WO2018137639A1 - 一种组蛋白甲基转移酶ezh2抑制剂、其制备方法及其医药用途 - Google Patents
一种组蛋白甲基转移酶ezh2抑制剂、其制备方法及其医药用途 Download PDFInfo
- Publication number
- WO2018137639A1 WO2018137639A1 PCT/CN2018/073946 CN2018073946W WO2018137639A1 WO 2018137639 A1 WO2018137639 A1 WO 2018137639A1 CN 2018073946 W CN2018073946 W CN 2018073946W WO 2018137639 A1 WO2018137639 A1 WO 2018137639A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- group
- alkyl
- optionally further
- groups
- hydrogen
- Prior art date
Links
- 0 CCN(C(CC1)CN1C(OC(C)(C)C)=O)c1cc(O)cc(*)c1C Chemical compound CCN(C(CC1)CN1C(OC(C)(C)C)=O)c1cc(O)cc(*)c1C 0.000 description 5
- JXWVLXPKVHFWFR-UHFFFAOYSA-N CC(C=C(C)N1)=C(CNC(c2c(C)c(N(CC3)CCN3C(C=C)=O)cc(Br)c2)=O)C1=O Chemical compound CC(C=C(C)N1)=C(CNC(c2c(C)c(N(CC3)CCN3C(C=C)=O)cc(Br)c2)=O)C1=O JXWVLXPKVHFWFR-UHFFFAOYSA-N 0.000 description 1
- BLPRODXOHQLOPB-MXAZLMSGSA-N CC12[C@H](CC3)CC3C[C@H]1C2 Chemical compound CC12[C@H](CC3)CC3C[C@H]1C2 BLPRODXOHQLOPB-MXAZLMSGSA-N 0.000 description 1
- KETURVDVFLNBIZ-UHFFFAOYSA-N CCN(C(CC1)CCN1C(C)=O)c1cc(Br)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C Chemical compound CCN(C(CC1)CCN1C(C)=O)c1cc(Br)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C KETURVDVFLNBIZ-UHFFFAOYSA-N 0.000 description 1
- YTXRGRPMWYUUNL-UHFFFAOYSA-N CCN(C(CC1)CCN1C(C=C)=O)c1c(C)c(C(NCC2=C(C)C=C(C)NC2=O)=O)cc(-c2cccc(CN3CCN(C)CCC3)c2)c1 Chemical compound CCN(C(CC1)CCN1C(C=C)=O)c1c(C)c(C(NCC2=C(C)C=C(C)NC2=O)=O)cc(-c2cccc(CN3CCN(C)CCC3)c2)c1 YTXRGRPMWYUUNL-UHFFFAOYSA-N 0.000 description 1
- UMUBVKSOGHPUHM-UHFFFAOYSA-N CCN(C(CC1)CCN1C(C=C)=O)c1cc(-c2ccc(CN(CC3)CCC3N(C)C)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C Chemical compound CCN(C(CC1)CCN1C(C=C)=O)c1cc(-c2ccc(CN(CC3)CCC3N(C)C)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C UMUBVKSOGHPUHM-UHFFFAOYSA-N 0.000 description 1
- XEPGEMYDUVQFDT-UHFFFAOYSA-N CCN(C(CC1)CCN1C(C=C)=O)c1cc(-c2ccc(CN(CC3)CCC3OCC)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C Chemical compound CCN(C(CC1)CCN1C(C=C)=O)c1cc(-c2ccc(CN(CC3)CCC3OCC)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C XEPGEMYDUVQFDT-UHFFFAOYSA-N 0.000 description 1
- MDJWSRCNTQIUIT-UHFFFAOYSA-N CCN(C(CC1)CN1C(C=C)=O)c1c(C)c(C(NCC2=C(C)C=C(C)NC2=O)=O)cc(-c(cc2)cc(Cl)c2C(N2CCN(C)CC2)=O)c1 Chemical compound CCN(C(CC1)CN1C(C=C)=O)c1c(C)c(C(NCC2=C(C)C=C(C)NC2=O)=O)cc(-c(cc2)cc(Cl)c2C(N2CCN(C)CC2)=O)c1 MDJWSRCNTQIUIT-UHFFFAOYSA-N 0.000 description 1
- DAVGMROJYVKLBW-UHFFFAOYSA-N CCN(C(CC1)CN1C(C=C)=O)c1cc(-c2cc(C(N3CCN(C)CC3)=O)ccc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C Chemical compound CCN(C(CC1)CN1C(C=C)=O)c1cc(-c2cc(C(N3CCN(C)CC3)=O)ccc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C DAVGMROJYVKLBW-UHFFFAOYSA-N 0.000 description 1
- PKMGEACWJZGQNR-UHFFFAOYSA-N CCN(C(CC1)CN1C(C=C)=O)c1cc(-c2ccc(C)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C Chemical compound CCN(C(CC1)CN1C(C=C)=O)c1cc(-c2ccc(C)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C PKMGEACWJZGQNR-UHFFFAOYSA-N 0.000 description 1
- AOUHOSDWLKEZMJ-UHFFFAOYSA-N CCN(C(CC1)CN1C(C=C)=O)c1cc(-c2ccc(CN3CCCC3)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C Chemical compound CCN(C(CC1)CN1C(C=C)=O)c1cc(-c2ccc(CN3CCCC3)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C AOUHOSDWLKEZMJ-UHFFFAOYSA-N 0.000 description 1
- WVWXXHPZFWEAQL-UHFFFAOYSA-N CCN(C(CC1)CN1C(C=C)=O)c1cc(-c2ccc(CN3CCCCC3)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C Chemical compound CCN(C(CC1)CN1C(C=C)=O)c1cc(-c2ccc(CN3CCCCC3)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C WVWXXHPZFWEAQL-UHFFFAOYSA-N 0.000 description 1
- ALOSGIRNGYSXSH-UHFFFAOYSA-N CCN(C(CC1)CN1C(C=C)=O)c1cc(-c2ccc(CO)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C Chemical compound CCN(C(CC1)CN1C(C=C)=O)c1cc(-c2ccc(CO)cc2)cc(C(NCC2=C(C)C=C(C)NC2=O)=O)c1C ALOSGIRNGYSXSH-UHFFFAOYSA-N 0.000 description 1
- NZTCEWZBMOTKCJ-UHFFFAOYSA-N CN(CC1)CCN1c(cc1)ncc1N Chemical compound CN(CC1)CCN1c(cc1)ncc1N NZTCEWZBMOTKCJ-UHFFFAOYSA-N 0.000 description 1
- DZIASOMGGPNYCU-UHFFFAOYSA-N CN(CC1)CCN1c(nc1)ccc1[N+]([O-])=O Chemical compound CN(CC1)CCN1c(nc1)ccc1[N+]([O-])=O DZIASOMGGPNYCU-UHFFFAOYSA-N 0.000 description 1
- PVOAHINGSUIXLS-UHFFFAOYSA-N CN1CCNCC1 Chemical compound CN1CCNCC1 PVOAHINGSUIXLS-UHFFFAOYSA-N 0.000 description 1
- CMZCBPLTJBYJHZ-UHFFFAOYSA-N Cc(c([N+]([O-])=O)cc(F)c1)c1C(O)=O Chemical compound Cc(c([N+]([O-])=O)cc(F)c1)c1C(O)=O CMZCBPLTJBYJHZ-UHFFFAOYSA-N 0.000 description 1
- JVBLXLBINTYFPR-UHFFFAOYSA-N Cc(ccc(F)c1)c1C(O)=O Chemical compound Cc(ccc(F)c1)c1C(O)=O JVBLXLBINTYFPR-UHFFFAOYSA-N 0.000 description 1
- FTAGZJAFWFNHAP-UHFFFAOYSA-N O=C(c(cc1)ccc1Br)N1CCOCC1 Chemical compound O=C(c(cc1)ccc1Br)N1CCOCC1 FTAGZJAFWFNHAP-UHFFFAOYSA-N 0.000 description 1
- BAZVFQBTJPBRTJ-UHFFFAOYSA-N [O-][N+](c(cc1)cnc1Cl)=O Chemical compound [O-][N+](c(cc1)cnc1Cl)=O BAZVFQBTJPBRTJ-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/14—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/44—Non condensed pyridines; Hydrogenated derivatives thereof
- A61K31/4412—Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D213/00—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
- C07D213/02—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
- C07D213/04—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
- C07D213/60—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
- C07D213/62—Oxygen or sulfur atoms
- C07D213/63—One oxygen atom
- C07D213/64—One oxygen atom attached in position 2 or 6
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
- C07D401/12—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D405/00—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
- C07D405/02—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
- C07D405/12—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D409/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
- C07D409/02—Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
- C07D409/12—Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D413/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
- C07D413/02—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
- C07D413/12—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D413/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
- C07D413/14—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D417/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
- C07D417/02—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
- C07D417/12—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D491/00—Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
- C07D491/02—Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
- C07D491/10—Spiro-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D491/00—Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
- C07D491/02—Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
- C07D491/10—Spiro-condensed systems
- C07D491/107—Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
Definitions
- the present invention relates to a novel histone methyltransferase EZH2 inhibitor, a process for the preparation thereof, and a pharmaceutical composition containing the same, and its use as a histone methyltransferase EZH2 inhibitor, in particular in prevention and/or prevention The use in the treatment of human diseases including cancer.
- Tumors including leukemia, are one of the major diseases that cause clinical death in humans.
- Malignant tumors such as lung cancer, stomach cancer, breast cancer, pancreatic cancer, liver cancer, colon cancer, ovarian cancer, cervical cancer, esophageal cancer, nasopharyngeal carcinoma, leukemia and malignant lymphoma have extremely high mortality rates.
- genetic screening, molecular diagnosis, and precision medicine for cancer have improved clinical early detection/correct diagnosis and treatment of cancer patients, most cancers, especially advanced, refractory, relapsed, and drug-resistant malignancies, have not been effective so far.
- the method or drug can be completely eradicated or cured.
- the occurrence, development, metastasis, deterioration, recurrence and drug resistance of cancer are related to many factors.
- the abnormality of molecular signaling pathway in normal cells is one of the important factors leading to cell transformation and canceration. Genetics (changes in gene expression levels due to genetic sequence changes, such as gene mutations, loss of gene heterozygosity, and microsatellite instability) and epigenetics (changes in gene expression levels due to non-gene sequence changes, such as DNA methylation And chromatin conformational changes, etc.) are two important methodologies for studying molecular signaling. Changes in gene sequences often directly lead to different types of human diseases including cancer. After decades of efforts, with the next generation of gene sequencing technology and genomic big data, many genetic variant genes related to human diseases have been discovered and identified as biomarker targets for clinical diagnosis and drug development, while many These targeted drugs show good results in the treatment of clinical diseases.
- Epigenetic research is a branch of genetics that does not involve changes in gene or protein expression of DNA sequence changes and can be stably transmitted during development and cell proliferation, including DNA methylation, histone covalent modification, and staining. Regulatory mechanisms such as remodeling, gene silencing and RNA editing.
- DNA is packaged with histones to form chromatin, and changes in the chromatin ordered structure can result in changes in the transcription of related genes. This process is highly precisely controlled because changes in gene expression patterns affect the normal physiological functions of cells such as differentiation, proliferation and apoptosis.
- the covalent modification of the N-terminal tail of histones mediates changes in chromatin structure, resulting in heritable changes in gene expression, but does not affect the sequence of the DNA itself.
- Enzymatically mediated covalent modification of amino acid side chains is one of the important functions of gene expression regulation.
- Histone methyltransferase controls the selective addition of methyl groups to specific amino acid sites on histones.
- the level of expression of a particular gene is affected by the presence or absence of one or more methyl groups at the relevant histone site.
- the specific effect of the methyl group at a particular histone site persists until the methyl group is removed by histone demethylase, or until the modified histone is replaced by nucleosome turnover for control of gene expression.
- HDAC Histone Deacetylase
- PRC2 Polycomb Repressive Complex 2 is one of two types of polycomb proteins or (PcG). This complex has histone methyltransferase activity, mainly histone H3 trimethylation on lysine 27 (ie H3K27me3), which serves as a marker for transcriptional silencing of chromatin.
- PRC2 has four subunits: Suz12 (zinc finger), Eed, Ezh1 or Ezh2 (the SET domain with histone methyltransferase activity) and RbAp48 (histone binding domain), of which EZH2 (Zeste Homolog 2 enhancement) Sub) belongs to the catalytic subunit.
- the catalytic site of EZH2 is present in the SET domain, except for the H3-K79 methyltransferase DOT1, which is characteristic of all known histone lysine methyltransferases.
- DOT1 H3-K79 methyltransferase
- the mutant EZH2 gene is heterozygous and all mutants can be incorporated into the polyprotein PRC2 complex, but the complex lacks the ability to catalyze H3-K27 methylation.
- Follicular lymphoma is one of the most common non-Hodgkin's lymphomas and is sensitive to immunochemotherapy, but many patients are highly recurrent, and clinical recurrence eventually leads to non-response to standard treatment, about 30%.
- the patient turned into an aggressive lymphoma, a diffuse large B-cell lymphoma (DLBCL).
- DLBCL diffuse large B-cell lymphoma
- epigenome mutations primarily target histone methyltransferases KMT2D (90%) and EZH2 (25%), as well as histone acetyltransferases CREBBP (30-60%) and EP300 (9%). ).
- anti-CD20 monoclonal antibody (rituximab) treatment has significantly improved disease in patients, DLBCL remains an incurable cancer, and patients with chemically tolerant or transformed diseases still do not have effective clinical treatment.
- EZH2 inhibitors such as EPZ-6438 can effectively inhibit the growth of non-Hodgkin's lymphoma cells with EZH2 mutation in vitro and in vivo, and have conducted clinical trials (Knutson SK et al.
- the present inventors have unexpectedly discovered EZH2 amino acids in the research and development of long-term epigenetic histone modification mutant enzyme inhibitors according to the EZH2 protein sequence, crystal structure and known EZH2 inhibitor binding sites and modes of action (reversible).
- EZH2 amino acids There is a unique cysteine Cys668 (NP_004447, hisine-lysine N-methyltransferase EZH2isoform a) in the SET sequence, and the corresponding site of EZH1 is Ser664 (NP_001982, hisine-lysine N-methyltransferase EZH1isoform 1).
- the compound of the present invention can form an irreversible covalent bond with EZH2, and effectively inhibits the growth of cancer cells expressed by EZH2 activating mutant genes (such as EZH2A677G and Y641N) and induces apoptosis. Therefore, the compounds of the present invention provide a new mechanism of action and direction for the development of EZH2 inhibitors.
- the object of the present invention is to provide a small molecule compound with good specificity, high activity and low toxicity, which can be used as a highly selective irreversible covalent binding inhibitor of histone methyltransferase EZH2 for preventing and/or treating human beings.
- Diseases include use in cancer.
- the invention provides a compound of formula (I), or a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof,
- R is selected from
- Ar is selected from a 5- or 6-membered aryl or heteroaryl group, and a fused ring formed from a 5- or 6-membered aryl or heteroaryl group, wherein Ar is optionally further substituted with one or more -Q-T groups;
- L is selected from a C 2 -C 8 saturated or unsaturated linear or branched hydrocarbon chain or cyclic structure, and any hydrogen atom in L may be replaced by a halogen, a cyano group, a hydroxyl group, a C 1 -C 6 alkoxy group, L Any of the carbon atoms may be replaced by N, O, S; L is optionally further substituted with one or more -QT groups;
- R 1 and R 2 are each independently selected from hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered hetero a cycloalkyl group, said alkyl, alkenyl, alkynyl, cycloalkyl and heterocyclyl optionally being further substituted by one or more -QT groups;
- R 1 may be attached to any position of L, together with the N atom to which they are attached, form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further one or more -QT group substitution;
- R 2 may be attached to any position of L, together with the N atom to which they are attached, to form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further one or more -QT group substitution;
- R 1 , R 2 , L and the two N atoms to which they are attached form a 4-12 membered heterocyclic group, which is optionally further substituted with one or more -QT groups;
- R 3 is selected from the group consisting of hydrogen, fluorine, and R a ;
- R 4 and R 5 are each independently selected from the group consisting of hydrogen and R a ;
- R 5 ' is selected from halogen, -OS(O) 2 -C 1 -C 6 alkyl or -OS(O) 2 -C 3 -C 6 cycloalkyl;
- R a is selected from a C 1 -C 6 alkyl group, a C 2 -C 6 alkenyl group, a C 2 -C 6 alkynyl group, a C 3 -C 6 cycloalkyl group, a 4-12 membered heterocyclic group;
- An alkenyl, alkynyl, cycloalkyl, and heterocyclic group is optionally further substituted with one or more -QT groups;
- R 6 and R 12 are each independently selected from the group consisting of hydrogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl;
- R 7 and R 9 are each independently selected from the group consisting of hydrogen, halogen, and R b ;
- R b is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclyl, said alkyl group,
- the alkenyl, alkynyl, cycloalkyl, and heterocyclic groups are optionally further selected from the group consisting of halogen, cyano, hydroxy, C 1 -C 6 alkoxy, C 1 -C 6 alkyl-OC(O)-, Substituting one or more groups of an amino group, a C 1 -C 6 alkylamino group, a bis C 1 -C 6 alkylamino group, a 4-12 membered heterocyclic group;
- R 8 , R 10 , R 11 are each independently selected from hydrogen, halogen, C 1 -C 3 alkyl or cycloalkyl;
- Q is selected from a bond or a C 1 -C 6 alkylene group, which is optionally further substituted with one or more groups selected from the group consisting of halogen, cyano, hydroxy, C 1 -C 6 alkoxy ;
- T is selected from the group consisting of hydrogen, halogen, cyano, hydroxy, -NR h R i , -C(O)R h , OR h , -C(O)OR h , C(O)NR h R i , -NR h C (O) R i , -NR j C(O)NR h R i , -NR h C(O)OR i , or R k ;
- R k is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclyl, 5 to 10 membered hetero
- An aryl group, an aryl group, R k is optionally further substituted with one or more -Q 1 -T 1 groups;
- R h , R i , R j are each independently selected from hydrogen or R l , and R l is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclyl, 5- or 6-membered heteroaryl, and aryl groups, R l optionally further substituted with one or more -Q 1 -T 1 groups;
- R h , R i together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further substituted by one or more —Q 1 —T 1 Group substitution
- Q 1 is a bond or a C 1 -C 6 alkylene group, which is optionally further substituted with one or more groups selected from the group consisting of halogen, cyano, hydroxy, C 1 -C 6 alkoxy ;
- T 1 is selected from the group consisting of hydrogen, halogen, cyano, hydroxy, -NR m R n , -C(O)R m , OR m , -C(O)OR m , C(O)NR m R n , -NR m C(O)R n , -NR o C(O)NR m R n , -NR m C(O)OR n , -O(CH 2 ) a NR m R n , -(CH 2 ) a NR m R n , -S(O) 2 NR m R n or R p ;
- R p is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclyl, 5 or 6 Heteroaryl, aryl;
- R m , R n , R o are each independently selected from hydrogen or R q , and R q is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 - C 6 cycloalkyl, 4-12 membered heterocyclyl, 5- or 6-membered heteroaryl group, an aryl group, R q is optionally further substituted selected from halogen, hydroxyl, 5- or 6-membered heteroaryl group, an aryl group, Substituting one or more groups, wherein the heteroaryl or aryl group is optionally further substituted with one or more groups selected from C 1 -C 6 alkyl, hydroxy, oxo;
- R m , R n together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further selected from the group consisting of halogen, hydroxy, oxo, C Substituting one or more groups of 1 -C 6 alkyl, -OR x , -NR x R y , -C(O)R x , -O(CH 2 ) a OR x ;
- R x and R y are each independently selected from hydrogen or R z , and R z is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 naphthenic a 4- to 4-membered heterocyclic group, a 5- or 6-membered heteroaryl group, an aryl group, and R q is optionally further selected from one or more of a halogen, a hydroxyl group, a 5- or 6-membered heteroaryl group, and an aryl group. a group substitution, wherein the heteroaryl or aryl group is optionally further substituted with one or more groups selected from the group consisting of C 1 -C 6 alkyl, hydroxy, oxo;
- R x , R y together with the N atom to which they are attached form a 4-12 membered heterocyclic group containing 0 or 1 additional hetero atom;
- a is an integer from 1 to 4.
- the compound of the formula (I), or a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof, according to the invention is a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof, according to the invention.
- Ar is selected from
- X is selected from CR 14 ;
- Y is selected from N or CR 15 ;
- Z is selected from N or CR 16 ;
- R 13 is selected from the group consisting of hydrogen, halogen, C 1 -C 6 alkyl or C 3 -C 6 cycloalkyl;
- R 14 , R 15 and R 16 are each independently selected from the group -QT;
- Q is selected from a bond or a C 1 -C 6 alkylene group
- T is selected from the group consisting of hydrogen, halogen, cyano, hydroxy, -NR h R i , -C(O)R h , OR h , -C(O)OR h , C(O)NR h R i , -NR h C (O) R i , -NR j C(O)NR h R i , -NR h C(O)OR i , or R k ;
- R k is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclyl, 5 to 10 membered hetero
- An aryl group, an aryl group, R k is optionally further substituted with one or more -Q 1 -T 1 groups;
- R h , R i , R j are each independently selected from hydrogen or R l , and R l is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclyl, 5- or 6-membered heteroaryl, and aryl groups, R l optionally further substituted with one or more -Q 1 -T 1 groups;
- R h , R i together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further substituted by one or more —Q 1 —T 1 Group substitution
- Q 1 is a bond or a C 1 -C 6 alkylene group
- T 1 is selected from the group consisting of hydrogen, halogen, cyano, hydroxy, -NR m R n , -C(O)R m , OR m , -C(O)OR m , C(O)NR m R n , -NR m C(O)R n , -NR o C(O)NR m R n , -NR m C(O)OR n , -O(CH 2 ) a NR m R n , -(CH 2 ) a NR m R n , -S(O) 2 NR m R n or R p ;
- R p is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclyl, 5 or 6 Heteroaryl, aryl;
- R m , R n , R o are each independently selected from hydrogen or R q , and R q is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C a 6 cycloalkyl group, a 4-12 membered heterocyclic group, a 5- or 6-membered heteroaryl group, an aryl group, and R q is optionally further selected from the group consisting of halogen, hydroxy, 5- or 6-membered heteroaryl, aryl Substituting a group or groups, wherein the heteroaryl or aryl group is optionally further substituted with one or more groups selected from the group consisting of C 1 -C 6 alkyl, hydroxy, oxo,
- R m , R n together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further selected from the group consisting of halogen, hydroxy, oxo, C Substituting one or more groups of 1 -C 6 alkyl, -OR x , -NR x R y , -C(O)R x , -O(CH 2 ) a OR x ;
- R x and R y are each independently selected from hydrogen or R z , and R z is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 naphthenic a 4- to 4-membered heterocyclic group, a 5- or 6-membered heteroaryl group, an aryl group, and R q is optionally further selected from one or more of a halogen, a hydroxyl group, a 5- or 6-membered heteroaryl group, and an aryl group. a group substitution, wherein the heteroaryl or aryl group is optionally further substituted with one or more groups selected from the group consisting of C 1 -C 6 alkyl, hydroxy, oxo;
- R x , R y together with the N atom to which they are attached form a 4-12 membered heterocyclic group containing 0 or 1 additional hetero atom;
- a is an integer from 1 to 4.
- the compound of the formula (I) or a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof according to the present invention is a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof according to the present invention.
- R 13 is selected from the group consisting of hydrogen, halogen, and C 1 -C 6 alkyl
- X is selected from CR 14 ;
- Y is selected from CR 15 ;
- Z is selected from CR 16 ;
- R 14 is selected from the group -QT
- R 15 and R 16 are each independently selected from hydrogen or halogen
- Q is selected from a bond or a C 1 -C 6 alkylene group
- T is selected from the group consisting of hydrogen, halogen, cyano, hydroxy, -NR h R i , -C(O)R h , OR h , -C(O)OR h , C(O)NR h R i , -NR h C (O) R i , -NR j C(O)NR h R i , -NR h C(O)OR i , or R k ;
- R k is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclyl, 5 to 10 membered hetero
- R h , R i , R j are each independently selected from hydrogen or R l , and R l is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclyl, 5- or 6-membered heteroaryl, and aryl groups, R l optionally further substituted with one or more -Q 1 -T 1 groups;
- R h , R i together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further substituted by one or more —Q 1 —T 1 Group substitution
- Q 1 is a bond or a C 1 -C 6 alkylene group
- T 1 is selected from the group consisting of hydrogen, halogen, cyano, hydroxy, -NR m R n , -C(O)R m , OR m , -C(O)OR m , C(O)NR m R n , -NR m C(O)R n , -NR o C(O)NR m R n , -NR m C(O)OR n , -O(CH 2 ) a NR m R n , -(CH 2 ) a NR m R n , -S(O) 2 NR m R n or R p ;
- R p is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclyl, 5 or 6 Heteroaryl, aryl;
- R m , R n , R o are each independently selected from hydrogen or R q , and R q is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C a 6 cycloalkyl group, a 4-12 membered heterocyclic group, a 5- or 6-membered heteroaryl group, an aryl group, and R q is optionally further selected from the group consisting of halogen, hydroxy, 5- or 6-membered heteroaryl, aryl Substituting a group or groups, wherein the heteroaryl or aryl group is optionally further substituted with one or more groups selected from the group consisting of C 1 -C 6 alkyl, hydroxy, oxo,
- R m , R n together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further selected from the group consisting of halogen, hydroxy, oxo, C Substituting one or more groups of 1 -C 6 alkyl, -OR x , -NR x R y , -C(O)R x , -O(CH 2 ) a OR x ;
- R x and R y are each independently selected from hydrogen or R z , and R z is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 naphthenic a 4- to 4-membered heterocyclic group, a 5- or 6-membered heteroaryl group, an aryl group, and R q is optionally further selected from one or more of a halogen, a hydroxyl group, a 5- or 6-membered heteroaryl group, and an aryl group. a group substitution, wherein the heteroaryl or aryl group is optionally further substituted with one or more groups selected from the group consisting of C 1 -C 6 alkyl, hydroxy, oxo;
- R x , R y together with the N atom to which they are attached form a 4-12 membered heterocyclic group containing 0 or 1 additional hetero atom;
- a is an integer from 1 to 4.
- the compound of the formula (I) or a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof according to the present invention wherein:
- Q is selected from a bond or a C 1 -C 6 alkylene group
- T is selected from the group consisting of hydrogen, halogen, -NR h R i , OR h or R k ;
- R k is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, 4-12 membered heterocyclic, 5 to 10 membered heteroaryl, C 6 -C 10 aryl Or the alkyl, alkenyl, alkynyl, heterocyclyl, heteroaryl or aryl group is optionally further substituted with one or more -Q 1 -T 1 groups;
- R h , R i are each independently selected from hydrogen or R l , R l is selected from C 1 -C 6 alkyl, 5- or 6-membered heteroaryl, and C 6 -C 10 aryl, R l optionally further Substituted by one or more -Q 1 -T 1 groups;
- R h , R i together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further substituted by one or more —Q 1 —T 1 Group substitution
- Q 1 is a bond or a C 1 -C 6 alkylene group
- T 1 is selected from the group consisting of hydrogen, halogen, hydroxyl, -NR m R n , OR m , -C(O)OR m , -C(O)NR m R n , -O(CH 2 ) a NR m R n ,- (CH 2 ) a NR m R n , -S(O) 2 NR m R n or R p ;
- R p is selected from a C 1 -C 6 alkyl group, a 4-12 membered heterocyclic group
- R m , R n are each independently selected from hydrogen, C 1 -C 6 alkyl, which is optionally further substituted with one or more groups selected from hydroxy or 5- or 6-membered heteroaryl groups, Wherein the heteroaryl group is optionally further substituted with one or more groups selected from C 1 -C 6 alkyl, oxo;
- R m , R n together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further selected from the group consisting of halogen, hydroxy, oxo, C 1 Substituting one or more groups of -C 6 alkyl, -OR x , -NR x R y , -C(O)R x , -O(CH 2 ) a OR x ;
- R x and R y are each independently selected from hydrogen or R z , and R z is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 naphthenic a 4- to 4-membered heterocyclic group, a 5- or 6-membered heteroaryl group, an aryl group, and R q is optionally further selected from one or more of a halogen, a hydroxyl group, a 5- or 6-membered heteroaryl group, and an aryl group. a group substitution, wherein the heteroaryl or aryl group is optionally further substituted with one or more groups selected from the group consisting of C 1 -C 6 alkyl, hydroxy, oxo;
- R x , R y together with the N atom to which they are attached form a 4-12 membered heterocyclic group containing 0 or 1 additional hetero atom;
- a is an integer from 1 to 4.
- the compound of the formula (I) or a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof according to the present invention wherein:
- Q is selected from a bond
- T is selected from the group consisting of hydrogen, halogen, -NR h R i , OR h or R k ;
- R k is selected from C 1 -C 6 alkyl, C 2 -C 6 alkynyl, or phenyl, pyrimidinyl, pyridyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl or thiophene [3,2- b] a thiophene group, said alkyl, alkynyl, phenyl, pyrimidinyl, pyridyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl or thiophene [3,2-b] thiophene group Further selected to be substituted by one or more -Q 1 -T 1 groups;
- R h , R i are each independently selected from the group consisting of hydrogen, pyridyl, pyrimidinyl and phenyl, optionally substituted by one or more —Q 1 —T 1 groups;
- R h , R i together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further substituted by one or more —Q 1 —T 1 Group substitution
- Q 1 is a bond or a C 1 -C 6 alkylene group
- T 1 is selected from the group consisting of hydrogen, halogen, hydroxyl, -NR m R n , OR m , -C(O)OR m , -C(O)NR m R n , -O(CH 2 ) a NR m R n ,- (CH 2 ) a NR m R n , -S(O) 2 NR m R n or R p ;
- R p is selected from C 1 -C 6 alkyl
- R m and R n are each independently selected from hydrogen, C 1 -C 6 alkyl, and the alkyl group is optionally further substituted with one or more groups selected from a hydroxyl group or a 5- or 6-membered heteroaryl group.
- the heteroaryl group is optionally further substituted with one or more groups selected from C 1 -C 6 alkyl, oxo;
- R m , R n together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further selected from the group consisting of halogen, hydroxy, oxo, C Substituting one or more groups of 1 -C 6 alkyl, -OR x , -NR x R y , -C(O)R x , -O(CH 2 ) a OR x ;
- R x and R y are each independently selected from hydrogen or R z , and R z is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 naphthenic a 4- to 4-membered heterocyclic group, a 5- or 6-membered heteroaryl group, an aryl group, and R q is optionally further selected from one or more of a halogen, a hydroxyl group, a 5- or 6-membered heteroaryl group, and an aryl group. a group substitution, wherein the heteroaryl or aryl group is optionally further substituted with one or more groups selected from the group consisting of C 1 -C 6 alkyl, hydroxy, oxo;
- R x , R y together with the N atom to which they are attached form a 4-12 membered heterocyclic group containing 0 or 1 additional hetero atom;
- a is an integer from 1 to 4.
- the compound of the formula (I) or a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof according to the present invention wherein:
- Q is selected from a bond
- T is selected from the group consisting of hydrogen, halogen, -NR h R i , OR h or R k ;
- R k is selected from C 1 -C 6 alkyl, C 2 -C 6 alkynyl, or phenyl, pyrimidinyl, pyridyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl or thiophene [3,2- b] a thiophene group, said alkyl, alkynyl, phenyl, pyrimidinyl, pyridyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl or thiophene [3,2-b] thiophene group Further selected to be substituted by one or more -Q 1 -T 1 groups;
- R h , R i are each independently selected from the group consisting of hydrogen, pyridyl, pyrimidinyl, and phenyl, which are optionally further substituted with one or more -Q 1 -T 1 groups;
- R h , R i together with the N atom to which they are attached form a piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl group, the piperidinyl, piperazinyl, pyrrolidinyl, morpholinyl optionally further Substituted by one or more -Q 1 -T 1 groups;
- Q 1 is a bond or a C 1 -C 6 alkylene group
- T 1 is selected from the group consisting of hydrogen, halogen, hydroxyl, -NR m R n , OR m , -C(O)OR m , -C(O)NR m R n , -O(CH 2 ) a NR m R n ,- (CH 2 ) a NR m R n , -S(O) 2 NR m R n or R p ;
- R p is selected from C 1 -C 6 alkyl
- R m , R n are each independently selected from hydrogen, C 1 -C 6 alkyl, which is optionally further substituted with one or more groups selected from hydroxy or 5- or 6-membered heteroaryl groups, Wherein the heteroaryl group is optionally further substituted with one or more groups selected from C 1 -C 6 alkyl, oxo;
- R m , R n together with the N atom to which they are attached form a 4-12 membered heterocyclyl containing 0 or 1 additional heteroatom, optionally further selected from the group consisting of halogen, hydroxy, oxo, C One or more groups of 1 -C 6 alkyl, -OR x , C 1 -C 6 alkoxy, -NR x R y , -C(O)R x , -O(CH 2 ) a OR x Substitute
- R x and R y are each independently selected from hydrogen or R z , and R z is selected from C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 naphthenic a 4- to 4-membered heterocyclic group, a 5- or 6-membered heteroaryl group, an aryl group, and R q is optionally further selected from one or more of a halogen, a hydroxyl group, a 5- or 6-membered heteroaryl group, and an aryl group. a group substitution, wherein the heteroaryl or aryl group is optionally further substituted with one or more groups selected from the group consisting of C 1 -C 6 alkyl, hydroxy, oxo;
- R x , R y together with the N atom to which they are attached form a 4-12 membered heterocyclic group containing 0 or 1 additional hetero atom;
- a is an integer from 1 to 4.
- the compound of the formula (I) or a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof according to the present invention wherein:
- Q is selected from a bond
- T is selected from the group consisting of hydrogen, halogen, -NR h R i , OR h or R k ;
- R k is selected from a C 1 -C 6 alkyl group, a C 2 -C 6 alkynyl group, or a phenyl, pyrimidinyl, pyridyl, or thiophene [3,2-b]thiophene group, said alkyl group, alkynyl group a phenyl, pyrimidinyl, pyridyl, or thiophene [3,2-b]thiophene group, optionally further substituted with one or more -Q 1 -T 1 groups;
- R h , R i are each independently selected from the group consisting of hydrogen, pyridyl, pyrimidinyl, and phenyl, which are optionally further substituted with one or more -Q 1 -T 1 groups;
- R h , R i together with the N atom to which they are attached form a piperidinyl, piperazinyl, morpholinyl group, optionally further one or more -Q 1 -T 1 group substitution;
- Q 1 is a bond or a C 1 -C 6 alkylene group
- T 1 is selected from the group consisting of hydrogen, halogen, hydroxyl, -NR m R n , OR m , -C(O)OR m , -C(O)NR m R n , -O(CH 2 ) a NR m R n ,- (CH 2 ) a NR m R n , -S(O) 2 NR m R n or R p ;
- R p is selected from C 1 -C 6 alkyl
- R m , R n are each independently selected from hydrogen, C 1 -C 6 alkyl, and the alkyl group is optionally further substituted with one or more groups selected from hydroxy or pyridyl, pyrimidinyl, wherein Pyridyl or pyrimidinyl is optionally further substituted with one or more groups selected from C 1 -C 6 alkyl, oxo;
- R m , R n together with the N atom to which they are attached form a piperidinyl, morpholinyl, pyrrolidinyl, piperazinyl, homopiperazinyl, azetidinyl, azaspiro ring, said piperidinyl, morpholinyl, pyrrolidinyl, piperazinyl, homopiperazinyl, azetidinyl, aza-spiro ring is optionally further selected from halo, oxo, C 1 -C 6 alkyl, Substituting one or more groups of -OR x , -NR x R y , -C(O)R x , -O(CH 2 ) a OR x ;
- R x and R y are each independently selected from hydrogen or R z , R z is selected from C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, and R q is optionally further selected from the group consisting of halogen and hydroxyl. Or a plurality of groups;
- R x , R y together with the N atom to which they are attached form a 4-12 membered heterocyclic group containing 0 or 1 additional hetero atom; preferably piperidinyl, morpholinyl, pyrrolidinyl, piperazinyl, homoperazine Azinyl group;
- a is an integer from 1 to 4.
- the compound of the formula (I) or a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof according to the present invention is of the following formula ( a compound represented by IIA), formula (IIB), formula (IIC), formula (IID), formula (IIE) or formula (IIF) or a pharmaceutically acceptable salt, solvate or metabolite thereof Or a prodrug,
- R 1 to R 12 and Ar are as defined in the above formula (I).
- the compound of the formula (I) or a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof according to the present invention is of the formula (IIA) a compound of the formula (IIB), formula (IIC), formula (IID), formula (IIE) or formula (IIF) or a pharmaceutically acceptable salt, solvate, metabolite thereof or Prodrug, among them,
- R 1 and R 2 are selected from the group consisting of hydrogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclic;
- R 3 and R 4 are selected from hydrogen
- R 5 is selected from hydrogen, C 1 -C 6 alkyl, which is optionally further substituted with one or more -QT groups;
- R 6 and R 12 are selected from hydrogen
- R 7 and R 9 are selected from the group consisting of hydrogen and C 1 -C 6 alkyl
- R 8 , R 10 and R 11 are selected from hydrogen
- -QT is a -NR h R i group
- R h , R i are each independently selected from hydrogen and C 1 -C 6 alkyl
- R h , R i together with the N atom to which they are attached form a 4-12 membered heterocyclic group containing 0 or 1 additional hetero atom; preferably piperidinyl, piperazinyl, morpholinyl, pyrrolidinyl;
- Ar is as defined in the formula (IIA), the formula (IIB), the formula (IIC), the formula (IID), the formula (IIE) or the formula (IIF).
- the compound of the formula (I) or a pharmaceutically acceptable salt, solvate, metabolite or prodrug thereof according to the present invention is of the formula (IIA) a compound of the formula (IIB), formula (IIC), formula (IID), formula (IIE) or formula (IIF) or a pharmaceutically acceptable salt, solvate, metabolite thereof or Prodrug, among them,
- Ar is a phenyl group, which is optionally further substituted with one or more -Q-T groups;
- R 1 and R 2 are selected from the group consisting of hydrogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, 4-12 membered heterocyclic;
- R 3 and R 4 are selected from hydrogen
- R 5 is selected from hydrogen, C 1 -C 6 alkyl, which is optionally further substituted with one or more -QT groups;
- R 6 and R 12 are selected from hydrogen
- R 7 and R 9 are selected from the group consisting of hydrogen and C 1 -C 6 alkyl
- R 8 , R 10 and R 11 are selected from hydrogen
- -QT is -phenyl, optionally substituted by one or more -Q 1 -T 1 groups
- Q 1 is a bond or a C 1 -C 6 alkylene group
- T 1 is selected from hydrogen, halo, hydroxy, -NR m R n, OR m , -C (O) OR m, -C (O) NR m R n, -O (CH 2) a NR m R n, - S(O) 2 NR m R n ;
- R m , R n are each independently selected from hydrogen or R q , R q is selected from C 1 -C 6 alkyl, and R q is optionally further selected from the group consisting of halogen, hydroxy, 5- or 6-membered heteroaryl Or a plurality of group substitutions, wherein the heteroaryl or aryl group is optionally further substituted with one or more groups selected from the group consisting of C 1 -C 6 alkyl, hydroxy, oxo;
- R m , R n together with the N atom to which they are attached form a 4-12 membered heterocyclic group containing 0 or 1 additional hetero atom, preferably piperidinyl, piperazinyl, homopiperazinyl, morpholinyl, pyrrole
- R x and R y are each independently selected from hydrogen or R z , and R z is selected from C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl;
- R x , R y together with the N atom to which they are attached form a 4-12 membered heterocyclic group containing 0 or 1 additional hetero atom; preferably piperidinyl, piperazinyl, morpholinyl, pyrrolidinyl;
- a is an integer from 1 to 4.
- Exemplary compounds of the invention include, but are not limited to, the compounds of Table 1 below, or pharmaceutically acceptable salts, solvates, metabolites or prodrugs thereof.
- Another aspect of the invention relates to a deuterated compound of the compound of the formula (I) according to the invention, wherein one or more H atoms of the compound of the formula (I) are independently replaced by a D atom,
- the compounds include, but are not limited to:
- Another aspect of the invention relates to a process for the preparation of a compound of the formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof according to the invention, which comprises the steps of:
- Step 1) compound (Ia) and compound (Ib) condensation reaction in the presence of a condensing agent such as HATU, to obtain compound (Ic);
- Step 2) Compound (Ic) is hydrolyzed under acidic conditions to obtain compound (Id);
- Step 3 Compound (Id) is reacted with the corresponding R to obtain a compound of the formula (I);
- reaction of R with an amino group may be a condensation reaction of a carboxylic acid form of R with an amine condensing agent such as HATU, or a form of an acid chloride, an acid anhydride or a mixed acid anhydride of R may be bonded to the amino group by an amine hydrolysis reaction.
- an amine condensing agent such as HATU
- a form of an acid chloride, an acid anhydride or a mixed acid anhydride of R may be bonded to the amino group by an amine hydrolysis reaction.
- the invention further relates to a pharmaceutical composition
- a pharmaceutical composition comprising a compound of the formula (I) according to the invention, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or according to the invention a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug deuterated compound thereof, and one or more pharmaceutically acceptable carriers.
- the invention further relates to a compound of the formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof, according to the invention, or a compound of the formula (I) according to the invention Or a pharmaceutically acceptable salt, solvate or prodrug deuterated compound, or a pharmaceutical composition comprising the same, for use in the preparation of an EZH2 histone methyltransferase inhibitor.
- Another aspect of the invention relates to a compound of the formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof, according to the invention, or to the formula (I) according to the invention
- the diseases associated with the EZH2 histone methyltransferase may be cancer, diabetes inflammation, immune system diseases, cardiovascular diseases, neurological diseases, and respiratory diseases.
- the cancer may be lung cancer, gastric cancer, liver cancer, breast cancer, nasopharyngeal cancer, pancreatic cancer, ovarian cancer, cervical cancer, colorectal cancer, glioma, melanoma, prostate cancer, kidney cancer, esophageal cancer, mesothelium.
- the invention further relates to a compound of the formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof, according to the invention, or a compound of the formula (I) according to the invention Or a pharmaceutically acceptable salt, solvate or prodrug deuterated compound, or a pharmaceutical composition comprising the same, for use as an EZH2 histone methyltransferase inhibitor.
- Another aspect of the invention relates to a compound of the formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof, according to the invention, or to the formula (I) according to the invention
- the diseases associated with the EZH2 histone methyltransferase may be cancer, diabetes inflammation, immune system diseases, cardiovascular diseases, neurological diseases, and respiratory diseases.
- the cancer may be lung cancer, gastric cancer, liver cancer, breast cancer, nasopharyngeal cancer, pancreatic cancer, ovarian cancer, cervical cancer, colorectal cancer, glioma, melanoma, prostate cancer, kidney cancer, esophageal cancer, mesothelium.
- Another aspect of the invention relates to a method of inhibiting EZH2 histone methyltransferase comprising administering to a patient in need thereof an effective amount of a compound of formula (I) according to the invention or a pharmaceutically acceptable compound thereof Accepted salt, solvate or prodrug, or a deuterated compound of the compound of the formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof according to the present invention, or a drug containing the same combination.
- Another aspect of the invention relates to a method of treating a disease associated with EZH2 histone methyltransferase, comprising administering to a patient in need thereof an effective amount of a compound of formula (I) according to the invention or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a deuterated compound of the compound of the formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof according to the present invention, or A pharmaceutical composition containing the same.
- the diseases associated with the EZH2 histone methyltransferase may be cancer, diabetes inflammation, immune system diseases, cardiovascular diseases, neurological diseases, and respiratory diseases.
- the cancer may be lung cancer, gastric cancer, liver cancer, breast cancer, nasopharyngeal cancer, pancreatic cancer, ovarian cancer, cervical cancer, colorectal cancer, glioma, melanoma, prostate cancer, kidney cancer, esophageal cancer, mesothelium.
- alkyl refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group having from 1 to 12 carbon atoms, more preferably from 1 to 6 carbons.
- the alkyl group of the atom is a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group having from 1 to 12 carbon atoms, more preferably from 1 to 6 carbons.
- Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1 ,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2- Methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3 - dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2 -methylhexyl, 3-methylhexyl, 4-methylhexyl,
- lower alkyl groups having from 1 to 6 carbon atoms, non-limiting examples including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl Base, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethyl Butyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl Base, 2,3-dimethylbutyl and the like.
- the alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably one or more of the following groups independently selected from the group consisting of an alkane Base, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, naphthenic An oxy group, a heterocycloalkoxy group, a cycloalkylthio group, a heterocycloalkylthio group, an oxo group, a carboxyl group or a carboxylate group.
- an alkane Base alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, hetero
- alkylene means that one hydrogen atom of the alkyl group is further substituted, for example, "methylene” refers to -CH 2 -, "ethylene” refers to -(CH 2 ) 2 -, "propylene” Refers to -(CH 2 ) 3 -, "butylene” means -(CH 2 ) 4 - and the like.
- alkenyl refers to an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon double bond, such as ethenyl, 1-propenyl, 2-propenyl, 1-, 2- or -butenyl and the like.
- the alkenyl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio group.
- alkynyl refers to an alkyl radical as defined above consisting of at least two carbon atoms and at least one carbon-carbon triple bond, such as ethynyl, propynyl, butynyl and the like.
- the alkynyl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio group.
- cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably from 3 to 6 carbon atoms. One carbon atom.
- Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene
- a polycycloalkyl group includes a spiro ring, a fused ring, and a cycloalkyl group.
- spirocycloalkyl refers to a polycyclic group that shares a carbon atom (referred to as a spiro atom) between 5 to 20 members of a single ring, which may contain one or more double bonds, but none of the rings have a fully conjugated ⁇ electronic system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
- the spirocycloalkyl group is classified into a monospirocycloalkyl group, a bispirocycloalkyl group or a polyspirocycloalkyl group, preferably a monospirocycloalkyl group and a bispirocycloalkyl group, depending on the number of common spiro atoms between the rings.
- spirocycloalkyl groups include:
- fused cycloalkyl refers to 5 to 20 members, and each ring in the system shares an all-carbon polycyclic group of an adjacent pair of carbon atoms with other rings in the system, wherein one or more of the rings may contain one or Multiple double bonds, but none of the rings have a fully conjugated ⁇ -electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
- fused cycloalkyl groups include:
- bridged cycloalkyl refers to an all-carbon polycyclic group of 5 to 20 members, any two rings sharing two carbon atoms which are not directly bonded, which may contain one or more double bonds, but none of the rings have complete Conjugate ⁇ -electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members. Depending on the number of constituent rings, it may be classified into a bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl group, preferably a bicyclic ring, a tricyclic ring or a tetracyclic ring, and more preferably a bicyclic ring or a tricyclic ring.
- bridged cycloalkyl groups include:
- the cycloalkyl ring may be fused to an aryl, heteroaryl or heterocycloalkyl ring, wherein the ring to which the parent structure is attached is a cycloalkyl group, non-limiting examples include indanyl, tetrahydronaphthalene Base, benzocycloheptyl and the like.
- the cycloalkyl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio a heterocycloalkylthio group, an oxo group, a carboxyl group or a carboxylate group.
- heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms wherein one or more ring atoms are selected from nitrogen, oxygen or S(O).
- a hetero atom of m (where m is an integer of 0 to 2), but excluding the ring moiety of -OO-, -OS- or -SS-, the remaining ring atoms being carbon.
- ring atoms Preferably comprising from 3 to 12 ring atoms, wherein from 1 to 4 are heteroatoms; most preferably from 3 to 8 ring atoms, wherein from 1 to 3 are heteroatoms; most preferably from 5 to 6 ring atoms, wherein 1 to 2 or 1 to 3 are heteroatoms.
- monocyclic heterocyclic groups include pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperidine.
- the group, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl and the like are preferably 1, 2, 5-oxadiazolyl, pyranyl or morpholinyl.
- Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
- spiroheterocyclyl refers to a polycyclic heterocyclic group in which one atom (called a spiro atom) is shared between 5 to 20 members of a single ring, wherein one or more ring atoms are selected from nitrogen, oxygen or S (O). ) m (where m is an integer 0 to 2) heteroatoms, and the remaining ring atoms are carbon. It may contain one or more double bonds, but none of the rings have a fully conjugated pi-electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
- the spiroheterocyclyl group is classified into a monospiroheterocyclic group, a dispiroheterocyclic group or a polyspirocyclic group according to the number of shared spiro atoms between the ring and the ring, and is preferably a monospiroheterocyclic group and a dispiroheterocyclic group. More preferably, it is 4 yuan / 4 yuan, 4 yuan / 5 yuan, 4 yuan / 6 yuan, 5 yuan / 5 yuan or 5 yuan / 6-membered monospiroheterocyclic group.
- Non-limiting examples of spiroheterocyclyl groups include:
- fused heterocyclyl refers to 5 to 20 members, and each ring in the system shares an adjacent pair of atomic polycyclic heterocyclic groups with other rings in the system, and one or more rings may contain one or more Double bond, but none of the rings have a fully conjugated ⁇ -electron system in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), and the remaining rings
- the atom is carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
- fused heterocyclic groups include:
- bridge heterocyclyl refers to a polycyclic heterocyclic group of 5 to 14 members, any two rings sharing two atoms which are not directly bonded, which may contain one or more double bonds, but none of the rings have a total A ⁇ -electron system of a yoke in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), the remaining ring atoms being carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
- bridge heterocyclic groups include:
- the heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heterocyclic group, non-limiting examples of which include:
- the heterocyclic group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio a heterocycloalkylthio group, an oxo group, a carboxyl group or a carboxylate group.
- aryl refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic ring (ie, a ring that shares a pair of adjacent carbon atoms) having a conjugated ⁇ -electron system, preferably 6 to 10 members, such as benzene. Base and naphthyl. More preferred is phenyl.
- the aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring to which the parent structure is attached is an aryl ring, non-limiting examples of which include:
- the aryl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle An alkylthio group, a carboxyl group or a carboxylate group.
- heteroaryl refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms are selected from the group consisting of oxygen, sulfur and nitrogen.
- the heteroaryl group is preferably 5 to 10 members, and has 1 to 3 hetero atoms; more preferably 5 or 6 members, and 1 to 2 hetero atoms; preferably, for example, imidazolyl, furyl, thienyl, thiazolyl, pyridyl An oxazolyl, oxazolyl, pyrrolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl, etc., preferably imidazolyl, thiazolyl, pyrazolyl or pyrimidinyl, thiazolyl; more selective Pyrazolyl or thiazolyl.
- the heteroaryl ring may be fused to an aryl, hetero
- the heteroaryl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio a heterocycloalkylthio group, a carboxyl group or a carboxylate group.
- alkoxy refers to -O-(alkyl) and -O-(unsubstituted cycloalkyl), wherein alkyl is as defined above.
- alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
- the alkoxy group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio a heterocycloalkylthio group, a carboxyl group or a carboxylate group.
- deuterated refers to a modification in which a hydrogen atom in an original compound is replaced with a deuterium atom.
- hydroxy refers to an -OH group.
- halogen means fluoro, chloro, bromo or iodo.
- amino means -NH 2.
- cyano refers to -CN.
- nitro refers to -NO 2 .
- heterocyclic group optionally substituted by an alkyl group means that an alkyl group may be, but not necessarily, present, and the description includes the case where the heterocyclic group is substituted with an alkyl group and the case where the heterocyclic group is not substituted with an alkyl group.
- Substituted refers to one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3, hydrogen atoms, independently of each other, substituted by a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and those skilled in the art will be able to determine (by experiment or theory) substitutions that may or may not be possible without undue effort. For example, an amino group or a hydroxyl group having a free hydrogen may be unstable when combined with a carbon atom having an unsaturated (e.g., olefinic) bond.
- “Pharmaceutical composition” means a mixture comprising one or more of the compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, as well as other components such as physiological/pharmaceutically acceptable carriers. And excipients.
- the purpose of the pharmaceutical composition is to promote the administration of the organism, which facilitates the absorption of the active ingredient and thereby exerts biological activity.
- “Pharmaceutically acceptable salt” refers to a salt of a compound of the invention which is safe and effective for use in a mammal and which possesses the desired biological activity.
- the present invention adopts the following technical solutions.
- the nitration or halogenation reaction is a conventional reaction well known in the art, for example, Reacting with NBS or KNO 3 in the presence of sulfuric acid;
- Step 2) The obtained intermediate a is reacted with ethyl bromide under basic conditions to obtain intermediate b, and the basic condition may be potassium carbonate;
- Step 3 reduction of the nitro compound of intermediate b to amino compound intermediate c, which is also known to those skilled in the art, such as the iron powder ammonium chloride process;
- Step 4) reductive amination reaction of intermediate c with 1-Boc-3-pyrrolidone in the presence of a catalyst under acidic conditions to obtain intermediate d, such as sodium triacetoxyborohydride;
- Step 5 another intermediate reductive reaction of intermediate d with acetaldehyde in the presence of a catalyst under acidic conditions to obtain intermediate e;
- Step 7) amidation of intermediate f with the corresponding aminomethylpyridone compound in the presence of a catalyst to obtain intermediate g, which may be HATU or N-methylmorpholine;
- Step 8) The Boc of the intermediate g is first hydrolyzed and then reacted with acrylic anhydride in the presence of a catalyst to prepare a compound of the invention, such as N-methylmorpholine.
- the compound of the present invention is first obtained by reacting the compound h obtained as in the above Scheme 1, and then reacting the compound h with the corresponding pinacol ester of phenylboronic acid in the presence of a catalyst.
- the intermediate i is prepared according to the method of the above scheme 1, and the intermediate i is reacted with the boronic acid pinacol ester in the presence of a catalyst to obtain an intermediate j; then the intermediate j is reacted with dichloropyrimidine to obtain an intermediate. k; intermediate k is reacted with a corresponding heterocyclic ring such as piperidine under basic conditions to obtain intermediate l; the ester of intermediate k is hydrolyzed to obtain intermediate m, and the ester of intermediate l is hydrolyzed to obtain intermediate n;
- the compound of the present invention can be obtained by substituting the intermediate m or n for the intermediate f according to the preparation method of the above scheme 1.
- intermediate i is reacted with pinacol hydroxymethylphenylboronic acid in the presence of a catalyst to obtain intermediate o; then intermediate o is reacted with methanesulfonyl chloride to obtain intermediate p; The various amines are reacted to produce intermediate q; then, the compound of the invention is prepared by substituting intermediate q for intermediate e according to the procedure of Scheme 1 above.
- the intermediate r is prepared by the above scheme 1 and then the intermediate r is reacted with the corresponding amine in the presence of a catalyst to prepare an intermediate s; then, using the preparation method of the scheme 1, the intermediate s is used instead of the intermediate g.
- the compounds of the invention are prepared.
- Scheme 6 is a process for the preparation of a particular class of compounds of the invention.
- the intermediate t is first prepared by Scheme 1, and then the intermediate t is reacted with 4-dimethylamino crotonate to prepare the compound of the present invention.
- R 7 , R 9 , R 13 , R m , R n , R h , R i and -Q 1 -T 1 are as defined in the formula (I).
- the pharmaceutically acceptable salt of the compound of the formula (I) of the present invention may be an acid addition salt or a basic addition salt.
- Acid-selectable inorganic acids include, but are not limited to, hydrochloric acid, sulfuric acid, phosphoric acid, hydrobromic acid; acids may also be selected from organic acids including, but not limited to, citric acid, maleic acid, oxalic acid, formic acid, acetic acid, propionic acid, glycolic acid, benzoic acid.
- Base selectable inorganic bases include, but are not limited to, sodium hydroxide, potassium hydroxide, magnesium hydroxide, calcium hydroxide; bases may also be selected from organic bases including, but not limited to, ammonium hydroxide, triethylamine, arginine or lysine .
- the compound of the formula (I) of the present invention and a pharmaceutically acceptable salt may be present in a solvated form or in an unsolvated form, for example, a hydrated form or the like.
- the compound represented by the formula (I) of the present invention should follow the design principle of the prodrug, and can release the original active formula by enzymatic hydrolysis, hydrolysis, acid hydrolysis or metabolic degradation under normal physiological conditions in the living body.
- Compound shown by I). This includes, but is not limited to, the lipidation of hydroxyl groups on the compound (such as the formation of phosphates and carbonates), the protection of amino groups and carboxyl groups.
- the compound of the formula (I) or a pharmaceutically acceptable salt or prodrug is prepared into a clinically usable pharmaceutical composition.
- the pharmaceutical preparations thereof include, but are not limited to, oral preparations such as tablets, gels, soft/hard capsules, emulsions, dispersible powders, granules, water/oil suspoemulsions; injections Including intravenous injection, intramuscular injection, intraperitoneal injection, rectal suppository, intracranial injection, these dosage forms may be aqueous solutions or oily solutions; topical preparations include creams, ointments, gels, water/oil solutions and packs Formulations; inhalation dosage forms include fine powders, liquid aerosols, and various dosage forms suitable for in vivo implantation.
- the pharmaceutical composition of the present invention is added to a conventional pharmaceutical excipient as needed.
- excipients should be in accordance with the pharmaceutical preparation process rules and compatible with the active ingredients.
- the solid oral preparation excipients are selected from, but not limited to, mannitol, lactose, starch, magnesium stearate, cellulose, glucose, sucrose, cyclodextrin, and the intestinal absorption molecular carrier vitamin E-PEG1000.
- Oral formulations may incorporate suitable colorants, sweeteners, flavoring agents, and preservatives.
- the compound of the formula (I) of the present invention is administered to a warm-blooded animal at a unit dose of 0.1 to 100 mg/kg.
- a pharmaceutical composition comprising the compound of the formula (I) of the present invention or a pharmaceutically acceptable salt as an active ingredient, mainly for treating a clinical disease associated with EZH2.
- a clinical disease associated with EZH2. include, but are not limited to, cancer, diabetes inflammation, immune system diseases, cardiovascular diseases, neurological diseases, and respiratory diseases.
- cancer includes, but is not limited to, lung cancer, gastric cancer, liver cancer, breast cancer, nasopharyngeal cancer, pancreatic cancer, ovarian cancer, cervical cancer, colorectal cancer, glioma, melanoma, prostate cancer, kidney cancer, esophagus.
- Cancer mesothelioma, head and neck cancer, bladder cancer, salivary gland cancer, anaplastic large cell lymphoma, leukemia, lymphoma, non-Hodgkin's lymphoma, and multiple myeloma.
- composition of the present invention may be used alone or in combination with one or more methods of surgery, radiation therapy, chemotherapy, immunotherapy, fusion tumor virus, RNAi, and cancer adjuvant therapy which are conventionally used clinically.
- Treatment including but not limited to the following anti-tumor drugs and treatments:
- alkylating agents such as cisplatin, cisplatin, oxaliplatin, chlorambucil, carbophosphoramide, nitrogen mustard, melphalan, temozolomide, busulfan, nitrosourea.
- anti-tumor antibiotics such as doxorubicin, bleomycin, doxorubicin, daunorubicin, epirubicin, idarubicin, mitomycin C, actinomycin, genus Anti-mitotic drugs such as vincristine, vinblastine, vindesine, vinorelbine, paclitaxel, taxotere, Polo kinase inhibitors.
- Antimetabolites and antifolates such as fluoropyrimidine, rametine, cytarabine, trebutose, hydroxyurea.
- Topoisomerase inhibitors such as epipodophyllotoxin and camptothecin.
- cytostatic agents such as antiestrogens/antiandrogens such as tamoxifen, fulvestrant, toremifene, raloxifene, ranoxifene, iodinefene; such as bicalutamide , flutamide, nilutamide, cyproterone acetate.
- LHRH antagonists or LHRH agonists such as goserelin, leuprolide, and buserelin, progestogens such as megestrol acetate.
- Aromatase inhibitors such as anastrozole, letrozole, vorozole, exemestane, 5a-reductase inhibitors such as finasteride.
- Anti-invasive agents such as c-Src kinase family inhibitors, metalloproteinase inhibitors, inhibitors of urokinase plasminogen activator receptor function or heparanase-like antibodies.
- inhibitors of growth function such as growth factor antibodies and growth factor receptor antibodies such as anti-HER2 antibody trastuzumab, anti-EGFR antibody panitumumab, anti-EGFR antibody cetuximab, etc.; Including other tyrosine kinase inhibitors and inhibitors of serine/threonine kinases such as Ras/Raf signaling inhibitors, cell signaling inhibitors of MEK and/or AKT kinase, c-kit inhibitors, abl kinase inhibitors , PI3 kinase inhibitors, FLT3 kinase inhibitors, CSF-1R kinase inhibitors, IGF receptor kinase inhibitors, Aurora kinase inhibitors, NTRKA/B/C kinase inhibitors, cyclin-dependent kinase inhibitors such as CDK2 and / or CDK4, a CDK6 inhibitor and a transcriptional kinase CDK7/9/12/13 inhibitor.
- An anti-angiogenic agent such as bevacizumab which inhibits the action of vascular endothelial growth factor and a VEGF receptor tyrosine kinase inhibitor.
- Tumor immunotherapy includes any in vitro and in vivo methods that increase the immunogenicity of a patient's tumor cells.
- cytokine IL-2, IL-4 or GM-CSF for transfection; methods for reducing T cell ineffectiveness such as anti-PD-1/PD-L mAb; transfected immune cells such as cytokine transfected trees Method of squamous cell; method of cytokine transfected tumor cell line; reduction of immunosuppressive cells such as regulatory T cells, myeloid suppressor cells, or dendrites expressing guanamine 2,3-deoxygenase Functional methods of cells; methods of agonists that increase the activity of immune cells, such as STING, and cancer vaccines composed of tumor-associated antigenic proteins or peptides.
- Tumor gene therapy such as CRISPR-Cas 9, RNAi and gene transduction.
- haloalkyl can contain one or more of the same or different halogens.
- the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS). NMR shifts are given in 10 -6 (ppm) units.
- the NMR was measured by a Brukerdps 400 type nuclear magnetic apparatus, and the solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), and the internal standard was tetramethylsilane (TMS).
- DMSO-d 6 dimethyl sulfoxide
- CDCl 3 deuterated chloroform
- TMS tetramethylsilane
- the MS was measured using a 1100 Series LC/MSD Trap (ESI) mass spectrometer (manufacturer: Agilent).
- Thin layer chromatography silica gel plate uses Qingdao Ocean Chemical GF254 silica gel plate.
- the specification of silica gel plate used for thin layer chromatography (TLC) is 0.15mm ⁇ 0.2mm.
- the specification for thin layer chromatography separation and purification is 0.4mm ⁇ 0.5. Mm.
- the reactions can all be carried out under an argon atmosphere or a nitrogen atmosphere.
- An argon atmosphere or a nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon having a volume of about 1 L.
- the solution means an aqueous solution.
- reaction temperature is room temperature and is 20 ° C to 30 ° C.
- the column chromatography eluent system and the thin layer chromatography developer system include: A: dichloromethane and methanol system, B: petroleum ether and ethyl acetate system, the volume ratio of the solvent according to the compound It can be adjusted by changing the polarity. It can also be adjusted by adding a small amount of alkaline or acidic reagent such as ammonia water and acetic acid.
- Step 4 Preparation of tert-butanol 3-(5-bromo-3-ethoxycarbonyl-2-methyl-phenylamino)-pyrrolidin-1-ylcarboxylate (1d)
- Step 5 Preparation of tert-butanol 3-((5-bromo-3-ethoxycarbonyl-2-methyl-phenyl)-ethyl-amino]-pyrrolidin-1-ylcarboxylate (1e)
- Step 6 Preparation of tert-butanol 3-((5-bromo-3-carboxylic acid-2-methyl-phenyl)-ethyl-amino]-pyrrolidin-1-ylcarboxylate (1f)
- Step 7 3-( ⁇ 5-Bromo-3-[(4,6-dimethyl-2-oxo-1,2-dihydro-pyridin-3-ylmethyl)-carbamoyl]-2 Of -methyl-phenyl ⁇ -ethyl-amino)-pyrrolidin-1-ylcarboxylic acid tert-butyl ester (1 g)
- Step 8 3-[(1-Acrylo-pyrrolidin-3-yl)-ethyl-amino]-5-bromo-N-(4,6-dimethyl-2-oxo-1,2- Preparation of dihydro-pyridin-3-ylmethyl)-2methyl-benzamide
- Step 1 Preparation of 4-(5-bromo-3-ethoxyxo-2-methyl-phenyl)-piperidine-1-carboxylic acid tert-butanol ester (8a)
- Step 1 Preparation of tert-butanol 3-((5-bromo-3-ethoxycarbonyl-2-methyl-phenyl)-methyl-amino]-pyrrolidin-1-ylcarboxylate (10a)
- Step 2 3-( ⁇ 5-[2-(4-Methyl-piperazin-1-yl)-pyridin-5-amino]-3-[(4,6-dimethyl-2-oxo- 1,2-Dihydro-pyridin-3-ylmethyl)-carbamoyl]-2-methyl-phenyl ⁇ -ethyl-amino)-pyrrolidin-1-ylcarboxylic acid tert-butanol ester (11b) Preparation
- Step 1 3-( ⁇ 5-(4-Methyl-piperazin-1-yl)-3-[(4,6-dimethyl-2-oxo-1,2-dihydro-pyridine-3) Of tert-butyl alcohol of -methylmethyl)-carbamoyl]-2-methyl-phenyl ⁇ -ethyl-amino)-pyrrolidine-1-ylcarboxylate
- Step 1 5-Bromo-3- ⁇ [1-(4-Dimethylamino-but-2-enoyl)-pyrrolidin-3-yl]-ethyl-amino ⁇ -N-(4,6-di Preparation of methyl-2-oxo-1,2-dihydro-pyridin-3-ylmethyl)-2-methyl-benzamide
- Step 2 Preparation of tert-butanol 3-((5-hydroxy-3-ethoxycarbonyl-2-methyl-phenyl)-ethyl-amino]-pyrrolidin-1-ylcarboxylate (16b)
- the preparation method was the same as that of the steps 3, 4, and 5 of Example 1, except that the compound 16b was replaced with the compound 16a to obtain the compound 16b.
- Step 3 Preparation of tert-butanol 3-[(5-methoxy-3-ethoxycarbonyl-2-methyl-phenyl)-ethyl-amino]-pyrrolidin-1-ylcarboxylate
- Step 2 N-(4,6-Dimethyl-2-oxo-1,2-dihydro-pyridin-3-ylmethyl)-5-[(1-tert-butoxycarbonyl-pyrrolidine-3 -yl)-ethyl-amino]-4'-hydroxymethyl-4-methyl-biphenyl-3-carboxamide
- Step 1 N-(4,6-Dimethyl-2-oxo-1,2-dihydro-pyridin-3-ylmethyl)-5-[(1-acryloyl-pyrrolidin-3-yl) Preparation of -ethyl-amino]-4'-(methanesulfonyloxymethyl)-4-methyl-biphenyl-3-carboxamide (18a)
- Step 2 N-(4,6-Dimethyl-2-oxo-1,2-dihydro-pyridin-3-ylmethyl)-5-[(1-acryloyl-pyrrolidin-3-yl) Preparation of ethyl-amino--4'-(piperidin-1-ylmethyl)-4-methyl-biphenyl-3-carboxamide (18b)
- Example 18 The same procedure as in the production of Example 18 was carried out except that morpholine was used instead of piperidine to obtain Compound 20.
- Step 3 Preparation of 4-(morpholin-4-ylmethyl)-4-bromo-2-chlorobenzene (21c) 4-bromo-2-chlorobenzyl bromide (0.95 g, 3.38 mmol), morpholine ( 0.59 g, 6.67 mmol), potassium carbonate (1.2 g, 8.45 mmol) was added to 20 mL of DMF, and the reaction was stirred at 70 ° C for 1 h. The reaction solution was cooled to room temperature, poured slowly over water and extracted with ethyl acetate. The organic phase was washed twice with aq. The residue was purified by column chromatography (EtOAc:EtOAc:EtOAc
- Step 5 N-(4,6-Dimethyl-2-oxo-1,2-dihydro-pyridin-3-ylmethyl)-5-[(1-acryloyl-pyrrolidin-3-yl) Preparation of ethyl-amino--3'-chloro-4'-(morpholin-4-ylmethyl)-4-methyl-biphenyl-3-carboxamide
- Step 1 3- ⁇ [3-ethoxyxo-2-methyl-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl) Preparation of tert-butanol ester of -phenyl]-ethyl-amino ⁇ -pyrrolidin-1-ylcarboxylate (26a)
- reaction solution was concentrated under reduced pressure and then extracted with water and ethyl acetate.
- organic phase was dried over anhydrous sodium
- residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate 40:1 to 3:1) to yield
- Step 1 2-Methyl-3-[(1-Boc-pyrrolidin-3-yl)-ethyl-amino]-5-[6-(4-methyl-piperazin-1-yl)-pyrimidine Preparation of -4-yl]-benzoic acid ethyl ester (27a)
- Ethyl 2-iodo-4-methylthiophene-3-carboxylate (1.37 g, 5.8 mmol) was dissolved in 20 mL of acetic anhydride, and the mixture was cooled with ice water, and 4 mL of nitric acid was added dropwise and stirred for 2 hours. The reaction mixture was extracted with water and ethyl acetate. The organic layer was washed with EtOAc (EtOAc) EtOAc.
- Step 2 5-[(1-Boc-Pyrrolidin-3-yl)-ethyl-amino]-4-methyl-4'-(morpholin-4-ylformyl)-[1,1'- Preparation of biphenyl]-3-carboxylate
- the compound was prepared in the same manner as in Example 39 except that N-methylpiperazine was used instead of morpholine.
- Example 41 The preparation method of Example 41 was carried out in the same manner except that 2-bromo-4-bromobenzoic acid was used instead of 4-bromobenzoic acid to obtain Compound 44.
- Step 1 Preparation of N-(2-hydroxyethyl)-N-methyl-5-bromo-thiophene [3,2-b] thiophene-2-carboxamide
- Step 1 4- ⁇ [3-Benzyloxy-2-methyl-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl) Preparation of tert-butyl alcohol ester of -phenyl]-ethyl-amino ⁇ -piperidin-1-ylcarboxylate (50a)
- the preparation method is the same as that of the intermediate 26a except that 4-[(3-benzyloxy-2-methyl-5-bromo-phenyl)-ethyl-amino]-piperidin-1-carboxylic acid tert-butyl is used.
- the alcohol ester prepared in the same manner as the intermediate 1e except that benzyl bromide was substituted for ethyl bromide, and 1-Boc-4-piperidone was used instead of 1-Boc-3-pyrrolidone) in place of 1e to give the title compound.
- Step 2 N-(2-Hydroxyethyl)-N-methyl-5- ⁇ 3-[(1-Boc-piperidin-4-yl)-ethyl-amino]-5-(benzyloxy) Preparation of -4-methyl-phenyl ⁇ -thiophene [3,2-b]thiophene-2-carboxamide
- the preparation method is the same as that of the intermediate 39a except that 4- ⁇ [3-benzyloxyyl-2-methyl-5-(4,4,5,5-tetramethyl-[1,3,2]dioxy is used.
- Step 3 N-(2-Hydroxyethyl)-N-methyl-5- ⁇ 3-[(1-Boc-piperidin-4-yl)-ethyl-amino]-5-carboxy-4-methyl Preparation of phenyl-phenyl ⁇ -thiophene [3,2-b]thiophene-2-carboxamide
- the compound 50b (0.35 g, 0.5 mmol) was dissolved in 15 mL of methanol, and 10% Pd-C (30 mg) was added, and the mixture was stirred with hydrogen, and stirred at room temperature for 3 hours, and the filtrate was filtered off. .
- Step 1, 2 Synthesis of ethyl 5-(3-hydroxy-prop-1-ynyl)-2-methyl-3-aminobenzoate (51b)
- Step 3, 4 3-[(1-Boc-pyrrolidin-3-yl)-ethyl-amino]-2-methyl-5-(3-hydroxypropyn-1-yl)-benzoic acid ethyl ester Synthesis of (51d)
- 2,5-Dibromopyridine (1.2 g, 5 mmol) was added to 3 mL of morpholine, and after nitrogen substitution, it was heated at 100 ° C for 4 hours. The reaction solution was cooled to room temperature and then extracted with water ethyl acetate. The ethyl acetate layer was concentrated under reduced pressure and the residue was purifiedjjjjjjjj
- Step 1 3-( ⁇ 4'-carboxy-5-[(4,6-dimethyl-2-oxo-1,2-dihydro-pyridin-3-ylmethyl)-acyl]-4- Preparation of tert-butyl alcohol methyl-biphenyl-3-yl ⁇ -ethyl-amino)-pyrrolidine-1-carboxylate
- Step 1 Preparation of 5-bromo-3-[2-(Boc-methyl-amino)-ethylamino]-2-methyl-benzoic acid ethyl ester
- Step 1 Preparation of 3-(5-bromo-3-ethoxycarbonyl-2-methyl-phenylamino)-pyrrolidin-1-ylcarboxylic acid 9H-indol-9-ylmethyl ester
- Step 2 3-[(5-Bromo-3-ethoxycarbonyl-2-methyl-phenyl)-(2,2,2-trifluoroethyl)-amino]-pyrrolidin-1-ylcarboxylic acid Preparation of 9H-fluoren-9-ylmethyl ester
- Step 3 3-[(5-Bromo-3-ethoxycarbonyl-2-methyl-phenyl)-(2,2,2-trifluoroethyl)-amino]-pyrrolidin-1-ylcarboxylic acid Preparation of tert-butanol ester
- Example 65 N-(4,6-Dimethyl-2-oxo-1,2-dihydro-pyridin-3-ylmethyl)-5-[(1-acryloyl-piperidin-4-yl) Of -ethyl-amino]-4-methyl-4'-(4-methoxypiperidin-1-ylmethyl)-[1,1'-biphenyl]-3-carboxamide
- Example 70 N-(4,6-Dimethyl-2-oxo-1,2-dihydro-pyridin-3-ylmethyl)-5-[(1-acryloyl-piperidin-4-yl) Of -ethyl-amino]-4-methyl-3'-(morpholin-4-ylmethyl)-[1,1'-biphenyl]-3-carboxamide
- Example 70 In the same manner as in the preparation of Example 70, except that the morpholine was replaced with 4-oxopipidine, the compound 83 was obtained.
- Example 34 The procedure of the preparation of Example 34 was carried out except that 3-isopropyloxy-1-azetidine was used instead of piperidine to obtain Compound 85.
- the preparation method was the same as in Example 21 except that 1-Boc-4-piperidone was used instead of 1-Boc-3-pyrrolidone, and 4-(morpholin-4-yl)-piperidine was used instead of morpholine to prepare a compound. 86.
- Test Example 1 Determination of 50% inhibition (GI50) concentration of in vitro growth of EZH2 wild type and mutant expression positive tumor cells by the compound of the present invention
- Tumor cell lines are an effective cell model for studying tumor growth inhibition in vitro, and a representative tumor cell line is selected for the determination of compound activity. Cell culture conditions and methods were performed as required for each cell line. No more than 3 passages per in vitro culture. Cell lines can be subjected to monoclonal purification and identification as needed.
- Cell culture medium was supplemented with 5-20% fetal bovine serum (Gibco), 1% double antibody and 2 mM glutamine using RPMI 1640 (Gibco).
- Human B cell non-Hodgkin's lymphoma Karpas 422 (ATCC) expressed EZH2 Y641N mutein and was cultured in suspension with RPMI 1640 plus 10% FBS and 1% double antibody and 2 mM glutamine.
- Human diffuse large cell lymphoma B lymphocytes Pfeiffer (ATCC) expressed EZH2 A677G mutein and was cultured in suspension with RPMI 1640 plus 10% FBS and 1% double antibody and 2 mM glutamine.
- Human B cell lymphoma SU-DHL-8 (ATCC) expressed EZH2 wild type and was cultured in suspension with RPMI 1640 plus 10% FBS and 1% double antibody and 2 mM glutamine.
- the suspension cultured cells were directly centrifuged (1700 rpm, 3 minutes), the supernatant was discarded, and the cells were counted.
- Different cell concentrations (1-10 x 10 4 cells per ml) were prepared according to each cell growth cycle, inoculated into 96-well plates (Corning), 200 ⁇ l per well, and cultured overnight at 37 ° C, 5% CO 2 . The next day, the test compound was added to the cultured cells in 2 wells.
- the final concentration of the organic solvent is not more than one thousandth, and the cells are further cultured for 6-12 days, and the MTT is determined.
- the compound of the present invention and the control compound EPZ-6438 were dissolved in DMSO, and the purity of the compound was 98% or more.
- the compound was stored at a concentration of 10 mM, stored at -20 ° C, and serially diluted 10 or 10 times before use.
- control compound EPZ-6438 was synthesized according to the method of US2012264734, and its structural formula is as follows.
- the MTT detection reagent is the Dojindo CCK8 kit, and the enzyme labeling instrument is THERMOMULTISKAN FC instrument.
- Suspension cells can be directly added to CCK8 reagent at a final concentration of 10%, and culture is continued for 1-4 hours.
- the solvent control wells appear dark yellow, the OD450nm light absorption value is measured, and the cell growth rate is calculated according to the following formula.
- T drug treatment cell hole optical density value - blank control hole optical density value
- T 0 cell hole optical density value before drug treatment - blank control hole optical density value
- C solvent control cell hole optical density - blank control hole light Density value.
- the drug concentration which is 50% inhibition of cell growth, is calculated as the GI50 by the drug concentration and cell growth rate curve. The experiment was repeated 3 times and the data was subjected to biological statistical analysis.
- Table 2 summarizes the results of in vitro growth inhibition (or induction of apoptosis) GI50 concentrations of some representative compounds of the compounds of the invention against EZH2 wild type and different mutant expression positive tumor cells.
- Test Example 2 Determination of in vivo growth inhibitory activity of the compound of the present invention against Pfeiffer of EZH2 mutant A677G expression-positive tumor cells
- NOD/SCID mice were subcutaneously inoculated with human diffuse large cell lymphoma B lymphocytes Pfeiffer (ATCC) expressing EZH2A677G mutant protein, and a human B cell lymphoma subcutaneous transplantation tumor model was established.
- mice NOD/SCID mice, female, 9 weeks (week age at the time of tumor cell inoculation), average body weight 20.3 g, 18 rats. Purchased from Shanghai Lingchang Biotechnology Co., Ltd., animal certificate number: 2002007656.
- the breeding environment is SPF.
- Test sample Inventive compound 34 (purity: 99.03%) and compound 35 (purity: 97.6%), solid powder, stored at -20 ° C.
- Pfeiffer cells were cultured in RPMI 1640 medium containing 10% fetal bovine serum. Pfeiffer cells in the exponential growth phase were collected and resuspended in PBS to 1 ⁇ 10 8 /ml cells for subcutaneous inoculation of NOD/SCID mice.
- mice in the day before cell inoculation were treated with Co60 irradiation at a dose of 200 rad, then 1 ⁇ 10 7 Pfeiffer cells were inoculated subcutaneously on the right side, and the cells were resuspended in 1:1 PBS and Matrigel (0.1 ml/mouse). .
- the tumor volume calculation formula is: long diameter ⁇ short diameter 2 /2.
- the day of tumor cell inoculation was defined as day 0.
- the test was divided into a vehicle control group and a drug-administered group (Compound 34 group and Compound 35 group), 6 rats in each group, orally administered once a day for 4 weeks.
- the efficacy was evaluated based on the relative tumor inhibition rate (TGI), and the safety was evaluated based on the animal's body weight change and death.
- TGI relative tumor inhibition rate
- Preparation of the test sample Weigh the compound 34 or 35, first add ultrapure water to mix, then add methanesulfonic acid to adjust, then the pH value is about 1.64, then add 4M NaOH to adjust the pH to 3.5, ultra-pure water to volume It is 20 mg/ml.
- T/C% is the relative tumor growth rate, that is, the percentage of tumor volume or tumor weight relative to the treatment group and the control group at a certain time point.
- T and C are the relative tumor volumes (RTV) of the treatment group and the control group at a specific time point, respectively.
- T/C% T RTV /C RTV *100%
- All test results are expressed as mean tumor volume ⁇ SEM (mean standard error). Tumor volume data 27 days after the start of drug administration were selected for statistical analysis between different groups. The independent sample T test method was used to compare the relative tumor volume of the treatment group with the control group. All data were analyzed using SPSS 18.0. p ⁇ 0.05 was considered to be a significant difference.
- Test compound 34 and compound 35 at the 200 mg/kg dose remained stable during the administration of the mice, and were well tolerated by the test drug.
- Test Example 3 Irreversible inhibition of in vitro growth of Pfeiffer of EZH2 mutant A677G expression-positive tumor cells by the compound of the present invention
- Diffuse large cell lymphoma B lymphocytes Pfeiffer (ATCC), were cultured in suspension with complete medium (RPMI 1640 plus 10% FBS and 1% double antibody and 2 mM glutamine). The Pfeiffer cells growing in log phase were collected by centrifugation (1700 rpm, 3 minutes), the supernatant was discarded, and the cells were counted. The concentration of 2 x 10 4 cells per ml was prepared in complete medium, inoculated into 96-well plates (Corning), 200 ⁇ l per well, and cultured overnight at 37 ° C, 5% CO 2 .
- test compound 34 different concentrations of the test compound (Compound 34) were added to the cultured cells in parallel with 4 wells, and the final concentration of the organic solvent DMSO was not more than one thousandth.
- the cells were centrifuged to collect the cells (2 wells in parallel), washed three times with serum-free cell culture medium, and resuspended in an equal volume (200 ⁇ l) of complete cell culture medium with no treatment holes.
- the cells were further cultured for 10 days at 37 ° C, 5% CO 2 .
- the MTT test was carried out in accordance with the method of Test Example 1. The test was repeated once, and the specific results are shown in Table 6.
- control product EPZ6438 was prepared in the same manner as in Test Example 1.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Epidemiology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Pyridine Compounds (AREA)
Abstract
本发明涉及一种组蛋白甲基转移酶EZH2抑制剂、其制备方法及其医药用途。特别地,本发明涉及通式(I)所示的化合物,其制备方法及含有其的药物组合物,以及其作为组蛋白甲基转移酶EZH2抑制剂,治疗与组蛋白甲基转移酶EZH2相关的疾病特别是癌症的用途。其中通式(I)中的各取代基的定义与说明书中的定义相同。
Description
本发明涉及一种新型的组蛋白甲基转移酶EZH2抑制剂、其制备方法及含有其的药物组合物,以及其作为组蛋白甲基转移酶EZH2抑制剂的用途,特别是在预防和/或治疗人类疾病包括癌症中的用途。
肿瘤包括白血病是导致人类临床死亡的重大疾病之一。恶性肿瘤如肺癌、胃癌、乳腺癌、胰腺癌、肝癌、肠癌、卵巢癌、宫颈癌、食管癌、鼻咽癌、白血病和恶性淋巴瘤等死亡率极高。尽管癌症的遗传筛查、分子诊断和精准医疗提高了癌症病人的临床早期发现/正确诊断和治疗效果,但至今为止多数癌症特别是晚期、难治、复发和耐药性恶性肿瘤仍无有效的方法或药物可完全根除或治愈。临床上急需特异性好、活性高、毒性小、无耐药性产生的优质抗癌药物。
癌症的发生、发展、转移、恶化、复发及耐药性产生与许多因素有关,其中正常细胞内分子信号传导途径的异常是导致细胞转化与癌化的重要因素之一。遗传学(基因序列改变所致基因表达水平变化,如基因突变、基因杂合丢失和微卫星不稳定等)和表观遗传学(非基因序列改变所致基因表达水平变化,如DNA甲基化和染色质构象变化等)是研究分子信号传导的两大重要方法学。基因序列的改变常常可直接导致不同类型的人类疾病包括癌症。经过几十年的努力,借助下一代基因测序技术和基因组大数据,人们已发现和鉴定出诸多与人类疾病相关的遗传变异基因可作为临床诊断及药物开发的生物标记靶点,同时许多作用于这些靶点的药物在临床疾病治疗中呈现良好的效果。
表观遗传学研究是不涉及DNA序列改变的基因或者蛋白质表达的变化,并可以在发育和细胞增殖过程中稳定传递的遗传学分支学科,主要包括DNA甲基化,组蛋白共价修饰,染色质重塑,基因沉默和RNA编辑等调控机制。在真核细胞中,DNA与组蛋白一起包装以形成染色质,染色质有序结构的变化可导致相关基因转录的改变。这个过程被高度精准控制,因为基因表达模式的变化影响细胞的正常生理功能,如分化、增殖和凋亡。其中组蛋白N-末端尾的共价修饰可介导染色质结构的改变,导致基因表达的可遗传变化,但不影响DNA本身的序列。酶促介导氨基酸侧链的共价修饰(例如甲基化、乙酰化、磷酸化和泛素化)是基因表达调控的重要功能之一。组蛋白甲基转移酶(HMT)控制甲基向组蛋白上的特定氨基酸位点的选择性添加。特定基因的表达水平受相关组蛋白位点处一个或多个甲基的存在或不存在的影响。甲基在特定组蛋白位点的特异性效应持续直到甲基被组蛋白去甲基化酶去除,或直到经修饰的组蛋白通过核小体周转被替换以此进行基因表达的控制。组蛋白甲基转移酶的异常表达和/或活性破坏时,导致疾病状态。 例如,在人类癌症中,失调的表观遗传酶活性促进癌症的发生和发展以及其它癌症相关表征(如细胞迁移和侵袭等)。除了癌症,越来越多的证据表明表观遗传酶在许多其它人类疾病中同样发挥作用,包括代谢性疾病(例如糖尿病)、炎性疾病(例如克罗恩病)、神经变性疾病(例如阿尔茨海默氏病)和心血管疾病。因此靶向表观遗传变异的临床诊断及药物开发已是当今基础研究和制药工业的热点。其中,有些药物已成功应用于临床如组蛋白去乙酰化酶(Histone Deacetylase,HDAC)抑制剂。
PRC2(Polycomb Repressive Complex 2,即多梳抑制复合物2)是两类多梳蛋白或(PcG)中的一种。该复合物具有组蛋白甲基转移酶活性,主要是赖氨酸27(即H3K27me3)上的组蛋白H3三甲基化,这种甲基化作为一种转录沉默染色质的标记。PRC2具有四个亚基:Suz12(锌指)、Eed、Ezh1或Ezh2(具有组蛋白甲基转移酶活性的SET结构域)和RbAp48(组蛋白结合结构域),其中EZH2(Zeste Homolog 2的增强子)属于催化亚基。EZH2的催化位点存在于SET结构域内,除了H3-K79甲基转移酶DOT1,SET结构域是所有已知组蛋白赖氨酸甲基转移酶的特征。研究发现EZH2的Y641(Y641F、Y641N、Y641C、Y641S和Y641H)、A677G和A687V的突变与弥漫性大B细胞淋巴瘤(DLBCL)的滤泡淋巴瘤(FL)和生发中心B细胞样(GCB)亚型相关(Morin等人(2010)Nat Genet 42:181-5;McCabe MT等人(2012)Proc Natl Acad Sci U S A.109(8):2989-94;Ott HM等人(2014)Mol Cancer Ther.13(12):3062-73;Majer CR等人(2012)FEBS Lett.586(19):3448-51),并且抑制EZH2可与许多癌细胞突变基因如ARID1A、KDM6A和SMARCA2/4等呈现协同致死效应(Bitler BG等人(2015)NatMed 21:231-8)。
一般来讲突变体EZH2基因是以杂合子出现,所有突变体都可以并入多蛋白PRC2复合物中,但复合物缺乏催化H3-K27甲基化的能力。滤泡性淋巴瘤是最常见的非霍奇金淋巴瘤之一,对免疫化学疗法可敏感应答,但许多患者极易复发,临床上的反复复发最终导致对标准治疗的不应答,约30%的患者转变为侵袭性淋巴瘤,即弥漫性大B细胞淋巴瘤(DLBCL)。在滤泡淋巴瘤中,表观基因组突变主要靶向组蛋白甲基转移酶KMT2D(90%)和EZH2(25%),以及组蛋白乙酰转移酶CREBBP(30-60%)和EP300(9%)。尽管抗CD20单克隆抗体(利妥昔单抗)治疗已经显着改善患者疾病,但DLBCL仍然是一种不可治愈的癌症,其中化学性耐受性或转化性疾病的患者仍然不能得到有效临床的治疗。近来研究发现EZH2抑制剂如EPZ-6438,能够有效地体内体外抑制具有EZH2突变的非霍奇金淋巴瘤细胞生长,并已开展其临床试验(Knutson SK等人(2012)Nat Chem Biol.8(11):890-6;McCabe MT等人(2012)Nature.492(7427):108-12;Knutson SK等人(2014)Mol Cancer Ther.13(4):842-54)。因此表观基因组蛋白修饰突变酶抑制剂将为弥漫性大B细胞淋巴瘤包括滤泡性淋巴瘤的临床治疗带来新的希望。
然而,现有EZH2抑制剂多为与EZH1/2的SET(Su(var)3-9,Enhancer-of-zeste, Trithorax)功能域结合,以下是EZH1/2SET功能域氨基酸序列的比较。
由序列的比较可以看出EZH1/2SET功能域氨基酸序列高度保守,95%的EZH1/2SET氨基酸序列相同,即122个氨基酸中116个氨基酸相同。因此,多数EZH2抑制剂不但能够抑制EZH2,同时也可抑制EZH1。
本发明人在长期表观基因组蛋白修饰突变酶抑制剂的研究与开发过程中,根据EZH2蛋白质序列、晶型结构以及已知EZH2抑制剂结合位点和作用方式(可逆),意外发现在EZH2氨基酸SET序列中存在一个独有半胱氨酸Cys668(NP_004447,histone-lysine N-methyltransferase EZH2isoform a),而EZH1相应位点为Ser664(NP_001982,histone-lysine N-methyltransferase EZH1isoform 1)。根据这一特征,本发明化合物可与EZH2形成不可逆共价结合,有效地抑制EZH2激活突变基因(如EZH2A677G和Y641N)表达的癌细胞生长,诱导其细胞凋亡。因此,本发明化合物为EZH2抑制剂的开发提供了一种新的作用机制和方向。
发明内容
本发明的目的是提供一种特异性好、活性高、毒性低的小分子化合物,其可以作为组蛋白甲基转移酶EZH2高选择性不可逆共价结合抑制剂,用于预防和/或治疗人类疾病包括癌症中的用途。
本发明一方面提供一种通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,
其中:
Ar选自5元或6元的芳基或杂芳基、及由5元或6元芳基或杂芳基形成的稠环,其中Ar任选进一步被一个或多个-Q-T基团取代;
L选自C
2-C
8饱和或不饱和的直链或支链烃链或环状结构,L中任意氢原子可被卤素、氰基、羟基、C
1-C
6烷氧基替换,L中任意碳原子可被N、O、S替换;L任选进一步被一个或多个-Q-T基团取代;
R
1、R
2各自独立地选自氢、C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷 基、4-12元杂环基,所述烷基、烯基、炔基、环烷基和杂环基任选进一步被一个或多个-Q-T基团取代;
或者,R
1可与L的任何位置连接,连同他们所连接的N原子形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q-T基团取代;
或者,R
2可与L的任何位置连接,连同它们所连接的N原子形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q-T基团取代;
或者,R
1、R
2、L及它们连接的两个N原子形成4-12元杂环基,所述杂环任选进一步被一个或多个-Q-T基团取代;
R
3选自氢、氟、R
a;
R
4、R
5各自独立地选自氢、R
a;
R
5’选自卤素、-OS(O)
2-C
1-C
6烷基或-OS(O)
2-C
3-C
6环烷基;
R
a选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基;所述烷基、烯基、炔基、环烷基、和杂环基任选进一步被一个或多个-Q-T基团取代;
R
6、R
12各自独立地选自氢、C
1-C
6烷基、C
3-C
6环烷基;
R
7、R
9各自独立地选自氢、卤素、R
b;
R
b选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基,所述烷基、烯基、炔基、环烷基、和杂环基任选进一步被选自卤素、氰基、羟基、C
1-C
6烷氧基、C
1-C
6烷基-OC(O)-、氨基、C
1-C
6烷基氨基、双C
1-C
6烷基氨基、4-12元杂环基的一种或多种基团取代;
R
8、R
10、R
11各自独立地选自氢、卤素、C
1-C
3烷基或环烷基;
Q选自一个键或C
1-C
6亚烷基,所述亚烷基任选进一步被选自卤素、氰基、羟基、C
1-C
6烷氧基的一种或多种基团取代;
T选自氢、卤素、氰基、羟基、-NR
hR
i、-C(O)R
h、OR
h、-C(O)O-R
h、C(O)NR
hR
i、-NR
hC(O)R
i、-NR
jC(O)NR
hR
i、-NR
hC(O)OR
i、或R
k;
或者-Q-T是氧代;
R
k选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5至10元杂芳基、芳基,R
k任选进一步被一个或多个-Q
1-T
1基团取代;
R
h、R
i、R
j各自独立地选自氢或R
l,R
l选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、和芳基,R
l任选进一步被一个或多个-Q
1-T
1基团取代;
或者R
h、R
i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q
1-T
1基团取代;
Q
1为一个键或C
1-C
6亚烷基,所述亚烷基任选进一步被选自卤素、氰基、羟基、C
1-C
6烷氧基的一种或多种基团取代;
T
1选自氢、卤素、氰基、羟基、-NR
mR
n、-C(O)R
m、OR
m、-C(O)O-R
m、C(O)NR
mR
n、 -NR
mC(O)R
n、-NR
oC(O)NR
mR
n、-NR
mC(O)OR
n、-O(CH
2)
aNR
mR
n、-(CH
2)
aNR
mR
n、-S(O)
2NR
mR
n或R
p;
或者-Q
1-T
1是氧代;
R
p选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基;
R
m、R
n、R
o各自独立地选自氢或R
q,R
q选自、C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R
q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C
1-C
6烷基、羟基、氧代的一种或多种基团取代;
或者R
m、R
n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C
1-C
6烷基、-OR
x、-NR
xR
y、-C(O)R
x、-O(CH
2)
aOR
x的一种或多种基团取代;
R
x和R
y各自独立地选自氢或R
z,R
z选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R
q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C
1-C
6烷基、羟基、氧代的一种或多种基团取代;
或者R
x、R
y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;
a为1至4的整数。
在本发明一个优选的实施方案中,根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,
其中:
Ar选自
X选自CR
14;
Y选自N或CR
15;
Z选自N或CR
16;
R
13选自氢、卤素、C
1-C
6烷基或C
3-C
6环烷基;
R
14、R
15、R
16各自独立地选自-Q-T基团;
Q选自一个键或C
1-C
6亚烷基;
T选自氢、卤素、氰基、羟基、-NR
hR
i、-C(O)R
h、OR
h、-C(O)O-R
h、C(O)NR
hR
i、-NR
hC(O)R
i、-NR
jC(O)NR
hR
i、-NR
hC(O)OR
i、或R
k;
R
k选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5至10元杂芳基、芳基,R
k任选进一步被一个或多个-Q
1-T
1基团取代;
R
h、R
i、R
j各自独立地选自氢或R
l,R
l选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、和芳基,R
l任选进一步被一个或多个-Q
1-T
1基团取代;
或者R
h、R
i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q
1-T
1基团取代;
Q
1为一个键或C
1-C
6亚烷基;
T
1选自氢、卤素、氰基、羟基、-NR
mR
n、-C(O)R
m、OR
m、-C(O)O-R
m、C(O)NR
mR
n、-NR
mC(O)R
n、-NR
oC(O)NR
mR
n、-NR
mC(O)OR
n、-O(CH
2)
aNR
mR
n、-(CH
2)
aNR
mR
n、-S(O)
2NR
mR
n或R
p;
R
p选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基;
R
m、R
n、R
o各自独立地选自氢或R
q,R
q选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R
q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C
1-C
6烷基、羟基、氧代的一种或多种基团取代,
或者R
m、R
n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C
1-C
6烷基、-OR
x、-NR
xR
y、-C(O)R
x、-O(CH
2)
aOR
x的一种或多种基团取代;
R
x和R
y各自独立地选自氢或R
z,R
z选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R
q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C
1-C
6烷基、羟基、氧代的一种或多种基团取代;
或者R
x、R
y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;
a为1至4的整数。
在本发明另一个优选的实施方案中,根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,
其中:
R
13选自氢、卤素、和C
1-C
6烷基;
X选自CR
14;
Y选自CR
15;
Z选自CR
16;
R
14选自-Q-T基团;
R
15、R
16各自独立地选自氢或卤素;
Q选自一个键或C
1-C
6亚烷基;
T选自氢、卤素、氰基、羟基、-NR
hR
i、-C(O)R
h、OR
h、-C(O)O-R
h、C(O)NR
hR
i、-NR
hC(O)R
i、-NR
jC(O)NR
hR
i、-NR
hC(O)OR
i、或R
k;
R
k选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5至10元杂芳基、芳基,所述杂环基、杂芳基或芳基任选进一步被一个或多个-Q
1-T
1基团取代;
R
h、R
i、R
j各自独立地选自氢或R
l,R
l选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、和芳基,R
l任选进一步被一个或多个-Q
1-T
1基团取代;
或者R
h、R
i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q
1-T
1基团取代;
Q
1为一个键或C
1-C
6亚烷基;
T
1选自氢、卤素、氰基、羟基、-NR
mR
n、-C(O)R
m、OR
m、-C(O)O-R
m、C(O)NR
mR
n、-NR
mC(O)R
n、-NR
oC(O)NR
mR
n、-NR
mC(O)OR
n、-O(CH
2)
aNR
mR
n、-(CH
2)
aNR
mR
n、-S(O)
2NR
mR
n或R
p;
R
p选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基;
R
m、R
n、R
o各自独立地选自氢或R
q,R
q选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R
q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C
1-C
6烷基、羟基、氧代的一种或多种基团取代,
或者R
m、R
n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C
1-C
6烷基、-OR
x、-NR
xR
y、-C(O)R
x、-O(CH
2)
aOR
x的一种或多种基团取代;
R
x和R
y各自独立地选自氢或R
z,R
z选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R
q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C
1-C
6烷基、羟基、氧代的一种或多种基团取代;
或者R
x、R
y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;
a为1至4的整数。
在本发明另一个优选的实施方案中,根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中:
Q选自一个键或C
1-C
6亚烷基;
T选自氢、卤素、-NR
hR
i、OR
h或R
k;
R
k选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、4-12元杂环基、5至10元杂芳基、C
6-C
10芳基,所述烷基、烯基、炔基、杂环基、杂芳基或芳基任选进一步被一个或多个-Q
1-T
1基团取代;
R
h、R
i各自独立地选自氢或R
l,R
l选自C
1-C
6烷基、5元或6元杂芳基、和C
6-C
10芳基,R
l任选进一步被一个或多个-Q
1-T
1基团取代;
或者R
h、R
i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q
1-T
1基团取代;
Q
1为一个键或C
1-C
6亚烷基;
T
1选自氢、卤素、羟基、-NR
mR
n、OR
m、-C(O)OR
m、-C(O)NR
mR
n、-O(CH
2)
aNR
mR
n、-(CH
2)
aNR
mR
n、-S(O)
2NR
mR
n或R
p;
R
p选自C
1-C
6烷基、4-12元杂环基;
R
m、R
n各自独立地选自氢、C
1-C
6烷基,所述烷基任选进一步被选自羟基或5元或6元杂芳基的一种或多种基团取代,其中所述杂芳基任选进一步被选自C
1-C
6烷基、氧代的一种或多种基团取代;
或者R
m、R
n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步选自卤素、羟基、氧代、C
1-C
6烷基、-OR
x、-NR
xR
y、-C(O)R
x、-O(CH
2)
aOR
x的一种或多种基团取代;
R
x和R
y各自独立地选自氢或R
z,R
z选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R
q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C
1-C
6烷基、羟基、氧代的一种或多种基团取代;
或者R
x、R
y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;
a为1至4的整数。
在本发明另一个优选的实施方案中,根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中:
Q选自一个键;
T选自氢、卤素、-NR
hR
i、OR
h或R
k;
R
k选自C
1-C
6烷基、C
2-C
6炔基、或苯基、嘧啶基、吡啶基、吡唑基、咪唑基、噻唑基、噁唑基或噻吩[3,2-b]噻吩基团,所述烷基、炔基、苯基、嘧啶基、吡啶基、、吡唑基、咪唑基、噻唑基、噁唑基或噻吩[3,2-b]噻吩基团任选进一步被一个或多个-Q
1-T
1基团取代;
R
h、R
i各自独立地选自氢、吡啶基、嘧啶基和苯基,所述吡啶基、嘧啶基或苯基任选进一步被一个或多个-Q
1-T
1基团取代;
或者R
h、R
i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q
1-T
1基团取代;
Q
1为一个键或C
1-C
6亚烷基;
T
1选自氢、卤素、羟基、-NR
mR
n、OR
m、-C(O)OR
m、-C(O)NR
mR
n、-O(CH
2)
aNR
mR
n、-(CH
2)
aNR
mR
n、-S(O)
2NR
mR
n或R
p;
R
p选自C
1-C
6烷基;
R
m、R
n各自独立地选自氢、C
1-C
6烷基,,所述烷基任选进一步被选自羟基或5元或6元杂芳基的一种或多种基团取代,其中所述杂芳基任选进一步被选自C
1-C
6烷基、氧代的一种或多种基团取代;
或者R
m、R
n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C
1-C
6烷基、-OR
x、-NR
xR
y、-C(O)R
x、-O(CH
2)
aOR
x的一种或多种基团取代;
R
x和R
y各自独立地选自氢或R
z,R
z选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R
q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C
1-C
6烷基、羟基、氧代的一种或多种基团取代;
或者R
x、R
y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;
a为1至4的整数。
在本发明另一个优选的实施方案中,根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中:
Q选自一个键;
T选自氢、卤素、-NR
hR
i、OR
h或R
k;
R
k选自C
1-C
6烷基、C
2-C
6炔基、或苯基、嘧啶基、吡啶基、吡唑基、咪唑基、噻唑基、噁唑基或噻吩[3,2-b]噻吩基团,所述烷基、炔基、苯基、嘧啶基、吡啶基、、吡唑基、咪唑基、噻唑基、噁唑基或噻吩[3,2-b]噻吩基团任选进一步被一个或多个-Q
1-T
1基团取代;
R
h、R
i各自独立地选自氢、吡啶基、嘧啶基、和苯基,所述吡啶基、嘧啶基或苯基任选进一步被一个或多个-Q
1-T
1基团取代;
或者R
h、R
i与它们相连的N原子一起形成哌啶基、哌嗪基、吡咯烷基、吗啉基,所述哌啶基、哌嗪基、吡咯烷基、吗啉基任选进一步被一个或多个-Q
1-T
1基团取代;
Q
1为一个键或C
1-C
6亚烷基;
T
1选自氢、卤素、羟基、-NR
mR
n、OR
m、-C(O)OR
m、-C(O)NR
mR
n、-O(CH
2)
aNR
mR
n、-(CH
2)
aNR
mR
n、-S(O)
2NR
mR
n或R
p;
R
p选自C
1-C
6烷基;
R
m、R
n各自独立地选自氢、C
1-C
6烷基,所述烷基任选进一步被选自羟基或5元或6元杂芳基的一种或多种基团取代,其中所述杂芳基任选进一步被选自C
1-C
6 烷基、氧代的一种或多种基团取代;
或者R
m、R
n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C
1-C
6烷基、-OR
x、C
1-C
6烷氧基、-NR
xR
y、-C(O)R
x、-O(CH
2)
aOR
x的一种或多种基团取代;
R
x和R
y各自独立地选自氢或R
z,R
z选自C
1-C
6烷基、C
2-C
6烯基、C
2-C
6炔基、C
3-C
6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R
q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C
1-C
6烷基、羟基、氧代的一种或多种基团取代;
或者R
x、R
y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;
a为1至4的整数。
在本发明另一个优选的实施方案中,根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中:
Q选自一个键;
T选自氢、卤素、-NR
hR
i、OR
h或R
k;
R
k选自C
1-C
6烷基、C
2-C
6炔基、或苯基、嘧啶基、吡啶基、或噻吩[3,2-b]噻吩基团,所述烷基、炔基、苯基、嘧啶基、吡啶基、或噻吩[3,2-b]噻吩基团任选进一步被一个或多个-Q
1-T
1基团取代;
R
h、R
i各自独立地选自氢、吡啶基、嘧啶基、和苯基,所述吡啶基、嘧啶基或苯基任选进一步被一个或多个-Q
1-T
1基团取代;
或者R
h、R
i与它们相连的N原子一起形成哌啶基、哌嗪基、吗啉基,所述哌啶基、哌嗪基、吗啉基任选进一步被一个或多个-Q
1-T
1基团取代;
Q
1为一个键或C
1-C
6亚烷基;
T
1选自氢、卤素、羟基、-NR
mR
n、OR
m、-C(O)OR
m、-C(O)NR
mR
n、-O(CH
2)
aNR
mR
n、-(CH
2)
aNR
mR
n、-S(O)
2NR
mR
n或R
p;
R
p选自C
1-C
6烷基;
R
m、R
n各自独立地选自氢、C
1-C
6烷基,所述烷基任选进一步被选自羟基或吡啶基、嘧啶基的一种或多种基团取代,其中所述吡啶基或嘧啶基任选进一步被选自C
1-C
6烷基、氧代的一种或多种基团取代;
或者R
m、R
n与它们相连的N原子一起形成哌啶基、吗啉基、吡咯烷基、哌嗪基、高哌嗪基、氮杂环丁烷基、氮杂螺环,所述哌啶基、吗啉基、吡咯烷基、哌嗪基、高哌嗪基、氮杂环丁烷基、氮杂螺环任选进一步被选自卤素、氧代、C
1-C
6烷基、-OR
x、-NR
xR
y、-C(O)R
x、-O(CH
2)
aOR
x的一种或多种基团取代;
R
x和R
y各自独立地选自氢或R
z,R
z选自C
1-C
6烷基、C
3-C
6环烷基,R
q任选进一步被选自卤素、羟基的一种或多种基团取代;
或者R
x、R
y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元 杂环基;优选哌啶基、吗啉基、吡咯烷基、哌嗪基、高哌嗪基;
a为1至4的整数。
在本发明另一个优选的实施方案中,根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其为以下通式(IIA)、通式(IIB)、通式(IIC)、通式(IID)、通式(IIE)或通式(IIF)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,
其中,R
1~R
12和Ar如上通式(I)中所定义。
在本发明另一个优选的实施方案中,根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其为通式(IIA)、通式(IIB)、通式(IIC)、通式(IID)、通式(IIE)或通式(IIF)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中,
R
1、R
2选自氢、C
1-C
6烷基、C
3-C
6环烷基、4-12元杂环基;
R
3、R
4选自氢;
R
5选自氢、C
1-C
6烷基,所述烷基任选进一步被一个或多个-Q-T基团取代;
R
6、R
12选自氢;
R
7、R
9选自氢、C
1-C
6烷基;
R
8、R
10、R
11选自氢;
-Q-T为-NR
hR
i基团;
R
h、R
i各自独立地选自氢和C
1-C
6烷基;
或者R
h、R
i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;优选哌啶基、哌嗪基、吗啉基、吡咯烷基;
Ar如通式(IIA)、通式(IIB)、通式(IIC)、通式(IID)、通式(IIE)或通式(IIF)中所定义。
在本发明另一个优选的实施方案中,根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其为通式(IIA)、通式(IIB)、通式(IIC)、通式(IID)、通式(IIE)或通式(IIF)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中,
Ar为苯基,所述苯基任选进一步被一个或多个-Q-T基团取代;
R
1、R
2选自氢、C
1-C
6烷基、C
3-C
6环烷基、4-12元杂环基;
R
3、R
4选自氢;
R
5选自氢、C
1-C
6烷基,所述烷基任选进一步被一个或多个-Q-T基团取代;
R
6、R
12选自氢;
R
7、R
9选自氢、C
1-C
6烷基;
R
8、R
10、R
11选自氢;
-Q-T为-苯基,所述苯基任选进一步被一个或多个-Q
1-T
1基团取代
Q
1为一个键或C
1-C
6亚烷基,
T
1选自氢、卤素、羟基、-NR
mR
n、OR
m、-C(O)OR
m、-C(O)NR
mR
n、-O(CH
2)
aNR
mR
n、-S(O)
2NR
mR
n;
或者-Q
1-T
1是氧代;
R
m、R
n各自独立地选自氢或R
q,R
q选自C
1-C
6烷基,R
q任选进一步被选自卤素、羟基、5元或6元杂芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C
1-C
6烷基、羟基、氧代的一种或多种基团取代;
或者R
m、R
n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,优选哌啶基、哌嗪基、高哌嗪基、吗啉基、吡咯烷基、氮杂环丁烷基、氮杂螺环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C
1-C
6烷基、-OR
x、-NR
xR
y、-C(O)R
x、-O(CH
2)
aOR
x的一种或多种基团取代;
R
x和R
y各自独立地选自氢或R
z,R
z选自C
1-C
6烷基、C
3-C
6环烷基,;
或者R
x、R
y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;优选哌啶基、哌嗪基、吗啉基、吡咯烷基;
a为1至4的整数。
本发明典型的化合物包括但不限于下表1中的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药。
表1
本发明另一方面涉及根据本发明所述的通式(I)所示的化合物的氘代化合物,其中通式(I)化合物中的一个或多个H原子独立地被D原子替代,其用于增加在体内的代谢稳定性,所述化合物包括但不限于:
本发明另一方面涉及根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的制备方法,其包括以下步骤:
步骤1)化合物(Ia)与化合物(Ib)在缩合剂如HATU的存在下发生缩合反应,制得化合物(Ic);
步骤2)化合物(Ic)在酸性条件下发生水解反应,制得化合物(Id);
步骤3)化合物(Id)与相应的R反应,制得通式(I)化合物;
其中,R与氨基的反应可以是R的羧酸形式与胺基缩合剂如HATU的存在下发生缩合反应,或者是R的酰氯、酸酐或混合酸酐的形式与氨基通过胺解反应完成连接。
本发明进一步涉及一种药物组合物,其包含根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的氘代化合物,以及一种或多种药学上可接受的载体。
本发明进一步涉及根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的氘代化合物,或含有其的药物组合物,在制备 EZH2组蛋白甲基转移酶抑制剂中的用途。
本发明另一方面涉及根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的氘代化合物,或含有其的药物组合物,在制备与EZH2组蛋白甲基转移酶相关的疾病的药物中的用途。其中所述与EZH2组蛋白甲基转移酶相关的疾病可以为癌症、糖尿病炎症、免疫系统疾病、心血管内疾病、神经类疾病和呼吸类疾病。其中所述癌症可以为肺癌、胃癌、肝癌、乳腺癌、鼻咽癌、胰腺癌、卵巢癌、宫颈癌、结肠直肠癌、胶质瘤、黑色素瘤、前列腺癌、肾癌、食道癌、间皮瘤、头颈癌、膀胱癌、唾腺癌、间变性大细胞淋巴瘤、白血病、淋巴瘤、非霍奇金淋巴瘤和多发性骨髓瘤。
本发明还涉及根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的氘代化合物,或含有其的药物组合物,其作为EZH2组蛋白甲基转移酶抑制剂的用途。
本发明另一方面涉及根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的氘代化合物,或含有其的药物组合物,其用作与EZH2组蛋白甲基转移酶相关的疾病的药物的用途。其中所述与EZH2组蛋白甲基转移酶相关的疾病可以为癌症、糖尿病炎症、免疫系统疾病、心血管内疾病、神经类疾病和呼吸类疾病。其中所述癌症可以为肺癌、胃癌、肝癌、乳腺癌、鼻咽癌、胰腺癌、卵巢癌、宫颈癌、结肠直肠癌、胶质瘤、黑色素瘤、前列腺癌、肾癌、食道癌、间皮瘤、头颈癌、膀胱癌、唾腺癌、间变性大细胞淋巴瘤、白血病、淋巴瘤、非霍奇金淋巴瘤和多发性骨髓瘤。
本发明另一方面涉及一种抑制EZH2组蛋白甲基转移酶的方法,其包含向需要其的患者施用有效量的根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的氘代化合物,或含有其的药物组合物。
本发明另一方面涉及一种治疗与EZH2组蛋白甲基转移酶相关的疾病的方法,其包含向需要其的患者施用有效量的根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药,或根据本发明所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物或前药的氘代化合物,或含有其的药物组合物。其中所述与EZH2组蛋白甲基转移酶相关的疾病可以为癌症、糖尿病炎症、免疫系统疾病、心血管内疾病、神经类疾病和呼吸类疾病。其中所述癌症可以为肺癌、胃癌、肝癌、乳腺癌、鼻咽癌、胰腺癌、卵巢癌、宫颈癌、结肠直肠癌、胶质瘤、黑色素瘤、前列腺癌、肾癌、食道癌、间皮瘤、头颈癌、膀胱癌、唾腺癌、间变性大细胞淋巴瘤、白血病、淋巴瘤、非霍奇金淋巴瘤和多发性 骨髓瘤。
发明的详细说明
在本说明书中,除非另有规定,本文使用的所有技术和科学术语具有与本领域技术人员的通常理解相同含义。倘若对于本文使用的术语有多个定义,除非另有说明,以本说明书中的为准。
术语“烷基”指饱和脂肪族烃基团,其为包含1至20个碳原子的直链或支链基团,优选含有1至12个碳原子的烷基,更优选含有1至6个碳原子的烷基。非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。更优选的是含有1至6个碳原子的低级烷基,非限制性实施例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基等。烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“亚烷基”是指烷基的一个氢原子进一步被取代,例如:“亚甲基”指-CH
2-、“亚乙基”指-(CH
2)
2-、“亚丙基”指-(CH
2)
3-、“亚丁基”指-(CH
2)
4-等。
术语“烯基”指由至少由两个碳原子和至少一个碳-碳双键组成的如上定义的烷基,例如乙烯基、1-丙烯基、2-丙烯基、1-、2-或3-丁烯基等。烯基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂 环烷硫基。
术语“炔基”指由至少由两个碳原子和至少一个碳-碳三键组成的如上定义的烷基,例如乙炔基、丙炔基、丁炔基等。炔基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个碳原子,优选包含3至12个碳原子,更优选包含3至6个碳原子。单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等;多环环烷基包括螺环、稠环和桥环的环烷基。
术语“螺环烷基”指5至20元的单环之间共用一个碳原子(称螺原子)的多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺环烷基分为单螺环烷基、双螺环烷基或多螺环烷基,优选为单螺环烷基和双螺环烷基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环烷基。螺环烷基的非限制性实例包括:
术语“稠环烷基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对碳原子的全碳多环基团,其中一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠环烷基,优选为双环或三环,更优选为5元/5元或5元/6元双环烷基。稠环烷基的非限制性实例包括:
术语“桥环烷基”指5至20元,任意两个环共用两个不直接连接的碳原子的全碳多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥环烷基,优选为双环、三环或四环,更有选为双环或三环。桥环烷基的非限制性实例包括:
所述环烷基环可以稠合于芳基、杂芳基或杂环烷基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等。环烷基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧或S(O)
m(其中m是整数0至2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包含3至12个环原子,其中1~4个是杂原子;最优选包含3至8个环原子,其中1~3个是杂原子;最优选包含5至6个环原子,其中1~2或1~3个是杂原子。单环杂环基的非限制性实例包括吡咯烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、二氢咪唑基、二氢呋喃基、二氢吡唑基、二氢吡咯基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基、吡喃基等,优选1、2、5-噁二唑基、吡喃基或吗啉基。多环杂环基包括螺环、稠环和桥环的杂环基。
术语“螺杂环基”指5至20元的单环之间共用一个原子(称螺原子)的多环杂环基团,其中一个或多个环原子为选自氮、氧或S(O)
m(其中m是整数0至2)的杂原子,其余环原子为碳。其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺杂环基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺杂环基和双螺杂环基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺杂环基。螺杂环基的非限制性实例包括:
术语“稠杂环基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,其中一个或多个环原子为选自氮、氧或S(O)
m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠杂环基,优 选为双环或三环,更优选为5元/5元或5元/6元双环稠杂环基。稠杂环基的非限制性实例包括:
术语“桥杂环基”指5至14元,任意两个环共用两个不直接连接的原子的多环杂环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,其中一个或多个环原子为选自氮、氧或S(O)
m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥杂环基,优选为双环、三环或四环,更有选为双环或三环。桥杂环基的非限制性实例包括:
所述杂环基环可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,其非限制性实例包括:
杂环基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“芳基”指具有共轭的π电子体系的6至14元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至10元,例如苯基和萘基。更优选苯基。所述芳基环可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环,其非限制性实例包括:
芳基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“杂芳基”指包含1至4个杂原子、5至14个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元,含1至3个杂原子;更优选为5元或6元,含1至2个杂原子;优选例如咪唑基、呋喃基、噻吩基、噻唑基、吡唑基、噁唑基、吡咯基、四唑基、吡啶基、嘧啶基、噻二唑、吡嗪基等,优选为咪唑基、噻唑基、吡唑基或嘧啶基、噻唑基;更有选吡唑基或噻唑基。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
杂芳基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“烷氧基”指-O-(烷基)和-O-(非取代的环烷基),其中烷基的定义如上所述。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基。烷氧基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“氘代”指用氘原子替换原化合物中的氢原子的修饰。
术语“羟基”指-OH基团。
术语“卤素”指氟、氯、溴或碘。
术语“氨基”指-NH
2。
术语“氰基”指-CN。
术语“硝基”指-NO
2。
术语“氧代”指=O。
术语“羧基”指-C(O)OH。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生的场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“可药用盐”是指本发明化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。
本发明化合物的合成方法
为了完成本发明的目的,本发明采用如下技术方案。
步骤1)使用硝基苯甲酸化合物或卤代苯甲酸化合物作为起始原料,分别经过卤化或硝化反应,制得中间体a;所述的硝化或卤化反应为本领域公知的常规反应,例如在硫酸存在下,与NBS或KNO
3反应;
步骤2)将得到的中间体a与溴乙烷在碱性条件下反应制得中间体b,所述碱性条件可以为碳酸钾;
步骤3)将中间体b的硝基化合物还原为氨基化合物中间体c,所述还原条件也是本领域技术人员所公知的,例如铁粉氯化铵法;
步骤4)将中间体c与1-Boc-3-吡咯烷酮在酸性条件下催化剂的存在下发生还原胺化反应,得到中间体d,所述催化剂例如三乙酰氧基硼氢化钠;
步骤5)将中间体d与乙醛在酸性条件下催化剂的存在下发生另一还原胺化反应,得到中间体e;
步骤6)中间体e的酯水解反应得到中间体f;
步骤7)将中间体f与相应的氨甲基吡啶酮化合物在催化剂的存在下发生酰胺化反应得到中间体g,所述催化剂可以为HATU或N-甲基吗啉;
步骤8)首先将中间体g的Boc水解,然后与丙烯酸酐在催化剂的存在下反应,制得本发明化合物,所述催化剂例如N-甲基吗啉。
对于Ar位置由芳基如苯基取代的化合物,可以通过以下方案2制备。
首先通过如上方案1制得的化合物h,然后通过化合物h与对应的苯硼酸频哪醇酯在催化剂存在下反应制得本发明化合物。
对于Ar位置由杂芳基如嘧啶基取代的化合物,且该杂芳基进一步被杂环取代,则可以参照以下方案3制备。
首先,根据如上方案1的方法制得中间体i,将中间体i与联硼酸频哪醇酯在催化剂的存在下反应制得中间体j;然后中间体j与二氯嘧啶反应制得中间体k;中间体k与对应的杂环如哌啶在碱性条件下反应制得中间体l;中间体k的酯水解制得中间体m,中间体l的酯水解制得中间体n;然后,按照如上方案1的制备方法,用中间体m或n代替中间体f制得本发明化合物。
对于Ar上为芳基如苯基取代,且该苯基进一步被取代的氨基烷基取代的化合物,可以参照如下方案4制备。
首先将中间体i与羟甲基苯硼酸频那醇酯在催化剂的存在下反应制得中间体o;然后将中间体o与甲磺酰氯反应制得中间体p;然后通过中间体p与对应的各种胺反应制得中间体q;然后,按照如上方案1的制备方法,用中间体q代替中间体e制得本发明化合物。
对于Ar上直接由氨基取代的化合物,可以参照以下方案5制备。
首先通过如上的方案1制备中间体r,然后将中间体r与相应的胺在催化剂的存在下反应,制得中间体s;然后,利用方案1的制备方法,用中间体s代替中间体g制得本发明的化合物。
方案6为本发明一类特殊化合物的制备方法,首先通过方案1制得中间体t,然后由中间体t与4-二甲氨基巴豆酸盐酸盐反应,制得本发明的化合物。
其中,R
7、R
9、R
13、R
m、R
n、R
h、R
i、-Q
1-T
1如通式(I)中所定义。
本发明通式(I)所示的化合物在药学上可接受的盐,可以为酸式加成盐或碱式加成盐。酸可选择无机酸包括但不限于盐酸、硫酸、磷酸、氢溴酸;酸还可选 择有机酸包括但不限于柠檬酸、马来酸、草酸,甲酸、乙酸、丙酸、乙醇酸、苯甲酸、富马酸、三氟乙酸、琥珀酸、酒石酸、乳酸、谷氨酸、天门冬氨酸、水杨酸、丙酮酸、甲磺酸、苯磺酸、对苯磺酸。碱可选择无机碱包括但不限于氢氧化钠、氢氧化钾、氢氧化镁、氢氧化钙;碱还可选择有机碱包括但不限于氢氧化铵、三乙胺、精氨酸或赖氨酸。
本发明通式(I)所示的化合物及药学上可接受的盐可以溶剂化形式或未溶剂化形式存在例如水合式等。
本发明通式(I)所示的化合物,其前药应遵照前药设计原则,在生物体内正常生理状况下,能够通过酶解、水解、酸解或代谢降解,释放出原活性通式(I)所示的化合物。这里包括但不限于化合物上羟基基团的脂化(如形成磷酸脂和碳酸脂),氨基基团和羧基基团的保护。前药设计参照(1)Karaman R,Prodrugs design based on inter-and intramolecular chemical processes.Chem Biol Drug Des.82(6):643-68,2013;(2)Rautio J等,Prodrugs:design and clinical applications.Nat Rev Drug Discov.7(3):255-70 2008;(3)Jampilek J.Prodrugs:pharmaceutical design and current perspectives.Curr Pharm Des.17(32):3480-1,2011;(4)Bundgaard H.Design of Progrugs.Elservier,1985。
本发明另一方面将通式(I)所示的化合物或药学上可接受的盐或前药制备成临床上可使用的药物组合物。根据临床适应症,给药途径与方式,其药用制剂包括但不限于口服制剂如片剂、凝胶剂、软/硬胶囊、乳剂、分散性粉剂、颗粒剂、水/油悬乳剂;注射剂包括静脉注射剂、肌肉注射剂、腹腔注射剂、直肠给药栓剂、颅内注射剂,这些剂型可为水溶液也可为油类溶液;局部制剂包括霜剂、软膏剂、凝胶剂、水/油溶液以及包合物制剂;吸入剂型包括细粉、液体气溶胶以及适合于体内植入的各种剂型。
本发明的药物组合物根据需要加入常规药用辅料。这些辅料应符合药物制剂制备工艺规则,与活性成分相兼容。固体口服制剂辅料选用但不限于甘露醇、乳糖、淀粉、硬脂酸镁、纤维素、葡萄糖、蔗糖、环糊精以及促进肠吸收分子载体维生素E-PEG1000。口服制剂可加入适当的着色剂、甜味剂、矫味剂及防腐剂。
本发明通式(I)所示的化合物按0.1-100mg/kg单位剂量给予温血动物。
包含本发明通式(I)所示的化合物或药学上可接受的盐作为有效成分的药物组合物,主要治疗与EZH2相关的临床疾病。其中包括但不限于癌症、糖尿病炎症、免疫系统疾病、心血管内疾病、神经类疾病以及呼吸类疾病。
上述临床疾病中,癌症包括但不限于肺癌、胃癌、肝癌、乳腺癌、鼻咽癌、胰腺癌、卵巢癌、宫颈癌、结肠直肠癌、胶质瘤、黑色素瘤、前列腺癌、肾癌、食道癌、间皮瘤、头颈癌、膀胱癌、唾腺癌、间变性大细胞淋巴瘤、白血病、淋巴瘤、非霍奇金淋巴瘤及多发性骨髓瘤。
本发明药物组合物在上述癌症治疗中,可单独使用,或与临床上常规使用的 手术、放射疗法、化学疗法、免疫疗法、融瘤病毒、RNAi、癌症辅助治疗的一种或多种方法联合治疗,其中包括但不限于以下抗肿瘤类药物和治疗方法:
1)烷化剂如顺铂、顺铂、奥沙利铂、苯丁酸氮芥、卡环磷酰胺,氮芥、美法仑、替莫唑胺、白消安、亚硝基脲类。
2)抗肿瘤抗生素类如阿霉素、博来霉素、多柔比星、道诺霉素、表柔比星、伊达比星、丝裂霉素C、放线菌素、光神霉素;抗有丝分裂药如长春新碱,长春碱,长春地辛,长春瑞滨,紫杉醇、泰索帝、Polo激酶抑制剂。
3)抗代谢和抗叶酸剂如氟嘧啶、雷甲氨蝶呤、阿糖胞苷、替曲塞、羟基脲。
4)拓扑异构酶抑制剂如表鬼臼毒素、喜树碱。
5)细胞生长抑制剂如抗雌激素/抗雄激素类药物如他莫昔芬、氟维司群、托瑞米芬、雷诺昔芬、屈诺昔芬、碘昔芬;如比卡鲁胺、氟他胺、尼鲁米特、醋酸环丙孕酮。
LHRH拮抗剂或LHRH激动剂如戈舍瑞林、亮丙瑞林、和布舍瑞林、孕激素类如醋酸甲地孕酮。
芳香酶抑制剂如阿那曲唑、来曲唑、伏罗唑、伊西美坦、5a-还原酶抑制剂如非那雄胺。
6)抗侵袭剂如c-Src激酶家族抑制剂,金属蛋白酶抑制剂,尿激酶纤溶酶原激活物受体功能的抑制剂或者类肝素酶的抗体。
7)生长功能的抑制剂如生长因子抗体和生长因子受体抗体如抗HER2抗体曲妥珠单抗、抗EGFR抗体帕尼单抗、抗EGFR抗体西妥昔单抗等;这种抑制剂还包括其它酪氨酸激酶抑制剂以及丝氨酸/苏氨酸激酶的抑制剂如Ras/Raf信号传导抑制剂,MEK和/或AKT激酶的细胞信号传导抑制剂、c-kit抑制剂、abl激酶抑制剂、PI3激酶抑制剂、FLT3激酶抑制剂、CSF-1R激酶抑制剂、IGF受体激酶抑制剂,极光激酶抑制剂,NTRKA/B/C激酶抑制剂,细胞周期蛋白依赖性激酶抑制剂如CDK2和/或CDK4,CDK6抑制剂及转录激酶CDK7/9/12/13抑制剂。
8)抗血管生成剂如抑制血管内皮生长因子作用的药剂贝伐珠单抗以及VEGF受体酪氨酸激酶抑制剂。
9)肿瘤免疫治疗法包括任何提高患者肿瘤细胞的免疫原性的体外和体内方法。如细胞因子IL-2、IL-4或者GM-CSF进行转染;降低T细胞无效能的方法如抗PD-1/PD-L单抗;使用转染的免疫细胞如细胞因子转染的树突状细胞的方法;使用细胞因子转染的肿瘤细胞系的方法;降低免疫抑制性细胞如调节性T细胞、髓源性抑制细胞、或表达吲哚胺2,3-脱氧酶的树突状细胞的功能方法;提高免疫细胞活性的激动剂如STING以及肿瘤相关抗原蛋白类或肽类组成的癌症疫苗的方法。
10)嵌合抗原受体T细胞免疫疗(CAR T)。
11)肿瘤基因治疗如CRISPR-Cas 9,RNAi和基因转导。
应予说明,如果任何给定取代基的数量没有规定(例如,卤代烷基),则可以存在一个或多个取代基。例如,“卤代烷基”可以含有一个或多个相同或不同的卤素。
在本文的描述中,如果化学结构和化学名称彼此矛盾时,则是以其化学结构为准。
当在本文使用时,对于任何保护基团和其他化合物的缩写,除非另有说明,以其常用的公认缩写表示,或根据IUPAC-IUB Commission on Biochemical Nomenclature表示(参见,Biochem.1972,77:942-944)。
以下结合实施例进一步描述本发明,但这些实施例并非限制着本发明的范围。
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移以10
-6(ppm)的单位给出。NMR的测定是用Brukerdps400型核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d
6),氘代氯仿(CDCl
3),内标为四甲基硅烷(TMS)。
MS的测定用1100Series LC/MSD Trap(ESI)质谱仪(生产商:Agilent)。
薄层层析硅胶板使用青岛海洋化工GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。
柱层析一般使用青岛海洋硅胶200~300目硅胶为载体。
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成,或商业途径获得;如无特殊说明,均来自商业途径获得。
实施例中无特殊说明,反应能够均在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
实施例中无特殊说明,溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂体系包括:A:二氯甲烷和甲醇体系,B:石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的氨水和醋酸等碱性或酸性试剂进行调节。
实施例
实施例1 3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
步骤1:5-溴-3-硝基-2-甲基苯甲酸(1a)的制备
将3-硝基-2-甲基苯甲酸(27.1g,150mmol)溶解于100mL硫酸中,加热至60℃,分批加入NBS(29.36g,165mmol),加完搅拌5小时,冷至室温。将反应液滴加至500mL冰水中,搅拌30分钟,过滤,滤饼用水淋洗,干燥得36g标题产物。
步骤2:5-溴-3-硝基-2-甲基苯甲酸乙酯(1b)的制备
将5-溴-3-硝基-2-甲基苯甲酸(36g,138mmol)、溴乙烷(22.64g,208mmol)、碳酸钾(38g,276mmol)加到120mL DMF中,室温搅拌15小时。反应液中加入水和乙酸乙酯萃取。有机层经无水硫酸钠干燥,过滤,滤液减压浓缩得39g油状标题产物,其不经纯化直接进行下一步。
步骤3:5-溴-3-氨基-2-甲基苯甲酸乙酯(1c)的制备
将5-溴-3-硝基-2-甲基苯甲酸乙酯(26g,90mmol)、铁粉(20.2g,360mmol)、氯化铵(19.3g,360mmol)加入260mL乙醇和100mL水的混合溶剂中,将混合物回流搅拌2小时。滤除铁泥,滤液减压浓缩。残余物通过柱层析(洗脱剂:石油醚:乙酸乙酯40:1~5:1)得产物19.5g标题产物。
步骤4:3-(5-溴-3-乙氧基羰基-2-甲基-苯基氨)-吡咯烷-1-基甲酸叔丁醇酯(1d)的制备
将5-溴-3-氨基-2-甲基苯甲酸乙酯(5.7g,22.1mmol)、1-Boc-3-吡咯烷酮(4.92g,26.6mmol)溶解于30mL乙酸中,分批加入三乙酰氧基硼氢化钠(9.4g,44.3mmol),加完搅拌2小时。向反应液中加入水和乙酸乙酯萃取。乙酸乙酯层用氢氧 化钠水溶液洗涤,有机层经无水硫酸钠干燥,过滤,滤液减压浓缩。残余物通过柱层析(洗脱剂:石油醚:乙酸乙酯50:1~5:1)得6.4g标题产物。
步骤5:3-[(5-溴-3-乙氧基羰基-2-甲基-苯基)-乙基-氨基]-吡咯烷-1-基甲酸叔丁醇酯(1e)的制备
将3-(5-溴-3-乙氧基羰基-2-甲基-苯基氨)-吡咯烷-1-基甲酸叔丁醇酯(3.2g,7.5mmol)、乙醛(0.66g,15mmol)溶解于20mL乙酸中,分批加入三乙酰氧基硼氢化钠(3.2g,15mmol),加完搅拌2小时。向反应液中加入水和乙酸乙酯萃取。乙酸乙酯层用氢氧化钠水溶液洗涤,有机层经无水硫酸钠干燥,过滤,滤液减压浓缩。残余物通过柱层析色谱法纯化(洗脱剂:石油醚:乙酸乙酯50:1~5:1),得3.3g标题产物。
步骤6:3-[(5-溴-3-甲酸基-2-甲基-苯基)-乙基-氨基]-吡咯烷-1-基甲酸叔丁醇酯(1f)的制备
将3-[(5-溴-3-乙氧基羰基-2-甲基-苯基)-乙基-氨基]-吡咯烷-1-基甲酸叔丁醇酯(3.3g,7.25mmol)、氢氧化钠(1g,25mmol)加入至30mL乙醇和10mL水的混合溶剂中,回流搅拌1小时,冷至室温。反应液用1M柠檬酸水溶液调PH至3-4,然后用乙酸乙酯萃取,乙酸乙酯层用水洗涤。有机层经无水硫酸钠干燥,过滤,滤液减压浓缩得3.15g标题产物。
步骤7:3-({5-溴-3-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-氨基甲酰基]-2-甲基-苯基}-乙基-氨基)-吡咯烷-1-基甲酸叔丁醇酯(1g)的制备
将3-[(5-溴-3-甲酸基-2-甲基-苯基)-乙基-氨基]-吡咯烷-1-基甲酸叔丁醇酯(3.15g,7.4mmol)、3-氨甲基-4,6-二甲基-1H-吡啶-2-酮盐酸盐(1.95g,10.4mmol)、N-甲基吗啉(6g,59.2mmol)加到18mL DMF中,搅拌溶解,加入HATU(4.22g,11.1mmol)搅拌两小时。向反应液中滴加18mL水,有固体析出。将固体过滤、干燥得4.3g标题产物。
步骤8:3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
将3-({5-溴-3-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-氨基甲酰基]-2-甲基-苯基}-乙基-氨基)-吡咯烷-1-基甲酸叔丁醇酯(280mg,0.5mmol)溶解于10mL二氯甲烷中,加入1mL三氟乙酸,回流10小时。将反应液减压浓缩,再加入10mL二氯甲烷、N-甲基吗啉(400mg,4mmol),冰水浴冷却下加入丙烯酸酐(75mg,0.6mmol),搅拌2小时,然后向其中加入水和二氯甲烷萃取。有机层经无水硫酸钠干燥,过滤,滤液减压浓缩。残余物通过薄层色谱法纯化(展开剂二氯甲烷:甲醇比20:1),得到30mg标题产物。
1H-NMR(CDCl
3)δ:0.87(3H,t),1.65-2.08(2H,m),2.27(3H,s),2.28(3H,s),2.41(3H,s),2.97-3.02(2H,m),3.22-3.35(1H,m),3.42-3.55(1H,m),3.65-3.85(3H,m),4.54(2H,d),5.65-5.71(1H,m),6.01(1H,s),6.34-6.42(2H,m),7.16-7.21(1H,t), 7.26-7.30(2H,m),12.00(1H,br)。
m/z ESI M+H
+517.0。
实施例2 3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-氟-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
步骤1:5-氟-3-硝基-2-甲基苯甲酸(2a)的制备
将5-氟-2-甲基苯甲酸(7.7g,50mmol)溶解于于60mL硫酸中,分批加入硝酸钾(6.1g,60mmol),加完室温搅拌2小时。将反应液倒入350mL冰水中,搅拌30分钟,过滤,滤饼用水淋洗,干燥得产物3.7g。
其余步骤与实施例1的步骤2至步骤8的操作相同,制得化合物2。
1H-NMR(CDCl
3)δ:0.88(3H,t),1.80-1.91(1H,m),2.01-2.09(1H,m),2.26(3H,s),2.28(3H,s),2.41(3H,s),2.98-3.05(2H,q),3.22-3.35(1H,m),3.40-3.55(1H,m),3.62-3.85(3H,m),4.54(2H,d),5.65-5.71(1H,m),5.97(1H,s),6.33-6.42(2H,m),6.87-6.94(2H,m),7.20(1H,t),11.05(1H,br)。
m/z ESI M+H
+454.9。
实施例3 3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-氯-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
与实施例2的制备方法相同,除了用5-氯-2-甲基苯甲酸替代5-氟-2-甲基苯甲酸,制得化合物3。
1H-NMR(CDCl
3)δ:0.88(3H,t),1.75-1.86(1H,m),1.99-2.08(1H,m),2.26(3H,s),2.29(3H,s),2.40(3H,s),2.96-3.04(2H,q),3.22-3.32(1H,m),3.40-3.55(1H,m),3.60-3.85(3H,m),4.54(2H,d),5.62-5.71(1H,m),5.99(1H,s),6.33-6.42(2H,m),7.11-7.15(2H,m),7.23(1H,t),12.01(1H,br)。
m/z ESI M+H
+470.8。
实施例4 3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
与实施例1的制备方法相同,除了用3-硝基-2-甲基苯甲酸替代5-溴3-硝基-2-甲基苯甲酸,制得化合物4。
1H-NMR(DMSO-d6)δ:0.81(3H,t),1.66-1.95(1H,m),1.98-2.07(1H,m),2.11(3H,s),2.20(3H,s),2.21(3H,s),2.95-3.03(2H,m),3.20-3.27(1H,m),3.46-3.55(2H,m),3.65-3.72(1H,m),3.75-3.85(1H,m),4.27(2H,d),5.58-5.66(1H,m),5.86(1H,s),6.05-6.14(1H,m),6.48-6.58(1H,m),7.01(1H,d),7.18(1H,t),7.28(1H,d),8.05-8.09(1H,t),11.46(1H,s)。
m/z ESI M+H
+437.2。
实施例5 3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-2,5-二溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-苯甲酰胺的制备
与实施例2的制备方法相同,除了用2,5-二溴苯甲酸替代5-氟-2-甲基苯甲酸,制得化合物5。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.80-1.92(1H,m),1.95-2.04(1H,m),2.11(3H,s),2.20(3H,s),3.05-3.13(2H,m),3.26-3.32(1H,m),3.46-3.58(2H,m),3.68-3.80(1H,m),3.88-3.96(1H,m),4.26(2H,d),5.60-5.67(1H,m),5.87(1H,s),6.08-6.12(1H,m),6.51-6.59(1H,m),7.21(1H,d),7.58(1H,d),8.36-8.42(1H,t),11.50(1H,br)。
m/z ESI M+H
+581.0。
实施例6 3-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-5-溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
与实施例1的制备方法相同,除了用N-Boc-哌啶-4-酮替代N-Boc-吡咯烷-3- 酮,制得化合物6。
1H-NMR(DMSO-d
6)δ:0.79(3H,t),1.32-1.50(2H,m),1.72(2H,br),2.12(2H,s),2.16(3H,s),2.20(3H,s),2.58-2.63(1H,m),2.95-3.10(4H,m),3.99(1H,br),4.26(2H,br),4.36(1H,br),5.65(1H,dd),5.87(1H,s),6.07(1H,dd),6.78(1H,dd),7.10(1H,d),7.32(1H,d),8.24(1H,br),11.48(1H,s)。
m/z ESI M+H
+529.0。
实施例7 3-[(1-丙烯酰基-氮杂环丁烷-3-基)-乙基-氨基]-5-溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
与实施例1的制备方法相同,除了用N-Boc-氮杂环丁烷-3-酮替代N-Boc-吡咯烷-3-酮,制得化合物7。
1H-NMR(DMSO-d
6)δ:0.82(3H,t),2.11(3H,s),2.17(3H,s),2.19(3H,s),2.95(2H,q),3.57(1H,m),3.80(1H,m),4.00(1H,m),4.20-4.35(4H,m),5.64(1H,dd),5.87(1H,s),6.08(1H,dd),6.29(1H,dd),7.12(1H,d),7.19(1H,d),8.27(1H,t),11.49(1H,s)。
m/z ESI M+H
+501.0。
实施例8 3-(4-丙烯酰基-哌嗪-1-基)-5-溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
步骤1:4-(5-溴-3-乙氧酰基-2-甲基-苯基)-哌啶-1-甲酸叔丁醇酯(8a)的制备
将5-溴-3-氨基-2-甲基苯甲酸乙酯(1c)(1.4g,5.5mmol)与二(2-氯乙基)胺盐酸盐(1.06g,6mmol)混合均匀。氮气氛下将混合物加热至170℃,熔融反应1小时。向反应物中补加二(2-氯乙基)胺盐酸盐(1.06g,6mmol)再反应1小时。将反应物冷却至室温,加入50mL二氯甲烷和25mL 1M氢氧化钠水溶液,搅拌下加入(Boc)
2O(3.3g,15mmol),搅拌1小时。有机层用无水硫酸钠干燥, 过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯40:1~10:1)纯化,得产物0.7g。
其余步骤与实施例1相同,除了用化合物8a替代实施例1的化合物1e制得化合物8。
1H-NMR(DMSO-d
6)δ:2.13(3H,s),2.20(6H,s),2.84(4H,br),3.69(4H,br),4.28(2H,d),5.69(1H,dd),5.85(1H,s),6.12(1H,dd),6.79(1H,dd),7.10(1H,d),7.18(1H,d),8.05(1H,br),11.29(1H,s)。
m/z ESI M+H
+489.1。
实施例9 3-[(1-丙烯酰基-吡咯烷-3-基)-丁基-氨基]-5-溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
与实施例1的制备方法相同,除了用丁醛替代乙醛,制得化合物9。
1H-NMR(DMSO-d
6)δ:0.81(3H,t),1.17-1.19(4H,m),1.80-1.88(1H,m),1.95-2.05(1H,m),2.12(3H,s),2.14(3H,s),2.19(3H,s),2.91-3.08(2H,m),3.22-3.32(1H,m),3.45-3.55(2H,m),3.65-3.82(2H,m),4.25(2H,d),5.60-5.67(1H,m),5.87(1H,s),6.05-6.13(1H,m),6.50-6.55(1H,m),7.13(1H,d),7.46(1H,d),8.26(1H,t),11.49(1H,s)。
m/z ESI M+H
+542.7。
实施例10 3-[(1-丙烯酰基-吡咯烷-3-基)-甲基-氨基]-5-溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
步骤1:3-[(5-溴-3-乙氧基羰基-2-甲基-苯基)-甲基-氨基]-吡咯烷-1-基甲酸叔丁醇酯(10a)的制备
将3-(5-溴-3-乙氧基羰基-2-甲基-苯基氨)-吡咯烷-1-基甲酸叔丁醇酯(1d)(1.5g,3.5mmol)、碘甲烷(0.98g,7mmol)、氢化钠(0.43g,10.6mmol)溶解于4mLDMF中,搅拌3小时。向反应液中加入水和乙酸乙酯萃取。有机层经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯50:1~10:1)纯化,得产物0.6g。
其余步骤与实施例1相同,除了用化合物10a替代实施例1的化合物1e,制得化合物10。
1H-NMR(CDCl
3)δ:1.85-1.96(1H,m),2.01-2.06(1H,m),2.25(3H,s),2.28(3H,s),2.41(3H,s),2.59(3H,s),3.37-3.41(1H,m),3.47-3.56(1H,m),3.62-3.81(3H,m),4.53(2H,d),5.65-5.70(1H,m),6.01(1H,s),6.35-6.41(2H,m),7.16(1H,t),7.23-7.28(2H,m),12.11(1H,br).
m/z ESI M+H
+501.1
实施例11 3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-5-[6-(4-甲基-哌嗪-1-基)-吡啶-3-基氨基]-苯甲酰胺的制备
步骤1 2-(4-甲基-哌嗪-1-基)-5-氨基吡啶(11a)的制备
将2-氯-5-硝基吡啶(3.17g,20mmol)、N-甲基哌嗪(3g,30mmol)、碳酸钾(5.52g,40mmol)加到5mL DMF中,于60℃加热1小时。将反应液冷却至室温,然后倒入50mL水中,有固体析出。将固体过滤并将产物加至5mL水和20mL乙醇的混合溶剂中,加入铁粉(4.48g,80mmol)和盐酸(2mL),回流2小时。向反应液中加入氢氧化钠调节PH至8,过滤,滤液减压浓缩。残余物通过柱层析色谱法纯化(洗脱剂:二氯甲烷:甲醇100:1~30:1),得到产物0.67g。
步骤2:3-({5-[2-(4-甲基-哌嗪-1-基)-吡啶-5-氨基]-3-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-氨基甲酰基]-2-甲基-苯基}-乙基-氨基)-吡咯烷-1-基甲酸叔丁醇酯(11b)的制备
将3-({5-溴-3-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-氨基甲酰基]-2-甲基-苯基}-乙基-氨基)-吡咯烷-1-基甲酸叔丁醇酯(1g)(170mg,0.3mmol)、2-(4-甲基-哌嗪-1-基)-5-氨基吡啶(11a)(64mg,0.33mmol)、Pd
2(dba)
3(28mg,0.03mmol)、X-phoes(29mg,0.06mmol)、叔丁醇钠(115mg,1.2mmol)加入至20mL甲苯中,氮气氛下90℃反应2小时。将反应液过滤,浓缩,残余物通过薄层色谱法(洗脱剂:二氯甲烷:甲醇80:1~25:1)纯化,得产物110mg。
其余步骤与实施例1相同,除了用化合物11b替代化合物1g,制得化合物11。
1H-NMR(DMSO-d
6)δ:0.84(3H,t),1.60-1.95(2H,m),2.08(3H,s),2.11(3H,s),2.17(3H,s),2.31(3H,s),2.55(4H,br),2.92-3.05(2H,m),3.23-3.31(H,m),3.52(2H,br),3.70(2H,br),4.24(2H,d),5.64(1H,m),5.86(1H,s),6.11(1H,m),6.50-6.56(2H,m),6.75(1H,s),6.82(1H,d),7.36(1H,d),7.75(1H,d),7.97-8.03(2H,m),11.46(1H,s)。
m/z ESI M+H
+628.2
实施例12 3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2-甲基-5-[4-(吗啉-4-基甲基)苯基氨基]-苯甲酰胺的制备
步骤1:4-(吗啉-4-基甲基)苯胺(12a)的制备
将4-硝基溴苄(10.8g,50mmol)、吗啉(4.6g,52.5mmol)、碳酸钾(10.35g,75mmol)加到25mL乙腈中,60℃加热4小时。将反应液冷却至室温,并倒入100mL水中,用乙酸乙酯萃取。将有机层用无水硫酸钠干燥,过滤,减压浓缩。将残余物加入至40mL水和100mL乙醇的混合溶剂中,加入铁粉(10.1g,180mmol)和盐酸(4mL),回流2小时。反应液加氢氧化钠调节PH至8,过滤,滤 液减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷:甲醇100:1~25:1)纯化,得到产物7.2g。
其余步骤与实施例11相同,除了用化合物12a替代化合物化合物11a,制得化合物12。
1H-NMR(CDCl
3)δ:0.89(3H,t),1.85-1.93(1H,m),2.01-2.05(1H,m),2.23(3H,s),2.26(3H,s),2.39(3H,s),2.95-3.03(2H,m),3.23-3.35(1H,m),3.38-3.85(12H,m),4.51(2H,d),5.63-5.71(1H,m),5.87(1H,br),5.92(1H,s),6.32-6.42(2H,m),6.86-6.99(4H,m),7.13-7.18(1H,br),7.21-7.28(2H,d),11.63(1H,br)。
m/z ESI M+H
+627.3。
实施例13 3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-(4-甲基-哌嗪-1-基)-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
步骤1:3-({5-(4-甲基-哌嗪-1-基)-3-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-氨基甲酰基]-2-甲基-苯基}-乙基-氨基)-吡咯烷-1-基甲酸叔丁醇酯的制备
将3-({5-溴-3-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-氨基甲酰基]-2-甲基-苯基}-乙基-氨基)-吡咯烷-1-基甲酸叔丁醇酯(1g)(0.84g,1.5mmol)、N-甲基哌嗪(0.23g,2.3mmol)、Pd
2(dba)
3(0.18g,0.2mmol)、X-phoes(0.19g,0.4mmol)加入至35mL甲苯中,再加入叔丁醇钠(0.77g,8mmol),氮气氛下于100℃搅拌反应15小时。反应液用乙酸乙酯和水萃取,有机层用无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷:甲醇100:1~30:1)纯化,得产物0.3g。
其余步骤与实施例1相同,除了用化合物13a替代化合物1g,制得化合物13。
1H-NMR(CDCl3)δ:0.86(3H,t),1.85-1.93(1H,m),2.01-2.08(1H,m),2.22(3H,s),2.25(3H,s),2.41(3H,s),2.49(3H,s),2.77(4H,br),2.98(2H,q),3.24-3.33(5H,m),3.41-3.82(4H,m),4.53(2H,d),5.61-5.68(1H,m),5.98(1H,s),6.32-6.42(2H,m), 6.74-6.77(2H,d),7.02-7.12(1H,br),11.63(1H,br)。
m/z ESI M+H
+535.0。
实施例14 5-溴-3-{[1-(4-二甲氨基-丁-2-烯酰基)-吡咯烷-3-基]-乙基-氨基}-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2-甲基-苯甲酰胺的制备
步骤1:5-溴-3-{[1-(4-二甲氨基-丁-2-烯酰基)-吡咯烷-3-基]-乙基-氨基}-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2-甲基-苯甲酰胺的制备
将5-溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-(乙基-吡咯烷-3-基-氨基)-2-甲基-苯甲酰胺(1g)(0.23g,0.5mmol)、4-二甲氨基巴豆酸盐酸盐(0.16g,1mmol)、N-甲基吗啉(0.3g,3mmol)溶解于4mL DMF中,搅拌下加入HATU(0.57g,1.5mmol),搅拌20小时。向反应液中加入水和乙酸乙酯萃取,有机层经无水硫酸钠干燥,过滤,减压浓缩。残余物通过薄层色谱法(展开剂二氯甲烷:甲醇比例15:1)纯化,得到产物15mg。
1H-NMR(CDCl
3)δ:0.88(3H,t),1.85-1.95(1H,m),1.98-2.08(1H,m),2.23(3H,s),2.29(3H,s),2.38(3H,s),2.52(6H,s),2.99(2H,q),3.16-3.78(7H,m),4.51(2H,d),5.96(1H,s),6.29-6.46(1H,m),6.80-6.91(1H,m),7.20-7.30(3H,m),12.05(1H,br)。
m/z ESI M+H
+571.8。
实施例15 5-溴-3-{[1-(1-氧-丁-2-炔基)-吡咯烷-3-基]-乙基-氨基}-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2-甲基-苯甲酰胺的制备
与实施例14的制备方法相同,除了用2-丁炔酸替代4-二甲氨基巴豆酸盐酸盐,制得化合物15。
1H-NMR(DMSO-d
6)δ:0.80(3H,t),1.72-1.83(1H,m),1.90-2.05(4H,m),2.12(3H,s),2.14(3H,s),2.19(3H,s),2.93-3.00(2H,m),3.20-3.30(1H,m),3.43-3.53(2H,m),3.65-3.72(1H,m),3.80-3.86(1H,m),4.25(2H,d),5.87(1H,s),7.15(1H,d),7.44(1H,d), 8.26(1H,t),11.49(1H,s)。
m/z ESI M+H
+528.7。
实施例16 3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-甲氧基-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2甲基-苯甲酰胺的制备
步骤1:5-羟基-3-硝基-2-甲基苯甲酸乙酯的制备
将5-溴-3-硝基-2-甲基苯甲酸乙酯(1b)(13.8g,47.9mmol)、联硼酸频哪醇酯(12.8g,50.3mmol)、乙酸钾(9.4g,95.8mmol)、Pd(dppf)
2Cl
2(0.5g)加到140mL二氧六环中,氮气氛下回流反应20小时。将反应液减压浓缩,残余物加入水和乙酸乙酯萃取,有机层减压浓缩,得黑色固体。将该固体加入至200mL丙酮中,加入75mL饱和Oxone水溶液中,室温搅拌1.5小时,然后加入亚硫酸钠水溶液淬灭过量的Oxone。将反应液减压浓缩,加入水和乙酸乙酯萃取。有机层经无水硫酸钠干燥,过滤,减压浓缩,制得粗产物16.6g,其未经纯化用于后续步骤。
步骤2:3-[(5-羟基-3-乙氧基羰基-2-甲基-苯基)-乙基-氨基]-吡咯烷-1-基甲酸叔丁醇酯(16b)的制备
与实施例1的步骤3、4、5的制备方法相同,除了用化合物16a代替化合物1b,制得化合物16b。
步骤3:3-[(5-甲氧基-3-乙氧基羰基-2-甲基-苯基)-乙基-氨基]-吡咯烷-1-基甲酸叔丁醇酯的制备
将3-[(5-羟基-3-乙氧基羰基-2-甲基-苯基)-乙基-氨基]-吡咯烷-1-基甲酸叔丁醇酯(0.4g,1mmol)、碘甲烷(0.52g,3mmol)、碳酸钾(0.42g,3mmol)加到2mLDMF中,室温搅拌16小时。向反应液中加入水和乙酸乙酯萃取,有机层经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯100:1~10:1)得产物0.32g。
其余步骤与实施例1的制备方法相同,除了用化合物16c替代化合物1e,制得化合物16。
1H-NMR(CDCl
3)δ:0.87(3H,t),1.82-1.90(1H,m),2.01-2.08(1H,m),2.23(3H,s),2.27(3H,s),2.43(3H,s),2.95-3.03(2H,m),3.20-3.35(1H,m),3.42-3.53(1H,m),3.62-3.84(6H,m),4.54(2H,d),5.62-5.69(1H,m),6.01(1H,s),6.33-6.42(2H,m),6.71-6.78(2H,m),7.03-7.10(1H,br),12.05(1H,br)。
m/z ESI M+H
+466.9。
实施例17N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-羟甲基-4-甲基-联苯-3-甲酰胺的制备
步骤1:4-羟甲基苯硼酸频哪醇酯的制备
将4-溴苄醇(15g,80.2mmol)、联硼酸频哪醇酯(30.6g,120.3mmol)、乙酸钾(23.6g,240mmol)、Pd(PPh
3)
2Cl
2(5.6g,8mmol)加入至150mL二氧六 环中,氮气氛下于85℃搅拌3小时。将反应液过滤,减压浓缩,然后加入水和乙酸乙酯萃取。有机层经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯60:1~10:1)纯化,得到16g标题产物。
步骤2:N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-叔丁氧酰基-吡咯烷-3-基)-乙基-氨基]-4'-羟甲基-4-甲基-联苯-3-甲酰胺
将3-({5-溴-3-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-氨基甲酰基]-2-甲基-苯基}-乙基-氨基)-吡咯烷-1-基甲酸叔丁醇酯(1g)(2.3g,4.1mmol)、4-羟甲基苯硼酸频哪醇酯(0.96g,4.1mmol)、四三苯基膦钯(0.47g,0.41mmol)、碳酸钾(1.13g,8.2mmol)加入至30mL乙腈和15mL水的混合溶剂中,氮气氛下回流4小时。将反应液过滤,减压浓缩,然后加水和乙酸乙酯萃取。有机层经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯50:1~1:1)纯化,得到产物0.62g。
其余步骤与实施例1的制备方法相同,除了用化合物17b替代化合物1g,制得化合物17。
1H-NMR(DMSO-d6)δ:0.86(3H,t),1.80-1.89(1H,m),1.95-2.05(1H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),3.01-3.08(2H,m),3.27-3.32(1H,m),3.48-3.55(2H,m),3.67-3.74(1H,m),3.91-3.98(1H,m),4.30(2H,d),4.54(2H,d),5.23(1H,t),5.57-5.65(1H,m),5.87(1H,s),6.05-6.13(1H,m),6.48--6.58(1H,m),7.28(1H,d),7.40(2H,d),7.52(1H,d),7.63(2H,d),8.20-8.26(1H,t),11.48(1H,s)。
m/z ESI M+H
+542.8。
实施例18N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-(哌啶-1-基甲基)-4-甲基-联苯-3-甲酰胺的制备
步骤1:N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷 -3-基)-乙基-氨基]-4'-(甲磺酰氧基甲基)-4-甲基-联苯-3-甲酰胺(18a)的制备
将N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-羟甲基-4-甲基-联苯-3-甲酰胺(0.58g,1mmol)、DIEA(0.26g,2mmol)溶解于10mL二氯甲烷中,冰水浴冷却下滴加甲磺酰氯(0.14g,1.2mmol)的2mL二氯甲烷溶液,加完搅拌2小时。向反应液中加入水和二氯甲烷萃取,有机层经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯50:1~2:1)纯化,得到产物0.14g。
步骤2:N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-(哌啶-1-基甲基)-4-甲基-联苯-3-甲酰胺(18b)的制备
将N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-甲磺酰氧基甲基-4-甲基-联苯-3-甲酰胺(0.14g,0.2mmol)加入至2mL哌啶中,搅拌2小时。反应液加入水和二氯甲烷萃取。有机层经无水硫酸钠干燥,过滤,减压浓缩。残余物通过薄层色谱法(展开剂二氯甲烷:甲醇比例20:1)纯化,得到产物90mg。
其余步骤与实施例1的制备方法相同,除了用化合物18b替代化合物1g,制得化合物18。
1H-NMR(DMSO-d
6)δ:0.85(3H,t),1.52-2.05(8H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.33-2.92(4H,m),2.95-3.12(2H,m),3.25-3.32(1H,m),3.52(2H,s),3.70-3.76(2H,m),3.90-3.97(2H,m),4.30(2H,d),5.60-5.67(1H,m),5.87(1H,s),6.07-6.12(1H,m),6.52-6.55(1H,m),7.13(1H,d),7.50-7.65(3H,m),7.71(2H,d),8.24(1H,t),11.48(1H,s)。
m/z ESI M+H
+609.9。
实施例19N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-(二甲氨基甲基)-4-甲基-联苯-3-甲酰胺的制备
与实施例18的制备方法相同,除了用二甲胺四氢呋喃溶液替代哌啶,制得化合物19。
1H-NMR(DMSO-d6)δ:0.85(3H,t),1.70-1.89(1H,m),1.95-2.05(1H,m),2.11(3H,s),2.21(3H,s),2.26(3H,s),2.69(6H,s),2.97-3.08(2H,m),3.24-3.31(1H,m),3.50-3.58(2H,m),3.67-3.77(1H,m),3.91-3.98(1H,m),4.22(2H,s),4.30(2H,d),5.59-5.67(1H,m),5.87(1H,s),6.07-6.12(1H,m),6.50-6.57(1H,m),7.33(1H,d),7.55-7.62(3H,m),7.75-7.81(2H,d),8.20-8.28(1H,t),11.48(1H,s)。
m/z ESI M/2+H
+285.8。
实施例20N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-(吗啉-4-基甲基)-4-甲基-联苯-3-甲酰胺的制备
与实施例18的制备方法相同,除了用吗啡啉替代哌啶,制得化合物20。
1H-NMR(DMSO-d
6)δ:0.85(3H,t),1.73-1.91(1H,m),1.96-2.05(1H,m),2.11(3H,s),2.21(3H,s),2.24(3H,s),2.38(4H,br),3.01-3.08(2H,m),3.25-3.32(1H,m),3.48-3.55(4H,m),3.58(4H,br),3.67-3.77(1H,m),3.91-3.98(1H,m),4.29(2H,d),5.57-5.65(1H,m),5.87(1H,s),6.05-6.13(1H,m),6.48--6.58(1H,m),7.28(1H,d),7.39(2H,d),7.52(1H,d),7.62(2H,d),8.20-8.26(1H,t),11.48(1H,s)。
m/z ESI M+H
+612.3。
实施例21N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-3'-氯-4'-(吗啉-4-基甲基)-4-甲基-联苯-3-甲酰胺的制备
步骤1:4-溴-2-氯苄醇(21a)的制备
将4-溴-2-氯苯甲酸(1.5g,6.41mmol)溶解于20ml THF中。氮气氛下,在冰水浴中,滴加15mL 1M硼烷四氢呋喃溶液,并搅拌3h。将反应液慢慢倒入水中,用乙酸乙酯萃取,有机相用饱和氯化钠溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩,得粗品1.6g标题产物。其无需纯化,直接用于下一步反应。
步骤2:4-溴-2-氯苄溴(21b)的制备
将4-溴-2-氯苄醇(1.6g,7.34mmol)、三苯基膦(3.85g,14.68mol)溶于50mL二氯甲烷中。在氮气氛下,在冰水浴中,慢慢加入NBS(2.74g,15.41mmol),于该温搅拌30分钟。将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯100:0~60:1)纯化,得0.95g标题产物。
步骤3:4-(吗啉-4-基甲基)-4-溴-2-氯苯(21c)的制备将4-溴-2-氯苄溴(0.95g,3.38mmol)、吗啡啉(0.59g,6.67mmol)、碳酸钾(1.2g,8.45mmol)加到20mL DMF中,于70℃搅拌反应1h。将反应液冷却至室温,并慢慢倒入水中,用乙酸乙酯萃取。有机相用用饱和氯化钠溶液洗涤两次,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷:甲醇100:1~25:1)纯化,得到0.95g标题产物。
步骤4:4-(吗啉-4-基甲基)-2-氯-苯基-4-硼酸频哪醇酯(21d)的制备
将4-(吗啉-4-基甲基)-4-溴-2-氯苯(0.95g,3.28mmol)、联硼酸频那醇酯(1.25g,4.93mmol)、乙酸钾(0.97g,9.86mmol)、Pd(PPh
3)
2Cl
2(0.23g,0.33mmol)加入至20mL二氧六环中,氮气氛下,于80℃搅拌反应3小时。将反应液冷却却至室温,然后倒入水中,用乙酸乙酯萃取。有机相用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷:甲醇100:1~30:1),得到0.42g标题产物。
步骤5:N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-3'-氯-4'-(吗啉-4-基甲基)-4-甲基-联苯-3-甲酰胺的制备
将3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2-甲基-苯甲酰胺(化合物1)(100mg,0.19mmol)、4-(吗啉-4-基甲基)-2-氯-苯基-4-硼酸频哪醇酯(131mg,0.39mmol)、碳酸钠(51mg,0.49mmol)、Pd(PPh
3)
4(22mg,0.02mmol)加入至10mL 1,4-二氧六环和2mL水的混合溶剂中,氮气氛下,于80℃搅拌反应小时。将反应液冷却至室温,然后倒入水中,用乙酸乙酯萃取。有机相用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷:甲醇100:1~25:1)纯化,得66mg标题化合物。
1H-NMR(CDCl
3)δ:0.89(3H,t),1.70-1.95(1H,m),2.03-2.09(1H,m),2.18(3H,s),2.36(3H,s),2.42(3H,s),2.54(4H,br),3.02-3.08(2H,m),3.27-3.31(1H,m),3.53-3.95(10H,m),4.53(2H,d),5.63-5.71(1H,m),5.93(1H,s),6.32-6.42(2H,m),7.18-7.22(1H,br),7.33-7.41(3H,m),7.48-7.55(2H,m),11.30(1H,br)。
m/z ESI M+H
+646.3。
实施例22N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-3'-甲氧基-4'-(吗啉-4-基甲基)-4-甲基-联苯-3-甲酰胺的制备
与实施例21的制备方法相同,除了用4-溴-2-甲氧基苯甲酸替代4-溴-2-氯苯 甲酸,制得化合物22。
1H-NMR(DMSO-d
6)δ:0.84(3H,t),1.67-2.05(2H,m),2.11(3H,s),2.21(3H,s),2.24(3H,s),2.40(4H,br),3.01-3.08(2H,m),3.25-3.31(1H,m),3.45-3.80(9H,m),3.83-3.98(4H,m),4.30(2H,d),5.57-5.67(1H,m),5.86(1H,s),6.05-6.13(1H,m),6.48--6.58(1H,m),7.15-7.23(2H,m),7.30(1H,s),7.38(1H,d),7.52(1H,d),8.18-8.25(1H,br),11.47(1H,s)。
m/z ESI M+H
+642.3。
实施例23N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-3'-氟-4'-(吗啉-4-基甲基)-4-甲基-联苯-3-甲酰胺的制备
与实施例21的制备方法相同,除了用4-溴-2-氟苯甲酸替代4-溴-2-氯苯甲酸,制得化合物23。
1H-NMR(DMSO-d
6)δ:0.84(3H,t),1.65-2.05(2H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.40(4H,br),3.01-3.08(2H,m),3.25-3.31(1H,m),3.49-3.60(8H,m),3.67-3.78(1H,m),3.92-3.98(4H,m),4.30(2H,d),5.58-5.66(1H,m),5.87(1H,s),6.05-6.13(1H,m),6.48-6.58(1H,m),7.33(1H,s),7.45-7.60(4H,m),8.20-8.28(1H,br),11.48(1H,s)。
m/z ESI M+H
+630.3
实施例24N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-(吗啉-4-基)-4-甲基-联苯-3-甲酰胺的制备
与实施例21的步骤4和5的制备方法相同,除了用4-(吗啉-4-基)-溴苯替代 4-(吗啉-4-基甲基)-4-溴-2-氯苯,制得化合物24。
1H-NMR(CDCl
3)δ:0.89(3H,t),1.65-1.93(1H,m),1.98-2.03(1H,m),2.16(3H,s),2.35(3H,s),2.42(3H,s),3.03-3..08(2H,m),3.15-3.55(6H,m),3.65-3.95(7H,m),4.57(2H,d),5.61-5.69(1H,m),5.92(1H,s),6.32-6.42(2H,m),6.91-6.97(1H,t),7.10-7.25(2H,m),7.32-7.48(4H,m),11.95(1H,br)。
m/z ESI M+H
+598.3。
实施例25N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-(4-甲基-哌嗪-1-基)-4-甲基-联苯-3-甲酰胺的制备
与实施例24的制备方法相同,除了用4-(4-甲基-哌嗪-1-基)-溴苯替代4-(吗啉-4-基)-溴苯,制得化合物25。
1H-NMR(CDCl
3)δ:0.88(3H,t),1.69-2.05(2H,m),2.16(3H,s),2.37(3H,s),2.43(3H,s),2.62(3H,s),2.94(4H,br),3.03-3..08(2H,m),3.28-3.55(6H,m),3.65-3.95(3H,m),4.56(2H,d),5.61-5.69(1H,m),5.95(1H,s),6.32-6.42(2H,m),6.77(1H,d),6.88(1H,d),7.30-7.45(5H,m),12.20(1H,br)。
m/z ESI M+H
+611.5。
实施例26N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2-甲基-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-(6-乙氧基-嘧啶-4-基)-苯甲酰胺的制备
步骤1:3-{[3-乙氧酰基-2-甲基-5-(4,4,5,5-四甲基-[1,3,2]二氧硼戊烷-2-基)-苯基]-乙基-氨基}-吡咯烷-1-基羧酸叔丁醇酯(26a)的制备
将3-[(5-溴-3-乙氧基羰基-2-甲基-苯基)-乙基-氨基]-吡咯烷-1-基甲酸叔丁醇酯(1e)(1.6g,3.5mmol)、联硼酸频哪醇酯(1.34g,5.3mmol)、乙酸钾(1g,10.5mmol)、二(三苯基膦)二氯化钯(0.25g,0.35mmol)加入至20mL二氧六环中,氮气氛下,于85℃搅拌4小时。将反应液减压浓缩浓缩,然后加入水和乙酸乙酯萃取。有机相经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯40:1~3:1)纯化,得到1.7g产物。
步骤2:2-甲基-3-[(1-Boc-吡咯烷-3-基)-乙基-氨基]-5-(6-氯-嘧啶-4-基)-苯甲酸乙酯(26b)的制备
将3-{[3-乙氧酰基-2-甲基-5-(4,4,5,5-四甲基-[1,3,2]二氧硼戊烷-2-基)-苯基]-乙基-氨基}-吡咯烷-1-基羧酸叔丁醇酯(1.7g,3.4mmol)、4,6-二氯嘧啶(2g,13.5mmol)、四三苯基膦钯(0.4g,0.34mmol)、碳酸钾(0.94g,6.8mmol)加入至30mL乙腈和15mL水的混合溶剂中,氮气氛下,于85℃搅拌1.5小时。将反应液减压浓缩,然后加入水和乙酸乙酯萃取。有机相经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯40:1~3:1)纯化,得到0.98g产物。
步骤3:2-甲基-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-(6-乙氧基-嘧啶-4-基)-苯甲酸(26c)的制备
将2-甲基-3-[(1-Boc-吡咯烷-3-基)-乙基-氨基]-5-(6-氯-嘧啶-4-基)-苯甲酸乙酯(1.3g,2.66mmol)、氢氧化钠(0.32g,8mmol)加入至15mL乙醇和5mL水 的混合溶剂中,室温搅拌20小时,再回流3小时。将反应液减压浓缩,然后加入1M柠檬酸水溶液调节PH至酸性,然后用乙酸乙酯萃取。有机相经无水硫酸钠干燥,过滤,减压浓缩,得1.17g产物。
其余步骤与实施例1的制备方法相同,除了用化合物26c替代化合物1f,制得化合物26。
1H-NMR(DMSO-d
6)δ:0.84(3H,t),1.37(3H,t),1.69-1.98(2H,m),2.12(3H,s),2.22(3H,s),2.27(3H,s),2.98-3.08(2H,m),3.25-3.31(1H,m),3.49-3.58(2H,m),3.68-3.76(1H,m),3.92-3.98(1H,m),4.31(2H,d),4.44(2H,m),5.57-5.67(1H,m),5.87(1H,s),6.07-6.13(1H,m),6.46-6.59(1H,m),7.55(1H,d),7.84(1H,d),8.04(1H,s),8.25(1H,t),8.83(1H,s),11.49(1H,s)。
m/z ESI M+H
+559.2。
实施例27N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2-甲基-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-[6-(4-甲基-哌嗪-1-基)-嘧啶-4-基]-苯甲酰胺的制备
步骤1:2-甲基-3-[(1-Boc-吡咯烷-3-基)-乙基-氨基]-5-[6-(4-甲基-哌嗪-1-基)-嘧啶-4-基]-苯甲酸乙酯(27a)的制备
将2-甲基-3-[(1-Boc-吡咯烷-3-基)-乙基-氨基]-5-(6-氯-嘧啶-4-基)-苯甲酸乙酯(26b)(0.72g,1.5mmol)、N-甲基哌嗪(0.22g,2.2mmol)、碳酸钾(0.42g,3mmol)溶解于10mL DMF中,于60℃搅拌2小时。反应物加入水和乙酸乙酯萃取。有机相经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:二氯甲烷:甲醇100:1~25:1)纯化,得到0.4g产物。
其余步骤与实施例1的制备方法相同,除了用化合物27a代替化合物1e,制得化合物27。
1H-NMR(DMSO-d
6)δ:0.84(3H,t),1.71-2.03(2H,m),2.11(3H,s),2.22-2.26(9H, t),2.40(4H,br),2.98-3.08(2H,m),3.25-3.30(1H,m),3.49-3.58(2H,m),3.68-3.78(5H,m),3.88-3.92(1H,m),4.30(2H,d),5.57-5.67(1H,m),5.87(1H,s),6.05-6.12(1H,m),6.48-6.58(1H,m),7.30(1H,s),,7.84(1H,d),8.04(1H,d),8.25(1H,br),8.56(1H,s),11.49(1H,s)。
m/z ESI M+H
+613.3。
实施例28N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-噻吩-3-甲酰胺的制备
步骤1:2-氨基-4-甲基噻吩-3-甲酸乙酯(28a)的制备
将氰乙酸乙酯(30g,265mmol)、硫(8.5g,265mmol)、吗啡啉(34.63g,400mmol)和丙酮(15.37g,265mmol)加入到200mL乙醇中,于50℃搅拌24小时。将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯50:1~5:1)纯化,得到7.3g产物。
步骤2:2-碘-4-甲基噻吩-3-甲酸乙酯(28b)的制备
将2-氨基-4-甲基噻吩-3-甲酸乙酯(13g,70mmol)加入至碘化亚铜(26.7g,140mmol)、亚硝酸叔丁醇酯(10.8g,105mmol)的300mL乙腈悬浮液中,搅拌4小时。滤除不溶物,滤液减压浓缩,然后加入水和乙酸乙酯萃取。有机相经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯100:1~5:1)纯化得1.5g产物。
步骤3:2-碘5-硝基-4-甲基噻吩-3-甲酸乙酯(28c)的制备
2-碘-4-甲基噻吩-3-甲酸乙酯(1.37g,5.8mmol)溶于20mL醋酸酐中,冰水浴冷却下,滴加4mL硝酸,搅拌2小时。反应液加入水和乙酸乙酯萃取。有机层用氢氧化钠洗至碱性,经无水硫酸钠干燥,过滤,减压浓缩,得到粗产物,其未经纯化而直接用于下一步。
步骤4:2-氨基-3-甲基噻吩-4-羧酸乙酯(28d)的制备
将得到的2-碘-5-硝基-4-甲基噻吩-3-甲酸乙酯粗品、雷尼镍(1.6g)加入至35mL乙醇中,氢气氛下搅拌20小时。反应液过滤,滤液减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯50:1~5:1)纯化,得产物0.6g。
其余步骤与实施例1的制备方法相同,除了用化合物28d替代化合物1c,制得化合物28。
1H-NMR(DMSO-d
6)δ:0.86(3H,t),1.72-1.92(1H,m),1.96-2.05(1H,m),2.12(3H,s),2.13(3H,s),2.18(3H,s),2.82-2.95(2H,m),3.10-3.18(1H,m),3.45-3.75(4H,m),4.23(2H,d),5.60-5.68(1H,m),5.87(1H,s),6.08-6.14(1H,m),6.48-6.56(1H,dd),7.71(1H,s),8.01(1H,br),11.50(1H,s)。
m/z ESI M+H
+443.2。
实施例29N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-4'-(吡咯烷-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例18的制备方法相同,除了用四氢吡咯替代哌啶,制得化合物29。
1H-NMR(DMSO-d
6)δ:0.84(3H,t),1.69-2.02(6H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.90-3.08(6H,m),3.25-3.31(1H,m),3.49-3.58(2H,m),3.68-3.78(1H,m),3.90-3.98(1H,m),4.15(2H,s),4.29(2H,d),5.57-5.65(1H,m),5.87(1H,s),6.07-6.13(1H,m),6.48-6.59(1H,m),7.31(1H,d),7.55(1H,d),7.58(2H,d),7.72(2H,d),8.25(1H,t),11.49(1H,s)。
m/z ESI M+H
+596.4。
实施例30N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基3'-(吗啉-4-基)甲基-[1,1'-联苯]-3-甲酰胺的制备
与实施例20的制备方法相同,除了用3-羟甲基苯硼酸频那醇酯替代4-羟甲基苯硼酸频那醇酯,制得化合物30。
1H-NMR(DMSO-d
6)δ:0.85(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.38(4H,br),3.01-3.08(2H,m),3.24-3.31(1H,m),3.50-3.81(9H,m),3.89-3.96(1H,m),4.30(2H,d),5.58-5.67(1H,m),5.87(1H,s),6.07-6.13(1H,m),6.48-6.58(1H,m),7.27-7.33(2H,m),7.42(1H,t),7.51-7.58(3H,m),8.25(1H,t),11.48(1H,s)。
m/z ESI M+H
+613.2。
实施例31N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-3'-(哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例18的制备方法相同,除了用3-羟甲基苯硼酸频那醇酯替代4-羟甲基苯硼酸频那醇酯,制得化合物31。
1H-NMR(DMSO-d
6)δ:0.85(3H,t),1.55-2.02(8H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.36-2.80(4H,m),3.01-3.08(2H,m),3.26-3.32(1H,m),3.50-4.02(6H,m),4.29(2H,d),5.58-5.67(1H,m),5.87(1H,s),6.07-6.13(1H,m),6.48-6.58(1H,m),7.32(1H,d),7.40-7.53(2H,m),7.56-7.75(3H,m),8.25(1H,t),11.49(1H,s)。六个氢与溶剂峰重合。
m/z ESI M+H
+610.3。
实施例32N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-4'-(4-羟基哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例18的制备方法相同,除了用4-羟基哌啶替代哌啶,制得化合物32。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.60-2.05(6H,m),2.11(3H,s),2.21(3H,s),2.25(3H,d),2.84-3.15(6H,m),3.22-3.28(1H,m),3.48-3.81(4H,m),3.90-3.98(1H,m),4.20-4.32(4H,m),5.02(1H,br),5.58-5.67(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.48-6.58(1H,m),7.32(1H,s),7.56(1H,s),7.73(4H,br),8.26(1H,t),10.97(1H,br),11.50(1H,s)。
m/z ESI M+H
+626.4。
实施例33N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-4'-{[(2-羟基乙基)-甲基-氨基]-甲基}-[1,1'-联苯]-3-甲酰胺的制备
与实施例18的制备方法相同,除了用N-甲基乙醇胺替代哌啶,制得化合物33。
1H-NMR(DMSO-d6)δ:0.85(3H,t),1.60-2.05(2H,m),2.11(3H,s),2.21(3H,s),2.25(3H,d),2.37(3H,s),2.67(2H,t),2.97-3.07(2H,m),3.22-3.28(1H,m),3.48-3.81(7H,m),3.90-3.98(1H,m),4.29(2H,d),4.68(1H,br),5.58-5.67(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.48-6.58(1H,m),7.30(1H,s),7.49-7.54(3H,m),7.67(2H,d),8.26(1H,t),11.49(1H,s)。
m/z ESI M+H
+600.3。
实施例34N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例18的制备方法相同,除了用1-Boc-4-哌啶酮替代1-Boc-3-吡咯烷酮,制得化合物34。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.35-1.82(10H,m),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.32(1H,m),2.41-2.80(4H,m),2.99-3.14(4H,m),3.63(2H,s),3.96-4.06(1H,m),4.27-4.41(3H,m),5.62-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.75-6.82(1H,m),7.26(1H,d),7.43(1H,d),7.53(2H,d),7.67(2H,d),8.22(1H,t),11.48(1H,s)。
m/z ESI M+H
+624.3。
实施例35N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(吗啉-4-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用吗啉替代哌啶,制得化合物35。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.35-1.48(2H,m),1.75-1.82(2H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.37(4H,br),2.61-2.71(1H,m),2.97-3.12(4H,m),3.49(2H,s),3.59(4H,br),3.97-4.05(1H,m),4.27-4.39(3H,m),5.62-5.66(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.74-6.82(1H,m),7.23(1H,d),7.36-7.42(3H,m),7.59(2H,d),8.20(1H,t),11.47(1H,s)。
m/z ESI M+H
+626.3。
实施例36N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-2-甲基-5-[6-(吗啉-4-基)-嘧啶-4-基]-苯甲酰胺
与实施例27的制备方法相同,除了用吗啉替代N-甲基哌嗪,制得化合物36。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.22(3H,s),2.25(3H,s),3.01-3.08(2H,m),3.25-3.31(1H,m),3.49-3.57(2H,m),3.58-3.65(1H,m),3.71(8H,s),3.86-3.95(1H,m),4.30(2H,d),5.57-5.66(1H,m),5.87(1H,s),6.05-6.13(1H,m),6.50-6.58(1H,m),7.31(1H,d),7.84(1H,d),8.03(1H,s),8.22(1H,t),8.59(1H,s),11.48(1H,s)。
m/z ESI M+H
+600.3。
实施例37N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-2-甲基-5-[2-(吗啉-4-基)嘧啶-4-基]-苯甲酰胺的制备
与实施例36的制备方法相同,除了用2,4-二氯嘧啶替代4,6-二氯嘧啶,制得化合物37。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),3.01-3.08(2H,m),3.24-3.31(1H,m),3.49-3.58(2H,m),3.67-3.78(9H,m),3.88-3.95(1H,m),4.29(2H,d),5.57-5.67(1H,m),5.87(1H,s),6.05-6.11(1H,m),6.48-6.58(1H,m),7.28(1H,t),7.77(1H,d),7.95(1H,d),8.25(1H,t),8.44(1H,d),11.48(1H,s)。
m/z ESI M+H
+600.3。
实施例38N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-2-甲基-5-[2-(4-甲基-哌嗪-1-基)嘧啶-4-基]-苯甲酰胺的制备
与实施例27的制备方法相同,除了用2,4-二氯嘧啶替代4,6-二氯嘧啶,制得化合物38。
1H-NMR(DMSO-d6)δ:0.83(3H,t),1.65-2.02(2H,m),2.12(3H,s),2.22(3H,s),2.26(3H,s),2.32(3H,s),2.55(4H,br),2.99-3.08(2H,m),3.24-3.31(1H,m),3.50-3.57(2H,m),3.65-3.98(7H,m),4.30(2H,d),5.58-5.67(1H,m),5.88(1H,s),6.07-6.13(1H,m),6.48-6.58(1H,m),7.27(1H,t),7.77(1H,d),7.94(1H,d),8.27(1H,t),8.43(1H,d),11.50(1H,s)。
m/z ESI M+H
+613.2。
实施例39N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-4'-(吗啉-4-基甲酰基)-[1,1'-联苯]-3-甲酰胺的制备
步骤1:4-溴苯甲酰吗啉的制备
4-溴苯甲酸(2g,10mmol)悬浮于20mL二氯甲烷中,加入草酰氯(2.55g,20mmol)、1滴DMF,回流反应1小时,减压除去二氯甲烷、草酰氯,残余物溶于20mL二氯甲烷中,冰水浴冷却下,加入吗啉(2.61g,30mmol),搅拌30分钟后,加入水、二氯甲烷萃取,二氯甲烷层拌样,通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯20:1~2:1)纯化,得产物4-溴苯甲酰吗啉1.9g。
步骤2:5-[(1-Boc-吡咯烷-3-基)-乙基-氨基]-4-甲基-4'-(吗啉-4-基甲酰基)-[1,1'-联苯]-3-甲酸乙酯的制备
3-{[3-乙氧酰基-2-甲基-5-(4,4,5,5-四甲基-[1,3,2]二氧硼戊烷-2-基)-苯基]-乙基-氨基}-吡咯烷-1-基羧酸叔丁醇酯26a(0.35g,0.7mmol)、4-溴苯甲酰吗啉(0.17g,0.63mmol)、Pd(PPh
3)
4(0.08g,0.07mmol)、碳酸钾(0.19g,1.4mmol)加到15mL乙腈、5mL水中,氮气置换后,回流反应18小时。将反应液减压浓缩,残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯20:1~1:1)纯化,得产物0.32g。
其余步骤与实施例27的制备方法相同,除了用化合物39a代替化合物27a,制得化合物39。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.99-3.12(2H,m),3.25-3.31(1H,m),3.41-3.81(10H,m),3.92-3.98(1H,m),4.30(2H,d),5.58-5.65(1H,m),5.87(1H,s),6.05-6.12(1H,m),6.48-6.58(1H,m),7.33(1H,d),7.50(2H,d),7.57(1H,d),7.75(2H,d),8.25(1H,t),11.49(1H,s)。
m/z ESI M+H
+626.2。
实施例40N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-3'-(吗啉-4-基甲酰基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例39的制备方法相同,除了用3-溴苯甲酸替代4-溴苯甲酸,制得化合物40。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.22(3H,s),2.26(3H,s),3.01-3.08(2H,m),3.25-3.31(1H,m),3.42-3.80(11H,m),3.92-3.98(1H,m),4.30(2H,d),5.57-5.66(1H,m),5.87(1H,s),6.07-6.13(1H,m),6.50-6.58(1H,m),7.31(1H,t),7.39(1H,d),7.51-7.58(2H,m),7.67-7.70(1H,m),7.73-7.78(1H,m),8.25(1H,t),11.48(1H,s)。
m/z ESI M+H
+626.2。
实施例41N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-4'-(4-甲基-哌嗪-1-基甲酰基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例39的制备方法相同,除了用N-甲基哌嗪代替吗啉,制得化合物41。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.21(6H,s),2.25-2.43(7H,m),3.01-3.08(2H,m),3.24-3.31(1H,m),3.45-3.80(7H,m),3.95-3.99(1H,m),4.30(2H,d),5.57-5.67(1H,m),5.87(1H,s),6.07-6.12(1H,m),6.50-6.58(1H,m),7.32(1H,d),7.45-7.58(2H,d),7.57(1H,d),7.74(2H,d),8.25(1H,t),11.48(1H,s)。
m/z ESI M+H
+639.2。
实施例42N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-3'-(4-甲基-哌嗪-1-基甲酰基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例41的制备方法相同,除了用3-溴苯甲酸替代4-溴苯甲酸,制得化合物42。
1H-NMR(DMSO-d6)δ:0.85(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.21(6H,d),2.26(3H,s),2.38(4H,br),3.01-3.08(2H,m),3.26-3.32(1H,m),3.50-3.81(7H,m),3.91-3.98(1H,m),4.30(2H,d),5.58-5.67(1H,m),5.87(1H,s),6.07-6.13(1H,m),6.48-6.58(1H,m),7.31-7.38(2H,m),7.50-7.57(2H,m),7.64(1H,d),7.76(1H,m),8.25(1H,t),11.49(1H,s)。
m/z ESI M+H
+639.2。
实施例43N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-(吗啉-4-基甲酰基)-3'-氯-4-甲基-[1,1'-联苯]-3-甲酰胺的制备
与实施例39的制备方法相同,除了用2-氯-4-溴苯甲酸代替4-溴苯甲酸,制得化合物43。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.34(3H,s),3.01-3.08(2H,m),3.19(2H,br),3.26-3.32(1H,m),3.48-3.58(4H,m),3.62-3.80(5H,m),3.93-4.01(1H,m),4.29(2H,d),5.58-5.67(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.48-6.58(1H,m),7.35(1H,s),7.47(1H,d),7.62(1H,s),7.72-7.78(1H,m),7.86(1H,m),8.24(1H,t),11.49(1H,s)。
m/z ESI M+H
+660.2。
实施例44N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-(4-甲基-哌嗪-1-基甲酰基)-3'-氯-4-甲基-[1,1'-联苯]-3-甲酰胺的制备
实施例41的制备方法相同,除了用2-氯-4-溴苯甲酸代替4-溴苯甲酸,制得化合物44。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.22(6H,d),2.26-2.40(7H,m),3.01-3.08(2H,m),3.17(2H,br),3.26-3.32(1H,m),3.48-3.75(5H,m),3.93-4.02(1H,m),4.30(2H,d),5.58-5.67(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.48-6.58(1H,m),7.35(1H,s),7.43(1H,d),7.62(1H,s),7.70-7.75(1H,m),7.85(1H,m),8.28(1H,t),11.49(1H,s)。
m/z ESI M/2+H
+337.2。
实施例45N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-4'-(吗啉-4-基磺酰基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例39的制备方法相同,除了用4-溴苯磺酰氯替代4-溴苯甲酸,制得化合物45。
1H-NMR(DMSO-d6)δ:0.85(3H,t),1.65-2.05(2H,m),2.12(3H,s),2.22(3H,s),2.27(3H,s),2.90(4H,br),3.02-3.07(2H,m),3.25-3.33(1H,m),3.50-3.81(7H,m),3.90-3.98(1H,m),4.31(2H,d),5.58-5.67(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.48-6.58(1H,m),7.40(1H,d),7.66(1H,d),7.79(2H,d),7.97(2H,d),8.30(1H,t),11.54(1H,s)。
m/z ESI M+H
+662.3。
实施例46N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-4'-(4-甲基-哌嗪-1-基磺酰基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例45的制备方法相同,除了用N-甲基哌嗪替代吗啉,制得化合物46。
1H-NMR(DMSO-d6)δ:0.85(3H,t),1.65-2.05(2H,m),2.11(3H,s),2.14(3H,s), 2.21(3H,s),2.26(3H,s),2.37(4H,br),2.91(4H,br),3.02-3.07(2H,m),3.25-3.33(1H,m),3.50-3.55(2H,m),3.68-3.81(1H,m),3.91-3.98(1H,m),4.29(2H,d),5.58-5.67(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.48-6.58(1H,m),7.38(1H,d),7.64(1H,d),7.78(2H,d),7.96(2H,m),8.29(1H,t),11.53(1H,s)。
m/z ESI M+H
+675.3。
实施例47N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-4'-(2-二甲氨基乙氧基)-[1,1'-联苯]-3-甲酰胺的制备
步骤1:[2-(4-溴苯氧基)-乙基]-二甲基胺的制备
4-溴苯酚(1g,5.8mmol)、(2-氯乙基)二甲胺盐酸盐(1.25g,8.7mmol)、碳酸铯(5.67g,17.4mmol)加到5mLDMA中,于90℃加热2小时。冷却至室温后,向反应液中加入乙酸乙酯和水萃取。将有机层通过无水硫酸钠干燥,过滤,减压浓缩,得到产物1.3g。
其余步骤与实施例39的制备方法相同,除了用[2-(4-溴苯氧基)-乙基]-二甲基胺代替4-溴苯甲酰吗啉,制得化合物47。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.05(2H,m),2.11(3H,s),2.21(3H,d),2.23(3H,s),2.34(6H,s),2.80(2H,t),2.96-3.04(2H,m),3.22-3.31(1H,m),3.48-3.55(2H,m),3.68-3.76(1H,m),3.88-3.97(1H,m),4.14(2H,t),4.29(2H,d),5.58-5.66(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.48-6.58(1H,m),7.03(2H,d),7.24(1H,s),7.47(1H,s),7.56-7.62(2H,m),8.24(1H,t),11.49(1H,s)。
m/z ESI M+H
+600.4。
实施例48N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4-甲基-4'-[2-(吗啉-4-基)-乙氧基]-[1,1'-联苯]-3-甲酰胺的制备
与实施例47的制备方法相同,除了用N-(2-氯乙基)吗啉盐酸盐替代(2-氯乙基)二甲胺盐酸盐,制得化合物48。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.05(2H,m),2.11(3H,s),2.21(3H,s),2.23(3H,s),2.47(4H,br),2.70(2H,t),2.99-3.05(2H,m),3.22-3.31(1H,m),3.48-3.61(6H,m),3.67-3.77(1H,m),3.88-3.97(1H,m),4.12(2H,t),4.29(2H,d),5.58-5.66(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.47-6.59(1H,m),7.02(2H,d),7.23(1H,s),7.47(1H,s),7.55-7.62(2H,m),8.20-8.28(1H,m),11.52(1H,s)。
m/z ESI M+H
+642.3。
实施例49N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-{3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-氨甲酰基]-4-甲基-苯基}-噻吩[3,2-b]噻吩-2-甲酰胺的制备
步骤1:N-(2-羟基乙基)-N-甲基-5-溴-噻吩[3,2-b]并噻吩-2-甲酰胺的制备
5-溴-噻吩[3,2-b]并噻吩-2-甲酸(0.5g,1.9mmol)悬浮于25mL二氯甲烷中,加入草酰氯(0.48g,3.8mmol)、1滴DMF,回流反应1小时,减压除去二氯甲烷、草酰氯,残余物溶于20mL二氯甲烷,冰水浴冷却,加入N-甲基乙醇胺(0.29g,3.8mmol),搅拌15分钟。将反应液通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯10:1~2:1)纯化,得产物0.37g。
其余步骤与实施例39的制备方法相同,除了用N-(2-羟基乙基)-N-甲基-5-溴-噻吩[3,2-b]并噻吩-2-甲酰胺代替4-溴苯甲酰吗啉,制得化合物49。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.05(2H,m),2.12(6H,d),2.19(3H,s),2.21(6H,s),3.01-3.06(2H,m),3.22-3.31(1H,m),3.48-3.58(2H,m),3.68-3.78(1H,m),3.88-3.97(1H,m),4.29(4H,d),5.58-5.66(1H,m),5.87(2H,d),6.06-6.13(1H,m),6.48-6.58(1H,m),7.27(2H,s),7.60(1H,s),7.98(1H,d),8.16(1H,s),8.25-8.33(1H,m),8.56(1H,t),11.53(2H,s)。
m/z ESI M+H
+753.2。
实施例50N-(2-羟基乙基)-N-甲基-5-{3-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-5-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-氨甲酰基]-4-甲基-苯基}-噻吩[3,2-b]噻吩-2-甲酰胺的制备
步骤1:4-{[3-苄氧酰基-2-甲基-5-(4,4,5,5-四甲基-[1,3,2]二氧硼戊烷-2-基)-苯基]-乙基-氨基}-哌啶-1-基羧酸叔丁醇酯(50a)的制备
与中间体26a的制备方法相同,除了用4-[(3-苄氧酰基-2-甲基-5-溴-苯基)-乙基-氨基]-哌啶-1-基羧酸叔丁醇酯(制备方法与中间体1e相同,除了用溴化苄替代溴乙烷,用1-Boc-4-哌啶酮替代1-Boc-3-吡咯烷酮)替代1e,制得标题化合物。
步骤2:N-(2-羟基乙基)-N-甲基-5-{3-[(1-Boc-哌啶-4-基)-乙基-氨基]-5-(苄氧酰基)-4-甲基-苯基}-噻吩[3,2-b]噻吩-2-甲酰胺的制备
与中间体39a的制备方法相同,除了用4-{[3-苄氧酰基-2-甲基-5-(4,4,5,5-四甲基-[1,3,2]二氧硼戊烷-2-基)-苯基]-乙基-氨基}-哌啶-1-基羧酸叔丁醇酯替代3-{[3-乙氧酰基-2-甲基-5-(4,4,5,5-四甲基-[1,3,2]二氧硼戊烷-2-基)-苯基]-乙基-氨基}-吡咯烷-1-基羧酸叔丁醇酯26a,用N-(2-羟基乙基)-N-甲基-5-溴-噻吩[3,2-b]并噻吩-2-甲酰胺(49a)代替4-溴苯甲酰吗啉,制得化合物50b。
步骤3:N-(2-羟基乙基)-N-甲基-5-{3-[(1-Boc-哌啶-4-基)-乙基-氨基]-5-羧基-4-甲基-苯基}-噻吩[3,2-b]噻吩-2-甲酰胺的制备
化合物50b(0.35g,0.5mmol)溶于15mL甲醇中,加入10%的Pd-C(30mg),氢气置换后室温搅拌3小时,滤除Pd-C,将滤液减压浓缩,得产物0.29g。
其余步骤与实施例26的制备方法相同,除了用化合物50c代替化合物26c,制得化合物50。
1H-NMR(DMSO-d6)δ:0.83(3H,t),1.38-1.49(2H,m),1.75-1.82(2H,m),2.12(3H,s),2.23(6H,d),2.66(1H,t),2.99-3.25(7H,m),3.57-3.67(4H,m),4.02(1H,d),4.29(2H,d),4.36(1H,d),4.81(1H,br),5.63-5.68(1H,m),5.88(1H,s),6.04-6.11(1H,m),6.75-6.82(1H,m),7.24(1H,d),7.50(1H,d),7.94(2H,d),8.28(1H,t),11.50(1H,s)。
m/z ESI M+H
+690.3。
实施例51N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-2-甲基-5-[3-(吗啉-4-基)-丙炔-1-基]-苯甲酰胺的制备
步骤1、2:5-(3-羟基-丙-1-炔基)-2-甲基-3-氨基苯甲酸乙酯(51b)的合成
5-溴-2-甲基-3-硝基苯甲酸乙酯(6g,20.8mmol)、丙炔醇(1.75g,31.2mmol)、Pd(PPh
3)
3Cl
2(0.73g,1.04mmol)加到40mL三乙胺中,氮气置换,于100℃加热1小时。将反应液冷却至室温,加入60mL乙酸乙酯稀释,用稀盐酸洗去三乙胺。乙酸乙酯层浓缩后溶于100mL乙醇、40mL水中,加入铁粉(4.65g,83mmol)、氯化铵(4.5g,83mmol),于60℃加热2.5小时。滤除铁泥,滤液减压浓缩,加入乙酸乙酯、水萃取。有机层经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯100:1~5:1)纯化,得产物0.83g。
步骤3、4:3-[(1-Boc-吡咯烷-3-基)-乙基-氨基]-2-甲基-5-(3-羟基丙炔-1-基)-苯甲酸乙酯(51d)的合成
与实施例1中1e的制备方法相同,除了用化合物51b替代化合物1c,制得化合物51d。
步骤5、6:3-[(1-Boc-吡咯烷-3-基)-乙基-氨基]-2-甲基-5-[3-(吗啉-4-基)丙炔-1-基]-苯甲酸乙酯(51f)的合成
与实施例18中18b的制备方法相同,除了用化合物51d替代化合物17a,用吗啉替代哌啶,制得化合物51f。
其余步骤与实施例39的制备方法相同,除了用化合物51f代替化合物39a,制得化合物51。
1H-NMR(DMSO-d6)δ:0.80(3H,t),1.65-1.95(2H,m),2.11(3H,s),2.19(3H,s),2.20(3H,d),2.94-3.00(2H,m),3.22-3.28(1H,m),3.48-3.55(4H,m),3.58-3.72(5H,m), 3.77-3.85(1H,m),4.26(2H,d),5.58-5.67(1H,m),5.86(1H,s),6.06-6.14(1H,m),6.48-6.58(1H,m),7.04(1H,s),7.32(1H,d),8.22(1H,t),11.47(1H,s)。
m/z ESI M+H
+560.2。
实施例52N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-2-甲基-5-[6-(吗啉-4-基)-吡啶-3-基]-苯甲酰胺的制备
步骤1:2-(吗啉-4-基)-5-溴吡啶的合成
将2,5-二溴吡啶(1.2g,5mmol)加到3mL吗啉中,氮气置换后,于100℃加热4小时。将反应液冷却至室温,然后加入水乙酸乙酯萃取。将乙酸乙酯层减压浓缩,残余物经柱层析色谱法(洗脱剂:二氯甲烷:甲醇100:1~40:1)纯化,得产物0.8g。
其余步骤与实施例47的制备方法相同,除了用2-(吗啉-4-基)-5-溴吡啶代替[2-(4-溴苯氧基)-乙基]-二甲基胺,制得化合物52。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.05(2H,m),2.11(3H,s),2.21(3H,s),2.23(3H,s),3.01-3.06(2H,m),3.22-3.31(1H,m),3.48-3.58(6H,m),3.68-3.73(5H,m),3.88-3.97(1H,m),4.29(2H,d),5.58-5.66(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.47-6.59(1H,m),6.93(1H,d),7.25(1H,s),7.50(1H,s),7.88-7.94(1H,m),8.25-8.33(1H,m),8.45-8.49(1H,m),11.49(1H,s)。
m/z ESI M+H
+599.3。
实施例53N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-2-甲基-5-[6-(4-甲基哌嗪-1-基)-吡啶-3-基]-苯甲酰胺的制备
与实施例52的制备方法相同,除了用N-甲基哌嗪替代吗啉,制得化合物53。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.05(2H,m),2.11(3H,s),2.21(3H,s),2.22(3H,s),2.36(3H,s),2.63(4H,br),2.99-3.05(2H,m),3.22-3.31(1H,m),3.48-3.67(7H,m),3.88-3.97(1H,m),4.28(2H,d),5.58-5.66(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.47-6.59(1H,m),6.94(1H,d),7.24(1H,s),7.49(1H,s),7.85-7.91(1H,m),8.18-8.25(1H,m),8.44-8.46(1H,m),11.51(1H,s)。
m/z ESI M+H
+612.3。
实施例54N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-2-甲基-5-(1-甲基-1H-吡唑-4-基)-苯甲酰胺的制备
与实施例39的制备方法相同,除了用1-甲基4-溴吡唑替代4-溴苯甲酰吗啉,制得化合物54。
1H-NMR(DMSO-d6)δ:0.83(3H,t),1.65-2.05(2H,m),2.11(3H,s),2.17(3H,d),2.21(3H,s),2.96-3.04(2H,m),3.22-3.31(1H,m),3.50-3.55(2H,m),3.68-3.74(1H,m),3.85-3.95(4H,m),4.28(2H,d),5.58-5.66(1H,m),5.87(1H,s),6.06-6.13(1H,m),6.48-6.58(1H,m),7.20(1H,s),7.45(1H,s),7.86(1H,d),8.08-8.18(2H,m),11.48(1H,s)。
m/z ESI M+H
+517.3。
实施例55N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4,3'-二甲基-[1,1'-联苯]-3-甲酰胺的制备
与实施例39的制备方法相同,除了用4-溴甲苯替代4-溴苯甲酰吗啉,制得化合物55。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.21(3H,d),2.24(3H,d),2.34(3H,s),3.01-3.08(2H,m),3.26-3.32(1H,m),3.48-3.57(2H,m),3.68-3.77(1H,m),3.91-3.98(1H,m),4.29(2H,d),5.57-5.67(1H,m),5.86(1H,s),6.06-6.13(1H,m),6.48-6.58(1H,m),7.26-7.29(3H,m),7.48-7.58(3H,m),8.24(1H,t), 11.48(1H,s)。
m/z ESI M+H
+527.2。
实施例56 3'-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-甲酰基]-5'-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-甲基-[1,1'-联苯]-4-甲酸乙酯的制备
与实施例50的制备方法相同,除了用4-溴苯甲酸乙酯替代N-(2-羟基乙基)-N-甲基-5-溴-噻吩[3,2-b]并噻吩-2-甲酰胺,用1-Boc-3-吡咯烷酮代替1-Boc-4-哌啶酮,制得化合物56。
1H-NMR(DMSO-d6)δ:0.85(3H,t),1.34(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.21(3H,s),2.26(3H,s),3.01-3.08(2H,m),3.24-3.31(1H,m),3.48-3.81(2H,m),3.68-3.78(1H,m),3.91-3.98(1H,m),4.28-4.37(4H,m),5.58-5.67(1H,m),5.87(1H,s),6.07-6.13(1H,m),6.48-6.58(1H,m),7.37(1H,d),7.62(1H,d),7.85(2H,d),8.03(2H,d),8.30(1H,t),11.49(1H,s)。
m/z ESI M+H
+585.2。
实施例57 3'-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-甲酰基]-5'-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-4'-甲基-[1,1'-联苯]-4-甲酸的制备
步骤1:3-({4'-羧基-5-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-酰基]-4-甲基-联苯-3-基}-乙基-氨基)-吡咯烷-1-甲酸叔丁醇酯的制备
3-({4'-乙氧酰基-5-[(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-氨甲酰基]-4-甲基-联苯-3-基}-乙基-氨基)-吡咯烷-1-甲酸叔丁醇酯56a(0.33g,0.5mmol)、氢氧化钠(0.1g,2.5mmol)溶于3mL乙醇、1mL水中搅拌过夜,加入1M柠檬酸水溶液20mL、乙酸乙酯萃取。将乙酸乙酯层浓缩,得到化合物57a。其未经纯化直接用 于下一步。
其余步骤与实施例56的制备方法相同,除了用化合物57a替代化合物56a,制得化合物57。
1H-NMR(DMSO-d6)δ:0.85(3H,t),1.65-2.02(2H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),3.01-3.08(2H,m),3.26-3.32(1H,m),3.50-3.58(2H,m),3.68-3.78(1H,m),3.91-3.98(1H,m),4.29(2H,d),5.58-5.67(1H,m),5.87(1H,s),6.07-6.13(1H,m),6.48-6.58(1H,m),7.31(1H,d),7.56(1H,d),7.62(2H,d),7.97(2H,d),8.27(1H,t),11.49(1H,s)。
m/z ESI M+H
+557.3。
实施例58 3-{[2-(丙烯酰基-甲基-氨基)-乙基]-乙基-氨基}-5-溴-N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-2-甲基-苯甲酰胺的制备
步骤1:5-溴-3-[2-(Boc-甲基-氨基)-乙基氨基]-2-甲基-苯甲酸乙酯的制备
将2-(Boc-甲基-氨基)-乙醇(0.19g,1mmol)溶于4mL二氯甲烷中,加入戴斯-马丁氧化剂(0.64g,1.5mmol)室温搅拌1小时。向反应液中加入3mL饱和硫代硫酸钠溶液、5mL饱和碳酸氢钠溶液,搅拌至澄清。然后,加入20mL二氯甲烷萃取,将二氯甲烷层减压浓缩。残余物溶于5mL醋酸中,加入5-溴-3-氨基-2-甲基苯甲酸乙酯1c(0.26g,1mmol),分批加入硼氢化钠(0.22g,6mmol),室温搅拌0.5小时。向反应液中加入水和乙酸乙酯萃取。乙酸乙酯层用氢氧化钠水溶液洗涤,有机层经无水硫酸钠干燥,过滤,滤液减压浓缩。残余物通过柱层析色谱法(洗脱剂:石油醚:乙酸乙酯30:1~3:1)纯化,得0.3g标题产物。
其余步骤与实施例1的制备方法相同,除了用化合物58a代替化合物1d,制得化合物58。
1H-NMR(DMSO-d
6)δ:0.86(3H,t),2.08(3H,d),2.11(3H,s),2.19(3H,s),2.83-3.04(5H,m),3.06-3.12(2H,m),3.40(2H,t),4.25(2H,d),5.52-5.67(1H,m),5.87(1H,s),6.00-6.12(1H,m),6.40-6.71(1H,m),7.06(1H,d),7.30(1H,d),8.22(1H,t),11.50(1H,s)。
m/z ESI M+H
+503.2。
实施例59N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-[(1-丙烯酰基-吡咯烷-3-基)-(2,2,2-三氟乙基)-氨基]-2-甲基-5-溴苯甲酰胺的制备
步骤1:3-(5-溴-3-乙氧基羰基-2-甲基-苯基氨)-吡咯烷-1-基甲酸9H-芴-9-基甲基酯的制备
将3-(5-溴-3-乙氧基羰基-2-甲基-苯基氨)-吡咯烷-1-基甲酸叔丁醇酯1b(1.6g,3.75mmol)溶解于20mL二氯甲烷中,加入2.5mL三氟乙酸,回流20小时后,冷却至室温,加入饱和碳酸氢钠溶液萃取。冰水浴冷却,向二氯甲烷层加入FmocCl(1.06g,4.1mmol)、1mL N-甲基吗啉,加完搅拌0.5小时。向反应液中加入二氯甲烷、1M稀盐酸萃取。有机层经无水硫酸钠干燥,过滤,滤液减压浓缩。残余物通过柱层析色谱法纯化(洗脱剂:石油醚:乙酸乙酯50:1~5:1),得1.48g标题产物。
步骤2:3-[(5-溴-3-乙氧基羰基-2-甲基-苯基)-(2,2,2-三氟乙基)-氨基]-吡咯烷-1-基甲酸9H-芴-9-基甲基酯的制备
将化合物59a(2.2g,4mmol)溶于20mL三氟乙酸中,20小时内分批加入硼氢化钠(3.7g,100mmol),加完后再搅拌5小时。向反应液中加入乙酸乙酯、水萃取,乙酸乙酯层用氢氧化钠水溶液洗去残留三氟乙酸后,经无水硫酸钠干燥,过滤,减压浓缩。残余物通过柱层析色谱法纯化(洗脱剂:石油醚:乙酸乙酯20:1~5:1),得产物0.86g。
步骤3:3-[(5-溴-3-乙氧基羰基-2-甲基-苯基)-(2,2,2-三氟乙基)-氨基]-吡咯烷-1-基甲酸叔丁醇酯的制备
化合物59b(0.86g,1.3mmol)溶于2.5mL DMF中,加入0.5mL哌啶搅拌20分钟。向反应液中加入乙酸乙酯、水萃取。将乙酸乙酯层减压浓缩,残余物溶于25mL二氯甲烷中,加入(Boc)
2O(1.75g,8mmol)搅拌15分钟。直接加入硅胶蒸干拌样,柱层析色谱法纯化(洗脱剂:石油醚:乙酸乙酯80:1~10:1),得0.4g标题产物。
其余步骤与实施例1的制备方法相同,除了用化合物59c代替化合物1e,制得化合物59。
1H-NMR(DMSO-d6)δ:1.75-2.05(2H,m),2.11(3H,s),2.15(3H,s),2.19(3H,s),3.15-3.26(1H,m),3.42-3.55(2H,m),3.63-3.81(2H,m),3.85-3.96(2H,m),4.25(2H,d),5.62-5.68(1H,m),5.87(1H,s),6.07-6.13(1H,m),6.48-6.58(1H,m),7.20(1H,d),7.67(1H,d),8.27(1H,t),11.49(1H,s)。
m/z ESI M+H
+569.1。
实施例60N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-[(1-丙烯酰基-吡咯烷-3-基)-乙基-氨基]-5-溴-4-氟-2-甲基-苯甲酰胺的制备
步骤1:5-溴-4-氟-2-甲基苯甲酸(60a)的制备
向30mL溴素中加入4-氟-2-甲基苯甲酸(10g,65mmol)、铁粉(0.56g,10mmol),搅拌30小时,反应液倒入400mL水中,过滤收集析出的固体,固体溶于90mL甲醇中,滴加250mL稀盐酸,固体析出,过滤收集得到粗品60a(含异构体3-溴-4-氟-2-甲基苯甲酸)12.7g,未经纯化直接进行下一步。
与实施例2的制备方法相同,除了用60a替代5-氟-2-甲基苯甲酸,制得化合物60。
1H-NMR(CDCl
3)δ:0.92(3H,t),1.64-2.05(2H,m),2.28(3H,s),2.33(3H,s),2.40(3H,s),3.02-3.10(2H,m),3.48-4.02(5H,m),4.53(2H,d),5.61-5.70(1H,m),5.99(1H,s),6.36-6.44(2H,m),7.22(1H,t),7.42(1H,d),12.01(1H,s)。
m/z ESI M+H
+533.1。
实施例61N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-3-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-5-溴-4-氟-2-甲基-苯甲酰胺的制备
与实施例60的制备方法相同,除了用1-Boc-4-哌啶酮替代1-Boc-3-吡咯烷酮,制得化合物61。
1H-NMR(CDCl
3)δ:0.89(3H,t),1.25-1.74(3H,m),1.97-2.02(1H,m),2.27(3H,s),2.33(3H,s),2.40(3H,s),2.59-2.64(1H,m),3.00-3.28(4H,m),3.88-4.02(1H,m),4.50-4.55(3H,m),5.68(1H,m),5.99(1H,s),6.28(1H,m),6.55(1H,m),7.20(1H,t),7.38(1H,d),11.71(1H,s)。
m/z ESI M+H
+547.1。
实施例62N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-3'-氟-4-甲基-4'-(吗啉-4-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例23的制备方法相同,除了用1-Boc-4-哌啶酮替代1-Boc-3-吡咯烷酮,制得化合物62。
1H-NMR(DMSO-d6)δ:0.83(3H,t),1.37-1.49(2H,m),1.78(2H,d),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.40(4H,br),2.65(1H,t),2.99-3.13(4H,m),3.55(2H,s),3.58(4H,br),4.01(1H,d),4.30(2H,d),4.36(1H,d),5.63-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.75-6.82(1H,m),7.28(1H,s),7.45-7.51(4H,m),8.22(1H,t),11.49(1H,s)。
m/z ESI M+H
+644.3。
实施例63N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(4,4-二氟-哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4,4-二氟哌啶替代哌啶,制得化合物63。
1H-NMR(DMSO-d6)δ:0.81-0.87(4H,m),1.21-1.51(5H,m),1.78(2H,t),1.93-2.01(4H,m),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.66(1H,t),3.00-3.13(4h,m),3.40-3.47(1H,m),3.58(2H,s),4.01(1H,d),4.28-4.38(3H,m),5.63-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.75-6.82(1H,m),7.23(1H,s),7.37-7.41(3H,m),7.60(2H,d),8.22(1H,t),11.49(1H,s)。
m/z ESI M+H
+660.3。
实施例64N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(4-乙酰基-哌嗪-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4-乙酰基哌嗪替代哌啶,制得化合物64。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.39-1.46(2H,t),1.98-2.02(2H,d),1.99(3H,s),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.32(2H,br),2.39(2H,br),2.66(1H,t),2.99-3.10(4H,m),3.43(4H,s),3.53(2H,s),3.99-4.03(1H,m),4.29-4.38(3H,m),5.63-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.75-6.82(1H,m),7.23(1H,s),7.38-7.41(3H,m),7.60(2H,d),8.21(1H,t),11.49(1H,s)。
m/z ESI M+H
+667.4。
实施例65N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(4-甲氧基哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4-甲氧基哌啶替代哌啶,制得化合物65。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.35-1.48(2H,m),1.60-1.83(3H,m),1.91-2.02(3H,m),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.65(1H,t),2.85-3.22(8H,m),3.25(3H,s),3.42(2H,s),3.99-4.03(1H,m),4.24-4.38(4H,m),5.63-5.67(1H,m),53.89(1H,s),6.05-6.12(1H,m),6.75-6.82(1H,m),7.27(1H,s),7.45(1H,s),7.62-7.76(4H,m),8.22(1H,t),11.49(1H,s)。
m/z ESI M+H
+654.4。
实施例66N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(4-二甲氨基-哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4-(二甲氨基)哌啶替代哌啶,制得化合物66。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.38-1.53(4H,m),1.78-1.86(2H,m),1.93-2.02(2H,m),2.11(3H,s),2.22(3H,s),2.25(3H,s),2.31(1H,m),2.44(6H,s),2.66(1H,t),2.91-3.12(8H,m),3.50(2H,s),3.97-4.02(1H,m),4.28-4.39(3H,m),5.63-5.66(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.75-6.82(1H,m),7.23(1H,d),7.35-7.41(3H,m),7.59(2H,d),8.19(1H,t),11.47(1H,s)。
m/z ESI M+H
+667.4。
实施例67N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(4-甲基高哌嗪-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4-甲基高哌嗪替代哌啶,制得化合物67。
1H-NMR(DMSO-d6)δ:0.83(3H,t),1.37-1.48(2H,m),1.75-1.81(2H,d),1.93-2.01(2H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.61-2.78(6H,m),2.84(2H,s),2.99-3.13(4H,m),3.20-3.32(4H,br),3.72(2H,s),4.00(1H,d),4.28-4.37(3H,m),5.62-5.67(1H,m),5.86(1H,s),6.04-6.09(1H,m),7.22(1H,s),7.40-7.45(3H,m), 7.60(2H,d),8.20(1H,t),11.47(1H,s)。
m/z ESI M+H
+653.4。
实施例68N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(3-氧代哌嗪-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用3-哌嗪酮替代哌啶,制得化合物68。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.36-1.17(2H,m),1.75-1.81(2H,m),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.57(2H,t),2.66(1H,t),2.93(2H,s),2.99-3.12(4H,m),3.16(2H,t),3.58(2H,s),3.97-4.03(1H,m),4.29-4.40(3H,m),5.62-5.66(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.74-6.82(1H,m),7.24(1H,d),7.38-7.42(3H,m),7.61(2H,d),7.67(1H,s),8.21(1H,t),11.48(1H,s)。
m/z ESI M+H
+639.3。
实施例69N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(4-甲基哌嗪-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4-甲基哌嗪替代哌啶,制得化合物69。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.39-1.47(2H,m),1.75-1.83(2H,m),2.11(3H,s),2.20(3H,s),2.22(3H,s),2.25(3H,s),2.39(8H,br),2.66(1H,t),2.99-3.12(4H,m),3.49(2H,s),4.01(1H,d),4.29-4.39(3H,m),5.63-5.66(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.70-6.82(1H,m),7.23(1H,d),7.35-7.41(3H,m),7.58(2H,d),8.20(1H,t),11.47(1H,s)。
m/z ESI M+H
+639.3。
实施例70N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-3'-(吗啉-4-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例30的制备方法相同,除了用1-Boc-4-哌啶酮替代1-Boc-3-吡咯烷酮,制得化合物70。
1H-NMR(DMSO-d6)δ:0.85(3H,t),1.37-1.50(2H,m),1.75-1.83(2H,d),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.39(4H,br),2.65(1H,t),2.99-3.12(4H,m),3.53(2H,s),3.58(4H,br),4.01(1H,d),4.29-4.38(3H,m),5.63-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.75-6.83(1H,m),7.22(1H,s),7.31(1H,d),7.39-7.45(2H,m),7.50-7.55(2H,m),8.23(1H,t),11.49(1H,s)。
m/z ESI M+H
+626.3。
实施例71N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-3'-氯-4-甲基-4'-(吗啉-4-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例21的制备方法相同,除了用1-Boc-4-哌啶酮替代1-Boc-3-吡咯烷酮,制得化合物71。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.37-1.48(2H,m),1.74-1.82(2H,d),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.44(4H,br),2.65(1H,t),2.99-3.12(4H,m),3.59(6H,br),4.01(1H,d),4.29-4.41(3H,m),5.62-5.66(1H,m),5.86(1H,s),6.04-6.09(1H,m),6.74-6.82(1H,m),7.25(1H,s),7.44(1H,s),7.54-7.62(2H,m),7.71(1H,s),8.22(1H,t),11.48(1H,s)。
m/z ESI M+H
+660.3。
实施例72N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-3'-(哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例70的制备方法相同,除了用哌啶替代吗啉,制得化合物72。
1H-NMR(DMSO-d6)δ:0.85(3H,t),1.37-1.50(2H,m),1.64-1.85(8H,d),2.12(3H,s),2.22(3H,s),2.26(3H,s),2.67(1H,t),2.85-3.15(10H,m),4.02(1H,d),4.29-4.41(5H,m),5.63-5.67(1H,m),5.88(1H,s),6.06-6.10(1H,m),6.75-6.83(1H,m),7.30(1H,s),7.48-7.57(3H,m),7.72-7.78(1H,m),7.88(1H,s),8.21(1H,t),9.90(1H,br),11.49(1H,s)。
m/z ESI M+H
+624.3。
实施例73N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-3'-(4-甲基哌嗪-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例70的制备方法相同,除了用N-甲基哌嗪替代吗啉,制得化合物73。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.37-1.50(2H,m),1.74-1.82(2H,d),2.12(3H,s),2.20(3H,s),2.25(3H,s),2.61-3.25(16H,m),3.62(2H,s),4.01(1H,d),4.29-4.42(3H,m),5.63-5.67(1H,m),5.88(1H,s),6.05-6.10(1H,m),6.75-6.83(1H,m),7.23(1H,s),7.32(1H,d),7.40-7.48(2H,m),7.52-7.59(2H,d),8.22(1H,t),11.49(1H,s)。
m/z ESI M+H
+639.4。
实施例74N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-3'-(4-乙酰哌嗪-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例70的制备方法相同,除了用N-乙酰基哌嗪替代吗啉,制得化合物74。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.38-1.48(2H,m),1.75-1.82(2H,d),1.98(3H,s),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.33(2H,br),2.40(2H,br),2.66(1H,t),2.99-3.13(4H,m),3.42(4H,br),3.58(2H,s),4.01(1H,d),4.29-4.42(3H,m),5.63-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.75-6.83(1H,m),7.22(1H,s), 7.31(1H,d),7.40-7.45(2H,m),7.52-7.55(2H,d),8.23(1H,t),11.48(1H,s)。
m/z ESI M+H
+667.3。
实施例75N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-3'-(4-甲基高哌嗪-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例70的制备方法相同,除了用N-甲基高哌嗪替代吗啉,制得化合物75。
1H-NMR(DMSO-d6)δ:0.77(3H,t),1.35-1.49(2H,m),1.75-1.83(2H,m),1.94-2.04(2H,m),2.04(3H,s),2.11(3H,s),2.18(3H,s),2.59-2.76(5H,m),2.80(1H,br),2.99-3.25(6H,m),3.70(2H,s),3.94(1H,d),4.22-4.31(3H,m),5.55-5.60(1H,m),5.80(1H,s),5.98-6.03(1H,m),6.67-6.75(1H,m),7.16(1H,s),7.33-7.61(5H,m),8.13(1H,t),11.40(1H,s)。
m/z ESI M+H
+653.3。
实施例76N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(3-甲氧基-1-氮杂环丁烷基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用3-甲氧氮杂环丁烷替代哌啶,制得化合物76。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.37-1.49(2H,m),1.78(2H,d),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.67(1H,t),2.99-3.13(4H,m),3.20(3H,s),3.40(2H,br),3.80(2H,br),3.87-4.13(4H,m),4.29-4.38(3H,m),5.63-5.66(1H,m),5.87(1H,s),6.04-6.10(1H,m),6.75-6.82(1H,m),7.24(1H,d),7.41-7.46(3H,m),7.63(2H,d),8.21(1H,t),11.48(1H,s)。
m/z ESI M+H
+626.3。
实施例77N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-[4-(2-甲氧基乙基氧基)-哌啶-1-基甲基]-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4-(2-甲氧基乙氧基)哌啶替代哌啶,制得化合物77。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.36-1.49(2H,m),1.78(2H,m),1.93-2.12(7H,m),2.22(3H,s),2.27(3H,s),2.66(1H,t),2.89-3.22(8H,m),3.24(3H,s),3.44(2H,br),3.53(2H,t),4.01(1H,d),4.26-4.38(5H,m),5.63-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.75-6.83(1H,m),7.28(1H,s),7.45(1H,s),7.67-7.75(4H,m),8.22(1H,t),11.49(1H,s)。
m/z ESI M+H
+698.5。
实施例78N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(2-氧杂-7-氮杂-螺[3.5]壬-7-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用2-氧杂-7-氮杂-螺[3,5]壬烷替代哌啶,制得化合物78。
1H-NMR(DMSO)δ:0.83(3H,t),1.37-1.17(2H,m),1.73-2.04(6H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.65(1H,t),2.99-3.09(4H,m),3.40(6H,br),4.01(1H,d),4.23-4.42(7H,m),5.62-5.66(1H,m),5.86(1H,s),6.04-6.09(1H,m),6.74-6.81(1H,m),7.25(1H,s),7.43(1H,s),7.44(2H,d),7.65(2H,d),8.20(1H,t),11.49(1H,s)。
m/z ESI M+H
+666.3。
实施例79N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-溴-3-{乙基-[1-(4-吗啉-4-基-丁-2-烯酰基)-哌啶-4-基]-氨基}-2-甲基-苯甲酰胺的制备
与实施例14的制备方法相同,除了用1-Boc-4-哌啶酮代替1-Boc-3-吡咯烷酮,并且用4-(吗啉-4-基)巴豆酸盐酸盐替代4-二甲氨基巴豆酸盐酸盐制得化合物79。
1H-NMR(DMSO-d6)δ:0.78(3H,t),1.32-1.47(2H,m),1.72(2H,d),2.11(3H,s),2.15(3H,s),2.19(3H,s),2.37(4H,br),2.59(1H,t),2.93-3.18(6H,m),3.58(4H,br),3.95-4.02(1H,m),4.25(2H,d),4.33-4.36(1H,m),5.86(1H,s),6.51-6.63(2H,m),7.09(1H,s),7.31(1H,s),8.23(1H,t),11.47(1H,s)。
m/z ESI M+H
+628.1。
实施例80N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-3'-[4-(吗啉-4-基)哌嗪-1-基甲基]-联苯-3-甲酰胺的制备
与实施例70的制备方法相同,除了用4-(哌啶-4-基)吗啉替代吗啉,制得化合物80。
1H-NMR(DMSO)δ:0.89(3H,t),1.35-1.48(4H,m),1.82(4H,d),2.16(3H,s),2.27(3H,s),2.30(3H,s),2.42(4H,br),2.70(1H,t),2.99-3.07(5H,m),3.62(4H,s),4.06(1H,d),4.33-4.42(3H,m),5.62-5.65(1H,m),5.87(1H,s),6.03-6.09(1H,m),6.74-6.80(1H,m),7.22(1H,s),7.30(1H,s),7.40-7.57(4H,m),8.19(1H,t),11.45(1H,s)。
m/z ESI M+H
+709.5。
实施例81N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-[4-(吗啉-4-基)哌嗪-1-基甲基]-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4-(哌啶-4-基)吗啉替代哌啶,制得化合物81。
1H-NMR(DMSO)δ:0.84(3H,t),1.37-1.49(3H,m),1.73-1.945(5H,m),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.45-2.68(6H,m),2.96-3.17(8H,m),3.61(6H,br),4.01(1H,d),4.28-4.39(3H,m),5.62-5.67(1H,m),5.87(1H,s),6.04-6.10(1H,dd),6.76-6.83(1H,m),7.25(1H,s),7.42-7.53(3H,m),7.63(2H,d),8.22(1H,t),11.50(1H,s)。
m/z ESI M+H
+709.5。
实施例82N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-3'-(4-甲氧基哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例70的制备方法相同,除了用4-甲氧基哌啶替代吗啉,制得化合物82。
1H-NMR(DMSO)δ:0.85(3H,t),1.38-1.47(3H,m),1.76-1.97(5H,m),2.12(3H,s),2.22(3H,s),2.26(3H,s),2.61-2.71(2H,m),2.97-3.20(7H,m),3.25(3H,s),3.34(2H,s),4.02(1H,d),4.30-4.39(4H,m),5.63-5.67(1H,m),5.88(1H,s),6.05-6.10(1H,m),6.75-6.83(1H,m),7.20-7.93(6H,m),8.21(1H,t),11.47(1H,s)。
m/z ESI M+H
+654.3。
实施例83N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(4-氧代-哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例70的制备方法相同,除了用4-氧代哌啶替代吗啉,制得化合物83。
1H-NMR(DMSO)δ:0.85(3H,t),1.35-1.48(2H,m),1.73-1.83(2H,m),2.12(3H,s),2.23(3H,s),2.26(3H,s),2.39(4H,br),2.60-2.76(5H,m),2.99-3.13(4H,m),3.63(2H,s),3.98-4.023(1H,m),4.26-4.41(3H,m),5.61-5.68(1H,m),5.88(1H,s),6.03-6.13(1H,m),6.72-6.83(1H,m),7.25(1H,s),7.38-7.51(3H,m),7.64(2H,d),8.23(1H,t),11.50(1H,s)。
m/z ESI M+H
+638.3。
实施例84N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(4-羟基哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4-哌啶醇替代哌啶,制得化合物84。
1H-NMR(DMSO)δ:0.83(3H,t),1.37-1.45(2H,m),1.58-1.81(4H,m),1.86-1.97(2H,m),2.11(3H,s),2.21(3H,s),2.25(3H,s),2.65(1H,t),2.82-3.18(8H,m),3.34(2H,s),4.01(1H,d),4.28-4.38(4H,m),5.62-5.66(1H,m),5.87(1H,s),6.04-6.09(1H,m),6.74-6.82(1H,m),7.27(1H,s),7.44(1H,s),7.60-7.75(4H,m),8.21(1H,t),11.48(1H,s)。
m/z ESI M+H
+640.3。
实施例85N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(3-异丙氧基-1-氮杂环丁烷基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用3-异丙氧基-1-氮杂环丁烷替代哌啶,制得化合物85。
1H-NMR(DMSO)δ:0.84(3H,t),1.08(6H,d),1.36-1.47(2H,m),1.78(2H,d),2.12(3H,s),2.22(3H,s),2.26(3H,s),2.65(1H,t),2.99-3.12(4H,m),3.61-3.72(2H,m),4.03-4.41(9H,m),5.63-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.75-6.83(1H,m),7.26(1H,s),7.43(1H,s),7.56(2H,d),7.69(2H,d),8.23(1H,t),11.50(1H,s)。
m/z ESI M+H
+654.3。
实施例86N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-3'-氯-4-甲基-4'-[4-(吗啉-4-基)-哌啶-1-基甲基]-[1,1'-联苯]-3-甲酰胺的制备
与实施例21的制备方法相同,除了用1-Boc-4-哌啶酮替代1-Boc-3-吡咯烷酮,用4-(吗啉-4-基)-哌啶替代吗啉,制得化合物86。
1H-NMR(DMSO)δ:0.84(3H,t),1.33-1.49(4H,m),1.71-1.82(4H,m),1.99-2.09(2H,m),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.45(4H,br),2.65(1H,t),2.90(2H,d),2.99-3.13(5H,m),3.56(6H,br),4.01(1H,d),4.29-4.38(3H,m),5.63-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.76-6.83(1H,m),7.26(1H,s),7.45(1H,s),7.54(1H,d),7.61(1H,d),7.71(1H,s),8.25(1H,t),11.50(1H,s)。
m/z ESI M+H
+743.3。
实施例87N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(1-氧杂-8-氮杂-螺[4.5]癸烷-8-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用1-氧-8-氮杂-螺[4.5]癸烷替代哌啶,制得化合物87。
1H-NMR(DMSO)δ:0.84(3H,t),1.36-1.47(2H,m),1.63-1.94(10H,m),2.11(3H,s),2.22(3H,s),2.26(3H,s),2.65(1H,t),2.99-3.43(9H,m),3.72(2H,t),4.02(1H,d),4.29-4.41(4H,m),5.64-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,s),6.76-6.83(1H,m),7.26(1H,s),7.44(1H,s),7.61-7.79(4H,m),8.24(1H,t),11.51(1H,s)。
m/z ESI M+H
+680.3。
实施例88N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(4-乙氧基哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4-乙氧基哌啶替代哌啶,制得化合物88。
1H-NMR(CDCl
3)δ:0.88(3H,t),1.20(3H,t),1.48-1.62(3H,m),1.78-1.82(5H,m),2.13(3H,s),2.17-2.32(2H,m),2.35(3H,s),2.41(3H,s),2.69(1H,t),2.92-3.11(6H,m),3.46(2H,q),3.57(1H,br),3.88-3.98(3H,m),4.50-4.58(3H,d),5.64-5.68(1H,m),5.91(1H,s),6.22-6.27(1H,s),6.52-6.59(1H,m),7.21(1H,t),7.28(1H,s),7.30(1H,s),7.49(2H,d),7.56(2H,d),11.67(1H,s)。
m/z ESI M+H
+668.4。
实施例89N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-[4-(四氢吡喃-4-基)-哌嗪-1-基甲基]-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用4-(四氢吡喃-4-基)-哌嗪替代哌啶,制得化合物89。
1H-NMR(DMSO)δ:0.83(3H,t),1.28-1.48(3H,m),1.72-1.95(5H,m),2.11(3H,s),2.21(3H,s),2.26(3H,s),2.53-3.20(13H,m),3.27(3H,t),3.57(2H,s),3.92(2H,d),4.01(1H,d),4.28-4.38(3H,m),5.63-5.67(1H,m),5.87(1H,s),6.05-6.10(1H,m),6.75-6.83(1H,m),7.23(1H,s),7.37-7.45(3H,m),7.62(2H,d),8.23(1H,t),11.51(1H,s)。
m/z ESI M+H
+710.0。
实施例90N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-3'-氯-4-甲基-4'-(4-甲氧基哌啶-4-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例86的制备方法相同,除了用4-甲氧基哌啶替代4-(吗啉-4-基)-哌啶,制得化合物90。
1H-NMR(CDCl
3)δ:0.91(3H,t),1.56-1.63(3H,m),1.77-1.92(5H,m),2.08-2.22(5H,m),2.36(3H,s),2.42(3H,s),2.72(1H,t),2.92-3.13(6H,m),3.36(3H,s),3.36-3.43(1H,m),3.92-4.02(3H,m),4.52-4.61(3H,m),5.66-5.70(1H,m),5.95(1H,s),6.24-6.30(1H,m),6.54-6.61(1H,m),7.22(1H,t),7.27(1H,d),7.30(1H,d),7.45(1H,d),7.51(1H,d),7.83(1H,br),11.40(1H,br)。
m/z ESI M+H
+688.3。
实施例91N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-3'-氯-4-甲基-4'-(3-甲氧基哌啶-4-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例86的制备方法相同,除了用3-甲氧基哌啶替代4-(吗啉-4-基)-哌啶,制得化合物91。
1H-NMR(CDCl
3)δ:0.91(3H,t),1.53-1.64(3H,m),1.79-1.93(5H,m),2.02-2.10(2H,m),2.20(3H,s),2.35(3H,s),2.42(3H,s),2.72(1H,t),2.89-3.18(6H,m),3.37(3H,s),3.51-3.61(1H,m),3.88-4.02(3H,m),4.52-4.61(3H,m),5.65-5.70(1H,m),5.97(1H,s),6.24-6.30(1H,m),6.54-6.61(1H,m),7.20(1H,t),7.28(1H,d),7.30(1H,d),7.44(1H,d),7.51(1H,d),7.76(1H,br),11.45(1H,br)。
m/z ESI M+H
+688.3。
实施例92N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-4-甲基-4'-(3-甲氧基哌啶-4-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例34的制备方法相同,除了用3-甲氧基哌啶替代哌啶,制得化合物92。
1H-NMR(CDCl
3)δ:0.90(3H,t),1.51-1.62(2H,m),1.74-1.90(4H,m),1.95-2.04(2H,m),2.06-2.22(5H,m),2.36(3H,s),2.42(3H,s),2.66-2.84(2H,m),2.98-3.12(5H,m),3.34(3H,s),3.41-3.51(1H,m),3.68(2H,br),3.93(1H,d),4.51-4.61(3H,m),5.65-5.68(1H,m),5.94(1H,s),6.23-6.28(1H,m),6.52-6.61(1H,m),7.15(1H,t),7.29(1H,d),7.31(1H,d),7.39(2H,d),7.46(2H,d),11.57(1H,br)。
m/z ESI M+H
+654.4。
实施例93N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-3'-氟-4-甲基-4'-(3-甲氧基哌啶-4-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例90的制备方法相同,除了2-氟-4-溴苯甲酸用替代2-氯-4-溴苯甲酸,制得化合物93。
1H-NMR(CDCl
3)δ:
1H-NMR(DMSO)0.83(3H,t),1.39-1.45(3H,m),1.78(2H,d),2.11(3H,s),2.21(4H,m),2.25(4H,m),2.30(4H,m),2.65(4H,m),3.03(4H,m),3.31(4H,m),4.01(1H,d),4.3-4.40(4H,m),5.64(1H,dd),5.87(1H,s),6.07(1H,dd),6.78(1H,dd),7.13-7.27(4H,m),7.43(1H,m),7.57(1H,m),7.71(1H,m),8.20(1H,s),10.15(1H,s),11.47(1H,s)。
m/z ESI M+H
+672.8。
实施例94N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-4-基)-乙基-氨基]-3',4-二甲基-4'-(3-甲氧基哌啶-4-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例90的制备方法相同,除了2-甲基-4-溴苯甲酸用替代2-氯-4-溴苯甲酸,制得化合物94。
1H-NMR(DMSO)δ:0.83(3H,t),1.39-1.45(3H,m),1.78(2H,d),2.11(3H,s),2.21(4H,m),2.25(4H,m),2.30(4H,m),2.65(4H,m),3.03(4H,m),3.31(4H,m),4.01(1H,d),4.3-4.40(4H,m),5.64(1H,dd),5.87(1H,s),6.07(1H,dd),6.78(1H,dd),7.13-7.27(4H,m),7.43(1H,m),7.57(1H,m),7.71(1H,m),8.20(1H,s),10.15(1H,s),11.47(1H,s)。
m/z ESI M+H
+668.9。
实施例95N-(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-5-[(1-丙烯酰基-哌啶-3-基)-乙基-氨基]-4-甲基-4'-(4-甲氧基哌啶-1-基甲基)-[1,1'-联苯]-3-甲酰胺的制备
与实施例65的制备方法相同,除了用1-Boc-3-哌啶酮替代1-Boc-4-哌啶酮,制得化合物95。
1H-NMR(DMSO-d6)δ:0.84(3H,t),1.43(3H,m),1.78(2H,m),2.00(H,m),2.11(3H,s)2.22(3H,s),2.25(2H,m),2.31(3H,s),2.38(1H,m),2.67(2H,s),3.08(4H,m),3.26(4H,m),4.0(1H,m),4.32(4H,m),5.64(1H,dd),5.87(1H,s),6.08(1H,dd),6.79(1H,q),7.17(2H,dd),67.26(2H,d),7.43(1H,d),7.57(1H,d),7.71(1H,m),8.20(1H,d),10.15(1H,s),11.48(1H,s)。
m/z ESI M+H
+654.3。
实施例96 5-{[1-(2-氰基-乙酰基)-哌啶-4-基]-乙基-氨基}-4-甲基-4'-吗啉-4-基甲基-联苯-3-甲酸1,3-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-酰胺的制备
以5-(乙基-哌啶-4-基-氨基)-4-甲基-4'-吗啉-4-基甲基-联苯-3-甲酸(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-酰胺为原料,其合成方法与实施例35相同。
100ml烧瓶中加入氰基乙酸(70mg,0.82mmol)、HATU(311mg,0.82mmol)、三乙胺(138mg,1.36mmol)、DCM 10ml、DMF 2.5ml,于室温搅拌30分钟。加入5-(乙基-哌啶-4-基-氨基)-4-甲基-4'-吗啉-4-基甲基-联苯-3-甲酸(4,6-二甲基-2-氧代-1,2-二氢-吡啶-3-基甲基)-酰胺(390mg,0.68mmol),于室温反应3小时。反应液加碳酸氢钠溶液20ml,乙酸乙酯20ml萃取3次,合并有机相用无水硫酸钠干燥,柱层析(DCM/MeOH=20:1)分离得到化合物96。
1H-NMR(DMSO)δ:11.49(s,1H),8.22(t,1H),7.35~7.64(m,5H),7.25(s,1H),5.87(s,1H),4.29(d,2H),4.22(d,1H),4.01(d,2H),3.60~3.63(m,7H),2.97~3.12(m,8H),2.65(t,1H),2.25(s,3H),2.21(s,3H),2.11(s,3H),1.74(d,2H),1.55(q,1H),1.38(q,1H),0.84(t,3H)。
m/z ESI M+H
+639.8。
生物学测试
试验例1本发明化合物对EZH2野生型和突变型表达阳性肿瘤细胞体外生长50%抑制(GI50)浓度值的测定
实验材料与方法
1)肿瘤细胞系及细胞培养
肿瘤细胞系是研究肿瘤体外生长抑制的有效细胞模型,本发明选择具有代表性的肿瘤细胞系用于化合物活性测定。细胞培养条件与方法按每种细胞系要求进行。每次体外培养不超过3次传代。根据需要,可对细胞系进行单克隆纯化与鉴定。
细胞培养基选用RPMI1640(Gibco)加入5-20%胎牛血清(Gibco)、1%双抗和2mM谷氨酰胺。
人类B细胞非霍奇金淋巴瘤Karpas 422(ATCC)表达EZH2 Y641N突变蛋白,用RPMI 1640加10%FBS和1%双抗及2mM谷氨酰胺悬浮培养。
人弥漫性大细胞淋巴瘤B淋巴细胞Pfeiffer(ATCC)表达EZH2 A677G突变蛋白,用RPMI 1640加10%FBS和1%双抗及2mM谷氨酰胺悬浮培养。
人B细胞淋巴瘤SU-DHL-8(ATCC)表达EZH2野生型,用RPMI 1640加10%FBS和1%双抗及2mM谷氨酰胺悬浮培养。
2)药物处理
悬浮培养细胞直接离心收集(1700rpm,3分钟),弃上清,计数细胞。根据每种细胞生长周期,配制不同的细胞浓度(每毫升1-10×10
4细胞),接种到96孔板(Corning),每孔200微升,37℃,5%CO
2培养过夜。第二天,加入待测化合物到培养细胞中,平行2孔。有机溶剂终浓度不超过千分之一,细胞继续培养6-12天,MTT测定。
本发明化合物与对照化合物EPZ-6438用DMSO溶解,化合物纯度达98%以上。化合物贮存浓度为10mM,-20℃保存,使用前对倍或者10倍系列稀释。
对照化合物EPZ-6438按照专利US2012264734方法合成,其结构式如下。
3)MTT检测及GI50计算
MTT检测试剂为Dojindo CCK8试剂盒,酶标测定仪为THERMOMULTISKAN FC仪。
悬浮细胞可直接加入CCK8试剂,终浓度为10%,继续培养1-4小时,当溶剂对照孔呈现暗黄色时,测OD450nm光吸收值,按下列公式计算细胞生长率。
细胞生长率%=100*(T-T
0)/(C-T
0)
T=药物处理细胞孔光密度值-空白对照孔光密度值;T
0=药物处理前细胞孔光密度值-空白对照孔光密度值;C=溶剂对照组细胞孔光密度-空白对照孔光密度值。
通过药物浓度与细胞生长率曲线,计算细胞生长50%抑制的药物浓度即GI50。试验重复进行3次,并对数据进行生物学统计分析。
实验结果
表2总结了本发明化合物中部分具有代表性化合物对EZH2野生型和不同突变型表达阳性肿瘤细胞体外生长抑制(或诱导细胞凋亡)GI50浓度的结果。其中GI50值越小,化合物活性越强。如果化合物对EZH2野生型细胞生长抑制(GI50)浓度高,突变型浓度低,即EZH2野生型GI50/ EZH2突变型GI50比值大,说明该化合选择性高。
表2
结论:结果显示所测本发明化合物对表达EZH2 A677G突变型细胞Pfeiffer具有高的生长抑制活性,GI50浓度可达亚纳摩尔(如化合物65、化合物77、化合物87、化合物88和化合物89);对表达EZH2Y641N突变细胞Karpas 422也呈现 强的生长抑制活性;对表达EZH2野生型细胞SU-DHL-8需要高浓度才呈现一定的生长抑制活性。
试验例2本发明化合物对EZH2突变型A677G表达阳性肿瘤细胞Pfeiffer的体内生长抑制活性的测定
NOD/SCID小鼠皮下接种表达EZH2A677G突变蛋白的人弥漫性大细胞淋巴瘤B淋巴细胞Pfeiffer(ATCC),建立人源B细胞淋巴瘤皮下移植肿瘤模型。
实验动物:NOD/SCID小鼠,雌性,9周(肿瘤细胞接种时的小鼠周龄),平均体重20.3g,18只。购自上海灵畅生物科技有限公司有限公司,动物合格证编号:2002007656。饲养环境为SPF级。
试验样品:本发明实施例化合物34(纯度:99.03%)和化合物35(纯度:97.6%),固体粉末,-20℃保存。
细胞和动物造模:
将Pfeiffer细胞培养在含10%胎牛血清的RPMI1640培养液中。收集指数生长期的Pfeiffer细胞,用PBS重悬至1×10
8/ml细胞用于NOD/SCID小鼠皮下接种。
细胞接种前一天所有实验小鼠进行Co60照射处理,照射剂量为200rad,然后于右侧皮下接种1×10
7Pfeiffer细胞,细胞重悬在1:1的PBS与基质胶中(0.1ml/只)。待肿瘤平均体积为134mm
3时,根据肿瘤大小随机分组。肿瘤体积计算公式为:长径×短径
2/2。肿瘤细胞接种当天定义为第0天。试验分为溶媒对照组和给药组(化合物34组和化合物35组),每组6只,口服给药,每天一次,共给药4周。根据相对肿瘤抑制率(TGI)进行疗效评价,根据动物体重变化和死亡情况进行安全性评价。
试验样品的配制:称取化合物34或35,先加入超纯水混匀,再加入甲磺酸调清,此时PH值约为1.64,再加入4M NaOH调PH至3.5,超纯水定容为20mg/ml。
结果判断标准:相对肿瘤抑制率TGI(%):TGI=1-T/C(%)。
T/C%为相对肿瘤增值率,即在某一时间点,治疗组和对照组相对肿瘤体积或瘤重的百分比值。T和C分别为治疗组和对照组在某一特定时间点的相对肿瘤体积(RTV)。计算公式如下:
T/C%=T
RTV/C
RTV*100%
T
RTV:治疗组平均RTV;C
RTV:溶媒对照组平均RTV;RTV=V
t/V
0,V
0为分组时该动物的瘤体积,V
t为治疗后该动物的瘤体积。统计分析:所有试验结果以平均瘤体积±SEM(平均标准误差)表示。选择给药开始后27天的肿瘤体积数据进行不同组间的统计分析,用独立样本T检验方法比较治疗组相对肿瘤体积与对照组相比有无显著性差异。所有的数据均用SPSS 18.0进行分析。p<0.05为具有显著性差异。
表3小鼠肿瘤体积(mm
3)随治疗时间的变化
表4给药开始后第27天时各组TGI和T/C值
表5各组瘤重表
结论:表3、表4和表5的结果显示,溶媒对照组在开始给药后第27天平均肿瘤体积为1869mm
3。与此相比,化合物34(200mg/kg)和化合物35(200mg/kg)治疗组均产生了显著的抗肿瘤效果,开始给药后第27天平均肿瘤体积分别为538 mm
3和1116mm
3,相对肿瘤抑制率TGI分别为67.1%和42.3%,相对溶媒对照组统计学上均有显著性差异(p值分别为0.005和0.006)。瘤重分析结果与相对瘤体积分析结果基本吻合。
测试化合物34和化合物35在200mg/kg剂量时小鼠在给药期间体重均保持平稳,对受试药物耐受良好。
试验例3本发明化合物对EZH2突变型A677G表达阳性肿瘤细胞Pfeiffer的体外生长的不可逆抑制
化合物短期暴露处理(washout)对表达EZH2A677G突变蛋白的人弥漫性大细胞淋巴瘤B淋巴细胞Pfeiffer(ATCC)的体外生长抑制试验按以下方法进行。
弥漫性大细胞淋巴瘤B淋巴细胞Pfeiffer(ATCC),用完全培养基(RPMI 1640加10%FBS和1%双抗及2mM谷氨酰胺)悬浮培养。生长对数期的Pfeiffer细胞直接离心收集(1700rpm,3分钟),弃上清,计数细胞。用完全培养基配制每毫升2×10
4细胞浓度,接种到96孔板(Corning),每孔200微升,37℃,5%CO
2培养过夜。第二天,加入不同浓度的待测化合物(化合物34)到培养细胞中,平行4孔,有机溶剂DMSO终浓度不超过千分之一。细胞继续培养12h之后,将试验一半孔分别离心收集细胞(平行2孔),用无血清细胞培养基洗三次,重新悬浮于等体积(200微升)的完全细胞培养基中,与无处理孔细胞一起,37℃,5%CO
2继续培养10天。MTT检测按照试验例1的方法进行。试验重复一次,具体结果见表6。
对照品EPZ6438按试验例1方法制备。
表6化合物34不可逆抑制Pfeiffer细胞的体外生长
表6结果表明,化合物34在持续存在或短期暴露处理Pfeiffer细胞都能够有效的抑制细胞体外生长,对照化合物EPZ6438在持续存在于细胞培养基中时可有效的抑制细胞体外生长,但药物短期暴露处理在测试浓度(25nM)不能够有效的抑制细胞的生长,结果说明化合物34具有潜在的不可逆抑制细胞体外生长抑制作用。
试验例4本发明化合物大鼠体内药代动力学的研究
雄性SD大鼠(上海西普尔-必凯实验动物有限公司)购入后,在上海中医药大学动物中心(上海中医药大学伦理委员会批准)适应性饲养7天。大鼠随机分为4组(体重~220g),每组3只。分别用于尾静脉(iv)注射给药和灌胃(po)给药。灌胃组大鼠给药前禁食12小时。大鼠给药后,采用眼眶静脉丛取血,取血时间为0min(给药前)、2min、5min、15min、30min、1h、2h、4h、6h、8h、24h。每只每时间点采血量约200ul,血样收集于加有肝素钠的1.5ml离心管中,4℃8000rpm离心3分钟,收集上层血浆液,-80℃保存待测。
试验样品的配制:称取化合物34或化合物35粉末125.0mg,加超纯水4ml,搅拌,期间加入甲磺酸25ul,继续搅拌至澄清透明(pH~3.32),再加入4M NaOH调pH至3.5,然后加超纯水定溶至5ml(25mg/ml)。取以上溶液1ml,加超纯水4ml,得5mg/ml的化合物34或化合物35溶液(pH=3.5)。
给药剂量和方式:单次口服(100mpk)和单次静脉(5mpk)。
给药体积:4ml/kg(口服),1ml/kg(静脉)。
血浆中化合物浓度由LC-MS/MS(AB SCIEXQTRAP
4500LC/MS/MS系统)进行方法开发和测定,药代动力学参数用药代动力学专业软件WinNonlin计算,试验重复一次。具体结果见表7。
表7本发明化合物的大鼠体内生物利用度结果
结论:测试化合物34和35在大鼠SD体内呈现良好的生物利用度。
Claims (19)
- 一种通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中:Ar选自5元或6元的芳基或杂芳基、及由5元或6元芳基或杂芳基形成的稠环,其中Ar任选进一步被一个或多个-Q-T基团取代;L选自C 2-C 8饱和或不饱和的直链或支链烃链或环状结构,L中任意氢原子可被卤素、氰基、羟基、C 1-C 6烷氧基替换,L中任意碳原子可被N、O、S替换;L任选进一步被一个或多个-Q-T基团取代;R 1、R 2各自独立地选自氢、C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基,所述烷基、烯基、炔基、环烷基和杂环基任选进一步被一个或多个-Q-T基团取代;或者,R 1可与L的任何位置连接,连同他们所连接的N原子形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q-T基团取代;或者,R 2可与L的任何位置连接,连同它们所连接的N原子形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q-T基团取代;或者,R 1、R 2、L及它们连接的两个N原子形成4-12元杂环基,所述杂环任选进一步被一个或多个-Q-T基团取代;R 3选自氢、氟、R a;R 4、R 5各自独立地选自氢、R a;R 5’选自卤素、-OS(O) 2-C 1-C 6烷基或-OS(O) 2-C 3-C 6环烷基;R a选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基;所述烷基、烯基、炔基、环烷基、和杂环基任选进一步被一个或多个-Q-T基团取代;R 6、R 12各自独立地选自氢、C 1-C 6烷基、C 3-C 6环烷基;R 7、R 9各自独立地选自氢、卤素、R b;R b选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基,所述烷基、烯基、炔基、环烷基、和杂环基任选进一步被选自卤素、氰基、羟基、 C 1-C 6烷氧基、C 1-C 6烷基-OC(O)-、氨基、C 1-C 6烷基氨基、双C 1-C 6烷基氨基、4-12元杂环基的一种或多种基团取代;R 8、R 10、R 11各自独立地选自氢、卤素、C 1-C 3烷基或环烷基;Q选自一个键或C 1-C 6亚烷基,所述亚烷基任选进一步被选自卤素、氰基、羟基、C 1-C 6烷氧基的一种或多种基团取代;T选自氢、卤素、氰基、羟基、-NR hR i、-C(O)R h、OR h、-C(O)O-R h、C(O)NR hR i、-NR hC(O)R i、-NR jC(O)NR hR i、-NR hC(O)OR i、或R k;或者-Q-T是氧代;R k选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5至10元杂芳基、芳基,R k任选进一步被一个或多个-Q 1-T 1基团取代;R h、R i、R j各自独立地选自氢或R l,R l选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、和芳基,R l任选进一步被一个或多个-Q 1-T 1基团取代;或者R h、R i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q 1-T 1基团取代;Q 1为一个键或C 1-C 6亚烷基,所述亚烷基任选进一步被选自卤素、氰基、羟基、C 1-C 6烷氧基的一种或多种基团取代;T 1选自氢、卤素、氰基、羟基、-NR mR n、-C(O)R m、OR m、-C(O)OR m、-C(O)NR mR n、-NR mC(O)R n、-NR oC(O)NR mR n、-NR mC(O)OR n、-O(CH 2) aNR mR n、-(CH 2) aNR mR n、-S(O) 2NR mR n或R p;或者-Q 1-T 1是氧代;R p选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基;R m、R n、R o各自独立地选自氢或R q,R q选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C 1-C 6烷基、羟基、氧代的一种或多种基团取代;或者R m、R n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C 1-C 6烷基、-OR x、-NR xR y、-C(O)R x、-O(CH 2) aOR x的一种或多种基团取代;R x和R y各自独立地选自氢或R z,R z选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C 1-C 6烷基、羟基、氧代的一种或多种基团取代;或者R x、R y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;a为1至4的整数。
- 根据权利要求1所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中:Ar选自X选自CR 14;Y选自N或CR 15;Z选自N或CR 16;R 13选自氢、卤素、C 1-C 6烷基或C 3-C 6环烷基;R 14、R 15、R 16各自独立地选自-Q-T基团;Q选自一个键或C 1-C 6亚烷基;T选自氢、卤素、氰基、羟基、-NR hR i、-C(O)R h、OR h、-C(O)O-R h、C(O)NR hR i、-NR hC(O)R i、-NR jC(O)NR hR i、-NR hC(O)OR i、或R k;R k选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5至10元杂芳基、芳基,R k任选进一步被一个或多个-Q 1-T 1基团取代;R h、R i、R j各自独立地选自氢或R l,R l选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、和芳基,R l任选进一步被一个或多个-Q 1-T 1基团取代;或者R h、R i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q 1-T 1基团取代;Q 1为一个键或C 1-C 6亚烷基;T 1选自氢、卤素、氰基、羟基、-NR mR n、-C(O)R m、OR m、-C(O)O-R m、-C(O)NR mR n、-NR mC(O)R n、-NR oC(O)NR mR n、-NR mC(O)OR n、-O(CH 2) aNR mR n、-(CH 2) aNR mR n、-S(O) 2NR mR n或R p;R p选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基;R m、R n、R o各自独立地选自氢或R q,R q选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C 1-C 6烷基、羟基、氧代的一种或多种基团取代;或者R m、R n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C 1-C 6烷基、-OR x、-NR xR y、 -C(O)R x、-O(CH 2) aOR x的一种或多种基团取代;R x和R y各自独立地选自氢或R z,R z选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C 1-C 6烷基、羟基、氧代的一种或多种基团取代;或者R x、R y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;a为1至4的整数。
- 根据权利要求1或2所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中:R 13选自氢、卤素、和C 1-C 6烷基;X选自CR 14;Y选自CR 15;Z选自CR 16;R 14选自-Q-T基团;R 15、R 16各自独立地选自氢或卤素;Q选自一个键或C 1-C 6亚烷基;T选自氢、卤素、氰基、羟基、-NR hR i、-C(O)R h、OR h、-C(O)O-R h、C(O)NR hR i、-NR hC(O)R i、-NR jC(O)NR hR i、-NR hC(O)OR i、或R k;R k选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5至10元杂芳基、芳基,所述杂环基、杂芳基或芳基任选进一步被一个或多个-Q 1-T 1基团取代;R h、R i、R j各自独立地选自氢或R l,R l选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、和芳基,R l任选进一步被一个或多个-Q 1-T 1基团取代;或者R h、R i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q 1-T 1基团取代;Q 1为一个键或C 1-C 6亚烷基;T 1选自氢、卤素、氰基、羟基、-NR mR n、-C(O)R m、OR m、-C(O)O-R m、C(O)NR mR n、-NR mC(O)R n、-NR oC(O)NR mR n、-NR mC(O)OR n、-O(CH 2) aNR mR n、-(CH 2) aNR mR n、-S(O) 2NR mR n或R p;R p选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基;R m、R n、R o各自独立地选自氢或R q,R q选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C 1-C 6烷基、羟基、氧代的一种或多种基团取代,或者R m、R n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C 1-C 6烷基、-OR x、-NR xR y、-C(O)R x、-O(CH 2) aOR x的一种或多种基团取代;R x和R y各自独立地选自氢或R z,R z选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C 1-C 6烷基、羟基、氧代的一种或多种基团取代;或者R x、R y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;a为1至4的整数。
- 根据权利要求1至3中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中:Q选自一个键或C 1-C 6亚烷基;T选自氢、卤素、-NR hR i、OR h或R k;R k选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、4-12元杂环基、5至10元杂芳基、C 6-C 10芳基,所述烷基、烯基、炔基、杂环基、杂芳基或芳基任选进一步被一个或多个-Q 1-T 1基团取代;R h、R i各自独立地选自氢或R l,R l选自C 1-C 6烷基、5元或6元杂芳基、和C 6-C 10芳基,R l任选进一步被一个或多个-Q 1-T 1基团取代;或者R h、R i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q 1-T 1基团取代;Q 1为一个键或C 1-C 6亚烷基;T 1选自氢、卤素、羟基、-NR mR n、OR m、-C(O)OR m、-C(O)NR mR n、-O(CH 2) aNR mR n、-(CH 2) aNR mR n、-S(O) 2NR mR n或R p;R p选自C 1-C 6烷基、4-12元杂环基;R m、R n各自独立地选自氢、C 1-C 6烷基,所述烷基任选进一步被选自羟基或5元或6元杂芳基的一种或多种基团取代,其中所述杂芳基任选进一步被选自C 1-C 6烷基、氧代的一种或多种基团取代;或者R m、R n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元 杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C 1-C 6烷基、-OR x、-NR xR y、-C(O)R x、-O(CH 2) aOR x的一种或多种基团取代;R x和R y各自独立地选自氢或R z,R z选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C 1-C 6烷基、羟基、氧代的一种或多种基团取代;或者R x、R y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;a为1至4的整数。
- 根据权利要求1至4中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中:Q选自一个键;T选自氢、卤素、-NR hR i、OR h或R k;R k选自C 1-C 6烷基、C 2-C 6炔基、或苯基、嘧啶基、吡啶基、吡唑基、咪唑基、噻唑基、噁唑基或噻吩[3,2-b]噻吩基团,所述烷基、炔基、苯基、嘧啶基、吡啶基、吡唑基、咪唑基、噻唑基、噁唑基或噻吩[3,2-b]噻吩基团任选进一步被一个或多个-Q 1-T 1基团取代;R h、R i各自独立地选自氢、吡啶基、嘧啶基、和苯基,所述吡啶基、嘧啶基或苯基任选进一步被一个或多个-Q 1-T 1基团取代;或者R h、R i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被一个或多个-Q 1-T 1基团取代;Q 1为一个键或C 1-C 6亚烷基;T 1选自氢、卤素、羟基、-NR mR n、OR m、-C(O)OR m、-C(O)NR mR n、-O(CH 2) aNR mR n、-(CH 2) aNR mR n、-S(O) 2NR mR n或R p;R p选自C 1-C 6烷基;R m、R n各自独立地选自氢、C 1-C 6烷基,所述烷基任选进一步被选自羟基或5元或6元杂芳基的一种或多种基团取代,其中所述杂芳基任选进一步被选自C 1-C 6烷基、氧代的一种或多种基团取代;或者R m、R n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C 1-C 6烷基、-OR x、-NR xR y、-C(O)R x、-O(CH 2) aOR x的一种或多种基团取代;R x和R y各自独立地选自氢或R z,R z选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C 1-C 6烷基、羟基、氧代的一种或多种基团取代;或者R x、R y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;a为1至4的整数。
- 根据权利要求1至5中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中:Q选自一个键;T选自氢、卤素、-NR hR i、OR h或R k;R k选自C 1-C 6烷基、C 2-C 6炔基、或苯基、嘧啶基、吡啶基、吡唑基、咪唑基、噻唑基、噁唑基或噻吩[3,2-b]噻吩基团,所述烷基、炔基、苯基、嘧啶基、吡啶基、吡唑基、咪唑基、噻唑基、噁唑基或噻吩[3,2-b]噻吩基团任选进一步被一个或多个-Q 1-T 1基团取代;R h、R i各自独立地选自氢、吡啶基、嘧啶基、和苯基,所述吡啶基、嘧啶基或苯基任选进一步被一个或多个-Q 1-T 1基团取代;或者R h、R i与它们相连的N原子一起形成哌啶基、哌嗪基、吡咯烷基、吗啉基,所述哌啶基、哌嗪基、吡咯烷基、吗啉基任选进一步被一个或多个-Q 1-T 1基团取代;Q 1为一个键或C 1-C 6亚烷基;T 1选自氢、卤素、羟基、-NR mR n、OR m、-C(O)OR m、-C(O)NR mR n、-O(CH 2) aNR mR n、-(CH 2) aNR mR n、-S(O) 2NR mR n或R p;R p选自C 1-C 6烷基;R m、R n各自独立地选自氢、C 1-C 6烷基,所述烷基任选进一步被选自羟基或5元或6元杂芳基的一种或多种基团取代,其中所述杂芳基任选进一步被选自C 1-C 6烷基、氧代的一种或多种基团取代;或者R m、R n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C 1-C 6烷基、-OR x、-NR xR y、-C(O)R x、-O(CH 2) aOR x的一种或多种基团取代;R x和R y各自独立地选自氢或R z,R z选自C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 6环烷基、4-12元杂环基、5元或6元杂芳基、芳基,R q任选进一步被选自卤素、羟基、5元或6元杂芳基、芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C 1-C 6烷基、羟基、氧代的一种或多种基团取代;或者R x、R y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;a为1至4的整数。
- 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其药学上可 接受的盐、溶剂化物、代谢物或前药,其中:Q选自一个键;T选自氢、卤素、-NR hR i、OR h或R k;R k选自C 1-C 6烷基、C 2-C 6炔基、或苯基、嘧啶基、吡啶基、或噻吩[3,2-b]噻吩基团,所述烷基、炔基、苯基、嘧啶基、吡啶基、或噻吩[3,2-b]噻吩基团任选进一步被一个或多个-Q 1-T 1基团取代;R h、R i各自独立地选自氢、吡啶基、嘧啶基、和苯基,所述吡啶基、嘧啶基或苯基任选进一步被一个或多个-Q 1-T 1基团取代;或者R h、R i与它们相连的N原子一起形成哌啶基、哌嗪基、吗啉基,所述哌啶基、哌嗪基、吗啉基任选进一步被一个或多个-Q 1-T 1基团取代;Q 1为一个键或C 1-C 6亚烷基;T 1选自氢、卤素、羟基、-NR mR n、OR m、-C(O)OR m、-C(O)NR mR n、-O(CH 2) aNR mR n、-(CH 2) aNR mR n、-S(O) 2NR mR n或R p;R p选自C 1-C 6烷基;R m、R n各自独立地选自氢、C 1-C 6烷基,所述烷基任选进一步被选自羟基或吡啶基、嘧啶基的一种或多种基团取代,其中所述吡啶基或嘧啶基任选进一步被选自C 1-C 6烷基、氧代的一种或多种基团取代;或者R m、R n与它们相连的N原子一起形成哌啶基、吗啉基、吡咯烷基、哌嗪基、高哌嗪基、氮杂环丁烷基、氮杂螺环,所述哌啶基、吗啉基、吡咯烷基、哌嗪基、高哌嗪基、氮杂环丁烷基、氮杂螺环任选进一步被选自卤素、氧代、C 1-C 6烷基、-OR x、-NR xR y、-C(O)R x、-O(CH 2) aOR x的一种或多种基团取代;R x和R y各自独立地选自氢或R z,R z选自C 1-C 6烷基、C 3-C 6环烷基,R q任选进一步被选自卤素、羟基的一种或多种基团取代;或者R x、R y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;优选哌啶基、吗啉基、吡咯烷基、哌嗪基、高哌嗪基;a为1至4的整数。
- 根据权利要求8所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中,R 1、R 2选自氢、C 1-C 6烷基、C 3-C 6环烷基、4-12元杂环基;R 3、R 4选自氢;R 5选自氢、C 1-C 6烷基,所述烷基任选进一步被一个或多个-Q-T基团取代;R 6、R 12选自氢;R 7、R 9选自氢、C 1-C 6烷基;R 8、R 10、R 11选自氢;-Q-T为-NR hR i基团;R h、R i各自独立地选自氢和C 1-C 6烷基;或者R h、R i与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;优选哌啶基、哌嗪基、吗啉基、吡咯烷基;Ar如权利要求1至5中任一项所定义。
- 根据权利要求8所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,其中,Ar为苯基,所述苯基任选进一步被一个或多个-Q-T基团取代;R 1、R 2选自氢、C 1-C 6烷基、C 3-C 6环烷基、4-12元杂环基;R 3、R 4选自氢;R 5选自氢、C 1-C 6烷基,所述烷基任选进一步被一个或多个-Q-T基团取代;R 6、R 12选自氢;R 7、R 9选自氢、C 1-C 6烷基;R 8、R 10、R 11选自氢;-Q-T为-苯基,所述苯基任选进一步被一个或多个-Q 1-T 1基团取代Q 1为一个键或C 1-C 6亚烷基,T 1选自氢、卤素、羟基、-NR mR n、OR m、-C(O)OR m、-C(O)NR mR n、-O(CH 2) aNR mR n、-S(O) 2NR mR n;或者-Q 1-T 1是氧代;R m、R n各自独立地选自氢或R q,R q选自C 1-C 6烷基,R q任选进一步被选自卤素、羟基、5元或6元杂芳基的一种或多种基团取代,其中,所述杂芳基或芳基任选进一步被选自C 1-C 6烷基、羟基、氧代的一种或多种基团取代;或者R m、R n与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基,优选哌啶基、哌嗪基、高哌嗪基、吗啉基、吡咯烷基、氮杂环丁烷基、氮杂螺环基,所述杂环基任选进一步被选自卤素、羟基、氧代、C 1-C 6烷基、-OR x、-NR xR y、-C(O)R x、-O(CH 2) aOR x的一种或多种基团取代;R x和R y各自独立地选自氢或R z,R z选自C 1-C 6烷基、C 3-C 6环烷基;或者R x、R y与它们相连的N原子一起形成含0或1个额外杂原子的4-12元杂环基;优选哌啶基、哌嗪基、吗啉基、吡咯烷基;a为1至4的整数。
- 一种根据权利要求1至11中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药的氘代化合物,其中通式(I)化合物中的一个或多个H原子独立地被D原子替代。
- 一种药物组合物,其包含根据权利要求1至11中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,或者根据权利要求12或13所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药的氘代化合物,以及一种或多种药学上可接受的载体。
- 根据权利要求1至11中任一项所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药,或者根据权利要求12或13所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药的氘代化合物,或者根据权利要求15所述的药物组合物,在制备组蛋白甲基转移酶EZH2抑制剂中的用途。
- 根据权利要求1至11中任一项所述的通式(I)所示的化合物或其药学上 可接受的盐、溶剂化物、代谢物或前药,或者根据权利要求12或13所述的通式(I)所示的化合物或其药学上可接受的盐、溶剂化物、代谢物或前药的氘代化合物,或者根据权利要求15所述的药物组合物,在制备与组蛋白甲基转移酶EZH2相关的疾病的药物中的用途。
- 根据权利要求17所述的用途,其中所述与组蛋白甲基转移酶EZH2相关的疾病选自癌症、糖尿病炎症、免疫系统疾病、心血管内疾病、神经类疾病和呼吸类疾病。
- 根据权利要求18所述的用途,其中所述癌症选自肺癌、胃癌、肝癌、乳腺癌、鼻咽癌、胰腺癌、卵巢癌、宫颈癌、结肠直肠癌、胶质瘤、黑色素瘤、前列腺癌、肾癌、食道癌、间皮瘤、头颈癌、膀胱癌、唾腺癌、间变性大细胞淋巴瘤、白血病、淋巴瘤、非霍奇金淋巴瘤和多发性骨髓瘤,优选间变性大细胞淋巴瘤、白血病、淋巴瘤、非霍奇金淋巴瘤。
Priority Applications (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201880004761.0A CN110191879A (zh) | 2017-01-25 | 2018-01-24 | 一种组蛋白甲基转移酶ezh2抑制剂、其制备方法及其医药用途 |
US16/478,472 US11214561B2 (en) | 2017-01-25 | 2018-01-24 | Histone methyltransferase EZH2 inhibitor, preparation method and pharmaceutical use thereof |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201710061026.0 | 2017-01-25 | ||
CN201710061026 | 2017-01-25 | ||
CN201710345609 | 2017-05-16 | ||
CN201710345609.6 | 2017-05-16 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2018137639A1 true WO2018137639A1 (zh) | 2018-08-02 |
Family
ID=62978081
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2018/073946 WO2018137639A1 (zh) | 2017-01-25 | 2018-01-24 | 一种组蛋白甲基转移酶ezh2抑制剂、其制备方法及其医药用途 |
Country Status (3)
Country | Link |
---|---|
US (1) | US11214561B2 (zh) |
CN (1) | CN110191879A (zh) |
WO (1) | WO2018137639A1 (zh) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2020526481A (ja) * | 2017-07-18 | 2020-08-31 | ロンザ リミテッドLonza Limited | 5−フルオロ−2−メチル−3−ニトロ安息香酸およびそのメチルエステルを製造する方法 |
WO2020192650A1 (zh) * | 2019-03-25 | 2020-10-01 | 上海华汇拓医药科技有限公司 | 酰胺类化合物制备方法及其在医药领域的应用 |
WO2021129629A1 (zh) * | 2019-12-23 | 2021-07-01 | 四川海思科制药有限公司 | Zeste增强子同源物2抑制剂及其用途 |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2021249305A1 (zh) * | 2020-06-08 | 2021-12-16 | 南京明德新药研发有限公司 | 联苯类化合物 |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2012149504A1 (en) * | 2011-04-29 | 2012-11-01 | Huawei Technologies Co., Ltd. | Data forwarder, network and method for centralized control and management for fiber channel over ehternet networks |
WO2013155464A1 (en) * | 2012-04-13 | 2013-10-17 | Epizyme, Inc. | Combination therapy for treating cancer |
CN104039956A (zh) * | 2011-11-04 | 2014-09-10 | 葛兰素史密斯克莱知识产权(第2号)有限公司 | 治疗方法 |
WO2016172199A1 (en) * | 2015-04-20 | 2016-10-27 | Epizyme, Inc. | Combination therapy for treating cancer |
US20160332969A1 (en) * | 2011-04-13 | 2016-11-17 | Epizyme, Inc. | Substituted benzene compounds |
Family Cites Families (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP5908493B2 (ja) * | 2010-12-01 | 2016-04-26 | グラクソスミスクライン・リミテッド・ライアビリティ・カンパニーGlaxoSmithKline LLC | インドール |
JO3438B1 (ar) | 2011-04-13 | 2019-10-20 | Epizyme Inc | مركبات بنزين مستبدلة بأريل أو أريل غير متجانس |
ES2745016T3 (es) * | 2012-04-13 | 2020-02-27 | Epizyme Inc | Bromhidrato de N-((4,6-dimetil-2-oxo-L,2-dihidropiridin-3-il)metil)-5-(etil(tetrahidro-2H-piran-4-il)amino)-4-metil-4'-(morfolinometil)-[L,1'-bifenil]-3-carboxamida para su uso en el tratamiento de un trastorno proliferativo celular del sistema hematológico |
EP2934531A4 (en) * | 2012-12-19 | 2016-06-15 | Glaxosmithkline Llc | COMBINATION |
-
2018
- 2018-01-24 US US16/478,472 patent/US11214561B2/en active Active
- 2018-01-24 WO PCT/CN2018/073946 patent/WO2018137639A1/zh active Application Filing
- 2018-01-24 CN CN201880004761.0A patent/CN110191879A/zh active Pending
Patent Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20160332969A1 (en) * | 2011-04-13 | 2016-11-17 | Epizyme, Inc. | Substituted benzene compounds |
WO2012149504A1 (en) * | 2011-04-29 | 2012-11-01 | Huawei Technologies Co., Ltd. | Data forwarder, network and method for centralized control and management for fiber channel over ehternet networks |
CN104039956A (zh) * | 2011-11-04 | 2014-09-10 | 葛兰素史密斯克莱知识产权(第2号)有限公司 | 治疗方法 |
WO2013155464A1 (en) * | 2012-04-13 | 2013-10-17 | Epizyme, Inc. | Combination therapy for treating cancer |
CN104768555A (zh) * | 2012-04-13 | 2015-07-08 | Epizyme股份有限公司 | 用于治疗癌症的联合治疗 |
WO2016172199A1 (en) * | 2015-04-20 | 2016-10-27 | Epizyme, Inc. | Combination therapy for treating cancer |
Cited By (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2020526481A (ja) * | 2017-07-18 | 2020-08-31 | ロンザ リミテッドLonza Limited | 5−フルオロ−2−メチル−3−ニトロ安息香酸およびそのメチルエステルを製造する方法 |
US11001552B2 (en) | 2017-07-18 | 2021-05-11 | Lonza Ltd | Method for preparation of 5-fluoro-2-methyl-3-nitrobenzoic acid and its methyl ester |
WO2020192650A1 (zh) * | 2019-03-25 | 2020-10-01 | 上海华汇拓医药科技有限公司 | 酰胺类化合物制备方法及其在医药领域的应用 |
CN113631541A (zh) * | 2019-03-25 | 2021-11-09 | 上海华汇拓医药科技有限公司 | 酰胺类化合物制备方法及其在医药领域的应用 |
JP2022528042A (ja) * | 2019-03-25 | 2022-06-08 | 上海華匯拓医薬科技有限公司 | アミド類化合物の調製方法及びその医薬分野の使用 |
JP2022528337A (ja) * | 2019-03-25 | 2022-06-10 | 上海華匯拓医薬科技有限公司 | アミド系化合物の調製方法及びその医学分野での使用 |
JP7396369B2 (ja) | 2019-03-25 | 2023-12-12 | 上海華匯拓医薬科技有限公司 | アミド系化合物の調製方法及びその医学分野での使用 |
JP7395807B2 (ja) | 2019-03-25 | 2023-12-12 | 上海華匯拓医薬科技有限公司 | アミド類化合物の調製方法及びその医薬分野の使用 |
WO2021129629A1 (zh) * | 2019-12-23 | 2021-07-01 | 四川海思科制药有限公司 | Zeste增强子同源物2抑制剂及其用途 |
Also Published As
Publication number | Publication date |
---|---|
US11214561B2 (en) | 2022-01-04 |
CN110191879A (zh) | 2019-08-30 |
US20190345139A1 (en) | 2019-11-14 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
TWI573790B (zh) | 作為活化激酶抑制劑之經取代吲唑衍生物 | |
CN107286077B (zh) | 一种选择性的c-kit激酶抑制剂 | |
CN115232154A (zh) | 杂环类衍生物抑制剂、其制备方法和应用 | |
WO2021129824A1 (zh) | 新型K-Ras G12C抑制剂 | |
JPWO2018143315A1 (ja) | キナゾリン化合物 | |
JP2021506966A (ja) | Nlrp3インフラマソーム調節剤としてのスルホニル尿素誘導体 | |
TW201716415A (zh) | 作為蛋白質激酶之調節劑的掌性二芳基巨環 | |
JP6339241B2 (ja) | Alkキナーゼ阻害剤並びにその調製方法及び使用 | |
KR20210065097A (ko) | 치환된 인돌 및 이의 사용 방법 | |
JP2017512783A (ja) | ピペリジン−ジオン誘導体 | |
WO2018137639A1 (zh) | 一种组蛋白甲基转移酶ezh2抑制剂、其制备方法及其医药用途 | |
CN110857293B (zh) | 一种新型的喹啉衍生物抑制剂 | |
JP2018517672A (ja) | イソオキサゾリル置換イミダゾピリジン類 | |
EA018385B1 (ru) | АМИДОФЕНОКСИИНДАЗОЛЫ В КАЧЕСТВЕ ИНГИБИТОРОВ c-Met | |
WO2017152874A1 (zh) | 一种脲类化合物、其制备方法及其医药用途 | |
EA029771B1 (ru) | Химические соединения | |
WO2020224652A1 (zh) | 一类泛素化特异性蛋白酶抑制剂及其制备方法与应用 | |
JP2018530527A (ja) | Ezh2の阻害剤 | |
KR20200011977A (ko) | 아미노피리미딘 화합물, 이의 제조방법 및 용도 | |
CN112745298A (zh) | 多取代异吲哚啉类化合物、其制备方法、药物组合物及用途 | |
CA2892927A1 (en) | Use of maleimide derivatives for preventing and treating leukemia | |
CN105884695A (zh) | 杂环衍生物类酪氨酸激酶抑制剂 | |
WO2019024876A1 (zh) | 一种具有fgfr4抑制活性的醛基吡啶衍生物、其制备方法和应用 | |
KR20220122597A (ko) | 약학적 화합물 | |
WO2019157959A1 (zh) | 一种嘧啶类化合物、其制备方法及其医药用途 |
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: 18744953 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 18744953 Country of ref document: EP Kind code of ref document: A1 |