WO2023125947A1 - Pharmaceutically acceptable salt of tetrahydroisoquinoline compound, and crystal form and use thereof - Google Patents

Pharmaceutically acceptable salt of tetrahydroisoquinoline compound, and crystal form and use thereof Download PDF

Info

Publication number
WO2023125947A1
WO2023125947A1 PCT/CN2022/143995 CN2022143995W WO2023125947A1 WO 2023125947 A1 WO2023125947 A1 WO 2023125947A1 CN 2022143995 W CN2022143995 W CN 2022143995W WO 2023125947 A1 WO2023125947 A1 WO 2023125947A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
acid
crystal form
Prior art date
Application number
PCT/CN2022/143995
Other languages
French (fr)
Chinese (zh)
Inventor
刘成祥
胡越
陈靖之
吴晖
肖承涛
唐任宏
任晋生
Original Assignee
江苏先声药业有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 江苏先声药业有限公司 filed Critical 江苏先声药业有限公司
Publication of WO2023125947A1 publication Critical patent/WO2023125947A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/438The ring being spiro-condensed with carbocyclic or heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00

Definitions

  • Patent application No. 202111664582.X filed with the State Intellectual Property Office of China on December 30, 2021.
  • the present disclosure relates to the field of medicinal chemistry, in particular, to pharmaceutically acceptable salts of tetrahydroisoquinoline compounds, crystal forms of the pharmaceutically acceptable salts, preparation methods thereof, pharmaceutical compositions containing them, and uses thereof.
  • Epigenetic changes are key mediators that drive and maintain the malignant phenotype of tumors. Changes in DNA methylation, histone acetylation and methylation, non-coding RNA, and post-translational modifications are all epigenetic drivers of cancer development independent of changes in DNA sequence. Arginine methylation is an important class of post-translational modifications that affect cell growth and proliferation, apoptosis, angiogenesis and metastasis by regulating transcription and post-transcriptional RNA processing.
  • methylarginine There are three types of methylarginine, namely ⁇ -NG-monomethylarginine (MMA), ⁇ -NG, N'G-asymmetric dimethylarginine (ADMA) and ⁇ -NG, N'G-symmetric dimethylarginine (SDMA).
  • MMA ⁇ -NG-monomethylarginine
  • ADMA N'G-asymmetric dimethylarginine
  • SDMA N'G-symmetric dimethylarginine
  • PRMT protein arginine methyltransferase
  • AdoMet S-adenosylmethionine
  • PRMT5 is primarily a type II enzyme that catalyzes the symmetric dimethylation of arginine. PRMT5 was first discovered in a two-hybrid assay detecting proteins interacting with Janus tyrosine kinase (Jak2).
  • PRMT5 is a general transcriptional repressor that forms a complex with other transcription factors, including BRG1 and hBRM, Blimp1, and Snail. PRMT5 participates in a variety of different cellular biological processes through the methylation of a variety of substrates in the cytoplasm and nucleus, including histone H4 residue Arg3 (H4R3) and H3 residue Arg8 (H3R8). H4R3 methylation is associated with transcriptional repression, while H3R8 methylation is thought to be associated with both transcriptional activation and transcriptional repression.
  • H4R3 methylation is associated with transcriptional repression
  • H3R8 methylation is thought to be associated with both transcriptional activation and transcriptional repression.
  • PRMT5 In addition to the direct induction of repressive histone marks by PRMT5, the enzyme's role in gene silencing is also mediated through the formation of multiple arrestin complexes, including NuRD components, HDACs, MDB proteins, and DNA methyltransferases. PRMT5 affects its substrate specificity by interacting with some binding proteins. A central component in this protein complex is MEP50. MEP50 is required for the enzymatic activity of PRMT5. The study found that PRMT5 can methylate proteins involved in RNA splicing, such as SmD3, which can be used to track the chemical activity of cellular biological PRMT5.
  • PRMT5 plays an important role in tumorigenesis. Studies have found that the expression of PRMT5 is upregulated in a variety of tumors, including lymphoma, lung cancer, breast cancer and colorectal cancer. In addition, PRMT5 expression was increased in mantle cell lymphoma (MCL) patient samples, and PRMT5 knockout could inhibit the proliferation of MCL cells, indicating that PRMT5 plays an important role in MCL. PRMT5 overexpression promotes cell proliferation, and in melanoma, breast cancer, and lung cancer cell lines, PRMT5 knockdown can inhibit the proliferation of these cells. Therefore, PRMT5 is a potential target for cancer therapy.
  • MCL mantle cell lymphoma
  • MTAP methylthioadenosine phosphorylase
  • PCT/CN2021/103597 (application date June 30, 2021) describes a compound 1-ethyl-4-((R)-2-hydroxy-2-((S)-1,2,3,4 -Tetrahydroisoquinolin-3-yl)ethyl)-8-(2-methoxy-7-azaspiro[3.5]nonane-7-carbonyl)-1,2,3,4-tetrahydro -5H-Benzo[e][1,4]diazepine -5-keto (compound I) hydrochloride, the study found that the compound I hydrochloride has good PRMT5 enzyme inhibition, cell proliferation inhibition and cell SDMA inhibitory activity, and good pharmacokinetic properties and liver function. Cell metabolism stability, and showed significant tumor growth inhibition in mouse subcutaneous xenograft Z-138 model, and showed a good dose-response relationship.
  • the present disclosure provides a pharmaceutically acceptable salt of Compound I, wherein the pharmaceutically acceptable salt is selected from monohydrochloride, hydrobromide, 1,5-naphthalene disulfonate, oxalate, citrate, Sulfate, Phosphate, L-Tartrate, L-Malate, Succinate, Adipate, Fumarate, Oxalate, Propionate, Benzoate, Acetate, Formic Acid salt or L-arginine salt, preferably monohydrochloride, hydrobromide or 1,5-naphthalene disulfonate, more preferably monohydrochloride.
  • the pharmaceutically acceptable salt is selected from monohydrochloride, hydrobromide, 1,5-naphthalene disulfonate, oxalate, citrate, Sulfate, Phosphate, L-Tartrate, L-Malate, Succinate, Adipate, Fumarate, Oxalate, Propionate, Benzoate, Acetate, Formic Acid salt
  • the molar ratio of Compound I to acid molecules is about 1:2 to 2:1, preferably selected from 1:2, 1:1 or 2:1, More preferably 1:1.
  • a monohydrochloride salt of Compound I wherein the molar ratio of Compound I to hydrochloric acid is about 1:1.
  • a hydrobromide salt of Compound I is provided.
  • a 1,5-naphthalene disulfonate salt of Compound I is provided.
  • the pharmaceutically acceptable salts of Compound I exist in unsolvated or solvated form.
  • the present disclosure also provides solid forms of the pharmaceutically acceptable salts of Compound I.
  • the solid form of the pharmaceutically acceptable salt of Compound I is selected from amorphous or crystalline forms.
  • the present disclosure also provides a method for preparing a pharmaceutically acceptable salt of the aforementioned compound I, comprising: a step of forming a salt of compound I with an acid, the acid being selected from hydrochloric acid, hydrobromic acid, 1,5-naphthalene disulfonic acid, oxalic acid , citric acid, sulfuric acid, phosphoric acid, L-tartaric acid, L-malic acid, succinic acid, adipic acid, fumaric acid, oxalic acid, propionic acid, benzoic acid, acetic acid, formic acid or L-arginine, preferably hydrochloric acid, hydrogen Bromic acid or 1,5-naphthalenedisulfonic acid, more preferably hydrochloric acid.
  • an acid being selected from hydrochloric acid, hydrobromic acid, 1,5-naphthalene disulfonic acid, oxalic acid , citric acid, sulfuric acid, phosphoric acid, L-tartaric acid, L-malic acid,
  • the solvent used in the salt-forming reaction described in the present disclosure is selected from at least one of ethyl acetate, tetrahydrofuran, acetonitrile, acetone, methyl acetate, isopropanol, methanol or toluene, preferably selected from isopropanol, methyl acetate, acetic acid ethyl ester, methanol or acetone, more preferably isopropanol or ethyl acetate.
  • the method for preparing the aforementioned pharmaceutically acceptable salt further includes at least one of the steps of volatilizing the solvent, crystallizing with stirring, filtering, washing or drying.
  • the pharmaceutically acceptable salt of Compound I is in crystalline form, preferably hydrochloride crystal form, hydrobromide salt crystal form, 1,5-naphthalene disulfonate salt crystal form, more preferably monohydrochloride Salt crystal form.
  • the hydrochloride crystal form of the compound I is the A crystal form of the compound I-hydrochloride
  • the X-ray powder diffraction pattern of the A crystal form represented by the diffraction angle 2 ⁇ is 5.42 ⁇ 0.2 o , 6.60 ⁇ 0.2o , 10.36 ⁇ 0.2o , 13.60 ⁇ 0.2o , 14.24 ⁇ 0.2o , 15.83 ⁇ 0.2o, 16.62 ⁇ 0.2o , 17.65 ⁇ 0.2o, 19.29 ⁇ 0.2o , 19.86 ⁇ 0.2o , 21 .37 ⁇ 0.2 o and 23.88 ⁇ 0.2 o have diffraction peaks.
  • the crystal form A has an X-ray powder diffraction pattern represented by diffraction angle 2 ⁇ at 5.42 ⁇ 0.2 ° , 6.60 ⁇ 0.2 ° , 9.76 ⁇ 0.2 ° , 10.36 ⁇ 0.2 ° , 13.00 ⁇ 0.2 ° , 13.60 ⁇ 0.2o , 14.24 ⁇ 0.2o , 15.83 ⁇ 0.2o , 16.62 ⁇ 0.2o, 17.65 ⁇ 0.2o, 18.28 ⁇ 0.2o, 19.29 ⁇ 0.2o , 19.86 ⁇ 0.2o , 20.61 ⁇ 0.2o, 21. 37 ⁇ 0.2o , 23.88 ⁇ 0.2 o , 25.08 ⁇ 0.2 o , 28.22 ⁇ 0.2 o have diffraction peaks.
  • the crystal form A has an X-ray powder diffraction pattern represented by diffraction angle 2 ⁇ at 5.42 ⁇ 0.2 ° , 6.60 ⁇ 0.2 ° , 6.83 ⁇ 0.2 ° , 9.76 ⁇ 0.2 ° , 10.36 ⁇ 0.2 ° , 13.00 ⁇ 0.2o , 13.60 ⁇ 0.2o , 14.24 ⁇ 0.2o , 15.27 ⁇ 0.2o , 15.83 ⁇ 0.2o, 16.62 ⁇ 0.2o, 17.30 ⁇ 0.2o , 17.65 ⁇ 0.2o, 18.28 ⁇ 0.2o , 19.
  • the crystal form A has an X-ray powder diffraction pattern represented by diffraction angle 2 ⁇ at 5.42 ⁇ 0.2 ° , 6.60 ⁇ 0.2 ° , 6.83 ⁇ 0.2 ° , 9.76 ⁇ 0.2 ° , 10.36 ⁇ 0.2 ° , 13.00 ⁇ 0.2o , 13.60 ⁇ 0.2o , 14.24 ⁇ 0.2o , 15.27 ⁇ 0.2o , 15.83 ⁇ 0.2o, 16.62 ⁇ 0.2o, 17.30 ⁇ 0.2o , 17.65 ⁇ 0.2o, 18.28 ⁇ 0.2o , 19.
  • the X-ray powder diffraction pattern of Form A is substantially as shown in FIG. 8 .
  • the crystalline Form A has a DSC spectrum with a peak at 259.27 ⁇ 5.0°C.
  • the DSC spectrum of Form A is substantially as shown in FIG. 9 .
  • the present disclosure also provides a method for preparing the A crystal form of compound I-hydrochloride, comprising the steps of:
  • step 3) after step 2) of the preparation method of the A crystal form, that is, a good solvent is used to dissolve the crystal obtained in step 2), and after the solution is obtained, a poor solvent is added dropwise to analyze crystal.
  • step 3' there is an optional step 3') after step 2) of the preparation method of crystal form A, that is, dissolving the crystal form obtained in step 2) in methanol and concentrating under reduced pressure to obtain compound I-hydrochloride free Shaped matter, and then dissolve the amorphous matter in solvent (ii), and then precipitate crystals by beating or adding anti-solvent dropwise.
  • the solvent (i) in step 1) of the preparation method for crystal form A is selected from methyl acetate, ethyl acetate, acetone or isopropanol, preferably isopropanol or ethyl acetate.
  • the volume (mL) of the solvent (i) in step 1) of the preparation method for crystal form A is 5 to 30 times, preferably 5 to 20 times, more preferably 10 to 30 times the weight of the compound (g). 15 times.
  • an organic solvent solution of hydrogen chloride preferably a hydrogen chloride-ethyl acetate solution, is added in step 2) of the preparation method for crystal form A.
  • the organic solvent solution of hydrogen chloride in step 2) of the preparation method of crystal form A is added dropwise to the reaction system at -10-10°C, preferably 0-10°C, and kept for reaction.
  • the molar amount of hydrogen chloride used in step 2) of the preparation method for crystal form A is 0.95-1.05 times the molar amount of compound I in step 1.
  • reaction time of step 2) of the preparation method for crystal form A is 0.5-3 hours, preferably 1-2 hours.
  • the good solvent in step 3) of the preparation method of crystal form A is selected from methanol, dichloromethane or chloroform, preferably methanol; the poor solvent is selected from isopropanol, acetone or isopropyl ether, preferably isopropyl alcohol.
  • crystals are prepared by beating in step 3') of the preparation method of crystal form A, and the solvent (ii) is selected from acetone, 4-methyl-2-pentanone, and 2-butanone , isopropyl ether, methyl tert-butyl ether, diethyl ether, n-propanol, isopropanol, ethanol, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, acetonitrile, tetrahydrofuran, cyclohexane, toluene , one or more of water or acetone aqueous solution, preferably ethyl acetate.
  • the solvent (ii) is selected from acetone, 4-methyl-2-pentanone, and 2-butanone , isopropyl ether, methyl tert-butyl ether, diethyl ether, n-propanol, isopropano
  • step 3') of the preparation method of crystal form A crystals are prepared by adding anti-solvent dropwise, and the solvent (ii) is selected from dichloromethane, chloroform or methanol, preferably methanol;
  • the anti-solvent is selected from acetone, isopropyl ether, methyl acetate, acetonitrile or tetrahydrofuran, preferably acetone.
  • the hydrochloride crystal form of the compound I is the B crystal form of the compound I-hydrochloride
  • the X-ray powder diffraction pattern of the B crystal form represented by the diffraction angle 2 ⁇ is at 5.16 ⁇ 0.2 o , 9.86 ⁇ 0.2o , 10.42 ⁇ 0.2o , 10.76 ⁇ 0.2o , 11.36 ⁇ 0.2o , 15.68 ⁇ 0.2o , 16.31 ⁇ 0.2o, 17.29 ⁇ 0.2o , 17.98 ⁇ 0.2o , 18.89 ⁇ 0.2o , 19 .83 ⁇ 0.2
  • the crystal form B has an X-ray powder diffraction pattern represented by diffraction angle 2 ⁇ at 5.16 ⁇ 0.2 ° , 9.86 ⁇ 0.2 ° , 10.42 ⁇ 0.2 ° , 10.76 ⁇ 0.2 ° , 11.36 ⁇ 0.2 ° , 14.46 ⁇ 0.2o , 15.68 ⁇ 0.2o , 16.31 ⁇ 0.2o , 17.29 ⁇ 0.2o , 17.98 ⁇ 0.2o , 18.89 ⁇ 0.2o , 19.83 ⁇ 0.2o , 20.99 ⁇ 0.2o, 22.13 ⁇ 0.2o , 22. 72 ⁇ 0.2o , 23.61 ⁇ 0.2 o and 25.94 ⁇ 0.2 o have diffraction peaks.
  • the crystal form B has an X-ray powder diffraction pattern represented by diffraction angle 2 ⁇ at 5.16 ⁇ 0.2 ° , 6.33 ⁇ 0.2 ° , 8.98 ⁇ 0.2 ° , 9.86 ⁇ 0.2 ° , 10.42 ⁇ 0.2 ° , 10.76 ⁇ 0.2o , 11.36 ⁇ 0.2o , 12.82 ⁇ 0.2o , 14.46 ⁇ 0.2o , 15.68 ⁇ 0.2o , 16.31 ⁇ 0.2o, 17.29 ⁇ 0.2o , 17.31 ⁇ 0.2o , 17.98 ⁇ 0.2o , 18.
  • the X-ray powder diffraction pattern of Form B is substantially as shown in FIG. 11 .
  • the DSC spectrum of Form B is substantially as shown in FIG. 12 .
  • the hydrobromide salt crystal form of Compound I is the C crystal form
  • the X-ray powder diffraction pattern of the C crystal form represented by the diffraction angle 2 ⁇ is at 6.45 ⁇ 0.2 ° , 9.49 ⁇ 0.2 ° , 10.01 ⁇ 0.2 o , 12.96 ⁇ 0.2 o , 13.45 ⁇ 0.2 o , 13.81 ⁇ 0.2 o , 15.50 ⁇ 0.2 o , 19.13 ⁇ 0.2 o , 20.13 ⁇ 0.2 o , 20.78 ⁇ 0.2 o , 23.31 ⁇ 0.2 o .
  • the crystal form C has an X-ray powder diffraction pattern represented by diffraction angle 2 ⁇ at 6.45 ⁇ 0.2 ° , 9.49 ⁇ 0.2 ° , 10.01 ⁇ 0.2 ° , 12.96 ⁇ 0.2 ° , 13.45 ⁇ 0.2 ° , 13.81 ⁇ 0.2o , 15.50 ⁇ 0.2o , 17.30 ⁇ 0.2o , 19.13 ⁇ 0.2o , 19.46 ⁇ 0.2o , 20.13 ⁇ 0.2o, 20.78 ⁇ 0.2o , 23.31 ⁇ 0.2o , 24.16 ⁇ 0.2o , 24. 55 ⁇ 0.2o , 24.80 ⁇ 0.2 o , 25.62 ⁇ 0.2 o , 27.00 ⁇ 0.2 o , 28.87 ⁇ 0.2 o , 28.95 ⁇ 0.2 o have diffraction peaks.
  • the crystal form C has an X-ray powder diffraction pattern represented by diffraction angle 2 ⁇ at 6.45 ⁇ 0.2 ° , 9.49 ⁇ 0.2 ° , 10.01 ⁇ 0.2 ° , 12.96 ⁇ 0.2 ° , 13.45 ⁇ 0.2 ° , 13.81 ⁇ 0.2o , 15.50 ⁇ 0.2o , 16.29 ⁇ 0.2o , 16.95 ⁇ 0.2o , 17.30 ⁇ 0.2o , 19.13 ⁇ 0.2o, 19.46 ⁇ 0.2o , 20.13 ⁇ 0.2o, 20.78 ⁇ 0.2o , 23.
  • the X-ray powder diffraction pattern of Form C is substantially as shown in FIG. 2 .
  • the crystalline Form C has a DSC spectrum with a peak at 238.84 ⁇ 5.0°C.
  • the DSC spectrum of Form C is substantially as shown in FIG. 3 .
  • the 1,5-naphthalene disulfonate salt of Compound I is the D crystal form
  • the X-ray powder diffraction pattern of the D crystal form represented by the diffraction angle 2 ⁇ is at 4.02 ⁇ 0.2 o , 9.89 ⁇ 0.2o , 11.49 ⁇ 0.2o , 12.74 ⁇ 0.2o , 15.68 ⁇ 0.2o, 15.69 ⁇ 0.2o , 16.15 ⁇ 0.2o, 18.62 ⁇ 0.2o , 18.93 ⁇ 0.2o, 19.67 ⁇ 0.2o , 19.79 ⁇ 0.2o o , 21.26
  • the crystal form D has an X-ray powder diffraction pattern represented by diffraction angle 2 ⁇ at 4.02 ⁇ 0.2 ° , 8.06 ⁇ 0.2 ° , 8.79 ⁇ 0.2 ° , 9.89 ⁇ 0.2 ° , 11.49 ⁇ 0.2 ° , 12.74 ⁇ 0.2o , 13.94 ⁇ 0.2o , 15.68 ⁇ 0.2o , 15.69 ⁇ 0.2o , 16.15 ⁇ 0.2o, 17.21 ⁇ 0.2o, 18.62 ⁇ 0.2o , 18.93 ⁇ 0.2o , 19.67 ⁇ 0.2o , 19. 79 ⁇ 0.2o , 20.57 ⁇ 0.2 o , 21.26 ⁇ 0.2 o , 24.36 ⁇ 0.2 o , 25.64 ⁇ 0.2 o , 27.56 ⁇ 0.2 o have diffraction peaks.
  • the crystal form D has an X-ray powder diffraction pattern represented by diffraction angle 2 ⁇ at 4.02 ⁇ 0.2 ° , 8.06 ⁇ 0.2 ° , 8.79 ⁇ 0.2 ° , 9.89 ⁇ 0.2 ° , 11.49 ⁇ 0.2 ° , 12.10 ⁇ 0.2o , 12.74 ⁇ 0.2o , 13.94 ⁇ 0.2o , 14.94 ⁇ 0.2o , 15.68 ⁇ 0.2o , 15.69 ⁇ 0.2o, 16.15 ⁇ 0.2o , 17.21 ⁇ 0.2o, 17.63 ⁇ 0.2o , 17.
  • the X-ray powder diffraction pattern of Form D is substantially as shown in FIG. 5 .
  • the crystalline Form D has a DSC spectrum with a peak at 231.85 ⁇ 5.0°C.
  • the DSC spectrum of Form D is substantially as shown in FIG. 6 .
  • the crystallization method of the crystal form in the present disclosure is conventional, such as crystallization from a volatile solvent, crystallization at lower temperature or crystallization at room temperature.
  • preparation method of the crystal form in the present disclosure also includes steps such as filtering, washing or drying.
  • the present disclosure provides a pharmaceutical composition, which comprises a pharmaceutically acceptable salt of Compound I described in the present disclosure, and pharmaceutically acceptable excipients.
  • the present disclosure provides a method for treating a disease or condition mediated by PRMT5 in a mammal, comprising administering a therapeutically effective amount of a pharmaceutically acceptable salt of Compound I of the present disclosure to a mammal in need of such treatment, preferably a human. , or a pharmaceutical composition thereof.
  • the present disclosure provides the use of a pharmaceutically acceptable salt of Compound I described in the present disclosure, or a pharmaceutical composition thereof, in the preparation of a medicament for preventing or treating a disease or disorder mediated by PRMT5.
  • the present disclosure provides the use of a pharmaceutically acceptable salt of Compound I described in the present disclosure, or a pharmaceutical composition thereof, in preventing or treating a disease or condition mediated by PRMT5.
  • the present disclosure provides a pharmaceutically acceptable salt of compound I of the present disclosure, or a pharmaceutical composition thereof, for preventing or treating a disease or condition mediated by PRMT5.
  • the present disclosure provides a pharmaceutically acceptable salt of compound I of the present disclosure, or a pharmaceutical composition thereof, for preventing or treating cancer.
  • the disease or condition mediated by PRMT5 includes, but is not limited to, cancer.
  • the room temperature mentioned in the present disclosure refers to 20 ⁇ 5°C.
  • the range "m ⁇ n” described in the present disclosure represents an abbreviated representation of any combination of real numbers between m and n, where both m and n are real numbers.
  • the numerical range “5-30” means that 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30;
  • “10 ⁇ 15” means that 10, 11, 12, 13, 14 or 15 have been listed in this article, "10 ⁇ 15 " is just an abbreviation for these numerical combinations.
  • the peak positions of their XRPD patterns are generally similar, and the relative intensity error may be large. It should also be pointed out that in the identification of mixtures, due to the decrease of content and other factors, some diffraction lines will be missing. At this time, there is no need to rely on all the diffraction peaks observed in high-purity samples, and even one diffraction peak may also affect the given is characteristic for a given crystal.
  • 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 diffraction peak 2 ⁇ is ⁇ 0.20°.
  • terapéuticaally effective amount means (i) treating or preventing a particular disease, condition or disorder, (ii) alleviating, ameliorating or eliminating one or more symptoms of a particular disease, condition or disorder, or (iii) preventing or An amount of a compound of the disclosure that delays the onset of one or more symptoms of a particular disease, condition or disorder described herein.
  • the amount of a compound of the present disclosure that constitutes a “therapeutically effective amount” will vary depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by one skilled in the art according to its own knowledge and this disclosure.
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms which, within the scope of sound medical judgment, are suitable for use in contact with human and animal tissues without excessive Toxicity, irritation, allergic reaction, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable excipients refers to those excipients that have no obvious stimulating effect on the organism and will not impair the biological activity and performance of the active compound. Suitable excipients are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
  • pharmaceutical composition refers to a mixture of one or more compounds of the present disclosure or pharmaceutically acceptable salts or solvates thereof and pharmaceutically acceptable excipients.
  • the purpose of the pharmaceutical composition is to facilitate administration of a compound of the present disclosure, or a pharmaceutically acceptable salt or solvate thereof, to an organism.
  • the pharmaceutical composition of the present disclosure can be prepared by combining the compound of the present disclosure or its pharmaceutically acceptable salt or its solvate with suitable pharmaceutically acceptable auxiliary materials, for example, it can be formulated into solid, semi-solid, liquid or gaseous preparations , such as tablets, pills, capsules, powders, granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres and aerosols, etc.
  • Typical routes of administration of a compound of the present disclosure, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising the compound or a pharmaceutically acceptable salt or solvate thereof include, but are not limited to, oral, rectal, topical, Inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous administration.
  • the pharmaceutical composition of the present disclosure can be produced by methods well known in the art, such as conventional mixing methods, dissolving methods, granulating methods, emulsifying methods, freeze-drying methods and the like.
  • the pharmaceutical composition is in oral form.
  • the pharmaceutical compositions can be formulated by mixing the active compounds with pharmaceutically acceptable excipients well known in the art. These excipients enable the disclosed compound or its pharmaceutically acceptable salt or its solvate to be formulated into tablets, pills, lozenges, dragees, capsules, liquids, gels, slurries, suspensions, etc. for oral administration to patients.
  • Solid oral compositions can be prepared by conventional methods of mixing, filling or tabletting. It can be obtained, for example, by mixing the active compound with solid excipients, optionally milling the resulting mixture, adding other suitable excipients if desired, and processing the mixture into granules to obtain tablets Or the core of the sugar coating.
  • Suitable auxiliary materials include but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, etc.
  • the pharmaceutical composition may also be adapted for parenteral administration as a suitable unit dosage form of sterile solutions, suspensions or lyophilized products.
  • the therapeutically effective amount of the pharmaceutically acceptable salt of Compound I contained in the pharmaceutical composition of the present disclosure is selected from 0.01 mg/kg to 100 mg/kg, in single or divided doses.
  • XRPD peak positions and/or intensities for a given crystalline form of the same compound will vary within error limits.
  • the 2 ⁇ values allow for an appropriate margin of error.
  • the margin of error is represented by " ⁇ ".
  • 5.42 ⁇ 0.2° 2 ⁇ means within the range of about 5.22 to 5.62° 2 ⁇ .
  • a suitable error range for the XRPD diffraction angle (2 ⁇ ) may be about ⁇ 0.20°, ⁇ 0.15°, ⁇ 0.10° , ⁇ 0.05° or less.
  • the term “substantially the same” or “substantially as shown” means comprising at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 90% Or a pattern of diffraction peaks with at least 99% of the diffraction angles within a standard deviation of ⁇ 0.2° 2 ⁇ .
  • measurements of a DSC spectrum for a given crystalline form of the same compound will vary within a margin of error.
  • Single-peak peak values (expressed in degrees Celsius) allow for a modest margin of error.
  • the margin of error is represented by " ⁇ ".
  • thermal transition temperatures and melting points are typically within about ⁇ 5.0°C in consecutive analyses.
  • a peak value of "259.27 ⁇ 5.0” indicates that it is in the range of 254.27 to 264.27.
  • appropriate error ranges for unimodal peaks may be ⁇ 5.0, ⁇ 4.0, ⁇ 3.0, ⁇ 2.0 or less.
  • Salt forms and/or crystalline forms of the present disclosure may also be isotopically labeled.
  • the disclosure also includes isotopically labeled disclosed compounds that are identical to those described herein, but with one or more atoms replaced by an atom of an atomic mass or mass number different from that normally found in nature.
  • isotopes that can be incorporated into compounds of the present disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 123 I, 125 I and 36 Cl, etc.
  • isotopically-labeled compounds of the disclosure are useful in compound and/or substrate tissue distribution assays. Tritiated (ie3H ) and carbon-14 ( ie14C ) isotopes are especially preferred for their ease of preparation and detectability.
  • Positron-emitting isotopes such as 15 O, 13 N, 11 C, and 18 F, can be used in positron emission tomography (PET) studies to determine substrate occupancy.
  • Isotopically labeled compounds of the disclosure can generally be prepared by following procedures similar to those disclosed in the Schemes and/or Examples below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • substitution with heavier isotopes such as deuterium may confer certain therapeutic advantages resulting from greater metabolic stability (e.g. increased in vivo half-life or reduced dosage requirements), and thus in some cases
  • deuterium substitution may be partial or complete, partial deuterium substitution means that at least one hydrogen is replaced by deuterium.
  • the compounds of the present disclosure or their pharmaceutically acceptable salts or their solvates can be prepared by various synthetic methods well known to those skilled in the art, including the specific embodiments listed below, and the compounds formed by combining them with other chemical synthesis methods Embodiments and equivalent replacements known to those skilled in the art, preferred embodiments include but are not limited to the examples of the present disclosure.
  • Sample pan aluminum pan, non-sealed gland
  • Heating rate starting from the initial temperature of 35°C, raise the temperature to 100°C at a rate of 20°C per minute, maintain for 5 minutes, then increase the temperature to 200°C at a rate of 20°C per minute, maintain for 1 minute, and then increase the temperature at a rate of 10°C per minute The rate of heating up to 300°C
  • the detection adopts DVS Intrinsic.
  • the pharmaceutically acceptable salts of Compound I described in the present disclosure have advantages in terms of physicochemical properties, preparation processability and bioavailability, and are suitable for preparing desired pharmaceutical compositions.
  • the compounds of the present disclosure can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by combining them with other chemical synthesis methods, and the methods well known to those skilled in the art As an equivalent alternative, preferred embodiments include but are not limited to the examples of the present disclosure.
  • Figure 1 is a ball-and-stick diagram of compound 1-8 single crystal.
  • Fig. 2 is the XRPD spectrum of compound I hydrobromide salt C crystal form.
  • Figure 3 is the DSC spectrum of compound I hydrobromide salt C crystal form.
  • Fig. 4 is the TGA spectrum of compound I hydrobromide salt C crystal form.
  • Figure 5 is the XRPD pattern of Compound I 1,5-naphthalene disulfonate D crystal form.
  • Figure 6 is the DSC spectrum of compound I 1,5-naphthalene disulfonate D crystal form.
  • Figure 7 is the TGA spectrum of compound I 1,5-naphthalene disulfonate D crystal form.
  • Fig. 8 is the XRPD pattern of compound I-hydrochloride A crystal form.
  • Fig. 9 is a DSC spectrum of compound I-hydrochloride A crystal form.
  • Figure 10 is the TGA pattern of compound I-hydrochloride A crystal form.
  • Figure 11 is the XRPD pattern of compound I-hydrochloride B crystal form.
  • Figure 12 is the DSC spectrum of compound I-hydrochloride B crystal form.
  • Figure 13 is the TGA pattern of compound I-hydrochloride B crystal form.
  • Figure 14 is the tumor growth curve of mice receiving the test compound in the Z-138 subcutaneous tumor model, in which compound 002 is the compound of Example 2.
  • Figure 15 is the curve of body weight change of mice receiving the test compound in the Z-138 subcutaneous tumor model, in which compound 002 is the compound of Example 2.
  • ratios indicated for mixed solvents are volume mixing ratios. Unless otherwise stated, % means wt%.
  • NMR nuclear magnetic resonance
  • MS mass spectroscopy
  • the following eluents can be mixed eluents formed by two or more solvents, and the ratio is the volume ratio of each solvent.
  • “0-10% methanol/dichloromethane” means that during the gradient elution process, the volume ratio of methanol and dichloromethane in the mixed eluent is 0:100-10:100.
  • reaction solution was reacted at room temperature for 4 hours. After the reaction was complete, the excess N,N-diisopropylethylamine and N,N-dimethylformamide were removed by rotary evaporation, then cooled in an ice bath, diluted with saturated brine (1L), extracted with ethyl acetate (200mL X 2), the combined organic phases were washed with 5% aqueous sodium carbonate solution (500mL X 2), and then washed with saturated brine (500mL).
  • the reaction system was cooled to room temperature, and Boc anhydride (122 mg, 0.56 mmol) was added to react for 1.5 hours.
  • the reaction system was cooled to 0°C, and the pH of the reaction solution was adjusted to 5.0 with 1mol/L hydrochloric acid aqueous solution, and then extracted with ethyl acetate (30mL X 3), the organic phase was dried with anhydrous sodium sulfate, filtered, The filtrate was concentrated under reduced pressure.
  • Single crystal preparation method Weigh compound 1-8 (10.0 mg) into a 3 mL screw-top glass bottle, add 2 mL of methanol, stir for 5 minutes, and then the solid dissolves. Add 0.5 mL of water to the glass bottle and continue stirring for 5 minutes. The solution was filtered through a 0.22 ⁇ m microporous membrane into a 3 mL screw-top glass bottle, and the mouth of the glass bottle was covered with plastic wrap. Prick 8 small holes at the mouth of the bottle with a needle and leave it at room temperature for 7 days to obtain a single crystal of the above compound.
  • the obtained single crystal sample was subjected to X-ray analysis, and the test results are shown in Table 1 and Fig. 1 .
  • Chromatographic information Chromatographic column—Shim-pack IC-A3; guard column—Shim-pack IC-GA3; eluent—8 mmol/L p-hydroxybenzoic acid, 3.2mmol/L Bis-Tris, 50mmol/L Boric acid solution; flow rate - 1.5mL/min; column temperature - 40°C; injection volume - 50 ⁇ L; conductivity detector mode - positive ion mode;
  • Reference solution (containing 6 ⁇ g/mL of chloride ion): Accurately measure 50 mg of sodium chloride into a 50 mL measuring bottle, add water to dissolve and dilute to the mark, shake well, precisely pipette 1.0 mL into a 100 mL measuring bottle, add water to dilute to the mark , shake well.
  • Test solution (0.1mg/mL): Accurately weigh 10mg of the sample to be tested into a 100mL measuring bottle, dissolve and dilute to the mark with water, and shake well.
  • Determination method Accurately measure the reference substance solution and the test solution respectively, inject them into a high-performance liquid chromatograph, use the eluent as the eluent to elute, and record the chromatogram.
  • Embodiment 4 Preparation of compound I hydrobromide C crystal form
  • the XRPD spectrum of the crystalline sample is shown in Figure 2, and the XRPD diffraction peak positions are shown in Table 2 below; the DSC spectrum is shown in Figure 3, and the peak is around 238.84°C; the TGA spectrum is shown in Figure 4.
  • Embodiment 6 Preparation of compound I-hydrochloride A crystal form
  • the content of hydrochloric acid in crystal form A was determined by potentiometric titration, and it was determined that the molar ratio of compound I to HCl in the salt was about 1:1.
  • the experimental instruments, reagents and methods for determining the content of hydrochloric acid by potentiometric titration are as follows.
  • Blank titration Measure 50 mL of solvent (60% methanol) into a 100 mL beaker, and titrate to the end point with sodium hydroxide titration solution (0.1 mol/L).
  • Titration of the sample to be tested Accurately weigh 300 mg of the sample to be tested, place it in a 100 mL beaker, add 50 mL of solvent (60% methanol), and titrate to the end point with sodium hydroxide titration solution (0.1 mol/L).
  • the hydrochloric acid content is calculated by the following formula:
  • V 0 blank consumes the volume of sodium hydroxide titration solution, unit (mL);
  • V the volume of sodium hydroxide titration solution consumed by the sample to be tested, unit (mL);
  • m the weighing amount of the sample to be tested, unit (mg);
  • the XRPD of the crystalline sample is shown in Figure 8, and the XRPD diffraction peak positions are shown in Table 4 below; the DSC spectrum is shown in Figure 9, and the peak is around 259.27°C; the TGA spectrum is shown in Figure 10.
  • Embodiment 7 Preparation of compound I-hydrochloride A crystal form
  • Embodiment 8 Preparation of compound I-hydrochloride A crystal form
  • Embodiment 9 Preparation of compound I-hydrochloride A crystal form
  • Embodiment 10 Preparation of compound I-hydrochloride amorphous
  • Example 10 At room temperature, add the compound I-hydrochloride amorphous (20 mg) prepared in Example 10 into a 4.5 mL centrifuge tube, add methanol (200 ⁇ L), dissolve and add acetone dropwise until solid precipitates, stir for several hours The precipitated solid was suction filtered, the filter cake was washed with 1 mL of acetone, and dried under vacuum at 25°C to obtain a solid (12 mg).
  • the XRPD of the crystalline sample is shown in Figure 11, and its XRPD diffraction peak positions are shown in Table 5 below; its DSC spectrum is shown in Figure 12; its TGA spectrum is shown in Figure 13.
  • PRMT5/MEP50 protein was purchased from BPS bioscience (USA); histone H4 peptide (Histone H4 Peptide) substrate was purchased from Sangon Bioengineering (Shanghai) Co., Ltd.; Anti-Histone H4 (symmetric dimethyl R3) antibody-ChIP Grade was purchased from Abcam (US); S-(5'-adenosyl)-L-methionine chloride dihydrochloride was purchased from Sigma (US); 384-well plate, AlphaScreen streptavidin AlphaScreen Streptavidin Donor beads, AlphaScreen Protein A Acceptor beads and Envision 2104 multi-label Reader were purchased from PerkinElmer Instruments Co., Ltd. (USA) Echo 550 pipette (Echo 550 Liquid Handler) was purchased from Labcyte (U.S.).
  • Detection of enzymatic activity the compound was injected into a 384-well plate by Echo, so that the final concentration was 0-1000 nM (initial concentration 1000 nM, 3-fold dilution, 10 points), and the DMSO content was 0.5%.
  • Prepare 6X detection reagent containing AlphaScreen Protein A Acceptor beads and Anti-Histone H4 (symmetric dimethyl R3) antibody add 5 ⁇ L to each well, and incubate at room temperature for 60 minutes.
  • Prepare 6X detection reagent containing AlphaScreen Streptavidin Donor beads add 5 ⁇ L to each well, and incubate at room temperature for 60 minutes.
  • Envision detection signal value The test results are shown in Table 6.
  • Test Test Example 2 Experiment of Inhibitory Activity of Compounds on Tumor Cell Proliferation
  • Z-138 cells were purchased from ATCC (US); IMDM medium and penicillin-streptomycin were purchased from Sigma (US); horse serum was purchased from Hyclone (US); 96-well plates were purchased from Corning ( U.S.); Cell-Titer Glo reagent was purchased from Promega (U.S.).
  • Cell culture Z-138 cells were cultured in IMDM medium containing 10% horse serum + 1% penicillin-streptomycin at 37°C and 5% CO 2 . Cells in the logarithmic growth phase can be used for experiments.
  • Cell-Titer Glo reagent was used to detect the proliferation inhibitory activity of the compound on Z-138 cells. Adjust the cell concentration, inoculate 96-well plates with 180 ⁇ L per well (500/well), and place them at 37° C. and 5% CO 2 to equilibrate for 10-15 minutes. 20 ⁇ L of compound-containing culture solution was added to each well to make the final concentration 0-300 nM (initial concentration 300 nM, 3-fold dilution, 10 points), and the DMSO content was 0.1%. Cell plates were incubated at 37°C, 5% CO 2 for 8 days.
  • the medium was changed on the fourth day: 100 ⁇ L of the supernatant was slowly aspirated, and 100 ⁇ L of fresh culture medium containing the compound was added to keep the concentration of the compound unchanged. Cell viability was detected by Cell-Titer Glo reagent. The test results are shown in Table 6.
  • Test Test Example 3 Compound's inhibitory activity experiment on SDMA
  • Z-138 cells were purchased from ATCC (US); IMDM medium and penicillin-streptomycin were purchased from Sigma (US); horse serum was purchased from Hyclone (US); Hoechst antibody was purchased from Invitrogen (US) ); Alexa Fluor 488 goat anti-rabbit IgG antibody was purchased from Invitrogen (U.S.); Anti-dimethyl-Arginine symmetric (SYM11) antibody was purchased from Merck (U.S.); DPBS was purchased from Gibco (U.S.); From Cell signaling technology company (USA); paraformaldehyde was purchased from Beijing Suo Laibao Technology Co., Ltd.; 384-well plate and Echo 550 Liquid Handler were purchased from Labcyte Company (USA); ImageXpress Nano was purchased from Molecular Devices company (USA).
  • Cell culture Z-138 cells were cultured in IMDM medium containing 10% horse serum + 1% penicillin-streptomycin at 37°C and 5% CO 2 . Cells in the logarithmic growth phase can be used for experiments.
  • Immunofluorescence detection Immunofluorescence was used to detect the effect of compounds on SDMA in Z-138 cells. The cell concentration was adjusted to 1*10 5 /mL, 40 ⁇ L per well was inoculated into a 384-well plate (4000/well), and placed at 37° C., 5% CO 2 to equilibrate for 10-15 minutes. The compound was injected into a 384-well plate by Echo, so that the final concentration was 0-300 nM (initial concentration 300 nM, 3-fold dilution, 10 points), and the DMSO content was 0.1%. Cell plates were incubated at 37°C, 5% CO 2 for 2 days.
  • CB17-SCID mice were purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd.; DMSO, HP- ⁇ -CD (hydroxypropyl- ⁇ -cyclodextrin), MC (methylcellulose), and acetonitrile were purchased from From Merck (USA), K 2 EDTA anticoagulant tube was purchased from Jiangsu Xinkang Medical Instrument Co., Ltd.
  • mice 6 female CB17-SCID mice (20-30g, 4-6 weeks) were randomly divided into 2 groups, 3 mice in each group.
  • Group 1 was given the compound by tail vein injection, the dose was 2mg/kg, the vehicle was 5% DMSO+95% 10% HP- ⁇ -CD aqueous solution, the second group was orally administered the compound, the dose was 10mg/kg, the vehicle was 0.5% MC aqueous solution.
  • Animals were fed and watered normally before the experiment. Venous blood was collected from mice in each group before administration and at 0.083 (intravenous injection group only), 0.25, 0.5, 1, 2, 4, 6, 8 and 24 hours after administration. The collected whole blood samples were placed in K 2 EDTA anticoagulant tubes, centrifuged for 5 minutes (4000 rpm, 4° C.) and the plasma was collected for testing.
  • mice plasma sample Take 10 ⁇ L of mouse plasma sample, add 150 ⁇ L of acetonitrile solvent (which contains internal standard compound) to precipitate protein, vortex for 0.5 min, centrifuge (4700 rpm, 4 ° C) for 15 min, and supernatant with 0.05% (v/v) formic acid Dilute 2 times with water, inject 3 ⁇ L into LC-MS/MS system (AB Sciex Triple Quad 6500+) for quantitative detection.
  • LC-MS/MS system ABS Sciex Triple Quad 6500+
  • human hepatocytes were purchased from Biopredic Company; mouse hepatocytes were purchased from BioIVT Company; acetonitrile and methanol were purchased from Merck Company; AOPI stain was purchased from Nexcelom Company; dexamethasone was purchased from NIFDC Company; Bao Technology Co., Ltd.; DPBS (10x), GlutaMAX TM -1 (100x) and human recombinant insulin were purchased from Gibco by Life Technologies; fetal bovine serum was purchased from Corning; formic acid was purchased from DIKMAPURE; Isotonic Percoll was purchased from GE Healthcare; Alprazolam was purchased from Supelco; caffeine was purchased from ChromaDex.inc; HEPES, tolbutamide and Williams' Medium E were purchased from Sigma.
  • the specific preparation information of the liver cell resuscitation solution is shown in Table 8 below.
  • the hepatocytes were quickly placed in a 37°C water bath and shaken until all ice crystals were dispersed, sprayed with 70% ethanol and transferred to a biological safety cabinet.
  • the contents of the hepatocyte tubules were poured into a centrifuge tube containing 50 mL of resuscitation medium, which was centrifuged at 100 g for 10 minutes. After centrifugation, aspirate the recovery medium and add enough incubation medium to obtain a cell suspension with a cell density of about 1.5 ⁇ 10 6 cells/mL.
  • Use Cellometer Vision to count liver cells and determine the density of live cells. The survival rate of liver cells must be greater than 75%. Dilute the hepatocyte suspension with incubation medium to a viable cell density of 0.5 ⁇ 106 viable cells/mL.
  • Z138 cells were purchased from ATCC; IMDM medium, penicillin and streptomycin and 0.25% trypsin-EDTA were purchased from Gibco; horse serum and PBS were purchased from Hyclone; Matrigel was purchased from Corning.
  • mice Female, 6-7 weeks old, weighing about 14-20 grams, were purchased from Shanghai Lingchang Biotechnology Co., Ltd. The mice were raised in an SPF-grade environment, and each cage was individually All animals had free access to a standard certified commercial laboratory diet and water ad libitum.
  • PBS phosphate-buffered saline without calcium and magnesium ions
  • Matrigel Matrigel
  • the dosage of the compound of Example 2 is 1.5mg/kg, 5mg/kg or 15mg/kg, PO, administered twice a day (BID) x 3 weeks. 6 mice per group.
  • Tumor diameters were measured twice a week with vernier calipers.
  • Mouse body weights were measured twice a week.
  • the antitumor efficacy of compounds was evaluated by tumor growth inhibition rate TGI (%).
  • TGI (%) [(1-(average tumor volume at the end of administration of a certain treatment group-average tumor volume at the beginning of administration of this treatment group)/(average tumor volume at the end of treatment of the solvent control group-at the beginning of treatment of the solvent control group Mean tumor volume)] x 100%.
  • the compound of Example 2 of the present disclosure had a significant inhibitory effect on tumor growth when administered twice a day at 1.5 mg/kg, 5 mg/kg and 15 mg/kg, and showed a better Dose-response relationship, administration of 15mg/kg twice a day has the effect of shrinking tumors.
  • the compound of Example 2 did not significantly affect the body weight of the mice at the dose tried in this pharmacodynamic experiment, nor did it cause any death of the mice, and the mice could tolerate it.
  • Hygroscopicity evaluation standard (according to the description of hygroscopicity characteristics and the definition of hygroscopicity weight gain in "9103 Drug Hygroscopicity Guiding Principles" in "Chinese Pharmacopoeia” 2015 Edition IV):
  • Hygroscopicity Classification Moisture absorption weight gain*( ⁇ W%) deliquescence Absorb enough water to form a liquid
  • Example 4 Crystal Form C
  • the sample of Example 4 has a moisture absorption weight gain of 2.072% under the condition of 80.0% RH, and has hygroscopicity.
  • Example 5 (crystal form D) has a moisture absorption weight gain of 14.470% under the condition of 80.0% RH, and has hygroscopicity.
  • Example 6 (crystal form A) has a hygroscopic weight gain of 2.318% under the condition of 80.0% RH, and has hygroscopicity.
  • Test instruments and reagents pH meter (model: 914PH; brand: Metrohm), electronic balance (model: XS205DU; brand: METTLER TOLEDO), constant temperature water bath oscillator (model: DHSZ-300A; brand: Taicang Wenerhuijin); Concentrated hydrochloric acid, potassium dihydrogen phosphate, phosphoric acid, sodium hydroxide, sodium acetate trihydrate, glacial acetic acid, 80% aqueous methanol.
  • pH1.0 buffer solution take 9mL of concentrated hydrochloric acid, add purified water to dilute to 1L, stir well, and measure pH;
  • pH4.5 buffer solution Weigh 2.99g of sodium acetate trihydrate and 1.68g of glacial acetic acid, add purified water to dilute to 1L, stir well, and measure pH;
  • pH6.8 buffer solution Weigh 6.805g of potassium dihydrogen phosphate and 0.896g of sodium hydroxide, add purified water to dilute to 1L, stir well, and measure pH.
  • Test instruments and reagents high performance liquid chromatography (model: Agilent 1260; brand: Agilent), constant temperature water bath oscillator (model: DHSZ-300A; brand: Taicang Wenerhuijin), pH meter (model: 914PH; brand Metrohm ); sodium chloride, sodium hydroxide (partially prepared as 1mol/L sodium hydroxide solution for use), concentrated hydrochloric acid (diluted to 1mol/L hydrochloric acid for use), sodium acetate, glacial acetic acid, anhydrous sodium dihydrogen phosphate , Pure Milk, FaSSGF/FaSSIF Powder.
  • pH5.8 medium appropriate amount of purified water.
  • FaSSGF fasting gastric juice
  • Postprandial gastric juice (FeSSGF) preparation Weigh 13.85g of sodium chloride, 2.441g of sodium acetate, 1.028g of glacial acetic acid, add 1000mL of water to dissolve, adjust the pH value to 1mol/L hydrochloric acid solution or 1mol/L sodium hydroxide solution 5.0 to obtain the buffer solution; take 100mL of pure milk and 100mL of the buffer solution, mix well, adjust the pH value to 5.0, and obtain the solution.
  • Fasting intestinal fluid (FaSSIF) preparation Weigh 0.42g sodium hydroxide, 6.19g sodium chloride, 3.44g anhydrous sodium dihydrogen phosphate, add 900mL water to dissolve, and use 1mol/L hydrochloric acid solution or 1mol/L sodium hydroxide solution Adjust the pH value to 6.5, then dilute to 1000mL with water to obtain the buffer; weigh about 112mg of FaSSIF powder into a 50mL measuring bottle, add 25mL of buffer and stir to dissolve, dilute to the mark with buffer, and stand for 2 hours to obtain .
  • FaSSIF Fasting intestinal fluid
  • Preparation of postprandial intestinal fluid (FeSSIF): Weigh 4.04g of sodium hydroxide, 11.87g of sodium chloride, and 8.65g of glacial acetic acid, add 900mL of water to dissolve, and adjust the pH value with 1mol/L hydrochloric acid solution or 1mol/L sodium hydroxide solution to 5.0, then diluted with water to 1000mL to obtain the buffer solution; weigh about 560mg of FaSSIF powder into a 50mL measuring bottle, add 25mL buffer solution and stir to dissolve, dilute to the mark with buffer solution, and stand still to obtain the solution.
  • FaSSIF postprandial intestinal fluid
  • the solubility of Compound I obtained in Example 3 was measured, and its solubility in purified water was measured to be less than 0.5 mg/mL.
  • Compound I and the A crystal form, C crystal form and D crystal form of compound I were packed in a double-layer polyethylene bag plus a layer of aluminum foil bag. Stability under nitrogen conditions.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides a pharmaceutically acceptable salt of a compound as a PRMT5 inhibitor, a crystal form of the pharmaceutically acceptable salt, a preparation method therefor, a pharmaceutical composition comprising the pharmaceutically acceptable salt or the crystal form thereof, and the use of the pharmaceutically acceptable salt or the pharmaceutical composition comprising the pharmaceutically acceptable salt in the preparation of a drug for preventing or treating related pharmacological conditions.

Description

四氢异喹啉类化合物的可药用盐、晶型及其用途Pharmaceutically acceptable salts, crystal forms and uses of tetrahydroisoquinoline compounds
相关申请的交叉引用Cross References to Related Applications
本申请要求以下1件中国发明专利申请的权益和优先权,在此将其全部内容以援引的方式整体并入本文中:This application claims the benefit and priority of the following 1 Chinese invention patent application, which is hereby incorporated by reference in its entirety:
2021年12月30日向中国国家知识产权局提交的第202111664582.X号专利申请。Patent application No. 202111664582.X filed with the State Intellectual Property Office of China on December 30, 2021.
技术领域technical field
本公开内容涉及药物化学领域,具体而言,涉及四氢异喹啉类化合物的可药用盐、所述可药用盐的晶型及其制备方法、包含其的药物组合物以及用途。The present disclosure relates to the field of medicinal chemistry, in particular, to pharmaceutically acceptable salts of tetrahydroisoquinoline compounds, crystal forms of the pharmaceutically acceptable salts, preparation methods thereof, pharmaceutical compositions containing them, and uses thereof.
背景技术Background technique
表观遗传学改变是驱动和维持肿瘤恶性化表型的关键介质。DNA甲基化、组蛋白乙酰化和甲基化、非编码RNA、翻译后修饰的变化都是癌症发生的表观遗传驱动力,而与DNA序列的变化无关。精氨酸甲基化是一类重要的翻译后修饰,通过调节转录和转录后RNA处理影响细胞生长和增殖,凋亡,血管生成和转移。存在三种类型的甲基精氨酸,即ω-NG-单甲基精氨酸(MMA)、ω-NG,N’G-不对称二甲基精氨酸(ADMA)和ω-NG,N’G-对称二甲基精氨酸(SDMA)。这种修饰是由蛋白质精氨酸甲基转移酶(PRMT)家族催化的,从S-腺苷甲硫氨酸(AdoMet)把甲基转移到组蛋白和非组蛋白的精氨酸侧链。在人类基因组中注释了九个PRMT基因,基于产生的甲基精氨酸类型分为I型(PRMT1,2,3,4,6和8)、II型(PRMT5和PRMT9)和III型酶(PRMT7)。PRMT5主要是II型酶,可催化精氨酸的对称二甲基化。PRMT5是在检测与Janus酪氨酸激酶(Jak2)相互作用蛋白的双杂交实验中被首次发现的。Epigenetic changes are key mediators that drive and maintain the malignant phenotype of tumors. Changes in DNA methylation, histone acetylation and methylation, non-coding RNA, and post-translational modifications are all epigenetic drivers of cancer development independent of changes in DNA sequence. Arginine methylation is an important class of post-translational modifications that affect cell growth and proliferation, apoptosis, angiogenesis and metastasis by regulating transcription and post-transcriptional RNA processing. There are three types of methylarginine, namely ω-NG-monomethylarginine (MMA), ω-NG, N'G-asymmetric dimethylarginine (ADMA) and ω-NG, N'G-symmetric dimethylarginine (SDMA). This modification is catalyzed by the protein arginine methyltransferase (PRMT) family, which transfers a methyl group from S-adenosylmethionine (AdoMet) to the arginine side chain of histones and non-histones. Nine PRMT genes were annotated in the human genome, classified based on the type of methylarginine produced into type I (PRMT1, 2, 3, 4, 6 and 8), type II (PRMT5 and PRMT9) and type III enzymes ( PRMT7). PRMT5 is primarily a type II enzyme that catalyzes the symmetric dimethylation of arginine. PRMT5 was first discovered in a two-hybrid assay detecting proteins interacting with Janus tyrosine kinase (Jak2).
PRMT5是一种通用的转录抑制因子,与其他转录因子形成复合物,包括BRG1和hBRM,Blimp1以及Snail。PRMT5通过对多种细胞质和细胞核中的底物的甲基化,包括组蛋白H4残基Arg3(H4R3)和H3残基Arg8(H3R8)而参与多种不同的细胞生物学过程。H4R3甲基化与转录抑制相关,而H3R8甲基化被认为既与转录激活有关,又和转录抑制有关。PRMT5除了直接诱导抑制性组蛋白标记外,该酶在基因沉默中的作用还通过形成多抑制蛋白复合物来介导,包括NuRD组分、HDACs、MDB蛋白和DNA甲基转移酶。PRMT5通过与一些结合蛋白的相互作用进而影响其底物特异性。这种蛋白质复合物中的核心成分是MEP50。MEP50对于PRMT5的酶学活性是必须的。研究发现,PRMT5可以甲基化参与RNA剪接的蛋白,比如SmD3,可用于跟踪细胞生物PRMT5的化学活性。PRMT5 is a general transcriptional repressor that forms a complex with other transcription factors, including BRG1 and hBRM, Blimp1, and Snail. PRMT5 participates in a variety of different cellular biological processes through the methylation of a variety of substrates in the cytoplasm and nucleus, including histone H4 residue Arg3 (H4R3) and H3 residue Arg8 (H3R8). H4R3 methylation is associated with transcriptional repression, while H3R8 methylation is thought to be associated with both transcriptional activation and transcriptional repression. In addition to the direct induction of repressive histone marks by PRMT5, the enzyme's role in gene silencing is also mediated through the formation of multiple arrestin complexes, including NuRD components, HDACs, MDB proteins, and DNA methyltransferases. PRMT5 affects its substrate specificity by interacting with some binding proteins. A central component in this protein complex is MEP50. MEP50 is required for the enzymatic activity of PRMT5. The study found that PRMT5 can methylate proteins involved in RNA splicing, such as SmD3, which can be used to track the chemical activity of cellular biological PRMT5.
PRMT5在肿瘤发生中起重要作用。研究发现PRMT5在多种肿瘤中的表达上调,包括淋巴瘤、肺癌、乳腺癌和结直肠癌。此外,PRMT5表达在套细胞淋巴瘤(MCL)病人样本中增高,而PRMT5敲除则可以抑制MCL细胞增殖,表明PRMT5在MCL中起重要作用。PRMT5过表达促进细胞增生,在黑色素瘤、乳腺癌和肺癌细胞系中,PRMT5敲除则可以抑制这些细胞的增殖。因此,PRMT5是癌症治疗的潜在靶点。PRMT5 plays an important role in tumorigenesis. Studies have found that the expression of PRMT5 is upregulated in a variety of tumors, including lymphoma, lung cancer, breast cancer and colorectal cancer. In addition, PRMT5 expression was increased in mantle cell lymphoma (MCL) patient samples, and PRMT5 knockout could inhibit the proliferation of MCL cells, indicating that PRMT5 plays an important role in MCL. PRMT5 overexpression promotes cell proliferation, and in melanoma, breast cancer, and lung cancer cell lines, PRMT5 knockdown can inhibit the proliferation of these cells. Therefore, PRMT5 is a potential target for cancer therapy.
甲基硫腺苷磷酸化酶(MTAP)的丧失赋予了细胞对PRMT5及其结合蛋白WDR77的选择性依赖。MTAP由于与通常缺失的肿瘤抑制基因CDKN2A靠近而经常丢失。携带MTAP缺失的细胞的胞内甲硫基腺苷(MTA,被MTAP裂解的代谢物)浓度增加。此外,MTA特异性抑制PRMT5的酶活性。与MTAP表达的细胞相比,MTA或PRMT5小分子抑制剂显著抑制MTAP缺失的癌细胞系的细胞活力。Loss of methylthioadenosine phosphorylase (MTAP) confers selective dependence on PRMT5 and its binding protein WDR77. MTAP is frequently lost due to its proximity to the commonly deleted tumor suppressor gene CDKN2A. Cells carrying MTAP deletions have increased intracellular concentrations of methylthioadenosine (MTA, a metabolite cleaved by MTAP). In addition, MTA specifically inhibits the enzymatic activity of PRMT5. MTA or PRMT5 small-molecule inhibitors significantly inhibited the cell viability of MTAP-deficient cancer cell lines compared with MTAP-expressing cells.
因此,本领域需要开发能够抑制PRMT5的活性并治疗各种PRMT5相关疾病的小分子活性化合物。Therefore, there is a need in the art to develop small molecule active compounds that can inhibit the activity of PRMT5 and treat various PRMT5-related diseases.
PCT/CN2021/103597(申请日2021年6月30日)描述了一种化合物1-乙基-4-((R)-2-羟基-2-((S)-1,2,3,4-四氢异喹啉-3-基)乙基)-8-(2-甲氧基-7-氮杂螺[3.5]壬烷-7-羰基)-1,2,3,4-四氢-5H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000001
-5-酮(化合物I)盐酸盐,研究发现,所述化合物I盐酸盐具有较好的PRMT5酶学抑制、细胞增殖抑制和细胞SDMA抑制活性,以及良好的药代动力学性质和肝细胞代谢稳定性,并在小鼠皮下移植瘤Z-138模型中显示出显著的肿瘤生长抑制作用,并呈现较好的剂量反应关系。
PCT/CN2021/103597 (application date June 30, 2021) describes a compound 1-ethyl-4-((R)-2-hydroxy-2-((S)-1,2,3,4 -Tetrahydroisoquinolin-3-yl)ethyl)-8-(2-methoxy-7-azaspiro[3.5]nonane-7-carbonyl)-1,2,3,4-tetrahydro -5H-Benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000001
-5-keto (compound I) hydrochloride, the study found that the compound I hydrochloride has good PRMT5 enzyme inhibition, cell proliferation inhibition and cell SDMA inhibitory activity, and good pharmacokinetic properties and liver function. Cell metabolism stability, and showed significant tumor growth inhibition in mouse subcutaneous xenograft Z-138 model, and showed a good dose-response relationship.
Figure PCTCN2022143995-appb-000002
Figure PCTCN2022143995-appb-000002
发明内容Contents of the invention
本公开内容提供了化合物I的可药用盐,其中所述可药用盐选自一盐酸盐、氢溴酸盐、1,5-萘二磺酸盐、草酸盐、柠檬酸盐、硫酸盐、磷酸盐、L-酒石酸盐、L-苹果酸盐、琥珀酸盐、己二酸盐、富马酸盐、草酸盐、丙酸盐、苯甲酸盐、乙酸盐、甲酸盐或L-精氨酸盐,优选一盐酸盐、氢溴酸盐或1,5-萘二磺酸盐,更优选一盐酸盐。The present disclosure provides a pharmaceutically acceptable salt of Compound I, wherein the pharmaceutically acceptable salt is selected from monohydrochloride, hydrobromide, 1,5-naphthalene disulfonate, oxalate, citrate, Sulfate, Phosphate, L-Tartrate, L-Malate, Succinate, Adipate, Fumarate, Oxalate, Propionate, Benzoate, Acetate, Formic Acid salt or L-arginine salt, preferably monohydrochloride, hydrobromide or 1,5-naphthalene disulfonate, more preferably monohydrochloride.
在一些实施方案中,所述化合物I的可药用盐中,化合物I与酸分子的摩尔比约为1:2~2:1,优选选自1:2、1:1或2:1,更优选为1:1。In some embodiments, in the pharmaceutically acceptable salt of Compound I, the molar ratio of Compound I to acid molecules is about 1:2 to 2:1, preferably selected from 1:2, 1:1 or 2:1, More preferably 1:1.
在一些实施方案中,提供了化合物I的一盐酸盐,其中化合物I与盐酸的摩尔比约为1:1。In some embodiments, there is provided a monohydrochloride salt of Compound I, wherein the molar ratio of Compound I to hydrochloric acid is about 1:1.
在一些实施方案中,提供了化合物I的氢溴酸盐。In some embodiments, a hydrobromide salt of Compound I is provided.
在一些实施方案中,提供了化合物I的1,5-萘二磺酸盐。In some embodiments, a 1,5-naphthalene disulfonate salt of Compound I is provided.
在一些实施方案中,所述化合物I的可药用盐以未溶剂化或溶剂化的形式存在。In some embodiments, the pharmaceutically acceptable salts of Compound I exist in unsolvated or solvated form.
本公开内容还提供了所述化合物I的可药用盐的固体形式。The present disclosure also provides solid forms of the pharmaceutically acceptable salts of Compound I.
在一些实施方案中,所述化合物I的可药用盐的固体形式选自无定型或结晶形式。In some embodiments, the solid form of the pharmaceutically acceptable salt of Compound I is selected from amorphous or crystalline forms.
本公开内容还提供了制备前述化合物I的可药用盐的方法,包括:化合物I与酸成盐的步骤,所述酸选自盐酸、氢溴酸、1,5-萘二磺酸、草酸、柠檬酸、硫酸、磷酸、L-酒石酸、L-苹果酸、琥珀酸、己二酸、富马酸、草酸、丙酸、苯甲酸、乙酸、甲酸或L-精氨酸,优选盐酸、氢溴酸或1,5-萘二磺酸,更优选盐酸。The present disclosure also provides a method for preparing a pharmaceutically acceptable salt of the aforementioned compound I, comprising: a step of forming a salt of compound I with an acid, the acid being selected from hydrochloric acid, hydrobromic acid, 1,5-naphthalene disulfonic acid, oxalic acid , citric acid, sulfuric acid, phosphoric acid, L-tartaric acid, L-malic acid, succinic acid, adipic acid, fumaric acid, oxalic acid, propionic acid, benzoic acid, acetic acid, formic acid or L-arginine, preferably hydrochloric acid, hydrogen Bromic acid or 1,5-naphthalenedisulfonic acid, more preferably hydrochloric acid.
本公开所述成盐反应所用溶剂选自乙酸乙酯、四氢呋喃、乙腈、丙酮、乙酸甲酯、异丙醇、甲醇或甲苯中的至少一种,优选选自异丙醇、乙酸甲酯、乙酸乙酯、甲醇或丙酮,更优选异丙醇或乙酸乙酯。The solvent used in the salt-forming reaction described in the present disclosure is selected from at least one of ethyl acetate, tetrahydrofuran, acetonitrile, acetone, methyl acetate, isopropanol, methanol or toluene, preferably selected from isopropanol, methyl acetate, acetic acid ethyl ester, methanol or acetone, more preferably isopropanol or ethyl acetate.
在一些实施方案中,制备前述可药用盐的方法还包括挥发溶剂、搅拌析晶、过滤、洗涤或干燥步骤中的至少一种。In some embodiments, the method for preparing the aforementioned pharmaceutically acceptable salt further includes at least one of the steps of volatilizing the solvent, crystallizing with stirring, filtering, washing or drying.
在一些实施方案中,所述化合物I的可药用盐为结晶形式,优选盐酸盐晶型、氢溴酸盐晶型、1,5-萘二磺酸盐晶型,更优选一盐酸盐晶型。In some embodiments, the pharmaceutically acceptable salt of Compound I is in crystalline form, preferably hydrochloride crystal form, hydrobromide salt crystal form, 1,5-naphthalene disulfonate salt crystal form, more preferably monohydrochloride Salt crystal form.
在一些实施方案中,所述化合物I的盐酸盐晶型为化合物I一盐酸盐的A晶型,所述A晶型以衍射角2θ表示的X-射线粉末衍射图谱,在5.42±0.2 o、6.60±0.2 o、10.36±0.2 o、13.60±0.2 o、14.24±0.2 o、15.83±0.2 o、16.62±0.2 o、17.65±0.2 o、19.29±0.2 o、19.86±0.2 o、21.37±0.2 o、23.88±0.2 o处有衍射峰。 In some embodiments, the hydrochloride crystal form of the compound I is the A crystal form of the compound I-hydrochloride, and the X-ray powder diffraction pattern of the A crystal form represented by the diffraction angle 2θ is 5.42 ± 0.2 o , 6.60± 0.2o , 10.36± 0.2o , 13.60±0.2o , 14.24±0.2o , 15.83±0.2o, 16.62 ±0.2o , 17.65±0.2o, 19.29±0.2o , 19.86±0.2o , 21 .37± 0.2 o and 23.88±0.2 o have diffraction peaks.
在一些实施方案中,所述A晶型,以衍射角2θ表示的X-射线粉末衍射图谱,在5.42±0.2 o、6.60±0.2 o、9.76±0.2 o、10.36±0.2 o、13.00±0.2 o、13.60±0.2 o、14.24±0.2 o、15.83±0.2 o、16.62±0.2 o、17.65±0.2 o、18.28±0.2 o、19.29±0.2 o、19.86±0.2 o、20.61±0.2 o、21.37±0.2 o、23.88±0.2 o、25.08±0.2 o、28.22±0.2 o处有衍射峰。 In some embodiments, the crystal form A has an X-ray powder diffraction pattern represented by diffraction angle 2θ at 5.42±0.2 ° , 6.60±0.2 ° , 9.76±0.2 ° , 10.36±0.2 ° , 13.00±0.2 ° , 13.60± 0.2o , 14.24± 0.2o , 15.83± 0.2o , 16.62±0.2o, 17.65±0.2o, 18.28±0.2o, 19.29 ± 0.2o , 19.86±0.2o , 20.61 ± 0.2o, 21. 37± 0.2o , 23.88±0.2 o , 25.08±0.2 o , 28.22±0.2 o have diffraction peaks.
在一些实施方案中,所述A晶型,以衍射角2θ表示的X-射线粉末衍射图谱,在5.42±0.2 o、6.60±0.2 o、6.83±0.2 o、9.76±0.2 o、10.36±0.2 o、13.00±0.2 o、13.60±0.2 o、14.24±0.2 o、15.27±0.2 o、15.83±0.2 o、16.62±0.2 o、17.30±0.2 o、17.65±0.2 o、18.28±0.2 o、19.29±0.2 o、19.86±0.2 o、20.61±0.2 o、21.37±0.2 o、21.88±0.2 o、23.88±0.2 o、25.08±0.2 o、25.56±0.2 o、26.51±0.2 o、27.22±0.2 o、28.22±0.2 o处有衍射峰。 In some embodiments, the crystal form A has an X-ray powder diffraction pattern represented by diffraction angle 2θ at 5.42±0.2 ° , 6.60±0.2 ° , 6.83±0.2 ° , 9.76±0.2 ° , 10.36±0.2 ° , 13.00± 0.2o , 13.60 ± 0.2o , 14.24± 0.2o , 15.27±0.2o , 15.83±0.2o, 16.62±0.2o, 17.30± 0.2o , 17.65±0.2o, 18.28± 0.2o , 19. 29± 0.2o , 19.86±0.2 o , 20.61 ±0.2 o , 21.37±0.2 o , 21.88±0.2 o , 23.88±0.2 o , 25.08±0.2 o , 25.56±0.2 o , 26.51±0.2 o , 27.22±0.2 o , 28. 22± 0.2o There are diffraction peaks.
在一些实施方案中,所述A晶型,以衍射角2θ表示的X-射线粉末衍射图谱,在5.42±0.2 o、6.60±0.2 o、6.83±0.2 o、9.76±0.2 o、10.36±0.2 o、13.00±0.2 o、13.60±0.2 o、14.24±0.2 o、15.27±0.2 o、15.83±0.2 o、16.62±0.2 o、17.30±0.2 o、17.65±0.2 o、18.28±0.2 o、19.29±0.2 o、19.86±0.2 o、20.15±0.2 o、20.61±0.2 o、21.37±0.2 o、21.88±0.2 o、23.23±0.2 o、23.88±0.2 o、25.08±0.2 o、25.56±0.2 o、26.51±0.2 o、27.22±0.2 o、28.22±0.2 o、29.42±0.2 o、30.21±0.2 o、30.74±0.2 o、34.79±0.2 o、37.74±0.2 o处有衍射峰。 In some embodiments, the crystal form A has an X-ray powder diffraction pattern represented by diffraction angle 2θ at 5.42±0.2 ° , 6.60±0.2 ° , 6.83±0.2 ° , 9.76±0.2 ° , 10.36±0.2 ° , 13.00± 0.2o , 13.60 ± 0.2o , 14.24± 0.2o , 15.27±0.2o , 15.83±0.2o, 16.62±0.2o, 17.30± 0.2o , 17.65±0.2o, 18.28± 0.2o , 19. 29± 0.2o , 19.86± 0.2o , 20.15± 0.2o , 20.61± 0.2o , 21.37±0.2o , 21.88±0.2o, 23.23±0.2o, 23.88± 0.2o , 25.08±0.2o , 25.56 ± 0.2o , 26. 51± 0.2o , 27.22±0.2 o , 28.22±0.2 o , 29.42±0.2 o , 30.21±0.2 o , 30.74±0.2 o , 34.79±0.2 o , 37.74±0.2 o have diffraction peaks.
在一些实施方案中,所述A晶型的X-射线粉末衍射图基本上如图8所示。In some embodiments, the X-ray powder diffraction pattern of Form A is substantially as shown in FIG. 8 .
在一些实施方案中,所述A晶型具有在259.27±5.0℃下的峰值的DSC谱图。In some embodiments, the crystalline Form A has a DSC spectrum with a peak at 259.27±5.0°C.
在一些实施方案中,所述A晶型的DSC谱图基本上如图9所示。In some embodiments, the DSC spectrum of Form A is substantially as shown in FIG. 9 .
本公开内容还提供了制备化合物I一盐酸盐的A晶型的方法,包括如下步骤:The present disclosure also provides a method for preparing the A crystal form of compound I-hydrochloride, comprising the steps of:
1)称取适量化合物I,加入溶剂(i)中,室温溶清或加热溶清;1) Weighing an appropriate amount of compound I, adding it to the solvent (i), and dissolving at room temperature or heating;
2)加入氯化氢水溶液或氯化氢的有机溶剂溶液,析晶。2) Add aqueous hydrogen chloride solution or organic solvent solution of hydrogen chloride to crystallize.
在一些实施方案中,所述A晶型制备方法的步骤2)之后还存在可选的步骤3),即采用良溶剂对步骤2)所得晶体进行溶解,得到溶液后再滴加不良溶剂,析晶。In some embodiments, there is an optional step 3) after step 2) of the preparation method of the A crystal form, that is, a good solvent is used to dissolve the crystal obtained in step 2), and after the solution is obtained, a poor solvent is added dropwise to analyze crystal.
在一些实施方案中,所述A晶型制备方法的步骤2)之后还存在可选的步骤3’),即将步骤2)所得晶型溶于甲醇后减压浓缩得到化合物I一盐酸盐无定型物,然后将该无定型物溶于溶剂(ii),继而通过打浆或反溶剂滴加的方式析出晶体。In some embodiments, there is an optional step 3') after step 2) of the preparation method of crystal form A, that is, dissolving the crystal form obtained in step 2) in methanol and concentrating under reduced pressure to obtain compound I-hydrochloride free Shaped matter, and then dissolve the amorphous matter in solvent (ii), and then precipitate crystals by beating or adding anti-solvent dropwise.
在一些实施方案中,所述A晶型制备方法步骤1)中的溶剂(i)选自乙酸甲酯、乙酸乙酯、丙酮或异丙醇,优选异丙醇或乙酸乙酯。In some embodiments, the solvent (i) in step 1) of the preparation method for crystal form A is selected from methyl acetate, ethyl acetate, acetone or isopropanol, preferably isopropanol or ethyl acetate.
在一些实施方案中,所述A晶型制备方法步骤1)中所述溶剂(i)的体积(mL)为化合物重量(g)的5~30倍,优选5~20倍,更优选10~15倍。In some embodiments, the volume (mL) of the solvent (i) in step 1) of the preparation method for crystal form A is 5 to 30 times, preferably 5 to 20 times, more preferably 10 to 30 times the weight of the compound (g). 15 times.
在一些实施方案中,所述A晶型制备方法步骤2)中加入氯化氢的有机溶剂溶液,优选加入氯化氢-乙酸乙酯溶液。In some embodiments, an organic solvent solution of hydrogen chloride, preferably a hydrogen chloride-ethyl acetate solution, is added in step 2) of the preparation method for crystal form A.
在一些实施方案中,所述A晶型制备方法步骤2)中所述氯化氢的有机溶剂溶液是在-10~10℃,优选0~10℃下滴加入反应体系,并保温反应。In some embodiments, the organic solvent solution of hydrogen chloride in step 2) of the preparation method of crystal form A is added dropwise to the reaction system at -10-10°C, preferably 0-10°C, and kept for reaction.
在一些实施方案中,所述A晶型制备方法步骤2)中的氯化氢摩尔用量为步骤1中化合物I摩尔用量的0.95~1.05倍。In some embodiments, the molar amount of hydrogen chloride used in step 2) of the preparation method for crystal form A is 0.95-1.05 times the molar amount of compound I in step 1.
在一些实施方案中,所述A晶型制备方法步骤2)的反应时间为0.5~3h,优选1~2h。In some embodiments, the reaction time of step 2) of the preparation method for crystal form A is 0.5-3 hours, preferably 1-2 hours.
在一些实施方案中,所述A晶型制备方法步骤3)中的良溶剂选自甲醇、二氯甲烷或氯仿,优选甲醇;不良溶剂选自异丙醇、丙酮或异丙醚,优选异丙醇。In some embodiments, the good solvent in step 3) of the preparation method of crystal form A is selected from methanol, dichloromethane or chloroform, preferably methanol; the poor solvent is selected from isopropanol, acetone or isopropyl ether, preferably isopropyl alcohol.
在一些实施方案中,所述A晶型制备方法步骤3’)中通过打浆的方式制备晶体,且所述溶剂(ii)选自丙酮、4-甲基-2-戊酮、2-丁酮、异丙醚、甲基叔丁基醚、乙醚、正丙醇、异丙醇、乙醇、乙酸甲酯、乙酸乙酯、乙酸异丙酯、乙酸丁酯、乙腈、四氢呋喃、环己烷、甲苯、水或丙酮水溶液中的一种或多种,优选乙酸乙酯。In some embodiments, crystals are prepared by beating in step 3') of the preparation method of crystal form A, and the solvent (ii) is selected from acetone, 4-methyl-2-pentanone, and 2-butanone , isopropyl ether, methyl tert-butyl ether, diethyl ether, n-propanol, isopropanol, ethanol, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, acetonitrile, tetrahydrofuran, cyclohexane, toluene , one or more of water or acetone aqueous solution, preferably ethyl acetate.
在一些实施方案中,所述A晶型制备方法步骤3’)中通过反溶剂滴加的方式制备晶体,且所述溶剂(ii)选自二氯甲烷、氯仿或甲醇,优选甲醇;所述反溶剂选自丙酮、异丙醚、乙酸甲酯、乙腈或四氢呋喃,优选丙酮。In some embodiments, in step 3') of the preparation method of crystal form A, crystals are prepared by adding anti-solvent dropwise, and the solvent (ii) is selected from dichloromethane, chloroform or methanol, preferably methanol; The anti-solvent is selected from acetone, isopropyl ether, methyl acetate, acetonitrile or tetrahydrofuran, preferably acetone.
在一些实施方案中,所述化合物I的盐酸盐晶型为化合物I一盐酸盐的B晶型,所述B晶型以衍射角2θ表示的X-射线粉末衍射图谱,在5.16±0.2 o、9.86±0.2 o、10.42±0.2 o、10.76±0.2 o、11.36±0.2 o、15.68±0.2 o、16.31±0.2 o、17.29±0.2 o、17.98±0.2 o、18.89±0.2 o、19.83±0.2 o、20.99±0.2 o处有衍射峰。 In some embodiments, the hydrochloride crystal form of the compound I is the B crystal form of the compound I-hydrochloride, and the X-ray powder diffraction pattern of the B crystal form represented by the diffraction angle 2θ is at 5.16±0.2 o , 9.86± 0.2o , 10.42± 0.2o , 10.76±0.2o , 11.36±0.2o , 15.68±0.2o , 16.31±0.2o, 17.29±0.2o , 17.98±0.2o , 18.89± 0.2o , 19 .83±0.2 There are diffraction peaks at o and 20.99±0.2 o .
在一些实施方案中,所述B晶型,以衍射角2θ表示的X-射线粉末衍射图谱,在5.16±0.2 o、9.86±0.2 o、10.42±0.2 o、10.76±0.2 o、11.36±0.2 o、14.46±0.2 o、15.68±0.2 o、16.31±0.2 o、17.29±0.2 o、17.98±0.2 o、18.89±0.2 o、19.83±0.2 o、20.99±0.2 o、22.13±0.2 o、22.72±0.2 o、23.61±0.2 o、25.94±0.2 o处有衍射峰。 In some embodiments, the crystal form B has an X-ray powder diffraction pattern represented by diffraction angle 2θ at 5.16±0.2 ° , 9.86±0.2 ° , 10.42±0.2 ° , 10.76±0.2 ° , 11.36±0.2 ° , 14.46± 0.2o , 15.68± 0.2o , 16.31± 0.2o , 17.29±0.2o , 17.98± 0.2o , 18.89± 0.2o , 19.83±0.2o , 20.99±0.2o, 22.13 ± 0.2o , 22. 72± 0.2o , 23.61±0.2 o and 25.94±0.2 o have diffraction peaks.
在一些实施方案中,所述B晶型,以衍射角2θ表示的X-射线粉末衍射图谱,在5.16±0.2 o、 6.33±0.2 o、8.98±0.2 o、9.86±0.2 o、10.42±0.2 o、10.76±0.2 o、11.36±0.2 o、12.82±0.2 o、14.46±0.2 o、15.68±0.2 o、16.31±0.2 o、17.29±0.2 o、17.31±0.2 o、17.98±0.2 o、18.89±0.2 o、19.30±0.2 o、19.83±0.2 o、20.99±0.2 o、21.73±0.2 o、22.13±0.2 o、22.72±0.2 o、23.61±0.2 o、23.73±0.2 o、24.71±0.2 o、25.94±0.2 o、26.38±0.2 o、37.29±0.2 o处有衍射峰。 In some embodiments, the crystal form B has an X-ray powder diffraction pattern represented by diffraction angle 2θ at 5.16±0.2 ° , 6.33±0.2 ° , 8.98±0.2 ° , 9.86±0.2 ° , 10.42±0.2 ° , 10.76± 0.2o , 11.36± 0.2o , 12.82± 0.2o , 14.46±0.2o , 15.68± 0.2o , 16.31±0.2o, 17.29± 0.2o , 17.31±0.2o , 17.98±0.2o , 18. 89± 0.2o , 19.30± 0.2o , 19.83± 0.2o , 20.99± 0.2o , 21.73±0.2o , 22.13±0.2o, 22.72± 0.2o , 23.61± 0.2o , 23.73±0.2o , 24.71± 0.2o , 25. 94± 0.2o , 26.38±0.2 o and 37.29±0.2 o have diffraction peaks.
在一些实施方案中,所述B晶型的X-射线粉末衍射图基本上如图11所示。In some embodiments, the X-ray powder diffraction pattern of Form B is substantially as shown in FIG. 11 .
在一些实施方案中,所述B晶型的DSC谱图基本上如图12所示。In some embodiments, the DSC spectrum of Form B is substantially as shown in FIG. 12 .
在一些实施方案中,所述化合物I的氢溴酸盐晶型为C晶型,所述C晶型以衍射角2θ表示的X-射线粉末衍射图谱,在6.45±0.2 o、9.49±0.2 o、10.01±0.2 o、12.96±0.2 o、13.45±0.2 o、13.81±0.2 o、15.50±0.2 o、19.13±0.2 o、20.13±0.2 o、20.78±0.2 o、23.31±0.2 o处有衍射峰。 In some embodiments, the hydrobromide salt crystal form of Compound I is the C crystal form, and the X-ray powder diffraction pattern of the C crystal form represented by the diffraction angle 2θ is at 6.45±0.2 ° , 9.49±0.2 ° , 10.01±0.2 o , 12.96±0.2 o , 13.45±0.2 o , 13.81±0.2 o , 15.50±0.2 o , 19.13±0.2 o , 20.13±0.2 o , 20.78±0.2 o , 23.31± 0.2 o .
在一些实施方案中,所述C晶型,以衍射角2θ表示的X-射线粉末衍射图谱,在6.45±0.2 o、9.49±0.2 o、10.01±0.2 o、12.96±0.2 o、13.45±0.2 o、13.81±0.2 o、15.50±0.2 o、17.30±0.2 o、19.13±0.2 o、19.46±0.2 o、20.13±0.2 o、20.78±0.2 o、23.31±0.2 o、24.16±0.2 o、24.55±0.2 o、24.80±0.2 o、25.62±0.2 o、27.00±0.2 o、28.87±0.2 o、28.95±0.2 o处有衍射峰。 In some embodiments, the crystal form C has an X-ray powder diffraction pattern represented by diffraction angle 2θ at 6.45±0.2 ° , 9.49±0.2 ° , 10.01±0.2 ° , 12.96±0.2 ° , 13.45±0.2 ° , 13.81± 0.2o , 15.50± 0.2o , 17.30± 0.2o , 19.13±0.2o , 19.46± 0.2o , 20.13±0.2o, 20.78± 0.2o , 23.31±0.2o , 24.16±0.2o , 24. 55± 0.2o , 24.80±0.2 o , 25.62±0.2 o , 27.00±0.2 o , 28.87±0.2 o , 28.95±0.2 o have diffraction peaks.
在一些实施方案中,所述C晶型,以衍射角2θ表示的X-射线粉末衍射图谱,在6.45±0.2 o、9.49±0.2 o、10.01±0.2 o、12.96±0.2 o、13.45±0.2 o、13.81±0.2 o、15.50±0.2 o、16.29±0.2 o、16.95±0.2 o、17.30±0.2 o、19.13±0.2 o、19.46±0.2 o、20.13±0.2 o、20.78±0.2 o、23.31±0.2 o、23.71±0.2 o、24.16±0.2 o、24.55±0.2 o、24.80±0.2 o、25.62±0.2 o、26.05±0.2 o、27.00±0.2 o、27.44±0.2 o、28.87±0.2 o、28.95±0.2 o、30.65±0.2 o、31.74±0.2 o、32.78±0.2 o、34.03±0.2 o、34.97±0.2 o处有衍射峰。 In some embodiments, the crystal form C has an X-ray powder diffraction pattern represented by diffraction angle 2θ at 6.45±0.2 ° , 9.49±0.2 ° , 10.01±0.2 ° , 12.96±0.2 ° , 13.45±0.2 ° , 13.81± 0.2o , 15.50± 0.2o , 16.29± 0.2o , 16.95±0.2o , 17.30± 0.2o , 19.13±0.2o, 19.46 ±0.2o , 20.13±0.2o, 20.78± 0.2o , 23. 31± 0.2o , 23.71± 0.2o , 24.16± 0.2o , 24.55± 0.2o , 24.80±0.2o , 25.62±0.2o, 26.05±0.2o, 27.00± 0.2o , 27.44±0.2o , 28.87± 0.2o , 28. 95± 0.2o , 30.65±0.2 o , 31.74±0.2 o , 32.78±0.2 o , 34.03±0.2 o , 34.97±0.2 o have diffraction peaks.
在一些实施方案中,所述C晶型的X-射线粉末衍射图谱基本上如图2所示。In some embodiments, the X-ray powder diffraction pattern of Form C is substantially as shown in FIG. 2 .
在一些实施方案中,所述C晶型具有在238.84±5.0℃下的峰值的DSC谱图。In some embodiments, the crystalline Form C has a DSC spectrum with a peak at 238.84±5.0°C.
在一些实施方案中,所述C晶型的DSC谱图基本上如图3所示。In some embodiments, the DSC spectrum of Form C is substantially as shown in FIG. 3 .
在一些实施方案中,所述化合物I的1,5-萘二磺酸盐为D晶型,所述D晶型以衍射角2θ表示的X-射线粉末衍射图谱,在4.02±0.2 o、9.89±0.2 o、11.49±0.2 o、12.74±0.2 o、15.68±0.2 o、15.69±0.2 o、16.15±0.2 o、18.62±0.2 o、18.93±0.2 o、19.67±0.2 o、19.79±0.2 o、21.26±0.2 o、24.36±0.2 o、27.56±0.2 o处有衍射峰。 In some embodiments, the 1,5-naphthalene disulfonate salt of Compound I is the D crystal form, and the X-ray powder diffraction pattern of the D crystal form represented by the diffraction angle 2θ is at 4.02±0.2 o , 9.89 ± 0.2o , 11.49± 0.2o , 12.74± 0.2o , 15.68±0.2o, 15.69± 0.2o , 16.15±0.2o, 18.62± 0.2o , 18.93±0.2o, 19.67± 0.2o , 19.79±0.2o o , 21.26 There are diffraction peaks at ±0.2 o , 24.36±0.2 o , and 27.56±0.2 o .
在一些实施方案中,所述D晶型,以衍射角2θ表示的X-射线粉末衍射图谱,在4.02±0.2 o、8.06±0.2 o、8.79±0.2 o、9.89±0.2 o、11.49±0.2 o、12.74±0.2 o、13.94±0.2 o、15.68±0.2 o、15.69±0.2 o、16.15±0.2 o、17.21±0.2 o、18.62±0.2 o、18.93±0.2 o、19.67±0.2 o、19.79±0.2 o、20.57±0.2 o、21.26±0.2 o、24.36±0.2 o、25.64±0.2 o、27.56±0.2 o处有衍射峰。 In some embodiments, the crystal form D has an X-ray powder diffraction pattern represented by diffraction angle 2θ at 4.02±0.2 ° , 8.06±0.2 ° , 8.79±0.2 ° , 9.89±0.2 ° , 11.49±0.2 ° , 12.74± 0.2o , 13.94± 0.2o , 15.68± 0.2o , 15.69± 0.2o , 16.15±0.2o, 17.21±0.2o, 18.62± 0.2o , 18.93±0.2o , 19.67± 0.2o , 19. 79± 0.2o , 20.57±0.2 o , 21.26±0.2 o , 24.36±0.2 o , 25.64±0.2 o , 27.56±0.2 o have diffraction peaks.
在一些实施方案中,所述D晶型,以衍射角2θ表示的X-射线粉末衍射图谱,在4.02±0.2 o、8.06±0.2 o、8.79±0.2 o、9.89±0.2 o、11.49±0.2 o、12.10±0.2 o、12.74±0.2 o、13.94±0.2 o、14.94±0.2 o、15.68±0.2 o、15.69±0.2 o、16.15±0.2 o、17.21±0.2 o、17.63±0.2 o、17.97±0.2 o、18.62±0.2 o、18.93±0.2 o、19.67±0.2 o、19.79±0.2 o、20.20±0.2 o、20.57±0.2 o、21.26±0.2 o、22.61±0.2 o、23.65±0.2 o、24.36±0.2 o、25.64±0.2 o、27.56±0.2 o、28.45±0.2 o、32.64±0.2 o、36.86±0.2 o、37.78±0.2 o处有衍射峰。 In some embodiments, the crystal form D has an X-ray powder diffraction pattern represented by diffraction angle 2θ at 4.02±0.2 ° , 8.06±0.2 ° , 8.79±0.2 ° , 9.89±0.2 ° , 11.49±0.2 ° , 12.10± 0.2o , 12.74± 0.2o , 13.94± 0.2o , 14.94± 0.2o , 15.68± 0.2o , 15.69±0.2o, 16.15± 0.2o , 17.21±0.2o, 17.63±0.2o , 17. 97± 0.2o , 18.62± 0.2o , 18.93± 0.2o , 19.67± 0.2o , 19.79± 0.2o , 20.20±0.2o, 20.57± 0.2o , 21.26 ± 0.2o , 22.61±0.2o , 23.65± 0.2o , 24. 36± 0.2o , 25.64±0.2 o , 27.56±0.2 o , 28.45±0.2 o , 32.64±0.2 o , 36.86±0.2 o , 37.78±0.2 o have diffraction peaks.
在一些实施方案中,所述D晶型的X-射线粉末衍射图基本上如图5所示。In some embodiments, the X-ray powder diffraction pattern of Form D is substantially as shown in FIG. 5 .
在一些实施方案中,所述D晶型具有在231.85±5.0℃下的峰值的DSC谱图。In some embodiments, the crystalline Form D has a DSC spectrum with a peak at 231.85±5.0°C.
在一些实施方案中,所述D晶型的DSC谱图基本上如图6所示。In some embodiments, the DSC spectrum of Form D is substantially as shown in FIG. 6 .
本公开中晶型的析晶方法是常规的,例如挥发溶剂析晶、降温析晶或室温下析晶。The crystallization method of the crystal form in the present disclosure is conventional, such as crystallization from a volatile solvent, crystallization at lower temperature or crystallization at room temperature.
进一步地,本公开中晶型的制备方法中还包括过滤、洗涤或干燥等步骤。Further, the preparation method of the crystal form in the present disclosure also includes steps such as filtering, washing or drying.
另一方面,本公开内容提供药物组合物,其包含本公开所述化合物I的可药用盐,以及药学上可接受的辅料。In another aspect, the present disclosure provides a pharmaceutical composition, which comprises a pharmaceutically acceptable salt of Compound I described in the present disclosure, and pharmaceutically acceptable excipients.
另一方面,本公开内容提供治疗哺乳动物由PRMT5介导的疾病或病症的方法,包括对需要该治疗的哺乳动物,优选人类,给予治疗有效量的本公开所述化合物I的可药用盐,或其药物组合物。In another aspect, the present disclosure provides a method for treating a disease or condition mediated by PRMT5 in a mammal, comprising administering a therapeutically effective amount of a pharmaceutically acceptable salt of Compound I of the present disclosure to a mammal in need of such treatment, preferably a human. , or a pharmaceutical composition thereof.
另一方面,本公开内容提供本公开所述化合物I的可药用盐、或其药物组合物在制备预防或者治疗由PRMT5介导的疾病或病症的药物中的用途。In another aspect, the present disclosure provides the use of a pharmaceutically acceptable salt of Compound I described in the present disclosure, or a pharmaceutical composition thereof, in the preparation of a medicament for preventing or treating a disease or disorder mediated by PRMT5.
另一方面,本公开内容提供本公开所述化合物I的可药用盐、或其药物组合物在预防或者治疗由PRMT5介导的疾病或病症中的用途。In another aspect, the present disclosure provides the use of a pharmaceutically acceptable salt of Compound I described in the present disclosure, or a pharmaceutical composition thereof, in preventing or treating a disease or condition mediated by PRMT5.
另一方面,本公开内容提供预防或者治疗由PRMT5介导的疾病或病症的本公开所述化合物I的可药用盐,或其药物组合物。In another aspect, the present disclosure provides a pharmaceutically acceptable salt of compound I of the present disclosure, or a pharmaceutical composition thereof, for preventing or treating a disease or condition mediated by PRMT5.
另一方面,本公开内容提供预防或者治疗癌症的本公开所述化合物I的可药用盐,或其药物组合物。In another aspect, the present disclosure provides a pharmaceutically acceptable salt of compound I of the present disclosure, or a pharmaceutical composition thereof, for preventing or treating cancer.
在一些实施方案中,所述由PRMT5介导的疾病或病症包括但不限于癌症。In some embodiments, the disease or condition mediated by PRMT5 includes, but is not limited to, cancer.
术语定义和说明Definitions and Explanations of Terms
除非另有说明,本公开中所用的术语具有下列含义,本公开中记载的基团和术语定义,包括其作为实例的定义、示例性的定义、优选的定义、表格中记载的定义、实施例中具体化合物的定义等,可以彼此之间任意组合和结合。一个特定的术语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照本领域普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。Unless otherwise stated, terms used in this disclosure have the following meanings, definitions of groups and terms recorded in this disclosure, including their definitions as examples, exemplary definitions, preferred definitions, definitions recorded in tables, examples The definitions of specific compounds in and etc. may be combined and combined with each other arbitrarily. A specific term should not be regarded as indeterminate or unclear if there is no special definition, but should be understood according to the ordinary meaning in the art. When a trade name appears herein, it is intended to refer to its corresponding trade name or its active ingredient.
术语“约”在本公开中用于指近似地、在……左右、大略地或大约。当术语“约”与数字范围结合使用时,通过扩大所陈述的数字范围的上下限值来对该范围来进行修饰。除非另作说明,否则术语“约”在本文中用于以10%偏差的数字值修饰所陈述的值的上限和下限。The term "about" is used in this disclosure to mean approximately, around, approximately, or approximately. When the term "about" is used in conjunction with a numerical range, it modifies that range by extending the limits of the stated numerical range. Unless otherwise indicated, the term "about" is used herein to modify the upper and lower limits of the stated value by a numerical value of 10% deviation.
除非另作说明,否则术语“包括(comprise)”、“含有(comprise)”或“包含(comprise)”及其英文变体例如comprises或comprising应理解为开放的、非排他性的意义,即“包括但不限于”。Unless otherwise stated, the terms "comprise", "comprise" or "comprise" and their English variants such as comprises or comprising are to be understood in an open, non-exclusive sense, ie "including but not limited to".
在整个本文中提到的“一些实施方案”或“实施方案”是指在至少一个实施方案中包括与该实施方案所述的相关的具体参考要素、结构或特征。因此,在整个本文中不同位置出现的短语“一些实施方案”或“实施方案”不必全部指同一实施方案。此外,具体要素、结构或特征可以任何适当的方式在一个或多个实施方案中结合。Reference throughout this text to "some embodiments" or "an embodiment" means that at least one embodiment includes a specific reference element, structure or feature described in relation to that embodiment. Thus, appearances of the phrase "some embodiments" or "an embodiment" in various places throughout this document are not necessarily all referring to the same embodiment. Furthermore, particular elements, structures or characteristics may be combined in any suitable manner in one or more embodiments.
术语“任选”或“任选地”是指随后描述的事件或情况可以发生或不发生,该描述包括发生所述事件或情况和不发生所述事件或情况。The term "optional" or "optionally" means that the subsequently described event or circumstance can or cannot occur, and that description includes that said event or circumstance occurs and that it does not.
本公开所述室温是指20±5℃。The room temperature mentioned in the present disclosure refers to 20±5°C.
本公开中所述的范围“m~n”表示m到n之间的任意实数组合的缩略表示,其中m和n都是实数。例如,数值范围“5~30”表示本文中已经列出了其中的5、6、7、8、9、10、11、12、13、14、15、16、17、18、19、20、21、22、23、24、25、26、27、28、29或30;“10~15”表示本文中已经列出了其中的10、11、12、13、14或15,“10~15”只是这些数值组合的缩略表示。The range "m∼n" described in the present disclosure represents an abbreviated representation of any combination of real numbers between m and n, where both m and n are real numbers. For example, the numerical range "5-30" means that 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30; "10~15" means that 10, 11, 12, 13, 14 or 15 have been listed in this article, "10~15 " is just an abbreviation for these numerical combinations.
本公开中所述的“X-射线粉末衍射图谱或XRPD图谱”是指根据布拉格公式2d Sinθ=nλ(式中,λ为X射线的波长,当使用Cu-Kα射线时,
Figure PCTCN2022143995-appb-000003
衍射的级数n为任何正整数,一般取一级衍射峰,n=1),当X射线以掠角θ(入射角的余角,又称为布拉格角)入射到晶体或部分晶体样品的某一具有d点阵平面间距的原子面上时,就能满足布拉格方程,从而测得了这组X射线粉末衍射图。
"X-ray powder diffraction pattern or XRPD pattern" described in the present disclosure refers to according to Bragg's formula 2d Sinθ=nλ (wherein, λ is the wavelength of X-ray, when using Cu-Kα ray,
Figure PCTCN2022143995-appb-000003
The order n of diffraction is any positive integer, generally take the first-order diffraction peak, n=1), when the X-ray is incident on the crystal or part of the crystal sample at the grazing angle θ (the complementary angle of the incident angle, also known as the Bragg angle) On an atomic plane with a d-lattice plane spacing, the Bragg equation can be satisfied, and thus this group of X-ray powder diffraction patterns has been measured.
对于同种化合物的同种晶型,其XRPD图谱的峰位置在整体上具有相似性,相对强度误差可能较大。还应指出的是,在混合物的鉴定中,由于含量下降等因素会造成部分衍射线的缺失,此时,无需依赖高纯试样中观察到的全部衍射峰,甚至一个衍射峰也可能对给定的晶体而言是特征性的。For the same crystal form of the same compound, the peak positions of their XRPD patterns are generally similar, and the relative intensity error may be large. It should also be pointed out that in the identification of mixtures, due to the decrease of content and other factors, some diffraction lines will be missing. At this time, there is no need to rely on all the diffraction peaks observed in high-purity samples, and even one diffraction peak may also affect the given is characteristic for a given crystal.
本公开中所述的“2θ或2θ角度”是指衍射角,θ为布拉格角,单位为°或度;每个衍射峰2θ的误差范围为±0.20°。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 diffraction peak 2θ is ±0.20°.
对于本领域技术人员而言,由于晶体缺陷、测量误差等因素,本公开化合物与酸/碱分子、溶剂合物中的溶剂分子的摩尔比往往存在一定程度的误差,一般而言,±10%均属于合理误差范围内。随其所用之处的上下文而有一定程度的误差变化,该误差变化不超过±10%,优选±5%。For those skilled in the art, due to crystal defects, measurement errors and other factors, there is often a certain degree of error in the molar ratio of the disclosed compound to the acid/base molecule and the solvent molecule in the solvate, generally speaking, ±10% are within the reasonable error range. There is a degree of error that will vary with the context in which it is used, but the error will vary by no more than ±10%, preferably ±5%.
术语“治疗上有效量”是指(i)治疗或预防特定疾病、病况或障碍,(ii)减轻、改善或消除特定疾病、病况或障碍的一种或多种症状,或(iii)预防或延迟本文中所述的特定疾病、病况或障碍的一种或多种症状发作的本公开化合物的用量。构成“治疗有效量”的本公开化合物的量取决于该化合物、疾病状态及其严重性、给药方式以及待被治疗的哺乳动物的年龄而改变,但可例行性地由本领域技术人员根据其自身的知识及本公开内容而确定。The term "therapeutically effective amount" means (i) treating or preventing a particular disease, condition or disorder, (ii) alleviating, ameliorating or eliminating one or more symptoms of a particular disease, condition or disorder, or (iii) preventing or An amount of a compound of the disclosure that delays the onset of one or more symptoms of a particular disease, condition or disorder described herein. The amount of a compound of the present disclosure that constitutes a "therapeutically effective amount" will vary depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by one skilled in the art according to its own knowledge and this disclosure.
术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。The term "pharmaceutically acceptable" refers to those compounds, materials, compositions and/or dosage forms which, within the scope of sound medical judgment, are suitable for use in contact with human and animal tissues without excessive Toxicity, irritation, allergic reaction, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
术语“药学上可接受的辅料”是指对有机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些辅料。合适的辅料是本领域技术人员熟知的,例如碳水化合物、蜡、水溶性和/或水可膨胀的聚合物、亲水性或疏水性材料、明胶、油、溶剂、水等。The term "pharmaceutically acceptable excipients" refers to those excipients that have no obvious stimulating effect on the organism and will not impair the biological activity and performance of the active compound. Suitable excipients are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
术语“药物组合物”是指一种或多种本公开的化合物或其可药用盐或其溶剂合物与药学上可接受的辅料组成的混合物。药物组合物的目的是有利于对有机体给予本公开的化合物或其可药用盐或其溶剂合物。The term "pharmaceutical composition" refers to a mixture of one or more compounds of the present disclosure or pharmaceutically acceptable salts or solvates thereof and pharmaceutically acceptable excipients. The purpose of the pharmaceutical composition is to facilitate administration of a compound of the present disclosure, or a pharmaceutically acceptable salt or solvate thereof, to an organism.
本公开的药物组合物可通过将本公开的化合物或其可药用盐或其溶剂合物与适宜的药学上可接受的辅料组合而制备,例如可配制成固态、半固态、液态或气态制剂,如片剂、丸剂、胶囊剂、粉剂、颗粒剂、膏剂、乳剂、悬浮剂、栓剂、注射剂、吸入剂、凝胶剂、微球及气溶胶等。The pharmaceutical composition of the present disclosure can be prepared by combining the compound of the present disclosure or its pharmaceutically acceptable salt or its solvate with suitable pharmaceutically acceptable auxiliary materials, for example, it can be formulated into solid, semi-solid, liquid or gaseous preparations , such as tablets, pills, capsules, powders, granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres and aerosols, etc.
给予本公开的化合物或其可药用盐或其溶剂合物,或包含所述化合物或其可药用盐或其溶剂合物的药物组合物的典型途径包括但不限于口服、直肠、局部、吸入、肠胃外、舌下、阴道内、鼻内、眼内、腹膜内、肌内、皮下、静脉内给药。Typical routes of administration of a compound of the present disclosure, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising the compound or a pharmaceutically acceptable salt or solvate thereof include, but are not limited to, oral, rectal, topical, Inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous administration.
本公开的药物组合物可以采用本领域众所周知的方法制造,如常规的混合法、溶解法、制粒法、乳化法、冷冻干燥法等。The pharmaceutical composition of the present disclosure can be produced by methods well known in the art, such as conventional mixing methods, dissolving methods, granulating methods, emulsifying methods, freeze-drying methods and the like.
在一些实施方案中,药物组合物是口服形式。对于口服给药,可以通过将活性化合物与本领域熟知的药学上可接受的辅料混合,来配制该药物组合物。这些辅料能使本公开的化合物或其可药用盐或其溶剂合物被配制成片剂、丸剂、锭剂、糖衣剂、胶囊剂、液体、凝胶剂、浆剂、悬浮剂等,用于对患者的口服给药。In some embodiments, the pharmaceutical composition is in oral form. For oral administration, the pharmaceutical compositions can be formulated by mixing the active compounds with pharmaceutically acceptable excipients well known in the art. These excipients enable the disclosed compound or its pharmaceutically acceptable salt or its solvate to be formulated into tablets, pills, lozenges, dragees, capsules, liquids, gels, slurries, suspensions, etc. for oral administration to patients.
可以通过常规的混合、填充或压片方法来制备固体口服组合物。例如,可通过下述方法获得:将所述的活性化合物与固体辅料混合,任选地碾磨所得的混合物,如果需要则加入其它合适的辅料,然后将该混合物加工成颗粒,得到了片剂或糖衣剂的核心。适合的辅料包括但不限于:粘合剂、稀释剂、崩解剂、润滑剂、助流剂、甜味剂或矫味剂等。Solid oral compositions can be prepared by conventional methods of mixing, filling or tabletting. It can be obtained, for example, by mixing the active compound with solid excipients, optionally milling the resulting mixture, adding other suitable excipients if desired, and processing the mixture into granules to obtain tablets Or the core of the sugar coating. Suitable auxiliary materials include but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, etc.
药物组合物还可适用于肠胃外给药,如合适的单位剂型的无菌溶液剂、混悬剂或冻干产品。The pharmaceutical composition may also be adapted for parenteral administration as a suitable unit dosage form of sterile solutions, suspensions or lyophilized products.
本公开的药物组合物中含有的化合物I的可药用盐的治疗有效量选自0.01mg/kg到100mg/kg,以单独或分开剂量的形式。The therapeutically effective amount of the pharmaceutically acceptable salt of Compound I contained in the pharmaceutical composition of the present disclosure is selected from 0.01 mg/kg to 100 mg/kg, in single or divided doses.
本领域技术人员认识到对于同一化合物的给定结晶形式的XRPD峰位置和/或强度的测量数据将在误差范围内变化。2θ值允许适当的误差范围。通常,误差范围是由“±”表示。例如,5.42±0.2°2θ表示在约5.22至5.62°2θ范围内。取决于样品制备技术、应用于仪器的校准技术、人类操作偏差等,本领域技术人员认识到对于XRPD衍射角(2θ)的适当的误差范围可为约±0.20°、±0.15°、±0.10°、±0.05°或更小。当用于描述XRPD图时,术语“基本上相同”或“基本上如……所示”是指包括至少50%、至少60%、至少70%、至少80%、至少90%、至少90%或至少99%的衍射角在±0.2°2θ的标准偏差范围内的衍射峰的图。Those skilled in the art recognize that measurements of XRPD peak positions and/or intensities for a given crystalline form of the same compound will vary within error limits. The 2Θ values allow for an appropriate margin of error. Usually, the margin of error is represented by "±". For example, 5.42±0.2° 2Θ means within the range of about 5.22 to 5.62° 2Θ. Depending on the sample preparation technique, the calibration technique applied to the instrument, human operator bias, etc., one skilled in the art recognizes that a suitable error range for the XRPD diffraction angle (2θ) may be about ±0.20°, ±0.15°, ±0.10° , ±0.05° or less. When used to describe an XRPD pattern, the term "substantially the same" or "substantially as shown" means comprising at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 90% Or a pattern of diffraction peaks with at least 99% of the diffraction angles within a standard deviation of ±0.2° 2Θ.
由于本领域技术人员认识到对于同一化合物的给定结晶形式的DSC谱图的测量数据将在误差容限内变化。单峰峰值(以摄氏度表示)允许适当的误差范围。通常,误差范围是由“±”表示。对于同种化合物的同种晶型,在连续的分析中,热转变温度和熔点误差典型地在约±5.0℃之内。例如,“259.27±5.0”的峰值表示在254.27至264.27范围内。取决于样 品制备技术、应用于仪器的校准技术、人类操作偏差等,本领域技术人员认识到对于单峰峰值的适当的误差范围可为±5.0、±4.0、±3.0、±2.0或更小。As one skilled in the art recognizes, measurements of a DSC spectrum for a given crystalline form of the same compound will vary within a margin of error. Single-peak peak values (expressed in degrees Celsius) allow for a modest margin of error. Usually, the margin of error is represented by "±". For the same crystalline form of the same compound, thermal transition temperatures and melting points are typically within about ±5.0°C in consecutive analyses. For example, a peak value of "259.27±5.0" indicates that it is in the range of 254.27 to 264.27. Depending on the sample preparation technique, the calibration technique applied to the instrument, human operator bias, etc., those skilled in the art will recognize that appropriate error ranges for unimodal peaks may be ±5.0, ±4.0, ±3.0, ±2.0 or less.
本公开的盐型和/或晶型也可以是同位素标记的。本公开还包括与本文中记载的那些相同的,但一个或多个原子被原子量或质量数不同于自然中通常发现的原子量或质量数的原子置换的同位素标记的公开化合物。可结合到本公开化合物的同位素的实例包括氢、碳、氮、氧、磷、硫、氟、碘和氯的同位素,诸如分别为 2H、 3H、 11C、 13C、 14C、 13N、 15N、 15O、 17O、 18O、 31P、 32P、 35S、 18F、 123I、 125I和 36Cl等。 Salt forms and/or crystalline forms of the present disclosure may also be isotopically labeled. The disclosure also includes isotopically labeled disclosed compounds that are identical to those described herein, but with one or more atoms replaced by an atom of an atomic mass or mass number different from that normally found in nature. Examples of isotopes that can be incorporated into compounds of the present disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 123 I, 125 I and 36 Cl, etc.
某些同位素标记的本公开化合物(例如用 3H及 14C标记的那些)可用于化合物和/或底物组织分布分析中。氚化(即 3H)和碳-14(即 14C)同位素对于由于它们易于制备和可检测性是尤其优选的。正电子发射同位素,诸如 15O、 13N、 11C和 18F可用于正电子发射断层扫描(PET)研究以测定底物占有率。通常可以通过与公开于下文的方案和/或实施例中的那些类似的下列程序,通过同位素标记试剂取代未经同位素标记的试剂来制备同位素标记的本公开化合物。 Certain isotopically-labeled compounds of the disclosure (eg, those labeled with3H and14C ) are useful in compound and/or substrate tissue distribution assays. Tritiated ( ie3H ) and carbon-14 ( ie14C ) isotopes are especially preferred for their ease of preparation and detectability. Positron-emitting isotopes, such as 15 O, 13 N, 11 C, and 18 F, can be used in positron emission tomography (PET) studies to determine substrate occupancy. Isotopically labeled compounds of the disclosure can generally be prepared by following procedures similar to those disclosed in the Schemes and/or Examples below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
此外,用较重同位素(诸如氘(即 2H))取代可以提供某些由更高的代谢稳定性产生的治疗优点(例如增加的体内半衰期或降低的剂量需求),并且因此在某些情形下可能是优选的,其中氘取代可以是部分或完全的,部分氘取代是指至少一个氢被氘取代。 Furthermore, substitution with heavier isotopes such as deuterium (i.e. 2 H) may confer certain therapeutic advantages resulting from greater metabolic stability (e.g. increased in vivo half-life or reduced dosage requirements), and thus in some cases The following may be preferred, where deuterium substitution may be partial or complete, partial deuterium substitution means that at least one hydrogen is replaced by deuterium.
本公开的化合物或其可药用盐或其溶剂合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本公开的实施例。The compounds of the present disclosure or their pharmaceutically acceptable salts or their solvates can be prepared by various synthetic methods well known to those skilled in the art, including the specific embodiments listed below, and the compounds formed by combining them with other chemical synthesis methods Embodiments and equivalent replacements known to those skilled in the art, preferred embodiments include but are not limited to the examples of the present disclosure.
本公开具体实施方式的化学反应是在合适的溶剂中完成的,所述的溶剂须适合于本公开的化学变化及其所需的试剂和物料。为了获得本公开的化合物或其可药用盐或其溶剂合物,有时需要本领域技术人员在已有实施方式的基础上对合成步骤或者反应流程进行修改或选择。The chemical reactions of the embodiments of the present disclosure are performed in suitable solvents that are suitable for the chemical changes of the present disclosure and the reagents and materials required therefor. In order to obtain the disclosed compound or its pharmaceutically acceptable salt or its solvate, it is sometimes necessary for those skilled in the art to modify or select the synthetic steps or reaction schemes based on the existing embodiments.
本公开实验所用仪器的测试条件:The test condition of the instrument used in this disclosure experiment:
1、单晶X-射线结晶学测量1. Single crystal X-ray crystallography measurement
(1)设备名称:单晶衍射仪(1) Equipment name: single crystal diffractometer
检测器型号:Bruker D8 AdvanceDetector model: Bruker D8 Advance
光源:Ga KαLight source: Ga Kα
波长:1.34139Wavelength: 1.34139
(2)设备名称:偏振光显微镜(2) Equipment name: polarized light microscope
仪器型号:奥林巴斯BX53Instrument model: Olympus BX53
光源:LEDLight source: LED
目镜倍数:10倍Eyepiece multiple: 10 times
2、X-射线粉末衍射(X-ray Powder Diffraction,XRPD)2. X-ray Powder Diffraction (XRPD)
仪器型号:Bruker D8FocusInstrument model: Bruker D8Focus
射线:Cu Kα,
Figure PCTCN2022143995-appb-000004
1.54060
Rays: Cu Kα,
Figure PCTCN2022143995-appb-000004
1.54060
狭缝( o):2.5 Slit ( o ): 2.5
扫描方式:θ/2θ,扫描范围:3-40 o Scanning mode: θ/2θ, scanning range: 3-40 o
停留时间(秒):0.12Dwell time (seconds): 0.12
扫描步长( o2θ):0.01 Scan step size ( o 2θ): 0.01
扫描流速:5 o/min Scanning flow rate: 5 o /min
电压:40kVVoltage: 40kV
电流:40mACurrent: 40mA
3、差示扫描量热仪(Differential Scanning Calorimeter,DSC)3. Differential Scanning Calorimeter (DSC)
仪器型号:Discovery DSC2500Instrument model: Discovery DSC2500
吹扫气:氮气Purge gas: Nitrogen
样品盘:铝盘,非密封压盖Sample pan: aluminum pan, non-sealed gland
方法:线性升温Method: linear heating
升温速率:10℃/minHeating rate: 10°C/min
温度范围:50℃~300℃Temperature range: 50℃~300℃
4、热重分析仪(Thermogravimetric Analysis,TGA)4. Thermogravimetric Analysis (TGA)
仪器型号:Discovery TA 55Instrument model: Discovery TA 55
吹扫气:氮气Purge gas: Nitrogen
样品盘:铂金,敞口Sample tray: Platinum, open
方法:线性升温Method: linear heating
升温速率:从起始温度35℃开始,以每分钟20℃的速率升温至100℃,维持5分钟,再以每分钟20℃的速率升温至200℃,维持1分钟,再以每分钟10℃的速率升温至300℃Heating rate: starting from the initial temperature of 35°C, raise the temperature to 100°C at a rate of 20°C per minute, maintain for 5 minutes, then increase the temperature to 200°C at a rate of 20°C per minute, maintain for 1 minute, and then increase the temperature at a rate of 10°C per minute The rate of heating up to 300°C
温度范围:35℃~300℃Temperature range: 35℃~300℃
5、动态水分吸附仪(Dynamic Vapour Sorption,DVS)5. Dynamic Vapor Sorption (DVS)
检测采用DVS Intrinsic。The detection adopts DVS Intrinsic.
本公开所述的化合物I的可药用盐,包括其结晶形式在内,在理化性质、制剂加工性能及生物利用度等方面具有优势,适合制备成为所期望的药物组合物。The pharmaceutically acceptable salts of Compound I described in the present disclosure, including its crystalline forms, have advantages in terms of physicochemical properties, preparation processability and bioavailability, and are suitable for preparing desired pharmaceutical compositions.
本公开的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本公开的实施例。The compounds of the present disclosure can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by combining them with other chemical synthesis methods, and the methods well known to those skilled in the art As an equivalent alternative, preferred embodiments include but are not limited to the examples of the present disclosure.
本公开具体实施方式的化学反应是在合适的溶剂中完成的,所述的溶剂须适合于本公开的化学变化及其所需的试剂和物料。为了获得本公开的化合物,有时需要本领域技术人员在已有实施方式的基础上对合成步骤或者反应流程进行修改或选择。The chemical reactions of the embodiments of the present disclosure are performed in suitable solvents that are suitable for the chemical changes of the present disclosure and the reagents and materials required therefor. In order to obtain the compounds of the present disclosure, it is sometimes necessary for those skilled in the art to modify or select synthetic steps or reaction schemes based on the existing embodiments.
附图说明Description of drawings
图1为化合物1-8单晶的球棍图。Figure 1 is a ball-and-stick diagram of compound 1-8 single crystal.
图2为化合物I氢溴酸盐C晶型的XRPD图谱。Fig. 2 is the XRPD spectrum of compound I hydrobromide salt C crystal form.
图3为化合物I氢溴酸盐C晶型的DSC图谱。Figure 3 is the DSC spectrum of compound I hydrobromide salt C crystal form.
图4为化合物I氢溴酸盐C晶型的TGA图谱。Fig. 4 is the TGA spectrum of compound I hydrobromide salt C crystal form.
图5为化合物I 1,5-萘二磺酸盐D晶型的XRPD图谱。Figure 5 is the XRPD pattern of Compound I 1,5-naphthalene disulfonate D crystal form.
图6为化合物I 1,5-萘二磺酸盐D晶型的DSC图谱。Figure 6 is the DSC spectrum of compound I 1,5-naphthalene disulfonate D crystal form.
图7为化合物I 1,5-萘二磺酸盐D晶型的TGA图谱。Figure 7 is the TGA spectrum of compound I 1,5-naphthalene disulfonate D crystal form.
图8为化合物I一盐酸盐A晶型的XRPD图谱。Fig. 8 is the XRPD pattern of compound I-hydrochloride A crystal form.
图9为化合物I一盐酸盐A晶型的DSC图谱。Fig. 9 is a DSC spectrum of compound I-hydrochloride A crystal form.
图10为化合物I一盐酸盐A晶型的TGA图谱。Figure 10 is the TGA pattern of compound I-hydrochloride A crystal form.
图11为化合物I一盐酸盐B晶型的XRPD图谱。Figure 11 is the XRPD pattern of compound I-hydrochloride B crystal form.
图12为化合物I一盐酸盐B晶型的DSC图谱。Figure 12 is the DSC spectrum of compound I-hydrochloride B crystal form.
图13为化合物I一盐酸盐B晶型的TGA图谱。Figure 13 is the TGA pattern of compound I-hydrochloride B crystal form.
图14为Z-138皮下瘤模型中接受受试化合物的小鼠的肿瘤生长曲线,图中化合物002即为实施例2化合物。Figure 14 is the tumor growth curve of mice receiving the test compound in the Z-138 subcutaneous tumor model, in which compound 002 is the compound of Example 2.
图15为Z-138皮下瘤模型中接受受试化合物的小鼠的体重变化曲线,图中化合物002即为实施例2化合物。Figure 15 is the curve of body weight change of mice receiving the test compound in the Z-138 subcutaneous tumor model, in which compound 002 is the compound of Example 2.
具体实施方式Detailed ways
下面通过实施例对本公开进行详细描述,但并不意味着对本公开的任何不利限制。本文已经详细地描述了本公开内容,其中也公开了其具体实施例方式,对本领域的技术人员而言,在不脱离本公开精神和范围的情况下针对本公开具体实施方式进行各种改变和改进将是显而易见的。本公开所使用的所有试剂是市售的,无需进一步纯化即可使用。The present disclosure will be described in detail through examples below, but it does not imply any adverse limitation to the present disclosure. The present disclosure has been described in detail herein, and its specific embodiments are also disclosed. For those skilled in the art, various changes and modifications can be made to the specific embodiments of the present disclosure without departing from the spirit and scope of the present disclosure. Improvements will be apparent. All reagents used in this disclosure are commercially available and used without further purification.
除非另作说明,混合溶剂表示的比例是体积混合比例。除非另作说明,否则,%是指wt%。Unless otherwise specified, the ratios indicated for mixed solvents are volume mixing ratios. Unless otherwise stated, % means wt%.
化合物经手工命名,市售化合物采用供应商目录名称。Compounds were named manually, and commercially available compounds used supplier catalog names.
化合物的结构是通过核磁共振(NMR)和/或质谱(MS)来确定的。NMR位移的单位为10 -6(ppm)。NMR测定的溶剂为氘代二甲基亚砜、氘代氯仿、氘代甲醇等,内标为四甲基硅烷(TMS);“IC 50”指半数抑制浓度,指达到最大抑制效果一半时的浓度。 Compound structures were determined by nuclear magnetic resonance (NMR) and/or mass spectroscopy (MS). The unit of NMR shift is 10 -6 (ppm). The solvents measured by NMR are deuterated dimethyl sulfoxide, deuterated chloroform, deuterated methanol, etc., and the internal standard is tetramethylsilane (TMS); concentration.
下文的洗脱剂可由两种或多种溶剂形成混合洗脱剂,其比值为各溶剂的体积比。例如,“0~10%甲醇/二氯甲烷”表示梯度洗脱过程中,混合洗脱剂中的甲醇与二氯甲烷的体积用量比为0:100~10:100。The following eluents can be mixed eluents formed by two or more solvents, and the ratio is the volume ratio of each solvent. For example, "0-10% methanol/dichloromethane" means that during the gradient elution process, the volume ratio of methanol and dichloromethane in the mixed eluent is 0:100-10:100.
实施例1:(S)-3-((S)-环氧乙烷-2-基)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(中间体1)的制备Example 1: (S)-3-((S)-oxiran-2-yl)-3,4-dihydroisoquinoline-2(1H)-tert-butyl carboxylate (Intermediate 1) preparation of
Figure PCTCN2022143995-appb-000005
Figure PCTCN2022143995-appb-000005
步骤1:化合物b的制备Step 1: Preparation of compound b
Figure PCTCN2022143995-appb-000006
Figure PCTCN2022143995-appb-000006
室温下将(S)-2-(叔丁氧羰基)-1,2,3,4-四氢异喹啉-3-羧酸(a)(55g,200mmol),二甲羟胺盐酸盐(29.4g,300mmol),2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(91g,240mmol)加入到1L单口瓶中,再加入无水N,N-二甲基甲酰胺(500mL),氮气保护后冰浴冷却,然后滴加N,N-二异丙基乙胺(104mL,600mmol)。反应液在室温反应4小时。反应完全后旋蒸除去过量的N,N-二异丙基乙胺和N,N-二甲基甲酰胺,然后冰浴冷却,用饱和食盐水(1L)稀释,用乙酸乙酯萃取(200mL X 2),有机相合并后用5%碳酸钠水溶液洗涤(500mL X 2),然后用饱和食盐水洗涤(500mL)。无水硫酸钠干燥,过滤,滤液减压浓缩后硅胶柱层析(洗脱剂梯度:石油醚/乙酸乙酯=2/1)纯化,得到目标中间体(S)-3-(甲氧基(甲基)氨基甲酰)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(b)(62g,收率:97%)。(S)-2-(tert-butoxycarbonyl)-1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (a) (55g, 200mmol), dimethylhydroxylamine hydrochloride ( 29.4g, 300mmol), 2-(7-azabenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (91g, 240mmol) was added in a 1L single-necked bottle, Anhydrous N,N-dimethylformamide (500 mL) was added again, cooled in an ice bath after nitrogen protection, and then N,N-diisopropylethylamine (104 mL, 600 mmol) was added dropwise. The reaction solution was reacted at room temperature for 4 hours. After the reaction was complete, the excess N,N-diisopropylethylamine and N,N-dimethylformamide were removed by rotary evaporation, then cooled in an ice bath, diluted with saturated brine (1L), extracted with ethyl acetate (200mL X 2), the combined organic phases were washed with 5% aqueous sodium carbonate solution (500mL X 2), and then washed with saturated brine (500mL). Dry over anhydrous sodium sulfate, filter, concentrate the filtrate under reduced pressure and purify by silica gel column chromatography (eluent gradient: petroleum ether/ethyl acetate=2/1) to obtain the target intermediate (S)-3-(methoxy (Methyl)carbamoyl)-3,4-dihydroisoquinoline-2(1H)-carboxylic acid tert-butyl ester (b) (62 g, yield: 97%).
LCMS:Rt:1.76min;MS m/z(ESI):321.3[M+H] +LCMS: Rt: 1.76 min; MS m/z (ESI): 321.3 [M+H] + .
手性HPLC:Rt:3.159min。Chiral HPLC: Rt: 3.159 min.
步骤2:化合物c的制备Step 2: Preparation of compound c
Figure PCTCN2022143995-appb-000007
Figure PCTCN2022143995-appb-000007
室温下将(S)-3-(甲氧基(甲基)氨基甲酰)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(b)(20g,62.5mmol)称量到500mL三口瓶中,加入无水四氢呋喃(200mL),冷却至-70℃,缓慢滴加二异丁基氢化铝(DIBAL-H)甲苯溶液(1.5mol/L,83mL,125mmol),反应液在-70℃搅拌1小时。反应完全后在-70℃缓慢加入饱和氯化铵溶液(100mL)淬灭,然后加入0.5mol/L盐酸水溶液稀释(200mL)。分层后有机相用饱和氯化钠水溶液洗涤(200mL X 2),然后用无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用硅胶柱层析(洗脱剂梯度:石油醚/乙酸乙酯=8/1)纯化,得中间体(S)-3-甲酰基-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(c)(15g,收率: 92%)。(S)-3-(methoxy(methyl)carbamoyl)-3,4-dihydroisoquinoline-2(1H)-carboxylic acid tert-butyl ester (b) (20g, 62.5mmol ) was weighed into a 500mL three-necked flask, anhydrous tetrahydrofuran (200mL) was added, cooled to -70°C, diisobutylaluminum hydride (DIBAL-H) toluene solution (1.5mol/L, 83mL, 125mmol) was slowly added dropwise, The reaction was stirred at -70°C for 1 hour. After the reaction was complete, slowly add saturated ammonium chloride solution (100 mL) at -70°C to quench, and then add 0.5 mol/L hydrochloric acid aqueous solution to dilute (200 mL). After layering, the organic phase was washed with saturated aqueous sodium chloride solution (200mL × 2), then dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the residue was subjected to silica gel column chromatography (eluent gradient: sherwood oil/acetic acid Ethyl ester=8/1) purification to obtain intermediate (S)-3-formyl-3,4-dihydroisoquinoline-2(1H)-tert-butyl carboxylate (c) (15g, yield: 92%).
LCMS:Rt:1.93min;MS m/z(ESI):206.1[M-56+H] +LCMS: Rt: 1.93 min; MS m/z (ESI): 206.1 [M-56+H] + .
手性HPLC:Rt:2.018min。Chiral HPLC: Rt: 2.018min.
步骤3:化合物d的制备Step 3: Preparation of compound d
Figure PCTCN2022143995-appb-000008
Figure PCTCN2022143995-appb-000008
将甲基三苯基溴化膦(238g,0.67mol)分散到无水四氢呋喃(1.5L)中,氮气保护下冷却至-70℃,缓慢滴加二(三甲基硅基)氨基钠(334mL,0.67mol),控制温度低于-50℃,滴加完毕后缓慢升至室温,搅拌2小时。重新冷却至-70℃,缓慢滴加(S)-3-甲酰基-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(c)(87g,0.33moL)的四氢呋喃溶液(200mL),控制温度低于-50℃,滴加完毕后缓慢升至室温过夜。TLC检测反应完毕后,冷却至0℃,加入饱和氯化铵溶液淬灭,缓慢加入1mol/L盐酸水溶液调节pH到3-4,加入乙酸乙酯(500mL)萃取。有机相用饱和食盐水洗涤(200mL X 2),无水硫酸钠干燥后过滤,滤液减压浓缩。残余物加入混合溶剂(乙酸乙酯/石油醚=1/4)重结晶,过滤除去析出的三苯基氧化膦,滤液浓缩后硅胶柱层析(乙酸乙酯/石油醚=1/8)得目标产物(S)-3-乙烯基-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(d)(85g,收率:98%)。Disperse methyltriphenylphosphine bromide (238g, 0.67mol) in anhydrous tetrahydrofuran (1.5L), cool to -70°C under nitrogen protection, slowly add sodium bis(trimethylsilyl)amide (334mL , 0.67mol), the temperature was controlled below -50°C, and after the dropwise addition was completed, it was slowly raised to room temperature and stirred for 2 hours. Re-cool to -70°C, slowly add (S)-3-formyl-3,4-dihydroisoquinoline-2(1H)-tert-butyl carboxylate (c) (87g, 0.33moL) in THF solution (200 mL), the temperature was controlled below -50°C, and after the dropwise addition was completed, it was slowly raised to room temperature overnight. After the reaction was detected by TLC, it was cooled to 0° C., quenched by adding saturated ammonium chloride solution, slowly added 1 mol/L hydrochloric acid aqueous solution to adjust the pH to 3-4, and extracted by adding ethyl acetate (500 mL). The organic phase was washed with saturated brine (200mL X 2), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure. The residue was recrystallized by adding a mixed solvent (ethyl acetate/petroleum ether=1/4), and the precipitated triphenylphosphine oxide was removed by filtration, and the filtrate was concentrated and then chromatographed on a silica gel column (ethyl acetate/petroleum ether=1/8) to obtain Target product (S)-tert-butyl 3-vinyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (d) (85 g, yield: 98%).
手性HPLC:Rt:1.883min。Chiral HPLC: Rt: 1.883 min.
步骤4:化合物e的制备Step 4: Preparation of compound e
Figure PCTCN2022143995-appb-000009
Figure PCTCN2022143995-appb-000009
将(S)-3-乙烯基-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(d)(25.9g,0.1mol)溶于乙酸乙酯/乙腈(500mL/500mL)溶液中,冷却至0℃,在10分钟内加入高碘酸钠(32.1g,0.15mol)与三氯化钌水合物(1.6g,7.7mmol)的水溶液,反应液在0℃搅拌10分钟。TLC检测反应完毕,加入硫代硫酸钠饱和溶液(150mL)淬灭,搅拌30分钟后分层,有机相用饱和食盐水洗涤(200mL X 2),无水硫酸钠干燥,过滤后滤液减压浓缩,残余物经硅胶柱层析纯化(洗脱剂:乙酸乙酯/石油醚=1/2-1/1),得产物(S)-3-((S)-1,2-二羟基乙基)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(e)(极性较小的产物,14g,收率48%)。加大洗脱剂极性(乙酸乙酯/石油醚=2/1-1/0),得产物(S)-3-((R)-1,2-二羟基乙基)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(e-1)(极性较大的产物,8g,收率28%)。Dissolve (S)-tert-butyl 3-vinyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (d) (25.9 g, 0.1 mol) in ethyl acetate/acetonitrile (500 mL/ 500mL) solution, cooled to 0°C, added sodium periodate (32.1g, 0.15mol) and ruthenium trichloride hydrate (1.6g, 7.7mmol) aqueous solution within 10 minutes, the reaction solution was stirred at 0°C for 10 minute. TLC detects that the reaction is complete, adding saturated sodium thiosulfate solution (150mL) to quench, stirring for 30 minutes and then layering, the organic phase is washed with saturated brine (200mL × 2), dried over anhydrous sodium sulfate, and the filtrate is concentrated under reduced pressure after filtration , the residue was purified by silica gel column chromatography (eluent: ethyl acetate/petroleum ether=1/2-1/1) to obtain the product (S)-3-((S)-1,2-dihydroxyethyl tert-butyl)-3,4-dihydroisoquinoline-2(1H)-carboxylate (e) (less polar product, 14 g, yield 48%). Increase the polarity of the eluent (ethyl acetate/petroleum ether=2/1-1/0) to obtain the product (S)-3-((R)-1,2-dihydroxyethyl)-3,4 -dihydroisoquinoline-2(1H)-tert-butyl carboxylate (e-1) (more polar product, 8 g, yield 28%).
步骤5:化合物f的制备Step 5: Preparation of compound f
Figure PCTCN2022143995-appb-000010
Figure PCTCN2022143995-appb-000010
将(S)-3-((S)-1,2-二羟基乙基)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(e)(14g,0.047mol)溶于二氯甲烷(150mL)中,加入三乙胺(9.90mL,0.072mol)后在搅拌状态下分批次加入对甲苯磺酰氯(10.0g,0.052mol),反应液在40℃搅拌过夜。将反应液冷却至室温,用饱 和食盐水洗涤(100mL X 2),有机相用无水硫酸钠干燥。过滤后滤液减压浓缩,残余物经硅胶柱层析(乙酸乙酯/石油醚=1/4)得目标产物(S)-3-((S)-1-羟基-2-(甲苯磺酰氧基)乙基)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(f)(12.6g,收率59%)。(S)-3-((S)-1,2-dihydroxyethyl)-3,4-dihydroisoquinoline-2(1H)-tert-butyl carboxylate (e) (14g, 0.047mol ) was dissolved in dichloromethane (150mL), triethylamine (9.90mL, 0.072mol) was added, and p-toluenesulfonyl chloride (10.0g, 0.052mol) was added in batches under stirring, and the reaction solution was stirred overnight at 40°C . The reaction solution was cooled to room temperature, washed with saturated brine (100mL × 2), and the organic phase was dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure, and the residue was subjected to silica gel column chromatography (ethyl acetate/petroleum ether=1/4) to obtain the target product (S)-3-((S)-1-hydroxy-2-(toluenesulfonyl Oxy)ethyl)-3,4-dihydroisoquinoline-2(1H)-carboxylic acid tert-butyl ester (f) (12.6 g, yield 59%).
步骤6:化合物中间体1的制备Step 6: Preparation of Compound Intermediate 1
Figure PCTCN2022143995-appb-000011
Figure PCTCN2022143995-appb-000011
将(S)-3-((S)-1-羟基-2-(甲苯磺酰氧基)乙基)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(f)(12.6g,28.2mmol)溶于N,N-二甲基甲酰胺(150mL)中,氮气保护下分批次加入氢化钠(1.70g,42.3mmol),反应液在40℃搅拌1小时,TLC检测反应完全。冷却至0℃,滴加饱和食盐水淬灭,反应液直接反相硅胶色谱柱(水/乙腈=50/10)纯化,柱层析所得溶液用乙酸乙酯萃取(200mL X 3),有机相经无水硫酸钠干燥后过滤,滤液减压浓缩得到中间体(S)-3-((S)-环氧乙烷-2-基)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(中间体1)(6.5g,收率72%)。(S)-3-((S)-1-hydroxyl-2-(tosyloxy)ethyl)-3,4-dihydroisoquinoline-2(1H)-carboxylic acid tert-butyl ester ( f) Dissolve (12.6g, 28.2mmol) in N,N-dimethylformamide (150mL), add sodium hydride (1.70g, 42.3mmol) in batches under nitrogen protection, and stir the reaction solution at 40°C for 1 hour , TLC detected that the reaction was complete. Cooled to 0°C, quenched by adding saturated brine dropwise, the reaction solution was directly purified by reverse-phase silica gel column (water/acetonitrile=50/10), the solution obtained by column chromatography was extracted with ethyl acetate (200mL X 3), and the organic phase was After drying over anhydrous sodium sulfate and filtering, the filtrate was concentrated under reduced pressure to obtain the intermediate (S)-3-((S)-oxirane-2-yl)-3,4-dihydroisoquinoline-2(1H )-tert-butylcarboxylate (Intermediate 1) (6.5 g, yield 72%).
实施例2:1-乙基-4-((R)-2-羟基-2-((S)-1,2,3,4-四氢异喹啉-3-基)乙基)-8-(2-甲氧基-7-氮杂螺[3.5]壬烷-7-羰基)-1,2,3,4-四氢-5H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000012
-5-酮(化合物I)盐酸盐的制备
Example 2: 1-ethyl-4-((R)-2-hydroxy-2-((S)-1,2,3,4-tetrahydroisoquinolin-3-yl)ethyl)-8 -(2-Methoxy-7-azaspiro[3.5]nonane-7-carbonyl)-1,2,3,4-tetrahydro-5H-benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000012
Preparation of -5-ketone (compound I) hydrochloride
Figure PCTCN2022143995-appb-000013
Figure PCTCN2022143995-appb-000013
步骤1:化合物1-2的制备Step 1: Preparation of Compound 1-2
Figure PCTCN2022143995-appb-000014
Figure PCTCN2022143995-appb-000014
室温下将4-溴-2-氟苯甲酸(1-1)(7.5g,34.2mmol)和N-叔丁氧羰基-1,2-乙二胺(16.4g,102.7mmol)加入到N-甲基吡咯烷酮(30mL)中。加入完毕后,升温至120℃反应16小时。反应完全后,将反应液缓慢加入到水(150mL)中,用2mol/L稀盐酸调节溶液pH至5~6,然后用乙酸乙酯萃取(150mL X 2)。合并有机相,用饱和食盐水(150mL X 2)洗涤,无水硫酸钠干燥,过滤。将滤液减压浓缩得到目标中间体4-溴-2-((2-((叔丁氧羰基)氨基)乙基)氨基)苯甲酸(1-2)(粗品,11.0g,收率:89%)。4-Bromo-2-fluorobenzoic acid (1-1) (7.5g, 34.2mmol) and N-tert-butoxycarbonyl-1,2-ethylenediamine (16.4g, 102.7mmol) were added to N- Methylpyrrolidone (30mL). After the addition was complete, the temperature was raised to 120° C. for 16 hours of reaction. After the reaction was complete, the reaction solution was slowly added to water (150mL), the pH of the solution was adjusted to 5-6 with 2mol/L dilute hydrochloric acid, and then extracted with ethyl acetate (150mL×2). Combine the organic phases, wash with saturated brine (150mL×2), dry over anhydrous sodium sulfate, and filter. The filtrate was concentrated under reduced pressure to obtain the target intermediate 4-bromo-2-((2-((tert-butoxycarbonyl)amino)ethyl)amino)benzoic acid (1-2) (crude product, 11.0g, yield: 89 %).
LCMS:Rt:1.836min;MS m/z(ESI):359.0[M+H] +LCMS: Rt: 1.836 min; MS m/z (ESI): 359.0 [M+H] + .
步骤2:化合物1-3的制备Step 2: Preparation of Compound 1-3
Figure PCTCN2022143995-appb-000015
Figure PCTCN2022143995-appb-000015
室温下将4-溴-2-((2-((叔丁氧羰基)氨基)乙基)氨基)苯甲酸(1-2)(11.0g,30.6mmol),加入到4mol/L盐酸/二氧六环溶液(30mL)中,然后在室温反应1小时。反应完全后将反应液减压浓缩,得到目标中间体2-((2-氨基乙基)氨基)-4-溴苯甲酸(1-3)(粗品,8g,收率:100%)。Add 4-bromo-2-((2-((tert-butoxycarbonyl)amino)ethyl)amino)benzoic acid (1-2) (11.0g, 30.6mmol) to 4mol/L hydrochloric acid/di Hexane solution (30 mL), and react at room temperature for 1 hour. After the reaction was complete, the reaction solution was concentrated under reduced pressure to obtain the target intermediate 2-((2-aminoethyl)amino)-4-bromobenzoic acid (1-3) (crude product, 8 g, yield: 100%).
LCMS:Rt:0.688min;MS m/z(ESI):259.0[M+H] +LCMS: Rt: 0.688 min; MS m/z (ESI): 259.0 [M+H] + .
步骤3:化合物1-4的制备Step 3: Preparation of Compounds 1-4
Figure PCTCN2022143995-appb-000016
Figure PCTCN2022143995-appb-000016
室温下将2-((2-氨基乙基)氨基)-4-溴苯甲酸(1-3)(3g,11.6mmol),2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(8.8g,23.2mmol)和三乙胺(4.7g,46.4mmol)加入到超干N,N-二甲基甲酰胺(20mL)中,室温反应1.5小时。反应完全后加入饱和食盐水(100mL),用乙酸乙酯萃取(100mL X 2),有机相用饱和食盐水洗涤(50mL X 2),无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用正相硅胶色谱柱(洗脱剂梯度:二氯甲烷/甲醇=20/1)纯化,得到目标中间体8-溴-1,2,3,4-四氢-5H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000017
-5-酮(1-4)(1.6g,收率:57%)。
2-((2-Aminoethyl)amino)-4-bromobenzoic acid (1-3) (3g, 11.6mmol), 2-(7-azabenzotriazole)-N,N , N',N'-Tetramethyluronium hexafluorophosphate (8.8 g, 23.2 mmol) and triethylamine (4.7 g, 46.4 mmol) were added to ultra-dry N,N-dimethylformamide (20 mL) , react at room temperature for 1.5 hours. After the reaction was complete, saturated brine (100 mL) was added, extracted with ethyl acetate (100 mL X 2), the organic phase was washed with saturated brine (50 mL X 2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by normal phase silica gel chromatography (eluent gradient: dichloromethane/methanol=20/1) to obtain the target intermediate 8-bromo-1,2,3,4-tetrahydro-5H-benzo[ e][1,4]diazepine
Figure PCTCN2022143995-appb-000017
-5-Kone (1-4) (1.6 g, yield: 57%).
LCMS:Rt:1.173min;MS m/z(ESI):241.0[M+H] +LCMS: Rt: 1.173 min; MS m/z (ESI): 241.0 [M+H] + .
步骤4:化合物1-5的制备Step 4: Preparation of Compounds 1-5
Figure PCTCN2022143995-appb-000018
Figure PCTCN2022143995-appb-000018
室温下将8-溴-1,2,3,4-四氢-5H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000019
-5-酮(1-4)(1.6g,6.6mmol),氰基硼氢化钠(834mg,13.3mmol)和乙醛(584mg,13.3mmol)加入到醋酸(2mL)和甲醇(20mL)混合溶液中,室温反应1小时。反应完全后加入2mol/L稀盐酸(1mL)和水(50.0mL),用乙酸乙酯(50.0mL X 2)萃取。有机相合并,用饱和食盐水(50mL X 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经正相硅胶色谱柱(洗脱剂梯度:石油醚/乙酸乙酯=1/2)纯化,得到目标中间体8-溴-1-乙基-1,2,3,4-四氢-5H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000020
-5-酮(1-5)(1.1g,收率:62%)。
8-Bromo-1,2,3,4-tetrahydro-5H-benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000019
-5-ketone (1-4) (1.6g, 6.6mmol), sodium cyanoborohydride (834mg, 13.3mmol) and acetaldehyde (584mg, 13.3mmol) were added to a mixed solution of acetic acid (2mL) and methanol (20mL) , react at room temperature for 1 hour. After the reaction was complete, 2mol/L dilute hydrochloric acid (1 mL) and water (50.0 mL) were added, and extracted with ethyl acetate (50.0 mL X 2). The organic phases were combined, washed with saturated brine (50 mL X 2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by normal phase silica gel chromatography (eluent gradient: petroleum ether/ethyl acetate=1/2) to obtain the target intermediate 8-bromo-1-ethyl-1,2,3,4-tetrahydro -5H-Benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000020
-5-Kone (1-5) (1.1 g, yield: 62%).
LCMS:Rt:1.499min;MS m/z(ESI):269.0[M+H] +LCMS: Rt: 1.499 min; MS m/z (ESI): 269.0 [M+H] + .
步骤5:化合物1-6的制备Step 5: Preparation of Compounds 1-6
Figure PCTCN2022143995-appb-000021
Figure PCTCN2022143995-appb-000021
室温下将8-溴-1-乙基-1,2,3,4-四氢-5H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000022
-5-酮(1-5)(400mg,1.52mmol)加入到超干N,N-二甲基甲酰胺(20mL)中,然后加入氢化钠(90.9mg,2.27mmol)。 添加完毕,加热至40℃搅拌1h后,再加入实施例1制得的中间体1(500mg,1.82mmol),反应16小时。反应完全后,加入水(100mL),用乙酸乙酯(100mL X 2)萃取。有机相合并,用饱和食盐水洗涤(50mL X 2),无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经正相硅胶色谱柱(洗脱剂梯度:二氯甲烷/甲醇=20/1)纯化,得到目标中间体(1R,10aS)-1-((8-溴-1-乙基-5-氧代-1,2,3,5-四氢-4H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000023
-4-基)甲基)-1,5,10,10a-四氢-3H-噁唑并[3,4-b]异喹啉-3-酮(1-6)(150mg,收率:21%)。
8-Bromo-1-ethyl-1,2,3,4-tetrahydro-5H-benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000022
-5-Kone(1-5) (400 mg, 1.52 mmol) was added to ultra-dry N,N-dimethylformamide (20 mL), followed by sodium hydride (90.9 mg, 2.27 mmol). After the addition was completed, the mixture was heated to 40° C. and stirred for 1 h, then the intermediate 1 (500 mg, 1.82 mmol) prepared in Example 1 was added, and reacted for 16 hours. After the reaction was complete, water (100 mL) was added and extracted with ethyl acetate (100 mL X 2). The organic phases were combined, washed with saturated brine (50 mL X 2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by normal phase silica gel chromatography (eluent gradient: dichloromethane/methanol=20/1) to obtain the target intermediate (1R,10aS)-1-((8-bromo-1-ethyl-5 -Oxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000023
-4-yl)methyl)-1,5,10,10a-tetrahydro-3H-oxazolo[3,4-b]isoquinolin-3-one (1-6) (150mg, yield: twenty one%).
LCMS:Rt:1.872min;MS m/z(ESI):470.1[M+H] +LCMS: Rt: 1.872 min; MS m/z (ESI): 470.1 [M+H] + .
步骤6:化合物1-7的制备Step 6: Preparation of Compounds 1-7
Figure PCTCN2022143995-appb-000024
Figure PCTCN2022143995-appb-000024
室温下将中间体(1R,10aS)-1-((8-溴-1-乙基-5-氧代-1,2,3,5-四氢-4H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000025
-4-基)甲基)-1,5,10,10a-四氢-3H-噁唑并[3,4-b]异喹啉-3-酮(1-6)(150mg,0.32mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯(11.7mg,0.02mmol)和醋酸钾(94mg,0.96mmol)加入到无水乙醇(10mL)中,CO置换3次,加热至70℃反应3.0小时。反应完全后冷却至室温,将反应液减压浓缩,加入饱和食盐水(50mL),然后用乙酸乙酯(50mL X 2)萃取,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经正相硅胶色谱柱(洗脱剂梯度:石油醚/乙酸乙酯=1/2)纯化,得到目标中间体1-乙基-5-氧代-4-(((1R,10aS)-3-羰基-1,5,10,10a-四氢-3H-噁唑并[3,4-b]异喹啉-1-基)甲基)-2,3,4,5-四氢-1H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000026
-8-羧酸乙酯(1-7)(130mg,收率:87%)。
The intermediate (1R,10aS)-1-((8-bromo-1-ethyl-5-oxo-1,2,3,5-tetrahydro-4H-benzo[e][1, 4] diazepine
Figure PCTCN2022143995-appb-000025
-4-yl)methyl)-1,5,10,10a-tetrahydro-3H-oxazolo[3,4-b]isoquinolin-3-one (1-6) (150mg, 0.32mmol) , [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (11.7mg, 0.02mmol) and potassium acetate (94mg, 0.96mmol) were added to absolute ethanol (10mL) and replaced by CO 3 times, heated to 70°C for 3.0 hours. After the reaction was complete, it was cooled to room temperature, the reaction solution was concentrated under reduced pressure, saturated brine (50 mL) was added, and then extracted with ethyl acetate (50 mL X 2), the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by normal phase silica gel chromatography (eluent gradient: petroleum ether/ethyl acetate=1/2) to obtain the target intermediate 1-ethyl-5-oxo-4-(((1R,10aS) -3-Carbonyl-1,5,10,10a-tetrahydro-3H-oxazolo[3,4-b]isoquinolin-1-yl)methyl)-2,3,4,5-tetrahydro -1H-Benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000026
-Ethyl 8-carboxylate (1-7) (130 mg, yield: 87%).
LCMS:Rt:1.784min;MS m/z(ESI):464.1[M+H] +LCMS: Rt: 1.784 min; MS m/z (ESI): 464.1 [M+H] + .
步骤7:化合物1-8的制备Step 7: Preparation of Compounds 1-8
Figure PCTCN2022143995-appb-000027
Figure PCTCN2022143995-appb-000027
室温下将1-乙基-5-氧代-4-(((1R,10aS)-3-羰基-1,5,10,10a-四氢-3H-噁唑并[3,4-b]异喹啉-1-基)甲基)-2,3,4,5-四氢-1H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000028
-8-羧酸乙酯(1-7)(130mg,0.28mmol)加入到甲醇(4mL)和水(4mL)的混合溶液中,然后加入氢氧化钠(90mg,2.24mmol),加热至70℃,反应16.0小时。反应完全后,将反应体系降温至室温,加入Boc酸酐(122mg,0.56mmol)反应1.5小时。反应完全后,将反应体系降温至0℃,用1mol/L的盐酸水溶液调节反应液的pH至5.0,后用乙酸乙酯(30mL X 3)萃取,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经反相硅胶色谱柱(洗脱剂梯度:乙腈/水=43%)得到目标中间体4-((R)-2-((S)-2-(叔丁氧基羰基)-1,2,3,4-四氢异喹啉-3-基)-2-羟基乙基)-1-乙基-5-氧代-2,3,4,5-四氢-1H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000029
-8-羧酸(1-8)(粗品,60mg,收率:42%)。
1-Ethyl-5-oxo-4-(((1R,10aS)-3-carbonyl-1,5,10,10a-tetrahydro-3H-oxazolo[3,4-b] Isoquinolin-1-yl)methyl)-2,3,4,5-tetrahydro-1H-benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000028
-Ethyl 8-carboxylate (1-7) (130mg, 0.28mmol) was added to a mixed solution of methanol (4mL) and water (4mL), then sodium hydroxide (90mg, 2.24mmol) was added and heated to 70°C , reacted for 16.0 hours. After the reaction was complete, the reaction system was cooled to room temperature, and Boc anhydride (122 mg, 0.56 mmol) was added to react for 1.5 hours. After the reaction was complete, the reaction system was cooled to 0°C, and the pH of the reaction solution was adjusted to 5.0 with 1mol/L hydrochloric acid aqueous solution, and then extracted with ethyl acetate (30mL X 3), the organic phase was dried with anhydrous sodium sulfate, filtered, The filtrate was concentrated under reduced pressure. The residue was subjected to reverse-phase silica gel chromatography (eluent gradient: acetonitrile/water=43%) to obtain the target intermediate 4-((R)-2-((S)-2-(tert-butoxycarbonyl)-1 ,2,3,4-tetrahydroisoquinolin-3-yl)-2-hydroxyethyl)-1-ethyl-5-oxo-2,3,4,5-tetrahydro-1H-benzo [e][1,4]diazepine
Figure PCTCN2022143995-appb-000029
-8-Carboxylic acid (1-8) (crude product, 60 mg, yield: 42%).
LCMS:Rt:1.722min;MS m/z(ESI):510.3[M+H] +LCMS: Rt: 1.722 min; MS m/z (ESI): 510.3 [M+H] + .
化合物1-8的单晶X-射线结构测定和单晶X-射线分析:Single crystal X-ray structure determination and single crystal X-ray analysis of compounds 1-8:
单晶制备方法:称取化合物1-8(10.0mg)放入3mL螺口玻璃瓶中,加入甲醇2mL, 搅拌5分钟后固体溶清。向玻璃瓶中加入0.5mL水,继续搅拌5分钟。溶液经0.22μm微孔滤膜滤至3mL螺口玻璃瓶中,玻璃瓶口用保鲜膜覆盖。用针头在瓶口处扎8个小孔,室温放置7天,制得上述化合物单晶。Single crystal preparation method: Weigh compound 1-8 (10.0 mg) into a 3 mL screw-top glass bottle, add 2 mL of methanol, stir for 5 minutes, and then the solid dissolves. Add 0.5 mL of water to the glass bottle and continue stirring for 5 minutes. The solution was filtered through a 0.22 μm microporous membrane into a 3 mL screw-top glass bottle, and the mouth of the glass bottle was covered with plastic wrap. Prick 8 small holes at the mouth of the bottle with a needle and leave it at room temperature for 7 days to obtain a single crystal of the above compound.
所得单晶样品进行X-射线分析,测试结果见表1和图1。The obtained single crystal sample was subjected to X-ray analysis, and the test results are shown in Table 1 and Fig. 1 .
表1化合物1-8的单晶样品和晶体数据Table 1 Single crystal samples and crystal data of compounds 1-8
Figure PCTCN2022143995-appb-000030
Figure PCTCN2022143995-appb-000030
通过上述X-射线晶体衍射实验,可以确定化合物1-8的化学结构和绝对构型如下:Through the above-mentioned X-ray crystal diffraction experiment, the chemical structure and absolute configuration of compound 1-8 can be determined as follows:
Figure PCTCN2022143995-appb-000031
Figure PCTCN2022143995-appb-000031
步骤8:化合物2-2的制备Step 8: Preparation of Compound 2-2
Figure PCTCN2022143995-appb-000032
Figure PCTCN2022143995-appb-000032
室温下将4-((R)-2-((S)-2-(叔丁氧基羰基)-1,2,3,4-四氢异喹啉-3-基)-2-羟基乙基)-1-乙基-5- 氧代-2,3,4,5-四氢-1H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000033
-8-羧酸(1-8)(100mg,0.196mmol),2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(231mg,0.608mmol)和N,N-二异丙基乙胺(157mg,1.216mmol)加入到超干N,N-二甲基甲酰胺(2.5mL)中,然后加入2-甲氧基-7-氮杂螺[3.5]壬烷盐酸盐(53mg,0.275mmol),室温搅拌反应1.0小时。反应完全后加入水(30mL),然后用乙酸乙酯(30mL X 3)萃取,合并有机相并用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用正相硅胶色谱柱(洗脱剂:二氯甲烷/甲醇=20/1)纯化,得到目标中间体(S)-3-((R)-2-(1-乙基-8-(2-甲氧基-7-氮杂螺[3.5]壬烷-7-羰基)-5-氧代-1,2,3,5-四氢-4H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000034
-4-基)-1-羟基乙基)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(2-2)(110mg,收率:86.7%)。
4-((R)-2-((S)-2-(tert-butoxycarbonyl)-1,2,3,4-tetrahydroisoquinolin-3-yl)-2-hydroxyethyl base)-1-ethyl-5-oxo-2,3,4,5-tetrahydro-1H-benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000033
-8-carboxylic acid (1-8) (100mg, 0.196mmol), 2-(7-azabenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate ( 231mg, 0.608mmol) and N,N-diisopropylethylamine (157mg, 1.216mmol) were added to ultra-dry N,N-dimethylformamide (2.5mL), then 2-methoxy-7 -Azaspiro[3.5]nonane hydrochloride (53 mg, 0.275 mmol), stirred at room temperature for 1.0 hour. After the reaction was complete, water (30 mL) was added, then extracted with ethyl acetate (30 mL X 3), the combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by normal phase silica gel chromatography (eluent: dichloromethane/methanol=20/1) to obtain the target intermediate (S)-3-((R)-2-(1-ethyl-8- (2-Methoxy-7-azaspiro[3.5]nonane-7-carbonyl)-5-oxo-1,2,3,5-tetrahydro-4H-benzo[e][1,4 ] diazepine
Figure PCTCN2022143995-appb-000034
-4-yl)-1-hydroxyethyl)-3,4-dihydroisoquinoline-2(1H)-tert-butyl carboxylate (2-2) (110 mg, yield: 86.7%).
LCMS:Rt:2.062min;MS m/z(ESI):647.3[M+H] +. LCMS: Rt: 2.062min; MS m/z (ESI): 647.3[M+H] + .
步骤9:化合物I盐酸盐的制备Step 9: Preparation of compound I hydrochloride
Figure PCTCN2022143995-appb-000035
Figure PCTCN2022143995-appb-000035
室温下将(S)-3-((R)-2-(1-乙基-8-(2-甲氧基-7-氮杂螺[3.5]壬烷-7-羰基)-5-氧代-1,2,3,5-四氢-4H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000036
-4-基)-1-羟基乙基)-3,4-二氢异喹啉-2(1H)-羧酸叔丁酯(2-2)(110mg,0.170mmol)加入到4mol/L盐酸/二氧六环(2.5mL)溶液中。室温搅拌反应1.0小时。反应完全后将反应液减压浓缩,残余物用高效液相制备色谱法(洗脱剂梯度:
(S)-3-((R)-2-(1-ethyl-8-(2-methoxy-7-azaspiro[3.5]nonane-7-carbonyl)-5-oxo Generation-1,2,3,5-tetrahydro-4H-benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000036
-4-yl)-1-hydroxyethyl)-3,4-dihydroisoquinoline-2(1H)-tert-butyl carboxylate (2-2) (110mg, 0.170mmol) was added to 4mol/L hydrochloric acid /dioxane (2.5mL) solution. The reaction was stirred at room temperature for 1.0 hour. After the reaction was complete, the reaction solution was concentrated under reduced pressure, and the residue was subjected to high performance liquid chromatography (eluent gradient:
Figure PCTCN2022143995-appb-000037
Figure PCTCN2022143995-appb-000037
)纯化,得到目标化合物1-乙基-4-((R)-2-羟基-2-((S)-1,2,3,4-四氢异喹啉-3-基)乙基)-8-(2-甲氧基-7-氮杂螺[3.5]壬烷-7-羰基)-1,2,3,4-四氢-5H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000038
-5-酮(化合物I)盐酸盐(39.81mg,收率:42.8%)。
) purification to obtain the target compound 1-ethyl-4-((R)-2-hydroxyl-2-((S)-1,2,3,4-tetrahydroisoquinolin-3-yl) ethyl) -8-(2-Methoxy-7-azaspiro[3.5]nonane-7-carbonyl)-1,2,3,4-tetrahydro-5H-benzo[e][1,4]di Aza
Figure PCTCN2022143995-appb-000038
-5-Kone (compound I) hydrochloride (39.81 mg, yield: 42.8%).
1H NMR(400MHz,CD 3OD)δ7.64(d,J=7.6Hz,1H),7.35-7.22(m,4H),7.12(d,J=8Hz,2H),4.51-4.36(m,2H),4.33-4.30(m,1H),4.04-3.89(m,2H),3.73-3.61(m,7H),3.51-3.50(m,1H),3.38-3.34(m,4H),3.31-3.30(m,2H),3.22(s,3H),2.29-2.23(m,2H),1.71-1.68(m,4H),1.55(s,2H),1.17(t,J=7.0Hz,3H). 1 H NMR (400MHz, CD 3 OD) δ7.64(d, J=7.6Hz, 1H), 7.35-7.22(m, 4H), 7.12(d, J=8Hz, 2H), 4.51-4.36(m, 2H),4.33-4.30(m,1H),4.04-3.89(m,2H),3.73-3.61(m,7H),3.51-3.50(m,1H),3.38-3.34(m,4H),3.31- 3.30(m,2H),3.22(s,3H),2.29-2.23(m,2H),1.71-1.68(m,4H),1.55(s,2H),1.17(t,J=7.0Hz,3H) .
LCMS:Rt:1.355min;MS m/z(ESI):547.4[M+H] +. LCMS: Rt: 1.355min; MS m/z (ESI): 547.4[M+H] + .
通过电导法测定所得化合物I盐酸盐中的盐酸含量,实验仪器、试剂、方法和结果如下。Determination of the hydrochloric acid content in the obtained compound I hydrochloride by conductometric method, experimental apparatus, reagent, method and result are as follows.
实验仪器与试剂:高效液相色谱仪(岛津LC-20AD)、电导检测器(CDD-10Avp)、电子天平(Sartorius,MAS125P-1CE-DU)、氯化钠、对羟基苯甲酸、Bis-Tris(HPLC级,阿拉丁A1910012)、硼酸、超纯水;Experimental instruments and reagents: high performance liquid chromatography (Shimadzu LC-20AD), conductivity detector (CDD-10Avp), electronic balance (Sartorius, MAS125P-1CE-DU), sodium chloride, p-hydroxybenzoic acid, Bis- Tris (HPLC grade, Aladdin A1910012), boric acid, ultrapure water;
色谱信息:色谱柱——Shim-pack IC-A3;保护柱——Shim-pack IC-GA3;淋洗液——8 mmol/L对羟基苯甲酸、3.2mmol/L Bis-Tris、50mmol/L硼酸溶液;流速——1.5mL/min;柱温——40℃;进样量——50μL;电导检测器模式——阳离子模式;Chromatographic information: Chromatographic column—Shim-pack IC-A3; guard column—Shim-pack IC-GA3; eluent—8 mmol/L p-hydroxybenzoic acid, 3.2mmol/L Bis-Tris, 50mmol/L Boric acid solution; flow rate - 1.5mL/min; column temperature - 40°C; injection volume - 50μL; conductivity detector mode - positive ion mode;
溶液配制:Solution preparation:
对照品溶液(含氯离子6μg/mL):精密量取氯化钠50mg至50mL量瓶中,加水溶解并稀释至刻度,摇匀,精密移取1.0mL至100mL量瓶中,加水稀释至刻度,摇匀。Reference solution (containing 6 μg/mL of chloride ion): Accurately measure 50 mg of sodium chloride into a 50 mL measuring bottle, add water to dissolve and dilute to the mark, shake well, precisely pipette 1.0 mL into a 100 mL measuring bottle, add water to dilute to the mark , shake well.
供试品溶液(0.1mg/mL):精密称取待测样品10mg至100mL量瓶中,用水溶解并稀释至刻度,摇匀。Test solution (0.1mg/mL): Accurately weigh 10mg of the sample to be tested into a 100mL measuring bottle, dissolve and dilute to the mark with water, and shake well.
测定方法:分别精密量取对照品溶液与供试品溶液,注入高效液相色谱仪,以淋洗液作为洗脱剂进行洗脱,记录色谱图。Determination method: Accurately measure the reference substance solution and the test solution respectively, inject them into a high-performance liquid chromatograph, use the eluent as the eluent to elute, and record the chromatogram.
实验结果:结果测得所述化合物I盐酸盐中的HCl含量为10.57%(化合物I一盐酸盐的HCl含量理论值为6.25%)。Experimental results: As a result, it was measured that the HCl content in the compound I hydrochloride was 10.57% (the theoretical value of the HCl content of the compound I monohydrochloride was 6.25%).
实施例3:1-乙基-4-((R)-2-羟基-2-((S)-1,2,3,4-四氢异喹啉-3-基)乙基)-8-(2-甲氧基-7-氮杂螺[3.5]壬烷-7-羰基)-1,2,3,4-四氢-5H-苯并[e][1,4]二氮杂
Figure PCTCN2022143995-appb-000039
-5-酮(化合物I)的制备
Example 3: 1-ethyl-4-((R)-2-hydroxy-2-((S)-1,2,3,4-tetrahydroisoquinolin-3-yl)ethyl)-8 -(2-Methoxy-7-azaspiro[3.5]nonane-7-carbonyl)-1,2,3,4-tetrahydro-5H-benzo[e][1,4]diazepine
Figure PCTCN2022143995-appb-000039
Preparation of -5-ketone (compound I)
室温下将按实施例2方法制得的3.0g化合物I盐酸盐加入100mL单口瓶中,加入12mL二氯甲烷,溶清后加入12mL纯化水,搅拌中加入20%氢氧化钠溶液调pH至水相pH为10左右,分液,用3ml二氯甲烷萃取三次水相,将有机相合并,用10mL纯化水反洗一次有机相,将有机相用无水硫酸钠干燥,减压浓缩至干,得2.5g泡沫状固体,即化合物I。At room temperature, add 3.0 g of compound I hydrochloride prepared according to the method of Example 2 into a 100 mL single-necked bottle, add 12 mL of dichloromethane, add 12 mL of purified water after dissolving, and add 20% sodium hydroxide solution during stirring to adjust the pH to The pH of the aqueous phase is about 10, separate the liquids, extract the aqueous phase three times with 3ml dichloromethane, combine the organic phases, backwash the organic phase once with 10mL purified water, dry the organic phase with anhydrous sodium sulfate, and concentrate to dryness under reduced pressure , to obtain 2.5g foamy solid, that is, compound I.
1H NMR(400MHz,DMSO-d 6)δ7.49-7.41(m,1H),7.17-7.04(m,3H),7.01(d,J=5.5Hz,1H),6.90-6.79(m,2H),5.01(d,J=5.5Hz,1H),4.03(q,J=7.1Hz,1H),3.98-3.79(m,4H),3.75(s,1H),3.63-3.35(m,6H),3.32-3.19(m,3H),3.18-3.12(m,2H),3.10(s,3H),2.84-2.65(m,3H),2.15(s,2H),1.70-1.39(m,6H),1.07(t,J=7.0Hz,3H). 1 H NMR (400MHz,DMSO-d 6 )δ7.49-7.41(m,1H),7.17-7.04(m,3H),7.01(d,J=5.5Hz,1H),6.90-6.79(m,2H ),5.01(d,J=5.5Hz,1H),4.03(q,J=7.1Hz,1H),3.98-3.79(m,4H),3.75(s,1H),3.63-3.35(m,6H) ,3.32-3.19(m,3H),3.18-3.12(m,2H),3.10(s,3H),2.84-2.65(m,3H),2.15(s,2H),1.70-1.39(m,6H) ,1.07(t,J=7.0Hz,3H).
实施例4:化合物I氢溴酸盐C晶型的制备Embodiment 4: Preparation of compound I hydrobromide C crystal form
室温下将实施例3制得的化合物I(350mg)加入50mL单口瓶中,加入异丙醇(3.5mL),搅拌溶清后,滴加107.9mg 40%氢溴酸水溶液,搅拌至析出大量固体,固体抽滤,滤饼用1mL异丙醇淋洗、25℃真空干燥,得到固体(230mg,HPLC纯度:99.19%)。Add the compound I (350mg) prepared in Example 3 into a 50mL single-necked bottle at room temperature, add isopropanol (3.5mL), stir to dissolve, then add 107.9mg of 40% hydrobromic acid aqueous solution dropwise, and stir until a large amount of solids are precipitated , the solid was suction-filtered, the filter cake was washed with 1 mL of isopropanol, and dried under vacuum at 25°C to obtain a solid (230 mg, HPLC purity: 99.19%).
1H NMR(400MHz,DMSO-d 6)δ9.21(s,1H),9.00(s,1H),7.46(d,J=7.7Hz,1H),7.35-7.16(m,4H),6.93-6.81(m,2H),5.90(d,J=6.0Hz,1H),4.44-4.15(m,4H),3.99-3.84(m,2H),3.52(s,5H),3.43-3.35(m,2H),3.22-3.13(m,4H),3.10(s,3H),2.15(s,2H),1.67-1.39(m,6H),1.05(dd,J=13.8,6.6Hz,5H). 1 H NMR (400MHz, DMSO-d 6 )δ9.21(s, 1H), 9.00(s, 1H), 7.46(d, J=7.7Hz, 1H), 7.35-7.16(m, 4H), 6.93- 6.81(m,2H),5.90(d,J=6.0Hz,1H),4.44-4.15(m,4H),3.99-3.84(m,2H),3.52(s,5H),3.43-3.35(m, 2H),3.22-3.13(m,4H),3.10(s,3H),2.15(s,2H),1.67-1.39(m,6H),1.05(dd,J=13.8,6.6Hz,5H).
该结晶样品的XRPD图谱见图2,其XRPD衍射峰位置如下表2所示;其DSC图谱见图3,峰值在238.84℃附近;其TGA图谱见图4。The XRPD spectrum of the crystalline sample is shown in Figure 2, and the XRPD diffraction peak positions are shown in Table 2 below; the DSC spectrum is shown in Figure 3, and the peak is around 238.84°C; the TGA spectrum is shown in Figure 4.
表2Table 2
Figure PCTCN2022143995-appb-000040
Figure PCTCN2022143995-appb-000040
Figure PCTCN2022143995-appb-000041
Figure PCTCN2022143995-appb-000041
实施例5:化合物I 1,5-萘二磺酸盐D晶型的制备Example 5: Preparation of Compound I 1,5-Naphthalene Disulfonate Form D
室温下将实施例3制得的化合物I(350mg)加入50mL单口瓶中,加入异丙醇(3.5mL),搅拌溶清,将1,5-萘二磺酸(184.8mg)溶于2.1mL纯化水中,然后滴入单口瓶中,搅拌过夜,将析出的固体抽滤,滤饼用1mL异丙醇淋洗、25℃真空干燥,得到固体(390mg,HPLC纯度:99.15%)。Add the compound I (350 mg) prepared in Example 3 into a 50 mL single-necked bottle at room temperature, add isopropanol (3.5 mL), stir to dissolve, and dissolve 1,5-naphthalene disulfonic acid (184.8 mg) in 2.1 mL Purified water was then dropped into a one-necked bottle, stirred overnight, and the precipitated solid was suction-filtered, the filter cake was rinsed with 1 mL of isopropanol, and vacuum-dried at 25°C to obtain a solid (390 mg, HPLC purity: 99.15%).
1H NMR(400MHz,DMSO-d 6)δ9.22(d,J=9.9Hz,1H),9.00(d,J=8.8Hz,1H),8.86(d,J=8.8Hz,2H),7.93(dd,J=7.0,1.0Hz,2H),7.52-7.37(m,3H),7.33-7.19(m,4H),6.92-6.80(m,2H),4.44-4.27(m,4H),4.04-3.70(m,3H),3.62-3.42(m,5H),3.43-3.20(m,4H),3.14(d,J=8.9Hz,4H),3.10(s,3H),2.15(s,2H),1.68-1.39(m,6H),1.05(t,J=7.0Hz,3H). 1 H NMR (400MHz, DMSO-d 6 ) δ9.22(d, J=9.9Hz, 1H), 9.00(d, J=8.8Hz, 1H), 8.86(d, J=8.8Hz, 2H), 7.93 (dd,J=7.0,1.0Hz,2H),7.52-7.37(m,3H),7.33-7.19(m,4H),6.92-6.80(m,2H),4.44-4.27(m,4H),4.04 -3.70(m,3H),3.62-3.42(m,5H),3.43-3.20(m,4H),3.14(d,J=8.9Hz,4H),3.10(s,3H),2.15(s,2H ),1.68-1.39(m,6H),1.05(t,J=7.0Hz,3H).
该结晶样品的XRPD见图5,其XRPD衍射峰位置如下表3所示;其DSC图谱见图6,其具有在231.85℃附近的峰值;其TGA图谱见图7。The XRPD of this crystalline sample is shown in Figure 5, and its XRPD diffraction peak positions are shown in Table 3 below; its DSC spectrum is shown in Figure 6, which has a peak around 231.85°C; its TGA spectrum is shown in Figure 7.
表3table 3
Figure PCTCN2022143995-appb-000042
Figure PCTCN2022143995-appb-000042
Figure PCTCN2022143995-appb-000043
Figure PCTCN2022143995-appb-000043
实施例6:化合物I一盐酸盐A晶型的制备Embodiment 6: Preparation of compound I-hydrochloride A crystal form
室温下将实施例3制得的化合物I(1.95g)加入100mL单口瓶中,加入乙酸甲酯(30mL),搅拌溶清后置于冰水浴中,缓慢滴加1.78mL 2mol/L氯化氢-乙酸乙酯溶液,搅拌至析出大量固体后抽滤,将滤饼用5mL乙酸甲酯淋洗、25℃真空干燥,得到固体(1.87g,HPLC纯度:98.98%)。Add the compound I (1.95g) prepared in Example 3 into a 100mL single-necked bottle at room temperature, add methyl acetate (30mL), stir to dissolve and place it in an ice-water bath, slowly add 1.78mL 2mol/L hydrogen chloride-acetic acid dropwise The ethyl ester solution was stirred until a large amount of solids were precipitated, and then suction-filtered, the filter cake was rinsed with 5 mL of methyl acetate, and vacuum-dried at 25°C to obtain a solid (1.87 g, HPLC purity: 98.98%).
1H NMR(400MHz,DMSO-d 6)δ9.73(s,1H),9.04(s,1H),7.45(d,J=7.7Hz,1H),7.26(dd,J=7.7,4.4Hz,4H),6.91-6.81(m,2H),5.94(d,J=5.7Hz,1H),4.28(d,J=16.0Hz,3H),3.99(dd,J=13.8,6.1Hz,1H),3.84(s,1H),3.48(d,J=30.3Hz,5H),3.39(dd,J=13.4,7.0Hz,2H),3.32-3.23(m,2H),3.22-3.12(m,5H),3.10(s,3H),2.15(s,2H),1.67-1.40(m,6H),1.04(t,J=7.0Hz,3H). 1 H NMR (400MHz, DMSO-d 6 )δ9.73(s, 1H), 9.04(s, 1H), 7.45(d, J=7.7Hz, 1H), 7.26(dd, J=7.7, 4.4Hz, 4H),6.91-6.81(m,2H),5.94(d,J=5.7Hz,1H),4.28(d,J=16.0Hz,3H),3.99(dd,J=13.8,6.1Hz,1H), 3.84(s,1H),3.48(d,J=30.3Hz,5H),3.39(dd,J=13.4,7.0Hz,2H),3.32-3.23(m,2H),3.22-3.12(m,5H) ,3.10(s,3H),2.15(s,2H),1.67-1.40(m,6H),1.04(t,J=7.0Hz,3H).
通过电位滴定法对A晶型中的盐酸含量进行测定,确定所述盐中化合物I与HCl的摩尔比约为1:1。The content of hydrochloric acid in crystal form A was determined by potentiometric titration, and it was determined that the molar ratio of compound I to HCl in the salt was about 1:1.
电位滴定法测定盐酸含量的实验仪器、试剂和方法如下。The experimental instruments, reagents and methods for determining the content of hydrochloric acid by potentiometric titration are as follows.
实验仪器与试剂:电位滴定仪(Metrohm,型号888)、电子天平(Mettler Toledo,型号XS205DU);甲醇(色谱级,稀释至60%甲醇作为溶剂和空白)、0.1mol/L氢氧化钠滴定液(南京化学试剂股份有限公司,批号210926428W);Experimental instruments and reagents: potentiometric titrator (Metrohm, model 888), electronic balance (Mettler Toledo, model XS205DU); methanol (chromatographic grade, diluted to 60% methanol as solvent and blank), 0.1mol/L sodium hydroxide titration solution (Nanjing Chemical Reagent Co., Ltd., batch number 210926428W);
滴定参数:Titration parameters:
Figure PCTCN2022143995-appb-000044
Figure PCTCN2022143995-appb-000044
滴定过程:Titration process:
空白滴定:量取溶剂(60%甲醇)50mL置于100mL烧杯中,用氢氧化钠滴定液(0.1mol/L)滴定至终点。Blank titration: Measure 50 mL of solvent (60% methanol) into a 100 mL beaker, and titrate to the end point with sodium hydroxide titration solution (0.1 mol/L).
待测样品滴定:精密称取待测样品300mg,置于100mL烧杯中,加入溶剂(60%甲醇)50mL,用氢氧化钠滴定液(0.1mol/L)滴定至终点。Titration of the sample to be tested: Accurately weigh 300 mg of the sample to be tested, place it in a 100 mL beaker, add 50 mL of solvent (60% methanol), and titrate to the end point with sodium hydroxide titration solution (0.1 mol/L).
盐酸含量通过以下公式计算:The hydrochloric acid content is calculated by the following formula:
Figure PCTCN2022143995-appb-000045
Figure PCTCN2022143995-appb-000045
备注:Remark:
V 0:空白消耗氢氧化钠滴定液的体积,单位(mL); V 0 : blank consumes the volume of sodium hydroxide titration solution, unit (mL);
V:待测样品消耗氢氧化钠滴定液的体积,单位(mL);V: the volume of sodium hydroxide titration solution consumed by the sample to be tested, unit (mL);
C:氢氧化钠滴定液的浓度,单位(mol/L);C: concentration of sodium hydroxide titration solution, unit (mol/L);
m:待测样品的称样量,单位(mg);m: the weighing amount of the sample to be tested, unit (mg);
3.646:氢氧化钠滴定液滴定度,mg/mL。3.646: titer of sodium hydroxide titration solution, mg/mL.
该结晶样品的XRPD见图8,其XRPD衍射峰位置如下表4所示;其DSC图谱见图9,峰值在259.27℃附近;其TGA图谱见图10。The XRPD of the crystalline sample is shown in Figure 8, and the XRPD diffraction peak positions are shown in Table 4 below; the DSC spectrum is shown in Figure 9, and the peak is around 259.27°C; the TGA spectrum is shown in Figure 10.
表4Table 4
Figure PCTCN2022143995-appb-000046
Figure PCTCN2022143995-appb-000046
Figure PCTCN2022143995-appb-000047
Figure PCTCN2022143995-appb-000047
实施例7:化合物I一盐酸盐A晶型的制备Embodiment 7: Preparation of compound I-hydrochloride A crystal form
室温下将实施例3制得的化合物I(500mg)加入50mL单口瓶中,加入乙酸乙酯(5mL),搅拌溶清后置于冰水浴中,缓慢滴加445μL 2mol/L氯化氢-乙酸乙酯溶液,搅拌至析出大量固体后抽滤,将滤饼用1mL乙酸乙酯淋洗、25℃真空干燥,得到固体(429mg)。Add the compound I (500mg) prepared in Example 3 into a 50mL single-necked bottle at room temperature, add ethyl acetate (5mL), stir to dissolve, place it in an ice-water bath, and slowly add 445 μL 2mol/L hydrogen chloride-ethyl acetate dropwise The solution was stirred until a large amount of solids were precipitated, and then suction-filtered, the filter cake was rinsed with 1 mL of ethyl acetate, and vacuum-dried at 25°C to obtain a solid (429 mg).
实施例8:化合物I一盐酸盐A晶型的制备Embodiment 8: Preparation of compound I-hydrochloride A crystal form
室温下将实施例3制得的化合物I(250mg)加入50mL单口瓶中,加入丙酮(2.5mL),搅拌溶清后置于冰水浴中,缓慢滴加220μL 2mol/L氯化氢-乙酸乙酯溶液,搅拌至析出大量固体后抽滤,将滤饼用1mL丙酮淋洗、25℃真空干燥,得到固体(166mg)。Add the compound I (250mg) prepared in Example 3 into a 50mL single-necked bottle at room temperature, add acetone (2.5mL), stir to dissolve, place in an ice-water bath, and slowly add 220 μL of 2mol/L hydrogen chloride-ethyl acetate solution dropwise , stirred until a large amount of solids were precipitated, then filtered with suction, rinsed the filter cake with 1 mL of acetone, and dried in vacuo at 25°C to obtain a solid (166 mg).
实施例9:化合物I一盐酸盐A晶型的制备Embodiment 9: Preparation of compound I-hydrochloride A crystal form
室温下将实施例3制得的化合物I(4.31g)加入500mL单口瓶中,加入异丙醇(43mL),搅拌溶清后置于冰水浴中,缓慢滴加3.9mL 2mol/L氯化氢-乙酸乙酯溶液,搅拌至析出大量固体后抽滤,将滤饼用5mL异丙醇淋洗、25℃真空干燥,得到固体(3.74g)。Add the compound I (4.31g) prepared in Example 3 into a 500mL single-necked bottle at room temperature, add isopropanol (43mL), stir to dissolve and place it in an ice-water bath, slowly add 3.9mL 2mol/L hydrogen chloride-acetic acid dropwise Ethyl ester solution, stirred until a large amount of solids were precipitated, then filtered with suction, rinsed the filter cake with 5 mL of isopropanol, and dried under vacuum at 25°C to obtain a solid (3.74 g).
实施例10:化合物I一盐酸盐无定型物的制备Embodiment 10: Preparation of compound I-hydrochloride amorphous
室温下将实施例6制得的化合物I一盐酸盐A晶型(300mg)加入甲醇(10mL)中,搅拌溶清后减压浓缩得泡沫状固体,经XRPD检测为化合物I一盐酸盐无定型物。Add the compound I-hydrochloride A crystal form (300mg) prepared in Example 6 into methanol (10mL) at room temperature, stir to dissolve, and concentrate under reduced pressure to obtain a foamy solid, which is detected as compound I-hydrochloride by XRPD Amorphous.
实施例11:化合物I一盐酸盐A晶型的制备Example 11: Preparation of compound I-hydrochloride A crystal form
室温下将实施例10制得的化合物I一盐酸盐无定型物(10mg)加入5mL离心管中,加入乙酸甲酯(100μL),形成混悬液,室温打浆过夜,固体抽滤,滤饼用1mL乙酸甲酯洗涤、25℃真空干燥得固体(8mg)。经XRPD检测为A晶型。Add compound I-amorphous hydrochloride (10mg) prepared in Example 10 into a 5mL centrifuge tube at room temperature, add methyl acetate (100μL) to form a suspension, beat at room temperature overnight, and filter the solid with suction. Wash with 1 mL of methyl acetate and dry under vacuum at 25°C to give a solid (8 mg). It was detected as crystal form A by XRPD.
实施例12:化合物I一盐酸盐A晶型的制备Example 12: Preparation of compound I-hydrochloride A crystal form
室温下,将实施例10制得的化合物I一盐酸盐无定型物(20mg)加入4.5mL离心管中,加入甲醇200μL),溶清后滴加丙酮至有固体析出,搅拌数小时后将析出的固体抽滤,滤饼用1mL丙酮洗涤、25℃真空干燥得固体(12mg)。At room temperature, add the compound I-hydrochloride amorphous (20 mg) prepared in Example 10 into a 4.5 mL centrifuge tube, add methanol (200 μL), dissolve and add acetone dropwise until solid precipitates, stir for several hours The precipitated solid was suction filtered, the filter cake was washed with 1 mL of acetone, and dried under vacuum at 25°C to obtain a solid (12 mg).
实施例13:化合物I一盐酸盐B晶型的制备Example 13: Preparation of compound I-hydrochloride B crystal form
室温下,将实施例10制得的化合物I一盐酸盐无定型物(20mg)加入4.5mL离心管中,加入99%甲醇水溶液(200μL),溶清后滴加乙酸甲酯(1.2mL)有固体析出,搅拌数小时后将析出的固体抽滤,滤饼用1mL乙酸甲酯洗涤、25℃真空干燥得固体(14mg)。At room temperature, add Compound I-amorphous hydrochloride (20 mg) prepared in Example 10 into a 4.5 mL centrifuge tube, add 99% aqueous methanol (200 μL), dissolve and add methyl acetate (1.2 mL) dropwise A solid was precipitated, and after stirring for several hours, the precipitated solid was suction-filtered, the filter cake was washed with 1 mL of methyl acetate, and dried in vacuo at 25°C to obtain a solid (14 mg).
该结晶样品的XRPD见图11,其XRPD衍射峰位置如下表5所示;其DSC图谱见图12;其TGA图谱见图13。The XRPD of the crystalline sample is shown in Figure 11, and its XRPD diffraction peak positions are shown in Table 5 below; its DSC spectrum is shown in Figure 12; its TGA spectrum is shown in Figure 13.
表5table 5
Figure PCTCN2022143995-appb-000048
Figure PCTCN2022143995-appb-000048
Figure PCTCN2022143995-appb-000049
Figure PCTCN2022143995-appb-000049
测试试验例1:PRMT5酶学活性抑制实验Test Test Example 1: PRMT5 Enzymatic Activity Inhibition Experiment
材料:PRMT5/MEP50蛋白购于BPS bioscience公司(美国);组蛋白H4多肽(Histone H4Peptide)底物购于生工生物工程(上海)股份有限公司;Anti-Histone H4(symmetric dimethyl R3)抗体-ChIP Grade购于艾博抗公司(美国);S-(5’-腺苷)-L-甲硫氨酸氯化物二盐酸盐购于西格玛公司(美国);384孔板、AlphaScreen链霉亲和素供体微珠(AlphaScreen Streptavidin Donor beads)、AlphaScreen蛋白A受体微珠(AlphaScreen Protein A Acceptor beads)和多功能酶标仪Envision 2104multi-label Reader购于珀金埃尔默仪器有限公司(美国);Echo 550移液器(Echo 550Liquid Handler)购于Labcyte公司(美国)。Materials: PRMT5/MEP50 protein was purchased from BPS bioscience (USA); histone H4 peptide (Histone H4 Peptide) substrate was purchased from Sangon Bioengineering (Shanghai) Co., Ltd.; Anti-Histone H4 (symmetric dimethyl R3) antibody-ChIP Grade was purchased from Abcam (US); S-(5'-adenosyl)-L-methionine chloride dihydrochloride was purchased from Sigma (US); 384-well plate, AlphaScreen streptavidin AlphaScreen Streptavidin Donor beads, AlphaScreen Protein A Acceptor beads and Envision 2104 multi-label Reader were purchased from PerkinElmer Instruments Co., Ltd. (USA) Echo 550 pipette (Echo 550 Liquid Handler) was purchased from Labcyte (U.S.).
酶学活性检测:利用Echo将化合物打入384孔板中,使终浓度为0-1000nM(起始浓度1000nM,3倍稀释,10个点),DMSO含量为0.5%。每孔加入10μL 2X PRMT5/MEP50溶液,常温孵育30分钟。每孔加入10μL 2X PRMT5/MEP50底物溶液启动反应,常温孵育60分钟。准备含AlphaScreen Protein A Acceptor beads和Anti-Histone H4(symmetric dimethyl R3)抗体的6X检测试剂,每孔加入5μL,常温孵育60分钟。准备含AlphaScreen Streptavidin Donor beads的6X检测试剂,每孔加入5μL,常温孵育60分钟。Envision检测信号值。测试结果见表6。Detection of enzymatic activity: the compound was injected into a 384-well plate by Echo, so that the final concentration was 0-1000 nM (initial concentration 1000 nM, 3-fold dilution, 10 points), and the DMSO content was 0.5%. Add 10 μL 2X PRMT5/MEP50 solution to each well and incubate at room temperature for 30 minutes. Add 10 μL 2X PRMT5/MEP50 substrate solution to each well to start the reaction, and incubate at room temperature for 60 minutes. Prepare 6X detection reagent containing AlphaScreen Protein A Acceptor beads and Anti-Histone H4 (symmetric dimethyl R3) antibody, add 5 μL to each well, and incubate at room temperature for 60 minutes. Prepare 6X detection reagent containing AlphaScreen Streptavidin Donor beads, add 5 μL to each well, and incubate at room temperature for 60 minutes. Envision detection signal value. The test results are shown in Table 6.
测试试验例2:化合物对肿瘤细胞增殖的抑制活性实验Test Test Example 2: Experiment of Inhibitory Activity of Compounds on Tumor Cell Proliferation
材料与细胞:Z-138细胞购于ATCC(美国);IMDM培养基和青霉素-链霉素购于西格玛公司(美国);马血清购于Hyclone公司(美国);96孔板购于康宁公司(美国);Cell-Titer Glo试剂购于普洛麦格公司(美国)。Materials and cells: Z-138 cells were purchased from ATCC (US); IMDM medium and penicillin-streptomycin were purchased from Sigma (US); horse serum was purchased from Hyclone (US); 96-well plates were purchased from Corning ( U.S.); Cell-Titer Glo reagent was purchased from Promega (U.S.).
细胞培养:Z-138细胞用含10%马血清+1%青霉素-链霉素的IMDM培养液于37℃、5%CO 2条件下培养。处于对数生长期细胞方可用于实验。 Cell culture: Z-138 cells were cultured in IMDM medium containing 10% horse serum + 1% penicillin-streptomycin at 37°C and 5% CO 2 . Cells in the logarithmic growth phase can be used for experiments.
细胞增殖活性检测:利用Cell-Titer Glo试剂检测化合物对Z-138细胞的增殖抑制活性。调整细胞浓度,每孔180μL接种96孔板(500/孔),置于37℃、5%CO 2条件下平衡10-15分钟。每孔加入20μL含化合物的培养液,使终浓度达到0-300nM(起始浓度300nM,3倍稀释,10个点),DMSO含量为0.1%。细胞板置于37℃、5%CO 2条件下孵育8天。其中第四天换液:缓慢吸去100μL上清,并补充100μL含有化合物的新鲜培养液,保持化合物浓度不变。通过Cell-Titer Glo试剂检测细胞活性。测试结果见表6。 Detection of cell proliferation activity: Cell-Titer Glo reagent was used to detect the proliferation inhibitory activity of the compound on Z-138 cells. Adjust the cell concentration, inoculate 96-well plates with 180 μL per well (500/well), and place them at 37° C. and 5% CO 2 to equilibrate for 10-15 minutes. 20 μL of compound-containing culture solution was added to each well to make the final concentration 0-300 nM (initial concentration 300 nM, 3-fold dilution, 10 points), and the DMSO content was 0.1%. Cell plates were incubated at 37°C, 5% CO 2 for 8 days. The medium was changed on the fourth day: 100 μL of the supernatant was slowly aspirated, and 100 μL of fresh culture medium containing the compound was added to keep the concentration of the compound unchanged. Cell viability was detected by Cell-Titer Glo reagent. The test results are shown in Table 6.
测试试验例3:化合物对SDMA的抑制活性实验Test Test Example 3: Compound's inhibitory activity experiment on SDMA
材料与细胞:Z-138细胞购于ATCC(美国);IMDM培养基和青霉素-链霉素购于西格玛公司(美国);马血清购于Hyclone公司(美国);Hoechst抗体购于invitrogen公司(美国);Alexa Fluor 488 goat anti-rabbit IgG抗体购于Invitrogen公司(美国);Anti-dimethyl-Arginine symmetric(SYM11)抗体购于Merck公司(美国);DPBS购于Gibco公司(美国);Nonfat Dry milk购于Cell signaling technology公司(美国);多聚甲醛购于北京索莱宝科技有限公司;384 孔板和Echo 550 Liquid Handler购于Labcyte公司(美国);ImageXpress Nano购于Molecular Devices公司(美国)。Materials and cells: Z-138 cells were purchased from ATCC (US); IMDM medium and penicillin-streptomycin were purchased from Sigma (US); horse serum was purchased from Hyclone (US); Hoechst antibody was purchased from Invitrogen (US) ); Alexa Fluor 488 goat anti-rabbit IgG antibody was purchased from Invitrogen (U.S.); Anti-dimethyl-Arginine symmetric (SYM11) antibody was purchased from Merck (U.S.); DPBS was purchased from Gibco (U.S.); From Cell signaling technology company (USA); paraformaldehyde was purchased from Beijing Suo Laibao Technology Co., Ltd.; 384-well plate and Echo 550 Liquid Handler were purchased from Labcyte Company (USA); ImageXpress Nano was purchased from Molecular Devices company (USA).
细胞培养:Z-138细胞用含10%马血清+1%青霉素-链霉素的IMDM培养液于37℃、5%CO 2条件下培养。处于对数生长期细胞方可用于实验。 Cell culture: Z-138 cells were cultured in IMDM medium containing 10% horse serum + 1% penicillin-streptomycin at 37°C and 5% CO 2 . Cells in the logarithmic growth phase can be used for experiments.
免疫荧光检测:利用免疫荧光检测化合物对Z-138细胞中SDMA的影响。调整细胞浓度为1*10 5/mL,每孔40μL接种384孔板(4000/孔),置于37℃、5%CO 2条件下平衡10-15分钟。利用Echo将化合物打入384孔板中,使终浓度为0-300nM(起始浓度300nM,3倍稀释,10个点),DMSO含量为0.1%。细胞板置于37℃、5%CO 2条件下孵育2天。每孔加入40μL 8%多聚甲醛,常温孵育30分钟。弃上清,DPBS洗板,每孔加入40μL 0.5%PBST(磷酸盐吐温缓冲液),常温孵育60分钟。弃上清,0.05%PBST洗板,每孔加入40μL封闭液(1%Nonfat milk in 0.05%PBST),常温孵育60分钟。弃上清,每孔加入20μL一抗(SYM11,1:500封闭液稀释),4℃过夜。弃上清,0.05%PBST洗板,每孔加入20μL二抗(Alexa Fluor 488goat anti-rabbit 1:1000和Hoechst 1:5000封闭液稀释),常温孵育60分钟。弃上清,0.05%PBST洗板,ImageXpress Nano检测荧光强度。测试结果见表6。 Immunofluorescence detection: Immunofluorescence was used to detect the effect of compounds on SDMA in Z-138 cells. The cell concentration was adjusted to 1*10 5 /mL, 40 μL per well was inoculated into a 384-well plate (4000/well), and placed at 37° C., 5% CO 2 to equilibrate for 10-15 minutes. The compound was injected into a 384-well plate by Echo, so that the final concentration was 0-300 nM (initial concentration 300 nM, 3-fold dilution, 10 points), and the DMSO content was 0.1%. Cell plates were incubated at 37°C, 5% CO 2 for 2 days. Add 40 μL of 8% paraformaldehyde to each well and incubate at room temperature for 30 minutes. Discard the supernatant, wash the plate with DPBS, add 40 μL of 0.5% PBST (phosphate Tween buffer) to each well, and incubate at room temperature for 60 minutes. Discard the supernatant, wash the plate with 0.05% PBST, add 40 μL of blocking solution (1% Nonfat milk in 0.05% PBST) to each well, and incubate at room temperature for 60 minutes. Discard the supernatant, add 20 μL primary antibody (SYM11, 1:500 dilution in blocking solution) to each well, and leave overnight at 4°C. Discard the supernatant, wash the plate with 0.05% PBST, add 20 μL of secondary antibody (diluted in Alexa Fluor 488goat anti-rabbit 1:1000 and Hoechst 1:5000 in blocking solution) to each well, and incubate at room temperature for 60 minutes. Discard the supernatant, wash the plate with 0.05% PBST, and detect the fluorescence intensity with ImageXpress Nano. The test results are shown in Table 6.
表6Table 6
Figure PCTCN2022143995-appb-000050
Figure PCTCN2022143995-appb-000050
测试试验例4:小鼠药代动力学实验Test Example 4: Pharmacokinetic experiment in mice
实验材料:CB17-SCID小鼠购自北京维通利华实验动物技术有限公司;DMSO、HP-β-CD(羟丙基-β-环糊精)、MC(甲基纤维素)、乙腈购自Merck公司(美国)、K 2EDTA抗凝管购自江苏新康医疗器械有限公司。 Experimental materials: CB17-SCID mice were purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd.; DMSO, HP-β-CD (hydroxypropyl-β-cyclodextrin), MC (methylcellulose), and acetonitrile were purchased from From Merck (USA), K 2 EDTA anticoagulant tube was purchased from Jiangsu Xinkang Medical Instrument Co., Ltd.
实验方法:雌性CB17-SCID小鼠6只(20-30g,4-6周),随机分成2组,每组3只。第1组尾静脉注射给予化合物,剂量为2mg/kg,溶媒为5%DMSO+95%10%HP-β-CD的水溶液,第2组口服给予化合物,剂量10mg/kg,溶媒为0.5%MC水溶液。动物实验前正常喂食喂水。每组小鼠于给药前及给药后0.083(仅静脉注射组)、0.25、0.5、1、2、4、6、8和24h进行静脉采血。收集的全血样品置于K 2EDTA抗凝管中,离心5min后(4000rpm,4℃)取血浆待测。 Experimental method: 6 female CB17-SCID mice (20-30g, 4-6 weeks) were randomly divided into 2 groups, 3 mice in each group. Group 1 was given the compound by tail vein injection, the dose was 2mg/kg, the vehicle was 5% DMSO+95% 10% HP-β-CD aqueous solution, the second group was orally administered the compound, the dose was 10mg/kg, the vehicle was 0.5% MC aqueous solution. Animals were fed and watered normally before the experiment. Venous blood was collected from mice in each group before administration and at 0.083 (intravenous injection group only), 0.25, 0.5, 1, 2, 4, 6, 8 and 24 hours after administration. The collected whole blood samples were placed in K 2 EDTA anticoagulant tubes, centrifuged for 5 minutes (4000 rpm, 4° C.) and the plasma was collected for testing.
取小鼠血浆样品10μL,加入150μL乙腈溶剂(其中含内标化合物)沉淀蛋白,涡旋0.5min后,离心(4700rpm,4℃)15min,上清液用含0.05%(v/v)甲酸的水稀释2倍,进样3μL于LC-MS/MS系统(AB Sciex Triple Quad 6500+)进行定量检测。在测定样品浓度时随行CB17-SCID小鼠血浆标准曲线(线性范围:0.5-1000ng/mL)和质控样品。对于10倍稀释样品的制备,取2μL小鼠血浆样品加入18μL的空白血浆,涡旋0.5min后,加入300μL乙腈溶剂(其中含内标化合物)沉淀蛋白,其余处理步骤同不稀释样品。Take 10 μL of mouse plasma sample, add 150 μL of acetonitrile solvent (which contains internal standard compound) to precipitate protein, vortex for 0.5 min, centrifuge (4700 rpm, 4 ° C) for 15 min, and supernatant with 0.05% (v/v) formic acid Dilute 2 times with water, inject 3 μL into LC-MS/MS system (AB Sciex Triple Quad 6500+) for quantitative detection. When determining the sample concentration, the CB17-SCID mouse plasma standard curve (linear range: 0.5-1000ng/mL) and quality control samples were followed. For the preparation of 10-fold diluted samples, take 2 μL of mouse plasma samples and add 18 μL of blank plasma, vortex for 0.5 min, add 300 μL of acetonitrile solvent (including internal standard compounds) to precipitate proteins, and the rest of the processing steps are the same as for undiluted samples.
药代动力学测试结果如表7所示。The pharmacokinetic test results are shown in Table 7.
表7小鼠药代动力学测试结果Table 7 Mouse pharmacokinetic test results
Figure PCTCN2022143995-appb-000051
Figure PCTCN2022143995-appb-000051
测试试验例5:肝细胞代谢稳定性测试Test Example 5: Hepatocyte Metabolic Stability Test
实验材料:人肝细胞购自Biopredic公司;小鼠肝细胞购自BioIVT公司;乙腈和甲醇购自Merck公司;AOPI染色剂购自Nexcelom公司;地塞米松购自NIFDC公司;DMSO购自北京索莱宝科技有限公司;DPBS(10x)、GlutaMAX TM-1(100x)和人重组胰岛素购自Gibco by Life Technologies;胎牛血清购自Corning公司;甲酸购自DIKMAPURE公司;Isotonic Percoll购自GE Healthcare公司;阿普唑仑购自Supelco公司;咖啡因购自ChromaDex.inc;HEPES、甲苯磺丁脲和Williams’Medium E购自Sigma公司。 Experimental materials: human hepatocytes were purchased from Biopredic Company; mouse hepatocytes were purchased from BioIVT Company; acetonitrile and methanol were purchased from Merck Company; AOPI stain was purchased from Nexcelom Company; dexamethasone was purchased from NIFDC Company; Bao Technology Co., Ltd.; DPBS (10x), GlutaMAX TM -1 (100x) and human recombinant insulin were purchased from Gibco by Life Technologies; fetal bovine serum was purchased from Corning; formic acid was purchased from DIKMAPURE; Isotonic Percoll was purchased from GE Healthcare; Alprazolam was purchased from Supelco; caffeine was purchased from ChromaDex.inc; HEPES, tolbutamide and Williams' Medium E were purchased from Sigma.
实验准备:Experiment preparation:
将受试物粉末用DMSO配制高浓度储备液,使用前用乙腈稀释到100μM的工作液,受试物终浓度为1μM。Prepare a high-concentration stock solution from the powder of the test substance in DMSO, and dilute to a working solution of 100 μM with acetonitrile before use, and the final concentration of the test substance is 1 μM.
肝细胞复苏液的具体制备信息见下表8。混合49.5mL Williams’E Medium和0.5mL GlutaMAX作为孵育液。将肝细胞复苏液和孵育液于使用前置于37℃水浴中至少预热15分钟。取一管超低温保存的肝细胞,确保肝细胞在复苏之前仍处于低温冰冻状态。将肝细胞迅速置于37℃水浴中并轻摇直至所有冰晶全部分散,喷洒70%乙醇后转移至生物安全柜中。将肝细胞小管的内容物倾入盛有50mL复苏培养基的离心管中,将其于100g离心10分钟。离心后,吸出复苏培养基并加入足量孵育培养基得到细胞密度约1.5×10 6个细胞/mL的细胞混悬液。用Cellometer Vision对肝细胞进行计数及确定活细胞密度,肝细胞成活率必须大于75%。利用孵育培养基稀释肝细胞混悬液至活细胞密度为0.5×10 6个活细胞/mL。 The specific preparation information of the liver cell resuscitation solution is shown in Table 8 below. Mix 49.5mL Williams'E Medium and 0.5mL GlutaMAX as incubation solution. Preheat the liver cell resuscitation solution and incubation solution in a 37°C water bath for at least 15 minutes before use. Take a tube of cryopreserved hepatocytes and make sure the hepatocytes are still cryogenically frozen before resuscitating. The hepatocytes were quickly placed in a 37°C water bath and shaken until all ice crystals were dispersed, sprayed with 70% ethanol and transferred to a biological safety cabinet. The contents of the hepatocyte tubules were poured into a centrifuge tube containing 50 mL of resuscitation medium, which was centrifuged at 100 g for 10 minutes. After centrifugation, aspirate the recovery medium and add enough incubation medium to obtain a cell suspension with a cell density of about 1.5×10 6 cells/mL. Use Cellometer Vision to count liver cells and determine the density of live cells. The survival rate of liver cells must be greater than 75%. Dilute the hepatocyte suspension with incubation medium to a viable cell density of 0.5× 106 viable cells/mL.
表8肝细胞复苏液制备Table 8 Preparation of liver cell resuscitation solution
Figure PCTCN2022143995-appb-000052
Figure PCTCN2022143995-appb-000052
实验方法:experimental method:
转移247.5μL活细胞(人肝细胞或鼠肝细胞)的混悬液或培养基到96孔深孔板,将深孔板置于涡旋上孵箱中预热10分钟。所有样品均采用双平行孵育。每孔加入2.5μL 100μM受试物进行反应起始,将深孔板放回孵箱涡旋器上。孵育样品,分别于0、15、30、60、90和120分钟,取25μL混悬液,加入125μL含内标的乙腈(100nM阿普唑仑,200nM咖啡因,100nM甲苯磺丁脲)终止反应。涡旋10分钟,于3220g、4℃条件离心30分钟,离心结束后转移100μL上清液到进样板,加入150μL纯水混匀,用于LC-MS/MS分析。Transfer 247.5 μL of live cell (human hepatocytes or mouse hepatocytes) suspension or culture medium to a 96-well deep-well plate, and place the deep-well plate in a vortex-topped incubator to preheat for 10 minutes. All samples were incubated in double parallel. Add 2.5 μL of 100 μM test substance to each well to initiate the reaction, and put the deep well plate back on the incubator vortexer. Incubate the samples at 0, 15, 30, 60, 90 and 120 minutes respectively, take 25 μL of the suspension, add 125 μL of acetonitrile containing internal standard (100 nM alprazolam, 200 nM caffeine, 100 nM tolbutamide) to terminate the reaction. Vortex for 10 minutes, centrifuge at 3220g, 4°C for 30 minutes, transfer 100 μL of supernatant to the sample plate after centrifugation, add 150 μL of pure water and mix well for LC-MS/MS analysis.
所有的数据计算均通过Microsoft Excel软件进行。通过提取离子图谱检测峰面积。通过对母药消除百分比的自然对数与时间进行线性拟合,检测母药的体外半衰期(t 1/2)。 All data calculations were performed by Microsoft Excel software. Peak areas were detected by extracted ion spectra. The in vitro half-life (t 1/2 ) of the parent drug was determined by linear fitting the natural logarithm of the percent elimination of the parent drug with time.
体外半衰期(t 1/2)通过斜率计算: In vitro half-life (t 1/2 ) is calculated by the slope:
in vitro t 1/2=0.693/k in vitro t 1/2 =0.693/k
实验结果如表9所示。The experimental results are shown in Table 9.
表9肝细胞代谢稳定性测试结果Table 9 Hepatocyte metabolic stability test results
化合物compound 人t 1/2(min) Person t 1/2 (min) 鼠t 1/2(min) Rat 1/2 (min)
实施例2Example 2 252.37252.37 94.6294.62
测试试验例6:小鼠体内药效实验Test Test Example 6: Drug efficacy experiment in mice
实验材料:Z138细胞购自ATCC;IMDM培养液、青链霉素和0.25%胰酶-EDTA购自Gibco公司;马血清、PBS购自Hyclone公司;Matrigel购自Corning公司。Experimental materials: Z138 cells were purchased from ATCC; IMDM medium, penicillin and streptomycin and 0.25% trypsin-EDTA were purchased from Gibco; horse serum and PBS were purchased from Hyclone; Matrigel was purchased from Corning.
动物信息:CB17-SCID小鼠,雌性,6-7周,体重约14-20克,动物购自上海灵畅生物科技有限公司,将小鼠饲养在SPF级的环境中,每个笼位单独送排风,所有动物都可以自由获 取标准认证的商业实验室饮食和自由饮水。Animal information: CB17-SCID mice, female, 6-7 weeks old, weighing about 14-20 grams, were purchased from Shanghai Lingchang Biotechnology Co., Ltd. The mice were raised in an SPF-grade environment, and each cage was individually All animals had free access to a standard certified commercial laboratory diet and water ad libitum.
实验方法:experimental method:
细胞培养:人套细胞淋巴瘤Z-138细胞株体外培养,培养条件为IMDM(细胞培养液)中加入10%马血清,1%青链霉素溶液,37℃、5%CO 2孵箱。一周两次用0.25%胰酶-EDTA消化液进行常规消化处理传代。当细胞饱和度为85%-90%,数量达到要求时,收取细胞,计数。 Cell culture: human mantle cell lymphoma Z-138 cell line was cultured in vitro, the culture condition was adding 10% horse serum, 1% penicillin and streptomycin solution to IMDM (cell culture medium), 37°C, 5% CO2 incubator. Routine digestion with 0.25% trypsin-EDTA digestion solution was performed twice a week for subculture. When the cell saturation is 85%-90% and the number reaches the requirement, collect the cells and count them.
细胞接种:将0.1mL/(含1×10 7)Z-138细胞悬液(PBS:Matrigel=1:1)皮下接种于每只小鼠的右后背。在接种后第18天,测量肿瘤平均体积达到约125mm 3时,依据肿瘤体积和动物体重采用随机分层分组方法开始分组给药。PBS为无钙镁离子的磷酸缓冲盐溶液,Matrigel是基质胶。 Cell inoculation: 0.1 mL/(containing 1×10 7 ) Z-138 cell suspension (PBS:Matrigel=1:1) was subcutaneously inoculated on the right back of each mouse. On the 18th day after inoculation, when the average volume of the measured tumor reached about 125 mm 3 , the grouping method was randomly stratified and administered according to the tumor volume and animal body weight. PBS is phosphate-buffered saline without calcium and magnesium ions, and Matrigel is Matrigel.
给药:实施例2化合物的给药剂量为1.5mg/kg、5mg/kg或15mg/kg,PO,每天两次给药(BID)x 3周。每组6只小鼠。Administration: The dosage of the compound of Example 2 is 1.5mg/kg, 5mg/kg or 15mg/kg, PO, administered twice a day (BID) x 3 weeks. 6 mice per group.
肿瘤测量和实验指标:Tumor measurements and experimental indicators:
每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a x b 2,a和b分别表示肿瘤的长径和短径。每周两次测量小鼠体重。 Tumor diameters were measured twice a week with vernier calipers. The formula for calculating the tumor volume is: V=0.5a x b 2 , where a and b represent the long diameter and short diameter of the tumor, respectively. Mouse body weights were measured twice a week.
化合物的抑瘤疗效用肿瘤生长抑制率TGI(%)来评价。The antitumor efficacy of compounds was evaluated by tumor growth inhibition rate TGI (%).
TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]x100%。TGI (%)=[(1-(average tumor volume at the end of administration of a certain treatment group-average tumor volume at the beginning of administration of this treatment group)/(average tumor volume at the end of treatment of the solvent control group-at the beginning of treatment of the solvent control group Mean tumor volume)] x 100%.
实验结果:Experimental results:
见表10、图14和图15。实验过程中无小鼠发病或死亡。See Table 10, Figure 14 and Figure 15. No mice became ill or died during the experiment.
表10 Z-138皮下瘤模型肿瘤体积Table 10 Z-138 subcutaneous tumor model tumor volume
Figure PCTCN2022143995-appb-000053
Figure PCTCN2022143995-appb-000053
实验结论:Experimental results:
在小鼠皮下移植瘤Z-138模型中,本公开实施例2化合物在1.5mg/kg、5mg/kg和15mg/kg一天两次给药对肿瘤生长都具有显著抑制作用,并呈现较好的剂量反应关系,15mg/kg一天两次给药,具有缩小肿瘤的效果。实施例2化合物在本次药效实验所尝试剂量下未显著影响小鼠体重,也未引起任何小鼠死亡,小鼠可以耐受。In the Z-138 model of subcutaneous tumor transplantation in mice, the compound of Example 2 of the present disclosure had a significant inhibitory effect on tumor growth when administered twice a day at 1.5 mg/kg, 5 mg/kg and 15 mg/kg, and showed a better Dose-response relationship, administration of 15mg/kg twice a day has the effect of shrinking tumors. The compound of Example 2 did not significantly affect the body weight of the mice at the dose tried in this pharmacodynamic experiment, nor did it cause any death of the mice, and the mice could tolerate it.
测试试验例7:吸湿性研究Test Test Example 7: Hygroscopicity Study
(1)实验仪器:动态水吸附仪DVS Intrinsic;(1) Experimental instrument: dynamic water adsorption instrument DVS Intrinsic;
(2)实验条件:分别取样品(适量)置于DVS样品盘内进行测试。(2) Experimental conditions: take samples (appropriate amount) and place them in the DVS sample tray for testing.
(3)DVS参数:(3) DVS parameters:
温度:25℃;Temperature: 25°C;
平衡:dm/dt≤0.002%/minBalance: dm/dt≤0.002%/min
RH(%)测试梯级:10%RH(%) test rung: 10%
RH(%)测试梯级范围:0%-90%-0%。RH (%) test step range: 0%-90%-0%.
吸湿性评价标准(依据《中国药典》2015年版四部中“9103药物引湿性指导原则”中引湿性特征描述与引湿性增重的界定):Hygroscopicity evaluation standard (according to the description of hygroscopicity characteristics and the definition of hygroscopicity weight gain in "9103 Drug Hygroscopicity Guiding Principles" in "Chinese Pharmacopoeia" 2015 Edition IV):
吸湿性分类Hygroscopicity Classification 吸湿增重*(ΔW%)Moisture absorption weight gain*(ΔW%)
潮解deliquescence 吸收足量水分形成液体Absorb enough water to form a liquid
极具吸湿性Very hygroscopic 15%≤ΔW%15%≤ΔW%
有吸湿性Hygroscopic 2%≤ΔW%<15%2%≤ΔW%<15%
略有吸湿性slightly hygroscopic 0.2%≤ΔW%<2%0.2%≤ΔW%<2%
无或几乎无吸湿性No or almost no hygroscopicity ΔW%<0.2%ΔW%<0.2%
*在25±1℃和80±2%RH下的吸湿增重。*Weight gain on moisture absorption at 25±1°C and 80±2% RH.
(4)实验结果:(4) Experimental results:
实施例4样品(C晶型)在80.0%RH条件下,吸湿增重为2.072%,有吸湿性。The sample of Example 4 (Crystal Form C) has a moisture absorption weight gain of 2.072% under the condition of 80.0% RH, and has hygroscopicity.
实施例5样品(D晶型)在80.0%RH条件下,吸湿增重为14.470%,有吸湿性。The sample of Example 5 (crystal form D) has a moisture absorption weight gain of 14.470% under the condition of 80.0% RH, and has hygroscopicity.
实施例6样品(A晶型)在80.0%RH条件下,吸湿增重为2.318%,有吸湿性。The sample of Example 6 (crystal form A) has a hygroscopic weight gain of 2.318% under the condition of 80.0% RH, and has hygroscopicity.
测试试验例8:饱和溶解度测定Test Test Example 8: Saturation Solubility Determination
一、表观溶解度测定1. Determination of Apparent Solubility
试验仪器与试剂:pH计(型号:914PH;品牌:Metrohm)、电子天平(型号:XS205DU;品牌:METTLER TOLEDO)、恒温水浴振荡器(型号:DHSZ-300A;品牌:太仓文尔惠金);浓盐酸、磷酸二氢钾、磷酸、氢氧化钠、三水合醋酸钠、冰醋酸、80%甲醇水溶液。Test instruments and reagents: pH meter (model: 914PH; brand: Metrohm), electronic balance (model: XS205DU; brand: METTLER TOLEDO), constant temperature water bath oscillator (model: DHSZ-300A; brand: Taicang Wenerhuijin); Concentrated hydrochloric acid, potassium dihydrogen phosphate, phosphoric acid, sodium hydroxide, sodium acetate trihydrate, glacial acetic acid, 80% aqueous methanol.
试验操作:Test operation:
1.介质配制:1. Medium preparation:
pH1.0缓冲液:取浓盐酸9mL,加入纯化水稀释至1L,搅匀,测pH;pH1.0 buffer solution: take 9mL of concentrated hydrochloric acid, add purified water to dilute to 1L, stir well, and measure pH;
pH4.5缓冲液:称取三水合醋酸钠2.99g、冰醋酸1.68g,加入纯化水稀释至1L,搅匀,测pH;pH4.5 buffer solution: Weigh 2.99g of sodium acetate trihydrate and 1.68g of glacial acetic acid, add purified water to dilute to 1L, stir well, and measure pH;
pH6.8缓冲液:称取磷酸二氢钾6.805g、氢氧化钠0.896g,加入纯化水稀释至1L,搅匀,测pH。pH6.8 buffer solution: Weigh 6.805g of potassium dihydrogen phosphate and 0.896g of sodium hydroxide, add purified water to dilute to 1L, stir well, and measure pH.
2.表观溶解度测定:称取待测样品约50mg至50mL量瓶中,平行称取3份;分别向容量瓶中加入上述3种介质各10mL,观察溶液现象,在37℃恒温水浴振荡器上振摇,在2h、24h时观察溶液现象;取样3mL,离心取上清液,作为供试品溶液。称取待测样品,用80%甲醇水溶液溶解并稀释成每1mL中约含0.2mg的溶液,作为对照品溶液。采用高效液相色谱法,分别量取对照品溶液与供试品溶液各5μL,注入液相色谱仪,记录色谱图,按外标法以峰面积计算本品含量。2. Determination of apparent solubility: Weigh about 50mg of the sample to be tested into a 50mL measuring bottle, and weigh 3 parts in parallel; add 10mL of each of the above three media to the volumetric flask, observe the solution phenomenon, and place in a constant temperature water bath oscillator at 37°C Shake on the top, observe the phenomenon of the solution at 2h and 24h; sample 3mL, centrifuge to get the supernatant, as the test solution. Weigh the sample to be tested, dissolve it with 80% aqueous methanol and dilute it to a solution containing about 0.2 mg per 1 mL, as the reference solution. Using high-performance liquid chromatography, measure 5 μL each of the reference substance solution and the test solution, inject it into the liquid chromatograph, record the chromatogram, and calculate the content of this product by the peak area according to the external standard method.
二、生物介质中的饱和溶解度测定2. Determination of Saturation Solubility in Biological Media
试验仪器与试剂:高效液相色谱仪(型号:Agilent 1260;品牌:安捷伦)、恒温水浴振荡器(型号:DHSZ-300A;品牌:太仓文尔惠金)、pH计(型号:914PH;品牌Metrohm);氯化钠、氢氧化钠(部分配制成1mol/L氢氧化钠溶液待用)、浓盐酸(稀释成1mol/L盐酸待用)、醋酸钠、冰醋酸、无水磷酸二氢钠、纯牛奶、FaSSGF/FaSSIF粉末。Test instruments and reagents: high performance liquid chromatography (model: Agilent 1260; brand: Agilent), constant temperature water bath oscillator (model: DHSZ-300A; brand: Taicang Wenerhuijin), pH meter (model: 914PH; brand Metrohm ); sodium chloride, sodium hydroxide (partially prepared as 1mol/L sodium hydroxide solution for use), concentrated hydrochloric acid (diluted to 1mol/L hydrochloric acid for use), sodium acetate, glacial acetic acid, anhydrous sodium dihydrogen phosphate , Pure Milk, FaSSGF/FaSSIF Powder.
试验操作:Test operation:
1.介质配制:1. Medium preparation:
pH5.8介质:纯化水适量。pH5.8 medium: appropriate amount of purified water.
空腹胃液(FaSSGF)配制:称取氯化钠2.0g,加900mL水使溶解,用1mol/L盐酸溶液调节pH值至1.6,用水稀释至1000mL,即得缓冲液;称取FaSSGF粉末约15mg至250mL量瓶中,加125mL缓冲液搅拌使溶解,用缓冲液稀释至刻度,静置即得。Preparation of fasting gastric juice (FaSSGF): Weigh 2.0g of sodium chloride, add 900mL of water to dissolve, adjust the pH value to 1.6 with 1mol/L hydrochloric acid solution, and dilute to 1000mL with water to obtain a buffer solution; weigh about 15mg of FaSSGF powder to In a 250mL measuring bottle, add 125mL of buffer solution and stir to dissolve, dilute to the mark with buffer solution, and let it stand still.
餐后胃液(FeSSGF)配制:称取氯化钠13.85g,乙酸钠2.441g,冰醋酸1.028g,加1000mL水使溶解,用1mol/L盐酸溶液或1mol/L氢氧化钠溶液调节pH值至5.0,即得缓冲液;取纯牛奶100mL与缓冲液100mL,混匀,调节pH值至5.0,即得。Postprandial gastric juice (FeSSGF) preparation: Weigh 13.85g of sodium chloride, 2.441g of sodium acetate, 1.028g of glacial acetic acid, add 1000mL of water to dissolve, adjust the pH value to 1mol/L hydrochloric acid solution or 1mol/L sodium hydroxide solution 5.0 to obtain the buffer solution; take 100mL of pure milk and 100mL of the buffer solution, mix well, adjust the pH value to 5.0, and obtain the solution.
空腹肠液(FaSSIF)配制:称取氢氧化钠0.42g、氯化钠6.19g、无水磷酸二氢钠3.44g,加900mL水使溶解,用1mol/L盐酸溶液或1mol/L氢氧化钠溶液调节pH值至6.5,再用水稀释至1000mL,即得缓冲液;称取FaSSIF粉末约112mg至50mL量瓶中,加25mL缓冲 液搅拌使溶解,用缓冲液稀释至刻度,静置2小时即得。Fasting intestinal fluid (FaSSIF) preparation: Weigh 0.42g sodium hydroxide, 6.19g sodium chloride, 3.44g anhydrous sodium dihydrogen phosphate, add 900mL water to dissolve, and use 1mol/L hydrochloric acid solution or 1mol/L sodium hydroxide solution Adjust the pH value to 6.5, then dilute to 1000mL with water to obtain the buffer; weigh about 112mg of FaSSIF powder into a 50mL measuring bottle, add 25mL of buffer and stir to dissolve, dilute to the mark with buffer, and stand for 2 hours to obtain .
餐后肠液(FeSSIF)配制:称取氢氧化钠4.04g、氯化钠11.87g、冰醋酸8.65g,加900mL水使溶解,用1mol/L盐酸溶液或1mol/L氢氧化钠溶液调节pH值至5.0,再用水稀释至1000mL,即得缓冲液;称取FaSSIF粉末约560mg至50mL量瓶中,加25mL缓冲液搅拌使溶解,用缓冲液稀释至刻度,静置即得。Preparation of postprandial intestinal fluid (FeSSIF): Weigh 4.04g of sodium hydroxide, 11.87g of sodium chloride, and 8.65g of glacial acetic acid, add 900mL of water to dissolve, and adjust the pH value with 1mol/L hydrochloric acid solution or 1mol/L sodium hydroxide solution to 5.0, then diluted with water to 1000mL to obtain the buffer solution; weigh about 560mg of FaSSIF powder into a 50mL measuring bottle, add 25mL buffer solution and stir to dissolve, dilute to the mark with buffer solution, and stand still to obtain the solution.
2.饱和溶解度测定:称取供试品约70mg至50mL量瓶中,平行称取5份;分别向容量瓶中加入上述5种介质各10mL,观察溶液现象,在37℃恒温水浴振荡器上振摇,在2h、8h和24h时观察溶液现象;取样3mL,离心,取上清液,作为供试品溶液。称取待测样品,用80%甲醇水溶液溶解并稀释成每1mL中约含0.2mg的溶液,作为对照品溶液。采用高效液相色谱法,分别量取对照品溶液与供试品溶液各5μL,注入液相色谱仪,记录色谱图,按外标法以峰面积计算本品含量。2. Determination of saturation solubility: Weigh about 70mg of the test product into a 50mL measuring bottle, and weigh 5 parts in parallel; add 10mL of the above-mentioned 5 kinds of media to the volumetric flask respectively, observe the solution phenomenon, and place it on a constant temperature water bath shaker at 37°C Shake, and observe the solution phenomenon at 2h, 8h and 24h; sample 3mL, centrifuge, and take the supernatant as the test solution. Weigh the sample to be tested, dissolve it with 80% aqueous methanol and dilute it to a solution containing about 0.2 mg per 1 mL, as the reference solution. Using high-performance liquid chromatography, measure 5 μL each of the reference substance solution and the test solution, inject it into the liquid chromatograph, record the chromatogram, and calculate the content of this product by the peak area according to the external standard method.
以上表观溶解度测定和生物介质中的饱和溶解度测定结果如下表11所示。The results of the above apparent solubility measurements and the saturation solubility measurements in biological media are shown in Table 11 below.
表11 A晶型在不同pH缓冲液中的溶解度测定结果Table 11 Solubility determination results of crystal form A in different pH buffers
Figure PCTCN2022143995-appb-000054
Figure PCTCN2022143995-appb-000054
同理,按照上述表观溶解度测定的方法,对实施例3所得化合物I进行溶解度测定,测得其在纯化水中的溶解度小于0.5mg/mL。Similarly, according to the above-mentioned apparent solubility determination method, the solubility of Compound I obtained in Example 3 was measured, and its solubility in purified water was measured to be less than 0.5 mg/mL.
测试试验例9:影响因素试验Test Test Example 9: Influencing Factor Test
将化合物I以及化合物I的A晶型、C晶型和D晶型均采用双层聚乙烯袋加一层铝箔袋包装,考察在25℃、40℃、60℃以及袋内充氮和不充氮条件下的稳定性。Compound I and the A crystal form, C crystal form and D crystal form of compound I were packed in a double-layer polyethylene bag plus a layer of aluminum foil bag. Stability under nitrogen conditions.
有关物质HPLC分析方法:Related substances HPLC analysis method:
色谱柱:YMC-Pack Pro C18 150×4.6mm,3μmChromatographic column: YMC-Pack Pro C18 150×4.6mm, 3μm
柱温:35℃Column temperature: 35°C
流速:1.0ml/minFlow rate: 1.0ml/min
检测波长:240nmDetection wavelength: 240nm
进样体积:5μlInjection volume: 5μl
流动相:A:0.1%三氟乙酸水溶液;B:乙腈Mobile phase: A: 0.1% trifluoroacetic acid in water; B: acetonitrile
梯度:gradient:
时间(min)time (min) 流动相B(%)Mobile phase B(%) 流动相A(%)Mobile phase A(%)
00 1515 8585
1818 4040 6060
2828 8585 1515
3535 8585 1515
35.1035.10 1515 8585
4040 1515 8585
稀释剂:90%甲醇Diluent: 90% Methanol
结果如表12和表13所示。The results are shown in Table 12 and Table 13.
表12 40℃和60℃放置条件下的影响因素试验结果Table 12 Test results of influencing factors under storage conditions of 40°C and 60°C
Figure PCTCN2022143995-appb-000055
Figure PCTCN2022143995-appb-000055
注:“-”代表未测定。Note: "-" means not determined.
表13 25℃放置条件下的影响因素试验结果Table 13 Test results of influencing factors under storage conditions at 25°C
Figure PCTCN2022143995-appb-000056
Figure PCTCN2022143995-appb-000056
注:“-”代表未测定; a所述放置时间为1个月。 Note: "-" means undetermined; a The storage time is 1 month.
此外,双层聚乙烯袋加铝箔包装的化合物I在60℃、不充氮、3天放置条件下,其纯度即从0时的97.4%降至94.8%;相较而言,同样包装的A晶型在相同放置条件下,其纯度从0时的98.97%仅降至98.92%。因此,化合物I在成盐后明显热稳定性更优。In addition, the purity of Compound I packaged in double-layer polyethylene bags and aluminum foil dropped from 97.4% at 0 to 94.8% at 60°C without nitrogen filling for 3 days; Under the same storage conditions, the purity of the crystal form only decreased from 98.97% at 0 to 98.92%. Therefore, compound I has better thermal stability after salt formation.
同理,将化合物I的B晶型采用双层聚乙烯袋加铝箔包装,也考察了其在30℃、40℃以及袋内充氮和不充氮条件下的稳定性。结果如表14所示。Similarly, the crystal form B of compound I was packaged in a double-layer polyethylene bag plus aluminum foil, and its stability at 30°C, 40°C, and nitrogen-filled or non-nitrogen-filled conditions in the bag was also investigated. The results are shown in Table 14.
表14 B晶型的影响因素试验结果Table 14 B crystal form influence factor test result
Figure PCTCN2022143995-appb-000057
Figure PCTCN2022143995-appb-000057
通过表14可以看出,B晶型在30℃和40℃、不充氮条件下杂质含量明显增大,相较以上A晶型、C晶型和D晶型在40℃条件下放置的结果,表明在该放置条件下,B晶型的化学稳定性稍差。It can be seen from Table 14 that the impurity content of Form B increases significantly at 30°C and 40°C without nitrogen filling, compared with the results of Form A, Form C and Form D placed at 40°C , indicating that under this storage condition, the chemical stability of Form B is slightly worse.
以上,对本公开内容的实施方式进行了说明。但是,本公开不限定于上述实施方式。凡在本公开的精神和原则之内,所做的任何修改、等同替换、改进等,均应包含在本公开的保护范围之内。The embodiments of the present disclosure have been described above. However, the present disclosure is not limited to the above-mentioned embodiments. Any modifications, equivalent replacements, improvements, etc. made within the spirit and principles of the present disclosure shall be included within the protection scope of the present disclosure.

Claims (21)

  1. 化合物I的可药用盐,其中所述可药用盐选自一盐酸盐、氢溴酸盐、1,5-萘二磺酸盐、草酸盐、柠檬酸盐、硫酸盐、磷酸盐、L-酒石酸盐、L-苹果酸盐、琥珀酸盐、己二酸盐、富马酸盐、草酸盐、丙酸盐、苯甲酸盐、乙酸盐、甲酸盐或L-精氨酸盐,优选一盐酸盐、氢溴酸盐或1,5-萘二磺酸盐,更优选一盐酸盐A pharmaceutically acceptable salt of compound I, wherein the pharmaceutically acceptable salt is selected from monohydrochloride, hydrobromide, 1,5-naphthalene disulfonate, oxalate, citrate, sulfate, phosphate , L-tartrate, L-malate, succinate, adipate, fumarate, oxalate, propionate, benzoate, acetate, formate or L-arginate Amino acid salt, preferably monohydrochloride, hydrobromide or 1,5-naphthalene disulfonate, more preferably monohydrochloride
    Figure PCTCN2022143995-appb-100001
    Figure PCTCN2022143995-appb-100001
  2. 根据权利要求1所述的化合物I的可药用盐,其中所述化合物与酸分子的摩尔比约为1:2~2:1,优选选自1:2、1:1或2:1,更优选为1:1。The pharmaceutically acceptable salt of compound I according to claim 1, wherein the molar ratio of the compound to the acid molecule is about 1:2 to 2:1, preferably selected from 1:2, 1:1 or 2:1, More preferably 1:1.
  3. 根据权利要求1或2所述的化合物I的可药用盐,其中所述可药用盐选自化合物I的一盐酸盐、化合物I的氢溴酸盐或化合物I的1,5-萘二磺酸盐。The pharmaceutically acceptable salt of compound I according to claim 1 or 2, wherein said pharmaceutically acceptable salt is selected from the monohydrochloride of compound I, the hydrobromide of compound I or the 1,5-naphthalene of compound I Disulfonate.
  4. 根据权利要求3所述的化合物I的可药用盐,其中所述可药用盐为无定型或结晶形式,优选为结晶形式。The pharmaceutically acceptable salt of compound I according to claim 3, wherein said pharmaceutically acceptable salt is in amorphous or crystalline form, preferably in crystalline form.
  5. 制备如权利要求1或2所述的化合物I的可药用盐的方法,包括:化合物I与酸成盐的步骤,所述酸选自盐酸、氢溴酸、1,5-萘二磺酸、草酸、柠檬酸、硫酸、磷酸、L-酒石酸、L-苹果酸、琥珀酸、己二酸、富马酸、草酸、丙酸、苯甲酸、乙酸、甲酸或L-精氨酸,优选盐酸、氢溴酸或1,5-萘二磺酸,更优选盐酸;所述成盐反应所用溶剂选自乙酸乙酯、四氢呋喃、乙腈、丙酮、乙酸甲酯、异丙醇、甲醇或甲苯中的至少一种,优选选自异丙醇、乙酸甲酯、乙酸乙酯、甲醇或丙酮,更优选异丙醇或乙酸乙酯。The method for preparing the pharmaceutically acceptable salt of compound I as claimed in claim 1 or 2, comprises: the step of compound I and acid salt formation, and described acid is selected from hydrochloric acid, hydrobromic acid, 1,5-naphthalene disulfonic acid , oxalic acid, citric acid, sulfuric acid, phosphoric acid, L-tartaric acid, L-malic acid, succinic acid, adipic acid, fumaric acid, oxalic acid, propionic acid, benzoic acid, acetic acid, formic acid or L-arginine, preferably hydrochloric acid , hydrobromic acid or 1,5-naphthalene disulfonic acid, more preferably hydrochloric acid; The solvent used in the salt-forming reaction is selected from ethyl acetate, tetrahydrofuran, acetonitrile, acetone, methyl acetate, isopropanol, methanol or toluene At least one, preferably selected from isopropanol, methyl acetate, ethyl acetate, methanol or acetone, more preferably isopropanol or ethyl acetate.
  6. 根据权利要求4所述的化合物I的可药用盐,其中所述可药用盐为化合物I一盐酸盐的A晶型,所述A晶型以衍射角2θ表示的X-射线粉末衍射图谱,在5.42±0.2°、6.60±0.2°、10.36±0.2°、13.60±0.2°、14.24±0.2°、15.83±0.2°、16.62±0.2°、17.65±0.2°、19.29±0.2°、19.86±0.2°、21.37±0.2°、23.88±0.2°处有衍射峰。The pharmaceutically acceptable salt of compound I according to claim 4, wherein the pharmaceutically acceptable salt is the A crystal form of compound I hydrochloride, and the X-ray powder diffraction of the A crystal form represented by diffraction angle 2θ Spectrum at 5.42±0.2°, 6.60±0.2°, 10.36±0.2°, 13.60±0.2°, 14.24±0.2°, 15.83±0.2°, 16.62±0.2°, 17.65±0.2°, 19.29±0.2°, 19.86± There are diffraction peaks at 0.2°, 21.37±0.2°, and 23.88±0.2°.
  7. 根据权利要求6所述的化合物I的可药用盐,其中所述A晶型以衍射角2θ表示的X-射线粉末衍射图谱,在5.42±0.2°、6.60±0.2°、9.76±0.2°、10.36±0.2°、13.00±0.2°、13.60±0.2°、14.24±0.2°、15.83±0.2°、16.62±0.2°、17.65±0.2°、18.28±0.2°、19.29±0.2°、19.86±0.2°、20.61±0.2°、21.37±0.2°、23.88±0.2°、25.08±0.2°、28.22±0.2°处有衍射峰。The pharmaceutically acceptable salt of Compound I according to claim 6, wherein the X-ray powder diffraction pattern represented by the diffraction angle 2θ of the A crystal form is at 5.42±0.2°, 6.60±0.2°, 9.76±0.2°, 10.36±0.2°, 13.00±0.2°, 13.60±0.2°, 14.24±0.2°, 15.83±0.2°, 16.62±0.2°, 17.65±0.2°, 18.28±0.2°, 19.29±0.2°, 19.86±0.2°, There are diffraction peaks at 20.61±0.2°, 21.37±0.2°, 23.88±0.2°, 25.08±0.2°, 28.22±0.2°.
  8. 根据权利要求6或7所述的化合物I的可药用盐,其中所述A晶型的X-射线粉末衍射图基本上如图8所示。The pharmaceutically acceptable salt of compound I according to claim 6 or 7, wherein the X-ray powder diffraction pattern of the crystal form A is substantially as shown in FIG. 8 .
  9. 根据权利要求6-8任一项所述的化合物I的可药用盐,其中所述A晶型具有在259.27±5.0℃下的峰值的DSC谱图。The pharmaceutically acceptable salt of Compound I according to any one of claims 6-8, wherein the crystal form A has a DSC spectrum with a peak at 259.27±5.0°C.
  10. 制备如权利要求6-9任一项所述的化合物I的可药用盐的方法,包括如下步骤:The method for preparing the pharmaceutically acceptable salt of compound I as described in any one of claims 6-9, comprises the steps:
    1)称取适量化合物I,加入溶剂(i)中,室温溶清或加热溶清;1) Weighing an appropriate amount of compound I, adding it to the solvent (i), and dissolving at room temperature or heating;
    2)加入氯化氢水溶液或氯化氢的有机溶剂溶液,析晶。2) Add aqueous hydrogen chloride solution or organic solvent solution of hydrogen chloride to crystallize.
  11. 根据权利要求10所述的制备方法,其中所述溶剂(i)选自乙酸甲酯、乙酸乙酯、丙酮或异丙醇,优选异丙醇或乙酸乙酯。The preparation method according to claim 10, wherein the solvent (i) is selected from methyl acetate, ethyl acetate, acetone or isopropanol, preferably isopropanol or ethyl acetate.
  12. 根据权利要求10或11所述的制备方法,其中所述步骤2)之后还存在可选的步骤3),即采用良溶剂对步骤2)所得晶体进行溶解,得到溶液后再滴加不良溶剂,析晶。The preparation method according to claim 10 or 11, wherein there is an optional step 3) after the step 2), that is, using a good solvent to dissolve the crystal obtained in step 2), and then adding the poor solvent dropwise after obtaining the solution, Crystallization.
  13. 根据权利要求10或11所述的制备方法,其中所述步骤2)之后还存在可选的步骤3’), 即将步骤2)所得晶型溶于甲醇后减压浓缩得到化合物I一盐酸盐无定型物,然后将该无定型物溶于溶剂(ii),继而通过打浆或反溶剂滴加的方式析出晶体。The preparation method according to claim 10 or 11, wherein there is an optional step 3') after the step 2), that is, dissolving the crystal form obtained in step 2) in methanol and concentrating under reduced pressure to obtain Compound I-hydrochloride Amorphous substance, and then dissolve the amorphous substance in solvent (ii), and then precipitate crystals by beating or adding anti-solvent dropwise.
  14. 根据权利要求4所述的化合物I的可药用盐,其中所述可药用盐为化合物I一盐酸盐的B晶型,所述B晶型以衍射角2θ表示的X-射线粉末衍射图谱,在5.16±0.2°、9.86±0.2°、10.42±0.2°、10.76±0.2°、11.36±0.2°、15.68±0.2°、16.31±0.2°、17.29±0.2°、17.98±0.2°、18.89±0.2°、19.83±0.2°、20.99±0.2°处有衍射峰。The pharmaceutically acceptable salt of compound I according to claim 4, wherein the pharmaceutically acceptable salt is the B crystal form of compound I-hydrochloride, and the X-ray powder diffraction of the B crystal form represented by diffraction angle 2θ Spectrum, at 5.16±0.2°, 9.86±0.2°, 10.42±0.2°, 10.76±0.2°, 11.36±0.2°, 15.68±0.2°, 16.31±0.2°, 17.29±0.2°, 17.98±0.2°, 18.89± There are diffraction peaks at 0.2°, 19.83±0.2°, and 20.99±0.2°.
  15. 根据权利要求14所述的化合物I的可药用盐,其中所述B晶型的X-射线粉末衍射图基本上如图11所示。The pharmaceutically acceptable salt of compound I according to claim 14, wherein the X-ray powder diffraction pattern of the crystal form B is substantially as shown in FIG. 11 .
  16. 根据权利要求4所述的化合物I的可药用盐,其中所述可药用盐为化合物I氢溴酸盐的C晶型,所述C晶型以衍射角2θ表示的X-射线粉末衍射图谱,在6.45±0.2°、9.49±0.2°、10.01±0.2°、12.96±0.2°、13.45±0.2°、13.81±0.2°、15.50±0.2°、19.13±0.2°、20.13±0.2°、20.78±0.2°、23.31±0.2°处有衍射峰。The pharmaceutically acceptable salt of compound I according to claim 4, wherein the pharmaceutically acceptable salt is the C crystal form of compound I hydrobromide, the X-ray powder diffraction of the C crystal form represented by diffraction angle 2θ Spectrum at 6.45±0.2°, 9.49±0.2°, 10.01±0.2°, 12.96±0.2°, 13.45±0.2°, 13.81±0.2°, 15.50±0.2°, 19.13±0.2°, 20.13±0.2°, 20.78± There are diffraction peaks at 0.2° and 23.31±0.2°.
  17. 根据权利要求16所述的化合物I的可药用盐,其中所述C晶型的X-射线粉末衍射图谱基本上如图2所示。The pharmaceutically acceptable salt of compound I according to claim 16, wherein the X-ray powder diffraction pattern of the crystal form C is substantially as shown in FIG. 2 .
  18. 根据权利要求4所述的化合物I的可药用盐,其中所述可药用盐为化合物I的1,5-萘二磺酸盐的D晶型,所述D晶型以衍射角2θ表示的X-射线粉末衍射图谱,在4.02±0.2°、9.89±0.2°、11.49±0.2°、12.74±0.2°、15.68±0.2°、15.69±0.2°、16.15±0.2°、18.62±0.2°、18.93±0.2°、19.67±0.2°、19.79±0.2°、21.26±0.2°、24.36±0.2°、27.56±0.2°处有衍射峰。The pharmaceutically acceptable salt of Compound I according to claim 4, wherein the pharmaceutically acceptable salt is the D crystal form of 1,5-naphthalene disulfonate of Compound I, and the D crystal form is represented by diffraction angle 2θ X-ray powder diffraction pattern at 4.02±0.2°, 9.89±0.2°, 11.49±0.2°, 12.74±0.2°, 15.68±0.2°, 15.69±0.2°, 16.15±0.2°, 18.62±0.2°, 18.93 There are diffraction peaks at ±0.2°, 19.67±0.2°, 19.79±0.2°, 21.26±0.2°, 24.36±0.2°, 27.56±0.2°.
  19. 根据权利要求18所述的化合物I的可药用盐,其中所述D晶型的X-射线粉末衍射图基本上如图5所示。The pharmaceutically acceptable salt of compound I according to claim 18, wherein the X-ray powder diffraction pattern of the D crystal form is substantially as shown in FIG. 5 .
  20. 一种药物组合物,含有根据权利要求1-4、6-9、14-19中任一项所述的化合物I的可药用盐,以及药学上可接受的辅料。A pharmaceutical composition, containing the pharmaceutically acceptable salt of compound I according to any one of claims 1-4, 6-9, 14-19, and pharmaceutically acceptable auxiliary materials.
  21. 权利要求1-4、6-9、14-19中任一项所述的化合物I的可药用盐或权利要求20所述的药物组合物在制备预防或者治疗由PRMT5介导的疾病或病症的药物中的用途;优选地,所述由PRMT5介导的疾病或病症为癌症。The pharmaceutically acceptable salt of compound I described in any one of claims 1-4, 6-9, 14-19 or the pharmaceutical composition described in claim 20 are used in the preparation of prevention or treatment of diseases or diseases mediated by PRMT5 Use in medicine; Preferably, the disease or disease mediated by PRMT5 is cancer.
PCT/CN2022/143995 2021-12-30 2022-12-30 Pharmaceutically acceptable salt of tetrahydroisoquinoline compound, and crystal form and use thereof WO2023125947A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111664582.X 2021-12-30
CN202111664582 2021-12-30

Publications (1)

Publication Number Publication Date
WO2023125947A1 true WO2023125947A1 (en) 2023-07-06

Family

ID=86998124

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/143995 WO2023125947A1 (en) 2021-12-30 2022-12-30 Pharmaceutically acceptable salt of tetrahydroisoquinoline compound, and crystal form and use thereof

Country Status (1)

Country Link
WO (1) WO2023125947A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016034673A1 (en) * 2014-09-03 2016-03-10 Ctxt Pty Ltd Tetrahydroisoquinoline derived prmt5-inhibitors
WO2017153520A1 (en) * 2016-03-09 2017-09-14 Ctxt Pty Limited Benzopiperidine derivatives and their use in the treatment of cancer and hemoglobinopathies
WO2019173804A1 (en) * 2018-03-09 2019-09-12 Pharmablock Sciences (Nanjing), Inc. Inhibitors of protein arginine methyltransferase 5 (prmt5), pharmaceutical products thereof, and methods thereof
CN110325188A (en) * 2016-10-26 2019-10-11 印第安纳大学理事会 (PRMT5) inhibitor of small protein arginine methyltransferase 5 and treatment method
WO2019219805A1 (en) * 2018-05-16 2019-11-21 Ctxone Pty Ltd Combination therapy
CN110650950A (en) * 2017-03-17 2020-01-03 阿古诺治疗有限公司 Compounds for treating or preventing PRMT5 mediated diseases
WO2022002142A1 (en) * 2020-06-30 2022-01-06 江苏先声药业有限公司 Tetrahydroisoquinoline compounds and use thereof

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016034673A1 (en) * 2014-09-03 2016-03-10 Ctxt Pty Ltd Tetrahydroisoquinoline derived prmt5-inhibitors
US20170298075A1 (en) * 2014-09-03 2017-10-19 Ctxt Pty Ltd Tetrahydroisoquinoline derived prmt5-inhibitors
WO2017153520A1 (en) * 2016-03-09 2017-09-14 Ctxt Pty Limited Benzopiperidine derivatives and their use in the treatment of cancer and hemoglobinopathies
CN110325188A (en) * 2016-10-26 2019-10-11 印第安纳大学理事会 (PRMT5) inhibitor of small protein arginine methyltransferase 5 and treatment method
CN110650950A (en) * 2017-03-17 2020-01-03 阿古诺治疗有限公司 Compounds for treating or preventing PRMT5 mediated diseases
WO2019173804A1 (en) * 2018-03-09 2019-09-12 Pharmablock Sciences (Nanjing), Inc. Inhibitors of protein arginine methyltransferase 5 (prmt5), pharmaceutical products thereof, and methods thereof
WO2019219805A1 (en) * 2018-05-16 2019-11-21 Ctxone Pty Ltd Combination therapy
WO2022002142A1 (en) * 2020-06-30 2022-01-06 江苏先声药业有限公司 Tetrahydroisoquinoline compounds and use thereof

Similar Documents

Publication Publication Date Title
EP2850082B1 (en) 1-(3,3-dimethylbutyl)-3-(2-fluoro-4-methyl-5-(7-methyl-2-(methylamino)pyrido(2,3-d)pyrimidin-6-yl)phenyl)urea as Raf kinase inhibitor for the treatment of cancer
AU2023208222A1 (en) Methods of treatment for cystic fibrosis
WO2021190467A1 (en) Spiro ring-containing quinazoline compound
CN107922425B (en) Methods of preparing PARP inhibitors, crystalline forms and uses thereof
CN109970745B (en) Substituted pyrrolotriazine compound, pharmaceutical composition and application thereof
CN109689641B (en) Crystal form and salt form of substituted 2-hydrogen-pyrazole derivative and preparation method thereof
CN115433187B (en) Crystalline form composition of salt of fused tetracyclic or pentacyclic dihydro-diazepin carbazolone and use thereof
WO2022002142A1 (en) Tetrahydroisoquinoline compounds and use thereof
WO2021143843A1 (en) Crystal of pde3/pde4 dual inhibitor and use thereof
KR20090092338A (en) Succinate salt of 2-((4-(1-methyl-4-(pyridin-4-yl)-1h-pyrazol-3-yl)phenoxy)methyl)quinoline
WO2023125947A1 (en) Pharmaceutically acceptable salt of tetrahydroisoquinoline compound, and crystal form and use thereof
WO2022068860A1 (en) Crystal form of pyrrolo heterocyclic derivative and preparation method therefor
WO2022022569A1 (en) Solid form of nitrogen-containing heterocyclic compound, and pharmaceutical composition and use thereof
CN114315837A (en) Crystalline form of ERK inhibitor and preparation method thereof
CN115427397A (en) Crystal form of nitroxoline prodrug, pharmaceutical composition containing nitroxoline prodrug, and preparation method and application of nitroxoline prodrug
WO2023131305A1 (en) Combination of prmt5 inhibitor and anti-cancer therapeutic agent
WO2019057142A1 (en) Crystalline sulfamide compound
CN114644616B (en) Pharmaceutically acceptable salt and crystal form of indazole derivative and preparation method thereof
WO2023104107A1 (en) Salt of 3,4-dihydroisoquinoline compound and use thereof
CN112334473B (en) Crystal form of heteroaryl [4,3-c ] pyrimidine-5-amine derivative and preparation method thereof
WO2022127904A1 (en) Pharmaceutically acceptable salt of indazole derivative, and crystalline form and preparation method therefor
WO2022171117A1 (en) Salt of nitrogen-containing fused heterocyclic compound or crystal form thereof, and preparation method therefor, pharmaceutical composition thereof, and use thereof
WO2023093861A1 (en) Mono-p-toluenesulfonate of axl kinase inhibitor and crystal form thereof
WO2021088992A1 (en) Tetrahydroisoquinoline spiro compound as prmt5 inhibitor
CN115232124A (en) Crystal form of ATX inhibitor and preparation method thereof

Legal Events

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

Ref document number: 22915195

Country of ref document: EP

Kind code of ref document: A1