WO2024012461A1 - 一类甲基转移酶抑制剂及其用途 - Google Patents

一类甲基转移酶抑制剂及其用途 Download PDF

Info

Publication number
WO2024012461A1
WO2024012461A1 PCT/CN2023/106840 CN2023106840W WO2024012461A1 WO 2024012461 A1 WO2024012461 A1 WO 2024012461A1 CN 2023106840 W CN2023106840 W CN 2023106840W WO 2024012461 A1 WO2024012461 A1 WO 2024012461A1
Authority
WO
WIPO (PCT)
Prior art keywords
unsubstituted
substituted
ring
group
cancer
Prior art date
Application number
PCT/CN2023/106840
Other languages
English (en)
French (fr)
Inventor
富兴年
米沅
吴海平
王猛
王龙
石慧
韩阳
Original Assignee
上海赛岚生物科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 上海赛岚生物科技有限公司 filed Critical 上海赛岚生物科技有限公司
Publication of WO2024012461A1 publication Critical patent/WO2024012461A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic 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
    • C07D401/12Heterocyclic 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic 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
    • C07D405/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention relates to the field of pharmaceutical compounds. Specifically, the present invention provides a class of compounds for inhibiting PRC2 complexes, monomers, or combinations thereof, and their use in medicines and medicine combinations.
  • PRC2 Polycomb family repressive complex 2
  • PRC2 plays a key gene silencing role by selectively occupying chromatin transcriptional silencing sites.
  • PRC2 consists of 4 core components and several cofactors that regulate its activity: enhancer of zeste 1 or 2 (EZH1/2), embryonic ectoderm development (EED), suppressor of zeste 12 (SUZ12), and retinoblastoma Tumor-associated proteins 46 and 48 (RBAP46/RBAP48).
  • EZH1/2 is the catalytic subunit of methyltransferase responsible for the mono-, di-, and trimethylation of lysine 27 of histone H3 (H3K27me1/2/3).
  • EED enhances the enzymatic activity of PRC2 by binding to H3K27me3.
  • SUZ12 interacts with all other subunits and contributes to the stability of the complex.
  • RBAP46/RBAP48 also known as RBBP4/7 recognize histones H3 and H4 and may modulate the substrate specificity of PRC2.
  • PRC2 is currently the only known H3K27 methyltransferase, and its trimethylated H3K27 (H3K27me3) ultimately leads to gene silencing at specific sites by changing the chromatin structure (chromatin compaction).
  • the PRC2 protein is often mutated or dysregulated.
  • solid tumors including but not limited to ovarian cancer, breast cancer, prostate cancer, endometrial cancer, melanoma, bladder cancer, lung cancer , liver cancer
  • PRC2 components are overexpressed.
  • specific missense mutations have also been found to enhance PRC2 in patients with two types of hematological malignancies, DLBCL (diffuse large B-cell lymphoma) and FL (follicular lymphoma). catalytic activity.
  • DLBCL diffuse large B-cell lymphoma
  • FL follicular lymphoma
  • catalytic activity At the same time, some in vivo and in vitro results also show that the proliferation and metastasis of many tumor cell lines depend on PRC2 activity.
  • EZH2 or other PRC2 core components can inhibit many types of tumor cell lines (such as leukemia). , lymphoma, prostate cancer, breast cancer, lung cancer, and kidney cancer).
  • tumor cell lines such as leukemia.
  • lymphoma lymphoma
  • prostate cancer breast cancer
  • lung cancer and kidney cancer.
  • Current preclinical studies have shown that secondary mutations in EZH2 can produce acquired resistance, and its homolog EZH1 also has methyltransferase activity, both of which will limit the activity of EZH2 inhibitors.
  • the EZH2 subunit itself is inactive and must be assembled with SUZ12 and EED to form a PRC2 protein complex to produce methyltransferase activity. Therefore, the development of new inhibitors targeting the entire PRC2 protein complex has attracted attention.
  • IFN- ⁇ plays a key role in the pathogenesis of many autoimmune diseases, and relative deficiency of IFN- ⁇ is a characteristic of some chronic infections and tumor resistance in humans.
  • Inhibition of EZH2 significantly increases IFN- ⁇ expression in naive CD4+ T cells, thereby potentially promoting IFN- ⁇ -dependent anti-tumor immunity, but this inhibition of EZH2 to promote IFN- ⁇ expression depends on other components of PRC2.
  • PRC2 provides a pharmacological target for solid tumors and some hematological malignancies.
  • Targeted inhibition of the methyltransferase activity of EZH2 has been proven to be a successful cancer treatment strategy and may also play an important role in tumor immunity. effect.
  • the object of the present invention is to provide a small molecule compound for inhibiting the PRC2 protein complex or one or more protein combinations constituting the complex.
  • a first aspect of the present invention provides a compound represented by the following formula I, or its pharmaceutically acceptable salt or deuterated product, racemate mixture and its optical monomer product:
  • a ring is selected from the following group: 5-10 membered bridged ring (including carbocyclic ring and heterocyclic ring), 6-10 membered aromatic ring, 5-10 membered heteroaromatic ring;
  • Ring B is selected from the following group: 6-10 membered aromatic ring, 5-14 membered heteroaromatic ring;
  • Ring C is selected from the following group: 5-10-membered aromatic ring or partially saturated aromatic ring, 5-10-membered heteroaromatic ring or partially saturated heteroaromatic ring, 5-10-membered saturated or partially unsaturated carbocyclic ring (including fused rings) , bridged ring), 5-10 membered saturated or partially unsaturated heterocyclic ring (including fused ring and bridged ring);
  • L 1 and L 2 are each independently -(L) p -, and each L is independently selected from the group consisting of: chemical bond or none, or -O-, -CHR-, -CHR-NH-, carbonyl, S, -NR-, -NHC(O)-, -NHS(O) 2 -, -NHC(O)NH-, -NHC(S)NH-, -COO-, -OS(O) 2 -, -CHR- NR-, -C(R) 2 NR-, -C(R) 2 -; wherein each R is independently selected from the following group: H, substituted or unsubstituted C 1-4 alkyl, substituted or unsubstituted C 3-4 cycloalkyl, or substituted or unsubstituted 3-4 membered heterocycle;
  • n is selected from the following group: 0, 1, 2, 3, 4 or 5;
  • n is selected from the following group: 0, 1, 2, 3 or 4;
  • p is selected from the following group: 0, 1 or 2;
  • Each R 1 , R 2 and R 3 is each independently selected from the group consisting of: H, halogen, cyano, amino, nitro, hydroxyl, mercapto, aldehyde, carboxyl, sulfonyl, substituted or unsubstituted C 1 -C 6 alkyl group, substituted or unsubstituted C 1 -C 6 alkoxy group, substituted or unsubstituted C 1 -C 6 alkylamine group, substituted or unsubstituted C 1 -C 6 alkyl amido group, substituted or unsubstituted C 1 -C 6 alkyl amido group Substituted C 1 -C 6 alkyl C(O)O, or substituted or unsubstituted C 1 -C 6 alkyl OC(O), substituted or unsubstituted amide group, substituted or unsubstituted amine group (NH 2 ), substituted or unsubstituted C 2 -
  • X and Y are each independently selected from the following group: chemical bond, or -O-, -C(R 5 ) 2 -, -S-, -NR 5 -;
  • Each R 5 is independently selected from the group consisting of: H, halogen, cyano, amino, nitro, hydroxy, thiol, aldehyde, carboxyl, sulfonyl, substituted or unsubstituted C 1 -C 6 alkyl, substituted or Unsubstituted C 3 -C 6 carbocyclic ring (including saturated or partially unsaturated), substituted or unsubstituted 3-6 membered heterocyclic ring, substituted or unsubstituted C 6-10 aromatic ring, substituted or unsubstituted Substituted 5-12 membered heteroaryl ring, substituted or unsubstituted C 1 -C 6 alkyl C(O)O, or substituted or unsubstituted C 1 -C 6 alkyl OC(O), substituted or unsubstituted Substituted 5-9 membered heterospirocycle;
  • R 1 , R 2 or R 3 connected to adjacent ring atoms on the ring together form a parallel ring structure selected from the following group: substituted or unsubstituted C 6-10 aromatic ring, substituted or unsubstituted 5-12 membered heteroaromatic ring, substituted or unsubstituted C 3 -C 8 carbocyclic ring (including saturated or partially unsaturated), substituted or unsubstituted 3-8 membered heterocyclic ring (including saturated or partially unsaturated unsaturated); or two connected R 1 , R 2 or R 3 connected to the same carbon atom of an adjacent ring together with the adjacent ring form a 3-8 membered saturated or partially unsaturated spiro ring structure, Or a 3-8 membered saturated or partially unsaturated heterospirocyclic structure (herein refers to the ring formed by the substituents being 3-8 members, excluding adjacent rings);
  • the ring skeleton of each of the above-mentioned heterocyclic rings may contain 1-3 heteroatoms selected from boron, oxygen, sulfur, phosphorus and nitrogen; particularly, when the atoms are boron, sulfur, phosphorus and nitrogen, the The ring skeleton atoms mentioned above may be oxidized, such as S(O) or S(O) 2 ;
  • each chiral atom in the molecule can be in R configuration, S configuration, or a combination thereof.
  • Ring B is a 6-10 membered heteroaromatic ring
  • the R 1 is selected from the following group: H, halogen, cyano, amino, nitro, hydroxyl, mercapto, aldehyde, carboxyl , sulfonyl group, substituted or unsubstituted C 1 -C 6 alkyl group, substituted or unsubstituted C 1 -C 6 alkoxy group, substituted or unsubstituted C 1 -C 6 alkylamino group; or two connected to R 1 on adjacent ring atoms together constitute a cyclic structure selected from the following group: substituted or unsubstituted C 3 -C 6 carbocyclic ring (including saturated or partially unsaturated), substituted or unsubstituted 3-6 Heterocyclic ring (including saturated and partially unsaturated).
  • the B ring has a structure selected from the following group (where the connection site can be located on any ring atom):
  • the A ring has a structure selected from the following group (where the connection site can be located on any ring atom):
  • the R 2 has the structure shown in the following formula:
  • L 3 is selected from the following group: -C(R 5 ) 2 -C(O)-, -C(R 5 ) 2 -;
  • R 3 is selected from the following group: H, substituted or unsubstituted C 1 -C 6 alkyl;
  • R 6 is selected from the following group: substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 6 carbocyclic ring (including saturated or partially unsaturated), substituted or unsubstituted 3- 6-membered heterocyclic ring; the ring skeleton of the heterocyclic ring may contain 1-3 heteroatoms selected from oxygen and sulfur; and the ring skeleton atoms may be oxidized;
  • substitution means that the hydrogen atom on the corresponding group is replaced by one or more substituents selected from the following group: deuterium, tritium, halogen, oxo, hydroxyl, carboxyl, mercapto, benzyl, cyano, unsubstituted Or halogenated C 1 -C 6 alkyl, unsubstituted or halogenated C 3 -C 8 cycloalkyl, C 2 -C 10 alkenyl, C 1 -C 6 alkoxy, C 1 -C 6 alkyl Base - amino group, C 6 -C 10 aryl group, five- or six-membered heteroaryl group, 3-8 membered non-aromatic heterocyclic group.
  • substituents selected from the following group: deuterium, tritium, halogen, oxo, hydroxyl, carboxyl, mercapto, benzyl, cyano, unsubstituted Or halogenated C 1 -C 6 al
  • the R 2 is -CHR-C(O)NH-R 6 , wherein R is H or substituted or unsubstituted C 1-4 alkyl, substituted or unsubstituted C 1 -C 6 alkoxy, hydroxyl, substituted or unsubstituted amino; R 6 is selected from the following group: substituted or unsubstituted C 3 -C 6 carbocyclic ring (including saturated or partially unsaturated), substituted or unsubstituted 3- 6-membered heterocycle (including saturated or partially unsaturated).
  • the compound has the structure shown in the following formula II:
  • X 1 , X 2 , X 3 or X 4 are each independently selected from the following group: N, CR 3 ;
  • X 1 , X 2 , X 3 or X 4 are each independently selected from the following group: O, S, N, NR 3 or CR 3 ;
  • the L 1 is selected from the following group: chemical bond, -O-, -CHR-, carbonyl group, S, -NH-;
  • the L 2 is selected from the following group: chemical bond, -CHR- NH-, -CHR-O-, -CHR-S-, -(CHR) 2 -.
  • the second aspect of the present invention provides a pharmaceutical composition containing a therapeutically effective amount of the compound as described in the first aspect of the present invention, its pharmaceutically acceptable salt, racemate, optical One or more isomers, stereoisomers or tautomers, and one or more pharmaceutically acceptable carriers, excipients, adjuvants, excipients and/or diluents.
  • the third aspect of the present invention provides a compound described in the first aspect of the present invention, its racemate, photon Chemical isomer monomers and mixtures or pharmaceutically acceptable salts thereof in the preparation of drugs for the treatment or prevention of diseases related to mutations, overexpression, or dysregulation of H3K27 methylation levels associated with PRC2 complexes, monomers, or combinations thereof the use of.
  • the disease is selected from the group consisting of tumors and autoimmune diseases.
  • the disease is selected from the following group: lymphoma, hematological malignancy, sarcoma, prostate cancer, breast cancer, renal cancer, urothelial cancer, gastric cancer, ovarian cancer, endometrial cancer, cervical cancer Cancer, lung cancer, liver cancer, pancreatic cancer, colon cancer, head and neck cancer, brain tumors, melanoma, mesothelioma, gastrointestinal stromal tumor, psoriasis and lupus erythematosus.
  • the disease is selected from the following group: non-Hodgkin's lymphoma, follicular lymphoma, mantle cell lymphoma, peripheral T-cell lymphoma, Burkitt's lymphoma, Hodgkin's lymphoma Lymphoma, chronic lymphocytic leukemia, acute and chronic myeloid leukemia, acute and chronic lymphocytic leukemia, multiple myeloma, myelodysplastic syndrome, epithelioid sarcoma, rhabdomyosarcoma, liposarcoma, prostate cancer, breast cancer, renal Cancer, bladder cancer, upper urothelial cancer, gastric cancer, ovarian cancer, endometrial cancer, cervical cancer, lung cancer, liver cancer, pancreatic cancer, colon cancer, head and neck cancer, blastoma, glioblastoma, schwannoma , melanoma, mesothelioma, gastrointestinal stromal tumor
  • the halogen is F, Cl, Br or I.
  • C1-C6 alkyl refers to a straight-chain or branched alkyl group with 1 to 6 carbon atoms, including without limitation methyl, ethyl, propyl, isopropyl, butyl, etc. ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl and tert-butyl.
  • C1-C6 alkoxy refers to a straight-chain or branched chain alkoxy group with 1 to 6 carbon atoms, including without limitation methoxy, ethoxy, propoxy, Isopropoxy and butoxy, etc.
  • C2-C6 alkenyl refers to a straight-chain or branched alkenyl group containing one double bond with 2 to 6 carbon atoms, including without limitation vinyl, propenyl, butenyl , isobutenyl, pentenyl and hexenyl, etc.
  • C2-C6 alkynyl refers to a straight-chain or branched-chain alkynyl group having 2 to 6 carbon atoms and containing a triple bond, including without limitation ethynyl, propynyl, butynyl base, isobutynyl, pentynyl and hexynyl, etc.
  • C3-C10 cycloalkyl refers to a cyclic alkyl group having 3 to 10 carbon atoms in the ring, including without limitation cyclopropyl, cyclobutyl, cyclopentyl, cyclopentyl, Hexyl, cycloheptyl, cyclooctyl and cyclodecyl, etc.
  • C3-C8 cycloalkyl “C3-C7 cycloalkyl”
  • C3-C6 cycloalkyl have similar meanings.
  • C3-C10 cycloalkenyl refers to a cyclic alkenyl group with 3 to 10 carbon atoms in the ring, including without limitation cyclopropenyl, cyclobutenyl, cyclopentenyl , cyclohexenyl, cycloheptenyl, cyclooctenyl and Cyclodecylene etc.
  • C3-C7 cycloalkenyl has a similar meaning.
  • C1-C12 alkoxycarbonyl refers to an alkoxycarbonyl group having 1 to 12 carbon atoms in the alkyl chain, including without limitation methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, Isopropoxycarbonyl, tert-butoxycarbonyl, benzyloxycarbonyl, etc.
  • C1-C12 alkylaminocarbonyl refers to an alkylaminocarbonyl group having 1 to 12 carbon atoms in the alkyl chain, including without limitation methylaminocarbonyl, ethylaminocarbonyl, propylaminocarbonyl, Isopropylaminocarbonyl, tert-butylaminocarbonyl, benzylaminocarbonyl, dimethylaminocarbonyl, etc.
  • aromatic ring or “aryl” has the same meaning, and preferably "aryl” is “C6-C12 aryl” or “C6-C10 aryl”.
  • aryl is “C6-C12 aryl” or “C6-C10 aryl”.
  • C6-C12 aryl refers to an aromatic ring group with 6 to 12 carbon atoms that does not contain heteroatoms on the ring, such as phenyl, naphthyl, etc.
  • C6-C10 aryl has a similar meaning.
  • aromatic heterocycle or “heteroaryl” have the same meaning and refer to heteroaromatic groups containing one to more heteroatoms.
  • Heteroatoms referred to here include oxygen, sulfur and nitrogen.
  • the heteroaryl ring may be fused to an aryl, heterocyclyl or cycloalkyl ring, where the ring attached to the parent structure is the heteroaryl ring.
  • Heteroaryl groups may be optionally substituted or unsubstituted.
  • 3-12-membered heterocyclyl refers to a saturated or unsaturated 3-12-membered cyclic group containing 1 to 3 heteroatoms selected from oxygen, sulfur and nitrogen on the ring, for example Dioxolyl etc.
  • 3-7 membered heterocyclyl has a similar meaning.
  • substituted means that one or more hydrogen atoms on a specific group are replaced by a specific substituent.
  • Specific substituents are the substituents described accordingly in the foregoing text, or the substituents appearing in each embodiment.
  • a substituted group may have a substituent selected from a specific group at any substitutable position of the group, and the substituents may be the same or different at each position.
  • a cyclic substituent, such as heterocycloalkyl can be linked to another ring, such as cycloalkyl, to form a spirobicyclic system, for example, both rings having a common carbon atom.
  • substituents contemplated by the present invention are those that are stable or chemically achievable.
  • the substituents are for example (but not limited to): C1-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, C3-8 cycloalkyl, 3- to 12-membered heterocyclyl, aryl, Heteroaryl, halogen, hydroxyl, carboxyl (-COOH), C1-8 aldehyde, C2-10 acyl, C2-10 ester, C1-C12 alkoxycarbonyl, amino, alkoxy, C1-10 sulfonyl, etc. .
  • the present invention provides a class of compounds with the effect of regulating H3K27 methylation levels:
  • a ring is selected from the following group: 5-10 membered bridged ring (including carbocyclic ring and heterocyclic ring), 6-10 membered aromatic ring, 5-10 membered heteroaromatic ring (preferably benzene ring or 5-6 aromatic heterocycle);
  • Ring B is selected from the following group: 6-10-membered aromatic ring, 5-14-membered heteroaromatic ring (preferably 8-14-membered aromatic heterocyclic ring) or partially saturated 5-12-membered heteroaromatic ring (preferably 8-14-membered aromatic heterocyclic ring) ring);
  • Ring C is selected from the following group: 5-10-membered aromatic ring or partially saturated aromatic ring, 5-10-membered aromatic heterocyclic ring or partially saturated aromatic heterocyclic ring, 5-10-membered saturated or partially unsaturated carbocyclic ring (including fused rings) , bridged ring), 5-10 membered saturated or partially unsaturated heterocycle (including including fused rings and bridged rings);
  • L 1 and L 2 are each independently -(L) p -, and each L is independently selected from the group consisting of: chemical bond or none, or -O-, -CHR-, -CHR-NH-, carbonyl, S, -NH-, -NHC(O)-, -NHS(O) 2 -, -NHC(O)NH-, -NHC(S)NH-, -COO-, -OS(O) 2 -, -NR- , -C(R) 2 NH-, -C(R) 2 NR-; wherein, R is selected from the following group: H or substituted or unsubstituted C 1-4 alkyl or cycloalkyl, or substituted or unsubstituted 3-4 membered heterocyclic ring;
  • n is selected from the following group: 0, 1, 2, 3, 4 or 5;
  • n is selected from the following group: 0, 1, 2, 3 or 4;
  • p is selected from the following group: 0, 1 or 2;
  • Each R 1 , R 2 and R 3 is each independently selected from the group consisting of: H, halogen, cyano, amino, nitro, hydroxyl, mercapto, aldehyde, carboxyl, sulfonyl, substituted or unsubstituted C 1 -C 6 alkyl group, substituted or unsubstituted C 1 -C 6 alkoxy group, substituted or unsubstituted C 1 -C 6 alkylamine group, substituted or unsubstituted C 1 -C 6 alkyl amido group or ester group, Substituted or unsubstituted amide group, substituted or unsubstituted amine group, substituted or unsubstituted C 2 -C 6 alkenyl group, substituted or unsubstituted C 2 -C 6 alkynyl group, substituted or unsubstituted C 3 - C 6 carbocyclic ring (including saturated or partially unsaturated), substituted
  • the ring skeleton of each of the above-mentioned heterocyclic rings may contain 1-3 heteroatoms selected from boron, oxygen, sulfur, phosphorus and nitrogen; particularly, when the atoms are boron, sulfur, phosphorus and nitrogen, the The ring skeleton atoms mentioned above may be oxidized, such as S(O) or S(O) 2 .
  • the substitution means that the hydrogen atom on the corresponding group is replaced by one or more substituents selected from the following group: deuterium, tritium, halogen, hydroxyl, carboxyl, mercapto, benzyl , C 1 -C 12 alkoxycarbonyl group, C 1 -C 6 aldehyde group, amino group, C 1 -C 6 amide group, nitro group, cyano group, unsubstituted or halogenated C 1 -C 6 alkyl group, unsubstituted Substituted or halogenated C 3 -C 8 cycloalkyl, C 2 -C 10 alkenyl, C 1 -C 6 alkoxy, C 1 -C 6 alkyl-amino, C 6 -C 10 aryl, Five- or six-membered heteroaryl, five- or six-membered non-aromatic heterocyclic group, -O-(C 6 -C
  • Each chiral atom in the molecule can be in R configuration, S configuration, or a combination thereof; the alkene configuration can be cis, trans, or a mixture thereof.
  • compositions and methods of administration are provided.
  • the compound of the present invention has excellent methyltransferase inhibitory activity
  • the compound of the present invention and its various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates, and compounds containing the compound of the present invention are mainly Pharmaceutical compositions of active ingredients can be used to treat, prevent and alleviate related diseases caused by abnormal activity or expression of RC2 complexes, monomers, or combinations thereof.
  • the pharmaceutical composition of the present invention contains a compound of the present invention or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient or carrier within a safe and effective amount.
  • the “safe and effective dose” refers to the amount of compound that is sufficient to significantly improve the condition without causing serious side effects.
  • the pharmaceutical composition contains 1-2000 mg of the compound of the present invention/dose, more preferably, it contains 5-200 mg of the compound of the present invention/dose.
  • the "dose" is a capsule or tablet.
  • “Pharmaceutically acceptable carrier” refers to one or more compatible solid or liquid fillers or gel substances that are suitable for human use and must be of sufficient purity and low enough toxicity. "Compatibility” here means that the components of the composition can be blended with the compounds of the present invention and with each other without significantly reducing the efficacy of the compounds.
  • Examples of pharmaceutically acceptable carriers include cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid , magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as ), wetting agents (such as sodium lauryl sulfate), colorants, flavorings, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • cellulose and its derivatives such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.
  • gelatin such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.
  • solid lubricants such as
  • administration mode of the compounds or pharmaceutical compositions of the present invention is not particularly limited.
  • Representative administration modes include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration. .
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or compatibilizers, for example, Starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) Binders, for example, hydroxymethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and gum arabic; (c) Humectants, For example, glycerol; (d) disintegrants, such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) retarder, such as paraffin; (f) Absorption accelerators, such as quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glyceryl mono
  • Solid dosage forms such as tablets, dragees, capsules, pills and granules may be prepared using coatings and shell materials such as enteric casings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be in a delayed manner in a certain part of the digestive tract. Examples of embedding components that can be used are polymeric substances and waxy substances. If necessary, the active compounds can also be in microencapsulated form with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • liquid dosage forms may contain inert diluents conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances.
  • inert diluents conventionally employed in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-butanediol, dimethylformamide and oils,
  • compositions may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents.
  • Suspensions may contain, in addition to the active compound, suspending agents, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar or mixtures of these substances and the like.
  • suspending agents for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar or mixtures of these substances and the like.
  • compositions for parenteral injection may contain physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
  • Dosage forms for topical administration of the compounds of this invention include ointments, powders, patches, sprays and inhalants.
  • the active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants that may be required.
  • the compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
  • the compounds of the present invention can be administered together with other small molecule compounds to form a PROTAC, or with other large molecule compounds such as monoclonal antibodies to form an ADC.
  • a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, and the dosage when administered is a pharmaceutically effective dosage.
  • a mammal such as a human
  • the daily dose is usually 1 to 2000 mg, preferably 5 to 500 mg.
  • the specific dosage should also take into account factors such as the route of administration and the patient's health condition, which are all within the skill of a skilled physician.
  • Example 1 Compound 2-[2-[[4-[(8-chloro-1,7-naphthyridin-2-yl)amino]-1-bicyclo[2.2.2]octyl]methylamino]pyrimidine- Synthesis of 5-yl]-N-(oxetan-3-yl)acetamide (PA046)
  • Step 1 Combine ((4-aminobicyclo[2.2.2]oct-1-yl)methyl)carbamic acid tert-butyl ester (PA046-a) (100 mg, 0.393 mmol) and 2-chloro-8-methyl Oxy-1,7-naphthyridine (78 mg, 0.393 mmol) was dissolved in 2 ml of dimethyl sulfoxide, then potassium fluoride (114 mg, 1.97 mmol, 46.05 ⁇ l) and 2-[2 -(2-methoxyethoxy)ethoxy]ethanol 65 mg, 0.393 mmol). The reaction solution was stirred at 100 degrees Celsius for 15 hours. LCMS detects that the reaction is complete.
  • PA046-a 2-chloro-8-methyl Oxy-1,7-naphthyridine
  • Step 2 ((4-((8-chloro-1,7-naphthyridin-2-yl)amino)bicyclo[2.2.2]oct-1-yl)methyl)carbamic acid tert-butyl ester (PA046- b) (150 mg, 0.360 mmol) was dissolved in 2 ml of methylene chloride, and trifluoroacetic acid (41 mg, 0.360 mmol, 26.6 ⁇ l) was added. The reaction solution was stirred at 25 degrees Celsius for 2 hours. LCMS detects that the reaction is complete. The reaction solution was concentrated to dryness under reduced pressure.
  • Step 3 N-[4-(aminomethyl)-1-bicyclo[2.2.2]octyl]-8-chloro-1,7-naphthyridin-2-amino (PA046-c) (110 mg, 0.347 mmol) and 2-(2-chloropyrimidin-5-yl)acetate tert-butyl ester (95 mg, 0.417 mmol) were dissolved in dimethyl sulfoxide, and potassium fluoride (101 mg, 1.74 mmol) was added. and 2-[2-(2-methoxyethoxy)ethoxy]ethanol (57 mg, 0.347 mmol). The reaction solution was stirred at 100 degrees Celsius for 15 hours. LCMS detects that the reaction is complete.
  • Step 4 Combine 2-[2-[[4-[(8-chloro-1,7-naphthyridin-2-yl)amino]-1-bicyclo[2.2.2]octyl]methylamino]pyrimidine-5 -tert-butyl acetate (PA046-d) (140 mg, 0.275 mmol) was dissolved in 2 ml of dichloromethane, and trifluoroacetic acid (1.54 g, 13.5 mmol, 1 ml) was added. The reaction was stirred at 25°C for 2 hours. The target product was detected by LCMS.
  • reaction solution was concentrated to dryness under reduced pressure, 10 ml of 1 molar hydrochloric acid was added, and the mixture was extracted twice with 10 ml of ethyl acetate.
  • the inorganic phase was adjusted to pH 8 with sodium carbonate, and then extracted twice with 10 ml of ethyl acetate.
  • Step 5 2-[2-[[4-[(8-chloro-1,7-naphthyridin-2-yl)amino]-1-bicyclo[2.2.2]octyl]methylamino]pyrimidine-5 -]acetic acid (PA046-e) (90 mg, 0.199 mmol) and oxetane-3-amino (22 mg, 0.298 mmol) were dissolved in 2 ml of N,N-dimethylformamide , then add N, N-diisopropylethylamine (128.41 mg, 993.52 ⁇ mol, 173.05 ⁇ L, 5 equivalents) dropwise, and finally add HATU ((151.11 mg, 397.41 ⁇ mol, 2 equivalents).
  • the reaction solution is at 25 Stir for 2 hours at degrees Celsius.
  • LCMS detects that the reaction is complete.
  • the reaction solution is concentrated to dryness under reduced pressure and separated by a high performance liquid chromatography column (column: Boston Prime C18 150 mm * 30 mm * 5 microns; mobile phase: [water (ammonia solution v/ v)-ACN]; B%: 26%-56%, 9min) to obtain white solid 2-[2-[[4-[(8-chloro-1,7-naphthyridin-2-yl)amino]-1 - Bicyclo[2.2.2]octyl]methylamino]pyrimidin-5-yl]-N-(oxetan-3-yl)acetamide (PA046) (40 mg mg, 39.13% yield).
  • Example 2 Racemic compound 2-[2-[[4-[(8-methoxy-1,7-naphthyridin-2-yl)amino]phenyl]methylamino]pyrimidin-5-yl] -Synthesis of N-(oxetan-3-yl)propionamide (PA041) and its enantiomers PA042 and PA043
  • Step 1 Add anhydrous tetrahydrofuran (5 ml) to methyl 2-(2-chloropyrimidin-5-yl)acetate (PA041-a) (170 mg, 0.911 mmol), then add potassium tert-butoxide (102 mg , 0.911 mmol). The reaction solution was reacted at 20 degrees Celsius for 30 minutes. Methane iodide (129 mg, 0.911 mmol, 57 ⁇ l) was added under cooling in an ice-water bath. The reaction solution was continued to stir in the ice-water bath for 1 hour. LCMS showed the target product was produced.
  • Step 2 2-Chloro-8-methoxy-1,7-naphthyridine (PA041-c) (200 mg, 1.03 mmol) and tert-butyl N-[(4-aminophenyl)methyl]carbamate Ester (c1) (456 mg, 2.06 mmol) was dissolved in anhydrous tetrahydrofuran (1 ml), and sodium tert-butoxide (98.7 mg, 1.03 mmol), BINAP (191 mg, 0.308 mmol) and Pd 2 ( dba) 3 (94.1 mg, 0.103 mmol). The reaction solution was heated to 65 degrees Celsius and reacted for 1.5 hours. LCMS showed that the reaction was complete.
  • Step 3 N-[[4-[(8-methoxy-1,7-naphthyridin-2-yl)amino]phenyl]methyl]carbamic acid tert-butyl ester (PA041-d) (620 mg, 0.815 mmol, 50% purity) was dissolved in anhydrous dichloromethane (10 ml), and trifluoroacetic acid (4.62 g, 40.52 mmol, 3 ml) was added. The reaction solution was reacted at 25 degrees Celsius for 2 hours, and LCMS showed that the reaction was complete.
  • PA041-d N-[[4-[(8-methoxy-1,7-naphthyridin-2-yl)amino]phenyl]methyl]carbamic acid tert-butyl ester (PA041-d) (620 mg, 0.815 mmol, 50% purity) was dissolved in anhydrous dichloromethane (10 ml), and trifluoroacetic acid
  • Step 4 Add N-[4-(aminomethyl)phenyl]-8-methoxy-1,7-naphthyridin-2-amino (PA041-e) (153 mg, 0.548 mmol), methyl 2-(2-chloropyrimidin-5-yl)propionate (b) (110 mg, 0.548 mmol), potassium fluoride (159 mg, 2.74 mmol, 64 ⁇ L), and 2-[2-(2-methoxyethoxy)ethoxy]ethanol (90.0 mg, 0.548 mmol), the reaction solution was reacted at 100 degrees Celsius for 5 hours. LCMS showed the reaction was complete.
  • Step 5 Methyl 2-[2-[[4-[(8-methoxy-1,7-naphthyridin-2-yl)amino]phenyl]methylamino]pyrimidin-5-yl]propionate (PA041-f) (110 mg, 0.247 mmol), add tetrahydrofuran (1 ml)/water (1 ml) mixture, add sodium hydroxide (19.8 mg, 0.495 mmol) with stirring, and react the reaction solution at 25 degrees Celsius for 15 hours, LCMS showed that the reaction was complete.
  • Step 6 2-[2-[[4-[(8-methoxy-1,7-naphthyridin-2-yl)amino]phenyl]methylamino]pyrimidin-5-yl]propionic acid (PA041- g) (100 mg, 0.232 mmol) and oxetane-3-amine (16.9 mg, 0.232 mmol) were dissolved in DMF (2 mL), and DIEA (90.0 mg, 0.697 mmol, 121 ⁇ L ) and HATU (132 mg, 0.348 mmol), the reaction solution was reacted at 25 degrees Celsius for 2 hours. LCMS showed the reaction was complete.
  • reaction solution was concentrated under reduced pressure, and the residue was purified by reversed-phase high performance liquid chromatography (chromatographic column: Boston Prime C18 150 mm * 30 mm * 5 micron; mobile phase: [water (ammonia solution v/v)-acetonitrile]; gradient: 21 %-51%, 9 minutes) to obtain a yellow solid racemic compound 2-[2-[[4-[(8-methoxy-1,7-naphthyridin-2-yl)amino]phenyl]methylamino] Pyrimidin-5-yl]-N-(oxetan-3-yl)propionamide (PA041) (47 mg).
  • Example 3 Racemic compound 2-(2-((1-(4-((8-methoxy-1,7-naphthyridin-2-yl)amino)phenyl)ethyl)amino)pyrimidine -5-yl)acetic acid N-(oxetan-3-yl)acetamide (PA039) and its enantiomer PA061 and Synthesis of PA062
  • Step 1 Combine 1-(4-aminophenyl)ethanone 1-(4-aminophenyl)ethanone (b) (180 mg, 1.33 mmol), 2-chloro-8-methoxy-1,7-naphthalene Add pyridine (PA041-c) (250 mg, 1.28 mmol.), sodium tert-butoxide (140 mg, 1.46 mmol) and anhydrous tetrahydrofuran (10 ml) into the reaction flask, remove oxygen under reduced pressure, and replace it with nitrogen.
  • pyridine PA041-c
  • sodium tert-butoxide 140 mg, 1.46 mmol
  • anhydrous tetrahydrofuran 10 ml
  • Step 2 1-(4-((8-methoxy-1,7-naphthyridin-2-yl)amino)phenyl)ethanone (PA039-c) (360 mg, 1.23 mmol) and ammonium acetate (1.89g, 24.47 mmol) in methanol (30 ml), add sodium cyanoborohydride (308 mg, 4.90 mmol), and react the reaction solution at 40 degrees Celsius for 16 hours. LCMS showed that the target product was produced and the starting material remained. Add 300 mg of sodium cyanoborohydride and react the reaction solution at 40 degrees Celsius for 48 hours. LCMS showed the reaction was complete.
  • Step 3 N-(4-(1-aminoethyl)phenyl)-8-methoxy-1,7-naphthyridin-2-amino (PA039-d) (150 mg, 0.51 mmol) and 2 -(2-Chloropyrimidin-5-yl)acetate (d1) (115 mg, 0.505 mmol) in DMSO (4 ml), add 2-[2-(2-methoxyethoxy) Ethoxy]ethanol (TGME) (92 mg, 0.561 mg mol) and potassium fluoride (148 mg, 2.55 mmol.), the reaction solution was reacted at 100 degrees Celsius for 16 hours. LCMS showed the reaction was complete.
  • Step 4 2-(2-((1-(4-((8-methoxy-1,7-naphthyridin-2-yl)amino)phenyl)ethyl)amino)pyrimidin-5-yl)
  • PA039-e tert-butyl acetate
  • trifluoroacetic acid/dichloromethane 3 ml
  • trifluoroacetic acid/dichloromethane 1:5, V/V] 10 ⁇ 15 Celsius reaction for 16 hours.
  • LCMS showed the target product was produced.
  • Step 5 2-(2-((1-(4-((8-methoxy-1,7-naphthyridin-2-yl)amino)phenyl)ethyl)amino)pyrimidin-5-yl) Acetic acid (PA039-f) (82 mg, 0.075 mmol) in DMF (1 ml), add oxetane-3-amine (10 mg, 0.137 mmol), DIPEA (50 mg, 0.387 mmol) and HATU (45 mg, 0.118 mmol), and the reaction solution was reacted at 10 to 15 degrees Celsius for 2 hours. LCMS showed the target product was produced.
  • reaction solution was concentrated under reduced pressure to obtain a crude product, which was purified by reversed-phase high performance liquid chromatography (chromatographic column: YMC-Triart Preparation C18 150*40 mm*7 microns; mobile phase: [water (ammonia + ammonium bicarbonate)-acetonitrile]; gradient :7%-47%, 9 minutes), freeze-dried to obtain an off-white solid product 2-(2-((1-(4-((8-methoxy-1,7-naphthyridin-2-yl)amino) )phenyl)ethyl)amino)pyrimidin-5-yl)acetic acid N-(oxetan-3-yl)acetamide (PA039) (12 mg, 0.024 mmol, 31.8% yield).
  • chromatographic column YMC-Triart Preparation C18 150*40 mm*7 microns
  • mobile phase [water (ammonia + ammonium bicarbonate)-acetonitrile]
  • Step 6 70 mg of 2-(2-((1-(4-((8-methoxy-1,7-naphthyridin-2-yl)amino)phenyl)ethyl)amino)pyrimidine-5- Chiral separation of N-(oxetan-3-yl)acetamide (PA039) acetate (chromatographic column: DAICEL CHIRALCEL OD-H (250 mm*30 mm, 5 ⁇ m); mobile phase: [0.1 % ammonia ethanol]; B%: 45%-45%, min) to obtain two optical enantiomeric compounds: white solid PA061 (25 mg, 35.7% yield).
  • Example 5 Compound 2-(2-(((S)-1-(4-((8-chloro-1,7-naphthyridin-2-yl)amino)bicyclo[2.2.2]octane-1 Synthesis of -ethyl)amino)pyrimidin-5-yl)propionic acid (PA169-11) and diastereomers PA166, PA207
  • PA166 1 H NMR(400MHz,DMSO-d6) ⁇ (ppm)8.75(d,1H),8.15(s,2H),7.96(d,1H),7.87(d,1H),7.53(d,1H) ,7.18(s,1H),7.02(d,1H),6.74(d,1H),4.79–4.64(m,3H),4.41(t,1H),4.36(t,1H),3.98–3.89(m ,1H),3.42(t,1H),2.11–2.05(m,6H),1.62–1.51(m,6H),1.30(d,3H),1.01(d,3H); (ESI)m/z. [M+H] + 536.0.
  • PA207 1 H NMR(400MHz,DMSO-d6) ⁇ (ppm)8.76(d,1H),8.15(s,2H),7.95(d,1H),7.86(d,1H),7.53(d,1H) ,7.18(s,1H),7.02(d,1H),6.75(d,1H),4.79–4.64(m,3H),4.41(t,1H),4.35(t,1H),3.99–3.90(m ,1H),3.41(q,1H),2.10–2.06(m,6H),1.64–1.50(m,6H),1.30(d,3H),1.00(d,3H); (ESI)m/z. [M+H] + 536.0.
  • McCoys 5A Modified medium (ATCC); FBS (Gibco); 0.25% Trypsin/EDTA (Gibco); PBS (Hyclone); Penicillin/Streptomycin (100x) (Gibco); DMSO (Sigma); 384 cell culture plate ( Corning); 384 assay plate (Greiner); 96-well plate (compound dilution) (Haimen, Qunchao); T75 cell culture flask (Corning)
  • G401 cells were seeded in a 384-well cell culture plate, incubated overnight in a 37°C, 5% CO 2 cell culture incubator, and then treated with different concentrations of compounds for 72 hours. Take out the 384 cell culture plate, invert the cell culture plate in a centrifuge and centrifuge at 300 rpm for 1 minute. After centrifugation, wash twice with PBS, then add lysis solution, lyse for 60 minutes, then add neutralization buffer for mixing, and continue incubation. 30 minutes. Take 20ul (for detecting K27) and 10ul (for detecting H3) cell lysate from the 384 cell culture plate, transfer it to the 384 ELISA detection plate, and incubate at 4°C overnight.
  • % inhibition rate 100-100*(absorbance of compound-absorbance of positive control)/(absorbance of negative control-absorbance of positive control). Then use “Correction” in GraphPad Prism 5 software. Curve fit the inhibition rate data using the "Number (Inhibitor) vs. Response-Variable Slope (Four Parameters)" model.
  • the compound inhibition rate is calculated as follows:
  • % inhibition rate 100-100*(Absorbance of compound-Absorbance of positive control)/(Absorbance of negative control-Absorbance of positive control)
  • the compounds of the present invention were tested according to the above experimental methods. Some results are shown in Table 3. The test results are classified by A, B and C. A represents IC 50 ⁇ 0.1uM; B represents 0.1uM ⁇ IC 50 ⁇ 1uM; C represents IC 50 >1uM

Landscapes

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

Abstract

一类具有甲基转移酶抑制活性的化合物,具有如下式I所示的结构,可用于与PRC2复合物或其组成单体相关的疾病的治疗、预防。

Description

一类甲基转移酶抑制剂及其用途 技术领域
本发明涉及药物化合物领域,具体地,本发明提供了一类用于抑制PRC2复合物、单体、或其组合的化合物,及其用于药物、药物组合的用途。
背景技术
组蛋白上的位点特异性赖氨酸甲基化是控制和介导许多基本生物学过程的重要表观遗传机制之一。
表观基因组在转录活性区域和沉默区域表现不同,多梳家族抑制复合物2(PRC2)通过选择性占据染色质转录沉默位点,起到关键的基因沉默作用。PRC2由4个核心成分及几个调节其活性的辅因子组成:zeste 1或2的增强子(EZH1/2),胚胎外胚层发育(EED),zeste 12的抑制因子(SUZ12)以及视网膜母细胞瘤相关蛋白46和48(RBAP46/RBAP48)。EZH1/2是负责组蛋白H3的27位赖氨酸单、二、三甲基化(H3K27me1/2/3)的甲基转移酶催化亚基。而EED通过与H3K27me3结合来增强PRC2的酶活性。SUZ12与所有其他亚基相互作用,并有助于复合物的稳定性。RBAP46/RBAP48(也称为RBBP4/7)可识别组蛋白H3和H4,并可能调节PRC2的底物特异性。PRC2是目前已知唯一的H3K27的甲基转移酶,其介导的三甲基化的H3K27(H3K27me3)通过改变染色质结构(染色质压实)最终导致特定位点的基因沉默。
在包括肿瘤在内的人类疾病中,PRC2蛋白往往会发生突变或失调研究发现在许多实体瘤(包括但不限于卵巢癌、乳腺癌、前列腺癌、子宫内膜癌、黑色素瘤、膀胱癌、肺癌、肝癌)PRC2组分存在过表达,另外在DLBCL(弥漫性大B细胞淋巴瘤)、FL(滤泡性淋巴瘤)这两类血液系统恶性肿瘤患者中也发现特定的错义突变可以增强PRC2的催化活性。同时一些体内和体外结果也表明,许多肿瘤细胞系增殖和转移依赖PRC2活性,因而用基因敲低或敲除或者小分子药物抑制EZH2或其他PRC2核心成分,可以抑制许多类型肿瘤细胞系(如白血病、淋巴瘤、前列腺癌、乳腺癌、肺癌和肾癌)的增殖。目前临床前研究表明EZH2的二次突变可产生获得性耐药,且其同源性EZH1也具有甲基转移酶活性,均会导致EZH2抑制剂活性受限。同时,EZH2亚基本身是不活跃的,必须与SUZ12和EED组装成PRC2蛋白复合体才可以产生甲基转移酶活性。因此,针对整个PRC2蛋白复合体的新型抑制剂的开发引起了人们的关注。
此外,IFN-γ在许多自身免疫性疾病的发病机制中起着关键作用,IFN-γ的相对缺乏是人类一些慢性感染和肿瘤耐受性的特征。抑制EZH2显著增加幼稚CD4+T细胞中IFN-γ的表达,从而可能促进IFN-γ依赖性抗肿瘤免疫,但是这种抑制EZH2促进IFN-γ的表达需要依赖PRC2其他组分。
因此,PRC2为实体瘤和部分血液系统恶性肿瘤提供了药理学靶点,靶向抑制EZH2的甲基转移酶活性已被证明是一种成功的癌症治疗策略,同时也可能在肿瘤免疫中发挥重要作用。
发明内容
本发明的目的是提供一种用于抑制PRC2蛋白复合物或组成该复合物的一个或多个蛋白组合的小分子化合物。
本发明的第一方面,提供了一种如下式I所示的化合物,或其药学上可接受的盐或氘代产物、消旋体混合物及其光学单体产物:
其中,A环选自下组:5-10元的桥环(包括碳环、杂环)、6-10元的芳环、5-10元杂芳环;
B环选自下组:6-10元芳环、5-14元杂芳环;
C环选自下组:5-10元芳环或部分饱和芳环、5-10元杂芳环或部分饱和杂芳环、5-10元饱和或部分不饱和的碳环(包括稠和环、桥环),5-10元饱和或部分不饱和的杂环(包括稠和环、桥环);
L1和L2各自独立地为-(L)p-,且各个L各自独立地选自下组:化学键或无,或-O-、-CHR-、-CHR-NH-、羰基、S、-NR-、-NHC(O)-、-NHS(O)2-、-NHC(O)NH-、-NHC(S)NH-、-COO-、-O-S(O)2-、-CHR-NR-、-C(R)2NR-、-C(R)2-;其中,各个R各自独立地选自下组:H、取代或未取代的C1-4烷基、取代或未取代的C3-4环烷基、或取代或未取代的3~4元杂环;
n选自下组:0、1、2、3、4或5;
m选自下组:0、1、2、3或4;
p选自下组:0、1或2;
各个R1、R2和R3各自独立地选自下组:H、卤素、氰基、氨基、硝基、羟基、巯基、醛基、羧基、磺酰基、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C1-C6烷胺基、取代或未取代的C1-C6烷基酰胺基、取代或未取代的C1-C6烷基C(O)O、或取代或未取代的C1-C6烷基OC(O)、取代或未取代的酰胺基、取代或未取代的胺基(NH2)、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环(包括饱和或部分不饱和的情况),或为-X-Z-Y-R5
其中,X和Y各自独立地选自下组:化学键,或-O-、-C(R5)2-、-S-、-NR5-;
Z选自下组:C(O)、NH、CH=CH、-C(R5)2-、S(O)、S(O)2
各个R5各自独立地选自下组:H、卤素、氰基、氨基、硝基、羟基、巯基、醛基、羧基、磺酰基、取代或未取代的C1-C6烷基、取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环、取代或未取代的C6-10的芳环、取代或未取代的5-12元的杂芳环、取代或未取代的C1-C6烷基C(O)O、或取代或未取代的C1-C6烷基OC(O)、取代或未取代的5-9元的杂螺环;
或连接于环上相邻环原子上的两个相连R1、R2或R3共同构成选自下组的并环结构:取代或未取代的C6-10的芳环、取代或未取代的5-12元的杂芳环、取代或未取代的C3-C8碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-8元的杂环(包括饱和或部分不饱和的情况);或连接于相邻环同一个碳原子上的两个相连的R1、R2或R3与相邻环共同构成3-8元饱和或部分不饱和的螺环结构、或3-8元饱和或部分不饱和的杂螺环结构(此处指取代基形成的环为3-8元,不包括相邻的环);
其中,上述各个杂环的环骨架可以含有1-3个选自硼、氧、硫、磷和氮中的杂原子;特别地,当所述的原子为硼、硫、磷和氮时,所述的环骨架原子可以是被氧化的,如S(O)或S(O)2
除非特别说明,上述各式中,所述的取代指对应基团被一个或多个选自下组的取代基所取代:氘、氚、卤素、氧代、=NH、=N(C1-8烷基)、羟基、羧基、巯基、苄基、C1-C12烷氧基羰基、C1-C6醛基、氨基、C1-C6酰胺基、硝基、氰基、未取代或卤代的C1-C6烷基、C1-C6烷基-CN、未取代或卤代的C3-C8环烷基、C2-C10烯基、C2-C10炔基、C1-C6烷氧基、C1-C6烷基-胺基、C6-C10芳基、五元或六元杂芳基、3-8元非芳香性杂环基、-O-(C6-C10芳基)、-O-(五元或六元杂芳基)、C1-C12烷氨基羰基、未取代或卤代的C2-C10酰基、磺酰基(-SO2-OH)、磷酰基(-PO3-OH)、未取代或卤代的C1-C4烷基-S(O)2-、未取代或卤代C1-C4烷基-SO-、未取代或卤代C1-C4烷基氨基-S(O)2-;
其中,分子中各个手性原子可以为R构型,S构型,或其组合。
在另一优选例中,B环为6-10元的杂芳环,且所述的R1选自下组:H、卤素、氰基、氨基、硝基、羟基、巯基、醛基、羧基、磺酰基、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C1-C6烷胺基;或两个连接于相邻环原子上的R1共同构成选自下组的环状结构:取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环(包括饱和与部分不饱和的情况)。
在另一优选例中,所述的B环具有选自下组的结构(其中,连接位点可以位于任何环原子上):
在另一优选例中,所述的A环具有选自下组的结构(其中,连接位点可以位于任何环原子上):
在另一优选例中,所述的R2具有如下式所示的结构:
其中,L3选自下组:-C(R5)2-C(O)-、-C(R5)2-;
R3选自下组:H、取代或未取代的C1-C6烷基;
R6选自下组:取代或未取代的C1-C6烷基、取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环;所述杂环的环骨架可以含有1-3个选自氧和硫中的杂原子;且所述的环骨架原子可以是被氧化的;
所述的取代指对应基团上的氢原子被一个或多个选自下组的取代基所取代:氘、氚、卤素、氧代、羟基、羧基、巯基、苄基、氰基、未取代或卤代的C1-C6烷基、未取代或卤代的C3-C8环烷基、C2-C10烯基、C1-C6烷氧基、C1-C6烷基-胺基、C6-C10芳基、五元或六元杂芳基、3-8元非芳香性杂环基。
在另一优选例中,所述的R2为-CHR-C(O)NH-R6,其中R为H或取代或未取代的C1- 4烷基、取代或未取代C1-C6烷氧基、羟基、取代或未取代氨基;R6选自下组:取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环(包括饱和或部分不饱和的情况)。
在另一优选例中,所述的化合物具有如下式II所示的结构:
其中,X,Y各自独立地选自下组:O、NR3、C(R3)2;-C(=O)-
或所述的化合物具有如下式III所示的结构:
其中,X1、X2、X3或X4各自独立地选自下组:N、CR3
或所述的化合物具有如下式IV所示的结构:
其中,X1、X2、X3或X4各自独立地选自下组:O、S、N、NR3或CR3
为单键或双键;
其余各基团的定义如本发明第一方面中所述。
在另一优选例中,所述的L1选自下组:化学键、-O-、-CHR-、羰基、S、-NH-;所述的L2选自下组:化学键、-CHR-NH-、-CHR-O-、-CHR-S-、-(CHR)2-。
本发明的第二方面,提供了一种药物组合物,所述的药物组合物含有治疗有效量的如本发明第一方面所述的化合物、其可药用的盐、外消旋体、光学异构体、立体异构体或互变异构体中的一种或多种,以及一种或多种可药用的载体、赋形剂、佐剂、辅料和/或稀释剂。
本发明的第三方面,提供了一种本发明第一方面所述的化合物、其外消旋体、光 学异构体单体及其混合物或可药用盐在制备治疗或预防与PRC2复合物、单体、或其组合相关突变、过高表达、或H3K27甲基化水平失调相关的疾病的药物中的用途。
在另一优选例中,所述的疾病选自下组:肿瘤、自身免疫性疾病。
在另一优选例中,所述的疾病选自下组:淋巴瘤,恶性血液病,肉瘤,前列腺癌,乳腺癌,肾癌,尿路上皮癌,胃癌,卵巢癌,子宫内膜癌,宫颈癌,肺癌,肝癌,胰腺癌,结肠癌,头颈癌,脑瘤,黑色素瘤,间皮瘤、胃肠间质瘤、银屑病和红斑狼疮。
在另一优选例中,所述的疾病选自下组:非霍奇金性淋巴瘤、滤泡性淋巴瘤、套细胞淋巴瘤、外周T细胞淋巴瘤、伯基特淋巴瘤、霍奇金淋巴瘤、慢性淋巴细胞白血病、急性和慢性髓系白血病、急性和慢性淋巴细胞白血病、多发性骨髓瘤、骨髓增生异常综合征、上皮样肉瘤、横纹肌肉瘤、脂肪肉瘤,前列腺癌,乳腺癌,肾癌,膀胱癌、上尿路上皮癌,胃癌,卵巢癌,子宫内膜癌,宫颈癌,肺癌,肝癌,胰腺癌,结肠癌,头颈癌,随母细胞瘤、胶质细胞瘤、神经鞘瘤,黑色素瘤,间皮瘤、胃肠间质瘤、银屑病和红斑狼疮。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。
具体实施方式
本发明人通过广泛而深入的研究,首次意外地发现一类具有H3K27甲基化水平调节作用的化合物。在此基础上完成了本发明。
术语
在本发明中,所述卤素为F、Cl、Br或I。
在本发明中,除非特别指出,所用术语具有本领域技术人员公知的一般含义。本发明中,如果没有特别指明,所有化学式意在涵盖可能的任何光学或几何异构体(例如R型、S型或外消旋体,或者烯烃的顺反异构体等)。
在本发明中,术语“C1-C6烷基”是指具有1至6个碳原子的直链或支链烷基,非限制性地包括甲基、乙基、丙基、异丙基、丁基、异丁基、仲丁基、叔丁基、戊基和已基等;优选乙基、丙基、异丙基、丁基、异丁基、仲丁基和叔丁基。
在本发明中,术语“C1-C6烷氧基”是指具有1至6个碳原子的直链或支链烷氧基,非限制性地包括甲氧基、乙氧基、丙氧基、异丙氧基和丁氧基等。
在本发明中,术语“C2-C6烯基”是指具有2至6个碳原子的含有一个双键的直链或支链烯基,非限制性地包括乙烯基、丙烯基、丁烯基、异丁烯基、戊烯基和己烯基等。
在本发明中,术语“C2-C6炔基”是指具有2至6个碳原子的含有一个三键的直链或支链炔基,非限制性地包括乙炔基、丙炔基、丁炔基、异丁炔基、戊炔基和己炔基等。
在本发明中,术语“C3-C10环烷基”是指在环上具有3至10个碳原子的环状烷基,非限制性地包括环丙基、环丁基、环戊基、环己基、环庚基、环辛基和环癸基等。术语“C3-C8环烷基”、“C3-C7环烷基”、和“C3-C6环烷基”具有类似的含义。
在本发明中,术语“C3-C10环烯基”是指在环上具有3至10个碳原子的环状烯基,非限制性地包括环丙烯基、环丁烯基、环戊烯基、环己烯基、环庚烯基、环辛烯基和 环癸基烯等。术语“C3-C7环烯基”具有类似的含义。
在本发明中,术语“C1-C12烷氧羰基”是指在烷基链上具有1至12个碳原子的烷氧羰基,非限制性地包括甲氧羰基、乙氧羰基、丙氧羰基、异丙氧羰基、叔丁氧羰基、苄氧羰基等。
在本发明中,术语“C1-C12烷氨基羰基”是指在烷基链上具有1至12个碳原子的烷氨基羰基,非限制性地包括甲氨基羰基、乙氨基羰基、丙氨基羰基、异丙氨基羰基、叔丁氨基羰基、苄氨基羰基、二甲氨基羰基等。
在本发明中,术语“芳环”或“芳基”具有相同的含义,优选地“芳基”为“C6-C12芳基”或“C6-C10芳基”。术语“C6-C12芳基”是指在环上不含杂原子的具有6至12个碳原子的芳香族环基,如苯基、萘基等。术语“C6-C10芳基”具有类似的含义。
在本发明中,术语“芳香杂环”或“杂芳基”具有相同的含义,指包含一个到多个杂原子的杂芳族基团。这里所指的杂原子包括氧、硫和氮。例如呋喃基、噻吩基、吡啶基、吡唑基、吡咯基、N-烷基吡咯基、嘧啶基、吡嗪基、咪唑基、四唑基等。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。杂芳基可以是任选取代的或未取代的。
在本发明中,术语“3-12元杂环基”是指在环上含有1~3个选自氧、硫和氮中的杂原子的饱和或不饱和的3-12元环基,例如二氧杂环戊基等。术语“3-7元杂环基”具有类似的含义。
在本发明中,术语“取代”指特定的基团上的一个或多个氢原子被特定的取代基所取代。特定的取代基为在前文中相应描述的取代基,或各实施例中所出现的取代基。除非特别说明,某个取代的基团可以在该基团的任何可取代的位点上具有一个选自特定组的取代基,所述的取代基在各个位置上可以是相同或不同的。环状取代基,例如杂环烷基,可以与另一个环相连,例如环烷基,从而形成螺二环系,例如,两个环具有一个共用碳原子。本领域技术人员应理解,本发明所预期的取代基的组合是那些稳定的或化学上可实现的组合。所述取代基例如(但并不限于):C1-8烷基、C2-8烯基、C2-8炔基、C3-8环烷基、3-至12-元杂环基,芳基、杂芳基、卤素、羟基、羧基(-COOH)、C1-8醛基、C2-10酰基、C2-10酯基、C1-C12烷氧羰基、氨基、烷氧基、C1-10磺酰基等。
H3K27甲基化水平调节剂化合物
本发明中提供了一类具有H3K27甲基化水平调节作用的化合物:
其中,A环选自下组:5-10元的桥环(包括碳环、杂环)、6-10元的芳环、5-10元的杂芳环(优选为苯环或5-6元芳杂环);
B环选自下组:6-10元的芳环、5-14元杂芳环(优选8-14元芳杂环)或部分饱和的5-12元杂环(优选8-14元芳杂环);
C环选自下组:5-10元芳环或部分饱和芳环、5-10元芳杂环或部分饱和芳杂环、5-10元饱和或部分不饱和的碳环(包括稠和环、桥环),5-10元饱和或部分不饱和的杂环(包 括稠和环、桥环);
L1和L2各自独立地为-(L)p-,且各个L各自独立地选自下组:化学键或无,或-O-、-CHR-、-CHR-NH-、羰基、S、-NH-、-NHC(O)-、-NHS(O)2-、-NHC(O)NH-、-NHC(S)NH-、-COO-、-O-S(O)2-、-NR-、-C(R)2NH-、-C(R)2NR-;其中,R选自下组:H或取代或未取代的C1-4烷基或环烷基、或取代或未取代的3~4元杂环;
n选自下组:0、1、2、3、4或5;
m选自下组:0、1、2、3或4;
p选自下组:0、1或2;
各个R1、R2和R3各自独立地选自下组:H、卤素、氰基、氨基、硝基、羟基、巯基、醛基、羧基、磺酰基、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C1-C6烷胺基、取代或未取代的C1-C6烷基酰胺基或酯基、取代或未取代的酰胺基、取代或未取代的胺基、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环,或为-X-C(O)Y-R5;其中,X和Y各自独立地选自下组:化学键,或-O-、-C(R5)2-、-S-、-NR5-;R5选自下组:H、卤素、氰基、氨基、硝基、羟基、巯基、醛基、羧基、磺酰基、取代或未取代的C1-C6烷基、取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环;或连接于环上相邻环原子上的两个相连R1、R2或R3共同构成选自下组的并环结构:取代或未取代的C6-10的芳环、取代或未取代的5-12元的杂芳环、取代或未取代的C3-C8碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-8元的杂环(包括饱和或部分不饱和的情况);或连接于相邻环同一个碳原子上的两个相连的R1、R2或R3与相邻环共同构成饱和或部分不饱和的螺环结构、或杂螺环结构;
其中,上述各个杂环的环骨架可以含有1-3个选自硼、氧、硫、磷和氮中的杂原子;特别地,当所述的原子为硼、硫、磷和氮时,所述的环骨架原子可以是被氧化的,如S(O)或S(O)2
除非特别说明,上述各式中,所述的取代指对应基团上的氢原子被一个或多个选自下组的取代基所取代:氘、氚、卤素、羟基、羧基、巯基、苄基、C1-C12烷氧基羰基、C1-C6醛基、氨基、C1-C6酰胺基、硝基、氰基、未取代或卤代的C1-C6烷基、未取代或卤代的C3-C8环烷基、C2-C10烯基、C1-C6烷氧基、C1-C6烷基-胺基、C6-C10芳基、五元或六元杂芳基、五元或六元非芳香性杂环基、-O-(C6-C10芳基)、-O-(五元或六元杂芳基)、C1-C12烷氨基羰基、未取代或卤代的C2-C10酰基、磺酰基(-SO2-OH)、磷酰基(-PO3-OH)、未取代或卤代的C1-C4烷基-S(O)2-、未取代或卤代C1-C4烷基-SO-;
其中,分子中各个手性原子可以为R构型,S构型,或其组合;烯烃构型可以为顺式、或反式、或其混合物。
药物组合物和施用方法
由于本发明化合物具有优异的甲基转移酶抑制的活性,因此本发明化合物及其各种晶型,药学上可接受的无机或有机盐,水合物或溶剂合物,以及含有本发明化合物为主要活性成分的药物组合物可用于治疗、预防以及缓解由于RC2复合物、单体、或其组合的活性或表达量异常引起的相关疾病。
本发明的药物组合物包含安全有效量范围内的本发明化合物或其药理上可接受的盐及药理上可以接受的赋形剂或载体。其中“安全有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg本发明化合物/剂,更佳地,含有5-200mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。
“药学上可以接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能和本发明的化合物以及它们之间相互掺和,而不明显降低化合物的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。
本发明化合物或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、瘤内、直肠、肠胃外(静脉内、肌肉内或皮下)、和局部给药。
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性化合物与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和阿拉伯胶;(c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;(e)缓溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性化合物外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。
除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。
除了活性化合物外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。
用于局部给药的本发明化合物的剂型包括软膏剂、散剂、贴剂、喷射剂和吸入剂。活性成分在无菌条件下与生理上可接受的载体及任何防腐剂、缓冲剂,或必要时可能需要的推进剂一起混合。
本发明化合物可以单独给药,或者与其他药学上可接受的化合物联合给药。在一些优选的实施方式中,本发明的化合物可以与其他小分子化合物一同形成PROTAC,或者与其他大分子化合物例如单抗共同形成ADC施用。
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选5~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。
实施例1:化合物2-[2-[[4-[(8-氯-1,7-萘啶-2-基)氨基]-1-双环[2.2.2]辛基]甲氨基]嘧啶-5-基]-N-(氧杂环丁烷-3-基)乙酰胺(PA046)的合成
步骤1:将((4-氨基双环[2.2.2]辛-1-基)甲基)氨基甲酸叔丁酯(PA046-a)(100毫克,0.393毫摩尔)和2-氯-8-甲氧基-1,7-萘啶(78毫克,0.393毫摩尔)溶于2毫升二甲基亚砜中,然后加入氟化钾(114毫克,1.97毫摩尔,46.05微升)和2-[2-(2-甲氧基乙氧基)乙氧基]乙醇65毫克,0.393毫摩尔)。反应液在100摄氏度下搅15个小时。LCMS检测反应完成。反应液用10毫升的水溶解,用10毫升的乙酸乙酯萃取2次。合并有机相,用硫酸镁干燥,过滤,减压浓缩旋后硅胶柱层析纯化得到黄色固体((4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛-1-基)甲基)氨基甲酸叔丁酯(PA046-b)(150毫克,91.6%产率).得到的产物将用于下一步反应。LCMS:m/z=417.2(M+H)+
步骤2:将((4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛-1-基)甲基)氨基甲酸叔丁酯(PA046-b)(150毫克,0.360毫摩尔)溶于2毫升二氯甲烷中,加入三氟醋酸(41毫克,0.360毫摩尔,26.6微升)。反应液在25摄氏度下搅拌2小时。LCMS检测反应完成。反应液减压浓缩干。加入10毫升1摩尔的盐酸,用10毫升的乙酸乙酯萃取2次。 无机相用氢氧化钠调pH至8~9,然后用10毫升的乙酸乙酯萃取两次。合并有机相,用硫酸镁干燥,过滤,减压浓缩旋干,得到白色固体粗产物N-[4-(氨基甲基)-1-双环[2.2.2]辛基]-8-氯-1,7-萘啶-2-氨基(PA046-c)(110毫克,96.4%产率)直接用于下一步反应,无需进一步纯化。LCMS:m/z=317.1(M+H)+
步骤3:将N-[4-(氨基甲基)-1-双环[2.2.2]辛基]-8-氯-1,7-萘啶-2-氨基(PA046-c)(110毫克,0.347毫摩尔)和2-(2-氯嘧啶-5-基)乙酸叔丁酯(95毫克,0.417毫摩尔)溶于二甲基亚砜中,加入氟化钾(101毫克,1.74毫摩尔)和2-[2-(2-甲氧基乙氧基)乙氧基]乙醇(57毫克,0.347毫摩尔)。反应液在100摄氏度下搅拌15小时。LCMS检测反应完成。反应液用10毫升的水溶解,用10毫升的乙酸乙酯萃取2次。合并有机相,用硫酸镁干燥,过滤,减压浓缩旋干。经硅胶柱层析纯化得黄色黏稠状液体。2-[2-[[4-[(8-氯-1,7-萘啶-2-基)氨基]-1-双环[2.2.2]辛基]甲氨基]嘧啶-5-基]乙酸叔丁酯(PA046-d)(140毫克,79.2%产率)。LCMS:m/z=509.3(M+H)+
步骤4:将2-[2-[[4-[(8-氯-1,7-萘啶-2-基)氨基]-1-双环[2.2.2]辛基]甲氨基]嘧啶-5-基]乙酸叔丁酯(PA046-d)(140毫克,0.275毫摩尔)溶于2毫升的二氯甲烷中,加入三氟醋酸(1.54克,13.5毫摩尔,1毫升)。反应在25摄氏度下搅拌2小时。LCMS检测目标产物生成。反应液减压浓缩干后,加入10毫升1摩尔的盐酸,用10毫升乙酸乙酯萃取2次。无机相用碳酸钠调pH至8,然后用10毫升的乙酸乙酯萃取两次。合并有机相,用硫酸镁干燥,过滤,减压浓缩旋干,得到黄色固体粗产物2-[2-[[4-[(8-氯-1,7-萘啶-2-基)氨基]-1-双环[2.2.2]辛基]甲氨基]嘧啶-5-基]乙酸(PA046-e)(90毫克,72.2%产率),无需进一步纯化直接用于下一步反应。LCMS:m/z=453.3(M+H)+
步骤5:将2-[2-[[4-[(8-氯-1,7-萘啶-2-基)氨基]-1-双环[2.2.2]辛基]甲氨基]嘧啶-5-基]乙酸(PA046-e)(90毫克,0.199毫摩尔)和氧杂环丁烷-3-氨基(22毫克,0.298毫摩尔)溶于2毫升的N,N-二甲基甲酰胺中,然后滴加N,N-二异丙基乙胺(128.41毫克,993.52微摩尔,173.05微升,5当量),最后加入HATU((151.11毫克,397.41微摩尔,2当量)。反应液在25摄氏度下搅拌2小时。LCMS检测到反应完成。反应液减压浓缩干,经高效液相色谱柱分离(柱子:Boston Prime C18 150毫米*30毫米*5微米;流动相:[水(氨水v/v)-ACN];B%:26%-56%,9min)得到白色固体2-[2-[[4-[(8-氯-1,7-萘啶-2-基)氨基]-1-双环[2.2.2]辛基]甲氨基]嘧啶-5-基]-N-(氧杂环丁烷-3-基)乙酰胺(PA046)(40毫克mg,39.13%产率)。LCMS:m/z=508.2(M+H)+1H NMR(400MHz,DMSO-d6)δ(ppm)8.78(br d,1H),8.11(s,2H),7.96(d,1H),7.87(d,1H),7.53(d,1H),7.15(s,1H),7.02(d,1H),6.92(br t,1H),4.73-4.83(m,1H),4.62-4.73(m,2H),4.42(t,2H),3.22-3.27(m,2H),3.13(br d,2H),2.01-2.18(m,6H),1.46-1.65(m,6H)
实施例2:外消旋化合物2-[2-[[4-[(8-甲氧基-1,7-萘啶-2-基)氨基]苯基]甲氨基]嘧啶-5-基]-N-(氧杂环丁烷-3-基)丙酰胺(PA041),及其对映异构体PA042与PA043的合成
步骤1:2-(2-氯嘧啶-5-基)乙酸甲酯(PA041-a)(170毫克,0.911毫摩尔)中加入无水四氢呋喃(5毫升),然后加入叔丁醇钾(102毫克,0.911毫摩尔).反应液20摄氏度反应30分钟,冰水浴冷却下加碘甲烷(129毫克,0.911毫摩尔,57微升),反应液冰水浴继续搅拌反应1小时。LCMS显示目标产物生成。反应液加饱和氯化铵溶液(30毫升),用乙酸乙酯(30毫升*2)萃取,合并有机相用无水硫酸镁干燥,抽滤,滤液浓缩得到粗品。硅胶柱层析纯化得无色黏胶状粗产物2-(2-氯嘧啶-5-基)丙酸甲酯(PA041-b)(110毫克,60.1%纯度)。LCMS:m/z=200.9(M+H)+
步骤2:2-氯-8-甲氧基-1,7-萘啶(PA041-c)(200毫克,1.03毫摩尔)和N-[(4-氨基苯基)甲基]氨基甲酸叔丁酯(c1)(456毫克,2.06毫摩尔)溶解在无水四氢呋喃(1毫升)中,加叔丁醇钠(98.7毫克,1.03毫摩尔),BINAP(191毫克,0.308毫摩尔)和Pd2(dba)3(94.1毫克,0.103毫摩尔).反应液加热到65摄氏度反应1.5小时.LCMS显示反应完全。反应液加水(10毫升),用乙酸乙酯(10毫升*2)萃取。合并有机相用无水硫酸镁干燥,抽滤,滤液浓缩得到粗品。硅胶柱层析纯化得红色固体粗产物N-[[4-[(8-甲氧基-1,7-萘啶-2-基)氨基]苯基]甲基]氨基甲酸叔丁酯(PA041-d)(620毫克,47.0%纯度),无需纯化直接用于下一步反应。LCMS:m/z=381.2(M+H)+
步骤3:N-[[4-[(8-甲氧基-1,7-萘啶-2-基)氨基]苯基]甲基]氨基甲酸叔丁酯(PA041-d)(620毫克,0.815毫摩尔,50%纯度)溶于无水二氯甲烷(10毫升)中,加三氟乙酸(4.62克,40.52毫摩尔,3毫升)。反应液25摄氏度反应2小时,LCMS显示反应完全。反应液减压浓缩,残留物加盐酸(10毫升,1M),用乙酸乙酯萃取(10毫升*2)。水相用氢氧化钠调节pH到8~9,用乙酸乙酯萃取(10毫升*2),合并有机相用无水硫酸镁干燥,抽滤,滤液浓缩得到黄色固体粗产物N-[4-(氨基甲基)苯基]-8-甲氧基-1,7-萘啶-2-氨基(PA041-e)(230毫克,97.29%产率),无需进一步纯化,直接用于下一步反应。 LCMS:m/z=281.1(M+H)+
步骤4:在DMSO(5毫升)中加入N-[4-(氨基甲基)苯基]-8-甲氧基-1,7-萘啶-2-氨基(PA041-e)(153毫克,0.548毫摩尔),2-(2-氯嘧啶-5-基)丙酸甲酯(b)(110毫克,0.548毫摩尔),氟化钾(159毫克,2.74毫摩尔,64微升),和2-[2-(2-甲氧基乙氧基)乙氧基]乙醇(90.0毫克,0.548毫摩尔),反应液在100摄氏度反应5小时。LCMS显示反应完全。反应液中加水(10毫升),用乙酸乙酯(10毫升*2)萃取,合并有机相用无水硫酸镁干燥,抽滤,滤液浓缩得到粗品。粗品经硅胶柱层析纯化得黄色固体产物2-[2-[[4-[(8-甲氧基-1,7-萘啶-2-基)氨基]苯基]甲基氨基]嘧啶-5-基]丙酸甲酯(PA041-f)(230毫克,82.88%产率,87.82%纯度)。LCMS:m/z=445.2(M+H)+
步骤5:2-[2-[[4-[(8-甲氧基-1,7-萘啶-2-基)氨基]苯基]甲基氨基]嘧啶-5-基]丙酸甲酯(PA041-f)(110毫克,0.247毫摩尔)中加入四氢呋喃(1毫升)/水(1毫升)混合液,搅拌下加入氢氧化钠(19.8毫克,0.495毫摩尔),反应液25摄氏度反应15小时,LCMS显示反应完全。反应液减压浓缩,残留物加二氯甲烷(5毫升),用三氟乙酸调节pH到4,减压浓缩得到黄色固粗产物2-[2-[[4-[(8-甲氧基-1,7-萘啶-2-基)氨基]苯基]甲氨基]嘧啶-5-基]丙酸(PA041-g)(100毫克,93.87%产率),无需纯化,直接用于下一步反应。LCMS:m/z=431.2(M+H)+
步骤6:2-[2-[[4-[(8-甲氧基-1,7-萘啶-2-基)氨基]苯基]甲氨基]嘧啶-5-基]丙酸(PA041-g)(100毫克,0.232毫摩尔)和氧杂环丁烷-3-胺(16.9毫克,0.232毫摩尔)溶解在DMF(2毫升)中,加DIEA(90.0毫克,0.697毫摩尔,121微升)和HATU(132毫克,0.348毫摩尔),反应液25摄氏度反应2小时。LCMS显示反应完全。反应液减压浓缩,残留物经反相高效液相色谱纯化(色谱柱:Boston Prime C18 150毫米*30毫米*5微米;流动相:[水(氨水v/v)-乙腈];梯度:21%-51%,9分钟)得黄色固体消旋化合物2-[2-[[4-[(8-甲氧基-1,7-萘啶-2-基)氨基]苯基]甲氨基]嘧啶-5-基]-N-(氧杂环丁烷-3-基)丙酰胺(PA041)(47毫克)。LCMS:m/z=486.2(M+H)+1H NMR(400MHz,DMSO-d6)δ(ppm)9.58(s,1H),8.75(d,1H),8.18-8.22(m,2H),8.02(d,1H),7.94(d,2H),7.84(d,1H),7.61(t,1H),7.22-7.29(m,4H),4.64-4.78(m,3H),4.45(d,2H),4.38-4.43(m,1H),4.34(t,1H),4.03(s,3H),3.39-3.46(m,1H),1.30(d,3H).
经手性SFC拆分(色谱柱:DAICEL CHIRALCEL OD-H(250毫米*30毫米,5微米);流动相:[0.1%氨水乙醇];B%:45%-45%,分钟)得到(PA042)(18毫克,15.42%产率)(PA043)(15毫克,13.11%产率)两个光学对映异构体黄色固体化合物;
(PA042)LCMS:m/z=486.2(M+H)+1H NMR(400MHz,DMSO-d6)δ(ppm)9.57(s,1H),8.73(br d,J=6.13Hz,1H),8.20(s,2H),8.02(d,J=8.88Hz,1H),7.93(br d,J=8.50Hz,2H),7.80-7.89(m,1H),7.58(br t,J=6.25Hz,1H),7.16-7.35(m,4H),4.55-4.80(m,3H),4.45(br d,J=6.25Hz,2H),4.39-4.42(m,1H),4.34(br t,J=5.94Hz,1H),4.03(s,3H),3.44-3.47(m,1H),1.30(br d,J=7.13Hz,3H).
(PA043)LCMS:m/z=486.2(M+H)+1H NMR(400MHz,DMSO-d6)δ(ppm)9.57(s,1H),8.73(br d,1H),8.20(s,2H),8.02(d,1H),7.93(br d,2H),7.84(d,1H),7.58(br t,1H),7.16-7.34(m,4H),4.55-4.80(m,3H),4.45(br d,2H),4.37-4.43(m,1H),4.30-4.37(m,1H),4.03(s,3H),3.46(br s,1H),1.30(br d,3H).
实施例3:外消旋化合物2-(2-((1-(4-((8-甲氧基-1,7-萘啶-2-基)氨基)苯基)乙基)氨基)嘧啶-5-基)乙酸N-(氧杂环丁烷-3-基)乙酰胺(PA039)及其对映异构体PA061与 PA062的合成
步骤1:将1-(4-aminophenyl)ethanone 1-(4-氨基苯基)乙酮(b)(180毫克,1.33毫摩尔),2-氯-8-甲氧基-1,7-萘啶(PA041-c)(250毫克,1.28毫摩尔.),叔丁醇钠(140毫克,1.46毫摩尔)和无水四氢呋喃(10毫升)加入反应瓶中,减压除氧气,用氮气置换几次,BINAP(160毫克,257微摩尔)和Pd2(dba)3(100毫克,109微摩尔)加到反应液,80摄氏度反应2小时。LCMS显示目标产物生成。反应降温至室温,加饱和食盐水(10毫升),用二氯甲烷(15毫升)萃取2次,合并有机相用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗品.粗品经硅胶柱层析纯化得橙色固体化合物1-(4-((8-甲氧基-1,7-萘啶-2-基)氨基)苯基)乙酮(PA039-c)(392毫克,94.6%产率)。LCMS:m/z=294.0[M+1]+1H NMR(400MHz,DMSO-d6)δ(ppm)10.08(s,1H),8.19(d,2H),8.13(d,1H),7.97(d,J=8.63Hz,2H),7.91(d,1H),7.33(d,1H),7.29(d,1H),4.08(s,3H),2.54(s,3H)
步骤2:1-(4-((8-甲氧基-1,7-萘啶-2-基)氨基)苯基)乙酮(PA039-c)(360毫克,1.23毫摩尔)和乙酸铵(1.89g,24.47毫摩尔)在甲醇(30毫升)中,加氰基硼氢化钠(308毫克,4.90毫摩尔),反应液40摄氏度反应16小时。LCMS显示目标产物生成,原料剩余。加300毫克氰基硼氢化钠,反应液40摄氏度反应48小时。LCMS显示反应完成。反应液减压浓缩,加50毫升饱和食盐水,用甲醇/二氯甲烷(1:10,75毫升)萃取,水相用二氯甲烷(50毫升)萃取2次,合并有机相用无水硫酸镁干燥,抽滤,滤液浓缩得到黄色固体粗产物N-(4-(1-氨基乙基)苯基)-8-甲氧基-1,7-萘啶-2-氨基(PA039-d)(372毫克),无需进一步纯化,直接用于下一步反应。LCMS m/z=295.1[M+1]+
步骤3:N-(4-(1-氨基乙基)苯基)-8-甲氧基-1,7-萘啶-2-氨基(PA039-d)(150毫克,0.51毫摩尔)和2-(2-氯嘧啶-5-基)乙酸叔丁酯(d1)(115毫克,0.505毫摩尔)在DMSO(4毫升)中,加2-[2-(2-甲氧基乙氧基)乙氧基]乙醇(TGME)(92毫克,0.561毫 摩尔)和氟化钾(148毫克,2.55毫摩尔.),反应液100摄氏度反应16小时。LCMS显示反应完全。反应液降至室温,加水(25毫升)稀释,抽滤,滤饼用水(20毫升)洗涤,收集滤饼,冻干得到棕色固体粗产物2-(2-((1-(4-((8-甲氧基-1,7-萘啶-2-基)氨基)苯基)乙基)氨基)嘧啶-5-基)乙酸叔丁酯(PA039-e)(183毫克)。LCMS m/z=487.2[M+1]+
步骤4:2-(2-((1-(4-((8-甲氧基-1,7-萘啶-2-基)氨基)苯基)乙基)氨基)嘧啶-5-基)乙酸叔丁酯(PA039-e)(40毫克,0.082毫摩尔)和三氟乙酸/二氯甲烷(3毫升)[三氟乙酸/二氯甲烷=1:5,V/V]混合物10~15摄氏度反应16小时。LCMS显示目标产物生成。减压浓缩得粗品,用甲基叔丁基醚(10毫升)打浆,用石油醚稀释(10毫升),静止寖壁掉溶液,残留物减压干燥得棕色浆状粗产物2-(2-((1-(4-((8-甲氧基-1,7-萘啶-2-基)氨基)苯基)乙基)氨基)嘧啶-5-基)乙酸(PA039-f)(82毫克,三氟乙酸盐),无需进一步纯化,直接用于下一步反应。LCMS m/z=431.1[M+1]+
步骤5:2-(2-((1-(4-((8-甲氧基-1,7-萘啶-2-基)氨基)苯基)乙基)氨基)嘧啶-5-基)乙酸(PA039-f)(82毫克,0.075毫摩尔)在DMF(1毫升)中,加氧杂环丁烷-3-胺(10毫克,0.137毫摩尔),DIPEA(50毫克,0.387毫摩尔)和HATU(45毫克,0.118毫摩尔),反应液在10~15摄氏度反应2小时。LCMS显示目标产物生成。反应液减压浓得粗品,经反相高效液相色谱纯化(色谱柱:YMC-Triart制备C18 150*40毫米*7微米;流动相:[水(氨水+碳酸氢铵)-乙腈];梯度:7%-47%,9分钟),冻干得类白色固体产物2-(2-((1-(4-((8-甲氧基-1,7-萘啶-2-基)氨基)苯基)乙基)氨基)嘧啶-5-基)乙酸N-(氧杂环丁烷-3-基)乙酰胺(PA039)(12毫克,0.024毫摩尔,31.8%产率)。LCMS m/z=486.2[M+1]+1H NMR(400MHz,DMSO-d6)δ(ppm)9.56(s,1H),8.78(d,1H),8.11(s,2H),8.01(d,1H),7.91(d,2H),7.83(d,1H),7.51(d,1H),7.33(d,2H),7.18-7.25(m,2H),5.00-5.09
(m,1H),4.70-4.80(m,.01H),4.64-4.69(m,2H),4.39(t,2H),4.03(s,3H),3.22(s,2H),1.43(d,3H)
步骤6:70毫克2-(2-((1-(4-((8-甲氧基-1,7-萘啶-2-基)氨基)苯基)乙基)氨基)嘧啶-5-基)乙酸N-(氧杂环丁烷-3-基)乙酰胺(PA039)经手性拆分(色谱柱:DAICEL CHIRALCEL OD-H(250毫米*30毫米,5微米);流动相:[0.1%氨水乙醇];B%:45%-45%,min)得两光学对映异构体化合物:白色固体PA061(25毫克,35.7%产率)。LCMS m/z=486.1[M+1]+;Chiral SFC:ee%=98.32%,1H NMR(400MHz,DMSO-d6)δ9.56(s,1H),8.78(d,1H),8.11(s,2H),8.01(d,1H),7.91(d,2H),7.83(d,1H),7.51(d,1H),7.33(d,2H),7.18-7.25(m,2H),5.00-5.09(m,1H),4.70-4.80(m,1H),4.64-4.69(m,2H),4.39(t,2H),4.03(s,3H),3.22(s,2H),1.43(d,H);白色固体化合物PA062(25毫克,35.7%产率)LCMS:m/z=486.1[M+1]+;Chiral SFC ee%=87.62%;1H NMR(400MHz,DMSO-d6)δ9.56(s,1H),8.78(d,1H),8.11(s,2H),8.01(d,1H),7.91(d,2H),7.83(d,1H),7.51(d,1H),7.33(d,2H),7.18-7.25(m,2H),5.00-5.09(m,1H),4.70-4.80(m,1H),4.64-4.69(m,2H),4.39(t,2H),4.03(s,3H),3.22(s,2H),1.43(d,3H)
实施例4:化合物(S)-3-(2-(1-(4-(8-氯-7-甲基喹啉-2-胺基)-3-氟苯基)乙基胺基)嘧啶-5-基甲基胺基)硫杂环丁烷-1,1-二酮(PA167)的合成
1.化合物(S,E)-N-(1-(3-氟-4-硝基苯基亚乙基)-2-甲基丙烷-2-磺酰亚胺(PA167-2)的制备
化合物3-氟-4-硝基苯乙酮(PA167-1)(5.00克,27.3毫摩尔)溶于50毫升四氢呋喃,加入(S)-叔丁基亚磺酰胺(4.96克,41.0毫摩尔)与Ti(OEt)4(12.46克,54.6毫摩尔,11.4毫升)。反应液75℃搅拌16小时,冷却到室温后冰-水(30毫升)淬灭反应,加入100毫升乙酸乙酯后室温搅拌15分钟。过滤,滤饼用3*20毫升乙酸乙酯洗涤。合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,浓缩得7.80克黄色(S,E)-N-(1-(3-氟-4-硝基苯基亚乙基)-2-甲基丙烷-2-亚磺酰胺(PA167-2)粗产品。(ESI)m/z.[M+H]+287.0。
2.(S)-N-((S)-1-(3-氟-4-硝基苯基)乙基)-2-甲基丙烷-2-亚磺酰胺(PA167-3)的制备
(S,E)-N-(1-(3-氟-4-硝基苯基亚乙基)-2-甲基丙烷-2-磺酰亚胺(PA167-2)粗产品(7.8克,27.2毫摩尔)溶于100毫升四氢呋喃,冷却到零下50℃,分批加入硼氢化钠(4.12克,109.0毫摩尔)。反应混合物缓慢升至0~25度搅拌3小时后加入20毫升饱和氯化铵水溶液小心淬灭,乙酸乙酯萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥,过滤、浓缩后硅胶柱层析纯化得到黄色油状(S)-N-((S)-1-(3-氟-4-硝基苯基)乙基)-2-甲基丙烷-2-亚磺酰胺(PA167-3)(5.17克,17.9毫摩尔,65.8%产率)。(ESI)m/z.[M+H]+289.0。
3.化合物(S)-1-(3-氟-4-硝基苯基)乙基-1-胺(PA167-4)的合成
化合物(S)-N-((S)-1-(3-氟-4-硝基苯基)乙基)-2-甲基丙烷-2-亚磺酰胺(PA167-3)(1.0克,3.47毫摩尔)溶于8毫升1,4-二氧六环,然后滴加8毫升4M的盐酸。反应液室温搅拌3小时后反应完全。旋去溶剂后加水,并用乙酸乙酯萃取。有机相用饱和碳酸氢钠水溶液洗涤,无水硫酸钠干燥,过滤、浓缩后得640毫克黄色粘液粗产物(S)-1-(3-氟-4-硝基苯基)乙基-1-胺(PA167-4)。(ESI)m/z.[M+H]+185.0。
4.化合物(S)-(1-(3-氟-4-硝基苯基)乙基)碳酸叔丁酯(PA167-5)的合成
(S)-1-(3-氟-4-硝基苯基)乙基-1-胺(PA167-4)(650毫克,3.5毫摩尔)溶于5毫升四氢呋喃,加入Boc2O(1.54克,7.06毫摩尔)与三乙胺(1.07克,10.59毫摩尔,3.77毫升)后,室温搅拌2小时。反应液浓缩后加水,用乙酸乙酯萃取,有机相用饱和实验 室洗涤,无水硫酸钠干燥,过滤、浓缩后硅胶柱层析纯化得黄色固体(S)-(1-(3-氟-4-硝基苯基)乙基)碳酸叔丁酯(PA167-5)(700.0毫克,2.5毫摩尔,69.8%产率)。(ESI)m/z.[M+H]+229.0。
5.化合物(S)-(1-(4-氨基-3-氟-苯基)乙基)碳酸叔丁酯(PA167-6)的合成
(S)-(1-(3-氟-4-硝基苯基)乙基)碳酸叔丁酯(PA167-5)(700.0毫克,2.5毫摩尔)溶于10毫升甲醇,加入70毫克Pd/C后室温搅拌2小时,反应完全。反应混合物用硅藻土过滤,甲醇洗涤。有机相浓缩得黄色粘液粗产物(S)-(1-(4-氨基-3-氟-苯基)乙基)碳酸叔丁酯(PA167-6)(540.0毫克,2.1毫摩尔,86.2%产率)。(ESI)m/z.[M+H]+277。
6.化合物(S)-(1-(4-((8-氯-7-甲基喹啉-2-基)氨基)-3-氟-苯基)乙基)碳酸叔丁酯(PA167-7)的合成
混合物2,8-二氯-7-甲基喹啉(366.9毫克,1.7毫摩尔),(S)-(1-(4-氨基-3-氟-苯基)乙基)碳酸叔丁酯(PA167-6)(400.0毫克,1.6毫摩尔),叔丁醇钠(151.2毫克,1.6毫摩尔),与BINAP(293.8毫克,471.9微摩尔)中加入10毫升干燥四氢呋喃,氮气置换后加入Pd2(dba)3(144.0毫克,157.3微摩尔)。反应在氮气保护下65℃搅拌16小时。反应液过滤后滤液中加入100毫升水,并用乙酸乙酯萃取2次,合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤、浓缩后硅胶柱层析纯化得棕色固体化合物(S)-(1-(4-((8-氯-7-甲基喹啉-2-基)氨基)-3-氟-苯基)乙基)碳酸叔丁酯(PA167-7)(400.0毫克,651.3微摩尔,41.4%产率)。(ESI)m/z.[M+H]+430.2
7.化合物(S)-N-(4-(1-氨基乙基)-2-氟苯基)-8-氯-7-甲基喹啉-2-胺(PA167-8)的合成
(S)-(1-(4-((8-氯-7-甲基喹啉-2-基)氨基)-3-氟-苯基)乙基)碳酸叔丁酯(PA167-7)(400.0毫克,93.4微摩尔)中加入3毫升二氯甲烷,室温搅拌下滴加5毫升三氟乙酸并搅拌2小时。反应混合物浓缩后加水稀释,并用饱和碳酸钠水溶液调节pH8-9,然后用二氯甲烷萃取,合并有机相用无水硫酸钠干燥,过滤、浓缩后硅胶柱层析纯化得黄色固体化合物(S)-N-(4-(1-氨基乙基)-2-氟苯基)-8-氯-7-甲基喹啉-2-胺(PA167-8)(190.0毫克,518.5微摩尔,55.7%产率)。(ESI)m/z.[M+H]+330.0。
8.化合物(S)-2-((1-(4-((8-氯-7-甲基喹啉-2-基)氨基)-3-氟苯基)乙基)氨基)嘧啶-5-甲醛(PA167-9)的合成
氮气保护下,混合物(S)-N-(4-(1-氨基乙基)-2-氟苯基)-8-氯-7-甲基喹啉-2-胺(PA167-8)(90毫克,272.9微摩尔),2-氯嘧啶-5-甲醛(38.9毫克,272.9微摩尔),TGME(44.8毫克,272.9微摩尔)中加入氟化钾(79.3毫克,1.4毫摩尔)与2毫升二甲基亚砜。反应混合物加热到110℃搅拌16小时。加入10毫升水后用乙酸乙酯萃取,合并有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤、浓缩后硅胶柱层析分离得黄色固体(S)-2-((1-(4-((8-氯-7-甲基喹啉-2-基)氨基)-3-氟苯基)乙基)氨基)嘧啶-5-甲醛(PA167-9)(55.0毫克,118.6微摩尔,43.5%产率)。(ESI)m/z.[M+H]+436.2。
9.化合物(S)-3-(((2-((1-(4-((8-氯-7-甲基喹啉-2-基)氨基)-3-氟苯基)乙基)氨基)嘧啶-5-基)甲基)氨基)-1,1-二氧代硫代环丁烷(PA167)的合成
化合物(S)-2-((1-(4-((8-氯-7-甲基喹啉-2-基)氨基)-3-氟苯基)乙基)氨基)嘧啶-5-甲醛(PA167-9)(55.0毫克,126.2微摩尔),1,1-二氧代硫杂环丁烷-3-胺(19.9毫克,164.0微摩尔)溶于3毫升干燥甲醇,50℃搅拌反应18小时后冷却到室温。加入1 毫升二氯甲烷与氰基硼氢化钠(15.9毫克,252.4微摩尔),室温搅拌反应5小时。加入2滴水淬灭反应,反应液后处理后加入甲醇并用制备HPL纯化得白色固体化合物(S)-3-(((2-((1-(4-((8-氯-7-甲基喹啉-2-基)氨基)-3-氟苯基)乙基)氨基)嘧啶-5-基)甲基)氨基)-1,1-二氧代硫代环丁烷(PA167)。1H NMR(400MHz,DMSO-d6)δ9.20(d,1H),9.08(t,1H),8.41(s,0H),8.16(s,2H),8.04(d,1H),7.59(dd,2H),7.30(d,1H),7.27–7.17(m,3H),5.05(p,1H),4.28–4.20(m,2H),3.84(ddd,2H),3.46–3.40(m,1H),3.36(s,2H),2.49(s,3H),1.41(d,3H);(ESI)m/z.[M+H]+541.2。
实施例5:化合物2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)氨基)嘧啶-5-基)丙酸(PA169-11)及非对映异构体PA166,PA207的合成
1.化合物4-(叔丁氧羰基氨基)双环[2.2.2]辛烷-1-羧酸甲酯(PA169-1)的合成
4-氨基双环[2.2.2]辛烷-1-羧酸甲酯(2克,10.9毫摩尔)溶于20毫升二氯甲烷,加入三乙胺(3.31克,32.7毫摩尔)与碳酸二叔丁酯(2.86克,13.1毫摩尔),反应液室温搅拌16小时。加水后用二氯甲烷萃取,有机相饱和食盐水洗涤,二氯甲烷萃取,无水硫酸钠干燥,过滤、浓缩得2.20克白色粗产物4-(叔丁氧羰基氨基)双环[2.2.2]辛烷-1-羧酸甲酯(PA169-1)。(ESI)m/z.[M+H]+228.0。
2.化合物4-(叔丁氧羰基氨基)双环[2.2.2]辛烷-1-羧酸(PA169-2)的合成
4-(叔丁氧羰基氨基)双环[2.2.2]辛烷-1-羧酸甲酯(PA169-1)(2.2g,7.76mmol)溶于15毫升甲醇,加入5毫升水和氢氧化锂(371.86mg,15.53mmol),室温搅拌4小 时。反应液用1N盐酸溶液条件pH6,乙酸乙酯萃取后饱和食盐水洗涤,有机相用无水硫酸钠干燥,过滤、浓缩后得白色粗产物4-(叔丁氧羰基氨基)双环[2.2.2]辛烷-1-羧酸(PA169-2)。(ESI)m/z.[M+H]+214.0。
3.化合物N-[4-[甲氧基(甲基)氨基甲酰基]-1-双环[2.2.2]辛基]碳酸叔丁酯(PA169-3)的合成
4-(叔丁氧羰基氨基)双环[2.2.2]辛烷-1-羧酸(PA169-2)(1.7g,6.31mmol)和二异丙基乙基胺(1.63g,12.62mmol)溶于20毫升DMF,加入N,O-二甲基羟胺(424.10mg,6.94mmol)与HATU(2.64g,6.94mmol),反应混合物室温搅拌1小时,反应液过滤后加水稀释,用乙酸乙酯萃取,有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤、浓缩后硅胶柱层析纯化得白色固体化合物N-[4-[甲氧基(甲基)氨基甲酰基]-1-双环[2.2.2]辛基]碳酸叔丁酯(PA169-3)(1.60g,4.61mmol,73.03%yield,90%purity)。(ESI)m/z.[M+H]+313.20。
4.化合物4-氨基-N-甲氧基-N-甲基双环[2.2.2]辛基-1-羧酸酰胺(PA169-4)的合成
化合物N-[4-[甲氧基(甲基)氨基甲酰基]-1-双环[2.2.2]辛基]碳酸叔丁酯(PA169-3)(1.6g,5.12mmol)溶于10毫升二氯甲烷,加入3毫升三氟乙酸,反应液室温搅拌2小时。反应液浓缩后加入碳酸氢钠水溶液调节pH10,二氯甲烷萃取后饱和食盐水洗涤,无水硫酸钠干燥,过滤、浓缩得1.00克白色固体粗产物4-氨基-N-甲氧基-N-甲基双环[2.2.2]辛基-1-羧酸酰胺(PA169-4)。(ESI)m/z.[M+H]+213.0。
5.化合物4-[(8-氯-1,7-萘啶-2-基)氨基]-N-甲氧基-N-甲基-双环[2.2.2]辛基-1-羧酸酰胺(PA169-5)的合成
化合物4-氨基-N-甲氧基-N-甲基双环[2.2.2]辛基-1-羧酸酰胺(PA169-4)(0.95g,4.48mmol)溶于15毫升二甲基亚砜,加入2,8-二氯-1,7-萘啶(1.25g,6.27mmol)、氟化钾(1.30g,22.38mmol)与TGME(734.80mg,4.48mmol)。反应液加热100度搅拌16小时。反应液过滤后加入30毫升水,用乙酸乙酯萃取后有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤、浓缩后粗产物用硅胶柱层析纯化得白色固体化合物4-[(8-氯-1,7-萘啶-2-基)氨基]-N-甲氧基-N-甲基-双环[2.2.2]辛基-1-羧酸酰胺(PA169-5)(1.00克,2.40毫摩尔,53.6%产率,90%纯度)。(ESI)m/z.[M+H]+375.0。
6.化合物1-[4-[(8-氯-1,7-萘啶-2-基)氨基]-1-双环[2.2.2]辛基]乙酮(PA169-6)的合成
化合物4-[(8-氯-1,7-萘啶-2-基)氨基]-N-甲氧基-N-甲基-双环[2.2.2]辛基-1-羧酸酰胺(PA169-5)(1g,2.67mmol)溶于10毫升干燥四氢呋喃,冷却到0度,氮气保护下滴加甲基溴化镁-四氢呋喃溶液(3M,4.45mL)。反应液缓慢升至40度反应16小时。冰水浴冷却下小心滴加15毫升饱和氯化铵水溶液,乙酸乙酯萃取后用饱和食盐水洗涤,无水硫酸钠干燥,过滤、浓缩得800毫克黄色固体粗产物1-[4-[(8-氯-1,7-萘啶-2-基)氨基]-1-双环[2.2.2]辛基]乙酮(PA169-6)。(ESI)m/z.[M+H]+330.10。
7.化合物(S,E)-N-(1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)亚乙基)-2-甲基丙烷-2-亚磺酰胺(PA169-7)的合成
化合物1-[4-[(8-氯-1,7-萘啶-2-基)氨基]-1-双环[2.2.2]辛基]乙酮(PA169-6)(0.15克,454.8微摩尔)溶于10毫升干燥四氢呋喃,加入四乙氧基钛(207.5毫克,909.6微摩尔),反应液加热75度搅拌16小时。反应液冷却后加入20毫升乙酸乙酯与10 毫升水,反应液继续搅拌半小时后用乙酸乙酯萃取,有机相用饱和食盐水洗涤,有机相用无水硫酸钠干燥,过滤、浓缩得150毫克白色固体粗产物(S,E)-N-(1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)亚乙基)-2-甲基丙烷-2-亚磺酰胺(PA169-7)。(ESI)m/z.[M+H]+433.20。
8.化合物(S)-N-((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)-2-甲基丙烷-2-亚磺酰胺(PA169-8)的合成
化合物(S,E)-N-(1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)亚乙基)-2-甲基丙烷-2-亚磺酰胺(PA169-7)(0.15克,346.4微摩尔)溶于5毫升四氢呋喃,冷却至-50度后加入硼氢化钠(52.4毫克,1.39毫摩尔),反应液缓慢升至室温后继续搅拌3小时。加入10毫升饱和氯化铵水溶液淬灭反应,乙酸乙酯萃取,饱和食盐水洗涤,无水硫酸钠干燥,过滤、浓缩后得120毫克黄色固体粗产物(S)-N-((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)-2-甲基丙烷-2-亚磺酰胺(PA169-8)。(ESI)m/z.[M+H]+435.1。
9.化合物(S)-N-(4-(1-氨基乙基)双环[2.2.2]辛烷-1-基)-8-氯-1,7-萘啶-2-胺(PA169-9)的合成
化合物(S)-N-((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)-2-甲基丙烷-2-亚磺酰胺(PA169-8)(0.12克,275.8微摩尔)溶于5毫升1,4-二氧六环,冷却到0度后滴加盐酸溶液(1,4-二氧六环溶液,2M浓度,1.1毫升)。反应液缓慢升至室温继续搅拌3小时,反应完全。反应液浓缩后用饱和碳酸氢钠水溶液调节pH8,乙酸乙酯萃取,有机相用饱和食盐水洗涤后用无水硫酸钠干燥,过滤、浓缩得75毫克黄色固体粗产物(S)-N-(4-(1-氨基乙基)双环[2.2.2]辛烷-1-基)-8-氯-1,7-萘啶-2-胺(PA169-9)。(ESI)m/z.[M+H]+331.0。
10.化合物2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)氨基)嘧啶-5-基)丙酸叔丁酯(PA169-10)的合成
(S)-N-(4-(1-氨基乙基)双环[2.2.2]辛烷-1-基)-8-氯-1,7-萘啶-2-胺(PA169-9(1.5克,4.53毫摩尔),2-(2-氯嘧啶-5-基)丙酸叔丁酯(1.21克,4.99毫摩尔)混合物中加入15毫升二甲基亚砜,然后加入氟化钾(1.32克,22.67毫摩尔)与TGME(744.4毫克,4.53毫摩尔),反应液加热到130℃搅拌反应16小时,反应液过滤后加入50毫升水,乙酸乙酯萃取后有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤、浓缩后硅胶柱层析纯化得白色固体化合物2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)氨基)嘧啶-5-基)丙酸叔丁酯(PA169-10)(1.15克,1.93毫摩尔,42.5%产率)。(ESI)m/z.[M+H]+537.2。
11.化合物2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)氨基)嘧啶-5-基)丙酸(PA169-11)的合成
化合物2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)氨基)嘧啶-5-基)丙酸叔丁酯(PA169-10)(1.1克,2.05毫摩尔)溶于10毫升二氯甲烷,室温搅拌下滴加5毫升三氟乙酸;继续搅拌反应2小时后浓缩反应液,得1.00克棕色固体粗产物2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)氨基)嘧啶-5-基)丙酸(PA169-11)。(ESI)m/z.[M+H]+481.20。
12.化合物2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)丙酰胺(PA169)的合成
化合物2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)氨 基)嘧啶-5-基)丙酸(PA169-11)(1克,2.08毫摩尔)溶于10毫升DMF,室温搅拌下加入二异丙基乙基胺(1.34克,10.40毫摩尔),氧杂环丁烷-3-胺(303.9毫克,4.16毫摩尔),与HATU(948.6毫克,2.49毫摩尔),反应搅拌1小时后反应完全。反应液过滤浓缩后制备HPLC纯化得白色固体化合物2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)丙酰胺(PA169)(620.0毫克,1.12毫摩尔,53.9%产率)。(ESI)m/z.[M+H]+536.1。
13.非对映异构体PA166与PA207的制备
化合物2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)双环[2.2.2]辛烷-1-基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)丙酰胺(PA169)(620.0毫克,1.16毫摩尔)经SFC手性柱拆分得两白色固体非对映异构体化合物PA166(205.0毫克,372.3微摩尔,32.2%产率)与PA207(219.2毫克,389.8微摩尔,33.7%产率),该两单体化合物酰胺邻位手性中心绝对构型暂时随机指认。
PA166:1H NMR(400MHz,DMSO-d6)δ(ppm)8.75(d,1H),8.15(s,2H),7.96(d,1H),7.87(d,1H),7.53(d,1H),7.18(s,1H),7.02(d,1H),6.74(d,1H),4.79–4.64(m,3H),4.41(t,1H),4.36(t,1H),3.98–3.89(m,1H),3.42(t,1H),2.11–2.05(m,6H),1.62–1.51(m,6H),1.30(d,3H),1.01(d,3H);(ESI)m/z.[M+H]+536.0。
PA207:1H NMR(400MHz,DMSO-d6)δ(ppm)8.76(d,1H),8.15(s,2H),7.95(d,1H),7.86(d,1H),7.53(d,1H),7.18(s,1H),7.02(d,1H),6.75(d,1H),4.79–4.64(m,3H),4.41(t,1H),4.35(t,1H),3.99–3.90(m,1H),3.41(q,1H),2.10–2.06(m,6H),1.64–1.50(m,6H),1.30(d,3H),1.00(d,3H);(ESI)m/z.[M+H]+536.0。
采用类似的方法,制备得到以下化合物:





















生物测试例
G401 ELISA测试:
实验主要仪器设备:
读板机:Envision(Perkinelmer);震板机:IKA MTS 2/4;洗板机:BioTek microplate wisher 405 select;细胞计数仪:Counterstar(Ruiyu-biotech);细胞培养箱:MCO-15AC(ThermoFisher);移液枪:BioHit Multichannel.Pipette:0.2-10μL,10-300μL 50-1200μL;离心机:Thermo Centrifuge ST 40R;纯水仪:Millipore Milli-Q Reference system;冰箱:海尔-80度、-20度和4度冰箱;细胞冻存盒:Mr.FrostyTM梯度降温盒Cat#5100-0001
实验细胞培养试剂、耗材及其来源:
McCoys 5A(Modified)培养基(ATCC);FBS(Gibco);0.25%Trypsin/EDTA(Gibco);PBS(Hyclone);Penicillin/Streptomycin(100x)(Gibco);DMSO(Sigma);384细胞培养板(Corning);384检测板(Greiner);96孔板(化合物稀释)(Haimen,Qunchao);T75细胞培养瓶(Corning)
实验操作:
将G401细胞种在384孔细胞培养板中,在37℃,5%CO2细胞培养箱内孵育过夜,之后加入不同浓度的化合物处理72小时。取出384细胞培养板,在离心机中倒扣细胞培养板以300rpm离心1分钟,离心后用PBS洗涤两次,再加入裂解液,裂解60分钟,之后再加入中和缓冲液进行混合,继续孵育30分钟。从384细胞培养板中吸取20ul(用于检测K27),10ul(用于检测H3)的细胞裂解液,转移到384 ELISA检测板中,4℃孵育过夜。第二天将384 ELISA板取出,在洗板机中加入TBST洗涤5遍,之后加入封闭液室温封闭1小时,弃去封闭液后加入一抗,室温孵育1小时。孵育完后在洗板机中加入TBST洗涤5遍,之后加入二抗,室温孵育1小时,孵育完后在洗板机中加入TBST洗涤5遍,最后加入显色剂显色,用Envision读板机进行读板。将得到的数据换算成抑制率,计算方法为:%抑制率=100-100*(化合物吸光度-阳性对照吸光度)/(阴性对照吸光度-阳性对照吸光度),之后在GraphPad Prism 5软件中以“对数(抑制剂)与反应-可变斜率(四个参数)”模型对抑制率数据进行曲线拟合。
化合物抑制率计算方法如下:
%抑制率=100-100*(化合物吸光度-阳性对照吸光度)/(阴性对照吸光度-阳性对照吸光度)
本发明化合物依据上述实验方法进行测试,部分结果请见表3,其中对测试结果用A、B、C进行分类,A代表IC50≤0.1uM;B代表0.1uM<IC50≤1uM;C代表IC50>1uM
表3:

在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (14)

  1. 一种如下式I所示的化合物,或其药学上可接受的盐或氘代产物、消旋体混合物及其光学单体产物:
    其中,A环选自下组:5-10元的桥环(包括碳环、杂环)、6-10元的芳环、5-10元杂芳环;
    B环选自下组:6-10元芳环、5-14元杂芳环;
    C环选自下组:5-10元芳环或部分饱和芳环、5-10元杂芳环或部分饱和杂芳环、5-10元饱和或部分不饱和的碳环(包括稠和环、桥环),5-10元饱和或部分不饱和的杂环(包括稠和环、桥环);
    L1和L2各自独立地为-(L)p-,且各个L各自独立地选自下组:化学键或无,或-O-、-CHR-、-CHR-NH-、羰基、S、-NR-、-NHC(O)-、-NHS(O)2-、-NHC(O)NH-、-NHC(S)NH-、-COO-、-O-S(O)2-、-CHR-NR-、-C(R)2NR-、-C(R)2-;其中,各个R各自独立地选自下组:H、取代或未取代的C1-4烷基、取代或未取代的C3-4环烷基、或取代或未取代的3~4元杂环;
    n选自下组:0、1、2、3、4或5;
    m选自下组:0、1、2、3或4;
    p选自下组:0、1或2;
    各个R1、R2和R3各自独立地选自下组:H、卤素、氰基、氨基、硝基、羟基、巯基、醛基、羧基、磺酰基、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C1-C6烷胺基、取代或未取代的C1-C6烷基酰胺基、取代或未取代的C1-C6烷基C(O)O、或取代或未取代的C1-C6烷基OC(O)、取代或未取代的酰胺基、取代或未取代的胺基(NH2)、取代或未取代的C2-C6烯基、取代或未取代的C2-C6炔基、取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环(包括饱和或部分不饱和的情况),或为-X-Z-Y-R5
    其中,X和Y各自独立地选自下组:化学键,或-O-、-C(R5)2-、-S-、-NR5-;
    Z选自下组:C(O)、NH、CH=CH、-C(R5)2-、S(O)、S(O)2
    各个R5各自独立地选自下组:H、卤素、氰基、氨基、硝基、羟基、巯基、醛基、羧基、磺酰基、取代或未取代的C1-C6烷基、取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环、取代或未取代的C6-10的芳环、取代或未取代的5-12元的杂芳环、取代或未取代的C1-C6烷基C(O)O、或取代或未取代的C1-C6烷基OC(O)、取代或未取代的5-9元的杂螺环;
    或连接于环上相邻环原子上的两个相连R1、R2或R3共同构成选自下组的并环结构:取代或未取代的C6-10的芳环、取代或未取代的5-12元的杂芳环、取代或未取代的C3-C8 碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-8元的杂环(包括饱和或部分不饱和的情况);或连接于相邻环同一个碳原子上的两个相连的R1、R2或R3与相邻环共同构成3-8元饱和或部分不饱和的螺环结构、或3-8元饱和或部分不饱和的杂螺环结构(此处指取代基形成的环为3-8元,不包括相邻的环);
    其中,上述各个杂环的环骨架可以含有1-3个选自硼、氧、硫、磷和氮中的杂原子;特别地,当所述的原子为硼、硫、磷和氮时,所述的环骨架原子可以是被氧化的,如S(O)或S(O)2
    除非特别说明,上述各式中,所述的取代指对应基团被一个或多个选自下组的取代基所取代:氘、氚、卤素、氧代、=NH、=N(C1-8烷基)、羟基、羧基、巯基、苄基、C1-C12烷氧基羰基、C1-C6醛基、氨基、C1-C6酰胺基、硝基、氰基、未取代或卤代的C1-C6烷基、C1-C6烷基-CN、未取代或卤代的C3-C8环烷基、C2-C10烯基、C2-C10炔基、C1-C6烷氧基、C1-C6烷基-胺基、C6-C10芳基、五元或六元杂芳基、3-8元非芳香性杂环基、-O-(C6-C10芳基)、-O-(五元或六元杂芳基)、C1-C12烷氨基羰基、未取代或卤代的C2-C10酰基、磺酰基(-SO2-OH)、磷酰基(-PO3-OH)、未取代或卤代的C1-C4烷基-S(O)2-、未取代或卤代C1-C4烷基-SO-、未取代或卤代C1-C4烷基氨基-S(O)2-;
    其中,分子中各个手性原子可以为R构型,S构型,或其组合。
  2. 如权利要求1所述的化合物,或其药学上可接受的盐或氘代产物,其特征在于,B环为6-10元的杂芳环,且所述的R1选自下组:H、卤素、氰基、氨基、硝基、羟基、巯基、醛基、羧基、磺酰基、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C1-C6烷胺基;或两个连接于相邻环原子上的R1共同构成选自下组的环状结构:取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环(包括饱和与部分不饱和的情况)。
  3. 如权利要求1所述的化合物,或其药学上可接受的盐或氘代产物,其特征在于,所述的B环具有选自下组的结构(其中,连接位点可以位于任何环原子上):
  4. 如权利要求1所述的化合物,或其药学上可接受的盐或氘代产物,其特征在于,所述的A环具有选自下组的结构(其中,连接位点可以位于任何环原子上):
  5. 如权利要求1所述的化合物,或其药学上可接受的盐或氘代产物,其特征在于,所述的R2具有如下式所示的结构:
    其中,L3选自下组:-C(R5)2-C(O)-、-C(R5)2-;
    R3选自下组:H、取代或未取代的C1-C6烷基;
    R6选自下组:取代或未取代的C1-C6烷基、取代或未取代的C3-C6碳环(包括饱和 或部分不饱和的情况)、取代或未取代的3-6元的杂环;所述杂环的环骨架可以含有1-3个选自氧和硫中的杂原子;且所述的环骨架原子可以是被氧化的;
    所述的取代指对应基团上的氢原子被一个或多个选自下组的取代基所取代:氘、氚、卤素、氧代、羟基、羧基、巯基、苄基、氰基、未取代或卤代的C1-C6烷基、未取代或卤代的C3-C8环烷基、C2-C10烯基、C1-C6烷氧基、C1-C6烷基-胺基、C6-C10芳基、五元或六元杂芳基、3-8元非芳香性杂环基。
  6. 如权利要求1所述的化合物,或其药学上可接受的盐或氘代产物,其特征在于,所述的R2为-CHR-C(O)NH-R6,其中R为H或取代或未取代的C1-4烷基、取代或未取代C1-C6烷氧基、羟基、取代或未取代氨基;R6选自下组:取代或未取代的C3-C6碳环(包括饱和或部分不饱和的情况)、取代或未取代的3-6元的杂环(包括饱和或部分不饱和的情况)。
  7. 如权利要求1所述的化合物,或其药学上可接受的盐或氘代产物,其特征在于,所述的化合物具有如下式II、III、IV所示的结构:
    其中,X,Y各自独立地选自下组:O、NR3、C(R3)2;-C(=O)-
    或所述的化合物具有如下式III所示的结构:
    其中,X1、X2、X3或X4各自独立地选自下组:N、CR3
    或所述的化合物具有如下式IV所示的结构:
    其中,X1、X2、X3或X4各自独立地选自下组:O、S、N、NR3或CR3
    为单键或双键;
    其余各基团的定义如权利要求1中所述。
  8. 如权利要求1所述的化合物,或其药学上可接受的盐或氘代产物,其特征在于,所述的L1选自下组:化学键、-O-、-CHR-、羰基、S、-NH-;所述的L2选自下组:化学键、-CHR-NH-、-CHR-O-、-CHR-S-、-(CHR)2-。
  9. 如权利要求1所述的化合物,或其药学上可接受的盐或氘代产物,其特征在于,所述的化合物选自下组:









  10. 一种药物组合物,其特征在于,所述的药物组合物含有治疗有效量的如权利要求1所述的化合物、其可药用的盐、外消旋体、光学异构体、立体异构体或互变异构体中的一种或多种,以及一种或多种可药用的载体、赋形剂、佐剂、辅料和/或稀释剂。
  11. 如权利要求1所述的化合物、其外消旋体、光学异构体单体及其混合物或可 药用盐在制备治疗或预防与PRC2复合物、单体、或其组合相关突变、过高表达、或H3K27甲基化水平失调相关的疾病的药物中的用途。
  12. 如权利要求11所述的用途,其特征在于,所述的疾病选自下组:肿瘤、自身免疫性疾病。
  13. 如权利要求11所述的用途,其特征在于,所述的疾病选自下组:淋巴瘤,恶性血液病,肉瘤,前列腺癌,乳腺癌,肾癌,尿路上皮癌,胃癌,卵巢癌,子宫内膜癌,宫颈癌,肺癌,肝癌,胰腺癌,结肠癌,头颈癌,脑瘤,黑色素瘤,间皮瘤、胃肠间质瘤、银屑病和红斑狼疮。
  14. 如权利要求11所述的用途,其特征在于,所述的疾病选自下组:非霍奇金性淋巴瘤、滤泡性淋巴瘤、套细胞淋巴瘤、外周T细胞淋巴瘤、伯基特淋巴瘤、霍奇金淋巴瘤、慢性淋巴细胞白血病、急性和慢性髓系白血病、急性和慢性淋巴细胞白血病、多发性骨髓瘤、骨髓增生异常综合征、上皮样肉瘤、横纹肌肉瘤、脂肪肉瘤,前列腺癌,乳腺癌,肾癌,膀胱癌、上尿路上皮癌,胃癌,卵巢癌,子宫内膜癌,宫颈癌,肺癌,肝癌,胰腺癌,结肠癌,头颈癌,随母细胞瘤、胶质细胞瘤、神经鞘瘤,黑色素瘤,间皮瘤、胃肠间质瘤、银屑病和红斑狼疮。
PCT/CN2023/106840 2022-07-12 2023-07-11 一类甲基转移酶抑制剂及其用途 WO2024012461A1 (zh)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202210822809.7 2022-07-12
CN202210822809 2022-07-12
CN202211729476.X 2022-12-30
CN202211729476.XA CN117384153A (zh) 2022-07-12 2022-12-30 一类甲基转移酶抑制剂及其用途

Publications (1)

Publication Number Publication Date
WO2024012461A1 true WO2024012461A1 (zh) 2024-01-18

Family

ID=89465459

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/106840 WO2024012461A1 (zh) 2022-07-12 2023-07-11 一类甲基转移酶抑制剂及其用途

Country Status (2)

Country Link
CN (1) CN117384153A (zh)
WO (1) WO2024012461A1 (zh)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017025493A1 (en) * 2015-08-12 2017-02-16 Bayer Pharma Aktiengesellschaft Quinoline ezh2 inhibitors
CN107108637A (zh) * 2014-12-23 2017-08-29 诺华股份有限公司 三唑并嘧啶化合物及其用途
WO2021057853A1 (en) * 2019-09-26 2021-04-01 Novartis Ag Aza-quinoline compounds and uses thereof
CN114555589A (zh) * 2020-08-10 2022-05-27 诺华公司 用于抑制ezh2的化合物和组合物

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107108637A (zh) * 2014-12-23 2017-08-29 诺华股份有限公司 三唑并嘧啶化合物及其用途
WO2017025493A1 (en) * 2015-08-12 2017-02-16 Bayer Pharma Aktiengesellschaft Quinoline ezh2 inhibitors
WO2021057853A1 (en) * 2019-09-26 2021-04-01 Novartis Ag Aza-quinoline compounds and uses thereof
CN114555589A (zh) * 2020-08-10 2022-05-27 诺华公司 用于抑制ezh2的化合物和组合物

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HE, HUALONG ET AL.: "Design and synthesis of (E)-1,2-diphenylethene-based EZH2 inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 30, no. 5, 7 January 2020 (2020-01-07), XP085993676, DOI: 10.1016/j.bmcl.2020.126957 *

Also Published As

Publication number Publication date
CN117384153A (zh) 2024-01-12

Similar Documents

Publication Publication Date Title
CN113365988B (zh) Shp2抑制剂及其应用
JP6916795B2 (ja) Lsd1阻害剤
CN108395452B (zh) 作为钠通道调节剂的吡啶酮酰胺的前药
WO2021143693A1 (zh) 芳基或杂芳基并吡啶酮或嘧啶酮类衍生物及其制备方法和应用
CN105601573B (zh) 2-氨基嘧啶类化合物及其药物组合物和应用
WO2016131381A1 (en) Fused-ring compounds, pharmaceutical composition and uses thereof
CA2840627A1 (en) Quinazolines as therapeutic compounds and related methods of use
CA3119526A1 (en) Small molecule degraders of helios and methods of use
EP2964223A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
CN111051300B (zh) 作为组蛋白脱乙酰基酶1和/或2(hdac1-2)的选择性抑制剂的新杂芳基酰胺衍生物
JP2018502841A (ja) Hdac1/2阻害剤としてのピペリジン誘導体
US20210269440A1 (en) Immunomodulators, compositions and methods thereof
KR20150008891A (ko) 알파 7 니코틴성 아세틸콜린 수용기 알로스테릭 조절제, 그의 유도체 및 그의 용도
JP7016471B2 (ja) ムスカリン性アセチルコリンレセプターm4のポジティブアロステリック調節因子
CN108530444B (zh) 一种新型nampt和ido双重抑制剂及其制备方法和医药用途
WO2021249563A1 (zh) 芳基或杂芳基并吡啶酮或嘧啶酮类衍生物及其制备方法和应用
JP6859358B2 (ja) テトラヒドロインダゾール及びその医学的使用
CN113166100A (zh) 免疫调节性化合物
CA3079188A1 (en) Antagonists of the muscarinic acetylcholine receptor m4
EP2760830A1 (en) Pyrrolinone carboxamide compounds useful as endothelial lipase inhibitors
CN110914253B (zh) 一类异吲哚酮-酰亚胺环-1,3-二酮-2-烯化合物、其组合物和用途
CN112513041B (zh) 三环化合物
JP6985271B2 (ja) アルキニルジヒドロキノリンスルホンアミド化合物
WO2016140501A1 (en) Pyridine n-oxide for enhancer of zeste homolog 2 inhibitors
JP2019059697A (ja) 置換された多環性ピリダジン誘導体およびそのプロドラッグ

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: 23838952

Country of ref document: EP

Kind code of ref document: A1