WO2017206924A1 - 抑制蛋白激酶活性化合物的晶型及其应用 - Google Patents

抑制蛋白激酶活性化合物的晶型及其应用 Download PDF

Info

Publication number
WO2017206924A1
WO2017206924A1 PCT/CN2017/086760 CN2017086760W WO2017206924A1 WO 2017206924 A1 WO2017206924 A1 WO 2017206924A1 CN 2017086760 W CN2017086760 W CN 2017086760W WO 2017206924 A1 WO2017206924 A1 WO 2017206924A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
disease
crystal form
crystalline form
Prior art date
Application number
PCT/CN2017/086760
Other languages
English (en)
French (fr)
Inventor
梁从新
马勇斌
何伟
Original Assignee
贝达药业股份有限公司
艾科睿控股公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to MYPI2018002245A priority Critical patent/MY188379A/en
Priority to EP17805869.9A priority patent/EP3470410B1/en
Priority to KR1020187038142A priority patent/KR102466958B1/ko
Priority to CA3026142A priority patent/CA3026142C/en
Priority to AU2017274110A priority patent/AU2017274110B2/en
Priority to EA201892687A priority patent/EA201892687A1/ru
Priority to BR112018074612-6A priority patent/BR112018074612A2/pt
Priority to US16/306,104 priority patent/US10899744B2/en
Application filed by 贝达药业股份有限公司, 艾科睿控股公司 filed Critical 贝达药业股份有限公司
Priority to SG11201810800VA priority patent/SG11201810800VA/en
Priority to RU2018145183A priority patent/RU2744264C2/ru
Priority to IL263356A priority patent/IL263356B/en
Priority to CN201780006852.3A priority patent/CN109195964B/zh
Priority to JP2018563619A priority patent/JP7054528B2/ja
Publication of WO2017206924A1 publication Critical patent/WO2017206924A1/zh
Priority to PH12018502529A priority patent/PH12018502529A1/en
Priority to ZA2018/08625A priority patent/ZA201808625B/en
Priority to HK19101605.4A priority patent/HK1259249A1/zh

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
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2059Starch, including chemically or physically modified derivatives; Amylose; Amylopectin; Dextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C403/00Derivatives of cyclohexane or of a cyclohexene or of cyclohexadiene, having a side-chain containing an acyclic unsaturated part of at least four carbon atoms, this part being directly attached to the cyclohexane or cyclohexene or cyclohexadiene rings, e.g. vitamin A, beta-carotene, beta-ionone
    • C07C403/06Derivatives of cyclohexane or of a cyclohexene or of cyclohexadiene, having a side-chain containing an acyclic unsaturated part of at least four carbon atoms, this part being directly attached to the cyclohexane or cyclohexene or cyclohexadiene rings, e.g. vitamin A, beta-carotene, beta-ionone having side-chains substituted by singly-bound oxygen atoms
    • C07C403/10Derivatives of cyclohexane or of a cyclohexene or of cyclohexadiene, having a side-chain containing an acyclic unsaturated part of at least four carbon atoms, this part being directly attached to the cyclohexane or cyclohexene or cyclohexadiene rings, e.g. vitamin A, beta-carotene, beta-ionone having side-chains substituted by singly-bound oxygen atoms by etherified hydroxy groups
    • 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
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic 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

Definitions

  • the present invention relates to a novel compound ⁇ 5-[(1R)-1-(2,6-dichloro-3-fluorophenyl)ethoxy]-6-aminopyridazin-3-yl ⁇ -N- ⁇ a new crystalline form of 4-[((3S,5R)-3,5-dimethylpiperazinyl)carbonyl]phenyl ⁇ carboxamide hydrochloride and its hydrate or solvate; the invention also relates to said compound And crystalline intermediate preparation methods and related intermediate compounds, pharmaceutical compositions containing the compounds, and their use in inhibiting the activity of protein kinase (PK).
  • the invention also relates to a method of treating a disease, disorder or condition associated with modulation of a protein kinase using a compound or a crystalline form and a pharmaceutical composition of at least one of the above.
  • Protein kinases are enzymes that catalyze the phosphorylation of proteins. In most cases, this phosphorylation occurs at the serine (ser), threonine (thr) and tyrosine (tyr) residues of the protein. Many aspects of cell life, such as cell growth, differentiation, proliferation, cell cycle, and survival, are dependent on the activity of protein kinases. Moreover, abnormal protein kinase activity is associated with many disorders such as cancer and inflammation.
  • protein kinases there are more than 500 protein kinases currently discovered. According to the types of amino acid residues whose substrate proteins are phosphorylated, they can be divided into five categories, namely: 1 serine/threonine (Ser/Thr) protein kinase: protein The hydroxyl group is phosphorylated; 2 tyrosine (Tyr) protein kinase: the phenolic hydroxyl group of the protein acts as a phosphorus acceptor; 3 histidine protein kinase: the basic group of the histidine, arginine or lysine of the protein Phosphorylated; 4 tryptophan protein kinase: a tryptophan residue of a protein as a phosphorus acceptor; 5 aspartyl/glutamyl protein kinase: an acyl group of a protein as a phosphorus acceptor.
  • Ser/Thr serine/threonine
  • Tyr tyrosine
  • PTK Protein tyrosine kinase
  • RTKs receptor tyrosine kinases
  • the MET proto-oncogene subfamily of the RTKs family has two MET and RON (receptor d' ridge nantais) member.
  • the C-Met proto-oncogene encodes a Met receptor tyrosine kinase.
  • the Met receptor is a 190 kDa glycosylated dimeric complex composed of a 50 kDa alpha chain linked to a beta strand of 145 by a disulfide bond. The alpha chain is found extracellularly, while the beta chain includes the transmembrane and cytoplasmic domains.
  • Met plays a role in tumorigenesis and tumor metastasis, Met transforms, tumorigenesis and metastasis with the expression of its ligand hepatocyte growth factor (HGF) (Jefferson, M. et al, Oncogene 1996, 13, 853-856; Michieli , P.
  • HGF hepatocyte growth factor
  • C-Met is overexpressed in a significant percentage of human cancers and expands during the transition between primary tumor and metastasis. Many studies have correlated the expression of C-Met and/or HGF/SF with the disease progression status of different types of cancer. Furthermore, overexpression of C-Met or HGF has been shown to be associated with poor prognosis and disease outcome in a variety of major human cancers including lung, liver, stomach and breast cancer. C-Met is also directly involved in cancers that have no successful treatment regimen, such as pancreatic cancer, glioma, and hepatocellular carcinoma.
  • the homologous genes of RON include Stk (murine) and Sea (chicken), and its ligand is macrophage stimulating protein (MSP), which is a serum protein and homologous to HGF.
  • MSP macrophage stimulating protein
  • the RON gene is located on human chromosome 3p21 and contains 20 exons and 19 introns.
  • the mature RON protein is a heterodimer composed of alpha and beta subunits with a molecular weight of approximately 185 kDa.
  • the RON gene product can be detected in a variety of normal human tissues.
  • RON is expressed in human epithelial cells, granulocytes, mononuclear macrophages, megakaryocytes, osteoclasts, and tonsil germinal layers, small intestine, colon, kidney, lung, and bone marrow. Recent studies have shown that in many human primary tumor and tumor cell lines including the digestive system, urinary system, lung and breast, the expression of RON changes significantly in quality and quantity.
  • the carcinogenic activity of RON is related to the activity of kinase.
  • the activity of RON kinase is significantly up-regulated by overexpression, mutation and cleavage mechanism, which leads to malignant transformation, growth and movement of cells. RON can also cause tumor invasion and metastasis alone or in combination with other factors (Journal of International Pathology and Clinical Medicine, 2005, 25(5): 441-443).
  • CSF1R colony stimulating factor 1 receptor
  • C-fms colony stimulating factor 1 receptor
  • Ig immunoglobulin motifs.
  • CSF1R is mainly expressed on cells of the monocyte cell line as well as in the cells of the female genital tract and placenta. It has also been found to be expressed in Langerhans cells, subgroups of smooth muscle cells, B cells and microglia in the skin. The major biological effects of CSF1R signaling are the differentiation, proliferation, migration and survival of precursor macrophages and osteoclasts derived from monocyte lines.
  • Axl belongs to a subfamily of receptor tyrosine kinases that also include Tyro 3 and Mer.
  • Axl overexpression has been reported in a variety of human cancers and is associated with infection and metastasis in lung, prostate, breast, gastric, renal cell carcinoma and glioblastoma.
  • Recent studies have shown that Axl overexpression via a "tyrosine kinase switch" causes imatinib resistance in gastrointestinal stromal tumors.
  • Axl expression is triggered by chemotherapeutic drugs, and overexpression of Axl causes acute myeloid leukemia The drug resistance, which suggests that Axl may be involved in the regulation of multiple aspects of tumorigenesis (Oncogene, 2009, 28: 3442).
  • EphA2 belongs to the largest subfamily EPH RTKs of receptor tyrosine kinases. Studies have shown that EphA2 is associated with a range of pathological conditions, including tumors (Pasquale EB. Eph receptors and ephrins in cancer: bidirectional signaling and beyond. Nat Rev Cancer 2010 ;10:165-80). Recently, studies have shown that EphA2 blockade can overcome the acquired resistance of EGFR kinase inhibitors in lung cancer (Amato et al. Cancer Res 2016; 76(2); 305-18).
  • ROS1 is a member of the insulin receptor family. Recently, ROS1 rearrangement was found to be present in a small number of lung cancer patients; and crizotinib, as an inhibitor of ROS1, is very effective in treating such patients (Bergethon et al. J. Clin. Oncol. 2012; 30 ( 8), 863).
  • NTRK1 rearrangements with tumorigenicity and drug sensitivity have also been found in lung cancer (Vaishnavil et al. Nature Medicine 2013; 19(11), 1469).
  • the NTRK1 gene encodes a high affinity nerve growth factor receptor (TRKA) protein.
  • Anaplastic lymphoma kinase belongs to the RTKs superfamily. Due to the ectopic t2 chromosome, the oncogenic constitutively active ALK fusion protein is expressed in anaplastic large cell lymphoma (ALCL) and inflammatory myofibroblastic tumor (IMT). ALK has recently been recognized as a proto-oncogene in a small proportion of non-small cell lung cancer and neuroblastoma (Choi et al, Cancer Res 2008; 68: (13); Webb et al, Expert Rev. Anticancer Ther. 2009; (3), 331-356).
  • ALK ATI alternative transcription initiation
  • ALK is also involved in neurological diseases, and results show that ALK acts in the adult brain to regulate the function of the frontal cortex and hippocampus and to identify ALK as a sign of psychosis (such as schizophrenia, depression, and substance (***e) addiction) Target.
  • Crizotinib has been reported to be a potent inhibitor of HGF receptor tyrosine kinase (C-Met) and ALK (WO2004076412; WO2006021881; WO2006021886).
  • crizotinib In Phase I clinical trials of crizotinib, 64% achieved an objective response (ORR) and 90% achieved disease control (J Clin Oncology 2010; 28:7S, Suppl; abstr3). Unfortunately, the violent response to crizotinib is only transient. Most patients develop resistance and disease progression after 6-18 months of treatment. In particular, a significant proportion of patients developed brain metastases that crizotinib could not treat.
  • the present invention also relates to various substantially pure crystalline forms of the compounds of the above formula I, hydrates thereof and/or solvates thereof.
  • the crystalline form of the compound of the formula I, the hydrate thereof and/or its solvate is present in one or more crystal forms.
  • the present invention first provides a crystalline form of a compound of the formula I, a hydrate thereof and/or a solvate thereof, having a diffraction angle 2 ⁇ of 4.9 ⁇ 0.2°, 10.0 ⁇ 0.2° and Characteristic peak of 19.3 ⁇ 0.2 °.
  • the invention is referred to as Form A.
  • the present invention further provides a preferred embodiment of the above Form A:
  • the X-ray powder diffraction pattern of the above Form A has characteristics of diffraction angles 2 ⁇ of 4.9 ⁇ 0.2°, 10.0 ⁇ 0.2°, 14.7 ⁇ 0.2°, 16.9 ⁇ 0.2°, 19.3 ⁇ 0.2°, and 20.3 ⁇ 0.2°. peak.
  • the X-ray powder diffraction pattern of the crystal form A has diffraction angles 2 ⁇ of 4.9 ⁇ 0.2°, 10.0 ⁇ 0.2°, 14.7 ⁇ 0.2°, 16.9 ⁇ 0.2°, 19.3 ⁇ 0.2°, 20.3 ⁇ 0.2°, 25.5. Characteristic peaks of ⁇ 0.2° and 30.7 ⁇ 0.2°.
  • the above Form A has an X-ray powder diffraction pattern as shown in Figure 1.
  • the present invention summarizes the X-ray powder diffraction pattern of Form A as shown in Table 1.
  • the Form A purity is ⁇ 85%.
  • the Form A purity is > 95%.
  • the Form A purity is > 99%.
  • the Form A purity is > 99.5%.
  • the crystalline form A is a dihydrate.
  • the present invention further provides another crystal form of the compound of the formula I, a hydrate thereof and/or a solvate thereof, which has a diffraction angle 2 ⁇ of 10.5 ⁇ 0.2° and 17.4 ⁇ 0.2 in the X-ray powder diffraction spectrum. ° and characteristic peaks of 21.1 ⁇ 0.2 °.
  • Form B the invention is referred to as Form B.
  • the crystal form B has an X-ray powder diffraction pattern having diffraction angles 2 ⁇ of 10.5 ⁇ 0.2°, 17.4 ⁇ 0.2°, 19.7 ⁇ 0.2°, 21.1 ⁇ 0.2°, 23.9 ⁇ 0.2°, and 25.5 ⁇ 0.2°. Characteristic peak.
  • the X-ray powder diffraction pattern of Form B has diffraction angles 2 ⁇ of 10.5 ⁇ 0.2°, 17.4 ⁇ 0.2°, 19.7 ⁇ 0.2°, 21.1 ⁇ 0.2°, 21.5 ⁇ 0.2°, 23.9 ⁇ 0.2°, 25.2. Characteristic peaks of ⁇ 0.2° and 25.5 ⁇ 0.2°.
  • Form B has an X-ray powder diffraction pattern as shown in Figure 2.
  • the present invention summarizes the X-ray powder diffraction pattern of Form B, as shown in Table 2.
  • the Form B purity is ⁇ 85%.
  • the Form B is ⁇ 95% pure.
  • the Form B purity is > 99%.
  • the Form B purity is > 99.5%.
  • the crystalline form B is a trihydrate.
  • the present invention further provides another crystal form of the compound of the formula I, a hydrate thereof and/or a solvate thereof, which has a diffraction angle 2 ⁇ of 10.2 ⁇ 0.2° and 20.6 ⁇ 0.2 in the X-ray powder diffraction spectrum. ° and characteristic peaks of 21.8 ⁇ 0.2 °.
  • Form C the invention is referred to as Form C.
  • the present invention further provides a preferred embodiment of the above Form C:
  • the X-ray powder diffraction pattern of the above Form C has diffraction angles 2 ⁇ of 10.2 ⁇ 0.2°, 14.7 ⁇ 0.2°, 19.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 21.8 ⁇ 0.2°, and 24.5 ⁇ 0.2°. Characteristic peaks.
  • the X-ray powder diffraction pattern of the above-mentioned Form C has diffraction angles 2 ⁇ of 8.7 ⁇ 0.2°, 10.2 ⁇ 0.2°, 14.7 ⁇ 0.2°, 19.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 21.8 ⁇ 0.2°, Characteristic peaks of 24.5 ⁇ 0.2 ° and 25.9 ⁇ 0.2 °.
  • the above crystalline form C has an X-ray powder diffraction pattern as shown in Fig. 3.
  • the present invention summarizes the X-ray powder diffraction pattern of Form C as shown in Table 3.
  • the Form C purity is > 85%.
  • the Form C purity is > 95%.
  • the Form C purity is > 99%.
  • the Form C purity is > 99.5%.
  • the crystal form C is a methanol solvent compound.
  • the present invention further provides another crystal form of the compound of the formula I, a hydrate thereof and/or a solvate thereof having a diffraction angle 2 ⁇ of 9.2 ⁇ 0.2° and 18.0 ⁇ 0.2 in the X-ray powder diffraction pattern. ° and characteristic peaks of 18.5 ⁇ 0.2 °.
  • Form D the invention is referred to as Form D.
  • the invention further provides a preferred embodiment of the above crystal form D:
  • the X-ray diffraction spectrum of the above-mentioned Form D has characteristics of diffraction angles 2 ⁇ of 4.5 ⁇ 0.2°, 9.2 ⁇ 0.2°, 18.0 ⁇ 0.2°, 18.5 ⁇ 0.2°, 19.5 ⁇ 0.2°, and 20.1 ⁇ 0.2°. peak.
  • the X-ray diffraction spectrum of the above-mentioned crystal form D has diffraction angles 2 ⁇ of 4.5 ⁇ 0.2°, 9.2 ⁇ 0.2°, 18.0 ⁇ 0.2°, 18.5 ⁇ 0.2°, 19.5 ⁇ 0.2°, 20.1 ⁇ 0.2°, 22.3. Characteristic peaks of ⁇ 0.2° and 23.1 ⁇ 0.2°.
  • the above crystal form D has an X-ray powder diffraction spectrum as shown in Fig. 4 .
  • the present invention summarizes the X-ray powder diffraction pattern of Form D, as shown in Table 4.
  • the crystal form D has a purity of ⁇ 85%.
  • the Form D purity is > 95%.
  • the Form D purity is > 99%.
  • the Form D purity is > 99.5%.
  • the crystalline form D is a dimethyl sulfoxide solvent compound.
  • the present invention further provides another crystal form of the compound of the formula I, a hydrate thereof and/or a solvate thereof, which has a diffraction angle 2 ⁇ of 4.8 ⁇ 0.2° and 9.6 ⁇ 0.2 in the X-ray powder diffraction pattern. ° and characteristic peaks of 25.8 ⁇ 0.2 °.
  • Form E the invention is referred to as Form E.
  • the present invention further provides a preferred embodiment of the above Form E:
  • the X-ray diffraction spectrum of the above Form E has characteristics of diffraction angles 2 ⁇ of 4.8 ⁇ 0.2°, 9.6 ⁇ 0.2°, 16.3 ⁇ 0.2°, 18.1 ⁇ 0.2°, 20.8 ⁇ 0.2°, and 25.8 ⁇ 0.2°. peak.
  • the X-ray diffraction spectrum of the above-mentioned crystal form D has diffraction angles 2 ⁇ of 4.8 ⁇ 0.2°, 9.6 ⁇ 0.2°, 16.3 ⁇ 0.2°, 18.1 ⁇ 0.2°, 19.3 ⁇ 0.2°, 20.8 ⁇ 0.2°, 25.8. Characteristic peaks of ⁇ 0.2° and 26.7 ⁇ 0.2°.
  • the above crystal form E has an X-ray powder diffraction spectrum as shown in Fig. 5.
  • the present invention summarizes the X-ray powder diffraction pattern of Form E, as shown in Table 5.
  • the Form E purity is > 85%.
  • the Form E purity is > 95%.
  • the Form E purity is > 99%.
  • the Form E purity is > 99.5%.
  • the crystalline form E is a dihydrate.
  • the invention further provides an amorphous form of the compound of formula I, a hydrate thereof and/or a solvate thereof, having an X-ray powder diffraction pattern as shown in Figure 6.
  • the invention still further provides a pharmaceutical composition comprising a therapeutically effective amount of Form A and/or Form B above.
  • the pharmaceutical composition contains a therapeutically effective amount of Form A or Form B provided herein, and a pharmaceutically acceptable adjuvant, adjuvant or carrier.
  • the pharmaceutical composition contains a therapeutically effective amount of Form A and Form B provided by the present invention, and a pharmaceutically acceptable adjuvant, adjuvant or carrier.
  • the pharmaceutical composition contains a therapeutically effective amount of Form A or Form B of the present invention, used in parallel Less one other active ingredient.
  • the pharmaceutical composition contains a therapeutically effective amount of Form A and Form B of the present invention in parallel with at least one other active ingredient.
  • the pharmaceutical composition is an oral preparation.
  • the pharmaceutical composition is a tablet or capsule.
  • the pharmaceutical composition comprises from 20 to 150 mg of Form A and/or Form B, formulated with a total amount of from about 50 mg to 500 mg with at least one adjuvant, adjuvant and/or carrier.
  • the adjuvant, adjuvant and/or carrier in the pharmaceutical composition is microcrystalline cellulose, mannitol, crospovidone, croscarmellose sodium, sodium carboxymethyl starch, Povidone, hydroxypropyl cellulose and/or stearic acid.
  • the pharmaceutical composition contains 0.01% by weight to 99% by weight of Form A or Form B of the present invention.
  • the pharmaceutical composition contains 0.01% by weight to 99% by weight of Form A and Form B of the present invention.
  • the pharmaceutical composition contains 0.1% by weight to 70% by weight of Form A or Form B of the present invention.
  • the pharmaceutical composition contains 0.1% by weight to 70% by weight of Form A and Form B of the present invention.
  • the pharmaceutical composition contains from 1% to 70% by weight of Form A or Form B of the present invention.
  • the pharmaceutical composition contains from 1% by weight to 70% by weight of Form A and Form B of the present invention.
  • the pharmaceutical composition contains from 1% to 50% by weight of Form A or Form B of the present invention.
  • the pharmaceutical composition contains from 1% by weight to 50% by weight of Form A and Form B of the present invention.
  • the pharmaceutical composition contains from 1% by weight to 30% by weight of Form A or Form B of the present invention.
  • the pharmaceutical composition contains from 1% by weight to 30% by weight of Form A and Form B of the present invention.
  • the pharmaceutical composition contains 10% by weight to 30% by weight of Form A or Form B of the present invention.
  • the pharmaceutical composition contains 10% by weight to 30% by weight of Form A and Form B of the present invention.
  • the invention also provides the use of Form A and/or Form B in the manufacture of a medicament for treating a disease, disorder or condition in a patient, wherein the disease, disorder or condition is c-Met, RON, Axl, CSF1R, EphA2, ROS1 or ROS1 fusion protein, TRKA or TRKA fusion protein, TRKB, TRKC, ALK, ALK ATI or ALK fusion protein.
  • the invention also provides a preferred embodiment of the above uses of Form A and/or Form B:
  • the ALK fusion protein is EML4-ALK or NPM-ALK kinase.
  • the disease, condition or condition is a cancer and/or a proliferative disease.
  • the disease, disorder or condition is lung cancer, melanoma, colon cancer, breast cancer, liver cancer, pancreatic cancer, brain cancer, kidney cancer, ovarian cancer, gastric cancer, skin cancer, bone cancer, glioma, Lymphoma, neuroblastoma, Hepatocellular carcinoma, papillary renal cell carcinoma, and/or head and neck squamous cell carcinoma.
  • the disease, condition or condition is non-small cell lung cancer (NSCLC) that is resistant to treatment with crizotinib.
  • NSCLC non-small cell lung cancer
  • the disease, condition or condition is melanoma.
  • the disease, condition or condition is a neurological disease, a psychiatric disease, an obesity, a diabetes and/or a cardiovascular disease.
  • the psychiatric disorder is schizophrenia, depression and/or substance addiction or abuse.
  • the substance addiction or abuse is addiction or abuse of ***e, tobacco or alcohol.
  • the invention also provides methods of treating a disease, disorder, or condition in a patient by administering to the patient a Form A and/or Form B provided herein.
  • the invention further provides a preferred embodiment of the above method of treating a disease, disorder or condition in a patient using Form A and/or Form B:
  • the disease, disorder or condition is a fusion protein of c-Met, RON, Axl, CSF1R, EphA2, ROS1 or ROS1, TRKA or TRKA fusion protein, TRKB, TRKC, ALK, ALK ATI or ALK fusion protein. guide.
  • the ALK fusion protein is EML4-ALK or NPM-ALK kinase.
  • the disease, condition or condition is a cancer and/or a proliferative disease
  • the disease, disorder or condition is lung cancer, melanoma, colon cancer, breast cancer, liver cancer, pancreatic cancer, brain cancer, kidney cancer, ovarian cancer, gastric cancer, skin cancer, bone cancer, glioma, Lymphoma, neuroblastoma, hepatocellular carcinoma, papillary renal cell carcinoma, and/or head and neck squamous cell carcinoma.
  • the disease, condition or condition is non-small cell lung cancer (NSCLC) that is resistant to treatment with crizotinib.
  • NSCLC non-small cell lung cancer
  • the disease, condition or condition is melanoma.
  • the disease, condition or condition is a neurological disease, a psychiatric disease, an obesity, a diabetes and/or a cardiovascular disease.
  • the psychiatric disorder is schizophrenia, depression and/or substance addiction or abuse.
  • the substance addiction or abuse is addiction or abuse of ***e, tobacco or alcohol.
  • the invention further provides a process for the preparation of a compound of formula I, as follows:
  • the invention further provides intermediates in the preparation of the compounds of formula I, as follows:
  • the present invention further provides a process for the preparation of Form A and Form B of the compound of Formula I, its hydrate and/or its solvate.
  • the preparation method of the crystal form A is as follows:
  • amorphous sample of the compound of the formula I is placed in a centrifuge tube, and allowed to stand in an atmosphere of ethanol or acetonitrile in a sealed solvent atmosphere for 6-10 days to obtain the crystal form A; or
  • the preparation method of the crystal form B is as follows:
  • Form A added to methanol, ethanol or water, dissolved into a clear solution, filtered, and then evaporated to dryness at room temperature (20 ° C) -40 ° C to obtain the crystal form B; or,
  • Form A added to methanol / water, methanol / acetone, methanol / ethyl acetate, methanol / methyl tert-butyl ether, methanol / tetrahydrofuran, methanol / dichloromethane, ethanol / water, ethanol / butanone, ethanol / Isopropyl acetate, ethanol/n-heptane, trifluoroethanol/water, trifluoroethanol/ethyl acetate, trifluoroethanol/tetrahydrofuran, water/methanol, water/ethanol, water/trifluoroethanol, water/isopropanol , water / acetone, water / tetrahydrofuran or water / acetonitrile, dissolved into a clear solution, filtered, placed at room temperature (20 ° C) -40 ° C open volatilization, to obtain the crystal form B; or,
  • Form A add to a lower alcohol, water, nitromethane, methyl ethyl ketone, ether, ethyl acetate, tetrahydrofuran, toluene or n-heptane to form a suspension, placed at room temperature -40 ° C, stirred 4 to 5 Day, the suspension is centrifuged to obtain the crystal form B; or,
  • Form A add water-saturated ethyl acetate layer, ethyl acetate-saturated aqueous layer, ethanol / diethyl ether, toluene / acetonitrile, butanone / ethanol or toluene / isopropyl ether to form a suspension, placed at 4 ° C Stirring at -40 ° C for 4-5 days, centrifuging, to obtain the crystal form B; or,
  • Form A add methanol, acetone / water (3:1v / v) or acetonitrile / water (3:2v / v) solvent at room temperature, dissolve into a clear solution, add hydroxypropyl cellulose, ethyl fiber After the compound, povidone K30, polyallylamine hydrochloride, carboxymethyl cellulose or polyvinyl alcohol, it is exposed to room temperature and volatilized to obtain the crystal form B; or
  • Form A at 60 ° C -70 ° C, add a lower alcohol or water, dissolve into a clear solution, filter, and stir at 4 ° C to precipitate the crystal form, to obtain the crystal form B; or,
  • Form A dissolved in acetone / trifluoroethanol, acetone / water, dioxane / water, acetonitrile / water or methyl tert-butyl ether / n-propanol at 55 ° C -70 ° C, dissolved into a clear solution , filtering, the filtrate is stirred at -20 ° C until the precipitated crystal form, and filtered to obtain the crystal form B; or
  • Form A at 60 ° C -70 ° C, add nitromethane / methanol, acetonitrile / methanol, methyl ethyl ketone / ethanol, ethyl acetate / ethanol, 1,4-dioxane / ethanol or tetrahydrofuran / water, Dissolving into a clear solution, filtering, and volatilizing at room temperature to obtain the crystal form B; or
  • Crystal form A add methanol, ethanol, water, trifluoroethanol, n-propanol or dimethyl sulfoxide at room temperature, dissolve into a clear solution, filter, add acetone and ethyl acetate to the filtrate with stirring. , methyl tert-butyl ether, diisopropyl ether, B Isopropyl acrylate, tetrahydrofuran, 1,4-dioxane, acetonitrile, n-heptane, dichloromethane or chloroform, to precipitate a large amount of crystal form, and filtered to obtain the crystal form B; or
  • amorphous sample of the compound of formula I is placed in a centrifuge tube which is then placed in n-butanol, water, nitromethane, ethyl acetate, methyl tert-butyl ether, tetrahydrofuran, dichloromethane , the chloroform or toluene is diffused in an atmosphere to obtain the crystal form B; or
  • the term "substantially pure” means that at least 85% by weight, preferably at least 95% by weight, more preferably at least 99% by weight, most preferably at least 99.5% by weight of the compound of formula I is present in the crystal of the present invention. In the form, especially in Form A and/or Form B.
  • Fig. 1 having an X-ray powder diffraction pattern as shown in Fig. 1
  • Fig. 2 having an X-ray powder diffraction pattern as shown in Fig. 2
  • the peak is shown in Fig. 1 or Fig. 2, wherein the main peak means that the relative intensity value exceeds 10%, preferably exceeds 30%, compared with the highest peak in Fig. 1 or Fig. 2 (the relative intensity is specified as 100%). Those peaks.
  • the "addition of methanol / acetone" or the like involved in the preparation method of the crystal form A or the form B means that the preparation is in the preparation
  • methanol is first added, followed by acetone.
  • ethanol/water means that ethanol is added first, followed by water;
  • trifluoroethanol/ethyl acetate means that trifluoroethanol is added first, followed by ethyl acetate.
  • solvent 1 / solvent 2 means that solvent 1 is added first, and then solvent 2 is added; and “solvent 2 / solvent 1” means that solvent 2 is added first, and then solvent 1 is added.
  • the term "therapeutically effective amount” means a compound which, when administered to a subject, is sufficient to affect a disease, disorder or condition for treating a disease, or at least one clinical condition of a disease or condition.
  • the amount of treatment can be a compound, a disease, a condition, and/or a symptom of a disease or condition, a disease, a condition, and/or a severity of a symptom of the disease or condition, the age of the patient being treated, and/or treated.
  • the patient's weight changes and so on. In any particular case, a suitable amount will be apparent to those skilled in the art, and may be determined by routine experimentation.
  • “therapeutically effective amount” refers to the total amount of a combination of subjects effective to treat a disease, disorder or condition.
  • compositions of the invention can be prepared by conventional methods in the pharmaceutical arts.
  • the active ingredient is combined with one or more excipients and then prepared into the desired dosage form.
  • “Pharmaceutically acceptable carrier” means a conventional pharmaceutical carrier suitable for the desired pharmaceutical formulation, for example, a diluent such as water, various organic solvents, or the like, an excipient; a filler such as starch, sucrose, or the like; a cellulose derivative, an alginate, a binder of gelatin and polyvinylpyrrolidone (PVP); a wetting agent such as glycerin; a disintegrant such as agar, calcium carbonate and sodium hydrogencarbonate; an absorption enhancer such as a quaternary ammonium compound; Surfactants such as cetyl alcohol; absorbent carriers such as kaolin and bentonite; lubricants such as talc, calcium stearate, magnesium stearate, and polyethylene glycol.
  • a diluent such as water, various organic solvents, or the like, an excipient
  • a filler such as starch, sucrose, or the like
  • PVP polyvinylpyrrol
  • disease refers to any disease, discomfort, disease, symptom or indication.
  • Figure 1 X-ray powder diffraction pattern of Form A of the compound of Formula I.
  • Figure 2 X-ray powder diffraction pattern of Form B of the compound of Formula I.
  • Figure 3 X-ray powder diffraction pattern of Form C of the compound of Formula I.
  • Figure 4 X-ray powder diffraction pattern of Form D of the compound of Formula I.
  • Figure 5 X-ray powder diffraction pattern of Form E of the compound of Formula I.
  • Figure 6 X-ray powder diffraction pattern of an amorphous form of the compound of formula I.
  • Figure 8 Dynamic moisture adsorption pattern of Form B of the compound of Structural Formula I.
  • Figure 10 Differential thermal analysis scan of Form A of the compound of Formula I.
  • Figure 11 Differential thermal analysis scan of Form B of the compound of Structural Formula I.
  • Figure 12 Differential thermal analysis scan of an amorphous form of a compound of formula I.
  • Figs. 1-6 The apparatus and method for detecting X-ray powder diffraction patterns shown in Figs. 1-6 are shown in Table 6.
  • DSC Differential Thermal Analysis Scanner
  • Boc tert-butoxycarbonyl
  • DCC dicyclohexylcarbodiimide
  • DIPEA diisopropylethylamine
  • EDCI 1-ethyl-(3-dimethylaminopropyl) carbonyldiimide hydrochloride
  • HATU 2-(7-azobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • NMM N-methylmorpholine
  • TFA trifluoroacetic acid
  • the organic phase was added with 20 mL of 1N diluted hydrochloric acid, stirred for 5 minutes, and left to stand for separation; the organic layer was washed with 20 mL of diluted hydrochloric acid, 6 mL of 1N KOH aqueous solution, and 35 mL of saturated brine. The organic layer was dried over anhydrous sodium sulfate, filtered and evaporated. The resulting residue was washed with EtOAc (EtOAc)EtOAc. The filter cake was dried to give 10.62 g of Compound M3 as a tan solid.
  • Form A Take about 5-10 mg of Form A, add the corresponding solvent, dissolve into a clear solution, filter, place at the corresponding temperature, and open to volatilize to obtain Form B.
  • Form A About 10 mg of Form A is taken, and after adding solvent 1, the solvent 2 is added, dissolved into a clear solution, filtered, and volatilized at the corresponding temperature to obtain Form B.
  • Form A About 15-30 mg of Form A is taken, a corresponding solvent is added to form a suspension, stirred at room temperature for 5 days, and filtered to obtain Form B.
  • Form A About 15-20 mg of Form A is taken, and a corresponding solvent is added to form a suspension, which is placed at 40 ° C, stirred for 5 days, and filtered to obtain Form B.
  • Form A About 15 to 40 mg of Form A is taken, and after adding solvent 2, solvent 1 is further added to form a suspension, which is stirred at the corresponding temperature for 4 days, and filtered to obtain Form B.
  • Form A About 15 to 50 mg of Form A is taken, and the corresponding solvent is added at the corresponding temperature, dissolved into a clear solution, and then filtered, and stirred at 4 ° C to precipitate a solid, which is filtered to obtain Form B.
  • isopropanol did not precipitate when stirred at 4 ° C, and then volatilized at room temperature to obtain crystal form B.
  • Form B preparation method 69-74 is stirred without precipitation, and then volatilized at room temperature to obtain Form B.
  • Form A Take about 10-15mg of Form A, add solvent 1 at room temperature, dissolve into a clear solution by ultrasonication, filter, stir The solvent 2 was added dropwise to the solution under stirring, and stirred to precipitate a large amount of solid, which was filtered to obtain a crystal form B.
  • Form B preparation methods 89 and 90 were stirred without precipitation, and then volatilized at room temperature to obtain Form B.
  • Form A A sample of Form A of 100 mg of the compound of Formula I was taken, 2.0 mL of ethyl acetate was added, and then 1.0 mL of dimethyl sulfoxide was added to form a suspension, which was stirred at room temperature for 1 day to obtain Form D.
  • Form A A sample of Form A of about 5 mg of the compound of the formula I was taken, dissolved in 0.1 mL of dimethyl sulfoxide, filtered, and opened to volatilize at a temperature of 40 ° C to obtain Form D.
  • Form A A sample of Form A of about 20 mg of the compound of Formula I was taken and placed in an 85% RH humidifier at room temperature for 26 days to obtain Form E.
  • Amorphous preparation method 1 is amorphous preparation method 1:
  • Amorphous preparation method 2 is amorphous preparation method 2:
  • Amorphous preparation method 3-5 is amorphous preparation method 3-5.
  • a sample of crystal form A of about 5 to 10 mg of the compound of the formula I is taken, dissolved in a corresponding solvent, filtered, and volatilized at a corresponding temperature to obtain an amorphous substance.
  • Amorphous preparation method 6-9 is amorphous preparation method 6-9:
  • a sample of Form A of about 10 mg of the compound of the formula I is taken, dissolved in a corresponding solvent, filtered, and volatilized at a corresponding temperature to obtain an amorphous substance.
  • Amorphous preparation method 10 is amorphous preparation method 10:
  • Amorphous preparation methods 11 and 12 are Amorphous preparation methods 11 and 12:
  • Amorphous preparation method 13 is amorphous preparation method 13:
  • a sample of Form A of about 10 to 15 mg of the compound of the formula I is taken, dissolved in a corresponding solvent, filtered, and rapidly spun at the corresponding temperature to obtain an amorphous substance.
  • the Form A and Form B samples were dried at 80 ° C for 24 hours, 25 ° C - 60% RH for 10 days, and 40 ° C - 75% RH for 10 days, and the crystal forms were unchanged.
  • Form C was almost completely converted to Form A after overnight drying under vacuum at room temperature.
  • Form D was air-dried at room temperature or vacuum-dried at room temperature overnight and most of it was converted to Form B, which was not stable.
  • Form E was placed in a desiccator for most of the day and turned into Form A, which was unstable.
  • Example 9 Dynamic moisture adsorption (DVS) determination
  • Form A The weight change in the range of 0%-80% RH is about 2.3%, the hydrate removes about 1.5% of water in the 0% RH drying stage, and the moisture absorption in the range of 0% RH-80% RH is about 2.3%. %, 1.5% moisture can be removed at 30% RH in the desorption stage, and the weight change is less than 2% in the range of 30-80% RH.
  • Form B The weight change in the range of 0%-80% RH is about 2.3%, and the hydrate can be removed below 10% RH. 4% moisture, in the adsorption phase can absorb about 4% moisture at 10% RH, and the weight change is less than 2% in the range of 10-80% RH.
  • Amorphous The weight change in the range of 0-80% RH is about 15.7%, which is extremely hygroscopic.
  • the solubility of the crystalline form A of the compound of the formula I in water at room temperature is 20-100 mg/mL, and the solubility of the free base amorphous form of the compound of structure I in water is ⁇ 1 mg/mL.
  • an oral drug about 20 to 150 mg of the polymorph described in Example 1 and/or 2 is prepared, and the total amount of microcrystalline cellulose and/or stearic acid is sufficiently finely divided to obtain a total amount of about 50 mg to 500mg to fill the type 0 capsule.
  • an oral drug about 20 to 150 mg of the polymorph described in Example 1 and/or 2 is used, and microcrystalline cellulose, mannitol, and the like are sufficiently finely divided by two or more of the following excipients.
  • a combination of vidosterone, croscarmellose sodium, sodium carboxymethyl starch, povidone, hydroxypropylcellulose and/or stearic acid yields a total amount of tablets or capsules of from about 50 mg to about 500 mg.
  • Rats were orally administered 50 mg/kg of the compound A in Form I and the free base amorph of Compound I shown in Structure I, with Tmax of 2.67 and 4.67h, respectively, Cmax of 1440 and 890 ng/mL, respectively, AUC last respectively It is 11551 and 8165h*ng/mL.
  • the crystalline form A of the compound represented by the structure I is better absorbed in the body than the free base amorphous form of the compound represented by the structure I.
  • the biochemical kinase activity assay of the compound of formula I is tested by Reaction Biology Corp., Malvern, PA, USA, and the assay protocol is described by Anastassiadis et al. Nat Biotechnol. 2011; (11): 1039-45.
  • the compound of formula I was found to potentially inhibit the following kinases:

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Diabetes (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Cardiology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

提供了结构式I所示化合物({5-[(1R)-1-(2,6-二氯-3-氟苯基)乙氧基]-6-氨基哒嗪-3-基}-N-{4-[((3S,5R)-3,5-二甲基哌嗪基)羰基]苯基}甲酰胺盐酸盐)及其水合物或溶剂化物的新晶型;还提供了所述化合物和晶型的制备方法以及相关的中间体化合物,含有所述化合物的药物组合物,以及所述化合物或晶型在制备治疗疾病、病症或病状的药物中的应用或用于治疗疾病、病症或病状的治疗方法。

Description

抑制蛋白激酶活性化合物的晶型及其应用 技术领域
本发明涉及一种新化合物{5-[(1R)-1-(2,6-二氯-3-氟苯基)乙氧基]-6-氨基哒嗪-3-基}-N-{4-[((3S,5R)-3,5-二甲基哌嗪基)羰基]苯基}甲酰胺盐酸盐及其水合物或溶剂化物新的晶型;本发明还涉及所述化合物和晶型的制备方法以及相关的中间体化合物,含有所述化合物的药物组合物,以及它们在抑制蛋白激酶(protein kinase,PK)的活性中的应用。本发明也涉及应用至少上述之一的化合物或晶型及药物组合物在治疗与蛋白激酶调节相关的疾病、病症或病状的方法。
背景技术
蛋白激酶是催化蛋白质磷酸化反应的酶,在大多数情况下,这一磷酸化反应是发生在蛋白质的丝氨酸(ser)、苏氨酸(thr)和酪氨酸(tyr)残基上。细胞生命的很多方面(例如细胞生长、分化、增殖、细胞周期和存活)均依赖于蛋白激酶的活性。而且,异常的蛋白激酶活性与许多的紊乱例如癌症和炎症相关。
目前发现的蛋白激酶约有500多种,根据其底物蛋白被磷酸化的氨基酸残基种类,可将他们分为5类,即:①丝氨酸/苏氨酸(Ser/Thr)蛋白激酶:蛋白质的羟基被磷酸化;②酪氨酸(Tyr)蛋白激酶:蛋白质的酚羟基作为磷受体;③组氨酸蛋白激酶:蛋白质的组氨酸、精氨酸或赖氨酸的碱性基团被磷酸化;④色氨酸蛋白激酶:以蛋白质的色氨酸残基作为磷受体;⑤天冬氨酰基/谷氨酰基蛋白激酶:以蛋白质的酰基作为磷受体。
酪氨酸蛋白激酶(protein tyrosine kinase,PTK),目前发现有100多个家族成员,在调节细胞的分化、生长和激活中起重要作用。根据PTK的结构,可分为受体型和非受体型PTK两大类,前者又称跨膜PTK,后者又称细胞内PTK。
人类肿瘤的发生、发展依赖于一系列癌基因的激活和抑癌基因的失活。在对上皮源性肿瘤的研究中发现,受体酪氨酸激酶(Receptor tyrosine kinase,RTKs)的跨膜蛋白质在调节细胞生长、分化和生存中起着基础性作用,并在肿瘤的发生发展中起重要作用。
RTKs家族中MET原癌基因亚家族有MET和RON(receptor d′ origine nantais)2个成 员。
C-Met原癌基因编码Met受体酪氨酸激酶。Met受体是190KDa的糖基化二聚复合物,由50KDa的α链以双硫键连接到145的β链而构成。α链在胞外发现,而β链包括跨膜和胞质域。Met在肿瘤发生和肿瘤转移中起作用,Met随其配体肝细胞生长因子(HGF)的表达而转化、致瘤和转移(Jefferson,M.等人,Oncogene 1996,13,853-856;Michieli,P.等人,Oncogene 1999,18,5221-5231)。C-Met在显著百分比的人类癌症中过表达并在原发肿瘤和转移之间的转变过程中扩大。很多研究已经将C-Met和/或HGF/SF的表达与不同种类癌症的疾病进展状态进行关联。此外,C-Met或HGF的过表达已经显示出与多种主要人类癌症(包括肺癌、肝癌、胃癌和乳腺癌)中的预后不佳和疾病转归相关联。C-Met还直接牵涉到没有成功治疗方案的癌症中,例如胰腺癌、神经胶质瘤和肝细胞癌。
RON的同源基因包括Stk(鼠)和Sea(鸡),其配体是巨噬细胞刺激蛋白(macrophage stimulating protein,MSP),为一血清蛋白质,与HGF同源。RON基因位于人类染色体3p21,含有20个外显子和19个内含子。成熟的RON蛋白是由α和β亚单位组成的杂二聚体,分子量约为185KDa。在多种正常人体组织中可以检测到RON基因产物。RON在人体上皮细胞、粒细胞、单核巨噬细胞、巨核细胞、破骨细胞以及扁桃体生发层、小肠、结肠、肾脏、肺、骨髓的细胞中均有表达。近年来研究表明,许多包括消化***、泌尿***、肺和乳腺等处的人类原发性肿瘤与肿瘤细胞系中,RON的表达在质和量上显著改变。RON的致癌活性与激酶的活性相关,通过过表达、突变和剪切机制可使RON激酶活性显著上调,从而引起细胞的恶性转化、生长与运动。RON同时也能单独或与其他因子合作引起肿瘤的侵袭和转移(国际病理科学与临床杂志,2005,25(5):441-443)。
CSF1R(colony stimulating factor 1 receptor,巨噬细胞集落刺激因子受体),也称为C-fms,是一种单链跨膜受体酪氨酸激酶,是含有免疫球蛋白(Ig)基序的RTKs家族的成员。CSF1R主要表达在单核细胞系的细胞上以及雌性生殖道和胎盘的细胞中。另外还发现表达在皮肤的郎格汉(Langerhans)细胞、平滑肌细胞的亚组、B细胞和小神经胶质细胞中。CSF1R信号转导的主要生物学效应是源自单核细胞系的前体巨噬细胞和破骨细胞的分化、增殖、迁移和存活。
Axl属于同样包括Tyro 3和Mer的受体酪氨酸激酶的亚族。Axl过表达已经在多种人类癌症中有过报道,且与肺癌、***癌、乳腺癌、胃癌、肾细胞癌和成胶质细胞瘤中的侵染和转移有关。近来有研究显示,经由“酪氨酸激酶开关”的Axl过表达在胃肠道间质瘤中引起伊马替尼抗性。Axl表达通过化疗药物而引发,且Axl的过表达造成急性髓性白血病中 的抗药性,这些表明,Axl可能参与肿瘤发生的多个方面的调控(Oncogene,2009,28:3442)。
EphA2属于受体酪氨酸激酶最大的亚族EPH RTKs,研究表明EphA2与一系列病理状况的调节有关联,包括肿瘤(Pasquale EB.Eph receptors and ephrins in cancer:bidirectional signaling and beyond.Nat Rev Cancer 2010;10:165-80)。近来,研究表明EphA2阻滞可以克服肺癌中EGFR激酶抑制剂的获得性抵抗(Amato et al.Cancer Res 2016;76(2);305-18)。
ROS1是胰岛素受体家族中的一员。最近,ROS1重排被发现存在于少量的肺癌患者中;而克唑替尼,作为ROS1的抑制剂,治疗这类患者是非常有效的(Bergethon et al.J.Clin.Oncol.2012;30(8),863)。
具有致瘤性和药物敏感性的NTRK1重排也被发现存在于肺癌中(Vaishnavil et al.Nature Medicine 2013;19(11),1469)。NTRK1基因编码高亲和性神经生长因子受体(TRKA)蛋白。
间变性淋巴瘤激酶(ALK)属于RTKs超家族。由于t2染色体异位,致癌的组成型活性的ALK融合蛋白在间变性大细胞淋巴瘤(ALCL)和炎性肌纤维母细胞瘤(IMT)中表达。ALK近来已被认为是小部分非小细胞肺癌和成神经细胞瘤中的原癌基因(Choi等人,Cancer Res 2008;68:(13);Webb等人,Expert Rev.Anticancer Ther.2009;9(3),331-356)。
近来,一种新的同种型ALK被发现表达于11%的黑色素瘤和偶发的其他人类癌症类型中,但是不在正常的组织中(Wiesner et al.Nature 2015;526,453)。这种新型的ALK转录物启动了位于ALK 19内含子的异常转录起始(alternative transcription initiation,ATI),称之为ALKATI
ALK还涉及神经***疾病中,有结果显示ALK在成人脑内作用于调控额叶皮质和海马的功能并将ALK确定为精神病迹象(例如精神***症、抑郁和物质(***)成瘾)的新目标物。
克唑替尼(crizotinib),已经被报道为HGF受体酪氨酸激酶(C-Met)和ALK的有效抑制剂(WO2004076412;WO2006021881;WO2006021886)。
Figure PCTCN2017086760-appb-000001
克唑替尼的I期临床试验中,64%达到客观应答(ORR),90%达到疾病控制(J Clin Oncology 2010;28:7S,Suppl;abstr3)。不幸的是,对克唑替尼的剧烈反应仅仅是短暂的。多数患者在治疗6-18个月后产生抗性和疾病进展。特别是,显著部分的患者产生克唑替尼无法治疗的脑转移。
之前的专利文本(WO2009/154769A1,WO2012/048259A2,CN103298806B)描述了取代的哒嗪羧酰胺化合物作为蛋白激酶抑制剂,这些化合物中的大部分有效地抑制c-Met和ALK,IC50为<100nM。因为在激酶介导疾病的治疗选择方面仍有未满足的需求,在此我们进一步筛选取代的哒嗪羧酰胺化合物的晶型以满足患者的医疗需求。
发明内容
本发明的目的在于提供一种如下结构式I的{5-[(1R)-1-(2,6-二氯-3-氟苯基)乙氧基]-6-氨基哒嗪-3-基}-N-{4-[((3S,5R)-3,5-二甲基哌嗪基)羰基]苯基}甲酰胺盐酸盐:
Figure PCTCN2017086760-appb-000002
本发明还涉及上述结构式I所示化合物、其水合物和/或其溶剂化物的多种基本上纯的晶型。
本发明中,结构式I所示化合物、其水合物和/或其溶剂化物的晶型,以一种或一种以上的晶型存在。
本发明首先提供了结构式I所示化合物、其水合物和/或其溶剂化物的一种晶型,该晶型X射线粉末衍射谱图具有衍射角2θ为4.9±0.2°、10.0±0.2°和19.3±0.2°的特征峰。为方便,本发明称之为晶型A。
其次,本发明进一步提供了上述晶型A的优选实施方式:
作为优选,上述晶型A的X射线粉末衍射谱图具有衍射角2θ为4.9±0.2°、10.0±0.2°、14.7±0.2°、16.9±0.2°、19.3±0.2°和20.3±0.2°的特征峰。
作为优选,该晶型A的X射线粉末衍射谱图具有衍射角2θ为4.9±0.2°、10.0±0.2°、14.7±0.2°、16.9±0.2°、19.3±0.2°、20.3±0.2°、25.5±0.2°和30.7±0.2°的特征峰。
作为优选,上述晶型A具有约如图1所示的X-射线粉末衍射图。
本发明总结了晶型A的X-射线粉末衍射图,如表1所示。
表1
2θ(°) 强度(I%)
4.9±0.2 13.7
10.0±0.2 100
14.7±0.2 17.7
16.9±0.2 20.3
19.3±0.2 63
20.3±0.2 22.4
25.5±0.2 10.8
30.7±0.2 14.9
作为优选,该晶型A纯度≥85%。
作为优选,该晶型A纯度≥95%。
作为优选,该晶型A纯度≥99%。
作为优选,该晶型A纯度≥99.5%。
作为优选,该晶型A为二水合物。
本发明进一步提供了结构式I所示化合物、其水合物和/或其溶剂化物的另一种晶型,该晶型X射线粉末衍射谱图中具有衍射角2θ为10.5±0.2°、17.4±0.2°和21.1±0.2°的特征峰。为方便,本发明称之为晶型B。
本发明还提供了上述晶型B的优先实施方式:
作为优选,所述晶型B的X射线粉末衍射谱图中具有衍射角2θ为10.5±0.2°、17.4±0.2°、19.7±0.2°、21.1±0.2°、23.9±0.2°和25.5±0.2°的特征峰。
作为优选,晶型B的X射线粉末衍射谱图中具有衍射角2θ为10.5±0.2°、17.4±0.2°、19.7±0.2°、21.1±0.2°、21.5±0.2°、23.9±0.2°、25.2±0.2°和25.5±0.2°的特征峰。
作为优选,晶型B具有约如图2所示的X-射线粉末衍射图。
本发明总结了晶型B的X-射线粉末衍射图,如表2所示。
表2
2θ(°) 强度(I%)
10.5±0.2° 100
17.4±0.2° 56.1
19.7±0.2° 26.4
21.1±0.2° 33.4
21.5±0.2° 23.6
23.9±0.2° 26.6
25.2±0.2° 25.8
25.5±0.2° 28.8
作为优选,该晶型B纯度≥85%。
作为优选,该晶型B纯度≥95%。
作为优选,该晶型B纯度≥99%。
作为优选,该晶型B纯度≥99.5%。
作为优选,该晶型B为三水合物。
本发明进一步提供了结构式I所示化合物、其水合物和/或其溶剂化物的另一种晶型,该晶型X射线粉末衍射谱图中具有衍射角2θ为10.2±0.2°、20.6±0.2°和21.8±0.2°的特征峰。为方便,本发明称之为晶型C。
其次,本发明进一步提供了上述晶型C的优选实施方式:
作为优选,上述晶型C的X射线粉末衍射谱图中具有衍射角2θ为10.2±0.2°、14.7±0.2°、19.4±0.2°、20.6±0.2°、21.8±0.2°和24.5±0.2°的特征峰。
作为优选,上述晶型C的X射线粉末衍射谱图中具有衍射角2θ为8.7±0.2°、10.2±0.2°、14.7±0.2°、19.4±0.2°、20.6±0.2°、21.8±0.2°、24.5±0.2°和25.9±0.2°的特征峰。
作为优选,上述晶型C具有约如图3所示的X-射线粉末衍射图。
本发明总结了晶型C的X-射线粉末衍射图,如表3所示。
表3
2θ(°) 强度(I%)
8.7±0.2 23.1
10.2±0.2 100
14.7±0.2 26.1
19.4±0.2 29.4
20.6±0.2 46.9
21.8±0.2 33.9
24.5±0.2 23.4
25.9±0.2 23.2
作为优选,该晶型C纯度≥85%。
作为优选,该晶型C纯度≥95%。
作为优选,该晶型C纯度≥99%。
作为优选,该晶型C纯度≥99.5%。
作为优选,该晶型C为甲醇溶剂化合物。
本发明进一步提供了结构式I所示化合物、其水合物和/或其溶剂化物的另一种晶型,该晶型X射线粉末衍射谱图中具有衍射角2θ为9.2±0.2°、18.0±0.2°和18.5±0.2°的特征峰。为方便,本发明称之为晶型D。
其次,本发明进一步提供了上述晶型D的优选实施方式:
作为优选,上述晶型D的X射线衍射谱图中具有衍射角2θ为4.5±0.2°、9.2±0.2°、18.0±0.2°、18.5±0.2°、19.5±0.2°和20.1±0.2°的特征峰。
作为优选,上述晶型D的X射线衍射谱图中具有衍射角2θ为4.5±0.2°、9.2±0.2°、18.0±0.2°、18.5±0.2°、19.5±0.2°、20.1±0.2°、22.3±0.2°和23.1±0.2°的特征峰。
作为优选,上述晶型D具有约如图4所示的X射线粉末衍射谱图。
本发明总结了晶型D的X射线粉末衍射谱图,如表4所示。
表4
2θ(°) 强度(I%)
4.5±0.2 21.9
9.2±0.2 100
18.0±0.2 54.5
18.5±0.2 23.4
19.5±0.2 13.2
20.1±0.2 13.1
22.3±0.2 11.8
23.1±0.2 11.9
作为优选,该晶型D纯度≥85%。
作为优选,该晶型D纯度≥95%。
作为优选,该晶型D纯度≥99%。
作为优选,该晶型D纯度≥99.5%。
作为优选,该晶型D为二甲亚砜溶剂化合物。
本发明进一步提供了结构式I所示化合物、其水合物和/或其溶剂化物的另一种晶型,该晶型X射线粉末衍射谱图中具有衍射角2θ为4.8±0.2°、9.6±0.2°和25.8±0.2°的特征峰。为方便,本发明称之为晶型E。
其次,本发明进一步提供了上述晶型E的优选实施方式:
作为优选,上述晶型E的X射线衍射谱图中具有衍射角2θ为4.8±0.2°、9.6±0.2°、16.3±0.2°、18.1±0.2°、20.8±0.2°和25.8±0.2°的特征峰。
作为优选,上述晶型D的X射线衍射谱图中具有衍射角2θ为4.8±0.2°、9.6±0.2°、16.3±0.2°、18.1±0.2°、19.3±0.2°、20.8±0.2°、25.8±0.2°和26.7±0.2°的特征峰。
作为优选,上述晶型E具有约如图5所示的X射线粉末衍射谱图。
本发明总结了晶型E的X射线粉末衍射谱图,如表5所示。
表5
2θ(°) 强度(I%)
4.8±0.2 47.1
9.6±0.2 100
16.3±0.2 34.1
18.1±0.2 28.3
19.3±0.2 24.9
20.8±0.2 26.8
25.8±0.2 41.6
26.7±0.2 20.9
作为优选,该晶型E纯度≥85%。
作为优选,该晶型E纯度≥95%。
作为优选,该晶型E纯度≥99%。
作为优选,该晶型E纯度≥99.5%。
作为优选,该晶型E为二水合物。
本发明进一步提供了结构式I所示化合物、其水合物和/或其溶剂化物的无定型物,该无定型物具有约如图6所示的X-射线粉末衍射图。
本发明还进一步提供了含有治疗有效量的上述晶型A和/或晶型B的药物组合物。
本发明还提供了上述药物组合物的优选实施方式:
作为优选,所述药物组合物含有治疗有效量的本发明提供的晶型A或晶型B,和药学上可接受的辅料、辅助剂或载体。
作为优选,所述药物组合物含有治疗有效量的本发明提供的晶型A和晶型B,和药学上可接受的辅料、辅助剂或载体。
作为优选,所述药物组合物,含有治疗有效量的本发明的晶型A或晶型B,并联用至 少一种其他有效成分。
作为优选,所述药物组合物,含有治疗有效量的本发明的晶型A和晶型B,并联用至少一种其他有效成分。
作为优选,所述药物组合物为口服制剂。
作为优选,所述药物组合物为片剂或胶囊。
作为优选,所述药物组合物中包含20至150mg晶型A和/或晶型B,用至少一种辅料、辅助剂和/或载体调配到总量约为50mg至500mg。
作为优选,所述药物组合物中的所述辅料、辅助剂和/或载体为微晶纤维素、甘露醇、交联聚维酮、交联羧甲基纤维素钠、羧甲基淀粉钠、聚维酮、羟丙基纤维素和/或硬脂酸。
作为优选,所述药物组合物含有0.01重量%~99重量%的本发明的晶型A或晶型B。
作为优选,所述药物组合物含有0.01重量%~99重量%的本发明的晶型A和晶型B。
作为优选,所述药物组合物含有0.1重量%~70重量%的本发明的晶型A或晶型B。
作为优选,所述药物组合物含有0.1重量%~70重量%的本发明的晶型A和晶型B。
作为优选,所述药物组合物含有1重量%~70重量%的本发明的晶型A或晶型B。
作为优选,所述药物组合物含有1重量%~70重量%的本发明的晶型A和晶型B。
作为优选,所述药物组合物含有1重量%~50重量%的本发明的晶型A或晶型B。
作为优选,所述药物组合物含有1重量%~50重量%的本发明的晶型A和晶型B。
作为优选,所述药物组合物含有1重量%~30重量%的本发明的晶型A或晶型B。
作为优选,所述药物组合物含有1重量%~30重量%的本发明的晶型A和晶型B。
作为优选,所述药物组合物含有10重量%~30重量%的本发明的晶型A或晶型B。
作为优选,所述药物组合物含有10重量%~30重量%的本发明的晶型A和晶型B。
本发明还提供了所述的晶型A和/或晶型B在制备治疗患者的疾病、病症或病状的药物中的用途,其中,所述的疾病、病症或病状由c-Met、RON、Axl、CSF1R、EphA2、ROS1或ROS1融合蛋白、TRKA或TRKA融合蛋白、TRKB、TRKC、ALK、ALKATI或ALK融合蛋白介导。
本发明还提供了所述晶型A和/或晶型B上述用途的优选实施方式:
作为优选,所述ALK融合蛋白是EML4-ALK或NPM-ALK激酶。
作为优选,所述疾病、病症或病状是癌症和/或增殖性疾病。
作为优选,所述的疾病、病症或病状是肺癌、黑色素瘤、结肠癌、乳腺癌、肝癌、胰腺癌、脑癌、肾癌、卵巢癌、胃癌、皮肤癌、骨癌、神经胶质瘤、淋巴瘤、成神经细胞瘤、 肝细胞癌、***状肾细胞癌和/或头颈部鳞状细胞癌。
作为优选,所述疾病、病症或病状是对克唑替尼(crizotinib)治疗有耐药性的非小细胞肺癌(NSCLC)。
作为优选,所述疾病、病症或病状是黑色素瘤。
作为优选,所述疾病、病症或病状是神经性疾病、精神性疾病、肥胖、糖尿病和/或心血管疾病。
作为优选,所述精神性疾病是精神***症、抑郁症和/或物质成瘾或滥用。
作为优选,所述物质成瘾或滥用是***、烟草或酒精的成瘾或滥用。
本发明还提供了通过对患者施用本发明提供的晶型A和/或晶型B治疗患者的疾病、病症或病状的方法。
本发明进一步提供了上述利用晶型A和/或晶型B治疗患者的疾病、病症或病状的方法的优选实施方式:
作为优选,所述的疾病、病症或病状是由c-Met、RON、Axl、CSF1R、EphA2、ROS1或ROS1融合蛋白、TRKA或TRKA融合蛋白、TRKB、TRKC、ALK、ALKATI或ALK融合蛋白介导。
作为优选,所述ALK融合蛋白是EML4-ALK或NPM-ALK激酶。
作为优选,所述的疾病、病症或病状是癌症和/或增殖性疾病
作为优选,所述的疾病、病症或病状是肺癌、黑色素瘤、结肠癌、乳腺癌、肝癌、胰腺癌、脑癌、肾癌、卵巢癌、胃癌、皮肤癌、骨癌、神经胶质瘤、淋巴瘤、成神经细胞瘤、肝细胞癌、***状肾细胞癌和/或头颈部鳞状细胞癌。
作为优选,所述疾病、病症或病状是对克唑替尼(crizotinib)治疗有耐药性的非小细胞肺癌(NSCLC)。
作为优选,所述疾病、病症或病状是黑色素瘤。
作为优选,所述疾病、病症或病状是神经性疾病、精神性疾病、肥胖、糖尿病和/或心血管疾病。
作为优选,所述精神性疾病是精神***症、抑郁症和/或物质成瘾或滥用。
作为优选,所述物质成瘾或滥用是***、烟草或酒精的成瘾或滥用。
本发明进一步提供了结构式I所示化合物的制备方法,如下:
方法一:
Figure PCTCN2017086760-appb-000003
方法二:
Figure PCTCN2017086760-appb-000004
方法三:
Figure PCTCN2017086760-appb-000005
本发明进一步提供了结构式I所示化合物制备过程中的中间体,如下:
本发明进一步提供了制备结构式I所示化合物、其水合物和/或其溶剂化物的晶型A和晶型B的方法。
其中,晶型A的制备方法如下:
结构式I所示化合物的无定型样品,置于离心管中,室温敞口置于乙醇或乙腈密闭溶剂气氛中静置扩散6-10天,制得该晶型A;或者,
结构式I所示化合物的无定型样品,加入乙醇溶剂后,置于4℃~25℃下,搅拌,过滤,制得该晶型A;或者,
4℃~25℃下,将结构式I所示化合物的无定型样品,加入到乙醇中,溶解成澄清溶液后过滤,在搅拌下向滤液内滴加正庚烷至析出大量晶型,过滤制得该晶型A;或者,
55℃~70℃下,将结构式I所示化合物的无定型样品,加入到甲基叔丁基醚/乙醇或正庚烷/乙醇中,溶解成澄清溶液后过滤,所得滤液置于-20℃,搅拌至析出固体,制得该晶型A;或者,
将结构式I所示化合物的无定型样品,加入到仲丁醇中,溶解成澄清溶液后过滤,置 于35℃-50℃下敞口挥发,制得该晶型A;或者,
将结构式I所示化合物的无定型样品,加入甲醇,溶解成澄清溶液,过滤,加入羧甲基纤维素,置于室温下敞口挥发,制得该晶型A。
其中,晶型B的制备方法如下:
将晶型A,加入甲醇、乙醇或水中,溶解成澄清溶液后过滤,置于室温(20℃)-40℃下敞口挥发干,制得该晶型B;或者,
将晶型A,加入甲醇/水、甲醇/丙酮、甲醇/乙酸乙酯、甲醇/甲基叔丁基醚、甲醇/四氢呋喃、甲醇/二氯甲烷、乙醇/水、乙醇/丁酮、乙醇/乙酸异丙酯、乙醇/正庚烷、三氟乙醇/水、三氟乙醇/乙酸乙酯、三氟乙醇/四氢呋喃、水/甲醇、水/乙醇、水/三氟乙醇、水/异丙醇、水/丙酮、水/四氢呋喃或水/乙腈,溶解成澄清溶液后过滤,置于室温(20℃)-40℃下敞口挥发干,制得该晶型B;或者,
将晶型A,加入低级醇、水、硝基甲烷、丁酮、***、乙酸乙酯、四氢呋喃、甲苯或正庚烷中,形成混悬液,置于室温-40℃下,搅拌4~5天,取混悬液离心,制得该晶型B;或者,
将晶型A,加入水饱和的乙酸乙酯层、乙酸乙酯饱和的水层、乙醇/***、甲苯/乙腈、丁酮/乙醇或甲苯/异丙醚,形成混悬液,置于4℃-40℃搅拌4-5天,离心,制得该晶型B;或者,
将晶型A,室温下加入甲醇、丙酮/水(3∶1v/v)或乙腈/水(3∶2v/v)溶剂中,溶解成澄清溶液后,添加羟丙基纤维素、乙基纤维素、聚维酮K30、聚烯丙基胺盐酸盐、羧甲基纤维素或聚乙烯醇后,置于室温下,敞口挥发,制得该晶型B;或者,
将晶型A,在60℃-70℃下,加入低级醇或水,溶解成澄清溶液,过滤,置于4℃搅拌至析出晶型,制得该晶型B;或者,
将晶型A,在55℃-70℃下,加入丙酮/三氟乙醇、丙酮/水、二恶烷/水、乙腈/水或甲基叔丁基醚/正丙醇中,溶解成澄清溶液,过滤,滤液置于-20℃搅拌至析出晶型,过滤,制得该晶型B;或者,
将晶型A,在60℃-70℃下,加入硝基甲烷/甲醇、乙腈/甲醇、丁酮/乙醇、乙酸乙酯/乙醇、1,4-二氧六环/乙醇或者四氢呋喃/水中,溶解成澄清溶液,过滤,室温下挥发,制得该晶型B;或者,
取晶型A,在室温下,加入甲醇、乙醇、水、三氟乙醇、正丙醇或二甲亚砜,溶解成澄清溶液后,过滤,在搅拌下向滤液内滴加丙酮、乙酸乙酯、甲基叔丁基醚、异丙醚、乙 酸异丙酯、四氢呋喃、1,4-二氧六环、乙腈、正庚烷、二氯甲烷或氯仿,至析出大量晶型,过滤,制得该晶型B;或者,
取晶型A,在室温下,加入甲醇或乙醇,溶解成澄清溶液后,过滤,在搅拌下向滤液内滴加二氯甲烷或四氢呋喃,室温挥发溶剂,制得该晶型B;或者,
取结构式I所示化合物的无定型样品,置于离心管中,然后将该离心管置于正丁醇、水、硝基甲烷、乙酸乙酯、甲基叔丁基醚、四氢呋喃、二氯甲烷、氯仿或甲苯气氛中扩散,制得该晶型B;或者,
取结构式I所示化合物的无定型样品,加入正丙醇、水、丁酮、乙酸乙酯、四氢呋喃、二氯甲烷、乙醇、异丙醇、正丁醇、丙酮、***、乙酸异丙酯、1,4-二氧六环、乙腈、氯仿、仲丁醇、硝基甲烷或甲苯中,然后置于4℃-40℃下,搅拌30分钟,过滤,制得该晶型B;或者,
取结构式I所示化合物的无定型样品,加入异丙醚/甲醇、乙酸乙酯/甲醇、1,4-二氧六环/甲醇、丁酮/乙醇、乙腈/乙醇、正庚烷/乙醇、硝基甲烷/三氟乙醇、***/三氟乙醇、四氢呋喃/三氟乙醇、丙酮/水、四氢呋喃/水、乙腈/水、甲基叔丁基醚/异丙醇、乙酸异丙酯/正丙醇、甲基环己烷/正丁醇、丙酮/二甲亚砜、乙酸乙酯/二甲亚砜、乙腈/二甲亚砜、甲基叔丁基醚/氯仿或者甲苯/乙酸乙酯,形成混悬液后,置于4℃-40℃下搅拌,过滤,制得该晶型B;或者,
取结构式I所示化合物的无定型样品,敞口静置于湿度为85%RH的室温下,静置10天,制得该晶型B;或者,
取晶型A,加入水或甲醇,溶解成澄清溶液,过滤,在40-60℃旋干,制得该晶型B。
本发明的所有晶型都是基本上纯的。
本发明所用的术语“基本上纯的”是指至少85重量%,优选至少95重量%,更优选至少99重量%,最优选至少99.5重量%的结构式I所示的化合物存在于本发明的晶型中,尤其是在晶型A和/或晶型B中。
上述的晶型仅仅概括了主要的峰。所述的主要的峰可再现并且在误差限度内(±0.2)。
本发明中,“具有约如图1所示的X-射线粉末衍射图”或“具有约如图2所示的X-射线粉末衍射图”,是指X-射线粉末衍射图示出的主要的峰如图1或图2所示,其中主要的峰是指与图1或图2中最高的峰(其相对强度指定为100%)相比,相对强度数值超过10%,优选超过30%的那些峰。
本发明中,晶型A或晶型B的制备方法中涉及的“加入甲醇/丙酮”等,是指在该制备 方法中,先加入甲醇,再加入丙酮。类似地,“乙醇/水”是指先加入乙醇,再加入水;而“三氟乙醇/乙酸乙酯”,是指先加入三氟乙醇,再加入乙酸乙酯。同理,类似地如“溶剂1/溶剂2”是指先加入溶剂1,再加入溶剂2;而“溶剂2/溶剂1”是指先加入溶剂2,再加入溶剂1。
本发明中,术语“治疗有效量”是指一个化合物施用于治疗对象时对于治疗一种疾病、或一种疾病或病症的至少一种临床症状时,足以影响对疾病、病症或症状的这种治疗的量。“治疗有效量”可以随着化合物,疾病、病症和/或疾病或病症的症状,疾病、病症和/或疾病或病症的症状的严重程度,被治疗的患者的年龄,和/或被治疗的患者的体重等变化。在任意特定的情况下,一个合适的量对那些本领域的技术人员可以是显而易见的,也可以是用常规实验确定的。在联合治疗的情况下,“治疗有效量”是指有效治疗疾病、病症或病状的联用对象的总量。
本发明的药物组合物的所有剂型都可以通过药学领域的常规方法制备。例如,将活性成分与一种或多种辅料混合,然后制成所需的剂型。
“药学上可接受的载体”是指适合于期望药物制剂的常规的药用载体,例如:诸如水、各种有机溶剂等的稀释剂、赋形剂;诸如淀粉、蔗糖等的填充剂;诸如纤维素衍生物、藻酸盐、明胶和聚乙烯吡咯烷酮(PVP)的黏合剂;诸如甘油的湿润剂;诸如琼脂、碳酸钙和碳酸氢钠的崩解剂;诸如季铵化合物的吸收促进剂;诸如十六烷醇的表面活性剂;诸如高岭土和膨润土的吸收载体;诸如滑石粉、硬脂酸钙、硬脂酸镁和聚乙二醇等的润滑剂。另外还可以在药物组合物中加入其它药学上可接受的辅料,如分散剂、稳定剂、增稠剂、络合剂、缓冲剂、渗透促进剂、聚合物、芳香剂、甜味剂和染料。优选使用适合期望剂型和期望给药方式的辅料。
术语“疾病”、“病症”或“病状”是指任意的疾病、不适、病、症状或者适应症。
附图说明
图1:结构式I所示化合物的晶型A的X-射线粉末衍射图谱。
图2:结构式I所示化合物的晶型B的X-射线粉末衍射图谱。
图3:结构式I所示化合物的晶型C的X-射线粉末衍射图谱。
图4:结构式I所示化合物的晶型D的X-射线粉末衍射图谱。
图5:结构式I所示化合物的晶型E的X-射线粉末衍射图谱。
图6:结构式I所示化合物的无定型物的X-射线粉末衍射图谱。
图7:结构式I所示化合物的晶型A的动态水分吸附图谱。
图8:结构式I所示化合物的晶型B的动态水分吸附图谱。
图9:结构式I所示化合物的无定型物的动态水分吸附图谱。
图10:结构式I所示化合物的晶型A的差热分析扫描图谱。
图11:结构式I所示化合物的晶型B的差热分析扫描图谱。
图12:结构式I所示化合物的无定型物的差热分析扫描图谱。
图1-6所示的X-射线粉末衍射图谱检测设备和方法如表6所示。
图7-9所示的动态水分吸附图谱检测设备和方法如表7所示。
图10-12所示的差热分析扫描图谱检测设备和方法如表8所示。
表6
Figure PCTCN2017086760-appb-000007
表7
Figure PCTCN2017086760-appb-000008
Figure PCTCN2017086760-appb-000009
表8
设备名称 差热分析扫描仪(DSC)
仪器 TA Instruments Q200DSC
控制软件 Thermal Adventage
分析软件 Universal Analysis
样品盘 铝坩埚(加盖打孔)
样品检测量 0.5-5mg
保护气体 氮气
气体流速 40mL/min
常用检测方法 Equilibrate at 20℃;Ramp 10℃/min to 250/300℃
具体实施方式
下面通过给出的实施例对本发明作出进一步说明,但所述实施例并不对本发明要求保护的范围构成任何限制。在本发明的具体实施例中,除非特别说明,所述技术或方法为本领域的常规技术或方法等。
缩略语:
Boc:叔丁氧羰基;
DCC:二环己基碳二亚胺;
DCM:二氯甲烷;
DIPEA:二异丙基乙基胺;
DMAP:4-二甲氨基吡啶;
DMF:N,N-二甲基甲酰胺;
EDCI:1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐;
HATU:2-(7-偶氮苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯;
HOBT:1-羟基苯并***;
IPA:异丙醇;
MeOH:甲醇;
MTBE:甲基叔丁基醚;
NMM:N-甲基吗啉;
N:mol/L;
TFA:三氟乙酸;
THF:四氢呋喃。
实施例1:结构式I所示化物的合成:
化合物M1的合成:
将8.57g(0.024mol,1.00eq)化合物SM1溶于85.7mL无水甲醇中,氮气保护下加入5%钯碳(0.86g),氢气置换三次,保持氢气气氛反应3小时。待原料反应完全,反应混合物过滤,滤液浓缩至固体,减压干燥,得类白色固体,即为化合物M1,收率为100%,纯度为97.32%。
LC-MS[M+H+]:334。
化合物M2的合成:
将9.60g(0.017mol,1.00eq)化合物SM2溶于53mL THF中,冷却反应液到-5~5℃,滴加1N KOH水溶液(1.40g KOH+25mL水),保温1-10℃,搅拌4小时。滴加1N稀盐酸调pH至5左右。分两次用乙酸乙酯(50mLx2)萃取,合并有机层,饱和食盐水洗涤有机层,无水硫酸钠干燥1小时,过滤,减压浓缩得粘稠油状物,加入23mL二氯甲烷溶解,再次减压浓缩得粘稠油状物。再次加入69mL二氯甲烷溶解,减压浓缩得中间体M2黄色固体9.10g,收率为99.7%。
LC-MS[M+H+]:546。
化合物M2的合成:
将9.60g(0.017mol,1.00eq)化合物SM2溶于53mL THF中,冷却反应液到0~10℃,滴加1N LiOH水溶液(1.05g LiOH+25mL水),30分钟滴加完毕后,逐渐升温至室温,搅拌反应过夜。反应完毕,减压浓缩。所得残余物加入100mL水溶解,向其中加入50mL甲基叔丁基醚,搅拌,分液。水相滴加1N稀盐酸调pH至5左右。分两次用乙酸乙酯(50mLx2) 萃取,合并有机层,饱和食盐水洗涤有机层,无水硫酸钠干燥1小时,过滤,减压浓缩得中间体M2黄色固体8.58g,收率为94%。
LC-MS[M+H+]:546。
化合物M3的合成:
将8.23g(0.015mol,1.15eq)化合物M2溶于66mL二氯甲烷,冷却至10-20℃,依次加入4.30g(0.013mol,1.00eq)化合物M1,3.00g(0.022mol,1.69eq)1-羟基苯并***(HOBT),4.58g(0.024mol,1.85eq)1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(EDCI)和6.90g(0.068mol,5.23eq)N-甲基吗啉(NMM)。20-30℃保温搅拌16小时,向反应液中加入16mL水,搅拌10-15分钟,静置分液,有机层加入16mL 1N稀盐酸,搅拌5分钟,静置分液;有机层分别经16mL 1N稀盐酸、5mL 1N KOH水溶液、30mL饱和食盐水洗涤,静置分液,有机相用无水硫酸钠干燥,过滤,减压浓缩。所得残余物分三次加入甲基叔丁基醚(20mL+20mL+6mL)洗涤,过滤,滤饼干燥,得7.95g类黄色固体,即为化合物M3,收率71.6%。
LC-MS[M+H+]:861。
化合物M3的合成:
将7.96g(0.015mol,1.15eq)化合物M2溶于40mLDMF,向其中加入5.23g(0.014mol,1.08eq)HATU,4.17g(0.013mol,1.00eq)化合物M1和2.17mL二异丙基乙基胺。室温搅拌反应过夜。向反应体系中加入50mL饱和碳酸钠水溶液,搅拌,过滤。固体用60mL水打浆,然后过滤。固体溶于30mL二氯甲烷,水洗两遍,有机相干燥,过滤,减压浓缩,干燥得10.03g化合物M3棕褐色固体,产率93.2%。
LC-MS[M+H+]:861。
化合物M3的合成:
将9.77g(0.018mol,1.20eq)化合物M2溶于75mL二氯甲烷,向其中加入6.21g(0.030mol,2.00eq)二环己基碳二亚胺,0.96g(0.0079mol,0.53eq)DMAP,5.00g(0.015mol,1.00eq)化合物M1。室温搅拌反应过夜。向反应体系中加入20mL水,搅拌,静置分液,有机相加入20mL 1N稀盐酸,搅拌5分钟,静置分液;有机层分别用20mL稀盐酸、6mL1N KOH水溶液、35mL饱和食盐水洗涤,静置分液,有机相用无水硫酸钠干燥,过滤,减压浓缩。所得残余物分三次加入甲基叔丁基醚(25mL+25mL+8mL)洗涤,过滤。滤饼干燥得10.62g化合物M3棕褐色固体,产率82.3%。
LC-MS[M+H+]:861。
结构式I所示化合物的合成:
将7.95g(0.092mol)化合物M3溶于30mL二氯甲烷中,冷却反应液至-5℃-5℃,滴加15mL三氟乙酸至反应液中,室温搅拌2小时,降温至10-20℃,缓慢加入60mL饱和碳酸钾水溶液,搅拌,静置分层,有机层用饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩。所得残余物加入41.2g异丙醇,搅拌溶解,降温至10-20℃,加入5mL浓盐酸,室温搅拌,过滤,滤饼用异丙醇洗涤,减压干燥,得到5.75g结构式I所示化合物。
LC-MS[M+H+]:561。
1H-NMR(300MHz,CDCl3):δ=1.25-1.43(m,6H),1.91(d,3H),3.15-3.48(m,4H),3.66-3.89(m,0.5H),4.55-4.78(m,0.5H),5.49(s,2H),6.26(q,1H),7.10(t,1H),7.33-7.44(m,4H),7.78(d,2H),9.93(s,1H)。
实施例2:晶型A的制备方法
晶型A制备方法1
取约5~10mg结构式I所示化合物的无定型样品,加入仲丁醇,溶解成澄清溶液后过滤,置于40℃下敞口挥发,制得晶型A。
晶型A制备方法2
取约10mg结构式I所示化合物的无定型样品,加入甲醇,溶解成澄清溶液后过滤,添加1mg羧甲基纤维素,置于室温下敞口挥发,制得晶型A。
晶型A制备方法3和4
取约10mg结构式I所示化合物的无定型样品,在相应温度下,加入溶剂2后再加入溶剂1后,溶解成澄清溶液后过滤,置于-20℃搅拌至析出固体,过滤,制得晶型A。
Figure PCTCN2017086760-appb-000010
晶型A制备方法5
取约10mg结构式I所示化合物的无定型样品,在室温下,加入乙醇,溶解成澄清溶液后过滤,在搅拌下向溶液内滴加正庚烷至析出大量固体,过滤,制得晶型A。
晶型A制备方法6和7
取约10mg结构式I所示化合物的无定型样品,置于2.0mL离心管中,室温敞口置 于相应密闭溶剂气氛中扩散6天,制得晶型A。
序号 样品量(mg) 溶剂 溶剂(体积,mL))
晶型A制备方法6 10 乙醇 4.0
晶型A制备方法7 10 乙腈 4.0
晶型A制备方法8
取约15mg结构式I所示化合物的无定型样品,于4℃下,加入乙醇0.2mL后,置于相应温度下搅拌30分钟,过滤,制得晶型A。
实施例3:晶型B的制备方法
第一种实验方式
取约5~10mg晶型A,加入相应溶剂,溶解成澄清溶液后过滤,置于相应温度下,敞口挥发,制得晶型B。
Figure PCTCN2017086760-appb-000011
第二种实验方式
取约10mg晶型A,加入溶剂1后,再加入溶剂2,溶解成澄清溶液后过滤,置于相应温度下敞口挥发,制得晶型B。
Figure PCTCN2017086760-appb-000012
Figure PCTCN2017086760-appb-000013
第三种实验方式
取约15~30mg晶型A,加入相应溶剂形成混悬液,置于室温搅拌5天,过滤,制得晶型B。
Figure PCTCN2017086760-appb-000014
第四种实验方式
取约15~20mg晶型A,加入相应溶剂形成混悬液,置于40℃下,搅拌5天,过滤,制得晶型B。
Figure PCTCN2017086760-appb-000015
Figure PCTCN2017086760-appb-000016
第五种实验方式
取约15~40mg晶型A,加入溶剂2后,再加入溶剂1,形成混悬液,置于相应温度下搅拌4天,过滤,制得晶型B。
Figure PCTCN2017086760-appb-000017
第六种实验方式
取约10mg晶型A,加入相应溶剂后,超声溶解成澄清溶液后过滤,添加1mg的高分子,置于室温下,敞口挥发,其中丙酮∶水=3∶1(体积比),乙腈∶水=3∶2(体积比),制得晶型B。
Figure PCTCN2017086760-appb-000018
Figure PCTCN2017086760-appb-000019
第七种实验方式
取约15~50mg晶型A,在相应温度下,加入相应溶剂,溶解成澄清溶液后过滤,置于4℃搅拌至析出固体,过滤,制得晶型B。其中,异丙醇在4℃搅拌时无析出,后室温挥发,制得晶型B。
序号 温度(℃) 晶型A质量(mg) 溶剂 溶剂(体积,mL)
晶型B制备方法58 60 50 甲醇 1.0
晶型B制备方法59 70 15 乙醇 1.2
晶型B制备方法60 70 15 正丙醇 1.4
晶型B制备方法61 70 10 正丁醇 2.0
晶型B制备方法62 70 50 0.4
晶型B制备方法63 70 10 异丙醇 2.0
第八种实验方式
取约10~15mg晶型A,在相应温度下,加入溶剂2后,再加入溶剂1,溶解成澄清溶液后过滤,置于-20℃搅拌至析出固体,制得晶型B。其中,晶型B制备方法69-74搅拌无析出,后室温下挥发制得晶型B。
Figure PCTCN2017086760-appb-000020
第九种实验方式
取约10~15mg晶型A,在室温下,加入溶剂1后,超声溶解成澄清溶液后过滤,搅 拌下向溶液内滴加溶剂2,搅拌至析出大量固体,过滤,制得晶型B。其中,晶型B制备方法89和90搅拌无析出,后来室温挥发,制得晶型B。
Figure PCTCN2017086760-appb-000021
第十种实验方式
取约10mg结构式I所示化合物的无定型样品,置于2.0mL离心管中,然后,室温下,将离心管置于相应溶剂气氛中,扩散6天,制得晶型B。
序号 样品质量(mg) 溶剂 溶剂(体积,mL)
晶型B制备方法91 10 正丁醇 4.0
晶型B制备方法92 10 4.0
晶型B制备方法93 10 硝基甲烷 4.0
晶型B制备方法94 10 乙酸乙酯 4.0
晶型B制备方法95 10 甲基叔丁基醚 4.0
晶型B制备方法96 10 四氢呋喃 4.0
晶型B制备方法97 10 二氯甲烷 4.0
晶型B制备方法98 10 氯仿 4.0
晶型B制备方法99 10 甲苯 4.0
第十一种实验方式
取约15~30mg结构式I所示化合物的无定型样品,加入相应溶剂后,置于相应温度下,搅拌30分钟,过滤,制得晶型B。
Figure PCTCN2017086760-appb-000022
第十二种实验方式
取约15~30mg结构式I所示化合物的无定型样品,加入溶剂2后,再加入溶剂1,制备成混悬液后,置于相应温度下搅拌30分钟,过滤,制得晶型B。
Figure PCTCN2017086760-appb-000023
Figure PCTCN2017086760-appb-000024
第十三种实验方式晶型B制备方法142
取约10mg结构式I所示化合物的无定型样品,在室温下,敞口静置于湿度为85%RH的气氛内10天,制得晶型B。
第十四种实验方式
取适量样品,加入相应溶剂,超声溶解成澄清溶液后过滤,在相应水浴温度下快速旋干,制得晶型B。
序号 温度(℃) 样品质量(mg) 溶剂 溶剂(体积,mL)
晶型B制备方法143 60 15 0.4
晶型B制备方法144 40 1000 甲醇 40.0
实施例4:晶型C的制备方法
晶型C制备方法1:
取100mg结构式I所示化合物的晶型A样品,加入2.8mL异丙醚后再加入2.8mL甲醇形成混悬液,室温搅拌4天,减压抽滤,得到73mg晶型C
晶型C制备方法2:
取100mg结构式I所示化合物的晶型A样品,加入1.0mL异丙醚后再加入3.0mL甲醇形成混悬液,室温搅拌5天,减压抽滤,即得到63mg晶型C。
晶型C制备方法3和4:
取约15mg结构式I所示化合物的晶型A样品,在相应温度下,加入溶剂2后再加入溶剂1后溶清,过滤,置于-20℃搅拌至析出固体,制得晶型C。
Figure PCTCN2017086760-appb-000025
晶型C制备方法5:
取约10mg结构式I所示化合物的晶型A样品,置于2.0mL离心管中,室温敞口置于4mL甲醇密闭溶剂气氛中扩散6天,制得晶型C。
实施例5:晶型D的制备方法
晶型D制备方法1:
取100mg结构式I所示化合物的晶型A样品,加入2.0mL乙酸乙酯后再加入1.0mL二甲亚砜形成混悬液,室温搅拌1天,制得晶型D。
晶型D制备方法2:
取约5mg结构式I所示化合物的晶型A样品,加入0.1mL二甲亚砜溶清后过滤,置于40℃温度下敞口挥发,制得晶型D。
晶型D制备方法3:
取约20mg结构式I所示化合物的晶型A样品,加入溶剂2后再加入溶剂1形成混悬液后置于相应温度下搅拌4天,制得晶型D。
温度(℃) 样品量(mg) 溶剂1 溶剂1(mL) 溶剂2 溶剂2(mL)
40 20 乙酸乙酯 1.0 二甲亚砜 0.5
晶型D制备方法4和5:
取约15mg结构式I所示化合物的晶型A样品,在室温下,加入溶剂1后超声溶清,过滤,在搅拌下向溶液内滴加溶剂2至析出大量固体,即得晶型D。
序号 样品量(mg) 溶剂1 溶剂1(mL) 溶剂2 溶剂2(mL)
制备方法4 15 二甲亚砜 0.2 丙酮 3.0
制备方法5 15 二甲亚砜 0.2 乙酸乙酯 5.0
实施例6:晶型E的制备方法
取约20mg结构式I所示化合物的晶型A样品,室温置于85%RH湿度器内26天,制得晶型E。
实施例7:无定型物的制备方法
无定型物制备方法1:
取200mg结构式I所示化合物的晶型A样品,加入0.6mL三氟乙醇,超声溶清过滤,40℃快速减压旋干,得到无定型物。
无定型物制备方法2:
取200mg结构式I所示化合物的晶型A样品,加入7.0mL甲醇,超声溶清过滤,40℃快速减压旋干,得到无定型。
无定型物制备方法3-5:
取约5~10mg结构式I所示化合物的晶型A样品,加入相应溶剂溶清后过滤,置于相应温度下敞口挥发,得到无定型物。
序号 温度(℃) 样品量(mg) 溶剂 溶剂用量(mL)
制备方法3 室温 5 三氟乙醇 0.4
制备方法4 40 5 正丙醇 1.0
制备方法5 40 10 三氟乙醇 0.4
无定型物制备方法6-9:
取约10mg结构式I所示化合物的晶型A样品,加入相应溶剂溶清后过滤,置于相应温度下敞口挥发,得到无定型物。
Figure PCTCN2017086760-appb-000026
无定型物制备方法10:
取约15mg结构式I所示化合物的晶型A样品,加入2.0mL正庚烷后置于室温搅拌5天,得到无定型物。
无定型物制备方法11和12:
取约10~15mg结构式I所示化合物的晶型A样品,在相应温度下,加入相应溶剂后溶清,过滤,置于-20℃搅拌至析出固体,得到无定型物。
Figure PCTCN2017086760-appb-000027
无定型物制备方法13:
取约15mg结构式I所示化合物的晶型A样品,在室温下,加入0.2mL三氟乙醇后超声溶清,过滤,在搅拌下向溶液内滴加1.0mL异丙醚至析出大量固体,得到无定型物。
无定型物制备方法14-17:
取约10~15mg结构式I所示化合物的晶型A样品,加入相应溶剂溶清后过滤,在相应温度下快速旋干,得到无定型物。
Figure PCTCN2017086760-appb-000028
实施例8:晶型稳定性的测定
晶型A和晶型B样品在80℃干燥放置24小时、25℃-60%RH放置10天、40℃-75%RH放置10天,晶型均不变。
晶型C在室温真空干燥过夜后几乎全部转为晶型A。
晶型D室温晾干或室温真空干燥过夜大部分转为晶型B,不能稳定存在。
晶型E放置于干燥器中2天大部分转为晶型A,不稳定。
实施例9:动态水分吸附(DVS)测定
晶型A:0%-80%RH范围内重量变化约为2.3%,该水合物在0%RH干燥阶段脱去约1.5%的水分,0%RH-80%RH范围内可吸湿约为2.3%,在脱吸附阶段在30%RH可脱去1.5%水分,在30-80%RH范围内重量变化小于2%。
晶型B:0%-80%RH范围内重量变化约为2.3%,该水合物在10%RH以下可脱去约 4%的水分,在吸附阶段在10%RH又可吸附约4%水分,在10-80%RH范围内重量变化小于2%。
无定型:0-80%RH范围内重量变化约为15.7%,极易吸湿。
实施例10:溶解度测定
结构式I所示化合物晶型A室温时在水中的溶解度是20-100mg/mL,结构I所示化合物游离碱无定型物在水中的溶解度<1mg/mL。
实施例11.胶囊的配方
作为一个口服药的明确具体化,取大约20至150mg实施例1和/或2记载的多晶型,用充分细微划分的微晶纤维素和/或硬脂酸调配得到总量约为50mg至500mg去填充0型胶囊。
实施例12.片剂或胶囊的配方
作为一个口服药的明确具体化,取大约20至150mg实施例1和/或2记载的多晶型,用二种或以上的下述辅料,充分细微划分的微晶纤维素、甘露醇、交联聚维酮、交联羧甲基纤维素钠、羧甲基淀粉钠、聚维酮、羟丙基纤维素和/或硬脂酸调配得到总量约为50mg至500mg的片剂或胶囊。
实施例13:药代动力学数据
雌性SD大鼠6只,分为两组,每组三只,分别单次灌胃50mg/kg给药结构I所示化合物晶型A和结构I所示化合物游离碱无定型物;分别在指定的时间点通过颈静脉采血,分离血浆,放入-80℃冰箱保存。
上述血浆样品,乙腈沉淀蛋白,取上清用水稀释3倍,取5uL至LC-MS/MS检测,试验数据如表9所示:
表9
Figure PCTCN2017086760-appb-000029
大鼠口服给予50mg/kg结构I所示化合物晶型A和结构I所示化合物游离碱无定型 物后,Tmax分别为2.67和4.67h,Cmax分别为1440和890ng/mL,AUClast分别为11551和8165h*ng/mL。
由以上结果提示,相较于结构I所示化合物游离碱无定型物,结构I所示化合物晶型A在体内吸收更好。
实施例14:结构式I所示化合物的生物化学激酶活性
结构式I所示化合物的生物化学激酶活性实验由位于美国宾夕法尼亚州(PA)莫尔文(Malvern)的反应生物有限公司(Reaction Biology Corp)检测,检测规程参见Anastassiadis等人的Nat Biotechnol.2011;29(11):1039-45。发现结构式I所示化合物潜在地抑制如下激酶:
Figure PCTCN2017086760-appb-000030
虽然本发明已通过其实施方式连同参考附图进行了全面的描述,但是值得注意的是,各种变化和修改对于本领域技术人员都是显而易见的。这样的变化和修改都应该包括在本发明所附权利要求的范围内。

Claims (51)

  1. 结构式I所示的化合物的制备方法,其特征在于,包括如下步骤
    Figure PCTCN2017086760-appb-100001
  2. 结构式I所示的化合物的制备方法,其特征在于,包括如下步骤
    Figure PCTCN2017086760-appb-100002
  3. 结构式I所示的化合物的制备方法,其特征在于,包括如下步骤
    Figure PCTCN2017086760-appb-100003
  4. 中间体化合物:
    Figure PCTCN2017086760-appb-100004
  5. 结构式I所示化合物、其水合物和/或其溶剂化物的晶型,
    Figure PCTCN2017086760-appb-100005
  6. 根据权利要求5所述的结构式I所示化合物的晶型,其特征在于:该晶型X射线衍射谱图具有衍射角2θ为4.9±0.2°、10.0±0.2°和19.3±0.2°的特征峰。
  7. 根据权利要求5或6所述的结构式I所示化合物的晶型,其特征在于:该晶型X射线粉末衍射谱图具有衍射角2θ为4.9±0.2°、10.0±0.2°、14.7±0.2°、16.9±0.2°、19.3±0.2°和20.3±0.2°的特征峰。
  8. 根据权利要求5-7任一项所述的结构式I所示化合物的晶型,其特征在于:该晶型X射线粉末衍射谱图具有衍射角2θ为4.9±0.2°、10.0±0.2°、14.7±0.2°、16.9±0.2°、19.3±0.2°、20.3±0.2°、25.5±0.2°和30.7±0.2°的特征峰。
  9. 根据权利要求5-8任一项所述的结构式I所示化合物的晶型,其特征在于:具有约如图1所示的X-射线粉末衍射图。
  10. 根据权利要求5-9任一项所述的结构式I所示化合物的晶型,其特征在于:该晶型为二水合物。
  11. 根据权利要求5所述的结构式I所示化合物的晶型,其特征在于:该晶型X射线粉末衍射谱图中具有衍射角2θ为10.5±0.2°、17.4±0.2°和21.1±0.2°的特征峰。
  12. 根据权利要求5或11所述的结构式I所示化合物的晶型,其特征在于:该晶型X射线粉末衍射谱图中具有衍射角2θ为10.5±0.2°、17.4±0.2°、19.7±0.2°、21.1±0.2°、23.9±0.2°和25.5±0.2°的特征峰。
  13. 根据权利要求5、11或12所述的结构式I所示化合物的晶型,其特征在于:该晶型X射线粉末衍射谱图中具有衍射角2θ为10.5±0.2°、17.4±0.2°、19.7±0.2°、21.1±0.2°、21.5±0.2°、23.9±0.2°、25.2±0.2°、和25.5±0.2°的特征峰。
  14. 根据权利要求5、11-13任一项所述的结构式I所示化合物的晶型,其特征在于:具有约如图2所示的X-射线粉末衍射图。
  15. 根据权利要求5、11-14任一项所述的结构式I所示化合物的晶型,其特征在于:该晶型为三水合物。
  16. 根据权利要求5-15任一项所述的结构式I所示化合物的晶型,其特征在于,所述晶型纯度≥85%。
  17. 根据权利要求5-16任一项所述的结构式I所示化合物的晶型,其特征在于,所述晶型纯度≥95%。
  18. 根据权利要求5-17任一项所述的结构式I所示化合物的晶型,其特征在于,所述晶型纯度≥99%。
  19. 一种药物组合物,其特征在于:含有治疗有效量的权利要求5-18任一项所述的晶型,和药学上可接受的辅料、辅助剂和/或载体。
  20. 一种药物组合物,其特征在于:含有治疗有效量的权利要求5-18任一项所述的晶型,联用至少一种其他有效成分。
  21. 根据权利要求19或20所述的药物组合物,其特征在于:所述药物组合物为口服制剂。
  22. 根据权利要求19-21任一项所述的药物组合物,其特征在于:所述药物组合物为片剂或胶囊。
  23. 根据权利要求19-22任一项所述的药物组合物,其特征在于:20至150mg权利要求5-18任一项所述的晶型,用至少一种辅料、辅助剂和/或载体调配到总量约为50mg至500mg。
  24. 根据权利要求19-23任一项所述的药物组合物,其特征在于:所述辅料、辅助剂和/或载体为微晶纤维素、甘露醇、交联聚维酮、交联羧甲基纤维素钠、羧甲基淀粉钠、聚维酮、羟丙基纤维素和/或硬脂酸。
  25. 根据权利要求19-24任一项所述的药物组合物,其特征在于:所述药物组合物中含有0.01重量%~99重量%的权利要求5-18任一项所述的晶型。
  26. 根据权利要求19-25任一项所述的药物组合物,其特征在于:所述药物组合物中含有0.1重量%~70重量%权利要求5-18任一项所述的晶型。
  27. 根据权利要求19-26任一项所述的药物组合物,其特征在于:所述药物组合物中含有1重量%~70重量%权利要求5-18任一项所述的晶型。
  28. 根据权利要求19-27任一项所述的药物组合物,其特征在于:所述药物组合物中含有1重量%~50重量%权利要求5-18任一项所述的晶型。
  29. 根据权利要求19-28任一项所述的药物组合物,其特征在于:所述药物组合物中含有1重量%~30重量%权利要求5-18任一项所述的晶型。
  30. 根据权利要求19-29任一项所述的药物组合物,其特征在于:所述药物组合物中含有10重量%~30重量%权利要求5-18任一项所述的晶型。
  31. 权利要求5-18任一项所述的晶型或权利要求19-30任一项所述的药物组合物在制备治疗患者的疾病、病症或病状的药物中的用途,其中所述疾病、病症或病状由c-Met、RON、Axl、CSF1R、EphA2、ROS1或ROS1融合蛋白、TRKA或TRKA融合蛋白、TRKB、TRKC、ALK、ALKATI或ALK融合蛋白介导。
  32. 根据权利要求31所述的用途,其中所述ALK融合蛋白是EML4-ALK或NPM-ALK激酶。
  33. 根据权利要求31或32所述的用途,其中所述疾病、病症或病状是癌症和/或增殖性疾病。
  34. 根据权利要求31-33任一项所述的用途,其中所述疾病、病症或病状是肺癌、黑色素瘤、结肠癌、乳腺癌、肝癌、胰腺癌、脑癌、肾癌、卵巢癌、胃癌、皮肤癌、骨癌、神经胶质瘤、淋巴瘤、成神经细胞瘤、肝细胞癌、***状肾细胞癌和/或头颈部鳞状细胞癌。
  35. 根据权利要求31-34任一项所述的用途,其中所述疾病、病症或病状是对克唑 替尼治疗有耐药性的非小细胞肺癌。
  36. 根据权利要求31-34任一项所述的用途,其中所述疾病、病症或病状是黑色素瘤。
  37. 根据权利要求31-34任一项所述的用途,其中所述疾病、病症或病状是神经性疾病、精神性疾病、肥胖、糖尿病和/或心血管疾病。
  38. 根据权利要求37所述的用途,其中,所述精神性疾病是精神***症、抑郁症和/或物质成瘾或滥用。
  39. 根据权利要求38所述的用途,其中所述物质成瘾或滥用是***、烟草或酒精的成瘾或滥用。
  40. 一种治疗患者疾病、病症或病状的方法,其特征在于:对患者施用权利要求5-18任一项所述的晶型或权利要求19-30任一项所述的药物组合物。
  41. 根据权利要求40所述的治疗患者疾病、病症或病状的方法,其特征在于:所述的疾病、病症或病状是由c-Met、RON、Axl、CSF1R、EphA2、ROS1或ROS1融合蛋白、TRKA或TRKA融合蛋白、TRKB、TRKC、ALK、ALKATI或ALK融合蛋白介导。
  42. 根据权利要求41所述的治疗患者疾病、病症或病状的方法,其特征在于:所述ALK融合蛋白是EML4-ALK或NPM-ALK激酶。
  43. 根据权利要求40-42任一项所述的治疗患者疾病、病症或病状的方法,其特征在于:所述的疾病、病症或病状是癌症和/或增殖性疾病。
  44. 根据权利要求40-43任一项所述的治疗患者疾病、病症或病状的方法,其特征在于:所述的疾病、病症或病状是肺癌、黑色素瘤、结肠癌、乳腺癌、肝癌、胰腺癌、脑癌、肾癌、卵巢癌、胃癌、皮肤癌、骨癌、神经胶质瘤、淋巴瘤、成神经细胞瘤、肝细胞癌、***状肾细胞癌和/或头颈部鳞状细胞癌。
  45. 根据权利要求40-44任一项所述的治疗患者疾病、病症或病状的方法,其特征在于:其中所述疾病、病症或病状是对克唑替尼治疗有耐药性的非小细胞肺癌。
  46. 根据权利要求40-44任一项所述的治疗患者疾病、病症或病状的方法,其中所述疾病、病症或病状是黑色素瘤。
  47. 根据权利要求40所述的治疗患者疾病、病症或病状的方法,其特征在于:所述疾病、病症或病状是神经性疾病、精神性疾病、肥胖、糖尿病和/或心血管疾病。
  48. 根据权利要求47所述的治疗患者疾病、病症或病状的方法,其特征在于:所述精神性疾病是精神***症、抑郁症和/或物质成瘾或滥用。
  49. 根据权利要求48所述的治疗患者疾病、病症或病状的方法,其特征在于:所述物质成瘾或滥用是***、烟草或酒精的成瘾或滥用。
  50. 制备权利要求6-10任一项所述的结构式I所示化合物的晶型的方法,其特征在于:包括如下的步骤:
    结构式I所示化合物的无定型样品,置于离心管中,室温敞口置于乙醇或乙腈密闭溶剂气氛中静置扩散6~10天,制得该晶型;或者,
    结构式I所示化合物的无定型样品,加入乙醇溶剂后,置于4℃~25℃下,搅拌,过滤,制得该晶型;或者,
    4℃~25℃下,将结构式I所示化合物的无定型样品,加入到乙醇中,溶解成澄清溶液后过滤,在搅拌下向滤液内滴加正庚烷至析出大量晶型,过滤制得该晶型;或者,
    55℃~70℃下,将结构式I所示化合物的无定型样品,加入到甲基叔丁基醚/乙醇或正庚烷/乙醇中,溶解成澄清溶液后过滤,所得滤液置于-20℃,搅拌至析出固体,制得该晶型;或者,
    将结构式I所示化合物的无定型样品,加入到甲醇中,溶解成澄清溶液后过滤,置于35℃-50℃下敞口挥发,制得该晶型;或者,
    将结构式I所示化合物的无定型样品,加入甲醇,溶解成澄清溶液,过滤,加入羧甲基纤维素,置于室温下敞口挥发,制得该晶型。
  51. 制备权利要求11-15任一项所述的结构式I所示化合物的晶型的方法,其特征在于:包括如下的步骤:
    将权利要求6-10任一项所述的结构式I所示化合物的晶型,加入甲醇、乙醇或水中,溶解成澄清溶液后过滤,置于室温(20℃)-40℃下敞口挥发干,制得该晶型;或者,
    将权利要求6-10任一项所述的结构式I所示化合物的晶型,加入甲醇/水、甲醇/丙酮、甲醇/乙酸乙酯、甲醇/甲基叔丁基醚、甲醇/四氢呋喃、甲醇/二氯甲烷、乙醇/水、乙醇/丁酮、乙醇/乙酸异丙酯、乙醇/正庚烷、三氟乙醇/水、三氟乙醇/乙酸乙酯、三氟乙醇/四氢呋喃、水/甲醇、水/乙醇、水/三氟乙醇、水/异丙醇、水/丙酮、水/四氢呋喃或水/乙腈,,溶解成澄清溶液后过滤,置于室温(20℃)-40℃下敞口挥发干,制得该晶型;或者,
    将权利要求6-10任一项所述的结构式I所示化合物的晶型,加入低级醇、水、硝基甲烷、丁酮、***、乙酸乙酯、四氢呋喃、甲苯或正庚烷中,形成混悬液,至于室温-40℃下,搅拌4~5天,取混悬液离心,制得该晶型;或者,
    将权利要求6-10任一项所述的结构式I所示化合物的晶型,加入水饱和的乙酸乙酯 层、乙酸乙酯饱和的水层、乙醇/***、甲苯/乙腈、丁酮/乙醇或甲苯/异丙醚,形成混悬液,置于4℃-40℃搅拌4-5天,离心,制得该晶型;或者,
    将权利要求6-10任一项所述的结构式I所示化合物的晶型,室温下加入甲醇、丙酮/水(3∶1v/v)或乙腈/水(3∶2v/v)溶剂中,溶解成澄清溶液后,添加羟丙基纤维素、乙基纤维素、聚维酮K30、聚烯丙基胺盐酸盐、羧甲基纤维素或聚乙烯醇后,置于室温下,敞口挥发,制得该晶型;或者,
    将权利要求6-10任一项所述的结构式I所示化合物的晶型,在60℃-70℃下,加入低级醇或水,溶解成澄清溶液,置于4℃搅拌至析出晶型,制得该晶型;或者,
    将权利要求6-10任一项所述的结构式I所示化合物的晶型,在55℃-70℃下,加入丙酮/三氟乙醇、丙酮/水、二恶烷/水、乙腈/水或甲基叔丁基醚/正丙醇中,溶解成澄清溶液,过滤,滤液置于-20℃搅拌至析出晶型,过滤,制得该晶型;或者,
    将权利要求6-10任一项所述的结构式I所示化合物的晶型,在60℃-70℃下,加入硝基甲烷/甲醇、乙腈/甲醇、丁酮/乙醇、乙酸乙酯/乙醇、1,4-二氧六环/乙醇或者四氢呋喃/水中,溶解成澄清溶液,过滤,室温下挥发,制得该晶型;或者,
    取权利要求6-10任一项所述的结构式I所示化合物的晶型,在室温下,加入甲醇、乙醇、水、三氟乙醇、正丙醇或二甲亚砜,溶解成澄清溶液后,过滤,向滤液内滴加丙酮、乙酸乙酯、甲基叔丁基醚、异丙醚、乙酸异丙酯、四氢呋喃、1,4-二氧六环、乙腈、正庚烷、二氯甲烷或氯仿,至析出大量晶型,制得该晶型;或者,
    取权利要求6-10任一项所述的结构式I所示化合物的晶型,在室温下,加入甲醇或乙醇,溶解成澄清溶液后,过滤,向滤液内滴加二氯甲烷或四氢呋喃,室温挥发溶剂,制得该晶型;或者,
    取结构式I所示化合物的无定型样品,置于离心管中,然后将该离心管置于正丁醇、水、硝基甲烷、乙酸乙酯、甲基叔丁基醚、四氢呋喃、二氯甲烷、氯仿或甲苯气氛中扩散,制得该晶型;或者,
    取结构式I所示化合物的无定型样品,加入正丙醇、水、丁酮、乙酸乙酯、四氯呋喃、二氯甲烷、乙醇、异丙醇、正丁醇、丙酮、***、乙酸异丙酯、1,4-二氧六环、乙腈、氯仿、仲丁醇、硝基甲烷或甲苯中,然后置于-4℃-40℃下,搅拌30分钟,过滤,制得该晶型;或者,
    取结构式I所示化合物的无定型样品,加入异丙醚/甲醇、乙酸乙酯/甲醇、1,4-二氧六环/甲醇、丁酮/乙醇、乙腈/乙醇、正庚烷/乙醇、硝基甲烷/三氟乙醇、***/三氟乙醇、四 氢呋喃/三氟乙醇、丙酮/水、四氢呋喃/水、乙腈/水、甲基叔丁基醚/异丙醇、乙酸异丙酯/正丙醇、甲基环己烷/正丁醇、丙酮/二甲亚砜、乙酸乙酯/二甲亚砜、乙腈/二甲亚砜、甲基叔丁基醚/氯仿或者甲苯/乙酸乙酯,形成混悬液后,置于4℃-40℃下搅拌,过滤,制得该晶型;或者,
    取结构式I所示化合物的无定型样品,敞口静置于湿度为85%RH的室温下,静置,制得该晶型;或者,
    取权利要求6-10任一项所述的结构式I所示化合物的晶型,加入水或甲醇,溶解成澄清溶液,过滤,旋干,制得该晶型。
PCT/CN2017/086760 2016-06-01 2017-06-01 抑制蛋白激酶活性化合物的晶型及其应用 WO2017206924A1 (zh)

Priority Applications (16)

Application Number Priority Date Filing Date Title
SG11201810800VA SG11201810800VA (en) 2016-06-01 2017-06-01 Crystalline form of compound suppressing protein kinase activity, and application thereof
EP17805869.9A EP3470410B1 (en) 2016-06-01 2017-06-01 Crystalline form of compound suppressing protein kinase activity, and application thereof
RU2018145183A RU2744264C2 (ru) 2016-06-01 2017-06-01 Кристаллическая форма соединения, подавляющего активность протеинкиназы, и её применение
AU2017274110A AU2017274110B2 (en) 2016-06-01 2017-06-01 Crystalline form of compound suppressing protein kinase activity, and application thereof
EA201892687A EA201892687A1 (ru) 2016-06-01 2017-06-01 Кристаллическая форма соединения, подавляющего активность протеинкиназы, и её применение
BR112018074612-6A BR112018074612A2 (pt) 2016-06-01 2017-06-01 forma cristalina de composto que suprimir a atividade de proteína quinase e aplicação da mesma
US16/306,104 US10899744B2 (en) 2016-06-01 2017-06-01 Crystalline form of compound suppressing protein kinase activity, and application thereof
MYPI2018002245A MY188379A (en) 2016-06-01 2017-06-01 Crystalline form of compound suppressing protein kinase activity, and application thereof
KR1020187038142A KR102466958B1 (ko) 2016-06-01 2017-06-01 단백질 키나제 활성을 억제하는 화합물의 결정 형태 및 이의 응용
CA3026142A CA3026142C (en) 2016-06-01 2017-06-01 Crystalline form of compound suppressing protein kinase activity, and application thereof
IL263356A IL263356B (en) 2016-06-01 2017-06-01 A crystalline form of a compound that suppresses protein kinase activity, and its application
CN201780006852.3A CN109195964B (zh) 2016-06-01 2017-06-01 抑制蛋白激酶活性化合物的晶型及其应用
JP2018563619A JP7054528B2 (ja) 2016-06-01 2017-06-01 プロテインキナーゼ活性を抑制する化合物の結晶形態、及びその適用
PH12018502529A PH12018502529A1 (en) 2016-06-01 2018-11-29 Crystalline form of compound suppressing protein kinase activity, and application thereof
ZA2018/08625A ZA201808625B (en) 2016-06-01 2018-12-20 Crystalline form of compound suppressing protein kinase activity, and application thereof
HK19101605.4A HK1259249A1 (zh) 2016-06-01 2019-01-30 抑制蛋白激酶活性化合物的晶型及其應用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNPCT/CN2016/084300 2016-06-01
CNPCT/CN2016/084300 2016-06-01

Publications (1)

Publication Number Publication Date
WO2017206924A1 true WO2017206924A1 (zh) 2017-12-07

Family

ID=60477983

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/086760 WO2017206924A1 (zh) 2016-06-01 2017-06-01 抑制蛋白激酶活性化合物的晶型及其应用

Country Status (17)

Country Link
US (1) US10899744B2 (zh)
EP (1) EP3470410B1 (zh)
JP (1) JP7054528B2 (zh)
KR (1) KR102466958B1 (zh)
CN (1) CN109195964B (zh)
AU (1) AU2017274110B2 (zh)
BR (1) BR112018074612A2 (zh)
CA (1) CA3026142C (zh)
EA (1) EA201892687A1 (zh)
HK (1) HK1259249A1 (zh)
IL (1) IL263356B (zh)
MY (1) MY188379A (zh)
PH (1) PH12018502529A1 (zh)
RU (1) RU2744264C2 (zh)
SG (1) SG11201810800VA (zh)
TW (1) TWI646094B (zh)
WO (1) WO2017206924A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113861191A (zh) * 2018-06-21 2021-12-31 贝达药业股份有限公司 抑制cdk4/6活性化合物的晶型及其应用
WO2022031636A1 (en) 2020-08-03 2022-02-10 Teva Pharmaceuticals International Gmbh Solid state forms of ensartinib and ensartinib salts
WO2023016321A1 (zh) * 2021-08-10 2023-02-16 贝达药业股份有限公司 恩沙替尼或其盐在治疗携带met 14外显子跳跃突变的疾病中的用途

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004076412A2 (en) 2003-02-26 2004-09-10 Sugen, Inc. Aminoheteroaryl compounds as protein kinase inhibitors
WO2006021881A2 (en) 2004-08-26 2006-03-02 Pfizer Inc. Pyrazole-substituted aminoheteroaryl compounds as protein kinase inhibitors
WO2006021886A1 (en) 2004-08-26 2006-03-02 Pfizer Inc. Aminoheteroaryl compounds as protein tyrosine kinase inhibitors
WO2009154769A1 (en) 2008-06-19 2009-12-23 Xcovery, Inc. Substituted pyridazine carboxamide compounds as kinase inhibitor compounds
WO2012048259A2 (en) 2010-10-08 2012-04-12 Xcovery Holding Company, Llc Substituted pyridazine carboxamide compounds

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2001233028A1 (en) * 2000-03-06 2001-10-03 Warner-Lambert Company 5-alkylpyrido(2,3-D)pyrimidines tyrosine kinase inhibitors
US8414489B2 (en) 2003-11-13 2013-04-09 Medtronic Minimed, Inc. Fabrication of multi-sensor arrays
WO2007066185A2 (en) * 2005-12-05 2007-06-14 Pfizer Products Inc. Polymorphs of a c-met/hgfr inhibitor
CN101652068A (zh) * 2007-01-19 2010-02-17 艾科睿制药公司 激酶抑制物化合物
JP5909236B2 (ja) * 2010-10-08 2016-04-26 エックスカバリー ホールディング カンパニー エルエルシー キナーゼ阻害剤化合物としての置換ピリダジンカルボキサミド化合物

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004076412A2 (en) 2003-02-26 2004-09-10 Sugen, Inc. Aminoheteroaryl compounds as protein kinase inhibitors
WO2006021881A2 (en) 2004-08-26 2006-03-02 Pfizer Inc. Pyrazole-substituted aminoheteroaryl compounds as protein kinase inhibitors
WO2006021886A1 (en) 2004-08-26 2006-03-02 Pfizer Inc. Aminoheteroaryl compounds as protein tyrosine kinase inhibitors
WO2009154769A1 (en) 2008-06-19 2009-12-23 Xcovery, Inc. Substituted pyridazine carboxamide compounds as kinase inhibitor compounds
CN102098917A (zh) * 2008-06-19 2011-06-15 艾科睿控股公司 作为激酶抑制剂化合物的取代的哒嗪羧酰胺化合物
WO2012048259A2 (en) 2010-10-08 2012-04-12 Xcovery Holding Company, Llc Substituted pyridazine carboxamide compounds
CN103298806A (zh) * 2010-10-08 2013-09-11 艾科睿控股公司 取代的哒嗪羧酰胺化合物
CN103298806B (zh) 2010-10-08 2015-03-18 贝达药业股份有限公司 取代的哒嗪羧酰胺化合物

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
"Pasquale EB. Eph receptors and ephrins in cancer: bidirectional signaling and beyond.", NAT REV CANCER, vol. 10, 2010, pages 165 - 80
AMATO ET AL., CANCER RES, vol. 76, no. 2, 2016, pages 305 - 18
ANASTASSIADIS, NAT BIOTECHNOL., vol. 29, no. 11, 2011, pages 1039 - 45
BERGETHON ET AL., J. CLIN. ONCOL., vol. 30, no. 8, 2012, pages 863
CHOI ET AL., CANCER RES, vol. 68, no. 13, 2008
INTERNATIONAL JOURNAL OF PATHOLOGY AND CLINICAL MEDIC, vol. 25, no. 5, 2005, pages 441 - 443
J CLIN ONCOLOGY, vol. 28, 2010, pages 7S
JEFFERSON, M. ET AL., ONCOGENE, vol. 13, 1996, pages 853 - 856
MICHIELI, P. ET AL., ONCOGENE, vol. 18, 1999, pages 5221 - 5231
ONCOGENE, vol. 28, 2009, pages 3442
See also references of EP3470410A4
VAISHNAVIL ET AL., NATURE MEDICINE, vol. 19, no. 11, 2013, pages 1469
WEBB ET AL., EXPERT REV. ANTICANCER THER., vol. 9, no. 3, 2009, pages 331 - 356
WIESNER ET AL., NATURE, vol. 526, 2015, pages 453

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113861191A (zh) * 2018-06-21 2021-12-31 贝达药业股份有限公司 抑制cdk4/6活性化合物的晶型及其应用
CN113861191B (zh) * 2018-06-21 2023-09-19 贝达药业股份有限公司 抑制cdk4/6活性化合物的晶型及其应用
WO2022031636A1 (en) 2020-08-03 2022-02-10 Teva Pharmaceuticals International Gmbh Solid state forms of ensartinib and ensartinib salts
WO2023016321A1 (zh) * 2021-08-10 2023-02-16 贝达药业股份有限公司 恩沙替尼或其盐在治疗携带met 14外显子跳跃突变的疾病中的用途

Also Published As

Publication number Publication date
BR112018074612A2 (pt) 2019-03-19
RU2744264C2 (ru) 2021-03-04
CA3026142C (en) 2024-02-13
RU2018145183A3 (zh) 2020-07-16
CN109195964B (zh) 2021-08-10
IL263356A (en) 2018-12-31
JP2019518026A (ja) 2019-06-27
CN109195964A (zh) 2019-01-11
CA3026142A1 (en) 2017-12-07
AU2017274110A1 (en) 2019-01-24
SG11201810800VA (en) 2019-01-30
KR102466958B1 (ko) 2022-11-14
EP3470410A1 (en) 2019-04-17
RU2018145183A (ru) 2020-07-14
AU2017274110B2 (en) 2021-05-06
EP3470410B1 (en) 2024-04-17
US20190135792A1 (en) 2019-05-09
EA201892687A1 (ru) 2019-06-28
IL263356B (en) 2022-08-01
TWI646094B (zh) 2019-01-01
PH12018502529A1 (en) 2019-04-08
JP7054528B2 (ja) 2022-04-14
EP3470410A4 (en) 2019-12-04
HK1259249A1 (zh) 2019-11-29
TW201742865A (zh) 2017-12-16
KR20190012231A (ko) 2019-02-08
US10899744B2 (en) 2021-01-26
MY188379A (en) 2021-12-07

Similar Documents

Publication Publication Date Title
CA2858033C (en) Quinolyl-containing hydroxamic acid compound and preparation method thereof, and pharmaceutical composition containing this compound and use thereof
WO2021190467A1 (zh) 含螺环的喹唑啉化合物
BRPI0911679B1 (pt) Composto aciltioureia ou um sal do mesmo, uso do dito composto para tratar câncer, bem como agente farmacêutico, agente antitumoral e composição farmacêutica compreendendo dito composto
WO2017206924A1 (zh) 抑制蛋白激酶活性化合物的晶型及其应用
US10870642B2 (en) Biheterocyclic compound
WO2017152707A1 (zh) 吡啶胺基嘧啶衍生物甲磺酸盐的结晶形式及其制备和应用
WO2016078160A1 (zh) 胞苷衍生物及其应用
CN111788197A (zh) 取代的二氨基杂环甲酰胺化合物及包含该化合物的组合物及其用途
JP2019509306A (ja) Jak関連疾患の治療又は予防に用いる薬物の塩酸塩の結晶形及びその製造方法
US20210205348A1 (en) Methods for the Treatment of Bladder Cancer
WO2021143954A2 (zh) 一种氟伐替尼或其甲磺酸盐的晶型及其制备方法
WO2022183964A1 (zh) 一种8-(吡啶三氮唑)取代香豆素类化合物及其制备方法和应用
WO2022194252A1 (zh) 一种化合物的多晶型及其制备方法和应用
WO2022183961A1 (zh) 一种8-(苯并噻唑酰胺)取代香豆素类化合物及其制备方法和应用
KR20200032152A (ko) N-페닐-2-아미노피리미딘 화합물의 결정형과 염형, 및 이의 제조 방법
JP7391226B2 (ja) ピリミジン化合物の結晶
TW202104216A (zh) Plk4抑制劑之結晶型
WO2016078397A1 (zh) 新型胞苷衍生物及其应用
WO2017076358A1 (zh) 咪唑基联苯基化合物盐的新晶型及其制备方法
NZ788961A (en) Crystalline form of compound suppressing protein kinase activity, and application thereof
EA041596B1 (ru) Кристаллическая форма соединения, подавляющего активность протеинкиназы, и её применение
JP6541635B2 (ja) Parp阻害剤としての縮合四環式または縮合五環式ジヒドロジアゼピノカルバゾロン
TW202120511A (zh) 雜環化合物

Legal Events

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

Ref document number: 17805869

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2018563619

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3026142

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112018074612

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20187038142

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017805869

Country of ref document: EP

Effective date: 20190102

ENP Entry into the national phase

Ref document number: 2017274110

Country of ref document: AU

Date of ref document: 20170601

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112018074612

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20181128