WO2025162419A1 - Crystal form of bicyclic derivative parp inhibitor, preparation method therefor, and use thereof - Google Patents
Crystal form of bicyclic derivative parp inhibitor, preparation method therefor, and use thereofInfo
- Publication number
- WO2025162419A1 WO2025162419A1 PCT/CN2025/075377 CN2025075377W WO2025162419A1 WO 2025162419 A1 WO2025162419 A1 WO 2025162419A1 CN 2025075377 W CN2025075377 W CN 2025075377W WO 2025162419 A1 WO2025162419 A1 WO 2025162419A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- crystalline form
- compound
- formula
- present
- day
- 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
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
- C07D487/04—Ortho-condensed systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic 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
- A61K31/4985—Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic 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
- A61K31/499—Spiro-condensed pyrazines or piperazines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07B—GENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
- C07B2200/00—Indexing scheme relating to specific properties of organic compounds
- C07B2200/13—Crystalline forms, e.g. polymorphs
Definitions
- the present invention relates to multiple crystal forms of a compound, a preparation method and applications thereof, and particularly to multiple crystal forms of a bicyclic derivative PARP inhibitor, a preparation method and applications thereof, belonging to the technical field of medicinal chemistry.
- BRCA1/2 genes are tumor suppressor genes that play an important role in DNA damage repair and normal cell growth. Mutations in these genes can inhibit the normal ability to repair DNA damage, causing homologous recombination deficiency (HRD). This is caused by loss of BRCA function or mutations or loss of function in other homologous recombination-related genes, preventing double-strand breaks from being repaired by homologous recombination (HRR), ultimately leading to cancer.
- HRD homologous recombination deficiency
- PARP Poly(ADP-ribose) polymerase
- PARP is a DNA repair enzyme that plays a key role in the DNA repair pathway. PARP is activated when DNA is damaged and broken. As a molecular sensor of DNA damage, it has the function of recognizing and binding to the site of DNA breakage, thereby activating and catalyzing the poly(ADP-ribosylation) of the receptor protein and participating in the DNA repair process. PARP plays a key role in the excision and repair of single-stranded DNA bases. In HRD tumor cells, double-stranded DNA cannot be repaired, and PARP inhibitors block single-strand repair, resulting in a "synthetic lethality" effect, leading to tumor cell death.
- PARP inhibitors have a "trapping" effect on PARP proteins, causing PARP proteins bound to damaged DNA to be trapped on the DNA and unable to get off. This directly prevents other DNA repair proteins from binding, ultimately leading to cell death.
- PARP inhibitors have been successfully developed, such as Olaparib, Rucaparib, and Niraparib.
- adverse reactions limit their ability to be used in combination with chemotherapy drugs. This may be related to the lack of selectivity of marketed PARP inhibitors for the PARP family.
- These side effects include intestinal toxicity caused by end-ankyropolymerase inhibition and hematotoxicity caused by PARP-2 inhibition. Therefore, it is of great clinical significance to develop highly selective PARP-1 inhibitors to reduce the related toxic side effects of non-selective PARP inhibitors.
- Patent WO2023227052 describes a compound of formula (I), which has good PARP inhibitory activity.
- the present invention provides compounds represented by formula (I), their crystal forms, preparation methods, pharmaceutical compositions, and their use in preparing medicaments for treating PARP-mediated diseases.
- the various crystal forms provided by the present invention have excellent properties, including high purity, good solubility, stable physical and chemical properties, ease of processing, crystallization, and handling, resistance to high temperatures, high humidity, and strong light, and low hygroscopicity.
- the present invention provides a crystalline form of a compound of formula (I):
- the crystalline form is crystalline form I, and its X-ray powder diffraction pattern using Cu-K ⁇ radiation has characteristic diffraction peaks at the following 2 ⁇ positions: 4.63° ⁇ 0.2°, 9.26° ⁇ 0.2°, 10.45° ⁇ 0.2°, 13.95° ⁇ 0.2° and 16.33° ⁇ 0.2°.
- the crystalline form is crystalline form I, and its X-ray powder diffraction pattern using Cu-K ⁇ radiation has characteristic diffraction peaks at the following 2 ⁇ positions: 4.63° ⁇ 0.2°, 9.26° ⁇ 0.2°, 10.45° ⁇ 0.2°, 13.95° ⁇ 0.2°, 15.97° ⁇ 0.2°, 16.33° ⁇ 0.2°, 17.14° ⁇ 0.2°, 20.62° ⁇ 0.2° and 22.37° ⁇ 0.2°.
- the crystalline form is Form I, and its X-ray powder diffraction pattern using Cu-K ⁇ radiation is substantially as shown in Figure 2.
- the differential scanning calorimetry (DSC) curve of Form I of the compound of formula (I) is substantially as shown in FIG3 .
- thermogravimetric analysis (TGA) curve of Form I of the compound of formula (I) shows a weight loss of about 0.13% before 100°C and a weight loss of about 0.34% from 100°C to 250°C.
- thermogravimetric analysis curve of Form I of the compound of formula (I) is substantially as shown in FIG4 .
- the hygroscopicity (DVS) isotherm curve of Form I of the compound of formula (I) shows that the water adsorption is 1.273% at 0 RH%-80% RH, indicating slight hygroscopicity.
- the isotherm adsorption curve is substantially as shown in FIG5 .
- the crystalline form is crystalline form II, and its X-ray powder diffraction pattern using Cu-K ⁇ radiation has characteristic diffraction peaks at the following 2 ⁇ positions: 7.59° ⁇ 0.2°, 11.41° ⁇ 0.2°, 11.79° ⁇ 0.2°, 14.53° ⁇ 0.2°, 17.40° ⁇ 0.2°, 21.54° ⁇ 0.2°, 24.93° ⁇ 0.2° and 25.45° ⁇ 0.2°.
- the crystalline form is crystalline form II, and its X-ray powder diffraction pattern using Cu-K ⁇ radiation has characteristic diffraction peaks at the following 2 ⁇ positions: 3.74° ⁇ 0.2°, 7.59° ⁇ 0.2°, 11.41° ⁇ 0.2°, 11.79° ⁇ 0.2°, 14.53° ⁇ 0.2°, 17.09° ⁇ 0.2°, 17.40° ⁇ 0.2°, 18.15° ⁇ 0.2°, 19.15° ⁇ 0.2°, 21.54° ⁇ 0.2°, 22.16° ⁇ 0.2°, 22.66° ⁇ 0.2°, 24.93° ⁇ 0.2°, 25.45° ⁇ 0.2° and 26.58° ⁇ 0.2°.
- Form II has an X-ray powder diffraction pattern substantially as shown in FIG6 using Cu-K ⁇ radiation.
- the differential scanning calorimetry curve of Form II of the compound of formula (I) is substantially as shown in FIG7 .
- thermogravimetric analysis (TGA) curve of Form II of the compound of formula (I) shows a weight loss of about 0.13% before 100°C and a weight loss of about 1.34% from 100°C to 270°C.
- thermogravimetric analysis curve of Form II of the compound of formula (I) is substantially as shown in FIG8 .
- the hygroscopicity (DVS) isotherm curve of Form II of the compound of formula (I) shows that the water adsorption at 0 RH%-80% RH is 1.148%, which is slightly hygroscopic.
- the isotherm adsorption curve is substantially as shown in FIG9 .
- the present invention also provides a pharmaceutical composition
- a pharmaceutical composition comprising a therapeutically effective amount of the aforementioned compound or any crystalline form thereof, and a pharmaceutically acceptable carrier and/or excipient, preferably wherein the therapeutically effective amount is 1-1440 mg.
- the pharmaceutical composition may be in the form of a unit dosage form (a unit dosage form is also referred to as a "dose strength").
- the present invention also provides use of the compound, crystal form, or composition of any of the aforementioned embodiments in the preparation of a medicament for treating a PARP-mediated disease.
- the PARP-mediated disease is selected from breast cancer, uterine cancer, cervical cancer, ovarian cancer, and prostate cancer.
- the present invention also provides a method for treating a PARP-mediated disease, comprising administering to a subject a therapeutically effective amount of a compound, crystalline form, or composition thereof according to any of the preceding embodiments, wherein the disease is preferably breast cancer, uterine cancer, cervical cancer, ovarian cancer, or prostate cancer, and wherein the therapeutically effective amount is preferably 1-1440 mg.
- the mammal of the present invention includes a human.
- an "effective amount” or “therapeutically effective amount” refers to administering a sufficient amount of a compound or crystalline form disclosed herein to alleviate, to some extent, one or more symptoms of the disease or condition being treated. In some embodiments, the result is a reduction and/or alleviation of the signs, symptoms, or causes of the disease, or any other desired change in a biological system.
- an "effective amount” for therapeutic uses is the amount of a composition comprising a compound or crystalline form disclosed herein required to provide a clinically significant reduction in disease symptoms.
- therapeutically effective amounts include, but are not limited to, 1-1440 mg, 1-1400 mg, 1-1300 mg, 1-1200 mg, 1-1000 mg, 1-900 mg, 1-800 mg, 1-700 mg, 1-600 mg, 1-500 mg, 1-400 mg, 1-300 mg, 1-250 mg, 1-200 mg, 1-150 mg, 1-125 mg, 1-100 mg, 1-80 mg, 1-60 mg, 1-50 mg, 1-40 mg, 1-25 mg, 1-20 mg, 5-1000 mg, 5-900 mg, 5-800 mg, 5-700 mg, 5-600 mg, 5 -500mg, 5-400mg, 5-300mg, 5-250mg, 5-200mg, 5-150mg, 5-125mg, 5-100mg, 5-90mg, 5-70mg, 5-80mg, 5-60mg, 5-50mg, 5-40mg, 5-30mg, 5-25mg, 5 -20mg, 10-1000mg, 10-900mg, 10-800mg, 10-700mg, 10-600mg, 10-500mg, 10-450mg, 10-400
- the pharmaceutical composition or formulation of the present invention contains the above-mentioned therapeutically effective amount of the compound or crystal form of the present invention
- the present invention relates to a pharmaceutical composition or pharmaceutical preparation comprising a therapeutically effective amount of a compound or crystalline form of the present invention and a carrier and/or excipient.
- the pharmaceutical composition may be in the form of a unit dosage form (the amount of the active ingredient in a unit dosage form is also referred to as the "dose strength").
- the pharmaceutical composition includes but is not limited to 1-1440 mg, 5-1000 mg, 10-800 mg, 20-600 mg, 25-500 mg, 40-200 mg, 50-100 mg, 1 mg, 1.25 mg, 2.5 mg, 5 mg, 10 mg, 12.5 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 125 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 , 700 mg, 725 mg, 750 mg, 775 mg, 800 mg, 850 mg, 900 mg, 950 mg, 1000 mg, 1100 mg, 1200 mg, 1300 mg, 1400 mg of a compound or crystalline form of the present invention.
- a method for treating a disease in a mammal comprising administering to a subject a therapeutically effective amount of a compound or crystalline form of the present invention, and a pharmaceutically acceptable carrier and/or excipient, the therapeutically effective amount preferably being 1-1500 mg, wherein the disease is selected from breast cancer, uterine cancer, cervical cancer, ovarian cancer, and prostate cancer.
- a method for treating a disease in a mammal comprising administering a compound or crystalline form of the present invention and a pharmaceutically acceptable carrier and/or excipient to a subject at a daily dose of 1-1440 mg/day, wherein the daily dose can be a single dose or divided doses.
- the daily dose includes but is not limited to 10-1440 mg/day, 20-1440 mg/day, 25-1440 mg/day, 50-1440 mg/day, 75-1440 mg/day, 100-1440 mg/day, 200-1440 mg/day, 10-1000 mg/day, 20-1000 mg/day, 25-1000 mg/day, 50-1000 mg/day, 75-1000 mg/day, 100-1000 mg/day, g/day, 200-1000 mg/day, 25-800 mg/day, 50-800 mg/day, 100-800 mg/day, 200-800 mg/day, 25-400 mg/day, 50-400 mg/day, 100-400 mg/day, 200-400 mg/day, in some embodiments, daily doses include but are not limited to 1 mg/day, 5 mg/day, 10 mg/day, 20 mg/day, 25 mg/day, 50 mg/day, 75 mg/day, 100 mg/day, 125 mg/day, 150 mg/day, 200 mg/day, 400 mg/day, 600 mg/day, 800 mg/
- the present invention relates to a kit, which may include a compound or a crystal form in a single-dose or multi-dose form.
- the kit contains the compound or crystal form of the present invention, and the amount of the compound or crystal form of the present invention is the same as that in the above-mentioned pharmaceutical composition.
- Preparation specifications refers to the weight of the main drug contained in each vial, tablet or other unit preparation.
- the compound or crystalline form of the present invention is present in an amount of about 5% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 10% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 15% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 20% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 25% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 30% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 35% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 40% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 45% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 50% to about 100% by weight of the drug substance; in certain embodiments, it is
- the drug substance is present at about 60% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 65% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 70% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 75% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 80% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 85% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 90% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 95% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 98% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 99% to about 100% by weight of the drug substance; in certain embodiments, substantially all of the drug substance is a substantially pure compound or
- the crystalline form of the present invention can be prepared by the following preparation method:
- Suspension method Add the compound of formula (I) to a selected single solvent or binary solvent until a suspension is formed. After suspension and stirring at room temperature to 50°C for a certain period of time (e.g., 1 hour to 3 days, or 2 hours to 24 hours, or 2 hours to 12 hours, or 3 hours to 5 hours), the suspension is centrifuged and dried to obtain the product.
- a certain period of time e.g., 1 hour to 3 days, or 2 hours to 24 hours, or 2 hours to 12 hours, or 3 hours to 5 hours
- Dissolution crystallization method dissolve the compound of formula (I) in a good solvent, add a certain amount of the solution dropwise to a poor solvent or add a poor solvent dropwise to the solution, stir to precipitate a solid, separate and dry to obtain the product.
- Cooling method dissolve a certain amount of sample in the corresponding solvent at high temperature, transfer the solution to room temperature for cooling, let it stand or stir for crystallization, separate, and dry to obtain the product.
- Thermal method experiment Take a certain amount of sample, place it on a glass slide and place it on a hot table. Heat it to the target temperature at a certain rate (such as 5-20°C/min, or 10-15°C/min), keep it at a constant temperature for a period of time (such as 0.5-5min, or 1-3min, or 1-2min), and then cool it naturally to room temperature to obtain a solid.
- a certain rate such as 5-20°C/min, or 10-15°C/min
- a constant temperature for a period of time such as 0.5-5min, or 1-3min, or 1-2min
- Vapor Diffusion Assay Add a suitable amount of a good solvent dropwise to a certain amount of the compound of formula (I) at room temperature until the sample is completely dissolved or a saturated solution of the good solvent is prepared. A certain amount of each solution is taken and the clear solution is placed in a poor solvent atmosphere and allowed to stand at room temperature until solid precipitates. This is followed by separation. Alternatively, the solid compound of formula (I) can be directly placed in a solvent atmosphere and allowed to stand at room temperature for 1 to 7 days to obtain the product.
- the good solvent and poor solvent of the present invention are relative.
- the one with higher solubility is a good solvent
- the one with lower solubility is a poor solvent.
- the good solvent is selected from ethylene glycol methyl ether, ethylene glycol dimethyl ether, dioxane, DMF, DMSO, methanol, ethanol, n-propyl alcohol, butyl formate, 4-methyl-2-pentanone, tetrahydrofuran, isopropyl alcohol, ethyl acetate, n-heptane, ether, water, acetonitrile, toluene, chloroform, acetone, butyl formate, MTBE, and cyclohexane with higher solubility
- the poor solvent is selected from the above-mentioned solvent with lower solubility.
- the good solvent is selected from ethylene glycol methyl ether, ethylene glycol dimethyl ether, dioxane, DMF, DMSO, methanol, ethanol, n-propyl alcohol, butyl formate, 4-methyl-2-pentanone, tetrahydrofuran, or a mixed solvent thereof.
- the poor solvent is selected from isopropyl alcohol, ethyl acetate, n-heptane, diethyl ether, water, acetonitrile, toluene, chloroform, acetone, butyl formate, MTBE, cyclohexane, or a mixed solvent thereof.
- the solvents of the evaporation method are water and acetone
- the solvent used in the dissolution crystallization method is dichloromethane and n-heptane; in some embodiments, the solvent used in the dissolution crystallization method is dichloromethane and isopropyl ether; in some embodiments, the solvent used in the dissolution crystallization method is ethyl acetate and n-heptane; in some embodiments, the solvent used in the dissolution crystallization method is 4-methyl-2-pentanone and n-heptane; in some embodiments, the solvent used in the dissolution crystallization method is dioxane and n-heptane;
- the solvents used in the cooling method are ethylene glycol dimethyl ether and cyclohexane; in some embodiments, the solvents used in the cooling method are toluene and cyclohexane; in some embodiments, the solvents used in the cooling method are isopropyl acetate and cyclohexane; in some embodiments, the solvents used in the cooling method are dioxane and cyclohexane; in some embodiments, the solvents used in the cooling method are dioxane and water; in some embodiments, the solvents used in the cooling method are DMSO and water.
- the vapor diffusion method employs vapor diffusion in ethyl acetate.
- the good solvent and poor solvent described in the present invention are relative.
- the one with higher solubility is a good solvent
- the one with lower solubility is a poor solvent.
- the solvent used in the above preparation method may be a single solvent or a combination of two or more solvents.
- IC50 refers to the half-maximal inhibitory concentration, which is the concentration at which half of the maximal inhibitory effect is achieved.
- the crystal of the present invention As used herein, “the crystal of the present invention”, “the crystal form of the present invention”, “the crystal form of the present invention” and the like can be used interchangeably.
- room temperature generally refers to 4-30°C, preferably 20 ⁇ 5°C.
- the crystalline structure of the present invention can be analyzed using various analytical techniques known to those skilled in the art, including but not limited to, X-ray powder diffraction (XRD), differential scanning calorimetry (DSC) and/or thermogravimetric analysis (TGA), also known as thermogravimetry (TG).
- XRD X-ray powder diffraction
- DSC differential scanning calorimetry
- TGA thermogravimetric analysis
- TG thermogravimetry
- 2 ⁇ or 2 ⁇ angle refers to the peak position expressed in degrees (°) based on the setup of an X-ray diffraction experiment, and is typically the unit of the abscissa in a diffraction pattern. If the incident beam forms an angle ⁇ with a certain lattice plane and the reflection is diffracted, the experimental setup requires recording the reflected beam in 2 ⁇ angles. It should be understood that the specific 2 ⁇ value of a specific crystal form mentioned herein is intended to represent the 2 ⁇ value (expressed in degrees) measured using the X-ray diffraction experimental conditions described herein, and the error range of the 2 ⁇ may be ⁇ 0.3, ⁇ 0.2, or ⁇ 0.1.
- crystal form of the present invention is not limited to the characteristic spectra that are exactly the same as the characteristic spectra described in the drawings disclosed in the present invention, such as XRD, DSC, TGA, and DVS. Any crystal form having characteristic spectra that are substantially the same or essentially the same as those described in the drawings falls within the scope of the present invention.
- the melting peak height of a DSC curve depends on many factors related to sample preparation and instrument geometry, while the peak position is relatively insensitive to experimental details. Therefore, in some embodiments, the crystalline compound of the present invention is characterized by a DSC pattern having a characteristic peak position, having substantially the same properties as the DSC pattern provided in the accompanying drawings of the present invention, with a measurement error tolerance of within ⁇ 5°C, generally required to be within ⁇ 3°C.
- Carrier refers to a system that does not cause significant irritation to the organism and does not eliminate the biological activity and properties of the administered compound, and can change the way the drug enters the human body and its distribution in the body, control the release rate of the drug and deliver the drug to the target organ.
- Non-limiting examples include microcapsules and microspheres, nanoparticles, liposomes, etc.
- excipient is a substance that is not itself a therapeutic agent but serves as a diluent, adjuvant, binder, and/or vehicle that is added to a pharmaceutical composition to improve its handling or storage properties or to allow or facilitate the formation of a compound or pharmaceutical composition into a unit dosage form for administration.
- pharmaceutical excipients can serve a variety of functions and can be described as wetting agents, buffers, suspending agents, lubricants, emulsifiers, disintegrants, absorbents, preservatives, surfactants, colorants, flavoring agents, and sweeteners.
- Examples of pharmaceutical excipients include, but are not limited to: (1) sugars such as lactose, glucose, and sucrose; (2) starches such as corn starch and potato starch; (3) cellulose and its derivatives such as sodium carboxymethylcellulose, ethylcellulose, cellulose acetate, hydroxypropyl methylcellulose, hydroxypropyl cellulose, microcrystalline cellulose, and cross-linked carboxymethylcellulose (e.g., cross-linked sodium carboxymethylcellulose); (4) tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients such as cocoa butter and suppository waxes; (9) oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn starch, and maltodextrin.
- sugars such as lactose, glucose, and sucrose
- starches such as corn starch and potato starch
- cellulose and its derivatives such as sodium carboxymethylcellulose, ethylcellulose,
- glycols such as propylene glycol
- polyols such as glycerol, sorbitol, mannitol and polyethylene glycol
- esters such as ethyl oleate and ethyl laurate
- agar such as agar
- buffers such as magnesium hydroxide and aluminum hydroxide
- FIG1 is an X-ray powder diffraction pattern of the amorphous compound represented by formula (I).
- Figure 2 is an X-ray powder diffraction pattern of Form I of the compound represented by formula (I).
- FIG3 is a differential scanning calorimetry analysis curve of Form I of the compound represented by formula (I).
- FIG4 is a thermogravimetric analysis spectrum of Form I of the compound represented by formula (I).
- FIG5 is an isothermal adsorption curve of Form I of the compound represented by formula (I).
- FIG6 is an X-ray powder diffraction pattern of Form II of the compound represented by formula (I).
- FIG7 is a differential scanning calorimetry analysis curve of Form II of the compound represented by formula (I).
- FIG8 is a thermogravimetric analysis spectrum of Form II of the compound represented by formula (I).
- FIG9 is an isothermal adsorption curve of Form II of the compound represented by formula (I).
- NMR nuclear magnetic resonance
- MS mass spectrometry
- MS was performed using (Agilent 6120B (ESI) and Agilent 6120B (APCI)).
- HPLC determination was performed using a LC-20AT (Shimadzu) high pressure liquid chromatograph (Shim-pack GIST C18, 4.6 ⁇ 250 mm (HSS), 5 ⁇ m).
- the known starting materials of the present invention can be synthesized by methods known in the art, or can be purchased from companies such as Titan Technology, Anage Chemical, Shanghai Demer, Chengdu Kelon Chemical, Shaoyuan Chemical Technology, and Bailingwei Technology.
- the solution refers to an aqueous solution.
- the room temperature is 20°C to 30°C.
- Step 3 tert-Butyl 4-(2-(difluoromethyl)imidazo[1,2-a]pyrazin-6-yl)piperazine-1-carboxylate (1C)
- Step 4 7-((4-(2-(difluoromethyl)imidazo[1,2-a]pyrazin-6-yl)piperazin-1-yl)methyl)-8-fluoro-3-methylquinoxalin-2(1H)-one (Formula I)
- Example 1 50 mg of the compound of formula (I) obtained in Example 1 was added to 20 mL of a mixed solvent of acetonitrile and water, dissolved and then freeze-dried.
- the amorphous form of the compound of formula (I) was characterized by XRD. Its X-ray powder diffraction pattern is shown in Figure 1.
- Form I of the compound represented by formula (I) was characterized by XRD, DSC, TGA and DVS, and its X-ray powder diffraction pattern, differential scanning calorimetry analysis curve, thermogravimetric analysis diagram, and isothermal adsorption curve are shown in Figures 2-5, respectively.
- thermodynamic stability of form I is better than that of form II.
- a TA Instruments Q200DSC differential scanning calorimeter was used. A 0.5–5 mg sample was accurately weighed and placed in a perforated DSC Tzero pan. The sample was heated to the final temperature at a rate of 10°C/min, with a nitrogen purge rate of 50 mL/min.
- thermogravimetric analyzer was a TA Instruments Q500TGA. Samples (1–10 mg) were placed in a pre-equilibrated open aluminum sample pan and automatically weighed within the TGA furnace. The samples were heated to the final temperature at a rate of 10°C/min, with nitrogen purge rates of 60 mL/min at the sample and 40 mL/min at the balance.
- 0.1 ⁇ L of compound in 100% DMSO was delivered to a 384-well plate (Corning 4514) using acoustic liquid delivery technology (Echo 655) and centrifuged at 1000 rpm for 1 minute.
- 5 ⁇ L of PARP enzyme solution was transferred to a 384-well plate and centrifuged at 1000 rpm for 1 minute, followed by incubation at 25°C for 10 minutes.
- 5 ⁇ L of substrate solution was transferred to a 384-well plate, centrifuged at 1000 rpm for 1 minute, and incubated at 25°C for 60 minutes.
- mP signal of FP (ex/em: 485 nm/520 nm) was read using the fluorescence polarization module of the BMG PHERAstar FSX.
- GraphPad Prism 8 software was then used to perform a four-parameter nonlinear regression fit to the IC50 value.
- IC50 values of the compound of formula (I) against PARP-1 and PARP-2 in vitro are expressed on a scale of A, B, C, or D, where A represents 0 ⁇ IC50 ⁇ 10 nM , B represents 10 nM ⁇ IC50 ⁇ 50 nM , C represents 50 nM ⁇ IC50 ⁇ 500 nM, and D represents 500 nM ⁇ IC50 .
- the compound of formula (I) of the present invention has a significant selective inhibitory effect on PARP-1 enzyme activity in vitro.
- mice Male Balb/c mice, 20-25 g, 12 mice per compound, purchased from Chengdu Dashuo Experimental Animal Co., Ltd.
- mice were randomly divided into groups according to body weight. They were fasted but not watered for 12-14 hours before administration and fed 4 hours after administration.
- Intravenous administration solvent 10% DMA + 10% Solutol + 80% Saline; oral administration solvent: 5% DMSO + 30% PEG400 + 65% (20% SBE-CD), Solutol is polyethylene glycol-15-hydroxystearate; Saline is normal saline
- the compound of formula (I) has excellent pharmacokinetic characteristics in mouse plasma.
- Crystal samples were taken and placed independently at 40°C + 75% RH and 30°C + 60% RH for 0 month, 1 month, 2 months, 3 months, 6 months and 9 months, respectively.
- the purity was tested by HPLC.
- the results showed that the chemical stability of crystal forms I and II of the compound of formula (I) of the present invention was good.
Landscapes
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Epidemiology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Description
本发明涉及一种化合物的多种晶型及其制备方法和应用,具体涉及一种双环衍生物PARP抑制剂的多种晶型及其制备方法和应用,属于药物化学技术领域。The present invention relates to multiple crystal forms of a compound, a preparation method and applications thereof, and particularly to multiple crystal forms of a bicyclic derivative PARP inhibitor, a preparation method and applications thereof, belonging to the technical field of medicinal chemistry.
大约5%的乳腺癌患者与BRCA1/2基因胚系突变相关(BRCA1基因3%,BRCA2基因2%)。BRCA1突变导致的乳腺癌大部分为三阴性乳腺癌(70%),而BRCA2突变更可能导致雌激素受体阳性乳腺癌(70%)。BRCA1/2基因是抑癌基因,在DNA损伤修复、细胞正常生长等方面均具有重要作用。该基因突变可抑制DNA损伤后正常修复能力,引起同源重组缺陷(homologous recombination deficiency,HRD),即BRCA功能缺失或其他同源重组相关基因发生突变或功能缺失,使双链断裂的DNA修复不能通过同源重组修复(homologous recombinant repair,HRR),最终导致癌变。Approximately 5% of breast cancer patients are associated with germline mutations in the BRCA1/2 genes (3% for BRCA1 and 2% for BRCA2). Breast cancers caused by BRCA1 mutations are mostly triple-negative (70%), while BRCA2 mutations are more likely to cause estrogen receptor-positive (ER-positive) breast cancer (70%). The BRCA1/2 genes are tumor suppressor genes that play an important role in DNA damage repair and normal cell growth. Mutations in these genes can inhibit the normal ability to repair DNA damage, causing homologous recombination deficiency (HRD). This is caused by loss of BRCA function or mutations or loss of function in other homologous recombination-related genes, preventing double-strand breaks from being repaired by homologous recombination (HRR), ultimately leading to cancer.
聚腺苷二磷酸核糖聚合酶(PARP)是一种DNA修复酶,在DNA修复通路中起关键作用。DNA损伤断裂时会激活PARP,它作为DNA损伤的一种分子感受器,具有识别、结合到DNA断裂位置的功能,进而激活、催化受体蛋白的聚ADP核糖基化作用,参与DNA的修复过程。PARP在DNA单链碱基切除、修复过程中发挥关键作用。在HRD肿瘤细胞中DNA双链无法修复,PARP抑制剂又阻断单链修复,从而形成“合成致死”效应,导致肿瘤细胞死亡。Poly(ADP-ribose) polymerase (PARP) is a DNA repair enzyme that plays a key role in the DNA repair pathway. PARP is activated when DNA is damaged and broken. As a molecular sensor of DNA damage, it has the function of recognizing and binding to the site of DNA breakage, thereby activating and catalyzing the poly(ADP-ribosylation) of the receptor protein and participating in the DNA repair process. PARP plays a key role in the excision and repair of single-stranded DNA bases. In HRD tumor cells, double-stranded DNA cannot be repaired, and PARP inhibitors block single-strand repair, resulting in a "synthetic lethality" effect, leading to tumor cell death.
PARP抑制剂对PARP蛋白有“诱捕”作用,导致与受损DNA结合的PARP蛋白被困在DNA上下不来了,直接造成其他的DNA修复蛋白也结合不上来了,最终导致细胞死亡。目前已有多款PARP抑制剂被成功开发,如奥拉帕利,卢卡帕利和尼拉帕利等,然而不良反应限制了其与化疗药物联合使用的能力。这可能与上市的PARP抑制剂缺少对PARP家族的选择性有关,这些副作用包括端锚聚合酶抑制引起的肠道毒性和PARP-2抑制导致的血液毒性。因此开发高选择性的PARP-1抑制剂,降低非选择性的PARP抑制剂的相关毒副作用具有重要的临床意义。PARP inhibitors have a "trapping" effect on PARP proteins, causing PARP proteins bound to damaged DNA to be trapped on the DNA and unable to get off. This directly prevents other DNA repair proteins from binding, ultimately leading to cell death. Currently, a number of PARP inhibitors have been successfully developed, such as Olaparib, Rucaparib, and Niraparib. However, adverse reactions limit their ability to be used in combination with chemotherapy drugs. This may be related to the lack of selectivity of marketed PARP inhibitors for the PARP family. These side effects include intestinal toxicity caused by end-ankyropolymerase inhibition and hematotoxicity caused by PARP-2 inhibition. Therefore, it is of great clinical significance to develop highly selective PARP-1 inhibitors to reduce the related toxic side effects of non-selective PARP inhibitors.
WO2023227052专利中记载了一种式(I)化合物,该化合物具有良好的PARP抑制活性。
Patent WO2023227052 describes a compound of formula (I), which has good PARP inhibitory activity.
选择稳定的、可重复制造的、并且具备有利于其作为治疗剂使用的物理化学特性的结晶剂形式是非常重要的。It is important to select a crystalline agent form that is stable, reproducible, and possesses physicochemical properties that are favorable for its use as a therapeutic agent.
本发明提供式(I)所示化合物、其晶型及制备方法、药物组合物,以及它们用于制备治疗PARP介导的疾病的药物的用途。本发明提供的多种晶型物具有纯度高、溶解性好、物理和化学性质稳定,易于加工和结晶、处理,能耐高温、高湿及强光照,引湿性低等优异特性。The present invention provides compounds represented by formula (I), their crystal forms, preparation methods, pharmaceutical compositions, and their use in preparing medicaments for treating PARP-mediated diseases. The various crystal forms provided by the present invention have excellent properties, including high purity, good solubility, stable physical and chemical properties, ease of processing, crystallization, and handling, resistance to high temperatures, high humidity, and strong light, and low hygroscopicity.
本发明提供一种式(I)化合物的晶型物:
The present invention provides a crystalline form of a compound of formula (I):
在某些具体实施方案中所述晶型物为晶型I,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:4.63°±0.2°、9.26°±0.2°、10.45°±0.2°、13.95°±0.2°和16.33°±0.2°。In certain specific embodiments, the crystalline form is crystalline form I, and its X-ray powder diffraction pattern using Cu-Kα radiation has characteristic diffraction peaks at the following 2θ positions: 4.63°±0.2°, 9.26°±0.2°, 10.45°±0.2°, 13.95°±0.2° and 16.33°±0.2°.
在某些具体实施方案中,所述晶型物为晶型I,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:4.63°±0.2°、9.26°±0.2°、10.45°±0.2°、13.95°±0.2°、15.97°±0.2°、16.33°±0.2°、17.14°±0.2°、20.62°±0.2°和22.37°±0.2°。In certain specific embodiments, the crystalline form is crystalline form I, and its X-ray powder diffraction pattern using Cu-Kα radiation has characteristic diffraction peaks at the following 2θ positions: 4.63°±0.2°, 9.26°±0.2°, 10.45°±0.2°, 13.95°±0.2°, 15.97°±0.2°, 16.33°±0.2°, 17.14°±0.2°, 20.62°±0.2° and 22.37°±0.2°.
在某些具体实施方案中,所述晶型物为晶型I,使用Cu-Kα辐射,其X-射线粉末衍射图基本如图2所示。In certain specific embodiments, the crystalline form is Form I, and its X-ray powder diffraction pattern using Cu-Kα radiation is substantially as shown in Figure 2.
在某些具体实施方案中,其差示扫描量热分析曲线(DSC)显示晶型I的峰值温度为274.95℃,ΔH=97.746J/g。In certain specific embodiments, the differential scanning calorimetry (DSC) curve thereof shows that the peak temperature of Form I is 274.95° C., and ΔH=97.746 J/g.
在某些具体实施方案中,式(I)化合物的晶型I,其差示扫描量热分析曲线(DSC)基本如图3所示。In certain specific embodiments, the differential scanning calorimetry (DSC) curve of Form I of the compound of formula (I) is substantially as shown in FIG3 .
在某些具体实施方案中,式(I)化合物的晶型I,其热重分析曲线(TGA)显示在100℃之前失重约0.13%,100℃到250℃失重约0.34%。In certain embodiments, the thermogravimetric analysis (TGA) curve of Form I of the compound of formula (I) shows a weight loss of about 0.13% before 100°C and a weight loss of about 0.34% from 100°C to 250°C.
在某些具体实施方案中,式(I)化合物的晶型I,其热重分析曲线基本如图4所示。In certain specific embodiments, the thermogravimetric analysis curve of Form I of the compound of formula (I) is substantially as shown in FIG4 .
在某些具体实施方案中,式(I)化合物的晶型I,其引湿性(DVS)等温曲线显示在0RH%-80%RH的水分吸附为1.273%,略有引湿性,其等温吸附曲线基本如图5所示。In certain specific embodiments, the hygroscopicity (DVS) isotherm curve of Form I of the compound of formula (I) shows that the water adsorption is 1.273% at 0 RH%-80% RH, indicating slight hygroscopicity. The isotherm adsorption curve is substantially as shown in FIG5 .
在某些具体实施方案中,所述晶型物为晶型II,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:7.59°±0.2°、11.41°±0.2°、11.79°±0.2°、14.53°±0.2°、17.40°±0.2°、21.54°±0.2°、24.93°±0.2°和25.45°±0.2°。In certain specific embodiments, the crystalline form is crystalline form II, and its X-ray powder diffraction pattern using Cu-Kα radiation has characteristic diffraction peaks at the following 2θ positions: 7.59°±0.2°, 11.41°±0.2°, 11.79°±0.2°, 14.53°±0.2°, 17.40°±0.2°, 21.54°±0.2°, 24.93°±0.2° and 25.45°±0.2°.
在某些具体实施方案中,所述晶型物为晶型II,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:3.74°±0.2°、7.59°±0.2°、11.41°±0.2°、11.79°±0.2°、14.53°±0.2°、17.09°±0.2°、17.40°±0.2°、18.15°±0.2°、19.15°±0.2°、21.54°±0.2°、22.16°±0.2°、22.66°±0.2°、24.93°±0.2°、25.45°±0.2°和26.58°±0.2°。In certain specific embodiments, the crystalline form is crystalline form II, and its X-ray powder diffraction pattern using Cu-Kα radiation has characteristic diffraction peaks at the following 2θ positions: 3.74°±0.2°, 7.59°±0.2°, 11.41°±0.2°, 11.79°±0.2°, 14.53°±0.2°, 17.09°±0.2°, 17.40°±0.2°, 18.15°±0.2°, 19.15°±0.2°, 21.54°±0.2°, 22.16°±0.2°, 22.66°±0.2°, 24.93°±0.2°, 25.45°±0.2° and 26.58°±0.2°.
在某些具体实施方案中,晶型II使用Cu-Kα辐射,其X-射线粉末衍射图基本如图6所示。In certain specific embodiments, Form II has an X-ray powder diffraction pattern substantially as shown in FIG6 using Cu-Kα radiation.
在某些具体实施方案中,其差示扫描量热分析曲线(DSC)显示晶型II的峰值温度为277.66℃,ΔH=89.910J/g。In certain specific embodiments, the differential scanning calorimetry (DSC) curve thereof shows that the peak temperature of Form II is 277.66° C., and ΔH=89.910 J/g.
在某些具体实施方案中,式(I)化合物的晶型II,其差示扫描量热分析曲线基本如图7所示。In certain specific embodiments, the differential scanning calorimetry curve of Form II of the compound of formula (I) is substantially as shown in FIG7 .
在某些具体实施方案中,式(I)化合物的晶型II,其热重分析曲线(TGA)显示在100℃之前失重约0.13%,100℃到270℃失重约1.34%。In certain embodiments, the thermogravimetric analysis (TGA) curve of Form II of the compound of formula (I) shows a weight loss of about 0.13% before 100°C and a weight loss of about 1.34% from 100°C to 270°C.
在某些具体实施方案中,式(I)化合物的晶型II,其热重分析曲线基本如图8所示。In certain specific embodiments, the thermogravimetric analysis curve of Form II of the compound of formula (I) is substantially as shown in FIG8 .
在某些具体实施方案中,式(I)化合物的晶型II,其引湿性(DVS)等温曲线显示在0RH%-80%RH的水分吸附为1.148%,略有引湿性,其等温吸附曲线基本如图9所示。In certain specific embodiments, the hygroscopicity (DVS) isotherm curve of Form II of the compound of formula (I) shows that the water adsorption at 0 RH%-80% RH is 1.148%, which is slightly hygroscopic. The isotherm adsorption curve is substantially as shown in FIG9 .
本发明还提供一种药物组合物,其中,所述药物组合物含有治疗有效量的前述化合物或任一晶型物,以及药学上可接受的载体和/或赋形剂,优选所述治疗有效量为1-1440mg。该药物组合物可以为单位制剂形式(单位制剂也被称为“制剂规格”)。The present invention also provides a pharmaceutical composition comprising a therapeutically effective amount of the aforementioned compound or any crystalline form thereof, and a pharmaceutically acceptable carrier and/or excipient, preferably wherein the therapeutically effective amount is 1-1440 mg. The pharmaceutical composition may be in the form of a unit dosage form (a unit dosage form is also referred to as a "dose strength").
本发明还提供前述任意一项方案所述的化合物、晶型物或组合物在制备治疗PARP介导的疾病的药物中的用途。进一步地,所述PARP介导的疾病选自乳腺癌、子宫癌、宫颈癌、卵巢癌、前列腺癌。The present invention also provides use of the compound, crystal form, or composition of any of the aforementioned embodiments in the preparation of a medicament for treating a PARP-mediated disease. Furthermore, the PARP-mediated disease is selected from breast cancer, uterine cancer, cervical cancer, ovarian cancer, and prostate cancer.
本发明还提供了一种用于治疗PARP介导的疾病的方法,所述方法包括给予受试者治疗有效量的前述任意一项方案所述的化合物、晶型物或其组合物,所述疾病优选为乳腺癌、子宫癌、宫颈癌、卵巢癌、前列腺癌,优选所述治疗有效量为1-1440mg。一些实施方案中,本发明中所述哺乳动物包括人。The present invention also provides a method for treating a PARP-mediated disease, comprising administering to a subject a therapeutically effective amount of a compound, crystalline form, or composition thereof according to any of the preceding embodiments, wherein the disease is preferably breast cancer, uterine cancer, cervical cancer, ovarian cancer, or prostate cancer, and wherein the therapeutically effective amount is preferably 1-1440 mg. In some embodiments, the mammal of the present invention includes a human.
本申请中所述“有效量”或“治疗有效量”是指给予足够量的本申请公开的化合物或晶型物,其将在某种程度上缓解所治疗的疾病或病症的一种或多种症状。在一些实施方案中,结果是减少和/或缓和疾病的体征、症状或原因,或生物系统的任何其它希望改变。例如,针对治疗用途的“有效量”是提供临床上显著的疾病症状降低所需的包含本申请公开的化合物或晶型物的组合物的量。治疗有效量的实例包括但不限于1-1440mg、1-1400mg、1-1300mg、1-1200mg、1-1000mg、1-900mg、1-800mg、1-700mg、1-600mg、1-500mg、1-400mg、1-300mg、1-250mg、1-200mg、1-150mg、1-125mg、1-100mg、1-80mg、1-60mg、1-50mg、1-40mg、1-25mg、1-20mg、5-1000mg、5-900mg、5-800mg、5-700mg、5-600mg、5-500mg、5-400mg、5-300mg、5-250mg、5-200mg、5-150mg、5-125mg、5-100mg、5-90mg、5-70mg、5-80mg、5-60mg、5-50mg、5-40mg、5-30mg、5-25mg、5-20mg、10-1000mg、10-900mg、10-800mg、10-700mg、10-600mg、10-500mg、10-450mg、10-400mg、10-300mg、10-250mg、10-200mg、10-150mg、10-125mg、10-100mg、10-90mg、10-80mg、10-70mg、10-60mg、10-50mg、10-40mg、10-30mg、10-20mg;20-1000mg、20-900mg、20-800mg、20-700mg、20-600mg、20-500mg、20-400mg、20-350mg、20-300mg、20-250mg、20-200mg、20-150mg、20-125mg、20-100mg、20-90mg、20-80mg、20-70mg、20-60mg、20-50mg、20-40mg、20-30mg;50-1000mg、50-900mg、50-800mg、50-700mg、50-600mg、50-500mg、50-400mg、50-300mg、50-250mg、50-200mg、50-150mg、50-125mg、50-100mg;100-1000mg、100-900mg、100-800mg、100-700mg、100-600mg、100-500mg、100-400mg、100-300mg、100-250mg、100-200mg;As used herein, an "effective amount" or "therapeutically effective amount" refers to administering a sufficient amount of a compound or crystalline form disclosed herein to alleviate, to some extent, one or more symptoms of the disease or condition being treated. In some embodiments, the result is a reduction and/or alleviation of the signs, symptoms, or causes of the disease, or any other desired change in a biological system. For example, an "effective amount" for therapeutic uses is the amount of a composition comprising a compound or crystalline form disclosed herein required to provide a clinically significant reduction in disease symptoms. Examples of therapeutically effective amounts include, but are not limited to, 1-1440 mg, 1-1400 mg, 1-1300 mg, 1-1200 mg, 1-1000 mg, 1-900 mg, 1-800 mg, 1-700 mg, 1-600 mg, 1-500 mg, 1-400 mg, 1-300 mg, 1-250 mg, 1-200 mg, 1-150 mg, 1-125 mg, 1-100 mg, 1-80 mg, 1-60 mg, 1-50 mg, 1-40 mg, 1-25 mg, 1-20 mg, 5-1000 mg, 5-900 mg, 5-800 mg, 5-700 mg, 5-600 mg, 5 -500mg, 5-400mg, 5-300mg, 5-250mg, 5-200mg, 5-150mg, 5-125mg, 5-100mg, 5-90mg, 5-70mg, 5-80mg, 5-60mg, 5-50mg, 5-40mg, 5-30mg, 5-25mg, 5 -20mg, 10-1000mg, 10-900mg, 10-800mg, 10-700mg, 10-600mg, 10-500mg, 10-450mg, 10-400mg, 10-300mg, 10-250mg, 10-200mg, 10-150mg, 10-12 5mg, 10-100mg, 10-90mg, 10-80mg, 10-70mg, 10-60mg, 10-50mg, 10-40mg, 10-30mg, 10-20mg; 20-1000mg, 20-900mg, 20-800mg, 20-700mg, 20-60 0mg, 20-500mg, 20-400mg, 20-350mg, 20-300mg, 20-250mg, 20-200mg, 20-150mg, 20-125mg, 20-100mg, 20-90mg, 20-80mg, 20-70mg, 20-60mg, 20- 50mg, 20-40mg, 20-30mg; 50-1000mg, 50-900mg, 50-800mg, 50-700mg, 50-600mg, 50-500mg, 50-400mg, 50-300mg, 50-250mg, 50-200mg, 50-150mg , 50-125mg, 50-100mg; 100-1000mg, 100-900mg, 100-800mg, 100-700mg, 100-600mg, 100-500mg, 100-400mg, 100-300mg, 100-250mg, 100-200mg;
在一些实施方案中,本发明的药物组合物或制剂含有上述治疗有效量的本发明化合物或晶型物;In some embodiments, the pharmaceutical composition or formulation of the present invention contains the above-mentioned therapeutically effective amount of the compound or crystal form of the present invention;
本发明涉及一种药物组合物或药物制剂,所述的药物组合物或药物制剂包含治疗有效量的本发明所述的化合物或晶型物以及载体和/或赋形剂。该药物组合物可以为单位制剂形式(单位制剂中主药的量也被称为“制剂规格”)。在一些实施方案中,该药物组合物包括但不限于1-1440mg、5-1000mg、10-800mg、20-600mg、25-500mg、40-200mg、50-100mg、1mg、1.25mg、2.5mg、5mg、10mg、12.5mg、15mg、20mg、25mg、30mg、35mg、40mg、45mg、50mg、55mg、60mg、65mg、70mg、75mg、80mg、85mg、90mg、95mg、100mg、110mg、120mg、125mg、130mg、140mg、150mg、160mg、170mg、180mg、190mg、200mg、210mg、220mg、230mg、240mg、250mg、275mg、300mg、325mg、350mg、375mg、400mg、425mg、450mg、475mg、500mg、525mg、550mg、575mg、600mg、625mg、650mg、675mg、700mg、725mg、750mg、775mg、800mg、850mg、900mg、950mg、1000mg、1100mg、1200mg、1300mg、1400mg、1440mg的本发明化合物或晶型物。The present invention relates to a pharmaceutical composition or pharmaceutical preparation comprising a therapeutically effective amount of a compound or crystalline form of the present invention and a carrier and/or excipient. The pharmaceutical composition may be in the form of a unit dosage form (the amount of the active ingredient in a unit dosage form is also referred to as the "dose strength"). In some embodiments, the pharmaceutical composition includes but is not limited to 1-1440 mg, 5-1000 mg, 10-800 mg, 20-600 mg, 25-500 mg, 40-200 mg, 50-100 mg, 1 mg, 1.25 mg, 2.5 mg, 5 mg, 10 mg, 12.5 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 125 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 , 700 mg, 725 mg, 750 mg, 775 mg, 800 mg, 850 mg, 900 mg, 950 mg, 1000 mg, 1100 mg, 1200 mg, 1300 mg, 1400 mg of a compound or crystalline form of the present invention.
一种用于治疗哺乳动物的疾病的方法,所述方法包括给予受试者治疗有效量的本发明化合物或晶型物,以及药学上可接受的载体和/或赋形剂,治疗有效量优选1-1500mg,所述的疾病选自乳腺癌、子宫癌、宫颈癌、卵巢癌、前列腺癌。A method for treating a disease in a mammal, comprising administering to a subject a therapeutically effective amount of a compound or crystalline form of the present invention, and a pharmaceutically acceptable carrier and/or excipient, the therapeutically effective amount preferably being 1-1500 mg, wherein the disease is selected from breast cancer, uterine cancer, cervical cancer, ovarian cancer, and prostate cancer.
一种用于治疗哺乳动物的疾病的方法,所述方法包括,将药物本发明化合物或晶型物,以及药学上可接受的载体和/或赋形剂,以1-1440mg/天的日剂量给予受试者,所述日剂量可以为单剂量或分剂量,在一些实施方案中,日剂量包括但不限于10-1440mg/天、20-1440mg/天、25-1440mg/天、50-1440mg/天、75-1440mg/天、100-1440mg/天、200-1440mg/天、10-1000mg/天、20-1000mg/天、25-1000mg/天、50-1000mg/天、75-1000mg/天、100-1000mg/天、200-1000mg/天、25-800mg/天、50-800mg/天、100-800mg/天、200-800mg/天、25-400mg/天、50-400mg/天、100-400mg/天、200-400mg/天,在一些实施方案中,日剂量包括但不限于1mg/天、5mg/天、10mg/天、20mg/天、25mg/天、50mg/天、75mg/天、100mg/天、125mg/天、150mg/天、200mg/天、400mg/天、600mg/天、800mg/天、1000mg/天、1200mg/天、1400mg/天、1440mg/天。A method for treating a disease in a mammal, comprising administering a compound or crystalline form of the present invention and a pharmaceutically acceptable carrier and/or excipient to a subject at a daily dose of 1-1440 mg/day, wherein the daily dose can be a single dose or divided doses. In some embodiments, the daily dose includes but is not limited to 10-1440 mg/day, 20-1440 mg/day, 25-1440 mg/day, 50-1440 mg/day, 75-1440 mg/day, 100-1440 mg/day, 200-1440 mg/day, 10-1000 mg/day, 20-1000 mg/day, 25-1000 mg/day, 50-1000 mg/day, 75-1000 mg/day, 100-1000 mg/day, g/day, 200-1000 mg/day, 25-800 mg/day, 50-800 mg/day, 100-800 mg/day, 200-800 mg/day, 25-400 mg/day, 50-400 mg/day, 100-400 mg/day, 200-400 mg/day, in some embodiments, daily doses include but are not limited to 1 mg/day, 5 mg/day, 10 mg/day, 20 mg/day, 25 mg/day, 50 mg/day, 75 mg/day, 100 mg/day, 125 mg/day, 150 mg/day, 200 mg/day, 400 mg/day, 600 mg/day, 800 mg/day, 1000 mg/day, 1200 mg/day, 1400 mg/day, 1440 mg/day.
本发明涉及一种试剂盒,该试剂盒可以包括单剂量或多剂量形式的化合物或晶型物,该试剂盒包含本发明化合物或晶型物,本发明化合物或晶型物的量与上述药物组合物中其量相同。The present invention relates to a kit, which may include a compound or a crystal form in a single-dose or multi-dose form. The kit contains the compound or crystal form of the present invention, and the amount of the compound or crystal form of the present invention is the same as that in the above-mentioned pharmaceutical composition.
本发明中本发明晶型物的量在每种情况下以游离碱的形式换算。The amounts of the crystalline forms according to the invention in the present invention are in each case calculated as the free base.
“制剂规格”是指每一支、片或其他每一个单位制剂中含有主药的重量。"Preparation specifications" refers to the weight of the main drug contained in each vial, tablet or other unit preparation.
本发明所述的化合物或晶型物,以原料药的约5重量%至约100重量%存在;在某些实施方案中,以原料药的约10重量%至约100重量%存在;在某些实施方案中,以原料药的约15重量%至约100重量%存在;在某些实施方案中,以原料药的约20重量%至约100重量%存在;在某些实施方案中,以原料药的约25重量%至约100重量%存在;在某些实施方案中,以原料药的约30重量%至约100重量%存在;在某些实施方案中,以原料药的约35重量%至约100重量%存在;在某些实施方案中,以原料药的约40重量%至约100重量%存在;在某些实施方案中,以原料药的约45重量%至约100重量%存在;在某些实施方案中,以原料药的约50重量%至约100重量%存在;在某些实施方案中,以原料药的约55重量%至约100重量%存在;在某些实施方案中,以原料药的约60重量%至约100重量%存在;在某些实施方案中,以原料药的约65重量%至约100重量%存在;在某些实施方案中,以原料药的约70重量%至约100重量%存在;在某些实施方案中,以原料药的约75重量%至约100重量%存在;在某些实施方案中,以原料药的约80重量%至约100重量%存在;在某些实施方案中,以原料药的约85重量%至约100重量%存在;在某些实施方案中,以原料药的约90重量%至约100重量%存在;在某些实施方案中,以原料药的约95重量%至约100重量%存在;在某些实施方案中,以原料药的约98重量%至约100重量%存在;在某些实施方案中,以原料药的约99重量%至约100重量%存在;在某些实施方案中,基本上所有的原料药都是基本纯的化合物或晶体。The compound or crystalline form of the present invention is present in an amount of about 5% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 10% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 15% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 20% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 25% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 30% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 35% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 40% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 45% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 50% to about 100% by weight of the drug substance; in certain embodiments, it is present in an amount of about 55% to about 100% by weight of the drug substance. In certain embodiments, the drug substance is present at about 60% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 65% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 70% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 75% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 80% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 85% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 90% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 95% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 98% to about 100% by weight of the drug substance; in certain embodiments, the drug substance is present at about 99% to about 100% by weight of the drug substance; in certain embodiments, substantially all of the drug substance is a substantially pure compound or crystal.
本发明的晶型物可以经如下的制备方法制备:The crystalline form of the present invention can be prepared by the following preparation method:
1、挥发实验:将式(I)化合物加入所选单一溶剂或二元溶剂中形成样品澄清溶液,在不同温度下敞口挥发至溶剂干。1. Volatilization experiment: Add the compound of formula (I) into a selected single solvent or binary solvent to form a clear sample solution, and evaporate it open at different temperatures until the solvent is dry.
2、混悬法:将式(I)化合物加入所选单一溶剂或二元溶剂中,直至形成悬浮液,在室温至50℃下悬浮搅拌一定时间(例如1h~3天,或者2h~24h,或者2h~12h,或者3~5h)后,将悬浮液离心分离,干燥,即得。2. Suspension method: Add the compound of formula (I) to a selected single solvent or binary solvent until a suspension is formed. After suspension and stirring at room temperature to 50°C for a certain period of time (e.g., 1 hour to 3 days, or 2 hours to 24 hours, or 2 hours to 12 hours, or 3 hours to 5 hours), the suspension is centrifuged and dried to obtain the product.
3、溶析结晶法:将式(I)化合物溶解在良溶剂中,取一定量的溶液滴加至不良溶剂中或者将不良溶剂滴加至溶液中,搅拌析出固体,分离干燥,即得。3. Dissolution crystallization method: dissolve the compound of formula (I) in a good solvent, add a certain amount of the solution dropwise to a poor solvent or add a poor solvent dropwise to the solution, stir to precipitate a solid, separate and dry to obtain the product.
4、降温法:在高温下将一定量的样品溶解到相应溶剂中,将溶液转移至室温冷却,静置或搅拌析晶,分离,干燥,即得。4. Cooling method: dissolve a certain amount of sample in the corresponding solvent at high temperature, transfer the solution to room temperature for cooling, let it stand or stir for crystallization, separate, and dry to obtain the product.
5、热方法实验:取一定量的样品置于玻璃片放在热台上,以一定的速率(如5~20℃/min,或者10~15℃/min)加热至目标温度,并恒温一段时间(如0.5~5min,或者1~3min,或者1~2min),然后自然降温冷却至室温得固体。5. Thermal method experiment: Take a certain amount of sample, place it on a glass slide and place it on a hot table. Heat it to the target temperature at a certain rate (such as 5-20℃/min, or 10-15℃/min), keep it at a constant temperature for a period of time (such as 0.5-5min, or 1-3min, or 1-2min), and then cool it naturally to room temperature to obtain a solid.
6、气相扩散实验:将一定量式(I)化合物室温下滴加适量良溶剂使样品完全溶解或配置为良溶剂的饱和溶液;分别取一定量溶液,将澄清溶液置于不良溶剂气氛中室温静置,直至有固体析出,分离,即得。或者直接将式(I)化合物固体置于溶剂气氛中室温静置1~7天后即得。6. Vapor Diffusion Assay: Add a suitable amount of a good solvent dropwise to a certain amount of the compound of formula (I) at room temperature until the sample is completely dissolved or a saturated solution of the good solvent is prepared. A certain amount of each solution is taken and the clear solution is placed in a poor solvent atmosphere and allowed to stand at room temperature until solid precipitates. This is followed by separation. Alternatively, the solid compound of formula (I) can be directly placed in a solvent atmosphere and allowed to stand at room temperature for 1 to 7 days to obtain the product.
本发明所述的良溶剂和不良溶剂是相对而言的,在一对溶剂中,溶解度较高者为良溶剂,溶解度较低者为不良溶剂。在一些实施方式中,良溶剂选自乙二醇甲醚、乙二醇二甲醚、二氧六环、DMF、DMSO、甲醇、乙醇、正丙醇、甲酸丁酯、4-甲基-2-戊酮、四氢呋喃、异丙醇、乙酸乙酯、正庚烷、乙醚、水、乙腈、甲苯、氯仿、丙酮、甲酸丁酯、MTBE、环己烷中的溶解度较高者,不良溶剂选自上述溶剂中溶解度较低者。在一些实施方式中,良溶剂选自乙二醇甲醚、乙二醇二甲醚、二氧六环、DMF、DMSO、甲醇、乙醇、正丙醇、甲酸丁酯、4-甲基-2-戊酮、四氢呋喃或者它们的混合溶剂。在一些实施方式中,不良溶剂选自异丙醇、乙酸乙酯、正庚烷、乙醚、水、乙腈、甲苯、氯仿、丙酮、甲酸丁酯、MTBE、环己烷或者它们的混合溶剂。The good solvent and poor solvent of the present invention are relative. In a pair of solvents, the one with higher solubility is a good solvent, and the one with lower solubility is a poor solvent. In some embodiments, the good solvent is selected from ethylene glycol methyl ether, ethylene glycol dimethyl ether, dioxane, DMF, DMSO, methanol, ethanol, n-propyl alcohol, butyl formate, 4-methyl-2-pentanone, tetrahydrofuran, isopropyl alcohol, ethyl acetate, n-heptane, ether, water, acetonitrile, toluene, chloroform, acetone, butyl formate, MTBE, and cyclohexane with higher solubility, and the poor solvent is selected from the above-mentioned solvent with lower solubility. In some embodiments, the good solvent is selected from ethylene glycol methyl ether, ethylene glycol dimethyl ether, dioxane, DMF, DMSO, methanol, ethanol, n-propyl alcohol, butyl formate, 4-methyl-2-pentanone, tetrahydrofuran, or a mixed solvent thereof. In some embodiments, the poor solvent is selected from isopropyl alcohol, ethyl acetate, n-heptane, diethyl ether, water, acetonitrile, toluene, chloroform, acetone, butyl formate, MTBE, cyclohexane, or a mixed solvent thereof.
在一些实施方案中,挥发法的溶剂为水和丙酮;In some embodiments, the solvents of the evaporation method are water and acetone;
在一些实施方案中,溶析结晶法采用的溶剂为二氯甲烷和正庚烷;在一些实施方案中,溶析结晶法采用的溶剂为二氯甲烷和异丙醚;在一些实施方案中,溶析结晶法采用的溶剂为乙酸乙酯和正庚烷;在一些实施方案中,溶析结晶法采用的溶剂为4-甲基-2-戊酮和正庚烷;在一些实施方案中,溶析结晶法采用的溶剂为二氧六环和正庚烷;In some embodiments, the solvent used in the dissolution crystallization method is dichloromethane and n-heptane; in some embodiments, the solvent used in the dissolution crystallization method is dichloromethane and isopropyl ether; in some embodiments, the solvent used in the dissolution crystallization method is ethyl acetate and n-heptane; in some embodiments, the solvent used in the dissolution crystallization method is 4-methyl-2-pentanone and n-heptane; in some embodiments, the solvent used in the dissolution crystallization method is dioxane and n-heptane;
在一些实施方案中,降温法采用的溶剂为乙二醇二甲醚和环己烷;在一些实施方案中,降温法采用的溶剂为甲苯和环己烷;在一些实施方案中,降温法采用的溶剂为乙酸异丙酯和环己烷;在一些实施方案中,降温法采用的溶剂为二氧六环和环己烷;在一些实施方案中,降温法采用的溶剂为二氧六环和水;在一些实施方案中,降温法采用的溶剂为DMSO和水。In some embodiments, the solvents used in the cooling method are ethylene glycol dimethyl ether and cyclohexane; in some embodiments, the solvents used in the cooling method are toluene and cyclohexane; in some embodiments, the solvents used in the cooling method are isopropyl acetate and cyclohexane; in some embodiments, the solvents used in the cooling method are dioxane and cyclohexane; in some embodiments, the solvents used in the cooling method are dioxane and water; in some embodiments, the solvents used in the cooling method are DMSO and water.
在一些实施方案中,气相扩散法采用在乙酸乙酯中气相扩散。In some embodiments, the vapor diffusion method employs vapor diffusion in ethyl acetate.
本发明所述的良溶剂和不良溶剂是相对而言的,在一对溶剂中,溶解度较高者为良溶剂,溶解度较低者为不良溶剂。The good solvent and poor solvent described in the present invention are relative. In a pair of solvents, the one with higher solubility is a good solvent, and the one with lower solubility is a poor solvent.
上述制备方法所采用的溶剂,在未指明时,可采用单一溶剂,也可以采用两种或两种以上的组合。The solvent used in the above preparation method, unless otherwise specified, may be a single solvent or a combination of two or more solvents.
本发明公开的X-射线粉末衍射或DSC图、TGA图,与其实质上相同的也属于本发明的范围。The X-ray powder diffraction, DSC pattern, and TGA pattern disclosed in the present invention, and those substantially the same also fall within the scope of the present invention.
除非有相反的陈述,在说明书和权利要求书中使用的术语具有下述含义。Unless stated otherwise, the terms used in the specification and claims have the following meanings.
“IC50”指半数抑制浓度,指达到最大抑制效果一半时的浓度。" IC50 " refers to the half-maximal inhibitory concentration, which is the concentration at which half of the maximal inhibitory effect is achieved.
如本发明所用,“本发明的晶体”、“本发明的晶型”、“本发明的晶型物”等可互换使用。As used herein, "the crystal of the present invention", "the crystal form of the present invention", "the crystal form of the present invention" and the like can be used interchangeably.
本发明所述“室温”一般指4-30℃,优选地指20±5℃。The "room temperature" mentioned in the present invention generally refers to 4-30°C, preferably 20±5°C.
本发明晶型结构可以使用本领域普通技术人员已知的各种分析技术分析,包括但不限于,X-射线粉末衍射(XRD)、示差扫描热法(DSC)和/或热重分析(Thermogravimetric Analysis,TGA),又叫热重法(Thermogravimetry,TG)。The crystalline structure of the present invention can be analyzed using various analytical techniques known to those skilled in the art, including but not limited to, X-ray powder diffraction (XRD), differential scanning calorimetry (DSC) and/or thermogravimetric analysis (TGA), also known as thermogravimetry (TG).
本发明所述的“2θ或2θ角度”是指基于X射线衍射实验中设置的以度数(°)表示的峰位,并且通常是在衍射图谱中的横坐标单位。如果入射束与某晶格面形成θ角时反射被衍射,则实验设置需要以2θ角记录反射束。应当理解,在本文中提到的特定晶型的特定2θ值意图表示使用本文所述的X射线衍射实验条件所测量的2θ值(以度数表示),所述2θ的误差范围可以是±0.3、±0.2或±0.1。As used herein, "2θ or 2θ angle" refers to the peak position expressed in degrees (°) based on the setup of an X-ray diffraction experiment, and is typically the unit of the abscissa in a diffraction pattern. If the incident beam forms an angle θ with a certain lattice plane and the reflection is diffracted, the experimental setup requires recording the reflected beam in 2θ angles. It should be understood that the specific 2θ value of a specific crystal form mentioned herein is intended to represent the 2θ value (expressed in degrees) measured using the X-ray diffraction experimental conditions described herein, and the error range of the 2θ may be ±0.3, ±0.2, or ±0.1.
可以理解的是,本发明描述的和保护的数值为近似值。数值内的变化可能归因于设备的校准、设备误差、晶体的纯度、晶体大小、样本大小以及其他因素。It is understood that the numerical values described and protected by the present invention are approximate values. Variations in the numerical values may be due to equipment calibration, equipment errors, crystal purity, crystal size, sample size and other factors.
可以理解的是,本发明的晶型不限于与本发明公开的附图中描述的特征图谱完全相同的特征图谱,比如XRD、DSC、TGA、DVS,具有与附图中描述的哪些图谱基本上相同或本质上相同的特征图谱的任何晶型均落入本发明的范围内。It is understood that the crystal form of the present invention is not limited to the characteristic spectra that are exactly the same as the characteristic spectra described in the drawings disclosed in the present invention, such as XRD, DSC, TGA, and DVS. Any crystal form having characteristic spectra that are substantially the same or essentially the same as those described in the drawings falls within the scope of the present invention.
可以理解的是,差示扫描量热(DSC)领域中所熟知的,DSC曲线的熔融峰高取决于与样品制备和仪器几何形状有关的许多因素,而峰位置对实验细节相对不敏感。因此,在一些实施方案中,本发明的结晶化合物的特征在于具有特征峰位置的DSC图,具有与本发明附图中提供的DSC图实质上相同的性质,测量值误差容限为±5℃内,一般要求在±3℃。It is understood that, as is well known in the art of differential scanning calorimetry (DSC), the melting peak height of a DSC curve depends on many factors related to sample preparation and instrument geometry, while the peak position is relatively insensitive to experimental details. Therefore, in some embodiments, the crystalline compound of the present invention is characterized by a DSC pattern having a characteristic peak position, having substantially the same properties as the DSC pattern provided in the accompanying drawings of the present invention, with a measurement error tolerance of within ±5°C, generally required to be within ±3°C.
“载体”指的是:不会对生物体产生明显刺激且不会消除所给予化合物的生物活性和特性,并能改变药物进入人体的方式和在体内的分布、控制药物的释放速度并将药物输送到靶向器官的体系,非限制性的实例包括微囊与微球、纳米粒、脂质体等。"Carrier" refers to a system that does not cause significant irritation to the organism and does not eliminate the biological activity and properties of the administered compound, and can change the way the drug enters the human body and its distribution in the body, control the release rate of the drug and deliver the drug to the target organ. Non-limiting examples include microcapsules and microspheres, nanoparticles, liposomes, etc.
“赋形剂”指的是:其本身并非治疗剂,用作稀释剂、辅料、粘合剂和/或媒介物,用于添加至药物组合物中以改善其处置或储存性质或允许或促进化合物或药物组合物形成用于给药的单位剂型。如本领域技术人员所已知的,药用赋形剂可提供各种功能且可描述为润湿剂、缓冲剂、助悬剂、润滑剂、乳化剂、崩解剂、吸收剂、防腐剂、表面活性剂、着色剂、矫味剂及甜味剂。药用赋形剂的实例包括但不限于:(1)糖,例如乳糖、葡萄糖及蔗糖;(2)淀粉,例如玉米淀粉及马铃薯淀粉;(3)纤维素及其衍生物,例如羧甲基纤维素钠、乙基纤维素、乙酸纤维素、羟丙基甲基纤维素、羟丙基纤维素、微晶纤维素及交联羧甲基纤维素(例如交联羧甲基纤维素钠);(4)黄蓍胶粉;(5)麦芽;(6)明胶;(7)滑石;(8)赋形剂,例如可可脂及栓剂蜡;(9)油,例如花生油、棉籽油、红花油、芝麻油、橄榄油、玉米油及大豆油;(10)二醇,例如丙二醇;(11)多元醇,例如甘油、山梨醇、甘露醇及聚乙二醇;(12)酯,例如油酸乙酯及月桂酸乙酯;(13)琼脂;(14)缓冲剂,例如氢氧化镁及氢氧化铝;(15)海藻酸;(16)无热原水;(17)等渗盐水;(18)林格溶液(Ringer’s solution);(19)乙醇;(20)pH缓冲溶液;(21)聚酯、聚碳酸酯和/或聚酐;及(22)其他用于药物制剂中的无毒相容物质。An "excipient" is a substance that is not itself a therapeutic agent but serves as a diluent, adjuvant, binder, and/or vehicle that is added to a pharmaceutical composition to improve its handling or storage properties or to allow or facilitate the formation of a compound or pharmaceutical composition into a unit dosage form for administration. As known to those skilled in the art, pharmaceutical excipients can serve a variety of functions and can be described as wetting agents, buffers, suspending agents, lubricants, emulsifiers, disintegrants, absorbents, preservatives, surfactants, colorants, flavoring agents, and sweeteners. Examples of pharmaceutical excipients include, but are not limited to: (1) sugars such as lactose, glucose, and sucrose; (2) starches such as corn starch and potato starch; (3) cellulose and its derivatives such as sodium carboxymethylcellulose, ethylcellulose, cellulose acetate, hydroxypropyl methylcellulose, hydroxypropyl cellulose, microcrystalline cellulose, and cross-linked carboxymethylcellulose (e.g., cross-linked sodium carboxymethylcellulose); (4) tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients such as cocoa butter and suppository waxes; (9) oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn starch, and maltodextrin. oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerol, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffers, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer’s solution; (19) ethanol; (20) pH buffer solutions; (21) polyesters, polycarbonates and/or polyanhydrides; and (22) other non-toxic compatible substances used in pharmaceutical preparations.
图1为式(I)所示化合物无定形的X-射线粉末衍射图谱。FIG1 is an X-ray powder diffraction pattern of the amorphous compound represented by formula (I).
图2为式(I)所示化合物晶型I的X-射线粉末衍射图谱。Figure 2 is an X-ray powder diffraction pattern of Form I of the compound represented by formula (I).
图3为式(I)所示化合物晶型I的差示扫描量热分析曲线图谱。FIG3 is a differential scanning calorimetry analysis curve of Form I of the compound represented by formula (I).
图4为式(I)所示化合物晶型I的热重分析图谱。FIG4 is a thermogravimetric analysis spectrum of Form I of the compound represented by formula (I).
图5为式(I)所示化合物晶型I的等温吸附曲线。FIG5 is an isothermal adsorption curve of Form I of the compound represented by formula (I).
图6为式(I)所示化合物晶型II的X-射线粉末衍射图谱。FIG6 is an X-ray powder diffraction pattern of Form II of the compound represented by formula (I).
图7为式(I)所示化合物晶型II的差示扫描量热分析曲线图谱。FIG7 is a differential scanning calorimetry analysis curve of Form II of the compound represented by formula (I).
图8为式(I)所示化合物晶型II的热重分析图谱。FIG8 is a thermogravimetric analysis spectrum of Form II of the compound represented by formula (I).
图9为式(I)所示化合物晶型II的等温吸附曲线。FIG9 is an isothermal adsorption curve of Form II of the compound represented by formula (I).
化合物的结构是通过核磁共振(NMR)或(和)质谱(MS)来确定的。NMR位移(δ)以10-6(ppm)的单位给出。NMR的测定是用(Bruker Avance III 400和Bruker Avance 300)核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代氯仿(CDCl3),氘代甲醇(CD3OD),内标为四甲基硅烷(TMS)。The structures of the compounds were confirmed by nuclear magnetic resonance (NMR) and/or mass spectrometry (MS). NMR shifts (δ) are given in units of 10-6 (ppm). NMR measurements were performed using a Bruker Avance III 400 and Bruker Avance 300 NMR spectrometer. The solvents used were deuterated dimethyl sulfoxide (DMSO- d6 ), deuterated chloroform ( CDCl3 ), and deuterated methanol ( CD3OD ), with tetramethylsilane (TMS) as the internal standard.
MS的测定用(Agilent 6120B(ESI)和Agilent 6120B(APCI))。MS was performed using (Agilent 6120B (ESI) and Agilent 6120B (APCI)).
HPLC的测定使用LC-20AT(岛津)高压液相色谱仪(Shim-pack GIST C18,4.6×250mm(HSS),5μm)。HPLC determination was performed using a LC-20AT (Shimadzu) high pressure liquid chromatograph (Shim-pack GIST C18, 4.6×250 mm (HSS), 5 μm).
晶型检测仪器信息和检测方法参数表(见下表1):Crystal form detection instrument information and detection method parameter table (see Table 1 below):
表1.仪器信息和检测方法参数表
Table 1. Instrument information and detection method parameters
本发明的己知的起始原料可以采用或按照本领域已知的方法来合成,或可购买于泰坦科技、安耐吉化学、上海德默、成都科龙化工、韶远化学科技、百灵威科技等公司。The known starting materials of the present invention can be synthesized by methods known in the art, or can be purchased from companies such as Titan Technology, Anage Chemical, Shanghai Demer, Chengdu Kelon Chemical, Shaoyuan Chemical Technology, and Bailingwei Technology.
实施例中无特殊说明,溶液是指水溶液。Unless otherwise specified in the examples, the solution refers to an aqueous solution.
实施例中无特殊说明,室温为20℃~30℃。Unless otherwise specified in the examples, the room temperature is 20°C to 30°C.
以下通过具体实施例详细说明本发明的实施过程和产生的有益效果,旨在帮助阅读者更好地理解本发明的实质和特点,不作为对本案可实施范围的限定。The following describes in detail the implementation process of the present invention and the beneficial effects produced by specific embodiments, which is intended to help readers better understand the essence and characteristics of the present invention and is not intended to limit the scope of implementation of this case.
实施例1:式(I)化合物的制备
Example 1: Preparation of compound of formula (I)
式(I)化合物合成:Synthesis of compound of formula (I):
第一步:3-溴-1,1-二氟丙-2-酮(1A)
Step 1: 3-Bromo-1,1-difluoropropan-2-one (1A)
将3-溴-1,1-二氟丙烷-2-酮(5g,40.27mmol)溶于无水的四氢呋喃中,置换氮气三次,加入二溴甲烷(14g,80.54mmol),将反应体系冷却至-78℃,滴加甲基锂(80.54mmol),在-78℃下反应2h。反应结束在0℃下加氯化铵饱和溶液淬灭反应,乙酸乙酯萃取(50ml×3),合并有机相,无水硫酸钠干燥,过滤浓缩后残留物柱层析快速分离纯化(洗脱剂比例:EA/PE=0%~20%)得化合物1A(5.01g,72%)。3-Bromo-1,1-difluoropropane-2-one (5 g, 40.27 mmol) was dissolved in anhydrous tetrahydrofuran, the nitrogen atmosphere was replaced three times, and dibromomethane (14 g, 80.54 mmol) was added. The reaction system was cooled to -78°C, and methyllithium (80.54 mmol) was added dropwise. The reaction was allowed to react at -78°C for 2 h. After completion of the reaction, saturated ammonium chloride solution was added at 0°C to quench the reaction. The mixture was extracted with ethyl acetate (50 ml x 3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was then purified by rapid column chromatography (eluent ratio: EA/PE = 0% to 20%) to obtain compound 1A (5.01 g, 72%).
LC-MS(ESI):m/z=173.2[M+1]+ LC-MS (ESI): m/z = 173.2 [M + 1 ] +
第二步:6-溴-2-(二氟甲基)咪唑并[1,2-a]吡嗪(1B)
Step 2: 6-Bromo-2-(difluoromethyl)imidazo[1,2-a]pyrazine (1B)
将化合物1A(49.42g,287.35mmol)、2-氨基-5-溴吡嗪(10g,57.47mmol)溶于1,4-二氧六环(100mL)中,于100℃下搅拌1h。LCMS监控反应,过滤收集滤饼,滤饼分散在300ml的1,4-二氧六环中,100℃下搅拌过夜,反应完毕后直接旋干,快速分离纯化(PE/EA=0-20%)得到化合物1B(10g,80%)。Compound 1A (49.42 g, 287.35 mmol) and 2-amino-5-bromopyrazine (10 g, 57.47 mmol) were dissolved in 1,4-dioxane (100 mL) and stirred at 100°C for 1 h. The reaction was monitored by LCMS. The filter cake was collected by filtration and dispersed in 300 mL of 1,4-dioxane. The mixture was stirred at 100°C overnight. After completion of the reaction, the mixture was directly spin-dried and purified by rapid separation (PE/EA = 0-20%) to obtain compound 1B (10 g, 80%).
MS m/z=248.0[M+1]+ MS m/z=248.0[M+1] +
第三步:4-(2-(二氟甲基)咪唑[1,2-a]吡嗪-6-基)哌嗪-1-羧酸叔丁酯(1C)
Step 3: tert-Butyl 4-(2-(difluoromethyl)imidazo[1,2-a]pyrazin-6-yl)piperazine-1-carboxylate (1C)
将化合物1B(5g,20.2mmol)、哌嗪-1-甲酸叔丁酯(5.6g,30mmol)、三(二亚苄基丙酮)二钯(1.92g,2mmol)、2-(二叔丁基膦)联苯(0.6g,2mmol)、叔丁醇钠(5.8g,60.6mmol)、甲苯(50mL)加至反应瓶内,在氮气保护下于110℃搅拌2h。反应结束后经乙酸乙酯萃取反应液三次、饱和氯化钠溶液洗涤、无水硫酸钠干燥、减压浓缩后快速柱层析(EA:PE=20%)得到目标化合物1C(3g,42%)。Compound 1B (5 g, 20.2 mmol), tert-butyl piperazine-1-carboxylate (5.6 g, 30 mmol), tris(dibenzylideneacetone)dipalladium (1.92 g, 2 mmol), 2-(di-tert-butylphosphino)biphenyl (0.6 g, 2 mmol), sodium tert-butoxide (5.8 g, 60.6 mmol), and toluene (50 mL) were added to a reaction flask and stirred at 110°C under nitrogen for 2 h. After completion of the reaction, the reaction solution was extracted three times with ethyl acetate, washed with saturated sodium chloride solution, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified by flash column chromatography (EA:PE = 20%) to obtain the target compound 1C (3 g, 42%).
LC-MS(ESI):m/z=354.2[M+1]+ LC-MS (ESI): m/z = 354.2 [M + 1] +
第三步:6-(哌嗪-1-基)-2-(二氟甲基)咪唑并[1,2-a]吡嗪(1D)
Step 3: 6-(piperazin-1-yl)-2-(difluoromethyl)imidazo[1,2-a]pyrazine (1D)
将化合物1C(330mg,0.89mmol)、三氟乙酸(3mL)加至反应瓶内,于25℃下搅拌1h。反应结束后减压浓缩得到粗品化合物1D(240mg,100%),直接投下一步。Compound 1C (330 mg, 0.89 mmol) and trifluoroacetic acid (3 mL) were added to the reaction flask and stirred at 25°C for 1 h. After the reaction, the mixture was concentrated under reduced pressure to obtain crude compound 1D (240 mg, 100%), which was directly used in the next step.
第四步:7-((4-(2-(二氟甲基)咪唑并[1,2-a]吡嗪-6-基)哌嗪-1-基)甲基)-8-氟-3-甲基喹喔啉-2(1H)-酮(式I)Step 4: 7-((4-(2-(difluoromethyl)imidazo[1,2-a]pyrazin-6-yl)piperazin-1-yl)methyl)-8-fluoro-3-methylquinoxalin-2(1H)-one (Formula I)
7-((4-(2-(difluoromethyl)imidazo[1,2-a]pyrazin-6-yl)piperazin-1-yl)methyl)-8-fluoro-3-methylquinoxalin-2(1H)-one
7-((4-(2-(difluoromethyl)imidazo[1,2-a]pyrazin-6-yl)piperazin-1-yl)methyl)-8-fluoro-3-methylquinoxalin-2(1H)-one
将1E(按照专利US20220009901A1合成)(1.28g,4.74mmol)、1D(1.0g,3.95mmol)、N,N-二异丙基乙胺(1.54g,11.86mmol)、碘化钾(61.42mg,0.37mmol)、乙腈(3mL)加至反应瓶内,于60℃下搅拌1h。LCMS监控反应,反应完毕后直接将反应体系送制备,制备HPLC分离方法:1.仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×250mm)2.样品用0.45μm滤头过滤,制成样品液。3.制备色谱条件:a.流动相A,B组成:流动相A:乙腈;流动相B:水(含0.1%乙酸铵)b.梯度洗脱,流动相A含量从10%-55%c.流量12mL/min。保留时间7.0min得到化合物式(I)(1.0g,57%)。1E (synthesized according to US20220009901A1) (1.28 g, 4.74 mmol), 1D (1.0 g, 3.95 mmol), N,N-diisopropylethylamine (1.54 g, 11.86 mmol), potassium iodide (61.42 mg, 0.37 mmol), and acetonitrile (3 mL) were added to a reaction flask and stirred at 60°C for 1 h. The reaction was monitored by LCMS. After completion, the reaction system was directly sent to preparative HPLC for separation: 1. Instrument: Waters 2767 Preparative HPLC; Chromatographic column: SunFire@Prep C18 (19 mm × 250 mm). 2. The sample was filtered through a 0.45 μm filter to prepare a sample solution. 3. Preparative Chromatography Conditions: a. Mobile Phase A, B Composition: Mobile Phase A: Acetonitrile; Mobile Phase B: Water (containing 0.1% ammonium acetate); b. Gradient elution, Mobile Phase A content from 10% to 55%; c. Flow rate 12 mL/min. Retention time 7.0 min to yield Compound (I) (1.0 g, 57%).
LCMS m/z=444.4[M+1]+ LCMS m/z=444.4[M+1] +
1H NMR(400MHz,DMSO-d6)δ8.90(s,1H),8.21(s,1H),7.93(d,1H),7.52(d,1H),7.30(d,1H),7.16(t,1H),3.70(s,2H),3.31(d,5H),2.63–2.55(m,4H),2.42(s,3H). 1 H NMR(400MHz,DMSO-d6)δ8.90(s,1H),8.21(s,1H),7.93(d,1H),7.52(d,1H),7.3 0(d,1H),7.16(t,1H),3.70(s,2H),3.31(d,5H),2.63–2.55(m,4H),2.42(s,3H).
实施例2式(I)所示化合物无定形的制备Example 2 Preparation of the amorphous compound represented by formula (I)
取实施例1所得到的式(I)化合物50mg,加入20mL乙腈与水的混合溶剂,溶清后冻干,通过XRD表征式(I)所示化合物的无定形,其X-射线粉末衍射图谱如图1。50 mg of the compound of formula (I) obtained in Example 1 was added to 20 mL of a mixed solvent of acetonitrile and water, dissolved and then freeze-dried. The amorphous form of the compound of formula (I) was characterized by XRD. Its X-ray powder diffraction pattern is shown in Figure 1.
实施例3式(I)所示化合物晶型I的制备Example 3 Preparation of Crystalline Form I of the Compound Represented by Formula (I)
取实施例2制得的式(I)所示化合物的无定形150mg,加入二氯甲烷(3mL)和甲醇(1ml),升温至50℃溶解,冷却至室温后缓慢加入甲基叔丁基醚(2.0mL),加完室温搅拌过夜,析出固体过滤,滤饼用少量甲基叔丁基醚洗涤后在50℃下真空干燥过夜,得到式(I)所示化合物晶型I,通过XRD、DSC、TGA和DVS表征式(I)所示化合物的晶型I,其X-射线粉末衍射图谱、差示扫描量热分析曲线、热重分析图、等温吸附曲线依次为图2-5。150 mg of the amorphous form of the compound represented by formula (I) obtained in Example 2 was taken, dichloromethane (3 mL) and methanol (1 ml) were added, the temperature was raised to 50°C to dissolve, and methyl tert-butyl ether (2.0 mL) was slowly added after cooling to room temperature. After the addition, the mixture was stirred at room temperature overnight. The precipitated solid was filtered, and the filter cake was washed with a small amount of methyl tert-butyl ether and then vacuum-dried at 50°C overnight to obtain Form I of the compound represented by formula (I). Form I of the compound represented by formula (I) was characterized by XRD, DSC, TGA and DVS, and its X-ray powder diffraction pattern, differential scanning calorimetry analysis curve, thermogravimetric analysis diagram, and isothermal adsorption curve are shown in Figures 2-5, respectively.
1H NMR(400MHz,DMSO-d6)δ8.90(s,1H),8.21(s,1H),7.93(d,1H),7.52(d,1H),7.30(d,1H),7.16(t,1H),3.70(s,2H),3.31(d,5H),2.63–2.55(m,4H),2.42(s,3H). 1 H NMR(400MHz,DMSO-d6)δ8.90(s,1H),8.21(s,1H),7.93(d,1H),7.52(d,1H),7.3 0(d,1H),7.16(t,1H),3.70(s,2H),3.31(d,5H),2.63–2.55(m,4H),2.42(s,3H).
式(I)化合物的晶型I的X-射线粉末衍射图谱(XRD)如图2所示。具体峰值如表2所示。The X-ray powder diffraction pattern (XRD) of the crystalline form I of the compound of formula (I) is shown in Figure 2. The specific peaks are shown in Table 2.
表2.晶型I的XRD具体峰值
Table 2. XRD specific peaks of Form I
实施例4式(I)所示化合物晶型II的制备Example 4 Preparation of Crystalline Form II of the Compound Represented by Formula (I)
取实施例2所得到的式(I)无定形化合物150mg,加入4mL乙腈与水的混合溶剂,在50℃下溶解,热过滤,所得滤液在室温下冷却结晶,室温搅拌过夜。过滤,所得固体在50℃下真空干燥过夜,得到式(I)所示化合物晶型II,通过XRD、DSC、TGA和DVS表征式(I)所示化合物的晶型II,其X-射线粉末衍射图谱、差示扫描量热分析曲线、热重分析图、等温吸附曲线依次为图6-9。150 mg of the amorphous compound of formula (I) obtained in Example 2 was added to 4 mL of a mixed solvent of acetonitrile and water, dissolved at 50°C, and hot filtered. The resulting filtrate was cooled at room temperature for crystallization and stirred overnight at room temperature. Filtered, the resulting solid was vacuum-dried at 50°C overnight to obtain Form II of the compound of formula (I). Form II of the compound of formula (I) was characterized by XRD, DSC, TGA, and DVS. Its X-ray powder diffraction pattern, differential scanning calorimetry curve, thermogravimetric analysis diagram, and isothermal adsorption curve are shown in Figures 6-9, respectively.
1H NMR(400MHz,DMSO-d6)δ8.90(s,1H),8.21(s,1H),7.93(d,1H),7.52(d,1H),7.30(d,1H),7.16(t,1H),3.70(s,2H),3.31(d,5H),2.63–2.55(m,4H),2.42(s,3H). 1 H NMR(400MHz,DMSO-d6)δ8.90(s,1H),8.21(s,1H),7.93(d,1H),7.52(d,1H),7.3 0(d,1H),7.16(t,1H),3.70(s,2H),3.31(d,5H),2.63–2.55(m,4H),2.42(s,3H).
式(I)化合物的晶型II的X-射线粉末衍射图谱(XRD)如图6所示。具体峰值如表3所示。The X-ray powder diffraction pattern (XRD) of the crystalline form II of the compound of formula (I) is shown in Figure 6. The specific peaks are shown in Table 3.
表3.晶型II的XRD具体峰值
Table 3. XRD specific peaks of Form II
实施例5相关晶型稳定性研究Example 5 Study on the stability of related crystal forms
1、式(I)化合物晶型I晶型稳定性1. Crystal stability of the compound of formula (I) crystal form I
测定条件见表4,实验结果见表5。The measurement conditions are shown in Table 4, and the experimental results are shown in Table 5.
表4.晶型I的固态稳定性实验
Table 4. Solid-state stability test of Form I
表5.晶型I固态稳定性实验的检测结果
Table 5. Test results of solid-state stability test of Form I
结论:式(I)化合物晶型I固态稳定性较好。Conclusion: The crystalline form I of the compound of formula (I) has good solid-state stability.
2、式(I)化合物晶型II晶型稳定性2. Crystal stability of Form II of the compound of formula (I)
实验条件见表6,实验结果见表7。The experimental conditions are shown in Table 6, and the experimental results are shown in Table 7.
表6.晶型II的固态稳定性实验
Table 6. Solid-state stability test of Form II
表7.晶型II固态稳定性实验的检测结果
Table 7. Test results of solid-state stability test of Form II
结论:式(I)化合物晶型II的固态稳定性良好。Conclusion: The solid-state stability of the crystalline form II of the compound of formula (I) is good.
实施例6式(I)化合物的晶型I和晶型II转化研究Example 6 Study on the transformation of the compound of formula (I) into Form I and Form II
分别取10mg晶型I和10mg晶型II样品,混合均匀,取样进行XRPD表征;将混样加入0.5mL二甲亚砜和水(1:1)混合溶剂中形成混悬液,在室温下搅拌5天,离心,40℃真空干燥过夜,取样进行XRPD表征。10 mg of Form I and 10 mg of Form II samples were taken respectively, mixed evenly, and sampled for XRPD characterization; the mixed sample was added to 0.5 mL of a mixed solvent of dimethyl sulfoxide and water (1:1) to form a suspension, stirred at room temperature for 5 days, centrifuged, and vacuum-dried at 40°C overnight, and sampled for XRPD characterization.
表8.晶型I和晶型II晶型转化实验结果表
Table 8. Crystal form conversion test results of Form I and Form II
结论:在室温条件下,晶型I热力学稳定性优于晶型II。Conclusion: At room temperature, the thermodynamic stability of form I is better than that of form II.
实施例7式(I)化合物晶型的DSC测试Example 7 DSC test of the crystal form of the compound of formula (I)
差示扫描量热分析仪的型号为TA Instruments Q200DSC。0.5-5mg样品经精确称重后置于扎孔的DSC Tzero样品盘中,以10℃/min的速率加热至最终温度,炉内氮气吹扫速度为50mL/min。A TA Instruments Q200DSC differential scanning calorimeter was used. A 0.5–5 mg sample was accurately weighed and placed in a perforated DSC Tzero pan. The sample was heated to the final temperature at a rate of 10°C/min, with a nitrogen purge rate of 50 mL/min.
式(I)化合物晶型1和晶型2测试结果见图3和图7所示。The test results of Form 1 and Form 2 of the compound of formula (I) are shown in Figures 3 and 7.
实施例8式(I)化合物晶型的TGA测试Example 8 TGA test of the crystal form of the compound of formula (I)
热重分析仪的型号为TA Instruments Q500TGA。将1-10mg样品置于已平衡的开口铝制样品盘中,在TGA加热炉内自动称量。样品以10℃/min的速率加热至最终温度,样品处氮气吹扫速度为60mL/min,天平处氮气吹扫速度为40mL/min。The thermogravimetric analyzer (TGA) was a TA Instruments Q500TGA. Samples (1–10 mg) were placed in a pre-equilibrated open aluminum sample pan and automatically weighed within the TGA furnace. The samples were heated to the final temperature at a rate of 10°C/min, with nitrogen purge rates of 60 mL/min at the sample and 40 mL/min at the balance.
式(I)化合物的晶型1和晶型2测试结果见图4和图8所示。The test results of Form 1 and Form 2 of the compound of formula (I) are shown in Figures 4 and 8.
实施例9 PARP酶活性测试实验Example 9 PARP enzyme activity test experiment
用FP方法测试化合物对PARP1/2的酶活性抑制实验。在反应缓冲液(50mM Tris(pH 8.0)、10mM MgCl2、150mM NaCl、0.001%Triox-100)中制备反应中的底物示踪剂Tracer(来自ICE)。将PARP1/2(BPS,Cat#80501/80502)加到反应Buffer中并轻轻混合。反应混合物中PARP1/2和Tracer的最终浓度分别为5/10nM和2.5nM。阳性参照品Olaparib起始为1μM,3倍稀释,10个浓度。通过声学液体输送技术(Echo655)将100% DMSO中的化合物0.1μL输送到384孔板(Corning4514)中,1000rpm离心1分钟;转移5μL PARP酶溶液到384反应板中并1000rpm离心1分钟,于25℃孵育10min;转移5μL底物溶液到384反应板中,1000rpm离心1分钟,于25℃孵育60min;最后用BMG PHERAstar FSX的荧光偏振模块读取FP(ex/em:485nm/520nm)的mP信号值。之后使用GraphPad Prism8的软件进行非线性回归曲线四参数拟合IC50值。Compounds were tested for inhibition of PARP1/2 enzyme activity using the FP method. The substrate tracer, Tracer (from ICE), was prepared in reaction buffer (50 mM Tris (pH 8.0), 10 mM MgCl2, 150 mM NaCl, 0.001% Triox-100). PARP1/2 (BPS, Cat#80501/80502) was added to the reaction buffer and gently mixed. The final concentrations of PARP1/2 and Tracer in the reaction mixture were 5/10 nM and 2.5 nM, respectively. The positive reference, Olaparib, started at 1 μM and was diluted 3-fold to 10 concentrations. 0.1 μL of compound in 100% DMSO was delivered to a 384-well plate (Corning 4514) using acoustic liquid delivery technology (Echo 655) and centrifuged at 1000 rpm for 1 minute. 5 μL of PARP enzyme solution was transferred to a 384-well plate and centrifuged at 1000 rpm for 1 minute, followed by incubation at 25°C for 10 minutes. 5 μL of substrate solution was transferred to a 384-well plate, centrifuged at 1000 rpm for 1 minute, and incubated at 25°C for 60 minutes. Finally, the mP signal of FP (ex/em: 485 nm/520 nm) was read using the fluorescence polarization module of the BMG PHERAstar FSX. GraphPad Prism 8 software was then used to perform a four-parameter nonlinear regression fit to the IC50 value.
实验结果:式(I)化合物在体外对PARP-1和PARP-2的IC50值用A、B、C、D等级表示,A表示0<IC50≤10nM,B表示10nM<IC50≤50nM,C表示50nM<IC50≤500nM,D表示500nM<IC50。Experimental results: The IC50 values of the compound of formula (I) against PARP-1 and PARP-2 in vitro are expressed on a scale of A, B, C, or D, where A represents 0 < IC50 ≤ 10 nM , B represents 10 nM < IC50 ≤ 50 nM , C represents 50 nM < IC50 ≤ 500 nM, and D represents 500 nM < IC50 .
表9.PARP酶活
Table 9. PARP enzyme activity
结论:本发明式(I)化合物在体外对PARP-1酶活性具有显著选择性抑制作用。Conclusion: The compound of formula (I) of the present invention has a significant selective inhibitory effect on PARP-1 enzyme activity in vitro.
实施例10小鼠药代动力学测试Example 10 Pharmacokinetic test in mice
试验动物:雄性Balb/c小鼠,20~25g,12只/化合物。购于成都达硕实验动物有限公司。Experimental animals: Male Balb/c mice, 20-25 g, 12 mice per compound, purchased from Chengdu Dashuo Experimental Animal Co., Ltd.
试验设计:试验当天,将Balb/c小鼠按体重随机分组。给药前1天禁食不禁水12~14h,给药后4h给食。Experimental Design: On the day of the experiment, Balb/c mice were randomly divided into groups according to body weight. They were fasted but not watered for 12-14 hours before administration and fed 4 hours after administration.
表10.给药信息
Table 10. Dosing Information
注:静脉给药溶媒:10%DMA+10%Solutol+80%Saline;灌胃给药溶媒:5%DMSO+30%PEG400+65%(20%SBE-CD),Solutol为聚乙二醇-15-羟基硬脂酸酯;Saline为生理盐水Note: Intravenous administration solvent: 10% DMA + 10% Solutol + 80% Saline; oral administration solvent: 5% DMSO + 30% PEG400 + 65% (20% SBE-CD), Solutol is polyethylene glycol-15-hydroxystearate; Saline is normal saline
于给药前及给药后异氟烷麻醉经眼眶取血0.06mL,置于EDTAK2离心管中,5000rpm,4℃离心10min,收集血浆。静脉组和灌胃组采血时间点均为:0,5,15,30min,1,2,4,6,8,24h。化合物12,15,18给药后分别于30min,2,24h取脑组织。化合物2,5于24h取脑组织,脑组织用冷生理盐水冲净表面残留血液,吸干后匀浆处理。分析检测前,所有样品存于-80℃,用LC-MS/MS对样品进行定量分析。Before and after administration, 0.06 mL of blood was collected from the orbital cavity under isoflurane anesthesia. The blood was placed in an EDTAK2 centrifuge tube and centrifuged at 5000 rpm at 4°C for 10 minutes to collect plasma. Blood was collected from both the intravenous and oral gavage groups at 0, 5, 15, 30 minutes, and 1, 2, 4, 6, 8, and 24 hours. Brain tissue was collected at 30 minutes, 2, and 24 hours after administration of compounds 12, 15, and 18, respectively. Brain tissue was collected at 24 hours after administration of compounds 2 and 5. The brain tissue was rinsed with cold saline to remove any residual blood, blotted dry, and homogenized. All samples were stored at -80°C prior to analysis and quantitative analysis was performed using LC-MS/MS.
表11.测试化合物在小鼠血浆中的药代动力学参数
Table 11. Pharmacokinetic parameters of test compounds in mouse plasma
结论:式(I)化合物在小鼠血浆中具有优异的药代动力学特征。Conclusion: The compound of formula (I) has excellent pharmacokinetic characteristics in mouse plasma.
实施例11晶型的稳定性研究Example 11 Stability Study of Crystal Form
取晶型样品分别在40℃+75%RH、30℃+60%RH条件下,各自独立地放置0月、1月、2月、3月、6月、9月,HPLC检测纯度,结果表明,本发明式(I)化合物晶型I、II的化学稳定性均良好。Crystal samples were taken and placed independently at 40°C + 75% RH and 30°C + 60% RH for 0 month, 1 month, 2 months, 3 months, 6 months and 9 months, respectively. The purity was tested by HPLC. The results showed that the chemical stability of crystal forms I and II of the compound of formula (I) of the present invention was good.
实施例12影响因素实验研究Example 12 Experimental Study of Influencing Factors
(1)取晶型样品分别在40℃、60℃、92.5%RH条件下,各自独立地放置0天、6天,HPLC检测纯度;(1) Crystal samples were placed at 40°C, 60°C, and 92.5% RH for 0 and 6 days respectively, and the purity was determined by HPLC.
(2)光照(4500Lx)下,将晶型样品分别在40℃、60℃、92.5%RH条件下,各种独立地放置6天、10天、30天,检测纯度。(2) Under light (4500 Lx), the crystal samples were placed independently at 40°C, 60°C, and 92.5% RH for 6 days, 10 days, and 30 days, and the purity was tested.
研究结果表明,本发明式(I)化合物晶型I、II在上述影响因素实验条件下均能保持良好的化学稳定性。The research results show that the crystal forms I and II of the compound of formula (I) of the present invention can maintain good chemical stability under the experimental conditions of the above-mentioned influencing factors.
Claims (8)
A crystalline form of a compound of formula (I):
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN202410135587.0A CN120398888A (en) | 2024-01-31 | 2024-01-31 | A crystal form of a bicyclic derivative PARP inhibitor, its preparation method and application |
| CN202410135587.0 | 2024-01-31 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025162419A1 true WO2025162419A1 (en) | 2025-08-07 |
Family
ID=96516227
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2025/075377 Pending WO2025162419A1 (en) | 2024-01-31 | 2025-01-27 | Crystal form of bicyclic derivative parp inhibitor, preparation method therefor, and use thereof |
Country Status (2)
| Country | Link |
|---|---|
| CN (1) | CN120398888A (en) |
| WO (1) | WO2025162419A1 (en) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2023046034A1 (en) * | 2021-09-22 | 2023-03-30 | 明慧医药(杭州)有限公司 | Nitrogen-containing heterocyclic compound, preparation method therefor, intermediate thereof, and application thereof |
| CN115919859A (en) * | 2022-07-14 | 2023-04-07 | 四川海思科制药有限公司 | Pharmaceutical composition of heteroaryl derivative and application thereof in medicine |
| WO2023227052A1 (en) * | 2022-05-25 | 2023-11-30 | 西藏海思科制药有限公司 | Bicyclic derivative parp inhibitor and use thereof |
-
2024
- 2024-01-31 CN CN202410135587.0A patent/CN120398888A/en active Pending
-
2025
- 2025-01-27 WO PCT/CN2025/075377 patent/WO2025162419A1/en active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2023046034A1 (en) * | 2021-09-22 | 2023-03-30 | 明慧医药(杭州)有限公司 | Nitrogen-containing heterocyclic compound, preparation method therefor, intermediate thereof, and application thereof |
| WO2023227052A1 (en) * | 2022-05-25 | 2023-11-30 | 西藏海思科制药有限公司 | Bicyclic derivative parp inhibitor and use thereof |
| CN115919859A (en) * | 2022-07-14 | 2023-04-07 | 四川海思科制药有限公司 | Pharmaceutical composition of heteroaryl derivative and application thereof in medicine |
Also Published As
| Publication number | Publication date |
|---|---|
| CN120398888A (en) | 2025-08-01 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| CN103476770B (en) | Salts and polymorphic forms of afatinib | |
| CN107286077A (en) | A kind of selective C-KIT kinase inhibitors | |
| WO2023227052A1 (en) | Bicyclic derivative parp inhibitor and use thereof | |
| TW202327593A (en) | Polymorph of kras inhibitor and preparation method for polymorph and use thereof | |
| US20240174669A1 (en) | Crystalline forms of a parp1 inhibitor | |
| WO2016155670A1 (en) | Cdk inhibitor, eutectic crystal of mek inhibitor, and preparation method therefor | |
| TWI875727B (en) | Crystalline and salt forms of an organic compound and pharmaceutical compositions thereof | |
| CN118005610A (en) | PARP-1 degrading agent and application thereof | |
| WO2025016457A1 (en) | New substituted heterocyclic compound as gspts/myc signal flux regulator | |
| WO2023185931A1 (en) | P2x3 inhibitor compound, salt thereof, polymorph thereof and use thereof | |
| CN115919859A (en) | Pharmaceutical composition of heteroaryl derivative and application thereof in medicine | |
| WO2025162419A1 (en) | Crystal form of bicyclic derivative parp inhibitor, preparation method therefor, and use thereof | |
| TW201920160A (en) | Solid forms of 2-(5-(4-(2-morpholinoethoxy) phenyl) pyridin-2-yl)-N-benzylacetamide | |
| WO2025162420A1 (en) | Crystal form of bicyclic derivative parp inhibitor, preparation method therefor, and use thereof | |
| CN106928218A (en) | Salts of morpholine derivatives and their crystal forms, their preparation methods, pharmaceutical compositions, and uses | |
| CN118359589A (en) | CDK12 degradation inhibitors and uses thereof | |
| KR20230157471A (en) | Polymorphs of compounds and their preparation methods and uses | |
| WO2024041608A1 (en) | Crystal form of heteroaryl derivative parp inhibitor and use thereof | |
| WO2025218780A1 (en) | Crystal form of heterocyclic deuterated compound as parp inhibitor, and preparation method therefor and use thereof | |
| WO2024041605A1 (en) | Pharmaceutically acceptable salt of heteroaryl derivative parp inhibitor and use thereof | |
| WO2025176139A1 (en) | Crystal form of quinazolinone derivative, preparation method therefor and use thereof | |
| WO2021143819A1 (en) | Crystal form of polycyclic anaplastic lymphoma kinase inhibitor | |
| CN114401720A (en) | Crystal form, preparation method and use of polytyrosine kinase inhibitor | |
| WO2025218777A1 (en) | Salt form of parp inhibitor-fused heterocyclic deuterated compound, and preparation method therefor and use thereof | |
| CN119431367A (en) | A crystalline form of a pyridopyrazole derivative and its preparation method and use |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 25747983 Country of ref document: EP Kind code of ref document: A1 |