WO2021036922A1 - 抑制并诱导降解egfr和alk的化合物 - Google Patents

抑制并诱导降解egfr和alk的化合物 Download PDF

Info

Publication number
WO2021036922A1
WO2021036922A1 PCT/CN2020/110442 CN2020110442W WO2021036922A1 WO 2021036922 A1 WO2021036922 A1 WO 2021036922A1 CN 2020110442 W CN2020110442 W CN 2020110442W WO 2021036922 A1 WO2021036922 A1 WO 2021036922A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
halogen
haloalkyl
compound
cycloalkyl
Prior art date
Application number
PCT/CN2020/110442
Other languages
English (en)
French (fr)
Inventor
赵焰平
王红军
王业明
范伏田
姜媛媛
王晓倩
梁会宁
谢勇
张衍豪
刘凯
冯泽旺
刘雪莲
Original Assignee
北京泰德制药股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 北京泰德制药股份有限公司 filed Critical 北京泰德制药股份有限公司
Priority to KR1020227009748A priority Critical patent/KR20220051244A/ko
Priority to EP20857158.8A priority patent/EP4019021A4/en
Priority to JP2022512798A priority patent/JP2022546375A/ja
Priority to US17/637,690 priority patent/US20220306659A1/en
Priority to CN202080059273.7A priority patent/CN114286678A/zh
Publication of WO2021036922A1 publication Critical patent/WO2021036922A1/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/645Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having two nitrogen atoms as the only ring hetero atoms
    • C07F9/6509Six-membered rings
    • C07F9/6512Six-membered rings having the nitrogen atoms in positions 1 and 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/662Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D471/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/645Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having two nitrogen atoms as the only ring hetero atoms
    • C07F9/6509Six-membered rings
    • C07F9/650952Six-membered rings having the nitrogen atoms in the positions 1 and 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings

Definitions

  • the present invention relates to the field of medicine. Specifically, the present invention provides a compound capable of inhibiting EGFR and ALK or inducing degradation of EGFR and ALK, and a preparation method and application thereof.
  • Non-Small Cell Lung Cancer is one of the most common malignant tumors. In 2018, there were 2.1 million new lung cancers worldwide, accounting for 11.6% of all new tumor cases; 1.8 million deaths, accounting for 18.4% of all tumor deaths. Among them, non-small cell lung cancer (Non-Small Cell Lung Cancer, NSCLC) accounts for 80%-85% of the total number of lung cancers. Epidermal Growth Factor Receptor (EGFR) and Anaplastic Lymphoma Kinase (ALK) are common driving genes for non-small cell lung cancer.
  • EGFR Epidermal Growth Factor Receptor
  • ALK Anaplastic Lymphoma Kinase
  • EGFR small molecule inhibitors are the first-line standard treatment for non-small cell lung cancer with mutations in the EGFR gene. They have been widely used in the field of lung cancer treatment. They inhibit the activation of tyrosine kinase by binding to EGFR competitively with endogenous ligands. Furthermore, it blocks the EGFR signaling pathway, inhibits the proliferation and metastasis of tumor cells, and promotes a series of biological effects such as tumor cell apoptosis.
  • the first-generation EGFR small molecule inhibitors Gefitinib and Erlotinib have been used to treat advanced non-small cell lung cancer with activated EGFR mutations (L858R, del E746-A750).
  • activated EGFR mutations L858R, del E746-A750.
  • patients will develop drug resistance after using Gefitinib and Erlotinib for 10-12 months.
  • more than 50% of drug-resistant patients are due to the secondary mutation of EGFR T790M leading to drug resistance.
  • Afatinib a second-generation EGFR irreversible inhibitor
  • Osimertinib a third-generation irreversible inhibitor, overcomes EGFR T790M resistance and can effectively treat patients with advanced non-small cell lung cancer who are resistant to EGFR T790M mutations.
  • Osimertinib has achieved great success in the treatment of non-small cell lung cancer with EGFR T790M mutations, some benefited patients have developed drug resistance after 9-14 months of treatment (Nature Medicine, 2015, 21(6) ,560-562). Studies have shown that up to 22% of drug-resistant patients are resistant to Osimertinib due to EGFR C797S mutations (JAMA Oncol. 2018; 4(11): 1527-1534). The EGFR C797S mutation causes the cysteine at position 797 to be mutated to serine, and Osimertinib cannot covalently bind to EGFR, eventually causing drug resistance. At present, there is still a lack of effective EGFR inhibitors against EGFR C797S alone in clinical practice. Therefore, the development of a new generation of EGFR inhibitors to meet the needs of clinical treatment is a problem that needs to be solved urgently.
  • Anaplastic lymphoma kinase is a receptor tyrosine protein kinase.
  • ALK gene rearrangement, point mutations and gene amplification can cause cancer in the body.
  • ALK rearrangement gene is a strong oncogenic driver gene, among which echinoderms microtubule-associated protein 4 (EML4-ALK) and nucleophosphate protein (NPM-ALK) are common types.
  • EML4-ALK microtubule-associated protein 4
  • NPM-ALK nucleophosphate protein
  • ALK gene rearrangement leads to the phosphorylation process of ALK before the formation of dimers. Therefore, the ALK fusion protein will continue to be activated and activate its downstream pathways, which will cause excessive cell proliferation and cause tumors. In non-small cell lung cancer, 3-7% of patients have ALK gene rearrangement.
  • Small molecule inhibitors that target ALK have been widely used in clinics. They compete with endogenous ligands to bind ALK. Inhibit the activation of tyrosine kinase, thereby blocking the ALK signal pathway, inhibiting a series of biological effects such as tumor cell proliferation and metastasis.
  • the ALK inhibitors currently on the market include crizotinib, ceritinib, alectinib, brigatinib, and lorlatinib, but it is inevitable that these inhibitors will develop resistance. Common ALK resistance mutations are L1196M, G1269A, S1206Y, G1202R, C1156Y, L1198F etc. Therefore, it is of great significance to develop new ALK inhibitors to meet the needs of clinical treatment.
  • Ubiquitin-Proteasome System is a multi-component system of protein degradation in cells, involved in cell growth and differentiation, DNA replication and repair, cell metabolism, immune response and other important physiological and biochemical processes.
  • Protein degradation mediated by the ubiquitin-proteasome pathway is an important mechanism for the body to regulate intracellular protein levels and functions, and it plays an important role in maintaining protein homeostasis in the body.
  • Through the intracellular ubiquitin-proteasome pathway it induces the degradation of EGFR or ALK, which provides a new idea for the treatment of cancer.
  • the present invention provides a compound capable of inhibiting EGFR and ALK, and/or inducing degradation of EGFR and ALK, and a preparation method and application thereof.
  • the present invention provides compounds of general formula (X), or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs thereof Types, prodrugs or isotopic variants, and mixtures thereof, can be used to treat EGFR and/or ALK kinase-mediated diseases, such as cancer.
  • Ring A is selected from the following optionally substituted groups: C 3-7 cycloalkyl, 4-8 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, wherein the substituent is selected from Halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 3-7 cycloalkyl, 4-8 membered heterocyclic group, -P(O)(C 1 -6 alkyl) 2 , -P(O)(C 2-6 alkenyl) 2 , -OC 1-6 alkyl, -OC 1-6 haloalkyl, -OC 2-6 alkenyl, -OC 3- 7 cycloalkyl, -O-4-8 membered heterocyclic group, -NH-C 1-6 alkyl, -NH-C 2-6 alkenyl, -NH-C 3-7 cycloalkyl, -NH- 4-8 membered heterocyclic group, -
  • Ring B is selected from the following groups:
  • connection position with other parts of the molecule can be located in the available position of the ring
  • Z 1 is an O, S, N or C atom, which is optionally substituted by one or two R Z1 ; or Z 1 is absent, so Z 4 is connected to Z 2 , Z 3 or the aromatic ring connected to Z 1
  • the C atom of, and the Z 2 connected to Z 1 and the C atom on the aromatic ring are respectively connected to R W ; or Z 1 , Z 2 and Z 3 do not exist, so Z 4 is connected to Z 1 or Z 3
  • One of the C atoms on the aromatic ring of, and the other C atom on the aromatic ring is connected to R W;
  • Z 2 is an O, S, N or C atom, which is optionally substituted by one or two R Z2;
  • Z 3 is an O, S, N or C atom, which is optionally substituted by one or two R Z3 ; with the proviso that, when When it is a double bond, Z 2 is a N or C atom, and Z 3 is a N or C atom;
  • Z 4 is N or CR Z4 ;
  • Z 5 is N or CR Z5 ;
  • R N1 is H, C 1-6 alkyl or C 1-6 haloalkyl, preferably H;
  • R Z4 is H, CN, halogen, -(CH 2 ) 0-5 -OR", -(CH 2 ) 0-5 -NR"R"', C 1-6 alkyl or C 1-6 haloalkyl;
  • R Z5 is H, CN, halogen, -(CH 2 ) 0-5 -OR", -(CH 2 ) 0-5 -NR"R"', C 1-6 alkyl, C 1-6 haloalkyl, -(CH 2 ) 0-5 -C 3-7 cycloalkyl or -(CH 2 ) 0-5 -4-8 membered heterocyclic group;
  • R w is H, CN, halogen, -(CH 2 ) 0-5 -OR", -(CH 2 ) 0-5 -NR"R"', -C 1-6 alkyl, C 1-6 haloalkane Group, -(CH 2 ) 0-5 -C 3-7 cycloalkyl, -(CH 2 ) 0-5 -4-8 membered heterocyclyl, C 2-6 alkenyl, C 2-6 alkynyl, -(CH 2 ) 0-5 -C 3-10 halocycloalkyl, -(CH 2 ) 0-5 -C 6-10 aryl or -(CH 2 ) 0-5 -5-14-membered heteroaromatic base;
  • R" is H, C 1-6 alkyl, C 1-6 haloalkyl or -(CH 2 ) 0-5 -C 3-7 cycloalkyl;
  • R"' is H, C 1-6 alkyl or C 1-6 haloalkyl
  • E is independently selected from: chemical bond, -C c R # R # '-C d R # R # '-C e R # R # ',
  • two E units can form -CH 2 CH 2 OCH 2 CH 2 -, -OCH 2 CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 O-,
  • H 1 and H 2 are N or C atoms, H 3 is O, S, N or C atom, and H 1 and H 3 , H 2 and H 3 are not heteroatoms at the same time;
  • H 4 and H 5 are N or C atoms
  • H 6 , H 7 , H 8 and H 9 are C or N atoms
  • p 0, 1 or 2;
  • q 1 or 2;
  • R * is H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, C 3-10 halocycloalkyl , 3-10 membered heterocyclic group, C 6-10 aryl group or 5-14 membered heteroaryl group;
  • R # is H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, C 3-10 halo ring Alkyl, 3-10 membered heterocyclyl, C 6-10 aryl or 5-14 membered heteroaryl;
  • R # ' is H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, C 3-10 halo Cycloalkyl, 3-10 membered heterocyclyl, C 6-10 aryl or 5-14 membered heteroaryl;
  • R # and R # on adjacent atoms can form a chemical bond
  • R # 'and R # ' on adjacent atoms can form a chemical bond
  • n 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10;
  • R s1 is selected from H, CN, halogen, -(CH 2 ) 0-5 -OR", -(CH 2 ) 0-5 -NR"R"', C 1-6 alkyl, C 1-6 haloalkyl , -(CH 2 ) 0-5 -4-8 membered heterocyclyl, C 2-6 alkenyl, C 2-6 alkynyl, -(CH 2 ) 0-5 -C 3-7 cycloalkyl,- (CH 2 ) 0-5 -C 3-10 halocycloalkyl, -(CH 2 ) 0-5 -C 6-10 aryl, -(CH 2 ) 0-5 -5-14 membered heteroaryl , -C(O)R W , -S(O)R W or -S(O) 2 R W ;
  • s1 is 0, 1, 2 or 3;
  • R' is selected from H, -C(O)-C 1-6 alkyl, -C(O)-C 1-6 haloalkyl, -C(O)-C 2-6 alkenyl or -C(O) -C 6-10 aryl;
  • L is a chemical bond, -O- or -NR-;
  • R is H or C 1-6 alkyl
  • T is selected from a chemical bond, a C 2-6 heteroalkylene group, a 4-12 membered heterocyclylene group or a 5-6 membered heterocyclylene group substituted with a 5-6 membered heterocyclylene group;
  • R 1 is selected from H, halogen, cyano, C 1-6 alkyl, C 3-7 cycloalkyl, 4-8 membered heterocyclic group, C 1-6 haloalkyl, C 2-6 alkenyl or C 2 -6 alkynyl;
  • R 1 and R together with the atoms to which they are attached form an optionally substituted 5-6 membered heterocyclic group, wherein the substituent is selected from halogen, oxo, C 1-6 alkane Group, C 1-6 haloalkyl, halogen monosubstituted or polysubstituted C 6-10 aryl;
  • R 2 is H, halogen, hydroxy, amino, C 1-6 alkyl or C 1-6 haloalkyl
  • R 1 and R 2 together with the atoms to which they are connected form a 5-6 membered heterocyclic group or a 5-6 membered heteroaryl group;
  • R 3 is selected from H, -OC 1-6 alkyl or -OC 1-6 haloalkyl
  • R 4 is selected from H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, -NHC(O)-C 1-6 alkyl or -NHC(O)-C 2-6 alkenyl;
  • each of the above groups is H or a H-containing group
  • the one or more H atoms may be substituted by D atoms
  • alkyl, alkylene, haloalkyl, alkenyl, alkynyl, cycloalkyl, halocycloalkyl, heterocyclyl, aryl, heteroaryl or contained in L 1 , E, L 2 , and T OH, NH, NH 2, CH , CH 2, CH 3 groups are each optionally substituted with three or more at each occurrence R s, wherein R s in each occurrence Independently selected from: halogen, hydroxy, amino, cyano, nitro, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 ring Alkyl, C 3-10 halocycloalkyl, 3-10 membered heterocyclic group, C 6-10 aryl, 5-14 membered heteroaryl, C 6-12 aralkyl, -OR a' ,- OC (O) R a ', -C (
  • n is independently 1 or 2 each time it appears;
  • R a'and R b' are each independently selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkyl-O-, C 1-6 alkyl-S-, C 3-10 cycloalkyl, 3-10 membered heterocyclic group, C 6-10 aryl, 5-14 membered heteroaryl, and C 6-12 aralkyl.
  • the present invention provides a pharmaceutical composition containing a compound of the present invention, and optionally a pharmaceutically acceptable excipient.
  • the present invention provides a pharmaceutical composition containing a compound of the present invention and a pharmaceutically acceptable excipient, which also contains other therapeutic agents.
  • the present invention provides a kit comprising a compound of the present invention, and other therapeutic agents and a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • the present invention provides the use of the compound of the present invention in the preparation of a medicament for the treatment and/or prevention of diseases mediated by EGFR and/or ALK kinase.
  • the present invention provides a method of treating and/or preventing a disease mediated by EGFR and/or ALK kinase in a subject, comprising administering a compound of the present invention or a composition of the present invention to the subject.
  • the present invention provides a compound of the present invention or a composition of the present invention for use in the treatment and/or prevention of diseases mediated by EGFR and/or ALK kinase.
  • the diseases treated by the present invention include cancers, such as ovarian cancer, cervical cancer, colorectal cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, melanoma, prostate cancer, leukemia, lymphoma Tumor, non-Hodgkin’s lymphoma, gastric cancer, lung cancer, hepatocellular carcinoma, gastric cancer, gastrointestinal stromal tumor (GIST), thyroid cancer, cholangiocarcinoma, endometrial cancer, kidney cancer, anaplastic large cell lymphoma, acute Myeloid leukemia (AML), multiple myeloma, melanoma, mesothelioma.
  • cancers such as ovarian cancer, cervical cancer, colorectal cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, melanoma, prostate cancer, leukemia, lymphoma Tumor, non-Hodgkin’s lymphoma, gastric cancer,
  • C 1-6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1-6 , C 1-5 , C 1-4 , C 1-3 , C 1 -2 , C 2-6 , C 2-5 , C 2-4 , C 2-3 , C 3-6 , C 3-5 , C 3-4 , C 4-6 , C 4-5 and C 5 -6 alkyl.
  • C 1-6 alkyl refers to a linear or branched saturated hydrocarbon group having 1 to 6 carbon atoms. In some embodiments, C 1-4 alkyl is preferred. Examples of C 1-6 alkyl groups include: methyl (C 1 ), ethyl (C 2 ), n-propyl (C 3 ), isopropyl (C 3 ), n-butyl (C 4 ), tert-butyl Base (C 4 ), sec-butyl (C 4 ), isobutyl (C 4 ), n-pentyl (C 5 ), 3-pentyl (C 5 ), pentyl (C 5 ), neopentyl ( C 5 ), 3-methyl-2-butyl (C 5 ), tert-amyl (C 5 ) and n-hexyl (C 6 ).
  • C 1-6 alkyl also includes heteroalkyls in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, Phosphorus) substitution.
  • the alkyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • alkyl abbreviations include: Me(-CH 3 ), Et(-CH 2 CH 3 ), iPr(-CH(CH 3 ) 2 ), nPr(-CH 2 CH 2 CH 3 ), n-Bu(-CH 2 CH 2 CH 2 CH 3 ) or i-Bu(-CH 2 CH(CH 3 ) 2 ).
  • C 2-6 alkenyl group refers to a straight or branched chain hydrocarbon group having 2 to 6 carbon atoms and at least one carbon-carbon double bond. In some embodiments, C 2-4 alkenyl is preferred. Examples of C 2-6 alkenyl groups include: vinyl (C 2 ), 1-propenyl (C 3 ), 2-propenyl (C 3 ), 1-butenyl (C 4 ), 2-butenyl (C 4 ), butadienyl (C 4 ), pentenyl (C 5 ), pentadienyl (C 5 ), hexenyl (C 6 ), etc.
  • C 2-6 alkenyl also includes heteroalkenyl groups in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, Phosphorus) substitution.
  • Alkenyl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 2-6 alkynyl refers to a straight or branched chain hydrocarbon group having 2 to 6 carbon atoms, at least one carbon-carbon triple bond, and optionally one or more carbon-carbon double bonds. In some embodiments, C 2-4 alkynyl is preferred. Examples of C 2-6 alkynyl groups include but are not limited to: ethynyl (C 2 ), 1-propynyl (C 3 ), 2-propynyl (C 3 ), 1-butynyl (C 4 ), 2-butynyl (C 4 ), pentynyl (C 5 ), hexynyl (C 6 ), etc.
  • C 2-6 alkynyl also includes heteroalkynyl groups in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, Phosphorus) substitution.
  • An alkynyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 1-6 alkylene, C 2-6 alkenylene or C 2-6 alkynylene refers to the above-defined "C 1-6 alkyl, C 2-6 alkenyl or C 2-6 Alkynyl" is a divalent group.
  • C 1-6 alkylene group refers to a divalent group formed by removing another hydrogen of the C 1-6 alkyl group, and may be substituted or unsubstituted. In some embodiments, C 1-4 alkylene is particularly preferred.
  • the unsubstituted alkylene group includes but is not limited to: methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), butylene Group (-CH 2 CH 2 CH 2 CH 2 -), pentylene (-CH 2 CH 2 CH 2 CH 2 CH 2 -), hexylene (-CH 2 CH 2 CH 2 CH 2 CH 2 CH 2 -) ,and many more.
  • alkylene groups substituted by one or more alkyl groups (methyl) include but are not limited to: substituted methylene (-CH(CH 3 )- , -C(CH 3 ) 2 -), substituted ethylene (-CH(CH 3 )CH 2 -, -CH 2 CH(CH 3 )-, -C(CH 3 ) 2 CH 2 -, -CH 2 C(CH 3 ) 2- ), substituted propylene (-CH(CH 3 )CH 2 CH 2 -, -CH 2 CH(CH 3 )CH 2 -, -CH 2 CH 2 CH(CH 3 ) -, -C(CH 3 ) 2 CH 2 CH 2 -, -CH 2 C(CH 3 ) 2 CH 2 -, -CH 2 CH 2 C(CH 3 ) 2 -), etc.
  • C 2-6 alkynylene group refers to a divalent group formed by removing another hydrogen of the C 2-6 alkynyl group, and may be substituted or unsubstituted. In some embodiments, C 2-4 alkynylene groups are particularly preferred. Exemplary alkynylene groups include, but are not limited to, ethynylene (-C ⁇ C-), substituted or unsubstituted propynylene (-C ⁇ CCH 2 -), and the like.
  • C 1-6 heteroalkyl refers to a C 1-6 alkyl group as defined herein, and in the parent chain, it further contains one or more (for example, 1, 2, 3, or 4) heteroatoms (for example, oxygen, sulfur, nitrogen, boron, silicon, phosphorus), wherein one or more heteroatoms are between adjacent carbon atoms in the parent carbon chain, and/or, one or more heteroatoms are in the carbon Between the atom and the parent molecule, that is, between the points of attachment.
  • the point of attachment of the C 1-6 heteroalkyl group to the parent molecule may be a carbon atom or a heteroatom.
  • C 2-6 heteroalkylene group refers to a divalent group formed by removing another hydrogen of a C 1-6 heteroalkyl group, and may be substituted or unsubstituted.
  • the point of attachment between the C 1-6 heteroalkylene group and other parts of the parent molecule can be two carbon atoms, two heteroatoms, or one carbon atom and one heteroatom.
  • Halo or "halogen” refers to fluorine (F), chlorine (Cl), bromine (Br) and iodine (I).
  • C 1-6 haloalkyl refers to the above-mentioned “C 1-6 alkyl", which is substituted with one or more halogen groups.
  • C 1-4 haloalkyl is particularly preferred, and C 1-2 haloalkyl is more preferred.
  • Exemplary haloalkyl groups include, but the are not limited to: -CF 3, -CH 2 F, -CHF 2, -CHFCH 2 F, -CH 2 CHF 2, -CF 2 CF 3, -CCl 3, -CH 2 Cl , -CHCl 2 , 2,2,2-trifluoro-1,1-dimethyl-ethyl, etc.
  • the haloalkyl group can be substituted at any available point of attachment, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 3-10 cycloalkyl refers to a non-aromatic cyclic hydrocarbon group having 3 to 10 ring carbon atoms and zero heteroatoms. In some embodiments, C 3-7 cycloalkyl and C 3-6 cycloalkyl are particularly preferred, and C 5-6 cycloalkyl is more preferred. Cycloalkyl also includes ring systems in which the above-mentioned cycloalkyl ring is fused with one or more aryl or heteroaryl groups, wherein the point of attachment is on the cycloalkyl ring, and in such cases, the number of carbons continues to indicate The number of carbons in the cycloalkyl system.
  • cycloalkyl groups include but are not limited to: cyclopropyl (C 3 ), cyclopropenyl (C 3 ), cyclobutyl (C 4 ), cyclobutenyl (C 4 ), cyclopentyl ( C 5 ), cyclopentenyl (C 5 ), cyclohexyl (C 6 ), cyclohexenyl (C 6 ), cyclohexadienyl (C 6 ), cycloheptyl (C 7 ), cycloheptene Group (C 7 ), cycloheptadienyl (C 7 ), cycloheptatrienyl (C 7 ), etc. Cycloalkyl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 3-10 halocycloalkyl group refers to the above-mentioned “C 3-10 cycloalkyl group", which is substituted with one or more halogen groups.
  • 3-12 membered heterocyclic group refers to a group of 3 to 12 membered non-aromatic ring system having ring carbon atoms and 1 to 5 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, Sulfur, boron, phosphorus and silicon.
  • the point of attachment may be a carbon or nitrogen atom.
  • a 4-12 membered heterocyclic group is preferred, which is a 4 to 12 membered non-aromatic ring system with ring carbon atoms and 1 to 5 ring heteroatoms; in some embodiments, 3-10 membered Heterocyclic group, which is a 3 to 10-membered non-aromatic ring system with ring carbon atoms and 1 to 5 ring heteroatoms; in some embodiments, 3-8 membered heterocyclic groups are preferred, which are ring carbon atoms and A 3 to 8 membered non-aromatic ring system with 1 to 4 ring heteroatoms; preferably a 3-6 membered heterocyclic group, which is a 3 to 6 membered non-aromatic ring system with ring carbon atoms and 1 to 3 ring heteroatoms; Preferably a 4-8 membered heterocyclic group, which is a 4 to 8-membered non-aromatic ring system having ring carbon atoms and 1 to 3 ring heteroatoms;
  • Heterocyclyl also includes a ring system in which the aforementioned heterocyclyl ring is fused with one or more cycloalkyl groups, wherein the point of attachment is on the cycloalkyl ring, or wherein the aforementioned heterocyclyl ring is connected to one or more aryl groups or Heteroaryl fused ring systems in which the point of attachment is on the heterocyclyl ring; and in this case, the number of ring members continues to indicate the number of ring members in the heterocyclyl ring system.
  • Exemplary 3-membered heterocyclic groups containing one heteroatom include, but are not limited to: aziridinyl, oxiranyl, and thiorenyl.
  • Exemplary 4-membered heterocyclic groups containing one heteroatom include, but are not limited to: azetidinyl, oxetanyl, and thietane.
  • Exemplary 5-membered heterocyclic groups containing one heteroatom include, but are not limited to: tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl and pyrrolyl-2, 5-dione.
  • Exemplary 5-membered heterocyclic groups containing two heteroatoms include, but are not limited to: dioxolane, oxasulfuranyl, disulfuranyl, and oxasulfuranyl.
  • Exemplary 5-membered heterocyclic groups containing three heteroatoms include, but are not limited to: triazolinyl, oxadiazolinyl, and thiadiazolinyl.
  • Exemplary 6-membered heterocyclic groups containing one heteroatom include, but are not limited to: piperidinyl, tetrahydropyranyl, dihydropyridyl, and thianyl.
  • Exemplary 6-membered heterocyclic groups containing two heteroatoms include, but are not limited to: piperazinyl, morpholinyl, dithiacyclohexyl, dioxanyl.
  • Exemplary 6-membered heterocyclic groups containing three heteroatoms include, but are not limited to: triazinanyl.
  • Exemplary 7-membered heterocyclic groups containing one heteroatom include, but are not limited to: azepanyl, oxepanyl, and thiepane.
  • Exemplary 5-membered heterocyclic groups fused to a C 6 aryl ring include, but are not limited to: indolinyl, isoindolinyl , Dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolinone, etc.
  • Exemplary 6-membered heterocyclic groups fused to a C 6 aryl ring include, but are not limited to: tetrahydroquinolinyl, tetrahydroisoquinolinyl, and many more.
  • the heterocyclyl group may be optionally substituted with one or more substituents, for example, 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 6-10 aryl refers to a monocyclic or polycyclic (e.g., bicyclic) 4n+2 aromatic ring system (e.g., having 6-10 ring carbon atoms and zero heteroatoms) (e.g., having a ring arrangement Shared 6 or 10 ⁇ electrons) groups.
  • an aryl group having six ring carbon atoms ( “C 6 aryl”; e.g., phenyl).
  • an aryl group has ten ring carbon atoms ("C 10 aryl”; for example, naphthyl, for example, 1-naphthyl and 2-naphthyl).
  • Aryl groups also include ring systems in which the above-mentioned aryl ring is fused with one or more cycloalkyl or heterocyclic groups, and the point of attachment is on the aryl ring. In this case, the number of carbon atoms continues to indicate The number of carbon atoms in the aryl ring system.
  • the aryl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • C 6-12 aralkyl means the group -R-R', where R is an alkyl moiety, R'is an aryl moiety, and the alkyl and aryl groups have 6-12 carbon atoms in total.
  • 5-14 membered heteroaryl group refers to a 5-14 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms (for example, having a shared ring arrangement 6, 10, or 14 ⁇ electrons), where each heteroatom is independently selected from nitrogen, oxygen, and sulfur.
  • the point of attachment may be a carbon or nitrogen atom.
  • Heteroaryl bicyclic ring systems can include one or more heteroatoms in one or both rings.
  • Heteroaryl groups also include ring systems in which the above-mentioned heteroaryl ring is fused with one or more cycloalkyl or heterocyclic groups, and the point of attachment is on the heteroaryl ring, in this case, the carbon atom The number continues to indicate the number of carbon atoms in the heteroaryl ring system.
  • a 5-10 membered heteroaryl group is preferred, which is a 5-10 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms.
  • a 5-6 membered heteroaryl group is particularly preferred, which is a 5-6 membered monocyclic or bicyclic 4n+2 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms .
  • Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyrrolyl, furyl, and thienyl.
  • Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to: imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl.
  • Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to: triazolyl, oxadiazolyl (for example, 1,2,4-oxadiazolyl), and thiadiazolyl.
  • Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to: tetrazolyl.
  • Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to: pyridyl.
  • Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to: pyridazinyl, pyrimidinyl, and pyrazinyl.
  • Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to, triazinyl and tetrazinyl, respectively.
  • Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to: azepinyl, oxepinyl, and thiepinyl.
  • Exemplary 5,6-bicyclic heteroaryl groups include, but are not limited to: indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl , Benzisofuranyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzoxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazolyl, Indenazinyl and purinyl.
  • Exemplary 6,6-bicyclic heteroaryl groups include, but are not limited to: naphthyridinyl, pterridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl, and quinazolinyl .
  • Heteroaryl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
  • alkyl group, alkenyl group, alkynyl group, cycloalkyl group, heterocyclic group, aryl group and heteroaryl group as defined herein are optionally substituted groups.
  • Each of Raa is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two Raa groups are combined to form a heterocyclic group or Heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl group is independently covered by 0, 1, 2, 3, 4 or 5 R dd groups Group replacement
  • Each of R cc is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two R cc groups are combined to form a heterocyclic ring Group or heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl group is independently covered by 0, 1, 2, 3, 4 or 5 R dd group substitution;
  • Each of R ee is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyl, and heteroaryl, wherein each alkyl, alkenyl, alkynyl, ring Alkyl, heterocyclyl, aryl and heteroaryl are independently substituted with 0, 1, 2, 3, 4 or 5 R gg groups;
  • Each of R ff is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or two R ff groups are combined to form a heterocyclic group Or heteroaryl ring, where each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl group is independently covered by 0, 1, 2, 3, 4 or 5 R gg Group substitution
  • cancer includes but is not limited to the following cancers: breast, ovary, cervix, prostate, testis, esophagus, stomach, skin, lung, bone, colon, pancreas, thyroid, bile duct, buccal cavity and pharynx (mouth), lips, Tongue, oral cavity, pharynx, small intestine, colorectal, large intestine, rectum, brain and central nervous system cancer, glioblastoma, neuroblastoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, adenocarcinoma, Adenoma, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder cancer, liver cancer, kidney cancer, bone marrow disorder, lymphatic disorder, Hodgkin's disease, hair cell carcinoma and leukemia.
  • treatment relates to reversing, reducing, inhibiting the progression of or preventing the disorder or condition to which the term applies, or one or more symptoms of such a disorder or condition.
  • treatment refers to the act of verb therapy, the latter being as just defined.
  • pharmaceutically acceptable salt refers to those carboxylate and amino acid addition salts of the compounds of the present invention, which are suitable for contact with patient tissues within the scope of reliable medical judgment, and will not cause inappropriate toxicity, The irritation, allergies, etc., commensurate with a reasonable benefit/risk ratio, are effective for their intended application, including (where possible) the zwitterionic form of the compounds of the invention.
  • Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali metal and alkaline earth metal hydroxides or organic amines.
  • metals used as cations are sodium, potassium, magnesium, calcium and the like.
  • suitable amines are N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine and procaine.
  • Base addition salts of acidic compounds can be prepared by contacting the free acid form with a sufficient amount of the required base in a conventional manner to form the salt.
  • the free acid can be regenerated by contacting the salt form with the acid in a conventional manner, and then separating the free acid.
  • the free acid forms are somewhat different from their respective salt forms in certain physical properties, such as solubility in polar solvents, but for the purposes of the present invention, the salts are still equivalent to their respective free acids.
  • the salt can be sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogen phosphate, dihydrogen phosphate, metaphosphate, pyrophosphate prepared from inorganic acid Salt, chloride, bromide, iodide, acid such as hydrochloric acid, nitric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, phosphoric acid, etc.
  • Representative salts include: hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurel Acid salt, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthoate, Methanesulfonate, glucoheptonate, lactobionate, laurylsulfonate and isethionate, etc.
  • Salts can also be prepared from organic acids, such as aliphatic mono- and di-carboxylic acids, phenyl-substituted alkanoic acids, hydroxyalkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, and the like.
  • organic acids such as aliphatic mono- and di-carboxylic acids, phenyl-substituted alkanoic acids, hydroxyalkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, and the like.
  • Representative salts include acetate, propionate, caprylate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, horse Lysoate, mandelate, benzoate, chlorobenzoic acid basin, methylbenzoate, dinitrobenzoate, naphthoate, benzenesulfonate, tosylate, phenylethyl Salt, citrate, lactate, maleate, tartrate, methanesulfonate, etc.
  • Pharmaceutically acceptable salts may include cations based on alkali metals and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, etc., as well as non-toxic ammonium, quaternary ammonium and amine cations, including but not limited to ammonium, tetramethyl Ammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, etc. Also encompasses salts of amino acids, such as arginine, gluconate, galacturonate, etc. (see, for example, Berge SMet al., "Pharmaceutical Salts," J. Pharm. Sci., 1977; 66:1- 19. This is incorporated as a reference).
  • Subjects to be administered include, but are not limited to: humans (ie, men or women of any age group, for example, pediatric subjects (e.g., infants, children, adolescents) or adult subjects (e.g., young Adults, middle-aged adults or older adults)) and/or non-human animals, for example, mammals, for example, primates (for example, cynomolgus monkeys, rhesus monkeys), cows, pigs, horses, sheep , Goats, rodents, cats and/or dogs.
  • the subject is a human.
  • the subject is a non-human animal.
  • the terms "human", “patient” and “subject” are used interchangeably herein.
  • treatment includes the effect that occurs when a subject suffers from a specific disease, disorder, or condition, which reduces the severity of the disease, disorder, or condition, or delays or slows the disease, disorder Or the development of a condition ("therapeutic treatment"), and also includes effects that occur before the subject begins to suffer from a specific disease, disorder, or condition ("prophylactic treatment").
  • the "effective amount" of a compound refers to an amount sufficient to cause a target biological response.
  • the effective amount of the compound of the present invention may vary according to the following factors: for example, the biological target, the pharmacokinetics of the compound, the disease to be treated, the mode of administration, and the subject’s Age health and symptoms.
  • the effective amount includes a therapeutically effective amount and a preventive effective amount.
  • the "therapeutically effective amount” of the compound used herein is an amount sufficient to provide a therapeutic benefit during the treatment of a disease, disorder, or condition, or to cause one or more symptoms associated with the disease, disorder, or condition The amount of delay or minimization.
  • the therapeutically effective amount of a compound refers to the amount of the therapeutic agent when used alone or in combination with other therapies, which provides therapeutic benefits in the treatment of diseases, disorders, or conditions.
  • the term “therapeutically effective amount” may include an amount that improves the overall treatment, reduces or avoids the symptoms or causes of a disease or disorder, or enhances the therapeutic effect of other therapeutic agents.
  • the “prophylactically effective amount” of the compound used herein is an amount sufficient to prevent a disease, disorder, or condition, or an amount sufficient to prevent one or more symptoms related to the disease, disorder, or condition, or prevent a disease , The amount of recurrence of the disorder or condition.
  • the prophylactically effective amount of a compound refers to the amount of the therapeutic agent when used alone or in combination with other agents, which provides a preventive benefit in the process of preventing diseases, disorders or conditions.
  • the term “prophylactically effective amount” may include an amount that improves overall prevention, or an amount that enhances the preventive effect of other preventive agents.
  • Combination and related terms refer to the simultaneous or sequential administration of the compound of the present invention and other therapeutic agents.
  • the compound of the present invention can be administered simultaneously or sequentially in separate unit dosage forms with other therapeutic agents, or simultaneously administered in a single unit dosage form with other therapeutic agents.
  • Figure 1 is a graph showing the effect of compound C176 on the protein level of EGFR L858R/T790M/C797S.
  • Figure 2 is a graph showing the effect of compound C213 on the protein level of EGFR Del19/T790M/C797S.
  • the "compound of the present invention” refers to the following compound of formula (X) (including sub-general formulas, such as formula (I), (I-1), (I-5-1), etc.), which are pharmaceutically acceptable Accepted salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof.
  • the present invention relates to a compound of general formula (X), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, Polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring A is selected from the following optionally substituted groups: C 3-7 cycloalkyl, 4-8 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, wherein the substituent is selected from Halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 3-7 cycloalkyl, 4-8 membered heterocyclic group, -P(O)(C 1 -6 alkyl) 2 , -P(O)(C 2-6 alkenyl) 2 , -OC 1-6 alkyl, -OC 1-6 haloalkyl, -OC 2-6 alkenyl, -OC 3- 7 cycloalkyl, -O-4-8 membered heterocyclic group, -NH-C 1-6 alkyl, -NH-C 2-6 alkenyl, -NH-C 3-7 cycloalkyl, -NH- 4-8 membered heterocyclic group, -
  • Ring B is selected from the following groups:
  • connection position with other parts of the molecule can be located in the available position of the ring
  • Z 1 is an O, S, N or C atom, which is optionally substituted by one or two R Z1 ; or Z 1 is absent, so Z 4 is connected to Z 2 , Z 3 or the aromatic ring connected to Z 1
  • the C atom of, and the Z 2 connected to Z 1 and the C atom on the aromatic ring are respectively connected to R W ; or Z 1 , Z 2 and Z 3 do not exist, so Z 4 is connected to Z 1 or Z 3
  • One of the C atoms on the aromatic ring of, and the other C atom on the aromatic ring is connected to R W;
  • Z 2 is an O, S, N or C atom, which is optionally substituted by one or two R Z2;
  • Z 3 is an O, S, N or C atom, which is optionally substituted by one or two R Z3 ; with the proviso that, when When it is a double bond, Z 2 is a N or C atom, and Z 3 is a N or C atom;
  • Z 4 is N or CR Z4 ;
  • Z 5 is N or CR Z5 ;
  • R N1 is H, C 1-6 alkyl or C 1-6 haloalkyl, preferably H;
  • R Z4 is H, CN, halogen, -(CH 2 ) 0-5 -OR", -(CH 2 ) 0-5 -NR"R"', C 1-6 alkyl or C 1-6 haloalkyl;
  • R Z5 is H, CN, halogen, -(CH 2 ) 0-5 -OR", -(CH 2 ) 0-5 -NR"R"', C 1-6 alkyl, C 1-6 haloalkyl, -(CH 2 ) 0-5 -C 3-7 cycloalkyl or -(CH 2 ) 0-5 -4-8 membered heterocyclic group;
  • R w is H, CN, halogen, -(CH 2 ) 0-5 -OR", -(CH 2 ) 0-5 -NR"R"', -C 1-6 alkyl, C 1-6 haloalkane Group, -(CH 2 ) 0-5 -C 3-7 cycloalkyl, -(CH 2 ) 0-5 -4-8 membered heterocyclyl, C 2-6 alkenyl, C 2-6 alkynyl, -(CH 2 ) 0-5 -C 3-10 halocycloalkyl, -(CH 2 ) 0-5 -C 6-10 aryl or -(CH 2 ) 0-5 -5-14-membered heteroaromatic base;
  • R" is H, C 1-6 alkyl, C 1-6 haloalkyl or -(CH 2 ) 0-5 -C 3-7 cycloalkyl;
  • R"' is H, C 1-6 alkyl or C 1-6 haloalkyl
  • E is independently selected from: chemical bond, -C c R # R # '-C d R # R # '-C e R # R # ',
  • two E units can form -CH 2 CH 2 OCH 2 CH 2 -, -OCH 2 CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 O-,
  • H 1 and H 2 are N or C atoms, H 3 is O, S, N or C atom, and H 1 and H 3 , H 2 and H 3 are not heteroatoms at the same time;
  • H 4 and H 5 are N or C atoms
  • H 6 , H 7 , H 8 and H 9 are C or N atoms
  • p 0, 1 or 2;
  • q 1 or 2;
  • R * is H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, C 3-10 halocycloalkyl , 3-10 membered heterocyclic group, C 6-10 aryl group or 5-14 membered heteroaryl group;
  • R # is H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, C 3-10 halo ring Alkyl, 3-10 membered heterocyclyl, C 6-10 aryl or 5-14 membered heteroaryl;
  • R # ' is H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, C 3-10 halo Cycloalkyl, 3-10 membered heterocyclyl, C 6-10 aryl or 5-14 membered heteroaryl;
  • R # and R # on adjacent atoms can form a chemical bond
  • R # 'and R # ' on adjacent atoms can form a chemical bond
  • n 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10;
  • R s1 is selected from H, CN, halogen, -(CH 2 ) 0-5 -OR", -(CH 2 ) 0-5 -NR"R"', C 1-6 alkyl, C 1-6 haloalkyl , -(CH 2 ) 0-5 -4-8 membered heterocyclyl, C 2-6 alkenyl, C 2-6 alkynyl, -(CH 2 ) 0-5 -C 3-7 cycloalkyl,- (CH 2 ) 0-5 -C 3-10 halocycloalkyl, -(CH 2 ) 0-5 -C 6-10 aryl, -(CH 2 ) 0-5 -5-14 membered heteroaryl , -C(O)R W , -S(O)R W or -S(O) 2 R W ;
  • s1 is 0, 1, 2 or 3;
  • R' is selected from H, -C(O)-C 1-6 alkyl, -C(O)-C 1-6 haloalkyl, -C(O)-C 2-6 alkenyl or -C(O) -C 6-10 aryl;
  • L is a chemical bond, -O- or -NR-;
  • R is H or C 1-6 alkyl
  • T is selected from a chemical bond, a C 2-6 heteroalkylene group, a 4-12 membered heterocyclylene group or a 5-6 membered heterocyclylene group substituted with a 5-6 membered heterocyclylene group;
  • R 1 is selected from H, halogen, cyano, C 1-6 alkyl, C 3-7 cycloalkyl, 4-8 membered heterocyclic group, C 1-6 haloalkyl, C 2-6 alkenyl or C 2 -6 alkynyl;
  • R 1 and R together with the atoms to which they are attached form an optionally substituted 5-6 membered heterocyclic group, wherein the substituent is selected from halogen, oxo, C 1-6 alkane Group, C 1-6 haloalkyl, halogen monosubstituted or polysubstituted C 6-10 aryl;
  • R 2 is H, halogen, hydroxy, amino, C 1-6 alkyl or C 1-6 haloalkyl
  • R 1 and R 2 together with the atoms to which they are connected form a 5-6 membered heterocyclic group or a 5-6 membered heteroaryl group;
  • R 3 is selected from H, -OC 1-6 alkyl or -OC 1-6 haloalkyl
  • R 4 is selected from H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, -NHC(O)-C 1-6 alkyl or -NHC(O)-C 2-6 alkenyl;
  • each of the above groups is H or a H-containing group
  • the one or more H atoms may be substituted by D atoms
  • alkyl, alkylene, haloalkyl, alkenyl, alkynyl, cycloalkyl, halocycloalkyl, heterocyclyl, aryl, heteroaryl or contained in L 1 , E, L 2 , and T OH, NH, NH 2, CH , CH 2, CH 3 groups are each optionally substituted with three or more at each occurrence R s, wherein R s in each occurrence Independently selected from: halogen, hydroxy, amino, cyano, nitro, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 ring Alkyl, C 3-10 halocycloalkyl, 3-10 membered heterocyclic group, C 6-10 aryl, 5-14 membered heteroaryl, C 6-12 aralkyl, -OR a' ,- OC (O) R a ', -C (
  • n is independently 1 or 2 each time it appears;
  • R a'and R b' are each independently selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkyl-O-, C 1-6 alkyl-S-, C 3-10 cycloalkyl, 3-10 membered heterocyclic group, C 6-10 aryl, 5-14 membered heteroaryl, and C 6-12 aralkyl.
  • the present invention relates to a compound of general formula (X), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate thereof , Polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring A is selected from the following optionally substituted groups: C 3-7 cycloalkyl, 4-8 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, wherein the substituent is selected from Halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 3-7 cycloalkyl, 4-8 membered heterocyclic group, -P(O)(C 1 -6 alkyl) 2 , -P(O)(C 2-6 alkenyl) 2 , -OC 1-6 alkyl, -OC 1-6 haloalkyl, -OC 2-6 alkenyl, -OC 3- 7 cycloalkyl, -O-4-8 membered heterocyclic group, -NH-C 1-6 alkyl, -NH-C 2-6 alkenyl, -NH-C 3-7 cycloalkyl, -NH- 4-8 membered heterocyclic group, -
  • Ring B is selected from the following groups:
  • R' is selected from H, -C(O)-C 1-6 alkyl, -C(O)-C 1-6 haloalkyl, -C(O)-C 2-6 alkenyl or -C(O) -C 6-10 aryl;
  • L is a chemical bond, -O- or -NR-;
  • R is H or C 1-6 alkyl
  • T is selected from a chemical bond, a C 2-6 heteroalkylene group, a 4-12 membered heterocyclylene group or a 5-6 membered heterocyclylene group substituted with a 5-6 membered heterocyclylene group;
  • n 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10;
  • L 2 is selected from chemical bond, -CH 2 -, -CH 2 CH 2 -, -OCH 2 -, -NHCH 2 -, -OC(O)-, -NHC(O)-, -CH 2 C(O)- Or -C(O)CH 2 -;
  • R 1 is selected from H, halogen, cyano, C 1-6 alkyl, C 3-7 cycloalkyl, 4-8 membered heterocyclic group, C 1-6 haloalkyl, C 2-6 alkenyl or C 2 -6 alkynyl;
  • R 1 and R together with the atoms to which they are attached form an optionally substituted 5-6 membered heterocyclic group, wherein the substituent is selected from halogen, oxo, C 1-6 alkane Group, C 1-6 haloalkyl, halogen monosubstituted or polysubstituted C 6-10 aryl;
  • R 2 is H, halogen, hydroxy, amino, C 1-6 alkyl or C 1-6 haloalkyl
  • R 1 and R 2 together with the atoms to which they are connected form a 5-6 membered heterocyclic group or a 5-6 membered heteroaryl group;
  • R 3 is selected from H, -OC 1-6 alkyl or -OC 1-6 haloalkyl
  • R 4 is selected from H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, -NHC(O)-C 1-6 alkyl or -NHC(O)-C 2-6 alkenyl;
  • alkyl, alkylene, alkenyl, alkynyl, cycloalkyl, heterocyclic, aryl, heteroaryl or L 1 , E, L 2 , and T contain OH, NH, NH 2 , CH, CH 2, CH 3 groups are each optionally substituted with three or more at each occurrence R s, wherein R s is independently at each occurrence selected from: halo, hydroxy, amino , Cyano, nitro, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, C 3-10 halo ring Alkyl, 3-10 membered heterocyclyl, C 6-10 aryl, 5-14 membered heteroaryl, C 6-12 aralkyl, -OR a , -OC(O)R a , -C(O )R a , -C(O)OR a
  • n is independently 1 or 2 each time it appears;
  • Each occurrence of R a and R b is independently selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkyl-O-, C 1 -6 alkyl-S-, C 3-10 cycloalkyl, 3-10 membered heterocyclic group, C 6-10 aryl, 5-14 membered heteroaryl, and C 6-12 aralkyl.
  • the present invention relates to compounds of formula (I) or (IG), or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates thereof Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • the present invention relates to compounds of general formula (I) or (IG), or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvents thereof Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring A is the following group:
  • R 7 is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 3-7 cycloalkyl, -P (O) (C 1-6 alkyl) 2 or -P (O)(C 2-6 alkenyl) 2 ;
  • R 8 is selected from H, -NH-C 1-6 alkyl, -NH-C 2-6 alkenyl, -NH-C 3-7 cycloalkyl, -NH-4-8 membered heterocyclic group, -NHC (O)-C 1-6 alkyl, -NHC(O)-C 2-6 alkenyl, -NHC(O)-C 3-7 cycloalkyl, -NHC(O)-4-8 membered heterocycle Group, -NHS(O) 2 -C 1-6 alkyl or -NHS(O) 2 -C 3-7 cycloalkyl;
  • R 9 is selected from H, halogen, -CN, C 1-6 haloalkyl, -OC 1-6 alkyl, -OC 1-6 haloalkyl, C 3-7 cycloalkyl, -OC 3-7 cycloalkyl , C 1-6 alkyl, -NH-C 1-6 alkyl, -NH-C 2-6 alkenyl, -NH-C 3-7 cycloalkyl, -NH-4-8 membered heterocyclic group, -NHC(O)-C 1-6 alkyl, -NHC(O)-C 2-6 alkenyl, -NHC(O)-C 3-7 cycloalkyl, -NHC(O)-4-8 member Heterocyclic group, -NHS(O) 2 -C 1-6 alkyl or -NHS(O) 2 -C 3-7 cycloalkyl;
  • R x is H, or R x together with R 8 and the C atom to which they are connected form a 5-6 membered heterocyclic group or a 5-6 membered heteroaryl group; preferably, R x together with R 8 and their connected The C atoms of together form a 5-6 membered heteroaryl group, preferably a pyrazinyl group;
  • X 1 is -CH(R X1 )- or -N(R X1 )-;
  • X 2 is -CH(R X2 )- or -N(R X2 )-;
  • R X1 is selected from H, C 1-6 alkyl, C 3-7 cycloalkyl, -OC 1-6 alkyl, -OC 3-7 cycloalkyl, -NH-C 1-6 alkyl, -NH-C 3-7 cycloalkyl, -S(O) 2 -C 1-6 alkyl, -S(O) 2 -C 3-7 cycloalkyl, -NHS(O) 2 -C 1- 6 alkyl, -NHS(O) 2 -C 3-7 cycloalkyl, -C(O)-C 1-6 alkyl, -C(O)-C 2-6 alkenyl, -C(O) -C 3-7 cycloalkyl, -NHC(O)-C 1-6 alkyl, -NHC(O)-C 3-7 cycloalkyl or -C(O)-4-8 membered heterocyclic group; R X2 is selected from H, C 1-6 alky
  • the present invention relates to compounds of general formula (I) or (IG), or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvents thereof Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • R 5 is H or C 1-6 alkyl
  • R 6 is H or C 1-6 alkyl
  • R 5 and R 6 are connected to form a C 1-6 alkylene group
  • the present invention relates to compounds of general formula (I) or (IG), or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvents thereof Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • R N is selected from C 1-6 alkyl or
  • R 11 is H or halogen
  • R 12 is H or halogen
  • R 13 is H or halogen
  • the present invention relates to compounds of general formula (I) or (IG), or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvents thereof Compounds, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • R 1 and R 2 together with the atoms to which they are connected form a 5-6 membered heteroaryl group; preferably form a pyrrolyl group; other groups are as defined above.
  • ring A is an optionally substituted C 3-7 cycloalkyl; in another specific embodiment, ring A is an optionally substituted 4-8 membered heterocyclyl; in another specific embodiment
  • ring A is an optionally substituted C 6-10 aryl group; in another specific embodiment, ring A is an optionally substituted 5-10 membered heteroaryl group; in another specific embodiment, ring A for In another specific embodiment, ring A is In another specific embodiment, ring A is In another specific embodiment, ring A is
  • the substituent of ring A is halogen; in another of the above embodiments, the substituent of ring A is -CN; in another of the above embodiments, the substituent of ring A is C 1-6 alkyl; in another above-mentioned specific embodiment, the substituent of ring A is C 1-6 haloalkyl; in another above-mentioned specific embodiment, the substituent of ring A is C 2-6 alkenyl; In another above-mentioned specific embodiment, the substituent of ring A is C 3-7 cycloalkyl; in another above-mentioned specific embodiment, the substituent of ring A is 4-8 membered heterocyclic group; in another above-mentioned In a specific embodiment, the substituent of ring A is -P(O)(C 1-6 alkyl) 2 ; in another specific embodiment described above, the substituent of ring A is -P(O)(C 2- 6 Alkenyl) 2 ; In another above-mentioned
  • L is a chemical bond; in another specific embodiment, L is -O-; in another specific embodiment, L is -NR-.
  • Y is -CH 2 -; in another specific embodiment, Y is -C(O)-.
  • T is C 2-6 heteroalkylene; in another specific embodiment, T is 4-12 membered heterocyclylene; in another specific embodiment, T is 5-6 5-6 membered heterocyclylene substituted with membered heterocyclylene; in another specific embodiment, T is In another specific embodiment, T is In another specific embodiment, T is In another specific embodiment, T is In another specific embodiment, T is In another specific embodiment, T is a chemical bond.
  • Z 1 is an O atom; in another specific embodiment, Z 1 is an S atom; in another specific embodiment, Z 1 is a N atom; in another specific embodiment, Z 1 is a C atom; in another specific embodiment, Z 1 is substituted by one R Z1 ; in another specific embodiment, Z 1 is substituted by two R Z1 ; in another specific embodiment, Z 1 is not present .
  • Z 2 is an O atom; in another specific embodiment, Z 2 is an S atom; in another specific embodiment, Z 2 is a N atom; in another specific embodiment, Z 2 is a C atom; in another specific embodiment, Z 2 is substituted by one R Z2 ; in another specific embodiment, Z 2 is substituted by two R Z2.
  • Z 3 is an O atom; in another specific embodiment, Z 3 is an S atom; in another specific embodiment, Z 3 is a N atom; in another specific embodiment, Z 3 is a C atom; in another specific embodiment, Z 3 is substituted by one R Z3 ; in another specific embodiment, Z 3 is substituted by two R Z3.
  • none of Z 1 , Z 2 and Z 3 are present.
  • Z 4 is N; in another specific embodiment, Z 4 is CR Z4 .
  • Z 5 is N; in another specific embodiment, Z 5 is CR Z5 .
  • R a is H; In another particular embodiment, R a is a halogen; In another particular embodiment, R a is OR '; In another particular embodiment, R a is NR'R "; in another particular embodiment, R a is C 1-6 alkyl; in another particular embodiment, R a is C 1-6 haloalkyl.
  • R b is H; in another specific embodiment, R b is halogen; in another specific embodiment, R b is OR'; in another specific embodiment, R b is NR'R"; In another specific embodiment, R b is C 1-6 alkyl; in another specific embodiment, R b is C 1-6 haloalkyl.
  • R c is H; in another specific embodiment, R c is halogen; in another specific embodiment, R c is OR'; in another specific embodiment, R c is NR'R"; in another specific embodiment, R c is C 1-6 alkyl; in another specific embodiment, R c is C 1-6 haloalkyl.
  • R Z4 is H; in another specific embodiment, R Z4 is CN; in another specific embodiment, R Z4 is halogen; in another specific embodiment, R Z4 is- (CH 2 ) 0-5 -OR'; in another specific embodiment, R Z4 is -(CH 2 ) 0-5 -NR'R"; in another specific embodiment, R Z4 is C 1- 6 Alkyl; In another specific embodiment, R Z4 is C 1-6 haloalkyl.
  • R Z5 is H; in another specific embodiment, R Z5 is CN; in another specific embodiment, R Z5 is halogen; in another specific embodiment, R Z5 is- (CH 2 ) 0-5 -OR'; in another specific embodiment, R Z5 is -(CH 2 ) 0-5 -NR'R"; in another specific embodiment, R Z5 is C 1- 6 alkyl; in another specific embodiment, R Z5 is C 1-6 haloalkyl; in another specific embodiment, R Z5 is -(CH 2 ) 0-5 -C 3-7 cycloalkyl; In another specific embodiment, R Z5 is -(CH 2 ) 0-5 -4-8 membered heterocyclyl.
  • the ring where Z 4 is located does not exist.
  • E is a chemical bond; in another specific embodiment, E is -C c R # R # '-C d R # R # '-C e R # R # '; in another specific embodiment In the embodiment, E is In another specific embodiment, E is In another specific embodiment, E is In another specific embodiment, E is In another specific embodiment, E is In another specific embodiment, E is In another specific embodiment, E is In another specific embodiment, E is a chemical bond; in another specific embodiment, E is -C c R # R # '-C d R # R # '-C e R # R # '; in another specific embodiment In the embodiment, E is In another specific embodiment, E is In another specific embodiment, E is In another specific embodiment, E is In another specific embodiment, E is In another specific embodiment, E is
  • two E units can form -CH 2 CH 2 OCH 2 CH 2 -; in another specific embodiment, two E units can form -OCH 2 CH 2 CH 2 CH 2 -; in another specific embodiment, two E units can form -CH 2 CH 2 CH 2 CH 2 O-; in another specific embodiment, two E units can form In another specific embodiment, two E units can form In another specific embodiment, two E units can form In another specific embodiment, two E units can form In another specific embodiment, two E units can form In another specific embodiment, two E units can form In another specific embodiment, two E units can form In another specific embodiment, two E units can form in another specific embodiment, two E units can form, two E units can form
  • R # and R # on adjacent atoms can form a chemical bond, and R # 'and R # ' on adjacent atoms can form a chemical bond ;
  • m is 0; in another specific embodiment, m is 1; in another specific embodiment, m is 2; in another specific embodiment, m is 3; in another In a specific embodiment, m is 4; in another specific embodiment, m is 5; in another specific embodiment, m is 6; in another specific embodiment, m is 7; in another specific embodiment In the scheme, m is 8; in another specific embodiment, m is 9; in another specific embodiment, m is 10.
  • R 1 is H; in another specific embodiment, R 1 is halogen; in another specific embodiment, R 1 is cyano; in another specific embodiment, R 1 is C 1-6 alkyl; in another embodiment, R 1 is C 3-7 cycloalkyl; in another embodiment, R 1 is 4-8 membered heterocyclyl; in another embodiment In the scheme, R 1 is C 1-6 haloalkyl; in another specific embodiment, R 1 is C 2-6 alkenyl; in another specific embodiment, R 1 is C 2-6 alkynyl.
  • R 2 is H; in another specific embodiment, R 2 is halogen; in another specific embodiment, R 2 is hydroxy; in another specific embodiment, R 2 is amino ; In another specific embodiment, R 2 is C 1-6 alkyl; in another specific embodiment, R 2 is C 1-6 haloalkyl; in another specific embodiment, R 1 , R 2 Together with the atoms to which they are attached, form a 5-6 membered heterocyclic group; in another specific embodiment, R 1 , R 2 and the atoms to which they are attached form a 5-6 membered heteroaryl group; in another specific embodiment , R 1 and R 2 form a pyrrolyl group together with the atoms to which they are attached.
  • R 3 is H; in another specific embodiment, R 3 is HOC 1-6 alkyl; in another specific embodiment, R 3 is -OC 1-6 haloalkyl.
  • R 4 is H; in another specific embodiment, R 4 is halogen; in another specific embodiment, R 4 is C 1-6 alkyl; in another specific embodiment , R 4 is C 1-6 haloalkyl; in another specific embodiment, R 4 is -NHC(O)-C 1-6 alkyl; in another specific embodiment, R 4 is -NHC(O ) -C 2-6 alkenyl.
  • any technical solution or any combination of any of the above specific embodiments can be combined with any technical solution or any combination of other specific embodiments.
  • any technical solution or any combination of ring A can be combined with any of L, Y, Z 1 -Z 5 , R', T, L 1 , E, m, L 2 , R 1 -R 4, etc.
  • Technical solutions or any combination thereof The present invention is intended to include all the combinations of these technical solutions, limited to space, and will not be listed one by one.
  • the present invention provides a compound of general formula (I) or (IG), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, Solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, in which,
  • R 7 is selected from -Me, cyclopropyl or -P(O)Me 2 ;
  • R 8 is selected from H, -NHMe, -NHC(O)CH 2 CH 3 or -NH-cyclopropyl;
  • R x is H, or R x together with R 8 and the C atom to which they are connected form a 5-6 membered heterocyclic group or a 5-6 membered heteroaryl group; preferably a 5-6 membered heteroaryl group (preferably Pyrazinyl);
  • X 1 is -CH 2 -or -N(R X1 )-;
  • X 2 is -CH(R X2 )- or -N(R X2 )-;
  • R X2 is H, -Me, -OMe, -NHMe, -C(O)CH 2 CH 3 ,- NHS(O) 2 CH 2 CH 3 or -NHC(O)CH 2 CH 3 ;
  • T is selected from the group consisting of chemical bond, C 2-6 heteroalkylene, 4-12 membered heterocyclylene, or 5-6 membered heterocyclylene substituted with 5-6 membered heterocyclylene; preferably the following groups:
  • R 5 is -Me
  • R 6 is -Me
  • R 5 and R 6 are connected to form -CH 2 CH 2 -;
  • R N is selected from -iPr, -Et or
  • R 11 is -Cl, -Br
  • R 12 is H or -F
  • R 13 is H or -F
  • R 2 is H
  • R 1 and R 2 together with the atoms to which they are connected form a 5-6 membered heterocyclic group or a 5-6 membered heteroaryl group; preferably form a pyrrolyl group;
  • R 3 is selected from H or -OMe
  • n 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10;
  • L 2 is selected from chemical bond, -CH 2 -, -CH 2 CH 2 -, -OCH 2 -, -NHCH 2 -, -OC(O)-, -NHC(O)-, -CH 2 C(O)- Or -C(O)CH 2 -;
  • the present invention provides a compound of general formula (I), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, Hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein the compound of general formula (I) has the following general structure:
  • the present invention provides general formulas (I-1), (I-1-A), (I-1-B), (I-1-C), (I-1-D ), (I-1-E), (I-1-F), (I-1-G), (I-1-H) or (I-1-I) compound, or pharmaceutically acceptable Salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants of, and mixtures thereof, wherein,
  • R 4 is H or -Me
  • R 8 is H, -NHMe, -NHC(O)CH 2 CH 3 or -NH-cyclopropyl
  • R x is H, or R x together with R 8 and the C atom to which they are attached form a pyrazinyl group;
  • the present invention provides a compound of general formula (IG), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, or solvate thereof , Hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • Ring A is the following group:
  • R 7 is -P(O)(C 1-6 alkyl) 2 ;
  • R 8 is H
  • R 9 is selected from H, halogen, C 1-6 alkyl, -CN or C 1-6 haloalkyl;
  • R x is H, or R x together with R 8 and the C atom to which they are connected form a 5-6 membered heteroaryl group;
  • it is pyrazinyl
  • L 1 is -C a R # R # '-C b R # R # '-;
  • L 2 is selected from chemical bond, -CR # R # '- or -C a R # R # '-C b R # R # '-;
  • C a R # R # 'or C b R # R # ' can be replaced by O, S(O) p or NR # ;
  • E is independently selected from: chemical bond or -C c R # R # '-C d R # R # '-C e R # R # ';
  • C c R # R # ', C d R # R # 'or C e R # R # ', or C c R # R # R # ' and C e R # R # ' both can be O, S (O) p or NR # replacement;
  • p 0, 1 or 2;
  • R # is H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R # ' is H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R # and R # on adjacent atoms can form a chemical bond
  • R # 'and R # ' on adjacent atoms can form a chemical bond
  • n 0, 1, 2, 3, 4 or 5;
  • R s1 is selected from H, CN, halogen, OH, NH 2 , C 2-6 alkenyl, C 2-6 alkynyl, -OC 1-6 alkyl, -OC 1-6 haloalkyl, -NH-C 1 -6 alkyl, C 1-6 alkyl or C 1-6 haloalkyl;
  • s1 is 0, 1, 2 or 3;
  • L is -NR-; wherein R is H or C 1-6 alkyl;
  • R 1 is selected from H, halogen, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl;
  • R 2 is H, halogen, hydroxy, amino, C 1-6 alkyl or C 1-6 haloalkyl
  • R 3 is selected from H, -OC 1-6 alkyl or -OC 1-6 haloalkyl
  • R 4 is selected from H, halogen, C 1-6 alkyl or C 1-6 haloalkyl
  • each of the above groups is H or a H-containing group
  • the one or more H atoms may be substituted by D atoms.
  • the present invention provides compounds of general formula (I-1-G), (I-1-H), (I-1-H'), (I-1-H”), Or its pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof:
  • R 1 is selected from H, halogen, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl; preferably, R 1 is H or halogen;
  • R 4 is selected from H, halogen, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R 4 is H;
  • R 8 is H
  • R 9 is selected from H, halogen, C 1-6 alkyl, -CN or C 1-6 haloalkyl; preferably, R 9 is H;
  • L 2 is selected from a chemical bond, -CH 2 -or -CH 2 CH 2 -, and one or more H atoms in the above groups may be substituted by D atoms;
  • E is -CH 2 CH 2 CH 2 -, -CH 2 CH 2 O-, -CH 2 OCH 2 -, -OCH 2 CH 2 -, -CH 2 CH 2 S-, -CH 2 SCH 2 -or -SCH 2 CH 2 -, and one or more H atoms in the above groups may be substituted by D atoms; preferably, E is -CH 2 CH 2 CH 2 -;
  • n 0, 1, 2 or 3; preferably, the chain length of -L 1 -(E) m -L 2 -is preferably 4-14 bond lengths, more preferably 5, 6, 7, 8, 9 or 10 key lengths;
  • R s1 is selected from H, CN, halogen, OH, NH 2 , C 2-6 alkenyl, C 2-6 alkynyl, -OC 1-6 alkyl, -OC 1-6 haloalkyl, -NH-C 1 -6 alkyl, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R s1 is H, CN or halogen;
  • s1 is 0, 1, or 2.
  • the present invention provides compounds of general formula (I-1-G), (I-1-H), (I-1-H'), (I-1-H”), Or its pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof:
  • R 1 is selected from H, halogen, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl; preferably, R 1 is halogen;
  • R 4 is selected from H, halogen, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R 4 is H;
  • R 8 is H
  • R 9 is selected from H, halogen, C 1-6 alkyl, -CN or C 1-6 haloalkyl; preferably, R 9 is H;
  • L 2 is selected from a chemical bond, -CH 2 -or -CH 2 CH 2 -, and one or more H atoms in the above groups may be substituted by D atoms;
  • E is -CH 2 CH 2 CH 2 -, -CH 2 CH 2 O-, -CH 2 OCH 2 -, -OCH 2 CH 2 -, -CH 2 CH 2 S-, -CH 2 SCH 2 -or -SCH 2 CH 2 -, and one or more H atoms in the above groups may be substituted by D atoms; preferably, E is -CH 2 CH 2 CH 2 -;
  • n 0, 1, 2 or 3; preferably, the chain length of -L 1 -(E) m -L 2 -is preferably 4-14 bond lengths, more preferably 5, 6, 7, 8, 9 or 10 key lengths;
  • R s1 is selected from H, CN, halogen, OH, NH 2 , C 2-6 alkenyl, C 2-6 alkynyl, -OC 1-6 alkyl, -OC 1-6 haloalkyl, -NH-C 1 -6 alkyl, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R s1 is H or halogen;
  • s1 is 0, 1, or 2.
  • the present invention provides compounds of general formula (I-1-G), (I-1-H), (I-1-H'), (I-1-H”), Or its pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof:
  • R 1 is selected from H, halogen, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl; preferably, R 1 is halogen;
  • R 4 is selected from H, halogen, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R 4 is H;
  • R 8 is H
  • R 9 is selected from H, halogen, C 1-6 alkyl, -CN or C 1-6 haloalkyl; preferably, R 9 is H;
  • L 2 is selected from a chemical bond, -CH 2 -or -CH 2 CH 2 -, and one or more H atoms in the above groups may be substituted by D atoms;
  • E is -CH 2 CH 2 CH 2 -, -CH 2 CH 2 O-, -CH 2 OCH 2 -, -OCH 2 CH 2 -, -CH 2 CH 2 S-, -CH 2 SCH 2 -or -SCH 2 CH 2 -, and one or more H atoms in the above groups may be substituted by D atoms; preferably, E is -CH 2 CH 2 CH 2 -;
  • m is 1, 2 or 3; preferably, the chain length of -L 1 -(E) m -L 2 -is less than 14 bond lengths; preferably, the chain length is 5-10 bond lengths, preferably 5, 6, 7, 8, 9 or 10 bond lengths;
  • R s1 is selected from H, CN, halogen, OH, NH 2 , C 2-6 alkenyl, C 2-6 alkynyl, -OC 1-6 alkyl, -OC 1-6 haloalkyl, -NH-C 1 -6 alkyl, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R s1 is H or halogen;
  • s1 is 0, 1, or 2.
  • the present invention provides formulas (I-1-I), (I-1-I'), (I-1-I"), (I-1-I"') Compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants thereof, and their mixture:
  • R 1 is selected from H, halogen, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl; preferably, R 1 is halogen;
  • R 4 is selected from H, halogen, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R 4 is selected from C 1-6 alkyl or C 1-6 haloalkyl;
  • R 9 is selected from H, halogen, C 1-6 alkyl, -CN or C 1-6 haloalkyl; preferably, R 9 is H;
  • L 2 is selected from a chemical bond, -CH 2 -or -CH 2 CH 2 -;
  • E is -CH 2 CH 2 CH 2 -, -CH 2 CH 2 O-, -CH 2 OCH 2 -, -OCH 2 CH 2 -, -CH 2 CH 2 S-, -CH 2 SCH 2 -or -SCH 2 CH 2 -;
  • E is -CH 2 CH 2 CH 2 -;
  • m is 1 or 2; preferably, the chain length of -L 1 -(E) m -L 2 -is 4-14 bond lengths, more preferably 5, 6, 7, 8, 9 or 10 bond lengths;
  • R s1 is selected from H, CN, halogen, OH, NH 2 , C 2-6 alkenyl, C 2-6 alkynyl, -OC 1-6 alkyl, -OC 1-6 haloalkyl, -NH-C 1 -6 alkyl, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R s1 is H, halogen or CN;
  • s1 is 0, 1, or 2.
  • the present invention provides formulas (I-1-I), (I-1-I'), (I-1-I"), (I-1-I"') Compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants thereof, and their mixture:
  • R 1 is selected from H, halogen, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl; preferably, R 1 is halogen;
  • R 4 is selected from H, halogen, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R 4 is selected from C 1-6 alkyl or C 1-6 haloalkyl;
  • R 9 is selected from H, halogen, C 1-6 alkyl, -CN or C 1-6 haloalkyl; preferably, R 9 is H;
  • L 2 is selected from a chemical bond, -CH 2 -or -CH 2 CH 2 -;
  • E is -CH 2 CH 2 CH 2 -;
  • m is 1 or 2; preferably, the chain length of -L 1 -(E) m -L 2 -is less than 14 bond lengths; preferably, 5, 6, 7, 8, 9 or 10 bond lengths are more preferred;
  • R s1 is selected from H, CN, halogen, OH, NH 2 , C 2-6 alkenyl, C 2-6 alkynyl, -OC 1-6 alkyl, -OC 1-6 haloalkyl, -NH-C 1 -6 alkyl, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R s1 is H or halogen;
  • s1 is 0, 1, or 2.
  • the present invention provides formulas (I-2-I), (I-2-I'), (I-2-I"), (I-2-I"') Compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants thereof, and their mixture:
  • R 1 is selected from H, halogen, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl or C 2-6 alkynyl; preferably, R 1 is halogen;
  • R 4 is selected from H, halogen, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R 4 is selected from C 1-6 alkyl or C 1-6 haloalkyl;
  • R 9 is selected from H, halogen, C 1-6 alkyl, -CN or C 1-6 haloalkyl; preferably, R 9 is H;
  • L 2 is selected from a chemical bond, -CH 2 -or -CH 2 CH 2 -;
  • E is -CH 2 CH 2 CH 2 -, -CH 2 CH 2 O-, -CH 2 OCH 2 -, -OCH 2 CH 2 -, -CH 2 CH 2 S-, -CH 2 SCH 2 -or -SCH 2 CH 2 -;
  • E is -CH 2 CH 2 CH 2 -;
  • m is 1 or 2; preferably, the chain length of -L 1 -(E) m -L 2 -is less than 14 bond lengths; preferably 5, 6, 7, 8, 9 or 10 bond lengths;
  • R s1 is selected from H, CN, halogen, C 1-6 alkyl or C 1-6 haloalkyl; preferably, R s1 is H or halogen;
  • s1 is 0, 1, or 2.
  • the present invention provides general formulas (I-2), (I-2-A), (I-2-B), (I-2-C), (I-2-D ), (I-2-E), (I-2-F), (I-2-G), (I-2-H) or (I-2-I) compounds, or pharmaceutically acceptable Salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants of, and mixtures thereof, wherein,
  • R 4 is H or -Me
  • R 8 is H, -NHMe, -NHC(O)CH 2 CH 3 or -NH-cyclopropyl
  • R x is H, or R x together with R 8 and the C atom to which they are attached form a pyrazinyl group;
  • the present invention provides general formulas (I-3), (I-3-A), (I-3-B), (I-3-C), (I-3-D ), (I-3-E), (I-3-F), (I-3-G) or (I-3-H) compound, or a pharmaceutically acceptable salt or enantiomer thereof , Diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • R 5 is -Me
  • R 6 is -Me
  • R 5 and R 6 are connected to form -CH 2 CH 2 -;
  • R 7 is -Me or cyclopropyl
  • the present invention provides general formulas (I-4), (I-4-A), (I-4-B), (I-4-C), (I-4-D ), (I-4-E), (I-4-F), (I-4-G) or (I-4-H) compound, or a pharmaceutically acceptable salt or enantiomer thereof , Diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • R 1 is -CF 3 ;
  • the present invention provides general formulas (I-5), (I-5-A), (I-5-B), (I-5-C), (I-5-D ), (I-5-E), (I-5-F), (I-5-G) or (I-5-H), or a pharmaceutically acceptable salt or enantiomer thereof , Diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • X 1 is -CH 2 -or -N(R X1 )-;
  • X 2 is -CH(R X2 )- or -N(R X2 )-;
  • R X2 is H, -Me, -OMe, -NHMe, -NHS(O) 2 CH 2 CH 3 , -C(O)CH 2 CH 3 or -NHC(O)CH 2 CH 3 ;
  • R 1 and R together with the atoms to which they are attached form an optionally substituted 5-6 membered heterocyclic group, wherein the substituent is selected from halogen, oxo, -iPr, -Et, halogen mono- or poly-substituted phenyl ;
  • R N is selected from -iPr, -Et or
  • R 11 is -Cl, -Br
  • R 12 is H or -F
  • R 13 is H or -F
  • R 3 is H or -OMe
  • R 4 is H or -Me
  • the present invention provides a compound of general formula (I-5-1) or (I'-5-1), or a pharmaceutically acceptable salt, enantiomer, or diastereomer Isomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • R N is -iPr or -Et
  • the present invention provides a compound of general formula (I-5-2) or (I'-5-2), or a pharmaceutically acceptable salt, enantiomer, or diastereomer Isomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • R 3 is H or -OMe
  • R 4 is H or -Me
  • the present invention provides a compound of general formula (I-5-3) or (I'-5-3), or a pharmaceutically acceptable salt, enantiomer, or diastereomer Isomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and their mixtures:
  • X 1 is -CH 2 -or -N(R X1 )-;
  • X 2 is -CH(R X2 )- or -N(R X2 )-;
  • R X1 is -C(O)CH 2 CH 3 ;
  • R X2 is H, -Me, -OMe, -NHMe, -C(O)CH 2 CH 3 , -NHS(O) 2 CH 2 CH 3 or -NHC(O)CH 2 CH 3 ;
  • R 11 is -Cl or -Br
  • R 12 is H or -F
  • R 13 is H or -F
  • the present invention provides general formulas (I-6), (I-6-A), (I-6-B), (I-6-C), (I-6-D ), (I-6-E), (I-6-F), (I-6-G) or (I-6-H), or a pharmaceutically acceptable salt or enantiomer thereof , Diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein,
  • L is -O- or -NH-
  • R 3 is H or -OMe
  • R 4 is H or -F
  • the present invention relates to all of the above general formula compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates thereof Compounds, polymorphs, prodrugs or isotopic variants, and their mixtures,
  • H atoms in the above groups may be substituted by D atoms.
  • the present invention relates to all of the above general formula compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates thereof Compounds, polymorphs, prodrugs or isotopic variants, and their mixtures,
  • H atoms in the above groups may be substituted by D atoms.
  • the present invention relates to all of the above general formula compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates thereof Compounds, polymorphs, prodrugs or isotopic variants, and their mixtures,
  • two E units, or two E'units can form -CH 2 CH 2 OCH 2 CH 2 -, -OCH 2 CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 O-,
  • H atoms in the above groups may be substituted by D atoms.
  • the present invention relates to all of the above general formula compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates thereof Compounds, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein m is 0, 1, 4, 5, 6, 7, 8, 9 or 10; preferably, m is 0, 1, 4, 5, 6, 7, or 8; preferably, m is 0, 1, 4, 5, or 6; preferably, m is 0, 1, 4, or 5.
  • the present invention relates to all of the above general formula compounds, or pharmaceutically acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates thereof Compounds, polymorphs, prodrugs or isotopic variants, and mixtures thereof, wherein the chain length of -L 1 -(E) m -L 2 -is 4 to 14 bond lengths; preferably, the chain length is less than 12 Bond length; preferably, the chain length is 5-10 bond lengths; preferably, the chain length is 5, 6, 7, 8, 9 or 10 bond lengths.
  • the present invention relates to a compound, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, poly Crystal forms, prodrugs or isotopic variants, and mixtures thereof, the compound is selected from the following:
  • the compounds of the present invention may include one or more asymmetric centers, and thus may exist in various stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms.
  • the compounds of the present invention may be individual enantiomers, diastereomers, or geometric isomers (such as cis and trans isomers), or may be in the form of a mixture of stereoisomers, Including racemate mixtures and mixtures rich in one or more stereoisomers.
  • the isomers can be separated from the mixture by methods known to those skilled in the art, including: chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts; or the preferred isomers can be separated by Prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • an organic compound can form a complex with a solvent, which reacts in the solvent or precipitates or crystallizes out of the solvent. These complexes are called “solvates”. When the solvent is water, the complex is called “hydrate”. The present invention covers all solvates of the compounds of the present invention.
  • solvate refers to a compound or a salt form thereof combined with a solvent, usually formed by a solvolysis reaction. This physical association may include hydrogen bonding.
  • solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether and the like.
  • Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric solvates and non-stoichiometric solvates. In some cases, the solvate will be able to separate, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid.
  • “Solvate” includes both solvates in solution and separable solvates. Representative solvates include hydrates, ethanolates, and methanolates.
  • hydrate refers to a compound that binds to water. Generally, the ratio of the number of water molecules contained in the hydrate of a compound to the number of molecules of the compound in the hydrate is determined. Therefore, a hydrate of a compound can be represented by, for example, the general formula R ⁇ x H 2 O, where R is the compound, and x is a number greater than zero.
  • a given compound can form more than one type of hydrate, including, for example, monohydrate (x is 1), lower hydrate (x is a number greater than 0 and less than 1, for example, hemihydrate (R ⁇ 0.5 H 2 O)) and polyhydrates (x is a number greater than 1, for example, dihydrate (R ⁇ 2 H 2 O) and hexahydrate (R ⁇ 6 H 2 O)).
  • monohydrate x is 1
  • lower hydrate x is a number greater than 0 and less than 1, for example, hemihydrate (R ⁇ 0.5 H 2 O)
  • polyhydrates x is a number greater than 1, for example, dihydrate (R ⁇ 2 H 2 O) and hexahydrate (R ⁇ 6 H 2 O)).
  • the compounds of the invention may be in amorphous or crystalline form (polymorphs).
  • the compounds of the present invention may exist in one or more crystalline forms. Therefore, the present invention includes all amorphous or crystalline forms of the compounds of the present invention within its scope.
  • polymorph refers to a crystalline form (or a salt, hydrate or solvate thereof) of a compound in a specific crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, photoelectric properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors can cause one crystalline form to dominate.
  • Various polymorphs of the compound can be prepared by crystallization under different conditions.
  • the present invention also includes isotopically-labeled compounds (isotopic variants), which are equivalent to those described in formula (I), but one or more atoms whose atomic mass or mass number is different from the atomic mass or mass number common in nature Replaced.
  • isotopes that can be introduced into the compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, and 18 respectively. O, 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • Tritium, i.e. 3 H and carbon-14, i.e. 14 C isotopes are particularly preferred because they are easy to prepare and detect. Further, substituted with heavier isotopes such as deuterium, i.e.
  • Isotopically-labeled compounds of formula (I) of the present invention and their prodrugs can generally be prepared in this way.
  • readily available isotope-labeled reagents are used instead of non-isotopes. Labeled reagents.
  • prodrugs are also included in the context of the present invention.
  • the term "prodrug” as used herein refers to a compound that is converted into its active form with a medical effect by, for example, hydrolysis in the blood in the body.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, ASSymposium Series Vol. 14, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and D. Fleisher, S. Ramon, and H. Barbra "Improved oral drug delivery: solubility limits overcome by the use of prodrugs", Advanced Drug Delivery Reviews (1996) 19(2) 115-130, each introduced This article serves as a reference.
  • a prodrug is any covalently bonded compound of the invention, and when such a prodrug is administered to a patient, it releases the parent compound in the body.
  • Prodrugs are usually prepared by modifying functional groups, and the modification is performed in such a way that the modification can be performed by conventional operations or cleavage in vivo to produce the parent compound.
  • Prodrugs include, for example, the compounds of the present invention in which a hydroxyl, amino, or sulfhydryl group is bonded to any group, which can be cleaved to form a hydroxyl, amino, or sulfhydryl group when administered to a patient.
  • prodrugs include, but are not limited to, acetate/amide, formate/amide, and benzoate/amide derivatives of the hydroxyl, sulfhydryl, and amino functional groups of the compound of formula (I).
  • esters such as methyl esters, ethyl esters, and the like can be used.
  • the ester itself can be active and/or can be hydrolyzed under human body conditions.
  • Suitable pharmaceutically acceptable in vivo hydrolyzable ester groups include those groups that are easily decomposed in the human body to release the parent acid or salt thereof.
  • the present invention also provides a pharmaceutical preparation comprising a therapeutically effective amount of a compound of formula (I) or a therapeutically acceptable salt thereof and a pharmaceutically acceptable carrier, diluent or excipient. All these forms belong to the present invention.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention (also referred to as an "active ingredient") and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprises an effective amount of a compound of the invention.
  • the pharmaceutical composition comprises a therapeutically effective amount of a compound of the invention.
  • the pharmaceutical composition comprises a prophylactically effective amount of a compound of the invention.
  • the pharmaceutically acceptable excipient used in the present invention refers to a non-toxic carrier, adjuvant or vehicle that does not destroy the pharmacological activity of the compound formulated together.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that can be used in the composition of the present invention include (but are not limited to) ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum white Protein), buffer substances (such as phosphate), glycine, sorbic acid, potassium sorbate, partial glyceride mixture of saturated plant fatty acids, water, salt or electrolyte (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate , Sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylate, wax, polyethylene-polyoxypropylene- Block polymers, polyethylene glycol and
  • kits e.g., pharmaceutical packaging.
  • the kit provided may include the compound of the present invention, other therapeutic agents, and first and second containers (for example, vials, ampoules, bottles, syringes, and/or dispersible packages or other containers) containing the compound of the present invention and other therapeutic agents. Suitable container).
  • the provided kit may also optionally include a third container, which contains pharmaceutical excipients for diluting or suspending the compound of the present invention and/or other therapeutic agents.
  • the compound of the invention and the other therapeutic agent provided in the first container and the second container are combined to form a unit dosage form.
  • parenteral administration includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, intrasternal administration , Intracerebrospinal membrane administration, intralesional administration, and intracranial injection or infusion technology.
  • an effective amount of the compound provided herein is administered.
  • the doctor can determine the amount of the compound actually administered .
  • the compound provided herein is administered to a subject at risk of developing the condition, typically based on the doctor's recommendation and under the supervision of the doctor, and the dosage level is as described above.
  • Subjects at risk of developing a specific condition generally include subjects with a family history of the condition, or those subjects who are particularly sensitive to developing the condition as determined by genetic testing or screening.
  • long-term administration refers to the administration of the compound or its pharmaceutical composition over a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or the administration can be continued indefinitely, For example, the rest of the subject's life.
  • long-term administration is intended to provide a constant level of the compound in the blood over a long period of time, for example, within a therapeutic window.
  • the pharmaceutical composition may be administered as a bolus, for example, to increase the concentration of the compound in the blood to an effective level.
  • the bolus dose depends on the target systemic level of the active ingredient passing through the body. For example, an intramuscular or subcutaneous bolus dose allows the active ingredient to be released slowly, while a bolus injection delivered directly to a vein (for example, by IV infusion) ) Can be delivered more quickly, so that the concentration of the active component in the blood quickly rises to an effective level.
  • the pharmaceutical composition may be administered in the form of a continuous infusion, for example, by IV infusion, so as to provide a steady-state concentration of the active ingredient in the subject's body.
  • a bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
  • Oral compositions can take the form of bulk liquid solutions or suspensions or bulk powders. However, more generally, in order to facilitate precise dosing, the composition is provided in unit dosage form.
  • unit dosage form refers to physically discrete units suitable as unit dosages for human patients and other mammals, each unit containing a predetermined number of active substances suitable for producing the desired therapeutic effect and suitable pharmaceutical excipients.
  • Typical unit dosage forms include pre-filled, pre-measured ampoules or syringes of liquid compositions, or pills, tablets, capsules, etc. in the case of solid compositions.
  • the compound is usually a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), and the remaining part is useful for forming the desired administration form.
  • Kinds of carriers or excipients and processing aids are used for forming the desired administration form.
  • the representative regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses.
  • each dose provides about 0.01 to about 20 mg/kg of the compound of the present invention, with preferred doses each providing about 0.1 to about 10 mg/kg, especially about 1 to about 5 mg/kg.
  • the transdermal dose is usually selected in an amount of about 0.01 to about 20% by weight, preferably about 0.1 to about 20% by weight, and preferably about 0.1 To about 10% by weight, and more preferably about 0.5 to about 15% by weight.
  • the injection dose level is in the range of about 0.1 mg/kg/hour to at least 10 mg/kg/hour.
  • a preload bolus of about 0.1 mg/kg to about 10 mg/kg or more can also be given.
  • the maximum total dose cannot exceed approximately 2 g/day.
  • Liquid forms suitable for oral administration may include suitable aqueous or non-aqueous carriers as well as buffers, suspending and dispersing agents, coloring agents, flavoring agents, and the like.
  • the solid form may include, for example, any of the following components, or compounds with similar properties: binders, for example, microcrystalline cellulose, tragacanth, or gelatin; excipients, for example, starch or lactose, disintegrants, For example, alginic acid, Primogel or corn starch; lubricants, for example, magnesium stearate; glidants, for example, colloidal silicon dioxide; sweeteners, for example, sucrose or saccharin; or flavoring agents, for example, mint, water Methyl salicylate or orange flavoring agent.
  • binders for example, microcrystalline cellulose, tragacanth, or gelatin
  • excipients for example, starch or lactose, disintegrants, For example, alginic acid, Primogel or corn starch
  • Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art.
  • the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
  • the transdermal composition is typically formulated as a topical ointment or cream containing the active ingredients.
  • the active ingredient When formulated as an ointment, the active ingredient is typically combined with paraffin wax or an ointment base that is miscible with water.
  • the active ingredient can be formulated as a cream with, for example, an oil-in-water cream base.
  • Such transdermal formulations are well known in the art, and usually include other components for enhancing the active ingredient or stable skin penetration of the formulation. All such known transdermal preparations and components are included within the scope provided by the present invention.
  • transdermal administration can be achieved using a reservoir or porous membrane type, or a variety of solid matrix patches.
  • compositions for oral administration, injection or topical administration are only representative.
  • Other materials and processing techniques are described in Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania in Part 8, which is incorporated herein by reference.
  • the compounds of the present invention can also be administered in a sustained release form or from a sustained release drug delivery system.
  • sustained-release materials can be found in Remington's Pharmaceutical Sciences.
  • the invention also relates to pharmaceutically acceptable formulations of the compounds of the invention.
  • the formulation contains water.
  • the formulation comprises a cyclodextrin derivative.
  • the most common cyclodextrins are ⁇ -, ⁇ - and ⁇ -cyclodextrins composed of 6, 7 and 8 ⁇ -1,4-linked glucose units, respectively, which optionally include one on the linked sugar moiety Or multiple substituents, including but not limited to: methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substitution.
  • the cyclodextrin is a sulfoalkyl ether ⁇ -cyclodextrin, for example, sulfobutyl ether ⁇ -cyclodextrin, also known as Captisol. See, for example, U.S. 5,376,645.
  • the formulation includes hexapropyl- ⁇ -cyclodextrin (e.g., 10-50% in water).
  • EGFR kinase is known to play a role in tumorigenesis and many other diseases.
  • the compounds of the present invention have potent anti-tumor activity, which is believed to be obtained by inhibiting EGFR kinase.
  • the compound of the present invention has value as an antitumor agent.
  • the compounds of the present invention have value as anti-proliferation, apoptosis and/or anti-invasive agents in the suppression and/or treatment of solid and/or liquid tumor diseases.
  • the compounds of the present invention are expected to be useful in the prevention or treatment of those tumors that are sensitive to inhibition of EGFR.
  • the compounds of the present invention are expected to be useful in the prevention or treatment of those tumors mediated solely or in part by EGFR. Therefore, the compounds can be used to produce EGFR enzyme inhibition in warm-blooded animals in need of such treatment.
  • inhibitors of EGFR kinase include cancers such as ovarian cancer, cervical cancer, colorectal cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, melanoma, prostate cancer, leukemia, lymphoma Tumor, non-Hodgkin’s lymphoma, gastric cancer, lung cancer, hepatocellular carcinoma, gastric cancer, gastrointestinal stromal tumor (GIST), thyroid cancer, cholangiocarcinoma, endometrial cancer, kidney cancer, anaplastic large cell lymphoma, acute Myeloid leukemia (AML), multiple myeloma, melanoma, mesothelioma.
  • cancers such as ovarian cancer, cervical cancer, colorectal cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, melanoma, prostate cancer, leukemia, lymphoma Tumor, non-Hodgkin’s lymphoma, gas
  • Anti-cancer effects useful for treating cancer in patients include, but are not limited to, anti-tumor effects, response rate, time to disease progression, and survival rate.
  • the anti-tumor effects of the treatment method of the present invention include, but are not limited to, inhibition of tumor growth, delay of tumor growth, tumor regression, tumor shrinkage, prolonged tumor regeneration for a long time after treatment is stopped, and slowing of disease progression.
  • Anti-cancer effects include preventive treatment and treatment of existing diseases.
  • EGFR kinase inhibitors or pharmaceutically acceptable salts thereof are also used in the treatment of cancer patients, including but not limited to blood cancers, such as leukemia, multiple myeloma; lymphomas, such as Hodgkin’s disease and non-Hodgkin’s Lymphoma (including mantle cell lymphoma) and myelodysplastic syndrome, as well as solid tumors and their metastases (metastases), such as breast cancer, lung cancer (non-small cell lung cancer (NSCL), small cell lung cancer (SCLC), Squamous cell carcinoma), endometrial cancer, central nervous system tumors (such as glioma, embryonic dysplastic neuroepithelial tumor, glioblastoma multiforme, mixed glioma, medulloblastoma, Retinoblastoma, neuroblastoma, germ cell tumor and teratoma, gastrointestinal cancer (such as stomach cancer), esophageal cancer, hepatocellular (liver)
  • the effective amount of the compound of the present invention is usually 0.01 mg to 50 mg of the compound per kilogram of the patient's body weight at an average daily dose, preferably 0.1 mg to 25 mg of the compound per kilogram of the patient's body weight, in single or multiple administrations.
  • the compound of the present invention can be administered to the patient in need of such treatment at a daily dose ranging from about 1 mg to about 3500 mg per patient, preferably 10 mg to 1000 mg.
  • the daily dose per patient can be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900 or 1000mg. It can be administered once or multiple times daily, weekly (or at intervals of several days) or on an intermittent schedule.
  • the compound may be administered on a weekly basis (e.g., every Monday), one or more times per day, variably or for several weeks, for example 4-10 weeks.
  • the compound may be administered daily for several days (for example, 2-10 days), and then the compound may not be administered for several days (for example, 1-30 days), and the cycle may be repeated indefinitely or a given number of times, such as 4-10 Cycles.
  • the compound of the present invention can be administered daily for 5 days, then 9 days intermittently, and then administered daily for 5 days, then 9 days intermittently, and so on, repeating the cycle arbitrarily or repeating 4-10 times in total.
  • the treatment defined herein may be applied as a stand-alone treatment, or may include conventional surgery or radiotherapy or chemotherapy in addition to the compounds of the invention. Therefore, the compounds of the present invention can also be used in combination with existing therapeutic agents for the treatment of cancer.
  • This chemotherapy and the compound of the present invention may be administered simultaneously, continuously, or separately, and may contain one or more of the following types of anti-tumor agents:
  • Anti-proliferative/anti-tumor drugs and their combinations used in medical oncology such as alkylating agents (such as cisplatin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, phenidine) Chlorambucil, busulfan, temozolomide, nitrosoureas); antimetabolites (e.g. gemcitabine and antifolates, such as fluoropyrimidines (e.g.
  • anti-tumor antibiotics such as anthracyclines, such as doxorubicin, bleomycin, doxorubicin, daunorubicin, epirubicin, idarubicin, Mitomycin C, Actinomycin, Mitomycin
  • Anti-mitotic agents for example, vinca alkaloids, such as vincristine, vinblastine, vindesine, vinorelbine; and taxanes, such as Paclitaxel, taxotecan, polo kinase inhibitors
  • topoisomerase inhibitors e.g. epipodophyllotoxin (such as etoposide, teniposide), amsacrine, topotecan, camptothecin);
  • Cell growth inhibitors such as anti-estrogens (such as tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene, iodoxifene (iodoxyfene)), antiandrogens Hormonal drugs (e.g. bicalutamide, flutamide, nilutamide, cyproterone acetate), LHRH antagonists or LHRH agonists (e.g. goserelin, leuprolide, and buserelin), Progestogens (e.g. megestrol acetate), aromatase inhibitors (e.g., anastrozole, letrozole, vorazole, isemestane), 5 ⁇ -reductase inhibitors (e.g. finasteride );
  • anti-estrogens such as tamoxifen, fulvestrant, toremifene, raloxifene, droloxi
  • Anti-invasion such as c-Src kinase family inhibitors, [such as 4-(6-chloro-2,3-methylenedioxyanilino)-7-[2-( 4-Methylpiperazin-1-yl)ethoxy]-5-tetrahydropyran-4-yloxyquinazoline [AZD0530(Secatinib)], N-(2-chloro-6- Methylphenyl)-2- ⁇ 6-[4-(2-hydroxyethyl)piperazin-1-yl]-2-methylpyrimidin-4-ylamino ⁇ thiazole-5-carboxamide (dasatidine Ni, BMS-354825) and Bosutinib (SKI-606), as well as metalloproteinase inhibitors (such as marimastat), inhibitors of urokinase plasminogen activator receptor function or heparanase ( heparanase) antibody];
  • Inhibitors of growth factor functions include growth factor antibodies and growth factor receptor antibodies (for example, anti-erbB2 antibody trastuzumab [Herceptin], anti-EGFR antibody panitumumab, anti- erbB1 antibody cetuximab [Erbitux, C225]; this inhibitor also includes: tyrosine kinase inhibitors, such as inhibitors of the epidermal growth factor family (for example, EGFR family tyrosine kinase inhibitors, such as N-( 3-chloro-4-fluorophenyl)-7-methoxy-6-(3-morpholinylpropoxy)-quinazolin-4-amine (gefitinib, ZD1839), N-(3 -Ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-amine (erlotinib, OSI-774), 6-acrylamido-N-(3 -Chloro
  • Anti-angiogenic agents such as agents that inhibit the action of vascular endothelial growth factor, [e.g. anti-human vascular endothelial cell growth factor antibody bevacizumab (Avastin) and e.g.
  • VEGF receptor tyrosine kinase inhibitors For example, vandetanib (ZD6474), vataranib (PTK787), sunitinib (SU11248), axitinib (AG-013736), pazopanib (GW786034), 4-(4-fluoro- 2-Methylindol-5-yloxy)-6-methoxy-7-(3-pyrrolidin-1-ylpropoxy)quinazoline (AZD2171), and compounds that act through other mechanisms (E.g. Ranolamide, inhibitor of integrin ⁇ v ⁇ 3 function and angiostatin)];
  • Vascular injury agents such as Comprutin A4
  • Endothelin receptor antagonists such as zipopertentan (ZD4054) or atrasentan;
  • Antisense therapeutics such as those directed to the targets listed above, such as ISIS2503 (an anti-ras antisense therapeutic);
  • Gene therapy methods including, for example, methods of replacing abnormal genes (such as abnormal p53 or abnormal BRCA1 or BRCA2); GDEPT (gene-directed enzyme prodrug therapy) methods, such as using cytosine deaminase, thymidine kinase or bacteria Those of nitroreductase; methods to improve the tolerance of patients to chemotherapy or radiotherapy, such as multidrug-resistant gene therapy; and
  • Immunotherapy methods including, for example, in vitro and in vivo methods to improve the immunogenicity of patient tumor cells, such as transfection with cytokines (such as interleukin 2, interleukin 4 or granulocyte macrophage colony stimulating factor) Transfection; methods to reduce the ineffectiveness of T cells; methods using transfected immune cells (such as cytokine-transfected dendritic cells); methods using cytokine-transfected tumor cell lines; methods using anti-idiotypic antibodies Methods of reducing the function of immunosuppressive cells (such as regulatory T cells, myeloid-derived suppressor cells, or dendritic cells expressing IDO (indoleamine 2,3-deoxygenase)); and A method of using a cancer vaccine composed of proteins or peptides derived from tumor-associated antigens (such as NY-ESO-1, mAGE-3, WT1, or Her2/neu).
  • cytokines such as interleukin 2, interleukin 4 or granulocyte macrophage
  • step 1
  • A1-1 (600mg, 1.79mmol), 6-alkynoheptanoic acid (450mg, 3.58mmol), CuI (69mg, 0.36mmol), Pd(PPh 3 ) 2 Cl 2 (501mg, 0.71mmol) and triethylamine ( 903mg, 8.94mmol) was added to DMF (15ml) and stirred at 70°C for 5 hours under nitrogen protection.
  • step 1
  • step 1
  • A3-1 (116 mg) was dissolved in TFA (1 ml) and stirred at room temperature for 1 hour. The solvent was evaporated to obtain crude product A3-2, which was directly used in the next step.
  • A3-4 (110mg, 0.13mmol) and TFA (1ml) were dissolved in dichloromethane and stirred at room temperature for 1 hour. The solvent was evaporated under reduced pressure to obtain crude product A3-5, which was directly used in the next step.
  • step 1
  • reaction solution was diluted with 50 ml of water, extracted three times with ethyl acetate (25 ml each time), combined the organic phases, washed three times with saturated brine (20 ml each time), and the organic phase was dried with anhydrous sodium sulfate for 2 hours The filtrate was filtered and concentrated under reduced pressure to obtain the crude product.
  • the crude product was separated by silica gel column chromatography to obtain yellow solid A6-3 (200 mg, yield 48%).
  • step 1
  • reaction solution was cooled and filtered to remove inorganic salts, diluted with 20 ml of water, extracted three times with ethyl acetate (25 ml of ethyl acetate each time), combined the organic phases, washed twice with saturated brine (10 mL each time), and then used Drying with anhydrous sodium sulfate, filtering, and concentrating the filtrate under reduced pressure to obtain a crude product, which was separated by silica gel column chromatography to obtain compound A8-2 (100 mg, yield: 24.9%).
  • step 1
  • the crude product was purified by RP-Flash chromatography to obtain 65 mg of a white solid product with a purity of about 60%.
  • step 1
  • reaction solution was diluted with 80 mL ethyl acetate, it was washed with saturated NH 4 Cl solution (20 mL x 1), H 2 O (20 mL x 3) and saturated brine (20 mL x 2) successively, and the organic layer was washed with anhydrous Na 2 SO 4 Dry, remove the desiccant by suction filtration, and concentrate the filtrate under reduced pressure to obtain a crude product.
  • Prep-TLC purification obtains 70 mg of A11-1 as a white solid product with a yield of 33.6%.
  • LCMS: [M+H] + 417.
  • step 1
  • step 1
  • A13-2 (228mg, 1.04mmol) and 3-(4-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione (A12-3) (270mg, 1.04mmol) ) was dissolved in DCM (10ml) and MeOH (5ml), and stirred at 50°C for 1 hour under nitrogen protection. Cool to room temperature, add sodium cyanoborohydride (98mg, 1.56mmol) and 2 drops of glacial acetic acid, raise to 50°C and continue stirring for 1.5 hours.
  • step 1
  • step 1
  • step 1
  • step 1
  • A6-5 (66.3 mg, 0.1 mmol) and 4-(2-bromoethyl) piperidine trifluoroacetate (30.5 mg, 0.1 mmol) were dispersed in 5.0 mL of anhydrous DCM, and T3P (159 mg, 0.25mmol) and DIPEA (64.5mg, 0.5mmol), after the addition, react at room temperature for 2h.
  • the reaction solution was diluted with 20 mL DCM, it was washed with saturated ammonium chloride solution (10 mL x 1) and saturated NaCl (10 mL x 1) successively, the organic layer was dried with anhydrous MgSO 4 , filtered with suction, and the filtrate was concentrated under reduced pressure to obtain a crude product.
  • step 1
  • reaction solution was diluted with 50 mL ethyl acetate, it was washed with saturated NH 4 Cl solution (20 mL x 1), H 2 O (20 mL x 3) and saturated brine (20 mL x 2) successively, and the organic layer was washed with anhydrous Na 2 SO 4 Dry, filter with suction, and concentrate the filtrate under reduced pressure to obtain a crude product.
  • step 1
  • step 1
  • step 1
  • step 1
  • step 1
  • step 1
  • A3-2 (223mg, 0.5mmol) and C99-1 (93mg, 0.5mmol) were dispersed in 10.0mL of anhydrous DCM, and T3P (636mg, 1.0mmol) and DIPEA (322.5mg, 2.5mmol) were sequentially added under nitrogen protection, React at room temperature for 2h.
  • T3P (636mg, 1.0mmol) and DIPEA (322.5mg, 2.5mmol) were sequentially added under nitrogen protection, React at room temperature for 2h.
  • the reaction solution was diluted with 30 mL ethyl acetate, it was washed with saturated NH 4 Cl (20 mL x 1) and saturated NaCl (20 mL x 2) successively, the organic layer was dried with anhydrous MgSO 4 , filtered with suction, and the filtrate was concentrated under reduced pressure to obtain a crude product .
  • the crude product was purified by Flash to obtain a white solid product C99-2 (230 mg, yield 92%)
  • step 1
  • step 1
  • step 1
  • step 1
  • step 1
  • A6-5 (51.3mg, 0.0818mmol), C110-4 (35mg, 0.0818mmol) were dispersed in a mixed solvent of 5.0mL of anhydrous DCM and 1.0mL of anhydrous DMF, under the protection of N 2 , and then added T3P (104mg, 0.1636) mmol) and DIPEA (63.3 mg, 0.4908 mmol), react at room temperature for 2 h.
  • LCMS detects that no raw material remains.
  • the reaction solution was concentrated under reduced pressure to obtain a crude product.
  • the crude product was purified by Prep-HPLC to obtain a white solid pure product C110, 25 mg, with a yield of 30.6%.
  • LCMS: [M+H] + 1002.
  • step 1
  • step 1
  • C116-1 (or C1-1) (500mg, 1.553mmol), CuI (59mg, 0.3106mmol) and Pd(dppf)Cl 2 (454mg, 0.6212mmol) were dispersed in 25mL of anhydrous DMF, under N 2 protection, in sequence Add 5-alkynyl-1-hexanol (380mg, 3.883mmol) and TEA (470mg, 4.659mmol), raise the temperature to 70°C and react for 20h. LCMS detected that the reaction was complete. The reaction solution was cooled to room temperature.
  • step 1
  • step 1
  • step 1
  • step 1
  • step 1
  • step 1

Landscapes

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

Abstract

一种通式(X)的新的抑制并诱导降解EGFR和ALK的化合物,以及含有所述化合物的药物组合物,它们可用于治疗与EGFR和ALK激酶相关的疾病,例如癌症,及上述化合物的制备和用途。

Description

抑制并诱导降解EGFR和ALK的化合物 技术领域
本发明涉及医药领域,具体地,本发明提供了能够抑制EGFR和ALK或诱导降解EGFR和ALK的化合物及其制备方法和应用。
背景技术
肺癌是最常见的恶性肿瘤之一,2018年全球新发肺癌210万例,占所有新发肿瘤病例的11.6%;死亡180万例,占所有肿瘤死亡病例的18.4%。其中,非小细胞肺癌(Non-Small Cell Lung Cancer,NSCLC)占肺癌总数的80%-85%。表皮生长因子受体(Epithelial Growth Factor Receptor,EGFR)和间变性淋巴瘤激酶(Anaplastic Lymphoma Kinase,ALK)为常见的非小细胞肺癌的驱动基因。
在非小细胞肺癌中,约50%的中国患者和11-16%的西方国家患者存在EGFR基因突变,其最常见的突变类型是19号外显子缺失突变(del E746-A750)和21号外显子L858R点突变,约占所有EGFR突变人群的90%。EGFR小分子抑制剂是一线治疗EGFR基因突变的非小细胞肺癌的标准疗法,已经在肺癌治疗领域广泛使用,它们通过与内源性的配体竞争性结合EGFR,抑制酪氨酸激酶的活化,进而阻断EGFR信号通路,抑制肿瘤细胞的增殖、转移并促进肿瘤细胞发生凋亡等一系列生物学效应。
第一代EGFR小分子抑制剂Gefitinib和Erlotinib已经被用于治疗携带激活型EGFR突变(L858R,del E746-A750)晚期非小细胞肺癌。然而患者在使用Gefitinib和Erlotinib 10-12个月后会产生耐药,其中,超过50%的耐药患者是由于EGFR发生了T790M二次突变导致耐药。第二代EGFR不可逆性抑制剂Afatinib对携带激活型EGFR突变(L858R,del E746-A750)的晚期非小细胞肺癌病人有效,但是无法解决EGFR T790M突变引起的临床耐药,并且Afatinib对野生型EGFR缺乏选择性,具有较大毒性。第三代不可逆性抑制剂Osimertinib克服了EGFR T790M耐药,临床上能够有效治疗EGFR T790M突变耐药的晚期非小细胞肺癌患者。尽管Osimertinib在临床上治疗EGFR T790M突变的非小细胞肺癌取得了较大的成功,但是部分受益患者在经过9-14个月治疗后又出现了耐药现象(Nature Medicine,2015,21(6),560-562)。研究表明,高达22%的耐药患者由于EGFR C797S突变导致了Osimertinib耐药(JAMA Oncol.2018;4(11):1527-1534)。EGFR C797S突变使得797位的半胱氨酸突变为丝氨酸,Osimertinib不能与EGFR共价结合,最终引起耐药。目前临床上尚缺乏针对EGFR C797S单独用药的有效EGFR抑制剂。因此,开发新一代EGFR抑制剂,满足临床治疗需求,是目前亟待解决的问题。
间变性淋巴瘤激酶(ALK)是一种受体酪氨酸蛋白激酶,ALK基因重排、点突变和基因扩增会导致机体产生癌变。ALK重排基因是一种强致癌驱动基因,其中棘皮动物微管相关类蛋白4(EML4-ALK)和核磷酸蛋白(NPM-ALK)为常见类型。ALK基因重排导致ALK在二聚体形成前可发生磷酸化过程,因此ALK融合蛋白将持续处于激活状态,并激活其下游通路,进而造成细胞过度增殖,导致肿瘤的发生。在非小细胞肺癌中,3-7%的患者出现了ALK基因重排,以ALK为靶点的小分子抑制剂已经广泛应用于临床,它们通过与内源性的配体竞争性结合ALK,抑制酪氨酸激酶的活化,进而阻断ALK信号通路,抑制肿瘤细胞的增殖、转移等一系列生物学效应。目前已上市的ALK抑制剂有crizotinib,ceritinib,alectinib、brigatinib和lorlatinib,但是不可避免的是这些抑制剂会出现耐药性,常见的ALK耐药性突变有L1196M,G1269A,S1206Y,G1202R,C1156Y,L1198F等。因此,开发新型ALK抑制剂以满足临床治疗需求具有重要意义。
泛素-蛋白酶体系统(Ubiquitin-Proteasome System,UPS)是细胞内蛋白质降解的多组分系统,参与细胞的生长和分化、DNA复制与修复、细胞代谢、免疫反应等重要生理生化过程。泛素-蛋白酶体途径介导的蛋白降解是机体调节细胞内蛋白水平与功能的一个重要机制,在维持体内蛋白稳态中发挥着 重要的作用。通过细胞内的泛素-蛋白酶体途径,诱导EGFR或ALK的降解,为治疗癌症提供了一种新的思路。
本发明提供了能够抑制EGFR和ALK,和/或诱导降解EGFR和ALK的化合物及其制备方法和应用。
发明内容
在一个方面,本发明提供通式(X)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,它们可用于治疗EGFR和/或ALK激酶介导的疾病,例如癌症。
Figure PCTCN2020110442-appb-000001
其中,
环A选自任选取代的以下基团:C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-10元杂芳基,其中所述取代基选自卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 3-7环烷基、4-8元杂环基、-P(O)(C 1-6烷基) 2、-P(O)(C 2-6烯基) 2、-O-C 1-6烷基、-O-C 1-6卤代烷基、-O-C 2-6烯基、-O-C 3-7环烷基、-O-4-8元杂环基、-NH-C 1-6烷基、-NH-C 2-6烯基、-NH-C 3-7环烷基、-NH-4-8元杂环基、-C(O)-C 1-6烷基、-C(O)-C 2-6烯基、-C(O)-C 3-7环烷基、-C(O)-4-8元杂环基、-S(O) 2-C 1-6烷基、-S(O) 2-C 2-6烯基、-S(O) 2-C 3-7环烷基、-S(O) 2-4-8元杂环基、-NHC(O)-C 1-6烷基、-NHC(O)-C 2-6烯基、-NHC(O)-C 3-7环烷基、-NHC(O)-4-8元杂环基、-NHS(O) 2-C 1-6烷基、-NHS(O) 2-C 2-6烯基、-NHS(O) 2-C 3-7环烷基或-NHS(O) 2-4-8元杂环基;
环B选自以下基团:
Figure PCTCN2020110442-appb-000002
Figure PCTCN2020110442-appb-000003
表示单键或双键;
Figure PCTCN2020110442-appb-000004
表示与分子其他部分的连接位置可以位于所在环的可用位置;
Z 1为O、S、N或C原子,其任选地被一个或两个R Z1取代;或者Z 1不存在,因而Z 4连接到Z 2、Z 3或与Z 1相连的芳环上的C原子,并且与Z 1相连的Z 2和芳环上的C原子分别与R W相连;或者Z 1、Z 2和Z 3都不存在,因而Z 4连接到与Z 1或Z 3相连的芳环上的C原子之一,并且芳环上的另一个C原子与R W相连;
Z 2为O、S、N或C原子,其任选地被一个或两个R Z2取代;
Z 3为O、S、N或C原子,其任选地被一个或两个R Z3取代;条件是,当
Figure PCTCN2020110442-appb-000005
为双键时,Z 2为N或C原子,Z 3为N或C原子;
Z 4为N或CR Z4
Z 5为N或CR Z5
R a、R b和R c独立地为H、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基或C 1-6卤代烷基; 或者R a、R b以及它们连接的C原子一起形成C=O、C 3-7环烷基或4-8元杂环基;或者R a和R c以及它们连接的C原子一起形成C 3-7环烷基或4-8元杂环基;或者R a和R c形成化学键;
R N1为H、C 1-6烷基或C 1-6卤代烷基,优选H;
R Z1为不存在、H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;或者两个R Z1与Z 1一起形成C=O、C 3-7环烷基或4-8元杂环基;
R Z2为不存在、H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;或者两个R Z2与Z 2一起形成C=O、C 3-7环烷基或4-8元杂环基;
R Z3为不存在、H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;或者两个R Z3与Z 3一起形成C=O、C 3-7环烷基或4-8元杂环基;
R Z4为H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基或C 1-6卤代烷基;
R Z5为H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;
或者Z 4所在的环不存在;
其中R w为H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、-C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基、-(CH 2) 0-5-4-8元杂环基、C 2-6烯基、C 2-6炔基、-(CH 2) 0-5-C 3-10卤代环烷基、-(CH 2) 0-5-C 6-10芳基或-(CH 2) 0-5-5-14元杂芳基;
R”为H、C 1-6烷基、C 1-6卤代烷基或-(CH 2) 0-5-C 3-7环烷基;
R”’为H、C 1-6烷基或C 1-6卤代烷基;
L 1选自化学键、-O-、-S(O) p-、-S(O)(=NR *)-、-NR #-、-CR #R #’-、-C aR #R #’-C bR #R #’-、-N=S(O)(R *)-或-S(O)(R *)=N-;
L 2选自化学键、-O-、-S(O) p-、-S(O)(=NR *)-、-NR #-、-CR #R #’-、-C aR #R #’-C bR #R #’-、-N=S(O)(R *)-或-S(O)(R *)=N-;
其中C aR #R #’或C bR #R #’中任一个可被O、S(O) p、S(O)(=NR *)或NR #替换,并且当C aR #R #’或C bR #R #’中任一个被O、S或NR #替换时,C aR #R #’或C bR #R #’中另一个还可被S(O) q替换;
E独立地选自:化学键、-C cR #R #’-C dR #R #’-C eR #R #’、
Figure PCTCN2020110442-appb-000006
Figure PCTCN2020110442-appb-000007
其中C cR #R #’、C dR #R #’或C eR #R #’中任一个,或者C cR #R #’和C eR #R #’二者可被O、S(O) p、S(O)(=NR *)或NR #替换,并且当C cR #R #’、C dR #R #’或C eR #R #’中任一个被O、S或NR #替换时,与之相邻的另一个或两个C cR #R #’、C dR #R #’或C eR #R #’还可被S(O) q替换;
或者两个E单元可以形成-CH 2CH 2OCH 2CH 2-、-OCH 2CH 2CH 2CH 2-、-CH 2CH 2CH 2CH 2O-、
Figure PCTCN2020110442-appb-000008
其中,
Figure PCTCN2020110442-appb-000009
表示与L 1或L 2的连接点;
H 1和H 2为N或C原子,H 3为O、S、N或C原子,并且H 1和H 3、H 2和H 3不同时为杂原子;
H 4和H 5为N或C原子;
H 6、H 7、H 8和H 9为C或N原子;
p为0、1或2;
q为1或2;
R *为H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基或5-14元杂芳基;
R #为H、卤素、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基或5-14元杂芳基;
R #’为H、卤素、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基或5-14元杂芳基;
或者,相邻原子上的R #和R #可以形成化学键,相邻原子上的R #’和R #’可以形成化学键;
或者,相同或不同原子上的R #和R #’可以一起形成=O、或任选被R x取代的C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-6元杂芳基,其中所述R x为H、CN、卤素、C 1-6烷基或C 1-6卤代烷基;
m为0、1、2、3、4、5、6、7、8、9或10;
R s1选自H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-4-8元杂环基、C 2-6烯基、C 2-6炔基、-(CH 2) 0-5-C 3-7环烷基、-(CH 2) 0-5-C 3-10卤代环烷基、-(CH 2) 0-5-C 6-10芳基、-(CH 2) 0-5-5-14元杂芳基、-C(O)R W、-S(O)R W或-S(O) 2R W
s1为0、1、2或3;
R’选自H、-C(O)-C 1-6烷基、-C(O)-C 1-6卤代烷基、-C(O)-C 2-6烯基或-C(O)-C 6-10芳基;
L为化学键、-O-或-NR-;
其中R为H或C 1-6烷基;
Z为-N=或-C(R 4)=;
T选自化学键、C 2-6亚杂烷基、4-12元亚杂环基或5-6元亚杂环基取代的5-6元亚杂环基;
R 1选自H、卤素、氰基、C 1-6烷基、C 3-7环烷基、4-8元杂环基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
或者,当L为-NR-时,R 1、R与它们连接的原子一起形成任选取代的5-6元杂环基,其中所述取代基选自卤素、氧代、C 1-6烷基、C 1-6卤代烷基、卤素单取代或多取代的C 6-10芳基;
R 2为H、卤素、羟基、氨基、C 1-6烷基或C 1-6卤代烷基;
或者R 1、R 2与它们连接的原子一起形成5-6元杂环基或5-6元杂芳基;
R 3选自H、-O-C 1-6烷基或-O-C 1-6卤代烷基;
R 4选自H、卤素、C 1-6烷基、C 1-6卤代烷基、-NHC(O)-C 1-6烷基或-NHC(O)-C 2-6烯基;
当上述各基团为H或含H的基团时,所述一个或多个H原子可以被D原子取代;
上述烷基、亚烷基、卤代烷基、烯基、炔基、环烷基、卤代环烷基、杂环基、芳基、杂芳基或L 1、E、L 2、和T中含有OH、NH、NH 2、CH、CH 2、CH 3的基团在每次出现时各自任选地被1、2、3或更多个R s取代,其中所述R s在每次出现时独立地选自:卤素、羟基、氨基、氰基、硝基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、-OR a’、-OC(O)R a’、-C(O)R a’、-C(O)OR a’、-C(O)NR a’R b’、-S(O) nR a’、-S(O) nOR a’、-S(O) nNR a’R b’、-NR a’R b’、-NR a’C(O)R b’、-NR a’-C(O)OR b’、-NR a’-S(O) n-R b’、-NR a’C(O)NR a’R b’、-C 1-6亚烷基-R a’、-C 1-6亚烷基-OR a’、-C 1-6亚烷基-OC(O)R a’、-C 1-6亚烷基-C(O)OR a’、-C 1-6亚烷基-S(O) nR a’、-C 1-6亚烷基-S(O) nOR a’、-C 1-6亚烷基-OC(O)NR a’R b’、-C 1-6亚烷基-C(O)NR a’R b’、-C 1-6亚烷基 -NR a’-C(O)NR a’R b’、-C 1-6亚烷基-OS(O) nR a’、-C 1-6亚烷基-S(O) nNR a’R b’、-C 1-6亚烷基-NR a’-S(O) nNR a’R b’、-C 1-6亚烷基-NR a’R b’和-O-C 1-6亚烷基-NR a’R b’,并且其中关于取代基R s所述的羟基、氨基、烷基、亚烷基、环烷基、杂环基、芳基、杂芳基和芳烷基进一步任选地被1、2、3或更多个独立地选自下列的取代基取代:卤素、OH、氨基、氰基、硝基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷基羟基、C 3-6环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基;
n每次出现时各自独立地为1或2;
R a’和R b’在每次出现时各自独立地选自H、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷基-O-、C 1-6烷基-S-、C 3-10环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基。
在另一个方面,本发明提供了一种药物组合物,所述药物组合物含有本发明化合物,和任选地药学上可接受的赋形剂。
在另一个方面,本发明提供了含有本发明化合物和药学上可接受的赋形剂的药物组合物,其还含有其它治疗剂。
在另一个方面,本发明提供了包含本发明化合物,和其它治疗剂以及药学上可接受的载剂、佐剂或媒剂的试剂盒。
在另一个方面,本发明提供了本发明化合物在制备用于治疗和/或预防EGFR和/或ALK激酶介导的疾病的药物中的用途。
在另一个方面,本发明提供了在受试者中治疗和/或预防EGFR和/或ALK激酶介导的疾病的方法,包括向所述受试者给药本发明化合物或本发明组合物。
在另一个方面,本发明提供了本发明化合物或本发明组合物,其用于治疗和/或预防EGFR和/或ALK激酶介导的疾病。
在具体实施方案中,本发明治疗的疾病包括癌症,例如卵巢癌、宫颈癌、结肠直肠癌、乳腺癌、胰腺癌、胶质瘤、胶质母细胞瘤、黑色素瘤、前列腺癌、白血病、淋巴瘤、非霍奇金淋巴瘤、胃癌、肺癌、肝细胞癌、胃癌、胃肠道基质瘤(GIST)、甲状腺癌、胆管癌、子宫内膜癌、肾癌、间变性大细胞淋巴瘤、急性髓细胞白血病(AML)、多发性骨髓瘤、黑色素瘤、间皮瘤。
由随后的具体实施方案、实施例和权利要求,本发明的其它目的和优点将对于本领域技术人员显而易见。
定义
化学定义
下面更详细地描述具体官能团和化学术语的定义。
当列出数值范围时,既定包括每个值和在所述范围内的子范围。例如“C 1-6烷基”包括C 1、C 2、C 3、C 4、C 5、C 6、C 1-6、C 1-5、C 1-4、C 1-3、C 1-2、C 2-6、C 2-5、C 2-4、C 2-3、C 3-6、C 3-5、C 3-4、C 4-6、C 4-5和C 5-6烷基。
“C 1-6烷基”是指具有1至6个碳原子的直链或支链饱和烃基团。在一些实施方案中,C 1-4烷基是优选的。C 1-6烷基的例子包括:甲基(C 1)、乙基(C 2)、正丙基(C 3)、异丙基(C 3)、正丁基(C 4)、叔丁基(C 4)、仲丁基(C 4)、异丁基(C 4)、正戊基(C 5)、3-戊基(C 5)、戊基(C 5)、新戊基(C 5)、3-甲基-2-丁基(C 5)、叔戊基(C 5)和正己基(C 6)。术语“C 1-6烷基”还包括杂烷基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。烷基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。常规烷基缩写包括:Me(-CH 3)、Et(-CH 2CH 3)、iPr(-CH(CH 3) 2)、nPr(-CH 2CH 2CH 3)、n-Bu(-CH 2CH 2CH 2CH 3)或i-Bu(-CH 2CH(CH 3) 2)。
“C 2-6烯基”是指具有2至6个碳原子和至少一个碳碳双键的直链或支链烃基团。在一些实施方案中,C 2-4烯基是优选的。C 2-6烯基的例子包括:乙烯基(C 2)、1-丙烯基(C 3)、2-丙烯基(C 3)、1-丁烯基(C 4)、2-丁烯基(C 4)、丁二烯基(C 4)、戊烯基(C 5)、戊二烯基(C 5)、己烯基(C 6),等等。术语“C 2-6烯基”还包括杂烯基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。烯基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C 2-6炔基”是指具有2至6个碳原子、至少一个碳-碳叁键以及任选地一个或多个碳-碳双键的直链或支链烃基团。在一些实施方案中,C 2-4炔基是优选的。C 2-6炔基的例子包括但不限于:乙炔基(C 2)、1-丙炔基(C 3)、2-丙炔基(C 3)、1-丁炔基(C 4)、2-丁炔基(C 4),戊炔基(C 5)、己炔基(C 6),等等。术语“C 2-6炔基”还包括杂炔基,其中一或多个(例如,1、2、3或4个)碳原子被杂原子(例如,氧、硫、氮、硼、硅、磷)替代。炔基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基”指的是上述定义的“C 1-6烷基、C 2-6烯基或C 2-6炔基”的二价基团。
“C 1-6亚烷基”是指除去C 1-6烷基的另一个氢而形成的二价基团,并且可以是取代或未取代的。在一些实施方案中,C 1-4亚烷基是特别优选的。未取代的所述亚烷基包括但不限于:亚甲基(-CH 2-)、亚乙基(-CH 2CH 2-)、亚丙基(-CH 2CH 2CH 2-)、亚丁基(-CH 2CH 2CH 2CH 2-)、亚戊基(-CH 2CH 2CH 2CH 2CH 2-)、亚己基(-CH 2CH 2CH 2CH 2CH 2CH 2-),等等。示例性的取代的所述亚烷基,例如,被一个或多个烷基(甲基)取代的所述亚烷基,包括但不限于:取代的亚甲基(-CH(CH 3)-、-C(CH 3) 2-)、取代的亚乙基(-CH(CH 3)CH 2-、-CH 2CH(CH 3)-、-C(CH 3) 2CH 2-、-CH 2C(CH 3) 2-)、取代的亚丙基(-CH(CH 3)CH 2CH 2-、-CH 2CH(CH 3)CH 2-、-CH 2CH 2CH(CH 3)-、-C(CH 3) 2CH 2CH 2-、-CH 2C(CH 3) 2CH 2-、-CH 2CH 2C(CH 3) 2-),等等。
“C 2-6亚烯基”是指除去C 2-6烯基的另一个氢而形成的二价基团,并且可以是取代或未取代的。在一些实施方案中,C 2-4亚烯基是特别优选的。示例性的未取代的所述亚烯基包括但不限于:亚乙烯基(-CH=CH-)和亚丙烯基(例如,-CH=CHCH 2-、-CH 2-CH=CH-)。示例性的取代的所述亚烯基,例如,被一个或多个烷基(甲基)取代的亚烯基,包括但不限于:取代的亚乙基(-C(CH 3)=CH-、-CH=C(CH 3)-)、取代的亚丙烯基(-C(CH 3)=CHCH 2-、-CH=C(CH 3)CH 2-、-CH=CHCH(CH 3)-、-CH=CHC(CH 3) 2-、-CH(CH 3)-CH=CH-、-C(CH 3) 2-CH=CH-、-CH 2-C(CH 3)=CH-、-CH 2-CH=C(CH 3)-),等等。
“C 2-6亚炔基”是指除去C 2-6炔基的另一个氢而形成的二价基团,并且可以是取代或未取代的。在一些实施方案中,C 2-4亚炔基是特别优选的。示例性的所述亚炔基包括但不限于:亚乙炔基(-C≡C-)、取代或未取代的亚丙炔基(-C≡CCH 2-),等等。
“C 1-6杂烷基”是指本文所定义的C 1-6烷基,并且在母体链内,它进一步含有一或多个(例如,1、2、3或4个)杂原子(例如,氧、硫、氮、硼、硅、磷),其中,一个或多个杂原子在所述母体碳链内的相邻碳原子之间,和/或,一个或多个杂原子在碳原子和母体分子之间,即,在连接点之间。C 1-6杂烷基与母体分子的连接点可为碳原子,也可为杂原子。
“C 2-6亚杂烷基”是指除去C 1-6杂烷基的另一个氢而形成的二价基团,并且可以是取代或未取代的。C 1-6亚杂烷基与母体分子其他部分的连接点可为两个碳原子,也可为两个杂原子,还可为一个碳原子和一个杂原子。
“卤代”或“卤素”是指氟(F)、氯(Cl)、溴(Br)和碘(I)。
因此,“C 1-6卤代烷基”是指上述“C 1-6烷基”,其被一个或多个卤素基团取代。在一些实施方案中, C 1-4卤代烷基是特别优选的,更优选C 1-2卤代烷基。示例性的所述卤代烷基包括但不限于:-CF 3、-CH 2F、-CHF 2、-CHFCH 2F、-CH 2CHF 2、-CF 2CF 3、-CCl 3、-CH 2Cl、-CHCl 2、2,2,2-三氟-1,1-二甲基-乙基,等等。卤代烷基基团可以在任何可用的连接点上被取代,例如,1至5个取代基、1至3个取代基或1个取代基。
“C 3-10环烷基”是指具有3至10个环碳原子和零个杂原子的非芳香环烃基团。在一些实施方案中,C 3-7环烷基和C 3-6环烷基是特别优选的,更优选C 5-6环烷基。环烷基还包括其中上述环烷基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在环烷基环上,且在这样的情况中,碳的数目继续表示环烷基体系中的碳的数目。示例性的所述环烷基包括但不限于:环丙基(C 3)、环丙烯基(C 3)、环丁基(C 4)、环丁烯基(C 4)、环戊基(C 5)、环戊烯基(C 5)、环己基(C 6)、环己烯基(C 6)、环已二烯基(C 6)、环庚基(C 7)、环庚烯基(C 7)、环庚二烯基(C 7)、环庚三烯基(C 7),等等。环烷基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C 3-10卤代环烷基”是指上述“C 3-10环烷基”,其被一个或多个卤素基团取代。
“3-12元杂环基”是指具有环碳原子和1至5个环杂原子的3至12元非芳香环系的基团,其中,每个杂原子独立地选自氮、氧、硫、硼、磷和硅。在包含一个或多个氮原子的杂环基中,只要化合价允许,连接点可为碳或氮原子。在一些实施方案中,优选4-12元杂环基,其为具有环碳原子和1至5个环杂原子的4至12元非芳香环系;在一些实施方案中,优选3-10元杂环基,其为具有环碳原子和1至5个环杂原子的3至10元非芳香环系;在一些实施方案中,优选3-8元杂环基,其为具有环碳原子和1至4个环杂原子的3至8元非芳香环系;优选3-6元杂环基,其为具有环碳原子和1至3个环杂原子的3至6元非芳香环系;优选4-8元杂环基,其为具有环碳原子和1至3个环杂原子的4至8元非芳香环系;更优选5-6元杂环基,其为具有环碳原子和1至3个环杂原子的5至6元非芳香环系。杂环基还包括其中上述杂环基环与一个或多个环烷基稠合的环体系,其中连接点在环烷基环上,或其中上述杂环基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在杂环基环上;且在这样的情况下,环成员的数目继续表示在杂环基环体系中环成员的数目。示例性的包含一个杂原子的3元杂环基包括但不限于:氮杂环丙烷基、氧杂环丙烷基、硫杂环丙烷基(thiorenyl)。示例性的含有一个杂原子的4元杂环基包括但不限于:氮杂环丁烷基、氧杂环丁烷基和硫杂环丁烷基。示例性的含有一个杂原子的5元杂环基包括但不限于:四氢呋喃基、二氢呋喃基、四氢噻吩基、二氢噻吩基、吡咯烷基、二氢吡咯基和吡咯基-2,5-二酮。示例性的包含两个杂原子的5元杂环基包括但不限于:二氧杂环戊烷基、氧硫杂环戊烷基(oxasulfuranyl)、二硫杂环戊烷基(disulfuranyl)和噁唑烷-2-酮。示例性的包含三个杂原子的5元杂环基包括但不限于:三唑啉基、噁二唑啉基和噻二唑啉基。示例性的包含一个杂原子的6元杂环基包括但不限于:哌啶基、四氢吡喃基、二氢吡啶基和硫杂环己烷基(thianyl)。示例性的包含两个杂原子的6元杂环基包括但不限于:哌嗪基、吗啉基、二硫杂环己烷基、二噁烷基。示例性的包含三个杂原子的6元杂环基包括但不限于:六氢三嗪基(triazinanyl)。示例性的含有一个杂原子的7元杂环基包括但不限于:氮杂环庚烷基、氧杂环庚烷基和硫杂环庚烷基。示例性的与C 6芳基环稠合的5元杂环基(在本文中也称作5,6-双环杂环基)包括但不限于:二氢吲哚基、异二氢吲哚基、二氢苯并呋喃基、二氢苯并噻吩基、苯并噁唑啉酮基,等等。示例性的与C 6芳基环稠合的6元杂环基(本文还指的是6,6-双环杂环基)包括但不限于:四氢喹啉基、四氢异喹啉基,等等。杂环基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“4-12元亚杂环基”和“5-6元亚杂环基”分别表示上述“4-12元杂环基”和“5-6元杂环基”,其中另一个氢被除去而形成的二价基团,并且可以是取代或未取代的。
“C 6-10芳基”是指具有6-10个环碳原子和零个杂原子的单环或多环的(例如,双环)4n+2芳族环体系(例如,具有以环状排列共享的6或10个π电子)的基团。在一些实施方案中,芳基具有六个环碳原子(“C 6芳基”;例如,苯基)。在一些实施方案中,芳基具有十个环碳原子(“C 10芳基”;例如,萘基,例如,1-萘基和2-萘基)。芳基还包括其中上述芳基环与一个或多个环烷基或杂环基稠合的环系统,而且连接点在所述芳基环上,在这种情况下,碳原子的数目继续表示所述芳基环系统中的碳原子数目。芳基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“C 6-12芳烷基”表示基团-R-R’,其中R为烷基部分,R’为芳基部分,并且烷基和芳基总共具有6-12个碳原子。
“5-14元杂芳基”是指具有环碳原子和1-4个环杂原子的5-14元单环或双环的4n+2芳族环体系(例如,具有以环状排列共享的6、10或14个π电子)的基团,其中每个杂原子独立地选自氮、氧和硫。在含有一个或多个氮原子的杂芳基中,只要化合价允许,连接点可以是碳或氮原子。杂芳基双环系统在一个或两个环中可以包括一个或多个杂原子。杂芳基还包括其中上述杂芳基环与一个或多个环烷基或杂环基稠合的环系统,而且连接点在所述杂芳基环上,在这种情况下,碳原子的数目继续表示所述杂芳基环系统中的碳原子数目。在一些实施方案中,5-10元杂芳基是优选的,其为具有环碳原子和1-4个环杂原子的5-10元单环或双环的4n+2芳族环体系。在另一些实施方案中,5-6元杂芳基是特别优选的,其为具有环碳原子和1-4个环杂原子的5-6元单环或双环的4n+2芳族环体系。示例性的含有一个杂原子的5元杂芳基包括但不限于:吡咯基、呋喃基和噻吩基。示例性的含有两个杂原子的5元杂芳基包括但不限于:咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基和异噻唑基。示例性的含有三个杂原子的5元杂芳基包括但不限于:三唑基、噁二唑基(例如,1,2,4-噁二唑基)和噻二唑基。示例性的含有四个杂原子的5元杂芳基包括但不限于:四唑基。示例性的含有一个杂原子的6元杂芳基包括但不限于:吡啶基。示例性的含有两个杂原子的6元杂芳基包括但不限于:哒嗪基、嘧啶基和吡嗪基。示例性的含有三个或四个杂原子的6元杂芳基分别包括但不限于:三嗪基和四嗪基。示例性的含有一个杂原子的7元杂芳基包括但不限于:氮杂环庚三烯基、氧杂环庚三烯基和硫杂环庚三烯基。示例性的5,6-双环杂芳基包括但不限于:吲哚基、异吲哚基、吲唑基、苯并三唑基、苯并噻吩基、异苯并噻吩基、苯并呋喃基、苯并异呋喃基、苯并咪唑基、苯并噁唑基、苯并异噁唑基、苯并噁二唑基、苯并噻唑基、苯并异噻唑基、苯并噻二唑基、茚嗪基和嘌呤基。示例性的6,6-双环杂芳基包括但不限于:萘啶基、喋啶基、喹啉基、异喹啉基、噌琳基、喹喔啉基、酞嗪基和喹唑啉基。杂芳基基团可以被一或多个取代基任选取代,例如,被1至5个取代基、1至3个取代基或1个取代基取代。
“氧代”表示=O。
本文定义的烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基等为任选取代的基团。
示例性的碳原子上的取代基包括但不局限于:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR aa、-ON(R bb) 2、-N(R bb) 2、-N(R bb) 3 +X -、-N(OR cc)R bb、-SH、-SR aa、-SSR cc、-C(=O)R aa、-CO 2H、-CHO、-C(OR cc) 2、-CO 2R aa、-OC(=O)R aa、-OCO 2R aa、-C(=O)N(R bb) 2、-OC(=O)N(R bb) 2、-NR bbC(=O)R aa、-NR bbCO 2R aa、-NR bbC(=O)N(R bb) 2、-C(=NR bb)R aa、-C(=NR bb)OR aa、-OC(=NR bb)R aa、-OC(=NR bb)OR aa、-C(=NR bb)N(R bb) 2、-OC(=NR bb)N(R bb) 2、-NR bbC(=NR bb)N(R bb) 2、-C(=O)NR bbSO 2R aa、-NR bbSO 2R aa、-SO 2N(R bb) 2、-SO 2R aa、-SO 2OR aa、-OSO 2R aa、-S(=O)R aa、-OS(=O)R aa、-Si(R aa) 3、-OSi(R aa) 3、-C(=S)N(R bb) 2、-C(=O)SR aa、-C(=S)SR aa、-SC(=S)SR aa、-SC(=O)SR aa、-OC(=O)SR aa、-SC(=O)OR aa、-SC(=O)R aa、-P(=O) 2R aa、-OP(=O) 2R aa、-P(=O)(R aa) 2、-OP(=O)(R aa) 2、-OP(=O)(OR cc) 2、-P(=O) 2N(R bb) 2、-OP(=O) 2N(R bb) 2、 -P(=O)(NR bb) 2、-OP(=O)(NR bb) 2、-NR bbP(=O)(OR cc) 2、-NR bbP(=O)(NR bb) 2、-P(R cc) 2、-P(R cc) 3、-OP(R cc) 2、-OP(R cc) 3、-B(R aa) 2、-B(OR cc) 2、-BR aa(OR cc)、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
或者在碳原子上的两个偕氢被基团=O、=S、=NN(R bb) 2、=NNR bbC(=O)R aa、=NNR bbC(=O)OR aa、=NNR bbS(=O) 2R aa、=NR bb或=NOR cc取代;
R aa的每个独立地选自烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R aa基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R bb的每个独立地选自:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R bb基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R cc的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R cc基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R dd的每个独立地选自:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR ee、-ON(R ff) 2、-N(R ff) 2,、-N(R ff) 3 +X -、-N(OR ee)R ff、-SH、-SR ee、-SSR ee、-C(=O)R ee、-CO 2H、-CO 2R ee、-OC(=O)R ee、-OCO 2R ee、-C(=O)N(R ff) 2、-OC(=O)N(R ff) 2、-NR ffC(=O)R ee、-NR ffCO 2R ee、-NR ffC(=O)N(R ff) 2、-C(=NR ff)OR ee、-OC(=NR ff)R ee、-OC(=NR ff)OR ee、-C(=NR ff)N(R ff) 2、-OC(=NR ff)N(R ff) 2、-NR ffC(=NR ff)N(R ff) 2、-NR ffSO 2R ee、-SO 2N(R ff) 2、-SO 2R ee、-SO 2OR ee、-OSO 2R ee、-S(=O)R ee、-Si(R ee) 3、-OSi(R ee) 3、-C(=S)N(R ff) 2、-C(=O)SR ee、-C(=S)SR ee、-SC(=S)SR ee、-P(=O) 2R ee、-P(=O)(R ee) 2、-OP(=O)(R ee) 2、-OP(=O)(OR ee) 2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代,或者两个偕R dd取代基可结合以形成=O或=S;
R ee的每个独立地选自烷基、卤代烷基、烯基、炔基、环烷基、芳基、杂环基和杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代;
R ff的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R ff基团结合形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代;
R gg的每个独立地是:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OC 1-6烷基、-ON(C 1-6烷基) 2、-N(C 1-6烷基) 2、-N(C 1-6烷基) 3 +X -、-NH(C 1-6烷基) 2 +X -、-NH 2(C 1-6烷基) +X -、-NH 3 +X -、-N(OC 1-6烷基)(C 1-6烷基)、-N(OH)(C 1-6烷基)、-NH(OH)、-SH、-SC 1-6烷基、-SS(C 1-6烷基)、-C(=O)(C 1-6烷基)、-CO 2H、-CO 2(C 1-6烷基)、-OC(=O)(C 1-6烷基)、-OCO 2(C 1-6烷基)、-C(=O)NH 2、-C(=O)N(C 1-6烷基) 2、-OC(=O)NH(C 1-6烷基)、-NHC(=O)(C 1-6烷基)、-N(C 1-6烷基)C(=O)(C 1-6烷基)、-NHCO 2(C 1-6烷基)、-NHC(=O)N(C 1-6烷基) 2、-NHC(=O)NH(C 1-6烷基)、-NHC(=O)NH 2、-C(=NH)O(C 1-6烷基)、-OC(=NH)(C 1-6烷基)、-OC(=NH)OC 1-6烷基、-C(=NH)N(C 1-6烷基) 2、-C(=NH)NH(C 1-6烷基)、-C(=NH)NH 2、 -OC(=NH)N(C 1-6烷基) 2、-OC(NH)NH(C 1-6烷基)、-OC(NH)NH 2、-NHC(NH)N(C 1-6烷基) 2、-NHC(=NH)NH 2、-NHSO 2(C 1-6烷基)、-SO 2N(C 1-6烷基) 2、-SO 2NH(C 1-6烷基)、-SO 2NH 2、-SO 2C 1-6烷基、-SO 2OC 1-6烷基、-OSO 2C 1-6烷基、-SOC 1-6烷基、-Si(C 1-6烷基) 3、-OSi(C 1-6烷基) 3、-C(=S)N(C 1-6烷基) 2、C(=S)NH(C 1-6烷基)、C(=S)NH 2、-C(=O)S(C 1-6烷基)、-C(=S)SC 1-6烷基、-SC(=S)SC 1-6烷基、-P(=O) 2(C 1-6烷基)、-P(=O)(C 1-6烷基) 2、-OP(=O)(C 1-6烷基) 2、-OP(=O)(OC 1-6烷基) 2、C 1-6烷基、C 1-6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 7环烷基、C 6-C 10芳基、C 3-C 7杂环基、C 5-C 10杂芳基;或者两个偕R gg取代基可结合形成=O或=S;其中,X -为反离子。
示例性的氮原子上取代基包括但不局限于:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR bb)R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者连接至氮原子的两个R cc基团结合形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代,且其中R aa、R bb、R cc和R dd如上所述。
其它定义
术语“癌症”包括但不限于下列癌症:乳腺、卵巢、子宫颈、前列腺、睾丸、食道、胃、皮肤、肺、骨、结肠、胰腺、甲状腺、胆道、颊腔与咽(口)、唇、舌、口腔、咽、小肠、结肠直肠、大肠、直肠、脑与中枢神经系统的癌症、成胶质细胞瘤、神经母细胞瘤、角化棘皮瘤、表皮样癌、大细胞癌、腺癌、腺瘤、滤泡癌、未分化的癌、乳头状癌、精原细胞瘤、黑色素瘤、肉瘤、膀胱癌、肝癌、肾癌、骨髓障碍、淋巴障碍、霍奇金氏病、毛细胞癌和白血病。
本文所用的术语“治疗”涉及逆转、减轻、抑制该术语适用的障碍或病症的进展或者预防之,或者这类障碍或病症的一种或多种症状。本文所用的名词“治疗”涉及动词治疗的动作,后者是如刚才所定义的。
本文所用的术语“药学上可接受的盐”表示本发明化合物的那些羧酸盐、氨基酸加成盐,它们在可靠的医学判断范围内适用于与患者组织接触,不会产生不恰当的毒性、刺激作用、变态反应等,与合理的益处/风险比相称,就它们的预期应用而言是有效的,包括(可能的话)本发明化合物的两性离子形式。
药学上可接受的碱加成盐是与金属或胺生成的,例如碱金属与碱土金属氢氧化物或有机胺。用作阳离子的金属的实例有钠、钾、镁、钙等。适合的胺的实例有N,N'-二苄基乙二胺、氯普鲁卡因、胆碱、二乙醇胺、乙二胺、N-甲基葡糖胺和普鲁卡因。
酸性化合物的碱加成盐可以这样制备,按照常规方式使游离酸形式与足量所需的碱接触,生成盐。按照常规方式使盐形式与酸接触,再分离游离酸,可以使游离酸再生。游离酸形式在某些物理性质上多少不同于它们各自的盐形式,例如在极性溶剂中的溶解度,但是出于本发明的目的,盐还是等价于它们各自的游离酸。
盐可以是从无机酸制备的硫酸盐、焦硫酸盐、硫酸氢盐、亚硫酸盐、亚硫酸氢盐、硝酸盐、磷酸盐、磷酸一氢盐、磷酸二氢盐、偏磷酸盐、焦磷酸盐、氯化物、溴化物、碘化物,酸例如盐酸、硝酸、硫酸、氢溴酸、氢碘酸、磷酸等。代表性盐包括:氢溴酸盐、盐酸盐、硫酸盐、硫酸氢盐、硝酸盐、乙酸盐、草酸盐、戊酸盐、油酸盐、棕榈酸盐、硬脂酸盐、月桂酸盐、硼酸盐、苯甲酸盐、乳酸盐、磷酸盐、甲苯磺酸盐、柠檬酸盐、马来酸盐、富马酸盐、琥珀酸盐、酒石酸盐、萘甲酸盐、甲磺酸盐、葡庚糖酸盐、乳糖酸盐、月桂基磺酸盐和羟乙磺酸盐等。盐也可以是从有机酸制备的,例如脂肪族一元与二元羧酸、苯基取代的烷酸、羟基烷酸、烷二酸、芳香族酸、脂肪族与芳香族磺酸等。代表性盐 包括乙酸盐、丙酸盐、辛酸盐、异丁酸盐、草酸盐、丙二酸盐、琥珀酸盐、辛二酸盐、癸二酸盐、富马酸盐、马来酸盐、扁桃酸盐、苯甲酸盐、氯苯甲酸盆、甲基苯甲酸盐、二硝基苯甲酸盐、萘甲酸盐、苯磺酸盐、甲苯磺酸盐、苯乙酸盐、柠檬酸盐、乳酸盐、马来酸盐、酒石酸盐、甲磺酸盐等。药学上可接受的盐可以包括基于碱金属与碱土金属的阳离子,例如钠、锂、钾、钙、镁等,以及无毒的铵、季铵和胺阳离子,包括但不限于铵、四甲基铵、四乙基铵、甲胺、二甲胺、三甲胺、三乙胺、乙胺等。还涵盖氨基酸的盐,例如精氨酸盐、葡糖酸盐、半乳糖醛酸盐等(例如参见Berge S.M.et al.,"Pharmaceutical Salts,”J.Pharm.Sci.,1977;66:1-19,引入此作为参考)。
给药的“受试者”包括但不限于:人(即,任何年龄组的男性或女性,例如,儿科受试者(例如,婴儿、儿童、青少年)或成人受试者(例如,年轻的成人、中年的成人或年长的成人))和/或非人的动物,例如,哺乳动物,例如,灵长类(例如,食蟹猴、恒河猴)、牛、猪、马、绵羊、山羊、啮齿动物、猫和/或狗。在一些实施方案中,受试者是人。在一些实施方案中,受试者是非人动物。本文可互换使用术语“人”、“患者”和“受试者”。
“疾病”、“障碍”和“病症”在本文中可互换地使用。
除非另作说明,否则,本文使用的术语“治疗”包括受试者患有具体疾病、障碍或病症时所发生的作用,它降低疾病、障碍或病症的严重程度,或延迟或减缓疾病、障碍或病症的发展(“治疗性治疗”),还包括在受试者开始患有具体疾病、障碍或病症之前发生的作用(“预防性治疗”)。
通常,化合物的“有效量”是指足以引起目标生物反应的数量。正如本领域普通技术人员所理解的那样,本发明化合物的有效量可以根据下列因素而改变:例如,生物学目标、化合物的药代动力学、所治疗的疾病、给药模式以及受试者的年龄健康情况和症状。有效量包括治疗有效量和预防有效量。
除非另作说明,否则,本文使用的化合物的“治疗有效量”是在治疗疾病、障碍或病症的过程中足以提供治疗益处的量,或使与疾病、障碍或病症有关的一或多种症状延迟或最小化的量。化合物的治疗有效量是指单独使用或与其它疗法联用时,治疗剂的量,它在治疗疾病、障碍或病症的过程中提供治疗益处。术语“治疗有效量”可以包括改善总体治疗、降低或避免疾病或病症的症状或病因、或增强其它治疗剂的治疗效果的量。
除非另作说明,否则,本文使用的化合物的“预防有效量”是足以预防疾病、障碍或病症的量,或足以预防与疾病、障碍或病症有关的一或多种症状的量,或防止疾病、障碍或病症复发的量。化合物的预防有效量是指单独使用或与其它药剂联用时,治疗剂的量,它在预防疾病、障碍或病症的过程中提供预防益处。术语“预防有效量”可以包括改善总体预防的量,或增强其它预防药剂的预防效果的量。
“组合”以及相关术语是指同时或依次给药本发明化合物和其它治疗剂。例如,本发明化合物可以与其它治疗剂以分开的单位剂型同时或依次给药,或与其它治疗剂一起在单一单位剂型中同时给药。
附图说明
图1为化合物C176对EGFR L858R/T790M/C797S蛋白水平的影响图。
图2为化合物C213对EGFR Del19/T790M/C797S蛋白水平的影响图。
具体实施方案
本文中,“本发明化合物”指的是以下的式(X)化合物(包括子通式,例如式(I)、(I-1)、(I-5-1)等)、其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物。
本文中,化合物使用标准的命名法命名。具有非对称中心的化合物,应该明白(除非另有说明)所有的光学异构体及其混合物均包含在内。此外,除非另有规定,本发明所包括的所有异构体化合物与 碳碳双键可能以Z和E的形式出现。在不同的互变异构形式存在的化合物,一个所述化合物并不局限于任何特定的互变异构体,而是旨在涵盖所有的互变异构形式。
在一个实施方案中,本发明涉及通式(X)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000010
其中,
环A选自任选取代的以下基团:C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-10元杂芳基,其中所述取代基选自卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 3-7环烷基、4-8元杂环基、-P(O)(C 1-6烷基) 2、-P(O)(C 2-6烯基) 2、-O-C 1-6烷基、-O-C 1-6卤代烷基、-O-C 2-6烯基、-O-C 3-7环烷基、-O-4-8元杂环基、-NH-C 1-6烷基、-NH-C 2-6烯基、-NH-C 3-7环烷基、-NH-4-8元杂环基、-C(O)-C 1-6烷基、-C(O)-C 2-6烯基、-C(O)-C 3-7环烷基、-C(O)-4-8元杂环基、-S(O) 2-C 1-6烷基、-S(O) 2-C 2-6烯基、-S(O) 2-C 3-7环烷基、-S(O) 2-4-8元杂环基、-NHC(O)-C 1-6烷基、-NHC(O)-C 2-6烯基、-NHC(O)-C 3-7环烷基、-NHC(O)-4-8元杂环基、-NHS(O) 2-C 1-6烷基、-NHS(O) 2-C 2-6烯基、-NHS(O) 2-C 3-7环烷基或-NHS(O) 2-4-8元杂环基;
环B选自以下基团:
Figure PCTCN2020110442-appb-000011
Figure PCTCN2020110442-appb-000012
表示单键或双键;
Figure PCTCN2020110442-appb-000013
表示与分子其他部分的连接位置可以位于所在环的可用位置;
Z 1为O、S、N或C原子,其任选地被一个或两个R Z1取代;或者Z 1不存在,因而Z 4连接到Z 2、Z 3或与Z 1相连的芳环上的C原子,并且与Z 1相连的Z 2和芳环上的C原子分别与R W相连;或者Z 1、Z 2和Z 3都不存在,因而Z 4连接到与Z 1或Z 3相连的芳环上的C原子之一,并且芳环上的另一个C原子与R W相连;
Z 2为O、S、N或C原子,其任选地被一个或两个R Z2取代;
Z 3为O、S、N或C原子,其任选地被一个或两个R Z3取代;条件是,当
Figure PCTCN2020110442-appb-000014
为双键时,Z 2为N或C原子,Z 3为N或C原子;
Z 4为N或CR Z4
Z 5为N或CR Z5
R a、R b和R c独立地为H、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基或C 1-6卤代烷基;或者R a、R b以及它们连接的C原子一起形成C=O、C 3-7环烷基或4-8元杂环基;或者R a和R c以及它们连接的C原子一起形成C 3-7环烷基或4-8元杂环基;或者R a和R c形成化学键;
R N1为H、C 1-6烷基或C 1-6卤代烷基,优选H;
R Z1为不存在、H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;或者两个R Z1与Z 1一起形成C=O、C 3-7环烷基或 4-8元杂环基;
R Z2为不存在、H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;或者两个R Z2与Z 2一起形成C=O、C 3-7环烷基或4-8元杂环基;
R Z3为不存在、H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;或者两个R Z3与Z 3一起形成C=O、C 3-7环烷基或4-8元杂环基;
R Z4为H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基或C 1-6卤代烷基;
R Z5为H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;
或者Z 4所在的环不存在;
其中R w为H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、-C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基、-(CH 2) 0-5-4-8元杂环基、C 2-6烯基、C 2-6炔基、-(CH 2) 0-5-C 3-10卤代环烷基、-(CH 2) 0-5-C 6-10芳基或-(CH 2) 0-5-5-14元杂芳基;
R”为H、C 1-6烷基、C 1-6卤代烷基或-(CH 2) 0-5-C 3-7环烷基;
R”’为H、C 1-6烷基或C 1-6卤代烷基;
L 1选自化学键、-O-、-S(O) p-、-S(O)(=NR *)-、-NR #-、-CR #R #’-、-C aR #R #’-C bR #R #’-、-N=S(O)(R *)-或-S(O)(R *)=N-;
L 2选自化学键、-O-、-S(O) p-、-S(O)(=NR *)-、-NR #-、-CR #R #’-、-C aR #R #’-C bR #R #’-、-N=S(O)(R *)-或-S(O)(R *)=N-;
其中C aR #R #’或C bR #R #’中任一个可被O、S(O) p、S(O)(=NR *)或NR #替换,并且当C aR #R #’或C bR #R #’中任一个被O、S或NR #替换时,C aR #R #’或C bR #R #’中另一个还可被S(O) q替换;
E独立地选自:化学键、-C cR #R #’-C dR #R #’-C eR #R #’、
Figure PCTCN2020110442-appb-000015
Figure PCTCN2020110442-appb-000016
其中C cR #R #’、C dR #R #’或C eR #R #’中任一个,或者C cR #R #’和C eR #R #’二者可被O、S(O) p、S(O)(=NR *)或NR #替换,并且当C cR #R #’、C dR #R #’或C eR #R #’中任一个被O、S或NR #替换时,与之相邻的另一个或两个C cR #R #’、C dR #R #’或C eR #R #’还可被S(O) q替换;
或者两个E单元可以形成-CH 2CH 2OCH 2CH 2-、-OCH 2CH 2CH 2CH 2-、-CH 2CH 2CH 2CH 2O-、
Figure PCTCN2020110442-appb-000017
其中,
Figure PCTCN2020110442-appb-000018
表示与L 1或L 2的连接点;
H 1和H 2为N或C原子,H 3为O、S、N或C原子,并且H 1和H 3、H 2和H 3不同时为杂原子;
H 4和H 5为N或C原子;
H 6、H 7、H 8和H 9为C或N原子;
p为0、1或2;
q为1或2;
R *为H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10 元杂环基、C 6-10芳基或5-14元杂芳基;
R #为H、卤素、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基或5-14元杂芳基;
R #’为H、卤素、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基或5-14元杂芳基;
或者,相邻原子上的R #和R #可以形成化学键,相邻原子上的R #’和R #’可以形成化学键;
或者,相同或不同原子上的R #和R #’可以一起形成=O、或任选被R x取代的C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-6元杂芳基,其中所述R x为H、CN、卤素、C 1-6烷基或C 1-6卤代烷基;
m为0、1、2、3、4、5、6、7、8、9或10;
R s1选自H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-4-8元杂环基、C 2-6烯基、C 2-6炔基、-(CH 2) 0-5-C 3-7环烷基、-(CH 2) 0-5-C 3-10卤代环烷基、-(CH 2) 0-5-C 6-10芳基、-(CH 2) 0-5-5-14元杂芳基、-C(O)R W、-S(O)R W或-S(O) 2R W
s1为0、1、2或3;
R’选自H、-C(O)-C 1-6烷基、-C(O)-C 1-6卤代烷基、-C(O)-C 2-6烯基或-C(O)-C 6-10芳基;
L为化学键、-O-或-NR-;
其中R为H或C 1-6烷基;
Z为-N=或-C(R 4)=;
T选自化学键、C 2-6亚杂烷基、4-12元亚杂环基或5-6元亚杂环基取代的5-6元亚杂环基;
R 1选自H、卤素、氰基、C 1-6烷基、C 3-7环烷基、4-8元杂环基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
或者,当L为-NR-时,R 1、R与它们连接的原子一起形成任选取代的5-6元杂环基,其中所述取代基选自卤素、氧代、C 1-6烷基、C 1-6卤代烷基、卤素单取代或多取代的C 6-10芳基;
R 2为H、卤素、羟基、氨基、C 1-6烷基或C 1-6卤代烷基;
或者R 1、R 2与它们连接的原子一起形成5-6元杂环基或5-6元杂芳基;
R 3选自H、-O-C 1-6烷基或-O-C 1-6卤代烷基;
R 4选自H、卤素、C 1-6烷基、C 1-6卤代烷基、-NHC(O)-C 1-6烷基或-NHC(O)-C 2-6烯基;
当上述各基团为H或含H的基团时,所述一个或多个H原子可以被D原子取代;
上述烷基、亚烷基、卤代烷基、烯基、炔基、环烷基、卤代环烷基、杂环基、芳基、杂芳基或L 1、E、L 2、和T中含有OH、NH、NH 2、CH、CH 2、CH 3的基团在每次出现时各自任选地被1、2、3或更多个R s取代,其中所述R s在每次出现时独立地选自:卤素、羟基、氨基、氰基、硝基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、-OR a’、-OC(O)R a’、-C(O)R a’、-C(O)OR a’、-C(O)NR a’R b’、-S(O) nR a’、-S(O) nOR a’、-S(O) nNR a’R b’、-NR a’R b’、-NR a’C(O)R b’、-NR a’-C(O)OR b’、-NR a’-S(O) n-R b’、-NR a’C(O)NR a’R b’、-C 1-6亚烷基-R a’、-C 1-6亚烷基-OR a’、-C 1-6亚烷基-OC(O)R a’、-C 1-6亚烷基-C(O)OR a’、-C 1-6亚烷基-S(O) nR a’、-C 1-6亚烷基-S(O) nOR a’、-C 1-6亚烷基-OC(O)NR a’R b’、-C 1-6亚烷基-C(O)NR a’R b’、-C 1-6亚烷基-NR a’-C(O)NR a’R b’、-C 1-6亚烷基-OS(O) nR a’、-C 1-6亚烷基-S(O) nNR a’R b’、-C 1-6亚烷基-NR a’-S(O) nNR a’R b’、-C 1-6亚烷基-NR a’R b’和-O-C 1-6亚烷基-NR a’R b’,并且其中关于取代基R s所述的羟基、氨基、烷基、亚烷基、环烷基、杂环基、芳基、杂芳基和芳烷基进一步任选地被1、2、3或更多个独立地选自下列的取代基取代:卤素、OH、氨基、氰基、硝基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷基羟基、C 3-6环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基;
n每次出现时各自独立地为1或2;
R a’和R b’在每次出现时各自独立地选自H、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷基-O-、C 1-6烷基-S-、C 3-10环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基。
在另一个实施方案中,本发明涉及通式(X)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000019
其中,
环A选自任选取代的以下基团:C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-10元杂芳基,其中所述取代基选自卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 3-7环烷基、4-8元杂环基、-P(O)(C 1-6烷基) 2、-P(O)(C 2-6烯基) 2、-O-C 1-6烷基、-O-C 1-6卤代烷基、-O-C 2-6烯基、-O-C 3-7环烷基、-O-4-8元杂环基、-NH-C 1-6烷基、-NH-C 2-6烯基、-NH-C 3-7环烷基、-NH-4-8元杂环基、-C(O)-C 1-6烷基、-C(O)-C 2-6烯基、-C(O)-C 3-7环烷基、-C(O)-4-8元杂环基、-S(O) 2-C 1-6烷基、-S(O) 2-C 2-6烯基、-S(O) 2-C 3-7环烷基、-S(O) 2-4-8元杂环基、-NHC(O)-C 1-6烷基、-NHC(O)-C 2-6烯基、-NHC(O)-C 3-7环烷基、-NHC(O)-4-8元杂环基、-NHS(O) 2-C 1-6烷基、-NHS(O) 2-C 2-6烯基、-NHS(O) 2-C 3-7环烷基或-NHS(O) 2-4-8元杂环基;
环B选自以下基团:
Figure PCTCN2020110442-appb-000020
其中Y为-CH 2-或-C(O)-;
R’选自H、-C(O)-C 1-6烷基、-C(O)-C 1-6卤代烷基、-C(O)-C 2-6烯基或-C(O)-C 6-10芳基;
Figure PCTCN2020110442-appb-000021
表示与L 1的连接点;
L为化学键、-O-或-NR-;
其中R为H或C 1-6烷基;
Z为-N=或-C(R 4)=;
T选自化学键、C 2-6亚杂烷基、4-12元亚杂环基或5-6元亚杂环基取代的5-6元亚杂环基;
L 1选自化学键、-O-、-NH-、-CH 2-、-C(O)-、-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-CH 2O-、-NHCH 2-、-CH 2NH-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-C(O)O-、-NHC(O)-或-C(O)NH-;
E独立地选自-CH 2CH 2CH 2-、-CH 2CH 2C(O)-、-CH 2C(O)CH 2-、-C(O)CH 2CH 2-、-C(O)CH=CH-、-C(O)C≡C-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-C(O)CH 2O-、-OCH 2C(O)-、-CH 2C(O)O-、-OC(O)CH 2-、-C(O)OCH 2-、-CH 2OC(O)-、-CH 2CH 2NH-、-CH 2NHCH 2-、-NHCH 2CH 2-、-C(O)CH 2NH-、-NHCH 2C(O)-、-CH 2C(O)NH-、-NHC(O)CH 2-、-C(O)NHCH 2-、-CH 2NHC(O)-、
Figure PCTCN2020110442-appb-000022
Figure PCTCN2020110442-appb-000023
其中,
Figure PCTCN2020110442-appb-000024
表示与L 1或L 2的连接点;
m为0、1、2、3、4、5、6、7、8、9或10;
L 2选自化学键、-CH 2-、-CH 2CH 2-、-OCH 2-、-NHCH 2-、-OC(O)-、-NHC(O)-、-CH 2C(O)-或-C(O)CH 2-;
R 1选自H、卤素、氰基、C 1-6烷基、C 3-7环烷基、4-8元杂环基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
或者,当L为-NR-时,R 1、R与它们连接的原子一起形成任选取代的5-6元杂环基,其中所述取代基选自卤素、氧代、C 1-6烷基、C 1-6卤代烷基、卤素单取代或多取代的C 6-10芳基;
R 2为H、卤素、羟基、氨基、C 1-6烷基或C 1-6卤代烷基;
或者R 1、R 2与它们连接的原子一起形成5-6元杂环基或5-6元杂芳基;
R 3选自H、-O-C 1-6烷基或-O-C 1-6卤代烷基;
R 4选自H、卤素、C 1-6烷基、C 1-6卤代烷基、-NHC(O)-C 1-6烷基或-NHC(O)-C 2-6烯基;
上述烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基或L 1、E、L 2、T中含有OH、NH、NH 2、CH、CH 2、CH 3的基团在每次出现时各自任选地被1、2、3或更多个R s取代,其中所述R s在每次出现时独立地选自:卤素、羟基、氨基、氰基、硝基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、-OR a、-OC(O)R a、-C(O)R a、-C(O)OR a、-C(O)NR aR b、-S(O) nR a、-S(O) nOR a、-S(O) nNR aR b、-NR aR b、-NR aC(O)R b、-NR a-C(O)OR b、-NR a-S(O) n-R b、-NR aC(O)NR aR b、-C 1-6亚烷基-R a、-C 1-6亚烷基-OR a、-C 1-6亚烷基-OC(O)R a、-C 1-6亚烷基-C(O)OR a、-C 1-6亚烷基-S(O) nR a、-C 1-6亚烷基-S(O) nOR a、-C 1-6亚烷基-OC(O)NR aR b、-C 1-6亚烷基-C(O)NR aR b、-C 1-6亚烷基-NR a-C(O)NR aR b、-C 1-6亚烷基-OS(O) nR a、-C 1-6亚烷基-S(O) nNR aR b、-C 1-6亚烷基-NR a-S(O) nNR aR b、-C 1-6亚烷基-NR aR b和-O-C 1-6亚烷基-NR aR b,并且其中关于取代基R s所述的卤素、羟基、氨基、烷基、亚烷基、环烷基、杂环基、芳基、杂芳基和芳烷基进一步任选地被1、2、3或更多个独立地选自下列的取代基取代:卤素、OH、氨基、氰基、硝基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷基羟基、C 3-6环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基;
n每次出现时各自独立地为1或2;
R a和R b在每次出现时各自独立地选自H、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷基-O-、C 1-6烷基-S-、C 3-10环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基。
在更具体的实施方案中,本发明涉及式(I)或(I-G)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000025
其中,各基团定义如上定义。
在更具体的实施方案中,本发明涉及通式(I)或(I-G)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
环A为以下基团:
Figure PCTCN2020110442-appb-000026
其中,
R 7选自C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 3-7环烷基、-P(O)(C 1-6烷基) 2或-P(O)(C 2-6烯基) 2
R 8选自H、-NH-C 1-6烷基、-NH-C 2-6烯基、-NH-C 3-7环烷基、-NH-4-8元杂环基、-NHC(O)-C 1-6烷基、-NHC(O)-C 2-6烯基、-NHC(O)-C 3-7环烷基、-NHC(O)-4-8元杂环基、-NHS(O) 2-C 1-6烷基或-NHS(O) 2-C 3-7环烷基;
R 9选自H、卤素、-CN、C 1-6卤代烷基、-O-C 1-6烷基、-O-C 1-6卤代烷基、C 3-7环烷基、-O-C 3-7环烷基、C 1-6烷基、-NH-C 1-6烷基、-NH-C 2-6烯基、-NH-C 3-7环烷基、-NH-4-8元杂环基、-NHC(O)-C 1-6烷基、-NHC(O)-C 2-6烯基、-NHC(O)-C 3-7环烷基、-NHC(O)-4-8元杂环基、-NHS(O) 2-C 1-6烷基或-NHS(O) 2-C 3-7环烷基;
X为-C(R x)=或-N=;
其中,R x为H,或者R x连同R 8以及它们所连接的C原子一起形成5-6元杂环基或5-6元杂芳基;优选地,R x连同R 8以及它们所连接的C原子一起形成5-6元杂芳基,优选吡嗪基;
X 1为-CH(R X1)-或-N(R X1)-;
X 2为-CH(R X2)-或-N(R X2)-;
其中,R X1选自H、C 1-6烷基、C 3-7环烷基、-O-C 1-6烷基、-O-C 3-7环烷基、-NH-C 1-6烷基、-NH-C 3-7环烷基、-S(O) 2-C 1-6烷基、-S(O) 2-C 3-7环烷基、-NHS(O) 2-C 1-6烷基、-NHS(O) 2-C 3-7环烷基、-C(O)-C 1-6烷基、-C(O)-C 2-6烯基、-C(O)-C 3-7环烷基、-NHC(O)-C 1-6烷基、-NHC(O)-C 3-7环烷基或-C(O)-4-8元杂环基;R X2选自H、C 1-6烷基、C 3-7环烷基、-O-C 1-6烷基、-O-C 3-7环烷基、-NH-C 1-6烷基、-NH-C 3-7环烷基、-C(O)-C 1-6烷基、-C(O)-C 3-7环烷基、-C(O)-4-8元杂环基、-S(O) 2-C 1-6烷基、-S(O) 2-C 3-7环烷基、-NHS(O) 2-C 1-6烷基、-NHS(O) 2-C 3-7环烷基、-NHC(O)-C 1-6烷基、-NHC(O)-C 2-6烯基、-NHC(O)-C 3-7环烷基或-NHC(O)-4-8元杂环基;
Figure PCTCN2020110442-appb-000027
表示与L的连接点;
其他各基团定义如上定义。
在更具体的实施方案中,本发明涉及通式(I)或(I-G)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
T为
化学键、
Figure PCTCN2020110442-appb-000028
其中,
R 5为H或C 1-6烷基;
R 6为H或C 1-6烷基;
或者R 5和R 6连接形成C 1-6亚烷基;
Figure PCTCN2020110442-appb-000029
表示与母核或L 2的连接点;
其他各基团定义如上定义。
在更具体的实施方案中,本发明涉及通式(I)或(I-G)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,
当L为-NR-时,R 1和R一起形成以下基团:-C(O)N(R N)C(O)-或-C(C 1-6烷基)=C(R N)C(O)-;
其中,
R N选自C 1-6烷基或
Figure PCTCN2020110442-appb-000030
R 11为H或卤素;
R 12为H或卤素;
R 13为H或卤素;
Figure PCTCN2020110442-appb-000031
表示连接点;
其他各基团定义如上定义。
在更具体的实施方案中,本发明涉及通式(I)或(I-G)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
其中,R 1、R 2与它们连接的原子一起形成5-6元杂芳基;优选形成吡咯基;其他各基团定义如上定义。
环A
在一个具体实施方案中,环A为任选取代的C 3-7环烷基;在另一个具体实施方案中,环A为任选取代的4-8元杂环基;在另一个具体实施方案中,环A为任选取代的C 6-10芳基;在另一个具体实施方案中,环A为任选取代的5-10元杂芳基;在另一个具体实施方案中,环A为
Figure PCTCN2020110442-appb-000032
在另一 个具体实施方案中,环A为
Figure PCTCN2020110442-appb-000033
在另一个具体实施方案中,环A为
Figure PCTCN2020110442-appb-000034
在一个上述具体实施方案中,环A的取代基为卤素;在另一个上述具体实施方案中,环A的取代基为-CN;在另一个上述具体实施方案中,环A的取代基为C 1-6烷基;在另一个上述具体实施方案中,环A的取代基为C 1-6卤代烷基;在另一个上述具体实施方案中,环A的取代基为C 2-6烯基;在另一个上述具体实施方案中,环A的取代基为C 3-7环烷基;在另一个上述具体实施方案中,环A的取代基为4-8元杂环基;在另一个上述具体实施方案中,环A的取代基为-P(O)(C 1-6烷基) 2;在另一个上述具体实施方案中,环A的取代基为-P(O)(C 2-6烯基) 2;在另一个上述具体实施方案中,环A的取代基为-O-C 1-6烷基;在另一个上述具体实施方案中,环A的取代基为-O-C 1-6卤代烷基;在另一个上述具体实施方案中,环A的取代基为-O-C 2-6烯基;在另一个上述具体实施方案中,环A的取代基为-O-C 3-7环烷基;在另一个上述具体实施方案中,环A的取代基为-O-4-8元杂环基;在另一个上述具体实施方案中,环A的取代基为-NH-C 1-6烷基;在另一个上述具体实施方案中,环A的取代基为-NH-C 2-6烯基;在另一个上述具体实施方案中,环A的取代基为-NH-C 3-7环烷基;在另一个上述具体实施方案中,环A的取代基为-NH-4-8元杂环基;在另一个上述具体实施方案中,环A的取代基为-C(O)-C 1-6烷基;在另一个上述具体实施方案中,环A的取代基为-C(O)-C 2-6烯基;在另一个上述具体实施方案中,环A的取代基为-C(O)-C 3-7环烷基;在另一个上述具体实施方案中,环A的取代基为-C(O)-4-8元杂环基;在另一个上述具体实施方案中,环A的取代基为-S(O) 2-C 1-6烷基;在另一个上述具体实施方案中,环A的取代基为-S(O) 2-C 2-6烯基;在另一个上述具体实施方案中,环A的取代基为-S(O) 2-C 3-7环烷基;在另一个上述具体实施方案中,环A的取代基为-S(O) 2-4-8元杂环基;在另一个上述具体实施方案中,环A的取代基为-NHC(O)-C 1-6烷基;在另一个上述具体实施方案中,环A的取代基为-NHC(O)-C 2-6烯基;在另一个上述具体实施方案中,环A的取代基为-NHC(O)-C 3-7环烷基;在另一个上述具体实施方案中,环A的取代基为-NHC(O)-4-8元杂环基;在另一个上述具体实施方案中,环A的取代基为-NHS(O) 2-C 1-6烷基;在另一个上述具体实施方案中,环A的取代基为-NHS(O) 2-C 2-6烯基;在另一个上述具体实施方案中,环A的取代基为-NHS(O) 2-C 3-7环烷基;在另一个上述具体实施方案中,环A的取代基为-NHS(O) 2-4-8元杂环基。
L
在一个具体实施方案中,L为化学键;在另一个具体实施方案中,L为-O-;在另一个具体实施方案中,L为-NR-。
Y
在一个具体实施方案中,Y为-CH 2-;在另一个具体实施方案中,Y为-C(O)-。
Z
在一个具体实施方案中,Z为-N=;在另一个具体实施方案中,Z为-C(R 4)=。
T
在一个具体实施方案中,T为C 2-6亚杂烷基;在另一个具体实施方案中,T为4-12元亚杂环基;在另一个具体实施方案中,T为5-6元亚杂环基取代的5-6元亚杂环基;在另一个具体实施方案中,T 为
Figure PCTCN2020110442-appb-000035
在另一个具体实施方案中,T为
Figure PCTCN2020110442-appb-000036
在另一个具体实施方案中,T为
Figure PCTCN2020110442-appb-000037
在另一个具体实施方案中,T为
Figure PCTCN2020110442-appb-000038
在另一个具体实施方案中,T为化学键。
Figure PCTCN2020110442-appb-000039
在一个具体实施方案中,
Figure PCTCN2020110442-appb-000040
表示单键;在另一个具体实施方案中,
Figure PCTCN2020110442-appb-000041
表示双键。
Z 1
在一个具体实施方案中,Z 1为O原子;在另一个具体实施方案中,Z 1为S原子;在另一个具体实施方案中,Z 1为N原子;在另一个具体实施方案中,Z 1为C原子;在另一个具体实施方案中,Z 1被一个R Z1取代;在另一个具体实施方案中,Z 1被两个R Z1取代;在另一个具体实施方案中,Z 1不存在。
Z 2
在一个具体实施方案中,Z 2为O原子;在另一个具体实施方案中,Z 2为S原子;在另一个具体实施方案中,Z 2为N原子;在另一个具体实施方案中,Z 2为C原子;在另一个具体实施方案中,Z 2被一个R Z2取代;在另一个具体实施方案中,Z 2被两个R Z2取代。
Z 3
在一个具体实施方案中,Z 3为O原子;在另一个具体实施方案中,Z 3为S原子;在另一个具体实施方案中,Z 3为N原子;在另一个具体实施方案中,Z 3为C原子;在另一个具体实施方案中,Z 3被一个R Z3取代;在另一个具体实施方案中,Z 3被两个R Z3取代。
在一个具体实施方案中,Z 1、Z 2和Z 3都不存在。
Z 4
在一个具体实施方案中,Z 4为N;在另一个具体实施方案中,Z 4为CR Z4
Z 5
在一个具体实施方案中,Z 5为N;在另一个具体实施方案中,Z 5为CR Z5
R a、R b和R c
在一个具体实施方案中,R a为H;在另一个具体实施方案中,R a为卤素;在另一个具体实施方案中,R a为OR’;在另一个具体实施方案中,R a为NR’R”;在另一个具体实施方案中,R a为C 1-6烷基;在另一个具体实施方案中,R a为C 1-6卤代烷基。
在一个具体实施方案中,R b为H;在另一个具体实施方案中,R b为卤素;在另一个具体实施方案中,R b为OR’;在另一个具体实施方案中,R b为NR’R”;在另一个具体实施方案中,R b为C 1-6烷 基;在另一个具体实施方案中,R b为C 1-6卤代烷基。
在一个具体实施方案中,R c为H;在另一个具体实施方案中,R c为卤素;在另一个具体实施方案中,R c为OR’;在另一个具体实施方案中,R c为NR’R”;在另一个具体实施方案中,R c为C 1-6烷基;在另一个具体实施方案中,R c为C 1-6卤代烷基。
在一个具体实施方案中,R a、R b以及它们连接的C原子一起形成C=O;在另一个具体实施方案中,R a、R b以及它们连接的C原子一起形成C 3-7环烷基;在另一个具体实施方案中,R a、R b以及它们连接的C原子一起形成4-8元杂环基;在另一个具体实施方案中,R a和R c形成化学键;在另一个具体实施方案中,R a和R c以及它们连接的C原子一起形成C 3-7环烷基或4-8元杂环基。
R Z1
在一个具体实施方案中,R Z1不存在;在另一个具体实施方案中,R Z1为H;在另一个具体实施方案中,R Z1为CN;在另一个具体实施方案中,R Z1为卤素;在另一个具体实施方案中,R Z1为-(CH 2) 0-5-OR’;在另一个具体实施方案中,R Z1为-(CH 2) 0-5-NR’R”;在另一个具体实施方案中,R Z1为C 1-6烷基;在另一个具体实施方案中,R Z1为C 1-6卤代烷基;在另一个具体实施方案中,R Z1为-(CH 2) 0-5-C 3-7环烷基;在另一个具体实施方案中,R Z1为-(CH 2) 0-5-4-8元杂环基;在另一个具体实施方案中,两个R Z1与Z 1一起形成C=O;在另一个具体实施方案中,两个R Z1与Z 1一起形成C 3-7环烷基;在另一个具体实施方案中,两个R Z1与Z 1一起形成4-8元杂环基。
R Z2
在一个具体实施方案中,R Z2不存在;在另一个具体实施方案中,R Z2为H;在另一个具体实施方案中,R Z2为CN;在另一个具体实施方案中,R Z2为卤素;在另一个具体实施方案中,R Z2为-(CH 2) 0-5-OR’;在另一个具体实施方案中,R Z2为-(CH 2) 0-5-NR’R”;在另一个具体实施方案中,R Z2为C 1-6烷基;在另一个具体实施方案中,R Z2为C 1-6卤代烷基;在另一个具体实施方案中,R Z2为-(CH 2) 0-5-C 3-7环烷基;在另一个具体实施方案中,R Z2为-(CH 2) 0-5-4-8元杂环基;在另一个具体实施方案中,两个R Z2与Z 2一起形成C=O;在另一个具体实施方案中,两个R Z2与Z 2一起形成C 3-7环烷基;在另一个具体实施方案中,两个R Z2与Z 2一起形成4-8元杂环基。
R Z3
在一个具体实施方案中,R Z3不存在;在另一个具体实施方案中,R Z3为H;在另一个具体实施方案中,R Z3为CN;在另一个具体实施方案中,R Z3为卤素;在另一个具体实施方案中,R Z3为-(CH 2) 0-5-OR’;在另一个具体实施方案中,R Z3为-(CH 2) 0-5-NR’R”;在另一个具体实施方案中,R Z3为C 1-6烷基;在另一个具体实施方案中,R Z3为C 1-6卤代烷基;在另一个具体实施方案中,R Z3为-(CH 2) 0-5-C 3-7环烷基;在另一个具体实施方案中,R Z3为-(CH 2) 0-5-4-8元杂环基;在另一个具体实施方案中,两个R Z3与Z 3一起形成C=O;在另一个具体实施方案中,两个R Z3与Z 3一起形成C 3-7环烷基;在另一个具体实施方案中,两个R Z3与Z 3一起形成4-8元杂环基。
R Z4
在一个具体实施方案中,R Z4为H;在另一个具体实施方案中,R Z4为CN;在另一个具体实施方案中,R Z4为卤素;在另一个具体实施方案中,R Z4为-(CH 2) 0-5-OR’;在另一个具体实施方案中,R Z4为-(CH 2) 0-5-NR’R”;在另一个具体实施方案中,R Z4为C 1-6烷基;在另一个具体实施方案中,R Z4为C 1-6卤代烷基。
R Z5
在一个具体实施方案中,R Z5为H;在另一个具体实施方案中,R Z5为CN;在另一个具体实施方案中,R Z5为卤素;在另一个具体实施方案中,R Z5为-(CH 2) 0-5-OR’;在另一个具体实施方案中,R Z5为-(CH 2) 0-5-NR’R”;在另一个具体实施方案中,R Z5为C 1-6烷基;在另一个具体实施方案中,R Z5为C 1-6卤代烷基;在另一个具体实施方案中,R Z5为-(CH 2) 0-5-C 3-7环烷基;在另一个具体实施方案中,R Z5为-(CH 2) 0-5-4-8元杂环基。
在一个具体实施方案中,Z 4所在的环不存在。
L 1
在一个具体实施方案中,L 1为化学键;在另一个具体实施方案中,L 1为-O-;在另一个具体实施方案中,L 1为-S(O) p-;在另一个具体实施方案中,L 1为-S(O)(=NR *)-;在另一个具体实施方案中,L 1为-NR #-;在另一个具体实施方案中,L 1为-CR #R #’-;在另一个具体实施方案中,L 1为-C aR #R #’-C bR #R #’-;在另一个具体实施方案中,L 1为-N=S(O)(R *)-;在另一个具体实施方案中,L 1为-S(O)(R *)=N-。
L 2
在一个具体实施方案中,L 2为化学键;在另一个具体实施方案中,L 2为-O-;在另一个具体实施方案中,L 2为-S(O) p-;在另一个具体实施方案中,L 2为-S(O)(=NR *)-;在另一个具体实施方案中,L 2为-NR #-;在另一个具体实施方案中,L 2为-CR #R #’-;在另一个具体实施方案中,L 2为-C aR #R #’-C bR #R #’-;在另一个具体实施方案中,L 2为-N=S(O)(R *)-;在另一个具体实施方案中,L 2为-S(O)(R *)=N-。
在另一个具体实施方案中,L 1或L 2中的C aR #R #’或C bR #R #’中任一个可被O、S(O) p、S(O)(=NR *)或NR #替换,并且当C aR #R #’或C bR #R #’中任一个被O、S或NR #替换时,C aR #R #’或C bR #R #’中另一个还可被S(O) q替换。
E
在一个具体实施方案中,E为化学键;在另一个具体实施方案中,E为-C cR #R #’-C dR #R #’-C eR #R #’;在另一个具体实施方案中,E为
Figure PCTCN2020110442-appb-000042
在另一个具体实施方案中,E为
Figure PCTCN2020110442-appb-000043
在另一个具体实施方案中,E为
Figure PCTCN2020110442-appb-000044
在另一个具体实施方案中,E为
Figure PCTCN2020110442-appb-000045
在另一个具体实施方案中,E为
Figure PCTCN2020110442-appb-000046
在另一个具体实施方案中,C cR #R #’、C dR #R #’或C eR #R #’中任一个,或者C cR #R #’和C eR #R #’二者可被O、S(O) p、S(O)(=NR *)或NR #替换,并且当C cR #R #’、C dR #R #’或C eR #R #’中任一个被O、S或NR #替换时,与之相邻的另一个或两个C cR #R #’、C dR #R #’或C eR #R #’还可被S(O) q替换;
在另一个具体实施方案中,两个E单元可以形成-CH 2CH 2OCH 2CH 2-;在另一个具体实施方案中,两个E单元可以形成-OCH 2CH 2CH 2CH 2-;在另一个具体实施方案中,两个E单元可以形成-CH 2CH 2CH 2CH 2O-;在另一个具体实施方案中,两个E单元可以形成
Figure PCTCN2020110442-appb-000047
在另一个具体 实施方案中,两个E单元可以形成
Figure PCTCN2020110442-appb-000048
在另一个具体实施方案中,两个E单元可以形成
Figure PCTCN2020110442-appb-000049
在另一个具体实施方案中,两个E单元可以形成
Figure PCTCN2020110442-appb-000050
在另一个具体实施方案中,两个E单元可以形成
Figure PCTCN2020110442-appb-000051
在另一个具体实施方案中,两个E单元可以形成
Figure PCTCN2020110442-appb-000052
在另一个具体实施方案中,在L 1、L 2或E的实施方案中,相邻原子上的R #和R #可以形成化学键,相邻原子上的R #’和R #’可以形成化学键;
在另一个具体实施方案中,在L 1、L 2或E的实施方案中,相同原子上的R #和R #’可以一起形成=O、或任选被R x取代的C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-6元杂芳基;在另一个具体实施方案中,在L 1、L 2或E的实施方案中,不同原子上的R #和R #’可以一起形成任选被R x取代的C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-6元杂芳基。
m
在一个具体实施方案中,m为0;在另一个具体实施方案中,m为1;在另一个具体实施方案中,m为2;在另一个具体实施方案中,m为3;在另一个具体实施方案中,m为4;在另一个具体实施方案中,m为5;在另一个具体实施方案中,m为6;在另一个具体实施方案中,m为7;在另一个具体实施方案中,m为8;在另一个具体实施方案中,m为9;在另一个具体实施方案中,m为10。
R 1
在一个具体实施方案中,R 1为H;在另一个具体实施方案中,R 1为卤素;在另一个具体实施方案中,R 1为氰基;在另一个具体实施方案中,R 1为C 1-6烷基;在另一个具体实施方案中,R 1为C 3-7环烷基;在另一个具体实施方案中,R 1为4-8元杂环基;在另一个具体实施方案中,R 1为C 1-6卤代烷基;在另一个具体实施方案中,R 1为C 2-6烯基;在另一个具体实施方案中,R 1为C 2-6炔基。在另一个具体实施方案中,当L为-NR-时,R 1、R与它们连接的原子一起形成任选取代的5-6元杂环基,其中所述取代基选自卤素、氧代、C 1-6烷基、C 1-6卤代烷基、卤素单取代或多取代的C 6-10芳基;在另一个具体实施方案中,R 1和R一起形成-C(O)N(R N)C(O)-;在另一个具体实施方案中,R 1和R一起形成-C(C 1-6烷基)=C(R N)C(O)-。
R 2
在一个具体实施方案中,R 2为H;在另一个具体实施方案中,R 2为卤素;在另一个具体实施方案中,R 2为羟基;在另一个具体实施方案中,R 2为氨基;在另一个具体实施方案中,R 2为C 1-6烷基;在另一个具体实施方案中,R 2为C 1-6卤代烷基;在另一个具体实施方案中,R 1、R 2与它们连接的原子一起形成5-6元杂环基;在另一个具体实施方案中,R 1、R 2与它们连接的原子一起形成5-6元杂芳基;在另一个具体实施方案中,R 1、R 2与它们连接的原子一起形成吡咯基。
R 3
在一个具体实施方案中,R 3为H;在另一个具体实施方案中,R 3为H-O-C 1-6烷基;在另一个具 体实施方案中,R 3为-O-C 1-6卤代烷基。
R 4
在一个具体实施方案中,R 4为H;在另一个具体实施方案中,R 4为卤素;在另一个具体实施方案中,R 4为C 1-6烷基;在另一个具体实施方案中,R 4为C 1-6卤代烷基;在另一个具体实施方案中,R 4为-NHC(O)-C 1-6烷基;在另一个具体实施方案中,R 4为-NHC(O)-C 2-6烯基。
以上任一具体实施方案中的任一技术方案或其任意组合,可以与其它具体实施方案中的任一技术方案或其任意组合进行组合。例如,环A的任一技术方案或其任意组合,可以与L、Y、Z 1-Z 5、R’、T、L 1、E、m、L 2、R 1-R 4等的任一技术方案或其任意组合进行组合。本发明旨在包括所有这些技术方案的组合,限于篇幅,不再一一列出。
在更具体的实施方案中,本发明提供了通式(I)或(I-G)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
环A选自任选取代的以下基团:C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-10元杂芳基,其中所述取代基选自-F、-Cl、-Br、-Me、-OMe、-CF 3、-OCF 3、-CN、-NHMe、环丙基、-P(O)Me 2、-NHC(O)CH 2CH 3、-C(O)CH=CH 2、-NHS(O) 2CH 2CH 3、-NH-环丙基、-NHC(O)CH=CH 2或-C(O)CH 2CH 3;环A优选为以下基团:
Figure PCTCN2020110442-appb-000053
其中,
R 7选自-Me、环丙基或-P(O)Me 2
R 8选自H、-NHMe、-NHC(O)CH 2CH 3或-NH-环丙基;
R 9选自H、-F、-Cl、-Br、-Me、-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH 2CH 3、-NHS(O) 2CH 2CH 3、-NHC(O)CH=CH 2或-NH-环丙基;
X为-C(R x)=或-N=;
其中,R x为H,或者R x连同R 8以及它们所连接的C原子一起形成5-6元杂环基或5-6元杂芳基;优选地为5-6元杂芳基(优选吡嗪基);
X 1为-CH 2-或-N(R X1)-;
X 2为-CH(R X2)-或-N(R X2)-;
其中,R X1为-C(O)CH 2CH 3或-C(O)CH=CH 2;R X2为H、-Me、-OMe、-NHMe、-C(O)CH 2CH 3、-NHS(O) 2CH 2CH 3或-NHC(O)CH 2CH 3
Figure PCTCN2020110442-appb-000054
表示与L的连接点;
T选自化学键、C 2-6亚杂烷基、4-12元亚杂环基或5-6元亚杂环基取代的5-6元亚杂环基;优选为以下基团:
Figure PCTCN2020110442-appb-000055
其中,
R 5为-Me;
R 6为-Me;
或者R 5和R 6连接形成-CH 2CH 2-;
Figure PCTCN2020110442-appb-000056
表示与母核或L 2的连接点;
R 1选自H、-Cl、-Br、-CH 3、-CF 3、环丙基或-CH=CH 2
或者,当L为-NR-时,R 1、R与它们连接的原子一起形成任选取代的5-6元杂环基,其中所述取代基选自卤素、氧代、-iPr、-Et、卤素单取代或多取代的苯基;R 1和R优选形成以下基团:-C(O)N(R N)C(O)-或-C(CH 3)=C(R N)C(O)-;
其中,
R N选自-iPr、-Et或
Figure PCTCN2020110442-appb-000057
R 11为-Cl、-Br;
R 12为H或-F;
R 13为H或-F;
Figure PCTCN2020110442-appb-000058
表示连接点;
R 2为H;
或者R 1、R 2与它们连接的原子一起形成5-6元杂环基或5-6元杂芳基;优选形成吡咯基;
R 3选自H或-OMe;
R 4选自H、-F、-Me、-CF 3或-NHC(O)CH=CH 2
其他各基团定义如上定义;
在上述通式(I)或(I-G)化合物的一个方案中,L 1选自化学键、-O-、-NH-、-CH 2-、-C(O)-、-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-CH 2O-、-NHCH 2-、-CH 2NH-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-C(O)O-、-NHC(O)-或-C(O)NH-;
E独立地选自-CH 2CH 2CH 2-、-CH 2CH 2C(O)-、-CH 2C(O)CH 2-、-C(O)CH 2CH 2-、-C(O)CH=CH-、-C(O)C≡C-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-C(O)CH 2O-、-OCH 2C(O)-、-CH 2C(O)O-、-OC(O)CH 2-、-C(O)OCH 2-、-CH 2OC(O)-、-CH 2CH 2NH-、-CH 2NHCH 2-、-NHCH 2CH 2-、-C(O)CH 2NH-、-NHCH 2C(O)-、-CH 2C(O)NH-、-NHC(O)CH 2-、-C(O)NHCH 2-、-CH 2NHC(O)-、
Figure PCTCN2020110442-appb-000059
Figure PCTCN2020110442-appb-000060
Figure PCTCN2020110442-appb-000061
m为0、1、2、3、4、5、6、7、8、9或10;
L 2选自化学键、-CH 2-、-CH 2CH 2-、-OCH 2-、-NHCH 2-、-OC(O)-、-NHC(O)-、-CH 2C(O)-或-C(O)CH 2-;
其中
Figure PCTCN2020110442-appb-000062
表示与L 1或L 2的连接点。
在更具体的实施方案中,本发明提供了通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物具有以下通式结构:
Figure PCTCN2020110442-appb-000063
Figure PCTCN2020110442-appb-000064
Figure PCTCN2020110442-appb-000065
Figure PCTCN2020110442-appb-000066
Figure PCTCN2020110442-appb-000067
Figure PCTCN2020110442-appb-000068
Figure PCTCN2020110442-appb-000069
其中各基团如上所定义。
在更具体的实施方案中,本发明提供了通式(I-1)、(I-1-A)、(I-1-B)、(I-1-C)、(I-1-D)、(I-1-E)、(I-1-F)、(I-1-G)、(I-1-H)或(I-1-I)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
R 1选自-Cl、-Br、-CF 3或-CH=CH 2;优选地,R 1选自-Cl或-Br;
R 4为H或-Me;
R 8为H、-NHMe、-NHC(O)CH 2CH 3或-NH-环丙基;
R 9为H、-F、-Cl、-Br、-Me、-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH 2CH 3、-NHS(O) 2CH 2CH 3或-NHC(O)CH=CH 2
X为-C(R x)=或-N=;
R x为H,或R x连同R 8以及它们所连接的C原子一起形成吡嗪基;
并且,其他基团如上所定义。
在更具体的实施方案中,本发明提供了通式(I-G)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000070
其中,
环A为以下基团:
Figure PCTCN2020110442-appb-000071
其中,
R 7为-P(O)(C 1-6烷基) 2
R 8为H;
R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;
X为-C(R x)=;
其中,R x为H,或者R x连同R 8以及它们所连接的C原子一起形成5-6元杂芳基;
优选地为吡嗪基;
Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2或C=O;优选地,Y为CH 2
L 1为-C aR #R #’-C bR #R #’-;
L 2选自化学键、-CR #R #’-或-C aR #R #’-C bR #R #’-;
其中C aR #R #’或C bR #R #’中任一个可被O、S(O) p或NR #替换;
E独立地选自:化学键或-C cR #R #’-C dR #R #’-C eR #R #’;
其中C cR #R #’、C dR #R #’或C eR #R #’中任一个,或者C cR #R #’和C eR #R #’二者可被O、S(O) p或NR #替换;
p为0、1或2;
R #为H、卤素、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R #’为H、卤素、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
或者,相邻原子上的R #和R #可以形成化学键,相邻原子上的R #’和R #’可以形成化学键;
或者,相同或不同原子上的R #和R #’可以一起形成=O;
m为0、1、2、3、4或5;
R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;
s1为0、1、2或3;
L为-NR-;其中R为H或C 1-6烷基;
Z为-C(R 4)=;
T为
Figure PCTCN2020110442-appb-000072
R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
R 2为H、卤素、羟基、氨基、C 1-6烷基或C 1-6卤代烷基;
R 3选自H、-O-C 1-6烷基或-O-C 1-6卤代烷基;
R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;
当上述各基团为H或含H的基团时,所述一个或多个H原子可以被D原子取代。
在更具体的实施方案中,本发明提供了通式(I-1-G)、(I-1-H)、(I-1-H’)、(I-1-H”)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000073
其中,
X为-CH=;
Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2或C=O;
R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为H或卤素;
R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4为H;
R 8为H;
R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-,并且上述基团中的一个或多个H原子可以被D原子取代;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-或-NHCH 2-;优选地,L 1选自-CH 2CH 2-、-C≡C-或-OCH 2-;
L 2选自化学键、-CH 2-或-CH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;
E为-CH 2CH 2CH 2-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-或-SCH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;优选地,E为-CH 2CH 2CH 2-;
m为0、1、2或3;优选地,-L 1-(E) m-L 2-的链长度优选地为4-14个键长,更优选5、6、7、8、9或10个键长;
R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H、CN或卤素;
s1为0、1或2。
在更具体的实施方案中,本发明提供了通式(I-1-G)、(I-1-H)、(I-1-H’)、(I-1-H”)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000074
其中,
X为-CH=;
Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2
R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为卤素;
R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4为H;
R 8为H;
R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-,并且上述基团中的一个或多个H原子可以被D原子取代;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-或-NHCH 2-;优选地,L 1选自-CH 2CH 2-、-C≡C-、-OCH 2-或-NHCH 2-;
L 2选自化学键、-CH 2-或-CH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;
E为-CH 2CH 2CH 2-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-或-SCH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;优选地,E为-CH 2CH 2CH 2-;
m为0、1、2或3;优选地,-L 1-(E) m-L 2-的链长度优选地为4-14个键长,更优选5、6、7、8、9或10个键长;
R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H或卤素;
s1为0、1或2。
在更具体的实施方案中,本发明提供了通式(I-1-G)、(I-1-H)、(I-1-H’)、(I-1-H”)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000075
其中,
X为-CH=;
Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2或C=O;
R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为卤素;
R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4为H;
R 8为H;
R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-,并且上述基团中的一个或多个H原子可以被D原子取代;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-或-NHCH 2-;优选地,L 1选自-C≡C-、-OCH 2-或-NHCH 2-;
L 2选自化学键、-CH 2-或-CH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;
E为-CH 2CH 2CH 2-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-或-SCH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;优选地,E为-CH 2CH 2CH 2-;
m为1、2或3;优选地,-L 1-(E) m-L 2-的链长度小于14个键长;优选地,链长度为5-10个键长,优选5、6、7、8、9或10个键长;
R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H或卤素;
s1为0、1或2。
在更具体的实施方案中,本发明提供了通式(I-1-I)、(I-1-I’)、(I-1-I”)、(I-1-I”’)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000076
其中,
Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2或C=O;
R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为卤素;
R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4选自C 1-6烷基或C 1-6卤代烷基;
R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-;优选地,L 1选 自-CH 2CH 2-、-CH=CH-、-C≡C-或-OCH 2-;优选地,L 1选自-CH 2CH 2-、-C≡C-或-OCH 2-;
L 2选自化学键、-CH 2-或-CH 2CH 2-;
E为-CH 2CH 2CH 2-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-或-SCH 2CH 2-;优选地,E为-CH 2CH 2CH 2-;
m为1或2;优选地,-L 1-(E) m-L 2-的链长度为4-14个键长,更优选5、6、7、8、9或10个键长;
R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H、卤素或CN;
s1为0、1或2。
在更具体的实施方案中,本发明提供了通式(I-1-I)、(I-1-I’)、(I-1-I”)、(I-1-I”’)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000077
其中,
Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2
R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为卤素;
R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4选自C 1-6烷基或C 1-6卤代烷基;
R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-或-OCH 2-;优选地,L 1选自-C≡C-或-OCH 2-;
L 2选自化学键、-CH 2-或-CH 2CH 2-;
E为-CH 2CH 2CH 2-;
m为1或2;优选地,-L 1-(E) m-L 2-的链长度小于14个键长;优选地,更优选5、6、7、8、9或10个键长;
R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H或卤素;
s1为0、1或2。
在更具体的实施方案中,本发明提供了通式(I-2-I)、(I-2-I’)、(I-2-I”)、(I-2-I”’)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000078
其中,
Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2
R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为卤素;
R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4选自C 1-6烷基或C 1-6卤代烷基;
R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-或-OCH 2-;优选地,L 1选自-C≡C-或-OCH 2-;
L 2选自化学键、-CH 2-或-CH 2CH 2-;
E为-CH 2CH 2CH 2-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-或-SCH 2CH 2-;优选地,E为-CH 2CH 2CH 2-;
m为1或2;优选地,-L 1-(E) m-L 2-的链长度小于14个键长;优选5、6、7、8、9或10个键长;
R s1选自H、CN、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H或卤素;
s1为0、1或2。
在更具体的实施方案中,本发明提供了通式(I-2)、(I-2-A)、(I-2-B)、(I-2-C)、(I-2-D)、(I-2-E)、(I-2-F)、(I-2-G)、(I-2-H)或(I-2-I)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
R 1为-Cl、-Br或-CH=CH 2
R 4为H或-Me;
R 8为H、-NHMe、-NHC(O)CH 2CH 3或-NH-环丙基;
R 9为H、-F、-Cl、-Br、-Me、-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH 2CH 3、-NHS(O) 2CH 2CH 3或-NHC(O)CH=CH 2
X为-C(R x)=;
R x为H,或R x连同R 8以及它们所连接的C原子一起形成吡嗪基;
并且,其他基团如上所定义。
在更具体的实施方案中,本发明提供了通式(I-3)、(I-3-A)、(I-3-B)、(I-3-C)、(I-3-D)、(I-3-E)、(I-3-F)、(I-3-G)或(I-3-H)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
R 1为H、-Cl或-CH=CH 2
R 4为-NHC(O)CH=CH 2
R 5为-Me;
R 6为-Me;
或者R 5和R 6连接形成-CH 2CH 2-;
R 7为-Me或环丙基;
并且,其他基团如上所定义。
在更具体的实施方案中,本发明提供了通式(I-4)、(I-4-A)、(I-4-B)、(I-4-C)、(I-4-D)、(I-4-E)、(I-4-F)、(I-4-G)或(I-4-H)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
R 1为-CF 3
R 9为H、-F、-Cl、-Br、-Me、-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH=CH 2或-NH-环丙基;
并且,其他基团如上所定义。
在更具体的实施方案中,本发明提供了通式(I-5)、(I-5-A)、(I-5-B)、(I-5-C)、(I-5-D)、(I-5-E)、(I-5-F)、(I-5-G)或(I-5-H)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
环A为任选取代的以下基团:C 3-7环烷基或C 6-10芳基,其中所述取代基选自-F、-Cl、-Br、-Me、-OMe、-CF 3、-OCF 3、-CN、-NHMe、-P(O)Me 2、-NHC(O)CH 2CH 3、-C(O)CH=CH 2、-NHS(O) 2CH 2CH 3、-NH-环丙基、-NHC(O)CH=CH 2或-C(O)CH 2CH 3;环A优选为以下基团:
Figure PCTCN2020110442-appb-000079
其中,
R 9为H、-F、-Cl、-Br、-Me、-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH 2CH 3、-NHS(O) 2CH 2CH 3或-NHC(O)CH=CH 2
X 1为-CH 2-或-N(R X1)-;
X 2为-CH(R X2)-或-N(R X2)-;
其中,
R X1为-C(O)CH 2CH 3或-C(O)CH=CH 2
R X2为H、-Me、-OMe、-NHMe、-NHS(O) 2CH 2CH 3、-C(O)CH 2CH 3或-NHC(O)CH 2CH 3
Figure PCTCN2020110442-appb-000080
表示连接点;
R 1、R与它们连接的原子一起形成任选取代的5-6元杂环基,其中所述取代基选自卤素、氧代、-iPr、-Et、卤素单取代或多取代的苯基;R 1和R优选形成以下基团:-C(O)N(R N)C(O)-或-C(CH 3)=C(R N)C(O)-;
其中,
R N选自-iPr、-Et或
Figure PCTCN2020110442-appb-000081
R 11为-Cl、-Br;
R 12为H或-F;
R 13为H或-F;
Figure PCTCN2020110442-appb-000082
表示连接点;
R 3为H或-OMe;
R 4为H或-Me;
并且,其他基团如上所定义。
在更具体的实施方案中,本发明提供了通式(I-5-1)或(I’-5-1)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000083
其中,
R 9为-NHC(O)CH 2CH 3或-NHC(O)CH=CH 2
R N为-iPr或-Et;
并且,其他基团如上所定义。
在更具体的实施方案中,本发明提供了通式(I-5-2)或(I’-5-2)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000084
其中,
R 3为H或-OMe;
R 4为H或-Me;
并且,其他基团如上所定义。
在更具体的实施方案中,本发明提供了通式(I-5-3)或(I’-5-3)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
Figure PCTCN2020110442-appb-000085
其中,
X 1为-CH 2-或-N(R X1)-;
X 2为-CH(R X2)-或-N(R X2)-;
其中,
R X1为-C(O)CH 2CH 3
R X2为H、-Me、-OMe、-NHMe、-C(O)CH 2CH 3、-NHS(O) 2CH 2CH 3或-NHC(O)CH 2CH 3
R 11为-Cl或-Br;
R 12为H或-F;
R 13为H或-F;
并且,其他基团如上所定义。
在更具体的实施方案中,本发明提供了通式(I-6)、(I-6-A)、(I-6-B)、(I-6-C)、(I-6-D)、(I-6-E)、(I-6-F)、(I-6-G)或(I-6-H)的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
L为-O-或-NH-;
R 3为H或-OMe;
R 4为H或-F;
R 9为-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH 2CH 3或-NHC(O)CH=CH 2
并且,其他基团如上所定义。
在更具体的实施方案中,本发明涉及以上所有的通式化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,
其中,
Figure PCTCN2020110442-appb-000086
选自以下基团:
Figure PCTCN2020110442-appb-000087
Figure PCTCN2020110442-appb-000088
并且上述基团中的一个或多个H原子可以被D原子取代。
在更具体的实施方案中,本发明涉及以上所有的通式化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,
其中L 1和L 2独立地选自化学键、-O-、-S-、-S(O)-、-S(O) 2-、-S(O)(=NH)-、-S(O)(=NMe)-、
Figure PCTCN2020110442-appb-000089
Figure PCTCN2020110442-appb-000090
-NH-、-N(Me)-、
Figure PCTCN2020110442-appb-000091
-N(CF 3)-、-CH 2-、-CH(OMe)-、-CH(Cl)-、-CH(F)-、-CF 2-、-CH(CF 3)-、-C(O)-、-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-CH 2O-、-SCH 2-、-CH 2S-、-S(O)CH 2-、-CH 2S(O)-、-S(O) 2CH 2-、-CH 2S(O) 2-、-NHCH 2-、-N(Me)CH 2-、-CH 2NH-、-CH 2N(Me)-、-C(O)CH 2-、-CH 2C(O)-、-C(O)CMe 2-、-CMe 2C(O)-、-OC(O)-、-C(O)O-、-SC(O)-、-C(O)S-、-NHC(O)-、-N(Me)C(O)-、-C(O)NH-、-C(O)N(Me)-、-S(O)=NH-、-NH=S(O)-、-N=S(O)Me-、-S(O)Me=N-、
Figure PCTCN2020110442-appb-000092
并且上述基团中的一个或多个H原子可以被D原子取代。
在更具体的实施方案中,本发明涉及以上所有的通式化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,
其中E选自化学键、-CH 2CH 2CH 2-、-CH 2CH=CH-、-CH=CHCH 2-、-CH 2C≡C-、-C≡CCH 2-、-CH 2CH 2C(O)-、-CH 2C(O)CH 2-、-C(O)CH 2CH 2-、-CH 2CH 2S(O) 2-、-CH 2S(O) 2CH 2-、-S(O) 2CH 2CH 2-、-C(O)CH=CH-、-C(O)C≡C-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-、-SCH 2CH 2-、 -C(O)CH 2O-、-OCH 2C(O)-、-CH 2C(O)O-、-C(O)CH 2S-、-SCH 2C(O)-、-CH 2C(O)S-、-OC(O)CH 2-、-C(O)OCH 2-、-CH 2OC(O)-、-SC(O)CH 2-、-C(O)SCH 2-、-CH 2SC(O)-、-CH 2CH 2NH-、-CH 2NHCH 2-、-NHCH 2CH 2-、-CH 2CH 2NMe-、-CH 2NMeCH 2-、-NMeCH 2CH 2-、-C(O)CH 2NH-、-NHCH 2C(O)-、-CH 2C(O)NH-、-NHC(O)CH 2-、-C(O)NHCH 2-、-CH 2NHC(O)-、
Figure PCTCN2020110442-appb-000093
Figure PCTCN2020110442-appb-000094
或者两个E单元,或者两个E’单元可以形成-CH 2CH 2OCH 2CH 2-、-OCH 2CH 2CH 2CH 2-、-CH 2CH 2CH 2CH 2O-、
Figure PCTCN2020110442-appb-000095
Figure PCTCN2020110442-appb-000096
并且上述基团中的一个或多个H原子可以被D原子取代。
在更具体的实施方案中,本发明涉及以上所有的通式化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中m为0、1、4、5、6、7、8、9或10;优选地,m为0、1、4、5、6、7或8;优选地,m为0、1、4、5或6;优选地,m为0、1、4或5。
在更具体的实施方案中,本发明涉及以上所有的通式化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中-L 1-(E) m-L 2-的链长度为4至14个键长;优选地,链长度为小于12个键长;优选地,链长度为5-10个键长;优选地,链长度为5、6、7、8、9或10个键长。
在更具体的实施方案中,本发明涉及一种化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,所述化合物选自以下:
Figure PCTCN2020110442-appb-000097
Figure PCTCN2020110442-appb-000098
Figure PCTCN2020110442-appb-000099
Figure PCTCN2020110442-appb-000100
Figure PCTCN2020110442-appb-000101
本发明化合物可包括一个或多个不对称中心,且因此可以存在多种立体异构体形式,例如,对映异构体和/或非对映异构体形式。例如,本发明化合物可为单独的对映异构体、非对映异构体或几何异构体(例如顺式和反式异构体),或者可为立体异构体的混合物的形式,包括外消旋体混合物和富含一种或多种立体异构体的混合物。异构体可通过本领域技术人员已知的方法从混合物中分离,所述方法包括:手性高压液相色谱法(HPLC)以及手性盐的形成和结晶;或者优选的异构体可通过不对称合成来制备。
本领域技术人员将理解,有机化合物可以与溶剂形成复合物,其在该溶剂中发生反应或从该溶剂中沉淀或结晶出来。这些复合物称为“溶剂合物”。当溶剂是水时,复合物称为“水合物”。本发明涵盖了本发明化合物的所有溶剂合物。
术语“溶剂合物”是指通常由溶剂分解反应形成的与溶剂相结合的化合物或其盐的形式。这个物理缔合可包括氢键键合。常规溶剂包括包括水、甲醇、乙醇、乙酸、DMSO、THF、乙醚等。本文所述的化合物可制备成,例如,结晶形式,且可被溶剂化。合适的溶剂合物包括药学上可接受的溶剂合物且进一步包括化学计量的溶剂合物和非化学计量的溶剂合物。在一些情况下,所述溶剂合物将能够分离,例如,当一或多个溶剂分子掺入结晶固体的晶格中时。“溶剂合物”包括溶液状态的溶剂合物和可 分离的溶剂合物。代表性的溶剂合物包括水合物、乙醇合物和甲醇合物。
术语“水合物”是指与水相结合的化合物。通常,包含在化合物的水合物中的水分子数与该水合物中该化合物分子数的比率确定。因此,化合物的水合物可用例如通式R·x H 2O代表,其中R是该化合物,和x是大于0的数。给定化合物可形成超过一种水合物类型,包括,例如,单水合物(x为1)、低级水合物(x是大于0且小于1的数,例如,半水合物(R·0.5 H 2O))和多水合物(x为大于1的数,例如,二水合物(R·2 H 2O)和六水合物(R·6 H 2O))。
本发明化合物可以是无定形或结晶形式(多晶型)。此外,本发明化合物可以以一种或多种结晶形式存在。因此,本发明在其范围内包括本发明化合物的所有无定形或结晶形式。术语“多晶型物”是指特定晶体堆积排列的化合物的结晶形式(或其盐、水合物或溶剂合物)。所有的多晶型物具有相同的元素组成。不同的结晶形式通常具有不同的X射线衍射图、红外光谱、熔点、密度、硬度、晶体形状、光电性质、稳定性和溶解度。重结晶溶剂、结晶速率、贮存温度和其他因素可导致一种结晶形式占优。化合物的各种多晶型物可在不同的条件下通过结晶制备。
本发明还包括同位素标记的化合物(同位素变体),它们等同于式(I)所述的那些,但一个或多个原子被原子质量或质量数不同于自然界常见的原子质量或质量数的原子所代替。可以引入本发明化合物中的同位素的实例包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,分别例如 2H、 3H、 13C、 11C、 14C、 15N、 18O、 17O、 31P、 32P、 35S、 18F和 36Cl。含有上述同位素和/或其它原子的其它同位素的本发明化合物、其前体药物和所述化合物或所述前体药物的药学上可接受的盐都属于本发明的范围。某些同位素标记的本发明化合物、例如引入放射性同位素(例如 3H和 14C)的那些可用于药物和/或底物组织分布测定。氚、即 3H和碳-14、即 14C同位素是特别优选的,因为它们容易制备和检测。进而,被更重的同位素取代,例如氘、即 2H,由于代谢稳定性更高可以提供治疗上的益处,例如延长体内半衰期或减少剂量需求,因而在有些情况下可能是优选的。同位素标记的本发明式(I)化合物及其前体药物一般可以这样制备,在进行下述流程和/或实施例与制备例所公开的工艺时,用容易得到的同位素标记的试剂代替非同位素标记的试剂。
此外,前药也包括在本发明的上下文内。本文所用的术语“前药”是指在体内通过例如在血液中水解转变成其具有医学效应的活性形式的化合物。药学上可接受的前药描述于T.Higuchi和V.Stella,Prodrugs as Novel Delivery Systems,A.C.S.Symposium Series的Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,以及D.Fleisher、S.Ramon和H.Barbra“Improved oral drug delivery:solubility limitations overcome by the use of prodrugs”,Advanced Drug Delivery Reviews(1996)19(2)115-130,每篇引入本文作为参考。
前药为任何共价键合的本发明化合物,当将这种前药给予患者时,其在体内释放母体化合物。通常通过修饰官能团来制备前药,修饰是以使得该修饰可以通过常规操作或在体内裂解产生母体化合物的方式进行的。前药包括,例如,其中羟基、氨基或巯基与任意基团键合的本发明化合物,当将其给予患者时,可以裂解形成羟基、氨基或巯基。因此,前药的代表性实例包括(但不限于)式(I)化合物的羟基、巯基和氨基官能团的乙酸酯/酰胺、甲酸酯/酰胺和苯甲酸酯/酰胺衍生物。另外,在羧酸(-COOH)的情况下,可以使用酯,例如甲酯、乙酯等。酯本身可以是有活性的和/或可以在人体体内条件下水解。合适的药学上可接受的体内可水解的酯基包括容易在人体中分解而释放母体酸或其盐的那些基团。
本发明还提供药物制剂,包含治疗有效量的式(I)化合物或其治疗学上可接受的盐和其药学上可接受的载体、稀释剂或赋形剂。所有这些形式都属于本发明。
药物组合物和试剂盒
在另一方面,本发明提供了药物组合物,其包含本发明化合物(还称为“活性组分”)和药学上可接受的赋形剂。在一些实施方案中,所述药物组合物包含有效量的本发明化合物。在一些实施方案中,所述药物组合物包含治疗有效量的本发明化合物。在一些实施方案中,所述药物组合物包含预防有效量的本发明化合物。
用于本发明的药学上可接受的赋形剂是指不会破坏一起调配的化合物的药理学活性的无毒载剂、佐剂或媒剂。可以用于本发明组合物中的药学上可接受的载剂、佐剂或媒剂包括(但不限于)离子交换剂、氧化铝、硬脂酸铝、卵磷脂、血清蛋白(如人类血清白蛋白)、缓冲物质(如磷酸盐)、甘氨酸、山梨酸、山梨酸钾、饱和植物脂肪酸的偏甘油酯混合物、水、盐或电解质(如硫酸鱼精蛋白)、磷酸氢二钠、磷酸氢钾、氯化钠、锌盐、硅胶、三硅酸镁、聚乙烯吡咯烷酮、基于纤维素的物质、聚乙二醇、羧甲基纤维素钠、聚丙烯酸酯、蜡、聚乙烯-聚氧丙烯-嵌段聚合物、聚乙二醇以及羊毛脂。
本发明还包括试剂盒(例如,药物包装)。所提供的试剂盒可以包括本发明化合物、其它治疗剂,以及含有本发明化合物、其它治疗剂的第一和第二容器(例如,小瓶、安瓿瓶、瓶、注射器和/或可分散包装或其它合适的容器)。在一些实施方案中,提供的试剂盒还可以任选包括第三容器,其含有用于稀释或悬浮本发明化合物和/或其它治疗剂的药用赋形剂。在一些实施方案中,提供在第一容器和第二容器中的本发明化合物和其它治疗剂组合形成一个单位剂型。
给药
本发明提供的药物组合物可以通过许多途径给药,包括但不限于:口服给药、肠胃外给药、吸入给药、局部给药、直肠给药、鼻腔给药、口腔给药、阴道给药、通过植入剂给药或其它给药方式。例如,本文使用的肠胃外给药包括皮下给药、皮内给药、静脉内给药、肌肉内给药、关节内给药、动脉内给药、滑膜腔内给药、胸骨内给药、脑脊髓膜内给药、病灶内给药、和颅内的注射或输液技术。
通常,给予有效量的本文所提供的化合物。按照有关情况,包括所治疗的病症、选择的给药途径、实际给予的化合物、个体患者的年龄、体重和响应、患者症状的严重程度,等等,可以由医生确定实际上给予的化合物的量。
当用于预防本发明所述病症时,给予处于形成所述病症危险之中的受试者本文所提供的化合物,典型地基于医生的建议并在医生监督下给药,剂量水平如上所述。处于形成具体病症的危险之中的受试者,通常包括具有所述病症的家族史的受试者,或通过遗传试验或筛选确定尤其对形成所述病症敏感的那些受试者。
还可以长期给予本文所提供的药物组合物(“长期给药”)。长期给药是指在长时间内给予化合物或其药物组合物,例如,3个月、6个月、1年、2年、3年、5年等等,或者可无限期地持续给药,例如,受试者的余生。在一些实施方案中,长期给药意欲在长时间内在血液中提供所述化合物的恒定水平,例如,在治疗窗内。
可以使用各种给药方法,进一步递送本发明的药物组合物。例如,在一些实施方案中,可以推注给药药物组合物,例如,为了使化合物在血液中的浓度提高至有效水平。推注剂量取决于通过身体的活性组分的目标全身性水平,例如,肌内或皮下的推注剂量使活性组分缓慢释放,而直接递送至静脉的推注(例如,通过IV静脉滴注)能够更加快速地递送,使得活性组分在血液中的浓度快速升高至有效水平。在其它实施方案中,可以以持续输液形式给予药物组合物,例如,通过IV静脉滴注,从而在受试者身体中提供稳态浓度的活性组分。此外,在其它实施方案中,可以首先给予推注剂量的药物组合物,而后持续输液。
口服组合物可以采用散装液体溶液或混悬剂或散装粉剂形式。然而,更通常,为了便于精确地剂量给药,以单位剂量形式提供所述组合物。术语“单位剂型”是指适合作为人类患者及其它哺乳动物的 单元剂量的物理离散单位,每个单位包含预定数量的、适于产生所需要的治疗效果的活性物质与合适药学赋形剂。典型的单位剂量形式包括液体组合物的预装填的、预先测量的安瓿或注射器,或者在固体组合物情况下的丸剂、片剂、胶囊剂等。在这种组合物中,所述化合物通常为较少的组分(约0.1至约50重量%,或优选约1至约40重量%),剩余部分为对于形成所需给药形式有用的各种载体或赋形剂以及加工助剂。
对于口服剂量,代表性的方案是,每天一个至五个口服剂量,尤其是两个至四个口服剂量,典型地是三个口服剂量。使用这些剂量给药模式,每个剂量提供大约0.01至大约20mg/kg的本发明化合物,优选的剂量各自提供大约0.1至大约10mg/kg,尤其是大约1至大约5mg/kg。
为了提供与使用注射剂量类似的血液水平,或比使用注射剂量更低的血液水平,通常选择透皮剂量,数量为大约0.01至大约20%重量,优选大约0.1至大约20%重量,优选大约0.1至大约10%重量,且更优选大约0.5至大约15%重量。
从大约1至大约120小时,尤其是24至96小时,注射剂量水平在大约0.1mg/kg/小时至至少10mg/kg/小时的范围。为了获得足够的稳定状态水平,还可以给予大约0.1mg/kg至大约10mg/kg或更多的预载推注。对于40至80kg的人类患者来说,最大总剂量不能超过大约2g/天。
适于口服给药的液体形式可包括合适的水性或非水载体以及缓冲剂、悬浮剂和分散剂、着色剂、调味剂,等等。固体形式可包括,例如,任何下列组份,或具有类似性质的化合物:粘合剂,例如,微晶纤维素、黄蓍胶或明胶;赋形剂,例如,淀粉或乳糖,崩解剂,例如,褐藻酸、Primogel或玉米淀粉;润滑剂,例如,硬脂酸镁;助流剂,例如,胶体二氧化硅;甜味剂,例如,蔗糖或糖精;或调味剂,例如,薄荷、水杨酸甲酯或橙味调味剂。
可注射的组合物典型地基于可注射用的无菌盐水或磷酸盐缓冲盐水,或本领域中已知的其它可注射的赋形剂。如前所述,在这种组合物中,活性化合物典型地为较少的组分,经常为约0.05至10%重量,剩余部分为可注射的赋形剂等。
典型地将透皮组合物配制为含有活性组分的局部软膏剂或乳膏剂。当配制为软膏剂时,活性组分典型地与石蜡或可与水混溶的软膏基质组合。或者,活性组分可与例如水包油型乳膏基质一起配制为乳膏剂。这种透皮制剂是本领域中公知的,且通常包括用于提升活性组分或制剂的稳定的皮肤渗透的其它组份。所有这种已知的透皮制剂和组份包括在本发明提供的范围内。
本发明化合物还可通过经皮装置给予。因此,经皮给药可使用贮存器(reservoir)或多孔膜类型、或者多种固体基质的贴剂实现。
用于口服给予、注射或局部给予的组合物的上述组份仅仅是代表性的。其它材料以及加工技术等阐述于Remington's Pharmaceutical Sciences,17th edition,1985,Mack Publishing Company,Easton,Pennsylvania的第8部分中,本文以引用的方式引入该文献。
本发明化合物还可以以持续释放形式给予,或从持续释放给药系统中给予。代表性的持续释放材料的描述可在Remington's Pharmaceutical Sciences中找到。
本发明还涉及本发明化合物的药学上可接受的制剂。在一个实施方案中,所述制剂包含水。在另一个实施方案中,所述制剂包含环糊精衍生物。最常见的环糊精为分别由6、7和8个α-1,4-连接的葡萄糖单元组成的α-、β-和γ-环糊精,其在连接的糖部分上任选包括一个或多个取代基,其包括但不限于:甲基化的、羟基烷基化的、酰化的和磺烷基醚取代。在一些实施方案中,所述环糊精为磺烷基醚β-环糊精,例如,磺丁基醚β-环糊精,也称作Captisol。参见,例如,U.S.5,376,645。在一些实施方案中,所述制剂包括六丙基-β-环糊精(例如,在水中,10-50%)。
治疗
如本文中所述,已知EGFR激酶在肿瘤发生及许多其它疾病中起作用。我们已经发现本发明化合物具有强有力的抗肿瘤活性,所述抗肿瘤活性被认为通过抑制EGFR激酶获得。
从而,本发明化合物具有作为抗肿瘤药剂的价值。尤其,本发明化合物具有在实体和/或液体肿瘤疾病的遏制和/或治疗中作为抗增殖、凋亡和/或抗侵袭药剂的价值。尤其,预期本发明化合物有用于预防或治疗对抑制EGFR敏感的那些肿瘤。此外,预期本发明化合物有用于预防或治疗单独或部分由EGFR介导的那些肿瘤。因此,所述化合物可用于在需要此类治疗温血动物中产生EGFR酶抑制作用。
如本文中所述,EGFR激酶的抑制剂包括癌症,例如卵巢癌、宫颈癌、结肠直肠癌、乳腺癌、胰腺癌、胶质瘤、胶质母细胞瘤、黑色素瘤、前列腺癌、白血病、淋巴瘤、非霍奇金淋巴瘤、胃癌、肺癌、肝细胞癌、胃癌、胃肠道基质瘤(GIST)、甲状腺癌、胆管癌、子宫内膜癌、肾癌、间变性大细胞淋巴瘤、急性髓细胞白血病(AML)、多发性骨髓瘤、黑色素瘤、间皮瘤。
有用于治疗患者中的癌症的抗癌作用包括但不局限于抗肿瘤作用、响应率、疾病进展的时间及存活率。本发明治疗方法的抗肿瘤作用包括但不局限于肿瘤生长的抑制、肿瘤生长的延迟、肿瘤的退化、肿瘤的收缩、治疗停止后肿瘤再生长时间的延长及疾病进展的减慢。抗癌作用包括预防性治疗以及现存在疾病的治疗。
EGFR激酶抑制剂或其药学上可接受的盐还有用于治疗癌症患者,所述癌症包括但不局限于血癌,如白血病、多发性骨髓瘤;淋巴瘤,如霍奇金病、非霍奇金淋巴瘤(包括套细胞淋巴瘤)和骨髓增生异常综合征,以及还有实体肿瘤及其转移灶(metastases),如乳腺癌、肺癌(非小细胞肺癌(NSCL)、小细胞肺癌(SCLC)、鳞状细胞癌)、子宫内膜癌、中枢神经系统肿瘤(如胶质瘤、胚胎期发育不良性神经上皮肿瘤、多形性成胶质细胞瘤、混合型胶质瘤、髓母细胞瘤、成视网膜细胞瘤、成神经细胞瘤、生殖细胞瘤和畸胎瘤、胃肠道癌(如胃癌)、食道癌、肝细胞(肝)癌、胆管癌、结肠和直肠癌、小肠癌、胰腺癌、皮肤癌如黑素瘤(尤其转移性黑素瘤)、甲状腺癌、头及颈癌和唾液腺癌、前列腺癌、睾丸癌、卵巢癌、子宫颈癌、子宫癌、外阴癌、膀胱癌、肾癌(包括肾细胞癌、明细胞和肾嗜酸细胞瘤)、鳞状细胞癌、肉瘤如骨肉瘤、软骨肉瘤、平滑肌肉瘤、软组织肉瘤、尤因肉瘤、胃肠道间质瘤(GIST)、卡波西肉瘤和儿科癌如横纹肌肉瘤和成神经细胞瘤。
本发明化合物的有效量通常在平均日剂量为0.01mg至50mg化合物/千克患者体重,优选0.1mg至25mg化合物/千克患者体重,以单次或多次给药。通常,本发明化合物可向该有此治疗需要的患者以每位患者约1mg至约3500mg的日剂量范围给药,优选10mg至1000mg。例如,每位患者的日剂量可为10、20、30、40、50、60、70、80、90、100、150、200、250、300、350、400、500、600、700、800、900或1000mg。可每天、每周(或间隔数天)或以间歇时间表,给药一次或多次。例如,可在每周的基础上(例如每周一),每天给予所述化合物一次或多次,不定地或持续几周,例如4-10周。或者,可每天给药持续几天(例如2-10天),然后几天(例如1-30天)不给药所述化合物,不定地重复该循环或重复给定的次数,例如4-10个循环。例如,本发明化合物可每天给药持续5天,然后间断9天,然后再每天给药持续5天,然后间断9天,以此类推,不定地重复该循环或共重复4-10次。
组合治疗
本文中定义的治疗可作为单独治疗应用,或除本发明化合物之外,可包括常规外科手术或放疗或化疗。因此,本发明化合物还可与用于治疗癌症的现有治疗药剂联合使用。
除了使用本发明化合物治疗以外,还涉及到常规的手术或放射疗法或化学疗法或免疫疗法。这种化学疗法与本发明化合物可以同时地、连续地、或分别地给药,并且可包含以下类型的抗肿瘤剂的一种或多种:
(i)医学肿瘤学中所使用的抗增殖/抗肿瘤药物及其组合,例如烷化剂(例如顺铂、奥沙利铂、卡铂、环磷酰胺、氮芥、美法仑、苯丁酸氮芥、白消安、替莫唑胺、亚硝基脲类);抗代谢药(例如吉西他滨和抗叶酸剂,例如氟嘧啶(如5-氟尿嘧啶和替加氟)、雷替曲塞、甲氨喋呤、阿糖胞苷、羟基脲);抗肿瘤抗生素(例如蒽环类,如阿霉素、博来霉素、多柔比星、道诺霉素、表柔比星、伊达比星、丝裂霉素C、放线菌素、光神霉素);抗有丝分裂药剂(例如长春花生物碱,如长春新碱、长春碱、长春地辛、长春瑞滨;以及紫杉烷类,如紫杉醇、泰索帝、polo激酶抑制剂);拓扑异构酶抑制剂(例如表鬼臼毒素(如依托泊苷、替尼泊苷),安吖啶、托泊替康、喜树碱);
(ii)细胞生长抑制剂,例如抗雌激素药(例如他莫昔芬、氟维司群、托瑞米芬、雷洛昔芬、屈洛昔芬、碘昔芬(iodoxyfene))、抗雄激素药(例如比卡鲁胺、氟他胺、尼鲁米特、醋酸环丙孕酮)、LHRH拮抗剂或LHRH激动剂(例如戈舍瑞林、亮丙瑞林、和布舍瑞林)、孕激素类(例如醋酸甲地孕酮)、芳香酶抑制剂(例如阿那曲唑、来曲唑、伏罗唑(vorazole)、伊西美坦)、5α-还原酶抑制剂(例如非那雄胺);
(iii)抗侵袭剂(anti-invasion),例如c-Src激酶家族抑制剂,[如4-(6-氯-2,3-亚甲基二氧基苯胺基)-7-[2-(4-甲基哌嗪-1-基)乙氧基]-5-四氢吡喃-4-基氧基喹唑啉[AZD0530(塞卡替尼)]、N-(2-氯-6-甲基苯基)-2-{6-[4-(2-羟乙基)哌嗪-1-基]-2-甲基嘧啶-4-基氨基}噻唑-5-甲酰胺(达沙替尼,BMS-354825)和波舒替尼(SKI-606),以及金属蛋白酶抑制剂(如马立马司他)、尿激酶纤溶酶原激活物受体功能的抑制剂或者类肝素酶(heparanase)的抗体];
(iv)生长因子功能的抑制剂:例如这种抑制剂包括生长因子抗体和生长因子受体抗体(例如抗erbB2抗体曲妥珠单抗[赫赛汀]、抗EGFR抗体帕尼单抗、抗erbB1抗体西妥昔单抗[Erbitux,C225];这种抑制剂还包括:酪氨酸激酶抑制剂,例如表皮生长因子家族的抑制剂(例如EGFR家族酪氨酸激酶抑制剂,如N-(3-氯-4-氟苯基)-7-甲氧基-6-(3-吗啉基丙氧基)-喹唑啉-4-胺(吉非替尼,ZD1839)、N-(3-乙炔基苯基)-6,7-双(2-甲氧基乙氧基)喹唑啉-4-胺(厄洛替尼,OSI-774)、6-丙烯酰胺基-N-(3-氯-4-氟苯基)-7-(3-吗啉基丙氧基)-喹唑啉-4-胺(CI1033)、erbB2酪氨酸激酶抑制剂(例如拉帕替尼);肝细胞生长因子家族的抑制剂;胰岛素生长因子家族的抑制剂;血小板衍生的生长因子家族的抑制剂,例如伊马替尼和/或尼洛替尼(AMN107);丝氨酸/苏氨酸激酶的抑制剂(例如Ras/Raf信号传导抑制剂,例如法呢基转移酶抑制剂,例如索拉非尼(BAY43-9006)、替匹法尼(R115777)、氯那法尼(SCH66336))、通过mEK和/或AKT激酶的细胞信号传导抑制剂、c-kit抑制剂、abl激酶抑制剂、PI3激酶抑制剂、Plt3激酶抑制剂、CSF-1R激酶抑制剂、IGF受体(胰岛素样生长因子)激酶抑制剂;极光激酶(aurora kinase)抑制剂(例如AZD1152、PH739358、VX-680、MLN8054、R763、MP235、MP529、VX-528、AX39459),细胞周期蛋白依赖性激酶抑制剂,例如CDK2和/或CDK4抑制剂;
(v)抗血管生成剂,例如抑制血管内皮生长因子作用的药剂,[例如抗人血管内皮细胞生长因子抗体贝伐珠单抗(阿瓦斯丁)以及例如VEGF受体酪氨酸激酶抑制剂,例如凡德他尼(ZD6474)、伐他拉尼(PTK787)、舒尼替尼(SU11248)、阿西替尼(AG-013736)、帕唑帕尼(GW786034)、4-(4-氟-2-甲基吲哚-5-基氧基)-6-甲氧基-7-(3-吡咯烷-1-基丙氧基)喹唑啉(AZD2171),以及通过其它机制起作用的化合物(例如雷诺胺,整联蛋白αvβ3功能的抑制剂和血管抑素(angiostatin))];
(vi)血管损伤剂,例如康普瑞汀A4;
(vii)内皮素受体拮抗剂,例如齐泊腾坦(ZD4054)或者阿曲生坦;
(viii)反义治疗剂,例如导向上面列出的靶点的那些,例如ISIS2503(一种anti-ras反义治疗剂);
(ix)基因治疗方法,包括例如替换异常基因(例如异常p53或者异常BRCA1或BRCA2)的方法;GDEPT(基因定向的酶前药治疗)法,例如使用胞嘧啶脱氨酶、胸苷激酶或者细菌硝基还原酶的那些;提高患者对化学治疗或放射治疗的耐受性的方法,例如多重耐药基因治疗;和
(x)免疫治疗方法,包括例如提高患者肿瘤细胞的免疫原性的体外和体内方法,例如用细胞因子(例如白细胞介素2、白细胞介素4或者粒细胞巨噬细胞集落刺激因子)进行转染;降低T细胞无效能的方法;使用转染的免疫细胞(例如细胞因子转染的树突状细胞)的方法;使用细胞因子转染的肿瘤细胞系的方法;使用抗独特型抗体的方法;降低免疫抑制性细胞(例如调节性T细胞、髓源性(myeloid-derived)抑制细胞、或表达IDO(吲哚胺2,3-脱氧酶)的树突状细胞)的功能的方法;以及使用衍生自肿瘤相关抗原(例如NY-ESO-1,mAGE-3、WT1或Her2/neu)的蛋白质类或肽类组成的癌症疫苗的方法。
实施例
本文所用的材料或试剂为可购买到的或由本领域通常已知的合成方法制备。
中间体的制备
2-(2,6-二氧代哌啶-3-基)-4-羟基异吲哚啉-1,3-二酮(A2-1)的制备
Figure PCTCN2020110442-appb-000102
将4-羟基异苯并呋喃-1,3-二酮(5.0g,30.5mmol),3-氨基哌啶-2,6-二酮(6.85g,41.8mmol)和醋酸钠(4.1g,50.0mmol)分散于100mL CH 3COOH中,氮气保护下,140℃搅拌回流8h。反应液冷却至室温后,减压浓缩除去CH 3COOH后,残渣中加入200mL水打浆搅拌2h。抽滤,固体产物用水淋洗两遍,烘干得灰白色固体产物7.4g,收率89.15%,LCMS:[M+H] +=275。 1H-NMR:(400MHz,DMSO-d 6)δ11.164(s,1H),11.080(s,1H),7.675(dd,J=7.2Hz,8.4Hz,1H),7.329(dd,J=7.2Hz,26.4Hz,2H),5.094(dd,J=5.2Hz,12.8Hz,1H),2.919-2.840(m,1H),2.619-2.530(m,2H),2.048-1.989(m,1H)。
2-(2,6-二氧代哌啶-3-基)-4-氟异吲哚啉-1,3-二酮(A6-1)的制备
Figure PCTCN2020110442-appb-000103
其制备方法同2-(2,6-二氧代哌啶-3-基)-4-羟基异吲哚啉-1,3-二酮的制备,以4-氟异苯并呋喃-1,3-二酮为原料,得到白色固体,LCMS:[M+H] +=277。
4-溴-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(A1-1)的制备
Figure PCTCN2020110442-appb-000104
其制备方法同2-(2,6-二氧代哌啶-3-基)-4-羟基异吲哚啉-1,3-二酮的制备,以4-溴异苯并呋喃-1,3-二酮为原料,得到白色固体,LCMS:[M+H] +=337,339。
2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙酸(A6-5)的制备
Figure PCTCN2020110442-appb-000105
向一个100毫升装有冷凝器的三口瓶中先后加入(2-((5-氯-2-((2-甲氧基-4-(4-(哌嗪-1-基)哌啶-1-基)苯基)氨基)嘧啶-4-基)氨基)苯基)二甲基氧化磷(300mg,0.527mmol),碳酸钾(145mg,1.054mmol)和溴乙酸叔丁酯(113.04mg,0.527mmol),溶于DMF(5mL),加热至60℃搅拌反应3小时。待反应液冷却至室温,加20毫升水稀释,乙酸乙酯萃取3次(每次20毫升),合并有机相,饱和食盐水洗涤三次,用无水硫酸钠干燥。减压浓缩得到粗品,用硅胶柱层析分离,得到120mg黄色固体。将上述得到的固体溶于1,4-二氧六环(5mL),加入2mL氯化氢/1,4-二氧六环溶液(4mol/L)室温搅拌2小时。LCMS检测反应结果,反应液基本为目标产物,减压浓缩得到黄色固体112mg,收率34%,LCMS:[M+H] +=628。
3-(4-溴-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C1-1)的制备
Figure PCTCN2020110442-appb-000106
将3-溴-2-甲基苯甲酸甲酯(1.14g,5.0mmol)溶于20.0mL CCl 4中,氮气保护下,加入NBS(1.34g,7.5mmol)和AIBN(164mg,1.0mmol),升高温度至85℃,回流反应20h。TLC检测无原料剩余。反应液冷却至室温后,抽滤,减压浓缩滤液得粗产物,Flash纯化得浅黄色油状产物1.35g。将此油状化合物(1.35g,4.41mmol)和3-氨基哌啶-2,6-二酮盐酸盐(941mg,5.74mmol)分散于25.0mL无水MeCN中,加入TEA(580mg,5.74mmol),升高温度至80℃,回流反应16h。LCMS检测反应完全。反应液冷却至室温后,抽滤,滤饼用MeCN淋洗三次,烘干固体得化合物C1-1(1.31g,收率92.3%)。LCMS:[M+H] +=323,325。
2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-N-(哌啶-4-基)乙酰胺(C88-4)的制备
Figure PCTCN2020110442-appb-000107
将化合物A6-5(75mg,0.12mmol),HATU(45mg,0.118mmol),DIEA(77mg,0.60mmol)溶于2.5mL的DMF中,氮气保护下室温搅拌1小时。然后加入4-氨基哌啶-1-羧酸叔丁酯(26mg,0.13mmol),继续搅拌2.5小时。减压除去溶剂,然后溶于二氯甲烷(4.0mL),滴加入TFA(1.0mL),室温搅拌2小时。减压除去溶剂,薄层色谱分离,得到化合物C88-4,LCMS:[M+H] +=710。
3-(5-溴-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C192-1)的制备
Figure PCTCN2020110442-appb-000108
将4-溴-2-(溴甲基)苯甲酸甲酯(300mg,0.974mmol),3-氨基哌啶-2,6-二酮盐酸盐(208mg,1.266mmol),三乙胺(128mg,1.266mmol)溶于5mL乙腈中,氮气保护下80℃搅拌过夜。冷至室温,减压除去溶剂,粗品用Flash纯化得到白色固体化合物C192-1(180mg,产率57.0%)。LCMS:[M+H] +=323,325。
4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-7-氧代庚基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(A1)的制备
Figure PCTCN2020110442-appb-000109
第1步:
将A1-1(600mg,1.79mmol),6-炔庚酸(450mg,3.58mmol),CuI(69mg,0.36mmol),Pd(PPh 3) 2Cl 2(501mg,0.71mmol)和三乙胺(903mg,8.94mmol)加入到DMF(15ml)中,氮气保护下于70℃搅拌5小时。TLC检测原料消耗,LCMS检测有目标产物生成。减压浓缩,粗品经柱色谱分离,得到固体A1-2(680mg,收率99.7%),LCMS:[M+H] +=383。
第2步:
将A1-2(340mg,0.89mmol)和Pd/C(10%,74mg)加入到甲醇(8ml)中,3MPa下室温加氢反应7小时。滤除固体,减压除去溶剂,得到固体A1-3(300mg,收率87.5%),LCMS:[M+H] +=387。
第3步:
将A1-3(85mg,0.22mmol),DIEA(85mg,0.66mmol)和HATU(84mg,0.22mmol)溶于DMF中,氮气保护下室温搅拌半小时。然后把A1-4(175mg,0.31mmol)加入到反应液中,继续搅拌3小时。TLC检测原料消耗,LCMS检测有目标产物生成。反应液直接经Flash色谱纯化得到白色固体A1(50mg,收率24.3%),LCMS:[M+H] +=938。
4-((7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-7-氧代庚基)氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(A2)的制备
Figure PCTCN2020110442-appb-000110
第1步:
将A2-1(200mg,0.73mmol),7-溴庚酸叔丁酯(232mg,0.87mmol),KI(12mg,0.072mmol)和碳酸氢钾(110mg)分散于DMF(5ml)中,氮气保护下60℃搅拌过夜。过滤,减压除去溶剂,Flash色谱纯化,得到160mg白色固体A2-2。
第2步:
将A2-2(160mg)溶于2ml TFA中,室温搅拌1小时。减压除去溶剂,得到粗品A2-3,直接用于下一步。
第3步:
将A2-3(140mg,0.348mmol),DIEA(225mg,1.744mmol),HATU(133mg,0.35mmol)溶于DMF(7ml)中,氮气保护下室温搅拌半小时。然后把A1-4(237mg,0.417mmol)加入到反应液中,继续搅拌2小时。反应液直接用Flash色谱纯化,得到A2(25mg,7.5%),LCMS:[M+H] +=954。
N-(2-(2-(2-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙氧基)乙氧基)乙氧基)乙基)-2-((2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-4-基)氧基)乙酰胺(A3)的制备
Figure PCTCN2020110442-appb-000111
第1步:
将A2-1(300mg,1.15mmol),溴乙酸叔丁酯(467mg,1.79mmol),KI(18mg,0.109mmol),碳酸氢钾(164mg,1.64mmol)分散于DMF中,氮气保护下于60℃搅拌过夜。过滤,减压除去溶剂,Flash制备色谱纯化,得到116mg产物A3-1,收率26.9%。
第2步:
将A3-1(116mg)溶于TFA(1ml)中,室温搅拌1小时。蒸除溶剂,得到粗品A3-2,直接用于下一步。
第3步:
氮气保护下将A1-4(150mg,0.21mmol),A3-3(94mg,0.26mmol),K 2CO 3(73mg,0.53mg)加入到乙腈中,90℃下搅拌过夜。滤除固体,减压蒸除溶剂,Flash色谱纯化,得到化合物A3-4(110mg,收率49.5%)。
第4步:
将A3-4(110mg,0.13mmol),TFA(1ml)溶于二氯甲烷中,室温搅拌1小时。减压蒸除溶剂,得粗品A3-5,直接用于下一步。
第5步:
将A3-2(99mg,0.31mmol),DIEA(192mg,1.49mmol),HATU(118mg,0.31mmol)溶于DMF中,氮气保护下室温搅拌半小时。然后把A3-5(222mg,0.30mmol)加入到反应液中,继续搅拌3小时。反应液直接Flash色谱纯化,得到白色固体A3(25mg,收率:18.1%),LCMS:[M+H] +=1059。
2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-N-(2-((2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-4-基)氨基)乙基)乙酰胺(A6)的制备
Figure PCTCN2020110442-appb-000112
第1步:
向一个装有冷凝器的三口烧瓶中先后加入化合物A6-1(276.2mg,1.0mmol),化合物A6-2(160.2mg,1.0mmol),DIPEA(387mg,3.0mmol)以及N,N-二甲基乙酰胺(5mL),在氮气保护下加热至90℃搅拌反应过夜。LCMS检测原料基本消耗,有目标产物生成。反应液冷却后用50毫升水稀释,用乙酸乙酯萃取三次(每次25毫升),合并有机相,用饱和食盐水洗涤三次(每次20毫升),有机相用无水硫酸钠干燥2小时,过滤、减压浓缩滤液得到粗品,粗品用硅胶柱色谱分离,得到黄色固体A6-3(200mg,收率48%)。
第2步:
向一个50毫升的单口烧瓶中加入化合物A6-3(100mg,0.24mmol)和1,4-二氧六环(5mL),搅拌溶解,加入氯化氢/1,4-二氧六环溶液(2mL,4M),室温搅拌反应2小时。TLC检测原料消耗,LCMS检测有目标产物生成。减压浓缩得到化合物A6-4的盐酸盐,为黄色固体(80mg,收率95%),LCMS:[M+H] +=317。
第3步:
向一个50毫升三口烧瓶中加入化合物A6-5(31.4mg,0.05mmol),HATU(23mg,0.06mmol)和二氯甲烷(3mL),在氮气保护下室温搅拌溶解,然后加入DIPEA(64mg,0.5mmol),室温搅拌反应30分钟。随后加入化合物A6-4(22mg,0.05mmol)的DMF(2.0mL)溶液,室温搅拌过夜。LCMS检测反应有目标产物生成。反应液减压浓缩除去二氯甲烷,用Flash制备柱色谱分离,得到浅黄色粉末即化合 物A6(14mg,收率30.4%),LCMS:[M+H] +=926。
2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-N-(2-(2-(2-(2-((2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-4-基)氨基)乙氧基)乙氧基)乙氧基)乙基)乙酰胺(A8)的制备
Figure PCTCN2020110442-appb-000113
第1步:
向20毫升微波反应管中先后加入化合物A3-3(260.7mg,0.732mmol),商业购买的化合物A8-1(200mg,0.732mmol),碳酸氢钠(232mg,2.196mmol)和N,N-二甲基甲酰胺(5mL),在氮气保护下用微波反应器加热至90℃反应2小时(反应器:CEM,功率100W)。通过薄层色谱检测化合物A8-1基本消失,液质检测有目标产物生成。反应液冷却过滤去除无机盐,加20毫升水稀释,用乙酸乙酯萃取三次(每次用乙酸乙酯25毫升),合并有机相,用饱和食盐水洗涤两次(每次10mL),然后用无水硫酸钠干燥,过滤、减压浓缩滤液得到粗品,粗品用硅胶柱层析分离,得到化合物A8-2(100mg,收率:24.9%)。
第2步:
向一个50毫升单口烧瓶中加入化合物A8-2(100mg,0.182mmol)和1,4-二氧六环(4mL),搅拌溶解,再加入三氟乙酸(2mL),室温搅拌反应2小时。TLC检测反应完毕。减压浓缩得到褐色油状物,用20毫升二氯甲烷溶解,用饱和碳酸氢钠溶液洗涤两次(每次5毫升),用10毫升水洗一次,无水硫酸镁干燥,过滤、减压浓缩滤液得到A8-3黄绿色固体55mg,收率68%,LCMS:[M+H] +=449。
第3步:
向一个50毫升三口烧瓶中加入化合物A6-5(70mg,0.1mmol),HATU(45.6mg,0.12mmol)和二氯甲烷(3mL),在氮气保护下室温搅拌溶解,然后加入DIPEA(64.5mg,0.5mmol),室温搅拌反应30分钟。随后加入化合物A8-3(45mg,0.1mmol),室温搅拌过夜。LCMS检测反应有目标产物生成。反应液用20毫升二氯甲烷稀释,用水洗涤两次(每次5毫升),减压浓缩得到粗品。粗品用硅胶制备板分离,得到浅黄色粉末A8(22mg,收率20.8%),LCMS:[M+H] +=1058。
4-((7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚基)氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(A10)的制备
Figure PCTCN2020110442-appb-000114
第1步:
将A2-1(274mg,1.0mmol),NaHCO 3(252mg,3.0mmol)和KI(83mg,0.5mmol)置于20mL反应瓶内,氮气保护下加入5.5mL无水DMF,搅拌中滴加7-溴-1-庚醇(292.5mg,1.5mmol),滴加完毕后,70℃反应20h。反应液用100mL乙酸乙酯稀释后,依次用饱和NH 4Cl溶液(30mL x 1),H 2O(30mL x 3)和饱和食盐水(30mL x 2)洗涤,有机层用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得粗产物,Prep-TLC纯化得A10-1白色固体产物300mg,产率77.3%,LCMS:[M+H] +=389。 1H-NMR:(400MHz,DMSO-d 6)δ11.092(s,1H),7.827(dd,J=7.6Hz,8.8Hz,1H),7.523(dd,J=8.4Hz,29.6Hz,2H),5.098-5.052(m,1H),4.358(t,J=5.2Hz,1H),4.218(t,J=6.4Hz,2H),2.926-2.836(m,1H),2.045-1.995(m,1H),1.791-1.722(m,2H),1.492-1.237(m,12H)。
第2步:
将A10-1(80mg,0.206mmol)溶于15mL无水DCM中,氮气保护下加入Dess-Martin试剂(262mg,0.618mmol),加毕后,55℃反应2h。反应液用30mL DCM稀释后,加入15mL饱和NaHCO 3溶液和15mL饱和Na 2S 2O 3溶液,搅拌5min后,分出有机层并用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得A10-2粗产物(75mg),LCMS:[M+H] +=387,该粗产物直接用于下一步反应。
第3步:
将A10-2(75mg,0.194mmol)和A1-4(110.3mg,0.194mmol)溶于12.0mL无水DCM中,氮气保护下,滴加2滴CH 3COOH后,室温搅拌5min,然后加入NaBH 4(73.7mg,1.94mmol),室温搅拌反应2h。反应液用30mL DCM稀释后,加入15mL饱和NH 4Cl溶液并搅拌5min,分出有机层先用15mL饱和食盐水洗一次,再用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得粗产物。粗产物用RP-Flash色谱纯化得纯度约60%白色固体产物65mg。该产物继续用Prep-TLC纯化,得纯度90%的白色固体产物30mg。再次用RP-Flash色谱纯化得白色固体纯产物15mg,产率8.2%,LCMS:[M+H] +=940.5。 1H-NMR:(400MHz,DMSO-d 6)δ11.164(s,1H),11.106(s,1H),8.480(s,1H),8.063-8.046(m,2H),7.827(t,J=8.0Hz,1H),7.555-7.502(m,2H),7.450-7.432(m,1H),7.391-7.336(m,2H),7.111(t,J=7.2Hz,1H),6.625(d,J=2.4Hz,1H),6.474(d,J=8.0Hz,1H),5.085(dd,J=5.6,12.8Hz,1H),4.204(t,J=6.4Hz,2H),3.757(s,3H),3.724-3.695(m,2H),2.884-2.851(m,1H),2.688-2.661(m,2H),2.630-2.566(m,3H),2.333-2.246(m,6H),2.227-1.990(m,2H),1.858-1.830(m,2H),1.779-1.745(m,9H),1.524-1.239(m,12H)。
4-((9-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)壬基)氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(A11)的制备
Figure PCTCN2020110442-appb-000115
第1步:
将A2-1(137mg,0.5mmol),NaHCO 3(126mg,1.5mmol)和KI(41.5mg,0.25mmol)置于20mL反应瓶内,氮气保护下加入5.0mL无水DMF,搅拌中滴加9-溴-1-壬醇(167.3mg,0.75mmol),滴加完毕后,70℃反应20h。反应液用80mL乙酸乙酯稀释后,依次用饱和NH 4Cl溶液(20mL x 1),H 2O(20mL x 3)和饱和食盐水(20mL x 2)洗涤,有机层用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得粗产物,Prep-TLC纯化得A11-1白色固体产物70mg,产率33.6%,LCMS:[M+H] +=417。
第2步:
将A11-1(60mg,0.144mmol)溶于12mL无水DCM中,氮气保护下加入Dess-Martin试剂(305.8mg,0.721mmol),加毕后,55℃反应3h。反应液用30mL DCM稀释后,加入15mL饱和NaHCO 3溶液和15mL饱和Na 2S 2O 3溶液,搅拌5min后,分出有机层并用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩,得A11-2粗产物(55mg),LCMS:[M+H] +=415,该粗产物直接用于下一步反应。
第3步:
将A11-2(50mg,0.121mmol)和A1-4(68.7mg,0.121mmol)溶于10.0mL无水DCM中,氮气保护下,滴加2滴CH 3COOH后,室温搅拌5min,然后加入NaBH 4(46mg,1.21mmol),室温搅拌反应2h。反应液用20mL DCM稀释后,加入15mL饱和NH 4Cl溶液并搅拌5min,分出有机层,先用15mL饱和食盐水洗一次,再用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得粗产物,粗产物用RP-Flash色谱纯化得白色固体产物20mg,产率17%,LCMS:[M+H] +=968.5。 1H-NMR:(400MHz,DMSO-d 6)δ11.169(s,1H),11.114(s,1H),8.475(s,1H),8.047(d,J=6.4Hz,2H),7.825-7.860(m,1H),7.555-7.502(m,3H),7.448-7.317(m,2H),7.092(t,J=7.6Hz,1H),6.624-6.450(m,2H),5.085(dd,J=5.2,12.8Hz,1H),4.201(t,J=6.4Hz,2H),3.725(t,J=12.8Hz,6H),3.287(s,2H),2.688-2.540(m,6H),2.328-2.195(m,6H),2.029-1.991(m,2H),1.861-1.830(m,2H),1.779-1.746(m,10H),1.661-1.274(m,4H),1.275-1.237(m,8H)。
3-(4-((7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚基)氨基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(A12)的制备
Figure PCTCN2020110442-appb-000116
第1步:
将7-溴-1-庚醇(200mg,1.03mmol)加入到氯铬酸吡啶鎓盐(333mg,1.54mmol)的THF(5ml)中,室温搅拌反应过夜。减压除去溶剂,加入20ml乙醚,使产品溶解,过滤,浓缩,得到粗品A12-2(160mg,收率80.8%),LCMS:[M+H] +=193,195。
第2步:
将A12-2(200mg,1.04mmol)和3-(4-氨基-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(A12-3)(270mg,1.04mmol)溶于DCM(10ml)和MeOH(5ml)中,氮气保护下50℃搅拌1小时。冷至室温,加入氰基硼氢化钠(98mg,1.56mmol)和2滴冰醋酸,升至50℃继续搅拌1.5小时。冷至室温,用饱和氯化铵水溶液淬灭,用二氯甲烷萃取,分层,合并有机相,无水硫酸钠干燥,静置,过滤,浓缩,粗品Flash纯化得到A12-4(100mg,收率22.1%),LCMS:[M+H] +=436,438。
第3步:
将A12-4(10mg,0.023mmol),A1-4(13mg,0.023mmol)和DIEA(16mg,0.124mmol)溶于DMF(0.5ml)中,氮气保护下于80℃搅拌7小时。减压除去溶剂,Flash纯化得到A12(2.97mg,收率14.0%),LCMS:[M+H] +=925。 1H NMR(400MHz,DMSO-d 6)δ11.17(s,1H),11.00(s,1H),8.48(s,1H),8.06(d,J=5.3Hz,1H),7.53(dd,J=13.9,7.4Hz,1H),7.43-7.24(m,4H),7.18(s,1H),7.09(t,J=7.4Hz,1H),6.92(d,J=7.4Hz,1H),6.74(d,J=8.1Hz,1H),6.62(d,J=2.5Hz,1H),6.47(d,J=8.3Hz,1H),5.55(s,1H),5.11(dd,J=13.1,5.1Hz,1H),4.23(d,J=17.0Hz,1H),4.12(d,J=17.2Hz,1H),3.76(s,3H),3.73(m,2H),3.52(m,1H),3.10(m,2H),2.93(m,1H),2.63(m,3H),2.33(m,3H),2.29-2.27(m,2H),2.03-1.98(m,4H),1.85(m,2H),1.78(s,3H),1.75(s,3H),1.57-1.50(m,6H),1.42(m,2H),1.34(m,2H),1.26(m,2H)。
3-(4-((9-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)壬基)氨基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(A13)的制备
Figure PCTCN2020110442-appb-000117
第1步:
将9-溴-1-壬醇(222mg,1.03mmol)加入到重氯铬酸吡啶鎓盐(333mg,1.54mmol)的THF(5ml)中,室温搅拌反应过夜。减压除去溶剂,加入20ml乙醚,使产品溶解,过滤,浓缩,得到粗品A13-2(150mg,收率68.2%),LCMS:[M+H] +=221,223。
第2步:
将A13-2(228mg,1.04mmol)和3-(4-氨基-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(A12-3)(270mg,1.04mmol)溶于DCM(10ml)和MeOH(5ml)中,氮气保护下50℃搅拌1小时。冷至室温,加入氰基硼氢化钠(98mg,1.56mmol)和2滴冰醋酸,升至50℃继续搅拌1.5小时。冷至室温,用饱和氯化铵水溶液淬灭,用二氯甲烷萃取,分层,合并有机相,无水硫酸钠干燥,静置,过滤,浓缩,粗品Flash纯化得到A13-3(100mg,收率20.9%),LCMS:[M+H] +=464,466。
第3步:
将A13-3(11mg,0.023mmol),A1-4(13mg,0.023mmol)和DIEA(16mg,0.124mmol)溶于DMF(0.5ml)中,氮气保护下于80℃搅拌7小时。减压除去溶剂,Flash纯化得到A13(2.49mg,收率11.0%), LCMS:[M+H] +=953。 1H-NMR(400MHz,DMSO-d 6)δ11.17(s,1H),11.01(s,1H),8.48(s,1H),8.06(s,1H),7.56-7.50(m,1H),7.40-7.25(m,4H),7.18(s,1H),7.09(t,J=7.5Hz,1H),6.90(dd,J=15.3,7.3Hz,1H),6.77-6.67(m,1H),6.62(d,J=2.6Hz,1H),6.47(d,J=9.2Hz,1H),5.54(t,1H),5.11(dd,J=13.2,5.0Hz,1H),4.23(d,J=17.2Hz,1H),4.12(d,J=17.2Hz,1H),3.76(s,3H),3.73(m,1H),3.70(m,1H),3.52(s,1H),3.11(d,J=6.1Hz,2H),2.99-2.86(m,1H),2.66(t,J=11.7Hz,3H),2.32(m,3H),2.26(m,2H),2.01(m,4H),1.85(d,J=12.2Hz,2H),1.78(s,3H),1.75(s,3H),1.59-1.49(m,4H),1.44-1.38(m,4H),1.34(m,2H),1.30(m,2H),1.27(m,2H),1.26(m,2H),1.25(m,2H)。
2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-N-(2-(2,6-二氧代哌啶-3-基)-1-氧代异吲哚啉-4-基)乙酰胺(A17)的制备
Figure PCTCN2020110442-appb-000118
第1步:
将A17-1(或A12-3)(200mg,0.772mmol)和TEA(234mg,2.32mmol)溶于DCM(3ml)中,氮气保护下0℃滴加氯乙酰氯(78mg,0.697mmol),滴加完毕,0℃继续搅拌2小时。LCMS监测反应完毕。加入1ml甲醇淬灭反应,40℃减压蒸除溶剂,粗品用Flash纯化得到白色固体A17-2(170mg,收率73.0%),LCMS:[M+H] +=336。
第2步:
将A17-2(30mg,0.090mmol),A1-4(51mg,0.090mmol)和DIEA(59mg,0.457mmol)加入到DMF(2ml)中,氮气保护下于60℃搅拌反应2小时。LCMS监测反应完毕。反应液用Flash纯化得到类白色固体A17(15mg,收率19.5%),LCMS:[M+H] +=869。 1H-NMR(400MHz,DMSO-d 6)δ11.17(s,1H),11.03(s,1H),8.49(s,1H),8.06(m,J=4.4Hz,2H),7.87-7.72(m,1H),7.58-7.49(m,3H),7.38(dd,J=15.7,8.0Hz,1H),7.10(t,J=7.6Hz,1H),6.71-6.60(m,1H),6.56-6.45(m,1H),5.15(dd,J=13.3,5.2Hz,2H),4.45-4.33(m,3H),3.97-3.89(m,1H),3.80-3.69(m,4H),3.49(d,J=15.9Hz,1H),3.38(s,1H),3.19(s,1H),3.02(s,4H),2.65(q,J=17.0,15.0Hz,8H),2.42-2.22(m,2H),1.90(d,J=12.9Hz,3H),1.78(s,3H),1.75(s,2H),1.28(d,J=15.9Hz,2H)。
3-(4-((2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-2-氧代乙基)氨基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(A18)的制备
Figure PCTCN2020110442-appb-000119
第1步:
将A18-1(或A12-3)(259.1mg,1mmol),2-溴乙酸叔丁酯(232.8mg,1.2mmol),KI(16.6mg,0.1 mmol)和NaHCO 3(126.0mg,1.5mmol)加入到DMF(7ml)中,氮气保护下60℃搅拌过夜。减压蒸除溶剂,粗品柱层析分离得到白色固体A18-2(150mg,收率40.2%)。
第2步:
0℃下将A18-2(44mg,0.118mmol)溶于1ml TFA中,室温搅拌1小时。40℃减压除去溶剂,得到的粗品A18-3,LCMS:[M+H] +=318。
第3步:
室温下将上述粗品A18-3分散于二氯甲烷(2ml)中,加入DIEA(81mg,0.628mmol)和HATU(44mg,0.116mmol),氮气保护下室温搅拌0.5小时。然后加入A1-4(51mg,0.090mmol)继续室温搅拌3小时。LCMS监测反应完毕。40℃减压蒸除溶剂,粗品用Flash纯化得到类白色固体A18(25mg,收率32.1%),LCMS:[M+H] +=869。 1H-NMR(400MHz,DMSO-d 6)δ11.16(s,1H),8.48(s,1H),8.05(d,J=7.4Hz,2H),7.56-7.48(m,1H),7.40-7.33(m,3H),7.19(t,J=7.6Hz,1H),7.09(t,J=7.4Hz,1H),6.92(d,J=7.4Hz,1H),6.80(d,J=7.9Hz,1H),6.68-6.60(m,1H),6.52-6.44(m,1H),5.45(s,1H),5.27(dd,J=13.3,5.2Hz,1H),4.51(d,J=6.1Hz,3H),4.24(d,J=16.9Hz,1H),4.02(d,J=16.8Hz,1H),3.75(s,4H),3.46(d,J=26.8Hz,3H),3.09(s,1H),2.84(d,J=17.2Hz,1H),2.66(t,J=12.1Hz,8H),2.30(d,J=13.6Hz,1H),2.15(s,1H),1.84(d,J=11.8Hz,1H),1.76(d,J=13.5Hz,6H),1.55(d,J=11.8Hz,1H),1.28(d,J=15.9Hz,2H),0.85(t,J=6.6Hz,1H)。
3-(4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C1)的制备
Figure PCTCN2020110442-appb-000120
第1步:
将C1-1(322mg,1mmol),庚-6-炔-1-醇(280mg,2.50mmol),CuI(38mmol,0.2mmol),Pd(PPh 3) 2Cl 2(280mg,0.4mmol)和TEA(303mg,3mmol)加入到DMF(10ml)中,氮气保护下于70℃搅拌反应过夜。冷却至室温,加入30ml水,用乙酸乙酯(20ml x 3)萃取,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品Flash纯化得到C1-2,200mg,收率56.5%,LCMS:[M+H] +=355。
第2步:
将C1-2(40mg,0.11mmol)和CBr 4(75.5mg,0.22mmol)溶于DCM(15ml)中,室温搅拌1小时。冷至0℃,氮气保护下滴加PPh 3(59.2mg,0.22mmol)的DCM(2ml)溶液,搅拌0.5小时。升至50℃继续搅拌3小时。减压除去溶剂,粗品Flash纯化得到Cl-3,200mg,收率63.8%,LCMS:[M+H] +=417,419。
第3步:
将C1-3(10mg,0.024mmol),A1-4(15mg,0.026mmol)和DIEA(16mg,0.124mmol)溶于DMF(0.5ml)中,氮气保护下于80℃搅拌7小时。减压除去溶剂,Flash纯化得到化合物C1,15mg,收率69.1%,LCMS:[M+H] +=906。
4-(2-(1-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙酰基)哌啶-4-基)乙氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C4)的制备
Figure PCTCN2020110442-appb-000121
第1步:
将A6-5(66.3mg,0.1mmol)和4-(2-溴乙基)哌啶三氟乙酸盐(30.5mg,0.1mmol)分散于5.0mL无水DCM中,依次加入T3P(159mg,0.25mmol)和DIPEA(64.5mg,0.5mmol),加毕后,室温反应2h。反应液用20mL DCM稀释后,依次用饱和氯化铵溶液(10mL x 1)和饱和NaCl洗(10mL x 1),有机层用无水MgSO 4干燥,抽滤,减压浓缩滤液得粗产物。该粗产物用Prep-TLC纯化得浅黄色固体产物C4-1,35mg,产率43.8%,LCMS:[M+H] +=801,803。
第2步:
将A2-1(11.3mg,0.041mmol),C4-1(30.0mg,0.038mmol),KI(3.1mg,0.019mmol)和NaHCO 3(9.45mg,0.113mmol)分散于2.0mL无水DMF中,氮气保护下,70℃反应20h。LCMS检测反应完全。反应液直接用Prep-HPLC纯化得浅黄色固体纯产物C4,13mg,产率35.1%,LCMS:[M+H] +=995。
2-((2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-4-基)氧基)乙酸3-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-2,2-二甲基-3-氧代丙基酯(C6)的制备
Figure PCTCN2020110442-appb-000122
第1步:
将A1-4(56.9mg,0.1mmol),3-羟基-2,2-二甲基丙酸(11.8mg,0.1mmol)和T3P(49.4mg,0.13mmol)分散于3.0mL无水DCM中,搅拌中加入DIPEA(51.6mg,0.4mmol),加毕后,室温反应2h。LCMS检测反应完全。反应液用30mL乙酸乙酯稀释后,依次用饱和NH 4Cl溶液(20mL x 1)和饱和食盐水(20mL x 1)洗涤,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物。Prep-TLC纯化得白色固体产物C6-1,65mg,产率97.0%,LCMS:[M+H] +=670。
第2步:
将C6-1(80.0mg,0.1144mmol),A3-2(57.0mg,0.1716mmol),DCC(28.3mg,0.1373mmol)和DMAP(14.0mg,0.1144mmol)分散于6.0mL无水DMF中,氮气保护下,室温搅拌8h。向反应液中补加DCC(35.3mg,0.1716mmol)和DMAP(14.0mg,0.1144mmol),继续室温反应12h。LCMS检测反应完全。反应液用50mL乙酸乙酯稀释后,依次用饱和NH 4Cl溶液(20mL x 1),H 2O(20mL x 3)和饱和食盐水(20mL x 2)洗涤,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-HPLC纯化得浅黄色固体纯产物20mg,产率17.8%,LCMS:[M+H] +=984。 1H-NMR:(400MHz,DMSO-d 6)δ11.168(s,1H),11.099(s,1H),8.477(s,1H),8.064(d,J=7.6Hz,2H),8.064(t,J =8.0Hz,1H),7.553-7.504(m,2H),7.499-7.321(m,3H),7.111(t,J=7.6Hz,1H),6.628(d,J=2.4Hz,1H),6.484(dd,J=2.4,8.8Hz,1H),5.123(s,2H),4.164(s,2H),3.760-3.710(m,5H),3.504(s,4H),2.938-2.846(m,1H),2.665-2.455(m,8H),2.373-2.324(m,1H),2.054-1.989(m,1H),1.857-1.746(m,8H),1.565-1.475(m,3H),1.192(s,6H)。
4-((9-(4-(1-(4-((4-((2-(二甲基磷酰基)苯基)氨基)-5-乙烯基嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)壬基)氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C25)的制备
Figure PCTCN2020110442-appb-000123
第1步:
将A11-1(120mg,0.288mmol),PPh 3(113.4mg,0.432mmol)和NBS(77.0mg,0.432mmol)溶于8.0mL无水DCM中,N 2保护下,室温反应2h。TLC检测无原料剩余。反应液用40mL乙酸乙酯稀释后,依次用H 2O(20mL x 1)和饱和食盐水(20mL x 1)洗涤,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物。Prep-TLC纯化得白色固体产物C25-1,100mg,产率72.6%。LCMS:[M+H] +=479,481。
第2步:
将C25-1(15mg,0.0314mmol)和C25-2(17.6mg,0.0314mmol)溶于2.0mL无水DMF中,N 2保护下,加入DIPEA(20.3mg,0.157mmol),加毕后,80℃反应8h。LCMS检测无原料剩余。反应液用20mL乙酸乙酯稀释后,依次用饱和NH 4Cl(10mL x 1),H 2O(10mL x 3)和饱和食盐水(10mL x 1)洗涤,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物。粗产物用Prep-HPLC纯化得浅黄色固体产物C25,3.0mg,产率10.0%,LCMS:[M+1] +=960。
4-((7-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)庚基)氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C47)的制备
Figure PCTCN2020110442-appb-000124
A10-2(46.3mg,0.12mmol)和C47-1(66.4mg,0.1mmol)溶于10.0mL无水DCM中,氮气保护下,加入CH 3COOH(6.0mg,0.1mmol,溶于0.5mL DCM中),室温搅拌1h。然后向反应液中加入固体NaBH 3CN(15.7mg,0.25mmol),室温继续反应2h。LCMS检测反应完全。反应液用20mL DCM稀释后,依次用饱和NH 4Cl(1 x 15mL)和饱和食盐水(1 x 15mL)洗,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-HPLC纯化得黄色固体纯产物40mg,产率38.7%。LCMS:[M+H] +=1035,1037。 1H-NMR:(400MHz,DMSO-d 6)δ12.763(s,1H),11.102(s,1H),9.017(br s,1H),8.886(d,J=1.6Hz,1H),8.856(d,J=2.0Hz,1H),8.401(s,1H),8.292(s,1H),7.951(d,J=9.6Hz,1H),7.841(t,J=8.0Hz,1H),7.532(d,J=8.4Hz,1H),7.466(d,J=7.2Hz,1H),7.380(s,1H),6.868(s,1H),5.102(dd,J=5.6,12.8Hz,1H),4.834(s,4H),4.238(t,J=6.0Hz,2H),3.797 (s,3H),3.398-3.369(m,2H),3.097-2.857(m,8H),2.575-2.542(m,1H),2.110(s,3H),2.048-2.012(m,7H),1.804-1.678(m,6H),1.572-1.371(m,10H)。
2-((1-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙酰基哌啶-4-基)氧基)-N-(2-(2,6-二氧代哌啶-3-基)-1-氧代异吲哚啉-4-基)乙酰胺(C78)的制备
Figure PCTCN2020110442-appb-000125
第1步:
C78-1(560mg,2.78mmol)溶于5.0mL无水THF中,0℃加入NaH(222.4mg,5.56mmol),室温搅拌反应2h。反应液冷却至0℃,向反应液中滴加溴乙酸(386mg,2.78mmol溶解于5.0mL无水THF中),滴加完毕后,恢复室温反应20h。0℃下,反应液用水(10mL)淬灭,然后用1M NaOH调节混合液pH=11-12后,用乙醚萃取混合液两次,残余水相用2N HCl调节pH=3-4,再用乙酸乙酯萃取(50mL x 3),合并乙酸乙酯层后,用饱和食盐水(50mL x 1)洗涤,有机层用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得粗产物C78-2(520mg,产率72.2%),该粗产物直接用于下一步反应。
第2步:
C78-2(104mg,0.4mmol)和C78-3(或A12-3)(104mg,0.4mmol)分散于8.0mL无水DCM和1.6mL无水DMF的混合溶剂中,氮气保护下依次加入T3P(508.8mg,0.8mmol)和DIPEA(258mg,2.0mmol),室温反应2h。反应液用30mL乙酸乙酯稀释后,依次用饱和NH 4Cl(20mL x 1),H 2O(20mL x 2)和饱和NaCl(20mL x 2)洗涤,有机层用无水MgSO 4干燥,抽滤,减压浓缩滤液得粗产物,该粗产物用Flash纯化得浅黄色油状产物C78-4(120mg,产率60%)。
第3步:
C78-4(120mg,0.24mmol)溶于3.0mL无水DCM中,搅拌中加入1.0mL TFA,室温反应2.0h。LCMS检测反应完全。反应液减压浓缩得粗产物,该粗产物RP-Flash纯化得白色固体产物C78-5(39mg,产率52.1%),LCMS:[M+H] +=401。
第4步:
C78-5(50mg,0.125mmol)和A6-5(78.4mg,0.125mmol)分散于5.0mL无水DCM和1.0mL无水DMF的混合溶剂中,氮气保护下,加入T3P(159mg,0.25mmol)和DIPEA(80.6mg,0.625mmol),室温搅拌2h。反应液减压浓缩得粗产物,该粗产物用RP-Flash纯化得黄色固体纯产物C78(20mg,产率15.9%),LCMS:[M+H] +=1010。
2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪 -1-基)-N-(1-(2-((2-(2,6-二氧代哌啶-3-基)-1-氧代异吲哚啉-4-基)氨基)-2-氧代乙基)哌啶-4-基)乙酰胺(C88)的制备
Figure PCTCN2020110442-appb-000126
第1步:
将化合物C88-1(或A12-3)(259.1mg,1mmol),溴乙酸叔丁酯(232.8mg,1.2mmol),碘化钾(16.6mg,0.1mmol)和碳酸氢钠(126.0mg,1.5mmol)加入到干燥的DMF(7ml)中,氮气保护下于60℃搅拌过夜。减压蒸除溶剂,粗品柱层析分离得到白色固体化合物C88-2(150mg,产率40.2%)。
第2步:
0℃下将化合物C88-2(44mg,0.118mmol)溶于1ml TFA中,室温搅拌1小时。40℃减压除去溶剂,再加入3ml甲苯,蒸除溶剂,得到的粗品C88-3,直接用于下一步,LCMS:[M+H] +=318。
第3步:
室温下将上述粗品C88-3分散于二氯甲烷(2ml)中,加入HATU(9.2mg,0.024mmol),氮气保护下室温搅拌0.5小时。然后加入DIEA(15mg,0.116mmol)和化合物C88-4(51mg,0.072mmol)继续室温搅拌4.5小时。反应完毕,40℃减压蒸除溶剂,粗品用Flash纯化得到类白色固体C88(5mg,产率19.7%),LCMS:[M+H] +=1009。
4-((1-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙酰基)氮杂环丁烷-3-基)氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C93)的制备
Figure PCTCN2020110442-appb-000127
第1步:
向一个50mL单口瓶中加入化合物C93-1(或A2-1)(116mg,0.42mmol),化合物C93-2(100mg,0.42mmol)和碳酸氢钠(106mg,1.26mmol),再加入DMF(3mL),加热至60℃,反应12小时。待反应液冷却后,加入水(30mL)用乙酸乙酯萃取三次(每次20mL),合并有机相用食盐水洗涤(15mL x 3),用无水硫酸钠干燥,过滤,减压浓缩滤液得到粗品。粗品用柱层析分离,得到浅白色固体C93-3,110mg,产率61%。
第2步:
向一个25mL单口烧瓶中加入化合物C93-3(110mg,0.256mmol)和氯化氢/1,4-二氧六环溶液(5mL,4mol/L),室温搅拌反应2小时。TLC监测反应情况。反应结束后,减压浓缩溶剂,用二氯甲烷 溶解(20mL)后,加入碳酸氢钠溶液调节pH=8-9,萃取分液后的二氯甲烷溶液用无水硫酸镁干燥,过滤,减压浓缩滤液得到浅黄色固体C93-4,51mg,产率60.7%,LCMS:[M+H] +=330。
第3步:
向一个25mL单口烧瓶中加入化合物C93-4(20mg,0.06mmol),化合物A6-5(38mg,0.06mmol),HATU(27.3mg,0.072mmol)和DIPEA(38.7mg,0.03mmol),再加入二氯甲烷(2mL)搅拌反应3小时。反应结束后,向反应液加入二氯甲烷(10mL),用水洗涤(5mL x 2),合并有机相用食盐水洗涤(5mL x 2),用无水硫酸钠干燥,过滤,减压浓缩滤液得到粗品,用prep-HPLC分离得到浅白色固体C93,5.6mg,产率10%,LCMS:[M+H] +=939。
4-((1-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙酰基)氮杂环丁烷-3-基)甲氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C94)的制备
Figure PCTCN2020110442-appb-000128
第1步:
将化合物C94-1(或A2-1)(100mg,0.365mmol),化合物C94-2(109mg,0.438mmol),碘化钾(6mg,0.036mmol)和碳酸氢钠(61mg,0.726mmol)加入到干燥的DMF(3ml)中,氮气保护下于60℃搅拌24小时。减压蒸除溶剂,粗品柱层析分离得到白色固体化合物C94-3(50mg,产率31.6%)。
第2步:
将化合物C94-3(50mg,0.113mmol)溶于三氟乙酸的二氯甲烷溶液(1ml)中,室温搅拌2小时。40℃减压除去溶剂,得到的粗品C94-4,直接用于下一步。LCMS:[M+H] +=344。
第3步:
室温下将上述粗品C94-4分散于DMF(2ml)中,加入DIEA(53mg,0.411mmol)和HATU(24mg,0.063mmol)中,氮气保护下室温搅拌1小时。然后加入化合物A6-5(40mg,0.064mmol),继续室温搅拌2.5小时。反应完毕后,40℃减压蒸除溶剂,粗品用Flash纯化得到类白色固体C94(20mg,产率36.0%),LCMS:[M+H] +=953。 1H NMR(400MHz,DMSO-d 6)δ11.16(s,1H),11.08(s,1H),8.47(d,1H),8.04(d,2H),7.87-7.81(m,1H),7.57-7.47(m,3H),7.43-7.30(m,2H),7.09(t,1H),6.62(t,1H),6.47(d,1H),5.08(m,1H),4.37(d,2H),4.32(t,1H),4.14(m,1H),3.98(t,1H),3.76(s,4H),3.71(d,2H),3.10-3.04(m,1H),2.97(s,2H),2.95-2.83(m,2H),2.70-2.60(m,4H),2.57(s,1H),2.44(s,3H),2.08-1.94(m,2H),1.83(d,2H),1.78(s,3H),1.74(s,3H),1.51(d,2H),1.25(m,3H)。
4-(2-(4-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-2-氧代乙基)哌嗪-1-基)-2-氧代乙氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C99)的制备
Figure PCTCN2020110442-appb-000129
第1步:
A3-2(223mg,0.5mmol)和C99-1(93mg,0.5mmol)分散于10.0mL无水DCM中,氮气保护下依次加入T3P(636mg,1.0mmol)和DIPEA(322.5mg,2.5mmol),室温反应2h。反应液用30mL乙酸乙酯稀释后,依次用饱和NH 4Cl(20mL x 1)和饱和NaCl(20mL x 2)洗,有机层用无水MgSO 4干燥,抽滤,减压浓缩滤液得粗产物。该粗产物用Flash纯化得白色固体产物C99-2(230mg,产率92%)。
第2步:
C99-2(230mg,0.46mmol)溶于6.0mL无水DCM中,搅拌中加入2.0mL HCl/二氧六环(4.0M),室温反应2.0h。LCMS检测反应完全。反应液减压浓缩得白色固体产物C99-3(250mg,产率100%),LCMS:[M+H] +=401。
第3步:
C99-3(109mg,0.25mmol),KI(20.8mg,0.125mmol)和NaHCO 3(84mg,1.0mmol)分散于5.0mL无水DMF中,氮气保护下,加入溴乙酸叔丁酯(48.8mg,0.25mmol),60℃反应20h。TLC检测反应完全。反应液用30mL乙酸乙酯稀释后,依次用饱和NH 4Cl(10mL x 1),H 2O(10mL x 2)和饱和NaCl(10mL x 2)洗涤,有机层用无水Na 2SO 4干燥,抽滤,减压浓缩滤液得粗产物。该粗产物用Prep-TLC纯化得浅黄色固体产物C99-4(95mg,产率74.2%),LCMS:[M+H] +=515。
第4步:
C99-4(95mg,0.185mmol)溶于3.0mL无水DCM中,搅拌中加入1.5mL HCl/二氧六环(4.0M),室温反应2.0h,抽滤,得浅黄色固体产物C99-5(70mg,产率82.6%),LCMS:[M+H] +=459。
第5步:
C99-5(70mg,0.142mmol)和A1-4(64.6mg,0.114mmol)分散于5.0mL无水DCM和1.0mL无水DMF中,氮气保护下,加入T3P(180.6mg,0.284mmol)和DIPEA(91.6mg,0.71mmol),室温搅拌2h。反应液减压浓缩得粗产物,该粗产物用Prep-HPLC纯化得黄色固体纯产物C99(25mg,产率17.5%),LCMS:[M+H] +=1010。
4-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-2-氧代乙氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C101)的制备
Figure PCTCN2020110442-appb-000130
A3-2(16.6mg,0.05mmol),A1-4(28.5mg,0.05mmol)和HATU(28.5mg,0.075mmol)分散于2.0mL无水DCM和0.2mL无水DMF的混合溶剂中,氮气保护下,滴加DIPEA(32.3mg,0.25mmol), 室温搅拌2h。反应液减压浓缩得粗产物,该粗产物用RP-Flash纯化得30mg粗产物,该粗产物继续用Prep-HPLC纯化得浅黄色固体纯产物C101(15mg,产率34.1%),LCMS:[M+H] +=884。
2-(((1-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-2-氧代乙基)环丙基)甲基)硫基)-N-(2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-4-基)乙酰胺(C103)的制备
Figure PCTCN2020110442-appb-000131
第1步:
氮气保护下0℃将化合物C103-2(或A8-1)(226mg,2.02mmol)缓慢滴加到化合物C103-1(500mg,1.83mmol)的干燥THF(10ml)中,然后升温回流4小时。降至室温,减压蒸除溶剂,粗品柱层析分离得到白色固体化合物C103-3(300mg,产率46.9%),LCMS:[M+H] +=350。
第2步:
将化合物C103-3(250mg,0.716mmol),化合物C103-4(105mg,0.716mmol)和醋酸钠(70mg,0.854mmol)加入到无水乙醇(5ml)中,搅拌回流3小时。40℃减压除去溶剂,粗品柱层析分离得到化合物C103-5(100mg,30.4%),LCMS:[M+H] +=460。
第3步:
室温下将化合物C103-5(40mg,0.087mmol),化合物A1-4(50mg,0.088mmol),DIEA(34mg,0.264mmol)和HATU(33mg,0.087mmol)溶于2ml DMF中,氮气保护下室温搅拌7小时。LCMS监测反应完毕。40℃减压蒸除溶剂,粗品用Flash纯化得到类白色固体C103(28mg,产率31.8%),LCMS:[M+H] +=1011, 1H NMR(400MHz,DMSO-d 6)δ11.16(s,1H),10.45(s,1H),8.63(d,1H),8.48(s,1H),8.05(d,2H),7.86(t,1H),7.63(d,1H),7.53(dd,1H),7.42-7.31(m,2H),7.09(t,1H),6.62(d,1H),6.47(d,1H),5.75(s,1H),5.17(dd,1H),3.76(s,3H),3.71(d,2H),3.57(s,2H),3.38(s,4H),2.79(d,2H),2.65(s,4H),2.43(s,5H),2.32(s,1H),2.13-2.05(m,1H),2.00(d,1H),1.82(s,1H),1.76(d,7H),1.48(s,3H),0.43(d,4H)。
1-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-羰基)环丙烷-1-羧酸2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-4-基酯(C104)的制备
Figure PCTCN2020110442-appb-000132
第1步:
将化合物C104-1(或A2-1)(100mg,0.365mmol),化合物C104-2(119mg,0.915mmol),DCC(90mg,0.436mmol)和DMAP(45mg,0.368mmol)溶于DMF(4ml)中,氮气保护下室温搅拌5小时。减压蒸除溶剂,粗品柱层析分离得到白色固体化合物C104-3(70mg,产率49.6%)。
第2步:
将化合物C104-3(40mg,0.104mmol),化合物A1-4(56mg,0.098mmol)HATU(40mg,0.105mmol)和DIEA(40mg,0.310mmol)加入到DMF(2ml)中,氮气保护下室温搅拌3小时。40℃减压除去溶剂,粗品用Flash纯化得到类白色固体C104(50mg,产率51.5%),LCMS:[M+H] +=938。 1H NMR(400MHz,DMSO-d 6)δ11.14(d,2H),8.48(s,1H),8.06(d,2H),7.96(t,1H),7.87(d,1H),7.70(d,1H),7.56-7.49(m,1H),7.39(d,1H),7.34(t,1H),7.09(t,1H),6.63(d,1H),6.47(dd,1H),5.14(dd,1H),3.76(s,3H),3.72(d,4H),3.51(s,2H),2.89(dt,1H),2.66(t,3H),2.39(s,1H),2.10-1.96(m,2H),1.83(d,2H),1.79-1.73(m,9H),1.55-1.50(m,3H),1.24(m,3H)。
4-((1-((4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)甲基)环丙基)甲氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C106)的制备
Figure PCTCN2020110442-appb-000133
第1步:
向一个50mL三口瓶中加入化合物C106-1(或A2-1)(274mg,1mmol),化合物C106-2(102mg,1mmol),三苯基膦(393mg,1.5mmol),再加入无水四氢呋喃(5mL),用氮气置换除氧,在氮气保护下搅拌并降温至0℃,逐滴加入DIAD(303mg,1.5mmol),加完后,升至室温搅拌反应3小时。液质监测反应,原料基本转化为产物。减压浓缩反应液得到粗品,用柱层析分离,得到目标产物C106-3,210mg,产率58.6%,LCMS:[M+H] +=359。
第2步:
向一个50mL的单口瓶中加入化合物C106-3(100mg,0.28mmol),溶于二氯甲烷中(15mL),再加入Dess-Martin试剂(305mg,0.72mmol),室温搅拌反应过夜。液质监测反应情况,原料基本消耗且有目标产物生成。先后向反应液加入碳酸氢钠饱和溶液和硫代硫酸钠饱和溶液各5mL,搅拌10分钟,体系澄清后,分液,有机相用无水硫酸钠干燥,过滤,减压浓缩滤液得到类白色固体,再用Prep-TLC分离得到浅白色固体C106-4,45mg,产率45%。LCMS:[M+H] +=357。
第3步:
向50mL单口瓶中加入化合物C106-4(41mg,0.115mmol),化合物A1-4(66mg,0.115mmol),用二氯甲烷溶解(3mL),再加入醋酸(0.05mL),室温搅拌反应0.5小时。然后加入氰基硼氢化钠(22mg,0.345mmol)反应过夜。液质监测反应情况,原料基本消耗完毕,有目标产物生成。向反应液中加入饱和氯化铵溶液(3mL),搅拌5分钟,分液,有机相用无水硫酸钠干燥,过滤,减压浓缩滤液得到粗品,再用prep-HPLC分离,得到白色粉末C106,10mg,产率9.6%。LCMS:[M+H] +=910。
2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪 -1-基)乙酸(1-(((2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-4-基)氧基)甲基)环丙基)甲基酯(C107)的制备
Figure PCTCN2020110442-appb-000134
向50mL单口烧瓶中加入化合物C106-3(28.5mg,0.08mmol)和化合物A6-5(50mg,0.08mmol),DCC(20mg,0.096mmol)和DMAP(10mg,0.08mmol),再溶于DMF中(3mL),室温搅拌反应过夜。液质监测反应情况,待原料基本转化停止反应。反应液减压浓缩得到粗品,用prep-TLC分离,得到浅黄色固体,再用prep-HPLC分离,得到白色粉末C107,10mg产率12.98%。LCMS:[M+H] +=968。
4-(((S)-1-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙酰基)吡咯烷-2-基)甲氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C109)的制备
Figure PCTCN2020110442-appb-000135
第1步:
C109-1(或A2-1)(82.2mg,0.3mmol),C109-2(66.3mg,0.33mmol)和PPh 3(94.3mg,0.36mmol)分散于7.0mL无水THF中,氮气保护下,滴加DIAD(78.8mg,0.39mmol),滴加完毕后,室温反应5.0h。反应液减压浓缩得粗产物,粗产物用Prep-HPLC纯化得白色固体产物C109-3,85mg,产率70.0%。LCMS:[M+H] +=408。
第2步:
C109-3(85mg,0.186mmol)溶于4.0mL无水DCM中,加入2.0mL 4.0M的HCl/二氧六环溶液,室温密封反应20h。LCMS检测反应完全。反应液减压浓缩得白色固体粗产物C109-4,100mg,产率100%,粗产物直接用于下一步反应。LCMS:[M+H] +=358。
第3步:
C109-4(42.8mg,0.12mmol)和A6-5(62.7mg,0.1mmol)分散于4.0mL无水DCM中,依次加入T3P(127.2mg,0.2mmol)和DIPEA(64.5mg,0.5mmol),室温反应2h。LCMS检测无原料剩余。反应液减压浓缩得粗产物,粗产物用Prep-HPLC纯化,得浅黄色固体纯产物C109,18mg,产率18.6%。LCMS:[M+H] +=967。 1H-NMR:(400MHz,DMSO-d 6)δ11.160(s,1H),11.093(d,J=3.6Hz,1H),8.476(s,1H),8.062(d,J=13.2Hz,2H),7.861-7.788(m,1H),7.577-7.320(m,5H),7.112(t,J= 7.2Hz,1H),6.622(d,J=2.8Hz,1H),6.476(t,J=2.0,1H),5.135-5.079(m,1H),4.411-4.238(m,3H),3.761(s,3H),3.707(d,J=12.0Hz,3H),3.550(d,J=5.2Hz,1H),3.179-3.122(m,1H),3.005-2.858(m,2H),2.674-2.615(m,3H),2.424-2.202(m,9H),1.997(s,4H),1.813-1.745(m,10H),1.509-1.453(m,2H)。
(2S)-1-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙酰基)-N-(2-(2,6-二氧代哌啶-3-基)-1-氧代异吲哚啉-4-基)-4,4-二氟吡咯烷-2-羧酰胺(C110)的制备
Figure PCTCN2020110442-appb-000136
第1步:
C110-1(或A12-3)(130mg,0.5mmol),C110-2(125.5mg,0.5mmol)分散于6.0mL无水DCM和1.2mL无水DMF的混合溶剂中,N 2保护下,依次加入T3P(636mg,1.0mmol)和DIPEA(258mg,2.0mmol),室温反应2h。LCMS检测无原料剩余。反应液减压浓缩除去DCM后,残余物用50mL乙酸乙酯稀释,依次用饱和NH 4Cl溶液(20mL x 1),H 2O(20mL x 3)和饱和食盐水(20mL x 1)洗涤,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物,Prep-TLC纯化得浅黄色油状产物C110-3,100mg,产率40.7%。 1H-NMR:(400MHz,DMSO-d 6)δ11.021(s,1H),10.080(s,1H),7.781-7.726(m,1H),7.575-7.523(m,2H),5.182-5.136(m,1H),4.574-4.255(m,2H),3.853(m,2H),2.962-2.890(m,2H),2.639-2.585(m,1H),2.341-2.253(m,1H),2.051-2.028(m,1H),1.416(s,3H),1.344(s,6H)。
第2步:
C110-3(100mg,0.203mmol)溶于4.0mL无水DCM,2.0mL无水THF和1.0mL无水1,4-二氧六环的混合溶剂中,加入3.0mL 4.0M的HCl/二氧六环溶液,室温密封反应2h。LCMS检测反应完全。反应液减压浓缩得白色固体粗产物C110-4,130mg,产率100%,粗产物直接用于下一步反应。LCMS:[M+H] +=393。
第3步:
A6-5(51.3mg,0.0818mmol),C110-4(35mg,0.0818mmol)分散于5.0mL无水DCM和1.0mL无水DMF的混合溶剂中,N 2保护下,依次加入T3P(104mg,0.1636mmol)和DIPEA(63.3mg,0.4908mmol),室温反应2h.LCMS检测无原料剩余。反应液减压浓缩得粗产物,粗产物用Prep-HPLC纯化得白色固体纯产物C110,25mg,产率30.6%。LCMS:[M+H] +=1002。 1H-NMR:(400MHz,DMSO-d 6)δ11.164(s,1H),11.037(s,1H),10.052(s,1H),8.477(s,1H),8.155-8.043(m,2H),7.776(t,J=6.8Hz,1H),7.566-7.499(m,3H),7.391-7.318(m,2H),7.112-7.075(m,2H),6.627-6.373(m,2H),5.201-5.130(m,1H),4.505-4.059(m,5H),3.773-3.703(m,5H),3.530(s,1H),3.286-3.275(m,2H),3.095-2.810(m,10H),2.640-2.598(m,1H),2.291–2.180(m,4H),2.008-1.838(m,3H),1.782-1.748(m, 1H),1.741(d,J=12.0Hz,6H),1.538-1.445(m,3H)。
2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)-N-(((2S)-1-((2-(2,6-二氧代哌啶-3-基)-1,3-二氧代异吲哚啉-4-基)氧基)-3,3-二甲基丁-2-基)乙酰胺(C112)的制备
Figure PCTCN2020110442-appb-000137
第1步:
向一个50mL三口瓶中加入化合物C112-1(或A2-1)(274mg,1mmol),化合物C112-2(217mg,1mmol),三苯基膦(393mg,1.5mmol),再加入无水四氢呋喃(5mL),用氮气置换除氧,在氮气保护下搅拌并降温至0℃。逐滴加入DIAD(303mg,1.5mmol),加完后,升至室温搅拌反应3小时。液质监测反应,反应结束后,减压浓缩反应液得到粗品,用柱层析分离,得到目标产物C112-3,240mg,产率50.7%,LCMS:[M+H] +=475。
第2步:
向一个25mL单口烧瓶中加入化合物C112-3(100mg,0.21mmol)和氯化氢/1,4-二氧六环溶液(5mL,4mol/L),室温搅拌反应2小时。TLC监测反应情况,反应结束后,减压浓缩除去氯化氢和大部分1,4-二氧六环,用二氯甲烷溶解(20mL)后加入碳酸氢钠溶液调节pH=8-9,分液后二氯甲烷溶液用无水硫酸镁干燥,过滤,减压浓缩滤液得到浅黄色固体C112-4,40mg,产率51%。LCMS:[M+H] +=374。
第3步:
向50mL单口烧瓶中加入化合物C112-4(40mg,0.107mmol)和A6-5(67mg,0.107mmol),溶于二氯甲烷中(3mL),再加入T 3P(136mg,0.214mmol),室温搅拌反应3小时。液质监测反应情况。反应结束后,向反应液加入二氯甲烷(10mL),加水(5mL)洗涤,分液,有机相用无水硫酸镁干燥,过滤,减压浓缩滤液得到粗品,用prep-TLC纯化,得到浅黄色固体,再用prep-HPLC分离,得到浅白色粉末C112,34mg。产率32.3%。LCMS:[M+H] +=983。
3-(4-(6-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)己-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C116)的制备
Figure PCTCN2020110442-appb-000138
第1步:
C116-1(或C1-1)(500mg,1.553mmol),CuI(59mg,0.3106mmol)和Pd(dppf)Cl 2(454mg,0.6212mmol)分散于25mL无水DMF中,N 2保护下,依次加入5-炔基-1-己醇(380mg,3.883mmol)和TEA(470mg,4.659mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,用150mL乙酸乙酯稀释反应液后,依次用饱和氯化铵(2 x 50mL),H 2O(2 x 50mL)和饱和食盐水(2 x 50mL)洗涤,有机层用无水硫酸钠干燥,抽滤,减压浓缩滤液得粗产物,RP-Flash纯化得浅黄色固体产物C116-2(255mg,48.3%)。LCMS:[M+H] +=341。
第2步:
C116-2(90mg,0.265mmol)分散于18.0mL无水DCM中,升高温度至40℃使反应液澄清,N 2保护下加入Dess-Martin试剂(168.5mg,0.3975mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入10mL饱和NaHCO 3溶液和10mL饱和Na 2S 2O 3溶液,室温剧烈搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-TLC纯化得浅黄色固体产物C116-3(35mg,39.1%)。LCMS:[M+H] +=339。
第3步:
C116-3(30mg,0.0888mmol)和A1-4(45.5mg,0.0799mmol)溶于4.0mL无水DCM和0.4mL无水甲醇的混合溶剂中,氮气保护下,加入CH 3COOH(5.3mg,0.0888mmol,溶于0.5mL DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(8.36mg,0.1332mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得白色固体产物C116,25mg,产率31.6%。LCMS:[M+H] +=892。 1H-NMR:(400MHz,DMSO-d 6)δ12.683(s,1H),11.119(s,1H),8.869(dd,J=2.0Hz,9.2Hz,3H),8.281(s,1H),8.263(s,1H),7.721(d,J=6.8Hz,1H),7.646(d,J=0.8Hz,1H),7.643(d,J=6.8Hz,1H),7.539(d,J=7.6Hz,1H),7.321(s,1H),6.809(s,1H),5.188(dd,J=5.2Hz,13.6Hz,1H),4.477(d,J=17.6Hz,1H),4.325(d,J=17.6Hz,1H),3.784(s,3H),2.513-2.495(m,10H),2.494-2.103(m,6H),2.075(s,3H),2.040(s,3H),2.004(s,3H),1.557-1.429(m,18H)。
3-(4-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)戊-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C126)的制备
Figure PCTCN2020110442-appb-000139
第1步:
C126-1(或C1-1)(322mg,1.0mmol),CuI(19mg,0.1mmol)和Pd(dppf)Cl 2(73.1mg,0.1mmol)分散于10.0mL无水DMF中,N 2保护下,依次加入4-炔基-1-戊醇(210mg,2.5mmol)和TEA(303mg,3.0mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,RP-Flash纯化得白色固体粗产物C126-2(410mg,产率94.1%)。LCMS:[M+H] +=327。
第2步:
C126-2(400mg,0.920mmol)溶解于150mL无水DCM和10mL无水THF的混合溶剂中,N 2保护下加入Dess-Martin试剂(1.04g,2.454mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入20mL饱和NaHCO 3溶液和20mL饱和Na 2S 2O 3溶液,室温剧烈搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Flash纯化得浅黄色固体产物C126-3(280mg,93.3%)。LCMS:[M+H] +=325。
第3步:
C126-3(48.6mg,0.15mmol)和A1-4(76.8mg,0.135mmol)溶于5.0mL无水DCM和0.5mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(13.5mg,0.225mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(14.1mg,0.225mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得白色固体纯产物C126(35mg,产率26.7%)。LCMS:[M+H] +=878。
4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C148)的制备
Figure PCTCN2020110442-appb-000140
第1步:
将化合物C148-1(或A1-1)(330mg,0.98mmol),化合物C148-2(220mg,1.96mmol),碘化 亚铜(38mg,0.20mmol),Pd(PPh 3) 2Cl 2(274mg,0.39mmol)和三乙胺(297mg,2.94mmol)溶于干燥的DMF(10ml)中,氮气保护下于70℃搅拌过夜.降至室温,减压蒸除溶剂,粗品柱层析分离得到化合物C148-3(90mg,产率24.9%)。LCMS:[M+H] +=369。
第2步:
将化合物C148-3(60mg,0.163mmol)和四溴化碳(108mg,0.326mmol)溶于二氯甲烷(20ml)中,室温搅拌2小时。降至0℃,滴加三苯基膦(86mg,0.326mmol)的二氯甲烷(2ml)溶液,自然升至室温,然后于55℃搅拌反应2小时。减压除去溶剂,粗品柱层析分离得到化合物C148-4(40mg,57.1%)。LCMS:[M+H] +=431,433。
第3步:
将化合物C148-4(40mg,0.093mmol),化合物A1-4(58mg,0.102mmol),DIEA(60mg,0.465mmol)溶于2ml DMF中,氮气保护下80℃搅拌5小时。LCMS监测反应完毕。40℃减压蒸除溶剂,粗品用Flash纯化得到C148(22mg,产率25.9%)。LCMS:[M+H] +=920。 1H NMR(400MHz,DMSO-d 6)δ11.13(d,2H),8.47(s,1H),8.04(d,2H),7.87(dd,3H),7.52(dd,1H),7.36(dd,2H),7.09(t,1H),6.62(d,1H),6.46(dd,1H),5.75(s,1H),5.14(dd,1H),3.76(s,3H),3.71(d,2H),3.26(s,1H),2.92-2.83(m,1H),2.69-2.60(m,4H),2.55(d,4H),2.34(d,5H),2.03(m,2H),1.84(d,2H),1.76(d,6H),1.60(m,3H),1.47(s,6H)。
4-(3-((7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚基)氧基)氮杂环丁烷-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C150)的制备
Figure PCTCN2020110442-appb-000141
第1步:
向100mL三口烧瓶中加入化合物C150-1(470mg,2.72mmol),溶于无水四氢呋喃中(6mL),在氮气保护下降温至0℃,再加入钠氢(120mg,2.98mmol),然后升至室温搅拌反应30分钟。再降温至0℃,加入化合物C150-2(842mg,3.26mmol),室温反应3小时。TLC监测反应情况,待原料基本转化后停止反应。向反应液加入冰水(3g),再用乙酸乙酯萃取2次(每次10mL),合并有机相用饱和食盐水洗涤(5mL x 2),用无水硫酸钠干燥,过滤,减压浓缩滤液得到粗品,用柱层析分离,得到浅黄色固体C150-3,100mg,产率10.6%。
第2步:
向50mL单口烧瓶中加入化合物C150-3(35mg,0.1mmol)和化合物A1-4(57mg,0.1mmol)DIPEA(38.7mg,0.3mmol)溶于DMF中(2mL)加热至70℃反应2小时,液质监测反应,反应结束后,冷却,向反应液加入乙酸乙酯(10mL)和水(5mL)分液后,再用乙酸乙酯萃取1次,合并有机相,用饱和食盐水洗涤(5mL x 2),用无水硫酸钠干燥,过滤,减压浓缩滤液得到粗品,用Prep-TLC 分离,得到浅白色固体C150-4,35mg,产率41.7%。
第3步:
向一个25mL单口烧瓶中加入化合物C150-4(30mg,0.035mmol)和氯化氢/1,4-二氧六环溶液(5mL,4mol/L),室温搅拌反应2小时。TLC监测反应情况。反应结束后,减压浓缩溶剂,用二氯甲烷溶解(20mL)后加入碳酸氢钠溶液调节pH=8-9,分液后二氯甲烷溶液用无水硫酸镁干燥,过滤,减压浓缩滤液得到浅黄色固体C150-5,25mg,产率99%。LCMS:[M+H] +=739。
第4步:
向一个25mL单口瓶中加入化合物C150-5(20mg,0.027mmol),C150-6(或A6-1)(8.8mg,0.032mmol),DIPEA(10.4mg,0.081mmol)和NMP(2mL),在氮气保护下加热至90℃反应3小时。液质监测有少量产品生成,反应液直接用prep-HPLC分离得到浅白色粉末C150,0.7mg,产率2.6%,LCMS:[M+H] +=995。
3-(4-((7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚基)氧基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C153)的制备
Figure PCTCN2020110442-appb-000142
第1步:
C153-1(或A12-3)(2.59g,10.0mmol)和联硼酸频那醇酯(2.794g,11.0mmol)分散于80.0mL无水MeCN中,氮气保护下滴加亚硝酸特丁酯(1.545g,15.0mmol),滴加完毕后,室温反应4.0h。反应液用80mL乙酸乙酯稀释后,搅拌五分钟,抽滤,滤液减压浓缩得粗产物,Flash纯化得浅黄色固体产物C153-2(2.1g,56.7%)。LCMS:[M+H] +=371。
第2步:
C153-2(2.1g,5.68mmol)分散于63.0H 2O 2(30%)中,搅拌中加入TBAB(548mg,1.70mmol),空气中反应1h。LCMS检测反应完全。反应液用150mL乙酸乙酯稀释后,搅拌五分钟,分出有机层,水相反萃一次,合并有机层后,用饱和食盐水(50mL x 2)洗涤,有机层用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得粗产物,Flash纯化得浅黄色固体产物C153-3(0.71g,48.2%)。LCMS:[M+H] +=261。
第3步:
C153-3(130mg,0.5mmol),K 2CO 3(103.5mg,0.75mmol)和KI(41.5mg,0.25mmol)分散于13.0mL无水MeCN中,加入7-溴-1-庚醇(195mg,1.0mmol),85℃反应1.5h。反应液冷却至室温后,抽滤除去固体残渣,滤液减压浓缩得粗产物。该粗产物Prep-TLC纯化得浅黄色固体产物C153-4(115mg,61.5%)。LCMS:[M+H] +=375。
第4步:
C153-4(75mg,0.2mmol)溶于10.0mL无水DCM中,加入Dess-Martin试剂(127.5mg,0.3mmol),50℃回流反应2h。反应液冷却至室温,用10mL DCM稀释后,加入5mL饱和NaHCO 3溶液和5mL饱和Na 2S 2O 3溶液,搅拌5min,分出有机层用饱和NaCl溶液(10mL x 1)洗,再用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得粗产物,用Prep-TLC纯化得浅黄色固体产物C153-5(40mg,53%)。LCMS:[M+H] +=373。
第5步:
C153-5(40mg,0.1075mmol)和A1-4(55.1mg,0.0968mmol)溶于8.0mL无水DCM中,氮气保护下,滴加CH 3COOH(6.45mg,0.1075mmol)后,室温搅拌30min。然后加入NaBH 3CN(62.8mg,0.1613mmol),室温搅拌反应2h。反应液用30mL DCM稀释后,依次用饱和NH 4Cl溶液(15mL x 1)和饱和NaCl溶液(10mL x 1)洗涤,有机层用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得粗产物,用Prep-HPLC纯化得白色固体纯产物C153(16mg,产率16.2%)。LCMS:[M+H] +=926。 1H-NMR:(400MHz,DMSO-d 6)δ11.163(s,1H),10.962(s,1H),8.480(brs,1H),8.061(d,J=13.2Hz,1H),7.534-7.492(m,2H),7.473-7.292(m,3H),7.241-7.221(m,1H),7.110(t,J=7.2Hz,1H),6.623(d,J=2.4Hz,1H),6.477(dd,J=2.4,8.8Hz,1H),5.127(dd,J=5.2,13.6Hz,1H),4.390(d,J=17.6Hz,1H),4.244(d,J=17.2Hz,1H),4.128(t,J=6.4Hz,2H),3.758(s,3H),3.720-3.690(m,2H),3.300-3.277(m,1H),2.954-2.864(m,1H),2.690-2.558(m,4H),2.328-2.204(m,7H),2.010-1.974(m,2H),1.855-1.745(m,10H),1.549-1.237(m,12H)。
4-((7-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)庚基)氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C158)的制备
Figure PCTCN2020110442-appb-000143
A10-2(46.3mg,0.12mmol)和C158-1(67.9mg,0.1mmol)溶于10.0mL无水DCM中,氮气保护下,加入CH 3COOH(6.0mg,0.1mmol,溶于0.5mL DCM中),室温搅拌1h。然后向反应液中加入固体NaBH 3CN(15.7mg,0.25mmol),室温继续反应2h。LCMS检测反应完全。反应液用20mL DCM稀释后,依次用饱和NH 4Cl(1 x 15mL)和饱和食盐水(1 x 15mL)洗,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-HPLC纯化得黄色固体纯产物C158,36mg,产率34.3%。LCMS:[M+H] +=1050。 1H-NMR:(400MHz,DMSO-d 6)δ11.749(s,1H),11.096(s,1H),8.887-8.849(m,3H),8.341(s,1H),8.288(s,1H),7.948(d,J=9.6Hz,1H),7.841(t,J=7.2Hz,1H),7.531(d,J=8.8Hz,1H),7.467(d,J=7.2Hz,1H),7.406(s,1H),6.767(s,1H),5.102(dd,J=5.2,12.8Hz,1H),4.583(s,6H),4.236(t,J=6.4Hz,3H),3.789(s,3H),3.198-2.700(m,9H),2.625-32.532(m,3H),2.111(s,3H),2.049(s,4H),2.013(s,3H),1.800-1.647(m,5H),1.495-1.376(m,6H),1.238(s,2H)。
3-(4-(1-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)戊基)-1H-吡唑-4-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C162)的制备
Figure PCTCN2020110442-appb-000144
第1步:
向50mL单口瓶中加入化合物C162-1(或C1-1)(300mg,1mmol),化合物C162-2(440mg,1.5mmol),Pd(dppf)Cl 2(73.1mg,0.1mmol)和无水碳酸钠(414mg,3mmol),再加入DMF(10mL)和水(2mL),用氮气脱气除氧并在氮气保护下加热至90℃用微波反应器反应1小时。待反应液冷却后,加入水(30ml)用乙酸乙酯萃取三次(每次20mL),合并有机相用食盐水洗涤(25mL x 3),用无水硫酸钠干燥,过滤,减压浓缩滤液得到粗品。粗品用柱层析分离,得到浅白色固体C162-3,140mg,产率34%。
第2步:
向25mL单口烧瓶中加入化合物C162-3(70mg,0.17mmol)和二氯甲烷(3mL),搅拌溶解后再加入三氟乙酸(2mL),室温搅拌反应1小时。TLC监测反应情况,反应结束后,减压浓缩除去二氯甲烷和大部分三氟乙酸,用二氯甲烷溶解(10mL)后加入碳酸氢钠溶液调节pH=8-9,分液后二氯甲烷溶液用无水硫酸镁干燥,过滤,减压浓缩滤液得到浅黄色固体C162-4,51mg,产率96%。LCMS:[M+H] +=311。
第3步:
向25mL单口烧瓶中加入化合物C162-4(50mg,0.16mmol)和化合物C162-5(36.8mg,0.16mmol)以及碳酸钾(66.24mg,0.48mmol),再加入DMF(3mL),搅拌并加热至45℃反应过夜。液质监测反应,反应完成后,反应液冷却,向其中加入水(10mL)用乙酸乙酯萃取三次(每次5mL),合并有机相用食盐水洗涤(5mL x 3),用无水硫酸钠干燥,过滤,减压浓缩滤液得到粗品。粗品用薄层制备色谱分离,得到浅黄色蜡状固体C162-6,22mg,产率30.1%,LCMS:[M+H] +=459,461。
第4步:
向25mL单口瓶中加入化合物C162-6(20mg,0.053mmol),化合物A1-4(30.5mg,0.053mmol)和二异丙基乙基胺(20.5mg,0.159mmol),再加入DMF(2mL),搅拌溶解并加热至70℃反应2小时。液质监测反应情况,反应结束后,反应液冷却后,减压浓缩得到粗品。粗品用高效制备液相分离得到白色固体C162,9mg,产率:17.9%。LCMS:[M+H] +=948。
3-(5-(7-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C163)的制备
Figure PCTCN2020110442-appb-000145
第1步:
将化合物C163-1(5g,16.35mmol),化合物C163-2(3.49g,21.25mmol),三乙胺(2.15g,21.25mmol)溶于80mL乙腈中,氮气保护下80℃搅拌过夜。冷至室温,过滤,固体用乙腈淋洗,母液浓缩,用少量乙腈洗,合并固体,干燥,得到白色固体化合物C163-3,直接用于下一步(7.03g,产率84.4%)。LCMS:[M+H] +=323,325。
第2步:
将化合物C163-3(322mg,1mmol),化合物C163-4(280mg,2.5mmol),碘化亚铜(38mg,0.2mmol),Pd(dppf)Cl 2(280mg,0.4mmol)和三乙胺(30.3mg,3mmol)溶于10mL无水DMF中,氮气保护下于70℃搅拌反应过夜。减压除去溶剂,粗品薄层色谱纯化得到类白色固体化合物C163-5(135mg,产率38.1%),LCMS:[M+H] +=355。
第3步:
将化合物C163-5(45mg,0.127mmol),Dess-Martin试剂(162mg,mmol)溶于20mL二氯甲烷中,于50℃搅拌2小时。降至室温,用饱和碳酸氢钠水溶液淬灭,分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C163-6(26.7mg,产率60.7%),LCMS:[M+H] +=353。
第4步:
将化合物C163-6(26.7mg,0.076mmol)和C163-7(或C158-1)(51.5mg,0.076mmol)溶于3mL二氯甲烷/甲醇(2/1)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(4.8mg,0.076mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C163(15mg,产率19.5%),LCMS:[M+H] +=1016,1018。
4-((7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚基-7,7-二氘)氧基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C164)的制备
Figure PCTCN2020110442-appb-000146
第1步:
A1-4(100mg,0.176mmol)和C164-1(30.8mg,0.211mmol)分散于10.0mL无水DCM中,氮 气保护下依次加入T3P(224mg,0.352mmol)和DIPEA(90.8mg,0.704mmol),室温反应2h。反应液用20mL二氯甲烷稀释后,依次用饱和NH 4Cl(10mL x 1)和饱和NaCl(10mL x 1)洗,有机层用无水MgSO 4干燥,抽滤,减压浓缩滤液得粗产物。该粗产物用Prep-TLC纯化得黄色固体产物C164-2(45mg,36.7%)。LCMS:[M+H] +=698。
第2步:
C164-2(45mg,0.065mmol)溶于10.0mL无水THF中,氮气保护下,加入LiAlD 4(27.1mg,0.65mmol),70℃反应2.0h。LCMS检测反应完全。反应液冷却至0℃,用1.0mL H2O淬灭反应,然后向反应液中加入5滴1mol/L NaOH溶液,搅拌5min,分出有机层后减压浓缩得粗产物C164-3(35mg,77.7%),该粗产物直接用于下一步反应。LCMS:[M+H] +=686。
第3步:
C164-3(35mg,0.051mmol),C164-4(或A2-1)(15.4mg,0.056mmol)和PPh 3(20mg,0.077mmol)溶于6.0mL无水THF中,氮气保护下,加入DIAD(15.4mg,0.077mmol),室温反应2h。LCMS检测反应完全。反应液减压浓缩得粗产物,该粗产物用Prep-HPLC纯化得浅黄色固体产物C164(10mg,产率20.8%)。LCMS:[M+H] +=942。
3-(4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-5-氟-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C166)的制备
Figure PCTCN2020110442-appb-000147
第1步:
向一个500mL单口瓶中加入化合物C166-1(10g,64.87mmol),再加入硫酸(200mL),搅拌溶解后再加入NBS(10.4g,58.44mmol),室温搅拌反应5小时。液质监测反应情况,反应结束后,把反应液加入冰水中(1000g)淬灭反应,析出白色固体,过滤收集,滤饼用水洗涤三次,每次200mL得到含水的白色固体,50℃真空干燥得到白色粉末C166-2混合物12g,直接用于下一步反应。
第2步:
向一个250mL的单口瓶中加入化合物C166-2混合物(6g,25.75mmol),溶于甲醇中(100mL),降温至0℃,再滴加二氯亚砜(7.67g,64.37mmol),滴加完毕,升温至回流反应2小时。TLC监测反应情况,待原料基本转化停止反应。将反应液减压浓缩除去大部分溶剂和二氯亚砜,再向浓缩后的溶液中加入冰水,用乙酸乙酯萃取(100mL x 3),合并有机相,先后用碳酸氢钠饱和溶液,水和食盐水洗涤(50mL x 2),无水硫酸钠干燥,过滤,减压浓缩滤液得到浅黄色油状物5.2g,产率:81.7%。直接用于下一步反应。
第3步:
向250mL单口瓶中加入化合物C166-3的混合物(3g,12.1mmol),用氯仿溶解(100mL),再 先后加入NBS(2.16g,12.1mmol)和AIBN(196.8mg,1.21mmol),搅拌并加热至回流反应5小时。TLC监测反应情况,待原料基本反应转化为产物。停止反应,反应液冷却后,过滤除去不溶物,母液用水洗涤三次(每次20mL),用无水硫酸镁干燥,过滤,减压浓缩滤液得到粗品,粗品用柱层析分离,得到C166-4无色油状物2.2g。
第4步:
向250mL单口瓶中先后加入化合物C166-4(2g,12.24mmol),化合物C166-5(4g,12.24mmol),然后加入乙腈(100mL),最后加入三乙胺(2.56mL,36.72mmol),加热至回流温度,反应过夜。待反应液冷却后析出固体,继续析晶5小时。然后过滤得到浅白色固体,用乙腈洗涤(20mL x 2),于40℃真空干燥得到C166-6浅白色粉末700mg,产率:16.8%。LCMS:[M+H] +=341,343。
第5步:
向100mL三口烧瓶中加入化合物C166-6(700mg,2.05mmol)和庚-6-炔-1-醇(392mg,3.5mmol),Pd(PPh 3) 2Cl 2(490mg,0.35mmol),碘化亚铜(66.8mg,0.35mmol)以及三乙胺(530mg,5.25mmol),再加入DMF(20mL)溶解后在氮气保护下加热至70℃反应5小时。液质监测反应情况,反应结束后,反应液冷却后减压浓缩得到粗品,用柱层析分离,得到C166-7浅黄色固体170mg,产率22.3%。LCMS:[M+H] +=373.2。
第6步:
向50mL装有冷凝器的三口瓶中加入化合物C166-7(170mg,0.46mmol)溶于THF中(3mL)再加入三溴氧膦(170mg,0.59mmol)加热至70℃反应2小时,液质监测反应情况,反应结束后,冷却,向反应液加入乙酸乙酯(10mL)然后用碳酸氢钠饱和溶液洗涤两次,每次5mL,再用食盐水洗涤(5mL x 2),最后用无水硫酸钠干燥,过滤,减压浓缩滤液得到粗品,粗品用薄层色谱制备色谱分离,得到C166-8浅白色固体39mg,产率19.5%,LCMS:[M+H] +=435,437。
第7步:
向一个装有冷凝管的25mL单口瓶中加入化合物C166-8(40mg,0.11mmol),A1-4(61.2mg,0.11mmol),二异丙基乙基胺(41.7mg,0.33mmol)和DMF(2mL),加热至70℃反应3小时。液质监测反应情况,待原料基本转化成目标产物,停止反应。反应液冷却后,减压浓缩得到粗品,用prep-HPLC分离,得到C166白色粉末10mg,产率10%。LCMS:[M+H] +=924。
3-(4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-6-氟-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C167)的制备
Figure PCTCN2020110442-appb-000148
第1步:
C167-1(1.0g,4.06mmol)溶于10.0mL CCl 4中,氮气保护下,加入NBS(1.1g,6.1mmol) 和AIBN(266.5mg,1.624mmol),升高温度至90℃,回流反应20h。LCMS检测无原料剩余。反应液冷却至室温后,抽滤,减压浓缩滤液得粗产物,Flash纯化得浅黄色油状产物C167-2(静置变为固体)1.2g。
第2步:
C167-2(1.2g,3.69mmol)和3-氨基哌啶-2,6-二酮盐酸盐(787mg,4.80mmol)分散于25.0mL无水MeCN中,加入TEA(485mg,4.80mmol),升高温度至80℃,回流反应16h。LCMS检测反应完全。反应液冷却至室温后,抽滤,滤饼用MeCN淋洗三次,烘干固体得产物C167-3(1.0g,产率80%)。LCMS:[M+H] +=341,343。
第3步:
C167-3(340mg,1.0mmol),CuI(38mg,0.2mmol)和Pd(dppf)Cl 2(292.4mg,0.4mmol)分散于17.0mL无水DMF中,N 2保护下,依次加入庚-6-炔-1-醇(281mg,2.5mmol)和TEA(303mg,3.0mmol),升高温度至70℃反应16h。LCMS检测反应完全。反应液冷却至室温,用80mL乙酸乙酯稀释反应液后,依次用饱和氯化铵(2 x 40mL),H 2O(2 x 40mL)和饱和食盐水(2 x 40mL)洗,有机层用无水硫酸钠干燥,抽滤,减压浓缩滤液得粗产物,RP-Flash纯化得浅黄色固体产物C167-4(95mg,25.5%)。LCMS:[M+H] +=373。
第4步:
C167-4(90mg,0.242mmol)分散于25.0mL无水DCM中,N 2保护下,加入Dess-Martin试剂(154mg,0.363mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入10mL饱和NaHCO 3溶液和10mL饱和Na 2S 2O 3溶液,室温搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-TLC纯化得浅黄色固体产物C167-5(55mg,60%)。LCMS:[M+H] +=371。
第5步:
C167-5(30.0mg,0.081mmol)和A1-4(46.1mg,0.081mmol)溶于5.5mL无水DCM中,氮气保护下,加入CH 3COOH(4.86mg,0.081mmol,溶于0.5mL DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(10.2mg,0.162mmol),室温继续反应2h。LCMS和TLC检测反应完全。反应液用20mL DCM稀释后,依次用饱和NH 4Cl(1 x 15mL)和饱和食盐水(1 x 15mL)洗,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物,用Prep-HPLC纯化得白色固体纯产物25mg,产率33.5%。LCMS:[M+H] +=924。 1H-NMR:(400MHz,DMSO-d 6)δ11.190(s,1H),11.051(s,1H),8.490(br s,1H),8.098(s,1H),8.066(s,1H),7.563-7.507(m,3H),7.397-7.315(m,2H),7.110(t,J=6.4Hz,1H),6.628(d,J=2.4Hz,1H),6.479(dd,J=2.4Hz,8.4Hz,1H),5.182(dd,J=5.2Hz,13.2Hz,1H),4.462(dd,J=17.2Hz,60.0Hz,2H),3.758(s,3H),3.725(d,J=12.8Hz,2H),2.967-2.876(m,2H),2.672-2.503(m,3H),2.501-2.397(m,6H),2.398-2.247(m,3H),2.032-1.974(m,1H),1.832-1.748(m,12H),1.5615-1.527(m,2H),1.498-1.436(m,6H)。
3-(4-(3-(2-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)乙氧基)丙-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C169)的制备
Figure PCTCN2020110442-appb-000149
第1步:
向100mL单口烧瓶中加入化合物C169-1(或C1-1)(309mg,1mmol),化合物C169-2(200mg,2mmol),Pd(dppf)Cl 2(280mg,0.4mmol),碘化亚铜(38.2mg,0.2mmol)以及三乙胺(303mg,3mmol),再加入DMF(20mL),溶解后用氮气置换三次除氧,在氮气保护下加热至70℃反应5小时。液质监测反应情况。反应结束后,冷却,减压浓缩得到粗品,用柱层析分离,得到C169-3浅黄色固体70mg,产率20.3%。LCMS:[M+H] +=343。
第2步:
向50mL装有冷凝器的三口瓶中加入化合物C169-3(70mg,0.2mmol),溶于THF中(2mL),再加入三溴氧膦(170mg,0.59mmol),加热至70℃反应2小时。液质监测反应情况。反应完成后,冷却,向反应液加入乙酸乙酯(10mL),然后用碳酸氢钠饱和溶液洗涤(5mL x 2),再用食盐水洗涤(5mL x 2),最后用无水硫酸钠干燥,过滤,减压浓缩滤液得到粗品。粗品用薄层制备色谱分离,得到C169-4浅白色固体24mg,产率29.7%,LCMS:[M+H] +=405,407。
第3步:
向一个装有冷凝管的25mL单口瓶中加入化合物C169-4(12mg,0.029mmol),化合物C169-5(或C158-1)(19.3mg,0.029mmol),二异丙基乙基胺(11.2mg,0.087mmol)和DMF(1mL),加热至70℃反应3小时。液质监测反应情况。反应结束后,冷却,减压浓缩得到粗品,用prep-HPLC分离,得到C169白色粉末10mg,产率34.5%。LCMS:[M+H] +=1004。
4-(7-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)庚-1-炔-1-基)-2-(2,6-二氧代哌啶-3-基)异吲哚啉-1,3-二酮(C171)的制备
Figure PCTCN2020110442-appb-000150
第1步:
将化合物C171-1(或C148-3)(56.6mg,0.154mmol),Dess-Martin试剂(196mg,0.461mmol)溶于二氯甲烷(20ml)中,50℃搅拌反应2h。降至室温,用饱和碳酸氢钠水溶液淬灭反应,分层,水相用二氯甲烷萃取三次,合并有机相,饱和食盐水洗涤(15mL x 2),无水硫酸钠干燥,过滤,减压除去溶剂,粗品柱层析分离得到化合物C171-2(45mg,产率80.4%)。LCMS:[M+H] +=367。
第2步:
将化合物C171-2(45mg,0.12mmol)和化合物C47-1(82mg,0.12mmol)溶于二氯甲烷和甲醇(2/1)(3ml)中,加入1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(7.8mg,0.12mmol),继续搅拌1小时。浓缩除去溶剂,粗品Flash纯化得到化合物C171(15mg,产率14.7%),LCMS:[M+H] +=1015,1017。
3-(4-(8-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)辛-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C172)的制备
Figure PCTCN2020110442-appb-000151
第1步:
向100mL单口烧瓶中加入化合物C172-1(或C1-1)(309mg,1mmol),化合物C172-2(252mg,2mmol),双三苯基膦二氯化钯(280mg,0.4mmol),碘化亚铜(38.4mg,0.2mmol)以及三乙胺(303mg,3mmol),再加入DMF(20mL),溶解后用氮气置换三次除氧,在氮气保护下加热至70℃反应5小时。液质监测反应情况。反应结束后,反应液冷却,减压浓缩得到粗品,用柱层析分离,得到C172-3浅黄色固体200mg,产率54.3%。LCMS:[M+H] +=369.4。
第2步:
向100mL单口烧瓶中加入化合物C172-3(200mg,0.543mmol)和二氯甲烷(40mmol),搅拌溶解后再加入Dess-Martin试剂(345mg,0.814mmol),加热至40℃反应2小时。液质监测反应情况。待原料基本转化为产物,停止反应,反应液用冰浴冷却至0-5℃,先后加入碳酸氢钠饱和溶液(10mL)和硫代硫酸钠溶液(10mL)搅拌约10分钟,分液后再用二氯甲烷萃取两次(每次20mL),合并有机相用饱和食盐水洗涤(10mL x 2),用无水硫酸钠干燥后,减压浓缩得到C172-4白色粘稠状固体180mg,直接用于下一步反应。LCMS:[M+H] +=367.2。
第3步:
向25mL单口烧瓶中加入化合物C172-4(40mg,0.11mmol)和化合物C47-1(59.7mg,0.09mmol)溶于二氯甲烷(5mL)中,再加入醋酸(10mg,0.16mmol)室温搅拌反应30分钟,再加入氰基硼氢化钠(9.5mg,0.15mmol)室温反应3小时,液质监测反应情况,待原料基本转化为产品,停止反应,向反应加入饱和氯化铵(5mL)搅拌5分钟,分液,再用二氯甲烷萃取两次,每次10mL,合并有机相,用饱和食盐水洗涤(5mL x 2),用无水硫酸钠干燥后,减压浓缩,用Prep-HPLC分离,得到C172黄色粉末20mg,产率18%。LCMS:[M+H] +=1001。
3-(4-(9-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)壬-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C173)的制备
Figure PCTCN2020110442-appb-000152
第1步:
C173-1(或C1-1)(644mg,2.0mmol),CuI(76mg,0.4mmol)和Pd(dppf)Cl 2(585mg,0.8mmol)分散于34mL无水DMF中,N 2保护下,依次加入8-炔基-1-壬醇(700mg,5.0mmol)和TEA(606mg,6.0mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,用150mLEA稀释反应液后,依次用饱和氯化铵(2 x 50mL),H 2O(2 x 50mL)和饱和食盐水(2 x 50mL)洗,有机层用无水硫酸钠干燥,抽滤,减压浓缩滤液得粗产物,RP-Flash纯化得浅黄色固体产物C173-2(210mg,27.5%)。LCMS:[M+H] +=383。
第2步:
C173-2(200mg,0.524mmol)分散于30.0mL无水DCM中,N 2保护下,加入Dess-Martin试剂(333mg,0.785mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入10mL饱和NaHCO 3溶液和10mL饱和Na 2S 2O 3溶液,室温搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物,该粗产物用Prep-TLC纯化得浅黄色固体产物C173-3(150mg,75%)。LCMS:[M+H] +=381。
第3步:
C173-3(45mg,0.1184mmol)和C47-1(70.8mg,0.1066mmol)溶于8.0mL无水DCM和0.8mL无水甲醇的混合溶剂中,氮气保护下,加入CH 3COOH(7.1mg,0.1184mmol,溶于0.5mL DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(11.2mg,0.1776mmol),室温继续反应2h。LCMS和TLC检测反应完全。反应液用20mL DCM稀释后,依次用饱和NH 4Cl(1 x 15mL)和饱和食盐水(1 x 15mL)洗,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物,用Prep-HPLC纯化得黄色固体纯产物45mg,产率40.0%。LCMS:[M+H] +=1029,1031。
1H-NMR:(400MHz,DMSO-d 6)δ12.683(s,1H),11.119(s,1H),8.869(dd,J=2.0Hz,9.2Hz,3H),8.281(s,1H),8.263(s,1H),7.721(d,J=6.8Hz,1H),7.646(d,J=0.8Hz,1H),7.643(d,J=6.8Hz,1H),7.539(d,J=7.6Hz,1H),7.321(s,1H),6.809(s,1H),5.188(dd,J=5.2Hz,13.6Hz,1H),4.477(d,J=17.6Hz,1H),4.325(d,J=17.6Hz,1H),3.784(s,3H),2.513-2.495(m,10H),2.494-2.103(m,6H),2.075(s,3H),2.040(s,3H),2.004(s,3H),1.557-1.429(m,18H)。
3-(4-(3-(2-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺并[5.5]十一烷-3-基)乙氧基)丙-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C174)的制备
Figure PCTCN2020110442-appb-000153
向一个装有冷凝管的25mL单口瓶中加入化合物C169-4(2mg,0.029mmol),化合物C47-1(1mg,0.029mmol),二异丙基乙基胺(11.2mg,0.087mmol)和DMF(1mL),加热至70度反应3小时。液质监测反应情况,反应结束后,冷却,减压浓缩得到粗品,用prep-HPLC分离,得到C174白色粉末9mg,产率31.2%。LCMS:[M+H] +=989,991。
3-(4-(3-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙氧基)丙-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C176)的制备
Figure PCTCN2020110442-appb-000154
第1步:
将化合物C176-1(或C1-1)(322mg,1.00mmol),化合物C176-2(200mg,2.00mmol),碘化亚铜(38mg,0.20mmol),Pd(PPh 3) 2Cl 2(280mg,0.40mmol)和三乙胺(303mg,3.00mmol)溶于干燥的DMF(10ml)中,氮气保护下于70℃搅拌过夜。降至室温,减压蒸除溶剂,粗品柱层析分离得到化合物C176-3(80mg,产率23.4%)。LCMS:[M+H] +=343。
第2步:
将化合物C176-3(52mg,0.152mmol)和四溴化碳(101mg,0.305mmol)溶于二氯甲烷(20ml)中,室温搅拌2小时。降至0℃,滴加PPh 3(80mg,0.305mmol)的二氯甲烷(2ml)溶液,自然升温至室温,然后于55℃搅拌反应2小时。减压除去溶剂,粗品柱层析分离得到化合物C176-4(20mg,产率32.8%)。LCMS:[M+H] +=405,407。
第3步:
将化合物C176-4(20mg,0.050mmol),化合物A1-4(31mg,0.054mmol),DIEA(32mg,0.248mmol)溶于1ml DMF中,氮气保护下80℃搅拌5小时。LCMS监测反应完毕。40℃减压蒸除溶剂,粗品用Flash纯化得到C176(18mg,产率40.9%)。LCMS:[M+H] +=894。 1H NMR(400MHz,DMSO-d 6)δ11.17(s,1H),11.01(s,1H),8.48(s,1H),8.05(d,2H),7.75(dd,2H),7.54(dt,2H),7.36(dd,2H),7.09(t,1H),6.62(d,1H),6.47(dd,1H),5.75(s,1H),5.16(dd,1H),4.50(d,1H),4.35(d,1H),3.76(s,3H),3.71(d,2H),3.65(t,2H),3.28(s,1H),3.00-2.87(m,2H),2.66(d,4H),2.57(s,1H),2.44(d,4H),2.29(s,2H),2.01(p,3H),1.84(d,2H),1.76(d,6H),1.57-1.42(m,3H)。
3-(4-(8-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)辛-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C177)的制备
Figure PCTCN2020110442-appb-000155
向25mL单口烧瓶中加入化合物C172-4(40mg,0.11mmol)和A1-4(51.2mg,0.09mmol)溶于二氯甲烷(5mL)中,再加入醋酸(10mg,0.16mmol)室温搅拌反应30分钟。再加入氰基硼氢化钠(9.5mg,0.15mmol),室温反应3小时。液质监测反应情况。反应结束后,向反应液中加入饱和氯化铵(5mL)搅拌5分钟,分液,再用二氯甲烷萃取(10mL x 2),合并有机相,用饱和食盐水洗涤(5mL x 2),用无水硫酸钠干燥后,减压浓缩,用Prep-HPLC分离,得到黄色粉末18mg,产 率19.5%。LCMS:[M+H] +=920。
3-(4-(9-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)壬-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C178)的制备
Figure PCTCN2020110442-appb-000156
C173-3(48mg,0.1263mmol)和A1-4(64.7mg,0.1137mmol)溶于8.0mL无水DCM和0.8mL无水甲醇的混合溶剂中,氮气保护下,加入CH 3COOH(7.6mg,0.1263mmol,溶于0.5mL DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(11.9mg,0.1895mmol),室温继续反应2h。LCMS和TLC检测反应完全。反应液用20mL DCM稀释后,依次用饱和NH 4Cl(1 x 15mL)和饱和食盐水(1 x 15mL)洗,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-TLC纯化得粗产物55mg,粗产物继续用Prep-HPLC纯化得白色固体纯产物35mg,产率29.7%。LCMS:[M+H] +=934。 1H-NMR:(400MHz,DMSO-d 6)δ11.190(s,1H),11.089(s,1H),8.481(brs,1H),8.092(d,J=10.0Hz,2H),7.721(d,J=7.6Hz,1H),7.644(d,J=7.6Hz,1H),7.539-7.502(m,2H),7.340(dd,J=8.4Hz,17.2Hz,2H),7.070(t,J=14.4Hz,1H),6.621(s,1H),6.476(d,J=8.8Hz,1H),5.184(dd,J=4.8Hz,13.2Hz,1H),4.474(d,J=17.6Hz,1H),4.321(d,J=17.6Hz,1H),3.757-3.694(m,5H),2.932-2.842(m,2H),2.636-2.530(m,3H),2.444-2.380(m,4H),2.357-2.160(m,7H),1.989-1.933(m,2H),1.809-780(m,2H),1.743(s,3H),1.709(s,3H),1.566-1.355(m,8H),1.319-1.157(m,5H)。
3-(4-(10-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)癸-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C179)的制备
Figure PCTCN2020110442-appb-000157
第1步:
C179-1(或C1-1)(322mg,1.0mmol),CuI(38mg,0.2mmol)和Pd(dppf)Cl 2(292.4mg,0.4mmol)分散于15mL无水DMF中,N 2保护下,依次加入9-炔基-1-癸醇(385mg,2.5mmol)和TEA(303mg,3.0mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,用80mL EA稀释反应液后,依次用饱和氯化铵(2 x 50mL),H 2O(2 x 50mL)和饱和食盐水(2 x 50mL)洗,有机层用无水硫酸钠干燥,抽滤,减压浓缩滤液得粗产物,RP-Flash纯化得浅黄色固体产 物C179-2(105mg,26.5%)。LCMS:[M+H] +=397。
第2步:
C179-2(35mg,0.088mmol)溶解于10.0mL无水DCM中,N 2保护下加入Dess-Martin试剂(75mg,0.176mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入10mL饱和NaHCO 3溶液和10mL饱和Na 2S 2O 3溶液,室温剧烈搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物,该粗产物用Prep-TLC纯化得浅黄色固体产物C179-3(15mg,42.8%)。LCMS:[M+H] +=395。
第3步:
C179-3(15mg,0.0381mmol)和A1-4(19.5mg,0.0343mmol)溶于3.0mL无水DCM中,氮气保护下,加入CH 3COOH(2.3mg,0.0381mmol,溶于0.5mL DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(3.59mg,0.0572mmol),室温继续反应2h。LCMS和TLC检测反应完全。反应液直接用Prep-TLC纯化得粗产物15mg,该粗产物继续用Prep-HPLC纯化得白色固体纯产物C179(7.0mg,产率19.5%)。LCMS:[M+H] +=948。
3-(4-(3-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)丙氧基)丙-1-炔-1-基)-1-氧代异吲哚啉-2-基哌啶-2,6-二酮(C181)的制备
Figure PCTCN2020110442-appb-000158
第1步:
0℃下将氢氧化钾(3.75g,66.8mmol),分批加入到化合物C181-1(3.68g,25.0mmol)和化合物C181-2(1.99g,26.2mmol)的混合物的甲苯溶液中,剧烈搅拌,然后升温至室温,继续搅拌1小时。反应液加水稀释,用100mL乙酸乙酯萃取,有机相用无水硫酸钠干燥,过滤,减压除去溶剂,粗品柱层析分离得到1.2g液体,产率42.9%。
第2步:
将化合物C181-4(或C1-1)(200mg,0.62mmol),化合物C181-3(566mg,4.97mmol),碘化亚铜(24mg,0.12mmol),Pd(dppf)Cl 2(174mg,0.25mmol)和三乙胺(188mg,1.86mmol)溶于5mL无水DMF中,氮气保护下于70℃搅拌反应5小时。减压除去溶剂,粗品薄层色谱纯化得到类白色固体化合物C181-4(60mg,产率27.1%),LCMS:[M+H] +=357。
第3步:
将化合物C181-4(20mg,0.056mmol),Dess-Martin试剂(36mg,0.084mmol)溶于20mL二氯甲烷中,于50℃搅拌2小时.降至室温,用饱和碳酸氢钠水溶液淬灭,分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C181-5 (18mg,产率90.0%),LCMS:[M+H] +=355。
第4步:
将化合物C181-5(18mg,0.051mmol)和化合物A1-4(32mg,0.056mmol)溶于3mL二氯甲烷/甲醇(2/1,v/v)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时.然后加入氰基硼氢化钠(3.2mg,0.051mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C181(15mg,产率32.6%),LCMS:[M+H] +=908。 1H NMR(400MHz,DMSO-d 6)δ11.15(s,1H),11.00(s,1H),8.50(dr,1H),8.04(d,2H),7.71(dd,1H),7.63(d,1H),7.57-7.44(m,2H),7.36(m,2H),7.06(d,1H),6.64(s,1H),6.43(d,1H),5.29(t,1H),5.13(dd,1H),4.46(d,1H),4.38(s,2H),4.31(d,1H),3.72(s,3H),3.67(d,2H),3.53(t,2H),2.98(m,1H),2.67(m,4H),2.59(m,3H),2.25(m,4H),2.02(m,2H),1.96(q,2H),1.69(s,3H),1.68(s,3H),1.45(s,2H),1.21(d,4H)。
3-(4-(3-(4-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)丁氧基)丙-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C182)的制备
Figure PCTCN2020110442-appb-000159
第1步:
0℃下将氢氧化钾(3.75g,66.8mmol),分批加入到化合物C182-1(3.68g,25.0mmol)和化合物C182-2(2.36g,26.2mmol)的混合物的甲苯溶液中,剧烈搅拌,然后升至室温,继续搅拌1小时。反应液加水稀释,用100mL乙酸乙酯萃取,有机相用无水硫酸钠干燥,过滤,减压除去溶剂,粗品柱层析分离得到1.4g液体C182-3,产率45.2%。
第2步:
将化合物C182-4(或C1-1)(200mg,0.62mmol),化合物C182-3(6366mg,4.97mmol),碘化亚铜(24mg,0.12mmol),Pd(dppf)Cl 2(174mg,0.25mmol)和三乙胺(188mg,1.86mmol)溶于5mL无水DMF中,氮气保护下于70℃搅拌反应5小时。减压除去溶剂,粗品薄层色谱纯化得到类白色固体化合物C182-5(60mg,产率26.1%),LCMS:[M+H] +=371。
第3步:
将化合物C182-5(30mg,0.081mmol),Dess-Martin试剂(52mg,0.1224mmol)溶于20mL二氯甲烷中,于50℃搅拌2小时。降至室温,用饱和碳酸氢钠水溶液淬灭,分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C182-6(28mg,产率93.3%),LCMS:[M+H] +=369。
第4步:
将化合物C182-6(20mg,0.051mmol)和化合物A1-4(32mg,0.056mmol)溶于3mL二氯甲烷/ 甲醇(2/1)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(3.2mg,0.051mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C182(10mg,产率20.8%),LCMS:[M+H] +=948。 1H NMR(400MHz,DMSO-d 6)δ11.18(s,1H),11.05(s,1H),8.50(dr,1H),8.08(d,2H),7.78(dd,1H),7.72(d,1H),7.56-7.44(m,2H),7.36(m,2H),7.09(d,1H),6.62(s,1H),6.48(d,1H),5.32(t,1H),5.15(dd,1H),4.53(d,1H),4.37(s,2H),4.32(d,1H),3.75(s,3H),3.69(d,2H),3.54(t,2H),2.97(m,1H),2.66(m,4H),2.56(m,3H),2.49(m,4H),2.02(m,2H),1.96(q,2H),1.69(s,3H),1.68(s,3H),1.45(s,3H),1.23(d,5H)。
3-(4-(9-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)壬-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C183)的制备
Figure PCTCN2020110442-appb-000160
C173-3(30mg,0.079mmol)和C183-1(或C158-1)(48.2mg,0.071mmol)溶于4.0mL无水DCM和0.4mL无水甲醇的混合溶剂中,氮气保护下,加入CH 3COOH(4.74mg,0.079mmol,溶于0.5mL DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(7.44mg,0.1185mmol),室温继续反应2h。LCMS和TLC检测反应完全。反应液用20mL DCM稀释后,依次用饱和NH 4Cl(1 x 15mL)和饱和食盐水(1 x 15mL)洗,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物,用Prep-HPLC纯化得黄色固体纯产物25mg,产率30.4%。LCMS:[M+H] +=1044,1046。 1H-NMR:(400MHz,DMSO-d 6)δ12.696(s,1H),11.089(s,1H),8.876(dd,J=2.0Hz,7.6Hz,3H),8.272(d,J=1.6Hz,2H),7.722(d,J=7.6Hz,1H),7.646(d,J=6.8Hz,1H),7.629(t,J=0.8Hz,1H),7.347(s,1H),6.748(s,1H),5.187(dd,J=5.2Hz,13.2Hz,1H),4.478(d,J=17.6Hz,1H),4.325(d,J=17.6Hz,1H),3.775(s,3H),3.345-2.831(m,4H),2.522-2.469(m,4H),2.468-2.185(m,9H),2.087(s,3H),2.043(s,3H),2.007(s,3H),1.574-1.560(m,2H),1.432-1.230(m,16H)。
3-(4-(3-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙氧基)丙-1-炔-1-基)-6-氟-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C184)的制备
Figure PCTCN2020110442-appb-000161
第1步:
C167-3(340mg,1.0mmol),CuI(19.0mg,0.10mmol)和Pd(dppf)Cl 2(73.1mg,0.10mmol)分散于17.0mL无水DMF中,N 2保护下,依次加入C184-1(200.2mg,2.0mmol)和TEA(303mg,3.0mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,用100mL乙酸乙酯稀释后,依次用饱和氯化铵(2 x 50mL),H 2O(2 x 50mL)和饱和食盐水(2 x 50mL)洗,有机层用无水硫酸钠干燥,抽滤,减压浓缩滤液得粗产物,RP-Flash纯化得白色固体产物C184-2(175mg,48.6%)。LCMS:[M+H] +=361。
第2步:
C184-2(70mg,0.195mmol)溶于14.0mL无水DCM中,N 2保护下加入Dess-Martin试剂(124mg,0.293mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入10mL饱和NaHCO 3溶液和10mL饱和Na 2S 2O 3溶液,室温剧烈搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-TLC纯化得浅黄色固体产物C184-3(40mg,57.2%)。LCMS:[M+H] +=359。
第3步:
C184-3(40mg,0.112mmol)和A1-4(51mg,0.0896mmol)溶于5.0mL无水DCM和0.5mL无水甲醇的混合溶剂中,氮气保护下,加入CH 3COOH(10.08mg,0.168mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(14.1mg,0.224mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得白色固体纯产物C184(20mg,产率19.6%)。LCMS:[M+H] +=912。
3-(4-(7-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)庚-1-炔-1-基)-6-氟-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C185)的制备
Figure PCTCN2020110442-appb-000162
第1步:
C167-5(18.0mg,0.0486mmol)和C47-1(32.3mg,0.0486mmol)溶于4.0mL无水DCM中,氮气保护下,加入CH 3COOH(2.9mg,0.0486mmol,溶于0.5mL DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(6.1mg,0.0972mmol),室温继续反应2h。LCMS和TLC检测反应完全。反应液用20mL DCM稀释后,依次用饱和NH 4Cl(1 x 15mL)和饱和食盐水(1 x 15mL)洗涤,有机层用无水Na 2SO 4干燥,抽滤,滤液减压浓缩得粗产物,用Prep-HPLC纯化得黄色固体纯产物13mg,产率26.3%。LCMS:[M+H] +=1019,1021。 1H-NMR:(400MHz,DMSO-d 6)δ11.688(s,1H),11.051(s,1H),8.877(dd,J=2.0Hz,11.2Hz,3H),8.307(s,1H),8.272(d,J=2.0Hz,1H),7.936(d,J=7.6Hz,1H),7.586-7.541(m,2H),7.328(s,1H),6.820(s,1H),5.191(dd,J=5.2Hz,13.2Hz,1H),4.474(dd,J=18.8Hz,60.0Hz,2H),3.787(s,3H),3.505(s,1H),3.347(s,3H),2.942-2.619(m,6H),2.570(s,4H),2.081-2.004(m,9H),1.639-1.432(m,11H),1.232(s,5H)。
3-(4-(6-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)己-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C186)的制备
Figure PCTCN2020110442-appb-000163
C116-3(25mg,0.0740mmol)和C183-1(或C158-1)(50.2mg,0.0740mmol)溶于4.0mL无水DCM和0.4mL无水甲醇的混合溶剂中,氮气保护下,加入CH 3COOH(4.44mg,0.0740mmol,溶于0.5DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(6.97mg,0.1110mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得黄色固体产物C186,23mg,产率31.0%。LCMS:[M+H] +=1002,1004。 1H-NMR:(400MHz,DMSO-d 6)δ11.027(s,1H),8.874-8.850(m,4H),8.271-8.255(m,2H),7.937(d,J=9.6Hz,1H),7.728(d,J=1.2Hz,1H),7.709(d,J=1.2Hz,1H),7.655-7.634(m,1H),7.357(s,1H),6.752(s,1H),5.181(dd,J=4.0Hz,13.6Hz,1H),4.484(d,J=17.6Hz,1H),4.335(d,J=17.6Hz,1H),3.776(s,3H),3.328-3.107(m,2H),2.967-2.892(m,1H),2.682-2.334(m,13H),2.091-2.007(m,12H),1.884-1.855(m,2H),1.600-1.567(m,6H),1.236(s,2H)。
3-(4-(6-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)己-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C187)的制备
Figure PCTCN2020110442-appb-000164
C116-3(25mg,0.0740mmol)和C47-1(49.1mg,0.0740mmol)溶于4.0mL无水DCM和0.4mL无水甲醇的混合溶剂中,氮气保护下,加入CH 3COOH(4.44mg,0.0740mmol,溶于0.5mL DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(6.97mg,0.1110mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得黄色固体纯产物C187,30mg,产率41.1%。LCMS:[M+H] +=987,989。 1H-NMR:(400MHz,DMSO-d 6)δ11.026(s,1H),8.864(dd,J=2.0Hz,9.6Hz,4H),8.265(s,2H),7.940(d,J=11.2Hz,1H),7.723(dd,J=1.2Hz,7.6Hz,1H),7.653(dd,J=1.2Hz,7.6Hz,1H),7.544(t,J=7.6Hz,1H),7.327(s,1H),6.815(s,1H),5.181-5.135(m,1H),4.483(d,J=16.8Hz,1H),4.334(d,J=17.2Hz,1H),3.783(s,3H),2.933-2.821(m,1H),2.807-2.797(m,4H),2.621-2.577(m,3H),2.503-2.335(m,6H),2.081(s,3H),2.040(s,4H),2.004(s,4H),1.600-1.528(m,12H)。
3-(4-(10-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)癸-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C188)的制备
Figure PCTCN2020110442-appb-000165
C179-3(30mg,0.076mmol)和C183-1(或C158-1)(43.9mg,0.0647mmol)溶于8.0mL无水DCM中,氮气保护下,加入CH 3COOH(4.56mg,0.076mmol,溶于0.5mL DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(7.16mg,0.114mmol),室温继续反应2h。LCMS和TLC检测反应完全。反应液直接用20mL DCM稀释后,依次用饱和氯化铵(1 x 10mL)和饱和食盐水(1 x 10mL)洗,有机层用无水硫酸钠干燥,抽滤,减压浓缩滤液得粗产物,用Prep-HPLC纯化得黄色固体纯产物C188(19.5mg,产率24.4%)。LCMS:[M+H] +=1058,1060。
3-(4-(10-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)癸-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C189)的制备
Figure PCTCN2020110442-appb-000166
C179-3(30mg,0.076mmol)和C47-1(40.4mg,0.0608mmol)溶于5.0mL无水DCM中,氮气保护下,加入CH 3COOH(4.56mg,0.076mmol,溶于0.5mL DCM中),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(7.16mg,0.114mmol),室温继续反应2h。LCMS和TLC检测反应 完全。用Prep-HPLC纯化得黄色固体纯产物C189(25mg,产率31.5%)。LCMS:[M+H] +=1043,1045。
3-(5-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C190)的制备
Figure PCTCN2020110442-appb-000167
将化合物C163-6(35mg,0.099mmol)和化合物A1-4(62mg,0.109mmol)溶于3mL二氯甲烷/甲醇(2/1)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(6.2mg,0.099mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C190(15mg,产率16.7%),LCMS:[M+H] +=906。
3-(5-(7-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)庚-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C191)的制备
Figure PCTCN2020110442-appb-000168
将化合物C163-6(35mg,0.099mmol)和化合物C47-1(73mg,0.109mmol)溶于3mL二氯甲烷/甲醇(2/1)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(6.2mg,0.099mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到黄色固体化合物C191(20mg,产率20.2%),LCMS:[M+H] +=1001,1003。
3-(5-(6-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)己-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C192)的制备
Figure PCTCN2020110442-appb-000169
第1步:
将化合物C192-1(100mg,0.31mmol),化合物C192-2(76mg,0.78mmol),碘化亚铜(12mg,0.06mmol),Pd(dppf)Cl 2(87mg,0.12mmol)和三乙胺(94mg,0.93mmol)溶于4mL无水DMF中,氮气保护下于70℃搅拌反应5小时。减压除去溶剂,粗品薄层色谱纯化得到类白色固体化合物C192-3(78mg,产率72.2%),LCMS:[M+H] +=341。
第2步:
将化合物C192-3(39mg,0.115mmol),Dess-Martin试剂(73mg,0.172mmol)溶于15mL二氯甲烷中,于50℃搅拌2小时。降至室温,用饱和碳酸氢钠水溶液淬灭,分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C192-4(30mg,产率77.5%),LCMS:[M+H] +=339。
第3步:
将化合物C192-4(26.6mg,0.079mmol)和化合物A1-4(49mg,0.087mmol)溶于3mL二氯甲烷/甲醇(2/1)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(5mg,0.079mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C192(10mg,产率14.3%),LCMS:[M+H] +=892。
3-(5-(3-(2-(4-(1-(4-((5-氯-4-((2-((二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙氧基)丙-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C193)的制备
Figure PCTCN2020110442-appb-000170
第1步:
将化合物C193-1(或C192-1)(100mg,0.31mmol),化合物C193-2(78mg,0.78mmol),碘化 亚铜(12mg,0.06mmol),Pd(dppf)Cl 2(87mg,0.12mmol)和三乙胺(94mg,0.93mmol)溶于4mL无水DMF中,氮气保护下于70℃搅拌反应5小时。减压除去溶剂,粗品薄层色谱纯化得到类白色固体化合物C193-3(70mg,产率66.0%),LCMS:[M+H] +=343。
第2步:
将化合物C193-3(23.2mg,0.068mmol),Dess-Martin试剂(43.1mg,0.102mmol)溶于15mL二氯甲烷中,于50℃搅拌2小时。降至室温,用饱和碳酸氢钠水溶液淬灭,分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C193-4(20mg,产率87.0%),LCMS:[M+H] +=341。
第3步:
将化合物C193-4(20mg,0.059mmol)和化合物A1-4(37mg,0.065mmol)溶于3mL二氯甲烷/甲醇(2/1)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(3.7mg,0.059mmol)继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C193(15mg,产率28.8%),LCMS:[M+H] +=894。
3-(5-(8-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)辛-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C194)的制备
Figure PCTCN2020110442-appb-000171
第1步:
将化合物C194-1(或C192-1)(100mg,0.31mmol),化合物C194-2(98mg,0.78mmol),碘化亚铜(12mg,0.06mmol),Pd(dppf)Cl 2(87mg,0.12mmol)和三乙胺(94mg,0.93mmol)溶于4mL无水DMF中,氮气保护下于70℃搅拌反应5小时.减压除去溶剂,粗品薄层色谱纯化得到类白色固体化合物C194-3(68mg,产率59.6%),LCMS:[M+H] +=369。
第2步:
将化合物C194-3(27mg,0.073mmol),Dess-Martin试剂(46.7mg,0.110mmol)溶于16mL二氯甲烷中,于50℃搅拌2小时。降至室温,用饱和碳酸氢钠水溶液淬灭,分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C194-4(20.6mg,产率76.6%),LCMS:[M+H] +=367。
第3步:
将化合物C194-4(20.6mg,0.056mmol)和化合物A1-4(35mg,0.062mmol)溶于3mL二氯甲烷/甲醇(2/1)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(3.5mg,0.056mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C194(15mg,产率29.0%),LCMS:[M+H] +=920。
3-(5-(9-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)壬-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C195)的制备
Figure PCTCN2020110442-appb-000172
第1步:
将化合物C195-1(或C192-1)(100mg,0.31mmol),化合物C195-2(109mg,0.78mmol),碘化亚铜(12mg,0.06mmol),Pd(dppf)Cl 2(87mg,0.12mmol)和三乙胺(94mg,0.93mmol)溶于4mL无水DMF中,氮气保护下于70℃搅拌反应5小时。减压除去溶剂,粗品薄层色谱纯化得到类白色固体化合物C195-3(72mg,产率61.0%),LCMS:[M+H] +=383。
第2步:
将化合物C195-3(23mg,0.060mmol),Dess-Martin试剂(38.3mg,0.090mmol)溶于16mL二氯甲烷中,于50℃搅拌2小时。降至室温,用饱和碳酸氢钠水溶液淬灭,分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C195-4(20mg,产率87.0%),LCMS:[M+H] +=381。
第3步:
将化合物C195-4(20mg,0.053mmol)和A1-4(33mg,0.058mmol)溶于3mL二氯甲烷/甲醇(2/1)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(3.5mg,0.053mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C195(15mg,产率30.6%),LCMS:[M+H] +=934。
3-(5-(10-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)癸-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C196)的制备
Figure PCTCN2020110442-appb-000173
第1步:
将化合物C196-1(或C192-1)(100mg,0.31mmol),化合物C196-2(120mg,0.78mmol),碘 化亚铜(12mg,0.06mmol),Pd(dppf)Cl 2(87mg,0.12mmol)和三乙胺(94mg,0.93mmol)溶于4mL无水DMF中,氮气保护下于70℃搅拌反应5小时.减压除去溶剂,粗品薄层色谱纯化得到类白色固体化合物C196-3(65mg,产率52.8%),LCMS:[M+H] +=397。
第2步:
将化合物C195-3(23.9mg,0.060mmol),Dess-Martin试剂(38.4mg,0.090mmol)溶于15mL二氯甲烷中,于50℃搅拌2小时。降至室温,用饱和碳酸氢钠水溶液淬灭,分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C196-4(20mg,产率84.0%),LCMS:[M+H] +=395。
第3步:
将化合物C196-4(20mg,0.051mmol)和化合物A1-4(32mg,0.056mmol)溶于3mL二氯甲烷/甲醇(2/1)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(3.2mg,0.051mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C196(10mg,产率20.8%),LCMS:[M+H] +=948。
3-(4-(3-(2-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)乙氧基)丙-1-炔-1-基)-1-氧代-6-(三氟甲基)异吲哚啉-2-基)哌啶-2,6-二酮(C200)的制备
Figure PCTCN2020110442-appb-000174
第1步:
0℃搅拌下向浓硫酸(20mL)中分批加入化合物C200-1(2g,9.8mmol),NBS(1.63g,9.16mmol)氮气保护下保持该温度搅拌反应3小时。升至室温搅拌过夜。将反应液缓慢倒入冰水中,混合物用乙酸乙酯萃取三次,合并有机相,用0.5M盐酸、饱和食盐水各洗一次,无水硫酸钠干燥,过滤,浓缩,粗品Flash纯化得到化合物C200-2(1.3g,产率47.1%)。LCMS:[M+H] +=283,285。
第2步:
0℃下向化合物C200-2(1.3g,4.61mmol)的20mL甲醇溶液中滴加氯化亚砜(2mL,25.57mmol),然后70℃加热搅拌过夜。降至室温,减压浓缩,粗品Flash纯化得到化合物C200-3(1.25g,产率91.9%)。
第3步:
将化合物C200-3(1.25g,4.22mmol),NBS(857mg,4.81mmol),AIBN(69mg,0.42mmol)溶于20mL氯仿中,氮气保护下95℃回流过夜。减压除去溶剂,粗品用Flash纯化,得到白色固体化 合物C200-4(1.3g,产率82.3%)。
第4步:
将化合物C200-4(1.33g,3.56mmol),3-氨基哌啶-2,6-二酮盐酸盐(759mg,4.62mmol),三乙胺(467mg,4.62mmol)溶于15mL乙腈中,氮气保护下80℃搅拌过夜。冷至室温,减压除去溶剂,粗品用Flash纯化得到白色固体化合物C200-5(790mg,产率56.9%)。LCMS:[M+H]+=391,393。
第5步:
将化合物C200-5(100mg,0.256mmol),化合物C169-2(64mg,0.641mmol),碘化亚铜(4.9mg,0.026mmol),Pd(dppf)Cl 2(18.8mg,0.026mmol)和三乙胺(77.7mg,0.769mmol)溶于3mL无水DMF中,氮气保护下于70℃搅拌过夜。减压除去溶剂,粗品薄层色谱纯化得到化合物C200-6(100mg,产率95.2%),LCMS:[M+H] +=411。
第6步:
将化合物C200-6(126mg,0.307mmol),Dess-Martin试剂(261mg,0.615mmol)溶于10mL二氯甲烷中,于50℃搅拌2小时。降至室温,顺序加入饱和碳酸氢钠水溶液和饱和硫代硫酸钠水溶液各搅拌5分钟使反应淬灭。分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C200-7(44mg,产率35.2%),LCMS:[M+H] +=409。
第7步:
将化合物C200-7(22mg,0.054mmol)和化合物A1-4(21.5mg,0.038mmol)溶于1.5mL二氯甲烷/甲醇(2/1,v/v)中,滴加冰醋酸(3.2mg,0.054mmol),氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(2.4mg,0.038mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C200(18mg,产率34.7%),LCMS:[M+H] +=962。 1H NMR(400MHz,DMSO-d 6)δ11.17(s,1H),11.04(s,1H),8.47(s,1H),8.11(d,1H),8.06(s,1H),8.05(d,2H),7.57-7.49(m,1H),7.38(d,1H),7.33(d,1H),7.09(t,1H),6.62(d,1H),6.47(dd,1H),5.34-5.30(m,1H),5.19(dd,1H),4.62(d,1H),4.48(s,3H),3.76(s,3H),3.73(s,1H),3.67(t,3H),3.29(m,3H),2.93(m,1H),2.70-2.61(m,4H),2.52(d,3H),2.01(m,4H),1.84(d,2H),1.76(d,6H),1.55-1.45(m,3H)。
3-(6-氯-4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C202)的制备
Figure PCTCN2020110442-appb-000175
第1步:
将化合物C202-1(1g,3.82mmol),NBS(679mg,4.35mmol),AIBN(62mg,0.38mmol)溶于20mL氯仿中,氮气保护下95℃回流反应5小时。减压除去溶剂,粗品用Flash纯化,得到白色固体化合物C202-2(600mg,产率46.2%)。
第2步:
将化合物C202-2(1.31g,3.85mmol),化合物C202-3(822mg,5.01mmol),三乙胺(506mg,5.01mmol)溶于10mL乙腈中,氮气保护下80℃搅拌过夜。冷至室温,减压除去溶剂,粗品用Flash纯化得到白色固体化合物C202-4(1.0g,产率73.0%)。LCMS:[M+H] +=357,359。
第3步:
将化合物C202-4(100mg,0.281mmol),化合物C202-5(79mg,0.702mmol),碘化亚铜(5.4mg,0.028mmol),Pd(dppf)Cl 2(21mg,0.028mmol)和三乙胺(85mg,0.843mmol)溶于2mL无水DMF中,氮气保护下于70℃搅拌反应5小时。减压除去溶剂,粗品薄层色谱色谱制备得到类白色固体化合物C202-6(94mg,产率87.9%),LCMS:[M+H] +=389。
第4步:
将化合物C202-6(50mg,0.129mmol),Dess-Martin试剂(82mg,0.193mmol)溶于10mL二氯甲烷中,于50℃搅拌2小时。降至室温,用饱和碳酸氢钠水溶液淬灭,分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C202-7(34mg,产率68.0%),LCMS:[M+H] +=387。
第5步:
将化合物C202-7(34mg,0.0881mmol)和化合物A1-4(35mg,0.062mmol)溶于1.5mL二氯甲烷/甲醇(2/1,v/v)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(3.9mg,0.062mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C202(10mg,产率12.1%),LCMS:[M+H] +=940。 1H NMR(400MHz,DMSO-d 6)δ11.18(s,1H),11.03(s,1H),8.48(s,1H),8.06(s,2H),7.72(dd,2H),7.57-7.49(m,1H),7.35(dd,2H),7.09(t,1H),6.62(d,1H),6.47(dd,1H),5.32(m,1H),5.15(dd,1H),4.46(d,1H),4.31(d,1H),3.76(s,3H),3.71(d,2H),2.97-2.87(m,1H),2.65(t,3H),2.59(d,3H),2.34-2.32(m,4H),2.27(m,3H),2.03-1.97(m,2H),1.82(d,2H),1.76(d,7H),1.59(t,2H),1.48(d,7H)。
7-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-2-(2,6-二氧代哌啶-3-基)-3-氧代异吲哚啉-5-腈(C204)的制备
Figure PCTCN2020110442-appb-000176
第1步:
C204-1(532.5mg,1.5mmol)溶于10.0mL CCl 4中,氮气保护下,加入NBS(351mg,1.95mmol)和AIBN(73.8mg,0.45mmol),升高温度至90℃,回流反应20h。TLC监测还有三分之一的原料剩余。向反应液中补加NBS(405mg,2.55mmol)和AIBN(123mg,0.75mmol),补加完毕后,继续回流反应20h。TLC检测无原料剩余。反应液冷却至室温后,抽滤,减压浓缩滤液得粗产物,Flash纯化得浅黄色油状产物,静置变为固体,C204-2(690mg,100%)。
第2步:
C204-2(690mg,1.6mmol)和C204-3(341mg,2.08mmol)分散于12.0mL无水MeCN中,加入TEA(210mg,2.08mmol),升高温度至80℃,回流反应16h。LCMS检测反应完全。反应液冷却至室温后,抽滤,滤饼用MeCN淋洗三次,烘干固体得目标产物C204-4(450mg,产率62.8%)。LCMS:[M+H] +=449,451。
第3步:
C204-4(380mg,0.848mmol)和Pd(PPh 3) 4(98mg,0.0848mmol)分散于20.0mL无水DMF中,N 2保护下,加入Zn(CN) 2(109.5mg,0.933mmol),升高温度至80℃反应2h。LCMS检测有原料剩余。向反应液中补加Zn(CN) 2(149.3mg,1.272mmol)和Pd(PPh 3) 4(284mg,0.2544mmol),补加完毕后,80℃继续反应18h。LCMS检测反应完全。反应液冷却至室温,直接RP-Flash纯化得褐色固体产物C204-5(170mg,57.8%)。LCMS:[M+H] +=348,350。
第4步:
C204-5(160mg,0.461mmol),CuI(17.5mg,0.092mmol)和Pd(dppf)Cl 2(67.4mg,0.092mmol)分散于20.0mL无水DMF中,N 2保护下,依次加入6-炔基-1-庚醇(129.1mg,1.153mmol)和TEA(140mg,1.383mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,直接RP-Flash纯化得棕黄色固体产物C204-6(140mg,91.4%)。LCMS:[M+H] +=380。
第5步:
C204-6(140mg,0.369mmol)溶解于28.0mL无水DCM中,N 2保护下,加入Dess-Martin试剂(313mg,0.738mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入20mL饱和NaHCO 3溶液和20mL饱和Na 2S 2O 3溶液,室温搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-TLC纯化得浅黄色固体产物C204-7(80mg,产率57.1%)。LCMS:[M+H] +=378。
第6步:
C204-7(45mg,0.119mmol)和A1-4(61mg,0.107mmol)溶于4.0mL无水DCM和0.4mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(10.7mg,0.179mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(15.0mg,0.238mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得白色固体纯产物C204(35mg,产率31.8%)。LCMS:[M+H] +=931。
3-(4-((7-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)庚基)氧基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C211)的制备
Figure PCTCN2020110442-appb-000177
C153-5(35mg,0.094mmol)和C47-1(56.2mg,0.0846mmol)溶于4.0mL无水DCM和0.4mL无水MeOH的混合溶剂中,氮气保护下,滴加CH 3COOH(8.46mg,0.1410mmol)后,室温搅拌30min。然后加入NaBH 3CN(8.8mg,0.1410mmol),室温搅拌反应2h。反应结束后,用Prep-HPLC纯化得黄色固体纯产物C211(25mg,产率26.1%)。LCMS:[M+H] +=1021,1023。
3-(4-((6-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺并[5.5]十一烷-3-基)己基)氧基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C212)的制备
Figure PCTCN2020110442-appb-000178
第1步:
C153-3(100mg,0.3846mmol),K 2CO 3(79.6mg,0.5769mmol)和KI(31.9mg,0.1923mmol)分散于15.0mL无水MeCN中,加入6-溴己-1-醇(125mg,0.6923mmol),80℃反应3.0h。反应液冷却至室温后,抽滤除去固体残渣,滤液减压浓缩得粗产物,粗产物Prep-TLC纯化得浅黄色固体产物C212-1(40mg,32.0%)。LCMS:[M+H] +=361。
第2步:
C212-1(40mg,0.111mmol)溶于15.0mL无水DCM中,加入Dess-Martin试剂(94.1mg,0.222mmol),50℃回流反应2h。反应液冷却至室温,用10mL DCM稀释后,加入8.0mL饱和NaHCO 3溶液和8.0mL饱和Na 2S 2O 3溶液,搅拌5min,分出有机层用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得粗产物,继续用Prep-TLC纯化得浅黄色油状产物C212-2(20mg,产率50.4%)。LCMS:[M+H] +=359。
第3步:
C212-2(20mg,0.056mmol)和C47-1(31.5mg,0.048mmol)溶于4.0mL无水DCM和0.4mL无水MeOH的混合溶剂中,氮气保护下,滴加CH 3COOH(5.03mg,0.084mmol,溶于0.5mL DCM中)后,室温搅拌30min。然后加入NaBH 3CN(7.02mg,0.112mmol),室温搅拌反应2h。反应液直接用Prep-TLC纯化得粗产物,该粗产物继续用Prep-HPLC纯化得黄色固体纯产物C212(20mg,产率35.7%)。LCMS:[M+H] +=1007,1009。
3-(4-((5-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺并[5.5]十一烷-3-基)戊基)氧基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C213)的制备
Figure PCTCN2020110442-appb-000179
第1步:
C153-3(130mg,0.5mmol),K 2CO 3(103.5mg,0.75mmol)和KI(41.5mg,0.25mmol)分散于15.0mL无水MeCN中,加入5-溴戊-1-醇(125.3mg,0.75mmol),80℃反应3.0h。反应液冷却至室温后,抽滤除去固体残渣,滤液减压浓缩得粗产物,粗产物Prep-TLC纯化得浅黄色固体产物C213-1(65mg,37.6%)。LCMS:[M+H] +=347。
第2步:
C213-1(65mg,0.188mmol)溶于15.0mL无水DCM中,加入Dess-Martin试剂(159.4mg,0.376mmol),50℃回流反应2h。反应液冷却至室温,用20mL DCM稀释后,加入10.0mL饱和NaHCO 3溶液和10.0mL饱和Na 2S 2O 3溶液,搅拌5min,分出有机层用无水Na 2SO 4干燥,抽滤除去干燥剂,滤液减压浓缩得粗产物C213-2(57mg,87.7%),该粗产物直接用于下一步反应。LCMS:[M+H] +=345。
第3步:
C213-2(34.4mg,0.1mmol)和C47-1(39.8mg,0.06mmol)溶于4.0mL无水DCM中,氮气保护下,滴加CH 3COOH(9.0mg,0.15mmol,溶于0.5mL DCM中)后,室温搅拌30min。然后加入NaBH 3CN(12.6mg,0.2mmol),室温搅拌反应2h。用Prep-HPLC纯化得黄色固体纯产物C213(13.0mg,产率13.1%)。LCMS:[M+H] +=993,995。
3-(4-((7-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)庚基)氧基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C214)的制备
Figure PCTCN2020110442-appb-000180
C153-5(30mg,0.081mmol)和C183-1(或C158-1)(49.3mg,0.073mmol)溶于4.0mL无水DCM和0.4mL无水MeOH的混合溶剂中,氮气保护下,滴加CH 3COOH(7.29mg,0.1215mmol)后,室温搅拌30min。然后加入NaBH 3CN(7.6mg,0.1215mmol),室温搅拌反应2h后。用Prep-HPLC纯化得黄色固体纯产物C214(25mg,产率29.9%)。LCMS:[M+H] +=1036,1038。
3-(4-(4-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)丁-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C215)的制备
Figure PCTCN2020110442-appb-000181
第1步:
C215-1(或C1-1)(322mg,1.0mmol),CuI(19.0mg,0.1mmol)和Pd(dppf)Cl 2(73.1mg,0.1mmol)分散于10.0mL无水DMF中,N 2保护下,依次加入3-炔基-1-丁醇(140.2mg,2.0mmol)和TEA(303mg,3.0mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,直接RP-Flash纯化得浅黄色固体产物C215-2(210mg,67.3%)。LCMS:[M+H] +=313。
第2步:
C215-2(112mg,0.359mmol)和PPh 3(103.5mg,0.395mmol)溶解于33.0mL无水THF中,N 2保护下,加入NBS(127.8mg,0.718mmol),室温反应5.0h。反应液减压浓缩得粗产物,该粗产物用Prep-TLC纯化得白色固体产物C215-3(105mg,78.3%)。LCMS:[M+H] +=375,377。
第3步:
C215-3(45mg,0.12mmol)和A1-4(54.8mg,0.096mmol)溶于4.5mL无水DMF中,氮气保护下,加入DIPEA(75.8mg,0.6mmol),70℃反应12h。用RP-Flash纯化得白色固体产物C215(12mg,产率9.2%)。LCMS:[M+H] +=864。
3-(5-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)戊-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C218)的制备
Figure PCTCN2020110442-appb-000182
第1步:
将化合物C218-1(300mg,0.974mmol),C218-2(208mg,1.266mmol),三乙胺(128mg,1.266mmol)溶于5mL乙腈中,氮气保护下80℃搅拌过夜。冷至室温,减压除去溶剂,粗品用Flash 纯化得到白色固体化合物C218-3(180mg,产率57.0%)。LCMS:[M+H] +=323,325。
第2步:
将化合物C218-3(90mg,0.281mmol),化合物C218-4(59mg,0.699mmol),碘化亚铜(5.3mg,0.028mmol),Pd(dppf)Cl 2(20.5mg,0.028mmol)和三乙胺(85mg,0.839mmol)溶于3mL无水DMF中,氮气保护下于70℃搅拌反应5小时。减压除去溶剂,粗品薄层色谱纯化得到类白色固体化合物C218-5(83mg,产率91.2%),LCMS:[M+H] +=327。
第3步:
将化合物C218-5(50mg,0.153mmol),Dess-Martin试剂(98mg,0.230mmol)溶于10mL二氯甲烷中,于50℃搅拌2小时。降至室温,用饱和碳酸氢钠水溶液淬灭,分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,浓缩,粗品TLC纯化得到白色固体化合物C218-6(38mg,产率76.0%),LCMS:[M+H] +=325。
第4步:
将化合物C218-6(38mg,0.119mmol)和化合物A1-4(40mg,0.095mmol)溶于3mL二氯甲烷/甲醇(2/1,v/v)中,滴加1滴冰醋酸,氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(6.0mg,0.095mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C218(25mg,产率24.3%),LCMS:[M+H] +=878。 1H NMR(400MHz,DMSO-d 6)δ11.17(s,1H),10.99(s,1H),8.48(dr,1H),8.06(d,2H),7.69(dd,1H),7.63(t,1H),7.56-7.49(m,2H),7.35(dd,2H),7.13-7.06(m,1H),6.63(d,1H),6.47(dd,1H),5.32(m,1H),5.11(dd,1H),4.48-4.28(m,2H),3.76(s,3H),3.72(d,2H),2.91(m,1H),2.72-2.60(m,4H),2.57(m,2H),2.40(m,7H),2.03-1.98(m,3H),1.86(d,2H),1.78(s,3H),1.74(s,3H),1.72m,2H),1.59-1.50(m,2H),1.50-1.43(m,1H)。
3-(4-(5-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)戊-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C227)的制备
Figure PCTCN2020110442-appb-000183
C126-3(32.4mg,0.1mmol)和C183-1(或C158-1)(61.1mg,0.09mmol)溶于4.0mL无水DCM和0.4mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(9.0mg,0.15mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(12.6mg,0.2mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得黄色固体纯产物C227(20mg,产率20.3%)。LCMS:[M+H] +=988,990。
3-(4-(5-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)戊-1-炔-1-基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C228)的制备
Figure PCTCN2020110442-appb-000184
C126-3(32.4mg,0.1mmol)和C47-1(59.8mg,0.09mmol)溶于4.0mL无水DCM和0.4mL 无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(9.0mg,0.15mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(12.6mg,0.2mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得黄色固体纯产物C228(30mg,产率30.9%)。LCMS:[M+H] +=973,975。
7-(7-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-2-(2,6-二氧代哌啶-3-基)-3-氧代异吲哚啉-5-腈(C229)的制备
Figure PCTCN2020110442-appb-000185
C204-7(35mg,0.093mmol)和C183-1(或C158-1)(56.7mg,0.0837mmol)溶于4.0mL无水DCM和0.4mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(8.36mg,0.1395mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(11.7mg,0.1860mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得黄色固体纯产物C229(25mg,产率25.8%)。LCMS:[M+H] +=1041,1043。
3-(5-(4-(3-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)丙基)苯基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C235)的制备
Figure PCTCN2020110442-appb-000186
第1步:
将化合物C235-1(600mg,2.84mmol),联硼酸频那醇酯(805mg,3.17mmol),醋酸钾(825mg,8.41mmol),Pd(dppf)Cl 2(62mg,0.08mmol)溶于10mL 1,4-二氧六环中,氮气保护下于100℃搅拌过夜。减压除去溶剂,粗品Flash纯化得到化合物C235-2(550mg,产率74.9%)。
第2步:
将C218-3(或C1-1)(80mg,0.248mmol),化合物C235-2(85mg,0.323mmol),磷酸钾(65mg,0.298mmol),Pd(dppf)Cl 2(18mg,0.025mmol)溶于1.5mL DMF中,氮气保护下于90℃搅拌过夜。减压除去溶剂,粗品Flash纯化得到化合物C235-3(52mg,产率45.2%)。LCMS:[M+H] +=379。
第3步:
将化合物C235-3(78mg,0.206mmol),Dess-Martin试剂(175mg,0.413mmol)溶于10mL二氯甲烷中,于50℃搅拌2小时。降至室温,顺序加入饱和碳酸氢钠水溶液和饱和硫代硫酸钠水溶液各搅拌5分钟使反应淬灭。分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C235-4(44mg,产率57.1%),LCMS:[M+H] +=377。
第4步:
将化合物C235-4(22mg,0.058mmol)和化合物A1-4(27mg,0.047mmol)溶于2mL二氯甲烷/甲醇(2/1,v/v)中,滴加冰醋酸(3.5mg,0.058mmol),氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(2.9mg,0.047mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C235(12mg,产率22.2%),LCMS:[M+H] +=930。
3-(5-(4-(4-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)丁基)苯基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C236)的制备
Figure PCTCN2020110442-appb-000187
第1步:
将化合物C236-1(1g,4.13mmol)溶于10mL干燥的四氢呋喃中,降至-10℃氮气保护下滴加硼烷的四氢呋喃络合物(1mol/L,8.3mL,8.26mmol),滴加完毕,升至室温搅拌2小时。将反应液倾倒10mL冷水中,用乙酸乙酯萃取,无水硫酸钠干燥,过滤,浓缩,粗品Flash纯化得到化合物C236-2(600mg,产率63.7%)。
第2步:
将化合物C236-2(635mg,2.78mmol),联硼酸频那醇酯(800mg,3.15mmol),醋酸钾(820mg,8.35mmol),Pd(dppf)Cl 2(61mg,0.08mmol)溶于10mL 1,4-二氧六环中,氮气保护下于100℃搅拌过夜。减压除去溶剂,粗品Flash纯化得到化合物C236-3(500mg,产率65.1%)。
第3步:
将C218-3(或C1-1)(100mg,0.311mmol),化合物C236-3(112mg,0.404mmol),磷酸钾(81mg,0.373mmol),Pd(dppf)Cl 2(23mg,0.031mmol)溶于2mL DMF中,氮气保护下于90℃搅拌过夜。减压除去溶剂,粗品Flash纯化得到化合物C236-4(60mg,产率49.6%)。LCMS:[M+H] +=393。
第4步:
将化合物C236-4(42mg,0.107mmol),Dess-Martin试剂(91mg,0.214mmol)溶于10mL二氯甲烷中,于50℃搅拌2小时。降至室温,顺序加入饱和碳酸氢钠水溶液和饱和硫代硫酸钠水溶液各搅拌5分钟使反应淬灭。分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C236-5(39mg,产率93.3%),LCMS:[M+H] +=391。
第5步:
将化合物C236-5(39mg,0.1mmol)和化合物A1-4(40mg,0.07mmol)溶于2mL二氯甲烷/甲 醇(2/1,v/v)中,滴加冰醋酸(6.0mg,0.1mmol),氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(4.4mg,0.07mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C236(2.6mg,产率2.8%),LCMS:[M+H] +=944。
3-(4-(6-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)己基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C238)的制备
Figure PCTCN2020110442-appb-000188
将C116(100mg,0.112mmol)溶于甲醇中(10mL),加入钯/碳(30mg,10%Pd),用氮气置换三次,再用氢气置换3次,用氢气加压至60Psi,室温搅拌反应1小时。液质监测反应,原料基本转化为目标产物。氮气置换脱去氢气,过滤除去钯/碳,滤液浓缩得到粗品,用Flash分离,得到白色固体C238(49mg,产率48.8%)。LCMS:[M+H] +=896。
3-(4-(8-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)辛基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C240)的制备
Figure PCTCN2020110442-appb-000189
将C177(50mg,0.051mmol)溶于甲醇中(10mL),加入钯/碳(20mg,10%Pd),用氮气置换三次,再用氢气置换3次,用氢气加压至60Psi,室温搅拌反应1小时。液质监测反应,原料基本转化为目标产物。氮气置换脱去氢气,过滤除去钯/碳,滤液浓缩得到粗品,用Flash分离,得到白色固体C240,19mg,产率40.4%。LCMS:[M+H] +=924。
3-(4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-7-氟-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C243)的制备
Figure PCTCN2020110442-appb-000190
第1步:
C243-1(1.0g,4.05mmol)溶于20.0mL CCl 4中,氮气保护下,加入NBS(1.09g,6.08mmol)和AIBN(265.7mg,1.62mmol),升高温度至90℃,回流反应20h。TLC监测还有五分之一的原料剩余。向反应液中补加NBS(1.09g,6.08mmol)和AIBN(265.7mg,1.62mmol),补加完毕后,继续回流反应20h。TLC检测无原料剩余。反应液冷却至室温后,抽滤,减压浓缩滤液得粗产物,Flash纯化得浅黄色油状产物C243-2(1.35g,100%)。
第2步:
C243-2(1.35g,4.18mmol)和C243-3(891mg,5.43mmol)分散于25.0mL无水MeCN中,加入TEA(548.8mg,5.43mmol),升高温度至80℃,回流反应16h。LCMS检测反应完全。反应液冷却至室温后,抽滤,滤饼用MeCN淋洗三次,烘干固体得目标产物C243-4(1.25g,产率88.0%)。LCMS:[M+H] +=341,343。
第3步:
C243-4(200mg,0.588mmol),CuI(22.3mg,0.118mmol)和Pd(dppf)Cl 2(86.2mg,0.118mmol)分散于10.0mL无水DMF中,N 2保护下,依次加入C243-5(131.8mg,1.176mmol)和TEA(178.2mg,1.764mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,直接RP-Flash纯化得浅黄色固体产物C243-6(160mg,73.1%)。LCMS:[M+H] +=373。
第4步:
C243-6(160mg,0.43mmol)溶解于80.0mL无水DCM中,N 2保护下,加入Dess-Martin试剂(365mg,0.86mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入20mL饱和NaHCO 3溶液和20mL饱和Na 2S 2O 3溶液,室温搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Flash纯化得浅黄色固体产物C243-7(75mg,46.9%)。LCMS:[M+H] +=371。
第5步:
C243-7(72mg,0.195mmol)和A1-4(99.9mg,0.176mmol)溶于7.0mL无水DCM和0.7mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(17.6mg,0.293mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(24.5mg,0.390mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得白色固体纯产物C243(45mg,产率25.0%)。LCMS:[M+H] +=924。
3-(4-(6-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)己-1-炔-1-基)-7-氟-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C244)的制备
Figure PCTCN2020110442-appb-000191
第1步:
C243-4(170mg,0.5mmol),CuI(19.0mg,0.1mmol)和Pd(dppf)Cl 2(73.1mg,0.1mmol)分散于8.5mL无水DMF中,N 2保护下,依次加入C244-1(122.5mg,1.25mmol)和TEA(151.5mg,1.5mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,直接RP-Flash纯化得浅黄色固体粗产物C244-2(210mg,产率93.8%),该粗产物直接用于下一步反应。LCMS:[M+H] +=359。
第2步:
C244-2(90mg,0.251mmol)溶解于45.0mL无水DCM中,N 2保护下,加入Dess-Martin试剂(159.6mg,0.377mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入15mL饱和NaHCO 3溶液和15mL饱和Na 2S 2O 3溶液,室温搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-TLC纯化得浅黄色固体产物C244-3(75mg,83.3%)。LCMS:[M+H] +=357。
第3步:
C244-3(71.2mg,0.2mmol)和A1-4(102.4mg,0.18mmol)溶于7.0mL无水DCM和0.7mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(18.0mg,0.3mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(25.1mg,0.4mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得白色固体纯产物C244(50mg,产率27.5%)。LCMS:[M+H] +=910。
3-(4-(7-(4-(1-(4-((4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C245)的制备
Figure PCTCN2020110442-appb-000192
将化合物C1(45mg,0.05mmol)溶于甲醇中(10mL),加入钯/碳(20mg,10%Pd),用氮气置换三次,再用氢气置换3次,用氢气加压至60Psi,室温搅拌反应1小时。液质监测反应,原料基本转化为目标产物。氮气置换脱去氢气,过滤除去钯/碳,滤液浓缩得到粗品,用Flash分离,得到白色固体7mg,产率16%。LCMS:[M+H] +=876。
3-(4-((5-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)戊基)氧基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C246)的制备
Figure PCTCN2020110442-appb-000193
C213-2(22mg,0.064mmol)和C183-1(或C158-1)(30.4mg,0.045mmol)溶于3.0mL无水DCM中,氮气保护下,滴加CH 3COOH(5.8mg,0.096mmol,溶于0.5mL无水DCM中)后,室温搅拌30min。然后加入NaBH 3CN(8.03mg,0.128mmol),室温搅拌反应2h。用Prep-HPLC纯化得黄色固体纯产物C246(8.0mg,产率12.5%)。LCMS:[M+H] +=1008,1010。
7-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)戊-1-炔-1-基)-2-(2,6-二氧代哌啶-3-基)-3-氧代异吲哚啉-5-腈(C258)的制备
Figure PCTCN2020110442-appb-000194
第1步:
C204-5(173.5mg,0.5mmol),CuI(19.0mg,0.1mmol)和Pd(dppf)Cl 2(73.1mg,0.1mmol)分散于10.0mL无水DMF中,N 2保护下,依次加入C258-1(105mg,2.5mmol)和TEA(151.5mg,1.5mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,直接RP-Flash纯化得浅黄色固体产物C258-2(180mg,100%)。LCMS:[M+H] +=352。
第2步:
C258-2(180mg,0.513mmol)溶解于90.0mL无水DCM中,N 2保护下,加入Dess-Martin试剂(435mg,1.026mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入20mL饱和NaHCO 3溶液和20mL饱和Na 2S 2O 3溶液,室温搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-TLC纯化得浅黄色固体产物C258-3(105mg,58.3%)。LCMS:[M+H] +=350。
第3步:
C258-3(93mg,0.266mmol)和A1-4(129mg,0.226mmol)溶于7.0mL无水DCM和0.7mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(24mg,0.399mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(33.5mg,0.532mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得白色固体纯产物C258(30mg,产率12.5%)。LCMS:[M+H] +=903。
7-(6-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)己-1-炔-1-基)-2-(2,6-二氧代哌啶-3-基)-3-氧代异吲哚啉-5-腈(C259)的制备
Figure PCTCN2020110442-appb-000195
第1步:
C204-5(173.5mg,0.5mmol),CuI(19.0mg,0.1mmol)和Pd(dppf)Cl 2(73.1mg,0.1mmol)分散于10.0mL无水DMF中,N 2保护下,依次加入C259-1(122.5mg,2.5mmol)和TEA(151.5mg,1.5mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,直接RP-Flash纯化得浅黄色固体粗产物C259-2(210mg,100%)。LCMS:[M+H] +=366。
第2步:
C259-2(210mg,0.575mmol)溶解于105mL无水DCM中,N 2保护下,加入Dess-Martin试剂(439mg,1.035mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入20mL饱和NaHCO 3溶液和20mL饱和Na 2S 2O 3溶液,室温搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Prep-TLC纯化得浅黄色固体产物C259-3(130mg,62.5%)。LCMS:[M+H] +=364。
第3步:
C259-3(120mg,0.33mmol)和A1-4(160mg,0.28mmol)溶于8.0mL无水DCM和0.8mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(29.8mg,0.495mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(41.5mg,0.66mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得白色固体纯产物C259(30mg,产率9.93%)。LCMS:[M+H] +=917。
3-(4-(7-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)庚-1-炔-1-基)-1-氧代-6-(三氟甲基)异吲哚啉-2-基)哌啶-2,6-二酮(C267)的制备
Figure PCTCN2020110442-appb-000196
第1步:
将化合物C267-1(或C200-5)(60mg,0.154mmol),化合物C200-4(43mg,0.384mmol),碘化亚铜(2.9mg,0.015mmol),Pd(dppf)Cl 2(11.3mg,0.015mmol)和三乙胺(46.6mg,0.461mmol),溶于2mL无水DMF中,氮气保护下于70℃搅拌过夜。减压除去溶剂,粗品用薄层色谱纯化得到化合物C267-2(61mg,产率94.0%),LCMS:[M+H] +=423。
第2步:
将化合物C267-2(81mg,0.192mmol),Dess-Martin试剂(163mg,0.384mmol)溶于10mL二氯甲烷中,于50℃搅拌2小时。降至室温,顺序加入饱和碳酸氢钠水溶液和饱和硫代硫酸钠水溶液各搅拌5分钟使反应淬灭。分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C267-3(60mg,产率74.4%),LCMS:[M+H] +=421。
第3步:
将化合物267-3(20mg,0.048mmol)和化合物A1-4(22mg,0.038mmol)溶于2mL二氯甲烷/甲醇(2/1,v/v)中,滴加冰醋酸(2.9mg,0.038mmol),氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(2.4mg,0.038mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C267(12mg,产率46.3%),LCMS:[M+H] +=974。 1H NMR(400MHz,DMSO-d 6)δ11.17(s,1H),11.04(s,1H),8.48(s,1H),8.06(d,2H),7.99-7.98(m,2H),7.57-7.49(m,1H),7.38(d,1H),7.33(d,1H),7.09(t,1H),6.62(d,1H),6.46(dd,1H),5.32(dd,1H),5.18(dd,1H),4.58(d,1H),4.43(d,1H),3.76(s,3H),3.70(d,2H),2.93(m,1H),2.69-2.60(m,4H),2.52(d,2H),2.46(m,3H),2.44-2.26(m,8H),1.99(m,4H),1.82(s,1H),1.78(m,3H),1.75(s,3H),1.60(d,2H),1.46(m,3H)。
3-(4-(6-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)己-1-炔-1-基)-1-氧代-6-(三氟甲基)异吲哚啉-2-基)哌啶-2,6-二酮(C268)的制备
Figure PCTCN2020110442-appb-000197
第1步:
将化合物C268-1(或C200-5)(100mg,0.256mmol),5-炔基-1-己醇(62.9mg,0.641mmol),碘化亚铜(4.9mg,0.026mmol),Pd(dppf)Cl 2(18.8mg,0.026mmol)和三乙胺(77.7mg,0.769mmol)溶于3mL无水DMF中,氮气保护下于70℃搅拌过夜.减压除去溶剂,粗品用薄层色谱纯化得到化合物C268-2(80mg,产率76.5%),LCMS:[M+H] +=409。
第2步:
将化合物C268-2(126mg,0.309mmol),Dess-Martin试剂(262mg,0.618mmol)溶于10mL二氯甲烷中,于50℃搅拌2小时。降至室温,顺序加入饱和碳酸氢钠水溶液和饱和硫代硫酸钠水溶液各搅拌5分钟使反应淬灭。分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C268-3(40mg,产率32.0%),LCMS:[M+H] +=407。
第3步:
将化合物C268-3(39mg,0.096mmol)和化合物A1-4(38.3mg,0.067mmol)溶于2mL二氯甲烷/甲醇(2/1,v/v)中,滴加冰醋酸(5.8mg,0.096mmol),氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(4.2mg,0.067mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C268(30mg,产率92.1%),LCMS:[M+H] +=960。 1H NMR(400MHz,DMSO-d 6)δ11.17(s,1H),11.04(s,1H),8.48(s,1H),8.06(d,2H),8.01-7.98(m,2H),7.53(m,1H),7.38(d,1H),7.33(d,1H),7.09(t,1H),6.62(d,1H),6.47(dd,1H),5.32(m,1H),5.18(dd,1H),4.57(d,1H),4.42(d,1H),3.76(s,3H),3.71(d,2H),2.93(m,1H),2.70-2.62(m,3H),2.53(d,3H),2.38(m,3H),2.30(s,4H),2.05-1.96(m,4H),1.85(d,2H),1.78(s,3H),1.75(s,3H),1.63-1.58(m,4H),1.53-1.46(m,2H)。
3-(4-(5-(4-(1-(4-((5-氯-4-((2-(二甲基磷酰基)苯基)氨基)嘧啶-2-基)氨基)-3-甲氧基苯基)哌啶-4-基)哌嗪-1-基)戊-1-炔-1-基)-1-氧代-6-(三氟甲基)异吲哚啉-2-基)哌啶-2,6-二酮(C269)的制备
Figure PCTCN2020110442-appb-000198
第1步:
将化合物C269-1(或C200-5)(60mg,0.154mmol),4-炔基-1-戊醇(32.3mg,0.384mmol),碘化亚铜(2.9mg,0.015mmol),Pd(dppf)Cl 2(11.3mg,0.015mmol)和三乙胺(46.6mg,0.461mmol)溶于2mL无水DMF中,氮气保护下于70℃搅拌过夜。减压除去溶剂,粗品薄层色谱纯化得到化合物C269-2(58mg,产率95.8%),LCMS:[M+H] +=395。
第2步:
将化合物C269-2(81mg,0.206mmol),Dess-Martin试剂(174mg,0.410mmol)溶于10mL二氯甲烷中,于50℃搅拌2小时。降至室温,顺序加入饱和碳酸氢钠水溶液和饱和硫代硫酸钠水溶液各搅拌5分钟使反应淬灭。分出有机相,水相用二氯甲烷萃取三次,合并有机相,无水硫酸钠干燥,过滤,浓缩,粗品TLC纯化得到白色固体化合物C269-3(60mg,产率74.4%),LCMS:[M+H] +=393。
第3步:
将C269-3(31mg,0.079mmol)和A1-4(36mg,0.063mmol)溶于2mL二氯甲烷/甲醇(2/1,v/v)中,滴加冰醋酸(4.7mg,0.079mmol),氮气保护下室温搅拌1小时。然后加入氰基硼氢化钠(4.0mg,0.063mmol),继续搅拌1小时。40℃以下减压除去溶剂,粗品Flash纯化得到类白色固体化合物C269(13mg,产率17.4%),LCMS:[M+H] +=946。 1H NMR(400MHz,DMSO-d 6)δ11.17(s,1H),11.04(s,1H),8.48(s,1H),8.06(d,2H),8.01-7.98(m,2H),7.53(dd,1H),7.38(d,1H),7.33(d,1H),7.09(t,1H),6.63(d,1H),6.47(dd,1H),5.32(t,1H),5.19(dd,1H),4.58(d,1H),4.42(d,1H),3.76(s,3H),3.72(d,2H),2.93(m,1H),2.70-2.62(m,4H),2.54(m,2H),2.52(d,3H),2.41(m,4H),1.99(dt,5H),1.86(d,2H),1.78(s,3H),1.74(s,3H),1.55-1.45(m,3H)。
3-(4-(5-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)戊基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C285)的制备
Figure PCTCN2020110442-appb-000199
第1步:
C126-2(250mg,0.767mmol)溶于100.0mL无水MeOH中,向混合液中加入10%Pd/C(125mg),向反应液内充入H 2(0.4atm),室温反应2h。LCMS检测反应完全。反应液抽滤除去固体催化剂,滤液减压除去溶剂得白色固体粗产物C285-1(230mg,92%)。LCMS:[M+H] +=331。
第2步:
C285-1(230mg,0.697mmol)溶解于150mL无水DCM中,N 2保护下,加入Dess-Martin试剂(591mg,1.394mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入30mL饱和NaHCO 3溶液和30mL饱和Na 2S 2O 3溶液,室温搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Flash纯化得白色固体产物C285-2(210mg,91.3%)。LCMS:[M+H] +=329。
第3步:
C285-2(49.2mg,0.15mmol)和C158-1(81.5mg,0.12mmol)溶于5.0mL无水DCM和0.5mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(13.5mg,0.225mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(18.8mg,0.3mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得黄色固体纯产物C285(38mg,产率25.6%)。LCMS:[M+H] +=992,994。
3-(4-(6-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)己基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C286)的制备
Figure PCTCN2020110442-appb-000200
第1步:
C116-2(150mg,0.441mmol)溶于60.0mL无水MeOH中,向混合液中加入10%Pd/C(150mg),向反应液内充入H 2(0.4atm),室温反应2h。LCMS检测反应完全。反应液抽滤除去固体催化剂,滤液减压除去溶剂得白色固体粗产物C286-1(150mg,98.5%),直接用于下一步反应。LCMS:[M+H] +=345。
第2步:
C286-1(150mg,0.436mmol)溶解于100mL无水DCM中,N 2保护下,加入Dess-Martin试剂(370mg,0.872mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入20mL饱和NaHCO 3溶液和20mL饱和Na 2S 2O 3溶液,室温搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Flash纯化得白色固体产物C286-2(100mg,66.7%)。LCMS:[M+H] +=343。
第3步:
C286-2(41mg,0.12mmol)和C158-1(65.2mg,0.096mmol)溶于4.0mL无水DCM和0.4mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(10.8mg,0.18mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(15.1mg,0.24mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得黄色固体纯产物C286(20mg,产率16.6%)。LCMS:[M+H] +=1006,1008。
3-(4-(6-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺[5.5]十一烷-3-基)己基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C299)的制备
Figure PCTCN2020110442-appb-000201
C187(20mg,0.02mmol)溶于20.0mL MeOH中,加入10%Pd/C(10mg),向反应体系内充入0.4atm H 2,并室温反应2h。LCMS检测反应完全。反应液过滤除去固体催化剂,滤液浓缩后得粗产物,直接RP-Flash纯化得浅黄色固体粗产物,该粗产物继续用Prep-HPLC纯化得白色固体产物C299(2.5mg,产率12.6%)。LCMS:[M+H] +=991,993。
3-(4-(4-(4-(1-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)哌啶-4-基)哌嗪-1-基)丁基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C301)的制备
Figure PCTCN2020110442-appb-000202
第1步:
C301-1(644mg,2.0mmol),CuI(38mg,0.2mmol)和Pd(dppf)Cl 2(146.2mg,0.2mmol)分散于20.0mL无水DMF中,N 2保护下,依次加入C301-2(280.4mg,4.0mmol)和TEA(606mg,6.0mmol),升高温度至70℃反应20h。LCMS检测反应完全。反应液冷却至室温,RP-Flash纯化得浅黄色固体粗产物C301-3(450mg,72.1%)。LCMS:[M+H] +=313。
第2步:
C301-3(450mg,1.44mmol)溶于90.0mL无水MeOH中,向混合液中加入10%Pd/C(225mg),向反应液内充入H 2(0.4atm),室温反应2h。LCMS检测反应完全。反应液抽滤除去固体催化剂,滤液减压除去溶剂得白色固体粗产物C301-4(450mg,98.9%),该粗产物直接用于下一步反应。LCMS:[M+H] +=317。
第3步:
C301-4(420mg,1.33mmol)溶解于150mL无水DCM中,N 2保护下,加入Dess-Martin试剂(1010mg,2.39mmol),升高温度至50℃,回流反应2.0h。TLC检测反应完全。反应液冷却至室温后,向反应液中加入30mL饱和NaHCO 3溶液和30mL饱和Na 2S 2O 3溶液,室温搅拌5min,分出有机层后用无水硫酸钠干燥,抽滤,滤液减压浓缩得粗产物。该粗产物用Flash纯化得白色固体产物C301-5(350mg,83.3%)。LCMS:[M+H] +=315。
第4步:
C301-5(47.1mg,0.15mmol)和C158-1(81.5mg,0.12mmol)溶于5.0mL无水DCM和0.5mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(13.5mg,0.225mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(18.8mg,0.3mmol),室温继续反应2h。LCMS和TLC检测反应完全。用RP-Flash纯化得黄色固体纯产物C301(35mg,产率23.8%)。LCMS:[M+H] +=978,980。
3-(4-(4-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺并[5.5]十一烷-3-基)丁基)-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C302)的制备
Figure PCTCN2020110442-appb-000203
C301-5(47.1mg,0.15mmol)和C47-1(79.7mg,0.12mmol)溶于5.0mL无水DCM和0.5mL无水MeOH的混合溶剂中,氮气保护下,加入CH 3COOH(13.5mg,0.225mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(18.8mg,0.3mmol),室温继续反应2h。LCMS和TLC检测反应完全。反应液直接用Prep-TLC纯化得粗产物,该粗产物继续用RP-Flash纯化得黄色固体纯产物C302(35mg,产率24.3%)。LCMS:[M+H] +=963,965。
3-(4-(3-(2-(9-(4-((5-溴-4-((5-(二甲基磷酰基)喹喔啉-6-基)氨基)嘧啶-2-基)氨基)-5-甲氧基-2-甲基苯基)-3,9-二氮杂螺并[5.5]十一烷-3-基)乙氧基)丙-1-炔-1-基)-6-氟-1-氧代异吲哚啉-2-基)哌啶-2,6-二酮(C310)的制备
Figure PCTCN2020110442-appb-000204
C184-3(40mg,0.112mmol)和C47-1(55.6mg,0.084mmol)溶于5.0mL无水DCM和0.5mL无水甲醇的混合溶剂中,氮气保护下,加入CH 3COOH(10.08mg,0.168mmol),室温搅拌0.5h。然后向反应液中加入固体NaBH 3CN(14.1mg,0.224mmol),室温继续反应2h。LCMS和TLC检测反应完全。用Prep-HPLC纯化得黄色固体纯产物C310(21mg,产率18.8%)。LCMS:[M+H] +=1007,1009。 1H-NMR:(400MHz,DMSO-d 6)δ11.026(s,1H),8.865(dd,J=2.0Hz,9.6Hz,3H),8.265(s,2H),7.938(d,J=9.6Hz,1H),7.676-7.611(m,3H),7.324(s,1H),6.810(s,1H),5.187(dd,J=5.2Hz,13.2Hz,1H),4.470(s,3H),4.360(d,J=17.6Hz,1H),3.782(s,3H),3.677(t,J=5.6Hz,2H),2.991-2.913(m,1H),2.8891-2.784(m,4H),2.681-2.503(m,4H),2.460-2.426(m,5H),2.076(s,3H),2.040(s,3H),2.004(s,3H),1.561-1.521(m,8H)。
EGFR抑制活性测试
1.实验方法
①配制化合物储备液,然后进行3x稀释得到化合物稀释液;用Echo 550取10nl化合物稀释液转移到384孔板(784075,Greiner)内;
②封闭板子,1,000g离心1min;
③用1x Kinase buffer分别制备2x EGFR Del19/T790M/C797S和EGFR L858R/T790M/C797S蛋白工作液;
④将5μl 2x EGFR蛋白工作液加入步骤②的384孔板中,1,000g离心30s,室温静置(充分混合)10min;
⑤用1x Kinase buffer制备2x TK-substrate-biotin(2μM)和ATP混合液;
⑥加入5μl TK-substrate-biotin和ATP(步骤⑤制备的混合液)至步骤④的384孔板中,起始反应;
⑦1,000g离心30s,封闭板子,室温静置(反应)40min;
⑧用detection buffer制备4X Sa-XL 665和TK-antibody-Cryptate;
⑨依次加入5μl Sa-XL 665和5μl TK-antibody-Cryptate至步骤⑦的384孔板中;
⑩1,000g离心30s,室温静置(反应)1h;
Figure PCTCN2020110442-appb-000205
酶标仪(PerkinElmer,74785)读取615nm和665nm的荧光。
2.数据分析
计算每个孔的Ratio(665/615)。
抑制率(%)公式:
Figure PCTCN2020110442-appb-000206
Ratio cmpd:测试化合物的Ratio(665/615)值。
Figure PCTCN2020110442-appb-000207
阳性对照的平均Ratio(665/615)值。
Figure PCTCN2020110442-appb-000208
阴性对照的平均Ratio(665/615)值。
用GraphPad Prism 8.0拟合抑制率(%)数值和化合物浓度的对数的非线性回归曲线(剂量响应-可变斜率),绘制化合物的效应-剂量曲线,计算IC 50值。
3.实验结果
本发明的化合物对突变型EGFR Del19/T790M/C797S和EGFR L858R/T790M/C797S的抑制活性IC 50值如下表所示。
Figure PCTCN2020110442-appb-000209
#注:A表示IC 50数值范围为1-100nM,B表示IC 50数值范围为101-200nM,C表示IC 50数值范围为201-300nM,D表示IC 50数值范围为>300nM。
结论:本发明的化合物对突变型EGFR Del19/T790M/C797S和EGFR L858R/T790M/C797S具有较强的抑制活性。
Ba/F3(EGFR Del19/T790M/C797S)和Ba/F3(EGFR L858R/T790M/C797S)细胞活性测试
1.实验方法
①Ba/F3(EGFR Del19/T790M/C797S)和Ba/F3(EGFR L858R/T790M/C797S)细胞分别按照ATCC的要求培养,37℃,5%CO 2条件下用孵箱孵育,细胞按照指数进行分析;细胞活力>90%可以用于实验,细胞以700个细胞/孔,30μl/孔接种在384孔板(PerkinElmer,6007680)上。
②配制化合物储备液,然后进行3x稀释得到化合物稀释液,用Echo(Labcyte,Echo550)取30nl化合物稀释液加入384孔板,细胞在37℃,5%CO 2条件下用孵箱孵育72小时。
③每孔加30μl CTG,在Plate shaker(QILINBEIER,QB-9002)上震荡384孔板,384孔板避光,37℃,5%CO 2条件下孵育30min,用Envision(PerkinElmer,EnVision 2104)读取化学发光数值。
2.数据分析
百分抑制率(%inhibition)用下面的公式计算
Figure PCTCN2020110442-appb-000210
LUM cmpd:测试化合物的发光值。
Figure PCTCN2020110442-appb-000211
浓度为10μM的阳性药平均LUM值。
Figure PCTCN2020110442-appb-000212
未加药物处理的阴性对照组平均LUM值。
用GraphPad Prism 8.0拟合抑制率(%)数值和化合物浓度的对数的非线性回归曲线(剂量响应-可变斜率),绘制化合物的效应-剂量曲线,计算IC 50值。
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50-X)*HillSlope))
X轴:化合物浓度的log对数;Y轴:抑制率(%inhibition)。
3.实验结果
本发明的化合物对Ba/F3(EGFR Del19/T790M/C797S)和Ba/F3(EGFR L858R/T790M/C797S)细胞的抑制活性IC 50值如下表所示。
Figure PCTCN2020110442-appb-000213
Figure PCTCN2020110442-appb-000214
Figure PCTCN2020110442-appb-000215
#注:A表示IC 50数值范围为1-100nM,B表示IC 50数值范围为101-200nM,C表示IC 50数值范围为201-300nM,D表示IC 50数值范围为>300nM。
结论:本发明的化合物对突变型Ba/F3(EGFR Del19/T790M/C797S)和Ba/F3(EGFR L858R/T790M/C797S)细胞具有较强的抑制活性。
化合物诱导EGFR L858R/T790M/C797S和EGFR Del19/T790M/C797S蛋白降解活性测试
为了进一步解释本发明的化合物对Ba/F3(EGFR L858R/T790M/C797S)和Ba/F3(EGFR Del19/T790M/C797S)细胞产生抑制活性的原因,选取代表性的化合物C176和C213,对化合物的作用机制进行研究,观察其对EGFR L858R/T790M/C797S和EGFR Del19/T790M/C797S蛋白水平的影响。
(1)细胞培养:
Ba/F3(EGFR L858R/T790M/C797S)和Ba/F3(EGFR Del19/T790M/C797S)细胞分别按照ATCC推荐的培养条件培养,并以指数分析。
完全培养基:1640培养基,10%FBS,1x谷氨酰胺,1x青霉素-链霉素。
培养条件:37℃,95%空气,5%CO 2孵箱中孵育。
(2)化合物储备液:10mM DMSO储备液,-20℃储存。
(3)细胞悬液制备:
收集细胞培养瓶中的细胞,细胞活力>90%可以用于实验。40μL细胞,1*10 5细胞/孔种在96孔板中。
(4)化合物处理:
用DMSO稀释化合物,化合物起始浓度分别为1.0mM(EGFR L858R/T790M/C797S实验)和5mM(EGFR Del19/T790M/C797S实验),3x稀释,10个浓度梯度,制备工作液。
(5)移取化合物加入96孔板处理细胞,在37℃,95%空气,5%CO 2孵箱中处理24小时。
(6)检测:
1)1μg/ml EGF激动细胞,处理10min;
2)化合物处理完成后,加lysis buffer裂解细胞;转移10μL细胞裂解液到384孔板,同时添加control lysate、negative control加5μL acceptor mix至384孔板,摇床上振摇1-2min;
3)每孔分别加5μL donor mix,封闭384孔板,摇床上振摇1-2min,室温避光过夜,用酶标仪读数。
(7)数据分析:
用Graphpad Prism 8.0,通过将化合物浓度对数处理、拟合Alpha Counts。
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50-X)*HillSlope))
X:化合物浓度的log对数;Y:Alpha Counts.
(8)实验结果:
本发明的化合物对突变型EGFR L858R/T790M/C797S和EGFR Del19/T790M/C797S蛋白水平的影响如图1和2所示。
如图1所示,实验结果表明,在0.01nM至1000nM范围内,随着化合物C176浓度的增加,EGFR L858R/T790M/C797S蛋白的水平随之降低,化合物C176显著地降低了EGFR L858R/T790M/C797S蛋白的水平,其DC 50为31.37nM,证明本发明的化合物对EGFR L858R/T790M/C797S蛋白具有显著的降解作用,并且具有剂量依赖性。
如图2所示,实验结果表明,在0.1nM至10000nM范围内,随着化合物C213浓度的增加,EGFR Del19/T790M/C797S蛋白的水平随之降低,化合物C213显著地降低了EGFR Del19/T790M/C797S蛋白的水平,其DC 50为11.28nM,证明本发明的化合物对EGFR Del19/T790M/C797S蛋白具有显著的降解作用,并且具有剂量依赖性。
结论:本发明的化合物在细胞内能够显著地诱导EGFR Del19/T790M/C797S和EGFR L858R/T790M/C797S蛋白的降解,并且具有剂量依赖性。
ALK激酶活性测试
1.实验方法
①配制化合物储备液;
②将化合物储备液进行3x稀释得化合物稀释液;
③用Echo 550转移100nl化合物稀释液至ProxiPlate-384 Plus(Perkin Elmer,Perkin Elmer),双复孔;
④用1x Kinase buffer分别制备2x ALK和ALK G1202R激酶工作液;
⑤将5μl 2x激酶工作液加入反应板中,1,000rpm离心1min,25℃孵育15min;
⑥用1x Kinase buffer制备2x TK-substrate-biotin(2μM)和ATP混合液;
⑦加入5μl STK-substrate-biotin和ATP(步骤⑥制备的混合液)起始反应;
⑧1,000g离心30S,封闭板子,25℃孵育60min;
⑨用detection buffer制备4X Sa-XL 665和TK-antibody-Cryptate;
⑩依次加入5μl Sa-XL 665和5μl TK-antibody-Cryptate;
Figure PCTCN2020110442-appb-000216
1,000g离心1min,25℃孵育60min;
Figure PCTCN2020110442-appb-000217
酶标仪(PerkinElmer,74785)读取615nm和665nm的荧光。
2.数据分析
计算每个孔Ratio(665/615)值。
抑制率(%)公式:
Figure PCTCN2020110442-appb-000218
Ratio cmpd:测试化合物的Ratio(665/615)值。
Figure PCTCN2020110442-appb-000219
阳性对照的平均Ratio(665/615)值。
Figure PCTCN2020110442-appb-000220
阴性对照的平均Ratio(665/615)值。
用GraphPad Prism 8.0拟合抑制率(%)数值和化合物浓度的对数做非线性回归曲线(剂量响 应-可变斜率),绘制化合物的效应-剂量曲线,计算IC 50值。
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50-X)*HillSlope))
X轴:化合物浓度的对数;Y轴:抑制率(%inhibition)。
3.实验结果
本发明的化合物对ALK和ALK G1202R抑制活性IC 50值如下表所示。
Figure PCTCN2020110442-appb-000221
#注:A表示IC 50数值范围为1-100nM,B表示IC 50数值范围为101-200nM,C表示IC 50数值范围为201-300nM,D表示IC 50数值范围为>300nM。
结论:本发明的化合物对ALK和ALK G1202R具有较强的抑制活性。
Ba/F3(EML4-ALK)细胞活性测试
1.实验方法
①Ba/F3(EML4-ALK)细胞按照ATCC的要求培养,37℃,5%CO 2条件下用孵箱孵育,细胞按照指数进行分析;细胞活力>90%可以用于实验,细胞以700个细胞/孔,30μl/孔接种在384孔板(PerkinElmer,6007680)上。
②配制化合物储备液,进行3x稀释得到化合物稀释液,用Echo(Labcyte,Echo550)将30nl梯度稀释液加入384孔板,细胞在37℃,5%CO 2条件下用孵箱孵育72小时。
③每孔加30μl CTG,在Plate shaker(QILINBEIER,QB-9002)上震荡384孔板,384孔板避光,37℃,5%CO 2条件下孵育30min,用Envision(PerkinElmer,EnVision 2104)读取化学发光数值。
2.数据分析
百分抑制率(%inhibition)用下面的公式计算
Figure PCTCN2020110442-appb-000222
LUM cmpd:测试化合物的发光值。
Figure PCTCN2020110442-appb-000223
浓度为10μM的阳性药平均LUM值。
Figure PCTCN2020110442-appb-000224
未加药物处理的阴性对照组平均LUM值。
用GraphPad Prism 8.0拟合抑制率(%)数值和化合物浓度的对数的非线性回归曲线(剂量响应-可变斜率),绘制化合物的效应-剂量曲线,计算IC 50值。
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50-X)*HillSlope))
X轴:化合物浓度的log对数;Y轴:抑制率(%inhibition)。
3.实验结果
本发明的化合物对Ba/F3(EML4-ALK)细胞的抑制活性IC 50值如下表所示。
Figure PCTCN2020110442-appb-000225
#注:A表示IC 50数值范围为1-100nM,B表示IC 50数值范围为101-200nM,C表示IC 50数值范围为201-300nM,D表示IC 50数值范围为>300nM。
结论:本发明的化合物对Ba/F3(EML4-ALK)细胞具有较强的抑制活性。
以上内容是结合具体的优选实施方式对本发明所作的进一步详细说明,不能认定本发明的具体实施只局限于这些说明。对于本发明所属技术领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干简单推演或替换,都应当视为属于本发明的保护范围。

Claims (33)

  1. 通式(X)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物:
    Figure PCTCN2020110442-appb-100001
    其中,
    环A选自任选取代的以下基团:C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-10元杂芳基,其中所述取代基选自卤素、-CN、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 3-7环烷基、4-8元杂环基、-P(O)(C 1-6烷基) 2、-P(O)(C 2-6烯基) 2、-O-C 1-6烷基、-O-C 1-6卤代烷基、-O-C 2-6烯基、-O-C 3-7环烷基、-O-4-8元杂环基、-NH-C 1-6烷基、-NH-C 2-6烯基、-NH-C 3-7环烷基、-NH-4-8元杂环基、-C(O)-C 1-6烷基、-C(O)-C 2-6烯基、-C(O)-C 3-7环烷基、-C(O)-4-8元杂环基、-S(O) 2-C 1-6烷基、-S(O) 2-C 2-6烯基、-S(O) 2-C 3-7环烷基、-S(O) 2-4-8元杂环基、-NHC(O)-C 1-6烷基、-NHC(O)-C 2-6烯基、-NHC(O)-C 3-7环烷基、-NHC(O)-4-8元杂环基、-NHS(O) 2-C 1-6烷基、-NHS(O) 2-C 2-6烯基、-NHS(O) 2-C 3-7环烷基或-NHS(O) 2-4-8元杂环基;
    环B选自以下基团:
    Figure PCTCN2020110442-appb-100002
    Figure PCTCN2020110442-appb-100003
    表示单键或双键;
    Figure PCTCN2020110442-appb-100004
    表示与分子其他部分的连接位置可以位于所在环的可用位置;
    Z 1为O、S、N或C原子,其任选地被一个或两个R Z1取代;或者Z 1不存在,因而Z 4连接到Z 2、Z 3或与Z 1相连的芳环上的C原子,并且与Z 1相连的Z 2和芳环上的C原子分别与R W相连;或者Z 1、Z 2和Z 3都不存在,因而Z 4连接到与Z 1或Z 3相连的芳环上的C原子之一,并且芳环上的另一个C原子与R W相连;
    Z 2为O、S、N或C原子,其任选地被一个或两个R Z2取代;
    Z 3为O、S、N或C原子,其任选地被一个或两个R Z3取代;条件是,当
    Figure PCTCN2020110442-appb-100005
    为双键时,Z 2为N或C原子,Z 3为N或C原子;
    Z 4为N或CR Z4
    Z 5为N或CR Z5
    R a、R b和R c独立地为H、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基或C 1-6卤代烷基;或者R a、R b以及它们连接的C原子一起形成C=O、C 3-7环烷基或4-8元杂环基;或者R a和R c以及它们连接的C原子一起形成C 3-7环烷基或4-8元杂环基;或者R a和R c形成化学键;
    R N1为H、C 1-6烷基或C 1-6卤代烷基,优选H;
    R Z1为不存在、H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;或者两个R Z1与Z 1一起形成C=O、C 3-7环烷基或 4-8元杂环基;
    R Z2为不存在、H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;或者两个R Z2与Z 2一起形成C=O、C 3-7环烷基或4-8元杂环基;
    R Z3为不存在、H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;或者两个R Z3与Z 3一起形成C=O、C 3-7环烷基或4-8元杂环基;
    R Z4为H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基或C 1-6卤代烷基;
    R Z5为H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基或-(CH 2) 0-5-4-8元杂环基;
    或者Z 4所在的环不存在;
    其中R w为H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、-C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-C 3-7环烷基、-(CH 2) 0-5-4-8元杂环基、C 2-6烯基、C 2-6炔基、-(CH 2) 0-5-C 3-10卤代环烷基、-(CH 2) 0-5-C 6-10芳基或-(CH 2) 0-5-5-14元杂芳基;
    R”为H、C 1-6烷基、C 1-6卤代烷基或-(CH 2) 0-5-C 3-7环烷基;
    R”’为H、C 1-6烷基或C 1-6卤代烷基;
    L 1选自化学键、-O-、-S(O) p-、-S(O)(=NR *)-、-NR #-、-CR #R #’-、-C aR #R #’-C bR #R #’-、-N=S(O)(R *)-或-S(O)(R *)=N-;
    L 2选自化学键、-O-、-S(O) p-、-S(O)(=NR *)-、-NR #-、-CR #R #’-、-C aR #R #’-C bR #R #’-、-N=S(O)(R *)-或-S(O)(R *)=N-;
    其中C aR #R #’或C bR #R #’中任一个可被O、S(O) p、S(O)(=NR *)或NR #替换,并且当C aR #R #’或C bR #R #’中任一个被O、S或NR #替换时,C aR #R #’或C bR #R #’中另一个还可被S(O) q替换;
    E独立地选自:化学键、-C cR #R #’-C dR #R #’-C eR #R #’、
    Figure PCTCN2020110442-appb-100006
    Figure PCTCN2020110442-appb-100007
    其中C cR #R #’、C dR #R #’或C eR #R #’中任一个,或者C cR #R #’和C eR #R #’二者可被O、S(O) p、S(O)(=NR *)或NR #替换,并且当C cR #R #’、C dR #R #’或C eR #R #’中任一个被O、S或NR #替换时,与之相邻的另一个或两个C cR #R #’、C dR #R #’或C eR #R #’还可被S(O) q替换;
    或者两个E单元可以形成-CH 2CH 2OCH 2CH 2-、-OCH 2CH 2CH 2CH 2-、-CH 2CH 2CH 2CH 2O-、
    Figure PCTCN2020110442-appb-100008
    其中,
    Figure PCTCN2020110442-appb-100009
    表示与L 1或L 2的连接点;
    H 1和H 2为N或C原子,H 3为O、S、N或C原子,并且H 1和H 3、H 2和H 3不同时为杂原子;
    H 4和H 5为N或C原子;
    H 6、H 7、H 8和H 9为C或N原子;
    p为0、1或2;
    q为1或2;
    R *为H、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10 元杂环基、C 6-10芳基或5-14元杂芳基;
    R #为H、卤素、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基或5-14元杂芳基;
    R #’为H、卤素、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基或5-14元杂芳基;
    或者,相邻原子上的R #和R #可以形成化学键,相邻原子上的R #’和R #’可以形成化学键;
    或者,相同或不同原子上的R #和R #’可以一起形成=O、或任选被R x取代的C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-6元杂芳基,其中所述R x为H、CN、卤素、C 1-6烷基或C 1-6卤代烷基;
    m为0、1、2、3、4、5、6、7、8、9或10;
    R s1选自H、CN、卤素、-(CH 2) 0-5-OR”、-(CH 2) 0-5-NR”R”’、C 1-6烷基、C 1-6卤代烷基、-(CH 2) 0-5-4-8元杂环基、C 2-6烯基、C 2-6炔基、-(CH 2) 0-5-C 3-7环烷基、-(CH 2) 0-5-C 3-10卤代环烷基、-(CH 2) 0-5-C 6-10芳基、-(CH 2) 0-5-5-14元杂芳基、-C(O)R W、-S(O)R W或-S(O) 2R W
    s1为0、1、2或3;
    R’选自H、-C(O)-C 1-6烷基、-C(O)-C 1-6卤代烷基、-C(O)-C 2-6烯基或-C(O)-C 6-10芳基;
    L为化学键、-O-或-NR-;
    其中R为H或C 1-6烷基;
    Z为-N=或-C(R 4)=;
    T选自化学键、C 2-6亚杂烷基、4-12元亚杂环基或5-6元亚杂环基取代的5-6元亚杂环基;
    R 1选自H、卤素、氰基、C 1-6烷基、C 3-7环烷基、4-8元杂环基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    或者,当L为-NR-时,R 1、R与它们连接的原子一起形成任选取代的5-6元杂环基,其中所述取代基选自卤素、氧代、C 1-6烷基、C 1-6卤代烷基、卤素单取代或多取代的C 6-10芳基;
    R 2为H、卤素、羟基、氨基、C 1-6烷基或C 1-6卤代烷基;
    或者R 1、R 2与它们连接的原子一起形成5-6元杂环基或5-6元杂芳基;
    R 3选自H、-O-C 1-6烷基或-O-C 1-6卤代烷基;
    R 4选自H、卤素、C 1-6烷基、C 1-6卤代烷基、-NHC(O)-C 1-6烷基或-NHC(O)-C 2-6烯基;
    当上述各基团为H或含H的基团时,所述一个或多个H原子可以被D原子取代;
    上述烷基、亚烷基、卤代烷基、烯基、炔基、环烷基、卤代环烷基、杂环基、芳基、杂芳基或L 1、E、L 2、和T中含有OH、NH、NH 2、CH、CH 2、CH 3的基团在每次出现时各自任选地被1、2、3或更多个R s及其同位素变体取代,其中所述R s在每次出现时独立地选自:卤素、羟基、氨基、氰基、硝基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、C 3-10环烷基、C 3-10卤代环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基、C 6-12芳烷基、-OR a’、-OC(O)R a’、-C(O)R a’、-C(O)OR a’、-C(O)NR a’R b’、-S(O) nR a’、-S(O) nOR a’、-S(O) nNR a’R b’、-NR a’R b’、-NR a’C(O)R b’、-NR a’-C(O)OR b’、-NR a’-S(O) n-R b’、-NR a’C(O)NR a’R b’、-C 1-6亚烷基-R a’、-C 1-6亚烷基-OR a’、-C 1-6亚烷基-OC(O)R a’、-C 1-6亚烷基-C(O)OR a’、-C 1-6亚烷基-S(O) nR a’、-C 1-6亚烷基-S(O) nOR a’、-C 1-6亚烷基-OC(O)NR a’R b’、-C 1-6亚烷基-C(O)NR a’R b’、-C 1-6亚烷基-NR a’-C(O)NR a’R b’、-C 1-6亚烷基-OS(O) nR a’、-C 1-6亚烷基-S(O) nNR a’R b’、-C 1-6亚烷基-NR a’-S(O) nNR a’R b’、-C 1-6亚烷基-NR a’R b’和-O-C 1-6亚烷基-NR a’R b’,并且其中关于取代基R s所述的羟基、氨基、烷基、亚烷基、环烷基、杂环基、芳基、杂芳基和芳烷基进一步任选地被1、2、3或更多个独立地选自下列的取代基及其同位素变体取代:卤素、OH、氨基、氰基、硝基、C 1-6烷基、C 1-6卤代烷基、C 1-6烷基羟基、C 3-6环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基;
    n每次出现时各自独立地为1或2;
    R a’和R b’在每次出现时各自独立地选自H、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷基-O-、C 1-6烷基-S-、C 3-10环烷基、3-10元杂环基、C 6-10芳基、5-14元杂芳基和C 6-12芳烷基。
  2. 权利要求1的通式(X)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其为通式(I)化合物:
    Figure PCTCN2020110442-appb-100010
    其中,各基团如权利要求1所定义。
  3. 权利要求2的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    环A为以下基团:
    Figure PCTCN2020110442-appb-100011
    其中,
    R 7选自C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 3-7环烷基、-P(O)(C 1-6烷基) 2或-P(O)(C 2-6烯基) 2
    R 8选自H、-NH-C 1-6烷基、-NH-C 2-6烯基、-NH-C 3-7环烷基、-NH-4-8元杂环基、-NHC(O)-C 1-6烷基、-NHC(O)-C 2-6烯基、-NHC(O)-C 3-7环烷基、-NHC(O)-4-8元杂环基、-NHS(O) 2-C 1-6烷基或-NHS(O) 2-C 3-7环烷基;
    R 9选自H、卤素、C 1-6烷基、-CN、C 1-6卤代烷基、-O-C 1-6烷基、-O-C 1-6卤代烷基、C 3-7环烷基、-O-C 3-7环烷基、-NH-C 1-6烷基、-NH-C 2-6烯基、-NH-C 3-7环烷基、-NH-4-8元杂环基、-NHC(O)-C 1-6烷基、-NHC(O)-C 2-6烯基、-NHC(O)-C 3-7环烷基、-NHC(O)-4-8元杂环基、-NHS(O) 2-C 1-6烷基或-NHS(O) 2-C 3-7环烷基;
    X为-C(R x)=或-N=;
    其中,R x为H,或者R x连同R 8以及它们所连接的C原子一起形成5-6元杂环基或5-6元杂芳基;优选地,R x连同R 8以及它们所连接的C原子一起形成5-6元杂芳基,优选吡嗪基;
    X 1为-CH(R X1)-或-N(R X1)-;
    X 2为-CH(R X2)-或-N(R X2)-;
    其中,R X1选自H、C 1-6烷基、C 3-7环烷基、-O-C 1-6烷基、-O-C 3-7环烷基、-NH-C 1-6烷基、-NH-C 3-7环烷基、-S(O) 2-C 1-6烷基、-S(O) 2-C 3-7环烷基、-NHS(O) 2-C 1-6烷基、-NHS(O) 2-C 3-7环烷基、-C(O)-C 1-6烷基、-C(O)-C 2-6烯基、-C(O)-C 3-7环烷基、-NHC(O)-C 1-6烷基、-NHC(O)-C 3-7环烷基或-C(O)-4-8元杂环基;R X2选自H、C 1-6烷基、C 3-7环烷基、-O-C 1-6烷基、-O-C 3-7环烷基、-NH-C 1-6烷基、-NH-C 3-7环烷基、-C(O)-C 1-6烷基、-C(O)-C 3-7环烷基、-C(O)-4-8元杂环基、-S(O) 2-C 1-6烷基、-S(O) 2-C 3-7环烷基、-NHS(O) 2-C 1-6烷基、-NHS(O) 2-C 3-7环烷基、-NHC(O)-C 1-6烷基、-NHC(O)-C 2-6烯基、-NHC(O)-C 3-7环烷基或-NHC(O)-4-8元杂环基;
    Figure PCTCN2020110442-appb-100012
    表示与L的连接点。
  4. 权利要求2或3的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    T为
    化学键、
    Figure PCTCN2020110442-appb-100013
    其中,
    R 5为H或C 1-6烷基;
    R 6为H或C 1-6烷基;
    或者R 5和R 6连接形成C 1-6亚烷基;
    Figure PCTCN2020110442-appb-100014
    表示与母核或L 2的连接点。
  5. 权利要求2-4中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    当L为-NR-时,R 1和R一起形成以下基团:-C(O)N(R N)C(O)-或-C(C 1-6烷基)=C(R N)C(O)-;
    其中,
    R N选自C 1-6烷基或
    Figure PCTCN2020110442-appb-100015
    R 11为H或卤素;
    R 12为H或卤素;
    R 13为H或卤素;
    Figure PCTCN2020110442-appb-100016
    表示连接点。
  6. 权利要求2-5中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,R 1、R 2与它们连接的原子一起形成5-6元杂芳基;优选形成吡咯基。
  7. 权利要求2的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,
    环A选自任选取代的以下基团:C 3-7环烷基、4-8元杂环基、C 6-10芳基或5-10元杂芳基,其中所述取代基选自-F、-Cl、-Br、-Me、-OMe、-CF 3、-OCF 3、-CN、-NHMe、环丙基、-P(O)Me 2、-NHC(O)CH 2CH 3、-C(O)CH=CH 2、-NHS(O) 2CH 2CH 3、-NH-环丙基、-NHC(O)CH=CH 2或-C(O)CH 2CH 3;环A优选为以下基团:
    Figure PCTCN2020110442-appb-100017
    其中,
    R 7选自-Me、环丙基或-P(O)Me 2
    R 8选自H、-NHMe、-NHC(O)CH 2CH 3或-NH-环丙基;
    R 9选自H、-F、-Cl、-Br、-Me、-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH 2CH 3、-NHS(O) 2CH 2CH 3、 -NHC(O)CH=CH 2或-NH-环丙基;
    X为-C(R x)=或-N=;
    其中,R x为H,或者R x连同R 8以及它们所连接的C原子一起形成5-6元杂环基或5-6元杂芳基;优选地为5-6元杂芳基(优选吡嗪基);
    X 1为-CH 2-或-N(R X1)-;
    X 2为-CH(R X2)-或-N(R X2)-;
    其中,R X1为-C(O)CH 2CH 3或-C(O)CH=CH 2;R X2为H、-Me、-OMe、-NHMe、-C(O)CH 2CH 3、-NHS(O) 2CH 2CH 3或-NHC(O)CH 2CH 3
    Figure PCTCN2020110442-appb-100018
    表示与L的连接点;
    T选自化学键、C 2-6亚杂烷基、4-12元亚杂环基或5-6元亚杂环基取代的5-6元亚杂环基;优选为以下基团:
    Figure PCTCN2020110442-appb-100019
    其中,
    R 5为-Me;
    R 6为-Me;
    或者R 5和R 6连接形成-CH 2CH 2-;
    Figure PCTCN2020110442-appb-100020
    表示与母核或L 2的连接点;
    R 1选自H、-Cl、-Br、-CH 3、CF 3、环丙基或-CH=CH 2
    或者,当L为-NR-时,R 1、R与它们连接的原子一起形成任选取代的5-6元杂环基,其中所述取代基选自卤素、氧代、-iPr、-Et、卤素单取代或多取代的苯基;R 1和R优选形成以下基团:-C(O)N(R N)C(O)-或-C(CH 3)=C(R N)C(O)-;
    其中,
    R N选自-iPr、-Et或
    Figure PCTCN2020110442-appb-100021
    R 11为-Cl、-Br;
    R 12为H或-F;
    R 13为H或-F;
    Figure PCTCN2020110442-appb-100022
    表示连接点;
    R 2为H;
    或者R 1、R 2与它们连接的原子一起形成5-6元杂环基或5-6元杂芳基;优选形成吡咯基;
    R 3选自H或-OMe;
    R 4选自H、-F、-Me、-CF 3或-NHC(O)CH=CH 2
    其他基团如权利要求2所定义。
  8. 权利要求2-7中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、 外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物具有以下通式结构:
    Figure PCTCN2020110442-appb-100023
    Figure PCTCN2020110442-appb-100024
    Figure PCTCN2020110442-appb-100025
    Figure PCTCN2020110442-appb-100026
    Figure PCTCN2020110442-appb-100027
    Figure PCTCN2020110442-appb-100028
    Figure PCTCN2020110442-appb-100029
    其中Y的定义与Z 1相同,并且其他基团如权利要求2-7所定义。
  9. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-1)、(I-1-A)、(I-1-B)、(I-1-C)、(I-1-D)、(I-1-E)、(I-1-F)、(I-1-G)、(I-1-H)或(I-1-I)的化合物:
    其中,
    R 1选自-Cl、-Br、-CF 3或-CH=CH 2;优选地,R 1选自-Cl或-Br;
    R 4为H或-Me;
    R 8为H、-NHMe、-NHC(O)CH 2CH 3或-NH-环丙基;
    R 9为H、-F、-Cl、-Br、-Me、-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH 2CH 3、-NHS(O) 2CH 2CH 3或-NHC(O)CH=CH 2
    X为-C(R x)=或-N=;
    R x为H,或R x连同R 8以及它们所连接的C原子一起形成吡嗪基;
    并且,其他基团如权利要求2-7所定义。
  10. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-G)的化合物:
    Figure PCTCN2020110442-appb-100030
    其中,
    环A为以下基团:
    Figure PCTCN2020110442-appb-100031
    其中,
    R 7为-P(O)(C 1-6烷基) 2
    R 8为H;
    R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;
    X为-C(R x)=;
    其中,R x为H,或者R x连同R 8以及它们所连接的C原子一起形成5-6元杂芳基;优选地为吡嗪基;
    Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2或C=O;优选地,Y为CH 2
    L 1为-C aR #R #’-C bR #R #’-;
    L 2选自化学键、-CR #R #’-或-C aR #R #’-C bR #R #’-;
    其中C aR #R #’或C bR #R #’中任一个可被O、S(O) p或NR #替换;
    E独立地选自:化学键或-C cR #R #’-C dR #R #’-C eR #R #’;
    其中C cR #R #’、C dR #R #’或C eR #R #’中任一个,或者C cR #R #’和C eR #R #’二者可被O、S(O) p或NR #替换;
    p为0、1或2;
    R #为H、卤素、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R #’为H、卤素、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    或者,相邻原子上的R #和R #可以形成化学键,相邻原子上的R #’和R #’可以形成化学键;
    或者,相同或不同原子上的R #和R #’可以一起形成=O;
    m为0、1、2、3、4或5;
    R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;
    s1为0、1、2或3;
    L为-NR-;其中R为H或C 1-6烷基;
    Z为-C(R 4)=;
    T为
    Figure PCTCN2020110442-appb-100032
    R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;
    R 2为H、卤素、羟基、氨基、C 1-6烷基或C 1-6卤代烷基;
    R 3选自H、-O-C 1-6烷基或-O-C 1-6卤代烷基;
    R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;
    当上述各基团为H或含H的基团时,所述一个或多个H原子可以被D原子取代。
  11. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-1-G)、(I-1-H)、(I-1-H’)、(I-1-H”)的化合物:
    Figure PCTCN2020110442-appb-100033
    其中,
    X为-CH=;
    Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2或C=O;
    R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为H或卤素;
    R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4为H;
    R 8为H;
    R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
    L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-,并且上述基团 中的一个或多个H原子可以被D原子取代;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-或-NHCH 2-;优选地,L 1选自-CH 2CH 2-、-C≡C-或-OCH 2-;
    L 2选自化学键、-CH 2-或-CH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;
    E为-CH 2CH 2CH 2-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-或-SCH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;优选地,E为-CH 2CH 2CH 2-;
    m为0、1、2或3;优选地,-L 1-(E) m-L 2-的链长度优选地为4-12个键长,更优选5、6、7、8、9或10个键长;
    R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H、CN或卤素;
    s1为0、1或2。
  12. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-1-G)、(I-1-H)、(I-1-H’)、(I-1-H”)的化合物:
    Figure PCTCN2020110442-appb-100034
    其中,
    X为-CH=;
    Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2
    R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为卤素;
    R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4为H;
    R 8为H;
    R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
    L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-,并且上述基团中的一个或多个H原子可以被D原子取代;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-或-NHCH 2-;优选地,L 1选自-CH 2CH 2-、-C≡C-、-OCH 2-或-NHCH 2-;
    L 2选自化学键、-CH 2-或-CH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;
    E为-CH 2CH 2CH 2-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-或-SCH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;优选地,E为-CH 2CH 2CH 2-;
    m为0、1、2或3;优选地,-L 1-(E) m-L 2-的链长度优选地为4-14个键长,优选5、6、7、8、9或10个键长;
    R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H或卤素;
    s1为0、1或2。
  13. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-1-G)、(I-1-H)、(I-1-H’)、(I-1-H”)的化合物:
    Figure PCTCN2020110442-appb-100035
    其中,
    X为-CH=;
    Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2或C=O;
    R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为卤素;
    R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4为H;
    R 8为H;
    R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
    L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-,并且上述基团中的一个或多个H原子可以被D原子取代;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-或-NHCH 2-;优选地,L 1选自-C≡C-、-OCH 2-或-NHCH 2-;
    L 2选自化学键、-CH 2-或-CH 2CH 2-,并且上述基团中的一个或多个H原子可以被D原子取代;
    E为-CH 2CH 2CH 2-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-或-SCH 2CH 2-, 并且上述基团中的一个或多个H原子可以被D原子取代;优选地,E为-CH 2CH 2CH 2-;
    m为1、2或3;优选地,-L 1-(E) m-L 2-的链长度小于14个键长;优选地,链长度为5-10个键长;
    R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H或卤素;
    s1为0、1或2。
  14. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-1-I)、(I-1-I’)、(I-1-I”)、(I-1-I”’)的化合物:
    Figure PCTCN2020110442-appb-100036
    其中,
    Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2或C=O;
    R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为卤素;
    R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4选自C 1-6烷基或C 1-6卤代烷基;
    R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
    L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-或-OCH 2-;优选地,L 1选自-CH 2CH 2-、-C≡C-或-OCH 2-;
    L 2选自化学键、-CH 2-或-CH 2CH 2-;
    E为-CH 2CH 2CH 2-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-或-SCH 2CH 2-;优选地,E为-CH 2CH 2CH 2-;
    m为1或2;优选地,-L 1-(E) m-L 2-的链长度为4-14个键长,更优选5、6、7、8、9或10个键长;
    R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H、卤素或CN;
    s1为0、1或2。
  15. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋 体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-1-I)、(I-1-I’)、(I-1-I”)、(I-1-I”’)的化合物:
    Figure PCTCN2020110442-appb-100037
    其中,
    Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2
    R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为卤素;
    R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4选自C 1-6烷基或C 1-6卤代烷基;
    R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
    L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-或-OCH 2-;优选地,L 1选自-C≡C-或-OCH 2-;
    L 2选自化学键、-CH 2-或-CH 2CH 2-;
    E为-CH 2CH 2CH 2-;
    m为1或2;优选地,-L 1-(E) m-L 2-的链长度小于10个键长;优选地,链长度为6-9个键长,更优选6、7、8或9个键长;
    R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H或卤素;
    s1为0、1或2。
  16. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-2-I)、(I-2-I’)、(I-2-I”)、(I-2-I”’)的化合物:
    Figure PCTCN2020110442-appb-100038
    其中,
    Y为C原子,其任选地被一个或两个R Z1取代,其中R Z1为H、CN或卤素;或者两个R Z1与Y一起形成C=O;优选地,Y为CH 2
    R 1选自H、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基或C 2-6炔基;优选地,R 1为卤素;
    R 4选自H、卤素、C 1-6烷基或C 1-6卤代烷基;优选地,R 4选自C 1-6烷基或C 1-6卤代烷基;
    R 9选自H、卤素、C 1-6烷基、-CN或C 1-6卤代烷基;优选地,R 9为H;
    L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-SCH 2-、-S(O)CH 2-、-S(O) 2CH 2-、-NHCH 2-、-N(Me)CH 2-、-C(O)CH 2-、-CH 2C(O)-、-OC(O)-、-SC(O)-、-NHC(O)-或-N(Me)C(O)-;优选地,L 1选自-CH 2CH 2-、-CH=CH-、-C≡C-或-OCH 2-;优选地,L 1选自-C≡C-或-OCH 2-;
    L 2选自化学键、-CH 2-或-CH 2CH 2-;
    E为-CH 2CH 2CH 2-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-或-SCH 2CH 2-;优选地,E为-CH 2CH 2CH 2-;
    m为1或2;优选地,-L 1-(E) m-L 2-的链长度小于10个键长;优选地,链长度为6-9个键长,更优选6、7、8或9个键长;
    R s1选自H、CN、卤素、OH、NH 2、C 2-6烯基、C 2-6炔基、-O-C 1-6烷基、-O-C 1-6卤代烷基、-NH-C 1-6烷基、C 1-6烷基或C 1-6卤代烷基;优选地,R s1为H或卤素;
    s1为0、1或2。
  17. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-2)、(I-2-A)、(I-2-B)、(I-2-C)、(I-2-D)、(I-2-E)、(I-2-F)、(I-2-G)、(I-2-H)或(I-2-I)的化合物:
    其中,
    R 1为-Cl、-Br或-CH=CH 2
    R 4为H或-Me;
    R 8为H、-NHMe、-NHC(O)CH 2CH 3或-NH-环丙基;
    R 9为H、-F、-Cl、-Br、-Me、-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH 2CH 3、-NHS(O) 2CH 2CH 3或-NHC(O)CH=CH 2
    X为-C(R x)=;
    R x为H,或R x连同R 8以及它们所连接的C原子一起形成吡嗪基;
    并且,其他基团如权利要求2-7所定义。
  18. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-3)、(I-3-A)、(I-3-B)、(I-3-C)、(I-3-D)、(I-3-E)、(I-3-F)、(I-3-G)或(I-3-H)的化合物:
    其中,
    R 1为H、-Cl或-CH=CH 2
    R 4为-NHC(O)CH=CH 2
    R 5为-Me;
    R 6为-Me;
    或者R 5和R 6连接形成-CH 2CH 2-;
    R 7为-Me或环丙基;
    并且,其他基团如权利要求2-7所定义。
  19. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-4)、(I-4-A)、(I-4-B)、(I-4-C)、(I-4-D)、(I-4-E)、(I-4-F)、(I-4-G)或(I-4-H)的化合物:
    其中,
    R 1为-CF 3
    R 9为H、-F、-Cl、-Br、-Me、-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH=CH 2或-NH-环丙基;
    并且,其他基团如权利要求2-7所定义。
  20. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-5)、(I-5-A)、(I-5-B)、(I-5-C)、(I-5-D)、(I-5-E)、(I-5-F)、(I-5-G)或(I-5-H)的化合物:
    其中,
    环A为任选取代的以下基团:C 3-7环烷基或C 6-10芳基,其中所述取代基选自-F、-Cl、-Br、-Me、-OMe、-CF 3、-OCF 3、-CN、-NHMe、-P(O)Me 2、-NHC(O)CH 2CH 3、-C(O)CH=CH 2、-NHS(O) 2CH 2CH 3、-NH-环丙基、-NHC(O)CH=CH 2或-C(O)CH 2CH 3;环A优选为以下基团:
    Figure PCTCN2020110442-appb-100039
    其中,
    R 9为H、-F、-Cl、-Br、-Me、-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH 2CH 3、-NHS(O) 2CH 2CH 3或-NHC(O)CH=CH 2
    X 1为-CH 2-或-N(R X1)-;
    X 2为-CH(R X2)-或-N(R X2)-;
    其中,
    R X1为-C(O)CH 2CH 3或-C(O)CH=CH 2
    R X2为H、-Me、-OMe、-NHMe、-NHS(O) 2CH 2CH 3、-C(O)CH 2CH 3或-NHC(O)CH 2CH 3
    Figure PCTCN2020110442-appb-100040
    表示连接点;
    R 1、R与它们连接的原子一起形成任选取代的5-6元杂环基,其中所述取代基选自卤素、氧代、 -iPr、-Et、卤素单取代或多取代的苯基;R 1和R优选形成以下基团:-C(O)N(R N)C(O)-或-C(CH 3)=C(R N)C(O)-;
    其中,
    R N选自-iPr、-Et或
    Figure PCTCN2020110442-appb-100041
    R 11为-Cl、-Br;
    R 12为H或-F;
    R 13为H或-F;
    Figure PCTCN2020110442-appb-100042
    表示连接点;
    R 3为H或-OMe;
    R 4为H或-Me;
    并且,其他基团如权利要求2-7所定义。
  21. 权利要求20的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-5-1)或(I’-5-1)化合物:
    Figure PCTCN2020110442-appb-100043
    其中,
    R 9为-NHC(O)CH 2CH 3或-NHC(O)CH=CH 2
    R N为-iPr或-Et;
    并且,其他基团如权利要求2-7所定义。
  22. 权利要求20的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-5-2)或(I’-5-2)化合物:
    Figure PCTCN2020110442-appb-100044
    其中,
    R 3为H或-OMe;
    R 4为H或-Me;
    并且,其他基团如权利要求2-7所定义。
  23. 权利要求20的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-5-3)或(I’-5-3)化合物:
    Figure PCTCN2020110442-appb-100045
    其中,
    X 1为-CH 2-或-N(R X1)-;
    X 2为-CH(R X2)-或-N(R X2)-;
    其中,
    R X1为-C(O)CH 2CH 3
    R X2为H、-Me、-OMe、-NHMe、-C(O)CH 2CH 3、-NHS(O) 2CH 2CH 3或-NHC(O)CH 2CH 3
    R 11为-Cl或-Br;
    R 12为H或-F;
    R 13为H或-F;
    并且,其他基团如权利要求2-7所定义。
  24. 权利要求8的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,其中,所述通式(I)化合物为通式(I-6)、(I-6-A)、(I-6-B)、(I-6-C)、(I-6-D)、(I-6-E)、(I-6-F)、(I-6-G)或(I-6-H)的化合物:
    其中,
    L为-O-或-NH-;
    R 3为H或-OMe;
    R 4为H或-F;
    R 9为-CF 3、-OMe、-OCF 3、-CN、-NHC(O)CH 2CH 3或-NHC(O)CH=CH 2
    并且,其他基团如权利要求2-7所定义。
  25. 权利要求1-7中任一项的通式(X)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,
    其中,
    Figure PCTCN2020110442-appb-100046
    选自以下基团:
    Figure PCTCN2020110442-appb-100047
    Figure PCTCN2020110442-appb-100048
    Figure PCTCN2020110442-appb-100049
    并且上述基团中的一个或多个H原子可以被D原子取代。
  26. 权利要求1-7中任一项的通式(X)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,
    其中L 1和L 2独立地选自化学键、-O-、-S-、-S(O)-、-S(O) 2-、-S(O)(=NH)-、-S(O)(=NMe)-、
    Figure PCTCN2020110442-appb-100050
    Figure PCTCN2020110442-appb-100051
    -NH-、-N(Me)-、
    Figure PCTCN2020110442-appb-100052
    -N(CF 3)-、-CH 2-、-CH(OMe)-、-CH(Cl)-、-CH(F)-、-CF 2-、-CH(CF 3)-、-C(O)-、-CH 2CH 2-、-CH=CH-、-C≡C-、-OCH 2-、-CH 2O-、-SCH 2-、-CH 2S-、-S(O)CH 2-、-CH 2S(O)-、-S(O) 2CH 2-、-CH 2S(O) 2-、-NHCH 2-、-N(Me)CH 2-、-CH 2NH-、-CH 2N(Me)-、-C(O)CH 2-、-CH 2C(O)-、-C(O)CMe 2-、-CMe 2C(O)-、-OC(O)-、-C(O)O-、-SC(O)-、-C(O)S-、-NHC(O)-、-N(Me)C(O)-、-C(O)NH-、-C(O)N(Me)-、-S(O)=NH-、-NH=S(O)-、-N=S(O)Me-、-S(O)Me=N-、
    Figure PCTCN2020110442-appb-100053
    并且上述基团中的一个或多个H原子可以被D原子取代。
  27. 权利要求1-7中任一项的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,
    其中E选自化学键、-CH 2CH 2CH 2-、-CH 2CH=CH-、-CH=CHCH 2-、-CH 2C≡C-、-C≡CCH 2-、-CH 2CH 2C(O)-、-CH 2C(O)CH 2-、-C(O)CH 2CH 2-、-CH 2CH 2S(O) 2-、-CH 2S(O) 2CH 2-、-S(O) 2CH 2CH 2-、-C(O)CH=CH-、-C(O)C≡C-、-CH 2CH 2O-、-CH 2OCH 2-、-OCH 2CH 2-、-CH 2CH 2S-、-CH 2SCH 2-、-SCH 2CH 2-、-C(O)CH 2O-、-OCH 2C(O)-、-CH 2C(O)O-、-C(O)CH 2S-、-SCH 2C(O)-、-CH 2C(O)S-、-OC(O)CH 2-、-C(O)OCH 2-、-CH 2OC(O)-、-SC(O)CH 2-、-C(O)SCH 2-、-CH 2SC(O)-、-CH 2CH 2NH-、-CH 2NHCH 2-、-NHCH 2CH 2-、-CH 2CH 2NMe-、-CH 2NMeCH 2-、-NMeCH 2CH 2-、-C(O)CH 2NH-、-NHCH 2C(O)-、-CH 2C(O)NH-、-NHC(O)CH 2-、-C(O)NHCH 2-、-CH 2NHC(O)-、
    Figure PCTCN2020110442-appb-100054
    Figure PCTCN2020110442-appb-100055
    Figure PCTCN2020110442-appb-100056
    或者两个E单元,或者两个E’单元可以形成-CH 2CH 2OCH 2CH 2-、-OCH 2CH 2CH 2CH 2-、-CH 2CH 2CH 2CH 2O-、
    Figure PCTCN2020110442-appb-100057
    Figure PCTCN2020110442-appb-100058
    并且上述基团中的一个或多个H原子可以被D原子取代。
  28. 权利要求1的通式(I)化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,以及它们的混合物,所述化合物选自以下:
    Figure PCTCN2020110442-appb-100059
    Figure PCTCN2020110442-appb-100060
    Figure PCTCN2020110442-appb-100061
    Figure PCTCN2020110442-appb-100062
    Figure PCTCN2020110442-appb-100063
    Figure PCTCN2020110442-appb-100064
  29. 药物组合物,其含有权利要求1-28中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体,和药学上可接受的赋形剂;优选地,其还含有其它治疗剂。
  30. 权利要求1-28中任一项的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体在制备用于治疗和/或预防EGFR和/或ALK激酶介导的疾病的药物中的用途。
  31. 一种在受试者中治疗和/或预防EGFR和/或ALK激酶介导的疾病的方法,所述方法包括向所述受试者给药权利要求1-28中任一项的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体或权利要求29的药物组合物。
  32. 权利要求1-28中任一项的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或同位素变体或权利要求29的药物组合物,其用于治疗和/或预防EGFR和/或ALK激酶介导的疾病。
  33. 权利要求30的用途或权利要求31的方法或权利要求32的化合物或组合物的用途,其中所述EGFR和/或ALK激酶介导的疾病包括癌症,例如卵巢癌、宫颈癌、结肠直肠癌、乳腺癌、胰腺癌、胶质瘤、胶质母细胞瘤、黑色素瘤、前列腺癌、白血病、淋巴瘤、非霍奇金淋巴瘤、胃癌、肺癌、肝细胞癌、胃癌、胃肠道基质瘤(GIST)、甲状腺癌、胆管癌、子宫内膜癌、肾癌、间变性大细胞淋巴瘤、急性髓细胞白血病(AML)、多发性骨髓瘤、黑色素瘤、间皮瘤。
PCT/CN2020/110442 2019-08-23 2020-08-21 抑制并诱导降解egfr和alk的化合物 WO2021036922A1 (zh)

Priority Applications (5)

Application Number Priority Date Filing Date Title
KR1020227009748A KR20220051244A (ko) 2019-08-23 2020-08-21 Egfr 및 alk의 억제 및 분해 유도 화합물
EP20857158.8A EP4019021A4 (en) 2019-08-23 2020-08-21 INHIBITING AND INDUCING DEGRADATION OF EGFR AND ALK
JP2022512798A JP2022546375A (ja) 2019-08-23 2020-08-21 Egfrおよびalkを阻害してそれらの分解を阻害する化合物
US17/637,690 US20220306659A1 (en) 2019-08-23 2020-08-21 Compound inhibiting and inducing degradation of egfr and alk
CN202080059273.7A CN114286678A (zh) 2019-08-23 2020-08-21 抑制并诱导降解egfr和alk的化合物

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN201910785651 2019-08-23
CN201910785651.9 2019-08-23
CN202010072446 2020-01-21
CN202010072446.0 2020-01-21
CN202010840485 2020-08-20
CN202010840485.0 2020-08-20

Publications (1)

Publication Number Publication Date
WO2021036922A1 true WO2021036922A1 (zh) 2021-03-04

Family

ID=74684046

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/110442 WO2021036922A1 (zh) 2019-08-23 2020-08-21 抑制并诱导降解egfr和alk的化合物

Country Status (7)

Country Link
US (1) US20220306659A1 (zh)
EP (1) EP4019021A4 (zh)
JP (1) JP2022546375A (zh)
KR (1) KR20220051244A (zh)
CN (1) CN114286678A (zh)
TW (1) TWI758832B (zh)
WO (1) WO2021036922A1 (zh)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022012623A1 (en) * 2020-07-16 2022-01-20 Beigene, Ltd. Degradation of (egfr) by conjugation of egfr inhibitors with e3 ligase ligand and methods of use
WO2022012622A1 (en) * 2020-07-16 2022-01-20 Beigene, Ltd. Degradation of (egfr) by conjugation of egfr inhibitors with e3 ligase ligand and methods of use
WO2022171123A1 (en) * 2021-02-10 2022-08-18 Beigene, Ltd. Egfr degraders and methods of use
WO2022228547A1 (zh) * 2021-04-30 2022-11-03 四川海思科制药有限公司 一种膦酰衍生物及其组合物和药学上的应用
WO2022228556A1 (en) * 2021-04-30 2022-11-03 Beigene, Ltd. Egfr degraders and associated methods of use
WO2022242725A1 (zh) * 2021-05-19 2022-11-24 和径医药科技(上海)有限公司 一类新型蛋白降解剂及其应用
WO2022268052A1 (en) * 2021-06-21 2022-12-29 Beigene, Ltd. (r) -glutarimide crbn ligands and methods of use
WO2022268229A1 (zh) * 2021-06-25 2022-12-29 和径医药科技(上海)有限公司 蛋白抑制剂或降解剂、包含其的药物组合物及医药上的用途
WO2023104155A1 (zh) * 2021-12-08 2023-06-15 标新生物医药科技(上海)有限公司 E3泛素连接酶配体化合物、基于该配体化合物开发的蛋白降解剂及它们的应用
EP4029499A4 (en) * 2019-06-12 2023-07-26 Shanghaitech University ALK PROTEIN REGULATOR AND ITS ANTITUMOR APPLICATION
WO2023198180A1 (zh) * 2022-04-15 2023-10-19 北京泰德制药股份有限公司 Egfr降解剂

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024032600A1 (zh) * 2022-08-08 2024-02-15 西藏海思科制药有限公司 一种杂环衍生物及其组合物和药学上的应用

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
WO2016105518A1 (en) * 2014-12-23 2016-06-30 Dana-Farber Cancer Institute, Inc. Methods to induce targeted protein degradation through bifunctional molecules
WO2018033556A1 (en) * 2016-08-18 2018-02-22 Glaxosmithkline Intellectual Property Development Limited Novel compounds
WO2018119441A1 (en) * 2016-12-23 2018-06-28 Arvinas, Inc. Egfr proteolysis targeting chimeric molecules and associated methods of use
CN109422733A (zh) * 2017-09-03 2019-03-05 上海美志医药科技有限公司 一类抑制并降解酪氨酸蛋白激酶alk的化合物
WO2019113071A1 (en) * 2017-12-05 2019-06-13 Icahn School Of Medicine At Mount Sinai Compositions and methods for treating alk-mediated cancer
CN109912655A (zh) * 2017-12-13 2019-06-21 上海科技大学 Alk蛋白降解剂及其抗肿瘤应用
CN109928956A (zh) * 2019-02-27 2019-06-25 杭州偶联医药科技有限公司 一种靶向泛素化降解egfr蛋白的化合物及其药物组合物和应用
WO2019196812A1 (zh) * 2018-04-09 2019-10-17 上海科技大学 蛋白降解靶向化合物、其抗肿瘤应用、其中间体及中间体应用
CN110684015A (zh) * 2018-07-06 2020-01-14 四川大学 靶向alk的protac及其应用
CN111285849A (zh) * 2018-12-07 2020-06-16 上海青东生物科技有限公司 一种靶向降解ALK,c-Met和ROS1蛋白的化合物及其制备方法

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9273077B2 (en) * 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
CN105985342B (zh) * 2015-02-06 2018-07-24 华东理工大学 作为egfr抑制剂的嘧啶并嘧啶二酮衍生物及其应用
WO2019015593A1 (zh) * 2017-07-19 2019-01-24 江苏奥赛康药业股份有限公司 嘧啶并吡啶酮或者吡啶并吡啶酮类化合物及其应用
CN110944989B (zh) * 2017-07-19 2021-06-25 正大天晴药业集团股份有限公司 作为egfr激酶抑制剂的芳基磷氧化合物
CN110204532B (zh) * 2019-05-15 2022-03-08 浙江工业大学 一种靶向egfr蛋白降解的化合物及其制备方法和应用
US20220257776A1 (en) * 2019-06-12 2022-08-18 Shanghaitech University Alk protein regulator and anti-tumor application thereof

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
WO2016105518A1 (en) * 2014-12-23 2016-06-30 Dana-Farber Cancer Institute, Inc. Methods to induce targeted protein degradation through bifunctional molecules
WO2018033556A1 (en) * 2016-08-18 2018-02-22 Glaxosmithkline Intellectual Property Development Limited Novel compounds
WO2018119441A1 (en) * 2016-12-23 2018-06-28 Arvinas, Inc. Egfr proteolysis targeting chimeric molecules and associated methods of use
CN109422733A (zh) * 2017-09-03 2019-03-05 上海美志医药科技有限公司 一类抑制并降解酪氨酸蛋白激酶alk的化合物
WO2019113071A1 (en) * 2017-12-05 2019-06-13 Icahn School Of Medicine At Mount Sinai Compositions and methods for treating alk-mediated cancer
CN109912655A (zh) * 2017-12-13 2019-06-21 上海科技大学 Alk蛋白降解剂及其抗肿瘤应用
WO2019196812A1 (zh) * 2018-04-09 2019-10-17 上海科技大学 蛋白降解靶向化合物、其抗肿瘤应用、其中间体及中间体应用
CN110684015A (zh) * 2018-07-06 2020-01-14 四川大学 靶向alk的protac及其应用
CN111285849A (zh) * 2018-12-07 2020-06-16 上海青东生物科技有限公司 一种靶向降解ALK,c-Met和ROS1蛋白的化合物及其制备方法
CN109928956A (zh) * 2019-02-27 2019-06-25 杭州偶联医药科技有限公司 一种靶向泛素化降解egfr蛋白的化合物及其药物组合物和应用

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
"Remington s Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
BERGE S. M. ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: 10.1002/jps.2600660104
CHELSEA E. POWELL, YANG GAO, LI TAN, KATHERINE A. DONOVAN, RADOSŁAW P. NOWAK, AMANDA LOEHR, MAGDA BAHCALL, ERIC S. FISCHER, PASI A: "Chemically Induced Degradation of Anaplastic Lymphoma Kinase (ALK)", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, vol. 61, no. 9, 10 May 2018 (2018-05-10), pages 4249 - 4255, XP055628330, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.7b01655 *
D. FLEISHERS. RAMONH. BARBRA: "Improved oral drug delivery: solubility limitations overcome by the use of prodrugs", ADVANCED DRUG DELIVERY REVIEWS, vol. 19, no. 2, 1996, pages 115 - 130
JAMA ONCOL, vol. 4, no. 11, 2018, pages 1527 - 1534
NATURE MEDICINE, vol. 21, no. 6, 2015, pages 560 - 562
See also references of EP4019021A4
T. HIGUCHIV. STELLA: "Prodrugs as Novel Delivery Systems", A.C.S. SYMPOSIUM SERIES, vol. 14

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4029499A4 (en) * 2019-06-12 2023-07-26 Shanghaitech University ALK PROTEIN REGULATOR AND ITS ANTITUMOR APPLICATION
WO2022012623A1 (en) * 2020-07-16 2022-01-20 Beigene, Ltd. Degradation of (egfr) by conjugation of egfr inhibitors with e3 ligase ligand and methods of use
WO2022012622A1 (en) * 2020-07-16 2022-01-20 Beigene, Ltd. Degradation of (egfr) by conjugation of egfr inhibitors with e3 ligase ligand and methods of use
WO2022171123A1 (en) * 2021-02-10 2022-08-18 Beigene, Ltd. Egfr degraders and methods of use
WO2022228547A1 (zh) * 2021-04-30 2022-11-03 四川海思科制药有限公司 一种膦酰衍生物及其组合物和药学上的应用
WO2022228556A1 (en) * 2021-04-30 2022-11-03 Beigene, Ltd. Egfr degraders and associated methods of use
WO2022242725A1 (zh) * 2021-05-19 2022-11-24 和径医药科技(上海)有限公司 一类新型蛋白降解剂及其应用
WO2022268052A1 (en) * 2021-06-21 2022-12-29 Beigene, Ltd. (r) -glutarimide crbn ligands and methods of use
WO2022268229A1 (zh) * 2021-06-25 2022-12-29 和径医药科技(上海)有限公司 蛋白抑制剂或降解剂、包含其的药物组合物及医药上的用途
WO2023104155A1 (zh) * 2021-12-08 2023-06-15 标新生物医药科技(上海)有限公司 E3泛素连接酶配体化合物、基于该配体化合物开发的蛋白降解剂及它们的应用
WO2023198180A1 (zh) * 2022-04-15 2023-10-19 北京泰德制药股份有限公司 Egfr降解剂

Also Published As

Publication number Publication date
JP2022546375A (ja) 2022-11-04
TWI758832B (zh) 2022-03-21
CN114286678A (zh) 2022-04-05
EP4019021A4 (en) 2022-11-02
TW202115026A (zh) 2021-04-16
EP4019021A1 (en) 2022-06-29
US20220306659A1 (en) 2022-09-29
KR20220051244A (ko) 2022-04-26

Similar Documents

Publication Publication Date Title
WO2021036922A1 (zh) 抑制并诱导降解egfr和alk的化合物
WO2021121261A1 (zh) 抑制并诱导降解egfr激酶的化合物
WO2021219091A2 (zh) 喹喔啉酮衍生物作为kras g12c突变蛋白的不可逆抑制剂
CN111471021B (zh) 取代的二氨基甲酰胺和二氨基甲腈嘧啶,其组合物,和用其治疗的方法
CN112047938B (zh) 作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物
CN104470898B (zh) 环烷基甲酸类衍生物、其制备方法及其在医药上的应用
TW202128668A (zh) 并環類衍生物、其製備方法及其在醫藥上的應用
TW202100512A (zh) 稠合芳香環類衍生物、其製備方法及其在醫藥上的應用
WO2021185256A1 (zh) 取代的嘧啶或吡啶胺衍生物、其组合物及医药上的用途
WO2023134266A1 (zh) 2-哌啶基或2-吡唑基取代的嘧啶化合物作为egfr抑制剂
TW202110831A (zh) 吡啶酮類衍生物、其製備方法及其在醫藥上的應用
WO2019149089A1 (zh) 含有羧酸基团的苯并氮杂环类化合物及其制备方法和用途
BR112021003949A2 (pt) derivado de anel fundido usado como inibidor de fgfr4
CN113912589B (zh) 抑制并诱导蛋白降解的化合物
WO2022012484A1 (zh) 作为atr激酶抑制剂的吡唑并嘧啶化合物
WO2021233376A1 (zh) 作为atr激酶抑制剂的2,4,6-三取代的嘧啶化合物
WO2023198180A1 (zh) Egfr降解剂
CN105524053A (zh) 四氢苯并噻吩化合物
WO2024067463A1 (zh) 苯并[7]环烯类衍生物、包含其的药物组合物及其医药用途
WO2020125391A1 (zh) 用于抑制蛋白激酶活性的氨基嘧啶类化合物
TW202214656A (zh) 氧氮雜環螺環衍生物、其製備方法及其醫藥用途
WO2023216910A1 (zh) 取代的双环杂芳基化合物作为usp1抑制剂

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: 20857158

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2022512798

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20227009748

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020857158

Country of ref document: EP

Effective date: 20220323