CN113527215B - Quinazoline compound, preparation method and application thereof - Google Patents

Quinazoline compound, preparation method and application thereof Download PDF

Info

Publication number
CN113527215B
CN113527215B CN202110395019.0A CN202110395019A CN113527215B CN 113527215 B CN113527215 B CN 113527215B CN 202110395019 A CN202110395019 A CN 202110395019A CN 113527215 B CN113527215 B CN 113527215B
Authority
CN
China
Prior art keywords
cancer
chloro
methyl
compound
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202110395019.0A
Other languages
Chinese (zh)
Other versions
CN113527215A (en
Inventor
张强
杨磊夫
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Beijing Scitech MQ Pharmaceuticals Ltd
Original Assignee
Beijing Scitech MQ Pharmaceuticals Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beijing Scitech MQ Pharmaceuticals Ltd filed Critical Beijing Scitech MQ Pharmaceuticals Ltd
Priority to TW110113803A priority Critical patent/TWI820414B/en
Priority to PCT/CN2021/087774 priority patent/WO2021209039A1/en
Publication of CN113527215A publication Critical patent/CN113527215A/en
Application granted granted Critical
Publication of CN113527215B publication Critical patent/CN113527215B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/06Anti-spasmodics, e.g. drugs for colics, esophagic dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Dermatology (AREA)
  • Immunology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides a quinazoline compound, a preparation method and application thereof, and in particular relates to a compound shown in a formula (I), an isomer, a hydrate and a solvate thereof, a pharmaceutically acceptable salt and a prodrug thereof, a preparation method and application thereof in preparing medicines serving as tyrosine kinase inhibitors. The compound has good inhibitory activity on EGFR and HER2 kinase, and simultaneously shows excellent blood brain barrier penetrating performance.

Description

Quinazoline compound, preparation method and application thereof
Technical Field
The invention belongs to the technical field of medicines, and relates to a quinazoline compound, a preparation method and application thereof.
Background
Epidermal growth factor receptor (epidermal growth factor receptor, EGFR) is a transmembrane glycoprotein belonging to the family of tyrosine kinase receptors, which is expressed very widely and plays an important role in growth and normal physiological function activities. In addition, EGFR and its mediated signaling pathways play an important role in the development and progression of tumors. However, EGFR expression is very unstable and gene amplification and rearrangement often occurs, altering the antigenic phenotype of the tumor cell surface, most commonly with epidermal growth factor receptor type III mutants (epidermal growthfactor receper variant III, EGFRvIII).
Egfrvlll is a recently discovered class of mutants of epidermal growth factor receptor (epidermal growth factor receptor, EGFR) that are expressed only on the surface of tumor cells, but not normal tissue cells. Aberrant expression of EGFR has been associated with the development of numerous malignant tumors, including glioma, small lung cell carcinoma, breast cancer, bladder cancer, ovarian cancer, and the like.
In contrast to the complete structure of EGFR, exons 2-7 encoding EGFRvIII extracellular ligand binding domain are deleted, resulting in a deletion of 801 base pairs, joining exons 1 and 8 and creating a new glycine at the binding site, resulting in a deletion of amino acids 6-273, thus losing the ability to bind ligand EGF. Egfrvlll, without ligand binding, dimerizes and autophosphorylates tyrosine kinases unregulated structural activation, induces downstream signaling, and stimulates tumor cell proliferation.
Studies have shown that: EGFRvIII can affect tumor development and progression by modulating a variety of signaling pathways, including Ras/Raf/MEK/ERK, PI3/AKT/mTOR, JAK/STAT, and PLC/PKC, among others. EGFRvIII positive tumor cells have obviously increased tumorigenicity, and the tumor cells can generate uncontrollable spontaneous proliferation and metastasis mainly through inhibiting apoptosis, promoting tumor angiogenesis, increasing invasiveness and migration and the like. In addition, egfrvlll plays a role in the escape-like function during radiotherapy and chemotherapy of tumors.
Gliomas are a common, highly aggressive malignancy, with Gliobastoma (GBM) being the most malignant type. The effects of radiotherapy and chemotherapy are not ideal, and recurrence is often caused after operation. Research at home and abroad shows that: 40% -60% of GBM significantly expresses EGFR, and its mutant form is predominantly EGFRvIII. EGFRvIII establishes a signal pathway regulation network through receptor-independent autophosphorylation and tyrosine kinase activity, and plays an important role in regulating the growth, metastasis and angiogenesis of GBM.
Recent studies have found that therapeutic approaches targeting egfrvlll molecules show good anti-tumor effects both in cell culture in vitro and in animal models in vivo. Thus, the development of new therapeutic agents targeted against egfrvlll molecules would provide more effective and economical treatment regimens for oncology patients, particularly glioma patients, and there is a great unmet clinical need.
The drug targeting egfrvlll for treating glioma needs to be capable of not only effectively penetrating the blood brain barrier, but also effectively inhibiting egfrvlll. At present, no report exists on the compound capable of penetrating the blood brain barrier and inhibiting the EGFRvIII, so that the research on the glioma driven by the EGFRvIII has important clinical value. In addition, most of the EGFR and HER2 kinase inhibitors on the market fail to penetrate the blood brain barrier, whereas EGFR-driven lung cancer and HER 2-driven breast cancer patients have a generally worse prognosis and a higher risk of brain metastasis. There is no effective drug approved for brain transfer therapy, and therefore there is a need to develop an EGFR inhibitor and/or HER2 inhibitor with a blood brain barrier that penetrates.
Disclosure of Invention
In one aspect, the invention provides a compound shown in formula (I), isomer, hydrate, solvate, pharmaceutically acceptable salt and prodrug thereof,
in the formula (I), m is 0, 1 or 2;
a is halogen, C 1 -C 3 An alkyl group; z is NH or O;
R 1 is hydrogen, hydroxy, 4-7 membered heteroalicyclic or-NR a R b
R a 、R b Each independently is hydrogen, C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl, C substituted by hydroxy 1 -C 6 Alkyl, quilt C 1- C 3 Alkoxy substituted C 1 -C 6 Alkyl, or C 3- C 6 Cycloalkyl-substituted C 1 -C 6 An alkyl group;
the 4-7 membered heteroalicyclic is a heteroalicyclic containing 1-2 heteroatoms selected from N, O or S, which heteroalicyclic is unsubstituted or C 1 -C 3 Alkyl, C 1 -C 4 Alkanoyl, hydroxy, cyano, aminoacyl, mono-or di-substituted C 1 -C 3 Aminoacyl, C 1- C 3 Alkyl sulfonyl, C 1- C 3 One or two of alkyl sulfoxide groups and oxo (=o) substitution;
R 2 、R 3 、R 4 、R 5 、R 6 each independently is hydrogen, halogen, C 1 -C 6 Alkyl, -O- (CH) 2 )n-R 7
R 7 Is hydrogen, C 1 -C 3 Alkyl, substituted by 1 to 3 groups selected from halogen, cyano, hydroxy, C 1 -C 3 Alkyl, C 1 -C 3 Alkoxy, halo C 1 -C 3 Alkyl, C 3 -C 4 Cycloalkyl, C 2 -C 3 Alkynyl, C 2 -C 3 An aryl or heteroaryl group substituted or unsubstituted by a substituent in alkenyl or-NR 'R', n is an integer from 0 to 3,
the aryl group is a monocyclic or bicyclic group containing 6 to 12 carbon ring atoms and having at least one aromatic ring, the heteroaryl group is a monocyclic or bicyclic group containing 1 to 3 heteroatoms selected from N, O, S as ring atoms and containing 5 to 10 ring atoms,
R ', R' are each independently H or C 1 -C 3 Is a hydrocarbon group.
Alternatively, when m is 1, the compound of formula (I) has the following formula (II),
in the formula (II), A is halogen or C 1 -C 3 An alkyl group; z is NH or O;
R 1 is hydrogen, hydroxy, 4-7 membered heteroalicyclic or-NR a R b
R a 、R b Each independently is hydrogen, C 1 -C 6 Alkyl, C 3 -C 6 Cycloalkyl, C substituted by hydroxy 1 -C 6 Alkyl, quilt C 1- C 3 Alkoxy substituted C 1 -C 6 Alkyl, or C 3- C 6 Cycloalkyl-substituted C 1 -C 6 An alkyl group;
the 4-7 membered heteroalicyclic is a heteroalicyclic containing 1-2 heteroatoms selected from N, O or S, which heteroalicyclic is unsubstituted or C 1 -C 3 Alkyl, C 1 -C 4 Alkanoyl, aminoacyl, hydroxy, cyano, mono-or di-substituted C 1 -C 3 Aminoacyl, C 1- C 3 Alkyl sulfonyl, C 1- C 3 One or two of alkyl sulfoxide groups and oxo (=o) substitution;
R 2 、R 3 、R 4 、R 5 、R 6 each independently is hydrogen, halogen, C 1 -C 6 Alkyl, -O- (CH) 2 )n-R 7
R 7 Is hydrogen, C 1 -C 3 Alkyl, substituted by 1 to 3 groups selected from halogen, cyano, hydroxy, C 1 -C 3 Alkyl, C 1 -C 3 Alkoxy, halo C 1 -C 3 Alkyl, C 3 -C 4 Cycloalkyl, C 2 -C 3 Alkynyl, C 2 -C 3 An aryl or heteroaryl group substituted or unsubstituted by a substituent in alkenyl or-NR 'R', n is an integer from 0 to 3,
the aryl group is a monocyclic or bicyclic group containing 6 to 12 carbon ring atoms and having at least one aromatic ring, the heteroaryl group is a monocyclic or bicyclic group containing 1 to 3 heteroatoms selected from N, O, S as ring atoms and containing 5 to 10 ring atoms,
R ', R' are each independently H or C 1 -C 3 Is a hydrocarbon group.
According to a preferred embodiment, a is Cl, F or methyl; z is NH.
More preferably, a is Cl; z is NH.
According to a preferred embodiment, R 1 Is 4-7 membered heterocyclyl or-NR a R b
R a 、R b Each independently is hydrogen, C 1 -C 3 Alkyl, C substituted by hydroxy 1 -C 3 Alkyl, quilt C 1 -C 3 Alkoxy substituted C 1 -C 3 An alkyl group;
the 4-7 membered heteroalicyclic group is pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, thiomorpholinyl, and the above groups are unsubstituted or substituted with one or two of methyl, ethyl, propyl, isopropyl, aldehyde, acetyl, propionyl, hydroxy, cyano, aminoacyl, methylsulfonyl, ethylsulfonyl, propylsulfonyl, isopropylsulfonyl, methylsulfonyl, ethylsulfoxide, propylsulfoxide, isopropylsulfoxide, oxo (=o).
More preferably, R 1 Is pyrrolidin-1-yl, piperidin-1-yl, 1-methylpiperazin-4-yl, 1-ethylpiperazin-4-yl, morpholinyl, tetrahydrofuran2-yl, tetrahydrofuran3-yl, tetrahydropyran 2-yl, tetrahydropyran 3-yl, thiomorpholinyl, dimethylamino, diethylamino, dipropylamino, diisopropylamino, methylethylamino, methylpropylamino or ethylpropylamino.
Most preferably, R 1 Is dimethylamino.
According to a preferred embodiment, m is 0 or 1,
R 1 is 4-7 membered heteroalicyclic or-NR a R b
R a 、R b Each independently is hydrogen, C 1 -C 3 Alkyl, C 3 -C 6 Cycloalkyl;
the 4-7 membered heteroalicyclic group is pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, tetrahydrofuranyl, tetrahydropyranyl, thiomorpholinyl, and the above groups are unsubstituted or substituted with one or two of methyl, ethyl, propyl, isopropyl, aldehyde, acetyl, propionyl, hydroxy, cyano, aminoacyl, methylsulfonyl, ethylsulfonyl, propylsulfonyl, isopropylsulfonyl, methylsulfonyl, ethylsulfoxide, propylsulfoxide, isopropylsulfoxide, oxo (=o).
More preferably, m is 0 or 1,
R 1 is 1-methyl-pyrrolidin-2-yl, 1-ethyl-pyrrolidin-2-yl, 1-isopropyl-pyrrolidin-2-yl, methylamino, ethylamino, propylamino, isopropylamino, cyclopropylamino, cyclobutylamino, methylisopropylamino, N-methyl-N-cyclopropylamino, N-methyl-N-cyclobutylamino, pyrrolidin-1-yl, piperidin-1-yl, 1-methylpiperazin-4-yl, 1-ethylpiperazin-4-yl, morpholino, tetrahydrofuran 2-yl, tetrahydrofuran 3-yl, tetrahydropyran 2-yl, tetrahydropyran 3-yl, thiomorpholino, dimethylamino, diethylamino, dipropylamino, diisopropylamino, methylethylamino, methylpropylamino or ethylpropylamino.
According to another preferred embodiment, R 2 、R 3 、R 4 、R 5 、R 6 Each independently is hydrogen, fluorine, chlorine, methyl, ethyl, propyl, isopropyl, -O- (CH) 2 )n-R 7 And R is 2 、R 3 、R 4 、R 5 、R 6 At least 3 of which are hydrogen,
R 7 is hydrogen, methyl, ethyl, propyl, isopropyl or is substituted with 1 to 3 groups selected from fluorine, chlorineSubstituted or unsubstituted aryl or heteroaryl groups with substituents of cyano, hydroxy, methyl, ethyl, methoxy, ethoxy, fluoromethyl, fluoroethyl, trifluoromethyl, cyclopropyl, ethynyl, vinyl or-NR 'R', n is an integer from 0 to 3,
r 'and R' are each independently H or methyl,
the aryl is phenyl, and the heteroaryl is pyridyl, pyrimidyl, pyrrolyl, thienyl, furyl and imidazolyl.
More preferably, R 2 、R 3 、R 4 、R 5 、R 6 Each independently hydrogen, fluorine, chlorine, -O- (CH) 2 )n-R 7 And R is 2 、R 3 、R 4 、R 5 、R 6 At least 3 of which are hydrogen,
R 7 aryl or heteroaryl substituted or unsubstituted by 1 to 3 substituents selected from fluorine, chlorine, cyano, hydroxy, methyl, ethyl, methoxy, ethoxy, fluoromethyl, fluoroethyl, trifluoromethyl, n is an integer from 0 to 3,
the aryl is phenyl and the heteroaryl is pyridyl.
In other preferred embodiments, R 2 、R 3 、R 4 、R 5 、R 6 Each independently is hydrogen, fluoro, chloro, phenoxy, 2-fluorophenoxy, 3-fluorophenoxy, 4-fluorophenoxy, pyridin-2-ylmethoxy, pyridin-3-ylmethoxy, pyridin-4-ylmethoxy, 3-fluorobenzyloxy, 2-fluorobenzyloxy, 4-fluorobenzyloxy, 3-chlorobenzyloxy, 2-chlorobenzyloxy, 4-chlorobenzyloxy, and R 2 、R 3 、R 4 、R 5 、R 6 At least 3 of which are hydrogen.
According to another preferred embodiment, R 2 、R 3 、R 5 、R 6 Each independently is hydrogen, fluorine, chlorine, methyl, ethyl, propyl, isopropyl, R 4 Is hydrogen, fluorine, chlorine, methyl, ethyl, propyl, isopropyl, -O- (CH) 2 )n-R 7 And R is 2 、R 3 、R 4 、R 5 、R 6 At least 2 of which are hydrogen,
R 7 is hydrogen, methyl, ethyl, propyl, isopropyl or aryl or heteroaryl substituted or unsubstituted by 1 to 3 substituents selected from fluorine, chlorine, cyano, hydroxy, methyl, ethyl, methoxy, ethoxy, fluoromethyl, fluoroethyl, trifluoromethyl, cyclopropyl, ethynyl, vinyl or-NR 'R', n is an integer from 0 to 3,
r 'and R' are each independently H or methyl,
the aryl is phenyl, and the heteroaryl is pyridyl, pyrimidinyl, pyrrolyl, thienyl, furyl and imidazolyl;
or, more preferably, R 2 、R 3 、R 5 、R 6 Each independently is hydrogen, fluorine, chlorine, R 4 Is hydrogen, fluoro, chloro, phenoxy, 2-fluorophenoxy, 3-fluorophenoxy, 4-fluorophenoxy, pyridin-2-ylmethoxy, pyridin-3-ylmethoxy, pyridin-4-ylmethoxy, 3-fluorobenzyloxy, 2-fluorobenzyloxy, 4-fluorobenzyloxy, 3-chlorobenzyloxy, 2-chlorobenzyloxy, 4-chlorobenzyloxy, and R 2 、R 3 、R 4 、R 5 、R 6 At least 2 of which are hydrogen.
Typical compounds according to the application are as follows:
in another aspect, the application provides a pharmaceutical composition comprising a compound of the application, a pharmaceutically acceptable salt, isomer, solvate, or prodrug thereof, and one or more pharmaceutically acceptable carriers or excipients.
The pharmaceutical compositions of the present application may also comprise one or more other therapeutic agents.
The application also relates to a method of treating EGFR, HER2, etc. kinase-mediated diseases or conditions, including those mentioned previously, comprising administering to a patient (human or other mammal, especially a human) in need thereof a therapeutically effective amount of a compound of the application or a salt thereof.
Detailed Description
Unless otherwise indicated, the following terms used in the present application (including the specification and claims) have the definitions set forth below. In the present application, the use of "or" and "means" and/or "unless stated otherwise. Furthermore, the use of the term "including" and other forms, such as "comprising," "containing," and "having," is not limiting. The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
Unless otherwise specified, alkyl represents a saturated straight-chain, branched-chain hydrocarbon group having the specified number of carbon atoms, the term C 1 -C 10 Alkyl represents an alkyl moiety having 1 to 10 carbon atoms, and C is the same as 1 -C 3 Alkyl represents an alkyl moiety having 1 to 3 carbon atoms, e.g., C 1 -C 6 Alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 3- (2-methyl) butyl, 2-pentyl, 2-methylbutyl, neopentyl, n-hexyl, 2-methylpentyl and the like.
When substituent terms such as "alkyl" are used in combination with other substituent terms, such as in the term "C 1 -C 3 Alkoxy C 1 -C 6 Alkylthio "or" hydroxy-substituted C 1 -C 10 In alkyl ", the term linking substituent (e.g., alkyl or alkylthio) is intended to encompass divalent moieties in which the point of attachment is through the linking substituent. "C 1 -C 3 Alkoxy C 1 -C 6 Examples of alkylthio groups include, but are not limited to, methoxymethylthio, methoxyethylthio, ethoxypropylthio, and the like. "hydroxy-substituted C 1 -C 10 Examples of alkyl "include, but are not limited to, hydroxymethyl, hydroxyethylAnd hydroxyisopropyl, etc.
Alkoxy is an alkyl-O-group formed from a straight or branched chain alkyl group as previously described with-O-, e.g., methoxy, ethoxy, and the like. Similarly, alkylthio is an alkyl-S-group formed from a straight or branched chain alkyl group as previously described with-S-, e.g., methylthio, ethylthio, and the like.
Alkenyl and alkynyl include straight chain, branched alkenyl or alkynyl, the term C 2 -C 6 Alkenyl or C 2 -C 6 Alkynyl means a straight or branched hydrocarbon group having at least one alkenyl or alkynyl group.
The term "haloalkyl", e.g. "halo C 1 -C 10 Alkyl "means a group having one or more halogen atoms, which may be the same or different, on one or more carbon atoms of an alkyl moiety comprising 1 to 10 carbon atoms. "halo C 1 -C 10 Examples of alkyl "groups may include, but are not limited to, -CF 3 (trifluoromethyl), -CCl 3 (trichloromethyl), 1-difluoroethyl, 2-trifluoroethyl, hexafluoroisopropyl, and the like. Similarly, the term "halo C 1 -C 10 Alkoxy "means a halogen represented by said C 1 -C 10 The haloalkyl-O-group formed by alkyl and-O-may be, for example, trifluoromethoxy, trichloromethoxy, and the like.
The term "C 1 -C 3 Acyl "includes formyl (-CHO), acetyl (CH) 3 CO-), acetyl (C) 2 H 5 CO-)。
"cycloalkyl" means a non-aromatic, saturated, cyclic hydrocarbon group containing the indicated number of carbon atoms. For example, the term "(C) 3 -C 6 ) Cycloalkyl "refers to a non-aromatic cyclic hydrocarbon ring having 3 to 6 ring carbon atoms. Exemplary "(C) 3 -C 6 ) Cycloalkyl "includes cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
The term "aryl" means a group or moiety comprising an aromatic, mono-or bicyclic hydrocarbon radical containing from 6 to 12 carbon ring atoms and having at least one aromatic ring. Examples of "aryl" are phenyl, naphthyl, indenyl and indanyl (indanyl). Typically, in the compounds of the present invention, aryl is phenyl.
The term "heteroalicyclic", as used herein, unless otherwise specified, means an unsubstituted or substituted stable 4 to 8 membered non-aromatic monocyclic saturated ring system consisting of carbon atoms and 1 to 3 heteroatoms selected from N, O, S, wherein the N, S heteroatoms may optionally be oxidized and the N heteroatoms may optionally be quaternized. Examples of such heterocycles include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrrolinyl, pyrazolidinyl, pyrazolinyl, imidazolidinyl, imidazolinyl, oxazolinyl, thiazolinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, 1, 3-dioxolanyl, piperidinyl, piperazinyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, 1, 3-dioxanyl, 1, 4-dioxanyl, 1, 3-oxathiolanyl, 1, 3-dithianyl, 1, 4-oxathiolanyl, 1, 4-dithianyl, morpholinyl, thiomorpholinyl.
The term "heteroaryl" as used herein means a group or moiety comprising an aromatic mono-or bi-cyclic radical (containing 5 to 10 ring atoms) comprising 1 to 3 heteroatoms independently selected from nitrogen, oxygen and sulfur. The term also includes bicyclic heteroaryl groups containing an aryl ring moiety fused to a heterocycloalkyl ring moiety or containing a heteroaryl ring moiety fused to a cycloalkyl ring moiety. Unless otherwise specified, represents an unsubstituted or substituted stable 5-or 6-membered monocyclic aromatic ring system, but also an unsubstituted or substituted benzoheteroaromatic ring system of 9 or 10 ring atoms or bicyclic heteroaromatic ring system, which consist of carbon atoms and from 1 to 3 heteroatoms from the group N, O, S, where the N, S heteroatoms may be oxidized and the N heteroatoms may also be quaternized. Heteroaryl groups may be attached to any heteroatom or carbon atom that results in the creation of a stable structure. Illustrative examples of heteroaryl groups include, but are not limited to, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, isothiazolyl, pyridyl, oxo-pyridyl (pyridyl-N-oxide), pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, benzofuryl, isobenzofuryl, 2, 3-dihydrobenzofuryl, 1, 3-benzodioxolyl, dihydrobenzodioxanyl, benzothienyl, indolizinyl, indolyl, isoindolyl indolinyl, benzimidazolyl, dihydrobenzimidazolyl, benzoxazolyl, dihydrobenzoxazolyl, benzothiazolyl, benzisothiazolyl, dihydrobenzisothiazolyl, indazolyl, imidazopyridinyl, pyrazolopyridinyl, benzotriazolyl, triazolopyridinyl, purinyl, quinolinyl, tetrahydroquinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, cinnolinyl, phthalazinyl, quinazolinyl, 1, 5-naphthyridinyl, 1, 6-naphthyridinyl, 1, 7-naphthyridinyl, 1, 8-naphthyridinyl and pteridinyl.
The term "carbonyl" refers to a-C (O) -group. The terms "halogen" and "halo" denote chloro, fluoro, bromo or iodo substituents. "oxo" means an oxygen moiety of a double bond; for example, if directly attached to a carbon atom, a carbonyl moiety is formed (c=o). "hydroxy" is intended to mean the radical-OH. The term "cyano" as used herein refers to the group-CN.
The term "each independently" means that when more than one substituent is selected from a number of possible substituents, those substituents may be the same or different.
It will be appreciated that the compounds of formula I, isomers, crystalline forms or prodrugs and pharmaceutically acceptable salts thereof may exist in solvated as well as unsolvated forms. For example, the solvated form may be a water-soluble form. The present application includes all such solvated and unsolvated forms.
The term "isomer" in the present application is a different compound having the same molecular formula and may include various isomeric forms such as stereoisomers, tautomers and the like. "stereoisomers" are isomers that differ only in the manner in which atoms are spatially arranged. Certain compounds described herein contain one or more asymmetric centers and thus can produce enantiomers, diastereomers, and other stereoisomeric forms that can be defined as (R) -or (S) -depending on absolute stereochemistry. The chemical entities, pharmaceutical compositions and methods of the present application are intended to include all such possible isomers, including racemic mixtures, optically pure forms and intermediate mixtures. The optically active (R) -and (S) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. The optical activity of a compound may be analyzed by any suitable method, including but not limited to chiral chromatography and polarimetry, and the degree of dominance of one stereoisomer over the other can be determined.
The individual isomers (or isomer-enriched mixtures) of the present invention can be resolved using methods known to those skilled in the art. For example, the splitting may be performed as follows: (1) By formation of diastereomeric salts, complexes or other derivatives; (2) By selective reaction with a stereoisomer-specific reagent, for example by enzymatic oxidation or reduction; or (3) by gas-liquid chromatography or liquid chromatography in a chiral environment, for example on a chiral support (e.g. silica gel with chiral ligands bound thereto) or in the presence of a chiral solvent. Those skilled in the art will appreciate that when converting a desired stereoisomer into another chemical entity by one of the separation methods described above, additional steps are required to release the desired form. Alternatively, specific stereoisomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by asymmetric transformation of one enantiomer into another.
When a compound described herein contains an olefinic double bond, it is intended that the compound include the various cis and trans isomers unless specified otherwise.
"tautomers" are structurally different isomers that can be converted to each other by tautomerization. "tautomerization" is a form of isomerization and includes proton transfer or proton transfer tautomerization, which can be considered a subset of acid-base chemistry. "proton transfer tautomerization" or "proton transfer tautomerization" refers to proton transfer accompanied by bond level transformations, often the exchange of single bonds with adjacent double bonds. When tautomerization may occur (e.g., in solution), chemical equilibrium of the tautomers may be reached. One example of tautomerism is keto-enol tautomerism.
The compounds of the present invention as active ingredients, as well as the process for preparing the compounds, are all the subject matter of the present invention. Furthermore, some crystalline forms of the compounds may exist as polymorphs and as such may be included in the present invention. In addition, some compounds may form solvates with water (i.e., hydrates) or common organic solvents, and such solvates are also included within the scope of this invention.
The compounds of the invention may be used in free form for therapy or, where appropriate, in the form of pharmaceutically acceptable salts or other derivatives. As used herein, the term "pharmaceutically acceptable salts" refers to organic and inorganic salts of the compounds of the present invention, which salts are suitable for use in humans and lower animals, without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts of amines, carboxylic acids, phosphonates, and other types of compounds are well known in the art. The salts may be formed by reacting the compounds of the present invention with a suitable free base or acid. Including but not limited to salts with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, perchloric acid, or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, malonic acid, or by using methods well known in the art, such as ion exchange methods. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartate, benzenesulfonates, benzoates, bisulphates, borates, butyrates, camphorites, camphorsulphonates, citrates, digluconates, dodecylsulphates, ethanesulphonates, formates, fumarates, glucoheptonates, glycerophosphate, gluconate, hemisulphates, caprates, hydroiodites, 2-hydroxyethanesulphonates, lactonates, lactates, laurates, laurylsulphates, malates, maleates, methanesulfonates, 2-naphthalenesulphonates, nicotinates, nitrates, oleates, palmitates, pamonates, pectates, persulphates, per 3-phenylpropionates, phosphates, picrates, propionates, stearates, sulphates, thiocyanates, p-toluene sulphonates, undecanoates and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Other pharmaceutically acceptable salts include suitable non-toxic ammonium, quaternary ammonium, and amine cations formed using, for example, halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, lower alkyl sulfonates, and aryl sulfonates.
In addition, the term "prodrug" as used herein means a compound that can be converted in vivo to a compound of the invention. This conversion is effected by hydrolysis of the prodrug in the blood or enzymatic conversion to the parent compound in the blood or tissue.
The pharmaceutical compositions of the invention comprise an additional active agent of a compound described herein or a pharmaceutically acceptable salt thereof, a kinase inhibitor (small molecule, polypeptide, antibody, etc.), an immunosuppressant, an anticancer agent, an antiviral agent, an anti-inflammatory agent, an antifungal agent, an antibiotic, or an anti-vascular hyperproliferative compound; and any pharmaceutically acceptable carrier, adjuvant or excipient.
The compounds of the present invention may be used alone or in combination with one or more other compounds of the present invention or with one or more other agents. When administered in combination, the therapeutic agents may be formulated for simultaneous administration or sequential administration at different times, or the therapeutic agents may be administered as a single composition. By "combination therapy" is meant the use of a compound of the invention in combination with another agent, either by simultaneous co-administration of each agent or by sequential administration of each agent, in either case with the objective of achieving the optimal effect of the drug. Co-administration includes simultaneous delivery of the dosage forms, as well as separate individual dosage forms for each compound. Thus, administration of the compounds of the present invention may be used concurrently with other therapies known in the art, for example, in cancer treatment using radiation therapy or additional therapies such as cytostatic agents, cytotoxic agents, other anticancer agents, etc., to ameliorate cancer symptoms. The invention is not limited to the order of administration; the compounds of the invention may be administered previously, concurrently, or after other anticancer or cytotoxic agents.
For the preparation of the pharmaceutical compositions of this invention, one or more compounds or salts of formula (I) as the active ingredient may be intimately admixed with pharmaceutical carriers according to conventional pharmaceutical compounding techniques, which carriers may take a wide variety of forms depending of the preparation formulated for administration (e.g. oral or parenteral). Suitable pharmaceutically acceptable carriers are well known in the art. A description of some of these pharmaceutically acceptable carriers can be found in the handbook of pharmaceutical excipients, published by the United states society of pharmacy and the United kingdom pharmaceutical society.
The pharmaceutical compositions of the invention may be in a form, for example, suitable for oral administration, such as tablets, capsules, pills, powders, sustained release forms, solutions or suspensions; for parenteral injection such as clear solutions, suspensions, emulsions; or for topical administration such as creams, creams; or as suppositories for rectal administration. The pharmaceutical ingredients may also be suitable in unit dosage form for single use administration of precise dosages. The pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and the compound prepared according to the present invention as an active ingredient, and may include other medical or pharmaceutical preparations, carriers, adjuvants, and the like.
The therapeutic compound may also be administered to a mammal other than a human. The dosage of the drug to be administered to a mammal will depend on the species of the animal and its disease condition or its state of imbalance. The therapeutic compound may be administered to the animal in the form of a capsule, bolus, or medicinal tablet. Therapeutic compounds may also be administered to animals by injection or infusion. We have prepared these pharmaceutical forms according to conventional means that meet the veterinary practice standards. Alternatively, the pharmaceutical composition may be mixed with an animal feed for feeding to the animal, and thus the concentrated feed additive or ready mix may be prepared for mixing with a conventional animal feed.
It is a further object of the present invention to provide a method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a composition comprising a compound of the present invention.
The invention also comprises the use of the compounds of the invention or pharmaceutically acceptable derivatives thereof for the manufacture of a medicament for the treatment of cancers and autoimmune diseases associated with the tyrosine kinases EGFR, HER 2. Agents for such cancers (including non-solid tumors, primary or metastatic cancers, as noted elsewhere herein, and including one or more other treatments that are resistant or refractory to cancer) and other diseases (including, but not limited to, ocular fundus disease, psoriasis, atherosclerosis, pulmonary fibrosis, liver fibrosis, myelofibrosis, etc.). Such cancers include, but are not limited to: non-small cell lung cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, ovarian cancer, cervical cancer, colorectal cancer, melanoma, endometrial cancer, prostate cancer, bladder cancer, leukemia, gastric cancer, liver cancer, gastrointestinal stromal tumor, thyroid cancer, chronic myelogenous leukemia, acute myelogenous leukemia, non-hodgkin lymphoma, nasopharyngeal carcinoma, esophageal cancer, brain tumor, B-cell and T-cell lymphoma, multiple myeloma, biliary sarcoma, cholangiocarcinoma.
Detailed Description
The present application also provides methods for preparing the corresponding compounds, the compounds described herein may be prepared using a variety of synthetic methods, including, but not limited to, the methods described below, and the compounds of the present application or pharmaceutically acceptable salts, isomers or hydrates thereof may be synthesized using the methods described below with synthetic methods known in the art of organic chemical synthesis, or by variations of these methods understood by those skilled in the art, preferred methods include, but are not limited to, the methods described below.
The present application will be described in further detail with reference to specific embodiments in order to make the objects, technical solutions and advantages of the present application more apparent. It should be understood that the specific embodiments described herein are for purposes of illustration only and are not intended to limit the scope of the application. The examples are not to be construed as limiting the specific techniques or conditions described in the literature in this field or as per the specifications of the product. The reagents or apparatus used were conventional products commercially available without the manufacturer's attention. The term "and/or" as used herein includes any and all combinations of one or more of the associated listed items. The examples provided below will better illustrate the application, all temperatures being in degrees celsius unless otherwise indicated. The nomenclature of some of the compounds of the application is translated according to chemdraw nomenclature.
Synthesis of intermediates
Synthesis of acid intermediates: (E) Synthesis method of (E) -4- (dimethylamino) but-2-enoic acid, (E) -4- (diethylamino) but-2-enoic acid and (E) -4- (piperidin-1-yl) but-2-enoic acid
Take (E) -4- (diethylamino) but-2-enoic acid as an example. To the flask was added 2g (27.34 mM) of diethylamine and 20ml of THF under argon, and 5g (27.93 mM) of methyl bromate and 9g (69.63 mM) of diisopropylethylamine under ice bath. Reacting for 3 hours, extracting with ethyl acetate and water; the solvent was removed by rotary evaporation and liquid base (2 g NaOH and 2ml H) was added to 20ml ethanol 2 O), stirring for 3h, adding concentrated hydrochloric acid dropwise to adjust the pH to 1-2, and concentrating under reduced pressure. Acetone (20 ml) was added for recrystallization to give a white solid.
Preparation of (R, E) -3- (1-methylpyrrolidin-2-yl) acryloyl chloride
(R, E) -3- (1-methylpyrrolidin-2-yl) acrylic acid (160 mg,1 mmol) was added to dried dichloromethane (3 ml), oxalyl chloride (130 mg,1 mmol), DMF (1 drop, catalytic amount) was added separately, stirred at room temperature for 3 hours, the reaction system became cloudy, clear, concentrated to give an off-white solid;
synthesis of amine intermediates B1-B5:
synthesis of 4- ((1H-benzo [ d ] [1,2,3] triazol-1-yl) oxy) -5-chloroquinazolin-6-amine
Conditions and reagents (a) EtOH, formamidine acetate, 80℃for 8h; (b) H 2 SO 4 ,HNO 3 -5 ℃, overnight; (c) CH (CH) 3 OH,Fe,NH 4 Cl,80 ℃, reflux/Ni, H 2 ,rt;(d)CH 3 CN,BOP,DBU,rt
Cyclisation with formamidine acetate at 80 ℃ using 2-amino-6-chlorobenzoic acid as starting material gives the compound 5-chloroquinazolin-4 (3H) -one, which is then dissolved in concentrated sulfuric acid solution, nitric acid is added at-10 ℃ and the compound 5-chloro-6-nitroquinazolin-4 (3H) -one is obtained by column chromatography. We then reduced the nitrated product with iron powder in an acidic environment (or reduced the nitrated product in a hydrogen environment using raney nickel) to give the compound 5-chloro-6-aminoquinazolin-4 (3H) -one. Then, by using BOP, 4- ((1H-benzo [ d ] [1,2,3] triazol-1-yl) oxy) -5-chloroquinazolin-6-amine is obtained.
Compound 4- ((1H-benzo [ d ] [1,2,3] triazol-1-yl) oxy) -5-chloroquinazolin-6-amine (100 mg,0.32 mM) and a series of amine compounds (71-90 mg,0.38 mM) were dissolved in i-PrOH (20 ml) solution, stirred at 90℃for 10 minutes, then TsOH (7 mg, 0.03 mM) was added to the mixture and reacted for 5 hours. And the progress of the reaction was monitored by TLC, and at the end of the reaction, water was added and filtered to obtain a solid. Purification by column chromatography (EA: pe=5:1) gives intermediates B1-B5 (brown or green solid), their structures and characterizations are shown in table 1 below.
TABLE 1 Structure, naming and characterization of intermediates B1-B5
Examples
The synthesis method comprises the following steps:
series 2-enoic acid (120 mg,0.9 mM) was dissolved in anhydrous DCM (5 ml) and oxalyl chloride (80. Mu.L, 0.9 mM) was added under ice-bath to react for 3h. The solvent was then evaporated to give an orange solid. An orange solid in DCM (1 ml) was then added under ice-bathB1-B5 (100 mg,0.3 mM) in NMP (2 ml) for 4h. And progress of the reaction was monitored by TLC. Then potassium carbonate solution was added to adjust the pH to 8-9, and the crude product was extracted with DCM. By column chromatography (DCM: CH 3 Oh=30: 1-10: 1) And (5) purifying. Examples 1-7 were synthesized using this procedure one.
The synthesis method II comprises the following steps:
(E) -4-Bromobut-2-enoic acid (150 mg,0.9 mM) was dissolved in anhydrous DCM (5 ml) and oxalyl chloride (80. Mu.L, 0.9 mM) was added under ice-bath to react for 3h. The solvent was then evaporated to give an orange solid. An orange solid in DCM (1 ml) was then added to a solution of B1-B5 (100 mg,0.3 mM) in NMP (2 ml) under ice-bath for 4h. And progress of the reaction was monitored by TLC. Then potassium carbonate solution was added to adjust the pH to 8-9, and the crude product was extracted with DCM. The pure product was isolated by column chromatography (EA: pe=5:1-3:1). Then, pyrrolidine (100-150 mg,1.5 mM) was added to the DMA solution of the product, and stirred at 0℃for 4 hours. And progress of the reaction was monitored by TLC. Then potassium carbonate solution was added to adjust the pH to 8-9, and the crude product was extracted with DCM. The pure product was isolated by column chromatography (DCM: ch3oh=30:1-10:1). Examples 8-10 were synthesized using this method two.
EXAMPLE 1 (E) -N- (5-chloro-4- ((3-chloro-4- (pyridin-2-ylmethoxy) phenyl) amino) quinazolin-6-yl) -4- (dimethylamino) but-2-enamide
White solid; mp 230-232 ℃. 1 H NMR(400MHz,DMSO-d 6 ,δppm):δ9.98(s,1H),9.51(s,1H),8.53(d,J=4.8Hz,1H),8.45(s,1H),8.05(d,J=9.0Hz,1H),7.84–7.81(m,2H),7.68(d,J=9.0Hz,1H),7.52(d,J=7.8Hz,2H),7.33–7.29(m,1H),7.20(d,J=9.0Hz,1H),6.75(dt,J=15.5,5.9Hz,1H),6.45(d,J=15.5Hz,1H),5.24(s,2H),3.03(d,J=5.6Hz,2H),2.13(s,6H). 13 C NMR(101MHz,DMSO-d 6 ,δppm):δ164.35(s),157.23(s),156.74(s),154.22(s),150.81(s),149.61(s),143.11(s),137.63(s),134.45(s),132.84(s),131.97(s),130.12(s),127.46(s),125.59(s), 125.50(s), 123.90(s), 123.59(s), 121.96(s), 121.52(s), 114.67(s), 113.36(s), 71.66(s), 60.20(s), 45.62(s) HRMS (ESI) m/z calculated value C 26 H 24 Cl 2 N 6 O 2 + [M+H] + 523.1416; found, 523.1456.
EXAMPLE 2 (E) -N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) -4- (dimethylamino) but-2-enamide
White solid; mp 235-237 ℃. 1 H NMR(400MHz,DMSO-d 6 ,δppm):δ10.02(s,1H),9.67(s,1H),8.53(s,1H),8.16(d,J=8.9Hz,1H),7.79(s,2H),7.46(s,1H),7.29(t,J=7.8Hz,1H),6.83(dt,J=15.4,5.8Hz,1H),6.53(d,J=15.5Hz,1H),3.10(dd,J=5.8,1.1Hz,2H),2.20(s,6H). 13 C NMR(101MHz,DMSO-d 6 Delta ppm of delta 164.30(s), 153.49(s), 153.07(s), 143.15(s), 134.72(s), 132.00(s), 130.10(s), 127.10(s), 126.08(s), 125.49(s), 125.45(s), 125.40(s), 121.47(s), 120.39(s), 120.22(s), 113.69(s), 109.82(s), 60.24(s), 45.66(s) HRMS (ESI) m/z calculated value C 20 H 18 Cl 2 FN 5 O + [M+H] + 434.0951; found, 434.0923.
EXAMPLE 3 (E) -N- (5-chloro-4- ((3-chloro-4-fluorophenyl) amino) quinazolin-6-yl) -4- (dimethylamino) but-2-enamide
White solid; mp 221-223 ℃. 1 H NMR(400MHz,DMSO-d 6 ,δppm):δ10.01(s,1H),9.66(s,1H),8.59(d,J=16.7Hz,1H),8.16(d,J=9.0Hz,1H),8.04(d,J=4.3Hz,1H),7.79–7.66(m,2H),7.45(t,J=9.0Hz,1H),6.82(dt,J=15.4,5.8Hz,1H),6.54(d,J=15.5Hz,1H),3.10(d,J=5.4Hz,2H),2.20(s,6H). 13 C NMR(101MHz,DMSO-d 6 ,δppm):δ164.43(s),156.77(s),150.82(s),149.61(s),144.08(s),137.60(s),134.56(s),132.03(s),130.11 (s), 127.45(s), 125.56(s), 125.11(s), 123.87(s), 123.57(s), 121.94(s), 121.55(s), 114.71(s), 71.72(s), 47.17(s) HRMS (ESI) m/z calculated value C 20 H 18 Cl 2 FN 5 O + [M+H] + 434.0951; found, 434.0984.
EXAMPLE 4 (E) -N- (5-chloro-4- ((4-phenoxyphenyl) amino) quinazolin-6-yl) -4- (diethylamino) but-2-enamide
White solid; mp.236-238 ℃. 1 H NMR(400MHz,DMSO-d 6 ,δppm):δ10.31(d,J=8.3Hz,1H),9.64(s,1H),8.51(s,1H),8.08(d,J=9.0Hz,1H),7.77–7.71(m,3H),7.40(t,J=7.9Hz,2H),7.12(d,J=7.4Hz,1H),7.07(d,J=8.9Hz,2H),7.04–6.99(m,2H),6.92–6.85(m,1H),6.66(d,J=14.9Hz,1H),3.29(d,J=7.1Hz,2H),2.69(s,4H),1.08(t,J=7.0Hz,6H), 13 C NMR(101MHz,DMSO-d 6 Delta ppm of delta 164.11(s), 157.58(s), 157.21(s), 154.29(s), 153.42(s), 150.16(s), 134.56(s), 134.48(s), 132.17(s), 130.51(s), 130.10(s), 129.11(s), 127.42(s), 125.43(s), 123.69(s), 121.98(s), 119.58(s), 118.59(s), 113.39(s), 63.25(s), 47.01(s), 11.32(s) HRMS (ESI) m/z calculated value C 28 H 28 ClN 5 O 2 + [M+H] + 502.2010; found, 502.2009.
EXAMPLE 5 (E) -N- (5-chloro-4- ((3-chloro-4- (pyridin-2-ylmethoxy) phenyl) amino) quinazolin-6-yl) -4- (piperidin-1-yl) but-2-enamide
White solid; mp 240-242 ℃. 1 H NMR(400MHz,DMSO-d 6 ,δppm):δ10.03(s,1H),9.57(s,1H),8.55(d,J=36.7Hz,2H),8.11(d,J=9.0Hz,1H),7.95–7.83(m,2H),7.73(d,J=8.9Hz,1H),7.58(d,J=8.0Hz,2H),7.42–7.32(m,1H),7.26(d,J=9.0Hz,1H),6.82(dt,J=15.3,5.8Hz,1H),6.52(d,J=15.9Hz,1H),5.30(s,2H),3.10(d,J=5.6Hz,2H),2.36(s,4H),1.59–1.34(m,6H). 13 C NMR(101MHz,DMSO-d 6 Delta ppm of delta 164.35(s), 157.18(s), 156.75(s), 154.17(s), 150.81(s), 149.57(s), 143.00(s), 137.58(s), 134.50(s), 132.86(s), 131.94(s), 127.48(s), 125.56(s), 125.52(s), 123.81(s), 123.57(s), 123.25(s), 121.94(s), 121.56(s), 121.45(s), 114.66(s), 71.71(s), 59.72(s), 54.56(s), 25.94(s), 24.27(s) HRMS (ESI) m/z calculated value C 29 H 28 Cl 2 FN 6 O 2 + [M+H] + 563.1729; found, 563.1796.
EXAMPLE 6 (E) -N- (5-chloro-4- ((4- ((3-fluorobenzyl) oxy) phenyl) amino) quinazolin-6-yl) -4- (piperidin-1-yl) but-2-enamide
White solid; mp 197-199 ℃. 1 H NMR(400MHz,DMSO-d 6 ,δppm):δ10.06(s,1H),9.57(s,1H),8.46(s,1H),8.08(d,J=9.0Hz,1H),7.72(d,J=9.0Hz,1H),7.60(d,J=8.9Hz,2H),7.45(td,J=8.0,6.1Hz,1H),7.30(t,J=7.8Hz,2H),7.16(td,J=8.6,2.2Hz,1H),7.06(d,J=9.0Hz,2H),6.82(dt,J=15.4,5.9Hz,1H),6.53(dd,J=14.1,8.6Hz,1H),5.16(s,2H),3.10(d,J=5.0Hz,2H),2.36(s,4H),1.55–1.36(m,6H). 13 C NMR(101MHz,DMSO-d 6 Delta ppm of delta 164.45(s), 163.48(s), 161.86(s), 155.61(s), 154.35(s), 149.96(s), 143.11(s), 140.57(s), 140.52(s), 134.27(s), 131.91(s), 130.96 (d, j=8.3 Hz), 127.36(s), 125.48(s), 123.86 (d, j=2.5 Hz), 115.38(s), 115.05(s), 114.91(s), 114.59(s), 114.45(s), 113.26(s), 69.01(s), 59.72(s), 54.52(s), 25.86(s), 24.22(s) HRMS (ESI) m/z calculated value C 30 H 29 ClFN 5 O 2 + [M+H] + 546.2072; found, 546.2019.
EXAMPLE 7 (E) -N- (5-chloro-4- ((3-chloro-4-fluorophenyl) amino) quinazolin-6-yl) -4- (piperidin-1-yl) but-2-enamide
White solid; mp 233-235 ℃. 1 H NMR(400MHz,DMSO-d 6 ,δppm):δ10.05(s,1H),9.68(s,1H),8.53(s,1H),8.12(d,J=9.0Hz,1H),8.01(s,1H),7.80–7.61(m,2H),7.49–7.39(m,1H),6.82(dt,J=15.4,5.9Hz,1H),6.52(d,J=15.7Hz,1H),3.10(d,J=5.5Hz,2H),2.35(s,4H),1.45(d,J=52.9Hz,6H). 13 C NMR(101MHz,DMSO-d 6 Delta ppm of delta 164.31(s), 157.07(s), 154.02(s), 150.11(s), 143.17(s), 136.19(s), 134.70(s), 132.07(s), 127.54(s), 125.35 (d, j=17.8 Hz), 124.22(s), 121.24(s), 119.35(s), 117.20(s), 116.98(s), 113.43(s), 59.76(s), 54.62(s), 26.02(s), 24.32(s) HRMS (ESI) m/z calculated value C 23 H 22 Cl 2 FN 5 O + [M+H] + 474.1264; found, 474.1271.
EXAMPLE 8 (E) -N- (5-chloro-4- ((3-chloro-4- (pyridin-2-ylmethoxy) phenyl) amino) quinazolin-6-yl) -4- (pyrrolidin-1-yl) but-2-enamide
White solid; 1 H NMR(400MHz,DMSO-d 6 ,δppm):δ10.04(s,1H),9.57(s,1H),8.61(d,J=3.3Hz,1H),8.53(s,1H),8.12(d,J=9.0Hz,1H),7.96–7.85(m,2H),7.76(d,J=9.0Hz,1H),7.59(d,J=6.8Hz,2H),7.38(dd,J=6.8,5.1Hz,1H),7.28(d,J=9.0Hz,1H),6.86(dt,J=15.3,5.7Hz,1H),6.56(d,J=15.6Hz,1H),5.32(s,2H),3.33–3.28(m,2H),2.66(dd,J=15.3,13.6Hz,4H),1.76(s,4H).MS:548.9[M+H] +
EXAMPLE 9 (E) -N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) -4- (pyrrolidin-1-yl) but-2-enamide
White solid; 1 H NMR(400MHz,DMSO-d 6 ,δppm):δ10.10(s,1H),9.65(s,1H),8.55(s,1H),8.17(d,J=9.0Hz,1H),7.83(dd,J=13.2,8.7Hz,1H),7.57–7.43(m,1H) 7.37-7.24 (m, 1H), 6.86 (dd, j=13.6, 7.7hz, 1H), 5.76 (s, 1H), 5.33 (d, j=4.7 hz, 1H), 3.31 (s, 2H), 2.68 (d, j=1.8 hz, 4H), 1.24 (s, 4H) HRMS (ESI) m/z calculated C 22 H 20 Cl 2 FN 5 O + [M+H] + 460.1107; found, 460.1101.
EXAMPLE 10 (E) -N- (5-chloro-4- ((3-chloro-4-fluorophenyl) amino) quinazolin-6-yl) -4- (pyrrolidin-1-yl) but-2-enamide
White solid; 1 H NMR(400MHz,DMSO-d 6 delta ppm delta 10.04 (s, 1H), 9.67 (s, 1H), 8.58 (s, 1H), 8.15 (d, J=9.0 Hz, 1H), 8.05 (dd, J=6.8, 2.6Hz, 1H), 7.78 (d, J=9.0 Hz, 1H), 7.71 (ddd, J=8.9, 4.3,2.7Hz, 1H), 7.47 (t, J=9.1 Hz, 1H), 6.87 (dt, J=15.4, 5.7Hz, 1H), 6.56 (d, J=15.4 Hz, 1H), 3.32-3.28 (m, 2H), 2.70-2.58 (m, 4H), 1.75 (s, 4H) HRMS (ESI) m/z calculated C 22 H 20 Cl 2 FN 5 O + [M+H] + 460.1107; found, 460.1100.
EXAMPLE 11 (E) -N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) -4- (isopropylamino) but-2-enamide
Step 1) Synthesis of 5-chloro-N- (3-chloro-2-fluorophenyl) -6-nitroquinazolin-4-amine
Suspending 5-chloro-6-nitroquinazolin-4-ol (1 g,4.5 mmol) in thionyl chloride (15 mL), adding DMF (0.5 mL) under stirring at normal temperature, heating the system to 105 ℃ for reaction, after the system is clear (about 3 h), heating the system to 90 ℃ for reflux reaction for 2h, monitoring LCMS (MeOH quenching system), and concentrating the system under reduced pressure directly after the reaction is finished to obtain pale yellow solid; suspending the above obtained solid (1 g,4.4 mmol) in dry acetonitrile (15 mL), dispersing uniformly by ultrasound, slowly dropwise adding 3-chloro-2-fluoroaniline (2.9 g,20 mmol) under ice bath condition, removing ice bath, addingHeating to 50deg.C for 2h, monitoring the reaction by LCMS, concentrating, adding MeOH, pulping, filtering, collecting filter cake to obtain 850mg of target product, and obtaining 53% of product with yield, MS:353[ M+H ]] +
Step 2) Synthesis of 5-chloro-N- (3-chloro-2-fluorophenyl) quinazoline-4, 6-diamine
5-chloro-N- (3-chloro-2-fluorophenyl) -6-nitroquinazolin-4-amine (350 mg,1 mmol), iron powder (280 mg,5 mmol) and ammonium chloride (530 mg,10 mmol) are added to a mixed solution of ethanol (10 ml) and water (1 ml), respectively, stirred and heated to 80 ℃ for reaction for 1 hour, diatomite is filtered, the filtrate is washed with ethyl acetate and saturated sodium bicarbonate respectively, the organic phase is dried, and concentrated to obtain an off-white solid 290mg which is directly used for the next step, MS:323[ M+H ] ] +
Step 3) Synthesis of (E) -4-bromo-N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) but-2-enamide to a solution of 5-chloro-N- (3-chloro-2-fluorophenyl) quinazoline-4, 6-diamine (65 mg,0.2 mmol) in NMP (2 ml) was added a solution of bromocrotonyl chloride (55 mg,0.3 mmol) under ice-water bath conditions, stirred for 30 minutes, quenched by addition of saturated sodium bicarbonate solution, precipitated solid, filtered, washed with ethyl acetate, dried and used directly in the next step; MS 469,471[ M+H ]] +
Step 4) Synthesis of (E) -N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) -4- (isopropylamino) but-2-enamide (0.46 g,1 mmol) but-2-enamide of (E) -4-bromo-N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) (isopropylamine (0.2 g,3 mmol) and diisopropylethylamine (0.3 g,3 mmol) were added to DMF (5 ml), heated to 50deg.C and stirred for 2 hours, cooled, water was added, ethyl acetate, the organic phase was washed with saturated brine, dried, concentrated, and purified by column chromatography to give 150mg of pale yellow solid product in 33 yield;
1 H NMR(400MHz,DMSO-d 6 )δ10.18(s,1H),9.66(s,1H),8.56(s,1H),8.14(d,J=9.0Hz,1H),7.82(d,J=8.9Hz,2H),7.49(t,J=7.6Hz,1H),7.31(t,J=8.1Hz,1H),6.89(dt,J=15.4,5.8Hz,1H),6.60(d,J=15.4Hz,1H),3.63(s,2H),3.33(br,1H),3.06(s,1H),1.15(d,J=6.3Hz,6H).MS:448[M+H] +
EXAMPLE 12 (E) -N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) -4- (cyclopropylamino) but-2-enamide
The synthesis was carried out in the same manner as in example 11 except that cyclopropylamine was used instead of isopropylamine of step 4); 1 H NMR(400MHz,DMSO-d 6 )δ10.00(s,1H),9.65(s,1H),8.54(s,1H),8.16(d,J=9.0Hz,1H),7.89–7.77(m,2H),7.49(ddd,J=8.3,6.8,1.6Hz,1H),7.30(td,J=8.1,1.4Hz,1H),6.92(dt,J=15.4,5.3Hz,1H),6.49(dt,J=15.5,1.8Hz,1H),3.43(dd,J=5.4,1.8Hz,2H),3.33(br,1H),2.16(tt,J=6.7,3.6Hz,1H),0.43-.039(m,2H),0.32–0.24(m,2H).MS:446[M+H] +
EXAMPLE 13 (E) -N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) -4- (methylamino) but-2-enamide monotrifluoroacetate salt
The synthesis was carried out in the same manner as in example 11 except that methylamine hydrochloride was used instead of isopropylamine in step 4) to carry out the reaction, and the product of monotrifluoroacetate was obtained by purifying the prepared liquid phase;
1 H NMR(400MHz,DMSO-d 6 )δ10.29(s,1H),9.71(s,1H),8.73(s,2H),8.57(s,1H),8.12(d,J=9.1Hz,1H),7.82(s,2H),7.49(s,1H),7.31(d,J=7.2Hz,1H),6.82(dt,J=15.5,6.5Hz,1H),6.65(d,J=15.5Hz,1H),3.84(q,J=5.9Hz,2H),2.67–2.53(m,3H).MS:420[M+H] +
EXAMPLE 14 (E) -N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) -4- (cyclobutylamino) but-2-enamide
/>
The synthesis was carried out in the same manner as in example 11 except that the reaction was carried out using cyclobutylamine instead of isopropylamine in step 4); 1 H NMR(400MHz,DMSO-d 6 )δ10.01(s,1H),9.66(s,1H),8.50(s,1H),8.14(d,J=9.0Hz,1H),7.78(s,2H),7.45(t,J=7.5Hz,1H),7.28(t,J=8.2Hz,1H),6.89(dt,J=15.4,5.3Hz,1H),6.51(d,J=15.4Hz,1H),3.29(d,J=5.3Hz,2H),3.33(br,1H),3.20(p,J=7.6Hz,1H),2.11(q,J=8.5,7.9Hz,2H),1.76-1.55(m,4H).MS:460[M+H] +
EXAMPLE 15 (E) -N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) -4- (isopropyl (methyl) amino) but-2-enamide
The synthesis was carried out in the same manner as in example 11 except that isopropylmethylamine was used instead of isopropylamine in step 4); 1 H NMR(400MHz,DMSO-d 6 )δ10.05(s,1H),9.66(s,1H),8.54(s,1H),8.16(d,J=9.1Hz,1H),7.87–7.75(m,2H),7.46(s,1H),7.28(t,J=8.2Hz,1H),6.83(dt,J=15.4,5.7Hz,1H),6.53(dt,J=15.4,1.8Hz,1H),3.21(dd,J=5.8,1.6Hz,2H),2.83(p,J=6.6Hz,1H),2.16(s,3H),0.99(d,J=6.5Hz,6H).MS:462[M+H] +
EXAMPLE 16 (E) -N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) -4- (cyclobutyl (methyl) amino) but-2-enamide
The synthesis was carried out in the same manner as in example 11 except that cyclobutylmethylamine was used instead of isopropylamine in step 4); 1 H NMR(400MHz,DMSO-d 6 )δ10.05(s,1H),9.66(s,1H),8.54(s,1H),8.17(d,J=9.1Hz,1H),7.88–7.76(m,2H),7.48(s,1H),7.29(t,J=8.1Hz,1H),6.84(dt,J=15.4,5.9Hz,1H),6.52(d,J=15.4Hz,1H),3.09–3.02(m,2H),2.93–2.81(m,1H),2.06(s,3H),1.99(dd,J=9.8,7.2Hz,2H),1.80(tt,J=11.2,8.9Hz,2H),1.61(ddt,J=18.1,10.4,8.1Hz,2H).MS:474[M+H] +
EXAMPLE 17 (E) -N- (5-chloro-4- ((3-chloro-2-fluorophenyl) amino) quinazolin-6-yl) -4- (cyclopropyl (methyl) amino) but-2-enamide
The synthesis was carried out in the same manner as in example 11 except that cyclopropylmethylamine was used instead of isopropylamine of step 4); 1 H NMR(400MHz,DMSO-d 6 )δ10.02(s,1H),9.66(s,1H),8.50(s,1H),8.14(s,1H),7.77(s,2H),7.48–7.42(m,1H),7.28(s,1H),6.85(s,1H),6.49(s,1H),4.11(s,1H),3.16(s,2H),2.29(s,3H),0.46(s,2H),0.35(s,2H).MS:460[M+H] +
EXAMPLE 18 (E) -N- (5-chloro-4- ((3-chloro-4- (pyridin-2-ylmethoxy) phenyl) amino) quinazolin-6-yl) -4- (cyclopropyl (methyl) amino) but-2-enamide
The synthesis was carried out in the same manner as in example 11 except that 3-chloro-4- (pyridin-2-ylmethoxy) aniline was used in place of 3-chloro-2-fluoroaniline in step 1), and cyclopropylmethylamine was used in place of isopropylamine in step 4);
1 H NMR(400MHz,DMSO-d 6 )δ10.00(s,1H),9.57(s,1H),8.60(ddd,J=4.8,1.8,0.9Hz,1H),8.52(s,1H),8.12(d,J=9.0Hz,1H),7.94–7.84(m,2H),7.75(d,J=9.0Hz,1H),7.62–7.54(m,2H),7.38(ddd,J=7.6,4.8,1.2Hz,1H),7.27(d,J=9.0Hz,1H),6.86(dt,J=15.4,6.1Hz,1H),6.49(dt,J=15.4,1.6Hz,1H),5.31(s,2H),3.38–3.30(m,2H),2.29(s,3H),1.77(tt,J=6.6,3.6Hz,1H),0.46(dt,J=6.1,3.0Hz,2H),0.39–0.31(m,2H).MS:549[M+H] +
EXAMPLE 19 (E) -N- (5-chloro-4- ((3-chloro-4- (pyridin-2-ylmethoxy) phenyl) amino) quinazolin-6-yl) -4- (cyclobutyl (methyl) amino) but-2-enamide
The synthesis was carried out in the same manner as in example 11 except that 3-chloro-4- (pyridin-2-ylmethoxy) aniline was used in place of 3-chloro-2-fluoroaniline in step 1), and cyclobutylmethylamine was used in place of isopropylamine in step 4);
1 H NMR(400MHz,DMSO-d 6 )δ10.01(s,1H),9.57(s,1H),8.60(dt,J=4.9,1.3Hz,1H),8.52(s,1H),8.12(d,J=9.0Hz,1H),7.94–7.84(m,2H),7.74(d,J=9.0Hz,1H),7.62–7.55(m,2H),7.37(ddd,J=7.7,4.8,1.2Hz,1H),7.27(d,J=9.0Hz,1H),6.83(dt,J=15.4,6.0Hz,1H),6.52(dt,J=15.4,1.8Hz,1H),5.31(s,2H),3.05(dd,J=6.0,1.6Hz,2H),2.93–2.78(m,1H),2.06(s,3H),2.03–1.95(m,2H),1.87–1.72(m,2H),1.60(tdd,J=15.0,7.0,4.9Hz,2H).MS:563[M+H] +
EXAMPLE 20 (R, E) -N- (5-chloro-4- ((3-chloro-4- (pyridin-2-ylmethoxy) phenyl) amino) quinazolin-6-yl) -3- (1-methylpyrrolidin-2-yl) acrylamide
The synthesis was carried out in the same manner as in example 11 except that 3-chloro-4- (pyridin-2-ylmethoxy) aniline was used in place of 3-chloro-2-fluoroaniline in step 1), and (R, E) -3- (1-methylpyrrolidin-2-yl) acryloyl chloride was used in place of bromocrotonyl chloride in step 3);
1 H NMR(400MHz,DMSO-d 6 )δ10.00(s,1H),9.57(s,1H),8.60(d,J=4.8Hz,1H),8.52(s,1H),8.13(d,J=9.0Hz,1H),7.89(dd,J=14.2,5.4Hz,2H),7.75(d,J=9.0Hz,1H),7.58(d,J=8.2Hz,2H),7.37(t,J=6.1Hz,1H),7.27(d,J=8.9Hz,1H),6.72(dd,J=15.3,7.5Hz,1H),6.52(d,J=15.3Hz,1H),5.31(s,2H),3.04(s,1H),2.79(q,J=8.0Hz,1H),2.22(s,3H),2.18(d,J=9.2Hz,1H),2.02(dq,J=14.4,8.3,7.8Hz,1H),1.74(q,J=8.4Hz,2H),1.61(d,J=18.1Hz,1H).MS:549[M+H] +
EXAMPLE 21 (E) -N- (5-chloro-4- ((3-chloro-2, 4-difluorophenyl) amino) quinazolin-6-yl) -4- (dimethylamino) but-2-enamide
The synthesis was carried out in the same manner as in example 11 except that 2, 4-difluoro-3-chloroaniline was used instead of 3-chloro-2-fluoroaniline in step 1), dimethylamine hydrochloride was used instead of isopropylamine in step 4);
1 H NMR(400MHz,DMSO-d 6 )δ10.02(s,1H),9.58(s,1H),8.50(s,1H),8.17(d,J=9.0Hz,1H),7.78(s,2H),7.40(s,1H),6.83(dt,J=15.5,5.8Hz,1H),6.53(dt,J=15.5,1.7Hz,1H),3.10(dd,J=5.9,1.6Hz,2H),2.21(s,6H).MS:452[M+H] +
EXAMPLE 22 (E) -N- (5-chloro-4- ((3-chloro-2, 4-difluorophenyl) amino) quinazolin-6-yl) -4- (isopropylamino) but-2-enamide
The synthesis was carried out in the same manner as in example 11 except that 2, 4-difluoro-3-chloroaniline was used instead of 3-chloro-2-fluoroaniline in step 1) to carry out the reaction;
1 H NMR(400MHz,DMSO-d 6 )δ10.04(s,1H),9.59(s,1H),8.46(s,1H),8.13(d,J=9.0Hz,1H),7.75(d,J=14.5Hz,2H),7.44–7.34(m,1H),6.91(dt,J=15.4,5.3Hz,1H),6.54(dt,J=15.4,1.8Hz,1H),3.43(dd,J=5.4,1.8Hz,2H),3.33(br,1H),2.82(p,J=6.2Hz,1H),1.05(d,J=6.2Hz,6H).MS:466[M+H] +
EXAMPLE 23 Synthesis of (E) -N- (4- ((3-chloro-2-fluorophenyl) amino) -5-methylquinazolin-6-yl) -4- (dimethylamino) but-2-enamide
Step 1) 5-Methylquinazolin-4-ol
2-amino-6-methyl-benzoic acid (4.53 g,30 mmol) and formamidine acetate (3.12 g,30.00 mmol) were added to ethanol (40 mL), heated to 80℃for 24h at reflux, LCMS monitored, and after completion of the reaction, the system was cooled to room temperature, and a large amount of solids were precipitated. Filtering, and washing the filter cake with a small amount of petroleum ether. Collecting and drying to obtain the product 5-methyl quinazoline-4-alcohol (3.66 g,22.85mmol, yield 76.17%); MS 161[ M+H ] ] +
Step 2) 5-methyl-6-nitroquinazolin-4-ol
5-Methylquinazolin-4-ol (3.66 g,22.85 mmol) was slowly added to H at ambient temperature 2 SO 4 (30 mL), cooling the ice salt bath to about-20deg.C, and adding KNO in portions 3 (2.54 g,25.14 mmol) (about 20 min), and controlling the system temperature below-10 ℃. The system is slowly heated to 10 ℃ for reaction for 2 hours, HPLC monitoring is carried out, the system is slowly poured into crushed ice after the reaction is finished, a large amount of solids are separated out, the filtration is carried out, a filter cake is washed with water for 3 times, the collection and the drying are carried out, and the product 5-methyl-6-nitroquinazolin-4-ol (3.2 g,15.60mmol, yield 68.26%) is obtained.
1 H NMR(400MHz,DMSO-d 6 )δ8.45(s,1H),8.14(d,J=8.9Hz,1H),7.92(s,1H)7.63(d,J=8.9Hz,1H),2.77(s,3H).
Step 3) N- (3-chloro-2-fluorophenyl) -5-methyl-6-nitroquinazolin-4-amine
Suspending 5-methyl-6-nitroquinazolin-4-ol (1 g,4.87 mmol) in thionyl chloride (15 mL), adding DMF (0.5 mL) under stirring at normal temperature, heating the system to 100 ℃ for reaction, after the system is clear (about 3 h), refluxing for 2h, monitoring LCMS (MeOH quenching system), and concentrating the system under reduced pressure directly after the reaction is finished to obtain brown solid; the solid (1 g,4.47 mmol) obtained above was suspended in 1, 2-dichloromethane (15 mL), the dispersion was made uniform by sonication, 3-chloro-2-fluoroaniline (2.60 g,17.89 mmol) was slowly added dropwise under ice bath conditions, the ice bath was removed after the dropwise addition, the reaction was heated to 50℃for 1h, LCMS was monitored to complete the reaction, the solvent was distilled off under reduced pressure, the remainder was added with MeOH, the dispersion was made uniform by sonication, and the filter cake was collected to give the product (990 mg,2.98mmol, yield 66.54%).
Step 4) N- (3-chloro-2-fluorophenyl) -5-methyl quinazoline-4, 6-diamine
N- (3-chloro-2-fluoro-phenyl) -5-methyl-6-nitro-quinolin-4-amine (990 mg,2.98 mmol) was suspended in methanol (10 mL) and Raney Ni (34.93 mg, 595.10. Mu. Mol) was added to displace H 2 The atmosphere was stirred for 30min at room temperature for 3 times, the system was gradually dissolved and clarified, and monitored by LCMS. After the completion of the reaction, the mixture was filtered through celite, and the filtrate was concentrated to give the product (890 mg,2.94mmol, yield 98.80%).
(E) -4-bromo-N- (4- ((3-chloro-2-fluorophenyl) amino) -5-methyl quinazolin-6-yl) but-2-enamide
N- (3-chloro-2-fluorophenyl) -5-methyl quinazoline-4, 6-diamine (50 mg,165.16 mu mol) is added into NMP (3 ml), under normal temperature stirring, (E) -bromocrotonyl chloride (45 mg,250 mu mol) acetonitrile solution is added dropwise for reaction for 15 minutes, LCMS monitoring, the system is quenched by excessive saturated sodium bicarbonate water solution, pH is regulated to be approximately equal to 8, a large amount of solid is precipitated, the solid is filtered, washed by ethyl acetate, and 700mg of crude product is obtained for standby, MS:449,451[ M+H ] is obtained after the reaction is completed] +
(E) -N- (4- ((3-chloro-2-fluorophenyl) amino) -5-methylquinazolin-6-yl) -4- (dimethylamino) but-2-enamide (E) -4-bromo-N- (4- ((3-chloro-2-fluorophenyl) amino) -5-methylquinazolin-6-yl) but-2-enamide (50 mg, 165.16. Mu. Mol) was added to DMF (3 mL), dimethylamine hydrochloride, diisopropylethylamine were added separately with stirring at room temperature, heated to 50℃for 2 hours, LCMS was monitored for reaction completion, the system was quenched with an excess of saturated aqueous sodium bicarbonate solution, pH was adjusted to approximately 8, a large amount of solids was precipitated, filtered, and the solid preparation plate was purified (DCM/=10:1) to give the target product (25 mg, 60.40. Mu. Mol, yield 36.57%).
1 H NMR(400MHz,DMSO-d 6 )δ9.76(s,1H),8.89(s,1H),8.41(s,1H),7.69(br,2H),7.19(br,3H),6.76(dt,J=15.4,5.8Hz,1H),6.40(d,J=15.5Hz,1H),3.08(d,J=5.9Hz,2H),2.72(s,3H),2.19(s,6H).MS:414[M+H] +
EXAMPLE 24 (E) -N- (4- ((3-chloro-2-fluorophenyl) amino) -5-methylquinazolin-6-yl) -4- (isopropylamino) but-2-enamide
The synthesis was carried out in the same manner as in example 23 except that isopropylamine was used instead of dimethylamine hydrochloride in step 6); 1 H NMR(400MHz,DMSO-d 6 )δ9.73(s,1H),8.91(s,1H),8.48-8.44(m,1H),7.81–7.51(m,2H),7.40–7.02(m,3H),6.90–6.78(m,1H),6.40(d,J=16.2Hz,1H),3.33(br,1H),2.82-2.79(m,4H),2.71(d,J=3.6Hz,2H),1.02(d,J=6.2Hz,6H).MS:428[M+H] +
EXAMPLE 25 (E) -N- (4- ((3-chloro-2-fluorophenyl) amino) -5-methylquinazolin-6-yl) -4- (cyclopropylamino) but-2-enamide
The synthesis was carried out in the same manner as in example 23 except that cyclopropylamine was used instead of the dimethylamine hydrochloride in step 6); 1 H NMR(400MHz,DMSO-d 6 )δ9.73(s,1H),8.06(s,1H),7.67(d,J=8.3Hz,1H),7.39(s,2H),7.33–7.12(m,3H),6.85(dt,J=15.4,5.1Hz,1H),6.39(d,J=15.4Hz,1H),3.38–3.33(m,4H),2.80–2.67(s,3H),1.01(d,J=6.2Hz,4H).MS:426[M+H] +
EXAMPLE 26 (E) -N- (4- ((3-chloro-2-fluorophenyl) amino) -5-methyl quinazolin-6-yl) -4- (methylamino) but-2-enamide
The synthesis was carried out in the same manner as in example 23 except that methylamine hydrochloride was used in place of dimethylamine hydrochloride in step 6); 1 H NMR(400MHz,DMSO-d 6 )δ9.83(s,1H),8.25(s,1H),7.64(br,3H),7.19(s,3H),6.80(d,J=15.6Hz,1H),6.43(d,J=15.5Hz,1H),3.45(br,3H),2.72(s,3H),2.40(s,3H).MS:400[M+H] +
EXAMPLE 27 (E) -N- (4- ((3-chloro-2-fluorophenyl) amino) -5-methyl quinazolin-6-yl) -4- (cyclobutylamino) but-2-enamide
The synthesis was carried out in the same manner as in example 23 except that cyclobutylamine was used instead of the dimethylamine hydrochloride in step 6); 1 H NMR(400MHz,DMSO-d 6 )δ9.74(s,1H),8.91(s,1H),8.38(br,1H),7.64(s,2H),7.43–6.90(m,3H),6.82(dt,J=15.6,5.3Hz,1H),6.39(d,J=15.4Hz,1H),3.33-3.27(m,3H),3.20(d,J=7.7Hz,1H),2.72(s,3H),2.11(q,J=8.2,5.8Hz,2H),1.79–1.48(m,4H).MS:440[M+H] +
EXAMPLE 28 (E) -N- (4- ((3-chloro-2-fluorophenyl) amino) -5-methylquinazolin-6-yl) -4- (isopropyl (methyl) amino) but-2-enamide
The synthesis was carried out in the same manner as in example 23 except that isopropylmethylamine was used instead of dimethylamine hydrochloride in step 6); 1 H NMR(400MHz,DMSO-d 6 )δ9.95-9.66(m,1H),8.91-8.45(m,1H),7.84(d,J=23.3Hz,1H),7.68–7.58(m,2H),7.36–6.99(m,3H),6.75(d,J=15.2Hz,1H),6.39(d,J=17.9Hz,1H),3.20(d,J=5.7Hz,2H),2.83(q,J=6.5Hz,1H),2.72(d,J=22.2Hz,3H),2.16(s,3H),0.99(d,J=6.5Hz,6H).MS:442[M+H] +
EXAMPLE 29 (R, E) -N- (4- ((3-chloro-2-fluorophenyl) amino) -5-methyl quinazolin-6-yl) -3- (1-methyl pyrrolidin-2-yl) acrylamide
The synthesis was carried out in the same manner as in example 23 except that (R, E) -3- (1-methylpyrrolidin-2-yl) acryloyl chloride was used in place of the bromocrotonyl chloride of step 5);
1 H NMR(400MHz,DMSO-d 6 )δ9.65(s,1H)8.46(s,1H),7.90–7.80(m,1H),7.61-7.46(m,2H),7.36-6.99(m,3H),6.66(td,J=19.5,17.8,7.4Hz,1H),6.39(dd,J=27.3,15.4Hz,1H),3.03(d,J=8.5Hz,1H),2.26–2.13(m,6H),2.00(d,J=11.3Hz,2H),1.77(td,J=6.6,6.0,2.8Hz,2H),1.59(s,2H).MS:440[M+H] +
EXAMPLE 30 (E) -N- (4- ((3-chloro-2, 4-difluorophenyl) amino) -5-methylquinazolin-6-yl) -4- (dimethylamino) but-2-enamide
The synthesis was carried out in the same manner as in example 23 except that 3-chloro-2-fluoroaniline in step 3) was replaced with 3-chloro-2, 4-difluoroaniline to obtain the product; 1 H NMR(400MHz,DMSO-d 6 )δ9.96(s,1H),9.74(s,1H),8.86(s,1H),8.41(s,1H),7.78(s,1H),7.62(s,1H),6.99(s,1H),6.80–6.71(m,1H),6.38(d,J=15.4Hz,1H),3.08(d,J=5.9Hz,2H),2.71(s,3H),2.19(s,6H).MS:432[M+H] +
EXAMPLE 31 (E) -N- (4- ((3-chloro-4-fluorophenyl) amino) -5-methyl quinazolin-6-yl) -4- (dimethylamino) but-2-enamide
The synthesis was carried out in the same manner as in example 23 except that 3-chloro-4-fluoroaniline was used in place of 3-chloro-2-fluoroaniline in step 3) to carry out the reaction;
1 H NMR(400MHz,DMSO-d 6 )δ9.90(s,1H),8.99(s,1H),8.51(s,1H),8.02(dd,J=6.9,2.6Hz,1H),7.83(d,J=8.8Hz,1H),7.72–7.59(m,2H),7.43(t,J=9.1Hz,1H),6.78(dt,J=15.4,5.9Hz,1H),6.45(d,J=15.5Hz,1H),3.12–3.06(m,2H),2.72(s,3H),2.20(s,6H).MS:414[M+H] +
EXAMPLE 32 (E) -N- (4- ((3-chloro-2, 4-difluorophenyl) amino) -5-methylquinazolin-6-yl) -4- (isopropylamino) but-2-enamide
The synthesis was carried out in the same manner as in example 23 except that 3-chloro-4-fluoroaniline was used instead of 3-chloro-2-fluoroaniline in step 3), and isopropylamine was used instead of dimethylamine hydrochloride in step 6) to obtain the product;
1 H NMR(400MHz,DMSO-d 6 )δ9.91(s,1H),8.33(s,1H),7.65(br,2H),7.28(br,3H),6.84(dt,J=15.4,5.7Hz,1H),6.48(d,J=15.4Hz,1H),3.53(d,J=5.7Hz,2H),3.33(br,1H),2.96(q,J=6.3Hz,1H),2.72(s,3H),1.11(d,J=6.3Hz,6H).MS:446[M+H] +
experimental example 1 Small molecule Compounds inhibit EGFR WT Assay for HER2 kinase Activity
Reagent and consumable: ULIght TM-labeled Ploy GT Peptide (Perkin Elmer, catalog number TRF-0100-M); ulight (TM) -clamped JAK-1 (Try 1023) Peptide (Perkin Elmer, catalog number TRF-0121-M); eu-W1024-labeled Anti-Phosphotyrosine Antibody (PT 66) (Perkin Elmer, catalog number AD 0068); 10 Xdetection Buffer (Perkin Elmer, catalog number CR 97-100); her2 kinase (Carna Biosciences, catalog number 08-016); EGFR kinase (Carna Biosciences, catalog number 08-115); HEPES (GIBCO, catalog number 15630-080); EGTA (Sigma, catalog number 03777-10G); EDTA (Sigma, catalog number EDS-100G); mgCl 2 (Sigma, catalog number 63069-100 ML); DTT (Sigma, catalog number 43816-10 ML); tween-20 (Sigma, catalog number P7949-100 ML); DMSO (Life Science, catalog number 0231-500 ML); 384 well plates (Perkin Elmer, catalog No. 607290); multifunctional reader (Perkin Elmer, catalog number Envision)
Compound solution configuration: test compounds were dissolved in DMSO to make a 10mM stock solution. The compound was diluted to 0.25mM (100-fold final concentration of dilution) in DMSO before use, and 3-fold concentration gradient dilutions, 11 gradients were made. The solution is diluted to 4 times of the final concentration by buffer solution when the medicine is added.
HER2 kinase assay: preparing a buffer solution, and preparing 40nM 4 XHer 2 kinase solution, 40. Mu.M 4 XATP solution, 400nM 4 XULTight TM Labeled Ploy GT Peptide substrate solution. After the preparation was completed, the enzyme was mixed with compounds of different concentrations prepared by pre-dilution, and left at room temperature for 5 minutes, with duplicate wells being set for each concentration. The corresponding substrate and ATP were added and reacted at room temperature for 120 minutes (with a negative and positive control set). After the completion of the reaction, PT66 detection antibody was added, and the reaction was incubated at room temperature for 60 minutes and then detected by Envision.
EGFR WT Kinase detection: preparing a buffer solution, and preparing 3.48nM 4 XEGFR kinase solution, 600. Mu.M 4 XATP solution, 400nM 4 XULTight using the buffer solution TM -a substrate solution of labeled JAK-1 (Try 1023) Peptide. After the preparation was completed, the enzyme was mixed with compounds of different concentrations prepared by pre-dilution, and left at room temperature for 5 minutes, with duplicate wells being set for each concentration. The corresponding substrate and ATP were added and reacted at room temperature for 120 minutes (with a negative and positive control set). After the completion of the reaction, PT66 detection antibody was added, and the reaction was incubated at room temperature for 60 minutes and then detected by Envision.
And (3) data calculation: well read and inhibition ratio were calculated using Excel table, well read = 10000 (well EU 665)/(well EU 615), inhibition ratio = [ (positive control well read-experimental well read)/(positive control well read-negative control well read)]*100%. Compound concentration and corresponding inhibition rate were input into GraphPad Prism treatment calculation IC 50 Values.
Table 2 tests show that the compounds of the application inhibit EGFR WT And HER2 tyrosine kinase activity, and in particular, some of the compounds exhibit potent inhibitory effects. The test results are summarized in table 2 below.
Table 2 shows EGFR pairs with some of the compounds of the application WT And HER2 tyrosine kinase inhibitory activity, wherein A represents IC 50 Less than or equal to 50nM, B represents IC 50 Greater than 50nM but less than or equal to 500nM, C represents IC 50 Greater than 500nM but less than or equal to 5000nM, D represents IC 50 Above 5000nM, NT indicates no relevant result.
TABLE 2 determination of EGFR and HER2 kinase inhibitory Activity of the Compounds of the application
Experimental example 2 test of small molecule Compounds for inhibiting cell proliferation
The present application employs CCK8 methods to examine the in vitro antiproliferative activity of the compounds of the present application on in vitro cultured BT474, NCI-N87, HCC-827 and Ba/F3EGFRvIII cell lines.
Reagent and consumable: RPMI1640 (ThermoFisher, catalog number C11875500 BT); DMEM (thermo fisher, C11995500 BT); fetal bovine serum (Hyclone, cat# SV 30087.03); 0.25% trypsin-EDTA (ThermoFisher, cat. 25200072); penicillin-streptomycin (Hyclone, catalog number SV 30010); DSMO (Life Science, catalog number 0231-500 ML); CCK8 test kit (Dojindo, catalog number CK 04-100); 96-well plates (Corning, catalog No. 3599); multifunctional reader (Perkin Elmer, catalog number Envision)
Cell line: BT474 (from cell bank of China academy of sciences), NCI-N87 (from ATCC) and HCC-827 (from ATCC), ba/F3EGFRvIII from Kang Yuanbo Biotechnology (Beijing); wherein BT474, NCI-N87 and Ba/F3EGFRvIII were cultured in RPMI1640 medium containing 10% fetal bovine serum, 100U/mL penicillin, 100. Mu.g/mL streptomycin, and HCC-827 was cultured in DMEM medium containing 10% fetal bovine serum, 100U/mL penicillin, 100. Mu.g/mL streptomycin.
The specific experimental method comprises the following steps:
1. the tested compound is dissolved by DSMO to form a storage solution, and the storage solution is subjected to gradient dilution, and then the storage solution is diluted by a corresponding culture medium to obtain a solution with 20 times of working concentration.
2. Cells in the logarithmic growth phase were diluted with culture medium to adjust to a specific cell concentration, and 80. Mu.L of cell suspension was added to a 96-well plate so that cell densities of BT474, NCI-N87, HCC-827 and Ba/F3EGFRvIII were 10000 cells/well, 8000 cells/well, 5000 cells/well and 8000 cells/well, respectively. Culturing overnight at 37deg.C in a 5% carbon dioxide incubator. Wherein Ba/F3EGFRvIII cells directly enter the next dosing treatment, and BT474, NCI-N87 and HCC-827 are required to be cultured overnight for adherence and then are subjected to the dosing treatment.
3. mu.L of drug solution was added to each well of the 96-well plate inoculated with cells. The highest concentration of the tested compound is 10 mu M,10 concentrations, 4-fold gradient dilution and double-compound hole. And a control group without adding medicines is arranged at the same time.
4. After 72 hours of cell culture, cell viability was measured with CCK 8. Dose-response curves were made and IC calculated using GraphPad Prism software 50
Table 3 shows the results of antiproliferative activity assays of representative compounds of the present application on BT474, NCI-N87, HCC-827 and Ba/F3EGFRvIII cells. Wherein A represents IC 50 Less than or equal to 50nM, B represents IC 50 Greater than 50nM but less than or equal to 500nM, C represents IC 50 Greater than 500nM but less than or equal to 5000nM, D represents IC 50 Above 5000nM, NT indicates no relevant result.
TABLE 3 antiproliferative activity assays of representative compounds of the application on BT474, NCI-N87, HCC-827 and Ba/F3EGFRvIII cells
/>
The results in Table 3 show that the above-mentioned compounds of the present application exhibit excellent to good antitumor proliferation activity against BT474, NCI-N87 and Ba/F3EGFRvIII cells. Furthermore, the compounds of the present application also exhibit excellent proliferation-inhibiting activity against HCC-827 cell line.
Experimental example 3 pharmacokinetic test of Small molecule Compounds
This test investigated the pharmacokinetic profile of a compound of the present application by administering a single oral and intravenous administration of a portion of the compound of the present application to SD rats and the ability of the compound of the present application to penetrate the blood brain barrier. At the same time, pyrroltinib (PYROTINIB), lenatinib (NERATINIB) were tested accordingly and compared with the compounds of the application.
Reagents, apparatus and animals for use in (I)
TABLE 4 test reagents
TABLE 5 test instrument
/>
TABLE 6 mice for test
(II) sample preparation formulation
1. Intravenous (IV) group: weighing a proper amount of the compound to be tested, and completely dissolving in a proper volume of solvent (DMSO/Solutol/H) 2 o=5/10/85V/V, added to 2meq HCl), stirred, vortexed and/or sonicated. After the solution was obtained, the solvent was gradually increased to a final volume to reach the target concentration, vortexed, sonicated to obtain a uniform solution, and filtered through a 0.22 μm PVDF filter.
2. Group (PO) orally: weighing a proper amount of the compound to be tested, and completely dissolving in a proper volume of solvent (DMSO/Solutol/H) 2 o=5/10/85V/V, added to 2meq HCl), stirred, vortexed and/or sonicated. After the solution is obtained, the solvent is gradually increased to a final volume to reach the target concentration, and the solution is obtained by vortex and ultrasonic treatment.
(III) administration and sampling of rats
Animals were randomly grouped according to animal weight, and the animals in each group had a comparable weight (no more than + -20% of the average weight) after the grouping. Meanwhile, group IV was not fasted, group PO was fasted overnight (> 12 hours) and food was given 2 hours after dosing. All animals were free to drink water. The dosing regimen and pharmacokinetic sampling regimen are given in tables 7 and 8, respectively, below.
TABLE 7 dosing regimen
TABLE 8 pharmacokinetic sampling protocol
/>
Rats were dosed as described above and blood and brain tissue samples were collected and processed at predetermined time points (collection and processing were performed as conventional in the art).
(IV) sample analysis
The brain was weighed and homogenized with 4 times of ultrapure water. The whole blood sample and brain homogenate were each added with 6 volumes of acetonitrile, vortexed for 1min, centrifuged at 4℃and 4500rpm for 15min, and the supernatant diluted 2-fold with ultrapure water, and the sample was analyzed by LC/MS.
And (V) data analysis:
pharmacokinetic parameter calculations will be performed using WinNonlin software. If applicable, the following pharmacokinetic parameters will be calculated as drug concentration-time data for plasma: CL (clearance); v (V) d (apparent distribution volume); t (T) 1/2 (elimination half-life); c (C) max (peak concentration); t (T) max (peak time); AUC (area under the blood concentration-time curve); MRT (mean residence time); f% (bioavailability).
The test results are shown in tables 9-15 below, which respectively show the rat blood concentrations of the compounds 1 and 2, 19 and 20, and the pyrroltinib and lenatinib of the present application at each time point, and the values of each pharmacokinetic parameter, and also show the concentrations of the compounds 1 and 2, 19 and 20, and the pyrroltinib and lenatinib of the present application in the brain and blood of the rats, and the ratio thereof. From the above results, it is clear that each of the compounds 1 and 2, 19 and 20 of the present application exhibits an excellent ability to penetrate the blood brain barrier, far superior to the commercially available pyrroltinib and glatiramib. This also demonstrates that the compounds of the present application have excellent EGFR and HER2 kinase inhibitory activity, and can inhibit cell proliferation at the cellular level, while having excellent ability to penetrate the blood brain barrier, and are expected to be applied to EGFR and/or HER2 kinase mediated diseases, particularly brain metastasis related diseases.
TABLE 9 rat plasma concentrations of Compounds 1 and 2 of the examples of the application
TABLE 10 rat drug parameters of the compounds of examples 1 and 2 of the application
TABLE 11 rat plasma concentrations of Compounds 19 and 20 of the examples of the application
TABLE 12 rat pharmaceutical parameters of the compounds of examples 19 and 20 of the application
/>
TABLE 13 rat plasma levels of pyrroltinib and Latinib
TABLE 14 rat pharmacokinetics of pyrroltinib and Latinib
TABLE 15 concentration and ratio of Compounds 1 and 2, 19 and 20 of the examples of the application, and of pyrroltinib and Latinib in brain and whole blood (PO 10mg/kg, sampling time, administration for 2 h)
Examples Blood concentration (ng/mL) Brain concentration (ng/g) Brain/blood ratio
Pyrotinib 884 25.8 0.0291
Neratinib 440 14.4 0.0366
1 256 373 1.46
2 616 3168 5.45
19 446 788 1.77
20 109 116 1.06
The results in tables 9 and 10 show that examples 1 and 2 have excellent pharmacokinetic parameters suitable for the development of oral inhibitors. The results in table 15 show that example 1 and example 2 have both very strong blood brain barrier penetrating properties and are suitable for the treatment of primary tumors in the brain as well as for the treatment of tumors in brain metastases. The results in tables 11-12 and 15 also show that examples 19 and 20 also have excellent pharmacokinetic properties while having very strong blood brain barrier penetrating properties. From a combination of the results in tables 3, 9, 10, 11-12 and 15, it can be seen that the compounds of the present application are expected to be developed as therapeutic agents for gliomas.
In summary, the compounds of the present application all show excellent inhibitory activity against EGFR kinase, and also show good to excellent inhibitory activity against HER2 kinase; in terms of cells, all the compounds of the application show excellent proliferation inhibition activity on Ba/F3EGFRvIII cell lines; at the same time, pharmacokinetic testing also finds that the compounds of the present application exhibit excellent ability to penetrate the blood brain barrier (far superior to marketed drugs), and thus, are expected to be therapeutic drugs for the above-mentioned diseases, especially in the case of EGFRvIII-induced tumors such as gliomas or EGFR/HER 2-driven tumor brain turnover.
Quinazoline drugs such as Gefitinib (Gefitinib), erlotinib (Erlotinib), icotinib (Icotinib), afatinib (Afatinib), and Lapatinib (Lapatinib) have been marketed in batches that are not effective in penetrating the blood brain barrier. Meanwhile, in the present compounds using quinazoline as a parent nucleus, most of the compounds are substituted at the 6 position and 7 position of the quinazoline ring, but almost none of the compounds are substituted at the 5 position, the inventor researches and discovers that an allylamide group is introduced at the 6 position of the quinazoline ring, and halogen (such as Cl) or alkyl (such as methyl) is introduced at the 5 position of the quinazoline ring for substitution, wherein the allylamide group can form irreversible covalent binding with EGFR or HER2 targets, and the halogen or alkyl introduced at the 5 position can lock the orientation of the allylamide group, so that the allylamide is more favorable for forming covalent binding with EGFR or HER2 targets. The design not only realizes the strong covalent binding capability of the compound to EGFR or HER2 targets, but also greatly improves the blood brain penetration capability of the compound.
While the foregoing is directed to the preferred embodiments of the present invention, it will be apparent to those skilled in the art that various changes and modifications can be made in the embodiments of the invention without departing from the spirit of the invention, and such changes and modifications should also be considered to be within the scope of the invention.

Claims (6)

1. A compound of formula (I), a pharmaceutically acceptable salt thereof,
in the formula (I), m is 0 and 1;
a is Cl or methyl; z is NH;
R 1 is 1-methyl-pyrrolidin-2-yl, methylamino, isopropylamino, cyclopropylamino, cyclobutylamino, methylisopropylamino, N-methyl-N-cyclopropylamino, N-methyl-N-cyclobutylamino, pyrrolidin-1-yl, piperidin-1-yl, dimethylamino and diethylamino;
R 2 、R 3 、R 5 、R 6 each independently is hydrogen, fluorine, chlorine,
R 4 is hydrogen, fluoro, phenoxy, pyridin-2-ylmethoxy, 3-fluorobenzyloxy, and R 2 、R 3 、R 4 、R 5 、R 6 At least 2 of which are hydrogen.
2. A compound according to claim 1, selected from the group consisting of:
3. a compound according to claim 1, selected from the group consisting of:
4. a pharmaceutical composition comprising a compound of any one of claims 1 to 3, a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers or excipients.
5. The pharmaceutical composition of claim 4, wherein the pharmaceutical composition further comprises one or more additional therapeutic agents.
6. Use of a compound according to any one of claims 1-3, a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer and autoimmune diseases associated with the tyrosine kinases EGFR, HER2, wherein the cancer and autoimmune diseases comprise: fundus disease, dry eye, psoriasis, vitiligo, dermatitis, alopecia areata, rheumatoid arthritis, colitis, multiple sclerosis, systemic lupus erythematosus, crohn's disease, atherosclerosis, pulmonary fibrosis, liver fibrosis, myelofibrosis, non-small cell lung cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, ovarian cancer, cervical cancer, colorectal cancer, melanoma, endometrial cancer, prostate cancer, bladder cancer, leukemia, gastric cancer, liver cancer, gastrointestinal stromal tumor, thyroid cancer, chronic myelogenous leukemia, acute myelogenous leukemia, non-Hodgkin's lymphoma, nasopharyngeal carcinoma, esophageal cancer, brain tumor, B-cell and T-cell lymphoma, multiple myeloma, biliary tract carcinoma sarcoma, cholangiocarcinoma.
CN202110395019.0A 2020-04-17 2021-04-13 Quinazoline compound, preparation method and application thereof Active CN113527215B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
TW110113803A TWI820414B (en) 2020-04-17 2021-04-16 Quinazoline compounds, preparation method and use thereof
PCT/CN2021/087774 WO2021209039A1 (en) 2020-04-17 2021-04-16 Quinazoline compound, preparation method therefor and application thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2020103048915 2020-04-17
CN202010304891 2020-04-17

Publications (2)

Publication Number Publication Date
CN113527215A CN113527215A (en) 2021-10-22
CN113527215B true CN113527215B (en) 2023-12-05

Family

ID=78124315

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110395019.0A Active CN113527215B (en) 2020-04-17 2021-04-13 Quinazoline compound, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN113527215B (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1218456A (en) * 1996-04-12 1999-06-02 沃尼尔·朗伯公司 Irreversible inhibitors of tyrosine kinases
US20050084905A1 (en) * 2002-03-21 2005-04-21 Prescott John C. Identification of kinase inhibitors
CN103998040A (en) * 2011-03-04 2014-08-20 江苏康缘药业股份有限公司 Alkyne substituted quinazoline compound and methods of use
CN106146412A (en) * 2015-03-31 2016-11-23 广州市恒诺康医药科技有限公司 Quinazoline derivant and its preparation method and application
CN110343090A (en) * 2018-04-08 2019-10-18 威尚(上海)生物医药有限公司 Quinazoline derivant salt form crystal form and preparation method and application
WO2020057511A1 (en) * 2018-09-18 2020-03-26 Suzhou Zanrong Pharma Limited Quinazoline derivatives as antitumor agents

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1218456A (en) * 1996-04-12 1999-06-02 沃尼尔·朗伯公司 Irreversible inhibitors of tyrosine kinases
US20050084905A1 (en) * 2002-03-21 2005-04-21 Prescott John C. Identification of kinase inhibitors
CN103998040A (en) * 2011-03-04 2014-08-20 江苏康缘药业股份有限公司 Alkyne substituted quinazoline compound and methods of use
CN106146412A (en) * 2015-03-31 2016-11-23 广州市恒诺康医药科技有限公司 Quinazoline derivant and its preparation method and application
CN110343090A (en) * 2018-04-08 2019-10-18 威尚(上海)生物医药有限公司 Quinazoline derivant salt form crystal form and preparation method and application
WO2020057511A1 (en) * 2018-09-18 2020-03-26 Suzhou Zanrong Pharma Limited Quinazoline derivatives as antitumor agents

Also Published As

Publication number Publication date
CN113527215A (en) 2021-10-22

Similar Documents

Publication Publication Date Title
CN111704611B (en) Aryl spiro SHP2 inhibitor compound, preparation method and application
JP6431593B2 (en) Pyridinylaminopyrimidine derivative, method for producing the same, and use thereof
CN114656482A (en) Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof
CN111484482B (en) Alkynyl pyrimidine or alkynyl pyridine compound, and composition and application thereof
JP2021501215A (en) Amino-substituted nitrogen-containing condensed ring compound, its preparation method and use
CN103421010A (en) Pteridinone derivative as EGFR inhibitor and application thereof
CN110857292A (en) EGFR kinase inhibitor and preparation method and application thereof
KR20170043546A (en) Quinazoline Derivative
CN111718332B (en) 2-substituted pyrazol amino-4-substituted amino-5-pyrimidine formamide compound, composition and application thereof
CN115322158B (en) As KRASG12CSubstituted quinazoline compounds of protein inhibitor
CN111825719A (en) Arylamine-substituted pyrrolopyrimidine compound, and preparation method and application thereof
CN113527215B (en) Quinazoline compound, preparation method and application thereof
CN113087671B (en) Cyano-substituted pyridine and cyano-substituted pyrimidine compounds, and preparation methods and applications thereof
CN115894381A (en) 2,4,5-trisubstituted pyrimidine compound and preparation method and application thereof
CN115894486B (en) Hydrogenated pyridoquinazoline compound, composition and application thereof
CN115894455B (en) Quinazoline compound, composition and application thereof
TWI820414B (en) Quinazoline compounds, preparation method and use thereof
CN115109036B (en) Quinazoline compound, composition and application thereof
CN115701429B (en) 4- (1H-indol-1-yl) pyrimidine-2-amino derivative, and preparation method and application thereof
CN112442045B (en) Acyl-substituted oxazine quinazoline compound, preparation method and application thereof
CN115894383A (en) Quinazoline compound, composition and application thereof
CN115803325B (en) EGFR inhibitor and preparation method and application thereof
CN112384506A (en) Indoline-1-formamide compound, preparation method and application thereof in medicine and pharmacology
CN112341378A (en) Urea substituted alkynyl pyrimidine or urea substituted alkynyl pyridine compound, and composition and application thereof
WO2013062071A1 (en) Piperazine derivative and salt thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant