WO2024056016A1 - 含氮杂环的多环化合物的自由碱晶型及其制备方法 - Google Patents

含氮杂环的多环化合物的自由碱晶型及其制备方法 Download PDF

Info

Publication number
WO2024056016A1
WO2024056016A1 PCT/CN2023/118743 CN2023118743W WO2024056016A1 WO 2024056016 A1 WO2024056016 A1 WO 2024056016A1 CN 2023118743 W CN2023118743 W CN 2023118743W WO 2024056016 A1 WO2024056016 A1 WO 2024056016A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
diffraction peak
alkyl
crystal form
diffraction
Prior art date
Application number
PCT/CN2023/118743
Other languages
English (en)
French (fr)
Inventor
杨彤
陈金瑶
Original Assignee
江苏豪森药业集团有限公司
上海翰森生物医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 江苏豪森药业集团有限公司, 上海翰森生物医药科技有限公司 filed Critical 江苏豪森药业集团有限公司
Publication of WO2024056016A1 publication Critical patent/WO2024056016A1/zh

Links

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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the invention belongs to the field of drug synthesis, and specifically relates to the crystal form of a nitrogen-containing heterocyclic polycyclic compound and its preparation method and application.
  • Orexin is a neuropeptide synthesized and secreted by orexin neurons in the lateral hypothalamus (LH). It is named for its strong appetite-promoting effect. Orexin is divided into They are Orexin A (Orexin A) and Orexin B (Orexin B). Orexin A and Orexin B both act on the G protein-coupled receptor orexin receptor OX1R and the orexin receptor OX2R. OX1R and OX2R act on the entire central nervous system. The system is widely expressed, among which OX1R has stronger binding ability to orexin A than orexin B, while OX2R has equivalent binding ability to orexin A and orexin B.
  • Orexin receptor antagonists have potential therapeutic advantages in the treatment of neurological diseases, including insomnia, depression, anxiety, and drug addiction.
  • OX1R and OX2R activate intracellular Ca 2+ through phospholipase C.
  • OX2R can also couple with Gi/Go and inhibit the production of cAMP by inhibiting adenylyl cyclase.
  • Nocturnal hypothalamic-pituitary-adrenal (HPA) hyperexcitability is the biggest difference between patients with depression and normal people. Excessive effects on the HPA system Downregulation of excitement will help improve depressive symptoms.
  • HPA hypothalamic-pituitary-adrenal
  • OX2R antagonists can have an antidepressant effect, and OX2R single receptor antagonists can also have sufficient effects on insomnia. Therefore, selective OX2R antagonists can avoid various side effects such as drowsiness caused by the effect on OX1R.
  • OX2R antagonist Seltorexant developed by Janssen is in the clinical stage, and its main indications are major depressive disorder (MDD), primary and secondary insomnia, etc.
  • Selective OX2R antagonists have the potential to treat neurological diseases such as insomnia, depression, and anxiety, and have huge clinical demand. Selective OX2R antagonists have good application prospects in the pharmaceutical industry as drugs scene.
  • PCT/CN2022/080829 discloses the structure of a series of nitrogen-containing heterocyclic polycyclic compounds.
  • PCT/CN2022/080829 discloses the structure of a series of nitrogen-containing heterocyclic polycyclic compounds.
  • the present invention conducted a comprehensive study on the free base crystal forms of the above compounds.
  • the object of the present invention is to provide a crystal form of the compound represented by general formula (I), the structure of which is as shown in formula (I):
  • X 1 is CR 6 or N
  • X 2 is CR 6 or does not exist
  • R 1 is selected from halogen
  • R 2 , R 3 , R 4 or R 6 are each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxyl, mercapto, cyano, carboxyl, sulfonate, oxo, thio, alkyl , deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, the Amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl , optionally may be further substituted;
  • R 2 , R 3 , R 4 or R 6 are each independently preferably hydrogen, deuterium, halogen, amino, nitro, hydroxyl, mercapto, cyano, carboxyl, sulfonic acid group, oxo group, thio group, C 1- 8 alkyl, C 1-8 deuterated alkyl, C 1-8 haloalkyl, C 1-8 hydroxyalkyl, C 1-8 alkoxy, C 1-8 haloalkoxy, C 1-8 hydroxyalkyl Oxygen group, C 2-8 alkenyl group, C 2-8 alkynyl group, C 3-12 cycloalkyl group, 3-12 membered heterocyclic group, C 6-14 aryl group or 5-14 membered heteroaryl group, the Amino, C 1-8 alkyl, C 1-8 deuterated alkyl, C 1-8 haloalkyl, C 1-8 hydroxyalkyl, C 1-8 alkoxy, C 1-8 haloalkoxy, C 1-8
  • any two or more of R 2 , R 3 , and R 4 are connected to the connected atom to form a cycloalkyl group, a heterocyclyl group, an aryl group, or a heteroaryl group, and the cycloalkyl group, heterocyclyl group, aryl group, or heteroaryl group is radicals and heteroaryl groups, optionally may be further substituted;
  • R 5 is selected from hydroxyalkyl or haloalkyl, and optionally can be further substituted.
  • the hydroxyalkyl is preferably
  • the haloalkyl group is preferably
  • X’ is halogen
  • R 7 are independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxyl, mercapto, cyano, carboxyl, sulfonate, oxo, thio, alkyl, deuterated alkyl, haloalkyl, Hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, the amino, alkyl, deuterated alkyl , haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, hydroxyalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, optionally may be further substituted; preferably Hydrogen, deuterium, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy,
  • a preferred embodiment of the present invention is to provide a crystal form of a compound represented by general formula (II),
  • X 1 is CH or N
  • X 2 is CH or does not exist
  • R 2 , R 3 or R 4 are each independently selected from hydrogen, deuterium, halogen, amino, hydroxyl, C 1-3 alkyl, C 1-3 deuterated alkyl, C 1-3 haloalkyl, C 1-3 Hydroxyalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy or C 1-3 hydroxyalkoxy;
  • R 5 is selected from
  • the substituent R 2 is preferably halogen, and the halogen is preferably located at the ortho or meta position of the triazole, more preferably at the ortho position,
  • the halogen is preferably fluorine
  • the halogen is preferably fluorine.
  • a preferred embodiment of the present invention is that the general formula (I) is selected from the following compounds:
  • the powder X-ray diffraction pattern of the crystalline form I has a diffraction peak at 2 ⁇ of 8.6 ⁇ 0.2°, or a diffraction peak at 2 ⁇ of 9.2 ⁇ 0.2°, or a diffraction peak at 2 ⁇ of 10.2 ⁇ 0.2°, Either have a diffraction peak at 2 ⁇ of 10.8 ⁇ 0.2°, or have a diffraction peak at 2 ⁇ of 12.0 ⁇ 0.2°, or have a diffraction peak at 2 ⁇ of 12.8 ⁇ 0.2°, or have a diffraction peak at 2 ⁇ of 13.7 ⁇ 0.2° Peak, either having a diffraction peak at 14.2 ⁇ 0.2° 2 ⁇ , or having a diffraction peak at 15.5 ⁇ 0.2° 2 ⁇ , or having a diffraction peak at 16.6 ⁇ 0.2° 2 ⁇ , or having a diffraction peak at 17.3 ⁇ 0.2° 2 ⁇ Has a diffraction peak, either at 18.4 ⁇ 0.2° 2 ⁇ , or at 19.0 ⁇ 0.2° 2 ⁇ , or at 19.5 ⁇
  • its powder X-ray diffraction pattern contains at least one or more diffraction peaks located at 2 ⁇ of 9.2 ⁇ 0.2°, 10.2 ⁇ 0.2°, 16.6 ⁇ 0.2° or 18.4 ⁇ 0.2°; preferably, it includes 2-4 of them. , more preferably 3-4 locations, most preferably 4 locations; optionally, further, 2 ⁇ may be 12.8 ⁇ 0.2°, 17.3 ⁇ 0.2°, 19.0 ⁇ 0.2°, 21.3 ⁇ 0.2° or 26.5 ⁇ 0.2°
  • One or more diffraction peaks in preferably including 2, 3, 4 or 5 of them; for example:
  • the powder X-ray diffraction pattern optionally further includes a position located at 2 ⁇ of 8.6 ⁇ 0.2°, 10.8 ⁇ 0.2°, 12.0 ⁇ 0.2°, 13.7 ⁇ 0.2°, 14.2 ⁇ 0.2° or 28.6 ⁇ 0.2°. or multiple diffraction peaks; preferably at least any 2-4 of them, or 5-6 of them, and more preferably, any 4 or 6 of them; for example:
  • the X-ray powder diffraction pattern of the crystalline form I has characteristic peaks at 2 ⁇ of 9.2 ⁇ 0.2° and 18.4 ⁇ 0.2°; preferably, it also includes 2 ⁇ of 10.2 ⁇ 0.2°, 16.6 ⁇ 0.2°, and 17.3 ⁇ 0.2 It has characteristic peaks at 12.8 ⁇ 0.2° and 21.3 ⁇ 0.2° in 2 ⁇ . It has characteristic peaks; further preferably, it also has characteristic peaks at 2 ⁇ , 26.5 ⁇ 0.2° and 28.6 ⁇ 0.2°, and further preferably, it also has characteristic peaks at 8.6 ⁇ 0.2°, 10.8 ⁇ 0.2°, and 12.0 ⁇ 0.2°. There are characteristic peaks at , 13.7 ⁇ 0.2°, 14.2 ⁇ 0.2°, 15.5 ⁇ 0.2°, 19.5 ⁇ 0.2°, 20.5 ⁇ 0.2°, 23.1 ⁇ 0.2°, 27.8 ⁇ 0.2° and 32.1 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the crystal form I of the compound 9 of the present invention is basically as shown in Figure 1
  • its DSC pattern is basically as shown in Figure 2
  • its TGA pattern is basically as shown in Figure 3.
  • the crystal form I of compound 9 according to the present invention is an anhydrate crystal form.
  • the powder X-ray diffraction pattern of the crystalline form II has a diffraction peak at 2 ⁇ of 9.9 ⁇ 0.2°, or a diffraction peak at 2 ⁇ of 10.8 ⁇ 0.2°, or a diffraction peak at 2 ⁇ of 13.2 ⁇ 0.2°, Either have a diffraction peak at 2 ⁇ of 14.9 ⁇ 0.2°, or have a diffraction peak at 2 ⁇ of 16.4 ⁇ 0.2°, or have a diffraction peak at 2 ⁇ of 17.1 ⁇ 0.2°, or have a diffraction peak at 2 ⁇ of 17.9 ⁇ 0.2° Peak, either having a diffraction peak at 2 ⁇ of 19.3 ⁇ 0.2°, or having a diffraction peak at 2 ⁇ of 19.8 ⁇ 0.2°, or having a diffraction peak at 2 ⁇ of 20.2 ⁇ 0.2°, or having a diffraction peak at 2 ⁇ of 20.5 ⁇ 0.2° Having a diffraction peak either at 21.0 ⁇ 0.2° 2 ⁇ , or having a diffraction peak at 2
  • its powder X-ray diffraction pattern contains at least one or more diffraction peaks located at 2 ⁇ of 16.4 ⁇ 0.2°, 17.1 ⁇ 0.2°, 17.9 ⁇ 0.2° or 25.8 ⁇ 0.2°; preferably 2-4 of them are included. , more preferably 3-4 locations, most preferably 4 locations; optionally, further, 2 ⁇ may be 10.8 ⁇ 0.2°, 14.9 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.9 ⁇ 0.2° or 26.5 ⁇ 0.2°
  • One or more diffraction peaks in preferably including 2, 3, 4 or 5 of them; for example:
  • the powder X-ray diffraction pattern optionally further includes a position located at 2 ⁇ of 9.9 ⁇ 0.2°, 13.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.2 ⁇ 0.2°, 20.5 ⁇ 0.2° or 21.0 ⁇ 0.2°. or multiple diffraction peaks; preferably at least any 2-4 of them, or 5-6 of them, and more preferably, any 4 or 6 of them; for example:
  • the X-ray powder diffraction pattern of the crystalline form II has characteristic peaks at 2 ⁇ of 16.4 ⁇ 0.2° and 25.8 ⁇ 0.2°; preferably, it also includes 2 ⁇ of 17.1 ⁇ 0.2 °, 17.9 ⁇ 0.2°, 21.9 ⁇ 0.2° and 26.5 ⁇ 0.2°; more preferably, it also has characteristic peaks at 2 ⁇ of 10.8 ⁇ 0.2° and 14.9 ⁇ 0.2°; further preferably, it also includes It has characteristic peaks at 2 ⁇ of 9.9 ⁇ 0.2° and 19.3 ⁇ 0.2°, and more preferably, it also includes peaks at 13.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.2 ⁇ 0.2°, 20.5 ⁇ 0.2°, 21.0 ⁇ 0.2° and There is a characteristic peak at 27.5 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the crystal form II of the compound 9 of the present invention is basically as shown in Figure 4, Its DSC spectrum is basically as shown in Figure 5, and its TGA spectrum is basically as shown in Figure 6.
  • the present invention also provides a method for preparing the crystal form of the compound represented by general formula (I), which specifically includes the following steps:
  • the poor solvent is selected from the group consisting of acetone, ethyl acetate, isopropyl acetate, acetonitrile, ethanol, tetrahydrofuran, 2-methyltetrahydrofuran, dichloromethane, 1,4-dioxane, benzene, toluene, and isopropyl alcohol. , n-butanol, isobutanol, N,N-dimethylformamide, N,N-dimethylacetamide, n-propanol, tert-butanol, 2-butanone or 3-pentanone, methyl tert.
  • the suspension density in step 1) is 50-200 mg/mL.
  • the temperature in step 2) is 0 to 50°C.
  • the shaking time in step 2) is selected from 1 to 10 days.
  • the invention also provides a method for preparing the crystal form of the compound represented by general formula (I), which specifically includes the following steps:
  • the good solvent is selected from methanol, acetone, ethyl acetate, acetonitrile, ethanol, tetrahydrofuran, dichloromethane, 1,4-dioxane, benzene, toluene, isopropanol, n-butanol, isobutanol, N,N-dimethylformamide, N,N-dimethylacetamide, n-propanol, tert-butanol, 2-butanone or 3-pentanone; preferably methanol, dichloromethane, and ethyl acetate.
  • the anti-solvent is selected from heptane, water, methyl tert-butyl ether, cyclohexane, and isopropyl acetate.
  • the temperature in step 2) is 0 to 25°C.
  • the invention also provides a method for preparing the crystal form of the compound represented by general formula (I), which specifically includes the following steps:
  • the good solvent is selected from methanol, acetone, ethyl acetate, acetonitrile, ethanol, tetrahydrofuran, dichloromethane, 1,4-dioxane, benzene, toluene, isopropanol, n-butanol, isobutanol, N,N-dimethylformamide, N,N-dimethylacetamide, n-propanol, tert-butanol, 2-butanone or tetrahydrofuran; acetone is preferred.
  • the temperature in step 2) is 0 to 25°C.
  • Another object of the present invention is to provide a pharmaceutical composition, which contains a therapeutically effective amount of the crystalline form of the above-mentioned compound, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present invention further relates to the application of the crystal form of any compound or the pharmaceutical composition of the present invention in the preparation of orexin receptor antagonists; preferably, the application in the preparation of OX2R selective receptor antagonists.
  • the present invention further relates to the crystal form of any compound or the pharmaceutical composition of the present invention or the application of the pharmaceutical composition of the present invention in the preparation of drugs for the treatment of neurological diseases;
  • the neurological diseases are preferably insomnia, depression, anxiety, drugs Addiction, more preferably major depressive disorder (MDD), primary and secondary insomnia, and depression accompanied by insomnia.
  • MDD major depressive disorder
  • the present invention further relates to a method for treating neurological diseases, wherein the present invention further relates to the crystal form of any compound or the pharmaceutical composition of the invention;
  • the neurological diseases are preferably insomnia, depression, anxiety, and drug addiction. , more preferably major depressive disorder (MDD), primary and secondary insomnia, and depression accompanied by insomnia.
  • MDD major depressive disorder
  • Figure 1 is an XRPD diagram of the free base crystal form I of compound 9.
  • Figure 2 is a DSC diagram of the free base crystal form I of compound 9.
  • Figure 3 is a TGA diagram of the free base crystal form I of compound 9.
  • Figure 4 is an XRPD diagram of the free base crystal form II of compound 9.
  • Figure 5 is a DSC diagram of the free base crystal form II of compound 9.
  • Figure 6 is a TGA diagram of the free base crystal form II of compound 9.
  • an alkyl group refers to a saturated aliphatic hydrocarbon group, which is a linear or branched chain group containing 1 to 20 carbon atoms, preferably an alkyl group containing 1 to 8 carbon atoms, and more preferably 1 to 6 carbon atoms.
  • Alkyl groups of carbon atoms most preferably alkyl groups of 1 to 3 carbon atoms.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1 ,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1, 2-Trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2- Ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methyl Hexyl, 4-methylhexyl, 5-methylhexy
  • Alkyl groups may be substituted or unsubstituted. When substituted, the substituents may be substituted at any available point of attachment.
  • the substituents are preferably one or more of the following groups, independently selected from alkyl groups: Base, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkyl Oxygen group, heterocycloalkyloxy group, cycloalkylthio group, heterocycloalkylthio group, oxo group, carboxyl group or carboxylate group.
  • methyl group, ethyl group, isopropyl group, tert-butyl group and haloalkyl group are preferred.
  • deuterated alkyl, alkoxy-substituted alkyl and hydroxyl-substituted alkyl; the hydroxyl-substituted alkyl can be 2-hydroxyisopropyl, 1-hydroxyethyl.
  • the cycloalkyl group refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent.
  • the cycloalkyl ring contains 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, and more preferably It contains 3 to 8 carbon atoms, and further preferably contains 3 to 6 carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene base, cyclooctyl, etc.; polycyclic cycloalkyl includes spirocycloalkyl, fused cycloalkyl and bridged cycloalkyl.
  • the cycloalkyl ring can be fused to an aryl, heteroaryl or heterocycloalkyl ring, where the ring connected to the parent structure is a cycloalkyl group, non-limiting examples include indanyl, tetralin base, benzocycloheptyl, etc. Cycloalkyl may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkyl, Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio group, oxo group, carboxyl group or carboxylate group.
  • groups which are independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkyl, Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalky
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic heterocyclyl, which contains 3 to 20 ring atoms, one or more of which are selected from nitrogen, oxygen or S(O).
  • m (where m is an integer from 0 to 2), excluding the ring portion of -OO-, -OS- or -SS-, and the remaining ring atoms are carbon. It preferably contains 3 to 12 ring atoms, of which 1 to 4 are heteroatoms; more preferably it contains 3 to 10 ring atoms; further preferably it contains 3 to 8 ring atoms.
  • Non-limiting examples of monocyclic heterocyclyl groups include pyrrolidinyl, pyrrole Alkanone group, piperidin-2-one group, 3,4-dihydropyridine-2(1H)-one group, 4,5-dihydropyridazine-3(2H)-one group, azetidine base, oxetanyl, oxanyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothienyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperazoyl Aldyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl, etc.; Preferred are rrolidinyl, pyrrolidonyl, piperidin-2-one, 3,4-dihydropyridin-2(1H)-one, and 4,5-
  • Polycyclic heterocyclyl groups include spirocyclic, fused-cyclic and bridged-cyclic heterocyclyl groups; the involved spirocyclic, fused-cyclic and bridged-cyclic heterocyclyl groups are optionally connected to other groups through a single bond, or through a ring. Any two or more atoms on are further connected to other cycloalkyl groups, heterocyclyl groups, aryl groups and heteroaryl groups.
  • Heterocyclyl may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkyl, Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio group, oxo group, carboxyl group or carboxylate group.
  • aryl refers to a 6 to 14-membered all-carbon monocyclic or fused polycyclic (that is, a ring that shares adjacent pairs of carbon atoms) group with a conjugated ⁇ electron system, preferably 6 to 10 members. More preferred are 6- to 8-membered ones, such as phenyl and naphthyl, with phenyl being preferred.
  • the aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, where the ring attached to the parent structure is an aryl ring, non-limiting examples of which include:
  • the aryl group may be substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio group, carboxyl group or carboxylate group.
  • heteroaryl refers to a heteroaromatic body containing 1 to 4 heteroatoms and 5 to 14 ring atoms. system, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen.
  • the heteroaryl group is preferably 5 to 10 yuan, more preferably 5 to 8 yuan, most preferably 5 yuan or 6 yuan, such as pyrazinyl, pyridazinyl, imidazolyl, furyl, thienyl, thiazolyl, pyrazolyl , oxazolyl, isoxazolyl, pyrrolyl, triazolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, oxadiazole, pyrazinyl, etc., preferably pyrimidinyl, pyrazolyl, oxazole base, isoxazolyl, oxadiazole, pyridine.
  • the heteroaryl
  • the heteroaryl group may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkyl, Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio group, carboxyl group or carboxylate group.
  • alkoxy refers to -O-(alkyl) and -O-(unsubstituted cycloalkyl), where alkyl is as defined above, preferably an alkyl group containing 1 to 8 carbon atoms, Alkyl groups of 1 to 6 carbon atoms are more preferred, and alkyl groups of 1 to 3 carbon atoms are most preferred.
  • alkoxy include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
  • the alkoxy group may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more of the following groups, which are independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkyl, Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio group, carboxyl group or carboxylate group;
  • Non-limiting examples of alkoxy groups also include: prop-2-oxy and the like.
  • haloalkyl refers to an alkyl group substituted by one or more halogens, wherein alkyl is as defined above.
  • Non-limiting examples of haloalkyl include: trifluoromethyl, trifluoroethyl;
  • Non-limiting examples of haloalkyl groups also include: difluoromethyl, 1,1,2,2-tetrafluoroethyl, perfluoroethyl, and the like.
  • haloalkoxy refers to an alkoxy group substituted by one or more halogens, wherein alkoxy is as defined above;
  • the halogenated alkoxy group can be fully halogenated or partially halogenated, and the number of halogenated groups can be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, etc.; the halogen is preferably F, Cl, Br, I; for example, it can be trifluoromethoxy, difluoromethoxy, 1,1,2,2-tetrafluoroethoxy, perfluoroethoxy, etc.
  • hydroxyalkyl refers to an alkyl group substituted by hydroxyl group, wherein alkyl group is as defined above.
  • alkenyl refers to an alkenyl group, also known as an alkenyl group. It is preferably an alkenyl group containing 2 to 8 carbon atoms, more preferably an alkenyl group containing 2 to 6 carbon atoms, and even more preferably an alkenyl group containing 2 to 4 carbon atoms. Alkenyl group, most preferably an alkenyl group of 2 to 3 carbon atoms. Non-limiting examples of alkenyl groups include: vinyl, propenyl.
  • alkenyl group mentioned therein can be further substituted by other related groups, such as: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, Cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate group.
  • other related groups such as: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, Cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate group
  • alkynyl refers to an alkynyl group, also known as an alkynyl group, and refers to an unsaturated hydrocarbon group containing -C ⁇ C-; an alkynyl group containing 2 to 8 carbon atoms is preferred, and an alkynyl group containing 2 to 6 carbon atoms is more preferred.
  • An alkynyl group is more preferably an alkynyl group having 2 to 4 carbon atoms, and most preferably an alkynyl group having 2 to 3 carbon atoms.
  • alkynyl group mentioned therein can be further substituted by other related groups, such as: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, Cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate group.
  • other related groups such as: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, Cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate
  • haloalkyl refers to an alkyl group substituted by one or more halogens, wherein alkyl is as defined above.
  • haloalkoxy refers to an alkoxy group substituted by one or more halogens, wherein alkoxy is as defined above.
  • hydroxyalkyl refers to an alkyl group substituted by hydroxyl group, wherein alkyl group is as defined above.
  • Halogen means fluorine, chlorine, bromine or iodine.
  • Amino refers to -NH2 .
  • Cyano refers to -CN.
  • Niro refers to -NO 2 .
  • THF tetrahydrofuran
  • DMSO dimethyl sulfoxide
  • IPA refers to isopropyl alcohol.
  • DMF N,N-dimethylformamide
  • DIPEA N,N-diisopropylethylamine
  • HPES 4-hydroxyethylpiperazineethanesulfonic acid
  • Substituted means that one or more hydrogen atoms, preferably up to 5, more preferably 1 to 3 hydrogen atoms in the group are replaced independently of each other by a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and the skilled person can determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, amino or hydroxy groups with free hydrogen may be unstable when combined with carbon atoms with unsaturated (such as olefinic) bonds.
  • Stepoisomerism includes three categories: geometric isomerism (cis-trans isomerism), optical isomerism, and conformational isomerism.
  • the hydrogen atoms described in the present invention can be replaced by its isotope deuterium, and any hydrogen atom in the example compounds involved in the present invention can also be replaced by a deuterium atom.
  • “Pharmaceutical composition” means a mixture containing one or more compounds described herein, or physiologically/pharmaceutically acceptable salts or prodrugs thereof, together with other chemical components, such as physiologically/pharmaceutically acceptable carriers and excipients.
  • the purpose of pharmaceutical compositions is to facilitate administration to living organisms and facilitate the absorption of active ingredients to exert biological activity.
  • X-ray powder diffraction pattern refers to the experimentally observed diffraction pattern or parameters derived from it.
  • the X-ray powder diffraction pattern is characterized by peak position (abscissa) and peak intensity (ordinate).
  • peak position abscissa
  • peak intensity ordinate
  • TGA thermogravimetric analysis
  • DSC differential scanning calorimetry
  • HPLC refers to high performance liquid chromatography (HPLC) experiments.
  • PK refers to pharmacokinetic (PK) experiments.
  • KF refers to the Karl Fischer Moisture Determination (KF) test
  • intermediate 3 For the preparation method of intermediate 3, refer to the preparation method of the third step, fourth step and fifth step of intermediate 2, and replace intermediate 2c with 6-chloro-4-methyl-2-acetyl-pyridine to obtain intermediate 3 (1.2g, colorless oil, 53.7%).
  • reaction solution was diluted with 30 mL of ethyl acetate, washed with water (30 mL*3), the organic phases were combined, dried and concentrated, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate system) to obtain (3aR, 6aS)-5-(2 -Fluoro-6-(2H-1,2,3-triazol-2-yl)benzoyl)hexahydropyrrole[3,4-c]pyrrole-2(1H)-carboxylic acid tert-butyl ester (compound 1b ) (452 mg, light yellow solid, 79.7%).
  • the synthesis method of compound 5 refers to the synthesis method of Example 4, using compound 2 as the raw material to replace compound 3, and obtaining compound 5 (30 mg, yield 66%).
  • Tributyl(1-ethoxyvinyl)stannane (11.90g, 32.95mmol) and compound 7a (5g, 29.95mmol) were dissolved in DMF (5mL), and then bistriphenylphosphine palladium chloride ( II) (1.05g, 1.50mmol), the reaction solution was stirred at 100°C for 2h. After the reaction, add saturated potassium fluoride (10ml) aqueous solution and stir for 1h, filter, and use methylene chloride (10mL*3) for the filter cake. Wash, combine the organic phases, dry, and concentrate to obtain a crude product, which is then column purified (petroleum ether/ethyl acetate system) to obtain compound 7b (5.6 g, yield 92.3%).
  • reaction solution was cooled to room temperature, 15 mL of water was added, filtered, the filtrate was concentrated under reduced pressure, 10 mL of ethanol was added, filtered to remove inorganic salts, and the filtrate was concentrated under reduced pressure to obtain crude product compound 14a (260 mg, Yield 73.1%), directly used in the next reaction.
  • Test Example 1 Determination of the effect of the compound of the present invention on calcium flow in cells stably expressing OX1 and OX2 receptors
  • DMEM high glucose
  • Cell seeding medium DMEM+10% FBS+1X PS;
  • Cell lines CHO-K1/human OX1R and CHO-K1/human OX2R cell lines.
  • CHO-K1/human OX1R and CHO-K1/human OX2R cell lines are cultured separately in complete culture medium at 37°C, 5% CO 2 to 70% to 90% confluence;
  • OX-A for OX1R assays
  • OX-B OX2R assays
  • the compound of the present invention shows a good inhibitory effect in the experiment on the influence of cells stably expressing OX2 receptor receptors on calcium flow, and the inhibitory effect of the compound on OX2 receptor is significantly better than that of OX1 receptor, and has good selectivity.
  • SD rats were used as test animals to study the pharmacokinetic behavior of the compound of the present invention in the rat body (plasma) after oral administration at a dose of 5 mg/kg.
  • the compounds of the embodiments of the present invention are self-made.
  • Drug formulation for oral administration 20% HP- ⁇ -CD in Water
  • the p.o. dose is 5 mg/kg and the administration volume is 10 mL/kg.
  • Blood collection 0.2 mL of blood was collected from the jugular vein of rats before and 0.25, 0.5, 1, 2, 4, 6, 8, and 24 hours after administration, placed in EDTA-K 2 anticoagulant tubes, and centrifuged at 6000 rpm for 6 min at 4°C. Separate plasma and store at -80°C; eat 4 hours after administration.
  • Brain tissue collection After experimental animals were euthanized by CO2 , brain tissue was collected at 0.25h, 0.5h, 1h, 2h, 4h, 6h, 8h, and 24h after administration. The brain tissue was rinsed with pre-cooled PBS, dried, and weighed. Heavy, store at -80°C.
  • Liquid A is 0.1% formic acid aqueous solution
  • liquid B is acetonitrile
  • This experiment uses the telemetry EEG and EMG system of DSI Company to detect the effect of the compound of the present invention on the sleep structure of SD rats.
  • DSI remote sensing pressure measurement system Implant model: TL11M2-F40-EET, Data Sciences International.HD-S02, Data Sciences International.
  • the weight of the animals when arriving at the facility is 190-210g (5-6W). After animals arrive at the facility, they are housed in a vivarium with strictly controlled environmental conditions for an acclimation period of 7-9 days. During the adaptation period, the experimental animals were placed in a 12-h light-dark alternating environment to adapt to adjust the rhythm time, and the animals' health status was monitored every day.
  • the number of animals must meet the requirements of at least 6 animals in each group, and meet the requirements of statistical testing and pharmacodynamic guidelines.
  • the distribution of awake/sleep states of the animals in the solvent control group was consistent with the circadian rhythm.
  • the solvent was not found to have a significant impact on the animal's sleep structure, and the distribution in different awake/sleep stages was consistent with the literature reports.
  • the model was successful and stable.
  • the different sleep stages of each animal in the vehicle group and the medication group were analyzed from 1 hour before administration to after administration.
  • the average value of each group was taken in units of 1 hour to analyze the time distribution changes of each group in different sleep stages.
  • the compounds of the embodiments of the present invention can significantly reduce the NREM/REM latency and the total duration of wakefulness at a lower dose; significantly increase the total NREM duration without affecting the total REM time, and have a low risk of drowsiness. It can be seen that the compound of the present invention has a good sleep-promoting effect on rats and has a low risk of drowsiness.
  • a human liver microsome incubation system was used to quickly predict the inhibitory status of compounds on CYP450 enzyme subtypes using a single-point method.
  • NADPH reduced nicotinamide adenine dinucleotide phosphate
  • the organic solvent can be MeOH, EtOH, EA, Acetone, IPA, IPAc, MEK, 2-Me-THF, MTBE, toluene, cyclohexane, heptane
  • the organic solvent can be MeOH, EtOH, EA, Acetone, IPA, IPAc, MEK, 2-Me-THF, MTBE, toluene, cyclohexane, heptane
  • the crystal form I and crystal form II of compound 9 were placed in saturated water vapor with different relative humidity to allow the compound and water vapor to reach dynamic equilibrium, and the percentage of moisture absorption weight gain of the compound after equilibrium was calculated.
  • the free base crystal form I has a hygroscopic weight gain of about 0.3263% under 80% RH conditions, and is slightly hygroscopic. After two cycles of moisture absorption and desorption under conditions of 0-95% relative humidity, the XRPD spectrum of the free base crystal form I has not changed, that is, the crystal form has not transformed.
  • the free base crystal form II absorbs moisture and gains weight by about 0.2053% under 80% RH conditions, and is slightly hygroscopic.
  • thermodynamic solubility of free base crystal form I in each medium system is higher than the solubility of crystal form II, and the solubility of crystal form I meets the drug concentration required for local treatment.
  • the free base crystal form I was suspended with a 0.5% CMCC-Na aqueous solution containing 0.5%, and the free base crystal form I was suspended with a 0.5% HPMC aqueous solution containing 0.5%. After uniformity, it was administered by gavage to the rats, and three animals were administered in parallel. For rats, the dosage is free base crystal form I (5 mg/kg, 30 mg/kg is suspension), free base crystal form II (30 mg/kg is suspension); free base crystal form I contains 20% The aqueous solution of HP- ⁇ -CD was dissolved clearly, filtered through a filter membrane, and then injected, and administered to three rats in parallel. 5.3 Experimental results

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (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

本发明涉及一种含氮杂环的多环化合物的自由碱晶型及其制备方法。具体涉及一种具有通式(I)化合物自由碱的晶型、制备方法和含有治疗有效量的该晶型的药物组合物,及其作为食欲素受体拮抗剂在制备治疗神经系统疾病相关药物中的用途。

Description

含氮杂环的多环化合物的自由碱晶型及其制备方法 技术领域
本发明属于药物合成领域,具体涉及一种含氮杂环的多环化合物的晶型及其制备方法和应用。
背景技术
下丘脑是调节饮食及能量平衡的中枢,食欲素(Orexin)是下丘脑外侧(LH)orexin神经元合成和分泌的神经肽,因其强烈的促食欲作用而得名,食欲素(Orexin)分为食欲素A(Orexin A)和食欲素B(Orexin B),Orexin A、Orexin B均作用于G蛋白偶联受体食欲素受体OX1R和食欲素受体OX2R,OX1R、OX2R在整个中枢神经系统广泛表达,其中,OX1R与食欲素A的结合能力强于食欲素B,而OX2R与食欲素A和食欲素B的结合能力相当。食欲素与其它影响进食的神经肽之间存在错综复杂的关系,它在增加摄食、饮水、调节睡眠觉醒周期、生殖、体温、血压和感觉等方面有广泛作用,例如通过调节两种不同的G蛋白偶联受体OX1R、OX2R来调节觉醒和觉醒。食欲素受体拮抗剂在治疗神经系统疾病中有潜在治疗优势,包括失眠、抑郁、焦虑、药物成瘾等。
OX1R和OX2R通过磷脂酶C活化胞内Ca2+,OX2R还可以偶联Gi/Go,通过抑制腺苷酸环化酶抑制cAMP的产生。研究发现,在OX1R、OX2R中,OX2R优先表达于下丘脑室旁核参与HPA轴调节,夜间下丘脑-垂体-肾上腺(HPA)过度兴奋是抑郁症病人与正常人的最大区别,对HPA系统过度兴奋的下调,将有助于改善抑郁症状。
目前,已有多个以OX1/2R为靶点处于临床阶段或已上市的药物,例如默克公司的Suvoraxant、卫材的Lemborexant等,但作为Orexin 1/2拮抗剂的药物对OX1R、OX2R两种受体均有拮抗作用,而作用于OX1R会对快速动眼期睡眠(NEM,大脑活动与清醒时一样)和非快速动眼睡眠(NEREM,深度睡眠)在正常生理结构发生改变,即牺牲NREM时间,延长REM时间,进而增加嗜睡的风险,且OX1R也并不能起到抗抑郁效果。
OX2R拮抗剂能够起到抗抑郁效果,OX2R单受体拮抗剂对失眠也能够起到足够的疗效,故选择性OX2R拮抗剂能够避免由于对OX1R作用所产出的嗜睡等多种副作用。目前,OX2R拮抗剂仅有杨森公司开发的的Seltorexant处于临床阶段,主要适应症为重度抑郁症(MDD)、原发和继发性失眠等。
选择性OX2R拮抗剂具有治疗失眠、抑郁、焦虑等神经系统疾病的潜力,具有庞大的临床需求。选择性OX2R拮抗剂作为药物在医药行业具有良好的应用前 景。
PCT专利(申请号:PCT/CN2022/080829)中公开了一系列含氮杂环的多环化合物的结构,在后续的研发中,为了产物易于处理、过滤和干燥,寻求适合的便于储存、产品长期稳定的晶体,本发明对上述化合物的自由碱晶型进行了全面的研究。
发明内容
专利PCT/CN2022/080829所涉及的所有内容均以引证的方式添加到本发明中。
本发明的目的在于提供一种通式(I)所示化合物的晶型,其结构如式(I)所示:
其中:
X1为CR6或N;
X2为CR6或不存在;
R1选自卤素;
R2、R3、R4或R6分别独立地选自氢、氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、羟基烷氧基、烯基、炔基、环烷基、杂环基、芳基或杂芳基,所述的氨基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、羟基烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,任选可以进一步被取代;
R2、R3、R4或R6各自独立地优选氢、氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、C1-8烷基、C1-8氘代烷基、C1-8卤代烷基、C1-8羟烷基、C1-8烷氧基、C1-8卤代烷氧基、C1-8羟基烷氧基、C2-8烯基、C2-8炔基、C3-12环烷基、3-12元杂环基、C6-14芳基或5-14元杂芳基,所述的氨基、C1-8烷基、C1-8氘代烷基、C1-8卤代烷基、C1-8羟烷基、C1-8烷氧基、C1-8卤代烷氧基、C1-8羟基烷氧基、C2-8烯基、C2-8炔基、C3-12环烷基、3-12元杂环基、C6-14芳基和5-14元杂芳基,任选进一步被氘、卤素、氨基、硝基、羟基、巯基、氰 基、羧基、磺酸基、氧代基、硫代基、C1-8烷基、C1-8氘代烷基、C1-8卤代烷基、C1-8羟烷基、C1-8烷氧基、C1-8卤代烷氧基、C2-8烯基、C2-8炔基、C3-12环烷基、3-12元杂环基、C6-14芳基和5-14元杂芳基中的一个或多个取代基所取代;
更优选氢、氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、C1-6烷基、C1-6氘代烷基、C1-6卤代烷基、C1-6羟烷基、C1-6烷氧基、C1-6卤代烷氧基、C1-6羟基烷氧基、C2-6烯基、C2-6炔基、C3-8环烷基、3-8元杂环基、C6-10芳基或5-10元杂芳基,所述的氨基、C1-6烷基、C1-6氘代烷基、C1-6卤代烷基、C1-6羟烷基、C1-6烷氧基、C1-6卤代烷氧基、C1-6羟基烷氧基、C2-6烯基、C2-6炔基、C3-8环烷基、3-8元杂环基、C6-10芳基和5-10元杂芳基,任选进一步被氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、C1-6烷基、C1-6氘代烷基、C1-6卤代烷基、C1-6羟烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-8环烷基、3-8元杂环基、C6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
或者,R2、R3、R4中的任意两个或多个与相连的原子连接形成环烷基、杂环基、芳基或杂芳基,所述环烷基、杂环基、芳基和杂芳基,任选可以进一步被取代;
R5选自羟烷基或卤代烷基,任选可以进一步被取代,所述羟烷基优选为所述卤代烷基优选为
X’为卤素;
R7分别独立地选自氢、氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、羟基烷氧基、烯基、炔基、环烷基、杂环基、芳基或杂芳基,所述的氨基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、羟基烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,任选可以进一步被取代;优选氢、氘、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、羟基烷氧基,任选一步的可以进一步被氘、卤素、C1-6烷基、C1-6氘代烷基、C1-6卤代烷基、C1-6羟烷基、C1-6烷氧基和C1-6卤代烷氧基中的一个或多个取代基所取代;进一步优选氢、氘、C1-6烷基、C1-6氘代烷基、C1-6卤代烷基、C1-6羟烷基、C1-6烷氧基和C1-6卤代烷氧基;再进一步优选氢、氘、C1-3烷基、C1-3氘代烷基、C1-3卤代烷基、C1-3羟烷基、C1-3烷氧基和C1-3卤代烷氧基;更优选氢、氘、C1-3烷基。
本发明优选的实施方案为,提供一种如通式(II)所示化合物的晶型,
其中,
X1为CH或N;
X2为CH或不存在;
R2、R3或R4分别独立地选自氢、氘、卤素、氨基、羟基、C1-3烷基、C1-3氘代烷基、C1-3卤代烷基、C1-3羟烷基、C1-3烷氧基、C1-3卤代烷氧基或C1-3羟基烷氧基;
R5选自
当X2不存在时,取代基R2优选为卤素,所述卤素优选位于三氮唑的邻位或间位,更优选位于邻位,
所述卤素优选为氟;
当X2为CR6时,取代基R2优选为卤素,所述卤素优选位于嘧啶的邻位或间位,更优选位于嘧啶的间位,和/或连接羰基的邻位,
所述卤素优选为氟。
本发明优选的实施方案为,所述通式(I)选自以下化合物:
在本发明优选的实施方式中,提供一种((3aR,6aS)-5-(5-氟-4-(2-羟基丙-2-基)嘧啶-2-基)六氢吡咯并[3,4-c]吡咯-2(1H)-基)(2-氟-6-(嘧啶-2-基)苯基)甲酮(化合物9)的晶型,化合物的结构如下:
所述晶型I的其粉末X射线衍射图谱在2θ为8.6±0.2°处具有衍射峰,或者在2θ为9.2±0.2°处具有衍射峰,或者在2θ为10.2±0.2°处具有衍射峰,或者在2θ为10.8±0.2°处具有衍射峰,或者在2θ为12.0±0.2°处具有衍射峰,或者在2θ为12.8±0.2°处具有衍射峰,或者在2θ为13.7±0.2°处具有衍射峰,或者在2θ为14.2±0.2°处具有衍射峰,或者在2θ为15.5±0.2°处具有衍射峰,或者在2θ为16.6±0.2°处具有衍射峰,或者在2θ为17.3±0.2°处具有衍射峰,或者在2θ为18.4±0.2°处具有衍射峰,或者在2θ为19.0±0.2°处具有衍射峰,或者在2θ为19.5±0.2°处具有衍射峰,或者在2θ为20.5±0.2°处具有衍射峰,或者在2θ为21.3±0.2°处具有衍射峰,或者在2θ为26.5±0.2°处具有衍射峰;或者在2θ为28.6±0.2°处具有衍射峰;优选的,包含其中任选的2处、4处、6处、8处或10处有衍射峰;
优选地,其粉末X射线衍射图谱至少包含位于2θ为9.2±0.2°、10.2±0.2°、16.6±0.2°或18.4±0.2°中的一处或多处衍射峰;优选包含其中2-4处,更优选包含3-4处,最优选包含4处;任选的,进一步,还可以包含2θ为12.8±0.2°、17.3±0.2°、19.0±0.2°、21.3±0.2°或26.5±0.2°中的一处或多处衍射峰,优选包含其中2处、3处、4处或5处;例如:
9.2±0.2°、10.2±0.2°、16.6±0.2°、18.4±0.2°;
9.2±0.2°、10.2±0.2°、16.6±0.2°、17.3±0.2°;
9.2±0.2°、12.8±0.2°、16.6±0.2°、18.4±0.2°;
9.2±0.2°、10.2±0.2°、17.3±0.2°、19.0±0.2°;
9.2±0.2°、12.8±0.2°、17.3±0.2°、19.0±0.2°;
10.2±0.2°、16.6±0.2°、17.3±0.2°、21.3±0.2°;
10.2±0.2°、12.8±0.2°、16.6±0.2°、17.3±0.2°;
12.8±0.2°、17.3±0.2°、19.0±0.2°、21.3±0.2°;
9.2±0.2°、10.2±0.2°、16.6±0.2°、18.4±0.2°;19.0±0.2°;
9.2±0.2°、10.2±0.2°、16.6±0.2°、17.3±0.2°;19.0±0.2°;
9.2±0.2°、12.8±0.2°、16.6±0.2°、18.4±0.2°;19.0±0.2°;
9.2±0.2°、10.2±0.2°、17.3±0.2°、19.0±0.2°;21.3±0.2°;
9.2±0.2°、12.8±0.2°、17.3±0.2°、19.0±0.2°;21.3±0.2°;
10.2±0.2°、16.6±0.2°、17.3±0.2°、21.3±0.2°;26.5±0.2°;
10.2±0.2°、12.8±0.2°、16.6±0.2°、17.3±0.2°;19.0±0.2°;
12.8±0.2°、17.3±0.2°、19.0±0.2°、21.3±0.2°;26.5±0.2°;
9.2±0.2°、10.2±0.2°、16.6±0.2°、18.4±0.2°;19.0±0.2°;21.3±0.2°;
9.2±0.2°、10.2±0.2°、16.6±0.2°、17.3±0.2°;19.0±0.2°;21.3±0.2°;
9.2±0.2°、12.8±0.2°、16.6±0.2°、18.4±0.2°;19.0±0.2°;21.3±0.2°;
9.2±0.2°、10.2±0.2°、17.3±0.2°、19.0±0.2°;21.3±0.2°;26.5±0.2°;
9.2±0.2°、12.8±0.2°、17.3±0.2°、19.0±0.2°;21.3±0.2°;26.5±0.2°;
10.2±0.2°、16.6±0.2°、17.3±0.2°、18.4±0.2°;21.3±0.2°;26.5±0.2°;
10.2±0.2°、12.8±0.2°、16.6±0.2°、17.3±0.2°;18.4±0.2°;19.0±0.2°;
12.8±0.2°、17.3±0.2°、18.4±0.2°;19.0±0.2°、21.3±0.2°;26.5±0.2°;
更优选地,其粉末X射线衍射图谱任选还包含位于2θ为8.6±0.2°、10.8±0.2°、12.0±0.2°、13.7±0.2°、14.2±0.2°或28.6±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-4处,或者5-6处,进一步优选,包含其中任意4处或6处;例如:
8.6±0.2°、10.8±0.2°、12.0±0.2°、13.7±0.2°;
8.6±0.2°、10.8±0.2°、12.0±0.2°、14.2±0.2°;
8.6±0.2°、10.8±0.2°、14.2±0.2°、28.6±0.2°;
10.8±0.2°、12.0±0.2°、13.7±0.2°、14.2±0.2°;
12.0±0.2°、13.7±0.2°、14.2±0.2°、28.6±0.2°;
8.6±0.2°、10.8±0.2°、12.0±0.2°、13.7±0.2°、14.2±0.2°;
8.6±0.2°、10.8±0.2°、12.0±0.2°、14.2±0.2°、28.6±0.2°;
8.6±0.2°、10.8±0.2°、12.0±0.2°、13.7±0.2°、14.2±0.2°、28.6±0.2°;
所述晶型I的X-射线粉末衍射图谱在2θ为9.2±0.2°和18.4±0.2°处具有特征峰;优选地,还包含在2θ为10.2±0.2°、16.6±0.2°、17.3±0.2°和19.0±0.2°处具有特征峰;更优选地,还包含在2θ为12.8±0.2°和21.3±0.2°处 具有特征峰;进一步优选地,还包含在2θ为、26.5±0.2°和28.6±0.2°处具有特征峰,更一步优选的,还包含在8.6±0.2°、10.8±0.2°、12.0±0.2°、13.7±0.2°、14.2±0.2°、15.5±0.2°、19.5±0.2°、20.5±0.2°、23.1±0.2°、27.8±0.2°和32.1±0.2°处具有特征峰。
使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表1所示。
表1

本发明所述的化合物9的晶型I,其X-射线粉末衍射图谱基本如图1所示,其DSC图谱基本如图2所示,其TGA图谱基本如图3所示。
本发明所述的化合物9的晶型I为无水合物晶型。
所述晶型II的其粉末X射线衍射图谱在2θ为9.9±0.2°处具有衍射峰,或者在2θ为10.8±0.2°处具有衍射峰,或者在2θ为13.2±0.2°处具有衍射峰,或者在2θ为14.9±0.2°处具有衍射峰,或者在2θ为16.4±0.2°处具有衍射峰,或者在2θ为17.1±0.2°处具有衍射峰,或者在2θ为17.9±0.2°处具有衍射峰,或者在2θ为19.3±0.2°处具有衍射峰,或者在2θ为19.8±0.2°处具有衍射峰,或者在2θ为20.2±0.2°处具有衍射峰,或者在2θ为20.5±0.2°处具有衍射峰,或者在2θ为21.0±0.2°处具有衍射峰,或者在2θ为21.9±0.2°处具有衍射峰,或者在2θ为25.8±0.2°处具有衍射峰,或者在2θ为26.5±0.2°处具有衍射峰,或者在2θ为27.5±0.2°处具有衍射峰;优选的,包含其中任选的2处、4处、6处、8处或10处有衍射峰;
优选地,其粉末X射线衍射图谱至少包含位于2θ为16.4±0.2°、17.1±0.2°、17.9±0.2°或25.8±0.2°中的一处或多处衍射峰;优选包含其中2-4处,更优选包含3-4处,最优选包含4处;任选的,进一步,还可以包含2θ为10.8±0.2°、14.9±0.2°、19.3±0.2°、21.9±0.2°或26.5±0.2°中的一处或多处衍射峰,优选包含其中2处、3处、4处或5处;例如:
16.4±0.2°、17.1±0.2°、17.9±0.2°、25.8±0.2°;
16.4±0.2°、17.1±0.2°、17.9±0.2°、26.5±0.2°;
16.4±0.2°、17.1±0.2°、25.8±0.2°、26.5±0.2°;
10.8±0.2°、14.9±0.2°、17.9±0.2°、25.8±0.2°;
10.8±0.2°、17.1±0.2°、19.3±0.2°、25.8±0.2°;
16.4±0.2°、17.1±0.2°、17.9±0.2°、21.9±0.2°;
10.8±0.2°、16.4±0.2°、17.1±0.2°、19.3±0.2°;
16.4±0.2°、17.1±0.2°、17.9±0.2°、25.8±0.2°;
16.4±0.2°、17.1±0.2°、17.9±0.2°、25.8±0.2°、26.5±0.2°;
16.4±0.2°、17.1±0.2°、17.9±0.2°、21.9±0.2°、26.5±0.2°;
16.4±0.2°、17.1±0.2°、19.3±0.2°、25.8±0.2°、26.5±0.2°;
10.8±0.2°、14.9±0.2°、17.9±0.2°、25.8±0.2°、26.5±0.2°;
10.8±0.2°、17.1±0.2°、19.3±0.2°、25.8±0.2°;26.5±0.2°;
16.4±0.2°、17.1±0.2°、17.9±0.2°、21.9±0.2°;25.8±0.2°;
10.8±0.2°、16.4±0.2°、17.1±0.2°、19.3±0.2°;21.9±0.2°;
16.4±0.2°、17.1±0.2°、17.9±0.2°、25.8±0.2°;26.5±0.2°;
14.9±0.2°、16.4±0.2°、17.1±0.2°、17.9±0.2°、25.8±0.2°;26.5±0.2°;
14.9±0.2°、16.4±0.2°、17.1±0.2°、17.9±0.2°、26.5±0.2°;26.5±0.2°;
14.9±0.2°、16.4±0.2°、17.1±0.2°、19.3±0.2°、25.8±0.2°、26.5±0.2°;
10.8±0.2°、14.9±0.2°、17.9±0.2°、21.9±0.2°、25.8±0.2°;26.5±0.2°;
10.8±0.2°、17.1±0.2°、19.3±0.2°、21.9±0.2°、25.8±0.2°;26.5±0.2°;
16.4±0.2°、17.1±0.2°、17.9±0.2°、21.9±0.2°;25.8±0.2°;26.5±0.2°;
10.8±0.2°、14.9±0.2°、16.4±0.2°、17.1±0.2°、19.3±0.2°;21.9±0.2°;
10.8±0.2°、16.4±0.2°、17.1±0.2°、17.9±0.2°、25.8±0.2°;26.5±0.2°;
更优选地,其粉末X射线衍射图谱任选还包含位于2θ为9.9±0.2°、13.2±0.2°、19.8±0.2°、20.2±0.2°、20.5±0.2°或21.0±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-4处,或者5-6处,进一步优选,包含其中任意4处或6处;例如:
9.9±0.2°、13.2±0.2°、19.8±0.2°、20.2±0.2°、
9.9±0.2°、13.2±0.2°、19.8±0.2°、20.5±0.2°、
9.9±0.2°、13.2±0.2°、20.5±0.2°、21.0±0.2°、
13.2±0.2°、19.8±0.2°、20.2±0.2°、20.5±0.2°、
9.9±0.2°、13.2±0.2°、19.8±0.2°、20.2±0.2°、20.5±0.2°、
13.2±0.2°、19.8±0.2°、20.2±0.2°、20.5±0.2°、21.0±0.2°;
9.9±0.2°、13.2±0.2°、19.8±0.2°、20.2±0.2°、20.5±0.2°或21.0±0.2°;
在本发明进一步优选的实施方式中,所述晶型II的X-射线粉末衍射图谱在2θ为16.4±0.2°和25.8±0.2°处具有特征峰;优选地,还包含在2θ为17.1±0.2°、17.9±0.2°、21.9±0.2°和26.5±0.2°处具有特征峰;更优选地,还包含在2θ为10.8±0.2°和14.9±0.2°处具有特征峰;进一步优选地,还包含在2θ为9.9±0.2°和19.3±0.2°处具有特征峰,更进一步优选的,还包含在13.2±0.2°、19.8±0.2°、20.2±0.2°、20.5±0.2°、21.0±0.2°和27.5±0.2°处具有特征峰。
使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表2所示。
表2

本发明所述化合物9的晶型II,其X-射线粉末衍射图谱基本如图4所示, 其DSC图谱基本如图5所示,其TGA图谱基本如图6所示。
本发明所述化合物9的晶型II(一水合物),KF测得水含量为3.1%(API:water=1:0.83,摩尔比)。
本发明还提供一种制备通式(I)所示化合物的晶型的方法,具体包括如下步骤:
1)称取适量的自由碱,用不良溶剂混悬;
2)将以上所得混悬液振摇;
3)将以上混悬液快速离心,去除上清液,将固体烘干至恒重得到目标产物;
其中:
所述的不良溶剂选自丙酮、乙酸乙酯、醋酸异丙酯、乙腈、乙醇、四氢呋喃、2-甲基四氢呋喃、二氯甲烷、1,4-二氧六环、苯、甲苯、异丙醇、正丁醇、异丁醇、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、正丙醇、叔丁醇、2-丁酮或3-戊酮、甲基叔丁基醚、水、庚烷、正戊烷;优选醋酸异丙酯、甲苯、甲基叔丁基醚、水、庚烷、正戊烷。
在本发明优选的方案中,步骤1)中的悬浮密度为50~200mg/mL。
在本发明优选的方案中,步骤2)中的温度为0~50℃。
在本发明优选的方案中,步骤2)中的振摇时间选自1~10天。
本发明还提供一种制备通式(I)所示化合物的晶型的方法,具体包括如下步骤:
1)称取适量的自由碱,用良溶剂溶解;
2)向以上所得溶液中加入反溶剂,搅拌至固体析出;
3)将以上混悬液快速离心,去除上清液,剩余固体烘干至恒重得到目标产物;
其中:
所述的良溶剂选自甲醇、丙酮、乙酸乙酯、乙腈、乙醇、四氢呋喃、二氯甲烷、1,4-二氧六环、苯、甲苯、异丙醇、正丁醇、异丁醇、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、正丙醇、叔丁醇、2-丁酮或3-戊酮;优选甲醇、二氯甲烷、乙酸乙酯。
所述的反溶剂选自庚烷、水、甲基叔丁基醚、环己烷、乙酸异丙酯。
在本发明优选的方案中,步骤2)中的温度为0~25℃。
本发明还提供一种制备通式(I)所示化合物的晶型的方法,具体包括如下步骤:
1)称取适量的自由碱,用良溶剂加热溶解;
2)将以上所得溶液迅速置于低温下,搅拌至固体析出;
3)将以上混悬液快速离心,去除上清液,剩余固体放入干燥箱中烘干至恒重得到目标产物;
其中:
所述的良溶剂选自甲醇、丙酮、乙酸乙酯、乙腈、乙醇、四氢呋喃、二氯甲烷、1,4-二氧六环、苯、甲苯、异丙醇、正丁醇、异丁醇、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、正丙醇、叔丁醇、2-丁酮或四氢呋喃;优选丙酮。
在本发明优选的方案中,步骤2)中的温度为0~25℃。
本发明另一目的在于,提供一种药物组合物,其含有治疗有效量的上述所述化合物的晶型,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
本发明进一步涉及任一化合物所述的晶型或本发明的药物组合物在制备食欲素受体拮抗剂中的应用;优选在制备OX2R选择性受体拮抗剂中的应用。
本发明进一步涉及任一化合物所述的晶型或本发明的药物组合物或本发明的药物组合物在制备治疗神经系统疾病药物中的应用;所述神经系统疾病优选失眠、抑郁、焦虑、药物成瘾,更优选为重度抑郁症(MDD)、原发和继发性失眠、伴随失眠的抑郁症。
本发明进一步涉及一种治疗神经系统疾病的方法,其中使用本发明进一步涉及任一化合物所述的晶型或本发明的药物组合物;所述神经系统疾病优选失眠、抑郁、焦虑、药物成瘾,更优选为重度抑郁症(MDD)、原发和继发性失眠、伴随失眠的抑郁症。
附图说明
图1为化合物9的自由碱晶型I的XRPD图示。
图2为化合物9的自由碱晶型I的DSC图示。
图3为化合物9的自由碱晶型I的TGA图示。
图4为化合物9的自由碱晶型II的XRPD图示。
图5为化合物9的自由碱晶型II的DSC图示。
图6为化合物9的自由碱晶型II的TGA图示。
具体实施方式
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
本发明中,烷基是指饱和脂肪族烃基团,其为包含1至20个碳原子的直链或支链基团,优选含有1至8个碳原子的烷基,更优选1至6个碳原子的烷基,最优选1至3个碳原子的烷基。非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、 2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。
烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基,本发明优选甲基、乙基、异丙基、叔丁基、卤代烷基、氘代烷基、烷氧基取代的烷基和羟基取代的烷基;所述羟基取代的烷基可为2-羟基异丙基、1-羟基乙基。
本发明中,所述环烷基是指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个碳原子,优选包含3至12个碳原子,更优选包含3至8个碳原子,进一步优选包含3至6个碳原子。
单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等;多环环烷基包括螺环烷基、稠环烷基和桥环烷基。
所述环烷基环可以稠合于芳基、杂芳基或杂环烷基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等。环烷基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
本发明中,杂环基是指饱和或部分不饱和单环或多环杂环基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包含3至12个环原子,其中1~4个是杂原子;更优选包含3至10个环原子;进一步优选包含3至8个环原子。单环杂环基的非限制性实例包括吡咯烷基、吡咯 烷酮基、哌啶-2-酮基、3,4-二氢吡啶-2(1H)-酮基、4,5-二氢哒嗪-3(2H)-酮基、氮杂环丁烷基、氧杂环丁烷基、氧杂环己烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、二氢咪唑基、二氢呋喃基、二氢吡唑基、二氢吡咯基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基、吡喃基、等;优选咯烷基、吡咯烷酮基、哌啶-2-酮基、3,4-二氢吡啶-2(1H)-酮基、4,5-二氢哒嗪-3(2H)-酮基、氮杂环丁烷基、氧杂环丁烷基、二氢吡咯基、四氢呋喃基、吡唑烷基、吗啉基、哌嗪基和吡喃基;更优选二氢吡咯基、咯烷基、吡咯烷酮基、哌啶-2-酮基、3,4-二氢吡啶-2(1H)-酮基、4,5-二氢哒嗪-3(2H)-酮基、氮杂环丁烷基、氧杂环丁烷基、氧杂环己烷基、吗啉基、哌啶基、哌嗪基、吡喃基。多环杂环基包括螺环、稠环和桥环的杂环基;其中涉及到的螺环、稠环和桥环的杂环基任选与其他基团通过单键相连接,或者通过环上的任意两个或者两个以上的原子与其他环烷基、杂环基、芳基和杂芳基进一步并环连接。
杂环基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
本发明中,芳基是指具有共轭的π电子体系的6至14元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至10元,更优选6至8元,例如苯基和萘基,优选苯基。所述芳基环可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环,其非限制性实例包括:
芳基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
本发明中,杂芳基是指包含1至4个杂原子、5至14个环原子的杂芳族体 系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元,更优选5至8元,最优选为5元或6元,例如吡嗪基、哒嗪基、咪唑基、呋喃基、噻吩基、噻唑基、吡唑基、噁唑基、异噁唑基、吡咯基、三唑基、四唑基、吡啶基、嘧啶基、噻二唑、噁二唑、吡嗪基等,优选嘧啶基、吡唑基、噁唑基、异噁唑基、噁二唑、吡啶。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
杂芳基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
本发明中,烷氧基是指-O-(烷基)和-O-(非取代的环烷基),其中烷基的定义如上所述,优选含有1至8个碳原子的烷基,更优选1至6个碳原子的烷基,最更优选1至3个碳原子的烷基。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基。烷氧基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基;
烷氧基的非限制性实例还包括:丙-2-氧基等。
本发明中,卤代烷基指被一个或多个卤素取代的烷基,其中烷基如上所定义。卤代烷基的非限制性实施例包括:三氟甲基、三氟乙基;
卤代烷基的非限制性实施例还包括:二氟甲基、1,1,2,2-四氟乙基、全氟乙基等。
本发明中,卤代烷氧基是指被一个或多个卤素取代的烷氧基,其中烷氧基如上所定义;
所述卤代烷氧基可为全卤代或部分卤代,卤代的个数可为1、2、3、4、5、6、7、8、9、10等;卤素优选为F、Cl、Br、I;例如可为三氟甲氧基、二氟甲氧基、1,1,2,2-四氟乙氧基、全氟乙氧基等。
本发明中,羟烷基是指被羟基取代的烷基,其中烷基如上所定义。
本发明中,烯基是指链烯基,又称烯烃基,优选含有2至8个碳原子的烯基,更优选2至6个碳原子的烯基,更进一步优选2至4个碳原子的烯基,最优选2至3个碳原子的烯基。烯基的非限制性实施例包括:乙烯基、丙烯基。其中所述的烯基可以进一步被其他相关基团取代,例如:烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
本发明中,炔基是指链炔基,又称炔烃基,指含有-C≡C-的不饱和烃基;优选含有2至8个碳原子的炔基,更优选2至6个碳原子的炔基,更进一步优选2至4个碳原子的炔基,最更优选2至3个碳原子的炔基。其中所述的炔基可以进一步被其他相关基团取代,例如:烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
本发明中,卤代烷基是指被一个或多个卤素取代的烷基,其中烷基如上所定义。
本发明中,卤代烷氧基是指被一个或多个卤素取代的烷氧基,其中烷氧基如上所定义。
本发明中,羟烷基是指被羟基取代的烷基,其中烷基如上所定义。
“羟基”指-OH基团。
“卤素”指氟、氯、溴或碘。
“氨基”指-NH2
“氰基”指-CN。
“硝基”指-NO2
“THF”指四氢呋喃。
“DMSO”指二甲基亚砜。
“IPA”指异丙醇。
“MeOH”指甲醇。
“EtOH”指乙醇。
“DMF”指N、N-二甲基甲酰胺。
“DIPEA”指N,N-二异丙基乙胺。
“HEPES”指4-羟乙基哌嗪乙磺酸。
“X选自A、B、或C”、“X选自A、B和C”、“X为A、B或C”、“X为A、B和C”等不同用语均表达了相同的意义,即表示X可以是A、B、C中的任意一种或几种。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生的场合。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有自由氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
“立体异构”包含几何异构(顺反异构)、旋光异构、构象异构三类。
本发明所述的氢原子均可被其同位素氘所取代,本发明涉及的实施例化合物中的任一氢原子也均可被氘原子取代。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
X-射线粉末衍射图谱(XRPD),是指实验观测到的衍射图或源自其的参数,通过峰位置(横坐标)及峰强度(纵坐标)表征X-射线粉末衍射图谱。本领域技术人员能够理解,其中的实验误差取决于仪器的条件、样品的准备和样品的纯度。特别是,本领域技术人员公知,X射线衍射图通常会随着仪器的条件而有所改变,本领域技术人员应理解XRPD合适的误差容限可以为:2θ±0.5°;2θ±0.4°;2θ±0.3°;2θ±0.2°。特别需要指出的是,X射线衍射图的相对强度也可能随着实验条件的变化而变化,所以峰强度的顺序不能作为唯一或决定性因素。另外,由于样品高度等实验因素的影响,会造成峰角度的整体偏移,通常允许一定的偏移。因而,本领域技术人员可以理解的是,任何具有与本发明图谱的特征峰相同或相似的晶型均属于本发明的范畴之内。
“TGA”是指热重分析(TGA)实验。
“DSC”是指差示扫描量热法(DSC)实验。
“HPLC”是指高效液相色谱(HPLC)实验。
“PK”是指药物代谢动力学(PK)实验。
“KF”是指卡尔费休水分测定(KF)实验
以下结合实施例进一步描述本发明,但这些实施例并非限制着本发明的范围。
实施例
中间体的制备:
中间体1:2-(2-氯-6-甲基嘧啶-4-基)丙醇的制备
-78℃下向甲基溴化镁(5mL,1M in THF)中加入中间体1a(674mg,3.6mmol)的THF(5mL)溶液,混合物在-78℃搅拌2小时,随后室温搅拌12小时。反应液缓慢加入饱和氯化铵溶液(20mL)淬灭,二氯甲烷萃取(20mL*3),有机相合并干燥浓缩,残渣用硅胶柱色谱法(石油醚/乙酸乙酯体系)纯化得到中间体1(362mg,无色油状物),产率53.7%。
MS m/z(ESI):187.1[M+1]
中间体2:2-(5-氟-2-(3aR,6aS)-六氢吡咯[3,4-c]吡咯-2(1H)-基)-6-甲基嘧啶-4-基)-丙-2-醇的制备
第一步
在氮保护下,向中间体2a(1.3g,7.18mmol)的无水DMF(15mL)溶液中加入三丁基-(1-乙氧基-乙烯基)-锡烷(2.7mL,7.99mmol)和二氯双(三苯基膦)钯(II)(100mg,0.142mmol)。将混合物在100℃加热16小时,冷却,加入氟化钾(aq)的饱和溶液并将混合物在室温搅拌1小时。通过硅藻土过滤后,用水彻底洗涤有 机相,再用乙酸乙酯萃取并浓缩。粗品通过过柱(PE:EA=10:1)获得中间体2b(1.3g,83.5%)。
MS m/z(ESI):217.0[M+1]
第二步
把中间体2b(1.3g,6.00mmol)溶在THF(10mL)中,加入3N的HCl(5mL)。将混合物在室温搅拌1小时,LCMS监测已经反应完全,用饱和的NaHCO3调节pH=7~8左右,用乙酸乙酯萃取,有机相浓缩得到中间体2c(1.1g,97.2%)。直接用作下一步。
MS m/z(ESI):189.0[M+1]
1H NMR(400MHz,Chloroform-d)δ2.69(s,3H),2.62(d,J=2.8Hz,3H).
第三步
在0℃下,将中间体2c(1.9g,10.07mmol),MeMgBr(3M,4.37mL)加入到THF(40mL)中。将反应溶液在25℃搅拌1时,加入饱和的NH4Cl(20ml),用乙酸乙酯(40ml)萃取,并将合并的萃取物经Na2SO4干燥,旋干,过柱(PE:EA=3:1)得到中间体2d(1.4g,产率67.9%)。
MS m/z(ESI):205.0[M+1]
第四步
将rac-(3aR,6aS)-2,3,3a,4,6,6a-六氢-1H-吡咯[3,4-c]吡咯-5-羧酸叔丁酯(500mg,2.36mmol),中间体2d(530.16mg,2.59mmol)和碳酸铯(1.55g,4.76mmol)加入到DMF(10mL)中。将反应溶液在100℃搅拌3时,加入水(20mL),再用二氯甲烷(30mL*2)萃取,并将合并的萃取物经Na2SO4干燥,旋干,粗品通过过柱(PE:EA=3:1)得到中间体2e(720mg,产率80.3%)。
MS m/z(ESI):381.2[M+1]
第五步
将中间体2e(1g,2.63mmol),三氟醋酸(2.69mmol,2mL)加入到DCM(3mL)中。反应溶液在25℃搅拌3小时旋干,得到中间体2(700mg,产率95.0%)。
MS m/z(ESI):281.2[M+1]
中间体3:2-(6-(3aR,6aS)-六氢吡咯[3,4-c]吡咯-2(1h)-基)-4-甲基吡啶-2-基)丙烷-2-醇的制备
中间体3的制备方法,参照中间体2第三步,第四步和第五步的制备方法,以6-氯-4-甲基-2-乙酰基-吡啶代替中间体2c,得到中间体3(1.2g,无色油状物,53.7%)。
MS m/z(ESI):262.2[M+1]
实施例1
化合物1:(2-氟-6-(2H-1,2,3-三唑-2-基)苯基)((3aR,6aS)-5-(4-(2-羟基丙烷-2-基)-6-甲基嘧啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮的制备
第一步
向1a(300mg,1.42mmol)和2-氟-6-(2H-1,2,3-三唑-2-基)苯甲酸(310mg,1.50mmol)的DMF(5.0mL)溶液中缓慢加入HATU(809mg,2.13mmol)和DIPEA(550mg,4.26mmol),混合物室温搅拌2小时。反应液加乙酸乙酯30mL稀释,水洗涤(30mL*3),有机相合并干燥浓缩,残渣用硅胶柱色谱法(石油醚/乙酸乙酯体系)纯化得到(3aR,6aS)-5-(2-氟-6-(2H-1,2,3-三唑-2-基)苯甲酰基)六氢吡咯[3,4-c]吡咯-2(1H)-羧酸叔丁酯(化合物1b)(452mg,淡黄色固体,79.7%)。
MS m/z(ESI):402.1[M+1]
第二步
0℃下向1b(450mg,1.12mmol)的DCM(6.0mL)溶液中缓慢加入TFA(3.0mL),混合物室温搅拌2小时。反应液浓缩,得到(2-氟-6-(2H-1,2,3-三唑-2-基)苯基)((3aR,6aS)-六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮(化合物1c)(330mg,淡黄色油状物),直接用于下一步。
MS m/z(ESI):302.0[M+1]
第三步
向圆底烧瓶中加入中间体1(61.5mg,0.33mmol),1c(100mg,0.33mmol), Pd2(dba)3(18mg,0.02mmol),BINAP(25mg,0.04mmol),碳酸铯(215mg,0.66mmol)和Dioxane(2mL),混合物在氮气保护下100℃搅拌12小时。反应液冷却后加入水(5mL)淬灭,二氯甲烷萃取(10mL*3),有机相合并干燥浓缩,残渣经制备HPLC纯化,得到(2-氟-6-(2H-1,2,3-三唑-2-基)苯基)((3aR,6aS)-5-(4-(2-羟基丙烷-2-基)-6-甲基嘧啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮(化合物1)(32.4mg,白色固体,21.8%)。
MS m/z(ESI):452.2[M+1]
1H NMR(400MHz,DMSO-d6)δ8.16(s,1H),7.94(s,1H),7.80(dd,J=17.2,8.2Hz,1H),7.66(td,J=8.3,6.1Hz,1H),7.44(dt,J=12.0,8.6Hz,1H),6.74(d,J=4.1Hz,1H),5.10(d,J=4.0Hz,1H),3.73(dtd,J=22.4,7.2,6.5,3.6Hz,2H),3.59–3.41(m,4H),3.35(d,J=4.8Hz,1H),3.16–2.90(m,3H),2.28(d,J=3.5Hz,3H),1.36(d,J=3.3Hz,6H).
实施例2
化合物2:((3aR,6aS)-5-(5-氟-4-(2-羟基丙烷-2-基)-6-甲基嘧啶-2-基)六氢吡咯并[3,4-c]吡咯-2(1H)-基)(2-氟-6-(2H-1,2,3-三唑-2-基)苯基)甲酮的制备
化合物2的合成方法,参考实施例1的合成方法。
MS m/z(ESI):470.2[M+1].
实施例3
化合物3:(2-氟-6-(嘧啶-2-基)苯基)((3aR,6aS)-5-(6-(2-羟基丙烷-2-基)-4-甲基吡啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮的制备
将2-氟-6-嘧啶-2-基苯甲酸(50mg,0.23mmol)和中间体3(65.88mg,0.25mmol)加入到MeCN(5mL)中。25℃下加入四甲基氯代脲六氟磷酸酯(128.33mg,0.46mmol),和1-甲基咪唑(56.45mg,0.69mmol)。反应溶液25℃下 搅拌2小时,加入水(20mL),再用二氯甲烷(30mL*2)萃取,并将合并的萃取物经Na2SO4干燥,旋干,粗品通过制备HPLC纯化得到化合物3(64mg,产率60.5%)。
MS m/z(ESI):462.2[M+1]
1H NMR(400MHz,DMSO-d6)δ8.93(d,J=4.9Hz,1H),8.82(d,J=4.9Hz,1H),8.05(dd,J=25.0,7.8Hz,1H),7.62(q,J=7.7Hz,1H),7.54–7.38(m,2H),6.85(d,J=9.8Hz,1H),6.68(d,J=22.0Hz,1H),3.80(ddd,J=37.8,12.6,7.8Hz,4H),3.61–3.51(m,3H),3.18(dt,J=10.8,5.1Hz,3H),2.36(d,J=3.4Hz,3H),1.50(d,J=5.2Hz,6H).
实施例4
化合物4:(2-氟-6-(嘧啶-2-基)苯基)((3aR,6aS)-5-(6-(2-氟丙烷-2-基)-4-甲基吡啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮的制备
在0℃下,将化合物3(45mg,0.1mmol)和DAST(23.57mg,0.15mmol)加入到DCM(5mL)中。反应溶液在25℃搅拌0.5小时,待反应结束后加入水(20ml),再用二氯甲烷(30mL*2)萃取,合并有机相并经Na2SO4干燥,旋干,粗品经制备HPLC纯化得到化合物4(31mg,产率68.6%)。
MS:m/z(ESI):464.2[M+1].
1H NMR(400MHz,DMSO-d6)δ8.92(d,J=4.9Hz,1H),8.76(d,J=4.9Hz,1H),8.03(dd,J=31.4,7.8Hz,1H),7.67–7.33(m,3H),6.56(d,J=3.8Hz,1H),6.20(d,J=5.4Hz,1H),3.85–3.65(m,2H),3.59–3.47(m,3H),3.27–2.90(m,5H),2.23(s,3H),1.60(dd,J=22.0,3.0Hz,6H).
实施例5
化合物5:((3aR,6aS)-5-(5-氟-4-(2-氟丙烷-2-基)-6-甲基嘧啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)(2-氟-6-(2H-1,2,3-三唑-2-基)苯基)甲酮的制备
化合物5的合成方法参考实施例4的合成方法,以化合物2为原料替换化合物3,得到化合物5(30mg,产率66%)。
MS:m/z(ESI):472.2[M+1].
1H NMR(400MHz,DMSO-d6)δ8.16(s,1H),7.96(s,1H),7.80(dd,J=14.7,8.2Hz,1H),7.66(td,J=8.3,6.0Hz,1H),7.44(q,J=9.2Hz,1H),3.76–3.64(m,2H),3.53(ddd,J=23.2,11.1,7.2Hz,5H),3.17–2.94(m,3H),2.36–2.23(m,3H),1.67(dd,J=21.9,4.0Hz,6H).
实施例6
化合物6:(2-氟-6-(2H-1,2,3-三唑-2-基)苯基)((3R,6S)-5-(6-(2-羟基丙烷-2-基)-4-甲基吡啶-2-基)六氢吡咯并[3,4-c]吡咯-2(1H)-基)甲酮的制备
第一步
化合物6-1(100.00mg,539.08μmol)溶于四氢呋喃(5.0mL)中,随后将甲基溴化镁(3M,538.77μL)加入反应液,反应液于0℃条件下搅拌反应0.5h。饱和氯化铵(5ml)加入反应液,二氯甲烷(3*10mL)萃取,有机相合并,干燥,浓缩得粗品,粗品柱纯化(石油醚/乙酸乙酯体系)得产物2-(6-氯-4-甲基吡啶-2-基)丙-2-醇(化合物6-2)(90mg,90.0%)。
MS m/z(ESI):186.1[M+1]
第二步
化合物6-2(65.95mg,0.36mmol),化合物3c(100mg,0.33mmol),4,5-双二苯基膦-9,9-二甲基氧杂蒽(34.22mg,59.20μmol),醋酸钯(6.63mg,29.61μmol)溶于1,4-二氧六环(2mL)中,随后将叔丁醇钾(33.16mg,296.07μmol)加入反应液,后将反应液于100℃微波条件下搅拌反应1h。饱和食盐水(10ml)加入反应液,二氯甲烷(3*10mL)萃取,有机相合并,干燥,浓缩得粗品,粗品制备纯化得(2-氟-6-(2H-1,2,3-三唑-2-基)苯基)((3R,6S)-5-(6-(2-羟基丙烷-2-基)-4-甲基吡啶-2-基)六氢吡咯并[3,4-c]吡咯-2(1H)-基)甲酮(化合物6)(8mg,6.00%产率)。
MS m/z(ESI):451.1[M+1].
1H NMR(400MHz,DMSO)δ7.95(s,1H),7.80(dd,J=18.7,8.2Hz,1H),7.73 –7.55(m,1H),7.40(m,1H),7.02(dd,J=58.3,50.0Hz,2H),6.66(d,J=6.9Hz,1H),6.11(s,1H),3.87–3.48(m,7H),3.27–2.90(m,3H),2.21(d,J=4.9Hz,3H),1.38(d,J=6.3Hz,6H).
实施例7
化合物7:((3aR,6aS)-5-(5-氟-4-(2-羟基丙-2-基)嘧啶-2-基)六氢吡咯并[3,4-c]吡咯-2(1H)-基)(2-氟-6-(2H-1,2,3-三唑-2-基)苯基)甲酮的制备
第一步
将三丁基(1-乙氧基乙烯基)锡烷(11.90g,32.95mmol)和化合物7a(5g,29.95mmol)溶于DMF(5mL)中,随后加入双三苯基膦氯化钯(II)(1.05g,1.50mmol),反应液于100℃下搅拌反应2h,反应结束后,加入饱和氟化钾(10ml)水溶液并搅拌1h,过滤,滤饼用二氯甲烷(10mL*3)洗涤,有机相合并,干燥,浓缩得粗品,粗品柱纯化(石油醚/乙酸乙酯体系)得到化合物7b(5.6g,产率92.3%)。
MS m/z(ESI):203.0[M+1]
第二步
化合物7b(5.6g,27.64mmol)溶于四氢呋喃(2mL)中,随后将盐酸(2mL,27.64mmol)加入反应液,后将反应液于50℃条件下搅拌反应2h,饱和氯化钠(10mL)加入反应液,二氯甲烷(10mL*3)萃取,有机相合并,干燥,浓缩得粗品,粗品柱纯化(石油醚/乙酸乙酯体系)得到化合物7c(4.4g,产率91.2%)。
MS m/z(ESI):175.0[M+1]
第三步
化合物7c(4.4g,25.21mmol)溶于四氢呋喃(5mL)中,-78℃下,将甲基溴化镁(3M,9.24mL)加入反应液,后将反应液于-78℃条件下搅拌反应15min,饱和氯化铵(10mL)加入反应液,二氯甲烷(10mL*3)萃取,有机相合并,干燥,浓缩得粗品,粗品柱纯化(石油醚/乙酸乙酯体系)得到7d(2.2g,产率45.8%)。
MS m/z(ESI):191.0[M+1]
第四步
化合物7的合成方法,参考化合物6的合成方法,以化合物1c和化合物7d为原料,得到化合物7(38mg,产率42.6%)。
MS:m/z(ESI):456.2[M+1]
1H NMR(400MHz,DMSO)δ8.21(t,J=23.7Hz,1H),7.99(d,J=73.9Hz,2H),7.73(dd,J=17.8,8.2Hz,1H),7.59(dd,J=14.6,8.1Hz,1H),7.37(dd,J=19.5,8.8Hz,1H),5.13(d,J=9.5Hz,1H),3.91–3.34(m,7H),3.14–2.87(m,3H),1.39(d,J=4.2Hz,6H).
实施例8
化合物8:(2-氟-6-(嘧啶-2-基)苯基)((3R,6S)-5-(6-(2-羟基丙烷-2-基)-5-甲基吡啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮的制备
第一步
化合物8-1(0.2g,1.75mmol),2-氟-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼烷-2-基)苯甲酸甲酯(586.94mg,2.10mmol),磷酸钾(1.11g,5.24mmol)溶于1,4-二氧六环(4.0mL)和H2O(1.0mL)中,随后将[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(142.49mg,174.62μmol)加入反应液,后将反应液于100℃下搅拌反应16h。饱和食盐水(10ml)加入反应液,二氯甲烷(3*20mL)萃取,有机相合并,干燥,浓缩得粗品,粗品柱纯化(石油醚/乙酸乙酯体系)得2-氟-6-(嘧啶-2-基)苯甲酸甲酯(化合物8-2)(0.2g,49.1%)。
MS m/z(ESI):233.1[M+1]
第二步
化合物8-2(0.8g,3.45mmol)溶于水(2mL)和甲醇(2mL)中,随后将氢氧化钠(413.39mg,10.33mmol)加入反应液,后将反应液于70℃条件下搅拌反应16h。2摩尔/升的盐酸溶液(10ml)加入反应液,调节反应液pH至酸性,乙酸乙酯(3*20mL)萃取,有机相合并,干燥,浓缩得粗品2-氟-6-(嘧啶-2-基)苯甲酸(化合物8-3)(260mg,34.6%)。粗品未纯化直接用于下一步。
MS m/z(ESI):219.1[M+1]
第三步
化合物8-3(0.2g,917.43μmol),N,N-二甲基甲酰胺(0.1ml)溶于二氯甲烷(2mL)中,随后将草酰氯(232.70mg,1.83mmol)加入反应液,后将反应液于室温条件下搅拌反应0.5h,反应液浓缩得粗品。将粗品,叔丁基(3R,6S)-2,3,3a,4,6,6a-六氢-1H-吡咯并[3,4-c]吡咯-5-羧酸酯(192.88mg,909.81μmol)溶于二氯甲烷(2mL)中,随后N-乙基-N-异丙基丙-2-胺(352.29mg,2.73mmol)加入反应液,后将反应液于室温条件下搅拌反应1h。饱和食盐水(10ml)加入反应液,二氯甲烷(3*10mL)萃取,有机相合并,干燥,浓缩得粗品,粗品柱纯化(石油醚/乙酸乙酯体系)得(3R,6S)-5-(2-氟-6-(嘧啶-2-基)苯甲酰基)六氢吡咯并[3,4-c]吡咯-2(1H)-羧酸叔丁酯(化合物8-4)(0.2g,53.4%)。
MS m/z(ESI):413.2[M+1]
第四步
化合物8-4(200.00mg,485.43μmol)溶于盐酸/1,4-二氧六环(4M,2mL)中,随后将反应液于室温条件下搅拌反应1h。反应液过滤,滤饼收集,干燥得(2-氟-6-(嘧啶-2-基)苯基)((3R,6S)-六氢吡咯并[3,4-c]吡咯-2(1H)-基)甲酮(化合物8-5)(120mg,79.2%)。
MS m/z(ESI):313.2[M+1]
第五步
化合物8的合成方法,参考化合物6的合成方法,以化合物8-5和化合物2-(6-氯-3-甲基吡啶-2-基)-丙-2-醇为原料,得到(2-氟-6-(嘧啶-2-基)苯基)((3R,6S)-5-(6-(2-羟基丙烷-2-基)-5-甲基吡啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮(化合物8)(1.8mg,2.6%)。
MS:m/z(ESI):462.2[M+1]
实施例9
化合物9:((3aR,6aS)-5-(5-氟-4-(2-羟基丙-2-基)嘧啶-2-基)六氢吡咯并[3,4-c]吡咯-2(1H)-基)(2-氟-6-(嘧啶-2-基)苯基)甲酮的制备
化合物9的合成方法,参考化合物6的合成方法,以化合物8-5和7d为原料,得到化合物9(22mg,产率28.6%)。
MS:m/z(ESI):467.2[M+1]
1H NMR(400MHz,DMSO)δ8.86(dd,J=48.0,4.9Hz,2H),8.32(s,1H),8.02(dd,J=35.7,7.8Hz,1H),7.60(dd,J=14.0,7.9Hz,1H),7.44(ddd,J=26.7,12.9,4.9Hz,2H),5.33–5.10(m,1H),3.89–3.46(m,7H),3.23–2.95(m,3H),1.46(s,6H).
实施例10
化合物10:(4-氟-2-(2H-1,2,3-三唑-2-基)苯基)((cis)-5-(5-氟-4-(2-羟基丙烷-2-基)-6-甲基嘧啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮的制备
将中间体2(80mg,0.285mmol)溶解在乙腈(2mL)中,加入4-氟-2-(三唑-2-基)苯甲酸(59mg,0.285mmol),N-甲基吗啉(36mg,0.356mmol)和N,N,N',N'-四甲基氯甲脒六氟磷酸盐(120mg,0.428mmol),反应液室温反应2小时。待反应结束将反应液倒入50mL水中,乙酸乙酯萃取(30mL*2)。合并有机相,依次用水(30mL)、饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残渣用反向制备HPLC色谱法(乙腈/水体系)纯化得到化合物10(16.3mg,产率12.2%)。
MS m/z(ESI):470.2[M+1]
1H NMR(400MHz,DMSO-d6)δ8.03(s,2H),7.74(dd,J=9.6,2.0Hz,1H),7.60-7.56(m,1H),7.43-7.38(m,1H),5.17(s,1H),3.75-3.65(m,2H),3.60-3.56(m,1H),3.48-3.44(m,3H),3.34-3.29(m,1H),3.02-2.93(m,3H),2.31(s,3H),1.46(s,6H).
实施例11
化合物11:(3-氟-2-(2H-1,2,3-三唑-2-基)苯基)((cis)-5-(5-氟-4-(2-羟基丙烷-2-基)-6-甲基嘧啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮的制备
第一步
将2,3-二氟苯甲腈(12.0g,86.3mmol)溶解在N,N-二甲基甲酰胺(100mL)中,加入1,2,3-三氮唑(5.96g,86.3mmol)和碳酸铯(28.1g,86.3mmol),反应液加热至120℃反应2小时。反应结束后,反应液冷却至室温,倒入200mL水中,甲基叔丁基醚萃取(100mL*2)。合并有机相,依次用水(100mL)、饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残渣用硅胶柱色谱法(石油醚/乙酸乙酯体系)纯化得到化合物11a(6.5g,产率40.0%)。
MS m/z(ESI):189.0[M+1]
1H NMR(400MHz,CDCl3)δ8.00(s,2H),7.67-7.65(m,1H),7.62-7.55(m,2H).
第二步
将化合物11a(1.5g,7.97mmol)溶解在1,4-二氧六环(10mL)和水(30mL)中,加入氢氧化钠(3.19g,79.8mmol),加热至110℃反应4小时。反应液冷却至0℃,稀盐酸调pH至1左右,乙酸乙酯萃取(100mL*2)。合并有机相,依次用水(100mL)、饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到化合物11b(1.5g,产率90.8%)。
MS m/z(ESI):208.0[M+1]
1H NMR(400MHz,CDCl3)δ7.89(s,2H),7.87(d,J=7.6Hz,1H),7.63-7.58(m,1H),7.52-7.49(m,1H).
第三步
将(cis)-2-Boc-六氢吡咯并[3,4-c]吡咯(120mg,0.565mmol)溶解在无水乙腈(2mL)中,加入化合物11b(117mg,0.565mmol),N,N,N',N'-四甲基氯甲脒六氟磷酸盐(238mg,0.848mmol)和N-甲基吗啉(86mg,0.850mmol),室温反应2小时。反应液倒入30mL水中,乙酸乙酯萃取(30mL*2)。合并有机相,依次用水(80mL)、饱和氯化钠溶液(80mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残渣用硅胶柱色谱法(石油醚/乙酸乙酯体系)纯化得到化合物11c(210mg,产率92.6%)。
MS m/z(ESI):402.2[M+1]
1H NMR(400MHz,DMSO-d6)δ8.07(s,2H),7.71-7.66(m,1H),7.63-7.58(m,1H),7.42(d,J=7.6Hz,1H),3.49-3.42(m,3H),3.40-3.36(m,1H),3.23-3.19(m,1H),3.16-3.01(m,3H),2.87-2.78(m,2H),1.41(s,9H).
第四步
将化合物11c(80mg,0.199mmol)溶解在无水二氯甲烷(2mL)中,加入盐酸二氧六环溶液(4M,2mL),室温反应2小时。反应液减压浓缩,得到化合物11d(60mg,产率99.9%)。
MS m/z(ESI):302.1[M+1]
第五步
将化合物11d(60mg,0.199mmol)溶解在无水N,N-二甲基甲酰胺(2mL)中,加入中间体2d(45mg,0.220mmol)和碳酸铯(130mg,0.399mmol),加热至110℃反应2小时。反应液冷却至室温,倒入30mL水中,乙酸乙酯萃取(30mL*2)。合并有机相,依次用水(30mL)、饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残渣用反向制备HPLC色谱法(乙腈/水体系)纯化得到化合物11(31.3mg,产率33.5%)。
MS m/z(ESI):470.2[M+1]
1H NMR(400MHz,DMSO-d6)δ8.00(s,2H),7.70-7.65(m,1H),7.62-7.57(m,1H),7.42(d,J=7.6Hz,1H),5.16(s,1H),3.73-7.69(m,1H),3.64-3.59(m,1H),3.57-3.49(m,2H),3.43-3.34(m,2H),3.29-3.25(m,1H),3.13-3.09(m,1H),2.99-2.93(m,2H),2.32(d,J=2.8Hz,3H),1.46(s,6H).
实施例12
化合物12:((3aR,6aS)-5-(5-氟-4-(2-羟基丙烷-2-基)-6-甲基嘧啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)(2-氟-6-(吡啶-2-基)苯基)甲酮的制备
第一步
N2保护下,在化合物12a(4g,17.16mmol)和2-(三丁基甲锡烷基)吡啶(6.97g,18.93mmol)的DMF(60mL)溶液中加入CuI(653.81mg,3.43mmol),Pd(PPh3)4(1.98g,1.71mmol)和CsF(5.21g,34.30mmol)。120℃反应5小时。冷却到室温后,加入150mL水,用乙酸乙酯(100mL*2)萃取。有机相合并后,用无水硫酸钠干燥,过滤,浓缩。得到的粗品经纯化得到化合物12b(2.8g,产率70.5%)。
MS m/z(ESI):232.1[M+1]
第二步
在化合物12b(800mg,3.46mmol)的水(5.0mL)和甲醇(5.0mL)的混合溶液中加入氢氧化钠(688.98mg,17.22mmol)。25℃反应3小时。加入2N HCl直至pH=2,用乙酸乙酯(20mL*2)萃取。有机相用无水硫酸钠干燥,过滤,旋干,得到化合物12c(650mg,产率86.5%)。
MS m/z(ESI):218.0[M+1]
第三步
在12c(100mg,0.46mmol)和中间体2(129mg,0.46mmol)的乙腈(3.0mL)溶液中加入四甲基氯代脲六氟磷酸酯(257mg,0.92mmol)和N-甲基咪唑(188mg,2.29mmol)。25℃下搅拌30分钟。在反应液中加入20mL水,乙酸乙酯萃取(20mL*2)。有机相合并后,用无水硫酸钠干燥,过滤,浓缩。得到的粗品用制备HPLC纯化,得到化合物12(40mg,产率18.1%)。
MS m/z(ESI):480.2[M+1]
1H NMR(400MHz,DMSO-d6)δ8.64–8.44(m,1H),7.87(dq,J=21.9,7.1,6.6Hz,1H),7.76–7.48(m,3H),7.43–7.19(m,2H),5.16(s,1H),3.67(dddd,J=38.5,25.2,11.5,7.2Hz,5H),3.23–2.95(m,5H),2.32(dd,J=11.6,2.8Hz,3H),1.46(d,J=11.0Hz,6H).
实施例13
化合物13:(5-氟-2-(2H-1,2,3-三唑-2-基)苯基)((cis)-5-(5-氟-4-(2-羟基丙烷-2-基)-6-甲基嘧啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮的制备
第一步
将5-氟-2-碘代苯甲酸(300mg,1.13mmol)溶解在无水N,N-二甲基甲酰胺(4mL)中,加入1,2,3-三氮唑(117mg,1.69mmol),碘化亚铜(258mg,1.35mmol),反式-N,N'-二甲基-1,2-环己烷二胺(193mg,0.97mmol)和碳酸铯(735mg,2.26mmol),微波120℃反应15分钟。反应液冷却至室温,倒入50mL水中,稀盐酸调pH至5,乙酸乙酯萃取(50mL*2)。合并有机相,依次用水(50mL)、饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残渣用反向C18柱层析色谱法(乙腈/水体系)纯化得到化合物13a(150mg,产率64.2%)。
MS m/z(ESI):208.0[M+1].
第二步
化合物13的制备方法,参照化合物12第三步的制备方法,以化合物13a替换化合物12c得到化合物13(60mg,产率30%)。
MS m/z(ESI):470.2[M+1].
1H NMR(400MHz,DMSO-d6)δ7.98(s,2H),7.93-7.89(m,1H),7.49-7.46(m,1H),7.37-7.35(m,1H),5.16(s,1H),3.81-3.57(m,3H),3.42-3.37(m,4H),3.04-2.94(m,3H),2.30(s,3H),1.46(s,6H).
实施例14
化合物14:(5-氟-2-(嘧啶-2-基)苯基)((cis)-5-(5-氟-4-(2-羟基丙烷-2-基)-6-甲基嘧啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)甲酮的制备
第一步
将2-氯嘧啶(280mg,2.44mmol)溶解在无水N,N-二甲基甲酰胺(4mL)和水(1mL)中,加入2-硼酸-5-氟苯甲酸(300mg,1.63mmol),四(三苯基膦)钯(86mg,0.074mmol)和碳酸铯(451mg,1.38mmol),氮气保护下加热至100℃反应2小时。反应液冷却至室温,加入15mL水,过滤,滤液减压浓缩,加入10mL乙醇,过滤除去无机盐,滤液减压浓缩,得到粗产品化合物14a(260mg, 产率73.1%),直接用于下步反应。
MS m/z(ESI):219.1[M+1].
第二步
化合物14b的制备方法,参照化合物11c的制备方法。
MS m/z(ESI):413.2[M+1].
第三步
化合物14c的制备方法,参照化合物11d的制备方法。
MS m/z(ESI):313.1[M+1].
第四步
化合物14的制备方法,参照化合物11的制备方法。
MS m/z(ESI):481.2[M+1].
1H NMR(400MHz,DMSO-d6)δ8.81(d,J=4.8Hz,2H),8.19(dd,J=8.8,5.6Hz,1H),7.42-7.37(m,2H),7.31–7.29(m,1H),5.16(s,1H),3.78-3.67(m,2H),3.62-3.46(m,5H),3.09-2.95(m,3H),2.31(d,J=2.4Hz,3H),1.46(s,6H).
实施例15
化合物15:((3aR,6aS)-5-(5-氟-4-(2-羟基丙烷-2-基)-6-甲基嘧啶-2-基)六氢吡咯[3,4-c]吡咯-2(1H)-基)(2-氟-6-(嘧啶-2-基)苯基)甲酮的制备
第一步
N2保护下,在化合物15a(4g,17.16mmol)和2-(三丁基甲锡烷基)嘧啶(6.97g,18.88mmol)的DMF(60mL)溶液中加入CuI(653.81mg,3.43mmol),Pd(PPh3)4(1.98g,1.71mmol)和CsF(5.21g,34.30mmol)。120℃反应5小时。冷却到室温后,加入150毫升水,用乙酸乙酯(100mL*2)萃取。有机相合并后,用无水硫酸钠干燥,过滤,浓缩。得到的粗品经纯化得到化合物15b(2.8g,产率70.3%)。
MS m/z(ESI):233.1[M+1].
第二步
在化合物15b(800mg,3.45mmol)的水(5.0mL)和甲醇(5.0mL)的混合溶液中加入氢氧化钠(688.98mg,17.22mmol)。25℃反应3小时。加入2N HCl直至pH=2,用乙酸乙酯(20mL*2)萃取。有机相用无水硫酸钠干燥,过滤,旋干,得到化合物15c(650mg,产率86.5%)。
MS m/z(ESI):219.0[M+1].
第三步
在106c(100mg,0.46mmol)和中间体2(129mg,0.46mmol)的乙腈(3.0mL)溶液中加入四甲基氯代脲六氟磷酸酯(257mg,0.92mmol)和N-甲基咪唑(188mg,2.29mmol)。25℃下搅拌30分钟。在反应液中加入20mL水,乙酸乙酯萃取(20mL*2)。有机相合并后,用无水硫酸钠干燥,过滤,浓缩。得到的粗品用制备HPLC纯化,得到化合物15(40mg,产率18.2%)。
MS m/z(ESI):481.2[M+1].
1H NMR(400MHz,DMSO-d6)δ8.85(dd,J=42.9,4.9Hz,2H),8.01(dd,J=36.2,7.8Hz,1H),7.59(td,J=8.1,5.8Hz,1H),7.51–7.37(m,2H),5.15(s,1H),3.85–3.47(m,7H),3.09(dddd,J=36.1,22.9,11.1,7.0Hz,3H),2.36–2.24(m,3H),1.44(d,J=3.3Hz,6H).
生物学测试评价
以下结合测试例进一步描述解释本发明,但这些实施例并非意味着限制本发明的范围。
一、细胞功能实验
测试例1、本发明化合物在稳定表达OX1和OX2受体细胞对钙流影响的测定
1.实验目的:
检测化合物对CHO-K1/human OX1R以及CHO-K1/human OX2R细胞钙流活动的抑制作用。
2.实验仪器和试剂
2.1仪器:
384孔-细胞培养板(Corning:3764);
读板仪FLIPR Tetra(Molecular Device)。
2.2试剂:
DMEM,high glucose(Gibco:12100);
胎牛血清(Biosera:FB-1058/500);
P/S(Biosera:XC-A4122);
5X Matrigel(Corning:354230);
HBSS(Sigma:H1387);
HEPES(Invitrogen:15630080);
Fluo-8AM(AAT Bioquest:21080);
Probenecid(Sigma:P8761);
Pluronic F-127(Sigma:P2443-250G);
1000X Fluo-8AM(2mM):Fluo-8AM溶于DMSO,振荡1-2min,分装后于-20℃保存;
完全培养基:DMEM+10%FBS+1X P/S;
细胞接种培养基:DMEM+10%FBS+1X PS;
实验缓冲液:1X HBSS+20mM HEPES+1mM Probenecid+0.025%Pluronic F-127;
1X Matrigel:用DMEM稀释5X Matrigel;
细胞株:CHO-K1/human OX1R和CHO-K1/human OX2R的细胞系。
3.实验方法:
1)CHO-K1/human OX1R以及CHO-K1/human OX2R细胞株分开培养于完全培养基中,37℃,5%CO2至70%~90%融合度;
2)用1X Matrigel包被384孔-细胞培养板,每孔包被5uL,室温10-30分钟;
3)将细胞消化处理重悬于细胞接种培养基中,分别接种CHO-K1/human OX1R以及CHO-K1/human OX2R细胞株8,000个细胞/孔/20μL至两块384孔-细胞培养板中,于37℃,5%CO2培养24小时;
4)将细胞培养板从CO2培养箱取出,于室温平衡10分钟;
5)取出1000X Fluo-8AM,用平衡至室温的实验缓冲液1稀释成1X Fluo-8AM,浓度2μM;
6)去除细胞培养板的培养基,每孔加入20μL 1X Fluo-8AM,室温300rpm离心60秒后,于室温避光孵育1小时;
7)确定OX1和OX2受体激动剂的EC80(OX-A for OX1R assays,OX-B for OX2R assays):分别配制稀释OX1和OX2受体激动剂工作液,用FLIPR Tetra将稀释的OX1和OX2受体激动剂分别加入CHO-K1/human OX1R以及CHO-K1/human OX2R细胞株相应384孔-细胞培养板的实验孔中,同时读取收集数据,根据下面实验数据处理方法分别得到OX-A和OX-B的EC80;
8)重复操作步骤1-7,配置阳性化合物(suvorexant和seltorexant)以及待测化合物,用FLIPR Tetra将稀释的化合物(11个浓度点)加入相应的两块384-孔细胞培养板的实验孔中15分钟后,根据先前得到的EC80分别在两种细胞株的块 孔板上加入OX1和OX2受体激动剂,同时读取收集数据,确定阳性化合物和待测化合物的IC50。
4.实验数据处理方法:
FLIPR Tetra读取收集荧光信号值(RFU),取最大RFU值,根据Low control(DMSO对照物)和High control(100nM阳性化合物)实验组的读值,计算百分比抑制(激活)的数据{%抑制(激活)率=(RFUsample-RFUlow control)/(RFUhigh control-RFUlow control)×100},使用Prism8拟合百分比抑制(激活)率和11个点浓度数据至参数非线性逻辑公式计算出化合物的IC50值。
5.实验结果:
本发明的化合物在稳定表达OX2受体受体细胞对钙流影响实验中显示出良好的抑制作用,且化合物对OX2受体的抑制效果显著优于OX1受体,具有较好的选择性。
二、药代动力学评价试验
大鼠药代动力学评价试验
1.研究目的:
以SD大鼠为受试动物,研究本发明化合物,在5mg/kg剂量下口服给药在大鼠体内(血浆)的药代动力学行为。
2.实验方案:
2.1实验药品:
本发明实施例化合物,自制。
2.2实验动物:
SD大鼠每组3只,雄性,上海杰思捷实验动物有限公司,动物生产许可证号(SCXK(沪)2013-0006 N0.311620400001794)。
2.3制剂处方:
口服给药药物配制:20%HP-β-CD in Water
称取20g HP-β-CD粉末,溶于100mL纯净水,涡旋、混匀、超声,得到澄 清溶液。
称取实施例化合物加入20mL玻璃瓶中,加入该溶液,超声10分钟,得到无色澄清溶液,浓度为0.5mg/mL。
2.4给药:
SD大鼠3只,雄性,禁食一夜后分别p.o.;
p.o.剂量为5mg/kg,给药体积10mL/kg。
2.5样品采集:
血液采集:大鼠于给药前和给药后0.25、0.5、1、2、4、6、8、24h颈静脉采血0.2mL,置于EDTA-K2抗凝管中,4℃6000rpm离心6min分离血浆,于-80℃保存;给药后4h进食。
脑组织采集:实验动物CO2安乐死后,于给药后0.25h、0.5h、1h、2h、4h、6h、8h、24h采集脑组织,脑组织用预冷PBS冲洗干净后,擦干,称重,于-80℃保存。
2.6样品处理:
1)血浆样品40μL加入160μL乙腈沉淀,混合后3500×g离心5~20分钟。
2)取处理后上清溶液进行LC/MS/MS分析待测化合物的浓度,LC/MS/MS分析仪器:AB Sciex API 4000Qtrap。
2.7液相分析:
●液相条件:Shimadzu LC-20AD泵
●色谱柱:Agilent ZORBAX XDB-C18(50×2.1mm,3.5μm)移动相:
A液为0.1%甲酸水溶液,B液为乙腈
●流速:0.4mL/min
●洗脱时间:0-4.0分钟,洗脱液如下:
3.试验结果与分析:
药代动力学主要参数用WinNonlin 6.1计算得到,大鼠药代实验结果如下所示:
4.实验结论:
数据显示,在大鼠药代动力学评价实验中,本发明实施例化合物口服给药后显示出较高的暴露量,尤其是在脑中的暴露量较高。
三、遥测脑电和肌电睡眠技术评价本发明化合物对大鼠睡眠结构的作用1.实验目的
本实验利用DSI公司的遥测脑电和肌电系统,检测本发明化合物对SD大鼠睡眠结构的影响。
2.实验仪器与试剂
2.1.主要仪器
DSI遥感测压系统:植入子型号:TL11M2-F40-EET,Data Sciences International.HD-S02,Data Sciences International.
数据采集软件:Ponemah Software 5.0,Data Sciences International.
数据分析软件:NeuroScore,Data Sciences International.
精密电子天平
电子天平
超声仪
恒温磁力搅拌器
漩涡仪
2.2.主要试剂
HP-β-CD
3.实验过程
3.1.实验动物购买和适应
SD大鼠,动物到设施时体重为190~210g(5-6W)。动物到达设施后,将其饲养于严格控制环境条件的动物饲养间中,适应期为7-9天。适应期内,将实验动物置于12h明暗交替环境中适应调整节律时间,每天监测动物的健康状况。
3.2.分组
所有动物按体重在到达动物设施,经适应性饲养,手术植入电极,只数满足每组至少6只,符合统计学检验的要求及药效学指导原则的要求。
3.3.实验流程
1)手术植入电极:将实验动物置于12h明暗交替环境中适应7-9天(关灯:07:00;开灯:19:00)。实验当天用舒泰(i.p.,20mg/kg),联合噻拉嗪(i.p.,8mg/kg)麻醉动物,麻醉后,脑立体定位仪固定,头部手术区域备皮,颅骨钻孔并埋植电极。同时将2根肌电电极分别平行穿入颈部肌肉内,并用缝合线固定两端,避免其末端相互接触。然后植入子体置于背部皮下,对手术伤口缝合并进行消毒。
2)术后护理:术后将大鼠小心放入干净恢复笼内,侧卧位,保证呼吸道通畅。12h明暗自动交替(关灯:07:00,开灯:19:00),恒温20-26℃,相对湿度40-70%。手术后给予动物3天护理,手术切口局部给予头孢拉定粉末处理,皮下给予庆大霉素4~8mg/kg,并连续3天皮下注射美洛昔康0.1ml/只,恢复7-10天后进行实验,并根据动物体重随机分组。
3)给药方案及指标监测:术后7-10天进行基础脑电、肌电记录,基础脑电、肌电记录完成后开始给药,连续给药七天。在给药第1天,第7天,当天11:00开始进行给药前1h脑电、肌电记录,并于12:00给药,之后进行给药后24h脑电、肌电记录。
4)实验指标:给药后,不同组别之间NREM、REM的潜伏期时间变化;给药后24小时内Wake、NREM和REM的时间变化。
4.数据采集及分析
原始数据由DSI系统Ponemah软件采集,用NeuroScore软件和GraphPad Prism软件分析。实验数据由平均值±标准误(Mean±SEM)表示,采用ANOVA进行统 计分析,P<0.05表示有显著性差异。
5.实验结果
溶媒对照组动物清醒/睡眠状态分布符合昼夜节律,未见溶媒对动物睡眠结构有明显影响,且在不同清醒/睡眠阶段的分布符合文献报道情况,模型成功且稳定。
对vehicle组及给药组每只动物在给药前1h至给药后不同睡眠阶段进行分析,以1h为单位取各组均值,分析各组在不同睡眠阶段的时间分布变化。
对关灯阶段,对给药后1h、3h、5h、7h,动物在不同睡眠阶段的总时间长度进行统计,分析不同给药组间药效影响。
对开灯阶段,对开灯后后1h、3h、5h、7h,动物在不同睡眠阶段的总时间长度进行统计,分析不同给药组间药效影响。
6.实验结论
本发明实施例化合物在较低的剂量即可显著降低NREM/REM潜伏期和清醒总时长;显著增加NREM总时长,对总REM时间无影响,嗜睡风险低。可见本发明化合物对大鼠有良好的促进睡眠作用,并且嗜睡风险低。
四、CYP酶单点抑制试验
1.实验目的
采用人肝微粒体孵育体系,利用单点法快速预测化合物对CYP450酶亚型的抑制情况。
2.实验步骤
2.1溶液配制
称取NADPH(还原型烟酰胺腺嘌呤二核苷酸磷酸)加100mM磷酸缓冲液,终浓度为2.5mM。50μL 20mg/mL微粒体,加4mL 100mM磷酸缓冲液,混匀得0.25mg/mL微粒体。
待测化合物反应液的配制:
称取待测实施例化合物,用DMSO稀释至10mM,再用100mM磷酸缓冲液稀释至100μM。
2.2实验流程:
1.在96孔板中,加入40μL肝微粒体、10μL底物、10μL待测化合物,预孵育3min。
2.加入NADPH 40μL。
3.在20min时加入300μL含有内标的乙腈终止液。
4.离心进样。
3.实验结果:
晶型研究
1.1实验仪器
1.1.1物理化学检测仪器的一些参数

1.1.2色谱条件
1.不同晶型的制备
1.1化合物9自由碱晶型I的制备
称量约20mg的自由碱到2mL的玻璃瓶中,加入100-200μL有机溶剂(有机溶剂可选MeOH、EtOH、EA、Acetone、IPA、IPAc、MEK、2-Me-THF、、MTBE、toluene、cyclohexane、heptane),在50℃下打浆7天,所得固体离心,去除上清液,40℃真空干燥,得到自由碱晶型I。经检测分析,其有如下如图1所示的XRPD图、如图2所示的DSC图及如图3所示的TGA图。
1.2化合物9自由碱晶型I的制备(工艺制备方法)
1)用20mL甲醇将15.3g粗品溶清,滴加30mL水,搅拌20min,析出固体,再滴加6mL水(甲醇:水=1:1.8),搅拌16h。
2)过滤,4mL甲醇+12mL水混合,洗滤饼,40℃干燥得浅褐色固体;
3)7.7g固体加入23mL甲醇中,加热到外温65℃,溶清,冷却到20℃搅拌0.5h,析出固体,滴加23mL水,搅拌4h。
4)过滤,10mL甲醇+30mL水混合,洗滤饼,40℃干燥得浅褐色固体;
5)7.3g固体加入22mL(3V)EA中,加热到回流搅拌1h,滴加22mL(3V)正庚烷,继续搅拌0.5h,冷却到20~24℃搅拌16h。
6)过滤,4mLEA+8mL正庚烷混合,洗滤饼,40℃干燥得6.5g浅褐色固体,得到自由碱晶型I。
1.3化合物9自由碱晶型II的制备
称量约20mg的自由碱到2mL的玻璃瓶中,加入50-300μL溶剂(溶剂可选ACN、THF、DCM、water),在50℃下打浆7天,在这个过程中所得澄清溶液在室温下挥发,所得固体离心,去除上清液,40℃真空干燥,得到自由碱晶型II。经检测分析,其有如下如图4所示的XRPD图、如图5所示的DSC图及如图6所示的TGA图。
2.固体稳定性实验
2.1实验目的:
考察自由碱晶型I、晶型II在高温60℃、高湿92.5%RH、高温高湿50℃75%RH条件下,化合物的物理化学稳定性,为化合物贮存提供依据。
2.2实验方案:
分别取自由碱晶型I和晶型II约2mg,于高温60℃、高湿92.5%RH、高温高湿50℃75%RH条件下,考察晶型在7天和14天的有关物质的变化情况,用HPLC,采用色谱峰面积归一化法计算有关物质的变化。
色谱条件:

2.3实验结果:
自由碱晶型I物理化学稳定性结果:
自由碱晶型II物理化学稳定性结果:

以上数据表明,化合物9自由碱晶型I和自由碱II在放置7天和14天后所有条件下均较稳定。
3引湿性实验
3.1实验目的
考察化合物9晶型I和晶型II在不同相对湿度条件下的引湿性,为化合物贮存提供依据。
3.2实验方案:
将化合物9晶型I和晶型II置不同相对湿度的饱和水蒸气中,使化合物与水蒸气达到动态平衡,并计算平衡后化合物吸湿增重的百分数。
3.3实验结果:
自由碱晶型I在80%RH条件下吸湿增重约0.3263%,略有引湿性。经0-95%相对湿度条件下吸湿与解吸湿循环2次,自由碱晶型I的XRPD谱图并未发生改变,即晶型未转变。
自由碱晶型II在80%RH条件下吸湿增重约0.2053%,略有引湿性。
4.不同介质中溶解度实验
4.1实验目的
比较自由碱晶型I和晶型II在pH1.0-8.0USP缓冲液、人工模拟胃液(FaSSGF)、禁食人工模拟肠液(FaSSIF)、非禁食人工模拟肠液(FeSSIF)以及纯水等媒介中溶解度大小,为可成药性评估提供依据。
4.2实验方案:
分别将约1mg自由碱晶型I和晶型II混悬至不同介质中2小时,用HPLC,外标法测定化合物37℃下的热力学溶解度。
4.3实验结果:如表3所示
表3

由以上实验结果可知,自由碱晶型I在各介质体系中的热力学溶解度均较晶型II的溶解度高,且晶型I的溶解度均满足局部治疗所需的药物浓度。
5.动物PK研究
5.1实验目的:
以SD大鼠为受试动物,研究自由碱晶型I及自由碱晶型II单次口服给药在大鼠体内(血浆)的药代动力学行为,比较暴露量的变化;研究自由碱晶型单次静脉给药的药代动力学,计算自由碱晶型I及自由碱晶型II口服给药的生物利用度。
5.2实验方案:
自由碱晶型I用含0.5%的0.5%CMCC-Na水溶液混悬,自由碱晶型I用含0.5%的0.5%HPMC水溶液混悬,均匀后,灌胃,大鼠给药,平行三只大鼠,给药剂量为自由碱晶型I(5mg/kg,30mg/kg为混悬液)、自由碱晶型II(30mg/kg为混悬液);自由碱晶型I用含20%HP-β-CD的水溶液溶清,滤膜过滤后,注射,大鼠给药,平行三只大鼠。5.3实验结果
表4自由碱晶型I和晶型II的动物PK实验结果

从以上数据可以看出,自由碱晶型I较自由碱晶型II的暴露量较高,晶型I 30mg/kg的暴露量比5mg/kg的暴露量明显较高。

Claims (10)

  1. 通式(I)所示化合物的晶型,其结构如下所示:
    其中:
    X1为CR6或N;
    X2为CR6或不存在;
    R1选自卤素;
    R2、R3、R4或R6分别独立地选自氢、氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、羟基烷氧基、烯基、炔基、环烷基、杂环基、芳基或杂芳基,所述的氨基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、羟基烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,任选可以进一步被取代;
    R2、R3、R4或R6各自独立地优选氢、氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、C1-8烷基、C1-8氘代烷基、C1-8卤代烷基、C1-8羟烷基、C1-8烷氧基、C1-8卤代烷氧基、C1-8羟基烷氧基、C2-8烯基、C2-8炔基、C3-12环烷基、3-12元杂环基、C6-14芳基或5-14元杂芳基,所述的氨基、C1-8烷基、C1-8氘代烷基、C1-8卤代烷基、C1-8羟烷基、C1-8烷氧基、C1-8卤代烷氧基、C1-8羟基烷氧基、C2-8烯基、C2-8炔基、C3-12环烷基、3-12元杂环基、C6-14芳基和5-14元杂芳基,任选进一步被氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、C1-8烷基、C1-8氘代烷基、C1-8卤代烷基、C1-8羟烷基、C1-8烷氧基、C1-8卤代烷氧基、C2-8烯基、C2-8炔基、C3-12环烷基、3-12元杂环基、C6-14芳基和5-14元杂芳基中的一个或多个取代基所取代;
    更优选氢、氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、C1-6烷基、C1-6氘代烷基、C1-6卤代烷基、C1-6羟烷基、C1-6烷氧基、C1-6卤代烷氧基、C1-6羟基烷氧基、C2-6烯基、C2-6炔基、C3-8环烷基、3-8元杂环基、C6-10芳基或5-10元杂芳基,所述的氨基、C1-6烷基、C1-6氘代烷基、C1-6卤代烷基、C1-6羟烷基、C1-6烷氧基、C1-6卤代烷氧基、C1-6羟基烷氧基、C2-6 烯基、C2-6炔基、C3-8环烷基、3-8元杂环基、C6-10芳基和5-10元杂芳基,任选进一步被氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、C1-6烷基、C1-6氘代烷基、C1-6卤代烷基、C1-6羟烷基、C1-6烷氧基、C1-6卤代烷氧基、C2-6烯基、C2-6炔基、C3-8环烷基、3-8元杂环基、C6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
    或者,R2、R3、R4中的任意两个或多个与相连的原子连接形成环烷基、杂环基、芳基或杂芳基,所述环烷基、杂环基、芳基和杂芳基,任选可以进一步被取代;
    R5选自羟烷基或卤代烷基,任选可以进一步被取代,所述羟烷基优选为所述卤代烷基优选为
    X’为卤素;
    R7分别独立地选自氢、氘、卤素、氨基、硝基、羟基、巯基、氰基、羧基、磺酸基、氧代基、硫代基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、羟基烷氧基、烯基、炔基、环烷基、杂环基、芳基或杂芳基,所述的氨基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、羟基烷氧基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,任选可以进一步被取代。
  2. 根据权利要求1所述的晶型,其特征在于,所述通式(I)进一步如通式(II)所示:
    其中,
    X1为CH或N;
    X2为CH或不存在;
    R2、R3或R4分别独立地选自氢、氘、卤素、氨基、羟基、C1-3烷基、C1-3氘代烷基、C1-3卤代烷基、C1-3羟烷基、C1-3烷氧基、C1-3卤代烷氧基或C1-3羟基烷氧基;
    R5选自
  3. 根据权利要求1或2所述的晶型,其特征在于,
    当X2不存在时,取代基R2优选为卤素,所述卤素优选位于三氮唑的邻位或间位,更优选位于邻位,
    所述卤素优选为氟;
    当X2为CR6时,取代基R2优选为卤素,所述卤素优选位于嘧啶的邻位或间位,更优选位于嘧啶的间位,和/或连接羰基的邻位,
    所述卤素优选为氟。
  4. 根据权利要求1所述的晶型,其特征在于,所述通式(I)选自以下化合物:
  5. ((3aR,6aS)-5-(5-氟-4-(2-羟基丙-2-基)嘧啶-2-基)六氢吡咯并[3,4-c]吡咯-2(1H)-基)(2-氟-6-(嘧啶-2-基)苯基)甲酮的晶型,包括晶型I和晶型II,其中:
    晶型I的粉末X射线衍射图谱在2θ为8.6±0.2°处具有衍射峰,或者在2θ为9.2±0.2°处具有衍射峰,或者在2θ为10.2±0.2°处具有衍射峰,或者在2θ为10.8±0.2°处具有衍射峰,或者在2θ为12.0±0.2°处具有衍射峰,或者在2θ为12.8±0.2°处具有衍射峰,或者在2θ为13.7±0.2°处具有衍射峰,或者在2θ为14.2±0.2°处具有衍射峰,或者在2θ为15.5±0.2°处具有衍射峰,或者在2θ为16.6±0.2°处具有衍射峰,或者在2θ为17.3±0.2°处具有衍射峰,或者在2θ为18.4±0.2°处具有衍射峰,或者在2θ为19.0±0.2°处具有衍射峰,或者在2θ为 19.5±0.2°处具有衍射峰,或者在2θ为20.5±0.2°处具有衍射峰,或者在2θ为21.3±0.2°处具有衍射峰,或者在2θ为26.5±0.2°处具有衍射峰;或者在2θ为28.6±0.2°处具有衍射峰;优选的,包含其中任选的2处、4处、6处、8处或10处有衍射峰;
    晶型II的粉末X射线衍射图谱在2θ为9.9±0.2°处具有衍射峰,或者在2θ为10.8±0.2°处具有衍射峰,或者在2θ为13.2±0.2°处具有衍射峰,或者在2θ为14.9±0.2°处具有衍射峰,或者在2θ为16.4±0.2°处具有衍射峰,或者在2θ为17.1±0.2°处具有衍射峰,或者在2θ为17.9±0.2°处具有衍射峰,或者在2θ为19.3±0.2°处具有衍射峰,或者在2θ为19.8±0.2°处具有衍射峰,或者在2θ为20.2±0.2°处具有衍射峰,或者在2θ为20.5±0.2°处具有衍射峰,或者在2θ为21.0±0.2°处具有衍射峰,或者在2θ为21.9±0.2°处具有衍射峰,或者在2θ为25.8±0.2°处具有衍射峰,或者在2θ为26.5±0.2°处具有衍射峰,或者在2θ为27.5±0.2°处具有衍射峰;优选的,包含其中任选的2处、4处、6处、8处或10处有衍射峰。
  6. 根据权利要求5所述的晶型,其特征在于,晶型I的粉末X射线衍射图谱至少包含位于2θ为9.2±0.2°、10.2±0.2°、16.6±0.2°或18.4±0.2°中的一处或多处衍射峰;优选包含其中2-4处,更优选包含3-4处,最优选包含4处;任选的,进一步,还可以包含2θ为12.8±0.2°、17.3±0.2°、19.0±0.2°、21.3±0.2°或26.5±0.2°中的一处或多处衍射峰,优选包含其中2处、3处、4处或5处;
    晶型II的粉末X射线衍射图谱至少包含位于2θ为16.4±0.2°、17.1±0.2°、17.9±0.2°或25.8±0.2°中的一处或多处衍射峰;优选包含其中2-4处,更优选包含3-4处,最优选包含4处;任选的,进一步,还可以包含2θ为10.8±0.2°、14.9±0.2°、19.3±0.2°、21.9±0.2°或26.5±0.2°中的一处或多处衍射峰,优选包含其中2处、3处、4处或5处。
  7. 根据权利要求5所述的晶型,其特征在于,晶型I的X-射线粉末衍射图谱在2θ为9.2±0.2°和18.4±0.2°处具有特征峰;优选地,还包含在2θ为10.2±0.2°、16.6±0.2°、17.3±0.2°和19.0±0.2°处具有特征峰;更优选地,还包含在2θ为12.8±0.2°和21.3±0.2°处具有特征峰;进一步优选地,还包含在2θ为、26.5±0.2°和28.6±0.2°处具有特征峰,更一步优选的,还包含在8.6±0.2°、10.8±0.2°、12.0±0.2°、13.7±0.2°、14.2±0.2°、15.5±0.2°、19.5±0.2°、20.5±0.2°、23.1±0.2°、27.8±0.2°和32.1±0.2°处具有特征峰;最优选地,其X-射线粉末衍射图谱基本如图1所示;其DSC图谱基本如图 2所示,其TGA图谱基本如图3所示;
    晶型II的X-射线粉末衍射图谱在2θ为16.4±0.2°和25.8±0.2°处具有特征峰;优选地,还包含在2θ为17.1±0.2°、17.9±0.2°、21.9±0.2°和26.5±0.2°处具有特征峰;更优选地,还包含在2θ为10.8±0.2°和14.9±0.2°处具有特征峰;进一步优选地,还包含在2θ为9.9±0.2°和19.3±0.2°处具有特征峰,更进一步优选的,还包含在13.2±0.2°、19.8±0.2°、20.2±0.2°、20.5±0.2°、21.0±0.2°和27.5±0.2°处具有特征峰;最优选地,其X-射线粉末衍射图谱基本如图4所示;其DSC图谱基本如图5所示,其TGA图谱基本如图6所示。
  8. 一种制备权利要求1-7任意一项所述化合物的晶型的方法,具体包括如下步骤:
    1)称取适量的自由碱,用不良溶剂混悬,悬浮密度优选为50~200mg/mL;
    2)以上所得混悬液在经振摇后,温度优选0~50℃,时间优选1~10天;
    3)将以上混悬液快速离心,去除上清液,剩余固体烘干至恒重得到目标产物;
    其中:
    所述的不良溶剂选自丙酮、乙酸乙酯、醋酸异丙酯、乙腈、乙醇、四氢呋喃、2-甲基四氢呋喃、二氯甲烷、1,4-二氧六环、苯、甲苯、异丙醇、正丁醇、异丁醇、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、正丙醇、叔丁醇、2-丁酮或3-戊酮、甲基叔丁基醚、水、庚烷或正戊烷;优选醋酸异丙酯、甲苯、甲基叔丁基醚、水、庚烷或正戊烷;
    或者,
    1)称取适量的自由碱,用良溶剂溶解;
    2)向以上所得溶液中加入反溶剂,搅拌至固体析出,温度优选0~25℃;
    3)将以上混悬液快速离心,去除上清液,剩余固体烘干至恒重得到目标产物;
    其中:
    所述的良溶剂选自甲醇、丙酮、乙酸乙酯、乙腈、乙醇、四氢呋喃、二氯甲烷、1,4-二氧六环、苯、甲苯、异丙醇、正丁醇、异丁醇、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、正丙醇、叔丁醇、2-丁酮或3-戊酮;优选甲醇、二氯甲烷或乙酸乙酯。
    所述的反溶剂选自庚烷、水、甲基叔丁基醚、环己烷或乙酸异丙酯;
    或者,
    1)称取适量的自由碱,用良溶剂加热溶解;
    2)将以上所得溶液迅速置于低温下,搅拌至固体析出,温度优选-10~5℃;
    3)将以上混悬液快速离心,去除上清液,剩余固体烘干至恒重得到目标产物;
    其中:
    所述的良溶剂选自甲醇、丙酮、乙酸乙酯、乙腈、乙醇、四氢呋喃、二氯甲烷、1,4-二氧六环、苯、甲苯、异丙醇、正丁醇、异丁醇、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、正丙醇、叔丁醇或2-丁酮或四氢呋喃;优选丙酮。
  9. 一种药物组合物,其含有治疗有效量的权利要求1-7中任一项所述化合物的晶型,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  10. 根据权利要求1-7中任一项所述的晶型或权利要求9所述的药物组合物在作为食欲素受体拮抗剂在制备治疗神经系统疾病药物中的应用,优选在制备OX2R选择性受体拮抗剂中的应用;
    更优选地,所述神经系统疾病选自失眠、抑郁、焦虑或药物成瘾,更优选为重度抑郁症、原发和继发性失眠或伴随失眠的抑郁症。
PCT/CN2023/118743 2022-09-14 2023-09-14 含氮杂环的多环化合物的自由碱晶型及其制备方法 WO2024056016A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211116574.6 2022-09-14
CN202211116574 2022-09-14

Publications (1)

Publication Number Publication Date
WO2024056016A1 true WO2024056016A1 (zh) 2024-03-21

Family

ID=90274319

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/118743 WO2024056016A1 (zh) 2022-09-14 2023-09-14 含氮杂环的多环化合物的自由碱晶型及其制备方法

Country Status (1)

Country Link
WO (1) WO2024056016A1 (zh)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102781942A (zh) * 2009-10-23 2012-11-14 詹森药业有限公司 用作食欲肽受体调节剂的二取代八氢吡咯并[3, 4-c]吡咯
CN106749269A (zh) * 2015-11-23 2017-05-31 广东东阳光药业有限公司 八氢吡咯并[3,4‑c]吡咯衍生物及其用途
CN108299437A (zh) * 2017-01-13 2018-07-20 广东东阳光药业有限公司 八氢吡咯并[3,4-c]吡咯衍生物及其用途
CN108883110A (zh) * 2016-03-10 2018-11-23 詹森药业有限公司 使用食欲素-2受体拮抗剂治疗抑郁症的方法
CN109988170A (zh) * 2017-12-29 2019-07-09 广东东阳光药业有限公司 八氢吡咯并[3,4-c]吡咯衍生物及其用途
WO2020100011A1 (en) * 2018-11-14 2020-05-22 Janssen Pharmaceutica Nv Improved synthetic methods of making fused heterocyclic compounds as orexin receptor modulators
WO2022194122A1 (zh) * 2021-03-16 2022-09-22 上海翰森生物医药科技有限公司 含氮杂环的多环化合物及其制备方法和应用

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102781942A (zh) * 2009-10-23 2012-11-14 詹森药业有限公司 用作食欲肽受体调节剂的二取代八氢吡咯并[3, 4-c]吡咯
CN106749269A (zh) * 2015-11-23 2017-05-31 广东东阳光药业有限公司 八氢吡咯并[3,4‑c]吡咯衍生物及其用途
CN108883110A (zh) * 2016-03-10 2018-11-23 詹森药业有限公司 使用食欲素-2受体拮抗剂治疗抑郁症的方法
CN108299437A (zh) * 2017-01-13 2018-07-20 广东东阳光药业有限公司 八氢吡咯并[3,4-c]吡咯衍生物及其用途
CN109988170A (zh) * 2017-12-29 2019-07-09 广东东阳光药业有限公司 八氢吡咯并[3,4-c]吡咯衍生物及其用途
WO2020100011A1 (en) * 2018-11-14 2020-05-22 Janssen Pharmaceutica Nv Improved synthetic methods of making fused heterocyclic compounds as orexin receptor modulators
WO2022194122A1 (zh) * 2021-03-16 2022-09-22 上海翰森生物医药科技有限公司 含氮杂环的多环化合物及其制备方法和应用

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DEFLORIAN FRANCESCA, PEREZ-BENITO LAURA, LENSELINK EELKE B, CONGREVE MILES, VAN VLIJMEN HERMAN W. T., MASON JONATHAN S., GRAAF CHR: "Accurate Prediction of GPCR Ligand Binding Affinity with Free Energy Perturbation", JOURNAL OF CHEMICAL INFORMATION AND MODELING, AMERICAN CHEMICAL SOCIETY , WASHINGTON DC, US, vol. 60, no. 11, 23 November 2020 (2020-11-23), US , pages 5563 - 5579, XP055968396, ISSN: 1549-9596, DOI: 10.1021/acs.jcim.0c00449 *
MICHAEL A. LETAVIC, PASCAL BONAVENTURE, NICHOLAS I. CARRUTHERS, CHRISTINE DUGOVIC, TATIANA KOUDRIAKOVA, BRIAN LORD, TIMOTHY W. LOV: "Novel Octahydropyrrolo[3,4- c ]pyrroles Are Selective Orexin-2 Antagonists: SAR Leading to a Clinical Candidate", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 58, no. 14, 23 July 2015 (2015-07-23), US , pages 5620 - 5636, XP055493378, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.5b00742 *
SONGLIANG WU, YU SUN, YI HU, HONGMEI ZHANG, LIJUAN HOU, XING LIU, YUFENG LI, HAIYING HE, ZHI LUO, YUAN CHEN, YUHE WANG, WEIHUA SHI: "Discovery of novel substituted octahydropyrrolo[3,4- c ]pyrroles as dual orexin receptor antagonists for insomnia treatment", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, ELSEVIER, AMSTERDAM NL, vol. 27, no. 6, 1 March 2017 (2017-03-01), Amsterdam NL , pages 1458 - 1462, XP055493385, ISSN: 0960-894X, DOI: 10.1016/j.bmcl.2017.01.075 *

Similar Documents

Publication Publication Date Title
JP7053760B2 (ja) ハンチントン病の治療方法
WO2021197464A1 (zh) 稠合咪唑类衍生物、其制备方法及其在医药上的应用
KR20140105445A (ko) 비아릴 에테르 술폰아미드 및 치료제로서의 그의 용도
WO2020238791A1 (zh) 氢化吡啶并嘧啶类衍生物、其制备方法及其在医药上的应用
TWI718113B (zh) 吡啶甲醯胺類衍生物、其製備方法及其在醫藥上的應用
WO2021143680A1 (zh) 杂芳基类衍生物及其制备方法和用途
TW202108582A (zh) 含二並環類衍生物抑制劑、其製備方法和應用
WO2016169421A1 (zh) 咪唑并异吲哚类衍生物、其制备方法及其在医药上的应用
TW201522330A (zh) 2-醯胺噻唑衍生物或其鹽
WO2017071516A1 (zh) 一种蛋白激酶抑制剂及其制备方法和医药用途
WO2021197452A1 (zh) 含氮杂芳类衍生物自由碱的晶型
WO2021249519A1 (zh) 吡啶酮并嘧啶类衍生物、其制备方法及其在医药上的应用
WO2021249475A1 (zh) 稠合喹唑啉类衍生物、其制备方法及其在医药上的应用
WO2023025320A1 (zh) 含氮杂环类衍生物抑制剂、其制备方法和应用
ES2379734T3 (es) Derivado de pirrol o sal del mismo
CN113493440A (zh) 含氮杂芳类衍生物的盐及其晶型
WO2022166860A1 (zh) Pim激酶抑制剂
WO2019062657A1 (zh) 氮杂环类衍生物、其制备方法及其医药用途
WO2021115225A1 (zh) 含吡唑多环类衍生物抑制剂、其制备方法和应用
WO2024056016A1 (zh) 含氮杂环的多环化合物的自由碱晶型及其制备方法
WO2021027647A1 (zh) 桥杂环基取代的嘧啶类化合物及其制备方法和医药用途
WO2022100738A1 (zh) 含二并环类衍生物抑制剂自由碱的晶型及其制备方法和应用
WO2023066363A1 (zh) Parp-1降解剂及其用途
WO2023125708A1 (zh) 一种p38 MAPK/MK2通路调节剂及其组合物、制备方法和用途
WO2022194122A1 (zh) 含氮杂环的多环化合物及其制备方法和应用

Legal Events

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

Ref document number: 23864748

Country of ref document: EP

Kind code of ref document: A1