CN107955019B - Salt form and crystal form of EGFR inhibitor and preparation method thereof - Google Patents

Salt form and crystal form of EGFR inhibitor and preparation method thereof Download PDF

Info

Publication number
CN107955019B
CN107955019B CN201710968068.2A CN201710968068A CN107955019B CN 107955019 B CN107955019 B CN 107955019B CN 201710968068 A CN201710968068 A CN 201710968068A CN 107955019 B CN107955019 B CN 107955019B
Authority
CN
China
Prior art keywords
degrees
compound
formula
reaction
stirring
Prior art date
Legal status (The legal status 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 status listed.)
Active
Application number
CN201710968068.2A
Other languages
Chinese (zh)
Other versions
CN107955019A (en
Inventor
丁照中
陈曙辉
胡利红
刘希乐
张路
李卫东
郭慎慎
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Guangdong Zhongsheng Pharmaceutical Co Ltd
Original Assignee
Guangdong Zhongsheng Pharmaceutical Co Ltd
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 Guangdong Zhongsheng Pharmaceutical Co Ltd filed Critical Guangdong Zhongsheng Pharmaceutical Co Ltd
Publication of CN107955019A publication Critical patent/CN107955019A/en
Application granted granted Critical
Publication of CN107955019B publication Critical patent/CN107955019B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Abstract

The invention discloses a salt form and a crystal form of an EGFR inhibitor and preparation methods thereof, and also discloses application of the salt form and the crystal form in preparation of a medicament for treating non-small cell lung cancer.

Description

Salt form and crystal form of EGFR inhibitor and preparation method thereof
Technical Field
The invention relates to a salt form and a crystal form of a T790M mutant EGFR inhibitor and preparation methods thereof, and also relates to application of the salt form and the crystal form in medicaments for treating non-small cell lung cancer.
Background
Protein tyrosine kinases are a class of enzymes that catalyze the transfer of a phosphate group located on a protein substrate from ATP or GTP to a tyrosine residue. Receptor tyrosine kinases activate secondary signaling pathways by transmitting signals from the outside of the cell to the inside of the cell causing phosphorylation. A variety of cellular processes are regulated by these signals, including proliferation, carbohydrate utilization, protein synthesis, angiogenesis, cell growth and cell survival. In addition, many diseases or conditions are associated with aberrant, abnormal or deregulated activity of one or more kinases.
The epidermal growth factor receptor belongs to the ErbB family of transmembrane tyrosine kinase receptors, which include EGFR (also known as ErbB or HER1), ErbB2 (HER2 or neu gene), ErbB3 (HER3) and ErbB4(HER 4). All have tyrosine kinase activity except HER 3. The EGFR/ErbB family tyrosine kinase receptors have an indispensable role in cell proliferation, differentiation and apoptosis, and thus are effective targets for preventing tumor growth and metastasis. The first generation of epidermal growth factor receptor tyrosine kinase inhibitors (in EGFR-TKI) including gefitinib (J Med 2004; 350:2129-39) and erlotinib (Lancet Oncol 2011; 12:735-42) have been shown to be effective in advanced NSCLC patients with somatic activating mutations. These mutations are point mutations (L858R) encoding in-frame deletions in the kinase domain encoding epidermal growth factor receptor, e.g., exon 19 of polynucleotide, and a substitution of leucine to arginine at position 858 on exon 21 (Nat Rev Cancer 2007; 7: 169-81). However, patients receiving the first generation of EGFR-TKIs eventually develop secondary tumor growth due to drug resistance. The secondary mutation of threonine at position 790 to methionine (T790M) is the most widely recognized mechanism of resistance. This mutation was detected in tumor cells from 50% to 60% of patients with disease progression (N Engl J Med 2005; 353: 207-8). Second generation EGFR-TKIs, such as Afatinib (Lancet Oncol 2014; 15:213-22) and dacomitinib (Cancer 2014; 120:1145-54), were developed to overcome drug resistance generated by the first generation TKIs. They may irreversibly bind covalently to cysteine at position 797 on EGFR. The covalent mechanism is thought to overcome the increased affinity of ATP by the double mutant. Cysteine-797 is however present in all forms of EGFR. These second generation compounds are therefore active not only against active mutant and second mutant EGFR, but also against wild type EGFR. Inhibition of wild-type EGFR is not considered to contribute to its clinical efficacy, but rather leads to side effects of rash and diarrhea (curr. med. chem.2006,13, 3483-.
Thus, third generation EGFR-TKIs, including AZD9291(Cancer Discov 2014; 4:1046-61), CO-1686(Cancer Res.2013; 19: 2240-. They are highly effective in T790M positive tumors, but they still have some toxicity, like still producing diarrhea, rash, nausea and even hyperglycemia clinical side effects ((J Clin Oncol 2014; 32: abstr 8009; J Clin Oncol 2014; 32: abstr 8010). it is clear that a compound with higher activity and lower toxicity brings greater benefit.
The Aslican AZD9291 is an oral small molecule third generation epidermal growth factor tyrosine kinase inhibitor, and the AZD9291 has stronger specific binding capacity for drug-resistant mutation T790M and sensitive mutation sites (19Del and L858R), so the AZD9291 has good treatment effect on drug-resistant non-small cell lung cancer patients after first-line TKI treatment, but has certain inhibition on EGFR wild type, so the side effects of diarrhea, rash and the like can be generated clinically.
Figure BDA0001435422930000021
CO1686 is a novel, oral, selective inhibitor for treating EGFR mutant non-small cell lung cancer patients, which can inhibit key activation and T790M drug resistance mutation, which leaves wild type EGFR signal idle and has good tolerance, and the metabolite produced by CO1686 has inhibitory effect on IGF1R and INSR two targets, so that the clinical side effect is shown as hyperglycemia.
Disclosure of Invention
The present invention provides compounds of formula (II).
Figure BDA0001435422930000022
The present invention also provides a crystalline form a of the compound of formula (II) characterised by an X-ray powder diffraction pattern having characteristic diffraction peaks at the following 2 Θ angles: 4.55 +/-0.2 degrees, 13.84 +/-0.2 degrees, 15.97 +/-0.2 degrees and 18.91 +/-0.2 degrees.
In some embodiments of the present invention, the above form a has an X-ray powder diffraction pattern with characteristic diffraction peaks at the following 2 Θ angles: 4.55 +/-0.2 degrees, 10.21 +/-0.2 degrees, 11.35 +/-0.2 degrees, 13.84 +/-0.2 degrees, 15.97 +/-0.2 degrees, 17.22 +/-0.2 degrees, 18.91 +/-0.2 degrees and 22.54 +/-0.2 degrees.
In some embodiments of the present invention, the above form a has an X-ray powder diffraction pattern with characteristic diffraction peaks at the following 2 Θ angles: 4.55 +/-0.2 degrees, 9.27 +/-0.2 degrees, 10.21 +/-0.2 degrees, 11.35 +/-0.2 degrees, 13.84 +/-0.2 degrees, 14.45 +/-0.2 degrees, 15.97 +/-0.2 degrees, 16.58 +/-0.2 degrees, 17.22 +/-0.2 degrees, 18.91 +/-0.2 degrees, 22.54 +/-0.2 degrees and 23.67 +/-0.2 degrees.
In some embodiments of the invention, the form a of the above form a has an XRPD pattern as shown in figure 1.
In some embodiments of the invention, the XRPD pattern analysis data for form a above is shown in table 1:
TABLE 1
Figure BDA0001435422930000023
Figure BDA0001435422930000031
In some embodiments of the invention, the differential scanning calorimetry curve of form A above has endothermic peaks at 71.51 ℃ ± 3 ℃ and 154.71 ℃ ± 3 ℃.
In some embodiments of the invention, the DSC pattern of form a above is shown in figure 2.
In some embodiments of the invention, the thermogravimetric analysis curve of form a above shows a weight loss of 2.676% at 126.44 ℃ ± 3 ℃ and a weight loss of 2.9411% at 172.01 ℃ ± 3 ℃.
In some embodiments of the invention, the TGA profile of the form a is as shown in figure 3.
The invention also provides a preparation method of the compound A crystal form of the formula (II), which comprises the following steps:
Figure BDA0001435422930000032
(a) adding a compound shown in the formula (I) into a solvent for dissolving;
(b) slowly adding a mixed solution of methanesulfonic acid and a solvent under stirring;
(c) stirring for 8-16 hours at 20-30 ℃;
(d) centrifuging and drying for 8-16 hours;
wherein the solvent is tetrahydrofuran, and the volume ratio of the methanesulfonic acid to the solvent in the mixed solution is 1: 9.
The invention also provides a crystal form B of the compound of formula (II), which is characterized in that the X-ray powder diffraction pattern thereof has characteristic diffraction peaks at the following 2 theta angles: 9.89 +/-0.2 degrees, 15.46 +/-0.2 degrees, 19.60 +/-0.2 degrees and 21.44 +/-0.2 degrees.
In some embodiments of the invention, the form B has an X-ray powder diffraction pattern with characteristic diffraction peaks at the following 2 Θ angles: 5.00 +/-0.2 degrees, 9.89 +/-0.2 degrees, 14.86 +/-0.2 degrees, 15.46 +/-0.2 degrees, 19.60 +/-0.2 degrees, 21.44 +/-0.2 degrees and 24.64 +/-0.2 degrees.
In some embodiments of the invention, the form B has an X-ray powder diffraction pattern with characteristic diffraction peaks at the following 2 Θ angles: 5.00 +/-0.2 degrees, 9.89 +/-0.2 degrees, 12.00 +/-0.2 degrees, 14.86 +/-0.2 degrees, 15.46 +/-0.2 degrees, 19.60 +/-0.2 degrees, 20.11 +/-0.2 degrees, 21.44 +/-0.2 degrees, 24.05 +/-0.2 degrees and 24.64 +/-0.2 degrees.
In some embodiments of the invention, the XRPD pattern of form B above is as shown in figure 5.
In some embodiments of the invention, the XRPD pattern analysis data for form B above is shown in table 2:
TABLE 2
Figure BDA0001435422930000041
In some embodiments of the invention, the differential scanning calorimetry curve of form B above has an endothermic peak at 199.07 ℃ ± 3 ℃.
In some embodiments of the invention, the DSC pattern of form B above is shown in figure 6.
In some embodiments of the present invention, the thermogravimetric analysis curve of the form B has a thermogravimetric analysis curve with a weight loss of 0.1283% at 173.87 ℃ ± 3 ℃ and a weight loss of 0.9514% at 212.74 ℃ ± 3 ℃.
In some embodiments of the invention, the TGA profile of form B above is shown in figure 7.
The invention also provides a preparation method of the compound B crystal form shown in the formula (II), which comprises the following steps:
(a) adding the compound of formula (II) to a solvent to form a suspension;
(b) stirring the suspension for 8-16 hours at 35-45 ℃;
(c) centrifuging and drying for 8-16 hours;
wherein the solvent is selected from the group consisting of acetone, isopropanol, ethyl acetate and 2-methyltetrahydrofuran.
The invention also provides application of the compound or the crystal form A or the crystal form B in preparation of a medicine for treating non-small cell lung cancer.
Technical effects
The compound has better PK property and oral absorption rate, stable crystal form, good hygroscopicity and small influence of light and heat.
The compounds of the present invention show excellent activity against sensitive mutant and double mutant EGFR (sensitive and T790M resistant) and have high selectivity against wild type EGFR. They are likely to provide more effective treatment of diseases caused by abnormalities in epidermal growth factor receptor enzymes.
Definitions and explanations
As used herein, the following terms and phrases are intended to have the following meanings unless otherwise indicated. A particular phrase or term should not be considered as ambiguous or unclear without special definition, but rather construed in a generic sense. When a trade name appears herein, it is intended to refer to its corresponding commodity or its active ingredient.
The intermediate compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof well known to those skilled in the art, with preferred embodiments including, but not limited to, the examples of the present invention.
The chemical reactions of the embodiments of the present invention are carried out in a suitable solvent that is compatible with the chemical changes of the present invention and the reagents and materials required therefor. In order to obtain the compounds of the present invention, it is sometimes necessary for a person skilled in the art to modify or select the synthesis steps or reaction schemes based on the existing embodiments.
The present invention will be specifically described below by way of examples, which are not intended to limit the present invention in any way.
All solvents used in the present invention are commercially available and can be used without further purification.
The invention employs the following abbreviations: r.t. represents room temperature; THF represents tetrahydrofuran; NMP stands for N-methylpyrrolidone; MeSO3H represents methane sulfonic acid; DME represents ethylene glycol dimethyl ether; DCM represents dichloromethane; xphos represents 2-dicyclohexylphosphine-2 ', 4 ', 6 ' -triisopropylbiphenyl; EtOAc for ethyl acetate; MeOH represents methanol; acetone represents acetone; 2-Me-THF represents 2-methyltetrahydrofuran; IPA stands for isopropyl alcohol.
The compound is made by hand or
Figure BDA0001435422930000051
The software names, and the commercial compounds are under the supplier catalog name.
The powder X-ray diffraction (XRPD) method of the invention
The instrument model is as follows: bruker D8advance X-ray diffractometer
The test method comprises the following steps: about 10-20 mg of sample was used for XRPD detection.
The detailed XRPD parameters are as follows:
light pipe: the concentration of Cu, k alpha,
Figure BDA0001435422930000052
voltage of light pipe: 40kV, light tube current: 40mA
Divergent slit: 0.60mm
Detector slit: 10.50mm
Anti-scattering slit: 7.10mm
Scanning range: 4-40deg
Step diameter: 0.02deg
Step length: 0.12 second
Sample pan rotation speed: 15rpm
Differential thermal analysis (DSC) method of the present invention
The instrument model is as follows: TA Q2000 differential scanning calorimeter
The test method comprises the following steps: a sample (. about.1 mg) was placed in a DSC aluminum pan for testing at 50mL/min N2The sample was heated from 30 deg.C (room temperature) to 300 deg.C (or 350 deg.C) at a ramp rate of 10 deg.C/min.
The present invention is a Thermal Gravimetric Analysis (TGA) method
The instrument model is as follows: TA Q5000IR thermogravimetric analyzer
The test method comprises the following steps: a sample (2-5 mg) was placed in a TGA platinum pan for testing at 25mL/min N2Under the condition, the sample is heated from room temperature to 350 ℃ or the weight loss is 20 percent at the heating rate of 10 ℃/min.
The invention relates to a Dynamic Vapor adsorption analysis (DVS) method
The instrument model is as follows: SMS DVS Advantage dynamic vapor adsorption instrument
And (3) testing conditions are as follows: samples (10-15 mg) were placed in a DVS sample pan for testing.
The detailed DVS parameters are as follows:
temperature: 25 deg.C
Balancing: dm/dt is 0.01%/min (shortest: 10min, longest: 180min)
And (3) drying: drying at 0% RH for 120min
RH (%) test step: 10 percent of
RH (%) test step range: 0 to 90 to 0 percent
The hygroscopicity evaluation was classified as follows:
moisture absorption classification ΔW%
Deliquescence Absorb sufficient water to form liquid
Is very hygroscopic ΔW%≥15%
Has moisture absorption 15%>ΔW%≥2%
Slightly hygroscopic 2%>ΔW%≥0.2%
No or almost no hygroscopicity ΔW%<0.2%
Note: Δ W% represents the moisture pick-up weight of the test article at 25. + -. 1 ℃ and 80. + -. 2% RH.
Drawings
FIG. 1 is an XRPD spectrum of Cu-Ka radiation of compound A form (II);
FIG. 2 is a DSC of form A of compound (II);
FIG. 3 is a TGA spectrum of the crystalline form of Compound A of (II);
figure 4 is a DVS spectrum of form a of compound (II);
FIG. 5 is a Cu-Ka radiation XRPD spectrum of compound B form (II);
FIG. 6 is a DSC of form B of Compound (II);
FIG. 7 is a TGA spectrum of form B of compound (II);
fig. 8 is a DVS spectrum of compound (II) form B.
Detailed Description
For better understanding of the present invention, the following description is given with reference to specific examples, but the present invention is not limited to the specific embodiments.
Example 1: preparation of Compounds of formula (I)
Figure BDA0001435422930000071
Step 1:
N2pyridine (1.74Kg, 22.01mol, 1.10 equivalents) in dichloromethane (20L) was cooled to-20 ℃ under protection, trifluoromethanesulfonic anhydride (5.87Kg,20.81mol,1.04 equivalents) was slowly added dropwise, the reaction system was stirred at-20 ℃ for 0.5 hour, 2-bromoethanol (2.50Kg,20.01mol,1.0 equivalents) was slowly added dropwise, the reaction temperature was maintained below 0 ℃ and stirred for 1.0 hour, TLC (petroleum ether: ethyl acetate: 5:1) indicated completion of the reaction. After the reaction, the reaction system was directly filtered, the filtrate was concentrated, petroleum ether (15L) was added, the precipitated solid was filtered, and the filtrate was concentrated to obtain compound b (4.80Kg, yield 93.33%) as a dark yellow oil.1HNMR(400MHz,CDCl3):4.77(t,J=6.40Hz,2H),3.63(t,J=6.40Hz,2H).
Step 2:
compound b (8.5Kg, 33.07mol, 1 eq.) and diphenyl sulfide (6.78Kg,36.38mol, 1.10 eq.) were placed in dry toluene (22.0L) and the reaction mixture was heated to 100 ℃ and stirred for 6 hours under nitrogen.1HNMR showed the reaction was complete. The reaction was cooled to room temperature, and methyl t-butyl ether (20L) was added, the precipitated solid was filtered, and the filter cake was dried in a vacuum oven at 40 ℃ to give compound c (7.4Kg, yield 50.48%) as a brown solid.1H NMR(400MHz,CDCl3):8.08-8.15(m,4H),7.69-7.83(m,6H),4.85-4.93(m,2H),3.66-3.72(m,2H).
And step 3:
compound c (7.4Kg, 16.69mol, 1 equivalent) and potassium bicarbonate (2.01Kg,20.03mol, 1.20 equivalents) were placed in tetrahydrofuran (18.2L) and H2O (9.1L), the reaction mixture was stirred at 15 ℃ for 3 hours.1HNMR showed the reaction was complete. Addition of H2O (10L), the aqueous phase was extracted with DCM (10L. times.4), the organic phases were combined and concentrated to give compound d (5.72Kg, yieldRate 94.57%) was a dark yellow oil.1H NMR(400MHz,CDCl3):7.87(d,J=7.60Hz,8H),7.62-7.78(m,12H),7.48(dd,J=16.00,8.80Hz,2H),6.68(dd,J=8.80,1.60Hz,2H),6.53(dd,J=16.00,2.20Hz,2H).
Figure BDA0001435422930000081
And 4, step 4:
indole-2-carboxylic acid (3Kg, 18.62mol, 1.0 eq) was dissolved in THF (15L), lithium aluminium hydride (900g, 23.7mol, 1.3 eq) was added in portions at 20 ℃, the temperature was programmed to 70 ℃, after the lithium aluminium hydride addition was completed, stirring was carried out at 70 ℃ for 3 hours, TLC (petroleum ether: ethyl acetate 1:1) monitored for completion of the reaction, the temperature was reduced to 20 ℃, water (900mL) was slowly added, NaOH (1N,900mL) was quenched, after complete quenching, suction filtration was carried out under reduced pressure, the filtrate was spin-dried to give a reddish brown oil, which was dissolved in EA (3L), petroleum ether (30L) was added under stirring, and a pale yellow solid precipitated from the solution. Filtration was carried out, the solid was slurried with petroleum ether and ethyl acetate 10:1(15L), suction filtered under reduced pressure, and the solid was dried in a vacuum oven at 50 ℃ to constant weight to give compound r (2.14kg, yield: 74%) as a pale yellow solid.1H NMR(400MHz,CDCl3)δ8.36(br s,1H),7.60(d,J=8.0Hz,1H),7.35(d,J=8.0Hz,1H),7.20(dt,J=1.6,8.0Hz,1H),7.15-7.09(m,1H),6.42(d,J=1.6Hz,1H),4.83(s,2H),1.93(br s,1H).
And 5:
in N2Under protection, dissolving compound r (2.14Kg,14.04mol, 1.0 equivalent) in DCM (50L), cooling to 0 deg.C, adding KOH (1.97Kg, 35.1mol, 2.5 equivalents), stirring at 0 deg.C for 30min, dissolving diphenyl (vinyl) sulfonium (6.36Kg, 17.54mol, 1.25 equivalents) in DCM (10L), slowly dropwise adding into the reaction system, controlling the reaction temperature at 0-5 deg.C, heating to 30 deg.C, stirring for 5 hr, monitoring reaction completion by TLC (petroleum ether: ethyl acetate ═ 2:1), adding water (30L) to quench the reaction, separating, washing the organic phase with saline (60L), drying, spin-drying to obtain black oily substance, adding DME (1.2L), stirring at-10 deg.C overnight, precipitating yellow solid, filtering, and collecting solidPulping with DME (1.2L) at-10 deg.C, filtering, pulping the filter cake with petroleum ether (800mL) once at room temperature, filtering, and drying the filter cake in a vacuum oven to constant weight to obtain compound e as white solid (1.1Kg, yield: 44.5%).1HNMR(400MHz,CDCl3)δ7.63-7.57(m,1H),7.31(dd,J=0.8,8.0Hz,1H),7.21(dt,J=0.8,8.0Hz,1H),7.18-7.11(m,1H),6.24(d,J=0.8Hz,1H),5.01(d,J=0.8Hz,2H),4.22-4.16(m,2H),4.13-4.07(m,2H)
Figure BDA0001435422930000082
Step 6:
5-fluoro-2, 4-dichloropyrimidine (2.12kg, 12.67mol, 1.05 eq.) was added to a solution of ethylene glycol dimethyl ether (10L), the solution was cooled to 0-5 ℃ and N was added2Aluminum trichloride (3.22kg,24.13mol, 2.0 eq.) was added in portions under protection and the reaction was stirred at 30 ℃ for 1 hour. Compound e (3.22kg,24.13mol,1.0 eq) was then slowly added to the reaction mixture, stirred at 20 ℃ for 15 hours, and the completion of the reaction was monitored by TLC (dichloromethane: methanol ═ 6: 1). Slowly adding the reaction solution into water (40L), separating out a large amount of solid, continuously stirring for 0.5h, filtering, adding the filter cake into methanol (16L), pulping at room temperature for 1 h, cooling to room temperature, and filtering. The filter cake was dried to give compound g (3.36kg, 90% yield) as a tan solid.1HNMR(400MHz,CDCl3)δ8.43–8.42(d,J=4.0Hz,1H),7.98-7.96(m,1H),7.40-7.32(m,3H),5.28(s,2H),4.27-4.22(m,4H)
LCMS(ESI)(20-80AB):m/z:304.1[M+1].
And 7:
30L of 1, 4-dioxane was charged into a 50L kettle (20 ℃ C.), and Compound (f) (3.0Kg, 9.88mol, 1.0 equivalent), Compound (g) (1.93Kg, 10.37mol,1.05 equivalent), Palladium acetate (110.91g, 0.494mol, 0.05 equivalent), XPhos (471g, 0.988mol, 0.1 equivalent), Potassium phosphate (4.19Kg, 19.76mol, 2.0 equivalent) were charged into the reaction kettle (20 ℃ C.), N2The replacement was performed 3 times. The temperature is programmed to 100 +/-5 ℃, and after the sampling HPLC central control detection reaction is finished, the temperature is cooled to the room temperature. The reaction solution was filtered under reduced pressure. Filter cakePulping with 40L purified water at 25 + -5 deg.C for 2 hr. The filter cake was slurried with methanol (30L) for 1 hour and dried under vacuum at 50 ℃ to constant weight. Compound i (4.18kg, 91.6% yield) was obtained as a yellow solid.1H NMR(400MHz,DMSO-d6)δ8.89(d,J=8.0Hz,1H),8.59(d,J=1.92Hz,1H),8.50(s,1H),7.78(dd,J=7.2,3.58Hz,1H),7.55(d,J=8.0Hz,1H),7.37(d,J=13.30Hz,1H),7.17-7.29(m,2H),5.11(s,2H),4.21(br dd,J=12.99,4.83Hz,4H),4.02(s,3H).LCMS(ESI)(20-80AB):m/z:454.2[M+1].
And 8:
10.4L N-Methylpyrrolidone was added to a 50L kettle (20 ℃ C.), and Compound (h) (4.15Kg, 9.15mol, 1.0 eq.), N, N, N-trimethylethylenediamine (1.12Kg, 10.98mol,1.2 eq.), potassium carbonate (2.53Kg, 18.30mol, 2.0 eq.) were added to the kettle (20 ℃ C.). The temperature is programmed to 100 +/-5 ℃, and after the sampling HPLC central control detection reaction is finished, the temperature is cooled to 25 +/-5 ℃. 10.4L of purified water was added dropwise to the above solution, and a large amount of red solid was precipitated. And (3) carrying out vacuum filtration, washing the filter cake with purified water (20L) until the filtrate is colorless, pulping the filter cake with 20L of methanol at 25 +/-5 ℃ for 1 hour, carrying out vacuum filtration, and carrying out vacuum drying on the filter cake at 50 ℃ until the weight is constant. Compound k (4.26kg, 86.4% yield) was obtained as a red solid.1H NMR(400MHz,CDCl3)δ9.06(s,1H),8.35(d,J=2.89Hz,1H),7.91(br,dd,J=4.96,3.70Hz,1H),7.54(s,1H),7.35-7.43(m,1H),7.27-7.33(m,3H),6.69(s,1H),5.29(s,2H),4.30(t,J=5.08Hz,2H),4.22(t,J=5.14Hz,2H),4.00(s,3H),3.28(t,J=7.15Hz,2H),2.89(s,3H),2.57(t,J=7.15Hz,2H),2.28(s,6H),1.69(s,3H).LCMS(ESI)(20-80AB):m/z:536.3[M+1].
And step 9:
NMP (6L) was charged to a 10L autoclave (RT), 0.75kg of compound (L) was charged to the autoclave (RT), N2Bubbling for 5 min. 75g of wet Pd-C (50%) are added to the reaction mixture, washed with H2Replace qi for 4 times. The reaction pressure is adjusted to 2.0MPa, and the reaction is heated to 25 ℃ under the stirring condition. Stirring at 25 deg.C for 20 hr (when R1 pressure is less than 1.5MPa, adding H2To 2.0MPa), central control sampling (HPLC), sampling (sampling method: dissolving 0.1mL of the extract in1mL MeOH). Stopping the reaction, and then reducing the pressure of the high-pressure kettle to normal pressure; the reaction solution was filtered through celite (750g) under reduced pressure; the filter cake was washed 1 time with 6L DCM and the filtrate was adjusted to pH 1 with aq.hcl (1M, 0.4L); DCM (6L x 5) was added to the filtrate for extraction and TLC check (sampling method: 0.1ml of aqueous phase was taken directly; developing solvent: DCM/MeOH 10:1, iodine color development, essentially no NMP was seen). Adding NaOH (5M) into water phase to adjust pH to 5, separating out solid, and stirring for 0.5 h. And (4) carrying out suction filtration under reduced pressure, washing a filter cake with water, and collecting the filter cake. The solid was dried under vacuum at 50 ℃ for 48 hours to give compound i.1HNMR(400MHz,CD3OD):δ,8.55(s,1H),8.41(d,J=3.14Hz,1H),7.83-7.89(m,1H),7.49(dd,J=7.22,1.19Hz,1H),7.27(ddd,J=8.82,7.62,1.13Hz,2H),7.16(s,1H),5.14(s,2H),4.18-4.30(m,4H),4.04(s,3H),3.37-3.49(m,4H),2.92(s,6H),2.74(s,3H)LCMS(ESI)(20-80AB):m/z:506.4[M+1].
Step 10:
10L of purified water, 458.76g of NaHCO315L of dichloromethane were added to a 50L reactor and stirred at room temperature until the solid was clear. Slowly adding 1.48Kg of compound (J), stirring for 10min, and standing the reaction solution for 10min for layering. The organic dichloromethane layer was washed 2 times with saturated brine (10L x 2). Dried over anhydrous sodium sulfate. The organic layer was spin dried to give 1.26Kg of brown solid. Free amine (1.26Kg,2.49mol,1.0 eq) was dissolved in 12.6L DME and 1.26L purified water and transferred to a 50L autoclave, cooled to-5-0 ℃, chloropropionyl chloride (347.77g, 2.74mol, 1.1 eq) was slowly added dropwise to the above solution, the addition was complete. Stirring was continued for 0.5 h. LCMS 20-80AB 35MIN monitors less than 1% of starting material remaining and the reaction is complete. Sodium hydroxide (398.74g, 9.96mol, 4.0 equiv) was added to the above solution, the temperature was raised to 70 ℃ and stirred for 6-8 hours, and the reaction was completed. The reaction solution was cooled to room temperature. The reaction solution was poured into 65L of purified water to precipitate a large amount of brown solid, which was then filtered under reduced pressure and the filter cake was dried under vacuum for 24 hours to constant weight to obtain the compound of formula (I) as a brown solid (1.16Kg, yield 78%).1H NMR(400MHz,CDCl3):δ,10.12(s,1H),9.45(s,1H),8.39(d,J=3.01Hz,1H),7.88-7.96(m,1H),7.55(s,1H),7.24-7.40(m,4H),6.81(s,1H),6.25-6.45(m,2H),5.68(dd,J=9.79,2.01Hz,1H),5.31(s,2H),4.14-4.27(m,4H),3.90(s,3H),2.86-2.92(m,2H),2.73(s,3H),2.25-2.32(m,8H)LCMS(ESI)(20-80AB):m/z:560.3[M+1].
Example 2: preparation of the Compound of formula (II)
Adding 8L of acetone into a 50L reaction kettle, adding the compound (1.10Kg,1.97mol, 1.0 equivalent) of the formula (I) into the reaction kettle at room temperature, stirring until the compound is dissolved and clarified, adding thiourea resin (220.44g, 20% by mass ratio) into the reaction solution, stirring overnight at 25 ℃, carrying out suction filtration under reduced pressure, and washing a filter cake with 3L of acetone. The acetone was combined, the solution was transferred to a 50L reactor, warmed to 65 ℃ and methanesulfonic acid (185.34g, 1.93mol, 0.98 eq.) was slowly added dropwise from a constant pressure dropping funnel, and the solution was clear after the addition. After 30min, an off-white solid precipitated from the solution. Stirring for 2 hours under heat preservation, and cooling to 25 ℃. N is a radical of2Suction filtration under reduced pressure under protection, vacuum drying of the filter cake to constant weight in a vacuum oven gave compound of formula (II) as a brown solid (965g, 73% yield).1H NMR(400MHz,CD3OD):δ,8.37(s,1H),8.34(d,J=3.07Hz,1H),7.87(dd,J=7.78,3.14Hz,1H),7.44(d,J=7.91Hz,1H),7.15-7.26(m,2H),6.96(s,1H),6.42-6.55(m,2H),5.86(dd,J=8.72,3.07Hz,1H),5.05(s,2H),4.13-4.22(m,4H),4.00(s,3H),3.49(t,J=5.65Hz,2H),3.28(t,J=5.71Hz,2H),2.87(s,6H),2.72(d,J=1.51Hz,6H).LCMS(ESI)(20-80AB):m/z:560.3[M+1].
Example 3: preparation of compound A crystal form of formula (II)
120mg of the compound of formula (I) are taken, dissolved in 0.5mL of tetrahydrofuran and then 0.14mL of methanesulfonic acid-tetrahydrofuran solution (V/V,1:9) is slowly added with stirring. Stirring overnight at 25 ℃ and then centrifuging, and placing the residual solid sample in a vacuum drying oven (30 ℃) to dry overnight to obtain the crystal form A of the compound shown in the formula (II).
Example 4: preparation of compound B crystal form of formula (II)
50mg of the compound of formula (II) was weighed into a 4.0mL glass vial, and an appropriate amount of acetone was added to make a suspension. And (3) adding magnetons, placing the suspension sample on a magnetic heating stirrer (at 40 ℃) for testing (keeping out of the sun), stirring at 40 ℃ overnight, centrifuging, and placing the residual solid sample in a vacuum drying oven (at 30 ℃) for drying overnight to obtain the crystal form B of the compound shown in the formula (II).
50mg of the compound of formula (II) was weighed into a 4.0mL glass vial and suspended in an appropriate amount of isopropanol. And (3) adding magnetons, placing the suspension sample on a magnetic heating stirrer (at 40 ℃) for testing (keeping out of the sun), stirring at 40 ℃ overnight, centrifuging, and placing the residual solid sample in a vacuum drying oven (at 30 ℃) for drying overnight to obtain the crystal form B of the compound shown in the formula (II).
50mg of the compound of formula (II) was weighed into a 4.0mL glass vial, and an appropriate amount of ethyl acetate was added to make a suspension. And (3) adding magnetons, placing the suspension sample on a magnetic heating stirrer (at 40 ℃) for testing (keeping out of the sun), stirring at 40 ℃ overnight, centrifuging, and placing the residual solid sample in a vacuum drying oven (at 30 ℃) for drying overnight to obtain the crystal form B of the compound shown in the formula (II).
50mg of the compound of formula (II) was weighed into a 4.0mL glass vial, and an appropriate amount of 2-methyltetrahydrofuran was added to make a suspension. And (3) adding magnetons, placing the suspension sample on a magnetic heating stirrer (at 40 ℃) for testing (keeping out of the sun), stirring at 40 ℃ overnight, centrifuging, and placing the residual solid sample in a vacuum drying oven (at 30 ℃) for drying overnight to obtain the crystal form B of the compound shown in the formula (II).
A compound of formula (II) (700g) and absolute ethanol (5.6L) were added to the reaction vessel at room temperature. Stirring and heating to 85 deg.c until the solid is completely dissolved. N-heptane (3.5L) was slowly added to the kettle and some solid precipitated from the solution. Stirring for 1 hour under heat preservation, and cooling the reaction solution to 25 ℃. N is a radical of2And (5) carrying out vacuum filtration under the protection, and drying the solid in a vacuum drying oven to constant weight. Adding the solid and 7L of acetone into a reaction kettle, stirring, heating to an internal temperature of 60 ℃, stirring for 12-40 hours, carrying out crystal transformation, and cooling to room temperature after crystal transformation is completed. N is a radical of2And (3) carrying out vacuum filtration under reduced pressure under protection, and drying a filter cake in a vacuum drying oven to constant weight to obtain the crystal form B (530g, yield 75%) of the compound shown in the formula (II).1H NMR(400MHz,CD3OD):δ,8.37(s,1H),8.34(d,J=3.07Hz,1H),7.87(dd,J=7.78,3.14Hz,1H),7.44(d,J=7.91Hz,1H),7.15-7.26(m,2H),6.96(s,1H),6.42-6.55(m,2H),5.86(dd,J=8.72,3.07Hz,1H),5.05(s,2H),4.13-4.22(m,4H),4.00(s,3H),3.49(t,J=5.65Hz,2H),3.28(t,J=5.71Hz,2H),2.87(s,6H),2.72(d,J=1.51Hz,6H).LCMS(ESI)(20-80AB):m/z:560.3[M+1].
Example 5: study of hygroscopicity of Compound A Crystal form of formula (II)
Experimental materials:
SMS DVS Advantage dynamic vapor adsorption instrument
The experimental method comprises the following steps:
10-15 mg of the compound A crystal form of the formula (II) is placed in a DVS sample tray for testing.
The experimental results are as follows:
the DVS spectrum of the form a of compound of formula (II) is shown in figure 4, with Δ W ═ 7.86%.
And (4) experimental conclusion:
the crystal form A of the compound of the formula (II) has the moisture absorption weight gain of 7.86% at 25 ℃ and 80% RH and is hygroscopic.
Example 6: study of hygroscopicity of Crystal form B of Compound of formula (II)
Experimental materials:
SMS DVS Advantage dynamic vapor adsorption instrument
The experimental method comprises the following steps:
and (3) placing 10-15 mg of the compound B crystal form shown in the formula (II) in a DVS sample tray for testing.
The experimental results are as follows:
the DVS spectrum of the form B of compound of formula (II) is shown in figure 8, with Δ W ═ 0.830%.
And (4) experimental conclusion:
the compound of formula (II) form B has a moisture pick-up of 0.83% at 25 ℃ and 80% RH, and is slightly hygroscopic.
Example 7: solid stability test of Compound B form of formula (II)
According to the guidelines of bulk drug and preparation stability tests (9001 of the four pharmacopoeias 2015 edition), the stability of the crystal form B of the compound of formula (II) under the conditions of high temperature (60 ℃, open), high humidity (92.5% of room temperature/relative humidity, open) and strong light (5000lx, closed) is investigated.
Compound B of formula (II) was weighed as 15mg, placed at the bottom of a glass sample bottle and spread into a thin layer. Sealing the bottle mouth of a sample placed under high-temperature and high-humidity conditions by using aluminum foil paper, and pricking small holes in the aluminum foil paper to ensure that the sample can be fully contacted with ambient air; the samples placed under intense light were sealed with a threaded cap. Samples placed under different conditions were sampled and tested (XRPD) at day 5 and day 10, and the test results were compared with the initial test results at day 0, and the test results are shown in table 3 below:
TABLE 3 solid stability test results for Compound B form of formula (II)
Figure BDA0001435422930000121
And (4) conclusion: the compound B crystal form of the formula (II) has good stability under the conditions of high temperature, high humidity and strong illumination.
In vivo pharmacodynamic study of form B of compound of formula (II) in the NCI-H1975 xenograft (CDX) model
Experimental materials:
BALB/c nude mice, female, 6-8 weeks, weighing about 18-22 grams, were kept in a special pathogen-free environment in single ventilated cages (10 mice per cage). All cages, bedding and water were sterilized prior to use. All animals were free to obtain a standard certified commercial laboratory diet. There were 100 mice purchased from Weitonglihua, Beijing for study. Each mouse was implanted subcutaneously in tumor tissue (20-30 cubic millimeters) in the right flank for tumor growth. The experiment was started when the mean tumor volume reached about 140-. The experimental method comprises the following steps:
in vivo efficacy was performed by subcutaneous implantation of NCI-H1975 lung cancer patient-derived xenograft (CDX) BALB/c nude mice. Experiment test compounds were administered orally daily for 21 consecutive days. Tumor volume was measured twice a week with a two-dimensional caliper, the volume being measured in cubic millimeters and calculated by the following formula: v ═ 0.5a × b2, where a and b are the major and minor diameters of the tumor, respectively. The anti-tumor efficacy was determined by dividing the mean tumor gain volume of animals treated with the compound by the mean tumor gain volume of untreated animals.
The experimental results are as follows: see table 4.
TABLE 4
Figure BDA0001435422930000131
And (4) experimental conclusion:
the compound B crystal form (12mg/Kg) of the formula (II) has the equivalent tumor inhibition activity to AZD9291(3mg/Kg) and CO1686(50 mg/Kg).
In vivo study of pharmaceutical efficacy of form B of Compound of formula (II) in A431 xenograft (CDX) model
Experimental materials:
BALB/c nude mice, female, 6-8 weeks, weighing about 18-20 grams, were kept in a special pathogen-free environment in a single ventilated cage (5 mice per cage). All cages, bedding and water were sterilized prior to use. All animals were free to obtain a standard certified commercial laboratory diet. All experimental mice were purchased from BK Laboratory Animal co, shanghai, LTD mice for study. Each mouse was implanted subcutaneously in tumor tissue (20-30 cubic millimeters) in the right flank for tumor growth. The experiment was started when the mean tumor volume reached about 150-
The experimental method comprises the following steps:
in vivo selectivity experiments were performed on subcutaneously implanted human skin squamous cell A431 xenograft (CDX) BALB/c nude mice.
Test compounds were administered orally for 11 consecutive days. Tumor volume was measured twice a week with a two-dimensional caliper, the volume being measured in cubic millimeters and calculated by the following formula: v ═ 0.5a × b2, where a and b are the major and minor diameters of the tumor, respectively. The anti-tumor efficacy was determined by dividing the mean tumor gain volume of animals treated with the compound by the mean tumor gain volume of untreated animals.
The experimental results are as follows: see table 5.
TABLE 5
Figure BDA0001435422930000132
Figure BDA0001435422930000141
And (4) experimental conclusion:
the compound B crystal form (12mg/Kg) of the formula (II) has better selectivity than AZD9291(3 mg/Kg).

Claims (8)

1. Form B of a compound of formula (II) characterized by an X-ray powder diffraction pattern having characteristic diffraction peaks at the following 2 θ angles: 5.00 +/-0.2 degrees, 9.89 +/-0.2 degrees, 10.04 +/-0.2 degrees, 12.00 +/-0.2 degrees, 14.86 +/-0.2 degrees, 15.46 +/-0.2 degrees, 19.60 +/-0.2 degrees, 20.11 +/-0.2 degrees, 21.44 +/-0.2 degrees, 24.05 +/-0.2 degrees, 24.64 +/-0.2 degrees, 24.82 +/-0.2 degrees
Figure FDA0003194267160000011
2. The form B according to claim 1, having an XRPD pattern as shown in figure 5.
3. Form B according to claim 1 or 2, having a differential scanning calorimetry curve with an endothermic peak at 199.07 ± 3 ℃.
4. The form B according to claim 3, having a DSC profile as shown in figure 6.
5. Form B according to claim 1 or 2 having a thermogravimetric analysis curve with a weight loss of 0.1283 at 173.87 ± 3 ℃ and a weight loss of 0.9514 at 212.74 ± 3 ℃.
6. The crystalline form B according to claim 5 having a TGA profile as shown in figure 7.
7. A process for preparing the crystalline form B of compound of formula (II) according to claims 1 to 6, comprising:
(a) adding the compound of formula (II) to a solvent to form a suspension;
(b) stirring the suspension for 8-16 hours at 35-45 ℃;
(c) centrifuging and drying for 8-16 hours;
wherein the solvent is selected from the group consisting of acetone, isopropanol, ethyl acetate and 2-methyltetrahydrofuran.
8. Use of the form B according to claims 1-6 in the preparation of a medicament for treating non-small cell lung cancer.
CN201710968068.2A 2016-10-17 2017-10-16 Salt form and crystal form of EGFR inhibitor and preparation method thereof Active CN107955019B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610903608 2016-10-17
CN2016109036084 2016-10-17

Publications (2)

Publication Number Publication Date
CN107955019A CN107955019A (en) 2018-04-24
CN107955019B true CN107955019B (en) 2021-09-14

Family

ID=61963482

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710968068.2A Active CN107955019B (en) 2016-10-17 2017-10-16 Salt form and crystal form of EGFR inhibitor and preparation method thereof

Country Status (1)

Country Link
CN (1) CN107955019B (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108285460A (en) * 2017-01-09 2018-07-17 南京圣和药业股份有限公司 A kind of pharmaceutical salts of EGFR kinase inhibitor and preparation method thereof and purposes
WO2019238088A1 (en) * 2018-06-13 2019-12-19 基石药业 Salt form and crystal form of pyridopyridone derivative
WO2020007219A1 (en) * 2018-07-02 2020-01-09 南京明德新药研发有限公司 Crystalline form of egfr inhibitor and preparation method therefor
CN112912380B (en) * 2018-12-21 2023-08-11 基石药业(苏州)有限公司 Crystal form and amorphous form of MEK inhibitor and application thereof
KR20220008313A (en) * 2019-05-13 2022-01-20 뤄신 헬스케어 사이언스 앤드 테크놀로지 디벨롭먼트 (베이징) 리미티드 Crystal form of quinazolinone-based compound and method for preparing the same

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013138495A1 (en) * 2012-03-15 2013-09-19 Celgene Avilomics Research, Inc. Solid forms of an epidermal growth factor receptor kinase inhibitor
CN103748096A (en) * 2012-08-06 2014-04-23 美国艾森生物科学公司 Novel pyrrolopyrimidine compounds as inhibitors of protein kinases
CN105348266A (en) * 2011-07-27 2016-02-24 阿斯利康(瑞典)有限公司 Substituted-3-chlorin-N-[3-(pyrimidine-2-ylamine)phenyl]propanamide or salts thereof
CN105377835A (en) * 2013-07-11 2016-03-02 贝达药业股份有限公司 Protein tyrosine kinase modulators and methods of use

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105348266A (en) * 2011-07-27 2016-02-24 阿斯利康(瑞典)有限公司 Substituted-3-chlorin-N-[3-(pyrimidine-2-ylamine)phenyl]propanamide or salts thereof
WO2013138495A1 (en) * 2012-03-15 2013-09-19 Celgene Avilomics Research, Inc. Solid forms of an epidermal growth factor receptor kinase inhibitor
CN103748096A (en) * 2012-08-06 2014-04-23 美国艾森生物科学公司 Novel pyrrolopyrimidine compounds as inhibitors of protein kinases
CN105377835A (en) * 2013-07-11 2016-03-02 贝达药业股份有限公司 Protein tyrosine kinase modulators and methods of use

Also Published As

Publication number Publication date
CN107955019A (en) 2018-04-24

Similar Documents

Publication Publication Date Title
CN107955019B (en) Salt form and crystal form of EGFR inhibitor and preparation method thereof
CN107531678B (en) EGFR inhibitor and pharmaceutically acceptable salts and polymorphs thereof and uses thereof
CN106928275B (en) Preparation method of spiro amine aryl phosphorus oxide compound and intermediate and crystal form thereof
WO2018214886A1 (en) Crystal form of deuterated azd9291, preparation method therefor, and use thereof
CN108602815B (en) Pyrido [1,2-a ] pyrimidinone analogs, crystalline forms thereof, intermediates thereof, and methods of making the same
CN109689641A (en) A kind of substituted 2- hydrogen-pyrazole derivatives crystal form, salt form and preparation method thereof
CN112119074A (en) EGFR inhibitors
CN110950847B (en) Novel crystal form of deuterated AZD9291 compound and application thereof
WO2021121146A1 (en) Crystal form a of aminopyrimidine mesylate compound, preparation method therefor, and application thereof
WO2023174400A1 (en) Salt of substituted amino six-membered nitric heterocyclic compound, crystal form thereof, method for preparing same, and use thereof
WO2020007219A1 (en) Crystalline form of egfr inhibitor and preparation method therefor
CN107266437B (en) The crystal form of N- phenyl -2- amino-metadiazine compound, salt form and preparation method thereof
US20230322687A1 (en) Salt of arylaminoquinazoline-containing compound, and preparation method therefor and use thereof
CN110950871A (en) 2-substituted-4-arylamino pyrazolotriazine derivative and application thereof in preparing antitumor drugs
CN109476634B (en) Crystal of salt of quinazoline derivative
WO2020147838A1 (en) Salt of egfr inhibitor, crystal form, and preparation method therefor
CN113784971B (en) Crystal form of EGFR inhibitor and preparation method thereof
CN111148747B (en) Salt form and crystal form of pyridopyrimidine compound and preparation method thereof
AU2015392050B2 (en) Fumarate of pyridylamine compound and crystals thereof
CN107778284A (en) HKI-272 compound
WO2023093859A1 (en) Salt of axl kinase inhibitor, preparation method therefor and use thereof
CN108299419B (en) Novel crystal forms of novel EGFR kinase inhibitor and preparation method thereof
CN114907342A (en) Polymorph of free base of nitrogen-containing fused heterocyclic compound and preparation method and application thereof
WO2023283206A1 (en) Crystalline polymorphs of epidermal growth factor receptor inhibitor, and compositions and methods thereof
CN116574100A (en) Novel crystal form of citric acid monosalt of pyridopyrimidinone compound, and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant