WO2021175270A1 - 新型hpk1抑制剂及其制备方法和应用 - Google Patents

新型hpk1抑制剂及其制备方法和应用 Download PDF

Info

Publication number
WO2021175270A1
WO2021175270A1 PCT/CN2021/078965 CN2021078965W WO2021175270A1 WO 2021175270 A1 WO2021175270 A1 WO 2021175270A1 CN 2021078965 W CN2021078965 W CN 2021078965W WO 2021175270 A1 WO2021175270 A1 WO 2021175270A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
substituted
methyl
cancer
unsubstituted
Prior art date
Application number
PCT/CN2021/078965
Other languages
English (en)
French (fr)
Inventor
江磊
冯志勇
金贤
刘胜洋
聂士常
石倩
寿建勇
汪兵
徐圆
张建华
张毅
赵海霞
Original Assignee
轶诺(浙江)药业有限公司
上海轶诺药业有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 轶诺(浙江)药业有限公司, 上海轶诺药业有限公司 filed Critical 轶诺(浙江)药业有限公司
Priority to CN202180019207.1A priority Critical patent/CN115279760A/zh
Publication of WO2021175270A1 publication Critical patent/WO2021175270A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • 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
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines 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
    • A61K31/519Pyrimidines; 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • 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
    • A61K31/5355Non-condensed oxazines 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
    • A61K31/535Heterocyclic 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
    • A61K31/537Heterocyclic 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 spiro-condensed or forming part of bridged 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
    • A61K31/535Heterocyclic 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
    • A61K31/535Heterocyclic 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
    • A61K31/53831,4-Oxazines, e.g. morpholine 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
    • A61K31/54Heterocyclic 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
    • 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
    • A61K31/547Heterocyclic 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 spiro-condensed or forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • 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 small molecule drugs. Specifically, the present invention relates to a kinase inhibitor and its preparation and use.
  • T cells, B cells and dendritic cells are powerful weapons used by the human immune system to resist foreign invaders such as viruses and bacteria, as well as self-infected cells or abnormal cells such as cancerous cells.
  • the human body has a complex and sophisticated control system to ensure the normal operation of the immune system.
  • cancerous cells appear in the human body, if the immune system cannot completely kill them, these immune escaped cancer cells will proliferate abnormally and form tumors.
  • Traditional tumor treatment is mainly through surgery, radiotherapy, chemotherapy and molecular targeted drugs. However, for many types of tumors or cancers, surgical resection is often not a viable option. While radiotherapy and chemotherapy target tumor cells, they also damage some healthy cells.
  • HPK1 Hematopoietic progenitor cell kinase 1
  • MAP4K1 a member of the MAP4K family
  • DCs dendritic cells
  • HPK1 is mainly expressed by hematopoietic cells, including early hematopoietic progenitor cells. In T cells, HPK1 is believed to be able to degrade these proteins by phosphorylating the Ser376 site of the downstream SLP76 protein and the Thr254 site of the Gads protein and recruiting 14-3-3 protein, thereby reducing the persistence of the signaling microclusters. To negatively regulate the role of T cell activation. HPK1 can also be activated in response to prostaglandins (PGE2) normally secreted by tumors, helping tumor cells escape from the immune system.
  • PGE2 prostaglandins
  • HPK1 can also inhibit AP-1, and AP-1 plays a role in promoting cell proliferation, inhibiting differentiation, and promoting tumor cell invasion and metastasis during tumor formation and development.
  • Targeted destruction of HPK1 kinase alleles can enable T cells to increase the production of Th1 cytokines (IL-2, IFN ⁇ , etc.) in the TCR response.
  • HPK1 has multiple roles in immunity and is related to the pathogenesis of autoimmune diseases, cancer and inflammation.
  • HPK1 kinase knockout (HPK1-/-) T cells proliferate much faster than monomeric wild-type, and mice transfected with HPK1-/-T cells can resist tumor growth, and dendritic cells (DCs) that have lost HPK1 kinase ) Compared with wild type, it has better antigen presentation ability and can better show anti-tumor immune response.
  • DCs dendritic cells
  • HPK1 kinase knockout T cells proliferate much faster than monomeric wild-type, and mice transfected with HPK1-/-T cells can resist tumor growth, and dendritic cells (DCs) that have lost HPK1 kinase )
  • DCs dendritic cells
  • HPK1 kinase knockout T cells proliferate much faster than monomeric wild-type, and mice transfected with HPK1-/-T cells can resist tumor growth, and dendritic cells (DCs) that have lost HPK1 kinase )
  • the purpose of the present invention is to provide a small molecule HPK1 inhibitor with selectivity and high activity.
  • the first aspect of the present invention provides a compound represented by the following formula I, or a pharmaceutically acceptable salt, optical isomer or hydrate thereof:
  • M 1 , M 2 or M 3 are each independently CH or N; and when said M 1 or M 2 is C, said It can be located on the M 1 or M 2 ;
  • Ring A is selected from the following group: C3-C8 cycloalkyl, 5-12 membered heterocyclic group, 6-10 membered aromatic ring, or 5-10 with 1-3 heteroatoms selected from N, S and O
  • a heteroaromatic ring of a member, or A is H;
  • n 1, 2, 3 or 4;
  • n 0, 1, 2, 3, 4 or 5;
  • p 0, 1 or 2;
  • Ra is selected from the group: halogen, CN, substituted or unsubstituted C1-C6 alkyl, or Wherein, the ring B is C3-C8 cycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, 3 with 1-3 heteroatoms selected from the group consisting of N, S and O. -10 membered heterocyclic group;
  • R 7 , R 8 , R 9 , R 10 , R 11 , and R 12 is independently selected from the following group: H, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy , Substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted 3-12 membered heterocyclic group having 1-3 heteroatoms selected from the group consisting of N, S and O; or said R 9 Together with R 10 or R 11 and R 12 and the nitrogen atom to which they are connected to form a substituted or unsubstituted 3-12 membered heterocyclic group with 1-3 heteroatoms selected from N, S and O;
  • each chiral center is R configuration or S configuration.
  • the A ring is selected from the following group: in,
  • Z is selected from the following group: NH, O or S;
  • q 0, 1 or 2;
  • Re and Rf are each independently selected from the following group: H, CN, halogen, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, phenyl, substituted or unsubstituted C3- C8 cycloalkyl, substituted or unsubstituted 3-12 membered heterocyclic group with 1-3 heteroatoms selected from the group consisting of N, S and O; or said Re and Rf and the atoms to which they are connected together form C3 -C8 cycloalkyl, or substituted or unsubstituted 3-12 membered heterocyclic group with 1-3 heteroatoms selected from N, S and O;
  • the Or R 3 can be located on M 1 or M 2 described above.
  • the A ring is selected from the following group: C3-C8 cycloalkyl, benzene ring, 5-6 membered heterocyclic ring, or heterocyclic ring having 1-3 selected from N, S and O A 5-10 membered heteroaromatic ring of atoms; and said R 2 is selected from the group consisting of H, halogen, CN, substituted or unsubstituted C1-C6 alkyl.
  • the M 1 is N, and the M 2 or M 3 is CH.
  • the X is O.
  • the Y is NH
  • the Y is -NH-C(O)NHCH 2 -.
  • the Y is -NH-C(S)NHCH 2 -.
  • the compound of formula I has the structure shown in the following formula II-1 or II-2:
  • Said Ra is independently selected from the following group: halogen, CN, substituted or unsubstituted C1-C6 alkyl;
  • the compound of formula I has the structure shown in the following formula III:
  • the compound of formula I is selected from the following group:
  • a pharmaceutical composition comprising (1) the compound according to claim 1 or its stereoisomer or tautomer, or its pharmaceutically Acceptable salts, hydrates or solvates; (2) pharmaceutically acceptable carriers.
  • the third aspect of the present invention provides the compound as described in the first aspect of the present invention or its stereoisomer or tautomer, or its pharmaceutically acceptable salt, hydrate or solvate, or as described in the present invention
  • the use of the pharmaceutical composition according to the second aspect of the invention is to prepare a pharmaceutical composition for preventing and/or treating diseases related to the activity or expression of HPK1 kinase.
  • the fourth aspect of the present invention provides a compound according to the first aspect of the present invention and its pharmaceutically acceptable salts, stereoisomers, esters, prodrugs, solvates and deuterated compounds and other tumor immunotherapy
  • the other tumor immunotherapeutic agents are selected from the following group: small molecule compounds and antibodies (including but not limited to PD-1, PD-L1, CTLA-4, TIM-3, TGF- ⁇ and Its receptors, LAG3 antagonists or TLR4, TLR7, TLR8, TLR9, STING agonists, etc.), chemotherapy regimens, radiotherapy regimens, tumor-targeted drugs, tumor vaccines, etc.
  • the diseases include but are not limited to cancer, metastatic cancer, inflammation and autoimmune related diseases.
  • the disease includes but is not limited to: lymphoma, blastoma, medulloblastoma, retinoblastoma, sarcoma, liposarcoma, synovial cell sarcoma, neuroendocrine tumor, carcinoid tumor, Gastrinoma, islet cell carcinoma, mesothelioma, schwannoma, acoustic neuroma, meningioma, adenocarcinoma, melanoma, leukemia or lymphoid malignancies, squamous cell carcinoma, epithelial squamous cell carcinoma, lung cancer, small Cell lung cancer, non-small cell lung cancer, adenocarcinoma lung cancer, lung squamous cell carcinoma, peritoneal cancer, hepatocellular carcinoma, gastric cancer, bowel cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, breast cancer Cancer, metastatic breast
  • the term “about” means that the value can vary from the recited value by no more than 5%.
  • the expression “about 100” includes all values between 95 and 105 (eg, 95.1, 95.2, 95.3, 95.4, etc.).
  • the term "containing” or “including (including)” can be open, semi-closed, and closed. In other words, the term also includes “substantially consisting of” or “consisting of”.
  • alkyl includes straight or branched chain alkyl groups.
  • C 1 -C 8 alkyl means a straight or branched alkyl group having 1-8 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl Wait.
  • alkenyl includes linear or branched alkenyl.
  • C 2 -C 6 alkenyl refers to a linear or branched alkenyl having 2-6 carbon atoms, such as vinyl, allyl, 1-propenyl, isopropenyl, 1-butenyl, 2 -Butenyl, or similar groups.
  • alkynyl includes straight-chain or branched alkynyl groups.
  • C 2 -C 6 alkynyl refers to a linear or branched alkynyl group having 2-6 carbon atoms, such as ethynyl, propynyl, butynyl, or the like.
  • C 3 -C 8 cycloalkyl refers to a cycloalkyl group having 3-8 carbon atoms. It may be a monocyclic ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or similar groups. It may also be in the form of a bicyclic ring, such as a bridged ring or a spiro ring.
  • C 1 -C 8 alkoxy refers to a linear or branched alkoxy group having 1-8 carbon atoms; for example, methoxy, ethoxy, propoxy, iso Propoxy, butoxy, isobutoxy, tert-butoxy, etc.
  • the term "3-12 membered heterocyclic group having 1-3 heteroatoms selected from the group consisting of N, S, and O" refers to a group having 3-12 atoms and 1-3 atoms are selected Saturated or partially saturated cyclic groups of heteroatoms from the following group of N, S, and O. It can be a single ring or a double ring form, such as a bridged ring or a spiro ring. Specific examples may be oxetane, azetidine, tetrahydro-2H-pyranyl, piperidinyl, tetrahydrofuranyl, morpholinyl, pyrrolidinyl and the like.
  • C 6 -C 10 aryl group refers to an aryl group having 6-10 carbon atoms, for example, a phenyl group or a naphthyl group and the like.
  • N, S, and O refers to those having 5-10 atoms and wherein 1-3 atoms are selected from The following group of N, S, and O heteroatoms are cyclic aromatic groups. It may be a monocyclic ring or a condensed ring form.
  • pyridyl pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3)-triazolyl and (1,2, 4)-Triazolyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, etc.
  • the groups of the present invention can be substituted by substituents selected from the group consisting of halogen, nitrile, nitro, hydroxyl, amino, C 1 -C 6 alkyl-amino, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxy, halo C 1 -C 6 alkyl, halo C 2 -C 6 alkenyl, halogen Substituted C 2 -C 6 alkynyl, halogenated C 1 -C 6 alkoxy, allyl, benzyl, C 6 -C 12 aryl, C 1 -C 6 alkoxy-C 1 -C 6 alkane Group, C 1 -C 6 alkoxy-carbonyl, phenoxycarbonyl, C 2 -C 6 alkynyl-carbonyl, C 2 -C 6 alkenyl-carbonyl, C
  • halogen or halogen atom refers to F, Cl, Br, and I. More preferably, the halogen or halogen atom is selected from F, Cl and Br. "Halogenated” refers to substitution with an atom selected from F, Cl, Br, and I.
  • the structural formula described in the present invention is intended to include all isomeric forms (such as enantiomers, diastereomers and geometric isomers (or configuration isomers)): The R and S configurations of the center of symmetry, the (Z) and (E) isomers of the double bond, etc. Therefore, a single stereochemical isomer of the compound of the present invention or a mixture of its enantiomers, diastereomers or geometric isomers (or configurational isomers) belongs to the scope of the present invention.
  • tautomers means that structural isomers with different energies can exceed the low energy barrier to convert into each other.
  • proton tautomers ie, proton transfer
  • Valence tautomers include interconversion through the recombination of some bonding electrons.
  • solvate refers to a complex in which the compound of the present invention coordinates with solvent molecules to form a specific ratio.
  • the present invention provides a compound represented by the following formula I:
  • M 1 , M 2 or M 3 are each independently CH or N; and when said M 1 or M 2 is C, said It can be located on the M 1 or M 2 ;
  • Ring A is selected from the following group: C3-C8 cycloalkyl, 5-12 membered heterocyclic group, 6-10 membered aromatic ring, or 5-10 with 1-3 heteroatoms selected from N, S and O
  • a heteroaromatic ring of a member, or A is H;
  • n 1, 2, 3 or 4;
  • n 0, 1, 2, 3, 4 or 5;
  • p 0, 1 or 2;
  • Ra is selected from the group: halogen, CN, substituted or unsubstituted C1-C6 alkyl, or Wherein, the ring B is C3-C8 cycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, 3 with 1-3 heteroatoms selected from the group consisting of N, S and O. -10 membered heterocyclic group;
  • R 7 , R 8 , R 9 , R 10 , R 11 , and R 12 is independently selected from the following group: H, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy , Substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted 3-12 membered heterocyclic group having 1-3 heteroatoms selected from the group consisting of N, S and O; or said R 9 Together with R 10 or R 11 and R 12 and the nitrogen atom to which they are connected to form a substituted or unsubstituted 3-12 membered heterocyclic group with 1-3 heteroatoms selected from N, S and O;
  • each chiral center is R configuration or S configuration.
  • R 8 , R 9 , R 10 , R 11 , and R 12 are each independently a corresponding group in a specific compound in each embodiment.
  • the compounds of the present invention can be used as HPK1 kinase inhibitors, in a preferred embodiment, they are HPK1 kinase selective inhibitors.
  • the compound of formula I of the present invention can be prepared by the following exemplary method:
  • the compound Ia is used for the cyclization reaction to obtain the compound of formula I'.
  • the compound of the present invention has excellent HPK1 kinase inhibitory activity, the compound of the present invention and its various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates, and containing the compound of the present invention as the main active ingredient
  • the pharmaceutical composition can be used to prevent and/or treat diseases related to the activity or expression of HPK1 kinase (e.g., cancer).
  • the pharmaceutical composition of the present invention contains a safe and effective amount of the compound of the present invention and a pharmaceutically acceptable excipient or carrier.
  • the "safe and effective amount” refers to: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects.
  • the pharmaceutical composition contains 1-2000 mg of the compound of the present invention/agent, more preferably, 10-200 mg of the compound of the present invention/agent.
  • the "one dose” is a capsule or tablet.
  • “Pharmaceutically acceptable carrier” refers to: one or more compatible solid or liquid fillers or gel substances, which are suitable for human use, and must have sufficient purity and sufficiently low toxicity. "Compatibility” here means that the components in the composition can be blended with the compound of the present invention and between them without significantly reducing the efficacy of the compound.
  • pharmaceutically acceptable carriers include cellulose and its derivatives (such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, talc, and solid lubricants (such as stearic acid).
  • Magnesium stearate calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as Tween) ), wetting agents (such as sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • vegetable oils such as soybean oil, sesame oil, peanut oil, olive oil, etc.
  • polyols such as propylene glycol, glycerin, mannitol, sorbitol, etc.
  • emulsifiers such as Tween
  • wetting agents such as sodium lauryl sulfate
  • coloring agents such as sodium lauryl sulfate
  • flavoring agents such as pepperminophen, sorbitol, etc.
  • antioxidants
  • the method of administration of the compound or pharmaceutical composition of the present invention is not particularly limited, and representative administration methods include (but are not limited to): oral, parenteral (intravenous, intramuscular, or subcutaneous) injection, and the like.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or mixed with the following ingredients: (a) fillers or compatibilizers, for example, Starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders such as hydroxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and gum arabic; (c) humectants, For example, glycerin; (d) disintegrants, such as agar, calcium carbonate, potato starch or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) Absorption accelerators, such as quaternary amine compounds; (g) wetting agents, such as cetyl alcohol and glycty
  • Solid dosage forms such as tablets, sugar pills, capsules, pills and granules can be prepared with coatings and shell materials, such as enteric coatings and other materials known in the art. They may contain opacifying agents, and the active compound or the release of the compound in such a composition may be released in a certain part of the digestive tract in a delayed manner. Examples of embedding components that can be used are polymeric substances and waxes. If necessary, the active compound can also be formed into microcapsules with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • the liquid dosage form may contain inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1 , 3-Butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances.
  • composition may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfumes.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents and perfumes.
  • the suspension may contain suspending agents, for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • suspending agents for example, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
  • composition for parenteral injection may contain physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
  • the compounds of the present invention can be administered alone or in combination with other pharmaceutically acceptable compounds.
  • the pharmaceutical composition When administered in combination, the pharmaceutical composition also includes one or more other pharmaceutically acceptable compounds.
  • One or more of the other pharmaceutically acceptable compounds may be administered simultaneously, separately or sequentially with the compound of the present invention.
  • 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.
  • the administered dose is usually 1 to 2000 mg, preferably 20 to 500 mg.
  • the specific dosage should also consider factors such as the route of administration and the patient's health status, which are all within the skill range of a skilled physician.
  • reaction solution was poured into 60 ml of water, and the mixture was extracted with ethyl acetate (20 ml ⁇ 3).
  • the combined organic phase was washed with water (20 mL ⁇ 3), saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product.
  • reaction solution was slowly added to water (40 mL), and extracted with ethyl acetate (20 mL ⁇ 3).
  • the combined organic phase was washed successively with water (20 ml ⁇ 3) and saturated aqueous salt solution (20 ml), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product.
  • reaction solution was poured into water (20 mL), and ethyl acetate (20 mL ⁇ 3) was added for extraction.
  • the combined organic phase was washed with water (10 ml ⁇ 3), saturated brine (10 ml), dried with anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product.
  • 2,6-Difluorophenol (1.7 g, 13 mmol) was dissolved in acetic acid (20 mL), and concentrated nitric acid (1.9 g, 20 mmol) was added dropwise.
  • the reaction solution was reacted at 70 degrees for 2 hours. TLC detects that the reaction is complete.
  • the reaction solution was poured into ice water (100 mL), and extracted with ethyl acetate (100 mL ⁇ 2). The combined organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product.
  • reaction solution was heated to 100 degrees and stirred for 2 hours. The reaction was checked by LCMS. After the reaction solution was cooled to room temperature, ethyl acetate (10 mL ⁇ 2) was added for extraction. The combined organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product.
  • Example 2 2-((3,5-Difluoro-4-((5-(2-fluoro-4-methoxyphenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl )Oxo)phenyl)ammonia Yl)-5-fluoro-5,6-dihydro-4H-1,3-oxazin-5-yl)methanol
  • reaction solution was cooled to room temperature, and then (1-(aminomethyl)cyclopropyl)methanol (59 mg, 0.58 mmol) was added.
  • the reaction was stirred at room temperature for 3 hours. The reaction was checked by LCMS.
  • reaction solution was cooled to room temperature, and then (1-(aminomethyl)cyclopropyl)methanol (111 mg, 1.09 mmol) was added.
  • the reaction was stirred at room temperature for 1 hour.
  • the reaction was checked by LCMS.
  • reaction solution was heated to 100 degrees and stirred for 3 hours. The reaction was checked by LCMS. After the reaction solution was cooled to room temperature, ethyl acetate (5 mL ⁇ 2) was added for extraction. The combined organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product.
  • reaction solution was cooled to room temperature, and then (1-(aminomethyl)cyclopropyl)methanol (56 mg, 0.56 mmol) was added.
  • the reaction was stirred at room temperature for 3 hours. The reaction was checked by LCMS.
  • Example 14 (2-((3,5-Difluoro-4-((5-isopropyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)phenyl)amino )-5-Fluoro- 5,6-Dihydro-4H-1,3-oxazin-5-yl)methanol
  • reaction solution was heated to 70 degrees and stirred for 14 hours. The reaction was checked by LCMS. After the reaction solution was cooled to room temperature, ethyl acetate (10 mL ⁇ 2) was added for extraction. The combined organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product.
  • Example 17 (2-((3,5-Difluoro-4-((5-(4-fluoro-3-methoxyphenyl)-7H-pyrrolo[2,3-d]pyrimidine-4 - (Yl)oxy)phenyl)amino)-5-methyl-5,6-dihydro-4H-1,3-oxazin-5-yl)methanol
  • reaction solution was heated to 70 degrees and stirred for 14 hours. The reaction was checked by LCMS. After the reaction solution was cooled to room temperature, ethyl acetate (5 mL ⁇ 2) was added for extraction. The combined organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product.
  • Example 27 (2-((3,5-Difluoro-4-(5-(5-isopropyl-1,3,4-oxadiazol-2-yl)-7H-pyrrolo[2, 3-d]pyrimidine -4-yl)oxy)phenyl)amino)-5-methyl-5,6-dihydro-4H-1,3-oxazin-5-yl)methanol
  • reaction solution was heated to 90 degrees and stirred for 14 hours. The reaction was checked by LCMS. After the reaction solution was cooled to room temperature, ethyl acetate (15 mL ⁇ 3) was added for extraction. The combined organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product.
  • reaction solution was stirred at zero for 20 minutes, and then diphenyl(methyl)sulfonium tetrafluoroborate (600mg, 1.02mmol) in anhydrous tetrahydrofuran (5.0ml) was added dropwise to the reaction solution. ) Solution.
  • the reaction was stirred at zero for 0.5 hours, the ice-salt bath was removed, and the reaction was placed at room temperature to continue stirring for 1 hour. The reaction was checked by LCMS. The reaction was quenched by adding saturated ammonium chloride solution (20 mL), and then extracted with ethyl acetate (15 mL ⁇ 3).
  • Example 32 (4-(4-(4-((5-oxa-7-azaspiro[2.5]oct-6-en-6-yl)amino)-2,6-difluorophenoxy )-7H-pyrrole And [2,3-d]pyrimidin-5-yl)phenyl)(2-oxa-6-azaspiro[3.3]heptan-6-yl)methanone
  • reaction solution was cooled to room temperature, and then (1-(aminomethyl)cyclopropyl)methanol (313 mg, 3.09 mmol) was added.
  • the reaction was stirred at room temperature for 1 hour.
  • the reaction was checked by LCMS.
  • reaction solution was placed in an ice-water bath to cool for 10 minutes, and then 3-(((ethylimino)methylene)amino)-N,N-dimethylpropane was added to the reaction solution -1-amine hydrochloric acid (533 mg, 2.78 mmol). The ice bath was removed and the reaction was stirred at room temperature for 24 hours. LCMS detects the reaction.
  • reaction solution was heated to 90 degrees and stirred for 3 hours. The reaction was checked by LCMS. After the reaction solution was cooled to room temperature, ethyl acetate (5 mL ⁇ 2) was added for extraction. The combined organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product.
  • Example 37 4-(2,6-Difluoro-4-((5-fluoro-5-(hydroxymethyl)-5,6-dihydro-4H-1,3-oxazin-2-yl) Amino)phenoxy Yl)-3-(4-fluorophenyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile
  • triphosgene (21 mg, 0.07 mmol) was added to the reaction solution, and the reaction solution was stirred at 0°C for 20 minutes.
  • (3-fluorooxetane-3-yl)methylamine (21 mg, 0.30 mmol) was added to the reaction solution, the ice bath was removed, and the reaction solution was stirred at room temperature for 30 minutes.
  • LCMS detects the reaction. Oily substance after concentration under reduced pressure.
  • Example 43 4-(2-Fluoro-4-((5-fluoro-5-(hydroxymethyl)-5,6-dihydro-4H-1,3-oxazin-2-yl)amino)benzene Oxy)-3-(trifluoro (Methyl)-1H-pyrrolo[2,3-b]pyridine-5-carbonitrile
  • Example 44 (2-((4-((5-chloro-3-(4-fluorophenyl)-1H-pyrrolo[2,3-b]pyridin-4-yl)oxy)-3- Fluorophenyl) ammonia Yl)-5-fluoro-5,6-dihydro-4H-1,3-oxazin-5-yl)methanol
  • reaction solution was heated to 70 degrees and stirred for 3 hours. The reaction was checked by LCMS. After the reaction solution was cooled to room temperature, ethyl acetate (5 mL ⁇ 3) was added for extraction. The combined organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product.
  • reaction solution was heated to 70 degrees and stirred for 12 hours.
  • the reaction was checked by LCMS.
  • saturated aqueous ammonium chloride solution was added to quench the reaction (2.0 mL), and dichloromethane (5 mL ⁇ 3) was added for extraction.
  • the combined organic phase was dried over anhydrous sodium sulfate and filtered.
  • the filtrate was concentrated under reduced pressure to obtain a crude product.
  • MS(ESI): m/z 577.8[M+H] + .
  • Example 62 4-(5-(3,5-Dimethyl-4-(4-methylpiperazin-1-yl)phenyl)-1H-pyrazolo[3,4-b]pyridine- 3-base) -N,N- Dimethylbenzamide
  • reaction solution was heated to 90 degrees and stirred for 3 hours. The reaction was checked by LCMS. After the reaction solution was cooled to room temperature, ethyl acetate (5 mL ⁇ 3) was added for extraction. The combined organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to obtain a crude product.
  • the enzyme activity test buffer contains 40mM Tris, pH 7.5; 20mM mg Cl2; 0.1 mg/ml BSA; 50 ⁇ M DTT.
  • the compound is dissolved in 100% DMSO, and the concentration of the mother liquor is 10 mM.
  • the DMSO solution of the compound starts from 100uM, and it is continuously diluted three-fold, with a total of eleven concentrations. After the diluted compound is diluted 1:20 with enzyme activity test buffer, add 1uL to the working well, each with two concentrations. Multiple holes. Add 1uL of DMSO solution diluted 1:20 to the negative control wells and the positive control wells.
  • the preparation of 2.5 ⁇ substrate/ATP working solution is an enzyme activity test buffer containing 0.25ug/uL MBP protein and 45uM ATP, and 2uL 2.5 ⁇ substrate/ATP working solution is added to each working well.
  • the enzyme activity test buffer contains 40mM Tris, pH 7.5; 20mM mg Cl2; 0.1 mg/ml BSA; 50 ⁇ M DTT.
  • the compound is dissolved in 100% DMSO, and the concentration of the mother liquor is 10 mM.
  • the DMSO solution of the compound is continuously diluted in three-fold gradient from 1 mM, with a total of eleven concentrations. After the diluted compound is diluted 1:20 with enzyme activity test buffer, 1uL is added to the working well, each concentration is two Multiple holes. Add 1uL of DMSO solution diluted 1:20 to the negative control wells and the positive control wells.
  • the preparation of 2.5 ⁇ substrate/ATP working solution is an enzyme activity test buffer containing 0.5ug/uL PKA substrate peptide and 105uM ATP, and 2uL 2.5 ⁇ substrate/ATP working solution is added to each working well.
  • A represents the IC 50 values ⁇ 50nM
  • B represents 50nM ⁇ IC 50 values ⁇ 500nM
  • C represents 500nM ⁇ IC 50 values ⁇ 00nMD.
  • Inhibition of HPK1 can inhibit the phosphorylation of downstream SLP76.
  • the phosphorylation of SLP76 protein uses Jurkat (ATCC, Clone E6-1 TIB-152 TM) cells were tested, the first day of the experiment cells were diluted with culture medium (RPMI 1640 + 0.5% FBS) to 106 / ml per well 100uL, 10 5 cells were plated in an amount of 96-well cell culture In the plate, starve for 4 hours and incubate. The compound is dissolved in 100% DMSO, and the concentration of the mother liquor is 4mM. The DMSO solution of the compound is continuously diluted in four times from 10 mM to a total of 9 concentrations.
  • 4 uL of the diluted compound is diluted to 196 uL of 37°C preheated RPMI 1640 and mixed.
  • Add 50uL of the final diluted compound to the cells incubate at 37°C for 20 minutes, and add 50uL of diluted human CD3/CD28T cell activator (Stemcell, catalog number: 10971) to make the final concentration volume of the activator equal to 1/ of the total system.

Abstract

本发明提供了一种HPK1激酶抑制剂及其制备和用途。具体地,本发明提供了一种如式I所示的化合物,其中,各基团的定义如说明书中所述。所述的化合物具有优异的HPK1抑制活性,因此可用于制备治疗癌症和其他HPK1活性相关疾病的药物组合物。

Description

新型HPK1抑制剂及其制备方法和应用 技术领域
本发明涉及小分子药物领域,具体地,本发明涉及一种激酶抑制剂及其制备和用途。
背景技术
T细胞、B细胞和树突状细胞(DCs)是人体免疫系统用于抵抗外来入侵者如病毒和细菌以及自身感染细胞或异常细胞如癌变细胞等的强有力武器。而人体存在一套复杂且精密的调控系统用于确保免疫系统的正常运行。当人体中出现癌变细胞的时候,如果免疫系统无法将其完全杀死,这些免疫逃逸的癌细胞异常增殖就会形成肿瘤。传统肿瘤治疗主要通过手术、放疗、化疗和分子靶向药物。然而,对于许多形式多样的肿瘤或癌症,手术切除往往并不是可行的选择。而放射疗法和化疗在靶向肿瘤细胞的同时,也会损害部分健康细胞。肿瘤细胞易于发生突变,并且可能对特异靶向肿瘤细胞的药物产生耐药性,使得癌症治疗困难重重。近年来利用患者自身的免疫系统来克服肿瘤细胞所采用的免疫逃逸策略,并增强机体抗肿瘤免疫力是一种新型的癌症治疗策略。其中一种策略是通过抑制通常起维持外周耐受作用的免疫反应的负调控因子,使肿瘤抗原被识别为非自身抗原,从而克服肿瘤细胞免疫逃逸。造血祖细胞激酶1(HPK1)又称MAP4K1(MAP4K家族成员),是一个树突状细胞(DCs)、T细胞和B细胞活化反应的负调控因子,抑制其活性可以针对性地增强机体抗肿瘤免疫力。HPK1主要由造血细胞,包括早期造血祖细胞表达。在T细胞中,HPK1被认为能够通过磷酸化下游SLP76蛋白的Ser376位点和Gads蛋白的Thr254位点并招募14-3-3蛋白来降解这些蛋白,从而降低信号微团簇的持久性,起到负调控T细胞活化的作用。HPK1还可以通过响应通常由肿瘤分泌的前列腺素(PGE2)而被激活,有助于肿瘤细胞从免疫系统逃逸。并且HPK1还能抑制AP-1,而AP-1在肿瘤形成及发展过程中,在促进细胞增殖、抑制分化、促进肿瘤细胞的侵袭和转移等过程中发挥作用。针对性地破坏HPK1激酶的等位基因可以使T细胞在TCR应答中提高Th1细胞因子(IL-2,IFNγ等)的产生。HPK1在免疫中具有多种作用,并与自身免疫性疾病,癌症和炎症反应的发病机理有关。HPK1激酶敲除(HPK1-/-)的T细胞增殖相对于单体野生型快很多,并且转染过HPK1-/-T细胞的老鼠能抵抗肿瘤的生长,失去HPK1激酶的树突细胞(DCs)与野生型相比具有更好的抗原提呈能力,能更好地表现出抗肿瘤免疫应答。此外,动物实验研究表明,HPK1的抑制和PD-1/PD-L1抗体药物具有明显的协同抗肿瘤活性。因此,HPK1激酶在疾病治疗特别是癌症治疗中具有关键作用。
目前针对该靶点尚未有药物上市,为了满足未来临床的巨大需求,我们希望通过设计开发出具有选择性和高活性的小分子HPK1抑制剂,为免疫相关疾病特别是肿瘤治疗提供新型的口服类药物,单独用药或者联合化疗、放疗、肿瘤靶向药、其他肿瘤免疫治疗剂(小分子化合物及抗体)以及肿瘤疫苗等。
发明内容
本发明的目的是提供一种具有选择性和高活性的小分子HPK1抑制剂。
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、光学异构体或水合物:
Figure PCTCN2021078965-appb-000001
其中,
X和Y各自独立地选自下组:无、NR、S、O、-NR-C(=O)R-、-NR-C(=O)NR-、、-NR-C(=O)C(=O)NR-、-NR-C(=S)NR-、-NR-C(=O)NRCH 2-、-NR-C(=S)NRCH 2-,其中,所述的R选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C3-C8环烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基;
M 1、M 2或M 3各自独立地为CH或N;且当所述的M 1或M 2为C时,所述的
Figure PCTCN2021078965-appb-000002
可以位于所述的M 1或M 2上;
A环选自下组:C3-C8环烷基、5-12元的杂环基、6-10元芳环,或具有1-3个选自N、S和O的杂原子的5-10元的杂芳环,或A为H;
m为1、2、3或4;
n为0、1、2、3、4或5;
p为0、1或2;
Ra选自下组:卤素、CN、取代或未取代的C1-C6烷基、或
Figure PCTCN2021078965-appb-000003
其中,所述的环B为C3-C8环烷基、6-10元芳基、5-10元的杂芳基、具有1-3个选自下组N、S和O的杂原子的3-10元杂环基;
各个R 1、R 2、R 3和R 4各自独立地选自下组:H、卤素、CN、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C3-C8环烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-10元杂芳基、R 7-C(=O)-、R 7-C(=O)具有1-3个选自下组N、S和O的杂原子的3-12元杂环基-、R 8-S(=O) 2-、R 9R 10N-C(=O)-、R 9R 10N-C(=O)NR-、R 11R 12N-S(=O) 2-、R 7O-、R 8-S(O) 2NR-;
各个R 7、R 8、R 9、R 10、R 11、R 12各自独立地选自下组:H、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C3-C8环烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基;或所述的R 9和R 10或R 11和R 12与其相连的氮原子共同构成取代或未取代的具有1-3个选自N、S和O的杂原子的3-12元杂环基;
除非特别说明,所述的“取代”是指被选自下组的一个或多个(例如2个、3个、4个等)取代基所取代:卤素、C1-C6烷氧基、卤代的C1-C6烷氧基、C3-C8环烷基、卤代的C3-C8环烷基、甲基砜基、氧代(=O)、-CN、羟基、-NH 2、-NHS(O) 2CH 3、C1-C6胺基、羧基、C1-C6酰胺基(-C(=O)-N(Rc) 2或-NH-C(=O)(Rc),Rc为H或C1-C5的烷基)、或取代或未取代的选自下组的基团:C1-C6烷基、C6-C10芳基、具有1-3个选自N、S和O的杂原子的5-10元杂芳基、-(CH 2)-C6-C10芳基、-(CH 2)-(具有1-3个选自N、S和O的杂原子的3-10元杂芳基)、-(具有1-3个选自N、S和O的杂原子的5-10元亚杂芳基)-(C1-C6烷基)、具有1-3个选自下组N、S和O的杂原子的3-12元杂环基(包括单环、螺环、桥环或并环),且所述的取代基选自下组:卤素、C1-C6烷基、C1-C6亚烷基-OH、C1-C6烷氧基、氧代、-S(O) 2CH 3、-CN、-OH、C6-C10芳基、具有1-3个选自N、S和O的杂原子的3-10元杂芳基、-C(O)CHNH 2、-C(O)CHOH;
且所述的式I化合物中,各个手性中心为R构型或S构型。
在另一优选例中,所述的“取代”是指被选自下组的一个或多个(例如2个、3个、4个等)取代基所取代:卤素、C1-C6烷氧基、卤代的C1-C6烷氧基、C3-C8环烷基、卤代的C3-C8环烷基、甲基砜基、氧代(=O)、-CN、羟基、-NH 2、C1-C6胺基、羧基、或取代或未取代的选自下组的基团:C1-C6烷基、C6-C10芳基、具有1-3个选自N、S和O的杂原子的5-10元杂芳基、且所述的取代基选自下组:卤素、C1-C6烷基、C1-C6亚烷基-OH、C1-C6烷氧基、氧代、-S(O) 2CH 3、-CN、-OH、C6-C10芳基、具有1-3个选自N、S和O的杂原子的3-10元杂芳基、-C(O)CHNH 2、-C(O)CHOH。
在另一优选例中,所述的A环选自下组:
Figure PCTCN2021078965-appb-000004
其中,
Z选自下组:NH、O或S;
q为0、1或2;
Re和Rf各自独立地选自下组:H、CN、卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、苯基、取代或未取代的C3-C8环烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基;或所述的Re和Rf与其相连的原子共同构成C3-C8环烷基、或取代或未取代的具有1-3个选自N、S和O的杂原子的3-12元杂环基;
且所述的
Figure PCTCN2021078965-appb-000005
还可以进一步被1或2个R 2取代。
在另一优选例中,当所述的M 1或M 2为CH时,所述的
Figure PCTCN2021078965-appb-000006
或者 R 3可以位于所述的M 1或M 2上。
在另一优选例中,所述的A环选自下组:C3-C8环烷基、苯环、5-6元的杂环、或具有1-3个选自N、S和O的杂原子的5-10元的杂芳环;且所述的R 2选自下组:H、卤素、CN、取代或未取代的C1-C6烷基。
在另一优选例中,所述的M 1为N,且所述的M 2或M 3为CH。
在另一优选例中,所述的X为O。
在另一优选例中,所述的Y为NH。
在另一优选例中,所述的Y为-NH-C(O)NHCH 2-。
在另一优选例中,所述的Y为-NH-C(S)NHCH 2-。
在另一优选例中,所述的B环选自下组:苯环、3-12元杂环基、5-6元的杂芳环、含有苯环结构单元的8-15元二环并环、含有5-6元杂芳环结构单元的8-15元二环并环;其中,所述的R 4选自下组:H、卤素、取代或未取代的C1-C6烷基、R 9R 10N-C(=O)-、R 9R 10N-C(=O)NR-、R 11R 12N-S(=O) 2-、R 7O-、R 8-S(O) 2NR-;其中,所述的取代优选包括一个或多个选自下组的基团:-NH 2、取代或未取代的C1-C6烷基。
在另一优选例中,所述的式I化合物具有如下式II-1或II-2所示的结构:
Figure PCTCN2021078965-appb-000007
其中,
所述的Ra独立地选自下组:卤素、CN、取代或未取代的C1-C6烷基;
在另一优选例中,所述的式I化合物具有如下式III所示的结构:
Figure PCTCN2021078965-appb-000008
其中,
所述的R 4选自下组:H、卤素、CN、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、R 9R 10N-C(=O)-、R 8-S(=O) 2-、R 9R 10N-C(=O)-、R 9R 10N-C(=O)NR-、R 11R 12N-S(=O) 2-、R 7O-、R 8-S(O) 2NR-;其中,所述的R 7、R 8、R 9、R 10、R 11、R 12各自独立地选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基;或所述的R 9和R 10或R 11和R 12与其相连的氮原子共同构成取代或未取代的具有1-3个选自N、S和O的杂原子的3-12元杂环基。 在另一优选例中,所述的式I化合物具有如下式III所示的结构:
Figure PCTCN2021078965-appb-000009
在另一优选例中,所述的式I化合物选自下组:
Figure PCTCN2021078965-appb-000010
Figure PCTCN2021078965-appb-000011
Figure PCTCN2021078965-appb-000012
Figure PCTCN2021078965-appb-000013
本发明的第二方面,提供了一种药物组合物,所述的药物组合物包括(1)如权利要求1所述的化合物或其立体异构体或互变异构体,或其药学上可接受的盐、水合物或溶剂化物;(2)药学上可接受的载体。
本发明的第三方面,提供了如本发明第一方面所述的化合物或其立体异构体或互变异构体,或其药学上可接受的盐、水合物或溶剂化物,或如本发明第二方面所述的药物组合物的用途,用于制备预防和/或治疗与HPK1激酶的活性或表达量相关的疾病的药物组合 物。
本发明的第四方面,提供了一种本发明第一方面所述的化合物及其药学上可接受的盐、立体异构体、酯、前药、溶剂化物和氘代化合物与其他肿瘤免疫治疗剂的联合肿瘤化疗方案,所述的其他肿瘤免疫治疗剂选自下组:小分子化合物及抗体(包括但不限于PD-1、PD-L1、CTLA-4、TIM-3、TGF-β及其受体、LAG3拮抗剂或TLR4、TLR7、TLR8、TLR9、STING激动剂等)、化疗方案、放疗方案、肿瘤靶向药、肿瘤疫苗等。
在另一优选例中,所述的疾病包括但不限于癌症、转移性癌症、炎症和自身免疫相关疾病。
在另一优选例中,所述疾病包括但不限于:淋巴瘤,母细胞瘤,髓母细胞瘤,视网膜母细胞瘤,肉瘤,脂肪肉瘤,滑膜细胞肉瘤,神经内分泌肿瘤,类癌肿瘤,胃泌素瘤,胰岛细胞癌,间皮瘤,神经鞘瘤,听神经瘤,脑膜瘤,腺癌,黑色素瘤,白血病或淋巴样恶性肿瘤,鳞状细胞癌,上皮鳞状细胞癌,肺癌,小细胞肺癌,非小细胞肺癌,腺癌肺癌,肺鳞癌,腹膜癌,肝细胞癌,胃癌,肠癌,胰腺癌,成胶质细胞瘤,子宫颈癌,卵巢癌,肝癌,膀胱癌,乳腺癌,转移性乳腺癌,结肠癌,直肠癌,结直肠癌,子宫癌,唾液腺癌,肾癌,前列腺癌,外阴癌,甲状腺癌,肛门癌,阴茎癌,梅克尔细胞癌,食管癌,胆道肿瘤,头颈部癌,血液恶性肿瘤,鼻咽癌,多发性骨髓瘤,大场绒毛腺瘤,非霍奇金淋巴瘤,骨癌,睾丸癌,霍奇金病,精元细胞瘤,口腔癌,脑癌,皮肤癌,乳腺导管癌,肾盂癌,肾母细胞瘤,食管腺瘤,视网膜细胞瘤,神经胶质瘤,神经纤维瘤,胃肠道间质瘤,原位癌,子宫内膜癌和骨髓增生异常综合征等。
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
本发明人经过长期而深入的研究,设计并合成了一类新型HPK1激酶抑制剂。在此基础上,发明人完成了本发明。
术语
除非另外定义,否则本文中所用的全部技术与科学术语均具有如本发明所属领域的普通技术人员通常理解的相同含义。
如本文所用,在提到具体列举的数值中使用时,术语“约”意指该值可以从列举的值变动不多于5%。例如,如本文所用,表述“约100”包括95和105和之间的全部值(例如,95.1、95.2、95.3、95.4等)。
如本文所用,术语“含有”或“包括(包含)”可以是开放式、半封闭式和封闭式的。换言之,所述术语也包括“基本上由…构成”、或“由…构成”。
定义
如本文所用,术语“烷基”包括直链或支链的烷基。例如C 1-C 8烷基表示具有1-8个碳原子的直链或支链的烷基,例如甲基、乙基、丙基、异丙基、丁基、异丁基、叔丁基等。
如本文所用,术语“烯基”包括直链或支链的烯基。例如C 2-C 6烯基指具有2-6个碳原子的直链或支链的烯基,例如乙烯基、烯丙基、1-丙烯基、异丙烯基、1-丁烯基、2-丁烯基、或类似基团。
如本文所用,术语“炔基”包括直链或支链的炔基。例如C 2-C 6炔基是指具有2-6个碳原子的直链或支链的炔基,例如乙炔基、丙炔基、丁炔基、或类似基团。
如本文所用,术语“C 3-C 8环烷基”指具有3-8个碳原子的环烷基。其可以是单环,例如环丙基、环丁基、环戊基、环己基、或类似基团。也可以是双环形式,例如桥环或螺环形式。
如本文所用,术语“C 1-C 8烷氧基”是指具有1-8个碳原子的直链或支链的烷氧基;例如,甲氧基、乙氧基、丙氧基、异丙氧基、丁氧基、异丁氧基、叔丁氧基等。
如本文所用,术语“具有1-3个选自下组N、S和O的杂原子的3-12元杂环基”是指具有3-12个原子的且其中1-3个原子为选自下组N、S和O的杂原子的饱和或部分饱和的环状基团。其可以是单环,也可以是双环形式,例如桥环或螺环形式。具体的实例可以为氧杂环丁烷、氮杂环丁烷、四氢-2H-吡喃基、哌啶基、四氢呋喃基、吗啉基和吡咯烷基等。
如本文所用,术语“C 6-C 10芳基”是指具有6-10个碳原子的芳基,例如,苯基或萘基等类似基团。
如本文所用,术语“具有1-3个选自下组N、S和O的杂原子的5-10元杂芳基”指具有5-10个原子的且其中1-3个原子为选自下组N、S和O的杂原子的环状芳香基团。其可以是单环,也可以是稠环形式。具体的实例可以为吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、吡咯基、吡唑基、咪唑基、(1,2,3)-三唑基以及(1,2,4)-三唑基、四唑基、呋喃基、噻吩基、异噁唑基、噻唑基、噁唑基等。
除非特别说明,否则本发明的基团均可被选自下组的取代基所取代:卤素、腈基、硝基、羟基、氨基、C 1-C 6烷基-胺基、C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷基、卤代C 2-C 6烯基、卤代C 2-C 6炔基、卤代C 1-C 6烷氧基、烯丙基、苄基、C 6-C 12芳基、C 1-C 6烷氧基-C 1-C 6烷基、C 1-C 6烷氧基-羰基、苯氧羰基、C 2-C 6炔基-羰基、C 2-C 6烯基-羰基、C 3-C 6环烷基-羰基、C 1-C 6烷基-磺酰基等。
如本文所用,“卤素”或“卤原子”指F、Cl、Br、和I。更佳地,卤素或卤原子选自F、Cl和Br。“卤代的”是指被选自F、Cl、Br、和I的原子所取代。
除非特别说明,本发明所描述的结构式意在包括所有的同分异构形式(如对映异构,非对映异构和几何异构体(或构型异构体)):例如含有不对称中心的R、S构型,双键的(Z)、(E)异构体等。因此,本发明化合物的单个立体化学异构体或其对映异构体、非对映异构体或几何异构体(或构型异构体)的混合物都属于本发明的范围。
如本文所用,术语“互变异构体”表示具有不同能量的结构同分异构体可以超过低能垒,从而互相转化。比如,质子互变异构体(即质子移变)包括通过质子迁移进行互变,如1H-吲唑与2H-吲唑。化合价互变异构体包括通过一些成键电子重组而进行互变。
如本文所用,术语“溶剂合物”是指本发明化合物与溶剂分子配位形成特定比例的配合物。
式I化合物
本发明提供了一种如下式I所示的化合物:
Figure PCTCN2021078965-appb-000014
其中,
X和Y各自独立地选自下组:无、NR、S、O、-NR-C(=O)R-、-NR-C(=O)NR-、、-NR-C(=O)C(=O)NR-、-NR-C(=S)NR-、-NR-C(=O)NRCH 2-、-NR-C(=S)NRCH 2-,其中,所述的R选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C3-C8环烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基;
M 1、M 2或M 3各自独立地为CH或N;且当所述的M 1或M 2为C时,所述的
Figure PCTCN2021078965-appb-000015
可以位于所述的M 1或M 2上;
A环选自下组:C3-C8环烷基、5-12元的杂环基、6-10元芳环,或具有1-3个选自N、S和O的杂原子的5-10元的杂芳环,或A为H;
m为1、2、3或4;
n为0、1、2、3、4或5;
p为0、1或2;
Ra选自下组:卤素、CN、取代或未取代的C1-C6烷基、或
Figure PCTCN2021078965-appb-000016
其中,所述的环B为C3-C8环烷基、6-10元芳基、5-10元的杂芳基、具有1-3个选自下组N、S和O的杂原子的3-10元杂环基;
各个R 1、R 2、R 3和R 4各自独立地选自下组:H、卤素、CN、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C3-C8环烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-10元杂芳基、R 7-C(=O)-、R 7-C(=O)具有1-3个选自下组N、S和O的杂原子的3-12元杂环基-、R 8-S(=O) 2-、R 9R 10N-C(=O)-、R 9R 10N-C(=O)NR-、R 11R 12N-S(=O) 2-、R 7O-、R 8-S(O) 2NR-;
各个R 7、R 8、R 9、R 10、R 11、R 12各自独立地选自下组:H、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C3-C8环烷基、取代或未取代的具 有1-3个选自下组N、S和O的杂原子的3-12元杂环基;或所述的R 9和R 10或R 11和R 12与其相连的氮原子共同构成取代或未取代的具有1-3个选自N、S和O的杂原子的3-12元杂环基;
除非特别说明,所述的“取代”是指被选自下组的一个或多个(例如2个、3个、4个等)取代基所取代:卤素、C1-C6烷氧基、卤代的C1-C6烷氧基、C3-C8环烷基、卤代的C3-C8环烷基、甲基砜基、氧代(=O)、-CN、羟基、-NH 2、-NHS(O) 2CH 3、C1-C6胺基、羧基、C1-C6酰胺基(-C(=O)-N(Rc) 2或-NH-C(=O)(Rc),Rc为H或C1-C5的烷基)、或取代或未取代的选自下组的基团:C1-C6烷基、C6-C10芳基、具有1-3个选自N、S和O的杂原子的5-10元杂芳基、-(CH 2)-C6-C10芳基、-(CH 2)-(具有1-3个选自N、S和O的杂原子的3-10元杂芳基)、-(具有1-3个选自N、S和O的杂原子的5-10元亚杂芳基)-(C1-C6烷基)、具有1-3个选自下组N、S和O的杂原子的3-12元杂环基(包括单环、螺环、桥环或并环),且所述的取代基选自下组:卤素、C1-C6烷基、C1-C6亚烷基-OH、C1-C6烷氧基、氧代、-S(O) 2CH 3、-CN、-OH、C6-C10芳基、具有1-3个选自N、S和O的杂原子的3-10元杂芳基、-C(O)CHNH 2、-C(O)CHOH;
且所述的式I化合物中,各个手性中心为R构型或S构型。
在另一优选例中,所述的“取代”是指被选自下组的一个或多个(例如2个、3个、4个等)取代基所取代:卤素、C1-C6烷氧基、卤代的C1-C6烷氧基、C3-C8环烷基、卤代的C3-C8环烷基、甲基砜基、氧代(=O)、-CN、羟基、-NH 2、C1-C6胺基、羧基、或取代或未取代的选自下组的基团:C1-C6烷基、C6-C10芳基、具有1-3个选自N、S和O的杂原子的5-10元杂芳基、且所述的取代基选自下组:卤素、C1-C6烷基、C1-C6亚烷基-OH、C1-C6烷氧基、氧代、-S(O) 2CH 3、-CN、-OH、C6-C10芳基、具有1-3个选自N、S和O的杂原子的3-10元杂芳基、-C(O)CHNH 2、-C(O)CHOH。
优选地,所述的X、Y、A、B、M 1、M 2、M 3、m、n、p、q、Ra、Re、Rf、R 1、R 2、R 3、R 4、R 7、R 8、R 9、R 10、R 11、R 12各自独立地为各个实施例中具体化合物中的对应基团。
本发明的化合物可以作为HPK1激酶抑制剂,在优选的实施例中,为HPK1激酶选择性抑制剂。
式I化合物的制备
本发明的式I化合物可以通过以下示例性的方法制备:
方法1:
Figure PCTCN2021078965-appb-000017
用五元并六元化合物进行取代、偶联反应,经取代基修饰后得到式I化合 物。
其中,一类A为不饱和环的化合物可以通过方法2制备:
Figure PCTCN2021078965-appb-000018
用Ia化合物进行环合反应,得到式I'化合物。
药物组合物和施用方法
由于本发明化合物具有优异的HPK1激酶的抑制活性,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预防和/或治疗与HPK1激酶活性或表达量相关的疾病(例如,癌症)。
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及药学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1-2000毫克本发明化合物/剂,更佳地,含有10-200毫克本发明化合物/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化合物以及它们之间相互掺和,而不明显降低化合物的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如吐温
Figure PCTCN2021078965-appb-000019
)、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、肠胃外(静脉内、肌肉内或皮下)注射等。
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。
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。
联合给药时,所述药物组合物还包括与一种或多种其他药学上可接受的化合物。该其他药学上可接受的化合物中的一种或多种可与本发明的化合物同时、分开或顺序地给药。
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1~2000毫克,优选20~500毫克。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。
各实施例中:
分析方法I
LCMS仪器:waters Acquity UPLC-MS,UV检测器:Acquity UPLC
色谱柱:Acquity UPLC HSS T3 1.8uM,柱温40℃
流动相:A:H2O(0.1%TFA),B:乙腈,梯度洗脱。
以下实施例中所用的实验材料和试剂如无特别说明均可从市售渠道获得。
中间体A1合成:
Figure PCTCN2021078965-appb-000020
5-碘-4-氯-7H-吡咯并[2,3-d]嘧啶
Figure PCTCN2021078965-appb-000021
在室温下,向4-氯-7H-吡咯并[2,3-d]嘧啶(10克,65.12毫摩尔)的N,N-二甲基甲酰胺(400毫升)溶液中分批加入N-碘代琥珀酰亚胺(15.7克,69.77毫摩尔)。将反应液于室温搅拌过夜,反应经LCMS检测。反应液浓缩至干,残留物悬浮在250毫升10%的亚硫酸钠水溶液中,过滤,滤饼用乙醇重结晶,得到白色固体5-碘-4-氯-7H-吡咯并[2,3-d]嘧啶(12.6克,纯度:90%,收率:69.3%)。MS(ESI):m/z=280.0,282.0[M+H] +.
4-氯-5-碘-7-(2-三甲基硅烷基-乙氧基甲基)-7H-吡咯并[2,3-d]嘧啶
Figure PCTCN2021078965-appb-000022
在冰水浴下,将60%的氢化钠(2.53克,6.22毫摩尔)分批加到5-碘-4-氯-7H-吡咯并[2,3-d]嘧啶(12.6克,45.16毫摩尔)的N,N-二甲基甲酰胺(120毫升)中,该反应混合物于0度下继续搅拌30分钟。然后将2-(三甲基硅烷基)乙氧甲基氯(9.77克,58.71毫摩尔)的N,N-二甲基甲酰胺(10毫升)滴加至反应液中,滴加完毕后,反应混合物在室温下搅拌1.5小时,反应经LCMS检测。将反应液倒入500毫升水中,用500毫升乙醚提取,有机相用无水硫酸钠干燥,抽滤,滤液浓缩,残留物通过正相硅胶层析柱,用溶剂(石油醚:乙酸乙酯=5:1)洗脱,得到的白色固体中间体4-氯-5-碘-7-(2-三甲基硅烷基-乙氧基甲基)-7H-吡咯并[2,3-d]嘧啶(14.0克,纯度:90%,收率:75.7%)。MS(ESI):m/z=410.1,412.1[M+H] +.
叔丁基(4-((5-碘-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯[2,3-d]嘧啶-4-基) 氧基)-3,5-二氟苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000023
向4-氯-5-碘-7-(2-三甲基硅烷基-乙氧基甲基)-7H-吡咯并[2,3-d]嘧啶(2.00克,4.88毫摩尔)和叔丁基(3,5-二氟-4-羟基苯基)氨基甲酸酯(中间体B1,1.26克,5.13毫摩尔)的二甲基亚砜(20毫升)溶液中加入碳酸钾(1.35克,9.76毫摩尔)。然后将该混合物加热至110度搅拌2小时。反应经LCMS检测,待反应结束后将反应冷却至室温。将反应液倒入60毫升水中,该混合物经乙酸乙酯(20毫升×3)萃取。合并的有机相依次经水(20毫升×3),饱和食盐水(20毫升)洗涤,无水硫酸钠干燥,过滤,滤液经减压浓缩得粗品。该粗品经快速硅胶分离(石油醚:乙酸乙酯=5:1),得到的白色固体叔丁基(4-((5-碘-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯[2,3-d]嘧啶-4-基)氧基)-3,5-二氟苯基)氨基甲酸酯(2.43克,收率:80.5%)。MS(ESI):m/z=619.0[M+H] +.
3,5-二氟-4-((5-碘-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧 代)苯胺
Figure PCTCN2021078965-appb-000024
将(4-((5-碘-7-((2-(三甲硅基)乙氧基)甲基)-7H吡咯[2,3-d]嘧啶-4-基)氧基)-3,5-二氟苯基)氨基甲酸酯(2.27克,3.67毫摩尔)的1,1,1,3,3,3-六氟丙烷-2-醇溶液(30毫升)加入到150毫升的密封管中。将该反应液加热至110度,搅拌1小时。反应经LCMS检测,待反应结束后,待反应液冷却至室温。减压浓缩得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=3:1)分离得浅黄色3,5-二氟-4-((5-碘-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯胺(1.6克,收率:84.1%)。MS(ESI):m/z=618.9[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.42(s,1H),7.40(s,1H),6.36–6.28(m,2H),5.60(s,2H),3.85(s,2H),3.59–3.50(m,2H),1.26(t,J=7.2Hz,1H),0.97–0.88(m,2H),-0.04(s,9H).
中间体A2合成:
Figure PCTCN2021078965-appb-000025
4-氯-5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶
Figure PCTCN2021078965-appb-000026
在氮气保护下,向4-氯-5-碘-7-(2-三甲基硅烷基-乙氧基甲基)-7H-吡咯并[2,3-d]嘧啶(1.10克,2.68毫摩尔),甲基2,2-二氟-2-(氟磺酰)乙酸酯(1.03克,2..37毫摩尔)的无水N,N-二甲基甲酰胺(10毫升)溶液中加入碘化亚铜(1.02克,5.37毫摩尔)固体。将该反应液加热至100度,搅拌2小时。反应经LCMS检测,待反应结束后,将反应液冷却室温。将反应液倒入30毫升水中,该混合物经乙酸乙酯(15毫升×3)萃取。合并的有机相依次经水(20毫升×3),饱和食盐水(20毫升)洗涤,无水硫酸钠干燥,过滤,滤液经减压浓缩得粗品。该粗品经快速硅胶分离(石油醚:乙酸乙酯=6:1),得到的白色固体4-氯-5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(670毫克,纯度:86%,收率:61%)。MS(ESI):m/z=352.0[M+H] +.
叔丁基(3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H 吡咯[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000027
向4-氯-5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶(450毫克,1.28毫摩尔)和叔丁基(3,5-二氟-4-羟基苯基)氨基甲酸酯(中间体B1,345毫克,1.41毫摩尔)的二甲基亚砜(5.0毫升)溶液中加入无水碳酸钾(353毫克,2.56毫摩尔)。然后将该混合物加热至110度搅拌2小时。反应经LCMS检测,待反应结束后将反应冷却至室温。将反应液倒入20毫升水中,该混合物经乙酸乙酯(10毫升×3)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩得粗品。该粗品经快速硅胶分离(石油醚:乙酸乙酯=3:1),得到的白色固体叔丁基(3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H吡咯[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(430毫克,收率:60%)。MS(ESI):m/z=561.1[M+H] +.
3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶- 4-基)氧代)苯胺
Figure PCTCN2021078965-appb-000028
将((3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H吡咯[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(330毫克,0.59毫摩尔)的1,1,1,3,3,3-六氟丙烷-2-醇溶液(8毫升)加入到20毫升的微波中。将该反应液加热至110度,搅拌1小时。反应经LCMS检测,待反应结束后,待反应液冷却至室温。减压浓缩得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=3:2)分离得浅黄色固体3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯胺(252毫克,收率:92.9%)。MS(ESI):m/z=461.0.[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.52(s,1H),7.67(s,1H),6.33(d,J=9.2Hz,2H),5.66(s,2H),3.64–3.56(m,2H),0.99–0.90(m,2H),-0.01–-0.04(m,9H).
中间体A3合成:
Figure PCTCN2021078965-appb-000029
3-溴-4-氯-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000030
将化合物4-氯-1H-吡咯并[2,3-b]吡啶-5-甲腈(860毫克,4.8毫摩尔)溶于N,N-二甲基甲酰胺(10毫升)中,在0度下,加入N-溴代丁二酰亚胺(1.03克,5.8毫摩尔)。反应缓慢升温到室温并搅拌1小时。LCMS检测反应。反应结束后,将反应液缓慢倒入水(30毫升)中,用乙酸乙酯(10毫升×3)萃取。合并的有机相依次经水(10毫升×2),饱和食盐水(10毫升)洗涤,无水硫酸钠干燥、过滤,滤液经减压浓缩得粗 品。该粗品经快速硅胶柱(石油醚:乙酸乙酯=1:1)分离纯化得到3-溴-4-氯-1H-吡咯并[2,3-b]吡啶-5-甲腈(1.2克,收率:98%)。MS(ESI):m/z=255.9,257.9[M+H] +.
3-溴-4-氯-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000031
将3-溴-4-氯-1H-吡咯并[2,3-b]吡啶-5-甲腈(1.2克,4.7毫摩尔)溶于N,N-二甲基甲酰胺(12毫升)中,在0度下,缓慢加入氢化钠(纯度:60%,282毫克,7.1毫摩尔)。反应在0度下搅拌30分钟,然后向溶液中缓慢滴加2-(三甲基硅烷基)乙氧甲基氯(1.02克,6.1毫摩尔)。滴加完毕后,将反应液于0度继续搅拌30分钟。LCMS检测反应反,待反应完成后,向反应液缓慢加入到水中(40毫升),用乙酸乙酯萃取(20毫升×3)。合并的有机相依次用水(20毫升×3)、饱和食盐水溶液(20毫升)洗涤,无水硫酸钠干燥,过滤,滤液经减压浓缩得粗品。该粗产品经快速硅胶柱纯化(石油醚:乙酸乙酯=5:1)得3-溴-4-氯-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(1.5克,收率:83%)。MS(ESI):m/z=386.0,388.0[M+H] +.
叔丁基(4-((3-溴-5-氰基-1-((2-(三甲硅基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧 基)-3,5-二氟苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000032
将3-溴-4-氯-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(300毫克,0.776毫摩尔)溶于二甲基亚砜(5.0毫升)中,再依次加入叔丁基(3,5-二氟-4-羟基苯基)氨基甲酸酯(323毫克,1.32毫摩尔)、无水碳酸钾(268毫克,1.94毫摩尔),氮气置换三次后,将该反应置于室温搅拌16小时。反应经LCMS检测。将反应液倒入水(20毫升)中,加乙酸乙酯(20毫升×3)萃取。合并的有机相依次用水(10毫升×3)、饱和食盐水(10毫升)洗涤、无水硫酸钠干燥、过滤,滤液经减压浓缩得粗品。该粗品经硅胶柱(石油醚:乙酸乙酯=1:4)分离纯化得到叔丁基(4-((3-溴-5-氰基-1-((2-(三甲硅基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3,5-二氟苯基)氨基甲酸酯(398毫克,产率:86.1%)。MS(ESI):m/z=595.3,597.3[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.38(s,1H),7.41(s,1H),7.26(s,2H),7.15(m,2H),6.61(s,1H),5.64(s,2H),3.57-3.52(m,2H),1.52(s,9H),0.94-0.90(m,2H),-0.04(s,9H).
4-(4-氨基-2,6-二氟苯氧基)-3-溴-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3- b]吡啶-5-腈
Figure PCTCN2021078965-appb-000033
将叔丁基(4-((3-溴-5-氰基-1-((2-(三甲硅基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3,5-二氟苯基)氨基甲酸酯(100毫克,0.168毫摩尔)溶于1,1,1,3,3,3-六氟丙烷-2-醇溶液(2.0毫升)中,反应在氮气保护下于100度加热搅拌16小时。LCMS检测反应。待反应液冷却至室温后,减压蒸除溶剂得油状物。该油状物经硅胶柱分离纯化(石油醚:乙酸乙酯=1:2)得到4-(4-氨基-2,6-二氟苯氧基)-3-溴-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(80毫克,收率:96.1%)。MS(ESI):m/z=495.3,497.3[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.37(s,1H),7.39(s,1H),7.26(s,1H),6.33-6.28(m,2H),5.63(s,2H),3.93(br s,2H),3.54(dd,J=7.7,8.9Hz,1H),3.56-3.52(m,1H),0.94-0.90(m,2H),-0.04(s,9H).
中间体A4合成:
Figure PCTCN2021078965-appb-000034
叔丁基(3-氯-4-羟基苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000035
在氩气保护下,向2-氯-4-硝基苯酚(3.00克,17.3毫摩尔)和二碳酸二叔丁酯(4.52克,20.8毫摩尔)的甲醇(50毫升)溶液中加入钯/碳(10%,1.0克),氢气置换三次。该反应于室温下,在氢气(30psi)气氛中反应6小时。LCMS检测反应完。过滤,滤液经减压浓缩得粗品。该粗品经快速硅胶柱分离纯化(石油醚:乙酸乙酯=1:4)得叔丁基(3-氯-4-羟基苯基)氨基甲酸酯(1.21克,收率:28.7%))MS(ESI):m/z=187.9[M-55] +.
1H NMR(400MHz,CDCl 3)δ7.56(br s,1H),7.02(dd,J=4.0,8.0Hz,1H),6.93(d,J=8.0Hz,1H),6.37(br s,1H),5.41(br s,1H),1.51(s,9H).
叔丁基(4-((3-溴-5-氰基-1-((2-(三甲硅基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4- 基)氧基)-3-氯苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000036
向叔丁基(3-氯-4-羟基苯基)氨基甲酸酯(208毫克,0.85毫摩尔),3-溴-4-氯-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-甲腈(300毫克,0.776毫摩尔)和无水碳酸钾(268毫克,1.94毫摩尔)的混合物中加入二甲基亚砜(5.0毫升)。氮气置换三次后,将该反应置于室温搅拌16小时。反应经LCMS检测。将反应液倒入水(20毫升)中,加乙酸乙酯(10毫升×3)萃取。合并的有机相依次用水(10毫升×3)、饱和食盐水(10毫升)洗涤、无水硫酸钠干燥、过滤,滤液经减压浓缩得粗品。该粗产品经硅胶柱(石油醚:乙酸乙酯=1:6)分离纯化得粗品叔丁基(4-((3-溴-5-氰基-1-((2-(三甲硅基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氯苯基)氨基甲酸酯(600毫克)。MS(ESI):m/z=593.2,595.2[M+H] +.
4-(4-氨基-2-氯苯氧基)-3-溴-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡 啶-5-腈
Figure PCTCN2021078965-appb-000037
将叔丁基(4-((3-溴-5-氰基-1-((2-(三甲硅基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氯苯基)氨基甲酸酯(500毫克,0.842毫摩尔)溶于1,1,1,3,3,3-六氟丙烷-2-醇溶液(25毫升)中,反应在氮气保护下于100度加热搅拌16小时。TLC检测反应。待反应液冷却至室温后,减压蒸除溶剂得油状物。该油状物经硅胶柱分离纯化(石油醚:乙酸乙酯=0-20%)得4-(4-氨基-2-氯苯氧基)-3-溴-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(285毫克,收率:57.0%)。MS(ESI):m/z=493.0,495.0[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.38(s,1H),7.39(s,1H),6.93(d,J=8.8Hz,1H),6.79(d,J=2.8Hz,1H),6.57(dd,J=2.8,8.8Hz,1H),5.63(s,2H),3.78(br,s,2H),3.59-3.51(m,2H),0.97-0.88(m,2H),-0.02--0.06(m,9H).
中间体A5合成:
Figure PCTCN2021078965-appb-000038
Figure PCTCN2021078965-appb-000039
4- 氯-3-碘-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000040
在室温下,向4-氯-1H-吡咯并[2,3-b]吡啶-5-腈(700毫克,3.95毫摩尔)的N,N-二甲基甲酰胺(15毫升)溶液中分批加入N-碘代琥珀酰亚胺(930毫克,4.15毫摩尔),该反应液在氮气保护下于零度搅拌1小时。反应经TLC检测。反应结束后,将反应液倒入水中(60毫升),加乙酸乙酯(20毫升×3)萃取。合并的有机相依次用水(10毫升×3)、饱和食盐水(10毫升)洗涤、无水硫酸钠干燥、过滤,滤液经减压浓缩得粗品。该粗品经快速硅胶分离(石油醚:乙酸乙酯=3:2)得4-氯-3-碘-1H-吡咯并[2,3-b]吡啶-5-腈(1.00克,收率:84%)。MS(ESI):m/z=304.0,306.0[M+H] +.
4-氯-3-碘-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000041
在冰水浴下,将60%的氢化钠(265毫克,6.6毫摩尔)分批加到4-氯-3-碘-1H-吡咯并[2,3-b]吡啶-5-腈(1.00克,3.3毫摩尔)的无水四氢呋喃溶液(20毫升)中,该反应混合物于0度下继续搅拌30分钟。然后滴加2-(三甲基硅烷基)乙氧甲基氯(826毫克,4.9毫摩尔),滴加加毕后,将该反应混合物在室温下搅拌1.5小时,反应经LCMS检测。将反应液倒入50毫升水中,乙酸乙酯(20毫升×3)萃取。合并的有机相用无水硫酸钠干燥,过滤,滤液经减压浓缩得粗品。该粗品硅胶分离(石油醚:乙酸乙酯=5:1)得到4-氯-3-碘-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(900毫克,收率:71.4%)。MS(ESI):m/z=424.0,426.0[M+H] +.
4-氯-3-(三氟甲基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000042
在氮气保护下,向4-氯-3-碘-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(900毫克,2.08毫摩尔),甲基2,2-二氟-2-(氟磺酰)乙酸酯(1.2克,6.25毫摩尔)的无水N,N-二甲基甲酰胺(10毫升)溶液中加入碘化亚铜(1.18克,6.25毫摩尔)固体。将该反应液加热至100度,搅拌4小时。反应经LCMS检测,待反应结束后,将反应液冷却室温。将反应液倒入30毫升水中,该混合物经乙酸乙酯(15毫升×3)萃取。合并的有机相依次经水(20毫升×3),饱和食盐水(20毫升)洗涤,无水硫酸钠干燥,过滤,滤液经减压浓缩得粗品。该粗品经快速硅胶分离(石油醚:乙酸乙酯=6:1-3:1),得4-氯-3-(三氟甲基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(650毫克,产率88.1%)。MS(ESI):m/z=376.1,378.1[M+H] +.
4-氯-3-(三氟甲基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000043
将4-氯-3-(三氟甲基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(700毫克,1.86毫摩尔)溶于二甲基亚砜(12毫升)中,再依次加入叔丁基(3-氟-4-羟基苯基)氨基甲酸酯(430毫克,2.24毫摩尔)、无水碳酸钾(513毫克,3.72毫摩尔),氮气置换三次后,将该反应置于室温搅拌14小时。反应经LCMS检测。将反应液倒入水(20毫升)中,加乙酸乙酯(20毫升×3)萃取。合并的有机相依次用水(10毫升×3)、饱和食盐水(10毫升)洗涤、无水硫酸钠干燥、过滤,滤液经减压浓缩得粗品。该粗品经硅胶柱(石油醚:乙酸乙酯=1:4)分离纯化得到4-氯-3-(三氟甲基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(570毫克,收率:53.9%)。MS(ESI):m/z=589.1[M+Na] +.
1H NMR(400MHz,DMSO-d 6)δ9.76(s,1H),8.85(s,1H),8.66(s,1H),7.72–7.58(m,1H),7.29-7.27(m,2H),5.80(s,2H),3.70–3.66(m,2H),1.55(s,9H),1.00–0.84(m,2H),0.04(s,9H).
4-(4-氨基-2-氟苯氧基)-3-(三氟甲基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯 并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000044
将叔丁基(4-((3-溴-5-氰基-1-((2-(三甲硅基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氯苯基)氨基甲酸酯(500毫克,0.842毫摩尔)溶于1,1,1,3,3,3-六氟丙烷-2-醇溶液(25毫升)中,反应在氮气保护下于100度加热搅拌16小时。TLC检测反应。待反应液冷却至室温后,减压蒸除溶剂得油状物。该油状物经硅胶柱分离纯化(石油醚:乙酸乙酯=1:4)得4-(4-氨基-2-氯苯氧基)-3-溴-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(285毫克,收率:57.0%)。MS(ESI):m/z=467.2[M+H] +.
中间体A6合成:
Figure PCTCN2021078965-appb-000045
叔丁基(4-((3-溴-5-氰基-1-((2-(三甲硅基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧 基)-3-氟苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000046
将3-溴-4-氯-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(1.50克,3.9毫摩尔)溶于二甲基亚砜(12毫升)中,再依次加入叔丁基(3-氟-4-羟基苯基)氨基甲酸酯(973毫克,4.3毫摩尔)、无水碳酸钾(1.3克,9.8毫摩尔),氮气置换三次后,将该反应置于室温搅拌24小时。反应经LCMS检测。将反应液倒入水(50毫升)中,加乙酸乙酯(20毫升×3)萃取。合并的有机相依次用水(20毫升×3)、饱和食盐水(30毫升)洗涤、无水硫酸钠干燥、过滤,滤液经减压浓缩得粗品。该粗品经硅胶柱(石油醚:乙酸乙酯=0-15%)分离纯化得叔丁基(4-((3-溴-5-氰基-1-((2-(三甲硅基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)氨基甲酸酯(1.50克,收率:69%)。MS(ESI):m/z=577.1,579.1[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.47(s,1H),7.56(d,J=12.4Hz,1H),7.45(s,1H),7.05–6.99(m,2H),6.59(s,1H),5.68(s,2H),3.59(dd,J=8.8,7.7Hz,2H),1.55(s,9H),0.98–0.94(m,2H),0.01(s,9H).
4-(4-氨基-2-氟苯氧基)-3-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡 啶-5-腈
将叔丁基(4-((3-溴-5-氰基-1-((2-(三甲硅基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)氨基甲酸酯(700毫克,1.26毫摩尔)溶于1,1,1,3,3,3-六氟丙烷-2-醇溶液(12毫升)中,反应在氮气保护下于100度加热搅拌2小时。TLC检测反应。待反应液冷却至室温后,减压蒸除溶剂得油状物。该油状物经硅胶柱分离纯化(石油醚:乙酸乙酯=1:3)得4-(4-氨基-2-氟苯氧基)-3-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(425毫克,收率:71.0%)。MS(ESI):m/z=477.1,479.1[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ8.67(s,1H),8.12(s,1H),6.98(t,J=9.2Hz,1H),6.56(dd,J=13.2,2.4Hz,1H),6.42(dd,J=8.8,2.0Hz,1H),5.71(s,2H),5.53(s,2H),3.65–3.59(m,2H),0.95–0.88(m,2H),-0.00(s,9H).
中间体B1合成:
Figure PCTCN2021078965-appb-000047
2,6-二氟-4-硝基苯酚
Figure PCTCN2021078965-appb-000048
将2,6-二氟苯酚(1.7克,13毫摩尔)溶于醋酸中(20毫升),滴加入浓硝酸(1.9克,20毫摩尔)。反应液在70度下反应2小时。TLC检测反应完成。将反应液倒入冰水中(100毫升),用乙酸乙酯萃取(100毫升×2)。合并的有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,滤液经减压浓缩得粗品。该粗品经硅胶柱(石油醚:乙酸乙酯=4:1)分离纯化,得到黄色固体2,6-二氟-4-硝基苯酚(1.57克,收率:69%)。
1H NMR(400MHz,DMSO-d 6)δ7.95(dd,J=6.8,2.0Hz,2H).
叔丁基(3,5-二氟-4-羟基苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000049
在氩气保护下,向2,6-二氟-4-硝基苯酚(1.57克,8.97毫摩尔)和二-叔-丁基二碳酸酯(2.9克,13mmol)甲醇溶液(30毫升)中加入钯/碳(500毫克)。将该反应液用氢气置换三次后,于室温条件下在氢气(15psi)气氛中搅拌反应16小时。TLC检测反应完成。反应液经过滤,滤液经减压浓缩得粗。该粗品经硅胶柱(油醚:乙酸乙酯=4:1)分离纯化,得到灰色固体(1.3克,收率:59%)。
1HNMR(400MHz,DMSO-d 6)δ9.64(s,1H),9.40(s,1H),7.12(d,J=9.6Hz,2H),1.46(s,9H).
实施例1:(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基) 氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000050
3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d] 嘧啶-4-基)氧代)苯胺
Figure PCTCN2021078965-appb-000051
在氩气保护下,向3,5-二氟-4-((5-碘-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯胺(A1,400毫克,0.945毫摩尔),(2-氟-4-甲氧苯基)硼酸(178毫克,1.04毫摩尔),磷酸钾(401毫克,1.89毫摩尔)和1,4-二氧六环(6.0毫升)/水(2.0毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(76毫 克,0.094毫摩尔)。将该反应液加热至100度搅拌2小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(10毫升×2)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=2:1)分离得3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯胺(439毫克,收率:83.9%)。MS(ESI):m/z=517.1[M+H] +.
1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并 [2,3-d]嘧啶-4-基)氧代)苯基)-3-((3-甲基噁丁环-3-基)甲基)脲
Figure PCTCN2021078965-appb-000052
向配有磁力搅拌器的50毫升圆底烧瓶中加入3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯胺(102毫克,0.197毫摩尔)和三乙胺(80毫克,0.79毫摩尔)的无水二氯甲烷(3.0毫升)溶液,氩气置换三次后将反应液置于冰浴中冷却。向反应液中加入三光气(41毫克,0.138毫摩尔),该反应液于0度搅拌20分钟。最后向反应液中加入(3-甲基噁丁环-3-基)甲胺(120毫克,1.18毫摩尔),撤掉冰浴,将反应液在于室温搅拌30分钟。LCMS检测反应。减压浓缩后油状物。该油状物经反向C-18硅胶柱(HCOOH)分离得1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((3-甲基噁丁环-3-基)甲基)脲(78毫克,收率61.4%)。MS(ESI):m/z=644.2[M+H] +.
(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲 基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000053
在0度下,向1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((3-甲基噁丁环-3-基)甲基)脲(78毫克,0.12毫摩尔)的无水二氯甲烷(3.0毫升)中加入三氟乙酸(1.0毫升)。撤去冰浴,该反应液于室温搅拌4小时。减压浓缩溶液的油状残留物,加入无水甲醇(4.0毫升),用冰浴冷却至0度后,向反应液中加入无水碳酸钾调节pH=10-11。开动搅拌器,搅拌30分钟后,过滤,滤液经减压浓缩得粗品。该粗品经反向C-18硅胶柱(NH 4HCO 3)分离得(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(51毫克,收率:82%)。MS(ESI):m/z=514.0.[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.25(s,1H),7.57(t,J=8.8Hz,1H),7.45(d,J=1.6Hz,1H),6.93(s,2H),6.80–6.74(m,2H),4.16(dd,J=10.6,1.6Hz,1H),3.94(d,J=10.6Hz,1H),3.81(s,3H),3.52(d,J=11.2Hz,1H),3.43(d,J=11.2Hz,1H),3.25(d,J=13.6Hz,1H),3.04(d,J=13.6Hz,1H),1.01(s,3H).
实施例2:2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨 基)-5-氟-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000054
1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d] 嘧啶-4-基)氧代)苯基)-3-((3-氟噁丁环-3-基)甲基)脲
Figure PCTCN2021078965-appb-000055
向配有磁力搅拌器的50毫升圆底烧瓶中加入3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯胺(100毫克,0.193毫摩尔)和三乙胺(79毫克,0.77毫摩尔)的无水二氯甲烷(3.0毫升)溶液,氩气置换三次,然后将反应液置于冰浴中冷却。然后向反应液中加入三光气(40毫克,0.135毫摩尔),该反应液于0度搅拌20分钟。然后向反应液中加入(3-氟噁丁环-3-基)甲胺(102毫克,0.969毫摩尔),撤掉冰浴,将反应液在于室温搅拌30分钟。LCMS检测反应。减压浓缩后油状物。该油状物经反向C-18硅胶柱(HCOOH)分离得白色固体1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((3-氟噁丁环-3-基)甲基)脲(80毫克,收率:63.8%)。MS(ESI):m/z=648.1[M+H] +.
(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-氟- 5,6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000056
以1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((3-氟噁丁环-3-基)甲基)脲(80毫克,0.123毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇的合成方法合成(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-氟-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(54毫克,收率:84.5%)。MS(ESI):m/z=518.2[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.26(s,1H),7.56(m,1H),7.44(d,J=1.6Hz,1H),7.14(s,2H),6.79–6.72(m,2H),4.34–4.21(m,2H),3.79(m,3H),3.76–3.44(m,5H).
实施例3:N-(3,5-二氟-4-((3-(2-氟-4-甲氧苯基)-1H-吡咯并[2,3-b]吡啶-4-基)氧代)苯基)- 5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺
Figure PCTCN2021078965-appb-000057
1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d] 啶-4-基)氧代)苯基)-3-((1-(羟甲基)环丙基)甲基)硫代脲
Figure PCTCN2021078965-appb-000058
在室温下,向配有磁力搅拌器的10毫升圆底烧瓶中加入3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯胺(150毫克,0.29毫摩尔)的无水二氯甲烷(3.0毫升)溶液中加入1,1'-硫羰基双(吡啶-2(1H)-酮)(74毫克,0.32毫摩尔),氩气置换三次,然后将反应液置于40度油浴中搅拌1小时。待原料转化完全后,将反应液冷却至室温,然后加入(1-(氨基甲基)环丙基)甲醇(59毫克,0.58毫摩尔)。该反应于室温搅拌3小时。反应经LCMS检测。减压浓缩溶剂得油状粗品,经反向C-18硅胶柱(HCOOH)分离得白色固体1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((1-(羟甲基)环丙基)甲基)硫代脲(180毫克,收率:94%)。MS(ESI):m/z=660.1[M+H] +.
N-(3,5-二氟-4-((3-(2-氟-4-甲氧苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并 [2,3-b]吡啶-4-基)氧代)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺
Figure PCTCN2021078965-appb-000059
向配有磁力搅拌器的10毫升烧瓶中依次加入1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((1-(羟甲基)环丙基)甲基)硫代脲(80毫克,0.12毫摩尔)和三乙胺(37毫克,0.36毫摩尔)和无水二氯甲烷(1.5毫升),氩气置换三次后,将反应液置于冰-水浴中冷却10分钟后,然后向反应液中加入3-(((乙基亚氨基)亚甲基)氨基)-N,N-二甲基丙烷-1-胺盐酸(47毫克,0.24毫摩 尔)。撤去冰浴,该反应于室温搅拌24小时。LCMS检测反应。减压浓缩溶剂得油状粗品,经反向C-18硅胶柱(HCOOH)分离得白色固体N-(3,5-二氟-4-((3-(2-氟-4-甲氧苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧代)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺(24毫克,收率:32%)。MS(ESI):m/z=626.1[M+H] +.
N-(3,5-二氟-4-((3-(2-氟-4-甲氧苯基)-1H-吡咯并[2,3-b]吡啶-4-基)氧代)苯基)-5-氧杂-7-氮 杂螺[2.5]辛-6-烯-6-胺
Figure PCTCN2021078965-appb-000060
以N-(3,5-二氟-4-((3-(2-氟-4-甲氧苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧代)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺(24毫克,0.038毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇的合成方法合成N-(3,5-二氟-4-((3-(2-氟-4-甲氧苯基)-1H-吡咯并[2,3-b]吡啶-4-基)氧代)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺(3.6毫克,收率:19%)。MS(ESI):m/z=496.1[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.25(s,1H),7.57(t,J=8.8Hz,1H),7.45(d,J=1.2Hz,1H),6.96(m,2H),6.80–6.73(m,2H),4.08(s,2H),3.81(s,3H),3.22(s,2H),0.74–0.64(m,4H).
实施例4:1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)- 3-((1-(羟甲基)环丙基)甲基)硫代脲
Figure PCTCN2021078965-appb-000061
以1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((1-(羟甲基)环丙基)甲基)硫代脲(20毫克,0.030毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇的合成方法合成1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((1-(羟甲基)环丙基)甲基)硫代脲(2.5毫克,收率:15.6%)。MS(ESI):m/z=530.0[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.27(s,1H),7.57(t,J=8.8Hz,1H),7.47(s,1H),7.32(d,J=9.6Hz,2H),6.82–6.73(m,2H),3.81(s,3H),3.65(s,2H),3.45(s,2H),0.59(m,2H),0.51(m,2H).
实施例5:(2-((3,5-二氟-4-((5-(三氟甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5- 氟-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000062
1-(3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧 啶-4-基)氧代)苯基)-3-((3-氟噁丁环-3-基)甲基)脲
Figure PCTCN2021078965-appb-000063
以3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯胺(77毫克,0.167毫摩尔)和(3-氟噁丁环-3-基)甲胺(88毫克,0.836毫摩尔)为原料参考化合物1-(3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((3-氟噁丁环-3-基)甲基)脲(化合物2-1)的方法合成1-(3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((3-氟噁丁环-3-基)甲基)脲(65毫克,收率:65.7%)MS(ESI):m/z=592.1[M+H] +.
(2-((3,5-二氟-4-((5-(三氟甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-氟-5,6-二 氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000064
以1-(3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((3-氟噁丁环-3-基)甲基)脲(65毫克,0.11毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇的合成方法合成(2-((3,5-二氟-4-((5-(三氟甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-氟-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(29毫克,收率:57.2%)。MS(ESI):m/z=462.0[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.35(s,1H),7.87(d,J=1.2Hz,1H),7.25–7.10(m,2H),4.29(m,2H),3.74–3.48(m,4H).
实施例6:N-(3,5-二氟-4-((5-(三氟甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-5-氧杂- 7-氮杂螺[2.5]辛-6-烯-6-胺
Figure PCTCN2021078965-appb-000065
1-(3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧 啶-4-基)氧代)苯基)-3-((1-(羟甲基)环丙基)甲基)硫代脲
Figure PCTCN2021078965-appb-000066
在室温下,向配有磁力搅拌器的10毫升圆底烧瓶中加入3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯胺(252毫克,0.55毫摩尔)的无水二氯甲烷(4.0毫升)溶液中加入1,1'-硫羰基双(吡啶-2(1H)-酮)(140毫克,0.60毫摩尔),氩气置换三次,然后将反应液置于40度油浴中搅拌1小时。待原料转化完全后,将反应液冷却至室温,然后加入(1-(氨基甲基)环丙基)甲醇(111毫克,1.09毫摩尔)。该反应于室温搅拌1小时。反应经LCMS检测。减压浓缩溶剂得油状粗品,经反向C-18硅胶柱(HCOOH)分离得1-(3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((1-(羟甲基)环丙基)甲基)硫代脲(292毫克,收率:88.4%)。MS(ESI):m/z=604.0[M+H] +.
N-(3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧 啶-4-基)氧代)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺
Figure PCTCN2021078965-appb-000067
向配有磁力搅拌器的10毫升烧瓶中依次加入1-(3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-3-((1-(羟甲基)环丙基)甲基)硫代脲(292毫克,0.483毫摩尔)和三乙胺(196毫克,1.93毫摩尔)和无水二氯甲烷(5.0毫升),氩气置换三次后,将反应液置于冰-水浴中冷却10分钟后,然后向反应液中加入3-(((乙基亚氨基)亚甲基)氨基)-N,N-二甲基丙烷-1-胺盐酸(185毫克,0.967毫摩 尔)。撤去冰浴,该反应于室温搅拌24小时。LCMS检测反应。减压浓缩溶剂得油状粗品,经反向C-18硅胶柱(HCOOH)分离得白色固体N-(3,5-二氟-4-((5-(三氟甲基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺(150毫克,收率:54.4%)。MS(ESI):m/z=570.0[M+H] +.
N-(3,5-二氟-4-((5-(三氟甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-5-氧杂-7-氮杂螺 [2.5]辛-6-烯-6-胺
Figure PCTCN2021078965-appb-000068
以N-(3,5-二氟-4-((3-(三氟甲基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧代)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺(150毫克,0.263毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇的合成方法合成N-(3,5-二氟-4-((5-(三氟甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺(45毫克,收率:38.9%)。MS(ESI):m/z=440.0[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.29(s,1H),7.86(d,J=1.2Hz,1H),7.16(d,J=8.4Hz,2H),4.43(s,2H),3.31(s,2H),0.82(m,4H).
以下化合物采用与实施例1类似的方法,替换相应原料获得。
Figure PCTCN2021078965-appb-000069
Figure PCTCN2021078965-appb-000070
实施例13:N-(3,5-二氟-4-((5-(三氟甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-5-氧杂 -7-氮杂螺[2.5]辛-6-烯-6-胺
Figure PCTCN2021078965-appb-000071
3,5-二氟-4-((5-(4-氟苯基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4- 基)氧基)苯胺
Figure PCTCN2021078965-appb-000072
在氩气保护下,向3,5-二氟-4-((5-碘-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯胺(A1,300毫克,0.578毫摩尔),(4-氟苯基)硼酸(97毫克,0.694毫摩尔),磷酸钾(245毫克,1.16毫摩尔)和1,4-二氧六环(4.0毫升)/水(1.0毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(46毫 克,0.057毫摩尔)。将该反应液加热至100度搅拌3小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(5毫升×2)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=2:1)分离得3,5-二氟-4-((5-(4-氟苯基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯胺(217毫克,收率:96.2%)。MS(ESI):m/z=487.1[M+H] +.
1-(3,5-二氟-4-((5-(4-氟苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶 -4-基)氧基)苯基)-3-((1-(羟甲基)环丙基)甲基)硫脲
Figure PCTCN2021078965-appb-000073
在室温下,向配有磁力搅拌器的10毫升圆底烧瓶中加入3,5-二氟-4-((5-(4-氟苯基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯胺(135毫克,0.28毫摩尔)的无水二氯甲烷(2.0毫升)溶液中加入1,1'-硫羰基双(吡啶-2(1H)-酮)(71毫克,0.31毫摩尔),氩气置换三次,然后将反应液置于40度油浴中搅拌1小时。待原料转化完全后,将反应液冷却至室温,然后加入(1-(氨基甲基)环丙基)甲醇(56毫克,0.56毫摩尔)。该反应于室温搅拌3小时。反应经LCMS检测。减压浓缩溶剂得油状粗品,经反向C-18硅胶柱(HCOOH)分离得白色固体1-(3,5-二氟-4-((5-(4-氟苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((1-(羟甲基)环丙基)甲基)硫脲(147毫克,收率:82.5%)。MS(ESI):m/z=630.1[M+H] +.
N-(3,5-二氟-4-((5-(4-氟苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧 啶-4-基)氧基)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺
Figure PCTCN2021078965-appb-000074
向配有磁力搅拌器的10毫升烧瓶中依次加入1-(3,5-二氟-4-((5-(4-氟苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((1-(羟甲基)环丙基)甲基)硫脲(147毫克,0.23毫摩尔)和三乙胺(95毫克,0.93毫摩尔)和无水二氯甲烷(2.5毫升),氩气置换三次后,将反应液置于冰-水浴中冷却10分钟后,然后向反应液中加入3-(((乙基亚氨基)亚甲基)氨基)-N,N-二甲基丙烷-1-胺盐酸(90毫克,0.47毫摩尔)。撤去冰浴,该反应于室温搅拌24小时。LCMS检测反应。减压浓缩溶剂得油状粗品,经反向C-18硅胶柱(HCOOH)分离得白色固体N-(3,5-二氟-4-((5-(4-氟苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺(75毫克,收率:54%)。MS(ESI):m/z=596.1[M+H] +.
N-(3,5-二氟-4-((5-(4-氟苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-5-氧杂-7-氮杂螺 [2.5]辛-6-烯-6-胺
Figure PCTCN2021078965-appb-000075
以N-(3,5-二氟-4-((5-(4-氟苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺(75毫克,0.125毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(实施例1)的合成方法合成N-(3,5-二氟-4-((5-(4-氟苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-胺(39毫克,收率:66.5%)。MS(ESI):m/z=446.0[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.24(s,1H),7.73–7.67(m,2H),7.49(s,1H),7.11–7.05(m,2H),6.98(d,J=9.6Hz,2H),4.10(s,2H),3.22(s,2H),0.74–0.64(m,4H).
实施例14:(2-((3,5-二氟-4-((5-异丙基-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基)-5-氟- 5,6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000076
叔丁基(3,5-二氟-4-((5-(丙-1-烯-2-基)-7-((2-(三甲基硅基)乙氧基)甲基)- 7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000077
在氩气保护下,向叔丁基(3,5-二氟-4-((5-碘-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(360毫克,0.582毫摩尔),4,4,5,5-四甲基-2-(丙-1-烯-2-基)-1,3,2-二氧杂环戊烷(195毫克,1.16毫摩尔),碳酸钾(241毫克,1.75毫摩尔)和1,4-二氧六环(4.5毫升)/水(1.5毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(47毫克,0.058毫摩尔)。将该反应液加热至70度搅拌14小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(10毫升×2)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=5:1)分离得叔丁基(3,5-二氟-4-((5-(丙-1-烯-2- 基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(246毫克,收率:79.3%)。MS(ESI):m/z=533.1[M+H] +.
叔丁基(3,5-二氟-4-((5-异丙基-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧 啶-4-基)氧基)苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000078
在氩气保护下,向叔丁基(3,5-二氟-4-((5-(丙-1-烯-2-基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(246毫克,0.461毫摩尔)的乙酸乙酯溶液(8.0毫升)中加入Pd/C(50毫克)。将该反应液用氢气置换三次后,于室温条件下在氢气(15psi)气氛中搅拌反应2小时。LCMS检测反应完成。反应液经过滤,滤液经减压浓缩得叔丁基(3,5-二氟-4-((5-异丙基-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(240毫克,收率:86.5%)。MS(ESI):m/z=535.1[M+H] +.
3,5-二氟-4-((5-异丙基-7-((2-(三甲硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基) 苯胺
Figure PCTCN2021078965-appb-000079
将叔丁基(3,5-二氟-4-((5-异丙基-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(240毫克,0.399毫摩尔)的1,1,1,3,3,3-六氟丙烷-2-醇溶液(5.0毫升)加入到20毫升的密封管中。将该反应液加热至110度,搅拌1小时。反应经LCMS检测,待反应结束后,待反应液冷却至室温。减压浓缩得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=2:1)分离得浅黄色固体3,5-二氟-4-((5-异丙基-7-((2-(三甲硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯胺(137毫克,收率:78.9%)。MS(ESI):m/z=435.1[M+H] +.
1-(3,5-二氟-4-((5-异丙基-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶- 4-基)氧基)苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲
Figure PCTCN2021078965-appb-000080
向配有磁力搅拌器的50毫升圆底烧瓶中加入3,5-二氟-4-((5-异丙基-7-((2-(三甲硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯胺(137毫克,0.315毫摩尔)和三乙胺 (127毫克,1.26毫摩尔)的无水二氯甲烷(3.0毫升)溶液,氩气置换三次,然后将反应液置于冰浴中冷却。然后向反应液中加入三光气(65毫克,0.221毫摩尔),该反应液于0度搅拌20分钟。然后向反应液中加入(3-氟噁丁环-3-基)甲胺(132毫克,1.26毫摩尔),撤掉冰浴,将反应液在于室温搅拌30分钟。LCMS检测反应。减压浓缩后油状物。该油状物经反向C-18硅胶柱(HCOOH)分离得白色固体1-(3,5-二氟-4-((5-异丙基-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲(135毫克,收率:74.0%)。MS(ESI):m/z=566.0[M+H] +.
(2-((3,5-二氟-4-((5-异丙基-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基)-5-氟-5,6-二氢- 4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000081
以1-(3,5-二氟-4-((5-异丙基-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲(135毫克,0.242毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-氟-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(实施例2)的合成方法合成(2-((3,5-二氟-4-((5-异丙基-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基)-5-氟-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(50毫克,收率:47.4%)。MS(ESI):m/z=436.1[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.17(s,1H),7.18(m,2H),7.12(s,1H),4.35–4.25(m,2H),3.74–3.50(m,4H),3.39–3.33(m,1H),1.39(d,J=6.8Hz,6H).
以下化合物采用与实施例14类似的方法,替换相应原料获得。
Figure PCTCN2021078965-appb-000082
实施例17:(2-((3,5-二氟-4-((5-(4-氟-3-甲氧基苯基)-7H-吡咯并[2,3-d]嘧啶-4- 基)氧基)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-恶嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000083
1-(3,5-二氟-4-((5-碘-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯[2,3-d]嘧啶-4- 基)氧基)苯基)-3-((3-甲基氧烷-3-基)甲基)脲
Figure PCTCN2021078965-appb-000084
向配有磁力搅拌器的50毫升圆底烧瓶中加入3,5-二氟-4-((5-碘-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯胺(550毫克,1.06毫摩尔)和三乙胺(429毫克,4.24毫摩尔)的无水二氯甲烷(8.0毫升)溶液,氩气置换三次,然后将反应液置于冰浴中冷却。然后向反应液中加入三光气(220毫克,0.743毫摩尔),该反应液于0度搅拌20分钟。然后向反应液中加入(3-甲基噁丁环-3-基)甲胺(429毫克,4.24毫摩尔),撤掉冰浴,将反应液在于室温搅拌30分钟。LCMS检测反应。减压浓缩后得油状物。该油状物经反向C-18硅胶柱(HCOOH)分离得白色固体1-(3,5-二氟-4-((5-碘-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲基氧烷-3-基)甲基)脲(590毫克,收率86.1%)。MS(ESI):m/z=646.0[M+H] +.
1-(3,5-二氟-4-((5-(4-氟-3-甲氧基苯基)-7-((2-(三甲基硅基)乙氧基)甲基) -7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲氧基-3-基)甲基)脲
Figure PCTCN2021078965-appb-000085
在氩气保护下,向1-(3,5-二氟-4-((5-碘-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲基氧烷-3-基)甲基)脲(70毫克,0.11毫摩尔),(4-氟-3-甲氧基苯基)硼酸(22毫克,0.13毫摩尔),碳酸钾(45毫克,0.33毫摩尔)和1,4-二氧六环(1.2毫升)/水(0.4毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(8.1毫克,0.01毫摩尔)。将该反应液加热至70度搅拌14小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(5毫升×2)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该 粗品经反向C-18硅胶柱(HCOOH)分离得1-(3,5-二氟-4-((5-(4-氟-3-甲氧基苯基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲氧基-3-基)甲基)脲(45毫克,收率:64.4%)。MS(ESI):m/z=644.1[M+H] +.
(2-((3,5-二氟-4-((5-(4-氟-3-甲氧基苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基) 苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-恶嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000086
以1-(3,5-二氟-4-((5-(4-氟-3-甲氧基苯基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲氧基-3-基)甲基)脲(45毫克,0.07毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(实施例1)的合成方法合成(2-((3,5-二氟-4-((5-(4-氟-3-甲氧基苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-恶嗪-5-基)甲醇(21.3毫克,收率:59.4%)。MS(ESI):m/z=514.0[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.27(s,1H),7.57–7.48(m,2H),7.23(m,1H),7.08(m,1H),6.96(d,J=m,2H),4.16(d,J=10.4Hz,1H),3.95(d,J=10.4Hz,1H),3.85(s,3H),3.52(d,J=11.2Hz,1H),3.44(d,J=11.2Hz,1H),3.26(d,J=13.2Hz,1H),3.05(d,J=13.6Hz,1H),1.01(s,3H).
以下化合物采用与实施例17类似的方法,替换相应原料获得。
Figure PCTCN2021078965-appb-000087
Figure PCTCN2021078965-appb-000088
Figure PCTCN2021078965-appb-000089
实施例27:(2-((3,5-二氟-4-(5-(5-异丙基-1,3,4-噁二唑-2-基)-7H-吡咯并[2,3-d]嘧啶 -4-基)氧基)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000090
4-(4-((叔丁氧基羰基)氨基)-2,6-二氟苯氧基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H- 吡咯并[2,3-d]嘧啶-5-羧酸甲酯
Figure PCTCN2021078965-appb-000091
氩气保护下,向叔丁基(3,5-二氟-4-((5-碘-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(2.30克,3.72毫摩尔)的甲醇(40毫升)溶液中,依次加入[1,1'-双(二苯基膦)二茂铁]二氯化钯(271毫克,0.37毫摩尔)、三乙胺(1.13克,11.2毫摩尔)。一氧化碳气体置换三次后,该反应在一氧化碳气氛(55psi)下于室温搅拌13小时。LCMS检测反应。原料转化完全后,减压浓缩溶剂得油状物。该油状物经快速硅胶柱(石油醚:乙酸乙酯=0~25%)分离纯化得4-(4-((叔丁氧基羰基)氨基)-2,6-二氟苯氧基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-5-羧酸甲酯(2.00克,收率:97.7%)。MS(ESI):m/z=551.2[M+H] +.
4-(4-((叔丁氧羰基)氨基)-2,6-二氟苯氧基)-7-((2-(三甲基硅基)乙氧基)甲 基)-7H-吡咯并[2,3-d]嘧啶-5-羧酸
Figure PCTCN2021078965-appb-000092
向4-(4-((叔丁氧基羰基)氨基)-2,6-二氟苯氧基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-5-羧酸甲酯(2.00克,3.63毫摩尔)的四氢呋喃/水(40毫升,V/V=1:1)混合溶剂中,加入一水氢氧化锂(305毫克,7.26毫摩尔),将该反应加热至60度搅拌13小时。反应经LCMS检测。反应结束后,减压浓缩蒸除四氢呋喃,水相用2N稀盐酸调至酸性pH=6,用乙酸乙酯(40毫升×3)萃取。合并的有机相经无水硫酸钠干燥、过滤,滤液减压下蒸除溶剂得4-(4-((叔丁氧羰基)氨基)-2,6-二氟苯氧基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-5-羧酸(1.8克,收率:92.5%)。MS(ESI):m/z=537.2[M+H] +
叔丁基(3,5-二氟-4-((5-(2-异丁基肼-1-羰基)-7-((2-(三甲基硅基)乙氧基)甲 基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000093
在氮气保护下,向4-(4-((叔丁氧羰基)氨基)-2,6-二氟苯氧基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-5-羧酸(1.30克,2.42毫摩尔)的吡啶(10毫升)溶液中依次加入盐酸异丙基肼(272毫克,2.66毫摩尔)和1-(3-二甲基氨丙基)-3-乙基碳二亚胺盐酸盐(696毫克,3.63毫摩尔),然后将反应液置于室温搅拌13小时。LCMS检测反应。待原料转化完全后,减压浓缩得残留物。向该残留物中加入加入甲醇(10毫升)打浆,过滤,滤饼经干燥得目标产物叔丁基(3,5-二氟-4-((5-(2-异丁基肼-1-羰基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(1.06克,收率:70.5%)。MS(ESI):m/z=621.2[M+H] +.
叔丁基(3,5-二氟-4-((5-(5-异丙基-1,3,4-恶二唑-2-基)-7-((2-(三甲基硅基)乙 氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000094
室温下,向化合物叔丁基(3,5-二氟-4-((5-(2-异丁基肼-1-羰基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(1.06克,1.7 毫摩尔)的甲苯溶液(15毫升)中,加入伯吉斯试剂(1.2克,5.1毫摩尔)。将该反应液加热至120度搅拌5小时。LCMS检测反应。待原料转化完全后,溶液经减压浓缩得粗品。该粗品经硅胶柱分离纯化(石油醚:乙酸乙酯=0~30%)得到叔丁基(3,5-二氟-4-((5-(5-异丙基-1,3,4-噁二唑-2-基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(880毫克,产率:85%)。MS(ESI):m/z=603.1[M+H] +
3,5-二氟-4-((5-(5-异丙基-1,3,4-噁二唑-2-基)-7-((2-(三甲基硅基)乙氧基)甲 基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯胺
Figure PCTCN2021078965-appb-000095
将叔丁基(3,5-二氟-4-((5-(5-异丙基-1,3,4-噁二唑-2-基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(800毫克,1.33毫摩尔)溶于1,1,1,3,3,3-六氟丙烷-2-醇溶液(10毫升)中,反应在氮气保护下于100度加热搅拌24小时。TLC检测反应。待反应液冷却至室温后,减压蒸除溶剂得油状物。该油状物经硅胶柱分离纯化(石油醚:乙酸乙酯=0-50%)得3,5-二氟-4-((5-(5-异丙基-1,3,4-噁二唑-2-基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯胺(480毫克,收率:72.0%)。MS(ESI):m/z=503.2[M+H]
1H NMR(400MHz,CDCl 3)δ8.52(s,1H),8.09(s,1H),6.34(d,J=9.2Hz,2H),5.71(s,2H),3.87(s,2H),3.62–3.57(m,2H),3.29–3.21(m,1H),1.40(t,J=6.8Hz,6H),0.97–0.91(m,2H),-0.03(s,9H).
1-(3,5-二氟-4-((5-(5-异丙基-1,3,4-噁二唑-2-基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H- 吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲基氧杂环丁烷-3-基)甲基)脲
Figure PCTCN2021078965-appb-000096
以3,5-二氟-4-((5-(5-异丙基-1,3,4-噁二唑-2-基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯胺(60毫克,0.12毫摩尔)为原料参照化合物1-(3,5-二氟-4-((5-碘-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲基氧烷-3-基)甲基)脲的制备方法合成1-(3,5-二氟-4-((5-(5-异丙基-1,3,4-噁二唑-2-基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲基氧杂环丁烷-3-基)甲基)脲(54毫克,收率:71.8%)MS(ESI):m/z=630.0[M+H] +.
(2-((3,5-二氟-4-(5-(5-异丙基-1,3,4-噁二唑-2-基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基) 苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000097
以1-(3,5-二氟-4-((5-(5-异丙基-1,3,4-噁二唑-2-基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲基氧杂环丁烷-3-基)甲基)脲(54毫克,0.086毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(实施例1)的合成方法合成(2-((3,5-二氟-4-(5-(5-异丙基-1,3,4-噁二唑-2-基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(31毫克,收率:72.4%)。MS(ESI):m/z=500.1[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.38(s,1H),8.18(s,1H),6.98(d,J=10.4Hz,2H),4.17(dd,J=10.4,1.2Hz,1H),3.96(d,J=10.8Hz,1H),3.53(d,J=11.2Hz,1H),3.44(d,J=11.2Hz,1H),3.25(m,2H),3.06(d,J=13.6Hz,1H),1.38(d,J=7.2Hz,6H),1.02(s,3H).
实施例28:(2-((3,5-二氟-4-(5-(5-异丙基-1,3,4-噁二唑-2-基)-7H-吡咯并[2,3-d]嘧啶-4-基) 氧基)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000098
叔丁基(3,5-二氟-4-((5-(3,3,3-三氟丙-1-烯-2-基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H- 吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000099
在氩气保护下,向叔丁基(3,5-二氟-4-((5-碘-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸(A1-3,1.10克,1.78毫摩尔),4,4,6-三甲基-2-(3,3,3-三氟丙-1-烯-2-基)-1,3,2-二氧硼烷(474毫克,2.13毫摩尔),碳酸钾(492毫克,3.56毫摩尔)和1,4-二氧六环(12毫升)/水(4.0毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(87毫克,1.07毫摩尔)。将该反应液加热至90度搅拌14小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(15毫升×3)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗 品经快速硅胶柱(石油醚:乙酸乙酯=4:1)分离得叔丁基(3,5-二氟-4-((5-(3,3,3-三氟丙-1-烯-2-基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(1.02克,收率:89.6%)。MS(ESI):m/z=587.8[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.43(s,1H),7.46(d,J=1.2Hz,1H),7.15(m,2H),6.59(s,1H),6.29(d,J=1.6Hz,1H),6.12(d,J=1.2Hz,1H),5.66(s,2H),3.62–3.56(m,2H),1.52(d,J=7.2Hz,9H),0.97–0.90(m,2H),0.00(s,3H),-0.04(s,6H).
叔丁基(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡 咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯
Figure PCTCN2021078965-appb-000100
在氩气保护下,向配有磁力搅拌器的50毫升圆底烧瓶中加入叔丁基(3,5-二氟-4-((5-(3,3,3-三氟丙-1-烯-2-基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(412毫克,1.43毫摩尔)和无水四氢呋喃(5.0毫升)。开动搅拌器,将反应液用冰盐浴冷却至零度,然后向反应液中滴加NaHMDS(2.0M,1.5毫升,3.0毫摩尔)的四氢呋喃溶液。滴加完毕后,将反应液于零度继续搅拌20分钟,然后向反应液中滴加二苯基(甲基)锍四氟硼酸盐(600毫克,1.02毫摩尔)的无水四氢呋喃(5.0毫升)溶液。待反应于零度搅拌0.5小时后,撤去冰盐浴,将反应置于室温继续搅拌1小时。反应经LCMS检测。加入饱和氯化铵溶液(20毫升)淬灭反应,然后加乙酸乙酯(15毫升×3)萃取。合并的有机相经饱和食盐(20毫升)水洗涤、无水硫酸钠干燥、过滤,滤液经减压浓缩溶剂得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=4:1)分离得叔丁基(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(378毫克,收率:46.1%)。MS(ESI):m/z=601.8[M+H] +.
1H NMR(400MHz,CDCl 3)δ8.40(s,1H),7.35(s,1H),7.14(d,J=9.2Hz,2H),6.62(s,1H),5.61(s,2H),3.60–3.53(m,2H),1.55–1.48(m,9H),1.45(m,2H),1.20(s,2H),0.92(m,2H),-0.05(s,7H).
3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并 [2,3-d]嘧啶-4-基)氧基)苯胺
Figure PCTCN2021078965-appb-000101
将叔丁基(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基甲酸酯(408毫克,0.68毫摩尔)的1,1,1,3,3,3-六氟丙烷-2-醇溶液(8.0毫升)加入到20毫升的密封管中。将该反应液加热至110度,搅拌1小时。反应经LCMS检测,待反应结束后,待反应液冷却至室温。减压浓缩得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=3:2)分离得3,5-二氟-4-((5-(1-(三氟甲基)环丙 基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯胺(190毫克,收率:72.5%)。MS(ESI):m/z=501.7[M+H] +.
1-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并 [2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲基氧杂环丁烷-3-基)甲基)脲
Figure PCTCN2021078965-appb-000102
向配有磁力搅拌器的50毫升圆底烧瓶中加入3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯胺(65毫克,0.13毫摩尔)和三乙胺(53毫克,0.52毫摩尔)的无水二氯甲烷(2.0毫升)溶液,氩气置换三次,然后将反应液置于冰浴中冷却。然后向反应液中加入三光气(27毫克,0.091毫摩尔),该反应液于0度搅拌30分钟。然后向反应液中加入(3-甲基噁丁环-3-基)甲胺(40毫克,0.39毫摩尔),撤掉冰浴,将反应液在于室温搅拌30分钟。LCMS检测反应。减压浓缩后油状物。该油状物经反向C-18硅胶柱(NH 4HCO 3)分离得1-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲基氧杂环丁烷-3-基)甲基)脲(51毫克,收率62.6%)。MS(ESI):m/z=628.9[M+H] +.
(2-((3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基)- 5-甲基-5,6-二氢-4H-1,3-氧嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000103
在0度下,向1-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-甲基氧杂环丁烷-3-基)甲基)脲(51毫克,0.081毫摩尔)的无水二氯甲烷(2.0毫升)中加入三氟乙酸(1.0毫升)。撤去冰浴,该反应液于室温搅拌4小时。减压浓缩溶液的油状残留物,加入无水甲醇(4.0毫升),用冰浴冷却至0度后,向反应液中加入无水碳酸钾调节pH=10-11。开动搅拌器,搅拌30分钟后,过滤,滤液经减压浓缩得粗品。该粗品经反向C-18硅胶柱(NH 4HCO 3)分离得白色固体(2-((3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-氧嗪-5-基)甲醇(28.8毫克,收率:71.3%)。MS(ESI):m/z=498.7[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.23(s,1H),7.48(s,1H),6.97(m,2H),4.19(dd,J=10.6,1.4Hz,1H),3.98(d,J=10.5Hz,1H),3.53(d,J=11.2Hz,1H),3.45(d,J=11.2Hz,1H),3.26(d,J=1.4Hz,1H),3.07(d,J=13.4Hz,1H),1.41(q,J=4.9Hz,2H),1.21(s,2H),1.03(s,3H).
实施例29:N-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯 基)-2,6-二氧杂-8-氮杂螺[3.5]壬-7-烯-7-胺
Figure PCTCN2021078965-appb-000104
1-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯 并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-(羟甲基)氧杂环丁烷-3-基)甲基)硫脲
Figure PCTCN2021078965-appb-000105
在室温下,向配有磁力搅拌器的10毫升圆底烧瓶中加入3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯胺(125毫克,0.25毫摩尔)的无水二氯甲烷(2.0毫升)溶液中加入1,1'-硫羰基双(吡啶-2(1H)-酮)(64毫克,0.27毫摩尔),氩气置换三次,然后将反应液置于40度油浴中搅拌1小时。待原料转化完全后,将反应液冷却至室温,然后依次加入二异丙基乙基胺(312毫克,2.41毫摩尔)和二-3-(氨甲基)氧杂环丁烷-3-基]甲醇草酸酯(121毫克,0.38毫摩尔)。该反应于室温搅拌16小时。反应经LCMS检测。减压浓缩溶剂得油状粗品,经反向C-18硅胶柱(HCOOH)分离得1-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-(羟甲基)氧杂环丁烷-3-基)甲基)硫脲(150毫克,收率:91.0%)。MS(ESI):m/z=660.8[M+H] +.
甲基(Z)-N-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)- 7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-N'-((3-(羟甲基)噁丁环-3-基)甲基)氨基甲酰亚胺基硫 酸酯
Figure PCTCN2021078965-appb-000106
室温中,向1-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((3-(羟甲基)氧杂环丁烷-3-基)甲基)硫脲(150毫克,0.227毫摩尔)和二异丙基乙基胺(88毫克,0.68毫摩尔)的丙酮(3.0毫升)溶液中加入碘甲烷(322毫克,2.27毫摩尔)。将该反应加热至55度搅拌2小时。反应经LCMS检测。过滤除去不溶物,收集滤液,滤液经减压浓缩得粗产物甲基(Z)-N-(3,5-二 氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-N'-((3-(羟甲基)噁丁环-3-基)甲基)氨基甲酰亚胺基硫酸酯(230毫克)。该粗品不经纯化直接用于下一步。MS(ESI):m/z=674.9[M+H] +
N-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基硅基)乙氧基) 甲基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯基)-2,6-二氧杂-8-氮杂螺环[3.5]壬-7-烯-7-胺
Figure PCTCN2021078965-appb-000107
在室温下,向甲基(Z)-N-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-N’-((3-(羟甲基)氧杂环丁烷-3-基)甲基)氨基甲酸酯(230毫克)的四氢呋喃溶液(4.0毫升)混合物中加入氢氧化钠(37毫克,0.91毫摩尔)。将该反应液于室温搅拌16小时,反应结束后,向反应液中加入水(10毫升)然后加入乙酸乙酯(10毫升×3)萃取。合并的有机相经饱和食盐(10毫升)水洗涤、无水硫酸钠干燥、过滤,滤液经减压浓缩溶剂得粗品,该粗品经C-18反向键合硅胶柱(NH 4HCO 3)分离得N-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯基)-2,6-二氧杂-8-氮杂螺环[3.5]壬-7-烯-7-胺(80毫克,总收率:56.3%)。MS(ESI):m/z=626.9[M+H] +.
N-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-2,6-二氧 杂-8-氮杂螺[3.5]壬-7-烯-7-胺
Figure PCTCN2021078965-appb-000108
以N-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯基)-2,6-二氧杂-8-氮杂螺环[3.5]壬-7-烯-7-胺(80毫克,0.13毫摩尔)为原料参考制备化合物(2-((3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-氧嗪-5-基)甲醇的方法合成N-(3,5-二氟-4-((5-(1-(三氟甲基)环丙基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)-2,6-二氧杂-8-氮杂螺[3.5]壬-7-烯-7-胺(34.5毫克,收率:54.4%)。MS(ESI):m/z=496.6[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.27(s,1H),7.56(s,1H),7.26(m,2H),4.92(s,2H),4.66(s,4H),3.81(s,2H),1.44(m,2H),1.25(m,2H).
以下化合物采用与实施例29类似的方法,替换相应原料获得。
Figure PCTCN2021078965-appb-000109
Figure PCTCN2021078965-appb-000110
实施例32:(4-(4-(4-((5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-基)氨基)-2,6-二氟苯氧基)-7H-吡咯 并[2,3-d]嘧啶-5-基)苯基)(2-氧杂-6-氮杂螺[3.3]庚-6-基)甲酮
Figure PCTCN2021078965-appb-000111
1-(3,5-二氟-4-((5-碘-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基) 氧基)苯基)-3-((1-(羟甲基)环丙基)甲基)硫脲
Figure PCTCN2021078965-appb-000112
在室温下,向配有磁力搅拌器的25毫升圆底烧瓶中加入3,5-二氟-4-((5-碘-7-((2-(三甲基硅基)乙氧基)甲基)-7H-吡咯[2,3-d]嘧啶-4-基)氧基)苯胺(800毫克,1.54毫摩尔)的无水二氯甲烷(10毫升)溶液中加入1,1'-硫羰基双(吡啶-2(1H)-酮)(394毫克,1.70毫摩尔),氩气置换三次,然后将反应液置于40度油浴中搅拌1小时。待原料转化完全后,将反应液冷却至室温,然后加入(1-(氨基甲基)环丙基)甲醇(313毫克,3.09毫摩尔)。该反应于室温搅拌1小时。反应经LCMS检测。减压浓缩溶剂得油状粗品,经反向C-18硅胶柱(NH 4HCO 3)分离得1-(3,5-二氟-4-((5-碘-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((1-(羟甲基)环丙基)甲基)硫脲(920毫克,收率:90.1%)。MS(ESI):m/z=662.1[M+H] +.
N-(3,5-二氟-4-((3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯[2,3-b]吡啶-4-基)氧 基)苯基)-5-氧杂-7-氮杂螺环[2.5]辛-6-烯-6-胺
Figure PCTCN2021078965-appb-000113
向配有磁力搅拌器的50毫升圆底烧瓶中依次加入1-(3,5-二氟-4-((5-碘-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-3-((1-(羟甲基)环丙基)甲基)硫脲(920毫克,1.39毫摩尔)和三乙胺(422毫克,4.17毫摩尔)和无水二氯甲烷(15毫升)。氩气置换三次后,将反应液置于冰-水浴中冷却10分钟,然后向反应液中加入3-(((乙基亚氨基)亚甲基)氨基)-N,N-二甲基丙烷-1-胺盐酸(533毫克,2.78毫摩尔)。撤去冰浴,该反应于室温搅拌24小时。LCMS检测反应。减压浓缩溶剂得油状粗品,经反向C-18硅胶柱(HCOOH)分离得N-(3,5-二氟-4-((3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯[2,3-b]吡啶-4-基)氧基)苯基)-5-氧杂-7-氮杂螺环[2.5]辛-6-烯-6-胺(520毫克,收率:59.6%)。MS(ESI):m/z=628.1[M+H] +.
(4-(4-(4-((5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-基)氨基)-2,6-二氟苯氧基)-7-(2-(三甲基甲硅 烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-5-基)苯基)(2-氧杂-6-氮杂螺[3.3]庚-6-基)甲酮
Figure PCTCN2021078965-appb-000114
在氩气保护下,向N-(3,5-二氟-4-((3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯[2,3-b]吡啶-4-基)氧基)苯基)-5-氧杂-7-氮杂螺环[2.5]辛-6-烯-6-胺(80毫克,0.127毫摩尔),(2-氧杂-6-氮杂螺环[3.3]庚-6-基)(4-(4,4,5,5-四甲基-1,3,2-二氧六环-2-基)苯基)甲酮(45毫克,0.14毫摩尔),磷酸钾(54毫克,0.254毫摩尔)和1,4-二氧六环(1.0毫升)/水(0.3毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(10.6毫克,0.013毫摩尔)。将该反应液加热至90度搅拌3小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(5毫升×2)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗品经反向C-18硅胶柱(NH 4HCO 3)分离得(4-(4-(4-((5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-基)氨基)-2,6-二氟苯氧基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-5-基)苯基)(2-氧杂-6-氮杂螺[3.3]庚-6-基)甲酮(32毫克,收率:35.7%)。MS(ESI):m/z=703.3[M+H] +.
(4-(4-(4-((5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-基)氨基)-2,6-二氟苯氧基)-7H-吡咯并[2,3-d]嘧 啶-5-基)苯基)(2-氧杂-6-氮杂螺[3.3]庚-6-基)甲酮
Figure PCTCN2021078965-appb-000115
以(4-(4-(4-((5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-基)氨基)-2,6-二氟苯氧基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-5-基)苯基)(2-氧杂-6-氮杂螺[3.3]庚-6-基)甲酮(32毫克,0.053毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(实施例1)的合成方法合成(4-(4-(4-((5-氧杂-7-氮杂螺[2.5]辛-6-烯-6-基)氨基)-2,6-二氟苯氧基)-7H-吡咯并[2,3-d]嘧啶-5-基)苯基)(2-氧杂-6-氮杂螺[3.3]庚-6-基)甲酮(7.0毫克,收率:26%)。MS(ESI):m/z=573.8[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.32(s,1H),7.89(d,J=8.4Hz,2H),7.73–7.67(m,3H),7.05–6.97(m,2H),4.62(s,2H),4.36(s,2H),4.09(s,2H),3.26(s,2H),0.72(m,4H).
以下化合物采用与实施例32类似的方法,替换相应原料获得。
Figure PCTCN2021078965-appb-000116
实施例37:4-(2,6-二氟-4-((5-氟-5-(羟甲基)-5,6-二氢-4H-1,3-噁嗪-2-基)氨基)苯氧 基)-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000117
4-(4-氨基-2,6-二氟苯氧基)-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯 并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000118
在氩气保护下,向4-(4-氨基-2,6-二氟苯氧基)-3-溴-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(63毫克,0.13毫摩尔),(4-氟苯基)硼酸(22毫克,0.15毫摩尔),磷酸钾(54毫克,0.25毫摩尔)和1,4-二氧六环(1.5毫升)/水(0.5毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(10毫克,0.012毫摩尔)。将该反应液加热至90度搅拌3小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(5毫升×3)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=2:1)分离得4-(4-氨基-2,6-二氟苯氧基)-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(51毫克,收率:78.5%)。MS(ESI):m/z=511.1[M+H] +.
1-(4-((5-氰基-3-(4-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶- 4-基)氧基)-3,5-二氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲
Figure PCTCN2021078965-appb-000119
向配有磁力搅拌器的10毫升圆底烧瓶中加入4-(4-氨基-2,6-二氟苯氧基)-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(51毫克,0.10毫摩尔)和三乙胺(40毫克,0.40毫摩尔)的无水二氯甲烷(1.5毫升)溶液,氩气置换三次,然后将反应液置于冰浴中冷却。然后向反应液中加入三光气(21毫克,0.07毫摩尔),该反应液于0度搅拌20分钟。然后向反应液中加入(3-氟噁丁环-3-基)甲胺(21毫克,0.30毫摩尔),撤掉冰浴,将反应液在于室温搅拌30分钟。LCMS检测反应。减压浓缩后油 状物。该油状物经反向C-18硅胶柱(HCOOH)分离得1-(4-((5-氰基-3-(4-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3,5-二氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲(32毫克,收率:50%)。MS(ESI):m/z=642.2[M+H] +.
4-(2,6-二氟-4-((5-氟-5-(羟甲基)-5,6-二氢-4H-1,3-恶嗪-2-基)氨基)苯氧基)-3-(4-氟苯基)- 1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000120
以1-(4-((5-氰基-3-(4-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3,5-二氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲(32毫克,0.05毫摩尔)为原料参考制备(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-氟-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(实施例2)的方法合成4-(2,6-二氟-4-((5-氟-5-(羟甲基)-5,6-二氢-4H-1,3-噁嗪-2-基)氨基)苯氧基)-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡啶-5-腈(15毫克,收率:59%)。MS(ESI):m/z=512.1[M+H] +.
1H NMR(400MHz,MeOD)δ8.39(s,1H),7.56–7.49(m,3H),7.20–7.09(m,2H),7.05(t,J=8.8Hz,2H),4.35–4.21(m,2H),3.79–3.56(m,3H),3.51(d,J=14.2Hz,1H).
以下化合物采用与实施例37类似的方法,替换相应原料获得。
Figure PCTCN2021078965-appb-000121
Figure PCTCN2021078965-appb-000122
实施例43:4-(2-氟-4-((5-氟-5-(羟甲基)-5,6-二氢-4H-1,3-氧嗪-2-基)氨基)苯氧基)-3-(三氟 甲基)-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000123
1-(4-((5-氰基-3-(三氟甲基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡 啶-4-基)氧基)-3-氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲
Figure PCTCN2021078965-appb-000124
以4-(4-氨基-2-氟苯氧基)-3-(三氟甲基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(100毫克,0.21毫摩尔)为原料参照化合物1-(4-((5-氰基-3-(4-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3,5-二氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲的制备方法合成1-(4-((5-氰基-3-(三氟甲基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲(98毫克,收率:76.5%)。MS(ESI):m/z=598.0[M+H] +.
4-(2-氟-4-((5-氟-5-(羟甲基)-5,6-二氢-4H-1,3-氧嗪-2-基)氨基)苯氧基)-3-(三氟甲基)-1H- 吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000125
以1-(4-((5-氰基-3-(三氟甲基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲(98毫克,0.16毫摩尔)为原料参考制备4-(2,6-二氟-4-((5-氟-5-(羟甲基)-5,6-二氢-4H-1,3-恶嗪-2-基)氨基)苯氧基)-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡啶-5-腈的方法合成4-(2-氟-4-((5-氟-5-(羟甲基)-5,6-二氢-4H-1,3-氧嗪-2-基)氨基)苯氧基)-3-(三氟甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(58.9毫克,收率:76.8%)。MS(ESI):m/z=468.0[M+H] +.
1H NMR(400MHz,MeOD)δ8.52(s,1H),8.00(d,J=1.2Hz,1H),7.59–7.45(m,1H),7.08(m,2H),4.36–4.26(m,2H),3.79–3.66(m,2H),3.65–3.57(m,1H),3.51(m,2H).
实施例44:(2-((4-((5-氯-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)氨 基)-5-氟-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000126
4-((5-氯-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基) 氧基)-3-氟苯胺
Figure PCTCN2021078965-appb-000127
在氩气保护下,向4-((5-氯-3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯[2,3-b]吡啶-4-基)氧基)-3-氟苯胺(80毫克,0.15毫摩尔),(4-氟苯基)硼酸(21毫克,0.15毫摩尔),碳酸钾(42毫克,0.30毫摩尔)和1,4-二氧六环(1.5毫升)/水(0.5毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(12毫克,0.015毫摩尔)。将该反应液加热至70度搅拌3小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(5毫升×3)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=3:1)分离得4-((5-氯-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯胺(66毫克,收率:87.7%)。MS(ESI):m/z=502.1,504.1.[M+H] +.
1-(4-((5-氯-3-(4-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶- 4-基)氧基)-3-氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲
Figure PCTCN2021078965-appb-000128
向配有磁力搅拌器的10毫升圆底烧瓶中加入4-((5-氯-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯胺(66毫克,0.13毫摩尔)和三乙胺(53毫克,0.53毫摩尔)的无水二氯甲烷(1.5毫升)溶液,氩气置换三次,然 后将反应液置于冰浴中冷却。然后向反应液中加入三光气(28毫克,0.09毫摩尔),该反应液于0度搅拌20分钟。然后向反应液中加入(3-氟噁丁环-3-基)甲胺(41毫克,0.39毫摩尔),撤掉冰浴,将反应液在于室温搅拌30分钟。LCMS检测反应。减压浓缩后油状物。该油状物经反向C-18硅胶柱(NH 4HCO 3)分离得1-(4-((5-氯-3-(4-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲(45毫克,收率:71%)。MS(ESI):m/z=633.8[M+H] +.
(2-((4-((5-氯-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)氨基)-5-氟-5, 6-二氢-4H-1,3-噁嗪-5-基)甲醇
Figure PCTCN2021078965-appb-000129
以1-(4-((5-氯-3-(4-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲(45毫克,0.071毫摩尔)为原料参考制备4-(2,6-二氟-4-((5-氟-5-(羟甲基)-5,6-二氢-4H-1,3-恶嗪-2-基)氨基)苯氧基)-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡啶-5-腈的方法合成(2-((4-((5-氯-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)氨基)-5-氟-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(21毫克,收率:58.8%)。MS(ESI):m/z=503.0[M+H] +.
1H NMR(400MHz,MeOD)δ8.31(d,J=7.2Hz,1H),7.44(s,1H),7.38(dd,J=8.4,5.6Hz,2H),7.11(d,J=12.8Hz,1H),6.92(t,J=8.8Hz,2H),6.66(d,J=8.4Hz,1H),6.18(t,J=9.12Hz,1H),4.32–4.16(m,2H),3.75–3.57(m,2H),3.56–3.37(m,2H).
实施例45:4-(2-氟-4-((5-(羟甲基)-5-甲基-5,6-二氢-4H-1,3-氧嗪-2-基)氨基)苯氧基)-3- (2-氟-4-甲氧基苯基)-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000130
4-(4-氨基-2-氟苯氧基)-3-(2-氟-4-甲氧基苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H- 吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000131
在氩气保护下,向4-(4-氨基-2-氟苯氧基)-3-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(80毫克,0.167毫摩尔),(2-氟-4-甲氧苯基)硼酸(34毫克,0.20毫摩尔),碳酸钾(46毫克,0.34毫摩尔)和1,4-二氧六环(1.2毫升)/水(0.4毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(13毫克,0.016毫摩尔)。将该反应液加热至100度搅拌4小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(5毫升×3)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=2:1)分离得4-(4-氨基-2-氟苯氧基)-3-(2-氟-4-甲氧基苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(74毫克,收率:84.5%)。MS(ESI):m/z=523.1.[M+H] +.
1-(4-((5-氰基-3-(2-氟-4-甲氧基苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并 [2,3-b]吡啶-4-基)氧基)-3-氟苯基)-3-((3-甲基氧杂环丁烷-3-基)甲基)脲
Figure PCTCN2021078965-appb-000132
以4-(4-氨基-2-氟苯氧基)-3-(2-氟-4-甲氧基苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(37毫克,0.071毫摩尔)为原料参考化合物1-(4-((5-氯-3-(4-氟苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)-3-(3-氟氧杂环丁烷-3-基)甲基)脲的制备方法合成1-(4-((5-氰基-3-(2-氟-4-甲氧基苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)-3-((3-甲基氧杂环丁烷-3-基)甲基)脲(28毫克,收率:61%)。MS(ESI):m/z=650.2[M+H] +.
4-(2-氟-4-((5-(羟甲基)-5-甲基-5,6-二氢-4H-1,3-恶嗪-2-基)氨基)苯氧基)-3-(2-氟-4-甲 氧基苯基)-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000133
以1-(4-((5-氰基-3-(2-氟-4-甲氧基苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-4-基)氧基)-3-氟苯基)-3-((3-甲基氧杂环丁烷-3-基)甲基)脲(28毫克,0.043毫摩尔)为原料参考制备4-(2,6-二氟-4-((5-氟-5-(羟甲基)-5,6-二氢-4H-1,3-恶嗪-2-基)氨基)苯氧基)-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡啶-5-腈的方法合成4-(2-氟-4-((5-(羟甲基)-5-甲基-5,6-二氢-4H-1,3-恶嗪-2-基)氨基)苯氧基)-3-(2-氟-4-甲氧基苯基)-1H-吡咯并[2,3-b]吡啶-5-腈(15.4毫克,收率:69%)。MS(ESI):m/z=520.7[M+H] +.
1H NMR(400MHz,MeOD)δ8.44(s,1H),7.43(s,1H),7.19(t,J=8.4Hz,1H),7.04–6.94(m,1H),6.63(m,4H),4.13(d,J=10.6Hz,1H),3.91(d,J=10.4Hz,1H),3.78(s,3H),3.50(d,J=11.2Hz,1H),3.41(d,J=11.2Hz,1H),3.20(d,J=13.8Hz,1H),3.00(d,J=13.8Hz,1H),1.00(s,3H).
以下化合物采用与实施例45类似的方法,替换相应原料获得。
Figure PCTCN2021078965-appb-000134
实施例50:N-(3,5-二氟-4-(5-(4-氟苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-1-甲 基哌啶-3-甲酰胺
Figure PCTCN2021078965-appb-000135
N-(3,5-二氟-4-((5-(4-氟苯基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d] 嘧啶-4-基)氧基)苯基)-1-甲基哌啶-3-甲酰胺
Figure PCTCN2021078965-appb-000136
在室温条件下,向化合物1-甲基哌啶-3-羧酸(220毫克,1.22毫摩尔)和三乙胺(248毫克,2.45毫摩尔)的N,N-二甲基甲酰胺(5.0毫升)溶液中加入2-(7-偶氮苯并三氮唑)-N,N,N,N-四甲基脲六氟磷酸酯(931毫克,2.45毫摩尔),反应于室温搅拌小时。然后加入3,5-二氟-4-((5-(4-氟苯基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯胺(596毫克,1.22毫摩尔)。反应加热至50度搅拌2小时。反应经LCMS检测。待反应结束后,将反应应用倒入水(30毫升),用乙酸乙酯(10毫升×3)萃取。合并的有机相依次用水(10毫升×3)、饱和食盐水洗涤(10毫升),无水硫酸钠干燥、过滤,滤液经减压浓缩得粗品。该粗品经硅胶柱分离纯化(二氯甲烷/甲醇=0~20%)得N-(3,5-二氟-4-((5-(4-氟苯基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-1-甲基哌啶-3-甲酰胺(200毫克,收率:27%)。MS(ESI):m/z=612.1[M+H] +.
N-(3,5-二氟-4-(5-(4-氟苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-1-甲基哌啶-3-甲 酰胺(0112-189-P1)
Figure PCTCN2021078965-appb-000137
在零度下,向化合物N-(3,5-二氟-4-((5-(4-氟苯基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-1-甲基哌啶-3-甲酰胺(50毫克,0.082毫摩尔)的二氯甲烷(2.0毫升)溶液中加入三氟乙酸(2.0毫升)。撤去冰浴,该反应液于室温搅拌4小时。减压浓缩溶液的油状残留物,加入无水甲醇(3.0毫升),用冰浴冷却至0度后,向反应液中加入无水碳酸钾调节pH=10-11。开动搅拌器,搅拌30分钟后,过滤,滤液经减压浓缩得粗品。该粗品经反向C-18硅胶柱(NH 4HCO 3)分离得N-(3,5-二氟-4-(5-(4-氟苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧基)苯基)-1-甲基哌啶-3-甲酰胺(26.5毫克,收率:67.1%)。MS(ESI):m/z=482.1[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ12.59(s,1H),10.38(s,1H),8.27(s,1H),8.15(s,1H),7.98–7.61(m,4H),7.42(m,2H),7.16(m,2H),2.87(m,1H),2.73(m,1H),2.57(s,1H),2.21(m,3H),2.09(m,1H),1.93(m,1H),1.79(s,1H),1.67(m,1H),1.40(m,3H).
以下化合物采用与实施例50类似的方法,替换相应原料获得。
Figure PCTCN2021078965-appb-000138
Figure PCTCN2021078965-appb-000139
实施例53:4-(2-氟-4-((四氢-2H-吡喃-4-基)氨基)苯氧基)-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡 啶-5-腈
Figure PCTCN2021078965-appb-000140
4-(4-氨基-2-氟苯氧基)-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2, 3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000141
在氩气保护下,向4-(4-氨基-2-氟苯氧基)-3-溴-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(380毫克,0.80毫摩尔),(4-氟苯基)硼酸(134毫克,0.96毫摩尔),磷酸钾(389毫克,1.60毫摩尔)和1,4-二氧六环(6.0毫升)/水(1.5毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(64毫克,0.079毫摩尔)。将该反应液加热至90度搅拌3小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(10毫升×3)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=2:1)分离得4-(4-氨基-2-氟苯氧基)-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(390毫克,收率:99%)。MS(ESI):m/z=493.1[M+H] +.
4-(2-氟-4-((四氢-2H-吡喃-4-基)氨基)苯氧基)-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧 基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000142
在氩气保护下,向4-(4-氨基-2-氟苯氧基)-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(50毫克,0.10毫摩尔)的1,2-二氯乙烷(1.0毫升)的溶液中滴加氯化锌的无水四氢呋喃溶液(1.0M,0.2毫升,0.2毫摩尔)。将反应液至于70度油浴中搅拌2小时。待反应液冷却至室温后,再加入氰基硼氢化钠(16毫克,0.30毫摩尔)。然后将反应液加热至70度继续搅拌12小时。反应经LCMS检测。待反应液冷却至室温后,加饱和氯化铵水溶液淬灭反应(2.0毫升),加二氯甲烷(5毫升×3)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=3:1)分离得标题化合物(45毫克,收率:76.8%)。MS(ESI):m/z=577.8[M+H] +.
4-(2-氟-4-((四氢-2H-吡喃-4-基)氨基)苯氧基)-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡啶-5-腈
Figure PCTCN2021078965-appb-000143
以4-(2-氟-4-((四氢-2H-吡喃-4-基)氨基)苯氧基)-3-(4-氟苯基)-1-(2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡咯并[2,3-b]吡啶-5-腈(45毫克,0.078毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇(实施例1)的合成方法合成4-(2-氟-4-((四氢-2H-吡喃-4-基)氨基)苯氧基)-3-(4-氟苯基)-1H-吡咯并[2,3-b]吡啶-5-腈(18.5毫克,收率:53.1%)。MS(ESI):m/z=447.6[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.40(s,1H),7.50–7.45(m,2H),7.03–6.96(m,2H),6.65(t,J=9.2Hz,1H),6.38(dd,J=13.6,2.8Hz,1H),6.28(m,1H),3.95(dt,J=11.6,3.6Hz,2H),3.51(td,J=11.6,2.2Hz,2H),3.40(s,1H),1.99–1.90(m,2H),1.50–1.38(m,2H).
以下化合物采用与实施例53类似的方法,替换相应原料获得。
Figure PCTCN2021078965-appb-000144
Figure PCTCN2021078965-appb-000145
(注:化合物57和化合物58为异构体,其绝对构型未确定,化合物57在HPLC分 离时先出峰)
以下化合物采用与实施例32类似的方法,替换相应原料获得。
Figure PCTCN2021078965-appb-000146
实施例62:4-(5-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-1H-吡唑并[3,4-b]吡啶-3-基)-N,N- 二甲基苯酰胺
Figure PCTCN2021078965-appb-000147
4-(5-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡 唑并[3,4-b]吡啶-3-基)-N,N-二甲基苯酰胺
Figure PCTCN2021078965-appb-000148
在氩气保护下,向5-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-b]吡啶(100毫克,0.173毫摩尔),(4-(二甲基氨基甲酰)苯基)硼酸(22毫克,0.21毫摩尔),磷酸钾(74毫克,0.35毫摩尔)和1,4-二氧六环(1.5毫升)/水(0.5毫升)的混合物中加入[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(14毫克,0.017毫摩尔)。将该反应液加热至90度搅拌3小时。反应经LCMS检测。待反应液冷却至室温后,加乙酸乙酯(5毫升×3)萃取。合并的有机相经无水硫酸钠干燥,过滤,滤液经减压浓缩溶剂得粗品,该粗品经快速硅胶柱(石油醚:乙酸乙酯=2:1)分离得4-(5-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-b]吡啶-3-基)-N,N-二甲基苯酰胺(32毫克,收率:30.9%)。MS(ESI):m/z=599.3[M+H] +.
4-(5-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-1H-吡唑并[3,4-b]吡啶-3-基)-N,N-二甲基苯酰
Figure PCTCN2021078965-appb-000149
以(4-(5-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-b]吡啶-3-基)-N,N-二甲基苯酰胺(32毫克,0.053毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇的合成方法合成4-(5-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-1H-吡唑并[3,4-b]吡啶-3-基)-N,N-二甲基苯酰胺(12毫克,收率:48%)。MS(ESI):m/z=469.3[M+H] +.
1H NMR(400MHz,CD 3OD)δ8.75(d,J=2.0Hz,1H),8.57(d,J=2.0Hz,1H),8.13(d,J=8.0Hz,1H),8.06(d,J=1.6Hz,1H),7.64(d,J=7.6Hz,1H),7.49(d,J=7.6Hz,1H),7.32(s,2H),3.21–3.16(m,4H),3.15(s,3H),3.08(s,3H),2.65–2.57(m,4H),2.41(s,6H),2.37(s,3H).
实施例63:4-(3-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-7H-吡咯并[2,3-c]哒嗪-5-基)- N-(3-甲氧基丙基)-N-甲基苯甲酰胺
Figure PCTCN2021078965-appb-000150
4-(3-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡 咯并[2,3-c]哒嗪-5-基)-N-(3-甲氧基丙基)-N-甲基苯甲酰胺
Figure PCTCN2021078965-appb-000151
以5-溴-3-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-7-((2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-c]哒嗪(120毫克,0.23毫摩尔)和N-(3-甲氧基丙基)-N-甲基-4-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)苯酰胺(90毫克,0.27毫摩尔)为原料参考化合物4-(5-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-b]吡啶-3-基)-N,N-二甲基苯酰胺的制备方法合成4-(3-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-7-(2-(三甲基甲硅烷基)乙氧基)甲基)-7H-吡咯并[2,3-c]哒嗪-5-基)-N-(3-甲氧基丙基)-N-甲基苯甲酰胺(20毫克,收率:17%)。MS(ESI):m/z=657.4[M+H] +.
4-(3-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-7H-吡咯并[2,3-c]哒嗪-5-基)-N-(3-甲氧基 丙基)-N-甲基苯甲酰胺
Figure PCTCN2021078965-appb-000152
以4-(5-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑并[3,4-b]吡啶-3-基)-N,N-二甲基苯酰胺(7.0毫克,0.011毫摩尔)为原料参考(2-((3,5-二氟-4-((5-(2-氟-4-甲氧苯基)-7H-吡咯并[2,3-d]嘧啶-4-基)氧代)苯基)氨基)-5-甲基-5,6-二氢-4H-1,3-噁嗪-5-基)甲醇的合成方法合成4-(3-(3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)-7H-吡咯并[2,3-c]哒嗪-5-基)-N-(3-甲氧基丙基)-N-甲基苯甲酰胺(2.2毫克,收率:39%)。MS(ESI):m/z=527.1[M+H] +.
1H NMR(400MHz,CD 3OD)δ11.52(s,1H),8.28(s,1H),7.95(s,1H),7.75–7.67(m,4H),7.55(d,J=7.4Hz,2H),3.66–3.48(m,4H),3.39–3.18(m,7H),3.10(s,3H),2.68(s,3H),2.46(s,6H),2.06–1.76(m,5H).
生物测试例1 HPK1 ADP-Glo酶学活性测试
制备酶活测试缓冲液包含40mM Tris,pH7.5;20mM毫克Cl2;0.1毫克/毫升BSA;50μM DTT。化合物溶于100%DMSO,母液浓度为10mM。化合物的DMSO溶液从100uM起,连续做三倍梯度稀释,共十一个浓度,将稀释后的化合物用酶活测试缓冲液以1:20稀释好后,取1uL加入工作孔,每个浓度两复孔。阴性对照孔和阳性对照孔加入1:20稀释的DMSO溶液1uL。制备2.5×底物/ATP工作液为包含0.25ug/uL MBP蛋白和45uM ATP的酶活测试缓冲液,每个工作孔中加入2uL 2.5×底物/ATP工作液。制备2.5×酶反应工作液为包含0.5ng/uL HPK1重组蛋白(Signalchem,货号M23-11G-10)的酶活测试缓冲液,每个工作孔中加入2uL 2.5×酶反应工作液,阴性对照孔仅加入2uL酶活测试缓冲液。贴好封板膜简单离心后置于室温反应30分钟,反 应结束后每孔加入5uL ADP‐Glo试剂(Promega,货号V1901)室温反应40分钟,随后加入10uL激酶检测试剂(Promega,货号V1901),室温反应20分钟后测定最终发光信号。
分别计算阳性孔和阴性孔的平均值,作为阳性对照值(Signal pos)和阴性对照值(Signal neg)。将工作孔信号值(Signal test)按公式Inhibition rate=(Signal pos-Signal test)/(Signal pos–Signal neg)×100%计算抑制率。求得的抑制率在GraphPad Prism软件中按非线性拟合绘制浓度-抑制率曲线,计算IC 50
生物测试例2 GLK ADP-Glo酶学活性测试
制备酶活测试缓冲液包含40mM Tris,pH7.5;20mM毫克Cl2;0.1毫克/毫升BSA;50μM DTT。化合物溶于100%DMSO,母液浓度为10mM。化合物的DMSO溶液从1mM起,连续做三倍梯度稀释,共十一个浓度,将稀释后的化合物用酶活测试缓冲液以1:20稀释好后,取1uL加入工作孔,每个浓度两复孔。阴性对照孔和阳性对照孔加入1:20稀释的DMSO溶液1uL。制备2.5×底物/ATP工作液为包含0.5ug/uL PKA底物多肽和105uM ATP的酶活测试缓冲液,每个工作孔中加入2uL 2.5×底物/ATP工作液。制备2.5×酶反应工作液为包含2.5ng/uL GLK重组蛋白(Signalchem,货号M25-11G-10)的酶活测试缓冲液,每个工作孔中加入2uL 2.5×酶反应工作液,阴性对照孔仅加入2uL酶活测试缓冲液。贴好封板膜简单离心后置于室温反应1小时,反应结束后每孔加入5uL ADP‐Glo试剂(Promega,货号V1901)室温反应40分钟,随后加入10uL激酶检测试剂(Promega,货号V1901),室温反应20分钟后测定最终发光信号。
分别计算阳性孔和阴性孔的平均值,作为阳性对照值(Signal pos)和阴性对照值(Signal neg)。将工作孔信号值(Signal test)按公式Inhibition rate=(Signal pos-Signal test)/(Signal pos–Signal neg)×100%计算抑制率。求得的抑制率在GraphPad Prism软件中按非线性拟合绘制浓度-抑制率曲线,计算IC 50。实验结果见下表:
表1 HPK1、GLK酶活实验结果
Figure PCTCN2021078965-appb-000153
Figure PCTCN2021078965-appb-000154
其中,A表示IC 50值≤50nM;B表示50nM<IC 50值≤500nM;C表示500nM<IC 50值≤00nMD。
生物测试例3 SLP76磷酸化细胞学测试
抑制HPK1可以抑制其下游SLP76的磷酸化。SLP76蛋白的磷酸化使用Jurkat(ATCC,Clone E6-1
Figure PCTCN2021078965-appb-000155
TIB-152 TM)细胞进行测试,实验的第一天将细胞用培养基 (RPMI 1640+0.5%FBS)稀释到10 6/毫升,按每孔100uL,10 5细胞的量铺在96孔细胞培养板中,饥饿4小时培养。化合物溶于100%DMSO,母液浓度为4mM。化合物的DMSO溶液从10mM起,连续做四倍梯度稀释,共9个浓度,将4uL稀释后的化合物稀释到196uL 37度预热的RPMI 1640并混匀。取50uL最终稀释后的化合物加入细胞中,37度孵育20分钟,加入50uL稀释后的人CD3/CD28T细胞激活剂(Stemcell,货号:10971),使激活剂的终浓度体积为总体系的1/40,37度孵育30分钟。反应结束后将细胞置于4度离心机1200rpm离心5分钟,吸走培养基,加入150uL细胞裂解液(ELISA kit中提供,Cell Signaling,货号30794C)在冰上放置30分钟使细胞充分裂解,裂解后的细胞吹打均匀后置于4度离心机以4000rpm离心5分钟,随后取50uL上清加入FastScan TM Phospho-SLP-76(Ser376)ELISA Kit(Cell Signaling,货号30794C)测试细胞SLP76的磷酸化水平。
分别计算阳性孔和阴性孔的平均值,作为阳性对照值(Signal pos)和阴性对照值(Signal neg)。将工作孔信号值(Signal test)按公式Inhibition rate=(Signal pos-Signal test)/(Signal pos–Signal neg)×100%计算抑制率。求得的抑制率在GraphPad Prism软件中按非线性拟合绘制浓度-抑制率曲线,计算IC 50
表2 SLP76磷酸化抑制实验结果
Figure PCTCN2021078965-appb-000156
Figure PCTCN2021078965-appb-000157
其中,A表示IC 50值≤1000nM;B表示1000nM<IC 50值≤20000nM。
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、光学异构体或水合物;
    Figure PCTCN2021078965-appb-100001
    其中,
    X和Y各自独立地选自下组:无、NR、S、O、-NR-C(=O)R-、-NR-C(=O)C(=O)NR-、-NR-C(=O)NR-、-NR-C(=S)NR-、-NR-C(=O)NRCH 2-、-NR-C(=S)NRCH 2-,其中,所述的R选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C3-C8环烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基;
    M 1、M 2或M 3各自独立地为CH或N;且当所述的M 1或M 2为C时,所述的
    Figure PCTCN2021078965-appb-100002
    可以位于所述的M 1或M 2上;
    A环选自下组:C3-C8环烷基、5-12元的杂环基、6-10元芳环,或具有1-3个选自N、S和O的杂原子的5-10元的杂芳环,或A为H;
    m为1、2、3或4;
    n为0、1、2、3、4或5;
    p为0、1或2;
    Ra选自下组:卤素、CN、取代或未取代的C1-C6烷基、或
    Figure PCTCN2021078965-appb-100003
    其中,所述的环B为C3-C8环烷基、6-10元芳基、5-10元的杂芳基、具有1-3个选自下组N、S和O的杂原子的3-10元杂环基;
    各个R 1、R 2、R 3和R 4各自独立地选自下组:H、卤素、OH、CN、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C3-C8环烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基、取代或未取代的C6-C10芳基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-10元杂芳基、R 7-C(=O)-、R 7-C(=O)具有1-3个选自下组N、S和O的杂原子的3-12元杂环基-、R 8-S(=O) 2-、R 9R 10N-C(=O)-、R 9R 10N-C(=O)NR-、R 11R 12N-S(=O) 2-、R 7O-、R 8-S(O) 2NR-;
    各个R 7、R 8、R 9、R 10、R 11、R 12各自独立地选自下组:H、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C3-C8环烷基、取代或未取代的具 有1-3个选自下组N、S和O的杂原子的3-12元杂环基;或所述的R 9和R 10或R 11和R 12与其相连的氮原子共同构成取代或未取代的具有1-3个选自N、S和O的杂原子的3-12元杂环基;
    除非特别说明,所述的“取代”是指被选自下组的一个或多个(例如2个、3个、4个等)取代基所取代:卤素、C1-C6烷氧基、卤代的C1-C6烷氧基、C3-C8环烷基、卤代的C3-C8环烷基、甲基砜基、氧代(=O)、-CN、羟基、-NH 2、-NHS(O) 2CH 3、C1-C6胺基、羧基、C1-C6酰胺基(-C(=O)-N(Rc) 2或-NH-C(=O)(Rc),Rc为H或C1-C5的烷基)、或取代或未取代的选自下组的基团:C1-C6烷基、C6-C10芳基、具有1-3个选自N、S和O的杂原子的5-10元杂芳基、-(CH 2)-C6-C10芳基、-(CH 2)-(具有1-3个选自N、S和O的杂原子的3-10元杂芳基)、-(具有1-3个选自N、S和O的杂原子的5-10元亚杂芳基)-(C1-C6烷基)、具有1-3个选自下组N、S和O的杂原子的3-12元杂环基(包括单环、螺环、桥环或并环),且所述的取代基选自下组:卤素、C1-C6烷基、C1-C6亚烷基-OH、C1-C6烷氧基、氧代、-S(O) 2CH 3、-CN、-OH、C6-C10芳基、具有1-3个选自N、S和O的杂原子的3-10元杂芳基、-C(O)CHNH 2、-C(O)CHOH;
    且所述的式I化合物中,各个手性中心为R构型或S构型。
  2. 如权利要求1所述的化合物,或其药学上可接受的盐、光学异构体或水合物,其特征在于,所述的A环选自下组:
    Figure PCTCN2021078965-appb-100004
    其中,
    Z选自下组:NH、O或S;
    q为0、1或2;
    Re和Rf各自独立地选自下组:H、CN、卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、苯基、取代或未取代的C3-C8环烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基;或所述的Re和Rf与其相连的原子共同构成C3-C8环烷基、或取代或未取代的具有1-3个选自N、S和O的杂原子的3-12元杂环基;
    且所述的
    Figure PCTCN2021078965-appb-100005
    还可以进一步被1或2个R 2取代。
  3. 如权利要求1所述的化合物,或其药学上可接受的盐、光学异构体或水合物,其特征在于,所述的A环选自下组:C3-C8环烷基、苯环、5-6元的杂环、或具有1-3个选自N、S和O的杂原子的5-10元的杂芳环;且所述的R 2选自下组:H、卤素、CN、取代或未取代的C1-C6烷基。
  4. 如权利要求1所述的化合物,或其药学上可接受的盐、光学异构体或水合物,其特征在于,所述的B环选自下组:苯环、3-12元杂环基、5-6元的杂芳环、含有苯环结构单元的8-15元二环并环、含有5-6元杂芳环结构单元的8-15元二环并环;其中,所述的R 4选自下组:H、卤素、取代或未取代的C1-C6烷基、R 9R 10N-C(=O)-、R 9R 10N-C(=O)NR-、R 11R 12N-S(=O) 2-、R 7O-、R 8-S(O) 2NR-;其中,所述的取代优选包括一个或多个选自下组的基团:-NH 2、取代或未取代的C1-C6烷基。
  5. 如权利要求1所述的化合物,或其药学上可接受的盐、光学异构体或水合物,其特征在于,所述的式I化合物具有如下式II-1或II-2所示的结构:
    Figure PCTCN2021078965-appb-100006
    其中,
    所述的Ra独立地选自下组:卤素、CN、取代或未取代的C1-C6烷基;
  6. 如权利要求5所述的化合物,或其药学上可接受的盐、光学异构体或水合物,其特征在于,所述的式I化合物具有如下式III所示的结构:
    Figure PCTCN2021078965-appb-100007
    其中,
    所述的R 4选自下组:H、卤素、CN、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、R 9R 10N-C(=O)-、R 8-S(=O) 2-、R 9R 10N-C(=O)-、R 9R 10N-C(=O)NR-、R 11R 12N-S(=O) 2-、R 7O-、R 8-S(O) 2NR-;其中,所述的R 7、R 8、R 9、R 10、R 11、R 12各自独立地选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的具有1-3个选自下组N、S和O的杂原子的3-12元杂环基;或所述的R 9和R 10或R 11和R 12与其相连的氮原子共同构成取代或未取代的具有1-3个选自N、S和O的杂原子的3-12元杂环基。
  7. 如权利要求1所述的化合物,或其药学上可接受的盐、光学异构体或水合物,其特征在于,所述的式I化合物具有如下式IV所示的结构:
    Figure PCTCN2021078965-appb-100008
  8. 如权利要求1所述的化合物,或其药学上可接受的盐、光学异构体或水合物,其特征在于,所述的式I化合物选自下组:
    Figure PCTCN2021078965-appb-100009
    Figure PCTCN2021078965-appb-100010
    Figure PCTCN2021078965-appb-100011
    Figure PCTCN2021078965-appb-100012
  9. 如权利要求1所述的化合物,或其药学上可接受的盐、光学异构体或水合物,其特征在于,所述的式I化合物选自下组:
    Figure PCTCN2021078965-appb-100013
    Figure PCTCN2021078965-appb-100014
    Figure PCTCN2021078965-appb-100015
    Figure PCTCN2021078965-appb-100016
  10. 一种药物组合物,其特征在于,所述的药物组合物包括(1)如权利要求1所述的化合物或其立体异构体或互变异构体,或其药学上可接受的盐、水合物或溶剂化物;(2)药学上可接受的载体。
  11. 如权利要求1所述的化合物或其立体异构体或互变异构体,或其药学上可接受的盐、水合物或溶剂化物,或如权利要求8所述的药物组合物的用途,其特征在于,用于制备预防和/或治疗与HPK1激酶的活性或表达量相关的疾病的药物组合物。
  12. 权利要求1所述的化合物及其药学上可接受的盐、立体异构体、酯、前药、溶剂化物和氘代化合物单独用药或者联合肿瘤化疗方案,其他肿瘤免疫治疗剂(小分子化合物及抗体包括但不限于PD-1、PD-L1、CTLA-4、TIM-3、TGF-β及其受体、LAG3拮抗剂或TLR4、TLR7、TLR8、TLR9、STING激动剂等),放疗方案,肿瘤靶向药,肿瘤疫苗等,可在所述药剂之前、之后或同时施用,或者可以与其他已知疗法共施用。也可用作疫苗佐剂。
  13. 权利要求1所述的化合物及其药学上可接受的盐、立体异构体、酯、前药、溶剂化物和氘代化合物与CAR-T免疫疗法相结合在癌症免疫疗法中的应用。
  14. 权利要求10-12任一所述的应用,所述的疾病包括但不限于:淋巴瘤,母细胞瘤,髓母细胞瘤,视网膜母细胞瘤,肉瘤,脂肪肉瘤,滑膜细胞肉瘤,神经内分泌肿瘤,类癌肿瘤,胃泌素瘤,胰岛细胞癌,间皮瘤,神经鞘瘤,听神经瘤,脑膜瘤,腺癌,黑色素瘤,白血病或淋巴样恶性肿瘤,鳞状细胞癌,上皮鳞状细胞癌,肺癌,小细胞肺癌,非小细胞肺癌,腺癌肺癌,肺鳞癌,腹膜癌,肝细胞癌,胃癌,肠癌,胰腺癌,成胶质细胞瘤,子宫颈癌,卵巢癌,肝癌,膀胱癌,乳腺癌,转移性乳腺癌,结肠癌,直肠癌,结直肠癌,子宫癌,唾液腺癌,肾癌,前列腺癌,外阴癌,甲状腺癌,肛门癌,阴茎癌,梅克尔细胞癌,食管癌,胆道肿瘤,头颈部癌,血液恶性肿瘤,鼻咽癌,多发性骨髓瘤,大场绒毛腺瘤,非霍奇金淋巴瘤,骨癌,睾丸癌,霍奇金病,精元细胞瘤,口腔癌,脑癌,皮肤癌,乳腺导管癌,肾盂癌,肾母细胞瘤,食管腺瘤,视网膜细胞瘤,神经胶质瘤,神经纤维瘤,胃肠道间质瘤,原位癌,子宫内膜癌和骨髓增生异常综合征等。
PCT/CN2021/078965 2020-03-03 2021-03-03 新型hpk1抑制剂及其制备方法和应用 WO2021175270A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180019207.1A CN115279760A (zh) 2020-03-03 2021-03-03 新型hpk1抑制剂及其制备方法和应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010140893.5A CN113354648A (zh) 2020-03-03 2020-03-03 新型hpk1抑制剂及其制备方法和应用
CN202010140893.5 2020-03-03

Publications (1)

Publication Number Publication Date
WO2021175270A1 true WO2021175270A1 (zh) 2021-09-10

Family

ID=77523188

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/078965 WO2021175270A1 (zh) 2020-03-03 2021-03-03 新型hpk1抑制剂及其制备方法和应用

Country Status (2)

Country Link
CN (2) CN113354648A (zh)
WO (1) WO2021175270A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022199652A1 (en) * 2021-03-24 2022-09-29 Impact Therapeutics (Shanghai) , Inc Five-membered heteroaryl-pyrimidine compounds as usp1 inhibitors and the use thereof
WO2022228549A1 (en) * 2021-04-30 2022-11-03 Chengdu Anticancer Bioscience, Ltd. Phenyl -o-quinoline, quinazoline, thienopyridine, thienopyrimidine, pyrrolopyridine, pyrrolopyrimidine compounds having anticancer activity
WO2022253311A1 (zh) * 2021-06-04 2022-12-08 武汉人福创新药物研发中心有限公司 Hpk1抑制剂及其应用
WO2024022519A1 (zh) * 2022-07-28 2024-02-01 先声再明医药有限公司 杂环并嘧啶类化合物及其应用

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230130909A1 (en) * 2020-04-20 2023-04-27 Wigen Biomedicine Technology (shanghai) Co., Ltd. Hpk1 inhibitor, preparation method therefor and use thereof
WO2022166920A1 (zh) * 2021-02-08 2022-08-11 杭州中美华东制药有限公司 一种吡咯并哒嗪类化合物及其制备方法和用途
EP4353721A1 (en) * 2021-05-11 2024-04-17 Evopoint Biosciences Co., Ltd. Nitrogen-containing heterocyclic compound
CN117003754A (zh) * 2022-04-28 2023-11-07 腾讯科技(深圳)有限公司 吡咯并[2,3-d]嘧啶或吡唑并[3,4-d]嘧啶衍生物及其用途
WO2024027755A1 (zh) * 2022-08-05 2024-02-08 杭州中美华东制药有限公司 一种protac嵌合化合物及其制备方法和用途
CN116162087A (zh) * 2023-02-20 2023-05-26 杭州医学院 一种hpk 1和/或lck激酶调节剂、制备方法及其应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019016071A1 (en) * 2017-07-18 2019-01-24 Bayer Pharma Aktiengesellschaft SUBSTITUTED PYRROLOPYRIDINE DERIVATIVES
WO2019238067A1 (en) * 2018-06-13 2019-12-19 Beigene, Ltd. Pyrrolo [2, 3-b] pyridines or pyrrolo [2, 3-b] pyrazines as hpk1 inhibitor and the use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019016071A1 (en) * 2017-07-18 2019-01-24 Bayer Pharma Aktiengesellschaft SUBSTITUTED PYRROLOPYRIDINE DERIVATIVES
WO2019238067A1 (en) * 2018-06-13 2019-12-19 Beigene, Ltd. Pyrrolo [2, 3-b] pyridines or pyrrolo [2, 3-b] pyrazines as hpk1 inhibitor and the use thereof

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022199652A1 (en) * 2021-03-24 2022-09-29 Impact Therapeutics (Shanghai) , Inc Five-membered heteroaryl-pyrimidine compounds as usp1 inhibitors and the use thereof
WO2022228549A1 (en) * 2021-04-30 2022-11-03 Chengdu Anticancer Bioscience, Ltd. Phenyl -o-quinoline, quinazoline, thienopyridine, thienopyrimidine, pyrrolopyridine, pyrrolopyrimidine compounds having anticancer activity
WO2022253311A1 (zh) * 2021-06-04 2022-12-08 武汉人福创新药物研发中心有限公司 Hpk1抑制剂及其应用
WO2024022519A1 (zh) * 2022-07-28 2024-02-01 先声再明医药有限公司 杂环并嘧啶类化合物及其应用

Also Published As

Publication number Publication date
CN115279760A (zh) 2022-11-01
CN113354648A (zh) 2021-09-07

Similar Documents

Publication Publication Date Title
WO2021175270A1 (zh) 新型hpk1抑制剂及其制备方法和应用
RU2747260C2 (ru) Ингибитор рфрф4, способ его получения и его фармацевтическое применение
US11661423B2 (en) Heterocyclic compounds as RET kinase inhibitors
KR102558066B1 (ko) Tam 억제제로서 피롤로트리아진 화합물
AU2017250448C1 (en) Heterocyclic compounds as RET kinase inhibitors
TWI503319B (zh) 吡啶酮和氮雜吡啶酮之化合物及其使用方法
AU2005332339B2 (en) Furanopyridine derivatives as ACK1 and Lck modulators
BR112019021992A2 (pt) inibidores de ahr de indol e usos dos mesmos
KR20210089716A (ko) 질소 함유 축합 헤테로고리 shp2억제제 화합물, 제조방법 및 용도
KR101469334B1 (ko) N-9-치환된 퓨린 화합물, 조성물 및 사용 방법
US9447101B2 (en) Pyrrolo[2,1-f][1,2,4]triazine compound, and preparation method and application thereof
WO2013134252A1 (en) 2-amino, 6 - phenyl substituted pyrido [2, 3 - d] pyrimidine derivatives useful as raf kinase inhibitors
MX2012009059A (es) Compuestos inhibidores de fosfoinositida 3-cinasa delta (pi3k) pirido [3,2-d]pirimidina y metodos de uso.
AU2009259867A1 (en) Triazolopyridine JAK inhibitor compounds and methods
TWI707855B (zh) 咪唑并嗒類化合物及其用途
CA3124678A1 (en) Aza-heterobicyclic inhibitors of mat2a and methods of use for treating cancer
CA2744154A1 (en) Pyrazolopyridine pi3k inhibitor compounds and methods of use
WO2018233527A1 (zh) 一种具有csf1r抑制活性的氮杂芳基衍生物、其制备方法和应用
WO2021175271A1 (zh) 新型hpk1抑制剂及其制备方法和应用
TW202200575A (zh) 一種免疫抑制劑、其製備方法和應用
WO2018214866A1 (zh) 一种氮杂芳基衍生物、其制备方法和在药学上的应用
JP2023145547A (ja) Cd73阻害剤、その製造方法と応用
CA3191362A1 (en) Pyrazolopyridazinone compound, and pharmaceutical composition and use thereof
WO2020057669A1 (zh) 一类具有激酶抑制活性的芳香杂环类化合物
WO2021239727A1 (en) 4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)-3,6-dihydropyridine-1-(2h)-carboxamide derivatives as limk and/or rock kinases inhibitors for use in the treatment of cancer

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: 21763929

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: 21763929

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 21763929

Country of ref document: EP

Kind code of ref document: A1