WO2020228642A9 - 一种用作iap抑制剂的smac模拟物的结晶及其制备方法 - Google Patents

一种用作iap抑制剂的smac模拟物的结晶及其制备方法 Download PDF

Info

Publication number
WO2020228642A9
WO2020228642A9 PCT/CN2020/089437 CN2020089437W WO2020228642A9 WO 2020228642 A9 WO2020228642 A9 WO 2020228642A9 CN 2020089437 W CN2020089437 W CN 2020089437W WO 2020228642 A9 WO2020228642 A9 WO 2020228642A9
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
crystal
liters
add
Prior art date
Application number
PCT/CN2020/089437
Other languages
English (en)
French (fr)
Other versions
WO2020228642A1 (zh
Inventor
刘迎春
徐招兵
胡利红
丁照中
朱兴训
陈曙辉
Original Assignee
正大天晴药业集团股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 正大天晴药业集团股份有限公司 filed Critical 正大天晴药业集团股份有限公司
Priority to BR112021022509A priority Critical patent/BR112021022509A2/pt
Priority to KR1020217040412A priority patent/KR20220006611A/ko
Priority to DK20805780.2T priority patent/DK3967702T3/da
Priority to CN202080030342.1A priority patent/CN113748119B/zh
Priority to MX2021013729A priority patent/MX2021013729A/es
Priority to CA3138411A priority patent/CA3138411A1/en
Priority to JP2021566285A priority patent/JP2022531794A/ja
Priority to AU2020274768A priority patent/AU2020274768A1/en
Priority to US17/594,737 priority patent/US20220177453A1/en
Priority to EP20805780.2A priority patent/EP3967702B1/en
Priority to FIEP20805780.2T priority patent/FI3967702T3/fi
Publication of WO2020228642A1 publication Critical patent/WO2020228642A1/zh
Publication of WO2020228642A9 publication Critical patent/WO2020228642A9/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/4045Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06026Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atom, i.e. Gly or Ala
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • This application belongs to the field of medicinal chemistry, and relates to a crystal of an SMAC mimic used as an IAP inhibitor and a preparation method thereof, and also includes the use of the crystal in the preparation of a medicine for treating cancers that benefit from cIAP1 inhibition.
  • Programmed cell death plays a key role in regulating the number of cells and removing stressed or damaged cells from normal tissues.
  • the apoptotic signaling network mechanisms inherent in most cell types provide a major barrier against the development and deterioration of human cancers.
  • the commonality of all cancer cells is that they cannot perform the apoptotic program and lack proper apoptosis due to the lack of normal apoptotic mechanisms.
  • most cancer therapies include chemotherapy, radiation therapy and immunotherapy, all of which work by indirectly inducing apoptosis of cancer cells.
  • the first type of regulatory factor is the Bcl-2 family of proteins, such as two potent anti-apoptotic molecules, Bcl-2 and Bcl-XL proteins.
  • IAPs apoptosis proteins
  • XIAP X-chromosome-linked inhibitor of apoptosis
  • NIK NF- ⁇ B-inducing kinase
  • cIAP1 and cIAP2 caused by frequent chromosomal amplification of the 11q21-q23 region (which covers two genes) has been observed in a variety of malignant diseases, including neuroblastoma and renal cell carcinoma , Colorectal cancer and gastric cancer.
  • the present application provides a crystalline composition of the compound of formula (I), wherein the crystals of the compound of formula (I) account for more than 50% of the weight of the crystalline composition, preferably more than 75%, more preferably It is 90% or more, preferably 95% or more.
  • the present application provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the crystal of the compound of formula (I) above, or the crystalline composition of the compound of formula (I);
  • the pharmaceutical composition may include at least one A pharmaceutically acceptable carrier or other excipients.
  • the present application provides a crystal of the compound of the above formula (I), a crystalline composition of the compound of the above formula (I), or the use of the above pharmaceutical composition in the preparation of a medicament for the treatment of cancer benefiting from the inhibition of cIAP1.
  • the present application provides the use of the crystal of the compound of formula (I) above, the crystalline composition of the compound of formula (I) above, or the use of the above pharmaceutical composition for the treatment of cancers that benefit from cIAP1 inhibition in mammals.
  • the present application provides a method for treating cancers that benefit from cIAP1 inhibition in mammals, which comprises administering to a mammal in need a therapeutically effective amount of crystals of the above-mentioned compound of formula (I), and the above-mentioned crystals of the compound of formula (I) Composition, or the above-mentioned pharmaceutical composition.
  • the present application provides a crystal of the compound of the above formula (I), a crystalline composition of the compound of the above formula (I), or the above pharmaceutical composition for use in the treatment of cancers that benefit from cIAP1 inhibition in a mammal.
  • One aspect of this application is to provide crystals of the compound of formula (I),
  • crystals described in this application may be in the form of unsolvated or solvated forms, such as hydrates.
  • the crystal of the compound of formula (I) is crystal A, and its X-ray powder diffraction pattern using Cu K ⁇ radiation has characteristic diffraction peaks at the following 2 ⁇ angles: 12.1° ⁇ 0.200°, 16.1° ⁇ 0.200 °, 18.5° ⁇ 0.200°, 20.2° ⁇ 0.200°, 21.3° ⁇ 0.200° and 23.0° ⁇ 0.200°.
  • the above-mentioned crystal A has characteristic diffraction peaks at the following 2 ⁇ angles in its X-ray powder diffraction pattern using Cu K ⁇ radiation: 12.1° ⁇ 0.200°, 16.1° ⁇ 0.200°, 18.5° ⁇ 0.200° , 18.8° ⁇ 0.200°, 19.2° ⁇ 0.200°, 19.8° ⁇ 0.200°, 20.2° ⁇ 0.200°, 21.3° ⁇ 0.200°, 23.0° ⁇ 0.200°, 26.6° ⁇ 0.200° and 27.4° ⁇ 0.200°.
  • the above-mentioned crystal A has characteristic diffraction peaks at the following 2 ⁇ angles in its X-ray powder diffraction pattern using Cu K ⁇ radiation: 7.0° ⁇ 0.200°, 8.7° ⁇ 0.200°, 12.1° ⁇ 0.200° , 13.2° ⁇ 0.200°, 13.9° ⁇ 0.200°, 16.1° ⁇ 0.200°, 16.7° ⁇ 0.200°, 18.5° ⁇ 0.200°, 18.8° ⁇ 0.200°, 19.2° ⁇ 0.200°, 19.8° ⁇ 0.200°, 20.2 ° ⁇ 0.200°, 21.0° ⁇ 0.200°, 21.3° ⁇ 0.200°, 23.0° ⁇ 0.200°, 24.3° ⁇ 0.200°, 25.3° ⁇ 0.200°, 26.6° ⁇ 0.200° and 27.4° ⁇ 0.200°.
  • the above-mentioned crystal A has characteristic diffraction peaks at the following 2 ⁇ angles in its X-ray powder diffraction pattern using Cu K ⁇ radiation: 7.0° ⁇ 0.200°, 8.7° ⁇ 0.200°, 12.1° ⁇ 0.200° , 13.2° ⁇ 0.200°, 13.9° ⁇ 0.200°, 16.1° ⁇ 0.200°, 16.5° ⁇ 0.200°, 16.7° ⁇ 0.200°, 18.5° ⁇ 0.200°, 18.8° ⁇ 0.200°, 19.2° ⁇ 0.200°, 19.8 ° ⁇ 0.200°, 20.2° ⁇ 0.200°, 21.0° ⁇ 0.200°, 21.3° ⁇ 0.200°, 23.0° ⁇ 0.200°, 23.2° ⁇ 0.200°, 24.3° ⁇ 0.200°, 25.3° ⁇ 0.200°, 26.6° ⁇ 0.200°, 26.9° ⁇ 0.200°, 27.4° ⁇ 0.200° and 29.4° ⁇ 0.200°.
  • the above-mentioned crystal A has characteristic diffraction peaks at the following 2 ⁇ angles in its X-ray powder diffraction pattern using Cu K ⁇ radiation: 7.0° ⁇ 0.200°, 8.7° ⁇ 0.200°, 9.7° ⁇ 0.200° , 10.6° ⁇ 0.200°, 11.4° ⁇ 0.200°, 12.1° ⁇ 0.200°, 13.2° ⁇ 0.200°, 13.9° ⁇ 0.200°, 16.1° ⁇ 0.200°, 16.5° ⁇ 0.200°, 16.7° ⁇ 0.200°, 17.5 ° ⁇ 0.200°, 18.5° ⁇ 0.200°, 18.8° ⁇ 0.200°, 19.2° ⁇ 0.200°, 19.5° ⁇ 0.200°, 19.8° ⁇ 0.200°, 20.2° ⁇ 0.200°, 21.0° ⁇ 0.200°, 21.3° ⁇ 0.200°, 22.5° ⁇ 0.200°, 23.0° ⁇ 0.200°, 23.2° ⁇ 0.200°, 23.8° ⁇ 0.200°, 24.3° ⁇ 0.200°, 24.6°
  • the peak positions and intensities of the characteristic peaks of the X-ray powder diffraction spectrum of the crystal A of the compound of formula (I) using Cu K ⁇ radiation are shown in Table 1:
  • Table 1 XRPD pattern characterization data of crystal A
  • the crystal A of the compound of formula (I) has an X-ray powder diffraction pattern using Cu K ⁇ radiation as shown in FIG. 1.
  • the crystalline A of the compound of formula (I) has an absorption peak in the differential scanning calorimetry (DSC) measurement chart at 202.5°C.
  • the crystal A of the compound of formula (I) has the initial position of the absorption peak in the differential scanning calorimetry (DSC) measurement chart at 200.8°C.
  • the differential scanning calorimetry (DSC) measurement chart of the crystal A of the compound of formula (I) is shown in FIG. 2.
  • thermogravimetric analysis (TGA) measurement chart of the crystal A of the compound of formula (I) is shown in FIG. 3.
  • this application provides a method for preparing crystalline A of the compound of formula (I), including:
  • the solvent in the above step (a) is selected from a mixed solvent of methanol, ethanol, isopropanol, ethyl acetate, acetonitrile, acetone, tetrahydrofuran, water, water and one of the above solvents; preferably methanol , Ethanol, isopropanol, or a mixed solvent with water; ethanol is more preferred.
  • this application also provides a method for preparing crystalline A of the compound of formula (I), including:
  • the solvent in the above step (d) is selected from a mixed solvent of methanol, ethanol, isopropanol, ethyl acetate, acetonitrile, acetone, tetrahydrofuran, water, water and one of the above solvents; preferably methanol , Ethanol, ethyl acetate, acetonitrile, acetone, tetrahydrofuran, water, a mixed solvent of water and methanol, a mixed solvent of water and ethanol, a mixed solvent of water and acetone.
  • the volume ratio of water to methanol, water to ethanol, and water to acetone is selected from 1:1 to 5, preferably 1:1 to 3.
  • the molar volume ratio of the compound of formula (I) to the solvent in the above step (a) is 1 mmol: 1-10 mL, preferably 1 mmol: 2-6 mL, more preferably 1 mmol: 2-4 mL.
  • the molar volume ratio of the compound of formula (I) to the solvent in step (d) is 1 mmol: 1-15 mL, preferably 1 mmol: 4-10 mL, more preferably 1 mmol: 8-10 mL.
  • the reflux temperature in step (b) above is 60-120°C, preferably 80-90°C.
  • the stirring temperature in the above step (e) is 30-50°C, preferably 40-50°C.
  • the present application provides a crystalline composition of the compound of formula (I), wherein the crystals of the compound of formula (I) account for more than 50% of the weight of the crystalline composition, preferably more than 75%, more preferably It is 90% or more, preferably 95% or more.
  • the crystalline composition may also contain a small amount of other crystalline or non-crystalline forms of the compound of formula (I).
  • the present application provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the crystal of the compound of formula (I) above, or the crystalline composition of the compound of formula (I);
  • the pharmaceutical composition may include at least one A pharmaceutically acceptable carrier or other excipients.
  • the present application provides a crystal of the compound of the above formula (I), a crystalline composition of the compound of the above formula (I), or the use of the above pharmaceutical composition in the preparation of a medicament for the treatment of cancer benefiting from the inhibition of cIAP1.
  • the present application provides the use of the crystal of the compound of formula (I) above, the crystalline composition of the compound of formula (I) above, or the use of the above pharmaceutical composition for the treatment of cancers that benefit from cIAP1 inhibition in mammals.
  • the present application provides a method for treating cancers that benefit from cIAP1 inhibition in mammals, which comprises administering to a mammal in need a therapeutically effective amount of crystals of the above-mentioned compound of formula (I), and the above-mentioned crystals of the compound of formula (I) Composition, or the above-mentioned pharmaceutical composition.
  • the present application provides a crystal of the compound of the above formula (I), a crystalline composition of the compound of the above formula (I), or the above pharmaceutical composition for use in the treatment of cancers that benefit from cIAP1 inhibition in a mammal.
  • the mammal is a human.
  • the cancer that benefits from inhibition of cIAP1 is selected from breast cancer.
  • the cancer that benefits from cIAP1 inhibition is selected from triple-negative breast cancer.
  • the crystals of the compound of formula (I) described in this application have excellent effects in at least one aspect of biological activity, safety, and bioavailability.
  • the crystal A of the compound of formula (I) has high stability and low hygroscopicity, and has excellent effects on cIAP1.
  • the inhibitory activity is better, and the value as a medicine is higher.
  • the crystalline A of the compound of formula (I) also has good pharmacokinetic properties and is suitable for use as a drug, wherein the pharmacokinetic properties can be measured in preclinical animal tests such as SD rats and beagle dogs , Can also be measured in clinical human trials.
  • the pharmaceutical composition can be made into a certain dosage form, and the route of administration is preferably oral administration, parenteral administration (including subcutaneous, intramuscular and intravenous), rectal administration and the like.
  • dosage forms suitable for oral administration include tablets, capsules, granules, powders, pills, powders, lozenges, syrups or suspensions
  • dosage forms suitable for parenteral administration include aqueous or non-aqueous injections.
  • dosage forms suitable for rectal administration include suppositories with hydrophilic or hydrophobic carriers.
  • the X-ray powder diffraction spectrum of the sample is measured under the following conditions: instrument: Bruker D8 ADVANCE X-ray diffractometer; target: Cu:K ⁇ ; wavelength 2 ⁇ angle range: 3-40°; scattering slit: 0.60mm; detector slit: 10.50mm; anti-scatter slit: 7.10mm; step diameter: 0.02°; step length: 0.12s; sample rotation speed: 15rpm; Cu target tube pressure and tube flow: 40KV, 40mA.
  • the DSC spectrum is measured under the following conditions: instrument: TA Q2000 differential scanning calorimeter; temperature range: 30 to 300°C; heating rate: 10°C/min.
  • TGA thermogravimetric analysis is measured under the following conditions: instrument: TA Q5000 thermogravimetric analyzer; temperature range: 25 to 300°C; heating rate: 10°C/min.
  • the position of the peak or the relative intensity of the peak may be different due to factors such as measuring instrument, measuring method/condition and so on.
  • the measurement error of the 2 ⁇ value may be ⁇ 0.2°. Therefore, when determining each crystal type, this error should be taken into account, and the error also belongs to the scope of this application.
  • the position of the endothermic peak of DSC may be different due to factors such as measuring instrument, measuring method/condition and so on.
  • there may be an error in the position of the endothermic peak with an error of ⁇ 5°C. Therefore, when determining each crystal type, this error should be taken into account, and the error also belongs to the scope of this application.
  • the location of the TGA weight loss temperature may be different due to factors such as the measuring instrument, the measuring method/condition and other factors.
  • the measuring instrument the measuring method/condition and other factors.
  • there may be an error in the position of the weight loss temperature with an error of ⁇ 5°C. Therefore, when determining each crystal type, this error should be taken into account, and the error also belongs to the scope of this application.
  • mammals include humans and domestic animals such as laboratory mammals and domestic pets (eg cats, dogs, pigs, sheep, cattle, sheep, goats, horses, rabbits), and non-domestic mammals such as wild mammals.
  • composition refers to a preparation of a compound of the present application and a medium generally accepted in the art for delivering a biologically active compound to a mammal such as a human.
  • the medium includes all pharmaceutically acceptable carriers for its use.
  • the pharmaceutical composition facilitates the administration of the compound to the organism.
  • terapéuticaally effective amount refers to a sufficient amount of a drug or agent that is non-toxic but can achieve the desired effect. The determination of the effective amount varies from person to person, and depends on the age and general conditions of the recipient, as well as the specific active substance. The appropriate effective amount in a case can be determined by those skilled in the art according to routine experiments.
  • pharmaceutically acceptable carriers refer to those carriers that are administered together with the active ingredient, have no obvious stimulating effect on the organism, and do not impair the biological activity and performance of the active compound.
  • pharmaceutically acceptable carriers refer to those carriers that are administered together with the active ingredient, have no obvious stimulating effect on the organism, and do not impair the biological activity and performance of the active compound.
  • Remington The Science and Practice of Pharmacy, 21st Ed., Lippincott, Williams & Wilkins (2005), and the content of this document is incorporated herein by reference.
  • room temperature means 20°C to 30°C.
  • FIG. 1 is an X-ray powder diffraction (XRPD) pattern of crystal A of the compound of formula (I) in Example 1.
  • FIG. 1 is an X-ray powder diffraction (XRPD) pattern of crystal A of the compound of formula (I) in Example 1.
  • FIG. 2 is a differential scanning calorimetry (DSC) chart of crystal A of the compound of formula (I) in Example 1.
  • DSC differential scanning calorimetry
  • FIG. 3 is a thermogravimetric analysis (TGA) chart of crystal A of the compound of formula (I) in Example 1.
  • TGA thermogravimetric analysis
  • the intermediate compounds of the present application can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by their combination with other chemical synthesis methods, and those of those skilled in the art.
  • Well-known equivalent alternatives, and preferred implementations include but are not limited to the examples of the present application.
  • step 1
  • dichloromethane (16.0 liters) to the reaction kettle, and start to cool down to 0°C, add compound 1 (2.0 kg, 9.94 moles, 1.0 equivalents) and stir, then add triethylamine (4.42 liters, 29.81 moles, 3.0 equivalents) And 4-dimethylaminopyridine (61.00 g, 496.86 mmol, 0.05 equivalent), add p-toluenesulfonyl chloride (2.18 kg, 11.43 mol, 1.15 equivalent) in batches, control the internal temperature at 0-10°C, and then add to the reactor Dichloromethane (4 liters) was added to the mixture, and the temperature was raised to 20°C and stirring was continued for 16 hours.
  • N,N-dimethylacetamide (6.0 liters) was added to the reaction kettle, and compound 2 (3.95 kg, 11.12 mol, 1.30 equivalent) and compound 4 (1.73 kg, 8.56 mol, 1.0 equivalent) were added at 25°C ), add cesium carbonate (4.18 kg, 12.85 mol, 1.50 equivalent) in batches, and add it in 10 minutes.
  • the wall of the reactor is rinsed with N,N-diethylacetamide (1.0 liter), and the temperature is raised to 80-85°C to react 16 Hour.
  • reaction solution was filtered, the filter cake and still wall solids were dissolved in methanol (7.0 liters), the organic phase was transferred to a rotary evaporator, and concentrated under reduced pressure to obtain a solid, which was suspended in ethyl acetate (8.85 liters) and stirred overnight.
  • the mixture is filtered, and the filter cake is vacuum-dried at 35-45° C. to a constant weight to obtain compound 9.
  • N,N-dimethylformamide (11.88 liters) into the reactor, adjust the temperature of the reactor to 0 ⁇ 5°C, add compound 10 (1.36 kg, 6.71 mol, 1.05 equivalent) and benzotriazole- N,N,N,N,-tetramethylurea hexafluorophosphate (2.54 kg, 6.71 mol, 1.05 equivalent), the solid cannot be completely dissolved, and a white suspension is obtained.
  • Add N,N-diisopropyl group dropwise Ethylamine (2.48 kg, 19.16 moles, 3.0 equivalents), control the temperature of the reactor to 0-5°C, and the solids gradually dissolve to obtain a clear solution.
  • Methyl tert-butyl ether (10.0 liters) was added to the reaction kettle, the reaction solution was stirred for 2.0 hours, the reaction solution was filtered, and the filter cake was drained. The filter cake and the solid on the wall of the kettle were dissolved in water (30.0 liters), the internal temperature was reduced to 5-10°C, and the mixture was extracted twice with ethyl acetate (10.0 liters*2), and the aqueous phase was collected.
  • Test conditions Take the compound crystal A (10-15 mg) of formula (I) and place it in the DVS sample pan for testing.
  • the DVS parameters are as follows:
  • the weight gain by dampening is not less than 15% Hygroscopic Moisture absorption weight gain is less than 15% but not less than 2%
  • Slightly hygroscopic Moisture absorption weight gain is less than 2% but not less than 0.2%
  • No or almost no hygroscopicity Moisture gain is less than 0.2%
  • the compound crystal A of formula (I) is slightly hygroscopic, easy to store, not suitable for deliquescent, deformation, mildew and other phenomena, and has a high pharmaceutical value.
  • Influencing factor experiment Weigh 12 parts of compound crystal A of formula (I), 1.50g each, and investigate 3 parts under each condition. Put each sample into an open weighing bottle, put it in a desiccator under different conditions at high temperature, and then put it in a corresponding stability box for investigation; put it in a stable box under high humidity conditions for investigation.
  • Light stability test Weigh 4 parts of compound crystal A of formula (I), each 1.50g, 2 parts are light samples (one is a 5-day light sample, the other is a 10-day light sample), and the other two are Control sample (one is a 5-day control sample, the other is a 10-day control sample), the illuminated sample is placed in a clean weighing bottle, flattened into a single layer, not covered by anything, covered with a transparent lid, Then put it into the light box for light.
  • the packaging method of the control sample is the same as that of the light sample, but the outside of the weighing bottle is covered with aluminum film.
  • Table 4 Test results of crystal A and related substances in the stability and light stability test of influencing factors
  • Table 5 Test results of crystal A and related substances in the accelerated stability test
  • the XRPD test results show that the crystal A of the compound of formula (I) is consistent with day 0, and the crystal A of the compound of formula (I) is stable under various test conditions.
  • Test buffer system 0.1 mol/L potassium phosphate, pH 7.5, 0.1% bovine serum albumin, 0.005% Triton X-100 and 1% dimethyl sulfoxide.
  • Probe ARPFAQ-K(5-FAM)-NH 2 .
  • cIAP1-BIR3-his RBC (Cat#APT-11-370), the BIR3 domain of human cIAP1 (covering amino acids 258 to 363; cIAP1 BIR3) was expressed and purified from E. coli as a GST-fusion protein.
  • XIAP-BIR3-his RBC (Cat#APT-11-374), the BIR3 domain of XIAP (covering amino acids 255 to 356; XIAP BIR3) was expressed and purified from E. coli as a GST-fusion protein.
  • Reaction conditions 5nM ARPFAQ-K(5-FAM)-NH 2 , 20nM cIAP1 BIR3 and 30nM XIAP BIR3.
  • the compound of formula (I) of the present application shows good binding activity of cIAP1 and BIR3, and shows good selectivity for cIAP1 and XIAP.
  • RPMI 1640 medium Invitrogen-22400089
  • fetal bovine serum Invitrogen-10099141
  • Trypsin 0.05% EDTA ⁇ 4Na
  • luminescence cell viability detection kit Promega-G7573
  • Dulbecco's phosphate buffer HyClone-SH30028.01B
  • 384-well plate Corning-6007680. Envision multi-label analyzer.
  • test compound is made at a high concentration of 10 ⁇ M, and the test compound is diluted by 5 times, and each compound is diluted by 10 concentration gradients), and then the cell plate is returned to the carbon dioxide incubator for culture 7 days.
  • the compound of formula (I) of the present application has good MDA-MB-231 cell anti-proliferation activity.
  • mice Female, 6-8 weeks old, weighing about 18-22 grams.
  • the mice are kept in a special pathogen-free environment and in a single ventilated cage (3 mice per cage). All cages, bedding and water are disinfected before use. All animals have free access to standard certified commercial laboratory diets.
  • a total of 48 mice purchased from Shanghai BK Laboratory Animal Co., LTD were used for research.
  • Each mouse was implanted with tumor cells (10 ⁇ 10 6 in 0.2 ml phosphate buffer) subcutaneously in the right flank for tumor growth. The administration was started when the average tumor volume reached approximately 147 cubic millimeters.
  • the test compound was orally administered daily at a dose of 30 mg/kg.
  • the anti-tumor efficacy is determined by dividing the average tumor increase volume of animals treated with the compound by the average tumor increase volume of untreated animals.
  • the compound of formula (I) of the present application exhibits better pharmacodynamics.
  • mice Female, 6-8 weeks old, weighing about 18-22 grams.
  • the mice are kept in a special pathogen-free environment and in a single ventilated cage (3 mice per cage). All cages, bedding and water are disinfected before use. All animals have free access to standard certified commercial laboratory diets.
  • a total of 48 mice purchased from Shanghai BK Laboratory Animal Co., LTD were used for research.
  • Each mouse was implanted with tumor cells (10 ⁇ 10 6 in 0.2 ml phosphate buffer) subcutaneously in the right flank for tumor growth. The administration was started when the average tumor volume reached approximately 110 cubic millimeters.
  • the test compound was orally administered daily at a dose of 30 mg/kg.
  • the anti-tumor efficacy is determined by dividing the average tumor increase volume of animals treated with the compound by the average tumor increase volume of untreated animals.
  • the compound of formula (I) of the present application exhibits better pharmacodynamics.

Abstract

一种用作IAP抑制剂的SMAC模拟物的结晶及其制备方法,还包括所述结晶在制备治疗受益于cIAP1抑制的癌症的药物中的用途。所述式(I)化合物的结晶稳定性高、吸湿性小,在物理性质、安全性和代谢稳定性方面具有优势,有较高的成药价值。

Description

一种用作IAP抑制剂的SMAC模拟物的结晶及其制备方法
相关申请的交叉引用
本申请要求于2019年5月10日向中国国家知识产权局提交的第201910389970.8号中国专利申请的优先权和权益,所有申请公开的内容通过引用整体并入本文中。
技术领域
本申请属于医药化学领域,涉及一种用作IAP抑制剂的SMAC模拟物的结晶及其制备方法,还包括所述结晶在制备治疗受益于cIAP1抑制的癌症的药物中的用途。
背景技术
程序性细胞死亡在调节细胞数量和从正常组织中清除应激或受损细胞方面起关键作用。实际上,大部分细胞类型中所固有的凋亡信号传导网络机制提供了抗人类癌症发展和恶化的主要屏障。然而,所有癌细胞的共同性在于它们无法执行凋亡程序,并由于正常凋亡机制的缺失而缺乏适当的凋亡。目前大多数癌症疗法包括化疗、放射疗法和免疫疗法,均通过间接诱导癌细胞凋亡发挥作用。因而,癌细胞由于正常凋亡机制的缺陷而无法执行凋亡程序通常与对化疗、放射疗法或免疫疗法诱导的细胞凋亡的抗性增强有关。因此,以直接抑制癌细胞凋亡中扮演了重要角色的关键负调控因子为目标将成为极有希望的用于新的抗癌药物设计的治疗策略。
目前已经确认了两类重要的细胞凋亡的负调节因子。第一类调节因子是Bcl-2族蛋白,例如两个有效的抗凋亡分子,Bcl-2和Bcl-XL蛋白。
细胞凋亡的第二类重要的负调节因子是细胞凋亡蛋白的抑制剂(IAPs)。IAP最初在杆状病毒中由于其能够取代P35蛋白质功能而被发现。这类蛋白包括:XIAP、cIAP1、cIAP2、ML-IAP、ILP-2、NAIP、Apollon和Survivin。其中X染色体连锁的细胞凋亡抑制剂(XIAP)通过直接抑制半胱天冬酶-3,半胱天冬酶-7和半胱天冬酶-9,起到对抗凋亡的作用。cIAPs则主要是通过阻断死亡受体通路抑制凋亡,随着cIAPs的降解,其底物NIK(NF-κB-inducing kinase)免于被降解而积聚,积聚的NIK通过非经典通路激活NF-κB;NF-κB的激活促进TNFα分泌,TNFα与TNF-R1(TNF receptor1)结合启动死亡受体通路,由于cIAPs的降解,RIPK1(receptor interacting protein kinase 1,受体相互作用蛋白激酶1)的分泌也会增加,其与FADD(Fas-associated death domin,Fas相关的死亡结构域)和caspase-8(半胱天冬酶-8)一起形成促凋亡RIPK1-FADD-caspase-8复合物,然后半胱天冬酶-3激活,导致细胞凋亡。
已在多种恶性疾病中观察到由11q21-q23区(其涵盖两种基因)的频繁染色体扩增而导致的cIAP1及cIAP2的过表达,该多种恶性疾病包括神经胚细胞瘤、肾细胞癌、结直肠癌及胃癌等。
发明概述
一方面,本申请提供式(I)化合物的结晶
Figure PCTCN2020089437-appb-000001
另一方面,本申请提供了一种式(I)化合物的结晶组合物,其中上述式(I)化合物的结晶占所述结晶组合物重量的50%以上,较好是75%以上,更好是90%以上,最好是95%以上。
另一方面,本申请提供一种药物组合物,其包含治疗有效量的上述式(I)化合物的结晶,或上述式(I)化合物的结晶组合物;所述药物组合物可以包含至少一种药学上可接受的载体或其他赋形剂。
另一方面,本申请提供了上述式(I)化合物的结晶,上述式(I)化合物的结晶组合物,或上述药物组合物在制备治疗受益于cIAP1抑制的癌症的药物中的用途。
另一方面,本申请提供了上述式(I)化合物的结晶,上述式(I)化合物的结晶组合物,或上述药物组合物用于治疗哺乳动物受益于cIAP1抑制的癌症的应用。
另一方面,本申请提供了治疗哺乳动物受益于cIAP1抑制的癌症的方法,其包括向有需要的哺乳动物给予治疗有效量的上述式(I)化合物的结晶,上述式(I)化合物的结晶组合物,或上述药物组合物。
另一方面,本申请提供了用于治疗哺乳动物受益于cIAP1抑制的癌症的上述式(I)化合物的结晶,上述式(I)化合物的结晶组合物,或上述药物组合物。
发明内容
本申请的一个方面在于提供式(I)化合物的结晶,
Figure PCTCN2020089437-appb-000002
本申请所述结晶可以是非溶剂合物的形式,也可以是溶剂合物的形式,例如水合物。
本申请的一个实施方案中,式(I)化合物的结晶为结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.1°±0.200°、16.1°±0.200°、18.5°±0.200°、20.2°±0.200°、21.3°±0.200°和23.0°±0.200°。
本申请的一个实施方案中,上述结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.1°±0.200°、16.1°±0.200°、18.5°±0.200°、18.8°±0.200°、19.2°±0.200°、19.8°±0.200°、20.2°±0.200°、21.3°±0.200°、23.0°±0.200°、26.6°±0.200°和27.4°±0.200°。
本申请的一个实施方案中,上述结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0°±0.200°、8.7°±0.200°、12.1°±0.200°、13.2°±0.200°、13.9°±0.200°、16.1°±0.200°、16.7°±0.200°、18.5°±0.200°、18.8°±0.200°、19.2°±0.200°、19.8°±0.200°、20.2°±0.200°、21.0°±0.200°、21.3°±0.200°、23.0°±0.200°、24.3°±0.200°、25.3°±0.200°、26.6°±0.200°和27.4°±0.200°。
本申请的一个实施方案中,上述结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0°±0.200°、8.7°±0.200°、12.1°±0.200°、13.2°±0.200°、13.9°±0.200°、16.1°±0.200°、16.5°±0.200°、16.7°±0.200°、18.5°±0.200°、18.8°±0.200°、19.2°±0.200°、19.8°±0.200°、20.2°±0.200°、21.0°±0.200°、21.3°±0.200°、23.0°±0.200°、23.2°±0.200°、24.3°±0.200°、25.3°±0.200°、26.6°±0.200°、26.9°±0.200°、27.4°±0.200°和29.4°±0.200°。
本申请的一个实施方案中,上述结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0°±0.200°、8.7°±0.200°、9.7°±0.200°、10.6°±0.200°、11.4°±0.200°、12.1°±0.200°、13.2°±0.200°、13.9°±0.200°、16.1°±0.200°、16.5°±0.200°、16.7°±0.200°、17.5°±0.200°、18.5°±0.200°、18.8°±0.200°、19.2°±0.200°、19.5°±0.200°、19.8°±0.200°、20.2°±0.200°、21.0°±0.200°、21.3°±0.200°、22.5°±0.200°、23.0°±0.200°、23.2°±0.200°、23.8°±0.200°、24.3°±0.200°、24.6°±0.200°、25.3°±0.200°、25.8°±0.200°、26.6°±0.200°、26.9°±0.200°、27.4°±0.200°、28.1°±0.200°、29.1°±0.200°、29.4°±0.200°、30.1°±0.200°、30.6°±0.200°、30.8°±0.200°、31.3°±0.200°、31.8°±0.200°、32.2°±0.200°、32.8°±0.200°、33.7°±0.200°、34.0°±0.200°、34.8°±0.200°、35.5°±0.200°、36.6°±0.200°、37.6°±0.200°、38.6°±0.200°和39.6°±0.200°。
本申请的一个实施方案中,式(I)化合物的结晶A的使用Cu Kα辐射的X-射线粉末衍射光谱特征峰的峰位置及强度如表1所示:
表1:结晶A的XRPD图谱表征数据
编号 2θ角(°) 相对强度(%) 编号 2θ角(°) 相对强度(%)
1 7.0 8.2 26 24.6 7.4
2 8.7 6.0 27 25.3 10.9
3 9.7 1.5 28 25.8 1.5
4 10.6 3.9 29 26.6 20.3
5 11.4 2.9 30 26.9 9.9
6 12.1 49.6 31 27.4 21.4
7 13.2 13.2 32 28.1 3.8
8 13.9 18.8 33 29.1 2.5
9 16.1 34.9 34 29.4 9.7
10 16.5 9.6 35 30.1 1.9
11 16.7 12.2 36 30.6 2.1
12 17.5 4.1 37 30.8 2.7
13 18.5 100.0 38 31.3 1.6
14 18.8 23.2 39 31.8 2.5
15 19.2 24.5 40 32.2 5.5
16 19.5 5.6 41 32.8 3.5
17 19.8 21.9 42 33.7 8.2
18 20.2 55.8 43 34.0 6.9
19 21.0 12.7 44 34.8 3.3
20 21.3 41.5 45 35.5 1.9
21 22.5 7.7 46 36.6 3.9
22 23.0 43.2 47 37.6 4.6
23 23.2 9.9 48 38.6 3.1
24 23.8 2.5 49 39.6 2.6
25 24.3 12.0      
本申请的一个实施方案中,式(I)化合物的结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱如图1所示。
本申请的一个实施方案中,式(I)化合物的结晶A,其差示扫描量热(DSC)测量图中吸收峰的峰值在202.5℃处。
本申请的一个实施方案中,式(I)化合物的结晶A,其差示扫描量热(DSC)测量图中吸收峰的起始位置在200.8℃处。
本申请的一个实施方案中,式(I)化合物的结晶A,其差示扫描量热(DSC)测量图如图2所示。
本申请的一个实施方案中,式(I)化合物的结晶A,其热重分析(TGA)测量图如图3所示。
另一方面,本申请提供一种式(I)化合物的结晶A的制备方法,包括:
(a)式(I)化合物于溶剂中,使其成混悬液或溶液;
(b)上述混悬液或溶液于加热回流,使其溶清;
(c)上述溶清液趁热过滤,冷却析晶,过滤,干燥,得到式(I)化合物的结晶A。
本申请的一个实施方案中,上述步骤(a)中的溶剂选自甲醇、乙醇、异丙醇、乙酸乙酯、乙腈、丙酮、四氢呋喃、水、水和上述一种溶剂的混合溶剂;优选甲醇、乙醇、异丙醇、或其与水的混合溶剂;更优选乙醇。
另一方面,本申请还提供一种式(I)化合物的结晶A的制备方法,包括:
(d)式(I)化合物于溶剂中,超声混匀,使其成混悬液或溶液;
(e)上述混悬液或溶液于恒温振荡仪上加热搅拌,离心,干燥,得到式(I)化合物的结晶A。
本申请的一个实施方案中,上述步骤(d)中的溶剂选自甲醇、乙醇、异丙醇、乙酸乙酯、乙腈、丙酮、四氢呋喃、水、水和上述一种溶剂的混合溶剂;优选甲醇、乙醇、乙酸乙酯、乙腈、丙酮、四氢呋喃、水、水和甲醇的混合溶剂、水和乙醇的混合溶剂、水和丙酮的混合溶剂。所述水和甲醇、水和乙醇、水和丙酮的体积比选自1:1~5,优选1:1~3。
本申请的一个实施方案中,上述步骤(a)中式(I)化合物与溶剂的摩尔体积比为1mmol:1~10mL,优选1mmol:2~6mL,更优选1mmol:2~4mL。
本申请的一个实施方案中,上述步骤(d)中式(I)化合物与溶剂的摩尔体积比为1mmol:1~15mL,优选1mmol:4~10mL,更优选1mmol:8~10mL。
本申请的一个实施方案中,上述步骤(b)所述回流温度为60~120℃,优选80~90℃。
本申请的一个实施方案中,上述步骤(e)所述搅拌温度为30~50℃,优选40~50℃。
另一方面,本申请提供了一种式(I)化合物的结晶组合物,其中上述式(I)化合物的结晶占所述结晶组合物重量的50%以上,较好是75%以上,更好是90%以上,最好是95%以上。所述结晶组合物中,还可能含有少量的式(I)化合物其它结晶或非结晶形式。
另一方面,本申请提供一种药物组合物,其包含治疗有效量的上述式(I)化合物的结晶,或上述式(I)化合物的结晶组合物;所述药物组合物可以包含至少一种药学上可接受的载体或其他赋形剂。
另一方面,本申请提供了上述式(I)化合物的结晶,上述式(I)化合物的结晶组合物,或上述药物组合物在制备治疗受益于cIAP1抑制的癌症的药物中的用途。
另一方面,本申请提供了上述式(I)化合物的结晶,上述式(I)化合物的结晶组合物,或上述药物组合物用于治疗哺乳动物受益于cIAP1抑制的癌症的应用。
另一方面,本申请提供了治疗哺乳动物受益于cIAP1抑制的癌症的方法,其包括向有需要的哺乳动物给予治疗有效量的上述式(I)化合物的结晶,上述式(I)化合物的结晶组合物,或上述药物组合物。
另一方面,本申请提供了用于治疗哺乳动物受益于cIAP1抑制的癌症的上述式(I)化合物的结晶,上述式(I)化合物的结晶组合物,或上述药物组合物。
本申请的一个实施方案中,所述哺乳动物为人类。
本申请的一个实施方案中,所述受益于cIAP1抑制的癌症选自乳腺癌。
本申请的一个实施方案中,所述受益于cIAP1抑制的癌症选自三阴性乳腺癌。
本申请所述式(I)化合物的结晶在生物活性、安全性及生物利用度等至少一方面具有优异的效果,式(I)化合物的结晶A稳定性高、吸湿性小,且对cIAP1的抑制活性较好,作为药物的价值较高。式(I)化合物的结晶A还具有良好的药代动力学性质,适合作为药物使用,其中所述药代动力学性质可以在临床前的例如SD大鼠、比格犬的动物试验中测得,也可以在临床的人体试验中测得。
本申请中,药物组合物可制成一定的剂型,给药途径优选经口服给药、肠胃外给药(包括皮下、肌肉内和静脉内)、直肠给药等。例如,适合经口给药的剂型包括片剂、胶囊剂、颗粒剂、散剂、丸剂、粉剂、锭剂、糖浆剂或混悬剂;适合肠胃外给药的剂型包括水性或非水性的注射用溶液或乳液;适合直肠给药的剂型包括使用亲水性或疏水性载体的栓剂。
本申请中,样品的X-射线粉末衍射光谱在下述条件下测定:仪器:Bruker D8 ADVANCE X射线衍射仪;靶:Cu:Kα;波长
Figure PCTCN2020089437-appb-000003
2θ角范围:3-40°;散射狭缝:0.60mm;探测器狭缝:10.50mm;防散射狭缝:7.10mm;步径:0.02°;步长:0.12s;样品旋转速度:15rpm;Cu靶管压及管流:40KV,40mA。
本申请中,DSC光谱在下述条件下测定:仪器:TA Q2000差示扫描量热仪;温度范围:30~300℃;升温速率:10℃/min。
本申请中,TGA热重分析在下述条件下测定:仪器:TA Q5000热重分析仪;温度范围:25~300℃;升温速率:10℃/min。
需要说明的是,在粉末X-射线衍射光谱中,峰的位置或峰的相对强度可能会因为测定仪器、测定方法/条件等因素而产生差异。对任何特定的晶型,峰的位置可能存在误差,2θ值的测定误差可以为±0.2°。因此,在确定每种晶型时,应该将此误差考虑在内,在误差内也属于本申请的范围。
需要说明的是,对于同种晶型,DSC的吸热峰出现位置可能会因为测定仪器、测定方法/条件等因素而产生差异。对任何特定的晶型,吸热峰的位置可能存在误差,误差为±5℃。因此,在确定每种晶型时,应该将此误差考虑在内,在误差内也属于本申请的范围。
需要说明的是,对于同种晶型,TGA的失重温度的出现位置可能会因为测定仪器、测定方法/条件等因素而产生差异。对任何特定的晶型,失重温度的位置可能存在误差,误差为±5℃。因此,在确定每种晶型时,应该将此误差考虑在内,在误差内也属于本申请的范围。
定义和说明
当用于本申请的说明书与权利要求中时,除非有相反的指定,否则下列术语具有所指示的意义:
“哺乳动物”包括人和家畜如实验室哺乳动物与家庭宠物(例如猫、狗、猪、羊、牛、绵羊、山羊、马、家兔),及非驯养哺乳动物,如野生哺乳动物等。
术语“药物组合物”是指本申请化合物与本领域中通常接受的用于传递生物活性化合物至哺乳动物例如人的介质的制剂。所述介质包括所有供其使用的药物可接受的载体。药物组合物有利于化合物向生物体的给药。
术语“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。有效量的确定因人而异,取决于受体的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
本申请中,“药学上可接受的载体”是指与活性成份一同给药的、对有机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些载体。关于载体的其他信息,可以参考Remington:The Science and Practice ofPharmacy,21st Ed.,Lippincott,Williams&Wilkins(2005),该文献的内容通过引用的方式并入本文。
本申请中,“室温“是指20℃~30℃。
附图说明
图1为实施例1中式(I)化合物的结晶A的X射线粉末衍射(XRPD)图。
图2为实施例1中式(I)化合物的结晶A的差示扫描量热(DSC)图。
图3为实施例1中式(I)化合物的结晶A的热重分析(TGA)图。
具体实施方式
下面的具体实施例,其目的是使本领域的技术人员能更清楚地理解和实施本申请。它们不应该被认为是对本申请范围的限制,而只是本申请的示例性说明和典型代表。
本申请的中间体化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本申请的实施例。
本申请具体实施方式的化学反应是在合适的溶剂中完成的,所述的溶剂须适合于本申请的化学变化及其所需的试剂和物料。为了获得本申请的化合物,有时需要本领域技术人员在已有实施方式的基础上对合成步骤或者反应流程进行修改或选择。
本申请所使用的所有溶剂是市售的,无需进一步纯化即可使用。
实施例1:式(I)化合物结晶A的制备
Figure PCTCN2020089437-appb-000004
第1步:
向反应釜中加入二氯甲烷(16.0升),并开始降温至0℃,加入化合物1(2.0千克,9.94摩尔,1.0当量)搅拌,再加入三乙胺(4.42升,29.81摩尔,3.0当量)和4-二甲氨基吡啶(61.00克,496.86毫摩尔,0.05当量),分批加入对甲苯磺酰氯(2.18千克,11.43摩尔,1.15当量),控制内温在0-10℃,再向反应釜中加入二氯甲烷(4升),升温至20℃继续搅拌16小时。向反应釜中加入水(20升),搅拌5分钟静置分液,除去水相,有机相用10%的柠檬酸水溶液(15.0升*2)和食盐水(15.0升*2)洗涤,用无水硫酸钠(2.0千克)干燥1小时,过滤,滤液转入旋转蒸发仪浓缩,得到化合物2。 1H NMR(400MHz,DMSO-d 6)δ7.78(br d,J=8.2Hz,2H),7.47(br d,J=7.9Hz,2H),4.13-3.93(m,2H),3.92-3.81(m,1H),3.22-3.11(m,2H),1.96-1.80(m,2H),1.74-1.65(m,2H),1.40(s,3H),1.38-1.23(m,9H)。
第2步:
向干燥的反应釜中加入二氯甲烷(1.30升),加入化合物3(1.28千克,8.49摩尔,1.0当量)搅拌溶解,并开始降温,冷却至内温0℃,氮气保护下滴加乙酰氯(606.0毫升,8.49摩尔,1.0当量),半小时加完,再向反应釜中滴加1M的二乙基氯化铝的正己烷溶液(8.50升,8.49摩尔,1.0当量),控制内温0-5℃,3小时加完,加完后反应1小时。将反应液缓慢分批倒入搅拌下的冰水中(20.0升),大量红色固体析出,将淬灭后的混合物转入旋转蒸发仪,30℃浓缩,除掉二氯甲烷和正己烷,残留物中加入2-甲基四氢呋喃(20.0升),转入分液器,搅拌15分钟后分液,除去水相,有机相用食盐水(20升*2)洗涤,用无水硫酸钠(3.0 千克)干燥,过滤,滤饼用2-甲基四氢呋喃(2升)洗涤,滤液转入旋转蒸发仪浓缩,残余物转入50升反应釜中,加入乙酸乙酯:正庚烷(V/V)=1:1(10.0升)搅拌10小时,过滤,滤饼用乙酸乙酯:正庚烷(V/V)=1:1(300毫升)淋洗,抽干,转入烘箱中35℃减压烘干得到化合物4。 1H NMR(400MHz,DMSO-d 6)δ12.10(br s,1H),8.38(d,J=3.2Hz,1H),8.16(d,J=2.2Hz,1H),7.50(d,J=8.7Hz,1H),7.23(dd,J=8.6,2.1Hz,1H),2.46(s,3H)。
第3步:
氮气保护下向反应釜中加入N,N-二甲基乙酰胺(6.0升),25℃下加入化合物2(3.95千克,11.12摩尔,1.30当量)和化合物4(1.73千克,8.56摩尔,1.0当量),分批加入碳酸铯(4.18千克,12.85摩尔,1.50当量),10分钟加完,反应釜壁用N,N-二乙基乙酰胺(1.0升)冲洗,升温至80-85℃反应16小时。降温至25℃,向反应液中缓慢加入水(30升),加入乙酸乙酯(20升)搅拌,转入分液器分液,水相用乙酸乙酯(10.0升)萃取1次,合并有机相,用水(15.0升*2)和食盐水(15.0升*2)洗涤,用无水硫酸钠(3.0千克)干燥1小时,过滤,滤饼用乙酸乙酯(2.0升)淋洗,滤液转入旋转蒸发仪浓缩得到粗品,转入反应釜中,加入乙酸乙酯:正庚烷(V/V)=1:2(9.0升)搅拌2小时,过滤,滤饼用乙酸乙酯:正庚烷(V/V)=1:2(500毫升)淋洗,抽干,转入烘箱中35℃减压烘干得到化合物5。 1H NMR(400MHz,DMSO-d 6)δ8.43-8.28(m,1H),8.17(s,1H),7.74-7.52(m,1H),7.27(br s,1H),4.40-4.05(m,3H),3.31-3.11(m,2H),2.50-2.35(m,3H),1.90-1.69(m,4H),1.39(br d,J=6.7Hz,5H),1.08(br s,4H)。
第4步:
向干燥的反应釜中加入乙酸乙酯(16.5升),加入化合物5(1.65千克,4.38摩尔,1.0当量)搅拌溶解,并开始降温,冷却至内温0℃,滴加新制的盐酸乙酸乙酯溶液(15.0升,4.0摩尔/升),1小时加完,恢复至室温20~30℃反应16小时。将反应液过滤,滤饼用乙酸乙酯(5升*2)漂洗,滤饼抽干得到粗品,粗品悬浮在乙酸乙酯中搅拌4小时。过滤,烘干得到化合物6。 1H NMR(400MHz,DMSO-d 6)δ10.00(br s,1H),9.54(br s,1H),8.76(s,1H),8.16(d,J=1.8Hz,1H),7.84(d,J=8.8Hz,1H),7.30(dd,J=8.7,1.8Hz,1H),4.80-4.72(m,1H),4.62(br d,J=5.4Hz,1H),3.92(br s,1H),3.27(br d,J=6.8Hz,1H),3.18-3.00(m,1H),2.44(s,3H),2.17-1.63(m,4H)。
第5步:
将化合物7(2.13千克,8.30摩尔,1.05当量)、N,N-二甲基甲酰胺(12.50升)依次加入反应釜中,调节反应釜温度至0~10℃,加入苯并三氮唑-N,N,N,N,-四甲基脲六氟磷酸酯(3.15千克,8.30摩尔,1.05当量),得到白色的悬浊液,滴加N,N-二异丙基乙胺(3.06千克,23.71摩尔,3.0当量),控制内温不超过10℃,期间固体逐渐溶解得到澄清溶液。加毕,控制内温5~10℃,搅拌反应0.5小时。分批加入化合物6(2.50千克,7.90摩尔,1.0当量),控制内温5~10℃,期间产生白烟。加毕,逐渐升温至25~30℃,反应1.5小时。搅拌下,将一半反应液缓慢滴加到装有水(30.0升)的反应釜中,大量固体析出,搅拌30分钟后,抽滤,滤饼 抽干,同样方法处理另一半反应液。滤饼一起于50℃下真空烘干至恒重,得到化合物8。 1H NMR(400MHz,DMSO-d 6)δ8.47-8.41(m,1H),8.15(d,J=2.1Hz,1H),7.97-7.91(m,1H),7.32(dd,J=8.7,2.0Hz,1H),6.87(br d,J=8.4Hz,1H),4.49-4.30(m,2H),4.15-3.89(m,2H),3.62(br dd,J=8.0,5.0Hz,2H),2.44(s,3H),2.17-1.97(m,1H),1.96-1.85(m,1H),1.78-1.52(m,9H),1.38-1.31(m,9H),1.20-1.06(m,4H)。
第6步:
将乙酸乙酯(17.70升)加入到50升的反应釜中,调节反应釜温度至0~5℃,加入化合物8(1.70千克,3.37摩尔,1.0当量),得到白色的悬浊液,用蠕动泵加入新制的盐酸乙酸乙酯溶液(17.70升,4摩尔/升,21.0当量),期间固体逐渐溶解得到红棕色溶液。加毕,逐渐升温至10~25℃反应2.5小时,有大量红棕色固体析出。反应液过滤,滤饼及釜壁固体用甲醇(7.0升)溶解,将有机相转入旋转蒸发仪,减压浓缩至得到固体,所得固体悬浮在乙酸乙酯(8.85升)中搅拌过夜。混合物过滤,滤饼于35~45℃下真空烘干至恒重,得到化合物9。 1H NMR(400MHz,DMSO-d 6)δ8.50(s,1H),8.30(br d,J=3.1Hz,3H),8.15(d,J=2.1Hz,1H),7.89(d,J=8.8Hz,1H),7.31(dd,J=2.1,8.7Hz,1H),4.56-4.45(m,1H),4.45-4.36(m,1H),4.15(dd,J=13.8,8.3Hz,1H),3.86(br t,J=5.1Hz,1H),3.70-3.62(m,1H),3.61-3.56(m,1H),2.44(s,3H),2.19-2.03(m,1H),1.96-1.87(m,1H),1.83-1.66(m,5H),1.60(br d,J=10.3Hz,3H),1.14-0.93(m,4H)。
第7步:
将N,N-二甲基甲酰胺(11.88升)加入到反应釜中,调节反应釜温度至0~5℃,加入化合物10(1.36千克,6.71摩尔,1.05当量)和苯并三氮唑-N,N,N,N,-四甲基脲六氟磷酸酯(2.54千克,6.71摩尔,1.05当量),固体不能完全溶清,得到白色悬浊液,滴加N,N-二异丙基乙胺(2.48千克,19.16摩尔,3.0当量),控制反应釜温度至0~5℃,固体逐渐溶解得到澄清溶液。加毕,控制反应釜温度0~5℃反应30分钟,缓慢加入化合物9(2.9千克,6.39摩尔,1.0当量),期间有白烟生成,放热明显,控制加料速度使反应温度控制在0~5℃,加毕,升温至25~30℃反应23小时。反应液分5次处理,搅拌下,将反应液(约5.0升)滴加到35.0升水中,大量白色固体析出,搅拌30分钟后过滤,滤饼抽干,合并滤饼。滤饼及釜壁固体用乙酸乙酯(30.0升)溶解后,无水硫酸钠干燥,过滤,滤液减压浓缩至得到化合物11。1H NMR(400MHz,CDCl 3)δ8.34(d,J=2.0Hz,1H),7.73(s,1H),7.67(br d,J=8.7Hz,1H),7.29-7.22(m,1H),6.78(br s,1H),4.78-4.45(m,4H),3.88-3.55(m,4H),2.84-2.76(m,3H),2.50(s,3H),2.06-1.94(m,2H),1.73(br d,J=10.3Hz,6H),1.51(s,9H),1.39(d,J=6.7Hz,2H),1.33(br d,J=7.0Hz,3H),1.16(br s,4H)。
第8步:
将乙酸乙酯(10.0升)加入到反应釜中,调节反应釜温度至0~5℃,将化合物11(3.84千克,6.03摩尔,1.0当量)用乙酸乙酯(9.20升)溶解后加入反应釜中。用蠕动泵加入新制的盐酸乙酸乙酯溶液(4摩尔/升,19.20升),流速为0.50升/分钟,加毕,逐渐升温至室温至25~30℃反应19小时,釜壁出现大量固体。反应釜中加入甲基叔丁基醚(10.0升),反应液搅拌2.0小时,反应液过滤,滤饼抽干。滤饼及釜壁固体用水 (30.0升)溶解,内温降至5~10℃,用乙酸乙酯(10.0升*2)萃取两次,收集水相。水相分两次处理,15.0升水加入反应釜中,内温降至5~10℃,滴加质量分数为10%碳酸钾水溶液至PH=9,反应液由***变为无色,固体析出,搅拌30分钟后过滤,如果固体太细,可加入乙酸乙酯(1.50升)搅拌30分钟后过滤,滤饼抽干后转入反应釜,加入异丙醇搅拌2.0小时。混合物过滤,滤饼于45℃下真空烘干至恒重,得到式(I)化合物。 1H NMR(400MHz,DMSO-d 6)δ8.50-8.39(m,1H),8.20-8.09(m,1H),7.96-7.82(m,2H),7.32(dd,J=8.7,2.1Hz,1H),4.58-4.34(m,3H),4.14-4.02(m,1H),3.79-3.54(m,2H),3.06-2.88(m,1H),2.48-2.41(m,3H),2.24-2.13(m,3H),2.09-1.95(m,3H),1.93-1.85(m,1H),1.79-1.54(m,8H),1.12-1.01(m,6H),0.99-0.86(m,1H)。
第9步:
将乙醇(14.0升)加入到反应釜中,开启搅拌,将式(I)化合物(1.74千克,3.41摩尔,1.0当量)加入反应釜中,固体不能完全溶解,得到悬浊液。设置循环浴温度90℃,釜内乙醇开始回流,固体逐渐溶清,保温15分钟后,趁热过滤,滤液转入反应釜中逐渐降温至室温,大量固体析出。反应液过滤,滤饼于35~45℃下真空烘干至恒重,得到式(I)化合物的结晶A。 1H NMR(400MHz,DMSO-d 6)δ8.47-8.39(m,1H),8.19-8.08(m,1H),7.97-7.67(m,2H),7.38-7.20(m,1H),4.59-4.29(m,3H),4.17-4.00(m,1H),3.63(td,J=7.6,3.8Hz,2H),2.97(q,J=6.8Hz,1H),2.47-2.39(m,3H),2.24-2.14(m,3H),2.11-1.96(m,2H),1.94-1.81(m,1H),1.78-1.49(m,8H),1.25-0.87(m,8H)。
实施例2:式(I)化合物结晶A的制备
分别称取约50mg的式(I)化合物各自加入1.5mL液相小瓶中,分别加入适量的溶剂或溶剂混合物(见表2),超声混匀或溶解。将悬浊液样品置于恒温振荡仪上(40℃)搅拌(避光)60小时。然后将样品液进行快速离心,离心后的固体置于35℃真空干燥箱中干燥过夜,得到的干燥样品进行XRPD检测。
表2:式(I)化合物结晶A的制备
编号 溶剂或溶剂混合物 体积 状态 结晶
1 甲醇 1mL 悬浊液 结晶A
2 乙醇 1mL 悬浊液 结晶A
3 乙酸乙酯 1mL 悬浊液 结晶A
4 乙腈 1mL 悬浊液 结晶A
5 丙酮 1mL 悬浊液 结晶A
6 四氢呋喃 1mL 悬浊液 结晶A
7 1mL 悬浊液 结晶A
8 甲醇:水=3:1(V/V) 1mL 悬浊液 结晶A
9 乙醇:水=1:1(V/V) 1mL 悬浊液 结晶A
10 丙酮:水=1:1(V/V) 1mL 悬浊液 结晶A
实验例1:式(I)化合物结晶A的吸湿性研究
实验条件:
仪器型号:SMSDVS Advantage动态气体吸附仪(DVS)。
测试条件:取式(I)化合物结晶A(10~15mg)置于DVS样品盘内进行测试。
DVS参数如下:
温度:25℃
平衡:dm/dt=0.01%/min(最短:10min,最长:180min)
干燥:0%RH下干燥120min
RH(%)测量梯度:10%
RH(%)测量梯度范围:0%~90%~0%
吸湿性评价标准见表3。
表3:吸湿性评价标准
吸湿性分类 吸湿增重*
潮解 吸收足量水分形成液体
极具引湿性 引湿增重不小于15%
有引湿性 引湿增重小于15%但不小于2%
略有引湿性 引湿增重小于2%但不小于0.2%
无或几乎无引湿性 引湿增重小于0.2%
注:*在25℃/80%RH下的吸湿增重。
实验结果:式(I)化合物结晶A在25℃、80%RH下的吸湿增重为0.72%。
实验结论:式(I)化合物结晶A属于略有引湿性,易于保存,不宜发生潮解、变形、发霉等现象,有很高的成药价值。
实验例2:式(I)化合物结晶A的稳定性试验
实验方法:
影响因素实验:称取式(I)化合物结晶A 12份,每份1.50g,每个条件下考察3份。每份样品放入敞口的称量瓶中,高温条件放入不同条件的保干器,然后放入相应的稳定箱中考察;高湿条件放入相应条件的稳定箱中考察。
光稳定性实验:称取式(I)化合物结晶A 4份,每份1.50g,2份为光照样品(一份作为5天光照样品,另一份为10天光照样品),另两份为对照样品(一份作为5天对照样品,另一份为10天对照样品), 光照样品放入干净的称量瓶中,平铺成单层,不被任何东西覆盖,盖上透明的盖子,然后放入光照箱中进行光照。对照样品的包装方式与光照样品一致,但在称量瓶外面覆盖铝膜。
长期加速实验:称取式(I)化合物结晶A 22份,每份1.50g。每份样品分别装入双层LDPE袋中,每层LDPE袋分别用扎扣密封,再放入铝箔袋中热封。在考察时间点,将相应的供试样品取出,用瓶盖盖好,0天的样品从冰箱中取出,待样品恢复至室温后进行分析。大约10mg供试品用于XRPD检测。考察化合物在以下条件放置并在不同的时间点取样检测性状,XRPD,含量和有关物质。研究条件和检测项目见表4和表5。
表4:影响因素稳定性及光照稳定性试验中结晶A及有关物质含量的检测结果
试验条件 结晶A含量(%) 杂质含量(%)
0天 98.2 1.19
60℃-5天 99 1.16
60℃-10天 98.2 1.14
60℃-30天 99.1 1.19
25℃-92.5%RH-5天 99.1 1.16
25℃-92.5%RH-10天 98.8 1.13
25℃-92.5%RH-30天 99.2 1.19
光照5天 97.8 1.12
光照10天 99.3 1.13
表5:加速稳定性试验中结晶A及有关物质含量的检测结果
试验条件 结晶A含量(%) 杂质含量(%)
0天 98.2 1.19
40℃-75%RH-1月 99.3 1.2
40℃-75%RH-2月 98.1 1.19
40℃-75%RH-3月 97.8 1.13
40℃-75%RH-6月 98.5 1.12
25℃-60%RH-3月 98 1.16
25℃-60%RH-6月 99.5 1.12
25℃-60%RH-9月 98.2 1.22
25℃-60%RH-12月 100.2 1.09
实验结论:
XRPD检测结果显示式(I)化合物的结晶A与0天一致,在各种测试条件下式(I)化合物结晶A均稳定。
实验例3:cIAP1 BIR3和XIAP BIR3结合实验
实验材料:
测试缓冲体系(cIAP1 BIR3或XIAP BIR3的缓冲液):0.1mol/L磷酸钾,pH 7.5,0.1%牛血清白蛋白,0.005%曲拉通X-100和1%二甲基亚砜。
探针:ARPFAQ-K(5-FAM)-NH 2
靶标:
cIAP1-BIR3-his:RBC(Cat#APT-11-370),人类cIAP1的BIR3域(涵盖氨基酸258至363;cIAP1 BIR3)作为GST-融合蛋白自大肠杆菌(E.coli)表达及纯化。
XIAP-BIR3-his:RBC(Cat#APT-11-374),XIAP的BIR3域(涵盖氨基酸255至356;XIAP BIR3)作为GST-融合蛋白自大肠杆菌(E.coli)表达及纯化。
反应条件:5nM ARPFAQ-K(5-FAM)-NH 2,20nM cIAP1 BIR3和30nM XIAP BIR3。
实验步骤:
首先制备cIAP1 BIR3或XIAP BIR3的新鲜缓冲液,加入2倍cIAP1 BIR3或XIAP BIR3溶液,再通过声学技术将100%DMSO溶解的待测化合物加入到cIAP1 BIR3或XIAP BIR3的缓冲溶液中,然后再加入2倍探针,室温下黑暗中混合孵化60分钟,测量荧光偏振和计算mP值,最后得到IC 50值。
实验结果:如表6所示。
实验结论:
本申请式(I)化合物表现出良好的cIAP1 BIR3结合活性,且对cIAP1和XIAP的表现出较好的选择性。
实验例4:体外细胞活性测试
实验材料:
RPMI 1640培养基(Invitrogen-22400089),胎牛血清(Invitrogen-10099141),Trypsin,0.05%EDTA·4Na(Invitrogen-25300062),发光法细胞活力检测试剂盒(Promega-G7573),杜氏磷酸盐缓冲液(HyClone-SH30028.01B),384孔板(Corning-6007680)。Envision多标记分析仪。
实验方法:
1.向384微孔板的孔中加30μL MDA-MB-231细胞悬液,其中包含250个MDA-MB-231细胞。
2、加入20μL受试化合物(受试化合物做高浓度为10μM,对受试化合物进行5倍梯度稀释,将每个化合物稀释10个浓度梯度),然后将细胞板放回到二氧化碳培养箱中培养7天。
3、将细胞板在室温下平放30分钟。
4、向细胞板中加入每孔20μL的Promega CellTiter-Glo试剂。
5、10分钟后采用Envision多标记分析仪读数。
实验结果:见表6。
实验结论:
本申请式(I)化合物具有较好的MDA-MB-231细胞抗增殖活性。
表6
受试化合物 cIAP1 BIR3IC 50(nM) XIAP BIR3IC 50(nM) MDA-MB-231 Cell IC 50(nM)
式(I)化合物 5.0 29.9 54.8
实验例5:体内药效研究1
在皮下植入MDA-MB-231三阴性乳腺癌患者来源的基于人源肿瘤细胞系的异种移植(CDX)BALB/c裸小鼠上进行体内药效实验
实验操作:
BALB/c裸鼠,雌性,6-8周,体重约18-22克,将小鼠保持在一个特殊的无病原体的环境中,且在单个通风笼中(3只小鼠每笼)。所有的笼子,铺垫和水在使用前进行消毒。所有的动物都可以自由获取标准认证的商业实验室饮食。共有48只购于上海西普尔必凯实验动物有限公司(Shanghai BK Laboratory Animal Co.,LTD)的小鼠用于研究。每只小鼠在右胁腹皮下植入肿瘤细胞(10×10 6在0.2毫升磷酸盐缓冲液中),用于肿瘤的生长。当平均肿瘤体积达到约147立方毫米时开始给药。将试验化合物每日口服给药,给药剂量为30毫克/公斤。肿瘤体积每3天用二维卡尺测量,体积以立方毫米计量,通过以下公式计算:V=0.5a×b 2,其中a和b分别是肿瘤的长径和短径。抗肿瘤药效是通过用化合物处理过的动物的平均肿瘤增加体积除以未处理过动物的平均肿瘤增加体积来确定。
实验结果:见表7。
实验结论:
在MDA-MB-231三阴性乳腺癌CDX体内药效模型中,本申请式(I)化合物展现较好的药效。
表7
Figure PCTCN2020089437-appb-000005
实验例6:体内药效研究2
在皮下植入MDA-MB-231三阴性乳腺癌患者来源的基于人源肿瘤细胞系的异种移植(CDX)BALB/c裸小鼠上进行体内药效实验
实验操作:
BALB/c裸鼠,雌性,6-8周,体重约18-22克,将小鼠保持在一个特殊的无病原体的环境中,且在单个通风笼中(3只小鼠每笼)。所有的笼子,铺垫和水在使用前进行消毒。所有的动物都可以自由获取标准认证的商业实验室饮食。共有48只购于上海西普尔必凯实验动物有限公司(Shanghai BK Laboratory Animal Co.,LTD)的小鼠用于研究。每只小鼠在右胁腹皮下植入肿瘤细胞(10×10 6在0.2毫升磷酸盐缓冲液中),用于肿瘤的生长。当平均肿瘤体积达到约110立方毫米时开始给药。将试验化合物每日口服给药,给药剂量为30毫克/公斤。肿瘤体积每3天用二维卡尺测量,体积以立方毫米计量,通过以下公式计算:V=0.5a×b 2,其中a和b分别是肿瘤的长径和短径。抗肿瘤药效是通过用化合物处理过的动物的平均肿瘤增加体积除以未处理过动物的平均肿瘤增加体积来确定。
实验结果:见表8。
实验结论:
在MDA-MB-231三阴性乳腺癌CDX体内药效模型中,本申请式(I)化合物展现较好的药效。
表8
Figure PCTCN2020089437-appb-000006

Claims (11)

  1. 式(I)化合物的结晶
    Figure PCTCN2020089437-appb-100001
  2. 权利要求1的结晶,所述结晶为式(I)化合物的结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.1°±0.200°、16.1°±0.200°、18.5°±0.200°、20.2°±0.200°、21.3°±0.200°和23.0°±0.200°。
  3. 权利要求2所述式(I)化合物的结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.1°±0.200°、16.1°±0.200°、18.5°±0.200°、18.8°±0.200°、19.2°±0.200°、19.8°±0.200°、20.2°±0.200°、21.3°±0.200°、23.0°±0.200°、26.6°±0.200°和27.4°±0.200°。
  4. 权利要求3所述式(I)化合物的结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0°±0.200°、8.7°±0.200°、12.1°±0.200°、13.2°±0.200°、13.9°±0.200°、16.1°±0.200°、16.7°±0.200°、18.5°±0.200°、18.8°±0.200°、19.2°±0.200°、19.8°±0.200°、20.2°±0.200°、21.0°±0.200°、21.3°±0.200°、23.0°±0.200°、24.3°±0.200°、25.3°±0.200°、26.6°±0.200°和27.4°±0.200°。
  5. 权利要求4所述式(I)化合物的结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0°±0.200°、8.7°±0.200°、12.1°±0.200°、13.2°±0.200°、13.9°±0.200°、16.1°±0.200°、16.5°±0.200°、16.7°±0.200°、18.5°±0.200°、18.8°±0.200°、19.2°±0.200°、19.8°±0.200°、20.2°±0.200°、21.0°±0.200°、21.3°±0.200°、23.0°±0.200°、23.2°±0.200°、24.3°±0.200°、25.3°±0.200°、26.6°±0.200°、26.9°±0.200°、27.4°±0.200°和29.4°±0.200°。
  6. 权利要求5所述式(I)化合物的结晶A,其使用Cu Kα辐射的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:7.0°±0.200°、8.7°±0.200°、9.7°±0.200°、10.6°±0.200°、11.4°±0.200°、12.1°±0.200°、13.2°±0.200°、13.9°±0.200°、16.1°±0.200°、16.5°±0.200°、16.7°±0.200°、17.5°±0.200°、18.5°±0.200°、18.8°±0.200°、19.2°±0.200°、19.5°±0.200°、19.8°±0.200°、20.2°±0.200°、21.0°±0.200°、21.3°±0.200°、22.5°±0.200°、23.0°±0.200°、23.2°±0.200°、23.8°±0.200°、24.3°±0.200°、24.6°±0.200°、25.3°±0.200°、25.8°±0.200°、26.6°±0.200°、26.9°±0.200°、27.4°±0.200°、28.1°±0.200°、29.1°±0.200°、29.4°±0.200°、30.1°±0.200°、30.6°±0.200°、30.8°±0.200°、31.3°±0.200°、31.8°±0.200°、32.2°±0.200°、32.8°±0.200°、 33.7°±0.200°、34.0°±0.200°、34.8°±0.200°、35.5°±0.200°、36.6°±0.200°、37.6°±0.200°、38.6°±0.200°和39.6°±0.200°。
  7. 权利要求2-6任一项所述的式(I)化合物的结晶A,其差示扫描量热测量图中吸收峰的峰值在202.5℃处。
  8. 权利要求2-6任一项所述的式(I)化合物的结晶A,其差示扫描量热测量图中吸收峰的起始位置在约200.8℃处。
  9. 结晶组合物,其中权利要求1所述的式(I)化合物结晶占结晶组合物重量的50%以上,较好是75%以上,更好是90%以上,最好是95%以上。
  10. 药物组合物,其包含权利要求1所述的式(I)化合物结晶或权利要求9所述的结晶组合物。
  11. 权利要求1所述的式(I)化合物结晶、权利要求9所述的结晶组合物,或权利要求10所述的药物组合物在制备治疗受益于cIAP1抑制的癌症的药物中的用途。
PCT/CN2020/089437 2019-05-10 2020-05-09 一种用作iap抑制剂的smac模拟物的结晶及其制备方法 WO2020228642A1 (zh)

Priority Applications (11)

Application Number Priority Date Filing Date Title
BR112021022509A BR112021022509A2 (pt) 2019-05-10 2020-05-09 Forma cristalina, forma cristalina a de fórmula (i), composição cristalina, composição farmacêutica e uso destas
KR1020217040412A KR20220006611A (ko) 2019-05-10 2020-05-09 Iap 억제제로서의 smac 모방물의 결정 및 그 제조방법
DK20805780.2T DK3967702T3 (da) 2019-05-10 2020-05-09 Krystallisering af smac-mimic anvendt som iap-inhibitor og fremgangsmåde til fremstilling deraf
CN202080030342.1A CN113748119B (zh) 2019-05-10 2020-05-09 一种用作iap抑制剂的smac模拟物的结晶及其制备方法
MX2021013729A MX2021013729A (es) 2019-05-10 2020-05-09 Cristalina de imitador smac utilizado como inhibidor de iap y metodo de preparacion del mismo.
CA3138411A CA3138411A1 (en) 2019-05-10 2020-05-09 Crystallization of smac mimic used as iap inhibitor and preparation method thereof
JP2021566285A JP2022531794A (ja) 2019-05-10 2020-05-09 Iap阻害剤としてのsmac模倣物の結晶及びその製造方法
AU2020274768A AU2020274768A1 (en) 2019-05-10 2020-05-09 Crystalline form of smac mimic used as iap inhibitor and preparation method thereof
US17/594,737 US20220177453A1 (en) 2019-05-10 2020-05-09 Crystallization of smac mimic used as iap inhibitor and preparation method thereof
EP20805780.2A EP3967702B1 (en) 2019-05-10 2020-05-09 Crystallization of smac mimic used as iap inhibitor and preparation method thereof
FIEP20805780.2T FI3967702T3 (fi) 2019-05-10 2020-05-09 IAP-inhibiittorina käytettävän SMAC-mimeetin kiteyttäminen ja menetelmä sen valmistamiseksi

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910389970 2019-05-10
CN201910389970.8 2019-05-10

Publications (2)

Publication Number Publication Date
WO2020228642A1 WO2020228642A1 (zh) 2020-11-19
WO2020228642A9 true WO2020228642A9 (zh) 2021-10-21

Family

ID=73290153

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/089437 WO2020228642A1 (zh) 2019-05-10 2020-05-09 一种用作iap抑制剂的smac模拟物的结晶及其制备方法

Country Status (13)

Country Link
US (1) US20220177453A1 (zh)
EP (1) EP3967702B1 (zh)
JP (1) JP2022531794A (zh)
KR (1) KR20220006611A (zh)
CN (1) CN113748119B (zh)
AU (1) AU2020274768A1 (zh)
BR (1) BR112021022509A2 (zh)
CA (1) CA3138411A1 (zh)
DK (1) DK3967702T3 (zh)
FI (1) FI3967702T3 (zh)
MX (1) MX2021013729A (zh)
PT (1) PT3967702T (zh)
WO (1) WO2020228642A1 (zh)

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100926203B1 (ko) * 2004-07-02 2009-11-09 제넨테크, 인크. Iap의 억제제
WO2006069063A1 (en) * 2004-12-20 2006-06-29 Genentech, Inc. Pyrrolidine inhibitors of iap
EP2049524A2 (en) * 2006-07-24 2009-04-22 Tetralogic Pharmaceuticals Corporation Iap inhibitors
CN101595121A (zh) * 2006-10-12 2009-12-02 诺瓦提斯公司 作为iap抑制剂的吡咯烷衍生物
JP2010513561A (ja) * 2006-12-19 2010-04-30 ジェネンテック, インコーポレイテッド Iapのイミダゾピリジンインヒビター
CN101951766A (zh) * 2008-01-24 2011-01-19 泰特拉洛吉克药业公司 凋亡抑制蛋白抑制剂
SG11202004377XA (en) * 2017-11-13 2020-06-29 Chia Tai Tianqing Pharmaceutical Group Co Ltd Smac mimetics used as iap inhibitors and use thereof

Also Published As

Publication number Publication date
AU2020274768A1 (en) 2022-01-06
JP2022531794A (ja) 2022-07-11
CN113748119A (zh) 2021-12-03
MX2021013729A (es) 2021-12-10
FI3967702T3 (fi) 2024-04-16
DK3967702T3 (da) 2024-04-15
CA3138411A1 (en) 2020-11-19
US20220177453A1 (en) 2022-06-09
BR112021022509A2 (pt) 2021-12-28
EP3967702B1 (en) 2024-03-20
WO2020228642A1 (zh) 2020-11-19
EP3967702A1 (en) 2022-03-16
PT3967702T (pt) 2024-04-09
KR20220006611A (ko) 2022-01-17
EP3967702A4 (en) 2023-01-11
CN113748119B (zh) 2024-04-02

Similar Documents

Publication Publication Date Title
ES2932867T3 (es) Degradadores selectivos de los receptores de estrógenos
CN108368110A (zh) 用于激酶调节的化合物和方法以及其适应症
CN109970745A (zh) 取代的吡咯并三嗪类化合物及其药物组合物及其用途
TWI718105B (zh) 6-(6,7-二甲氧基喹唑啉-4-基氧基)-n,2-二甲基苯並呋喃-3-甲醯胺晶型
JP2008519049A (ja) 増殖性疾患の治療のためのsrcキナーゼインヒビターおよびbcr−ablインヒビターの組み合わせ医薬
BR112018001688B1 (pt) Uso de um composto
CN113336768B (zh) 一种多靶点酪氨酸激酶抑制剂
CN111902405B (zh) 靶向cdk4/6激酶抑制剂的晶型
WO2021121146A1 (zh) 氨基嘧啶类化合物甲磺酸盐的晶型a及其制备方法和应用
WO2020228642A9 (zh) 一种用作iap抑制剂的smac模拟物的结晶及其制备方法
JP5860125B2 (ja) 新規な多形形態の6−(1−メチル−1h−ピラゾール−4−イル)−2−{3−[5−(2−モルホリン−4−イル−エトキシ)−ピリミジン−2−イル]−ベンジル}−2h−ピリダジン−3−オンジヒドロゲンホスファートおよびその製造方法
CN107778336B (zh) 吡喃葡萄糖基衍生物的结晶形式
AU2022350669A1 (en) Egfr inhibitor polymorph forms
EP4116300A1 (en) Crystal of tricyclic compound acting on crbn protein and preparation method therefor
CN115448874A (zh) 固体形式的周期蛋白依赖性激酶9抑制剂及其用途
CN101239990B (zh) N-(2,3,4,5,6-五羟基己基)-l-氨基酸合铂、其制备方法及应用
CN107663173A (zh) 恩杂鲁胺及其制备方法和用途
CN113825506A (zh) 结合细胞周期蛋白依赖性激酶抑制剂1b(p27kip1)的小分子
WO2018099451A1 (zh) 化合物的晶型
WO2024055994A1 (zh) 萘并呋喃取代的戊二酰亚胺类化合物的晶型、制备方法及其应用
CN115246863B (zh) 一种川芎嗪四价铂类化合物、其制备方法及其用途
WO2022247772A1 (zh) 一种含氧杂环化合物的晶型、其制备方法及应用
WO2023006057A1 (zh) 氨基吡唑并嘧啶化合物的晶体
WO2021143819A1 (zh) 多环类间变性淋巴瘤激酶抑制剂的晶型
WO2019238088A1 (zh) 吡啶并吡啶酮衍生物的盐型及晶型

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: 20805780

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3138411

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021566285

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021022509

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20217040412

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020805780

Country of ref document: EP

Effective date: 20211210

ENP Entry into the national phase

Ref document number: 112021022509

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20211109

ENP Entry into the national phase

Ref document number: 2020274768

Country of ref document: AU

Date of ref document: 20200509

Kind code of ref document: A