WO2022143671A1 - 吗啉取代的苯并嘧啶类化合物的晶型及其制备方法 - Google Patents

吗啉取代的苯并嘧啶类化合物的晶型及其制备方法 Download PDF

Info

Publication number
WO2022143671A1
WO2022143671A1 PCT/CN2021/142101 CN2021142101W WO2022143671A1 WO 2022143671 A1 WO2022143671 A1 WO 2022143671A1 CN 2021142101 W CN2021142101 W CN 2021142101W WO 2022143671 A1 WO2022143671 A1 WO 2022143671A1
Authority
WO
WIPO (PCT)
Prior art keywords
crystal form
compound
angles
ray powder
formula
Prior art date
Application number
PCT/CN2021/142101
Other languages
English (en)
French (fr)
Inventor
陈新海
陈兆国
于衍新
胡伯羽
夏尚华
王永鑫
Original Assignee
南京明德新药研发有限公司
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 南京明德新药研发有限公司 filed Critical 南京明德新药研发有限公司
Priority to CN202180087676.7A priority Critical patent/CN116670128A/zh
Publication of WO2022143671A1 publication Critical patent/WO2022143671A1/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
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings

Definitions

  • the invention relates to a crystal form of a morpholine-substituted benzopyrimidine compound and a preparation method thereof.
  • DNA double-strand breaks can cause loss of genetic material, genetic recombination, and then cancer or cell death.
  • DDR DNA damage response
  • DNA double-strand break repair mainly includes two types: homologous end linking (HR) repair and non-homologous end linking (NHEJ) repair.
  • DDR initial damage factors such as MRN will detect and recognize the damage site, recruit phosphatidylinositol kinase family members (ATM, ATR, DNA-PK), phosphorylate H2AX to promote the formation of ⁇ H2AX, and recruit related signaling proteins (such as 53BP1, Chk1, Chk2, BRCA1, NBS1) and other transduction damage signals, so that cells enter a cell cycle arrest state and recruit related repair proteins to repair damaged DNA.
  • ATM phosphatidylinositol kinase family members
  • ATR phosphatidylinositol kinase family members
  • DNA-PK phosphorylate H2AX to promote the formation of ⁇ H2AX
  • related signaling proteins such as 53BP1, Chk1, Chk2, BRCA1, NBS1
  • DNA-PK is mainly aimed at non-homologous end double-strand breaks. It consists of six core factors: KU70, KU80, DNA-PKcs, CRCC4, ligase IV and Artemis.
  • KU molecules are specifically linked to the double-strand damage through a pre-formed channel, respectively It binds to the end of the DNA chain, and then slides along the DNA chain for a distance in an ATP-dependent manner to form a KU-DNA complex, which attracts DNA-PKcs to the damaged site, binds with it and activates the kinase activity, and then phosphorylates to participate in A series of proteins that repair and damage signaling to complete the repair.
  • the present invention aims to discover a DNA-PK small molecule inhibitor, which can inhibit DNA-PK activity through combined use with radiotherapy and chemotherapy drugs, thereby greatly reducing tumor DNA repair and inducing cells to enter apoptosis process. To a large extent, it can overcome the problem of resistance to radiotherapy and chemotherapy, and enhance the inhibitory effect on various tumors such as small cell lung cancer, head and neck cancer, colorectal cancer, and pancreatic cancer. Such compounds have good activity and show excellent effects and functions, and have broad prospects.
  • the present invention provides a compound represented by formula (I),
  • n is selected from 0-2, preferably 0-1, more preferably 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4 or 1.5, most preferably 0.5.
  • the present invention provides crystal form A of the compound represented by formula (I), characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 11.22 ⁇ 0.20°, 24.28 ⁇ 0.20°, 27.07 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form A has characteristic diffraction peaks at the following 2 ⁇ angles: 11.22 ⁇ 0.20°, 13.59 ⁇ 0.20°, 15.73 ⁇ 0.20°, 17.25 ⁇ 0.20°, 21.68 ⁇ 0.20 °, 24.28 ⁇ 0.20°, 26.27 ⁇ 0.20°, 27.07 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above-mentioned A crystal form has characteristic diffraction peaks at the following 2 ⁇ angles: 5.26 ⁇ 0.20°, 11.22 ⁇ 0.20°, 13.59 ⁇ 0.20°, 15.73 ⁇ 0.20°, 17.25 ⁇ 0.20 °, 18.55 ⁇ 0.20°, 21.68 ⁇ 0.20°, 23.47 ⁇ 0.20°, 24.28 ⁇ 0.20°, 25.02 ⁇ 0.20°, 26.27 ⁇ 0.20°, 27.07 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above-mentioned Form A has characteristic diffraction peaks at the following 2 ⁇ angles: 11.22 ⁇ 0.20°, 24.28 ⁇ 0.20°, and/or 27.07 ⁇ 0.20°, and/or 13.59 ⁇ 0.20°, and/or 15.73 ⁇ 0.20°, and/or 17.25 ⁇ 0.20°, and/or 21.68 ⁇ 0.20°, and/or 26.27 ⁇ 0.20°, and/or 5.26 ⁇ 0.20°, and/or 18.55 ⁇ 0.20° , and/or 23.47 ⁇ 0.20°, and/or 25.02 ⁇ 0.20°, and/or 7.60 ⁇ 0.20°, and/or 8.24 ⁇ 0.20°, and/or 10.55 ⁇ 0.20°, and/or 14.97 ⁇ 0.20°, and /or 16.59 ⁇ 0.20°, and/or 19.71 ⁇ 0.20°, and/or 23.80 ⁇ 0.20°, and/or 27.43 ⁇ 0.20°, and/or 28.37
  • the X-ray powder diffraction pattern of the above-mentioned A crystal form has characteristic diffraction peaks at the following 2 ⁇ angles: 5.26°, 7.60°, 8.24°, 10.55°, 11.22°, 13.59°, 14.97°, 15.73° , 16.59°, 17.25°, 18.55°, 19.71°, 21.68°, 23.47°, 23.80°, 24.28°, 25.02°, 26.27°, 27.07°, 27.43°, 28.37°, 29.21°, 30.90°, 34.44°, 36.68 °, 37.53°.
  • the XRPD pattern of the above-mentioned crystal form A is basically as shown in FIG. 1 .
  • the differential scanning calorimetry curve has an endothermic peak starting point at 144.2 ⁇ 3.0°C.
  • the DSC spectrum of the above-mentioned crystal form A is shown in FIG. 2 .
  • the above-mentioned crystal form A has a weight loss of 2.19% at 170.0 ⁇ 3.0°C in its thermogravimetric analysis curve.
  • the present invention provides the B crystal form of the compound represented by formula (I), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.55 ⁇ 0.20°, 11.42 ⁇ 0.20°, 16.30 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 4.55 ⁇ 0.20°, 9.09 ⁇ 0.20°, 11.42 ⁇ 0.20°, 15.24 ⁇ 0.20°, 16.30 ⁇ 0.20 °, 18.65 ⁇ 0.20°, 23.94 ⁇ 0.20°, 25.18 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 4.55 ⁇ 0.20°, 9.09 ⁇ 0.20°, 11.42 ⁇ 0.20°, 12.68 ⁇ 0.20°, 15.24 ⁇ 0.20 °, 16.30 ⁇ 0.20°, 18.65 ⁇ 0.20°, 22.97 ⁇ 0.20°, 23.94 ⁇ 0.20°, 25.18 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above-mentioned Form A has characteristic diffraction peaks at the following 2 ⁇ angles: 4.55 ⁇ 0.20°, 11.42 ⁇ 0.20°, and/or 16.30 ⁇ 0.20°, and/or 9.09 ⁇ 0.20° 0.20°, and/or 15.24 ⁇ 0.20°, and/or 18.65 ⁇ 0.20°, and/or 23.94 ⁇ 0.20°, and/or 25.18 ⁇ 0.20°, and/or 12.68 ⁇ 0.20°, and/or 17.83 ⁇ 0.20° , and/or 22.97 ⁇ 0.20°, and/or 27.14 ⁇ 0.20°.
  • the above-mentioned crystal form B its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 4.55°, 9.09°, 11.42°, 12.68°, 15.24°, 16.30°, 17.83°, 18.65° °, 22.97°, 23.94°, 25.18°, 27.14°.
  • the XRPD pattern of the above-mentioned crystal form B is basically as shown in FIG. 4 .
  • the above-mentioned crystal form B has an onset of an endothermic peak at 128.2 ⁇ 3.0°C and a peak of an endothermic peak at 198.2 ⁇ 3.0°C in its differential scanning calorimetry curve.
  • the DSC spectrum of the above-mentioned crystal form B is shown in FIG. 5 .
  • the above-mentioned crystal form B has a weight loss of 2.35% at 170.0 ⁇ 3.0°C in its thermogravimetric analysis curve.
  • the above-mentioned crystal form B its TGA spectrum is shown in FIG. 6 .
  • the present invention provides the C crystal form of the compound represented by formula (II), which is characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 14.76 ⁇ 0.20°, 18.03 ⁇ 0.20°, 23.19 ⁇ 0.20°;
  • the X-ray powder diffraction pattern of the above crystal form C has characteristic diffraction peaks at the following 2 ⁇ angles: 10.44 ⁇ 0.20°, 12.22 ⁇ 0.20°, 14.76 ⁇ 0.20°, 18.03 ⁇ 0.20°, 19.09 ⁇ 0.20 °, 20.73 ⁇ 0.20°, 23.19 ⁇ 0.20°, 28.66 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above crystal form C has characteristic diffraction peaks at the following 2 ⁇ angles: 10.44 ⁇ 0.20°, 12.22 ⁇ 0.20°, 14.76 ⁇ 0.20°, 18.03 ⁇ 0.20°, 19.09 ⁇ 0.20 °, 20.73 ⁇ 0.20°, 23.19 ⁇ 0.20°, 25.67 ⁇ 0.20°, 27.43 ⁇ 0.20°, 28.66 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above-mentioned Form A has characteristic diffraction peaks at the following 2 ⁇ angles: 14.76 ⁇ 0.20°, 18.03 ⁇ 0.20°, and/or 23.19 ⁇ 0.20°, and/or 10.44 ⁇ 0.20°, and/or 12.22 ⁇ 0.20°, and/or 19.09 ⁇ 0.20°, and/or 20.73 ⁇ 0.20°, and/or 28.66 ⁇ 0.20°, and/or 25.67 ⁇ 0.20°, and/or 27.43 ⁇ 0.20° , and/or 7.08 ⁇ 0.20°, and/or 8.71 ⁇ 0.20°, and/or 13.20 ⁇ 0.20°, and/or 23.37 ⁇ 0.20°, and/or 24.24 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above crystal form C has characteristic diffraction peaks at the following 2 ⁇ angles: 7.08°, 8.71°, 10.44°, 12.22°, 13.20°, 14.76°, 18.03°, 19.09° , 20.73°, 23.19°, 23.37°, 24.24°, 25.67°, 27.43°, 28.66°.
  • the XRPD pattern of the above-mentioned crystal form C is basically as shown in FIG. 7 .
  • the differential scanning calorimetry curve of the differential scanning calorimetry curve has an onset of an endothermic peak at 197.8 ⁇ 3.0°C.
  • the DSC spectrum of the above-mentioned crystal form C is shown in FIG. 8 .
  • the present invention also provides the following experimental methods:
  • test compound was mixed with 10% DMSO/50% PEG400/40% water, vortexed and sonicated to prepare a nearly clear solution of 0.2 mg/mL or 0.4 mg/mL, which was filtered through a microporous membrane for use.
  • Balb/c female mice of 18 to 20 grams were selected and administered the candidate compound solution by intravenous injection at a dose of 1 or 2 mg/kg.
  • the test compound was mixed with 10% DMSO/50% PEG400/40% water, vortexed and sonicated to prepare a 0.2 mg/mL or 1 mg/mL approximately clear solution, which was filtered through a microporous membrane for use.
  • mice Balb/c female mice of 18 to 20 grams were selected and the candidate compound solution was orally administered at a dose of 2 or 10 mg/kg.
  • Whole blood was collected for a certain period of time, plasma was prepared, drug concentration was analyzed by LC-MS/MS method, and pharmacokinetic parameters were calculated by Phoenix WinNonlin software (Pharsight, USA).
  • 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.
  • SXRD single crystal X-ray diffractometry
  • the cultivated single crystal is collected by Bruker D8venture diffractometer
  • the light source is CuK ⁇ radiation
  • the scanning method is as follows: After scanning and collecting relevant data, the crystal structure was further analyzed by the direct method (Shelxs97), and the absolute configuration could be confirmed.
  • the solvent used in the present invention is commercially available.
  • DCM stands for dichloromethane
  • DMF stands for N,N-dimethylformamide
  • DMSO stands for dimethyl sulfoxide
  • EtOH stands for ethanol
  • MeOH stands for methanol
  • TFA trifluoroacetic acid
  • ATP stands for Adenosine triphosphate
  • HEPES 4-hydroxyethylpiperazineethanesulfonic acid
  • MgCl 2 stands for magnesium dichloride
  • Pd(PPh 3 ) 2 Cl 2 stands for bistriphenylphosphine palladium dichloride.
  • the crystal form of the compound of the present invention has good stability and is easy to prepare medicine; the compound crystal form of the present invention has excellent DNA-PK kinase inhibitory activity.
  • X-ray powder diffraction X-ray powder diffractometer, XRPD
  • Test Method Approximately 10 mg of sample was used for XRPD detection.
  • Light tube voltage 45kV
  • light tube current 40mA
  • DSC Differential Scanning Calorimeter
  • Test method Take a sample (about 1-5mg) and place it in a DSC aluminum pan for testing. Under the condition of 50mL/min N2 , at a heating rate of 10°C/min, heat the sample from 25°C (room temperature) to before the sample decomposes. .
  • Test method Take a sample (about 1-5mg) and place it in a TGA aluminum pan for testing. Under the condition of 10mL/min N2 , at a heating rate of 10°C/min, heat the sample from room temperature to 350°C.
  • the present invention 's dynamic moisture adsorption (Dynamic Vapor Sorption, DVS)
  • Test conditions take a sample (10-20 mg) and place it in the DVS sample tray for testing.
  • RH (%) test step 10% (0%RH-90%RH, 90%RH-0%RH)
  • the hygroscopicity evaluation is classified as follows:
  • 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% No or almost no hygroscopicity ⁇ W% ⁇ 0.2%
  • ⁇ W% represents the hygroscopic weight gain of the test product at 25 ⁇ 1°C and 80 ⁇ 2%RH.
  • Fig. 1 is the XRPD spectrum of the Cu-K ⁇ radiation of compound A of formula (I);
  • Fig. 2 is the DSC spectrogram of formula (I) compound A crystal form
  • Fig. 3 is the TGA spectrum of formula (I) compound A crystal form
  • Fig. 4 is the XRPD spectrum of the Cu-K ⁇ radiation of compound B of formula (I);
  • Fig. 5 is the DSC spectrogram of formula (I) compound B crystal form
  • Fig. 6 is the TGA spectrum of formula (I) compound B crystal form
  • Fig. 7 is the XRPD spectrum of the Cu-K ⁇ radiation of compound C of formula (II);
  • Fig. 8 is the DSC spectrogram of the crystal form of compound C of formula (II);
  • Figure 9 is the DVS spectrum of the crystalline form of compound A of formula (I).
  • the filter cake was washed three times with 3 L of xylene, and the filtrate was collected.
  • the above-mentioned filtrate containing intermediate 3 was slowly added to the reactor, respectively, cesium carbonate (4.31kg, 13.22mol), compound 4 (858g, 9.86mol), 4,5-bis(diphenylphosphorus)-9,9- Dimethylxanthene (101.60 g, 0.18 mol). It was replaced with nitrogen for 3 times, then palladium acetate (19.81 g, 0.09 mol) was added, and it was continued to be replaced with nitrogen for 3 times, and then the temperature was raised to 115-125° C. and reacted for 16 hours. After the reaction was completed, the temperature of the reactor was lowered to 25° C. under stirring, and then filtered. The filter cake was washed three times with 3 L of xylene, and the filtrate was collected. The compound 5 filtrate 25L containing the theoretical value of 3.31kg was directly put into the next step.
  • the mixture in the reaction kettle was transferred to a 50L liquid separator, stirred, left standing, and liquid-separated extraction was performed.
  • the organic phase was collected, the aqueous phase was extracted three times with 5 L of dichloromethane, the organic phases were combined, and transferred to a 50 L rotary evaporator for concentration under reduced pressure.
  • the solid was collected and dried in a vacuum oven to obtain compound 7.
  • the crude product was dispersed with 13 liters of ethyl acetate and 6.5 liters of water. The liquids were separated, and the organic phase was further washed with 6.5 liters of water. The organic phase was spun dry. The obtained crude product was purified by column chromatography to obtain compound 9.
  • the reaction solution was slowly added to a mixture of 4.1 kg of sodium carbonate in 27 kg of ice-water and a solid appeared. Filter, transfer the filter cake to the kettle, add 30 liters of water, stir at room temperature for 1 hour (wash salt), filter, transfer the filter cake to the kettle, add 25 liters of tert-butyl methyl ether, stir at room temperature for 0.3 hours, and filter. The filter cake was transferred to the kettle again, 15 liters of tert-butyl methyl ether was added, the mixture was stirred at room temperature for 0.3 hours, and filtered. Dry in a vacuum oven ( ⁇ 0.09MPa, 50°C, 15 hours) to obtain compound 10.
  • a 3L three-necked round-bottomed flask containing a magnetic stirrer was fixed on the magnetic stirrer, an internal temperature thermometer and an oxygen bag connected with a tee were fixed, and the air tightness was checked.
  • 1.6 L of acetonitrile was added to the reaction flask and stirring was initiated.
  • Compound 15 (199.8 g, 0.38 mol) and potassium carbonate (124.8 g, 0.91 mol) were added.
  • the air in the reaction flask was replaced with oxygen to keep the reaction in an oxygen atmosphere.
  • the temperature was maintained at 20 ⁇ 2°C for 51.5 hours. When the reaction is complete, stop the reaction.
  • the organic phase was concentrated under reduced pressure ( ⁇ 0.03MPa, 50°C), and the obtained solid was further dried in a vacuum oven ( ⁇ 0.01MPa, 50°C, 13 hours) to obtain a crude product (970.3 g).
  • methanol (15.0L) into the 50L reactor, start stirring, add crude product (968g), heat up to 64°C, keep stirring for 2 hours, lower the temperature to 25°C, filter, collect the filter cake and vacuum dry ( ⁇ 0.01 MPa, 55°C, 20 hours).
  • the obtained crude product (832 g) was added to a 50 L reaction kettle filled with dichloromethane (17 L), and stirring was started.
  • the crystal form of compound A of formula (I) was placed under the conditions of high temperature (60° C., closed mouth) and high humidity (92.5% RH, wrapped with parafilm and tied with 5 small holes) for 6 days and 11 days respectively.
  • ICH conditions The total illuminance of visible light reaches 1.2E+06Lux ⁇ hrs, and the total illuminance of ultraviolet light reaches 200W ⁇ hrs/m 2 ) closed and placed under visible light and ultraviolet light (the samples of the shading control group were placed at the same time and wrapped with tin foil), at 60°C/75% Placed for 1, 2 months under the condition of RH (wrapped with parafilm and tied with 5 small holes), and placed under the condition of 40°C/75% RH (wrapped with 5 small holes with parafilm) for 1, 2, and 3 months.
  • the samples were tested for chemical purity (HPLC area purity), relative content (relative to the 0-day sample stored at -20°C) and crystal form at each sampling point to determine the physical/chemical stability of the samples.
  • relative chromatographic purity purity of stability sample/purity of initial sample ⁇ 100%; content change relative to the content of initial sample, mass of initial sample/peak area of initial sample* peak area of stable sample/mass of stable sample; Peaks with a peak area less than 0.05% were not integrated.
  • DNA-PK (h) was incubated in assay buffer containing 50 nM GST-cMyc-p53, test compound and Mg/ATP (concentration as required). The reaction was initiated by the addition of a Mg/ATP mixture. After 30 minutes of incubation at room temperature, the reaction was stopped by the addition of stop solution containing EDTA. Finally, detection buffer (containing labeled anti-GST monoclonal antibody and europium-labeled anti-phospho-Ser15 antibody against phosphorylated p53) was added.
  • HTRF homogeneous time-resolved fluorescence

Landscapes

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

Abstract

提供了一种吗啉取代的苯并嘧啶类化合物的晶型及其制备方法。

Description

吗啉取代的苯并嘧啶类化合物的晶型及其制备方法
本申请主张如下优先权
CN202011596979.5,申请日:2020年12月28日。
技术领域
本发明涉及一种吗啉取代的苯并嘧啶类化合物的晶型及其制备方法。
背景技术
DNA双链断裂(DSBs)作为一种严重的DNA损伤,会造成遗传物质的丢失、基因重组,从而导致癌症或细胞死亡。生物体为维持基因稳定性与细胞活性进化出了DNA损伤应答(DDR)机制进行损伤的检测、信号传导以及损伤修复。DNA双链断裂修复主要包括两种:同源末端链接(HR)修复和非同源末端链接(NHEJ)修复。DDR初期损伤因子如MRN等会检测识别损伤位点,募集膦酯酰肌醇激酶家族成员(ATM、ATR、DNA-PK),磷酸化H2AX促进γH2AX形成,募集相关信号蛋白(如53BP1、Chk1、Chk2、BRCA1、NBS1)等传导损伤信号,使细胞进入细胞周期阻滞状态并募集相关修复蛋白,使损伤DNA修复。
DNA-PK作为DNA损伤修复的重要成员,主要针对非同源末端双链的断裂。由六个核心因子组成:KU70、KU80、DNA-PKcs、CRCC4、连接酶IV和Artemis,DNA双链损伤修复时,KU分子通过一个预先形成的通道特异性地连接到双链损伤处,分别识别并结合DNA链末端,然后以ATP依赖的方式沿DNA链分别向两端滑动一段距离,形成KU-DNA复合物,吸引DNA-PKcs到损伤部位,与之结合并激活激酶活性,进而磷酸化参与修复及损伤信号传导的一系列蛋白,完成修复。
目前,通过放疗和化疗(拓扑异构酶II、博来霉素、阿霉素、依托泊苷)等方式来诱导DNA损伤来是控制肿瘤的生长主要手段之一。但研究表明,经过放化疗后的肿瘤组织中DNA-PK高表达,不断修复放化疗损伤的肿瘤细胞,成为放化疗耐药的主要原因之一。
本发明旨在发现一种DNA-PK小分子抑制剂,通过与放化疗药物联用,抑制DNA-PK活性,从而大大减少肿瘤DNA修复,诱导细胞进入凋亡程序。可以在很大程度上克服放化疗耐药问题,增强对小细胞肺癌、头颈癌、结直肠癌、胰腺癌等各种肿瘤的抑制作用。此类化合物具有良好的活性,并表现出了优异的效果和作用,具有广阔的前景。
发明内容
本发明提供了式(I)所示化合物,
Figure PCTCN2021142101-appb-000001
其中,n选自0~2,优选为0~1,更优选为0.5、0.6、0.7、0.8、0.9、1、1.1、1.2、1.3、1.4或1.5,最优选为0.5。
本发明提供了式(I)所示化合物的A晶型,其特征在于其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:11.22±0.20°,24.28±0.20°,27.07±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:11.22±0.20°,13.59±0.20°,15.73±0.20°,17.25±0.20°,21.68±0.20°,24.28±0.20°,26.27±0.20°,27.07±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:5.26±0.20°,11.22±0.20°,13.59±0.20°,15.73±0.20°,17.25±0.20°,18.55±0.20°,21.68±0.20°,23.47±0.20°,24.28±0.20°,25.02±0.20°,26.27±0.20°,27.07±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:11.22±0.20°,24.28±0.20°,和/或27.07±0.20°,和/或13.59±0.20°,和/或15.73±0.20°,和/或17.25±0.20°,和/或21.68±0.20°,和/或26.27±0.20°,和/或5.26±0.20°,和/或18.55±0.20°,和/或23.47±0.20°,和/或25.02±0.20°,和/或7.60±0.20°,和/或8.24±0.20°,和/或10.55±0.20°,和/或14.97±0.20°,和/或16.59±0.20°,和/或19.71±0.20°,和/或23.80±0.20°,和/或27.43±0.20°,和/或28.37±0.20°,和/或29.21±0.20°,和/或30.90±0.20°,和/或34.44±0.20°,和/或36.68±0.20°,和/或37.53±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:5.26°,7.60°,8.24°,10.55°,11.22°,13.59°,14.97°,15.73°,16.59°,17.25°,18.55°,19.71°,21.68°,23.47°,23.80°,24.28°,25.02°,26.27°,27.07°,27.43°,28.37°,29.21°,30.90°,34.44°,36.68°,37.53°。
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晶型的XRPD图谱解析数据如表1所示:
表1式(I)化合物A晶型的XRPD解析数据
Figure PCTCN2021142101-appb-000002
Figure PCTCN2021142101-appb-000003
本发明的一些方案中,上述A晶型,其差示扫描量热曲线在144.2±3.0℃有一个吸热峰的起始点。
本发明的一些方案中,上述A晶型,其DSC图谱如图2所示。
本发明的一些方案中,上述A晶型,其热重分析曲线在170.0±3.0℃处失重达2.19%。
本发明的一些方案中,上述A晶型,其TGA图谱如图3所示。
本发明提供了式(I)所示化合物的B晶型,其特征在于其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.55±0.20°,11.42±0.20°,16.30±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:4.55±0.20°,9.09±0.20°,11.42±0.20°,15.24±0.20°,16.30±0.20°,18.65±0.20°,23.94±0.20°,25.18±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:4.55±0.20°,9.09±0.20°,11.42±0.20°,12.68±0.20°,15.24±0.20°,16.30±0.20°,18.65±0.20°,22.97±0.20°,23.94±0.20°,25.18±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:4.55±0.20°,11.42±0.20°,和/或16.30±0.20°,和/或9.09±0.20°,和/或15.24±0.20°,和/或18.65±0.20°,和/或23.94±0.20°,和/或25.18±0.20°,和/或12.68±0.20°,和/或17.83±0.20°,和/或22.97±0.20°,和/或27.14±0.20°。
本发明的一些方案中,上述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:4.55°,9.09°,11.42°,12.68°,15.24°,16.30°,17.83°,18.65°,22.97°,23.94°,25.18°,27.14°。
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晶型的XRPD图谱解析数据如表2所示:
表2式(I)化合物B晶型的XRPD解析数据
Figure PCTCN2021142101-appb-000004
本发明的一些方案中,上述B晶型,其差示扫描量热曲线在128.2±3.0℃有一个吸热峰的起始点,在198.2±3.0℃有一个吸热峰的峰值。
本发明的一些方案中,上述B晶型,其DSC图谱如图5所示。
本发明的一些方案中,上述B晶型,其热重分析曲线在170.0±3.0℃处失重达2.35%。
本发明的一些方案中,上述B晶型,其TGA图谱如图6所示。
本发明提供了式(II)所示化合物的C晶型,其特征在于其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:14.76±0.20°,18.03±0.20°,23.19±0.20°;
Figure PCTCN2021142101-appb-000005
本发明的一些方案中,上述C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:10.44±0.20°,12.22±0.20°,14.76±0.20°,18.03±0.20°,19.09±0.20°,20.73±0.20°,23.19±0.20°,28.66±0.20°。
本发明的一些方案中,上述C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:10.44±0.20°,12.22±0.20°,14.76±0.20°,18.03±0.20°,19.09±0.20°,20.73±0.20°,23.19±0.20°,25.67±0.20°,27.43±0.20°,28.66±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:14.76±0.20°,18.03±0.20°,和/或23.19±0.20°,和/或10.44±0.20°,和/或12.22±0.20°,和/或19.09±0.20°,和/或20.73±0.20°,和/或28.66±0.20°,和/或25.67±0.20°,和/或27.43±0.20°,和/或7.08±0.20°,和/或8.71±0.20°,和/或13.20±0.20°,和/或23.37±0.20°,和/或24.24±0.20°。
本发明的一些方案中,上述C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°,8.71°,10.44°,12.22°,13.20°,14.76°,18.03°,19.09°,20.73°,23.19°,23.37°,24.24°,25.67°,27.43°,28.66°。
本发明的一些方案中,上述C晶型,其XRPD图谱基本如图7所示。
本发明的一些方案中,上述C晶型的XRPD图谱解析数据如表3所示:
表3式(II)化合物C晶型的XRPD解析数据
Figure PCTCN2021142101-appb-000006
本发明的一些方案中,上述C晶型,其差示扫描量热曲线在197.8±3.0℃有一个吸热峰的起始点。
本发明的一些方案中,上述C晶型,其DSC图谱如图8所示。
本发明还提供了下述实验方法:
实验例:药代动力学评价
实验方法:
受试化合物与10%DMSO/50%PEG400/40%水混合,涡旋并超声,制备得到0.2mg/mL或0.4mg/mL近似澄清溶液,微孔滤膜过滤后备用。选取18至20克的Balb/c雌性小鼠,静脉注射给予候选化合物溶液,剂量为1或2mg/kg。受试化合物与10%DMSO/50%PEG400/40%水混合,涡旋并超声,制备得到0.2mg/mL或1mg/mL近似澄清溶液,微孔滤膜过滤后备用。选取18至20克的Balb/c雌性小鼠,口服给予候选化合物溶液,剂量为2或10mg/kg。收集一定时间的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并用Phoenix WinNonlin软件(美国Pharsight公司)计算药代参数。
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 D8venture衍射仪收集衍射强度数据,光源为CuKα辐射,扫描方式:
Figure PCTCN2021142101-appb-000007
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。
本发明所使用的溶剂可经市售获得。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;TFA代表三氟乙酸;ATP代表三磷酸腺苷;HEPES代表4-羟乙基哌嗪乙磺酸;MgCl 2代表二氯化镁;Pd(PPh 3) 2Cl 2代表双三苯基膦二氯化钯。
技术效果
本发明化合物的晶型稳定性好,易于成药;本发明化合晶型具有优异的DNA-PK激酶抑制活性。
本发明X-射线粉末衍射(X-ray powder diffractometer,XRPD)
仪器型号:PANalytical(帕纳科)公司的X’Pert 3型X-射线衍射仪
测试方法:大约10mg样品用于XRPD检测。
详细的XRPD参数如下:
射线源:Cu,kα(
Figure PCTCN2021142101-appb-000008
Kα2/Kα1强度比例:0.5)
光管电压:45kV,光管电流:40mA
发散狭缝:固定1/8deg
第一索拉狭缝:0.04rad,第二索拉狭缝:0.04rad
接收狭缝:无,防散射狭缝:7.5mm
测量时间:5min
扫描角度范围:4-40deg
步宽角度:0.0263deg
步长:46.665秒
样品盘转速:15rpm
本发明差示扫描量热(Differential Scanning Calorimeter,DSC)
仪器型号:TA Discovery DSC 2500差示扫描量热仪
测试方法:取样品(约1-5mg)置于DSC铝盘内进行测试,在50mL/min N 2条件下,以10℃/min的升温速率,加热样品从25℃(室温)到样品分解前。
本发明热重分析(Thermal Gravimetric Analyzer,TGA)
仪器型号:TA Discovery TGA 5500热重分析仪
测试方法:取样品(约1-5mg)置于TGA铝盘内进行测试,在10mL/min N 2条件下,以10℃/min的升温速率,加热样品从室温到350℃。
本发明动态水分吸附(Dynamic Vapor Sorption,DVS)
仪器型号:SMS Intrinsic型动态水分吸附仪
测试条件:取样品(10~20mg)置于DVS样品盘内进行测试。
详细的DVS参数如下:
温度:25℃
平衡:dm/dt=0.002%/min(最短:10min,最长:180min)
RH(%)测试梯级:10%(0%RH-90%RH,90%RH-0%RH)
5%(90%RH-95%RH,95%RH-90%RH)
RH(%)测试梯级范围:0%RH-95%RH-0%RH
引湿性评价分类如下:
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。
附图说明
图1为式(I)化合物A晶型的Cu-Kα辐射的XRPD谱图;
图2为式(I)化合物A晶型的DSC谱图;
图3为式(I)化合物A晶型的TGA谱图;
图4为式(I)化合物B晶型的Cu-Kα辐射的XRPD谱图;
图5为式(I)化合物B晶型的DSC谱图;
图6为式(I)化合物B晶型的TGA谱图;
图7为式(II)化合物C晶型的Cu-Kα辐射的XRPD谱图;
图8为式(II)化合物C晶型的DSC谱图;
图9为式(I)化合物A晶型的DVS谱图。
具体实施方式
为了更好的理解本发明的内容,下面结合具体实施例来做进一步的说明,但具体的实施方式并不是对本发明的内容所做的限制。
实施例1:式(I)化合物A晶型的制备
Figure PCTCN2021142101-appb-000009
第一步
将17.5L甲苯加入反应釜中,启动搅拌,转速186rpm。然后分别加入碳酸铯(4.31kg,13.2mol),化合物1(2.5kg,9.2mol),化合物2(1.66kg,9.17mol)和4,5-双(二苯基磷)-9,9-二甲基氧杂蒽(101.60g,0.18mol)。使用氮气置换3次后,再加入醋酸钯(19.81g,0.09mol),继续使用氮气置换3次后,升温至80~90℃保温反应60小时。反应完全,搅拌下将反应釜温度降低至25℃后,过滤,滤饼使用二甲苯3L分3次洗涤, 收集滤液。将上述含有中间体3的滤液缓慢加入反应釜,分别加入碳酸铯(4.31kg,13.22mol),化合物4(858g,9.86mol),4,5-双(二苯基磷)-9,9-二甲基氧杂蒽(101.60g,0.18mol)。使用氮气置换3次,然后加入醋酸钯(19.81g,0.09mol),继续使用氮气置换3次后升温至115~125℃反应16小时。反应完全,搅拌下将反应釜温度降低至25℃后,过滤,滤饼使用二甲苯3L分3次洗涤,收集滤液。将含有理论值3.31kg的化合物5滤液25L直接投入下一步。
第二步
将上述含有化合物5的25L二甲苯溶液加入50L反应釜中,启动搅拌,转速192rpm。缓慢滴加880mL盐酸,并控制反应温度于20℃~40℃之间,反应12~16小时。反应完全后,过滤,滤饼使用乙酸乙酯3L洗涤一次。收集滤饼。将滤饼用乙酸乙酯10L稀释,转移至50L分液器中,开启搅拌。然后将803g的氢氧化钠完全溶解的10L水溶液加入分液器后搅拌10分钟。分液萃取,收集有机相,水相用乙酸乙酯10L萃取2次,合并有机相,进行减压浓缩。收集固体。固体用10L甲基叔丁基醚在25℃下搅拌打浆16小时。过滤,收集滤饼。继续将滤液减压浓缩至0.5L。过滤,收集滤饼。合并两次滤饼,室温晾干得到化合物6。
MS-ESI计算值[M+H] +215,实测值215。
1H NMR(400MHz,CDCl 3)δ:6.17-6.12(m,1H),6.07-6.01(m,1H),3.89-3.82(m,4H),3.78(br s,2H),2.99-3.09(m,4H)。
第三步
将12.5L二氯甲烷加入反应釜中,启动搅拌,转速176rpm。然后依次加入化合物6(1.45kg,6.78mol),碳酸氢钠(700g,6.6mol)。将氯化碘(1110g,6.77mol)溶解到二氯甲烷2L中,转移至滴液漏斗并嵌入反应釜。开启滴液漏斗开始滴加,保持滴加速度为1L/30min,温度范围保持在20~35℃,然后保持该温度搅拌1小时。反应完全,将配置好的含970g亚硫酸钠的水溶液10L加入反应釜中,搅拌30min。将反应釜中的混合物转移至50L分液器中搅拌,静置,分液萃取。收集有机相,水相用二氯甲烷5L萃取3次,合并有机相,转移至50L旋转蒸发仪中进行减压浓缩。收集固体,然后放置于真空干燥箱干燥得到化合物7。
MS-ESI计算值[M+H] +341,实测值341。
1H NMR(400MHz,CDCl 3)δ:6.32-6.09(m,1H),3.78(br s,2H),3.97-3.80(m,4H),3.19-2.99(m,4H)。
第四步
在20℃,氮气保护下,向干燥的50L反应釜中加入25.5升三乙胺,启动搅拌,转速100-150rpm。然后依次加入化合物7(5120g,15.1mol),化合物8(1373g,14.0mol),碘化亚铜(44.1g,0.23mol),80.6克Pd(PPh 3) 2Cl 2。升温至55-65℃,反应16小时。反应完全。将反应液过滤,滤饼用6.5升石油醚淋洗,将滤液旋干,得到粗品。粗品用13升乙酸乙酯和6.5升水分散。分液,有机相继续用6.5升水洗涤。有 机相旋干。得到的粗品柱层析纯化,得到化合物9。
MS-ESI计算值[M+H] +311实测值311。
第五步
在20℃下,向50L反应釜中加入16.5升四氢呋喃,启动搅拌,转速100-150rpm。然后加入化合物9(3306g,10.7mol),内温冷却至0-10℃。控制内温为0-40℃将4升浓盐酸缓慢滴入釜中。滴完立即LCMS检测,化合物9含量含量小于2%。控制内温为10-15℃,将亚硝酸叔丁酯(1028g,9.97mol)滴加入釜中。滴完立即LCMS检测,反应完全。将反应液缓慢加入4.1千克碳酸钠的27千克的冰水混合物中,固体出现。过滤,将滤饼转移至釜中,加入30升水,室温搅拌1小时(洗盐),过滤,滤饼转移至釜中,加入25升叔丁基甲醚,室温搅拌0.3小时,过滤。滤饼再次转移至釜中,加入15升叔丁基甲醚,室温搅拌0.3小时,过滤。真空烘箱(<0.09MPa,50℃,15小时)干燥,得化合物10。
MS-ESI计算值[M+H] +286,288实测值286,288。
第六步
将13L四氢呋喃加入30L反应釜中,氮气置换10分钟,保持微弱氮气流,启动搅拌,转速150-200rpm,依次加入甲氧基[环辛二烯]合铱二聚体(99.75g,150.25mmol)、4,4-二叔丁基-2,2,-二吡啶(80.89g,300.5mmol),化合物11(2602.1g,15.03mol,98%纯度),双联频哪醇硼酸酯(3112.5g,12.02mol,98%纯度),开启加热,保持釜内温度70-75℃下搅拌260小时,反应完全,开启冷却,降温至25~35℃,将反应液减压除去溶剂,加入13L[石油醚/乙酸乙酯=10:1]混合溶剂打浆,过滤,将滤饼放入真空烘箱,减压蒸去有机溶剂(<0.07MPa,50℃),得到12。
1H NMR(400MHz,DMSO-d 6)δ:7.84(d,J=6.4Hz,1H),7.48(d,J=8.8Hz,1H),4.11(s,2H),1.17(s,12H)。
第七步
将24L甲苯和5L水加入50L反应釜中,启动搅拌,转速200rpm。然后依次加入碳酸氢钾(1.083kg,10.83nol),化合物10(2.81kg,9.86mol),化合物12(1.90kg,6.42mol)。调节反应釜搅拌桨转速至50rpm,使用聚四氟乙烯管导入氮气流至反应釜中液面以下,使用微弱氮气流鼓泡10分钟。加入75.77g二氯双(三苯基膦)钯(II),盖上釜盖,升温至80~95℃反应14小时。LCMS与HPLC分析化合物10≤5%后,搅拌下将反应釜温度降低至50℃。将11L的水加入反应釜中,在50℃下搅拌1小时。然后降低反应釜温度至25℃后,进行过滤,收集滤饼。滤饼使用水3L洗涤1次。将滤饼再次加入50L反应釜中,再加入10L甲醇。盖上釜盖,升温至50℃搅拌打浆3小时。然后降低温度至25℃。进行过滤,收集滤饼。滤饼使用甲醇6L分2次洗涤。收集固体,然后放置于真空干燥箱干燥得到化合物13。
MS-ESI计算值[M+H] +419,实测值419。
1H NMR(400MHz,CDCl3)δ:9.10(s,1H),7.55(d,J=7.20Hz,1H),7.35(d,J=8.80Hz,1H),7.20(dd,J=6.4,12.8Hz,1H),3.98-3.88(m,6H),3.45-3.38(m,4H)。
第八步
将15L四氢呋喃加入50L反应釜中,启动搅拌,转速189rpm。依次加入化合物13(2.0kg,4.78mol),化合物14(713g,4.79mol),开启冷却循环浴,调节内温为20℃后开始分3批次加入叔丁醇钾1.15kg。加料频率:0.383kg/10min。盖上釜盖,关闭冷却循环浴,反应液在室温下反应1小时。LCMS与HPLC分析化合物13≤1%后,将反应液倒入含有15kg一水合柠檬酸的150L水中。搅拌均匀后过滤,收集滤饼放置于真空干燥箱干燥,将得到的粗品使用硅胶柱层析析纯化(洗脱剂:乙酸乙酯/二氯甲烷:20%~35%),浓缩后得到固体使用2(千克/升)倍体积二氯甲烷打浆,得到化合物15。
MS-ESI计算值[M+H] +531,实测值531。
1H NMR(400MHz,CDCl3)δ:9.15(d,J=36.4Hz,1H),7.72(br t,J=6.0Hz,1H),7.65-7.61(m,1H),7.59(s,1H),7.37(d,J=8.8Hz,1H),7.15-7.25(m,1H),5.98(s,1H),3.97-3.91(m,4H),3.45-3.38(m,4H)。
第九步
将盛有磁力搅拌子的3L三口圆底烧瓶固定于磁力搅拌器上,固定内温温度计和连有三通的氧气袋,检查气密性。向反应瓶中加入1.6L乙腈,并启动搅拌。加入化合物15(199.8g,0.38mol)和碳酸钾(124.8g,0.91mol)。用氧气置换反应瓶中的空气,使反应处于氧气氛围。保持温度在20±2℃反应51.5小时。反应完全,停止反应。平行投料9批,合并反应液(20L),将之缓慢加入到水(100L)中,边加边搅拌,同时析出大量固体。过滤,收集固体,真空干燥(<0.01MPa,50℃,17小时)得粗品。将甲醇(0.9L)和叔丁基甲基醚(2.7L)加入反应釜中,向其中加入粗品,20℃搅拌2.5小时。过滤,收集滤饼,真空干燥(<0.01MPa,50℃,19小时)得到化合物16。
MS-ESI计算值[M+H] +520,522实测值520,522。
1H NMR(400MHz,CDCl 3)δ:9.16(s,1H),8.23(d,J=8.0Hz,1H),7.77(d,J=8.0Hz,1H),7.69(d,J=8.0Hz,1H),7.38(d,J=12.0Hz,1H),7.24-7.19(m,1H),4.00-3.90(m,4H),3.48-3.38(m,4H)。
第十步
将二氯甲烷(18.0L)和甲醇(9.0L)加入到50L反应釜中,启动搅拌,用氮气置换体系中的空气,并维持氮气流,保证反应液置于氮气氛围下。向反应釜中加入化合物16(1350.3g,2.6mol)和碳酸钾(674.5g,4.89mol)。反应液于22±2℃条件下,搅拌36.5小时。反应完全,向反应液加入柠檬酸(1000g),调节pH值至6。减压浓缩(<0.02MPa,25℃)祛除大部分二氯甲烷。向浓缩后的溶液中加入水(90.0L),然后加入碳酸氢钠(500g),搅拌0.5小时,使溶液调节pH值至8,过滤,收集滤饼。再次将滤饼加入水(10.0L)中,搅拌1小时,过滤,收集固体,真空干燥(<0.02MPa,50℃,17小时)得到化合物17。MS-ESI计算值[M+H] +516,518实测值516,518。
1H NMR(400MHz,CDCl 3)δ:9.19(s,1H),8.22(d,J=9.2Hz,1H),7.64(d,J=7.2Hz,1H),7.37(d,J=9.2Hz,1H),7.25-7.22(m,1H),7.19(d,J=9.2Hz,1H),4.27(s,3H),3.97-3.95(m,4H),3.45-3.43(m,4H)。
第十一步
将二氯甲烷(9.4L)和水(3.2L)加入到50L反应釜中,启动搅拌。向反应釜中加入化合物17(1250.9g,2.42mol)、甲酸钠(950.9g,14.2mol)、十六烷基三甲基溴化铵(85.1g,0.23mol)和催化剂(S,S)-N-(对甲苯磺酸)-1,2-二苯乙烷二胺(对异丙基笨)氯化钌(74.4g,0.12mol)。氮气置换并维持微弱的氮气流,保证反应体系处于氮气氛围。反应液于40℃保温搅拌1小时。反应完全,停止反应,降低反应液内温至23℃。向反应液中加入二氯甲烷(5.0L)和水(5.0L)。搅拌、静置、分液,收集有机相(14.5L)。向有机相中加入100-200目硅胶(2.5kg),减压(<0.03MPa,50℃)浓缩制沙。安装硅胶柱(硅胶6.0kg),用二氯甲烷/乙酸乙酯(4/1至1/1)快速洗脱,收集有机相(305L)。减压浓缩有机相(<0.03MPa,50℃),将得到的固体置于真空烘箱中进一步干燥(<0.01MPa,50℃,13小时)得粗品(970.3g)。将甲醇(15.0L)加入到50L反应釜中,启动搅拌,加入粗品(968g),加热升温至64℃,保温搅拌2小时,降低温度至25℃,过滤,收集滤饼并真空干燥(<0.01MPa,55℃,20小时)。将得到的粗品(832g)加入到盛有二氯甲烷(17L)的50L反应釜中,启动搅拌。加入除金属硅胶iMoLboX-LMat-EH01x(75.3g),反应液于18℃搅拌17小时。过滤,减压浓缩虑液(0.02MPa,45℃),向固体中加入甲醇(12.5L)加热搅拌(65℃,19.5小时),降温至25℃,过滤,将得到的粗品重复此步骤一次。固体真空干燥(<0.01MPa,65℃,5天)。将干燥后所得粗品(760g)加入到盛有纯水(15.0L)的50L反应釜中,启动搅拌,保温搅拌(54℃,63.5小时)。降温至25℃、过滤、干燥(<0.01MPa,60℃,9天)得到式(I)化合物A晶型。
MS-ESI计算值[M+H] +518,520实测值518,520。
1H NMR(400MHz,CDCl 3)δ:9.03(d,J=64.0Hz,1H),7.59-7.55(m,1H),7.38(dd,J=8.0,16.0Hz,1H),7.29-7.27(d,J=8.0Hz,1H),7.21-7.06(m,1H),6.98(dd,J=8.0,16.0Hz,1H),6.42(s,1H),5.14(s,1H),4.12(d,J=16.0Hz,3H),3.92(s,4H),3.39(s,4H)。
实施例2:式(I)化合物B晶型的制备
称约5mg式(I)化合物A晶型和5mg式(II)化合物C晶型至HPLC小瓶中,加入0.5mL预饱和的丙酮溶液,室温搅拌约3天后获得式(I)化合物B晶型。
实施例3:式(II)化合物C晶型的制备
称约500mg式(I)化合物A晶型至20mL小瓶中,加入2.0mL乙腈,室温搅拌约2天后获得式(II)化合物C晶型。
实施例4:A晶型和C晶型的竞争实验
在室温(25±3℃)条件下,配置不同水活度的EtOH/H 2O和Acetone/H 2O溶剂体系(a w=0~1),分别将式(I)化合物A晶型和式(II)化合物C晶型各约5mg加到0.5mL的饱和溶液中形成悬浊液,在室温下磁力搅拌10 00转分2天后取样测试XRPD。
溶剂体系(体积比,v/v) 晶型
Acetone B晶型
Acetone/H 2O(94/6,aw=0.2) A晶型
Acetone/H 2O(86/14,aw=0.4) A晶型
Acetone/H 2O(73/27,aw=0.6) A晶型
Acetone/H 2O(50/50,aw=0.8) A晶型
H 2O A晶型
EtOH A晶型
EtOH/H 2O(93/7,aw=0.2) A晶型
EtOH/H 2O(83/17,aw=0.4) A晶型
EtOH/H 2O(68/32,aw=0.6) A晶型
EtOH/H 2O(55/45,aw=0.8) A晶型
结论:在室温水活度0-1范围内,式(I)化合物A晶型均较式(II)化合物C晶型稳定。
实施例5:式(I)化合物A晶型的固体稳定性试验
将式(I)化合物A晶型分别在高温(60℃,闭口)、高湿(92.5%RH,封口膜包裹并扎5个小孔)条件下各放置6天、11天,按照ICH条件(可见光总照度达到1.2E+06Lux·hrs、紫外光总照度达到200W·hrs/m 2)闭口放置在可见光及紫外光下(遮光对照组样品同时放置并用锡箔纸包裹),在60℃/75%RH(封口膜包裹并扎5个小孔)条件下放置1、2个月,在40℃/75%RH(封口膜包裹并扎5个小孔)条件下放置1、2、3个月。在每个取样点分别测试样品化学纯度(HPLC面积纯度)、相对含量(相对-20℃保存的0天样品)及晶型,以确定样品的物理/化学稳定性。
表4:式(I)化合物A晶型的固体稳定性试验结果
Figure PCTCN2021142101-appb-000010
Figure PCTCN2021142101-appb-000011
注:相对色谱纯度=稳定性样品纯度/起始样品纯度×100%;相对起始样品含量的含量变化,起始样品质量/起始样品峰面积*稳定性样品峰面积/稳定性样品质量;峰面积小于0.05%的峰不进行积分。
结论:式(I)化合物A晶型在高温高湿光照以及加速条件下均稳定。
实施例6:式(I)化合物A晶型的吸湿性研究
实验材料:
SMS Intrinsic型动态水分吸附仪
实验方法:
取式(I)化合物A晶型10~20mg置于DVS样品盘内进行测试。
实验结果:
式(I)化合物A晶型的DVS谱图如图9所示,△W=1.263%。
实验结论:
式(I)化合物A晶型在25℃和80%RH下的吸湿增重为1.263%,略有吸湿性。
生物测试数据
实验例1:体外评价DNA-PK激酶抑制活性
本实验测试于Eurofins Pharma Discovery Service,Reaction Biology Corp.(RBC)
实验材料及方法:
人源DNA-PK;Mg/ATP;GST-cMyc-p53;EDTA;Ser15抗体;ATP:10μM;生物素化磷脂酰肌醇-3,4,5-三磷酸;GST标签的GRP1PH域;链霉亲和素别藻蓝蛋白;铕标记的GST单克隆抗体。
实验方法(Eurofins Pharma Discovery Service):
将DNA-PK(h)在含有50nM GST-cMyc-p53,受试化合物和Mg/ATP(根据需要的浓度)的测定缓冲液中温育。通过添加Mg/ATP混合物引发反应。在室温下温育30分钟后,加入含有EDTA的终止溶液终止反应。最后,添加检测缓冲液(含有标记的抗GST单克隆抗体和针对磷酸化p53的铕标记的抗磷酸Ser15抗体)。然后以时间分辨荧光模式读板,并根据公式HTRF=10000×(Em665nm/Em620nm)测定均匀时间分辨荧光(HTRF)信号。
实验结果:
表5DNA-PK激酶活性测试结果
供试品 DNA-PK激酶抑制活性IC 50(nM)
式(I)化合物A晶型 2.0
结论:式(I)化合物A晶型具有显著甚至意料不到的DNA-PK激酶抑制活性。

Claims (26)

  1. 式(I)所示化合物,
    Figure PCTCN2021142101-appb-100001
    其中,n选自0~1,优选为0.5。
  2. 式(I)所示化合物的A晶型,其特征在于其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:11.22±0.20°,24.28±0.20°,27.07±0.20°;
    Figure PCTCN2021142101-appb-100002
  3. 根据权利要求2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:11.22±0.20°,13.59±0.20°,15.73±0.20°,17.25±0.20°,21.68±0.20°,24.28±0.20°,26.27±0.20°,27.07±0.20°。
  4. 根据权利要求3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:5.26±0.20°,11.22±0.20°,13.59±0.20°,15.73±0.20°,17.25±0.20°,18.55±0.20°,21.68±0.20°,23.47±0.20°,24.28±0.20°,25.02±0.20°,26.27±0.20°,27.07±0.20°。
  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:5.26°,7.60°,8.24°,10.55°,11.22°,13.59°,14.97°,15.73°,16.59°,17.25°,18.55°,19.71°,21.68°,23.47°,23.80°,24.28°,25.02°,26.27°,27.07°,27.43°,28.37°,29.21°,30.90°,34.44°,36.68°,37.53°。
  6. 根据权利要求5所述的A晶型,其XRPD图谱基本如图1所示。
  7. 根据权利要求2~6任意一项所述的A晶型,其差示扫描量热曲线在144.2±3.0℃有一个吸热峰的起始点。
  8. 根据权利要求7所述的A晶型,其DSC图谱如图2所示。
  9. 根据权利要求2~6任意一项所述的A晶型,其热重分析曲线在170.0±3.0℃处失重达2.19%。
  10. 根据权利要求9所述的A晶型,其TGA图谱如图3所示。
  11. 式(I)所示化合物的B晶型,其特征在于其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:4.55±0.20°,11.42±0.20°,16.30±0.20°;
    Figure PCTCN2021142101-appb-100003
  12. 根据权利要求11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:4.55±0.20°,9.09±0.20°,11.42±0.20°,15.24±0.20°,16.30±0.20°,18.65±0.20°,23.94±0.20°,25.18±0.20°。
  13. 根据权利要求12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:4.55±0.20°,9.09±0.20°,11.42±0.20°,12.68±0.20°,15.24±0.20°,16.30±0.20°,18.65±0.20°,22.97±0.20°,23.94±0.20°,25.18±0.20°。
  14. 根据权利要求13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:4.55°,9.09°,11.42°,12.68°,15.24°,16.30°,17.83°,18.65°,22.97°,23.94°,25.18°,27.14°。
  15. 根据权利要求14所述的B晶型,其XRPD图谱基本如图4所示。
  16. 根据权利要求11~15任意一项所述的B晶型,其差示扫描量热曲线在128.2±3.0℃有一个吸热峰的起始点,在198.2±3.0℃有一个吸热峰的峰值。
  17. 根据权利要求16所述的B晶型,其DSC图谱如图5所示。
  18. 根据权利要求11~15任意一项所述的B晶型,其热重分析曲线在170.0±3.0℃处失重达2.35%。
  19. 根据权利要求18所述的B晶型,其TGA图谱如图6所示。
  20. 式(II)所示化合物的C晶型,其特征在于其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:14.76±0.20°,18.03±0.20°,23.19±0.20°;
    Figure PCTCN2021142101-appb-100004
  21. 根据权利要求20所述的C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:10.44±0.20°,12.22±0.20°,14.76±0.20°,18.03±0.20°,19.09±0.20°,20.73±0.20°,23.19±0.20°,28.66±0.20°。
  22. 根据权利要求21所述的C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:10.44±0.20°,12.22±0.20°,14.76±0.20°,18.03±0.20°,19.09±0.20°,20.73±0.20°,23.19±0.20°,25.67±0.20°,27.43±0.20°,28.66±0.20°。
  23. 根据权利要求22所述的C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°,8.71°,10.44°,12.22°,13.20°,14.76°,18.03°,19.09°,20.73°,23.19°,23.37°,24.24°,25.67°,27.43°,28.66°。
  24. 根据权利要求23所述的C晶型,其XRPD图谱基本如图7所示。
  25. 根据权利要求20~24任意一项所述的C晶型,其差示扫描量热曲线在197.8±3.0℃有一个吸热峰的起始点。
  26. 根据权利要求25所述的C晶型,其DSC图谱如图8所示。
PCT/CN2021/142101 2020-12-28 2021-12-28 吗啉取代的苯并嘧啶类化合物的晶型及其制备方法 WO2022143671A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180087676.7A CN116670128A (zh) 2020-12-28 2021-12-28 吗啉取代的苯并嘧啶类化合物的晶型及其制备方法

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011596979 2020-12-28
CN202011596979.5 2020-12-28

Publications (1)

Publication Number Publication Date
WO2022143671A1 true WO2022143671A1 (zh) 2022-07-07

Family

ID=82259107

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/142101 WO2022143671A1 (zh) 2020-12-28 2021-12-28 吗啉取代的苯并嘧啶类化合物的晶型及其制备方法

Country Status (2)

Country Link
CN (1) CN116670128A (zh)
WO (1) WO2022143671A1 (zh)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105358552A (zh) * 2013-05-11 2016-02-24 默克专利股份公司 芳基喹唑啉
WO2018114776A1 (en) * 2016-12-19 2018-06-28 Merck Patent Gmbh Combination of a protein kinase inhibitor and an additional chemotherapeutic agent
WO2018178129A1 (en) * 2017-03-30 2018-10-04 Merck Patent Gmbh Crystalline form of (s)-[2-chloro-4-fluoro-5-(7-morpholin-4-ylquinazolin-4-yl)phenyl]-(6-methoxy-pyridazin-3-yl)-methanol
WO2018178133A1 (en) * 2017-03-30 2018-10-04 Merck Patent Gmbh Solid form of (s)-[2-chloro-4-fluoro-5-(7-morpholin-4-ylquinazolin-4-yl)phenyl]-(6-methoxy-pyridazin-3-yl)methanol
CN111909144A (zh) * 2019-05-10 2020-11-10 山东轩竹医药科技有限公司 喹唑啉类dna-pk抑制剂
WO2020259613A1 (zh) * 2019-06-27 2020-12-30 南京明德新药研发有限公司 作为dna-pk抑制剂的喹啉和噌啉衍生物

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105358552A (zh) * 2013-05-11 2016-02-24 默克专利股份公司 芳基喹唑啉
WO2018114776A1 (en) * 2016-12-19 2018-06-28 Merck Patent Gmbh Combination of a protein kinase inhibitor and an additional chemotherapeutic agent
WO2018178129A1 (en) * 2017-03-30 2018-10-04 Merck Patent Gmbh Crystalline form of (s)-[2-chloro-4-fluoro-5-(7-morpholin-4-ylquinazolin-4-yl)phenyl]-(6-methoxy-pyridazin-3-yl)-methanol
WO2018178133A1 (en) * 2017-03-30 2018-10-04 Merck Patent Gmbh Solid form of (s)-[2-chloro-4-fluoro-5-(7-morpholin-4-ylquinazolin-4-yl)phenyl]-(6-methoxy-pyridazin-3-yl)methanol
CN111909144A (zh) * 2019-05-10 2020-11-10 山东轩竹医药科技有限公司 喹唑啉类dna-pk抑制剂
WO2020259613A1 (zh) * 2019-06-27 2020-12-30 南京明德新药研发有限公司 作为dna-pk抑制剂的喹啉和噌啉衍生物

Also Published As

Publication number Publication date
CN116670128A (zh) 2023-08-29

Similar Documents

Publication Publication Date Title
US11680061B2 (en) Crystal forms C and E of pyrazin-2(1H)-one compound and preparation method therefor
CN109689641A (zh) 一种取代的2-氢-吡唑衍生物的晶型、盐型及其制备方法
TWI812223B (zh) 雜環取代的嘌呤酮衍生物的鹽型及晶型
WO2021023272A1 (zh) 一种atr抑制剂的晶型及其应用
WO2020061996A1 (zh) 氘代azd9291化合物的新晶型及其用途
WO2021143843A1 (zh) 一种pde3/pde4双重抑制剂的结晶及其应用
WO2022143671A1 (zh) 吗啉取代的苯并嘧啶类化合物的晶型及其制备方法
CN107793371B (zh) 一类溴结构域识别蛋白抑制剂及其制备方法和用途
WO2022199591A1 (zh) 一种氟取代的吡啶并吡唑类化合物的晶型及其制备方法
CN118076614A (zh) 一种egfr抑制剂的多晶型
CN114524812A (zh) 1,4-二氢-1,6-萘啶化合物的晶型制备以及合成方法
CN106632421A (zh) 1‑(2‑吡啶)‑9‑(4‑甲基苄基)‑β‑咔啉的硝酸铜配合物及其合成方法和应用
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
WO2023179793A1 (zh) 一种1H-吡咯并[2,3-c]吡啶类化合物的晶型及其制备方法
WO2024032558A1 (zh) 一种5,6-二氢噻吩并[3,4-h]喹唑啉类化合物的盐型、晶型及其制备方法
US11708357B2 (en) Crystal form of pyrrolidinyl urea derivative and application thereof
WO2023046072A1 (zh) 咪唑啉酮衍生物的晶型
WO2022099442A1 (zh) 5-氟-1-(4-氟-3-(4-(嘧啶-2-基)哌嗪-1-羰基)苄基)喹唑啉-2,4(1h,3h)-二酮的结晶形式及其制备
WO2020192762A1 (zh) 一种a2a受体拮抗剂的盐型、晶型及其制备方法
CN106478678B (zh) 1‑(2‑吡啶)‑9‑(萘‑2‑甲基)‑β‑咔啉的硝酸铜配合物及其合成方法和应用
CN106632415B (zh) 1‑(2‑吡啶)‑9‑(萘‑2‑甲基)‑β‑咔啉的氯化铜配合物及其合成方法和应用
CN106432288B (zh) 1‑(2‑吡啶)‑9‑(2‑苄氧基乙基)‑β‑咔啉的硝酸铜配合物及合成方法和应用
CN106478676B (zh) 1‑(2‑吡啶)‑9‑(2‑苯氧基乙基)‑β‑咔啉的硝酸铜配合物及合成方法和应用
CN106478689B (zh) 1-(2-吡啶)-9-(4-甲基苄基)-β-咔啉的氯化铜配合物及其合成方法和应用
CN106478677B (zh) 1‑(2‑吡啶)‑9‑(2‑苯氧基乙基)‑β‑咔啉的氯化铜配合物及合成方法和应用

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: 21914405

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202180087676.7

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21914405

Country of ref document: EP

Kind code of ref document: A1

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 11/12/2023)