WO2025124502A1 - Nav1.8 inhibitor compound, salt thereof, polymorph thereof, and use thereof - Google Patents
Nav1.8 inhibitor compound, salt thereof, polymorph thereof, and use thereof Download PDFInfo
- Publication number
- WO2025124502A1 WO2025124502A1 PCT/CN2024/138893 CN2024138893W WO2025124502A1 WO 2025124502 A1 WO2025124502 A1 WO 2025124502A1 CN 2024138893 W CN2024138893 W CN 2024138893W WO 2025124502 A1 WO2025124502 A1 WO 2025124502A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- formula
- crystalline form
- solvent
- free
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D239/00—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
- C07D239/02—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
- C07D239/24—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
- C07D239/28—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
- C07D239/32—One oxygen, sulfur or nitrogen atom
- C07D239/42—One nitrogen atom
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/04—Centrally acting analgesics, e.g. opioids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07B—GENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
- C07B2200/00—Indexing scheme relating to specific properties of organic compounds
- C07B2200/13—Crystalline forms, e.g. polymorphs
Definitions
- the present invention belongs to the field of medicine and relates to a Nav1.8 inhibitor compound and its salt, polymorph, and their preparation methods and applications.
- Pain is "an unpleasant feeling and emotional sensation, accompanied by actual or potential tissue damage, and it is a subjective feeling". Pain can serve as a warning signal to alert the body to potential dangers and has an indispensable protective effect on the body's normal life activities. At the same time, pain is also a common clinical symptom. After the external stimulus that causes pain disappears, strong or persistent pain can cause physiological dysfunction and seriously affect the quality of life of the living body. According to statistics, about one-fifth of the world's people suffer from moderate to severe chronic pain. The global analgesic market was approximately US$36 billion in 2018 and is expected to reach US$56 billion in 2023.
- Chronic pain is the main driving force for the continued growth of the global pain market in the next decade.
- action potentials nerve impulses
- DRG dorsal root ganglion
- the generation and conduction of action potentials in neurons depends on voltage-gated sodium channels (NaV) on the cell membrane. When the cell membrane is depolarized, the sodium ion channels are activated, the channels open, causing sodium ions to flow in, further depolarizing the cell membrane, and leading to the generation of action potentials. Therefore, inhibiting abnormal sodium ion channel activity helps to treat and relieve pain.
- NaV voltage-gated sodium channels
- Human sodium ions are a type of transmembrane ion channel protein, consisting of an ⁇ subunit with a molecular weight of 260kD and a ⁇ subunit with a molecular weight of 30-40kD. According to the different ⁇ subunits, it can be divided into 9 subtypes, namely Nav1.1 ⁇ Nav1.9.
- Nav1.5, Nav1.8 and Nav1.9 are tetrodotoxin (TTX)-insensitive sodium channels.
- Nav1.5 is mainly present in myocardial cells
- Nav1.8 and Nav1.9 are present in the peripheral nervous system.
- Nav1.8 is an important ion channel involved in chronic pain, atrial fibrillation, and Budd-Chiari syndrome, and is a highly selective target for pain treatment.
- Nav1.8 The gene encoding Nav1.8 is SCN10A, which is located in the human chromosome 3p21-22 region and mainly encodes the ⁇ subunit. Studies have found that the homology of human and rat Nav1.8 genes is as high as 93%. Nav1.8 is mainly present in trigeminal ganglion neurons and DRG neurons, and has the electrophysiological characteristics of slow inactivation and rapid recovery. In neurons expressing Nav1.8, the rise of action potential is mainly composed of Nav1.8 current. In the model of neuropathic pain, nerve injury will increase the expression level of Nav1.8 in axons and neuronal cell bodies. The use of Nav1.8 antisense oligonucleotides can significantly relieve pain while reducing Nav1.8 expression.
- Nav1.8 knockout mice cannot show normal visceral inflammatory pain.
- human Nav1.8 gene produces a gain-of-function mutation, it will cause peripheral neuropathy.
- selective inhibition of Nav1.8 has the potential to become a new type of analgesic therapy, which can be used to treat various types of pain, such as inflammatory pain, neuralgia, postoperative pain and cancer pain.
- Nav1.8 is mainly limited to neurons that perceive pain, selective Nav1.8 blockers are unlikely to induce adverse reactions common to non-selective Nav's blockers. Therefore, the art still needs to develop new Nav1.8 selective inhibitors, preferably Nav channel inhibitors with better selectivity, more effective, increased metabolic stability, increased solubility and fewer side effects to Nav1.8.
- the compound of formula I can effectively antagonize the activity of Nav1.8 receptors and has broad application prospects in the preparation of drugs for treating diseases related to Nav1.8. Therefore, further research on the compound of formula I and its salt form and crystal form is of great significance for the development of effective therapeutic drugs.
- the present invention provides a crystalline form of a compound of formula I or a pharmaceutically acceptable salt thereof, wherein the structure of the compound of formula I is as follows:
- the present invention provides a free crystalline form A of the compound of formula I, wherein the X-ray powder diffraction spectrum of the free crystalline form A represented by a diffraction angle of 2 ⁇ 0.2° has diffraction peaks at 17.79°, 18.13°, 20.52°, 21.63°, and 25.97°; further, the X-ray powder diffraction spectrum of the free crystalline form A represented by a diffraction angle of 2 ⁇ 0.2° also has diffraction peaks at one or more of the following: 12.85°, 16.20°, 24.46°, and 25.00°; further, the X-ray powder diffraction spectrum of the free crystalline form A represented by a diffraction angle of 2 ⁇ 0.2° has diffraction peaks at 12.85°, 16.20°, 17.79°, 18.13°, 20.52°, 21.63°, and 24.46°.
- the X-ray powder diffraction spectrum of the free crystalline form A represented by a diffraction angle of 2 ⁇ 0.2° is 5.78°, 11.56°, 12.85°, 16.20°, 17.39°, 17.79°, 18.13°, 20.52°, 21.63°, 24.46°, 25.00°, 25.97°.
- the free crystalline Form A has one, two or three of the following characteristics:
- TGA curve of free form A shows a weight loss of approximately 0.38 ⁇ 1% at 150.0 ⁇ 3°C;
- the DSC graph of the free crystalline form A is shown in Figure 1-2; the TGA graph of the free crystalline form A is shown in Figure 1-3.
- the free-state crystalline form A is an anhydrous crystalline form.
- the present invention provides a free crystalline form B of the compound of formula I, wherein the free crystalline form B has an X-ray powder diffraction spectrum represented by a diffraction angle of 2 ⁇ 0.2° at 11.64°, 12.60°, 17.46°, 20.93°, 25.16°, and 26.56°; further, the free crystalline form B has an X-ray powder diffraction spectrum represented by a diffraction angle of 2 ⁇ 0.2° at 11.64°, 12.60°, 17.46°, 20.93°, 25.16°, and 26.56°.
- the X-ray powder diffraction spectrum of the free crystalline form B also has diffraction peaks at one or more of the following positions: 5.80°, 29.25°, 28.22°, 28.51°, and 35.32°; further, the X-ray powder diffraction spectrum of the free crystalline form B represented by a diffraction angle of 2 ⁇ 0.2° has diffraction peaks at 5.80°, 11.64°, 12.60°, 17.46°, 20.93°, 22.19°, 25.16 The free form B has diffraction peaks at 5.80°, 11.64°, 12.60°, 16.09°, 17.46°, 18.37°, 20.9 There are diffraction peaks at 3°, 22.19°, 23.03°, 23.82°, 25.16°, 26.56°, 28.22°, 28.51°, 29.25°, 29.79°, 32.20°, 33.23°, 34.42°, 35.32°, and 38.52°; further,
- the free crystalline Form B has one, two or three of the following characteristics:
- the TGA curve of free form B shows a weight loss of approximately 0.23 ⁇ 1% at 150.0 ⁇ 3°C;
- the DSC curve of the free form B has an endothermic peak starting point at 165.8 ⁇ 3°C;
- the DSC curve of free form B has an endothermic peak at 168.47 ⁇ 3°C.
- the DSC graph of the free crystalline form B is shown in Figure 2-2; the TGA graph of the free crystalline form B is shown in Figure 2-3.
- the free-state crystalline form B is an anhydrous crystalline form.
- the present invention provides a method for preparing the free crystalline form A of the compound of formula I, which comprises the following methods:
- Method 1 Place the first sample bottle containing the compound of formula I in an open position in a second sample bottle containing a solvent, seal the second sample bottle, and let it stand at room temperature; the solvent does not cover the mouth of the first sample bottle;
- the solvent is selected from one or more of ethanol, acetone, methyl tert-butyl ether, ethyl acetate, dichloromethane, tetrahydrofuran, acetonitrile, n-heptane, and toluene;
- Method 2 Place the first sample bottle containing the solution of the compound of formula I in the second sample bottle containing the anti-solvent, seal the second sample bottle, and let it stand at room temperature; the anti-solvent does not cover the mouth of the first sample bottle;
- the solvent in the solution of the compound of formula I is selected from one or more of dichloromethane, tetrahydrofuran, 1,4-dioxane, dimethyl sulfoxide, and N,N-dimethylformamide;
- the anti-solvent is selected from one or more of n-heptane, methyl tert-butyl ether, and water;
- Method 3 Add a solvent to the compound of formula I at room temperature, stir magnetically, and collect the solid;
- the solvent is selected from one or more of water, ethanol, isopropanol, ethyl acetate, isopropyl acetate, tetrahydrofuran, 1,4-dioxane, methyl tert-butyl ether, acetone, methyl ethyl ketone, dimethyl sulfoxide, acetonitrile, toluene, N,N-dimethylformamide, and N-methylpyrrolidone;
- Method 4 Add solvent to the compound of formula I at 50°C, stir magnetically, and collect the solid;
- the solvent is selected from one or more of water, ethanol, isopropanol, ethyl acetate, isopropyl acetate, tetrahydrofuran, 1,4-dioxane, methyl tert-butyl ether, acetone, methyl ethyl ketone, dimethyl sulfoxide, acetonitrile, toluene, N,N-dimethylformamide, and N-methylpyrrolidone;
- Method 5 Add the compound of formula I into organic solvent I, filter after dissolving, and evaporate at room temperature:
- the organic solvent I is selected from one or more of methanol, ethanol, ethyl acetate, tetrahydrofuran, 1,4-dioxane, acetone, methyl ethyl ketone, dichloromethane and acetonitrile;
- Method 6 Completely dissolve the compound of formula I in a good solvent, filter, and dropwise add an antisolvent to the clear solution until solid precipitates;
- the good solvent is selected from one or more of dimethyl sulfoxide, N,N-dimethylformamide, dichloromethane, tetrahydrofuran, methyl ethyl ketone, N-methylpyrrolidone, N,N-dimethylacetamide, ethanol, and isopropyl acetate;
- the anti-solvent is selected from one or more of water, toluene, methyl tert-butyl ether, and n-heptane;
- the good solvent when the anti-solvent is selected from water, the good solvent is selected from one of dimethyl sulfoxide, N,N-dimethylformamide, tetrahydrofuran, N-methylpyrrolidone, and N,N-dimethylacetamide; when the anti-solvent is selected from methyl tert-butyl ether, the good solvent is selected from dichloromethane; when the anti-solvent is selected from n-heptane, the good solvent is selected from methyl ethyl ketone or ethanol.
- Method 7 Dissolve the compound of formula I completely in a good solvent, filter, and add the clear solution to an anti-solvent;
- the good solvent is selected from one or more of dimethyl sulfoxide, N,N-dimethylformamide, dichloromethane, ethyl acetate, 1,4-dioxane, methyl ethyl ketone, and tetrahydrofuran;
- the anti-solvent is selected from one or more of water, n-heptane, methyl tert-butyl ether, and toluene;
- the good solvent when the anti-solvent is selected from water, the good solvent is selected from dimethyl sulfoxide or N,N-dimethylformamide; when the anti-solvent is selected from methyl tert-butyl ether, the good solvent is selected from 1,4-dioxane or methyl ethyl ketone; when the anti-solvent is selected from n-heptane, the good solvent is selected from ethyl acetate; when the anti-solvent is selected from toluene, the good solvent is selected from methyl ethyl ketone or tetrahydrofuran.
- Method 8 Add the compound of formula I into organic solvent I at 50°C, filter after dissolving, stir magnetically, and cool at room temperature;
- the organic solvent I is selected from one or more of methanol, ethanol, isopropyl acetate, acetonitrile, ethyl acetate, and acetone;
- Method 9 Add the compound of formula I into a mortar or add the compound of formula I and a solvent into a mortar and grind;
- the solvent is selected from one or more of water, methyl tert-butyl ether and n-heptane.
- the present invention provides a method for preparing the free crystalline form B of the compound of formula I, which comprises the following methods:
- Method 1 Dissolve the compound of formula I completely in a good solvent, filter, and add an antisolvent dropwise to the clear solution until solid precipitates;
- the good solvent is selected from one or two of tetrahydrofuran and N,N-dimethylacetamide, and the anti-solvent is selected from n-heptane.
- the anti-solvent is selected from one or more of water, toluene, methyl tert-butyl ether and n-heptane.
- Method 2 completely dissolve the compound of formula I in a good solvent, filter, and add the clear solution into an anti-solvent;
- the good solvent is selected from one or two of dichloromethane and 1,4-dioxane; the anti-solvent is selected from n-heptane.
- Method 3 Add the compound of formula I into isopropanol at 40-60° C. (eg 50° C.), dissolve, filter, stir, and cool at room temperature.
- Method 4 Completely dissolve the compound of formula I in tetrahydrofuran, filter, add n-heptane to the clear solution with stirring until solid precipitates, then add free form B seed crystals and stir.
- the free-state crystal form B seed can be prepared by one of the methods one, two, three, and four.
- the present invention provides a pharmaceutically acceptable salt of the compound of formula I, wherein the pharmaceutically acceptable salt is selected from the salts formed by the compound of formula I and an acid or base.
- the pharmaceutically acceptable salt of the compound of formula I is selected from the salts formed by the compound of formula I and the following acids or bases: hydrochloric acid, sulfuric acid, maleic acid, phosphoric acid, fumaric acid, tartaric acid, citric acid, L-malic acid, succinic acid, p-toluenesulfonic acid, methanesulfonic acid, sodium hydroxide, and arginine.
- the molar ratio of the acid or the base to the compound of formula I can be 5:1 to 1:5, for example, 5:1, 4.5:1, 4:1, 3.5:1, 3:1, 2.5:1, 2:1, 1.5:1, 1:1, 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5, 1:5.
- the pharmaceutically acceptable salt of the compound of formula I is selected from the maleate salt of the compound of formula I; in some embodiments, in the maleate salt, the molar ratio of the compound of formula I to maleic acid is 1:1.
- the pharmaceutically acceptable salt of the compound of formula I is selected from the sodium salt of the compound of formula I; in some embodiments, in the sodium salt, the molar ratio of the compound of formula I to sodium is 1:1.
- the present invention provides a crystalline form of a pharmaceutically acceptable salt of the compound of formula I.
- the present invention provides a maleate salt form A of a compound of formula I, wherein the maleate salt form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2 ⁇ 0.2° and has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, and 18.44°; further, the maleate salt form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2 ⁇ 0.2° and has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, 18.44°, 20.96°, 25.62°, and 26.78°; further, the maleate salt form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2 ⁇ 0.2° and has diffraction peaks at 2 ⁇ 0.2°.
- the figure has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, 18.44°, 20.96°, 21.61°, 25.29°, 25.62°, and 26.78°; further, the X-ray powder diffraction spectrum of the maleate salt form A represented by a diffraction angle of 2 ⁇ 0.2° has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, 18.44°, 20.45°, 20.96°, 21.61°, 25.29°, 25.62°, 26.19°, and 26.78°; further, the X-ray powder diffraction spectrum of the maleate salt form A represented by a diffraction angle of 2 ⁇ 0.2° has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, 18.44°, 20.45°, 20.96°, 21.61°, 25.29°, 25.62°, 26.19°, and 26.78°; further,
- the maleate salt form A has one or two of the following characteristics:
- the TGA curve of the maleate salt form A shows a weight loss of 0.5-3% at 120 ⁇ 3°C, preferably 0.5, 1.0, 1.5, 2.0, 2.5, 3.0%, for example 1.41%;
- the maleate salt form A has an endothermic peak at 139.1 ⁇ 3°C.
- the TGA/DSC graph of the maleate salt form A is substantially as shown in FIG. 3-2 ; the 1 H NMR graph of the maleate salt form A is substantially as shown in FIG. 3-3 .
- the present invention provides a sodium salt crystalline form A of the compound of formula I, wherein the sodium salt crystalline form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2 ⁇ 0.2° at 16.70°, 17.02°, 21.23°, 22.33°, 24.39°, 25.50°, and 25.89° having diffraction peaks; further, the sodium salt crystalline form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2 ⁇ 0.2° at 16.70°, 17.02°, 17.67°, 18.51°, 21.23°, 22.33°, 24.39°, 25.50°, 25.89°, and 29.73° having diffraction peaks; further, the sodium salt crystalline form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2 ⁇ 0.2° at There are diffraction peaks at 16.70°, 17.02°, 17.67°, 18.51°, 21.23
- the sodium salt form A has an XRPD spectrum substantially as shown in Figure 4-1.
- the sodium salt form A has one or two of the following characteristics:
- the TGA curve of the sodium salt form A shows a weight loss of 1.0-5.0% at 150 ⁇ 3°C, preferably 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0%, for example 2.75%;
- the sodium salt form A has two endothermic peaks at 191.0 ⁇ 3°C and 215.0 ⁇ 3°C.
- the TGA/DSC graph of the sodium salt crystalline form A is substantially as shown in FIG. 4-2 ; the 1 H NMR graph of the sodium salt crystalline form A is substantially as shown in FIG. 4-3 .
- the present invention provides a method for preparing a pharmaceutically acceptable salt (including a crystalline form of the salt) of the compound of formula I, comprising mixing the compound of formula I with a suitable acid or base to obtain the pharmaceutically acceptable salt.
- the preparation method comprises the following steps: mixing the compound of formula I with an equimolar amount of maleic acid or sodium hydroxide, stirring in an organic solvent A, and separating to obtain a solid.
- the stirring time is 1-5 days, for example, 3 days, and the stirring temperature is room temperature;
- the organic solvent A is selected from one or more of isopropanol, ethyl acetate, and 2-methyltetrahydrofuran.
- the compound of formula I is mixed with maleic acid or sodium hydroxide, a suspension is obtained in an organic solvent A, and the suspension is stirred;
- the preparation method comprises the following steps: mixing the compound of formula I with an equimolar amount of maleic acid, obtaining a suspension in an organic solvent A, suspending and stirring, and separating to obtain a solid.
- the suspension and stirring time is 1-5 days, for example, 3 days, and the suspension and stirring temperature is room temperature; the organic solvent A is selected from ethyl acetate.
- the preparation method comprises the following steps: mixing the compound of formula I with an equimolar amount of sodium hydroxide, obtaining a suspension in an organic solvent A, suspending and stirring, and separating to obtain a solid.
- the suspension and stirring time is 1-5 days, for example, 3 days, and the suspension and stirring temperature is room temperature;
- the organic solvent A is selected from one or more of isopropanol, ethyl acetate, and 2-methyltetrahydrofuran.
- the present invention provides a pharmaceutical composition
- a pharmaceutical composition comprising one or more of the free crystalline forms of the compound of formula I as described above (e.g., free crystalline form A, free crystalline form B), and the pharmaceutically acceptable salts of the compound of formula I as described above (including their crystalline forms).
- the pharmaceutical composition further comprises a pharmaceutically acceptable excipient or carrier.
- the present invention provides a pharmaceutical composition, comprising substance A and a pharmaceutically acceptable carrier, wherein substance A is a free crystalline form of a compound of formula I or a crystalline form of a pharmaceutically acceptable salt of a compound of formula I as described in any one of the above items; preferably, substance A is free crystalline form A of a compound of formula I, free crystalline form B of a compound of formula I, maleate crystalline form A, or sodium salt crystalline form A.
- the present invention provides the use of the free crystalline form of the compound of formula I (e.g., free crystalline form A, free crystalline form B), a pharmaceutically acceptable salt of the compound of formula I (including its crystalline form) or the pharmaceutical composition in the preparation of a drug for treating and/or preventing voltage-gated sodium channel-related diseases.
- free crystalline form of the compound of formula I e.g., free crystalline form A, free crystalline form B
- a pharmaceutically acceptable salt of the compound of formula I including its crystalline form
- the pharmaceutical composition in the preparation of a drug for treating and/or preventing voltage-gated sodium channel-related diseases.
- the voltage-gated sodium ion channel-related disease is a Nav1.8-related disease.
- the free crystalline form of the compound of formula I e.g., free crystalline form A, free crystalline form B
- a pharmaceutically acceptable salt of the compound of formula I including its crystalline form
- the pharmaceutical composition described in the present invention can provide patients in need with better and more effective clinical treatment drugs or regimens.
- the present invention also provides a method for treating and/or preventing diseases related to Nav1.8, which comprises administering to a patient a therapeutically effective dose of a crystalline form of the compound of formula I as described above, a pharmaceutically acceptable salt of the compound of formula I as described above, a crystalline form of the salt as described above, or a pharmaceutical composition as described above; preferably, a pharmaceutical preparation comprising a free crystalline form of the compound of formula I as described above (e.g., free crystalline form A, free acid crystalline form B), a pharmaceutically acceptable salt of the compound of formula I (including its crystalline form, such as maleate crystalline form A, sodium salt crystalline form A) or the pharmaceutical composition.
- a pharmaceutical preparation comprising a free crystalline form of the compound of formula I as described above (e.g., free crystalline form A, free acid crystalline form B), a pharmaceutically acceptable salt of the compound of formula I (including its crystalline form, such as maleate crystalline form A, sodium salt crystalline form A) or the pharmaceutical composition.
- the Nav1.8-related diseases include: pain.
- the pain includes: acute pain, chronic pain, inflammatory pain, cancer pain, neuropathic pain, musculoskeletal pain, primary pain, intestinal pain and idiopathic pain.
- the numerical ranges recorded in this specification and claims are equivalent to recording at least each specific integer value therein.
- two or more represent 2, 3, 4, 5, 6, 7, 8, 9, 10 or more.
- numbers When certain numerical ranges are defined or understood as “numbers", they should be understood to record the two endpoints of the range, each integer within the range, and each decimal within the range.
- "a number from 0 to 10” should be understood to record not only each integer of 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 and 10, but also at least the sum of each integer therein and 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, respectively.
- about 10 represents: values within the range of 10 ⁇ 1.5, that is, values within the range of 8.5 to 11.5; values within the range of 10 ⁇ 1.0, that is, values within the range of 9.0 to 11.0; and values within the range of 10 ⁇ 0.5, that is, values within the range of 9.5 to 10.5.
- the "2 ⁇ or 2 ⁇ angle" mentioned in the present disclosure refers to the diffraction angle, ⁇ is the Bragg angle, and the unit is ° or degree; the error range of each characteristic peak 2 ⁇ is ⁇ 0.20 (preferably ⁇ 0.10) (including the case where the numbers exceeding 2 decimal places are rounded off).
- the salts and polymorphs of the compound of formula I of the present invention may be used in combination with other active ingredients as long as it does not cause other adverse effects such as allergic reactions.
- composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
- patient refers to any animal including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, pigs, cows, sheep, horses or primates, and most preferably humans.
- terapéuticaally effective dose refers to the amount of an active compound or drug that elicits the biological or medical response that a researcher, veterinarian, physician or other clinician is seeking in a tissue, system, animal, individual or human, and includes one or more of the following: (1) preventing disease: for example, preventing a disease, disorder or condition in an individual who is susceptible to the disease, disorder or condition but has not yet experienced or developed the pathology or symptoms of the disease; (2) inhibiting disease: for example, inhibiting the disease, disorder or condition in an individual who is experiencing or developing the pathology or symptoms of the disease, disorder or condition (i.e., preventing further development of the pathology and/or symptoms); (3) alleviating disease: for example, alleviating the disease, disorder or condition in an individual who is experiencing or developing the pathology or symptoms of the disease, disorder or condition (i.e., reversing the pathology and/or symptoms).
- pharmaceutically acceptable means that the formulation components or active ingredients have no undue adverse effect on health for the general purpose of treatment.
- pharmaceutically acceptable excipient or carrier means one or more compatible solid or liquid fillers or gel substances, which are suitable for human use and must have sufficient purity and sufficiently low toxicity.
- Cosmetic here means that the components in the composition can be mixed with the compounds of the present invention and with each other without significantly reducing the efficacy of the compounds.
- pharmacologically acceptable excipients or carriers include cellulose and its derivatives (such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers, wetting agents (such as sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
- cellulose and its derivatives such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.
- gelatin such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.
- the pharmaceutical composition can be specially formulated in solid or liquid form for oral administration, for parenteral injection or for rectal administration.
- the pharmaceutical composition can be formulated into a variety of dosage forms for easy administration, for example, oral preparations (such as tablets, capsules, solutions or suspensions), injectable preparations (such as injectable solutions or suspensions, or injectable dry powders that can be used immediately after adding a pharmaceutical solvent before injection).
- the total daily dosage of the salts, polymorphs and pharmaceutical compositions of the compounds of formula I of the present invention shall be determined by the attending physician within the scope of sound medical judgment.
- the specific therapeutically effective dosage level shall be determined based on a variety of factors, including the disorder being treated and the severity of the disorder; the activity of the specific compound used; the specific composition used; the patient's age, weight, general health, sex and diet; the administration time, route of administration and excretion rate of the specific compound used; the duration of treatment; drugs used in combination or concurrently with the specific compound used; and similar factors known in the medical field. For example, it is practiced in the art to start the dosage of the compound from a level lower than that required to obtain the desired therapeutic effect and gradually increase the dosage until the desired effect is obtained.
- free state refers to a form of a compound that has not been further salted, such as the compound of Formula I itself herein. It will be understood by those skilled in the art that when the “free state” forms a salt with a certain acid, the “free state” can be understood as the “free base” corresponding to the salt formed by the acid. That is, “free base” and “free state” have equivalent meanings, and similarly, “free acid” can also have equivalent meanings to “free state” (when the free state forms a salt with a base).
- the present invention provides salts of compounds of formula I, and crystal forms of compounds of formula I and their salts, which have good pharmaceutical properties.
- Figure 1-1 is an XRPD diagram of free form A of the compound of formula I;
- Figure 1-2 is a DSC diagram of the free form A of the compound of formula I;
- Figure 1-3 is a TGA diagram of the free form A of the compound of formula I;
- Figure 2-1 is an XRPD diagram of free form B of the compound of formula I;
- Figure 2-2 is a DSC diagram of the free form B of the compound of formula I;
- Figure 2-3 is a TGA diagram of the free form B of the compound of formula I;
- the instrument and detection method adopted by the present invention are as follows:
- the XRPD pattern was collected on an X-ray powder diffraction analyzer produced by Bruker and PANalytacal, and the scanning parameters are shown in Tables A-1a and A-1b below, respectively.
- TGA Thermogravimetric analysis
- DSC differential scanning calorimetry
- Dynamic moisture sorption (DVS) curves were collected on the DVS IntrInsic of SMS (Surface Measurement Systems). The relative humidity at 25°C was calibrated using the deliquescent points of LiCl, Mg(NO 3 ) 2 and KCl. The DVS test parameters are listed in Table A-3.
- Liquid-state NMR spectra were collected on a Bruker 400M NMR spectra using DMSO- d6 as solvent.
- Step 1 Synthesis of 4,5-dichloro-2-(4-(trifluoromethoxy)phenoxy)benzoic acid
- Step 2 Synthesis of 4,5-dichloro-N-(pyrimidin-5-yl)-2-(4-(trifluoromethoxy)phenoxy)benzamide
- Table 1-4 XRPD diffraction peak data of free form A
- the XRPD characterization results of the obtained solid show (Table 2-1) that a total of 2 salt form samples were obtained in the salt screening test, namely maleate form A and sodium salt form A.
- the screened salt samples were characterized by TGA/DSC, and the salt formation molar ratio was determined by 1 H NMR and HPLC/IC.
- the salt characterization results are summarized in Table 2-2.
- a The sample was clarified after stirring at room temperature and then stirred at 5°C;
- b The sample was clarified after stirring at 5°C and then 1.0 mL of n-heptane was added to induce crystallization;
- c The sample became gel after adding n-heptane to induce crystallization and then the temperature cycle was performed from 50°C to 5°C;
- d The sample was clarified after adding n-heptane to induce crystallization and then stirred at 5°C;
- e The sample remained clarified after stirring at 5°C and then volatilized at room temperature.
- Maleate Form A is obtained by slurrying a free Form A sample and an equimolar amount of maleic acid in EtOAc at room temperature for 3 days, centrifuging and vacuum drying the solid sample at 50°C.
- the XRPD and TGA/DSC results of the maleate Form A sample are shown in Figures 3-1 and 3-2, respectively.
- the TGA results show that the sample has a weight loss of 1.41% when heated to 120°C, and the DSC results show that an endothermic signal is observed at 139.1°C (starting point temperature).
- the 1 H NMR results are shown in Figure 3-3.
- the molar ratio of maleic acid to the compound of formula I in the sample is 1:1, and no residual EtOAc solvent is found.
- the XRPD diffraction peak data of maleate Form A are shown in Table 2-3.
- Sodium salt form A is obtained by slurrying a free form A sample and an equimolar amount of sodium hydroxide in 2-MeTHF at room temperature for 3 days, centrifuging and separating the solid sample, and vacuum drying at 50°C.
- the XRPD and TGA/DSC results of the sodium salt form A sample are listed in Figures 4-1 and 4-2, respectively.
- the TGA results show that the sample has a 2.75% weight loss when heated to 150°C
- the DSC results show that the sample has two endothermic peaks at 191.0°C and 215.0°C (peak temperature).
- the 1 H NMR results are shown in Figure 4-3, and no 2-MeTHF solvent residue was found in the sample.
- HPLC/IC results show that the molar ratio of Na + to the compound of formula I in sodium salt form A is 1:1.
- the XRPD diffraction peak data of sodium salt form A are shown in Table 2-4 below.
- the solid feed concentration was 10 mg/mL (in free form) and the mixture was mixed by rotation at 37°C.
- the solubility of each sample in H 2 O, SGF, FaSSIF and FeSSIF was measured at different time points (1, 2, 4 and 24 hours). After sampling at each time point, the samples were centrifuged (10000 rpm) and filtered (0.45 ⁇ m PTFE). The HPLC concentration and pH value of the filtrate were measured, and the solid samples after centrifugation were tested by XRPD.
- the solubility test results are summarized in Table 2-5, and the solubility curve is shown in Figure 5.
- the XRPD results show that the free form A does not change its crystalline form after the dynamic solubility test (Figure 6-1 to Figure 6-4).
- the hygroscopicity of free crystalline form A and maleate crystalline form A was evaluated by dynamic moisture sorption instrument (DVS).
- the percentage change in mass of the samples was collected when the humidity changed (0% RH-95% RH) under a constant temperature of 25°C.
- the DVS test results and the XRPD results of the samples before and after the DVS test are shown in Figures 7-1 to 7-4.
- the results show that the moisture adsorption of free crystalline form A and maleate crystalline form A at 25°C/80% RH are 0.1698% and 0.224%, respectively.
- the crystal form of all samples remained unchanged after the DVS test, especially the free crystalline form A has almost no hygroscopicity.
- the free form A and the maleate form A were placed at 80°C/closed for one day and at 25°C/60%RH and 40°C/75%RH for one week, respectively, and the physical and chemical stability of the samples was tested by XRPD and HPLC.
- the purity data are listed in Table 2-6, and the XRPD results are listed in Figures 8-1 and 8-2. The results show that no obvious purity changes were observed for all samples, and the crystal form remained unchanged.
- Test Example 1 Detection of the inhibitory activity of compounds on Nav1.8 ion channels
- test compound and control compound solutions contained 1 ⁇ M TTX.
- the intracellular solution was: aspartic acid, 140 mM; magnesium chloride, 2; ethylene glycol tetraacetic acid (EGTA), 11 mM; N-2-hydroxyethylpiperazine-N'-2-ethanesulfonic acid (HEPES), 10 mM.
- the pH was adjusted to 7.4 with cesium hydroxide.
- test compound was dissolved in dimethyl sulfoxide (DMSO) at a concentration of 9 mM. On the day of the test, it was dissolved in the extracellular fluid to the required concentration.
- DMSO dimethyl sulfoxide
- the test article is administered by perfusion using a perfusion system that uses its own gravity.
- the peak current amplitude is observed for at least 1 minute until it stabilizes.
- the CV% of all peak current amplitudes should be less than 10% to exclude the ups and downs of the initial current.
- the average of the peak current amplitudes recorded in the last 10 times during the initial recording period is used as the current peak of the negative control.
- the test sample is administered from a low concentration until the peak current recorded in 10 times stabilizes again or after continuous administration for 5 minutes, the peak current after administration is "unchanged" from that before administration.
- the peak current average of the last 10 scans for each concentration is used as the peak current for data analysis. If a steady state cannot be reached within 5 minutes, the peak current average of the last 10 scans at this time is used as the peak current for data analysis. At the same time, the cell should be discarded and no longer used for detection of higher concentrations. At least two cells are tested for each concentration of the compound.
- pCLAMP 10 Molecular Devices, Union City, CA.
- Current stability refers to the current changing within a limited range over time.
- the inhibitory activity (IC 50 ) of the drug on the Nav1.8 ion channel is calculated by plotting the dose-effect relationship between the drug's gradient dilution series concentration and the stable current value generated by its action on HEK293/ Nav1.8 .
- test results show that the compound of the present invention has good pharmacokinetic characteristics.
- Mechanical pain threshold detection method The test animal is continuously stimulated with Von-Frey fibers to make the fibers bend, and the animal's paw retraction reaction is observed.
- the test animals are stimulated one by one in the order of the fiber weight from small to large, and each fiber weight is stimulated 5 times in a row. If the positive reaction is less than 3 times, repeat the above operation with a larger fiber. When the positive reaction occurs 3 times or more for the first time, the fiber is the pain threshold of the animal (each animal is tested 3 times and the average value is taken).
- Fiber weight 0.6, 1.0, 1.4, 2.0, 4.0, 6.0, 8.0, 10.0, 15.0; the cut-off value is 15.0g.
- Table B-3 Effects of compounds on pain threshold in rats with spinal nerve ligation One-way ANOVA, ***P ⁇ 0.001, **P ⁇ 0.01, *P ⁇ 0.05 NA: No test is scheduled at this time
- test results show that the compound of the present invention can significantly improve the reduction of the mechanical pain threshold of animals caused by spinal nerve ligation modeling in rats and has excellent analgesic effect.
- mice Male SD rats weighing 200-250g were anesthetized and fixed in a prone position. The soles of their hind limbs were flattened upwards, and the toes were fixed with surgical tape and disinfected. A scalpel was used to cut the skin fascia from 0.5cm behind the heel of the animal's foot to the toe tip, and a longitudinal incision of about 1cm was made. After lifting the short flexor digitorum with surgical curved forceps, a longitudinal incision was made on the belly of the muscle with a scalpel without completely cutting the muscle. The skin was sutured and disinfected. On the second day of modeling, the animals were divided into different groups, with 8 rats in each group, and different compounds were orally gavaged. The mechanical pain threshold of the animals was detected with Von-Frey fibers at different time points after administration. For specific grouping dosage and detection time, please see Table B-4 below.
- Table B-4 Effects of compounds on pain threshold in rats with incisional pain model One-way ANOVA, vs Vehicle group, ***P ⁇ 0.001
- test results show that the compound of the present invention can significantly improve the reduction of the mechanical pain threshold of animals caused by the incision pain modeling in rats and has excellent analgesic effect.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Pain & Pain Management (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Neurosurgery (AREA)
- Neurology (AREA)
- Rheumatology (AREA)
- Epidemiology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Description
本申请要求享有申请人:This application requires the applicant to:
于2023年12月13日向中国国家知识产权局提交的,专利申请号为202311723384.5,名称为“一种Nav1.8抑制剂化合物及其盐、多晶型和用途”的在先申请的优先权权益;The priority benefit of the prior application, patent application number 202311723384.5, entitled “A Nav1.8 inhibitor compound and its salt, polymorph and use”, filed with the State Intellectual Property Office of China on December 13, 2023;
于2024年12月4日向中国国家知识产权局提交的,专利申请号为202411777862.5,名称为“一种Nav1.8抑制剂化合物及其盐、多晶型和用途”的在先申请的优先权权益;The priority benefit of the prior application, patent application number 202411777862.5, entitled “A Nav1.8 inhibitor compound and its salt, polymorph and use”, filed with the State Intellectual Property Office of China on December 4, 2024;
所述在先申请的全文通过引用的方式结合于本申请中。The entirety of said prior application is incorporated into the present application by reference.
本发明属于医药领域,涉及一种Nav1.8抑制剂化合物及其盐、多晶型,及它们的制备方法和应用。The present invention belongs to the field of medicine and relates to a Nav1.8 inhibitor compound and its salt, polymorph, and their preparation methods and applications.
疼痛是“一种令人不快的感觉和情绪上的感受,伴有实质上的或潜在的组织损伤,它是一种主观感受”。疼痛可以作为一种警戒信号,提醒机体注意潜在的危险,对机体正常的生命活动具有不可或缺的保护作用。同时,疼痛也是一种常见的临床症状,在引发疼痛的外界刺激消失后,强烈或持久的疼痛会造成生理功能的紊乱,严重影响生命体的生活质量。据统计,全世界约五分之一的人患有中度至重度慢性疼痛。2018年全球镇痛药市场约为360亿美元,预计2023年将达到560亿美元。其中急性中重度未来将以2.5%的年复合增长率稳定增长,慢性疼痛未来市场将18%左右的年复合增长率增长,慢性疼痛是驱动未来十年全球疼痛市场持续增长的主要推动力。Pain is "an unpleasant feeling and emotional sensation, accompanied by actual or potential tissue damage, and it is a subjective feeling". Pain can serve as a warning signal to alert the body to potential dangers and has an indispensable protective effect on the body's normal life activities. At the same time, pain is also a common clinical symptom. After the external stimulus that causes pain disappears, strong or persistent pain can cause physiological dysfunction and seriously affect the quality of life of the living body. According to statistics, about one-fifth of the world's people suffer from moderate to severe chronic pain. The global analgesic market was approximately US$36 billion in 2018 and is expected to reach US$56 billion in 2023. Among them, acute, moderate and severe pain will grow steadily at a compound annual growth rate of 2.5% in the future, and the chronic pain market will grow at a compound annual growth rate of about 18%. Chronic pain is the main driving force for the continued growth of the global pain market in the next decade.
疼痛起源于周围神经系统的伤害感受器。这是一种游离的神经末梢,广泛分布于全身的皮肤、肌肉、关节和内脏组织中,它可以将感受到的热的、机械的或化学的刺激转化为神经冲动(动作电位)并经由传入神经纤维传递到其位于背根神经节(dorsal rootganglia,DRG)的胞体部分,最终传递到高级神经中枢,引起痛觉。而神经元中动作电位的产生和传导又依赖于细胞膜上的电压门控钠离子通道(voltage-gated sodiumchannels,NaV)。当细胞膜去极化时,钠离子通道激活,通道打开,引起钠离子内流,使细胞膜进一步去极化,导致动作电位的产生。因此,抑制异常的钠离子通道活动有助于疼痛的治疗、缓解。Pain originates from nociceptors in the peripheral nervous system. This is a free nerve ending widely distributed in the skin, muscles, joints and visceral tissues throughout the body. It can convert the thermal, mechanical or chemical stimulation it senses into nerve impulses (action potentials) and transmit them to the cell body part located in the dorsal root ganglion (DRG) via afferent nerve fibers, and finally to the higher nerve center, causing pain. The generation and conduction of action potentials in neurons depends on voltage-gated sodium channels (NaV) on the cell membrane. When the cell membrane is depolarized, the sodium ion channels are activated, the channels open, causing sodium ions to flow in, further depolarizing the cell membrane, and leading to the generation of action potentials. Therefore, inhibiting abnormal sodium ion channel activity helps to treat and relieve pain.
人类钠离子是一类跨膜离子通道蛋白,由分子量260kD的α亚基和分子量为30-40kD的β亚基组成,根据α亚基的不同可以分为9种亚型,分别为Nav1.1~Nav1.9,Nav1.5、Nav1.8和Nav1.9是河豚毒素(tetrodotoxin,TTX)不敏感性钠通道,Nav1.5主要存在于心肌细胞中,Nav1.8、Navl.9存在于外周神经系统。其中Nav1.8是参与慢性疼痛、心房纤维性颤动、布加综合征的重要离子通道,是疼痛治疗的高选择性作用靶点。Human sodium ions are a type of transmembrane ion channel protein, consisting of an α subunit with a molecular weight of 260kD and a β subunit with a molecular weight of 30-40kD. According to the different α subunits, it can be divided into 9 subtypes, namely Nav1.1~Nav1.9. Nav1.5, Nav1.8 and Nav1.9 are tetrodotoxin (TTX)-insensitive sodium channels. Nav1.5 is mainly present in myocardial cells, and Nav1.8 and Nav1.9 are present in the peripheral nervous system. Among them, Nav1.8 is an important ion channel involved in chronic pain, atrial fibrillation, and Budd-Chiari syndrome, and is a highly selective target for pain treatment.
Nav1.8编码基因为SCN10A,位于人类染色体3p21-22区域,主要编码α亚单位。研究发现人与大鼠Nav1.8基因的同源性高达93%。Nav1.8主要存在于三叉神经节神经元和DRG神经元中,具有慢速失活、迅速恢复的电生理特征。在表达Nav1.8的神经元内,动作电位的上升主要由Nav1.8电流构成。在神经性疼痛的模型中,神经损伤会使Nav1.8在轴突和神经元胞体中的表达水平上升。使用Nav1.8反义寡核苷酸在降低Nav1.8表达的同时可以明显地缓解疼痛。大鼠爪内注射角叉菜胶后,DRG神经元中Nav1.8的表达有所上升。Nav1.8敲除小鼠不能表现出正常的内脏炎症痛。人类的Nav1.8基因产生功能增益突变后,会导致外周神经痛。根据一系列动物实验以及人类基因证据,选择性抑制Nav1.8具有成为新型镇痛疗法的潜力,可以用于炎性疼痛、神经疼痛、手术后疼痛和癌痛等多种疼痛类型的治疗。The gene encoding Nav1.8 is SCN10A, which is located in the human chromosome 3p21-22 region and mainly encodes the α subunit. Studies have found that the homology of human and rat Nav1.8 genes is as high as 93%. Nav1.8 is mainly present in trigeminal ganglion neurons and DRG neurons, and has the electrophysiological characteristics of slow inactivation and rapid recovery. In neurons expressing Nav1.8, the rise of action potential is mainly composed of Nav1.8 current. In the model of neuropathic pain, nerve injury will increase the expression level of Nav1.8 in axons and neuronal cell bodies. The use of Nav1.8 antisense oligonucleotides can significantly relieve pain while reducing Nav1.8 expression. After carrageenan was injected into the rat paw, the expression of Nav1.8 in DRG neurons increased. Nav1.8 knockout mice cannot show normal visceral inflammatory pain. When the human Nav1.8 gene produces a gain-of-function mutation, it will cause peripheral neuropathy. Based on a series of animal experiments and human genetic evidence, selective inhibition of Nav1.8 has the potential to become a new type of analgesic therapy, which can be used to treat various types of pain, such as inflammatory pain, neuralgia, postoperative pain and cancer pain.
一些已知的Nav’s抑制剂的主要缺点是它们的治疗窗口差,这可能是它们缺乏同种型选择性的结果。由于Navl.8主要限于感知疼痛的神经元,因此选择性Nav1.8阻断剂不太可能诱导非选择性Nav’s阻断剂常见的不良反应。因此,本领域仍然需要开发新的Nav1.8选择性抑制剂,优选对Nav1.8选择性更好、更有效、代谢稳定性增加、溶解度增加和副作用更少的Nav通道抑制剂。The major drawback of some known Nav's inhibitors is that their therapeutic window is poor, which may be the result of their lack of isotype selectivity. Since Nav1.8 is mainly limited to neurons that perceive pain, selective Nav1.8 blockers are unlikely to induce adverse reactions common to non-selective Nav's blockers. Therefore, the art still needs to develop new Nav1.8 selective inhibitors, preferably Nav channel inhibitors with better selectivity, more effective, increased metabolic stability, increased solubility and fewer side effects to Nav1.8.
中国专利申请CN202310743287.6公开了式I化合物的结构:
Chinese patent application CN202310743287.6 discloses the structure of the compound of formula I:
该式I化合物能够有效拮抗Nav1.8受体活性,在制备治疗与Nav1.8相关疾病的药物方面具有广阔的应用前景,因此,进一步研究式I化合物及其盐型、晶型对于开发有效的治疗药物具有重要意义。The compound of formula I can effectively antagonize the activity of Nav1.8 receptors and has broad application prospects in the preparation of drugs for treating diseases related to Nav1.8. Therefore, further research on the compound of formula I and its salt form and crystal form is of great significance for the development of effective therapeutic drugs.
为解决现有技术中存在的问题,一方面,本发明提供一种式I化合物或其药学上可接受的盐的晶型,其中,式I化合物的结构如下所示:
In order to solve the problems existing in the prior art, on the one hand, the present invention provides a crystalline form of a compound of formula I or a pharmaceutically acceptable salt thereof, wherein the structure of the compound of formula I is as follows:
在一些实施方案中,本发明提供所述式I化合物的游离态晶型A,所述游离态晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在17.79°、18.13°、20.52°、21.63°、25.97°处有衍射峰;进一步的,所述游离态晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图还在以下一处或多处有衍射峰:12.85°、16.20°、24.46°、25.00°;进一步的,所述游离态晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在12.85°、16.20°、17.79°、18.13°、20.52°、21.63°、24.46°、25.00°、25.97°处有衍射峰;更进一步的,所述游离态晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在5.78°、11.56°、12.85°、16.20°、17.39°、17.79°、18.13°、20.52°、21.63°、24.46°、25.00°、25.97°、27.33°、27.77°、28.83°、29.03°、30.33°、30.62°、34.25°、34.64°处有衍射峰;还进一步的,所述游离态晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在5.78°、10.77°、11.56°、12.85°、16.20°、17.39°、17.79°、18.13°、20.52°、21.63°、22.01°、24.46°、25.00°、25.56°、25.97°、27.33°、27.77°、28.83°、29.03°、30.33°、30.62°、31.77°、33.33°、34.25°、34.64°、35.19°、36.01°、38.66°处有衍射峰;还更进一步的,所述游离态晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在5.78°、8.10°、9.06°、10.77°、11.56°、12.85°、16.20°、17.39°、17.79°、18.13°、18.45°、18.93°、19.55°、20.52°、21.63°、22.01°、23.20°、23.89°、24.46°、25.00°、25.56°、25.97°、26.39°、27.33°、27.77°、28.83°、29.03°、29.60°、30.33°、30.62°、31.14°、31.77°、32.23°、33.33°、33.77°、34.25°、34.64°、35.19°、36.01°、38.66°、32.86°处有衍射峰;还更进一步的,所述游离态晶型A具有基本如图1-1所示的XRPD谱图。In some embodiments, the present invention provides a free crystalline form A of the compound of formula I, wherein the X-ray powder diffraction spectrum of the free crystalline form A represented by a diffraction angle of 2θ±0.2° has diffraction peaks at 17.79°, 18.13°, 20.52°, 21.63°, and 25.97°; further, the X-ray powder diffraction spectrum of the free crystalline form A represented by a diffraction angle of 2θ±0.2° also has diffraction peaks at one or more of the following: 12.85°, 16.20°, 24.46°, and 25.00°; further, the X-ray powder diffraction spectrum of the free crystalline form A represented by a diffraction angle of 2θ±0.2° has diffraction peaks at 12.85°, 16.20°, 17.79°, 18.13°, 20.52°, 21.63°, and 24.46°. Furthermore, the X-ray powder diffraction spectrum of the free crystalline form A represented by a diffraction angle of 2θ±0.2° is 5.78°, 11.56°, 12.85°, 16.20°, 17.39°, 17.79°, 18.13°, 20.52°, 21.63°, 24.46°, 25.00°, 25.97°. There are diffraction peaks at 25.97°, 27.33°, 27.77°, 28.83°, 29.03°, 30.33°, 30.62°, 34.25°, and 34.64°; further, the X-ray powder diffraction spectrum of the free crystalline form A represented by a diffraction angle of 2θ±0.2° is 5.78°, 10.77°, 11.56°, 12.85°, 16.20°、17.39°、17.79°、18.13°、20.52°、21.63°、22.01°、24.46°、25.00°、25.56°、25.97°、27.33°、27.77°、28.83°、29.03°、30.33°、30.62°、31.77°、33.33°、3 The diffraction peaks are at 4.25°, 34.64°, 35.19°, 36.01°, and 38.66°; further, the X-ray powder diffraction spectrum of the free crystalline form A represented by a diffraction angle of 2θ±0.2° is 5.78°, 8.10°, 9.06°, 10.77°, 11.56°, 12.85°, 16.20°, 17.39°, 17 .79°, 18.13°, 18.45°, 18.93°, 19.55°, 20.52°, 21.63°, 22.01°, 23.20°, 23.89°, 24.46°, 25.00°, 25.56°, 25.97°, 26.39°, 27.33°, 27.77°, 28.83°, 29.03°, 29. There are diffraction peaks at 60°, 30.33°, 30.62°, 31.14°, 31.77°, 32.23°, 33.33°, 33.77°, 34.25°, 34.64°, 35.19°, 36.01°, 38.66°, and 32.86°; further, the free crystalline form A has an XRPD spectrum substantially as shown in Figure 1-1.
在一些实施方案中,所述游离态晶型A具有以下的一个、两个或三个特征:In some embodiments, the free crystalline Form A has one, two or three of the following characteristics:
(1)游离态晶型A的TGA曲线在150.0±3℃时失重约0.38±1%;(1) The TGA curve of free form A shows a weight loss of approximately 0.38±1% at 150.0±3°C;
(2)游离态晶型A的DSC曲线在169.5±3℃处有一个吸热峰的起始点;(2) The DSC curve of the free form A has an endothermic peak starting point at 169.5±3℃;
(3)游离态晶型A的DSC曲线在171.0±3℃处有一个吸热峰。(3) The DSC curve of free form A has an endothermic peak at 171.0±3℃.
在一些实施方案中,所述游离态晶型A的DSC图如图1-2所示;所述游离态晶型A的TGA图如图1-3所示。In some embodiments, the DSC graph of the free crystalline form A is shown in Figure 1-2; the TGA graph of the free crystalline form A is shown in Figure 1-3.
根据本发明的实施方案,所述游离态晶型A为无水晶型。According to an embodiment of the present invention, the free-state crystalline form A is an anhydrous crystalline form.
在一些实施方案中,本发明提供所述式I化合物的游离态晶型B,所述游离态晶型B以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在11.64°、12.60°、17.46°、20.93°、25.16°、26.56°处有衍射峰;进一步的,所述游离态晶型B以2θ±0.2°衍射角表示的X-射线粉末衍射谱图还在以下一处或多处有衍射峰:5.80°、29.25°、28.22°、28.51°、35.32°;更进一步的,所述游离态晶型B以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在5.80°、11.64°、12.60°、17.46°、20.93°、22.19°、25.16°、26.56°、28.22°、28.51°、29.25°、33.23°、35.32°、38.52°处有衍射峰;还进一步的,所述游离态晶型B以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在5.80°、11.64°、12.60°、16.09°、17.46°、18.37°、20.93°、22.19°、23.03°、23.82°、25.16°、26.56°、28.22°、28.51°、29.25°、29.79°、32.20°、33.23°、34.42°、35.32°、38.52°处有衍射峰;还更进一步的,所述游离态晶型B具有基本如图2-1所示的XRPD谱图。In some embodiments, the present invention provides a free crystalline form B of the compound of formula I, wherein the free crystalline form B has an X-ray powder diffraction spectrum represented by a diffraction angle of 2θ±0.2° at 11.64°, 12.60°, 17.46°, 20.93°, 25.16°, and 26.56°; further, the free crystalline form B has an X-ray powder diffraction spectrum represented by a diffraction angle of 2θ±0.2° at 11.64°, 12.60°, 17.46°, 20.93°, 25.16°, and 26.56°. The X-ray powder diffraction spectrum of the free crystalline form B also has diffraction peaks at one or more of the following positions: 5.80°, 29.25°, 28.22°, 28.51°, and 35.32°; further, the X-ray powder diffraction spectrum of the free crystalline form B represented by a diffraction angle of 2θ±0.2° has diffraction peaks at 5.80°, 11.64°, 12.60°, 17.46°, 20.93°, 22.19°, 25.16 The free form B has diffraction peaks at 5.80°, 11.64°, 12.60°, 16.09°, 17.46°, 18.37°, 20.9 There are diffraction peaks at 3°, 22.19°, 23.03°, 23.82°, 25.16°, 26.56°, 28.22°, 28.51°, 29.25°, 29.79°, 32.20°, 33.23°, 34.42°, 35.32°, and 38.52°; further, the free form B has an XRPD spectrum substantially as shown in Figure 2-1.
在一些实施方案中,所述游离态晶型B具有以下的一个、两个或三个特征:In some embodiments, the free crystalline Form B has one, two or three of the following characteristics:
(1)游离态晶型B的TGA曲线在150.0±3℃时失重约0.23±1%;(1) The TGA curve of free form B shows a weight loss of approximately 0.23±1% at 150.0±3°C;
(2)游离态晶型B的DSC曲线在165.8±3℃处有一个吸热峰的起始点;(2) The DSC curve of the free form B has an endothermic peak starting point at 165.8±3℃;
(3)游离态晶型B的DSC曲线在168.47±3℃处有一个吸热峰。(3) The DSC curve of free form B has an endothermic peak at 168.47±3℃.
在一些实施方案中,所述游离态晶型B的DSC图如图2-2所示;所述游离态晶型B的TGA图如图2-3所示。In some embodiments, the DSC graph of the free crystalline form B is shown in Figure 2-2; the TGA graph of the free crystalline form B is shown in Figure 2-3.
根据本发明的实施方案,所述游离态晶型B为无水晶型。According to an embodiment of the present invention, the free-state crystalline form B is an anhydrous crystalline form.
另一方面,本发明提供所述式I化合物的游离态晶型A的制备方法,其包括如下几种方法:On the other hand, the present invention provides a method for preparing the free crystalline form A of the compound of formula I, which comprises the following methods:
方法一、将盛有式I化合物的第一样品瓶敞口置于盛有溶剂的第二样品瓶中,密封第二样品瓶,室温静置;所述溶剂不没过第一样品瓶的瓶口;Method 1: Place the first sample bottle containing the compound of formula I in an open position in a second sample bottle containing a solvent, seal the second sample bottle, and let it stand at room temperature; the solvent does not cover the mouth of the first sample bottle;
所述溶剂选自乙醇、丙酮、甲基叔丁基醚、乙酸乙酯、二氯甲烷、四氢呋喃、乙腈、正庚烷、甲苯中的一种或多种;The solvent is selected from one or more of ethanol, acetone, methyl tert-butyl ether, ethyl acetate, dichloromethane, tetrahydrofuran, acetonitrile, n-heptane, and toluene;
方法二、将盛有式I化合物溶液的第一样品瓶敞口置于盛有反溶剂的第二样品瓶中,密封第二样品瓶,室温静置;所述反溶剂不没过第一样品瓶的瓶口;Method 2: Place the first sample bottle containing the solution of the compound of formula I in the second sample bottle containing the anti-solvent, seal the second sample bottle, and let it stand at room temperature; the anti-solvent does not cover the mouth of the first sample bottle;
所述式I化合物溶液中的溶剂选自二氯甲烷、四氢呋喃、1,4-二氧六环、二甲基亚砜、N,N-二甲基甲酰胺中的一种或多种;The solvent in the solution of the compound of formula I is selected from one or more of dichloromethane, tetrahydrofuran, 1,4-dioxane, dimethyl sulfoxide, and N,N-dimethylformamide;
所述反溶剂选自正庚烷、甲基叔丁基醚、水中的一种或多种;The anti-solvent is selected from one or more of n-heptane, methyl tert-butyl ether, and water;
方法三、在室温下,向式I化合物加入溶剂,磁力搅拌,收集固体;Method 3: Add a solvent to the compound of formula I at room temperature, stir magnetically, and collect the solid;
所述溶剂选自水、乙醇、异丙醇、乙酸乙酯、乙酸异丙酯、四氢呋喃、1,4-二氧六环、甲基叔丁基醚、丙酮、甲基乙基酮、二甲基亚砜、乙腈、甲苯、N,N-二甲基甲酰胺、N-甲基吡咯烷酮中的一种或多种;The solvent is selected from one or more of water, ethanol, isopropanol, ethyl acetate, isopropyl acetate, tetrahydrofuran, 1,4-dioxane, methyl tert-butyl ether, acetone, methyl ethyl ketone, dimethyl sulfoxide, acetonitrile, toluene, N,N-dimethylformamide, and N-methylpyrrolidone;
方法四、在50℃下,向式I化合物加入溶剂,磁力搅拌,收集固体;Method 4: Add solvent to the compound of formula I at 50°C, stir magnetically, and collect the solid;
所述溶剂选自水、乙醇、异丙醇、乙酸乙酯、乙酸异丙酯、四氢呋喃、1,4-二氧六环、甲基叔丁基醚、丙酮、甲基乙基酮、二甲基亚砜、乙腈、甲苯、N,N-二甲基甲酰胺、N-甲基吡咯烷酮中的一种或多种;The solvent is selected from one or more of water, ethanol, isopropanol, ethyl acetate, isopropyl acetate, tetrahydrofuran, 1,4-dioxane, methyl tert-butyl ether, acetone, methyl ethyl ketone, dimethyl sulfoxide, acetonitrile, toluene, N,N-dimethylformamide, and N-methylpyrrolidone;
方法五、将式I化合物加入有机溶剂I中,溶解后过滤,于室温下挥发:Method 5: Add the compound of formula I into organic solvent I, filter after dissolving, and evaporate at room temperature:
优选地,所述有机溶剂I选自甲醇、乙醇、乙酸乙酯、四氢呋喃、1,4-二氧六环、丙酮、甲基乙基酮、二氯甲烷、乙腈中的一种或多种;Preferably, the organic solvent I is selected from one or more of methanol, ethanol, ethyl acetate, tetrahydrofuran, 1,4-dioxane, acetone, methyl ethyl ketone, dichloromethane and acetonitrile;
方法六、将式I化合物在良溶剂中完全溶解,过滤,向澄清溶液中滴加反溶剂,至有固体析出;Method 6: Completely dissolve the compound of formula I in a good solvent, filter, and dropwise add an antisolvent to the clear solution until solid precipitates;
所述良溶剂选自二甲基亚砜、N,N-二甲基甲酰胺、二氯甲烷、四氢呋喃、甲基乙基酮、N-甲基吡咯烷酮、N,N-二甲基乙酰胺、乙醇、乙酸异丙酯中的一种或多种;The good solvent is selected from one or more of dimethyl sulfoxide, N,N-dimethylformamide, dichloromethane, tetrahydrofuran, methyl ethyl ketone, N-methylpyrrolidone, N,N-dimethylacetamide, ethanol, and isopropyl acetate;
所述反溶剂选自水、甲苯、甲基叔丁基醚、正庚烷中的一种或多种;The anti-solvent is selected from one or more of water, toluene, methyl tert-butyl ether, and n-heptane;
在一些实施方案中,所述反溶剂选自水时,良溶剂选自二甲基亚砜、N,N-二甲基甲酰胺、四氢呋喃、N-甲基吡咯烷酮、N,N-二甲基乙酰胺中的一种;所述反溶剂选自甲基叔丁基醚时,良溶剂选自二氯甲烷;所述反溶剂选自正庚烷时,良溶剂选自甲基乙基酮或乙醇。In some embodiments, when the anti-solvent is selected from water, the good solvent is selected from one of dimethyl sulfoxide, N,N-dimethylformamide, tetrahydrofuran, N-methylpyrrolidone, and N,N-dimethylacetamide; when the anti-solvent is selected from methyl tert-butyl ether, the good solvent is selected from dichloromethane; when the anti-solvent is selected from n-heptane, the good solvent is selected from methyl ethyl ketone or ethanol.
方法七、将式I化合物在良溶剂中完全溶解,过滤,将澄清溶液加入反溶剂中;Method 7: Dissolve the compound of formula I completely in a good solvent, filter, and add the clear solution to an anti-solvent;
所述良溶剂选自二甲基亚砜、N,N-二甲基甲酰胺、二氯甲烷、乙酸乙酯、1,4-二氧六环、甲基乙基酮、四氢呋喃中的一种或多种;The good solvent is selected from one or more of dimethyl sulfoxide, N,N-dimethylformamide, dichloromethane, ethyl acetate, 1,4-dioxane, methyl ethyl ketone, and tetrahydrofuran;
所述反溶剂选自水、正庚烷、甲基叔丁基醚、甲苯中的一种或多种;The anti-solvent is selected from one or more of water, n-heptane, methyl tert-butyl ether, and toluene;
在一些实施方案中,所述反溶剂选自水时,良溶剂选自二甲基亚砜或N,N-二甲基甲酰胺;所述反溶剂选自甲基叔丁基醚时,良溶剂选自1,4-二氧六环或甲基乙基酮;所述反溶剂选自正庚烷时,良溶剂选自乙酸乙酯;所述反溶剂选自甲苯时,良溶剂选自甲基乙基酮或四氢呋喃。In some embodiments, when the anti-solvent is selected from water, the good solvent is selected from dimethyl sulfoxide or N,N-dimethylformamide; when the anti-solvent is selected from methyl tert-butyl ether, the good solvent is selected from 1,4-dioxane or methyl ethyl ketone; when the anti-solvent is selected from n-heptane, the good solvent is selected from ethyl acetate; when the anti-solvent is selected from toluene, the good solvent is selected from methyl ethyl ketone or tetrahydrofuran.
方法八、在50℃下,将式I化合物加入有机溶剂I中,溶解后过滤,磁力搅拌,室温下冷却;Method 8: Add the compound of formula I into organic solvent I at 50°C, filter after dissolving, stir magnetically, and cool at room temperature;
优选地,所述有机溶剂I选自甲醇、乙醇、乙酸异丙酯、乙腈、乙酸乙酯、丙酮中的一种或多种;Preferably, the organic solvent I is selected from one or more of methanol, ethanol, isopropyl acetate, acetonitrile, ethyl acetate, and acetone;
方法九、将式I化合物加入研钵中或者将式I化合物和溶剂加入研钵中,研磨;Method 9: Add the compound of formula I into a mortar or add the compound of formula I and a solvent into a mortar and grind;
优选地,所述溶剂选自水、甲基叔丁基醚、正庚烷中的一种或多种。Preferably, the solvent is selected from one or more of water, methyl tert-butyl ether and n-heptane.
另一方面,本发明提供所述式I化合物的游离态晶型B的制备方法,其包括如下几种方法:On the other hand, the present invention provides a method for preparing the free crystalline form B of the compound of formula I, which comprises the following methods:
方法一、将式I化合物在良溶剂中完全溶解,过滤,向澄清溶液中滴加反溶剂,至有固体析出;Method 1: Dissolve the compound of formula I completely in a good solvent, filter, and add an antisolvent dropwise to the clear solution until solid precipitates;
所述良溶剂选自四氢呋喃、N,N-二甲基乙酰胺中的一种或两种,反溶剂选自正庚烷。The good solvent is selected from one or two of tetrahydrofuran and N,N-dimethylacetamide, and the anti-solvent is selected from n-heptane.
所述反溶剂选自水、甲苯、甲基叔丁基醚、正庚烷中的一种或多种。The anti-solvent is selected from one or more of water, toluene, methyl tert-butyl ether and n-heptane.
方法二、将式I化合物在良溶剂中完全溶解,过滤,将澄清溶液加入反溶剂中;Method 2: completely dissolve the compound of formula I in a good solvent, filter, and add the clear solution into an anti-solvent;
所述良溶剂选自二氯甲烷、1,4-二氧六环中的一种或两种;所述反溶剂选自正庚烷。The good solvent is selected from one or two of dichloromethane and 1,4-dioxane; the anti-solvent is selected from n-heptane.
方法三、在40-60℃(例如50℃)下,将式I化合物加入异丙醇中,溶解后过滤,搅拌,室温下冷却。Method 3: Add the compound of formula I into isopropanol at 40-60° C. (eg 50° C.), dissolve, filter, stir, and cool at room temperature.
方法四、将式I化合物在四氢呋喃中完全溶解,过滤,搅拌下向澄清溶液中加入正庚烷,至有固体析出,再加入游离态晶型B晶种,搅拌。Method 4: Completely dissolve the compound of formula I in tetrahydrofuran, filter, add n-heptane to the clear solution with stirring until solid precipitates, then add free form B seed crystals and stir.
根据本发明的实施方案,所述游离态晶型B晶种可采用所述方法一、二、三、四中的一种制备得到。According to an embodiment of the present invention, the free-state crystal form B seed can be prepared by one of the methods one, two, three, and four.
另一方面,本发明提供所述式I化合物药学上可接受的盐,其中,所述药学上可接受的盐选自式I化合物与酸或碱性成的盐,在一些实施方案中,所述式I化合物药学上可接受的盐选自式I化合物与以下酸或碱形成的盐:盐酸、硫酸、马来酸、磷酸、富马酸、酒石酸、柠檬酸、L-苹果酸、琥珀酸、对甲苯磺酸、甲磺酸、氢氧化钠、精氨酸。On the other hand, the present invention provides a pharmaceutically acceptable salt of the compound of formula I, wherein the pharmaceutically acceptable salt is selected from the salts formed by the compound of formula I and an acid or base. In some embodiments, the pharmaceutically acceptable salt of the compound of formula I is selected from the salts formed by the compound of formula I and the following acids or bases: hydrochloric acid, sulfuric acid, maleic acid, phosphoric acid, fumaric acid, tartaric acid, citric acid, L-malic acid, succinic acid, p-toluenesulfonic acid, methanesulfonic acid, sodium hydroxide, and arginine.
当所述式I化合物与酸或碱形成盐时,所述酸或碱与式I化合物的摩尔比可以为5:1~1:5,例如5:1、4.5:1、4:1、3.5:1、3:1、2.5:1、2:1、1.5:1、1:1、1:1.5、1:2、1:2.5、1:3、1:3.5、1:4、1:4.5、1:5。When the compound of formula I forms a salt with an acid or a base, the molar ratio of the acid or the base to the compound of formula I can be 5:1 to 1:5, for example, 5:1, 4.5:1, 4:1, 3.5:1, 3:1, 2.5:1, 2:1, 1.5:1, 1:1, 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5, 1:5.
在一些实施方案中,所述式I化合物药学上可接受的盐选自式I化合物的马来酸盐;在一些实施方案中,所述马来酸盐中,式I化合物与马来酸的摩尔比为1:1。In some embodiments, the pharmaceutically acceptable salt of the compound of formula I is selected from the maleate salt of the compound of formula I; in some embodiments, in the maleate salt, the molar ratio of the compound of formula I to maleic acid is 1:1.
在一些实施方案中,所述式I化合物药学上可接受的盐选自式I化合物的钠盐;在一些实施方案中,所述钠盐中,式I化合物与钠的摩尔比为1:1。In some embodiments, the pharmaceutically acceptable salt of the compound of formula I is selected from the sodium salt of the compound of formula I; in some embodiments, in the sodium salt, the molar ratio of the compound of formula I to sodium is 1:1.
另一方面,本发明提供所述式I化合物药学上可接受的盐的晶型。In another aspect, the present invention provides a crystalline form of a pharmaceutically acceptable salt of the compound of formula I.
在一些实施方案中,本发明提供式I化合物的马来酸盐晶型A,所述马来酸盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在8.39°、13.78°、16.31°、17.07°、18.44°处有衍射峰;进一步的,所述马来酸盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在8.39°、13.78°、16.31°、17.07°、18.44°、20.96°、25.62°、26.78°处有衍射峰;进一步的,所述马来酸盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在8.39°、13.78°、16.31°、17.07°、18.44°、20.96°、21.61°、25.29°、25.62°、26.78°处有衍射峰;进一步的,所述马来酸盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在8.39°、13.78°、16.31°、17.07°、18.44°、20.45°、20.96°、21.61°、25.29°、25.62°、26.19°、26.78°处有衍射峰;进一步的,所述马来酸盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在8.39°、13.78°、16.31°、17.07°、18.44-°、19.01°、20.45°、20.96°、21.61°、22.65°、23.19°、24.62°、25.29°、25.62°、26.19°、26.78°、27.03°、27.41°、28.47°、28.96°、30.05°、31.25°、31.61°、33.91°处有衍射峰;进一步的,所述马来酸盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在8.39°、10.65°、13.78°、16.31°、17.07°、18.44°、19.01°、20.45°、20.96°、21.61°、22.65°、23.19°、24.62°、25.29°、25.62°、26.19°、26.78°、27.03°、27.41°、28.47°、28.96°、30.05、30.74°、31.25°、31.61°、32.70°、33.13°、33.91°、34.49°、35.22°、36.21°、38.48°处有衍射峰;进一步的,所述马来酸盐晶型A具有基本如图3-1所示的XRPD谱图;In some embodiments, the present invention provides a maleate salt form A of a compound of formula I, wherein the maleate salt form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2θ±0.2° and has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, and 18.44°; further, the maleate salt form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2θ±0.2° and has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, 18.44°, 20.96°, 25.62°, and 26.78°; further, the maleate salt form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2θ±0.2° and has diffraction peaks at 2θ±0.2°. The figure has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, 18.44°, 20.96°, 21.61°, 25.29°, 25.62°, and 26.78°; further, the X-ray powder diffraction spectrum of the maleate salt form A represented by a diffraction angle of 2θ±0.2° has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, 18.44°, 20.45°, 20.96°, 21.61°, 25.29°, 25.62°, 26.19°, and 26.78°; further, the X-ray powder diffraction spectrum of the maleate salt form A represented by a diffraction angle of 2θ±0.2° has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, 18.44°, 20.45°, 20.96°, 21.61°, 25.29°, 25.62°, 26.19°, and 26.78° The X-ray powder diffraction spectrum of the maleate salt form A has diffraction peaks at 8.39°, 13.78°, 16.31°, 17.07°, 18.44°, 19.01°, 20.45°, 20.96°, 21.61°, 22.65°, 23.19°, 24.62°, 25.29°, 25.62°, 26.19°, 26.78°, 27.03°, 27.41°, 28.47°, 28.96°, 30.05°, 31.25°, 31.61°, and 33.91°; further, the X-ray powder diffraction spectrum of the maleate salt form A represented by a diffraction angle of 2θ±0.2° has diffraction peaks at 8.39°, 10.65°, 13.78°, 16.31°, 17.07°, 18.44°, 19.01°, 20.45°, 20.96°, 21.61°, 22.65°, 23.19°, 24.62°, .31°, 17.07°, 18.44°, 19.01°, 20.45°, 20.96°, 21.61°, 22.65°, 23.19°, 24.62°, 25.29°, 25.62°, 26.19°, 26.78°, 27.03°, 27.41°, 28.47°, 28.96°, 30.05, 30.74°, 31.25°, 31.61°, 32.70°, 33.13°, 33.91°, 34.49°, 35.22°, 36.21°, and 38.48°; further, the maleate salt form A has an XRPD spectrum substantially as shown in Figure 3-1;
在一些实施方案中,所述马来酸盐晶型A具有以下特征中的一个或两个:In some embodiments, the maleate salt form A has one or two of the following characteristics:
(1)所述马来酸盐晶型A的TGA曲线在120±3℃时失重0.5-3%,优选的为0.5,1.0,1.5,2.0,2.5,3.0%,例如为1.41%;(1) The TGA curve of the maleate salt form A shows a weight loss of 0.5-3% at 120±3°C, preferably 0.5, 1.0, 1.5, 2.0, 2.5, 3.0%, for example 1.41%;
(2)所述马来酸盐晶型A在139.1±3℃处有一个吸热峰。(2) The maleate salt form A has an endothermic peak at 139.1±3°C.
在一些实施方案中,所述马来酸盐晶型A的TGA/DSC图基本如图3-2所示;所述马来酸盐晶型A的1H NMR图基本如图3-3所示。In some embodiments, the TGA/DSC graph of the maleate salt form A is substantially as shown in FIG. 3-2 ; the 1 H NMR graph of the maleate salt form A is substantially as shown in FIG. 3-3 .
在一些实施方案中,本发明提供式I化合物的钠盐晶型A,所述钠盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在16.70°、17.02°、21.23°、22.33°、24.39°、25.50°、25.89°处有衍射峰;进一步的,所述钠盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在16.70°、17.02°、17.67°、18.51°、21.23°、22.33°、24.39°、25.50°、25.89°、29.73°处有衍射峰;进一步的,所述钠盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在16.70°、17.02°、17.67°、18.51°、21.23°、22.33°、24.39°、25.50°、25.89°、29.73°、30.76°处有衍射峰;进一步的,所述钠盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在4.85°、9.70°、12.13°、13.55°、14.20°、14.55、1670°、17.02°、17.67°、18.51°、21.23°、22.33°、24.39°、25.50°、25.89°、26.72°、27.74°、28.44°、29.73°、30.76°处有衍射峰;进一步的,所述钠盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在4.85°、9.70°、12.13°、13.55°、14.20°、14.55、16.70°、17.02°、17.67°、18.51°、20.27°、21.23°、22.33°、23.35°、24.39°、25.50°、25.89°、26.72°、27.74°、28.44°、29.73°、30.76°、31.43°、31.88°、32.46°、39.71°处有衍射峰;进一步的,所述钠盐晶型A以2θ±0.2°衍射角表示的X-射线粉末衍射谱图在4.85°、9.70°、12.13°、13.55°、14.20°、14.55°、16.70°、17.02°、17.67°、18.51°、20.27°、21.23°、22.33°、23.35°、24.39°、25.50°、25.89°、26.72°、27.74°、28.44°、28.87°、29.73°、30.76°、31.43°、31.88°、32.46°、32.98°、34.06°、35.13°、35.68°、36.33°、38.30°、39.71°处有衍射峰;进一步的,所述钠盐晶型A具有基本如图4-1所示的XRPD谱图。In some embodiments, the present invention provides a sodium salt crystalline form A of the compound of formula I, wherein the sodium salt crystalline form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2θ±0.2° at 16.70°, 17.02°, 21.23°, 22.33°, 24.39°, 25.50°, and 25.89° having diffraction peaks; further, the sodium salt crystalline form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2θ±0.2° at 16.70°, 17.02°, 17.67°, 18.51°, 21.23°, 22.33°, 24.39°, 25.50°, 25.89°, and 29.73° having diffraction peaks; further, the sodium salt crystalline form A has an X-ray powder diffraction spectrum represented by a diffraction angle of 2θ±0.2° at There are diffraction peaks at 16.70°, 17.02°, 17.67°, 18.51°, 21.23°, 22.33°, 24.39°, 25.50°, 25.89°, 29.73°, and 30.76°; further, the X-ray powder diffraction spectrum of the sodium salt form A expressed at a diffraction angle of 2θ±0.2° is at 4.85°, 9.70°, , 12.13°, 13.55°, 14.20°, 14.55, 1670°, 17.02°, 17.67°, 18.51°, 21.23°, 22.33°, 24.39°, 25.50°, 25.89°, 26.72°, 27.74°, 28.44°, 29.73°, and 30.76° have diffraction peaks; In the step, the X-ray powder diffraction spectrum of the sodium salt form A represented by a diffraction angle of 2θ±0.2° is 4.85°, 9.70°, 12.13°, 13.55°, 14.20°, 14.55, 16.70°, 17.02°, 17.67°, 18.51°, 20.27°, 21.23°, 22.33°, 23.35° , 24.39°, 25.50°, 25.89°, 26.72°, 27.74°, 28.44°, 29.73°, 30.76°, 31.43°, 31.88°, 32.46°, and 39.71°; further, the X-ray powder diffraction spectrum of the sodium salt form A represented by a diffraction angle of 2θ±0.2° is at 4.85 °, 9.70°, 12.13°, 13.55°, 14.20°, 14.55°, 16.70°, 17.02°, 17.67°, 18.51°, 20.27°, 21.23°, 22.33°, 23.35°, 24.39°, 25.50°, 25.89°, 26.72°, 27.74°, 28. There are diffraction peaks at 44°, 28.87°, 29.73°, 30.76°, 31.43°, 31.88°, 32.46°, 32.98°, 34.06°, 35.13°, 35.68°, 36.33°, 38.30°, and 39.71°; further, the sodium salt form A has an XRPD spectrum substantially as shown in Figure 4-1.
在一些实施方案中,所述钠盐晶型A具有以下特征中的一个或两个:In some embodiments, the sodium salt form A has one or two of the following characteristics:
(1)所述钠盐晶型A的TGA曲线在150±3℃时失重1.0-5.0%,优选的为1.0,1.5,2.0,2.5,3.0,3.5,4.0,4.5,5.0%,例如为2.75%;(1) The TGA curve of the sodium salt form A shows a weight loss of 1.0-5.0% at 150±3°C, preferably 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0%, for example 2.75%;
(2)所述钠盐晶型A在191.0±3℃和215.0±3℃处有两个吸热峰。(2) The sodium salt form A has two endothermic peaks at 191.0±3°C and 215.0±3°C.
在一些实施方案中,所述钠盐晶型A的TGA/DSC图基本如图4-2所示;所述钠盐晶型A的1H NMR图基本如图4-3所示。In some embodiments, the TGA/DSC graph of the sodium salt crystalline form A is substantially as shown in FIG. 4-2 ; the 1 H NMR graph of the sodium salt crystalline form A is substantially as shown in FIG. 4-3 .
另一方面,本发明提供所述式I化合物药学上可接受的盐(包括盐的晶型)的制备方法,包括将式I化合物和合适的酸或碱混合,得到所述药学上可接受的盐。On the other hand, the present invention provides a method for preparing a pharmaceutically acceptable salt (including a crystalline form of the salt) of the compound of formula I, comprising mixing the compound of formula I with a suitable acid or base to obtain the pharmaceutically acceptable salt.
在一些实施方案中,所述式I化合物药学上可接受的盐为马来酸盐或钠盐时,制备方法包括如下步骤:将式I化合物和等摩尔量的马来酸或氢氧化钠混合,在有机溶剂A搅拌,分离得到固体。在一些实施方案中,所述搅拌的时间为1-5天,例如为3天,搅拌的温度为室温;所述有机溶剂A选自异丙醇、乙酸乙酯、2-甲基四氢呋喃中的一种或多种。在一些实施方案中,将式I化合物和马来酸或氢氧化钠混合,在有机溶剂A中得到悬浊液,悬浮搅拌;In some embodiments, when the pharmaceutically acceptable salt of the compound of formula I is a maleate or sodium salt, the preparation method comprises the following steps: mixing the compound of formula I with an equimolar amount of maleic acid or sodium hydroxide, stirring in an organic solvent A, and separating to obtain a solid. In some embodiments, the stirring time is 1-5 days, for example, 3 days, and the stirring temperature is room temperature; the organic solvent A is selected from one or more of isopropanol, ethyl acetate, and 2-methyltetrahydrofuran. In some embodiments, the compound of formula I is mixed with maleic acid or sodium hydroxide, a suspension is obtained in an organic solvent A, and the suspension is stirred;
在一些实施方案中,所述式I化合物药学上可接受的盐为马来酸盐晶型A时,制备方法包括如下步骤:将式I化合物和等摩尔量的马来酸混合,在有机溶剂A中得到悬浊液,悬浮搅拌,分离得到固体。在一些实施方案中,所述悬浮搅拌的时间为1-5天,例如为3天,悬浮搅拌的温度为室温;所述有机溶剂A选自乙酸乙酯。In some embodiments, when the pharmaceutically acceptable salt of the compound of formula I is maleate crystalline form A, the preparation method comprises the following steps: mixing the compound of formula I with an equimolar amount of maleic acid, obtaining a suspension in an organic solvent A, suspending and stirring, and separating to obtain a solid. In some embodiments, the suspension and stirring time is 1-5 days, for example, 3 days, and the suspension and stirring temperature is room temperature; the organic solvent A is selected from ethyl acetate.
在一些实施方案中,所述式I化合物药学上可接受的盐为钠盐晶型A时,制备方法包括如下步骤:将式I化合物和等摩尔量的氢氧化钠混合,在有机溶剂A中得到悬浊液,悬浮搅拌,分离得到固体。在一些实施方案中,所述悬浮搅拌的时间为1-5天,例如为3天,悬浮搅拌的温度为室温;所述有机溶剂A选自异丙醇、乙酸乙酯、2-甲基四氢呋喃中的一种或多种。In some embodiments, when the pharmaceutically acceptable salt of the compound of formula I is sodium salt crystal form A, the preparation method comprises the following steps: mixing the compound of formula I with an equimolar amount of sodium hydroxide, obtaining a suspension in an organic solvent A, suspending and stirring, and separating to obtain a solid. In some embodiments, the suspension and stirring time is 1-5 days, for example, 3 days, and the suspension and stirring temperature is room temperature; the organic solvent A is selected from one or more of isopropanol, ethyl acetate, and 2-methyltetrahydrofuran.
又一方面,本发明提供了一种药物组合物,所述药物组合物包含如上所述式I化合物游离态晶型(例如游离态晶型A、游离态晶型B)、如上所述式I化合物药学上可接受的盐(包括其晶型形式)中的一种或多种。In another aspect, the present invention provides a pharmaceutical composition comprising one or more of the free crystalline forms of the compound of formula I as described above (e.g., free crystalline form A, free crystalline form B), and the pharmaceutically acceptable salts of the compound of formula I as described above (including their crystalline forms).
在一些实施方案中,所述药物组合物还包含药学上可以接受的赋形剂或载体。In some embodiments, the pharmaceutical composition further comprises a pharmaceutically acceptable excipient or carrier.
又一方面,本发明提供了一种药物组合物,所述药物组合物包含物质A以及药学上可接受的载体,所述的物质A为如上任一项所述的式I化合物的游离态晶型、式I化合物的药学上可接受的盐的晶型;优选地,所述物质A为式I化合物的游离态晶型A、式I化合物的游离态晶型B、马来酸盐晶型A、钠盐晶型A。In another aspect, the present invention provides a pharmaceutical composition, comprising substance A and a pharmaceutically acceptable carrier, wherein substance A is a free crystalline form of a compound of formula I or a crystalline form of a pharmaceutically acceptable salt of a compound of formula I as described in any one of the above items; preferably, substance A is free crystalline form A of a compound of formula I, free crystalline form B of a compound of formula I, maleate crystalline form A, or sodium salt crystalline form A.
又一方面,本发明提供了所述式I化合物游离态晶型(例如游离态晶型A、游离态晶型B)、式I化合物药学上可接受的盐(包括其晶型形式)或者所述的药物组合物在制备药物中的用途,所述药物用于治疗和/或预防电压门控钠离子通道相关疾病。In another aspect, the present invention provides the use of the free crystalline form of the compound of formula I (e.g., free crystalline form A, free crystalline form B), a pharmaceutically acceptable salt of the compound of formula I (including its crystalline form) or the pharmaceutical composition in the preparation of a drug for treating and/or preventing voltage-gated sodium channel-related diseases.
根据本发明的实施方案,所述压门控钠离子通道相关疾病为Nav1.8相关疾病。According to an embodiment of the present invention, the voltage-gated sodium ion channel-related disease is a Nav1.8-related disease.
根据本发明的实施方案,利用本发明所述式I化合物游离态晶型(例如游离态晶型A、游离态晶型B)、式I化合物药学上可接受的盐(包括其晶型形式)或者所述的药物组合物,可以为有需要的患者提供更优、更有效的临床治疗药物或方案。According to an embodiment of the present invention, the free crystalline form of the compound of formula I (e.g., free crystalline form A, free crystalline form B), a pharmaceutically acceptable salt of the compound of formula I (including its crystalline form) or the pharmaceutical composition described in the present invention can provide patients in need with better and more effective clinical treatment drugs or regimens.
本发明还提供治疗和/或预防与Nav1.8相关疾病的方法,该方法包括向患者施用治疗有效剂量的包含如上所述式I化合物的晶型、如上所述式I化合物药学上可接受的盐、如上所述盐的晶型、或者如上所述药物组合物;优选地,包括如上所述式I化合物游离态晶型(例如游离态晶型A、游离酸晶型B)、式I化合物药学上可接受的盐(包括其晶型形式,例如马来酸盐晶型A、钠盐晶型A)或者所述的药物组合物的药物制剂。The present invention also provides a method for treating and/or preventing diseases related to Nav1.8, which comprises administering to a patient a therapeutically effective dose of a crystalline form of the compound of formula I as described above, a pharmaceutically acceptable salt of the compound of formula I as described above, a crystalline form of the salt as described above, or a pharmaceutical composition as described above; preferably, a pharmaceutical preparation comprising a free crystalline form of the compound of formula I as described above (e.g., free crystalline form A, free acid crystalline form B), a pharmaceutically acceptable salt of the compound of formula I (including its crystalline form, such as maleate crystalline form A, sodium salt crystalline form A) or the pharmaceutical composition.
根据本发明的实施方案,所述Nav1.8相关疾病包括:疼痛。According to an embodiment of the present invention, the Nav1.8-related diseases include: pain.
优选地,所述疼痛包括:急性疼痛、慢性疼痛、炎性疼痛、癌症疼痛、神经性疼痛、肌肉骨骼痛、原发性疼痛、肠痛和特发性疼痛。Preferably, the pain includes: acute pain, chronic pain, inflammatory pain, cancer pain, neuropathic pain, musculoskeletal pain, primary pain, intestinal pain and idiopathic pain.
术语定义和解释Definitions and Explanations of Terms
本发明使用的各种术语和短语具有本领域技术人员公知的一般含义,即便如此,本发明仍然希望在此对这些术语和短语作更详尽的说明和解释,提及的术语和短语如有与公知含义不一致的,以本发明所表述的含义为准。The various terms and phrases used in the present invention have general meanings known to those skilled in the art. Even so, the present invention still hopes to provide a more detailed description and explanation of these terms and phrases herein. If the mentioned terms and phrases are inconsistent with the known meanings, the meanings expressed in the present invention shall prevail.
除非另有说明,本说明书和权利要求书记载的数值范围相当于至少记载了其中每一个具体的整数数值。例如,两种以上代表2、3、4、5、6、7、8、9、10或更多种。当某些数值范围被定义或理解为“数”时,应当理解为记载了该范围的两个端点、该范围内的每一个整数以及该范围内的每一个小数。例如,“0~10的数”应当理解为不仅记载了0、1、2、3、4、5、6、7、8、9和10的每一个整数,还至少记载了其中每一个整数分别与0.1、0.2、0.3、0.4、0.5、0.6、0.7、0.8、0.9的和。Unless otherwise stated, the numerical ranges recorded in this specification and claims are equivalent to recording at least each specific integer value therein. For example, two or more represent 2, 3, 4, 5, 6, 7, 8, 9, 10 or more. When certain numerical ranges are defined or understood as "numbers", they should be understood to record the two endpoints of the range, each integer within the range, and each decimal within the range. For example, "a number from 0 to 10" should be understood to record not only each integer of 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 and 10, but also at least the sum of each integer therein and 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, respectively.
本说明书和权利要求书记载“约”某个数值时,包括了该数值本身,以及本领域可接受的该数值前后范围内的数值,例如该数值±15%范围内的数值,该数值±10%范围内的数值,该数值±5%范围内的数值等。例如,约10,代表了包括:10±1.5范围内、即8.5~11.5范围内的数值;10±1.0范围内、即9.0~11.0范围内的数值;以及10±0.5范围内、即9.5~10.5范围内的数值。When the description and claims record "about" a certain value, it includes the value itself, as well as the values within the range of the value acceptable in the art, such as the values within the range of ±15% of the value, the values within the range of ±10%, the values within the range of ±5%, etc. For example, about 10 represents: values within the range of 10±1.5, that is, values within the range of 8.5 to 11.5; values within the range of 10±1.0, that is, values within the range of 9.0 to 11.0; and values within the range of 10±0.5, that is, values within the range of 9.5 to 10.5.
本公开所述的“2θ或2θ角度”是指衍射角,θ为布拉格角,单位为°或度;每个特征峰2θ的误差范围为±0.20(优选为±0.10)(包括超过2位小数的数字经过四舍五入后的情况)。The "2θ or 2θ angle" mentioned in the present disclosure refers to the diffraction angle, θ is the Bragg angle, and the unit is ° or degree; the error range of each characteristic peak 2θ is ±0.20 (preferably ±0.10) (including the case where the numbers exceeding 2 decimal places are rounded off).
本发明的式I化合物的盐、多晶型物可以与其它活性成分组合使用,只要它不产生其他不利作用,例如过敏反应。The salts and polymorphs of the compound of formula I of the present invention may be used in combination with other active ingredients as long as it does not cause other adverse effects such as allergic reactions.
本发明所用的术语“组合物”意指包括包含指定量的各指定成分的产品,以及直接或间接从指定量的各指定成分的组合产生的任何产品。As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
术语“患者”是指包括哺乳动物在内的任何动物,优选小鼠、大鼠、其它啮齿类动物、兔、狗、猫、猪、牛、羊、马或灵长类动物,最优选人。The term "patient" refers to any animal including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, pigs, cows, sheep, horses or primates, and most preferably humans.
术语“治疗有效剂量”是指研究人员、兽医、医师或其他临床医师正在组织、系统、动物、个体或人中寻找的引起生物学或医学反应的活性化合物或药物的量,它包括以下一项或多项:(1)预防疾病:例如在易感染疾病、紊乱或病症但尚未经历或出现疾病病理或症状的个体中预防疾病、紊乱或病症;(2)抑制疾病:例如在正经历或出现疾病、紊乱或病症的病理或症状的个体中抑制疾病、紊乱或病症(即阻止病理和/或症状的进一步发展);(3)缓解疾病:例如在正经历或出现疾病、紊乱或病症的病理或症状的个体中缓解疾病、紊乱或病症(即逆转病理和/或症状)。The term "therapeutically effective dose" refers to the amount of an active compound or drug that elicits the biological or medical response that a researcher, veterinarian, physician or other clinician is seeking in a tissue, system, animal, individual or human, and includes one or more of the following: (1) preventing disease: for example, preventing a disease, disorder or condition in an individual who is susceptible to the disease, disorder or condition but has not yet experienced or developed the pathology or symptoms of the disease; (2) inhibiting disease: for example, inhibiting the disease, disorder or condition in an individual who is experiencing or developing the pathology or symptoms of the disease, disorder or condition (i.e., preventing further development of the pathology and/or symptoms); (3) alleviating disease: for example, alleviating the disease, disorder or condition in an individual who is experiencing or developing the pathology or symptoms of the disease, disorder or condition (i.e., reversing the pathology and/or symptoms).
术语“药学上可接受的”是指处方组分或活性成分对一般治疗目标的健康没有过分的有害影响。The term "pharmaceutically acceptable" means that the formulation components or active ingredients have no undue adverse effect on health for the general purpose of treatment.
术语“药学上可以接受的赋形剂或载体”意指一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能与本发明的化合物以及它们之间相互掺和,而不明显降低化合物的药效。药理上可以接受的赋形剂或载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等))、乳化剂、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。所述药物组合物可特别专门配制成以固体或液体形式供口服给药、供胃肠外注射或供直肠给药。所述的药物组合物可配制成多种剂型,便于给药,例如,口服制剂(如片剂、胶囊剂、溶液或混悬液),可注射的制剂(如可注射的溶液或混悬液,或者是可注射的干燥粉末,在注射前加入药物溶媒后可立即使用)。The term "pharmaceutically acceptable excipient or carrier" means one or more compatible solid or liquid fillers or gel substances, which are suitable for human use and must have sufficient purity and sufficiently low toxicity. "Compatibility" here means that the components in the composition can be mixed with the compounds of the present invention and with each other without significantly reducing the efficacy of the compounds. Some examples of pharmacologically acceptable excipients or carriers include cellulose and its derivatives (such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers, wetting agents (such as sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc. The pharmaceutical composition can be specially formulated in solid or liquid form for oral administration, for parenteral injection or for rectal administration. The pharmaceutical composition can be formulated into a variety of dosage forms for easy administration, for example, oral preparations (such as tablets, capsules, solutions or suspensions), injectable preparations (such as injectable solutions or suspensions, or injectable dry powders that can be used immediately after adding a pharmaceutical solvent before injection).
当用于上述治疗和/或预防用途时,本发明式I化合物的盐、多晶型物和药物组合物的总日用量须由主诊医师在可靠的医学判断范围内作出决定。对于任何具体的患者,具体的治疗有效剂量水平须根据多种因素而定,所述因素包括所治疗的障碍和该障碍的严重程度;所采用的具体化合物的活性;所采用的具体组合物;患者的年龄、体重、一般健康状况、性别和饮食;所采用的具体化合物的给药时间、给药途径和排泄率;治疗持续时间;与所采用的具体化合物组合使用或同时使用的药物;及医疗领域公知的类似因素。例如,本领域的做法是,化合物的剂量从低于为得到所需治疗效果而要求的水平开始,逐渐增加剂量,直到得到所需的效果。When used for the above-mentioned treatment and/or prevention purposes, the total daily dosage of the salts, polymorphs and pharmaceutical compositions of the compounds of formula I of the present invention shall be determined by the attending physician within the scope of sound medical judgment. For any particular patient, the specific therapeutically effective dosage level shall be determined based on a variety of factors, including the disorder being treated and the severity of the disorder; the activity of the specific compound used; the specific composition used; the patient's age, weight, general health, sex and diet; the administration time, route of administration and excretion rate of the specific compound used; the duration of treatment; drugs used in combination or concurrently with the specific compound used; and similar factors known in the medical field. For example, it is practiced in the art to start the dosage of the compound from a level lower than that required to obtain the desired therapeutic effect and gradually increase the dosage until the desired effect is obtained.
术语“游离态”是指化合物未进一步成盐的形式,如本文中对应式I化合物本身。本领域技术人员可以理解,当“游离态”和某种酸成盐,则该“游离态”可以理解为该酸形成的盐对应的“游离碱”。即“游离碱”和“游离态”具有等同含义,同理,“游离酸”也可与“游离态”具有等同含义(当游离态与碱成盐时)。The term "free state" refers to a form of a compound that has not been further salted, such as the compound of Formula I itself herein. It will be understood by those skilled in the art that when the "free state" forms a salt with a certain acid, the "free state" can be understood as the "free base" corresponding to the salt formed by the acid. That is, "free base" and "free state" have equivalent meanings, and similarly, "free acid" can also have equivalent meanings to "free state" (when the free state forms a salt with a base).
本发明提供了式I化合物的盐,式I化合物及其盐的晶型,具有良好的药用性质。The present invention provides salts of compounds of formula I, and crystal forms of compounds of formula I and their salts, which have good pharmaceutical properties.
图1-1为式I化合物游离态晶型A的XRPD图;Figure 1-1 is an XRPD diagram of free form A of the compound of formula I;
图1-2为式I化合物游离态晶型A的DSC图;Figure 1-2 is a DSC diagram of the free form A of the compound of formula I;
图1-3为式I化合物游离态晶型A的TGA图;Figure 1-3 is a TGA diagram of the free form A of the compound of formula I;
图2-1为式I化合物游离态晶型B的XRPD图;Figure 2-1 is an XRPD diagram of free form B of the compound of formula I;
图2-2为式I化合物游离态晶型B的DSC图;Figure 2-2 is a DSC diagram of the free form B of the compound of formula I;
图2-3为式I化合物游离态晶型B的TGA图;Figure 2-3 is a TGA diagram of the free form B of the compound of formula I;
图3-1马来酸盐晶型A的XRPD图Figure 3-1 XRPD diagram of maleate salt form A
图3-2马来酸盐晶型A的TGA/DSC图Figure 3-2 TGA/DSC graph of maleate crystal form A
图3-3马来酸盐晶型A的1H NMR图Figure 3-3 1 H NMR graph of maleate salt form A
图4-1钠盐晶型A的XRPD图Figure 4-1 XRPD diagram of sodium salt form A
图4-2钠盐晶型A的TGA/DSC图Figure 4-2 TGA/DSC diagram of sodium salt form A
图4-3钠盐晶型A的1H NMR图Figure 4-3 1 H NMR spectrum of sodium salt form A
图5 37℃动态溶解度曲线图Figure 5 Dynamic solubility curve at 37℃
图6-1游离态晶型A在H2O中溶解度样品的XRPD叠图Figure 6-1 XRPD overlay of solubility samples of free form A in H 2 O
图6-2游离态晶型A在SGF中溶解度样品的XRPD叠图Figure 6-2 XRPD overlay of solubility samples of free form A in SGF
图6-3游离态晶型A在FaSSIF中溶解度样品的XRPD叠图Figure 6-3 XRPD overlay of solubility samples of free form A in FaSSIF
图6 4游离态晶型A在FeSSIF中溶解度样品的XRPD叠图Figure 6 XRPD overlay of solubility samples of free form A in FeSSIF
图7-1游离态晶型A的DVS图Figure 7-1 DVS diagram of free crystal form A
图7-2游离态晶型A在DVS测试前后的XRPD叠图Figure 7-2 XRPD overlay of free form A before and after DVS test
图7-3马来酸盐晶型A的DVS图Figure 7-3 DVS diagram of maleate crystal form A
图7-4马来酸盐晶型A在DVS测试前后的XRPD叠图Figure 7-4 XRPD overlay of maleate crystal form A before and after DVS test
图8-1游离态晶型A稳定性评估样品的XRPD叠图Figure 8-1 XRPD overlay of the stability evaluation sample of free form A
图8-2马来酸盐晶型A稳定性评估样品的XRPD叠图Figure 8-2 XRPD overlay of maleate crystal form A stability evaluation sample
下文将结合具体实施例对本发明的技术方案做更进一步的详细说明。应当理解,下列实施例仅为示例性地说明和解释本发明,而不应被解释为对本发明保护范围的限制。凡基于本发明上述内容所实现的技术均涵盖在本发明旨在保护的范围内。The technical scheme of the present invention will be further described in detail below in conjunction with specific embodiments. It should be understood that the following embodiments are only exemplary descriptions and explanations of the present invention and should not be construed as limiting the scope of protection of the present invention. All technologies implemented based on the above content of the present invention are included in the scope that the present invention is intended to protect.
除非另有说明,以下实施例中使用的原料和试剂均为市售商品,或者可以通过已知方法制备。Unless otherwise specified, the raw materials and reagents used in the following examples are commercially available or can be prepared by known methods.
本发明所采用的仪器和检测方法如下:The instrument and detection method adopted by the present invention are as follows:
一、X射线粉末衍射(XRPD)1. X-ray powder diffraction (XRPD)
XRPD图在Bruker、PANalytacal生产的X射线粉末衍射分析仪上采集,扫描参数分别如下表A-1a和A-1b所示。The XRPD pattern was collected on an X-ray powder diffraction analyzer produced by Bruker and PANalytacal, and the scanning parameters are shown in Tables A-1a and A-1b below, respectively.
表A-1a XRPD测试参数
Table A-1a XRPD test parameters
表A-1b XRPD测试参数
Table A-1b XRPD test parameters
二、热重分析(TGA)和差示扫描量热(DSC)2. Thermogravimetric analysis (TGA) and differential scanning calorimetry (DSC)
2.1 TGA和DSC图分别在TA Q500热重分析仪和TA Q2000差示扫描量热仪上采集,下表A-2a列出了测试参数。2.1 TGA and DSC images were collected on a TA Q500 thermogravimetric analyzer and a TA Q2000 differential scanning calorimeter, respectively. The test parameters are listed in Table A-2a below.
表A-2a DSC和TGA测试参数
Table A-2a DSC and TGA test parameters
2.2 TGA和DSC图分别在TA 5500热重分析仪和TA 2500差示扫描量热仪上采集,下表A-2b列出了测试参数。2.2 TGA and DSC graphs were collected on a TA 5500 thermogravimetric analyzer and a TA 2500 differential scanning calorimeter, respectively. The test parameters are listed in Table A-2b below.
表A-2b TGA和DSC测试参数
Table A-2b TGA and DSC test parameters
三、动态水分吸附(DVS)3. Dynamic moisture adsorption (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(NO3)2和KCl的潮解点校正。DVS测试参数列于表A-3。Dynamic moisture sorption (DVS) curves were collected on the DVS IntrInsic of SMS (Surface Measurement Systems). The relative humidity at 25°C was calibrated using the deliquescent points of LiCl, Mg(NO 3 ) 2 and KCl. The DVS test parameters are listed in Table A-3.
表A-3 DVS测试参数
Table A-3 DVS test parameters
四、液态核磁(1H NMR)4. Liquid Nuclear Magnetics ( 1H NMR)
液态核磁谱图在Bruker 400M核磁共振仪上采集,以DMSO-d6作为溶剂。Liquid-state NMR spectra were collected on a Bruker 400M NMR spectra using DMSO- d6 as solvent.
五、超高效液相色谱和离子色谱(UPLC/IC)5. Ultra-high performance liquid chromatography and ion chromatography (UPLC/IC)
试验中纯度测试、动态溶解度和稳定性测试是由沃特世H-Class超高效液相色谱仪测试,离子的成盐摩尔比测试是由离子色谱测试,分析条件如表A-4和表A-5所示。In the experiment, the purity test, dynamic solubility and stability test were performed by Waters H-Class ultra high performance liquid chromatograph, and the ion salt formation molar ratio test was performed by ion chromatography. The analysis conditions are shown in Table A-4 and Table A-5.
表A-4高效液相色谱测试条件
Table A-4 HPLC test conditions
表A-5离子色谱测试条件
Table A-5 Ion chromatography test conditions
六、生物溶媒的配制6. Preparation of Biosolvent
模拟胃液的配制(SGF)Preparation of simulated gastric fluid (SGF)
称取100mg NaCl和50mg曲纳通X-100至50-mL容量瓶中,加入纯化水溶清。加入816μL 1M盐酸,用1M的盐酸或1M的NaOH溶液调节pH至1.8。加纯化水定容。Weigh 100 mg NaCl and 50 mg Trinaton X-100 into a 50-mL volumetric flask and add purified water to dissolve. Add 816 μL 1 M hydrochloric acid and adjust the pH to 1.8 with 1 M hydrochloric acid or 1 M NaOH solution. Add purified water to volume.
模拟禁食状态肠液的配制(FaSSIF)Preparation of fasting-simulated intestinal fluid (FaSSIF)
称取340mg无水NaH2PO4和620mg NaCl至100-mL容量瓶中。加入纯化水溶清,后加入55.44μL 50%的NaOH溶液,用1M的盐酸或1M的NaOH溶液调节pH至6.5。加纯化水定容。后称110mg SIF粉末至50-mL容量瓶中,用上述溶液溶清并定容。Weigh 340 mg of anhydrous NaH 2 PO 4 and 620 mg of NaCl into a 100-mL volumetric flask. Add purified water to dissolve, then add 55.44 μL of 50% NaOH solution, and adjust the pH to 6.5 with 1M hydrochloric acid or 1M NaOH solution. Add purified water to make up to volume. Then weigh 110 mg of SIF powder into a 50-mL volumetric flask, dissolve with the above solution and make up to volume.
模拟喂食状态肠液的配制(FeSSIF)Preparation of simulated fed intestinal fluid (FeSSIF)
取0.82mL冰醋酸,1.18g NaCl至100-mL容量瓶中,加入纯化水溶清,后加入528μL 50%的NaOH溶液,用1M的盐酸或1M的NaOH溶液调节pH至5.0,加纯化水定容。后称560mg SIF粉末至50-mL容量瓶中,用上述溶液溶清并定容。Take 0.82 mL of glacial acetic acid and 1.18 g of NaCl into a 100-mL volumetric flask, add purified water to dissolve, then add 528 μL of 50% NaOH solution, adjust the pH to 5.0 with 1 M hydrochloric acid or 1 M NaOH solution, and add purified water to make up to volume. Then weigh 560 mg of SIF powder into a 50-mL volumetric flask, dissolve with the above solution and make up to volume.
七、本发明使用的试剂如下表A-6VII. The reagents used in the present invention are shown in Table A-6 below
表A-6试验中选用溶剂的中英文名称对照表
Table A-6 Comparison table of Chinese and English names of solvents used in the test
实施例1:式I化合物的制备Example 1: Preparation of Compounds of Formula I
5-(4,5-二氯-2-(4-(三氟甲氧基)苯氧基)苯甲酰氨基)嘧啶1-氧化物
5-(4,5-dichloro-2-(4-(trifluoromethoxy)phenoxy)benzamido)pyrimidine 1-oxide
式I化合物的合成路线如下所示:
The synthetic route of the compound of formula I is as follows:
第一步:4,5-二氯-2-(4-(三氟甲氧基)苯氧基)苯甲酸的合成
Step 1: Synthesis of 4,5-dichloro-2-(4-(trifluoromethoxy)phenoxy)benzoic acid
室温下,将4,5-二氯-2-氟苯甲酸(1.0g,4.78mmol),碳酸铯(4.68g,14.35mmol)和4-(三氟甲氧基)苯酚(8mL)加入到20毫升微波管中,密封微波管,在150℃下反应1小时。冷却至室温,向反应液中加入水(10mL),用EtOAc(20mL×3)萃取,有机相用饱和食盐水(20mL)洗1次,并用无水硫酸钠干燥,过滤,减压浓缩旋干,粗品经柱层析(SiO2;PE:EA=10:1)纯化得4,5-二氯-2-(4-(三氟甲氧基)苯氧基)苯甲酸(3)(1.0g;产率56.9%)。At room temperature, 4,5-dichloro-2-fluorobenzoic acid (1.0 g, 4.78 mmol), cesium carbonate (4.68 g, 14.35 mmol) and 4-(trifluoromethoxy)phenol (8 mL) were added to a 20 mL microwave tube, which was sealed and reacted at 150°C for 1 hour. After cooling to room temperature, water (10 mL) was added to the reaction solution, and the mixture was extracted with EtOAc (20 mL×3). The organic phase was washed once with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure and dried, and the crude product was purified by column chromatography (SiO 2 ; PE:EA=10:1) to obtain 4,5-dichloro-2-(4-(trifluoromethoxy)phenoxy)benzoic acid (3) (1.0 g; yield 56.9%).
第二步:4,5-二氯-N-(嘧啶-5-基)-2-(4-(三氟甲氧基)苯氧基)苯甲酰胺的合成
Step 2: Synthesis of 4,5-dichloro-N-(pyrimidin-5-yl)-2-(4-(trifluoromethoxy)phenoxy)benzamide
将4,5-二氯-2-(4-(三氟甲氧基)苯氧基)苯甲酸(1.0g,2.72mmol),5-氨基嘧啶(310.88mg,3.27mmol)和HATU(2.07g,5.45mmol)加入到DMF(10mL)中,然后加入DIPEA(1.06g,8.17mmol),加完后,反应液在室温下反应16小时。LC-MS显示反应完成。向反应液中加入NH4Cl溶液(15mL),用EtOAc(20mL×3)萃取,合并有机相,分别用水(20mL×2)和饱和食盐水(10mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液浓缩旋干,粗品通过正相硅胶柱层析分离(SiO2;EtOAc/PE=1:1)得到4,5-二氯-N-(嘧啶-5-基)-2-(4-(三氟甲氧基)苯氧基)苯甲酰胺(1.10g;产率90.9%)。4,5-Dichloro-2-(4-(trifluoromethoxy)phenoxy)benzoic acid (1.0 g, 2.72 mmol), 5-aminopyrimidine (310.88 mg, 3.27 mmol) and HATU (2.07 g, 5.45 mmol) were added to DMF (10 mL), and then DIPEA (1.06 g, 8.17 mmol) was added. After the addition was complete, the reaction solution was reacted at room temperature for 16 hours. LC-MS showed that the reaction was complete. NH 4 Cl solution (15 mL) was added to the reaction solution, and the mixture was extracted with EtOAc (20 mL×3). The organic phases were combined, washed with water (20 mL×2) and saturated brine (10 mL), respectively. The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated and dried by spin drying. The crude product was separated by normal phase silica gel column chromatography (SiO 2 ; EtOAc/PE=1:1) to give 4,5-dichloro-N-(pyrimidin-5-yl)-2-(4-(trifluoromethoxy)phenoxy)benzamide (1.10 g; yield 90.9%).
第三步:5-(4,5-二氯-2-(4-(三氟甲氧基)苯氧基)苯甲酰氨基)嘧啶1-氧化物(式I化合物)
Step 3: 5-(4,5-dichloro-2-(4-(trifluoromethoxy)phenoxy)benzamido)pyrimidine 1-oxide (compound of formula I)
室温下,分别称取4,5-二氯-N-(嘧啶-5-基)-2-(4-(三氟甲氧基)苯氧基)苯甲酰胺(1.0g,2.09mmol)于DCM(10mL)中,再缓慢加入m-CPBA(849.89mg,4.19mmol;85%)。反应液在室温条件下反应16小时。反应完成后,反应液用二氯甲烷(20mL)稀释,然后用饱和碳酸氢钠水溶液(15mL×2)洗涤,有机相用Na2SO4干燥,浓缩后送高压液相色谱制备(氨水-乙腈)得到5-(4,5-二氯-2-(4-(三氟甲氧基)苯氧基)苯甲酰氨基)嘧啶1-氧化物(86.6mg,产率8.38%)。At room temperature, 4,5-dichloro-N-(pyrimidin-5-yl)-2-(4-(trifluoromethoxy)phenoxy)benzamide (1.0 g, 2.09 mmol) was weighed into DCM (10 mL), and then m-CPBA (849.89 mg, 4.19 mmol; 85%) was slowly added. The reaction solution was reacted at room temperature for 16 hours. After the reaction was completed, the reaction solution was diluted with dichloromethane (20 mL), and then washed with saturated sodium bicarbonate aqueous solution (15 mL×2). The organic phase was dried over Na 2 SO 4 , concentrated, and sent to high pressure liquid chromatography (ammonia-acetonitrile) to obtain 5-(4,5-dichloro-2-(4-(trifluoromethoxy)phenoxy)benzamido)pyrimidine 1-oxide (86.6 mg, yield 8.38%).
1H NMR(400MHz,DMSO):δ8.85-8.84(m,2H),8.43-8.42(m,1H),8.04(s,1H),7.45(s,1H),7.40-7.37(m,2H),7.20-7.18(m,2H)。 1 H NMR (400MHz, DMSO): δ8.85-8.84(m,2H),8.43-8.42(m,1H),8.04(s,1H),7.45(s,1H),7.40-7.37(m,2H),7.20-7.18(m,2H).
LC-MS,M/Z(ESI):458.1[M-H]-。LC-MS,M/Z(ESI):458.1[MH] - .
实施例2:气固扩散法制备晶型Example 2: Preparation of crystal form by gas-solid diffusion method
称取大约40mg的式I化合物分别放入2mL的透明样品瓶中,另分别在20mL样品瓶加入表1-1中的溶剂体系(3mL),将2mL透明样品瓶敞口置于20mL样品瓶中,密封20mL样品瓶并于室温下静置,过滤得到固体样品。室温真空干燥后,对样品进行XRPD测试。结果表明,在表1-1的溶剂体系下,均得到了游离态晶型A。Weigh about 40 mg of the compound of formula I and put it into a 2 mL transparent sample bottle, add the solvent system (3 mL) in Table 1-1 to the 20 mL sample bottle, place the 2 mL transparent sample bottle open in the 20 mL sample bottle, seal the 20 mL sample bottle and let it stand at room temperature, and filter to obtain a solid sample. After vacuum drying at room temperature, the sample was subjected to XRPD testing. The results show that free form A was obtained under the solvent system in Table 1-1.
表1-1:气固扩散法晶型筛选结果
Table 1-1: Crystal screening results of gas-solid diffusion method
实施例3:气液扩散法制备晶型Example 3: Preparation of crystal form by gas-liquid diffusion method
称取大约40mg的式I化合物分别放入2mL的透明样品瓶中,在室温下分别向2mL样品瓶中加入少量的表2中的良溶剂,震荡使样品充分溶解,得到的溶液用0.22μm的滤膜过滤至另一2mL透明样品瓶中。另取20mL样品瓶向其中加入4mL的表2中对应的反溶剂,将装有过滤后澄清溶液的2mL透明样品瓶敞口置于该20mL样品瓶中,密封20mL样品瓶并于室温下静置以析出固体。室温真空干燥后,对样品进行XRPD测试。结果表明,在表1-2的溶剂体系下,均得到了游离态晶型A。当良溶剂为DMSO,反溶剂为H2O时,得到了游离态晶型A和B的混合物。Weigh about 40 mg of the compound of formula I and put them into 2 mL transparent sample bottles respectively. Add a small amount of good solvents in Table 2 to the 2 mL sample bottles at room temperature, shake to fully dissolve the samples, and filter the obtained solution into another 2 mL transparent sample bottle with a 0.22 μm filter membrane. Take another 20 mL sample bottle and add 4 mL of the corresponding anti-solvent in Table 2 thereto. Place the 2 mL transparent sample bottle containing the clear solution after filtration into the 20 mL sample bottle with an open mouth. Seal the 20 mL sample bottle and let it stand at room temperature to precipitate solids. After vacuum drying at room temperature, perform XRPD test on the sample. The results show that free crystalline form A is obtained under the solvent system of Table 1-2. When the good solvent is DMSO and the anti-solvent is H 2 O, a mixture of free crystalline forms A and B is obtained.
表1-2:气液扩散法晶型筛选结果
Table 1-2: Crystal screening results of gas-liquid diffusion method
实施例4:室温混悬法制备晶型Example 4: Preparation of crystal form by room temperature suspension method
称取大约50mg的式I化合物分别放入2mL的透明样品瓶中,在室温下分别向样品瓶中加入0.1-0.5mL的表3中的溶剂,磁力搅拌约两周,过滤得到固体样品。室温真空干燥后,对固体样品进行XRPD测试。结果表明,在表1-3的溶剂体系下,均得到了游离态晶型A。选取在溶剂EtOH下得到的游离态晶型A进行XRPD、TGA、DSC测试,其XRPD图谱如图1-1所示,XRPD衍射峰数据如表1-4所示;DSC图如图1-2所示,TGA图如图1-3所示。Weigh about 50 mg of the compound of formula I and put them into 2 mL transparent sample bottles respectively, add 0.1-0.5 mL of the solvent in Table 3 to the sample bottles at room temperature, stir magnetically for about two weeks, and filter to obtain a solid sample. After vacuum drying at room temperature, the solid sample was subjected to XRPD test. The results show that free crystalline form A was obtained under the solvent system of Table 1-3. The free crystalline form A obtained in the solvent EtOH was selected for XRPD, TGA, and DSC tests, and its XRPD spectrum is shown in Figure 1-1, and the XRPD diffraction peak data is shown in Table 1-4; the DSC graph is shown in Figure 1-2, and the TGA graph is shown in Figure 1-3.
表1-3:室温混悬法晶型筛选结果
Table 1-3: Room temperature suspension method crystal screening results
表1-4:游离态晶型A的XRPD衍射峰数据
Table 1-4: XRPD diffraction peak data of free form A
实施例5:50℃混悬法制备晶型Example 5: Preparation of crystal form by 50°C suspension method
分别向2mL的透明样品瓶中称取大约50mg的式I化合物,在50℃下分别向样品瓶中加入表1-5中的0.1-0.5mL溶剂,磁力搅拌约一周,过滤得到固体样品。室温真空干燥后,对样品进行XRPD测试。结果表明,在表1-5的溶剂体系下,均得到了游离态晶型A。Weigh approximately 50 mg of the compound of formula I into a 2 mL transparent sample bottle, add 0.1-0.5 mL of the solvent in Table 1-5 to the sample bottle at 50°C, stir magnetically for about one week, and filter to obtain a solid sample. After vacuum drying at room temperature, the sample was subjected to XRPD testing. The results show that free crystalline form A was obtained in the solvent system in Table 1-5.
表1-5:50℃混悬晶型筛选结果
Table 1-5: Screening results of 50℃ suspension crystals
实施例6:缓慢挥发法制备晶型Example 6: Preparation of crystal form by slow evaporation method
分别向2mL的透明样品瓶中称取大约40mg的式I化合物,分别向样品瓶中加入表1-6中2-7mL的溶剂,得到的溶液用0.22μm的滤膜过滤,过滤后的澄清溶液放置在室温条件下缓慢挥发,得到固体。室温真空干燥后,对样品进行XRPD测试。结果表明,在表1-6的溶剂体系下,均得到了游离态晶型A。当溶剂为1,4-dioxane时,得到了游离态晶型B和少量游离态晶型A的混合物。Weigh approximately 40 mg of the compound of formula I into a 2 mL transparent sample bottle, add 2-7 mL of the solvent in Table 1-6 to the sample bottle, filter the resulting solution with a 0.22 μm filter membrane, and slowly volatilize the filtered clear solution at room temperature to obtain a solid. After vacuum drying at room temperature, the sample was subjected to XRPD testing. The results show that free crystalline form A was obtained under the solvent system in Table 1-6. When the solvent is 1,4-dioxane, a mixture of free crystalline form B and a small amount of free crystalline form A was obtained.
表1-6:缓慢挥发法晶型筛选结果
Table 1-6: Slow evaporation method crystal screening results
实施例7:反溶剂法制备晶型Example 7: Preparation of Crystalline Form by Anti-Solvent Method
分别向2mL的透明样品瓶中称取大约40mg的式I化合物,在室温下用少量的良溶剂将样品溶解,得到的溶液用0.22μm的滤膜过滤。然后将2-4倍的反溶剂缓慢加入到过滤后的澄清溶液中直到有固体析出,过滤得到固体样品。室温真空干燥后,对样品进行XRPD测试。结果表明,在表1-7的溶剂体系下,得到了游离态晶型A和游离态晶型B。Weigh approximately 40 mg of the compound of formula I into a 2 mL transparent sample bottle, dissolve the sample with a small amount of good solvent at room temperature, and filter the resulting solution with a 0.22 μm filter membrane. Then slowly add 2-4 times the anti-solvent to the filtered clear solution until solids precipitate, and filter to obtain a solid sample. After vacuum drying at room temperature, the sample was subjected to XRPD testing. The results show that free crystalline form A and free crystalline form B were obtained under the solvent system of Table 1-7.
表1-7:反溶剂法晶型筛选结果
Table 1-7: Anti-solvent method crystal screening results
实施例8:反反溶剂法制备晶型Example 8: Preparation of Crystalline Form by Anti-Anti-Solvent Method
分别向2mL的透明样品瓶中称取大约40mg的式I化合物,在室温下用少量的良溶剂将样品溶解,得到的溶液用0.22μm的滤膜过滤。然后将过滤后的澄清溶液迅速加到2-4倍的反溶剂中,析出固体,过滤得到固体样品。室温真空干燥后,对样品进行XRPD测试。结果表明,在表1-8的溶剂体系下,得到了游离态晶型A和游离态晶型B。当良溶剂为1,4-dioxane,反溶剂为H2O时,得到了游离态晶型B和少量游离态晶型A的混合物。Weigh approximately 40 mg of the compound of formula I into a 2 mL transparent sample bottle, dissolve the sample with a small amount of good solvent at room temperature, and filter the resulting solution with a 0.22 μm filter membrane. Then quickly add the filtered clear solution to 2-4 times the anti-solvent, precipitate the solid, and filter to obtain a solid sample. After vacuum drying at room temperature, the sample was subjected to XRPD testing. The results show that free crystalline form A and free crystalline form B were obtained under the solvent system of Table 1-8. When the good solvent is 1,4-dioxane and the anti-solvent is H 2 O, a mixture of free crystalline form B and a small amount of free crystalline form A was obtained.
表1-8:反反溶剂法晶型筛选结果
Table 1-8: Anti-antisolvent method crystal screening results
实施例9:冷却法制备晶型Example 9: Preparation of crystal form by cooling method
分别向2mL的透明样品瓶中称取大约40mg的式I化合物,在50℃下用少量的溶剂将样品溶解,得到的溶液用0.22μm的滤膜过滤。磁力搅拌下将过滤后的澄清溶液放置于室温条件缓慢冷却,析出固体,过滤得到固体样品,室温真空干燥后,对样品进行XRPD测试。结果表明,在表1-9的溶剂体系下,得到了游离态晶型A和游离态晶型B。Weigh approximately 40 mg of the compound of formula I into a 2 mL transparent sample bottle, dissolve the sample with a small amount of solvent at 50°C, and filter the resulting solution with a 0.22 μm filter membrane. The filtered clear solution was slowly cooled at room temperature under magnetic stirring to precipitate a solid, and a solid sample was obtained by filtration. After vacuum drying at room temperature, the sample was subjected to XRPD testing. The results showed that free crystalline form A and free crystalline form B were obtained under the solvent system of Table 1-9.
表1-9:冷却法晶型筛选结果
Table 1-9: Cooling method crystal screening results
实施例10:研磨法制备晶型Example 10: Preparation of crystal form by grinding method
分别称取大约40mg的式I化合物于研钵中,在不同条件下研磨约10分钟。对研磨后的固体室温真空干燥后,对样品进行XRPD测试。结果表明,在表1-10的溶剂体系下,得到了含大量无定形的游离态晶型A。About 40 mg of the compound of formula I was weighed in a mortar and ground for about 10 minutes under different conditions. After the ground solid was dried under vacuum at room temperature, the sample was subjected to XRPD testing. The results showed that under the solvent system of Table 1-10, a large amount of amorphous free-state crystalline form A was obtained.
表1-10:研磨法晶型筛选结果
Table 1-10: Crystal screening results by grinding method
实施例11:游离态晶型B的制备Example 11: Preparation of Free Form B
游离态晶型B的制备过程如下:The preparation process of free crystal form B is as follows:
称取500mg的式I化合物到圆底烧瓶中。室温下向圆底烧瓶中加入6.25mL的THF,超声使样品溶解,得到的溶液用0.22μm的滤膜过滤到另一100mL圆底烧瓶中,在磁力搅拌下将18.75mL的heptane缓慢加入到过滤后的澄清溶液中,溶液中有白色固体析出,再加入约5mg游离态晶型B的晶种(实施例7反溶剂法THF/heptane得到)继续搅拌。室温下搅拌约三小时后抽滤得到白色固体,室温真空干燥后进行XRPD、TGA、DSC测试。游离态晶型B的XRPD图谱如图2-1所示,XRPD衍射峰数据如表1-12所示;DSC图如图2-2所示,TGA图如图2-3所示。Weigh 500 mg of the compound of formula I into a round-bottom flask. Add 6.25 mL of THF to the round-bottom flask at room temperature, dissolve the sample by ultrasound, and filter the resulting solution into another 100 mL round-bottom flask with a 0.22 μm filter membrane. Slowly add 18.75 mL of heptane to the filtered clear solution under magnetic stirring. A white solid precipitates in the solution, and then add about 5 mg of seed crystals of free form B (obtained by the anti-solvent method THF/heptane in Example 7) and continue stirring. After stirring at room temperature for about three hours, filter and obtain a white solid. After vacuum drying at room temperature, perform XRPD, TGA, and DSC tests. The XRPD spectrum of free form B is shown in Figure 2-1, and the XRPD diffraction peak data is shown in Table 1-12; the DSC graph is shown in Figure 2-2, and the TGA graph is shown in Figure 2-3.
表1-11:游离态晶型B的表征
Table 1-11: Characterization of free crystal form B
表1-12:实施例11游离态晶型B的XRPD衍射峰数据
Table 1-12: XRPD diffraction peak data of free form B of Example 11
实施例12盐型筛选Example 12 Salt type screening
以游离态晶型A样品为起始,选用13种配体(酸碱投料比为1:1)和3种溶剂共设置了39个盐型筛选试验。筛选试验具体步骤如下:称取约20mg游离态晶型A样品和等摩尔量的对应配体至HPLC小瓶中,加入0.5mL溶剂混合得到悬浊液,液体酸先用相应溶剂稀释后再与起始样品混合。室温悬浮搅拌约3天后,离心分离固体并于50℃下真空干燥。所得固体XRPD表征结果显示(表2-1),在盐型筛选试验中共得到2种盐型晶型样品,分别为马来酸盐晶型A和钠盐晶型A。分别对筛选得到的盐型样品进行TGA/DSC表征,并用1H NMR和HPLC/IC确定成盐摩尔比,盐型表征结果汇总于表2-2。Starting with the free form A sample, 13 ligands (acid-base feed ratio of 1:1) and 3 solvents were selected to set up a total of 39 salt screening tests. The specific steps of the screening test are as follows: weigh about 20 mg of the free form A sample and an equimolar amount of the corresponding ligand into an HPLC vial, add 0.5 mL of solvent to mix to obtain a suspension, and the liquid acid is diluted with the corresponding solvent before mixing with the starting sample. After stirring at room temperature for about 3 days, the solid is separated by centrifugation and vacuum dried at 50°C. The XRPD characterization results of the obtained solid show (Table 2-1) that a total of 2 salt form samples were obtained in the salt screening test, namely maleate form A and sodium salt form A. The screened salt samples were characterized by TGA/DSC, and the salt formation molar ratio was determined by 1 H NMR and HPLC/IC. The salt characterization results are summarized in Table 2-2.
表2-1盐型筛选试验结果总结
*:配体与游离态的投料摩尔比为1:1;#:观察到样品变成橙红色。
a:样品在室温搅拌后澄清,后转5℃搅拌;b:样品在5℃搅拌后澄清,后加入1.0mL n-heptane诱导析
晶;c:加入n-heptane诱导析晶后样品成胶,后转50℃~5℃温度循环;d:加入n-heptane诱导析晶后样品澄清,后转5℃搅拌;e:样品在5℃搅拌后仍澄清,后转室温挥发。Table 2-1 Summary of salt type screening test results
*: The molar ratio of the ligand to the free state was 1:1; # : The sample was observed to turn orange-red.
a : The sample was clarified after stirring at room temperature and then stirred at 5°C; b : The sample was clarified after stirring at 5°C and then 1.0 mL of n-heptane was added to induce crystallization; c : The sample became gel after adding n-heptane to induce crystallization and then the temperature cycle was performed from 50°C to 5°C; d : The sample was clarified after adding n-heptane to induce crystallization and then stirred at 5°C; e : The sample remained clarified after stirring at 5°C and then volatilized at room temperature.
表2-2筛选所得盐型表征结果汇总
#:由1H NMR或HPLC/IC计算所得;ND:未检测出。Table 2-2 Summary of the characterization results of the salt types obtained by screening
# : calculated by 1 H NMR or HPLC/IC; ND: not detected.
12.1马来酸盐晶型A12.1 Maleate Crystalline Form A
马来酸盐晶型A是通过将游离态晶型A样品和等摩尔量的马来酸于EtOAc中室温打浆3天后,离心分离固体样品于50℃真空干燥后获得。马来酸盐晶型A样品的XRPD和TGA/DSC结果分别列于图3-1和图3-2。TGA结果显示样品加热至120℃时有1.41%的失重,DSC结果显示样品在139.1℃(起点温度)观察一个吸热信号。1H NMR结果如图3-3所示,样品中马来酸与式I化合物摩尔比为1:1,未发现EtOAc溶剂残留。马来酸盐晶型A的XRPD衍射峰数据如下表2-3。Maleate Form A is obtained by slurrying a free Form A sample and an equimolar amount of maleic acid in EtOAc at room temperature for 3 days, centrifuging and vacuum drying the solid sample at 50°C. The XRPD and TGA/DSC results of the maleate Form A sample are shown in Figures 3-1 and 3-2, respectively. The TGA results show that the sample has a weight loss of 1.41% when heated to 120°C, and the DSC results show that an endothermic signal is observed at 139.1°C (starting point temperature). The 1 H NMR results are shown in Figure 3-3. The molar ratio of maleic acid to the compound of formula I in the sample is 1:1, and no residual EtOAc solvent is found. The XRPD diffraction peak data of maleate Form A are shown in Table 2-3.
表2-3马来酸盐晶型A的XRPD衍射峰数据
Table 2-3 XRPD diffraction peak data of maleate crystal form A
12.2钠盐晶型A的制备12.2 Preparation of Sodium Salt Form A
钠盐晶型A是通过将游离态晶型A样品和等摩尔量的氢氧化钠于2-MeTHF中室温打浆3天后,离心分离固体样品于50℃真空干燥后获得。钠盐晶型A样品的XRPD和TGA/DSC结果分别列于图4-1和图4-2。TGA结果显示样品加热至150℃时有2.75%的失重,DSC结果显示样品在191.0℃和215.0℃(峰值温度)有2个吸热峰。1H NMR结果如图4-3所示,样品中未发现2-MeTHF溶剂残留。HPLC/IC结果显示钠盐晶型A中Na+与式I化合物的摩尔比为1:1。钠盐晶型A的XRPD衍射峰数据如下表2-4。Sodium salt form A is obtained by slurrying a free form A sample and an equimolar amount of sodium hydroxide in 2-MeTHF at room temperature for 3 days, centrifuging and separating the solid sample, and vacuum drying at 50°C. The XRPD and TGA/DSC results of the sodium salt form A sample are listed in Figures 4-1 and 4-2, respectively. The TGA results show that the sample has a 2.75% weight loss when heated to 150°C, and the DSC results show that the sample has two endothermic peaks at 191.0°C and 215.0°C (peak temperature). The 1 H NMR results are shown in Figure 4-3, and no 2-MeTHF solvent residue was found in the sample. HPLC/IC results show that the molar ratio of Na + to the compound of formula I in sodium salt form A is 1:1. The XRPD diffraction peak data of sodium salt form A are shown in Table 2-4 below.
表2-4钠盐晶型A的XRPD衍射峰数据
Table 2-4 XRPD diffraction peak data of sodium salt form A
实施例13评估试验Example 13 Evaluation Test
13.1动态溶解度13.1 Dynamic Solubility
以10mg/mL(以游离态计)的固体投料浓度在37℃下旋转混合,并在不同时间点(1、2、4和24小时)测定各样品在H2O、SGF、FaSSIF和FeSSIF中的溶解度。每个时间点取样后经离心(10000rpm)过滤(0.45μmPTFE),测定滤液的HPLC浓度和pH值,离心后的固体样品测试XRPD。溶解度试验结果总结于表2-5,溶解度曲线见图5。XRPD结果显示游离态晶型A在动态溶解度测试后晶型不变(图6-1至图6-4)。The solid feed concentration was 10 mg/mL (in free form) and the mixture was mixed by rotation at 37°C. The solubility of each sample in H 2 O, SGF, FaSSIF and FeSSIF was measured at different time points (1, 2, 4 and 24 hours). After sampling at each time point, the samples were centrifuged (10000 rpm) and filtered (0.45 μm PTFE). The HPLC concentration and pH value of the filtrate were measured, and the solid samples after centrifugation were tested by XRPD. The solubility test results are summarized in Table 2-5, and the solubility curve is shown in Figure 5. The XRPD results show that the free form A does not change its crystalline form after the dynamic solubility test (Figure 6-1 to Figure 6-4).
表2-5 37℃动态溶解度测试结果总结
S:溶解度(mg/mL)。Table 2-5 Summary of 37℃ dynamic solubility test results
S: solubility (mg/mL).
13.2、引湿性13.2. Hygroscopicity
通过动态水分吸附仪(DVS)对游离态晶型A和马来酸盐晶型A进行引湿性评估。收集了25℃恒温条件下,随湿度变化(0%RH-95%RH)时,样品的质量变化百分比。DVS测试结果及DVS测试前后样品的XRPD结果如图7-1至图7-4所示。结果表明,游离态晶型A和马来酸盐晶型A在25℃/80%RH的水分吸附分别为0.1698%和0.224%,所有样品在DVS测试后晶型均不变,特别是游离态晶型A几乎无引湿性。The hygroscopicity of free crystalline form A and maleate crystalline form A was evaluated by dynamic moisture sorption instrument (DVS). The percentage change in mass of the samples was collected when the humidity changed (0% RH-95% RH) under a constant temperature of 25°C. The DVS test results and the XRPD results of the samples before and after the DVS test are shown in Figures 7-1 to 7-4. The results show that the moisture adsorption of free crystalline form A and maleate crystalline form A at 25°C/80% RH are 0.1698% and 0.224%, respectively. The crystal form of all samples remained unchanged after the DVS test, especially the free crystalline form A has almost no hygroscopicity.
13.3、固态稳定性13.3 Solid-state stability
将游离态晶型A和马来酸盐晶型A分别在80℃/闭口放置一天及25℃/60%RH和40℃/75%RH敞口放置1周,通过XRPD和HPLC检测样品的物理和化学稳定性。纯度数据列于表2-6,XRPD结果列于图8-1和图8-2。结果显示所有样品均未观察到明显的纯度变化,且晶型不变。The free form A and the maleate form A were placed at 80°C/closed for one day and at 25°C/60%RH and 40°C/75%RH for one week, respectively, and the physical and chemical stability of the samples was tested by XRPD and HPLC. The purity data are listed in Table 2-6, and the XRPD results are listed in Figures 8-1 and 8-2. The results show that no obvious purity changes were observed for all samples, and the crystal form remained unchanged.
表2-6固态稳定性评估总结
Table 2-6 Summary of solid state stability evaluation
测试例1:化合物对Nav1.8离子通道抑制活性检测Test Example 1: Detection of the inhibitory activity of compounds on Nav1.8 ion channels
试剂除用于酸碱滴定的NaOH和KOH外,均从Sigma(St.Louis,MO)公司购买。测试化合物的最终浓度均在当天配制,再溶于细胞外液。细胞外液(mM)为:NaCl,137mM;KCl,4mM;CaCl2,1.8mM;MgCl2,1mM;HEPES,10mM;glucose,10mM;pH 7.4(NaOH滴定)。所有测试化合物和对照化合物溶液均含1μM TTX。细胞内液(mM)为:天冬氨酸,140mM;氯化镁,2;乙二醇四乙酸(EGTA),11mM;N-2-羟乙基哌嗪-N’-2-乙磺酸(HEPES),10mM。用氢氧化铯调整pH到7.4。All reagents except NaOH and KOH for acid-base titration were purchased from Sigma (St. Louis, MO). The final concentrations of the test compounds were prepared on the day and dissolved in the extracellular solution. The extracellular solution (mM) was: NaCl, 137 mM; KCl, 4 mM; CaCl 2 , 1.8 mM; MgCl 2 , 1 mM; HEPES, 10 mM; glucose, 10 mM; pH 7.4 (NaOH titration). All test compound and control compound solutions contained 1 μM TTX. The intracellular solution (mM) was: aspartic acid, 140 mM; magnesium chloride, 2; ethylene glycol tetraacetic acid (EGTA), 11 mM; N-2-hydroxyethylpiperazine-N'-2-ethanesulfonic acid (HEPES), 10 mM. The pH was adjusted to 7.4 with cesium hydroxide.
测试化合物溶于二甲基亚砜(DMSO),浓度为9mM。测试当天再溶于细胞外液,配制成要求浓度。电生理实验步骤:The test compound was dissolved in dimethyl sulfoxide (DMSO) at a concentration of 9 mM. On the day of the test, it was dissolved in the extracellular fluid to the required concentration. Electrophysiological experimental steps:
将细胞转移到灌流槽中,用细胞外液进行灌流。细胞内液实验当天融化。电极用PC–10(Narishige,Japan)拉制。全细胞膜片钳记录,噪音用采样频率的五分之一进行过滤。电极内加入1/4电极管长的细胞内液,将电极安装在探针上。设置好所需要的Protocol,将界面调成Membrane test,Stage调成Bath。电极内施加正压,将电极尖端接触到细胞,抽气装置三通阀调成三通状态,然后对电极施加负压,使得电极与细胞形成高阻封接。Stage调成Patch,leak控制在-200pA内,继续施加负压,使得细胞膜破裂,形成电流通路。打开抽滤装置和细胞外液阀门进行灌流,观察细胞电流,待细胞电流稳定开始加药(至少3个sweep的电流曲线重叠)。从低浓度往高浓度加药,每个浓度加药时间不少于2min且等到电流稳定再更换浓度加药。Transfer the cells to the perfusion tank and perfuse with extracellular solution. The intracellular solution was thawed on the day of the experiment. The electrodes were pulled with PC-10 (Narishige, Japan). Whole-cell patch clamp recording was performed, and the noise was filtered at one-fifth of the sampling frequency. 1/4 of the length of the electrode tube was added to the electrode, and the electrode was installed on the probe. Set the required Protocol, adjust the interface to Membrane test, and adjust the Stage to Bath. Apply positive pressure to the electrode, touch the tip of the electrode to the cell, adjust the three-way valve of the vacuum device to the three-way state, and then apply negative pressure to the electrode to form a high-resistance seal between the electrode and the cell. Adjust the Stage to Patch, control the leak within -200pA, and continue to apply negative pressure to rupture the cell membrane and form a current path. Open the vacuum filter and the extracellular solution valve for perfusion, observe the cell current, and start adding drugs when the cell current is stable (the current curves of at least 3 sweeps overlap). Add medicine from low concentration to high concentration. The adding time for each concentration should be no less than 2 minutes and wait until the current is stable before changing the concentration.
供试品给药采用利用自身重力的灌流系统进行灌流。在初始记录期间,观察峰值电流幅度至少1分钟直到其稳定。在此期间,所有峰值电流幅度的CV%应小于10%以排除初始电流的上下波动。初始记录期间最后10次记录的峰值电流幅度的平均值作为阴性对照的电流峰值。待初始电流稳定后,试验样品从低浓度开始给药直到10次记录的峰值电流再次稳定或者持续给药5min后,给药后和给药前峰值电流“不变”。我们将下面两种情况定义为“稳定”或者“不变”:1)如果连续10次扫描的峰值电流的绝对平均值超过200pA而CV值小于10%,2)或者连续10次扫描的峰值电流的平均值在200pA和50pA之间而且CV值小于30%。然后给予下一个更高浓度的检测。The test article is administered by perfusion using a perfusion system that uses its own gravity. During the initial recording period, the peak current amplitude is observed for at least 1 minute until it stabilizes. During this period, the CV% of all peak current amplitudes should be less than 10% to exclude the ups and downs of the initial current. The average of the peak current amplitudes recorded in the last 10 times during the initial recording period is used as the current peak of the negative control. After the initial current stabilizes, the test sample is administered from a low concentration until the peak current recorded in 10 times stabilizes again or after continuous administration for 5 minutes, the peak current after administration is "unchanged" from that before administration. We define the following two situations as "stable" or "unchanged": 1) if the absolute average of the peak current for 10 consecutive scans exceeds 200pA and the CV value is less than 10%, 2) or the average of the peak current for 10 consecutive scans is between 200pA and 50pA and the CV value is less than 30%. Then the next higher concentration is administered for testing.
每个浓度最后10次扫描的峰值电流平均值作为该浓度的峰值电流,用于数据分析。如果5分钟内不能达到稳定状态,那么此时的最后10次扫描的峰值电流平均值作为该浓度的峰值电流用于数据分析。同时该细胞要丢弃,不再用于更高浓度的检测。化合物每个浓度至少测试两个细胞。The peak current average of the last 10 scans for each concentration is used as the peak current for data analysis. If a steady state cannot be reached within 5 minutes, the peak current average of the last 10 scans at this time is used as the peak current for data analysis. At the same time, the cell should be discarded and no longer used for detection of higher concentrations. At least two cells are tested for each concentration of the compound.
电压脉冲程序:Voltage pulse procedure:
将细胞钳制在–80mV,然后用持续10毫秒方波去极化到10mV,以得到NaV1.8电流。这一程序每5秒重复一次。检测方波引发的最大电流,待其稳定后,灌流测试化合物,当反应稳定后,计算阻断的强度。The cells were clamped at -80 mV and then depolarized to 10 mV with a 10 ms square wave to obtain NaV1.8 current. This procedure was repeated every 5 seconds. The maximum current induced by the square wave was measured, and after it stabilized, the test compound was perfused. When the response stabilized, the intensity of the blockade was calculated.
数据处理和拟合Data processing and fitting
资料采集和分析将用pCLAMP 10(Molecular Devices,Union City,CA)。电流稳定指的是电流随时间变化在有限的范围内。通过绘制药物的梯度稀释系列浓度和其作用在HEK293/Nav1.8上产生的稳定电流值之间的量效关系,进而计算该药物对Nav1.8离子通道的抑制活性(IC50)。Data collection and analysis will be performed using pCLAMP 10 (Molecular Devices, Union City, CA). Current stability refers to the current changing within a limited range over time. The inhibitory activity (IC 50 ) of the drug on the Nav1.8 ion channel is calculated by plotting the dose-effect relationship between the drug's gradient dilution series concentration and the stable current value generated by its action on HEK293/ Nav1.8 .
表B-1:化合物对于Nav1.8离子通道的抑制活性
Table B-1: Inhibitory activity of compounds on Nav1.8 ion channels
试验结果表明,本发明化合物具有较强的对Nav1.8离子通道抑制活性。The test results show that the compound of the present invention has strong inhibitory activity on Nav1.8 ion channel.
测试例2:小鼠药代动力学试验Test Example 2: Pharmacokinetics test in mice
小鼠药代动力学试验,采用雄性ICR小鼠3只,禁食过夜,口服灌胃给药10mg/kg。在给药前和在给药后15、30分钟以及1、2、4、6、8、24小时采血。血液样品8000转/分钟4℃离心6分钟,收集血浆,于-20℃保存。取各时间点血浆,加入3-5倍量含内标的乙腈溶液混合,涡旋混合1分钟,13000转/分钟4℃离心10分钟,取上清液加入3倍量水混合,取适量混合液进行LC-MS/MS分析。主要药代动力学参数用WinNonlin 7.0软件非房室模型分析。For the pharmacokinetic test in mice, three male ICR mice were fasted overnight and given 10 mg/kg by oral gavage. Blood was collected before administration and at 15, 30 minutes, and 1, 2, 4, 6, 8, and 24 hours after administration. The blood samples were centrifuged at 8000 rpm and 4°C for 6 minutes, and the plasma was collected and stored at -20°C. The plasma at each time point was taken, mixed with 3-5 times the amount of acetonitrile solution containing the internal standard, vortexed for 1 minute, centrifuged at 13000 rpm and 4°C for 10 minutes, the supernatant was taken, mixed with 3 times the amount of water, and an appropriate amount of the mixed solution was taken for LC-MS/MS analysis. The main pharmacokinetic parameters were analyzed by non-compartmental model using WinNonlin 7.0 software.
表B-2:化合物的小鼠药代动力学试验结果
Table B-2: Results of pharmacokinetic tests of compounds in mice
试验结果表明,本发明化合物具有良好的药代动力学特征。The test results show that the compound of the present invention has good pharmacokinetic characteristics.
测试例3:大鼠脊神经结扎神经性疼痛模型Test Example 3: Rat spinal nerve ligation neuropathic pain model
将体重为180~220g的雄性SD大鼠,麻醉后俯卧位放置在手术台上,在动物髋骨附近沿脊柱开口,分离筋膜和肌肉。用镊子小心咬断L5横突,用玻璃分针分离L5神经,使用5-0结扎线结扎L5神经。缝合肌肉和皮肤,碘伏消毒。造模14天后,将动物分为不同组别,每组10只,分别口服灌胃不同化合物,给药后不同时间点用Von-Frey纤维丝检测动物的机械痛阈。具体给药剂量和检测时间,详见下表B-3。Male SD rats weighing 180-220g were placed in a prone position on the operating table after anesthesia. An opening was made along the spine near the hip bone of the animal to separate the fascia and muscles. The L5 transverse process was carefully bitten off with forceps, the L5 nerve was separated with a glass needle, and the L5 nerve was ligated with a 5-0 ligature. The muscles and skin were sutured and disinfected with iodine. After 14 days of modeling, the animals were divided into different groups, with 10 in each group, and different compounds were orally gavaged. The mechanical pain threshold of the animals was detected with Von-Frey fibers at different time points after administration. For specific dosage and detection time, please see Table B-3 below.
机械痛阈检测方法:将试验动物用Von-Frey纤维丝持续刺激待测后肢足底使纤维丝弯曲,观察动物缩足反应。按照纤维丝克数从小到大的顺序逐一刺激受试动物,每一个克数的纤维丝连续刺激5次。若出现的阳性反应小于3次,则使用较大一级的纤维丝重复上述操作,当测试第一次出现3次或3次以上的阳性反应,则该纤维丝为该动物的疼痛阈值(每只动物测3次测试,取其平均值)。纤维丝克数:0.6,1.0,1.4,2.0,4.0,6.0,8.0,10.0,15.0;切断值为15.0g。Mechanical pain threshold detection method: The test animal is continuously stimulated with Von-Frey fibers to make the fibers bend, and the animal's paw retraction reaction is observed. The test animals are stimulated one by one in the order of the fiber weight from small to large, and each fiber weight is stimulated 5 times in a row. If the positive reaction is less than 3 times, repeat the above operation with a larger fiber. When the positive reaction occurs 3 times or more for the first time, the fiber is the pain threshold of the animal (each animal is tested 3 times and the average value is taken). Fiber weight: 0.6, 1.0, 1.4, 2.0, 4.0, 6.0, 8.0, 10.0, 15.0; the cut-off value is 15.0g.
表B-3:化合物对脊神经结扎大鼠疼痛阈值的药效
One-way ANOVA,***P<0.001,**P<0.01,*P<0.05
NA:该时间点未安排检测Table B-3: Effects of compounds on pain threshold in rats with spinal nerve ligation
One-way ANOVA, ***P<0.001, **P<0.01, *P<0.05
NA: No test is scheduled at this time
试验结果表明,本发明化合物可显著改善大鼠脊神经结扎造模引起的动物机械痛阈降低,具有优良的镇痛药效。The test results show that the compound of the present invention can significantly improve the reduction of the mechanical pain threshold of animals caused by spinal nerve ligation modeling in rats and has excellent analgesic effect.
测试例4:大鼠切口痛模型Test Example 4: Rat Incisional Pain Model
将体重为200~250g的雄性SD大鼠,麻醉后俯卧位固定,将其侧后肢脚掌朝上展平,手术胶带固定脚趾,消毒。在动物足底脚后跟0.5cm处用手术刀向趾端划开皮肤筋膜,作一1cm左右的纵向切口。用手术弯镊抬起趾短屈肌后,用手术刀对肌肉腹部进行纵向切口,不将肌肉完全切断。对皮肤进行缝合,消毒。造模第二天,将动物分为不同组别,每组8只,分别口服灌胃不同化合物,给药后不同时间点用Von-Frey纤维丝检测动物的机械痛阈。具体分组给药剂量和检测时间,详见下表B-4。Male SD rats weighing 200-250g were anesthetized and fixed in a prone position. The soles of their hind limbs were flattened upwards, and the toes were fixed with surgical tape and disinfected. A scalpel was used to cut the skin fascia from 0.5cm behind the heel of the animal's foot to the toe tip, and a longitudinal incision of about 1cm was made. After lifting the short flexor digitorum with surgical curved forceps, a longitudinal incision was made on the belly of the muscle with a scalpel without completely cutting the muscle. The skin was sutured and disinfected. On the second day of modeling, the animals were divided into different groups, with 8 rats in each group, and different compounds were orally gavaged. The mechanical pain threshold of the animals was detected with Von-Frey fibers at different time points after administration. For specific grouping dosage and detection time, please see Table B-4 below.
机械痛阈检测方法:将试验动物用Von-Frey纤维丝持续刺激待测后肢足底使纤维丝弯曲,观察动物缩足反应。按照纤维丝克数从小到大的顺序逐一刺激受试动物,每一个克数的纤维丝连续刺激5次。若出现的阳性反应小于3次,则使用较大一级的纤维丝重复上述操作,当测试第一次出现3次或3次以上的阳性反应,则该纤维丝为该动物的疼痛阈值(每只动物测3次测试,取其平均值)。纤维丝克数:0.6,1.0,1.4,2.0,4.0,6.0,8.0,10.0,15.0;切断值为15.0g。Mechanical pain threshold detection method: The test animal is continuously stimulated with Von-Frey fibers to make the fibers bend, and the animal's paw retraction reaction is observed. The test animals are stimulated one by one in the order of the fiber weight from small to large, and each fiber weight is stimulated 5 times in a row. If the positive reaction is less than 3 times, repeat the above operation with a larger fiber. When the positive reaction occurs 3 times or more for the first time, the fiber is the pain threshold of the animal (each animal is tested 3 times and the average value is taken). Fiber weight: 0.6, 1.0, 1.4, 2.0, 4.0, 6.0, 8.0, 10.0, 15.0; the cut-off value is 15.0g.
表B-4:化合物对切口痛模型大鼠疼痛阈值的药效
One-way ANOVA,vs Vehicle组,***P<0.001Table B-4: Effects of compounds on pain threshold in rats with incisional pain model
One-way ANOVA, vs Vehicle group, ***P<0.001
试验结果表明,本发明化合物可显著改善大鼠切口痛造模引起的动物机械痛阈降低,具有优良的镇痛药效。The test results show that the compound of the present invention can significantly improve the reduction of the mechanical pain threshold of animals caused by the incision pain modeling in rats and has excellent analgesic effect.
Claims (10)
A crystalline form of a compound of formula I or a pharmaceutically acceptable salt thereof, wherein the structure of the compound of formula I is as follows:
A pharmaceutically acceptable salt of a compound of formula I, wherein the structure of the compound of formula I is as follows:
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202311723384.5 | 2023-12-13 | ||
CN202311723384 | 2023-12-13 | ||
CN202411777862.5 | 2024-12-04 | ||
CN202411777862 | 2024-12-04 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2025124502A1 true WO2025124502A1 (en) | 2025-06-19 |
Family
ID=95959557
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2024/138893 WO2025124502A1 (en) | 2023-12-13 | 2024-12-12 | Nav1.8 inhibitor compound, salt thereof, polymorph thereof, and use thereof |
Country Status (2)
Country | Link |
---|---|
CN (1) | CN120136793A (en) |
WO (1) | WO2025124502A1 (en) |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN111065383A (en) * | 2017-07-11 | 2020-04-24 | 沃泰克斯药物股份有限公司 | Carboxamides as Sodium Channel Modulators |
CN112225695A (en) * | 2020-12-15 | 2021-01-15 | 上海济煜医药科技有限公司 | Oxynitride and preparation method and application thereof |
WO2021047622A1 (en) * | 2019-09-12 | 2021-03-18 | 上海济煜医药科技有限公司 | Pyridine oxynitride, preparation method therefor and use thereof |
WO2022188872A1 (en) * | 2021-03-11 | 2022-09-15 | 上海济煜医药科技有限公司 | Crystal form of pyridine nitrogen oxide compound and use thereof |
WO2023246867A1 (en) * | 2022-06-22 | 2023-12-28 | 武汉人福创新药物研发中心有限公司 | Nav1.8 inhibitor |
-
2024
- 2024-12-12 WO PCT/CN2024/138893 patent/WO2025124502A1/en unknown
- 2024-12-12 CN CN202411831729.3A patent/CN120136793A/en active Pending
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN111065383A (en) * | 2017-07-11 | 2020-04-24 | 沃泰克斯药物股份有限公司 | Carboxamides as Sodium Channel Modulators |
WO2021047622A1 (en) * | 2019-09-12 | 2021-03-18 | 上海济煜医药科技有限公司 | Pyridine oxynitride, preparation method therefor and use thereof |
CN112225695A (en) * | 2020-12-15 | 2021-01-15 | 上海济煜医药科技有限公司 | Oxynitride and preparation method and application thereof |
WO2022188872A1 (en) * | 2021-03-11 | 2022-09-15 | 上海济煜医药科技有限公司 | Crystal form of pyridine nitrogen oxide compound and use thereof |
WO2023246867A1 (en) * | 2022-06-22 | 2023-12-28 | 武汉人福创新药物研发中心有限公司 | Nav1.8 inhibitor |
Also Published As
Publication number | Publication date |
---|---|
CN120136793A (en) | 2025-06-13 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP6254730B2 (en) | Benzamides | |
CN112566909B (en) | 3-aryloxy-3-five-membered heteroaryl-propylamine compound and application thereof | |
CN104662007A (en) | Cystathionine-upsilon-lyase (cse) inhibitors | |
KR100716389B1 (en) | N-Hydroxy??????????5?isopropyl???methyl??,??thiazol?4?yl?phenoxy?pentoxy?benzamidine ? methansulfonic acid salt | |
CN103957710A (en) | Compounds for use in treatment of mucositis | |
JP2025096383A (en) | Amorphous melanocortin receptor agonists and methods for their preparation | |
CA3119313A1 (en) | Pharmaceutical composition comprising histone deacetylase 6 inhibitors | |
WO2020228789A1 (en) | 3-aryloxyl-3-five-membered heteroaryl propylamine compound, and crystal form and use thereof | |
US20250214986A1 (en) | P2x3 inhibitor compound, salt thereof, polymorph thereof and use thereof | |
JP5726301B2 (en) | New antihistamine | |
CN111943943A (en) | 3-aryloxy-3-pentabasic heteroaryl-propylamine compound and crystal form and application thereof | |
CN114072381B (en) | Application of aminothiol compound as cerebral nerve or heart protecting agent | |
WO2025124502A1 (en) | Nav1.8 inhibitor compound, salt thereof, polymorph thereof, and use thereof | |
KR20220052955A (en) | Salts and crystalline forms of activin receptor-like kinase inhibitors | |
KR20180032585A (en) | Piperazine derivative | |
US9963457B2 (en) | Crystal of 5-((2-(6-amino)-9H-purin-9-yl)ethyl)amino)pentan-1-o1 | |
TW200803856A (en) | Method of treating parkinson's disease with diarylmethylpiperazine compounds exhibiting delta receptor agonist activity | |
CN108948018A (en) | Benzodiazepine * * derivative and its salt and related crystalline form, preparation method and purposes | |
JP6691135B2 (en) | Diarylmethylidene piperidine derivatives and their use as delta opioid receptor agonists | |
TW202521117A (en) | Solid forms of a compound for treating or preventing hyperuricemia or gout | |
KR20250026776A (en) | Solid crystalline form of a compound for the treatment or prevention of hyperuricemia or gout | |
CN119859142A (en) | Crystal forms of heterocyclic compounds and pharmaceutical compositions thereof | |
CN120058684A (en) | Crystalline form of substituted tetrahydrofuran compound and preparation method thereof | |
CN116635033A (en) | Crystal form III of melanocortin receptor agonist compound and its preparation method | |
EA041240B1 (en) | A1 ION CHANNEL INHIBITORS WITH TRANSITOR RECEPTOR POTENTIAL |
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: 24902919 Country of ref document: EP Kind code of ref document: A1 |