CN114539226A - Crystal form containing indole derivative free base and preparation method and application thereof - Google Patents

Crystal form containing indole derivative free base and preparation method and application thereof Download PDF

Info

Publication number
CN114539226A
CN114539226A CN202111346076.6A CN202111346076A CN114539226A CN 114539226 A CN114539226 A CN 114539226A CN 202111346076 A CN202111346076 A CN 202111346076A CN 114539226 A CN114539226 A CN 114539226A
Authority
CN
China
Prior art keywords
diffraction peak
amino
diffraction
ray powder
alkyl
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.)
Pending
Application number
CN202111346076.6A
Other languages
Chinese (zh)
Inventor
李媛媛
呙临松
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.)
Jiangsu Hansoh Pharmaceutical Group Co Ltd
Shanghai Hansoh Biomedical Co Ltd
Original Assignee
Jiangsu Hansoh Pharmaceutical Group Co Ltd
Shanghai Hansoh Biomedical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jiangsu Hansoh Pharmaceutical Group Co Ltd, Shanghai Hansoh Biomedical Co Ltd filed Critical Jiangsu Hansoh Pharmaceutical Group Co Ltd
Publication of CN114539226A publication Critical patent/CN114539226A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • C07D403/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • 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
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Abstract

The invention relates to a crystal form of indole derivative free base, a preparation method and application thereof. The invention relates to a compound shown in a general formula (I), a crystal form thereof, a preparation method, a pharmaceutical composition containing a therapeutically effective amount of the compound or the crystal form thereof, and application of the compound as a kinase inhibitor, in particular as a receptor Tyrosine Kinase Inhibitor (TKI), more particularly as an EGFR or HER2 inhibitor in treating related diseases such as cancer, inflammation, chronic liver disease, diabetes, cardiovascular disease and AIDS, wherein the compound shows good inhibitory activity in EGFR and HER 220 exon mutation.
Figure DDA0003354163650000011

Description

Crystal form containing indole derivative free base and preparation method and application thereof
Technical Field
The invention belongs to the field of drug synthesis, and particularly relates to a crystal form of indole derivative free base, and a preparation method and application thereof.
Background
The existence of multiple signaling pathways in a cell that interact to control cell proliferation, growth, migration, and apoptosis. Abnormal activation of the signaling pathway can lead to tumorigenesis. Receptor tyrosine kinases play an important role in the regulation of cells. The Epidermal Growth Factor Receptor (EGFR) is a member of the ErbB receptor family (including ErbB1, ErbB2, ErbB3, ErbB4) which are transmembrane protein tyrosine kinases, and can form homodimers on membranes by binding to their ligand, Epidermal Growth Factor (EGF), or heterodimers with other receptors in the ErbB family (such as ErbB2, ErbB3, ErbB4), resulting in activation of EGFR tyrosine kinase activity. Activated EGFR can phosphorylate different substrates, so that the downstream PI3K-AKT pathway and RAS-MAPK pathway are activated, and the activated EGFR plays a role in multiple processes such as cell survival, proliferation and apoptosis.
Dysregulation of the EGFR signaling pathway, including increased expression of ligands and receptors, amplification and mutation of the EGFR gene, and the like, can promote cellular transformation to malignancy, leading to the development of a variety of tumors. About 35% of non-small cell lung cancer (NSCLC) patients in china are EGFR mutations, with the most common types of mutations being a 19 exon deletion mutation (Del19) and a 21 exon L858R activating mutation, both of which account for about 80% of EGFR mutations. The EGFR20 exon insertion mutation is another large mutation of EGFR mutation, which accounts for 4% -10% of EGFR mutation in NSCLC, the mutation types are dozens of types, and the common mutation types are Ex20Ins D770_ N771InsSVD, Ex20Ins V769_ D770InsASV and the like.
Over the years, a large number of targeted drugs have been developed for EGFR mutation in NSCLC, such as a generation of reversible tyrosinase inhibitors (TKI) gefitinib and erlotinib against classical Del19 mutation and L858R mutation, a generation of irreversible covalent binding inhibitors afatinib, and a generation of inhibitor Axictinib against drug-resistant mutation EGFR T790M, which all have very good clinical effects. However, the EGFR inhibitor on the market has poor effect on insertion mutation of EGFR20 exon, the life cycle of patients is short, the target point needs a more specific inhibitor, and a larger clinical demand exists.
HER2 is another member of the ErbB family, whose amplification and mutation occur in a variety of cancers. Among them, HER2 mutations account for about 4% in NSCLC, whereas HER2 mutations account for about 90% for 20 exon insertion mutations, the most common type of mutation being p.a775_ G776insYVMA, to which EGFR inhibitors currently on the market generally exert their effects.
At present, a plurality of domestic and foreign drug enterprises have actively researched for EGFR & HER 220 exon insertion mutation, wherein Poziotiib of Spectrum company, TAK-788 of Takeda and Tarloxotiib of Rain Therapeutics enter clinical research, and a compound TAS-6417 of Cullinan & Taiho company shows better activity in preclinical experiments. Because many EGFR inhibitors have strong inhibition effect on EGFR wild type, side effects such as rash and the like appear clinically, and the inhibition activities on EGFR20 exon insertion mutation and HER 220 exon insertion mutation targets are also to be improved, the compounds with obvious mutation effects on EGFR and HER 220 exons and high selectivity on wild type EGFR still have great demand and have good market prospects.
The patent PCT/CN2020/091558 discloses a series of structures of indole derivative-containing inhibitors, and in subsequent development, in order to make the product easy to handle, filter and dry, and to find suitable crystals which are convenient to store, stable for a long time and high in bioavailability, the invention has comprehensively studied the free base crystal form of the above compounds.
Disclosure of Invention
All matters hithertofore set forth in PCT/CN2020/091558 are hereby incorporated by reference.
The invention aims to provide a crystal form of a compound shown as a general formula (I),
Figure BDA0003354163630000021
wherein:
R1selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -C (O) NRaa(CH2)nRbbOr- (CH)2)nN=S(O)RaaRbb(ii) a The amino, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, and heteroaryl, optionally further substituted with one or more substituents of deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
R2selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, or heteroaryl;
R3selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, or heteroaryl;
Raaand RbbEach independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl; the amino, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, optionally further substituted with one or more substituents of deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
x is an integer of 0-4; and is
n is an integer of 0 to 4.
In a preferred embodiment of the invention, the crystal form shown in the general formula (I) is shown in the specification, wherein R is1Selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-8Cycloalkyl, 3-8 membered heterocyclyl, C6-10Aryl, 5-10 membered heteroaryl, -C (O) NRaa(CH2)nRbbOr- (CH)2)nN=S(O)RaaRbb(ii) a The amino group, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-8Cycloalkyl, 3-8 membered heterocyclyl, C6-10Aryl and 5-10 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitroHydroxy, cyano, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl, 3-8 membered heterocyclyl, C6-10Aryl and 5-10 membered heteroaryl;
preferably, R1Selected from hydrogen, deuterium, halogen, cyano, C1-3Alkyl radical, C1-3Deuterated alkyl, C1-3Haloalkyl, C1-3Alkoxy radical, C1-3Haloalkoxy, 5-6 membered heteroaryl, -C (O) NRaa(CH2)nRbbOr- (CH)2)nN=S(O)RaaRbbSaid C is1-3Alkyl radical, C1-3Deuterated alkyl, C1-3Haloalkyl, C1-3Alkoxy radical, C1-3Haloalkoxy and 5-6 membered heteroaryl, optionally further substituted with deuterium, halogen, C1-3Alkyl radical, C1-3Deuterated alkyl, C1-3Haloalkyl, C1-3Alkoxy and C1-6One or more substituents of haloalkoxy;
more preferably, R1Selected from hydrogen, deuterium, cyano, 5-6 membered heteroaryl, -C (O) NRaa(CH2)nRbbOr- (CH)2)nN=S(O)RaaRbbSaid 5-6 membered heteroaryl is optionally further substituted by deuterium, halogen, C1-3Alkyl and C1-3Substituted with one or more substituents in deuterated alkyl;
most preferably, R1Selected from hydrogen, cyano, oxazolyl, pyrazolyl,
Figure BDA0003354163630000031
Figure BDA0003354163630000032
RaaAnd RbbEach independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-8Cycloalkyl, 3-8 membered heterocyclyl, C6-10Aryl or 5-10 membered heteroaryl; the amino group, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-8Cycloalkyl, 3-8 membered heterocyclyl, C6-10Aryl and 5-10 membered heteroaryl, optionally further substituted by deuterium, halogen, amino, nitro, hydroxy, cyano, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-8Cycloalkyl, 3-8 membered heterocyclyl, C6-10Aryl and 5-10 membered heteroaryl;
x is an integer of 0-2; and is
n is an integer of 0 to 2.
In a preferred embodiment of the invention, the crystal form shown in the general formula (I) is shown in the specification, wherein R is2Selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-8Cycloalkyl or 3-8 membered heterocyclyl;
preferably hydrogen, deuterium, halogen, cyano, C1-3Alkyl radical, C1-3Deuterated alkyl, C1-3Haloalkyl, C1-3Alkoxy radical, C1-3Haloalkoxy, C3-6Cycloalkyl or 3-6 membered heterocyclyl;
more preferably hydrogen, deuterium, methyl, ethyl, propyl, cyclopropyl or oxetanyl;
most preferred are hydrogen, methyl, cyclopropyl or oxetanyl.
In a preferred embodiment of the invention, the crystal form shown in the general formula (I), R3Selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy or C1-6A haloalkoxy group;
preferably hydrogen, deuterium, halogen, cyano, C1-3Alkyl radical, C1-3Deuterated alkyl, C1-3Haloalkyl, C1-3Alkoxy or C1-3A haloalkoxy group;
more preferably hydrogen, deuterium, fluorine, chlorine, bromine, methoxy or ethoxy;
most preferred is hydrogen, fluorine or methoxy.
In a more preferred embodiment of the invention, the compound is N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, N- (5- ((5-cyano-4- (6-methoxy-1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, or mixtures thereof, N- (5- ((5-cyano-4- (1-cyclopropyl-5-fluoro-6-methoxy-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, N- (2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) -5- (oxazol-2-yl) pyrimidin-2-yl) amino) phenyl) acrylamide.
In a more preferred embodiment of the invention, a crystalline form of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide is provided.
In a further preferred embodiment of the present invention, there is provided crystalline form a of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (example 1).
Form a has an X-ray powder diffraction pattern comprising at least one or more diffraction peaks, preferably 2, more preferably 3, at 2 Θ, located in 7.1 ± 0.2 °, 14.1 ± 0.2 °, 18.9 ± 0.2 ° 2 Θ; optionally, the composition can further comprise at least one position with 2 theta of 22.7 +/-0.2 degrees, 20.4 +/-0.2 degrees, 22.3 +/-0.2 degrees, 13.4 +/-0.2 degrees and 12.9 +/-0.2 degrees, preferably comprises 2 positions, 3 positions, 4 positions or 5 positions;
the X-ray powder diffraction pattern of form a optionally further comprises one or more diffraction peaks at 17.5 ± 0.2 °, 20.8 ± 0.2 °, 9.6 ± 0.2 °, 22.0 ± 0.2 °, 25.6 ± 0.2 °, 14.9 ± 0.2 °, 15.6 ± 0.2 ° 2 Θ; preferably at least any 2-3, or 4-5, or 6-7 thereof; further preferably, any 2, 3, 4, 5, 6, 7 thereof;
the X-ray powder diffraction pattern of the crystal form A comprises one or more diffraction peaks which are positioned in the diffraction peaks with the 2 theta of 7.1 +/-0.2 degrees, 14.1 +/-0.2 degrees, 18.9 +/-0.2 degrees, 22.7 +/-0.2 degrees, 20.4 +/-0.2 degrees, 22.3 +/-0.2 degrees, 13.4 +/-0.2 degrees, 12.9 +/-0.2 degrees, 17.5 +/-0.2 degrees, 20.8 +/-0.2 degrees, 9.6 +/-0.2 degrees, 22.0 +/-0.2 degrees, 25.6 +/-0.2 degrees, 14.9 +/-0.2 degrees, 15.6 +/-0.2 degrees, 28.1 +/-0.2 degrees, 9.4 +/-0.2 degrees, 24.1 +/-0.2 degrees, 16.2 +/-0.2 degrees and 19.3 +/-0.2 degrees; preferably, the compound comprises diffraction peaks at 4, 5, 6, 8 or 10 optionally;
most preferably, using Cu-K.alpha.radiation, the characteristic diffraction peaks of X-rays expressed in terms of 2 theta angle and interplanar spacing d are shown in Table 1.
TABLE 1
Figure BDA0003354163630000051
Further preferably, the compound shown in example 1, N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, in crystalline form a, has an X-ray powder diffraction pattern substantially as shown in figure 1; the DSC pattern is basically shown in figure 2; the TGA profile is substantially as shown in figure 3.
In a further preferred embodiment of the present invention, there is provided crystalline form B of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (example 1).
The X-ray powder diffraction pattern of the crystal form B has a diffraction peak at the 2 theta of 6.5 +/-0.2 degrees; or a diffraction peak at 20.7 ± 0.2 °; or a diffraction peak at 20.9 ± 0.2 °; or a diffraction peak at 21.0 ± 0.2 °; or a diffraction peak at 21.4 ± 0.2 °; or a diffraction peak at 15.6 ± 0.2 °; or a diffraction peak at 23.8 ± 0.2 °; or a diffraction peak at 21.6 ± 0.2 °; or a diffraction peak at 9.2 ± 0.2 °; or a diffraction peak at 25.1 ± 0.2 °; preferably comprises any 2-5, or 3-6, or 3-8, or 5-8, or 6-8 of the diffraction peaks, more preferably comprises any 6, 7 or 8 thereof;
form B has an X-ray powder diffraction pattern comprising at least one, preferably two, more preferably three, diffraction peaks, at 2 Θ of 6.5 ± 0.2 °, 20.7 ± 0.2 °, 20.9 ± 0.2 °; optionally, at least one of 21.0 ± 0.2 °, 21.4 ± 0.2 °, 15.6 ± 0.2 °, 23.8 ± 0.2 °, and 21.6 ± 0.2 ° may be further included; preferably 2, 3, 4 or 5 thereof;
the X-ray powder diffraction pattern of form B optionally further comprises one or more diffraction peaks at 9.2 ± 0.2 °, 25.1 ± 0.2 °, 10.7 ± 0.2 °, 12.4 ± 0.2 °, 17.4 ± 0.2 °, 17.6 ± 0.2 °, 14.5 ± 0.2 ° 2 Θ; preferably at least any 2-3, or 4-5, or 6-7 thereof; further preferably, any 2, 3, 4, 5, 6, 7 thereof;
the X-ray powder diffraction pattern of the crystal form B comprises diffraction peaks at one or more positions of which the 2 theta is 6.5 +/-0.2 degrees, 20.7 +/-0.2 degrees, 20.9 +/-0.2 degrees, 21.0 +/-0.2 degrees, 21.4 +/-0.2 degrees, 15.6 +/-0.2 degrees, 23.8 +/-0.2 degrees, 21.6 +/-0.2 degrees, 9.2 +/-0.2 degrees, 25.1 +/-0.2 degrees, 10.7 +/-0.2 degrees, 12.4 +/-0.2 degrees, 17.4 +/-0.2 degrees, 17.6 +/-0.2 degrees, 14.5 +/-0.2 degrees, 23.4 +/-0.2 degrees, 20.5 +/-0.2 degrees, 13.5 +/-0.2 degrees, 16.9 +/-0.2 degrees and 24.7 +/-0.2 degrees; preferably, the compound comprises diffraction peaks at 4, 5, 6, 8 and 10;
most preferably, using Cu-K.alpha.radiation, the characteristic diffraction peaks for X-rays, expressed in terms of 2 θ angle and interplanar spacing d, are shown in Table 2.
TABLE 2
Figure BDA0003354163630000061
Figure BDA0003354163630000071
Further preferably, the compound shown in example 1, N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, in crystalline form B, has an X-ray powder diffraction pattern substantially as shown in figure 4; the DSC pattern is basically shown in figure 5; the TGA profile is substantially as shown in figure 6.
In a further preferred embodiment of the present invention, crystalline form C of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (example 1) is provided.
The X-ray powder diffraction pattern of the crystal form C has a diffraction peak at the 2 theta of 7.1 +/-0.2 degrees; or a diffraction peak at 18.1 ± 0.2 °; or a diffraction peak at 9.0 ± 0.2 °; or a diffraction peak at 22.7 ± 0.2 °; or a diffraction peak at 14.1 ± 0.2 °; or a diffraction peak at 18.9 ± 0.2 °; or a diffraction peak at 22.5 ± 0.2 °; or a diffraction peak at 20.3 ± 0.2 °; or a diffraction peak at 22.2 ± 0.2 °; or a diffraction peak at 23.2 ± 0.2 °; preferably comprises any 2-5, or 3-6, or 3-8, or 5-8, or 6-8 of the diffraction peaks, more preferably comprises any 6, 7 or 8 thereof;
form C has an X-ray powder diffraction pattern comprising at least one, preferably two, more preferably three, diffraction peaks, at 2 Θ of 7.1 ± 0.2 °, 18.1 ± 0.2 °, 9.0 ± 0.2 °; optionally, at least one of 22.7 ± 0.2 °, 14.1 ± 0.2 °, 18.9 ± 0.2 °, 22.5 ± 0.2 °, and 20.3 ± 0.2 ° may be further included; preferably 2, 3, 4 or 5 thereof;
the X-ray powder diffraction pattern of form C optionally further comprises one or more diffraction peaks at 22.2 ± 0.2 °, 23.2 ± 0.2 °, 23.1 ± 0.2 °, 20.7 ± 0.2 °, 22.0 ± 0.2 °, 17.5 ± 0.2 °, 12.9 ± 0.2 ° 2 Θ; preferably at least any 2-3, or 4-5, or 6-7 thereof; further preferably, any 2, 3, 4, 5, 6, 7 thereof;
the X-ray powder diffraction pattern of the crystal form C comprises one or more diffraction peaks positioned at one or more of 7.1 +/-0.2 degrees, 18.1 +/-0.2 degrees, 9.0 +/-0.2 degrees, 22.7 +/-0.2 degrees, 14.1 +/-0.2 degrees, 18.9 +/-0.2 degrees, 22.5 +/-0.2 degrees, 20.3 +/-0.2 degrees, 22.2 +/-0.2 degrees, 23.2 +/-0.2 degrees, 23.1 +/-0.2 degrees, 20.7 +/-0.2 degrees, 22.0 +/-0.2 degrees, 17.5 +/-0.2 degrees, 12.9 +/-0.2 degrees, 21.9 +/-0.2 degrees, 13.4 +/-0.2 degrees and 10.7 +/-0.2 degrees of 2 degrees; preferably, the compound comprises diffraction peaks at 4, 5, 6, 8 or 10 optionally;
most preferably, using Cu-K.alpha.radiation, the characteristic diffraction peaks of X-rays expressed in terms of 2 theta angle and interplanar spacing d are shown in Table 3.
TABLE 3
Figure BDA0003354163630000081
Further preferably, the compound shown in example 1, N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, has the crystalline form C and the X-ray powder diffraction pattern substantially as shown in fig. 7; the DSC pattern is basically shown in figure 8; the TGA profile is substantially as shown in figure 9.
In a further preferred embodiment of the present invention, there is provided crystalline form D of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (example 1).
The X-ray powder diffraction pattern of the crystal form D has a diffraction peak at the 2 theta of 26.2 +/-0.2 degrees; or a diffraction peak at 22.2 ± 0.2 °; or a diffraction peak at 10.6 ± 0.2 °; or a diffraction peak at 14.5 ± 0.2 °; or a diffraction peak at 21.3 ± 0.2 °; or a diffraction peak at 15.9 ± 0.2 °; or a diffraction peak at 7.0 ± 0.2 °; or a diffraction peak at 9.0 ± 0.2 °; or a diffraction peak at 19.0 ± 0.2 °; or a diffraction peak at 8.3 ± 0.2 °; preferably comprises any 2-5, or 3-6, or 3-8, or 5-8, or 6-8 of the diffraction peaks, more preferably comprises any 6, 7 or 8 thereof;
form D has an X-ray powder diffraction pattern comprising at least one, preferably two, more preferably three, diffraction peaks, at 26.2 ± 0.2 °, 22.2 ± 0.2 °, 10.6 ± 0.2 ° 2 Θ; optionally, at least one of 14.5 ± 0.2 °, 21.3 ± 0.2 °, 15.9 ± 0.2 °, 7.0 ± 0.2 °, and 9.0 ± 0.2 ° may be further included; preferably 2, 3, 4 or 5 thereof;
the X-ray powder diffraction pattern of form D optionally further comprises one or more diffraction peaks at 19.0 ± 0.2 °, 8.3 ± 0.2 °, 26.9 ± 0.2 °, 18.1 ± 0.2 °, 20.1 ± 0.2 °, 21.8 ± 0.2 °, 19.8 ± 0.2 ° 2 Θ; preferably at least any 2-3, or 4-5, or 6-7 thereof; further preferably, any 2, 3, 4, 5, 6, 7 thereof;
the X-ray powder diffraction pattern of the crystal form D comprises one or more diffraction peaks positioned in 26.2 +/-0.2 degrees, 22.2 +/-0.2 degrees, 10.6 +/-0.2 degrees, 14.5 +/-0.2 degrees, 21.3 +/-0.2 degrees, 15.9 +/-0.2 degrees, 7.0 +/-0.2 degrees, 9.0 +/-0.2 degrees, 19.0 +/-0.2 degrees, 8.3 +/-0.2 degrees, 26.9 +/-0.2 degrees, 18.1 +/-0.2 degrees, 20.1 +/-0.2 degrees, 21.8 +/-0.2 degrees, 19.8 +/-0.2 degrees and 18.3 +/-0.2 degrees of 2 theta; preferably, the compound comprises diffraction peaks at 4, 5, 6, 8 or 10 optionally;
most preferably, using Cu-K.alpha.radiation, the characteristic diffraction peaks of X-rays expressed in terms of 2 theta angle and interplanar spacing d are shown in Table 4.
TABLE 4
Figure BDA0003354163630000091
Further preferably, the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide in crystalline form D has an X-ray powder diffraction pattern substantially as shown in figure 10; the DSC pattern is basically shown in figure 11; the TGA profile is substantially as shown in figure 12.
In a further preferred embodiment of the present invention, crystalline form E of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (example 1) is provided.
The X-ray powder diffraction pattern of the crystal form E has a diffraction peak at the 2 theta of 6.3 +/-0.2 degrees; or a diffraction peak at 12.6 ± 0.2 °; or a diffraction peak at 9.9 ± 0.2 °; or a diffraction peak at 21.1 ± 0.2 °; or a diffraction peak at 13.2 ± 0.2 °; or a diffraction peak at 26.2 ± 0.2 °; or a diffraction peak at 20.0 ± 0.2 °; or a diffraction peak at 16.9 ± 0.2 °; or a diffraction peak at 15.5 ± 0.2 °; or a diffraction peak at 25.3 ± 0.2 °; preferably comprises any of 2 to 5, or 3 to 6, or 3 to 8, or 5 to 8, or 6 to 8 of the diffraction peaks; more preferably any 6, 7 or 8 thereof;
form E has an X-ray powder diffraction pattern comprising at least one, preferably two, more preferably three, diffraction peaks, at 2 Θ of 6.3 ± 0.2 °, 12.6 ± 0.2 °, 9.9 ± 0.2 °; optionally, at least one of 21.1 ± 0.2 °, 13.2 ± 0.2 °, 26.2 ± 0.2 °, 20.0 ± 0.2 °, and 16.9 ± 0.2 ° may be further included; preferably 2, 3, 4 or 5 thereof;
the X-ray powder diffraction pattern of form E optionally further comprises one or more diffraction peaks at 15.5 ± 0.2 °, 25.3 ± 0.2 °, 27.4 ± 0.2 ° 2 Θ; preferably at least any 2 to 3 thereof; further preferably, any 2 or 3 thereof;
the X-ray powder diffraction pattern of the crystal form E comprises one or more diffraction peaks positioned in 6.3 +/-0.2 degrees, 12.6 +/-0.2 degrees, 9.9 +/-0.2 degrees, 21.1 +/-0.2 degrees, 13.2 +/-0.2 degrees, 26.2 +/-0.2 degrees, 20.0 +/-0.2 degrees, 16.9 +/-0.2 degrees, 15.5 +/-0.2 degrees, 25.3 +/-0.2 degrees and 27.4 +/-0.2 degrees of 2 theta; preferably, the compound comprises diffraction peaks at 4, 5, 6, 8 or 10 optionally;
most preferably, using Cu-K.alpha.radiation, the characteristic diffraction peaks of X-rays expressed in terms of 2 theta angle and interplanar spacing d are shown in Table 5.
TABLE 5
Figure BDA0003354163630000101
Further preferably, the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide is in crystalline form E having an X-ray powder diffraction pattern substantially as shown in figure 13; the DSC pattern is basically as shown in figure 14; the TGA profile is substantially as shown in figure 15.
In a further preferred embodiment of the present invention, the diffraction peak positions of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, which have the first ten strong relative peak intensities in the X-ray powder diffraction pattern of form a, form B, form C, form D, form E, have a 2 θ error of ± 0.2 ° to ± 0.5 °, preferably ± 0.2 ° to ± 0.3 °, most preferably ± 0.2 °, from the diffraction peak positions corresponding to fig. 1,4, 7, 10 and 13, respectively.
In a preferred embodiment of the invention, the compound of formula (I) is in a crystalline form, with or without a solvent, wherein the solvent is selected from one or more of water, methanol, acetone, ethyl acetate, acetonitrile, ethanol, 88% acetone, tetrahydrofuran, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, benzene, toluene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, N-methylpyrrolidone, dimethyl sulfoxide, N-propanol, tert-butanol, 2-butanone, 3-pentanone, N-heptane, ethyl formate, isopropyl acetate, cyclohexane, methyl tert-butyl ether or isopropyl ether.
In a preferred embodiment of the invention, the number of solvents in the crystalline form of any one of the compounds of formula (I) is 0.2 to 3, preferably 0.2, 0.5, 1, 1.5, 2, 2.5 or 3, more preferably 0.5, 1,2 or 3.
In a preferred embodiment of the present invention, the crystalline form of the compound of formula (I) is a non-solvent crystalline form, preferably an anhydrous crystalline form.
In a preferred embodiment of the invention, the crystalline form of the compound of formula (I) is a hydrate crystalline form, the number of water is 0.2 to 3, preferably 0.2, 0.5, 1, 1.5, 2, 2.5 or 3, more preferably 0.5, 1,2 or 3.
In a more preferred embodiment of the invention, the crystalline form of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide is a non-solvated compound, preferably an anhydrate or a hydrate.
In a more preferred embodiment of the invention, the number of water in the crystalline form of the hydrate of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide is between 0.2 and 3, preferably 0.2, 0.5, 1, 1.5, 2, 2.5 or 3, more preferably 0.5, 1,2 or 3.
It is well known to those skilled in the art that XRPD can produce certain displacements and intensity deviations due to the detection method, conditions and instrumentation. As a specific example of the crystal form of the present invention, XRPD is shown as pattern X, but it is understood by those of ordinary skill that when the deviation of the critical characteristic peak shift 2 theta is about + -0.5, especially + -0.2, all can be identified as the same crystal form.
In another aspect, the present invention also relates to a process for preparing a crystalline form of the compound of formula (I), comprising the steps of:
1) weighing a proper amount of free alkali, and suspending with a poor solvent, wherein the suspension density is preferably 50-200 mg/mL;
2) shaking the obtained suspension at a certain temperature for a certain time, preferably at the temperature of 0-50 ℃ for 1-15 days;
3) quickly centrifuging the suspension, removing supernatant, drying the residual solid to constant weight to obtain a target product, and for example, drying the target product in a vacuum drying oven at the temperature of 45-55 ℃ to constant weight to obtain the target product;
wherein:
the poor solvent is selected from one or more of methanol, acetone, ethyl acetate, tetrahydrofuran, acetonitrile, ethanol, 88% acetone, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, methyl tert-butyl ether, N-heptane, benzene, toluene, chlorobenzene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, dimethyl sulfoxide, N-propanol, ethyl formate, isopropyl acetate, tert-butanol, 2-butanone and 3-pentanone; one or more of toluene, 1, 4-dioxane, 3-pentanone, ethyl acetate, methanol and ethyl formate are preferred.
The invention also relates to a method for preparing the crystal form of the compound shown in the general formula (I), which specifically comprises the following steps:
1) weighing a proper amount of free alkali, and dissolving the free alkali in a good solvent;
2) adding an anti-solvent into the obtained solution, and stirring until a solid is separated out; preferably, adding an anti-solvent into the obtained solution at the temperature of 0-50 ℃;
3) quickly centrifuging the suspension, removing supernatant, drying the residual solid to constant weight to obtain a target product, and for example, drying the target product in a vacuum drying oven at the temperature of 45-55 ℃ to constant weight;
wherein:
the benign solvent is selected from one or more of methanol, acetone, ethyl acetate, tetrahydrofuran, acetonitrile, ethanol, 88% acetone, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, benzene, toluene, chlorobenzene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, dimethyl sulfoxide, N-propanol, ethyl formate, isopropyl acetate, tert-butanol, 2-butanone and 3-pentanone; preferably one or more of dichloromethane, tetrahydrofuran, 2-methyl-tetrahydrofuran, 1, 4-dioxane, dimethyl sulfoxide, acetonitrile and 2-butanone.
The poor solvent is selected from one or more of methanol, ethanol, ethyl acetate, acetone, isopropanol, toluene, n-heptane, water, isopropyl acetate, cyclohexane, methyl tert-butyl ether and isopropyl ether; one or more of water, n-heptane, cyclohexane and methyl tert-butyl ether are preferred.
The invention also aims to provide a pharmaceutical composition which contains a therapeutically effective amount of the crystal form of the compound shown in the general formula (I) and one or more pharmaceutically acceptable carriers or excipients.
The invention also aims to provide the crystal form of the compound shown in the general formula (I) and application of the pharmaceutical composition in preparation of kinase inhibitor drugs.
The invention also aims to provide a crystal form of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide shown in the general formula (I) and application of a pharmaceutical composition in preparation of a kinase inhibitor medicament.
The kinase inhibitor is a receptor tyrosine kinase inhibitor, preferably a HER2 inhibitor, an EGFR monoclonal antibody and a combination related medicament thereof, more preferably a HER 220 exon mutant inhibitor, an EGFR20 exon mutant inhibitor, an EGFR20 exon mutant monoclonal antibody and a combination related medicament thereof.
The invention aims to provide the crystal form of the compound shown in the general formula (I) and the application of the pharmaceutical composition in medicaments for treating cancers, inflammations, chronic liver diseases, diabetes, cardiovascular diseases and AIDS-related diseases, preferably, the cancers, the inflammations, the chronic liver diseases, the diabetes, the cardiovascular diseases and the AIDS-related diseases are diseases mediated by HER 220 exon mutation and/or EGFR20 exon mutation.
The invention also aims to provide a crystal form of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide shown as the general formula (I) and application of the pharmaceutical composition in medicines for treating cancer, inflammation, chronic liver disease, diabetes, cardiovascular disease and AIDS-related diseases, preferably, the cancer, inflammation, chronic liver disease, diabetes, cardiovascular disease and AIDS-related diseases are diseases mediated by HER 220 exon mutation and/or EGFR20 exon mutation.
The cancer is selected from breast cancer, cervical cancer, colon cancer, lung cancer, gastric cancer, rectal cancer, pancreatic cancer, brain cancer, liver cancer, solid tumor, glioma, glioblastoma, leukemia, lymphoma, myeloma and non-small cell lung cancer.
The third-generation EGFR inhibitor mainly aims at the EGFR activating mutant and the T790M drug-resistant mutant, and compared with the third-generation EGFR inhibitor, the compound of the invention has the following remarkable advantages in the aspect of EGFR and/or HER 220 exon insertion mutation targets:
1. the inhibitory activity of the Ba/F3 EGFR mutant cell strain is obviously improved, and the activity of the optimized compound is higher than 10 times, even 20 times;
2. the selectivity of the proliferation inhibition activity of the EGFR mutant cell line and the A431 cell line in Ba/F3 is improved, and the compound is preferably more than 3 times or even 10 times higher;
3. also shows significant advantages in the in vivo pharmacodynamic tumor inhibition rate on a mouse primary B cell Ba/F3 EGFR-D770-N771ins _ SVD transplantation tumor model.
Detailed description of the invention
Unless stated to the contrary, terms used in the specification and claims have the following meanings.
The term "alkyl" refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing 1 to 20 carbon atoms, preferably an alkyl group containing 1 to 8 carbon atoms, more preferably an alkyl group of 1 to 6 carbon atoms, and most preferably an alkyl group of 1 to 3 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, various branched chain isomers thereof, and the like.
The term "cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably from 3 to 8 carbon atoms, and most preferably from 3 to 6 carbon atoms. Non-limiting examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl.
The term "heterocyclyl" refers to a saturated or partially unsaturated mono-or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms wherein one or more of the ring atoms is selected from nitrogen, oxygen, or S (O)m(wherein m is an integer from 0 to 2) but excludes the ring moiety of-O-O-, -O-S-, or-S-S-, the remaining ring atoms being carbon. Preferably 3 to 12 ring atoms, of which 1 to 4 are heteroatoms; more preferably from 3 to 8 ring atoms; most preferably from 3 to 6 ring atoms. Non-limiting examples of heterocyclyl groups include oxetanyl, thietanyl, pyrrolidinyl, pyrrolidinonyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl.
The term "aryl" refers to a 6 to 14 membered all carbon monocyclic or fused polycyclic (i.e., rings which share adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 10 membered, such as phenyl and naphthyl. More preferably phenyl.
The term "heteroaryl" refers to a heteroaromatic system comprising 1 to 4 heteroatoms, 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen. Heteroaryl is preferably 5 to 10 membered, more preferably 5 or 6 membered, such as imidazolyl, furyl, thienyl, thiazolyl, pyrazolyl, oxazolyl, pyrrolyl, triazolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl and the like, preferably triazolyl, thienyl, imidazolyl, pyrazolyl, oxazolyl, pyrimidinyl or thiazolyl; more preferably pyrazolyl and oxazolyl.
The term "alkoxy" refers to-O- (alkyl) and-O- (unsubstituted cycloalkyl), wherein alkyl is as defined above. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
"haloalkyl" refers to an alkyl group substituted with one or more halogens, wherein alkyl is as defined above.
"haloalkoxy" refers to an alkoxy group substituted with one or more halogens, wherein the alkoxy group is as defined above.
"hydroxy" refers to an-OH group.
"halogen" means fluorine, chlorine, bromine or iodine.
"amino" means-NH2
"cyano" means-CN.
"nitro" means-NO2
"carboxy" refers to-C (O) OH.
"THF" refers to tetrahydrofuran.
"MeOH" refers to methanol.
“Pd2(dba)3"refers to tris (dibenzylideneacetone) dipalladium.
Different terms such as "X is selected from A, B or C", "X is selected from A, B and C", "X is A, B or C", "X is A, B and C" and the like all express the same meaning, that is, X can be any one or more of A, B, C.
All hydrogen atoms described in the present invention can be replaced by deuterium, which is an isotope thereof, and any hydrogen atom in the compound of the embodiment related to the present invention can also be replaced by a deuterium atom.
"optional" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not. For example, "a heterocyclic group optionally substituted with an alkyl" means that an alkyl may, but need not, be present, and the description includes the case where the heterocyclic group is substituted with an alkyl and the heterocyclic group is not substituted with an alkyl.
"substituted" means that one or more, preferably up to 5, more preferably 1 to 3, hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (experimentally or theoretically) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable in combination with carbon atoms having unsaturated (e.g., olefinic) bonds. Optional substituents include deuterium, halogen, amino, hydroxy, cyano, oxo, thioxo, alkyl, alkenyl, alkynyl, deuterated alkyl, haloalkylHydroxyalkyl, alkoxy, alkylthio, haloalkoxy, cycloalkyl, heterocyclyl, aryl and heteroaryl, preferably deuterium, halogen, amino, hydroxy, cyano, oxo, thio, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Hydroxyalkyl radical, C1-6Alkoxy radical, C1-6Alkylthio radical, C1-6Haloalkoxy, C3-12Cycloalkyl, 3-12 membered heterocyclyl, C6-14Aryl and 5-14 membered heteroaryl.
"pharmaceutical composition" means a mixture containing one or more compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof in admixture with other chemical components, as well as other components such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of the active ingredient and exert biological activity.
"pharmaceutically acceptable salts" refers to salts of the compounds of the present invention which are safe and effective for use in the body of a mammal and which possess the requisite biological activity.
Drawings
Figure 1 is an XRPD representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form a.
Figure 2 is a DSC representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form a.
Figure 3 is a TGA representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form a.
Figure 4 is a graphical representation of the XRPD of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form B.
Figure 5 is a DSC representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form B.
Figure 6 is a TGA representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form B.
Figure 7 is a graphical representation of the XRPD of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form C.
Figure 8 is a DSC representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form C.
Figure 9 is a TGA representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form C.
Figure 10 is a graphical representation of the XRPD of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form D.
Figure 11 is a DSC representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form D.
Figure 12 is a TGA representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form D.
Figure 13 is a graphical representation of the XRPD of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form E.
Figure 14 is a DSC representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form E.
Figure 15 is a TGA representation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystalline form E.
Detailed Description
The present invention is further described below with reference to examples, which are not intended to limit the scope of the present invention.
Preparation of compounds
Examples
The structure of the compounds of the invention is determined by Nuclear Magnetic Resonance (NMR) or/and liquid mass chromatography (LC-MS). NMR chemical shifts (δ) are given in parts per million (ppm). NMR was measured using a Bruker AVANCE-400 NMR spectrometer using deuterated dimethyl sulfoxide (DMSO-d)6) Deuterated methanol (CD)3OD) and deuterated chloroform (CDCl)3) Internal standard is Tetramethylsilane (TMS).
LC-MS was measured using an Agilent 1200Infinity Series Mass spectrometer. HPLC was carried out using an Agilent 1200DAD high pressure liquid chromatograph (Sunfire C18150X 4.6mm column) and a Waters 2695-2996 high pressure liquid chromatograph (Gimini C18150X 4.6mm column).
The thin layer chromatography silica gel plate adopts a tobacco yellow sea HSGF254 or Qingdao GF254 silica gel plate, the specification adopted by TLC is 0.15 mm-0.20 mm, and the specification adopted by the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm. The column chromatography generally uses 200-300 mesh Titai Huanghai silica gel as a carrier.
The starting materials in the examples of the present invention are known and commercially available, or can be synthesized according to methods known in the art.
All reactions of the present invention are carried out under continuous magnetic stirring in a dry nitrogen or argon atmosphere, without specific indication, the solvent is a dry solvent, and the reaction temperature is given in degrees celsius.
Example 1
N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide
Figure BDA0003354163630000171
The first step is as follows: preparation of 2-chloro-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-5-carbonitrile
Figure BDA0003354163630000172
2, 4-dichloropyrimidine-5-carbonitrile (2g,12mmol) was dissolved in dichloroethane (30mL), cooled to 0 deg.C, ferric trichloride (3.9g,24mmol) was added, the reaction was stirred at room temperature for half an hour, 1-cyclopropyl-1H-indole (2.17g,14mmol) was added, and the reaction was stirred at 60 deg.C for 2 hours. Water (30mL) was added, filtration was performed, the filtrate was extracted with dichloromethane (30mL x2), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, spun-dried, and the crude product was purified by column chromatography (petroleum ether/ethyl acetate: 100/1-3/1) to give 2-chloro-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-5-carbonitrile (1.6g, yield: 47%) as a brown solid.
The second step is that: preparation of 4- (1-cyclopropyl-1H-indol-3-yl) -2- ((4-fluoro-2-methoxy-5-nitrophenyl) amino) pyrimidine-5-carbonitrile
Figure BDA0003354163630000173
2-chloro-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-5-carbonitrile (1g,3.4mmol), 4-fluoro-2-methoxy-5-nitroaniline (0.7g,3.7mmol) and p-toluenesulfonic acid (0.7g,3.7mmol) were dissolved in 2-pentanol (40mL) and the reaction was stirred at 100 ℃ overnight. Cooling to room temperature, adding water (50mL), extracting with dichloromethane (50mL × 2), drying the organic phase with anhydrous sodium sulfate, filtering, spin-drying, and purifying the crude product by column chromatography (petroleum ether/ethyl acetate: 100/1-1/1-dichloromethane/ethyl acetate: 100/1-10/1) to obtain 4- (1-cyclopropyl-1H-indol-3-yl) -2- ((4-fluoro-2-methoxy-5-nitrophenyl) amino) pyrimidine-5-carbonitrile (1g, 67%) as a brown solid.
The third step: preparation of 4- (1-cyclopropyl-1H-indol-3-yl) -2- ((4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitrophenyl) amino) pyrimidine-5-carbonitrile
Figure BDA0003354163630000181
4- (1-cyclopropyl-1H-indol-3-yl) -2- ((4-fluoro-2-methoxy-5-nitrophenyl) amino) pyrimidine-5-carbonitrile (0.4g,0.9mmol) was dissolved in acetonitrile (20mL), potassium carbonate (0.37g,2.7mmol) and N1, N1, N2-trimethylethane-1, 2-diamine (0.11g,1.1mmol) were added, and the reaction was stirred at 80 ℃ for 1H. The reaction solution was cooled to room temperature, filtered, and the filtrate was spin-dried to give crude 4- (1-cyclopropyl-1H-indol-3-yl) -2- ((4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitrophenyl) amino) pyrimidine-5-carbonitrile (0.48g, yield: 100% crede) as a yellow solid.
The fourth step: preparation of 2- ((5-amino-4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxyphenyl) amino) -4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-5-carbonitrile
Figure BDA0003354163630000182
4- (1-cyclopropyl-1H-indol-3-yl) -2- ((4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitrophenyl) amino) pyrimidine-5-carbonitrile (0.48g,0.97mmol) was dissolved in ethanol (20mL), a saturated sodium bicarbonate solution (10mL) and iron powder (0.54g,9.7mmol) were added, and the reaction was stirred at 80 ℃ for 2 hours. Cooled to room temperature, the reaction was filtered, the filtrate was extracted with dichloromethane (20mL × 2), the organic phase was dried over anhydrous sodium sulfate, filtered and dried by spinning to give 2- ((5-amino-4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxyphenyl) amino) -4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-5-carbonitrile (0.4g, yield: 88%) as a yellow solid.
The fifth step: preparation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide
Figure BDA0003354163630000191
2- ((5-amino-4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxyphenyl) amino) -4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-5-carbonitrile (0.4g,0.8mmol) was dissolved in tetrahydrofuran (10mL), cooled to 0 deg.C, triethylamine (0.12g,1.2mmol) was added, 3-chloropropionylchloride (0.12g,0.97mmol) was added, and the reaction stirred at 0 deg.C for 1H. 3M sodium hydroxide solution (3mL) was added and the reaction stirred at room temperature for 1 hour. Water (30mL) was added, extracted with dichloromethane (30mL x2), the organic phase was dried over anhydrous sodium sulfate, filtered, spun dry, and the crude product was purified by prep-TLC separation (dichloromethane/MeOH: 20/1) to afford N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (0.15g, yield: 34%) as a yellow solid.
MS m/z(ESI):551.4[M+H]+.
1H NMR(400MHz,CDCl3)δ10.08(s,1H),9.42(s,1H),8.68(s,1H),8.49-8.47(m,2H),7.76(s,1H),7.61(d,J=8.0Hz,1H),7.30-7.26(m,2H),6.81(s,1H),6.39(s,2H),5.69-5.67(m,1H),3.89(s,3H),3.48-3.44(m,1H),2.93-2.91(m,2H),2.73(s,3H),2.35-2.30(m,8H),1.17-1.11(m,4H).
Example 2
N- (5- ((4- (1-cyclopropyl-1H-indol-3-yl) -5- (oxazol-2-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide
Figure BDA0003354163630000192
Example 2 the preparation is as in example 1, with the following synthetic route:
Figure BDA0003354163630000201
MS m/z(ESI):593.1[M+H]+.
1H NMR(400MHz,CDCl3)δ9.65(s,1H),8.86(s,1H),8.14–7.98(m,1H),7.76(s,1H),7.57–7.44(m,2H),7.21–7.12(m,2H),7.02–6.94(m,1H),6.73(s,1H),6.47(d,J=17.2Hz,1H),5.73(d,J=11.7Hz,1H),5.40–5.22(m,1H),3.88(s,3H),3.43–3.36(m,1H),3.22–3.04(m,2H),2.88(s,2H),2.73(s,3H),2.70–2.35(m,6H),2.25–2.18(m,1H),2.05–1.96(m,1H),1.15–1.01(m,4H).
example 6
N- (5- ((5-cyano-4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide
Figure BDA0003354163630000202
Figure BDA0003354163630000211
The first step is as follows: preparation of 3- (5-bromo-2-chloropyrimidin-4-yl) -1H-indole
Figure BDA0003354163630000212
Indole (5.0g,54.86mmol) was dissolved in 2-methyltetrahydrofuran (50mL) under ice-cooling, methylmagnesium bromide (3.0M 2-methyltetrahydrofuran solution, 18.3mL,54.86mmol) was added dropwise thereto, the internal temperature was maintained at 30 ℃ or less, and after the addition was completed, the mixture was stirred at room temperature for 30 minutes. Then, a solution of 2, 4-dichloro-5-bromopyrimidine in 2-methyltetrahydrofuran (5.0g,21.94mmol,10mL solvent) was added dropwise thereto, and after completion of the addition, the reaction was stirred at room temperature for half an hour, then heated to 70 ℃ and reacted for 14 hours. Cooling to room temperature, pouring the reaction solution into saturated ammonium chloride aqueous solution, separating out solid, and filtering to obtain solid. The resulting solid was resuspended in water, sonicated, filtered to give a solid, dried to give the product 3- (5-bromo-2-chloropyrimidin-4-yl) -1H-indole (4.0g, yield: 59%, yellow solid).
MS m/z(ESI):308.0[M+H]+.
1H NMR(400MHz,DMSO-d6)δ12.21(s,1H),8.86(s,1H),8.80(s,1H),8.46(d,J=7.2Hz,1H),7.55(d,J=7.2Hz,1H),7.30-7.23(m,2H).
The second step is that: preparation of 5-bromo-N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (1H-indol-3-yl) pyrimidin-2-amine
Figure BDA0003354163630000221
3- (5-bromo-2-chloropyrimidin-4-yl) -1H-indole (0.50g,1.62mmol), 4-fluoro-2-methoxy-5-nitroaniline (0.33g,1.78mmol) and p-toluenesulfonic acid (0.34g,1.78mmol) were dissolved in 2-pentanol (25mL) and reacted at 100 ℃ for 14 hours. Cooled to room temperature, the reaction was neutralized with saturated sodium bicarbonate solution, stirred at room temperature for 20 minutes, filtered, the filter cake was washed with 2-pentanol and finally with petroleum ether, and the filter cake was dried to give 5-bromo-N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (1H-indol-3-yl) pyrimidin-2-amine (0.40g, 54%, as an earth yellow solid).
MS m/z(ESI):458.0,460.0[M+H]+.
The third step: preparation of N1- (5-bromo-4- (1H-indol-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl) -2-methoxy-N4-methyl-5-nitrobenzene-1, 4-diamine
Figure BDA0003354163630000222
5-bromo-N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (1H-indol-3-yl) pyrimidin-2-amine (0.40g,0.87mmol) was dissolved in acetonitrile (20mL), potassium carbonate (0.36g,2.61mmol) and N1, N1, N2-trimethylethane-1, 2-diamine (0.10g,0.96mmol) were added and the reaction stirred at 80 ℃ for two hours. The reaction mixture was cooled to room temperature, diluted with ethyl acetate, washed with saturated brine, and the organic phase was dried over anhydrous sodium sulfate, filtered, and dried by spinning to give N1- (5-bromo-4- (1H-indol-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl) -2-methoxy-N4-methyl-5-nitrophenyl-1, 4-diamine (0.40g, yield: 87%, red solid).
MS m/z(ESI):540.2,542.2[M+H]+.
The fourth step: preparation of N1- (5-bromo-4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl) -2-methoxy-N4-methyl-5-nitrobenzene-1, 4-diamine
Figure BDA0003354163630000223
N1- (5-bromo-4- (1H-indol-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl) -2-methoxy-N4-methyl-5-nitrobenzene-1, 4-diamine (0.30g,0.56mmol), cesium carbonate (0.13g,0.72mmol) and 3-iodo-oxetane (0.36g,1.11mmol) were dissolved in N, N-dimethylformamide (8mL) at room temperature, reacted with microwave at 110 ℃ for 1.5 hours, and cooled to room temperature. The reaction solution was diluted with ethyl acetate, filtered, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, spun-dried, and the crude product was isolated on a silica gel preparation plate (dichloromethane: methanol: 20: 1) to give N1- (5-bromo-4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl) -2-methoxy-N4-methyl-5-nitrobenzene-1, 4-diamine (0.12g, yield: 36%, red solid).
MS m/z(ESI):596.0[M+H]+.
The fifth step: preparation of 2- ((4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitrophenyl) amino) -4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidine-5-carbonitrile
Figure BDA0003354163630000231
N1- (5-bromo-4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidin-2-yl) -N4- (2- (dimethylamino) ethyl) -2-methoxy-N4-methyl-5-nitrobenzene-1, 4-diamine (0.12g,0.20mmol), Pd, was added at room temperature2(dba)3(18.4mg, 0.02mmol), X-Phos (19mg,0.04mmol), zinc cyanide (23.5mg, 0.20mmol) and zinc powder (13mg, 0.20mmol)) Dissolved in N, N-dimethylacetamide (3mL), the nitrogen gas was replaced, and the reaction was carried out by microwave at 110 ℃ for 1.5 hours. Cooled to room temperature, filtered, the solid was washed with ethyl acetate, the organic phase was concentrated under reduced pressure, and the crude product was isolated with a preparative plate (dichloromethane: methanol: 20: 1) to give 2- ((4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitrophenyl) amino) -4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidine-5-carbonitrile (0.096g, yield: 88%, yellow solid).
MS m/z(ESI):543.1[M+H]+.
And a sixth step: preparation of 2- ((5-amino-4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxyphenyl) amino) -4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidine-5-carbonitrile
Figure BDA0003354163630000232
2- ((4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitrophenyl) amino) -4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidine-5-carbonitrile (0.12g,0.22mmol) was dissolved in ethanol (10mL) and then aqueous ammonium chloride (0.06g,1.1mmol, water, 2mL) and iron powder (0.12g,2.2mmol) were added and the reaction stirred at 80 ℃ for 2H. Cooled to room temperature, the reaction was filtered, the filter cake was washed with dichloromethane, the organic solvent was removed under reduced pressure, the residue was dissolved with dichloromethane and water, the layers were separated, the aqueous phase was extracted with dichloromethane (20mL x2) again, the organic phases were combined, the organic phase was dried over anhydrous sodium sulfate, filtered, and dried by spinning to give 2- ((5-amino-4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxyphenyl) amino) -4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidine-5-carbonitrile (0.095g, yield: 83.8%, yellow oil).
MS m/z(ESI):513.2[M+H]+.
The seventh step: preparation of N- (5- ((5-cyano-4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide
Figure BDA0003354163630000241
2- ((5-amino-4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxyphenyl) amino) -4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidine-5-carbonitrile (0.075g,0.146mmol) was dissolved in dichloromethane (5mL), cooled to 0 deg.C, triethylamine (0.03g,0.292mmol) was added, 3-chloropropionylchloride (0.024g,0.19mmol) was added, and the reaction was stirred at 0 deg.C for 1H. After the reaction, the reaction was dried by spinning, the crude product was dissolved in dichloromethane, washed with saturated sodium bicarbonate solution, washed with saturated brine, and the organic phase was dried over anhydrous sodium sulfate, filtered and dried by spinning. The crude product was dissolved in acetonitrile (5mL), and then aqueous sodium hydroxide (0.058g,1.46mmol, water, 0.5mL) was added and reacted at 40 ℃ for two hours. The reaction mixture was cooled to room temperature, diluted with ethyl acetate, washed with saturated brine, the organic phase was dried over anhydrous sodium sulfate, filtered, spun-dried and the crude product was purified by preparative chromatography to give N- (5- ((5-cyano-4- (1- (oxetan-3-yl) -1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide (0.04g, yield: 48%, yellow solid).
MS m/z(ESI):567.2[M+H]+.
1H NMR(400MHz,CDCl3)δ10.15-9.82(br,1H),9.45(s,1H),8.69(s,2H),8.55-8.38(br,1H)7.80(s,1H),7.61(d,J=8.0Hz,1H),7.33-7.29(m,2H),6.78(s,1H),6.42-6.38(m,1H),5.72-5.66(m,2H),5.28-5.13(m,5H),3.90(s,3H),3.13-2.95(br,2H),2.74(s,3H),2.61-2.24(br,6H),1.85-1.53(m,2H).
Example 9
N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide
Figure BDA0003354163630000242
Figure BDA0003354163630000251
The first step is as follows: preparation of 6-chloro-2-methoxypyridin-3-amine
Figure BDA0003354163630000252
6-chloro-2-methoxy-3-nitropyridine (5g,26.6mmol) was dissolved in ethanol (100mL) and water (30mL), ammonium chloride (7.0g,133mmol) was added, iron powder (7.5g,133mmol) was added in portions, and the reaction was stirred at 85 ℃ for 2 hours. The reaction solution was cooled to room temperature, filtered through celite, and ethyl acetate (150mL) and saturated brine (120mL) were added to the filtrate to separate an organic layer, which was dried over anhydrous sodium sulfate, filtered, and dried by spinning to give 6-chloro-2-methoxypyridin-3-amine (4g, yield: 95%) as a brown solid.
MS m/z(ESI):159.1[M+H]+.
The second step is that: preparation of N- (6-chloro-2-methoxypyridin-3-yl) acetamide
Figure BDA0003354163630000253
6-chloro-2-methoxypyridin-3-amine (4.0g,25.0mmol) was dissolved in dichloromethane (100mL), diisopropylethylamine (4.8g,37.5mmol) was added, cooled to 0 deg.C, acetyl chloride (2.4g,30.0mmol) was added and stirring continued for 2 h. The reaction mixture was washed with 80mL of water, 80mL of 1N hydrochloric acid and 80mL of saturated brine in this order, dried over anhydrous sodium sulfate, filtered and spun to give N- (6-chloro-2-methoxypyridin-3-yl) acetamide (4.0g, yield: 79%) as a brown solid.
MS m/z(ESI):201.1[M+H]+.
The third step: preparation of N- (6-chloro-2-methoxy-5-nitropyridin-3-yl) acetamide
Figure BDA0003354163630000261
6-chloro-2-methoxypyridin-3-amine (2.0g,10.0mmol) was dissolved in trifluoroacetic anhydride (20mL), cooled to-10 deg.C, fuming nitric acid (0.5mL,10mmol) was added dropwise, and stirring was continued for 2 hours. Crushed ice was added to the reaction solution, which was extracted with dichloromethane (50mL), dried over anhydrous sodium sulfate, filtered, and spin-dried to give N- (6-chloro-2-methoxy-5-nitropyridin-3-yl) acetamide (1.6g, yield: 65%) as a brown solid.
MS m/z(ESI):244.1[M-H]+.
The fourth step: preparation of N- (6- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitropyridin-3-yl) acetamide
Figure BDA0003354163630000262
N- (6-chloro-2-methoxy-5-nitropyridin-3-yl) acetamide (1.6g,6.5mmol) was dissolved in acetonitrile (30mL), N1, N1, N2-trimethylethane-1, 2-diamine (1g,9.8mmol) was added and the reaction stirred at 80 ℃ for 3 hours. The solvent was dried by spinning, and the crude product was purified by column chromatography (dichloromethane/methanol: 100/1-10/1) to give N- (6- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitropyridin-3-yl) acetamide (0.9g, yield: 45%) as a brown solid.
MS m/z(ESI):312.1[M+H]+.
The fifth step: preparation of N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine
Figure BDA0003354163630000263
N- (6- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitropyridin-3-yl) acetamide (0.9g,2.9mmol) was dissolved in methanol (30mL), concentrated hydrochloric acid (5mL) and the reaction stirred at 60 ℃ for 3 h. The solvent was dried by spinning, dichloromethane (50mL) and saturated sodium bicarbonate (50mL) were added, stirring was carried out until no bubble was precipitated, the organic layer was separated, dried over anhydrous sodium sulfate, filtered, spun-dried, and the crude product was purified by column chromatography (dichloromethane/methanol: 100/1-10/1) to give N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (0.15g, yield: 19%) as a brown solid.
MS m/z(ESI):270.1[M+H]+.
And a sixth step: preparation of 4- (1-cyclopropyl-1H-indol-3-yl) -2- ((6- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitropyridin-3-yl) amino) pyrimidine-5-carbonitrile
Figure BDA0003354163630000271
N2- (2- (dimethylamino) ethyl) -6-methoxy-N2-methyl-3-nitropyridine-2, 5-diamine (0.11g,0.41mmol), 2-chloro-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-5-carbonitrile (0.12g,0.41mmol), tris (dibenzylideneacetone) dipalladium (0.18g,0.2mmol), x-phos (0.2g,0.41mmol), sodium tert-butoxide (0.12g,1.2mmol) were dissolved in dioxane (5mL), nitrogen was replaced, and the reaction was stirred at 140 ℃ under microwave for 1 hour. The reaction mixture was cooled to room temperature, filtered, the filtrate was dried by spinning, and the crude product was isolated by prep-TLC (dichloromethane/methanol: 20/1) to give 4- (1-cyclopropyl-1H-indol-3-yl) -2- ((4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitrophenyl) amino) pyrimidine-5-carbonitrile (0.1g, yield: 47%) as a yellow solid.
MS m/z(ESI):528.1[M+H]+.
The seventh step: preparation of 2- ((5-amino-6- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxypyridin-3-yl) amino) -4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-5-carbonitrile
Figure BDA0003354163630000272
4- (1-cyclopropyl-1H-indol-3-yl) -2- ((4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitrophenyl) amino) pyrimidine-5-carbonitrile (80mg,0.15mmol) was dissolved in methanol (10mL), raney nickel (80mg) and 85% hydrazine hydrate (90mg,1.5mmol) were added at 0 ℃, and the reaction was stirred at room temperature for 2 hours. The reaction was filtered, the filtrate was spun dry, water (15mL) was added, extracted with dichloromethane (15mL x2), the organic phase was dried over anhydrous sodium sulfate, filtered, and spun dry to give 2- ((5-amino-6- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxypyridin-3-yl) amino) -4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-5-carbonitrile (80mg, yield: 100% crude) as a brown solid.
MS m/z(ESI):498.1[M+H]+.
Eighth step: preparation of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide
Figure BDA0003354163630000281
2- ((5-amino-6- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxypyridin-3-yl) amino) -4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-5-carbonitrile (80mg,0.16mmol) was dissolved in dichloromethane (10mL), cooled to 0 deg.C, triethylamine (24mg,0.24mmol) was added, 3-chloropropionyl chloride (25mg,0.19mmol) was added, and the reaction stirred at 0 deg.C for 1H. Water (10mL) was added, extracted with dichloromethane (10mL x2), the organic phase was dried over anhydrous sodium sulfate, filtered, spun dry, tetrahydrofuran (5mL) was added, a solution of sodium hydroxide (64mg,1.6mmol) in water (0.5mL) was added, and the reaction stirred at 40 ℃ overnight. Water (10mL) was added, extracted with ethyl acetate (10mL x2), the organic phase was dried over anhydrous sodium sulfate, filtered, spun dry, and the crude product was isolated by prep-HPLC to give N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -6-methoxypyridin-3-yl) acrylamide (0.9mg, yield: 0.7%) as a yellow solid.
MS m/z(ESI):552.1[M+H]+.
The preparation of other examples is as follows:
Figure BDA0003354163630000282
Figure BDA0003354163630000291
Figure BDA0003354163630000301
Figure BDA0003354163630000311
II,Biological test evaluation
The present invention is further described and explained below in conjunction with test examples, which are not intended to limit the scope of the present invention.
1. Testing of enzymology experiments
Test example 1 determination of inhibitory Effect of the Compound of the present invention on EGFR20 exon insertion mutation kinase Activity
Purpose of the experiment: the purpose of this test example was to measure the inhibitory ability of compounds on the EGFR20 exon insertion mutant kinase activity.
An experimental instrument: centrifuge (Eppendorf 5810R), microplate reader (BioTek Synergy H1), pipette (Eppendorf & Rainin)
The experimental method comprises the following steps: in the experiment, the inhibiting activity of the compound on EGFR20 exon insertion mutation kinase is researched by adopting a TR-FRET (time-resolved fluorescence resonance energy transfer) method. The experiment was performed in 384 well plates and experiment buffer (50mM HEPES, 1mM EGTA, 10mM MgCl)22mM DTT, 0.01% Tween-20), compounds were gradient diluted to different concentrations using assay buffer, 2.5 μ L per well were added to 384 well plates, 2.5 μ L of diluted EGFR kinase solution (0.001-0.5nM) was added, incubation was performed at room temperature for 10 minutes, 5 μ L of ULight-poly GT/ATP mixed solution was added, incubation was performed at room temperature for 30 minutes to 60 minutes, 5 μ L of EDTA-terminated reaction and 5 μ L of Eu-labeled antibody detection solution were added, incubation was performed at room temperature for 1 hour, and 665nM fluorescence signal values were measured for each well plate with a microplate reader.
The experimental data processing method comprises the following steps:
inhibition ((positive control well value-sample well value)/(positive control well value-negative control well value)) -100% was calculated using the fluorescence signal value at 665nm, and the concentration and inhibition were subjected to nonlinear regression curve using Graphpad Prism softwareLine fitting to obtain IC50The values, specific data are shown in table 1 below:
EGFR20 exon insertion mutant kinase inhibitory Activity IC with the Compounds of Table 150
Figure BDA0003354163630000321
And (4) experimental conclusion:
the scheme shows that the compound of the embodiment of the invention has better inhibition effect in EGFR20 exon insertion mutation kinase activity inhibition experiments.
Test example 2 determination of inhibitory Effect of the Compound of the present invention on EGFR wild-type kinase Activity
Purpose of the experiment: the purpose of this test example was to measure the inhibitory ability of compounds on EGFR wild-type kinase activity.
An experimental instrument: centrifuge (Eppendorf 5810R), microplate reader (BioTek Synergy H1), pipette (Eppendorf & Rainin)
The experimental method comprises the following steps: in the experiment, the inhibiting activity of the compound on EGFR wild type kinase is researched by adopting a TR-FRET (time-resolved fluorescence resonance energy transfer) method. The experiment was performed in 384 well plates and experiment buffer (50mM HEPES, 1mM EGTA, 10mM MgCl)22mM DTT, 0.01% Tween-20), compounds were gradient diluted to different concentrations using assay buffer, 2 μ L per well were added to 384 well plates, 4 μ L of diluted EGFR kinase solution (0.001-0.5nM) was added, incubation was performed at room temperature for 10 minutes, 4 μ LULight-poly GT/ATP mixed solution was added, incubation was performed at room temperature for 30 to 60 minutes, 5 μ L of EDTA-terminated reaction and 5 μ L of Eu-labeled antibody detection solution were added, incubation was performed at room temperature for 1 hour, and 665nM fluorescence signal values of each well plate were measured with a microplate reader.
The experimental data processing method comprises the following steps:
the inhibition ratio ((positive control well value-sample well value)/(positive control well value-negative control well value)). 100% was calculated using the fluorescence signal value at 665nm, and the concentration and inhibition ratio were fitted to a nonlinear regression curve using Graphpad Prism software to obtain IC50Value, concrete dataAs shown in table 2 below:
table 2 Compound inhibitory Activity on EGFR wild-type kinase IC50
Figure BDA0003354163630000331
And (4) experimental conclusion:
the scheme shows that the compounds of the embodiment of the invention have small inhibition effect in an EGFR wild type kinase inhibition test.
Test example 3 measurement of proliferation inhibitory Effect of the Compound of the present invention on Ba/F3 EGFR mutant cell line and A431 cell line
Purpose of the experiment: the purpose of this test example was to measure the inhibitory effect of the compounds on the proliferation activity of Ba/F3 EGFR mutant cell line and A431 cell line.
An experimental instrument: enzyme-labeling instrument (BioTek Synergy H1), pipette (Eppendorf & Rainin)
The experimental method comprises the following steps:
culturing Ba/F3 EGFR mutant cells to appropriate density, collecting cells, adjusting the cell concentration to appropriate using complete medium, spreading the cell suspension in 96-well plate at 90. mu.L/well, placing at 37 deg.C and 5% CO2Adhering the incubator to the wall overnight, preparing compound solutions with different concentrations by using DMSO and a culture medium, setting a solvent control, adding the compound solutions into a 96-well plate, placing 10 mu L of each well, placing at 37 ℃ and 5% CO2And (3) continuously culturing for 72-144H in the incubator, adding CellTiter-Glo solution, shaking and mixing uniformly, incubating for 10 minutes in a dark place, and reading by using a BioTek Synergy H1 enzyme-labeling instrument.
The experimental method comprises the following steps:
culturing A431 cells to appropriate density, collecting cells, adjusting the cell concentration to appropriate using complete medium, spreading the cell suspension in 96-well plate, 90 μ L per well, placing at 37 deg.C and 5% CO2Adhering the incubator to the wall overnight, preparing compound solutions with different concentrations by using DMSO and a culture medium, setting a solvent control, adding the compound solutions into a 96-well plate, placing 10 mu L of each well, placing at 37 ℃ and 5% CO2After the culture in the incubator is continued for 72H, CellTiter-Glo solution is added, after shaking and mixing uniformly, the mixture is incubated for 10 minutes in the dark, and reading is carried out by a BioTek Synergy H1 enzyme-labeling instrument.
The experimental data processing method comprises the following steps:
calculating the inhibition rate by using the luminescence signal value, and fitting the concentration and the inhibition rate by using Graphpad Prism software to obtain IC50The values, specific data are shown in table 3 below:
proliferation inhibitory Activity of the Compounds of Table 3 on Ba/F3 EGFR mutant cell line and A431 cell line IC50
Figure BDA0003354163630000341
And (4) experimental conclusion:
the scheme shows that the compound of the embodiment of the invention has good inhibition effect in the inhibition test of the proliferation activity of Ba/F3 EGFR mutant cells and has poor inhibition effect on A431 cells, and the comparative data show that the series of embodiments of the invention have high selectivity on the inhibition of the proliferation activity of Ba/F3 EGFR mutant cells.
Third, research on crystal form
1. Laboratory apparatus
1.1 some parameters of the physicochemical measuring instrument
Figure BDA0003354163630000342
Figure BDA0003354163630000351
2. Preparation of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide in different crystalline forms
2.1 preparation of form A
Figure BDA0003354163630000352
66.0g of 4- (1-cyclopropyl-1H-indol-3-yl) -2- ((4- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methoxy-5-nitrophenyl) amino) pyrimidine-5-carbonitrile, 660mL of tetrahydrofuran, was added to a 2L three-necked flask; palladium on carbon (6.6g), N were added2Bubbling for half an hour below the liquid level; replacing 3 times with hydrogen, heating to 35-40 ℃, reacting for 20-24 h, filtering, washing filter cake with tetrahydrofuran (100mL), and filtering to finish N2Protecting the filtrate, cooling to 0-5 deg.C, and adding triethylamine (12.6 g); controlling the temperature to be 0-5 ℃, and dropwise adding a tetrahydrofuran (120mL) solution of 3-chloropropionyl chloride (23.7g) for about 45 minutes; the reaction was maintained at 0-5 ℃ for 2-3 hours, filtered, and the filter cake was washed with tetrahydrofuran (100mL) and then rinsed with 100mL of N-heptane to give N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) 3-chloropropanamide hydrochloride (98.9g, 100% yield).
Adding 50.0g of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) 3-chloropropamide hydrochloride and 420mL of THF to a 2L three-necked flask and stirring; 42mL of potassium hydroxide 42.0g aqueous solution is dripped, and after dripping is finished in about 10min, the mixture is kept at room temperature and stirred for 30 min; heating to 35-40 ℃ for reaction for 1h, and completely reacting the raw materials; cooling to room temperature, adding 500mL of water, stirring for 10 minutes, standing for layering, retaining an organic phase, and extracting a water phase with 300mL of 2-methyltetrahydrofuran; the organic phases are combined and washed by 200mL of water and 200mL of saturated saline solution in sequence; adding 2.5g of activated carbon and 25g of anhydrous sodium sulfate into the organic phase, and stirring for 1h at room temperature (20 +/-5 ℃); filtering, concentrating the filtrate to obtain yellow solid about 70g, adding 150mL of 2-methyltetrahydrofuran, heating to 80 ℃ for refluxing, heating and stirring for 1 hour, and naturally cooling to room temperature; the mixture was cooled in an ice bath (0-5 ℃), stirred for 1 hour and filtered, and the filter cake was washed with 2-methyltetrahydrofuran (50ml) to give the desired product N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide as crystalline form a (24.0g, 69.7% yield) with an XRPD pattern as shown in fig. 1, a DSC pattern as shown in fig. 2, and a TGA pattern as shown in fig. 3, which were detected and analyzed.
2.2 preparation of form B
Weighing 10mg of N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form A, adding 100 muL of toluene solvent, pulping at 25 ℃ for two weeks, centrifuging, drying at 50 ℃ in vacuum to constant weight to obtain N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form B, and (5) detecting and analyzing. Has an XRPD pattern as shown in figure 4, a DSC pattern as shown in figure 5 and a TGA pattern as shown in figure 6.
2.3 preparation of form C
Weighing 10mg of compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form a, adding 100 μ L of 1, 4-dioxane solvent, pulping at 25 ℃ for two weeks, centrifuging, and drying in vacuo at 50 ℃ to constant weight to obtain compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form C. And (5) detecting and analyzing. Has an XRPD pattern as shown in figure 7, a DSC pattern as shown in figure 8 and a TGA pattern as shown in figure 9.
2.4 preparation of form D
Weighing 10mg of compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form a, adding 100 μ L of ethyl formate solvent, pulping at 25 ℃ for two weeks, centrifuging, and vacuum drying at 50 ℃ to constant weight to obtain compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form D. By detection and analysis, the XRPD pattern shown in figure 10, the DSC pattern shown in figure 11 and the TGA pattern shown in figure 12 are as follows.
2.5 preparation of form E
Weighing 10mg of compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form a, adding 100 μ L of ethyl formate solvent, pulping for one day at 50 ℃, centrifuging, and vacuum drying at 50 ℃ to constant weight to obtain compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form E. By detection analysis, it has the following XRPD pattern as shown in FIG. 13, DSC pattern as shown in FIG. 14 and TGA pattern as shown in FIG. 15.
3. Solid stability test
3.1 purpose of experiment:
the physical and chemical stability of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form A under the conditions of 5000lx of illumination, 60 ℃ of high temperature, 92.5% RH of high humidity and 50 ℃ of 75% RH of high humidity is considered, and a basis is provided for compound storage.
3.2 Instrument and conditions for liquid phase analysis
3.2.1 instruments and devices:
name of instrument Model number
Analytical balance Sartorius BSA224S-CW
Water purifier Milli-Q Plus,Millipore
High performance liquid chromatograph Agilent1260
Pump Agilent G1311B
Sample injector G1329B
Column oven G1316A
Detector G1315D
3.2.2 chromatographic conditions:
Figure BDA0003354163630000371
3.3 protocol:
taking about 1mg of crystal form A of a compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, inspecting for 5 days and 10 days under the conditions of 5000lx illumination, 60 ℃ high temperature, 92.5% RH high humidity and 50 ℃ high humidity, 75% RH high temperature, measuring the content by using an HPLC (high performance liquid chromatography) external standard method, and calculating the change of related substances by using a chromatographic peak area normalization method.
3.4 Experimental results:
Figure BDA0003354163630000381
3.5 conclusion of the experiment:
the data show that the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form A is relatively stable under the illumination condition, is degraded under the conditions of high temperature of 60 ℃, high humidity of 92.5 percent RH and high temperature of 50 ℃ and 75 percent RH, and needs to be stored in shade in the later storage process.
4. Dynamic hygroscopicity test
4.1 purpose of experiment:
the hygroscopicity of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form A under different relative humidity conditions is examined, and a basis is provided for compound storage.
4.2 Experimental protocol:
the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form A is placed in saturated water vapor with different relative humidity, so that the compound and the water vapor reach dynamic equilibrium, and the percentage of the compound which absorbs moisture and gains weight after the equilibrium is calculated.
4.3 Experimental results and conclusions:
the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, form a, had a hygroscopic weight gain of about 0.521% at RH 80%, a slightly hygroscopic property. After the moisture absorption and desorption cycle is carried out for 2 times under the condition of 0-95% relative humidity, the XRPD spectrogram of the crystal form A is not changed, namely the crystal form is not transformed.
5. Solubility experiments in different media
5.1 purpose of experiment:
comparison of the solubility of the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide form a in USP buffer, pH1-8, artificial simulated gastric fluid (FaSSGF), fasted artificial simulated intestinal fluid (FaSSIF), non-fasted artificial simulated intestinal fluid (FeSSIF), fasted artificial simulated colon fluid (fasscuf), non-fasted artificial simulated colon fluid (FeSSCoF), and purified water provides a basis for the evaluation of drugability.
5.2 protocol:
compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, form A1 mg, was suspended in different media for 24 hours and the thermodynamic solubility of compound at 37 ℃ was determined by HPLC, external standard method.
5.3 Experimental results:
sample name Solubility of form A (mg/mL)
pH1 >1
pH 2 >1
pH 3 0.02
pH 4 0.01
pH 5 0.01
pH 6 0.00
pH 7 0.00
pH 8 0.00
H2O 0.00
FaSSGF >1
FaSSIF 0.16
FeSSIF >1
FaSSCoF 0.00
FeSSCoF 0.09
5.4 conclusion of the experiment:
the solubility of the comparative compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide form a in different media, form a having better solubility under acidic conditions, was compared.
6. Thermodynamic stability test
6.1 purpose of experiment:
through polycrystal screening, a relatively stable compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form is found.
6.2 protocol:
selecting an organic solvent and water with certain solubility, suspending different crystal forms in a solvent system, stirring and pulping for 2 weeks or 1 day at 25 ℃, centrifuging, discarding supernatant, vacuum drying the solid at 50 ℃ (-0.1Mpa) for 16 hours, and measuring XRPD, DSC and TGA of the solid for comparison.
6.3 Experimental results:
Figure BDA0003354163630000391
Figure BDA0003354163630000401
6.4 conclusion of the experiment:
the preparation method comprises the steps of pulping, changing a crystallization solvent and a crystallization mode to obtain 5 crystal forms which are respectively a crystal form A, a crystal form B, a crystal form C, a crystal form D and a crystal form E of a compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide. By comparing DSC spectra of different crystal forms, the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidine-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide crystal form A is judged to have better thermodynamic stability and to be an anhydrous compound.
7. Pharmacokinetic study in rats
7.1 purpose of the experiment:
rat pharmacokinetic parameters were studied for compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide free base crystalline form a.
7.2 laboratory instruments and reagents:
7.2.1 Instrument:
Figure BDA0003354163630000402
7.2.2 reagents:
Figure BDA0003354163630000403
Figure BDA0003354163630000411
7.3 Experimental animals:
animal species Line of Age (age) Sex Suppliers of goods
Rat SD
7 weeks, body weight 200g Male sex JOINN LABORATORIES (SUZHOU) Co.,Ltd.
7.4 test compounds:
the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide free base crystalline form a.
7.5 protocol:
the compound N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide free base form a was suspended homogeneously in an aqueous solution containing 0.5% HPMC (hydroxypropylmethylcellulose) K4M (1% Tween80), and then the mixture was gavaged, administered to rats, and three rats in parallel at a dose of 30mg/kg, with the amount of compound being converted to the same amount of free base.
7.6 Experimental results:
TABLE 7.1 rat pharmacokinetic experiment results
Figure BDA0003354163630000412
7.7 conclusion of the experiment:
as can be seen from the results of the rat pharmacological experiments in the table, compound 5N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide of the present invention exhibits good metabolic properties at a dose of 30 mg/kg.

Claims (10)

1. A crystal form of a compound shown as a general formula (I),
Figure FDA0003354163620000011
wherein:
R1selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, -C (O) NRaa(CH2)nRbbOr- (CH)2)nN=S(O)RaaRbb(ii) a Said amino, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, and heteroaryl, optionally further substituted with one or more substituents of deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
preferably, R1Selected from hydrogen, deuterium, halogen, cyano、C1-3Alkyl radical, C1-3Deuterated alkyl, C1-3Haloalkyl, C1-3Alkoxy radical, C1-3Haloalkoxy, 5-6 membered heteroaryl, -C (O) NRaa(CH2)nRbbOr- (CH)2)nN=S(O)RaaRbbSaid C is1-3Alkyl radical, C1-3Deuterated alkyl, C1-3Haloalkyl, C1-3Alkoxy radical, C1-3Haloalkoxy and 5-6 membered heteroaryl, optionally further substituted with deuterium, halogen, C1-3Alkyl radical, C1-3Deuterated alkyl, C1-3Haloalkyl, C1-3Alkoxy and C1-6One or more substituents of haloalkoxy;
more preferably, R1Selected from hydrogen, deuterium, cyano, 5-6 membered heteroaryl, -C (O) NRaa(CH2)nRbbOr- (CH)2)nN=S(O)RaaRbbSaid 5-6 membered heteroaryl, optionally, further substituted with deuterium, halogen, C1-3Alkyl and C1-3Substituted with one or more substituents in deuterated alkyl;
most preferably, R1Selected from hydrogen, cyano, oxazolyl, pyrazolyl,
Figure FDA0003354163620000012
Figure FDA0003354163620000013
R2Selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, or heteroaryl;
preferably hydrogen, deuterium, halogen, cyano, C1-3Alkyl radical, C1-3Deuterated alkyl, C1-3Haloalkyl, C1-3Alkoxy radical, C1-3Haloalkoxy, C3-6Cycloalkyl or 3-6 membered heterocyclyl;
more preferably hydrogen, deuterium, methyl, ethyl, propyl, cyclopropyl or oxetanyl;
most preferably hydrogen, methyl, cyclopropyl or oxetanyl;
R3selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, or heteroaryl;
preferably hydrogen, deuterium, halogen, cyano, C1-3Alkyl radical, C1-3Deuterated alkyl, C1-3Haloalkyl, C1-3Alkoxy or C1-3A haloalkoxy group;
more preferably hydrogen, deuterium, fluorine, chlorine, bromine, methoxy or ethoxy;
most preferably hydrogen, fluoro or methoxy;
Raaand RbbEach independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl; said amino, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, optionally further substituted with one or more substituents of deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
preferably, RaaAnd RbbEach independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-8Cycloalkyl, 3-8 membered heterocyclyl, C6-10Aryl or 5-10 membered heteroaryl; the amino group, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-8Cycloalkyl, 3-8 membered heterocyclyl, C6-10Aryl and 5-10 membered heteroaryl, optionally further substituted with deuterium, halogenAmino, nitro, hydroxy, cyano, C1-6Alkyl radical, C1-6Deuterated alkyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-8Cycloalkyl, 3-8 membered heterocyclyl, C6-10Aryl and 5-10 membered heteroaryl;
x is an integer of 0-4;
preferably, x is an integer of 0-2; and is
n is an integer of 0 to 4; preferably an integer of 0 to 2.
2. A crystalline form of the compound according to claim 1, characterized in that the specific compound has the following structure:
Figure FDA0003354163620000021
Figure FDA0003354163620000031
3. a crystalline form of the compound according to claim 1 or 2, characterized in that the compound is N- (5- ((5-cyano-4- (1-cyclopropyl-1H-indol-3-yl) pyrimidin-2-yl) amino) -2- ((2- (dimethylamino) ethyl) (methyl) amino) -4-methoxyphenyl) acrylamide, characterized in that the crystalline form is form a, form B, form C, form D, form E, wherein:
the X-ray powder diffraction pattern of the crystal form A has a diffraction peak at the 2 theta of 7.1 +/-0.2 degrees; or a diffraction peak at 14.1 ± 0.2 °; or a diffraction peak at 18.9 ± 0.2 °; or a diffraction peak at 22.7 ± 0.2 °; or a diffraction peak at 20.4 ± 0.2 °; or a diffraction peak at 22.3 ± 0.2 °; or a diffraction peak at 13.4 ± 0.2 °; or a diffraction peak at 12.9 ± 0.2 °; or a diffraction peak at 17.5 ± 0.2 °; or a diffraction peak at 20.8 ± 0.2 °; preferably comprises any of 2 to 5, or 3 to 6, or 3 to 8, or 5 to 8, or 6 to 8 of the diffraction peaks; more preferably any 6, 7 or 8 thereof;
preferably, form a has an X-ray powder diffraction pattern comprising at least one or more diffraction peaks, preferably 2, more preferably 3, located at 2 Θ of 7.1 ± 0.2 °, 14.1 ± 0.2 °, 18.9 ± 0.2 °; optionally, the compound can further comprise at least one position with 2 theta of 22.7 +/-0.2 degrees, 20.4 +/-0.2 degrees, 22.3 +/-0.2 degrees, 13.4 +/-0.2 degrees and 12.9 +/-0.2 degrees, preferably comprises 2 positions, 3 positions, 4 positions or 5 positions;
more preferably, the X-ray powder diffraction pattern of form a optionally further comprises one or more diffraction peaks at 17.5 ± 0.2 °, 20.8 ± 0.2 °, 9.6 ± 0.2 °, 22.0 ± 0.2 °, 25.6 ± 0.2 °, 14.9 ± 0.2 °, 15.6 ± 0.2 ° 2 Θ; preferably at least any 2-3, or 4-5, or 6-7 thereof; further preferably, any 2, 3, 4, 5, 6, 7 thereof;
further preferably, the X-ray powder diffraction pattern of form a comprises one or more diffraction peaks located at 7.1 ± 0.2 °, 14.1 ± 0.2 °, 18.9 ± 0.2 °, 22.7 ± 0.2 °, 20.4 ± 0.2 °, 22.3 ± 0.2 °, 13.4 ± 0.2 °, 12.9 ± 0.2, 17.5 ± 0.2 °, 20.8 ± 0.2 °, 9.6 ± 0.2 °, 22.0 ± 0.2 °, 25.6 ± 0.2 °, 14.9 ± 0.2 °, 15.6 ± 0.2 ° of 2 Θ; preferably, the compound comprises diffraction peaks at 4, 5, 6, 8 or 10 optionally;
most preferably, form a has an X-ray powder diffraction pattern as shown in figure 1;
the X-ray powder diffraction pattern of the crystal form B has a diffraction peak at the 2 theta of 6.5 +/-0.2 degrees; or a diffraction peak at 20.7 ± 0.2 °; or a diffraction peak at 20.9 ± 0.2 °; or a diffraction peak at 21.0 ± 0.2 °; or a diffraction peak at 21.4 ± 0.2 °; or a diffraction peak at 15.6 ± 0.2 °; or a diffraction peak at 23.8 ± 0.2 °; or a diffraction peak at 21.6 ± 0.2 °; or a diffraction peak at 9.2 ± 0.2 °; or a diffraction peak at 25.1 ± 0.2 °; preferably comprises any 2-5, or 3-6, or 3-8, or 5-8, or 6-8 of the diffraction peaks, more preferably comprises any 6, 7 or 8 thereof;
preferably, form B has an X-ray powder diffraction pattern comprising at least one, preferably two, more preferably three, diffraction peaks, located at one or more of 6.5 ± 0.2 °, 20.7 ± 0.2 °, 20.9 ± 0.2 ° 2 Θ; optionally, at least one of 21.0 ± 0.2 °, 21.4 ± 0.2 °, 15.6 ± 0.2 °, 23.8 ± 0.2 °, and 21.6 ± 0.2 ° may be further included; preferably 2, 3, 4 or 5 thereof;
more preferably, the X-ray powder diffraction pattern of form B optionally further comprises one or more diffraction peaks at 9.2 ± 0.2 °, 25.1 ± 0.2 °, 10.7 ± 0.2 °, 12.4 ± 0.2 °, 17.4 ± 0.2 °, 17.6 ± 0.2 °, 14.5 ± 0.2 ° 2 Θ; preferably at least any 2-3, or 4-5, or 6-7 thereof; further preferably, any 2, 3, 4, 5, 6, 7 thereof;
further preferably, the X-ray powder diffraction pattern of form B comprises diffraction peaks at one or more of 6.5 ± 0.2 °, 20.7 ± 0.2 °, 20.9 ± 0.2 °, 21.0 ± 0.2 °, 21.4 ± 0.2 °, 15.6 ± 0.2 °, 23.8 ± 0.2 °, 21.6 ± 0.2 °, 9.2 ± 0.2 °, 25.1 ± 0.2 °, 10.7 ± 0.2 °, 12.4 ± 0.2 °, 17.4 ± 0.2 °, 17.6 ± 0.2 °, 14.5 ± 0.2 ° of 2 Θ; preferably, the compound comprises diffraction peaks at 4, 5, 6, 8 and 10;
most preferably, form B has an X-ray powder diffraction pattern as shown in figure 4;
the X-ray powder diffraction pattern of the crystal form C has a diffraction peak at the 2 theta of 7.1 +/-0.2 degrees; or a diffraction peak at 18.1 ± 0.2 °; or a diffraction peak at 9.0 ± 0.2 °; or a diffraction peak at 22.7 ± 0.2 °; or a diffraction peak at 14.1 ± 0.2 °; or a diffraction peak at 18.9 ± 0.2 °; or a diffraction peak at 22.5 ± 0.2 °; or a diffraction peak at 20.3 ± 0.2 °; or a diffraction peak at 22.2 ± 0.2 °; or a diffraction peak at 23.2 ± 0.2 °; preferably comprises any 2-5, or 3-6, or 3-8, or 5-8, or 6-8 of the diffraction peaks, more preferably comprises any 6, 7 or 8 thereof;
preferably, form C has an X-ray powder diffraction pattern comprising at least one, preferably two, more preferably three, diffraction peaks, at 2 Θ of 7.1 ± 0.2 °, 18.1 ± 0.2 °, 9.0 ± 0.2 °; optionally, at least one of 22.7 ± 0.2 °, 14.1 ± 0.2 °, 18.9 ± 0.2 °, 22.5 ± 0.2 °, and 20.3 ± 0.2 ° may be further included; preferably 2, 3, 4 or 5 thereof;
more preferably, the X-ray powder diffraction pattern of form C optionally further comprises one or more diffraction peaks at 22.2 ± 0.2 °, 23.2 ± 0.2 °, 23.1 ± 0.2 °, 20.7 ± 0.2 °, 22.0 ± 0.2 °, 17.5 ± 0.2 °, 12.9 ± 0.2 ° 2 Θ; preferably at least any 2-3, or 4-5, or 6-7 thereof; further preferably, any 2, 3, 4, 5, 6, 7 thereof;
further preferably, the X-ray powder diffraction pattern of form C comprises one or more diffraction peaks located at 7.1 ± 0.2 °, 18.1 ± 0.2 °, 9.0 ± 0.2 °, 22.7 ± 0.2 °, 14.1 ± 0.2 °, 18.9 ± 0.2 °, 22.5 ± 0.2 °, 20.3 ± 0.2 °, 22.2 ± 0.2 °, 23.2 ± 0.2 °, 23.1 ± 0.2 °, 20.7 ± 0.2 °, 22.0 ± 0.2 °, 17.5 ± 0.2 °, 12.9 ± 0.2 ° of 2 Θ; preferably, the compound comprises diffraction peaks at 4, 5, 6, 8 or 10 optionally;
most preferably, form C has an X-ray powder diffraction pattern as shown in figure 7;
the X-ray powder diffraction pattern of the crystal form D has a diffraction peak at the 2 theta of 26.2 +/-0.2 degrees; or a diffraction peak at 22.2 ± 0.2 °; or a diffraction peak at 10.6 ± 0.2 °; or a diffraction peak at 14.5 ± 0.2 °; or a diffraction peak at 21.3 ± 0.2 °; or a diffraction peak at 15.9 ± 0.2 °; or a diffraction peak at 7.0 ± 0.2 °; or a diffraction peak at 9.0 ± 0.2 °; or a diffraction peak at 19.0 ± 0.2 °; or a diffraction peak at 8.3 ± 0.2 °; preferably comprises any 2-5, or 3-6, or 3-8, or 5-8, or 6-8 of the diffraction peaks, more preferably comprises any 6, 7 or 8 thereof;
preferably, form D has an X-ray powder diffraction pattern comprising at least one, preferably two, more preferably three, diffraction peaks, located at one or more of 26.2 ± 0.2 °, 22.2 ± 0.2 °, 10.6 ± 0.2 ° 2 Θ; optionally, at least one of 14.5 ± 0.2 °, 21.3 ± 0.2 °, 15.9 ± 0.2 °, 7.0 ± 0.2 °, and 9.0 ± 0.2 ° may be further included; preferably 2, 3, 4 or 5 thereof;
more preferably, the X-ray powder diffraction pattern of form D optionally further comprises one or more diffraction peaks at 19.0 ± 0.2 °, 8.3 ± 0.2 °, 26.9 ± 0.2 °, 18.1 ± 0.2 °, 20.1 ± 0.2 °, 21.8 ± 0.2 °, 19.8 ± 0.2 ° 2 Θ; preferably at least any 2-3, or 4-5, or 6-7 thereof; further preferably, any 2, 3, 4, 5, 6, 7 thereof;
further preferably, the X-ray powder diffraction pattern of form D comprises one or more diffraction peaks located at 26.2 ± 0.2 °, 22.2 ± 0.2 °, 10.6 ± 0.2 °, 14.5 ± 0.2 °, 21.3 ± 0.2 °, 15.9 ± 0.2 °, 7.0 ± 0.2 °, 9.0 ± 0.2 °, 19.0 ± 0.2 °, 8.3 ± 0.2 °, 26.9 ± 0.2 °, 18.1 ± 0.2 °, 20.1 ± 0.2 °, 21.8 ± 0.2 °, 19.8 ± 0.2 ° 2 Θ; preferably, the compound comprises diffraction peaks at 4, 5, 6, 8 or 10 optionally;
most preferably, form D has an X-ray powder diffraction pattern as shown in figure 10;
the X-ray powder diffraction pattern of the crystal form E has a diffraction peak at the 2 theta of 6.3 +/-0.2 degrees; or a diffraction peak at 12.6 ± 0.2 °; or a diffraction peak at 9.9 ± 0.2 °; or a diffraction peak at 21.1 ± 0.2 °; or a diffraction peak at 13.2 ± 0.2 °; or a diffraction peak at 26.2 ± 0.2 °; or a diffraction peak at 20.0 ± 0.2 °; or a diffraction peak at 16.9 ± 0.2 °; or a diffraction peak at 15.5 ± 0.2 °; or a diffraction peak at 25.3 ± 0.2 °; preferably comprises any of 2 to 5, or 3 to 6, or 3 to 8, or 5 to 8, or 6 to 8 of the diffraction peaks; more preferably any 6, 7 or 8 thereof;
preferably, form E has an X-ray powder diffraction pattern comprising at least one, preferably two, more preferably three, diffraction peaks, located at one or more of 6.3 ± 0.2 °, 12.6 ± 0.2 °, 9.9 ± 0.2 ° 2 Θ; optionally, at least one of 21.1 ± 0.2 °, 13.2 ± 0.2 °, 26.2 ± 0.2 °, 20.0 ± 0.2 °, and 16.9 ± 0.2 ° may be further included; preferably 2, 3, 4 or 5 thereof;
more preferably, the X-ray powder diffraction pattern of form E optionally further comprises one or more diffraction peaks at 15.5 ± 0.2 °, 25.3 ± 0.2 °, 27.4 ± 0.2 ° 2 Θ; preferably at least any 2 to 3 thereof; further preferably, any 2 or 3 thereof;
further preferably, the X-ray powder diffraction pattern of form E comprises one or more diffraction peaks located at 6.3 ± 0.2 °, 12.6 ± 0.2 °, 9.9 ± 0.2 °, 21.1 ± 0.2 °, 13.2 ± 0.2 °, 26.2 ± 0.2 °, 20.0 ± 0.2 °, 16.9 ± 0.2 °, 15.5 ± 0.2 °, 25.3 ± 0.2 °, 27.4 ± 0.2 ° of 2 Θ; preferably, the compound comprises diffraction peaks at 4, 5, 6, 8 or 10 optionally;
most preferably, form E has an X-ray powder diffraction pattern as shown in figure 13.
4. A crystalline form of the compound according to claim 3, characterized in that the diffraction peak positions with first ten strong relative peak intensities in the X-ray powder diffraction patterns of form a, form B, form C, form D, form E have diffraction peaks with 2 θ errors of ± 0.2 ° to ± 0.5 °, preferably ± 0.2 ° to ± 0.3 °, most preferably ± 0.2 ° from the corresponding positions of fig. 1, fig. 4, fig. 7, fig. 10 and fig. 13, respectively.
5. A crystalline form of the compound of claim 3, characterized in that form a has a DSC profile as shown in figure 2; or a TGA profile as shown in figure 3; the form B has a DSC pattern as shown in figure 5; or a TGA profile as shown in figure 6; the form C has a DSC pattern as shown in figure 8; or a TGA profile as shown in figure 9; the form D has a DSC pattern as shown in figure 11; or a TGA profile as shown in figure 12; the form E has a DSC profile as shown in figure 14; or a TGA profile as shown in figure 15.
6. A process for preparing a crystalline form of the compound according to any one of claims 1 to 5, comprising in particular the steps of:
1) weighing a proper amount of free alkali, and suspending with a poor solvent;
2) shaking the suspension obtained in the step 1) for a certain time at a certain temperature;
3) quickly centrifuging the suspension obtained in the step 2), removing supernatant, and drying the residual solid to constant weight to obtain a target product;
wherein:
the poor solvent is selected from one or more of methanol, acetone, ethyl acetate, tetrahydrofuran, acetonitrile, ethanol, 88% acetone, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, methyl tert-butyl ether, N-heptane, benzene, toluene, chlorobenzene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, dimethyl sulfoxide, N-propanol, ethyl formate, isopropyl acetate, tert-butanol, 2-butanone and 3-pentanone; one or more of toluene, 1, 4-dioxane, 3-pentanone, ethyl acetate, methanol and ethyl formate are preferred.
7. A process for preparing a crystalline form of the compound according to any one of claims 1 to 5, comprising in particular the steps of:
1) weighing a proper amount of free alkali, and dissolving the free alkali in a good solvent;
2) adding an anti-solvent into the obtained solution, and stirring until a solid is separated out;
3) quickly centrifuging, removing supernatant, and drying the residual solid to constant weight;
wherein:
the benign solvent is selected from one or more of methanol, acetone, ethyl acetate, tetrahydrofuran, acetonitrile, ethanol, 88% acetone, 2-methyl-tetrahydrofuran, dichloromethane, 1, 4-dioxane, benzene, toluene, chlorobenzene, isopropanol, N-butanol, isobutanol, N-dimethylformamide, N-dimethylacetamide, dimethyl sulfoxide, N-propanol, ethyl formate, isopropyl acetate, tert-butanol, 2-butanone and 3-pentanone; preferably one or more of dichloromethane, tetrahydrofuran, 2-methyl-tetrahydrofuran, 1, 4-dioxane, dimethyl sulfoxide, acetonitrile and 2-butanone;
the poor solvent is selected from one or more of methanol, ethanol, ethyl acetate, acetone, isopropanol, toluene, n-heptane, water, isopropyl acetate, cyclohexane, methyl tert-butyl ether and isopropyl ether; one or more of water, n-heptane, cyclohexane and methyl tert-butyl ether are preferred.
8. A pharmaceutical composition comprising a therapeutically effective amount of a crystalline form of a compound of any one of claims 1-5, and one or more pharmaceutically acceptable carriers or excipients.
9. Use of a crystalline form of a compound according to any one of claims 1-5, and a pharmaceutical composition according to claim 8, for the manufacture of a medicament for a kinase inhibitor; preferably, the kinase inhibitor is a receptor tyrosine kinase inhibitor, more preferably a HER2 inhibitor, an EGFR inhibitor and an EGFR monoclonal antibody and their combination related drugs, and further preferably a HER 220 exon mutant inhibitor, an EGFR20 exon mutant inhibitor and an EGFR20 exon mutant monoclonal antibody and their combination related drugs.
10. Use of a crystalline form of a compound according to any one of claims 1-5, and a pharmaceutical composition according to claim 8 for the manufacture of a medicament for the treatment of cancer, inflammation, chronic liver disease, diabetes, cardiovascular disease and AIDS related disease, preferably, the cancer, inflammation, chronic liver disease, diabetes, cardiovascular disease and AIDS related disease is a disease mediated by HER 220 exon mutation and/or EGFR20 exon mutation; more preferably, the cancer is selected from the group consisting of breast cancer, cervical cancer, colon cancer, lung cancer, stomach cancer, rectal cancer, pancreatic cancer, brain cancer, liver cancer, solid tumors, gliomas, glioblastoma, leukemia, lymphoma, myeloma, and non-small cell lung cancer.
CN202111346076.6A 2020-11-19 2021-11-15 Crystal form containing indole derivative free base and preparation method and application thereof Pending CN114539226A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011307267 2020-11-19
CN2020113072677 2020-11-19

Publications (1)

Publication Number Publication Date
CN114539226A true CN114539226A (en) 2022-05-27

Family

ID=81668874

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111346076.6A Pending CN114539226A (en) 2020-11-19 2021-11-15 Crystal form containing indole derivative free base and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN114539226A (en)

Similar Documents

Publication Publication Date Title
JP6691281B2 (en) EGFR inhibitors, their manufacture and applications
JP6431593B2 (en) Pyridinylaminopyrimidine derivative, method for producing the same, and use thereof
DK2651918T3 (en) Crystalline forms of 5-chloro-N2- (2-isopropoxy-5-methyl-4-piperidin-4-yl-phenyl) -N4- [2- (propane-2-sulfonyl) -phenyl] -pyrimidine-2,4 diamine
CN113773304B (en) Preparation method of anti-drug-resistance anti-tumor EGFR inhibitor
CA2561406C (en) Heterocyclic compound and anti-malignant-tumor agent comprising the same as effective component
CN112552294B (en) Piperazine heterocyclic derivative-containing inhibitor, preparation method and application thereof
KR20100016432A (en) 2-morpholin-4-yl-pyrimidines as pi3k inhibitors
KR20200115583A (en) 2H-indazole derivatives as CDK4 and CDK6 inhibitors and their therapeutic use
US20200361908A1 (en) Crystals of aniline pyrimidine compound serving as egfr inhibitor
CN110698461A (en) Preparation method of third-generation EGFR inhibitor
CN111606889B (en) Process for the preparation of 4- (1-cyclopropyl-1H-indol-3-yl) -N-phenylpyrimidin-2-amine derivatives
CN111825658A (en) Novel EGFR (epidermal growth factor receptor) triple-mutation inhibitor and application thereof
CN114539226A (en) Crystal form containing indole derivative free base and preparation method and application thereof
CN116528859A (en) Crystal form containing free alkali of di-fused ring derivative inhibitor and preparation method and application thereof
CN111303128A (en) Polycyclic compound, preparation method and application thereof
WO2022105882A1 (en) Salt and crystal form of indole-containing derivative, and preparation methods therefor and applications thereof
CN110256429A (en) A kind of amino-metadiazine compound and its preparation method and application with spirane structure
CN115803326B (en) EGFR inhibitor, preparation method thereof and application thereof in pharmacy
WO2022161447A1 (en) Dicarboxamide compound, preparation method therefor, and pharmaceutical use 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