CN111100117B - Crystal form A of aminopyrimidine compound mesylate and preparation method and application thereof - Google Patents

Crystal form A of aminopyrimidine compound mesylate and preparation method and application thereof Download PDF

Info

Publication number
CN111100117B
CN111100117B CN201911311034.1A CN201911311034A CN111100117B CN 111100117 B CN111100117 B CN 111100117B CN 201911311034 A CN201911311034 A CN 201911311034A CN 111100117 B CN111100117 B CN 111100117B
Authority
CN
China
Prior art keywords
degrees
compound
mesylate
crystal form
aminopyrimidine
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
CN201911311034.1A
Other languages
Chinese (zh)
Other versions
CN111100117A (en
Inventor
汤春
M·N·格雷科
M·J·科斯坦佐
张晓霞
J·彭
D·张
Y·卢
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.)
BETA PHARMA (SHANGHAI) Co.,Ltd.
Original Assignee
Beta Pharma Shanghai 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 Beta Pharma Shanghai Co ltd filed Critical Beta Pharma Shanghai Co ltd
Priority to CN201911311034.1A priority Critical patent/CN111100117B/en
Publication of CN111100117A publication Critical patent/CN111100117A/en
Priority to PCT/CN2020/135661 priority patent/WO2021121146A1/en
Priority to TW109145163A priority patent/TW202128666A/en
Priority to ARP200103555A priority patent/AR120821A1/en
Application granted granted Critical
Publication of CN111100117B publication Critical patent/CN111100117B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • C07D403/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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

Landscapes

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

Abstract

The invention provides a crystal form A of amino pyrimidine compound mesylate, wherein X-ray powder diffraction represented by a 2 theta angle has diffraction peaks at the vicinity of 11.06 +/-0.2 degrees, 12.57 +/-0.2 degrees, 13.74 +/-0.2 degrees, 14.65 +/-0.2 degrees, 15.48 +/-0.2 degrees, 16.58 +/-0.2 degrees, 17.83 +/-0.2 degrees, 19.20 +/-0.2 degrees, 19.79 +/-0.2 degrees, 20.88 +/-0.2 degrees, 22.05 +/-0.2 degrees, 23.06 +/-0.2 degrees, 24.23 +/-0.2 degrees, 25.10 +/-0.2 degrees, 25.71 +/-0.2 degrees, 26.15 +/-0.2 degrees, 27.37 +/-0.2 degrees and 27.42 +/-0.2 degrees (as shown in figure 3). The crystal form A of the aminopyrimidine compound mesylate has good solubility and high bioavailability in animal bodies. It can be used for the treatment or prevention of diseases or medical conditions mediated by mutations (activation or drug resistance) of the Epidermal Growth Factor (EGFR) in mammals, especially humans, especially cancer.

Description

Crystal form A of aminopyrimidine compound mesylate and preparation method and application thereof
Technical Field
The invention belongs to the field of drug synthesis, and particularly relates to a crystal form A of aminopyrimidine compound mesylate, and a preparation method and application thereof.
Background
Epidermal Growth Factor Receptor (EGFR) is a type of transmembrane receptor tyrosine kinase in human body, and the activation (i.e. phosphorylation) of the region has important significance for the inhibition of the proliferation, angiogenesis, tumor invasion, metastasis and apoptosis of tumor cells. EGFR kinases are involved in the disease progression of most cancers and these receptors are overexpressed in many major human tumors. Over-expression, mutation or high expression of ligands that bind to these family members can lead to the development of a number of neoplastic diseases, such as non-small cell lung cancer, colorectal cancer, breast cancer, head and neck cancer, cervical cancer, bladder cancer, thyroid cancer, gastric cancer, and renal cancer.
In recent years, epidermal growth factor receptor tyrosine kinase has become one of the most attractive targets for the research of antitumor drugs at present. In 2003, FDA approved the first epidermal growth receptor tyrosine kinase inhibitor (EGFR-TKI) class of drugs Iressa in the United states
Figure BDA0002324528400000011
(gefitinib) for the treatment of advanced non-small cell lung cancer (NSCLC) opened the development of first generation EGFR inhibitors. A plurality of clinical tests prove that the treatment effect of the molecular targeted medicament for EGFR positive mutation non-small cell lung cancer patients is obviously superior to that of the traditional chemotherapy.
Although the first generation of EGFR-inhibiting class of targeted drugs responded well to the initial treatment of numerous non-small cell lung cancer (NSCLC) patients, most patients eventually developed disease progression due to development of drug resistance (e.g., EGFR secondary T790M mutation). Drug resistance arises through a variety of mechanisms based on mutations in the activity of the original EGFR pathway. For drug resistance studies of first generation EGFR inhibitors, the leading edge has been irreversible third generation EFGR inhibitors.
However, to date, the third generation of EGFR inhibitors worldwide has been excluded, except for oxitinib (terixate) by the company asikang
Figure BDA0002324528400000012
) The development is successful, no medicine which is effective for the T790M drug-resistant mutation patient is approved for clinical treatment, and a plurality of candidate medicines for the T790M mutation are in the clinical development stage. This type of third generation EGFR inhibitor is chemically distinct from the first generation. The main difference from the first generation EGFR inhibitors is that they all use a highly selective core structure in place of the less selective aminoquinoline of the first, second generation EGFR-TKIA core structure. These third generation compounds all have high specific selectivity for the T790M mutation after EGFR positive resistance relative to wild-type EGFR.
The patent application CN201580067776.8 discloses a compound with the structure shown in the formula I, which also belongs to the third generation EGFR-TKI small molecule targeted drugs. The compound has high inhibition effect on non-small cell lung cancer (NSCLC) cells with single-activity mutation and T790M double-mutation EGFR, and the effective inhibition concentration of the compound is obviously lower than that required for inhibiting the activity of wild type EGFR tyrosine kinase. Has the characteristics of good selectivity, lower toxic and side effects and good safety.
Figure BDA0002324528400000021
Patent application CN201780050034.3 of the same company also discloses various salts and corresponding crystal forms of the compounds of the structure of formula I as above, and example 2 thereof discloses two crystal forms of the mesylate salt of the compound of formula I, 2A and 2B, respectively.
Disclosure of Invention
The invention aims to provide a novel crystal form A of amino pyrimidine compound mesylate, and a preparation method and application thereof. Relates to the mesylate salt, substantially pure novel crystalline form, and pharmaceutically acceptable salts of the compound of formula I.
Compared with the crystals 2A and 2B disclosed in the patent example 2 of CN201780050034.3 published by the company, the crystal form of the application has different XPRD characteristic peak numbers, positions and intensities, and has obvious difference in melting points, and the crystal form obtained by the preparation process has higher purity and is more stable, thereby being more beneficial to the application of the crystal form in the field of medicine.
In order to achieve the purpose, the invention adopts the following technical scheme:
the invention provides a crystal form A of the mesylate salt of an aminopyrimidine compound of formula I having an XRPD pattern with diffraction peaks at 11.06, 12.57, 13.74, 14.65, 15.48, 16.58, 17.83, 19.20, 19.79, 20.88, 22.05, 23.06, 24.23, 25.10, 25.71, 26.15, 27.37 and 27.42 (relative intensity greater than 10%),
Figure BDA0002324528400000022
preferably, the XRPD pattern of form a is as shown in figure 3.
Preferably, the infrared spectrum of the form A is shown in figure 6.
The invention also provides a preparation method of the crystal form A of the amino pyrimidine compound mesylate, which comprises the following steps: dissolving the mesylate of the compound shown in the formula I by using an organic solvent, stirring, crystallizing, filtering and drying to obtain a target crystal form A, wherein the organic solvent comprises dimethyl sulfoxide, tetrahydrofuran, N-methylpyrrolidone, N-dimethylformamide and N, N-dimethylacetamide.
Preferably, the method further comprises the following steps: the compound of the formula I and methanesulfonic acid are mixed according to a feeding molar ratio of 1.05-0.97 to prepare the compound of the formula I, i.e. methanesulfonic acid salt.
Preferably, the crystallization is carried out by adding an anti-solvent and stirring, wherein the anti-solvent comprises methyl tert-butyl ether, isopropyl acetate, ethyl acetate, methyl isobutyl ketone, acetonitrile, toluene, acetone and water.
Preferably, the filtration is followed by atmospheric or vacuum drying at 30 to 70 deg.C (more preferably 40 to 50 deg.C).
The invention also provides a pharmaceutical composition, which comprises the crystal form A of the amino pyrimidine compound mesylate and a pharmaceutically acceptable carrier.
The invention also provides a crystal form A of the amino pyrimidine compound mesylate, and application of the pharmaceutical composition or the combined medicine in preparing antitumor drugs.
Preferably, the tumor is lung cancer.
The invention has the following beneficial effects:
experiments show that the crystal form A of the aminopyrimidine compound mesylate has high purity, stable property, good solubility and high bioavailability in animal bodies. Their use in pharmaceutical combinations and combinations for the treatment or prevention of diseases or medical conditions mediated by mutations (activated or drug-resistant forms) of the Epidermal Growth Factor (EGFR) in mammals, especially humans, and in particular cancer.
Drawings
FIG. 1 is a nuclear magnetic hydrogen spectrum of a compound of formula I.
Fig. 2 is a nuclear magnetic hydrogen spectrum of form a.
Figure 3 is an XRPD pattern and table for form a.
Figure 4 is a Differential Scanning Calorimetry (DSC) profile of form a.
Figure 5 is a thermogravimetric analysis (TGA) profile of form a.
Figure 6 is an Infrared (IR) spectrum of form a.
FIG. 7 is a comparison of powder X-ray diffraction patterns (XPRD) of form A obtained by different preparation methods.
FIG. 8 is a graph of blood concentration versus time after a single administration to SD rats.
FIG. 9 is a plot of tumor volume change versus time for subcutaneous transplantable tumors of human lung carcinoma H1975 nude mice, given as single and combined doses, of the mesylate salt of the compound of formula (I), prepared in example 2 (i.e., form A).
FIG. 10 is a graph of weight change versus time for human lung cancer H1975 nude mice treated with single and combination doses of the mesylate salt of the compound of formula (I), prepared in example 2 (i.e., form A).
Detailed Description
The advantageous effects of the present invention will now be further described by the following examples, which are for illustrative purposes only and do not limit the scope of the present invention, and variations and modifications apparent to those skilled in the art according to the present invention are also included in the scope and the like of the present invention.
In the following examples, the "room temperature" may be in the range of 15 to 25 ℃.
Preparation of (mono) N- (2- (2- (dimethylamino) ethoxy) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide (compound of formula I)
Figure BDA0002324528400000041
Known (see for example CN201580067776.8) N- (2- (2- (dimethylamino) ethoxy) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamides (compounds of formula I) can be prepared by the following synthetic route:
Figure BDA0002324528400000042
step 1-preparation of intermediate J:
Figure BDA0002324528400000051
preparation: in a 10L reaction flask, 6L of anhydrous tetrahydrofuran solvent was added, protected with nitrogen, and cooled to 0 ℃. While stirring, 101g of sodium hydride (101g, 2.52mol) were slowly added, while the internal temperature was not more than 10 ℃ and 234g of dimethylaminoethanol (234g, 2.62mol) were added. After the addition, the temperature is adjusted to room temperature to prepare the sodium alkoxide solution.
In a 30L reaction flask, N- (4-fluoro-2-methoxy-5-nitrophenyl) -4- (1-methyl-1H-indol-3-yl) -2-pyrimidinamine (starting material B) (430g, 1.10mol) was added, then 9L of tetrahydrofuran was added, stirred to dissolve and clear, the temperature was controlled at 10. + -. 10 ℃ and the prepared sodium alkoxide solution was slowly added dropwise. Controlling the temperature at 10 +/-10 ℃, and preserving the temperature for 5.0 h. When the content of the raw materials is less than or equal to 0.5 percent, the reaction is finished. And controlling the temperature to be 10 +/-10 ℃, slowly dropwise adding a 3% hydrochloric acid solution, adjusting the pH value of the solution to 6-7, stirring for 1.5h, standing for layering, separating an organic phase, and concentrating to 15-20V. After the temperature is reduced to 20 +/-5 ℃, 4.3kg of water is slowly dripped, and the mixture is filtered and dried to obtain 497g of yellow powder intermediate J, wherein the yield is 98.0 percent and the HPLC purity is 99.3 percent. MS m/z: 463.2[ M +1]]. Nuclear magnetic data:1HNMR(d6-DMSO):δppm:8.78(s,1H);8.42-8.28(m,3H);8.16(s,1H);7.53(d,1H,J=8.28);7.29-7.20(m,2H);7.13-7.07(m,1H);7.01(s,1H);4.33(t,2H,J=5.65);4.02(s,3H);3.88(s,3H);2.71(t,2H,J=5.77);2.27(s,6H)。
step 2-preparation of intermediate K:
Figure BDA0002324528400000052
preparation: adding 5L of tetrahydrofuran and an intermediate J (350g, 108mmol) into a 10L hydrogenation reaction kettle, adding 17.5g of wet palladium-carbon, performing hydrogen replacement on the hydrogenation kettle, adjusting the pressure value to be 0.2Mpa, controlling the temperature to be 25 ℃, performing heat preservation reaction for 9 hours, monitoring the reaction progress by HPLC (high performance liquid chromatography), and stopping the reaction, wherein the substrate is less than or equal to 0.5%. Filtering, concentrating the filtrate under reduced pressure until the solvent volume is about 2L, adjusting the internal temperature to room temperature, slowly dropwise adding n-heptane 4L within 4-7 h, filtering, and drying under reduced pressure to obtain 285g of off-white powder intermediate K, with yield of 86% and HPLC purity of 99.60%. MS m/z: 433.3[ M +1 ].
Nuclear magnetic data:1HNMR(CDCl3):δppm:8.42(d,1H,J=7.78),8.28(s,1H),8.26-8.23(m,1H),7.78(s,1H),7.51(d,1H,J=8.28),7.41(s,1H),7.26-7.23(m,1H),7.19-7.11(m,2H),6.72(s,1H),4.38(br,2H),4.06(t,2H,J=5.77),3.88(s,3H),3.75(s,3H),2.63(t,2H,J=5.77),2.26(s,6H).
step 3-preparation of compounds of formula I:
Figure BDA0002324528400000061
preparation: into a reaction flask, 250mL of anhydrous tetrahydrofuran solvent and intermediate K (14g, 32mmol) were added and stirred, cooled to 0-5 ℃ and 10% hydrochloric acid (12mL) was added and stirred for 20 minutes. 3-chloropropionyl chloride (5.6g, 45mmol) was slowly added dropwise at 0-5 deg.C into the reaction flask. Stirring for 3 hours, after sampling and testing (K/(U + K) < 0.5%) to be qualified, adding 36% potassium hydroxide aqueous solution (75ml, 480mmol), heating to 23-25 ℃, and stirring for 12 hours. Heating to 50-60 deg.c and stirring for 4 hr. And (4) after the sampling test is qualified (U/(U + L) is less than or equal to 0.1 percent), standing and separating the liquid. The organic phase was separated, washed three times with 10% saline, dried, filtered and the organic phase was concentrated to 150 ml. The temperature is increased to 40 ℃, 150ml of n-heptane is slowly dropped into the mixture, and the mixture is cooled to room temperature, and crystals are separated out. Filtration and drying gave 10.71g of a light brown solid (compound of formula I), 68% yield, HPLC purity: 99.8% (all single hetero not exceeding 0.15%). MS m/z: 487.3[ M +1 ].
Nuclear magnetic data (figure 1):1HNMR(d6-DMSO):δppm:9.84(s,1H),8.90~8.82(m,1H),8.32-8.25(m,2H),7.89(s,1H),7.51(d,1H,J=8.25),7.27~7.10(m,1H),6.94(s,1H),6.49(dd,1H,J=16.88,10.13),6.25(dd,1H,J=16.95,1.81),5.80~5.75(m,1H),4.19(t,2H,J=5.57),3.88(d,6H,J=14.63,6H),3.34(s,3H),2.58(d,2H,J=5.5),2.28(s,6H)。
preparation of (di) N- (2- (2- (dimethylamino) ethoxy) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide mesylate (form a)
Example 1
Figure BDA0002324528400000071
The compound of formula I (3g, 6.1mmol) was dissolved in 24ml dimethylsulfoxide DMSO solvent, warmed to 65 ℃ and the solution was stirred. An equivalent amount of methanesulfonic acid (0.59g, 6.1mmol) was added to the system. The temperature is reduced to 50 ℃, and 12ml of isopropyl acetate IPAc is slowly added. Stirring at 50 deg.C for 1 hr, and cooling to 15 deg.C. 21ml of IPAc was added over 4 hours. Stirring the solution at 15 ℃ for crystallization, performing vacuum filtration, washing a filter cake with isopropyl acetate, pulping and washing with acetone, and reducing the DMSO solvent residue. Forced air drying at 50 ℃ (or vacuum drying at 50 ℃) to obtain 3.16g of light yellow solid (crystal form A). HPLC purity 100%, yield 88%, DMSO: <100 ppm; IPAc <100 ppm. MS m/z: 487.2[ M +1-MsOH ]. Melting point: 242 ℃ and 244 ℃.
Nuclear magnetic data (fig. 2):1HNMR(d6-DMSO):δppm:9.57(brs,1H),9.40(s,1H),8.71(s,1H),8.48(s,1H),8.32(d,1H,J=7.9),8.29(d,1H,J=5.3),7.96(s,1H),7.51(d,1H,J=8.2),7.23(ddd,1H,J=7.9,7.1,0.8),7.19(d,1H,J=5.4),7.15(ddd,1H,J=7.8,7.3,0.5),6.94(s,1H),6.67(dd,1H,J=16.9,10.2),6.27(dd,1H,J=16.9,1.8),5.57(dd,1H,J=16.9,1.7),4.44(t,2H,J=4.6),3.89(s,3H),3.88(s,3H),3.58(t,2H,J=4.6),2.93(s,6H),2.39(s,3H)。
through detection, the X-ray diffraction pattern of the crystal form A powder obtained in the embodiment has characteristic peaks at diffraction angle 2 theta values of 11.06 +/-0.2 degrees, 12.57 +/-0.2 degrees, 13.74 +/-0.2 degrees, 14.65 +/-0.2 degrees, 15.48 +/-0.2 degrees, 16.58 +/-0.2 degrees, 17.83 +/-0.2 degrees, 19.20 +/-0.2 degrees, 19.79 +/-0.2 degrees, 20.88 +/-0.2 degrees, 22.05 +/-0.2 degrees, 23.06 +/-0.2 degrees, 24.23 +/-0.2 degrees, 25.10 +/-0.2 degrees, 25.71 +/-0.2 degrees, 26.15 +/-0.2 degrees, 27.37 +/-0.2 degrees and 27.42 +/-0.2 degrees; the XRPD pattern is shown in figure 3 and attached table, the DSC pattern is shown in figure 4, the TGA pattern is shown in figure 5, and the infrared spectrum IR pattern is shown in figure 6.
Example 2
Figure BDA0002324528400000072
The compound of formula I (28.25g, 58.1mmol) was dissolved in 224ml dimethylsulfoxide DMSO solvent, warmed to 15-35 ℃ and the solution was stirred. To the system was added 0.97 equivalents of methanesulfonic acid (5.4g, 0.97mmol) in portions. 448ml of methyl isobutyl ketone (MIBK) are slowly added. Stirring for 1 hour, and then cooling to 10-15 ℃. The solution is subjected to salt forming reaction at 10-15 ℃, samples are taken, and HPLC (high performance liquid chromatography) is used for detecting the residue of the compound in the formula I in the mother solution (less than or equal to 0.4%). After the reaction, the crude product of the mesylate of the compound of the formula I is obtained by vacuum filtration, namely 32 g.
Adding 3g of the crude product of the mesylate of the compound shown in the formula I into 24ml of dimethyl sulfoxide DMSO solvent, stirring at 65 ℃ for clearing, cooling, slowly dropwise adding 48ml of methyl isobutyl ketone (MIBK), stirring for crystallization for 6-8 hours, carrying out suction filtration under reduced pressure, and carrying out forced air drying at 60 ℃ (or vacuum drying at 60 ℃) to obtain the target crystal form A. Melting point: 242 ℃ and 244 ℃. The XRPD pattern of the crystalline form is consistent with fig. 3 (fig. 7), with all characteristic peaks within error.
Example 3
Adding 3g of the crude mesylate of the compound of the formula I (prepared in reference example 2) into 24ml of dimethyl sulfoxide DMSO solvent, stirring at 65 ℃ for clearing, cooling, slowly dropwise adding 48ml of ethyl acetate, stirring for crystallization for 6-8 hours, carrying out vacuum filtration, washing a filter cake with ethyl acetate, and carrying out forced air drying at 70 ℃ (or vacuum drying at 60 ℃) to obtain 2.8g of a target crystal form A. Melting point: 242 ℃ and 244 ℃. The XRPD pattern of the crystal form is consistent with fig. 3, and all characteristic peaks are within the error range.
Example 4
Adding 3g of the crude product of the mesylate of the compound shown in the formula I into 24ml of dimethyl sulfoxide DMSO solvent, stirring at 65 ℃ for clearing, cooling, slowly dropwise adding 12ml of methyl tert-butyl ether (MTBK), stirring for crystallization for 6-8 hours, carrying out vacuum filtration, and carrying out forced air drying at 50 ℃ (or vacuum drying at 30 ℃) to obtain the target crystal form A. Melting point: 242 ℃ and 244 ℃. The XRPD pattern of the crystal form is consistent with fig. 3, and all characteristic peaks are within the error range.
Example 5
Adding 3g of the crude product of the mesylate of the compound shown in the formula I into 24ml of dimethyl sulfoxide DMSO solvent, stirring at 65 ℃ for clearing, cooling, slowly dropwise adding 48ml of Acetonitrile (ACN), stirring for crystallization for 6-8 hours, carrying out suction filtration under reduced pressure, and carrying out forced air drying at 50 ℃ or vacuum drying at 60 ℃ under reduced pressure to obtain the target crystal form A. Melting point: 242 ℃ and 244 ℃. The XRPD pattern of the crystal form is consistent with fig. 3, and all characteristic peaks are within the error range.
Example 6
Adding 3g of the crude product of the mesylate of the compound shown in the formula I into 24ml of dimethyl sulfoxide DMSO solvent, stirring at 65 ℃ for clearing, cooling, slowly dropwise adding 48ml of toluene (PhMe), stirring for crystallization for 6-8 hours, carrying out suction filtration under reduced pressure, and carrying out forced air drying at 50 ℃ or vacuum drying at 50 ℃ under reduced pressure to obtain the target crystal form A. Melting point: 242 ℃ and 244 ℃. The XRPD pattern of the crystal form is consistent with fig. 3, and all characteristic peaks are within the error range.
Example 7
Adding 10g of the crude product of the mesylate of the compound shown in the formula I into 100ml of tetrahydrofuran THF solvent, stirring at 65 ℃ for dissolving, cooling to room temperature, stirring for crystallization for 6-8 hours, filtering, and drying at 50 ℃ to obtain 8.8g of target crystal form A. Melting point: 242 ℃ and 244 ℃. The XRPD pattern of the crystal form is consistent with fig. 3, and all characteristic peaks are within the error range.
Example 8
Adding 2g of the crude product of the mesylate of the compound shown in the formula I into 20ml of N-methylpyrrolidone NMP solvent, stirring at 65 ℃ for dissolving, cooling to room temperature, stirring for crystallization for 6-8 hours, performing suction filtration under reduced pressure, and performing forced air drying at 50 ℃ (or vacuum drying at 50 ℃) to obtain the target crystal form A. Melting point: 242 ℃ and 244 ℃. The XRPD pattern of the crystal form is consistent with fig. 3, and all characteristic peaks are within the error range.
Example 9
Adding 2g of the crude product of the mesylate of the compound shown in the formula I into 30ml of N, N-dimethylformamide DMF solvent, stirring at 65 ℃ for dissolving, cooling to room temperature, stirring for crystallization for 6-8 hours, performing suction filtration under reduced pressure, and performing forced air drying (or vacuum drying at 60 ℃) at 65 ℃ to obtain the target crystal form A. Melting point: 242 ℃ and 244 ℃. The XRPD pattern of the crystal form is consistent with fig. 3, and all characteristic peaks are within the error range.
Example 10
Adding 2g of the crude product of the mesylate of the compound shown in the formula I into 50ml of N, N-dimethylacetamide DMAc solvent, stirring at 65 ℃ for dissolving, cooling to room temperature, stirring for crystallizing for 6-8 hours, carrying out suction filtration under reduced pressure, and carrying out forced air drying at 60 ℃ (or vacuum drying at 60 ℃) to obtain the target crystal form A. Melting point: 242 ℃ and 244 ℃. The XRPD pattern of the crystal form is consistent with fig. 3, and all characteristic peaks are within the error range.
Example 11
Adding 2g of the crude mesylate of the compound shown in the formula I into 50ml of an acetone/water mixed solvent (acetone/water is 1/1v/v), heating to 50 ℃, stirring and pulping, cooling to room temperature, stirring and crystallizing for 6-8 hours, performing suction filtration under reduced pressure, and performing forced air drying at 50 ℃ (or vacuum drying at 50 ℃) to obtain the target crystal form A. Melting point: 242 ℃ and 244 ℃. The XRPD pattern of the crystal form is consistent with fig. 3, and all characteristic peaks are within the error range.
Structure confirmation study of (tri) N- (2- (2- (dimethylamino) ethoxy) -4-methoxy-5- ((4- (1-methyl-1H-indol-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide mesylate (form a)
Form a prepared by the method of example 2 (purity greater than 99.80%) was tested for structure determination using the following example method.
Test example 1
And respectively testing the percentage content of C, H, N, S element contained in the crystal form A by adopting an element analysis method, testing twice in parallel, and taking the average value.
CHN test instrument: an Elementar Vario EL model III elemental analyzer (C, H, N measurement);
CHN test method: the percentage content of C, H, N is obtained by burning, decomposing, quantitatively converting, detecting and processing the data of the sample;
the S titration method comprises the following steps: precisely weighing crystal form A sample, respectively wrapping with ash-free filter paper, decomposing by combustion method, adding pure water and 30% H2O2Absorbed as an absorption liquid. After complete absorption, the flask was transferred to a 250mL volumetric flask, and the flask was rinsed with ethanol and the rinse was also transferred to a 250mL volumetric flask. Then adding a certain amount of HNO with known concentration3And an azochlorophosphine III indicator, followed by a known concentration of Ba (ClO)4)2·3H2O titrate until the solution changes color from purple to blue. Record Ba (ClO)4)2·3H2O consumption volume, calculate the percentage of S.
TABLE 1 elemental analysis test results for form A
Figure BDA0002324528400000101
The determination result shows that the crystal form A (C)27H30N6O3·CH4O3S) the difference between the observed value and the theoretical value of the C, H, N and the S content of the sample is less than 0.3%, indicating that the observed value is consistent with the theoretical value and is a salt with 1 molecule of methanesulfonic acid. In agreement with the data reported in example 2 of the earlier published patent CN201780050034.3 by this company.
Test example 2
The purpose is as follows: and testing the specific crystal structure of the crystal form A by adopting a powder X-ray diffraction analysis method.
The instrument comprises the following steps: bruker D8 advanced powder X-ray diffractometer
TABLE 2 test condition parameter table
Figure BDA0002324528400000111
Measurement and analysis: from the powder X-ray diffraction pattern of form A (FIG. 3 and the attached table), it can be seen that: the compound has a specific crystal structure. The angle 2 theta (degree) of the compound has stronger peaks (more than 10 percent of relative strength) at 11.06, 12.57, 13.74, 14.65, 15.48, 16.58, 17.83, 19.20, 19.79, 20.88, 22.05, 23.06, 24.23, 25.10, 25.71, 26.15, 27.37 and 27.42.
TABLE 3 comparison of example 2 of patent CN201780050034.3 disclosed earlier by this company
Different crystal forms of mesylate Total number of characteristic peaks Number of characteristic peaks with relative intensity greater than 10%
Crystal form A 44 18
2A 36 16
2B 12 12
Test example 3
The purpose is as follows: the melting point of form a was tested by differential thermal analysis DSC.
The instrument comprises the following steps: TAQ2000 differential calorimeter
Parameters are as follows: temperature rise range: 30-300 ℃; the heating rate is as follows: 10 ℃/min
Measurement and analysis: from the DSC curve (FIG. 4), the melting point of form A is 242-244 ℃.
TABLE 4 comparison with the mesylate salt of example 2 of patent CN201780050034.3
Different crystal forms of mesylate Melting Point
Crystal form A 242-244℃
2A 233.3-238.1℃
2B 157.3-180.2℃
Test example 4
The purpose is as follows: and testing whether the crystal form A contains water or solvent compounds by adopting a thermogravimetric analysis method.
The instrument comprises the following steps: TAQ5000 type thermogravimetric analyzer
Parameters are as follows: temperature rise range: 30-300 ℃; the heating rate is as follows: 10 ℃/min
Measurement and analysis: as can be seen from the TSA graph (figure 5), the weight loss of the crystal form A is only small at 33.40-120.00 ℃, and DSC and elemental analysis show that the crystal form A contains no water or solvent.
TABLE 5 comparison with the mesylate salt of example 2 of patent CN201780050034.3
Different crystal forms of mesylate Thermal weight loss (weight loss wt%)
Crystal form A -0.035%(33.4-120℃)
2A -0.435%(32.8-210.0℃)
2B -8.063%(29.7-193.4℃)
Test example 5
The purpose is as follows: and testing the infrared spectrogram of the crystal form A by adopting an infrared absorption spectrometry.
The instrument comprises the following steps: nicolet 380FT-IR type infrared spectrometer YP-2 tablet press
The method comprises the following steps: KBr pellet process
Measurement and analysis: as can be seen from the infrared absorption spectrum (figure 6), the product contains aromatic ring, aromatic heterocycle, alkenyl, -CH3, -CH2-, amino, amine salt, amide, ether group and sulfonate structure. The above infrared IR spectrum results are consistent with the structure of the crystal form A.
Test example 6
The purpose is as follows: the substance stability of the compound of the crystal form A prepared in the patent method example 2 is examined.
The method comprises the following steps: the stability data of the substance of the crystal form a are tested by referring to the requirements of the Chinese pharmacopoeia (2015 edition) on the stability test of the bulk drug and adopting the influence factors and the accelerated/long-term stability investigation method (the test design is shown in the following table 6).
The instrument comprises the following steps: a climatic condition cabinet; illumination box
TABLE 6 stability test design
Figure BDA0002324528400000131
Remarks are as follows: influential factor test samples at 40 ℃ were tested only if the test results at 60 ℃ did not meet the acceptance criteria. Samples at 25 ℃/75% RH were tested only if the 25 ℃/90% RH test did not meet the acceptance criteria.
TABLE 7 influence factor test results- -high temperature test
Figure BDA0002324528400000132
TABLE 8 influence factor test results-high humidity test
Figure BDA0002324528400000133
Figure BDA0002324528400000141
TABLE 9 influence factor test results- -light test
Figure BDA0002324528400000142
TABLE 10 Effect test results- -light test
Figure BDA0002324528400000143
TABLE 11 influence factor test results-Long term test
Figure BDA0002324528400000151
Measurement and analysis: influence factors, long-term and accelerated stability research data show that after the crystal form A is placed for 36 months for a long time, all detection indexes have no obvious change compared with 0 day, and all the detection indexes meet established standards and have no obvious adverse trend. The crystal form A prepared by the process has good stability under the existing packaging condition, and various detection indexes can meet the requirements of quality standards.
(IV) research on pharmaceutical properties of crystal form A
Test example 1
The purpose is as follows: comparative test for evaluating solubility of form A in different biological media
Experiment design: about 2.5mg of the compound of formula I and form A (mesylate salt of the compound of formula I) were weighed into 3 glass vials, and 200. mu.L of SGF, FaSSIF and FeSSIF were added to the vials, respectively. The suspension was stirred for about 30min at 37 ℃. (1) If a clear solution is obtained, the addition of the compound is continued until a suspension is obtained; (2) if not, the suspension is stirred for a further 24 h. At 24h, the pH was measured and the appearance visually inspected. If the suspension is still present after 24 hours, the solid is separated by centrifugation, the supernatant is diluted with 50% aqueous acetonitrile, and the dilution is then tested for its concentration by HPLC. The results are shown in the following table.
TABLE 12 comparison of the solubilities of Compound of formula I and form A in different biological media
Figure BDA0002324528400000161
And (4) conclusion: form a of the mesylate salt of the compound of formula I shows a solubility in biologically relevant media superior to that of the compound of formula I, especially in simulated gastric fluid SGF media with a significant improvement. The crystal form A is suitable for developing oral gastric soluble quick release solid preparation.
Test example 2
The purpose is as follows: evaluation of comparison of pharmacokinetic parameters after Single oral administration of form A and Compound of formula I in SD rats
Experiment design: healthy SD rats were randomly divided into a compound of formula I (free base) group and a mesylate salt form A group of the compound of formula I, both groups were male, 21 rats each, weighing 190-210 g. 60mg/kg of each test compound was administered orally in a single dose in the fasted state, whole blood was collected intravenously at different time points at 0.5, 1, 2, 4, 8, 12 and 24 hours after administration and plasma was separated, the drug concentration of form a or formula I compound in plasma was measured using liquid chromatography/tandem mass spectrometry (LC-MS/MS) and relevant pharmacokinetic parameters were calculated using a non-compartmental model. The main pharmacokinetic parameters are shown in the following table:
TABLE 13 Main pharmacokinetic parameters of Compound group I and Compound mesylate form A of formula I
Pharmacokinetic parameters Free base Crystal form A
Cmax(ng/mL) 496 622
Tmax(h) 4.00 2.00
T1/2(h) 5.02 3.31
AUC0-24(ng·h/ml) 5899 6754
AUC0-∞(ng·h/ml) 6236 6846
MRT0-24(h) 8.29 7.46
MRT0-∞(h) 9.53 7.75
CmaxThe maximum concentration of drug in plasma; t ismax: the peak time of the highest concentration of the drug; t is1/2: elimination of half-life; AUC is the area under the blood concentration-time curve; MRT mean residence time of drug
From the above table and fig. 8, it can be concluded that: the bioavailability of form a is significantly better than that of the free base.
Test example 3
The purpose is as follows: test for evaluating pharmaceutical activity of crystal form A as lung cancer drug
1) Inhibition and selection specificity assays for specific cell growth
The method comprises the following steps: and detecting whether the crystal form A has antiproliferative activity on human epidermal carcinoma cells (A431, wild type EGFR), human lung cancer cells (HCC827, EGFR 19 exon deletion type activation mutation) and human lung cancer cells (H1975, EGFR L858R/T790M drug-resistant type mutation) by adopting a fluorescent cell proliferation technology and a cell microscope visualization detection technology (Alma blue method).
Experiment design: the cells in the logarithmic growth phase were seeded in a 96-well plate (cell concentration: 2000/well; cell suspension: 200. mu.l/well) at 30 ℃ with 5% CO2Cells were allowed to adhere for 24 hours in culture. The DMSO solutions of test compounds were diluted with the cell culture medium solution from high to low to different solubilities, 3-fold each time, to give a total of 9 different concentrations. Test drug solutions of different concentrations were added to a 96-well cell plate containing the above cancer cells, with 3 duplicate wells for each concentration. A cell control well containing DMSO only cell culture medium was also provided. Continuing at 37 ℃ with 5% CO2After culturing for 72 hours, the culture was terminated.
Cell proliferation was detected after staining with Alamar Blue reagent (resazurin). Resazurin (resazurin) is reduced into a resorufin reagent (resorufin) by cells to become a fluorescent substance (544nm excitation, 612nm color development), and the fluorescence intensity is in direct proportion to the number of cells. The resazurin reagent was dissolved in PBS solution and prepared as a stock solution at a concentration of 440. mu.M. After 5 hours of incubation, the cell culture plates were returned to normal culture conditions and fluorescence measurements were read after 72 hours using a staining 3 multimode microplate reader (Biotek).
Fluorescence measurements were normalized to cell-free (background) well readings and the average of total growth over 72 hours was determined to vehicle control wells (n-3). The results are shown in the following table:
TABLE 14 results of experiments on inhibition and selection specificity of form A and oxitinib for specific cell growth
Figure BDA0002324528400000171
Data source: table 4, P21, PHARMACOLOGY REVIEW (S), APPLICATION NUMBER 208065Orig1S000, CDER of FDA;https://www.accessdata.fda.gov/drugsatfda_docs/ nda/2015/208065Orig1s000PharmR.pdf
the results show that the compound of the crystal form A has excellent activity of inhibiting the proliferation and growth of cancer cells for H1975 and HCC827, has no obvious inhibition on the growth of wild cells, and is equivalent to a product (oxitinib) on the market abroad; but form a showed superior selectivity of inhibition (relative to wild-type a431 cancer cells) for H1975 and HCC827 cells.
2) Inhibition selectivity for different kinase targets
Kinase inhibitors are used as drugs for clinical treatment, and it is necessary to avoid the inhibition of some non-targeted kinases in addition to targeted kinases, and possibly to induce some cytotoxic reactions (so-called "off-target effects"). This experiment was performed by KINOMEscan's in vitro competitive binding assay*1For 97 kinases (andhuman disease associated mutant site kinase profiling) was subjected to high throughput screening (kinase profiling).
TABLE 15 results of high throughput screening of 97 kinase zymograms by form A and oxitinib
Figure BDA0002324528400000181
*1In vitro binding assays reference is made in detail to: fabian, M.A. et al.A. small molecule-kinase interaction map for clinical kinase inhibitors Nat.Biotechnol.23,329-336(2005).
*2The data source is as follows: p15, P43, PHARMACOLOGY REVIEW (S), APPLICATION NUMBER 208065Orig1S000, CDER of FDA;https://www.accessdata.fda.gov/drugsatfda_docs/ nda/2015/208065Orig1s000PharmR.pdf
the results show that the selectivity of the inhibitory activity of the non-target kinase of the compound in the crystal form A is better than that of the marketed drug of oxitinib.
Test example 4 evaluation of pharmaceutical safety of crystalline form a
1) Safety pharmacological test
The purpose is as follows: the clinical medication route is simulated, and the influence on the central nervous system of an SD rat, the respiratory system of the SD rat and the cardiovascular system of a beagle dog is observed after the oral administration preparation prepared by the single oral administration of the crystal form A. The potential undesirable adverse physiological (central nervous system, respiratory system and cardiovascular system) effects of the crystalline form a compound at doses within or above the therapeutic range were investigated.
The method comprises the following steps:
[ Central nervous experiment design ] SD rats of 7 weeks of age, half male and female, and between the weight of 170-. Animals were given a single oral dose of vehicle (0.5% (w/v) methylcellulose), 10, 40 or 160mg/kg of a suspension of form a compound dissolved in 1% (w/v) methylcellulose (vehicle), with vehicle alone in control animals. All animals were dosed at a volume of 10 mL/kg. The effect of different doses of the test article on the central nervous system function of an animal is assessed using a functional Observation combination (FOB) comprising tests on the animal's motor function, behavioral changes, coordination function, sensory/motor reflexes and body temperature. FOB observations were made before the experiment, about 2 hours, 4 hours and 24 hours after dosing.
[ design of respiratory System experiment ] SD rats of 7-8 weeks old, half male and female, with a body weight of 168-287g, were divided into 4 groups of 40 rats (20 male and female) each, and 5 male and female in each group. Animals were given a single oral dose of vehicle (0.5% (w/v) methylcellulose), 10, 40 or 160mg/kg of a suspension of form a compound dissolved in 0.5% (w/v) methylcellulose (vehicle), with vehicle alone in control animals. All animals were dosed at a volume of 10 mL/kg. On the day of dosing, animals were first acclimated in an "open-head" plethysmograph for at least 5 minutes prior to data acquisition, and immediately following acclimation, respiratory data (respiratory rate, tidal volume, minute ventilation) were acquired at time points of about 2 hours (± 10min), 4 hours (± 10min) and 24 hours post-dose prior to dosing, with each acquisition lasting about 15 minutes in duration. Changes in tidal volume, respiratory volume, and respiratory rate before and after dosing were recorded. Time points of 0h, 2h, 4h and 24h are set. Each index after administration was compared to its own control and subjected to statistical analysis.
[ cardiovascular System Experimental design ] A22-24-month-old beagle dog, half a beagle dog, 9.3-10.5kg of body weight, 6 beagle dogs in total, is pre-implanted into a remote sensing sub-device. The vehicle alone was administered to control animals in a dose escalating fashion, with each dog being given 1 vehicle and 10, 30, 100mg/kg of a form a compound suspension dissolved in 0.5% (w/v) methylcellulose (vehicle). The interval between the two doses was 3 days (except for the control, which was 2 days). The volume administered was 5ml/kg, the actual dose administered was based on the animal's most recent weight weighed. Electrocardiographic data are collected before administration, 2 hours, 4 hours, 6 hours, 8 hours and 24 hours after administration using telemetry, which is obtained for veterinary evaluation by an electrocardiograph.
As a result: the oral crystal form A has low toxicity, is negative to test results of a central nervous system, a respiratory system and a cardiovascular system of experimental animals, and has small adverse reaction and higher safety.
2) Genotoxicity test
The purpose is as follows: genotoxicity assays can be used to detect somatic cell mutagens, germ cell mutagens, and potential carcinogens. The method combining in vitro and in vivo genotoxicity tests is adopted, and three tests, namely a bacterium (including salmonella typhimurium and escherichia coli) back mutation in vitro test (Ames test), a Chinese hamster ovary cell chromosome aberration in vitro test and a rat gavage administration bone marrow micronucleus in vivo test, are selected to comprehensively evaluate the potential genotoxicity of the crystal form A administration preparation.
The method comprises the following steps:
the potential mutagenic capacity of the crystal form a compound was evaluated by testing its ability to induce reversion of selected four histidine-deficient salmonella typhimurium (TA98, TA100, TA1535 and TA1537) and tryptophan-deficient Escherichia Coli WP2 uvrA with and without the addition of exogenous metabolic activation system (Aroclor 1254-induced rat liver S9) at different doses. And a negative/solvent control group and a positive control group are also arranged. The experimental dose was set as: the series of additions and subtractions of S9 for TA98, TA100, TA1535 and TA1537 were 10, 25, 50, 100, 250 and 1000. mu.g/dish; the series of additions and deletions of S9 for WP2 uvrA were 100, 250, 500, 1000, 2500 and 5000. mu.g/dish.
Chromosome aberration experimental design the ability of chinese hamster ovary cells (CHO-WBL) to produce chromosome aberrations was determined by testing different doses of the crystalline form a compound with and without the addition of an exogenous metabolic activation system (Aroclor1254 induced rat liver S9). Adding S9 to activate the experiment system, and treating the cells for 3 hours; cells were dosed for 3 and 22 hours without the addition of the S9 assay system. After 22 hours of culture after dosing, all cells were harvested. And a negative/solvent control group and a positive control group are also arranged. The experimental dose was set as: the activation system added with S9 is as follows: 2.5, 12 and 14. mu.g/ml; the inactive lines were 1, 3 and 6. mu.g/ml without S9.
[ design of bone marrow micronucleus experiment ] A total of 46 male rats between 7-8 weeks old and 241-306g in weight were randomly divided into 7 groups of 6 animals (5 &6 groups of 8). Vehicle (0.5% (w/v) methylcellulose) (groups 1, 2), 500, 1000, 2000, or 2000mg/kg of crystalline form a compound suspension dissolved in 0.5% (w/v) methylcellulose (vehicle) (groups 3-6), and 20mg/kg of positive control drug (a physiological saline solution of cyclophosphamide monohydrate, administered by a single intraperitoneal injection) (group 7) were each administered orally in a single dose. All animals were dosed at a volume of 10 mL/kg. 1. Animals in groups 3, 4, 5 and 7 were sacrificed at about 24 hours after the administration, and animals in groups 2 and 6 were examined for the formation rate of micronucleated pleochromocytes (MN-PCE) in their bone marrow after about 48 hours after the administration.
As a result: the compound of the crystal form A has no genotoxicity (Ames test, mouse micronucleus test and chromosome aberration test) in all genotoxicity studies, and the results are all negative. It can be concluded that form a has no mutagenicity and obvious genetic hazard. The crystal form A has no obvious genotoxicity and mutagenic effect.
Clinical application example 1 human clinical trial of crystalline form a
The purpose is as follows: evaluating the safety and curative effect of the oral preparation prepared from the crystal form A on the administration in Chinese patients with locally advanced or metastatic non-small cell lung cancer after EGFR-TKI treatment.
Materials and methods: capsules prepared by the crystal form A (auxiliary materials are microcrystalline cellulose, lactose, croscarmellose sodium, colloidal silicon dioxide and magnesium stearate; and a No. 3 gelatin hollow capsule is directly filled and filled according to the required medicament dosage after being uniformly mixed) are adopted, and 128 cases of patients with non-small cell lung cancer are added in total to evaluate the safety and the curative effect.
As a result: clinical safety studies after single and multiple oral administrations of 6 dose groups of 30, 60, 120, 180, 240, 300mg, etc. have been completed, and the results show that doses of 30mg-300mg are safe.
The middle-stage efficacy analysis of clinical trials shows that from 30-300mg of QD once a day, a minimum initial dose of 30mg shows efficacy (disease control rate DCR 80.0%), and a recommended phase II dose of 180mg has a disease remission rate ORR of 73.1%, which is higher than that of the marketed drug oxitinib (66.1%); the disease remission rate DCR is 96.2 percent and is also higher than the marketed drug oxitinib (91 percent). The specific efficacy evaluations are as follows:
TABLE 16 evaluation of therapeutic efficacy of different dose groups
Dosage form 30mg 60mg 120mg 180mg 240mg 300mg Total
Number of cases to be grouped 10 6 26 52 3 2 128
CR 0 0 0 0 0 0 0
PR 4(40.0) 1(16.7) 16(61.5) 29(55.8) 11(34.4) 1(50.0) 62(48.4)
SD 4(40.0) 3(50.0) 6(23.1) 12(23.1) 13(40.6) 1(50.0) 39(30.5)
PD 2(20.0) 1(16.7) 2(7.7) 2(3.8) 1(3.1) 0 8(6.3)
ORR,n(%) 4(40.0) 2(33.3) 18(69.2) 38(73.1) 18(56.3) 1(50.0) 81(63.3)
DCR,n(%) 8(80.0) 5(83.3) 24(92.3) 50(96.2) 31(96.9) 2(100) 120(93.8)
The terms referred to above, are to be interpreted with the precision: CR: complete remission, PR: partial remission, SD: disease stability, PD: disease progression, ORR: objective remission rate, DCR: the rate of disease control.
The mid-term efficacy analysis of clinical trials shows that from 30-300mg of QD once a day, the incidence (proportion and variety) of adverse reactions is significantly lower than that of the marketed drug, oxitinib. The specific safety evaluations are as follows:
TABLE 17 comparison of AE incidence rates of adverse reactions of crystal form A and oxitinib
Figure BDA0002324528400000211
Figure BDA0002324528400000221
TABLE 18 comparative listing of adverse event TEAE species greater than 10% for form A and Oxitinib
Figure BDA0002324528400000222
Data source: page 4, the instructions for use of oxitinib mesylate tablet and Page 89, medicine&STATISTICAL REVIEW(S),APPLICATION NUMBER:208065Orig1s000,CDER ofFDA;https:// www.accessdata.fda.gov/drugsatfda_docs/nda/2015/208065Orig1s000MedR.pdf
Clinical application example 2 application of drug combination study of crystal form a
The purpose is as follows: the inhibiting effect of the single medicine or the combined medicine of the crystal form A prepared in the embodiment 2 and the compound BPI-7722 independently designed by the company on the human lung cancer H1975 nude mouse subcutaneous transplantation tumor is observed.
Materials and methods:
cell culture: h1975 (human Lung adenocarcinoma) was purchased from cell banks of Chinese academy of sciences and cultured in CO2A constant temperature incubator. The cell culture medium is modified RPMI; 10% fetal bovine serum; 100U/ml penicillin and 100. mu.g/ml streptomycin. Cells were digested with 0.25% pancreatin and passaged every other day. After the cells have expanded to the desired cell number and the cells are in the logarithmic growth phase, cell counts are collected for seeding.
Experimental animals: BALB/c nude male mice, 24, 6-7 weeks, 16-18g, 6 per group, were purchased from Shanghai Ling Biotech, Inc. The tumor volume of the nude mice is selected to be 80-254mm 21 days after H1975 cell inoculation 324 mice, randomly divided into 4 groups of 6 animals each based on tumor volume and body weight. Respectively as follows: group 1: a 0.5% sodium carboxymethylcellulose solvent control group; group 2: example 2 single drug group of form a (5 mg/kg); group 3: BPI-7722(100mg/kg) single drug group; group 4: form A (5mg/kg)/BPI-7722(25mg/kg) combination.
The experimental method comprises the following steps: human lung cancer H1975 cell line (5X 10)6One mouse/mouse) is inoculated in the subcutaneous part of the right back of an experimental mouse, the growth condition of the tumor is regularly observed, the diameter of the transplanted tumor is measured by a vernier caliper, and after 21 days of inoculation, the tumor grows to be average (80-254 mm)3) Groups were randomized according to tumor size and mouse weight. The administration volume of each group is 10ml/kg according to the latest weight of the mice. Once daily for 21 days. Whole solidIn the course of the test, the body weight and tumor size of the mice were measured three times per week to observe whether or not a toxic reaction occurred. Tumor volume calculation formula: tumor volume TV (mm)3) 0.5 × (tumor major diameter × tumor minor diameter)2). The tumor growth curves of the experimental groups are shown in the following table and the attached figure 9, and the body weight curves are shown in the following table and the attached figure 10.
TABLE 19 summary of mean body tumor volume versus time for each dose group
Figure BDA0002324528400000231
Blank group animals had been euthanized due to tumor overload, so no data collection was performed on day 21.
TABLE 20 summary of mean body weight-time relationship for each dose group
Figure BDA0002324528400000232
Blank group animals had been euthanized due to tumor overload, so no data collection was performed on day 21.
And (4) conclusion: the single medicine of the crystal form A compound of the embodiment 2 has good inhibition effect on the growth of subcutaneous transplantation tumor of a human lung cancer H1975 nude mouse; after the combination of BPI-7722, the inhibition effect is obviously better than the growth inhibition effect of single drug; and exhibits good safety.
Although the present invention has been described to a certain extent by the embodiments thereof with reference to the accompanying drawings, it is to be noted that various modifications, alterations and adaptations of the present invention may occur to one skilled in the art without departing from the spirit and scope of the present invention after reading the foregoing disclosure of the present application, and such changes and adaptations are intended to be included within the scope of the appended claims.

Claims (9)

1. A crystalline form a of an aminopyrimidine mesylate salt, wherein the aminopyrimidine mesylate salt has a structure represented by the formula:
Figure FDA0002822050290000011
and
the XRPD pattern of form a is shown in figure 3.
2. The aminopyrimidine mesylate form A of claim 1 having an IR spectrum according to FIG. 6.
3. A process for preparing form a of the mesylate salt of an aminopyrimidine according to claim 1 or 2 comprising: dissolving the aminopyrimidine compound mesylate by using an organic solvent, stirring, crystallizing, filtering and drying to obtain the crystal form A, wherein the organic solvent comprises dimethyl sulfoxide, tetrahydrofuran, N-methylpyrrolidone, N-dimethylformamide and N, N-dimethylacetamide.
4. The method of manufacturing according to claim 3, further comprising: preparing an aminopyrimidine compound and methanesulfonic acid according to a feeding molar ratio of 1.05-0.97 to obtain a methanesulfonic acid salt of the aminopyrimidine compound, wherein the aminopyrimidine compound has the following formula:
Figure FDA0002822050290000021
5. the method according to claim 3, wherein the crystallization is carried out by adding an anti-solvent selected from the group consisting of methyl t-butyl ether, isopropyl acetate, ethyl acetate, methyl isobutyl ketone, acetonitrile, toluene, acetone and water.
6. The method according to claim 3, wherein the filtration is followed by drying at 30 to 70 ℃ under normal pressure or vacuum.
7. A pharmaceutical composition comprising crystalline form a of the mesylate salt of an aminopyrimidine compound of claim 1 or 2 and a pharmaceutically acceptable carrier.
8. The use of the crystalline form a of the mesylate of an aminopyrimidine compound according to claim 1 or 2 or the pharmaceutical composition according to claim 7 for preparing an anti-tumor medicament.
9. The use of claim 8, wherein the tumor is lung cancer.
CN201911311034.1A 2019-12-18 2019-12-18 Crystal form A of aminopyrimidine compound mesylate and preparation method and application thereof Active CN111100117B (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CN201911311034.1A CN111100117B (en) 2019-12-18 2019-12-18 Crystal form A of aminopyrimidine compound mesylate and preparation method and application thereof
PCT/CN2020/135661 WO2021121146A1 (en) 2019-12-18 2020-12-11 Crystal form a of aminopyrimidine mesylate compound, preparation method therefor, and application thereof
TW109145163A TW202128666A (en) 2019-12-18 2020-12-18 Crystal form a of aminopyrimidine compound mesylate and preparation method and application thereof
ARP200103555A AR120821A1 (en) 2019-12-18 2020-12-18 CRYSTALLINE FORM A OF THE METANSULFONATE SALT OF AN AMINOPYRIMIDINE COMPOUND AND METHOD OF PREPARATION AND APPLICATION OF THE SAME

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201911311034.1A CN111100117B (en) 2019-12-18 2019-12-18 Crystal form A of aminopyrimidine compound mesylate and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN111100117A CN111100117A (en) 2020-05-05
CN111100117B true CN111100117B (en) 2021-02-19

Family

ID=70422824

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201911311034.1A Active CN111100117B (en) 2019-12-18 2019-12-18 Crystal form A of aminopyrimidine compound mesylate and preparation method and application thereof

Country Status (4)

Country Link
CN (1) CN111100117B (en)
AR (1) AR120821A1 (en)
TW (1) TW202128666A (en)
WO (1) WO2021121146A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111100117B (en) * 2019-12-18 2021-02-19 上海倍而达药业有限公司 Crystal form A of aminopyrimidine compound mesylate and preparation method and application thereof
WO2023287783A1 (en) * 2021-07-13 2023-01-19 ACEA Therapeutics, Inc. Heterocyclic compounds and uses thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3191469B1 (en) * 2014-07-24 2021-12-15 Beta Pharma, Inc. 2-h-indazole derivatives as cyclin-dependent kinase (cdk) inhibitors and therapeutic uses thereof
CN111170998B (en) * 2014-11-05 2023-04-11 益方生物科技(上海)股份有限公司 Pyrimidine or pyridine compound, preparation method and medical application thereof
CN109715164A (en) * 2016-05-11 2019-05-03 贝达医药公司 2- anilino-pyrimidine derivative as the therapeutic agent for treating the cancer of the brain
CN109937043A (en) * 2016-06-17 2019-06-25 贝达医药公司 The drug salts and its crystal form of N- (2- (2- (dimethylamino) ethyoxyl) -4- methoxyl group -5- ((4- (1- Methyl-1H-indole -3- base) pyrimidine -2-base) amino) phenyl) acrylamide
JP7045726B2 (en) * 2017-06-16 2022-04-01 ベータ ファーマ,インコーポレイテッド N- (2- (2- (dimethylamino) ethoxy) -4-methoxy-5-((4- (1-methyl-1H-indole-3-yl) pyrimidin-2-yl) amino) phenyl) acrylamide and Pharmaceutical formulation of the salt
CN111100117B (en) * 2019-12-18 2021-02-19 上海倍而达药业有限公司 Crystal form A of aminopyrimidine compound mesylate and preparation method and application thereof

Also Published As

Publication number Publication date
WO2021121146A1 (en) 2021-06-24
AR120821A1 (en) 2022-03-23
TW202128666A (en) 2021-08-01
CN111100117A (en) 2020-05-05

Similar Documents

Publication Publication Date Title
CN109963844B (en) Compound for inhibiting and degrading tyrosine protein kinase ALK
EP3312180B1 (en) Use of pteridinone derivative serving as egfr inhibitor
CN107531678B (en) EGFR inhibitor and pharmaceutically acceptable salts and polymorphs thereof and uses thereof
US20090093495A1 (en) Combination of a src kinase inhibitor and a bcr-abl inhibitor for the treatment of proliferative diseases
CN112010839B (en) Crystalline forms of a targeted silk/threonine kinase inhibitor
US7166722B2 (en) N-{2-chloro-4-[(6,7-dimethoxy-4-quinolyl)oxy]phenyl}-n′-(5-methyl-3-isoxazolyl)urea salt in crystalline form
CN111100117B (en) Crystal form A of aminopyrimidine compound mesylate and preparation method and application thereof
CN113861191B (en) Crystal forms of CDK4/6 active compound and application thereof
CN113966332B (en) Polymorphs of a CDK9 inhibitor, methods of making and using the same
CN116768856A (en) Salt of substituted amino six-membered nitrogen heterocyclic compound, crystal form, preparation method and application thereof
CN111362924B (en) Deuterated pyrimidine derivatives and uses thereof
JP2023543281A (en) Salts of arylaminoquinazoline-containing compounds, and their preparation and use
CN109438279B (en) Small molecule compound for overcoming EGFR drug-resistant mutation and preparation method and application thereof
CN104119321B (en) The 2-maleate and its polymorph of indolinone derivative
CN101003513A (en) Compound in quinazoline class or its pharmaceutical salt preparation method, and medical usage
WO2024067085A1 (en) Citrate salt of cyclin-dependent kinase (cdk4/6) inhibitor, crystal form thereof, preparation method therefor and use thereof
WO2021143819A1 (en) Crystal form of polycyclic anaplastic lymphoma kinase inhibitor
WO2024027565A1 (en) Crystal form of spirocyclic amine aryl phosphorus oxide compound and preparation method therefor
WO2024097848A1 (en) Malonate and glycolate salts of an egfr inhibitor
KR20230152118A (en) Drug composition and its preparation method and use
CN118146164A (en) Hydrazone-containing quinazoline derivative or pharmaceutically acceptable salt thereof, 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
CB03 Change of inventor or designer information
CB03 Change of inventor or designer information

Inventor after: Tang Chun

Inventor after: M.N. Greco

Inventor after: M.J. Costanzo

Inventor after: Zhang Xiaoxia

Inventor after: J.Peng

Inventor after: D.Zhang

Inventor after: Y.Lu

Inventor before: Tang Chun

Inventor before: M.N. Greco

Inventor before: M.J. Costanzo

Inventor before: Zhang Xiaoxia

Inventor before: J.Peng

Inventor before: D.Zhang

TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20210122

Address after: 201203 Block H, 5th floor, 780 Cailun Road, Pudong New Area pilot Free Trade Zone, Shanghai

Applicant after: BETA PHARMA (SHANGHAI) Co.,Ltd.

Address before: Room d411, building D, 398 Ruoshui Road, Suzhou Industrial Park, 215000, Jiangsu Province

Applicant before: BEIERDA PHARMACY (SUZHOU) Co.,Ltd.

GR01 Patent grant
GR01 Patent grant
PE01 Entry into force of the registration of the contract for pledge of patent right
PE01 Entry into force of the registration of the contract for pledge of patent right

Denomination of invention: Crystal form A of amino pyrimidine compound methanesulfonate and its preparation method and application

Effective date of registration: 20230926

Granted publication date: 20210219

Pledgee: Bank of Shanghai Limited by Share Ltd. Pudong branch

Pledgor: BETA PHARMA (SHANGHAI) Co.,Ltd.

Registration number: Y2023310000612

PC01 Cancellation of the registration of the contract for pledge of patent right
PC01 Cancellation of the registration of the contract for pledge of patent right

Granted publication date: 20210219

Pledgee: Bank of Shanghai Limited by Share Ltd. Pudong branch

Pledgor: BETA PHARMA (SHANGHAI) Co.,Ltd.

Registration number: Y2023310000612

PE01 Entry into force of the registration of the contract for pledge of patent right
PE01 Entry into force of the registration of the contract for pledge of patent right

Denomination of invention: Crystal Form A of Aminopyrimidine Compound Methanesulfonate and Its Preparation Method and Application

Granted publication date: 20210219

Pledgee: Bank of Shanghai Limited by Share Ltd. Pudong branch

Pledgor: BETA PHARMA (SHANGHAI) Co.,Ltd.

Registration number: Y2024310000902