WO2022127904A1 - Sel pharmaceutiquement acceptable de dérivé d'indazole, forme cristalline et procédé de préparation associé - Google Patents

Sel pharmaceutiquement acceptable de dérivé d'indazole, forme cristalline et procédé de préparation associé Download PDF

Info

Publication number
WO2022127904A1
WO2022127904A1 PCT/CN2021/139125 CN2021139125W WO2022127904A1 WO 2022127904 A1 WO2022127904 A1 WO 2022127904A1 CN 2021139125 W CN2021139125 W CN 2021139125W WO 2022127904 A1 WO2022127904 A1 WO 2022127904A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound represented
pharmaceutically acceptable
crystal form
maleate
Prior art date
Application number
PCT/CN2021/139125
Other languages
English (en)
Chinese (zh)
Inventor
王林
邵启云
冯君
贺峰
赵苗苗
杜振兴
Original Assignee
江苏恒瑞医药股份有限公司
上海恒瑞医药有限公司
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
Application filed by 江苏恒瑞医药股份有限公司, 上海恒瑞医药有限公司 filed Critical 江苏恒瑞医药股份有限公司
Priority to CN202180084474.7A priority Critical patent/CN116615418A/zh
Publication of WO2022127904A1 publication Critical patent/WO2022127904A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic 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
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms

Definitions

  • the present disclosure relates to a pharmaceutically acceptable salt, a crystalline form and a preparation method of an indazole derivative, belonging to the field of medicine.
  • Breast cancer is one of the most common malignant tumors in women. According to the 2012 GLOBALCAN statistics (CA CANCER J CLIN 2015; 65:87–108), there are about 1.7 million new cancer cases and 520,000 deaths worldwide every year. Both morbidity and mortality rank first among female malignant tumors. According to the 2017 China Cancer Registry Annual Report released by the National Cancer Center, breast cancer ranks first in the incidence of female malignant tumors, with about 279,000 new cases each year, and an annual increase of about 2%.
  • Endocrine therapy plays an important role in the treatment of this part of breast cancer patients.
  • Endocrine therapy is mainly divided into three categories, namely aromatase inhibitor (AI), which can inhibit the conversion of androgens into estrogen, reduce the level of estrogen in the body, and selective estrogen receptor modulator (selective estrogen receptor modulator).
  • SERM selective estrogen receptor modulator
  • SERM selective estrogen receptor degrader
  • SELD selective estrogen receptor degrader
  • endocrine therapy is the treatment of choice for estrogen receptor-positive breast cancer
  • about 30% of patients receiving adjuvant therapy will relapse, and almost all patients with metastatic breast cancer will develop drug resistance and progress.
  • the mechanisms of resistance to endocrine therapy are mainly divided into two categories. One is focused on the estrogen receptor signaling pathway itself, including the activating mutation, amplification, and fusion with other genes of the gene ESR1 encoding the estrogen receptor. Other mechanisms include the activation of signaling pathways that cross-react with the estrogen receptor signaling pathway, such as the growth factor receptor pathway, etc. (Nat Rev Clin Oncol. 2015 Oct. 2015). ; 12(10):573-83).
  • ESR1 gene mutations may be one of the mechanisms of drug resistance in estrogen-positive breast cancer (Nat Rev Clin Oncol. 2015 Oct;12(10):573-83 and Nat Genet 2013;45:1439-45).
  • ESR1 gene mutations were found in patients with estrogen receptor-positive metastatic breast cancer, and the mutation rate was about 30%.
  • ER Y537S and ER D538G mutations were found in 29% of the ctDNA of patients with estrogen receptor-positive metastatic breast cancer that progressed after AIs therapy.
  • PFS progression free survival
  • OS overall survival
  • ESR1 gene mutations mostly occur in metastatic estrogen receptor-positive breast cancer patients who have progressed after AIs therapy. These patients are no longer sensitive to AIs therapy. Therefore, it is necessary to develop estrogen receptors targeting ESR1 gene mutations. antagonist.
  • the first-in-class estrogen receptor covalent binding antagonist H3B-6545 developed by Eisai Company has strong inhibitory activity on wild-type and mutant estrogen receptors, and can bind to the receptor through covalent binding. To exert longer-term efficacy, clinical phase I and II trials are currently underway.
  • WO2016196346 and WO2016196342 patents for estrogen receptor antagonists with mutations in the ESR1 gene are currently underway.
  • PCT/CN2020/096744 provides an indazole derivative whose chemical name is (E)-1-morpholinyl-4-((1-(((5-((Z)-4,4,4 -Trifluoro-1-(3-fluoro-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)pyridin-2-yl)oxy)methyl)cyclopropyl ) amino) but-2-en-1-one (Formula I), providing patients with a new treatment option.
  • the present disclosure provides a pharmaceutically acceptable salt of a compound represented by formula (I), the pharmaceutically acceptable salt is selected from maleate or hydrochloride,
  • the present disclosure provides a maleic acid salt of a compound shown in formula (I), wherein the mol ratio of compound shown in formula (I) to maleic acid is 1:2-2:1.
  • the present disclosure provides a maleate salt of a compound represented by formula (I), wherein the molar ratio of the compound represented by formula (I) to maleic acid is 1:1.
  • the present disclosure also provides a method for preparing the pharmaceutically acceptable salt of the aforementioned formula (I), comprising: the step of forming a salt between the compound of formula (I) and an acid selected from maleic acid, hydrochloric acid or their solution, the solvent used in the salt-forming reaction is selected from water, methanol, n-propanol, isopropanol, ethanol, isopropyl ether, tetrahydrofuran, isopropyl acetate, acetone, butanone, methyl tert-butyl ether, acetonitrile, One or more of 1,4-dioxane, ethyl acetate and n-hexane.
  • the present disclosure further provides Form I of the maleate salt of the compound represented by formula (I), whose X-ray powder diffraction pattern has characteristic peaks at 2 ⁇ angles of 16.448, 16.956, 19.332, 20.135, 21.645, 22.257 and 22.696 .
  • the present disclosure further provides Form I of the maleate salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 7.853, 16.448, 16.956, 19.332, 20.135, 20.835, 21.645, 22.257, 22.696 and a characteristic peak at 25.879.
  • the present disclosure further provides Form I of the maleate salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 6.643, 7.853, 16.448, 16.956, 18.671, 19.332, 20.135, 20.835, 21.645 , 22.257, 22.696, 25.879 and 29.015 have characteristic peaks.
  • the present disclosure further provides Form II of the maleate salt of the compound represented by formula (I), whose X-ray powder diffraction pattern has characteristic peaks at 2 ⁇ angles of 7.440, 15.005, 15.503, 17.599, 18.763, 20.471 and 26.259 .
  • the present disclosure further provides Form II of the maleate salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 7.440, 8.724, 15.005, 15.503, 17.599, 18.136, 18.763, 20.471, 26.259 and a characteristic peak at 28.925.
  • the present disclosure further provides Form II of the maleate salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 7.440, 8.724, 15.005, 15.503, 17.599, 18.136, 18.763, 20.471, 22.600 , 23.556, 24.643, 26.259 and 28.925 have characteristic peaks.
  • the present disclosure further provides Form III of the maleate salt of the compound represented by formula (I), whose X-ray powder diffraction pattern has characteristic peaks at 2 ⁇ angles of 6.830, 16.440, 17.358, 19.295, 19.919, 20.946 and 26.340 .
  • the present disclosure further provides Form III of the maleate salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 6.830, 7.916, 16.440, 17.358, 19.295, 19.919, 20.946, 23.702, 25.820 and characteristic peaks at 26.340.
  • the present disclosure further provides Form III of the maleate salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 6.830, 7.916, 14.150, 16.440, 17.358, 19.295, 19.919, 20.946, 22.943 , 23.702, 25.820, 26.340 and 29.177 have characteristic peaks.
  • Another aspect of the present disclosure provides a method for preparing the I crystal form of the maleate salt of the compound represented by formula (I), the method comprising the following steps:
  • Another aspect of the present disclosure provides a method for preparing the II crystal form of the maleate salt of the compound represented by the formula (I), the method comprising the following steps:
  • Another aspect of the present disclosure provides a method for preparing the maleate salt form III of the compound represented by formula (I), the method comprising raising the temperature of the maleate salt form I of the compound represented by formula (I) to 110°C.
  • the present disclosure provides a hydrochloride salt of a compound represented by formula (I), wherein the molar ratio of the compound represented by formula (I) to hydrochloric acid is 1:2-2:1.
  • the present disclosure provides a hydrochloride salt of the compound represented by formula (I), wherein the molar ratio of the compound represented by formula (I) to hydrochloric acid is 1:1.
  • the present disclosure further provides a crystal form of the hydrochloride salt of the compound represented by formula (I), whose X-ray powder diffraction pattern has characteristic peaks at 2 ⁇ angles of 6.253, 12.578, 13.583, 18.151, 19.174, 20.027 and 26.978.
  • the molar ratio of the compound represented by formula (I) to hydrochloric acid is 1:2-2:1, preferably 1:1.
  • the present disclosure further provides a crystal form of the hydrochloride salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 6.253, 8.647, 11.210, 12.578, 13.583, 18.151, 19.174, 20.027, 26.676 and There is a characteristic peak at 26.978.
  • the molar ratio of the compound represented by formula (I) to hydrochloric acid is 1:2-2:1, preferably 1:1.
  • the present disclosure further provides a crystal form of the hydrochloride salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 6.253, 8.647, 11.210, 12.578, 13.583, 18.151, 19.174, 20.027, 24.105, There are characteristic peaks at 25.004, 25.375, 26.676 and 26.978.
  • the molar ratio of the compound represented by formula (I) to hydrochloric acid is 1:2-2:1, preferably 1:1.
  • the present disclosure further provides the b crystal form of the hydrochloride salt of the compound represented by formula (I), whose X-ray powder diffraction pattern has characteristic peaks at 2 ⁇ angles of 12.613, 15.871, 16.013, 17.839, 18.144, 19.186 and 20.074.
  • the molar ratio of the compound represented by formula (I) to hydrochloric acid is 1:2-2:1, preferably 1:1.
  • the present disclosure further provides the b crystal form of the hydrochloride salt of the compound represented by formula (I), the X-ray powder diffraction pattern of which is 12.613, 15.871, 16.013, 17.839, 18.144, 19.186, 20.074, 20.773, 21.186 and There is a characteristic peak at 26.977.
  • the mol ratio of the compound shown in the formula (I) and hydrochloric acid is 1:2-2:1, preferably 1:1.
  • the present disclosure further provides the b crystal form of the hydrochloride salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 12.613, 15.871, 16.013, 17.839, 18.144, 19.186, 20.074, 20.773, 21.186, There are characteristic peaks at 22.512, 24.181, 26.599 and 26.977.
  • the molar ratio of the compound represented by formula (I) to hydrochloric acid is 1:2-2:1, preferably 1:1.
  • the present disclosure further provides the crystal form c of the hydrochloride salt of the compound represented by formula (I), whose X-ray powder diffraction pattern has characteristic peaks at 2 ⁇ angles of 15.515, 17.137, 19.743, 20.471, 21.525, 23.442 and 25.987.
  • the present disclosure further provides the crystal form c of the hydrochloride salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 7.353, 13.066, 14.042, 15.515, 17.137, 19.743, 20.471, 21.525, 23.442 and There is a characteristic peak at 25.987.
  • the present disclosure further provides the crystal form c of the hydrochloride salt of the compound represented by formula (I), whose X-ray powder diffraction pattern at 2 ⁇ angles is 6.970, 7.353, 9.527, 13.066, 14.042, 15.515, 17.137, 19.743, 20.471, There are characteristic peaks at 21.525, 23.442, 25.987 and 29.252.
  • Another aspect of the present disclosure provides a method for preparing the a-crystal form of the hydrochloride salt of the compound represented by formula (I), the method comprising the following steps:
  • the molar ratio of the compound represented by formula (I) to hydrochloric acid is 1:2-2:1, preferably 1:1.
  • Another aspect of the present disclosure provides a method for preparing the b crystal form of the hydrochloride salt of the compound represented by formula (I), the method comprising the following steps:
  • compound shown in formula (I) is dissolved in at least one solvent selected from isopropanol, isopropyl ether, n-hexane, ethyl acetate and acetonitrile,
  • the molar ratio of the compound represented by formula (I) to hydrochloric acid is 1:2-2:1, preferably 1:1.
  • Another aspect of the present disclosure provides a method for preparing the crystal form c of the hydrochloride salt of the compound represented by formula (I), the method comprising the following steps:
  • the preparation method of the crystal form described in the present disclosure further comprises the steps of filtration, washing or drying.
  • the present disclosure also provides a pharmaceutical composition prepared from the pharmaceutically acceptable salt or the crystal form of the pharmaceutically acceptable salt of the compound represented by the aforementioned formula (I).
  • the present disclosure also provides a pharmaceutical composition, comprising the following components: i) a pharmaceutically acceptable salt of the compound represented by the aforementioned formula (I) or a crystalline form of a pharmaceutically acceptable salt, and ii) optionally selected from a pharmaceutically acceptable salt acceptable carrier, diluent or excipient.
  • the present disclosure also provides a method for preparing a pharmaceutical composition, comprising the steps of mixing the aforementioned component i) and component ii).
  • the present disclosure also provides a pharmaceutically acceptable salt of the compound represented by the aforementioned formula (I) or a crystal form of a pharmaceutically acceptable salt of the compound represented by the aforementioned formula (I), or the aforementioned composition, or prepared by the aforementioned method Use of the obtained composition in the preparation of estrogen receptor modulators.
  • the present disclosure also provides the pharmaceutically acceptable salt of the compound represented by the aforementioned formula (I) or the crystal form of the pharmaceutically acceptable salt of the aforementioned compound represented by the formula (I) or the aforementioned composition or prepared by the aforementioned method.
  • Use of the composition in the preparation of a medicament for the prevention and/or treatment of an estrogen receptor-mediated or dependent disease or condition preferably the estrogen receptor-mediated or dependent disease or condition is cancer, more Breast cancer, ovarian cancer, endometrial cancer, prostate cancer or uterine cancer are preferred, and breast cancer is most preferred.
  • the "2 ⁇ or 2 ⁇ angle" mentioned in this disclosure refers to the diffraction angle, and ⁇ is the Bragg angle, in degrees or degrees; the error range of each characteristic peak 2 ⁇ is ⁇ 0.20, which can be -0.20, -0.19, -0.18, -0.17, -0.16, -0.15, -0.14, -0.13, -0.12, -0.11, -0.10, -0.09, -0.08, -0.07, -0.06, -0.05, -0.04, -0.03, -0.02, -0.01 , 0.00, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20.
  • Crystallization in the present disclosure includes, but is not limited to, stirring crystallization, beating crystallization and volatile crystallization.
  • the preparation method of the crystal form described in the present disclosure also includes steps such as filtration and drying.
  • the drying temperature mentioned in the present disclosure is generally 25°C-100°C, preferably 40°C-70°C, and drying under normal pressure or under reduced pressure is possible.
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • the MS was measured with an Agilent 1200/1290 DAD-6110/6120 Quadrupole MS LC/MS instrument (manufacturer: Agilent, MS model: 6110/6120 Quadrupole MS).
  • HPLC High performance liquid chromatography
  • Chiral HPLC analysis was determined using an Agilent 1260 DAD high performance liquid chromatograph.
  • HPLC preparations used Waters 2545-2767, Waters 2767-SQ Detector2, Shimadzu LC-20AP and Gilson GX-281 preparative chromatographs.
  • XRPD is X-ray powder diffraction detection: the measurement is carried out with a BRUKER D8Discover X-ray diffractometer, specific information collected: Cu anode (40kV, 40mA), Cu-K ⁇ rays Scanning mode: ⁇ /2 ⁇ , scanning range (2 ⁇ range): 3 ⁇ 50°.
  • DSC is differential scanning calorimetry: METTLER TOLEDO DSC 3+ differential scanning calorimeter is used for measurement, the heating rate is 10°C/min, and the specific temperature range refers to the corresponding spectrum (mostly 25-300 or 25-350°C), nitrogen purge Speed 50mL/min.
  • TGA thermogravimetric analysis: METTLER TOLEDO TGA 2 type thermogravimetric analyzer was used for detection, the heating rate was 10°C/min, the specific temperature range was referred to the corresponding spectrum (mostly 25-300°C), and the nitrogen purge rate was 50mL/min.
  • DVS dynamic moisture adsorption: SMS DVS Advantage is used for detection. At 25°C, the humidity changes from 50%-95%-0%-95%-50% in steps of 10% (the last step is 5%) (the specific humidity range is The corresponding spectrum shall prevail, and most of the methods are listed here), and the judgment standard is that dm/dt is not more than 0.002%.
  • the solution refers to an aqueous solution.
  • reaction temperature is room temperature, which is 20°C to 30°C.
  • the monitoring of the reaction progress in the embodiment adopts thin layer chromatography (TLC), the developing solvent used in the reaction, the eluent system of the column chromatography used for purifying the compound and the developing solvent system of the thin layer chromatography method include: A: Dichloromethane/methanol system, B: n-hexane/ethyl acetate system, the volume ratio of the solvent is adjusted according to the polarity of the compound, and a small amount of basic or acidic reagents such as triethylamine and acetic acid can also be added for adjustment.
  • TLC thin layer chromatography
  • the second step (Z)-(1-(((5-(4,4,4-trifluoro-1-(3-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H -Indazol-5-yl)-2-phenylbut-1-en-1-yl)pyridin-2-yl)oxy)methyl)cyclopropyl)carbamate tert-butyl ester 1f
  • the third step (Z)-(1-(((5-(4,4,4-trifluoro-1-(3-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H -Indazol-5-yl)-2-phenylbut-1-en-1-yl)pyridin-2-yl)oxy)methyl)cyclopropyl)-1-amine 1g
  • the fourth step (E)-1-morpholinyl-4-((1-(((5-((Z)-4,4,4-trifluoro-1-(3-fluoro-1-(tetrafluoro) Hydro-2H-pyran-2-yl)-1H-indazol-5-yl)-2-phenylbut-1-en-1-yl)pyridin-2-yl)oxy)methyl)cyclopropane base)amino)but-2-en-1-one 1i
  • Test Example 1 Determination of Inhibitory Effect of Compound of Formula I on Estrogen Receptor Reporter Gene Activity
  • the purpose of this experiment is to test the inhibitory effect of the disclosed compounds on the activity of the estrogen receptor reporter gene, and to evaluate the in vitro activity of the compounds according to the IC 50 size.
  • MCF7 cells (ATCC, HTB-22) expressing the estrogen receptor response element-controlled luciferase reporter gene ERE-luc (synthesized by Jinweizhi Biotechnology Co., Ltd.) MCF7/ERE-luc containing 10% fetal bovine serum and 500 ⁇ g /ml G418 in MEM (GE Healthcare, SH30024.01) medium for culture.
  • ERE-luc the estrogen receptor response element-controlled luciferase reporter gene
  • MCF7/ERE-luc containing 10% fetal bovine serum and 500 ⁇ g /ml G418 in MEM (GE Healthcare, SH30024.01) medium for culture.
  • MCF7/ERE-luc cells were seeded in a 96-well plate at a density of 30,000 cells/well using incomplete MEM medium containing 10% activated carbon-treated fetal bovine serum (BioSun, BS-0004-500).
  • the 96-well plate was taken out, and 100 ⁇ l of ONE-Glo TM Luciferase Assay system (Promega, E6110) was added to each well to detect the activity of luciferase. After 3 minutes at room temperature until the cells were fully lysed, the multi-labeled microplate was used for enzyme labeling. The luminescence signal value was read by an instrument (PerkinElmer, VICTOR 3), and the IC50 value of the inhibitory activity of the compound was calculated according to the concentration of the compound and the luminescence signal value with Graphpad Prism software.
  • the inhibitory effect of the compounds in the present disclosure on the activity of the estrogen receptor reporter gene was determined by the above experiments, and the chemiluminescence signal value was plotted with the logarithmic concentration of the compound using Graghpad Prism, and the IC50 value of the compound of formula I was determined to be 1 nM .
  • the disclosed compounds have a significant inhibitory effect on the estrogen receptor reporter gene.
  • Test Example 2 Inhibitory effect of the compounds of the present disclosure on the proliferation of MCF7 cells
  • the purpose of this experiment is to determine the inhibitory activity of the disclosed compounds on the proliferation of MCF7 cells, and to evaluate the in vitro activity of the compounds according to the IC 50 size.
  • MCF7 cells (ATCC, HTB-22) were cultured in MEM (GE Healthcare, SH30024.01) complete medium containing 10% fetal bovine serum.
  • MEM GE Healthcare, SH30024.01
  • MCF7 cells were seeded in a 96-well plate at a density of 3,000 cells/well in complete medium, 100 ⁇ l of cell suspension per well, and placed in a cell incubator at 37°C and 5% CO 2 overnight. The next day, the medium was aspirated, and each well was replaced with 135 ⁇ l of MEM incomplete medium containing 2% fetal bovine serum. At the same time, 15 ⁇ l of different concentrations of the compounds to be tested prepared in incomplete medium were added to each well.
  • the final concentration of the compounds was 9 concentration points of 4-fold serial dilution starting from 100 nM, set up blank control containing 0.5% DMSO, and placed in a cell incubator at 37 °C, 5% CO 2 for 144 hours.
  • the IC 50 value of the compound was obtained as 0.5 nM, and the result showed that the compound of the present disclosure has a significant inhibitory effect on the proliferation of MCF7 cells.
  • Test Example 3 Experimental Biological Evaluation of Expression of ER ⁇ Mutant MCF7 Cell Proliferation Inhibition
  • the purpose of this experiment was to determine the inhibitory activity of the disclosed compounds on the proliferation of MCF7 cells expressing ER ⁇ mutants.
  • mutants ER ⁇ Y537S and ER ⁇ D538G of human estrogen receptor ⁇ (ER ⁇ ) protein were obtained by double-primer PCR using the cDNA (Accession No. NM000125) of the wild-type ESR1 gene as a template for site-directed mutagenesis.
  • the primer sequences used for mutation are as follows (the underlined nucleotides are the sites of mutation): Y537S: F-AAG AAC GTG GTG CCC CTC T C T GAC CTG CTG CTG GAG ATG; R-CAT CTC CAG CAG GTC A G A GAG GGG CAC CAC GTT CTT; D538G: F-AAC GTG GTG CCC CTC TAT G G C CTG CTG CTG GAG ATG CTG; R-CAG CAT CTC CAG CAG CAG G C C ATA GAG GGG CAC CAC GTT.
  • the cDNA of mutant ESR1 was cloned into the target lentiviral vector pCDH-CMV-MCS-EF1-Puro.
  • the lentiviral plasmids carrying the mutant ESR1 gene sequences and lentiviral packaging plasmids were then transfected into HEK-293T cells (ATCC, CRL-3216) by Lipofectamine 3000 Transfection Reagent (ThermoFisher Scientific, Cat# L3000075).
  • the virus-containing medium supernatant was filtered and ultracentrifuged to obtain the virus pellet, resuspended and dissolved with an appropriate amount of medium, added to MCF7 cells (ATCC, HTB-22), and added to the final concentration Incubate overnight with 8 ⁇ g/ml polybrene.
  • MCF7 cells ATCC, HTB-22
  • puromycin was added to the cell culture medium for resistance screening, and about two weeks later, the MCF7 cell line capable of stably expressing ER ⁇ Y537S and ER ⁇ D538G mutants was obtained.
  • MCF7 cells expressing ER ⁇ mutants were cultured in MEM (GE Healthcare, SH30024.01) complete medium containing 10% fetal bovine serum.
  • MEM GE Healthcare, SH30024.01
  • complete medium containing 10% fetal bovine serum.
  • cells were seeded in a 96-well plate at a density of 3,000 cells/well in complete medium, 100 ⁇ l of cell suspension per well, and cultured overnight in a cell incubator at 37°C, 5% CO 2 .
  • the medium was aspirated, and each well was replaced with 135 ⁇ l of MEM incomplete medium containing 2% fetal bovine serum.
  • 15 ⁇ l of different concentrations of the compounds to be tested prepared in incomplete medium were added to each well.
  • the final concentration of the compounds was 9 concentration points of 4-fold serial dilution starting from 100 nM, set up blank control containing 0.5% DMSO, and placed in a cell incubator at 37 °C, 5% CO 2 for 144 hours.
  • IC 50 value of the inhibitory activity of the compound, the IC 50 of the inhibitory effect of the compound of the present disclosure on the proliferation of MCF7 D538G cells expressing ER ⁇ mutant is 2nM
  • the IC 50 of the inhibitory effect on the proliferation of MCF7ER ⁇ Y537S cells expressing ER ⁇ mutant is 3nM
  • mice Female, were divided into 4 groups on average, and 9 mice were divided into 1 group. They were purchased from Jisijie Laboratory Animal Co., Ltd., and the animal production license number was SCXK (Shanghai) 2013-0006.
  • mice After a night of fasting, the mice were administered by intragastric administration respectively, and the administration volume was 0.2 ml/10 g, and the administration dose of the compound of formula I was 30 mg/kg.
  • the purpose of this experiment was to determine the covalent modification of the estrogen receptor ER ⁇ wild type and ER ⁇ Y537S mutant by the compounds of the present disclosure.
  • the ligand binding domain (LBD, ligand binding domain, aa296-554) of estrogen receptor ER ⁇ wild type and ER ⁇ Y537S mutant was expressed and purified from E. coli. Add 2 ⁇ M ER ⁇ wild-type or ER ⁇ Y537S mutant protein and 10 ⁇ M compound to a buffer containing 50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1 mM TCEP, 5% glycerol, mix well, and incubate at 4°C for 24 hours. High-resolution mass spectrometry detection.
  • test compounds have good covalent modification effect on ER ⁇ wild type or ER ⁇ Y537S mutant protein.
  • the product is defined as crystal form I
  • the XRPD spectrum is shown in Figure 1
  • the peak positions are shown in Table 1.
  • DSC spectrum shows endothermic peaks at 61.47°C and 115.48°C; TGA spectrum shows 2.81% weight loss at 25°C-140°C.
  • the obtained product was characterized by 1 H-NMR, and the nuclear magnetic data showed that the molar ratio of the main component and maleic acid in the salt was 1:1.
  • the product is defined as crystal form II, as shown in Figure 2, and the peak positions are shown in Table 2.
  • the DSC spectrum shows an endothermic peak at 136.02°C; the TGA spectrum shows a weight loss of 0.58% at 25°C-85°C, 3.73% at 85°C-150°C, and 4.39% at 150°C-210°C.
  • the crystal form of compound I represented by formula I was heated to 110° C. by DSC, and detected by X-ray powder diffraction.
  • the crystal form of the product was transformed and defined as crystal form III, as shown in FIG.
  • the obtained product was characterized by 1 H-NMR, and the nuclear magnetic data showed that the molar ratio of the main component and maleic acid in the salt was 1:1.
  • Embodiment 7 the preparation of hydrochloride a crystal form
  • Embodiment 8 the preparation of hydrochloride a crystal form
  • the compound represented by formula I (1.5 g, 2.36 mmol) was added to 10 mL of butanone, stirred, heated to 60 ° C, dissolved, and then added 0.5 mL of n-hexane clear liquid, cooled to 40 ° C, and slowly added dropwise 229.36 ⁇ L concentrated Hydrochloric acid (270.41 mg, 2.60 mmol, 35%) was dissolved, then cooled to room temperature and stirred at room temperature for 24 hours. No solid was precipitated.
  • the compound hydrochloride salt of formula I was added (Example 11), and gradually A white solid was precipitated, stirred for 16 hours, and gradually formed a white cloudy liquid, which was filtered, and the filter cake was collected and dried in vacuo to obtain the title product (1 g, yield: 63%).
  • the chloride ion content was 4.99% as detected by ion chromatography, indicating that the molar ratio of the main component and hydrochloric acid in the salt was 1:1.
  • the product was found to be crystal form a by X-ray powder diffraction.
  • the DSC spectrum shows an endothermic peak at 173.48°C; the TGA spectrum shows a weight loss of 2.81% at 25°C-170°C and a weight loss of 2.07% at 170°C-200°C.
  • DVS test shows that under normal storage conditions (ie, 25°C, 60%RH), the sample has a hygroscopic weight gain of about 0.89%; under accelerated experimental conditions (ie, 70%RH), the hygroscopic weight gain of the sample is about 1.02%; under extreme conditions At low temperature (90% RH), the hygroscopic weight gain was about 1.49%.
  • the crystal form was re-measured, and the crystal form did not change.
  • Embodiment 9 the preparation of hydrochloride a crystal form
  • the chloride ion content was 5.04% as detected by ion chromatography, indicating that the molar ratio of the main component and hydrochloric acid in the salt was 1:1.
  • the product is defined as crystal form b, the XRPD spectrum is shown in Figure 5, and the peak positions are shown in Table 5.
  • the DSC spectrum shows that the endothermic peak is not obvious, and the exothermic peak peak is 183.54°C;
  • DVS test shows that under normal storage conditions (ie, 25°C, 60% RH), the sample has a hygroscopic weight gain of about 0.84%; under accelerated experimental conditions (ie, 70% RH), the hygroscopic weight gain of the sample is about 0.99%; under extreme conditions At low temperature (90% RH), the hygroscopic weight gain was about 1.8%.
  • the desorption process and the adsorption process of this sample were basically coincident during the humidity change from 0% to 95% RH. After the DVS test, the crystal form was re-measured, and the crystal form did not change.
  • the compound represented by formula I (50 mg, 78.66 ⁇ mol) was added to 2 mL of acetonitrile, stirred, and dissolved, and 7.21 ⁇ L of concentrated hydrochloric acid (12M, 86.5 mmol, 35%) was slowly added dropwise, still dissolved, and stirred at room temperature for 24 hours. After that, a white cloudy liquid gradually formed, filtered, and the filter cake was collected and dried in vacuo to obtain the title product (40 mg, yield: 75.66%). The product was found to be crystal form b by X-ray powder diffraction.
  • the DSC spectrum shows that the endothermic peaks are 94.07°C and 147.06°C; the TGA spectrum shows that the weight loss is 5.30% at 25°C-120°C.
  • DVS test shows that under normal storage conditions (ie, 25°C, 60% RH), the sample has a hygroscopic weight gain of about 8.64%; under accelerated experimental conditions (ie, 70% RH), the hygroscopic weight gain of the sample is about 9.32%; under extreme conditions (90% RH), the hygroscopic weight gain was about 12.32%.
  • the desorption process and the adsorption process of this sample were basically coincident during the humidity change from 0% to 95% RH. After the DVS test, the crystal form was re-measured, and the crystal form did not change.
  • Embodiment 15 Experiment on influencing factors of hydrochloride crystal form a
  • Embodiment 16 Long-term accelerated stability experiment of hydrochloride crystal form a

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

L'invention concerne un sel pharmaceutiquement acceptable d'un dérivé d'indazole, une forme cristalline et un procédé de préparation associé. Plus particulièrement, la présente invention concerne une forme cristalline d'un sel pharmaceutiquement acceptable d'un composé représenté par la formule (I), et un procédé de préparation associé. La forme cristalline fournie du sel pharmaceutiquement acceptable du composé représenté par la formule (I) a une bonne stabilité et peut être mieux utilisée pour un traitement clinique.
PCT/CN2021/139125 2020-12-18 2021-12-17 Sel pharmaceutiquement acceptable de dérivé d'indazole, forme cristalline et procédé de préparation associé WO2022127904A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180084474.7A CN116615418A (zh) 2020-12-18 2021-12-17 一种吲唑类衍生物的药学上可接受的盐、结晶形式及其制备方法

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011510169.3 2020-12-18
CN202011510169 2020-12-18

Publications (1)

Publication Number Publication Date
WO2022127904A1 true WO2022127904A1 (fr) 2022-06-23

Family

ID=82058945

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/139125 WO2022127904A1 (fr) 2020-12-18 2021-12-17 Sel pharmaceutiquement acceptable de dérivé d'indazole, forme cristalline et procédé de préparation associé

Country Status (3)

Country Link
CN (1) CN116615418A (fr)
TW (1) TW202241879A (fr)
WO (1) WO2022127904A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107847498A (zh) * 2015-05-29 2018-03-27 卫材R&D管理有限公司 四取代的烯烃化合物及其用途
WO2018098251A1 (fr) * 2016-11-24 2018-05-31 Eisai R&D Management Co., Ltd. Composés d'alcène tétrasubstitués et leur utilisation
CN110300751A (zh) * 2016-11-24 2019-10-01 卫材 R&D 管理有限公司 四取代烯烃化合物及其用于治疗乳腺癌的用途
WO2020253762A1 (fr) * 2019-06-19 2020-12-24 江苏恒瑞医药股份有限公司 Dérivé d'indazole, son procédé de préparation et son application pharmaceutique

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107847498A (zh) * 2015-05-29 2018-03-27 卫材R&D管理有限公司 四取代的烯烃化合物及其用途
WO2018098251A1 (fr) * 2016-11-24 2018-05-31 Eisai R&D Management Co., Ltd. Composés d'alcène tétrasubstitués et leur utilisation
CN110300751A (zh) * 2016-11-24 2019-10-01 卫材 R&D 管理有限公司 四取代烯烃化合物及其用于治疗乳腺癌的用途
WO2020253762A1 (fr) * 2019-06-19 2020-12-24 江苏恒瑞医药股份有限公司 Dérivé d'indazole, son procédé de préparation et son application pharmaceutique

Also Published As

Publication number Publication date
TW202241879A (zh) 2022-11-01
CN116615418A (zh) 2023-08-18

Similar Documents

Publication Publication Date Title
JP6059723B2 (ja) 4−(8−メトキシ−1−((1−メトキシプロパン−2−イル)−2−(テトラヒドロ−2H−ピラン−4−イル)−1H−イミダゾ[4,5−c]キノリン−7−イル)−3,5−ジメチルイソオキサゾールおよびブロモドメイン阻害剤としてのその使用
WO2018045993A1 (fr) Forme cristalline, type de sel d'un dérivé de 2-hydro-pyrazole substitué et son procédé de préparation
KR20210131371A (ko) 1-(1-옥소-1,2-디하이드로이소퀴놀린-5-일)-5-(트리플루오로메틸)-n-(2-(트리플루오로메틸)피리딘-4-일)-1h-피라졸-4-카복사미드 일수화물의 결정질 형태
TW200916098A (en) Agent for lowering uric acid level
WO2020228635A1 (fr) Inhibiteur de kinase egfr et son utilisation dans la préparation d'un médicament anticancéreux
WO2015021894A1 (fr) Nouveau dérivé d'acide hydroximique et son application médicale
CN107501279B (zh) 呋喃并喹啉二酮类化合物及其医药用途
WO2021143843A1 (fr) Cristal d'inhibiteur double de pde3/pde4 et son utilisation
WO2022127904A1 (fr) Sel pharmaceutiquement acceptable de dérivé d'indazole, forme cristalline et procédé de préparation associé
WO2022068860A1 (fr) Forme cristalline d'un dérivé pyrrolo-hétérocyclique et son procédé de préparation
WO2021004487A1 (fr) Dérivé de thiazolidinedione et composition pharmaceutique le comprenant
CN114644616B (zh) 一种吲唑类衍生物的药学上可接受的盐、结晶形式及其制备方法
WO2019242642A1 (fr) Sel pharmaceutiquement acceptable et forme cristalline d'un inhibiteur d'otr et procédé de préparation associé
WO2016169417A1 (fr) Dérivés de purinyl-n-hydroxylpyrimidine-formamide, leur procédé de préparation et leur utilisation
CN114644615B (zh) 一种吲唑类衍生物的结晶形式及其制备方法
WO2023125947A1 (fr) Sel pharmaceutiquement acceptable de composé tétrahydroisoquinoléine, forme cristalline et utilisation associée
WO2023151660A1 (fr) Forme cristalline d'un antagoniste du récepteur p2x3 et son procédé de préparation
TW200927107A (en) ABCG2 inhibitor
WO2023185869A1 (fr) Forme cristalline de fumarate de dérivé pyrrolohétérocyclique et son procédé de préparation
WO2024067781A1 (fr) Sel pharmaceutiquement acceptable et forme cristalline de dérivé de tétrahydronaphtalène, et procédé de préparation
TWI777380B (zh) 2-吲哚啉螺環酮類化合物之結晶形式
WO2023116879A1 (fr) Forme cristalline d'agoniste du récepteur glp-1 et son procédé de préparation
WO2023284788A1 (fr) Sel pharmaceutiquement acceptable d'un agoniste du récepteur mor, polymorphe de celui-ci et utilisation associée
WO2023236877A1 (fr) Sel pharmaceutiquement acceptable d'un composé benzo[c]chromane, forme polymorphe et utilisation d'un sel pharmaceutiquement acceptable
WO2022048551A1 (fr) Polymorphe d'un inhibiteur de tyrosine kinase de bruton, son procédé de préparation et son utilisation

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: 21905829

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202180084474.7

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21905829

Country of ref document: EP

Kind code of ref document: A1