WO2023083292A1 - Salt of pyrrolidine compound, crystal form thereof, and preparation method therefor - Google Patents

Salt of pyrrolidine compound, crystal form thereof, and preparation method therefor Download PDF

Info

Publication number
WO2023083292A1
WO2023083292A1 PCT/CN2022/131313 CN2022131313W WO2023083292A1 WO 2023083292 A1 WO2023083292 A1 WO 2023083292A1 CN 2022131313 W CN2022131313 W CN 2022131313W WO 2023083292 A1 WO2023083292 A1 WO 2023083292A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
crystal form
ether
pharmaceutically acceptable
Prior art date
Application number
PCT/CN2022/131313
Other languages
French (fr)
Chinese (zh)
Inventor
胡越
刘成祥
冯爱娟
薛黎婷
杨桂梅
陈平
古鹏
唐任宏
任晋生
Original Assignee
先声药业有限公司
江苏先声药业有限公司
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 先声药业有限公司, 江苏先声药业有限公司 filed Critical 先声药业有限公司
Publication of WO2023083292A1 publication Critical patent/WO2023083292A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • 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
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the disclosure belongs to the field of medicine, and relates to pharmaceutically acceptable salts of pyrrolidine compounds, crystals of the pharmaceutically acceptable salts, and corresponding preparation methods and applications.
  • Estrogen (E2) and estrogen alpha receptor (ER ⁇ ) are important drivers of breast cancer development. More than 2/3 of breast cancer patients express ER transcription factors, and in most ER-positive patients, ER is still a key driver even in tumors that progress after early endocrine therapy, so ER is A major target for breast cancer therapy (Pharmacology & Therapeutics 186 (2016) 1–24).
  • the purpose of endocrine therapy is to reduce the activity of ER.
  • SERMs selective estrogen receptor modulators
  • tamoxifen tamoxifen
  • Aromatase inhibitors by inhibiting the conversion of androgen into estrogen, reduce the level of estrogen in the body; and selective estrogen receptor down-regulators, such as fulvestrant (fulvestrant), not only as ER Antagonists inhibit its activity and also induce ER protein degradation.
  • fulvestrant fulvestrant
  • Fulvestrant is the first and only SERD drug clinically approved for the treatment of postmenopausal patients with ER-positive, metastatic breast cancer after progression on tamoxifen or an aromatase inhibitor.
  • a number of research data show that patients treated with fulvestrant have not completely degraded ER in vivo.
  • intramuscular injection caused obvious reactions such as pain, swelling, and redness at the injection site, and the absorption was slow and the exposure in vivo was limited. And other characteristics limit its clinical application, so patients with ER-positive breast cancer urgently need new treatment options.
  • PCT/CN2021/093736 (application date May 14, 2021) describes a compound N-((S)-1-(3-fluoropropyl)pyrrolidin-3-yl)-6-((1S, 3R)-3-methyl-2-(2,2,2-trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indole-1- Base) pyridin-3-amine (compound of formula (I)), the compound of formula (I) is a selective estrogen receptor down-regulator, and the in vivo and in vitro models show that the compound of (I) has a wide range of inhibitory effects on ER-positive cells.
  • the pharmaceutically acceptable salt is selected from sulfate, phosphate, benzoate, succinate, adipate, fumarate, L-malate and citrate.
  • the present disclosure also provides the crystal form C of the adipate salt of the compound of the above formula (I).
  • the X-ray powder diffraction pattern represented by the diffraction angle 2 ⁇ of the crystal form C there are 4.46 ⁇ 0.2°, 6.30 ⁇ 0.2°, 6.30 ⁇ 0.2°, There are diffraction peaks at 8.90 ⁇ 0.2° and 19.10 ⁇ 0.2°.
  • the present disclosure also provides the crystal form B of the benzoate salt of the compound of the above formula (I).
  • the X-ray powder diffraction pattern represented by the diffraction angle 2 ⁇ of the crystal form B there are 7.19 ⁇ 0.2°, 10.01 ⁇ 0.2°, There are diffraction peaks at 11.39 ⁇ 0.2° and 18.78 ⁇ 0.2°.
  • the present disclosure also provides the crystal form A of the succinate salt of the compound of the above formula (I).
  • the X-ray powder diffraction pattern represented by the diffraction angle 2 ⁇ of the crystal form A at 7.13 ⁇ 0.2°, 9.30 ⁇ 0.2°, 11.29 There are diffraction peaks at ⁇ 0.2°, 15.51 ⁇ 0.2°, and 19.89 ⁇ 0.2°.
  • the present disclosure also provides a method for preparing a pharmaceutically acceptable salt of the compound of formula (I), the preparation method comprising the step of reacting the compound of formula (I) with a corresponding acid to form a salt.
  • the present disclosure also provides a method for preparing the above crystal form C, comprising the following steps:
  • the present disclosure also provides a method for preparing the above crystal form B, comprising the following steps:
  • the present disclosure also provides a method for preparing the above crystal form A, comprising the following steps:
  • the present disclosure also provides a pharmaceutical composition, which comprises a pharmaceutically acceptable salt of the compound of formula (I) or the above crystal form, and pharmaceutically acceptable auxiliary materials.
  • the present disclosure also provides the use of the pharmaceutically acceptable salt of the compound of the above formula (I), the above crystal form or the above pharmaceutical composition in the preparation of a drug for preventing or treating estrogen receptor-related diseases.
  • Fig. 1 is the NOESY spectrum of the compound of formula (I).
  • Fig. 2 is the XRPD spectrum of the crystal form C of adipate salt of the compound of formula (I).
  • Fig. 3 is the DSC spectrum of the crystal form C of adipate salt of the compound of formula (I).
  • Fig. 4 is the TGA spectrum of the crystal form C of adipate salt of the compound of formula (I).
  • Fig. 5 is the XRPD spectrum of the benzoic acid salt B crystal form of the compound of formula (I).
  • Fig. 6 is the DSC spectrum of the crystal form B of the benzoate salt of the compound of formula (I).
  • Fig. 7 is the TGA spectrum of the benzoic acid salt B crystal form of the compound of formula (I).
  • Fig. 8 is the XRPD spectrum of the crystal form A of succinate salt of the compound of formula (I).
  • Fig. 9 is the DSC spectrum of the crystal form A of the compound succinate of formula (I).
  • Fig. 10 is the TGA spectrum of the crystal form A of succinate salt of the compound of formula (I).
  • Fig. 11 is a three-dimensional structural ellipsoid diagram of crystal form A of succinate salt of the compound of formula (I).
  • Fig. 12 is a packing diagram of the unit cell structure of the crystal form A of the compound succinate of formula (I).
  • Fig. 13 is a picture of anti-tumor growth effect of compound succinate of formula (I) on MCF-7 xenograft subcutaneous xenograft tumor mouse model.
  • Fig. 14 is a graph showing the change in body weight of mice with MCF-7 xenogeneic xenograft tumors transplanted with succinate of the compound of formula (I).
  • Fig. 15 is the effect diagram of anti-tumor growth of compound succinate of formula (I) on T47D xenograft subcutaneous xenograft tumor mouse model.
  • Fig. 16 is a graph showing the change in body weight of T47D xenograft subcutaneously transplanted tumor mice with succinate of the compound of formula (I).
  • Fig. 17 The survival curve of the compound succinate of formula (I) on MCF-7 heterogeneous brain orthotopic tumor-bearing mice.
  • Fig. 18 is a graph showing the survival curve of the compound of formula (I) on MCF-7 xenogeneic brain orthotopic tumor-bearing mice.
  • Fig. 19 is a graph showing the change in body weight of MCF-7 heterogeneous brain orthotopic tumor-bearing mice by the compound of formula (I).
  • the present disclosure provides a pharmaceutically acceptable salt of a compound of formula (I) with excellent physical and chemical properties and/or pharmaceutical properties and crystals thereof,
  • the present disclosure provides a pharmaceutically acceptable salt of the compound represented by formula (I), wherein the pharmaceutically acceptable salt is selected from sulfate, phosphate, benzoate, succinate, adipate, fumaric acid salt, L-malate and citrate, preferably benzoate, succinate, adipate, fumarate, L-malate and citrate, more preferably benzoate, succinate salts and adipates.
  • the molar ratio of the compound of formula (I) to the acid molecule is about 0.5-2.
  • the molar ratio of the compound of formula (I) to the acid molecule is about 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3 , 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, and 2.0.
  • the molar ratio of the compound of formula (I) to the acid molecule is about 0.5, 1.0, 1.5 and 2.0.
  • the succinate salt of the compound of formula (I) above is represented by formula (II), wherein, x is selected from 0.5-2,
  • the benzoate salt of the compound of formula (I) above is represented by formula (III), wherein, y is selected from 0.5 to 2,
  • the adipate salt of the compound of formula (I) above is represented by formula (IV), wherein, z is selected from 0.5-2,
  • x, y, z are independently selected from 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, and 2.0.
  • x, y, z are independently selected from 0.8, 0.9, 1.0, 1.1 and 1.2.
  • x, y, z are independently selected from 1.0.
  • the present disclosure also provides a method for preparing a pharmaceutically acceptable salt of the compound of formula (I), comprising: the step of forming a salt of the compound shown in formula (I) with an acid, wherein the acid is selected from sulfuric acid, phosphoric acid, benzoic acid, succinic acid, Adipic acid, fumaric acid, L-malic acid and citric acid, preferably benzoic acid, succinic acid, adipic acid, fumaric acid, L-malic acid and citric acid, more preferably benzoic acid, succinic acid and adipic acid.
  • the acid is selected from sulfuric acid, phosphoric acid, benzoic acid, succinic acid, Adipic acid, fumaric acid, L-malic acid and citric acid, preferably benzoic acid, succinic acid, adipic acid, fumaric acid, L-malic acid and citric acid, more preferably benzoic acid, succinic acid and adipic acid.
  • the solvent used in the salt-forming reaction described in the present disclosure is selected from cyclohexane, n-heptane, toluene, chloroform, ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran, ethylene glycol dimethyl ether, acetone, methyl acetate , ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, preferably isopropyl ether, methyl tert-butyl ether, methanol, isopropanol and tetrahydrofuran at least one of the
  • the method for preparing a pharmaceutically acceptable salt of the compound of formula (I) further includes one or a combination of solvent volatilization, stirring crystallization, filtration, drying, etc.
  • the present disclosure also provides a crystalline form of the adipate salt of the compound of formula (I).
  • the crystalline form of the adipate salt of the compound of formula (I) is an anhydrate.
  • the present disclosure provides the C crystal form of the adipate salt of the compound of formula (I), the C crystal form, in the X-ray powder diffraction pattern represented by the diffraction angle 2 ⁇ , at 4.46 ⁇ 0.2°, 6.30 ⁇ 0.2°, There are diffraction peaks at 8.90 ⁇ 0.2° and 19.10 ⁇ 0.2°.
  • the crystal form C is at 4.46 ⁇ 0.2°, 6.30 ⁇ 0.2°, 7.03 ⁇ 0.2°, 8.90 ⁇ 0.2°, 16.45 ⁇ 0.2 °, 17.78 ⁇ 0.2°, 19.10 ⁇ 0.2° and 21.03 ⁇ 0.2° have diffraction peaks.
  • the crystal form C is at 4.46 ⁇ 0.2°, 6.30 ⁇ 0.2°, 7.03 ⁇ 0.2°, 8.90 ⁇ 0.2°, 10.65 ⁇ 0.2 °, 11.95 ⁇ 0.2°, 12.55 ⁇ 0.2°, 13.34 ⁇ 0.2°, 14.20 ⁇ 0.2°, 16.02 ⁇ 0.2°, 16.45 ⁇ 0.2°, 17.78 ⁇ 0.2°, 18.33 ⁇ 0.2°, 19.10 ⁇ 0.2°, 19.77 ⁇ 0.2 °, 20.12 ⁇ 0.2°, 21.03 ⁇ 0.2°, 21.62 ⁇ 0.2°, 24.30 ⁇ 0.2°, 25.97 ⁇ 0.2°, 27.39 ⁇ 0.2° and 27.98 ⁇ 0.2° have diffraction peaks.
  • the crystal form C is at 4.46 ⁇ 0.2°, 6.30 ⁇ 0.2°, 7.03 ⁇ 0.2°, 8.90 ⁇ 0.2°, 10.65 ⁇ 0.2 °, 11.95 ⁇ 0.2°, 12.55 ⁇ 0.2°, 12.96 ⁇ 0.2°, 13.34 ⁇ 0.2°, 14.20 ⁇ 0.2°, 15.20 ⁇ 0.2°, 16.02 ⁇ 0.2°, 16.45 ⁇ 0.2°, 17.78 ⁇ 0.2°, 18.33 ⁇ 0.2 °, 19.10 ⁇ 0.2°, 19.77 ⁇ 0.2°, 20.12 ⁇ 0.2°, 21.03 ⁇ 0.2°, 21.62 ⁇ 0.2°, 22.44 ⁇ 0.2°, 22.52 ⁇ 0.2°, 22.54 ⁇ 0.2°, 23.35 ⁇ 0.2°, 23.98 ⁇ 0.2
  • the X-ray powder diffraction pattern of the crystal form C is substantially consistent with that in FIG. 2 .
  • the DSC spectrum of the crystal form C has an endothermic peak at 149.89 ⁇ 5°C.
  • the DSC spectrum of Form C is substantially the same as that of FIG. 3 .
  • the present disclosure also provides a method for preparing the crystal form C of adipate salt of the compound of formula (I), comprising the step of reacting the compound of formula (I) with adipic acid to form a salt.
  • the preparation method of the above crystal form C includes the following steps:
  • the solvent in step (1-a) and step (2-a) is selected from cyclohexane, n-heptane, toluene, chloroform, diethyl ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran , at least one of ethylene glycol dimethyl ether, acetone, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, preferably isopropyl ether, At least one of methyl tert-butyl ether, methanol, isopropanol and tetrahydrofuran, more preferably isopropanol.
  • the molar ratio of the compound of formula (I) to adipic acid is 1:2-2:1, preferably 1:1.
  • the step (1-a) is performed under heating.
  • the step (1-a) is carried out at a heating temperature of about 40-80°C, preferably 60°C.
  • the step (4-a) is carried out at 15-30°C, preferably at 20-25°C.
  • the present disclosure also provides a crystalline form of the benzoate salt of the compound of formula (I).
  • the crystalline form of the benzoate salt of the compound of formula (I) is an anhydrate.
  • the present disclosure provides the crystal form B of the benzoate salt of the compound of formula (I).
  • the crystal form B is at 7.19 ⁇ 0.2°, 10.01 ⁇ 0.2°, 11.39 ⁇
  • the X-ray powder diffraction pattern represented by the diffraction angle 2 ⁇ of the B crystal form there are ° has a diffraction peak.
  • the X-ray powder diffraction pattern represented by the diffraction angle 2 ⁇ of the B crystal form at 7.19 ⁇ 0.2°, 10.01 ⁇ 0.2°, 11.39 ⁇ 0.2°, 18.78 ⁇ 0.2°, 19.01 ⁇ 0.2° , 19.66 ⁇ 0.2°, 19.91 ⁇ 0.2°, 20.09 ⁇ 0.2° have diffraction peaks.
  • the X-ray powder diffraction pattern represented by the diffraction angle 2 ⁇ of the B crystal form at 4.69 ⁇ 0.2°, 6.34 ⁇ 0.2°, 7.19 ⁇ 0.2°, 10.01 ⁇ 0.2°, 11.39 ⁇ 0.2 °, 18.78 ⁇ 0.2°, 19.01 ⁇ 0.2°, 19.66 ⁇ 0.2°, 19.91 ⁇ 0.2° and 20.09 ⁇ 0.2° have diffraction peaks.
  • the X-ray powder diffraction pattern represented by the diffraction angle 2 ⁇ of the B crystal form at 4.69 ⁇ 0.2°, 6.34 ⁇ 0.2°, 7.19 ⁇ 0.2°, 10.01 ⁇ 0.2°, 11.11 ⁇ 0.2 °, 11.39 ⁇ 0.2°, 14.78 ⁇ 0.2°, 18.78 ⁇ 0.2°, 19.01 ⁇ 0.2°, 19.25 ⁇ 0.2°, 19.66 ⁇ 0.2°, 19.91 ⁇ 0.2°, 20.09 ⁇ 0.2°, 20.80 ⁇ 0.2°, 21.66 ⁇ 0.2 ° has a diffraction peak.
  • the X-ray powder diffraction pattern represented by the diffraction angle 2 ⁇ of the B crystal form at 4.69 ⁇ 0.2°, 6.34 ⁇ 0.2°, 7.19 ⁇ 0.2°, 9.83 ⁇ 0.2°, 10.01 ⁇ 0.2° , 11.11 ⁇ 0.2°, 11.39 ⁇ 0.2°, 14.78 ⁇ 0.2°, 18.78 ⁇ 0.2°, 19.01 ⁇ 0.2°, 19.25 ⁇ 0.2°, 19.66 ⁇ 0.2°, 19.91 ⁇ 0.2°, 20.09 ⁇ 0.2°, 20.80 ⁇ 0.2° , There is a diffraction peak at 21.66 ⁇ 0.2°.
  • the X-ray powder diffraction pattern represented by the diffraction angle 2 ⁇ of the B crystal form at 4.69 ⁇ 0.2°, 4.91 ⁇ 0.2°, 6.34 ⁇ 0.2°, 7.19 ⁇ 0.2°, 9.37 ⁇ 0.2 °, 9.83 ⁇ 0.2°, 10.01 ⁇ 0.2°, 10.32 ⁇ 0.2°, 11.11 ⁇ 0.2°, 11.39 ⁇ 0.2°, 12.06 ⁇ 0.2°, 12.69 ⁇ 0.2°, 13.52 ⁇ 0.2°, 14.07 ⁇ 0.2°, 14.36 ⁇ 0.2 °, 14.78 ⁇ 0.2°, 17.30 ⁇ 0.2°, 18.78 ⁇ 0.2°, 19.01 ⁇ 0.2°, 19.25 ⁇ 0.2°, 19.66 ⁇ 0.2°, 19.91 ⁇ 0.2°, 20.09 ⁇ 0.2°, 20.80 ⁇ 0.2°, 21.15 ⁇ 0.2 °, 21.66 ⁇ 0.2°, 22.35 ⁇ 0.2°, 23.57 ⁇ 0.2°, 23.70 ⁇ 0.2°, 24.63 ⁇ 0.2°, 25.68 ⁇ 0.2°, 27.05 ⁇ 0.2° have diffraction peaks.
  • the X-ray powder diffraction pattern of the crystal form B is substantially consistent with FIG. 5 .
  • the DSC spectrum of the crystal form B has an endothermic peak at 150.08 ⁇ 5°C.
  • the DSC spectrum of Form B is substantially consistent with FIG. 6 .
  • the present disclosure also provides a preparation method of the crystal form B of the benzoate salt of the compound of formula (I), comprising the step of reacting the compound of formula (I) with benzoic acid to form a salt.
  • the preparation method of the above-mentioned B crystal form includes the following steps:
  • the solvent in step (1-b) is selected from cyclohexane, n-heptane, toluene, chloroform, diethyl ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran, ethylene glycol dimethyl ether , acetone, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, preferably isopropyl ether, methyl tert-butyl ether, At least one of methanol, isopropanol and tetrahydrofuran, more preferably isopropyl ether.
  • the molar ratio of the compound of formula (I) to benzoic acid is 1:2-2:1, preferably 1:1.
  • the step (1-b) is carried out at 15-30°C, preferably at 20-25°C.
  • the stirring in step (3-b) is carried out under heating conditions, preferably the heating temperature is about 40-68°C, more preferably 60-68°C.
  • the preparation method of the benzoate B crystal form of the compound of formula (I) also includes the following steps:
  • the solvent in step (5-b) is selected from at least one of isopropyl ether, methyl tert-butyl ether, methanol, isopropanol and tetrahydrofuran, preferably isopropyl ether.
  • the step (6-b) is carried out at 15-30°C, preferably at 20-25°C.
  • the present disclosure also provides a crystalline form of the succinate salt of the compound of formula (I).
  • the crystalline form of the succinate salt of the compound of formula (I) is an anhydrate.
  • the present disclosure provides the crystal form A of succinate salt of the compound of formula (I).
  • the crystal form A is at 7.13 ⁇ 0.2°, 9.30 ⁇ 0.2°, 11.29 ⁇ 0.2 °, 15.51 ⁇ 0.2°, 19.89 ⁇ 0.2° have diffraction peaks.
  • the crystal form A is at 7.13 ⁇ 0.2°, 9.30 ⁇ 0.2°, 11.29 ⁇ 0.2°, 15.51 ⁇ 0.2°, 17.10 ⁇ 0.2 °, 19.89 ⁇ 0.2°, 20.26 ⁇ 0.2°, 20.91 ⁇ 0.2°, 21.51 ⁇ 0.2°, 22.69 ⁇ 0.2°, 23.58 ⁇ 0.2°, 25.42 ⁇ 0.2° have diffraction peaks.
  • the crystal form A is at 5.01 ⁇ 0.2°, 7.13 ⁇ 0.2°, 9.30 ⁇ 0.2°, 10.09 ⁇ 0.2°, 11.29 ⁇ 0.2 °, 11.73 ⁇ 0.2°, 13.75 ⁇ 0.2°, 14.30 ⁇ 0.2°, 15.18 ⁇ 0.2°, 15.51 ⁇ 0.2°, 16.00 ⁇ 0.2°, 17.10 ⁇ 0.2°, 18.25 ⁇ 0.2°, 18.69 ⁇ 0.2°, 19.89 ⁇ 0.2 °, 20.26 ⁇ 0.2°, 20.91 ⁇ 0.2°, 21.12 ⁇ 0.2°, 21.51 ⁇ 0.2°, 22.69 ⁇ 0.2°, 23.05 ⁇ 0.2°, 23.58 ⁇ 0.2°, 24.58 ⁇ 0.2°, 25.42 ⁇ 0.2°, 26.66 ⁇ 0.2 °, 27.24 ⁇ 0.2° and 29.47 ⁇ 0.2° have diffraction peaks.
  • the crystal form A is at 5.01 ⁇ 0.2°, 7.13 ⁇ 0.2°, 9.30 ⁇ 0.2°, 10.09 ⁇ 0.2°, 11.29 ⁇ 0.2 °, 11.73 ⁇ 0.2°, 13.75 ⁇ 0.2°, 14.30 ⁇ 0.2°, 15.18 ⁇ 0.2°, 15.51 ⁇ 0.2°, 16.00 ⁇ 0.2°, 17.10 ⁇ 0.2°, 18.25 ⁇ 0.2°, 18.69 ⁇ 0.2°, 19.89 ⁇ 0.2 °, 20.26 ⁇ 0.2°, 20.91 ⁇ 0.2°, 21.12 ⁇ 0.2°, 21.51 ⁇ 0.2°, 22.69 ⁇ 0.2°, 23.05 ⁇ 0.2°, 23.58 ⁇ 0.2°, 24.58 ⁇ 0.2°, 25.42 ⁇ 0.2°, 26.66 ⁇ 0.2 °, 27.24 ⁇ 0.2°, 28.65 ⁇ 0.2°, 29.47 ⁇ 0.2°, 30.90 ⁇ 0.2°, 32.31 ⁇ 0.2°, 34.45 ⁇ 0.2° have diffraction peaks.
  • the X-ray powder diffraction pattern of Form A is substantially consistent with FIG. 8 .
  • the DSC spectrum of the crystal form A has an endothermic peak at 186.82 ⁇ 5°C.
  • the DSC spectrum of Form A is substantially consistent with FIG. 9 .
  • the present disclosure also provides a method for preparing the succinate A crystal form of the compound of formula (I), comprising the following steps:
  • the solvent in step (1-c) is selected from cyclohexane, n-heptane, toluene, chloroform, diethyl ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran, ethylene glycol dimethyl ether , acetone, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, preferably isopropyl ether, methyl tert-butyl ether, At least one of methanol, isopropanol and tetrahydrofuran, more preferably isopropyl ether.
  • the molar ratio of the compound of formula (I) to succinic acid is 1:2-2:1, preferably 1:1.
  • the step (3-c) is carried out at 15-30°C, preferably at 20-25°C.
  • the reaction time of the step (3-c) is 1-48 hours, preferably 3-24 hours, more preferably 8-16 hours.
  • preparation method of the succinate A crystal form of the compound of formula (I) also includes the following steps:
  • the solvent in step (5-c) is at least one selected from methanol, ethanol, isopropanol, tetrahydrofuran and water, preferably a mixed solvent of ethanol/water.
  • the volume ratio of ethanol/water is about 1:1 ⁇ 50:1, preferably 2:1 ⁇ 20:1; in some specific embodiments of the present disclosure, the volume ratio of ethanol/water is about 10 :1, 9:1, 8:1, 7:1, 6:1, 5:1 or 4:1.
  • the step (6-c) is carried out at 15-30°C, preferably at 20-25°C.
  • the stirring crystallization time of the step (6-c) is 0.5-48 hours, preferably 1-12 hours, more preferably 2-4 hours.
  • the present disclosure also provides a pharmaceutical composition, which comprises a pharmaceutically acceptable salt of the compound of formula (I) above or the crystal form above and pharmaceutically acceptable excipients.
  • the present disclosure also relates to the use of the pharmaceutically acceptable salt of the compound of formula (I) above or the above crystal form, or the above pharmaceutical composition in the preparation of drugs for preventing or treating estrogen receptor-related diseases.
  • the present disclosure relates to the use of a pharmaceutically acceptable salt of the compound of formula (I) above or the above crystal form, or the above pharmaceutical composition in the prevention or treatment of estrogen receptor-related diseases.
  • the present disclosure relates to a pharmaceutically acceptable salt of the compound of the above formula (I) or the above crystal form, or the above pharmaceutical composition for preventing or treating estrogen receptor-related diseases.
  • the present disclosure also relates to a method for treating estrogen receptor-related diseases, the method comprising administering to an individual a therapeutically effective dose of the pharmaceutically acceptable salt of the compound of formula (I) or the above-mentioned crystal form, the above-mentioned pharmaceutical composition, or the above-mentioned Pharmaceutically acceptable salts of the compound of formula (I) or pharmaceutical preparations of the above crystal forms.
  • the present disclosure also relates to the use of the pharmaceutically acceptable salt of the compound of the above formula (I) or the above crystal form, or the above pharmaceutical composition in the preparation of drugs for preventing or treating tumors.
  • the present disclosure relates to the use of a pharmaceutically acceptable salt of the compound of formula (I) or the above crystal form, or the above pharmaceutical composition in the prevention or treatment of tumors.
  • the present disclosure relates to a pharmaceutically acceptable salt of the compound of formula (I) or the above crystal form, or the above pharmaceutical composition for preventing or treating tumors.
  • the present disclosure also relates to a method for treating tumors, the method comprising administering to an individual a therapeutically effective dose of a pharmaceutically acceptable salt of the above-mentioned compound of formula (I) or the above-mentioned crystal form, the above-mentioned pharmaceutical composition or a compound containing the above-mentioned formula (I) of the present disclosure Pharmaceutically acceptable salts or pharmaceutical preparations of the above crystal forms.
  • the estrogen receptor-related diseases include but are not limited to tumors.
  • the estrogen receptor-related disease or tumor is breast cancer.
  • the estrogen receptor-related disease or tumor is ER-positive breast cancer.
  • the estrogen receptor-related disease or tumor is brain metastases from ER-positive breast cancer.
  • the individual is a cancer patient.
  • the individual is a breast cancer patient.
  • the individual is a patient with ER-positive breast cancer.
  • the individual is a patient with brain metastases from ER-positive breast cancer.
  • the pharmaceutically acceptable salts of the compounds of formula (I) and their crystals provided by the present disclosure have at least one of the advantages of good water solubility, high purity, good stability, and low hygroscopicity;
  • benzoate, succinate or adipate and crystal forms thereof of the compound of formula (I) can also effectively inhibit the generation of isomers, and/or have better thermal stability;
  • the pharmaceutically acceptable salt of the compound of formula (I) or its crystal form provided by the present disclosure also has at least one of the advantages of simple preparation method, mild crystallization conditions, high crystallinity, good solubility, good stability, and difficult to absorb moisture, etc., It is suitable for preparing the desired pharmaceutical composition.
  • the daily dosage of the pharmaceutically acceptable salt of the compound of formula (I) of the present disclosure is 0.01 mg/kg to 200 mg/kg body weight, preferably 0.05 mg/kg to 50 mg/kg body weight, more preferably 0.1 mg/kg to 30 mg/kg body weight. kg body weight, in single or divided doses.
  • Typical routes of administration of the pharmaceutically acceptable salts of the present disclosure or their crystalline forms or their polymorphs, or their polymorphic compositions, or their pharmaceutical compositions include, but are not limited to, oral, rectal, topical , inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous administration.
  • the pharmaceutical composition is in oral form.
  • the pharmaceutical compositions can be formulated by mixing the active compounds with pharmaceutically acceptable excipients well known in the art. These excipients enable the compounds of the present application to be formulated into tablets, pills, lozenges, dragees, capsules, liquids, gels, slurries, suspensions, etc. for oral administration to patients.
  • Solid oral compositions can be prepared by conventional methods of mixing, filling or tabletting. It can be obtained, for example, by mixing the active compound with solid excipients, optionally milling the resulting mixture, adding other suitable excipients if desired, and processing the mixture into granules to obtain tablets Or the core of the sugar coating.
  • Suitable auxiliary materials include but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, etc.
  • the room temperature mentioned in the present disclosure refers to 20 ⁇ 5°C.
  • the range “m ⁇ n” described in the present disclosure represents an abbreviated representation of any combination of real numbers between m and n, where both m and n are real numbers.
  • the numerical range “0.5 ⁇ 2” includes but is not limited to 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 or 2.0 .
  • the "X-ray powder diffraction pattern" described in this disclosure is measured using Cu-K ⁇ radiation.
  • the crystalline forms disclosed in the present disclosure are substantially pure crystals.
  • the term "substantially pure” as used herein means that the crystalline form has a purity of at least 85 wt%, preferably at least 90 wt%, more preferably at least 95 wt%.
  • the peak positions of their XRPD patterns are generally similar, and the relative intensity error may be large. It should also be pointed out that in the identification of mixtures, due to the decrease of content and other factors, some diffraction lines will be missing. At this time, there is no need to rely on all the diffraction peaks observed in high-purity samples, and even one diffraction peak may also affect the given is characteristic for a given crystal.
  • the "2 ⁇ or 2 ⁇ angle" mentioned in the present disclosure refers to the diffraction angle, ⁇ is the Bragg angle, and the unit is ° or degree; the error range of each diffraction peak 2 ⁇ is ⁇ 0.20°.
  • Differential scanning calorimetry or DSC in this disclosure refers to the measurement of the temperature difference and heat flow difference between the sample and the reference object during the heating or constant temperature of the sample to characterize all the physical changes related to thermal effects and Chemical changes, to obtain the phase transition information of the sample.
  • the drying temperature described in the present disclosure is generally 20°C-100°C, preferably 25°C-70°C, more preferably 40°C-60°C, and can be dried under normal pressure or reduced pressure. Preferably, drying is performed under reduced pressure.
  • treating means administering a compound or formulation described herein to improve or eliminate a disease or one or more symptoms associated with the disease, and includes:
  • prevention means administering a compound or formulation described herein to prevent a disease or one or more symptoms associated with the disease, and includes:
  • a disease or disease state from occurring in a subject (eg, a mammal), particularly when such a subject is susceptible to the disease state but has not yet been diagnosed as having the disease state.
  • terapéuticaally effective dose means (i) treating a particular disease, condition or disorder, (ii) alleviating, ameliorating or eliminating one or more symptoms of a particular disease, condition or disorder, or (iii) delaying the The amount of a compound of the disclosure required for the onset of one or more symptoms of a particular disease, condition or disorder as described.
  • the amount of a compound of the disclosure that constitutes a “therapeutically effective dose” will vary depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by one skilled in the art. Based on its own knowledge and this disclosure.
  • pharmaceutically acceptable excipients refers to those excipients that have no obvious stimulating effect on the organism and will not impair the biological activity and performance of the active compound. Suitable excipients are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
  • mammals include mammals and non-mammals.
  • mammals include, but are not limited to, any member of the class Mammalia: humans, non-human primates (such as chimpanzees and other apes and monkeys); livestock such as cattle, horses, sheep, goats, pigs; domesticated animals , such as rabbits, dogs, and cats; laboratory animals, including rodents, such as rats, mice, and guinea pigs.
  • non-human mammals include, but are not limited to, birds, fish, and the like.
  • Sample pan aluminum pan, non-sealed gland
  • Mobile phase A 0.01mol/L ammonium chloride solution (adjust the pH to 10.0 with ammonia water)
  • XRPD peak positions and/or intensities for a given crystalline form of the same compound will vary within error.
  • the 2 ⁇ values allow for an appropriate margin of error.
  • the margin of error is represented by " ⁇ ".
  • 5.92 ⁇ 0.2° 2 ⁇ represents a range of about 6.12 to 5.72.
  • suitable error ranges for XRPD diffraction angles (2 ⁇ ) may be about ⁇ 0.20°, ⁇ 0.15°, ⁇ 0.10° , ⁇ 0.05° or less.
  • the term “substantially identical” or “substantially as shown” means comprising at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 90% Or a pattern of diffraction peaks with at least 99% of the diffraction angles within a standard deviation of ⁇ 0.2° 2 ⁇ .
  • measurements of a DSC thermogram for a given crystalline form of the same compound will vary within a margin of error.
  • Single-peak peak values (expressed in degrees Celsius) allow for a modest margin of error.
  • the margin of error is represented by " ⁇ ".
  • thermal transition temperatures and melting points are typically within about ⁇ 5.0°C in consecutive analyses.
  • the peak value of "186.82 ⁇ 5°C” indicates that it is in the range of 181.82 to 191.82.
  • suitable error ranges for unimodal peaks may be ⁇ 5.0, ⁇ 4.0, ⁇ 3.0, ⁇ 2.0 or less.
  • Lux (lux, legal symbol lx) is the unit of luminance.
  • the luminous flux obtained by an object uniformly illuminated by light is 1 lumen on an area of 1 square meter, its illuminance is 1 lux.
  • NMR nuclear magnetic resonance
  • MS mass spectroscopy
  • Example 1 N-((S)(1-(3-fluoropropyl)pyrrolidin-3-yl)-6-((1S,3R)-3-methyl-2-(2,2,2 Synthesis of -trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indol-1-yl)pyridin-3-amine (formula (I) compound)
  • Step 3 Synthesis of (R)-1-(1H-indol-3-yl)-N-(2,2,2-trifluoroethyl)propane-2-amine
  • Step 4 (1S,3R)-1-(5-Bromopyridin-2-yl)-3-methyl-2-(2,2,2-trifluoroethyl)-2,3,4,9- Synthesis of Tetrahydro-1H-pyrido[3,4-b]indole
  • Step 5 N-((S)-1-(3-fluoropropyl)pyrrolidin-3-yl)-6-(((1S,3R)-3-methyl-2-(2,2,2 Synthesis of -trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indol-1-yl)pyridin-3-amine (compound of formula (I))
  • the NOESY spectrum (Fig. 1) shows that the methyl hydrogen on the 3-position of the compound of formula (I) and the hydrogen on the 1-position have a significant NOE effect, which proves that both are on the same side, and the pyridyl group on the 1-position and the hydrogen on the 3-position
  • the relative configuration of the methyl group on the 6-membered piperidine ring is trans, and the absolute configuration of the carbon atom at the 3-position is known as R, so the absolute configuration of the carbon atom at the 1-position is S.
  • Embodiment 2 the preparation of formula (I) compound sulfate
  • Embodiment 3 the preparation of formula (I) compound phosphate
  • Embodiment 4 the preparation of formula (I) compound fumarate
  • Embodiment 5 the preparation of formula (I) compound L-malate
  • Embodiment 6 the preparation of formula (I) compound citrate
  • Embodiment 7 the preparation of formula (I) compound adipate C crystal form
  • Embodiment 8 the preparation of formula (I) compound benzoate B crystal form
  • the XRPD spectrum of the crystalline solid is shown in Figure 5, its DSC spectrum is shown in Figure 6, and the TGA spectrum is shown in Figure 7. Its DSC endothermic peak peak is at 150.08 Around °C, the XRPD diffraction peak positions are shown in Table 2 below.
  • Embodiment 9 the preparation of formula (I) compound succinate
  • Embodiment 10 Preparation of crystal form A of compound succinate of formula (I)
  • Example 10-1 The succinate obtained in Example 9 was dissolved in methanol, and concentrated under reduced pressure to obtain a foamy solid. Weigh 500 mg of foamy solid, add ethanol (4.5 ml) and water (0.5 ml) into a 10 ml single-necked bottle, stir at room temperature, the solid dissolves, keep stirring for 2 hours to precipitate a solid, filter it with suction, and dry the obtained solid in vacuum at room temperature for 3 hours to obtain 0.22 g Crystal form A of the succinate salt of the compound of formula (I). The obtained solid was confirmed by X-ray powder diffraction spectrum. The XRPD spectrum of the crystalline solid is shown in Figure 8, its DSC spectrum is shown in Figure 9, and its TGA spectrum is shown in Figure 10. As shown in Table 3 below.
  • the crystal form A of compound succinate of formula (I) can also be prepared by the following examples: add 100 mg of the above-mentioned foamy solid and 500 ⁇ L of solvent (see Table 4) to a 5 ml centrifuge tube, stir at room temperature for 1-24 hours, and filter out solid and dried under vacuum at room temperature. See Table 4 for details.
  • Example solvent crystal form 10-2 isopropyl ether Form A 10-3 methyl tert-butyl ether Form A
  • Embodiment 11 Research on single crystal of compound succinate of formula (I)
  • a single crystal of the succinate of the compound of formula (I) can be prepared by the following method: take about 10 mg of the succinate of the compound of the formula (I) (prepared in Example 9) in a 3 mL sample bottle, add 1500 ⁇ L of isopropyl ether and 900 ⁇ L tetrahydrofuran, after ultrasonic dissolution, seal the bottle mouth with a parafilm and pierce a small hole in the parafilm with a needle, leave it at room temperature for 3 days, and precipitate crystals, and the obtained single crystal sample is detected by X-powder diffraction as A crystal form.
  • the single crystal belongs to the tetragonal crystal system and the space group I 4.
  • the stoichiometric formula is C 30 H 37 F 4 N 5 O 4 , and the calculated molecular weight of the asymmetric unit is 607.64.
  • Experimental method Accurately weigh about 10 mg of the sample to be tested, put it in a 10 ml centrifuge tube, add 1 ml of water, and shake in a water bath constant temperature shaker (37 ° C, 150 rpm). If the sample is completely dissolved, continue to add 10 mg of the sample and shake for about After 24 hours, take the supernatant, perform HPLC detection after appropriate dilution with acetonitrile, and determine its concentration by the external standard method.
  • Test example 2 investigation test of isomer impurity of formula (I) compound and pharmaceutically acceptable salt thereof
  • Test method formula (I) compound succinate (embodiment 10-1, A crystal form), formula (I) compound adipate (embodiment 7, C crystal form), formula (I) compound (implementation Example 1) are packaged with two layers of polyethylene and one layer of pharmaceutical composite film, focusing on the changes in the content of isomer impurities of the compound of formula (I) under conditions such as high temperature, light, high humidity, and acceleration.
  • the results are shown in Table 7 shown.
  • the isomer impurities are isomer 1 and isomer 2, respectively, and the structures are shown below. N.D means the impurity was not detected.
  • Test method the formula (I) compound succinate (embodiment 10-1, A crystal form), the formula (I) compound benzoate (embodiment 8, B crystal form), the formula (I) compound hexadiene Salt (embodiment 7, C crystal form) all adopts two layers of polyethylene, one layer of pharmaceutical composite film packing, investigates the change of impurity amount and impurity total amount under conditions such as high temperature, illumination, acceleration and high humidity, the result As shown in Table 8.
  • formula (I) compound succinate, formula (I) compound benzoate and formula (I) compound adipate all can control total impurity content preferably, especially formula (I) compound succinic acid
  • the salt and the adipate salt of the compound of formula (I) perform well in both aspects of total impurity content and impurity growth rate under conditions such as high temperature, light, acceleration and high humidity.
  • Test Example 4 Rat Pharmacokinetic Evaluation of Compounds of Formula (I) and Pharmaceutically Acceptable Salts thereof
  • MC methylcellulose
  • Acetonitrile was purchased from Merck (USA).
  • formula (I) compound benzoate, formula (I) compound succinate and formula (I) compound adipate have achieved more excellent PK characteristics, and have good oral bioavailability .
  • MCF-7, CAMA-1, HCC1500, T47D, and SK-BR-3 cells were purchased from ATCC, and EFM-19 and MCF-7 ESR1 Y537S mutant strains were purchased from Kebai Biotech.
  • the cell culture conditions refer to ATCC or Kebai product instructions, the medium is purchased from Invitrogen, and the serum is purchased from Gibico.
  • MCF-7 medium is DMEM+10%FBS+0.01mg/ml human insulin (Yisheng)+1% non-essential amino acid.
  • CAMA-1 medium is EMEM+10% FBS.
  • HCC1500 and EFM-19 medium are RPMI-1640+10% FBS.
  • T47D medium is RPMI-1640+10% HI-FBS+2 unit/mL bovine insulin (Solarbio).
  • MCF-7 ESR1 Y537S medium is MEM + 10% FBS + 1mM sodium pyruvate + 1% non-essential amino acids.
  • SK-BR-3 medium is McCoy's 5A+10% FBS. Fulvestrant was purchased from MCE.
  • the crystal form A of succinate salt of the compound of formula (I) was prepared according to the method in Example 10-1.
  • Use Echo650 (Beckman) to transfer 120nL of 3-fold diluted compound to the cell plate, add 40 ⁇ L of corresponding medium, after 7 days of compound treatment, add CellTiter-Glo (Promega) to detect cell viability, the cell well is used as 0% inhibition control, 0.1 ⁇ M fluorine Wells treated with fulvestrant were used as 100% inhibition control (wells treated with 2 ⁇ M fulvestrant for MCF-7 ESR1 Y537S were used as 100% inhibition control, and medium wells were used as 100% inhibition control for SK-BR-3). Using IDBS XLfit to perform 4-parameter fitting to calculate IC 50 and maximum inhibition rate data.
  • the succinate A crystal form of the compound of formula (I) showed strong proliferation inhibitory effects on different ER-positive breast cancer cell lines, with IC 50 ⁇ 1nM, and the IC 50 for proliferation inhibition of ESR1 Y537S mutant strains was 27.6nM, SK-BR-3 has no inhibitory effect on the proliferation of ER-negative cells at the highest concentration of 2 ⁇ M.
  • Test example 6 formula (I) compound succinate A crystal form to MCF-7 and MCF-7 ESR1 Y537S mutant strain ER protein
  • MCF-7 was purchased from ATCC, and the MCF-7 ESR1 Y537S mutant strain was purchased from Kebai Biotech.
  • the cell culture conditions refer to ATCC or Kebai product instructions, the medium is purchased from Invitrogen, and the serum is purchased from Gibico.
  • MCF-7 medium is DMEM+10%FBS+0.01mg/ml human insulin (Yisheng)+1% non-essential amino acid.
  • CAMA-1 medium is EMEM+10% FBS.
  • MCF-7 ESR1 Y537S medium is MEM + 10% FBS + 1mM sodium pyruvate + 1% non-essential amino acids. Fulvestrant was purchased from MCE.
  • the crystal form A of succinate salt of the compound of formula (I) was prepared according to the method in Example 10-1.
  • IRDye 800 goat anti-rabbit IgG antibody and IRDye 680 goat anti-mouse IgG antibody (LI-COR) and incubate in the dark for 45 minutes, wash off the secondary antibody, invert 1200 rpm and centrifuge for 1 minute to remove residual liquid, use Sapphire RGBNIR (Azure) detects 800nM and 680nM signals.
  • Use AzureSpot for fluorescence quantitative analysis calculate Chanel 800(ER)/Chanel 680(GAPDH) value, cell wells as 0% inhibition control, MCF-7 use 0.1 ⁇ M fulvestrant treatment wells as 100% inhibition control, MCF-7 ESR1 Y537S wells were treated with 2 ⁇ M fulvestrant as 100% degradation.
  • IDBS XLfit was used to perform 4-parameter fitting to calculate DC 50 and maximum degradation rate data.
  • Human breast cancer MCF-7 cells ECACC, 86012803
  • EMEM medium ATCC, Cat No.: 30-2003
  • Fetal bovine serum ExCell; Cat No.: FND500
  • Double antibody Gibco, Cat No.: 15240-062
  • Fulvestrant injection (Fulvestrant): AstraZeneca, 250mg/5mL/bottle
  • mice Female, 6-8 weeks old, weighing about 18-22 grams, were purchased from Shanghai Lingchang Biotechnology Co., Ltd., and the mice were raised in an SPF-grade environment. Each cage Separate exhaust ventilation was provided and all animals had free access to a standard certified commercial laboratory diet and water ad libitum.
  • Cell culture human breast cancer MCF-7 cell line was cultured in vitro, and the culture conditions were 10% fetal bovine serum and 1% double antibody in EMEM (cell culture medium), 37° C., 5% CO 2 incubator. Routine digestion with 0.25% trypsin-EDTA digestion solution was performed twice a week for subculture. When the confluence of the cells is 80%-90% and the number reaches the requirement, the cells are collected and counted.
  • Cell inoculation Inoculate 0.2ml/(containing 1 ⁇ 107 ) MCF-7 cell suspension (DPBS: Matrigel, volume ratio 1:1) subcutaneously on the right back of each mouse, and inoculate 17 ⁇ -estradiol tablets were subcutaneously inoculated two days before. On the 6th day after cell inoculation, when the average tumor volume reached 173.65 mm 3 , group administration began, and random grouping was administered according to the tumor volume. The day of group administration was Day 0 (Day 0).
  • the administration dose of formula (I) compound succinate (Example 10-1, A crystal form) is 0.3, 1, 3 or 10 mg/kg (calculated as free base), orally administered (PO), Once-daily dosing (QD) x21 times.
  • the dosage of fulvestrant (Fulvestrant) is 250 mg/kg, subcutaneous injection (SC), once a week (QW) x 4 times. 8 mice per group.
  • Tumor diameters were measured twice a week with vernier calipers.
  • Mouse body weights were measured twice a week.
  • TGI (%) [(1-(average tumor volume at the end of administration of a certain treatment group-average tumor volume at the beginning of administration of this treatment group)/(average tumor volume at the end of treatment of the solvent control group-at the beginning of treatment of the solvent control group Mean tumor volume)] x 100%.
  • mice have more than 10% weight loss, it has nothing to do with the dose of the test product formula (I) compound succinate, and the body weight of the mice in the test drug group is not particularly significant compared with the vehicle control group. Considering that the model is affected by the estrogen patch and may affect the state and body weight of the mice, the weight loss of these individual mice has nothing to do with the test product formula (I) compound succinate. No mouse died in each group of the experiment.
  • test results show that on the 20th day (Day 20) after the start of administration, subcutaneous injection of Fulvestrant 250 mg/kg into human breast cancer MCF-7 tumor-bearing mice once a week has a significant effect on the tumor growth of the MCF-7 xenograft tumor model. Weak inhibitory effect (P ⁇ 0.05).
  • formula (I) compound succinate shows significantly better than reference substance Fulvestrant (250mg/kg, SC) at a dose of 1mg/kg and above (PO, QD). , QW) antitumor activity (P ⁇ 0.0001).
  • Test Example 8 Pharmacodynamic study of compound succinate of formula (I) on human ER-positive breast cancer T47D xenograft subcutaneous xenograft tumor mouse model
  • T47D Human breast cancer xxT47D cells are T47D (ATCC, HTB-133) cells that have been passaged twice in vivo
  • RPMI-1640 culture medium Gibco, Cat No.: C22400500BT
  • Bovine insulin Yuanye, Cat.No.:11070-73-8
  • Double antibody Gibco, Cat No.: 15240-062
  • Fulvestrant injection (Fulvestrant): AstraZeneca, 250mg/5mL/bottle
  • mice Female, 6-8 weeks old, weighing about 18-22 grams, were purchased from Shanghai Lingchang Biotechnology Co., Ltd., and the mice were raised in an SPF-grade environment. Each cage Separate exhaust ventilation was provided and all animals had free access to a standard certified commercial laboratory diet and water ad libitum.
  • Cell culture Human breast cancer xxT47D cells were cultured in vitro. The culture conditions were RPMI-1640 medium plus 10% fetal bovine serum, 0.2 Units/mL bovine insulin, 1% double antibody, and cultured in a 5% CO 2 incubator at 37°C. Routine digestion with trypsin-EDTA was performed twice a week for passaging. When the confluence of the cells is 80%-90% and the number reaches the requirement, the cells are collected and counted.
  • Cell inoculation Inoculate 0.2mL (containing 1 ⁇ 107 ) xxT47D cell suspension (DPBS: Matrigel, volume ratio 1:1) subcutaneously on the right back of each mouse, and two days before cell inoculation Subcutaneous inoculation of 17 ⁇ -estradiol tablets. On the 6th day after cell inoculation, when the average volume of the tumor reached 137.09 mm 3 , group administration began, and the group was randomly assigned according to the tumor volume, and the day of group administration was Day 0 (Day 0).
  • DPBS Matrigel, volume ratio 1:1
  • the dosage of the compound succinate of formula (I) (Example 10-1, A crystal form) is 0.3, 1 or 3mg/kg, administered orally (PO), administered once a day (QD) x28 Second-rate.
  • the dosage of fulvestrant (Fulvestrant) is 250 mg/kg, subcutaneous injection (SC), once a week (QW) x 4 times. 8 mice per group.
  • Tumor diameters were measured twice a week with vernier calipers.
  • Mouse body weights were measured twice a week.
  • TGI (%) [(1-(average tumor volume at the end of administration of a certain treatment group-average tumor volume at the beginning of administration of this treatment group)/(average tumor volume at the end of treatment of the solvent control group-at the beginning of treatment of the solvent control group Mean tumor volume)] x 100%.
  • test results showed that on the 28th day (Day 28) after the start of administration, subcutaneous injection of 250mg/kg Fulvestrant to human breast cancer T47D tumor-bearing mice once a week showed a certain anti-tumor effect compared with the vehicle control group.
  • the effect of growth (P ⁇ 0.0001).
  • formula (I) compound succinate shows significantly better than reference substance Fulvestrant (250mg/kg, SC, QW) under the dose (PO, QD) of 1mg/kg and above. ) antitumor activity (P ⁇ 0.0001).
  • Human breast cancer MCF-7 cells ATCC, HTB-22
  • EMEM medium ATCC, Cat No.: 30-2003
  • Fetal bovine serum Gibco, Cat No.: 10099-141C
  • DPBS Hyclone, Cat. No.: SH30256.01
  • Fulvestrant injection (Fulvestrant): AstraZeneca, 250mg/5mL/bottle
  • mice Female, 6-7 weeks old, weighing about 17-23 grams, were purchased from Beijing Weitongda Biotechnology Co., Ltd., and the mice were raised in an SPF-grade environment, and each cage was sent separately All animals had free access to a standard certified commercial laboratory diet and water ad libitum.
  • Cell culture In vitro culture of human breast cancer MCF-7 cell line, the culture conditions are 10% fetal bovine serum, 10 ⁇ g/ml recombinant human insulin and 1% P/S in EMEM (cell culture medium), 37 ° C, 5% CO 2 incubators. Passage with routine digestion with 0.25% trypsin-EDTA once or twice a week. When the number reaches the requirement and the cells are in the logarithmic growth phase, the cells are collected and counted.
  • MCF-7 0.015ml/(containing 2 ⁇ 10 6 ) MCF-7 cell suspension was inoculated in the brain of each mouse (1mm in front of bregma, 2mm on the right side of the raphe, 3mm below the dura mater plane of the skull) , and 17 ⁇ -estradiol tablets were subcutaneously inoculated three days before cell inoculation. On the 8th day after cell inoculation, group administration began, and animals were randomly divided into groups according to body weight, and the day of grouping was Day 0.
  • the dosage of the compound succinate of formula (I) (Example 10-1, crystal form A) is 3, 10 or 30 mg/kg, administered orally (PO), administered once a day (QD), Dosing continued until Day 60.
  • the dosage of fulvestrant (Fulvestrant) was 250mg/kg, subcutaneously injected (SC), once a week (QW), and continued until Day 60. 8 mice per group.
  • mice were weighed twice a week.
  • the antitumor efficacy of compounds was evaluated by the median survival time.
  • Test Example 10 Research on the effect of compounds of formula (I) on the degradation of estrogen receptors in MCF7 cells
  • the purpose of this experiment is to measure the degradation activity of the compound of formula (I) on the endogenously expressed estrogen receptor in MCF7 cells, and evaluate the activity of the compound according to DC 50 and maximum degradation efficiency.
  • MCF7 cells (ATCC, HTB-22) were cultured with DMEM (Gibco, 11995-065) complete medium containing 10% fetal bovine serum. On the first day of the experiment, MCF7 cells were seeded in a 384-well plate at a density of 3000 cells/well using complete medium, and cultured in a 5% CO 2 cell incubator at 37°C.
  • DMEM Gibco, 11995-065
  • the compound to be tested was dissolved in DMSO with a storage concentration of 10mM, diluted with Echo 550 (Labcyte Inc.) and added to the cell culture plate, the initial concentration of the compound to be tested was 100nM, 3-fold serial dilution, 9 concentration points, set A blank control containing 0.5% DMSO was used, and a double-well control was set at each concentration point.
  • DMSO fetal sulfate
  • Echo 550 Echo 550
  • the wells treated with 0.1 ⁇ M fulvestrant were used as the 100% degradation control, and the degradation rate at each concentration point was calculated.
  • XlLfit was used to analyze the processing data, and the degradation activity DC50 and the maximum degradation rate Imax of the compound were calculated. See Table 15 for data analysis.
  • Table 15 formula (I) compound is to MCF7 intracellular estrogen receptor degradation activity
  • Test Example 11 Study on the inhibitory effect of the compound of formula (I) on the proliferation of MCF7 cells
  • the purpose of this experiment is to determine the inhibitory effect of the compound of formula (I) on the proliferation of MCF7 cells in vitro, and to evaluate the activity of the compound according to IC 50 .
  • MCF7 cells (ATCC, HTB-22) were cultured with DMEM (Gibco, 11995-065) complete medium containing 10% fetal bovine serum. On the first day of the experiment, MCF7 cells were seeded in a 384-well plate at a density of 500 cells/well using complete medium, and cultured overnight at 37° C. in a 5% CO 2 cell incubator. The next day, add the compound to be tested for drug treatment, and use Echo550 (Labcyte Inc.) to dilute the compound solution with a storage concentration of 10 mM and transfer it to each cell culture well.
  • DMEM Gibco, 11995-065
  • Echo550 Echo550
  • the initial concentration of the compound to be tested in the cells is 100nM, 3-fold serial dilution, 9 concentration points, a blank control containing 0.3% DMSO was set, and double-well controls were set at each concentration point.
  • Test Example 12 Apparent Solubility of Compound of Formula (I) in Phosphate Buffered Saline at pH 7.4
  • the compounds to be tested were prepared according to the methods described.
  • the control drug progesterone was purchased from Sigma.
  • Phosphate buffer with a pH value of 7.4 was prepared by our laboratory.
  • Acetonitrile and methanol were purchased from Fisher.
  • Other reagents were purchased from the market.
  • Test Example 13 The blood-brain barrier (BBB) penetration ability of the compound of formula (I) in rats
  • Drugs can pass through the blood-brain barrier of animals and have sufficient exposure in the brain is the key to the effectiveness of drugs on brain metastases. Therefore, by measuring drug concentrations in plasma and brain tissue after administration to animals, the effect of drugs in the brain Distribution, and then judge whether the drug can inhibit tumor growth in the brain orthotopic model.
  • SD female rats were purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd.
  • MC methylcellulose
  • acetonitrile was purchased from Merck (USA).
  • PBS phosphate buffered saline
  • Rat brain tissue samples were first homogenized with 4 times the mass volume of PBS homogenate. Take 20 ⁇ L of brain tissue homogenate sample, add 20 ⁇ L of blank mouse plasma to dilute and mix, then add 600 ⁇ L of acetonitrile solvent (including internal standard compound) to precipitate protein, vortex for 5 min, centrifuge (14,000 rpm) for 5 min, supernatant with Diluted 2 times with 0.1% (v/v) FA in water, and carried out quantitative detection on LC-MS/MS system (AB Sciex Triple Quad 6500+).
  • the compound of formula (I) exhibits excellent blood-brain barrier penetration ability, and the drug exposure in rat brain tissue is relatively high.
  • the BBB test results are as follows:
  • Test Example 14 Growth inhibition experiment of the compound of formula (I) on MCF-7 mouse subcutaneous tumor model
  • Human breast cancer MCF-7 cells ATCC, HTB-22
  • EMEM medium ATCC, Cat No.: 30-2003
  • Fetal bovine serum Gbico; Cat No.:1099-141C
  • Pen Strep (Pen Strep): Gibco, Cat No.: 15240-122
  • D-PBS phosphate buffered saline without calcium and magnesium ions
  • mice Female, 6-7 weeks old, weighing about 19-28 grams, were purchased from Beijing Weitongda Biotechnology Co., Ltd., and the mice were raised in an SPF-grade environment, and each cage was sent separately All animals had free access to a standard certified commercial laboratory diet and water ad libitum.
  • Cell culture In vitro culture of human breast cancer MCF-7 cell line, the culture conditions are 10% fetal bovine serum, 1% Pen Strep, 10 ⁇ g/ml recombinant human insulin in EMEM (cell culture medium), 37°C, 5% CO 2 incubator. Routine digestion with 0.25% trypsin-EDTA digestion solution once a week for passage. When the cell saturation is 80%-90% and the number reaches the requirement, collect the cells and count them.
  • Cell inoculation Subcutaneously inoculate 0.1ml/(containing 1 ⁇ 10 7 ) MCF-7 cell suspension (D-PBS: Matrigel, volume ratio 1:1) on the right back of each mouse, and inoculate In the first four days, 17 ⁇ -estradiol tablets were subcutaneously inoculated. On the 24th day after cell inoculation, the drugs were randomly divided into groups according to the tumor volume, and the grouping day was Day 0 (Day 0).
  • the dosage of the compound of formula (I) is 1, 3 or 10 mg/kg, administered orally (PO), administered once a day (QD) x 3 weeks. There were 8 mice in the vehicle group and 6 mice in the administration group.
  • Tumor diameters were measured twice a week with vernier calipers.
  • Mouse body weights were measured twice a week.
  • TGI (%) [(1-(average tumor volume at the end of administration of a certain treatment group-average tumor volume at the beginning of administration of this treatment group)/(average tumor volume at the end of treatment of the solvent control group-at the beginning of treatment of the solvent control group Mean tumor volume)] x 100%.
  • the compound of formula (I) has a significant inhibitory effect on tumor growth (P ⁇ 0.01) at 1 mg/kg, 3 mg/kg, or 10 mg/kg orally administered once a day, and has It has a good dose-response relationship, and it has the effect of shrinking tumors at the doses of 3mg/kg and 10mg/kg.
  • Oral administration of the compound of formula (I) once a day at 10 mg/kg has a significant inhibitory effect on tumor growth (P ⁇ 0.01), and has the effect of shrinking tumors.
  • the compound of formula (I) did not significantly affect the body weight of mice at the doses tried.
  • Test Example 15 Inhibitory experiment of the compound of formula (I) on the growth of mouse MCF-7 brain orthotopic tumor model
  • Human breast cancer MCF-7 cells ATCC, HTB-22
  • EMEM medium ATCC, Cat No.: 30-2003
  • Fetal bovine serum Gibco, Cat.No.:1099-141C
  • Pen Strep (Pen Strep): Gibco, Cat No.: 15240-122
  • Micro injection pump KDS, Cat No.: Legato130
  • mice Female, 6-8 weeks old, weighing about 17-29 grams, were purchased from Beijing Weitongda Biotechnology Co., Ltd., and the mice were raised in an SPF-grade environment, and each cage was sent separately All animals had free access to a standard certified commercial laboratory diet and water ad libitum.
  • Cell culture In vitro culture of human breast cancer MCF-7 cell line, the culture conditions are 10% fetal bovine serum, 1% Pen Strep, 10 ⁇ g/ml recombinant human insulin in EMEM (cell culture medium), 37°C, 5% CO 2 incubator. Routine digestion with 0.25% trypsin-EDTA digestion solution was performed twice a week for subculture. When the cell saturation is 80%-90% and the number reaches the requirement, collect the cells and count them.
  • MCF-7 15 ⁇ l/(containing 2 ⁇ 10 6 ) MCF-7 cell suspension was inoculated into the mouse skull using a brain localizer, a micro-injection pump and a miniature hand-held cranial drill, and 17 ⁇ was subcutaneously inoculated three days before cell inoculation - Estradiol tablets. On the 8th day after cell inoculation, mice were randomly divided into groups according to body weight for administration, and the grouping day was Day 0 (Day 0).
  • Fulvestrant (Fulvestrant, AstraZeneca) dosage is 250mg/kg, subcutaneous injection (SC), administration (QW) once a week, the dosage of formula (I) compound is 30mg/kg , administered orally (PO), administered once a day (QD).
  • SC subcutaneous injection
  • QW administration
  • PO administration
  • QD administration
  • mice There were 11 mice in the vehicle group and 8 mice in the administration group. All groups continued administration until the mice died, were euthanized due to poor condition or the experiment ended.
  • mice The body weight of the mice was measured twice a week, and the survival status of the mice was observed.
  • mice in the Fulvestrant group continued to decrease, and the survival status of the mice was not significantly different from that of the solvent control group (median survival period, vehicle 29 days in the control group and 29.5 days in the Fulvestrant group).
  • the mice in the compound group of formula (I) (30 mg/kg administered orally once a day) had a stable body weight and no abnormal state. Until the end of the experiment, the mice in the compound group of formula (I) did not die.
  • the compound It has a significant inhibitory effect on MCF-7 brain orthotopic tumor model mice, and the survival period of mice is significantly prolonged (P ⁇ 0.01).

Abstract

A pharmaceutically acceptable salt of the compound of formula (I), a crystal thereof, a preparation method therefor, and a use thereof. The pharmaceutically acceptable salt is selected from sulfate, phosphate, benzoate, succinate, adipate, fumarate, L-malate, and citrate, preferably benzoate, succinate, and adipate.

Description

吡咯烷类化合物的盐、晶型及其制备方法Salt, crystal form and preparation method of pyrrolidine compound
相关申请的交叉引用Cross References to Related Applications
本申请要求于2021年11月12日向中国国家知识产权局提交的第202111339152.0号中国专利申请的优先权和权益,所述申请公开的内容通过援引整体并入本文中。This application claims the priority and rights of Chinese Patent Application No. 202111339152.0 filed with the State Intellectual Property Office of China on November 12, 2021, the disclosure of which is incorporated herein by reference in its entirety.
技术领域technical field
本公开属于医药领域,涉及吡咯烷类化合物的可药用盐、所述可药用盐的结晶以及相应的制备方法和用途。The disclosure belongs to the field of medicine, and relates to pharmaceutically acceptable salts of pyrrolidine compounds, crystals of the pharmaceutically acceptable salts, and corresponding preparation methods and applications.
背景技术Background technique
雌激素(E2)及雌激素α受体(ERα)是乳腺癌发生发展的重要驱动因子。在乳腺癌患者中有超过2/3的患者表达ER转录因子,并且在大多数ER阳性患者中,即使经过早期的内分泌治疗后进展的肿瘤中,ER仍是一个关键的驱动因子,因此ER是乳腺癌治疗的一个主要靶点(Pharmacology&Therapeutics 186(2018)1–24)。内分泌治疗目的是降低ER活性,主要有三类,包括选择性雌激素受体调节剂(SERMs),比如他莫昔芬(tamoxifen),是ER的别构调节剂,同ER结合后抑制其转录活性;芳香化酶抑制剂(aromatase inhibitors,AIs),通过抑制雄激素转化为雌激素,减低体内雌激素水平;以及选择性雌激素受体下调剂,比如氟维司群(fulvestrant),不仅作为ER的拮抗剂抑制其活性,还具有诱导ER蛋白降解的作用。虽然内分泌治疗是雌激素受体阳性乳腺癌患者的首选,但是约有30%的治疗后病人会发生复发,并且几乎所有的转移性乳腺癌患者都会产生耐药而发生进展。Estrogen (E2) and estrogen alpha receptor (ERα) are important drivers of breast cancer development. More than 2/3 of breast cancer patients express ER transcription factors, and in most ER-positive patients, ER is still a key driver even in tumors that progress after early endocrine therapy, so ER is A major target for breast cancer therapy (Pharmacology & Therapeutics 186 (2018) 1–24). The purpose of endocrine therapy is to reduce the activity of ER. There are three main types, including selective estrogen receptor modulators (SERMs), such as tamoxifen (tamoxifen), which is an allosteric regulator of ER and inhibits its transcriptional activity after binding to ER. ; Aromatase inhibitors (aromatase inhibitors, AIs), by inhibiting the conversion of androgen into estrogen, reduce the level of estrogen in the body; and selective estrogen receptor down-regulators, such as fulvestrant (fulvestrant), not only as ER Antagonists inhibit its activity and also induce ER protein degradation. Although endocrine therapy is the first choice for patients with estrogen receptor-positive breast cancer, about 30% of patients will relapse after treatment, and almost all patients with metastatic breast cancer will develop drug resistance and progress.
临床上,约70-80%的乳腺癌检测雌激素受体(ER)呈阳性,这类乳腺癌细胞的增殖严重依赖ER,且50%的乳腺癌死亡病例均为该类分型。早期ER阳性乳腺癌预后较好,5年生存率超过90%。术后内分泌治疗(TAM或AI药物)的病人10年内约30%出现复发,但仍然可以接受标准的内分泌治疗。Clinically, about 70-80% of breast cancers are positive for estrogen receptor (ER), the proliferation of such breast cancer cells is heavily dependent on ER, and 50% of breast cancer deaths are of this type. Early ER-positive breast cancer has a better prognosis, with a 5-year survival rate of over 90%. About 30% of patients who received postoperative endocrine therapy (TAM or AI drugs) relapsed within 10 years, but they could still receive standard endocrine therapy.
氟维司群是首个也是唯一经临床批准用于他莫昔芬或芳香化酶抑制剂进展后治疗ER阳性、转移性乳腺癌的绝经后患者的SERD类药物。多项研究数据显示经氟维司群治疗的患者体内并未能完全实现ER的降解,此外肌肉注射方式造成的注射部位疼痛、肿胀、发红等明显反应,且吸收缓慢、体内暴露量受限等特点限制了其临床应用,因此ER阳性乳腺癌患者亟需新的治疗选择。Fulvestrant is the first and only SERD drug clinically approved for the treatment of postmenopausal patients with ER-positive, metastatic breast cancer after progression on tamoxifen or an aromatase inhibitor. A number of research data show that patients treated with fulvestrant have not completely degraded ER in vivo. In addition, intramuscular injection caused obvious reactions such as pain, swelling, and redness at the injection site, and the absorption was slow and the exposure in vivo was limited. And other characteristics limit its clinical application, so patients with ER-positive breast cancer urgently need new treatment options.
PCT/CN2021/093736(申请日2021年5月14日)描述了一种化合物N-((S)-1-(3-氟丙基)吡咯烷-3-基)-6-((1S,3R)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)吡啶-3-胺(式(I)化合物),所述式(I)化合物为选择性雌激素受体下调剂,体内外模型显示(I)化合物对ER阳性细胞具有广泛的抑制效果。PCT/CN2021/093736 (application date May 14, 2021) describes a compound N-((S)-1-(3-fluoropropyl)pyrrolidin-3-yl)-6-((1S, 3R)-3-methyl-2-(2,2,2-trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indole-1- Base) pyridin-3-amine (compound of formula (I)), the compound of formula (I) is a selective estrogen receptor down-regulator, and the in vivo and in vitro models show that the compound of (I) has a wide range of inhibitory effects on ER-positive cells.
发明内容Contents of the invention
本公开提供了式(I)化合物的可药用盐,The present disclosure provides pharmaceutically acceptable salts of compounds of formula (I),
Figure PCTCN2022131313-appb-000001
Figure PCTCN2022131313-appb-000001
其中,所述可药用盐选自硫酸盐、磷酸盐、苯甲酸盐、琥珀酸盐、己二酸盐、富马酸盐、L-苹果酸盐和柠檬酸盐。Wherein, the pharmaceutically acceptable salt is selected from sulfate, phosphate, benzoate, succinate, adipate, fumarate, L-malate and citrate.
本公开还提供了上述式(I)化合物的己二酸盐C晶型,所述C晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在4.46±0.2°、6.30±0.2°、8.90±0.2°、19.10±0.2°处有衍射峰。The present disclosure also provides the crystal form C of the adipate salt of the compound of the above formula (I). In the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the crystal form C, there are 4.46±0.2°, 6.30±0.2°, 6.30±0.2°, There are diffraction peaks at 8.90±0.2° and 19.10±0.2°.
本公开还提供了上述式(I)化合物的苯甲酸盐B晶型,所述B晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在7.19±0.2°、10.01±0.2°、11.39±0.2°、18.78±0.2°处有衍射峰。The present disclosure also provides the crystal form B of the benzoate salt of the compound of the above formula (I). In the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the crystal form B, there are 7.19±0.2°, 10.01±0.2°, There are diffraction peaks at 11.39±0.2° and 18.78±0.2°.
本公开还提供了上述式(I)化合物的琥珀酸盐A晶型,所述A晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在7.13±0.2°、9.30±0.2°、11.29±0.2°、15.51±0.2°、19.89±0.2°处有衍射峰。The present disclosure also provides the crystal form A of the succinate salt of the compound of the above formula (I). In the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the crystal form A, at 7.13±0.2°, 9.30±0.2°, 11.29 There are diffraction peaks at ±0.2°, 15.51±0.2°, and 19.89±0.2°.
本公开还提供了制备上述式(I)化合物的可药用盐的方法,所述制备方法包括了式(I)所示化合物与相应酸成盐反应的步骤。The present disclosure also provides a method for preparing a pharmaceutically acceptable salt of the compound of formula (I), the preparation method comprising the step of reacting the compound of formula (I) with a corresponding acid to form a salt.
本公开还提供了制备上述C晶型的方法,包含以下步骤:The present disclosure also provides a method for preparing the above crystal form C, comprising the following steps:
(1-a)将己二酸和溶剂混合;(1-a) mixing adipic acid and a solvent;
(2-a)将式(I)化合物和溶剂混合;(2-a) mixing the compound of formula (I) and a solvent;
(3-a)混合步骤(1-a)的混合物与步骤(2-a)的混合物;(3-a) mixing the mixture of step (1-a) and the mixture of step (2-a);
(4-a)将混合了所述已二酸和所述式(I)化合物的所得混合物搅拌,析晶;(4-a) Stir the resulting mixture mixed with the adipic acid and the compound of formula (I), and crystallize;
(5-a)过滤出固体,收集滤饼,干燥。(5-a) The solid was filtered out, and the filter cake was collected and dried.
本公开还提供了制备上述B晶型的方法,包含以下步骤:The present disclosure also provides a method for preparing the above crystal form B, comprising the following steps:
(1-b)将式(I)化合物和溶剂混合;(1-b) mixing the compound of formula (I) and a solvent;
(2-b)向步骤(1-b)的混合物中加入苯甲酸;(2-b) adding benzoic acid to the mixture of step (1-b);
(3-b)将混合了所述式(I)化合物和所述苯甲酸的所得混合物搅拌,析晶;(3-b) Stir the resulting mixture mixed with the compound of formula (I) and the benzoic acid, and crystallize;
(4-b)过滤出并收集固体。(4-b) The solid was filtered off and collected.
本公开还提供了制备上述A晶型的方法,包含以下步骤:The present disclosure also provides a method for preparing the above crystal form A, comprising the following steps:
(1-c)将式(I)化合物和溶剂混合;(1-c) mixing the compound of formula (I) and a solvent;
(2-c)向步骤(1-c)的混合物中加入琥珀酸;(2-c) adding succinic acid to the mixture of step (1-c);
(3-c)将混合了所述式(I)化合物和所述琥珀酸的所得混合物搅拌,析出固体;(3-c) stirring the mixture obtained by mixing the compound of formula (I) and the succinic acid to precipitate a solid;
(4-c)过滤出并收集固体。(4-c) The solid was filtered off and collected.
本公开还提供了一种药物组合物,其包含上述式(I)化合物的可药用盐或上述晶型,以及药学上可接受的辅料。The present disclosure also provides a pharmaceutical composition, which comprises a pharmaceutically acceptable salt of the compound of formula (I) or the above crystal form, and pharmaceutically acceptable auxiliary materials.
本公开还提供了上述式(I)化合物的可药用盐、上述晶型或上述药物组合物在制备用于预防或者治疗雌激素受体相关疾病的药物中的用途。The present disclosure also provides the use of the pharmaceutically acceptable salt of the compound of the above formula (I), the above crystal form or the above pharmaceutical composition in the preparation of a drug for preventing or treating estrogen receptor-related diseases.
附图说明Description of drawings
图1为式(I)化合物NOESY图谱。Fig. 1 is the NOESY spectrum of the compound of formula (I).
图2为式(I)化合物的己二酸盐C晶型的XRPD图谱。Fig. 2 is the XRPD spectrum of the crystal form C of adipate salt of the compound of formula (I).
图3为式(I)化合物的己二酸盐C晶型的DSC图谱。Fig. 3 is the DSC spectrum of the crystal form C of adipate salt of the compound of formula (I).
图4为式(I)化合物的己二酸盐C晶型的TGA图谱。Fig. 4 is the TGA spectrum of the crystal form C of adipate salt of the compound of formula (I).
图5为式(I)化合物的苯甲酸盐B晶型的XRPD图谱。Fig. 5 is the XRPD spectrum of the benzoic acid salt B crystal form of the compound of formula (I).
图6为式(I)化合物的苯甲酸盐B晶型的DSC图谱。Fig. 6 is the DSC spectrum of the crystal form B of the benzoate salt of the compound of formula (I).
图7为式(I)化合物的苯甲酸盐B晶型的TGA图谱。Fig. 7 is the TGA spectrum of the benzoic acid salt B crystal form of the compound of formula (I).
图8为式(I)化合物琥珀酸盐A晶型的XRPD图谱。Fig. 8 is the XRPD spectrum of the crystal form A of succinate salt of the compound of formula (I).
图9为式(I)化合物琥珀酸盐A晶型的DSC图谱。Fig. 9 is the DSC spectrum of the crystal form A of the compound succinate of formula (I).
图10为式(I)化合物琥珀酸盐A晶型的TGA图谱。Fig. 10 is the TGA spectrum of the crystal form A of succinate salt of the compound of formula (I).
图11为式(I)化合物琥珀酸盐A晶型的立体结构椭球图。Fig. 11 is a three-dimensional structural ellipsoid diagram of crystal form A of succinate salt of the compound of formula (I).
图12为式(I)化合物琥珀酸盐A晶型的晶胞结构堆积图。Fig. 12 is a packing diagram of the unit cell structure of the crystal form A of the compound succinate of formula (I).
图13式(I)化合物琥珀酸盐在MCF-7异种皮下移植瘤小鼠模型上的抗肿瘤生长效果图。Fig. 13 is a picture of anti-tumor growth effect of compound succinate of formula (I) on MCF-7 xenograft subcutaneous xenograft tumor mouse model.
图14式(I)化合物琥珀酸盐对MCF-7异种皮下移植瘤小鼠体重变化图。Fig. 14 is a graph showing the change in body weight of mice with MCF-7 xenogeneic xenograft tumors transplanted with succinate of the compound of formula (I).
图15式(I)化合物琥珀酸盐在T47D异种皮下移植瘤小鼠模型上的抗肿瘤生长的效果图。Fig. 15 is the effect diagram of anti-tumor growth of compound succinate of formula (I) on T47D xenograft subcutaneous xenograft tumor mouse model.
图16式(I)化合物琥珀酸盐对T47D异种皮下移植瘤小鼠体重变化图。Fig. 16 is a graph showing the change in body weight of T47D xenograft subcutaneously transplanted tumor mice with succinate of the compound of formula (I).
图17式(I)化合物琥珀酸盐对MCF-7异种脑原位荷瘤小鼠生存曲线图。Fig. 17 The survival curve of the compound succinate of formula (I) on MCF-7 heterogeneous brain orthotopic tumor-bearing mice.
图18为式(I)化合物对MCF-7异种脑原位荷瘤小鼠生存曲线图。Fig. 18 is a graph showing the survival curve of the compound of formula (I) on MCF-7 xenogeneic brain orthotopic tumor-bearing mice.
图19为式(I)化合物对MCF-7异种脑原位荷瘤小鼠体重变化图。Fig. 19 is a graph showing the change in body weight of MCF-7 heterogeneous brain orthotopic tumor-bearing mice by the compound of formula (I).
具体实施方式Detailed ways
本公开提供一种理化性质和/或药学性质优异的式(I)化合物的可药用盐及其结晶,The present disclosure provides a pharmaceutically acceptable salt of a compound of formula (I) with excellent physical and chemical properties and/or pharmaceutical properties and crystals thereof,
Figure PCTCN2022131313-appb-000002
Figure PCTCN2022131313-appb-000002
本公开提供了式(I)所示化合物的可药用盐,其中,所述可药用盐选自硫酸盐、磷酸盐、苯甲酸盐、琥珀酸盐、己二酸盐、富马酸盐、L-苹果酸盐和柠檬酸盐,优选苯甲酸盐、琥珀酸盐、己二酸盐、富马酸盐、L-苹果酸盐和柠檬酸盐,更优选苯甲酸盐、琥珀酸盐和己二酸盐。The present disclosure provides a pharmaceutically acceptable salt of the compound represented by formula (I), wherein the pharmaceutically acceptable salt is selected from sulfate, phosphate, benzoate, succinate, adipate, fumaric acid salt, L-malate and citrate, preferably benzoate, succinate, adipate, fumarate, L-malate and citrate, more preferably benzoate, succinate salts and adipates.
在一些实施方案中,所述式(I)化合物的可药用盐中,式(I)化合物与酸分子的摩尔比约为0.5~2。In some embodiments, in the pharmaceutically acceptable salt of the compound of formula (I), the molar ratio of the compound of formula (I) to the acid molecule is about 0.5-2.
在一些实施方案中,所述式(I)化合物的可药用盐中,式(I)化合物与酸分子的摩尔比约为0.5、0.6、0.7、0.8、0.9、1.0、1.1、1.2、1.3、1.4、1.5、1.6、1.7、1.8、1.9和2.0。In some embodiments, in the pharmaceutically acceptable salt of the compound of formula (I), the molar ratio of the compound of formula (I) to the acid molecule is about 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3 , 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, and 2.0.
在一些实施方案中,所述式(I)化合物的可药用盐中,式(I)化合物与酸分子的摩尔比约为0.5、1.0、1.5和2.0。In some embodiments, in the pharmaceutically acceptable salt of the compound of formula (I), the molar ratio of the compound of formula (I) to the acid molecule is about 0.5, 1.0, 1.5 and 2.0.
在一些实施方案中,上述式(I)化合物的琥珀酸盐如式(II)所示,其中,x选自0.5~2,In some embodiments, the succinate salt of the compound of formula (I) above is represented by formula (II), wherein, x is selected from 0.5-2,
Figure PCTCN2022131313-appb-000003
Figure PCTCN2022131313-appb-000003
在一些实施方案中,上述式(I)化合物的苯甲酸盐如式(III)所示,其中,y选自0.5~2,In some embodiments, the benzoate salt of the compound of formula (I) above is represented by formula (III), wherein, y is selected from 0.5 to 2,
Figure PCTCN2022131313-appb-000004
Figure PCTCN2022131313-appb-000004
在一些实施方案中,上述式(I)化合物的己二酸盐如式(IV)所示,其中,z选自0.5~2,In some embodiments, the adipate salt of the compound of formula (I) above is represented by formula (IV), wherein, z is selected from 0.5-2,
Figure PCTCN2022131313-appb-000005
Figure PCTCN2022131313-appb-000005
在一些实施方案中,x、y、z独立地选自0.5、0.6、0.7、0.8、0.9、1.0、1.1、1.2、1.3、1.4、1.5、1.6、1.7、1.8、1.9和2.0。In some embodiments, x, y, z are independently selected from 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, and 2.0.
在一些实施方案中,x、y、z独立地选自0.8、0.9、1.0、1.1和1.2。In some embodiments, x, y, z are independently selected from 0.8, 0.9, 1.0, 1.1 and 1.2.
在一些实施方案中,x、y、z独立地选自1.0。In some embodiments, x, y, z are independently selected from 1.0.
本公开还提供了制备上述式(I)化合物可药用盐的方法,包括:式(I)所示化合物与酸成盐的步骤,所述酸选自硫酸、磷酸、苯甲酸、琥珀酸、己二酸、富马酸、L-苹果酸和柠檬酸,优选苯甲酸、琥珀酸、己二酸、富马酸、L-苹果酸和柠檬酸,更优选苯甲酸、琥珀酸和己二酸。The present disclosure also provides a method for preparing a pharmaceutically acceptable salt of the compound of formula (I), comprising: the step of forming a salt of the compound shown in formula (I) with an acid, wherein the acid is selected from sulfuric acid, phosphoric acid, benzoic acid, succinic acid, Adipic acid, fumaric acid, L-malic acid and citric acid, preferably benzoic acid, succinic acid, adipic acid, fumaric acid, L-malic acid and citric acid, more preferably benzoic acid, succinic acid and adipic acid.
本公开所述成盐反应所用溶剂选自环己烷、正庚烷、甲苯、氯仿、乙醚、异丙醚、甲基叔丁基醚、四氢呋喃、乙二醇二甲醚、丙酮、乙酸甲酯、乙酸乙酯、乙酸异丙酯、乙酸丁酯、甲醇、乙醇、异丙醇、正丁醇和乙腈中的至少一种,优选异丙醚、甲基叔丁基醚、甲醇、异丙醇和四氢呋喃中的至少一种。The solvent used in the salt-forming reaction described in the present disclosure is selected from cyclohexane, n-heptane, toluene, chloroform, ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran, ethylene glycol dimethyl ether, acetone, methyl acetate , ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, preferably isopropyl ether, methyl tert-butyl ether, methanol, isopropanol and tetrahydrofuran at least one of the
在可选实施方案中,制备上述式(I)化合物可药用盐的方法还包括挥发溶剂或搅拌析晶、过滤、干燥等中的一种或其组合的步骤。In an optional embodiment, the method for preparing a pharmaceutically acceptable salt of the compound of formula (I) further includes one or a combination of solvent volatilization, stirring crystallization, filtration, drying, etc.
本公开还提供了式(I)化合物的己二酸盐的晶型。The present disclosure also provides a crystalline form of the adipate salt of the compound of formula (I).
在一些实施方案中,式(I)化合物的己二酸盐的晶型为无水合物。In some embodiments, the crystalline form of the adipate salt of the compound of formula (I) is an anhydrate.
本公开提供了上述式(I)化合物己二酸盐的C晶型,所述C晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在4.46±0.2°、6.30±0.2°、8.90±0.2°和19.10±0.2°处有衍射峰。The present disclosure provides the C crystal form of the adipate salt of the compound of formula (I), the C crystal form, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ, at 4.46±0.2°, 6.30±0.2°, There are diffraction peaks at 8.90±0.2° and 19.10±0.2°.
在一些实施方案中,所述C晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在4.46±0.2°、6.30±0.2°、7.03±0.2°、8.90±0.2°、16.45±0.2°、17.78±0.2°、19.10±0.2°和21.03±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ, the crystal form C is at 4.46±0.2°, 6.30±0.2°, 7.03±0.2°, 8.90±0.2°, 16.45±0.2 °, 17.78±0.2°, 19.10±0.2° and 21.03±0.2° have diffraction peaks.
在一些实施方案中,所述C晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在4.46±0.2°、6.30±0.2°、7.03±0.2°、8.90±0.2°、10.65±0.2°、11.95±0.2°、12.55±0.2°、13.34±0.2°、14.20±0.2°、16.02±0.2°、16.45±0.2°、17.78±0.2°、18.33±0.2°、19.10±0.2°、19.77±0.2°、20.12±0.2°、21.03±0.2°、21.62±0.2°、24.30±0.2°、25.97±0.2°、27.39±0.2°和27.98±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ, the crystal form C is at 4.46±0.2°, 6.30±0.2°, 7.03±0.2°, 8.90±0.2°, 10.65±0.2 °, 11.95±0.2°, 12.55±0.2°, 13.34±0.2°, 14.20±0.2°, 16.02±0.2°, 16.45±0.2°, 17.78±0.2°, 18.33±0.2°, 19.10±0.2°, 19.77±0.2 °, 20.12±0.2°, 21.03±0.2°, 21.62±0.2°, 24.30±0.2°, 25.97±0.2°, 27.39±0.2° and 27.98±0.2° have diffraction peaks.
在一些实施方案中,所述C晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在4.46±0.2°、6.30±0.2°、7.03±0.2°、8.90±0.2°、10.65±0.2°、11.95±0.2°、12.55±0.2°、12.96±0.2°、 13.34±0.2°、14.20±0.2°、15.20±0.2°、16.02±0.2°、16.45±0.2°、17.78±0.2°、18.33±0.2°、19.10±0.2°、19.77±0.2°、20.12±0.2°、21.03±0.2°、21.62±0.2°、22.44±0.2°、22.52±0.2°、22.54±0.2°、23.35±0.2°、23.98±0.2°、24.30±0.2°、24.86±0.2°、25.37±0.2°、25.97±0.2°、27.39±0.2°、27.98±0.2°和28.81±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ, the crystal form C is at 4.46±0.2°, 6.30±0.2°, 7.03±0.2°, 8.90±0.2°, 10.65±0.2 °, 11.95±0.2°, 12.55±0.2°, 12.96±0.2°, 13.34±0.2°, 14.20±0.2°, 15.20±0.2°, 16.02±0.2°, 16.45±0.2°, 17.78±0.2°, 18.33±0.2 °, 19.10±0.2°, 19.77±0.2°, 20.12±0.2°, 21.03±0.2°, 21.62±0.2°, 22.44±0.2°, 22.52±0.2°, 22.54±0.2°, 23.35±0.2°, 23.98±0.2 There are diffraction peaks at 24.30±0.2°, 24.86±0.2°, 25.37±0.2°, 25.97±0.2°, 27.39±0.2°, 27.98±0.2° and 28.81±0.2°.
在一些实施方案中,所述C晶型,其X-射线粉末衍射图谱与图2基本上一致。In some embodiments, the X-ray powder diffraction pattern of the crystal form C is substantially consistent with that in FIG. 2 .
在一些实施方案中,所述C晶型的DSC谱图在149.89±5℃处具有吸热峰。In some embodiments, the DSC spectrum of the crystal form C has an endothermic peak at 149.89±5°C.
在一些实施方案中,所述C晶型的DSC谱图与图3基本上一致。In some embodiments, the DSC spectrum of Form C is substantially the same as that of FIG. 3 .
本公开还提供了式(I)化合物的己二酸盐C晶型的制备方法,包括式(I)化合物与己二酸成盐反应步骤。The present disclosure also provides a method for preparing the crystal form C of adipate salt of the compound of formula (I), comprising the step of reacting the compound of formula (I) with adipic acid to form a salt.
本公开的一些方案中,上述C晶型的制备方法包括以下步骤:In some schemes of the present disclosure, the preparation method of the above crystal form C includes the following steps:
(1-a)将己二酸和溶剂混合;(1-a) mixing adipic acid and a solvent;
(2-a)将式(I)化合物和溶剂混合;(2-a) mixing the compound of formula (I) and a solvent;
(3-a)混合步骤(1-a)的混合物与步骤(2-a)的混合物;(3-a) mixing the mixture of step (1-a) and the mixture of step (2-a);
(4-a)将混合了所述已二酸和所述式(I)化合物的所得混合物搅拌,析晶;(4-a) Stir the resulting mixture mixed with the adipic acid and the compound of formula (I), and crystallize;
(5-a)过滤出固体,收集滤饼(即所述固体),干燥。(5-a) Filter out the solid, collect the filter cake (ie the solid), and dry.
在一些实施方案中,步骤(1-a)和步骤(2-a)中的溶剂选自环己烷、正庚烷、甲苯、氯仿、乙醚、异丙醚、甲基叔丁基醚、四氢呋喃、乙二醇二甲醚、丙酮、乙酸甲酯、乙酸乙酯、乙酸异丙酯、乙酸丁酯、甲醇、乙醇、异丙醇、正丁醇和乙腈中的至少一种,优选异丙醚、甲基叔丁基醚、甲醇、异丙醇和四氢呋喃中的至少一种,更优选异丙醇。In some embodiments, the solvent in step (1-a) and step (2-a) is selected from cyclohexane, n-heptane, toluene, chloroform, diethyl ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran , at least one of ethylene glycol dimethyl ether, acetone, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, preferably isopropyl ether, At least one of methyl tert-butyl ether, methanol, isopropanol and tetrahydrofuran, more preferably isopropanol.
在一些实施方案中,所述式(I)化合物与己二酸的摩尔比为1:2~2:1,优选摩尔比为1:1。In some embodiments, the molar ratio of the compound of formula (I) to adipic acid is 1:2-2:1, preferably 1:1.
在一些实施方案中,所述步骤(1-a)在加热条件下进行。In some embodiments, the step (1-a) is performed under heating.
在一些实施方案中,所述步骤(1-a)在加热温度约为40-80℃,优选60℃下进行。In some embodiments, the step (1-a) is carried out at a heating temperature of about 40-80°C, preferably 60°C.
在一些实施方案中,所述步骤(4-a)在15-30℃条件下进行,优选在20-25℃条件下进行。In some embodiments, the step (4-a) is carried out at 15-30°C, preferably at 20-25°C.
本公开还提供了式(I)化合物的苯甲酸盐的晶型。The present disclosure also provides a crystalline form of the benzoate salt of the compound of formula (I).
在一些实施方案中,式(I)化合物的苯甲酸盐的晶型为无水合物。In some embodiments, the crystalline form of the benzoate salt of the compound of formula (I) is an anhydrate.
本公开提供了式(I)化合物的苯甲酸盐B晶型,所述B晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在7.19±0.2°、10.01±0.2°、11.39±0.2°、18.78±0.2°处有衍射峰。The present disclosure provides the crystal form B of the benzoate salt of the compound of formula (I). In the X-ray powder diffraction pattern represented by the diffraction angle 2θ, the crystal form B is at 7.19±0.2°, 10.01±0.2°, 11.39± There are diffraction peaks at 0.2° and 18.78±0.2°.
在一些实施方案中,所述B晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在7.19±0.2°、10.01±0.2°、11.39±0.2°、18.78±0.2°和20.09±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the B crystal form, there are ° has a diffraction peak.
在一些实施方案中,所述B晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在7.19±0.2°、10.01±0.2°、11.39±0.2°、18.78±0.2°、19.01±0.2°、19.66±0.2°、19.91±0.2°、20.09±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the B crystal form, at 7.19±0.2°, 10.01±0.2°, 11.39±0.2°, 18.78±0.2°, 19.01±0.2° , 19.66±0.2°, 19.91±0.2°, 20.09±0.2° have diffraction peaks.
在一些实施方案中,所述B晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在4.69±0.2°、6.34±0.2°、7.19±0.2°、10.01±0.2°、11.39±0.2°、18.78±0.2°、19.01±0.2°、19.66±0.2°、19.91±0.2°和20.09±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the B crystal form, at 4.69±0.2°, 6.34±0.2°, 7.19±0.2°, 10.01±0.2°, 11.39±0.2 °, 18.78±0.2°, 19.01±0.2°, 19.66±0.2°, 19.91±0.2° and 20.09±0.2° have diffraction peaks.
在一些实施方案中,所述B晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在4.69±0.2°、6.34±0.2°、7.19±0.2°、10.01±0.2°、11.11±0.2°、11.39±0.2°、14.78±0.2°、18.78±0.2°、19.01±0.2°、19.25±0.2°、19.66±0.2°、19.91±0.2°、20.09±0.2°、20.80±0.2°、21.66±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the B crystal form, at 4.69±0.2°, 6.34±0.2°, 7.19±0.2°, 10.01±0.2°, 11.11±0.2 °, 11.39±0.2°, 14.78±0.2°, 18.78±0.2°, 19.01±0.2°, 19.25±0.2°, 19.66±0.2°, 19.91±0.2°, 20.09±0.2°, 20.80±0.2°, 21.66±0.2 ° has a diffraction peak.
在一些实施方案中,所述B晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在4.69±0.2°、6.34±0.2°、7.19±0.2°、9.83±0.2°、10.01±0.2°、11.11±0.2°、11.39±0.2°、14.78±0.2°、18.78±0.2°、19.01±0.2°、19.25±0.2°、19.66±0.2°、19.91±0.2°、20.09±0.2°、20.80±0.2°、21.66±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the B crystal form, at 4.69±0.2°, 6.34±0.2°, 7.19±0.2°, 9.83±0.2°, 10.01±0.2° , 11.11±0.2°, 11.39±0.2°, 14.78±0.2°, 18.78±0.2°, 19.01±0.2°, 19.25±0.2°, 19.66±0.2°, 19.91±0.2°, 20.09±0.2°, 20.80±0.2° , There is a diffraction peak at 21.66±0.2°.
在一些实施方案中,所述B晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在4.69±0.2°、4.91±0.2°、6.34±0.2°、7.19±0.2°、9.37±0.2°、9.83±0.2°、10.01±0.2°、10.32±0.2°、11.11±0.2°、11.39±0.2°、12.06±0.2°、12.69±0.2°、13.52±0.2°、14.07±0.2°、14.36±0.2°、14.78±0.2°、 17.30±0.2°、18.78±0.2°、19.01±0.2°、19.25±0.2°、19.66±0.2°、19.91±0.2°、20.09±0.2°、20.80±0.2°、21.15±0.2°、21.66±0.2°、22.35±0.2°、23.57±0.2°、23.70±0.2°、24.63±0.2°、25.68±0.2°、27.05±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the B crystal form, at 4.69±0.2°, 4.91±0.2°, 6.34±0.2°, 7.19±0.2°, 9.37±0.2 °, 9.83±0.2°, 10.01±0.2°, 10.32±0.2°, 11.11±0.2°, 11.39±0.2°, 12.06±0.2°, 12.69±0.2°, 13.52±0.2°, 14.07±0.2°, 14.36±0.2 °, 14.78±0.2°, 17.30±0.2°, 18.78±0.2°, 19.01±0.2°, 19.25±0.2°, 19.66±0.2°, 19.91±0.2°, 20.09±0.2°, 20.80±0.2°, 21.15±0.2 °, 21.66±0.2°, 22.35±0.2°, 23.57±0.2°, 23.70±0.2°, 24.63±0.2°, 25.68±0.2°, 27.05±0.2° have diffraction peaks.
在一些实施方案中,所述B晶型,其X-射线粉末衍射图谱与图5基本上一致。In some embodiments, the X-ray powder diffraction pattern of the crystal form B is substantially consistent with FIG. 5 .
在一些实施方案中,所述B晶型的DSC谱图在150.08±5℃处具有吸热峰。In some embodiments, the DSC spectrum of the crystal form B has an endothermic peak at 150.08±5°C.
在一些实施方案中,所述B晶型的DSC谱图与图6基本上一致。In some embodiments, the DSC spectrum of Form B is substantially consistent with FIG. 6 .
本公开还提供了式(I)化合物的苯甲酸盐B晶型的制备方法,包括式(I)化合物与苯甲酸成盐反应步骤。The present disclosure also provides a preparation method of the crystal form B of the benzoate salt of the compound of formula (I), comprising the step of reacting the compound of formula (I) with benzoic acid to form a salt.
本公开的一些方案中,上述B晶型的制备方法包括以下步骤:In some schemes of the present disclosure, the preparation method of the above-mentioned B crystal form includes the following steps:
(1-b)将式(I)化合物和溶剂混合;(1-b) mixing the compound of formula (I) and a solvent;
(2-b)向步骤(1-b)的混合物中加入苯甲酸;(2-b) adding benzoic acid to the mixture of step (1-b);
(3-b)将混合了所述式(I)化合物和所述苯甲酸的所得混合物搅拌,析晶;(3-b) Stir the resulting mixture mixed with the compound of formula (I) and the benzoic acid, and crystallize;
(4-b)过滤出并收集固体。(4-b) The solid was filtered off and collected.
在一些实施方案中,步骤(1-b)中的溶剂选自环己烷、正庚烷、甲苯、氯仿、乙醚、异丙醚、甲基叔丁基醚、四氢呋喃、乙二醇二甲醚、丙酮、乙酸甲酯、乙酸乙酯、乙酸异丙酯、乙酸丁酯、甲醇、乙醇、异丙醇、正丁醇和乙腈中的至少一种,优选异丙醚、甲基叔丁基醚、甲醇、异丙醇和四氢呋喃中的至少一种,更优选异丙醚。In some embodiments, the solvent in step (1-b) is selected from cyclohexane, n-heptane, toluene, chloroform, diethyl ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran, ethylene glycol dimethyl ether , acetone, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, preferably isopropyl ether, methyl tert-butyl ether, At least one of methanol, isopropanol and tetrahydrofuran, more preferably isopropyl ether.
在一些实施方案中,所述式(I)化合物与苯甲酸的摩尔比为1:2~2:1,优选摩尔比为1:1。In some embodiments, the molar ratio of the compound of formula (I) to benzoic acid is 1:2-2:1, preferably 1:1.
在一些实施方案中,所述步骤(1-b)在15-30℃条件下进行,优选在20-25℃条件下进行。In some embodiments, the step (1-b) is carried out at 15-30°C, preferably at 20-25°C.
在一些实施方案中,所述步骤(3-b)的搅拌在加热条件下进行,优选加热温度约为40-68℃,更优选60-68℃。In some embodiments, the stirring in step (3-b) is carried out under heating conditions, preferably the heating temperature is about 40-68°C, more preferably 60-68°C.
进一步,式(I)化合物的苯甲酸盐B晶型的制备方法还包括以下步骤:Further, the preparation method of the benzoate B crystal form of the compound of formula (I) also includes the following steps:
(5-b)将步骤(4-b)收集的固体和溶剂混合;(5-b) mixing the solid collected in step (4-b) with a solvent;
(6-b)将所得混合物搅拌;(6-b) stirring the resulting mixture;
(7-b)过滤出并收集固体,干燥。(7-b) was filtered off and the solid was collected and dried.
在一些实施方案中,步骤(5-b)中的溶剂选自异丙醚、甲基叔丁基醚、甲醇、异丙醇和四氢呋喃中的至少一种,优选异丙醚。In some embodiments, the solvent in step (5-b) is selected from at least one of isopropyl ether, methyl tert-butyl ether, methanol, isopropanol and tetrahydrofuran, preferably isopropyl ether.
在一些实施方案中,所述步骤(6-b)在15-30℃条件下进行,优选在20-25℃条件下进行。In some embodiments, the step (6-b) is carried out at 15-30°C, preferably at 20-25°C.
本公开还提供了式(I)化合物琥珀酸盐的晶型。The present disclosure also provides a crystalline form of the succinate salt of the compound of formula (I).
在一些实施方案中,式(I)化合物琥珀酸盐的晶型为无水合物。In some embodiments, the crystalline form of the succinate salt of the compound of formula (I) is an anhydrate.
本公开提供了式(I)化合物琥珀酸盐A晶型,所述A晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在7.13±0.2°、9.30±0.2°、11.29±0.2°、15.51±0.2°、19.89±0.2°处有衍射峰。The present disclosure provides the crystal form A of succinate salt of the compound of formula (I). In the X-ray powder diffraction pattern represented by the diffraction angle 2θ, the crystal form A is at 7.13±0.2°, 9.30±0.2°, 11.29±0.2 °, 15.51±0.2°, 19.89±0.2° have diffraction peaks.
在一些实施方案中,所述A晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在7.13±0.2°、9.30±0.2°、11.29±0.2°、15.51±0.2°、17.10±0.2°、19.89±0.2°、20.26±0.2°、20.91±0.2°、21.51±0.2°、22.69±0.2°、23.58±0.2°、25.42±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ, the crystal form A is at 7.13±0.2°, 9.30±0.2°, 11.29±0.2°, 15.51±0.2°, 17.10±0.2 °, 19.89±0.2°, 20.26±0.2°, 20.91±0.2°, 21.51±0.2°, 22.69±0.2°, 23.58±0.2°, 25.42±0.2° have diffraction peaks.
在一些实施方案中,所述A晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在5.01±0.2°、7.13±0.2°、9.30±0.2°、10.09±0.2°、11.29±0.2°、11.73±0.2°、13.75±0.2°、14.30±0.2°、15.18±0.2°、15.51±0.2°、16.00±0.2°、17.10±0.2°、18.25±0.2°、18.69±0.2°、19.89±0.2°、20.26±0.2°、20.91±0.2°、21.12±0.2°、21.51±0.2°、22.69±0.2°、23.05±0.2°、23.58±0.2°、24.58±0.2°、25.42±0.2°、26.66±0.2°、27.24±0.2°和29.47±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ, the crystal form A is at 5.01±0.2°, 7.13±0.2°, 9.30±0.2°, 10.09±0.2°, 11.29±0.2 °, 11.73±0.2°, 13.75±0.2°, 14.30±0.2°, 15.18±0.2°, 15.51±0.2°, 16.00±0.2°, 17.10±0.2°, 18.25±0.2°, 18.69±0.2°, 19.89±0.2 °, 20.26±0.2°, 20.91±0.2°, 21.12±0.2°, 21.51±0.2°, 22.69±0.2°, 23.05±0.2°, 23.58±0.2°, 24.58±0.2°, 25.42±0.2°, 26.66±0.2 °, 27.24±0.2° and 29.47±0.2° have diffraction peaks.
在一些实施方案中,所述A晶型,以衍射角2θ表示的X-射线粉末衍射图谱中,在5.01±0.2°、7.13±0.2°、9.30±0.2°、10.09±0.2°、11.29±0.2°、11.73±0.2°、13.75±0.2°、14.30±0.2°、15.18±0.2°、15.51±0.2°、16.00±0.2°、17.10±0.2°、18.25±0.2°、18.69±0.2°、19.89±0.2°、20.26±0.2°、20.91±0.2°、21.12±0.2°、21.51±0.2°、22.69±0.2°、23.05±0.2°、23.58±0.2°、24.58±0.2°、25.42±0.2°、26.66±0.2°、27.24±0.2°、28.65±0.2°、29.47±0.2°、30.90±0.2°、32.31±0.2°、34.45±0.2°处有衍射峰。In some embodiments, in the X-ray powder diffraction pattern represented by the diffraction angle 2θ, the crystal form A is at 5.01±0.2°, 7.13±0.2°, 9.30±0.2°, 10.09±0.2°, 11.29±0.2 °, 11.73±0.2°, 13.75±0.2°, 14.30±0.2°, 15.18±0.2°, 15.51±0.2°, 16.00±0.2°, 17.10±0.2°, 18.25±0.2°, 18.69±0.2°, 19.89±0.2 °, 20.26±0.2°, 20.91±0.2°, 21.12±0.2°, 21.51±0.2°, 22.69±0.2°, 23.05±0.2°, 23.58±0.2°, 24.58±0.2°, 25.42±0.2°, 26.66±0.2 °, 27.24±0.2°, 28.65±0.2°, 29.47±0.2°, 30.90±0.2°, 32.31±0.2°, 34.45±0.2° have diffraction peaks.
在一些实施方案中,所述A晶型的X-射线粉末衍射图谱与图8基本上一致。In some embodiments, the X-ray powder diffraction pattern of Form A is substantially consistent with FIG. 8 .
在一些实施方案中,所述A晶型的DSC谱图在186.82±5℃处具有吸热峰。In some embodiments, the DSC spectrum of the crystal form A has an endothermic peak at 186.82±5°C.
在一些实施方案中,所述A晶型的DSC谱图与图9基本上一致。In some embodiments, the DSC spectrum of Form A is substantially consistent with FIG. 9 .
在一些实施方案中,所述A晶型的晶胞参数为
Figure PCTCN2022131313-appb-000006
Figure PCTCN2022131313-appb-000007
α=90°,β=90°,γ=90°。
In some embodiments, the unit cell parameters of the A crystal form are
Figure PCTCN2022131313-appb-000006
Figure PCTCN2022131313-appb-000007
α=90°, β=90°, γ=90°.
本公开还提供了制备式(I)化合物琥珀酸盐A晶型的方法,包括以下步骤:The present disclosure also provides a method for preparing the succinate A crystal form of the compound of formula (I), comprising the following steps:
(1-c)将式(I)化合物和溶剂混合;(1-c) mixing the compound of formula (I) and a solvent;
(2-c)向步骤(1-c)的混合物中加入琥珀酸;(2-c) adding succinic acid to the mixture of step (1-c);
(3-c)将混合了所述式(I)化合物和所述琥珀酸的所得混合物搅拌,析出固体;(3-c) stirring the mixture obtained by mixing the compound of formula (I) and the succinic acid to precipitate a solid;
(4-c)过滤出并收集固体。(4-c) The solid was filtered off and collected.
在一些实施方案中,步骤(1-c)中的溶剂选自环己烷、正庚烷、甲苯、氯仿、乙醚、异丙醚、甲基叔丁基醚、四氢呋喃、乙二醇二甲醚、丙酮、乙酸甲酯、乙酸乙酯、乙酸异丙酯、乙酸丁酯、甲醇、乙醇、异丙醇、正丁醇和乙腈中的至少一种,优选异丙醚、甲基叔丁基醚、甲醇、异丙醇和四氢呋喃中的至少一种,更优选异丙醚。In some embodiments, the solvent in step (1-c) is selected from cyclohexane, n-heptane, toluene, chloroform, diethyl ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran, ethylene glycol dimethyl ether , acetone, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, preferably isopropyl ether, methyl tert-butyl ether, At least one of methanol, isopropanol and tetrahydrofuran, more preferably isopropyl ether.
在一些实施方案中,所述式(I)化合物与琥珀酸的摩尔比为1:2~2:1,优选摩尔比为1:1。In some embodiments, the molar ratio of the compound of formula (I) to succinic acid is 1:2-2:1, preferably 1:1.
在一些实施方案中,所述步骤(3-c)在15-30℃条件下进行,优选在20-25℃条件下进行。In some embodiments, the step (3-c) is carried out at 15-30°C, preferably at 20-25°C.
在一些实施方案中,所述步骤(3-c)的反应时间为1-48小时,优选3-24小时,更优选8-16小时。In some embodiments, the reaction time of the step (3-c) is 1-48 hours, preferably 3-24 hours, more preferably 8-16 hours.
进一步,式(I)化合物的琥珀酸盐A晶型的制备方法还包括以下步骤:Further, the preparation method of the succinate A crystal form of the compound of formula (I) also includes the following steps:
(5-c)将步骤(4-c)收集的固体和溶剂混合;(5-c) mixing the solid collected in step (4-c) with a solvent;
(6-c)将所得混合物搅拌析晶;(6-c) stirring and crystallizing the resulting mixture;
(7-c)过滤出并收集固体,干燥。(7-c) was filtered off and the solid was collected and dried.
在一些实施方案中,步骤(5-c)中的溶剂选自甲醇、乙醇、异丙醇、四氢呋喃和水中的至少一种,优选乙醇/水的混合溶剂。In some embodiments, the solvent in step (5-c) is at least one selected from methanol, ethanol, isopropanol, tetrahydrofuran and water, preferably a mixed solvent of ethanol/water.
在一些实施方案中,乙醇/水的体积比约为1:1~50:1,优选2:1~20:1;在本公开的一些具体实施方案中,乙醇/水的体积比约为10:1、9:1、8:1、7:1、6:1、5:1或4:1。In some embodiments, the volume ratio of ethanol/water is about 1:1~50:1, preferably 2:1~20:1; in some specific embodiments of the present disclosure, the volume ratio of ethanol/water is about 10 :1, 9:1, 8:1, 7:1, 6:1, 5:1 or 4:1.
在一些实施方案中,所述步骤(6-c)在15-30℃条件下进行,优选在20-25℃条件下进行。In some embodiments, the step (6-c) is carried out at 15-30°C, preferably at 20-25°C.
在一些实施方案中,所述步骤(6-c)的搅拌析晶时间为0.5-48小时,优选1-12小时,更优选2-4小时。In some embodiments, the stirring crystallization time of the step (6-c) is 0.5-48 hours, preferably 1-12 hours, more preferably 2-4 hours.
本公开还提供药物组合物,其包含上述式(I)化合物的可药用盐或上述晶型和药学上可接受的辅料。The present disclosure also provides a pharmaceutical composition, which comprises a pharmaceutically acceptable salt of the compound of formula (I) above or the crystal form above and pharmaceutically acceptable excipients.
本公开还涉及上述式(I)化合物的可药用盐或上述晶型、或上述药物组合物在制备用于预防或者治疗雌激素受体相关疾病的药物中的用途。The present disclosure also relates to the use of the pharmaceutically acceptable salt of the compound of formula (I) above or the above crystal form, or the above pharmaceutical composition in the preparation of drugs for preventing or treating estrogen receptor-related diseases.
进一步,本公开涉及上述式(I)化合物的可药用盐或上述晶型、或上述药物组合物在预防或者治疗雌激素受体相关疾病中的用途。Further, the present disclosure relates to the use of a pharmaceutically acceptable salt of the compound of formula (I) above or the above crystal form, or the above pharmaceutical composition in the prevention or treatment of estrogen receptor-related diseases.
进一步,本公开涉及用于预防或者治疗雌激素受体相关疾病的上述式(I)化合物的可药用盐或上述晶型、或上述药物组合物。Further, the present disclosure relates to a pharmaceutically acceptable salt of the compound of the above formula (I) or the above crystal form, or the above pharmaceutical composition for preventing or treating estrogen receptor-related diseases.
本公开还涉及治疗雌激素受体相关疾病的方法,该方法包括给以个体治疗上有效剂量的上述式(I)化合物的可药用盐或上述晶型、上述药物组合物或者包含本公开上述式(I)化合物可药用盐或上述晶型的药物制剂。The present disclosure also relates to a method for treating estrogen receptor-related diseases, the method comprising administering to an individual a therapeutically effective dose of the pharmaceutically acceptable salt of the compound of formula (I) or the above-mentioned crystal form, the above-mentioned pharmaceutical composition, or the above-mentioned Pharmaceutically acceptable salts of the compound of formula (I) or pharmaceutical preparations of the above crystal forms.
本公开还涉及上述式(I)化合物的可药用盐或上述晶型、或上述药物组合物在制备用于预防或者治疗肿瘤的药物中的用途。The present disclosure also relates to the use of the pharmaceutically acceptable salt of the compound of the above formula (I) or the above crystal form, or the above pharmaceutical composition in the preparation of drugs for preventing or treating tumors.
进一步,本公开涉及上述式(I)化合物的可药用盐或上述晶型、或上述药物组合物在预防或者治疗肿瘤中的用途。Further, the present disclosure relates to the use of a pharmaceutically acceptable salt of the compound of formula (I) or the above crystal form, or the above pharmaceutical composition in the prevention or treatment of tumors.
进一步,本公开涉及用于预防或者治疗肿瘤的上述式(I)化合物的可药用盐或上述晶型、或上述药物组合物。Further, the present disclosure relates to a pharmaceutically acceptable salt of the compound of formula (I) or the above crystal form, or the above pharmaceutical composition for preventing or treating tumors.
本公开还涉及治疗肿瘤的方法,该方法包括给以个体治疗上有效剂量的上述式(I)化合物的可药用盐或上述晶型、上述药物组合物或者包含本公开上述式(I)化合物可药用盐或上述晶型的药物制剂。The present disclosure also relates to a method for treating tumors, the method comprising administering to an individual a therapeutically effective dose of a pharmaceutically acceptable salt of the above-mentioned compound of formula (I) or the above-mentioned crystal form, the above-mentioned pharmaceutical composition or a compound containing the above-mentioned formula (I) of the present disclosure Pharmaceutically acceptable salts or pharmaceutical preparations of the above crystal forms.
在一些实施方案中,所述的雌激素受体相关疾病包括但不限于肿瘤。In some embodiments, the estrogen receptor-related diseases include but are not limited to tumors.
在一些实施方案中,所述的雌激素受体相关疾病或肿瘤为乳腺癌。In some embodiments, the estrogen receptor-related disease or tumor is breast cancer.
在一些实施方案中,所述的雌激素受体相关疾病或肿瘤为ER阳性乳腺癌。In some embodiments, the estrogen receptor-related disease or tumor is ER-positive breast cancer.
在一些实施方案中,所述的雌激素受体相关疾病或肿瘤为ER阳性乳腺癌脑转移。In some embodiments, the estrogen receptor-related disease or tumor is brain metastases from ER-positive breast cancer.
在一些实施方案中,所述个体为肿瘤患者。In some embodiments, the individual is a cancer patient.
在一些实施方案中,所述个体为乳腺癌患者。In some embodiments, the individual is a breast cancer patient.
在一些实施方案中,所述个体为ER阳性乳腺癌患者。In some embodiments, the individual is a patient with ER-positive breast cancer.
在一些实施方案中,所述个体为ER阳性乳腺癌脑转移患者。In some embodiments, the individual is a patient with brain metastases from ER-positive breast cancer.
技术效果technical effect
本公开提供的式(I)化合物的可药用盐及其结晶具有下述至少一种优势:The pharmaceutically acceptable salts of the compound of formula (I) and crystals thereof provided by the present disclosure have at least one of the following advantages:
本公开提供的式(I)化合物的可药用盐及其结晶具有水溶性好、纯度高、稳定性好、吸湿性小等中的至少一种优点;The pharmaceutically acceptable salts of the compounds of formula (I) and their crystals provided by the present disclosure have at least one of the advantages of good water solubility, high purity, good stability, and low hygroscopicity;
相对于游离碱或其它盐型,式(I)化合物的苯甲酸盐、琥珀酸盐或己二酸盐及其晶型还能够有效抑制异构体的产生,和/或具有更优的热稳定性;Compared with free base or other salt forms, benzoate, succinate or adipate and crystal forms thereof of the compound of formula (I) can also effectively inhibit the generation of isomers, and/or have better thermal stability;
本公开提供的式(I)化合物的可药用盐或其晶型还具有制备方法简便、结晶条件温和、结晶度高、溶解性好、稳定性好和不易吸湿等中的至少一种优点,适合制备成为所期望的药物组合物。The pharmaceutically acceptable salt of the compound of formula (I) or its crystal form provided by the present disclosure also has at least one of the advantages of simple preparation method, mild crystallization conditions, high crystallinity, good solubility, good stability, and difficult to absorb moisture, etc., It is suitable for preparing the desired pharmaceutical composition.
定义和说明Definition and Description
本公开的式(I)化合物的可药用盐每天给药的剂量为0.01mg/kg到200mg/kg体重,优选为0.05mg/kg到50mg/kg体重,更优选0.1mg/kg到30mg/kg体重,以单独或分开剂量的形式。The daily dosage of the pharmaceutically acceptable salt of the compound of formula (I) of the present disclosure is 0.01 mg/kg to 200 mg/kg body weight, preferably 0.05 mg/kg to 50 mg/kg body weight, more preferably 0.1 mg/kg to 30 mg/kg body weight. kg body weight, in single or divided doses.
本公开的可药用盐或其晶型或它们的多晶型物,或它们的多晶型物的组合物,或它们的药物组合物的典型给药途径包括但不限于口服、直肠、局部、吸入、肠胃外、舌下、阴道内、鼻内、眼内、腹膜内、肌内、皮下、静脉内给药。Typical routes of administration of the pharmaceutically acceptable salts of the present disclosure or their crystalline forms or their polymorphs, or their polymorphic compositions, or their pharmaceutical compositions include, but are not limited to, oral, rectal, topical , inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous administration.
在可选的实施方案中,药物组合物是口服形式。对于口服给药,可以通过将活性化合物与本领域熟知的药学上可接受的辅料混合,来配制该药物组合物。这些辅料能使本申请的化合物被配制成片剂、丸剂、锭剂、糖衣剂、胶囊剂、液体、凝胶剂、浆剂、悬浮剂等,用于对患者的口服给药。In an alternative embodiment, the pharmaceutical composition is in oral form. For oral administration, the pharmaceutical compositions can be formulated by mixing the active compounds with pharmaceutically acceptable excipients well known in the art. These excipients enable the compounds of the present application to be formulated into tablets, pills, lozenges, dragees, capsules, liquids, gels, slurries, suspensions, etc. for oral administration to patients.
可以通过常规的混合、填充或压片方法来制备固体口服组合物。例如,可通过下述方法获得:将所述的活性化合物与固体辅料混合,任选地碾磨所得的混合物,如果需要则加入其它合适的辅料,然后将该混合物加工成颗粒,得到了片剂或糖衣剂的核心。适合的辅料包括但不限于:粘合剂、稀释剂、崩解剂、润滑剂、助流剂、甜味剂或矫味剂等。Solid oral compositions can be prepared by conventional methods of mixing, filling or tabletting. It can be obtained, for example, by mixing the active compound with solid excipients, optionally milling the resulting mixture, adding other suitable excipients if desired, and processing the mixture into granules to obtain tablets Or the core of the sugar coating. Suitable auxiliary materials include but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, etc.
术语“约”在本公开中用于指近似地、在……左右。当术语“约”与数字范围结合使用时,通过扩大所陈述的数字范围的上下限值来对该范围来进行修饰。除非另作说明,否则术语“约”在本文中用于以10%偏差的数字值修饰所陈述的值的上限和下限。The term "about" is used in this disclosure to mean approximately, around. When the term "about" is used in conjunction with a numerical range, it modifies that range by extending the limits of the stated numerical range. Unless otherwise indicated, the term "about" is used herein to modify the upper and lower limits of the stated value by a numerical value of 10% deviation.
除非另作说明,否则术语“包括(comprise)”、“含有(comprise)”或“包含(comprise)”及其英文变体例如comprises或comprising应理解为开放的、非排他性的意义,即“包括但不限于”。Unless otherwise stated, the terms "comprise", "comprise" or "comprise" and their English variants such as comprises or comprising are to be understood in an open, non-exclusive sense, ie "including but not limited to".
在整个本说明书中提到的“一些实施方案”、“可选实施方案”或“实施方案”是指在至少一实施方案中包括与该实施方案所述的相关的具体参考要素、结构或特征。因此,在整个说明书中不同位置出现的短语“一些实施方案”、“可选实施方案”或“实施方案”不必全部指同一实施方案。此外,具体要素、结构或特征可以任何适当的方式在一个或多个实施方案中结合。References throughout this specification to "some embodiments", "alternative embodiments" or "embodiments" means that at least one embodiment includes specific reference elements, structures or features described in relation to that embodiment . Thus, appearances of the phrases "some embodiments," "alternative embodiments," or "an embodiment" in various places throughout the specification are not necessarily all referring to the same embodiment. Furthermore, particular elements, structures or characteristics may be combined in any suitable manner in one or more embodiments.
本公开所述室温是指20±5℃。The room temperature mentioned in the present disclosure refers to 20±5°C.
本公开中所述的范围“m~n”表示m到n之间的任意实数组合的缩略表示,其中m和n都 是实数。数值范围“0.5~2”包括但不限于本文中已经列出了其中的0.5、0.6、0.7、0.8、0.9、1.0、1.1、1.2、1.3、1.4、1.5、1.6、1.7、1.8、1.9或2.0。The range "m∼n" described in the present disclosure represents an abbreviated representation of any combination of real numbers between m and n, where both m and n are real numbers. The numerical range "0.5~2" includes but is not limited to 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 or 2.0 .
本公开中所述的“X-射线粉末衍射图谱”为使用Cu-Kα辐射测量得到。The "X-ray powder diffraction pattern" described in this disclosure is measured using Cu-Kα radiation.
本公开中所述的“X-射线粉末衍射图谱或XRPD图谱”是指根据布拉格公式2d Sinθ=nλ(式中,λ为X射线的波长,
Figure PCTCN2022131313-appb-000008
衍射的级数n为任何正整数,一般取一级衍射峰,n=1),当X射线以掠角θ(入射角的余角,又称为布拉格角)入射到晶体或部分晶体样品的某一具有d点阵平面间距的原子面上时,就能满足布拉格方程,从而测得了这组X射线粉末衍射图。
"X-ray powder diffraction pattern or XRPD pattern" described in the present disclosure refers to according to Bragg's formula 2d Sinθ=nλ (wherein, λ is the wavelength of X-ray,
Figure PCTCN2022131313-appb-000008
The order n of diffraction is any positive integer, generally take the first-order diffraction peak, n=1), when the X-ray is incident on the crystal or part of the crystal sample at the grazing angle θ (the complementary angle of the incident angle, also known as the Bragg angle) On an atomic plane with a d-lattice plane spacing, the Bragg equation can be satisfied, and thus this group of X-ray powder diffraction patterns has been measured.
本公开公开的晶型为大致纯的结晶。本申请使用的术语“大致纯的”是指所述晶型的纯度至少为85wt%,优选至少为90wt%、更优选至少为95wt%。The crystalline forms disclosed in the present disclosure are substantially pure crystals. The term "substantially pure" as used herein means that the crystalline form has a purity of at least 85 wt%, preferably at least 90 wt%, more preferably at least 95 wt%.
对于同种化合物的同种晶型,其XRPD图谱的峰位置在整体上具有相似性,相对强度误差可能较大。还应指出的是,在混合物的鉴定中,由于含量下降等因素会造成部分衍射线的缺失,此时,无需依赖高纯试样中观察到的全部衍射峰,甚至一个衍射峰也可能对给定的晶体而言是特征性的。For the same crystal form of the same compound, the peak positions of their XRPD patterns are generally similar, and the relative intensity error may be large. It should also be pointed out that in the identification of mixtures, due to the decrease of content and other factors, some diffraction lines will be missing. At this time, there is no need to rely on all the diffraction peaks observed in high-purity samples, and even one diffraction peak may also affect the given is characteristic for a given crystal.
本公开中所述的“2θ或2θ角度”是指衍射角,θ为布拉格角,单位为°或度;每个衍射峰2θ的误差范围为±0.20°。The "2θ or 2θ angle" mentioned in the present disclosure refers to the diffraction angle, θ is the Bragg angle, and the unit is ° or degree; the error range of each diffraction peak 2θ is ±0.20°.
本公开中所述的“差示扫描量热分析或DSC”是指在样品升温或恒温过程中,测量样品与参考物之间的温度差、热流差,以表征所有与热效应有关的物理变化和化学变化,得到样品的相变信息。"Differential scanning calorimetry or DSC" in this disclosure refers to the measurement of the temperature difference and heat flow difference between the sample and the reference object during the heating or constant temperature of the sample to characterize all the physical changes related to thermal effects and Chemical changes, to obtain the phase transition information of the sample.
本公开中所述干燥温度一般为20℃~100℃,优选25℃~70℃,更优选40℃~60℃,可以常压干燥,也可以减压干燥。优选地,在减压下干燥。The drying temperature described in the present disclosure is generally 20°C-100°C, preferably 25°C-70°C, more preferably 40°C-60°C, and can be dried under normal pressure or reduced pressure. Preferably, drying is performed under reduced pressure.
对于本领域技术人员而言,本公开化合物与酸分子的摩尔比测定存在一定程度的误差,一般而言,±10%均属于合理误差范围内。随其所用之处的上下文而有一定程度的误差变化,该误差变化不超过±10%,优选±5%。For those skilled in the art, there is a certain degree of error in the determination of the molar ratio of the disclosed compound to the acid molecule, and generally speaking, ±10% is within a reasonable error range. There is a degree of error that will vary with the context in which it is used, but the error will vary by no more than ±10%, preferably ±5%.
术语“治疗”意为将本申请所述化合物或制剂进行给药以改善或消除疾病或与所述疾病相关的一个或多个症状,且包括:The term "treating" means administering a compound or formulation described herein to improve or eliminate a disease or one or more symptoms associated with the disease, and includes:
(i)抑制疾病或疾病状态,即遏制其发展;(i) inhibiting a disease or disease state, i.e. arresting its development;
(ii)缓解疾病或疾病状态,即使该疾病或疾病状态消退。(ii) amelioration of a disease or disease state, even if the disease or disease state regresses.
术语“预防”意为将本申请所述化合物或制剂进行给药以预防疾病或与所述疾病相关的一个或多个症状,且包括:The term "prevention" means administering a compound or formulation described herein to prevent a disease or one or more symptoms associated with the disease, and includes:
预防疾病或疾病状态在对象(例如哺乳动物)中出现,特别是当这类对象易患有该疾病状态,但尚未被诊断为已患有该疾病状态时。Preventing a disease or disease state from occurring in a subject (eg, a mammal), particularly when such a subject is susceptible to the disease state but has not yet been diagnosed as having the disease state.
术语“治疗上有效剂量”是指(i)治疗特定疾病、病况或障碍,(ii)减轻、改善或消除特定疾病、病况或障碍的一种或多种症状,或(iii)延迟本文中所述的特定疾病、病况或障碍的一种或多种症状发作的本公开化合物的用量。构成“治疗上有效剂量”的本公开化合物的量取决于该化合物、疾病状态及其严重性、给药方式以及待被治疗的哺乳动物的年龄而改变,但可例行性地由本领域技术人员根据其自身的知识及本公开内容而确定。The term "therapeutically effective dose" means (i) treating a particular disease, condition or disorder, (ii) alleviating, ameliorating or eliminating one or more symptoms of a particular disease, condition or disorder, or (iii) delaying the The amount of a compound of the disclosure required for the onset of one or more symptoms of a particular disease, condition or disorder as described. The amount of a compound of the disclosure that constitutes a "therapeutically effective dose" will vary depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by one skilled in the art. Based on its own knowledge and this disclosure.
术语“药学上可接受的辅料”是指对有机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些辅料。合适的辅料是本领域技术人员熟知的,例如碳水化合物、蜡、水溶性和/或水可膨胀的聚合物、亲水性或疏水性材料、明胶、油、溶剂、水等。The term "pharmaceutically acceptable excipients" refers to those excipients that have no obvious stimulating effect on the organism and will not impair the biological activity and performance of the active compound. Suitable excipients are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
本申请中,术语“个体”或“患者”包括哺乳动物和非哺乳动物。哺乳动物的实例包括但不限于哺乳动物纲的任何成员:人,非人的灵长类动物(例如黑猩猩和其它猿类和猴);家畜,例如牛、马、绵羊、山羊、猪;家养动物,例如兔、狗和猫;实验室动物,包括啮齿类动物,例如大鼠、小鼠和豚鼠等。非人哺乳动物的实例包括但不限于鸟类和鱼类等。As used herein, the term "individual" or "patient" includes mammals and non-mammals. Examples of mammals include, but are not limited to, any member of the class Mammalia: humans, non-human primates (such as chimpanzees and other apes and monkeys); livestock such as cattle, horses, sheep, goats, pigs; domesticated animals , such as rabbits, dogs, and cats; laboratory animals, including rodents, such as rats, mice, and guinea pigs. Examples of non-human mammals include, but are not limited to, birds, fish, and the like.
本公开实验所用仪器的测试条件:The test condition of the instrument used in this disclosure experiment:
1、差示扫描量热仪(Differential Scanning Calorimeter,DSC)1. Differential Scanning Calorimeter (DSC)
仪器型号:TA DSC2500Instrument model: TA DSC2500
吹扫气:氮气Purge gas: Nitrogen
样品盘:铝盘,非密封压盖Sample pan: aluminum pan, non-sealed gland
方法:线性升温Method: linear heating
升温速率:10℃/minHeating rate: 10°C/min
温度范围:30℃~设置终点温度Temperature range: 30 ℃ ~ set the end temperature
2、X-射线粉末衍射谱(X-ray Powder Diffraction,XRPD)2. X-ray Powder Diffraction (XRPD)
仪器型号:Bruker D8 FocusInstrument model: Bruker D8 Focus
射线:Cu Kα,Kα1
Figure PCTCN2022131313-appb-000009
:1.54060;Kα2
Figure PCTCN2022131313-appb-000010
:1.54439
Rays: Cu Kα, Kα1
Figure PCTCN2022131313-appb-000009
: 1.54060; Kα2
Figure PCTCN2022131313-appb-000010
: 1.54439
Kα1/Kα2:2Kα1/Kα2:2
狭缝(°):2.5Slit (°): 2.5
扫描方式:θ/2θ,扫描范围:3-40°Scanning mode: θ/2θ, scanning range: 3-40°
停留时间(秒):0.12Dwell time (seconds): 0.12
扫描步长(°2θ):0.01Scan step size (°2θ): 0.01
电压:40kV,电流:40mAVoltage: 40kV, Current: 40mA
3、热重分析(Thermogravimetric Analysis,TGA)3. Thermogravimetric Analysis (TGA)
仪器型号:TA TGA550Instrument model: TA TGA550
吹扫气:氮气Purge gas: Nitrogen
样品盘:铂金,敞口Sample tray: Platinum, open
方法:线性升温Method: linear heating
升温速率:10℃/minHeating rate: 10°C/min
温度范围:30℃~设置终点温度Temperature range: 30 ℃ ~ set the end temperature
4、饱和溶解度试验的高效液相色谱(HPLC)4. High performance liquid chromatography (HPLC) for saturation solubility test
色谱柱:Water XBridge C18(4.6*100mm,3.5μm)Chromatographic column: Water XBridge C18 (4.6*100mm, 3.5μm)
流动相A:0.01mol/L氯化铵溶液(pH10.0)Mobile phase A: 0.01mol/L ammonium chloride solution (pH10.0)
流动相B:乙腈Mobile Phase B: Acetonitrile
等度条件:流动相A:B=50:50,等度15minIsocratic conditions: mobile phase A:B=50:50, isocratic for 15min
流速:1.5mL/minFlow rate: 1.5mL/min
波长:260nmWavelength: 260nm
柱温:35℃Column temperature: 35°C
进样量:10μlInjection volume: 10μl
稀释剂:乙腈Diluent: Acetonitrile
5、异构体杂质检测的高效液相色谱(HPLC)5. High performance liquid chromatography (HPLC) for detection of isomer impurities
色谱柱:CHIRALPAK AD-H(4.6*250mm,5μm)Chromatographic column: CHIRALPAK AD-H (4.6*250mm, 5μm)
流动相A:正己烷Mobile Phase A: n-Hexane
流动相B:0.1%二乙胺-乙醇Mobile phase B: 0.1% diethylamine-ethanol
等度条件:流动相A:流动相B=70:30,等度运行15minIsocratic conditions: mobile phase A: mobile phase B = 70:30, run isocratically for 15 minutes
流速:1.0mL/minFlow rate: 1.0mL/min
波长:260nmWavelength: 260nm
柱温:35℃Column temperature: 35°C
进样量:10μlInjection volume: 10μl
稀释剂:0.1%二乙胺-乙醇Diluent: 0.1% diethylamine-ethanol
式(I)化合物可药用盐稳定性考察试验的高效液相色谱(HPLC)The high performance liquid chromatography (HPLC) of formula (I) compound pharmaceutically acceptable salt stability investigation test
色谱柱:Water XBridge C18(4.6*150mm,3.5μm)Chromatographic column: Water XBridge C18 (4.6*150mm, 3.5μm)
流动相A:0.01mol/L氯化铵溶液(氨水调pH10.0)Mobile phase A: 0.01mol/L ammonium chloride solution (adjust the pH to 10.0 with ammonia water)
流动相B:乙腈Mobile Phase B: Acetonitrile
流速:1.0mL/minFlow rate: 1.0mL/min
波长:285nmWavelength: 285nm
柱温:35℃Column temperature: 35°C
进样量:20μlInjection volume: 20μl
本领域技术人员认识到对于同一化合物的给定结晶形式的XRPD峰位置和/或强度的测量数据将在误差范围内变化。2θ值允许适当的误差范围。通常,误差范围是由“±”表示。例如,5.92±0.2°2θ表示在约6.12至5.72范围内。取决于样品制备技术、应用于仪器的校准技术、人类操作偏差等,本领域技术人员认识到对于XRPD衍射角(2θ)的适当的误差范围可为约±0.20°、±0.15°、±0.10°、±0.05°或更小。当用于描述XRPD图时,术语“基本上一致”或“基本上如……所示”是指包括至少50%、至少60%、至少70%、至少80%、至少90%、至少90%或至少99%的衍射角在±0.2°2θ的标准偏差范围内的衍射峰的图。Those skilled in the art recognize that measurements of XRPD peak positions and/or intensities for a given crystalline form of the same compound will vary within error. The 2Θ values allow for an appropriate margin of error. Usually, the margin of error is represented by "±". For example, 5.92 ± 0.2° 2Θ represents a range of about 6.12 to 5.72. Depending on the sample preparation technique, the calibration technique applied to the instrument, human operator bias, etc., one skilled in the art recognizes that suitable error ranges for XRPD diffraction angles (2Θ) may be about ±0.20°, ±0.15°, ±0.10° , ±0.05° or less. When used to describe an XRPD pattern, the term "substantially identical" or "substantially as shown" means comprising at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 90% Or a pattern of diffraction peaks with at least 99% of the diffraction angles within a standard deviation of ±0.2° 2Θ.
由于本领域技术人员认识到对于同一化合物的给定结晶形式的DSC热谱图的测量数据将在误差容限内变化。单峰峰值(以摄氏度表示)允许适当的误差范围。通常,误差范围是由“±”表示。对于同种化合物的同种晶型,在连续的分析中,热转变温度和熔点误差典型地在约±5.0℃之内。例如,“186.82±5℃”的峰值表示在181.82至191.82范围内。取决于样品制备技术、应用于仪器的校准技术、人类操作偏差等,本领域技术人员认识到对于单峰峰值的适当的误差范围可为±5.0、±4.0、±3.0、±2.0或更小。As one skilled in the art recognizes, measurements of a DSC thermogram for a given crystalline form of the same compound will vary within a margin of error. Single-peak peak values (expressed in degrees Celsius) allow for a modest margin of error. Usually, the margin of error is represented by "±". For the same crystalline form of the same compound, thermal transition temperatures and melting points are typically within about ±5.0°C in consecutive analyses. For example, the peak value of "186.82±5°C" indicates that it is in the range of 181.82 to 191.82. Depending on the sample preparation technique, the calibration technique applied to the instrument, human operator bias, etc., one skilled in the art will recognize that suitable error ranges for unimodal peaks may be ±5.0, ±4.0, ±3.0, ±2.0 or less.
勒克斯(lux,法定符号lx)是照度(luminance)的单位。被光均匀照射的物体,在1平方米面积上所得的光通量是1流明时,它的照度是1勒克斯。Lux (lux, legal symbol lx) is the unit of luminance. When the luminous flux obtained by an object uniformly illuminated by light is 1 lumen on an area of 1 square meter, its illuminance is 1 lux.
实施例Example
以下实施例详细说明发明的技术方案,但本公开的保护范围包括但不限于此。The following examples describe the technical solutions of the invention in detail, but the protection scope of the present disclosure includes but is not limited thereto.
化合物的结构是通过核磁共振(NMR)和/或质谱(MS)来确定的。NMR位移的单位为10 -6(ppm)。NMR测定的溶剂为氘代二甲基亚砜、氘代氯仿、氘代甲醇等,内标为四甲基硅烷(TMS);“IC 50”指半数抑制浓度,指达到最大抑制效果一半时的浓度。 Compound structures were determined by nuclear magnetic resonance (NMR) and/or mass spectroscopy (MS). The unit of NMR shift is 10 -6 (ppm). The solvents measured by NMR are deuterated dimethyl sulfoxide, deuterated chloroform, deuterated methanol, etc., and the internal standard is tetramethylsilane (TMS); concentration.
实施例1:N-((S)(1-(3-氟丙基)吡咯烷-3-基)-6-((1S,3R)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)吡啶-3-胺(式(I)化合物)的合成Example 1: N-((S)(1-(3-fluoropropyl)pyrrolidin-3-yl)-6-((1S,3R)-3-methyl-2-(2,2,2 Synthesis of -trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indol-1-yl)pyridin-3-amine (formula (I) compound)
Figure PCTCN2022131313-appb-000011
Figure PCTCN2022131313-appb-000011
合成方法:resolve resolution:
Figure PCTCN2022131313-appb-000012
Figure PCTCN2022131313-appb-000012
步骤1:(S)-叔丁基(1-(3-氟丙基)吡咯烷-3-基)氨基甲酯的合成Step 1: Synthesis of (S)-tert-butyl(1-(3-fluoropropyl)pyrrolidin-3-yl)aminomethyl ester
将(S)-叔丁基吡咯烷-3-基氨基甲酯(500.00mg,2.68mmol)溶于四氢呋喃(10mL)中,加入氢氧化钠溶液(5mol﹒L -1,1.07mL)和1-碘-3-氟丙烷(529.88mg,2.82mmol)。反应液于25℃搅拌反应16小时。TLC检测原料反应完全后,将反应液用乙酸乙酯(50mL)稀释后,用饱和氯化铵溶液(10mL)洗涤,分别收集水相和有机相。水相用乙酸乙酯(20mL)萃取三次后,将所有有机相合并,用硫酸钠干燥后,有机相减压浓缩干,然后柱层析纯化(二氧化硅,二氯甲烷/甲醇=100/1)得到产物(S)-叔丁基(1-(3-氟丙基)吡咯烷-3-基)氨基甲酯(480.00mg)。 Dissolve (S)-tert-butylpyrrolidin-3-ylaminomethyl ester (500.00mg, 2.68mmol) in tetrahydrofuran (10mL), add sodium hydroxide solution (5mol·L -1 , 1.07mL) and 1- Iodo-3-fluoropropane (529.88 mg, 2.82 mmol). The reaction solution was stirred and reacted at 25°C for 16 hours. After the reaction of the raw materials was detected by TLC, the reaction solution was diluted with ethyl acetate (50 mL), washed with saturated ammonium chloride solution (10 mL), and the aqueous phase and the organic phase were collected respectively. After the aqueous phase was extracted three times with ethyl acetate (20 mL), all the organic phases were combined and dried over sodium sulfate. The organic phase was concentrated to dryness under reduced pressure, and then purified by column chromatography (silica, dichloromethane/methanol=100/ 1) The product (S)-tert-butyl(1-(3-fluoropropyl)pyrrolidin-3-yl)aminomethyl ester (480.00 mg) was obtained.
1H NMR(400MHz,METHANOL-d 4)δ4.58-4.53(m,1H),4.46-4.40(m,1H),4.14-4.04(m,1H),2.93-2.85(m,1H),2.77-2.67(m,1H),2.61(dd,J=7.78,5.52Hz,3H),2.47-2.40(m,1H),2.29-2.17(m,1H),1.99-1.82(m,2H),1.71-1.61(m,1H),1.45(s,9H). 1 H NMR (400MHz, METHANOL-d 4 )δ4.58-4.53(m,1H),4.46-4.40(m,1H),4.14-4.04(m,1H),2.93-2.85(m,1H),2.77 -2.67(m,1H),2.61(dd,J=7.78,5.52Hz,3H),2.47-2.40(m,1H),2.29-2.17(m,1H),1.99-1.82(m,2H),1.71 -1.61(m,1H),1.45(s,9H).
步骤2:(S)-1-(3-氟丙基)吡咯烷-3-胺盐酸盐的合成Step 2: Synthesis of (S)-1-(3-fluoropropyl)pyrrolidin-3-amine hydrochloride
将(S)-叔丁基(1-(3-氟丙基)吡咯烷-3-基)氨基甲酯(480.00mg,1.93mmol)溶于1,4-二氧六环(3mL)中,然后加入盐酸-1,4-二氧六环溶液(4moL﹒L -1,4.94mL),反应液为黄色透明溶液。反应液在25℃下搅拌3小时。TLC检测原料反应完全后,将反应溶液减压浓缩得化合物(S)-1-(3-氟丙基)吡咯烷-3-胺盐酸盐(450.00mg)。 (S)-tert-butyl(1-(3-fluoropropyl)pyrrolidin-3-yl)aminomethyl ester (480.00 mg, 1.93 mmol) was dissolved in 1,4-dioxane (3 mL), Then hydrochloric acid-1,4-dioxane solution (4moL·L -1 , 4.94mL) was added, and the reaction solution was a yellow transparent solution. The reaction solution was stirred at 25°C for 3 hours. After the reaction of the raw materials was detected by TLC, the reaction solution was concentrated under reduced pressure to obtain compound (S)-1-(3-fluoropropyl)pyrrolidin-3-amine hydrochloride (450.00 mg).
1H NMR(400MHz,DMSO-d 6)δ8.80-8.42(m,3H),4.62(s,1H),4.51(s,1H),4.12-3.45(m,3H),3.17(br s,3H),2.35-1.99(m,4H). 1 H NMR (400MHz,DMSO-d 6 )δ8.80-8.42(m,3H),4.62(s,1H),4.51(s,1H),4.12-3.45(m,3H),3.17(br s, 3H),2.35-1.99(m,4H).
步骤3:(R)-1-(1H-吲哚-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺的合成Step 3: Synthesis of (R)-1-(1H-indol-3-yl)-N-(2,2,2-trifluoroethyl)propane-2-amine
将(2R)-1-(1H-吲哚-3-基)丙烷-2-胺(600mg,3.44mmol)和N,N-二异丙基乙胺(445.05mg,3.44mmol)溶于1,4-二氧六环(10mL)中,在25℃下加入溶在1,4-二氧六环(5mL)中的三氟甲磺酸三氟乙酯(1.20g,5.17mmol),反应液于75℃搅拌反应16小时。将反应液减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=3/1)得到产物(R)-1-(1H-吲哚-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺(0.69g)。Dissolve (2R)-1-(1H-indol-3-yl)propane-2-amine (600 mg, 3.44 mmol) and N,N-diisopropylethylamine (445.05 mg, 3.44 mmol) in 1, To 4-dioxane (10mL), add trifluoroethyl trifluoromethanesulfonate (1.20g, 5.17mmol) dissolved in 1,4-dioxane (5mL) at 25°C, and the reaction solution The reaction was stirred at 75°C for 16 hours. The reaction solution was concentrated to dryness under reduced pressure. Then column chromatography purification (silica, petroleum ether/ethyl acetate = 3/1) to obtain the product (R)-1-(1H-indol-3-yl)-N-(2,2,2-tri Fluoroethyl)propan-2-amine (0.69g).
MS m/z(ESI):257.2[M+H] +MS m/z(ESI):257.2[M+H] + ;
1H NMR(400MHz,METHANOL-d 4)δ7.56-7.54(d,J=8.0Hz,1H),7.36-7.34(d,J=8.0Hz,1H),7.12-7.08(m,2H),7.03-7.00(m,1H),3.26-3.23(m,2H),3.12-3.10(m,1H),2.93-2.88(m, 1H),2.80-2.78(m,1H),1.11(d,J=6.0Hz,3H) 1 H NMR (400MHz, METHANOL-d 4 ) δ7.56-7.54(d, J=8.0Hz, 1H), 7.36-7.34(d, J=8.0Hz, 1H), 7.12-7.08(m, 2H), 7.03-7.00(m,1H),3.26-3.23(m,2H),3.12-3.10(m,1H),2.93-2.88(m,1H),2.80-2.78(m,1H),1.11(d,J =6.0Hz,3H)
步骤4:(1S,3R)-1-(5-溴吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚的合成Step 4: (1S,3R)-1-(5-Bromopyridin-2-yl)-3-methyl-2-(2,2,2-trifluoroethyl)-2,3,4,9- Synthesis of Tetrahydro-1H-pyrido[3,4-b]indole
将(R)-1-(1H-吲哚-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺(120.00mg,468.26μmol)溶于甲苯(2mL)中,加入5-溴-吡啶-2-甲醛(87.10mg,468.26μmol)和乙酸(562.40mg,9.37mmol),反应液为黄色透明溶液。反应液在90℃下搅拌10小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩干,经薄层层析纯化(二氧化硅,石油醚/乙酸乙酯=4/1)得(1S,3R)-1-(5-溴吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚(110.00mg)。Dissolve (R)-1-(1H-indol-3-yl)-N-(2,2,2-trifluoroethyl)propan-2-amine (120.00 mg, 468.26 μmol) in toluene (2 mL) 5-bromo-pyridine-2-carbaldehyde (87.10mg, 468.26μmol) and acetic acid (562.40mg, 9.37mmol) were added, and the reaction solution was a yellow transparent solution. The reaction solution was stirred at 90° C. for 10 hours. After the completion of the reaction detected by LCMS, the reaction solution was cooled to room temperature, concentrated to dryness under reduced pressure, and purified by thin layer chromatography (silica, petroleum ether/ethyl acetate=4/1) to obtain (1S,3R)-1- (5-bromopyridin-2-yl)-3-methyl-2-(2,2,2-trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4 -b] Indole (110.00 mg).
MS m/z(ESI):424.1,426.1[M+H] +MS m/z (ESI): 424.1, 426.1 [M+H] + .
1H NMR(400MHz,METHANOL-d 4)δ8.59(s,1H),8.01-7.94(m,1H),7.54(d,J=8.53Hz,1H),7.46(d,J=7.78Hz,1H),7.30(d,J=8.03Hz,1H),7.08(d,J=7.59Hz,1H),7.03-6.96(m,1H),5.08(s,1H),3.58-3.46(m,1H),3.38-3.34(m,1H),3.13-2.99-(m,1H),2.80(d,J=4.52Hz,1H),2.70-2.61(m,1H),1.26(d,J=6.78Hz,3H). 1 H NMR (400MHz, METHANOL-d 4 ) δ8.59(s, 1H), 8.01-7.94(m, 1H), 7.54(d, J=8.53Hz, 1H), 7.46(d, J=7.78Hz, 1H), 7.30(d, J=8.03Hz, 1H), 7.08(d, J=7.59Hz, 1H), 7.03-6.96(m, 1H), 5.08(s, 1H), 3.58-3.46(m, 1H ),3.38-3.34(m,1H),3.13-2.99-(m,1H),2.80(d,J=4.52Hz,1H),2.70-2.61(m,1H),1.26(d,J=6.78Hz ,3H).
步骤5:N-((S)-1-(3-氟丙基)吡咯烷-3-基)-6-(((1S,3R)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)吡啶-3-胺(式(I)化合物)的合成Step 5: N-((S)-1-(3-fluoropropyl)pyrrolidin-3-yl)-6-(((1S,3R)-3-methyl-2-(2,2,2 Synthesis of -trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indol-1-yl)pyridin-3-amine (compound of formula (I))
将(1S,3R)-1-(5-溴吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚(140.00mg,263.99μmol)溶于四氢呋喃(3mL)中,加入(S)-1-(3-氟丙基)吡咯烷-3-胺盐酸盐(86.77mg,316.79μmol),叔丁醇钠(152.22mg,1.58mmol)和甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2,4,6-三异丙基-1,1-联苯)(2-氨基-1,1-联苯-2-基)钯(II)(23.93mg,26.40μmol),氮气环境下反应液在80℃搅拌4小时。LCMS检测反应完成后,冷却至室温,过滤,滤液减压浓缩后,经过制备液相色谱纯化(Phenomenex Gemini C18柱,3um二氧化硅,30mm直径,75mm长度);(使用水(含有0.225%甲酸)和乙腈的极性递减的混合物作为洗脱液)纯化得到化合物N-((S)-1-(3-氟丙基)吡咯烷-3-基)-6-(((1S,3R)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-1-基)吡啶-3-胺(37.79mg)。(1S,3R)-1-(5-bromopyridin-2-yl)-3-methyl-2-(2,2,2-trifluoroethyl)-2,3,4,9-tetrahydro -1H-pyrido[3,4-b]indole (140.00 mg, 263.99 μmol) was dissolved in tetrahydrofuran (3 mL), and (S)-1-(3-fluoropropyl)pyrrolidin-3-amine salt was added salt (86.77mg, 316.79μmol), sodium tert-butoxide (152.22mg, 1.58mmol) and methanesulfonic acid (2-dicyclohexylphosphine)-3,6-dimethoxy-2,4,6-tri Isopropyl-1,1-biphenyl)(2-amino-1,1-biphenyl-2-yl)palladium(II) (23.93 mg, 26.40 μmol), the reaction solution was stirred at 80°C for 4 hours under nitrogen atmosphere . After the LCMS detection reaction is completed, cool to room temperature, filter, and the filtrate is concentrated under reduced pressure, and purified by preparative liquid chromatography (Phenomenex Gemini C18 column, 3um silica, 30mm diameter, 75mm length); (using water (containing 0.225% formic acid ) and a mixture of decreasing polarity of acetonitrile as eluent) to obtain compound N-((S)-1-(3-fluoropropyl)pyrrolidin-3-yl)-6-(((1S,3R) -3-Methyl-2-(2,2,2-trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indol-1-yl) Pyridin-3-amine (37.79 mg).
MS m/z(ESI):365.1[M+H] +MS m/z (ESI): 365.1 [M+H] + .
1H NMR(400MHz,METHANOL-d 4)δ7.89(d,J=2.76Hz,1H),7.46(d,J=7.78Hz,1H),7.25(d,J=8.53Hz,2H),7.09-7.03(m,2H),7.02-6.97(m,1H),4.98(s,1H),4.60(t,J=5.65Hz,1H),4.49(t,J=5.65Hz,1H),4.18(br s,1H),3.51-3.35(m,4H),3.14-2.99(m,5H),2.92(dd,J=15.18,4.89Hz,1H),2.65(dd,J=16.06,6.78Hz,1H),2.53-2.42(m,1H),2.12-1.92(m,3H),1.20(d,J=6.78Hz,3H). 1 H NMR (400MHz, METHANOL-d 4 ) δ7.89(d, J=2.76Hz, 1H), 7.46(d, J=7.78Hz, 1H), 7.25(d, J=8.53Hz, 2H), 7.09 -7.03(m,2H),7.02-6.97(m,1H),4.98(s,1H),4.60(t,J=5.65Hz,1H),4.49(t,J=5.65Hz,1H),4.18( br s,1H),3.51-3.35(m,4H),3.14-2.99(m,5H),2.92(dd,J=15.18,4.89Hz,1H),2.65(dd,J=16.06,6.78Hz,1H ),2.53-2.42(m,1H),2.12-1.92(m,3H),1.20(d,J=6.78Hz,3H).
式(I)化合物的绝对构型鉴定Absolute Configuration Identification of Compounds of Formula (I)
Figure PCTCN2022131313-appb-000013
Figure PCTCN2022131313-appb-000013
NOESY图谱(图1)显示,式(I)化合物的3位上的甲基氢与1位上的氢有明显的NOE效应,证明两者在同侧,1位上的吡啶基与3位上的甲基在6元哌啶环上的相对构型为反式,已知3位碳原子的绝对构型为R,因此1位碳原子的绝对构型为S。The NOESY spectrum (Fig. 1) shows that the methyl hydrogen on the 3-position of the compound of formula (I) and the hydrogen on the 1-position have a significant NOE effect, which proves that both are on the same side, and the pyridyl group on the 1-position and the hydrogen on the 3-position The relative configuration of the methyl group on the 6-membered piperidine ring is trans, and the absolute configuration of the carbon atom at the 3-position is known as R, so the absolute configuration of the carbon atom at the 1-position is S.
实施例2:式(I)化合物硫酸盐的制备Embodiment 2: the preparation of formula (I) compound sulfate
将500mg式(I)化合物、10ml异丙醚加入50ml单口瓶中,室温搅拌至固体完全溶清,加入100mg浓硫酸,升温至反应液回流,保温反应1h,降至室温搅拌过夜,抽滤,固体40℃真空干燥3h,得0.58g固体。Add 500mg of the compound of formula (I) and 10ml of isopropyl ether into a 50ml single-necked bottle, stir at room temperature until the solid is completely dissolved, add 100mg of concentrated sulfuric acid, heat up to the reflux of the reaction solution, keep the reaction for 1h, cool down to room temperature and stir overnight, and filter with suction. The solid was dried under vacuum at 40°C for 3 hours to obtain 0.58 g of solid.
实施例3:式(I)化合物磷酸盐的制备Embodiment 3: the preparation of formula (I) compound phosphate
将0.5g式(I)化合物、10ml异丙醚加入50ml单口瓶中,室温搅拌至固体完全溶清,加入118mg磷酸,升温至反应液回流,保温反应1h,降至室温搅拌过夜,抽滤,固体40℃真空干燥3h,得0.53g固体。Add 0.5g of the compound of formula (I) and 10ml of isopropyl ether into a 50ml single-necked bottle, stir at room temperature until the solid is completely dissolved, add 118mg of phosphoric acid, heat up to the reflux of the reaction solution, keep the reaction for 1h, cool down to room temperature and stir overnight, and filter with suction. The solid was dried under vacuum at 40°C for 3 hours to obtain 0.53 g of solid.
实施例4:式(I)化合物富马酸盐的制备Embodiment 4: the preparation of formula (I) compound fumarate
将0.5g式(I)化合物、10ml异丙醇加入50ml单口瓶中,室温搅拌至固体完全溶清,加入118.5mg富马酸,升温至反应液回流,保温反应1h,降至室温搅拌过夜,抽滤,固体40℃真空干燥3h,得0.3g固体。Add 0.5g of the compound of formula (I) and 10ml of isopropanol into a 50ml single-necked bottle, stir at room temperature until the solid is completely dissolved, add 118.5mg of fumaric acid, heat up to the reflux of the reaction solution, keep the reaction for 1h, lower it to room temperature and stir overnight, After suction filtration, the solid was dried under vacuum at 40°C for 3 hours to obtain 0.3 g of solid.
实施例5:式(I)化合物L-苹果酸盐的制备Embodiment 5: the preparation of formula (I) compound L-malate
将50mg式(I)化合物、1ml异丙醚加入5ml离心管,室温搅拌至固体完全溶清,加入13.7mg L-苹果酸,室温搅拌过夜,抽滤,固体室温真空干燥3h,得30mg固体。Add 50 mg of the compound of formula (I) and 1 ml of isopropyl ether into a 5 ml centrifuge tube, stir at room temperature until the solid is completely dissolved, add 13.7 mg of L-malic acid, stir at room temperature overnight, filter with suction, and dry the solid in vacuum at room temperature for 3 hours to obtain 30 mg of solid.
实施例6:式(I)化合物柠檬酸盐的制备Embodiment 6: the preparation of formula (I) compound citrate
将50mg式(I)化合物、1ml甲基叔丁基醚加入5ml离心管,室温搅拌至固体完全溶清,加入21.5mg柠檬酸,室温搅拌3h,抽滤,固体室温真空干燥3h,得26mg固体。Add 50 mg of compound of formula (I) and 1 ml of methyl tert-butyl ether into a 5 ml centrifuge tube, stir at room temperature until the solid is completely dissolved, add 21.5 mg of citric acid, stir at room temperature for 3 h, filter with suction, and dry the solid in vacuum at room temperature for 3 h to obtain 26 mg of solid .
实施例7:式(I)化合物己二酸盐C晶型的制备Embodiment 7: the preparation of formula (I) compound adipate C crystal form
将0.7g己二酸和19ml异丙醇加入100ml单口瓶中,升温至60℃,固体溶清。将2.36g式(I)化合物和19ml异丙醇加入100ml单口瓶中,室温搅拌溶清后滴入己二酸异丙醇溶液,室温搅拌过夜,析晶,抽滤,室温真空干燥3h,得1.66g固体。所得固体经X-射线粉末衍射谱(X-ray Powder Diffraction,XRPD)确认为晶体形态(C晶型),C晶型的XRPD图谱见图2,其DSC图谱见图3,TGA图谱见图4,其DSC吸热峰峰值在149.89℃附近,其XRPD衍射峰位置如下表1所示。Add 0.7g of adipic acid and 19ml of isopropanol into a 100ml single-necked bottle, raise the temperature to 60°C, and dissolve the solid. Add 2.36g of the compound of formula (I) and 19ml of isopropanol into a 100ml single-necked bottle, stir at room temperature to dissolve, then drop into adipate isopropanol solution, stir at room temperature overnight, crystallize, filter with suction, and dry in vacuum at room temperature for 3h to obtain 1.66 g solid. The obtained solid was confirmed to be a crystal form (C crystal form) by X-ray Powder Diffraction (XRPD). , its DSC endothermic peak is around 149.89°C, and its XRPD diffraction peak position is shown in Table 1 below.
表1Table 1
Figure PCTCN2022131313-appb-000014
Figure PCTCN2022131313-appb-000014
Figure PCTCN2022131313-appb-000015
Figure PCTCN2022131313-appb-000015
1H NMR(400MHz,DMSO)δ10.60(s,1H),7.85(d,J=2.7Hz,1H),7.39(t,J=9.5Hz,1H),7.26(d,J=8.0Hz,1H),7.11–6.86(m,4H),5.99(d,J=6.8Hz,1H),4.91(s,1H),4.53(t,J=6.0Hz,1H),4.42(t,J=6.0Hz,1H),3.85(d,J=3.2Hz,1H),3.55(dq,J=20.2,10.0Hz,1H),3.35–3.25(m,1H),3.01(dq,J=19.3,9.6Hz,1H),2.81(dd,J=9.2,7.0Hz,1H),2.71–2.52(m,3H),2.49–2.41(m,3H),2.34(dd,J=9.3,4.5Hz,1H),2.25–2.13(m,5H),1.87–1.72(m,2H),1.61–1.52(m,1H),1.52–1.41(m,4H),1.13(d,J=6.7Hz,3H). 1 H NMR (400MHz, DMSO) δ10.60(s, 1H), 7.85(d, J=2.7Hz, 1H), 7.39(t, J=9.5Hz, 1H), 7.26(d, J=8.0Hz, 1H), 7.11–6.86(m, 4H), 5.99(d, J=6.8Hz, 1H), 4.91(s, 1H), 4.53(t, J=6.0Hz, 1H), 4.42(t, J=6.0 Hz,1H),3.85(d,J=3.2Hz,1H),3.55(dq,J=20.2,10.0Hz,1H),3.35–3.25(m,1H),3.01(dq,J=19.3,9.6Hz ,1H),2.81(dd,J=9.2,7.0Hz,1H),2.71–2.52(m,3H),2.49–2.41(m,3H),2.34(dd,J=9.3,4.5Hz,1H), 2.25–2.13(m,5H),1.87–1.72(m,2H),1.61–1.52(m,1H),1.52–1.41(m,4H),1.13(d,J=6.7Hz,3H).
实施例8:式(I)化合物苯甲酸盐B晶型的制备Embodiment 8: the preparation of formula (I) compound benzoate B crystal form
将3.0g式(I)化合物、100ml异丙醚加入250ml单口瓶中,室温搅拌至固体完全溶清,加入0.75g苯甲酸,室温搅拌过夜,抽滤,将滤饼及30ml异丙醚加100ml单口瓶中室温搅拌1h,析晶,抽滤,室温真空干燥所得滤饼3h,得3.11g固体。所得固体经X-射线粉末衍射谱确认为晶体形态(B晶型),该结晶固体的XRPD图谱见图5,其DSC图谱见图6,TGA图谱见图7,其DSC吸热峰峰值在150.08℃附近,其XRPD衍射峰位置如下表2所示。Add 3.0g of compound of formula (I) and 100ml of isopropyl ether into a 250ml single-necked bottle, stir at room temperature until the solid is completely dissolved, add 0.75g of benzoic acid, stir at room temperature overnight, filter with suction, add 100ml of filter cake and 30ml of isopropyl ether Stir in a one-necked flask at room temperature for 1 h, crystallize, filter with suction, and vacuum-dry the obtained filter cake at room temperature for 3 h to obtain 3.11 g of solid. The obtained solid is confirmed to be a crystal form (B crystal form) by X-ray powder diffraction spectrum. The XRPD spectrum of the crystalline solid is shown in Figure 5, its DSC spectrum is shown in Figure 6, and the TGA spectrum is shown in Figure 7. Its DSC endothermic peak peak is at 150.08 Around ℃, the XRPD diffraction peak positions are shown in Table 2 below.
表2Table 2
Figure PCTCN2022131313-appb-000016
Figure PCTCN2022131313-appb-000016
Figure PCTCN2022131313-appb-000017
Figure PCTCN2022131313-appb-000017
1H NMR(400MHz,DMSO)δ10.60(s,1H),7.98–7.91(m,2H),7.85(d,J=2.7Hz,1H),7.62(t,J=7.4Hz,1H),7.50(t,J=7.6Hz,2H),7.40(d,J=7.6Hz,1H),7.26(d,J=8.0Hz,1H),7.09–6.99(m,2H),6.97–6.89(m,2H),6.00(d,J=6.8Hz,1H),4.90(s,1H),4.54(t,J=6.0Hz,1H),4.42(t,J=6.0Hz,1H),3.85(s,2H),3.01(dt,J=25.3,9.6Hz,2H),2.82(dd,J=9.2,6.9Hz,1H),2.71–2.54(m,3H),2.45(dd,J=11.9,7.3Hz,3H),2.35(dd,J=9.4,4.5Hz,1H),2.19(dt,J=13.4,7.8Hz,1H),1.91–1.73(m,2H),1.55(dt,J=12.7,6.3Hz,1H),1.13(d,J=6.7Hz,3H). 1 H NMR (400MHz,DMSO)δ10.60(s,1H),7.98–7.91(m,2H),7.85(d,J=2.7Hz,1H),7.62(t,J=7.4Hz,1H), 7.50(t, J=7.6Hz, 2H), 7.40(d, J=7.6Hz, 1H), 7.26(d, J=8.0Hz, 1H), 7.09–6.99(m, 2H), 6.97–6.89(m ,2H),6.00(d,J=6.8Hz,1H),4.90(s,1H),4.54(t,J=6.0Hz,1H),4.42(t,J=6.0Hz,1H),3.85(s ,2H),3.01(dt,J=25.3,9.6Hz,2H),2.82(dd,J=9.2,6.9Hz,1H),2.71–2.54(m,3H),2.45(dd,J=11.9,7.3 Hz, 3H), 2.35(dd, J=9.4, 4.5Hz, 1H), 2.19(dt, J=13.4, 7.8Hz, 1H), 1.91–1.73(m, 2H), 1.55(dt, J=12.7, 6.3Hz, 1H), 1.13(d, J=6.7Hz, 3H).
实施例9:式(I)化合物琥珀酸盐的制备Embodiment 9: the preparation of formula (I) compound succinate
将5.0g式(I)化合物、100ml异丙醚装入250ml单口瓶中,室温搅拌至固体完全溶清。加入1.21g琥珀酸,室温搅拌过夜,抽滤,室温真空干燥3h,得5.4g的式(I)化合物琥珀酸盐。Put 5.0 g of the compound of formula (I) and 100 ml of isopropyl ether into a 250 ml single-necked bottle, and stir at room temperature until the solid is completely dissolved. Add 1.21 g of succinic acid, stir overnight at room temperature, filter with suction, and dry in vacuum at room temperature for 3 hours to obtain 5.4 g of the compound of formula (I) succinate.
实施例10:式(I)化合物琥珀酸盐A晶型的制备Embodiment 10: Preparation of crystal form A of compound succinate of formula (I)
实施例10-1:将实施例9得到的琥珀酸盐加入甲醇溶清,减压浓缩得泡沫状固体。称取泡沫状固体500mg,将乙醇(4.5ml)、水(0.5ml)加入10ml单口瓶中,室温搅拌,固体溶清,保温搅拌2h析出固体,抽滤,室温真空干燥所得固体3h,得0.22g的式(I)化合物琥珀酸盐A晶型。所得固体经X-射线粉末衍射谱确认,该结晶固体的XRPD图谱见图8,其DSC图谱见图9,TGA图谱见图10,其DSC吸热峰峰值在186.82℃附近,其XRPD衍射峰位置如下表3所示。Example 10-1: The succinate obtained in Example 9 was dissolved in methanol, and concentrated under reduced pressure to obtain a foamy solid. Weigh 500 mg of foamy solid, add ethanol (4.5 ml) and water (0.5 ml) into a 10 ml single-necked bottle, stir at room temperature, the solid dissolves, keep stirring for 2 hours to precipitate a solid, filter it with suction, and dry the obtained solid in vacuum at room temperature for 3 hours to obtain 0.22 g Crystal form A of the succinate salt of the compound of formula (I). The obtained solid was confirmed by X-ray powder diffraction spectrum. The XRPD spectrum of the crystalline solid is shown in Figure 8, its DSC spectrum is shown in Figure 9, and its TGA spectrum is shown in Figure 10. As shown in Table 3 below.
表3table 3
Figure PCTCN2022131313-appb-000018
Figure PCTCN2022131313-appb-000018
Figure PCTCN2022131313-appb-000019
Figure PCTCN2022131313-appb-000019
1H NMR(400MHz,DMSO)δ10.59(s,1H),7.86(d,J=2.6Hz,1H),7.40(d,J=7.7Hz,1H),7.26(d,J=7.9Hz,1H),7.08(d,J=8.5Hz,1H),7.02(t,J=7.5Hz,1H),6.98–6.91(m,2H),6.01(d,J=6.4Hz,1H),4.91(s,1H),4.54(t,J=6.0Hz,1H),4.42(t,J=6.0Hz,1H),3.89(s,1H),3.54(dq,J=20.1,10.0Hz,1H),3.31(dq,J=13.3,6.5Hz,1H),3.02(dq,J=19.3,9.6Hz,1H),2.89(dd,J=9.4,6.9Hz,1H),2.67(dt,J=8.8,6.4Hz,2H),2.59–2.52(m,4H),2.43(dd,J=9.2,4.1Hz,1H),2.40(s,4H),2.21(td,J=13.7,7.8Hz,1H),1.90–1.75(m,2H),1.59(td,J=12.6,7.0Hz,1H),1.14(d,J=6.7Hz,3H). 1 H NMR (400MHz, DMSO) δ10.59(s, 1H), 7.86(d, J=2.6Hz, 1H), 7.40(d, J=7.7Hz, 1H), 7.26(d, J=7.9Hz, 1H), 7.08(d, J=8.5Hz, 1H), 7.02(t, J=7.5Hz, 1H), 6.98–6.91(m, 2H), 6.01(d, J=6.4Hz, 1H), 4.91( s,1H),4.54(t,J=6.0Hz,1H),4.42(t,J=6.0Hz,1H),3.89(s,1H),3.54(dq,J=20.1,10.0Hz,1H), 3.31(dq, J=13.3,6.5Hz,1H),3.02(dq,J=19.3,9.6Hz,1H),2.89(dd,J=9.4,6.9Hz,1H),2.67(dt,J=8.8, 6.4Hz, 2H), 2.59–2.52(m, 4H), 2.43(dd, J=9.2, 4.1Hz, 1H), 2.40(s, 4H), 2.21(td, J=13.7, 7.8Hz, 1H), 1.90–1.75(m,2H),1.59(td,J=12.6,7.0Hz,1H),1.14(d,J=6.7Hz,3H).
式(I)化合物琥珀酸盐A晶型还可通过以下实施例制备得到:在5ml离心管中加入100mg上述泡沫状固体与500μL溶剂(见表4),室温搅拌1-24小时,抽滤出固体,并将其于室温真空干燥。详细见表4。The crystal form A of compound succinate of formula (I) can also be prepared by the following examples: add 100 mg of the above-mentioned foamy solid and 500 μL of solvent (see Table 4) to a 5 ml centrifuge tube, stir at room temperature for 1-24 hours, and filter out solid and dried under vacuum at room temperature. See Table 4 for details.
表4Table 4
实施例Example 溶剂solvent 晶型crystal form
10-210-2 异丙醚isopropyl ether A晶型Form A
10-310-3 甲基叔丁基醚methyl tert-butyl ether A晶型Form A
实施例11:式(I)化合物琥珀酸盐单晶研究Embodiment 11: Research on single crystal of compound succinate of formula (I)
可通过如下方法制备式(I)化合物琥珀酸盐的单晶:取约10mg的上述式(I)化合物琥珀酸盐(由实施例9制备得到)置于3mL样品瓶中,加入1500μL异丙醚与900μL四氢呋喃,超声溶解后,用封口膜将瓶口密封并用针头在封口膜上扎一个小孔,室温静置3天,析出晶体,制得,所得单晶样品经X-粉末衍射检测为A晶型。A single crystal of the succinate of the compound of formula (I) can be prepared by the following method: take about 10 mg of the succinate of the compound of the formula (I) (prepared in Example 9) in a 3 mL sample bottle, add 1500 μL of isopropyl ether and 900 μL tetrahydrofuran, after ultrasonic dissolution, seal the bottle mouth with a parafilm and pierce a small hole in the parafilm with a needle, leave it at room temperature for 3 days, and precipitate crystals, and the obtained single crystal sample is detected by X-powder diffraction as A crystal form.
对所得单晶进行单晶X-射线衍射数据收集并解析其单晶结构。其单晶结构数据如表5所示。Single crystal X-ray diffraction data collection was performed on the obtained single crystal and its single crystal structure was analyzed. Its single crystal structure data are shown in Table 5.
根据单晶衍射结果分析,单晶属四方晶系,空间群I 4。晶胞参数:
Figure PCTCN2022131313-appb-000020
Figure PCTCN2022131313-appb-000021
α=90°,β=90°,γ=90°,晶胞体积
Figure PCTCN2022131313-appb-000022
可靠因子R1=0.0891,wR2=0.2852,S=0.992。化学计量式为C 30H 37F 4N 5O 4,计算不对称单元分子量为607.64,单个晶胞中含有8个不对称单元,每个不对称单元含有1个游离碱分子与1个琥珀酸分子,如图11和图12所示。
According to the analysis of the single crystal diffraction results, the single crystal belongs to the tetragonal crystal system and the space group I 4. Cell parameters:
Figure PCTCN2022131313-appb-000020
Figure PCTCN2022131313-appb-000021
α=90°, β=90°, γ=90°, unit cell volume
Figure PCTCN2022131313-appb-000022
Reliability factor R1=0.0891, wR2=0.2852, S=0.992. The stoichiometric formula is C 30 H 37 F 4 N 5 O 4 , and the calculated molecular weight of the asymmetric unit is 607.64. There are 8 asymmetric units in a single unit cell, and each asymmetric unit contains 1 free base molecule and 1 succinic acid molecules, as shown in Figure 11 and Figure 12.
确认式(I)化合物琥珀酸盐A晶型的绝对构型为:Confirm that the absolute configuration of formula (I) compound succinate A crystal form is:
Figure PCTCN2022131313-appb-000023
Figure PCTCN2022131313-appb-000023
表5:单晶X-射线衍射数据Table 5: Single crystal X-ray diffraction data
Figure PCTCN2022131313-appb-000024
Figure PCTCN2022131313-appb-000024
Figure PCTCN2022131313-appb-000025
Figure PCTCN2022131313-appb-000025
生物学活性及相关性质的测试Tests for Biological Activity and Related Properties
测试例1、式(I)化合物及其可药用盐饱和溶解度试验Test example 1, formula (I) compound and its pharmaceutically acceptable salt saturation solubility test
实验仪器:回旋式水浴恒温振荡器(太仓文尔惠金,型号DSHZ-300A);高效液相色谱仪(安捷伦1260)。Experimental equipment: rotary water bath constant temperature oscillator (Taicang Wener Huijin, model DSHZ-300A); high performance liquid chromatography (Agilent 1260).
实验方法:精密称取供试样品约10mg,置10ml离心管中,加水1ml,至水浴恒温振荡器中振摇(37℃,150rpm),若样品完全溶解,则继续加入样品10mg,振摇约24小时,取上清液,用乙腈进行适当稀释后进行HPLC检测,按外标法测定其浓度。Experimental method: Accurately weigh about 10 mg of the sample to be tested, put it in a 10 ml centrifuge tube, add 1 ml of water, and shake in a water bath constant temperature shaker (37 ° C, 150 rpm). If the sample is completely dissolved, continue to add 10 mg of the sample and shake for about After 24 hours, take the supernatant, perform HPLC detection after appropriate dilution with acetonitrile, and determine its concentration by the external standard method.
实验结果:待测化合物的饱和溶解度如表6所示,可见式(I)化合物琥珀酸盐、苯甲酸盐和己二酸盐的24h饱和溶解度显著优于游离碱。Experimental results: the saturated solubility of the compounds to be tested is shown in Table 6. It can be seen that the 24h saturated solubility of the compound of formula (I) succinate, benzoate and adipate is significantly better than that of the free base.
表6:饱和溶解度试验结果Table 6: Saturated Solubility Test Results
Figure PCTCN2022131313-appb-000026
Figure PCTCN2022131313-appb-000026
测试例2、式(I)化合物及其可药用盐的异构体杂质考察试验Test example 2, investigation test of isomer impurity of formula (I) compound and pharmaceutically acceptable salt thereof
试验方法:将式(I)化合物琥珀酸盐(实施例10-1,A晶型)、式(I)化合物己二酸盐(实施例7,C晶型)、式(I)化合物(实施例1)均采用两层聚乙烯、一层药用复合膜包装,重点考察在高温、光照、高湿、加速等条件下式(I)化合物的异构体杂质的含量变化,结果如表7所示。异构体杂质分别为异构体1和异构体2,结构如下所示。N.D表示未检测到该杂质。Test method: formula (I) compound succinate (embodiment 10-1, A crystal form), formula (I) compound adipate (embodiment 7, C crystal form), formula (I) compound (implementation Example 1) are packaged with two layers of polyethylene and one layer of pharmaceutical composite film, focusing on the changes in the content of isomer impurities of the compound of formula (I) under conditions such as high temperature, light, high humidity, and acceleration. The results are shown in Table 7 shown. The isomer impurities are isomer 1 and isomer 2, respectively, and the structures are shown below. N.D means the impurity was not detected.
试验结果:式(I)化合物琥珀酸盐和式(I)化合物己二酸盐在高温、光照、加速和高湿条件下,其异构体杂质均未明显增长,游离状态的式(I)化合物在5天的高温条件下,其异构体1杂质的含量出现较明显的增长。另外游离状态的式(I)化合物在高温、光照、加速和高湿条件下检测到新的异构体2杂质。该试验证明化合物的上述盐型热稳定性更优,且相较于游离碱能更有效地抑制异构体杂质的产生。Test result: formula (I) compound succinate and formula (I) compound adipate are under high temperature, light, acceleration and high humidity conditions, and its isomer impurity all does not increase obviously, the free state formula (I) The content of the isomer 1 impurity of the compound increased significantly under the high temperature condition for 5 days. In addition, the compound of formula (I) in the free state is detected as a new impurity of isomer 2 under high temperature, light, acceleration and high humidity conditions. This test proves that the above-mentioned salt form of the compound has better thermal stability, and can more effectively suppress the generation of isomer impurities compared with the free base.
Figure PCTCN2022131313-appb-000027
Figure PCTCN2022131313-appb-000027
表7:异构体杂质考察试验结果Table 7: Test results of isomer impurity investigation
Figure PCTCN2022131313-appb-000028
Figure PCTCN2022131313-appb-000028
Figure PCTCN2022131313-appb-000029
Figure PCTCN2022131313-appb-000029
测试例3、式(I)化合物可药用盐稳定性考察试验Test example 3, formula (I) compound pharmaceutically acceptable salt stability investigation test
试验方法:将式(I)化合物琥珀酸盐(实施例10-1,A晶型)、式(I)化合物苯甲酸盐(实施例8,B晶型)、式(I)化合物己二酸盐(实施例7,C晶型)均采用两层聚乙烯、一层药用复合膜包装,考察在高温、光照、加速和高湿等条件下的杂质数量和杂质总量的变化,结果如表8所示。Test method: the formula (I) compound succinate (embodiment 10-1, A crystal form), the formula (I) compound benzoate (embodiment 8, B crystal form), the formula (I) compound hexadiene Salt (embodiment 7, C crystal form) all adopts two layers of polyethylene, one layer of pharmaceutical composite film packing, investigates the change of impurity amount and impurity total amount under conditions such as high temperature, illumination, acceleration and high humidity, the result As shown in Table 8.
试验结果:式(I)化合物琥珀酸盐、式(I)化合物苯甲酸盐和式(I)化合物己二酸盐均能较好地控制总杂含量,特别是式(I)化合物琥珀酸盐和式(I)化合物己二酸盐在高温、光照、加速和高湿等条件下,其总杂含量和杂质增长率这两方面均表现出色。Test result: formula (I) compound succinate, formula (I) compound benzoate and formula (I) compound adipate all can control total impurity content preferably, especially formula (I) compound succinic acid The salt and the adipate salt of the compound of formula (I) perform well in both aspects of total impurity content and impurity growth rate under conditions such as high temperature, light, acceleration and high humidity.
表8:稳定性考察试验结果Table 8: Stability investigation test results
Figure PCTCN2022131313-appb-000030
Figure PCTCN2022131313-appb-000030
测试例4:式(I)化合物及其可药用盐的大鼠药代动力学评价Test Example 4: Rat Pharmacokinetic Evaluation of Compounds of Formula (I) and Pharmaceutically Acceptable Salts thereof
实验材料:Experimental Materials:
SD雌性大鼠购自北京维通利华实验动物技术有限公司。SD female rats were purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd.
MC(甲基纤维素)购自Sigma。MC (methylcellulose) was purchased from Sigma.
乙腈购自Merck(USA)。Acetonitrile was purchased from Merck (USA).
实验方法:experimental method:
雌性SD大鼠12只(200-300g,6-8周),随机分成4组,每组3只。分别口服给予式(I)化合物、式(I)化合物苯甲酸盐、式(I)化合物琥珀酸盐和式(I)化合物己二酸盐,剂量24mg/kg,溶媒0.5%甲基纤维素水溶液。动物实验前正常喂水,禁食过夜,在给药后4h恢复食物供给。每组大鼠于给药前及给药后0.25、0.5、1、2、4、6、8、24h和48h进行颈静脉采血。收集的全血样品置于K 2EDTA抗凝管中,离心5min后(12000rpm,4℃)取血浆待测。 Twelve female SD rats (200-300g, 6-8 weeks) were randomly divided into 4 groups, with 3 rats in each group. Oral administration of formula (I) compound, formula (I) compound benzoate, formula (I) compound succinate and formula (I) compound adipate, dose 24mg/kg, vehicle 0.5% methylcellulose aqueous solution. Animals were fed water normally before the experiment, fasted overnight, and food supply was resumed 4 hours after administration. Rats in each group were collected from the jugular vein before administration and at 0.25, 0.5, 1, 2, 4, 6, 8, 24h and 48h after administration. The collected whole blood samples were placed in K 2 EDTA anticoagulant tubes, centrifuged for 5 minutes (12000 rpm, 4° C.) and plasma was collected for testing.
取大鼠血浆样品10μL,加入150μL乙腈溶剂(其中含内标化合物)沉淀蛋白,涡旋5min后,离心(14000rpm)5min,上清液用含0.1%(v/v)FA的水稀释2倍,于LC-MS/MS系统 (AB Sciex Triple Quad 6500+)进行定量检测。在测定样品浓度时随行SD大鼠血浆标准曲线和质控样品。对10x稀释样品,取2μL样品加入18μL的空白血浆,涡旋0.5min后,加入300μL乙腈溶剂(其中含内标化合物)沉淀蛋白,其余处理步骤同不稀释样品。Take 10 μL of rat plasma sample, add 150 μL of acetonitrile solvent (including internal standard compound) to precipitate protein, vortex for 5 minutes, centrifuge (14000rpm) for 5 minutes, and dilute the supernatant 2 times with water containing 0.1% (v/v) FA , quantitatively detected on LC-MS/MS system (AB Sciex Triple Quad 6500+). SD rat plasma standard curve and quality control samples were accompanied by the determination of sample concentration. For 10x diluted samples, take 2 μL sample and add 18 μL blank plasma, vortex for 0.5 min, add 300 μL acetonitrile solvent (including internal standard compound) to precipitate protein, and the rest of the processing steps are the same as for undiluted samples.
实验结果:Experimental results:
相对于游离碱,式(I)化合物苯甲酸盐、式(I)化合物琥珀酸盐和式(I)化合物己二酸盐均取得了更为优异的PK特性,具有良好的口服生物利用度。Compared with free base, formula (I) compound benzoate, formula (I) compound succinate and formula (I) compound adipate have achieved more excellent PK characteristics, and have good oral bioavailability .
表9式(I)化合物及其可药用盐的大鼠PK测试结果The rat PK test result of table 9 formula (I) compound and pharmaceutically acceptable salt thereof
Figure PCTCN2022131313-appb-000031
Figure PCTCN2022131313-appb-000031
测试例5式(I)化合物琥珀酸盐A晶型对乳腺癌细胞的体外增殖抑制研究Test Example 5 Study on In vitro Proliferation Inhibition of Formula (I) Compound Succinate A Crystal Form on Breast Cancer Cells
实验材料:MCF-7,CAMA-1,HCC1500,T47D,及SK-BR-3细胞均购自ATCC,EFM-19及MCF-7 ESR1 Y537S突变株购自科佰生物。细胞培养条件参照ATCC或科佰产品说明书,培养基购自Invitrogen,血清购自Gibico。MCF-7培养基为DMEM+10%FBS+0.01mg/ml人胰岛素(翌圣)+1%非必需氨基酸。CAMA-1培养基为EMEM+10%FBS.HCC1500及EFM-19培养基为RPMI-1640+10%FBS。T47D培养基为RPMI-1640+10%HI-FBS+2unit/mL牛胰岛素(Solarbio)。MCF-7 ESR1 Y537S培养基为MEM+10%FBS+1mM丙酮酸钠+1%非必需氨基酸。SK-BR-3培养基为McCoy’s 5A+10%FBS。氟维司群购自MCE。式(I)化合物琥珀酸盐A晶型根据实施例10-1方法制备得到。Experimental materials: MCF-7, CAMA-1, HCC1500, T47D, and SK-BR-3 cells were purchased from ATCC, and EFM-19 and MCF-7 ESR1 Y537S mutant strains were purchased from Kebai Biotech. The cell culture conditions refer to ATCC or Kebai product instructions, the medium is purchased from Invitrogen, and the serum is purchased from Gibico. MCF-7 medium is DMEM+10%FBS+0.01mg/ml human insulin (Yisheng)+1% non-essential amino acid. CAMA-1 medium is EMEM+10% FBS. HCC1500 and EFM-19 medium are RPMI-1640+10% FBS. T47D medium is RPMI-1640+10% HI-FBS+2 unit/mL bovine insulin (Solarbio). MCF-7 ESR1 Y537S medium is MEM + 10% FBS + 1mM sodium pyruvate + 1% non-essential amino acids. SK-BR-3 medium is McCoy's 5A+10% FBS. Fulvestrant was purchased from MCE. The crystal form A of succinate salt of the compound of formula (I) was prepared according to the method in Example 10-1.
实验方法:培养细胞融合率达到70%以上时,消化细胞,MCF-7,T47D,MCF ESR1 Y537S细胞密度调整为500/20μL/孔,HCC1500细胞密度调整为2000/20μL/孔,CAMA-1,EFM-19以及SK-BR-3细胞密度调整为1000/20μL/孔种在384孔细胞培养板中(Corning)培养过夜。使用Echo650(Beckman)转移120nL 3倍稀释化合物至细胞板中,补加40μL相应培养基,化合物处理7天后,加入CellTiter-Glo(Promega)检测细胞活力,细胞孔作为0%抑制对照,0.1μM氟维司群处理孔作为100%抑制对照(其中MCF-7 ESR1 Y537S使用2μM氟维司群处理孔作为100%抑制对照,SK-BR-3使用培养基孔作为100%抑制对照)。使用IDBS XLfit进行4参数拟合计算IC 50及最大抑制率数据。 Experimental method: when the fusion rate of the cultured cells reaches more than 70%, digest the cells, adjust the density of MCF-7, T47D, MCF ESR1 Y537S cells to 500/20 μL/well, adjust the density of HCC1500 cells to 2000/20 μL/well, CAMA-1, EFM-19 and SK-BR-3 cells were adjusted to a density of 1000/20 μL/well and cultured overnight in a 384-well cell culture plate (Corning). Use Echo650 (Beckman) to transfer 120nL of 3-fold diluted compound to the cell plate, add 40 μL of corresponding medium, after 7 days of compound treatment, add CellTiter-Glo (Promega) to detect cell viability, the cell well is used as 0% inhibition control, 0.1 μM fluorine Wells treated with fulvestrant were used as 100% inhibition control (wells treated with 2 μM fulvestrant for MCF-7 ESR1 Y537S were used as 100% inhibition control, and medium wells were used as 100% inhibition control for SK-BR-3). Using IDBS XLfit to perform 4-parameter fitting to calculate IC 50 and maximum inhibition rate data.
实验结果:式(I)化合物琥珀酸盐A晶型对不同ER阳性乳腺癌细胞株均表现出较强的增殖抑制作用,IC 50<1nM,对ESR1 Y537S突变株增殖抑制IC 50为27.6nM,对ER阴性细胞SK-BR-3最高2μM浓度下无增殖抑制作用。 Experimental results: the succinate A crystal form of the compound of formula (I) showed strong proliferation inhibitory effects on different ER-positive breast cancer cell lines, with IC 50 <1nM, and the IC 50 for proliferation inhibition of ESR1 Y537S mutant strains was 27.6nM, SK-BR-3 has no inhibitory effect on the proliferation of ER-negative cells at the highest concentration of 2 μM.
表10式(I)化合物琥珀酸盐A晶型对乳腺癌细胞增殖抑制作用Table 10 formula (I) compound succinate A crystal form inhibits breast cancer cell proliferation
细胞株cell line IC 50_nM IC50_nM 最大抑制率%Maximum inhibition rate%
MCF7MCF7 0.780.78 99.5999.59
MCF-7 ESR1 Y537SMCF-7 ESR1 Y537S 27.6027.60 100.96100.96
HCC1500HCC1500 0.940.94 90.3290.32
CAMA-1CAMA-1 0.380.38 97.6697.66
EFM-19EFM-19 0.400.40 110.99110.99
T47DT47D 0.550.55 94.2994.29
SK-BR-3(ER -) SK-BR-3(ER - ) >2000>2000 -2.5-2.5
测试例6式(I)化合物琥珀酸盐A晶型对MCF-7及MCF-7 ESR1 Y537S突变株ER蛋白Test example 6 formula (I) compound succinate A crystal form to MCF-7 and MCF-7 ESR1 Y537S mutant strain ER protein
降解作用Degradation
实验材料:MCF-7购自ATCC,MCF-7 ESR1 Y537S突变株购自科佰生物。细胞培养条件参照ATCC或科佰产品说明书,培养基购自Invitrogen,血清购自Gibico。MCF-7培养基为DMEM+10%FBS+0.01mg/ml人胰岛素(翌圣)+1%非必需氨基酸。CAMA-1培养基为EMEM+10%FBS。MCF-7 ESR1 Y537S培养基为MEM+10%FBS+1mM丙酮酸钠+1%非必需氨基酸。氟维司群购自MCE。式(I)化合物琥珀酸盐A晶型根据实施例10-1方法制备得到。Experimental materials: MCF-7 was purchased from ATCC, and the MCF-7 ESR1 Y537S mutant strain was purchased from Kebai Biotech. The cell culture conditions refer to ATCC or Kebai product instructions, the medium is purchased from Invitrogen, and the serum is purchased from Gibico. MCF-7 medium is DMEM+10%FBS+0.01mg/ml human insulin (Yisheng)+1% non-essential amino acid. CAMA-1 medium is EMEM+10% FBS. MCF-7 ESR1 Y537S medium is MEM + 10% FBS + 1mM sodium pyruvate + 1% non-essential amino acids. Fulvestrant was purchased from MCE. The crystal form A of succinate salt of the compound of formula (I) was prepared according to the method in Example 10-1.
实验方法:培养细胞融合率达到70%以上时,消化细胞,调整细胞密度为5000/20μL/孔,种在384孔细胞培养板中(Greiner)培养过夜。使用Echo650(Beckman)转移100nL 3倍稀释化合物至细胞板中,24小时后加入20μL 8%甲醛(沪试)室温固定20分钟,加入50μL冰甲醇(Millipore)透膜处理10分钟,加入50μL Licor blocking buffer室温封闭1小时,加入20μL ER抗体(CST)及GAPDH抗体(Abcam)4度孵育过夜。洗掉一抗,加入IRDye 800羊抗兔IgG抗体及IRDye 680羊抗鼠IgG抗体(LI-COR)避光孵育45分钟,洗掉二抗,倒置1200转每分离心1分钟去除残余液体,使用Sapphire RGBNIR(Azure)检测800nM及680nM信号。使用AzureSpot进行荧光定量分析,计算Chanel 800(ER)/Chanel 680(GAPDH)数值,细胞孔作为0%抑制对照,MCF-7使用0.1μM氟维司群处理孔作为100%抑制对照,MCF-7 ESR1 Y537S使用2μM氟维司群处理孔作为100%降解。使用IDBS XLfit进行4参数拟合计算DC 50及最大降解率数据。 Experimental method: when the fusion rate of the cultured cells reaches more than 70%, digest the cells, adjust the cell density to 5000/20 μL/well, seed in a 384-well cell culture plate (Greiner) and culture overnight. Use Echo650 (Beckman) to transfer 100nL 3-fold diluted compound to the cell plate, add 20μL 8% formaldehyde (Shanghai test) after 24 hours to fix at room temperature for 20 minutes, add 50μL ice methanol (Millipore) to permeate the membrane for 10 minutes, add 50μL Licor blocking The buffer was blocked at room temperature for 1 hour, and 20 μL of ER antibody (CST) and GAPDH antibody (Abcam) were added to incubate overnight at 4°C. Wash off the primary antibody, add IRDye 800 goat anti-rabbit IgG antibody and IRDye 680 goat anti-mouse IgG antibody (LI-COR) and incubate in the dark for 45 minutes, wash off the secondary antibody, invert 1200 rpm and centrifuge for 1 minute to remove residual liquid, use Sapphire RGBNIR (Azure) detects 800nM and 680nM signals. Use AzureSpot for fluorescence quantitative analysis, calculate Chanel 800(ER)/Chanel 680(GAPDH) value, cell wells as 0% inhibition control, MCF-7 use 0.1μM fulvestrant treatment wells as 100% inhibition control, MCF-7 ESR1 Y537S wells were treated with 2 μM fulvestrant as 100% degradation. IDBS XLfit was used to perform 4-parameter fitting to calculate DC 50 and maximum degradation rate data.
实验结果:式(I)化合物琥珀酸盐A晶型对MCF-7及ESR1 Y537S突变株均表现出较高的ER降解作用。Experimental results: The succinate A crystal form of the compound of formula (I) showed a high ER degradation effect on MCF-7 and ESR1 Y537S mutant strains.
表11式(I)化合物琥珀酸盐A晶型对MCF-7及ESR1 Y537S突变株ER降解作用Table 11 Formula (I) compound succinate A crystal form on MCF-7 and ESR1 Y537S mutant strain ER degradation
细胞株cell line DC 50_nM DC 50_nM 最大降解率%Maximum degradation rate%
MCF7MCF7 0.730.73 105.09105.09
MCF-7 ESR Y537SMCF-7 ESR Y537S 47.3247.32 99.5799.57
测试例7式(I)化合物琥珀酸盐的人ER阳性乳腺癌MCF-7异种皮下移植瘤小鼠模型药效学研究Pharmacodynamic study of human ER-positive breast cancer MCF-7 xenograft subcutaneous xenograft tumor mouse model of test example 7 formula (I) compound succinate
实验材料:Experimental Materials:
人乳腺癌MCF-7细胞:ECACC,86012803Human breast cancer MCF-7 cells: ECACC, 86012803
17β-雌二醇片:Innovative Research of America,Cat No.:SE-121,60-day release,0.18mg/pellet17β-estradiol tablets: Innovative Research of America, Cat No.: SE-121, 60-day release, 0.18mg/pellet
EMEM培养液:ATCC,Cat No.:30-2003EMEM medium: ATCC, Cat No.: 30-2003
胎牛血清:ExCell;Cat No.:FND500Fetal bovine serum: ExCell; Cat No.: FND500
双抗:Gibco,Cat No.:15240-062Double antibody: Gibco, Cat No.: 15240-062
0.25%胰酶-EDTA:Gibco,Cat No.:25200-0720.25% Trypsin-EDTA: Gibco, Cat No.: 25200-072
DPBS:Corning,Cat.No.:21-031-CVRDPBS: Corning, Cat. No.: 21-031-CVR
基质胶:Corning,Cat.No.:354234Matrigel: Corning, Cat.No.: 354234
氟维司群注射液(Fulvestrant):阿斯利康(AstraZeneca),250mg/5mL/支Fulvestrant injection (Fulvestrant): AstraZeneca, 250mg/5mL/bottle
实验方法:experimental method:
动物信息:Balb/c nude小鼠,雌性,6-8周,体重约18-22克,动物购自上海灵畅生物科技有限公司,将小鼠饲养在SPF级的环境中,每个笼位单独送排风,所有动物都可以自由获取标准认证的商业实验室饮食和自由饮水。Animal information: Balb/c nude mice, female, 6-8 weeks old, weighing about 18-22 grams, were purchased from Shanghai Lingchang Biotechnology Co., Ltd., and the mice were raised in an SPF-grade environment. Each cage Separate exhaust ventilation was provided and all animals had free access to a standard certified commercial laboratory diet and water ad libitum.
细胞培养:人乳腺癌MCF-7细胞株体外培养,培养条件为EMEM(细胞培养液)中加入10%胎牛血清,1%双抗,37℃,5%CO 2孵箱。一周两次用0.25%胰酶-EDTA消化液进行常规消化处理传代。当细胞汇合度为80%-90%,数量达到要求时,收取细胞,计数。 Cell culture: human breast cancer MCF-7 cell line was cultured in vitro, and the culture conditions were 10% fetal bovine serum and 1% double antibody in EMEM (cell culture medium), 37° C., 5% CO 2 incubator. Routine digestion with 0.25% trypsin-EDTA digestion solution was performed twice a week for subculture. When the confluence of the cells is 80%-90% and the number reaches the requirement, the cells are collected and counted.
细胞接种:将0.2ml/(含1×10 7个)MCF-7细胞悬液(DPBS:基质胶,体积比为1:1)皮下接种于每只小鼠的右后背,并于细胞接种前两天皮下接种17β-雌二醇片。在接种细胞后第6天,肿瘤平均体积达到173.65mm 3时开始分组给药,依据肿瘤体积随机分组给药,分组给药当天为第0天(Day 0)。 Cell inoculation: Inoculate 0.2ml/(containing 1× 107 ) MCF-7 cell suspension (DPBS: Matrigel, volume ratio 1:1) subcutaneously on the right back of each mouse, and inoculate 17β-estradiol tablets were subcutaneously inoculated two days before. On the 6th day after cell inoculation, when the average tumor volume reached 173.65 mm 3 , group administration began, and random grouping was administered according to the tumor volume. The day of group administration was Day 0 (Day 0).
给药:式(I)化合物琥珀酸盐(实施例10-1,A晶型)的给药剂量为0.3,1,3或10mg/kg(以游离碱计),口服给药(PO),每天一次给药(QD)x21次。氟维司群(Fulvestrant)的给药剂量为250mg/kg,皮下注射(SC),每周一次给药(QW)x4次。每组8只小鼠。Administration: the administration dose of formula (I) compound succinate (Example 10-1, A crystal form) is 0.3, 1, 3 or 10 mg/kg (calculated as free base), orally administered (PO), Once-daily dosing (QD) x21 times. The dosage of fulvestrant (Fulvestrant) is 250 mg/kg, subcutaneous injection (SC), once a week (QW) x 4 times. 8 mice per group.
肿瘤测量和实验指标:Tumor measurements and experimental indicators:
每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a x b 2,a和b分别表示肿瘤的长径和短径。每周两次测量小鼠体重。 Tumor diameters were measured twice a week with vernier calipers. The formula for calculating the tumor volume is: V=0.5a x b 2 , where a and b represent the long diameter and short diameter of the tumor, respectively. Mouse body weights were measured twice a week.
化合物的抑瘤疗效用肿瘤生长抑制率TGI(%)来评价。TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]x100%。The antitumor efficacy of compounds was evaluated by tumor growth inhibition rate TGI (%). TGI (%)=[(1-(average tumor volume at the end of administration of a certain treatment group-average tumor volume at the beginning of administration of this treatment group)/(average tumor volume at the end of treatment of the solvent control group-at the beginning of treatment of the solvent control group Mean tumor volume)] x 100%.
实验结果:Experimental results:
见表12,图13和图14。虽然有个别小鼠出现了超过10%的体重下降,但是与供试品式(I)化合物琥珀酸盐的剂量无关,而且受试药组的小鼠体重和溶媒对照组相比并无特别显著的降低,考虑到该模型受到雌激素贴片的影响可能会影响小鼠状态和体重,所以这些个别小鼠的体重下降与供试品式(I)化合物琥珀酸盐无关。实验各组均无小鼠死亡。See Table 12, Figure 13 and Figure 14. Although individual mice have more than 10% weight loss, it has nothing to do with the dose of the test product formula (I) compound succinate, and the body weight of the mice in the test drug group is not particularly significant compared with the vehicle control group. Considering that the model is affected by the estrogen patch and may affect the state and body weight of the mice, the weight loss of these individual mice has nothing to do with the test product formula (I) compound succinate. No mouse died in each group of the experiment.
试验结果表明,开始给药后第20天(Day 20),一周一次皮下注射给予人乳腺癌MCF-7荷瘤小鼠250mg/kg对照品Fulvestrant,对MCF-7异种移植肿瘤模型的肿瘤生长有较弱的抑制效果(P<0.05)。一天一次口服给予荷瘤小鼠0.3mg/kg的供试品式(I)化合物琥珀酸盐,和溶媒对照组相比,显示出一定的抑制肿瘤生长的作用(P<0.0001),一天一次口服给予荷瘤小鼠1mg/kg,3mg/kg或10mg/kg的供试品式(I)化合物琥珀酸盐,和溶媒对照组相比,显示出显著的抑制肿瘤生长的作用(P<0.0001),并呈现较好的剂量依赖性,在1mg/kg及以上的剂量下,肿瘤开始消退。在人乳腺癌MCF-7皮下瘤模型上,式(I)化合物琥珀酸盐在1mg/kg及以上的剂量下(PO,QD),显示出了显著优于对照品Fulvestrant(250mg/kg,SC,QW)的抗肿瘤活性(P<0.0001)。The test results show that on the 20th day (Day 20) after the start of administration, subcutaneous injection of Fulvestrant 250 mg/kg into human breast cancer MCF-7 tumor-bearing mice once a week has a significant effect on the tumor growth of the MCF-7 xenograft tumor model. Weak inhibitory effect (P<0.05). Orally administered 0.3mg/kg of the test product formula (I) compound succinate to tumor-bearing mice once a day, compared with the vehicle control group, showed a certain effect of inhibiting tumor growth (P<0.0001), once a day orally Giving tumor-bearing mice 1mg/kg, 3mg/kg or 10mg/kg of the test product formula (I) compound succinate, compared with the vehicle control group, showed a significant effect of inhibiting tumor growth (P<0.0001) , and showed a good dose dependence, at the dose of 1mg/kg and above, the tumor began to regress. On human breast cancer MCF-7 subcutaneous tumor model, formula (I) compound succinate shows significantly better than reference substance Fulvestrant (250mg/kg, SC) at a dose of 1mg/kg and above (PO, QD). , QW) antitumor activity (P<0.0001).
表12 MCF-7皮下瘤模型肿瘤体积Table 12 MCF-7 subcutaneous tumor model tumor volume
Figure PCTCN2022131313-appb-000032
Figure PCTCN2022131313-appb-000032
测试例8式(I)化合物琥珀酸盐的人ER阳性乳腺癌T47D异种皮下移植瘤小鼠模型药效学研究Test Example 8 Pharmacodynamic study of compound succinate of formula (I) on human ER-positive breast cancer T47D xenograft subcutaneous xenograft tumor mouse model
实验材料:Experimental Materials:
人乳腺癌xxT47D细胞,为T47D(ATCC,HTB-133)经过两次体内传代的细胞Human breast cancer xxT47D cells are T47D (ATCC, HTB-133) cells that have been passaged twice in vivo
17β-雌二醇片:Innovative Research of America,Cat No.:SE-121,60-day release,0.18mg/pellet17β-estradiol tablets: Innovative Research of America, Cat No.: SE-121, 60-day release, 0.18mg/pellet
RPMI-1640培养液:Gibco,Cat No.:C22400500BTRPMI-1640 culture medium: Gibco, Cat No.: C22400500BT
胎牛血清:HyClone;Cat No.:SV30087.03Fetal bovine serum: HyClone; Cat No.:SV30087.03
牛胰岛素:源叶,Cat.No.:11070-73-8Bovine insulin: Yuanye, Cat.No.:11070-73-8
双抗:Gibco,Cat No.:15240-062Double antibody: Gibco, Cat No.: 15240-062
0.25%胰酶-EDTA:Gibco,Cat No.:252000720.25% Trypsin-EDTA: Gibco, Cat No.: 25200072
DPBS:Corning,Cat.No.:21-031-CVRDPBS: Corning, Cat. No.: 21-031-CVR
基质胶:Corning,Cat.No.:354234Matrigel: Corning, Cat.No.: 354234
氟维司群注射液(Fulvestrant):阿斯利康(AstraZeneca),250mg/5mL/支Fulvestrant injection (Fulvestrant): AstraZeneca, 250mg/5mL/bottle
实验方法:experimental method:
动物信息:Balb/c nude小鼠,雌性,6-8周,体重约18-22克,动物购自上海灵畅生物科技有限公司,将小鼠饲养在SPF级的环境中,每个笼位单独送排风,所有动物都可以自由获取标准认证的商业实验室饮食和自由饮水。Animal information: Balb/c nude mice, female, 6-8 weeks old, weighing about 18-22 grams, were purchased from Shanghai Lingchang Biotechnology Co., Ltd., and the mice were raised in an SPF-grade environment. Each cage Separate exhaust ventilation was provided and all animals had free access to a standard certified commercial laboratory diet and water ad libitum.
细胞培养:人乳腺癌xxT47D细胞体外培养,培养条件为RPMI-1640培养基中加10%胎牛血清,0.2Units/mL牛胰岛素,1%双抗,37℃5%CO 2孵箱培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞汇合度为80%~90%,数量到达要求时,收取细胞,计数。 Cell culture: Human breast cancer xxT47D cells were cultured in vitro. The culture conditions were RPMI-1640 medium plus 10% fetal bovine serum, 0.2 Units/mL bovine insulin, 1% double antibody, and cultured in a 5% CO 2 incubator at 37°C. Routine digestion with trypsin-EDTA was performed twice a week for passaging. When the confluence of the cells is 80%-90% and the number reaches the requirement, the cells are collected and counted.
细胞接种:将0.2mL(含1×10 7个)xxT47D细胞悬液(DPBS:基质胶,体积比为1:1)皮下接种于每只小鼠的右后背,并于细胞接种前两天皮下接种17β-雌二醇片。在接种细胞后第6天,肿瘤平均体积达到137.09mm 3时开始分组给药,依据肿瘤体积随机分组给药,分组给药当天为第0天(Day 0)。 Cell inoculation: Inoculate 0.2mL (containing 1× 107 ) xxT47D cell suspension (DPBS: Matrigel, volume ratio 1:1) subcutaneously on the right back of each mouse, and two days before cell inoculation Subcutaneous inoculation of 17β-estradiol tablets. On the 6th day after cell inoculation, when the average volume of the tumor reached 137.09 mm 3 , group administration began, and the group was randomly assigned according to the tumor volume, and the day of group administration was Day 0 (Day 0).
给药:式(I)化合物琥珀酸盐(实施例10-1,A晶型)的给药剂量为0.3,1或3mg/kg,口服给药(PO),每天一次给药(QD)x28次。氟维司群(Fulvestrant)的给药剂量为250mg/kg,皮下注射(SC),每周一次给药(QW)x4次。每组8只小鼠。Administration: The dosage of the compound succinate of formula (I) (Example 10-1, A crystal form) is 0.3, 1 or 3mg/kg, administered orally (PO), administered once a day (QD) x28 Second-rate. The dosage of fulvestrant (Fulvestrant) is 250 mg/kg, subcutaneous injection (SC), once a week (QW) x 4 times. 8 mice per group.
肿瘤测量和实验指标:Tumor measurements and experimental indicators:
每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a x b 2,a和b分别表示肿瘤的长径和短径。每周两次测量小鼠体重。 Tumor diameters were measured twice a week with vernier calipers. The formula for calculating the tumor volume is: V=0.5a x b 2 , where a and b represent the long diameter and short diameter of the tumor, respectively. Mouse body weights were measured twice a week.
化合物的抑瘤疗效用肿瘤生长抑制率TGI(%)来评价。TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]x100%。The antitumor efficacy of compounds was evaluated by tumor growth inhibition rate TGI (%). TGI (%)=[(1-(average tumor volume at the end of administration of a certain treatment group-average tumor volume at the beginning of administration of this treatment group)/(average tumor volume at the end of treatment of the solvent control group-at the beginning of treatment of the solvent control group Mean tumor volume)] x 100%.
实验结果:Experimental results:
见表13,图15和图16。See Table 13, Figure 15 and Figure 16.
试验结果表明,在开始给药后第28天(Day 28),一周一次皮下注射给予人乳腺癌T47D荷瘤小鼠250mg/kg对照品Fulvestrant,和溶媒对照组相比,显示出一定的抗肿瘤生长的作用(P<0.0001)。一天一次口服给予荷瘤小鼠0.3mg/kg,1mg/kg或3mg/kg的供试品式(I)化合物琥珀酸盐,和溶媒对照组相比有一定或显著的抑制肿瘤生长的作用(P<0.0001),并呈现较好的剂量依赖性。在人乳腺癌T47D皮下瘤模型上,式(I)化合物琥珀酸盐在1mg/kg及以上的剂量下(PO,QD),显示出了显著优于对照品Fulvestrant(250mg/kg,SC,QW)的抗肿瘤活性(P<0.0001)。The test results showed that on the 28th day (Day 28) after the start of administration, subcutaneous injection of 250mg/kg Fulvestrant to human breast cancer T47D tumor-bearing mice once a week showed a certain anti-tumor effect compared with the vehicle control group. The effect of growth (P<0.0001). Once a day orally administered to tumor-bearing mice 0.3mg/kg, 1mg/kg or 3mg/kg of the test product formula (I) compound succinate, compared with the vehicle control group, there is a certain or significant effect of inhibiting tumor growth ( P<0.0001), and showed a good dose dependence. On human breast cancer T47D subcutaneous tumor model, formula (I) compound succinate shows significantly better than reference substance Fulvestrant (250mg/kg, SC, QW) under the dose (PO, QD) of 1mg/kg and above. ) antitumor activity (P<0.0001).
表13 T47D皮下瘤模型肿瘤体积Table 13 T47D subcutaneous tumor model tumor volume
Figure PCTCN2022131313-appb-000033
Figure PCTCN2022131313-appb-000033
Figure PCTCN2022131313-appb-000034
Figure PCTCN2022131313-appb-000034
测试例9式(I)化合物琥珀酸盐的人ER阳性乳腺癌MCF-7异种脑原位移植瘤小鼠模型药效学研究Test Example 9 Pharmacodynamic study of compound succinate of formula (I) on human ER-positive breast cancer MCF-7 xenograft brain orthotopic transplantation mouse model
实验材料Experimental Materials
人乳腺癌MCF-7细胞:ATCC,HTB-22Human breast cancer MCF-7 cells: ATCC, HTB-22
17β-雌二醇片:Innovative Research of America,SE-121,60-day release,0.72mg/pellet17β-estradiol tablets: Innovative Research of America, SE-121, 60-day release, 0.72mg/pellet
EMEM培养液:ATCC,Cat No.:30-2003EMEM medium: ATCC, Cat No.: 30-2003
胎牛血清:Gibco,Cat No.:10099-141CFetal bovine serum: Gibco, Cat No.: 10099-141C
重组人胰岛素:上海翊圣生物科技有限公司,Cat No.:40112ES60Recombinant human insulin: Shanghai Yisheng Biotechnology Co., Ltd., Cat No.: 40112ES60
青链霉素(P/S):Gibco,Cat No.:15140-122Penicillin (P/S): Gibco, Cat No.: 15140-122
0.25%胰酶-EDTA:Gibco,Cat No.:25200-0720.25% Trypsin-EDTA: Gibco, Cat No.: 25200-072
DPBS:Hyclone,Cat.No.:SH30256.01DPBS: Hyclone, Cat. No.: SH30256.01
氟维司群注射液(Fulvestrant):阿斯利康(AstraZeneca),250mg/5mL/支Fulvestrant injection (Fulvestrant): AstraZeneca, 250mg/5mL/bottle
实验方法experimental method
动物信息:NPG小鼠,雌性,6-7周,体重约17-23克,动物购自北京维通达生物技术有限公司,将小鼠饲养在SPF级的环境中,每个笼位单独送排风,所有动物都可以自由获取标准认证的商业实验室饮食和自由饮水。Animal information: NPG mice, female, 6-7 weeks old, weighing about 17-23 grams, were purchased from Beijing Weitongda Biotechnology Co., Ltd., and the mice were raised in an SPF-grade environment, and each cage was sent separately All animals had free access to a standard certified commercial laboratory diet and water ad libitum.
细胞培养:人乳腺癌MCF-7细胞株体外培养,培养条件为EMEM(细胞培养液)中加入10%胎牛血清,10μg/ml重组人胰岛素和1%P/S,37℃,5%CO 2孵箱。一周一到两次用0.25%胰酶-EDTA进行常规消化处理传代。数量到达要求以及细胞在对数生长期时,收取细胞,计数。 Cell culture: In vitro culture of human breast cancer MCF-7 cell line, the culture conditions are 10% fetal bovine serum, 10 μg/ml recombinant human insulin and 1% P/S in EMEM (cell culture medium), 37 ° C, 5% CO 2 incubators. Passage with routine digestion with 0.25% trypsin-EDTA once or twice a week. When the number reaches the requirement and the cells are in the logarithmic growth phase, the cells are collected and counted.
细胞接种:将0.015ml/(含2×10 6个)MCF-7细胞悬液脑原位接种于每只小鼠的大脑(前囟前1mm,中缝右侧2mm,颅骨硬脑膜平面下3mm),并于细胞接种前三天皮下接种17β-雌二醇片。在接种细胞后第8天,开始分组给药,依据动物体重随机分组给药,分组当天为Day 0。 Cell inoculation: 0.015ml/(containing 2×10 6 ) MCF-7 cell suspension was inoculated in the brain of each mouse (1mm in front of bregma, 2mm on the right side of the raphe, 3mm below the dura mater plane of the skull) , and 17β-estradiol tablets were subcutaneously inoculated three days before cell inoculation. On the 8th day after cell inoculation, group administration began, and animals were randomly divided into groups according to body weight, and the day of grouping was Day 0.
给药:式(I)化合物琥珀酸盐(实施例10-1,A晶型)的给药剂量为3,10或30mg/kg,口服给药(PO),每天一次给药(QD),持续给药到Day 60。氟维司群(Fulvestrant)的给药剂量为250mg/kg,皮下注射(SC),每周一次给药(QW),持续给药到Day 60。每组8只小鼠。Administration: the dosage of the compound succinate of formula (I) (Example 10-1, crystal form A) is 3, 10 or 30 mg/kg, administered orally (PO), administered once a day (QD), Dosing continued until Day 60. The dosage of fulvestrant (Fulvestrant) was 250mg/kg, subcutaneously injected (SC), once a week (QW), and continued until Day 60. 8 mice per group.
体重测量和实验指标:Body weight measurements and experimental indicators:
每周两次称量小鼠体重。Mice were weighed twice a week.
化合物的抑瘤疗效用中位生存期来评价。The antitumor efficacy of compounds was evaluated by the median survival time.
实验结果:Experimental results:
见表14和图17。与溶剂对照组相比,一周一次皮下注射给予MCF-7脑原位肿瘤模型动物250mg/kg Fulvestrant对动物的生存率没有显著提高,而一天一次口服给予MCF-7脑原位肿瘤模型动物10或30mg/kg式(I)化合物琥珀酸盐可以显著提高小鼠生存率(P<0.001),改善动物状态,药效明显强于Fulvestrant(P<0.001)。See Table 14 and Figure 17. Compared with the solvent control group, giving MCF-7 brain orthotopic tumor model animals 250 mg/kg Fulvestrant subcutaneously once a week did not significantly improve the survival rate of the animals, while oral administration of MCF-7 brain orthotopic tumor model animals once a day for 10 or 30mg/kg formula (I) compound succinate can significantly increase the survival rate of mice (P<0.001), improve the state of animals, and the drug effect is significantly stronger than that of Fulvestrant (P<0.001).
表14 MCF-7脑原位模型各治疗组对小鼠生存率的影响Table 14 Effect of MCF-7 brain orthotopic model of each treatment group on the survival rate of mice
Figure PCTCN2022131313-appb-000035
Figure PCTCN2022131313-appb-000035
测试例10:式(I)化合物对MCF7细胞内雌激素受体降解效果研究试验Test Example 10: Research on the effect of compounds of formula (I) on the degradation of estrogen receptors in MCF7 cells
1.实验目的1. Purpose of the experiment
本实验的目的是测定式(I)化合物对MCF7细胞内内源表达的雌激素受体的降解活性,根据DC 50及最大降解效率评价化合物的活性。 The purpose of this experiment is to measure the degradation activity of the compound of formula (I) on the endogenously expressed estrogen receptor in MCF7 cells, and evaluate the activity of the compound according to DC 50 and maximum degradation efficiency.
2.实验方法2. Experimental method
MCF7细胞(ATCC,HTB-22)用含10%胎牛血清的DMEM(Gibco,11995-065)完全培养基进行培养。实验第一天,使用完全培养基将MCF7细胞以3000个/孔的密度种于384孔板,37℃,5%CO 2细胞培养箱培养。待测化合物溶解于DMSO,储存浓度为10mM,用Echo 550(Labcyte Inc.)稀释并加入细胞培养板内,待测化合物处理的起始浓度为100nM,3倍梯度稀释,9个浓度点,设置含0.5%DMSO的空白对照,各浓度点设双复孔对照。37℃,5%CO 2细胞培养箱培养24小时。各细胞培养孔内加入多聚甲醛至细胞培养液内,多聚甲醛终浓度约3.7%以固定细胞,作用30分钟后,弃上清,加入50μL PBS每孔洗涤一次;加入PBS含0.5%v/v Tween-20处理细胞30分钟,PBS洗涤一次;加入封闭液(PBS内含5%BSA,0.05%Tween-20)室温孵育1小时;去封闭液加入一抗混合液(抗-ER单抗,Estrogen Receptorα(D8H8)Rabbit mAb,GST,#8644S,1:1000稀释;抗-GAPDH单抗,GAPDH(D4C6R)Mouse mAb,GST,#97166S,1:2000稀释)室温孵育3小时;用PBST(PBS内含0.05%Tween-20)洗涤3次;加入检测二抗(800CW-羊抗兔IgG,LI-COR,P/N:926-32211,1:1000稀释;680RD-羊抗鼠IgG,LI-COR,#925-68070,1:1000稀释),室温,避光孵育45分钟;PBST洗涤3次,使用Odyssey CLx读取各孔荧光信号,计算Chanel 800(ER)/Chanel 680(GAPDH)数值。以0.1μM fulvestrant处理孔作为100%降解对照,计算各浓度点的降解率,使用XlLfit分析处理数据,计算化合物的降解活性DC 50及最大降解率Imax。数据分析见表15。 MCF7 cells (ATCC, HTB-22) were cultured with DMEM (Gibco, 11995-065) complete medium containing 10% fetal bovine serum. On the first day of the experiment, MCF7 cells were seeded in a 384-well plate at a density of 3000 cells/well using complete medium, and cultured in a 5% CO 2 cell incubator at 37°C. The compound to be tested was dissolved in DMSO with a storage concentration of 10mM, diluted with Echo 550 (Labcyte Inc.) and added to the cell culture plate, the initial concentration of the compound to be tested was 100nM, 3-fold serial dilution, 9 concentration points, set A blank control containing 0.5% DMSO was used, and a double-well control was set at each concentration point. Incubate in a 37°C, 5% CO 2 cell incubator for 24 hours. Add paraformaldehyde to the cell culture medium in each cell culture well. The final concentration of paraformaldehyde is about 3.7% to fix the cells. After 30 minutes of action, discard the supernatant and add 50 μL PBS to each well to wash once; add PBS containing 0.5% v /v Tween-20 to treat cells for 30 minutes, wash once with PBS; add blocking solution (5% BSA, 0.05% Tween-20 in PBS) and incubate at room temperature for 1 hour; remove blocking solution and add primary antibody mixture (anti-ER monoclonal antibody , Estrogen Receptorα (D8H8) Rabbit mAb, GST, #8644S, 1:1000 dilution; anti-GAPDH monoclonal antibody, GAPDH (D4C6R) Mouse mAb, GST, #97166S, 1:2000 dilution) at room temperature for 3 hours; with PBST ( PBS containing 0.05% Tween-20) and washed 3 times; add detection secondary antibody (800CW-goat anti-rabbit IgG, LI-COR, P/N:926-32211, 1:1000 dilution; 680RD-goat anti-mouse IgG, LI -COR, #925-68070, 1:1000 dilution), room temperature, incubate in the dark for 45 minutes; wash 3 times with PBST, use Odyssey CLx to read the fluorescence signal of each well, and calculate the value of Chanel 800(ER)/Chanel 680(GAPDH) . The wells treated with 0.1 μM fulvestrant were used as the 100% degradation control, and the degradation rate at each concentration point was calculated. XlLfit was used to analyze the processing data, and the degradation activity DC50 and the maximum degradation rate Imax of the compound were calculated. See Table 15 for data analysis.
表15式(I)化合物对MCF7细胞内雌激素受体降解活性Table 15 formula (I) compound is to MCF7 intracellular estrogen receptor degradation activity
化合物编号Compound number ER level DC 50(nM) ER level DC 50 (nM) 最大降解率maximum degradation rate
式(I)化合物Compound of formula (I) 0.150.15 104%104%
测试例11:式(I)化合物对MCF7细胞增殖的抑制效果研究试验Test Example 11: Study on the inhibitory effect of the compound of formula (I) on the proliferation of MCF7 cells
1.实验目的1. Purpose of the experiment
本实验的目的是测定式(I)化合物对MCF7细胞体外增殖的抑制影响,根据IC 50评价化合物的活性。 The purpose of this experiment is to determine the inhibitory effect of the compound of formula (I) on the proliferation of MCF7 cells in vitro, and to evaluate the activity of the compound according to IC 50 .
2.实验方法2. Experimental method
MCF7细胞(ATCC,HTB-22)用含10%胎牛血清的DMEM(Gibco,11995-065)完全培养基进行培养。实验第一天,使用完全培养基将MCF7细胞以500个/孔的密度种于384孔板, 37℃,5%CO 2细胞培养箱过夜培养。第二天,加入待测化合物进行药物处理,采用Echo550(Labcyte Inc.)将储存浓度为10mM的化合物溶液进行稀释及转移至各细胞培养孔内,待测化合物在细胞内的处理起始浓度为100nM,3倍梯度稀释,9个浓度点,设置含0.3%DMSO的空白对照,各浓度点设双复孔对照。37℃,5%CO 2细胞培养箱培养7天,第八天,取出细胞培养板。加入
Figure PCTCN2022131313-appb-000036
Luminescent Cell Viability Assay(Promega,G7573),室温放置10分钟后,使用多标记酶标仪EnVision(PerkinElmer)读取发光信号值,用XLfit根据化合物的浓度和发光信号值计算各化合物的抑制活性IC 50
MCF7 cells (ATCC, HTB-22) were cultured with DMEM (Gibco, 11995-065) complete medium containing 10% fetal bovine serum. On the first day of the experiment, MCF7 cells were seeded in a 384-well plate at a density of 500 cells/well using complete medium, and cultured overnight at 37° C. in a 5% CO 2 cell incubator. The next day, add the compound to be tested for drug treatment, and use Echo550 (Labcyte Inc.) to dilute the compound solution with a storage concentration of 10 mM and transfer it to each cell culture well. The initial concentration of the compound to be tested in the cells is 100nM, 3-fold serial dilution, 9 concentration points, a blank control containing 0.3% DMSO was set, and double-well controls were set at each concentration point. Culture in a 37°C, 5% CO 2 cell incubator for 7 days, and take out the cell culture plate on the eighth day. join in
Figure PCTCN2022131313-appb-000036
Luminescent Cell Viability Assay (Promega, G7573), after standing at room temperature for 10 minutes, read the luminescence signal value using a multi-labeled microplate reader EnVision (PerkinElmer), and use XLfit to calculate the inhibitory activity IC 50 of each compound according to the concentration and luminescence signal value of the compound .
3.数据分析见表163. Data analysis see Table 16
表16式(I)化合物对MCF7细胞增殖的抑制活性The inhibitory activity of table 16 formula (I) compound to MCF7 cell proliferation
化合物编号Compound number MCF-7细胞增殖抑制IC 50(nM) MCF-7 cell proliferation inhibition IC 50 (nM)
式(I)化合物Compound of formula (I) 0.270.27
测试例12:式(I)化合物在pH值为7.4的磷酸盐缓冲液中的表观溶解度Test Example 12: Apparent Solubility of Compound of Formula (I) in Phosphate Buffered Saline at pH 7.4
为了使口服化合物到达作用部位,并且为了肠道的有效吸收,该化合物需处于溶液状态,因此具有高固有溶解度的化合物可能更适合于药物用途。In order for an orally administered compound to reach the site of action, and for efficient intestinal absorption, the compound needs to be in solution, so compounds with high intrinsic solubility may be more suitable for pharmaceutical use.
一、材料和试剂1. Materials and Reagents
待测化合物按记载方法制备。对照药孕酮从Sigma购买。pH值为7.4的磷酸盐缓冲液由本实验室配制。乙腈和甲醇从Fisher购买。其他试剂从市场购买。The compounds to be tested were prepared according to the methods described. The control drug progesterone was purchased from Sigma. Phosphate buffer with a pH value of 7.4 was prepared by our laboratory. Acetonitrile and methanol were purchased from Fisher. Other reagents were purchased from the market.
1.5毫升平底玻璃小瓶(BioTech Solutions);聚四氟乙烯/硅有机树脂瓶塞(BioTech Solutions);聚四氟乙烯包被搅拌棒;MultiScreenHTS HV(0.45μm)96 well plate过滤板(Millipore,MSHVN4510 or MSHVN4550);Eppendorf Thermomixer Comfort;Vacuum Manifold ORVMN96(Orochem)。1.5 ml flat-bottomed glass vial (BioTech Solutions); PTFE/silicone stopper (BioTech Solutions); PTFE-coated stirring rod; MultiScreenHTS HV (0.45 μm) 96 well plate filter plate (Millipore, MSHVN4510 or MSHVN4550); Eppendorf Thermomixer Comfort; Vacuum Manifold ORVMN96 (Orochem).
二、实验步骤2. Experimental steps
储备液的配制Preparation of stock solution
用DMSO配制待测物和对照药孕酮的10mM储备液。10mM stock solutions of the test substance and the control drug progesterone were prepared in DMSO.
表观溶解度测定步骤Apparent Solubility Determination Procedure
取30μL 10mM待测物储备液,以指定顺序加到对应96孔板的对应位置。在样品板的对应小瓶加入970μL的pH值7.4的磷酸盐缓冲液。实验为双平行。在每个小瓶中加一根搅拌棒,并盖上聚四氟乙烯/硅有机树脂瓶塞。随后将样品盘放进Eppendorf Thermomixer Comfort,以1100转的转速在25度条件下震荡2个小时。2小时后,去除瓶塞,用一块大磁铁吸走搅拌棒,然后从样品板转移样品至过滤板。用真空泵产生负压,过滤样品。转移5μL滤液到新的样品板,然后加入5μL DMSO和490μL 50%ACN(IS).H 2O(内标乙腈:水=1:1)。根据峰形情况,可能用一定比例的50%ACN(IS).H 2O来稀释样品稀释液以获得更好的峰形。稀释倍数可能因待测物溶解性的大小或其液质响应信号强弱而调整。 Take 30 μL of 10mM stock solution of the analyte and add it to the corresponding position of the corresponding 96-well plate in the specified order. Add 970 μL of pH 7.4 phosphate buffer to the corresponding vial of the sample plate. The experiments were performed in double parallel. Add a stir bar to each vial and cap with a Teflon/silicone stopper. Then put the sample tray into the Eppendorf Thermomixer Comfort, and vibrate for 2 hours at 1100 rpm at 25°C. After 2 hours, remove the stopper, remove the stir bar with a large magnet, and transfer the sample from the sample plate to the filter plate. Use a vacuum pump to generate negative pressure and filter the sample. Transfer 5 μL of filtrate to a new sample plate, then add 5 μL DMSO and 490 μL 50% ACN(IS).H 2 O (internal standard acetonitrile:water=1:1). Depending on the peak shape, it may be possible to dilute the sample diluent with a certain percentage of 50% ACN(IS).H 2 O to obtain better peak shape. The dilution factor may be adjusted due to the solubility of the analyte or the strength of its liquid-mass response signal.
样品分析步骤Sample Analysis Procedure
将进样板放进自动进样器的进样盘中,通过液质分析评估样品。Place the sampling plate into the sampling tray of the autosampler and evaluate the sample by LC/MS.
三、实验步骤3. Experimental steps
通过Microsoft Excel进行所有的计算。样品滤液的分析和定量,是通过使用液质对已知浓度的标准品峰的定性和定量完成的。计算得到的式(I)化合物在PH值为7.4的磷酸盐缓冲液中的表观溶解度值。All calculations were performed by Microsoft Excel. The analysis and quantification of the sample filtrate is accomplished by using LC/MS to identify and quantify the peaks of standard products of known concentration. The calculated apparent solubility value of the compound of formula (I) in phosphate buffer solution with a pH value of 7.4.
表17式(I)化合物在pH值为7.4的磷酸盐缓冲液中的表观溶解度值The apparent solubility value of the compound of table 17 formula (I) in the phosphate buffer saline buffered saline that pH value is 7.4
化合物编号Compound number pH=7.4表观溶解度(μM)pH=7.4 apparent solubility (μM)
式(I)化合物Compound of formula (I) 9292
测试例13:式(I)化合物在大鼠中血脑屏障(BBB)透过能力Test Example 13: The blood-brain barrier (BBB) penetration ability of the compound of formula (I) in rats
药物能透过动物的血脑屏障在脑中有足够的暴露量是药物对脑转移病灶有效的关键,因此通过测量动物给药后血浆和脑组织中的药物浓度,可以评估药物在脑中的分布情况,进而判断药物能否在脑原位模型中起到抑制肿瘤生长的效果。Drugs can pass through the blood-brain barrier of animals and have sufficient exposure in the brain is the key to the effectiveness of drugs on brain metastases. Therefore, by measuring drug concentrations in plasma and brain tissue after administration to animals, the effect of drugs in the brain Distribution, and then judge whether the drug can inhibit tumor growth in the brain orthotopic model.
实验材料Experimental Materials
SD雌性大鼠购自北京维通利华实验动物技术有限公司。MC(甲基纤维素)购自Sigma;乙腈购自Merck(USA)。PBS(磷酸盐缓冲盐)购自生工生物。SD female rats were purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd. MC (methylcellulose) was purchased from Sigma; acetonitrile was purchased from Merck (USA). PBS (phosphate buffered saline) was purchased from Sangon Biotech.
实验方法experimental method
雌性SD大鼠6只(200-300g,6-8周),随机分成2组,每组3只。分别给予式(I)化合物,溶媒0.5%甲基纤维素水溶液。动物实验前正常喂水,禁食过夜,给药后四小时恢复给食。每组大鼠于给药后2h收集血浆和脑组织。收集的全血样品置于K 2EDTA抗凝管中,离心5min后(12,000rpm,4℃)取血浆待测;组织采集后用滤纸吸干,样品保存在-80度冰箱待测。 Six female SD rats (200-300g, 6-8 weeks) were randomly divided into 2 groups with 3 rats in each group. The compound of formula (I) and the vehicle of 0.5% methylcellulose aqueous solution were administered respectively. Animals were fed water normally before the experiment, fasted overnight, and resumed feeding four hours after administration. Plasma and brain tissue were collected from rats in each group 2 hours after administration. The collected whole blood samples were placed in K 2 EDTA anticoagulant tubes, centrifuged for 5 minutes (12,000rpm, 4°C) to collect plasma for testing; tissues were collected and blotted dry with filter paper, and the samples were stored in a -80°C refrigerator for testing.
取大鼠血浆样品10μL,加入150μL乙腈溶剂(其中含内标化合物)沉淀蛋白,涡旋5min后,离心(14,000rpm)5min,上清液用含0.1%(v/v)FA的水稀释2倍,于LC-MS/MS系统(AB Sciex Triple Quad 6500+)进行定量检测。对10x稀释样品,取2μL样品加入18μL的空白血浆,涡旋0.5min后,加入300μL乙腈溶剂(其中含内标化合物)沉淀蛋白,其余处理步骤同不稀释样品。在测定血浆样品浓度时随行SD大鼠血浆标准曲线和血浆质控样品。Take 10 μL of rat plasma sample, add 150 μL of acetonitrile solvent (including internal standard compound) to precipitate protein, vortex for 5 min, centrifuge (14,000 rpm) for 5 min, and dilute the supernatant with water containing 0.1% (v/v) FA for 2 times, quantitative detection was performed on LC-MS/MS system (AB Sciex Triple Quad 6500+). For 10x diluted samples, take 2 μL sample and add 18 μL blank plasma, vortex for 0.5 min, add 300 μL acetonitrile solvent (including internal standard compound) to precipitate protein, and the rest of the processing steps are the same as for undiluted samples. When determining the concentration of plasma samples, SD rat plasma standard curve and plasma quality control samples were accompanied.
大鼠脑组织样品先用4倍质量体积的PBS匀浆液进行匀浆。取脑组织匀浆液样品20μL,加入20μL空白小鼠血浆进行稀释混匀再加入600μL乙腈溶剂(其中含内标化合物)沉淀蛋白,涡旋5min后,离心(14,000rpm)5min,上清液用含0.1%(v/v)FA的水稀释2倍,于LC-MS/MS系统(AB Sciex Triple Quad 6500+)进行定量检测。式(I)化合物表现出优秀的血脑屏障透过能力,在大鼠的脑组织中药物暴露量较高。Rat brain tissue samples were first homogenized with 4 times the mass volume of PBS homogenate. Take 20 μL of brain tissue homogenate sample, add 20 μL of blank mouse plasma to dilute and mix, then add 600 μL of acetonitrile solvent (including internal standard compound) to precipitate protein, vortex for 5 min, centrifuge (14,000 rpm) for 5 min, supernatant with Diluted 2 times with 0.1% (v/v) FA in water, and carried out quantitative detection on LC-MS/MS system (AB Sciex Triple Quad 6500+). The compound of formula (I) exhibits excellent blood-brain barrier penetration ability, and the drug exposure in rat brain tissue is relatively high.
BBB测试结果如下所示:The BBB test results are as follows:
表18式(I)化合物大鼠血脑屏障透过实验Table 18 formula (I) compound rat blood-brain barrier penetration test
Figure PCTCN2022131313-appb-000037
Figure PCTCN2022131313-appb-000037
测试例14:式(I)化合物对MCF-7小鼠皮下肿瘤模型的生长抑制实验Test Example 14: Growth inhibition experiment of the compound of formula (I) on MCF-7 mouse subcutaneous tumor model
实验试剂experimental reagent
人乳腺癌MCF-7细胞:ATCC,HTB-22Human breast cancer MCF-7 cells: ATCC, HTB-22
17β-雌二醇片:Innovative Research of America,Cat No.:SE-121,60-day release,0.72mg/pellet17β-estradiol tablets: Innovative Research of America, Cat No.: SE-121, 60-day release, 0.72mg/pellet
EMEM培养液:ATCC,Cat No.:30-2003EMEM medium: ATCC, Cat No.: 30-2003
胎牛血清:Gbico;Cat No.:1099-141CFetal bovine serum: Gbico; Cat No.:1099-141C
青链霉素(Pen Strep):Gibco,Cat No.:15240-122Pen Strep (Pen Strep): Gibco, Cat No.: 15240-122
重组人胰岛素:上海翊圣,Cat.No.:40112ES60Recombinant human insulin: Shanghai Yisheng, Cat.No.: 40112ES60
0.25%胰酶-EDTA:Gibco,Cat No.:25200-0720.25% Trypsin-EDTA: Gibco, Cat No.: 25200-072
D-PBS(无钙镁离子磷酸盐缓冲液):Hyclone,Cat.No.:SH30256.01D-PBS (phosphate buffered saline without calcium and magnesium ions): Hyclone, Cat.No.: SH30256.01
Matrigel:Corning,Cat.No.:356237Matrigel: Corning, Cat. No.: 356237
实验方法experimental method
动物信息:NPG小鼠,雌性,6-7周,体重约19-28克,动物购自北京维通达生物技术有限公司,将小鼠饲养在SPF级的环境中,每个笼位单独送排风,所有动物都可以自由获取标准认证的商业实验室饮食和自由饮水。Animal information: NPG mice, female, 6-7 weeks old, weighing about 19-28 grams, were purchased from Beijing Weitongda Biotechnology Co., Ltd., and the mice were raised in an SPF-grade environment, and each cage was sent separately All animals had free access to a standard certified commercial laboratory diet and water ad libitum.
细胞培养:人乳腺癌MCF-7细胞株体外培养,培养条件为EMEM(细胞培养液)中加入10%胎牛血清,1%Pen Strep,10μg/ml重组人胰岛素,37℃、5%CO 2孵箱。一周一次用0.25%胰酶-EDTA消化液进行常规消化处理传代。当细胞饱和度为80%-90%,数量达到要求时,收取细胞,计数。 Cell culture: In vitro culture of human breast cancer MCF-7 cell line, the culture conditions are 10% fetal bovine serum, 1% Pen Strep, 10 μg/ml recombinant human insulin in EMEM (cell culture medium), 37°C, 5% CO 2 incubator. Routine digestion with 0.25% trypsin-EDTA digestion solution once a week for passage. When the cell saturation is 80%-90% and the number reaches the requirement, collect the cells and count them.
细胞接种:将0.1ml/(含1×10 7)MCF-7细胞悬液(D-PBS:Matrigel,体积比为1:1)皮下接种于每只小鼠的右后背,并于细胞接种前四天皮下接种17β-雌二醇片。在接种细胞后第24天,依据肿瘤体积随机分组给药,分组当天为第0天(Day 0)。 Cell inoculation: Subcutaneously inoculate 0.1ml/(containing 1×10 7 ) MCF-7 cell suspension (D-PBS: Matrigel, volume ratio 1:1) on the right back of each mouse, and inoculate In the first four days, 17β-estradiol tablets were subcutaneously inoculated. On the 24th day after cell inoculation, the drugs were randomly divided into groups according to the tumor volume, and the grouping day was Day 0 (Day 0).
给药:式(I)化合物的给药剂量为1,3或10mg/kg,口服给药(PO),每天一次给药(QD)x3周。溶媒组8只小鼠,给药组6只小鼠。Administration: The dosage of the compound of formula (I) is 1, 3 or 10 mg/kg, administered orally (PO), administered once a day (QD) x 3 weeks. There were 8 mice in the vehicle group and 6 mice in the administration group.
肿瘤测量和实验指标:Tumor measurements and experimental indicators:
每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a x b 2,a和b分别表示肿瘤的长径和短径。每周两次测量小鼠体重。 Tumor diameters were measured twice a week with vernier calipers. The formula for calculating the tumor volume is: V=0.5a x b 2 , where a and b represent the long diameter and short diameter of the tumor, respectively. Mouse body weights were measured twice a week.
化合物的抑瘤疗效用肿瘤生长抑制率TGI(%)来评价。TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]x100%。The antitumor efficacy of compounds was evaluated by tumor growth inhibition rate TGI (%). TGI (%)=[(1-(average tumor volume at the end of administration of a certain treatment group-average tumor volume at the beginning of administration of this treatment group)/(average tumor volume at the end of treatment of the solvent control group-at the beginning of treatment of the solvent control group Mean tumor volume)] x 100%.
实验结果:Experimental results:
见表19。在小鼠皮下移植瘤MCF-7模型中,式(I)化合物在1mg/kg,3mg/kg,或10mg/kg一天一次口服给药对肿瘤生长具有显著抑制作用(P<0.01),且具有较好的剂量反应关系,在3mg/kg和10mg/kg剂量下具有缩小肿瘤的效果。式(I)化合物在10mg/kg一天一次口服给药对肿瘤生长具有显著抑制作用(P<0.01),且具有缩小肿瘤的效果。式(I)化合物在所尝试剂量下未显著影响小鼠体重。See Table 19. In the subcutaneous transplanted tumor MCF-7 model in mice, the compound of formula (I) has a significant inhibitory effect on tumor growth (P<0.01) at 1 mg/kg, 3 mg/kg, or 10 mg/kg orally administered once a day, and has It has a good dose-response relationship, and it has the effect of shrinking tumors at the doses of 3mg/kg and 10mg/kg. Oral administration of the compound of formula (I) once a day at 10 mg/kg has a significant inhibitory effect on tumor growth (P<0.01), and has the effect of shrinking tumors. The compound of formula (I) did not significantly affect the body weight of mice at the doses tried.
表19 MCF-7皮下瘤模型肿瘤体积Table 19 MCF-7 subcutaneous tumor model tumor volume
Figure PCTCN2022131313-appb-000038
Figure PCTCN2022131313-appb-000038
测试例15:式(I)化合物对小鼠MCF-7脑原位肿瘤模型生长的抑制实验Test Example 15: Inhibitory experiment of the compound of formula (I) on the growth of mouse MCF-7 brain orthotopic tumor model
实验试剂/仪器:Experimental reagents/instruments:
人乳腺癌MCF-7细胞:ATCC,HTB-22Human breast cancer MCF-7 cells: ATCC, HTB-22
17β-雌二醇片:Innovative Research of America,Cat No.:SE-121,60-day release,0.72mg/pellet17β-estradiol tablets: Innovative Research of America, Cat No.: SE-121, 60-day release, 0.72mg/pellet
EMEM培养液:ATCC,Cat No.:30-2003EMEM medium: ATCC, Cat No.: 30-2003
胎牛血清:Gibco,Cat.No.:1099-141CFetal bovine serum: Gibco, Cat.No.:1099-141C
青链霉素(Pen Strep):Gibco,Cat No.:15240-122Pen Strep (Pen Strep): Gibco, Cat No.: 15240-122
重组人胰岛素:上海翊圣,Cat.No.:40112ES60Recombinant human insulin: Shanghai Yisheng, Cat.No.: 40112ES60
0.25%胰酶-EDTA:Gibco,Cat No.:25200-0720.25% Trypsin-EDTA: Gibco, Cat No.: 25200-072
脑立体定位仪:瑞沃德,Cat No.:标准型/数显/单臂/小鼠/68055Brain Stereotaxic Instrument: Reward, Cat No.: Standard/Digital Display/Single Arm/Mouse/68055
微量注射泵:KDS,Cat No.:Legato130Micro injection pump: KDS, Cat No.: Legato130
微型手持颅钻:瑞沃德,Cat No.:78001Miniature Handheld Skull Drill: Reward, Cat No.:78001
实验方法:experimental method:
动物信息:NPG小鼠,雌性,6-8周,体重约17-29克,动物购自北京维通达生物技术有限公司,将小鼠饲养在SPF级的环境中,每个笼位单独送排风,所有动物都可以自由获取标准认证的商业实验室饮食和自由饮水。Animal information: NPG mice, female, 6-8 weeks old, weighing about 17-29 grams, were purchased from Beijing Weitongda Biotechnology Co., Ltd., and the mice were raised in an SPF-grade environment, and each cage was sent separately All animals had free access to a standard certified commercial laboratory diet and water ad libitum.
细胞培养:人乳腺癌MCF-7细胞株体外培养,培养条件为EMEM(细胞培养液)中加入10%胎牛血清,1%Pen Strep,10μg/ml重组人胰岛素,37℃、5%CO 2孵箱。一周两次用0.25%胰酶-EDTA消化液进行常规消化处理传代。当细胞饱和度为80%-90%,数量达到要求时,收取细胞,计数。 Cell culture: In vitro culture of human breast cancer MCF-7 cell line, the culture conditions are 10% fetal bovine serum, 1% Pen Strep, 10 μg/ml recombinant human insulin in EMEM (cell culture medium), 37°C, 5% CO 2 incubator. Routine digestion with 0.25% trypsin-EDTA digestion solution was performed twice a week for subculture. When the cell saturation is 80%-90% and the number reaches the requirement, collect the cells and count them.
细胞接种:将15μl/(含2×10 6)MCF-7细胞悬液利用脑定位仪,微量注射泵和微型手持颅钻,接种于小鼠颅内,并于细胞接种前三天皮下接种17β-雌二醇片。在接种细胞后第8天,依据小鼠体重随机分组给药,分组当天为第0天(Day 0)。 Cell inoculation: 15μl/(containing 2×10 6 ) MCF-7 cell suspension was inoculated into the mouse skull using a brain localizer, a micro-injection pump and a miniature hand-held cranial drill, and 17β was subcutaneously inoculated three days before cell inoculation - Estradiol tablets. On the 8th day after cell inoculation, mice were randomly divided into groups according to body weight for administration, and the grouping day was Day 0 (Day 0).
给药:Fulvestrant(氟维司群,阿斯利康)给药剂量为250mg/kg,皮下注射(SC),每周一次给药(QW),式(I)化合物的给药剂量为30mg/kg,口服给药(PO),每天一次给药(QD)。溶媒组11只小鼠,给药组8只小鼠。所有组持续给药直到小鼠死亡,因状态差安乐死或者实验结束。Administration: Fulvestrant (Fulvestrant, AstraZeneca) dosage is 250mg/kg, subcutaneous injection (SC), administration (QW) once a week, the dosage of formula (I) compound is 30mg/kg , administered orally (PO), administered once a day (QD). There were 11 mice in the vehicle group and 8 mice in the administration group. All groups continued administration until the mice died, were euthanized due to poor condition or the experiment ended.
实验观察和结束:Experimental observation and end:
每周两次测量小鼠体重,并观察小鼠生存状态。The body weight of the mice was measured twice a week, and the survival status of the mice was observed.
第48天(Day 48)结束实验,安乐死所有小鼠。On the 48th day (Day 48), the experiment was terminated, and all mice were euthanized.
实验结果:Experimental results:
见图18和图19。在小鼠脑原位MCF-7模型中,Fulvestrant组小鼠(250mg/kg一周一次皮下注射给药)体重持续下降,小鼠的生存状况和溶剂对照组无明显差别(中位生存期,溶媒对照组29天,Fulvestrant组29.5天)。式(I)化合物组小鼠(30mg/kg一天一次口服给药)体重平稳,状态无异常,直到实验终点,式(I)化合物组小鼠均未出现死亡,相对于溶剂对照或者Fulvestrant,化合物对MCF-7脑原位肿瘤模型小鼠具有显著的抑制作用,小鼠的生存期显著的延长(P<0.01)。See Figure 18 and Figure 19. In the orthotopic MCF-7 model of the mouse brain, the body weight of the mice in the Fulvestrant group (250mg/kg administered subcutaneously once a week) continued to decrease, and the survival status of the mice was not significantly different from that of the solvent control group (median survival period, vehicle 29 days in the control group and 29.5 days in the Fulvestrant group). The mice in the compound group of formula (I) (30 mg/kg administered orally once a day) had a stable body weight and no abnormal state. Until the end of the experiment, the mice in the compound group of formula (I) did not die. Compared with the solvent control or Fulvestrant, the compound It has a significant inhibitory effect on MCF-7 brain orthotopic tumor model mice, and the survival period of mice is significantly prolonged (P<0.01).

Claims (26)

  1. 式(I)化合物的可药用盐,A pharmaceutically acceptable salt of a compound of formula (I),
    Figure PCTCN2022131313-appb-100001
    Figure PCTCN2022131313-appb-100001
    其中,所述可药用盐选自硫酸盐、磷酸盐、苯甲酸盐、琥珀酸盐、己二酸盐、富马酸盐、L-苹果酸盐和柠檬酸盐。Wherein, the pharmaceutically acceptable salt is selected from sulfate, phosphate, benzoate, succinate, adipate, fumarate, L-malate and citrate.
  2. 根据权利要求1所述的式(I)化合物的可药用盐,所述式(I)化合物的可药用盐选自其苯甲酸盐、琥珀酸盐和己二酸盐。The pharmaceutically acceptable salt of the compound of formula (I) according to claim 1, which is selected from the group consisting of benzoate, succinate and adipate.
  3. 根据权利要求1所述的式(I)化合物的可药用盐,所述式(I)化合物的琥珀酸盐如式(II)所示,其中,x选自0.5~2,The pharmaceutically acceptable salt of the compound of formula (I) according to claim 1, the succinate of the compound of formula (I) is shown in formula (II), wherein, x is selected from 0.5~2,
    Figure PCTCN2022131313-appb-100002
    Figure PCTCN2022131313-appb-100002
  4. 根据权利要求1所述的式(I)化合物的可药用盐,所述式(I)化合物的苯甲酸盐如式(III)所示,其中,y选自0.5~2,According to the pharmaceutically acceptable salt of the compound of formula (I) according to claim 1, the benzoate salt of the compound of formula (I) is shown in formula (III), wherein, y is selected from 0.5~2,
    Figure PCTCN2022131313-appb-100003
    Figure PCTCN2022131313-appb-100003
  5. 根据权利要求1所述的式(I)化合物的可药用盐,所述式(I)化合物的己二酸盐如式(IV)所示,其中,z选自0.5~2,According to the pharmaceutically acceptable salt of the compound of formula (I) according to claim 1, the adipate salt of the compound of formula (I) is shown in formula (IV), wherein, z is selected from 0.5~2,
    Figure PCTCN2022131313-appb-100004
    Figure PCTCN2022131313-appb-100004
  6. 根据权利要求3所述的式(I)化合物的可药用盐,其中x选自0.5、0.6、0.7、0.8、0.9、1.0、1.1、1.2、1.3、1.4、1.5、1.6、1.7、1.8、1.9和2.0;优选地,x选自0.8、0.9、1.0、1.1和1.2。The pharmaceutically acceptable salt of the compound of formula (I) according to claim 3, wherein x is selected from 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 and 2.0; preferably, x is selected from 0.8, 0.9, 1.0, 1.1 and 1.2.
  7. 根据权利要求4所述的式(I)化合物的可药用盐,其中y选自0.5、0.6、0.7、0.8、0.9、1.0、1.1、1.2、1.3、1.4、1.5、1.6、1.7、1.8、1.9和2.0;优选地,y选自0.8、0.9、1.0、1.1和1.2。The pharmaceutically acceptable salt of the compound of formula (I) according to claim 4, wherein y is selected from 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 and 2.0; preferably, y is selected from 0.8, 0.9, 1.0, 1.1 and 1.2.
  8. 根据权利要求5所述的式(I)化合物的可药用盐,其中z选自0.5、0.6、0.7、0.8、0.9、1.0、1.1、1.2、1.3、1.4、1.5、1.6、1.7、1.8、1.9和2.0;优选地,z选自0.8、0.9、1.0、1.1和1.2。The pharmaceutically acceptable salt of the compound of formula (I) according to claim 5, wherein z is selected from 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 and 2.0; preferably z is selected from 0.8, 0.9, 1.0, 1.1 and 1.2.
  9. 式(I)化合物的己二酸盐C晶型,The adipate salt C crystal form of the compound of formula (I),
    Figure PCTCN2022131313-appb-100005
    Figure PCTCN2022131313-appb-100005
    所述C晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在4.46±0.2°、6.30±0.2°、8.90±0.2°、19.10±0.2°处有衍射峰。In the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the crystal form C, there are diffraction peaks at 4.46±0.2°, 6.30±0.2°, 8.90±0.2°, and 19.10±0.2°.
  10. 根据权利要求9所述C晶型,其中:The C crystal form according to claim 9, wherein:
    所述C晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在4.46±0.2°、6.30±0.2°、7.03±0.2°、8.90±0.2°、16.45±0.2°、17.78±0.2°、19.10±0.2°、21.03±0.2°处有衍射峰;或者In the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the crystal form C, at 4.46±0.2°, 6.30±0.2°, 7.03±0.2°, 8.90±0.2°, 16.45±0.2°, 17.78±0.2°, Diffraction peaks at 19.10±0.2° and 21.03±0.2°; or
    所述C晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在4.46±0.2°、6.30±0.2°、7.03±0.2°、8.90±0.2°、10.65±0.2°、11.95±0.2°、12.55±0.2°、13.34±0.2°、14.20±0.2°、16.02±0.2°、16.45±0.2°、17.78±0.2°、18.33±0.2°、19.10±0.2°、19.77±0.2°、20.12±0.2°、21.03±0.2°、21.62±0.2°、24.30±0.2°、25.97±0.2°、27.39±0.2°和27.98±0.2°处有衍射峰;或者In the X-ray powder diffraction pattern of the crystal form C represented by diffraction angle 2θ, at 4.46±0.2°, 6.30±0.2°, 7.03±0.2°, 8.90±0.2°, 10.65±0.2°, 11.95±0.2°, 12.55±0.2°, 13.34±0.2°, 14.20±0.2°, 16.02±0.2°, 16.45±0.2°, 17.78±0.2°, 18.33±0.2°, 19.10±0.2°, 19.77±0.2°, 20.12±0.2°, Diffraction peaks at 21.03±0.2°, 21.62±0.2°, 24.30±0.2°, 25.97±0.2°, 27.39±0.2° and 27.98±0.2°; or
    所述C晶型的X-射线粉末衍射图谱与图2基本上一致。The X-ray powder diffraction pattern of the C crystal form is basically consistent with that in FIG. 2 .
  11. 根据权利要求9或10所述C晶型,所述C晶型的DSC谱图在149.89±5℃处具有吸热峰。According to the crystal form C according to claim 9 or 10, the DSC spectrum of the crystal form C has an endothermic peak at 149.89±5°C.
  12. 式(I)化合物的苯甲酸盐B晶型,The benzoate B crystal form of the compound of formula (I),
    Figure PCTCN2022131313-appb-100006
    Figure PCTCN2022131313-appb-100006
    所述B晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在7.19±0.2°、10.01±0.2°、11.39±0.2°、18.78±0.2°处有衍射峰。In the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the crystal form B, there are diffraction peaks at 7.19±0.2°, 10.01±0.2°, 11.39±0.2°, and 18.78±0.2°.
  13. 根据权利要求12所述B晶型,其中:The B crystal form according to claim 12, wherein:
    所述B晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在7.19±0.2°、10.01±0.2°、11.39±0.2°、18.78±0.2°、19.01±0.2°、19.66±0.2°、19.91±0.2°、20.09±0.2°处有衍射峰;或者In the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the B crystal form, at 7.19±0.2°, 10.01±0.2°, 11.39±0.2°, 18.78±0.2°, 19.01±0.2°, 19.66±0.2°, Diffraction peaks at 19.91±0.2° and 20.09±0.2°; or
    所述B晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在4.69±0.2°、6.34±0.2°、7.19±0.2°、9.83±0.2°、10.01±0.2°、11.11±0.2°、11.39±0.2°、14.78±0.2°、18.78±0.2°、19.01±0.2°、19.25±0.2°、19.66±0.2°、19.91±0.2°、20.09±0.2°、20.80±0.2°、21.66±0.2°处有衍射峰;或者In the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the B crystal form, at 4.69±0.2°, 6.34±0.2°, 7.19±0.2°, 9.83±0.2°, 10.01±0.2°, 11.11±0.2°, 11.39±0.2°, 14.78±0.2°, 18.78±0.2°, 19.01±0.2°, 19.25±0.2°, 19.66±0.2°, 19.91±0.2°, 20.09±0.2°, 20.80±0.2°, 21.66±0.2° have diffraction peaks; or
    所述B晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在4.69±0.2°、4.91±0.2°、6.34±0.2°、7.19±0.2°、9.37±0.2°、9.83±0.2°、10.01±0.2°、10.32±0.2°、11.11±0.2°、11.39±0.2°、12.06±0.2°、12.69±0.2°、13.52±0.2°、14.07±0.2°、14.36±0.2°、14.78±0.2°、17.30±0.2°、18.78±0.2°、19.01±0.2°、19.25±0.2°、19.66±0.2°、19.91±0.2°、20.09±0.2°、20.80±0.2°、21.15±0.2°、21.66±0.2°、22.35±0.2°、23.57±0.2°、23.70±0.2°、24.63±0.2°、25.68±0.2°、27.05±0.2°处有衍射峰;或者In the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the B crystal form, at 4.69±0.2°, 4.91±0.2°, 6.34±0.2°, 7.19±0.2°, 9.37±0.2°, 9.83±0.2°, 10.01±0.2°, 10.32±0.2°, 11.11±0.2°, 11.39±0.2°, 12.06±0.2°, 12.69±0.2°, 13.52±0.2°, 14.07±0.2°, 14.36±0.2°, 14.78±0.2°, 17.30±0.2°, 18.78±0.2°, 19.01±0.2°, 19.25±0.2°, 19.66±0.2°, 19.91±0.2°, 20.09±0.2°, 20.80±0.2°, 21.15±0.2°, 21.66±0.2°, Diffraction peaks at 22.35±0.2°, 23.57±0.2°, 23.70±0.2°, 24.63±0.2°, 25.68±0.2°, 27.05±0.2°; or
    所述B晶型的X-射线粉末衍射图谱与图5基本上一致。The X-ray powder diffraction pattern of the crystal form B is basically consistent with that in FIG. 5 .
  14. 根据权利要求12或13所述B晶型,所述B晶型的DSC谱图在150.08±5℃处具有吸热峰。According to the crystal form B according to claim 12 or 13, the DSC spectrum of the crystal form B has an endothermic peak at 150.08±5°C.
  15. 式(I)化合物的琥珀酸盐A晶型,The crystal form A of the succinate salt of the compound of formula (I),
    Figure PCTCN2022131313-appb-100007
    Figure PCTCN2022131313-appb-100007
    所述A晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在7.13±0.2°、9.30±0.2°、11.29±0.2°、15.51±0.2°、19.89±0.2°处有衍射峰。In the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the crystal form A, there are diffraction peaks at 7.13±0.2°, 9.30±0.2°, 11.29±0.2°, 15.51±0.2°, and 19.89±0.2°.
  16. 根据权利要求15所述A晶型,其中:A crystal form according to claim 15, wherein:
    所述A晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在7.13±0.2°、9.30±0.2°、11.29±0.2°、15.51±0.2°、17.10±0.2°、19.89±0.2°、20.26±0.2°、20.91±0.2°、21.51±0.2°、22.69±0.2°、23.58±0.2°、25.42±0.2°处有衍射峰;或者In the X-ray powder diffraction pattern of the crystal form A represented by diffraction angle 2θ, at 7.13±0.2°, 9.30±0.2°, 11.29±0.2°, 15.51±0.2°, 17.10±0.2°, 19.89±0.2°, Diffraction peaks at 20.26±0.2°, 20.91±0.2°, 21.51±0.2°, 22.69±0.2°, 23.58±0.2°, 25.42±0.2°; or
    所述A晶型以衍射角2θ表示的X-射线粉末衍射图谱中,在5.01±0.2°、7.13±0.2°、9.30±0.2°、10.09±0.2°、11.29±0.2°、11.73±0.2°、13.75±0.2°、14.30±0.2°、15.18±0.2°、15.51±0.2°、16.00±0.2°、17.10±0.2°、18.25±0.2°、18.69±0.2°、19.89±0.2°、20.26±0.2°、20.91±0.2°、21.12±0.2°、21.51±0.2°、 22.69±0.2°、23.05±0.2°、23.58±0.2°、24.58±0.2°、25.42±0.2°、26.66±0.2°、27.24±0.2°和29.47±0.2°处有衍射峰;或者In the X-ray powder diffraction pattern of the crystal form A represented by the diffraction angle 2θ, at 5.01±0.2°, 7.13±0.2°, 9.30±0.2°, 10.09±0.2°, 11.29±0.2°, 11.73±0.2°, 13.75±0.2°, 14.30±0.2°, 15.18±0.2°, 15.51±0.2°, 16.00±0.2°, 17.10±0.2°, 18.25±0.2°, 18.69±0.2°, 19.89±0.2°, 20.26±0.2°, 20.91±0.2°, 21.12±0.2°, 21.51±0.2°, 22.69±0.2°, 23.05±0.2°, 23.58±0.2°, 24.58±0.2°, 25.42±0.2°, 26.66±0.2°, and Diffraction peak at 29.47±0.2°; or
    所述A晶型的X-射线粉末衍射图谱与图8基本上一致。The X-ray powder diffraction pattern of the crystal form A is basically consistent with that in FIG. 8 .
  17. 根据权利要求15或16所述A晶型,所述A晶型的DSC谱图在186.82±5℃处具有吸热峰。According to the crystal form A according to claim 15 or 16, the DSC spectrum of the crystal form A has an endothermic peak at 186.82±5°C.
  18. 根据权利要求15~17中任一项所述A晶型,所述A晶型的晶胞参数为
    Figure PCTCN2022131313-appb-100008
    Figure PCTCN2022131313-appb-100009
    α=90°,β=90°,γ=90°。
    According to the crystal form A described in any one of claims 15-17, the unit cell parameters of the crystal form A are:
    Figure PCTCN2022131313-appb-100008
    Figure PCTCN2022131313-appb-100009
    α=90°, β=90°, γ=90°.
  19. 制备权利要求1~8中任一项式(I)化合物的可药用盐的方法,所述制备方法包含了式(I)所示化合物与相应酸成盐反应的步骤。A method for preparing a pharmaceutically acceptable salt of a compound of formula (I) according to any one of claims 1 to 8, said preparation method comprising the step of reacting the compound of formula (I) with a corresponding acid to form a salt.
  20. 根据权利要求19所述制备方法,所述成盐反应的溶剂选自环己烷、正庚烷、甲苯、氯仿、乙醚、异丙醚、甲基叔丁基醚、四氢呋喃、乙二醇二甲醚、丙酮、乙酸甲酯、乙酸乙酯、乙酸异丙酯、乙酸丁酯、甲醇、乙醇、异丙醇、正丁醇和乙腈中的至少一种。According to the preparation method described in claim 19, the solvent of the salt-forming reaction is selected from cyclohexane, n-heptane, toluene, chloroform, ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran, ethylene glycol dimethyl At least one of ether, acetone, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile.
  21. 根据权利要求19或20所述制备方法,所述制备方法还包括搅拌析晶,过滤和干燥中的一种或其组合的步骤。According to the preparation method described in claim 19 or 20, the preparation method further comprises the steps of one or a combination of stirring and crystallization, filtering and drying.
  22. 制备权利要求9~11中任一项所述的C晶型的方法,包含以下步骤:The method for preparing the C crystal form described in any one of claims 9-11, comprising the following steps:
    (1-a)将己二酸和溶剂混合;(1-a) mixing adipic acid and a solvent;
    (2-a)将式(I)化合物和溶剂混合;(2-a) mixing the compound of formula (I) and a solvent;
    (3-a)混合步骤(1-a)的混合物与步骤(2-a)的混合物;(3-a) mixing the mixture of step (1-a) and the mixture of step (2-a);
    (4-a)将混合了所述已二酸和所述式(I)化合物的所得混合物搅拌,析晶;(4-a) Stir the resulting mixture mixed with the adipic acid and the compound of formula (I), and crystallize;
    (5-a)过滤出结晶,收集滤饼,干燥;(5-a) filtering out the crystals, collecting the filter cake, and drying;
    优选地,步骤(1-a)和步骤(2-a)中的溶剂选自环己烷、正庚烷、甲苯、氯仿、乙醚、异丙醚、甲基叔丁基醚、四氢呋喃、乙二醇二甲醚、丙酮、乙酸甲酯、乙酸乙酯、乙酸异丙酯、乙酸丁酯、甲醇、乙醇、异丙醇、正丁醇和乙腈中的至少一种,更优选异丙醚、甲基叔丁基醚、甲醇、异丙醇和四氢呋喃中的至少一种,最优选异丙醇;Preferably, the solvent in step (1-a) and step (2-a) is selected from cyclohexane, n-heptane, toluene, chloroform, diethyl ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran, ethylene di At least one of alcohol dimethyl ether, acetone, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, more preferably isopropyl ether, methyl At least one of tert-butyl ether, methanol, isopropanol and tetrahydrofuran, most preferably isopropanol;
    优选地,所述式(I)化合物与己二酸的摩尔比为1:2~2:1,更优选摩尔比为1:1;Preferably, the molar ratio of the compound of formula (I) to adipic acid is 1:2 to 2:1, more preferably the molar ratio is 1:1;
    优选地,所述步骤(1-a)在加热条件下进行,更优选加热温度约为40-80℃;Preferably, the step (1-a) is carried out under heating conditions, more preferably the heating temperature is about 40-80°C;
    优选地,所述步骤(4-a)在15-30℃条件下进行。Preferably, the step (4-a) is carried out at 15-30°C.
  23. 制备权利要求12~14中任一项所述的B晶型的方法,包含以下步骤:The method for preparing the B crystal form described in any one of claims 12-14, comprising the following steps:
    (1-b)将式(I)化合物和溶剂混合;(1-b) mixing the compound of formula (I) and a solvent;
    (2-b)向步骤(1-b)的混合物中加入苯甲酸;(2-b) adding benzoic acid to the mixture of step (1-b);
    (3-b)将混合了所述式(I)化合物和所述苯甲酸的所得混合物搅拌,析晶;(3-b) Stir the resulting mixture mixed with the compound of formula (I) and the benzoic acid, and crystallize;
    (4-b)过滤出并收集固体;(4-b) filtering out and collecting the solid;
    优选地,步骤(1-b)中的溶剂选自环己烷、正庚烷、甲苯、氯仿、乙醚、异丙醚、甲基叔丁基醚、四氢呋喃、乙二醇二甲醚、丙酮、乙酸甲酯、乙酸乙酯、乙酸异丙酯、乙酸丁酯、甲醇、乙醇、异丙醇、正丁醇和乙腈中的至少一种,更优选异丙醚、甲基叔丁基醚、甲醇、异丙醇和四氢呋喃中的至少一种,最优选异丙醚;Preferably, the solvent in step (1-b) is selected from cyclohexane, n-heptane, toluene, chloroform, ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran, ethylene glycol dimethyl ether, acetone, At least one of methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, more preferably isopropyl ether, methyl tert-butyl ether, methanol, At least one of isopropanol and tetrahydrofuran, most preferably isopropyl ether;
    优选地,所述式(I)化合物与苯甲酸的摩尔比为1:2~2:1,更优选摩尔比为1:1;Preferably, the molar ratio of the compound of formula (I) to benzoic acid is 1:2 to 2:1, more preferably the molar ratio is 1:1;
    优选地,所述步骤(1-b)在15-30℃条件下进行;Preferably, the step (1-b) is carried out at 15-30°C;
    优选地,所述步骤(3-b)的搅拌在加热条件下进行,更优选加热温度约为40-68℃,最优选60-68℃;Preferably, the stirring in step (3-b) is carried out under heating conditions, more preferably the heating temperature is about 40-68°C, most preferably 60-68°C;
    任选地,所述B晶型的制备方法还包括以下步骤:Optionally, the preparation method of the B crystal form also includes the following steps:
    (5-b)将步骤(4-b)收集的固体和溶剂混合;(5-b) mixing the solid collected in step (4-b) with a solvent;
    (6-b)将所得混合物搅拌;(6-b) stirring the resulting mixture;
    (7-b)过滤出并收集固体,干燥;(7-b) filter out and collect solid, dry;
    优选地,步骤(5-b)中的溶剂选自异丙醚、甲基叔丁基醚、甲醇、异丙醇和四氢呋喃中的至少一种,优选异丙醚;Preferably, the solvent in step (5-b) is selected from at least one of isopropyl ether, methyl tert-butyl ether, methanol, isopropanol and tetrahydrofuran, preferably isopropyl ether;
    优选地,步骤(6-b)在15-30℃条件下进行。Preferably, step (6-b) is carried out at 15-30°C.
  24. 制备权利要求15~18中任一项所述的A晶型的方法,包含以下步骤:The method for preparing the crystal form A described in any one of claims 15-18, comprising the following steps:
    (1-c)将式(I)化合物和溶剂混合;(1-c) mixing the compound of formula (I) and a solvent;
    (2-c)向步骤(1-c)的混合物中加入琥珀酸;(2-c) adding succinic acid to the mixture of step (1-c);
    (3-c)将混合了所述式(I)化合物和所述琥珀酸的所得混合物搅拌,析出固体;(3-c) stirring the mixture obtained by mixing the compound of formula (I) and the succinic acid to precipitate a solid;
    (4-c)过滤出并收集固体;(4-c) filtering out and collecting the solid;
    优选地,步骤(1-c)中的溶剂选自环己烷、正庚烷、甲苯、氯仿、乙醚、异丙醚、甲基叔丁基醚、四氢呋喃、乙二醇二甲醚、丙酮、乙酸甲酯、乙酸乙酯、乙酸异丙酯、乙酸丁酯、甲醇、乙醇、异丙醇、正丁醇和乙腈中的至少一种,更优选异丙醚、甲基叔丁基醚、甲醇、异丙醇和四氢呋喃中的至少一种,最优选异丙醚;Preferably, the solvent in step (1-c) is selected from cyclohexane, n-heptane, toluene, chloroform, ether, isopropyl ether, methyl tert-butyl ether, tetrahydrofuran, ethylene glycol dimethyl ether, acetone, At least one of methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, methanol, ethanol, isopropanol, n-butanol and acetonitrile, more preferably isopropyl ether, methyl tert-butyl ether, methanol, At least one of isopropanol and tetrahydrofuran, most preferably isopropyl ether;
    优选地,所述式(I)化合物与琥珀酸的摩尔比为1:2~2:1,更优选摩尔比为1:1;Preferably, the molar ratio of the compound of formula (I) to succinic acid is 1:2 to 2:1, more preferably the molar ratio is 1:1;
    优选地,所述步骤(3-c)在15-30℃条件下进行;Preferably, the step (3-c) is carried out at 15-30°C;
    优选地,所述步骤(3-c)的反应时间为1-48小时,更优选3-24小时;Preferably, the reaction time of the step (3-c) is 1-48 hours, more preferably 3-24 hours;
    任选地,所述A晶型的制备方法还包括以下步骤:Optionally, the preparation method of the A crystal form also includes the following steps:
    (5-c)将步骤(4-c)收集的固体和溶剂混合;(5-c) mixing the solid collected in step (4-c) with a solvent;
    (6-c)将所得混合物搅拌析晶;(6-c) stirring and crystallizing the resulting mixture;
    (7-c)过滤出并收集固体,干燥;(7-c) filter out and collect solid, dry;
    优选地,步骤(5-c)中的溶剂选自甲醇、乙醇、异丙醇、四氢呋喃和水中的至少一种,更优选乙醇/水的混合溶剂;Preferably, the solvent in step (5-c) is selected from at least one of methanol, ethanol, isopropanol, tetrahydrofuran and water, more preferably a mixed solvent of ethanol/water;
    优选地,所述乙醇/水的体积比约为1:1~50:1,更优选2:1~20:1,最优选10:1、9:1、8:1、7:1、6:1、5:1或4:1;Preferably, the volume ratio of ethanol/water is about 1:1-50:1, more preferably 2:1-20:1, most preferably 10:1, 9:1, 8:1, 7:1, 6 :1, 5:1 or 4:1;
    优选地,所述步骤(6-c)在15-30℃条件下进行;Preferably, the step (6-c) is carried out at 15-30°C;
    优选地,所述步骤(6-c)的反应时间为0.5-48小时。Preferably, the reaction time of the step (6-c) is 0.5-48 hours.
  25. 一种药物组合物,其包含权利要求1~8任一项所述式(I)化合物的可药用盐或权利要求9~18任一项所述晶型,以及药学上可接受的辅料。A pharmaceutical composition, which comprises the pharmaceutically acceptable salt of the compound of formula (I) according to any one of claims 1-8 or the crystal form according to any one of claims 9-18, and pharmaceutically acceptable auxiliary materials.
  26. 权利要求1~8任一项所述式(I)化合物的可药用盐、权利要求9~18任一项所述晶型或权利要求25所述的药物组合物在制备用于预防或者治疗雌激素受体相关疾病的药物中的用途;The pharmaceutically acceptable salt of the compound of formula (I) according to any one of claims 1 to 8, the crystal form according to any one of claims 9 to 18, or the pharmaceutical composition according to claim 25 is used for preventing or treating Use in drugs for estrogen receptor-related diseases;
    优选地,所述雌激素受体相关疾病为肿瘤;Preferably, the estrogen receptor-related disease is a tumor;
    更优选地,所述肿瘤为乳腺癌;More preferably, the tumor is breast cancer;
    更优选地,所述肿瘤为ER阳性乳腺癌;More preferably, the tumor is ER-positive breast cancer;
    更优选地,所述肿瘤为ER阳性乳腺癌脑转移。More preferably, the tumor is brain metastases from ER-positive breast cancer.
PCT/CN2022/131313 2021-11-12 2022-11-11 Salt of pyrrolidine compound, crystal form thereof, and preparation method therefor WO2023083292A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111339152 2021-11-12
CN202111339152.0 2021-11-12

Publications (1)

Publication Number Publication Date
WO2023083292A1 true WO2023083292A1 (en) 2023-05-19

Family

ID=86335127

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/131313 WO2023083292A1 (en) 2021-11-12 2022-11-11 Salt of pyrrolidine compound, crystal form thereof, and preparation method therefor

Country Status (2)

Country Link
TW (1) TW202327576A (en)
WO (1) WO2023083292A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019223715A1 (en) * 2018-05-23 2019-11-28 江苏恒瑞医药股份有限公司 Benzopiperidine or heteroarylpiperidine derivative, preparation method therefor, and medical application thereof
CN111499614A (en) * 2019-01-31 2020-08-07 江苏恒瑞医药股份有限公司 Tetrahydroisoquinoline derivatives, preparation method and medical application thereof
WO2021228210A1 (en) * 2020-05-15 2021-11-18 江苏先声药业有限公司 Pyrrolidine compound and use thereof
WO2021249533A1 (en) * 2020-06-12 2021-12-16 江苏先声药业有限公司 Estrogen receptor modulator compound and use thereof
WO2022166983A1 (en) * 2021-02-08 2022-08-11 贝达药业股份有限公司 Heteroarylopiperidine derivative, and pharmaceutical composition thereof and use thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019223715A1 (en) * 2018-05-23 2019-11-28 江苏恒瑞医药股份有限公司 Benzopiperidine or heteroarylpiperidine derivative, preparation method therefor, and medical application thereof
CN111499614A (en) * 2019-01-31 2020-08-07 江苏恒瑞医药股份有限公司 Tetrahydroisoquinoline derivatives, preparation method and medical application thereof
WO2021228210A1 (en) * 2020-05-15 2021-11-18 江苏先声药业有限公司 Pyrrolidine compound and use thereof
WO2021249533A1 (en) * 2020-06-12 2021-12-16 江苏先声药业有限公司 Estrogen receptor modulator compound and use thereof
WO2022166983A1 (en) * 2021-02-08 2022-08-11 贝达药业股份有限公司 Heteroarylopiperidine derivative, and pharmaceutical composition thereof and use thereof

Also Published As

Publication number Publication date
TW202327576A (en) 2023-07-16

Similar Documents

Publication Publication Date Title
EP3378861B1 (en) Acrylic acid derivative, preparation method and use in medicine thereof
TWI642667B (en) Pyridino pyrimidine derivatives, preparation method and medical use thereof
WO2021068898A1 (en) Novel kras g12c protein inhibitor, preparation method therefor, and use thereof
RU2183457C2 (en) Method for applying 5_ht5-ht1f-antagonist for inhibiting 5_ht5-ht1f-mediated neurogenic meningeal transudation
TWI651321B (en) Acrylic derivative, process for producing the same and medical use thereof
JP7021394B2 (en) Compounds as Neurokinin-1 Receptor Antagonists and Their Use
KR20150048140A (en) Crystal of dispiropyrrolidine derivative
RU2556205C2 (en) Purified pyrroloquinolinyl-pyrrolidine-2,5-dione compositions and methods of obtaining and application thereof
WO2004072080A1 (en) Certain 8-heteroaryl-6-phenyl-imidazo[1,2-a]pyrazines as modulators of hsp90 complex activity
JP6307088B2 (en) Quinazolinone and isoquinolinone derivatives
AU2014336929B9 (en) Salt and crystal forms of PLK-4 inhibitor
US10106523B2 (en) Amide compound
TWI781607B (en) A kind of immunosuppressant, its preparation method and application
WO2021228210A1 (en) Pyrrolidine compound and use thereof
WO2022237815A1 (en) Novel forms of Compound I and use thereof
KR20100091149A (en) Quinolone compound and pharmaceutical composition
TW201311672A (en) Quinazoline-7-ether compounds and methods of use
WO2021083328A1 (en) Polysubstituted isoindoline compounds, preparation method therefor, pharmaceutical composition thereof and use thereof
WO2023083292A1 (en) Salt of pyrrolidine compound, crystal form thereof, and preparation method therefor
CA3219940A1 (en) Heterocyclic compounds and methods of preparation thereof
KR20230044416A (en) Determination of indolo heptamyl oxime analogs as PARP inhibitors and method for their preparation
WO2023083283A1 (en) Drug combination for treating tumor, and application thereof
WO2022242753A1 (en) Pharmaceutically acceptable salt of pyrazoloheteroaryl derivative and crystal form thereof
CN113912608B (en) Pyrimidopyrimidinone derivatives, preparation method thereof and application thereof in medicines
JP2019519561A5 (en)

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: 22892085

Country of ref document: EP

Kind code of ref document: A1