WO2020169054A1 - CRYSTALLINE FORM OF (E)-α,β-UNSATURED AMIDE COMPOUND, PREPARATION METHOD THEREFOR, AND USE THEREOF - Google Patents

CRYSTALLINE FORM OF (E)-α,β-UNSATURED AMIDE COMPOUND, PREPARATION METHOD THEREFOR, AND USE THEREOF Download PDF

Info

Publication number
WO2020169054A1
WO2020169054A1 PCT/CN2020/075918 CN2020075918W WO2020169054A1 WO 2020169054 A1 WO2020169054 A1 WO 2020169054A1 CN 2020075918 W CN2020075918 W CN 2020075918W WO 2020169054 A1 WO2020169054 A1 WO 2020169054A1
Authority
WO
WIPO (PCT)
Prior art keywords
crystal form
compound
formula
volume ratio
ray powder
Prior art date
Application number
PCT/CN2020/075918
Other languages
French (fr)
Chinese (zh)
Inventor
李娜
陈连蔚
吴忠伟
陈磊
杨志清
张鑫龙
龚永祥
Original Assignee
浙江海正药业股份有限公司
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 浙江海正药业股份有限公司 filed Critical 浙江海正药业股份有限公司
Publication of WO2020169054A1 publication Critical patent/WO2020169054A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/62Compounds containing any of the groups, X being a hetero atom, Y being any atom, e.g. N-acylcarbamates
    • C07C271/64Y being a hydrogen or a carbon atom, e.g. benzoylcarbamates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/325Carbamic acids; Thiocarbamic acids; Anhydrides or salts thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C225/00Compounds containing amino groups and doubly—bound oxygen atoms bound to the same carbon skeleton, at least one of the doubly—bound oxygen atoms not being part of a —CHO group, e.g. amino ketones
    • C07C225/02Compounds containing amino groups and doubly—bound oxygen atoms bound to the same carbon skeleton, at least one of the doubly—bound oxygen atoms not being part of a —CHO group, e.g. amino ketones having amino groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C225/14Compounds containing amino groups and doubly—bound oxygen atoms bound to the same carbon skeleton, at least one of the doubly—bound oxygen atoms not being part of a —CHO group, e.g. amino ketones having amino groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being unsaturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/16Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms
    • C07C233/17Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/20Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom having the carbon atom of the carboxamide group bound to a carbon atom of an acyclic unsaturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/28Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and unsaturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C269/00Preparation of derivatives of carbamic acid, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C269/08Separation; Purification; Stabilisation; Use of additives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/12Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the invention relates to the field of chemical pharmacy. More specifically, the present invention relates to polymorphs of (E)- ⁇ , ⁇ -unsaturated amide compounds and methods for preparing the crystal forms, pharmaceutical compositions containing them, and their pharmaceutical uses.
  • the compound of formula I is a representative of this class of new (E)- ⁇ , ⁇ -unsaturated amide compounds, which have been demonstrated in animal models of stroke, Alzheimer’s disease, Parkinson’s disease and multiple sclerosis It has very good efficacy and has a bright future for treating multiple diseases with one medicine.
  • the compound of formula I is a polymorphic compound.
  • different crystal forms have different physical and chemical properties, including melting point, chemical stability, apparent solubility, dissolution rate, optical and mechanical properties, etc. These physical and chemical properties directly determine the efficacy of a particular crystal form, and directly affect the quality of raw materials and preparations.
  • crystal form I (hereinafter referred to as "crystal form I") of the compound of formula I with good chemical and physical stability.
  • the crystal form I has excellent properties in terms of chemical stability and processing (filtration, drying) adaptability.
  • the X-ray powder diffraction pattern of the crystal form I of the present invention has characteristic peaks at the following diffraction angles 2 ⁇ : 8.82 ⁇ 0.2°, 14.56 ⁇ 0.2°, 16.82 ⁇ 0.2°, 17.72 ⁇ 0.2°, 20.22 ⁇ 0.2°, 22.50 ⁇ 0.2°, 26.24 ⁇ 0.2°, 29.40 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the crystal form I has characteristic peaks at the following diffraction angles 2 ⁇ : 10.06 ⁇ 0.2°, 24.22 ⁇ 0.2°, 25.00 ⁇ 0.2°, 26.76 ⁇ 0.2°, 28.16 ⁇ 0.2°, 30.56 ⁇ 0.2°, 31.14 ⁇ 0.2°, 33.74 ⁇ 0.2°, 38.66 ⁇ 0.2°, 40.20 ⁇ 0.2°, 44.84 ⁇ 0.2°, 46.04 ⁇ 0.2°.
  • the X-ray powder diffraction spectrum of crystal form I of the present invention has 2 ⁇ , d and relative intensity data as shown in Table 1 below:
  • the X-ray powder diffraction spectrum of the crystal form I of the present invention is basically as shown in FIG. 1.
  • the peak value of the differential scanning calorimetry (DSC) spectrum of the crystalline form I of the present invention is 192.7°C.
  • crystal form I of the present invention has a DSC spectrum as shown in FIG. 2.
  • the crystal form I of the present invention has a TGA pattern as shown in FIG. 3.
  • Another object of the present invention is to provide a method for preparing the crystal form I, and the method is selected from any one of the following methods:
  • Method (1) which includes the following steps:
  • Method (2) which includes the following steps:
  • the compound of formula I is dissolved in any solvent of alcohol, ketone, ester, acetonitrile, dioxane, dimethylacetamide, dimethylformamide, and dimethylsulfoxide, and the temperature is lowered to crystallize;
  • the alcohol is a C2-C4 alcohol;
  • the ketone is a C3-C6 ketone;
  • the ester is a C3-C6 ester;
  • the dissolution temperature is 40-70°C;
  • the mass-volume ratio of the compound of formula I to the solvent is (mg /ml) 10: 0.4 ⁇ 3; the temperature of said crystallization is 0 ⁇ 20°C;
  • the compound of formula I is dissolved in a mixed solvent of any organic solvent among ethanol, isopropanol, dioxane, acetone, and acetonitrile and water, and the temperature is lowered to crystallize; the dissolution temperature is 40-70°C;
  • the volume ratio of the organic solvent to water is 1:0.1-9;
  • the mass-volume ratio of the compound of formula I and the mixed solvent is (mg/ml) 10:1-1.5;
  • the crystallization temperature is 0-5°C ;
  • crystal form II of the compound of formula I (hereinafter referred to as "crystal form II")
  • the X-ray powder diffraction (XRD) pattern of the crystal form II has characteristic peaks at the following diffraction angle 2 ⁇ : 7.34 ⁇ 0.2°, 14.72 ⁇ 0.2°, 16.98 ⁇ 0.2°, 20.30 ⁇ 0.2°, 22.16 ⁇ 0.2°, 25.84 ⁇ 0.2°, 38.44 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the crystal form II has characteristic peaks at the following diffraction angles 2 ⁇ : 9.20 ⁇ 0.2°, 13.90 ⁇ 0.2°, 18.46 ⁇ 0.2°, 23.74 ⁇ 0.2°, 29.72 ⁇ 0.2°, 34.42 ⁇ 0.2°.
  • the X-ray powder diffraction spectrum of the crystal form II of the present invention has the 2 ⁇ data shown in Table 2 below:
  • the X-ray powder diffraction spectrum of the crystal form II of the present invention is basically shown in FIG. 4.
  • the peak value of the differential scanning calorimetry (DSC) spectrum of the crystalline form II of the present invention is 188.3°C.
  • crystal form II of the present invention has a DSC spectrum as shown in FIG. 5.
  • the crystal form II of the present invention has a TGA pattern as shown in FIG. 6.
  • Another object of the present invention is to provide a method for preparing the crystal form II, the method is selected from any one of the following methods:
  • Method (1) which includes the following steps:
  • Method (2) which includes the following steps:
  • the unit of the mass-volume ratio of the compound of formula I to the solvent may be mg/ml, g/L, etc., depending on the specific scale of operation.
  • Another object of the present invention is to provide a pharmaceutical composition, which comprises an effective dose of crystal form I or crystal form II; the pharmaceutical composition also comprises one or more pharmaceutically acceptable carriers.
  • the present invention also relates to the application of crystal form I or crystal form II, or their pharmaceutical composition in the preparation of a medicine for treating diseases related to Nrf2 activation, wherein the diseases related to Nrf2 activation are stroke and neurodegeneration Sexual diseases, diabetes, diabetic nephropathy, coronary heart disease, atherosclerosis or non-alcoholic fatty liver.
  • the present invention also relates to the use of crystal form I or crystal form II, or their pharmaceutical composition in the preparation of a medicament for the treatment of stroke.
  • the present invention also relates to the application of the crystal form I or the crystal form II, or their pharmaceutical composition in the preparation of a medicament for the treatment of neurodegenerative diseases.
  • the neurodegenerative disease is selected from multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), muscle atrophy Lateral sclerosis (ALS), Friedreich’s ataxia (FRDA), spinal muscular atrophy (SMA), optic neuromyelitis (NMO) and spinocerebellar ataxia (SCA).
  • MS multiple sclerosis
  • AD Alzheimer's disease
  • PD Parkinson's disease
  • HD Huntington's disease
  • ALS muscle atrophy Lateral sclerosis
  • FRDA muscle atrophy Lateral sclerosis
  • SMA spinal muscular atrophy
  • NMO optic neuromyelitis
  • SCA spinocerebellar ataxia
  • the present invention also relates to the use of crystal form I or crystal form II, or their pharmaceutical composition in the preparation of a medicament for the treatment of diseases related to immune regulation, wherein the diseases related to immune regulation are preferably From psoriasis, rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, transplant rejection and inflammatory diseases.
  • a method for treating diseases associated with Nrf2 activation comprising administering an effective dose of crystal form I or crystal form II, or a pharmaceutical composition thereof, to a patient in need.
  • a method for treating stroke comprising administering an effective dose of crystal form I or crystal form II, or a pharmaceutical composition thereof to a patient in need.
  • a method of treating neurodegenerative diseases comprising administering an effective dose of crystal form I or crystal form II, or a pharmaceutical composition thereof to a patient in need.
  • a method for treating diseases related to immune regulation comprising administering an effective dose of crystal form I or crystal form II, or a pharmaceutical composition thereof, to a patient in need.
  • the inventors After a lot of research, the inventors have discovered the crystal form I and crystal form II of the compound of formula I, which have good solubility, simple crystallization process, easy operation, low pollution, can realize industrial production, and have crystal form I or crystal form II.
  • the compound of formula I has the advantages of high product purity, excellent physical and chemical properties, good chemical stability, and reproducible processing (filtering, drying,).
  • Fig. 1 is an X-ray powder diffraction pattern of the crystal form I of the compound of formula I obtained in Example 1.
  • Example 2 is a DSC chart of the crystal form I of the compound of formula I obtained in Example 1.
  • Example 4 is an X-ray powder diffraction pattern of the crystal form II of the compound of formula I obtained in Example 5.
  • Fig. 7 shows that the compound of formula I induces Nrf2 to enter the nucleus in HT22 cells (4h).
  • Figure 8 shows that the compound of formula I up-regulates the expression of HO-1 protein in HT22 cells (4h).
  • Figure 9 shows the inhibitory effect of the compound of formula I on mouse EAE (***p ⁇ 0.001, **p ⁇ 0.01, *p ⁇ 0.05, compared with the solvent group (one-way ANOVA/Dunnett)).
  • Figure 10 is the effect of Morris water maze experiment formula I compound on the escape latency of AD rat model (***p ⁇ 0.001, **p ⁇ 0.01, *p ⁇ 0.05, compared with the model group (one-way ANOVA/Dunnett) )).
  • Figure 11 is the effect of Morris water maze experiment formula I compound on the number of times the AD rat model crosses the target platform (***p ⁇ 0.001, **p ⁇ 0.01, *p ⁇ 0.05, compared with the model group (one-way ANOVA) /Dunnett)).
  • Figure 12 shows the effect of the compound of formula I on the latency in the 6-OHDA-induced PD rat rotarod experiment (***p ⁇ 0.001, **p ⁇ 0.01, *p ⁇ 0.05, compared with the model group (one-way) ANOVA/Dunnett)).
  • Figure 13 is the effect of the compound of formula I on the climbing time in the 6-OHDA-induced PD rat climbing experiment (***p ⁇ 0.001, **p ⁇ 0.01, *p ⁇ 0.05, compared with the model group (one -way ANOVA/Dunnett)).
  • Figure 14 is the effect of formula I compound on the rotation speed of PD rats induced by apomorphine (***p ⁇ 0.001, **p ⁇ 0.01, *p ⁇ 0.05, compared with the model group (one-way ANOVA/Dunnett) )).
  • Figure 15 is the effect of the compound of formula I on the neurological score of acute cerebral artery ischemia-reperfusion injury in rats (***p ⁇ 0.001, **p ⁇ 0.01, *p ⁇ 0.05, compared with the model group (one-way) ANOVA/Dunnett)).
  • Figure 16 shows the effect of compound of formula I on the weight ratio of cerebral infarction area/whole brain in rats with acute cerebral artery ischemia reperfusion injury (***p ⁇ 0.001, **p ⁇ 0.01, *p ⁇ 0.05, similar to the model group Than (one-way ANOVA/Dunnett)).
  • Figure 17 shows the sensitization rate of the compound of formula I to the skin of guinea pigs.
  • the raw materials of the compound of formula I used in the method of the present invention are derived from the following synthetic methods:
  • Step 1 Add 250g (1.92mol) (E)-4-methoxy-4-oxo-but-2-enoic acid, 225g (2.31mol) N,O-dimethylhydroxylamine into the reaction flask Hydrochloride and 2500 ml of dichloromethane, reduce the temperature to 0 ⁇ 10°C, add 550 g (2.88 mol) of 1-ethyl-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) , Warm up to room temperature, react for 1 hour, the reaction is complete. Wash with 1500 ml of water, collect the organic phase, decolorize with activated carbon, filter, and concentrate the filtrate to dryness.
  • EDCI 1-ethyl-(3-dimethylaminopropyl) carbodiimide hydrochloride
  • Step 2 Add 1250 ml of ammonia to the concentrate obtained above, cool to 0-10°C, react for 20 minutes, after the reaction is complete, filter, and recrystallize the solid with ethanol and ethyl acetate to obtain 142.3 g of compound Ia.
  • Step 1 Put 5.0 g (31.6 mmol) (E)-N'-methoxy-N'-methyl-but-2-ene diamide (compound Ia) and 50 ml 1,2-into the reaction flask Dichloroethane, cooled to 0-10°C, added 6 ml (70.9 mmol) of oxalyl chloride, reacted at room temperature for 6 hours, heated to 65°C, reacted for 15 minutes, concentrated to dryness, and set aside.
  • Step 2 Add 30 milliliters of anhydrous methanol to another reaction flask, lower the temperature to 0-10°C, add the concentrated solution obtained in Step 1, and react for 10 minutes to precipitate a solid to obtain 3.56 g of formula I compound.
  • the solvent used in the present invention is not particularly limited, and commercially available conventional solvents can be used.
  • the "stirring" described in the method of the present invention can adopt conventional methods in the art.
  • the stirring method includes magnetic stirring and mechanical stirring, and the stirring speed is 50-300 rpm/min, preferably 100-200 rpm/min.
  • the X-ray powder diffraction instrument and test conditions involved in the present invention are: X-ray diffraction instrument model MiniFlex600Cu target; operation method: scanning speed 20°/min, scanning step width 0.02°.
  • the DSC test conditions involved in the present invention are: DSC detector model: NETZSCH DSC 214 Polyma; operation method: heating rate 10°C/min, temperature range: 25-250°C.
  • the TGA test conditions involved in the present invention are: the model of the TGA detector is: METTLER TOLEDO TGA2; the operation method: the heating rate is 10°C/min, and the temperature range: 25-250°C.
  • HPLC purity test conditions of the compound of formula I involved in the present invention are: chromatographic column: Welch Ultimate XB-C18 250*4.6mm 5 ⁇ m; mobile phase A: 0.01% trifluoroacetic acid-water; mobile phase B: acetonitrile; filtration and degassing ; Detection wavelength: 226nm; Flow rate: 1.0ml/min; Injection volume: 10 ⁇ l; Column temperature: 25°C; Mobile phase conditions are shown in Table 3:
  • Example 5 Take the crystalline form I prepared in Example 1 and the crystalline form II prepared in Example 8, and conduct a stability test at 60° C. for 10 days.
  • the HPLC purity and maximum single impurity content of the compound before and after storage were tested, and the results are shown in Table 4; the crystal form changes of the samples after 10 days of storage were analyzed by X-ray powder diffraction detection, and the results are shown in Table 5.
  • Crystal Form II XRD shows a mixed crystal of crystalline form I and II
  • the compound induces Nrf2 to enter the nucleus and HO-1 expression in the cytoplasm of HT22 cells
  • HT22 cells that proliferate well and are in the logarithmic growth phase, and count after trypsin digestion and dispersion.
  • the cell density is adjusted with RPMI 1640 medium containing 5% fetal bovine serum, and 1000 cells/well are inoculated into a 96-well plate. Incubate in an incubator at 37°C, 5% CO 2 , and 100% relative humidity for 24 hours. Compounds of different concentrations were added to each well, after 24 hours of continuous cultivation, a certain concentration of L-glutamate monosodium salt was added, and after another 24 hours of cultivation, the CellTiter-Glo kit was used to detect cell viability.
  • the experimental results show that in the HT22 cell injury model induced by sodium glutamate, the compound of the present invention has a protective effect on HT22 cells injured by sodium L-glutamate (Table 8).
  • mice Female C57BL/6 mice aged 6-8 weeks were randomly grouped. On day 0, 100 ⁇ L of MOG35-55 (1mg/mL) and Freund's complete adjuvant (2mg/mL) were injected into the hind limbs and back muscles of the mice on day 0. The immune emulsion was injected with pertussis toxin (200ng) into the abdominal cavity 48h later to induce the development of EAE model.
  • the compound improves the learning and memory of Alzheimer's disease (AD) rats injected with A ⁇ into the lateral ventricle
  • mice Male wistar rats, 12 weeks old, were injected with 6-OHDA into the medial forebrain tract to prepare a PD rat model. 21 days after 6-OHDA injection, apomorphine was used to induce rotation to verify the success of the model. Mice were randomly divided into groups and given different doses of compound by gavage. The sham operation group and model group were given double distilled water, L-dopa was used as the control drug, and behavioral testing (including the rotating rod test) was performed after 10 days. , Pole climbing experiment and apomorphine-induced rotation experiment) to evaluate the efficacy of the test compound.
  • the compound of formula I can significantly alleviate the PD-like behavioral disorder in rats induced by 6-OHDA.
  • MCAO acute cerebral artery ischemia-reperfusion injury model
  • the test drug in the administration group and the petroleum jelly in the control group were washed and washed to observe whether there was skin reaction, and the degree of reaction was scored and photographed and recorded, and the sensitization rate of each group was counted. There was no significant reaction in the Control group.
  • DMF caused 90% of the guinea pigs to have obvious skin erythema and edema.
  • the formula I compound group had no obvious skin reaction, only slight edema ( Figure 17). The results show that DMF has extremely strong allergenicity, while the compound of formula I only has extremely weak allergenicity.

Abstract

The present invention relates to crystalline forms I and II of a (E)-α,β-unsatured amide compound, preparation methods therefor, and uses thereof. The crystalline forms all have excellent properties in physical and chemical stability and processing adaptability.

Description

(E)-α,β-不饱和酰胺化合物的晶型及其制备方法和用途(E)-α,β-Unsaturated amide compound crystal form and its preparation method and application 技术领域Technical field
本发明涉及化学制药领域。更具体地说,本发明涉及(E)-α,β-不饱和酰胺化合物的多晶型以及所述晶型的制备方法、含有它们的药物组合物以及它们的制药用途。The invention relates to the field of chemical pharmacy. More specifically, the present invention relates to polymorphs of (E)-α,β-unsaturated amide compounds and methods for preparing the crystal forms, pharmaceutical compositions containing them, and their pharmaceutical uses.
技术背景technical background
本发明申请人之前的专利申请(PCT/CN2018/102824,该申请全文被本申请引用作为参考)报道了一类新的(E)-α,β-不饱和酰胺化合物,这类化合物是Nrf2途径激活剂,能有效地保护神经细胞免受氧化损伤。这类化合物也有一定的免疫调节活性。The applicant’s previous patent application (PCT/CN2018/102824, the entire application is cited as a reference in this application) reports a new class of (E)-α,β-unsaturated amide compounds, which are Nrf2 pathways The activator can effectively protect nerve cells from oxidative damage. Such compounds also have certain immunomodulatory activity.
Figure PCTCN2020075918-appb-000001
Figure PCTCN2020075918-appb-000001
式I化合物是这类新的(E)-α,β-不饱和酰胺化合物中的一个代表,它在脑卒中、阿尔茨海默氏病、帕金森病和多发性硬化症动物模型上都展示出很好的药效,具有一药治多病的光明前景。The compound of formula I is a representative of this class of new (E)-α,β-unsaturated amide compounds, which have been demonstrated in animal models of stroke, Alzheimer’s disease, Parkinson’s disease and multiple sclerosis It has very good efficacy and has a bright future for treating multiple diseases with one medicine.
式I化合物是一个多晶型化合物,而对于多晶型化合物而言,不同的晶型具有不同的物理化学性质,包括熔点、化学稳定性、表观溶解度、溶解速率、光学和机械性质等,而这些物化性能直接决定某特定晶型的药效,并且直接影响到原料药和制剂的质量。The compound of formula I is a polymorphic compound. For a polymorphic compound, different crystal forms have different physical and chemical properties, including melting point, chemical stability, apparent solubility, dissolution rate, optical and mechanical properties, etc. These physical and chemical properties directly determine the efficacy of a particular crystal form, and directly affect the quality of raw materials and preparations.
因此,有必要对成药化合物的各个晶型的性质进行研究,以满足药物制备的需要。Therefore, it is necessary to study the properties of each crystal form of the drug compound to meet the needs of drug preparation.
发明内容Summary of the invention
Figure PCTCN2020075918-appb-000002
Figure PCTCN2020075918-appb-000002
式I化合物的化学名为N-[(E)-4-[甲氧基(甲基)氨基]-4-氧代-丁-2-烯酰基]氨基甲 酸甲酯,代号为MS-77。The chemical name of the compound of formula I is N-[(E)-4-[methoxy(methyl)amino]-4-oxo-but-2-enoyl]carbamate methyl ester, and the code is MS-77.
本发明的目的之一在于提供了化学和物理稳定性很好的式I化合物的晶型I(以下称为“晶型I”)。所述晶型I在化学稳定性和加工(过滤、干燥)适应性方面具有优异的性质。One of the objectives of the present invention is to provide the crystal form I (hereinafter referred to as "crystal form I") of the compound of formula I with good chemical and physical stability. The crystal form I has excellent properties in terms of chemical stability and processing (filtration, drying) adaptability.
本发明的晶型I的X-射线粉末衍射图在以下衍射角2θ处具有特征峰:8.82±0.2°、14.56±0.2°、16.82±0.2°、17.72±0.2°、20.22±0.2°、22.50±0.2°、26.24±0.2°、29.40±0.2°。The X-ray powder diffraction pattern of the crystal form I of the present invention has characteristic peaks at the following diffraction angles 2θ: 8.82±0.2°, 14.56±0.2°, 16.82±0.2°, 17.72±0.2°, 20.22±0.2°, 22.50± 0.2°, 26.24±0.2°, 29.40±0.2°.
进一步地,所述晶型I的X-射线粉末衍射图在以下衍射角2θ处有特征峰:10.06±0.2°、24.22±0.2°、25.00±0.2°、26.76±0.2°、28.16±0.2°、30.56±0.2°、31.14±0.2°、33.74±0.2°、38.66±0.2°、40.20±0.2°、44.84±0.2°、46.04±0.2°。Further, the X-ray powder diffraction pattern of the crystal form I has characteristic peaks at the following diffraction angles 2θ: 10.06±0.2°, 24.22±0.2°, 25.00±0.2°, 26.76±0.2°, 28.16±0.2°, 30.56±0.2°, 31.14±0.2°, 33.74±0.2°, 38.66±0.2°, 40.20±0.2°, 44.84±0.2°, 46.04±0.2°.
更进一步地,本发明的晶型I的X-射线粉末衍射谱图具有如下表1所示的2θ、d和相对强度数据:Furthermore, the X-ray powder diffraction spectrum of crystal form I of the present invention has 2θ, d and relative intensity data as shown in Table 1 below:
表1Table 1
Figure PCTCN2020075918-appb-000003
Figure PCTCN2020075918-appb-000003
Figure PCTCN2020075918-appb-000004
Figure PCTCN2020075918-appb-000004
非限制性地,本发明的晶型I的X-射线粉末衍射谱图基本上如图1所示。Without limitation, the X-ray powder diffraction spectrum of the crystal form I of the present invention is basically as shown in FIG. 1.
本发明所述晶型I的差示扫描量热(DSC)图谱峰值为192.7℃。The peak value of the differential scanning calorimetry (DSC) spectrum of the crystalline form I of the present invention is 192.7°C.
非限制性地,本发明的晶型I具有如图2所示的DSC图谱。Without limitation, the crystal form I of the present invention has a DSC spectrum as shown in FIG. 2.
非限制性地,本发明的晶型I具有如图3所示的TGA图谱。Without limitation, the crystal form I of the present invention has a TGA pattern as shown in FIG. 3.
本发明的另一目的还在于提供所述晶型I的制备方法,所述方法选自下述方法中的任意一种:Another object of the present invention is to provide a method for preparing the crystal form I, and the method is selected from any one of the following methods:
方法(1),其包括以下步骤:Method (1), which includes the following steps:
1)将式I化合物溶解于醇、酮、酯、乙腈、二氧六环中的任一溶剂中,溶清,蒸发析晶;所述醇为C2-C4醇;所述酮为C3-C6酮;所述酯为C3-C6酯;所述溶解的温度为25~45℃;所述式I化合物与溶剂的质量体积比为(mg/ml)10:1.5~30;1) Dissolve the compound of formula I in any solvent of alcohol, ketone, ester, acetonitrile and dioxane, dissolve it, and evaporate and crystallize; the alcohol is a C2-C4 alcohol; the ketone is a C3-C6 Ketone; the ester is a C3-C6 ester; the dissolution temperature is 25-45°C; the mass-volume ratio of the compound of formula I to the solvent is (mg/ml) 10:1.5-30;
2)过滤,得晶型I。2) Filter to obtain crystal form I.
方法(2),其包括以下步骤:Method (2), which includes the following steps:
1)将式I化合物溶解于醇、酮、酯、乙腈、二氧六环、二甲基乙酰胺、二甲基甲酰胺、二甲亚砜中的任一溶剂中,降温析晶;所述醇为C2-C4醇;所述酮为C3-C6酮;所述酯为C3-C6酯;所述溶解的温度为40~70℃;所述式I化合物与溶剂的质量体积比为(mg/ml)10:0.4~3;所述析晶的温度为0~20℃;1) The compound of formula I is dissolved in any solvent of alcohol, ketone, ester, acetonitrile, dioxane, dimethylacetamide, dimethylformamide, and dimethylsulfoxide, and the temperature is lowered to crystallize; The alcohol is a C2-C4 alcohol; the ketone is a C3-C6 ketone; the ester is a C3-C6 ester; the dissolution temperature is 40-70°C; the mass-volume ratio of the compound of formula I to the solvent is (mg /ml) 10: 0.4~3; the temperature of said crystallization is 0~20℃;
或将式I化合物溶解于乙醇、异丙醇、二氧六环、丙酮、乙腈中的任一有机溶剂与水的混合溶剂中,降温析晶;所述溶解的温度为40~70℃;所述有机溶剂与水的体积比为1: 0.1~9;所述式I化合物与混合溶剂的质量体积比为(mg/ml)10:1~1.5;所述析晶的温度为0~5℃;Or the compound of formula I is dissolved in a mixed solvent of any organic solvent among ethanol, isopropanol, dioxane, acetone, and acetonitrile and water, and the temperature is lowered to crystallize; the dissolution temperature is 40-70°C; The volume ratio of the organic solvent to water is 1:0.1-9; the mass-volume ratio of the compound of formula I and the mixed solvent is (mg/ml) 10:1-1.5; the crystallization temperature is 0-5°C ;
2)过滤,得晶型I。2) Filter to obtain crystal form I.
本发明的另一目的在于提供式I化合物的晶型II(以下称为“晶型II”),所述晶型II的X-射线粉末衍射(XRD)图在以下衍射角2θ处有特征峰:7.34±0.2°、14.72±0.2°、16.98±0.2°、20.30±0.2°、22.16±0.2°、25.84±0.2°、38.44±0.2°。Another object of the present invention is to provide crystal form II of the compound of formula I (hereinafter referred to as "crystal form II"), the X-ray powder diffraction (XRD) pattern of the crystal form II has characteristic peaks at the following diffraction angle 2θ : 7.34±0.2°, 14.72±0.2°, 16.98±0.2°, 20.30±0.2°, 22.16±0.2°, 25.84±0.2°, 38.44±0.2°.
进一步地,所述晶型II的X-射线粉末衍射图谱在以下衍射角2θ处有特征峰:9.20±0.2°、13.90±0.2°、18.46±0.2°、23.74±0.2°、29.72±0.2°、34.42±0.2°。Further, the X-ray powder diffraction pattern of the crystal form II has characteristic peaks at the following diffraction angles 2θ: 9.20±0.2°, 13.90±0.2°, 18.46±0.2°, 23.74±0.2°, 29.72±0.2°, 34.42±0.2°.
更进一步地,本发明的晶型II的X-射线粉末衍射谱图具有如下表2所示的2θ数据:Furthermore, the X-ray powder diffraction spectrum of the crystal form II of the present invention has the 2θ data shown in Table 2 below:
表2Table 2
Figure PCTCN2020075918-appb-000005
Figure PCTCN2020075918-appb-000005
非限制性地,本发明的晶型II的X-射线粉末衍射谱图基本上如图4所示。Without limitation, the X-ray powder diffraction spectrum of the crystal form II of the present invention is basically shown in FIG. 4.
非限制性地,本发明的晶型II的差示扫描量热(DSC)图谱峰值为188.3℃。Without limitation, the peak value of the differential scanning calorimetry (DSC) spectrum of the crystalline form II of the present invention is 188.3°C.
非限制性地,本发明的晶型II具有如图5所示的DSC图谱。Without limitation, the crystal form II of the present invention has a DSC spectrum as shown in FIG. 5.
非限制性地,本发明的晶型II具有如图6所示的TGA图谱。Without limitation, the crystal form II of the present invention has a TGA pattern as shown in FIG. 6.
本发明的另一目的还在于提供所述晶型II的制备方法,所述方法选自下述方法中的任意一种:Another object of the present invention is to provide a method for preparing the crystal form II, the method is selected from any one of the following methods:
方法(1),其包括以下步骤:Method (1), which includes the following steps:
1)将式I化合物加入CH2Cl2中,回流溶解;所述式I化合物与CH2Cl2的质量体积比(mg/ml)为10:1.4~1.6;1) Add the compound of formula I to CH2Cl2 and dissolve under reflux; the mass-volume ratio (mg/ml) of the compound of formula I to CH2Cl2 is 10:1.4-1.6;
2)将上述溶液加至-15~-10℃的乙酸乙酯(EA)中;所述EA与CH2Cl2的体积比为0.9~1.1:1;2) Add the above solution to ethyl acetate (EA) at -15~-10℃; the volume ratio of the EA and CH2Cl2 is 0.9~1.1:1;
3)搅拌析晶,过滤,得晶型II。3) Stir to crystallize, filter to obtain crystal form II.
方法(2),其包括以下步骤:Method (2), which includes the following steps:
1)将式I化合物溶解于二氧六环中;所述溶解的温度为45~60℃;所述式I化合物与二氧六环的质量体积比(mg/ml)为10:1.4~1.6;1) The compound of formula I is dissolved in dioxane; the temperature of the dissolution is 45-60°C; the mass-volume ratio (mg/ml) of the compound of formula I to dioxane is 10:1.4-1.6 ;
2)将上述溶液加至4~6℃的正庚烷中;所述正庚烷与二氧六环的体积比为3~3.5:1;2) Add the above solution to n-heptane at 4-6°C; the volume ratio of n-heptane to dioxane is 3 to 3.5:1;
3)搅拌析晶,过滤,得晶型II。3) Stir to crystallize, filter to obtain crystal form II.
在上述方法中,式I化合物与溶剂的质量体积比的单位可以为mg/ml、g/L等,可以视具体的操作规模而定。In the above method, the unit of the mass-volume ratio of the compound of formula I to the solvent may be mg/ml, g/L, etc., depending on the specific scale of operation.
本发明的另一目的还在于提供药物组合物,所述药物组合物包含有效剂量的晶型I或晶型II;药物组合物还包含一种或多种药学上可接受的载体。Another object of the present invention is to provide a pharmaceutical composition, which comprises an effective dose of crystal form I or crystal form II; the pharmaceutical composition also comprises one or more pharmaceutically acceptable carriers.
本发明还涉及晶型I或晶型II,或它们的药物组合物在制备用于治疗与Nrf2激活相关的疾病的药物中的应用,其中所述与Nrf2激活相关的疾病为脑卒中、神经退行性疾病、糖尿病、糖尿病肾病、冠心病、动脉粥样硬化或非酒精性脂肪肝。The present invention also relates to the application of crystal form I or crystal form II, or their pharmaceutical composition in the preparation of a medicine for treating diseases related to Nrf2 activation, wherein the diseases related to Nrf2 activation are stroke and neurodegeneration Sexual diseases, diabetes, diabetic nephropathy, coronary heart disease, atherosclerosis or non-alcoholic fatty liver.
在另一方面,本发明还涉及晶型I或晶型II,或它们的药物组合物在制备用于治疗脑卒中的药物中的应用。In another aspect, the present invention also relates to the use of crystal form I or crystal form II, or their pharmaceutical composition in the preparation of a medicament for the treatment of stroke.
在另一方面,本发明还涉及晶型I或晶型II,或它们的药物组合物在制备用于治疗神经退行性疾病的药物中的应用。In another aspect, the present invention also relates to the application of the crystal form I or the crystal form II, or their pharmaceutical composition in the preparation of a medicament for the treatment of neurodegenerative diseases.
在优选的方案中,其中所述的神经退行性疾病选自多发性硬化症(MS)、阿尔茨海默 氏病(AD)、帕金森病(PD)、亨廷顿氏病(HD)、肌萎缩侧索硬化(ALS)、弗里德赖希氏共济失调(FRDA)、脊髓性肌萎缩(SMA)、视神经脊髓炎(NMO)和脊髓小脑性共济失调(SCA)。In a preferred embodiment, the neurodegenerative disease is selected from multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), muscle atrophy Lateral sclerosis (ALS), Friedreich’s ataxia (FRDA), spinal muscular atrophy (SMA), optic neuromyelitis (NMO) and spinocerebellar ataxia (SCA).
在另一方面,本发明还涉及晶型I或晶型II,或它们的药物组合物在制备用于治疗与免疫调节相关的疾病的药物中的应用,其中所述与免疫调节相关的疾病优选自银屑病、类风湿关节炎、系统性红斑狼疮、桥本氏甲状腺炎、移植排斥和炎性疾病。In another aspect, the present invention also relates to the use of crystal form I or crystal form II, or their pharmaceutical composition in the preparation of a medicament for the treatment of diseases related to immune regulation, wherein the diseases related to immune regulation are preferably From psoriasis, rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, transplant rejection and inflammatory diseases.
在本发明的另一方面,提供了一种治疗与Nrf2激活相关的疾病的方法,所述方法包括向有需要的患者施用有效剂量的晶型I或晶型II,或它们的药物组合物。In another aspect of the present invention, there is provided a method for treating diseases associated with Nrf2 activation, the method comprising administering an effective dose of crystal form I or crystal form II, or a pharmaceutical composition thereof, to a patient in need.
在本发明的另一方面,提供了一种治疗脑卒中的方法,所述方法包括向有需要的患者施用有效剂量的晶型I或晶型II,或它们的药物组合物。In another aspect of the present invention, there is provided a method for treating stroke, the method comprising administering an effective dose of crystal form I or crystal form II, or a pharmaceutical composition thereof to a patient in need.
在本发明的另一方面,提供了一种治疗神经退行性疾病的方法,所述方法包括向有需要的患者施用有效剂量的晶型I或晶型II,或它们的药物组合物。In another aspect of the present invention, there is provided a method of treating neurodegenerative diseases, the method comprising administering an effective dose of crystal form I or crystal form II, or a pharmaceutical composition thereof to a patient in need.
在本发明的另一方面,提供了一种治疗与免疫调节相关的疾病的方法,所述方法包括向有需要的患者施用有效剂量的晶型I或晶型II,或它们的药物组合物。In another aspect of the present invention, there is provided a method for treating diseases related to immune regulation, the method comprising administering an effective dose of crystal form I or crystal form II, or a pharmaceutical composition thereof, to a patient in need.
本发明人经过大量研究发现了式I化合物的晶型I和晶型II,其溶解性良好、结晶工艺简单、便于操作、污染小、可实现工业化生产,而且具有晶型I或晶型II的式I化合物具备产品纯度高、理化性质优异、化学稳定性良好、加工(过滤、干燥、)可再现的优点。After a lot of research, the inventors have discovered the crystal form I and crystal form II of the compound of formula I, which have good solubility, simple crystallization process, easy operation, low pollution, can realize industrial production, and have crystal form I or crystal form II. The compound of formula I has the advantages of high product purity, excellent physical and chemical properties, good chemical stability, and reproducible processing (filtering, drying,).
附图说明:Description of the drawings:
图1为实施例1所得式I化合物的晶型I的X-射线粉末衍射图谱。Fig. 1 is an X-ray powder diffraction pattern of the crystal form I of the compound of formula I obtained in Example 1.
图2为实施例1所得式I化合物的晶型I的DSC图谱。2 is a DSC chart of the crystal form I of the compound of formula I obtained in Example 1.
图3为实施例1所得式I化合物的晶型I的TGA图谱。3 is a TGA spectrum of the crystal form I of the compound of formula I obtained in Example 1.
图4为实施例5所得式I化合物的晶型II的X-射线粉末衍射图谱。4 is an X-ray powder diffraction pattern of the crystal form II of the compound of formula I obtained in Example 5.
图5为实施例5所得式I化合物的晶型II的DSC图谱。5 is a DSC chart of the crystal form II of the compound of formula I obtained in Example 5.
图6为实施例5所得式I化合物的晶型II的TGA图谱。6 is the TGA spectrum of the crystal form II of the compound of formula I obtained in Example 5.
图7是式I化合物诱导HT22细胞中Nrf2入核(4h)。Fig. 7 shows that the compound of formula I induces Nrf2 to enter the nucleus in HT22 cells (4h).
图8是式I化合物上调HT22细胞HO-1蛋白的表达(4h)。Figure 8 shows that the compound of formula I up-regulates the expression of HO-1 protein in HT22 cells (4h).
图9是式I化合物对小鼠EAE的抑制作用(***p<0.001,**p<0.01,*p<0.05,与溶剂组相比(one-way ANOVA/Dunnett))。Figure 9 shows the inhibitory effect of the compound of formula I on mouse EAE (***p<0.001, **p<0.01, *p<0.05, compared with the solvent group (one-way ANOVA/Dunnett)).
图10是Morris水迷宫实验式I化合物对AD大鼠模型逃避潜伏期的影响(***p<0.001,**p<0.01,*p<0.05,与模型组相比(one-way ANOVA/Dunnett))。Figure 10 is the effect of Morris water maze experiment formula I compound on the escape latency of AD rat model (***p<0.001, **p<0.01, *p<0.05, compared with the model group (one-way ANOVA/Dunnett) )).
图11是Morris水迷宫实验式I化合物对AD大鼠模型跨越目标平台次数的影响(***p<0.001,**p<0.01,*p<0.05,与模型组相比(one-way ANOVA/Dunnett))。Figure 11 is the effect of Morris water maze experiment formula I compound on the number of times the AD rat model crosses the target platform (***p<0.001, **p<0.01, *p<0.05, compared with the model group (one-way ANOVA) /Dunnett)).
图12是式I化合物对6-OHDA引发的PD大鼠转棒实验中潜伏期的影响(***p<0.001,**p<0.01,*p<0.05,与模型组相比(one-way ANOVA/Dunnett))。Figure 12 shows the effect of the compound of formula I on the latency in the 6-OHDA-induced PD rat rotarod experiment (***p<0.001, **p<0.01, *p<0.05, compared with the model group (one-way) ANOVA/Dunnett)).
图13是式I化合物对6-OHDA引发的PD大鼠爬杆实验中爬杆时间的影响(***p<0.001,**p<0.01,*p<0.05,与模型组相比(one-way ANOVA/Dunnett))。Figure 13 is the effect of the compound of formula I on the climbing time in the 6-OHDA-induced PD rat climbing experiment (***p<0.001, **p<0.01, *p<0.05, compared with the model group (one -way ANOVA/Dunnett)).
图14是式I化合物对阿扑吗啡诱导PD大鼠旋转的速度的影响(***p<0.001,**p<0.01,*p<0.05,与模型组相比(one-way ANOVA/Dunnett))。Figure 14 is the effect of formula I compound on the rotation speed of PD rats induced by apomorphine (***p<0.001, **p<0.01, *p<0.05, compared with the model group (one-way ANOVA/Dunnett) )).
图15是式I化合物对急性脑动脉缺血再灌注损伤大鼠神经功能评分的影响(***p<0.001,**p<0.01,*p<0.05,与模型组相比(one-way ANOVA/Dunnett))。Figure 15 is the effect of the compound of formula I on the neurological score of acute cerebral artery ischemia-reperfusion injury in rats (***p<0.001, **p<0.01, *p<0.05, compared with the model group (one-way) ANOVA/Dunnett)).
图16是式I化合物对急性脑动脉缺血再灌注损伤大鼠脑梗区/全脑重量比的影响(***p<0.001,**p<0.01,*p<0.05,与模型组相比(one-way ANOVA/Dunnett))。Figure 16 shows the effect of compound of formula I on the weight ratio of cerebral infarction area/whole brain in rats with acute cerebral artery ischemia reperfusion injury (***p<0.001, **p<0.01, *p<0.05, similar to the model group Than (one-way ANOVA/Dunnett)).
图17是式I化合物对豚鼠皮肤的致敏率。Figure 17 shows the sensitization rate of the compound of formula I to the skin of guinea pigs.
具体实施例Specific embodiment
下列实施例是为了进一步解释说明本发明,而不是构成对本发明范围的限制或限定。The following examples are intended to further explain the present invention, but not to limit or limit the scope of the present invention.
本发明方法中所使用的式I化合物原料来源于以下合成方法:The raw materials of the compound of formula I used in the method of the present invention are derived from the following synthetic methods:
制备例:式I化合物原料的合成方法Preparation example: Synthesis method of the compound of formula I
(E)-N'-甲氧基-N'-甲基-丁-2-烯二酰胺(化合物Ia)的制备(E) Preparation of-N'-methoxy-N'-methyl-but-2-ene diamide (compound Ia)
Figure PCTCN2020075918-appb-000006
Figure PCTCN2020075918-appb-000006
步骤一:向反应瓶中加入250克(1.92摩尔)(E)-4-甲氧基-4-氧代-丁-2-烯酸,225克(2.31摩尔)N,O-二甲基羟胺盐酸盐和2500毫升二氯甲烷,降温至0~10℃,加入550克(2.88摩尔)1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(EDCI),升温至室温,反应1小时,反应完毕。1500毫升水洗涤,收集有机相,活性炭脱色,过滤,滤液浓缩至干。Step 1: Add 250g (1.92mol) (E)-4-methoxy-4-oxo-but-2-enoic acid, 225g (2.31mol) N,O-dimethylhydroxylamine into the reaction flask Hydrochloride and 2500 ml of dichloromethane, reduce the temperature to 0~10℃, add 550 g (2.88 mol) of 1-ethyl-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) , Warm up to room temperature, react for 1 hour, the reaction is complete. Wash with 1500 ml of water, collect the organic phase, decolorize with activated carbon, filter, and concentrate the filtrate to dryness.
步骤二:向上述得到的浓缩物加入1250毫升氨水,冷却至0~10℃,反应20分钟,反应完毕,过滤,固体用乙醇和乙酸乙酯重结晶得142.3克化合物Ia。Step 2: Add 1250 ml of ammonia to the concentrate obtained above, cool to 0-10°C, react for 20 minutes, after the reaction is complete, filter, and recrystallize the solid with ethanol and ethyl acetate to obtain 142.3 g of compound Ia.
N-[(E)-4-[甲氧基(甲基)氨基]-4-氧代-丁-2-烯酰基]氨基甲酸甲酯(式I化合物)的制备Preparation of methyl N-[(E)-4-[methoxy(methyl)amino]-4-oxo-but-2-enoyl]carbamate (compound of formula I)
Figure PCTCN2020075918-appb-000007
Figure PCTCN2020075918-appb-000007
步骤一:向反应瓶中投入5.0克(31.6毫摩尔)(E)-N'-甲氧基-N'-甲基-丁-2-烯二酰胺(化合物Ia)和50毫升1,2-二氯乙烷,冷却至0~10℃,加入6毫升(70.9毫摩尔)草酰氯,室温反应6小时,升温至65℃,反应15分钟,浓缩至干,备用。Step 1: Put 5.0 g (31.6 mmol) (E)-N'-methoxy-N'-methyl-but-2-ene diamide (compound Ia) and 50 ml 1,2-into the reaction flask Dichloroethane, cooled to 0-10°C, added 6 ml (70.9 mmol) of oxalyl chloride, reacted at room temperature for 6 hours, heated to 65°C, reacted for 15 minutes, concentrated to dryness, and set aside.
步骤二:向另一反应瓶中加入30毫升无水甲醇,降温至0~10℃,加入步骤一所得的浓缩液,反应10分钟,析出固体,得3.56克式I化合物。Step 2: Add 30 milliliters of anhydrous methanol to another reaction flask, lower the temperature to 0-10°C, add the concentrated solution obtained in Step 1, and react for 10 minutes to precipitate a solid to obtain 3.56 g of formula I compound.
本发明所使用的溶剂没有特别的限制,可采用商购的常规溶剂。The solvent used in the present invention is not particularly limited, and commercially available conventional solvents can be used.
除非另有说明,本发明方法中所述的“搅拌”可以采用本领域的常规方法,例如搅拌方式包括磁力搅拌、机械搅拌,搅拌速度为50-300rpm/min,优选100-200rpm/min。Unless otherwise specified, the "stirring" described in the method of the present invention can adopt conventional methods in the art. For example, the stirring method includes magnetic stirring and mechanical stirring, and the stirring speed is 50-300 rpm/min, preferably 100-200 rpm/min.
本发明所涉及的X-射线粉末衍射仪器及测试条件为:X-衍射仪器型号MiniFlex600Cu靶;操作方法:扫描速度20°/min,扫描步宽0.02°。The X-ray powder diffraction instrument and test conditions involved in the present invention are: X-ray diffraction instrument model MiniFlex600Cu target; operation method: scanning speed 20°/min, scanning step width 0.02°.
本发明涉及的DSC测试条件为:DSC检测仪型号为:NETZSCH DSC 214 Polyma;;操作方法:升温速率10℃/min,温度范围:25-250℃。The DSC test conditions involved in the present invention are: DSC detector model: NETZSCH DSC 214 Polyma; operation method: heating rate 10°C/min, temperature range: 25-250°C.
本发明涉及的TGA测试条件为:TGA检测仪型号为:METTLER TOLEDO TGA2;操作方法:升温速率10℃/min,温度范围:25-250℃。The TGA test conditions involved in the present invention are: the model of the TGA detector is: METTLER TOLEDO TGA2; the operation method: the heating rate is 10°C/min, and the temperature range: 25-250°C.
本发明涉及的式I化合物HPLC的纯度测试条件为:色谱柱:Welch Ultimate XB-C18 250*4.6mm 5μm;流动相A:0.01%三氟乙酸-水;流动相B:乙腈;过滤并脱气;检测波长:226nm;流速:1.0ml/min;进样量:10μl;柱温:25℃;流动相条件如表3所示:The HPLC purity test conditions of the compound of formula I involved in the present invention are: chromatographic column: Welch Ultimate XB-C18 250*4.6mm 5μm; mobile phase A: 0.01% trifluoroacetic acid-water; mobile phase B: acetonitrile; filtration and degassing ; Detection wavelength: 226nm; Flow rate: 1.0ml/min; Injection volume: 10μl; Column temperature: 25°C; Mobile phase conditions are shown in Table 3:
表3table 3
t(min)t(min) 流动相A(%)Mobile phase A (%) 流动相B(%)Mobile phase B (%)
00 8383 1717
3030 8383 1717
应当强调的是,本发明技术方案中所涉及的数值或数值端点,其含义或意欲的保护范围并不局限于该数字本身,本领域技术人员能够理解,它们包含了那些已被本领域广为接受的可允许误差范围,例如实验误差、测量误差、统计误差和随机误差等等,而这些误差范围均包含在本发明的范围之内。It should be emphasized that the meaning or intended protection scope of the numerical value or numerical endpoint involved in the technical solution of the present invention is not limited to the number itself. Those skilled in the art can understand that they include those that have been widely used in the art. Acceptable allowable error ranges, such as experimental errors, measurement errors, statistical errors and random errors, etc., and these error ranges are all included in the scope of the present invention.
实施例1Example 1
将制备例获得的原料1g加入300ml乙醇中,45℃溶解,45℃蒸发析晶,过滤,得0.52g晶体,HPLC=99.89%,经测X-射线粉末衍射图谱(XRD),确认为晶型I。Add 1 g of the raw material obtained in the preparation example to 300 ml of ethanol, dissolve at 45°C, evaporate and crystallize at 45°C, and filter to obtain 0.52g crystals, HPLC=99.89%, by measuring X-ray powder diffraction pattern (XRD), it is confirmed that the crystal form I.
该晶型的X-射线粉末衍射、DSC以及TGA谱图分别如图1-3所示,在本发明中将其命名为晶型I。The X-ray powder diffraction, DSC and TGA spectra of this crystal form are shown in Figures 1-3, respectively, and are named crystal form I in the present invention.
实施例2Example 2
将制备例获得的原料10mg加入1.5ml二氧六环中,40℃溶清,40℃蒸发析晶,过滤,得5.3mg晶体,HPLC=99.84%,经测X-射线粉末衍射图谱(XRD),确认为晶型I。Add 10 mg of the raw material obtained in the preparation example to 1.5 ml of dioxane, dissolve at 40°C, evaporate and crystallize at 40°C, and filter to obtain 5.3 mg of crystals, HPLC=99.84%, measured X-ray powder diffraction pattern (XRD) , Confirmed as crystal form I.
实施例3Example 3
将制备例获得的原料10mg加入30ml乙酸异丙酯中,25℃溶清,25℃蒸发析晶,过滤,得6.4mg晶体,HPLC=99.76%,经测X-射线粉末衍射图谱(XRD),确认为晶型I。Add 10 mg of the raw materials obtained in the preparation example to 30 ml of isopropyl acetate, dissolve at 25°C, evaporate and crystallize at 25°C, and filter to obtain 6.4 mg of crystals, HPLC=99.76%, measured X-ray powder diffraction pattern (XRD), Confirmed as crystal form I.
实施例4Example 4
将制备例获得的原料10mg加入3ml乙醇中,70℃溶解,降温至20℃析晶,过滤,得4.5mg晶体,HPLC=99.86%,经测X-射线粉末衍射图谱(XRD),确认为晶型I。Add 10 mg of the raw material obtained in the preparation example to 3 ml of ethanol, dissolve at 70°C, cool to 20°C to crystallize, and filter to obtain 4.5 mg of crystals. HPLC=99.86%. The X-ray powder diffraction pattern (XRD) was measured and confirmed to be crystals. Type I.
实施例5Example 5
将制备例获得的原料50mg加入2ml二甲基乙酰胺中,40℃溶解,降温至0℃析晶,过滤,得3.8mg晶体,HPLC=99.86%,经测X-射线粉末衍射图谱(XRD),确认为晶型I。Add 50mg of the raw material obtained in the preparation example to 2ml of dimethylacetamide, dissolve at 40°C, cool to 0°C to crystallize, and filter to obtain 3.8mg crystals, HPLC=99.86%, measured X-ray powder diffraction pattern (XRD) , Confirmed as crystal form I.
实施例6Example 6
将制备例获得的原料10mg加入1.5ml甲醇水的混合物中,40℃溶解,甲醇水的体积比为10:1,降温至0℃析晶,过滤,得4.9mg晶体,HPLC=99.81%,经测X-射线粉末衍射图谱(XRD),确认为晶型I。Add 10 mg of the raw material obtained in the preparation example to 1.5 ml of methanol-water mixture, dissolve at 40°C, the volume ratio of methanol-water is 10:1, cool to 0°C to crystallize, filter to obtain 4.9mg crystals, HPLC=99.81%, The X-ray powder diffraction pattern (XRD) was measured, and it was confirmed to be crystal form I.
实施例7Example 7
将制备例获得的原料10mg加入1ml乙腈水的混合物中,70℃溶解,乙腈水的体积比为1:9,降温至5℃析晶,过滤,得6.3mg晶体,HPLC=99.76%,经测X-射线粉末衍射图谱(XRD),确认为晶型I。Add 10 mg of the raw material obtained in the preparation example to 1 ml of acetonitrile water mixture, dissolve at 70°C, the volume ratio of acetonitrile water is 1:9, cool to 5°C to crystallize, filter to obtain 6.3mg crystals, HPLC=99.76%, measured X-ray powder diffraction pattern (XRD), confirmed to be crystal form I.
实施例8Example 8
将制备例获得的原料1g加入150ml CH2Cl2中,回流溶解,将溶液加至-10℃的150ml EA中,搅拌析晶,过滤,得0.21g晶体,HPLC=99.62%,经测X-射线粉末衍射图谱(XRD),确认为晶型II。Add 1g of the raw material obtained in the preparation example to 150ml CH2Cl2, dissolve under reflux, add the solution to 150ml EA at -10°C, stir and crystallize, filter to obtain 0.21g crystals, HPLC=99.62%, measured by X-ray powder diffraction The spectrum (XRD) confirmed that it was form II.
该晶型的X-射线粉末衍射、DSC以及TGA谱图分别如图4-6所示,在本发明中将其命名为晶型II。The X-ray powder diffraction, DSC and TGA spectra of this crystal form are shown in Figures 4-6, respectively, and are named crystal form II in the present invention.
实施例9Example 9
将制备例获得的原料10mg加入1.4ml CH2Cl2中,回流溶解,将溶液加至-10℃的1.26ml EA中,搅拌析晶,过滤,得2.9mg晶体,HPLC=99.61%,经测X-射线粉末衍射图谱(XRD),确认为晶型II。Add 10mg of the raw material obtained in the preparation example to 1.4ml CH2Cl2, dissolve under reflux, add the solution to 1.26ml EA at -10°C, stir and crystallize, filter to obtain 2.9mg crystals, HPLC=99.61%, measured by X-ray Powder diffraction pattern (XRD), confirmed to be crystal form II.
实施例10Example 10
将制备例获得的原料10mg加入1.6ml CH2Cl2中,回流溶解,将溶液加至-15℃的1.76ml EA中,搅拌析晶,过滤,得3.5mg晶体,HPLC=99.62%,经测X-射线粉末衍射图谱(XRD),确认为晶型II。Add 10mg of the raw material obtained in the preparation example to 1.6ml CH2Cl2, dissolve under reflux, add the solution to 1.76ml EA at -15°C, stir and crystallize, filter to obtain 3.5mg crystals, HPLC=99.62%, measured by X-ray Powder diffraction pattern (XRD), confirmed to be crystal form II.
实施例11Example 11
将制备例获得的原料10mg加入1.4ml二氧六环中,45℃溶解,将溶液加至5℃的4.2ml正庚烷中,搅拌析晶,过滤,得3.6mg晶体,HPLC=99.61%,经测X-射线粉末衍射图谱(XRD),确认为晶型II。Add 10mg of the raw material obtained in the preparation example to 1.4ml dioxane, dissolve at 45°C, add the solution to 4.2ml n-heptane at 5°C, stir and crystallize and filter to obtain 3.6mg crystals, HPLC=99.61%, After measuring the X-ray powder diffraction pattern (XRD), it was confirmed to be crystal form II.
实施例12Example 12
将制备例获得的原料10mg加入1.5ml二氧六环中,50℃溶解,将溶液加至6℃的5ml正庚烷中,搅拌析晶,过滤,得3.4mg晶体,HPLC=99.62%,经测X-射线粉末衍射图谱(XRD),确认为晶型II。Add 10 mg of the raw material obtained in the preparation example to 1.5 ml of dioxane, dissolve at 50°C, add the solution to 5 ml of n-heptane at 6°C, stir and crystallize, and filter to obtain 3.4 mg of crystals. HPLC=99.62%. The X-ray powder diffraction pattern (XRD) was measured, and it was confirmed to be crystal form II.
实施例13Example 13
将制备例获得的原料10mg加入1.6ml二氧六环中,60℃溶解,将溶液加至4℃的5.6ml正庚烷中,搅拌析晶,过滤,得3.9mg晶体,HPLC=99.61%,经测X-射线粉末衍射图谱(XRD),确认为晶型II。Add 10 mg of the raw material obtained in the preparation example to 1.6 ml of dioxane, dissolve at 60°C, add the solution to 5.6 ml of n-heptane at 4°C, stir and crystallize, filter to obtain 3.9 mg of crystals, HPLC=99.61%, After measuring the X-ray powder diffraction pattern (XRD), it was confirmed to be crystal form II.
稳定性实验Stability experiment
取实施例1制备的晶型I和实施例8制备的晶型II,进行60℃条件下放置10天的稳定性实验。检测化合物在放置前后的HPLC纯度和最大单杂含量,结果见表4;通过X-射线粉末衍射检测分析放置10天后的样品的晶型变化,结果见表5。Take the crystalline form I prepared in Example 1 and the crystalline form II prepared in Example 8, and conduct a stability test at 60° C. for 10 days. The HPLC purity and maximum single impurity content of the compound before and after storage were tested, and the results are shown in Table 4; the crystal form changes of the samples after 10 days of storage were analyzed by X-ray powder diffraction detection, and the results are shown in Table 5.
表4Table 4
Figure PCTCN2020075918-appb-000008
Figure PCTCN2020075918-appb-000008
从表4的60℃条件下放置10天的稳定性数据可以看出:10天后,晶型I和晶型II的HPLC纯度和最大单杂含量变化值均较小,两种晶型化学稳定性较优;而晶型I的HPLC纯度和最大单杂含量变化小于晶型II的HPLC纯度和最大单杂含量变化,该结果表明,本发明获得的晶型I在60℃条件下的稳定性优于晶型II。From the stability data of 10 days at 60°C in Table 4, it can be seen that after 10 days, the HPLC purity and maximum single impurity content of crystal form I and crystal form II are both small, and the chemical stability of the two crystal forms Better; and the HPLC purity and maximum single impurity content changes of crystal form I are smaller than the HPLC purity and maximum single impurity content changes of crystal form II. This result shows that the crystal form I obtained by the present invention has excellent stability at 60°C于晶型II.
表5table 5
 To 10天10 days
晶型IForm I XRD图谱无变化No change in XRD pattern
晶型IICrystal Form II XRD显示为晶型I和II的混晶XRD shows a mixed crystal of crystalline form I and II
由表5的60℃条件下放置10天的XRD分析结果可以看出:10天后晶型I未发生变化;而晶型II发生了转晶,部分转晶为晶型I,该结果表明,本发明的晶型I在60℃条件下的物理稳定性优于晶型II。From the XRD analysis results of 10 days at 60℃ in Table 5, it can be seen that the crystal form I has not changed after 10 days; while the crystal form II has been transformed, and some of the crystals are transformed into the crystal form I. This result shows that The physical stability of the invented crystalline form I at 60°C is better than that of the crystalline form II.
吸湿性实验Hygroscopicity test
取干燥的具塞玻璃称量瓶(外径为50mm,高为15mm),于试验前一天置于适宜的25℃±1℃恒温干燥器(下部放置氯化钠饱和溶液),分别取同样重量的实施例1中制备得到的晶型I和实施例8中制备得到的晶型II,平铺于上述称量瓶中,在温度为25℃±1℃、相对湿度为75%±2%条件下放置24h,进行引湿性实验。结果见表6。Take a dry stoppered glass weighing bottle (outer diameter of 50mm, height of 15mm), place it in a suitable 25°C±1°C constant temperature desiccator (place a saturated sodium chloride solution at the bottom) the day before the test, and take the same weight The crystal form I prepared in Example 1 and the crystal form II prepared in Example 8 were laid flat in the above weighing bottle at a temperature of 25℃±1℃ and a relative humidity of 75%±2%. Place it for 24h and conduct a moisture absorption test. The results are shown in Table 6.
表6Table 6
 To 晶型IForm I 晶型IICrystal Form II
吸湿性(%)Hygroscopicity (%) 00 00
由表6的吸湿性实验结果可知:本发明获得的晶型I和晶型II无吸湿性。It can be seen from the results of the hygroscopicity experiment in Table 6 that the crystal form I and crystal form II obtained in the present invention have no hygroscopicity.
匀浆实验Homogenization experiment
取30mg实施例8制备的晶型II,加入4ml乙醇,20℃搅拌1h,过滤,经测X-射线粉末衍射图谱(XRD),确认为晶型I。Take 30 mg of the crystal form II prepared in Example 8, add 4 ml of ethanol, stir at 20° C. for 1 hour, and filter. The X-ray powder diffraction pattern (XRD) is measured, and the crystal form I is confirmed.
由匀浆实验结果可知:本发明获得的晶型II在乙醇溶液中容易转晶为晶型I,表明晶型I的稳定性优于晶型II。From the homogenization experiment results, it is known that the crystal form II obtained in the present invention is easily transformed into crystal form I in an ethanol solution, indicating that the stability of the crystal form I is better than that of the crystal form II.
溶解度实验Solubility test
样品溶液:取适量实施例1所得的晶型I和实施例8所得的晶型II至20ml量瓶,加10ml溶媒(盐酸水溶液,pH=1.0、pH=2.0、pH=4.5、pH=6.8)混匀,在37℃条件下振摇10min,立即过滤,取滤液60℃烘干至恒重,采用重量法获得相应晶型的溶解度,结果见表7。Sample solution: Take an appropriate amount of the crystal form I obtained in Example 1 and the crystal form II obtained in Example 8 to a 20ml measuring flask, and add 10ml of solvent (aqueous hydrochloric acid, pH=1.0, pH=2.0, pH=4.5, pH=6.8) Mix well, shake for 10 min at 37°C, filter immediately, take the filtrate and dry it to constant weight at 60°C, and obtain the solubility of the corresponding crystal form by gravimetric method. The results are shown in Table 7.
表7Table 7
Figure PCTCN2020075918-appb-000009
Figure PCTCN2020075918-appb-000009
Figure PCTCN2020075918-appb-000010
Figure PCTCN2020075918-appb-000010
由表7结果可知,晶型I和晶型II的在上述溶媒中的溶解度均随pH值的增加而增大,但晶型II的溶解度优于晶型I的溶解度。It can be seen from the results in Table 7 that the solubility of crystal form I and crystal form II in the above-mentioned solvent both increase with the increase of pH value, but the solubility of crystal form II is better than that of crystal form I.
活性例Active example
式I化合物的活性实验,按下列方式进行The activity experiment of the compound of formula I is carried out in the following way
一.化合物诱导HT22细胞中Nrf2入核及细胞质中HO-1表达1. The compound induces Nrf2 to enter the nucleus and HO-1 expression in the cytoplasm of HT22 cells
取增殖良好处于对数生长期的小鼠海马神经元细胞HT22细胞,经胰酶消化分散后计数,采用含5%胎牛血清的RPMI 1640培养基调整细胞密度,将1×10 6个细胞接种至T25培养瓶中,置于37℃,5%CO 2,100%相对湿度培养箱中培养24h后,加入一定浓度的化合物。继续培养,并于4h后收取细胞,提取核蛋白和细胞总蛋白,Western Blot检测核内Nrf2的含量和细胞质中HO-1的表达量。实验结果表明式I化合物可显著地提高HT22细胞核内Nrf2蛋白的含量,提示式I化合物具有诱导细胞质中Nrf2蛋白进入细胞核的作用(图7),同浓度下,式I化合物的作用强于富马酸二甲酯(DMF);而且式I化合物能诱导细胞质中HO-1的表达,从而显著增加细胞质中HO-1的表达量(图8)。 Take the mouse hippocampal neuron cells HT22 cells that proliferate well and are in the logarithmic growth phase, and count them after trypsinization and dispersion. The cell density is adjusted with RPMI 1640 medium containing 5% fetal bovine serum, and 1×10 6 cells are inoculated Into the T25 culture flask, place it in a 37°C, 5% CO 2 , 100% relative humidity incubator for 24 hours, and then add a certain concentration of compound. Continue to culture, and harvest the cells after 4 hours, extract nuclear protein and total cell protein, Western Blot to detect the content of Nrf2 in the nucleus and the expression of HO-1 in the cytoplasm. The experimental results show that the compound of formula I can significantly increase the content of Nrf2 protein in the nucleus of HT22 cells, suggesting that the compound of formula I has the effect of inducing Nrf2 protein in the cytoplasm to enter the nucleus (Figure 7). At the same concentration, the compound of formula I is stronger than fumar Dimethyl Acid (DMF); and the compound of formula I can induce the expression of HO-1 in the cytoplasm, thereby significantly increasing the expression of HO-1 in the cytoplasm (Figure 8).
二.化合物对L-谷氨酸钠损伤的HT22细胞的保护作用2. The protective effect of the compound on HT22 cells injured by sodium L-glutamate
取增殖良好处于对数生长期的HT22细胞,经胰酶消化分散后计数,采用含5%胎牛血清的RPMI 1640培养基调整细胞密度,以1000个细胞/孔接种至96孔板,置于37℃,5%CO 2,100%相对湿度培养箱中培养24h。各孔加入不同浓度的化合物,继续培养24h后,加入一定浓度的L-谷氨酸单钠盐,再培养24h后使用CellTiter-Glo kit检测细胞活力。实验结果表明,在谷氨酸钠诱导的HT22细胞损伤模型上,本发明化合物对L-谷氨酸钠损伤的HT22细胞均有保护作用(表8)。 Take HT22 cells that proliferate well and are in the logarithmic growth phase, and count after trypsin digestion and dispersion. The cell density is adjusted with RPMI 1640 medium containing 5% fetal bovine serum, and 1000 cells/well are inoculated into a 96-well plate. Incubate in an incubator at 37°C, 5% CO 2 , and 100% relative humidity for 24 hours. Compounds of different concentrations were added to each well, after 24 hours of continuous cultivation, a certain concentration of L-glutamate monosodium salt was added, and after another 24 hours of cultivation, the CellTiter-Glo kit was used to detect cell viability. The experimental results show that in the HT22 cell injury model induced by sodium glutamate, the compound of the present invention has a protective effect on HT22 cells injured by sodium L-glutamate (Table 8).
表8 化合物对谷氨酸钠诱导的HT22细胞损伤模型的保护作用Table 8 The protective effects of compounds on the HT22 cell injury model induced by sodium glutamate
CmpsCmps EC 50(μM) EC 50 (μM)
DMFDMF 0.330.33
式I化合物Formula I compound 0.180.18
三.化合物对IFN-γ诱导Hacat细胞分泌CXCL9的抑制作用3. The compound's inhibitory effect on IFN-γ-induced Hacat cells to secrete CXCL9
取增殖良好处于对数生长期的Hacat细胞,经胰酶消化分散后计数,制成细胞悬液,采用含10%胎牛血清的MEM培养液调整细胞密度,以1.2×10 5个细胞/孔接种至24孔板中,于37℃,5%CO 2,100%相对湿度培养箱中培养16h,然后同时加入IFN-γ及不同浓度的化合物,继续培养24h,收集细胞上清液,用Human CXCL9/MIG Elisa kit检测CXCL9的分泌量。实验结果表明,在IFN-γ诱导Hacat细胞分泌CXCL9的细胞模型上,式I化合物对CXCL9的分泌具有一定的抑制作用(表9)。 Take Hacat cells that proliferate well and are in the logarithmic growth phase. After trypsin digestion and dispersion, they are counted to prepare a cell suspension. The cell density is adjusted with MEM medium containing 10% fetal bovine serum to 1.2×10 5 cells/well Inoculate into a 24-well plate, incubate in a 37°C, 5% CO 2 , 100% relative humidity incubator for 16 hours, then add IFN-γ and different concentrations of compounds at the same time, continue to incubate for 24 hours, collect the cell supernatant, and use Human CXCL9/MIG Elisa kit detects the secretion of CXCL9. The experimental results showed that in the cell model where IFN-γ induced Hacat cells to secrete CXCL9, the compound of formula I had a certain inhibitory effect on the secretion of CXCL9 (Table 9).
表9 化合物对IFN-γ诱导Hacat细胞分泌CXCL9的抑制作用Table 9 Inhibitory effects of compounds on IFN-γ-induced Hacat cells to secrete CXCL9
CmpsCmps IC 50(μM) IC 50 (μM)
DMFDMF 30.0430.04
式I化合物Formula I compound 15.9715.97
四.化合物对LPS诱导Ana-1细胞分泌TNF-α的抑制作用4. The inhibitory effect of the compound on LPS-induced TNF-α secretion in Ana-1 cells
取增殖良好处于对数生长期的Ana-1细胞,经胰酶消化分散后计数,制成细胞悬液,采用含10%胎牛血清的RPMI1640培养液调整细胞密度,以0.8×10 5个细胞/孔接种至24孔板,于37℃,5%CO 2,100%相对湿度培养箱中培养24h后,加入LPS及不同浓度的化合物溶液,继续培养3h,收集细胞上清液,用Mouse TNF-αElisa kit检测TNF-α的分泌量。实验结果表明,在LPS诱导的Ana-1细胞分泌TNF-α的细胞模型上,式I化合物对TNF-α的分泌具有一定的抑制作用(表10)。 Take Ana-1 cells that proliferate well and are in the logarithmic growth phase, and count them after trypsin digestion and dispersion to prepare a cell suspension. Use RPMI1640 culture medium containing 10% fetal bovine serum to adjust the cell density to 0.8×10 5 cells Inoculate each well into a 24-well plate, incubate in a 37°C, 5% CO 2 , 100% relative humidity incubator for 24 hours, add LPS and compound solutions of different concentrations, continue to incubate for 3 hours, collect the cell supernatant, and use Mouse TNF -αElisa kit detects the secretion of TNF-α. The experimental results show that in the cell model of Ana-1 cells secreting TNF-α induced by LPS, the compound of formula I has a certain inhibitory effect on the secretion of TNF-α (Table 10).
表10 化合物对LPS诱导Ana-1细胞分泌TNF-α的抑制作用Table 10 Inhibitory effect of compounds on LPS-induced TNF-α secretion in Ana-1 cells
CmpsCmps IC 50(μM) IC 50 (μM)
DMFDMF 52.6252.62
式I化合物Formula I compound 3.183.18
五.化合物对MOG诱导的C57BL/6小鼠实验性变态反应性脑脊髓炎(EAE)的抑制作用5. The inhibitory effect of the compound on MOG-induced experimental allergic encephalomyelitis (EAE) in C57BL/6 mice
6-8周龄的雌性C57BL/6小鼠,随机分组后,第0天在小鼠后肢和背部肌肉注射100μL用MOG35-55(1mg/mL)和弗氏完全佐剂(2mg/mL)制备的免疫乳剂,48h后腹腔注射百日咳毒素(200ng),诱导EAE模型的发生。第3天-第30天灌胃给予不同剂量的化合物,并根据临床症状对小鼠的EAE病情进程进行评分,考察药物对小鼠EAE进程的抑制作用并计算抑制率(抑制率=(1-(Mean AUC of clinical score(Test/Vehicle)))*100)。实验结果表明,在小鼠EAE模型上,相对于模型组,式I化合物(10mg/kg,qd)能够显著性抑制小鼠EAE的发展和进行,抑制率为61.37%,而DMF(15mg/kg,bid)的抑制率为41.39%。 显然,式I化合物(10mg/kg,qd)的抑制作用强于DMF(15mg/kg,bid)的抑制作用(图9)。Female C57BL/6 mice aged 6-8 weeks were randomly grouped. On day 0, 100μL of MOG35-55 (1mg/mL) and Freund's complete adjuvant (2mg/mL) were injected into the hind limbs and back muscles of the mice on day 0. The immune emulsion was injected with pertussis toxin (200ng) into the abdominal cavity 48h later to induce the development of EAE model. From the 3rd day to the 30th day, different doses of compounds were given by gavage, and the EAE progress of the mice was scored according to the clinical symptoms, the inhibitory effect of the drugs on the EAE progress of the mice was investigated and the inhibition rate was calculated (inhibition rate = (1- (Mean AUC of clinical score(Test/Vehicle)))*100). The experimental results show that in the mouse EAE model, compared with the model group, the compound of formula I (10mg/kg, qd) can significantly inhibit the development and progress of mouse EAE, the inhibition rate is 61.37%, while DMF (15mg/kg , bid) The inhibition rate was 41.39%. Obviously, the inhibitory effect of the compound of formula I (10 mg/kg, qd) is stronger than that of DMF (15 mg/kg, bid) (Figure 9).
六.化合物对侧脑室注射Aβ的阿尔兹海默症(AD)大鼠学习记忆的改善作用6. The compound improves the learning and memory of Alzheimer's disease (AD) rats injected with Aβ into the lateral ventricle
雄性wistar大鼠,12周龄,侧脑室手术注射凝聚态的寡聚体Aβ25-35(10nM)制备AD大鼠模型,第2天起开始灌胃给予不同剂量的化合物,以多奈哌齐作为对照药,10天后开始进行行为学实验(Morris水迷宫)以评价药物对大鼠学习记忆能力的改善作用。Morris水迷宫实验分为定位航行和空间探索两部分。第1天开始定位航行训练,共训练3天,每天连续重复训练两次。第4天测试最后一次逃避潜伏期,然后撤去水下平台,进行空间探索实验。Male wistar rats, 12 weeks old, were surgically injected with condensed oligomer Aβ25-35 (10 nM) into the lateral ventricle to prepare AD rat models. From the second day onwards, they were given different doses of compounds by gavage. Donepezil was used as the control drug. Ten days later, a behavioral experiment (Morris water maze) was started to evaluate the improvement effect of the drugs on the learning and memory ability of rats. The Morris water maze experiment is divided into two parts: positioning navigation and space exploration. Positioning and sailing training began on the first day, training for a total of 3 days, repeated training twice a day. On the 4th day, the test escaped the incubation period for the last time, and then the underwater platform was removed for space exploration experiments.
在Morris水迷宫实验中,在第4天时,假手术组动物与空白组动物相比,学习成绩未见显著变化,提示手术操作并未对大鼠学习记忆能力产生影响(p>0.05)。与假手术组相比,模型大鼠到达平台的潜伏期显著延长(p<0.001),表明AD动物模型制备成功。与模型组相比,式I化合物(15mg/kg,qd)和多奈哌齐(3mg/kg,qd)均可显著缩短大鼠到达平台潜伏期(图10);在第4天的空间探索实验中,药物治疗对动物跨越目标平台次数产生显著影响,与模型组相比,式I化合物和多奈哌齐组大鼠跨越次数显著增加(图11)。In the Morris water maze experiment, on the 4th day, animals in the sham operation group had no significant changes in academic performance compared with the animals in the blank group, suggesting that the operation did not affect the learning and memory abilities of rats (p>0.05). Compared with the sham operation group, the latency of model rats to reach the platform was significantly longer (p<0.001), indicating that the AD animal model was successfully prepared. Compared with the model group, the compound of formula I (15mg/kg, qd) and donepezil (3mg/kg, qd) can significantly shorten the latency of rats to reach the platform (Figure 10); in the space exploration experiment on day 4, the drug The treatment had a significant effect on the number of times the animals crossed the target platform. Compared with the model group, the number of crossings of the rats in the compound of formula I and donepezil group was significantly increased (Figure 11).
Morris水迷宫实验表明式I化合物(15mg/kg,qd)能够显著性地改善AD模型大鼠的学习记忆。Morris water maze experiment showed that the compound of formula I (15mg/kg, qd) can significantly improve the learning and memory of AD model rats.
七.化合物对6-OHDA诱导的帕金森病(PD)大鼠的行为学改善作用Seven. Compounds improve the behavior of rats with Parkinson's disease (PD) induced by 6-OHDA
雄性wistar大鼠,12周龄,采用中脑内侧前脑束定位注射6-OHDA以制备PD大鼠模型,6-OHDA注射21天后采用阿扑吗啡诱导旋转以验证模型是否成功,将成功旋转大鼠随机分组,并灌胃给予不同剂量的化合物,而假手术组和模型组则给予双蒸水,以左旋多巴(L-dopa)作为对照药,10天后进行行为学检测(包括转棒试验、爬杆实验和阿扑吗啡诱导的旋转实验),以评价待测化合物药效。Male wistar rats, 12 weeks old, were injected with 6-OHDA into the medial forebrain tract to prepare a PD rat model. 21 days after 6-OHDA injection, apomorphine was used to induce rotation to verify the success of the model. Mice were randomly divided into groups and given different doses of compound by gavage. The sham operation group and model group were given double distilled water, L-dopa was used as the control drug, and behavioral testing (including the rotating rod test) was performed after 10 days. , Pole climbing experiment and apomorphine-induced rotation experiment) to evaluate the efficacy of the test compound.
在转棒实验中,与假手术组相比,模型组大鼠跌落潜伏期显著缩短(p<0.001),提示其运动功能出现障碍,PD模型制备成功。与模型组相比,对照药左旋多巴(10mg/kg,qd)和式I化合物(10mg/kg,qd)可显著延长大鼠跌落潜伏期(图12);In the rotating rod experiment, compared with the sham operation group, the rats in the model group had a significantly shorter fall latency (p<0.001), indicating that their motor function was impaired and the PD model was successfully prepared. Compared with the model group, the control drugs levodopa (10mg/kg, qd) and the compound of formula I (10mg/kg, qd) can significantly prolong the incubation period of rats' fall (Figure 12);
在爬杆实验中,与假手术组相比,模型组大鼠爬杆时间显著延长(p<0.01),提示其运动功能出现障碍,PD模型制备成功。与模型组相比,对照药左旋多巴(10mg/kg,qd)和式I化合物(10mg/kg,qd)均可显著性缩短大鼠爬杆时间(图13)。In the rod climbing experiment, compared with the sham operation group, the rod climbing time of rats in the model group was significantly longer (p<0.01), indicating that their motor function was impaired and the PD model was successfully prepared. Compared with the model group, the control drug levodopa (10mg/kg, qd) and the compound of formula I (10mg/kg, qd) can significantly shorten the climbing time of rats (Figure 13).
在阿扑吗啡诱导PD大鼠旋转实验中,与假手术组相比,模型组大鼠旋转速度显著增加(p<0.01),提示其DA系统功能出现障碍,PD模型制备成功。与模型组相比,对照药左旋多巴(10mg/kg,qd)和式I化合物(10mg/kg,qd)均可显著性降低大鼠旋转速度(图14)。In the rotation experiment of PD rats induced by apomorphine, compared with the sham operation group, the rotation speed of rats in the model group was significantly increased (p<0.01), suggesting that the DA system function was impaired and the PD model was successfully prepared. Compared with the model group, the control drug levodopa (10mg/kg, qd) and the compound of formula I (10mg/kg, qd) can significantly reduce the rat's rotation speed (Figure 14).
综合上述结果,式I化合物可显著缓解由6-OHDA诱发的大鼠PD样行为学障碍。Based on the above results, the compound of formula I can significantly alleviate the PD-like behavioral disorder in rats induced by 6-OHDA.
八.化合物对急性脑动脉缺血再灌注损伤大鼠的神经保护作用8. The neuroprotective effect of the compound on acute cerebral artery ischemia-reperfusion injury in rats
SD大鼠经戊巴比妥钠腹腔注射麻醉后,采用线栓法以制备急性脑动脉缺血再灌注损伤模型(MCAO)。在缺血2h后,拔出造模线栓形成再灌注损伤,并于5min内静脉给予药物干预,以依达拉奉为阳性对照药。第2天继续给药。第3天采用Zea-Longa 5级标准评分对术后大鼠进行行为学评分(0分:正常,无神经功能缺损;1分:左侧前爪不能完全伸展,轻度神经功能缺损;2分:行走时,大鼠向左侧(瘫痪侧)转圈,中度神经功能缺损;3分:行走时,大鼠身体向左侧(瘫痪侧)倾倒,重度神经功能缺损;4分:不能自发行走,意识丧失)。在试验结束后,将大鼠麻醉,然后断头取脑,脑组织用多聚甲醛固定。在使用生理盐水清洗全脑后,将其放置于干净的培养皿中,并于-20℃冰箱中速冻30min。以脑前极与视交叉连线中点处为起点,每隔2mm切一片,将大脑切成5-6片,然后将脑切片放入1%TTC溶液中,37℃温育染色10~15min。用手术刀切下不着色梗死区组织,计算梗死区和全脑重量比,以此评价化合物对急性脑动脉缺血再灌注损伤大鼠的神经保护作用。行为学评分结果表明:在式I化合物(10mg/kg)干预后,SD大鼠的行为评分显著低于模型组,且低于对照药依达拉奉(6mg/kg)组(图15)。就梗死区/全脑重量比而言,依达拉奉组和式I化合物组均明显小于模型组。上述结果表明:依达拉奉和式I化合物均能有效地保护脑动脉缺血再灌注损伤大鼠的神经,且式I化合物的效果优于依达拉奉(图16)。After SD rats were anesthetized by intraperitoneal injection of sodium pentobarbital, a suture method was used to prepare an acute cerebral artery ischemia-reperfusion injury model (MCAO). After 2 hours of ischemia, the model thread plug was pulled out to cause reperfusion injury, and intravenous drug intervention was given within 5 minutes, with edaravone as the positive control drug. The administration was continued on the second day. On the third day, Zea-Longa 5-level standard scoring was used to score the postoperative rats' behavior (0 points: normal, no neurological deficit; 1 point: left front paw incomplete extension, mild neurological deficit; 2 points : When walking, the rat turns to the left side (paralyzed side), with moderate neurological deficit; 3 points: When walking, the rat's body falls to the left side (paralyzed side), with severe neurological deficit; 4 points: unable to walk spontaneously , Loss of consciousness). After the test, the rats were anesthetized, and then the brain was decapitated, and the brain tissue was fixed with paraformaldehyde. After washing the whole brain with normal saline, place it in a clean petri dish and quickly freeze it in a refrigerator at -20°C for 30 min. Starting from the midpoint of the line between the anterior pole of the brain and the optic chiasm, slice the brain every 2 mm, and cut the brain into 5-6 slices, then put the brain slices in 1% TTC solution, incubate at 37°C for 10-15 minutes . The non-stained infarct area tissue was cut with a scalpel, and the weight ratio of the infarct area to the whole brain was calculated to evaluate the neuroprotective effect of the compound on acute cerebral artery ischemia-reperfusion injury in rats. The behavioral score results showed that: after the intervention of the compound of formula I (10mg/kg), the behavior score of SD rats was significantly lower than that of the model group, and lower than the control drug edaravone (6mg/kg) group (Figure 15). In terms of the infarct area/whole brain weight ratio, both the edaravone group and the formula I compound group were significantly smaller than the model group. The above results indicate that both edaravone and the compound of formula I can effectively protect the nerves of rats with cerebral artery ischemia-reperfusion injury, and the effect of the compound of formula I is better than that of edaravone (Figure 16).
九.化合物致敏性试验9. Compound sensitization test
于实验前一天,将雄性豚鼠要与药物接触部位的毛发剃除。将0.1g受试药物(诱导剂量)与适量凡士林搅拌均匀,于第0天、第7天和第14天涂皮,每次诱导时间为6h,而对照组的豚鼠则用凡士林涂皮。距末次涂皮诱导14天后,于豚鼠诱导部位对侧皮肤涂皮给予0.08g受试药物(激发剂量),而于对照组的豚鼠的相应部位对侧皮肤仍用凡士林涂皮,6h后分别去掉给药组的受试药物和对照组的凡士林并清洗,观察有无皮肤反应,并对反应程度评分及拍照记录,统计各组致敏率。Control组无显著反应,DMF引起90%的豚鼠的皮肤明显红斑和水肿,式I化合物组无明显的皮肤反应,仅一只有轻微的水肿(图17)。结果表明DMF具有极强的致敏性,而式I化合物仅具有极弱的致敏性。On the day before the experiment, the male guinea pigs were shaved off the areas where they were in contact with the drug. Stir 0.1 g of the test drug (inducing dose) and appropriate amount of petroleum jelly, and smear the skin on the 0th, 7th and 14th day, each induction time is 6h, while the guinea pigs in the control group are smeared with petroleum jelly. 14 days after the last induction of smearing, 0.08g of the test drug (provocation dose) was applied to the contralateral skin of the guinea pig induction site, while the contralateral skin of the corresponding site of the guinea pig in the control group was still smeared with petroleum jelly, which was removed after 6 hours. The test drug in the administration group and the petroleum jelly in the control group were washed and washed to observe whether there was skin reaction, and the degree of reaction was scored and photographed and recorded, and the sensitization rate of each group was counted. There was no significant reaction in the Control group. DMF caused 90% of the guinea pigs to have obvious skin erythema and edema. The formula I compound group had no obvious skin reaction, only slight edema (Figure 17). The results show that DMF has extremely strong allergenicity, while the compound of formula I only has extremely weak allergenicity.

Claims (14)

  1. 一种式I化合物的晶型I,A crystal form I of a compound of formula I,
    Figure PCTCN2020075918-appb-100001
    Figure PCTCN2020075918-appb-100001
    其特征在于,其X-射线粉末衍射图在以下衍射角2θ处具有特征峰:8.82±0.2°、14.56±0.2°、16.82±0.2°、17.72±0.2°、20.22±0.2°、22.50±0.2°、26.24±0.2°、29.40±0.2°。It is characterized in that its X-ray powder diffraction pattern has characteristic peaks at the following diffraction angles 2θ: 8.82±0.2°, 14.56±0.2°, 16.82±0.2°, 17.72±0.2°, 20.22±0.2°, 22.50±0.2° , 26.24±0.2°, 29.40±0.2°.
  2. 根据权利要求1所述的晶型I,其特征在于,其X-射线粉末衍射图进一步在以下衍射角2θ处有特征峰:10.06±0.2°、24.22±0.2°、25.00±0.2°、26.76±0.2°、28.16±0.2°、30.56±0.2°、31.14±0.2°、33.74±0.2°、38.66±0.2°、40.20±0.2°、44.84±0.2°、46.04±0.2°。The crystalline form I of claim 1, wherein the X-ray powder diffraction pattern further has characteristic peaks at the following diffraction angles 2θ: 10.06±0.2°, 24.22±0.2°, 25.00±0.2°, 26.76± 0.2°, 28.16±0.2°, 30.56±0.2°, 31.14±0.2°, 33.74±0.2°, 38.66±0.2°, 40.20±0.2°, 44.84±0.2°, 46.04±0.2°.
  3. 根据权利要求1或2所述的晶型I,其特征在于,其X-射线粉末衍射谱图基本上如图1所示。The crystalline form I according to claim 1 or 2, characterized in that its X-ray powder diffraction spectrum is basically as shown in Fig. 1.
  4. 一种制备权利要求1~3任一项所述的晶型I的方法,所述方法选自下述方法中的任意一种:A method for preparing the crystal form I according to any one of claims 1 to 3, the method being selected from any of the following methods:
    方法(1),其包括以下步骤:Method (1), which includes the following steps:
    1)将式I化合物溶解于醇、酮、酯、乙腈、二氧六环中的任一溶剂中,溶清,蒸发析晶;所述醇为C2-C4醇;所述酮为C3-C6酮;所述酯为C3-C6酯;所述溶解的温度为25~45℃;所述式I化合物与溶剂的质量体积比为(mg/ml)10:1.5~30;1) Dissolve the compound of formula I in any solvent of alcohol, ketone, ester, acetonitrile and dioxane, dissolve it, and evaporate and crystallize; the alcohol is a C2-C4 alcohol; the ketone is a C3-C6 Ketone; the ester is a C3-C6 ester; the dissolution temperature is 25-45°C; the mass-volume ratio of the compound of formula I to the solvent is (mg/ml) 10:1.5-30;
    2)过滤,得晶型I;2) Filter to obtain crystal form I;
    方法(2),其包括以下步骤:Method (2), which includes the following steps:
    1)将式I化合物溶解于醇、酮、酯、乙腈、二氧六环、二甲基乙酰胺、二甲基甲酰胺、二甲亚砜中的任一溶剂中,降温析晶;所述醇为C2-C4醇;所述酮为C3-C6酮;所述酯为C3-C6酯;所述溶解的温度为40~70℃;所述式I化合物与溶剂的质量体积比为(mg/ml)10:0.4~3;所述析晶的温度为0~20℃;1) The compound of formula I is dissolved in any solvent of alcohol, ketone, ester, acetonitrile, dioxane, dimethylacetamide, dimethylformamide, and dimethylsulfoxide, and the temperature is lowered to crystallize; The alcohol is a C2-C4 alcohol; the ketone is a C3-C6 ketone; the ester is a C3-C6 ester; the dissolution temperature is 40-70°C; the mass-volume ratio of the compound of formula I to the solvent is (mg /ml) 10: 0.4~3; the temperature of said crystallization is 0~20℃;
    或将式I化合物溶解于乙醇、异丙醇、二氧六环、丙酮、乙腈中的任一有机溶剂与水的混合溶剂中,降温析晶;所述溶解的温度为40~70℃;所述有机溶剂与水的体积比为1: 0.1~9;所述式I化合物与混合溶剂的质量体积比为(mg/ml)10:1~1.5;所述析晶的温度为0~5℃;Or the compound of formula I is dissolved in a mixed solvent of any organic solvent among ethanol, isopropanol, dioxane, acetone, and acetonitrile and water, and the temperature is lowered to crystallize; the dissolution temperature is 40-70°C; The volume ratio of the organic solvent to water is 1:0.1-9; the mass-volume ratio of the compound of formula I and the mixed solvent is (mg/ml) 10:1-1.5; the crystallization temperature is 0-5°C ;
    2)过滤,得晶型I。2) Filter to obtain crystal form I.
  5. 一种式I化合物的晶型II,A crystal form II of the compound of formula I,
    Figure PCTCN2020075918-appb-100002
    Figure PCTCN2020075918-appb-100002
    其特征在于,其X-射线粉末衍射图在以下衍射角2θ处有特征峰:7.34±0.2°、14.72±0.2°、16.98±0.2°、20.30±0.2°、22.16±0.2°、25.84±0.2°、38.44±0.2°。It is characterized in that its X-ray powder diffraction pattern has characteristic peaks at the following diffraction angles 2θ: 7.34±0.2°, 14.72±0.2°, 16.98±0.2°, 20.30±0.2°, 22.16±0.2°, 25.84±0.2° , 38.44±0.2°.
  6. 根据权利要求5所述的晶型II,其特征在于,其X-射线粉末衍射图进一步在以下衍射角2θ处有特征峰:9.20±0.2°、13.90±0.2°、18.46±0.2°、23.74±0.2°、29.72±0.2°、34.42±0.2°。The crystal form II according to claim 5, wherein the X-ray powder diffraction pattern further has characteristic peaks at the following diffraction angles 2θ: 9.20±0.2°, 13.90±0.2°, 18.46±0.2°, 23.74± 0.2°, 29.72±0.2°, 34.42±0.2°.
  7. 根据权利要求5或6所述的晶型II,其特征在于,其X-射线粉末衍射谱图基本上如图4所示。The crystal form II according to claim 5 or 6, characterized in that its X-ray powder diffraction spectrum is basically as shown in Fig. 4.
  8. 一种制备权利要求5~7任一项所述的晶型II的方法,所述方法选自下述方法中的任意一种:A method for preparing the crystal form II according to any one of claims 5 to 7, the method is selected from any one of the following methods:
    方法(1),其包括以下步骤:Method (1), which includes the following steps:
    1)将式I化合物加入CH 2Cl 2中,回流溶解;所述式I化合物与CH 2Cl 2的质量体积比(mg/ml)为10:1.4~1.6; 1) Add the compound of formula I to CH 2 Cl 2 and dissolve under reflux; the mass-volume ratio (mg/ml) of the compound of formula I to CH 2 Cl 2 is 10:1.4-1.6;
    2)将上述溶液加至-15~-10℃的乙酸乙酯(EA)中;所述EA与CH 2Cl 2的体积比为0.9~1.1:1; 2) Add the above solution to ethyl acetate (EA) at -15~-10°C; the volume ratio of the EA to CH 2 Cl 2 is 0.9-1.1:1;
    3)搅拌析晶,过滤,得晶型II;3) Stir and crystallize, filter to obtain crystal form II;
    方法(2),其包括以下步骤:Method (2), which includes the following steps:
    1)将式I化合物溶解于二氧六环中;所述溶解的温度为45~60℃;所述式I化合物与二氧六环的质量体积比(mg/ml)为10:1.4~1.6;1) The compound of formula I is dissolved in dioxane; the temperature of the dissolution is 45-60°C; the mass-volume ratio (mg/ml) of the compound of formula I to dioxane is 10:1.4-1.6 ;
    2)将上述溶液加至4~6℃的正庚烷中;所述正庚烷与二氧六环的体积比为3~3.5:1;2) Add the above solution to n-heptane at 4-6°C; the volume ratio of n-heptane to dioxane is 3 to 3.5:1;
    3)搅拌析晶,过滤,得晶型II。3) Stir to crystallize, filter to obtain crystal form II.
  9. 一种药物组合物,其中包括有效剂量的权利要求1~3中任一项所述的晶型I或权 利要求5~7中任一项所述的晶型II。A pharmaceutical composition comprising an effective dose of the crystal form I according to any one of claims 1 to 3 or the crystal form II according to any one of claims 5 to 7.
  10. 权利要求1~3中任一项所述的晶型I或权利要求5~7中任一项所述的晶型II,或权利要求9所述的药物组合物在制备用于治疗与Nrf2激活相关的疾病的药物中的应用,其中所述与Nrf2激活相关的疾病为脑卒中、神经退行性疾病、糖尿病、糖尿病肾病、冠心病、动脉粥样硬化或非酒精性脂肪肝。The crystal form I according to any one of claims 1 to 3 or the crystal form II according to any one of claims 5 to 7, or the pharmaceutical composition according to claim 9 is prepared for treatment and activation of Nrf2 Application in medicine for related diseases, wherein the diseases related to Nrf2 activation are stroke, neurodegenerative diseases, diabetes, diabetic nephropathy, coronary heart disease, atherosclerosis or non-alcoholic fatty liver.
  11. 权利要求1~3中任一项所述的晶型I或权利要求5~7中任一项所述的晶型II,或权利要求9所述的药物组合物在制备用于治疗脑卒中的药物中的应用。The crystal form I according to any one of claims 1 to 3 or the crystal form II according to any one of claims 5 to 7, or the pharmaceutical composition according to claim 9 is prepared for the treatment of stroke Application in medicine.
  12. 权利要求1~3中任一项所述的晶型I或权利要求5~7中任一项所述的晶型II,或权利要求9所述的药物组合物在制备用于治疗神经退行性疾病的药物中的应用。The crystal form I according to any one of claims 1 to 3 or the crystal form II according to any one of claims 5 to 7, or the pharmaceutical composition according to claim 9 is prepared for the treatment of neurodegeneration Application of medicines for diseases.
  13. 根据权利要求10或12所述的应用,其中所述的神经退行性疾病选自多发性硬化症(MS)、阿尔茨海默氏病(AD)、帕金森病(PD)、亨廷顿氏病(HD)、肌萎缩侧索硬化(ALS)、弗里德赖希氏共济失调(FRDA)、脊髓性肌萎缩(SMA)、视神经脊髓炎(NMO)和脊髓小脑性共济失调(SCA)。The use according to claim 10 or 12, wherein the neurodegenerative disease is selected from multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease ( HD), Amyotrophic Lateral Sclerosis (ALS), Friedreich’s Ataxia (FRDA), Spinal Muscular Atrophy (SMA), Optic Neuromyelitis (NMO) and Spinocerebellar Ataxia (SCA).
  14. 权利要求1~3中任一项所述的晶型I或权利要求5~7中任一项所述的晶型II,或权利要求9所述的药物组合物在制备用于治疗与免疫调节相关的疾病的药物中的应用,其中所述与免疫调节相关的疾病优选自银屑病、类风湿关节炎、系统性红斑狼疮、桥本氏甲状腺炎、移植排斥和炎性疾病。The crystal form I according to any one of claims 1 to 3 or the crystal form II according to any one of claims 5 to 7, or the pharmaceutical composition according to claim 9 is prepared for treatment and immunomodulation Application in medicines for related diseases, wherein the diseases related to immune regulation are preferably selected from psoriasis, rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, transplant rejection and inflammatory diseases.
PCT/CN2020/075918 2019-02-22 2020-02-20 CRYSTALLINE FORM OF (E)-α,β-UNSATURED AMIDE COMPOUND, PREPARATION METHOD THEREFOR, AND USE THEREOF WO2020169054A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910132052.7A CN111606828B (en) 2019-02-22 2019-02-22 (E) Crystal forms of alpha, beta-unsaturated amide compound, preparation method and application thereof
CN201910132052.7 2019-02-22

Publications (1)

Publication Number Publication Date
WO2020169054A1 true WO2020169054A1 (en) 2020-08-27

Family

ID=72144321

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/075918 WO2020169054A1 (en) 2019-02-22 2020-02-20 CRYSTALLINE FORM OF (E)-α,β-UNSATURED AMIDE COMPOUND, PREPARATION METHOD THEREFOR, AND USE THEREOF

Country Status (2)

Country Link
CN (1) CN111606828B (en)
WO (1) WO2020169054A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH326535A (en) * 1954-04-29 1957-12-31 Sandoz Ag Process for the preparation of dicarboxamides
JPH01156958A (en) * 1987-09-02 1989-06-20 Nippon Shokubai Kagaku Kogyo Co Ltd Method for transporting and storing acrylonitrile solution of maleimides
CN103889412A (en) * 2011-08-04 2014-06-25 尼尔米斯有限公司 Novel aniline derivatives and use thereof
CN103998035A (en) * 2011-12-19 2014-08-20 阿雷斯贸易股份有限公司 Pharmaceutical compositions comprising glitazones and NRF2 activators
CN104487415A (en) * 2013-05-08 2015-04-01 杨永亮 Maleic amide compound, preparation method therefor and application thereof
WO2019042301A1 (en) * 2017-08-29 2019-03-07 浙江海正药业股份有限公司 (E)-α,β-UNSATURATED AMIDE COMPOUND AND PREPARATION METHOD AND USE THEREOF

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH326535A (en) * 1954-04-29 1957-12-31 Sandoz Ag Process for the preparation of dicarboxamides
JPH01156958A (en) * 1987-09-02 1989-06-20 Nippon Shokubai Kagaku Kogyo Co Ltd Method for transporting and storing acrylonitrile solution of maleimides
CN103889412A (en) * 2011-08-04 2014-06-25 尼尔米斯有限公司 Novel aniline derivatives and use thereof
CN103998035A (en) * 2011-12-19 2014-08-20 阿雷斯贸易股份有限公司 Pharmaceutical compositions comprising glitazones and NRF2 activators
CN104487415A (en) * 2013-05-08 2015-04-01 杨永亮 Maleic amide compound, preparation method therefor and application thereof
WO2019042301A1 (en) * 2017-08-29 2019-03-07 浙江海正药业股份有限公司 (E)-α,β-UNSATURATED AMIDE COMPOUND AND PREPARATION METHOD AND USE THEREOF

Also Published As

Publication number Publication date
CN111606828B (en) 2023-07-28
CN111606828A (en) 2020-09-01

Similar Documents

Publication Publication Date Title
JP5841672B2 (en) N-benzylaniline derivatives and uses thereof
KR20140129136A (en) Morpholinoalkyl fumarate compounds, pharmaceutical compositions, and methods of use
US20210122740A1 (en) Solid forms of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile
CN108602761B (en) Salts of tetracyclines
US9789114B2 (en) Compounds as HIF-1α inhibitors and manufacturing process thereof
RU2624446C9 (en) Tricyclic compounds, compositions containing these compounds and their use
EP3838887B1 (en) 2-(1-acyloxypentyl) benzoic acid salt formed by basic amino acid or aminoguanidine, preparation method therefor and uses thereof
US11859019B2 (en) Bicyclic peptidyl inhibitor of tumor necrosis factor-alpha
WO2023165094A1 (en) Hydroxypentyl benzoic acid diester compound, and preparation method therefor and use thereof
JP2018115218A (en) Crystalline forms of neurotrophin mimetic compounds and their salts
WO2019129249A1 (en) Triterpenoid-containing pharmaceutical composition and use thereof
CN114206878B (en) Wu Pati Ni crystal form and preparation method and application thereof
WO2020169054A1 (en) CRYSTALLINE FORM OF (E)-α,β-UNSATURED AMIDE COMPOUND, PREPARATION METHOD THEREFOR, AND USE THEREOF
CA2086441A1 (en) N-acyl derivatives of aminoalcohols with polycarboxylic acids able to modulate mast cells in inflammatory processes having meuroimmunogenic origin
WO2024060373A1 (en) S-(+)-flurbiprofen salt, method for preparing same, pharmaceutical composition thereof, and use thereof
CN114644618A (en) Salt form of isoquinolinone compound as ROCK protein kinase inhibitor and preparation method thereof
JP2015535842A (en) Crystal forms of neurotrophin mimetic compounds and their salts
CN111065619B (en) (E) -alpha, beta-unsaturated amide compound, preparation method and application thereof
CN108794386B (en) Pharmaceutical compositions comprising mono-tartrate salts and uses thereof
US20180030058A1 (en) Crystal of 5-( (2-(6-amino)-9H-purin-9-yl) ethyl) amino) pentan-1-o1
WO2017028762A1 (en) Crystal form of naphthalene cyclic compound
CN106478616B (en) Crystalline form of GPR40 agonist and preparation method thereof
AU2019218153A1 (en) Therapeutic drug for neurodegenerative disease and application thereof
US9359314B2 (en) Thiazine amide derivative and pharmaceutical composition and use thereof
CN109422723B (en) Crystal form of intestinal 2B type sodium phosphate cotransporter inhibitor and preparation method thereof

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: 20759678

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20759678

Country of ref document: EP

Kind code of ref document: A1