WO2023061372A1 - Malate salt of xanomeline derivative, crystalline form a, and preparation method therefor and use thereof - Google Patents

Malate salt of xanomeline derivative, crystalline form a, and preparation method therefor and use thereof Download PDF

Info

Publication number
WO2023061372A1
WO2023061372A1 PCT/CN2022/124662 CN2022124662W WO2023061372A1 WO 2023061372 A1 WO2023061372 A1 WO 2023061372A1 CN 2022124662 W CN2022124662 W CN 2022124662W WO 2023061372 A1 WO2023061372 A1 WO 2023061372A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound
malate
crystal form
preparation
Prior art date
Application number
PCT/CN2022/124662
Other languages
French (fr)
Chinese (zh)
Inventor
刘飞
吴刚
李先朝
陈庆
王晓波
赵欣
冷超群
Original Assignee
南京迈诺威医药科技有限公司
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 南京迈诺威医药科技有限公司 filed Critical 南京迈诺威医药科技有限公司
Publication of WO2023061372A1 publication Critical patent/WO2023061372A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • 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
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C51/00Preparation of carboxylic acids or their salts, halides or anhydrides
    • C07C51/41Preparation of salts of carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C51/00Preparation of carboxylic acids or their salts, halides or anhydrides
    • C07C51/42Separation; Purification; Stabilisation; Use of additives
    • C07C51/43Separation; Purification; Stabilisation; Use of additives by change of the physical state, e.g. crystallisation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C59/00Compounds having carboxyl groups bound to acyclic carbon atoms and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C59/235Saturated compounds containing more than one carboxyl group
    • C07C59/245Saturated compounds containing more than one carboxyl group containing hydroxy or O-metal groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the present invention relates to the field of medicinal chemistry, in particular to a malate salt of a jenomeline derivative, a crystal form A and a preparation method thereof, which also includes the preparation of the malate salt and a crystal form A of a jenomeline derivative
  • a malate salt of a jenomeline derivative a malate salt of a jenomeline derivative
  • a crystal form A a preparation method thereof, which also includes the preparation of the malate salt and a crystal form A of a jenomeline derivative
  • Neurotransmitters are chemical messengers secreted by neurons to facilitate the flow of information and communicate with other cells in the central and peripheral nervous systems, such as muscle or similar nerve cells.
  • Acetylcholine one of the key neurotransmitters in the brain, has two distinct receptor classes: muscarinic receptors (M receptors, G protein-coupled receptors) and nicotinic receptors (N receptors , ion channel receptors).
  • the M receptor family includes five subtypes from M1 to M5, all of which are expressed in the brain and peripheral tissues and play many key physiological roles in cognition, behavior, sensory, motor and autonomic processes. Disruption of M-type receptor signaling leads to memory impairment and cognitive impairment, triggers a variety of diseases including schizophrenia and Alzheimer's disease (AD), and exacerbates psychosis. On the contrary, third-party preclinical and clinical data show that the enhancement of M-type receptor signaling can improve these symptoms. In addition, M-type receptors, especially M1, M2, and M4 receptors, are also considered to be related to analgesia.
  • Xanomeline is a partial agonist of muscarinic receptors, which can produce agonistic effects on all five subtypes of muscarinic receptors without selectivity. Jointly developed and marketed by Eli Lilly and Novo Nordisk, it is mainly used clinically for the treatment of Alzheimer's disease.
  • the chemical name of Normeline is 3-[(4-hexyloxy)-1,2,5 -Thiadiazol-3-yl]-1,2,5,6-tetrahydro-1-picoline, the chemical structure is as follows:
  • Patent document CN94192681.8 discloses that oxalate can be converted into oxalate, but oxalate has potential side effects on the patient's kidney function, so it is not suitable for medicine, especially when treating the elderly. And it is further disclosed that in the series of twelve kinds of pharmaceutically acceptable acids (the name of the specific pharmaceutically acceptable acid is not disclosed), only zomeline tartrate has good bioavailability, good handling properties and reproducible crystal form .
  • the screening of drug salt form is a difficult semi-empirical choice, and the hygroscopicity of the drug will seriously affect the fluidity of the drug, and even affect the stability of the drug.
  • the solubility of drugs has a crucial impact on the preparation of pharmaceuticals, drug dissolution, absorption and so on. But how to improve the solubility of the drug, without the expense of the hygroscopicity of the drug, it is difficult to obtain a candidate drug salt with suitable stability, solubility and hygroscopicity.
  • Xanomeline as an M-receptor activator has shown encouraging therapeutic efficacy in the clinic for the treatment of psychosis and related behavioral symptoms in patients with schizophrenia and AD, but its potential has been limited by cholinergic side effects, including salivation , nausea, dizziness, etc., which are thought to be due to stimulation of M receptors in peripheral nervous tissue.
  • the metabolism of Zanameline in the human body is complex and unpredictable, and Zanameline lacks selectivity for muscarinic receptor subtypes, thus causing difficulties in the drug development of Zanameline.
  • the object of the present invention is to provide a malate salt of a zanmeline derivative shown in formula I, which can be used for the preparation of medicines for preventing or treating central nervous system disorders and the dissolution of this derivative Improved ability, stable storage, convenient administration, improved drug metabolism, drug toxicity and side effects, and high patient compliance during administration.
  • Another object of the present invention is to provide a crystal form A of the malate salt of phenomeline derivatives shown in formula I, the crystal form A can have excellent thermodynamic stability and mechanical stability, good repeatability, Suitable for commercial mass production.
  • the present invention provides a malate salt of a compound of formula I.
  • the malate salt of the compound of formula I above is shown in formula II, wherein x is selected from 0.5-2.
  • the above-mentioned x is 0.5, 1.0, 1.5, 2.0.
  • x in the compound of formula II above is 1.0.
  • the present invention provides the A crystal form of the malate salt of the compound of formula I, whose X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 12.007 ⁇ 0.2°, 14.827 ⁇ 0.2°, 16.796 ⁇ 0.2° , 20.012 ⁇ 0.2° and 21.942 ⁇ 0.2°.
  • the crystal form A of the malate salt of the compound of formula I is the compound of formula II with x being 1.0, and the malic acid therein is L-malic acid.
  • the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2 ⁇ angles: 12.007 ⁇ 0.2°, 14.827 ⁇ 0.2°, 16.796 ⁇ 0.2°, 20.012 ⁇ 0.2°, 21.942 ⁇ 0.2 °, 22.473 ⁇ 0.2° and 25.333 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2 ⁇ angles: 12.007 ⁇ 0.2°, 13.288 ⁇ 0.2°, 14.827 ⁇ 0.2°, 16.796 ⁇ 0.2°, 20.012 ⁇ 0.2 °, 21.942 ⁇ 0.2°, 22.473 ⁇ 0.2°, and 25.333 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2 ⁇ angles: 12.007 ⁇ 0.2°, 13.288 ⁇ 0.2°, 14.827 ⁇ 0.2°, 15.399 ⁇ 0.2°, 16.796 ⁇ 0.2 °, 20.012 ⁇ 0.2°, 20.327 ⁇ 0.2°, 21.942 ⁇ 0.2°, 22.473 ⁇ 0.2°, and 25.333 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2 ⁇ angles: 12.007 ⁇ 0.2°, 13.288 ⁇ 0.2°, 14.827 ⁇ 0.2°, 15.399 ⁇ 0.2°, 16.796 ⁇ 0.2 °, 20.012 ⁇ 0.2°, 20.327 ⁇ 0.2°, 21.942 ⁇ 0.2°, 22.473 ⁇ 0.2°, 24.996 ⁇ 0.2°, 25.333 ⁇ 0.2°, and 27.718 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2 ⁇ angles: 12.007 ⁇ 0.2°, 12.755 ⁇ 0.2°, 13.288 ⁇ 0.2°, 13.903 ⁇ 0.2°, 14.827 ⁇ 0.2 °, 15.399 ⁇ 0.2°, 16.796 ⁇ 0.2°, 17.686 ⁇ 0.2°, 18.906 ⁇ 0.2°, 20.012 ⁇ 0.2°, 20.327 ⁇ 0.2°, 21.942 ⁇ 0.2°, 22.473 ⁇ 0.2°, 24.996 ⁇ 0.2°, 25.333 ⁇ 0.2 ° and 27.718 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2 ⁇ angles: 12.007 ⁇ 0.2°, 12.755 ⁇ 0.2°, 13.288 ⁇ 0.2°, 13.903 ⁇ 0.2°, 14.827 ⁇ 0.2 °, 15.399 ⁇ 0.2°, 16.796 ⁇ 0.2°, 17.686 ⁇ 0.2°, 18.906 ⁇ 0.2°, 20.012 ⁇ 0.2°, 20.327 ⁇ 0.2°, 20.660 ⁇ 0.2°, 21.942 ⁇ 0.2°, 22.473 ⁇ 0.2°, 24.996 ⁇ 0.2 °, 25.333 ⁇ 0.2°, 27.382 ⁇ 0.2°, and 27.718 ⁇ 0.2°.
  • the above crystal form A has an XRPD pattern as shown in FIG. 1 .
  • the above crystal form A has a DSC spectrum substantially as shown in FIG. 2 .
  • the differential scanning calorimetry curve of the above crystal form A has an endothermic peak at 111.03 ⁇ 3°C.
  • the above crystal form A has a TGA curve substantially as shown in FIG. 3 .
  • the present invention also provides a method for preparing malate of the compound of formula I, comprising reacting the compound of formula I with malic acid.
  • the preparation method of the A crystal form of the compound malate of the above formula I comprises the following steps:
  • the solvent is selected from ethanol, isopropanol, n-propanol, butanol, acetone, ethyl acetate, n-heptyl
  • the solvent is selected from ethanol, isopropanol, n-propanol, butanol, acetone, ethyl acetate, n-heptyl
  • One or more mixtures of alkanes, acetonitrile, tetrahydrofuran preferably one or more mixtures of ethanol, isopropanol, ethyl acetate, n-heptane; or
  • the first solvent is selected from one or more mixtures of methanol, ethanol, isopropanol, n-butanol;
  • the second solvent is selected from methyl tert-butyl ether, esters, acetonitrile, alkanes
  • the mixture of one or more than one of them means that the esters are selected from ethyl acetate and/or, the lipids are, for example, isopropyl acetate; the alkanes are selected from n-heptane and/or n-hexane.
  • the heating temperature is 50-80°C, such as 60-70°C.
  • the compound of formula I, malic acid and the solvent used to dissolve the compound or the first solvent are mixed and then stirred.
  • the stirring time may be 1-5 hours, preferably 2-3 hours.
  • the weight/volume ratio (g/mL) of the compound of formula I to the solvent used to dissolve the compound or the first solvent is 1:4-10, preferably 1:6-8, such as 1:6, 1:7 , 1:8.
  • the cooling may be to 0-30°C, preferably to 2-8°C, for example to 5°C. It can be filtered once before cooling.
  • stirring is carried out during the cooling process, and the stirring time may be 6 to 24 hours, preferably 18 hours.
  • the step of washing with a solvent is also included before drying, and the washing solvent is selected from ethanol, isopropanol, n-propanol, One or a mixture of acetone, ethyl acetate, acetonitrile, tetrahydrofuran.
  • the molar ratio of the compound of formula I to malic acid is 1:0.9-1.3, preferably, the molar ratio of the compound of formula I to malic acid is 1:1-1.1, for example 1: 1. 1:1.05, 1:1.1.
  • the present invention provides a pharmaceutical composition, the pharmaceutical composition comprising the malate of the compound of formula I or the malate A crystal form of the compound of formula I, and optional pharmaceutically acceptable excipients agent.
  • the present invention provides the malate of the compound of formula I, the malate A crystal form of the compound of formula I above, or the pharmaceutical composition comprising the malate of the compound of formula I, or the malate of the compound of formula I Use of the pharmaceutical composition of crystal form A in the preparation of medicines for treating disorders of the central nervous system.
  • the disorders of the central nervous system include but are not limited to schizophrenia, Alzheimer's disease, Parkinson's disease, depression, movement disorders, drug addiction, pain and neurodegeneration (eg Dow disease or synucleinopathies).
  • the central nervous system disorder is schizophrenia.
  • the present invention provides a method for treating or preventing central nervous system disorders in mammals (such as humans), the method comprising administering a therapeutically effective amount of malic acid of a compound of formula I to mammals (such as humans) salt, malate A crystal form of the compound of formula I, a pharmaceutical composition comprising the malate salt of the compound of formula I and a pharmaceutical composition comprising the malate A crystal form of the compound of formula I.
  • the disorders of the central nervous system include but are not limited to schizophrenia, Alzheimer's disease, Parkinson's disease, depression, movement disorders, drug addiction, pain and neurodegeneration (eg Dow disease or synucleinopathies).
  • the central nervous system disorder is schizophrenia.
  • the present invention provides the malate of the compound of formula I for the first time; the malate of the compound of formula I has at least one aspect such as physical stability, solubility, hygroscopicity, biological activity, safety, bioavailability, toxic side effect Excellent effect.
  • the crystal form A of the malate salt of the compound of formula I has good stability, low hygroscopicity, good water solubility, small batch-to-batch variation, and good prospects for making a drug.
  • the preparation process of the salt form and crystal form of the present invention is simple and suitable for industrial production.
  • Fig. 1 is the XRPD pattern of A crystal form of compound L-malate of formula I;
  • Fig. 2 is the DSC spectrum of the A crystal form of formula I compound L-malate
  • Fig. 3 is the TGA spectrum of the A crystal form of formula I compound L-malate
  • Fig. 4 is the XRPD patterns of crystal form A of compound L-malate of formula I at high temperature (60° C.) for 0 days, 6 days, 14 days, and 30 days;
  • Fig. 5 is the XRPD patterns of crystal form A of compound L-malate of formula I under high humidity (RH92.5%) conditions at 0 days, 6 days, 14 days, and 30 days;
  • Fig. 6 is the XRPD pattern of crystal form A of L-malate of the compound of formula I under light conditions (4500 ⁇ 500Lux) at 0 days, 6 days, 14 days and 30 days.
  • Fig. 7 is the pharmacokinetic curve of the compound of formula I in rat plasma after oral administration of crystal form A of compound L-malate of formula I (sample of Example 2) and compound of formula I (sample of Example 1).
  • rt stands for room temperature
  • aq stands for aqueous solution
  • DCM dichloromethane
  • THF tetrahydrofuran
  • MeOH stands for methanol
  • EtOH stands for ethanol
  • IPA isopropanol
  • DSC differential scanning calorimeter
  • TGA thermogravimetric analysis
  • 1 H-NMR hydrogen nuclear magnetic resonance spectrum
  • XRPD X-ray powder diffraction
  • eq stands for molar equivalent.
  • Powder X-ray diffraction also known as "X-ray powder diffraction", X-ray powder diffractometer, XRPD
  • Test method About 10-20 mg of sample is used for XRPD detection.
  • Phototube voltage 40kV
  • phototube current 40mA
  • the diffraction pattern obtained from a crystalline compound is often characteristic for a particular crystal, where the relative intensities of the bands (especially at low angles) may vary. Varies due to the effect of preferred orientation due to differences in crystallization conditions, particle size, and other measurement conditions. Therefore, the relative intensity of the diffraction peaks is not characteristic of the targeted crystal, and when judging whether it is the same as a known crystal, more attention should be paid to the relative positions of the peaks rather than their relative intensities. Furthermore, for any given crystal there may be slight errors in the position of the peaks, as is well known in the art of crystallography.
  • the position of the peak can move, and the measurement error of the 2 ⁇ value is sometimes about ⁇ 0.2°. Therefore, this error should be taken into account when determining each crystal structure.
  • the peak positions of their XRPD spectra are similar on the whole, and the relative intensity error may be large.
  • Measurement variance associated with such X-ray powder diffraction analysis results arises from a variety of factors including: (a) errors in sample preparation (e.g., sample height), (b) instrument errors, (c) calibration differences, ( d) operator errors (including errors in determining peak positions), and (e) properties of the species (eg, preferred orientation errors). Calibration errors and sample height errors often cause all peaks to shift in the same direction. Small differences in sample height will result in large shifts in XRPD peak positions when using flat supports. Systematic studies have shown that a sample height difference of 1 mm can result in a peak shift in 2 ⁇ of up to 1°.
  • shifts can be identified from the X-ray diffraction pattern and can be eliminated by compensating for them (using a system calibration factor for all peak position values) or recalibrating the instrument. Measurement errors from different instruments can be corrected for by applying system calibration factors to make peak positions consistent, as described above.
  • Differential thermal analysis also known as “differential scanning calorimetry”, Differential Scanning Calorimeter, DSC) method
  • Test method Take a sample ( ⁇ 1mg) and place it in a DSC aluminum pot for testing. Under the condition of 50mL/ minN2 , heat the sample from 25°C to 300°C at a heating rate of 10°C/min.
  • DSC differential scanning calorimetry
  • the error of thermal transition temperature and melting point is typically within about 5°C, usually within about 3°C, when we say that a compound has a given DSC peak or melting point, this means ⁇ 5°C of the DSC peak or melting point.
  • DSC provides an auxiliary method for distinguishing between different crystals. Different crystalline forms can be identified based on their different transition temperature characteristics. It should be pointed out that for mixtures, the DSC peak or melting point may vary in a larger range. In addition, since the melting process of the substance is accompanied by decomposition, the melting temperature is related to the heating rate.
  • TGA Thermal Gravimetric Analyzer
  • Test method Take a sample (2 ⁇ 5mg) and place it in a TGA platinum pot for testing. Under the condition of 25mL/ minN2 , heat the sample from room temperature to 350°C at a heating rate of 10°C/min.
  • the instrument used is Waters ARC 2998HPLC; Chromatographic column: Agilent Poroshell bonus-RP 4.6 ⁇ 100mm, 2.7 ⁇ m;
  • the measurement conditions are as follows:
  • Mobile phase A 0.2% phosphoric acid aqueous solution
  • the inventor found that the hygroscopicity of the drug will seriously affect the fluidity of the drug, and even affect the stability of the drug.
  • the solubility of drugs has a crucial impact on the preparation of pharmaceuticals, drug dissolution, absorption and so on. But how to improve the solubility of the drug, without the expense of the hygroscopicity of the drug, it is difficult to obtain a drug candidate salt with suitable drug stability, solubility and hygroscopicity.
  • the inventor unexpectedly found that the malate of the compound of formula I has a higher melting point as determined by DSC, and the stability experiment confirmed that the malate of the compound of formula I has higher stability.
  • the malate salt of the compound of formula I has low hygroscopicity and moderate solubility.
  • the combination of high stability, low hygroscopicity and appropriate solubility of compound malate of formula I is unexpected, which makes compound malate of formula I (especially compound L-malate of formula I) suitable for the preparation of oral solid dosage forms , especially the preparation of tablets.
  • solubility of the active ingredient is of great importance for the choice of dosage form because the dosage form has a direct effect on bioavailability.
  • it is generally considered to be advantageous to have a higher solubility of the active ingredient because it leads to increased bioavailability.
  • the inventors have discovered the A crystal form of malate of the compound of formula I, which has advantages in at least one of physical stability, thermodynamic stability and mechanical stability; the prepared formula I compound malate
  • the crystal form A of the acid salt has a suitable crystal size and is suitable for industrial production and preparation.
  • a crystalline form of malate A of the compound of formula I can significantly improve the water solubility of the compound of formula I, and has good stability and extremely low hygroscopicity, which is beneficial for making oral solid preparations.
  • the preparation made of malate of the compound of formula I can maintain the effective physiological concentration of the free base of formula I in the body for a long time and is convenient to administer, so it has the advantages of improving the compliance of patients and the like.
  • 3-Pyridinecarbaldehyde (6.00g, 56.02mmol, 1.0eq), glacial acetic acid (3.36g, 56.02mmol, 1.0eq) and 6mL of pure water were successively added into a 100mL single-port reaction flask, and stirred and mixed at room temperature.
  • step 3 3-(4-chloro-1,2,5-thiadiazol-3-yl)pyridine (intermediate 3)
  • Step 5 Preparation of iodide-1-deuteromethyl-3-(4-hexyloxy-1,2,5-thiadiazol-3-yl)pyridine (intermediate 5)
  • Step 6 the preparation of deuterated phenomeline (compound of formula I)
  • the obtained white solid was submitted for inspection by XRPD, DSC and TGA. After testing, it was found that the obtained white solid existed in the form of crystals, and the obtained crystal form was named as the compound of formula I L-malate A crystal form, referred to as A crystal form.
  • the XRPD pattern, DSC pattern and TGA pattern of the obtained crystal form A are basically shown in Fig. 1 , Fig. 2 and Fig. 3 respectively.
  • the peak positions and intensities of the characteristic peaks are shown in Table 2;
  • the diffraction angle data of the XRPD pattern of the obtained crystal form are basically shown in Table 3, and the error range of the 2 ⁇ value is ⁇ 0.2°.
  • Embodiment 7 the preparation of formula I compound nitrate
  • Solubility measurement experiment results the solubility of the compound L-malate of the formula I is 40.48 mg/mL, and the solubility of the compound L-malate of the formula I is better.
  • formula I compound L-malate has higher fusing point (promptly having higher stability), lower hygroscopicity, and suitable solubility is arranged, and this makes formula I compound malate be suitable for preparing oral Preparation of solid dosage forms, especially tablets.
  • Test example 2 The physical stability research of the prepared salt of embodiment 2,7-10
  • the formula I compound L-malate A crystal form (the sample obtained in Example 2), the formula I compound nitrate (the sample obtained in Example 7), the formula I compound succinate (the sample obtained in Example 8), the formula I Compound p-toluenesulfonate (samples obtained in Example 9), compound tartrate of formula I (samples obtained in Example 10) are respectively exposed and laid flat, and placed under high temperature (60° C.), high humidity (RH92.5%), light Carry out the stability test of sample under (4500 ⁇ 500Lux) condition, measure in different sampling time (0 day, 6 days, 14 days, 30 days) in the sample active substance (formula I compound or phenomeline) and related substances content, as shown in Table 6.
  • nitrate of the compound of formula 1 is liquid under high temperature conditions, and its physical properties are unstable. NA means not determined.
  • the crystal form A of compound L-malate of formula I of the present invention has very low impurity content, and has very good stability under high temperature, high humidity and light conditions, and has a good prospect for pharmaceutical preparation;
  • the malate A crystal form of the compound of formula I is more stable under light conditions than other salt forms, which is convenient for the storage of raw materials and pharmaceutical preparations.
  • Test example 3 Toxic and side effects research of different salt types
  • Zanomeline is an M receptor activator, which can not only act on the central nervous system, but also stimulate M receptors in peripheral nervous tissue, resulting in cholinergic side effects, such as salivation, nausea , dizziness, etc.
  • the toxic and side effects of each compound were evaluated by observing the salivation of SD rats in each group. According to the experimental results, it can be seen that the cholinergic side effect of the crystal form A of the compound L-malate of formula I is small and has good safety.
  • mice male SD rats (weight 180-220g, purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd., production license number: SCXK (Beijing) 2016-0006), formula I compound L-malate A crystal Type (Example 2), formula I compound (Example 1), purified water (self-made).
  • mice 6 male SD rats were randomly divided into 2 groups (3 rats in each group), free to drink water during the test period, fasted for more than 12 hours before administration, and fed 4 hours after administration.
  • Oral gavage administration two groups of SD rats were given formula I compound malate A crystal form A aqueous solution and formula I compound aqueous solution at a dose of 30 mg/kg (calculated as free base).
  • 0min before administration 15min, 30min, 45min, 1.0h, 1.5h, 2h, 3h, 4h, 6h, 8h, and 10h after administration
  • blood samples were collected into K 2 EDTA anticoagulant tubes, and temporarily stored on ice until centrifugal.
  • Plasma needs to be centrifuged within 60 minutes after blood collection (at 2-8°C, centrifuge at 8000rpm for 5min), after centrifugation, transfer the plasma to a 96-well plate or centrifuge tube, transport it in an ice box, and store it at ⁇ -15°C for LC-MS/MS detection.
  • the LC-MS/MS bioanalysis method was used to detect the drug concentration in SD rat plasma, and the non-compartmental model was used to analyze the blood drug concentration-time data using WinNonlin TM (Version8.3, Certara, USA) to evaluate its SD rat pharmacokinetic (PK) characteristics in vivo, the data are shown in Table 10.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Psychiatry (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Pain & Pain Management (AREA)
  • Addiction (AREA)
  • Psychology (AREA)
  • Hospice & Palliative Care (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to the field of medicinal chemistry, and in particular, to a malate salt of a xanomeline derivative, the malate salt having a structure as shown in formula (I). The malate salt of the compound of formula (I) has excellent effects on at least one aspect of physical stability, solubility, hygroscopicity, biological activity, safety, bioavailability, toxic and side effects, etc.

Description

占诺美林衍生物的苹果酸盐、A晶型及其制备方法和用途Malate salt of zanameline derivative, crystal form A, preparation method and use thereof
本申请要求以下在先申请的优先权:2021年10月14日向中国国家知识产权局提交的专利申请号为202111198168.4,发明名称为“占诺美林衍生物的苹果酸盐、A晶型及其制备方法和用途”的在先申请。所述在先申请的全文通过引用的方式结合于本申请中。This application claims the priority of the following earlier application: the patent application number 202111198168.4 filed with the State Intellectual Property Office of China on October 14, 2021, and the title of the invention is "malate, crystal form A of zanomeline derivatives and its Preparation method and use of "prior application. The entirety of said prior application is incorporated by reference into this application.
技术领域technical field
本发明涉及药物化学领域,具体涉及一种占诺美林衍生物的苹果酸盐、A晶型及其制备方法,还包括所述占诺美林衍生物的苹果酸盐、A晶型在制备预防或治疗中枢神经系统紊乱疾病药物中的应用。The present invention relates to the field of medicinal chemistry, in particular to a malate salt of a jenomeline derivative, a crystal form A and a preparation method thereof, which also includes the preparation of the malate salt and a crystal form A of a jenomeline derivative The application of drugs for the prevention or treatment of disorders of the central nervous system.
背景技术Background technique
神经递质是神经元分泌的化学信使,用以促进信息流动并与中枢神经系统和周围神经系统中的其它细胞(例如肌肉或类似神经细胞)进行通讯。乙酰胆碱是大脑中的关键神经递质之一,其有两种不同的受体类别:毒蕈碱型受体(M受体,G蛋白偶联受体)和烟碱型受体(N受体,离子通道受体)。Neurotransmitters are chemical messengers secreted by neurons to facilitate the flow of information and communicate with other cells in the central and peripheral nervous systems, such as muscle or similar nerve cells. Acetylcholine, one of the key neurotransmitters in the brain, has two distinct receptor classes: muscarinic receptors (M receptors, G protein-coupled receptors) and nicotinic receptors (N receptors , ion channel receptors).
M受体家族包含M1至M5五种亚型,它们都在大脑和周围组织中表达,并在认知、行为、感觉、运动和自主神经过程中起着许多关键的生理作用。M型受体信号的破坏会导致记忆障碍和认知障碍,引发包括精神分裂症和阿尔茨海默症(AD)在内的多种疾病,并加剧精神病。相反,第三方的临床前和临床数据表明,M型受体信号传导的增强则会改善这些症状,此外M型受体尤其是M1、M2、M4受体也被认为与镇痛有关。The M receptor family includes five subtypes from M1 to M5, all of which are expressed in the brain and peripheral tissues and play many key physiological roles in cognition, behavior, sensory, motor and autonomic processes. Disruption of M-type receptor signaling leads to memory impairment and cognitive impairment, triggers a variety of diseases including schizophrenia and Alzheimer's disease (AD), and exacerbates psychosis. On the contrary, third-party preclinical and clinical data show that the enhancement of M-type receptor signaling can improve these symptoms. In addition, M-type receptors, especially M1, M2, and M4 receptors, are also considered to be related to analgesia.
占诺美林(xanomeline)是一种毒蕈碱受体的部分激动剂,可对毒蕈碱受体的5个亚型都产生激动作用,不具备选择性。由美国礼来公司及Novo Nordisk公司共同开发并上市,临床主要用于阿尔茨海默症的治疗,占诺美林的化学名称为3-[(4-己氧基)-1,2,5-噻二唑-3-基]-1,2,5,6-四氢-1-甲基吡啶,化学结构式如下:Xanomeline is a partial agonist of muscarinic receptors, which can produce agonistic effects on all five subtypes of muscarinic receptors without selectivity. Jointly developed and marketed by Eli Lilly and Novo Nordisk, it is mainly used clinically for the treatment of Alzheimer's disease. The chemical name of Normeline is 3-[(4-hexyloxy)-1,2,5 -Thiadiazol-3-yl]-1,2,5,6-tetrahydro-1-picoline, the chemical structure is as follows:
Figure PCTCN2022124662-appb-000001
Figure PCTCN2022124662-appb-000001
专利文献CN94192681.8公开了占诺美林可以转化为草酸盐,但草酸盐对病人的肾功能具有潜在的副作用,因而在药学上不宜,特别是在治疗老年人时尤为不宜。并进一步公开了在十二种可药用酸(未公开具体药用酸的名称)系列中,只有占诺美林酒石酸盐才具备好的生物利用率,好的处理性质和可重复的晶型。Patent document CN94192681.8 discloses that oxalate can be converted into oxalate, but oxalate has potential side effects on the patient's kidney function, so it is not suitable for medicine, especially when treating the elderly. And it is further disclosed that in the series of twelve kinds of pharmaceutically acceptable acids (the name of the specific pharmaceutically acceptable acid is not disclosed), only zomeline tartrate has good bioavailability, good handling properties and reproducible crystal form .
目前,公知常识普遍教导,选择具有所需的性能组合的盐仍然是一个困难的半经验性的选择,需要盐形式的性质的折衷选择,但是仍然存在评估哪种盐形式为最适合筛选特定候选药物的困难。Currently, common general knowledge generally teaches that selecting a salt with a desired combination of properties remains a difficult semi-empirical choice, requiring a compromise in the properties of the salt form, but there remains the question of which salt form is most suitable for screening a particular candidate Medication difficulties.
药物盐型的筛选是一个困难的半经验性选择,药物吸湿性会严重影响药物的流动性,甚至会影响药物的稳定性。药物的溶解度对药剂的制备、药物溶出、吸收等都具有至关重要的影响。但如何提高药物的溶解度,不以药物的吸湿性为代价,得到稳定性、溶解度及吸湿性均合适的候选药物盐是困难的。The screening of drug salt form is a difficult semi-empirical choice, and the hygroscopicity of the drug will seriously affect the fluidity of the drug, and even affect the stability of the drug. The solubility of drugs has a crucial impact on the preparation of pharmaceuticals, drug dissolution, absorption and so on. But how to improve the solubility of the drug, without the expense of the hygroscopicity of the drug, it is difficult to obtain a candidate drug salt with suitable stability, solubility and hygroscopicity.
另外,Xanomeline作为一种M受体激活剂在临床上治疗精神分裂症和AD患者的精神病和相关行为症状方面具有令人鼓舞的治疗效果,但其潜力一直受到胆碱能副作用的限制,包括流涎、恶心、头晕等,这被认为是由于刺激外周神经组织中的M受体所导致的。占诺美林在人体内的代谢情况复杂且不可预测,且占诺美林对毒蕈碱受体亚型缺乏选择性,因此对占诺美林的药物开发造成了困难。Alternatively, Xanomeline as an M-receptor activator has shown encouraging therapeutic efficacy in the clinic for the treatment of psychosis and related behavioral symptoms in patients with schizophrenia and AD, but its potential has been limited by cholinergic side effects, including salivation , nausea, dizziness, etc., which are thought to be due to stimulation of M receptors in peripheral nervous tissue. The metabolism of Zanameline in the human body is complex and unpredictable, and Zanameline lacks selectivity for muscarinic receptor subtypes, thus causing difficulties in the drug development of Zanameline.
因此,需要进一步寻找具有良好疗效、副作用小、具有更好的药代动力学性质的适于成药的新型化合物;还需要开发出该化合物具有适合可靠的配制和制备特性的结晶盐及其多晶型物。Therefore, it is necessary to further search for novel compounds suitable for drug-making with good curative effect, small side effects, and better pharmacokinetic properties; it is also necessary to develop crystalline salts and polymorphs thereof with suitable and reliable formulation and preparation characteristics. Models.
发明内容Contents of the invention
本发明的目的在于提供一种式I所示的占诺美林衍生物的苹果酸盐,所述苹果酸盐能够用于制备预防或治疗中枢神经系统紊乱疾病的药物并且这种衍生物的溶解能力改善、储存稳定、给药方便、药物代谢、药物毒副作用改善,并且给药时患者的依从度高。The object of the present invention is to provide a malate salt of a zanmeline derivative shown in formula I, which can be used for the preparation of medicines for preventing or treating central nervous system disorders and the dissolution of this derivative Improved ability, stable storage, convenient administration, improved drug metabolism, drug toxicity and side effects, and high patient compliance during administration.
本发明的另一目的在于提供一种式I所示占诺美林衍生物的苹果酸盐的A晶型,所述A晶型能够具备优异的热力学稳定性和机械稳定性,重复性好,适于商业化大生产。Another object of the present invention is to provide a crystal form A of the malate salt of phenomeline derivatives shown in formula I, the crystal form A can have excellent thermodynamic stability and mechanical stability, good repeatability, Suitable for commercial mass production.
第一方面,本发明提供了式I化合物的苹果酸盐。In a first aspect, the present invention provides a malate salt of a compound of formula I.
Figure PCTCN2022124662-appb-000002
Figure PCTCN2022124662-appb-000002
在本发明的一些方案中,上述式I化合物的苹果酸盐,如式Ⅱ所示,其中,x选自0.5~2。In some solutions of the present invention, the malate salt of the compound of formula I above is shown in formula II, wherein x is selected from 0.5-2.
Figure PCTCN2022124662-appb-000003
Figure PCTCN2022124662-appb-000003
本发明的一些方案中,上述x为0.5、1.0、1.5、2.0。In some solutions of the present invention, the above-mentioned x is 0.5, 1.0, 1.5, 2.0.
本发明的一些方案中,上述式Ⅱ化合物中x为1.0。In some aspects of the present invention, x in the compound of formula II above is 1.0.
本发明的一些方案中,上述式Ⅱ化合物,其中苹果酸为L-苹果酸。In some solutions of the present invention, the compound of formula II above, wherein the malic acid is L-malic acid.
第二方面,本发明提供了式I化合物的苹果酸盐的A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、14.827±0.2°、16.796±0.2°、20.012±0.2°和21.942±0.2°。所述式I化合物的苹果酸盐的A晶型是x为1.0的式Ⅱ化合物,且其中的苹果酸为L-苹果酸。In a second aspect, the present invention provides the A crystal form of the malate salt of the compound of formula I, whose X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 14.827±0.2°, 16.796±0.2° , 20.012±0.2° and 21.942±0.2°. The crystal form A of the malate salt of the compound of formula I is the compound of formula II with x being 1.0, and the malic acid therein is L-malic acid.
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、14.827±0.2°、16.796±0.2°、20.012±0.2°、21.942±0.2°、22.473±0.2°和25.333±0.2°。In some solutions of the present invention, the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 14.827±0.2°, 16.796±0.2°, 20.012±0.2°, 21.942±0.2 °, 22.473±0.2° and 25.333±0.2°.
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、13.288±0.2°、14.827±0.2°、16.796±0.2°、20.012±0.2°、21.942±0.2°、22.473±0.2°和25.333±0.2°。In some solutions of the present invention, the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 13.288±0.2°, 14.827±0.2°, 16.796±0.2°, 20.012±0.2 °, 21.942±0.2°, 22.473±0.2°, and 25.333±0.2°.
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、13.288±0.2°、14.827±0.2°、15.399±0.2°、16.796±0.2°、20.012±0.2°、20.327±0.2°、21.942±0.2°、22.473±0.2°和25.333±0.2°。In some solutions of the present invention, the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 13.288±0.2°, 14.827±0.2°, 15.399±0.2°, 16.796±0.2 °, 20.012±0.2°, 20.327±0.2°, 21.942±0.2°, 22.473±0.2°, and 25.333±0.2°.
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、13.288±0.2°、14.827±0.2°、15.399±0.2°、16.796±0.2°、20.012±0.2°、20.327±0.2°、21.942±0.2°、22.473±0.2°、24.996±0.2°、25.333±0.2°和27.718±0.2°。In some solutions of the present invention, the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 13.288±0.2°, 14.827±0.2°, 15.399±0.2°, 16.796±0.2 °, 20.012±0.2°, 20.327±0.2°, 21.942±0.2°, 22.473±0.2°, 24.996±0.2°, 25.333±0.2°, and 27.718±0.2°.
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、12.755±0.2°、13.288±0.2°、13.903±0.2°、14.827±0.2°、15.399±0.2°、16.796±0.2°、17.686±0.2°、18.906±0.2°、20.012±0.2°、20.327±0.2°、21.942±0.2°、22.473±0.2°、24.996±0.2°、25.333±0.2°和27.718±0.2°。In some solutions of the present invention, the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 12.755±0.2°, 13.288±0.2°, 13.903±0.2°, 14.827±0.2 °, 15.399±0.2°, 16.796±0.2°, 17.686±0.2°, 18.906±0.2°, 20.012±0.2°, 20.327±0.2°, 21.942±0.2°, 22.473±0.2°, 24.996±0.2°, 25.333±0.2 ° and 27.718±0.2°.
本发明的一些方案中,上述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、12.755±0.2°、13.288±0.2°、13.903±0.2°、14.827±0.2°、15.399±0.2°、16.796±0.2°、17.686±0.2°、18.906±0.2°、20.012±0.2°、20.327±0.2°、20.660±0.2°、21.942±0.2°、22.473±0.2°、24.996±0.2°、25.333±0.2°、27.382±0.2°和27.718±0.2°。In some solutions of the present invention, the X-ray powder diffraction pattern of the above crystal form A has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 12.755±0.2°, 13.288±0.2°, 13.903±0.2°, 14.827±0.2 °, 15.399±0.2°, 16.796±0.2°, 17.686±0.2°, 18.906±0.2°, 20.012±0.2°, 20.327±0.2°, 20.660±0.2°, 21.942±0.2°, 22.473±0.2°, 24.996±0.2 °, 25.333±0.2°, 27.382±0.2°, and 27.718±0.2°.
本发明的一些方案中,上述A晶型,其具有基本如图1所示的XRPD图谱。In some solutions of the present invention, the above crystal form A has an XRPD pattern as shown in FIG. 1 .
本发明的一些方案中,上述A晶型,其具有基本如图2所示的DSC图谱。In some solutions of the present invention, the above crystal form A has a DSC spectrum substantially as shown in FIG. 2 .
本发明的一些方案中,上述A晶型的差示扫描量热曲线在111.03±3℃处具有吸热峰。In some solutions of the present invention, the differential scanning calorimetry curve of the above crystal form A has an endothermic peak at 111.03±3°C.
本发明的一些方案中,上述A晶型,其具有基本如图3所示的TGA曲线。In some solutions of the present invention, the above crystal form A has a TGA curve substantially as shown in FIG. 3 .
第三方面,本发明还提供式I化合物的苹果酸盐的制备方法,包括将式I化合物与苹果酸反应。In a third aspect, the present invention also provides a method for preparing malate of the compound of formula I, comprising reacting the compound of formula I with malic acid.
本发明的一些方案中,上述式I化合物苹果酸盐的A晶型的制备方法包括如下步骤:In some schemes of the present invention, the preparation method of the A crystal form of the compound malate of the above formula I comprises the following steps:
将式I化合物、苹果酸与溶剂混合,加热至完全溶解,再冷却,析出晶体;其中,所述溶剂选自乙醇、异丙醇、正丙醇、丁醇、丙酮、乙酸乙酯、正庚烷、乙腈、四氢呋喃中的一种或一种以上的混合物;优选为乙醇、异丙醇、乙酸乙酯、正庚烷中的一种或一种以上的混合物;或者Mix the compound of formula I, malic acid and a solvent, heat until completely dissolved, then cool to precipitate crystals; wherein, the solvent is selected from ethanol, isopropanol, n-propanol, butanol, acetone, ethyl acetate, n-heptyl One or more mixtures of alkanes, acetonitrile, tetrahydrofuran; preferably one or more mixtures of ethanol, isopropanol, ethyl acetate, n-heptane; or
将式I化合物、苹果酸与第一溶剂混合,加热至完全溶解,再加入第二溶剂,冷却,析晶。所述第一溶剂选自甲醇、乙醇、异丙醇、正丁醇中的一种或一种以上的混合物;所述第二溶剂选自甲基叔丁基醚、酯类、乙腈、烷烃类中的一种或一种以上的混合物,说是酯类选自乙酸乙酯和/或,所述脂类例如为醋酸异丙酯;所述烷烃类选自正庚烷和/或正己烷。Mix the compound of formula I, malic acid and the first solvent, heat until completely dissolved, then add the second solvent, cool and crystallize. The first solvent is selected from one or more mixtures of methanol, ethanol, isopropanol, n-butanol; the second solvent is selected from methyl tert-butyl ether, esters, acetonitrile, alkanes The mixture of one or more than one of them means that the esters are selected from ethyl acetate and/or, the lipids are, for example, isopropyl acetate; the alkanes are selected from n-heptane and/or n-hexane.
本发明的一些方案中,加热温度为50~80℃,例如60~70℃。In some solutions of the present invention, the heating temperature is 50-80°C, such as 60-70°C.
本发明的一些方案中,将式I化合物、苹果酸与溶解该化合物所用的溶剂或第一溶剂混合后进行搅拌,所述搅拌时间可以是1~5小时,优选2~3小时。In some solutions of the present invention, the compound of formula I, malic acid and the solvent used to dissolve the compound or the first solvent are mixed and then stirred. The stirring time may be 1-5 hours, preferably 2-3 hours.
所述式I化合物与溶解该化合物所用的溶剂或第一溶剂的重量/体积比(g/mL)为1:4-10,优选地为1:6-8,例如1:6、1:7、1:8。The weight/volume ratio (g/mL) of the compound of formula I to the solvent used to dissolve the compound or the first solvent is 1:4-10, preferably 1:6-8, such as 1:6, 1:7 , 1:8.
本发明的一些方案中,所述冷却可以是冷却至0~30℃,优选地,冷却至2~8℃,例如冷却至5℃。在冷却之前可以先进行一次过滤。In some solutions of the present invention, the cooling may be to 0-30°C, preferably to 2-8°C, for example to 5°C. It can be filtered once before cooling.
本发明的一些方案中,在冷却过程中进行搅拌,所述搅拌的时间可以是6~24小时,优 选18小时。In some solutions of the present invention, stirring is carried out during the cooling process, and the stirring time may be 6 to 24 hours, preferably 18 hours.
根据本发明,在析出晶体后进行过滤,干燥;在本发明的一些方案中,干燥之前还包括任选地用溶剂洗涤的步骤,所述洗涤溶剂选自乙醇、异丙醇、正丙醇、丙酮、乙酸乙酯、乙腈、四氢呋喃中的一种或一种以上的混合物。According to the present invention, after the crystals are precipitated, filter and dry; in some solutions of the present invention, the step of washing with a solvent is also included before drying, and the washing solvent is selected from ethanol, isopropanol, n-propanol, One or a mixture of acetone, ethyl acetate, acetonitrile, tetrahydrofuran.
本发明的一些方案中,所述式I化合物与苹果酸的摩尔比为1:0.9-1.3,优选地,所述式I化合物与苹果酸的摩尔比为1:1-1.1,例如为1:1、1:1.05、1:1.1。In some schemes of the present invention, the molar ratio of the compound of formula I to malic acid is 1:0.9-1.3, preferably, the molar ratio of the compound of formula I to malic acid is 1:1-1.1, for example 1: 1. 1:1.05, 1:1.1.
第四方面,本发明提供了一种药物组合物,所述药物组合物包含式I化合物的苹果酸盐或者式I化合物的苹果酸盐A晶型,和任选的药学上可接受的赋形剂。In a fourth aspect, the present invention provides a pharmaceutical composition, the pharmaceutical composition comprising the malate of the compound of formula I or the malate A crystal form of the compound of formula I, and optional pharmaceutically acceptable excipients agent.
第五方面,本发明提供了式I化合物的苹果酸盐、上述式I化合物的苹果酸盐A晶型、或包含式I化合物苹果酸盐的药物组合物、或包含式I化合物的苹果酸盐A晶型的药物组合物在制备用于治疗中枢神经系统紊乱疾病的药物中的用途。In a fifth aspect, the present invention provides the malate of the compound of formula I, the malate A crystal form of the compound of formula I above, or the pharmaceutical composition comprising the malate of the compound of formula I, or the malate of the compound of formula I Use of the pharmaceutical composition of crystal form A in the preparation of medicines for treating disorders of the central nervous system.
本发明的一些方案中,所述中枢神经系统紊乱疾病包括但不限于精神分裂症、阿尔茨海默氏病、帕金森氏病、抑郁症、运动障碍、吸毒成瘾、疼痛和神经退行性变(例如陶氏病或突触核蛋白病)。In some aspects of the present invention, the disorders of the central nervous system include but are not limited to schizophrenia, Alzheimer's disease, Parkinson's disease, depression, movement disorders, drug addiction, pain and neurodegeneration (eg Dow disease or synucleinopathies).
本发明的一些方案中,所述中枢神经系统紊乱疾病为精神分裂症。In some aspects of the present invention, the central nervous system disorder is schizophrenia.
第六方面,本发明提供了一种治疗或预防哺乳动物(例如人)的中枢神经系统紊乱疾病的方法,所述方法包括向哺乳动物(例如人)给予治疗有效量的式I化合物的苹果酸盐、式I化合物的苹果酸盐A晶型、包含式I化合物的苹果酸盐的药物组合物和包含式I化合物的苹果酸盐A晶型的药物组合物。In a sixth aspect, the present invention provides a method for treating or preventing central nervous system disorders in mammals (such as humans), the method comprising administering a therapeutically effective amount of malic acid of a compound of formula I to mammals (such as humans) salt, malate A crystal form of the compound of formula I, a pharmaceutical composition comprising the malate salt of the compound of formula I and a pharmaceutical composition comprising the malate A crystal form of the compound of formula I.
本发明的一些方案中,所述中枢神经系统紊乱疾病包括但不限于精神分裂症、阿尔茨海默氏病、帕金森氏病、抑郁症、运动障碍、吸毒成瘾、疼痛和神经退行性变(例如陶氏病或突触核蛋白病)。In some aspects of the present invention, the disorders of the central nervous system include but are not limited to schizophrenia, Alzheimer's disease, Parkinson's disease, depression, movement disorders, drug addiction, pain and neurodegeneration (eg Dow disease or synucleinopathies).
本发明的一些方案中,所述中枢神经系统紊乱疾病为精神分裂症。In some aspects of the present invention, the central nervous system disorder is schizophrenia.
有益效果Beneficial effect
1.本发明首次提供了式I化合物的苹果酸盐;式I化合物的苹果酸盐在物理稳定性、溶解性、吸湿性、生物活性、安全性、生物利用度、毒副作用等至少一方面具有优异的效果。1. The present invention provides the malate of the compound of formula I for the first time; the malate of the compound of formula I has at least one aspect such as physical stability, solubility, hygroscopicity, biological activity, safety, bioavailability, toxic side effect Excellent effect.
2.式I化合物苹果酸盐的A晶型的稳定性好,吸湿性小,水溶性好,批次间差异小、成药前景良好。2. The crystal form A of the malate salt of the compound of formula I has good stability, low hygroscopicity, good water solubility, small batch-to-batch variation, and good prospects for making a drug.
3.本发明的盐型、晶型制备工艺简单,适宜工业化生产。3. The preparation process of the salt form and crystal form of the present invention is simple and suitable for industrial production.
附图说明Description of drawings
图1为式I化合物L-苹果酸盐的A晶型的XRPD图谱;Fig. 1 is the XRPD pattern of A crystal form of compound L-malate of formula I;
图2为式I化合物L-苹果酸盐的A晶型的DSC图谱;Fig. 2 is the DSC spectrum of the A crystal form of formula I compound L-malate;
图3为式I化合物L-苹果酸盐的A晶型的TGA图谱;Fig. 3 is the TGA spectrum of the A crystal form of formula I compound L-malate;
图4为式I化合物L-苹果酸盐的A晶型在高温(60℃)条件下0天、6天、14天、30天的XRPD图谱;Fig. 4 is the XRPD patterns of crystal form A of compound L-malate of formula I at high temperature (60° C.) for 0 days, 6 days, 14 days, and 30 days;
图5为式I化合物L-苹果酸盐的A晶型在高湿(RH92.5%)条件下0天、6天、14天、30天的XRPD图谱;Fig. 5 is the XRPD patterns of crystal form A of compound L-malate of formula I under high humidity (RH92.5%) conditions at 0 days, 6 days, 14 days, and 30 days;
图6为式I化合物L-苹果酸盐的A晶型在光照条件(4500±500Lux)条件下0天、6天、14天、30天的XRPD图谱。Fig. 6 is the XRPD pattern of crystal form A of L-malate of the compound of formula I under light conditions (4500±500Lux) at 0 days, 6 days, 14 days and 30 days.
图7为口服给予式I化合物L-苹果酸盐的A晶型(实施例2样品)、式I化合物(实施例1样品)后大鼠血浆中式I化合物的药代动力曲线。Fig. 7 is the pharmacokinetic curve of the compound of formula I in rat plasma after oral administration of crystal form A of compound L-malate of formula I (sample of Example 2) and compound of formula I (sample of Example 1).
具体实施方式Detailed ways
下文将结合具体实施例对本发明的通式化合物及其制备方法和应用做更进一步的详细说明。应当理解,下列实施例仅为示例性地说明和解释本发明,而不应被解释为对本发明保护范围的限制。凡基于本发明上述内容所实现的技术均涵盖在本发明旨在保护的范围内。The compound of the general formula of the present invention and its preparation method and application will be further described in detail below in conjunction with specific examples. It should be understood that the following examples are only for illustrating and explaining the present invention, and should not be construed as limiting the protection scope of the present invention. All technologies realized based on the above contents of the present invention are covered within the scope of protection intended by the present invention.
除非另有说明,以下实施例中使用的原料和试剂均为市售商品,或者可以通过已知方法制备。Unless otherwise stated, the raw materials and reagents used in the following examples are commercially available or can be prepared by known methods.
本发明采用下述缩略词:r.t.代表室温;aq代表水溶液;DCM代表二氯甲烷;THF代表四氢呋喃;MeOH代表甲醇;EtOH代表乙醇;IPA代表异丙醇;DSC代表差示扫描量热仪;TGA代表热重分析; 1H-NMR代表核磁共振氢谱;XRPD代表X-射线粉末衍射;eq代表摩尔当量。 The present invention adopts the following abbreviations: rt stands for room temperature; aq stands for aqueous solution; DCM stands for dichloromethane; THF stands for tetrahydrofuran; MeOH stands for methanol; EtOH stands for ethanol; IPA stands for isopropanol; DSC stands for differential scanning calorimeter; TGA stands for thermogravimetric analysis; 1 H-NMR stands for hydrogen nuclear magnetic resonance spectrum; XRPD stands for X-ray powder diffraction; eq stands for molar equivalent.
化合物经手工或者ChemDraw软件命名,市售化合物采用供应商目录名称。The compounds were named manually or by ChemDraw software, and the commercially available compounds used the supplier catalog names.
下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。For the experimental methods without specific conditions indicated in the following examples, the conventional conditions or the conditions suggested by the manufacturer are usually followed. Percentages and parts are by weight unless otherwise indicated.
本发明实施例对盐和晶体进行表征所采用的测试方法和仪器如下:The test methods and instruments used to characterize salts and crystals in the embodiments of the present invention are as follows:
粉末X-射线衍射(又称“X-射线粉末衍射”,X-ray powder diffractometer,XRPD)方法Powder X-ray diffraction (also known as "X-ray powder diffraction", X-ray powder diffractometer, XRPD) method
仪器型号:布鲁克D8advanceX-射线衍射仪Instrument model: Bruker D8advance X-ray diffractometer
测试方法:大约10~20mg样品用于XRPD检测。Test method: About 10-20 mg of sample is used for XRPD detection.
详细的XRPD参数如下:The detailed XRPD parameters are as follows:
光管:Cu,kα,
Figure PCTCN2022124662-appb-000004
Light pipe: Cu, kα,
Figure PCTCN2022124662-appb-000004
光管电压:40kV,光管电流:40mAPhototube voltage: 40kV, phototube current: 40mA
发散狭缝:0.60mmDivergence slit: 0.60mm
探测器狭缝:10.50mmDetector slit: 10.50mm
防散射狭缝:7.10mmAnti-scatter slit: 7.10mm
扫描范围:4-40degScanning range: 4-40deg
步径:0.02degStep: 0.02deg
步长:0.12秒Step size: 0.12 seconds
需要说明的是,在X-射线粉末衍射光谱(XRPD)中,由结晶化合物得到的衍射谱图对于特定的结晶往往是特征性的,其中谱带(尤其是在低角度)的相对强度可能会因为结晶条件、粒径和其它测定条件的差异而产生的优势取向效果而变化。因此,衍射峰的相对强度对所针对的结晶并非是特征性的,判断是否与已知的结晶相同时,更应该注意的是峰的相对位置而不是它们的相对强度。此外,对任何给定的结晶而言,峰的位置可能存在轻微误差,这在结晶学领域中也是公知的。例如,由于分析样品时温度的变化、样品移动、或仪器的标定等,峰的位置可以移动,2θ值的测定误差有时约为±0.2°。因此,在确定每种结晶结构时,应该将此误差考虑在内。在XRPD图谱中通常用2θ角或晶面距d表示峰位置,两者之间具有简单的换算关系:d=λ/2sinθ,其中d代表晶面距(又称“面间距”),λ代表入射X射线的波长,θ为衍射角。对于同种化合物的同种结晶,其XRPD谱的峰位置在整体上具有相似性,相对强度误差可能较大。还应指出的是,在混合物的鉴定中,由于含量下降等因素会造成部分衍射线的缺失,此时,无需依赖高纯试样中观察到的全部谱带,甚至一条谱带也可能对给定的结晶是特征性的。It should be noted that in X-ray powder diffraction (XRPD), the diffraction pattern obtained from a crystalline compound is often characteristic for a particular crystal, where the relative intensities of the bands (especially at low angles) may vary. Varies due to the effect of preferred orientation due to differences in crystallization conditions, particle size, and other measurement conditions. Therefore, the relative intensity of the diffraction peaks is not characteristic of the targeted crystal, and when judging whether it is the same as a known crystal, more attention should be paid to the relative positions of the peaks rather than their relative intensities. Furthermore, for any given crystal there may be slight errors in the position of the peaks, as is well known in the art of crystallography. For example, due to temperature changes, sample movement, or instrument calibration when analyzing samples, the position of the peak can move, and the measurement error of the 2θ value is sometimes about ±0.2°. Therefore, this error should be taken into account when determining each crystal structure. In the XRPD spectrum, the 2θ angle or the crystal plane distance d is usually used to represent the peak position, and there is a simple conversion relationship between the two: d=λ/2sinθ, where d represents the crystal plane distance (also known as "planar distance"), and λ represents The wavelength of the incident X-ray, θ is the diffraction angle. For the same crystal of the same compound, the peak positions of their XRPD spectra are similar on the whole, and the relative intensity error may be large. It should also be pointed out that in the identification of mixtures, due to factors such as content decline, some diffraction lines will be missing. At this time, it is not necessary to rely on all the bands observed in the high-purity sample, and even one band may affect the given Certain crystallization is characteristic.
由包括以下的多种因素产生与这类X射线粉末衍射分析结果相关的测量差异:(a)样品制备物(例如样品高度)中的误差,(b)仪器误差,(c)校准差异,(d)操作人员误差(包括在测定峰位置时出现的误差),和(e)物质的性质(例如优选的定向误差)。校准误差和样品高度误差经常导致所有峰在相同方向中的位移。当使用平的支架时,样品高度的小差异将导致XRPD峰位置的大位移。系统研究显示1mm的样品高度差异可以导致高至1°的2θ的峰位移。可以从X射线衍射图鉴定这些位移,并且可以通过针对所述位移进行补偿(将系统校准因子用于所有峰位置值)或再校准仪器消除所述位移。如上所述,通过应用系统校准因子使峰位置一致,可校正来自不同仪器的测量误差。Measurement variance associated with such X-ray powder diffraction analysis results arises from a variety of factors including: (a) errors in sample preparation (e.g., sample height), (b) instrument errors, (c) calibration differences, ( d) operator errors (including errors in determining peak positions), and (e) properties of the species (eg, preferred orientation errors). Calibration errors and sample height errors often cause all peaks to shift in the same direction. Small differences in sample height will result in large shifts in XRPD peak positions when using flat supports. Systematic studies have shown that a sample height difference of 1 mm can result in a peak shift in 2θ of up to 1°. These shifts can be identified from the X-ray diffraction pattern and can be eliminated by compensating for them (using a system calibration factor for all peak position values) or recalibrating the instrument. Measurement errors from different instruments can be corrected for by applying system calibration factors to make peak positions consistent, as described above.
差热分析(又称“差示扫描量热法”,Differential Scanning Calorimeter,DSC)方法Differential thermal analysis (also known as "differential scanning calorimetry", Differential Scanning Calorimeter, DSC) method
仪器型号:METTLER TOLEDO DSC3+差示扫描量热仪Instrument Model: METTLER TOLEDO DSC3+ Differential Scanning Calorimeter
测试方法:取样品(~1mg)置于DSC铝锅内进行测试,在50mL/minN 2条件下,以10℃/min的升温速率,加热样品从25℃到300℃。本发明中采用差式扫描量热法(DSC)来测定熔点。DSC测定当结晶由于其结晶结构发生变化或结晶熔融而吸收或释放热时的转变温度。对于同种化合物的同种结晶,在连续的分析中,热转变温度和熔点误差典型的在约5℃之内,通常在约3℃之内,当我们说一个化合物具有一给定的DSC峰或熔点时,这是指该DSC峰或熔点±5℃。DSC提供了一种辨别不同结晶的辅助方法。不同的结晶形态可根据其不同的转变温度特征而加以识别。需要指出的是对于混合物而言,其DSC峰或熔点可能会在更大的范围内变动。此外,由于在物质熔化的过程中伴有分解,因此熔化温度与升温速率相关。 Test method: Take a sample (~1mg) and place it in a DSC aluminum pot for testing. Under the condition of 50mL/ minN2 , heat the sample from 25°C to 300°C at a heating rate of 10°C/min. In the present invention, differential scanning calorimetry (DSC) is used to determine the melting point. DSC measures the transition temperature when a crystal absorbs or releases heat due to a change in its crystal structure or melting of the crystal. For the same crystal of the same compound, in consecutive analysis, the error of thermal transition temperature and melting point is typically within about 5°C, usually within about 3°C, when we say that a compound has a given DSC peak or melting point, this means ±5°C of the DSC peak or melting point. DSC provides an auxiliary method for distinguishing between different crystals. Different crystalline forms can be identified based on their different transition temperature characteristics. It should be pointed out that for mixtures, the DSC peak or melting point may vary in a larger range. In addition, since the melting process of the substance is accompanied by decomposition, the melting temperature is related to the heating rate.
热重分析(Thermal Gravimetric Analyzer,TGA)方法Thermal Gravimetric Analyzer (TGA) method
仪器型号:TAQ5000IR热重分析仪Instrument Model: TAQ5000IR Thermogravimetric Analyzer
测试方法:取样品(2~5mg)置于TGA铂金锅内进行测试,在25mL/minN 2条件下,以10℃/min的升温速率,加热样品从室温到350℃。 Test method: Take a sample (2~5mg) and place it in a TGA platinum pot for testing. Under the condition of 25mL/ minN2 , heat the sample from room temperature to 350°C at a heating rate of 10°C/min.
高效液相色谱(HPLC)分析方法:High performance liquid chromatography (HPLC) analysis method:
所用仪器为Waters ARC 2998HPLC;色谱柱:Agilent Poroshell bonus-RP 4.6×100mm,2.7μm;The instrument used is Waters ARC 2998HPLC; Chromatographic column: Agilent Poroshell bonus-RP 4.6×100mm, 2.7μm;
测定条件如下:The measurement conditions are as follows:
进样体积:10ul;Injection volume: 10ul;
流速:1.0mL/min;Flow rate: 1.0mL/min;
检测波长:275nm;Detection wavelength: 275nm;
样品浓度:1.0mg/mL;Sample concentration: 1.0mg/mL;
稀释液:20%乙腈水溶液;Diluent: 20% acetonitrile aqueous solution;
柱温:40℃;Column temperature: 40°C;
流动相A:0.2%磷酸水溶液;Mobile phase A: 0.2% phosphoric acid aqueous solution;
流动相B:乙腈-甲醇(1:1)溶液;Mobile phase B: acetonitrile-methanol (1:1) solution;
洗脱梯度如表1所示:The elution gradient is shown in Table 1:
表1洗脱梯度条件Table 1 Elution gradient conditions
时间(min)time (min) %A%A %B%B
00 9090 1010
1515 6060 4040
2020 2525 7575
3030 2525 7575
3131 9090 1010
4040 9090 1010
目前,公知常识普遍教导,选择具有所需的性能组合的盐仍然是一个困难的半经验性的选择,需要盐形式的性质的折衷选择,但是仍然存在评估哪种盐形式为最适合筛选特定候选药物的困难。Currently, common general knowledge generally teaches that selecting a salt with a desired combination of properties remains a difficult semi-empirical choice, requiring a compromise in the properties of the salt form, but there remains the question of which salt form is most suitable for screening a particular candidate Medication difficulties.
在实验过程中,发明人发现药物吸湿性会严重影响药物的流动性,甚至会影响药物的稳定性。药物的溶解度对药剂的制备、药物溶出、吸收等都具有至关重要的影响。但如何提高药物的溶解度,不以药物的吸湿性为代价,得到药物稳定性、溶解度及吸湿性均合适的候选药物盐是困难的。During the experiment, the inventor found that the hygroscopicity of the drug will seriously affect the fluidity of the drug, and even affect the stability of the drug. The solubility of drugs has a crucial impact on the preparation of pharmaceuticals, drug dissolution, absorption and so on. But how to improve the solubility of the drug, without the expense of the hygroscopicity of the drug, it is difficult to obtain a drug candidate salt with suitable drug stability, solubility and hygroscopicity.
发明人经过广泛而深入的研究,出乎意料地发现式I化合物苹果酸盐经DSC测定具有较高的熔点且经过稳定性实验证实:式I化合物苹果酸盐具有较高的稳定性。另外,式I化合物苹果酸盐有较低的吸湿性,并且有适当的溶解度。式I化合物苹果酸盐的高稳定性、低吸湿度和适当溶解度的结合是不可预料的,这使得式I化合物苹果酸盐(尤其是式I化合物L-苹果酸盐)适用于制备口服固体制剂,特别是片剂的制备。After extensive and in-depth research, the inventor unexpectedly found that the malate of the compound of formula I has a higher melting point as determined by DSC, and the stability experiment confirmed that the malate of the compound of formula I has higher stability. In addition, the malate salt of the compound of formula I has low hygroscopicity and moderate solubility. The combination of high stability, low hygroscopicity and appropriate solubility of compound malate of formula I is unexpected, which makes compound malate of formula I (especially compound L-malate of formula I) suitable for the preparation of oral solid dosage forms , especially the preparation of tablets.
活性成份的溶解度对于剂型的选择具有重要意义,这是因为剂型对生物利用度具有直接影响。对于口服剂型而言,通常认为活性组分具有较高的溶解度是有利的,这是因为其使得生物利用度提高。The solubility of the active ingredient is of great importance for the choice of dosage form because the dosage form has a direct effect on bioavailability. For oral dosage forms, it is generally considered to be advantageous to have a higher solubility of the active ingredient because it leads to increased bioavailability.
在此基础上,发明人发现式I化合物苹果酸盐的A晶型,所述A晶型在物理稳定性、热力学稳定性和机械稳定性等至少一个方面具备优势;制备得到的式I化合物苹果酸盐的A晶型,晶体大小合适,适于工业化生产制备。On this basis, the inventors have discovered the A crystal form of malate of the compound of formula I, which has advantages in at least one of physical stability, thermodynamic stability and mechanical stability; the prepared formula I compound malate The crystal form A of the acid salt has a suitable crystal size and is suitable for industrial production and preparation.
一种式I化合物苹果酸盐A晶型,能够显著改善式I化合物的水溶性、且具有良好稳定性和极低的吸湿性,有利于制成口服固体制剂。该式I化合物苹果酸盐制成的制剂可以较长时间维持体内有效生理浓度的式I游离碱并且给药方便,从而具备提高患者的依从度等优点。A crystalline form of malate A of the compound of formula I can significantly improve the water solubility of the compound of formula I, and has good stability and extremely low hygroscopicity, which is beneficial for making oral solid preparations. The preparation made of malate of the compound of formula I can maintain the effective physiological concentration of the free base of formula I in the body for a long time and is convenient to administer, so it has the advantages of improving the compliance of patients and the like.
实施例1.式I化合物的合成Embodiment 1. the synthesis of formula I compound
Figure PCTCN2022124662-appb-000005
Figure PCTCN2022124662-appb-000005
步骤1、2-羟基-2-(3-吡啶基)乙腈(中间体1)的制备The preparation of step 1,2-hydroxyl-2-(3-pyridyl)acetonitrile (intermediate 1)
向100mL单口反应瓶中依次加入3-吡啶甲醛(6.00g,56.02mmol,1.0eq)、冰醋酸(3.36g,56.02mmol,1.0eq)和6mL纯水,在室温搅拌混匀。于2~8℃滴入三甲基氰硅烷(7.42g,74.42mmol,1.3eq)的水溶液中,搅拌反应,TLC(薄层色谱)板确认反应结束,冰盐浴降温至-5℃搅拌析晶体。过滤,用冰水(5mL*3)洗涤滤饼,得白色固体(6.72g,收率89%)。3-Pyridinecarbaldehyde (6.00g, 56.02mmol, 1.0eq), glacial acetic acid (3.36g, 56.02mmol, 1.0eq) and 6mL of pure water were successively added into a 100mL single-port reaction flask, and stirred and mixed at room temperature. Add dropwise trimethylsilyl cyanide (7.42g, 74.42mmol, 1.3eq) into an aqueous solution of trimethylsilyl cyanide (7.42g, 74.42mmol, 1.3eq) at 2-8°C, stir to react, confirm the completion of the reaction on a TLC (thin-layer chromatography) plate, cool down to -5°C in an ice-salt bath, stir and analyze crystals. After filtering, the filter cake was washed with ice water (5 mL*3) to obtain a white solid (6.72 g, yield 89%).
1H NMR(400MHz,CDCl 3)δ8.66–8.52(m,2H),7.96(m,1H),7.45(dd,J=8.0,4.9Hz,1H),5.64(s,1H).MS m/z:134.9[M+H] +. 1 H NMR (400MHz, CDCl 3 )δ8.66–8.52(m,2H),7.96(m,1H),7.45(dd,J=8.0,4.9Hz,1H),5.64(s,1H).MS m /z:134.9[M+H] + .
步骤2、2-氨基-2-(3-吡啶基)乙腈(中间体2)的制备The preparation of step 2,2-amino-2-(3-pyridyl)acetonitrile (intermediate 2)
向50mL二口反应瓶依次加入NH 4Cl(1.81g,33.80mmol,1.5eq)、水(15.00mL)和氨水(4.80mL,25%,1.1eq),室温搅拌溶清后加入中间体1(3.00g,22.37mmol,1.0eq),继续搅拌20小时。用二氯甲烷(15mL*7)萃取,合并有机相,2g无水硫酸钠干燥5分钟。过滤,减压蒸干得红棕色油状物(1.30g,收率43%)。未纯化,直接用于下一步反应。 Add NH 4 Cl (1.81g, 33.80mmol, 1.5eq), water (15.00mL) and ammonia water (4.80mL, 25%, 1.1eq) to a 50mL two-necked reaction flask in turn, stir at room temperature to dissolve, then add intermediate 1 ( 3.00 g, 22.37 mmol, 1.0 eq), stirring was continued for 20 hours. Extract with dichloromethane (15mL*7), combine the organic phases, and dry with 2g of anhydrous sodium sulfate for 5 minutes. Filter and evaporate to dryness under reduced pressure to obtain a reddish-brown oil (1.30 g, yield 43%). Without purification, it was directly used in the next reaction.
步骤3、3-(4-氯-1,2,5-噻二唑-3-基)吡啶(中间体3)的制备Preparation of step 3, 3-(4-chloro-1,2,5-thiadiazol-3-yl)pyridine (intermediate 3)
向250mL三口反应瓶依次加入S 2Cl 2(13.99g,103.62mmol,2.0eq)、DMF(56.00mL)冰水浴降温搅拌,0~5℃滴入DMF(28.00mL)溶清的中间体2(6.90g,51.82mmol,1.0eq)。搅拌反应40分钟,同温滴加9M NaOH(60mL),过滤。水相用二氯甲烷(120mL*3)萃取,有机相合并用纯水(80mL*3)洗涤,无水硫酸钠干燥,过滤。减压蒸干得棕褐色固体(7.99g,收率78%)。 Add S 2 Cl 2 (13.99g, 103.62mmol, 2.0eq) and DMF (56.00mL) to a 250mL three-necked reaction flask in turn, and stir in an ice-water bath, then drop in DMF (28.00mL) at 0-5°C to dissolve Intermediate 2 ( 6.90 g, 51.82 mmol, 1.0 eq). Stir the reaction for 40 minutes, add 9M NaOH (60 mL) dropwise at the same temperature, and filter. The aqueous phase was extracted with dichloromethane (120 mL*3), and the combined organic phases were washed with pure water (80 mL*3), dried over anhydrous sodium sulfate, and filtered. Evaporated to dryness under reduced pressure to obtain a tan solid (7.99 g, yield 78%).
1H NMR(400MHz,CDCl 3)δ9.24(s,1H),8.75(d,J=4.8Hz,1H),8.29(m,1H),7.46(dd,J=8.1,4.9Hz,1H).MS m/z:197.8,199.8[M+H] +. 1 H NMR (400MHz, CDCl 3 ) δ9.24(s, 1H), 8.75(d, J=4.8Hz, 1H), 8.29(m, 1H), 7.46(dd, J=8.1, 4.9Hz, 1H) .MS m/z: 197.8, 199.8[M+H] + .
步骤4、3-(4-己氧基-1,2,5-噻二唑-3-基)吡啶(中间体4)的制备Preparation of Step 4, 3-(4-hexyloxy-1,2,5-thiadiazol-3-yl)pyridine (intermediate 4)
向100mL三口反应瓶依次加入60%NaH(6.21g,155.28mmol,9.0eq)、四氢呋喃(9.00mL),0~5℃滴入四氢呋喃(18.00mL)稀释的正己醇(6.27g,61.47mmol,3.0eq),室温搅拌2小时。将四氢呋喃(15.00mL)溶清的中间体3(3.41g,17.25mmol,1.0eq)于室温滴入体系, 磁力搅拌3小时。反应液加饱和碳酸氢钠水溶液(30mL)洗涤,水相用二氯甲烷(30mL*3)萃取,减压浓缩。粗品柱层析(石油醚(60-90)/乙酸乙酯5:1-3:1)得类白色固体(3.61g,收率79%)。Add 60% NaH (6.21g, 155.28mmol, 9.0eq) and tetrahydrofuran (9.00mL) to a 100mL three-necked reaction flask in sequence, and add n-hexanol (6.27g, 61.47mmol, 3.0 eq), stirred at room temperature for 2 hours. Intermediate 3 (3.41 g, 17.25 mmol, 1.0 eq) dissolved in tetrahydrofuran (15.00 mL) was dropped into the system at room temperature, and magnetically stirred for 3 hours. The reaction solution was washed with saturated aqueous sodium bicarbonate (30 mL), the aqueous phase was extracted with dichloromethane (30 mL*3), and concentrated under reduced pressure. Column chromatography of the crude product (petroleum ether (60-90)/ethyl acetate 5:1-3:1) gave an off-white solid (3.61 g, yield 79%).
1H NMR(400MHz,CDCl 3)δ9.40(d,J=2.2Hz,1H),8.65(dd,J=4.8,1.7Hz,1H),8.43(dt,J=8.0,2.0Hz,1H),7.40(dd,J=8.1,4.8Hz,1H),4.53(t,J=6.6Hz,2H),1.89(m,2H),1.71(s,2H),1.44–1.29(m,4H),0.97–0.87(m,3H).MS m/z:264.0[M+H] +. 1 H NMR (400MHz, CDCl 3 ) δ9.40(d, J=2.2Hz, 1H), 8.65(dd, J=4.8, 1.7Hz, 1H), 8.43(dt, J=8.0, 2.0Hz, 1H) ,7.40(dd,J=8.1,4.8Hz,1H),4.53(t,J=6.6Hz,2H),1.89(m,2H),1.71(s,2H),1.44–1.29(m,4H), 0.97–0.87(m,3H).MS m/z:264.0[M+H] + .
步骤5、碘化-1-氘代甲基-3-(4-己氧基-1,2,5-噻二唑-3-基)吡啶(中间体5)的制备 Step 5, Preparation of iodide-1-deuteromethyl-3-(4-hexyloxy-1,2,5-thiadiazol-3-yl)pyridine (intermediate 5)
向100mL三口反应瓶依次加入中间体4(3.47g,13.2mmol,1.0eq)、丙酮(50.00mL)和氘代碘甲烷(5.72g,39.5mmol,3.0eq)于室温搅拌24小时。中间体5从体系析出,过滤得亮黄色固体(5.18g,收率96%)。Intermediate 4 (3.47g, 13.2mmol, 1.0eq), acetone (50.00mL) and deuteroiodomethane (5.72g, 39.5mmol, 3.0eq) were sequentially added to a 100mL three-neck reaction flask and stirred at room temperature for 24 hours. Intermediate 5 was precipitated from the system and filtered to obtain a bright yellow solid (5.18 g, yield 96%).
1H NMR(400MHz,CDCl 3)δ9.68(m,1H),9.44(m,1H),9.14(dt,J=8.4,1.5Hz,1H),8.29(dd,J=8.3,6.0Hz,1H),4.61(t,J=6.9Hz,2H),1.95(m,2H),1.55–1.45(m,2H),1.45–1.28(m,4H),0.92(t,J=7.0Hz,3H).MS m/z:281.0. 1 H NMR (400MHz, CDCl 3 ) δ9.68(m, 1H), 9.44(m, 1H), 9.14(dt, J=8.4, 1.5Hz, 1H), 8.29(dd, J=8.3, 6.0Hz, 1H), 4.61(t, J=6.9Hz, 2H), 1.95(m, 2H), 1.55–1.45(m, 2H), 1.45–1.28(m, 4H), 0.92(t, J=7.0Hz, 3H ).MS m/z:281.0.
步骤6、氘代占诺美林(式I化合物)的制备 Step 6, the preparation of deuterated phenomeline (compound of formula I)
向250mL三口反应瓶依次加入中间体5(2.00g,4.9mmol,1.0eq)、乙醇(24.00mL)搅拌溶清,-5~0℃滴加NaBH 4(371mg,9.8mmol,2.0eq)的乙醇(16.00mL)悬浮液,同温搅拌1小时。加纯水(100.00mL)淬灭,水相用二氯甲烷(100mL*3)萃取。合并有机相,无水硫酸钠干燥,过滤,减压浓缩。粗品柱层析(二氯甲烷/甲醇20:1)得棕褐色固体(1.11g,收率79%)。熔点:38.26℃。 Add intermediate 5 (2.00g, 4.9mmol, 1.0eq) and ethanol (24.00mL) to a 250mL three-necked reaction flask in sequence, stir to dissolve, and add NaBH 4 (371mg, 9.8mmol, 2.0eq) in ethanol dropwise at -5~0°C (16.00 mL) suspension, stirred at the same temperature for 1 hour. Purified water (100.00 mL) was added to quench, and the aqueous phase was extracted with dichloromethane (100 mL*3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. Column chromatography (dichloromethane/methanol 20:1) of the crude product gave a tan solid (1.11 g, yield 79%). Melting point: 38.26°C.
1H NMR(400MHz,CDCl 3)δ7.07(m,1H),4.44(t,J=6.6Hz,2H),3.46(m,2H),2.58(t,J=5.7Hz,2H),2.46(m,2H),1.88–1.79(m,2H),1.46(m,2H),1.34(m,4H),0.91–1.79(m,3H).MS m/z:285.1[M+H] +. 1 H NMR (400MHz, CDCl 3 ) δ7.07(m, 1H), 4.44(t, J=6.6Hz, 2H), 3.46(m, 2H), 2.58(t, J=5.7Hz, 2H), 2.46 (m,2H),1.88–1.79(m,2H),1.46(m,2H),1.34(m,4H),0.91–1.79(m,3H).MS m/z:285.1[M+H] + .
实施例2.式I化合物L-苹果酸盐A晶型的制备 Embodiment 2. The preparation of formula I compound L-malate A crystal form
称取式I化合物(10.0g,35.2mmol)、L-苹果酸(4.81g,35.8mmol)于烧瓶中,加入异丙醇70mL,升温至60~70℃,搅拌混合,使得混合物完全溶解,趁热过滤,得澄清液,冷却、将温度降至2~8℃、搅拌析晶,过滤得产物,用冰异丙醇洗涤,得到湿产物,将湿产物置于40℃真空干燥,干燥后得到白色固体(13.83g,收率93.9%),熔点1 11.03℃。 1H NMR(400MHz,CDCl 3)δ7.22(m,1H),4.47(t,J=6.7Hz,2H),4.25(dd,J=9.6,4.5Hz,1H),4.12(s,2H),3.29(s,2H),2.85–2.69(m,4H),1.85(m,2H),1.50–1.41(m,2H),1.35(m,4H),0.95–0.86(t,J=6.5Hz 3H).MS m/z:285.1[M+H] +. Weigh the compound of formula I (10.0g, 35.2mmol) and L-malic acid (4.81g, 35.8mmol) into a flask, add 70mL of isopropanol, heat up to 60-70°C, stir and mix, so that the mixture is completely dissolved. Filtrate hot to obtain a clear liquid, cool, lower the temperature to 2-8°C, stir and crystallize, filter to obtain the product, wash with ice isopropanol to obtain a wet product, place the wet product at 40°C for vacuum drying, and obtain White solid (13.83g, yield 93.9%), melting point 11.03°C. 1 H NMR (400MHz, CDCl 3 ) δ7.22(m, 1H), 4.47(t, J=6.7Hz, 2H), 4.25(dd, J=9.6, 4.5Hz, 1H), 4.12(s, 2H) ,3.29(s,2H),2.85–2.69(m,4H),1.85(m,2H),1.50–1.41(m,2H),1.35(m,4H),0.95–0.86(t,J=6.5Hz 3H).MS m/z:285.1[M+H] + .
将所得白色固体送检XRPD、DSC和TGA,经测试,发现所得白色固体以晶体形态存在,所得晶型命名为式I化合物L-苹果酸盐A晶型,简称A晶型。所得A晶型的XRPD图、DSC图和TGA图基本上分别如图1、图2和图3所示。所得晶体的X射线粉末衍射图谱中,特征 峰的峰位置及强度如表2所示;所得晶型的XRPD图谱的衍射角数据基本如表3所示,其中2θ值误差范围为±0.2°。The obtained white solid was submitted for inspection by XRPD, DSC and TGA. After testing, it was found that the obtained white solid existed in the form of crystals, and the obtained crystal form was named as the compound of formula I L-malate A crystal form, referred to as A crystal form. The XRPD pattern, DSC pattern and TGA pattern of the obtained crystal form A are basically shown in Fig. 1 , Fig. 2 and Fig. 3 respectively. In the X-ray powder diffraction pattern of the obtained crystal, the peak positions and intensities of the characteristic peaks are shown in Table 2; the diffraction angle data of the XRPD pattern of the obtained crystal form are basically shown in Table 3, and the error range of the 2θ value is ±0.2°.
表2.A晶型的X射线粉末衍射图谱的特征峰的峰位置及强度The peak position and intensity of the characteristic peaks of the X-ray powder diffraction pattern of Table 2.A crystal form
编号serial number 2θ角(°)2θ angle (°) 相对强度(%)Relative Strength(%) 编号serial number 2θ角(°)2θ angle (°) 相对强度(%)Relative Strength(%)
11 12.00712.007 26.326.3 1010 20.01220.012 39.939.9
22 12.75512.755 3.93.9 1111 20.32720.327 26.326.3
33 13.28813.288 4.74.7 1212 20.66020.660 8.68.6
44 13.90313.903 7.47.4 1313 21.94221.942 100.0100.0
55 14.82714.827 25.725.7 1414 22.47322.473 24.024.0
66 15.39915.399 20.320.3 1515 24.99624.996 21.121.1
77 16.79616.796 21.621.6 1616 25.33325.333 30.430.4
88 17.68617.686 6.16.1 1717 27.38227.382 7.87.8
99 18.90618.906 11.311.3 1818 27.71827.718 17.517.5
表3.A晶型的XRPD解析数据Table 3. XRPD analysis data of crystal form A
Figure PCTCN2022124662-appb-000006
Figure PCTCN2022124662-appb-000006
实施例3、式I化合物L-苹果酸盐A晶型的制备Embodiment 3, preparation of formula I compound L-malate A crystal form
向100mL烧瓶中加入式I化合物(10.0g,35.2mmol),L-苹果酸(4.72g,35.2mmol)和异丙醇40mL,加热混合物直至完全溶解,趁热过滤、得澄清液。在搅拌下缓慢冷却溶液至2~8℃析晶,过滤收集产物,冷异丙醇洗涤,在40℃真空干燥,得到白色固体(13.3g,收率90.3%)。经测试所得产物为式I化合物L-苹果酸盐A晶型。Add the compound of formula I (10.0 g, 35.2 mmol), L-malic acid (4.72 g, 35.2 mmol) and 40 mL of isopropanol into a 100 mL flask, heat the mixture until completely dissolved, and filter while hot to obtain a clear liquid. The solution was cooled slowly under stirring to 2-8°C for crystallization, and the product was collected by filtration, washed with cold isopropanol, and dried in vacuum at 40°C to obtain a white solid (13.3 g, yield 90.3%). The product obtained through testing is the crystal form A of compound L-malate of formula I.
实施例4.式I化合物L-苹果酸盐A晶型的制备Example 4. Preparation of Form A of Compound L-Malate of Formula I
向100mL烧瓶中加入式I化合物(4.9g,17.2mmol),L-苹果酸(2.54g,18.9mmol) 和甲醇5mL,加热混合物直至完全溶解,趁热过滤、得澄清液。然后向其加入34mL乙酸乙酯。在搅拌下缓慢冷却溶液至2~8℃析晶,过滤收集产物,冷乙酸乙酯洗涤,在40℃真空干燥,得到白色固体(6.6g,收率91.7%)。经测试所得产物为式I化合物L-苹果酸盐A晶型。Add the compound of formula I (4.9 g, 17.2 mmol), L-malic acid (2.54 g, 18.9 mmol) and 5 mL of methanol into a 100 mL flask, heat the mixture until it is completely dissolved, and filter while hot to obtain a clear liquid. Then 34 mL of ethyl acetate was added thereto. The solution was cooled slowly under stirring to 2-8°C for crystallization, and the product was collected by filtration, washed with cold ethyl acetate, and dried in vacuum at 40°C to obtain a white solid (6.6 g, yield 91.7%). The product obtained through testing is the crystal form A of compound L-malate of formula I.
实施例5.式I化合物L-苹果酸盐A晶型的制备Example 5. Preparation of Form A of Compound L-Malate of Formula I
向100mL烧瓶中加入式I化合物(5.0g,17.6mmol),L-苹果酸(2.83g,21.1mmol)和异丙醇12mL,加热混合物直至完全溶解,趁热过滤、得澄清液。然后向其加入38mL乙酸乙酯。在搅拌下缓慢冷却溶液至2~8℃析晶,过滤收集产物,冷异丙醇洗涤,在40℃真空干燥,得到白色固体(6.8g,收率92.4%)。经测试所得产物为式I化合物苹果酸盐A晶型。Add the compound of formula I (5.0 g, 17.6 mmol), L-malic acid (2.83 g, 21.1 mmol) and 12 mL of isopropanol into a 100 mL flask, heat the mixture until completely dissolved, and filter while hot to obtain a clear liquid. Then 38 mL of ethyl acetate was added thereto. The solution was cooled slowly under stirring to 2-8°C for crystallization, and the product was collected by filtration, washed with cold isopropanol, and dried in vacuum at 40°C to obtain a white solid (6.8 g, yield 92.4%). The product obtained through testing is the crystal form A of malate salt of the compound of formula I.
实施例6.式I化合物L-苹果酸盐A晶型的制备Example 6. Preparation of Form A of Compound L-Malate of Formula I
向50mL烧瓶中加入式I化合物(1.0g,3.52mmol),L-苹果酸(0.6g,4.47mmol)和甲醇0.5mL,甲基叔丁基醚0.5mL,加热混合物直至完全溶解,趁热过滤、得澄清液。然后向溶液中补加5mL甲基叔丁基醚。在搅拌下缓慢冷却溶液至2~8℃析晶,过滤收集产物,冷甲基叔丁基醚洗涤,在40℃真空干燥,得到白色固体(1.3g,收率88.4%)。经测试所得产物为式I化合物苹果酸盐A晶型。Add formula I compound (1.0g, 3.52mmol), L-malic acid (0.6g, 4.47mmol) and 0.5mL of methanol, 0.5mL of methyl tert-butyl ether into a 50mL flask, heat the mixture until completely dissolved, and filter while hot , Obtain clarification liquid. An additional 5 mL of methyl tert-butyl ether was then added to the solution. The solution was cooled slowly under stirring to 2-8°C for crystallization, and the product was collected by filtration, washed with cold methyl tert-butyl ether, and dried in vacuum at 40°C to obtain a white solid (1.3 g, yield 88.4%). The product obtained through testing is the crystal form A of malate salt of the compound of formula I.
实施例7.式I化合物硝酸盐的制备Embodiment 7. the preparation of formula I compound nitrate
向25mL烧瓶中加入式I化合物1085mg,乙酸乙酯3mL,搅拌混合物直至完全溶解,趁热过滤、得澄清液。在搅拌下向其加入硝酸至pH值介于3~4之间,然后向其补加甲基叔丁基醚12mL,搅拌析晶,过滤收集产物,冷甲基叔丁基醚洗涤,在40℃真空干燥,得到白色固体1000mg,收率75.5%。经测试所得产物为式I化合物硝酸盐。熔点:64.5℃(DSC)。 1H NMR(400MHz,CDCl 3):δ12.31(brs,1H),7.26–7.23(m,1H),4.75–4.38(m,1H),4.47(t,J=6.7Hz,2H),4.00–3.56(m,2H),3.26–2.86(m,2H),2.73–2.49(m,1H),1.85(quint,J=6.8Hz,2H),1.57–1.25(m,6H),0.91(t,J=6.9Hz,3H).MS m/z:285.2[M+H] +. Add 1085 mg of the compound of formula I and 3 mL of ethyl acetate into a 25 mL flask, stir the mixture until it is completely dissolved, and filter while hot to obtain a clear liquid. Add nitric acid to it under stirring until the pH value is between 3 and 4, then add 12 mL of methyl tert-butyl ether to it, stir and crystallize, collect the product by filtration, wash with cold methyl tert-butyl ether, °C and dried in vacuo to obtain 1000 mg of white solid with a yield of 75.5%. The product obtained through testing is the compound nitrate of formula I. Melting point: 64.5°C (DSC). 1 H NMR (400MHz, CDCl 3 ): δ12.31 (brs, 1H), 7.26–7.23 (m, 1H), 4.75–4.38 (m, 1H), 4.47 (t, J=6.7Hz, 2H), 4.00 –3.56(m,2H),3.26–2.86(m,2H),2.73–2.49(m,1H),1.85(quint,J=6.8Hz,2H),1.57–1.25(m,6H),0.91(t ,J=6.9Hz,3H).MS m/z:285.2[M+H] + .
实施例8.式I化合物丁二酸盐的制备 Embodiment 8. Preparation of formula I compound succinate
向50mL烧瓶中加入式I化合物505mg,丁二酸210mg和异丙醇2mL,加热混合物直至完全溶解,趁热过滤、得澄清液,然后向其加入18mL乙酸乙酯。在搅拌下缓慢冷却溶液至2~8℃析晶,过滤收集产物,冷乙酸乙酯洗涤,在40℃真空干燥,得到白色固体630mg,收率88.2%。经测试所得产物为式I化合物丁二酸盐。熔点:93.4℃(DSC)。 1H NMR(400MHz,CDCl 3):δ7.23–7.12(m,1H),4.46(t,J=6.7Hz,2H),4.05–3.85(m,2H),3.22–2.98(m,2H),2.77–2.52(m,2H),2.57(s,4H),1.85(quint,J=6.8Hz,2H),1.55–1.27(m,6H),0.91(t,J=6.6Hz,3H).MS m/z:285.1[M+H] +. Add 505 mg of the compound of formula I, 210 mg of succinic acid and 2 mL of isopropanol to a 50 mL flask, heat the mixture until it is completely dissolved, filter while hot to obtain a clear liquid, and then add 18 mL of ethyl acetate to it. The solution was slowly cooled under stirring to 2-8°C for crystallization, and the product was collected by filtration, washed with cold ethyl acetate, and dried in vacuum at 40°C to obtain 630 mg of white solid with a yield of 88.2%. The product obtained through testing is the compound succinate of formula I. Melting point: 93.4°C (DSC). 1 H NMR (400MHz, CDCl 3 ): δ7.23–7.12 (m, 1H), 4.46 (t, J=6.7Hz, 2H), 4.05–3.85 (m, 2H), 3.22–2.98 (m, 2H) ,2.77–2.52(m,2H),2.57(s,4H),1.85(quint,J=6.8Hz,2H),1.55–1.27(m,6H),0.91(t,J=6.6Hz,3H). MS m/z:285.1[M+H] + .
实施例9.式I化合物对甲苯磺酸盐的制备Embodiment 9. Preparation of formula I compound tosylate
向50mL烧瓶中加入式I化合物501mg,对甲苯磺酸337mg和异丙醇2mL,加热混合物直至完全溶解,趁热过滤、得澄清液,然后向其加入18mL乙酸乙酯。在搅拌下缓慢冷却溶液至2~8℃析晶,过滤收集产物,冷乙酸乙酯洗涤,在40℃真空干燥,得到白色固体720mg,收率90%。经测试所得产物为式I化合物对甲苯磺酸盐。 1H NMR(400MHz,CDCl 3):δ11.41(brs,1H),7.72(d,J=7.9Hz,2H),7.23–7.16(m,1H),7.12(d,J=7.9Hz,2H),4.60–4.40(m,1H),4.45(t,J=6.7Hz,2H),3.90–3.66(m,2H),3.20–2.94(m,2H),2.70–2.45(m,1H),2.33(s,3H),1.84(quint,J=6.8Hz,1H),1.56–1.18(m,6H),0.91(t,J=6.8Hz,3H).MS m/z:285.1[M+H] +. Add 501 mg of the compound of formula I, 337 mg of p-toluenesulfonic acid and 2 mL of isopropanol into a 50 mL flask, heat the mixture until it is completely dissolved, filter while hot to obtain a clear liquid, and then add 18 mL of ethyl acetate to it. The solution was cooled slowly under stirring to 2-8°C for crystallization, and the product was collected by filtration, washed with cold ethyl acetate, and dried in vacuum at 40°C to obtain 720 mg of a white solid with a yield of 90%. The product obtained through testing is the p-toluenesulfonate of the compound of formula I. 1 H NMR (400MHz, CDCl 3 ): δ11.41(brs, 1H), 7.72(d, J=7.9Hz, 2H), 7.23–7.16(m, 1H), 7.12(d, J=7.9Hz, 2H ),4.60–4.40(m,1H),4.45(t,J=6.7Hz,2H),3.90–3.66(m,2H),3.20–2.94(m,2H),2.70–2.45(m,1H), 2.33(s,3H),1.84(quint,J=6.8Hz,1H),1.56–1.18(m,6H),0.91(t,J=6.8Hz,3H).MS m/z:285.1[M+H ] + .
实施例10.式I化合物酒石酸盐的制备 Embodiment 10. Preparation of formula I compound tartrate
向100mL烧瓶中加入式I化合物3.0g,L-酒石酸1.6g和异丙醇15mL,加热混合物直至完全溶解,趁热过滤、得澄清液,然后向其加入45mL乙酸乙酯。在搅拌下缓慢冷却溶液至2~8℃析晶,过滤收集产物,冷乙酸乙酯洗涤,在40℃真空干燥,得到白色固体4.2g,收率91.5%。经测试所得产物为式I化合物酒石酸盐。 1H NMR(400MHz,CDCl 3)δ7.16(1H,brs),4.43(4H,m),4.09(2H,brs),3.29(2H,brs),2.69(2H,brs),1.83(2H,m),1.43(2H,m),1.40(4H,m),0.9(t,J=6.5Hz,3H).MS m/z:285.1[M+H] +. Add 3.0 g of the compound of formula I, 1.6 g of L-tartaric acid and 15 mL of isopropanol to a 100 mL flask, heat the mixture until it is completely dissolved, filter while hot to obtain a clear liquid, and then add 45 mL of ethyl acetate to it. The solution was cooled slowly under stirring to 2-8°C for crystallization, and the product was collected by filtration, washed with cold ethyl acetate, and dried in vacuum at 40°C to obtain 4.2 g of white solid, with a yield of 91.5%. The product obtained through testing is the compound tartrate of formula I. 1 H NMR (400MHz, CDCl 3 ) δ7.16 (1H, brs), 4.43 (4H, m), 4.09 (2H, brs), 3.29 (2H, brs), 2.69 (2H, brs), 1.83 (2H, m),1.43(2H,m),1.40(4H,m),0.9(t,J=6.5Hz,3H).MS m/z:285.1[M+H] + .
实施例11.式I化合物其他盐的制备Embodiment 11. Preparation of other salts of compounds of formula I
选取甲磺酸、苯磺酸、苯甲酸、丙二酸、己二酸,参照实施例2-10的制备方法与式I化合物进行成盐,但均未得到固体形式的式I化合物的盐。Select methanesulfonic acid, benzenesulfonic acid, benzoic acid, malonic acid, adipic acid, and refer to the preparation method of Example 2-10 to form a salt with the compound of formula I, but none of the salts of the compound of formula I in solid form were obtained.
测试例1实施例1-10合成的各化合物和盐的理化性质测定实验The physical and chemical property determination experiment of each compound and salt synthesized in Test Example 1 Example 1-10
1.熔点测定实验1. Melting point determination experiment
通过差式扫描量热仪(DSC)测定实施例1-10合成的式I化合物(实施例1所得样品)、式I化合物L-苹果酸盐A晶型(实施例2所得样品)、式I化合物硝酸盐(实施例7所得样品)、式I化合物丁二酸盐(实施例8所得样品)、式I化合物对甲苯磺酸盐(实施例9所得样品)、式I化合物酒石酸盐(实施例10所得样品)的熔点,不同盐型的熔点见表4。Measure the compound of formula I synthesized in Examples 1-10 (the sample obtained in Example 1), the compound of formula I L-malate A crystal form (the sample obtained in Example 2) and the compound of formula I by differential scanning calorimetry (DSC). Compound nitrate (samples gained in Example 7), compound succinate of formula I (samples gained in Example 8), compound p-toluenesulfonate of formula I (samples gained in Example 9), compound tartrate of formula I (samples gained in Example 8) 10 obtained sample), the melting points of different salt forms are shown in Table 4.
2.溶解度测定实验2. Solubility determination experiment
取七份2mL纯化水分别逐次向其中加入式I化合物(实施例1所得样品)、式I化合物L-苹果酸盐A晶型(实施例2所得样品)、式I化合物硝酸盐(实施例7所得样品)、式I化合物丁二酸盐(实施例8所得样品)、式I化合物对甲苯磺酸盐(实施例9所得样品)、 式I化合物酒石酸盐(实施例10所得样品)至不溶,并记录相应用量m,目测溶解度S=m/2mL。另搅拌24h后,取上层清液送HPLC分析、所测得溶液浓度即为HPLC分析方法测定溶解度。不同盐型在水中溶解度结果见表4。本实验溶解度均在环境温度(实测环境温度为10℃)中测定。Get seven parts of 2mL purified water and add the compound of formula I (the sample obtained in Example 1), the compound of formula I L-malate A crystal form (the sample obtained in Example 2), the compound nitrate of formula I (the sample obtained in Example 7) to it successively. Gained sample), formula I compound succinate (embodiment 8 gained sample), formula I compound p-toluenesulfonate (embodiment 9 gained sample), formula I compound tartrate (embodiment 10 gained sample) until insoluble, And record the corresponding dosage m, visual solubility S=m/2mL. After stirring for another 24 hours, take the supernatant and send it to HPLC for analysis, and the measured solution concentration is the HPLC analysis method to determine the solubility. The solubility results of different salt types in water are shown in Table 4. The solubility in this experiment was measured at ambient temperature (the measured ambient temperature was 10°C).
3、吸湿性考察3. Investigation of hygroscopicity
取上述实施例1、2、7-10合成的各样品适量,精密称定,称得重量为m1。将样品平摊放置于扁形称量瓶中,称得总重为m2,于25±1℃、80±1%RH条件下放置24h后,称重为m3。通过下式计算得到吸湿增重ΔW,ΔW=(m3-m2)/m1*100%,不同盐型吸湿增重结果见表4。Take an appropriate amount of each sample synthesized in the above-mentioned Examples 1, 2, 7-10, accurately weigh it, and weigh it as m1. Place the sample flat in a flat weighing bottle, and weigh the total weight as m2. After placing it at 25±1°C and 80±1% RH for 24 hours, weigh it as m3. The moisture absorption weight gain ΔW is calculated by the following formula, ΔW=(m3-m2)/m1*100%, and the results of moisture absorption weight gain of different salt types are shown in Table 4.
吸湿性评价指标参见表5:See Table 5 for hygroscopicity evaluation index:
表4.各样品的理化性质Table 4. Physicochemical properties of each sample
样品sample 熔点/℃Melting point/℃ 溶解度S(mg/mL)Solubility S(mg/mL) 吸湿增重ΔW/%Moisture absorption weight gain ΔW/%
式I化合物(实施例1)Formula I compound (embodiment 1) 38.2638.26 S<1.07 a S< 1.07a NANA
式I化合物L-苹果酸盐A晶型(实施例2)Formula I compound L-malate A crystal form (Example 2) 111.03111.03 40.48 a 40.48 a 1.971.97
式I化合物硝酸盐(实施例7)Formula I compound nitrate (embodiment 7) 64.5964.59 S>244.2 b S> 244.2b 吸湿后形成液体liquid after moisture absorption
式I化合物丁二酸盐(实施例8)Formula I compound succinate (embodiment 8) 93.4393.43 7.99 a 7.99a 17.0017.00
式I化合物对甲苯磺酸盐(实施例9)Formula I compound tosylate (embodiment 9) 112.50112.50 5.39 a 5.39a 1.001.00
式I化合物酒石酸盐(实施例10)Formula I compound tartrate (embodiment 10) 95.995.9 86.05<S<98.9 b 86.05<S< 98.9b 3.033.03
表4中,a:使用HPLC法测定溶解度;b:使用目测法测定溶解度。NA表示未测定。In Table 4, a: Solubility was measured by HPLC; b: Solubility was measured by visual inspection. NA means not determined.
表5.吸湿性评价指标Table 5. Hygroscopicity evaluation index
吸湿性分类Hygroscopicity Classification 吸湿增重(ΔW%)Moisture weight gain (ΔW%)
潮解deliquescence 吸收足量的水分形成液体Absorb enough water to form a liquid
极具吸湿性Very hygroscopic ΔW%≥15%ΔW%≥15%
有吸湿性Hygroscopic 2%≤ΔW%<15%2%≤ΔW%<15%
略有吸湿性slightly hygroscopic 0.2%≤ΔW%<2%0.2%≤ΔW%<2%
无或几乎无吸湿性No or almost no hygroscopicity ΔW%<0.2%ΔW%<0.2%
熔点测定实验结果:式I化合物L-苹果酸盐的熔点为111.03℃,式I化合物L-苹果酸盐的稳定性较好。Experimental results of melting point determination: the melting point of compound L-malate of formula I is 111.03°C, and the compound L-malate of formula I has good stability.
溶解度测定实验结果:式I化合物L-苹果酸盐的溶解度为40.48mg/mL,式I化合物L- 苹果酸盐的溶解度较好。Solubility measurement experiment results: the solubility of the compound L-malate of the formula I is 40.48 mg/mL, and the solubility of the compound L-malate of the formula I is better.
吸湿性实验结果:式I化合物L-苹果酸盐在25±1℃、80±1%RH条件下的吸湿增重为1.97%,说明式I化合物L-苹果酸盐为略有吸湿性。Hygroscopicity test results: the moisture absorption weight gain of compound L-malate of formula I was 1.97% under the conditions of 25±1° C. and 80±1% RH, indicating that compound L-malate of formula I was slightly hygroscopic.
实验结果分析:式I化合物L-苹果酸盐具有较高的熔点(即具备较高稳定性)、较低的吸湿性、并且有适当的溶解度,这使得式I化合物苹果酸盐适用于制备口服固体制剂,特别是片剂的制备。Experimental result analysis: formula I compound L-malate has higher fusing point (promptly having higher stability), lower hygroscopicity, and suitable solubility is arranged, and this makes formula I compound malate be suitable for preparing oral Preparation of solid dosage forms, especially tablets.
测试例2.实施例2、7-10所制备的盐的物理稳定性研究Test example 2. The physical stability research of the prepared salt of embodiment 2,7-10
将式I化合物L-苹果酸盐A晶型(实施例2所得样品)、式I化合物硝酸盐(实施例7所得样品)、式I化合物丁二酸盐(实施例8所得样品)、式I化合物对甲苯磺酸盐(实施例9所得样品)、式I化合物酒石酸盐(实施例10所得样品)分别敞口平摊放置,在高温(60℃)、高湿(RH92.5%)、光照(4500±500Lux)条件下进行样品的稳定性试验,测定在不同取样时间(0天、6天、14天、30天)样品中活性物质(式I化合物或占诺美林)和有关物质的含量,具体如表6所示。The formula I compound L-malate A crystal form (the sample obtained in Example 2), the formula I compound nitrate (the sample obtained in Example 7), the formula I compound succinate (the sample obtained in Example 8), the formula I Compound p-toluenesulfonate (samples obtained in Example 9), compound tartrate of formula I (samples obtained in Example 10) are respectively exposed and laid flat, and placed under high temperature (60° C.), high humidity (RH92.5%), light Carry out the stability test of sample under (4500 ± 500Lux) condition, measure in different sampling time (0 day, 6 days, 14 days, 30 days) in the sample active substance (formula I compound or phenomeline) and related substances content, as shown in Table 6.
表6.盐型的稳定性实验Table 6. Stability experiments of salt forms
Figure PCTCN2022124662-appb-000007
Figure PCTCN2022124662-appb-000007
Figure PCTCN2022124662-appb-000008
Figure PCTCN2022124662-appb-000008
表6中,a:式1化合物的硝酸盐,在高温条件下为液态,物理性质不稳定。NA表示未测定。In Table 6, a: the nitrate of the compound of formula 1 is liquid under high temperature conditions, and its physical properties are unstable. NA means not determined.
由上表6中的结果可见,本发明的式I化合物L-苹果酸盐A晶型的杂质含量很低,且在高温、高湿及光照条件下稳定性非常好,具有良好的成药前景;出乎意料的,式I化合物苹果酸盐A晶型在光照条件下相对其他盐型更加稳定,便于原料药及药物制剂的储存。From the results in Table 6 above, it can be seen that the crystal form A of compound L-malate of formula I of the present invention has very low impurity content, and has very good stability under high temperature, high humidity and light conditions, and has a good prospect for pharmaceutical preparation; Unexpectedly, the malate A crystal form of the compound of formula I is more stable under light conditions than other salt forms, which is convenient for the storage of raw materials and pharmaceutical preparations.
测试例3.不同盐型的毒副作用研究Test example 3. Toxic and side effects research of different salt types
实验材料:SD大鼠(购自北京维通利华实验动物技术有限公司,生产许可证号:SCXK(京)2016-0006)、式I化合物(实施例1)、式I化合物苹果酸盐(实施例2)、式I化合物酒石酸盐(实施例10)、纯化水(自制)。Experimental materials: SD rats (purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd., production license number: SCXK (Beijing) 2016-0006), formula I compound (Example 1), formula I compound malate ( Embodiment 2), formula I compound tartrate (embodiment 10), purified water (self-made).
实验方法:将24只SD大鼠随机分为4组(每组6只),经口灌胃给药,分别按30mg/kg(以游离碱量计)的剂量给予式I化合物苹果酸盐(实施例2所得样品)水溶液、式I化合物酒石酸盐(实施例10所得样品)水溶液、占诺美林(实施例1所得样品)水溶液及适量的纯化水。给药前、后分别观察SD大鼠的精神状态,并观察给药后各组SD大鼠的流涎情况,记录流涎大鼠的只数。给药前及给药过程中如发生任何异常,需及时记录,具体结果见表7。Experimental method: 24 SD rats are randomly divided into 4 groups (6 in every group), administered by oral gavage, and given formula I compound malate ( Example 2 obtained sample) aqueous solution, formula I compound tartrate (embodiment 10 obtained sample) aqueous solution, phenomeline (embodiment 1 obtained sample) aqueous solution and an appropriate amount of purified water. The mental state of the SD rats was observed before and after the administration, and the salivation of the SD rats in each group was observed after the administration, and the number of the salivating rats was recorded. If any abnormality occurs before and during the administration, it should be recorded in time, and the specific results are shown in Table 7.
表7.不同盐型诱导的副反应Table 7. Side effects induced by different salt types
组别group 流涎大鼠的只数number of salivating rats
式I化合物L-苹果酸盐A晶型(实施例2)Formula I compound L-malate A crystal form (Example 2) 00
式I化合物酒石酸盐(实施例10)Formula I compound tartrate (embodiment 10) 11
式I化合物(实施例1)Formula I compound (embodiment 1) 11
纯化水purified water 00
实验结果分析:占诺美林是一种M受体激活剂,其不仅能作用于中枢神经系统,其也可以刺激外周神经组织中的M受体,从而导致胆碱能副作用,如流涎、恶心、头晕等。本实验通过观察各组SD大鼠的流涎情况来评价各化合物的毒副作用。通过实验结果可知式I化合物 L-苹果酸盐A晶型的胆碱能副作用小,具有良好的安全性。Analysis of experimental results: Zanomeline is an M receptor activator, which can not only act on the central nervous system, but also stimulate M receptors in peripheral nervous tissue, resulting in cholinergic side effects, such as salivation, nausea , dizziness, etc. In this experiment, the toxic and side effects of each compound were evaluated by observing the salivation of SD rats in each group. According to the experimental results, it can be seen that the cholinergic side effect of the crystal form A of the compound L-malate of formula I is small and has good safety.
测试例4式I化合物L-苹果酸盐的多晶型研究Polymorphic Research of Test Example 4 Formula I Compound L-Malate
将通过实施例2方法制备得到的式I化合物L-苹果酸盐A晶型在表8所示溶剂中悬浮打浆,在40℃下避光搅拌2天,将溶液离心去沉淀干燥后经XRPD检测,结果如表8所示:The crystal form A of compound L-malate prepared by the method in Example 2 was suspended and beaten in the solvent shown in Table 8, stirred at 40°C in the dark for 2 days, the solution was centrifuged to remove the precipitate and dried, and then detected by XRPD , the results are shown in Table 8:
表8.测试溶剂Table 8. Test Solvents
Figure PCTCN2022124662-appb-000009
Figure PCTCN2022124662-appb-000009
经上表中的结果分析得出:晶型A稳定性良好,在不同溶剂体系下均能保持稳定。According to the analysis of the results in the above table, it can be concluded that the crystal form A has good stability and can be kept stable under different solvent systems.
测试例5.式I化合物L-苹果酸盐A晶型的稳定性加速实验研究Test Example 5. Accelerated Experimental Study on the Stability of Formula I Compound L-Malate A Crystal Form
将实施例2方法制得的式I化合物L-苹果酸盐A晶型样品敞口平摊放置,考察在高温(60℃)、高湿(RH92.5%)和光照条件(4500±500Lux)下样品的化学稳定性,分别在0天、7天、14天和30天取一定量样品检测样品纯度(用HPLC检测)及样品晶型,来评价氘代占诺美林苹果酸盐A晶型稳定性,实验结果见表9。Place the sample of formula I compound L-malate A crystal form prepared by the method of Example 2 open and flat, and investigate the conditions of high temperature (60°C), high humidity (RH92.5%) and light conditions (4500 ± 500Lux) Under the chemical stability of the sample, a certain amount of samples were taken at 0 days, 7 days, 14 days and 30 days to detect the purity of the samples (detected by HPLC) and the crystal form of the samples to evaluate the deuterium phenomeline malate A crystal Type stability, the experimental results are shown in Table 9.
表9.A晶型物理化学稳定性实验结果Table 9. Physicochemical stability test results of crystal form A
Figure PCTCN2022124662-appb-000010
Figure PCTCN2022124662-appb-000010
经过实验及图4、图5及图6所示的XRPD图谱可知,式I化合物苹果酸盐A晶型在高温(60℃)、高湿(RH92.5%)和光照条件(4500±500Lux)条件下,0天、6天、14天、30 天时刻点的XRPD图谱与图1中A晶型的XRPD图谱基本相同,A晶型的稳定性良好。在高温(60℃)、高湿(RH92.5%)和光照条件(4500±500Lux)下,经过30天的稳定性加速实验,A晶型的纯度基本保持不变,没有杂质产生,A晶型没有发生改变。Through experiments and the XRPD patterns shown in Fig. 4, Fig. 5 and Fig. 6, it can be seen that the crystal form of malate A of the compound of formula I is stable under high temperature (60°C), high humidity (RH92.5%) and light conditions (4500±500Lux) Under these conditions, the XRPD patterns at the time points of 0 day, 6 days, 14 days, and 30 days are basically the same as the XRPD patterns of crystal form A in Figure 1, and the stability of crystal form A is good. Under high temperature (60°C), high humidity (RH92.5%) and light conditions (4500±500Lux), after 30 days of accelerated stability experiments, the purity of Form A remained basically unchanged, and no impurities were produced. The type has not changed.
测试例6.不同盐型的药代动力学研究Test example 6. Pharmacokinetic study of different salt forms
实验材料:雄性SD大鼠(体重180-220g,购自北京维通利华实验动物技术有限公司,生产许可证号:SCXK(京)2016-0006)、式I化合物L-苹果酸盐A晶型(实施例2)、式I化合物(实施例1)、纯化水(自制)。Experimental materials: male SD rats (weight 180-220g, purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd., production license number: SCXK (Beijing) 2016-0006), formula I compound L-malate A crystal Type (Example 2), formula I compound (Example 1), purified water (self-made).
实验方法:将6只雄性SD大鼠随机分为2组(每组3只),试验期间自由饮水,给药前禁食12小时以上,给药后4小时喂食。经口灌胃给药,对两组SD大鼠分别按30mg/kg(以游离碱量计)的剂量给予式I化合物苹果酸盐A晶型水溶液、式I化合物水溶液。给药前0min,给药后15min、30min、45min、1.0h、1.5h、2h、3h、4h、6h、8h、和10h各采集血样至K 2EDTA抗凝管中,在冰上暂存至离心。采血后60min内需离心出血浆(2-8℃条件下,以8000rpm离心5min),离心后将血浆转移至96孔板或离心管中,冰盒转运,≤-15℃保存至LC-MS/MS检测。采用LC-MS/MS生物分析方法检测SD大鼠血浆中的药物浓度,采用非房室模型,使用WinNonlin TM(Version8.3,Certara,USA)对血药浓度-时间数据进行分析,评估其在SD大鼠体内药物代谢动力学(PK)特性,数据见表10。 Experimental method: 6 male SD rats were randomly divided into 2 groups (3 rats in each group), free to drink water during the test period, fasted for more than 12 hours before administration, and fed 4 hours after administration. Oral gavage administration, two groups of SD rats were given formula I compound malate A crystal form A aqueous solution and formula I compound aqueous solution at a dose of 30 mg/kg (calculated as free base). 0min before administration, 15min, 30min, 45min, 1.0h, 1.5h, 2h, 3h, 4h, 6h, 8h, and 10h after administration, blood samples were collected into K 2 EDTA anticoagulant tubes, and temporarily stored on ice until centrifugal. Plasma needs to be centrifuged within 60 minutes after blood collection (at 2-8°C, centrifuge at 8000rpm for 5min), after centrifugation, transfer the plasma to a 96-well plate or centrifuge tube, transport it in an ice box, and store it at ≤ -15°C for LC-MS/MS detection. The LC-MS/MS bioanalysis method was used to detect the drug concentration in SD rat plasma, and the non-compartmental model was used to analyze the blood drug concentration-time data using WinNonlin TM (Version8.3, Certara, USA) to evaluate its SD rat pharmacokinetic (PK) characteristics in vivo, the data are shown in Table 10.
表10.不同化合物的药代动力学参数Table 10. Pharmacokinetic parameters of different compounds
Figure PCTCN2022124662-appb-000011
Figure PCTCN2022124662-appb-000011
通过表10中数据可以看出:对于大鼠给予式I化合物L-苹果酸盐A晶型后,T max(达峰时间)时间短、能够快速的达到治疗有效浓度;C max(达峰浓度)适中,在较好起到治疗效果的同时,能够有效的降低胆碱能副作用,且具有较好的生物利用度。 As can be seen from the data in Table 10: after giving rats the compound L-malate A crystal form of formula I, the Tmax (peak time) time is short and can quickly reach the therapeutically effective concentration; Cmax (peak concentration ) is moderate, it can effectively reduce cholinergic side effects while playing a good therapeutic effect, and has good bioavailability.
以上,对本发明的实施方式进行了说明。但是,本发明不限定于上述实施方式。凡在本发明的精神和原则之内,所做的任何修改、等同替换、改进等,均应包含在本发明的保护范围之内。The embodiments of the present invention have been described above. However, the present invention is not limited to the above-mentioned embodiments. Any modifications, equivalent replacements, improvements, etc. made within the spirit and principles of the present invention shall be included within the protection scope of the present invention.

Claims (10)

  1. 式I化合物的苹果酸盐,The malate salt of a compound of formula I,
    Figure PCTCN2022124662-appb-100001
    Figure PCTCN2022124662-appb-100001
  2. 如权利要求1所述的式I化合物的苹果酸盐,其中,所述式I化合物的苹果酸盐如式Ⅱ所示,其中,x选自0.5~2。The malate of the compound of formula I according to claim 1, wherein the malate of the compound of formula I is represented by formula II, wherein x is selected from 0.5-2.
    Figure PCTCN2022124662-appb-100002
    Figure PCTCN2022124662-appb-100002
  3. 如权利要求1所述的式I化合物的苹果酸盐,其中,x为0.5、1.0、1.5、2.0;The malate of the compound of formula I as claimed in claim 1, wherein x is 0.5, 1.0, 1.5, 2.0;
    优选的,x为1.0;Preferably, x is 1.0;
    优选的,所述苹果酸盐为L-苹果酸盐。Preferably, the malate is L-malate.
  4. 如权利要求2所述的式I化合物的苹果酸盐,其中,所述式I化合物的苹果酸盐为A晶型,其是x为1.0的式Ⅱ化合物,且其中的苹果酸为L-苹果酸,所述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、14.827±0.2°、16.796±0.2°、20.012±0.2°和21.942±0.2°。The malate of the compound of formula I as claimed in claim 2, wherein the malate of the compound of formula I is crystal form A, which is the compound of formula II with x being 1.0, and wherein the malic acid is L-malate Acid, the X-ray powder diffraction pattern of the A crystal form has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 14.827±0.2°, 16.796±0.2°, 20.012±0.2° and 21.942±0.2°.
  5. 如权利要求4所述的式I化合物的苹果酸盐,其中,所述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、14.827±0.2°、16.796±0.2°、20.012±0.2°、21.942±0.2°、22.473±0.2°和25.333±0.2°。The malate salt of the compound of formula I as claimed in claim 4, wherein the X-ray powder diffraction pattern of the A crystal form has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 14.827±0.2°, 16.796± 0.2°, 20.012±0.2°, 21.942±0.2°, 22.473±0.2°, and 25.333±0.2°.
  6. 如权利要求4所述的式I化合物的苹果酸盐,其中,所述A晶型的X射线粉末衍射图 谱在下列2θ角处具有特征衍射峰:12.007±0.2°、13.288±0.2°、14.827±0.2°、16.796±0.2°、20.012±0.2°、21.942±0.2°、22.473±0.2°和25.333±0.2°;The malate salt of the compound of formula I according to claim 4, wherein the X-ray powder diffraction pattern of the crystal form A has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 13.288±0.2°, 14.827± 0.2°, 16.796±0.2°, 20.012±0.2°, 21.942±0.2°, 22.473±0.2° and 25.333±0.2°;
    优选的,所述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、13.288±0.2°、14.827±0.2°、15.399±0.2°、16.796±0.2°、20.012±0.2°、20.327±0.2°、21.942±0.2°、22.473±0.2°和25.333±0.2°。Preferably, the X-ray powder diffraction pattern of the A crystal form has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 13.288±0.2°, 14.827±0.2°, 15.399±0.2°, 16.796±0.2°, 20.012 ±0.2°, 20.327±0.2°, 21.942±0.2°, 22.473±0.2°, and 25.333±0.2°.
    优选的,所述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、13.288±0.2°、14.827±0.2°、15.399±0.2°、16.796±0.2°、20.012±0.2°、20.327±0.2°、21.942±0.2°、22.473±0.2°、24.996±0.2°、25.333±0.2°和27.718±0.2°。Preferably, the X-ray powder diffraction pattern of the A crystal form has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 13.288±0.2°, 14.827±0.2°, 15.399±0.2°, 16.796±0.2°, 20.012 ±0.2°, 20.327±0.2°, 21.942±0.2°, 22.473±0.2°, 24.996±0.2°, 25.333±0.2°, and 27.718±0.2°.
    优选的,所述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、12.755±0.2°、13.288±0.2°、13.903±0.2°、14.827±0.2°、15.399±0.2°、16.796±0.2°、17.686±0.2°、18.906±0.2°、20.012±0.2°、20.327±0.2°、21.942±0.2°、22.473±0.2°、24.996±0.2°、25.333±0.2°和27.718±0.2°。Preferably, the X-ray powder diffraction pattern of the A crystal form has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 12.755±0.2°, 13.288±0.2°, 13.903±0.2°, 14.827±0.2°, 15.399 ±0.2°, 16.796±0.2°, 17.686±0.2°, 18.906±0.2°, 20.012±0.2°, 20.327±0.2°, 21.942±0.2°, 22.473±0.2°, 24.996±0.2°, 25.333±0.2° and 27.718 ±0.2°.
    优选的,所述A晶型的X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:12.007±0.2°、12.755±0.2°、13.288±0.2°、13.903±0.2°、14.827±0.2°、15.399±0.2°、16.796±0.2°、17.686±0.2°、18.906±0.2°、20.012±0.2°、20.327±0.2°、20.660±0.2°、21.942±0.2°、22.473±0.2°、24.996±0.2°、25.333±0.2°、27.382±0.2°和27.718±0.2°。Preferably, the X-ray powder diffraction pattern of the A crystal form has characteristic diffraction peaks at the following 2θ angles: 12.007±0.2°, 12.755±0.2°, 13.288±0.2°, 13.903±0.2°, 14.827±0.2°, 15.399 ±0.2°, 16.796±0.2°, 17.686±0.2°, 18.906±0.2°, 20.012±0.2°, 20.327±0.2°, 20.660±0.2°, 21.942±0.2°, 22.473±0.2°, 24.996±0.2°, 25.333 ±0.2°, 27.382±0.2°, and 27.718±0.2°.
    优选的,所述A晶型具有基本如图1所示的XRPD图谱。Preferably, the crystal form A has an XRPD pattern substantially as shown in FIG. 1 .
    优选的,所述A晶型,其具有基本如图2所示的DSC图谱。Preferably, the crystal form A has a DSC spectrum substantially as shown in FIG. 2 .
    优选的,所述A晶型的差示扫描量热曲线在111.03±3℃处具有吸热峰。Preferably, the differential scanning calorimetry curve of the crystal form A has an endothermic peak at 111.03±3°C.
    优选的,所述A晶型,其具有基本如图3所示的TGA曲线。Preferably, the crystal form A has a TGA curve substantially as shown in FIG. 3 .
  7. 权利要求1-6任一项所述的式I化合物的苹果酸盐的制备方法,包括将式I化合物与苹果酸反应。The preparation method of the malate of the compound of formula I according to any one of claims 1-6, comprising reacting the compound of formula I with malic acid.
  8. 如权利要求7所述的制备方法,其中,式I化合物苹果酸盐的A晶型的制备方法包括如下步骤:将式I化合物、苹果酸与溶剂混合,加热,再冷却,析出晶体;其中,所述溶剂选自乙醇、异丙醇、正丙醇、丁醇、丙酮、乙酸乙酯、正庚烷、乙腈、四氢呋喃中的一种或一种以上的混合物;或者;The preparation method according to claim 7, wherein, the preparation method of the A crystal form of malate of the compound of formula I comprises the following steps: mixing the compound of formula I, malic acid and a solvent, heating, cooling, and precipitation of crystals; wherein, The solvent is selected from one or more mixtures of ethanol, isopropanol, n-propanol, butanol, acetone, ethyl acetate, n-heptane, acetonitrile, tetrahydrofuran; or;
    将式I化合物、苹果酸与第一溶剂混合,加热至完全溶解,再加入第二溶剂,冷却,析晶;其中,所述第一溶剂选自甲醇、乙醇、异丙醇、正丁醇的一种或一种以上的混合物;所述第二溶剂选自甲基叔丁基醚;酯类,如乙酸乙酯、醋酸异丙酯;乙腈;烷烃类,如正庚烷、 正己烷等的一种或一种以上的混合物。Mix the compound of formula I, malic acid and the first solvent, heat until completely dissolved, then add the second solvent, cool, and crystallize; wherein, the first solvent is selected from methanol, ethanol, isopropanol, n-butanol One or more mixtures; the second solvent is selected from methyl tert-butyl ether; esters, such as ethyl acetate, isopropyl acetate; acetonitrile; alkanes, such as n-heptane, n-hexane, etc. One or more than one mixture.
  9. 一种药物组合物,其中,所述药物组合物包含权利要求1-6任一项所述的式I化合物的苹果酸盐和任选的药学上可接受的赋形剂。A pharmaceutical composition, wherein the pharmaceutical composition comprises the malate salt of the compound of formula I according to any one of claims 1-6 and optional pharmaceutically acceptable excipients.
  10. 权利要求1-6任一项所述的式I化合物的苹果酸盐、或权利要求9所述的药物组合物在制备用于治疗中枢神经系统紊乱疾病的药物中的用途;Use of the malate salt of the compound of formula I described in any one of claims 1-6, or the pharmaceutical composition described in claim 9 in the preparation of medicines for the treatment of central nervous system disorders;
    优选的,所述中枢神经系统紊乱疾病包括但不限于精神分裂症、阿尔茨海默氏病、帕金森氏病、抑郁症、运动障碍、吸毒成瘾、疼痛和神经退行性变(例如陶氏病或突触核蛋白病)。Preferably, the disorders of the central nervous system include but are not limited to schizophrenia, Alzheimer's disease, Parkinson's disease, depression, movement disorders, drug addiction, pain and neurodegeneration (such as Dow's disease or synucleinopathies).
PCT/CN2022/124662 2021-10-14 2022-10-11 Malate salt of xanomeline derivative, crystalline form a, and preparation method therefor and use thereof WO2023061372A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111198168 2021-10-14
CN202111198168.4 2021-10-14

Publications (1)

Publication Number Publication Date
WO2023061372A1 true WO2023061372A1 (en) 2023-04-20

Family

ID=85961311

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/124662 WO2023061372A1 (en) 2021-10-14 2022-10-11 Malate salt of xanomeline derivative, crystalline form a, and preparation method therefor and use thereof

Country Status (2)

Country Link
CN (1) CN115974863A (en)
WO (1) WO2023061372A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996013168A1 (en) * 1994-10-31 1996-05-09 Eli Lilly And Company Method for treating anxiety
CN1208348A (en) * 1995-12-07 1999-02-17 伊莱利利公司 Method for treating pain
WO2020172516A1 (en) * 2019-02-22 2020-08-27 Karuna Therapeutics, Inc. Compounds and methods of deuterated xanomeline for treating neurological disorders
WO2021097427A1 (en) * 2019-11-15 2021-05-20 Karuna Therapeutics, Inc. Xanomeline derivatives and methods for treating neurological disorders

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5043345A (en) * 1989-02-22 1991-08-27 Novo Nordisk A/S Piperidine compounds and their preparation and use
AU698673B2 (en) * 1993-06-04 1998-11-05 Novo Nordisk A/S Heterocyclic chemistry
CZ285030B6 (en) * 1993-08-19 1999-05-12 Novo Nordisk A/S The use of thiadiazole and oxadiazole compounds for preparing a pharmaceutical preparation for treating schizophrenia or schizophrenic diseases
WO2012033956A1 (en) * 2010-09-08 2012-03-15 Mithridion, Inc. Cognition enhancing compounds and compositions, methods of making, and methods of treating

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996013168A1 (en) * 1994-10-31 1996-05-09 Eli Lilly And Company Method for treating anxiety
CN1208348A (en) * 1995-12-07 1999-02-17 伊莱利利公司 Method for treating pain
WO2020172516A1 (en) * 2019-02-22 2020-08-27 Karuna Therapeutics, Inc. Compounds and methods of deuterated xanomeline for treating neurological disorders
WO2021097427A1 (en) * 2019-11-15 2021-05-20 Karuna Therapeutics, Inc. Xanomeline derivatives and methods for treating neurological disorders

Also Published As

Publication number Publication date
CN115974863A (en) 2023-04-18

Similar Documents

Publication Publication Date Title
US20200039990A1 (en) Short-acting benzodiazepine salts and their polymorphic forms
EP3610875B1 (en) Opioid receptor (mor) agonist salt, fumarate salt i crystal form thereof and preparation method thereof
US8933114B2 (en) Polymorphic forms of asenapine maleate and processes for their preparation
TW202220962A (en) Crystal form of selective nav inhibitor and preparation method thereof
WO2023193563A1 (en) Crystal form a of thienopyridine compound, and preparation method therefor and pharmaceutical composition thereof
JP7152122B2 (en) edaravone salt
US8859567B2 (en) Hydrate of 1-{(2S)-2-amino-4-[2,4-bis(trifluoromethyl)-5,8-di-hydropyrido[3,4-D]pyrimidin-7(6H)-yl]-4-oxobutyl}-5,5-difluoro-piperidin-2-one tartrate
TWI802654B (en) Crystalline form of oxopicolinamide derivative and a preparation method thereof
WO2023061372A1 (en) Malate salt of xanomeline derivative, crystalline form a, and preparation method therefor and use thereof
US20220389012A1 (en) Salts of a compound and the crystalline forms thereof
WO2018149309A1 (en) Crystal form of 4-phenylthiazole derivative and preparation method thereof
EP4083040A1 (en) Irak4 inhibitor crystal and preparation method therefor
US20220281879A1 (en) Salt and crystal forms of an activin receptor-like kinase inhibitor
CN108948018B (en) Benzodiazepine derivatives, their salts and related crystalline forms, preparation and use
CN114853762A (en) Solid form of imidazotriazine compound and preparation method and application thereof
US9643950B2 (en) Solid forms of {s-3-(4-amino-1-oxo-isoindolin-2-yl)(piperidine-3,4,4,5,5-d5)-2,6-dione}
US20060194833A1 (en) Crystalline 1H-imidazo[4,5-b]pyridin-5-amine, 7-[5-[(cyclohexylmethylamino)-methyl]-1H-indol-2-yl]-2-methyl, sulfate (1:1), trihydrate and its pharmaceutical uses
US20160090385A1 (en) Crystalline forms of n,n-dicyclopropyl-4-(1,5-dimethyl-1h-pyrazol-3-ylamino)-6-ethyl-1-methyl-1,6-dihydroimidazo[4,5-d]pyrrolo[2,3-b]pyridine-7-carboximide for the treatment of myeloproliferative disorders
WO2022099442A1 (en) Crystalline forms of 5-fluoro-1-(4-fluoro-3-(4-(pyrimidin-2-yl)piperazine-1-carbonyl)benzyl)quinazoline-2,4(1h,3h)-dione and preparation therefor
CN114853750A (en) Pamoic acid salt and crystal form of pennomelin, preparation method and application thereof
CN113149998B (en) Amorphous or crystalline forms of 2-indoline spiroketones or salts, solvates thereof
WO2020073985A1 (en) Crystalline structure of mu opioid receptor (mor) agonist and preparation method therefor
TW202327593A (en) Polymorph of kras inhibitor and preparation method for polymorph and use thereof
WO2019086008A1 (en) Crystal form of benzotriazole derivative and preparation method and use thereof
JP2024505736A (en) Crystalline forms of 3-hydroxy-5-pregnan-20-one derivatives and their preparation and use

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22880309

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE