WO2020224626A1 - 用作激酶抑制剂的化合物及其应用 - Google Patents

用作激酶抑制剂的化合物及其应用 Download PDF

Info

Publication number
WO2020224626A1
WO2020224626A1 PCT/CN2020/089067 CN2020089067W WO2020224626A1 WO 2020224626 A1 WO2020224626 A1 WO 2020224626A1 CN 2020089067 W CN2020089067 W CN 2020089067W WO 2020224626 A1 WO2020224626 A1 WO 2020224626A1
Authority
WO
WIPO (PCT)
Prior art keywords
mono
substituted
compound
alkyl
poly
Prior art date
Application number
PCT/CN2020/089067
Other languages
English (en)
French (fr)
Other versions
WO2020224626A9 (zh
Inventor
李钧
牛成山
梁阿朋
吴豫生
Original Assignee
浙江同源康医药股份有限公司
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
Priority to EP20802185.7A priority Critical patent/EP3967696A4/en
Priority to KR1020217040056A priority patent/KR20220007111A/ko
Priority to AU2020270303A priority patent/AU2020270303B2/en
Priority to CN202080004473.2A priority patent/CN112867717B/zh
Application filed by 浙江同源康医药股份有限公司 filed Critical 浙江同源康医药股份有限公司
Priority to JP2021566283A priority patent/JP7420403B2/ja
Priority to BR112021022255A priority patent/BR112021022255A2/pt
Priority to MX2021013576A priority patent/MX2021013576A/es
Priority to CA3142088A priority patent/CA3142088C/en
Priority to SG11202112381VA priority patent/SG11202112381VA/en
Publication of WO2020224626A1 publication Critical patent/WO2020224626A1/zh
Publication of WO2020224626A9 publication Critical patent/WO2020224626A9/zh
Priority to TW110106655A priority patent/TW202142541A/zh
Priority to US17/521,153 priority patent/US20220089603A1/en
Priority to IL287908A priority patent/IL287908A/en
Priority to ZA2021/09737A priority patent/ZA202109737B/en

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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • 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
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention relates to the technical field of medicine, in particular to a compound used as a tropomyosin receptor kinase inhibitor, a preparation method thereof, and application in the preparation of drugs for treating ROS1, NTRK, ALK and other kinase-mediated diseases.
  • the tropomyosin receptor kinase (TRK) family belongs to the transmembrane receptor tyrosine kinases (RTKs), which are involved in regulating the synaptic growth and functional maintenance of the mammalian nervous system, the occurrence and development of memory, and the protection of neurons from damage. .
  • TRK kinase is a type of nerve growth factor receptor, and its family consists of highly homologous tropomyosin-related kinase A (TRKA), tropomyosin-related kinase B (Tropomyosin-related kinase B, TRKB), tropomyosin-related kinase C (Tropomyosin-related kinase C, TRKC), which are respectively encoded by NTRK1, NTRK2 and NTRK3 genes.
  • TRKA tropomyosin-related kinase A
  • TRKB tropomyosin-related kinase B
  • TRKC tropomyosin-related kinase C
  • the complete TRK kinase includes three parts: the extracellular domain, the transmembrane domain, and the intracellular domain.
  • TRK kinase Like other RTKs, the extracellular domain of TRK kinase combines with the corresponding ligand to form a dimer, which can cause TRK kinase Autophosphorylation occurs in the intracellular region to activate its own kinase activity and further activate the downstream signal transduction pathway. TRK kinase affects cell proliferation, differentiation, metabolism and apoptosis through downstream pathways such as Ras/MAPK, PI3K/AKT and PLC ⁇ .
  • the NTRKs gene When the NTRKs gene is fused or mutated, it will change or eliminate the extracellular domain receptor (Greco, A.et.al, Mol.Cell.Biol.1995,15,6118; Oncogene 1998,16,809), and the fusion or mutation TRK protein is in a highly activated kinase activity state without ligand binding, which can continuously activate downstream signal transduction pathways, which can lead to abnormal regulation of TRK kinase downstream signal pathways, induce cell proliferation, and promote The occurrence and development of tumors.
  • NTRKs gene fusion occurs in a variety of adult and child solid tumors, including breast cancer, colorectal cancer, non-small cell lung cancer, papillary thyroid cancer, Spitz-like melanoma, glioma, and various sarcomas.
  • breast cancer colorectal cancer
  • non-small cell lung cancer papillary thyroid cancer
  • Spitz-like melanoma glioma
  • various sarcomas various sarcomas.
  • the incidence of NTRK gene fusion is relatively low, about 1%-3%, but in some rare cancers, such as infantile fibrosarcoma, breast secretion Type cancer, etc., the incidence of NTRK gene fusion can reach more than 90%.
  • the earliest TPM3-TRKA fusion protein was found in colon cancer cells.
  • NTRK fusion protein has become an effective anti-cancer target and a hot spot in the research and development of anti-cancer drugs.
  • TRK fusion protein types and mutation types have been discovered (Russo, M.et.al Cancer Discovery, 2016, 6, 36; Drilon, A.et.al ,Annals of Oncology,2016,27,920), so there is an urgent need for clinical development of new NTRK inhibitors with better activity and wider effects, so as to solve the problem of tumor treatment caused by these NTRK protein fusions or mutations.
  • ROS1 c-ros oncogene 1 receptor kinase
  • ROS1 c-ros oncogene 1 receptor kinase
  • the acid kinase active region, transmembrane region and extracellular region are composed of three parts, which encode chimeric proteins with tyrosine kinase activity.
  • the basic structure consists of the extracellular N-terminal ligand binding domain (amino acids 1-1861), the transmembrane domain (amino acids 1862-1882) and the intracellular C-terminal 464 amino acid tyrosine kinase active region (amino acids 1883-2347). )composition.
  • the ROS1 gene When the ROS1 gene is rearranged, the extracellular region is lost, and the transmembrane region and intracellular tyrosine kinase region are retained.
  • the rearrangement site mainly occurs in the 32-36 exons of the ROS1 gene. Mutations in the ROS1 gene mainly occur in lung cancer patients, with a proportion of 1%-2%.
  • the ROS1 gene is mainly fused with SLC34A2 and CD74, and continuously activates the ROS1 tyrosine kinase domain and downstream signal pathways such as JAK/STAT, PI3K/AKT, RAS/MAPK, etc., which in turn causes the occurrence of tumors. It has been confirmed in a large number of literatures and clinically that by inhibiting the activity of mutant ROS1 kinase, it is possible to treat diseases caused by overactivation of ROS1, especially cancer.
  • the currently marketed therapeutic drugs for ROS1-positive non-small cell lung cancer are crizotinib and entritinib, both of which belong to the first generation of small molecule ROS1 inhibitors.
  • ALK anaplastic lymphoma kinase
  • Anaplastic lymphoma kinase is a receptor type protein tyrosine phosphokinase of the insulin receptor superfamily.
  • the invention provides a new, high-efficiency and broad-spectrum kinase inhibitor that can simultaneously act on oncogenic proteins such as NTRK, ALK and/or ROS1.
  • a compound represented by formula I or a tautomer, or a meso form, a racemate, and a mixture of meso and racemates, or Its enantiomers, diastereomers and mixtures of enantiomers and diastereomers, or its pharmaceutically acceptable salts, or its deuterated products:
  • X is independently selected from the following group: NR 6 , O, CR 1 R 2 , S, S(O) or S(O) 2 ;
  • B is selected from the following group: monocyclic aromatic hydrocarbons, bicyclic aromatic hydrocarbons, monocyclic heteroaromatic hydrocarbons or bicyclic heteroaromatic hydrocarbons, wherein the H on any carbon atom of B can be substituted by the following substituents: halogen, hydroxyl, amino, cyano , Acyl, ester, alkyl, cycloalkyl, alkylamino, alkoxy, cycloalkoxy, aryl, heteroaryl, mono- or poly-substituted alkyl, mono- or poly-substituted alkoxy, Mono- or multi-substituted cycloalkyl, mono- or multi-substituted cycloalkoxy, mono- or multi-substituted aryl, mono- or multi-substituted heteroaryl; said mono- or multi-substituted alkyl, mono- or multi-substituted The substituents of substituted alkoxy, mono-
  • C is independently Wherein, Y is independently selected from the group: O, NR A, or CR 1 R 2, Represents Z or E;
  • R 1 and R 2 are each independently selected from the following group: hydrogen atom, halogen, amino, cyano, hydroxyl, acyl, ester, alkyl, alkoxy, cycloalkyl, aryl, heteroaryl, Mono- or multi-substituted alkyl, mono- or multi-substituted alkoxy, mono- or multi-substituted cycloalkyl, mono- or multi-substituted aryl, mono- or multi-substituted heteroaryl; said mono- or multi-substituted
  • the substituents of alkyl, mono- or poly-substituted alkoxy, mono- or poly-substituted cycloalkyl, mono- or poly-substituted aryl, mono- or poly-substituted heteroaryl are independently selected from the following group: deuterium, halogen , Amino, cyano, hydroxy, acyl, ester, alkyl,
  • R 3 and R 4 are each independently selected from the following group: hydrogen atom, amino, hydroxy, acyl, ester, alkyl, cycloalkyl, aryl, heteroaryl, mono- or poly-substituted alkyl, mono-substituted or Multi-substituted alkoxy, mono- or multi-substituted cycloalkyl, mono- or multi-substituted aryl, mono- or multi-substituted heteroaryl; said mono- or multi-substituted alkyl, mono- or multi-substituted alkoxy , Mono- or poly-substituted cycloalkyl, mono- or poly-substituted aryl, mono- or poly-substituted heteroaryl substituents are independently selected from the following group: halogen, amino, cyano, hydroxy, acyl, ester, Alkyl, haloalkyl, cycloalkyl, halocycl
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 , and Z 7 are each independently selected from the following group: N, CR 5 or NR 6 ;
  • R 5 is independently arbitrarily selected from: hydrogen atom, halogen, amino, cyano, hydroxy, acyl, ester, alkyl, cycloalkyl, aryl, heteroaryl, mono- or poly-substituted alkyl, mono-substituted or Multi-substituted cycloalkyl, mono- or multi-substituted aryl, mono- or multi-substituted heteroaryl; said mono- or multi-substituted alkyl, mono- or multi-substituted cycloalkyl, mono- or multi-substituted aryl,
  • the substituents of mono- or multi-substituted heteroaryl groups are independently selected from halogen, amino, cyano, hydroxy, acyl, ester, alkyl, haloalkyl, cycloalkyl, halocycloalkyl, alkoxy, Halogenated alkoxy, aryl and heteroaryl;
  • R 6 and R A are each independently and arbitrarily selected from: hydrogen atom, acyl group, ester group, alkyl group, cycloalkyl group, aryl group, heteroaryl group, monosubstituted or polysubstituted alkyl group, monosubstituted or polysubstituted cycloalkyl group , Mono- or poly-substituted aryl, mono- or poly-substituted heteroaryl; said mono- or poly-substituted alkyl, mono- or poly-substituted cycloalkyl, mono- or poly-substituted aryl, mono- or poly-substituted
  • the substituents of the heteroaryl group are independently selected from halogen, amino, cyano, hydroxy, acyl, ester, alkyl, haloalkyl, cycloalkyl, halocycloalkyl, alkoxy, haloalkoxy, aryl Group and
  • A is Among them, * represents the chiral center; R 1 , R 2 , and X are defined as described above.
  • A is Wherein, * represents the R configuration, and the definitions of R 1 , R 2 and X are as described above.
  • X is NH or O.
  • R 1 and R 2 are each independently H, alkyl, haloalkyl, or cycloalkyl.
  • C is selected from the following group: among them,
  • Z is O
  • C is R 1, R 2, R 3 , R 4, R A is defined as described above, or R 1 and R C atom 4 together with their attached fused to form a substituted or unsubstituted 3-7 membered cycloalkane, nitrogen Heterocycloalkane, oxacycloalkane or thioheteroalkane, wherein said substitution refers to substitution by one or more groups selected from the group consisting of alkyl, acyl, ester, sulfonyl, and sulfinyl.
  • C is R 3, R & lt 4, R A is defined as described above, or R 3 and R C atoms together with their attached fused 4 form a substituted or unsubstituted 3-7 membered cycloalkane, aza-cycloalkane, oxa Cycloalkanes or thioheteroalkanes, wherein the substitution refers to substitution by one or more groups selected from the following group: alkyl, acyl, ester, sulfonyl, and sulfinyl.
  • A is wherein, the X is NR 6 , O, CR 1 R 2 , S, S(O) or S(O) 2 ;
  • B is arbitrarily selected from monocyclic aromatic hydrocarbons, bicyclic aromatic hydrocarbons, monocyclic heteroaromatic hydrocarbons or bicyclic heteroaromatic hydrocarbons, wherein H on any carbon atom of B can be substituted by the following substituents: halogen, hydroxyl, amino, cyano, ester group , Alkyl, haloalkyl, alkylamino, alkoxy, aryl or heteroaryl;
  • R 1 , R 2 , R 3 , R 4 are each independently selected from hydrogen atom, halogen, amino, cyano, hydroxyl, acyl, ester group, alkyl group, halogenated alkyl group, cycloalkyl group, halogenated ring Alkyl, aryl, heteroaryl, mono- or poly-substituted alkyl, mono- or poly-substituted cycloalkyl, mono- or poly-substituted aryl, mono- or poly-substituted heteroaryl;
  • the substituents of substituted alkyl, mono- or poly-substituted cycloalkyl, mono- or poly-substituted aryl, mono- or poly-substituted heteroaryl are independently selected from halogen, amino, cyano, hydroxy, acyl, ester group , Alkyl, haloalkyl, cycloalkyl, halo
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 , and Z 7 are each independently selected from N, CR 5 or NR 6 ;
  • X is selected from: NR 6 , O, CR 1 R 2 , S, S(O) or S(O) 2 ;
  • R 1 and R 2 are different, and are each independently selected from the following group: hydrogen atom, halogen, amino, cyano, hydroxyl, alkyl, haloalkyl;
  • R 6 is independently selected from the following group: hydrogen atom, alkyl group, mono- or multi-substituted alkyl, and the substituent of said mono- or multi-substituted alkyl is independently selected from the group: halogen, amino, cyano, hydroxyl , Acyl, ester, alkyl, haloalkyl, cycloalkyl, halocycloalkyl, alkoxy, haloalkoxy, aryl and heteroaryl;
  • Z 1 , Z 4 , and Z 5 are all N.
  • Z 2 , Z 4 , and Z 6 are all N.
  • Z 2 , Z 3 , Z 4 , and Z 6 are all N.
  • Z 3 , Z 6 , and Z 7 are all CR 5 , wherein R 5 is independently selected from the following group: hydrogen atom, halogen, amino, cyano, hydroxyl, acyl, ester, alkyl , Cycloalkyl, aryl, heteroaryl, mono- or poly-substituted alkyl, mono- or poly-substituted cycloalkyl, mono- or poly-substituted aryl, mono- or poly-substituted heteroaryl; said mono-substituted
  • the substituents of or polysubstituted alkyl, monosubstituted or polysubstituted cycloalkyl, monosubstituted or polysubstituted aryl, monosubstituted or polysubstituted heteroaryl are independently selected from halogen, amino, cyano, hydroxy, acyl, an ester group, an alkyl, haloalkyl, cyclol
  • Z 8 and Z 9 are each independently selected from: CR 11 or N;
  • P is independently selected from: O, NH, S;
  • Q when When it is a double bond, Q is independently selected from: CR 11 or N; when When it is a single bond, Q is independently selected from: O, S, CR 11 R 12 or NH;
  • Each R 7 is independently selected from the following group: hydrogen atom, halogen, amino, cyano, hydroxyl, acyl, ester, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, monosubstituted or poly Substituted alkyl, mono- or poly-substituted alkoxy, mono- or poly-substituted cycloalkyl, mono- or poly-substituted aryl, mono- or poly-substituted heteroaryl; said mono- or poly-substituted alkyl, mono- Substituents of substituted or polysubstituted alkoxy, monosubstituted or polysubstituted cycloalkyl, monosubstituted or polysubstituted aryl, monosubstituted or polysubstituted heteroaryl are independently selected from the following group: deuterium, halogen, amino, cyanide Group, hydroxy, acyl
  • R 11 and R 12 are each independently selected from the following group: H, hydroxy, halogen, amino, cyano, acyl, alkyl, haloalkyl, alkoxy, and haloalkoxy;
  • e 0, 1, 2, 3, or 4.
  • B is independently selected from the following group: Wherein, Z 8 and Z 9 are each independently selected from: CR 11 or N;
  • Each R 7 is independently selected from the following group: hydrogen atom, halogen, amino, cyano, hydroxyl, acyl, ester, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, monosubstituted or poly Substituted alkyl, mono- or poly-substituted alkoxy, mono- or poly-substituted cycloalkyl, mono- or poly-substituted aryl, mono- or poly-substituted heteroaryl; said mono- or poly-substituted alkyl, mono-
  • the substituents of substituted or multi-substituted alkoxy, mono- or multi-substituted cycloalkyl, mono- or multi-substituted aryl, mono- or multi-substituted heteroaryl are independently selected from deuterium, halogen, amino, cyano, Hydroxy, acyl, ester, alkyl, haloalky
  • Each R 11 is independently selected from the following group: H, hydroxy, halogen, amino, cyano, acyl, alkyl, haloalkyl, alkoxy, and haloalkoxy;
  • e 0, 1, 2;
  • B is independently Among them, Z 9 is CR 11 or N;
  • Each R 7 is independently selected from the following group: hydrogen atom, halogen, amino, cyano, hydroxyl, acyl, ester, alkyl, cycloalkyl, alkoxy, aryl, heteroaryl, monosubstituted or poly Substituted alkyl, mono- or poly-substituted alkoxy, mono- or poly-substituted cycloalkyl, mono- or poly-substituted aryl, mono- or poly-substituted heteroaryl; said mono- or poly-substituted alkyl, mono- Substituents of substituted or multi-substituted alkoxy, mono- or multi-substituted cycloalkyl, mono- or multi-substituted aryl, mono- or multi-substituted heteroaryl are independently selected from the following group: deuterium, halogen, amino, Cyano, hydroxy, acyl, ester, alkyl,
  • Each R 11 is independently selected from the following group: H, hydroxy, halogen, amino, cyano, acyl, alkyl, haloalkyl, alkoxy, and haloalkoxy;
  • e 0, 1, 2.
  • R 3 and R 4 are as described above.
  • R 3 and R 4 are each independently selected from the following group: hydrogen atom, halogen, amino, cyano, hydroxyl, acyl, ester, alkyl, halogenated alkyl, cycloalkyl, halogenated ring Alkyl, mono- or poly-substituted alkyl, mono- or poly-substituted cycloalkyl; the substituents of said mono- or poly-substituted alkyl, mono- or poly-substituted cycloalkyl are independently selected from the following group: halogen, Amino, cyano, hydroxy, acyl, ester, alkyl, haloalkyl, cycloalkyl, halocycloalkyl, alkoxy, and haloalkoxy.
  • the substitution refers to substitution by one or more groups selected from the following group: alkyl, acyl, ester, sulfonyl, and sulfinyl.
  • the compound of formula I or its tautomer, or its meso, racemate, mixture of meso and racemate, or its enantiomer Conformers, diastereomers and mixtures of enantiomers and diastereomers, or pharmaceutically acceptable salts thereof, or deuterated products thereof have one or more characteristics selected from the following group:
  • A is wherein, X is NH or O; R 1 and R 2 are each independently H, alkyl, or haloalkyl;
  • Z 9 is CR 11 or N;
  • Each R 7 is independently selected from the following group: hydrogen atom, halogen, hydroxy, acyl, alkyl, cycloalkyl, alkoxy, mono- or poly-substituted alkyl, mono- or poly-substituted alkoxy;
  • the substituents of mono- or poly-substituted alkyl, mono- or poly-substituted alkoxy are independently selected from the following group: deuterium, halogen, amino, cyano, hydroxy, alkyl, haloalkyl, cycloalkyl, halo Cycloalkyl, alkoxy, haloalkoxy;
  • Each R 11 is independently selected from the following group: H, hydroxy, halogen, alkyl, halogenated alkyl, alkoxy, halogenated alkoxy;
  • e 0, 1, 2;
  • C is independently Preferably C is wherein, Y is selected from the following group: O, CR 1 R 2 ; R 3 and R 4 are each independently selected from the following group: H, alkyl, mono- or poly-substituted alkyl, phenyl, pyridyl, mono-substituted or Multi-substituted phenyl, mono- or multi-substituted pyridyl; or R 3 and R 4 together with the C atom to which they are connected form a substituted or unsubstituted 3-8 membered cycloalkyl or heterocyclic group, and the substitution means the selected Substituting one or more groups from the following group: halogen, alkoxy, ester, sulfonyl;
  • Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 , and Z 7 are each independently N or CR 5 , wherein R 5 is selected from the group consisting of H and halogen.
  • the compound of formula I or its tautomer, or its meso, racemate, mixture of meso and racemate, or its enantiomer Conformers, diastereomers, mixtures of enantiomers and diastereomers, or pharmaceutically acceptable salts thereof, or deuterated products thereof, which have the structure shown in Formula III,
  • A, B, C, Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 , and Z 7 are specific groups corresponding to each specific compound in the embodiment.
  • the compound, or its tautomer, or its meso, racemate, mixture of meso and racemate, or its enantiomer Isomers, diastereomers and mixtures of enantiomers and diastereomers, or pharmaceutically acceptable salts thereof, or deuterated compounds thereof characterized in that: the compound represented by formula I is arbitrarily selected from The following compounds:
  • the compound represented by formula I is selected from the compounds shown in the embodiments of the present invention.
  • a pharmaceutically acceptable salt of the compound of formula I is an inorganic acid salt or an organic acid salt, and the inorganic acid salt is selected from hydrochloride and hydrobromide. , Hydroiodide, sulfate, bisulfate, nitrate, phosphate, acid phosphate; the organic acid salt is selected from formate, acetate, trifluoroacetate, propionate, acetone Acid salt, glycolate, oxalate, malonate, fumarate, maleate, lactate, malate, citrate, tartrate, methanesulfonate, ethanesulfonate Acid salt, isethionate, besylate, salicylate, picrate, glutamate, ascorbate, camphorate, camphorsulfonate.
  • the third aspect of the present invention provides a pharmaceutical composition, which contains a therapeutically effective amount of the compound described in the first aspect, or its tautomer, or its mesomer, racemate and meso Mixtures of isomers and racemates, or mixtures of enantiomers, diastereomers, enantiomers and diastereomers, or pharmaceutically acceptable salts thereof, or deuterium thereof Substitutes, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the fourth aspect of the present invention provides a compound as described in the first aspect, or a tautomer, or a mixture of meso, racemate, and meso and racemate, or Its enantiomers, diastereomers, and mixtures of enantiomers and diastereomers, or pharmaceutically acceptable salts thereof, or deuterated compounds thereof, or containing compounds represented by formula I
  • the composition is used in the preparation of drugs for the prevention and/or treatment of diseases with pathological characteristics mediated by ROS1, NTRK, ALK and the like.
  • the diseases with pathological characteristics mediated by ROS1, NTRK, ALK, etc. include cancer, sarcoma and pain.
  • the cancer is breast cancer, cervical cancer, colon cancer, lung cancer, gastric cancer, rectal cancer, pancreatic cancer, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer Any one of cancer, bladder cancer, liver cancer, fallopian tube tumor, peritoneal tumor, melanoma, glioma, glioblastoma, head and neck cancer, mastoid nephroma, leukemia, lymphoma, myeloma, and thyroid tumorkind.
  • the pharmaceutical composition provided by the present invention can be made into a suitable dosage form for administration.
  • These dosage forms include those suitable for oral, rectal, topical, intraoral, and other parenteral administration (e.g., subcutaneous, intramuscular, intravenous, etc.).
  • the pharmaceutical composition of the present invention can be formulated, quantified and administered in a manner that conforms to medical practice standards.
  • the "effective amount" of the compound administered is determined by factors such as the specific condition to be treated, the individual to be treated, the cause of the condition, the target of the drug, and the mode of administration.
  • the inventor of the present application unexpectedly discovered a new compound that has excellent inhibitory activity against ROS1, NTRK and ALK and their drug-resistant mutations, especially for drug-resistant mutations. It is active and has better pharmacodynamics, pharmacokinetic properties and lower toxic and side effects. It has the potential to develop into an effective drug for drug-resistant patients that is urgently needed in clinical practice.
  • Alkyl refers to a monovalent linear or branched saturated hydrocarbon group containing 1 to 12 carbon atoms composed only of carbon and hydrogen atoms.
  • Alkyl is preferably an alkyl group of 1 to 6 carbon atoms, that is, a C 1 -C 6 alkyl group, more preferably a C 1 -C 4 alkyl group.
  • alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, isobutyl, sec-butyl, tert-butyl, pentyl, n-hexyl, octyl, dodecyl and the like.
  • the alkyl group is also intended to include a deuterated alkyl group, and examples of the deuterated alkyl group include, but are not limited to, CD 3 , CD 2 CD 3 , CD 2 CD 2 CD 3 .
  • Alkoxy refers to a group of formula -OR or -R'-OR, where R is an alkyl group as defined herein and R'is an alkylene group.
  • alkoxy groups include, but are not limited to, methoxy, ethoxy, isopropoxy, tert-butoxy, -CH 2 O-CH 3 , -CH 2 CH 2 -O-CH 3 , -CH 2 -O-CH 2 CH 3 and so on.
  • Halogen refers to a fluorine, chlorine, bromine or iodine substituent.
  • Haloalkyl refers to an alkyl group as defined herein in which one or more hydrogens are replaced by the same or different halogens.
  • the "haloalkyl” is preferably a halogenated C 1 -C 6 alkyl group, more preferably a halogenated C 1 -C 4 alkyl group.
  • Examples of the halogenated alkyl group include -CH 2 Cl, -CH 2 CF 3 , -CH 2 CCl 3 , Perfluoroalkyl (for example, -CF 3 -, -CF 2 CF 3 ), etc.
  • Haloalkoxy refers to a group of formula -OR, where R is a haloalkyl group as defined herein.
  • Examples of haloalkoxy groups include, but are not limited to, trifluoromethoxy, difluoromethoxy, 2,2,2-trifluoroethoxy, and the like.
  • Cycloalkyl refers to a monovalent saturated carbocyclic group consisting of a mono- or bicyclic ring, which has 3-12 (C 3 -C 12 ), preferably 3-10 (C 3 -C 10 ), More preferably, 3-6 ring atoms (C 3 -C 6 ). Cycloalkyl groups may be optionally substituted with one or more substituents, where each substituent is independently a hydroxy, alkyl, alkoxy, halogen, haloalkyl, amino, monoalkylamino, or dialkylamino. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like.
  • Cycloalkoxy refers to a group of formula -OR, where R is a cycloalkyl as defined herein.
  • exemplary cycloalkyloxy groups include cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
  • Acyl refers to a group of formula -C(O)R, where R is alkyl or alkylamino as defined herein.
  • “Acyl” is preferably -C(O)C 1 -C 6 alkyl, -C(O)NH 2 , -C(O)NHC 1 -C 6 alkyl, -C(O)N(C 1- C 6 alkyl) 2 , more preferably -C(O)C 1 -C 3 alkyl, -C(O)NH 2 , -C(O)NHC 1 -C 3 alkyl, -C(O) N(C 1 -C 3 alkyl) 2 , exemplary acyl groups include acetyl, n-propionyl, isopropionyl, n-butyryl, isobutyryl, tert-butyryl, -C(O)NH 2 , -C (O)NHCH 3 , -C(O)N
  • Alkylamino refers to a group of formula -NRaRb, wherein Ra and Rb are the same or different, and each is independently H or an alkyl group as defined herein.
  • Ester group refers to a group of formula -C(O)OR, where R is an alkyl group as defined herein.
  • the ester group is preferably -C(O)OC 1 -C 6 alkyl, more preferably -C(O)OC 1 -C 4 alkyl.
  • Exemplary ester groups include -C(O)OMe, -C (O)OEt, -C(O)OC(CH 3 ) 3, etc.
  • Sulfonyl refers to the formula -S(O) 2- R, where R is an alkyl group as defined herein.
  • the sulfonyl group is preferably -S(O) 2 -C 1 -C 6 alkyl, exemplarily including -S(O) 2 -Me, -S(O) 2 -Et and the like.
  • Sulfonyl refers to the formula -SO-R, where R is an alkyl group as defined herein.
  • the sulfinyl group is preferably -SO-C 1 -C 6 alkyl, exemplarily including -SO-Me, -SO-Et and the like.
  • Alkylthio refers to a group of formula -SRa, wherein Ra is H or an alkyl group as defined herein.
  • Cycloalkylamino refers to a group of formula -NRaRb, wherein Ra is H, alkyl as defined herein, or cycloalkyl as defined herein, and Rb is cycloalkyl as defined herein; or Ra and Rb and the N atom to which it is attached together form a 3-6 membered N-containing heterocyclic group, such as tetrahydropyrrolyl.
  • Heterocycle (base) refers to a fully saturated or partially unsaturated cyclic group (including but not limited to, for example, 3-7 membered monocyclic ring, 6-11 membered bicyclic ring, or 8-16 membered tricyclic ring system) , Where at least one heteroatom is present in a ring with at least one carbon atom.
  • Each heterocyclic ring containing heteroatoms has 1, 2, 3 or 4 heteroatoms selected from nitrogen, oxygen or sulfur atoms, where nitrogen or sulfur atoms can be oxidized, and nitrogen atoms can also be quaternized .
  • Heterocycloalkane (base) refers to a fully saturated heterocycle (base).
  • the heterocyclic group can be attached to any heteroatom or carbon atom residue of the ring or ring system molecule.
  • Typical monocyclic heterocycles include, but are not limited to, azetidinyl, pyrrolidinyl, oxetanyl, pyrazolinyl, imidazolinyl, imidazolidinyl, oxazolidinyl, isoxazolidine Group, thiazolidinyl, isothiazolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, hexahydroazepine Inyl, 4-piperidinone, tetrahydropyranyl, morpholino, thiomorpholino, thiomorpholinosulfoxide, thiomorpholinosulfone, 1,3-dioxanyl
  • Polycyclic heterocyclic groups include spiro, condensed and bridged heterocyclic groups; the spiro, condensed and bridged heterocyclic groups are optionally connected to other groups through a single bond, or through a ring Any two or more of the above atoms are further connected to other cycloalkyl groups, heterocyclic groups, aryl groups and heteroaryl groups.
  • Aromatic hydrocarbon (base) refers to an aromatic cyclic hydrocarbon compound group with 1-5 rings, especially monocyclic and bicyclic groups, where there are two or more aromatic rings (bicyclic, etc.), aryl groups
  • the aromatic ring of the group can be connected by a single bond (such as biphenyl), or fused (such as naphthalene, anthracene, etc.).
  • the aryl group is preferably a C6-C12 aryl group, which refers to an aromatic cyclic hydrocarbon compound group containing 6, 7, 8, 9, 10, 11 or 12 ring carbon atoms.
  • aryl groups examples include, but are not limited to, phenyl, biphenyl, or naphthyl.
  • the aryl group can be condensed with the heterocyclic group through a single bond or any two adjacent ring C atoms, for example: chromanyl, chromanyl, benzodioxanyl, Wait.
  • Heteroaromatic (base) refers to a monocyclic, bicyclic or tricyclic group of 5 to 12 ring atoms (5-12 members), which contains at least one (such as 1, 2 or 3) selected from The ring heteroatoms of N, O or S and the remaining ring atoms are the aromatic ring of C. It should be clear that the point of attachment of the heteroaryl group should be on the heteroaromatic ring.
  • the heteroaryl group preferably has specifically 5-8 ring atoms (5-8 members), and more preferably has 5-6 ring atoms (5-6 members).
  • heteroaryl groups include, but are not limited to: imidazolyl, Azolyl, iso Azolyl, thiazolyl, isothiazolyl, Diazolyl, thiadiazolyl, pyrazinyl, thienyl, furyl, pyranyl, pyridyl, pyrrolyl, pyrazolyl, pyrimidinyl, quinolinyl, isoquinolinyl, benzofuranyl, , Benzothienyl, benzothiopyranyl, benzimidazolyl, benzo Azolyl, benzo Diazolyl, benzothiazolyl, benzothiadiazolyl, benzopyranyl, indolyl, isoindolyl, triazolyl, triazinyl, quinoxalinyl, purinyl, quinazoline Group, quinazinyl, naphthyridinyl, pterridinyl, carbazoly
  • Multiple substitution refers to two or more substitutions.
  • the alkyl, alkoxy, cycloalkyl, heterocyclic, aryl, heteroaryl and other groups include substituted alkyl, alkoxy, and cycloalkane unless otherwise specified.
  • Groups, heterocyclic groups, aryl groups, heteroaryl groups, etc., the substituents such as (but not limited to): halogen, hydroxyl, cyano, acyl, sulfonyl, ester, sulfinyl, alkyl, cycloalkane Groups, heterocyclic groups, aryl groups, heteroaryl groups, acyl groups, ester groups, etc.
  • Deuterated compound refers to a compound obtained by replacing one hydrogen atom (H) or multiple hydrogen atoms (H) with deuterium atoms (D) in a compound.
  • the terms "compounds of the present invention” or “active ingredients of the present invention” are used interchangeably and refer to compounds of formula I, or tautomers thereof, or meso, racemate, and endo Mixtures of racemates and racemates, or mixtures of enantiomers, diastereomers, and enantiomers and diastereomers, or pharmaceutically acceptable salts thereof, or Its deuterated substance.
  • the compound of formula I or its tautomer, or its meso, racemate and mixture of meso and racemate, or its enantiomers, diastereomers And a mixture of enantiomers and diastereomers, or a pharmaceutically acceptable salt thereof, or a deuterated product thereof has the following structure,
  • A, B, C, Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 , and Z 7 are defined as described above.
  • the compound of formula I, or its tautomer, or its meso, racemate, and mixture of meso and racemate, or its enantiomers, diastereomers Isomers and mixtures of enantiomers and diastereomers, or their pharmaceutically acceptable salts, or their deuterated products have the structure shown in Formula III,
  • salts that the compounds of the present invention may form also belong to the scope of the present invention. Unless otherwise specified, the compounds in the present invention are understood to include their salts.
  • the term "salt” as used herein refers to a salt in the acid or basic form formed with an inorganic or organic acid and a base.
  • the compound of the present invention contains a basic fragment, it includes but is not limited to pyridine or imidazole, and when it contains an acidic fragment, including but not limited to carboxylic acid, the zwitterion (“internal salt”) that may be formed is contained in Within the scope of the term "salt”.
  • salts are preferred, although other salts are also useful, for example, they can be used in separation or purification steps in the preparation process.
  • the compound of the present invention may form a salt.
  • the compound I can be obtained by reacting with a certain amount of acid or base, such as an equivalent amount of acid or base, and salting out in the medium, or freeze-dried in an aqueous solution.
  • the basic fragments contained in the compounds of the present invention may form salts with organic or inorganic acids.
  • Typical acids that can form salts include hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, nitrate, phosphate, and acid phosphate; the organic acid salt is selected from formate , Acetate, trifluoroacetate, propionate, pyruvate, glycolate, oxalate, malonate, fumarate, maleate, lactate, malic acid Salt, citrate, tartrate, methanesulfonate, ethanesulfonate, isethionate, benzenesulfonate, salicylate, picrate, glutamate, ascorbate, camphorate , Camphor sulfonate and so on.
  • the acidic fragments that some compounds of the present invention may contain, including but not limited to carboxylic acids, may form salts with various organic or inorganic bases.
  • Typical salts formed by bases include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium, magnesium salts, and salts formed by organic bases (such as organic amines), such as benzathine and dicyclohexylamine , Hypamine (a salt formed with N,N-bis(dehydroabietyl)ethylenediamine), N-methyl-D-glucamine, N-methyl-D-glucamide, tert-butyl Amines, and salts with amino acids such as arginine, lysine, etc.
  • Basic nitrogen-containing groups can be combined with halide quaternary ammonium salts, such as small molecular alkyl halides (such as methyl, ethyl, propyl and butyl chloride, bromide and iodide), dialkyl sulfate (E.g., dimethyl sulfate, diethyl, dibutyl and dipentyl sulfate), long chain halides (such as chlorides and bromides of decyl, dodecyl, tetradecyl and tetradecyl And iodides), aralkyl halides (such as benzyl and phenyl bromides) and so on.
  • small molecular alkyl halides such as methyl, ethyl, propyl and butyl chloride, bromide and iodide
  • dialkyl sulfate E.g., dimethyl sulfate, diethyl,
  • prodrugs and solvates of the compounds of the present invention are also covered.
  • prodrug herein refers to a compound that undergoes metabolic or chemical transformation to produce the compound, salt, or solvate of the present invention when treating related diseases.
  • the compounds of the present invention include solvates such as hydrates.
  • the compounds, salts or solvates of the present invention may exist in tautomeric forms (such as amides and imine ethers). All these tautomers are part of the invention.
  • All stereoisomers of compounds (for example, those asymmetric carbon atoms that may exist due to various substitutions), including their enantiomeric forms and diastereomeric forms, fall within the scope of the present invention.
  • the independent stereoisomers of the compound in the present invention may not coexist with other isomers (for example, as a pure or substantially pure optical isomer with special activity), or may be a mixture, such as Racemates, or mixtures with all other stereoisomers or part of them.
  • the chiral center of the present invention has two configurations, S or R, and is determined by the International Union of Theoretical and Applied Chemistry
  • racemic form can be resolved by physical methods, such as fractional crystallization, or separation of crystallization by derivatization into diastereomers, or separation by chiral column chromatography.
  • Individual optical isomers can be obtained from racemates by suitable methods, including but not limited to traditional methods, such as salt formation with an optically active acid and recrystallization.
  • the weight content of the compound obtained by successive preparation, separation and purification is equal to or greater than 90%, for example, equal to or greater than 95%, equal to or greater than 99% ("very pure" compound), as described in the text Listed.
  • very pure compounds of the invention are also part of the invention.
  • All configuration isomers of the compounds of the present invention are within the scope of coverage, whether in mixture, pure or very pure form.
  • the definition of the compound of the present invention includes two olefin isomers, cis (Z) and trans (E), as well as cis and trans isomers of carbocyclic and heterocyclic rings.
  • Certain compounds of the present invention may exist in specific geometric or stereoisomeric forms.
  • the present invention covers all compounds, including their cis and trans isomers, R and S enantiomers, diastereomers, (D) isomers, (L) isomers, and exogenous Spin the mixture and other mixtures.
  • the asymmetric carbon atom may represent a substituent, such as an alkyl group. All isomers and their mixtures are included in the present invention.
  • the mixture of isomers can contain various isomer ratios.
  • a mixture of only two isomers can have the following combinations: 50:50, 60:40, 70:30, 80:20, 90:10, 95:5, 96:4, 97:3, 98: 2, 99:1, or 100:0, all ratios of isomers are within the scope of the present invention. Similar ratios that are easily understood by those skilled in the art and ratios that are mixtures of more complex isomers are also within the scope of the present invention.
  • the present invention also includes isotopically labeled compounds, which are equivalent to the original compounds disclosed herein. However, in fact, it usually occurs when one or more atoms are replaced by atoms whose atomic weight or mass number is different.
  • isotopes of compounds that can be included in the present invention include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine isotopes, such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, and 18 O, respectively. , 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • the compounds of the present invention or enantiomers, diastereomers, isomers, or pharmaceutically acceptable salts or solvates, which contain isotopes or other isotopic atoms of the above compounds are all within the scope of the present invention.
  • Certain isotope-labeled compounds of the present invention such as radioisotopes of 3 H and 14 C, are also among them, which are useful in tissue distribution experiments of drugs and substrates.
  • Isotopically-labeled compounds can be prepared by a general method, by replacing readily available isotope-labeled reagents with non-isotopic reagents, using the protocol disclosed in the example.
  • a specific enantiomer of the compound of the present invention can be prepared by asymmetric synthesis, or derivatized with a chiral adjuvant, separating the resulting diastereomeric mixture, and then removing the chiral adjuvant.
  • the pure enantiomer if the molecule contains a basic functional group, such as an amino acid, or an acidic functional group, such as a carboxyl group, a suitable optically active acid or base can be used to form a diastereomeric salt with it, and then through separation crystallization or chromatography, etc. After separation by conventional means, the pure enantiomers are obtained.
  • the compounds of the present invention can be combined with any number of substituents or functional groups to expand their coverage.
  • the general formula including substituents in the formula of the present invention refers to the replacement of hydrogen radicals with designated structural substituents.
  • each position of the substituents may be the same or different.
  • substitution as used herein includes all permissible substitution of organic compounds. Broadly speaking, the permissible substituents include acyclic, cyclic, branched unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic organic compounds.
  • the heteroatom nitrogen may have a hydrogen substituent or any permitted organic compound as described above to supplement its valence.
  • the present invention is not intended to limit the permitted substitution of organic compounds in any way.
  • the present invention believes that the combination of substituents and variable groups is excellent in the treatment of diseases in the form of stable compounds.
  • stable refers to a compound that is stable and can be tested for a long enough time to maintain the structural integrity of the compound, preferably for a long enough time to be effective, and is used herein for the above purpose.
  • the compounds of the present invention can be conveniently prepared by combining various synthetic methods described in this specification or known in the art, and such combinations can be easily performed by those skilled in the art to which the present invention belongs.
  • each reaction is usually carried out in an inert solvent at -60°C to 100°C, preferably -60°C to 80°C.
  • the reaction time is usually 0.1 hour to 60 hours, preferably 0.5 to 48 hours.
  • the preferred synthetic route is as follows:
  • A, B, C, Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 , Z 7 , R 3 , R 4 , R A are defined as described above;
  • Route 1 (1) In an inert solvent (such as ethanol, methanol), compound 1 and compound 2 are under the action of a base (such as sodium carbonate, potassium carbonate, sodium hydroxide, triethylamine, pyridine, etc.), A nucleophilic substitution reaction occurs to produce compound 3; (2) In an inert solvent (such as ethanol, methanol), under the action of a base (such as sodium carbonate, potassium carbonate, sodium hydroxide, triethylamine, pyridine, etc.), the compound 3 reacts with hydroxylamine hydrochloride to produce compound 4; (3) In an inert solvent (such as 1,2-dichloroethane and/or glacial acetic acid), compound 4 reacts with dimethoxy acetal to obtain the final product 5 .
  • a base such as sodium carbonate, potassium carbonate, sodium hydroxide, triethylamine, pyridine, etc.
  • a base such as sodium carbonate, potassium carbonate, sodium hydroxide, triethy
  • the starting materials of the present invention are all known and commercially available, or can be synthesized according to literature reports in the field.
  • the pharmaceutical composition of the present invention is used to prevent and/or treat the following diseases: inflammation, cancer, cardiovascular disease, infection, immune disease, and metabolic disease.
  • the compounds of the present invention can be combined with other drugs known to treat or improve similar conditions.
  • the original drug administration mode and dosage can remain unchanged, while the compound of the present invention is administered simultaneously or subsequently.
  • a pharmaceutical composition containing one or more known drugs and the compound of the present invention can be preferably used.
  • the drug combination also includes taking the compound of the present invention and one or more other known drugs in overlapping time periods.
  • the dose of the compound of the present invention or a known drug may be lower than the dose of the compound alone.
  • the dosage form of the pharmaceutical composition of the present invention includes (but is not limited to): injection, tablet, capsule, aerosol, suppository, film, dripping pill, external liniment, controlled release or sustained release or nano preparation.
  • the pharmaceutical composition of the present invention contains the compound of the present invention or a pharmacologically acceptable salt thereof and a pharmacologically acceptable excipient or carrier within a safe and effective amount.
  • the "safe and effective amount” refers to: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects.
  • the pharmaceutical composition contains 1-2000 mg of the compound of the present invention/agent, more preferably, 10-1000 mg of the compound of the present invention/agent.
  • the "one dose" is a capsule or tablet.
  • “Pharmaceutically acceptable carrier” refers to: one or more compatible solid or liquid fillers or gel substances, which are suitable for human use, and must have sufficient purity and sufficiently low toxicity. "Compatibility” here means that the components in the composition can be blended with the compound of the present invention and between them without significantly reducing the efficacy of the compound.
  • pharmaceutically acceptable carriers include cellulose and its derivatives (such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, talc, and solid lubricants (such as stearic acid).
  • Magnesium stearate calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as Tween) ), wetting agents (such as sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • vegetable oils such as soybean oil, sesame oil, peanut oil, olive oil, etc.
  • polyols such as propylene glycol, glycerin, mannitol, sorbitol, etc.
  • emulsifiers such as Tween
  • wetting agents such as sodium lauryl sulfate
  • coloring agents such as sodium lauryl sulfate
  • flavoring agents such as pepperminophen, sorbitol, etc.
  • antioxidants
  • the method of administration of the compound or pharmaceutical composition of the present invention is not particularly limited.
  • Representative administration methods include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration .
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or mixed with the following ingredients: (a) fillers or compatibilizers, for example, Starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, such as hydroxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and gum arabic; (c) humectant, For example, glycerin; (d) disintegrants, such as agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) Absorption accelerators, such as quaternary amine compounds; (g) wetting agents, such as cetyl alcohol and gly
  • Solid dosage forms such as tablets, sugar pills, capsules, pills and granules can be prepared with coatings and shell materials, such as enteric coatings and other materials known in the art. They may contain opacifying agents, and the active compound or the release of the compound in the composition may be released in a certain part of the digestive tract in a delayed manner. Examples of embedding components that can be used are polymeric substances and waxes. If necessary, the active compound can also be formed into microcapsules with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • the liquid dosage form may contain inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-Butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances.
  • composition may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfumes.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfumes.
  • the suspension may contain suspending agents, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • suspending agents for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • composition for parenteral injection may contain physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
  • the dosage form of the compound of the present invention for topical administration includes ointment, powder, patch, spray and inhalant.
  • the active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants that may be required if necessary.
  • the treatment method of the present invention can be administered alone or in combination with other treatment means or therapeutic drugs.
  • a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, wherein the dosage is the pharmaceutically effective dosage considered to be administered.
  • the daily dose administered is usually 1-2000 mg, preferably 10-1000 mg.
  • the specific dosage should also consider factors such as the route of administration, the patient's health status, etc., which are within the skill range of a skilled physician.
  • the present invention also provides a method for preparing a pharmaceutical composition, which includes the steps of: subjecting a pharmaceutically acceptable carrier to the compound of the present invention or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof. Mix to form a pharmaceutical composition.
  • the present invention also provides a method of treatment, which comprises the steps of: administering the compound described in the present invention, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, to a subject in need of treatment, or administering the compound
  • the pharmaceutical composition of the invention is used to selectively inhibit the fusion mutations of ROS1, NTRK, ALK, etc. and their resistance mutations.
  • the compound of the present invention has a good inhibitory ability on ROS1, NTRK, and ALK kinases, especially the activity against these target resistance mutations;
  • the compound of the present invention has better pharmacodynamics, pharmacokinetic properties and lower toxic and side effects;
  • the compounds of the present invention have great potential to be developed into effective drugs for drug-resistant patients that are urgently needed in clinical practice.
  • Example 11 1ml of glacial acetic acid and 1,2-dichloroethane (1ml) were added to compound 5 (70mg, 1.0eq), and then 2,2-dimethoxypropane (81mg, 4eq) was added, Replace nitrogen and react at 80°C for 1 hour. TLC monitors that the reaction is complete, spin dry the solvent, then add sodium bicarbonate aqueous solution to the system, adjust the pH to 7-8, then add EA (10mlX3) for extraction, combine the EA phases and add anhydrous sodium sulfate to dry, filter, spin-dry the column , To obtain 15 mg (19% yield).
  • Example 12 Add glacial acetic acid (1ml) and 1,2-dichloroethane (1ml) to compound 8 (100mg, 1.0eq), and then add 2,2-dimethoxypropane (112mg, 4eq) ), replace nitrogen, react at 80°C for 1h. TLC monitors that the reaction is complete, spin-dry the solvent, then add sodium bicarbonate aqueous solution to the system, adjust the pH to 7-8, then add EA (20mlX3) for extraction, combine the EA phases and add anhydrous sodium sulfate to dry, filter, spin-dry the column , Obtained 40 mg of Example 14 (yield 36%).
  • Example 13 To compound 8 (700mg, 1.0eq), glacial acetic acid (7ml) and 1,2-dichloroethane (7ml) were added, and then 2,2-dimethoxypropane (832mg, 4.0 eq), replacing nitrogen, reacting at 80°C for 1 hour. TLC monitors the reaction to complete, spin dry the solvent, then add aqueous sodium bicarbonate solution to the system, adjust the pH to 7-8, then add EA (30mlX3) for extraction, combine the EA phases and add anhydrous sodium sulfate to dry, filter, spin dry, and pass Column, 42 mg (yield 5%) were obtained.
  • TLC monitors the reaction to complete, spin dry the solvent, then add aqueous sodium bicarbonate solution to the system, adjust the pH to 7-8, then add EA (30mlX3) for extraction, combine the EA phases and add anhydrous sodium sulfate to dry, filter, spin dry, and pass Column, 42 mg (yield 5%) were obtained
  • Example 14 Weigh compound 6 (0.2g) into a 50ml round bottom flask, then add 2,2-dimethoxypropane (0.22g, 4eq), measure 1,2-dichloroethane (4ml) and acetic acid (4ml) are mixed solvents, heated under reflux at 80°C for 2 hours, monitoring the completion of the reaction. A small amount of water was added, and then saturated sodium bicarbonate was added to neutralize the acetic acid in the reaction system, followed by extraction with dichloromethane. Then, it was passed through the column with dichloromethane: methanol (30:1) to obtain 66 mg of the final compound.
  • Example 15 (10) Synthesis of Example 15: Place compound 10 (41mg, 0.1mmol) in a 50ml round-bottomed flask, then add 2,2-dimethoxypropane (61.95mg, 6eq, 0.6mmol), and measure 1 , 2-Dichloroethane (2mL) and acetic acid (2mL) were mixed solvents, heated at 80°C under reflux for 2 hours, and monitored the completion of the reaction. A small amount of water was added, and then saturated sodium bicarbonate was added to neutralize the acetic acid in the reaction system, followed by extraction with dichloromethane. The column was then passed through the column with dichloromethane: methanol (30:1) to obtain 15 mg of the final compound.
  • Example 16 Synthesis of Example 16: Compound 7 (420mg, 1.1mmol) was placed in a 50ml round-bottomed flask, then 2,2-dimethoxypropane (0.46g, 4eq, 4.44mmol) was added, and 1,2- Dichloroethane (3mL) and acetic acid (3mL) were mixed solvents, heated at 80°C under reflux, and monitored after the reaction was completed. A small amount of water was added, and then saturated sodium bicarbonate was added to neutralize the acetic acid in the reaction system, followed by extraction with dichloromethane. Then the column was passed through the column with dichloromethane: methanol (30:1) to obtain 26 mg of the final compound.
  • Examples 17-83 were synthesized, as shown in Table 1-2:
  • Example 84 Example 25 and its enantiomers
  • Example 84 Refer to the synthesis of chiral amine intermediate compound 4 in Example 12 and the synthesis of Example 84, using p-fluoroacetophenone and (R)-tert-butylsulfinamide as raw materials, after generating imines, then sodium borohydride Reduction, two steps, to obtain a pair of diastereoisomers compound 3 and compound 3'. The two compounds are separated by column chromatography, and then the tert-butylsulfinyl group is removed to obtain two R and S configurations. A chiral amine intermediate. The two chiral amine intermediates are reacted separately to obtain the compound with two configurations of R (ie, Example 25) and S in Example 84. The two chiral amine intermediates are mixed to obtain Example 84, which is Racemic body.
  • the two compounds are separated by preparing the liquid phase under the following conditions:
  • Solvent methanol, sample concentration 12 mg/ml
  • Injection volume 0.5 ml, delay time: 24 seconds;
  • Threshold 20000, timetable: 2.00,
  • Mobile phase A: water (0.1% trifluoroacetic acid), B; methanol.
  • Test Example 1 Inhibitory activity of the compound of the present invention on ROS1, NTRK, ALK and their drug-resistant kinases
  • the compound's activity inhibition experiment on protein kinase was carried out on the HotSpot kinase experimental platform radiolabeled by Reaction Biology Corporation.
  • reaction solution after incubation was transferred to P81 ion exchange chromatography paper (Whatman#3698-915), eluted with 0.75% phosphoric acid solution, and the amount of phosphorylated substrate containing radioactivity remaining on the chromatography paper was detected.
  • Table 2 shows the IC 50 value of the inhibitory activity of the compounds of the present invention on ROS1, NTRK and ALK and their resistant kinases, where A ⁇ 0.5nM, 0.5nM ⁇ B ⁇ 5.0nM, 5.0nM ⁇ C ⁇ 50nM, 50nM ⁇ D ⁇ 500nM,E>500nM;
  • the kinase activity test shows that the series of compounds of the present invention have good inhibitory activity against ROS1, NTRK and ALK and their drug-resistant mutations, especially the inhibition of drug-resistant mutations.
  • the compound of the present invention has better inhibitory activity against ROS1, NTRK, ALK and one or more of their drug-resistant mutations than the current clinical drugs.
  • Most of the compounds of the present invention have one or more activities against ROS1, NTRK and ALK and their drug-resistant mutations, which are superior or equivalent to current clinical drugs.
  • the compound of the present invention has great potential to be applied to the treatment of diseases mediated by ROS1, NTRK and ALK.
  • Test Example 2 Inhibition of cell proliferation by compound
  • the compound's cell proliferation inhibition experiment was carried out in Hefei Zhongke Puruisheng Biomedical Technology Co., Ltd.
  • the Ba/F3 engineered cell lines stably transfected with different kinase genes were recovered with RPMI 1640 medium (Biological Industries, Israel) + 10% fetal bovine serum (Biological Industries, Israel) + 1% double antibody (Penicillin Streptomycin solution, Coring, USA) was cultured for two generations, the logarithmic growth phase cell suspension was taken, and 2000 cells/well were inoculated on 96-well white cell culture plates (Corning 3917, NY, USA) with a volume of 95 ⁇ L per well.
  • Table 3 shows the compounds of the present invention to Ba ROS1, NTRK thereof, and ALK or resistance mutations / F3 inhibiting activity IC 50 value of the engineered cells.
  • the series of compounds of the present invention have good inhibitory activity against ROS1, NTRK and ALK and their resistant mutant Ba/F3 engineered cell lines, especially the inhibition of resistant mutations.
  • the compounds of the present invention have good inhibitory activity against ROS1, NTRK and ALK and their resistant mutant Ba/F3 engineering cell lines, and most of the compounds of the present invention are against ROS1, NTRK and ALK and their resistant mutations.
  • the Ba/F3 engineered cell line is also very active and has the potential to be applied to the treatment of diseases mediated by ROS1, NTRK and ALK.

Abstract

一种式I所示的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物。式I化合物可用作激酶抑制剂,作为用于治疗ROS1、NTRK、ALK等激酶介导的疾病的药物。

Description

用作激酶抑制剂的化合物及其应用 技术领域
本发明涉及医药技术领域,具体涉及用作原肌球蛋白受体激酶抑制剂的化合物,其制备方法,以及在制备用于治疗ROS1、NTRK、ALK等激酶介导的疾病的药物方面的应用。
背景技术
原肌球蛋白受体激酶(TRK)家族属于跨膜受体酪氨酸激酶(RTKs),参与调节哺乳动物神经***的突触生长与功能维持、记忆的发生发展以及保护神经元免受损伤等。TRK激酶是一类神经生长因子受体,其家族由高度同源性的原肌球蛋白相关激酶A(Tropomyosin-related kinase A,TRKA)、原肌球蛋白相关激酶B(Tropomyosin-related kinase B,TRKB)、原肌球蛋白相关激酶C(Tropomyosin-related kinase C,TRKC)组成,分别有NTRK1、NTRK2和NTRK3基因编码。完整的TRK激酶包括胞外区、跨膜区和胞内区三个部分,和其他的RTKs一样,TRK激酶的胞外区与相应的配体结合之后,形成二聚体,能够引起TRK激酶的胞内区发生自体磷酸化从而激活自身的激酶活性,进一步激活下游的信号转导通路。TRK激酶通过Ras/MAPK、PI3K/AKT和PLCγ等下游通路影响细胞的增殖、分化、代谢和凋亡。当NTRKs基因发生融合或突变后,会改变或消除胞外区受体(Greco,A.et.al,Mol.Cell.Biol.1995,15,6118;Oncogene 1998,16,809),而融合或突变的TRK蛋白在不需要配体结合的情况下,自身处于高度活化的激酶活性状态,从而能够持续性的激活下游的信号转导通路,可导致TRK激酶下游信号通路调控失常,诱导细胞的增殖,促进肿瘤的发生和发展。NTRKs基因融合出现在多种成人和儿童实体瘤中,包括乳腺癌、结直肠癌、非小细胞肺癌、***状甲状腺癌、Spitz样黑色素瘤、神经胶质瘤以及各种肉瘤等。在常见的癌症中,如非小细胞肺癌、结直肠癌等中,NTRK基因融合的发生率较低,大致为1%-3%,但在一些罕见的癌症中,如婴儿纤维肉瘤、乳腺分泌型癌等,NTRK基因融合的发生率可达90%以上。最早的TPM3-TRKA融合蛋白是在结肠癌细胞中发现的。后来在不同的临床肿瘤病人样本如乳腺癌、非小细胞肺癌、***状甲状腺癌、Spitz样黑色素瘤、神经胶质瘤等中发现了更多类型的NTRK融合蛋白,如CD74-NTRKA、MPRIP-NTEKA、QKI-NTRKB、ETV6-NTRKC、BTB1-NTRKC等。因此,近年来,NTRK融合蛋白成为了一个有效的抗癌靶点,成为了抗癌药物研发的一个热点。随着近年来人们对TRK激酶的进一步的深入了解,发现了更多的TRK融合蛋白类型及突变类型(Russo,M.et.al Cancer Discovery,2016,6,36;Drilon,A.et.al,Annals of Oncology,2016,27,920),所以临床上急需开发活性更好,作用更广泛的新型NTRK抑制剂,从而解决这些NTRK蛋白融合或突变所引起的肿瘤的治疗问题。
ROS1(c-ros oncogene 1 receptor kinase)是一种人体内由ROS1原癌基因编码的酪氨酸蛋白激酶,定位于6q22.1染色体,属于酪氨酸激酶胰岛素受体基因,由胞内酪氨酸激酶活性区、跨膜区及胞外区3部分组成,编码具有酪氨酸激酶活性的嵌合蛋白。基本结构由胞外N-末端配体结合区(氨基酸1-1861)、跨膜区(氨基酸1862-1882)及胞内C-末端464个氨基酸构 成的酪氨酸激酶活性区(氨基酸1883-2347)组成。ROS1基因发生重排时丢失细胞外区域,保留跨膜区和胞内酪氨酸激酶区域,重排位点主要发生在ROS1基因的32~36外显子。ROS1基因突变主要发生在肺癌患者中,患者比例为1%-2%。在NSCLC中ROS1基因主要与SLC34A2、CD74发生融合,并持续激活ROS1酪氨酸激酶区及下游JAK/STAT、PI3K/AKT、RAS/MAPK等信号通路,进而引起肿瘤的发生。在大量地文献和临床上均已经证实,通过抑制突变的ROS1激酶的活性,就可以达到治疗由ROS1过度激活所导致的疾病,尤其是癌症。目前上市的用于ROS1阳性非小细胞肺癌的治疗药物有克唑替尼和恩曲替尼,他们均属于第一代的小分子ROS1抑制剂。但是,在服用克唑替尼或恩曲替尼的治疗过程中,大约在15个月左右会产生耐药,发生疾病进展。在发生耐药的患者中,最为常见的耐药突变就是G2032R等溶剂前沿突变,对于耐药的患者,目前还没有治疗药物上市。所以目前急需研发针对ROS1的新抑制剂,尤其是针对使用克唑替尼或恩曲替尼等一代ROS1抑制剂产生耐药的新的ROS1抑制剂药物用于临床的治疗。
NSCLC中有2-5%的病例为间变性淋巴瘤激酶(ALK)重排型,间变性淋巴瘤激酶是胰岛素受体超家族的一个受体型蛋白质酪氨酸磷酸激酶。最初人们是在间变性大细胞淋巴瘤中以一种激活的融合癌基因的形式发现了ALK,随后连续的研究在多种癌症中发现了ALK的融合形式,其中包括***性组织异常增生、炎性肌纤维细胞癌、非小细胞肺癌等。ALK在多种癌症中的突变和异常的活性,已经使其成为一个治疗ALK阳性癌症的药物靶点。目前上市了多个ALK激酶抑制剂,随着这些药物在临床上的应用,患者都会发生耐药突变,如果G1202R等耐药突变,导致这些药物失去疗效。
随着近年来人们对ROS1、NTRK、ALK等激酶的进一步的深入了解,以及临床耐药患者的增多,所以临床上急需开发活性更好,作用更广泛的新型酪氨酸激酶抑制剂,从而解决这些由ROS1、NTRK、ALK等激酶蛋白融合或突变所引起的肿瘤的治疗问题。
发明内容
本发明提供了一种能同时作用于NTRK、ALK和/或ROS1等致癌蛋白的新的、高效、广谱的激酶抑制剂。
本发明第一方面,提供了一种式Ⅰ所示的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物:
Figure PCTCN2020089067-appb-000001
式Ⅰ中:
A为
Figure PCTCN2020089067-appb-000002
其中,X独立地选自下组:NR 6、O、CR 1R 2、S、S(O)或S(O) 2
B选自下组:单环芳烃、双环芳烃、单环杂芳烃或双环杂芳烃,其中,所述B的任意碳原子上的H均可以被以下取代基取代:卤素、羟基、氨基、氰基、酰基、酯基、烷基、环烷基、烷胺基、烷氧基、环烷氧基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代环烷氧基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代环烷氧基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立地选自下组:氘、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
C独立地为
Figure PCTCN2020089067-appb-000003
其中,Y独立地选自下组:O、NR A或CR 1R 2
Figure PCTCN2020089067-appb-000004
代表Z式或E式;
其中,R 1、R 2各自独立地选自下组:氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、烷氧基、环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立地选自下组:氘、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;或者R 1和R 2与其连接的C原子一起连接成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或硫杂环烷烃;其中,所述取代是指被选自下组的一个或多个基团取代:烷基、酰基、酯基、磺酰基、亚磺酰基;
R 3和R 4各自独立地选自下组:氢原子、氨基、羟基、酰基、酯基、烷基、环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立地选自下组:卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
或者R 3和R 4与其连接的C原子一起连接成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃、硫杂环烷烃或氧代基(=O);或者R A和R 4与他们邻接的原子一起形成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或硫杂环烷烃;或者R 3与Y稠合形成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或硫杂环烷烃;其中,所述取代是指被选自下组的一个或多个基团取代:烷基、酰基、酯基、磺酰基、亚磺酰基;
Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7各自独立地选自下组:N、CR 5或NR 6
R 5独立地任意选自:氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
R 6和R A各自独立地任意选自:氢原子、酰基、酯基、烷基、环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基。
在另一优选例中,A为
Figure PCTCN2020089067-appb-000005
其中,*表示手性中心;R 1、R 2、X的定义如上所述。
在另一优选例中,A为
Figure PCTCN2020089067-appb-000006
其中,*表示R构型,R 1、R 2、X的定义如上所述。
在另一优选例中,X为NH、O。
在另一优选例中,R 1和R 2各自独立地为H、烷基、卤代烷基、环烷基。
在另一优选例中,C选自下组:
Figure PCTCN2020089067-appb-000007
其中,
Z为O;
R 1、R 2、R 3、R 4和R A的定义如上所述。
在另一优选例中,C为
Figure PCTCN2020089067-appb-000008
R 1、R 2、R 3、R 4、R A的定义如上所述,或者R 1和R 4与他们连接的C原子一起稠合形成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或硫杂环烷烃,其中,所述取代是指被选自下组的一个或多个基团取代:烷基、酰基、酯基、磺酰基、亚磺酰基。
在另一优选例中,C为
Figure PCTCN2020089067-appb-000009
R 3、R 4、R A的定义如上所述,或者R 3和R 4与他们连接的C原子一起稠合形成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或硫杂环烷烃,其中,所述取代是指被选自下组的一个或多个基团取代:烷基、酰基、酯基、磺酰基、亚磺酰基。
在另一优选例中,
Figure PCTCN2020089067-appb-000010
部分为
Figure PCTCN2020089067-appb-000011
在另一优选例中,所述的式Ⅰ化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物:
Figure PCTCN2020089067-appb-000012
式Ⅰ中:
A为
Figure PCTCN2020089067-appb-000013
其中,所述X为NR 6、O、CR 1R 2、S、S(O)或S(O) 2
B任意选自单环芳烃、双环芳烃、单环杂芳烃或双环杂芳烃,其中所述B的任意碳原子上的H均可以被以下取代基取代:卤素、羟基、氨基、氰基、酯基、烷基、卤代烷基、烷胺基、烷氧基、芳基或杂芳基;
C任意选自
Figure PCTCN2020089067-appb-000014
其中所述R 1、R 2、R 3、R 4各自独立的任意选自氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基,R 1和R 2、R 2和R 3、R 3和R 4或者R 1和R 4可以连接成3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或硫杂环烷烃;
Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7各自独立的任意选自N、CR 5或NR 6
R 5、R 6各自独立的任意选自氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、环烷基、卤代烷基、卤代环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基。
在另一优选例中,所述的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物:
Figure PCTCN2020089067-appb-000015
式II中,
*表示手性中心;
X选自:NR 6、O、CR 1R 2、S、S(O)或S(O) 2
R 1和R 2不同,且各自独立地选自下组:氢原子、卤素、氨基、氰基、羟基、烷基、卤代烷基;
R 6独立地选自下组:氢原子、烷基、单取代或多取代烷基,所述单取代或多取代烷基的取代基独立地选自下组:卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
B、C、Z 1、Z 2、Z 3、Z 4、Z 5、Z 6和Z 7的定义如上所述。
在另一优选例中,Z 1、Z 4、Z 5均为N。
在另一优选例中,Z 2、Z 4、Z 6均为N。
在另一优选例中,Z 2、Z 3、Z 4、Z 6均为N。
在另一优选例中,Z 3、Z 6、Z 7均为CR 5,其中,R 5独立地选自下组:氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;优选地R 5为H或卤素。
在另一优选例中,所述的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,其中,B独立地选自下组:
Figure PCTCN2020089067-appb-000016
Figure PCTCN2020089067-appb-000017
其中,
Figure PCTCN2020089067-appb-000018
为单键或双键;
Z 8、Z 9各自独立地选自:CR 11或N;
P独立地选自:O、NH、S;
Figure PCTCN2020089067-appb-000019
为双键时,Q独立地选自:CR 11或N;当
Figure PCTCN2020089067-appb-000020
为单键时,Q独立地选自:O、S、CR 11R 12或NH;
各R 7各自独立地选自下组:氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、环烷基、烷氧基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立地选自下组:氘、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
R 11和R 12各自独立地选自下组:H、羟基、卤素、氨基、氰基、酰基、烷基、卤代烷基、烷氧基、卤代烷氧基;
e为0、1、2、3或4。
在另一优选例中,B独立地选自下组:
Figure PCTCN2020089067-appb-000021
Figure PCTCN2020089067-appb-000022
其中,Z 8、Z 9各自独立地选自:CR 11或N;
各R 7各自独立地选自下组:氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、环烷基、烷氧基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自氘、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
各R 11独立地选自下组:H、羟基、卤素、氨基、氰基、酰基、烷基、卤代烷基、烷氧基、卤代烷氧基;
e为0、1、2;
P、Q、
Figure PCTCN2020089067-appb-000023
的定义如上所述。
在另一优选例中,B独立地为
Figure PCTCN2020089067-appb-000024
其中,Z 9为CR 11或N;
各R 7各自独立地选自下组:氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、环烷基、烷氧基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自下组:氘、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
各R 11独立地选自下组:H、羟基、卤素、氨基、氰基、酰基、烷基、卤代烷基、烷氧基、卤代烷氧基;
e为0、1、2。
在另一优选例中,所述的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,其中,C为
Figure PCTCN2020089067-appb-000025
其中,R 3和R 4的定义如上所述。
在另一优选例中,R 3和R 4各自独立地选自下组:氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、单取代或多取代烷基、单取代或多取代环烷基;所述单取代或多取代烷基、单取代或多取代环烷基的取代基独立地选自下组:卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基。
在另一优选例中,R 3和R 4与其连接的C原子一起连接成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或氧代基(=O);其中,所述取代是指被选自下组的一个或多个基团取代:烷基、酰基、酯基、磺酰基、亚磺酰基。
在另一优选例中,所述的式I化合物或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,具有选自下组的一个或多个特征:
A为
Figure PCTCN2020089067-appb-000026
其中,X为NH或O;R 1和R 2各自独立地为H、烷基、卤代烷基;
B独立地为
Figure PCTCN2020089067-appb-000027
其中,Z 9为CR 11或N;
各R 7各自独立地选自下组:氢原子、卤素、羟基、酰基、烷基、环烷基、烷氧基、单取 代或多取代烷基、单取代或多取代烷氧基;所述单取代或多取代烷基、单取代或多取代烷氧基的取代基独立的任意选自下组:氘、卤素、氨基、氰基、羟基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基;
各R 11独立地选自下组:H、羟基、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基;
e为0、1、2;
C独立地为
Figure PCTCN2020089067-appb-000028
优选地C为
Figure PCTCN2020089067-appb-000029
其中,Y选自下组:O、CR 1R 2;R 3和R 4各自独立地选自下组:H、烷基、单取代或多取代烷基、苯基、吡啶基、单取代或多取代苯基、单取代或多取代吡啶基;或者R 3和R 4与其连接的C原子一起构成取代或未取代的3-8元环烷基或杂环基,所述取代是指被选自下组的一个或多个基团取代:卤素、烷氧基、酯基、磺酰基;
Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7各自独立地为N或CR 5,其中,R 5选自下组:H、卤素。
在另一优选例中,所述的式I化合物或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,其具有式III所示的结构,
Figure PCTCN2020089067-appb-000030
式中,
B、X、Y、R 1、R 2、R 3和R 4的定义如上所述。
在另一优选例中,A、B、C、Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7为实施例中各具体化合物相对应的具体基团。
在另一优选例中,所述的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,其特征在于:式Ⅰ所示的化合物任意选自如下化合物:
Figure PCTCN2020089067-appb-000031
Figure PCTCN2020089067-appb-000032
在另一优选例中,式Ⅰ所示的化合物选自本发明实施例所示的化合物。
本发明第二方面,提供一种式Ⅰ化合物的可药用的盐,所述可药用的盐为无机酸盐或有机酸盐,所述无机酸盐选自盐酸盐、氢溴酸盐、氢碘酸盐、硫酸盐、硫酸氢盐、硝酸盐、磷酸盐、酸式磷酸盐;所述有机酸盐选自甲酸盐、乙酸盐、三氟乙酸盐、丙酸盐、丙酮酸盐、羟乙酸盐、乙二酸盐、丙二酸盐、富马酸盐、马来酸盐、乳酸盐、苹果酸盐、柠檬酸盐、酒石酸盐、甲磺酸盐、乙磺酸盐、羟乙磺酸盐、苯磺酸盐、水杨酸盐、苦味酸盐、谷氨酸盐、抗坏血酸盐、樟脑酸盐、樟脑磺酸盐。
本发明第三方面,提供一种药物组合物,其含有治疗有效量的第一方面所述的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,以及一种或多种药学上可以接受的载体、稀释剂或赋形剂。
本发明第四方面,提供一种第一方面所述的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,或含有式Ⅰ所示的化合物的组合物在制备用于预防和/或治疗ROS1、NTRK、ALK等介导的病理学特征的疾病的药物方面的用 途。
在另一优选例中,所述ROS1、NTRK、ALK等介导的病理学特征的疾病包括癌症、肉瘤和疼痛。
在另一优选例中,所述的癌症是乳腺癌、***、结肠癌、肺癌、胃癌、直肠癌、胰腺癌、脑癌、皮肤癌、口腔癌、***癌、骨癌、肾癌、卵巢癌、膀胱癌、肝癌、输卵管肿瘤、腹膜肿瘤、黑色素瘤、神经胶质瘤、神经胶母细胞瘤、头颈癌、乳突肾性瘤、白血病、淋巴瘤、骨髓瘤、甲状腺瘤中的任一种。
本发明提供的药物组合物可以制成适合的剂型施用。这些剂型包括适用于口服、直肠给药、局部给药、口内给药以及其他非胃肠道施用(例如,皮下、肌肉、静脉等)的那些。
本发明的药物组合物可以以符合医学实践规范的方式配制,定量和给药。给予化合物的“有效量”由要治疗的具体病症、治疗的个体、病症的起因、药物的靶点以及给药方式等因素决定。
具体实施方式
本申请发明人经过广泛而深入的研究,意外发现一种新的化合物,这些化合物对ROS1、NTRK和ALK及它们的耐药突变具有优异的抑制活性,尤其是针对耐药突变具有较好的抑制活性,且具有更好的药效学、药代动力学性能和更低的毒副作用,非常有潜力开发成目前临床急需的用于耐药病人的有效药物。
术语
除非特别说明,否则在本申请(包括说明书和权利要求书)所用的以下术语具有下面所给出的定义。
“烷基”指的是仅由碳和氢原子组成的含有1至12个碳原子的单价直链或支链饱和烃基团。“烷基”优选为1至6个碳原子的烷基基团,即C 1-C 6烷基,更优选为C 1-C 4烷基。烷基基团的实例包括但不限于甲基、乙基、丙基、异丙基、异丁基、仲丁基、叔丁基、戊基、正己基、辛基、十二烷基等。在本发明中,烷基还意在包括氘代烷基,氘代烷基实例包括但不限于CD 3、CD 2CD 3、CD 2CD 2CD 3
“烷氧基”指的是式-OR或-R'-OR基团,其中R是本文所定义的烷基基团,R'为亚烷基。烷氧基基团的实例包括但不限于甲氧基、乙氧基、异丙氧基、叔丁氧基、-CH 2O-CH 3、-CH 2CH 2-O-CH 3、-CH 2-O-CH 2CH 3等。
“卤素(卤代)”是指氟、氯、溴或碘取代基。
“卤代烷基”指的是其中一个或多个氢被相同或不同的卤素代替的本文所定义的烷基。“卤代烷基”优选卤代C 1-C 6烷基,更优选地为卤代C 1-C 4烷基,卤代烷基的实例包括-CH 2Cl、-CH 2CF 3、-CH 2CCl 3、全氟烷基(例如,-CF 3-、-CF 2CF 3)等。
“卤代烷氧基”指的是式-OR基团,其中R是本文所定义的卤代烷基基团。卤代烷氧基基 团的实例包括但不限于三氟甲氧基、二氟甲氧基、2,2,2-三氟乙氧基等。
“环烷基”指的是由单-或二环组成的单价饱和碳环基团,其具有3-12个(C 3-C 12)、优选3-10个(C 3-C 10)、更优选3-6个环原子(C 3-C 6)。环烷基可以任选地被一个或多个取代基所取代,其中各取代基独立地为羟基、烷基、烷氧基、卤素、卤代烷基、氨基、单烷基氨基或二烷基氨基。环烷基基团的实例包括但不限于环丙基、环丁基、环戊基、环己基、环庚基等。
“环烷氧基”指的是式-OR基团,其中R为如本文所定义的环烷基。示例性的环烷基氧基包括环丙基氧基、环丁基氧基、环戊基氧基、环己基氧基等。
“酰基”指的是式-C(O)R基团,其中R为如本文所定义的烷基或烷氨基。“酰基”优选地为-C(O)C 1-C 6烷基、-C(O)NH 2、-C(O)NHC 1-C 6烷基、-C(O)N(C 1-C 6烷基) 2,更优选地为-C(O)C 1-C 3烷基、-C(O)NH 2、-C(O)NHC 1-C 3烷基、-C(O)N(C 1-C 3烷基) 2,示例性的酰基包括乙酰基、正丙酰基、异丙酰基、正丁酰基、异丁酰基、叔丁酰基、-C(O)NH 2、-C(O)NHCH 3、-C(O)N(CH 3) 2等。
“烷氨基”指的是式-NRaRb基团,其中Ra和Rb相同或不同,且各自独立地为H或如本文所定义的烷基。
酯基是指式-C(O)OR的基团,其中R为如本文所定义的烷基。酯基优选地为-C(O)OC 1-C 6烷基,更优选地为-C(O)OC 1-C 4烷基,示例性的酯基包括-C(O)OMe、-C(O)OEt、-C(O)O-C(CH 3) 3等。
磺酰基是指式-S(O) 2-R,其中R为如本文所定义的烷基。磺酰基优选地为-S(O) 2-C 1-C 6烷基,示例性地包括-S(O) 2-Me、-S(O) 2-Et等。
磺酰基指式-SO-R,其中R为如本文所定义的烷基。亚磺酰基优选地为-SO-C 1-C 6烷基,示例性地包括-SO-Me、-SO-Et等。
“烷硫基”指的是式-SRa基团,其中Ra为H或如本文所定义的烷基。
“环烷氨基”指的是式-NRaRb基团,其中Ra为H、如本文所定义的烷基或如本文所定义的环烷基,Rb为如本文所定义的环烷基;或者Ra和Rb与其连接的N原子一起形成3-6元含N杂环基,如四氢吡咯基。
“杂环(基)”是指完全饱和的或部分不饱和的的环状基团(包含但不限于如3-7元单环,6-11元双环,或8-16元三环***),其中至少有一个杂原子存在于至少有一个碳原子的环中。每个含有杂原子的杂环带有1、2、3或4个选自氮原子、氧原子或硫原子的杂原子,其中氮原子或硫原子可以被氧化,氮原子也可以被季铵化。杂环烷烃(基)是指完全饱和的杂环(基)。杂环基团可以连接到环或环系分子的任何杂原子或碳原子的残基上。典型的单环杂环包括但不限于氮杂环丁烷基、吡咯烷基、氧杂环丁烷基、吡唑啉基、咪唑啉基、咪唑烷基、噁唑烷基、异噁唑烷基、噻唑烷基、异噻唑烷基、四氢呋喃基、哌啶基、哌嗪基、2-氧代哌嗪基、2-氧代哌啶基、2-氧代吡咯烷基、六氢吖庚因基、4-哌啶酮基、四氢吡喃基、***啉基、硫代***啉基、硫代***啉亚砜基、硫代***啉砜基、1,3-二噁烷基和四氢-1,1-二氧噻吩等。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。
“芳烃(基)”是指芳香环状烃类化合物基团,具有1-5个环,尤其指单环和双环基团,凡含有两个或两个以上芳香环(双环等),芳基基团的芳香环可由单键联接(如联苯),或稠合(如萘、蒽等等)。芳基优选地为C6-C12芳基,是指包含6、7、8、9、10、11或12个环碳原子的芳香环状烃类化合物基团。芳基(尤其指单环和双环基团)的实例包括但不限于苯基、联苯基或萘基。芳基可以与杂环基通过单键或者任意两个相邻的环C原子稠合,例如:苯并四氢呋喃基、苯并四氢吡喃基、苯并二氧六环基、
Figure PCTCN2020089067-appb-000033
等。
“杂芳烃(基)”指的是5至12个环原子(5-12元)的单环、二环或三环基团,其含有至少1个(如1、2或3个)选自N、O或S的环杂原子、剩余的环原子是C的芳环,应当清楚地是,杂芳基的连接点应当位于杂芳环上。杂芳基优选具体5-8个环原子(5-8元),更优选具有5-6个环原子(5-6元)。杂芳基基团的实例包括但不限于:咪唑基、
Figure PCTCN2020089067-appb-000034
唑基、异
Figure PCTCN2020089067-appb-000035
唑基、噻唑基、异噻唑基、
Figure PCTCN2020089067-appb-000036
二唑基、噻二唑基、吡嗪基、噻吩基、呋喃基、吡喃基、吡啶基、吡咯基、吡唑基、嘧啶基、喹啉基、异喹啉基、苯并呋喃基、、苯并噻吩基、苯并噻喃基、苯并咪唑基、苯并
Figure PCTCN2020089067-appb-000037
唑基、苯并
Figure PCTCN2020089067-appb-000038
二唑基、苯并噻唑基、苯并噻二唑基、苯并吡喃基、吲哚基、异吲哚基、***基、三嗪基、喹喔啉基、嘌呤基、喹唑啉基、喹嗪基、萘啶基、蝶啶基、咔唑基、氮杂
Figure PCTCN2020089067-appb-000039
基、二氮杂
Figure PCTCN2020089067-appb-000040
基、吖啶基等。
“多取代”是指包括两个或两个以上的取代。
本发明中,所述烷基、烷氧基、环烷基、杂环基、芳基、杂芳基等基团,在未特别说明的情况下包括取代的烷基、烷氧基、环烷基、杂环基、芳基、杂芳基等,所述取代基例如(但并不限于):卤素、羟基、氰基、酰基、磺酰基、酯基、亚磺酰基、烷基、环烷基、杂环基、芳基、杂芳基、酰基、酯基等。
“氘代物”指的是化合物中一个氢原子(H)或多个氢原子(H)被氘原子(D)取代后所得到的化合物。
活性成分
如本文所用,术语“本发明的化合物”或“本发明的活性成分”可互换使用,指式I化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物。
式I化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物具有如下结构,
Figure PCTCN2020089067-appb-000041
其中,A、B、C、Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7的定义如上所述。
优选地,式I化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物具有式III所示的结构,
Figure PCTCN2020089067-appb-000042
式中,
B、X、Y、R 1、R 2、R 3和R 4的定义如上所述。
本发明中的化合物可能形成的盐也是属于本发明的范围。除非另有说明,本发明中的化合物被理解为包括其盐类。在此使用的术语“盐”,指用无机或有机酸和碱形成酸式或碱式的盐。此外,当本发明中的化合物含一个碱性片段时,它包括但不限于吡啶或咪唑,含一个酸性片段时,包括但不限于羧酸,可能形成的两性离子(“内盐”)包含在术语“盐”的范围内。药学上可接受的(即无毒,生理可接受的)盐是首选,虽然其他盐类也有用,例如可以用在制备过程中的分离或纯化步骤。本发明的化合物可能形成盐,例如,化合物I与一定量如等当量的酸或碱反应,在介质中盐析出来,或在水溶液中冷冻干燥得来。
本发明中的化合物含有的碱性片段,包括但不限于胺或吡啶或咪唑环,可能会和有机或无机酸形成盐。可以成盐的典型的酸包括盐酸盐、氢溴酸盐、氢碘酸盐、硫酸盐、硫酸氢盐、硝酸盐、磷酸盐、酸式磷酸盐;所述有机酸盐选自甲酸盐、乙酸盐、三氟乙酸盐、丙酸盐、丙酮酸盐、羟乙酸盐、乙二酸盐、丙二酸盐、富马酸盐、马来酸盐、乳酸盐、苹果酸盐、柠檬酸盐、酒石酸盐、甲磺酸盐、乙磺酸盐、羟乙磺酸盐、苯磺酸盐、水杨酸盐、苦味酸盐、谷氨酸盐、抗坏血酸盐、樟脑酸盐、樟脑磺酸盐等等。
本发明的某些化合物可能含有的酸性片段,包括但不限于羧酸,可能会和各种有机或无机碱形成盐。典型的碱形成的盐包括铵盐、碱金属盐如钠、锂、钾盐,碱土金属盐如钙、镁盐和有机碱形成的盐(如有机胺),如苄星、二环已基胺、海巴胺(与N,N-二(去氢枞 基)乙二胺形成的盐)、N-甲基-D-葡糖胺、N-甲基-D-葡糖酰胺、叔丁基胺,以及和氨基酸如精氨酸、赖氨酸等等形成的盐。碱性含氮基团可以与卤化物季铵盐,如小分子烷基卤化物(如甲基、乙基、丙基和丁基的氯化物、溴化物及碘化物),二烷基硫酸盐(如,硫酸二甲酯、二乙酯,二丁酯和二戊酯),长链卤化物(如癸基、十二烷基、十四烷基和十四烷基的氯化物、溴化物及碘化物),芳烷基卤化物(如苄基和苯基溴化物)等等。
本发明中化合物的前药及溶剂合物也在涵盖的范围之内。此处术语“前药”是指一种化合物,在治疗相关疾病时,经过代谢或化学过程的化学转化而产生本发明中的化合物、盐、或溶剂合物。本发明的化合物包括溶剂合物,如水合物。
本发明中的化合物、盐或溶剂合物,可能存在的互变异构形式(例如酰胺和亚胺醚)。所有这些互变异构体都是本发明的一部分。
所有化合物的立体异构体(例如,那些由于对各种取代可能存在的不对称碳原子),包括其对映体形式和非对映形式,都属于本发明的设想范围。本发明中的化合物独立的立体异构体可能不与其他异构体同时存在(例如,作为一个纯的或者实质上是纯的光学异构体具有特殊的活性),或者也可能是混合物,如消旋体,或与所有其他立体异构体或其中的一部分形成的混合物。本发明的手性中心有S或R两种构型,由理论与应用化学国际联合会
(IUPAC)1974年建议定义。外消旋形式可通过物理方法解决,例如分步结晶,或通过衍生为非对映异构体分离结晶,或通过手性柱色谱法分离。单个的光学异构体可通过合适的方法由外消旋体得到,包括但不限于传统的方法,例如与光学活性酸成盐后再结晶。
本发明中的化合物,依次通过制备、分离纯化获得的该化合物其重量含量等于或大于90%,例如,等于或大于95%,等于或大于99%(“非常纯”的化合物),在正文描述列出。此处这种“非常纯”本发明的化合物也作为本发明的一部分。
本发明的化合物所有的构型异构体都在涵盖的范围之内,无论是混合物、纯的或非常纯的形式。在本发明化合物的定义包含顺式(Z)和返式(E)两种烯烃异构体,以及碳环和杂环的顺式和反式异构体。
在整个说明书中,基团和取代基可以被选择以提供稳定的片段和化合物。
特定官能团和化学术语定义都详细介绍如下。对本发明来说,化学元素与Periodic Table of the Elements,CAS version,Handbook of Chemistry and Physics,75 th Ed.中定义的一致。特定官能团的定义也在其中描述。此外,有机化学的基本原则以及特定官能团和反应性在“Organic Chemistry”,Thomas Sorrell,University Science Books,Sausalito:1999,也有说明,其全部内容纳入参考文献之列。
本发明的某些化合物可能存在于特定的几何或立体异构体形式。本发明涵盖所有的化合物,包括其顺式和反式异构体、R和S对映异构体、非对映体、(D)型异构体、(L)型异构体、外消旋混合物和其它混合物。另外不对称碳原子可表示取代基,如烷基。所有异构体以及它们的混合物,都包涵在本发明中。
按照本发明,同分异构体的混合物含有异构体的比率可以是多样的。例如,在只有两 个异构体的混合物可以有以下组合:50:50,60:40,70:30,80:20,90:10,95:5,96:4,97:3,98:2,99:1,或100:0,异构体的所有比率都在本发明范围之内。本专业内一般技术人员容易理解的类似的比率,及为更复杂的异构体的混合物的比率也在本发明范围之内。
本发明还包括同位素标记的化合物,等同于原始化合物在此公开。不过实际上对一个或更多的原子被与其原子量或质量序数不同的原子取代通常会出现。可以列为本发明的化合物同位素的例子包括氢,碳,氮,氧,磷,硫,氟和氯同位素,分别如 2H、 3H、 13C、 11C、 14C、 15N、 18O、 17O、 31P、 32P、 35S、 18F和 36Cl。本发明中的化合物,或对映体,非对映体,异构体,或药学上可接受的盐或溶剂化物,其中含有上述化合物的同位素或其他同位素原子都在本发明的范围之内。本发明中某些同位素标记化合物,例如 3H和 14C的放射性同位素也在其中,在药物和底物的组织分布实验中是有用的。氚,即 3H和碳-14,即 14C,它们的制备和检测比较容易。是同位素中的首选。此外,较重同位素取代如氘,即 2H,由于其很好的代谢稳定性在某些疗法中有优势,例如在体内增加半衰期或减少用量,因此,在某些情况下可以优先考虑。同位素标记的化合物可以用一般的方法,通过用易得的同位素标记试剂替换为非同位素的试剂,用批露在示例中的方案可以制备。
如果要设计一个本发明的化合物特定的对映体的合成,它可以不对称合成制备,或用手性辅剂衍生化,将所产生的非对映混合物分离,再除去手性辅剂而得到纯的对映体。另外,如果分子中含有一个碱性官能团,如氨基酸,或酸性官能团,如羧基,可以用合适的光学活性的酸或碱的与之形成非对映异构体盐,再通过分离结晶或色谱等常规手段分离,然后就得到了纯的对映体。
如本文所述,本发明中的化合物可与任何数量取代基或官能团取而扩大其包涵范围。通常,术语“取代”不论在术语“可选”前面或后面出现,在本发明配方中包括取代基的通式,是指用指定结构取代基,代替氢自由基。当特定结构中的多个在位置被多个特定的取代基取代时,取代基每一个位置可以是相同或不同。本文中所使用的术语“取代”包括所有允许有机化合物取代。从广义上讲,允许的取代基包括非环状的、环状的、支链的非支链的、碳环的和杂环的,芳环的和非芳环的有机化合物。在本发明中,如杂原子氮可以有氢取代基或任何允许的上文所述的有机化合物来补充其价态。此外,本发明是无意以任何方式限制允许取代有机化合物。本发明认为取代基和可变基团的组合在以稳定化合物形式在疾病的治疗上是很好的。此处术语“稳定”是指具有稳定的化合物,在足够长的时间内检测足以维持化合物结构的完整性,最好是在足够长的时间内都在效,本文在此用于上述目的。
本申请所涉及的化合物及其药学可接受的盐的代谢产物,以及可以在体内转变为本申请所涉及的化合物及其药学可接受的盐的结构的前药,也包含在本申请的权利要求中。
制备方法
本发明化合物可以任选将在本说明书中描述的或本领域已知的各种合成方法组合起来而方便地制得,这样的组合可由本发明所属领域的技术人员容易地进行。
通常,在制备流程中,各反应通常在惰性溶剂中,在-60℃~100℃,优选-60℃~80℃下进行。反应时间通常为0.1小时-60小时,较佳地为0.5-48小时。
优选的合成路线如下:
Figure PCTCN2020089067-appb-000043
式中,Z为O;R为C1-C6烷基;
A、B、C、Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7、R 3、R 4、R A的定义如上所述;
其中:路线1中:(1)在惰性溶剂(如乙醇、甲醇)中,化合物1和化合物2在碱(如碳酸钠、碳酸钾、氢氧化钠、三乙胺、吡啶等)的作用下,发生亲核取代反应,生成化合物3;(2)在惰性溶剂(如乙醇、甲醇)中,在碱(例如碳酸钠、碳酸钾、氢氧化钠、三乙胺、吡啶等)的作用下,化合物3与盐酸羟胺反应,生成化合物4;(3)在惰性溶剂(如1,2-二氯乙烷和/或冰醋酸)中,化合物4与二甲氧基缩丙酮发生反应,得到最终产物5。
路线2中:(1)在惰性溶剂(如乙醇、甲醇)中,化合物1和化合物2在碱(例如碳酸钠、碳酸钾、氢氧化钠、三乙胺、吡啶等)的作用下,发生亲核取代反应,生成化合物3;(2)在惰性溶剂(如乙醇、甲醇)中,在碱(例如碳酸钠、碳酸钾、氢氧化钠、三乙胺、吡啶等)的作用下,化合物3与盐酸羟胺反应,生成化合物4;(3)在惰性溶剂(如1,2-二氯乙烷和/或冰醋酸)中,化合物4与二甲氧基缩丙酮,得到最终产物5。
路线3中:(1)在惰性溶剂(如甲苯)中,化合物1和化合物2在碱(例如叔丁醇钠、叔丁醇钾、氢化钠、氢化钾、碳酸钾、碳酸铯、磷酸钾、氢氧化钾、氢氧化钠等)的作用下,发生亲核取代反应,生成化合物3;(2)在惰性溶剂(如甲苯)中,三甲基铝存在下,化合物3与
Figure PCTCN2020089067-appb-000044
发生反应,得到最终产物4。
本发明的起始原料都是已知并有市售的,或者可以按照本领域已报道的文献资料合成的。
药物组合物和施用方法
本发明所述的药物组合物用于预防和/或治疗以下疾病:炎症、癌症、心血管疾病、感染、免疫性疾病、代谢性疾病。
本发明所述化合物可以与已知的治疗或改进相似病状的其他药物联用。联合给药时,原来药物的给药方式和剂量可以保持不变,而同时或随后服用本发明的化合物。当本发明化合物与其它一种或几种药物同时服用时,可以优选使用同时含有一种或几种已知药物和本发明化合物的药用组合物。药物联用也包括在重叠的时间段服用本发明化合物与其它一种或几种已知药物。当本发明化合物与其它一种或几种药物进行药物联用时,本发明化合物或已知药物的剂量可能比它们单独用药的剂量低。
本发明所述药物组合物的剂型包括(但并不限于):注射剂、片剂、胶囊剂、气雾剂、栓剂、膜剂、滴丸剂、外用擦剂、控释型或缓释型或纳米制剂。
本发明的药物组合物包含安全有效量范围内的本发明化合物或其药理上可接受的盐及药理上可以接受的赋形剂或载体。其中“安全有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg本发明化合物/剂,更佳地,含有10-1000mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。
“药学上可以接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能和本发明的化合物以及它们之间相互掺和,而不明显降低化合物的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如吐温
Figure PCTCN2020089067-appb-000045
)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
本发明化合物或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、瘤内、直肠、肠胃外(静脉内、肌肉内或皮下)、和局部给药。
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性化合物与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和***胶;(c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;(e)缓溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性化合物外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。
除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。
除了活性化合物外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。
用于局部给药的本发明化合物的剂型包括软膏剂、散剂、贴剂、喷射剂和吸入剂。活性成分在无菌条件下与生理上可接受的载体及任何防腐剂、缓冲剂,或必要时可能需要的推进剂一起混合。
本发明治疗方法可以单独施用,或者与其它治疗手段或者治疗药物联用。
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1-2000mg,优选10-1000mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。
本发明还提供了一种药物组合物的制备方法,包括步骤:将药学上可接受的载体与本发明所述化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药进行混合,从而形成药物组合物。
本发明还提供了一种治疗方法,它包括步骤:给需要治疗的对象施用本发明中所述化合物,或其药学上可接受的盐、立体异构体、溶剂化物或前药,或施用本发明所述的药物组合物,用于选择性地抑制ROS1、NTRK、ALK等的融合突变及其耐药突变。
本发明具有以下主要优点:
(1)本发明化合物对ROS1、NTRK、ALK激酶均有很好的抑制能力,尤其是对这些靶点耐药突变的活性非常出色;
(2)本发明化合物具有更好的药效学、药代动力学性能和更低的毒副作用;
(3)本发明化合物非常有潜力开发成目前临床急需的用于耐药病人的有效药物。
下面对本发明的技术方案作进一步的说明,但本发明的保护范围不限于此。
下面列举一些具体实施例进行说明。
实施例1
合成路线:
Figure PCTCN2020089067-appb-000046
反应步骤:
(1)化合物2的合成:100mL单口瓶,冷凝管,氩气保护。称取化合物1(5.2g),加入的甲醇(50mL)和的四氢呋喃(25mL),氩气保护下升温到60℃,缓慢滴加1M/L甲醇钠的甲醇溶液(32mL)(自制),1小时滴完,然后60℃下搅拌过夜。次日,旋干溶剂,加水和乙酸乙酯分液,然后用乙酸乙酯再萃取一次,将有机相合并,干燥,旋干,过柱,得到4.21g油状物产品。 1H NMR(400MHz,CDCl 3)δ10.48(d,J=1.0Hz,1H),7.31(dd,J=9.2,8.2Hz,1H),6.88(dd,J=9.2,3.7Hz,1H),3.92(s,3H)。
(2)化合物3的合成:250mL单口瓶,冷凝管上面密封,加入化合物2(4.01g)、(R)-叔丁亚磺酰胺(3.87g,1.5eq)、钛酸四乙酯(9.73g,2.0eq)、四氢呋喃(100mL),80℃下搅拌过夜,次日降温。加入大量的饱和盐水和乙酸乙酯,分液,水相再用二氯甲烷萃取一次,有机相合并,干燥,旋干,过柱,得到产品4.73g油状物, 1H NMR(400MHz,CDCl 3)δ8.93(s,1H),7.23(dd,J=9.1,8.4Hz,1H),6.85(dd,J=9.2,3.9Hz,1H),3.88(s,3H),1.30(s,9H)。
(3)化合物4的合成:250mL三口瓶中,加入化合物3(4.73g)、四氢呋喃(200mL),氩气保护,先室温搅拌10min,降温到-10℃,加入3M的甲基氯化镁的四氢呋喃溶液(25ml,3eq),反应慢慢升温到室温,搅拌过夜。次日,TLC监测反应完全。加入水和乙酸乙酯分液,然后用乙酸乙酯再萃取一次,有机相合并,干燥,旋干,过柱,得到4.525g固体产品 1H NMR(400MHz,CDCl 3)δ7.01(td,J=9.2,8.4Hz,1H),6.76(ddd,J=9.1,6.9,4.1Hz,1H),5.33–4.39(m,2H),3.87(d,J=6.2Hz,3H),1.57(dd,J=56.9,7.0Hz,3H),1.17(d,J=28.1Hz,9H)。
(4)化合物5的合成:500mL单口瓶,加入化合物4(4.525g)、盐酸/二氧六环(150mL)。室温搅拌4小时,TLC监测,原料反应完。直接旋干溶剂,加水,用碳酸钠水溶液调pH到9-10。用乙酸乙酯萃取,萃取两次,干燥,浓缩,得到2.86g浅黄色油状物。
(5)化合物6的合成:100mL单口瓶,化合物5(1.06g)、5-氯吡唑并嘧啶-3-甲腈 (0.93g,1.0eq)、乙醇(60mL)、三乙胺(1.581g,3.0eq)加入单口瓶中,加冷凝管,氩气保护,先室温搅拌10min,之后55℃反应过夜。次日,TLC监测反应完全,直接抽滤,得到0.93g粉末固体产品。 1H NMR(400MHz,DMSO)δ8.57(d,J=7.6Hz,1H),8.46(d,J=7.3Hz,1H),8.23(s,1H),7.26(t,J=9.0Hz,1H),7.02(dd,J=9.2,4.3Hz,1H),6.59(d,J=7.6Hz,1H),5.82(q,J=7.1Hz,1H),3.89(s,3H),1.56(d,J=7.2Hz,3H)。
(6)化合物7的合成:100mL单口瓶中,加入化合物6(0.93g)、无水碳酸钾(1.12g,3eq)、盐酸羟胺(0.563g,3eq)、乙醇(40mL)、二氧六环(20mL),80℃反应过夜。次日,TLC监测反应完全。直接旋干溶剂,加入水和乙酸乙酯,水相再用二氯甲烷萃取一次,有机相合并,干燥,旋干,过柱,得到纯品0.411g, 1H NMR(400MHz,DMSO)δ9.02(s,1H),8.46(d,J=7.6Hz,1H),8.13(d,J=7.5Hz,1H),7.88(s,1H),7.26(t,J=9.0Hz,1H),7.04(dd,J=9.2,4.3Hz,1H),6.44(d,J=7.6Hz,1H),5.89–5.61(m,3H),3.87(s,3H),1.54(d,J=7.2Hz,3H)。
化合物实施例1的合成:称取化合物7(0.411g),加入二甲氧基缩丙酮(0.457g,4eq)、1,2-二氯乙烷(15mL)、冰醋酸(7.5mL),80℃下搅拌4小时,TLC监测反应完全,直接旋干溶剂,加入水和二氯甲烷,干燥,旋干,过柱,得到170mg最终产品。 1H NMR(400MHz,CDCl 3)δ8.25–8.12(m,2H),7.05(dd,J=9.1,8.2Hz,1H),6.80(dd,J=9.1,4.0Hz,1H),6.08(t,J=30.1Hz,4H),3.91(s,2H),1.58(t,J=5.8Hz,8H)。
实施例2
合成路线:
Figure PCTCN2020089067-appb-000047
反应步骤:
(1)化合物2的合成:500mL三口瓶,加温度计,冷凝管,氩气保护。称取化合物1(14.77g),加入甲醇(200mL)和四氢呋喃(85mL),氩气保护下升温到60℃,缓慢滴加1M/L甲醇钠的甲醇溶液(85mL)(自制),1小时滴完。之后60℃下搅拌过夜。次日,旋干溶剂,加水和乙酸乙酯萃取,然后用乙酸乙酯再萃取一次,得到12g油状物产品。
(2)化合物3的合成:500mL单口瓶,冷凝管上面密封,加入化合物2(12g)、(R)-叔丁亚磺酰胺(19.52g,2.5eq)、钛酸四乙酯(36.8g,2.5eq)、四氢呋喃(300mL)。80℃下搅拌过夜,次日降温。加入大量的饱和盐水和乙酸乙酯分层,水相再用二氯甲烷萃取一次,有机相合并,干燥,旋干,过柱,得到产品3.0g油状物。
(3)化合物4的合成:500mL单口瓶中,加入化合物3(3.0g)、四氢呋喃(200mL),氩气保护,先室温搅拌10min,之后干冰降温到-60℃,加入硼氢化钠(1.2g, 3.0eq),反应慢慢升温到室温,搅拌过夜。次日,TLC监测反应完全。加入水和乙酸乙酯萃取,然后用乙酸乙酯再萃取一次。合并有机相,干燥,旋干,过柱,得到2.25g油状物产品。
(4)化合物5的合成:100mL单口瓶,加入化合物4(2.25g)、盐酸/二氧六环(50mL),室温搅拌4小时,检测,原料反应完。直接旋干溶剂,加水,用碳酸钠水溶液调pH到9-10。用乙酸乙酯萃取,萃取两次,干燥,浓缩得到1.8g浅黄色油状物。
(5)化合物6的合成:100mL单口瓶,化合物5(0.92g)、5-氯吡唑并嘧啶-3-甲腈(0.81g,1.1eq)、乙醇(40mL)、三乙胺(1.25g,3.0eq)加入其中,加冷凝管,氩气保护,先室温搅拌10min,之后55℃反应过夜。次日,TLC监测反应完全,直接旋干,加水和乙酸乙酯萃取,合并有机相,干燥,旋干,过柱,得到0.95g油状物产品。
(6)化合物7的合成:100mL单口瓶中,加入化合物6(0.95g)、无水碳酸钾(0.8g,2eq)、盐酸羟胺(0.4g,2eq)、乙醇(40mL)、二氧六环(20mL)。80℃反应过夜,次日,TLC监测反应完全。直接旋干溶剂,加入水和乙酸乙酯,水相再用二氯甲烷萃取一次。合并有机相,干燥,旋干,过柱,得到纯品0.4g, 1H NMR(400MHz,DMSO)δ9.02(s,1H),8.46(d,J=7.6Hz,1H),8.23(d,J=7.1Hz,1H),7.89(s,1H),7.20(ddd,J=11.1,9.2,5.2Hz,1H),6.98(td,J=9.6,3.8Hz,1H),6.40(d,J=7.6Hz,1H),5.78(d,J=11.3Hz,2H),5.55–5.33(m,1H),3.91(d,J=1.7Hz,3H),1.59(d,J=7.1Hz,3H)。
(7)化合物实施例2的合成:称取化合物7(0.3g),加入二甲氧基缩丙酮(0.345g,4eq)、1,2-二氯乙烷(10mL)、冰醋酸(5mL),80℃下搅拌4小时,TLC监测反应完全,直接旋干溶剂,加入水和二氯甲烷。干燥,旋干,过柱,得到130mg最终产物。 1H NMR(400MHz,CDCl 3)δ8.20(s,1H),8.15(d,J=7.6Hz,1H),6.97(ddd,J=10.8,9.2,5.3Hz,1H),6.75(td,J=9.4,3.7Hz,1H),6.34(s,1H),6.06(d,J=7.5Hz,1H),5.79–5.59(m,2H),4.03(d,J=1.8Hz,2H),1.72–1.64(m,5H),1.60(s,3H)。
实施例3
合成路线:
Figure PCTCN2020089067-appb-000048
反应步骤:
(1)化合物2的合成:500mL三口瓶,加温度计,冷凝管,氩气保护。称取化合物1(9.65g),加入二氯甲烷(350mL)和对甲苯磺酰氯(23.84g,1.3eq),氩气保护下降温到0℃,缓慢滴加三乙胺(29.24g,3.0eq),10min滴完,之后室温下搅拌过夜。次日,加水和二氯甲烷,二氯甲烷再萃取一次,干燥,旋干,过柱,得到20g产品。
(2)化合物4的合成:500mL三口瓶,加温度计,冷凝管,氩气保护。称取化合物2 (20g),加入N,N-二甲基甲酰胺(350mL)和化合物3(12.13g,1eq),再加入无水碳酸钾(54.33g,5eq),氩气保护下升温到60℃,搅拌过夜。次日,加水和乙酸乙酯,乙酯再萃取一次,干燥,旋干,过柱,得到13g油状物产品。收率70.3%。 1H NMR(400MHz,CDCl 3)δ7.51(dd,J=8.8,3.3Hz,1H),7.19(ddd,J=9.0,7.2,3.3Hz,1H),6.88(dd,J=9.0,3.9Hz,1H),4.43(q,J=7.9Hz,2H),2.63(s,3H)。
(3)化合物5的合成:500mL单口瓶,冷凝管上面密封,加入化合物4(13g)、(R)-叔丁亚磺酰胺(13.33g,2eq)、钛酸四乙酯(25.13g,2eq)、四氢呋喃(300mL),80℃下搅拌过夜,次日降温。加入大量的饱和盐水和乙酸乙酯,分层,水相再用二氯甲烷萃取一次,有机相合并,干燥,旋干,过柱,得到产品9.6g油状物,收率51.6%。
(4)化合物6的合成:250mL单口瓶中,加入化合物5(9.6g)、四氢呋喃(150mL),氩气保护,先室温搅拌10min,之后干冰降温到-60℃,加入硼氢化钠(3.23g,3eq),反应慢慢升温到室温,搅拌过夜。次日,TLC检测。加入饱和氯化铵水溶液和乙酸乙酯,乙酸乙酯再萃取一次。合并有机相,干燥,旋干,过柱,得到0.9g油状物产品。 1H NMR(400MHz,CDCl 3)δ7.05(dd,J=8.8,3.1Hz,1H),6.94(ddd,J=8.9,7.7,3.1Hz,1H),6.78(dd,J=9.0,4.3Hz,1H),4.67(p,J=6.8Hz,1H),4.47–4.32(m,2H),3.79(d,J=6.9Hz,1H),1.50(d,J=6.8Hz,3H),1.21(s,9H).
(5)化合物7的合成:100mL单口瓶,加入化合物6(0.9g)、盐酸/二氧六环(50mL),室温搅拌4小时,TLC检测,原料反应完。直接旋干溶剂,得到0.865g浅黄色固体。
(6)化合物9的合成:100mL单口瓶,加入化合物7(0.865g)、5-氯吡唑并嘧啶-3-甲腈(0.562g,1eq)、乙醇(40mL)、三乙胺(0.96g,3eq)。加冷凝管,氩气保护,先室温搅拌10min,之后55℃反应过夜。次日,直接旋干,加水和乙酸乙酯萃取,干燥,旋干,过柱,得到0.988g油状物产品。
(7)化合物10的合成:100mL单口瓶中,加入化合物9(0.988g)、无水碳酸钾(1.08g,3eq)、盐酸羟胺(0.544g,3eq)、乙醇(40mL)、二氧六环(20mL),80℃反应过夜,次日,TLC检测。直接旋干溶剂,加入水和乙酸乙酯,水相再用二氯甲烷萃取一次,合并有机相,干燥,旋干,过柱,得到纯品0.65g产品,收率60.5%。 1H NMR(400MHz,DMSO)δ9.00(s,1H),8.55(dd,J=38.2,7.8Hz,1H),8.28(d,J=6.5Hz,1H),7.92(d,J=33.3Hz,1H),7.18–7.02(m,3H),6.47(dd,J=77.2,7.8Hz,1H),5.63(s,2H),5.44–5.27(m,1H),5.08–4.74(m,2H),1.42(d,J=6.9Hz,3H).
化合物实施例3的合成:化合物10(0.65g)、二甲氧基缩丙酮(0.656g,4eq)、1,2-二氯乙烷(15mL)、冰醋酸(7.5mL)加入反应瓶中,80℃下搅拌4小时。直接旋干溶剂,加入水和二氯甲烷萃取,干燥,旋干,过柱,得到180mg最终产物。液相纯度99%。 1H NMR(400MHz,CDCl 3)δ8.29–8.10(m,2H),7.03(dd,J=8.6,2.9Hz,1H),7.00–6.92(m,1H),6.83(dd,J=9.0,4.2Hz,1H),6.08(d,J=7.6Hz,1H),5.79(s,1H),5.51(d,J=5.5Hz,1H),5.25 (s,1H),4.53–4.29(m,2H),1.62(s,3H),1.59(s,3H),1.47(s,3H)。
实施例4
合成路线:
Figure PCTCN2020089067-appb-000049
反应步骤:
(1)化合物2的合成:将化合物1(6.6g,42.8mmol,1eq)溶于乙腈(100mL)溶液中,加入CD 3OTs(9.72g,51.4mmol,1.2eq)和K 2CO 3(8.88g,64.2mmol,1.5eq),80℃下反应12h,待原料反应完毕后,加入水和乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,得到化合物2(7.0g,40.9mmol,收率95.5%)。
(2)化合物3的合成:将化合物2(6.0g,35.1mmol,1.0eq)溶于60mL干燥THF溶液中,加入(R)-(+)-叔丁基亚磺酰胺(8.5g,70.1mmol,2eq)和Ti(OEt) 4(16.0g,70.1mmol,2eq),70℃下反应12h,待原料反应完毕后,加入水和乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,柱层析纯化(石油醚:乙酸乙酯=4:1)得到化合物3(8.0g,29.1mmol,收率83.2%)。
(3)化合物4的合成:将化合物3(3.0g,10.9mmol,1.0eq)溶于30mL干燥THF溶液中,-50℃加入NaBH 4(1.24g,32.8mmol,3eq),-50℃下继续反应4h,待原料反应完毕后,加入饱和氯化铵水溶液淬灭,乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,柱层析纯化(石油醚:乙酸乙酯=8:1)得到化合物4(1g,3.62mmol,收率33.1%)。
(4)化合物5的合成:将化合物4(1.0g,3.62mmol,1eq)冰浴下加入4M盐酸二氧六环(10mL),0℃继续反应1h,待原料反应完毕后,加入饱和碳酸钠水溶液淬灭,乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,得到化合物5(0.5g,黄色油状液体,收率80.2%)。
(5)化合物6的合成:将化合物5(500mg,2.9mmol,1eq)溶于乙醇(8mL)中,然后加入化合物5a(622mg,3.48mmol,1.2eq)和三乙胺(881mg,8.71mmol,2eq),然后升温至60℃反应2h,待原料反应完毕后,旋干溶剂,柱层析纯化(石油醚:乙酸乙酯=2:1),得到化合物6(0.75g,2.39mmol,收率82.2%)。
MS:300(M+H+)。
(6)化合物7的合成:将化合物6(700mg,2.23mmol,1eq)溶于乙醇(10mL)中,然后加入盐酸羟胺(310mg,4.45mmol,2eq)和碳酸钾(616mg,4.45mmol,2eq),然后升温至80℃反应12h,待原料反应完毕后,加入水和乙酸乙酯萃取,用盐水洗三次,合并有机 相,无水硫酸钠干燥,旋干有机相,柱层析纯化(二氯甲烷:甲醇=50:1)得到化合物7(700mg,2.02mmol,收率90.5%)。
化合物实施例4的合成:将化合物7(400mg,1.15mmol,1eq)溶于5mL醋酸和1,2-二氯乙烷(5mL)中,然后加入化合物7a(480mg,4.61mmol,4eq),然后升温至80℃反应2h,待原料反应完毕后,加入饱和碳酸钠水溶液淬灭,乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,柱层析纯化(石油醚:乙酸乙酯=0:1),得到化合物实施例4(300mg,白色固体,0.77mmol,收率67.2%)。 1H NMR(400MHz,CDCl 3)δ8.17(d,J=7.6Hz,1H),8.15(s,1H),6.98(dd,J=8.8,3.3Hz,1H),6.95–6.88(m,1H),6.86(dd,J=8.8,4.4Hz,1H),6.17(d,J=6.4Hz,1H),5.95(s,1H),5.86(d,J=5.4Hz,1H),1.62(s,3H),1.54(d,J=6.9Hz,3H),1.44(s,3H)。
实施例5
合成路线:
Figure PCTCN2020089067-appb-000050
反应步骤:
(1)化合物2的合成:将化合物1(8g,32.6mmol,1eq)溶于干燥四氢呋喃(60mL)中,-70℃下滴加甲基溴化镁溶液(21mL,65.2mmol,3M,2eq),滴加完毕后继续保温反应2h,待原料反应完毕后,加入饱和氯化铵水溶液淬灭,乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,柱层析纯化(石油醚:乙酸乙酯=4:1)得到化合物2(2g,黄色固体,收率23.5%)。
(2)化合物3的合成:将化合物2(1.5g,5.74mmol,1eq)冰浴下加入4M盐酸二氧六环(10mL),0℃继续反应1h,待原料反应完毕后,加入饱和碳酸钠水溶液淬灭,乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,得到化合物3(0.8g,黄色油状液体,收率88.7%)。
(3)化合物4的合成:将化合物3(200mg,1.27mmol,1eq)溶于乙醇(4mL)中,然后加入化合物3a(272mg,1.53mmol,1.2eq)和三乙胺(257mg,2.55mmol,2eq),然后升温至55℃反应2h,待原料反应完毕后,旋干溶剂,柱层析纯化(石油醚:乙酸乙酯=2:1),得到化合物4(150mg白色固体,收率39.4%)。
MS:300(M+H+)。
(4)化合物5的合成:将化合物4(150mg,0.5mmol,1eq)溶于乙醇(2mL)中,然后加入盐酸羟胺(70mg,1.0mmol,2eq)和碳酸钾(138mg,1.0mmol,2eq),然后升温至80℃ 反应12h,待原料反应完毕后,加入水和乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,柱层析纯化(二氯甲烷:甲醇=50:1)得到化合物5(50mg,棕色油状液体,收率30.0%)。
化合物实施例5的合成:将化合物5(50mg,0.15mmol,1eq)溶于醋酸(0.5mL)和1,2-二氯乙烷(0.5mL)中,然后加入化合物5a(62mg,0.6mmol,4eq),然后升温至80℃反应2h,待原料反应完毕后,加入饱和碳酸钠水溶液淬灭,乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,柱层析纯化(石油醚:乙酸乙酯=0:1),得到实施例5(20mg,白色固体,收率35.7%)。 1H NMR(400MHz,CDCl 3)δ8.21(d,J=7.6Hz,1H),8.18(s,1H),7.09–6.99(m,2H),6.92(m,1H),6.16(d,J=7.6Hz,1H),5.94(s,1H),5.55(d,J=5.7Hz,1H),5.38(m,1H),1.64(s,3H),1.60(s,3H),1.50(s,3H)。MS:373(M+H+)。
实施例6
合成路线:
Figure PCTCN2020089067-appb-000051
反应步骤:
(1)化合物2的合成:将化合物1a(10g,92.5mmol,1eq)和化合物1(17.3g,97.1mmol,1.05eq)溶于200mL乙醇中,加入EtONa(8.81g,129.5mmol,1.4eq),然后升温至80℃反应12h,待原料反应完毕后,旋干溶剂,加入水,用1M HCl调节pH为2-3,有沉淀析出,沉淀过滤,干燥得到化合物2(14g,72.12mmol收率66.8%)。
(2)化合物3的合成:将化合物2(14g,72.12mmol,1eq)加入POCl 3(100mL),100℃反应12h,待原料反应完毕后,旋干溶剂,柱层析纯化得到化合物3(2.2g,9.52mmol,收率13.2%)。
(3)化合物4的合成:将化合物3(2.2g,9.52mmol,1eq)溶于乙醇(42mL)、四氢呋喃(14mL)和水(28mL)中,然后加入Zn粉(3.11g,47.6mmol,5eq)和NH 4Cl(2.04g,38.1mmol,4eq),然后20℃反应10分钟,待原料反应完毕后,加入水和乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,柱层析纯化得到化合物4(1.0g,5.09mmol,收率53.4%)。
(4)化合物5的合成:将化合物4a(500mg,3.22mmol,1eq)溶于乙醇(6mL)中,然后加入化合物4(696mg,3.54mmol,1.1eq)和三乙胺(978mg,9.67mmol,3eq),然后升温至60℃反应2h,待原料反应完毕后,旋干溶剂,柱层析纯化(石油醚:乙酸乙酯=2:1),得到化合物5(600mg白色固体,收率59.0%)。
(5)化合物6的合成:将化合物5(600mg,1.9mmol,1eq)溶于6mL乙腈中,然后加入CD 3OTs(432mg,2.28mmol,1.2eq)和碳酸钾(395mg,2.85mmol,2eq),然后升温至80℃反应2h,待原料反应完毕后,加入水和乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,柱层析纯化得到化合物6(400mg,1.2mmol,收率63.2%)。
(6)化合物7的合成:将化合物6(250mg,0.75mmol,1eq)溶于乙醇(5mL)中,然后加入盐酸羟胺(105mg,1.5mmol,2eq)和碳酸钾(208mg,1.5mmol,2eq),然后升温至80℃反应12h,待原料反应完毕后,加入水和乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,柱层析纯化(二氯甲烷:甲醇=50:1)得到化合物7(250mg,棕色油状液体,收率91.0%)。
化合物实施例6的合成:将化合物7(250mg,0.68mmol,1eq)溶于醋酸(2mL)和1,2-二氯乙烷(2mL)中,然后加入化合物7a(285mg,2.74mmol,4eq),然后升温至80℃反应2h,待原料反应完毕后,加入饱和碳酸钠水溶液淬灭,乙酸乙酯萃取,用盐水洗三次,合并有机相,无水硫酸钠干燥,旋干有机相,柱层析纯化(石油醚:乙酸乙酯=0:1),得到实施例6(50mg,白色固体,收率18.0%)。 1H NMR(400MHz,CDCl 3)δ8.27(d,J=5.6Hz,1H),8.18(s,1H),6.94(m,2H),6.88(m,1H),5.87(d,J=6.0Hz,1H),5.71(s,1H),5.36(m,1H),1.62(m,6H),1.44(s,3H)。
实施例7
合成路线:
Figure PCTCN2020089067-appb-000052
反应步骤:
(1)化合物2的合成:在100mL三口烧瓶中将6g化合物1和三乙胺(4.97g,1.2eq)溶于二氯甲烷中,0℃下缓慢加入乙酰氯(3.86g,1.2eq),TLC监测反应至完全。加水,乙酸乙酯萃取,无水硫酸钠干燥,旋干,柱层析得7g化合物2。GC-MS[M]为188。
(2)化合物3的合成:在100mL圆底烧瓶中加入化合物2(7g),三氯化铝(14.86g,3eq),升温至160℃,搅拌反应1h,TLC监测反应完全。加盐酸(6mol/L),乙酸乙酯萃取,无水硫酸钠干燥,旋干,柱层析,得6.16g化合物3。GC-MS[M]为188。
(3)化合物4的合成:在100mL圆底烧瓶中将化合物3(2g)和碳酸钾(7.3g,5eq)溶于丙酮中,搅拌下加入碘甲烷(7.5g,5eq),升温至60℃,TLC监测反应至完全。 乙酸乙酯萃取,无水硫酸钠干燥,旋干,柱层析分离,得2.05g化合物4。LC-MS[M+1]为203。
(4)化合物5的合成:在100mL圆底烧瓶中将化合物4(2.05g)和R-叔丁基亚磺酰胺(2.42g,2eq)溶于无水四氢呋喃中,搅拌下加入钛酸乙酯(4.56g,2eq),升温至60℃。TLC监测反应至完全,加水,抽滤,乙酸乙酯萃取,旋干,柱层析分离得2.63g化合物5。LC-MS[M+1]为306。
(5)化合物6的合成:在100mL三口烧瓶中将化合物5(2.63g)溶于无水四氢呋喃中,-50℃下加入硼氢化钠(0.98g,3eq),TLC监测反应至完全。氯化铵水溶液淬灭,加乙酸乙酯萃取,无水硫酸钠干燥有机相,柱层析分离得1.76g化合物6。LC-MS[M+1]为308。
(6)化合物7的合成:将化合物6(1.76g)加在100mL圆底烧瓶中,加入盐酸二氧六环(10mL),搅拌1h后,TLC显示反应结束。抽滤,得滤饼1.1g化合物7。
(7)化合物8的合成:在100mL圆底烧瓶中将化合物7(1.1g)和5-氯-3-氰基吡唑并[1,5-α]嘧啶(0.98g,1.2eq)溶解在无水乙醇中,搅拌下滴加三乙胺(1.8g,4eq),升温至60℃,TLC监测反应至完全。旋干溶剂,加入水和乙酸乙酯萃取,无水硫酸钠干燥有机相,柱层析分离,得1.35g化合物8。LC-MS[M+1]为346。
(8)化合物9的合成:在100mL圆底烧瓶中分别加入化合物8(1.35g)、盐酸羟胺(1g,4eq)、碳酸钾(2g,4eq)、乙醇(10mL)。升温至80℃,TLC监测反应至完全。旋干溶剂,加入水和乙酸乙酯萃取,无水硫酸钠干燥有机相,柱层析分离,得0.73g化合物9。LC-MS[M+1]为379。
实施例7的合成:在100mL圆底烧瓶中将化合物9(0.73g)溶于乙酸(4mL)和1,2-二氯乙烷(4mL)中,搅拌下加入2,2-二甲氧基丙烷(1g,5eq),升温至80℃,TLC监测反应至完全。加入碳酸钠水溶液和乙酸乙酯萃取,无水硫酸钠干燥有机相,柱层析分离,得0.36g实施例8。LC-MS[M+1]=419。 1H NMR(400MHz,CDCl 3)δ8.14(dd,J=7.7,1.9Hz,2H),7.07(dt,J=7.9,3.9Hz,1H),7.01(dd,J=7.4,2.6Hz,1H),6.33–6.21(m,2H),5.58(d,J=5.7Hz,1H),3.98(s,3H),1.72(s,3H),1.62–1.55(m,6H)。
实施例8
合成路线:
Figure PCTCN2020089067-appb-000053
反应步骤:
(1)化合物2的合成:将叔丁醇钠(0.07g,1.5eq)溶于5ml甲苯中,0℃加入化合物1(0.1g,1eq),5min后将化合物1’(5-氯吡唑并[1,5-a]嘧啶-3-羧酸乙酯)(0.13g,1.2eq)加 入反应体系,逐步升至rt反应2h。点板反应完毕,氯化铵溶液淬灭,EA萃取干燥,拌样过柱,得到0.1g化合物2,收率50%。
化合物实施例8的合成:将1,2-二氨基-2-甲基丙烷(1.5eq)溶于干燥的甲苯(3ml)中,Ar保护下,0℃滴加三甲基铝(5eq),滴毕,升至RT反应2h,再降温到0℃滴加化合物2(0.1g,1eq)的甲苯溶液(3ml),保温反应30min后升至80℃反应3h。TLC监测反应至完全,甲醇淬灭,调pH=8-9,EA萃取干燥,制备板分离得到20mg,收率20%。 1H NMR(400MHz,CDCl 3)δ9.54(s,1H),8.63(d,J=7.5Hz,1H),7.36–7.28(m,2H),7.13(dd,J=8.9,7.9Hz,1H),6.71(d,J=7.5Hz,1H),6.61(q,J=6.8Hz,1H),3.71(dd,J=27.5,10.7Hz,2H),1.86(d,J=6.9Hz,3H),1.58(d,J=4.4Hz,6H)。
实施例9
合成路线:
Figure PCTCN2020089067-appb-000054
反应步骤:
(1)化合物2的合成:将叔丁醇钠(0.35g,1.5eq)溶于25ml甲苯中,0℃加入化合物1(0.5g,1eq),5min后将化合物1’(0.51g,1.2eq)加入反应体系,逐步升至rt反应2h。TLC监测反应至完全,氯化铵溶液淬灭,EA萃取干燥,拌样过柱,得到0.7g,收率83%。
(2)化合物3的合成:将化合物2(0.7g,1eq)、盐酸羟胺(0.28g,2eq)和碳酸钾(0.56g,2eq)依次加入到无水乙醇(7ml)中,80℃反应过夜,反应完毕后,加水,EA萃取干燥拌样过柱得到0.3g,收率39.5%。LCMS(384.0,386.0)。
化合物实施例9的合成:将化合物3(0.1g,1eq)和2,2-二甲氧基丙烷(0.11g,4eq)加入到乙酸(4ml)中,50℃反应过夜,待反应完毕,碳酸氢钠溶液调碱,EA萃取,干燥,拌样过柱,得到0.07g,收率63.6%。 1H NMR(400MHz,CDCl 3)δ8.43(d,J=7.5Hz,1H),8.32(s,1H),7.31–7.27(m,1H),7.07(dd,J=8.8,8.0Hz,1H),6.59(q,J=6.9Hz,1H),6.49(d,J=7.5Hz,1H),5.61(s,1H),1.81(d,J=6.9Hz,3H),1.63(s,3H),1.55(s,3H)。
实施例10
合成路线:
Figure PCTCN2020089067-appb-000055
反应步骤:
(1)化合物2的合成:将化合物1(5g,1eq)溶于THF(50ml)中,加入R-叔丁基亚磺酰胺(7.25g,2eq),后将钛酸四乙酯(13.75g,2eq)加入反应体系,在60℃反应过夜。TLC监测反应至完全,拌样过柱(PE:EA=10:1—5:1),得到4.3g化合物2,收率53.7%。
(2)化合物3的合成:将化合物2(4.4g,1eq)溶于THF(35ml)中,-50℃下分批加入硼氢化钠(1.85g,3eq),后逐步升至RT反应5h,待反应完毕,后处理:加水,EA萃取,拌样过柱,得到2.6g+1g交叉(含其非对映异构体),收率59.1%。
(3)化合物4的合成:取化合物3(0.8g,1eq)加入8ml 4M的盐酸二氧六环中,RT反应4h,TLC监测反应至完全,加入碳酸钠溶液调pH=9-10,EA萃取,干燥旋干,得到0.5g化合物4,收率98%。
(4)化合物5的合成:取化合物4(0.5g,1eq)加入15ml无水乙醇,随后加入5-氯吡唑并[1,5-a]嘧啶-3-氰基(0.58g,1.1eq)和三乙胺(0.9g,3eq),在60℃反应过夜。TLC监测反应至完全,加入PE,过滤,得到0.4g,收率43.5%。
(5)化合物6的合成:将化合物5(0.4g,1eq),盐酸羟胺(0.18g,2eq)和碳酸钾(0.36g,2eq)依次加入到无水乙醇:二氧六环=2:1的混合溶剂(15ml)中,80℃反应过夜,反应完毕后,加水,EA萃取,干燥,拌样过柱,得到0.4g,收率91%。
化合物实施例10的合成:将化合物6(0.2g,1eq)和2,2-二甲氧基丙烷(0.25g,4eq)加入到乙酸:1,2-二氯乙烷=1:1的混合溶剂(6ml)中,80℃反应2h,待反应完毕,碳酸氢钠溶液调碱,EA萃取,干燥,拌样过柱,得到0.13g,收率59.1%。 1H NMR(400MHz,CDCl 3)δ8.19(d,J=7.8Hz,2H),6.92(m,J=13.3,8.8,3.7Hz,3H),6.09(d,J=7.4Hz,1H),5.87(s,1H),5.47(s,1H),5.29(s,1H),3.91(s,3H),1.62(s,3H),1.56(d,J=6.7Hz,6H)。
实施例11
合成路线:
Figure PCTCN2020089067-appb-000056
反应步骤:
(1)化合物2合成:向化合物1(1.0g,1.5eq)中加入10ml无水乙醇,随后分别加入INT-1(947mg,1.0eq)与TEA(1.6ml,3.0eq),置换氮气后于60℃反应18h,TLC监控反应完毕,旋干乙醇,然后向反应体系中加入水(50ml)后,加入EA(50mlX3)萃取,合并EA相,加入无水硫酸钠干燥,过滤,旋干过柱,得到1.1g化合物2(收率86%)。
(2)化合物3的合成:向150ml三颈烧瓶中加入50ml甲苯,降温至-10~0℃之间,然后向甲苯中鼓入氨气至饱合,之后于0℃下滴加三甲基铝(12.4ml,4.5eq),滴毕,升至室温搅拌2h,然后降温至0℃滴加化合物2(1.1g,1.0eq)的甲苯溶液,滴毕,升温至80℃反应18h。TLC监控反应完毕后,过滤,EA洗滤饼,收集滤液,向滤液中加水,分液,收集有机相,加入无水硫酸钠干燥,过滤,旋干过柱,得到600mg化合物3(收率51%)。
(3)化合物4的合成:向化合物3(600mg,1.0eq)中加入三氯氧磷(10ml),80℃搅拌5h,TLC监控反应完全,旋干三氯氧磷,然后用碳酸氢钠水溶液调pH至7~8,之后加入EA(40X3)萃取,分液,合并EA相,加入无水硫酸钠干燥,过滤,旋干过柱,得到120mg化合物4(收率21%)。
(4)化合物5的合成:向化合物4(80mg,1.0eq)中加入无水乙醇(3ml)与1,4-二氧六环(3ml),然后加入盐酸羟胺(42.6mg,2.0eq)与碳酸钾(85mg,2eq),置换氮气,80℃反应16h,TLC监控反应完全,过滤,旋干,直接过柱,得到70mg化合物5(收率79%)。
实施例11的合成:向化合物5(70mg,1.0eq)中加入1ml冰乙酸与1,2-二氯乙烷(1ml),之后加入2,2-二甲氧基丙烷(81mg,4eq),置换氮气,80℃反应1h。TLC监控反应完全,旋干溶剂,然后向体系中加入碳酸氢钠水溶液,调pH至7~8,然后加入EA(10mlX3)萃取,合并EA相加入无水硫酸钠干燥,过滤,旋干过柱,得到15mg(收率19%)。 1H NMR(400MHz,CDCl 3)δ7.91(d,J=7.4Hz,1H),7.02–6.82(m,3H),5.84(d,J=7.3Hz,1H),5.78(s,1H),5.24–5.13(m,1H),4.93(s,2H),3.90(s,3H),3.75(t,J=6.7Hz,1H),1.61(s,3H),1.53(d,J=6.7Hz,6H)。
实施例12
合成路线:
Figure PCTCN2020089067-appb-000057
反应步骤:
(1)化合物2的合成:向化合物1(15g,1.0eq)中加入BAST(23g,1.5eq),置换氮气,70℃下反应18h。TLC监控原料消失,向反应体系中加入水(100ml),然后加入***(100ml)萃取,收集***相,然后加入10%柠檬酸水溶液洗,分液,然后加入碳酸氢钠水溶液洗,分液,加入盐水洗一次,收集有机相干燥,低温旋干***,纯石油醚过柱,得到9.6g化合物2(收率58%)。
(2)化合物3的合成:向化合物2(5g)中加入无水THF(50ml),降温至-78℃,然后缓慢滴加n-BuLi(10.08ml,1.2eq),滴毕,保持低温搅拌1h,之后滴加INT-1(1.6g, 1.2eq)的THF溶液,滴毕,保温反应1h。TLC监控反应完毕,向反应体系中加入氯化铵水溶液淬灭,之后加入EA萃取,收集EA相,加入无水硫酸钠干燥,过滤,旋干过柱,得到680mg化合物3(收率16%)。
(3)化合物4的合成:将化合物3(680mg,1.0eq)溶于无水THF(8ml),然后加入R-叔丁基亚磺酰胺(814.6mg,2eq),之后加入钛酸四乙酯(1.56g,2eq),60℃反应2h。TLC监控反应完全,将反应液倾入水中,有固体析出,过滤,收集滤液,加水和EA萃取,收集EA相,加入无水硫酸钠干燥,旋干过柱,得到900mg化合物4(收率87%)。
(4)化合物5的合成:将化合物4(900mg,1.0eq)溶于THF(10ml)中,降温至-50℃后,分批加入硼氢化钠(224mg,2eq),加毕,逐渐升至室温反应2h,TLC监控反应完毕,向反应体系中加入水,然后加入EA(30mlX3)萃取,收集EA相,加入无水硫酸钠干燥,旋干过柱,得到115mg化合物5(收率12.7%)。
(5)化合物6的合成:向化合物5(115mg,1.0eq)中加入盐酸二氧六环(2ml),室温反应2h,TLC监控反应完全,旋干溶剂,加入饱和碳酸氢钠水溶液调pH至7~8,然后加入二氯甲烷与甲醇,萃取多次,收集有机相,加入无水硫酸钠干燥,过滤旋干得到80mg化合物6(收率95%)。
(6)化合物7的合成:向化合物6(80mg,1.0eq)中加入无水乙醇(10ml),随后分别加入INT-2(84mg,1.1eq)与TEA(0.17ml,3.0eq),置换氮气后于60℃反应18h,TLC监控反应完毕,旋干乙醇,然后向反应体系中加入水后,加入EA(10mlX3)萃取,合并EA相,加入无水硫酸钠干燥,过滤,旋干过柱,得到120mg化合物7(收率88%)。
(7)化合物8的合成:向化合物4(120mg,1.0eq)中加入无水乙醇(1.2ml)与1,4-二氧六环(0.4ml),然后加入盐酸羟胺(63.9mg,2.0eq)与碳酸钾(127.5mg,2.0eq),置换氮气,80℃反应16h,TLC监控反应完全,过滤,旋干,直接过柱,得到100mg化合物8(收率76%)。
实施例12的合成:向化合物8(100mg,1.0eq)中加入冰乙酸(1ml)与1,2-二氯乙烷(1ml),之后加入2,2-二甲氧基丙烷(112mg,4eq),置换氮气,80℃反应1h。TLC监控反应完全,旋干溶剂,然后向体系中加入碳酸氢钠水溶液,调pH至7~8,然后加入EA(20mlX3)萃取,合并EA相加入无水硫酸钠干燥,过滤,旋干过柱,得到40mg实施例14(收率36%)。 1H NMR(400MHz,CDCl 3)δ8.21(d,J=7.6Hz,1H),8.19(s,1H),7.50(d,J=7.0Hz,1H),7.38(s,1H),7.17–7.08(m,1H),6.13(d,J=7.6Hz,1H),5.96(s,1H),5.49(s,1H),5.43(s,1H),1.87(t,J=18.2Hz,3H),1.67–1.58(m,6H),1.53(s,3H)。
实施例13
合成路线:
Figure PCTCN2020089067-appb-000058
反应步骤:
(1)化合物2的合成:将化合物1(3g,1.0eq)溶于无水THF(10ml),然后加入R-叔丁基亚磺酰胺(4.17g,2.0eq),之后加入钛酸四乙酯(7.86g,2.0eq),60℃反应2h。TLC监控反应完全,将反应液倾入水中,有固体析出,过滤,收集滤液,加水和EA(150mgX3)萃取,收集EA相,加入无水硫酸钠干燥,旋干过柱,得到3.8g化合物2(收率97%)。
(2)化合物3的合成:将化合物2(3.8g,1.0eq)溶于THF(40ml)中,降温至-50℃后,分批加入硼氢化钠(1.04g,2.0eq),加毕,逐渐升至室温反应2h,TLC监控反应完毕,向反应体系中加入水,然后加入EA(100mlX3)萃取,收集EA相,加入无水硫酸钠干燥,旋干过柱,得到1.2g化合物3(收率31%)。
(3)化合物4的合成:向化合物3(1.2g,1.0eq)中加入盐酸二氧六环,室温反应2h,TLC监控反应完全,旋干溶剂,加入饱和碳酸氢钠水溶液调PH至7~8,然后加入二氯甲烷与甲醇,萃取多次,收集有机相,加入无水硫酸钠干燥,过滤,旋干,得到700mg化合物4(收率93%)。
(4)化合物5的合成:向化合物4(700mg,1.0eq)中加入10ml无水乙醇,随后分别加入INT-1(783mg,1.1eq)与TEA(1.2ml,3.0eq),置换氮气后于60℃反应18h,TLC监控反应完毕,旋干乙醇,然后向反应体系中加入水后,加入EA(50mlX3)萃取,合并EA相,加入无水硫酸钠干燥,过滤,旋干过柱,得到900mg化合物5(收率71%)。
(5)化合物6的合成:向化合物5(900mg,1.0eq)中加入无水乙醇(8ml)与1,4-二氧六环(4ml),然后加入盐酸羟胺(394.6mg,2.0eq)与碳酸钾(783.6mg,2.0eq),置换氮气,80℃反应16h,TLC监控反应完全,过滤,旋干直接过柱,得到730mg化合物6(收率74%)。
实施例13的合成:向化合物8(700mg,1.0eq)中加入冰乙酸(7ml)与1,2-二氯乙烷(7ml),之后加入2,2-二甲氧基丙烷(832mg,4.0eq),置换氮气,80℃反应1h。TLC监控反应完全,旋干溶剂,然后向体系中加入碳酸氢钠水溶液,调pH至7~8,然后加入EA(30mlX3)萃取,合并EA相加入无水硫酸钠干燥,过滤,旋干,过柱,得到42mg(收率5%)。 1H NMR(400MHz,CDCl 3)δ8.23(d,J=7.6Hz,1H),8.19(s,1H),6.85(dd,J=8.3,4.9 Hz,2H),6.17(d,J=7.6Hz,1H),5.85(s,1H),5.60(d,J=5.6Hz,1H),5.49–5.37(m,1H),1.60(d,J=7.0Hz,6H),1.51(s,3H)。
实施例14
合成路线:
Figure PCTCN2020089067-appb-000059
反应步骤:
(1)化合物2的合成:在250mL圆底烧瓶中将10g化合物1和MsCl(7.1g,1.3eq)溶于甲苯溶剂中,再加入三乙胺(7.3g,1.5eq)作碱,室温条件下反应4小时,TLC监测反应至完全。萃取,干燥,旋干有机相,用石油醚:乙酸乙酯(10:1)过柱,得13.1g淡黄色液态化合物2。
(2)化合物3的合成:将化合物2(13g)置于100mL圆底烧瓶中,加入DMF(60ml)作溶剂,随后加入NaN 3(5.9g,2.0eq),50℃条件下反应3.5小时,TLC监测反应至完全。加水和乙酸乙酯萃取,干燥有机相,旋干。用石油醚:乙酸乙酯(10:1)过柱,得10.2g化合物3。
(3)化合物4的合成:将化合物3(10.2g)置于250mL圆底烧瓶中,加入乙醇(102ml)和水(34ml)(3:1)作混合溶剂,再加入Zn(3.7g,1.3eq)和NH 4Cl(5.85g,2.5eq),80℃回流加热6小时,监测反应结束。过滤,乙酸乙酯萃取,干燥有机相,旋干。用石油醚:乙酸乙酯(10:1)过柱,得7.3g化合物4。
(4)化合物5的合成:取化合物4(1.5g)置于100ml圆底烧瓶中,随后加入化合物a(1.56g,1.2eq),加三乙胺(3ml,3eq)和乙醇(50ml)作溶剂,回流加热,2小时左右监测反应结束。旋去少量乙醇,加水和乙酸乙酯萃取。随后用石油醚:乙酸乙酯(3:1)过柱,得2.1g化合物5。
(5)化合物6的合成:取化合物5(1.0g)置于100ml圆底烧瓶中,再加入盐酸羟胺(1.12g,5.6eq)和无水碳酸钾(2.2g,5.6eq),随后加入乙醇(50ml)作溶剂,80℃回流加热过夜,监测反应结束。旋去少量乙醇,加水和乙酸乙酯萃取。随后用石油醚:乙酸乙酯(3:1)过柱,得0.6g化合物6。
实施例14的合成:称取化合物6(0.2g)置于50ml圆底烧瓶中,再加入2,2-二甲氧基丙烷(0.22g,4eq),量取1,2-二氯乙烷(4ml)和乙酸(4ml)为混合溶剂,80℃回流加热2小时,监测反应结束。加入少量水,再加饱和碳酸氢钠中和反应体系中的乙酸,随后 使用二氯甲烷萃取。随后用二氯甲烷:甲醇(30:1)过柱,得66mg最终化合物。 1H NMR(400MHz,DMSO)δ:8.59(d,J=6.0Hz,1H),8.55(d,J=7.6Hz,1H),7.97(s,1H),7.52(brs,1H),7.40(t,J=8.7Hz,1H),6.51(d,J=7.6Hz,1H),5.65-5.59(m,1H),1.59(d,J=7.2Hz,3H),1.48(s,3H),1.39(s,3H)。
实施例15
合成路线:
Figure PCTCN2020089067-appb-000060
反应步骤:
(1)化合物2的合成:在100mL圆底烧瓶中加入DMF(20mL)作反应溶剂,随后降温到0℃,缓慢加入NaH(1.71g,2.5eq,42.9mmol),加入完毕后降温30min左右,分批少量加入2-氯-5-氟烟酸(3g,17.1mmol),然后升温到室温反应,待反应4h后,加热至75℃过夜,可以得到化合物2,不需要进行处理,直接进行下一步。
(2)化合物3的合成:在化合物2的基础上,滴加碘乙烷(4.01g,1.5eq,25.7mmol),然后反应半个小时,停止反应。先用旋蒸旋去大量的DMF,然后用乙酸乙酯萃取,Na 2SO 4干燥,旋干溶剂,用石油醚:乙酸乙酯(20:1)过柱,提纯处理,得到1.3g化合物3。
(3)化合物4的合成:把化合物3(1.3g,4.87mmol)置于100mL三口瓶中,加入DCM(20mL)为反应溶剂,氮气保护,随后降温到-78℃,待稳定后,逐滴加入DIBAL-H(3.4mL,1.05eq,5.11mmol),保持温度在-78℃,1h左右,检测反应结束。加水和甲醇淬灭反应,产生难溶固体,加少量NaOH溶液,固体消失,使用DCM萃取反应,Na 2SO 4干燥,旋干溶剂,用石油醚:乙酸乙酯(20:1)过柱提纯处理,得到0.63g化合物4。
(4)化合物5的合成:将化合物4(0.63g,2.8mmol)置于100mL圆底烧瓶中,加入乙酸乙酯(10mL)作反应溶剂,随后加入IBX(1.88g,2.4eq,6.72mmol),80℃加热反应,2h左右反应结束,用砂芯漏斗抽滤,乙酸乙酯洗涤,收集滤液,旋干浓缩,得到0.35g化合物5。液质产生的质谱峰分子量比化合物多了18,等于结合了一个水,不影响下一步反应。
(5)化合物6的合成:把化合物5(0.35g,1.57mmol)加入100mL圆底烧瓶中,随后称取(R)-叔丁基亚磺酰胺(0.29g,1.5eq,2.35mmol)和碳酸铯(0.36g,0.7eq,1.1mmol)加入圆底烧瓶中,加入二氯甲烷(10mL)为反应溶剂,室温反应,2h左右反应结束,加二氯甲烷萃取,Na 2SO 4干燥,旋干溶剂,用石油醚:乙酸乙酯(10:1)过柱提纯处理,得到0.6g化合物6。
(6)化合物7的合成:把(0.6g,1.84mmol)化合物6置于100mL三口烧瓶中,加入无水THF(10mL)作反应溶剂,氮气保护,降温到-20℃,温度恒定后,缓慢滴加甲基溴化镁的四氢呋喃溶液(2.2mL,1.2eq,2.21mmol),随后升温反应,过夜反应后还有大量原料剩余,然后再补加甲基溴化镁的四氢呋喃溶液(2.2mL),温度恢复室温后,检测反应结束。加入饱和氯化铵水溶液淬灭反应。乙酸乙酯萃取,Na 2SO 4干燥,旋干溶剂,用石油醚:乙酸乙酯(1.5:1)过柱,提纯处理,得到0.2g化合物7。
(7)化合物8的合成:把化合物7(0.2g,0.3mmol)加入到50mL圆底烧瓶中,加入HCl/1,4-dioxane(3mL)和甲醇(3mL),室温反应1h左右,反应结束,NaHCO 3溶液中和反应,加入乙酸乙酯萃取,Na 2SO 4干燥,旋干溶剂,得到0.1g化合物8。
(8)化合物9的合成:取化合物8(0.1g,0.42mmol)置于50mL圆底烧瓶中,随后加入氯氰基化合物(90mg,1.2eq,0.51mmol),加三乙胺(0.13g,3eq)和乙醇(10mL)作溶剂,回流加热,2小时左右监测反应结束。旋去少量乙醇,加水和乙酸乙酯萃取。随后用石油醚:乙酸乙酯(3:1)过柱,得55mg化合物9。
(9)化合物10的合成:取化合物9(55mg,0.15mmol)置于50mL圆底烧瓶中,再加入盐酸羟胺(56mg,5.6eq)和无水碳酸钾(113mg,5.6eq),随后加入乙醇(5ml)作溶剂,80℃回流加热,监测反应结束。旋去少量乙醇,加水和乙酸乙酯萃取。随后用石油醚:乙酸乙酯(3:1)过柱,得41mg化合物10。
(10)实施例15的合成:将化合物10(41mg,0.1mmol)置于50ml圆底烧瓶中,再加入2,2-二甲氧基丙烷(61.95mg,6eq,0.6mmol),量取1,2-二氯乙烷(2mL)和乙酸(2mL)为混合溶剂,80℃回流加热2小时,监测反应结束。加入少量水,再加饱和碳酸氢钠中和反应体系中的乙酸,随后使用二氯甲烷萃取。随后用二氯甲烷:甲醇(30:1)过柱,得15mg最终化合物。
实施例16
合成路线:
Figure PCTCN2020089067-appb-000061
反应步骤:
(1)化合物2的合成:在250mL三口烧瓶中加入化合物1(5g,32.5mmol),加入乙腈和水(1:1)的混合溶液(100mL)作反应溶剂,氮气保护,随后降温到-78℃,在降温的过程中,-50℃时反应体系冻成一块,随后缓慢滴加溴氟甲基膦酸二乙酯(17.3g,2eq,65mmol),加入完毕后升至室温搅拌,反应4h左右,监测反应结束,加乙酸乙酯萃取, Na 2SO 4干燥,旋干溶剂,用石油醚:乙酸乙酯(19:1)过柱提纯处理,得到5.6g化合物2,产物无质谱吸收峰。
(2)化合物3的合成:把化合物2(3.04g,14.9mmol)加入100mL圆底烧瓶中,加入(R)-叔丁基亚磺酰胺(3.62g,2eq,29.8mmol)和钛酸四乙脂(8.5g,2.5eq,37.3mmol),取无水THF(20mL)为反应溶剂,80℃条件下反应,4h左右监测反应结束,加水之后反应体系产生大量固体,用硅藻土抽滤,洗涤,然后用乙酸乙酯萃取,Na 2SO 4干燥,旋干溶剂,用石油醚:乙酸乙酯(4:1)过柱,提纯处理,得到4.827g化合物3。
(3)化合物4的合成:把化合物3(4.827g,15.7mmol)加入100mL圆底烧瓶中,取无水甲醇(10mL)为反应溶剂,冰浴条件下缓慢加入硼氢化钠(1.487g,2.5eq,39.3mmol),加入完毕后升温搅拌30min,反应结束,缓慢加水和乙酸乙酯萃取,Na 2SO 4干燥,旋干溶剂,用石油醚:乙酸乙酯(1.5:1)过柱,提纯处理,得到1.82g化合物4。
(4)化合物5的合成:把化合物4(1.82g,0.3mmol)加入到100mL圆底烧瓶中,加入HCl/1,4-二氧六环(5mL)和甲醇(5mL),室温反应1h左右,反应结束,NaHCO 3溶液中和反应,加入乙酸乙酯萃取,Na 2SO 4干燥,旋干溶剂,得到1.4g化合物6。
(5)化合物6的合成:取化合物5(0.5g,2.44mmol)置于50mL圆底烧瓶中,随后加入氯氰基化合物(0.52g,1.2eq,2.93mmol),加三乙胺(0.65mL,2eq)和乙醇(10mL)作溶剂,回流加热,2小时左右监测反应结束。旋去少量乙醇,加水和乙酸乙酯萃取。随后用石油醚:乙酸乙酯(3:1)过柱,得0.53g化合物6。
(6)化合物7的合成:取化合物6(0.53g,1.53mmol)置于50mL圆底烧瓶中,再加入盐酸羟胺(0.59g,5.6eq)和无水碳酸钾(1.18g,5.6eq),随后加入乙醇(10ml)作溶剂,80℃回流加热,监测反应结束。旋去少量乙醇,加水和乙酸乙酯萃取。随后用二氯甲烷:甲醇(30:1)过柱,得420mg化合物7。
实施例16的合成:将化合物7(420mg,1.1mmol)置于50ml圆底烧瓶中,再加入2,2-二甲氧基丙烷(0.46g,4eq,4.44mmol),量取1,2-二氯乙烷(3mL)和乙酸(3mL)为混合溶剂,80℃回流加热,监测反应结束后。加入少量水,再加饱和碳酸氢钠中和反应体系中的乙酸,随后使用二氯甲烷萃取。随后用二氯甲烷:甲醇(30:1)过柱,得最终化合物26mg。
实施例1-16汇总如下表1-1:
表1-1
Figure PCTCN2020089067-appb-000062
Figure PCTCN2020089067-appb-000063
Figure PCTCN2020089067-appb-000064
Figure PCTCN2020089067-appb-000065
Figure PCTCN2020089067-appb-000066
同时,参照以上实施例,合成了实施例17-83,具体见表1-2:
表1-2
Figure PCTCN2020089067-appb-000067
Figure PCTCN2020089067-appb-000068
Figure PCTCN2020089067-appb-000069
Figure PCTCN2020089067-appb-000070
Figure PCTCN2020089067-appb-000071
Figure PCTCN2020089067-appb-000072
Figure PCTCN2020089067-appb-000073
Figure PCTCN2020089067-appb-000074
Figure PCTCN2020089067-appb-000075
实施例84实施例25及其对映异构体
Figure PCTCN2020089067-appb-000076
手性胺中间体的合成路线:
Figure PCTCN2020089067-appb-000077
参照实施例12中手性胺中间体化合物4的合成,实施例84的合成,以对氟苯乙酮和(R)-叔丁基亚磺酰胺为原料,经过生成亚胺,然后硼氢化钠还原,两个步,得到一对非对映异构体化合物3和化合物3’,通过柱层析将两个化合物分开,然后脱掉叔丁基亚磺酰基就得到R和S构型的两个手性胺中间体。两个手性胺中间体各自反应,就能得到实施例84为 R(即,实施例25)和S两个构型的化合物,两个手性胺中间体混合,得到实施例84,其为消旋体。
实施例85实施例48的拆分
Figure PCTCN2020089067-appb-000078
两个化合物通过制备液相分离,分离条件如下:
仪器:waters2525&waters2767;
色谱柱:Innoval ODS-2(30 X 100毫米,5微米);
流速:15.0毫升/分钟,检测波长254纳米;
溶剂:甲醇,样品浓度12毫克/毫升;
进样量:0.5毫升,延迟时间:24秒;
阈值:20000,时间表:2.00,
流动相:A:水(0.1%三氟乙酸),B;甲醇。
梯度程序:
t(min) A相 B相
0 41 59
22 41 59
25 10 90
27 10 90
28 41 59
30 41 59
测试例1:本发明化合物对ROS1、NTRK和ALK及它们的耐药激酶的抑制活性
化合物对蛋白激酶的活性抑制实验在Reaction Biology Corporation放射性标记的HotSpot激酶实验平台开展。制备含相应底物的新鲜反应液(20mM HEPESpH 7.5,10mM MgCl 2,1mM EGTA,0.02%Brij35,0.02mg/mL BSA,0.1mM Na 3VO 4,2mM DTT,1%DMSO),加入所需辅因子和待测激酶至上述溶液并轻轻混匀,使用Echo550移液***向每孔加入待测化合物DMSO溶液(空白对照组加入相应体积DMSO),加入33P-ATP(最终比活度0.01μCi/μL)以开始反应,反应液于室温孵育120分钟。将孵育后的反应液转移至P81离子交换层析纸(Whatman#3698-915)上,用0.75%的磷酸溶液洗脱,检测层析纸上剩余含放射性的磷酸化底物的量。
表2给出了本发明化合物对ROS1、NTRK和ALK及它们的耐药激酶的抑制活性IC 50值,其中,A<0.5nM,0.5nM≤B≤5.0nM,5.0nM<C<50nM,50nM≤D≤500nM,E>500nM;
表2
Figure PCTCN2020089067-appb-000079
Figure PCTCN2020089067-appb-000080
经激酶活性测试,表明本发明的系列化合物对ROS1、NTRK和ALK及它们的耐药突变有很好的抑制活性,尤其是对耐药突变的抑制更出色。
本发明化合物对ROS1、NTRK和ALK及它们的耐药突变中的一种或多种的活性均比目前在临床上的药物有更好的抑制剂活性。
本发明大部分化合物对ROS1、NTRK和ALK及它们的耐药突变的一种或多种活性优于或与目前在临床上的药物相当。
本发明化合物非常有潜力应用于由ROS1、NTRK和ALK等所介导的疾病的治疗。
测试例2:化合物对细胞的增殖抑制
化合物对细胞的增殖抑制实验在合肥中科普瑞昇生物医药科技有限公司开展。稳定转染不同激酶基因的Ba/F3工程细胞株复苏后采用RPMI 1640培养基(Biological Industries,Israel)+10%胎牛血清(Biological Industries,Israel)+1%双抗(Penicillin Streptomycin solution,Coring,USA)培养两代,取对数生长期细胞悬液,以2000细胞/孔,接种于96孔白色细胞培养板(Corning 3917,NY,USA),每孔体积为95μL。取5μL 20×待测化合物DMSO溶液加入上述含95μL细胞悬液的培养板中,空白对照组加入相应体积DMSO,混匀,于37℃、5%CO 2培养箱中孵育72小时,采用CellTiter-Glo检测细胞活力。
表3给出了本发明化合物对ROS1、NTRK和ALK或它们的耐药突变的Ba/F3工程细胞株的抑制活性IC 50值。
表3
Figure PCTCN2020089067-appb-000081
经细胞活性测试,发现本发明的系列化合物对ROS1、NTRK和ALK及它们的耐药突变的Ba/F3工程细胞株有很好的抑制活性,尤其是对耐药突变的抑制更出色。本发明的化合物对ROS1、NTRK和ALK及它们的耐药突变的Ba/F3工程细胞株活性均很好的抑制剂活性,且本发明大部分化合物对ROS1、NTRK和ALK及它们的耐药突变的Ba/F3工程细胞株活性也非常优异,非常有潜力应用于由ROS1、NTRK和ALK等所介导的疾病的治疗。
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (11)

  1. 式Ⅰ所示的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物:
    Figure PCTCN2020089067-appb-100001
    式Ⅰ中:
    A为
    Figure PCTCN2020089067-appb-100002
    其中,X独立地选自下组:NR 6、O、CR 1R 2、S、S(O)或S(O) 2
    B选自下组:单环芳烃、双环芳烃、单环杂芳烃或双环杂芳烃,其中,所述B的任意碳原子上的H均可以被以下取代基取代:卤素、羟基、氨基、氰基、酰基、酯基、烷基、环烷基、烷胺基、烷氧基、环烷氧基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代环烷氧基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代环烷氧基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立地选自下组:氘、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
    C独立地为
    Figure PCTCN2020089067-appb-100003
    其中,Y独立地选自下组:O、NR A或CR 1R 2
    Figure PCTCN2020089067-appb-100004
    代表Z式或E式;
    其中,R 1、R 2各自独立地选自下组:氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、烷氧基、环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立地选自下组:氘、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;或者R 1和R 2与其连接的C原子一起连接成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或硫杂 环烷烃;其中,所述取代是指被选自下组的一个或多个基团取代:烷基、酰基、酯基、磺酰基、亚磺酰基;
    R 3和R 4各自独立地选自下组:氢原子、氨基、羟基、酰基、酯基、烷基、环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立地选自下组:卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
    或者R 3和R 4与其连接的C原子一起连接成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃、硫杂环烷烃或氧代基(=O);或者R A和R 4与他们邻接的原子一起形成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或硫杂环烷烃;或者R 3与Y稠合形成取代或未取代的3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或硫杂环烷烃;其中,所述取代是指被选自下组的一个或多个基团取代:烷基、酰基、酯基、磺酰基、亚磺酰基;
    Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7各自独立地选自下组:N、CR 5或NR 6
    R 5独立地任意选自:氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、环烷基、烷胺基、烷氧基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
    R 6和R A各自独立地任意选自:氢原子、酰基、酯基、烷基、环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基。
  2. 如权利要求1的所述的式Ⅰ化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物:
    Figure PCTCN2020089067-appb-100005
    式Ⅰ中:
    A为
    Figure PCTCN2020089067-appb-100006
    其中,所述X为NR 6、O、CR 1R 2、S、S(O)或S(O) 2
    B任意选自单环芳烃、双环芳烃、单环杂芳烃或双环杂芳烃,其中所述B的任意碳原子上的H均可以被以下取代基取代:卤素、羟基、氨基、氰基、酯基、烷基、卤代烷基、烷胺基、烷氧基、芳基或杂芳基;
    C任意选自
    Figure PCTCN2020089067-appb-100007
    其中,所述R 1、R 2、R 3、R 4各自独立的任意选自氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基,R 1和R 2、R 2和R 3、R 3和R 4或者R 1和R 4可以连接成3-7元的环烷烃、氮杂环烷烃、氧杂环烷烃或硫杂环烷烃;
    Z 1、Z 2、Z 3、Z 4、Z 5、Z 6、Z 7各自独立的任意选自N、CR 5或NR 6
    R 5、R 6各自独立的任意选自氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、环烷基、卤代烷基、卤代环烷基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立的任意选自卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基。
  3. 如权利要求1所述的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物:
    Figure PCTCN2020089067-appb-100008
    式II中,
    *表示手性中心;
    X选自:NR 6、O、CR 1R 2、S、S(O)或S(O) 2
    R 1和R 2不同,且各自独立地选自下组:氢原子、卤素、氨基、氰基、羟基、烷基、卤代烷基;
    R 6独立地选自下组:氢原子、烷基、单取代或多取代烷基,所述单取代或多取代烷基的取代基独立地选自下组:卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
    B、C、Z 1、Z 2、Z 3、Z 4、Z 5、Z 6和Z 7的定义如权利要求1所述。
  4. 如权利要求1所述的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,其特征在于,B独立地选自下组:
    Figure PCTCN2020089067-appb-100009
    其中,
    Figure PCTCN2020089067-appb-100010
    为单键或双键;
    Z 8、Z 9各自独立地选自:CR 11或N;
    P独立地选自:O、NH、S;
    Figure PCTCN2020089067-appb-100011
    为双键时,Q独立地选自:CR 11或N;当
    Figure PCTCN2020089067-appb-100012
    为单键时,Q独立地选自:O、S、CR 11R 12或NH;
    各R 7各自独立地选自下组:氢原子、卤素、氨基、氰基、羟基、酰基、酯基、烷基、环烷基、烷氧基、烷氨基、芳基、杂芳基、单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基;所述单取代或多取代烷基、单取代或多取代烷氧基、单取代或多取代环烷基、单取代或多取代芳基、单取代或多取代杂芳基的取代基独立地选自下组:氘、卤素、氨基、氰基、羟基、酰基、酯基、烷基、卤代烷基、环烷基、卤代环烷基、烷氧基、卤代烷氧基、芳基和杂芳基;
    R 11和R 12各自独立地选自下组:H、羟基、卤素、氨基、氰基、酰基、烷基、卤代烷基、烷氧基、卤代烷氧基;
    e为0、1、2、3或4。
  5. 如权利要求1所述的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合 物,或其可药用的盐,或其氘代物,其特征在于,C为
    Figure PCTCN2020089067-appb-100013
    其中,R 3和R 4的定义如权利要求1所述。
  6. 如权利要求1所述的化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,其特征在于:式Ⅰ所示的化合物任意选自如下化合物:
    Figure PCTCN2020089067-appb-100014
    Figure PCTCN2020089067-appb-100015
  7. 如权利要求1所述的式Ⅰ化合物的可药用的盐,其特征在于:所述可药用的盐为无机酸盐或有机酸盐,所述无机酸盐选自盐酸盐、氢溴酸盐、氢碘酸盐、硫酸盐、硫酸氢盐、硝酸盐、磷酸盐、酸式磷酸盐;所述有机酸盐选自甲酸盐、乙酸盐、三氟乙酸盐、丙酸盐、丙 酮酸盐、羟乙酸盐、乙二酸盐、丙二酸盐、富马酸盐、马来酸盐、乳酸盐、苹果酸盐、柠檬酸盐、酒石酸盐、甲磺酸盐、乙磺酸盐、羟乙磺酸盐、苯磺酸盐、水杨酸盐、苦味酸盐、谷氨酸盐、抗坏血酸盐、樟脑酸盐、樟脑磺酸盐。
  8. 一种药物组合物,其含有治疗有效量的权利要求1所述的式Ⅰ化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,以及一种或多种药学上可以接受的载体、稀释剂或赋形剂。
  9. 如权利要求1所述的式Ⅰ化合物,或其互变异构体,或其内消旋体、外消旋体及内消旋体和外消旋体的混合物,或其对映异构体、非对映异构体及对映异构体和非对映异构体的混合物,或其可药用的盐,或其氘代物,或含有式Ⅰ所示的化合物的组合物在制备用于预防和/或治疗ROS1、NTRK、ALK等介导的病理学特征的疾病的药物方面的用途。
  10. 如权利要求9所述的用途,其特征在于,所述ROS1、NTRK、ALK等介导的病理学特征的疾病包括癌症、肉瘤和疼痛。
  11. 如权利要求10所述的用途,其特征在于,所述的癌症是乳腺癌、***、结肠癌、肺癌、胃癌、直肠癌、胰腺癌、脑癌、皮肤癌、口腔癌、***癌、骨癌、肾癌、卵巢癌、膀胱癌、肝癌、输卵管肿瘤、腹膜肿瘤、黑色素瘤、神经胶质瘤、神经胶母细胞瘤、头颈癌、乳突肾性瘤、白血病、淋巴瘤、骨髓瘤、甲状腺瘤中的任一种。
PCT/CN2020/089067 2019-05-08 2020-05-07 用作激酶抑制剂的化合物及其应用 WO2020224626A1 (zh)

Priority Applications (13)

Application Number Priority Date Filing Date Title
BR112021022255A BR112021022255A2 (pt) 2019-05-08 2020-05-07 Composto usado como inibidor de quinase e aplicação do mesmo
AU2020270303A AU2020270303B2 (en) 2019-05-08 2020-05-07 Compound used as kinase inhibitor and application thereof
CN202080004473.2A CN112867717B (zh) 2019-05-08 2020-05-07 用作激酶抑制剂的化合物及其应用
CA3142088A CA3142088C (en) 2019-05-08 2020-05-07 Compound used as kinase inhibitor and application thereof
JP2021566283A JP7420403B2 (ja) 2019-05-08 2020-05-07 キナーゼ阻害剤として使用される化合物およびその応用
KR1020217040056A KR20220007111A (ko) 2019-05-08 2020-05-07 키나아제 억제제로 사용되는 화합물 및 이의 응용
MX2021013576A MX2021013576A (es) 2019-05-08 2020-05-07 Compuesto utilizado como inhibidor de la quinasa y aplicación del mismo.
EP20802185.7A EP3967696A4 (en) 2019-05-08 2020-05-07 COMPOUND USED AS A KINASE INHIBITOR AND ITS APPLICATION
SG11202112381VA SG11202112381VA (en) 2019-05-08 2020-05-07 Compound used as kinase inhibitor and application thereof
TW110106655A TW202142541A (zh) 2020-05-07 2021-02-25 用作激酶抑制劑的化合物及其應用
US17/521,153 US20220089603A1 (en) 2019-05-08 2021-11-08 Compound used as kinase inhibitor and application thereof
IL287908A IL287908A (en) 2019-05-08 2021-11-08 A compound used as a kinase inhibitor and its use
ZA2021/09737A ZA202109737B (en) 2019-05-08 2021-11-29 Compound used as kinase inhibitor and application thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910379104.0 2019-05-08
CN201910379104 2019-05-08

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/521,153 Continuation-In-Part US20220089603A1 (en) 2019-05-08 2021-11-08 Compound used as kinase inhibitor and application thereof

Publications (2)

Publication Number Publication Date
WO2020224626A1 true WO2020224626A1 (zh) 2020-11-12
WO2020224626A9 WO2020224626A9 (zh) 2020-12-30

Family

ID=73051434

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/089067 WO2020224626A1 (zh) 2019-05-08 2020-05-07 用作激酶抑制剂的化合物及其应用

Country Status (13)

Country Link
US (1) US20220089603A1 (zh)
EP (1) EP3967696A4 (zh)
JP (1) JP7420403B2 (zh)
KR (1) KR20220007111A (zh)
CN (1) CN112867717B (zh)
AU (1) AU2020270303B2 (zh)
BR (1) BR112021022255A2 (zh)
CA (1) CA3142088C (zh)
IL (1) IL287908A (zh)
MX (1) MX2021013576A (zh)
SG (1) SG11202112381VA (zh)
WO (1) WO2020224626A1 (zh)
ZA (1) ZA202109737B (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022095910A1 (zh) * 2020-11-04 2022-05-12 浙江同源康医药股份有限公司 用作激酶抑制剂的化合物及其应用
CN114957259A (zh) * 2021-02-25 2022-08-30 南京明德新药研发有限公司 氰基取代的芳香双环类化合物及其应用
WO2022262671A1 (zh) * 2021-06-15 2022-12-22 中国医药研究开发中心有限公司 杂环大环化合物及其医药用途

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102596957A (zh) * 2009-07-09 2012-07-18 阵列生物制药公司 作为TRK激酶抑制剂的被取代的吡唑并[1,5-a]嘧啶化合物
CN104619708A (zh) * 2012-07-10 2015-05-13 武田药品工业株式会社 氮杂吲哚衍生物
CN107207514A (zh) * 2014-12-15 2017-09-26 康联制药有限公司 稠环杂芳基化合物及其作为trk抑制剂的用途
CN111171019A (zh) * 2018-11-13 2020-05-19 上海轶诺药业有限公司 一类五元并六元杂环化合物及其作为蛋白受体激酶抑制剂的用途
WO2020114499A1 (en) * 2018-12-07 2020-06-11 Betta Pharmaceuticals Co., Ltd Tyrosine kinase inhibitors, compositions and methods there of

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102005042742A1 (de) * 2005-09-02 2007-03-08 Schering Ag Substituierte Imidazo[1,2b]pyridazine als Kinase-Inhibitoren, deren Herstellung und Verwendung als Arzneimittel
UY33597A (es) 2010-09-09 2012-04-30 Irm Llc Compuestos y composiciones como inhibidores de la trk
TWI585088B (zh) 2012-06-04 2017-06-01 第一三共股份有限公司 作爲激酶抑制劑之咪唑并[1,2-b]嗒衍生物
KR20150011838A (ko) 2012-06-20 2015-02-02 에프. 호프만-라 로슈 아게 탄키라아제 억제제로서의 피롤로피라존

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102596957A (zh) * 2009-07-09 2012-07-18 阵列生物制药公司 作为TRK激酶抑制剂的被取代的吡唑并[1,5-a]嘧啶化合物
CN104619708A (zh) * 2012-07-10 2015-05-13 武田药品工业株式会社 氮杂吲哚衍生物
CN107207514A (zh) * 2014-12-15 2017-09-26 康联制药有限公司 稠环杂芳基化合物及其作为trk抑制剂的用途
CN111171019A (zh) * 2018-11-13 2020-05-19 上海轶诺药业有限公司 一类五元并六元杂环化合物及其作为蛋白受体激酶抑制剂的用途
WO2020114499A1 (en) * 2018-12-07 2020-06-11 Betta Pharmaceuticals Co., Ltd Tyrosine kinase inhibitors, compositions and methods there of

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
GRECO, A, CELL. BIOL., vol. 15, 1995, pages 6118
KATAYAMA, RYOHEI ET AL.: "The new-generation selective ROS1/NTRK inhibitor DS-6051b overcomes crizotinib resistant ROS1-G2032R mutation in preclinical models", NATURE COMMUNICATIONS, vol. 10, no. 1,, 9 August 2019 (2019-08-09), XP055751643 *
RUSSO, M: "6", CANCER DISCOVERY, 2016, pages 36
THOMAS SORRELL: "Handbook of Chemistry and Physics", 1999, UNIVERSITY SCIENCE BOOKS

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022095910A1 (zh) * 2020-11-04 2022-05-12 浙江同源康医药股份有限公司 用作激酶抑制剂的化合物及其应用
CN114957259A (zh) * 2021-02-25 2022-08-30 南京明德新药研发有限公司 氰基取代的芳香双环类化合物及其应用
WO2022262671A1 (zh) * 2021-06-15 2022-12-22 中国医药研究开发中心有限公司 杂环大环化合物及其医药用途
CN115884776A (zh) * 2021-06-15 2023-03-31 中国医药研究开发中心有限公司 杂环大环化合物及其医药用途

Also Published As

Publication number Publication date
AU2020270303A1 (en) 2022-01-06
CN112867717A (zh) 2021-05-28
JP7420403B2 (ja) 2024-01-23
EP3967696A1 (en) 2022-03-16
BR112021022255A2 (pt) 2022-01-18
KR20220007111A (ko) 2022-01-18
WO2020224626A9 (zh) 2020-12-30
ZA202109737B (en) 2022-07-27
SG11202112381VA (en) 2021-12-30
IL287908A (en) 2022-01-01
EP3967696A4 (en) 2023-08-09
CA3142088A1 (en) 2020-11-12
CN112867717B (zh) 2023-01-10
MX2021013576A (es) 2021-12-15
CA3142088C (en) 2023-02-28
US20220089603A1 (en) 2022-03-24
AU2020270303B2 (en) 2023-09-07
JP2022532141A (ja) 2022-07-13

Similar Documents

Publication Publication Date Title
AU2017335242C1 (en) Pyridine compound
WO2020259432A1 (zh) Kras-g12c抑制剂
WO2020224626A1 (zh) 用作激酶抑制剂的化合物及其应用
CN111171049B (zh) 酪氨酸激酶抑制剂及其用途
JP2023535746A (ja) Cdk7キナーゼ阻害剤として使用される化合物およびその適用
WO2017152707A1 (zh) 吡啶胺基嘧啶衍生物甲磺酸盐的结晶形式及其制备和应用
WO2021213317A1 (zh) Hpk1抑制剂及其制备方法和用途
WO2021249563A1 (zh) 芳基或杂芳基并吡啶酮或嘧啶酮类衍生物及其制备方法和应用
CN116390728B (zh) 喹唑啉衍生物及其制备方法和用途
WO2023078451A1 (zh) 用作cdk7激酶抑制剂的化合物及其应用
CN109988151B (zh) 一种炔类化合物、制备方法及其应用
CN116134016A (zh) 三环杂环
WO2022095910A1 (zh) 用作激酶抑制剂的化合物及其应用
CA3090876C (en) Dioxinoquinoline compounds, preparation method and uses thereof
JP2009073743A (ja) 新規な縮合環式ピリミジン化合物又はその塩、及びその医薬組成物
WO2021110010A1 (zh) 新型吡咯化合物
WO2017049711A1 (zh) 喹啉类衍生物、其药物组合物、制备方法及应用
WO2023025164A1 (zh) 芳基磷氧化合物的晶型、制备方法及用途
EA045833B1 (ru) Соединение, используемое как ингибитор киназы, и его применение
KR20240021239A (ko) Cdk 키나아제 억제제로 사용되는 화합물 및 이의 용도
WO2024027695A1 (zh) 作为her2抑制剂的化合物
CN115322158A (zh) 作为krasg12c蛋白抑制剂的取代喹唑啉类化合物
CN114805371A (zh) 含2-氨基嘧啶大环类化合物及其制备方法和用途
CN113354630A (zh) 一种5,6-二氢苯并[h]喹唑啉类化合物及其应用
CN116462680A (zh) 一种抗肿瘤化合物及其应用

Legal Events

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

Ref document number: 20802185

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021566283

Country of ref document: JP

Kind code of ref document: A

Ref document number: 3142088

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021022255

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20217040056

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020802185

Country of ref document: EP

Effective date: 20211208

ENP Entry into the national phase

Ref document number: 2020270303

Country of ref document: AU

Date of ref document: 20200507

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112021022255

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20211105