WO2022171044A1 - 一种氧氮杂螺环化合物、其盐型及其晶型 - Google Patents

一种氧氮杂螺环化合物、其盐型及其晶型 Download PDF

Info

Publication number
WO2022171044A1
WO2022171044A1 PCT/CN2022/075259 CN2022075259W WO2022171044A1 WO 2022171044 A1 WO2022171044 A1 WO 2022171044A1 CN 2022075259 W CN2022075259 W CN 2022075259W WO 2022171044 A1 WO2022171044 A1 WO 2022171044A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
crystal form
alternatively
angles
ray powder
Prior art date
Application number
PCT/CN2022/075259
Other languages
English (en)
French (fr)
Inventor
吴凌云
贾海飞
汪秋燕
陈曙辉
陶琳
李永华
刘成洪
夏龙军
王晓霞
张晓丽
王真
赵岩
邹阳
熊剑
姚培水
张芳
文万高
刘冬梅
邝振英
Original Assignee
南昌弘益药业有限公司
南昌弘益科技有限公司
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 南昌弘益药业有限公司, 南昌弘益科技有限公司 filed Critical 南昌弘益药业有限公司
Priority to CN202280003890.4A priority Critical patent/CN115485282A/zh
Publication of WO2022171044A1 publication Critical patent/WO2022171044A1/zh

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
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • 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
    • A61K31/438The ring being spiro-condensed with carbocyclic or heterocyclic ring systems
    • 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
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring

Definitions

  • the present invention relates to an oxazaspiro compound, its salt form, its crystal form and preparation method, in particular to compound III and its pharmaceutically acceptable salt, and its salt form crystal form.
  • Lysine specific demethylase 1 (LSD1, also known as KDM1A) is the first reported histone lysine demethylase, which regulates the methylation of histone lysine by regulating the methylation of histone lysine. It is widely involved in transcriptional regulation and affects many physiological processes such as cell proliferation and differentiation, and pluripotency of embryonic stem cells. [Yujiang Shi, Fei Lan, Caitlin Matson et al., Cell, 2004, 941–953] [Daniel P. Mould, Alison E. McGonagle, Daniel H.
  • the LSD1 structure consists of three main parts: the N-terminal SWIRM domain, the C-terminal aminooxidase domain (AOL) and the central Tower domain. [Ruchi Anand, Ronen Marmorstein, Journal of Biological Chemistry, 2007, 35425–35429].
  • the C-terminal aminooxidase domain includes two active pockets, one for FAD binding and the other for recognition and substrate binding [Pete Stavropoulos, Günter Blobel, André Hoelz, Nature Structral & Molecular Biology , 2006, 626-632].
  • the function of the SWIRM domain has not yet been clearly concluded.
  • the Tower domain is the binding domain of LSD1 to other protein factors. After LSD1 binds to different protein factors, it acts on different substrates, thereby exerting different regulatory effects on histone and gene expression. In addition, LSD1 also regulates the methylation status of some non-histone substrates, including tumor suppressor gene p53 and DNA methyltransferase 1 (DNMT1), etc. [Yi Chao Zheng, Jinlian Ma, Zhiru Wang, Medicinal Research Reviews, 2015, 1032–1071].
  • DNMT1 DNA methyltransferase 1
  • LSD1 is a FAD-dependent aminooxidase, of which proton transfer is considered as the most likely oxidation mechanism [Zheng Y C, Yu B, Chen Z S, et al. Epigenomics, 2016, 8, 651-666.].
  • proton transfer the N-CH 3 bond of the substrate is converted into an imine bond, and this imide ion intermediate undergoes a hydrolysis reaction to generate demethylated amine on one side and formaldehyde on the other side.
  • LSD1 is aberrantly expressed in many different types of tumors.
  • LSD1 is highly expressed in acute myeloid leukemia (AML) subtypes and is an important factor in maintaining the potential of leukemia stem cells (LSC).
  • LSD1 is highly expressed in various solid tumors, such as lung, breast, prostate, liver and pancreatic cancer, and is closely related to poor tumor prognosis.
  • LSD1 inhibits the expression of cadherin and is closely related to tumor invasion and epithelial-mesenchymal transition (EMT) [Hosseini A, Minucci S. Epigenomics, 2017, 9, 1123-1142.].
  • EMT epithelial-mesenchymal transition
  • the present invention provides compound III:
  • the present invention provides compound IV':
  • n is 0-2, preferably 1.5-2.
  • the present invention provides crystal form A of compound IV, and its X-ray powder diffraction (XRPD) pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 7.42 ⁇ 0.20°, 16.90 ⁇ 0.20° and 19.99 ⁇ 0.20°;
  • the X-ray powder diffraction pattern of the above-mentioned A crystal form has characteristic diffraction peaks at the following 2 ⁇ angles: 7.42 ⁇ 0.20°, 8.46 ⁇ 0.20°, 15.88 ⁇ 0.20°, 16.90 ⁇ 0.20°, 17.80 ⁇ 0.20 °, 18.77 ⁇ 0.20°, 19.99 ⁇ 0.20° and 22.65 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above-mentioned A crystal form has characteristic diffraction peaks at the following 2 ⁇ angles: 7.42 ⁇ 0.20°, 8.46 ⁇ 0.20°, 15.88 ⁇ 0.20°, 16.90 ⁇ 0.20°, 17.80 ⁇ 0.20 °, 18.27 ⁇ 0.20°, 18.77 ⁇ 0.20°, 19.99 ⁇ 0.20°, 21.98 ⁇ 0.20° and 22.65 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above-mentioned A crystal form has characteristic diffraction peaks at the following 2 ⁇ angles: 7.42 ⁇ 0.20°, 8.46 ⁇ 0.20°, 10.79 ⁇ 0.20°, 11.69 ⁇ 0.20°, 13.66 ⁇ 0.20 degrees °, 21.64 ⁇ 0.20°, 21.98 ⁇ 0.20°, 22.30 ⁇ 0.20°, 22.65 ⁇ 0.20°, 23.15 ⁇ 0.20° and 23.44 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above-mentioned A crystal form has characteristic diffraction peaks at the following 2 ⁇ angles: 7.42°, 8.46°, 10.79°, 11.69°, 13.66°, 14.78°, 15.88°, 16.40° , 16.90°, 17.80°, 18.27°, 18.77°, 19.57°, 19.99°, 21.23°, 21.64°, 21.98°, 22.30°, 22.65°, 23.15° and 23.44°.
  • the X-ray powder diffraction pattern of the above-mentioned A crystal form has characteristic diffraction peaks at the following 2 ⁇ angles: 7.42 ⁇ 0.20°, 19.99 ⁇ 0.20°, and/or 16.90 ⁇ 0.20°, and/or 8.46 ⁇ 0.20°, and/or 10.79 ⁇ 0.20°, and/or 11.69 ⁇ 0.20°, and/or 13.66 ⁇ 0.20°, and/or 14.78 ⁇ 0.20°, and/or 15.88 ⁇ 0.20°, and/or 16.40 ⁇ 0.20° , and/or 17.80 ⁇ 0.20°, and/or 18.27 ⁇ 0.20°, and/or 18.77 ⁇ 0.20°, and/or 19.57 ⁇ 0.20°, and/or 21.23 ⁇ 0.20°, and/or 21.64 ⁇ 0.20°, and /or 21.98 ⁇ 0.20°, and/or 22.30 ⁇ 0.20°, and/or 22.65 ⁇ 0.20°, and/or 23.15 ⁇ 0.20°, and/or 23.44 ⁇
  • the XRPD pattern of the above-mentioned crystal form A is basically as shown in FIG. 1 .
  • the XRPD of the above-mentioned crystal form A uses Cu-K ⁇ radiation.
  • the differential scanning calorimetry curve has an endothermic peak starting point at 215.4 ⁇ 5°C.
  • the DSC analysis method of the above crystal form A is as follows: the scanning rate is 10°C/min, and the temperature range is 25-350°C.
  • the DSC spectrum of the above-mentioned crystal form A is shown in FIG. 2 .
  • thermogravimetric analysis curve (TGA) of the above-mentioned crystal form A has a weight loss of 0.89% at 180 ⁇ 5°C.
  • the TGA analysis method of the above-mentioned crystal form A is as follows: the scanning rate is 10°C/min, and the temperature range is room temperature-350°C.
  • the TGA spectrum of the above-mentioned A crystal form is shown in FIG. 3 .
  • the present invention provides compound V:
  • the invention provides the B crystal form of compound V, and its X-ray powder diffraction (XRPD) pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 16.33 ⁇ 0.20°, 18.98 ⁇ 0.20° and 22.27 ⁇ 0.20°;
  • the X-ray powder diffraction pattern of the above crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 7.08 ⁇ 0.20°, 10.51 ⁇ 0.20°, 14.54 ⁇ 0.20°, 16.33 ⁇ 0.20°, 17.37 ⁇ 0.20 °, 17.91 ⁇ 0.20°, 18.98 ⁇ 0.20° and 22.27 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 7.08 ⁇ 0.20°, 10.51 ⁇ 0.20°, 14.54 ⁇ 0.20°, 16.33 ⁇ 0.20°, 17.37 ⁇ 0.20 °, 17.91 ⁇ 0.20°, 18.98 ⁇ 0.20°, 19.56 ⁇ 0.20°, 21.30 ⁇ 0.20°, 22.27 ⁇ 0.20°, 23.58 ⁇ 0.20° and 26.66 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 7.08 ⁇ 0.20°, 7.40 ⁇ 0.20°, 10.51 ⁇ 0.20°, 12.27 ⁇ 0.20°, 12.47 ⁇ 0.20 degrees degrees degrees °.
  • the X-ray powder diffraction pattern of the above crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 7.08°, 7.40°, 10.51°, 12.27°, 12.47°, 14.15°, 14.54°, 16.33° , 17.37°, 17.91°, 18.60°, 18.98°, 19.20°, 19.56°, 20.13°, 21.06°, 21.30°, 22.27°, 22.74°, 23.58°, 24.68°, 25.16°, 25.84°, 26.66°, 27.77 °, 29.03°, 30.18°, 30.70°, 31.54°, 32.18°, 32.96°, 34.03°, 34.53°, 36.26° and 37.41°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 16.33 ⁇ 0.20°, 18.98 ⁇ 0.20°, and/or 7.08 ⁇ 0.20°, and/or 7.40 ⁇ 0.20°, and/or 10.51 ⁇ 0.20°, and/or 12.27 ⁇ 0.20°, and/or 12.47 ⁇ 0.20°, and/or 14.15 ⁇ 0.20°, and/or 14.54 ⁇ 0.20°, and/or 17.37 ⁇ 0.20° , and/or 17.91 ⁇ 0.20°, and/or 18.60 ⁇ 0.20°, and/or 19.20 ⁇ 0.20°, and/or 19.56 ⁇ 0.20°, and/or 20.13 ⁇ 0.20°, and/or 21.06 ⁇ 0.20°, and /or 21.30 ⁇ 0.20°, and/or 22.27 ⁇ 0.20°, and/or 22.74 ⁇ 0.20°, and/or 23.58 ⁇ 0.20°, and/or 24.68 ⁇
  • the XRPD pattern of the above-mentioned crystal form B is basically as shown in FIG. 4 .
  • the XRPD of the above-mentioned crystal form B uses Cu-K ⁇ radiation.
  • the differential scanning calorimetry curve has an endothermic peak at 127.4 ⁇ 5°C.
  • the DSC analysis method of the above crystal form B is as follows: the scanning rate is 10°C/min, and the temperature range is 25-350°C.
  • the DSC spectrum of the above-mentioned crystal form B is shown in FIG. 5 .
  • thermogravimetric analysis curve (TGA) of the above-mentioned crystal form B has a weight loss of 8.84% at 150 ⁇ 5°C.
  • the TGA analysis method of the above crystal form B is as follows: the scanning rate is 10°C/min, and the temperature range is room temperature-350°C.
  • the TGA spectrum of the above-mentioned B crystal form is shown in FIG. 6 .
  • the present invention also provides a preparation method of compound II,
  • the organic solvent is methanol, ethanol, isopropanol, acetonitrile or isopropyl acetate;
  • the above-mentioned preparation method of compound II comprises the following steps:
  • the organic solvent is methanol, ethanol, isopropanol, acetonitrile or isopropyl acetate;
  • Solvent 1 is saturated sodium bicarbonate solution
  • the extractant is dichloromethane.
  • the above-mentioned organic solvent is acetonitrile.
  • the above The molar ratio to compound I is 0.5-1:1; preferably 1:1.
  • the volume-to-mass ratio V:m of the above-mentioned organic solvent to compound I is 5-20 (mL/g).
  • the volume-to-mass ratio V:m of the above-mentioned acetonitrile and compound I is 5-20 (mL/g).
  • reaction temperature of the above-mentioned compound I to prepare the compound I-A is 25-85°C.
  • the present invention also provides the application of compound III, compound IV', compound IV, compound V, crystal form A of compound IV and crystal form B of compound V in the preparation of medicines for treating LSD1-related diseases.
  • the present invention also provides applications of Compound III, Compound IV', Compound IV, Compound V, Crystal Form A of Compound IV, and Crystal Form B of Compound V in the preparation of medicines for treating LSD1-related diseases;
  • the diseases are hematological tumors, Small cell lung cancer, squamous non-small cell lung cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, glioma, or Ewing's sarcoma.
  • the hematological tumor is preferably human acute myeloid leukemia.
  • Compound III, compound IV', compound V, crystal form A of compound IV and crystal form B of compound V of the present invention have good pharmacokinetic properties in rodents, including good oral bioavailability, oral exposure , half-life and clearance rate, etc.; the A crystal form of compound IV has good in vitro activity and in vivo efficacy; the A crystal form of compound IV is stable, less affected by light, heat and humidity, and has broad drug prospects; the benefits of intermediate splitting: A single configuration with better optical purity was obtained by screening chemical resolution conditions.
  • the intermediate compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by their combination with other chemical synthesis methods, and those skilled in the art.
  • Well-known equivalents, preferred embodiments include, but are not limited to, the examples of the present invention.
  • the structure of the compound of the present invention can be confirmed by conventional methods well known to those skilled in the art. If the present invention relates to the absolute configuration of the compound, the absolute configuration can be confirmed by conventional technical means in the art. For example, single crystal X-ray diffraction method (SXRD), the cultured single crystal is collected by Bruker D8 venture diffractometer, the light source is CuK ⁇ radiation, and the scanning mode is: After scanning and collecting relevant data, the crystal structure was further analyzed by the direct method (Shelxs97), and the absolute configuration could be confirmed.
  • SXRD single crystal X-ray diffraction method
  • the cultured single crystal is collected by Bruker D8 venture diffractometer
  • the light source is CuK ⁇ radiation
  • the scanning mode is: After scanning and collecting relevant data, the crystal structure was further analyzed by the direct method (Shelxs97), and the absolute configuration could be confirmed.
  • DCM dichloromethane
  • DMF N,N-dimethylformamide
  • DMSO dimethyl sulfoxide
  • EtOH for ethanol
  • MeOH for methanol
  • 2-MeTHF 2-methyl Dioxane for dioxane
  • ACN for acetonitrile
  • Toluene for toluene
  • Acetone for acetone
  • EtOAc for ethyl acetate
  • THF tetrahydrofuran
  • H 2 O for water
  • TosOH for p-toluenesulfonic acid
  • FaSSIF for simulated fasting artificial intestinal fluid
  • FeSSIF stands for simulated satiety artificial intestinal fluid
  • SGF stands for simulated artificial gastric fluid
  • LOQ stands for limit of quantitation.
  • Figure 7 Single crystal ellipsoid diagram of compound IV.
  • X-ray powder diffraction (X-ray powder diffractometer, XRPD) method of the present invention test parameters are shown in Table 3.
  • DSC Differential Thermal Analysis
  • Thermogravimetric analysis (Thermal Gravimetric Analyzer, TGA) method of the present invention test parameters are shown in Table 5.
  • DVS Dynamic Vapor Sorption
  • Hygroscopic classification ⁇ W% deliquescence Absorbs enough water to form a liquid Very hygroscopic ⁇ W% ⁇ 15% hygroscopic 15%> ⁇ W% ⁇ 2% slightly hygroscopic 2%> ⁇ W% ⁇ 0.2%
  • ⁇ W% represents the hygroscopic weight gain of the test product at 25 ⁇ 1°C and 80 ⁇ 2%RH.
  • the stirring uniformity of the precipitated solid is better, and the ee value is kept above 90%. It is found that the crystallization temperature should not be too low through the screening of the above conditions. Therefore, the final resolution condition is 1eq of chiral acid, and the solvent acetonitrile is used. The amount was 20 V, and the temperature was resolved at 60-25 °C to obtain a single configuration.
  • reaction solution was stirred at 56 ° C for 2 hours, naturally cooled to 25 ° C and stirred for 12 hours, the reaction solution was filtered, the solid was washed with acetonitrile (600 mL x 2), dried under vacuum at 40 ° C, to the solid Acetonitrile (3858 mL) was added to the mixture, and the reaction solution was stirred at 85°C for 1.5 hours, at 60°C for 1 hour, at 50°C for 0.5 hours, at 40°C for 1 hour, at 30°C for 1 hour, and at 30°C for 1 hour.
  • reaction solution was diluted with ethyl acetate (250 mL), washed successively with hydrochloric acid (1%, 100 mL ⁇ 4), saturated sodium bicarbonate (100 mL ⁇ 1) and saturated brine (100 mL ⁇ 1), and the organic phase was washed with anhydrous sodium sulfate Dry, filter, and concentrate under reduced pressure to give crude product.
  • Example 5 Stability test of crystal form A of compound IV in different solvents
  • Test number solvent appear solid crystal form 1 Isopropyl acetate suspension Form A 2 1,4-Dioxane suspension Form A 3 2-Methyltetrahydrofuran suspension Form A 4 n-heptane suspension Form A 5 Acetonitrile/n-heptane, 1:1 suspension Form A 6 Acetonitrile/2-Methyltetrahydrofuran, 1:1 suspension Form A 7 Acetonitrile/1,4-dioxane, 1:1 suspension Form A 8 Acetonitrile/Isopropyl Acetate, 1:1 suspension Form A 9 Water/2-Methyltetrahydrofuran, 1/35 Precipitation after dissolving Form A 10 tetrahydrofuran suspension Form A 11 Ethyl acetate suspension Form A 12 Acetonitrile suspension Form A 13 Methyl tert-butyl ether suspension Form A 14 2-Methyltetrahydrofuran/n-heptane, 1:1 suspension Form A 15 2-Methyltetra
  • the crystal form A of compound IV has good stability in solvent, and the crystal form A belongs to the stable crystal form.
  • Example 7 Study on the hygroscopicity of A crystal form of compound IV
  • Two samples of crystal form A of compound IV were weighed respectively, one was an illumination sample, and the other was an illumination control sample.
  • the lighted sample is placed in a clean weighing bottle, flattened into a single layer, not covered by anything, and placed in a light box for light exposure.
  • the light conditions are 5000 ⁇ 500lux (visible light) and 90 ⁇ w/cm 2 (ultraviolet) .
  • the control samples were packaged in the same way as the illuminated samples, but the weighing bottle was covered with aluminum foil.
  • the two samples were investigated simultaneously for 10 days, and the XRPD detection results are shown in Table 15 below.
  • the crystal form A of compound IV has good stability under the conditions of high temperature, high humidity and strong light, and the crystal form A belongs to the stable crystal form.
  • Example 9 Solubility determination of crystal form A of compound IV in buffers with different pH values
  • Step 1 Weigh about 3 mg of the crystal form A sample of compound IV, put it into a 1.5 mL liquid phase vial, add 1 mL of medium, and place it on a constant temperature mixer (37 °C, 800 rpm) to observe the dissolution. Dissolve under certain medium conditions, proceed to step 2, otherwise go to step 4;
  • Step 2 Weigh about 7 mg of the crystal form A sample of Compound IV, put it into a 1.5 mL liquid phase vial, add 1 mL of medium, and place it on a constant temperature mixer (37 °C, 800 rpm) to observe the dissolution. Dissolve under certain medium conditions, continue to step 3, otherwise go to step 4;
  • Step 3 Weigh about 10 mg of the crystal form A sample of Compound IV, put it in a 1.5 mL liquid phase vial, add 1 mL of medium, and place it on a constant temperature mixer (37°C, 800 rpm);
  • Step 4 Sampling at 18 hours, recording the dissolution, all the sample solutions are clarified, the pH value is measured, and the concentration is measured by HPLC after dilution. The data are shown in Table 16.
  • Intravenous and oral vehicle is a mixed vehicle of 10% dimethyl sulfoxide and 90% 10% hydroxypropyl beta cyclodextrin.
  • This project uses four male SD rats and two SD rats for intravenous injection at a dose of 0.5mg/kg. Plasma samples, the other two SD rats were given oral gavage at a dose of 1 mg/kg.
  • CD-1 mice male, 7-9 weeks old, Shanghai Institute of Family Planning Science
  • Intravenous and oral vehicle is a mixed vehicle of 10% dimethyl sulfoxide and 90% 10% hydroxypropyl beta cyclodextrin.
  • This project used four male CD-1 mice, two mice were administered intravenously at a dose of 1 mg/kg, collected at 0h (before administration) and 0.0833, 0.25, 0.5, 1, 2 after administration , 4, 8, 24h plasma samples, the other two mice were given oral gavage at a dose of 2mg/kg, collected at 0h (before administration) and 0.25, 0.5, 1, 2, 4 after administration. 8, 24h plasma samples, collect whole blood samples within 24h, and quantitatively analyze the blood drug concentration by LC-MS/MS analysis method.
  • Form A of the compound IV of the present invention has good pharmacokinetic properties in rodents, including good oral bioavailability, oral exposure, half-life and clearance rate.
  • the pharmacokinetics of the compounds after oral administration were tested in beagle dogs by standard protocols.
  • the candidate compounds were formulated into clear solutions and administered to beagle dogs by single intravenous injection and oral administration.
  • the vehicle for intravenous injection is a mixed solvent of 10% dimethyl sulfoxide and 90% 10% hydroxypropyl beta cyclodextrin.
  • the oral vehicle was 0.5% methylcellulose (4000 cp).
  • This project uses four male beagle dogs, two beagle dogs for intravenous injection, the dosage is 0.5mg/kg, after the collection of administration, 0.033, 0.0833, 0.25, 0.5, 1, 2, 4, 8, 12, 24h plasma samples, two beagle dogs were administered orally by gavage, the dose was 1mg/kg, and the plasma samples were collected at 0.083, 0.25, 0.5, 1, 2, 4, 8, 12, 24h after administration , Quantitative analysis of blood drug concentration by LC-MS/MS analysis method, and calculation of pharmacokinetic parameters, such as peak concentration (C max ), clearance rate (CL), half-life (T 1/2 ), tissue distribution (Vdss), Area under the drug-time curve (AUC 0-last ), bioavailability (F), etc.
  • C max peak concentration
  • CL clearance rate
  • T 1/2 half-life
  • Vdss tissue distribution
  • AUC 0-last Area under the drug-time curve
  • bioavailability bioavailability
  • the crystal form A of the compound IV of the present invention has good pharmacokinetic properties in large animals, including good oral bioavailability, oral exposure, half-life and clearance rate.
  • LSD1 enzyme binds to histone H3K4 methylated peptide substrate to generate demethylation activity to generate H 2 O 2 .
  • the inhibitory effect of compounds on LSD1 activity was detected by combining peroxidase and fluorescent reagent Amplex Red to detect H 2 O 2 generated by the enzymatic reaction.
  • Compound working solution preparation According to the compound arrangement diagram, take 1 ⁇ L of compound per well from the 1st to 10th column of the compound plate into a new corresponding well of the compound plate, and add 39 ⁇ L of 1X LSD1 buffer per well; positive control well Take 1 ⁇ L of 100% DMSO per well and add 39 ⁇ L of 1X LSD1 buffer to each well for later use.
  • substrate mixed solution includes 7.5 ⁇ L of 2X LSD1 buffer and 2.5 ⁇ L of histone H3K4 monomethylated peptide substrate solution.
  • the test plate is sealed The membrane was placed at 25°C and incubated for 60 minutes.
  • the blank control is a control well without enzyme; the positive control is a control well containing enzyme, substrate and 0.5% DMSO.
  • the results are shown in Table 20.
  • RPMI 1640 medium was purchased from Ekosai Biotechnology Co., Ltd., and penicillin/streptomycin antibiotics were purchased from HyClone.
  • CellTiter-Glo Luminescent Cell Viability Assay chemiluminescence detection reagent for cell viability
  • Fetal bovine serum (FBS) Kasumi-1 cells were purchased from American type culture collection (ATCC, American type culture collection). Nivo Multilabel Analyzer (PerkinElmer).
  • IC 50 was calculated using the software Prism 8 for the inhibition rate, and the results are shown in Table 21.
  • Fetal bovine serum (FBS) was purchased from Ekosai Biotechnology Co., Ltd.
  • IMDM medium was purchased from American type culture collection (ATCC, American type culture collection), and penicillin/streptomycin double antibody was purchased from HyClone .
  • CellTiter-Glo Luminescent Cell Viability Assay chemiluminescence detection reagent for cell viability
  • KG-1 cells were purchased from the European Standard Cell Collection (ECACC, European Collection of Authenticated Cell Cultures). Nivo Multilabel Analyzer (PerkinElmer).
  • IC 50 was calculated using the software GraphPad Prism 9 for the inhibition rate, and the results are shown in Table 22.
  • the purpose of this experiment was to evaluate the antitumor effect of compound IV on human acute myeloid leukemia Kasumi-1 cells in CB-17 SCID mouse subcutaneous tumor model.
  • mice Mice; Strain: CB-17 SCID mice; Age and body weight: 6-8 weeks old, body weight 16-21 grams; Gender: female; Supplier: Shanghai Jihui Laboratory Animal Breeding Co., Ltd.
  • Human acute myeloid leukemia Kasumi-1 cells were cultured in suspension in RPMI-1640 medium with 20% fetal bovine serum, 1% penicillin and streptomycin, and cultured at 37°C and 5% CO 2 . When cell saturation is 80%-90%, harvest cells, count, adjust to 10 x 106 cells/mL and resuspend in phosphate buffered saline PBS.
  • Kasumi-1 cells (plus Matrigel, volume 1:1) were subcutaneously inoculated into the right back of each mouse, when the average tumor volume reached about 135mm, group administration was started and random grouping was used. , start dosing.
  • the experimental solvent is 0.5% methyl cellulose solution: Weigh 2.5 g of methyl cellulose, dissolve it in 400 mL of ultrapure water, stir evenly, dilute to 500 mL with ultrapure water, and store at 4°C.
  • Azacitidine (5-Azacytidine, manufacturer MedChemExpress, batch number 28452) formulation preparation: Weigh 1 mg of 5-azacytidine, add 14.2 mL of PBS, dissolve to obtain a clear solution, and store at 4°C.
  • Preparation of compound IV preparation Weigh 14.2 mg of compound IV, add 16.026 mL of 0.5% MC, dissolve to obtain a clear solution with a concentration of 0.45 mg/mL, and store at 4°C.
  • the purpose of this experiment was to evaluate the antitumor effect of compound IV on human leukemia KG-1 cells in BALB/c nude mice subcutaneously transplanted tumor model.
  • mice mouse; strain: BALB/c nude mice; age and body weight: 6-8 weeks old, body weight 18-24 grams; gender: female; supplier: Shanghai Lingchang Biotechnology Co., Ltd.
  • Human acute myeloid leukemia KG-1 cells were cultured in monolayer in vitro, and the culture conditions were 20% fetal bovine serum, 1% penicillin and streptomycin in IMDM medium, 37°C, 5% CO 2 for culture and passage. When the cell saturation was 80%-90%, the cells were harvested, counted, and the number of cell suspension was adjusted to 5 ⁇ 10 7 cells/mL.
  • 0.2mL (10 ⁇ 10 6 cells) KG-1 cells were subcutaneously inoculated on the right back of each mouse, when the average tumor volume reached about 106mm 3 , random grouping was used, and administration was started .
  • the experimental solvent is 0.5% methyl cellulose solution: Weigh 2.5 g of methyl cellulose, dissolve it in 400 mL of ultrapure water, stir evenly, dilute to 500 mL with ultrapure water, and store at 4°C.
  • Azacitidine (5-Azacytidine, manufacturer MedChemExpress, batch number 103024) formulation preparation: Weigh 1.6 mg of 5-azacytidine, add 16 mL of PBS, dissolve to obtain a clear solution, and store at 4°C.
  • Preparation of compound IV preparation Weigh 5 mg of compound IV, add 4.224 mL of 0.5% MC, dissolve to obtain a clear solution with a concentration of 0.6 mg/mL, and store at 4°C.
  • the single drug of compound IV has inhibitory effect on the growth of human leukemia KG-1 cells in BALB/c nude mice subcutaneous xenografted tumors.
  • the combined administration of compound IV and azacitidine has a significant inhibitory effect on tumor growth, and the tumor-inhibiting effect is obviously better than that of compound IV and azacitidine alone.
  • Tumor-bearing mice showed good tolerance to Compound IV at all doses.
  • the purpose of this experiment was to evaluate the antitumor effect of compound IV on human leukemia MV-4-11-Luc cells in NCG mouse xenograft model.
  • mice mouse; strain: NCG mouse; age and body weight: 6-7 weeks old, body weight 19.1-24.7 g; gender: female; supplier: Jiangsu Jicui Yaokang Biotechnology Co., Ltd.
  • MV-4-11-Luc cells were cultured in IMDM medium containing 10% fetal bovine serum. MV-4-11-Luc cells in exponential growth phase were collected and resuspended in PBS to an appropriate concentration for inoculation into the tail vein of mice.
  • mice Female NCG mice were inoculated with 1 ⁇ 10 7 MV-4-11-Luc cells in the tail vein. All mice were imaged on day 7 after inoculation and grouped according to fluorescence values, and the day of group administration was defined as day 0.
  • the experimental solvent is 0.5% methyl cellulose solution: Weigh 2.5 g of methyl cellulose, dissolve it in 400 mL of ultrapure water, stir evenly, dilute to 500 mL with ultrapure water, and store at 4°C.
  • Azacytidine (5-Azacytidine, manufacturer MedChemExpress, batch number 103024) preparation preparation: Weigh 0.675 mg of 5-Azacytidine, add 9 mL of normal saline, vortex, and ultrasonically shake to obtain a clear solution, which is stored at 4°C.
  • Compound IV preparation preparation Weigh 4.8 mg of Compound IV, add 24.219 mL of 0.5% methylcellulose (4000 cps), vortex, and ultrasonically shake to obtain a clear solution, which is stored at 4°C.
  • the experimental indicator is to examine whether tumor growth is inhibited, delayed or cured.
  • tumor growth was monitored by in vivo bioluminescence imaging in mice.
  • the imaging frequency was twice a week after group administration.
  • the imaging system used was the IVIS Lumina III mouse in vivo imager (PerkinElmer, USA).
  • the specific experimental steps are as follows:
  • Imaging mice were subcutaneously injected with D-Luciferin imaging substrate (PerkinElmer, XenoLight D-Luciferin (K+Salt), Cat. No. 122799) through the neck at a dose of 150 mg/kg and an injection volume of 5 ⁇ l/g;
  • D-Luciferin imaging substrate PerkinElmer, XenoLight D-Luciferin (K+Salt), Cat. No. 122799
  • mice to be imaged After the mice to be imaged enter anesthesia, transfer the mice to the imager, and keep the animals under anesthesia during the imaging process by placing their mouths and noses in the cannula of the anesthesia system. Large, placed from left to right, belly facing up, mouse tail placed in black light-shielding sleeve;
  • TGI The antitumor efficacy of the compounds was evaluated by TGI (%), which reflected the tumor growth inhibition rate.
  • TV was replaced by the fluorescence signal value.
  • Azacitidine at the dose of 0.75mg/kg has no significant antitumor effect on human acute myeloid leukemia MV-4-11-Luc cells in the NCG mouse system xenograft model.
  • Compound IV has significant tumor suppressing effect on human acute myeloid leukemia MV-4-11-Luc cells in the NCG mouse system transplanted tumor model at doses of 0.5 mg/kg and 1.0 mg/kg.
  • Compound IV (0.50mg/kg) combined with azacitidine (0.75mg/kg) and compound IV (1.0mg/kg) combined with azacitidine (0.75mg/kg) had significant tumor inhibition. Effect.

Landscapes

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

Abstract

涉及一种氧氮杂螺环化合物、其盐型及其晶型。具体涉及化合物Ⅲ及其药学上可接受的盐,及其盐型的晶型以及制备方法,及其作为LSD1抑制剂在治疗血液肿瘤、小细胞肺癌、鳞状非小细胞肺癌、乳腺癌、前列腺癌、肝癌、胰腺癌、胶质瘤或尤文氏肉瘤等疾病中的应用。

Description

一种氧氮杂螺环化合物、其盐型及其晶型
本申请主张如下优先权:
CN202110179260.X,申请日2021年2月9日。
技术领域
本发明涉及一种氧氮杂螺环化合物、其盐型及其晶型以及制备方法,具体涉及化合物Ⅲ及其药学上可接受的盐,及其盐型的晶型。
背景技术
组蛋白的甲基化状态由组蛋白甲基转移酶和组蛋白去甲基化酶共同调控。赖氨酸特异性去甲基化酶(Lysine specific demethylase 1,LSD1,又名KDM1A)是第一个被报道的组蛋白赖氨酸去甲基化酶,通过调控组蛋白赖氨酸的甲基化状态,广泛参与转录调控,影响细胞增殖和分化、胚胎干细胞多能性等诸多生理过程。[Yujiang Shi,Fei Lan,Caitlin Matson et al.,Cell,2004,941–953][Daniel P.Mould,Alison E.McGonagle,Daniel H.Wiseman et al.,Medicinal Research Reviews,2015,35,586–618]。LSD1结构包括三个主要部分:N-末端的SWIRM结构域,C-末端的氨基氧化酶结构域(AOL)和中央的Tower域。[Ruchi Anand,Ronen Marmorstein,Journal of Biological Chemistry,2007,35425–35429]。C-末端的氨基氧化酶结构域包括两个活性口袋,一个是FAD结合的位点,另一个是用于识别并与底物结合的位点[Pete Stavropoulos,Günter Blobel,André Hoelz,Nature Structral&Molecular Biology,2006,626-632]。SWIRM结构域的功能还没有明确的结论,它不直接参与FAD或者底物的结合,但是这个区域的突变或者是去除都会降低LSD1的活性,因此推测该区域可能是通过调整构象,影响活性区域的作用。[Yong Chen,Yuting Yang,Feng Wang et al.,Biochemistry,2006,13956–13961]。Tower结构域是LSD1与其他蛋白因子的结合域。LSD1与不同蛋白因子相结合后,作用于不同底物,从而对组蛋白以及基因表达起到不同的调控作用。此外,LSD1还调控部分非组蛋白底物的甲基化状态,包括抑癌基因p53和DNA甲基转移酶1(DNA methyltransferase 1,DNMT1)等[Yi Chao Zheng,Jinlian Ma,Zhiru Wang,Medicinal Research Reviews,2015,1032–1071]。
LSD1是FAD依赖的氨基氧化酶,其中质子转移被认为是其最可能的氧化机理[Zheng Y C,Yu B,Chen Z S,et al.Epigenomics,2016,8,651-666.]。首先通过质子转移,将底物的N-CH 3键转化成亚胺键,这个亚胺离子中间体发生水解反应,一边生成去甲基的胺,另一边生成甲醛。在这个催化循环过程中,FAD被还原成FADH2,随后又被一分子的氧气氧化回到FAD,同时生成一分子H2O2[Yujiang Shi,Fei Lan,Caitlin Matson,Cell,2004,941–953]。
LSD1在多种不同类型的肿瘤中异常表达。LSD1在急性髓性白血病(acute myeloid leukemia,AML)亚型中高表达,是维持白血病干细胞(leukemia stem cell,LSC)潜能的重要因素。LSD1在多种实体瘤如肺癌、乳腺癌、前列腺癌、肝癌和胰腺癌中高表达,与肿瘤的预后不良密切相关。LSD1抑制钙粘蛋白的表达,与肿瘤的侵袭和上皮-间质转移(epithelial-mesenchymal transition,EMT)密切相关[Hosseini A,Minucci S.Epigenomics,2017,9,1123-1142.]。
LSD1抑制剂目前没有药物获批上市,已有8个药物处于临床研究阶段,主要用于血液肿瘤、小细胞肺癌和尤文氏肉瘤等疾病的治疗。然而,面对巨大的未满足市场,该领域仍然需要活性更好,药代动力学参数更优的候选化合物推进临床试验,以满足治疗需求。
发明内容
本发明提供了化合物Ⅲ:
Figure PCTCN2022075259-appb-000001
本发明提供了化合物Ⅳ’:
Figure PCTCN2022075259-appb-000002
其中,n为0-2,优选为1.5-2。
本发明的一些方案中,上述化合物Ⅳ’具有化合物Ⅳ的结构:
Figure PCTCN2022075259-appb-000003
本发明提供了化合物Ⅳ的A晶型,其X射线粉末衍射(XRPD)图谱在下列2θ角处具有特征衍射峰:7.42±0.20°、16.90±0.20°和19.99±0.20°;
Figure PCTCN2022075259-appb-000004
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.42±0.20°、8.46±0.20°、15.88±0.20°、16.90±0.20°、17.80±0.20°、18.77±0.20°、19.99±0.20°和22.65±0.20°。
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.42±0.20°、8.46±0.20°、15.88±0.20°、16.90±0.20°、17.80±0.20°、18.27±0.20°、18.77±0.20°、19.99±0.20°、21.98±0.20°和22.65±0.20°。
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.42±0.20°、8.46±0.20°、10.79±0.20°、11.69±0.20°、13.66±0.20°、14.78±0.20°、15.88±0.20°、16.40±0.20°、16.90±0.20°、17.80±0.20°、18.27±0.20°、18.77±0.20°、19.57±0.20°、19.99±0.20°、21.23±0.20°、21.64±0.20°、21.98±0.20°、22.30±0.20°、22.65±0.20°、23.15±0.20°和23.44±0.20°。
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.42°、8.46°、10.79°、11.69°、13.66°、14.78°、15.88°、16.40°、16.90°、17.80°、18.27°、18.77°、19.57°、19.99°、21.23°、21.64°、21.98°、22.30°、22.65°、23.15°和23.44°。
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.42±0.20°、19.99±0.20°、和/或16.90±0.20°、和/或8.46±0.20°、和/或10.79±0.20°、和/或11.69±0.20°、和/或13.66±0.20°、和/或14.78±0.20°、和/或15.88±0.20°、和/或16.40±0.20°、和/或17.80±0.20°、和/或18.27±0.20°、和/或 18.77±0.20°、和/或19.57±0.20°、和/或21.23±0.20°、和/或21.64±0.20°、和/或21.98±0.20°、和/或22.30±0.20°、和/或22.65±0.20°、和/或23.15±0.20°、和/或23.44±0.20°。
本发明的一些方案中,上述A晶型,其XRPD图谱基本如图1所示。
本发明的一些方案中,上述A晶型,其XRPD使用Cu-Kα辐射。
本发明的一些方案中,上述A晶型的XRPD图谱解析数据如表1所示。
表1 化合物Ⅳ的A晶型的XRPD解析数据
Figure PCTCN2022075259-appb-000005
本发明的一些方案中,上述A晶型,其差示扫描量热曲线在215.4±5℃有一个吸热峰的起始点。
本发明的一些方案中,上述A晶型的DSC分析方法如下:扫描速率为10℃/分钟,温度范围为25-350℃。
本发明的一些方案中,上述A晶型,其DSC图谱如图2所示。
在本发明的一些方案中,上述A晶型的热重分析曲线(TGA)在180±5℃时失重达0.89%。
在本发明的一些方案中,上述A晶型的TGA分析方法如下:扫描速率为10℃/分钟,温度范围为室温-350℃。
在本发明的一些方案中,上述A晶型的TGA图谱如图3所示。本发明提供了化合物Ⅴ:
Figure PCTCN2022075259-appb-000006
本发明提供了化合物Ⅴ的B晶型,其X射线粉末衍射(XRPD)图谱在下列2θ角处具有特征衍射峰:16.33±0.20°、18.98±0.20°和22.27±0.20°;
Figure PCTCN2022075259-appb-000007
本发明的一些方案中,上述B晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.08±0.20°、10.51±0.20°、14.54±0.20°、16.33±0.20°、17.37±0.20°、17.91±0.20°、18.98±0.20°和22.27±0.20°。
本发明的一些方案中,上述B晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.08±0.20°、10.51±0.20°、14.54±0.20°、16.33±0.20°、17.37±0.20°、17.91±0.20°、18.98±0.20°、19.56±0.20°、21.30±0.20°、22.27±0.20°、23.58±0.20°和26.66±0.20°。
本发明的一些方案中,上述B晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.08±0.20°、7.40±0.20°、10.51±0.20°、12.27±0.20°、12.47±0.20°、14.15±0.20°、14.54±0.20°、16.33±0.20°、17.37±0.20°、17.91±0.20°、18.60±0.20°、18.98±0.20°、19.20±0.20°、19.56±0.20°、20.13±0.20°、21.06±0.20°、21.30±0.20°、22.27±0.20°、22.74±0.20°、23.58±0.20°、24.68±0.20°、25.16±0.20°、25.84±0.20°、26.66±0.20°、27.77±0.20°、29.03±0.20°、30.18±0.20°、30.70±0.20°、31.54±0.20°、32.18±0.20°、32.96±0.20°、34.03±0.20°、34.53±0.20°、36.26±0.20°和37.41±0.20°。
本发明的一些方案中,上述B晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.08°、7.40°、10.51°、12.27°、12.47°、14.15°、14.54°、16.33°、17.37°、17.91°、18.60°、18.98°、19.20°、19.56°、20.13°、21.06°、21.30°、22.27°、22.74°、23.58°、24.68°、25.16°、25.84°、26.66°、27.77°、29.03°、30.18°、30.70°、31.54°、32.18°、32.96°、34.03°、34.53°、36.26°和37.41°。
本发明的一些方案中,上述B晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:16.33±0.20°、18.98±0.20°、和/或7.08±0.20°、和/或7.40±0.20°、和/或10.51±0.20°、和/或12.27±0.20°、和/或12.47±0.20°、和/或14.15±0.20°、和/或14.54±0.20°、和/或17.37±0.20°、和/或17.91±0.20°、和/或18.60±0.20°、和/或19.20±0.20°、和/或19.56±0.20°、和/或20.13±0.20°、和/或21.06±0.20°、和/或21.30±0.20°、和/或22.27±0.20°、和/或22.74±0.20°、和/或23.58±0.20°、和/或24.68±0.20°、和/或25.16±0.20°、和/或25.84±0.20°、和/或26.66±0.20°、和/或27.77±0.20°、和/或29.03±0.20°、和/或30.18±0.20°、和/或30.70±0.20°、和/或31.54±0.20°、和/或32.18±0.20°、和/或32.96±0.20°、和/或34.03±0.20°、和/或34.53±0.20°、和/或36.26±0.20°、和/或37.41±0.20°。
本发明的一些方案中,上述B晶型,其XRPD图谱基本如图4所示。
本发明的一些方案中,上述B晶型,其XRPD使用Cu-Kα辐射。
本发明的一些方案中,上述B晶型的XRPD图谱解析数据如表2所示。
表2 化合物Ⅴ的B晶型的XRPD解析数据
Figure PCTCN2022075259-appb-000008
Figure PCTCN2022075259-appb-000009
本发明的一些方案中,上述B晶型,其差示扫描量热曲线在127.4±5℃有一个吸热峰的峰值。
本发明的一些方案中,上述B晶型的DSC分析方法如下:扫描速率为10℃/分钟,温度范围为25-350℃。
本发明的一些方案中,上述B晶型,其DSC图谱如图5所示。
在本发明的一些方案中,上述B晶型的热重分析曲线(TGA)在150±5℃时失重达8.84%。
在本发明的一些方案中,上述B晶型的TGA分析方法如下:扫描速率为10℃/分钟,温度范围为室温-350℃。
本发明的一些方案中,上述B晶型的TGA图谱如图6所示。
本发明还提供了化合物Ⅱ的制备方法,
Figure PCTCN2022075259-appb-000010
其包含如下步骤:
Figure PCTCN2022075259-appb-000011
其中,
有机溶剂为甲醇、乙醇、异丙醇、乙腈或乙酸异丙酯;
手性酸为
Figure PCTCN2022075259-appb-000012
本发明的一些方案中,上述化合物Ⅱ制备方法,其包含如下步骤:
Figure PCTCN2022075259-appb-000013
其中,
有机溶剂为甲醇、乙醇、异丙醇、乙腈或乙酸异丙酯;
手性酸为
Figure PCTCN2022075259-appb-000014
溶剂1为饱和碳酸氢钠溶液;
萃取剂为二氯甲烷。
本发明的一些方案中,上述有机溶剂为乙腈。
本发明的一些方案中,上述
Figure PCTCN2022075259-appb-000015
与化合物Ⅰ的摩尔比为0.5-1:1;优选为1:1。
本发明的一些方案中,上述有机溶剂与化合物Ⅰ的体积质量比V:m为5-20(mL/g)。
本发明的一些方案中,上述乙腈与化合物Ⅰ的体积质量比V:m为5-20(mL/g)。
本发明的一些方案中,上述化合物Ⅰ制备化合物Ⅰ-A的反应温度为25-85℃。
本发明还提供了化合物Ⅲ、化合物Ⅳ’、化合物Ⅳ、化合物Ⅴ、化合物Ⅳ的A晶型以及化合物Ⅴ的B晶型在制备治疗LSD1相关疾病的药物中的应用。
本发明还提供了化合物Ⅲ、化合物Ⅳ’、化合物Ⅳ、化合物Ⅴ、化合物Ⅳ的A晶型以及化合物Ⅴ的B晶型在制备治疗LSD1相关疾病的药物中的应用;所述疾病为血液肿瘤、小细胞肺癌、鳞状非小细胞肺癌、乳腺癌、前列腺癌、肝癌、胰腺癌、胶质瘤或尤文氏肉瘤。所述血液肿瘤优选为人急性髓系白血病。
技术效果
本发明化合物Ⅲ、化合物Ⅳ’、化合物Ⅴ、化合物Ⅳ的A晶型以及化合物Ⅴ的B晶型在啮齿类动物体内具有良好的药代动力学性质,包括良好的口服生物利用度,口服暴露量,半衰期和清除率等;化合物Ⅳ的A晶型具有良好的体外活性和体内药效;化合物Ⅳ的A晶型稳定、受光热湿度影响小、成药前景广阔;中间体拆分的有益之处:通过化学拆分条件筛选得到光学纯度较好的单一构型。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在含有下列含义。一个特定的短语或术语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文出现商品名时,旨在指代其对应的商品或其活性成分。
本发明的中间体化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明具体实施方式的化学反应是在合适的溶剂中完成的,所述的溶剂须适合于本发明的化学变化及其所需的试剂和物料。为了获得本发明的化合物,有时需要本领域技术人员在已有实施方式的基础上对合成步骤或者反应流程进行修改或选择。
本发明的化合物可以通过本领域技术人员所熟知的常规方法来确认结构,如果本发明涉及化合物的绝对构型,则该绝对构型可以通过本领域常规技术手段予以确证。例如单晶X射线衍射法(SXRD),把培养出的单晶用Bruker D8 venture衍射仪收集衍射强度数据,光源为CuKα辐射,扫描方式:
Figure PCTCN2022075259-appb-000016
扫描,收 集相关数据后,进一步采用直接法(Shelxs97)解析晶体结构,便可以确证绝对构型。
下面会通过实施例具体描述本发明,这些实施例并不意味着对本发明的任何限制。
本发明所使用的所有溶剂是市售的,无需进一步纯化即可使用。
本发明采用下述缩略词:DCM代表二氯甲烷;DMF代表N,N-二甲基甲酰胺;DMSO代表二甲亚砜;EtOH代表乙醇;MeOH代表甲醇;2-MeTHF代表2-甲基四氢呋喃;Dioxane代表二氧六环;ACN代表乙腈;Toluene代表甲苯;Acetone代表丙酮;EtOAc代表乙酸乙酯;THF代表四氢呋喃;H 2O代表水;TosOH代表对甲苯磺酸;FaSSIF代表模拟空腹人工肠液;FeSSIF代表模拟饱腹人工肠液;SGF代表模拟人工胃液;LOQ代表定量限。
化合物依据本领域常规命名原则或者使用
Figure PCTCN2022075259-appb-000017
软件命名,市售化合物采用供应商目录名称。
附图说明
图1:化合物Ⅳ的A晶型的XRPD图谱;
图2:化合物Ⅳ的A晶型的DSC图谱;
图3:化合物Ⅳ的A晶型的TGA图谱;
图4:化合物Ⅴ的B晶型的XRPD图谱;
图5:化合物Ⅴ的B晶型的DSC图谱;
图6:化合物Ⅴ的B晶型的TGA图谱;
图7:化合物Ⅳ的单晶椭球图。
具体实施方式
本发明X-射线粉末衍射(X-ray powder diffractometer,XRPD)方法,测试参数见表3。
表3 XRPD测试参数
Figure PCTCN2022075259-appb-000018
本发明差热分析(Differential Scanning Calorimeter,DSC)方法,测试参数见表4。
表4 DSC测试参数
Figure PCTCN2022075259-appb-000019
本发明热重分析(Thermal Gravimetric Analyzer,TGA)方法,测试参数见表5。
表5 TGA测试参数
Figure PCTCN2022075259-appb-000020
本发明动态蒸汽吸附分析(Dynamic Vapor Sorption,DVS)方法,测试参数见表6。
表6 DVS测试参数
仪器厂家型号 SMS/DVS intrinsic
测试条件 称取约10mg样品,进行测试
温度 25℃
平衡 dm/dt:0.01%/min
干燥 25℃,0%RH干燥2h
RH(%)测试梯级 5%RH
RH(%)测试梯级范围 0%-95%-0%RH
引湿性评价分类如下表7所示:
表7 引湿性评价分类表
吸湿性分类 ΔW%
潮解 吸收足量水分形成液体
极具吸湿性 ΔW%≥15%
有吸湿性 15%>ΔW%≥2%
略有吸湿性 2%>ΔW%≥0.2%
无或几乎无吸湿性 ΔW%<0.2%
注:ΔW%表示受试品在25±1℃和80±2%RH下的吸湿增重。
实施例1:化合物I的制备
Figure PCTCN2022075259-appb-000021
合成路线:
Figure PCTCN2022075259-appb-000022
第一步
将化合物1(200g,706mmol)溶于二氯甲烷(600mL)形成混悬液,将反应液冷却至0℃,缓慢的向反应液中加入氢氧化钠溶液(1mol/L,706mL),反应液在25℃下搅拌1小时,反应液用水H2O(200mL)稀释,用二氯甲烷萃取(600mL x 2),有机相用水(500mL x 1),饱和食盐水(1000mL x 1)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物2。 1H NMR(400MHz,CDCl 3)δ7.18-7.14(m,2H),7.08-7.06(m,1H),6.95-6.93(m,2H),2.48-2.44(m,1H),1.76-1.77(m,1H),0.97-0.94(m,1H),0.92-0.88(m,1H)。
第二步
将化合物2(86.6g,650mmol)溶解在二氯甲烷(2000mL)中,向反应液中加入化合物3(182.6g,715mmol),醋酸硼氢化钠(344.5g,1.63mol),反应液在25℃下搅拌3小时,用饱和碳酸氢钠(2500mL)淬灭,用二氯甲烷(1500mL x 2)萃取,合并有机相用饱和食盐水(1000mL x 1)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品化合物I。 1H NMR(400MHz,CDCl 3)δ7.19-7.18(m,2H),7.17-7.00(m,1H),6.96-6.94(m,2H),3.91-3.90(m,1H),3.58-3.51(m,1H),3.50-3.24(m,3H),3.23-3.22(m,2H),2.26-2.22(m,1H),1.96(m,1H),1.56-1.55(m,1H),1.53-1.52(m,4H),1.51-1.50(m,1H),1.39-1.38(m,9H),1.00-0.99(m,1H),0.92(m,1H)。MS-ESI计算值[M+H] +373实测值373。
实施例2:化合物II的制备
Figure PCTCN2022075259-appb-000023
合成路线:
Figure PCTCN2022075259-appb-000024
第一步
化学拆分条件筛选-溶剂和手性酸种类筛选
室温条件下,分别在不同的反应瓶中加入原料I(50mg)和不同的溶剂各1mL,再加入不同手性酸(1eq)试剂。该反应先在50-60℃搅拌2h,后降温至25℃,搅拌12h,有固体析出的收集固体,固体用NaOH(1mol/L)溶液游离送SFC检测(手性柱:Chiralpak AD-3 50×4.6mm I.D.,3μm;流动相:[CO 2-0.05%的二乙胺甲醇溶液];梯度:40%(0.05%的二乙胺甲醇溶液),3mL/minn),第一个峰保留时间=0.425min,第二个峰保留时间=0.678min。实验结果如下表8所示:
Figure PCTCN2022075259-appb-000025
表8 溶剂和手性酸种类筛选
Figure PCTCN2022075259-appb-000026
结论:以(-)-O,O-二对甲苯甲酰基-L-酒石酸(手性酸5)为手性酸,乙腈为溶剂会有较高的选择性。以此为基础继续优化拆分条件,“-”表示未见明显固体析出。
手性酸当量筛选:
室温条件下,在反应瓶中加入原料I(500mg),乙腈(5mL),再加入不同当量的(-)-O,O-二对甲苯甲酰基-L-酒石酸。在50-60℃下搅拌2h,降温至25℃搅拌12小时,有固体析出,收集固体,固体用NaOH(1mol/L)溶液游离送SFC检测,实验结果如下表9所示:
表9 手性酸当量筛选
酸当量 0.5eq 0.6eq 0.8eq 1.0eq
产率 32.12% 40.61% 44.77% 44.61%
第二个峰ee值 98.674% 97.582% 95.840% 96.672%
结论:手性酸为1eq时产率和ee值相对较优,以此为基础继续优化。
溶剂乙腈体积与原料I质量比V mL/m筛选
室温条件下,向不同的反应瓶中加入原料I(500mg),(-)-O,O-二对甲苯甲酰基-L-酒石酸(1eq),不同体积数的乙腈,反应在50-60℃下搅拌2h,0℃下静置12小时,有固体析出,收集固体,固体用NaOH(1mol/L)溶液游离送SFC检测。实验结果如下表10所示:
表10 溶剂乙腈体积与原料I质量比筛选
Figure PCTCN2022075259-appb-000027
当V/m=20时析出固体搅拌均一性比较好,且ee值保持在90%以上,通过以上条件筛选发现析晶温度不宜过低,故拆分条件最终为手性酸1eq,溶剂乙腈使用量为20V,温度在60-25℃下拆分得到单一构型。
化合物I-A的制备
将化合物I(121.1g x 2,299.6mmol x 2)溶解在乙腈(2422mL x 2)中,向反应液中加入化合物(-)-O,O-二对甲苯甲酰基-L-酒石酸(115.8g x 2,299.6mmol x 2),反应液在56℃下搅拌2小时,自然降温至25℃搅拌12小时,将反应液过滤,固体用乙腈(600mL x 2)洗涤,在40℃下真空干燥,向固体中加入乙腈(3858mL),反应液在85℃下搅拌1.5小时,在60℃下搅拌1小时,在50℃下搅拌0.5小时,在40℃下搅拌1小时,在30℃下搅拌1小时,在26℃下搅拌0.5小时,在26℃下静置12小时,将反应液过滤,固体用乙腈(500mL x 2)洗涤后真空干燥得到化合物I-A。送SFC检测(手性柱:Chiralpak AD-3 50×4.6mm I.D.,3μm;流动相:[CO 2-0.05%的二乙胺甲醇溶液];梯度:40%(0.05%的二乙胺甲醇溶液),3mL/minn),第二个峰:保留时间=0.676min,ee值99.208%。
第二步
将化合物I-A(101.1g,133mmol)溶解在饱和碳酸氢钠溶液(1250mL)中,反应液在25℃下搅拌0.5小时,反应液用二氯甲烷(900mL x 2)萃取,有机相用饱和食盐水(1247mL x 1)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到化合物II。 1H NMR(400MHz,CD 3OD)7.14-7.10(m,2H),7.03-7.00(m,1H),6.96-6.94(m,2H),3.90-3.87(m,1H),3.54-3.43(m,4H),3.22-3.21(m,2H),2.21-2.17(m,1H),2.05-1.96(m,1H),1.85-1.80(m,1H),1.62-1.48(m,4H),1.43-1.35(m,1H),1.35(s,9H),0.97-0.88(m,2H)。MS-ESI计算值[M+H] +373实测值373。
实施例3:化合物III的制备
Figure PCTCN2022075259-appb-000028
合成路线:
Figure PCTCN2022075259-appb-000029
第一步
将化合物II(48.7g,128mmol)溶解在二氯甲烷(600mL)中,将反应液冷却到0℃,向反应液中加入三乙胺(19.4g,192mmol)和三氟乙酸酐(40.4g,192mmol),反应液在26℃下反应10小时,反应液依次用饱和碳酸氢钠(800mL),稀盐酸(0.1mol/L,800mL),水(800mL)和饱和食盐水(800mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到化合物III-A。 1H NMR(400MHz,CDCl 3)δ7.26-7.23(m,2H),7.19-7.15(m,1H),7.00-6.98(m,2H),4.58-4.54(m,1H),4.03-3.99(m,1H),3.90-3.86(m,1H),3.50-3.47(m,2H),3.22-3.17(m,2H),3.02-2.65(m,1H),2.3-2.26(m,1H),2.01-1.99(s,2H),1.62-1.52(m,6H),1.38(m,9H)。MS-ESI[M+Na] +491,实测值491。
第二步
将化合物III-A(57.2g,117mmol)溶解在乙酸乙酯(306mL)中,将反应液冷却至0℃,向反应液中加入氯化氢乙酸乙酯溶液(4mol/L,117mL),反应液在25℃下搅拌2小时,将反应液过滤,滤饼用正庚烷(150mL x 2)洗涤得到化合物III-B。 1H NMR(400MHz,CD 3OD)7.33-7.29(m,2H),7.24-7.20(m,1H),7.15-7.13(m,2H),4.71-4.64(m,1H),4.15-4.04(m,2H),3.25-3.22(m,5H),2.49-2.44(m,1H),2.30-2.18(m,2H),2.03-1.77(m,4H),1.66-1.61(m,1H),1.49-1.43(m,1H)。MS-ESI[M+H] +369,实测值369。
第三步
将化合物III-C(10.92g,51.56mmol),羰基二咪唑(9.16g,56.47mmol)和二异丙基乙胺(19.04g,147.32mmol)溶于乙腈(100mL)中,在28℃下搅拌3小时。向反应液中加入III-B(20g,49.11mmol),反应液在28℃下搅拌12小时。反应液用乙酸乙酯(250mL)稀释,依次用盐酸(1%,100mL×4),饱和碳酸氢钠(100mL×1)和饱和食盐水(100mL×1)洗涤,有机相用无水硫酸钠干燥,过滤,减压浓缩得到粗产品。粗产品经硅胶柱层析法(石油醚/乙酸乙酯=4/1-1/1)分离纯化得III-D。 1H NMR(400MHz,CD 3OD)δ7.31-7.28(m,2H),7.22-7.18(m,1H),7.13-7.11(m,2H),4.69-4.65(m,1H),4.13-4.09(m,1H),4.03-3.99(m,1H),3.86-3.78(m,2H),3.47-3.33(m,2H),3.23-3.12(m,1H),2.46-2.41(m,1H),2.20-2.08(m,2H),1.77-1.62(m,4H),1.56-1.50(m,1H),1.46-1.44(m,1H),1.41(s,9H),1.25-1.22(m,2H),0.89-0.91(m,2H)。MS-ESI计算值[M+H] +552,实 测值552。
第四步
将化合物III-D(20.0g,36.09mmol)溶于乙酸乙酯(50mL)中,0℃下加入硫酸(10.84g,108.27mmol)的乙酸乙酯(50mL)溶液,在25℃下反应2小时。0℃下向反应液加入饱和碳酸钠(30mL)和饱和碳酸氢钠(20mL)。5分钟后用乙酸乙酯(100mL×2)萃取,合并有机相,有机相用饱和食盐水(50mL×1)洗,无水硫酸钠干燥,过滤,减压浓缩得粗产品,粗产品经硅胶柱层析法(二氯甲烷/甲醇=100/1-20/1)纯化得化合物III-E。 1H NMR(400MHz,CD 3OD)δ7.31-7.27(m,2H),7.22-7.18(m,1H),7.13-7.11(m,2H),4.69-4.65(m,1H),4.14-4.10(m,1H),4.04-4.00(m,1H),3.95-3.87(m,2H),3.47-3.38(m,2H),3.21-3.15(m,2H),2.46-2.41(m,1H),2.23-2.18(m,1H),2.14-2.09(m,1H),1.79-1.72(m,2H),1.62-1.54(m,2H),1.46-1.41(m,1H),0.94-0.91(m,2H),0.82-0.79(m,2H)。MS-ESI计算值[M+H] +452,实测值452。
第五步
将化合物III-E(15.1g,33.15mmol)溶于甲醇(150mL)中,加入碳酸钾(9.16g,66.30mmol),28℃下反应2小时。将反应液过滤,滤液用水(150mL)稀释,二氯甲烷(200mL,100mL,50mL x 2)萃取,合并有机相,有机相用饱和食盐水(100mL×1)洗,无水硫酸钠干燥,过滤,减压浓缩得III。 1H NMR(400MHz,CD 3OD)δ7.24-7.20(m,2H),7.13-7.09(m,1H),7.06-7.04(m,2H),4.02-3.99(m,1H),3.94-3.89(m,2H),3.67-3.58(m,2H),3.48-3.45(m,2H),2.31-2.27(m,1H),2.15-2.10(m,1H),1.95-1.90(m,1H),1.81-1.74(m,2H),1.70-1.65(m,2H),1.61-1.58(m,1H),1.08-0.99(m,2H),0.95-0.92(m,2H),0.82-0.79(m,2H)。MS-ESI计算值[M+H] +356,实测值356。
实施例4:化合物Ⅳ及其A晶型制备
Figure PCTCN2022075259-appb-000030
合成路线:
Figure PCTCN2022075259-appb-000031
将化合物III-D(2.8g,5.08mmol)溶于甲醇(30m第L),一步向溶液中加入碳酸钾(1.40g,10.15mmol),反应液在28℃下搅拌12小时,过滤,滤液中加入水(30mL),用乙酸乙酯(130mL x 2)萃取,合并有机相用饱和食盐水(50mL x 1)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到化合物III-F。MS-ESI计算值[M+H] + 456,实测值456。
第二步
将化合物III-F(1g,2.19mmol)与2-MeTHF(10mL)搅拌混匀,向悬浊液中加入一水合对甲苯磺酸(1.07g,5.49mmol)的2-MeTHF(5mL)溶液,反应液在50℃下搅拌12小时,反应液中有大量固体析出,过滤,滤饼用2-MeTH(3mL x 3)洗涤,真空干燥得到化合物Ⅳ的A晶型。 1H NMR(400MHz,CD3OD)δ7.70(d,J=8.0Hz,4H),7.33-7.26(m,2H),7.26-7.20(m,5H),7.18-7.13(m,2H),4.21-4.09(m,2H),4.08-4.02(m,1H),4.01-3.88(m,2H),3.49-3.35(m,2H),3.03-2.99(m,1H),2.56-2.50(m,1H),2.42-2.32(m,7H),2.03-1.96(m,1H),1.91-1.76(m,3H),1.65-1.50(m,2H),1.42-1.28(m,5H)。MS-ESI计算值[M+H] +356,实测值356。XRPD,DSC,TGA检测结果依次如图1、图2和图3。
取适量化合物Ⅳ配制成甲醇的饱和溶液,静置培养单晶,约4天,有透明晶体生成,送SC-XRD测试,单晶结构解析结果如图7所示。
实施例5:化合物Ⅳ的A晶型在不同溶剂中的稳定性试验
分别称取约100mg的化合物Ⅳ的A晶型在不同溶剂中配制成50℃的悬浊液,在50℃下搅拌24小时,反应液冷却至室温,过滤收集所得固体并进行XRPD测试,实验结果如下表11所示:
表11 化合物Ⅳ的A晶型在不同溶剂中的稳定性试验结果
试验编号 溶剂 显现 固体晶型
1 醋酸异丙酯 悬浊液 A晶型
2 1,4-二氧六环 悬浊液 A晶型
3 2-甲基四氢呋喃 悬浊液 A晶型
4 正庚烷 悬浊液 A晶型
5 乙腈/正庚烷,1:1 悬浊液 A晶型
6 乙腈/2-甲基四氢呋喃,1:1 悬浊液 A晶型
7 乙腈/1,4-二氧六环,1:1 悬浊液 A晶型
8 乙腈/醋酸异丙酯,1:1 悬浊液 A晶型
9 水/2-甲基四氢呋喃,1/35 溶清后析出 A晶型
10 四氢呋喃 悬浊液 A晶型
11 乙酸乙酯 悬浊液 A晶型
12 乙腈 悬浊液 A晶型
13 甲基叔丁基醚 悬浊液 A晶型
14 2-甲基四氢呋喃/正庚烷,1:1 悬浊液 A晶型
15 2-甲基四氢呋喃/甲基叔丁基醚,1:1 悬浊液 A晶型
结论:化合物Ⅳ的A晶型在溶剂中具有良好的稳定性,A晶型属于稳定晶型。
实施例6:化合物Ⅳ的A晶型含盐系数测试
基于对甲苯磺酸与主成分结构差异,使用HPLC方法测试化合物Ⅳ的A晶型中对甲苯磺酸含量,HPLC分析方法具体色谱条件见表12,测试结果见表13。
表12 对甲苯磺酸含量测试色谱条件
Figure PCTCN2022075259-appb-000032
表13 化合物Ⅳ的A晶型对甲苯磺酸含量测试结果
Figure PCTCN2022075259-appb-000033
结论:化合物Ⅳ的A晶型各批次对甲苯磺酸含量的实测值与理论值相符,实测值与理论值误差小于0.03,本品含两个对甲苯磺酸。
实施例7:化合物Ⅳ的A晶型引湿性研究
参考药物引湿性试验指导原则(《中国药典》2020年版(四部)通则9103),对化合物Ⅳ的A晶型样品进行引湿性考察。引湿性检测结果见表14。
表14 引湿性试验结果
Figure PCTCN2022075259-appb-000034
结论:化合物Ⅳ的A晶型根据引湿增重结果,本品略有引湿性。
实施例8:化合物Ⅳ的A晶型固体预稳定性试验研究
参考中国药典2020年版四部9001“原料药物与制剂稳定性试验指导原则”和国家食品药品监督管理局药品审评中心颁布的《化学药物(原料药和制剂)稳定性研究技术指导原则(修订)》以及ICH Q1B的规定要求,对化合物Ⅳ的A晶型进行影响因素试验。
1.光稳定性试验:
分别称取化合物Ⅳ的A晶型样品两份,1份为光照样品,1份为光照对照样品。光照样品放入干净的称量瓶中,平铺成单层,不被任何东西覆盖,敞口放入光照箱中进行光照,光照条件为5000±500lux(可见光)与90μw/cm 2(紫外)。对照样品的包装方式与光照样品一致,但在称量瓶外面覆盖铝膜。两份样品同时考察10天,XRPD检测结果见下表15。
2.温度影响试验:
称取化合物Ⅳ的A晶型样品放入敞口的称量瓶中,60℃条件下放置30天,XRPD检测结果见下表15。
3.湿度影响试验:
每份样品放入敞口的称量瓶中,在25℃/92.5%RH条件下放置30天,XRPD检测结果见下表15。
表15 化合物Ⅳ的A晶型的固体稳定性试验结果
Figure PCTCN2022075259-appb-000035
结论:化合物Ⅳ的A晶型在高温、高湿、强光照条件下具有良好的稳定性,A晶型属于稳定晶型。
实施例9:化合物Ⅳ的A晶型在不同pH值缓冲液中溶解度测定
步骤一:称取化合物Ⅳ的A晶型样品约3mg,置于1.5mL的液相小瓶内,加入1mL媒介,置于恒温混匀仪(37℃,800rpm)上,观察溶解情况,如果化合物在某媒介条件下溶解,则继续步骤二,否则进入步骤四;
步骤二:称取化合物Ⅳ的A晶型样品约7mg,置于1.5mL的液相小瓶内,加入1mL媒介,置于恒温混匀仪(37℃,800rpm)上,观察溶解情况,如果化合物在某媒介条件下溶解,则继续步骤三,否则进入步骤四;
步骤三:称取化合物Ⅳ的A晶型样品约10mg,置于1.5mL的液相小瓶内,加入1mL媒介,置于恒温混匀仪(37℃,800rpm)上;
步骤四:18h取样,记录溶解情况,样品溶液全部澄清,测定pH值,稀释后用HPLC测定浓度,数据见表16。
表16 化合物Ⅳ的A晶型溶解度试验结果(37℃)
Figure PCTCN2022075259-appb-000036
结论:化合物Ⅳ的A晶型pH7.4以下缓冲溶液和纯化水中都表现出高溶解性。
生物活性评价
实验例1:化合物Ⅳ的A晶型药代动力学评价
1.1化合物Ⅳ的A晶型啮齿类动物药代动力学评价
1.1.1化合物在SD大鼠体内的药代动力学评价
实验目的:测试化合物在SD大鼠体内的药代动力学
实验材料:
SD大鼠(雄性,150-180g,北京维通利华)
实验操作:
以标准方案测试化合物静脉注射及口服给药后的啮齿类动物药代特征,实验中候选化合物配成澄清溶液,给予SD大鼠单次静脉注射及口服给药。静注及口服溶媒为10%二甲基亚砜与90%的10%的羟丙基β环糊精配成的混合溶媒。该项目使用四只雄性SD大鼠,两只SD大鼠进行静脉注射给药,给药剂量为0.5mg/kg,收集给药后0.083,0.25,0.5,1,2,4,8,24h的血浆样品,另外两只SD大鼠口服灌胃给药,给药剂量为1mg/kg,收集给药后0.25,0.5,1,2,4,8,24h的血浆样品,以LC-MS/MS分析方法定量分析血药浓度,并计算药代参数, 如达峰浓度(C max),清除率(CL),半衰期(T 1/2),组织分布(Vdss),药时曲线下面积(AUC 0-last),生物利用度(F)等。实验结果如表17所示:
表17 药代动力学测试结果
Figure PCTCN2022075259-appb-000037
1.1.2化合物在CD-1小鼠体内的药代动力学评价
实验目的:测试化合物在CD-1小鼠体内的药代动力学
实验材料:
CD-1小鼠(雄性,7-9周龄,上海市计划生育科学研究所)
实验操作:
以标准方案测试化合物静脉注射及口服给药后的啮齿类动物药代特征,实验中候选化合物配成澄清溶液,给予小鼠单次静脉注射及口服给药。静注及口服溶媒为10%二甲基亚砜与90%的10%的羟丙基β环糊精配成的混合溶媒。该项目使用四只雄性CD-1小鼠,两只小鼠进行静脉注射给药,给药剂量为1mg/kg,收集0h(给药前)和给药后0.0833,0.25,0.5,1,2,4,8,24h的血浆样品,另外两只小鼠口服灌胃给药,给药剂量为2mg/kg,收集0h(给药前)和给药后0.25,0.5,1,2,4,8,24h的血浆样品,收集24小时内的全血样品,以LC-MS/MS分析方法定量分析血药浓度。并计算药代参数,如达峰浓度(C max),清除率(CL),半衰期(T 1/2),组织分布(Vdss),药时曲线下面积(AUC 0-last),生物利用度(F)等。实验结果如表18:
表18 药代动力学测试结果
Figure PCTCN2022075259-appb-000038
结论:本发明化合物Ⅳ的A晶型在啮齿类动物体内具有良好的药代动力学性质,包括良好的口服生物利用度,口服暴露量,半衰期和清除率等。
1.2化合物Ⅳ的A晶型大动物药代动力学评价
实验目的:测试化合物在比格犬体内的药代动力学
实验材料:
比格犬(雄性,5-15kg,北京玛斯生物技术有限公司)
实验操作:
以标准方案测试化合物口服给药后的比格犬的药代特征,实验中候选化合物配成澄清溶液,给予比格犬单次静脉注射和口服给药。静注溶媒为10%二甲基亚砜与90%的10%的羟丙基β环糊精配成的混合溶媒。口服溶媒为0.5%甲基纤维素(4000cp)。该项目使用四只雄性比格犬,两只比格犬进行静脉注射给药,给药剂量为0.5mg/kg,收集给药后0.033,0.0833,0.25,0.5,1,2,4,8,12,24h的血浆样品,两只比格犬口服灌胃给药,给药剂量为1mg/kg,收集给药后0.083,0.25,0.5,1,2,4,8,12,24h的血浆样品,以LC-MS/MS分析方法定量分析血药浓度,并计算药代参数,如达峰浓度(C max),清除率(CL),半衰期(T 1/2),组织分布(Vdss),药时曲 线下面积(AUC 0-last),生物利用度(F)等。实验结果如表19所示:
表19 药代动力学测试结果
Figure PCTCN2022075259-appb-000039
结论:本发明化合物Ⅳ的A晶型在大动物体内具有良好的药代动力学性质,包括良好的口服生物利用度,口服暴露量,半衰期和清除率等。
实验例2:化合物的Ⅳ酶活性评价
实验目的:利用酶荧光偶联法分析受试化合物Ⅳ对LSD1酶活性的抑制作用。
实验原理:LSD1酶与组蛋白H3K4单甲基化肽(methylated peptide)底物相结合产生去甲基化活性生成H 2O 2。通过过氧化物酶(peroxidase)和荧光试剂Amplex Red联合检测酶反应生成的H 2O 2,来检测化合物对LSD1活性的抑制作用。
实验方法:本实验设双复孔,实验重复两次。
化合物工作液浓度的配制:
1)将10mM待测化合物用100%DMSO稀释至2mM,即取5μL化合物储存液放入386孔化合物板第1列,再加入20μL 100%DMSO混匀;
2)在384孔化合物板的第2列至第10列中分别加入20μL 100%DMSO;
3)从第1列取10μL化合物到第2列混匀,再从第2列取10μL化合物到第3列混匀,重复以上步骤至第10列,完成化合物的梯度稀释;
4)化合物工作液配制:按照化合物排布图,从化合物板第1列至第10列中取1μL每孔化合物到新的对应化合物板孔中,加入39μL每孔1X LSD1缓冲液;阳性对照孔取1μL每孔100%DMSO,加入39μL每孔1X LSD1缓冲液备用。
实验步骤:
1)取5μL每孔化合物工作液,按照实验排布图加入到384孔测试板中,阳性对照加入5μL每孔1X LSD1缓冲液含有2.5%DMSO,空白对照加入5μL每孔1X LSD1缓冲液;
2)LSD1酶原液,冰上解冻,并且在实验过程中酶溶液需要一直置于冰上;
3)酶完全溶解后,用1X LSD1缓冲液将酶原液稀释到12.5ng/μL,即取2.6μL酶原液(4090ng/μL)用848μL 1X LSD1缓冲液稀释;
4)取10μL每孔酶溶液加入到384孔板中,空白对照孔加入10μL每孔1X LSD1缓冲液,酶量为125ng每孔。
5)酶和化合物置于25℃孵育30分钟;
6)将组蛋白H3K4单甲基化肽底物的干粉溶解在500μL水中,放置于冰上备用。
7)化合物和酶结束孵育后向测试板加入10μL每孔底物混合溶液,10μL底物混合溶液包括7.5μL 2X LSD1缓冲液,2.5μL组蛋白H3K4单甲基化肽底物溶液,测试板封膜置于25℃,孵育60分钟。
8)取20μL 10mM Amplex Red和40μL 10U/mL Peroxidase加入1940μL 1X LSD1缓冲液进行检测混合液配制。结束孵育后,取检测混合液25μL每孔加入测试板,测试板置于25℃,孵育5分钟。
9)结束孵育后,马上使用Nivo进行荧光值检测(检测波长:excitation 530nm,emission 580nm)。
数据分析:由原始读值根据以下公式计算得到抑制率百分比,然后利用Prism作图统计并计算化合物的IC 50值。
%抑制率=100-(FI 化合物-FI 空白对照)/(FI 阳性对照-FI 空白对照)×100%
其中空白对照为不含酶对照孔;阳性对照为含有酶,底物和0.5%DMSO对照孔。结果如表20所示。
表20 化合物对LSD1酶活性的抑制作用
Figure PCTCN2022075259-appb-000040
结论:本发明化合物Ⅳ对LSD1抑制活性明显。
实验例3:化合物Ⅳ的细胞活性评价
1.1 kasumi-1细胞活性评价
实验目的:分析化合物Ⅳ对人急性髓系白血病Kasumi-1细胞的增殖抑制作用。
实验材料:RPMI 1640培养基购自依科赛生物科技有限公司,盘尼西林/链霉素抗生素购自HyClone。CellTiter-Glo Luminescent Cell Viability Assay(细胞活率化学发光检测试剂)试剂购自Promega。胎牛血清(FBS),Kasumi-1细胞购自美国模式培养物集存库(ATCC,American type culture collection)。Nivo多标记分析仪(PerkinElmer)。
实验方法:
1)将Kasumi-1细胞种于96孔板中,80μL细胞悬液每孔,其中1×10 4个细胞/mL。细胞板置于二氧化碳培养箱中培养。
2)使用细胞培养液将化合物稀释到50μM,放入化合物板的第1列(1.5μL 10mM母液+300μL细胞培养液)。在第2列到第9列的孔中,加入80μL细胞培养液,从第1列取20μL化合物加入到第2列混合均匀,再从第2列取20μL化合物加入到第3列混合均匀,重复此步骤到第9列。
3)从化合物板取20μL每孔梯度稀释完成的化合物放入细胞培养板的相应位置,此时化合物的最终浓度为10μM至0.128nM。将细胞板放回含5%的二氧化碳培养箱继续孵育6天。
4)细胞培养6天后,取出96孔细胞培养板,加入CTG试剂,50μL/孔,混匀离心,室温孵育15分钟。使用Envision多标记分析仪读数。
数据分析:
1)抑制率计算:
%抑制率=(RFU样品-RFU阴性对照)/(RFU阳性对照-RFU阴性对照)×100%
2)IC 50计算:将抑制率使用软件Prism 8进行IC 50计算,结果如表21所示。
表21 化合物对kasumi-1的增殖抑制作用
Figure PCTCN2022075259-appb-000041
Figure PCTCN2022075259-appb-000042
结论:本发明化合物Ⅳ在Kasumi-1细胞上显示出了显著的抗增殖活性。
1.2 KG-1细胞活性评价
实验目的:分析化合物Ⅳ对人急性髓系白血病KG-1细胞的增殖抑制作用。
实验材料:胎牛血清(FBS)购自依科赛生物科技有限公司,IMDM培养基购自美国模式培养物集存库(ATCC,American type culture collection),青霉素/链霉素双抗购自HyClone。CellTiter-Glo Luminescent Cell Viability Assay(细胞活率化学发光检测试剂)试剂购自Promega。KG-1细胞购自欧洲标准细胞收藏中心(ECACC,European Collection of Authenticated Cell Cultures)。Nivo多标记分析仪(PerkinElmer)。
实验方法:
1)将KG-1细胞种于96孔板中,100μL细胞悬液每孔,其中1×10 4个细胞/mL。细胞板置于二氧化碳培养箱中培养。
2)将配制好的化合物转移至相应的细胞板孔中(细胞板终浓度以10μM为起始,5X递减,9个浓度)。将细胞版置于含5%CO 2的细胞培养箱中37℃培养6天。
3)第6天取出96孔细胞培养板,加入CellTiter-Glo Luminescent Cell Viability Assay试剂,50μL/孔,混匀震板10分钟,室温孵育5分钟。使用Envision Multilabel Plate Reader读细胞板荧光值。
数据分析:
1)抑制率计算:
%抑制率=(RFU样品-RFU阴性对照)/(RFU阳性对照-RFU阴性对照)×100%
2)IC 50计算:将抑制率使用软件GraphPad Prism 9进行IC 50计算,结果如表22所示。
表22 化合物对细胞增殖的抑制作用
Figure PCTCN2022075259-appb-000043
结论:本发明化合物Ⅳ在KG-1细胞上显示出了显著的抗增殖活性。
实验例4:化合物Ⅳ对人急性髓系白血病Kasumi-1细胞在CB-17 SCID小鼠皮下移植瘤模型中的体内药效学研究
4.1实验目的
本实验的目的是评价化合物Ⅳ对人急性髓系白血病Kasumi-1细胞在CB-17 SCID小鼠皮下移植瘤模型中的抑瘤效果。
4.2实验动物
种属:小鼠;品系:CB-17 SCID小鼠;周龄及体重:6-8周龄,体重16-21克;性别:雌性;供应商:上海吉辉实验动物饲养有限公司。
4.3实验方法与步骤
4.3.1细胞培养
人急性髓系白血病Kasumi-1细胞悬浮培养,培养条件为RPMI-1640培养基中加20%胎牛血清,加1%青霉素和链霉素,37℃ 5%CO 2培养。当细胞饱和度为80%-90%时,收取细胞,计数,调整10×10 6个细胞/mL重悬于磷酸盐缓冲溶液PBS。
4.3.2肿瘤细胞接种
0.2mL(10×10 6个)Kasumi-1细胞(加基质胶,体积1:1)皮下接种于每只小鼠的右后背,肿瘤平均体积达到约135mm 3时开始分组给药采用随机分组,开始给药。
4.3.3受试物的配制
实验用溶媒为0.5%甲基纤维素溶液配制:称取2.5g甲基纤维素,溶解于400mL超纯水中,搅拌均匀后用超纯水定容至500mL,于4℃保存。
阿扎胞苷(5-Azacytidine,生产商MedChemExpress,批号28452)制剂配制:称量1mg 5-azacytidine,加入14.2mL PBS,溶解得到澄清溶液,于4℃保存。
化合物Ⅳ制剂配制:称取14.2mg化合物Ⅳ,加入16.026mL 0.5%MC,溶解得到浓度0.45mg/mL澄清溶液,于4℃保存。
4.3.4肿瘤测量和实验指标
实验指标是考察肿瘤生长是否被抑制、延缓或治愈。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×b 2,a和b分别表示肿瘤的长径和短径。
化合物的抑瘤疗效用TGI(%)评价。TGI(%),反映肿瘤生长抑制率。TGI(%)的计算:TGI(%)=【1-(某处理组给药结束时肿瘤平均体积-该处理组开始给药时肿瘤平均体积)/(溶剂对照组治疗结束时肿瘤平均体积-溶剂对照组开始治疗时肿瘤平均体积)】×100%。
统计分析:各实验组与溶剂对照间采用one tailed T Test进行分析,p<0.05认为有显著性差异。
4.4实验结果:基于给药后第21天肿瘤体积计算得出的实验结果见表23。
表23 化合物Ⅳ对人急性髓系白血病Kasumi-1细胞在CB-17 SCID小鼠皮下移植瘤模型中的抑瘤药效评价
Figure PCTCN2022075259-appb-000044
结论:化合物Ⅳ单药治疗组及与阿扎胞苷联用给药均对Kasumi-1移植肿瘤生长有明显抑制作用。化合物Ⅳ与阿扎胞苷联用,较化合物Ⅳ、阿扎胞苷单药具有更显著的抑瘤效果。荷瘤鼠对化合物Ⅳ在所有剂量下都显示出较好的耐受性。
实验例5:化合物Ⅳ对人急性髓系白血病KG-1细胞在BALB/c nude小鼠皮下移植瘤模型中的体内药效学研究
5.1实验目的
本实验的目的是评价化合物Ⅳ对人白血病KG-1细胞在BALB/c nude小鼠皮下移植瘤模型中的抑瘤效果。
5.2实验动物
种属:小鼠;品系:BALB/c nude小鼠;周龄及体重:6-8周龄,体重18-24克;性别:雌性;供应商:上海灵畅生物科技有限公司。
5.3实验方法与步骤
5.3.1细胞培养
人急性髓系白血病KG-1细胞体外单层培养,培养条件为IMDM培养基中加20%胎牛血清,1%青霉素和链霉素,37℃、5%CO 2培养传代。当细胞饱和度为80%-90%时,收取细胞,计数,调整细胞悬液数目为5×10 7个/mL。
5.3.2肿瘤细胞接种
0.2mL(10×10 6个)KG-1细胞(加基质胶,体积1:1)皮下接种于每只小鼠的右后背,肿瘤平均体积达到约106mm 3时采用随机分组,开始给药。
5.3.3受试物的配制
实验用溶媒为0.5%甲基纤维素溶液配制:称取2.5g甲基纤维素,溶解于400mL超纯水中,搅拌均匀后用超纯水定容至500mL,于4℃保存。
阿扎胞苷(5-Azacytidine,生产商MedChemExpress,批号103024)制剂配制:称量1.6mg 5-azacytidine,加入16mL PBS,溶解得到澄清溶液,于4℃保存。
化合物Ⅳ制剂配制:称取5mg化合物Ⅳ,加入4.224mL 0.5%MC,溶解得到浓度0.6mg/mL澄清溶液,于4℃保存。
5.3.4肿瘤测量和实验指标
实验指标是考察肿瘤生长是否被抑制、延缓或治愈。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×b 2,a和b分别表示肿瘤的长径和短径。
化合物的抑瘤疗效用TGI(%)评价。TGI(%),反映肿瘤生长抑制率。TGI(%)的计算:TGI(%)=【1-(某处理组给药结束时肿瘤平均体积-该处理组开始给药时肿瘤平均体积)/(溶剂对照组治疗结束时肿瘤平均体积-溶剂对照组开始治疗时肿瘤平均体积)】×100%。
统计分析:各实验组与溶剂对照间采用Prism 6.02进行分析,p<0.05认为有显著性差异。
5.4实验结果:基于给药后第21天肿瘤体积计算得出的实验结果见表24。
表24 化合物Ⅳ对人急性髓系白血病KG-1细胞在BALB/c nude小鼠皮下移植瘤模型中的抑瘤药效评价
Figure PCTCN2022075259-appb-000045
Figure PCTCN2022075259-appb-000046
结论:受试物化合物Ⅳ单药对人白血病KG-1细胞在BALB/c nude小鼠皮下异种移植肿瘤生长均有抑制作用。化合物Ⅳ与阿扎胞苷联合给药对肿瘤生长具有显著的抑制作用,抑瘤效果明显优于化合物Ⅳ单药和阿扎胞苷单药。荷瘤鼠对化合物Ⅳ所有剂量下都显示出较好的耐受性。
实验例6:化合物Ⅳ对系统性人急性髓系白血病MV-4-11-Luc细胞在NCG小鼠异种移植瘤模型中的药效学评价
6.1实验目的
本实验的目的是评价化合物Ⅳ对人白血病MV-4-11-Luc细胞在NCG小鼠异种系统性移植瘤模型中的抑瘤效果。
6.2实验动物
种属:小鼠;品系:NCG小鼠;周龄及体重:6-7周龄,体重19.1-24.7克;性别:雌性;供应商:江苏集萃药康生物科技有限公司。
6.3实验方法与步骤
6.3.1细胞培养
MV-4-11-Luc细胞培养在含10%胎牛血清的IMDM培养液中。收集指数生长期的MV-4-11-Luc细胞,PBS重悬至适合浓度后用于小鼠尾静脉接种。
6.3.2肿瘤细胞接种
雌性NCG小鼠尾静脉接种1×10 7MV-4-11-Luc细胞。接种后第7天将所有小鼠成像,根据荧光值分组,分组给药当天定义为第0天。
6.3.3受试物的配制
实验用溶媒为0.5%甲基纤维素溶液配制:称取2.5g甲基纤维素,溶解于400mL超纯水中,搅拌均匀后用超纯水定容至500mL,于4℃保存。
阿扎胞苷(5-Azacytidine,生产商MedChemExpress,批号103024)制剂配制:称取0.675mg 5-Azacytidine,加入9mL的生理盐水,涡旋,超声振荡得到澄清溶液,于4℃保存。
化合物Ⅳ制剂配制:称取4.8mg化合物Ⅳ,加入24.219mL的0.5%甲基纤维素(4000cps),涡旋,超声振荡得到澄清溶液,于4℃保存。
6.3.4肿瘤测量和实验指标
实验指标是考察肿瘤生长是否被抑制、延缓或治愈。本实验中通过小鼠活体生物发光体内成像进行监测肿瘤生长,成像频率为分组给药后每周两次,所使用成像系统为IVIS Lumina III小鼠活体成像仪(PerkinElmer,USA)。具体的实验步骤如下:
1)成像小鼠通过颈部皮下注射D-Luciferin成像底物(PerkinElmer,XenoLight D-Luciferin(K+Salt),货号122799),注射剂量为150mg/kg,注射体积为5μl/g;
2)底物注射10分钟后,成像小鼠置于异氟烷麻醉箱中进行麻醉;
3)待成像小鼠进入麻醉状态后,将小鼠转移至成像仪中,通过将小鼠口鼻置于麻醉系统套管中维持动物 在成像过程中处于麻醉状态,小鼠按照耳号从小到大,从左到右放置,腹部朝上,小鼠尾巴置于黑色遮光套管中;
4)IVIS Lumina III成像软件中选择生物发光成像,曝光时间设置为自动,进行小鼠成像.
5)成像结束后,将小鼠转移至饲养笼盒内,待全部动物确认苏醒后放回小鼠饲养架中。
化合物的抑瘤疗效用TGI(%)评价,反映肿瘤生长抑制率。T/C%=T RTV/C RTV×100%;TGI%=(1-T/C)×100%。本实验中TV用荧光信号值代替。
统计分析:p<0.05认为有显著性差异。
6.4实验结果:基于给药后第21天肿瘤荧光强度计算得出的实验结果见表25。
表25 化合物Ⅳ对人急性髓系白血病MV-4-11-Luc细胞在NCG小鼠系统移植瘤模型中的抑瘤药效评价
Figure PCTCN2022075259-appb-000047
结论:阿扎胞苷在0.75mg/kg剂量下对人急性髓系白血病MV-4-11-Luc细胞在NCG小鼠系统移植瘤模型中无显著性的抑瘤效果。化合物Ⅳ在0.5mg/kg及1.0mg/kg剂量下对对人急性髓系白血病MV-4-11-Luc细胞在NCG小鼠系统移植瘤模型均有显著性的抑瘤效果。化合物Ⅳ(0.50mg/kg)与阿扎胞苷(0.75mg/kg)联合、化合物Ⅳ(1.0mg/kg)与阿扎胞苷(0.75mg/kg)联合后,均有显著性的抑瘤效果。

Claims (17)

  1. 化合物Ⅲ:
    Figure PCTCN2022075259-appb-100001
  2. 化合物Ⅳ’:
    Figure PCTCN2022075259-appb-100002
    其中,n为0-2,优选为1.5-2。
  3. 根据权利要求2所述的化合物Ⅳ’,其具有化合物Ⅳ的结构:
    Figure PCTCN2022075259-appb-100003
  4. 化合物Ⅳ的A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.42±0.20°、16.90±0.20°和19.99±0.20°;
    Figure PCTCN2022075259-appb-100004
  5. 根据权利要求4所述的A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.42±0.20°、8.46±0.20°、15.88±0.20°、16.90±0.20°、17.80±0.20°、18.77±0.20°、19.99±0.20°和22.65±0.20°;
    或者,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.42±0.20°、8.46±0.20°、15.88±0.20°、16.90±0.20°、17.80±0.20°、18.27±0.20°、18.77±0.20°、19.99±0.20°、21.98±0.20°和22.65±0.20°;
    或者,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.42±0.20°、8.46±0.20°、10.79±0.20°、11.69±0.20°、13.66±0.20°、14.78±0.20°、15.88±0.20°、16.40±0.20°、16.90±0.20°、17.80±0.20°、18.27±0.20°、18.77±0.20°、19.57±0.20°、19.99±0.20°、21.23±0.20°、21.64±0.20°、21.98±0.20°、22.30±0.20°、22.65±0.20°、23.15±0.20°和23.44±0.20°;
    或者,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.42°、8.46°、10.79°、11.69°、13.66°、14.78°、15.88°、16.40°、16.90°、17.80°、18.27°、18.77°、19.57°、19.99°、21.23°、21.64°、21.98°、22.30°、22.65°、23.15°和23.44°。
  6. 根据权利要求4所述的A晶型,其差示扫描量热曲线在215.4±5℃有一个吸热峰的起始点;
    或者,其热重分析曲线在180±5℃时失重达0.89%。
  7. 根据权利要求4所述的A晶型,其XRPD图谱基本如图1所示;
    或者,其DSC图谱如图2所示;
    或者,其TGA图谱如图3所示。
  8. 化合物Ⅴ:
    Figure PCTCN2022075259-appb-100005
  9. 化合物Ⅴ的B晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:16.33±0.20°、18.98±0.20°和22.27±0.20°;
    Figure PCTCN2022075259-appb-100006
  10. 根据权利要求9所述的B晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.08±0.20°、10.51±0.20°、14.54±0.20°、16.33±0.20°、17.37±0.20°、17.91±0.20°、18.98±0.20°和22.27±0.20°;
    或者,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.08±0.20°、10.51±0.20°、14.54±0.20°、16.33±0.20°、17.37±0.20°、17.91±0.20°、18.98±0.20°、19.56±0.20°、21.30±0.20°、22.27±0.20°、23.58±0.20°和26.66±0.20°;
    或者,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.08±0.20°、7.40±0.20°、10.51±0.20°、12.27±0.20°、12.47±0.20°、14.15±0.20°、14.54±0.20°、16.33±0.20°、17.37±0.20°、17.91±0.20°、18.60±0.20°、18.98±0.20°、19.20±0.20°、19.56±0.20°、20.13±0.20°、21.06±0.20°、21.30±0.20°、22.27±0.20°、22.74±0.20°、23.58±0.20°、24.68±0.20°、25.16±0.20°、25.84±0.20°、26.66±0.20°、27.77±0.20°、29.03±0.20°、30.18±0.20°、30.70±0.20°、31.54±0.20°、32.18±0.20°、32.96±0.20°、34.03±0.20°、34.53±0.20°、36.26±0.20°和37.41±0.20°;
    或者,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.08°、7.40°、10.51°、12.27°、12.47°、14.15°、14.54°、16.33°、17.37°、17.91°、18.60°、18.98°、19.20°、19.56°、20.13°、21.06°、21.30°、22.27°、22.74°、23.58°、24.68°、25.16°、25.84°、26.66°、27.77°、29.03°、30.18°、30.70°、31.54°、32.18°、32.96°、34.03°、34.53°、36.26°和37.41°。
  11. 根据权利要求9所述的B晶型,其差示扫描量热曲线在127.4±5℃有一个吸热峰的峰值;
    或者,其热重分析曲线在150±5℃时失重达8.84%。
  12. 根据权利要求9所述的B晶型,其XRPD基本图谱如图4所示;
    或者,其DSC图谱如图5所示;
    或者,其TGA图谱如图6所示。
  13. 化合物Ⅱ的制备方法,
    Figure PCTCN2022075259-appb-100007
    其包含如下步骤:
    Figure PCTCN2022075259-appb-100008
    其中,
    有机溶剂为甲醇、乙醇、异丙醇、乙腈或乙酸异丙酯;
    手性酸为
    Figure PCTCN2022075259-appb-100009
  14. 根据权利要求13所述的制备方法,其包含如下步骤:
    Figure PCTCN2022075259-appb-100010
    其中,
    有机溶剂为甲醇、乙醇、异丙醇、乙腈或乙酸异丙酯;
    手性酸为
    Figure PCTCN2022075259-appb-100011
    溶剂1为饱和碳酸氢钠溶液;
    萃取剂为二氯甲烷。
  15. 根据权利要求13或14所述的制备方法,其中有机溶剂为乙腈;
    或者,
    Figure PCTCN2022075259-appb-100012
    与化合物Ⅰ的摩尔比为0.5-1:1;优选为1:1;
    或者,有机溶剂与化合物Ⅰ的体积质量比V:m为5-20(mL/g);
    或者,化合物Ⅰ制备化合物Ⅰ-A的反应温度为25-85℃。
  16. 权利要求1所述的化合物Ⅲ、权利要求2所述的化合物IV’、权利要求3所述的化合物Ⅳ、权利要求8所述的化合物Ⅴ、权利要求4-7任意一项所述的A晶型以及权利要求9-12任意一项所述的B晶型在制备治疗LSD1相关疾病的药物中的应用。
  17. 根据权利要求16所述的应用,所述疾病为血液肿瘤、小细胞肺癌、鳞状非小细胞肺癌、乳腺癌、前列腺癌、肝癌、胰腺癌、胶质瘤或尤文氏肉瘤,优选为人急性髓系白血病。
PCT/CN2022/075259 2021-02-09 2022-01-30 一种氧氮杂螺环化合物、其盐型及其晶型 WO2022171044A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280003890.4A CN115485282A (zh) 2021-02-09 2022-01-30 一种氧氮杂螺环化合物、其盐型及其晶型

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110179260 2021-02-09
CN202110179260.X 2021-02-09

Publications (1)

Publication Number Publication Date
WO2022171044A1 true WO2022171044A1 (zh) 2022-08-18

Family

ID=82837441

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/075259 WO2022171044A1 (zh) 2021-02-09 2022-01-30 一种氧氮杂螺环化合物、其盐型及其晶型

Country Status (2)

Country Link
CN (1) CN115485282A (zh)
WO (1) WO2022171044A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023217758A1 (en) 2022-05-09 2023-11-16 Oryzon Genomics, S.A. Methods of treating malignant peripheral nerve sheath tumor (mpnst) using lsd1 inhibitors
WO2023217784A1 (en) 2022-05-09 2023-11-16 Oryzon Genomics, S.A. Methods of treating nf1-mutant tumors using lsd1 inhibitors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103857393A (zh) * 2011-03-25 2014-06-11 葛兰素史密斯克莱知识产权(第2号)有限公司 环丙基胺作为lsd1抑制剂
CN104203914A (zh) * 2011-10-20 2014-12-10 奥瑞泽恩基因组学股份有限公司 作为lsd1抑制剂的(杂)芳基环丙胺化合物
CN109153636A (zh) * 2016-05-09 2019-01-04 朱比连特比利斯有限公司 作为双重lsd1/hdac抑制剂的环丙基-酰胺化合物
WO2020052649A1 (zh) * 2018-09-13 2020-03-19 南京明德新药研发有限公司 作为lsd1抑制剂的环丙胺类化合物及其应用

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103857393A (zh) * 2011-03-25 2014-06-11 葛兰素史密斯克莱知识产权(第2号)有限公司 环丙基胺作为lsd1抑制剂
CN104203914A (zh) * 2011-10-20 2014-12-10 奥瑞泽恩基因组学股份有限公司 作为lsd1抑制剂的(杂)芳基环丙胺化合物
CN109153636A (zh) * 2016-05-09 2019-01-04 朱比连特比利斯有限公司 作为双重lsd1/hdac抑制剂的环丙基-酰胺化合物
WO2020052649A1 (zh) * 2018-09-13 2020-03-19 南京明德新药研发有限公司 作为lsd1抑制剂的环丙胺类化合物及其应用

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023217758A1 (en) 2022-05-09 2023-11-16 Oryzon Genomics, S.A. Methods of treating malignant peripheral nerve sheath tumor (mpnst) using lsd1 inhibitors
WO2023217784A1 (en) 2022-05-09 2023-11-16 Oryzon Genomics, S.A. Methods of treating nf1-mutant tumors using lsd1 inhibitors

Also Published As

Publication number Publication date
CN115485282A (zh) 2022-12-16

Similar Documents

Publication Publication Date Title
WO2022171044A1 (zh) 一种氧氮杂螺环化合物、其盐型及其晶型
EP3825309B1 (en) Salt of lsd1 inhibitor and a polymorph thereof
US11680061B2 (en) Crystal forms C and E of pyrazin-2(1H)-one compound and preparation method therefor
TWI812223B (zh) 雜環取代的嘌呤酮衍生物的鹽型及晶型
CN108602815A (zh) 吡啶并[1,2-a]嘧啶酮类似物、其晶型、其中间体及其制备方法
CN114728973A (zh) 一种核蛋白抑制剂的晶型及其应用
WO2021218912A1 (zh) 含苯基并内磺酰胺的化合物
WO2020147838A1 (zh) 一种egfr抑制剂的盐、晶型及其制备方法
US11465986B2 (en) Crystal form of c-MET inhibitor and salt form thereof and preparation method therefor
WO2020164603A1 (zh) 固体形式的fgfr抑制剂化合物及其制备方法
EP3896063A1 (en) Salt of syk inhibitor and crystalline form thereof
SG183869A1 (en) Agomelatine hydrobromide hydrate and preparation thereof
RU2794977C2 (ru) Соль ингибитора lsd1 и её полиморфная форма
US20230045991A1 (en) Salt form and crystal form of mutant idh1 inhibitor and preparation method therefor
WO2019057142A1 (zh) 一种内磺酰胺化合物结晶
WO2022237682A1 (zh) 吡咯并三嗪类化合物的盐型、其晶型及其制备方法
WO2021139794A1 (zh) 一种吡咯烷基脲衍生物的晶型及其应用
CN109422798A (zh) 一种苯基丙酰胺类衍生物的游离碱晶型及其制备方法
WO2024027825A1 (zh) 一种cdk抑制剂及其磷酸盐的多晶型
US20230365596A1 (en) Crystal forms of pyridopyrazole compounds and preparation method therefor
CN114106042B (zh) 一种氘代化合物及其制备方法和应用
WO2020224585A1 (zh) 一种mTORC1/2双激酶活性抑制剂的盐型、晶型及其制备方法
WO2019141245A1 (zh) 一种三并环化合物的晶型、盐型及其制备方法
TW202408510A (zh) Cdk抑制劑及其磷酸鹽的多晶型、其製備方法、包含其的醫藥組合物及其用途
CN117645603A (zh) p300溴结构域抑制剂及其药用组合物及其应用

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: 22752205

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22752205

Country of ref document: EP

Kind code of ref document: A1