.
  • the compositions may contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfumes.
  • the suspension may contain suspending agents, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar or mixtures of these and the like.
  • suspending agents for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar or mixtures of these and the like.
  • compositions for parenteral injection may comprise a physiologically acceptable sterile aqueous or nonaqueous solution, dispersion, suspension or emulsion, and a sterile powder for reconstitution into a sterile injectable solution or dispersion.
  • Suitable aqueous and nonaqueous vehicles, diluents, solvents or vehicles include water, ethanol, polyols, and suitable mixtures thereof.
  • the compounds of the invention may be administered alone or in combination with other therapeutic agents such as hypoglycemic agents.
  • a safe and effective amount of a compound of the invention is administered to a mammal (e.g., a human) in need of treatment wherein the dosage is a pharmaceutically effective effective dosage, for a 60 kg body weight
  • the dose to be administered is usually from 1 to 2000 mg, preferably from 20 to 500 mg.
  • specific doses should also consider factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
  • the compounds of formula I can be combined with other agents known to treat or ameliorate similar conditions.
  • the mode of administration and dosage of the original drug remain unchanged while the compound of formula I is administered simultaneously or subsequently.
  • a pharmaceutical composition comprising both one or several known drugs and a compound of formula I.
  • medicine The combination of the compounds also includes the administration of a compound of formula I with one or more other known drugs over an overlapping period of time.
  • the dose of the compound of formula I or a known drug may be lower than when they are administered alone.
  • Drugs or active ingredients which may be combined with a compound of formula I include, but are not limited to:
  • the pharmaceutical or active ingredient that can be administered in combination with a compound of Formula I includes, but is not limited to, adiponectin, alendronate, interferon, atrexine, allopurinol, allopurinol Sodium, palonosetron hydrochloride, hexamethylene melamine, aminoglumidine, amifostine, amrubicin, amsacrine, anatoxazole, dolasetron, aranesp, arglabin, arsenic trioxide, Arnoldin, 5-azacytidine, azathioprine, BCG or tici BCG, betahidine, betamethasone acetate, betamethasone sodium phosphate, bexarotene, bleomycin sulfate, bromide, Bortezomib, busulfan, calcitonin, alemtuzumab injection, capecitabine, carboplatin, constance, cefesone, simmein, daun
  • This kind of compound can effectively inhibit the growth of a variety of tumor cells, and has an inhibitory effect on EGFR and IGF1R protease, and can be used for preparing an antitumor drug.
  • This kind of compound can overcome the resistance of the existing drugs gefitinib, erlotinib and the like, and is selective for wild-type non-small cell lung cancer, and has good pharmacokinetic properties.
  • the synthesis method for synthesizing the intermediate 6 from the starting material 1 is as in Step 1 of Example 1.
  • the synthesis method of the starting material 8 synthesis intermediate 9 is as in Step 1-2 of Example 1.
  • Step 1 N 2 -(2-Methoxy-5-nitrophenyl)-N 4 -(naphthalen-2-yl)-5-isopropenylpyrimidine-2,4-diamine (Intermediate 10)
  • the synthesis method of the starting material 1 synthesis intermediate 19 is as in Step 1-2 of Example 1.
  • the synthesis method of the starting material 1 of the starting material 1 was as in Example 41.
  • the enzyme reaction substrate poly(glutamate, tyrosine) 4:1 (Sigma) was diluted with potassium-free PBS (10 mM sodium phosphate buffer, 150 mM NaCl, pH 7.2-7.4) to 20 ⁇ g/mL, 125 ⁇ L.
  • the wells were coated with the ELISA plate and reacted at 37 ° C for 12-16 hours. After discarding the liquid in the well, the plate was washed three times with 200 ⁇ L/well of T-PBS (PBS containing 0.1% Tween-20) for 5 minutes each, and then the plate was dried in an oven at 37 ° C for 1-2 hours.
  • reaction buffer 50 mM HEPES pH 7.4, 50 mM MgCl 2 , 5 mM MnCl 2 , 0.2 mM Na 3 VO 4 , 1 mM DTT
  • reaction buffer 50 mM HEPES pH 7.4, 50 mM MgCl 2 , 5 mM MnCl 2 , 0.2 mM Na 3 VO 4 , 1 mM DTT
  • Compounds were diluted to the appropriate concentration with 1% DMSO, 10 ⁇ L per well, and different types of tyrosine kinase proteins (including EGFR wt , EGFR T790M/L858R and IGF-1R) diluted in 40 ⁇ L of reaction buffer were added. Since Millipore).
  • the above reaction system was reacted at 37 ° C on a shaker (100 rpm) for 1 hour. No enzyme control wells and DMSO solvent control wells were required for each experiment. After the reaction was completed, the plate was washed three times with T-PBS. Add anti-phosphotyrosine primary antibody PY99 (Santa Cruz) 100 ⁇ L / well, antibody diluted with BSA 5 mg / mL T-PBS 1:1000), shake at 37 ° C for 0.5 hours, then wash with T-PBS Plate three times.
  • IC 50 values by fitting a four-parameter is calculated in inhibition curves.
  • Some compounds show potent kinase inhibitory activity, which is 20-100 times more potent against EGFR T790M/L858R than EGFR WT , and against EGFR T790M
  • the inhibitory activity of /L858R was stronger than the positive control compounds CO1686 and AZD-9291 (EGFR inhibitor).
  • human epidermal carcinoma A431 cell line and human non-small cell lung cancer NCI-H1975 cell line were purchased from the American Standard Biological Collection Center (ATCC).
  • the sulforhodamine B (SRB) method was as follows: a certain number of different tumor cells in the logarithmic growth phase were inoculated into a 96-well culture plate, and after 24 hours of cell attachment, different concentrations of the present invention were added. For the test compound, three replicate wells were set for each concentration, and the corresponding concentration of DMSO solvent control and cell-free zero-adjustment were set. After treating the cells with the drug for 72 hours, the culture solution was decanted, and the cells were fixed by adding 100 ⁇ L of ice-cold 10% trichloroacetic acid solution, and allowed to stand at 4 ° C for 1 hour, and then washed 5 times with distilled water, and naturally dried in the air.
  • SRB sulforhodamine B
  • Inhibition rate (%) (OD control - OD dosing) / OD control x 100%.
  • the half-inhibitory concentration (IC 50 ) value was calculated based on the growth inhibitory effect of the 2-aminopyrimidine compound on these cells, as shown in Table 2.
  • Detection was carried out using a conventional Western Blot (immunoblotting) as follows. A431 cells and NCI-H1975 cells in logarithmic growth phase were planted in 6-well plates in a certain number, and cultured overnight in an incubator. After serum-free medium was starved for 24 hours, a certain concentration of compound was added for 2 hours. EGF stimulating factor, 50 ng / mL for 10 min, lysate the cells with lysate. Then, an appropriate amount of sample was taken for SDS-PAGE electrophoresis.
  • the protein was transferred to a nitrocellulose membrane by a semi-dry electrotransfer system, and the nitrocellulose membrane was placed in a blocking solution (5% skim milk powder was diluted in 0.1% Tween 20). The cells were blocked at room temperature for 2 h in TBS), and then the membranes were separately placed in a primary antibody solution (1:500 diluted in TBS containing 0.1% Tween 20) and incubated overnight at 4 °C. Wash three times with TBS containing 0.1% Tween 20 for 15 min each time.
  • a blocking solution 5% skim milk powder was diluted in 0.1% Tween 20.
  • the cells were blocked at room temperature for 2 h in TBS), and then the membranes were separately placed in a primary antibody solution (1:500 diluted in TBS containing 0.1% Tween 20) and incubated overnight at 4 °C. Wash three times with TBS containing 0.1% Tween 20 for 15 min each time.
  • CCB120067 can significantly inhibit the phosphorylation of EGFR T790M/L858R and the activation of downstream signaling pathway proteins AKT and ERK in NCI-H1975 cells, and the inhibitory activity is superior to the positive control compound.
  • CO1686 As shown, CCB120067 inhibited the phosphorylated EGFR T790M/L858R and its downstream phosphorylated Akt and Erk at a concentration of 1 nM, which was comparable or slightly superior to the inhibitory activity at CO1686100 nM.
  • the compound CCB120067 has a weak inhibitory activity against phosphorylation of EGFR wt in A431 cells.
  • the tumor tissue in the vigorous growth period was cut into small pieces of about 1.5 mm 3 and inoculated subcutaneously under sterile conditions.
  • the nude mice were randomly grouped until the tumor grew to about 100 mm 3 .
  • the test compound CCB120067 was administered at a dose of 10 mg/kg, and the positive control compound CO1686 was administered at a dose of 100 mg/kg. Both were administered orally, once a day for 11 days, and a solvent control group (including 1% Tween) was established. 80 water for injection).
  • the diameter of the transplanted tumor was measured twice with a vernier caliper twice a week, and the body weight of the mice was weighed.
  • the formula for calculating tumor volume is:
  • CCB12006710 mg/kg administered by gavage significantly inhibited the growth of NCI-H1975 nude mice xenografts, and the tumor completely resolved after 7 days of administration.
  • CCB120067 had a tumor growth inhibitory activity at a dose of 10 mg/kg stronger than that of CO1686 at a dose of 100 mg/kg.

Abstract

本发明公开了一种2-氨基嘧啶类化合物及其药物组合物和应用,2-氨基嘧啶类化合物的结构如I所示,式中,R1、R2、R3、R4、R5、X、Y、Z、W、(i)的定义如说明书和权利要求书中所述,该类化合物可以有效抑制多种肿瘤细胞的生长,并对EGFR及IGF1R蛋白酶产生抑制作用,可用于制备抗肿瘤药物,并可以克服现有药物吉非替尼,厄洛替尼等诱发的耐药,对肿瘤尤其是野生型非小细胞肺癌具有选择性,具有良好的药代动力学性质。

Description

2-氨基嘧啶类化合物及其药物组合物和应用 技术领域
本发明属于化学医药领域,特别是涉及2-氨基嘧啶类化合物及其药物组合物和应用。
背景技术
肿瘤分子靶向治疗是基于对肿瘤生长密切相关的关键分子通过化学或生物学手段选择性杀伤肿瘤细胞的一种治疗方法。靶向治疗的特点为:特异性高,选择性强,毒副作用较轻;联合应用时,它可加强传统化疗、放疗的疗效,减少术后复发。肿瘤靶向治疗是肿瘤治疗的热点和发展趋势。
蛋白酪氨酸激酶(PTKs)是一类能够催化多种重要蛋白质的酪氨酸残基上的酚羟基发生磷酸化,进而激活功能蛋白的功能的蛋白质酶系。研究表明,半数以上的原癌基因和癌基因的激活都与蛋白酪氨酸激酶相关。以酪氨酸激酶为靶点进行抗肿瘤药物研发成为国际上的一个热点,也是各国药物开发机构研究投入的重点。
表皮生长因子受体(EGFR),一种受体酪氨酸蛋白激酶,调控了细胞的增殖,存活,粘连,迁移与分化。EGFR在多种肿瘤细胞中过度活化或持续活化,比如肺癌,乳腺癌,***癌等。阻断EGFR和Erb-B2的活化已被临床验证为主导的方法来靶向***细胞。两种靶向EGFR的小分子抑制剂,吉非替尼和厄洛替尼,得到了美国FDA的快速批准用于治疗晚期非小细胞肺癌(NSCLC)患者,这些患者对常规化疗已经失去反应。
已有多项前瞻性临床研究证实,EGFR活化突变阳性的NSCLC患者对EGFR-TKI(EGFR-酪氨酸激酶抑制剂)的反应率显著高于EGFR野生型NSCLC患者,无进展生存(PFS)期和总生存(OS)期也显著延长。但尽管如此,大部分EGFR突变阳性患者的PFS不超过12~14个月,即对TKI发生了耐药。获得性耐药的机制及其临床应对策略成为靶向治疗领域的又一研究热点。
针对耐药性,临床上采用的策略是:策略1—继续使用EGFR-TKI,吉非替尼和厄洛替尼的交叉使用。总之,TKI进展后继续使用TKI有一定益处,但获益程度非常有限。策略2—开发新型EGFR-TKI。策略3—针对其他靶点治疗。由于“旁路激活途径”在EGFR-TKI耐药中发挥重要作用,针对这些旁路的靶向药物不断涌现。但是,目前的EGFR-TKI仍不能解决药物耐药性所引起的临床压力,而且现有的药物多是以喹唑啉或者喹啉胺类为基本母核的EGFR可逆或不可逆抑制剂,其对野生型细胞的选择性差带来的毒副作用也是不可避免的。
因此,本领域迫切需要新类型的,尤其是新颖骨架的化合物来解决耐药性,选择性差,药代性质差等问题。
发明内容
本发明的目的在于提供一种结构新颖的2-氨基嘧啶类化合物及药物组合物和应用。
本发明的第一方面,提供一种式I化合物,其药学上可接受的盐或立体异构体或其前药分子,
Figure PCTCN2015094954-appb-000001
其中,R1和R2各自独立地为H、卤素、氰基、取代或未取代的C1-C6烷基、取代或未取代的C3-C6环烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C6环烷氧基;
R3为H或-(CH2)mNR8R9;R4和R5独立地为H、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、卤素、-(CH2)mNR8R9、-(CH2)mCR6R8R9;其中,各m独立地为0、1、2或3;R6为H或C1-C3烷基;各R8和各R9独立地选自H、取代或未取代的C1-C6烷基,或者R8、R9和相连的N或C一同形成未取代或取代的含有1、2或3个选自O、N、S杂原子的3-8元单环或稠环;
W为NH、N(C1-C3烷基)、O或S;
X、Y、Z各自独立地为N或-CR10,其中,R10为氢、卤素、取代或未取代的C1-C3烷基、取代或未取代的C1-C3烷氧基;
Figure PCTCN2015094954-appb-000002
为含有0、1、2或3个选自O、N或S的杂原子的取代或未取代的5-7元芳香环;
其中,各取代独立地指被选自下组的取代基取代:卤素、羟基、氨基、C1-C3烷基、C1-C3烷氧基、-NH(C1-C3烷基)、-N(C1-C3烷基)(C1-C3烷基)、-C(=O)(C1-C3烷基)。
在另一优选例中,所述盐选自无机酸盐或有机酸盐,如盐酸盐、硫酸盐、氢溴酸盐、磷酸盐、硝酸盐,乙酸盐、马来酸盐、对甲苯磺酸盐、甲磺酸盐或三氟乙酸。
在另一优选例中,所述各取代独立地为单取代、二取代、三取代或四取代。
在另一优选例中,所述未取代或取代的3-8元单环或稠环上,任选地具有1、2或3个选自O、N、S的杂原子,所述取代是指被选自下组的取代基取代:卤素、C1-C3烷基、-NH(C1-C3烷基)、-N(C1-C3烷基)(C1-C3烷基)、-C(=O)(C1-C3烷基)。
在另一优选例中,W为NH、O、S或-N(CH3)。
在另一优选例中,X为N或-CH-。
在另一优选例中,Y为-CH-。
在另一优选例中,Z为-CH-。
在另一优选例中,R1和R2各自独立地为H、卤素、氰基、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、C3-C6环烷氧基、卤代C1-C6烷基或卤代C1-C6烷氧基。
在另一优选例中,R1为H、氟、氯、溴、氰基、C1-C4烷基、氟代C1-C3烷基、氟代C1-C3烷氧基或C1-C3烷氧基。
在另一优选例中,氟代C1-C3烷基为三氟甲基。
在另一优选例中,
Figure PCTCN2015094954-appb-000003
为苯基、呋喃基、噻吩基、吡咯基、吡唑基、噻唑基、异噻唑基、噁唑基、异噁唑基、吡嗪基、哒嗪基、吡啶基、咪唑基或嘧啶基。
在另一优选例中,
Figure PCTCN2015094954-appb-000004
为苯基、吡咯基或吡啶基。
在另一优选例中,式I化合物具有以下一个或多个特征:
(1)R3为H、-(CH2)sN(C1-C3烷基)(C1-C3烷基)、或
Figure PCTCN2015094954-appb-000005
其中s为1或2或3;
(2)R4为H、C1-C3烷基、卤代C1-C3烷基、C1-C3烷氧基、卤代C1-C3烷氧基、卤素、-(CH2)mNR8R9、或
Figure PCTCN2015094954-appb-000006
(3)R5为H、C1-C3烷基、卤代C1-C3烷基、C1-C3烷氧基、卤代C1-C3烷氧基、卤素、-(CH2)mNR8R9
Figure PCTCN2015094954-appb-000007
其中,各m独立地为0、1、2或3;
各R8和各R9独立地选自H、取代或未取代的C1-C3烷基,所述取代是指被选自下组的取代基取代:卤素、-NH(C1-C3烷基)、-N(C1-C3烷基)(C1-C3烷基)、-C(=O)(C1-C3烷基);
各n1、各n2、各n3独立地为0、1、2或3;
各V独立地为CH、C(C1-C3烷基)或N;
各U独立地为无、O、S、CR11R12或NR13,其中R11、R12、R13独立地为H、C1-C3烷基、C3-C6环烷基、-NH(C1-C3烷基)、-N(C1-C3烷基)(C1-C3烷基)或-C(=O)(C1-C3烷基)。
在另一优选例中,
Figure PCTCN2015094954-appb-000008
在另一优选例中,式I化合物具有以下一个或多个特征:
(1)R3为H、
Figure PCTCN2015094954-appb-000009
(2)R4为H、-CH3
Figure PCTCN2015094954-appb-000010
(3)R5为H、-CH3、F、
Figure PCTCN2015094954-appb-000011
Figure PCTCN2015094954-appb-000012
在另一优选例中,W为NH;X、Y和Z为CH;
Figure PCTCN2015094954-appb-000013
为苯环,式I化合物的结构如式II所示:
Figure PCTCN2015094954-appb-000014
其中,R1、R2、R3、R4、R5的定义如前所述。
在另一优选例中,式I化合物中各取代基为实施例中各具体化合物中对应位置的取代基。
在另一优选例中,所述式I化合物为:
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((喹啉-6-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((喹啉-3-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((吲哚-5-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-(N-甲基-(萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((萘-2-基)氧基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((萘-2-基)硫基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-(4-((萘-2-基)氨基)嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氟-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-甲基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-甲氧基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氰基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-三氟甲基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-异丙基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-氟苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-乙氧基苯基)-4-(二甲氨基)-2-丁烯酰 胺;
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-异丙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-氟-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-氟苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-乙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-异丙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((5-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
(E)-N-((5-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-氟-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺;
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(哌啶-1-基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(吗啉-1-基)-2-丁烯酰胺;
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(4-乙酰基哌嗪-1-基)-2-丁烯酰胺;
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-吗啉甲基苯基)丙烯酰胺;
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-(二甲氨基甲基)苯基)丙烯酰胺;
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-((N-甲基-N-二甲氨基乙基)甲基胺)苯基)丙烯酰胺;
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-((4-甲基哌嗪-1-基)甲基)苯基)丙烯酰胺;
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-((4-甲基高哌嗪-1-基)甲基)苯基)丙烯酰胺;
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-((N-甲基-N-二甲氨基乙基)氨基)-4-甲氧基苯基)丙烯酰胺;
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-乙酰基哌嗪)-4-甲氧基苯基)丙烯酰胺;
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-甲基哌嗪)-4-甲氧基苯基)丙烯酰胺;
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-吗啉基-4-甲氧基苯基)丙烯酰胺;
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-甲基高哌嗪基)-4-甲氧基苯基)丙烯酰胺;
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-二甲氨基哌啶)-4-甲氧基苯基)丙烯酰胺;
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(3-二甲氨基吡咯烷-1-基)-4-甲氧基苯基)丙烯酰胺;
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(3-二甲氨基氮杂环丁烷-1-基)-4-甲氧基苯基)丙烯酰胺;或
N-((5-((5-氯-4-(萘-2-基氨基)嘧啶-2-基)氨基)-4-甲氧基-2-(甲基(2-甲基氨基)乙基)氨基)苯基)丙烯酰胺。
本发明的第二方面,提供第一方面所述的式I化合物的制备方法,采用式III化合物或其盐为原料,与式A化合物或其盐反应得到式I化合物,或者与式B化合物或其盐反应得到式I化合物,
Figure PCTCN2015094954-appb-000015
其中,R1、R2、R3、R4、R5、W、X、Y、Z和
Figure PCTCN2015094954-appb-000016
的定义同前;
R7为卤素、-OCOR14或OR14,R14为C1-C6烷基、卤代C1-C6烷基、C6-C10芳基或C6-C10芳基C1-C6烷基,优选为C1-C3烷基、卤代C1-C3烷基、苯基或苯基C1-C3烷基。
本发明的第三方面,提供一种药物组合物,包括:
本发明第一方面所述的式I化合物,其药学上可接受的盐或立体异构体或其前药分子;以及药学上可接受的载体。
本发明的第四方面,提供第一方面所述的式I化合物,其药学上可接受的盐或立体异构体或其前药分子,或者第三方面所述的药物组合物的用途,用作:
(1)制备***的药物;
(2)制备EGFR蛋白酶抑制剂的药物;
(3)制备IGF1R蛋白酶抑制剂的药物。
在另一优选例中,所述肿瘤选自:非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、***癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌、鼻咽癌、头颈部肿瘤、结肠癌、直肠癌、胶质瘤。
本发明的第五方面,提供一种***的方法,对需要的对象给予第一方面所述的式I 化合物,其药学上可接受的盐或立体异构体或其前药分子,或者第三方面所述的药物组合物。
本发明的第六方面,提供一种抑制EGFR蛋白酶的方法,对需要的对象给予第一方面所述的式I化合物,其药学上可接受的盐或立体异构体或其前药分子,或者第三方面所述的药物组合物。
本发明的第七方面,提供一种抑制IGF1R蛋白酶的方法,对需要的对象给予第一方面所述的式I化合物,其药学上可接受的盐或立体异构体或其前药分子,或者第三方面所述的药物组合物。
在另一优选例中,所述需要的对象为非人哺乳动物或人,较佳地,为人、小鼠或大鼠。
本发明的第八方面,提供一种式III化合物或其盐:
Figure PCTCN2015094954-appb-000017
其中,R1、R2、R4、R5
Figure PCTCN2015094954-appb-000018
W、X、Y和Z的定义同前,条件是式III化合物不包括如下结构:
Figure PCTCN2015094954-appb-000019
在另一优选例中,所述盐选自无机酸盐或有机酸盐,如盐酸盐、硫酸盐、氢溴酸盐、磷酸盐、硝酸盐,乙酸盐、马来酸盐、对甲苯磺酸盐、甲磺酸盐或三氟乙酸。
在一优选例中,
Figure PCTCN2015094954-appb-000020
为苯基、吡咯基或吡啶基。
在另一优选例中,
Figure PCTCN2015094954-appb-000021
本发明的式I化合物及其药学上可接受的盐,可以抑制多种肿瘤细胞的生长,对EGFR,Her家族蛋白酶产生抑制作用,尤其是能够选择性作用于EGFRL858R/T790M以及EGFRDel E745_A750肺癌细胞。对比野生型癌细胞,该类化合物的具有大于30倍的选择性。该类化合物可用于制备抗肿瘤药物,并可以克服现有药物吉非替尼,厄洛替尼等诱发的耐药,是一类新颖的能够克服现有EGFR酪氨酸激酶抑制剂耐药的并具有选择性和良好药代性质的蛋白激酶抑制剂。如本领域技术人员所理解的,本发明的化合物及其药学可接受的盐可用于制备治疗人类及其它哺乳动物的肿瘤等过度增殖性疾病。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。
附图说明
图1为化合物CCB120067及CO1686对含EGFRT790M/L858R突变的NCI-H1975细胞中EGFRT790M/L858R激酶及其下游信号通路蛋白磷酸化影响测试结果图。
图2为化合物CCB120067及CO1686对野生型EGFR高表达的A431细胞中EGFR激酶及其下游信号通路蛋白磷酸化影响测试结果图。
图3为化合物CCB120067及CO1686对人肺癌NCI-H1975裸小鼠皮下移植瘤的生长抑制作用结果图。
具体实施方式
本申请的发明人经过广泛而深入地研究,首次意外研发出一种结构新颖的2-氨基嘧啶类化合物,能够解决耐药性,选择性差,药代性质差等问题。在此基础上,完成了本发明。
本发明所述化合物中,当任何变量(例如R8、R9等)在任何组分中出现超过一次,则其每次出现的定义独立于其它每次出现的定义。同样,允许取代基及变量的组合,只要这种组合使化合物稳定。自取代基划入环***的线表示所指的键可连接到任何能取代的环原子上。如果环***为多环,其意味着这种键仅连接到邻近环的任何适当的碳原子上。要理解本领域普通技术人员可选择本发明化合物的取代基及取代型式而提供化学上稳定的并可通过本领域技术和下列提出的方法自可容易获得的原料容易的合成的化合物。如果取代基自身被超过一个基团取代,应理解这些基团可在相同碳原子上或不同碳原子上,只要使结构稳定。短语“被选自下组的取代基取代”被认为与短语“被至少一个取代基取代”相当,且在此情况下优选的实施方案将具有1-4个取代基。
本文所用术语“烷基”意指包括具有特定碳原子数目的支链的和直链的饱和脂肪烃基。例如,“C1-C6烷基”中“C1-C6”的定义包括以直链或支链排列的具有1、2、3、4、5或6个碳原子的基团。例如,“C1-C6烷基”具体包括甲基、乙基、正丙基、异丙基、正丁基、叔丁基、异丁基、戊基、己基。术语“环烷基”指具有特定碳原子数目的单环饱和脂肪烃基。例如“环烷基”包括环丙基、环丁基、环戊基或环己基等。术语“烷氧基”指具有-O-烷基结构的基团,如-OCH3、-OCH2CH3、-OCH2CH2CH3、-O-CH2CH(CH3)2、-OCH2CH2CH2CH3、-O-CH(CH3)2等。本文中,“含有0、1、2或3个选自O、N或S的杂原子的5~7元芳香环”是指苯环或杂芳环,本发明范围内的杂芳环包括但不限于:咪唑基、***基、吡唑基、呋喃基、噻吩基、噁唑基、异噁唑基、吡嗪基、哒嗪基、吡啶基、嘧啶基、吡咯基。
正如本领域技术人员所理解的,本文中所用“卤素”(“halo”)或“卤”意指氯、氟、溴和碘。
本发明提供一种式I化合物,其药学上可接受的盐或立体异构体或其前药分子,
Figure PCTCN2015094954-appb-000022
其中,R1、R2、R3、W、R4、R5、X、Y、Z、
Figure PCTCN2015094954-appb-000023
的定义同前。
本发明包括式Ⅰ化合物的游离形式,也包括其药学上可接受的盐及立体异构体。本文中一些特定的示例性化合物为胺类化合物的质子化的盐。术语“游离形式”指以非盐形式的胺类化合物。包括在内的药学上可接受盐不仅包括本文所述特定化合物的示例性盐,也包括所有式Ⅰ化合物游离形式的典型的药学上可接受的盐。可使用本领域已知技术分离所述化合物特定盐的游离形式。例如,可通过用适当的碱稀水溶液例如NaOH稀水溶液、碳酸钾稀水溶液、稀氨水及碳酸氢钠稀水溶液处理该盐使游离形式再生。游离形式在某些物理性质例如在极性溶剂中溶解度上与其各自盐形式多少有些区别,但是为发明的目的这种酸盐及碱盐在其它药学方面与其各自游离形式相当。
可通过常规化学方法自含有碱性部分或酸性部分的本发明化合物合成本发明的药学上可接受的盐。通常,通过离子交换色谱或通过游离碱和化学计算量或过量的所需盐形式的无机或有机酸在适当溶剂或多种溶剂的组合中反应制备碱性化合物的盐。类似的,通过和适当的无机或有机碱反应形成酸性化合物的盐。
因此,本发明化合物的药学上可接受的盐包括通过碱性本发明化合物和无机或有机酸反应形成的本发明化合物的常规无毒盐。例如,常规的无毒盐包括得自无机酸例如盐酸、氢溴酸、硫酸、氨基磺酸、磷酸、硝酸等的盐,也包括自有机酸例如乙酸、丙酸、琥珀酸、乙醇酸、硬脂酸、乳酸、苹果酸、酒石酸、柠檬酸、抗坏血酸、扑酸、马来酸、羟基马来酸、苯乙酸、谷氨酸、苯甲酸、水杨酸、对氨基苯磺酸、2-乙酰氧基一苯甲酸、富马酸、甲苯磺酸、甲磺酸、乙烷二磺酸、草酸、羟乙基磺酸、三氟乙酸等制备的盐。
如果本发明化合物为酸性的,则适当的“药学上可接受的盐”指通过药学上可接受的无毒碱包括无机碱及有机碱制备的盐.得自无机碱的盐包括铝盐、铵盐、钙盐、铜盐、铁盐、亚铁盐、锂盐、镁盐、锰盐、亚锰盐、钾盐、钠盐、锌盐等。特别优选铵盐、钙盐、镁盐、钾盐和钠盐。得自药学上可接受的有机无毒碱的盐,所述碱包括伯胺、仲胺和叔胺的盐,取代的胺包括天然存在的取代胺、环状胺及碱性离子交换树脂例如精氨酸、甜菜碱、咖啡因、胆碱、N,N'-二苄基乙二胺、二乙胺、2-二乙基氨基乙醇、2-二甲基氨基乙醇、氨基乙醇、乙醇胺、乙二胺、N-乙基吗啉、N-乙基哌啶、葡萄糖胺、氨基葡萄糖、组氨酸、羟钴胺、异丙基胺、赖氨酸、甲基葡萄糖胺、吗啉、哌嗪,哌啶、呱咤、多胺树脂、普鲁卡因、嘌呤、可可碱、三乙胺、三甲胺、三丙胺、氨基丁三醇等。
Berg等,“Pharmaceutical Salts”J.Pharm.Sci.’1977:66:1-19更详细描述了上文所述药学上可接受的盐及其它典型的药学上可接受的盐的制备。
由于在生理条件下化合物中脱质子化的酸性部分例如羧基可为阴离子的,而这种带有的电荷然后可被内部带有阳离子的质子化了的或烷基化的碱性部分例如四价氮原子平衡抵消,所以应注意本发明化合物是潜在的内盐或两性离子。
除在文献中已知的或在实验程序中例证的标准方法外,可采用如下列方案中显示的反应制备本发明化合物。因此,下列说明性方案是为说明的目的而不是局限于所列化合物或任何特定的取代基。方案中显示的取代基数目并不必需符合权利要求中所用的数目,且为清楚起见,显示单取代基连接到在上文中式I的定义下允许有多取代基的化合物上。
如发明所述式Ⅰ化合物可通过如下反应路线制备:
Figure PCTCN2015094954-appb-000024
其中,所述式A化合物和式B化合物的结构如下:
Figure PCTCN2015094954-appb-000025
其中,R1、R2、R3、R4、R5、W、X、Y、Z和
Figure PCTCN2015094954-appb-000026
的定义如前所述;
R7为卤素、-OCOR14或OR14,R14为C1-C6烷基、卤代C1-C6烷基、C6-C10芳基或C6-C10芳基C1-C6烷基,优选为C1-C3烷基、卤代C1-C3烷基、苯基或苯基C1-C3烷基。
还原
式IV化合物经还原得到式III化合物或其盐,所述还原反应的条件可以是:加入还原剂,例如铁粉、锌粉或氯化亚锡;也可以是在氢化催化剂存在条件下,通入氢气进行反应,氢化催化剂可以为钯碳、活性镍或二氧化铂等;所述还原反应的溶剂为醋酸、盐酸、硫酸、甲醇、乙醇、水、乙酸乙酯、乙腈和四氢呋喃中的一种或几种。
式III化合物或其盐和式A化合物或其盐的反应在缩合剂和合适的碱存在下,在有或无催化剂存在下,在合适的溶剂中进行。所述缩合剂优选为选自N,N'-二环己基碳二亚胺(DCC)、N-(3-二甲氨基丙基)-N'-乙基碳二亚胺或其盐酸盐(EDC或EDC.HCl),碳酰二咪唑(CDI)、N,N'-二异丙基碳二亚胺(DIC)、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯(TBTU)、O-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(HATU)、苯并三氮唑-N,N,N',N'-四甲基脲六氟磷酸酯(HBTU)、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐(BOP)和六氟磷酸苯并***-1-基-氧基三吡咯烷基(PyBOP)中的一种或几种;所述催化剂为1-羟基-苯并三氮唑(HOBt)或4-二甲基氨基吡啶(DMAP);所述碱优选为选自三乙胺、二乙胺、三正丁胺、三丙基胺、二异丙基胺、二异丙基乙胺(DIPEA)、三甲胺、吡啶、2,6-二甲基吡啶、4-二甲氨基吡啶、哌啶、吡咯烷、喹啉、吗啉、N-甲基吗啉(NMM)、N-乙基吗啉、二异丙基胺、二异丙基乙胺、1,8-二氮杂环[5,4,0]十一烯-7和1,5-二氮杂二环[4.3.0]-壬-5-烯中的一种或几种;反应溶剂优选选自苯、二甲苯、甲苯、二氯甲烷、氯仿、四氢呋喃、***、丙酮、1,4-二氧六环、N,N-二甲基甲酰胺、N,N-二乙基甲酰胺、N,N-二甲基乙酰胺、乙腈、二甲亚砜和上述溶剂的混合物,更优选为四氢呋喃、乙腈、二氯甲烷、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、二甲亚砜和上述溶剂的混合物;反应温度优选在-20℃~200℃,更优选-10℃~100℃之间。
式III化合物或其盐和式B化合物或其盐的反应在碱存在下,在适当的溶剂中进行;优选地,所述碱为选自吡啶、哌啶、吡咯烷、咪唑、吗啉、N-甲基吗啉、喹啉、4-二甲氨基吡啶、三乙胺、二乙胺、三正丁胺、三丙基胺、二异丙基胺、二异丙基乙胺、甲醇钠、乙醇钠、 叔丁醇钾、丁基锂、1,8-二氮杂环[5,4,0]十一烯-7、N-甲基吗啉、喹啉、4-二甲氨基吡啶、钠氢、氢氧化钠、氢氧化钾、氢氧化锂、碳酸氢钠、碳酸氢钾、碳酸钠和碳酸钾中的一种或几种;优选地,所述反应的溶剂选自芳烃类溶剂、醚类溶剂、卤代烃类溶剂、其他溶剂和其组合;优选地,所述芳烃类溶剂为选自苯、甲苯、二甲苯等中的一种或几种;所述醚类溶剂为选自四氢呋喃、***、乙二醇二甲醚、二乙二醇二甲醚、乙二醇单甲醚、二氧六环中的一种或几种;所述卤代烃类溶剂为选自二氯甲烷、氯仿、四氯化碳、二氯乙烷中的一种或几种;所述其他溶剂为选自甲醇、乙醇、乙二醇、正己烷、环己烷、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、二甲亚砜、N-甲基吡咯烷酮、丙酮、乙腈、乙酸乙酯、水中的一种或几种;所述反应的温度优选在-30℃~150℃,更优选-10℃~120℃之间;所述反应的时间优选在10分钟~24小时之间。
所述式III化合物的盐、式A化合物的盐和式B化合物的盐选自无机酸盐或有机酸盐中,如盐酸盐、硫酸盐、氢溴酸盐、磷酸盐、硝酸盐,乙酸盐、马来酸盐、对甲苯磺酸盐、甲磺酸盐或三氟乙酸中。
在一个实施方案中,本申请提供了一种利用具有式Ⅰ的化合物及其药学可接受的盐治疗人或其它哺乳动物肿瘤等过渡增殖性疾病或症状。
在一个实施方案中,本申请所设计的化合物及其药学可接受的盐可以用于治疗或控制非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、***癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌、鼻咽癌、头颈部肿瘤、结肠癌、直肠癌、胶质瘤等过渡增殖性疾病。
药物代谢物及前药
本申请所涉及的化合物及其药学可接受的盐的代谢产物,以及可以在体内转变为本申请所涉及的化合物及其药学可接受的盐的结构的前药,也包含在本申请的权利要求中。
药物组合物
本发明还提供了一种药物组合物,它包含安全有效量范围内的活性成分,以及药学上可接受的载体。
本发明所述的“活性成分”是指本发明所述的式I化合物。
本发明所述的“活性成分”和药物组合物可用作EGFR及IGF1R蛋白酶抑制剂。在另一优选例中,用于制备预防和/***的药物。
“安全有效量”指的是:活性成分的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg活性成分/剂,更佳地,含有10-200mg活性成分/剂。较佳地,所述的“一剂”为一个药片。
“药学上可接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。
“相容性”在此指的是组合物中各组份能和本发明的活性成分以及它们之间相互掺和,而不明显降低活性成分的药效。
药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如
Figure PCTCN2015094954-appb-000027
)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
在另一优选例中,本发明式I化合物可与大分子化合物或高分子通过非键合作用形成复 合物。在另一优选例中,本发明式I化合物作为小分子还可通过化学键与大分子化合物或高分子相连接。所述大分子化合物可以是生物大分子如高聚糖、蛋白、核酸、多肽等。
本发明的活性成分或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、瘤内、直肠、肠胃外(静脉内、肌肉内或皮下)等。
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。
在这些固体剂型中,活性成分与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:
(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;
(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和***胶;
(c)保湿剂,例如,甘油;
(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;
(e)缓溶剂,例如石蜡;
(f)吸收加速剂,例如,季胺化合物;
(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;
(h)吸附剂,例如,高岭土;和
(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。
所述的固体剂型还可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性成分的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性成分外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。
除了活性成分外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。
本发明化合物可以单独给药,或者与其他治疗药物(如降糖药)联合给药。
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选20~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。
联合用药
式Ⅰ化合物可以与已知的治疗或改进相似病状的其它药物联用。联合给药时,原来药物的给药方式和剂量保持不变,而同时或随后服用式Ⅰ化合物。当式Ⅰ化合物与其它一种或几种药物同时服用时,优选使用同时含有一种或几种已知药物和式I化合物的药用组合物。药 物联用也包括在重叠的时间段服用式Ⅰ化合物与其它一种或几种已知药物。当式Ⅰ化合物与其它一种或几种药物进行药物联用时,式Ⅰ化合物或已知药物的剂量可能比它们单独用药时的剂量较低。
可以与式Ⅰ化合物进行药物联用的药物或活性成分包括但不局限为:
***受体调节剂、雄激素受体调节剂、视网膜样受体调节剂、细胞毒素/细胞抑制剂、抗增殖剂、蛋白转移酶抑制剂、HMG-CoA还原酶抑制剂、HIV蛋白激酶抑制剂、逆转录酶抑制剂、血管生成抑制剂、细胞增殖及生存信号抑制剂、干扰细胞周期关卡的药物和细胞凋亡诱导剂,细胞毒类药物、酪氨酸蛋白抑制剂、EGFR抑制剂、VEGFR抑制剂、丝氨酸/苏氨酸蛋白抑制剂、Bcr-Abl抑制剂、c-Kit抑制剂、Met抑制剂、Raf抑制剂、MEK抑制剂、MMP抑制剂、拓扑异构酶抑制剂、组氨酸去乙酰化酶抑制剂、蛋白酶体抑制剂、CDK抑制剂、Bcl-2家族蛋白抑制剂、MDM2家族蛋白抑制剂、IAP家族蛋白抑制剂、STAT家族蛋白抑制剂、PI3K抑制剂、AKT抑制剂、整联蛋白阻滞剂、干扰素-α、白介素-12、COX-2抑制剂、p53、p53激活剂、VEGF抗体、EGF抗体等。
在一个实施方案中,可以与式Ⅰ化合物进行药物联用的药物或活性成分包括但不局限为:阿地白介素、阿仑膦酸、干扰素、阿曲诺英、别嘌醇、别嘌醇钠、帕洛诺司琼盐酸盐、六甲蜜胺、氨基格鲁米特、氨磷汀、氨柔比星、安丫啶、阿纳托唑、多拉司琼、aranesp、arglabin、三氧化二砷、阿诺新、5-氮胞苷、硫唑嘌呤、卡介苗或tice卡介苗、贝他定、醋酸倍他米松、倍他米松磷酸钠制剂、贝沙罗汀、硫酸博来霉素、溴尿甘、bortezomib、白消安、降钙素、阿来佐单抗注射剂、卡培他滨、卡铂、康士得、cefesone、西莫白介素、柔红霉素、苯丁酸氮芥、顺铂、克拉屈滨、克拉屈滨、氯屈磷酸、环磷酰胺、阿糖胞昔、达卡巴嗪、放线菌素D、柔红霉素脂质体、***、磷酸***、戊酸***、地尼白介素2、狄波美、地洛瑞林、地拉佐生、己烯雌酚、大扶康、多西他奇、去氧氟尿苷、阿霉素、屈***酚、钦-166-壳聚糖复合物、eligard、拉布立酶、盐酸表柔比星、阿瑞吡坦、表阿霉素、阿法依伯汀、红细胞生成素、依铂、左旋咪唑片、***制剂、17-β-***、雌莫司汀磷酸钠、炔雌醇、氨磷汀、羟磷酸、凡毕复、依托泊甙、法倔唑、他莫昔芬制剂、非格司亭、非那司提、非雷司替、氟尿苷、氟康唑、氟达拉滨、5-氟脱氧尿嘧啶核苷一磷酸盐、5-氟尿嘧啶、氟***、氟他胺、福麦斯坦、1-β-D-阿糖呋喃糖胞噻啶-5’-硬脂酰磷酸酯、福莫司汀、氟维司群、丙种球蛋白、吉西他滨、吉妥单抗、甲磺酸伊马替尼、卡氮芥糯米纸胶囊剂、戈舍瑞林、盐酸格拉尼西隆、组氨瑞林、和美新、氢化可的松、赤型-羟基壬基腺嘌呤、羟基脲、替坦异贝莫单抗、伊达比星、异环磷酰胺、干扰素α、干扰素-α2、干扰素α-2A、干扰素α-2B、干扰素α-nl、干扰素α-n3、干扰素β、干扰素γ-la、白细胞介素-2、内含子A、易瑞沙、依立替康、凯特瑞、硫酸香菇多糖、来曲唑、甲酰四氢叶酸、亮丙瑞林、亮丙瑞林醋酸盐、左旋四咪唑、左旋亚叶酸钙盐、左甲状腺素钠、左甲状腺素钠制剂、洛莫司汀、氯尼达明、屈***酚、氮芥、甲钴胺、甲羟孕酮醋酸酯、醋酸甲地孕酮、美法仑、酯化***、6-琉基嘌呤、美司钠、氨甲蝶呤、氨基乙酰丙酸甲酯、米替福新、美满霉素、丝裂霉素C、米托坦、米托葱醌、曲洛司坦、柠檬酸阿霉素脂质体、奈达铂、聚乙二醇化非格司亭、奥普瑞白介素、neupogen、尼鲁米特、三苯氧胺、NSC-631570、重组人白细胞介素1-β、奥曲肽、盐酸奥丹西隆、去氢氢化可的松口服溶液剂、奥沙利铂、紫杉醇、***磷酸钠制剂、培门冬酶、派罗欣、喷司他丁、溶链菌制剂、盐酸匹鲁卡品、毗柔比星、普卡霉素、卟吩姆钠、泼尼莫司汀、司替***龙、***、倍美力、丙卡巴脐、重组人类红细胞生成素、雷替曲塞、利比、依替膦酸铼-186、美罗华、力度伸-A、罗莫肽、盐酸毛果芸香碱片剂、奥曲肽、沙莫司亭、司莫司汀、西佐喃、索布佐生、唬钠甲强龙、帕福斯酸、干细胞治疗、链佐星、氯化锶-89、左旋甲状 腺素钠、他莫昔芬、坦舒洛辛、他索那明、tastolactone、泰索帝、替西硫津、替莫唑胺、替尼泊苷、丙酸睾酮、***、硫鸟嘌呤、噻替哌、促甲状腺激素、替鲁膦酸、拓扑替康、托瑞米芬、托西莫单抗、曲妥珠单抗、曲奥舒凡、维A酸、甲氨喋呤片剂、三甲基密胺、三甲曲沙、乙酸曲普瑞林、双羟萘酸曲普瑞林、优福定、尿苷、戊柔比星、维司力农、长春碱、长春新碱、长春酰胺、长春瑞滨、维鲁利秦、右旋丙亚胺、净司他丁斯酯、枢复宁、紫杉醇蛋白质稳定制剂、acolbifene、干扰素r-lb、affinitak、氨基喋呤、阿佐昔芬、asoprisnil、阿他美坦、阿曲生坦、BAY43-9006、阿瓦斯丁、CCI-779、CDC-501、西乐葆、西妥昔单抗、克立那托、环丙孕酮醋酸酯、地西他滨、DN-101、阿霉素-MTC、dSLIM、度他雄胺、edotecarin、依氟鸟氨酸、依喜替康、芬维A胺、组胺二盐酸盐、组氨瑞林水凝胶植入物、钬-166DOTMP、伊班膦酸、干扰素γ、内含子-PEG、ixabepilone、匙孔形血蓝蛋白、L-651582、兰乐肽、拉索昔芬、libra、lonafamib、米泼昔芬、米诺屈酸酯、MS-209、脂质体MTP-PE、MX-6、那法瑞林、奈莫柔比星、新伐司他、诺拉曲特、奥利默森、onco-TCS、osidem、紫杉醇聚谷氨酸酯、帛米酸钠、PN-401、QS-21、夸西洋、R-1549、雷洛昔芬、豹蛙酶、13-顺维A酸、沙铂、西奥骨化醇、T-138067、tarceva、二十二碳六烯酸紫杉醇、胸腺素αl、嘎唑呋林、tipifarnib、替拉扎明、TLK-286、托瑞米芬、反式MID-lo7R、伐司朴达、伐普肽、vatalanib、维替泊芬、长春氟宁、Z-100和唑来麟酸或它们的组合。
本发明提到的上述特征,或实施例提到的特征可以任意组合。本案说明书所揭示的所有特征可与任何组合物形式并用,说明书中所揭示的各个特征,可以被任何提供相同、均等或相似目的的替代性特征取代。因此除有特别说明,所揭示的特征仅为均等或相似特征的一般性例子。
本发明的有益之处在于:
(1)提供一种结构新颖的2-氨基嘧啶类化合物。
(2)该类化合物可以有效抑制多种肿瘤细胞的生长,并对EGFR及IGF1R蛋白酶产生抑制作用,可用于制备抗肿瘤药物。
(3)该类化合物可以克服现有药物吉非替尼,厄洛替尼等诱发的耐药,对野生型非小细胞肺癌具有选择性,具有良好的药代动力学性质。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。
除非另行定义,文中所使用的所有专业与科学用语与本领域熟练人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本发明方法中。文中所述的较佳实施方法与材料仅作示范之用。
实施例1
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB118563)
Figure PCTCN2015094954-appb-000028
步骤1.2,5-二氯-N-(萘-2-基)嘧啶-4-胺(2)
2,4,5-三氯嘧啶(6g,32.7mmol),2-萘胺(4.92g,34.4mmol),碳酸钠(6.94g,65.4mmol)溶于无水乙醇(100mL)中,室温搅拌过夜。搅拌下加入冰水(300mL),大量固体析出。减压过滤,真空干燥得棕色固体(8.38g,收率88%)。
1H NMR(400MHz,DMSO-d6):δ9.74(s,1H),8.42(s,1H),8.09(d,J=1.6Hz,1H),7.95-7.86(m,3H),7.73(d,J=8.8Hz,1H),7.54-7.47(m,2H).
MS(ESI):m/z 291[M+H]+.
步骤2.5-氯-N2-(2-甲氧基-5-硝基苯基)-N4-(萘基-2-基)嘧啶-2,4-二胺(3)
于封管中加入2,5-二氯-N-(萘-2-基)嘧啶-4-胺(2)(3g,10.3mmol),2-甲氧基-5-硝基苯胺(1.91g,11.4mmol),对甲苯磺酸一水合物(2.95g,15.5mmol),加入无水仲丁醇(30mL),密封,120℃搅拌过夜。冷却至室温,加入10%NaHCO3水溶液,二氯甲烷萃取,旋至几乎干,加入无水乙醇打浆,抽滤,真空干燥得黄色固体(3.96g,收率:91%)。
1H NMR(400MHz,DMSO-d6):δ9.09(s,1H),8.84(d,J=2.8Hz,1H),8.27(s,2H),8.21(s,1H),7.95(dd,J=8.8Hz,2.8Hz,1H),7.83(d,J=7.2Hz,1H),7.80(s,2H),7.68(d,J=8.8Hz,1H),7.46-7.39(m,2H),7.23(d,J=8.8Hz,1H),3.95(s,3H).
MS(ESI):m/z 422[M+H]+.
步骤3.N2-(5-氨基-2-甲氧基苯基)-5-氯-N4-(萘-2-基)嘧啶-2,4-二胺(4)
5-氯-N2-(2-甲氧基-5-硝基苯基)-N4-(萘基-2-基)嘧啶-2,4-二胺(3)(1g,2.4mmol),铁粉(1.33g,24mmol)溶于乙醇(12mL)中,加入氯化铵饱和溶液(1mL),加热回流搅拌2h。冷却至室温。硅藻土抽滤,旋干,柱层析分离得固体(705mg,收率:75%)。
1H NMR(400MHz,DMSO-d6):δ8.95(s,1H),8.25(s,1H),8.13(s,1H),7.89-7.78(m,5H),7.49-7.40(m,2H),7.15(d,J=2.4Hz,1H),6.73(d,J=8.8Hz,1H),6.24(dd,J=8.8Hz,2.4Hz,1H),4.26(br,2H),3.67(s,3H).
MS(ESI):m/z 392[M+H]+.
步骤4.(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB118563)
N2-(5-氨基-2-甲氧基苯基)-5-氯-N4-(萘-2-基)嘧啶-2,4-二胺(4)(500mg,1.28mmol),(E)-4-(二甲氨基)丁-2-烯酸盐酸盐(254mg,1.53mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲 基脲六氟磷酸酯(727mg,1.91mmol)溶于乙腈(10mL)中,加入二异丙基乙胺(0.441mL),室温搅拌过夜。旋干,加入的10%NaHCO3溶液(10mL),二氯甲烷萃取三次,合并有机相,再用饱和食盐水洗涤一遍,旋干,柱层析分离得固体(397mg,收率:62%)。
1H NMR(400MHz,DMSO-d6):δ9.88(s,1H),8.88(s,1H),8.28(s,1H),8.15(s,2H),7.97(s,1H),7.82(d,J=8.8Hz,1H),7.77(m,2H),7.67(d,J=8.0Hz,1H),7.53(d,J=8.8Hz,1H),7.42-7.35(m,2H),7.00(d,J=8.8Hz,1H),6.68-6.61(m,1H),6.17(d,J=15.6Hz,1H),3.76(s,3H),3.01(d,J=5.6Hz,2H),2.14(s,6H).
MS(ESI):m/z 503[M+H]+.
实施例2
(E)-N-((3-((5-氯-4-((喹啉-6-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145213)
Figure PCTCN2015094954-appb-000029
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.85(s,1H),9.00(s,1H),8.73(s,1H),8.39(s,1H),8.27(s,1H),8.18(s,1H),8.07-8.01(m,2H),7.98(s,1H),7.87(d,J=8.8Hz,2H),7.39(m,1H),7.02(d,J=8.8Hz,1H),6.64-6.58(m,1H),6.12(d,J=15.6Hz,1H),3.76(s,3H),3.00(d,J=4.8Hz,2H),2.14(s,6H).MS(ESI);m/z 504[M+H]+.
实施例3
(E)-N-((3-((5-氯-4-((喹啉-3-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145221)
Figure PCTCN2015094954-appb-000030
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.81(s,1H),9.17(s,1H),9.15(d,J=2.4Hz,1H),8.71(d,J=2.0Hz,1H),8.28(s,1H),8.19(s,1H),7.97(d,J=2.0Hz,1H),7.91(d,J=8.8Hz,1H),7.69(d,J=7.6Hz,1H),7.60(t,J=8.0Hz,1H),7.56(dd,J=8.8Hz,2.4Hz,1H),7.49(t,J=8.0Hz,1H),7.01(d,J=8.8Hz,1H),6.64-6.57(m,1H),6.13(d,J=15.6Hz,1H),3.76(s,3H),3.01(d,J=5.6Hz,2H),2.15(s,6H).MS(ESI):m/z 504[M+H]+.
实施例4
(E)-N-((3-((5-氯-4-((吲哚-5-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145231)
Figure PCTCN2015094954-appb-000031
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.77(s,1H),8.62(s,1H),8.07(s,1H),8.05(s,1H),7.75(s,1H),7.70(s,1H),7.45(d,J=8.8Hz,1H),7.30-7.24(m,3H),6.94(d,J=8.8Hz,1H),6.71-6.64(m,1H),6.34(s,1H),6.21(d,J=15.6Hz,1H),3.75(s,3H),3.04(d,J=6.0Hz,2H),2.17(s,6H).MS(ESI):m/z 492[M+H]+.
实施例5
(E)-N-((3-((5-氯-4-(N-甲基-(萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145295)
Figure PCTCN2015094954-appb-000032
中间体5的合成步骤如下:
2,5-二氯-N-(萘-2-基)嘧啶-4-胺(2)(3g,10.3mmol),氢氧化钠(824mg,20.6mmol)溶于的四氢呋喃(40mL)中,冰浴下滴加碘甲烷(0.96mL,15.45mmol),升到室温搅拌2h。旋干,加入水(10mL),二氯甲烷萃取三次。无水Na2SO4干燥,旋干,柱层析分离得固体(1.57g,收率:50%)。
由中间体5合成终产物CCB145295的合成方法如实施例1中步骤2-4。
Figure PCTCN2015094954-appb-000033
1H NMR(400MHz,DMSO-d6)δ9.89(s,1H),8.47(s,1H),8.06(s,1H),8.01(s,1H),7.93(d,J=8.8Hz,1H),7.90(d,J=7.2Hz,1H),7.85(d,J=7.2Hz,1H),7.63(s,1H),7.48(m,2H),7.43(d,J=8.8Hz,1H),7.31(d,J=8.8Hz,1H),6.99(d,J=8.8Hz,1H), 6.71-6.64(m,1H),6.25(d,J=15.6Hz,1H),3.86(s,3H),3.53(s,3H),3.02(d,J=5.6Hz,2H),2.15(s,6H).MS(ESI):m/z 517[M+H]+.
实施例6
(E)-N-((3-((5-氯-4-((萘-2-基)氧基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145260)
Figure PCTCN2015094954-appb-000034
由起始原料1合成中间体6的合成方法如实施例1中步骤1。
其中,中间体7的合成步骤如下:
将中间体6(870mg,3mmol),2-甲氧基-5-硝基苯胺(504mg,3mmol),醋酸钯(13.5mg,0.06mmol),4,5-双二苯基膦-9,9-二甲基氧杂蒽(87mg,0.15mmol),碳酸铯(1.96g,6mmol)溶于二氧六环(20mL)中,氩气置换三次,加热回流搅拌过夜。自然回复室温,旋干,加入水(10mL),二氯甲烷萃取三次。无水Na2SO4干燥,旋干,柱层析分离得固体(1.04g,收率:82%)。
中间体7合成终产物CCB145260的合成方法如实施例1中步骤3-4。
Figure PCTCN2015094954-appb-000035
1H NMR(400MHz,DMSO-d6)δ9.77(s,1H),8.43(s,1H),8.23(s,1H),7.96(d,J=8.8Hz,1H),7.91(m,2H),7.84(s,1H),7.79(d,J=2.0Hz,1H),7.55-7.51(m,2H),7.46(dd,J=8.8Hz,2.0Hz,1H),7.33(dd,J=8.8Hz,2.0Hz,1H),6.87(d,J=8.8Hz,1H),6.71-6.65(m,1H),6.22(d,J=15.6Hz,1H),3.68(s,3H),3.01(d,J=6.0Hz,2H),2.18(s,6H).MS(ESI):m/z 504[M+H]+.
实施例7
(E)-N-((3-((5-氯-4-((萘-2-基)硫基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145291)
Figure PCTCN2015094954-appb-000036
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.73(s,1H),8.27(s,2H),8.16(s,1H),7.90(d,J=8.0Hz,2H),7.82(d,J=8.0Hz,1H),7.66(s,1H),7.60-7.54(m,3H),7.29(d,J=8.8Hz,1H),6.71-6.65(m,2H),6.20(d,J=15.6Hz,1H),3.57(s,3H),3.05(d,J=5.2Hz,2H),2.18(s,6H).MS(ESI):m/z 520[M+H]+.
实施例8
(E)-N-((3-(4-((萘-2-基)氨基)嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145242)
Figure PCTCN2015094954-appb-000037
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.84(s,1H),9.34(s,1H),8.72(s,1H),8.26-8.25(m,2H),7.85-7.84(m,2H),7.79-7.76(m,2H),7.52(s,1H),7.47-7.39(m,2H),7.21(s,1H),7.09(s,1H),6.70-6.63(m,1H),6.21(d,J=15.2Hz,1H),3.02(d,J=6.0Hz,2H),2.15(s,6H),1.91(s,3H).
MS(ESI):m/z 469[M+H]+.
实施例9
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145286)
Figure PCTCN2015094954-appb-000038
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.83(s,1H),8.61(s,1H),8.24(s,1H),8.22(s,1H),8.15(s,1H),7.96(s,1H),7.80-7.78(m,3H),7.67(d,J=7.6Hz,1H),7.51(d,J=8.8Hz,1H),7.43-7.35(m,2H),6.99(d,J=8.8Hz,1H),6.68-6.61(m,1H),6.15(d,J=15.6Hz,1H),3.75(s,3H),3.01(d,J=5.6Hz,2H),2.15(s,6H).
MS(ESI);m/z 547[M+H]+.
实施例10
(E)-N-((3-((5-氟-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145287)
Figure PCTCN2015094954-appb-000039
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.85(s,1H),9.50(s,1H),8.43(s,1H),8.12(d,J=3.6Hz,1H),8.07(d,J=2.4Hz,1H),7.97(s,1H),7.84(dd,J=8.8Hz,2.4Hz,1H),7.79-7.76(m,2H),7.67(d,J=7.6Hz,1H),7.53(dd,J=8.8Hz,2.4Hz,1H),7.40(t,J=6.8Hz,1H),7.35(t,J=6.8Hz,1H),7.00(d,J=8.8Hz,1H),6.67-6.60(m,1H),6.15(d,J=15.6Hz,1H),3.79(s,3H),3.00(d,J=5.6Hz,2H),2.14(s,6H).
MS(ESI);m/z 487[M+H]+.
实施例11
(E)-N-((3-((5-甲基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145289)
Figure PCTCN2015094954-appb-000040
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.79(s,1H),8.44(s,1H),8.31(s,1H),8.20(s,1H),7.93(s,1H),7.86(d,J=8.8Hz,1H),7.79-7.76(m,2H),7.70(d,J=8.0Hz,1H),7.60(s,1H),7.47(d,J=8.8Hz,1H),7.40(t,J=7.2Hz,1H),7.36(t,J=7.2Hz,1H),6.96(d,J=8.8Hz,1H),6.67-6.61(m,1H),6.16(d,J=15.2Hz,1H),3.80(s,3H),3.00(d,J=5.6Hz,2H),2.17(s,3H),2.15(s,6H).
MS(ESI):m/z 483[M+H]+.
实施例12
(E)-N-((3-((5-甲氧基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145268)
Figure PCTCN2015094954-appb-000041
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.84(s,1H),8.92(s,1H),8.49(s,1H),8.27(s,1H),7.90-7.87(m,2H),7.79-7.76(m,2H),7.70(d,J=7.6Hz,1H),7.58(s,1H),7.45-7.32(m,3H),6.97(d,J=8.8Hz,1H),6.67-6.61(m,1H),6.17(d,J=15.6Hz,1H),3.90(s,3H),3.82(s,3H),3.03(d,J=5.2Hz,2H),2.16(s,6H).
MS(ESI):m/z 499[M+H]+.
实施例13
(E)-N-((3-((5-氰基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145293)
Figure PCTCN2015094954-appb-000042
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.90(s,1H),9.46(br,1H),9.02(br,1H),8.49(s,1H),8.17(br,1H),7.79(s,1H),7.73(s,3H),7.60-7.58(m,2H),7.40-7.35(m,2H),7.04(d,J=9.2Hz,1H),6.70-6.63(m,1H),6.17(d,J=15.6Hz,1H),3.73(s,3H),3.01(d,J=5.2Hz,2H),2.15(s,6H).
MS(ESI):m/z 494[M+H]+.
实施例14
(E)-N-((3-((5-三氟甲基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145274)
Figure PCTCN2015094954-appb-000043
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.83(s,1H),8.63(br,1H),8.56(s,1H),8.36(s,1H),8.07(s,1H),7.83(d,J=8.0Hz,1H),7.78-7.67(m,4H),7.51(d,J=8.8Hz,1H),7.42-7.39(m,2H),6.99(d,J=8.8Hz,1H),6.69-6.63(m,1H),6.17(d,J=15.2Hz,1H),3.72(s,3H),3.03(d,J=5.6Hz,2H),2.16(s,6H).
MS(ESI):m/z 537[M+H]+.
实施例15
(E)-N-((3-((5-异丙基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145283)
Figure PCTCN2015094954-appb-000044
起始原料8合成中间体9的合成方法如实施例1中步骤1-2。
中间体11的合成步骤如下:
步骤1.N2-(2-甲氧基-5-硝基苯基)-N4-(萘-2-基)-5-异丙烯基嘧啶-2,4-二胺(中间体10)
于封管中加入中间体9(771mg,1.66mmol),1,1'-双(二苯基磷)二茂铁二氯化钯(61mg,0.08mmol),碳酸钾(686mg,4.97mmol),加入二氧六环/水(20mL,v/v=3/1),氩气置换3次,最后加入异丙烯基硼酸频哪醇酯(0.62mL,3.32mmol),密封,80℃搅拌过夜。冷却至室温,加入10%NaHCO3水溶液,二氯甲烷萃取,无水Na2SO4干燥,旋干,柱层析分离得固体(507mg,收率:71%)。
步骤2.5-异丙基-N2-(2-甲氧基-5-氨基苯基)-N4-(萘-2-基)嘧啶-2,4-二胺(中间体11)
于中间体10(500mg,1.17mmol),10%Pd/C(50mg)的反应瓶中加入的乙醇(10mL)溶液,氢气置换三次,氢气球下室温反应3h。硅藻土抽滤,旋干,柱层析分离得固体(303mg,收率:65%)。
中间体11合成终产物CCB145283的合成方法如实施例1步骤4。
1H NMR(400MHz,DMSO-d6):δ9.76(s,1H),8.51(s,1H),8.24(s,1H),8.23(s,1H),8.02(s,1H),7.85(d,J=8.8Hz,1H),7.77(d,J=8.8Hz,2H),7.71(d,J=8.0Hz,1H),7.56(s,1H),7.44-7.33(m,3H),6.95(d,J=8.8Hz,1H),6.67-6.60(m,1H),6.15(d,J=15.2Hz,1H),3.79(s,3H),3.27(m,J=6.8Hz,1H),3.01(d,J=5.6Hz,2H),2.15(s,6H),1.26(s,J=6.8Hz,6H).
MS(ESI):m/z 511[M+H]+.
实施例16
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145333)
Figure PCTCN2015094954-appb-000045
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.92(s,1H),9.38(s,1H),8.93(br,1H),8.37(s,1H),8.18(s,1H),7.84(s,3H),7.79(s,2H),7.48-7.39(m,3H),7.28(d,J=8.0Hz,1H),7.04(t,J=8.0Hz,1H),6.72-6.65(m,1H),6.23(d,J=15.6Hz,1H),3.02(d,J=5.2Hz,2H),2.16(s,6H).
MS(ESI):m/z 473[M+H]+.
实施例17
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145340)
Figure PCTCN2015094954-appb-000046
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.98(s,1H),8.75(s,1H),8.73(s,1H),8.29(s,1H),8.11(s,1H),7.78-7.68(m,4H),7.54-7.51(m,2H),7.38-7.31(m,2H),7.17(d,J=8.4Hz,1H),6.71-6.64(m,1H),6.20(d,J=15.6Hz,1H),3.01(d,J=5.6Hz,2H),2.14(s,3H),2.13(s,6H).
MS(ESI):m/z 487[M+H]+.
实施例18
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-氟苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145329)
Figure PCTCN2015094954-appb-000047
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ10.21(s,1H),9.03(s,1H),8.87(s,1H),8.30(s,1H),8.15(s,1H),7.86(d,J=6.8Hz,1H),7.81-7.73(m,3H),7.62(d,J=8.0Hz,1H),7.53(s,1H),7.42-7.34(m,2H),7.20(t,J=9.6Hz,1H),6.72-6.65(m,1H),6.23(d,J=15.6Hz,1H),3.02(d,J=5.2Hz,2H),2.15(s,6H).
MS(ESI):m/z 491[M+H]+.
实施例19
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-乙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145373)
Figure PCTCN2015094954-appb-000048
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6)δ9.81(s,1H),8.90(s,1H),8.31(s,1H),8.17(s,1H),8.08(s,1H),8.00(s,1H),7.82(d,J=8.8Hz,1H),7.77(m,2H),7.64(d,J=8.0Hz,1H),7.49(dd,J=8.8Hz,1.6Hz,1H),7.41(d,J=6.8Hz,1H),7.38-7.34(m,1H),6.98(d,J=8.8Hz,1H),6.72-6.60(m,1H),6.14(d,J=15.2Hz,1H),4.01(q,2H),3.01(d,J=6.0Hz,2H),2.15(s,6H),1.27(t,3H).
MS(ESI):m/z 517[M+H]+.
实施例20
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-异丙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145384)
Figure PCTCN2015094954-appb-000049
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ9.80(s,1H),8.93(s,1H),8.32(s,1H),8.17(s,1H),8.05(s,1H),7.98(s,1H),7.83(d,J=8.8Hz,1H),7.77(d,J=8.8Hz,2H),7.65(d,J=8.0Hz,1H),7.47(d,J=8.8Hz,1H),7.43-7.35(m,2H),7.00(d,J=8.8Hz,1H),6.67-6.60(m,1H),6.14(d,J=15.2Hz,1H),4.51(m,1H),3.01(d,J=6.0Hz,2H),2.15(s,6H),1.24(s,3H),1.22(s,3H).
MS(ESI):m/z 531[M+H]+.
实施例21
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145344)
Figure PCTCN2015094954-appb-000050
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ9.85(s,1H),9.34(s,1H),8.97(s,1H),8.33(s,1H),8.18(s, 1H),7.86-7.81(m,3H),7.76(d,J=8.0Hz,1H),7.55(s,1H),7.47-7.39(m,2H),7.23(s,1H),7.11(s,1H),6.70-6.63(m,1H),6.22(d,J=15.2Hz,1H),3.02(d,J=5.6Hz,2H),2.15(s,6H),1.95(s,3H).
MS(ESI):m/z 487[M+H]+.
实施例22
(E)-N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145346)
Figure PCTCN2015094954-appb-000051
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ9.36(s,1H),9.33(s,1H),8.97(s,1H),8.32(s,1H),8.18(s,1H),7.87-7.79(m,4H),7.64(s,1H),7.50-7.49(m,2H),7.43(t,J=7.2Hz,1H),6.93(d,J=8.0Hz,1H),6.71-6.64(m,1H),6.33(d,J=15.2Hz,1H),3.14(d,J=4.0Hz,2H),2.24(s,6H),2.10(s,3H).
MS(ESI):m/z 487[M+H]+.
实施例23
(E)-N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-氟-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145380)
Figure PCTCN2015094954-appb-000052
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ9.63(s,1H),8.88(s,1H),8.20(s,2H),8.12(s,1H),8.07(d,J=8.0Hz,1H),7.83-7.75(m,3H),7.69(d,J=8.0Hz,1H),7.46-7.37(m,2H),7.05(d,J=12.4Hz,1H),6.68-6.61(m,1H),6.31(d,J=15.2Hz,1H),3.76(s,3H),3.02(d,J=5.6Hz,2H),2.16(s,6H).
MS(ESI):m/z 521[M+H]+.
实施例24
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145335)
Figure PCTCN2015094954-appb-000053
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ9.98(s,1H),9.41(s,1H),8.71(s,1H),8.32(s,1H),8.26(s,1H),7.85(d,J=7.6Hz,1H),7.79(d,J=8.4Hz,1H),7.48-7.42(m,3H),7.27(d,J=7.6Hz,1H),6.99(t,J=7.6Hz,1H),6.70-6.66(m,1H),6.24(d,J=15.6Hz,1H),3.03(d,J=4.8Hz,2H),2.16(s,6H).
MS(ESI):m/z 517[M+H]+.
实施例25
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145339)
Figure PCTCN2015094954-appb-000054
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ10.02(s,1H),8.79(s,1H),8.47(s,1H),8.25(s,1H),8.20(s,1H),7.73-7.68(m,4H),7.53(d,J=8.0Hz,2H),7.37-7.34(m,2H),7.17(d,J=8.0Hz,1H),6.70-6.66(m,1H),6.22(d,J=15.2Hz,1H),3.07(d,J=4.0Hz,2H),2.18(s,6H),2.14(s,3H).
MS(ESI):m/z 531[M+H]+.
实施例26
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-氟苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145330)
Figure PCTCN2015094954-appb-000055
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ10.14(s,1H),9.03(s,1H),8.59(s,1H),8.24(s,1H),8.22(s,1H),7.84(d,J=7.2Hz,1H),7.78-7.73(m,3H),7.63(d,J=8.0Hz,1H),7.52(d,J=8.0Hz,1H),7.43-7.34(m,2H),7.19(t,J=9.6Hz,1H),6.72-6.65(m,1H),6.21(d,J=15.6Hz,1H),3.03 (d,J=5.6Hz,2H),2.15(s,6H).
MS(ESI):m/z 535[M+H]+.
实施例27
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-乙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145374)
Figure PCTCN2015094954-appb-000056
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ9.80(s,1H),8.63(s,1H),8.26(s,1H),8.24(s,1H),8.09(s,1H),7.98(s,1H),7.81-7.75(m,3H),7.64(d,J=7.6Hz,1H),7.48(d,J=8.8Hz,1H),7.42-7.35(m,2H),6.97(d,J=8.8Hz,1H),6.67-6.61(m,1H),6.14(d,J=15.6Hz,1H),4.02(q,2H),3.02(d,J=6.0Hz,2H),2.15(s,6H),1.27(t,3H).
MS(ESI):m/z 561[M+H]+.
实施例28
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-异丙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145385)
Figure PCTCN2015094954-appb-000057
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ9.79(s,1H),8.66(s,1H),8.27(s,1H),8.25(s,1H),8.03(s,1H),7.98(s,1H),7.81-7.76(m,3H),7.65(d,J=7.6Hz,1H),7.45(d,J=8.8Hz,1H),7.43-7.35(m,2H),7.00(d,J=8.8Hz,1H),6.67-6.60(m,1H),6.14(d,J=15.2Hz,1H),4.51(m,1H),3.01(d,J=5.6Hz,2H),2.15(s,6H),1.23(s,3H),1.22(s,3H).
MS(ESI):m/z 575[M+H]+.
实施例29
(E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145342)
Figure PCTCN2015094954-appb-000058
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ9.84(s,1H),9.34(s,1H),8.72(s,1H),8.26(s,2H),7.85-7.84(m,2H),7.79-7.76(m,2H),7.52(s,1H),7.47-7.39(m,2H),7.21(s,1H),7.09(s,1H),6.70-6.63(m,1H),6.22(d,J=15.2Hz,1H),3.02(d,J=5.6Hz,2H),2.15(s,6H),1.91(s,3H).
MS(ESI):m/z 531[M+H]+.
实施例30
(E)-N-((5-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145348)
Figure PCTCN2015094954-appb-000059
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ9.32(s,2H),8.71(s,1H),8.25(s,2H),7.88(d,J=8.4Hz,2H),7.80(d,J=8.4Hz,2H),7.61(s,1H),7.51-7.42(m,3H),6.88(d,J=8.8Hz,1H),6.70-6.63(m,1H),6.31(d,J=14.4Hz,1H),3.03(d,J=4.8Hz,2H),2.17(s,6H),2.09(s,3H).
MS(ESI):m/z 531[M+H]+.
实施例31
(E)-N-((5-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-氟-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺(CCB145381)
Figure PCTCN2015094954-appb-000060
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ9.63(s,1H),8.61(s,1H),8.20(s,2H),8.16(s,1H),8.06(d,J=8.0Hz,1H),7.80-7.75(m,3H),7.69(d,J=8.0Hz,1H),7.46-7.37(m,2H),7.04(d,J=12.4Hz,1H),6.68-6.61(m,1H),6.30(d,J=15.6Hz,1H),3.75(s,3H),3.02(d,J=5.6Hz,2H),2.16(s,6H).
MS(ESI):m/z 565[M+H]+.
实施例32
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺(CCB120027)
Figure PCTCN2015094954-appb-000061
中间体4的合成方法如实施例1。终产物CCB120027的合成步骤如下:
中间体4(780mg,2mmol)溶于的二氯甲烷(10mL)中,加入二异丙基乙胺(0.41mL,2.4mmol),0℃下搅拌5分钟,滴加丙烯酰氯(0.16mL,2mmol),继续搅拌2h。旋干,加入水(10mL),二氯甲烷萃取三次。无水Na2SO4干燥,旋干,柱层析分离得固体(405mg,收率:45%)。
1H NMR(400MHz,DMSO-d6)δ9.94(s,1H),8.89(s,1H),8.28(s,1H),8.15(s,2H),7.99(s,1H),7.83-7.78(m,3H),7.68(d,J=8.4Hz,1H),7.54(d,J=8.4Hz,1H),7.41-7.37(m,2H),7.01(d,J=8.8Hz,1H),6.34(dd,J=16.8Hz,9.6Hz,1H),6.18(d,J=16.8Hz,1H),5.67(d,J=9.6Hz,1H),3.77(s,3H).MS(ESI):m/z 446[M+H]+.
实施例33
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(哌啶-1-基)-2-丁烯酰胺(CCB120024)
Figure PCTCN2015094954-appb-000062
中间体14的合成步骤如下:
步骤1.(E)-4-(哌啶-1-基)丁-2-烯酸乙酯(中间体13)
原料12(965mg,4mmol)溶于的四氢呋喃(20mL)中,0℃下搅拌5分钟,滴加哌啶(1mL,10mmol),室温继续搅拌2h。旋干,二氯甲烷溶解,饱和NaCl溶液洗涤2次。无水Na2SO4干燥,旋干,柱层析分离得液体(673mg,收率:85%)。
步骤2.(E)-4-(哌啶-1-基)丁-2-烯酸(中间体14)
10%的NaOH溶液(400mg/4mL)滴入中间体13(650mg,3.3mmol)的甲醇(10mL)溶液中,45℃中搅拌1h。用6N HCl调pH至1-2,旋干,加入乙醇,抽滤,滤液旋干,加入异丙醇,抽滤,得固体直接用于下一步。
中间体4合成终产物CCB120024的合成方法如实施例1中步骤4。
1H NMR(400MHz,DMSO-d6)δ9.82(s,1H),8.88(s,1H),8.29(s,1H),8.15(s,1H),8.14(s,1H),7.97(d,J=2.4Hz,1H),7.82(dd,J=8.8Hz,1.6Hz,1H),7.78-7.76(m,2H),7.66(d,J=8.0Hz,1H),7.52(dd,J=8.8Hz,2.4Hz,1H),7.42-7.34(m,2H),7.00(d,J=8.8Hz,1H),6.68-6.61(m,1H),6.14(d,J=15.2Hz,1H),3.76(s,3H),3.03(d,J=5.6Hz,2H),2.31(br,4H),1.51-1.49(br,4H),1.38(br,2H).
MS(ESI):m/z 543[M+H]+.
实施例34
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(吗啉-1-基)-2-丁烯酰胺(CCB120025)
Figure PCTCN2015094954-appb-000063
合成方法如实施例33。
1H NMR(400MHz,DMSO-d6)δ9.84(s,1H),8.88(s,1H),8.30(s,1H),8.15(s,2H),7.98(d,J=2.4Hz,1H),7.82(dd,J=9.2Hz,2.0Hz,1H),7.78-7.76(m,2H),7.66(d,J=7.2Hz,1H),7.53(dd,J=9.2Hz,2.0Hz,1H),7.42-7.35(m,2H),7.01(d,J=8.8Hz,1H),6.66-6.59(m,1H),6.16(d,J=15.6Hz,1H),3.76(s,3H),3.58(m,4H),3.07(d,J=5.6Hz,2H),2.35(br,4H).
MS(ESI):m/z 545[M+H]+.
实施例35
(E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(4-乙酰基哌嗪-1-基)-2-丁烯酰胺(CCB120029)
Figure PCTCN2015094954-appb-000064
合成方法如实施例33。
1H NMR(400MHz,DMSO-d6)δ9.85(s,1H),8.88(s,1H),8.29(s,1H),8.15(s,1H),8.14(s,1H),7.98(d,J=2.0Hz,1H),7.82(dd,J=8.8Hz,2.0Hz,1H),7.78-7.76(m,2H),7.66(d,J=7.6Hz,1H),7.53(dd,J=8.8Hz,2.0Hz,1H),7.42-7.35(m,2H),7.00(d,J=8.8Hz,1H),6.68-6.61(m,1H),6.16(d,J=15.2Hz,1H),3.76(s,3H),3.42(m,4H),3.11(d,J=5.6Hz,2H),2.37(m,2H),2.30(m,2H),1.98(s,3H).
MS(ESI):m/z 586[M+H]+.
实施例36
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-吗啉甲基苯基)丙烯酰胺(CCB120106)
Figure PCTCN2015094954-appb-000065
中间体17的合成步骤如下:
步骤1.(3,5-二硝基苯基)(吗啉)甲酮(中间体16)
原料15(424mg,2mmol),***啉(0.174mL),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(912mg,2.4mmol)溶于乙腈(10mL)中,加入二异丙基乙胺(1.04mL),室温搅拌过夜。旋干,加入的10%NaHCO3溶液(10mL),二氯甲烷萃取三次,合并有机相,再用饱和食盐水洗涤一遍,干燥,旋干,柱层析分离得固体(355mg,收率:63%)。
步骤2.3-(吗啉甲基)-5-硝基苯胺(中间体17)
中间体16(350mg,1.26mmol)溶于无水THF(5mL),0℃下缓慢滴加2M的BH3-Me2S溶液(3.79mL,7.58mmol),室温搅拌3h,回流过夜。自然冷却到室温,加入6NHCl溶液室温下搅拌1h,回流2h。加入8NNaOH水溶液调至中性,加水稀释,乙酸乙酯萃取三次,饱和NaCl溶液洗涤,干燥,旋干,柱层析分离得固体(150mg,收率:50%)。
中间体2合成中间体18的合成方法如实施例1中步骤2-3。
中间体18合成终产物CCB120106的合成方法如实施例32。
1H NMR(400MHz,DMSO-d6):δ9.99(s,1H),9.38(s,1H),8.98(s,1H),8.31(s,1H),8.18(s,1H),7.86-7.76(m,5H),7.48-7.40(m,2H),7.32(s,1H),7.24(s,1H),6.40(dd,J=16.8Hz,10.0Hz,1H),6.21(dd,J=16.8Hz,2.0Hz,1H),5.70(dd,J=10.0Hz,2.0Hz,1H),3.47(m,4H),3.05(s,2H),2.14(s,4H).
MS(ESI):m/z 515[M+H]+.
实施例37
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-(二甲氨基甲基)苯基)丙烯酰胺(CCB120081)
Figure PCTCN2015094954-appb-000066
合成方法如实施例36。
1H NMR(400MHz,DMSO-d6)δ9.98(s,1H),9.38(s,1H),8.97(s,1H),8.32(s,1H),8.19(s,1H),7.86-7.78(m,4H),7.74(s,1H),7.48-7.40(m,2H),7.31(s,1H),7.25(s,1H),6.40(dd,J=16.8Hz,10.0Hz,1H),6.20(dd,J=16.8Hz,2.0Hz,1H),5.69(dd,J=10.0Hz,2.0Hz,1H),2.99(s,2H),1.98(s,6H).
MS(ESI):m/z 473[M+H]+.
实施例38
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-((N-甲基-N-二甲氨基乙基)甲基胺)苯基)丙烯酰胺(CCB120069)
Figure PCTCN2015094954-appb-000067
合成方法如实施例36。
1H NMR(400MHz,DMSO-d6)δ9.99(s,1H),9.38(s,1H),8.96(s,1H),8.33(s,1H),8.18(s,1H),7.85-7.80(m,3H),7.78-7.77(m,2H),7.47-7.39(m,2H),7.32(s,1H),7.23(s,1H),6.41(dd,J=16.8Hz,10.0Hz,1H),6.21(dd,J=16.8Hz,1.6Hz,1H),5.69(dd,J=10.0Hz,1.6Hz,1H),3.07(s,2H),2.23(s,4H),2.06(s,6H),1.96(s,3H).
MS(ESI):m/z 530[M+H]+.
实施例39
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-((4-甲基哌嗪-1-基)甲基)苯基)丙烯酰胺(CCB120103)
Figure PCTCN2015094954-appb-000068
合成方法如实施例36。
1H NMR(400MHz,DMSO-d6)δ9.98(s,1H),9.38(s,1H),8.97(s,1H),8.32(s,1H),8.18(s,1H),7.85-7.83(m,3H),7.79-7.77(m,2H),7.47-7.40(m,2H),7.29(s,1H),7.22(s,1H),6.40(dd,J=16.8Hz,10.0Hz,1H),6.20(dd,J=16.8Hz,2.0Hz,1H),5.70(dd,J=10.0Hz,2.0Hz,1H),3.06(s,2H),2.19(br,8H),2.11(s,3H).
MS(ESI):m/z 528[M+H]+.
实施例40
N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-((4-甲基高哌嗪-1-基)甲基)苯基)丙烯酰胺(CCB120105)
Figure PCTCN2015094954-appb-000069
合成方法如实施例36。
1H NMR(400MHz,Acetone-d6)δ9.20(br,1H),8.64(br,1H),8.41(s,1H),8.33(br,1H),8.14(s,1H),7.93(s,1H),7.87-7.82(m,3H),7.76(d,J=8.0Hz,1H),7.50-7.42(m,4H),6.39(dd,J=16.8Hz,10.0Hz,1H),6.30(dd,J=16.8Hz,2.0Hz,1H),5.65(dd,J=10.0Hz,2.0Hz,1H),3.38(s,2H),2.56-2.51(m,6H),2.47-2.46(m,2H),2.43(s,3H),1.71-1.67(m,2H).
MS(ESI):m/z 542[M+H]+.
实施例41
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-((N-甲基-N-二甲氨基乙基)氨基)-4-甲氧基苯基)丙烯酰胺(CCB120067)
Figure PCTCN2015094954-appb-000070
原料1合成中间体19的合成方法如实施例1中步骤1-2。
中间体20的合成步骤如下:
中间体19(330mg,0.75mmol)溶于MeCN(5mL)中,加入N,N-二异丙基乙胺(0.259mL,1.5mmol)及三甲基乙二胺(0.195mL,1.5mmol),80℃搅拌2h。旋干,加入的10%NaHCO3溶液(10mL),二氯甲烷萃取三次,合并有机相,再用饱和食盐水洗涤一遍,干燥,旋干,柱层析分离得固体(370mg,收率:95%)。
中间体20合成终产物CCB120067的合成方法如实施例32。
1H NMR(500MHz,DMSO-d6)δ10.02(s,1H),8.80(s,1H),8.46(s,1H),8.26(s,1H),8.23(s,1H),8.11(s,1H),7.81(d,J=8.5Hz,1H),7.76(d,J=7.5Hz,1H),7.72(d,J=8.5Hz,1H),7.64(d,J=7.5Hz,1H),7.42-7.35(m,2H),7.00(s,1H),6.34(dd,J=17.0Hz,10.0Hz,1H),6.14(d,J=17.0Hz,1H),5.68(d,J=10.0Hz,1H),3.75(s,3H),2.88-2.86(m,2H),2.72(s,3H),2.31-2.29(m,2H),2.19(s,6H).
MS(ESI):m/z 546[M+H]+.
实施例42
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-乙酰基哌嗪)-4-甲氧基苯基)丙烯酰胺(CCB120111)
Figure PCTCN2015094954-appb-000071
合成方法如实施例41。
1H NMR(400MHz,DMSO-d6)δ9.03(s,1H),8.81(s,1H),8.25(s,1H),8.23(s,1H),8.22(s,1H),8.11(s,1H),7.81-7.74(m,3H),7.62(d,J=7.6Hz,1H),7.43-7.36(m,2H),6.89(s,1H),6.62(dd,J=16.8Hz,10.4Hz,1H),6.15(d,J=16.8Hz,1H),5.68(d,J=10.4Hz,1H),3.75(s,3H),3.66(br,4H),2.85-2.80(m,4H),2.06(s,3H).
MS(ESI):m/z 572[M+H]+.
实施例43
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-甲基哌嗪)-4-甲氧基苯基)丙烯酰胺(CCB120115)
Figure PCTCN2015094954-appb-000072
合成方法如实施例41。
1H NMR(400MHz,DMSO-d6)δ8.89(s,1H),8.79(s,1H),8.26(s,1H),8.23(s,1H),8.11(s,1H),8.10(s,1H),7.79-7.74(m,3H),7.61(d,J=7.2Hz,1H),7.43-7.36(m,2H),6.86(s,1H),6.54(dd,J=16.0Hz,10.0Hz,1H),6.12(d,J=16.0Hz,1H),5.67(d,J=10.0Hz,1H),3.75(s,3H),2.87(s,4H),2.55(s,4H),2.26(s,3H).
MS(ESI):m/z 544[M+H]+.
实施例44
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-吗啉基-4-甲氧基苯基)丙烯酰胺(CCB120117)
Figure PCTCN2015094954-appb-000073
合成方法如实施例41。
1H NMR(400MHz,DMSO-d6)δ9.00(s,1H),8.81(s,1H),8.26(s,1H),8.23(s,1H),8.17(s,1H),8.11(s,1H),7.82-7.74(m,3H),7.63(d,J=7.2Hz,1H),7.43-7.36(m,2H),6.88(s,1H),6.59(dd,J=16.8Hz,10.4Hz,1H),6.14(d,J=16.8Hz,1H),5.67(d,J=10.4 Hz,1H),3.81(br,4H),3.76(s,3H),2.87(br,4H).
MS(ESI):m/z 531[M+H]+.
实施例45
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-甲基高哌嗪基)-4-甲氧基苯基)丙烯酰胺(CCB120120)
Figure PCTCN2015094954-appb-000074
合成方法如实施例41。
1H NMR(400MHz,DMSO-d6)δ9.06(s,1H),8.78(s,1H),8.27(s,1H),8.20(s,1H),8.10(s,1H),7.98(s,1H),7.81-7.74(m,3H),7.63(d,J=7.2Hz,1H),7.43-7.36(m,2H),6.85(s,1H),6.51(dd,J=16.8Hz,10.0Hz,1H),6.13(d,J=16.8Hz,1H),5.67(d,J=10.0Hz,1H),3.74(s,3H),3.16-3.14(m,4H),2.69-2.67(m,4H),2.33(s,3H),1.89-1.86(m,2H).
MS(ESI):m/z 558[M+H]+.
实施例46
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-二甲氨基哌啶)-4-甲氧基苯基)丙烯酰胺(CCB120121)
Figure PCTCN2015094954-appb-000075
合成方法如实施例41。
1H NMR(400MHz,DMSO-d6)δ8.91(s,1H),8.78(s,1H),8.27(s,1H),8.23(s,1H),8.14(s,1H),8.10(s,1H),7.81-7.73(m,3H),7.61(d,J=7.2Hz,1H),7.43-7.36(m,2H),6.84(s,1H),6.61(dd,J=16.8Hz,10.4Hz,1H),6.14(d,J=16.8Hz,1H),5.67(d,J=10.4Hz,1H),3.74(s,3H),3.06-3.04(m,2H),2.71-2.65(m,2H),2.32(s,6H),2.23-2.17(m,1H),1.87-1.84(m,2H),1.74-1.66(m,2H).
MS(ESI):m/z 572[M+H]+.
实施例47
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(3-二甲氨基吡咯烷-1-基)-4-甲氧基苯基)丙烯酰胺(CCB120137)
Figure PCTCN2015094954-appb-000076
合成方法如实施例41。
1H NMR(400MHz,DMSO-d6):δ9.26(s,1H),8.73(s,1H),8.29(s,1H),8.14(s,1H),8.07(s,1H),7.82-7.76(m,3H),7.65(d,J=6.8Hz,1H),7.43-7.36(m,3H),6.51(s,1H),6.45(dd,J=16.8Hz,10.0Hz,1H),6.13(d,J=16.8Hz,1H),5.64(d,J=10.0Hz,1H),3.73(s,3H),3.42-3.35(m,1H),3.24-3.16(m,3H),2.72-2.68(m,1H),2.17(s,6H),2.13-2.09(m,1H),1.77-1.72(m,1H).
MS(ESI):m/z 558[M+H]+.
实施例48
N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(3-二甲氨基氮杂环丁烷-1-基)-4-甲氧基苯基)丙烯酰胺(CCB120138)
Figure PCTCN2015094954-appb-000077
合成方法如实施例41。
1H NMR(400MHz,DMSO-d6)δ9.20(s,1H),8.73(s,1H),8.28(s,1H),8.12(s,1H),8.06(s,1H),7.81-7.75(m,3H),7.65(d,J=7.6Hz,1H),7.51(t,J=7.6Hz,1H),7.38(t,J=7.6Hz,1H),7.33(s,1H),6.43(dd,J=16.8Hz,10.4Hz,1H),6.23(s,1H),6.14(d,J=16.8Hz,1H),5.64(d,J=10.4Hz,1H),3.98(t,J=7.2Hz,2H),3.72(s,3H),3.59(t,J=7.2Hz,2H),3.07(m,1H),2.10(s,6H).
MS(ESI):m/z 544[M+H]+.
实施例49
N-((5-((5-氯-4-(萘-2-基氨基)嘧啶-2-基)氨基)-4-甲氧基-2-(甲基(2-甲基氨基)乙基)氨基)苯基)丙烯酰胺(CCB4580030)
Figure PCTCN2015094954-appb-000078
原料1合成中间体21的合成方法如实施例41。
终产物CCB4580030的合成步骤如下:
中间体21(378mg,0.60mmol)溶于DCM(5mL)中,冷却至0℃下滴加三氟醋酸(446μL,6.0mmol),恢复室温继续搅拌2h。旋干,加入的10%NaHCO3溶液(10mL),二氯甲烷萃取三次,合并有机相,再用饱和食盐水洗涤一遍,干燥,旋干,柱层析分离得固体(293mg,收率:92%)。
1H NMR(400MHz,DMSO-d6)δ9.39(s,1H),8.88(s,1H),8.24-8.20(m,3H),8.16(s,1H),8.12(s,1H),7.85-7.80(m,3H),7.78-7.70(m,1H),7.46-7.36(m,2H),6.94(s,1H),6.56(dd,J=16.8Hz,10.4Hz,1H),6.21(d,J=16.8Hz,1H),5.74(d,J=10.4Hz,1H),3.80(s,3H),3.24-3.18(m,2H),3.10-3.04(m,2H),2.59(s,3H),2.57(s,3H).
MS(ESI):m/z 532[M+H]+.
实施例50
2-氨基嘧啶类化合物激酶抑制活性测试
将酶反应底物聚(谷氨酸,酪氨酸)4:1(Sigma公司)用无钾离子的PBS(10mM磷酸钠缓冲液,150mM NaCl,pH 7.2-7.4)稀释成20μg/mL,125μL/孔包被酶标板,置37℃反应12-16小时。弃去孔中液体后,用200μL/孔的T-PBS(含0.1%Tween-20的PBS)洗板三次,每次5分钟,然后于37℃烘箱中干燥酶标板1-2小时。每孔加入用反应缓冲液(50mM HEPES pH 7.4,50mM MgCl2,5mM MnCl2,0.2mM Na3VO4,1mM DTT)稀释的ATP溶液50μL,终浓度5μM。化合物分别用1%DMSO稀释成合适的浓度,每孔加入10μL,再分别加入用40μL反应缓冲液稀释的不同种类的酪氨酸激酶蛋白(包括EGFRwt,EGFRT790M/L858R和IGF-1R,购自Millipore公司)。将上述反应体系置37℃摇床(100rpm)反应1小时。每次实验需设无酶对照孔及DMSO溶剂对照孔。反应结束后,用T-PBS洗板三次。加入抗磷酸化酪氨酸的一抗PY99(Santa Cruz公司)100μL/孔,抗体用含BSA 5mg/mL的T-PBS 1:1000稀释),37℃摇床反应0.5小时,然后T-PBS洗板三次。加入二抗辣根过氧化物酶标记羊抗鼠的IgG 100μL/孔(抗体用含BSA 5mg/mL的T-PBS 1:2000稀释),37℃摇床反应0.5小时,T-PBS洗板三次。加入2mg/mL的OPD显色液100μL/孔(用含有0.03%H2O2的0.1M柠檬酸-柠檬酸钠缓冲液(pH=5.4)稀释),25℃避光反应1-10分钟。加入2M H2SO450μL/孔中止反应,用可调波长式微孔板酶标仪SPECTRA MAX 190读数,波长为490nm。
Figure PCTCN2015094954-appb-000079
IC50值通过抑制曲线以四参数拟合计算。
表1中所列为化合物编号以及对应激酶活性结果。
表1化合物激酶抑制活性
Figure PCTCN2015094954-appb-000080
Figure PCTCN2015094954-appb-000081
由激酶活性检测结果可知,2-氨基嘧啶类化合物存在与蛋白半胱氨酸位点形成不可逆迈克尔加成反应,本发明化合物对EGFR两种亚型激酶都表现出较高的抑制活性。部分化合物(如CCB120027,CCB120024,CCB120067,CCB120115,CCB120120,CCB120069,CCB120137等)显示强效激酶抑制活性,相对于EGFRWT,其对EGFRT790M/L858R抑制活性强20-100倍以上,且对EGFRT790M/L858R的抑制活性强于阳性对照化合物CO1686和AZD-9291(EGFR抑制剂)。
实施例51
化合物对高表达野生型EGFR的人表皮癌A431细胞株及高表达EGFR T790M/L858R突变的人非小细胞肺癌NCI-H1975细胞株的体外增殖抑制作用
细胞株:人表皮癌A431细胞株及人非小细胞肺癌NCI-H1975细胞株购自美国标准生物品收藏中心(ATCC)。
方法:磺酰罗单明B(sulforhodamine B,SRB)法,具体如下:将一定数量处于对数生长期的不同肿瘤细胞接种于96孔培养板,培养24h细胞贴壁后,加入不同浓度的本发明的受试化合物,每个浓度设三复孔,并设定相应浓度的DMSO溶剂对照及无细胞调零孔。用药物处理细胞72h后,倾去培养液,加入100μL冰预冷的10%的三氯乙酸溶液固定细胞,4℃放置1h后用蒸馏水洗涤5次,空气中自然干燥。然后加入100μL SRB(4mg/mL)(Sigma公司)溶液,室温中染色15min,去染色液,用1%冰醋酸洗涤5次,空气干燥。最后加入150μL 10mM的Tris溶液(pH 10.5),可调波长式微孔板酶标仪(VERSAmaxTM,Molecular Device Corporation,Sunnyvale,CA,USA)在515nm波长下测定OD值。以下列公式计算药物对细胞生长的抑制率:
抑制率(%)=(OD对照-OD加药)/OD对照×100%。
根据2-氨基嘧啶类化合物对这些细胞的生长抑制作用,计算出其半数抑制浓度(IC50)值,如表2所示。
表2化合物细胞活性
Figure PCTCN2015094954-appb-000082
Figure PCTCN2015094954-appb-000083
结果发现(见表2),2-氨基嘧啶类化合物可显著抑制NCI-H1975癌细胞的增殖,而且化合物对A431细胞的抑制活性远低于其对NCI-H1975细胞的活性,表现出化合物的选择性。部分化合物对NCI-H1975细胞的抑制活性,优于阳性对照化合物CO1686。
实施例52
2-氨基嘧啶类化合物对NCI-H1975细胞及A431细胞中EGFR激酶磷酸化及下游信号通路活化的影响
使用常规Western Blot(免疫印迹法)进行检测,具体如下。分别将处于对数生长期的A431细胞和NCI-H1975细胞按一定数量种于6孔板,培养箱内贴壁培养过夜后,换无血清培养液饥饿24h,加入一定浓度的化合物作用2h,加入EGF刺激因子,50ng/mL作用10min,用裂解液裂解细胞收样。然后取适量样品进行SDS-PAGE电泳,电泳结束后用半干电转移***将蛋白转移至硝酸纤维素膜,将硝酸纤维素膜置于封闭液(5%脱脂奶粉稀释于含0.1%Tween 20的TBS)中室温封闭2h,然后将膜分别置于一抗溶液(1:500稀释于含0.1%Tween 20的TBS)中4℃孵育过夜。用含0.1%Tween 20的TBS洗涤三次,每次15min。将膜置 于二抗溶液(辣根过氧化物酶标记羊抗兔的IgG,1:2000稀释于含0.1%Tween 20的TBS)中室温反应1h。同上洗膜三次后,用ECL plus试剂发色,Image Quant LAS 4000拍照。
通过图1可以发现,2-氨基嘧啶类化合物中,CCB120067能显著抑制NCI-H1975细胞中EGFRT790M/L858R的磷酸化及其下游信号通路蛋白AKT及ERK的活化,且抑制活性优于阳性对照化合物CO1686。如图所示,CCB120067在1nM浓度下对磷酸化EGFRT790M/L858R及其下游磷酸化Akt和Erk的抑制活性,与CO1686100nM浓度下的抑制活性相当或略优。此外,由图2结果可以发现,化合物CCB120067对A431细胞中EGFRwt的磷酸化抑制活性较弱。
实施例53
2-氨基嘧啶类化合物对人肺癌NCI-H1975裸小鼠皮下移植瘤生长抑制作用
取生长旺盛期的肿瘤组织,剪切成1.5mm3左右的小块,于无菌条件下接种于裸小鼠皮下。待肿瘤生长至100mm3左右将裸小鼠随机分组。受试化合物CCB120067给药剂量为10mg/kg,阳性对照化合物CO1686给药剂量为100mg/kg,均口服灌胃给药,每天一次,连续给药11天,同时设立溶剂对照组(含1%Tween 80的注射用水)。整个实验过程中,每周2次用游标卡尺测量移植瘤直径,称量小鼠体重。肿瘤体积的计算公式为:
TV=1/2×a×b2(其中a、b分别表示长、宽)
结果如图3所示。通过图3可以发现,2-氨基嘧啶类化合物中,CCB12006710mg/kg(灌胃给药)可显著抑制NCI-H1975裸小鼠移植瘤的生长,并在给药7天后肿瘤完全消退。同阳性对照化合物相比,CCB120067在10mg/kg给药剂量下,对肿瘤生长抑制活性强于CO1686在100mg/kg给药剂量的抑瘤效果。
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (11)

  1. 一种式I化合物,其药学上可接受的盐或立体异构体或其前药分子,
    Figure PCTCN2015094954-appb-100001
    其中,R1和R2各自独立地为H、卤素、氰基、取代或未取代的C1-C6烷基、取代或未取代的C3-C6环烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C6环烷氧基;
    R3为H或-(CH2)mNR8R9;R4和R5独立地为H、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、卤素、-(CH2)mNR8R9、-(CH2)mCR6R8R9;其中,各m独立地为0、1、2或3;R6为H或C1-C3烷基;各R8和各R9独立地选自H、取代或未取代的C1-C6烷基,或者R8、R9和相连的N或C一同形成未取代或取代的含有1、2或3个选自O、N、S杂原子的3-8元单环或稠环;
    W为NH、N(C1-C3烷基)、O或S;
    X、Y、Z各自独立地为N或-CR10,其中,R10为氢、卤素、取代或未取代的C1-C3烷基、取代或未取代的C1-C3烷氧基;
    Figure PCTCN2015094954-appb-100002
    为含有0、1、2或3个选自O、N或S的杂原子的取代或未取代的5-7元芳香环;
    其中,各取代独立地指被选自下组的取代基取代:卤素、羟基、氨基、C1-C3烷基、C1-C3烷氧基、-NH(C1-C3烷基)、-N(C1-C3烷基)(C1-C3烷基)、-C(=O)(C1-C3烷基)。
  2. 如权利要求1所述的式I化合物,其特征在于,R1和R2各自独立地为H、卤素、氰基、C1-C6烷基、C1-C6烷氧基、C3-C6环烷基、C3-C6环烷氧基、卤代C1-C6烷基或卤代C1-C6烷氧基。
  3. 如权利要求1所述的式I化合物,其特征在于,
    Figure PCTCN2015094954-appb-100003
    为苯基、呋喃基、噻吩基、吡咯基、吡唑基、噻唑基、异噻唑基、噁唑基、异噁唑基、吡嗪基、哒嗪基、吡啶基、咪唑基或嘧啶基。
  4. 如权利要求1所述的式I化合物,其特征在于,式I化合物具有以下一个或多个特征:
    (1)R3为H、-(CH2)sN(C1-C3烷基)(C1-C3烷基)、或
    Figure PCTCN2015094954-appb-100004
    其中s为1或2或3;
    (2)R4为H、C1-C3烷基、卤代C1-C3烷基、C1-C3烷氧基、卤代C1-C3烷氧基、卤素、-(CH2)mNR8R9、或
    Figure PCTCN2015094954-appb-100005
    (3)R5为H、C1-C3烷基、卤代C1-C3烷基、C1-C3烷氧基、卤代C1-C3烷氧基、卤素、 -(CH2)mNR8R9
    Figure PCTCN2015094954-appb-100006
    其中,各m独立地为0、1、2或3;
    各R8和各R9独立地选自H、取代或未取代的C1-C3烷基,所述取代是指被选自下组的取代基取代:卤素、-NH(C1-C3烷基)、-N(C1-C3烷基)(C1-C3烷基)、-C(=O)(C1-C3烷基);
    各n1、各n2、各n3独立地为0、1、2或3;
    各V独立地为CH、C(C1-C3烷基)或N;
    各U独立地为无、O、S、CR11R12或NR13,其中R11、R12、R13独立地为H、C1-C3烷基、C3-C6环烷基、-NH(C1-C3烷基)、-N(C1-C3烷基)(C1-C3烷基)或-C(=O)(C1-C3烷基)。
  5. 如权利要求4所述的式I化合物,其特征在于,式I化合物具有以下一个或多个特征:
    (1)R3为H、
    Figure PCTCN2015094954-appb-100007
    (2)R4为H、-CH3
    Figure PCTCN2015094954-appb-100008
    (3)R5为H、-CH3、F、
    Figure PCTCN2015094954-appb-100009
    Figure PCTCN2015094954-appb-100010
  6. 如权利要求1所述的式I化合物,其特征在于,所述式I化合物为:
    (E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((喹啉-6-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((喹啉-3-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((吲哚-5-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-(N-甲基-(萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)氧基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)硫基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-(4-((萘-2-基)氨基)嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氟-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-甲基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-甲氧基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氰基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-三氟甲基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-异丙基-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-氟苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-乙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-异丙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-氟-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-氟苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-乙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-异丙氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((3-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((5-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-甲基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    (E)-N-((5-((5-溴-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-氟-4-甲氧基苯基)-4-(二甲氨基)-2-丁烯酰胺;
    N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(哌啶-1-基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(吗啉-1-基)-2-丁烯酰胺;
    (E)-N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-4-甲氧基苯基)-4-(4-乙酰基哌嗪-1-基)-2-丁烯酰胺;
    N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-吗啉甲基苯基)丙烯酰胺;
    N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-(二甲氨基甲基)苯基)丙烯酰胺;
    N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-((N-甲基-N-二甲氨基乙基)甲基胺)苯基)丙烯酰胺;
    N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-((4-甲基哌嗪-1-基)甲基)苯基)丙烯酰胺;
    N-((3-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-5-((4-甲基高哌嗪-1-基)甲基)苯基)丙烯酰胺;
    N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-((N-甲基-N-二甲氨基乙基)氨基)-4-甲氧基苯基)丙烯酰胺;
    N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-乙酰基哌嗪)-4-甲氧基苯基)丙烯酰胺;
    N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-甲基哌嗪)-4-甲氧基苯基)丙烯酰胺;
    N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-吗啉基-4-甲氧基苯基)丙烯酰胺;
    N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-甲基高哌嗪基)-4-甲氧基苯基)丙烯酰胺;
    N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(4-二甲氨基哌啶)-4-甲氧基苯基)丙烯酰胺;
    N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(3-二甲氨基吡咯烷-1-基)-4-甲氧基苯基)丙烯酰胺;
    N-((5-((5-氯-4-((萘-2-基)氨基))嘧啶-2-基)氨基)-2-(3-二甲氨基氮杂环丁烷-1-基)-4-甲氧基苯基)丙烯酰胺;或
    N-((5-((5-氯-4-(萘-2-基氨基)嘧啶-2-基)氨基)-4-甲氧基-2-(甲基(2-甲基氨基)乙基)氨基)苯基)丙烯酰胺。
  7. 如权利要求1-6中任一所述的式I化合物的制备方法,其特征在于,所述制备方法采用式III化合物或其盐为原料,与式A化合物或其盐反应得到式I化合物,或者与式B化合物或其盐反应得到式I化合物,
    Figure PCTCN2015094954-appb-100011
    其中,R1、R2、R3、R4、R5、W、X、Y、Z和
    Figure PCTCN2015094954-appb-100012
    的定义同权利要求1-6;
    R7为卤素、-OCOR14或OR14,R14为C1-C6烷基、卤代C1-C6烷基、C6-C10芳基或C6-C10芳 基C1-C6烷基,优选为C1-C3烷基、卤代C1-C3烷基、苯基或苯基C1-C3烷基。
  8. 一种药物组合物,其特征在于,所述药物组合物包括:
    权利要求1所述的式I化合物,其药学上可接受的盐或立体异构体或其前药分子;以及
    药学上可接受的载体。
  9. 如权利要求1所述的式I化合物,其药学上可接受的盐或立体异构体或其前药分子,或者权利要求8所述的药物组合物的用途,其特征在于,用作:
    (1)制备***的药物;
    (2)制备EGFR蛋白酶抑制剂的药物;
    (3)制备IGF1R蛋白酶抑制剂的药物。
  10. 如权利要求9所述的用途,其特征在于,所述肿瘤选自:非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、***癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌、鼻咽癌、头颈部肿瘤、结肠癌、直肠癌、胶质瘤。
  11. 一种式III化合物或其盐:
    Figure PCTCN2015094954-appb-100013
    其中,R1、R2、R4、R5
    Figure PCTCN2015094954-appb-100014
    W、X、Y和Z的定义同权利要求1-5所述,条件是式III化合物不包括如下结构:
    Figure PCTCN2015094954-appb-100015
PCT/CN2015/094954 2014-11-24 2015-11-18 2-氨基嘧啶类化合物及其药物组合物和应用 WO2016082713A1 (zh)

Priority Applications (11)

Application Number Priority Date Filing Date Title
US15/529,333 US10059694B2 (en) 2014-11-24 2015-11-18 2-aminopyrimidine compound and pharmaceutical composition and use thereof
EP15863532.6A EP3225619B1 (en) 2014-11-24 2015-11-18 2-aminopyrimidine compound and pharmaceutical composition and use thereof
AU2015353210A AU2015353210B2 (en) 2014-11-24 2015-11-18 2-aminopyrimidine compound and pharmaceutical composition and use thereof
CN201580016514.9A CN106458930B (zh) 2014-11-24 2015-11-18 2-氨基嘧啶类化合物及其药物组合物和应用
KR1020177017442A KR101941929B1 (ko) 2014-11-24 2015-11-18 2-아미노피리미딘계 화합물 및 이의 약물 조성물과 응용
CA2968633A CA2968633C (en) 2014-11-24 2015-11-18 2-aminopyrimidine compound and pharmaceutical composition and use thereof
SG11201704244SA SG11201704244SA (en) 2014-11-24 2015-11-18 2-aminopyrimidine compound and pharmaceutical composition and use thereof
NZ733009A NZ733009A (en) 2014-11-24 2015-11-18 2-aminopyrimidine compound and pharmaceutical composition and use thereof
RU2017122062A RU2704129C2 (ru) 2014-11-24 2015-11-18 Соединение 2-аминопиримидина и фармацевтическая композиция и применение данного соединения
JP2017528838A JP6474068B2 (ja) 2014-11-24 2015-11-18 2−アミノピリミジン系化合物およびその薬物組成物と使用
IL252477A IL252477B (en) 2014-11-24 2017-05-24 History of 2-[(ethenyl-carbonyl-amino)anilino]-pyrimidine, process for their preparation and pharmaceutical preparations containing them

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201410682756.9 2014-11-24
CN201410682756.9A CN105601573B (zh) 2014-11-24 2014-11-24 2-氨基嘧啶类化合物及其药物组合物和应用

Publications (1)

Publication Number Publication Date
WO2016082713A1 true WO2016082713A1 (zh) 2016-06-02

Family

ID=55982000

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/094954 WO2016082713A1 (zh) 2014-11-24 2015-11-18 2-氨基嘧啶类化合物及其药物组合物和应用

Country Status (12)

Country Link
US (1) US10059694B2 (zh)
EP (1) EP3225619B1 (zh)
JP (1) JP6474068B2 (zh)
KR (1) KR101941929B1 (zh)
CN (2) CN105601573B (zh)
AU (1) AU2015353210B2 (zh)
CA (1) CA2968633C (zh)
IL (1) IL252477B (zh)
NZ (1) NZ733009A (zh)
RU (1) RU2704129C2 (zh)
SG (1) SG11201704244SA (zh)
WO (1) WO2016082713A1 (zh)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2020527151A (ja) * 2017-07-19 2020-09-03 チア タイ ティエンチン ファーマシューティカル グループ カンパニー リミテッドChia Tai Tianqing Pharmaceutical Group Co., Ltd. Egfrキナーゼ阻害薬としてのアリールホスフィンオキシド
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
CN113304325A (zh) * 2020-02-27 2021-08-27 四川大学 一种集可注射与抗菌于一体的双功能水凝胶及其制备方法和用途
WO2021244502A1 (zh) * 2020-06-03 2021-12-09 江苏先声药业有限公司 多芳基化合物及应用

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105601573B (zh) * 2014-11-24 2021-07-02 中国科学院上海药物研究所 2-氨基嘧啶类化合物及其药物组合物和应用
CN106928150B (zh) * 2015-12-31 2020-07-31 恩瑞生物医药科技(上海)有限公司 丙烯酰胺苯胺衍生物及其药学上的应用
CN107417626B (zh) * 2016-05-23 2020-01-24 江苏奥赛康药业有限公司 一种2-氨基嘧啶类化合物的多晶型形式
CN106083736A (zh) * 2016-06-21 2016-11-09 郑州泰基鸿诺医药股份有限公司 一种嘧啶类化合物、egfr抑制剂及其应用
CN108498477A (zh) * 2017-02-27 2018-09-07 江苏奥赛康药业股份有限公司 一种2-氨基嘧啶类化合物的药用组合物及其制备方法
KR20190108079A (ko) * 2018-03-13 2019-09-23 보로노이바이오 주식회사 2,4,5-치환된 피리미딘 유도체, 이의 제조방법 및 이를 유효성분으로 포함하는 암의 예방 또는 치료용 약학적 조성물
CN110305161A (zh) * 2018-03-20 2019-10-08 暨南大学 2-氨基嘧啶类化合物及其应用
WO2019196838A1 (zh) * 2018-04-13 2019-10-17 江苏奥赛康药业有限公司 一种2-氨基嘧啶类化合物的药学上可接受的盐
CN110856719B (zh) * 2018-08-23 2021-08-10 江苏奥赛康药业有限公司 一种2-氨基嘧啶类化合物的药用组合物
CN113179640A (zh) * 2019-11-26 2021-07-27 上海翰森生物医药科技有限公司 含氮多环类衍生物抑制剂、其制备方法和应用
CN113493420A (zh) * 2020-03-18 2021-10-12 南京药石科技股份有限公司 Egfr酪氨酸激酶抑制剂及其用途
CN115536699B (zh) * 2022-12-01 2023-05-23 北京鑫开元医药科技有限公司 一种新型EGFR-TKIs、制备方法、药物组合物及其应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102083800A (zh) * 2008-06-27 2011-06-01 阿维拉制药公司 杂芳基化合物和其用途
CN102740847A (zh) * 2009-12-29 2012-10-17 阿维拉制药公司 杂芳基化合物和其用途
WO2013169401A1 (en) * 2012-05-05 2013-11-14 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in egfr-driven cancers
CN103717602A (zh) * 2011-05-17 2014-04-09 加利福尼亚大学董事会 激酶抑制剂

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6939874B2 (en) 2001-08-22 2005-09-06 Amgen Inc. Substituted pyrimidinyl derivatives and methods of use
US20090149467A1 (en) * 2005-09-15 2009-06-11 Merck & Co., Inc. Tyrosine Kinase Inhibitors
MX2009011199A (es) * 2007-04-16 2010-03-17 Hutchison Medipharma Entpr Ltd Derivados de pirimidina.
CN105601573B (zh) * 2014-11-24 2021-07-02 中国科学院上海药物研究所 2-氨基嘧啶类化合物及其药物组合物和应用

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102083800A (zh) * 2008-06-27 2011-06-01 阿维拉制药公司 杂芳基化合物和其用途
CN102740847A (zh) * 2009-12-29 2012-10-17 阿维拉制药公司 杂芳基化合物和其用途
CN103717602A (zh) * 2011-05-17 2014-04-09 加利福尼亚大学董事会 激酶抑制剂
WO2013169401A1 (en) * 2012-05-05 2013-11-14 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in egfr-driven cancers

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3225619A4 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2020527151A (ja) * 2017-07-19 2020-09-03 チア タイ ティエンチン ファーマシューティカル グループ カンパニー リミテッドChia Tai Tianqing Pharmaceutical Group Co., Ltd. Egfrキナーゼ阻害薬としてのアリールホスフィンオキシド
US11390625B2 (en) 2017-07-19 2022-07-19 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. Aryl-phosphorus-oxygen compound as EGFR kinase inhibitor
JP7233410B2 (ja) 2017-07-19 2023-03-06 チア タイ ティエンチン ファーマシューティカル グループ カンパニー リミテッド Egfrキナーゼ阻害薬としてのアリールホスフィンオキシド
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
CN113304325A (zh) * 2020-02-27 2021-08-27 四川大学 一种集可注射与抗菌于一体的双功能水凝胶及其制备方法和用途
CN113304325B (zh) * 2020-02-27 2022-03-11 四川大学 一种集可注射与抗菌于一体的双功能水凝胶及其制备方法和用途
WO2021244502A1 (zh) * 2020-06-03 2021-12-09 江苏先声药业有限公司 多芳基化合物及应用

Also Published As

Publication number Publication date
EP3225619A1 (en) 2017-10-04
RU2704129C2 (ru) 2019-10-24
JP6474068B2 (ja) 2019-02-27
IL252477A0 (en) 2017-07-31
US20170283398A1 (en) 2017-10-05
KR20170131341A (ko) 2017-11-29
RU2017122062A (ru) 2018-12-26
CA2968633A1 (en) 2016-06-02
AU2015353210A1 (en) 2017-07-13
JP2017535589A (ja) 2017-11-30
AU2015353210B2 (en) 2019-05-02
EP3225619B1 (en) 2020-10-14
SG11201704244SA (en) 2017-06-29
NZ733009A (en) 2018-10-26
EP3225619A4 (en) 2018-05-23
IL252477B (en) 2021-04-29
CN105601573B (zh) 2021-07-02
CN106458930B (zh) 2019-10-11
US10059694B2 (en) 2018-08-28
CA2968633C (en) 2020-06-30
CN106458930A (zh) 2017-02-22
KR101941929B1 (ko) 2019-01-24
RU2017122062A3 (zh) 2018-12-26
CN105601573A (zh) 2016-05-25

Similar Documents

Publication Publication Date Title
WO2016082713A1 (zh) 2-氨基嘧啶类化合物及其药物组合物和应用
WO2017028797A1 (zh) 取代喹诺酮类衍生物或其药学上可接受的盐或立体异构体及其药用组合物和应用
CN103012399B (zh) 7-氧代吡啶并嘧啶类化合物及其药用组合物和应用
ES2558780T3 (es) Imidazopiridazinas como inhibidores de la cinasa Akt
WO2012000304A1 (zh) 杂环炔苯类化合物及其药用组合物和应用
WO2018133716A1 (zh) 杂环脲类化合物及其药物组合物和应用
WO2012167415A1 (zh) 嘧啶并嘧啶酮类化合物及其药用组合物和应用
ES2911040T3 (es) Nuevos derivados de heteroaril amida como inhibidores selectivos de histona deacetilasa 1 y 2 (HDAC1/2)
ES2660215T3 (es) Potenciador de efecto antitumoral que comprende un compuesto de imidazooxazina
CN107660202B (zh) 水溶性前药
CN113166110B (zh) 2-氨基嘧啶类化合物及其应用
TW200916458A (en) Heterocyclic compounds and methods of use thereof
AU2018338098A1 (en) Cyclic iminopyrimidine derivatives as kinase inhibitors
WO2017202311A1 (zh) 一种2-氨基嘧啶类化合物的多晶型形式
WO2019185064A1 (zh) 喹啉或喹唑啉类化合物及其应用
CN112313213B (zh) 3-氨基吡唑类化合物及其应用
WO2021180008A1 (zh) 含脲结构的三芳环化合物及其应用
CN110386901B (zh) 含磺酰苯胺嘧啶结构的化合物及其作为抗肿瘤药物的应用
US20230099912A1 (en) Benzamide compound and preparation method, use, and pharmaceutical composition thereof
WO2022188755A1 (zh) 吡啶并嘧啶类化合物及其应用

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: 15863532

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2968633

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2017528838

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 15529333

Country of ref document: US

Ref document number: 252477

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 11201704244S

Country of ref document: SG

REEP Request for entry into the european phase

Ref document number: 2015863532

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20177017442

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2017122062

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2015353210

Country of ref document: AU

Date of ref document: 20151118

Kind code of ref document: A