WO2022095930A1 - Atx抑制剂氘代衍生物及其应用 - Google Patents

Atx抑制剂氘代衍生物及其应用 Download PDF

Info

Publication number
WO2022095930A1
WO2022095930A1 PCT/CN2021/128735 CN2021128735W WO2022095930A1 WO 2022095930 A1 WO2022095930 A1 WO 2022095930A1 CN 2021128735 W CN2021128735 W CN 2021128735W WO 2022095930 A1 WO2022095930 A1 WO 2022095930A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
deuterium
ethyl
formula
fluorophenyl
Prior art date
Application number
PCT/CN2021/128735
Other languages
English (en)
French (fr)
Inventor
李瑶
张国彪
张晓波
唐平明
陈娅姝
余彦
张晨
严庞科
Original Assignee
四川海思科制药有限公司
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 四川海思科制药有限公司 filed Critical 四川海思科制药有限公司
Priority to JP2023526670A priority Critical patent/JP2023548181A/ja
Priority to EP21888622.4A priority patent/EP4242208A1/en
Priority to CN202180072781.3A priority patent/CN116437917A/zh
Publication of WO2022095930A1 publication Critical patent/WO2022095930A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the invention belongs to the field of medicine, and in particular relates to a deuterated derivative of an ATX inhibitor, its stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal, and its preparation and treatment related Use in medicine for disease.
  • ATX is an extracellular secretase (also known as ENPP2). Its main physiological function is to hydrolyze lysophosphatidylcholine (LPC) to generate biologically active lysophosphatidic acid (LPA) and choline. ATX hydrolyzed LPC is the main source of LPA in blood. LPA produces a series of physiological activities by acting on LPA receptors (there are at least 6 LPA 1-6 ), including cell proliferation, survival, movement and the like. The ATX-LPA signaling pathway is involved in numerous pathological processes, including angiogenesis, autoimmune diseases, inflammation, fibrosis, neurodegeneration, and pain.
  • fibrosis and tumors especially idiopathic pulmonary fibrosis (IPF).
  • IPF idiopathic pulmonary fibrosis
  • Pirfenidone pirfenidone
  • Nintedanib Nintedanib
  • the main role is to prevent the further development and deterioration of IPF. It has obvious curative effect, so people have been working to find more effective drugs for IPF treatment.
  • the clinical progress of ATX inhibitor drug GLPG-1690 is relatively advanced (clinical phase III), and it is used for the treatment of IPF, and its phase II clinical has shown good curative effect.
  • the purpose of the present invention is to provide a deuterated ATX inhibitor, which has the advantages of novel structure, good drug effect, high bioavailability, small side effects, fast onset of action and long-term effect.
  • the present invention relates to a compound represented by formula (I), its stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal,
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 are each independently selected from hydrogen or deuterium;
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 at least one group is selected from deuterium
  • the compound of formula (I) is not the following structure:
  • R 22 is selected from H
  • R 1 is selected from deuterium
  • R 2 , R 3 are selected from deuterium
  • R 6 , R 7 are selected from deuterium
  • R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 are selected from deuterium;
  • R 16 is selected from deuterium
  • R 17 is selected from deuterium
  • R 18 , R 19 , R 20 , R 21 are selected from deuterium;
  • R 23 , R 24 are selected from deuterium
  • At least one of R 25 , R 26 , R 27 is selected from deuterium
  • R 25 , R 26 , R 27 are selected from deuterium
  • R 28 , R 29 , R 30 are selected from deuterium
  • R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 are selected from deuterium;
  • R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 25 , R 26 , R 27 are selected from deuterium;
  • R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 is selected from deuterium;
  • R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 are selected from deuterium;
  • R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 18 , R 19 , R 20 , R 21 are selected from deuterium;
  • R 23 , R 24 , R 28 , R 29 , R 30 , R 18 , R 19 , R 20 , R 21 are selected from deuterium;
  • R 25 , R 26 , R 27 , R 18 , R 19 , R 20 , R 21 are selected from deuterium;
  • R 1 , R 25 , R 26 , R 27 are selected from deuterium
  • R 1 , R 23 , R 24 , R 28 , R 29 , R 30 are selected from deuterium;
  • R 6 , R 7 , R 25 , R 26 , R 27 are selected from deuterium;
  • R 6 , R 7 , R 23 , R 24 , R 28 , R 29 , R 30 are selected from deuterium;
  • R 2 , R 3 , R 25 , R 26 , R 27 are selected from deuterium;
  • R 2 , R 3 , R 23 , R 24 , R 28 , R 29 , R 30 are selected from deuterium.
  • the present invention further provides a more specific crystal form I of the compound of formula (A), the crystal form has high purity, low hygroscopicity, good solubility, high temperature resistance, high humidity and strong light, high stability, and suitable particle size, It is easy to filter and has good fluidity, and is suitable for the preparation of pharmaceutical dosage forms.
  • the crystalline form I of the compound of formula (A) also has characteristic diffraction peaks at the following 2 ⁇ positions: 17.29 ⁇ 0.2°, 18.26 ⁇ 0.2°, 20.58 ⁇ 0.2°.
  • the crystalline form I of the compound of formula (A) also has characteristic diffraction peaks at the following 2 ⁇ positions: 6.92 ⁇ 0.2°, 7.23 ⁇ 0.2°, 13.46 ⁇ 0.2°, 19.47 ⁇ 0.2°, 24.85 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the crystalline form I of the compound of formula (A) is basically as shown in FIG. 1 .
  • the TGA curve of the crystal form I of the compound of formula (A) is basically as shown in FIG. 2
  • the DSC chart thereof is basically as shown in FIG. 3 .
  • the crystal form I of the compound of formula (A) of the present invention has a crystal particle size of less than 100 ⁇ m; in some embodiments, the crystal particle size is less than 90 ⁇ m; in some embodiments, the crystal particle size is less than 80 ⁇ m; in some embodiments , the crystal particle size is less than 70 ⁇ m; in some embodiments, the crystal particle size is less than 60 ⁇ m; in some embodiments, the crystal particle size is less than 50 ⁇ m.
  • the present invention also provides a method for preparing the crystal form I of the compound of formula (A), the method comprising: adding the crude product of compound (A) to an alcohol solvent, heating up and stirring, dissolving and cooling to room temperature, crystallization, filtering , that is.
  • the alcoholic solvent is selected from methanol, ethanol, propanol, isopropanol or a combination thereof.
  • the ratio of compound (A) to alcohol solvent is 1:5-8 g/mL. In some embodiments, the ratio of compound (A) to alcoholic solvent is 1:5-6 g/mL.
  • the temperature in the above preparation method, is raised to 60-90°C. In some embodiments, the temperature is raised to 70-80°C. In some embodiments, the temperature is raised to 75°C.
  • Form I of Compound (A) of the present invention is present at about 5% to about 100% by weight of the drug substance; in certain embodiments, it is present at about 10% to about 100% by weight of the drug substance; in In certain embodiments, from about 15% to about 100% by weight of the drug substance; in certain embodiments, from about 20% to about 100% by weight of the drug substance; in certain embodiments, Present at about 25% to about 100% by weight of the drug substance; in certain embodiments, at about 30% to about 100% by weight of the drug substance; in certain embodiments, at about 35% by weight of the drug substance Present at % by weight to about 100% by weight; in certain embodiments, present at about 40% by weight to about 100% by weight of the drug substance; in certain embodiments, at about 45% by weight to about 100% by weight of the drug substance % present; in certain embodiments, present at about 50% to about 100% by weight of the drug substance; in certain embodiments, present at about 55% to about 100% by weight of the drug substance; in certain In embodiments, from about 60% to about 100% by weight of the drug substance; in
  • the present invention further provides compounds of formula (II-A), formula (II-B) and formula (II-C) as shown below:
  • the present invention further provides a method for preparing the compound of formula (A), comprising the following steps
  • the compound of formula (II-C) is dissolved in an organic solvent A, and is preferably mixed with 2-chloro-1-(3hydroxyazetidine-1-yl)ethanone, a base in React at 72-88°C for 3-10 hours; more preferably at 75-85°C for 4-8 hours; more preferably at 80°C for 6 hours.
  • the organic solvent A is selected from at least one of acetonitrile, 2-methyltetrahydrofuran, ethylbenzene, methyl ethyl formate, and diethylene glycol tert-butyl ether.
  • the organic solvent A is selected from acetonitrile.
  • the alkali is at least one of potassium carbonate and sodium carbonate.
  • the base is potassium carbonate.
  • preparation method of the compound of formula (A) also comprises the following steps
  • preparation method of the compound of formula (A) further comprises the following steps
  • preparation method of the compound of formula (A) further comprises the following steps
  • the preparation of the compound of formula (A) of the present invention also comprises the following steps,
  • step i The compound (II-A) obtained in step i is dissolved in the organic solvent B, cooled to 0° C. under nitrogen protection, a reducing agent is added, and methyl iodide-d 3 is added after the reaction is completed to obtain compound (II-B );
  • step iii Dissolving the compound (II-B) obtained in step ii in the organic solvent C, adding trifluoroacetic acid, and reacting to obtain the compound of formula (II-C).
  • the preparation method of the compound of formula (A) of the present invention comprises the following steps,
  • the aprotic polar solvent is at least one of N,N-dimethylacetamide, dimethylformamide, hexamethylphosphoramide, acetonitrile, acetone, and dimethyl sulfoxide .
  • aprotic polar solvent is N,N-dimethylacetamide.
  • the organic solvent B is at least one of tetrahydrofuran, N,N-dimethylacetamide, dimethylformamide, acetonitrile, and acetone.
  • organic solvent B is tetrahydrofuran.
  • the reducing agent is sodium hydride.
  • the organic solvent C is at least one of methylene chloride, methyl acetate, dimethyl carbonate, propylene glycol methyl ether acetate, and dimethyl nylon acid.
  • organic solvent C is dichloromethane.
  • the present invention provides a pharmaceutical composition, which contains the compound described in any one of the foregoing technical solutions, its stereoisomers, solvates, prodrugs, metabolites, pharmaceutically acceptable compounds The accepted salt or co-crystal, or Form I of the compound of formula (A), and a pharmaceutically acceptable carrier and/or excipient.
  • the present invention provides the compound described in any one of the foregoing technical solutions, its stereoisomer, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal, or Use of the aforementioned composition in the preparation of a medicament for treating ATX-mediated diseases.
  • the ATX-mediated disease is idiopathic pulmonary fibrosis.
  • thermogravimetric analysis TGA
  • DSC differential scanning calorimetry
  • the melting peak heights of TGA and DSC curves depend on many factors related to sample preparation and instrument geometry , while the peak positions are relatively insensitive to experimental details.
  • the crystalline compounds of the present invention have TGA and DSC patterns of characteristic peak positions, having substantially the same properties as the TGA and DSC patterns provided in the accompanying drawings of the present invention, with a measurement error tolerance of Within ⁇ 5°C, generally within ⁇ 3°C.
  • the crystal form of the present invention is not limited to the characteristic pattern that is exactly the same as the characteristic pattern described in the accompanying drawings disclosed in the present invention, such as XRD, DSC, TGA, DVS, and isothermal adsorption curves, which have the same characteristics as those described in the accompanying drawings. Any crystalline form of which patterns are substantially the same or substantially the same characteristic pattern falls within the scope of the present invention.
  • a “therapeutically effective amount” refers to the amount of a compound that elicits a physiological or medical response in a tissue, system, or subject, which amount is sought, including one or a The amount of compound at which several symptoms occur or are alleviated to some extent.
  • “Pharmaceutically acceptable salt” means that a compound of the present invention retains the biological availability and properties of a free acid or free base that is treated with a non-toxic inorganic or organic base, and the free base is treated with Salts obtained by the reaction of non-toxic inorganic or organic acids.
  • “Pharmaceutical composition” means a mixture of one or more of the compounds of the present invention or stereoisomers, solvates, pharmaceutically acceptable salts, co-crystals, deuterated compounds, and other components, wherein the other components Physiologically/pharmaceutically acceptable carriers and/excipients are included.
  • Carrier means one that does not significantly irritate the organism and does not eliminate the biological activity and properties of the administered compound, and can change the way the drug enters the human body and its distribution in the body, controls the release rate of the drug and releases the drug
  • Non-limiting examples of systems for delivery to targeted organs include microcapsules and microspheres, nanoparticles, liposomes, and the like.
  • Excipient means: not itself a therapeutic agent, but used as a diluent, adjuvant, binder and/or vehicle for addition to a pharmaceutical composition to improve its handling or storage properties or to allow or facilitate The compound or pharmaceutical composition is formed in unit dosage form for administration.
  • pharmaceutical excipients can serve various functions and can be described as wetting agents, buffers, suspending agents, lubricants, emulsifying agents, disintegrating agents, absorbing agents, preservatives , surfactants, colorants, flavors and sweeteners.
  • Examples of pharmaceutical excipients include, but are not limited to: (1) sugars such as lactose, glucose and sucrose; (2) starches such as corn starch and potato starch; (3) cellulose and derivatives thereof such as carboxymethyl Sodium cellulose, ethyl cellulose, cellulose acetate, hydroxypropyl methylcellulose, hydroxypropyl cellulose, microcrystalline cellulose and croscarmellose (e.g.
  • croscarmellose sodium (4) tragacanth powder; (5) malt; (6) gelatin; (7) talc; (8) excipients such as cocoa butter and suppository wax; (9) oils such as peanut oil, cottonseed oil, red Flower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols such as propylene glycol; (11) polyols such as glycerol, sorbitol, mannitol and polyethylene glycol; (12) esters such as oils (13) agar; (14) buffers such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; ( 18) Ringer's solution; (19) Ethanol; (20) pH buffer solution; (21) Polyester, polycarbonate and/or polyanhydride; Compatible substances.
  • oils such as peanut oil, cottonseed oil, red Flower oil, sesame oil, olive oil, corn oil and soybean oil
  • Steps refer to isomers resulting from different arrangements of atoms in a molecule in space, including cis-trans isomers, enantiomers and conformational isomers.
  • Solvate refers to a substance formed by a compound of the present invention or a salt thereof and a stoichiometric or non-stoichiometric solvent bound non-covalently by intermolecular forces.
  • the solvent is water, it is a hydrate.
  • Co-crystal refers to a crystal formed by the combination of an active pharmaceutical ingredient (API) and a co-crystal former (CCF) under the action of hydrogen bonds or other non-covalent bonds, wherein the pure states of API and CCF are both at room temperature solid, and there is a fixed stoichiometric ratio between the components.
  • a co-crystal is a multicomponent crystal that includes both binary co-crystals formed between two neutral solids and multi-component co-crystals formed between neutral solids and salts or solvates.
  • Fig. 1 is the X-ray powder diffraction pattern of compound A crystal form I using Cu-K ⁇ radiation;
  • Fig. 2 is the TGA curve diagram of compound A crystal form I
  • Fig. 3 is the DSC curve diagram of compound A crystal form I
  • Fig. 4 is the PLM image of compound A crystal form I
  • Figure 5 is a DVS graph of Compound A Form I.
  • the raw materials were purchased from companies such as Titan Technology, Annagy Chemical, Shanghai Demo, Chengdu Kelon Chemical, Shaoyuan Chemical Technology, Nanjing Yaoshi, WuXi AppTec and Bailingwei Technology.
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • HPLC HPLC-based high pressure liquid chromatograph
  • the thin layer chromatography silica gel plate uses Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate, the size of the silica gel plate used for thin layer chromatography (TLC) is 0.15mm-0.20mm, and the specification used for TLC separation and purification products is 0.4mm -0.5mm;
  • the XRPD patterns were acquired on an X-ray powder diffraction analyzer manufactured by PANalytacal, and the scan parameters are shown in the table below.
  • TGA Thermogravimetric Analysis
  • DSC Differential Scanning Calorimetry
  • TGA and DSC graphs were collected on a TA Q5000/5500 Thermogravimetric Analyzer and a TA 2500 Differential Scanning Calorimeter, respectively, and the test parameters are listed in the table below.
  • Dynamic moisture sorption (DVS) curves were collected on the DVS Intrinsic of SMS (Surface Measurement Systems). The relative humidity at 25°C was corrected for the deliquescence points of LiCl, Mg( NO3 ) 2 and KCl. DVS test parameters are listed in the table below.
  • Polarized light microscopy data were collected at room temperature with an Axio Lab.A1 upright microscope.
  • NMP N-methylpyrrolidone
  • HATU 2-(7-azabenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate
  • PE petroleum ether
  • Acetone acetone
  • ACN acetonitrile
  • Trimethylsilylacetylene (1a) (10.07 g, 102.5 mmol) was dissolved in dry tetrahydrofuran (45 mL) at room temperature, the system was cooled to -78 °C and n-butyllithium (45 mL, 2M n-hexane) was added dropwise to it. solution), transfer to an ice-water bath and continue to react for 20 minutes after the dropwise addition is complete. After cooling to -78 ° C, hexamethylphosphoric acid triamine (18.37 g, 102.5 mmol) was added dropwise, and stirring was continued for 30 minutes. After the addition was completed, the reaction was naturally raised to room temperature overnight.
  • Acetic acid-d4(3a) (1.0 g, 15.6 mmol) was dissolved in trifluoroacetic anhydride (6 mL), and after stirring uniformly, 4-dimethylaminopyridine (30 mg, 0.25 mmol) was added, and the system was heated to 60 °C, Bromine (0.98 mL) was slowly added dropwise under this strip for 1 hour. After the dropwise addition was completed, the reaction was continued for 1 hour under these conditions, then cooled to room temperature, and nitrogen gas was introduced into the system until the volume of the system was reduced to about 1 mL. Ethanol (20 mL) was added, reacted at 60°C overnight, and cooled to room temperature.
  • Methyl 4-fluorobenzoate-2,3,5,6-d4(4b) (0.96g, 6.1mmol) was dissolved in toluene (8mL), cooled to -10°C under nitrogen protection, and acetonitrile (1.46g) was added. , 36.3 mmol), sodium bis(trimethylsilyl)amide (6 mL, 12 mmol, 2M in THF) was slowly added dropwise to the system. After the addition was complete, hydrochloric acid (20 mL, 1N aqueous solution) was quenched.
  • Cupric chloride (0.60 g, 4.5 mmol) was dissolved in acetonitrile (8 mL), tert-butyl nitrite (0.67 g, 5.6 mmol) was added dropwise to the system, and the mixture was stirred at room temperature for 30 minutes.
  • To the system was added 2-amino-4-(4-fluorophenyl-2,3,5,6-d4)thiazole-5-carbonitrile (4d) (0.82 g, 3.67 mmol) in portions. After the addition was complete, the reaction was continued for 1 hour at room temperature. Hydrochloric acid (10 mL, 1 N aqueous solution) was quenched.
  • reaction solution added water (150 mL), the aqueous phase was extracted with ethyl acetate (200 mL ⁇ 2), the combined organic phases were washed with saturated sodium chloride (100 mL), dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the residue was subjected to silica gel column chromatography Isolation and purification to obtain the target compound 4-(2-(ethyl-d5)-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylic acid tert-butyl ester (1F), white Solid (7.53 g, 71% yield).
  • the reaction solution was added with water (200 mL), the aqueous phase was extracted with ethyl acetate (200 mL ⁇ 2), the combined organic phases were washed with saturated sodium chloride (100 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and separated by silica gel column chromatography to obtain The target compound, tert-butyl 4-(2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate (3B), white solid (7.8 g, yield 68%).
  • reaction solution was added with water (150 mL), the aqueous phase was extracted with ethyl acetate (200 mL ⁇ 2), the combined organic phases were washed with saturated sodium chloride (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated to obtain 4D crude product (2.0 g) .
  • the crude 4D product (2.0 g, about 5.6 mmol) from the previous step was dissolved in methanol (10.0 mL) and acetic acid (2.0 mL), cooled to 5-10 °C, and then an aqueous solution of sodium nitrite (0.77 g, 11.2 mmol) ( 2.0 mL) was added dropwise to the reaction solution, the addition was completed, and the reaction was carried out at room temperature for 16 hours. Water (20 mL) was added dropwise to the reaction, the addition was completed, filtered, the filter cake was washed with water (5 mL ⁇ 2), and the crude product of 4E (2.00 g) was obtained by suction.
  • Compound 4H can also be synthesized by the following methods:
  • the eighth step 2-((2-ethyl-6-fluoro-5-(piperazin-1-yl-2,2,3,3,5,5,6,6-d8)pyrazole[1, 5-a]Pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile (4I)
  • Trifluoroacetic acid (5.0 mL) was added dropwise to the solution of compound 4H (1.20 g crude product) obtained in the previous step in dichloromethane (15.0 mL) under nitrogen protection at 0°C, then the temperature was naturally raised to room temperature, and stirring was continued for 2 h.
  • the trifluoroacetate salt of compound 9C (0.58 g) was dissolved in acetonitrile (15 mL), and potassium carbonate (0.41 g, 3 mmol), 2-chloro-1-(3hydroxyazetidine-1) were added to the system in sequence -yl) ethyl ketone (1M) (0.22 g, 1.5 mmol), and the temperature was raised to 80° C. to react for 3 hours.
  • the dynamic solubility of Compound A Form I (Example 11) in water and three biological vehicles was evaluated.
  • the dynamic solubility (1, 2, 4 and 24 hours) of the samples in four solvent systems of water, SGF, FaSSIF and FeSSIF was determined by rotary mixing (25 rpm) at a feed concentration of 5-10 mg/mL at 37 °C.
  • the samples at each time point were centrifuged (12000 rpm, 5 min) and filtered (0.45 ⁇ m PTFE filter), and the HPLC concentration and pH of the filtrate were determined.
  • the solubility test results are summarized in Table 2.
  • the starting pH of H2O , SGF, FaSSIF and FeSSIF were 6.9, 1.8, 6.5 and 5.0, respectively.
  • simulated fasting state intestinal fluid FaSSIF: Weigh 0.17 g of anhydrous NaH 2 PO 4 , 0.021 g of NaOH and 0.31 g of NaCl into a 50 mL volumetric flask. About 48 mL of purified water-soluble clear was added, and the pH was adjusted to 6.5 with 1 M hydrochloric acid or 1 M NaOH solution. Add purified water to volume, and weigh in 0.11 g of SIF powder to dissolve.
  • simulated feeding state intestinal fluid Take 0.41 mL of glacial acetic acid, 0.20 g of NaOH and 0.59 g of NaCl into a 50 mL volumetric flask. About 48 mL of purified water-soluble clear was added, and the pH was adjusted to 5.0 with 1 M hydrochloric acid or 1 M NaOH solution. Add purified water to volume, and weigh in 0.56g of SIF powder to dissolve.
  • Autotaxin is a plasma phosphodiesterase that converts lysophosphatidylcholine (LPC) to lysophosphatidic acid (LPA), so LPA formation was used to evaluate the efficacy of autotaxin inhibitors. The efficacy of the compounds was evaluated in pooled isolated human plasma.
  • Plasma was collected after centrifugation of heparin-anticoagulated whole blood. 5- ⁇ L of the compound to be tested or DMSO was added to 95- ⁇ L plasma, incubated at 37° C. for 2 h, and then a stop solution (40 mM disodium hydrogen phosphate buffer containing 30 mM citric acid, pH 4) was added. LPA in plasma was detected by LC-MSMS before and after incubation. To determine LPA 18:2 or 20:4 concentrations in study plasma, calibration standards for LPA 18:2 or 20:4 were prepared by serial dilution in butanol: 20000, 10000, 5000, 2000, 1000, 500, 200 , 100, 50, 20 and 10 ng/mL.
  • Inhibition rate % (average relative concentration of control wells-average relative concentration of test wells)/average relative concentration of control wells ⁇ 100%, the curve is drawn as inhibition rate (y-axis) and compound concentration (x-axis), and passed through GraphPad Prism 7.0 was fitted with log (inhibitor) and normalized responses (variable slope).
  • Example LPA formation (18:2) LPA formation (20:4) 1 46.05 48.51 2 51.96 55.27 3 42.66 44.47 4 51.79 54.81
  • test substance was administered to SD rats through a single dose of intravenous and intragastric administration, the concentration of the test substance in the rat plasma was determined, and the pharmacokinetic characteristics and bioavailability of the test substance in rats were evaluated.
  • mice male SD rats, about 220 g, 6-8 weeks old, 6 rats/compound. Purchased from Chengdu Dashuo Laboratory Animal Co., Ltd.
  • Sample collection time points of group G2 0, 15, 30 min, 1, 2, 4, 6, 8, 24 h.
  • the compounds of the present invention especially compounds 2, 9, and 10, have good pharmacokinetics and high bioavailability in rats, and compounds 5, 6, and 7 have fast onset of action.
  • Test animals C57 mice, ⁇ 25 g, male, 6-8 weeks old, 36 mice, purchased from Chengdu Dashuo Laboratory Animal Co., Ltd., production license number: SCXK (Chuan) 2020-030.
  • Test subjects compound I-1 (patent WO2019228403A1 compound 101) and compound 3.
  • Vehicle for intravenous administration 5% DMA+5% Solutol+90% Saline; vehicle for intragastric administration: 0.5% MC
  • the compounds of the present invention especially compound 3, have good pharmacokinetics and high bioavailability in mice.
  • Test animals Beagle dogs, 8-10kg, male, 0.5-1.5 years old, 12, purchased from Beijing Masi Biotechnology Co., Ltd., production license number: SCXK (Beijing) 2016-001.
  • Test subjects compound I-1 (patent WO2019228403A1 compound 101) and compound 3.
  • Vehicle for intravenous administration 5% DMSO + 5% Solutol + 90% Saline;
  • Oral administration vehicle 1% DMSO + 1% Solutol + 98% (0.5% MC);
  • the compounds of the present invention especially compound 3, have good pharmacokinetics and high bioavailability in dogs.
  • Test animals cynomolgus monkeys, 2.4-5.9kg, male, 3-5.5 years old, 12, purchased from Suzhou Xishan Zhongke Laboratory Animal Co., Ltd., production license number: SCXK (Su) 2018-0001.
  • Test subjects compound I-1 (patent WO2019228403A1 compound 101) and compound 3.
  • Vehicle for intravenous administration 5% DMSO + 5% Solutol + 90% Saline;
  • Oral administration vehicle 1% DMSO + 1% Solutol + 98% (0.5% CMC-Na)
  • the compounds of the present invention especially compound 3, have good pharmacokinetics and high bioavailability in monkeys.
  • Test compounds compound I-1 (patent WO2019228403A1 compound 101) and example compound 3.
  • HEK293 cell line stably expressing hERG ion channel was purchased from Invitrogen Company. The cell line was cultured in a solution containing 85% DMEM, 10% dialyzed fetal bovine serum, 0.1 mM MEM non-essential amino acid solution, 100 U/mL penicillin-streptomycin solution, 25 mM HEPES, 5 ⁇ g/mL blasticidin and 400 ⁇ g/mL blasticidin. mL of Geneticin (G418). When the cell density increased to 40% to 80% of the bottom area of the culture dish, digested by trypsin, and passaged three times a week.
  • the cells were cultured in a 6 cm dish at a density of 5 ⁇ 10 5 , and induced with 1 ⁇ g/mL doxycycline for 48 hours, then the cells were digested and seeded on glass slides for subsequent manual patch clamp experiments. .
  • Extracellular fluid (in mM): 132 sodium chloride, 4 potassium chloride, 3 calcium chloride, 0.5 magnesium chloride, 11.1 glucose, 10 HEPES (pH adjusted to 7.35 with sodium hydroxide).
  • Test compounds were first dissolved in DMSO and formulated to a final concentration of 30 mM stock solutions. The original stock solution was then diluted with DMSO at a certain ratio into the other 4 gradient series solutions at the concentrations: 10, 3.33, 1.11 and 0.37 mM, respectively. Before the start of the experiment, the gradient series solutions of the compounds to be tested were diluted again at a ratio of 1:1000 into a series of working solutions of gradient concentrations with extracellular fluid. Five different gradient concentrations of working solutions were used to determine the potential inhibition of the hERG potassium channel by compounds and to fit dose-response curves and calculate IC50s .
  • the hERG current test method is as follows: depolarize the membrane potential from -80mV to +30mV by applying a depolarization command voltage for 4.8 seconds, and then momentarily apply a repolarization voltage for 5.2 seconds to reduce the membrane potential to -50mV to remove channel failure.
  • the hERG tail current was observed.
  • the peak value of the tail current is the magnitude of the hERG current.
  • the hERG currents used to detect the test compounds were recorded continuously for 120 seconds before administration to evaluate the stability of the hERG currents produced by the test cells. Only stable cells within the acceptance range of the evaluation criteria can be used for subsequent compound testing.
  • Test of the inhibitory effect of the test compound on hERG current first, the hERG current measured in the extracellular fluid containing 0.1% DMSO was used as the detection baseline. After the hERG current remained stable for at least 5 minutes, the solution containing the test compound was perfused from low to high concentration around the cells. Wait approximately 5 minutes after the end of each perfusion to allow the compound to fully act on the cells and to simultaneously record hERG currents. After the recorded current became stable, the last 5 hERG current values were recorded, and the average value was taken as its final current value at a specific concentration. After testing the compounds, 150 nM of dofetilide was added to the same cell to completely inhibit its current as a positive control for that cell. At the same time, the positive compound Dofelide was simultaneously detected with the same patch clamp system before and after the end of the test drug experiment to ensure the reliability and sensitivity of the entire detection system.
  • the current inhibition percentage was calculated by the following formula, the data was output by PatchMaster or Clampex 10.2 software, the dose-response curve was fitted by Graphpad Prism 8.0 software, and the IC 50 value was calculated.
  • the experimental results are shown in Table 11.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Cardiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

公开一种式(I)的化合物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,或含它们的药物组合物,及其作为ATX抑制剂在制备治疗相关疾病的药物中的用途,式(I)中各基团如说明书之定义。

Description

ATX抑制剂氘代衍生物及其应用 技术领域
本发明属于药物领域,尤其涉及一种ATX抑制剂的氘代衍生物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,及其在制备治疗相关疾病的药物中的用途。
背景技术
Autotaxin(ATX)是一种胞外分泌酶(也被称为ENPP2)。其主要的生理功能是水解溶血磷脂酰胆碱(LPC)生成具有生物活性的溶血磷脂酸(LPA)和胆碱(choline)。ATX水解LPC是血中LPA的最主要来源。LPA通过作用于LPA受体(至少有6种LPA 1-6)而产生具有一系列的生理活性,包括细胞增殖、存活、运动等。在众多病理过程中都存在ATX-LPA信号通路的参与,包括血管生成、自体免疫性疾病、炎症、纤维化、神经退行性病变和疼痛。其中研究最为广泛的是纤维化和肿瘤,特别是特发性肺纤维化(idiopathic pulmonary fibrosis,IPF)。2014年FDA批准了Pirfenidone(吡非尼酮)和三重血管激酶抑制剂Nintedanib(尼达尼布)两种新药用于IPF的治疗,主要作用是防止IPF的进一步发展和恶化,对治疗IPF本身没有明显疗效,所以人们一直致力于寻找IPF治疗更加有效的药物。ATX抑制剂药物GLPG-1690临床进展比较靠前(临床三期),用于IPF的治疗,其二期临床已显示出良好的疗效。
发明内容
本发明的目的是提供一种氘代的ATX抑制剂,具有结构新颖、药效好、生物利用度高、副作用小、起效快、长效的优点。
本发明涉及一种式(I)所示的化合物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
Figure PCTCN2021128735-appb-000001
其中,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 16、R 17、R 18、R 19、R 20、R 21、R 22、R 23、R 24、R 25、R 26、R 27、R 28、R 29、R 30各自独立地选自氢或氘;
条件是,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 16、R 17、R 18、R 19、R 20、R 21、R 22、R 23、R 24、R 25、R 26、R 27、R 28、R 29、R 30中至少一个基团选自氘且式(I)化合物不为如下结构:
Figure PCTCN2021128735-appb-000002
在某些实施方案中,R 22选自H;
在某些实施方案中,R 1选自氘;
在某些实施方案中,R 2、R 3选自氘;
在某些实施方案中,R 6、R 7选自氘;
在某些实施方案中,R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15选自氘;
在某些实施方案中,R 16选自氘;
在某些实施方案中,R 17选自氘;
在某些实施方案中,R 18、R 19、R 20、R 21选自氘;
在某些实施方案中,R 23、R 24选自氘;
在某些实施方案中,R 25、R 26、R 27中的至少一个选自氘;
在某些实施方案中,R 25、R 26、R 27选自氘;
在某些实施方案中,R 28、R 29、R 30选自氘;
在某些实施方案中,R 23、R 24、R 25、R 26、R 27、R 28、R 29、R 30选自氘;
在某些实施方案中,R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 25、R 26、R 27选自氘;
在某些实施方案中,R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 23、R 24、R 25、R 26、R 27、R 28、R 29、R 30选自氘;
在某些实施方案中,R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 16、R 17选自氘;
在某些实施方案中,R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 18、R 19、R 20、R 21选自氘;
在某些实施方案中,R 23、R 24、R 28、R 29、R 30、R 18、R 19、R 20、R 21选自氘;
在某些实施方案中,R 25、R 26、R 27、R 18、R 19、R 20、R 21选自氘;
在某些实施方案中,R 1、R 25、R 26、R 27选自氘;
在某些实施方案中,R 1、R 23、R 24、R 28、R 29、R 30选自氘;
在某些实施方案中,R 6、R 7、R 25、R 26、R 27选自氘;
在某些实施方案中,R 6、R 7、R 23、R 24、R 28、R 29、R 30选自氘;
在某些实施方案中,R 2、R 3、R 25、R 26、R 27选自氘;
在某些实施方案中,R 2、R 3、R 23、R 24、R 28、R 29、R 30选自氘。
本发明所述的式(I)的化合物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中所述化合物选自以下结构之一:
Figure PCTCN2021128735-appb-000003
Figure PCTCN2021128735-appb-000004
本发明还进一步提供了更具体的式(A)化合物的晶型I,该晶型纯度高,吸湿性低,溶解性好,能耐高温、高湿及强光照,稳定性高,粒径适宜,易过滤、流动性好,适于药物剂型的制备。
Figure PCTCN2021128735-appb-000005
式(A)化合物的晶型I,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:9.35±0.2°、10.32±0.2°、12.08±0.2°、15.10±0.2°。
进一步地,式(A)化合物的晶型I,其X-射线粉末衍射图谱还在以下2θ位置具有特征衍射峰:17.29±0.2°、18.26±0.2°、20.58±0.2°。
进一步地,式(A)化合物的晶型I,其X-射线粉末衍射图谱还在以下2θ位置具有特征衍射峰:6.92±0.2°、7.23±0.2°、13.46±0.2°、19.47±0.2°、24.85±0.2°。
进一步地,式(A)化合物的晶型I,其X-射线粉末衍射图谱基本如图1所示。
进一步地,式(A)化合物的晶型I,其X-射线粉末衍射峰基本如表1所示。
进一步地,式(A)化合物的晶型I,其TGA曲线基本如图2所示,其DSC图基本如图3所示。
进一步地,本发明式(A)化合物的晶型I,其晶体粒径小于100μm;在一些实施例中,晶体粒径小于90μm;在一些实施例中,晶体粒径小于80μm;在一些实施例中,晶体粒径小于70μm;在一些实施例中,晶体粒径小于60μm;在一些实施例中,晶体粒径小于50μm。
本发明还提供了式(A)化合物的晶型I的制备方法,所述方法包括:将化合物(A)粗品加到醇类溶剂中,升温并搅拌,溶解后冷却至室温,析晶,过滤,即得。
在一些实施方案中,上述制备方法,所述醇类溶剂选自甲醇、乙醇、丙醇、异丙醇或其组合。
在一些实施方案中,上述制备方法,化合物(A)与醇类溶剂的比例为1:5-8g/mL。在一些实施例中,化合物(A)与醇类溶剂的比例为1:5-6g/mL。
在一些实施方案中,上述制备方法,所述升温是升温至60-90℃。在一些实施例中,升温至70-80℃。在一些实施例中,升温至75℃。
本发明的化合物(A)的晶型I,以原料药的约5重量%至约100重量%存在;在某些实施方案中,以原料药的约10重量%至约100重量%存在;在某些实施方案中,以原料药的约15重量%至约100重量%存在;在某些实施方案中,以原料药的约20重量%至约100重量%存在;在某些实施方案中,以原料药的约25重量%至约100重量%存在;在某些实施方案中,以原料药的约30重量%至约100重量%存在;在某些实施方案中,以原料药的约35重量%至约100重量%存在;在某些实施方案中,以原料药的约40重量%至约100 重量%存在;在某些实施方案中,以原料药的约45重量%至约100重量%存在;在某些实施方案中,以原料药的约50重量%至约100重量%存在;在某些实施方案中,以原料药的约55重量%至约100重量%存在;在某些实施方案中,以原料药的约60重量%至约100重量%存在;在某些实施方案中,以原料药的约65重量%至约100重量%存在;在某些实施方案中,以原料药的约70重量%至约100重量%存在;在某些实施方案中,以原料药的约75重量%至约100重量%存在;在某些实施方案中,以原料药的约80重量%至约100重量%存在;在某些实施方案中,以原料药的约85重量%至约100重量%存在;在某些实施方案中,以原料药的约90重量%至约100重量%存在;在某些实施方案中,以原料药的约95重量%至约100重量%存在;在某些实施方案中,以原料药的约98重量%至约100重量%存在;在某些实施方案中,以原料药的约99重量%至约100重量%存在;在某些实施方案中,基本上所有的原料药都是本发明化合物(A)的晶型I,即原料药基本上是相纯晶体。
本发明还进一步提供了如下所示式(II-A)、式(II-B)、式(II-C)化合物:
Figure PCTCN2021128735-appb-000006
(II-A)、式(II-B)、式(II-C)化合物作为化合物(A)的中间体。
本发明还进一步提供了一种制备式(A)化合物的方法,包含如下步骤
Figure PCTCN2021128735-appb-000007
将式(II-C)化合物溶于有机溶剂A中,与2-氯-1-(3羟基氮杂环丁烷-1-基)乙酮、碱,在70~90℃下反应,得到式(A)化合物。
在一些实施方案中,上述制备方法,式(II-C)化合物溶于有机溶剂A中,与2-氯-1-(3羟基氮杂环丁烷-1-基)乙酮、碱优选在72~88℃下反应3~10小时;进一步优选在75~85℃下 反应4~8h;更优选在80℃下反应6小时。
在一些实施方案中,上述制备方法,所述有机溶剂A选自乙腈、2-甲基四氢呋喃、乙苯、甲酰甲酸甲乙酯、二乙二醇叔丁醚中的至少一种。
在一些实施方案中,上述制备方法,所述有机溶剂A选自乙腈。
在一些实施方案中,上述制备方法,所述碱为碳酸钾、碳酸钠中的至少一种。
在一些实施方案中,上述制备方法,所述碱为碳酸钾。
进一步的,式(A)化合物的制备方法,还包含如下步骤
Figure PCTCN2021128735-appb-000008
化合物(II-B)溶于有机溶剂C中,加入三氟乙酸,反应得到式(II-C)化合物。
进一步的,式(A)化合物的制备方法,还进一步包含如下步骤
Figure PCTCN2021128735-appb-000009
将化合物(II-A),溶于有机溶剂B中,在氮气保护下冷却至0℃,加入还原剂,反应完成后加入碘甲烷-d 3,得到化合物(II-B)。
进一步的,式(A)化合物的制备方法,还进一步包含如下步骤
Figure PCTCN2021128735-appb-000010
式(II-D)化合物、2-氯-4-(4-氟苯基)噻唑-5-氰基、2,6-二甲基吡啶和非质子极性溶剂,在60~80℃下反应,得到化合物(II-A)。
本发明式(A)化合物的制备还包含如下步骤,
Figure PCTCN2021128735-appb-000011
i、式(II-D)化合物、2-氯-4-(4-氟苯基)噻唑-5-氰基、2,6-二甲基吡啶和非质子极性溶剂,在60~80℃下反应,得到化合物(II-A);
ii、将步骤i得到的化合物(II-A),溶于有机溶剂B中,在氮气保护下冷却至0℃,加入还原剂,反应完成后加入碘甲烷-d 3,得到化合物(II-B);
iii、将步骤ii得到的化合物(II-B)溶于有机溶剂C中,加入三氟乙酸,反应得到式(II-C)化合物。
本发明式(A)化合物的制备方法包含如下步骤,
Figure PCTCN2021128735-appb-000012
进一步的,上述制备方法,所述非质子极性溶剂为N,N-二甲基乙酰胺、二甲基甲酰胺、六甲基磷酰胺、乙腈、丙酮、二甲亚砜中的至少一种。
更进一步的,所述非质子极性溶剂为N,N-二甲基乙酰胺。
进一步的,上述制备方法,所述有机溶剂B为四氢呋喃、N,N-二甲基乙酰胺、二甲基甲酰胺、乙腈、丙酮中的至少一种。
更进一步的,所述有机溶剂B为四氢呋喃。
进一步的,上述制备方法,所述还原剂为氢化钠。
进一步的,上述制备方法,所述有机溶剂C为二氯甲烷、乙酸甲酯、碳酸二甲酯、丙二醇甲醚乙酸酯、尼龙酸二甲酯中的至少一种。
更进一步的,所述有机溶剂C为二氯甲烷。
作为本发明的又一技术方案,本发明提供了一种药物组合物,其含有前述任意一项技术方案所述的化合物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,或者式(A)化合物的晶型I,以及药学上可接受的载体和/或赋形剂。
作为本发明的再一技术方案,本发明提供了前述任意一项技术方案所述的化合物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,或者前述组合物在制备治疗ATX介导的疾病的药物中的应用。所述ATX介导的疾病为特发性肺纤维化。
可以理解的是,热重分析(TGA)和差示扫描量热(DSC)为本领域中所熟知的,TGA曲线和DSC曲线的熔融峰高取决于与样品制备和仪器几何形状有关的许多因素,而峰位置对实验细节相对不敏感。因此,在一些实施方案中,本发明的结晶化合物具有特征峰位置的TGA图和DSC图,具有与本发明附图中提供的TGA图和DSC图实质上相同的性质,测量值误差容限为±5℃内,一般要求在±3℃内。
可以理解的是,本发明描述的和保护的数值为近似值。数值内的变化可能归因于设备的校准、设备误差、晶体的纯度、晶体大小、样本大小以及其他因素。
可以理解的是,本发明的晶型不限于与本发明公开的附图中描述的特征图谱完全相同的特征图谱,比如XRD、DSC、TGA、DVS、等温吸附曲线图,具有与附图中描述的哪些图谱基本上相同或本质上相同的特征图谱的任何晶型均落入本发明的范围内。
术语
“治疗有效量”指引起组织、系统或受试者生理或医学反应的化合物的量,此量是所寻求的,包括在受治疗者身上施用时足以预防受治疗的疾患或病症的一种或几种症状发生或使其减轻至某种程度的化合物的量。
“室温”:10℃-30℃,>30%RH。
“药学上可接受的盐”是指本发明化合物保持游离酸或者游离碱的生物有效性和特性,且所述的游离酸通过与无毒的无机碱或者有机碱,所述的游离碱通过与无毒的无机酸或者有机酸反应获得的盐。
“药物组合物”表示一种或多种本发明所述化合物或其立体异构体、溶剂化物、药学上可接受的盐、共晶、氘代物,与其他组成成分的混合物,其中其他组分包含生理学/药学上可接受的载体和/赋形剂。
“载体”指的是:不会对生物体产生明显刺激且不会消除所给予化合物的生物活性和特性,并能改变药物进入人体的方式和在体内的分布、控制药物的释放速度并将药物输送到靶向器官的体系,非限制性的实例包括微囊与微球、纳米粒、脂质体等。
“赋形剂”指的是:其本身并非治疗剂,用作稀释剂、辅料、粘合剂和/或媒介物,用于添加至药物组合物中以改善其处置或储存性质或允许或促进化合物或药物组合物形成用于给药的单位剂型。如本领域技术人员所已知的,药用赋形剂可提供各种功能且可描述为润湿剂、缓冲剂、助悬剂、润滑剂、乳化剂、崩解剂、吸收剂、防腐剂、表面活性剂、着色剂、矫味剂及甜味剂。药用赋形剂的实例包括但不限于:(1)糖,例如乳糖、葡萄糖及蔗糖; (2)淀粉,例如玉米淀粉及马铃薯淀粉;(3)纤维素及其衍生物,例如羧甲基纤维素钠、乙基纤维素、乙酸纤维素、羟丙基甲基纤维素、羟丙基纤维素、微晶纤维素及交联羧甲基纤维素(例如交联羧甲基纤维素钠);(4)黄蓍胶粉;(5)麦芽;(6)明胶;(7)滑石;(8)赋形剂,例如可可脂及栓剂蜡;(9)油,例如花生油、棉籽油、红花油、芝麻油、橄榄油、玉米油及大豆油;(10)二醇,例如丙二醇;(11)多元醇,例如甘油、山梨醇、甘露醇及聚乙二醇;(12)酯,例如油酸乙酯及月桂酸乙酯;(13)琼脂;(14)缓冲剂,例如氢氧化镁及氢氧化铝;(15)海藻酸;(16)无热原水;(17)等渗盐水;(18)林格溶液(Ringer’s solution);(19)乙醇;(20)pH缓冲溶液;(21)聚酯、聚碳酸酯和/或聚酐;及(22)其他用于药物制剂中的无毒相容物质。
“立体异构体”是指由分子中原子在空间上排列方式不同所产生的异构体,包括顺反异构体、对映异构体和构象异构体。
“溶剂化物”指本发明化合物或其盐与分子间非共价力结合的化学计量或非化学计量的溶剂形成的物质。当溶剂为水时,则为水合物。
“共晶”是指活性药物成分(API)和共晶形成物(CCF)在氢键或其他非共价键的作用下结合而成的晶体,其中API和CCF的纯态在室温下均为固体,并且各组分间存在固定的化学计量比。共晶是一种多组分晶体,既包含两种中性固体之间形成的二元共晶,也包含中性固体与盐或溶剂化物形成的多元共晶。
附图说明
图1是化合物A晶型I使用Cu-Kα辐射的X-射线粉末衍射图谱;
图2是化合物A晶型I的TGA曲线图;
图3是化合物A晶型I的DSC曲线图;
图4是化合物A晶型I的PLM图;
图5是化合物A晶型I的DVS图。
具体实施方式
以下将通过实施例对本发明的内容进行详细描述。实施例中未注明具体条件的,按照常规条件的实验方法进行。所举实施例是为了更好地对本发明的内容进行说明,但并不能理解为本发明的内容仅限于所举实例。本领域常规技术人员根据上述发明内容对实施方案进行非本质的改进和调整,仍属于本发明的保护范围。
无特殊说明,原料购买于泰坦科技、安耐吉化学、上海德默、成都科龙化工、韶远化 学科技、南京药石、药明康德和百灵威科技等公司。
仪器信息和方法
化合物的结构是通过核磁共振(NMR)或(和)质谱(MS)来确定的。NMR位移(δ)以10-6(ppm)的单位给出。NMR的测定是用(Bruker Avance III 400和Bruker Avance 300)核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代氯仿(CDCl 3),氘代甲醇(CD 3OD),内标为四甲基硅烷(TMS);
MS的测定用(Agilent 6120B(ESI)和Agilent 6120B(APCI));
HPLC的测定使用Agilent 1260DAD高压液相色谱仪(Zorbax SB-C18 100×4.6mm,3.5μM);
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm-0.20mm,薄层层析分离纯化产品采用的规格是0.4mm-0.5mm;
柱层析一般使用烟台黄海硅胶200-300目硅胶为载体。
X射线粉末衍射
XRPD图在PANalytacal生产的X射线粉末衍射分析仪上采集,扫描参数如下表所示。
Figure PCTCN2021128735-appb-000013
热重分析(TGA)和差示扫描量热(DSC)
TGA和DSC图分别在TA Q5000/5500热重分析仪和TA 2500差示扫描量热仪上采集,下表列出了测试参数。
参数 TGA DSC
方法 线性升温 线性升温
样品盘 铝盘,敞开 铝盘,压盖/不压盖
温度范围 室温-设置终点温度 25℃-设置终点温度
扫描速率(℃/min) 10 10
保护气体 氮气 氮气
动态水分吸附(DVS)
动态水分吸附(DVS)曲线在SMS(Surface Measurement Systems)的DVS Intrinsic上采集。在25℃时的相对湿度用LiCl,Mg(NO 3) 2和KCl的潮解点校正。DVS测试参数列于下表。
Figure PCTCN2021128735-appb-000014
偏光显微镜(PLM)
偏光显微数据通过Axio Lab.A1正置式显微镜在室温下进行采集。
简写说明:
NMP:N-甲基吡咯烷酮;
THF:四氢呋喃;
HATU:2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯;
PE:石油醚;
EA:乙酸乙酯;
DCM:二氯甲烷;
MeOH:甲醇;
Acetone:丙酮;
EtOH:乙醇;
ACN:乙腈;
H 2O:水。
中间体1:
(丁-1-炔-1-基-d5)三甲基硅烷(中间体1)
(but-1-yn-1-yl-d5)trimethylsilane
Figure PCTCN2021128735-appb-000015
室温条件下将三甲基硅基乙炔(1a)(10.07g,102.5mmol)溶于干燥四氢呋喃(45mL)中,体系冷却至-78℃后向其中滴加正丁基锂(45mL,2M正己烷溶液),滴加完全后转移至冰水浴中继续反应20分钟。之后冷却至-78℃,滴加六甲基磷酸三胺(18.37g,102.5mmol)在此条件下继续搅拌30分钟后滴加碘乙烷-d5(1b)(15.00g,93.2mmol),滴加完全后自然升至室温反应过夜。向反应液中加入水(20mL)淬灭反应,分液,水相用乙醚萃取(20mL),合并有机相,有机相用饱和食盐水洗涤(20mL),无水硫酸钠干燥,过滤,滤液常压蒸馏,收集80℃至100℃馏分。得到标题化合物(丁-1-炔-1-基-d5)三甲基硅烷(中间体1),无色透明液体(10.0g,产率74%)。
中间体2:
2-氯-1-(3-羟基氮杂环丁烷-1-基-3-d)乙-1-酮(中间体2)
2-chloro-1-(3-hydroxyazetidin-1-yl-3-d)ethan-1-one
Figure PCTCN2021128735-appb-000016
第一步:叔丁基3-羟基氮杂环丁烷-1-羧酸酯-3-d(2b)
tert-butyl 3-hydroxyazetidine-1-carboxylate-3-d
Figure PCTCN2021128735-appb-000017
将1-Boc-3-氮杂环丁酮(2a)(5.13g,30mmol)溶于四氢呋喃(50mL)中,搅拌均匀后,分批加入硼氢化钠-d4(1.26g,30mmol),室温反应1小时。加入水(20mL)淬灭反应,搅拌10分钟后,加入乙酸乙酯(100mL)和饱和碳酸氢钠(100mL),萃取分层,有机相用饱和食盐水洗(100mL),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物叔丁基3-羟基氮杂环丁烷-1-羧酸酯-3-d(2b),透明油状物(4.6g,收率87%)。
1H NMR(400MHz,CDCl 3)δ4.14(d,2H),3.79(d,2H),1.44(s,9H)。
第二步:
氮杂环丁烷-3-d-3-醇(2c)
azetidin-3-d-3-ol
Figure PCTCN2021128735-appb-000018
将化合物叔丁基3-羟基氮杂环丁烷-1-羧酸酯-3-d(2b)(4.6g,26.3mmol)溶于二氯甲烷(24mL)中,向体系滴加三氟乙酸(8mL),滴加完全后,室温反应1h,减压浓缩得到氮杂环丁烷-3-d-3-醇(2c)的三氟乙酸盐,棕色油状物,(4.94g,收率100%),粗产品直接进行下一步反应。
第三步:
2-氯-1-(3-羟基氮杂环丁烷-1-基-3-d)乙-1-酮(中间体2)
2-chloro-1-(3-hydroxyazetidin-1-yl-3-d)ethan-1-one
Figure PCTCN2021128735-appb-000019
将氮杂环丁烷-3-d-3-醇(2c)的三氟乙酸盐(4.94g,26.3mmol)溶于水(40mL)中,搅拌均匀后,分批加入碳酸钾(12.4g,60mmol),待体系不再冒泡后加入二氯甲烷(40mL)。冰浴条件下滴加氯乙酰氯(3.39g,30mmol),滴加完全后室温反应2小时。加入乙酸乙酯(100mL)和水(100mL),萃取分层,水相用乙酸乙酯(100mL)萃取。合并有机相,有机相用饱和食盐水洗(100mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离得到目标化合物2-氯-1-(3-羟基氮杂环丁烷-1-基-3-d)乙-1-酮(中间体2),白色固体(0.85g,收率21%)。
1H NMR(400MHz,CDCl 3)δ4.49(d,1H),4.30(d,1H),4.15(d,1H),3.94(d,1H),3.90(s,2H),3.01(s,1H)。
中间体3:
乙基2-溴乙酸酯-d2(中间体3)
ethyl 2-bromoacetate-d2
Figure PCTCN2021128735-appb-000020
将乙酸-d4(3a)(1.0g,15.6mmol)溶于三氟乙酸酐(6mL)中,搅拌均匀后,加入4-二甲氨基吡啶(30mg,0.25mmol),将体系加热至60℃,在此条加下缓慢滴加溴素(0.98mL),滴加时间1小时。滴加完全后继续在此条件下反应1小时后冷却至室温,向体系通入氮气, 直到体系体积减少至约1mL左右。加入乙醇(20mL),60℃条件下反应过夜后冷却至室温。减压浓缩,加入乙醚(100mL),饱和食盐水洗(100mL),无水硫酸钠干燥,过滤,滤液减压浓缩得到乙基2-溴乙酸酯-d2(中间体3),淡黄色液体(1.65g,收率62.6%)。
1H NMR(400MHz,CDCl 3)δ4.24(q,J=7.1Hz,1H),1.30(t,J=7.1Hz,2H)。
中间体4:
Figure PCTCN2021128735-appb-000021
第一步:
4-氟苯甲酸甲酯-2,3,5,6-d4(4b)
methyl 4-fluorobenzoate-2,3,5,6-d4
Figure PCTCN2021128735-appb-000022
将4-氟苯甲酸--2,3,5,6-d4(4a)(1.0g,6.94mmol)溶于N,N-二甲基甲酰胺(10mL)中,冷却至0℃,加入碳酸钾(1.92g,13.9mmol),此条件下反应5分钟后,向体系滴加硫酸二甲酯(1.05g,8.33mmol)。加毕,室温反应1小时。加入水(20mL)淬灭反应,乙酸乙酯萃取(50mL×2),合并有机相,饱和食盐水洗(50mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离(石油醚:乙酸乙酯(v/v)=8:1)得到目标化合物4-氟苯甲酸甲酯-2,3,5,6-d4(4b),为透明液体(0.96g,收率87%)。
1H NMR(400MHz,CDCl 3)δ3.91(s,3H)。
第二步:
3-(4-氟苯基-2,3,5,6-d4)-3-氧代丙腈(4c)
3-(4-fluorophenyl-2,3,5,6-d4)-3-oxopropanenitrile
Figure PCTCN2021128735-appb-000023
将4-氟苯甲酸甲酯-2,3,5,6-d4(4b)(0.96g,6.1mmol)溶于甲苯(8mL)中,氮气保护下冷却至-10℃,加入乙腈(1.46g,36.3mmol),向体系缓慢滴加二(三甲基硅基)氨基钠(6mL,12mmol,2M in THF)。滴加完全后,盐酸(20mL,1N水溶液)淬灭反应。升至室温,乙酸乙酯萃取(50mL×2),合并有机相,饱和食盐水洗(50mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物3-(4-氟苯基-2,3,5,6-d4)-3-氧代丙腈(4c),为淡黄色固体(0.92g,收率91%)。
1H NMR(400MHz,CDCl 3)δ4.05(s,2H)。
第三步:
2-氨基-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(4d)
2-amino-4-(4-fluorophenyl-2,3,5,6-d4)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000024
将3-(4-氟苯基-2,3,5,6-d4)-3-氧代丙腈(4c)(0.92g,5.5mmol)溶于乙醇(12mL)中,加入吡啶(0.44g,5.5mmol)后升温至70℃反应15分钟。冷却至室温,向体系缓慢滴加硫脲(0.84g,11mmol)和碘单质(1.4g,5.5mmol)的乙醇混悬液(8mL)。滴加完全后,室温反应1小时。硫代硫酸钠(10mL,1N水溶液)淬灭反应。过滤,滤饼干燥得到目标化合物2-氨基-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(4d),为白色固体(0.82g,收率67%)。
第四步:
2-氯-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(中间体4)
2-chloro-4-(4-fluorophenyl-2,3,5,6-d4)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000025
将氯化铜(0.60g,4.5mmol)溶于乙腈(8mL)中,向体系滴加亚硝酸叔丁酯(0.67g,5.6mmol),室温下搅拌30分钟后。向体系分批加入2-氨基-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(4d)(0.82g,3.67mmol)。加入完全后,室温继续反应1小时。盐酸(10mL,1N水溶液)淬灭反应。加入乙酸乙酯(80mL),饱和食盐水洗(50mL),无水硫酸钠干燥,过滤,滤液 减压浓缩,残留物经硅胶柱层析分离(二氯甲烷)得到目标化合物2-氯-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(中间体4),为淡黄色固体(0.68g,收率76%)。
实施例1:
2-((2-(乙基-d5)-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物1)
2-((2-(ethyl-d5)-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000026
第一步:
4-(2-氯-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(1C)
tert-butyl 4-(2-chloro-5-fluoropyridin-4-yl)piperazine-1-carboxylate(1C)
Figure PCTCN2021128735-appb-000027
500mL反应瓶中,加入NMP(220mL),开启搅拌,再依次加入2-氯-5-氟-4-碘吡啶(1A)(44.00g,170.9mmol)、1-Boc-哌嗪(1B)(47.75g,256.4mmol)和碳酸钾(47.24g,341.8mmol),升温至150℃反应3小时后停止反应。反应液冷却至室温,缓慢将反应物加入到冰水(500mL)中,白色固体析出,搅拌0.5小时,过滤。滤饼用水洗涤后使用正己烷(200mL)打浆,过滤,干燥得到目标化合物4-(2-氯-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(1C),白色固体(50g,产率64%)。
LCMS m/z=316.2[M+1] +
第二步:
4-(2-(2-(丁-1-炔-1-基-d5)-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(1D)
tert-butyl 4-(2-(but-1-yn-1-yl-d5)-5-fluoropyridin-4-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000028
将4-(2-氯-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(1C)(23.18g,71.6mmo),(丁-1-炔-1-基-d5)三甲基硅烷(中间体1)(9.4g,131.3mmol),双三苯基磷二氯化钯(5.03g,7.2mmol),1,3-双(二苯基膦)丙烷(4.43g,10.7mmol)和氟化铯(22.8g,143.3mmol)依次加入到二甲基亚砜(184mL)中,体系用氮气置换三次,95℃下反应4小时。反应结束后冷却至室温,加入水(200mL),水相用乙酸乙酯(200mL×3)萃取,合并有机相用饱和氯化钠水溶液(200mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残留物经硅胶柱层析分离(PE:EA=20:1~5:1)得到4-(2-(2-(丁-1-炔-1-基-d5)-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(1D),淡棕色粘稠液体(10.15g,产率41.9%)。
LCMS m/z=339.2[M+1] +
第三步:
4-(2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-甲酸叔丁酯(1F)
tert-butyl 4-(2-(ethyl-d5)-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000029
250mL反应瓶中,加入乙醇(100mL)和4-(2-(2-(丁-1-炔-1-基-d5)-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(1D)(10.15g,30.0mmol),冷却至0℃,分批加入2-[(氨基氧基)磺酰]-1,3,5-三甲基苯(1E)(11.00g,51.1mmol),再将碳酸氢钠(5.04g,60.0mmol)加入反应中,加毕室温反应2小时后再将碳酸钾(8.29g,60.0mmol)加入反应中,室温反应过夜。反应液加入水(150mL),水相用乙酸乙酯(200mL×2)萃取,合并有机相用饱和氯化钠(100mL)洗 涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离纯化得到目标化合物4-(2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-甲酸叔丁酯(1F),白色固体(7.53g,产率71%)。
LCMS m/z=354.3[M+1] +
第四步:
4-(2-(乙基-d5)-6-氟-3-亚硝基吡唑并[1,5-a]吡啶基-5-基)哌嗪-1-羧酸叔丁酯(1G)
tert-butyl
4-(2-(ethyl-d5)-6-fluoro-3-nitrosopyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000030
将4-(2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-甲酸叔丁酯(1F)(7.50g,21.2mmol)溶于甲醇(37.5mL)和乙酸(7.5mL),降温至5℃,再将亚硝酸钠(2.93g,42.4mmol)溶于水(10mL)中,滴加到反应液中,加毕,室温反应16小时。向反应液滴加水(30mL),加毕,过滤,滤饼用水(10mL×2)洗涤,干燥得到目标化合物4-(2-(乙基-d5)-6-氟-3-亚硝基吡唑并[1,5-a]吡啶基-5-基)哌嗪-1-羧酸叔丁酯(1G)的粗品,墨绿色固体(11.5g)。
LCMS m/z=383.3[M+1] +
第五步:
4-(3-氨基-2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸叔丁酯(1H)
tert-butyl
4-(3-amino-2-(ethyl-d5)-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000031
将4-(2-(乙基-d5)-6-氟-3-亚硝基吡唑并[1,5-a]吡啶基-5-基)哌嗪-1-羧酸叔丁酯(1G)(11.5g,粗品)溶于乙醇(40mL)和水(20mL)中,再将氯化铵(15.78g,295mmol)和铁粉(8.23g,147mmol)加入反应中,65℃反应20分钟,过滤。滤液用二氯甲烷萃取(50mL×3),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物4-(3-氨基-2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸叔丁酯(1H)的粗品,黄色固体(7.85g)。
LCMS m/z=369.2[M+1] +
第六步:
叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)氨基)-2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5- 基)哌嗪-1-羧酸酯(1J)
tert-butyl 4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)amino)-2-(ethyl-d5)-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000032
将4-(3-氨基-2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸叔丁酯(1H)(7.85g,21.3mmol)、2-氯-4-(4-氟苯基)噻唑-5-氰基(1I)(6.1g,25.6mmol)(参考文献J.Med.Chem.2017,60,3580-3590,DOI:10.1021/acs.jmedchem.7b00032)、2,6-二甲基吡啶(3.42g,31.9mmol)和N,N-二甲基乙酰胺(40mL),升温至70℃下反应4小时。加入10%氯化钠溶液(100mL)淬灭反应,乙酸乙酯萃取(150mL×3),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)氨基)-2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(1J)的粗品,黄棕色粘稠物(15.50g)。
LCMS m/z=571.2[M+1] +
第七步:
叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(1K)
tert-butyl4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000033
将叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)氨基)-2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(1J)的粗品(15.50g)溶于四氢呋喃(60mL)中,氮气保护下冷却至0℃,分批加入钠氢(1.3g,32.6mmol,60%wt),加毕,此条加下反应10分钟后,向体系滴加碘甲烷(4.63g,32.6mmol)。加毕,室温反应30分钟。加入水(100mL)淬灭反应,乙酸乙酯萃取(100mL×2),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩残留物经硅胶柱层析分离得到目标化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(1K),黄色固体 (11.56g,四步产率93%)。
第八步:
2-((2-(乙基-d5)-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(1L)
2-((2-(ethyl-d5)-6-fluoro-5-(piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile hydrochloride
Figure PCTCN2021128735-appb-000034
将化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(1K)(11.56g,19.8mmol)溶于二氯甲烷(110mL)中,向体系滴加三氟乙酸(35mL),反应约3小时后,减压浓缩。残留物中加入二氯甲烷(200mL),饱和碳酸氢钠调节pH至8,分液,水相用二氯甲烷萃取(200mL×2),合并有机相,饱和食盐水洗(300mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物2-((2-(乙基-d5)-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(1L),黄色固体(10.7g)。
第九步:
2-((2-(乙基-d5)-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物1)
2-((2-(ethyl-d5)-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000035
将2-((2-(乙基-d5)-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(1L)(10.7g)溶于乙腈(100mL)中,向体系依次加入2-氯-1-(3羟基氮杂环丁烷-1-基)乙酮(1M)(3.63g,24.3mmol,参考文献J.Med.Chem.2017,60,3580-3590的方法合成)、碳酸钾(6.11g,44.2mmol),升温至80℃下反应过夜。加入水(100mL)淬灭反应,乙酸乙酯萃取(150mL×3),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩硅胶柱层析分离(二氯甲烷/甲醇=20:1)得到标题化合物2-((2-(乙基-d5)-6- 氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物1)(6.2g,两步53%)。
1H NMR(400MHz,CDCl 3)δ8.27(d,1H),8.18–8.10(m,2H),7.20–7.11(m,2H),6.45(d,1H),4.67(s,1H),4.50-4.40(m,1H),4.32-4.23(m,1H),4.15-4.07(m,1H),3.93-3.85(m,1H),3.58(s,3H),3.30-3.20m,4H),3.19–3.06(m,2H),2.85-2.67(m,5H)。
LCMS m/z=598.1[M+1] +
实施例2:
2-((2-(乙基-d5)-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物2)
2-((2-(ethyl-d5)-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000036
第一步:
叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨)-2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(2A)
tert-butyl 4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl-d3)amino)-2-(ethyl-d5)-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000037
将叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)氨基)-2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(1J)的粗品(9.2g,16.1mmol)溶于四氢呋喃(60mL)中,氮气保护下冷却至0℃,分批加入钠氢(0.77g,19.3mmol,60%wt),加毕,此条件下反应10分钟后,向体系滴加碘甲烷-d3(2.80g,19.3mmol)。加毕,室温反应30分钟。加入水(100mL)淬灭反应,乙酸乙酯萃取(100mL×2),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩残留物经硅胶柱层析分离得到目标化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨)-2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(2A),黄色固体(8.0g,产率84.5%)。
第二步:
2-((2-(乙基-d5)-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨)-4-(4-氟苯基)噻唑-5-甲腈(2B)
2-((2-(ethyl-d5)-6-fluoro-5-(piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000038
将化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨)-2-(乙基-d5)-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(2A)(8.0g,13.6mmol)溶于二氯甲烷(90mL)中,向体系滴加三氟乙酸(30mL),反应约3小时后,减压浓缩。残留物中加入二氯甲烷(100mL),饱和碳酸氢钠调节pH至8,分液,水相用二氯甲烷萃取(100mL×3),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物2-((2-(乙基-d5)-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨)-4-(4-氟苯基)噻唑-5-甲腈(2B),黄色固体(6.40g,收率96.3%)。
第三步:
2-((2-(乙基-d5)-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物2)
2-((2-(ethyl-d5)-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl)pyrazol o[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000039
将2-((2-(乙基-d5)-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨)-4-(4-氟苯基)噻唑-5-甲腈(2B)(6.40g,13.1mmol)溶于乙腈(100mL)中,向体系依次加入2-氯-1-(3羟基氮杂环丁烷-1-基)乙酮(1M)(2.16g,14.4mmol)、碳酸钾(3.62g,26.2mmol),升温至80℃下反应5小时。加入水(60mL)淬灭反应,乙酸乙酯萃取(100mL×3),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩硅胶柱层析分离(二氯甲烷/甲醇=20:1)得到标题化合物2-((2-(乙基-d5)-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-腈(化合物2)(5.1g,两步64.8%)。
1H NMR(400MHz,CDCl 3)δ8.27(d,1H),8.20–8.11(m,2H),7.20–7.11(m,2H),6.45(d,1H),4.67(s,1H),4.50-4.40(m,1H),4.32-4.24(m,1H),4.15-4.07(m,1H),3.94-3.86(m,1H),3.31–3.06(m,6H),2.86–2.66(m,5H)。
LCMS m/z=601.2[M+1] +
实施例3:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物3)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000040
Figure PCTCN2021128735-appb-000041
第一步:
4-(2-(丁-1-炔-1-基)-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(3A)
tert-butyl 4-(2-(but-1-yn-1-yl)-5-fluoropyridin-4-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000042
将4-(2-氯-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(1C)(20.1g,62mmo),(丁-1-炔-1-基)三甲基硅烷(9.3g,74mmol),双三苯基磷二氯化钯(4.4g,6.3mmol),1,3-双(二苯基膦)丙烷(3.84g,9.3mmol)和氟化铯(19.0g,125mmol)依次加入到二甲基亚砜(160mL)中,体系用氮气置换三次,95℃下反应4小时。反应结束后冷却至室温,加入水(160mL),水相用乙酸乙酯(200mL×3)萃取,合并有机相用饱和氯化钠水溶液(200mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残留物经硅胶柱层析分离(PE:EA=20:1~5:1)得到4-(2-(丁-1-炔-1-基)-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(3A),淡棕色粘稠液体(11.0g,产率53%)。
LCMS m/z=334.2[M+1] +
第二步:
4-(2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-甲酸叔丁酯(3B)
tert-butyl 4-(2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000043
250mL反应瓶中,加入乙醇(100mL)和4-(2-(丁-1-炔-1-基)-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(3A)(11.0g,33mmol),冷却至0℃,分批加入2-[(氨基氧基)磺酰]-1,3,5-三甲基苯(1E)(10.7g,49.5mmol),再将碳酸氢钠(5.5g,66mmol)加入反应中,加毕室温反应2小时后再将碳酸钾(9.12g,66mmol)加入反应中,室温反应过夜。反应液加入水(200mL),水相用乙酸乙酯(200mL×2)萃取,合并有机相用饱和氯化钠(100mL)洗涤,无水硫酸钠干燥,过滤,浓缩,硅胶柱层析分离得到目标化合物4-(2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-甲酸叔丁酯(3B),白色固体(7.8g,产率68%)。
LCMS m/z=349.2[M+1] +
第三步:
4-(2-乙基-6-氟-3-亚硝基吡唑并[1,5-a]吡啶基-5-基)哌嗪-1-羧酸叔丁酯(3C)
tert-butyl 4-(2-ethyl-6-fluoro-3-nitrosopyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000044
将4-(2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-甲酸叔丁酯(3B)(7.40g,22.4mmol)溶于甲醇(40mL)和乙酸(8mL),降温至5℃,再将亚硝酸钠(3.09g,44.8mmol)溶于水(10mL)中,缓慢滴加到反应液中,加毕,室温反应16小时。向反应液滴加水(30mL),滴加完全后,过滤,滤饼用水(10mL×2)洗涤,干燥得到目标化合物4-(2-乙基-6-氟-3-亚硝基吡唑并[1,5-a]吡啶基-5-基)哌嗪-1-羧酸叔丁酯(3C)的粗品,墨绿色固体(10.8g)。
LCMS m/z=378.3[M+1] +
第四步:
4-(3-氨基-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸叔丁酯(3D)
tert-butyl 4-(3-amino-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000045
将4-(2-乙基-6-氟-3-亚硝基吡唑并[1,5-a]吡啶基-5-基)哌嗪-1-羧酸叔丁酯(3C)(10.8g,粗品)溶于乙醇(40mL)和水(20mL)中,再将氯化铵(14.98g,0.28mol)和铁粉(7.84g,0.14mol)加入反应中,65℃反应20分钟,过滤。滤液用二氯甲烷萃取(60mL×3),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物4-(3-氨基-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸叔丁酯(3D)的粗品,黄色固体 (7.12g,两步收率87.5%)。
LCMS m/z=364.2[M+1] +
第五步:
叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(3E)
tert-butyl 4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000046
将4-(3-氨基-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸叔丁酯(3D)(7.12g,19.6mmol)、2-氯-4-(4-氟苯基)噻唑-5-氰基(1I)(5.61g,23.5mmol)、2,6-二甲基吡啶(3.15g,29.4mmol)和N,N-二甲基乙酰胺(40mL),升温至70℃下反应5小时。加入10%氯化钠溶液(100mL)淬灭反应,乙酸乙酯萃取(150mL×3),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离得到目标化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(3E),黄棕色固体(10.0g,收率90.3%)。
LCMS m/z=566.2[M+1] +
第六步:
叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(3F)
tert-butyl 4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl-d3)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000047
将叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(3E)(10.0g,17.7mmol)溶于四氢呋喃(60mL)中,氮气保护下冷却至0℃,分批加入钠氢(0.85g,21.2mmol,60%wt),加毕,此条加下反应10分钟后,向体系滴加碘甲烷-d3(3.07g,21.2mmol)。加毕,室温反应30分钟。加入水(100mL)淬灭反应,乙酸乙酯萃取(100mL×2),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤, 滤液减压浓缩得到目标化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(3F)的粗品,黄色固体(11.7g)。
LCMS m/z=583.2[M+1] +
第七步:
2-((2-乙基-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(3G)
2-((2-ethyl-6-fluoro-5-(piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile hydrochloride
Figure PCTCN2021128735-appb-000048
将化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(3F)的粗品(11.7g)溶于二氯甲烷(100mL)中,向体系滴加三氟乙酸(33mL),反应约3小时后,减压浓缩。残留物中加入二氯甲烷(200mL),饱和碳酸氢钠调节pH至8,分液,水相用二氯甲烷萃取(200mL×2),合并有机相,饱和食盐水洗(300mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物2-((2-乙基-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(3G)的粗品,黄色固体(10.0g)。
LCMS m/z=483.2[M+1] +
第八步:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物3)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000049
将2-((2-乙基-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(3G)的粗品(10.0g)溶于乙腈(100mL)中,向体系依次加入2-氯-1-(3羟基氮杂环丁烷-1-基)乙酮(1M)(3.35g,22.4mmol)、碳酸钾(5.55g,40.1mmol),升温至80℃下反应6小时。加入水(100mL)淬灭反应,乙酸乙酯萃取(150mL×3),合并有机相,饱和食 盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离(二氯甲烷/甲醇=20:1)得到标题化合物2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物3)(7.6g,三步72.1%)。
1H NMR(400MHz,CDCl 3)δ8.27(d,1H),8.20–8.10(m,2H),7.20–7.11(m,2H),6.45(d,1H),4.67(s,1H),4.50-4.40(m,1H),4.32-4.24(m,1H),4.16-4.08(m,1H),3.94-3.86(m,1H),3.29–3.07(m,6H),2.78(s,4H),2.72(q,2H),2.58(s,1H),1.32(t,3H)。
LCMS m/z=596.3[M+1] +
实施例4:
2-((2-(乙基)-6-氟-5-(4-(2-(3-羟基氮杂环丁基-1-基)-2-氧代乙基)哌嗪-1-基-2,2,3,3,5,5,6,6-d8)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物4)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl-2,2,3,3,5,5,6,6-d8)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000050
Figure PCTCN2021128735-appb-000051
第一步:
叔丁基4-(2-氯-5-氟吡啶-4-基)哌嗪-1-羧酸酯-2,2,3,3,5,5,6,6-d8(4B)
tert-butyl 4-(2-chloro-5-fluoropyridin-4-yl)piperazine-1-carboxylate-2,2,3,3,5,5,6,6-d8
Figure PCTCN2021128735-appb-000052
250mL反应瓶中,加入NMP(93mL),再依次加入2-氯-5-氟-4-碘吡啶(1A)(9.3g,36.2mmol)、哌嗪-1碳酸叔丁酯-2,2,3,3,5,5,6,6-氘8(4A)(4.5g,24.2mmol)和碳酸钾(6.7g,48.3mmol),升温至150℃反应3小时后停止反应。反应液冷却至室温,缓慢将反应物加入到冰水(200mL)中,白色固体析出,搅拌0.5小时,过滤。滤饼用水洗涤后使用正己烷(100mL)打浆,过滤,干燥得到目标化合物叔丁基4-(2-氯-5-氟吡啶-4-基)哌嗪-1-羧酸酯-2,2,3,3,5,5,6,6-d8(4B),白色固体(10.01g,产率85%)。
LCMS m/z=324.3[M+1] +
第二步:
4-(2-(叔-1-炔-1-基)-5-氟吡啶-4-基)哌嗪-1-甲酸叔丁酯-2,2,3,3,5,5,6,6-d8(4C)
tert-butyl
4-(2-(but-1-yn-1-yl)-5-fluoropyridin-4-yl)piperazine-1-carboxylate-2,2,3,3,5,5,6,6-d8
Figure PCTCN2021128735-appb-000053
将4-(2-氯-5-氟吡啶-4-基)哌嗪-1-羧酸叔丁酯(4B)(2.00g,6.2mmo),(丁-1-炔-1-基)三甲基硅烷(0.94g,7.4mmol),双三苯基磷二氯化钯(0.44g,0.6mmol),1,3-双(二苯基膦)丙烷(0.38g,0.9mmol)和氟化铯(1.89g,12.4mmol)依次加入到二甲基亚砜(16mL)中,体系用氮气置换三次,95℃下反应5小时。反应结束后冷却至室温,加入水(20mL),水相用乙酸乙酯(20mL×3)萃取,合并有机相用饱和氯化钠水溶液(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残留物经硅胶柱层析分离(PE:EA=15:1~5:1)得到4C(1.20g,产率57%)。
LCMS m/z=342.3[M+1] +
第三步:
4-(2-乙基-6-氟吡唑[1,5-a]吡啶-5-基)哌嗪-1-甲酸叔丁酯-2,2,3,3,5,5,6,6-d8(4D)
tert-butyl 4-(2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate-2,2,3,3,5,5,6,6-d8
Figure PCTCN2021128735-appb-000054
100mL反应瓶中,加入乙醇(12mL)和4C(1.20g,3.5mmol),冷却至0℃,分批加入2-[(氨基氧基)磺酰]-1,3,5-三甲基苯(1E)(1.14g,5.3mmol),再将碳酸氢钠(0.59g,7.0mmol)加入反应中,加毕室温反应2小时后再将碳酸钾(0.97g,7.0mmol)加入反应中,室温反应过夜。反应液加入水(150mL),水相用乙酸乙酯(200mL×2)萃取,合并有机相用饱和氯化钠(100mL洗涤,无水硫酸钠干燥,过滤,浓缩,得4D粗品(2.0g)。
LCMS m/z=357.2[M+1] +
第四步:
4-(2-乙基-6-氟-3-亚硝基吡唑[1,5-a]并吡啶-5-基)哌嗪-1-甲酸叔丁酯-2,2,3,3,5,5,6,6-d8(4E)
tert-butyl 4-(2-ethyl-6-fluoro-3-nitrosopyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate-2,2,3,3,5,5,6,6-d8
Figure PCTCN2021128735-appb-000055
将上一步的4D粗品(2.0g,约5.6mmol)溶于甲醇(10.0mL)和乙酸(2.0mL),降温至 5-10℃,再将亚硝酸钠(0.77g,11.2mmol)的水溶液(2.0mL)滴加到反应液中,加毕,室温反应16小时。向反应液滴加水(20mL),加毕,过滤,滤饼用水(5mL×2)洗涤,抽干得4E的粗品(2.00g)。
第五步:
4-(3-胺基-2-乙基-6-氟吡唑[1,5-a]并吡啶-5-基)哌嗪-1-甲酸叔丁酯-2,2,3,3,5,5,6,6-d8(4F)
tert-butyl 4-(3-amino-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate-2,2,3,3,5,5,6,6-d8
Figure PCTCN2021128735-appb-000056
将上一步的4E粗品(2.00g,约5.2mmol)溶于乙醇(10mL)和水(10mL)中,再将氯化铵(2.78g,52.0mmol)和铁粉(1.46g,26.0mmol)加入反应中,65℃反应2h,过滤。滤液用EA萃取(10mL×3),合并有机相,饱和食盐水洗(10mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩后柱层析纯化(DCM:MeOH=30:1),得到4F(0.92g,收率48%)。
LCMS m/z=372.3[M+1] +
第六步:
4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)胺基)-2-乙基-6-氟吡唑[1,5-a]并吡啶-5-基)哌嗪-1-甲酸叔丁酯-2,2,3,3,5,5,6,6-d8(4G)
tert-butyl 4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate-2,2,3,3,5,5,6,6-d8
Figure PCTCN2021128735-appb-000057
将4F(0.92g,2.5mmol)、2-氯-4-(4-氟苯基)噻唑-5-甲腈(1I)(0.72g,3.0mmol)、2,6-二甲基吡啶(0.40g,3.8mmol)和N,N-二甲基乙酰胺(5mL),升温至70℃下反应过夜。加入10%氯化钠溶液(20mL)淬灭反应,乙酸乙酯萃取(20mL×3),合并有机相,饱和食盐水洗(20mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩后柱层析(DCM:MeOH(v/v)=30:1),得到4G(1.00g,收率70%)。
LCMS m/z=574.3[M+1] +
第七步:
4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)胺基)-2-乙基-6-氟吡唑[1,5-a]吡啶-5-基)哌嗪 -1-甲酸叔丁酯-2,2,3,3,5,5,6,6-d8(4H)
tert-butyl 4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate-2,2,3,3,5,5,6,6-d8
Figure PCTCN2021128735-appb-000058
将4G(15.50g)溶于四氢呋喃(10mL)中,氮气保护下冷却至0℃,分批加入氢化钠(0.10g,2.1mmol,60%wt),加毕,此条件下反应10分钟后,向体系滴加碘甲烷(0.16mL,2.1mmol)。加毕自然升至室温反应1h。加入10mL甲醇淬灭反应,旋干反应液得4H粗品(1.20g),无需纯化,直接下一步反应。
LCMS m/z=588.3[M+1] +
化合物4H还可通过如下方法合成:
Figure PCTCN2021128735-appb-000059
室温下将原料4J(参照WO2019228403A1,Intermediate 3合成)(948mg,2.0mmol)、N-Boc-哌嗪-d8(580mg,3.0mmol)、三二亚苄基丙酮二钯(183mg,0.2mmol)、4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(230mg,0.4mmol)和叔丁醇钠(576mg,6.0mmol)加入到干燥的1,4-二氧六环(20mL)中,氮气置换3次,然后升温至105℃反应2h。冷至室温后旋干,直接柱层析(PE:EA=5:1~2:1),得棕色泡沫状固体4H(530mg,收率45%)。
LCMS m/z=588.3[M+1] +
第八步:2-((2-乙基-6-氟-5-(哌嗪-1-基-2,2,3,3,5,5,6,6-d8)吡唑[1,5-a]吡啶-3-基)(甲基)胺基)-4-(4-氟苯基)噻唑-5-甲腈(4I)
2-((2-ethyl-6-fluoro-5-(piperazin-1-yl-2,2,3,3,5,5,6,6-d8)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000060
0℃氮气保护下将三氟乙酸(5.0mL)滴加到上一步得到化合物4H(1.20g粗品)的二 氯甲烷(15.0mL)溶液中,然后自然升温至室温,继续搅拌2h。旋干反应液后加入10mL二氯甲烷,然后用饱和碳酸钠中和至pH=8,二氯甲烷萃取(10mL×2),水洗(10mL×1),饱和氯化钠洗(10mL×1),无水硫酸钠干燥,过滤,减压浓缩后得4I的粗品(0.90g),无需纯化,直接投入下一步反应。
LCMS m/z=488.3[M+1] +
第九步:
2-((2-(乙基)-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基-2,2,3,3,5,5,6,6-d8)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物4)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl-2,2,3,3,5,5,6,6-d8)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000061
室温下将化合物4I的粗品(860mg,约1.8mmol)、2-氯-1-(3-羟基吖啶-1-基)乙-1-酮(296mg,2.0mmol)和碳酸钾(500mg,3.6mmol)加入到乙腈(50mL)中,然后升温至80℃反应5h。冷至室温后抽滤,二氯甲烷洗涤滤饼(30mL×2),滤液浓缩后柱层析(DCM:MeOH=30:1),得化合物4(880mg,收率81%)。
1H NMR(400MHz,CDCl 3)δ8.27(d,1H),8.19–8.11(m,2H),7.20–7.12(m,2H),6.44(d,1H),4.67(s,1H),4.50-4.40(m,1H),4.32-4.24(m,1H),4.16–4.09(m,1H),3.94-3.86(m,1H),3.58(s,3H),3.24–3.06(m,2H),2.72(q,2H),2.59(s,1H),1.32(t,3H)。
LCMS m/z=601.2[M+1] +
实施例5:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基-3-d)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5甲腈(化合物5)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl-3-d)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000062
Figure PCTCN2021128735-appb-000063
第一步:
叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(5A)
tert-butyl 4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000064
将叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(3E)(2.8g,5.0mmol)溶于四氢呋喃(30mL)中,氮气保护下冷却至0℃,分批加入钠氢(0.24g,6.0mmol,60%wt),加毕,此条件下反应10分钟后,向体系滴加碘甲烷(1.42g,10mmol)。加毕,室温反应30分钟。加入水(50mL)淬灭反应,乙酸乙酯萃取(50mL×2),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(5A)的粗品,黄色固体(2.8g)。
LCMS m/z=580.3[M+1] +
第二步:
2-((2-乙基-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(5B)
2-((2-ethyl-6-fluoro-5-(piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000065
叔丁基4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(5A)的粗品(2.8g)溶于二氯甲烷(30mL)中,向体系滴加三氟乙酸(15mL),反应约3小时后,减压浓缩。残留物中加入二氯甲烷(100mL),饱和碳酸氢钠调节pH至8,分液,水相用二氯甲烷萃取(100mL×2),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物2-((2-乙基-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(5B),黄色固体(2.0g,两步收率83.3%)。
LCMS m/z=480.1[M+1] +
第三步:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基-3-d)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物5)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl-3-d)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000066
将2-((2-乙基-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(0.31g,0.65mmol)溶于乙腈(10mL)中,向体系依次加入2-氯-1-(3-羟基氮杂环丁烷-1-基-3-d)乙-1-酮(中间体2)(0.15g,1.0mmol)、碳酸钾(0.27g,2.0mmol),升温至80℃下反应6小时。加入水(50mL)淬灭反应,二氯甲烷萃取(50mL×3),合并有机相,饱和食盐水洗(50mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离(二氯甲烷/甲醇=20:1)得到标题化合物2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基-3-d)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物5)(90mg,23%)。
1H NMR(400MHz,CDCl 3)δ8.27(d,1H),8.18–8.10(m,2H),7.20–7.10(m,2H),6.45(d,1H),4.43(d,1H),4.25(d,1H),4.11(dd,1H),3.89(d,1H),3.58(s,3H),3.28–3.06(m,7H),2.81–2.66(m,6H),1.31(t,3H)。
LCMS m/z=594.2[M+1] +
实施例6:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基-1,1-d2)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物6)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl-1,1-d2)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000067
第一步:
乙基2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸酯-d2(6A)
ethyl 2-(4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazin-1-yl)acetate-d2
Figure PCTCN2021128735-appb-000068
向2-((2-乙基-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(5B)(0.31g,0.65mmol)的乙腈(10mL)溶液中依次添加碳酸钾(0.14g,1mmol)和乙基2-溴乙酸酯-d2(中间体3)(0.17g,1mmol),将混合物在室温下搅拌1小时。反应完成 后,将混合物用饱和食盐水(30mL)稀释,并用乙酸乙酯(30mL×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。残余物通过硅胶柱色谱法(乙酸乙酯:石油醚=1:1)纯化得到乙基2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸酯-d2(6A),淡黄色固体(0.32g,86.7%)。
LC-MS(ESI):m/z=568.2[M+H] +
第二步:
2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸-2,2-d2(6B)
2-(4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazin-1-yl)acetic-2,2-d2acid
Figure PCTCN2021128735-appb-000069
将乙基2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸酯-d2(6A)(0.32g,0.56mmol)溶于四氢呋喃(6mL),加入甲醇(2mL)和水(2mL),搅拌均匀后加入氢氧化锂一水合物(84mg,2mmol),室温反应1小时,用盐酸(1N水溶液)调节pH至5,乙酸乙酯萃取(50mL×2),合并有机相,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩得到2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸-2,2-d2(6B),淡黄色固体(0.24g,产率79%)。
LCMS m/z=540.2[M+1] +
第三步:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基-1,1-d2)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物6)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl-1,1-d2)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000070
将2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸-2,2-d2(6B)(0.24g,0.45mmol)溶于N,N-二甲基甲酰胺(5mL)中,向体系 依次加入三乙胺(0.15g,1.5mmol)、HATU(0.19g,0.5mmol),搅拌10分钟后,加入4-羟基氮杂环丁烷盐酸盐(0.7mmol,76mg)的N,N-二甲基甲酰胺(5mL)溶液,室温反应1小时。加入水(30mL)淬灭反应,二氯甲烷萃取(50mL×3),合并有机相,饱和食盐水洗(50mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离(二氯甲烷/甲醇=20:1)得到标题化合物2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基-1,1-d2)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物6)(0.12g,45%)。
1H NMR(400MHz,CDCl 3)δ8.27(d,1H),8.19–8.10(m,2H),7.20–7.12(m,2H),6.45(d,1H),4.67(s,1H),4.49–4.40(m,1H),4.30–4.24(m,1H),4.16–4.08(m,1H),3.96–3.87(m,1H),3.58(s,3H),3.26(s,4H),2.84(s,4H),2.72(q,2H),2.59(s,1H),1.32(t,3H)。
LCMS m/z=595.3[M+1] +
实施例7:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基-氮杂环丁基-1-基-3-d)-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)胺基)-4-(4-氟苯基)噻唑-5-甲腈(化合物7)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl-3-d)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000071
将2-((2-乙基-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(3G)的粗品(300mg)溶于乙腈(5mL)中,向体系依次加入中间体2(103mg,0.7mmol)、碳酸钾(172mg,1.3mmol),升温至80℃下反应6小时。加入水(10mL)淬灭反应,乙酸乙酯萃取(10mL×3),合并有机相,饱和食盐水洗(10mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩硅胶柱层析分离(二氯甲烷/甲醇(v/v)=20:1)得到白色固体化合物7(200mg,54%)。
1H NMR(400MHz,CDCl 3)δ8.27(d,1H),8.17-8.13(m,2H),7.18-7.14(m,2H),6.45(d,1H),4.46-4.44(m,1H),4.29-4.26(m,1H),4.14-4.10(m,1H),3.92-3.89(m,1H),3.26-3.15(m,6H),2.83(s,4H),2.75-2.69(m,2H),2.54(s,1H),1.32(t,3H)。
LCMS m/z=597.3[M+1] +
实施例8:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基-1,1-d2)哌嗪-1-基)吡唑并 [1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物8)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl-1,1-d2)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000072
第一步:
乙基2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸酯-d2(8A)
ethyl 2-(4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl-d3)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazin-1-yl)acetate-d2
Figure PCTCN2021128735-appb-000073
向化合物3G(0.35g,0.73mmol)的乙腈(10mL)溶液中依次添加碳酸钾(0.21g,1.5mmol)和乙基2-溴乙酸酯-d2(中间体3)(0.17g,1mmol),将混合物在室温下搅拌1小时。反应完成后,将混合物用饱和食盐水(30mL)稀释,并用乙酸乙酯(30mL×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。残余物通过硅胶柱色谱法(乙酸乙酯:石油醚(v/v)=1:1)纯化,得到淡黄色固体(0.38g,收率92%)。
第二步:
2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸-2,2-d2(8B)
2-(4-(3-((5-cyano-4-(4-fluorophenyl)thiazol-2-yl)(methyl-d3)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazin-1-yl)acetic-2,2-d2acid
Figure PCTCN2021128735-appb-000074
将乙基2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸酯-d2(8A)(0.38g,0.67mmol)溶于四氢呋喃(9mL),加入甲醇(3mL)和水(3mL),搅拌均匀后加入氢氧化锂一水合物(126mg,3mmol),室温反应1小时,用盐酸(1N水溶液)调节pH至5,乙酸乙酯萃取(50mL×2),合并有机相,有机相用无水硫酸钠干燥,浓缩,得到2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸-2,2-d2(8B),淡黄色固体(0.32g,产率88%)。
第三步:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基-1,1-d2)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物8)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl-1,1-d2)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000075
将2-(4-(3-((5-氰基-4-(4-氟苯基)噻唑-2-基)(甲基-d3)氨)-2-乙基-6-氟吡唑并[1,5-a]哌啶-5-基)哌嗪-1-基)乙酸-2,2-d2(8B)(0.32g,0.59mmol)溶于N,N-二甲基甲酰胺(10mL)中,向体系依次加入三乙胺(0.20g,2.0mmol)、HATU(0.27g,0.7mmol),搅拌10分钟后,加入4-羟基氮杂环丁烷盐酸盐(1mmol,0.11g)的N,N-二甲基甲酰胺(5mL)溶液,室温反应1小时。加入水(30mL)淬灭反应,二氯甲烷萃取(50mL×3),合并有机相,饱和食盐水洗(50mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩硅胶柱层析分离(二氯甲烷/甲醇=20:1)得到2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基-1,1-d2)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物8)(0.28g,79%)。
1H NMR(400MHz,CDCl 3)δ8.27(d,1H),8.19–8.11(m,2H),7.20–7.11(m,2H),6.44(d,1H),4.67(s,1H),4.49–4.41(m,1H),4.32–4.24(m,1H),4.15–4.08(m,1H),3.94–3.84 (m,1H),3.23(s,4H),2.79–2.66(m,6H),2.62(s,1H),1.32(t,3H)。
LCMS m/z=598.2[M+1] +
实施例9:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(化合物9)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl-2,3,5,6-d4)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000076
第一步:
叔丁基4-(3-((5-氰基-4-(4-氟苯基-2,3,5,6-d4)噻唑-2-基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(9A)
tert-butyl 4-(3-((5-cyano-4-(4-fluorophenyl-2,3,5,6-d4)thiazol-2-yl)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000077
将4-(3-氨基-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸叔丁酯(3D)(1.02g,2.8mmol)、2-氯-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(中间体4)(0.68g,2.8mmol)和2,6-二甲基吡啶(0.60g,5.6mmol)依次加入到N,N-二甲基乙酰胺(20mL)中,升温至70℃下反应5 小时。加入10%氯化钠溶液(60mL)淬灭反应,乙酸乙酯萃取(60mL×3),合并有机相,饱和食盐水洗(60mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离(石油醚:乙酸乙酯(v/v)=3:1)得到目标化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基-2,3,5,6-d4)噻唑-2-基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(9A),黄棕色固体(1.02g,收率64%)。
LCMS m/z=570.2[M+1] +
第二步:
叔丁基4-(3-((5-氰基-4-(4-氟苯基-2,3,5,6-d4)噻唑-2-基)(甲基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(9B)
tert-butyl 4-(3-((5-cyano-4-(4-fluorophenyl-2,3,5,6-d4)thiazol-2-yl)(methyl)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000078
将叔丁基4-(3-((5-氰基-4-(4-氟苯基-2,3,5,6-d4)噻唑-2-基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(9A)(0.51g,0.89mmol)溶于N,N-二甲基甲酰胺(10mL)中,加入碳酸钾(0.27g,2mmo)。向体系滴加碘甲烷(0.19g,1.34mmol)。加毕,室温反应30分钟。加入水(50mL)淬灭反应,乙酸乙酯萃取(50mL×2),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基-2,3,5,6-d4)噻唑-2-基)(甲基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(9B),黄色固体(0.52g,收率100%)。
LCMS m/z=584.2[M+1] +
第三步:
2-((2-乙基-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(9C)
2-((2-ethyl-6-fluoro-5-(piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl-2,3,5,6-d4)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000079
将化合物(3-((5-氰基-4-(4-氟苯基-2,3,5,6-d4)噻唑-2-基)(甲基)氨基)-2-乙基-6-氟吡唑并 [1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(9B)(0.52g,0.89mmol)溶于二氯甲烷(10mL)中,向体系滴加三氟乙酸(3mL),反应约2小时后,减压浓缩得到目标化合物9C的三氟乙酸盐(0.58g,棕色粘稠物),粗产品直接进行下一步反应。
LCMS m/z=484.2[M+1] +
第四步:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(化合物9)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl)amino)-4-(4-fluorophenyl-2,3,5,6-d4)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000080
将化合物9C的三氟乙酸盐(0.58g)溶于乙腈(15mL)中,向体系依次加入碳酸钾(0.41g,3mmol)、2-氯-1-(3羟基氮杂环丁烷-1-基)乙酮(1M)(0.22g,1.5mmol),升温至80℃下反应3小时。加入水(50mL)淬灭反应,乙酸乙酯萃取(50mL×3),合并有机相,饱和食盐水洗(50mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离(二氯甲烷/甲醇=20:1)得到标题化合物2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基)氨基)-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(化合物9)(0.39g,两步73%)。
1H NMR(400MHz,CDCl 3)δ8.27(d,1H),6.45(d,1H),4.67(s,1H),4.50–4.41(m,1H),4.32-4.24(m,1H),4.15–4.08(m,1H),3.94-3.86(m,1H),3.58(s,3H),3.22(s,4H),3.17–3.10(m,2H),2.78(s,4H),2.72(q,2H),2.65(s,1H),1.32(t,3H)。
LCMS m/z=597.3[M+1] +
实施例10:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(化合物10)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl-2,3,5,6-d4)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000081
第一步:
叔丁基4-(3-((5-氰基-4-(4-氟苯基-2,3,5,6-d4)噻唑-2-基)(甲基-d3)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(10A)
tert-butyl 4-(3-((5-cyano-4-(4-fluorophenyl-2,3,5,6-d4)thiazol-2-yl)(methyl-d3)amino)-2-ethyl-6-fluoropyrazolo[1,5-a]pyridin-5-yl)piperazine-1-carboxylate
Figure PCTCN2021128735-appb-000082
将叔丁基4-(3-((5-氰基-4-(4-氟苯基-2,3,5,6-d4)噻唑-2-基)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(9A)(0.51g,0.89mmol)溶于N,N-二甲基甲酰胺(10mL)中,加入碳酸钾(0.27g,2mmo),向体系滴加碘甲烷-d3(0.19g,1.34mmol)。加毕,室温反应30分钟。加入水(50mL)淬灭反应,乙酸乙酯萃取(50mL×2),合并有机相,饱和食盐水洗(100mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩得到目标化合物叔丁基4-(3-((5-氰基-4-(4-氟苯基-2,3,5,6-d4)噻唑-2-基)(甲基-d3)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(10A),黄色固体(0.51g,收率98%)。
LCMS m/z=587.2[M+1] +
第二步:
2-((2-乙基-6-氟-5-(哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基 -2,3,5,6-d4)噻唑-5-甲腈(10B)
2-((2-ethyl-6-fluoro-5-(piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl-2,3,5,6-d4)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000083
叔丁基4-(3-((5-氰基-4-(4-氟苯基-2,3,5,6-d4)噻唑-2-基)(甲基-d3)氨基)-2-乙基-6-氟吡唑并[1,5-a]吡啶-5-基)哌嗪-1-羧酸酯(10A)(0.51g,0.88mmol)溶于二氯甲烷(10mL)中,向体系滴加三氟乙酸(3mL),反应约2小时后,减压浓缩得到目标化合物10B的三氟乙酸盐(0.62g,棕色粘稠物),粗产品直接进行下一步反应。
LCMS m/z=487.3[M+1] +
第三步:
2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(化合物10)
2-((2-ethyl-6-fluoro-5-(4-(2-(3-hydroxyazetidin-1-yl)-2-oxoethyl)piperazin-1-yl)pyrazolo[1,5-a]pyridin-3-yl)(methyl-d3)amino)-4-(4-fluorophenyl-2,3,5,6-d4)thiazole-5-carbonitrile
Figure PCTCN2021128735-appb-000084
将化合物10B的三氟乙酸盐(0.62g)溶于乙腈(15mL)中,向体系依次加入碳酸钾(0.46g,3.3mmol)、2-氯-1-(3羟基氮杂环丁烷-1-基)乙酮(1M)(0.22g,1.5mmol),升温至80℃下反应4小时。加入水(60mL)淬灭反应,乙酸乙酯萃取(60mL×3),合并有机相,饱和食盐水洗(60mL×1),无水硫酸钠干燥,过滤,滤液减压浓缩,残留物经硅胶柱层析分离(二氯甲烷/甲醇(v/v)=20:1)得到标题化合物2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基-2,3,5,6-d4)噻唑-5-甲腈(化合物10)(0.35g,两步66%)。
1H NMR(400MHz,CDCl 3)δ8.27(d,1H),6.44(d,1H),4.66(s,1H),4.50–4.40(m,1H),4.30–4.22(m,1H),4.15–4.05(m,1H),3.93–3.85(m,1H),3.21(s,4H),3.18–3.04(m,2H),2.90(s,1H),2.77–2.66(m,6H),1.32(t,3H)。
LCMS m/z=600.3[M+1] +
实施例11:化合物A晶型I的制备
将实施例3制备的2-((2-乙基-6-氟-5-(4-(2-(3-羟基氮杂环丁烷-1-基)-2-氧代乙基)哌嗪-1-基)吡唑并[1,5-a]吡啶-3-基)(甲基-d3)氨基)-4-(4-氟苯基)噻唑-5-甲腈(化合物A即化合物3)的游离碱(9.1g,15.3mmol)加入到乙醇(50mL)中,升温到75℃,搅拌2h后溶解,缓慢搅拌下渐冷却至室温,析出大量白色固体。抽滤,滤饼用乙醇(10mL)洗涤后40℃真空干燥24h得到化合物A的晶型I,白色固体(6.2g,收率68%)。其XRPD图如图1所示,XRPD衍射峰出峰列表列于表1。TGA图列于图2,DSC图列于图3,结果显示样品加热至150℃有1.3%的失重,并在178.7℃(起始温度)有1个吸热峰。动态水分吸附仪(DVS)图列于图4,在25℃/80%RH是水分吸附为0.20%。PLM图列于图5,结果显示结晶粒径小于50μm。
表1 化合物A晶型I的XRPD衍射峰列表
Figure PCTCN2021128735-appb-000085
Figure PCTCN2021128735-appb-000086
测试例
1.溶解性试验
对化合物A晶型I(实施例11)在水和三种生物溶媒中的动态溶解度进行了评估。以5~10mg/mL的投料浓度在37℃条件下利用旋转混合的方式(25rpm)测定样品在水、SGF、FaSSIF和FeSSIF四种溶剂体系的动态溶解度(1、2、4和24小时)。每个时间点的样品经离心(12000rpm,5min)过滤(0.45μm PTFE滤头),测定滤液的HPLC浓度和pH值。溶解度试验结果总结于表2。
表2 动态溶解度测试结果总结
Figure PCTCN2021128735-appb-000087
S:溶解度(mg/mL);
LOQ:0.03μg/mL
H 2O、SGF、FaSSIF和FeSSIF的起始pH分别为6.9、1.8、6.5和5.0。
生物溶媒的配制
模拟胃液的配制(SGF):称取0.1g NaCl和0.05g曲纳通X-100至50mL容量瓶中,加入纯化水溶清。加入67.5μL浓盐酸(12M),用1M的盐酸或1M的NaOH溶液调节pH至1.8。加纯化水定容。
模拟禁食状态肠液的配制(FaSSIF):称取0.17g无水NaH 2PO 4、0.021g NaOH和0.31g NaCl至50mL容量瓶中。加入约48mL纯化水溶清,用1M的盐酸或1M的NaOH溶液调节pH至6.5。加纯化水定容,并称入0.11g SIF粉末溶清。
模拟喂食状态肠液的配制(FeSSIF):取0.41mL冰醋酸、0.20g NaOH和0.59g NaCl至 50mL容量瓶中。加入约48mL纯化水溶清,用1M的盐酸或1M的NaOH溶液调节pH至5.0。加纯化水定容,并称入0.56g SIF粉末溶清。
2.稳定性试验
将晶型I(实施例11)在25℃/60%RH和40℃/75%RH条件下放置12天后,检测样品纯度并观察晶型转变情况,结果见表3。
表3 晶型稳定性评估
Figure PCTCN2021128735-appb-000088
结论:本发明化合物A的晶型I具有较好的稳定性。
生物实验
1、生物体外实验
自分泌运动因子是一种血浆磷酸二酯酶,其将溶血磷脂酰胆碱(LPC)转化为溶血磷脂酸(LPA),因此使用LPA形成来评价自分泌运动因子抑制剂的效力。在混合的离体人血浆中评价了化合物的效力。
采肝素抗凝全血离心后收集血浆。95-μL血浆中加入5-μL梯度稀释的待测化合物或DMSO,37℃孵育2h后加入终止液(40mM磷酸氢二钠buffer containing 30mM柠檬酸,pH=4)。孵育前后血浆中LPA用LC-MSMS检测。为了确定研究血浆中的LPA18:2或20:4浓度,通过在丁醇中进行系列稀释制备LPA 18:2或20:4的校准标准品:20000、10000、5000、2000、1000、500、200、100、50、20和10ng/mL。向1.5mL微量离心管中的27.0μL空白血浆中加入3μL校准标准溶液,以生成1X校准标准品。向1.5mL微量离心管中添加30.0μL标准品或研究血浆。向每个装有研究血浆或校准标准品的1.5mL微量离心管中加入200μL丁醇(含有25.0ng/mLLPA17:0用于内部控制)。涡旋振荡1分钟并以10000rpm离心10min后,将180μL上清液转移到96孔板中,并使用LC/MS/MS与标准品一起对血浆中的LPA18:2浓度进行定量。简言之,注入8μL溶液以供使用ACQUITY UPLC BEH C18柱(2.1x50mm,1.7μm)进行LC-MS/MS分析,其中使用流动相A[20mM NH 4OAC水溶液(0.1%FA)]和流动相B[5mM NH 4OAC水溶液/ACN中的0.2%FA=5:95]。针对LPA18:2的质谱仪参数优化通过去质子化分子离子进行,对于LPA18:2在m/z 433.2([M-H]-)处,对于LPA20:4在457.2处,对于LPA18:2和LPA20:4均获得了在m/z 152.8处的丰富产物离子。以多重反应监测(MRM)负电喷射电离模式获取定量数据。
通过比较孵育和未孵育的血浆中LPA的水平改变来确定不同浓度化合物对LPA生成的抑制率,计算化合物IC 50为计算抑制率,各孔的相对浓度=各孔的浓度-基线的平均浓度。
抑制率%=(对照孔的平均相对浓度-待测孔的平均相对浓度)/对照孔的平均相对浓度×100%,曲线绘制为抑制率(y轴)与化合物浓度(x轴),并通过GraphPad Prism7.0用对数(抑制剂)与归一化响应(可变斜率)进行拟合。
表4 测试化合物抑制LPA形成的IC 50(nM)
实施例 LPA formation(18:2) LPA formation(20:4)
1 46.05 48.51
2 51.96 55.27
3 42.66 44.47
4 51.79 54.81
2、药代动力学测试
2.1大鼠药代动力学测试
实验目的:通过单剂量静脉和灌胃给予受试物于SD大鼠,测定大鼠血浆中受试物的浓度,评价受试物在大鼠体内药代特征和生物利用度。
实验对象:本发明实施例3化合物。
实验动物:雄性SD大鼠,220g左右,6~8周龄,6只/化合物。购于成都达硕实验动物有限公司。
实验方法:试验当天,6只SD大鼠按体重随机分组。给药前1天禁食不禁水12~14h,给药后4h给食。按照表5给药。
表5 给药信息
Figure PCTCN2021128735-appb-000089
*剂量以游离碱计。
2.2生物样品采集
于给药前及给药后异氟烷麻醉经眼眶取血0.1mL,置于EDTAK2离心管中。5000rpm,4℃离心10min,收集血浆。
G1组样品采集时间点:0,5,15,30min,1,2,4,6,8,24h。
G2组样品采集时间点:0,15,30min,1,2,4,6,8,24h。
分析检测前,所有样品存于-80℃。
2.3样品前处理
取30μL血浆样品、标曲和质控样品,加入200μL的含内标乙腈溶液,涡旋混匀之后,于4℃,12000rpm离心10min。取170μL上清液于96孔板中,LC-MS/MS分析,进样量为2μL。
主要药代动力学参数用WinNonlin 8.0软件非房室模型分析。实验结果如表6所示。
表6 大鼠药代动力学
Figure PCTCN2021128735-appb-000090
结论:本发明化合物,特别是化合物2、9、10,在大鼠中具有良好的药代动力学,生物利用度高,化合物5、6、7起效快。
2.2小鼠药代动力学测试
试验目的:评价受试物在小鼠体内药代特征和生物利用度。
试验动物:C57小鼠,~25g,雄性,6~8周龄,36只,购于成都达硕实验动物有限公司,生产许可证号:SCXK(川)2020-030。
试验对象:化合物I-1(专利WO2019228403A1化合物101)和化合物3。
试验设计:
表7 给药信息
Figure PCTCN2021128735-appb-000091
Figure PCTCN2021128735-appb-000092
静脉给药溶媒:5%DMA+5%Solutol+90%Saline;灌胃给药溶媒:0.5%MC
于给药前及给药后异氟烷麻醉经眼眶取血0.06mL,置于EDTAK2离心管中,5000rpm,4℃离心10min,收集血浆。采血时间点:0,5,15,30min,1,2,4,6,8,10,24h。分析检测前,所有血浆样品存于-80℃。实验结果见表8。
表8 小鼠药代动力学
Figure PCTCN2021128735-appb-000093
结论:本发明化合物,特别是化合物3,在小鼠中具有良好的药代动力学,生物利用度高。
2.3犬药代动力学测试
试验目的:评价受试物在犬体内药代特征和生物利用度。
试验动物:Beagle犬,8~10kg,雄性,0.5~1.5周岁,12只,购于北京玛斯生物技术有限公司,生产许可证号:SCXK(京)2016-001。
试验对象:化合物I-1(专利WO2019228403A1化合物101)和化合物3。
试验设计:
表9 给药信息
Figure PCTCN2021128735-appb-000094
静脉给药溶媒:5%DMSO+5%Solutol+90%Saline;
灌胃给药溶媒:1%DMSO+1%Solutol+98%(0.5%MC);
于给药前及给药后经前肢静脉取血1.0mL,置于EDTAK2离心管中,5000rpm,4℃离心10min,收集血浆。采血时间点:0,5,15,30min,1,2,4,6,8,10,12,24h。分析检测前,所有血浆样品存于-80℃。实验结果见表10。
表10 犬药代动力学
Figure PCTCN2021128735-appb-000095
结论:本发明化合物,特别是化合物3,在犬中具有良好的药代动力学,生物利用度高。
2.4猴药代动力学测试
试验目的:评价受试物在猴体内药代特征和生物利用度。
试验动物:食蟹猴,2.4~5.9kg,雄性,3~5.5周岁,12只,购于苏州西山中科实验动物有限公司,生产许可证号:SCXK(苏)2018-0001。
试验对象:化合物I-1(专利WO2019228403A1化合物101)和化合物3。
试验设计:
表11 给药信息
Figure PCTCN2021128735-appb-000096
静脉给药溶媒:5%DMSO+5%Solutol+90%Saline;
灌胃给药溶媒:1%DMSO+1%Solutol+98%(0.5%CMC-Na)
于给药前及给药后经前肢静脉取血1.0mL,置于EDTAK2离心管中,5000rpm,4℃离心10min,收集血浆。采血时间点:0,5,15,30min,1,2,4,6,8,10,12,24h。分析检测前,所有血浆样品存于-80℃。实验结果见表12。
表12 猴药代动力学
Figure PCTCN2021128735-appb-000097
结论:本发明化合物,特别是化合物3,在猴中具有良好的药代动力学,生物利用度高。
2.5hERG钾通道作用测试
测试化合物:化合物I-1(专利WO2019228403A1化合物101)及实施例化合物3。
使用电生理手动膜片钳方法测试实施例化合物对hERG钾通道(human Ether-à-go-go Related Gene potassium channel)电流的影响。
表13 实验材料及仪器
实验材料 提供商(货号)
6cm细胞培养皿 上海华雅思创生物科技有限公司(150288)
3.5cm细胞培养皿 上海华雅思创生物科技有限公司(153066)
透析胎牛血清 上海博晗生物科技有限公司(BS-0005-500)
DMSO 德国默克集团(102952)
DMEM培养基 赛默飞世尔科技(中国)有限公司(10569)
HEPES 赛默飞世尔科技(中国)有限公司(15630080)
胰蛋白酶 赛默飞世尔科技(中国)有限公司(12604)
磷酸盐缓冲液(不含钙镁离子) 赛默飞世尔科技(中国)有限公司(14190)
青霉素-链霉素溶液 赛默飞世尔科技(中国)有限公司(15140-122)
MEM非必需氨基酸溶液 赛默飞世尔科技(中国)有限公司(11140)
遗传霉素(G418) 赛默飞世尔科技(中国)有限公司(11811031)
杀稻瘟菌素 赛默飞世尔科技(中国)有限公司(R21001)
多聚赖氨酸 赛默飞世尔科技(中国)有限公司(P4832)
多菲莱德 北京伊普瑞斯技术开发有限公司(D525700)
强力霉素 西格玛奥德里奇(上海)贸易有限公(D9891)
实验仪器 提供商(型号)
二氧化碳培养箱 赛默飞世尔科技(中国)有限公司(371)
拉制仪 美国Sutter公司(P-97)
微操纵器 美国Siskiyou公司(MC1000e)
  美国Sutter公司(ROE-200;MP285)
放大器 德国HEKA公司(EPC10)
放大器 美国AXON公司(Multiclamp 700B)
显微镜 奥林巴斯中国有限公司(IX51/71/73)
灌流系统 美国ALA公司(VM8型重力给药系统)
细胞株及细胞培养:
稳定表达hERG离子通道的HEK293细胞株购自Invitrogen公司。该细胞株被培养在含有85%DMEM,10%透析胎牛血清,0.1mM MEM非必需氨基酸溶液,100U/mL青霉素-链霉素溶液,25mM HEPES,5μg/mL杀稻瘟菌素和400μg/mL遗传霉素(G418)的培养基中。待细胞密度增长至培养皿底面积的40%~80%时,通过胰蛋白酶进行消化,每周传代三次。在实验前,细胞按照5×10 5的密度培养在6cm培养皿中,加入1μg/mL强力霉素诱导48小时,然后将细胞进行消化,接种在玻片上以备后续的手动膜片钳的实验。
溶液配制:
细胞外液(以mM为单位):132氯化钠,4氯化钾,3氯化钙,0.5氯化镁,11.1葡萄糖,10HEPES(采用氢氧化钠将pH调至7.35)。
待测化合物溶液配制:
测试化合物首先用DMSO溶解并配制成终浓度为30mM的储备液。原始储备液随后以一定的比例用DMSO稀释成其他4个梯度系列溶液,浓度分别为:10,3.33,1.11和0.37mM。实验开始前,用细胞外液将待测化合物梯度系列溶液再次按1:1000的比例稀释成一系列梯度浓度的工作溶液,其终浓度分别为:30,10,3.33,1.11和0.37μM。5个不同梯度浓度的工作溶液被用于测定化合物对hERG钾离子通道的潜在抑制作用并用以拟合量效曲线和计算IC 50
实验步骤:
1.将培养皿中载有HEK293细胞的小玻片放置于显微操作台的灌流槽中。
2.在Olympus IX51、IX71或IX73倒置显微镜下将合适的细胞调置于视野中央,使用×10倍物镜找到玻璃电极的尖端,并置于视野的中央。然后使用微操纵器下移电极,同时调整粗准焦螺旋,使电极慢慢接近细胞。
3.当快接近细胞时,转换为×40倍物镜进行观察,通过微操纵器微调档,使电极逐渐接近细胞的表面。
4.给予负压,使电极尖与细胞膜之间形成电阻高于1GΩ的封接。
5.在电压钳模式下对瞬时电容电流C fast进行补偿。然后重复给予短促的负压进行破膜,最终形成全细胞记录模式。
6.在膜电位钳制于-60mV的条件下,对缓慢电容电流C slow,细胞膜电容(Cm)和输入膜电阻(Ra)分别进行补偿。
7.细胞稳定后,将钳制电压改为-90mV,采样频率设置为20kHz,过滤频率为10kHz。漏电流的检测条件为钳制电压转为-80mV,时程500ms。
8.hERG电流测试方法如下:施加4.8秒去极化命令电压将膜电位从-80mV去极化至+30mV,然后瞬间施加5.2秒的复极化电压使膜电位降至-50mV以去除通道失活,从而得以观 察到hERG尾电流。尾电流的峰值为hERG电流的大小。
9.用于检测待测化合物的hERG电流在给药前均被持续记录120秒以评估受试细胞产生hERG电流的稳定性。只有在评价标准接受范围以内的稳定细胞才能进入后续化合物检测。
10.待测化合物对hERG电流抑制作用的测试:首先将在含0.1%DMSO的细胞外液中测定得到的hERG电流作为检测基线。在hERG电流保持稳定至少5分钟后将含有待测化合物的溶液从低浓度到高浓度依次灌注于细胞周围。每次灌流结束后等待约5分钟以使化合物充分作用于细胞并同步记录hERG电流。待记录电流趋于稳定后记录最后5个hERG电流值,并取其平均值作为其最终在特定浓度下的电流值。在测试完化合物后,加入150nM多菲莱德至同一个细胞上,将其电流完全抑制,作为该细胞的阳性对照。同时,阳性化合物多菲莱德在测试药实验结束前后用同一膜片钳系统进行同步检测,以确保整个检测系统的可靠性和灵敏性。
数据分析:电流抑制百分率通过以下公式进行计算,数据由PatchMaster或Clampex10.2软件输出,量效曲线通过Graphpad Prism 8.0软件进行拟合并计算IC 50值。实验结果见表11。
Figure PCTCN2021128735-appb-000098
表14 测试结果
序号 测试样品 hERG IC 50[μM]
1 化合物I-1 9.368
2 Dofetilide 0.016
3 化合物3 23.113
评价化合物是否对hERG钾离子通道具有抑制作用,被广泛认同和使用的判断标准如下:抑制作用不显著:IC 50>10μM;中等抑制:1μM<IC 50<10μM;显著抑制作用:IC 50<1μM。从测试结果看,化合物3对hERG钾离子通道抑制作用不显著。

Claims (18)

  1. 一种式(I)所示的化合物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
    Figure PCTCN2021128735-appb-100001
    R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 16、R 17、R 18、R 19、R 20、R 21、R 22、R 23、R 24、R 25、R 26、R 27、R 28、R 29和R 30各自独立地选自氢或氘;
    条件是,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 16、R 17、R 18、R 19、R 20、R 21、R 22、R 23、R 24、R 25、R 26、R 27、R 28、R 29和R 30中至少一个选自氘原子且式(I)化合物不为如下结构:
    Figure PCTCN2021128735-appb-100002
  2. 根据权利要求1所述的式(I)化合物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中
    R 22选自H。
  3. 根据权利要求1-2任意一项所述的式(I)化合物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中,
    R 1选自氘;或者
    R 2和R 3选自氘;或者
    R 6和R 7选自氘;或者
    R 8、R 9、R 10、R 11、R 12、R 13、R 14和R 15选自氘;或者
    R 16选自氘;或者
    R 17选自氘;或者
    R 18、R 19、R 20和R 21选自氘;或者
    R 23和R 24选自氘;或者
    R 25、R 26和R 27选自氘;或者
    R 28、R 29和R 30选自氘。
  4. 根据权利要求1-2任意一项所述的式(I)化合物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中,
    R 23、R 24、R 25、R 26、R 27、R 28、R 29和R 30选自氘;或者
    R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 25、R 26和R 27选自氘;或者
    R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 23、R 24、R 25、R 26、R 27、R 28、R 29和R 30选自氘;或者
    R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 16和R 17选自氘;或者
    R 8、R 9、R 10、R 11、R 12、R 13、R 14、R 15、R 18、R 19、R 20和R 21选自氘;或者
    R 23、R 24、R 28、R 29、R 30、R 18、R 19、R 20和R 21选自氘;或者
    R 25、R 26、R 27、R 18、R 19、R 20和R 21选自氘;或者
    R 1、R 25、R 26和R 27选自氘;或者
    R 1、R 23、R 24、R 28、R 29和R 30选自氘;或者
    R 6、R 7、R 25、R 26和R 27选自氘;或者
    R 6、R 7、R 23、R 24、R 28、R 29和R 30选自氘;或者
    R 2、R 3、R 25、R 26和R 27选自氘;或者
    R 2、R 3、R 23、R 24、R 28、R 29和R 30选自氘。
  5. 根据权利要求1所述的式(I)的化合物,其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中所述化合物选自以下结构之一:
    Figure PCTCN2021128735-appb-100003
    Figure PCTCN2021128735-appb-100004
    Figure PCTCN2021128735-appb-100005
  6. 式(A)化合物的晶型I,
    Figure PCTCN2021128735-appb-100006
    其特征在于,使用Cu-Kα辐射,其X-射线粉末衍射图谱在以下2θ位置具有特征衍射峰:9.35±0.2°、10.32±0.2°、12.08±0.2°和15.10±0.2°。
  7. 根据权利要求6所述的式(A)化合物的晶型I,其X-射线粉末衍射图谱还在以下2θ位置具有特征衍射峰:17.29±0.2°、18.26±0.2°和20.58±0.2°。
  8. 根据权利要求7所述的式(A)化合物的晶型I,其X-射线粉末衍射图谱还在以下2θ位置具有特征衍射峰:6.92±0.2°、7.23±0.2°、13.46±0.2°、19.47±0.2°和24.85±0.2°。
  9. 根据权利要求8所述的式(A)化合物的晶型I,其X-射线粉末衍射图谱基本如图1所示。
  10. 根据权利要求6所述的式(A)化合物的晶型I,其TGA曲线基本如图2所示,其DSC图基本如图3所示。
  11. 如下所示式(II-A)、式(II-B)、式(II-C)化合物
    Figure PCTCN2021128735-appb-100007
  12. 一种制备式(A)化合物的方法,其特征在于,包含如下步骤
    Figure PCTCN2021128735-appb-100008
    将式(II-C)化合物溶于有机溶剂A中,与2-氯-1-(3羟基氮杂环丁烷-1-基)乙酮、碱,在70~90℃下反应,得到式(A)化合物。
  13. 根据权利要求12所述制备方法,其特征在于,所述有机溶剂A选自乙腈、2-甲基四氢呋喃、乙苯、甲酰甲酸甲乙酯和二乙二醇叔丁醚中的至少一种;所述碱为碳酸钾和碳酸钠中的至少一种。
  14. 根据权利要求12所述制备方法,其特征在于,式(A)化合物的制备方法还包含如下步骤,
    Figure PCTCN2021128735-appb-100009
    i、式(II-D)化合物、2-氯-4-(4-氟苯基)噻唑-5-氰基、2,6-二甲基吡啶和非质子极性溶剂,在60~80℃下反应,得到化合物(II-A);
    ii、将步骤i得到的化合物(II-A),溶于有机溶剂B中,在氮气保护下冷却至0℃,加入还原剂,反应完成后加入碘甲烷-d 3,得到化合物(II-B);
    iii、将步骤ii得到的化合物(II-B)溶于有机溶剂C中,加入三氟乙酸,反应得到式(II-C)化合物。
  15. 根据权利要求14所述制备方法,其特征在于,所述非质子极性溶剂为N,N-二甲基乙酰胺、二甲基甲酰胺、六甲基磷酰胺、乙腈、丙酮和二甲亚砜中的至少一种;所述有机溶剂B为四氢呋喃、N,N-二甲基乙酰胺、二甲基甲酰胺、乙腈和丙酮中的至少一种;所述还原剂为氢化钠;所述有机溶剂C为二氯甲烷、乙酸甲酯、碳酸二甲酯、丙二醇甲醚乙酸酯和尼龙酸二甲酯中的至少一种。
  16. 一种药物组合物,其特征在于,含有权利要求1-5任意一项所述的化合物、其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶、或者含有权利要求6-10任意一项所述的晶型I,以及药学上可接受的载体和/或赋形剂。
  17. 权利要求1-5任意一项所述的化合物、其立体异构体、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶、或者含有权利要求6-10任意一项所述的晶型I、或者权利要求16所述的组合物在制备治疗ATX介导的疾病的药物中的应用。
  18. 根据权利要求17所述的应用,其特征在于,所述ATX介导的疾病为特发性肺纤维化。
PCT/CN2021/128735 2020-11-04 2021-11-04 Atx抑制剂氘代衍生物及其应用 WO2022095930A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2023526670A JP2023548181A (ja) 2020-11-04 2021-11-04 Atx阻害剤の重水素化誘導体及びその応用
EP21888622.4A EP4242208A1 (en) 2020-11-04 2021-11-04 Deuterated derivative as atx inhibitor, and application thereof
CN202180072781.3A CN116437917A (zh) 2020-11-04 2021-11-04 Atx抑制剂氘代衍生物及其应用

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202011213775.9 2020-11-04
CN202011213775 2020-11-04
CN202011562922 2020-12-25
CN202011562922.3 2020-12-25
CN202110085643 2021-01-22
CN202110085643.0 2021-01-22

Publications (1)

Publication Number Publication Date
WO2022095930A1 true WO2022095930A1 (zh) 2022-05-12

Family

ID=81456966

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/128735 WO2022095930A1 (zh) 2020-11-04 2021-11-04 Atx抑制剂氘代衍生物及其应用

Country Status (4)

Country Link
EP (1) EP4242208A1 (zh)
JP (1) JP2023548181A (zh)
CN (1) CN116437917A (zh)
WO (1) WO2022095930A1 (zh)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105143221A (zh) * 2013-03-14 2015-12-09 加拉帕戈斯股份有限公司 用于治疗炎性障碍的化合物及其药物组合物
CN105339370A (zh) * 2013-06-19 2016-02-17 加拉帕戈斯股份有限公司 用于治疗炎症性疾病的新化合物和及其药物组合物
CN110156772A (zh) * 2018-02-14 2019-08-23 苏州信诺维医药科技有限公司 一种杂环化合物、其应用及含其的药物组合物
WO2019228403A1 (en) 2018-05-29 2019-12-05 Fronthera U.S. Pharmaceuticals Llc Autotaxin inhibitors and uses thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106068262B (zh) * 2014-04-11 2019-10-29 海思科医药集团股份有限公司 喹唑啉衍生物及其制备方法和在医药上的应用
CN111170995A (zh) * 2018-11-09 2020-05-19 山东轩竹医药科技有限公司 Ask1抑制剂及其应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105143221A (zh) * 2013-03-14 2015-12-09 加拉帕戈斯股份有限公司 用于治疗炎性障碍的化合物及其药物组合物
CN105339370A (zh) * 2013-06-19 2016-02-17 加拉帕戈斯股份有限公司 用于治疗炎症性疾病的新化合物和及其药物组合物
CN110156772A (zh) * 2018-02-14 2019-08-23 苏州信诺维医药科技有限公司 一种杂环化合物、其应用及含其的药物组合物
WO2019228403A1 (en) 2018-05-29 2019-12-05 Fronthera U.S. Pharmaceuticals Llc Autotaxin inhibitors and uses thereof
CN111801328A (zh) * 2018-05-29 2020-10-20 弗朗塞拉美国制药有限责任公司 自分泌运动因子抑制剂及其用途

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
J. MED. CHEM., vol. 60, 2017, pages 3580 - 3590

Also Published As

Publication number Publication date
CN116437917A (zh) 2023-07-14
JP2023548181A (ja) 2023-11-15
EP4242208A1 (en) 2023-09-13

Similar Documents

Publication Publication Date Title
US11498900B2 (en) Salts of an LSD1 inhibitor
JP6059723B2 (ja) 4−(8−メトキシ−1−((1−メトキシプロパン−2−イル)−2−(テトラヒドロ−2H−ピラン−4−イル)−1H−イミダゾ[4,5−c]キノリン−7−イル)−3,5−ジメチルイソオキサゾールおよびブロモドメイン阻害剤としてのその使用
AU2014265279B2 (en) Bipyrazole derivatives as JAK inhibitors
WO2016124067A1 (zh) 一种周期素依赖性蛋白激酶抑制剂的羟乙基磺酸盐、其结晶形式及制备方法
KR20230057386A (ko) Apol1 억제제 및 사용 방법
TWI723480B (zh) 用作fgfr4抑制劑的稠環衍生物
WO2022095930A1 (zh) Atx抑制剂氘代衍生物及其应用
WO2016155573A1 (zh) 一种杂环衍生物及其制备方法和在医药上的用途
TW202313604A (zh) 吡唑並雜芳基類衍生物的可藥用鹽及其結晶形式
CN112457296B (zh) 嘧啶类化合物及其制备方法
CN112759538B (zh) 3-(二甲氨基甲基)环己-4-醇衍生物及其制备方法和药物用途
TW202214644A (zh) 含氮雜環化合物的固體形式及其藥物組合物和用途
RU2640047C2 (ru) Соль производного пирролидин-3-ил-уксусной кислоты и ее кристаллы
JP2022517396A (ja) Egfr阻害剤の塩、結晶形及びその製造方法
CN112759546A (zh) 3-(二甲氨基甲基)哌啶-4-醇衍生物及其制备方法和药物用途
WO2023025270A1 (zh) 一种吲哚类化合物的固体形式及其制备方法和用途
WO2019015640A1 (zh) 一种氮杂环酰胺衍生物的盐、其晶型及其制备方法和用途
WO2021233427A1 (zh) 并环化合物及其制备方法、药物组合物和应用
WO2023116879A1 (zh) 一种glp-1受体激动剂的结晶形式及其制备方法
EP4339194A1 (en) Acid addition salt of rock inhibitor, and crystal form, composition and pharmaceutical use thereof
CN115368354A (zh) Atx抑制剂的药用盐及其组合物和用途
WO2023116882A1 (zh) 一种glp-1受体激动剂的可药用盐、结晶形式及其制备方法
WO2022012534A1 (zh) 含氮杂环化合物、药物组合物和应用
TW202333694A (zh) 稠環衍生物的晶型、其製備方法及其應用
CN113943296A (zh) 吡咯并嘧啶衍生物及其应用

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: 21888622

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2023526670

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021888622

Country of ref document: EP

Effective date: 20230605