CN115385923A - Crystal form of oxygen-containing heterocyclic compound, preparation method and application thereof - Google Patents

Crystal form of oxygen-containing heterocyclic compound, preparation method and application thereof Download PDF

Info

Publication number
CN115385923A
CN115385923A CN202210560179.0A CN202210560179A CN115385923A CN 115385923 A CN115385923 A CN 115385923A CN 202210560179 A CN202210560179 A CN 202210560179A CN 115385923 A CN115385923 A CN 115385923A
Authority
CN
China
Prior art keywords
formula
compound
cancer
solvent
degrees
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.)
Pending
Application number
CN202210560179.0A
Other languages
Chinese (zh)
Inventor
许祖盛
楼杨通
唐佑海
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.)
Shanghai Yingli Pharmaceutical Co Ltd
Original Assignee
Shanghai Yingli 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 Shanghai Yingli Pharmaceutical Co Ltd filed Critical Shanghai Yingli Pharmaceutical Co Ltd
Publication of CN115385923A publication Critical patent/CN115385923A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/052Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being six-membered
    • 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

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

Abstract

The invention discloses a crystal form of an oxygen-containing heterocyclic compound, and a preparation method and application thereof. The crystal form of the compound is a crystal form A of the compound shown in the formula (I), a crystal form B of the compound shown in the formula (I) and a crystal form C of the compound solvate shown in the formula (I). The crystal form disclosed by the invention is simple in preparation method, suitable for industrial production, not easy to absorb moisture, good in stability and beneficial to preparation of a preparation and long-term storage of a medicament.
Figure DDA0003656248880000011

Description

Crystal form of oxygen-containing heterocyclic compound, preparation method and application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to a crystal form of an oxygen-containing heterocyclic compound, and a preparation method and application thereof.
Background
Ras (Rat sarcoma viral oncogene), the earliest of which was found in Rat sarcoma. There are three members of the mammalian ras gene family, H-ras, K-ras, and N-ras, in which the fourth exon of K-ras has two variants, A and B. Ras gene is widely present in various eukaryotes such as mammals, fruit flies, fungi, nematodes and yeasts, and is expressed to various degrees in different tissues, among which H-Ras is mainly expressed in skin and skeletal muscle, K-Ras is mainly expressed in colon and thymus, and N-Ras is expressed to a high degree in testis. Ras protein is used as a molecular switch in the cell signal transduction process, and regulates and controls signal transduction by combining with GTP/GDP for switching, thereby regulating the life processes of cell proliferation, differentiation, aging, apoptosis and the like.
Ras mutations are closely associated with the development and progression of tumors. Ras is mutated in more than 30% of human tumors and is considered one of the most powerful cancer drivers. Ras proto-oncogene mutation is mainly performed by means of point mutation. More than 150 different Ras point mutations have been found, with mutations at glycine 12 and 13 and glutamine 61 being the most common.
For decades, efforts have been made to develop small molecule inhibitors targeting Ras, however, the development of related drugs has progressed slowly. Scientists have hoped to develop competitive inhibitors of GTP that act directly on Ras proteins, but this has not been successful because of the strong affinity between GTP and Ras (pmol/L scale), the high concentration of GTP in cells (0.5 mM), and the lack of pockets in the Ras protein structure that facilitate small molecule binding. In recent years, people use K-Ras G12C mutant allosteric site for drug development has made some progress. In 2013, a research group reported the discovery of K-Ras G12C small molecule inhibitors (Nature, 2013,503, 548-551). They identified a novel binding pocket from the K-Ras G12C mutant located below the region of molecular switch II, to which these inhibitors bind and form covalent bonds with nearby Cys12, thereby selectively inhibiting K-Ras G12C activation. Other researchers have reported KRas inhibitors with cellular activity (Science, 2016,351, 604-608). The compound sotoraib (AMG 510) from Amgen binds to cysteine thiol group 12 of KRAS-G12C mutant via acrylamide Michael addition acceptor structure, locking the G12C mutant KRAS protein in an inactive GDP binding state to specifically and irreversibly inhibit its pro-proliferative activity. Sotoraib obtained FDA approval to market at 2021 month 5 and became the first global targeted therapy for treating patients with locally advanced or metastatic non-small cell lung cancer (NSCLC) carrying KRas G12C mutation.
It is well known to those skilled in the art that chemical drugs exhibit polymorphism due to the different periodic arrangement of their internal particles, and that different solid forms of the same drug, including amorphous and crystalline forms, can exhibit widely different physical, chemical and spectroscopic properties, such as differences in melting point, hygroscopicity, dissolution rate, solubility, processability and stability, which in turn leads to pharmaceutical products that also exhibit differences in safety and efficacy. Therefore, there are many practical benefits to conducting an exhaustive study of drug polymorphism and to evaluate the physicochemical properties of different solid forms of a drug, selecting the appropriate solid form for drug development.
An oxygen-containing heterocyclic compound, the structure of which is shown in formula (I)
Figure BDA0003656248860000021
(hereinafter, referred to as a compound represented by the formula (I)), can be used for treating and/or preventing various diseases mediated by Ras. The compound is independently designed and synthesized by the inventor
Figure BDA0003656248860000022
Chiral resolution of an OJ column (20X 250mm,10 μm; brand Daicel) produced a solid form which was determined to be amorphous by XRPD. It has been found that the compounds of formula (I) have the following disadvantages when present in the amorphous solid form: the moisture absorption performance is high; has poor stability, and particularly has obviously reduced purity under high temperature and illumination。
In view of the fact that the potential for developing the compound shown as the formula (I) into a medicament is poor when the compound shown as the formula (I) exists in an amorphous solid form, the development of a novel crystal form of the compound shown as the formula (I) with more advantageous properties has very important practical significance.
Disclosure of Invention
The invention aims to solve the technical problems of single solid form, high hygroscopicity and poor stability of an oxygen-containing heterocyclic compound in the prior art, and provides a crystal form of the oxygen-containing heterocyclic compound, and a preparation method and application thereof. The crystal forms are simple in preparation method, suitable for industrial production, not prone to moisture absorption, good in stability and beneficial to preparation of preparations and long-term storage of medicines.
The present invention solves the above-mentioned problems by the following technical means.
The invention provides a crystal form A of a compound shown as a formula (I), which has an X-ray powder diffraction pattern expressed by a 2 theta angle, and has characteristic peaks at 4.8 +/-0.2 degrees, 12.4 +/-0.2 degrees, 15.6 +/-0.2 degrees and 23.3 +/-0.2 degrees, wherein the X-ray powder diffraction pattern is measured by using a K alpha spectral line of a Cu target;
Figure BDA0003656248860000023
in some embodiments, the crystalline form a of the compound of formula (I), having an X-ray powder diffraction pattern expressed in terms of 2 Θ angles, may have characteristic peaks at 4.8 ± 0.2 °, 9.1 ± 0.2 °, 9.7 ± 0.2 °, 12.4 ± 0.2 °, 15.6 ± 0.2 °, 19.8 ± 0.2 °, 20.4 ± 0.2 °, 21.1 ± 0.2 °, 23.3 ± 0.2 °, 24.7 ± 0.2 ° and 26.3 ± 0.2 °.
In some embodiments, the crystalline form a of the compound of formula (I) has characteristic peaks at 4.8 ± 0.2 °, 9.1 ± 0.2 °, 9.7 ± 0.2 °, 12.4 ± 0.2 °, 14.5 ± 0.2 °, 15.6 ± 0.2 °, 16.5 ± 0.2 °, 17.2 ± 0.2 °, 17.5 ± 0.2 °, 19.8 ± 0.2 °, 20.4 ± 0.2 °, 21.1 ± 0.2 °, 21.4 ± 0.2 °, 22.1 ± 0.2 °, 22.4 ± 0.2 °, 23.3 ± 0.2 °, 24.7 ± 0.2 °, 26.3 ± 0.2 °, 27.6 ± 0.2 ° and 27.9 ± 0.2 ° in its X-ray powder diffraction pattern, expressed in terms of 2 Θ angle.
In some embodiments, the compound of formula (I) is in crystalline form a, having an X-ray powder diffraction pattern expressed in terms of 2 Θ angles, and further having diffraction peaks and percentages of peak area as follows:
numbering 2θ(±0.2°) Percentage of Peak area (%)
1 4.839 100.0
2 9.141 40.6
3 9.694 21.5
4 12.433 37.6
5 14.451 26.0
6 14.644 8.6
7 15.644 84.3
8 16.545 8.4
9 17.249 41.7
10 17.456 9.9
11 19.801 36.8
12 20.406 24.2
13 21.098 48.2
14 21.399 8.9
15 22.134 14.7
16 22.407 4.2
17 23.302 42.3
18 24.685 54.9
19 26.257 30.2
20 27.552 15.8
21 27.948 17.6
22 28.520 2.3
23 29.742 2.2
24 30.746 5.3
In some embodiments, the X-ray powder diffraction pattern of form a of the compound of formula (I) expressed as 2 Θ angles can also be substantially as shown in figure 3.
In some embodiments, the compound of formula (I) may also have an endothermic peak at 167.7-171.0 ℃ in a Differential Scanning Calorimetry (DSC) profile of form a.
In some embodiments, the Differential Scanning Calorimetry (DSC) of form a of the compound of formula (I) can also be substantially as shown in figure 4.
In some embodiments, the thermogravimetric analysis (TGA) of the crystalline form a of the compound of formula (I) can also be substantially as shown in figure 5. In the TGA diagram, the form a is substantially free of weight loss prior to heating to the decomposition temperature, which shows that the form a is an anhydrate.
In some embodiments, the dynamic water sorption profile (DVS) of form a of the compound of formula (I) may further increase the weight of form a by about 0.30% over the initial weight in the range of 0% to 95% relative humidity. It was shown that said form a is substantially non-hygroscopic.
In some embodiments, the dynamic water sorption profile (DVS) of the crystalline form a of the compound of formula (I) can also be substantially as shown in figure 6.
In some embodiments, the form a of the compound of formula (I) is substantially pure. For example, the form a may be present in an amount of at least 99%, at least 95%, or less to 90% by weight. Alternatively still, the amount by weight of form a is at least 80%, or at least 70%, or even less than 60%. Or further, the content of the crystal form A is at least 50 percent by weight.
In another aspect, the invention provides a preparation method of the crystalline form a of the compound represented by formula (I), which is method 1, method 2 or method 3:
the method comprises the following steps: cooling a heat saturated solution formed by the compound shown as the formula (I) and a solvent to room temperature at 40-80 ℃, and crystallizing; wherein the solvent is selected from one or more of ethanol, ethyl acetate, methyl isobutyl ketone, acetonitrile and methyl tert-butyl ether;
the method 2 comprises the following steps: mixing a solution formed by a compound shown as a formula (I) and a solvent A with a solvent B at 40-80 ℃, and crystallizing; wherein the solvent A is one or more of ethanol, isopropanol, acetone, ethyl acetate, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran and 1,4-dioxane, and the solvent B is one or more selected from methyl tert-butyl ether, n-heptane and water;
the method 3 comprises the following steps: placing excessive compound shown as formula (I) in solvent to form suspension, suspending, separating solid phase from liquid phase, and drying; wherein the solvent is selected from one or more of ethanol, methyl isobutyl ketone, methyl tert-butyl ether, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane.
In some embodiments, the compound of formula (I) in method 1, method 2, or method 3 may be any solid form of the compound of formula (I), for example, an amorphous solid form of the compound of formula (I).
Wherein the content of the first and second substances,
in the method 1, the raw material is subjected to a thermal treatment,
the amount of the solvent may be an amount conventionally used in the art for such procedures, as long as a saturated solution is formed, and in some embodiments, the volume-to-mass ratio of the solvent to the compound represented by formula (I) is 10.0 to 30.0mL/g, for example, 16.7mL/g.
The crystallization can be carried out in a cooling mode at the speed of 5-30 ℃/hour.
In some embodiments, in said method 1, said hot saturated solution is filtered prior to said crystallization. The filtration treatment may be by means of filtration conventional to such operations in the art, preferably by thermal filtration.
The crystallization can also comprise solid-liquid separation and drying operations.
The solid-liquid separation may be filtration.
The drying may be drying under reduced pressure, for example, drying under reduced pressure at 40 ℃.
In the method 2, the raw material is treated,
the volume-mass ratio of the solvent A to the compound shown in the formula (I) is 8.0-11.0 mL/g, for example, 9.5mL/g.
The temperature of 40-80 ℃ can be 70-75 ℃ or 45-50 ℃.
The volume ratio of the solvent A to the solvent B is (0.2-5): 1, e.g., (0.38-1.9): 1.
the mixing timing is not particularly limited, and a solution of the compound represented by the formula (I) and the solvent a may be mixed with the solvent B, or a solution of the compound represented by the formula (I) and the solvent a may be cooled until a solid is precipitated and then mixed with the solvent B.
The mixing manner may be a dropping manner, for example, dropping the solvent B into a solution of the compound represented by the formula (I) and the solvent A.
In some embodiments, in the method 2, preferably, when the solvent B is n-heptane, the solvent a is ethanol and/or ethyl acetate; when the solvent B is methyl tert-butyl ether, the solvent A is one or more selected from ethanol, isopropanol and 2-methyltetrahydrofuran.
In some embodiments, the method 3, the solvent is selected from ethanol and/or methyl isobutyl ketone, e.g., methyl isobutyl ketone.
In the method 3, the amount of the solvent is not limited, and may be conventional in the art as long as it forms a suspension with the compound represented by the formula (I), for example, when the solvent is methyl isobutyl ketone, the volume-to-mass ratio of the solvent to the compound represented by the formula (I) is 0.2g/mL.
The temperature of the suspension equilibrium may be conventional, as long as it is not above the boiling point of the solvent system, and may be from 30 ℃ to 60 ℃, e.g., 50 ℃.
In some embodiments, in the method 3, the time for the suspension equilibration may depend on the reaction scale, generally from 2 hours to 14 days; for example, in laboratory lab scale, the reaction can be completed in about 2-24 hours at room temperature; the reaction time after the amplification is prolonged, for example, from 24 hours to 14 days, preferably from 7 days to 14 days, for example, 7 days, 14 days.
The invention provides a crystal form B of a compound shown as a formula (I), which has an X-ray powder diffraction pattern expressed by a 2 theta angle, and has characteristic peaks at 5.0 +/-0.2 degrees, 9.8 +/-0.2 degrees, 14.7 +/-0.2 degrees, 19.6 +/-0.2 degrees, 24.6 +/-0.2 degrees and 31.0 +/-0.2 degrees, wherein the X-ray powder diffraction pattern is measured by using a Ka spectral line of a Cu target;
Figure BDA0003656248860000051
in some embodiments, the crystalline form B of the compound of formula (I), having an X-ray powder diffraction pattern expressed in terms of 2 Θ angles, may also have the following diffraction peaks and peak area percentages:
numbering 2θ(±0.2°) Percentage of Peak area (%)
1 4.958 100.0
2 9.838 19.8
3 14.718 1.1
4 19.631 4.6
5 24.582 2.9
6 31.047 1.1
In some embodiments, the X-ray powder diffraction pattern of crystalline form B of the compound of formula (I) expressed in terms of 2 Θ angles can also be substantially as shown in fig. 7.
In some embodiments, the compound of formula (I) may also have an endothermic peak at 132.0-142.5 ℃ in a Differential Scanning Calorimetry (DSC) of form B.
In some embodiments, a Differential Scanning Calorimetry (DSC) profile of form B of the compound of formula (I) may also be substantially as shown in figure 8.
In some embodiments, the thermogravimetric analysis (TGA) of the crystalline form B of the compound of formula (I) may also be substantially as shown in figure 9. In the TGA plot, the form B is substantially free of weight loss prior to heating to the decomposition temperature, which shows that the form B is an anhydrate.
In some embodiments, the dynamic water sorption profile (DVS) of form B of the compound of formula (I) may further increase the weight of form B by about 0.95% over the initial weight in the range of 0% to 95% relative humidity. It is shown that said form B is substantially non-hygroscopic.
In some embodiments, the dynamic water sorption profile (DVS) of the crystalline form a of the compound of formula (I) can also be substantially as shown in figure 10.
In some embodiments, the crystalline form B of the compound of formula (I) is substantially pure. For example, the form B may be present in an amount of at least 99%, at least 95%, or less to 90% by weight. Alternatively still, the amount by weight of form B is at least 80%, or at least 70%, or even less than 60%. Or further, the content of the crystal form B by weight is at least 50%.
In another aspect, the invention provides a method for preparing the crystalline form B of the compound of formula (I), which is method a or method B:
the method A comprises the following steps: forming a hot saturated solution of the compound shown as the formula (I) and methanol at 50-70 ℃, and volatilizing the solvent at room temperature for crystallization;
the method B comprises the following steps: forming a suspension of the compound shown as the formula (I) and a solvent, after the suspension is balanced, separating a solid phase from a liquid phase, and drying; the solvent is methanol or a mixed solution of methanol and water.
In some embodiments, the compound of formula (I) in process a or process B may be in any solid form, for example, in an amorphous solid form.
In some embodiments, in the method a, the hot saturated solution is filtered before the volatile solvent is crystallized. The filtration treatment may be by means of filtration conventional to such operations in the art, preferably by thermal filtration.
In some embodiments, the method a may further include a drying operation after the crystallization. The drying is reduced pressure drying.
In some embodiments, in the method B, when the solvent is a mixed solution of methanol and water, the volume ratio of methanol to water may be (5-95): 5, e.g. 5:5 or 95:5.
the temperature of the suspension equilibrium may be conventional, as long as it is not higher than the boiling point of the solvent system, and may be from 10 ℃ to 40 ℃, for example, room temperature.
In some embodiments, the time for said suspension equilibration in said method B may depend on the scale of the reaction, generally between 2 days and 14 days; for example, in a laboratory lab scale, the test can be completed in about 2 days at room temperature; the reaction time after the amplification is prolonged, for example, from 2 days to 14 days, preferably from 7 days to 14 days, for example, 7 days, 14 days.
In some embodiments, in method B, the suspension may be heated while suspending, and the heating temperature should be no higher than the boiling point of the solvent system, e.g., about 40 ℃ to about 50 ℃. The heating may promote the transformation of the solid in the suspension to form B of the compound of formula (I).
The invention also provides a crystal form C of the compound solvate shown as the formula (I), wherein the structure of the crystal form C is shown as follows:
Figure BDA0003656248860000071
it belongs to a triclinic system, the space group is P1, and the unit cell parameters are as follows:
Figure BDA0003656248860000072
α =95.12 (3) °, β =93.82 (3) °, γ =90.43 (3) °; cell volume
Figure BDA0003656248860000073
The number of asymmetric units in the unit cell Z =1.
In one embodiment, the crystal structure data collected by X-ray single crystal diffraction of form C is shown in table 4 below:
TABLE 4
Figure BDA0003656248860000074
Figure BDA0003656248860000081
The invention also provides a pharmaceutical composition, which comprises one or more of the crystal form A of the compound shown in the formula (I), the crystal form B of the compound shown in the formula (I), the crystal form C of the compound solvate shown in the formula (I), and pharmaceutically acceptable auxiliary materials.
In the invention, one or more of the crystal form A of the compound shown in the formula (I), the crystal form B of the compound shown in the formula (I) and the crystal form C of the compound solvate shown in the formula (I) can be combined with one or more other active ingredients for use; when used in combination, the active ingredients may be separate compositions for simultaneous administration by the same or different routes of administration or for separate administration at different times in therapy, or they may be administered together in the same pharmaceutical composition.
In the present invention, the method of administration of the pharmaceutical composition is not particularly limited, and various dosage forms of the preparation may be selected for administration according to the age, sex, and other conditions and symptoms of the patient; for example, oral administration of tablets, pills, solutions, suspensions, emulsions, granules or capsules; the injection can be administered alone or mixed with injectable delivery solution (such as glucose solution and amino acid solution) for intravenous injection; the suppository is administered to the rectum.
In another aspect, the invention provides the use of a compound of formula (I) or a pharmaceutical composition as described above for the preparation of a medicament for the treatment and/or prevention of a Ras-mediated disease.
In some embodiments, the compound of formula (I) is form a of the compound of formula (I) described above, form B of the compound of formula (I) described above, or form C of the compound solvate of formula (I) described above.
In some embodiments, the Ras is, for example, a G12C mutation of one or more of K-Ras, H-Ras and N-Ras, and further for example a G12C mutation of K-Ras.
In some embodiments, the Ras-mediated disease, such as cancer. Such as one or more of colon cancer, appendiceal cancer, pancreatic cancer, MYH-related polyposis, hematological cancer, breast cancer, endometrial cancer, gallbladder cancer, bile duct cancer, prostate cancer, lung cancer, brain cancer, ovarian cancer, cervical cancer, testicular cancer, kidney cancer, head or neck cancer, bone cancer, skin cancer, rectal cancer, liver cancer, esophageal cancer, stomach cancer, thyroid cancer, bladder cancer, lymphoma, leukemia, and melanoma.
In another aspect, the invention provides the use of a compound of formula (I) or a pharmaceutical composition as described above in the preparation of a Ras inhibitor.
In some embodiments, the compound of formula (I) is form a of the compound of formula (I) described above, form B of the compound of formula (I) described above, or form C of the compound solvate of formula (I) described above.
In some embodiments, the Ras is, for example, a G12C mutation of one or more of K-Ras, H-Ras and N-Ras, and further for example a G12C mutation of K-Ras.
In some embodiments, the Ras inhibitors can be used in mammalian organisms; also useful in vitro, primarily for experimental purposes, for example: provide comparison as a standard sample or a control sample, or prepare a kit according to the conventional method in the field, and provide rapid detection for Ras inhibition effect.
The invention also provides the application of the compound shown in the formula (I) or the pharmaceutical composition in preparing a medicament for treating and/or preventing cancer.
In some embodiments, the compound of formula (I) is form a of the compound of formula (I) described above, form B of the compound of formula (I) described above, or form C of the compound solvate of formula (I) described above.
In some embodiments, the cancer is, for example, one or more of colon cancer, appendiceal cancer, pancreatic cancer, MYH-related polyposis, hematological cancer, breast cancer, endometrial cancer, gallbladder cancer, bile duct cancer, prostate cancer, lung cancer, brain cancer, ovarian cancer, cervical cancer, testicular cancer, kidney cancer, head or neck cancer, bone cancer, skin cancer, rectal cancer, liver cancer, esophageal cancer, stomach cancer, thyroid cancer, bladder cancer, lymphoma, leukemia, and melanoma.
Unless otherwise defined, the terms used in the present invention have the following meanings:
the "compound represented by the formula (I)" used in the present invention means a compound having a chemical structure represented by the following formula (I):
Figure BDA0003656248860000091
as used herein, "amorphous" or "amorphous" refers to a solid form of a compound of formula (I) as an amorphous form.
As used herein, "crystal form" and "polymorph" are used interchangeably herein and specifically refer to form a of the compound of formula (I) as described above, form B of the compound of formula (I) as described above, or form C of the solvate of the compound of formula (I) as described above.
The crystalline forms of the invention may be identified by one or more solid state analysis methods. Such as X-ray powder diffraction, single crystal X-ray diffraction, infrared absorption spectrum, differential scanning calorimetry, thermogravimetric curve, etc. Those skilled in the art will appreciate that the peak intensity and/or peak condition of X-ray powder diffraction may vary depending on the experimental conditions. Meanwhile, due to different accuracies of the instruments, the measured 2 theta value has an error of about +/-0.2 degrees. The relative intensity values of the peaks depend more on certain properties of the measured sample, such as the size of the crystals and the purity than the position of the peaks, so that the measured peak intensities may deviate by about + -20%. One skilled in the art can obtain sufficient information to identify each crystal form from the X-ray powder diffraction data provided herein, despite experimental, instrumental, and orientation preferences. In infrared spectrometry, the shape of the spectrum and the position of an absorption peak are affected to a certain extent due to different instrument performances of various models, different grinding degrees or different water absorption degrees during preparation of a test article, and the like. In DSC measurements, however, the initial temperature, maximum temperature and heat of fusion data of the endothermic peak obtained by actual measurement are subject to some degree of variability, depending on the heating rate, crystal shape and purity and other measurement parameters.
As used herein, "anhydrate" means that the sample contains no more than 1.0% (weight percent) or no more than 0.5% (weight percent) water as determined by TGA.
As used herein, "about" for a parameter such as amount, angle, temperature, time, etc., means up to + -10%, preferably within + -5%, and most preferably within + -2% of the specifically given value; however, when measuring the onset temperature and peak temperature of a thermal event in a Differential Scanning Calorimetry (DSC) plot, the term "about" indicates that the onset temperature or peak temperature can generally be within ± 3 ℃ of each other, regardless of the absolute value of the onset temperature or peak temperature. As will be appreciated by those skilled in the art, the use of a number for a non-critical parameter is for illustrative purposes only and is not limiting.
As used herein, "substantially pure" when used to describe a polymorph of a compound of formula (I) means that the solid form of the compound contains this polymorph and does not substantially contain other polymorphs of the compound. Typical substantially pure polymorphs contain other polymorphs at less than 50%, preferably less than 40%, preferably less than 30%, preferably less than 20%, preferably less than 10%, preferably less than 5%, preferably less than 1% by weight.
As used herein, "substantially non-hygroscopic" when used to describe the polymorphic form of a compound of formula (I) means that the polymorphic form of a compound of formula (I) increases in mass by less than 2%, preferably less than 1%, in the range of 0-95% relative humidity compared to the initial mass as measured using dynamic moisture sorption (DVS) techniques.
As used herein, "hot saturated solution" means a solution of a solute in one or more solvents, wherein the solute is in excess, formed by heating the solution to form a supersaturated solution. The heating temperature is typically at or below the boiling point of the solvent by about 5 to 30 deg.C, for example, the heating temperature may be 40 deg.C, 50 deg.C, 60 deg.C, 70 deg.C, 75 deg.C, 80 deg.C.
As used herein, "hot filtration" means filtration of the hot saturated solution through a pinhole filter, wherein the membrane is preferably polyvinylidene fluoride (PVDF) or nylon, and the pore size of the membrane is preferably 0.45 μm.
As used herein, "hot clarified liquid" means a solution of a solute completely dissolved in one or more solvents by heating, wherein the solvent is in excess. The heating temperature is typically about 5 to 30 ℃ below the boiling point of the solvent, for example, the heating temperature may be 40 ℃,50 ℃,60 ℃, 70 ℃, 75 ℃, 80 ℃.
As used herein, a "suspension" refers to a solution of a solute in one or more solvents in which the solute is present in a significant excess, such as a suspension prepared in a suspension equilibrium experiment.
In the present invention, "suspension equilibration" used may be accomplished by methods conventional in the art, for example, by rotating the prepared suspension vial by 360 °, or by stirring the prepared suspension. The temperature of the suspension equilibrium is usually room temperature unless otherwise specified.
In the present invention, the "room temperature" means 10 to 30 ℃.
In the present invention, the operation of the "solid-liquid phase separation" can be performed by a method conventional in the art, such as filtration and centrifugation. When the "solid-liquid separation" is accomplished by the filtration operation, the filtration means, unless otherwise specified, suction filtration under reduced pressure. The specific operation of the centrifugation is as follows: the pre-separated sample is placed in a centrifuge, typically at a centrifugation rate of 3000-15000 rpm, preferably 6000-12000 rpm. The solid obtained by said "isolation" may be further washed, preferably with the same solvent as used in the crystal production process, and the amount of the washing solvent is generally 0.1 to 1 times the volume of the solvent used in the crystal production process.
In the present invention, the "drying" can be carried out by a method conventional in the art, and the drying method is, for example, drying under atmospheric pressure, drying under reduced pressure, preferably drying under reduced pressure. Without limitation, the degree of vacuum of the reduced pressure drying may be-0.09 MPa; the temperature of the reduced pressure drying may be 30 to 80 deg.C, preferably 40 to 70 deg.C, for example 40 deg.C, 50 deg.C. The drying period is typically from 1 hour to overnight, i.e. 1-24 hours, e.g. 2 hours, 16 hours.
In the present invention, the "stirring" can be performed by a method conventional in the art, such as magnetic stirring and mechanical stirring, and the stirring speed is 50 to 1200 rpm, preferably 200 to 500 rpm.
In the present invention, "volatilizing" means that the mouth of a vial containing a solution filtered by a filter membrane is covered with an aluminum foil and punctured with a small hole, and the solvent is slowly volatilized when left standing in a laboratory environment, for example, the mouth of a vial containing a solution filtered by a filter membrane is covered with an aluminum foil and punctured with a small hole, and left standing in a fume hood to slowly volatilize the solvent.
As used herein, "treating" or "treatment" refers to ameliorating a disease or disorder (i.e., arresting the disease or reducing the manifestation, extent or severity of clinical symptoms thereof); alternatively, at least one physical parameter, which may not be perceived by the subject, is improved; or slow disease progression.
In the context of the present invention, "prevention" refers to a reduction in the risk of acquiring or developing a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not occurring in a subject who may be exposed to a disease causing agent or predisposing disease prior to the onset of the disease).
The term "pharmaceutically acceptable excipients" refers to excipients and additives used in the manufacture of pharmaceutical products and in the formulation of pharmaceutical formulations, and is intended to include all substances contained in pharmaceutical formulations, except the active ingredient. See pharmacopoeia of the people's republic of China (2020 Edition), or Handbook of Pharmaceutical Excipients (Raymond C Rowe,2009 Sixth Edition).
The above preferred conditions can be arbitrarily combined to obtain preferred embodiments of the present invention without departing from the common general knowledge in the art.
The reagents and starting materials used in the present invention are commercially available.
The positive progress effects of the invention are as follows:
the crystal form of the compound shown as the formula (I) has one or more improved characteristics, particularly shows that the crystal form has good purity, improves the stability to high temperature and illumination, improves the hygroscopicity under high humidity conditions, is not easy to generate crystal transformation, can better resist the problems of purity reduction, impurity increase and the like of active ingredients caused by factors such as environmental temperature, humidity, illumination and the like in the processes of production, storage, transportation and the like of medicaments, and reduces the risk of curative effect reduction and safety risk brought by the problems; and can better resist the risk of the change of the bioavailability of the medicament caused by the transformation of the solid form of the active ingredient during the long-term storage of the medicament preparation, and is suitable for the development of the preparation of the medicament as the active ingredient. The preparation method provided by the invention is simple and easy to operate, and is suitable for industrial production.
Drawings
FIG. 1 is an amorphous X-ray powder diffraction (XRPD) pattern of a compound of formula (I).
FIG. 2 is an ellipsoid diagram of a unimolecular solid structure of a crystal form C of a compound solvate shown as a formula (I).
Figure 3 is an XRPD pattern of form a of the compound of formula (I).
FIG. 4 is a Differential Scanning Calorimetry (DSC) profile of form A of the compound of formula (I).
Figure 5 is a thermogravimetric analysis (TGA) profile of crystalline form a of the compound shown in formula (I).
FIG. 6 is a dynamic moisture sorption (DVS) profile of form A of the compound of formula (I); wherein 1 is a moisture absorption curve and 2 is a moisture removal curve.
Figure 7 is an XRPD pattern of form B of the compound of formula (I).
FIG. 8 is a DSC of form B of the compound of formula (I).
Figure 9 is a TGA profile of crystalline form B of the compound shown in formula (I).
FIG. 10 is a DVS plot of form B of a compound according to formula (I); wherein 1 is a moisture absorption curve and 2 is a moisture removal curve.
FIG. 11 is a DVS plot of the amorphous state of a compound of formula (I); wherein 1 is a moisture absorption curve and 2 is a moisture removal curve.
FIG. 12 is a graph of the change in body weight of mice in the lung cancer cell NCI-H358 subcutaneous xenograft tumor model.
FIG. 13 is a graph showing the change in tumor volume in mice in the lung cancer cell NCI-H358 subcutaneous xenograft tumor model.
FIG. 14 is a graph of body weight changes of mice in a pancreatic cancer cell MIA PaCa-2 subcutaneous xenograft tumor model.
FIG. 15 is a graph showing the change in tumor volume of mice in the pancreatic cancer cell MIA PaCa-2 subcutaneous xenograft tumor model.
FIG. 16 is a graph of the change in body weight of mice in the model of subcutaneous xenograft tumors of human colon cancer cells SW 837.
FIG. 17 is a graph showing the change in tumor volume of mice in the model of subcutaneous xenograft tumor of human colon cancer cells SW 837.
Detailed Description
The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention. The experimental methods without specifying specific conditions in the following examples were selected according to the conventional methods and conditions, or according to the commercial instructions.
The following examples employ as starting material a compound of formula (I) in amorphous solid form.
The solvents referred to in the following examples are all analytically pure or chromatographically pure, and when the solvents are mixed solvents, they are in volume ratios unless otherwise indicated.
The test instrument and test conditions used in the experiment of the invention are as follows:
(1) X-Ray Powder Diffraction (XRPD)
The detection is carried out by adopting a D8 advanced X-ray powder diffractometer of Bruker company, and the Kalpha spectral line of a Cu target is adopted
Figure BDA0003656248860000121
Figure BDA0003656248860000131
The voltage is 40 kilovolts, the current is 40 milliamps, the divergence slit is 1.0mm, the cable-stayed slit is 0.4 degrees, the scanning mode is continuous scanning, the scanning angle range is 3 degrees to 45 degrees, the step length is 0.02 degrees, the scanning speed is 8 degrees/min, and the detector: lynxEye.
(2) Differential Scanning Calorimetry (DSC)
The temperature was measured by DSC25 type differential scanning calorimeter from TA instruments under nitrogen at a temperature rise rate of 10 deg.C/min and in the temperature range of 25-300 deg.C.
(3) Thermogravimetric Analysis (Thermo Gravimetric Analysis, TGA)
The temperature was measured by a Q500 thermogravimetric analyzer (TA instruments Co.) under nitrogen at a temperature rate of 10 ℃/min to 350 ℃.
(4) Dynamic moisture adsorption (DVS)
The method is characterized in that an Advantage 1.0 type dynamic moisture adsorption instrument of SMS company is adopted for detection, the temperature is 25 ℃, the relative humidity range is 0-95%, the humidity change step length is 5%, when the value of the mass change rate dm/dt is less than 0.002%, balance is regarded as balance, when the mass change rate dm/dt within 5 minutes is less than 0.01%/minute, the balance standard in the detection process is regarded, and the longest balance time is 2 hours.
Preparation example 1 Synthesis of Compound represented by the formula (I)
(1) Synthesis of Compound 1
Synthetic route to compound 1:
Figure BDA0003656248860000132
Figure BDA0003656248860000141
synthesis of Compound 1-j
The compound 1-bromo-8-chloronaphthalene (500mg, 2.07mmol) was dissolved in THF (20 mL), cooled to-78 deg.C, and n-BuLi (2.5M, 1.66mL, 4.14mmol) was added dropwise under nitrogen. After the addition was complete, the mixture was stirred at-78 ℃ for 10 minutes, and then DMF (800. Mu.L, 10.35 mmol) was added dropwise at-78 ℃. After the addition was complete, the reaction mixture was stirred at-78 ℃ for 30 minutes, warmed to room temperature and stirred for 2 hours, quenched with 50mL of saturated ammonium chloride solution, and extracted with ethyl acetate (50ml × 2). The organic phase was washed with saturated brine (50ml × 2), treated with anhydrous sodium sulfate, filtered, and concentrated to give the crude product. The crude product was isolated and purified by flash column separation (EA/PE = 1/10) to give compound 1-j (330mg, 84% yield) as a white solid. LC-MS (ESI) m/z =191.0[ M + H ]] +1 H NMR(400MHz,CDCL 3 ):δ11.31(s,1H),8.03(dd,1H,J 1 =1.2Hz,J 2 =8.4Hz),7.92(dd,1H,J 1 =1.2Hz,J 2 =7.2Hz),7.86(1H,J=8.4Hz),7.70(dd,1H,J 1 =1.2Hz,J 2 =7.6Hz),7.59(t,1H,J=7.6Hz),7.47(t,1H,J=8Hz).
Synthesis of Compound 1-i
NaH (60%, 242mg, 6.05mmol) was added to 6mL of THF at room temperature. Methyl acetoacetate (543. Mu.L, 5.04 mmol) was then added under nitrogen at room temperature, and after the mixture was stirred for 30 minutes under nitrogen at room temperature, n-BuLi (2.5M, 2.4mL, 6.05mmol) was added dropwise at-15 ℃ to-10 ℃. After the addition was complete, the mixture was kept at this temperature for 30 minutes, and then a solution of compound 1-j (320mg, 1.68mmol) in THF (10 mL) was added dropwise. After the addition was completed, the mixture was stirred at low temperature (-10 ℃ C. -0 ℃ C.) for 2 hours, and then quenched with a saturated ammonium chloride solution (50 mL), followed by extraction with ethyl acetate (50mL. Multidot.2). The organic phase was washed with saturated brine (50ml × 2), treated with anhydrous sodium sulfate, filtered, and concentrated to give the crude product, which was purified by flash column separation (EA/DCM = 1/10) to give compound 1-i (510mg, 99% yield) as a white solid. LC-MS (ESI) m/z =329.1[ m + Na ]] +1 H NMR(400MHz,CDCl 3 ):δ8.06(d,1H,J=6.4Hz),7.79(d,2H,J=8Hz),7.58(dd,1H,J 1 =7.6Hz,J 2 =1.6Hz),7.53(t,1H,J=7.6Hz),7.34(t,1H,J=7.6Hz),6.91(dd,1H,J 1 =9.2Hz,J 2 =2.4Hz),3.74(s,3H),3.54(s,2H),3.36(dd,1H,J 1 =18Hz,J 2 =1.6Hz),3.24(d,1H,J=3.6Hz),2.85-2.75(m,1H).
Synthesis of Compounds 1-h
Compound 1-i (510mg, 1.66mmol) was dissolved in DCM (18 mL) at room temperature, and DMF-DMA (245. Mu.L, 1.83 mmol) was added under nitrogen at room temperature. The reaction mixture was stirred at room temperature for 45 minutes, and BF was added thereto 3 ·Et 2 O (232. Mu.L, 1.83 mmol). After the addition was completed, the mixture was stirred at room temperature for 1 hour, and then diluted with 100mL of ethyl acetate. The organic phase was successively saturated with NaHCO 3 The solution (100 mL) was washed with saturated brine (100ml × 2), treated with anhydrous sodium sulfate, filtered, and concentrated to give crude compound 1-h (520 mg). The crude product is used directly without purificationAnd (4) carrying out one-step reaction. LC-MS (ESI) m/z =317.1[ 2[ M ] +1 ]] + .
Synthesis of Compounds 1-g
Compound 1-h (520mg, 1.64mmol) was dissolved in THF (20 mL) at room temperature, and lithium tri-sec-butylborohydride (1M, 1.64mL, 1.64mmol) was added dropwise under nitrogen at-78 ℃. After the addition was complete, the mixture was stirred at-78 ℃ for 1 hour, quenched with saturated ammonium chloride solution (50 mL), extracted with ethyl acetate (50ml × 2), the organic phase was washed with saturated brine (50ml × 2), treated with anhydrous sodium sulfate, filtered, concentrated to give crude product, which was purified by flash column separation (PE/EA = 4/1) to give compound 1-g (338mg, 65% yield) as a yellow oil. LC-MS (ESI) m/z =319.0[ m +1 ]] + .
Synthesis of Compound 1-f
Compound 1-g (338mg, 1.06mmol) was dissolved in methanol (20 mL) at room temperature, and then sodium methoxide (286mg, 5.3mmol) and the compound 2-methyl-2-mercaptourea sulfate (265mg, 0.954mmol) were added in this order under nitrogen at 0 ℃. After the addition was complete, the mixture was warmed to room temperature and stirred for 20 hours. The reaction solution was adjusted to pH 5 with 1N dilute hydrochloric acid, a solid precipitated, filtered, the filter cake washed with water (5 ml × 2), the solid collected and dried under vacuum to give crude 1-f (313 mg) as a white solid. LC-MS (ESI) m/z =359.1[ 2 ] M +1] + .
Synthesis of Compound 1-e
Compound 1-f (313mg, 0.87mmol) was dissolved in DCM (10 mL) at room temperature, and DIPEA (431. Mu.L, 2.61 mmol), triflic anhydride (219. Mu.L, 1.31 mmol) were added sequentially under nitrogen in an ice-water bath. After the addition was complete, the reaction mixture was stirred for 2 hours in an ice-water bath, quenched with saturated sodium bicarbonate solution (50 mL), extracted with DCM (50ml × 2), and the organic phase was treated with anhydrous sodium sulfate, filtered, concentrated to give crude product which was purified by flash column separation (EA/PE = 1/10) to give compound 1-e (83mg, 16% from 2 steps) as a white solid. LC-MS (ESI) m/z =491.0[ M +1 ]] + .
Synthesis of Compound 1-d
Compound 1-e (83mg, 0.1699 mmol) was dissolved in DMF (10 mL) at room temperatureDIPEA (84. Mu.L, 0.507 mmol), (S) -2-cyanomethylpiperazine-1-carboxylic acid benzyl ester hydrochloride (59.9mg, 0.203mmol) was then added in this order. After the addition was completed, the mixture was stirred at 100 ℃ for 1 hour under nitrogen, cooled to room temperature, quenched with saturated brine (50 mL), and extracted with ethyl acetate (50ml × 2). The organic phase was washed with saturated brine (50ml × 3), then treated with anhydrous sodium sulfate, filtered, and concentrated to give crude product, which was purified by flash column separation (EA/PE = 1/1) to give compound 1-d (101mg, 99% yield) as a white solid. LC-MS (ESI) m/z =600.2[ 2 ], [ M +1 ]] + .
Synthesis of Compound 1-c
Compounds 1-d (101mg, 0.168mmol) were dissolved in ethyl acetate (10 mL) at room temperature, and MCPBA (85%, 88.4mg,0.437 mmol) was added at room temperature. After the addition was complete, the mixture was stirred at room temperature for 2 hours, quenched with saturated sodium bicarbonate solution (20 mL), extracted with ethyl acetate (25ml × 2), the organic phase was treated with anhydrous sodium sulfate, filtered, and concentrated to give a crude product which was purified by flash column separation (EA/PE = 1/4) to give compound 1-c (88mg, 82% yield) as a white solid. LC-MS (ESI) m/z =632.1[ 2 ] M +1] + .
Synthesis of Compound 1-b
Compound 1-c (88mg, 0.139mmol) was dissolved in toluene (10 mL) at room temperature, and the reaction was cooled to 0 ℃ and N-methylprolinol (29. Mu.L, 0.243 mmol), t-BuONa (27mg, 0.278mmol) were added in that order. After addition, the reaction mixture was stirred under nitrogen in an ice-water bath for 0.5 h, quenched with water (20 mL) and extracted with ethyl acetate (30ml × 2). The organic phase was treated with anhydrous sodium sulfate, filtered, and concentrated to give the crude product, which was purified by flash column separation (MeOH/DCM = 1/10) to give compound 1-b (78mg, 84% yield) as a white solid. LC-MS (ESI) m/z =667.3[ M +1 ]] + .
Synthesis of Compound 1-a
Compound 1-b (72mg, 0.108mmol) was dissolved in methanol (50 mL) at room temperature, the reaction solution was cooled to-78 deg.C and purged with nitrogen 2 times, and then Pd/C (150 mg), znBr was added 2 (24.3mg, 0.108mmol), hydrogen substitution 3 times, reactionThe reaction solution was warmed to room temperature and stirred under hydrogen for 5 hours. The reaction was filtered and concentrated to give crude which was isolated and purified by flash column separation (MeOH/DCM = 1:4) to give compound 1-a (20mg, 35% yield) as a white solid. LC-MS (ESI) m/z =533.0[ m +1 ]] + .
Synthesis of Compound 1
Compound 2-fluoroacrylic acid (5.1mg, 0.0563mmol) was dissolved in DMF (2 mL) at room temperature, then HATU (25.6 mg, 0.0675mmol), DIPEA (18.6. Mu.L, 0.113 mmol) were added sequentially at 0 ℃ and after the addition, the reaction mixture was stirred at 0 ℃ under nitrogen for 20 minutes, then compound 1-a (20mg, 0.0375mmol) in DMF (3 mL) was added to the above reaction mixture, warmed to room temperature and stirred for 5 hours. Quenched with saturated brine (20 mL), extracted with ethyl acetate (25ml × 2), and the organic phase washed with saturated brine (50ml × 3), treated with anhydrous sodium sulfate, filtered, and concentrated to give the crude product which was purified by PREP-TLC separation (MeOH/DCM = 1/10) to give compound 1 (6 mg,26% yield) as a white solid. LC-MS (ESI) m/z =605.2[ 2 ], [ M +1 ]] +1 H NMR(400MHz,CDCl 3 ):δ7.99-7.93(m,1H),7.83(t,2H,J=8.8Hz),7.62-7.49(m,2H),7.36(t,1H,J=7.6Hz),6.55-6.44(m,1H),5.51-5.31(m,1H),5.25(d,1H,J=16.8Hz),5.02-4.93(m,1H),4.82(dd,1H,J 1 =2.4Hz,J 2 =13.6Hz),4.48-4.38(m,1H),4.32-4.19(m,1H),4.17-4.04(m,1H),4.00(d,1H,J=14Hz),3.87-3.70(m,1H),3.66-3.36(m,2H),3.31-3.16(m,2H),3.14-2.98(m,1H),2.96-2.69(m,4H),2.59(d,3H,J=18Hz),2.52-2.34(m,1H),2.15-2.06(m,1H),1.87-1.74(m,2H),0.93-0.76(m,2H).
(2) Resolution of Compound 1
Synthesis of Compounds 1-1 and 1-2
Figure BDA0003656248860000171
The chiral resolution of the compound 1 to obtain the compound shown as the formula (I) is difficult. Despite the various conditions tried, the two isomers of compound 1 were not separated on thin layer chromatography plates and could not be separated by thin layer chromatography means; even in HPLC, the two isomers of compound 1 are poorly separated and cannot be separated by preparative HPLC; finally having to resort to chiral resolution, a number of conditions were tried (table 1 below), and finally chiral resolution conditions 9 were found, enabling the separation of the compound of formula (I) from its diastereoisomers.
TABLE 1
Figure BDA0003656248860000172
Figure BDA0003656248860000181
Figure BDA0003656248860000191
The newly prepared compound 1 (260mg, 0.43mmol) was subjected to chiral resolution under the conditions shown in Table 2 below. Compound 1-1 (76mg, 29% yield) was obtained as a white solid; compound 1-2 (67mg, 26% yield) was obtained as a white solid. Wherein the compound 1-2, i.e., the compound represented by the formula (I), can be determined by analyzing a single crystal structure of the compound represented by the formula (I) shown below.
TABLE 2
Figure BDA0003656248860000192
1-1:LC-MS(ESI):m/z=605.3[M+1] +1 H NMR(400MHz,CDCl 3 )δ7.96(d,1H,J=7.2Hz),7.83(t,2H,J=8.4Hz),7.65-7.50(m,2H),7.36(t,1H,J=8.0Hz),6.47(dd,1H,J 1 =10.8Hz,J 2 =3.2Hz),5.42(d,1H,J=49.2Hz),5.26(dd,1H,J 1 =3.6Hz,J 2 =16.8Hz),5.05-4.76(m,1H),4.97(d,1H,J=13.6Hz),4.84(d,1H,J=13.6Hz),4.36(dd,1H,J 1 =4.8Hz,J 2 =10.4Hz),4.17(dd,1H,J 1 =6.8Hz,J 2 =10.8Hz),4.06-3.87(m,1H),3.77(d,1H,J=10Hz),3.59(dd,1H,J 1 =2.4Hz,J 2 =17.6Hz),3.50-3.15(m,3H),3.14-2.99(m,2H),2.96-2.82(m,2H),2.72-2.59(m,1H),2.47(s,3H),2.32-2.21(m,1H),2.10-1.98(m,1H),1.89-1.67(m,4H).
1-2:LC-MS(ESI):m/z=605.2[M+1] +1 H NMR(400MHz,CDCl 3 )δ7.97(d,1H,J=7.2Hz),7.83(t,2H,J=9.2Hz),7.63-7.51(m,2H),7.36(t,1H,J=7.6Hz),6.52(dd,1H,J 1 =3.2Hz,J 2 =10.8Hz),5.42(d,1H,J=47.2Hz),5.25(dd,1H,J 1 =3.6Hz,J 2 =16.4Hz),4.99(d,1H,J=14.0Hz),4.82(d,1H,J=13.6Hz),5.05-4.72(m,1H),4.38(dd,1H,J 1 =4.8Hz,J 2 =10.4Hz),4.15(dd,1H,J 1 =6.8Hz,J 2 =10.8Hz),3.98(d,1H,J=14Hz),3.87-3.73(m,1H),3.60(dd,1H,J 1 =2.4Hz,J 2 =18.4Hz),3.66-3.54(m,1H),3.54-3.41(m,1H),3.16-2.98(m,2H),2.95-2.71(m,3H),2.71-2.61(m,1H),2.46(s,3H),2.33-2.19(m,1H),2.10-1.98(m,1H),1.90-1.66(m,4H).
The solid form of compound 1-2 (compound represented by formula (I)) prepared by the above method was determined to be amorphous by XRPD, and its XRPD pattern is shown in fig. 1.
Example 1 preparation of crystalline form C of a solvate of a compound of formula (I)
Yet another difficulty with this work is the structural identification of the compounds of formula (I). The compound shown as the formula (I) and the diastereoisomer thereof have obvious difference in vitro cell activity, and the chiral structure is difficult to confirm by methods such as chemistry, general spectrum and the like. Finally, the technique of X-ray diffraction was used, and for this reason, the single crystal was cultured under various conditions without success. Finally, fortunately, colorless transparent plate-like crystals satisfying the requirements of the single crystal X-ray diffraction experiments were obtained by the solvent evaporation method under the condition of the solvent methanol-water (20) (condition corresponding to number 7 in table 3 below). By analysis, the compound 1-2 is determined to be the compound shown in the formula (I), and the compound 1-1 is a diastereoisomer thereof.
The single crystal culture process comprises the following steps: single crystal growth experiments were performed at room temperature using the methods and solvent conditions shown in table 3 below.
TABLE 3
Figure BDA0003656248860000201
Single crystal test conditions: the measurement is carried out according to the first method of 0451 in the four-part general rules of the 2020 edition of Chinese pharmacopoeia, and a D8 Venture type single crystal X-ray diffractometer of Bruke company, moK alpha radiation and omega scanning are adopted. The number of the lumped diffraction points is 18322 in the experimental acceptance of single crystal diffraction, the number of the independent diffraction points is 10763, and the number of the observable points (| F | Y 2 ≥2σ|F| 2 ) 7231.
Analyzing the single crystal structure: the crystal structure is analyzed by adopting a direct method (Shelxs 97), the crystal structure data of the crystal form C of the compound solvate shown in the formula (I) is shown in a table 4, the crystal structure data prove that the crystal form C is a solvate solid form of the compound shown in the formula (I), and the compound shown in the formula (I) in the solvate combines 1 molecule of methanol and 1 molecule of water, and the specific structure is shown as follows:
Figure BDA0003656248860000211
the ellipsoid of the monomolecular stereo structure of the crystal form C is shown in figure 2.
TABLE 4
Figure BDA0003656248860000212
Figure BDA0003656248860000221
Example 2 preparation of form A of the Compound of formula (I)
Dissolving 1.2g of the compound shown as the formula (I) in 20mL of ethanol at about 60 ℃ to obtain a hot saturated solution, carrying out hot filtration, cooling the filtrate to room temperature at the speed of 5 ℃/h, and stirring overnight; the precipitated solid was filtered off, dried at 40 ℃ under reduced pressure for 16 hours to obtain 0.89g of white powder, HPLC purity 99.6%, yield: 74.2 percent.
The obtained powder sample is the crystal form A of the compound shown as the formula (I), the X-ray powder diffraction pattern of the crystal form A is shown as figure 3, and the 2 theta angle, the d spacing, the peak height percentage and the peak area percentage of the main diffraction peak in the X-ray powder diffraction pattern expressed by the 2 theta angle, the d spacing, the peak height percentage and the peak area percentage are shown as the following table 5, wherein the characteristic peak (2 theta +/-0.2 DEG) is 4.839 °, 9.141 °, 9.694 °, 12.433 °, 14.451 °, 15.644 °, 16.545 °, 17.249 °, 17.456 °, 19.801 °, 20.406 °, 21.098 °, 3256 °, 3456 zxft 3238 zxft 5749 °, 3438 zxft 6598 ° 3495 °, 3495 ° and 34zxft 3498 °.
TABLE 5
Figure BDA0003656248860000222
Figure BDA0003656248860000231
The differential scanning calorimetry pattern is shown in FIG. 4. DSC test shows that the crystal form A has an endothermic peak at 167.73-170.96 ℃.
The thermogravimetric analysis thereof is shown in FIG. 5. As can be seen from fig. 5, the weight loss of form a is 0.122% when heated to 220 ℃.
The dynamic water absorption diagram is shown in figure 6. DVS testing showed that the moisture absorption weight gain mass percentage of form a was 0.3048% when varied from 0% to 95% relative humidity.
Example 3 preparation of form A of a Compound of formula (I)
Dissolving 2.0g of a compound shown as a formula (I) in 19mL of ethanol at the temperature of 70-75 ℃ to obtain a thermal clear solution, stirring for 10 minutes, slowly cooling to 45-55 ℃, separating out solids, stirring for 1 hour at the temperature, dropwise adding 10mL of n-heptane, stirring for 1 hour after dropwise adding, slowly cooling to room temperature, stirring for 2 hours, and filtering out the separated solids; drying at 50 deg.C under reduced pressure for 16 hr to obtain white powder 1.80g with yield of 90.0%. The sample obtained by the method is determined to be the crystal form A of the compound shown as the formula (I) by comparing X-ray powder diffraction patterns.
Example 4 preparation of form A of a Compound of formula (I)
Dissolving 2.0g of a compound shown as a formula (I) in 19mL of ethanol at the temperature of 70-75 ℃ to obtain a thermal clear solution, stirring for 10 minutes, slowly cooling to 45-50 ℃, separating out solids, keeping the temperature, stirring for 1 hour, dropwise adding 50mL of methyl tert-butyl ether, stirring for 1 hour after dropwise adding, slowly cooling to room temperature, stirring for 2 hours, and filtering out the separated solids; drying at 50 deg.C under reduced pressure for 16 hr to obtain white powder 1.54g, yield 77.0%. The sample obtained by the method is determined to be the crystal form A of the compound shown as the formula (I) by comparing X-ray powder diffraction patterns.
Example 5 preparation of form A of the Compound of formula (I)
2.0g of the compound represented by the formula (I) was dispersed in 10mL of methyl isobutyl ketone, capped and sealed, and then suspended and equilibrated at about 50 ℃ for 2 days. The solid was filtered off and dried under reduced pressure at 40 ℃ for 16 hours to give 1.87g of a white powder, yield: 93.5 percent. The sample obtained by the method is determined to be the crystal form A of the compound shown as the formula (I) by comparing X-ray powder diffraction patterns.
Example 6 preparation of form B of the Compound of formula (I)
0.93g of the compound represented by the formula (I) was dissolved in 80mL of methanol at about 60 ℃ to obtain a hot saturated solution, which was filtered hot, the filtrate was transferred to a 250mL round-bottomed flask, the mouth of which was covered with aluminum foil and perforated with a small hole, and the solvent was slowly evaporated at room temperature. The solid obtained after complete evaporation of the solvent was dried at 40 ℃ under reduced pressure for 16 hours to obtain 0.82g of white powder with a yield of 88.2%.
The obtained powder sample is the crystal form B of the compound shown as the formula (I), the X-ray powder diffraction pattern of the crystal form B is shown as a figure 7, and the 2 theta angle, the d spacing, the peak height percentage and the peak area percentage of main diffraction peaks in the X-ray powder diffraction pattern expressed by the 2 theta angle, the d spacing, the peak height percentage and the peak area percentage are shown as the following table 6, wherein the characteristic peaks (2 theta +/-0.2 DEG) are 4.958 degrees, 9.838 degrees, 14.718 degrees, 19.631 degrees, 24.582 degrees and 31.047 degrees.
TABLE 6
Figure BDA0003656248860000241
The differential scanning calorimetry thereof is shown in FIG. 8. DSC tests show that the crystal form B has an endothermic peak at 131.98-142.51 ℃.
The thermogravimetric analysis thereof is shown in FIG. 9. From fig. 9, it can be seen that the weight loss of form B is 0.207% when heated to 220 ℃.
The dynamic water absorption diagram is shown in figure 10. DVS testing showed that the moisture absorption weight gain mass percentage of form B was 0.9504% when varied from 0% to 95% relative humidity.
Example 7 preparation of form B of a Compound of formula (I)
0.5g of the compound represented by the formula (I) was suspended in 2mL of methanol, capped and sealed, and then suspended and equilibrated at room temperature for 2 days. The solid was filtered off and dried under vacuum at 40 ℃ for 16 hours to give 0.37g of a white powder with a yield of 74.0%. The sample obtained by the method is determined to be the crystal form B of the compound shown as the formula (I) by comparing X-ray powder diffraction patterns.
Effect example 1 stability
1.1 stability of form A of the compound of formula (I) in solution
Respectively taking a proper amount of crystal form A samples of the compound shown as the formula (I) into small bottles, respectively adding 1mL of single solvent or mixed solvent into the small bottles, and performing ultrasonic dispersion to prepare suspension. The suspension was covered with a lid and closed, and suspended at room temperature for equilibrium for 14 days. Centrifugation, supernatant discarded and the solid dried at 40 ℃ under reduced pressure for XRPD determination.
The results show that the crystal form is not changed in various solvents and still is the crystal form A. The single solvent comprises ethanol, n-propanol, isopropanol, n-butanol, acetone, 2-butanone, methyl isobutyl ketone, ethyl acetate, isopropyl ether, methyl tert-butyl ether, acetonitrile, 2-methyltetrahydrofuran, tetrahydrofuran, dichloromethane, toluene, 1,4-dioxane, water, n-heptane, n-hexane, cyclohexane and cyclopentane; the mixed solvent comprises a solvent mixture and a solvent mixture, wherein the volume ratio of the solvent mixture to the solvent mixture is respectively 15: 85. 65:35 and 95:5, ethanol and water in a volume ratio of 35: 65. 80:20 and 95:5 isopropyl alcohol and water.
1.2 stability of form B of the compound of formula (I) in solution
Respectively taking a proper amount of crystal form B samples of the compound shown as the formula (I) into a small bottle, respectively adding 1mL of single solvent or mixed solvent into the small bottle, and ultrasonically dispersing to prepare suspension. The suspension was covered with a lid and closed, and suspended at room temperature for equilibrium for 14 days. Centrifugation was carried out, the supernatant was discarded, and the solid was dried at 40 ℃ under reduced pressure to determine XRPD.
The results show that the crystal form is not changed in various solvents and is still the crystal form B, and the single solvent or the mixed solvent comprises methanol and the volume ratio of the methanol to the mixed solvent is respectively 50: 50. 75:25 or 95:5, the volume ratio of methanol to water is 50:50 of methanol and acetonitrile, methanol and acetone, methanol and ethyl acetate, methanol and isopropyl ether, methanol and methyl tert-butyl ether and methanol and dichloromethane.
Effect example 2 Competition experiment
Respectively taking equal weight of crystal form A and crystal form B samples of the compound shown as the formula (I) into a same vial, adding 1mL of solvent into the vial, and ultrasonically dispersing to prepare suspension, wherein the solvent comprises ethanol, acetone, ethyl acetate and acetonitrile. The suspension was suspended and equilibrated at room temperature for 7 days after being capped and sealed. Centrifugation, supernatant discarded and the solid dried at 40 ℃ under reduced pressure for XRPD determination.
The results show that the final solid forms are all form a of the compound of formula (I), which is seen to be a thermodynamically more stable polymorph of the compound of formula (I).
Effect example 3 stability of Crystal form A, crystal form B and amorphous form of the Compound represented by the formula (I) under high temperature, high humidity, light conditions
A proper amount of a sample of the crystal form A or the crystal form B or an amorphous state of the compound shown as the formula (I) is placed in a glass vial and is placed in an open way under the conditions of high temperature (60 ℃), high humidity (RH 92.5 percent, 25 ℃) and illumination (4500 +/-500Lux, 25 ℃). Samples were taken after 2 weeks and the samples were analyzed by X-ray powder diffraction and High Performance Liquid Chromatography (HPLC).
The method of X-ray powder diffraction analysis is as described above.
Reverse phase HPLC analysis was carried out using an Agilent ZORBAX SB-Phenyl column (3.5 μm, 4.6X 150 mm) with mobile phase components of (A) water containing 0.1% perchloric acid and (B) acetonitrile. The elution gradient was that mobile phase B increased from 25% to 95% in 12 minutes and held at 95% for 3 minutes, then the system was equilibrated at 25% for 5 minutes. The flow rate was 1.0 mL/min, the injection volume was 2. Mu.L, the column temperature was 25 ℃ and the detection wavelength was 228nm. The test article was prepared with methanol to a final concentration of 0.5mg/mL. And (3) calculating the total impurity content in the sample and the purity of the compound shown as the formula (I) by adopting an area normalization method, and neglecting the chromatographic peak of which the peak area is less than 0.05% in the chromatogram of the sample. The results of the examination are shown in tables 7 to 9 below.
TABLE 7 influence factor test results of form A
Figure BDA0003656248860000251
TABLE 8 influence factor test results for form B
Figure BDA0003656248860000252
TABLE 9 influence factor test results for amorphous form
Figure BDA0003656248860000253
Figure BDA0003656248860000261
The data in tables 7-9 show that the purity of the compound shown in the formula (I) can be improved to about 99.6% by the crystal form A, and the content of active ingredients of the crystal form A is higher than that of the amorphous and crystal form B.
Compared with the crystal form before the influence factor test treatment, the chemical purity and the crystal form of the crystal form A and the crystal form B are not changed and the total impurities are not increased after being respectively placed for 2 weeks under the high-temperature condition, and the XRPD measurement shows that the amorphous state is transformed, and the chemical purity is reduced by 2.7 percent.
Respectively standing for 2 weeks under high humidity condition, wherein the crystal forms of the crystal form A, the crystal form B and the amorphous crystal form are not changed; the chemical purity of the crystal form A and the crystal form B is not changed, and the chemical purity of the amorphous state is slightly reduced.
The crystal forms A, B and amorphous crystal forms are not changed after being respectively placed for 2 weeks under the illumination condition, and the chemical purity of the crystal forms A and B is only reduced by 0.4-0.5%; the chemical purity of the amorphous state decreases by 3.4% and the total impurity growth increases by 3.34%.
Therefore, compared with an amorphous solid form, the crystal form A and the crystal form B of the invention obviously improve the stability of the compound shown as the formula (I) to high temperature and illumination.
Effect example 4 moisture-wicking Properties
DVS assay was performed on amorphous samples of the compound of formula (I) using the same DVS assay method as for form a of the compound of formula (I) in example 2 and form B of the compound of formula (I) in example 6, and the dynamic water uptake figures are shown in fig. 11. The DVS hygroscopicity results for form a, form B and the amorphous form are summarized in table 10.
TABLE 10 DVS hygroscopicity results for form A, form B and amorphous form
Figure BDA0003656248860000262
As can be seen from table 10, when the relative humidity is changed from 0% to 95%, the moisture absorption weight percentages of the crystal form a, the crystal form B and the amorphous form are 0.3048%, 0.9504% and 4.376, respectively, and the moisture absorption weight percentages of the amorphous form are 14.4 times and 4.6 times of those of the crystal form a and the crystal form B, respectively, which shows that the crystal form a and the crystal form B of the present invention have lower hygroscopicity under high humidity conditions.
Therefore, compared with an amorphous solid form, the crystal form A and the crystal form B of the invention obviously improve the moisture-inducing performance of the compound shown as the formula (I), and are more favorable for storing medicaments.
Effect example 5 in vitro cell Activity of Compounds represented by formula (I) and diastereomers thereof
CTG method detection compound on NCI-H358, MIA PaCa-2, A549 and A375 cell line proliferation inhibition experiment.
NCI-H358 was a human non-small cell lung cancer cell with KRas G12C mutation, MIA PaCa-2 was a human pancreatic cancer cell with KRas G12C mutation, a549 was a human non-small cell lung cancer cell with KRas G12V mutation, a375 was a wild-type malignant melanoma cell. The inhibition of different mutations by the compounds was evaluated by examining their proliferation inhibitory activity on four cell lines.
To 384-well plates (except for peripheral wells) were added 40 μ L of test cell suspension (plate 1. The plates were placed in a carbon dioxide incubator overnight. The prepared compound (10 concentration gradients of compound obtained by 3-fold dilution) was added to each well. The cell plates were incubated in a carbon dioxide incubator for 120 hours. To a 384 well plate 25. Mu.L of CellTiter Glo reagent was added, shaken for 10 minutes in the dark, and incubated for 10 minutes. Place the plate in an EnVision read plate, draw a pharmacodynamic inhibition curve using XLFit and calculate IC 50 The value is obtained.
In vitro cell activity data for the compounds of formula (I) and their diastereomers (compounds 1-1) are shown in table 11 below.
TABLE 11 proliferation inhibitory Activity of Compounds of formula (I) and diastereomers on H358 cells, MIA PaCa-2 cells, A549 cells and A375 cells
Figure BDA0003656248860000271
The results showed that the proliferation inhibitory activity of the compound represented by the formula (I) was 614 times that of its diastereomer (compound 1-1) on H358 cells, and 326 times that of its diastereomer (compound 1-1) on MIA PaCa-2 cells.
The result shows that the inhibitory activity of the compound shown as the formula (I) on KRas G12C mutant cells is far better than that of the diastereoisomer (compound 1-1).
Effect example 6 pharmacodynamic study of Compound represented by formula (I) in Lung cancer cell NCI-H358 subcutaneous xenograft tumor model
The experimental criteria were to investigate whether tumor growth could be inhibited, delayed or cured. Tumor diameters were measured twice weekly using a vernier caliper.
The tumor volume was calculated as: v =0.5 × a × b 2 And a and b represent the major and minor diameters of the tumor, respectively.
Evaluation of tumor suppression therapeutic efficacy T/C (%) of the test article. T/C% = T RTV /C RTV ×100%(T RTV : treatment group RTV; c RTV : solvent control RTV).
The calculation formula of the relative tumor volume RTV is RTV = V T /V 1 . Wherein V 1 The resulting tumor volume, V, was measured for cage administration (i.e., day 1) T Tumor volume measured on day T.
The percentage value of T/C (%) reflects the curative effect, and the tested substance is considered to be effective according to the guiding principle of the central anti-tumor drug of the drug trial that T/C% is less than or equal to 40%. Tumor volume therapeutic effect TGI V (%) evaluation, TGI V (%)=(1-(TV T-Dn -TV T-D1 )/(TV C-Dn -TV C-D1 ))×100%,TV C : tumor volume of solvent control group, TV T : tumor volume of treatment group.
BALB/c nude mice were used for the experiments. In the experiment, 0.5% of MC aqueous solution was added to a sample of the form A of the compound represented by the formula (I), and vortexed to mix well, thereby obtaining suspensions having concentrations of 2.5, 5.0, and 10.0mg/mL, respectively, for administration in 25mg/kg, 50mg/kg, and 100mg/kg groups, respectively. The administration is carried out by intragastric administration.
The experiment was divided into solvent control groups, such as 25mg/kg, 50mg/kg and 100mg/kg groups of the compound of formula (I), and administered orally by gavage for 21 days, once a day. During the administration period, the mice in each group showed good tolerance and no obvious abnormality. The graph of the body weight change of the mice is shown in FIG. 12.
The graph of the change in tumor volume in mice is shown in FIG. 13. When the tumor volumes of the treatment groups and the solvent control group are analyzed statistically, the compound shown as the formula (I) shows a very significant difference between the 50mg/kg group and the 100mg/kg group from the 5 th day of the administration to the end of the administration.
On day 22 of the initial administration of the test substance, the T/C values of the compound represented by the formula (I) in the 25mg/kg, 50mg/kg and 100mg/kg groups were 66.58% (TGI%: 42.39%), 36.10% (TGI%: 81.00%) and 13.36% (TGI%: 109.91%), respectively, as compared with the solvent control group.
Experiments show that the compounds shown in the formula (I) in 50mg/kg group and 100mg/kg group have obvious tumor inhibition effect in a lung cancer cell NCI-H358 subcutaneous xenograft tumor model.
Effect example 7 pharmacodynamic study of Compound represented by the formula (I) in pancreatic cancer cell MIA PaCa-2 subcutaneous xenograft tumor model
BALB/c nude mice were used for the experiments. In the experiment, 0.5% of MC aqueous solution was added to a sample of the form A of the compound represented by the formula (I), and vortexed to mix well, thereby obtaining suspensions having concentrations of 1.5, 3.0, and 10.0mg/mL, respectively, for administration in 15mg/kg, 30mg/kg, and 100mg/kg groups, respectively. Administration is by intragastric administration.
The experiment was divided into solvent control group, group 15mg/kg, 30mg/kg and 100mg/kg of the compound of formula (I), and oral gavage was administered once a day for 21 days. During the administration period, the mice in each group showed good tolerance and no obvious abnormality. The graph of the body weight change of the mice is shown in FIG. 14.
The change in tumor volume in mice is shown in FIG. 15. The statistical analysis of the tumor volumes of the treatment groups and the solvent control group shows that the compounds shown in the formula (I) in the 15mg/kg group, the 30mg/kg group and the 100mg/kg group have very significant differences from the 5 th day of the beginning of the administration to the end of the administration.
On day 22 of the initial administration of the compound, the T/C values of the compound represented by the formula (I) in the 15mg/kg, 30mg/kg and 100mg/kg groups were 36.91% (TGI%: 75.39%), 13.55% (TGI%: 103.21%) and 0.56% (TGI%: 118.78%), respectively, as compared with the solvent control group. The compounds of formula (I) showed very good anticancer activity in 15mg/kg, 30mg/kg and 100mg/kg groups.
Experiments show that the compounds shown as the formula (I) in 15mg/kg, 30mg/kg and 100mg/kg groups all show obvious tumor inhibition effect in a pancreatic cancer cell MIA PaCa-2 subcutaneous xenograft tumor model.
Effect example 8 pharmacodynamic study of Compound represented by formula (I) in human Colon cancer SW837 subcutaneous xenograft tumor model
NOD SCID mice were used for the experiments. In the experiment, 0.5 percent aqueous solution of MC is added into a sample of the crystal form A of the compound shown as the formula (I), and the mixture is fully mixed by vortex to obtain suspensions with the concentration of 10.0mg/mL respectively. Administration is by intragastric administration.
The experiment was divided into a solvent control group, a 100mg/kg group of the compound of formula (I), orally administered by gavage for 28 days, once a day. During the administration period, the mice in each group showed good tolerance and no obvious abnormality. The graph of the body weight change of the mice is shown in FIG. 16.
The graph of the change in tumor volume in mice is shown in FIG. 17. Statistical analysis is carried out on the tumor volumes of the treatment group and the solvent control group, and the compound shown as the formula (I) in the 100mg/kg group shows very significant difference from the 4 th day after the beginning of the administration to the end of the administration.
On the 28 th day after the start of administration of the compound, the T/C value and the TGI% value of the compound represented by the formula (I) in the 100mg/kg group were 6.48% and 106.4%, respectively, as compared with those of the solvent control group. The compound of formula (I) showed good anticancer activity in the 100mg/kg group.
Experiments show that the compound shown as the formula (I) in a group of 100mg/kg shows a remarkable tumor inhibition effect in a human colon cancer SW837 subcutaneous xenograft tumor model.
It is to be understood that the examples described herein are for illustrative purposes only and are intended to provide further understanding of the present invention, but are not intended to limit the scope of the present invention. Many modifications, both to materials and methods, may be made by one skilled in the art without departing from the scope of the invention, and such changes or modifications are intended to be included within the spirit and scope of this application and the scope of the appended claims.

Claims (17)

1. A crystalline form a of a compound of formula (I) characterized by an X-ray powder diffraction pattern expressed in terms of 2 Θ angles having characteristic peaks at 4.8 ± 0.2 °, 12.4 ± 0.2 °, 15.6 ± 0.2 ° and 23.3 ± 0.2 °, said X-ray powder diffraction pattern being measured using the ka spectral line of a Cu target;
Figure FDA0003656248850000011
2. form A of the compound of formula (I) according to claim 1, having characteristic peaks at 4.8 ± 0.2 °, 9.1 ± 0.2 °, 9.7 ± 0.2 °, 12.4 ± 0.2 °, 15.6 ± 0.2 °, 19.8 ± 0.2 °, 20.4 ± 0.2 °, 21.1 ± 0.2 °, 23.3 ± 0.2 °, 24.7 ± 0.2 ° and 26.3 ± 0.2 ° in an X-ray powder diffraction pattern, expressed in terms of 2 θ angle.
3. The crystalline form a of the compound of formula (I) according to claim 1, wherein the crystalline form a of the compound of formula (I) satisfies one or more of the following conditions:
(1) An X-ray powder diffraction pattern expressed by a 2 theta angle has characteristic peaks at 4.8 +/-0.2 degrees, 9.1 +/-0.2 degrees, 9.7 +/-0.2 degrees, 12.4 +/-0.2 degrees, 14.5 +/-0.2 degrees, 15.6 +/-0.2 degrees, 16.5 +/-0.2 degrees, 17.2 +/-0.2 degrees, 17.5 +/-0.2 degrees, 19.8 +/-0.2 degrees, 20.4 +/-0.2 degrees, 21.1 +/-0.2 degrees, 21.4 +/-0.2 degrees, 22.1 +/-0.2 degrees, 22.4 +/-0.2 degrees, 23.3 +/-0.2 degrees, 24.7 +/-0.2 degrees, 26.3 +/-0.2 degrees, 27.6 +/-0.2 degrees and 27.9 +/-0.2 degrees;
(2) The Differential Scanning Calorimetry (DSC) of the crystal form A of the compound shown as the formula (I) also has an endothermic peak at 167.7-171.0 ℃;
(3) In the dynamic water sorption Drawing (DVS) of the crystalline form a of the compound of formula (I), the weight gain of the crystalline form a is 0.30% in the range of 0% to 95% relative humidity compared to the initial weight.
4. Form A of the compound of formula (I) according to claim 3, characterized by an X-ray powder diffraction pattern expressed in 2 θ angles with diffraction peaks and percentage peak areas as shown in the following table:
Figure FDA0003656248850000012
Figure FDA0003656248850000021
5. the crystalline form a of the compound of formula (I) according to claim 4, wherein the crystalline form a of the compound of formula (I) satisfies one or more of the following conditions:
(1) The X-ray powder diffraction pattern of the crystal form A of the compound shown as the formula (I) expressed by 2 theta angles is basically shown as figure 3;
(2) A Differential Scanning Calorimetry (DSC) of said form a of the compound of formula (I) substantially as shown in figure 4;
(3) A thermogravimetric analysis (TGA) of crystalline form a of the compound of formula (I) is substantially as shown in figure 5;
(4) The dynamic water sorption Drawing (DVS) of form a of the compound of formula (I) may also be substantially as shown in figure 6.
6. A process for the preparation of form a of the compound of formula (I) according to any one of claims 1 to 5, which is process 1, process 2 or process 3:
the method comprises the following steps: cooling a heat saturated solution formed by the compound shown as the formula (I) and a solvent to room temperature at 40-80 ℃, and crystallizing; wherein the solvent is selected from one or more of ethanol, ethyl acetate, methyl isobutyl ketone, acetonitrile and methyl tert-butyl ether;
the method 2 comprises the following steps: mixing a solution formed by a compound shown as a formula (I) and a solvent A with a solvent B at 40-80 ℃, and crystallizing; wherein, the solvent A is one or more of ethanol, isopropanol, acetone, ethyl acetate, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran and 1,4-dioxane, and the solvent B is one or more of methyl tert-butyl ether, n-heptane and water;
the method 3 comprises the following steps: placing excessive compound shown as formula (I) in solvent to form suspension, suspending, separating solid phase from liquid phase, and drying; wherein the solvent is selected from one or more of ethanol, methyl isobutyl ketone, methyl tert-butyl ether, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran, 1,4-dioxane.
7. The process for the preparation of the compound of formula (I) in crystalline form a according to claim 6, wherein the process for the preparation of the compound of formula (I) in crystalline form a satisfies one or more of the following conditions:
(1) In the method 1, the method 2 or the method 3, the compound represented by the formula (I) is in an amorphous solid form;
(2) In the method 1, the volume-mass ratio of the solvent to the compound shown in the formula (I) is 10.0-30.0 mL/g;
(3) In the method 1, the crystallization is carried out in a cooling mode at a speed of 5-30 ℃/hour;
(4) In the method 1, before the crystallization, the hot saturated solution is filtered;
(5) In the method 2, the volume-mass ratio of the solvent A to the compound shown in the formula (I) is 8.0-11.0 mL/g;
(6) In the method 2, the temperature of 40-80 ℃ is 70-75 ℃ or 45-50 ℃;
(7) In the method 2, the volume ratio of the solvent A to the solvent B is (0.2-5): 1;
(8) In the method 2, the mixing is carried out by mixing a solution formed by the compound shown in the formula (I) and the solvent A with the solvent B, or cooling the solution formed by the compound shown in the formula (I) and the solvent A until solids are separated out and then mixing the solution with the solvent B;
(9) In the method 3, the solvent is selected from ethanol and/or methyl isobutyl ketone, for example, methyl isobutyl ketone;
(10) The suspension equilibrium temperature is 30-60 ℃;
(11) In the method 3, the suspension balancing time is 2 hours to 14 days.
8. A crystalline form B of the compound of formula (I) characterized by an X-ray powder diffraction pattern expressed in terms of 2 Θ angles having characteristic peaks at 5.0 ± 0.2 °, 9.8 ± 0.2 °, 14.7 ± 0.2 °, 19.6 ± 0.2 °, 24.6 ± 0.2 ° and 31.0 ± 0.2 °, said X-ray powder diffraction pattern being measured using the ka spectral line of the Cu target;
Figure FDA0003656248850000031
9. the crystalline form B of the compound of formula (I) according to claim 8, which satisfies one or more of the following conditions:
(1) Its X-ray powder diffraction pattern, expressed in terms of 2 theta angles, has the diffraction peaks and peak area percentages as shown in the following table:
numbering 2θ(±0.2°) Percent peak area (%)) 1 4.958 100.0 2 9.838 19.8 3 14.718 1.1 4 19.631 4.6 5 24.582 2.9 6 31.047 1.1
(2) The compound shown as the formula (I) has an endothermic peak at 132.0-142.5 ℃ in a Differential Scanning Calorimetry (DSC) of the crystal form B of the compound shown as the formula (I);
(3) In the dynamic water sorption Drawing (DVS) of the crystalline form B of the compound of formula (I), the weight gain of the crystalline form B is 0.95% in the range of 0% to 95% relative humidity compared to the initial weight.
10. The crystalline form B of the compound of formula (I) according to claim 9, which satisfies one or more of the following conditions:
(1) The X-ray powder diffraction pattern of the crystal form B of the compound shown as the formula (I) expressed by the 2 theta angle is shown as figure 7;
(2) The Differential Scanning Calorimetry (DSC) of the crystal form B of the compound shown in the formula (I) is shown in figure 8;
(3) The thermogravimetric analysis (TGA) of the crystalline form B of the compound represented by the formula (I) is shown in figure 9;
(4) The dynamic water absorption Diagram (DVS) of the crystal form A of the compound shown as the formula (I) is shown as a figure 10.
11. A process for the preparation of form B of the compound of formula (I) according to any one of claims 8 to 10, which is process a or process B:
the method A comprises the following steps: forming a hot saturated solution of the compound shown as the formula (I) and methanol at 50-70 ℃, and volatilizing the solvent at room temperature for crystallization;
the method B comprises the following steps: forming a suspension of the compound shown as the formula (I) and a solvent, after the suspension is balanced, separating a solid phase from a liquid phase, and drying; the solvent is methanol or a mixed solution of methanol and water.
12. The process for the preparation of the compound of formula (I) in crystalline form B according to claim 11, wherein said compound of formula (I) in crystalline form B is prepared in a manner that satisfies one or more of the following conditions:
(1) In the method A or the method B, the compound shown in the formula (I) is in an amorphous solid form;
(2) In the method A, before the volatile solvent crystallization step, the hot saturated solution is filtered;
(3) In the method a, the post-processing step is further included after the crystallization: drying;
(4) In the method B, when the solvent is a mixed solution of methanol and water, the volume ratio of the methanol to the water is (5-95): 5;
(5) In the method B, the suspension balancing time is 2 days to 14 days.
13. A crystal form C of a compound solvate shown as a formula (I) is characterized in that the compound solvate shown as the formula (I) is a methanol monohydrate of the compound shown as the formula (I),
Figure FDA0003656248850000041
the crystal form C belongs to a triclinic crystal system, a space group is P1, and unit cell parameters are as follows:
Figure FDA0003656248850000051
Figure FDA0003656248850000052
α =95.12 (3) °, β =93.82 (3) °, γ =90.43 (3) °; cell volume
Figure FDA0003656248850000053
The number of asymmetric units in the unit cell Z =1.
14. A pharmaceutical composition, which comprises one or more of the crystalline form a of the compound of formula (I) according to any one of claims 1 to 5, the crystalline form B of the compound of formula (I) according to any one of claims 8 to 10, and the crystalline form C of the solvate of the compound of formula (I) according to claim 13, and pharmaceutically acceptable excipients.
15. Use of a compound of formula (I) or a pharmaceutical composition of claim 14 for the preparation of a medicament for the treatment and/or prevention of a Ras-mediated disease;
Figure FDA0003656248850000054
16. use of a compound according to formula (I) or a pharmaceutical composition according to claim 14 for the preparation of a medicament for the treatment and/or prevention of cancer.
17. Use according to claim 15 or 16, wherein the use satisfies one or more of the following conditions:
(1) The compound shown as the formula (I) is the crystal form A of the compound shown as the formula (I) as described in any one of claims 1 to 5, the crystal form B of the compound shown as the formula (I) as described in any one of claims 8 to 10 or the crystal form C of the compound solvate shown as the formula (I) as described in claim 13;
(2) Ras is a G12C mutation of one or more of K-Ras, H-Ras and N-Ras, e.g., a G12C mutation of K-Ras;
(3) The Ras-mediated disease is cancer;
(4) The cancer is one or more of colon cancer, appendiceal cancer, pancreatic cancer, polyposis related to MYH, hematological cancer, breast cancer, endometrial cancer, gallbladder cancer, bile duct cancer, prostate cancer, lung cancer, brain cancer, ovarian cancer, cervical cancer, testicular cancer, kidney cancer, head or neck cancer, bone cancer, skin cancer, rectal cancer, liver cancer, esophageal cancer, stomach cancer, thyroid cancer, bladder cancer, lymphoma, leukemia and melanoma.
CN202210560179.0A 2021-05-24 2022-05-23 Crystal form of oxygen-containing heterocyclic compound, preparation method and application thereof Pending CN115385923A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021105675408 2021-05-24
CN202110567540 2021-05-24

Publications (1)

Publication Number Publication Date
CN115385923A true CN115385923A (en) 2022-11-25

Family

ID=84115516

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210560179.0A Pending CN115385923A (en) 2021-05-24 2022-05-23 Crystal form of oxygen-containing heterocyclic compound, preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN115385923A (en)
WO (1) WO2022247772A1 (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112390788A (en) * 2019-08-13 2021-02-23 苏州闻天医药科技有限公司 Compound for inhibiting KRASG12C mutant protein and preparation method and application thereof
AU2020395397A1 (en) * 2019-12-02 2022-06-09 Shanghai Yingli Pharmaceutical Co., Ltd Oxygen-containing heterocyclic compound, preparation method and application thereof
CN112979664B (en) * 2019-12-02 2024-03-01 上海璎黎药业有限公司 Oxygen-containing heterocyclic compound, preparation method and application thereof
US20230023009A1 (en) * 2020-06-05 2023-01-26 Sparcbio Llc Heterocyclic compounds and methods of use thereof

Also Published As

Publication number Publication date
WO2022247772A1 (en) 2022-12-01

Similar Documents

Publication Publication Date Title
JP7383652B2 (en) B-RAF Kinase Maleate Salt, Crystal Form, Preparation Method, and Use thereof
CN109970745B (en) Substituted pyrrolotriazine compound, pharmaceutical composition and application thereof
TWI634115B (en) Pladienolide pyridine compounds and methods of use
CN109232440B (en) Diphenylaminopyrimidine compounds for inhibiting kinase activity
JP7379680B2 (en) Novel salts of terphenyl compounds
CN108366994A (en) Novel Jak1 selective depressants and application thereof
CN105980389B (en) A kind of crystal form of disulfate of jak kinase inhibitor and preparation method thereof
TW201307345A (en) Crystalline (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-(1-methyl-1H-1,2,4-triazol-5-yl)-8,9-dihydro-2H-pyrido[4,3,2-de]phthalazin-3(7H)-one tosylate salt
EP3191475B1 (en) Crystalline forms of 6-((6, 7-dimethoxyquinazolin-4-yl)oxy) -n,2-dimethylbenzofuran-3-carboxamide
JP2024518429A (en) Solid forms of salts of 4-[5-[(3S)-3-aminopyrrolidine-1-carbonyl]-2-[2-fluoro-4-(2-hydroxy-2-ethylpropyl)phenyl]phenyl]-2-fluoro-benzonitrile
US9453011B2 (en) Crystal form of dabrafenib mesylate and preparation method thereof
CN112424202A (en) Crystal forms of active compounds inhibiting CDK4/6 and uses thereof
JP7054528B2 (en) Crystal form of compound that suppresses protein kinase activity and its application
WO2022247772A1 (en) Crystal forms of oxygen-containing heterocyclic compound, preparation method therefor and application thereof
CN110256408B (en) Diaryl pyrazole compound, composition containing compound and application of diaryl pyrazole compound
WO2018099451A1 (en) Crystal form of compound
EP3008071B1 (en) Polymorphic form of icotinib and uses thereof
WO2023116895A1 (en) Polymorph of kras inhibitor, preparation method therefor, and use thereof
WO2021073494A1 (en) The salts of a compound and the crystalline forms thereof
US20240174669A1 (en) Crystalline forms of a parp1 inhibitor
CN113861195B (en) Multi-condensed-ring EGFR inhibitor as well as preparation method and application thereof
WO2023093861A1 (en) Mono-p-toluenesulfonate of axl kinase inhibitor and crystal form thereof
JP5308030B2 (en) Solid forms of chemokine receptor antagonists and methods of use thereof
EP3008070B1 (en) Polymorph forms of icotinib maleate and uses thereof
CN117986263A (en) Crystal form of oxygen-containing heterocyclic compound, preparation method and application thereof

Legal Events

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