[go: up one dir, main page]

WO2024011883A1 - Wee1抑制剂及其制备和用途 - Google Patents

Wee1抑制剂及其制备和用途 Download PDF

Info

Publication number
WO2024011883A1
WO2024011883A1 PCT/CN2023/072296 CN2023072296W WO2024011883A1 WO 2024011883 A1 WO2024011883 A1 WO 2024011883A1 CN 2023072296 W CN2023072296 W CN 2023072296W WO 2024011883 A1 WO2024011883 A1 WO 2024011883A1
Authority
WO
WIPO (PCT)
Prior art keywords
μmol
compound
add
alkyl
reaction
Prior art date
Legal status (The legal status 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 status listed.)
Ceased
Application number
PCT/CN2023/072296
Other languages
English (en)
French (fr)
Inventor
宋利
唐海
马晓慧
周水平
蔡金勇
董利明
宋壮
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Jiangsu Tasly Diyi Pharmaceutical Co Ltd
Original Assignee
Jiangsu Tasly Diyi Pharmaceutical Co Ltd
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 Jiangsu Tasly Diyi Pharmaceutical Co Ltd filed Critical Jiangsu Tasly Diyi Pharmaceutical Co Ltd
Priority to US18/867,250 priority Critical patent/US20250304587A1/en
Priority to EP23838385.5A priority patent/EP4570807A1/en
Priority to KR1020247043576A priority patent/KR20250034312A/ko
Priority to CA3248185A priority patent/CA3248185A1/en
Priority to AU2023306869A priority patent/AU2023306869A1/en
Priority to JP2024568139A priority patent/JP2025525290A/ja
Publication of WO2024011883A1 publication Critical patent/WO2024011883A1/zh
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53861,4-Oxazines, e.g. morpholine spiro-condensed or forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/14Ortho-condensed systems
    • C07D491/147Ortho-condensed systems the condensed system containing one ring with oxygen as ring hetero atom and two rings with nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention relates to a class of compounds with inhibitory activity of WEE1 kinase and their use in treating diseases mediated by WEE1.
  • WEE1 tyrosine kinase is a checkpoint in the G2 phase of the cell cycle.
  • the cell cycle is tightly regulated. When the cell's DNA is not damaged, the G1 phase, S phase, and G 2 phase checkpoints promote cells to enter the division phase and ensure the smooth completion of the cell cycle. (Clinical Cancer Research, 2011, 17(13): 4200-4207.)
  • the cell cycle is regulated by cyclin-dependent kinases (CDKs).
  • CDKs cyclin-dependent kinases
  • the CDKs family has 14 serine/threonine protein kinases. CDK activity is regulated by phosphorylation and binding to different cycle proteins.
  • CDK1 also known as CDC2
  • WEE1 phosphorylation of CDK1
  • MYT1 myelin transcription factor
  • WEE1 is a negative regulator of the cell cycle.
  • WEE1 By preventing cyclin B and activated CDK1 complexes from entering the nucleus, WEE1 negatively regulates cells from the G2 phase to the division phase.
  • the expression and activity of WEE1 are increased in S phase and G2 phase, and decreased in the highly phosphorylated M phase.
  • polo-like protein kinase 1 phosphorylates WEE1 and degrades WEE1 through the ubiquitin ligase complex.
  • PLK1 also phosphorylates and activates the protein phosphatase cell division cycle 25 analog (CDC25), which activates CDK1 through dephosphorylation.
  • Active CDK1 can bind to cyclin B and promote cells to enter the division phase (Molecular & Cellular Biology, 2012, 32(20): 4226.).
  • the G1, S and G2 phase checks delay the cell's entry into the division phase, buying time for the cell to repair the damaged DNA before it enters division, thereby ensuring the integrity of the genome.
  • P53 a key regulator of the G1 phase checkpoint, is mutated in many malignant tumor cells. (Proceedings of the National Academy of Sciences of the United States of America, 2007, 104(10): 3753-3758.) Tumor cells with defective P53 function cannot arrest the cell cycle in the G1 phase when their DNA is damaged. Therefore, it relies more on the G2 phase checkpoint.
  • the G2 phase checkpoint In response to DNA damage, the G2 phase checkpoint inhibits the phosphorylation of CDK1 through two parallel and interconnected pathways, thereby delaying cells from entering the division phase.
  • ataxia telangiectasia variant (ATM) protein kinase or ataxia telangiectasia related (ATR) protein kinase is activated (Oncotarget, 2016, 7(31): 49902-49916.) .
  • ATM is activated by ionizing radiation, radioactive agents, and agents that cause double-stranded DNA breaks.
  • ATM phosphorylates and activates checkpoint kinase 2 (CHK2), which phosphorylates Ser216 of cell division cycle 25C phosphatase (CDC25C).
  • CHK2 checkpoint kinase 2
  • CDC25C cell division cycle 25C phosphatase
  • Inhibiting the activity of CDC25C results in the inhibition of the phosphorylation of the CDK1/CDK2-binding cyclin B complex, leaving CDK1 in an inactive form and inhibiting cells from entering the division phase (Molecular Cancer, 2014, 13(1): 72.).
  • ATR is activated by a wide range of genotoxic stimuli that lead to single-stranded DNA breaks.
  • ATR is the primary kinase responsible for phosphorylating and activating CHK1.
  • CHK1 can be activated by both ATM and ATR.
  • CHK1 simultaneously phosphorylates WEE1 and CDC25C, activating WEE1 kinase activity and inhibiting the phosphatase activity of CDC25C.
  • WEE1 phosphorylates CDK1-binding cyclin B, causing cell cycle arrest in the G2 phase, providing time for DNA repair (Drug News & Perspectives, 2010, 23(7): 425.).
  • WEE1 is overexpressed in many malignant tumors, such as liver cancer, breast cancer, malignant glioma, melanoma, and brain tumors in adults and children. Some tumor cells have abnormal G1 checkpoints. If WEE1 activity is inhibited, G2 checkpoint failure will occur. At this time, cells with unrepaired damaged DNA will continue to divide and eventually divide and die. (Molecular Cancer Therapeutics, 2013, 12(12): 2675-2684.) Whether through pyrimidine derivatives (PD0166285) or small interfering RNA knockdown, inhibiting the activity of WEE1 will cause ovarian cancer, colon cancer, cervical cancer, and osteosarcoma.
  • malignant tumors such as liver cancer, breast cancer, malignant glioma, melanoma, and brain tumors in adults and children. Some tumor cells have abnormal G1 checkpoints. If WEE1 activity is inhibited, G2 checkpoint failure will occur. At this time, cells with unrepaired damaged DNA will continue to divide and eventually divide and die.
  • Patents WO2007126122, WO2008133866, WO2013012681, WO2013126656, WO2014167347, WO2015092431, WO2018011569, WO2018011570, WO2018090939, WO2018133829, WO2 018171633 et al. describe small molecule compounds with WEE1 kinase inhibitory activity.
  • the most leading compound at present is AZD1775, which has entered clinical phase II trials and has shown good cancer treatment effects.
  • the object of the present invention is to provide a compound represented by formula I, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof: in,
  • R 1 is selected from -C 1 ⁇ 6 alkyl, -C 2 ⁇ 6 alkenyl, -C 2 ⁇ 6 alkynyl, -C 0 ⁇ 2 alkylene-CN, -C 0 ⁇ 2 alkylene -(3 ⁇ 10 membered cycloalkyl), -C 0 ⁇ 2 alkylene-(3 ⁇ 10 membered heterocycloalkyl);
  • R 2 is selected from
  • the X is selected from O, NH or CH 2 ;
  • the X 1 is selected from CH or N;
  • R 21 , R 22 , and R 29 are each independently selected from hydrogen, deuterium, halogen, cyano, nitro, -OH, -C 1 ⁇ 6 alkyl, halogen-substituted C 1 ⁇ 6 alkyl, -C 0 ⁇ 2 alkylene -OH, -O (C 1 ⁇ 6 alkyl), -O (halogen substituted C 1 ⁇ 6 alkyl), -NH 2 , -C 0 ⁇ 2 alkylene -NH (C 1 ⁇ 6 alkyl), -C 0 ⁇ 2 alkylene-N(C 1 ⁇ 6 alkyl)(C 1 ⁇ 6 alkyl), -C 0 ⁇ 2 alkylene-(3 ⁇ 10 membered cycloalkyl) , -C 0 ⁇ 2 alkylene-(3 ⁇ 10 membered heterocycloalkyl);
  • R 27 and R 28 together with the atoms directly connected to them form a 3 to 10-membered carbocyclic ring and a 3 to 10-membered heterocyclic ring;
  • R 3 is selected from hydrogen, deuterium, halogen, cyano, nitro, -C 1 ⁇ 6 alkyl, halogen-substituted C 1 ⁇ 6 alkyl, -C 0 ⁇ 2 alkylene -OH, -O(C 1 ⁇ 6 alkyl), -O (halogen-substituted C 1 ⁇ 6 alkyl), -NH 2 , -C 0 ⁇ 2 alkylene -NH (C 1 ⁇ 6 alkyl), -C 0 ⁇ 2 alkylene Base-N(C 1 ⁇ 6 alkyl)(C 1 ⁇ 6 alkyl);
  • the R 4 is selected from 3 to 12-membered heterocycloalkyl; the heterocycloalkyl can be further substituted by one, two, three or four independent R 41 ;
  • R 41 is selected from hydrogen, halogen, cyano, nitro, -OH, -C 1 ⁇ 6 alkyl, halogen-substituted C 1 ⁇ 6 alkyl, -C 0 ⁇ 2 alkylene -OH, - O (C 1 to 6 alkyl), -O (halogen-substituted C 1 to 6 alkyl), -NH 2 , -C 0 to 2 alkylene -NH (C 1 to 6 alkyl), -C 0 ⁇ 2 alkylene-N(C 1 ⁇ 6 alkyl)(C 1 ⁇ 6 alkyl), -C(O)C 1 ⁇ 6 alkyl, 3 ⁇ 10 membered carbocyclic ring, 3 ⁇ 10 membered heterocyclic ring;
  • the carbocyclic ring and heterocyclic ring may be further substituted by one, two, three or four independent R 31 ;
  • the R 3 and R 4 and the atoms directly connected to them together form a 3-10-membered carbocyclic ring or a 3-10-membered heterocyclic ring; the described carbocyclic ring or heterocycloalkyl group can be further replaced by one, two or three members.
  • R 31 is selected from hydrogen, halogen, cyano, nitro, -OH, -C 1 ⁇ 6 alkyl, halogen-substituted C 1 ⁇ 6 alkyl, -C 0 ⁇ 2 alkylene -OH, - O (C 1 to 6 alkyl), -O (halogen-substituted C 1 to 6 alkyl), -NH 2 , -C 0 to 2 alkylene -NH (C 1 to 6 alkyl), -C 0 ⁇ 2Alkylene -N(C 1 ⁇ 6 alkyl)(C 1 ⁇ 6 alkyl).
  • the compound of the present invention or its stereoisomer, or its pharmaceutically acceptable salt, said R 1 is selected from Methyl, ethyl,
  • R 21 , R 22 , and R 29 are independently selected from hydrogen, deuterium, cyano, methyl, and ethanol.
  • base -OH, trifluoromethyl, cyclopropyl, -CH 2 OH, -NH 2 .
  • the R 23 and R 24 together with the atoms directly connected to them form a cyclopropyl group, a cyclobutyl group, or a cyclobutyl group.
  • R 25 and R 26 together with the atoms directly connected to them form cyclopropyl, cyclobutyl or cyclopentyl;
  • R 27 and R 28 together with the atoms directly connected to them form cyclopropyl, cyclobutyl or cyclopentyl.
  • the compound of the present invention, or its stereoisomer, or its pharmaceutically acceptable salt, said R 2 is selected from
  • the R 3 is selected from hydrogen, fluorine, methyl, -CH 2 OH, and methoxy.
  • the compound of the present invention or its stereoisomer, or its pharmaceutically acceptable salt, said R 4 is selected from the group consisting of a nitrogen-containing 6-membered heterocycle, a 7-membered nitrogen-containing bridged ring, an 8-membered nitrogen-containing bridged ring, Nitrogen bridged ring, 9-membered nitrogen-containing heterospirocycle, 11-membered nitrogen-containing heterospirocycle.
  • R 4 is selected from
  • R 4 is selected from
  • the R 3 and R 4 together with the atoms directly connected to them form a 6-membered nitrogen-containing heterocyclic ring.
  • R 31 is selected from methyl.
  • the compound of the present invention, or its stereoisomer, or its pharmaceutically acceptable salt, the compound of Formula I is specifically:
  • the compound of the present invention, or its stereoisomer, or its pharmaceutically acceptable salt, the compound described in Formula I is specifically:
  • the present invention also provides a compound described in any one of the above, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof Use in the preparation of drugs for the treatment of WEE1-mediated diseases.
  • the WEE1-mediated disease is one or more of the diseases related to inflammation, autoimmune diseases, infectious diseases, cancer, and precancerous syndromes.
  • the present invention also provides a pharmaceutical composition, which is prepared from any of the above-mentioned compounds, or its stereoisomers, or its pharmaceutically acceptable salts, plus pharmaceutically acceptable auxiliary materials. preparation.
  • Cancer or “malignancy” means any of a variety of diseases characterized by uncontrolled abnormal proliferation of cells, the ability of affected cells to spread to other sites locally or through the bloodstream and lymphatic system body (i.e. metastasis) and any of a number of characteristic structural and/or molecular characteristics.
  • Cancer cells refer to cells that undergo early, intermediate or late stages of multi-step tumor progression. Cancers include sarcomas, breast cancer, lung cancer, brain cancer, bone cancer, liver cancer, kidney cancer, colon cancer, and prostate cancer.
  • compounds of Formula I are used to treat a cancer selected from the group consisting of colon cancer, brain cancer, breast cancer, fibrosarcoma, and squamous cell carcinoma.
  • the cancer is selected from melanoma, breast cancer, colon cancer, lung cancer, and ovarian cancer.
  • the cancer treated is metastatic cancer.
  • autoimmune diseases are caused by the body's immune response to substances and tissues normally present in the body.
  • autoimmune diseases include myocarditis, lupus nephritis, primary biliary cirrhosis, psoriasis, type I diabetes, Grave's disease, celiac disease, Crohn's disease, autoimmune neutropenia, juvenile form Arthritis, rheumatoid arthritis, fibromyalgia, Guillain-Barré syndrome, multiple sclerosis and autoimmune retinopathy.
  • Some embodiments of the invention relate to the treatment of autoimmune diseases such as psoriasis or multiple sclerosis.
  • Inflammatory diseases include a variety of conditions characterized by histopathological inflammation.
  • inflammatory diseases include acne vulgaris, asthma, celiac disease, chronic prostatitis, glomerulonephritis, inflammatory bowel disease, pelvic inflammatory disease, reperfusion injury, rheumatoid arthritis, sarcoidosis, vasculitis, Airway inflammation and interstitial cystitis caused by house dust mites.
  • Some embodiments of the invention relate to the treatment of the inflammatory disease asthma.
  • the immune system is often involved in inflammatory disorders, manifesting itself in allergic reactions and some myopathies, and many immune system disorders result in abnormal inflammation.
  • IL-17A-mediated diseases also include autoimmune inflammatory diseases.
  • substitution means that a hydrogen atom in a molecule is replaced by a different atom or molecule.
  • C a to b alkyl indicates any alkyl group containing "a" to "b” carbon atoms.
  • C 1-4 alkyl refers to an alkyl group containing 1 to 4 carbon atoms.
  • Alkyl refers to a saturated hydrocarbon chain having the specified number of member atoms.
  • C 1-6 alkyl refers to an alkyl group having 1 to 6 member atoms, such as 1 to 4 member atoms.
  • Alkyl groups can be straight or branched. Representative branched alkyl groups have one, two or three branches. Alkyl groups may be optionally substituted with one or more substituents as defined herein. Alkyl groups include methyl, ethyl, propyl (n-propyl and isopropyl), butyl (n-butyl, isobutyl and tert-butyl), pentyl (n-pentyl, isopentyl and neopentyl). base) and hexyl base. Alkyl groups may also be part of other groups, such as C 1 -C 6 alkoxy groups.
  • Cycloalkyl and “cycloalkane” refer to saturated or partially saturated cyclic groups having carbon atoms and no ring heteroatoms and having a single ring or multiple rings (including fused, combined and bridged rings).
  • cycloalkyl applies when the point of attachment is at a nonaromatic carbon atom (e.g., 5,6,7,8,-tetra Hydronaphthalene-5-yl).
  • cycloalkyl includes cycloalkenyl groups such as cyclohexenyl.
  • cycloalkyl groups include, for example, adamantyl, cyclopropyl, cyclobutyl, cyclohexyl, cyclopentyl, cyclooctyl, cyclopentenyl and cyclohexenyl. Includes polybicycloalkyl rings
  • cycloalkyl groups of the system are bicyclohexyl, bicyclopentyl, bicyclooctyl, etc. For example
  • (Ca-Cb)alkenyl refers to an alkenyl group having a to b carbon atoms and is intended to include, for example, vinyl, propenyl, isopropenyl, 1,3-butadienyl, and the like.
  • Alkynyl refers to a straight chain monovalent hydrocarbon group or a branched chain monovalent hydrocarbon group containing at least one triple bond.
  • alkynyl is also intended to include those hydrocarbyl groups having one triple bond and one double bond.
  • (C2-C6)alkynyl is intended to include ethynyl, propynyl, and the like.
  • Halogen is fluorine, chlorine, bromine or iodine.
  • Haloalkyl means that the hydrogen atoms in the alkyl group may be replaced by one or more halogen atoms.
  • C 1-4 halogen alkyl refers to an alkyl group containing 1 to 4 carbon atoms in which hydrogen atoms are replaced by one or more halogen atoms.
  • Heterocycle refers to a saturated ring or a non-aromatic unsaturated ring containing at least one heteroatom; where the heteroatom refers to a nitrogen atom, an oxygen atom, or a sulfur atom;
  • Aromatic heterocycle refers to an aromatic unsaturated ring containing at least one heteroatom; where the heteroatom refers to a nitrogen atom, an oxygen atom, or a sulfur atom;
  • Steps include enantiomers and diastereomers
  • the compounds of the present invention may contain asymmetric or chiral centers and thus exist as different stereoisomers. All stereoisomeric forms of the compounds of the present invention, including but not limited to, diastereomers, enantiomers, atropisomers, and mixtures thereof, such as racemic mixtures, constitute the present invention. part. Many organic compounds exist in optically active forms, that is, they have the ability to rotate the plane of plane-polarized light. When describing optically active compounds, the prefixes D, L or R, S are used to indicate the absolute configuration of the chiral center of the molecule. The chemical structures of these stereoisomers are the same, but their three-dimensional structures are different.
  • stereoisomers can be enantiomers, and mixtures of isomers are often called enantiomeric mixtures.
  • a 50:50 mixture of enantiomers is called a racemic mixture or racemate, which can result in no stereoselectivity or stereospecificity during a chemical reaction.
  • racemic mixture and “racemate” refer to an equimolar mixture of two enantiomers that lacks optical activity.
  • pharmaceutically acceptable means a carrier, carrier, diluent, excipient, and/or salt formed that is generally chemically or physically compatible with the other ingredients constituting a pharmaceutical dosage form and is physiologically compatible Compatible with receptors.
  • the pharmaceutical composition of the present invention can be in any reusable pharmaceutical preparation form, such as oral, injection, external use, etc.
  • the oral dosage forms include but are not limited to: tablets, capsules, oral liquids, granules, Pills, suspensions, and injections are selected from water injections and powder injections, and external preparations are selected from patches and ointments. All preparations can be prepared according to conventional pharmaceutical techniques, such as using any of the compounds of the present invention, or its stereoisomers, or its pharmaceutically acceptable salts as pharmaceutical active ingredients, and adding pharmaceutically acceptable salts when necessary.
  • the carrier is prepared into the above-mentioned pharmaceutical dosage form suitable for taking, wherein the unit dose of the active pharmaceutical ingredient can be 0.1 mg-1000 mg, such as each tablet contains 0.1 mg-1000 mg, preferably 5-500 mg of the active pharmaceutical ingredient.
  • salts and “pharmaceutically acceptable salt” refer to the above-mentioned compounds or their stereoisomers, acidic and/or basic salts formed with inorganic and/or organic acids and bases, and also include zwitterionic salts (within salts), also includes quaternary ammonium salts, such as alkylammonium salts. These salts can be obtained directly from the final isolation and purification of the compounds. It can also be obtained by appropriately mixing the above compound, or its stereoisomer, with a certain amount of acid or base (for example, equivalent amounts).
  • salts may form a precipitate in the solution and be collected by filtration, or may be recovered after evaporation of the solvent, or may be obtained by reacting in an aqueous medium and then freeze-drying.
  • the salt mentioned in the present invention can be the hydrochloride, sulfate, citrate, benzenesulfonate, hydrobromide, hydrofluoride, phosphate, acetate, propionate, butylene salt of the compound. Acid, oxalate, malate, succinate, fumarate, maleate acid salt, tartrate or trifluoroacetate.
  • one or more compounds of the invention may be used in combination with each other.
  • the compounds of the present invention can also be optionally used in combination with any other active agent to prepare drugs or pharmaceutical compositions for regulating cell function or treating diseases. If a panel of compounds is used, the compounds can be administered to the subject simultaneously, separately, or sequentially.
  • step 1
  • R P represents a hydrogen atom or a protecting group of an imino group.
  • the protecting group of the imino group of R P benzyl group, p-methoxybenzyl group, tert-butoxycarbonyl group, benzyloxy group is preferred. basecarbonyl etc.
  • the R 1 group in formula B, formula C and formula D refers to the above definition and is the same as the previous definition.
  • reaction In preparation method 1, the compound represented by formula A and the hydrazine derivative represented by formula B are reacted in the presence of a base to obtain the compound represented by formula C.
  • the reaction can usually be carried out in the presence of triethylamine or diisopropylethylamine.
  • the reaction can be carried out in the presence of organic bases such as DIPEA, pyridine, 4-dimethylaminopyridine, or inorganic bases such as sodium hydroxide, potassium hydroxide, sodium carbonate, potassium carbonate, and sodium bicarbonate; for example, dichloromethane, chloroform, It is carried out in inert solvents such as tetrahydrofuran, diethyl ether, benzene, toluene, xylene, dimethylformamide, etc. or mixed solvents. Then, a deprotection reaction is performed, and the compound represented by formula D is formed by cyclization of the compound.
  • organic bases such as DIPEA, pyridine, 4-dimethylaminopyridine
  • inorganic bases such as sodium hydroxide, potassium hydroxide, sodium carbonate, potassium carbonate, and sodium bicarbonate
  • inert solvents such as tetrahydrofuran, diethyl ether, benzene, toluene, x
  • the usage amount of the above-mentioned base is preferably equimolar to excess mole relative to 1 mole of the compound of formula A, more preferably 1 molar amount to 5 molar amount, and most preferably 1 molar amount to 3 molar amount.
  • the base when the base is liquid, the base can be used as both a solvent and a base.
  • the reaction temperature is usually -78°C to 200°C, preferably 20-100°C.
  • the reaction time is usually 5 minutes to 7 days, preferably 8 hours to 96 hours.
  • the compound of formula C is subjected to deprotection and cyclization reaction to obtain the compound of formula D.
  • the reagents are selected from trifluoroacetic acid, hydrochloric acid solution, etc.
  • the solvent is selected from methanol, dichloromethane or 1,4-dioxane, etc.
  • the protecting group is preferably removed by TFA/CH 2 Cl 2 ; if Boc is used as the protecting group, the deprotection reaction can be carried out under standard conditions, for example, dichloromethane/trifluoro It is carried out in an acetic acid system and a saturated hydrogen chloride dioxane solution;
  • the reaction conditions of the cyclization reaction are alkaline conditions, and the alkaline conditions are selected from a certain concentration of sodium hydroxide solution, potassium hydroxide solution, and sodium carbonate solution.
  • the deprotection and cyclization reaction temperature is usually -78°C to 200°C, preferably 20-100°C, and the reaction time is usually 5 minutes to 7 days, preferably 8 hours to 96 hours.
  • the compound represented by formula G can be prepared by step 2-1 or step 2-2.
  • the preparation method of step 2-1 or step 2-2 is as follows:
  • Step 2-1
  • the substituents R 1 and R 2 groups of the compounds described in Formula E and Formula G refer to the above definitions and are the same as the previous definitions.
  • the compound of formula E and formula D are reacted through a CN coupling reaction to prepare the compound of formula G.
  • the reaction solvent is It is carried out in 1,4 dioxane, tetrahydrofuran, diethyl ether, benzene, toluene, xylene, etc. or a mixed solvent.
  • the reaction temperature is 0-200 degrees Celsius, preferably 20-150 degrees Celsius.
  • Step 2-2
  • the substituents R 1 and R 2 of the compounds described in Formula F and Formula G refer to the above definitions, which are the same as the previous definitions.
  • the halogen atoms are F, Cl, Br, and I.
  • the compound represented by formula F and the compound represented by formula D undergo a CN coupling reaction to obtain the compound represented by formula G.
  • the reaction solvent is 1,4 dioxane, tetrahydrofuran, diethyl ether, benzene, toluene, xylene, etc. or a mixed solvent.
  • the reaction temperature is 0-200 degrees Celsius, preferably 20-150 degrees Celsius.
  • the CN coupling reaction in this step is a coupling method commonly used in the art to construct CN bonds, such as Ullmann reaction, Buchwald reaction, preferably Ullmann reaction, more preferably copper iodide/potassium carbonate/N,N-diiso
  • the reaction conditions of propylethylamine DMEDA/1,4-dioxane are CuI, DMEDA, K2CO3, 1,4-dioxane), or more preferably CuI/K 2 CO 3 /N, N'-dimethyl-1,2-cyclohexanediamine, anisole/NaI/microwave, or more preferably CuI/K 2 CO 3 /anisole/NaI/microwave.
  • the compounds R 2 B(OH) 2 and R 2 -halogen atoms in steps 2-1 and 2-2 can be prepared from simple and easily available raw materials by common synthesis methods in the field of organic chemistry.
  • the substituents R 1 , R 2 , R 3 and R 4 of the compounds described in formula H and formula G refer to the above definitions and are the same as the previous definitions.
  • the compound represented by formula G first forms a highly active intermediate sulfoxide under the action of an oxidizing agent, and then undergoes a substitution reaction with the compound represented by formula H to obtain the compound represented by formula I.
  • the reaction solvent is selected from dichloromethane, chloroform, tetrahydrofuran, diethyl ether, benzene, toluene, xylene, dimethylformamide, etc.
  • the oxidizing agent is preferably m-chloroperoxybenzoic acid m- CPBA;
  • the conditions of the substitution reaction are the reaction conditions commonly used for substitution in this field, such as alkaline conditions or acidic conditions.
  • the alkaline conditions are preferably diisopropylethylamine DIPEA.
  • the acidic conditions are preferably trifluoroacetic acid.
  • the reaction temperature is - 20-200°C, preferably 20-150°C, most preferably room temperature.
  • step 3 the substituted aniline compound represented by formula H can be prepared from simple and easily available raw materials by common synthesis methods in the field of organic chemistry.
  • Step 4 If the compound of formula I prepared in step 3 contains a chiral center, those skilled in the art can combine known separation techniques to obtain pure chiral compounds through chromatography or other separation methods. For example, two chiral compounds containing one chiral center can be obtained through SFC separation. If the compound of formula I obtained in step 3 does not contain a chiral center, there is no need to perform the resolution process of step 4.
  • the structure of the compound was determined by nuclear magnetic resonance (NMR) and mass spectrometry (MS). NMR shifts ( ⁇ ) are given in units of 10-6 (ppm). NMR was measured using (Bruker AvanceIII 400 and Bruker Avance 300) nuclear magnetic instruments. The measurement solvents were deuterated dimethyl sulfoxide (DMSO-d6), deuterated chloroform (CDCl 3 ), deuterated methanol (CD3OD), and internal standards. It is tetramethylsilane (TMS).
  • DMSO-d6 deuterated dimethyl sulfoxide
  • CDCl 3 deuterated chloroform
  • CD3OD deuterated methanol
  • TMS tetramethylsilane
  • LC-MS was measured using Shimadzu LC-MS 2020 (ESI).
  • HPLC measurement used a Shimadzu high-pressure liquid chromatograph (Shimadzu LC-20A).
  • MPLC medium pressure preparative chromatography
  • Thin layer chromatography silica gel plates use Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plates. The specifications used for thin layer chromatography separation and purification products are 0.4mm ⁇ 0.5mm.
  • Column chromatography generally uses Yantai Huanghai Silica Gel 200 ⁇ 300 mesh silica gel as the carrier.
  • the known starting materials of the present invention can be synthesized by methods known in the art, or can be purchased from companies such as Anaiji Chemical, Chengdu Kelon Chemical, Shaoyuan Chemical Technology, Bailingwei Technology, etc.
  • THF tetrahydrofuran
  • DIPEA N,N-diisopropylethylamine
  • DCM dichloromethane
  • TFA trifluoroacetic acid
  • m-CPBA m-chloroperoxybenzoic acid
  • DMF dimethylformamide
  • PTSA p-toluenesulfonamide
  • DMSO dimethyl sulfoxide
  • NBS N-bromosuccinimide
  • AIBN azobisisobutyronitrile
  • DMP dimethyl phthalate
  • TBAHS tetrabutylammonium hydrogen sulfate
  • HPLC test conditions are as follows:
  • Step 1 Synthesis of Compounds 1-3:
  • step 3 in Example 3 replace 3-3 in step 3 with 4-8 (40 mg, 190.77 ⁇ mol), and the synthetic method is the same to obtain compound 4-9 (39 mg, 98.62 ⁇ mol, 51.69% yield ), LCMS(ESI + )m/z:396.0[M+H] + .
  • step 2 in Example 3 replace 3-2 in step 2 with 6-2 (141 mg, 679.01 ⁇ mol).
  • the synthetic method is the same to obtain compound 6-3 (150 mg, crude).
  • step 3 in Example 3 replace 3-3 in step 3 with 6-3 (95.39 mg, 429.15 ⁇ mol), and the synthetic method is the same to obtain compound 6-4 (75 mg, 183.15 ⁇ mol, 51.21% product rate), LCMS (ESI + )m/z: 410.4[M+H] + .
  • step 4 in Example 3 replace 3-4 in step 4 with 6-4 (15 mg, 36.63 ⁇ mol), and replace 1-8 with 7-5 (8.94 mg, 43.96 ⁇ mol).
  • the synthesis method is the same. , compound 7 (10.4 mg, 16.21 ⁇ mol, 44.25% yield) was obtained.
  • step 4 in Example 3 replace 3-4 in step 4 with 6-4 (15 mg, 36.63 ⁇ mol), and replace 1-8 with 8-1 (9.02 mg, 43.96 ⁇ mol).
  • the synthesis method is the same.
  • compound 8 (7.4 mg, 12.14 ⁇ mol, 33.15% yield) was obtained.
  • Substrate 14-3 (200 mg, 663.57 ⁇ mol) was added to a dry one-neck bottle and dissolved in DCM (1 mL). TFA (756.61 mg, 6.64 mmol) was added, and the reaction was stirred at room temperature for 0.5 hours, and monitored by LC-MS. After the reaction was completed, the solvent was spin-dried to obtain crude product 14-4 (100 mg, 496.81 ⁇ mol, 74.87% yield), LCMS (ESI+) m/z: 202 [M+H]+.
  • step 5 in Example 2 replace 2-5 in step 5 with 16-1 (40 mg, 0.15 mmol), and the synthetic method is the same to obtain compound 16-2 (30 mg, 73.43 ⁇ mol, 48.83% yield ), LCMS(ESI + )m/z:409[M+H] + .
  • step 5 in Example 2 replace 2-5 in step 5 with 17-1 (70 mg, 250 ⁇ mol), and the synthesis method is the same to obtain compound 17-2 (25 mg, 59.17 ⁇ mol, 23.69% yield) , LCMS(ESI + )m/z:423[M+H] + .
  • reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by preparative high-performance liquid chromatography to obtain compound 20a (1.0 mg, 1.47 ⁇ mol) (20a and 20b are diastereomers, and the product was obtained by direct separation.
  • the structure is written randomly, and the absolute configuration is not determined. It is based on the retention time, the same below).
  • Step 1 Synthesis of Compounds 36a & 36b
  • Step 20 Synthesis of compounds 37a & 37b:
  • reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by preparative high-performance liquid chromatography (eluting with acetonitrile/0.01% ammonium bicarbonate aqueous solution, 60 ml/min) to obtain the compound, which was then purified by supercritical liquid chromatography.
  • Step 1 Synthesis of compounds 38a & 38b:
  • Step 1 Synthesis of Compounds 39a & 39b
  • Step 1 Synthesis of Compounds 41a & 41b
  • Step 1 Synthesis of Compounds 48a & 48b
  • Step 1 Synthesis of Compounds 50a & 50b
  • Step 1 Synthesis of Compounds 51a & 51b
  • Step 1 Synthesis of Compounds 52a & 52b
  • Step 1 Synthesis of compounds 54a & 54b:
  • Step 1 Synthesis of compounds 56a & 56b:
  • Step 1 Synthesis of compounds 57a & 57b:
  • Step 1 Synthesis of compounds 58a & 58b:
  • Step 3 Synthesis of compounds 59a & 59b:
  • Example 62 Synthesis of Compounds 62a and 62b
  • Step 1 Synthesis of Compounds 64a & 64b
  • Step 1 Synthesis of compounds 66a & 66b:
  • Step 1 Synthesis of compounds 68a & 68b:
  • AZD1775 and ZnC3 used in some biological evaluation experiments in this part of the examples are used as controls.
  • the structural information of AZD1775 (CAS No.: 955365-80-7) and ZnC3 (CAS No.: 2376146-48-2) is as follows. :
  • WEE1 (Thermo Fisher, Cat#PR7373A) protein final reaction concentration is 15nM), in compounds of different concentrations, reaction substrate Tracer 178 (Invitrogen, PV5593) and MAb Anti-GST-Eu crypate (Cisbio, 61GSTKLA) were added to a 384-well plate (Corning, cat#3574), centrifuged at 1000 rpm for 1 min, and the 384-well reaction plate was placed on a constant temperature shaker and incubated for 60 min, 25 °C, 300rpm.
  • Tracer 178 and MAb Anti-GST-Eu crypate are configured in buffer (50mM HEPES pH 7.5, 10mM MgCl2, 1mM EGTA, 0.01% Brij-35), and the final reaction concentration of Tracer178 is 50nM, MAb Anti-GST-Eu crypate The final concentration was 2 nM, in which an equal amount of buffer was used for the negative control (minimum signal control) instead of the protein solution.
  • the Wee1 inhibitory activity of the example compounds was tested according to the above method. The test results are shown in Table 1. The IC 50 of each compound was determined and classified as follows according to the instructions:
  • the compound of the present invention has good Wee1 kinase inhibitory activity.
  • DMSO was dissolved to a concentration of 10mM test compound and 10mM reference compound AZD1775, and the compounds were serially diluted in the culture medium for a total of 9 dose points, and 2 parallel replicates were set for each concentration.
  • the cell growth group without compound addition was used as a positive control (maximum signal control), and the culture medium was used as a negative control (minimum signal control).
  • maximum signal control maximum signal control
  • minimum signal control minimum signal control
  • IC 50 of the compound's inhibition of cell activity was calculated through GraphPad Prism 6 using log(inhibitor) vs. response–Variable slope mode fitting.
  • the example compounds were subjected to BxPC3, HT-29 and OVCAR-3 in vitro anti-cell proliferation tests.
  • the test results are shown in Table 2.
  • the IC 50 of each compound was determined and classified as follows according to the instructions:
  • the compounds of the present invention have good inhibitory effect on tumor cell proliferation, and the anti-proliferation activity data of some compounds are equivalent to or better than the control compound AZD1775.
  • the test method is the same as the evaluation of anti-proliferative activity on tumor cells.
  • the test results of the two normal cell lines are shown in Table 3.
  • the compound of the present invention has weak inhibitory activity on the proliferation of normal HUVEC cells and has a certain inhibitory effect on HK2 cells. use. Compared with the control compounds, most of the compounds of the present invention show equivalent or lower proliferation inhibitory activity and have better safety.
  • the reaction is carried out on an incubator and shaking device. Use a row gun to draw 15 ⁇ L/sample of the starting solution and add it to the reaction plate. Shake and mix slightly to start the reaction, use a timer to accurately time and record;
  • Caco-2 cells were purchased from the American Model Tissue Cell Collection (Rockville, MD). The cell culture medium was modified Eagle's medium (MEM) containing 10% inactivated fetal calf serum and 1% non-essential amino acids. Cells were seeded on polycarbonate filters (Cat. No.: 3396) and cultured in a 37°C, 5% CO2 incubator.
  • MEM modified Eagle's medium
  • Cells can be cultured for 21 to 28 days after seeding for transport experiments, and the apparent permeability coefficient (P app ) of Lucifer Yellow is used to characterize and verify the compactness of the cell monolayer.
  • P app apparent permeability coefficient
  • the compound was dissolved in DMSO to prepare a 10mM stock solution, and diluted with Hanks' balanced salt solution (HBSS, Invitrogen, Cat#14025-092) containing 25mM HEPES (pH 7.4) to obtain a working solution. Add 10 ⁇ M working solution of the compound to be tested to the apical side and basolateral side of Caco-2 and incubate at 37°C for 90 minutes.
  • HBSS Hanks' balanced salt solution
  • HEPES pH 7.4
  • the membrane permeability of the compounds of the present invention on the Caco2 model is comparable to that of the control compounds. Overall, the compounds of the present invention and the control compounds have poor cell permeability and have certain efflux properties.
  • the dialysis device and T5 plate were placed in a microplate constant-temperature shaker and incubated for 5 hours (37°C, use 300 rpm or the minimum rotation speed). After the incubation, add 300 ⁇ L acetonitrile (Verapamil-HCl, 4ng/mL), and then add 50 ⁇ L PBS solution. After the dialysis incubation is completed, take a new 96-well deep-well plate.
  • ⁇ Plasma protein binding rate [(Rpe-Rb)/Rpe] ⁇ 100%
  • ⁇ R pe Ratio of peak area of test sample on plasma side to internal standard
  • ⁇ R b Ratio of the peak area of the test sample on the buffer side to the internal standard
  • ⁇ R 5 Ratio of incubator stability sample peak area to internal standard
  • the compound of the present invention has good plasma protein binding ability.
  • Enzymology experiments use the fluorescence produced by the oxidation of substrates by cytochrome P450 to quantitatively detect the inhibition of the enzyme activity of each isoform of CYP450 by small molecule inhibitors.
  • the experiment was performed in a 384-well plate (Corning, Cat#3575), and the reaction buffer used was: 142.86mM Potassium Phosphate, pH 7.4.
  • the components of Solution A used in the experiment are: 26.13mM NADP+ (Sigma-aldrich, Cat#N0505), 65.77mM G6P (J&K, Cat#968161) and 65.42mM MgCl2 (Sigma-aldrich, Cat#M2670).
  • the components of Solution B used in the experiment are: 40U/mL G6PDH (Sigma-aldrich, Cat#G6378).
  • the components of the substrate mixed solution are: 0.05X Solution A, 0.01X Solution B, 50mM Potassium Phosphate, 0.01mM BOMCC/0.01mM EOMCC/0.001mM DBOMF.
  • the reaction system is 50 ⁇ L or 20 ⁇ L respectively, including 3nM CYP3A4 or 120nM CYP2C9, BOMCC substrate mixed solution and different concentrations of test compounds.
  • the reaction system is 20 ⁇ L, including 12.5nM CYP2C19, 80nM CYP2D6 or 1nM CYP1A2, EOMCC substrate mixed solution and different concentrations of test compounds.
  • the reaction system is 50 ⁇ L, including 1.5 nM CYP2C8, DBOMF substrate mixed solution and different concentrations of test compounds.
  • BOMCC/EOMCC Ex430nm/Em480nm, DBOMF Ex490nm/Em520nm After preincubating the compound and enzyme for 10 minutes, add the substrate, and use BMG PHERAStar to read the fluorescence signals in different bands according to different substrates (BOMCC/EOMCC Ex430nm/Em480nm, DBOMF Ex490nm/Em520nm).
  • the reaction interval is 30 seconds or more ( Set according to the actual experimental well number), and the reaction time is 30 minutes.
  • the experimental data were analyzed and processed by GraphPad Prism 6 software to obtain the IC50 value.
  • A.CHO Choinese hamster ovary cells
  • the cell lines used in the patch clamp experiment were 10th generation CHO cells overexpressing hERG potassium channel cDNA.
  • CHO hERG cells are cultured in a culture dish or culture bottle in a 37°C, 5% CO2 incubator. 24-48 hours before the electrophysiological experiment, drop the cells on a round glass slide and culture them in cell culture medium. The cells will be used for experiments after they adhere to the wall.
  • Hygromycin B hygromycin
  • the compound powder is dissolved in the extracellular fluid and undergoes conventional sonication and shaking for 5 to 10 minutes to ensure complete dissolution of the compound.
  • the final concentrations of compounds used for electrophysiological detection were 5 and 20 ⁇ M, and the final concentration of DMSO was 0.1%.
  • the peak value of the tail current is the current value after adding the drug.
  • Cisapride C4740-10mg, Sigma
  • hERG inhibitory activity of some compounds of the present invention is weaker than that of the control compound AZD1775, and their risk of cardiotoxicity is relatively lower.
  • This experiment aims to study the pharmacokinetics of the test compound in the plasma of male ICR mice after a single intravenous and single oral administration.
  • the purpose of this test is to obtain the pharmacokinetics of the test compound in ICR mice (including intravenous and oral)
  • test article-detection, DMPK animal test, and DMPK analysis of this trial were all completed in Chengdu. All tests followed this trial protocol and the relevant SOPs of relevant institutions.
  • Intravenous injection solvent 5% DMSO-10% Solutol-85% HPBCD (20%, W/V)
  • Intravenous injection dosage 1mg/kg, dosage volume 5mL/kg
  • Intragastric administration dosage 10mg/kg, dosage volume 10mL/kg
  • Blood (40-50ul) was collected via orbital venous plexus puncture at each time point at 5 minutes (IV only), 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, and 24 hours after administration to contain the premedicated dose.
  • anticoagulant tubes sprayed with EDTA-K2, centrifuge blood samples at 10,000 rpm for 20 minutes within 1 hour (store on wet ice before and after centrifugation). Take the supernatant liquid, which is plasma, and freeze it in a refrigerator at -20°C or below for LC-MS preparation. /MS analysis.
  • mice The pharmacokinetic evaluation method in mice was used to evaluate the pharmacokinetics of male beagle dogs.
  • Basic information is as follows:
  • Solvent 5% DMSO+5% Solutol+90% (20% HP- ⁇ -CD in saline)
  • Test purpose To evaluate the anti-tumor efficacy of compound 37b and AZD1775 on xenogeneic tumor-bearing (HT-29) nude mice.
  • Test method Use BALB/c nude mice (provided by Vitong Lever), 7-8 weeks old, weighing 19-22g
  • HT-29 cells Prepare and culture HT-29 cells, count them, mix 5 ⁇ 10 6 HT-29 cells in 0.1 mL PBS solution, and inoculate subcutaneously into the right wing of the mouse.
  • grouping was started and drug administration was started.
  • the test indicator is to examine whether tumor growth is inhibited.
  • TGI (%) [1-(Average tumor volume at the end of administration in a certain treatment group - Average tumor volume at the beginning of administration in this treatment group)/(Average tumor volume at the end of administration in the vehicle control group) -The average tumor volume in the vehicle control group at the beginning of administration)] ⁇ 100%.
  • compound 37b of the present invention has comparable tumor inhibitory effects to the control compound AZD1775; at a dose of 120 mg/kg, compound 37b of the present invention has significant tumor growth inhibitory effect.
  • Test method Use BALB/c nude mice (provided by Vitong Lever), 7-8 weeks old, weighing 19-22g
  • BxPC3 cells Prepare and culture BxPC3 cells, count them, mix 1 ⁇ 10 7 BxPC3 cells in 0.1 mL PBS solution, and inoculate subcutaneously into the right wing of the mouse.
  • groups were started and drug administration started.
  • the test indicator is to examine whether tumor growth is inhibited.
  • TGI (%) [1-(Average tumor volume at the end of administration in a certain treatment group - Average tumor volume at the beginning of administration in this treatment group)/(Average tumor volume at the end of administration in the vehicle control group) -The average tumor volume in the vehicle control group at the beginning of administration)] ⁇ 100%.
  • compound 37b of the present invention has better tumor inhibitory effect than the control compound AZD1775; at the dose of 120 mg/kg, compound 37b has significant tumor growth inhibitory effect.
  • Test purpose Evaluation of rat tolerance under single administration of compounds AZD1775 and 37b
  • mice (provided by Vitong Lever), 6-8 weeks old, 180-220 grams.
  • Test method Give rats a single oral dose according to the predetermined dose, and then observe the survival of the rats (the longest continuous observation is 14 days). Please see Table 15 for the test plan and results.
  • Test purpose Rat tolerance performance and compound toxicity evaluation of compounds AZD1775, ZnC3 and 37b under continuous administration
  • mice (provided by Vitong Lever), 6-8 weeks old, 180-220 grams.
  • Test method Rats were administered at the predetermined dose, administered orally once a day, and the survival and weight changes of the rats were followed up (maximum continuous administration for 14 days). Please see Table 16 for the test protocol.
  • the sampling for the toxicokinetics study is as follows: first and last dose sampling; sampling time points: 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours and 24 hours.
  • the compound 37b of the present invention has obvious advantages in drug safety compared with the control compound AZD1775 and the control compound ZnC3.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

本发明涉及WEE1抑制剂及其制备和用途,所述WEE1抑制剂结构为式(I)所示,本发明涉及式(I)化合物、或其立体异构体、或其药学上可接受的盐,及其在制备用来治疗WEE1活性相关的疾病的药物中的用途。

Description

WEE1抑制剂及其制备和用途 技术领域
本发明涉及一类具有抑制WEE1激酶活性的化合物,以及其在治疗由WEE1介导的疾病中的用途。
背景技术
WEE1酪氨酸激酶是细胞周期G2期检查点。细胞周期受到严密调控,当细胞的DNA未受到损伤时,G1期、S期和G 2期检查点促进细胞进入分裂期,保证细胞周期顺利完成。(Clinical Cancer Research,2011,17(13):4200-4207.)细胞周期受细胞周期蛋白依赖激酶(CDKs)调控,CDKs家族有14个丝氨酸/苏氨酸蛋白激酶。CDK的活性被磷酸化和结合不同周期蛋白所调控。细胞从G2期到进入分裂期的转换被CDK1(也被称为CDC2)的磷酸化及其相关周期蛋白B正调控。细胞分裂前,CDK1处于非活性状态,它的第15位酪氨酸被WEE1磷酸化,然后它的第14位苏氨酸被髓鞘转录因子(MYT1)磷酸化。因此,WEE1是细胞周期的负调控因子,通过阻止周期蛋白B和激活的CDK1复合物进入细胞核,负调控细胞从G2期进入分裂期。WEE1在S期和G2期的表达量和活性都提高,在高度磷酸化的M期降低。当细胞进入G2期,没有DNA损伤发生时,polo样蛋白激酶1(PLK1)磷酸化WEE1,通过泛素连接酶复合物将WEE1降解。PLK1也磷酸化并且激活蛋白磷酸酶细胞分裂周期25类似物(CDC25),CDC25通过去磷酸化激活CDK1。活性的CDK1可以和周期蛋白B结合,促进细胞进入分裂期(Molecular&CellularBiology,2012,32(20):4226.)。
当细胞的DNA受到损伤时,G1期、S期和G2期检查延迟细胞进入分裂期,为细胞进入分裂之前修复损伤的DNA争取了时间,从而保证了基因组的完整性。G1期检查点的关键调控因子P53在很多恶性肿瘤细胞中是突变形式。(Proceedings of the National Academy of Sciences of the United States of America,2007,104(10):3753-3758.)P53功能缺陷的肿瘤细胞,在DNA受到损伤时,不能将细胞周期阻滞在G1期,因此更加依赖于G2期检查点。针对DNA损伤,G2期检查点通过平行且互相联系的两条路径抑制CDK1的磷酸化,从而延迟细胞进入分裂期。根据DNA损伤类型,共济失调性毛细血管扩张症变异(ATM)蛋白激酶或者共济失调性毛细血管扩张相关(ATR)蛋白激酶被激活(Oncotarget,2016,7(31):49902-49916.)。
ATM被离子辐射、放射剂和引起双链DNA断裂的试剂激活。ATM磷酸化和激活检查点激酶2(CHK2),CHK2磷酸化细胞分裂周期25C磷酸酶(CDC25C)的Ser216。这就导致CDC25C的向核输出和细胞质分离,从而抑制它的磷酸化活性。抑制CDC25C的活性导致CDK1/CDK2结合周期蛋白B复合物磷酸化被抑制,使CDK1处于失活形式,抑制细胞进入分裂期(MolecularCancer,2014,13(1):72.)。
ATR由广泛的导致单链DNA断裂的基因毒性刺激因素激活的。ATR是负责磷酸化和激活CHK1的主要激酶。与CHK2只能被ATM激活相比,CHK1可以被ATM和ATR激活。CHK1同时磷酸化WEE1和CDC25C,激活WEE1激酶活性,抑制CDC25C的磷酸酶活性。WEE1磷酸化CDK1-结合周期蛋白B,导致细胞周期阻滞在G2期,为DNA修复提供时间(Drug News&Perspectives,2010,23(7):425.)。
WEE1在很多恶性肿瘤中过表达,比如肝癌、乳腺癌、恶性胶质瘤、黑色素瘤、成人和儿童脑瘤。其中一部分肿瘤细胞G1检查点异常,如果抑制WEE1活性会导致G2期检查点故障,此时带有没有修复的损伤DNA的细胞会持续进行分裂,最终分裂致死。(Molecular Cancer Therapeutics,2013,12(12):2675-2684.)无论通过嘧啶衍生物(PD0166285)或者小干扰RNA敲低方式,抑制WEE1的活性都会使卵巢癌、结肠癌、宫颈癌、骨肉瘤、恶性胶质瘤和肺癌细胞对放射和拓扑异构酶抑制产生的DNA损伤更敏感。因此,WEE1抑制剂单药和联合用药 都具有广阔的发展空间(Cancer Biology&Therapy,2010,9(7):523-525.)。
专利WO2007126122,WO2008133866,WO2013012681,WO2013126656,WO2014167347,WO2015092431,WO2018011569,WO2018011570,WO2018090939,WO2018133829,WO2018171633等中描述了具有WEE1激酶抑制活性的小分子化合物。目前最领先的化合物是AZD1775,已经进入临床II期试验,显示出良好的癌症治疗效果。
发明内容
本发明的目的是提供了一种如式I所示的化合物、或其立体异构体、或其药学上可接受的盐:

其中,
所述的R1选自-C1~6烷基、-C2~6烯基、-C2~6炔基、-C0~2亚烷基-CN、-C0~2亚烷基-(3~10元环烷基)、-C0~2亚烷基-(3~10元杂环烷基);
R2选自
所述的X选自O、NH或CH2
所述的X1选自CH或N;
R21、R22、R29分别独立地选自氢、氘、卤素、氰基、硝基、-OH、-C1~6烷基、卤素取代的C1~6烷基、-C0~2亚烷基-OH、-O(C1~6烷基)、-O(卤素取代的C1~6烷基)、-NH2、-C0~2亚烷基-NH(C1~6烷基)、-C0~2亚烷基-N(C1~6烷基)(C1~6烷基)、-C0~2亚烷基-(3~10元环烷基)、-C0~2亚烷基-(3~10元杂环烷基);
所述的R23、R24与其直接相连的原子一起形成3~10元碳环、3~10元杂环;
所述的R25、R26与其直接相连的原子一起形成3~10元碳环、3~10元杂环;
所述的R27、R28与其直接相连的原子一起形成3~10元碳环、3~10元杂环;
R3选自氢、氘、卤素、氰基、硝基、-C1~6烷基、卤素取代的C1~6烷基、-C0~2亚烷基-OH、-O(C1~6烷基)、-O(卤素取代的C1~6烷基)、-NH2、-C0~2亚烷基-NH(C1~6烷基)、-C0~2亚烷基-N(C1~6烷基)(C1~6烷基);
所述的R4选自3~12元杂环烷基;所述的杂环烷基可进一步被一个、两个、三个或四个独立的R41取代;
所述的R41选自氢、卤素、氰基、硝基、-OH、-C1~6烷基、卤素取代的C1~6烷基、-C0~2亚烷基-OH、-O(C1~6烷基)、-O(卤素取代的C1~6烷基)、-NH2、-C0~2亚烷基-NH(C1~6烷基)、-C0~2亚烷基-N(C1~6烷基)(C1~6烷基)、-C(O)C1~6烷基、3~10元碳环、3~10元杂环;所述碳环、杂环可进一步被一个、两个、三个或四个独立的R31取代;
或者,所述的R3、R4与其直接相连的原子一起形成3~10元碳环、3~10元杂环;所述的碳环、杂环烷基可进一步被一个、两个、三个或四个独立的R31取代;
所述的R31选自氢、卤素、氰基、硝基、-OH、-C1~6烷基、卤素取代的C1~6烷基、-C0~2亚烷基-OH、-O(C1~6烷基)、-O(卤素取代的C1~6烷基)、-NH2、-C0~2亚烷基-NH(C1~6烷基)、-C0~2亚烷基-N(C1~6烷基)(C1~6烷基)。
优选的,本发明所述的化合物、或其立体异构体、或其药学上可接受的盐,所述的R1选自甲基、乙基、
优选的,本发明所述的化合物、或其立体异构体、或其药学上可接受的盐,R21、R22、R29分别独立地选自氢、氘、氰基、甲基、乙基、-OH、三氟甲基、环丙基、-CH2OH、-NH2
优选的,本发明所述的化合物、或其立体异构体、或其药学上可接受的盐,所述的R23、R24与其直接相连的原子一起形成环丙基、环丁基、环戊基;
所述的R25、R26与其直接相连的原子一起形成环丙基、环丁基、环戊基;
所述的R27、R28与其直接相连的原子一起形成环丙基、环丁基、环戊基。
优选的,本发明所述的化合物、或其立体异构体、或其药学上可接受的盐,所述的R2选自
优选的,本发明所述的化合物、或其立体异构体、或其药学上可接受的盐,所述的R3选自氢、氟、甲基、-CH2OH、甲氧基。
优选的,本发明所述的化合物、或其立体异构体、或其药学上可接受的盐,所述的R4选自含氮6元杂环、7元含氮桥环、8元含氮桥环、9元含氮杂螺环、11元含氮杂螺环。
进一步地:所述的R4选自
进一步地:所述的R4选自
优选的,本发明所述的化合物、或其立体异构体、或其药学上可接受的盐,所述的R3、R4与其直接相连的原子一起形成6元含氮杂环。
更优选的,本发明所述的化合物、或其立体异构体、或其药学上可接受的盐,所述的R3、R4与其直接相连的原子一起形成
进一步地:所述的R31选自甲基。
优选的,本发明所述的化合物、或其立体异构体、或其药学上可接受的盐,式I所述的化合物具体为:






























更优选,本发明的化合物、或其立体异构体、或其药学上可接受的盐,式I所述的化合物具体为:
本发明还提供一种上述任一项所述的化合物、或其立体异构体、或其药学上可接受的盐 在制备治疗WEE1介导的疾病的药物中的用途。
所述WEE1介导的疾病是与炎症、自身免疫性疾病、感染性疾病、癌症、癌前期综合征相关的疾病中的一种或几种。
本发明还提供一种药物组合物,它是以上述任一项所述的化合物、或其立体异构体、或其药学上可接受的盐,加上药学上可接受的辅料制备而成的制剂。
以下为对本发明的名词术语的解释和说明:
“癌症”或“恶性肿瘤”是指以不受控制的细胞异常增殖为特征的多种疾病中的任何一种,受影响的细胞在局部或通过血流和淋巴系统扩散到其他部位的能力的身体(即转移)以及许多特征结构和/或分子特征中的任何一个。“癌细胞”是指经历多步骤肿瘤进展的早期,中期或晚期阶段的细胞。癌症包括肉瘤、乳腺癌、肺癌、脑癌、骨癌、肝癌、肾癌、结肠癌和前列腺癌。在一些实施方案中,式I的化合物用于治疗选自结肠癌、脑癌、乳腺癌、纤维肉瘤和鳞状细胞癌的癌症。在一些实施方案中,癌症选自黑素瘤、乳腺癌、结肠癌、肺癌和卵巢癌。在一些实施方案中,所治疗的癌症是转移性癌症。
自身免疫性疾病是由身体对体内正常存在的物质和组织的免疫反应引起的。自身免疫疾病的例子包括心肌炎、狼疮性肾炎、原发性胆汁性肝硬化、牛皮癣、I型糖尿病、格雷夫氏病、腹腔疾病、克罗恩病、自身免疫性中性白细胞减少症、幼年型关节炎、类风湿性关节炎、纤维肌痛、吉兰巴利综合征、多发性硬化症和自身免疫性视网膜病变。本发明的一些实施方案涉及治疗自身免疫疾病如牛皮癣或多发性硬化症。
炎症疾病包括以组织病理性炎症为特征的多种病症。炎性疾病的例子包括寻常性痤疮、哮喘、腹腔疾病、慢性前列腺炎、肾小球性肾炎、炎症性肠病、盆腔炎、再灌注损伤、类风湿性关节炎、结节病、血管炎、房尘螨引起的气道炎症和间质性膀胱炎。炎性疾病与自身免疫性疾病之间存在显著重叠。本发明的一些实施方案涉及炎性疾病哮喘的治疗。免疫系统通常涉及炎症性疾病,在过敏反应和一些肌病中都有表现,许多免疫系统疾病导致异常炎症。IL-17A介导的疾病也包括自身免疫性炎症性疾病。
本发明中提供的化合物和衍生物可以根据IUPAC(国际纯粹与应用化学联合会)或CAS(化学文摘服务社,Columbus,OH)命名系统命名。
关于本发明的使用术语的定义:除非另有说明,本文中基团或者术语提供的初始定义适用于整篇说明书的该基团或者术语;对于本文没有具体定义的术语,应该根据公开内容和上下文,给出本领域技术人员能够给予它们的含义。
“取代”是指分子中的氢原子被其它不同的原子或分子所替换。
碳氢基团中碳原子含量的最小值和最大值通过前缀表示,例如,前缀Ca~b烷基表明任何含“a”至“b”个碳原子的烷基。因此,例如,“C1~4烷基”是指包含1~4个碳原子的烷基。
“烷基”是指具有指定数目的成员原子的饱和烃链。例如,C1~6烷基是指具有1至6个成员原子,例如1至4个成员原子的烷基基团。烷基基团可以是直链或支链的。代表性的支链烷基基团具有一个、两个或三个支链。烷基基团可任选地被一个或多个如本文所定义的取代基取代。烷基包括甲基、乙基、丙基(正丙基和异丙基)、丁基(正丁基、异丁基和叔丁基)、戊基(正戊基、异戊基和新戊基)和己基。烷基基团也可以是其他基团的一部分,所述其他基团为例如C1~C6烷氧基。
“环烷基”、“环烷烃”是指具有碳原子且没有环杂原子且具有单个环或多个环(包括稠合、并和、桥环)的饱和或部分饱和的环状基团。对于具有不含环杂原子的芳族和非芳族环的多环体系,当连接点位于非芳族碳原子时,适用术语“环烷基”(例如5,6,7,8,-四氢化萘-5-基)。术语“环烷基”包括环烯基基团,诸如环己烯基。环烷基基团的实例包括例如,金刚烷基、环丙基、环丁基、环己基、环戊基、环辛基、环戊烯基和环己烯基。包括多双环烷基环 体系的环烷基基团的实例是双环己基、双环戊基、双环辛基等。例如
“烯基”是指具有2至10个碳原子和在一些实施方案中2至6个碳原子或2至4个碳原子且具有至少1个乙烯基不饱和位点(>C=C<)的直链或支链烃基基团。例如,(Ca-Cb)烯基是指具有a至b个碳原子的烯基基团并且意在包括例如乙烯基、丙烯基、异丙烯基、1,3-丁二烯基等。
“炔基”是指含有至少一个三键的直链一价烃基或支链一价烃基。术语“炔基”还意在包括具有一个三键和一个双键的那些烃基基团。例如,(C2-C6)炔基意在包括乙炔基、丙炔基等。
“卤素”为氟、氯、溴或碘。
“卤素烷基”指烷基中的氢原子可被一个或多个卤素原子取代。例如C1~4卤素烷基指氢原子被一个或多个卤素原子取代的包含1~4个碳原子的烷基。
“杂环”、“杂环烷基”、“杂环烷烃”指包含至少一个杂原子的饱和环或非芳香性的不饱和环;其中杂原子指氮原子、氧原子、硫原子;
“芳杂环”指包含至少一个杂原子的芳香性不饱和环;其中杂原子指氮原子、氧原子、硫原子;
“立体异构体”包括对映异构体和非对映异构体;
本发明的化合物可以包含不对称中心或手性中心,因此存在不同的立体异构体。本发明的化合物所有的立体异构形式,包括但绝不限于,非对映体,对映异构体,阻转异构体,和它们的混合物,如外消旋混合物,组成了本发明的一部分。很多有机化合物都以光学活性形式存在,即它们有能力旋转平面偏振光的平面。在描述光学活性化合物时,前缀D、L或R、S用来表示分子手性中心的绝对构型。这些立体异构体的化学结构是相同的,但是它们的立体结构不一样。特定的立体异构体可以是对映体,异构体的混合物通常称为对映异构体混合物。50:50的对映体混合物被称为外消旋混合物或外消旋体,这可能导致化学反应过程中没有立体选择性或立体定向性。术语“外消旋混合物”和“外消旋体”是指等摩尔的两个对映异构体的混合物,缺乏光学活性。
术语“药学上可接受的”是指某载体、运载物、稀释剂、辅料,和/或所形成的盐通常在化学上或物理上与构成某药物剂型的其它成分相兼容,并在生理上与受体相兼容。
本发明所述药物组合物,可以是任意一种可复用的药物制剂形式,如:口服,注射,外用等形式,口服剂型包括但不限于:片剂,胶囊剂,口服液,颗粒剂,丸剂,混悬剂,注射剂选自水针,粉针,外用制剂选自贴剂,膏剂。所有制剂均可以按照制剂学常规技术制备,如以本发明化合物,或其立体异构体、或其药学上可接受的盐中的任意一项作为药物活性成份,必要时加入药学上可接受的载体,制备成适于服用的上述药物剂型,其中,单位剂量的药物活性成份可以是0.1mg-1000mg,如片剂的每片含有0.1mg-1000mg优选5-500mg的药物活性成份。
术语“盐”和“可药用的盐”是指上述化合物或其立体异构体,与无机和/或有机酸和碱形成的酸式和/或碱式盐,也包括两性离子盐(内盐),还包括季铵盐,例如烷基铵盐。这些盐可以是在化合物的最后分离和纯化中直接得到。也可以是通过将上述化合物,或其立体异构体,与一定数量的酸或碱适当(例如等当量)进行混合而得到。这些盐可能在溶液中形成沉淀而以过滤方法收集,或在溶剂蒸发后回收而得到,或在水介质中反应后冷冻干燥制得。本发明中所述盐可以是化合物的盐酸盐、硫酸盐、枸橼酸盐、苯磺酸盐、氢溴酸盐、氢氟酸盐、磷酸盐、乙酸盐、丙酸盐、丁二酸盐、草酸盐、苹果酸盐、琥珀酸盐、富马酸盐、马来 酸盐、酒石酸盐或三氟乙酸盐。
在某些实施方式中,本发明的一种或多种化合物可以彼此联合使用。也可选择将本发明的化合物与任何其它的活性试剂结合使用,用于制备调控细胞功能或治疗疾病的药物或药物组合物。如果使用的是一组化合物,则可将这些化合物同时、分别或有序地对受试对象进行给药。
对本发明的化合物的制备方法进行说明,具体步骤如下:
步骤1:
式B、式C中RP表示氢原子或亚氨基的保护基团,作为RP的亚氨基的保护基团,优选为苄基、对甲氧基苄基、叔丁氧基羰基、苄氧基羰基等。式B、式C和式D中的R1基团参见上述的定义,与前文的定义相同。
在制备方法1中,使式A所示化合物与式B所示的肼衍生物在碱的存在下反应得到式C所示化合物,反应通常可以在三乙基胺、二异丙基乙基胺DIPEA、吡啶、4-二甲基氨基吡啶等有机碱或氢氧化钠、氢氧化钾、碳酸钠、碳酸钾、碳酸氢钠等无机碱的存在下进行反应;例如可以在二氯甲烷、氯仿、四氢呋喃、乙醚、苯、甲苯、二甲苯、二甲基甲酰胺等或混合溶剂等惰性溶剂中进行。然后进行脱保护反应,通过该化合物环化形成式D所示的化合物。上述碱的用量相对于式A化合物1摩尔,优选为等摩尔~过量摩尔,更优选为1摩尔量-5摩尔量,最优选为1摩尔-3摩尔。此外,该碱为液体时,可以将该碱兼用作溶剂和碱。反应温度通常为-78℃-200℃,优选为20-100℃。反应时间通常为5分钟-7天,优选为8小时-96小时。
在制备方法1中,使式C化合物进行脱保护和环化反应得到式D所示的化合物,其中脱保护反应中反应试剂选自三氟乙酸、盐酸溶液等、溶剂选择甲醇、二氯甲烷或1,4-二氧六环等,优选TFA/CH2Cl2的方式脱除保护基;如果使用Boc作为保护基时,脱保护反应可以在标准条件下进行,例如,二氯甲烷/三氟乙酸体系、饱和的氯化氢二氧六环溶液中进行;其中所述的环化反应的反应条件为碱性条件,碱性条件选自一定浓度的氢氧化钠溶液、氢氧化钾溶液、碳酸钠溶液、碳酸钾溶液或碳酸氢钠溶液等,优选为氢氧化钠溶液。脱保护和环化反应温度通常为-78℃-200℃,优选为20-100℃,反应时间通常为5分钟-7天,优选为8小时-96小时。
步骤2:
式G所示的化合物可以通过步骤2-1或步骤2-2的方式制备得到,步骤2-1或步骤2-2的制备方法如下:
步骤2-1:
式E和式G所述化合物的取代基R1和R2基团参见上述的定义,与前文的定义相同。该反应中通过C-N偶联反应将式E和式D化合物反应制备得到式G所示化合物。反应溶剂为 1,4二氧六环、四氢呋喃、乙醚、苯、甲苯、二甲苯等或混合溶剂中进行,反应温度为0-200摄氏度,优选为20-150℃。
步骤2-2:
式F和式G所述化合物的取代基R1和R2基团参见上述的定义,与前文的定义相同,卤原子为F、Cl、Br、I。式F所示化合物与式D所示化合物经C-N偶联反应得到式G所示化合物,反应溶剂为1,4二氧六环、四氢呋喃、乙醚、苯、甲苯、二甲苯等或混合溶剂中进行,反应温度为0-200摄氏度,优选为20-150℃。该步骤中所述C-N偶联反应为本领域惯常用于构建C-N键的偶联方法,例如Ullmann反应,Buchwald反应,优选Ullmann反应,更优选碘化亚铜/碳酸钾/N,N-二异丙基乙胺DMEDA/1,4-二氧六环的反应条件(偶联反应的条件为CuI,DMEDA,K2CO3,1,4-dioxane),或更优选为CuI/K2CO3/N,N'-二甲基-1,2-环己二胺、苯甲醚/NaI/微波,或更优选为CuI/K2CO3/苯甲醚/NaI/微波。
步骤2-1和2-2中化合物R2B(OH)2和R2-卤原子可以通过有机化学领域中通常的合成方法以简单易得的原料制备得到。
步骤3:
式H和式G所述化合物的取代基R1、R2、R3、R4基团参见上述的定义,与前文的定义相同。式G所示化合物先在氧化剂的作用下形成活性较高的中间体亚砜,然后再与式H所示化合物进行取代反应得到式I所示化合物。其中反应溶剂选自二氯甲烷、氯仿、四氢呋喃、乙醚、苯、甲苯、二甲苯、二甲基甲酰胺等或其混合溶剂;在氧化反应中所述氧化剂优选为间氯过氧苯甲酸m-CPBA;所述取代反应的条件为本领域惯常用于取代的反应条件,例如碱性条件或酸性条件,碱性条件优选二异丙基乙胺DIPEA,酸性条件优选三氟乙酸,反应温度为-20-200℃,优选为20-150℃,最优选为室温。
在步骤3中式H所示取代苯胺类化合物可以通过有机化学领域中通常的合成方法以简单易得的原料制备得到。
步骤4:如若步骤3中制备得到式I化合物含有手性中心,本领域技术人员可结合已知的分离技术通过色谱方法或其他拆分方法得到纯手性化合物。例如可通过SFC拆分得到含有一个手性中心的两个手性化合物。如果步骤3中得到的式I化合物不含有手性中心,就不需要进行步骤4的拆分过程。
显然,根据本发明的上述内容,按照本领域的普通技术知识和惯用手段,在不脱离本发明上述基本技术思想前提下,还可以做出其它多种形式的修改、替换或变更。
以下通过实施例形式的具体实施方式,对本发明的上述内容再作进一步的详细说明。但不应将此理解为本发明上述主题的范围仅限于以下的实例。凡基于本发明上述内容所实现的技术均属于本发明的范围。
具体实施方式
化合物的结构是通过核磁共振(NMR)和质谱(MS)来确定的。NMR位移(δ)以10-6(ppm)的单位给出。NMR的测定是用(Bruker AvanceIII 400和Bruker Avance 300)核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代氯仿(CDCl3),氘代甲醇(CD3OD),内标为四甲基硅烷(TMS)。
LC-MS的测定使用岛津液质联用仪(Shimadzu LC-MS 2020(ESI))。HPLC的测定使用岛津高压液相色谱仪(Shimadzu LC-20A)。MPLC(中压制备色谱)使用Gilson GX-281反相制备色谱仪。薄层层析硅胶板用烟台黄海HSGF254或青岛GF254硅胶板,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。柱层析一般使用烟台黄海硅胶200~300目硅胶为载体。
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买于安耐吉化学、成都科龙化工、韶远化学科技、百灵威科技等公司。
实施例中无特殊说明,反应在氮气氛围下进行。实施例中无特殊说明,溶液是指水溶液。实施例中无特殊说明,反应的温度为室温。实施例中无特殊说明,M是摩尔每升。
THF:四氢呋喃;DIPEA:N,N-二异丙基乙胺;
DCM:二氯甲烷;TFA:三氟乙酸;m-CPBA:间氯过氧苯甲酸;
DMF:二甲基甲酰胺;PTSA:对甲苯磺酰胺;
DMSO:二甲基亚砜;NBS:N-溴代琥珀酰亚胺;AIBN:偶氮二异丁腈;
DMP:邻苯二甲酸二甲酯;TBAHS:四丁基硫酸氢铵;
BMS:4-苯甲酰基-4"-甲基-二苯硫醚;
实施例中无特殊说明时,HPLC测试条件如下:
method A:
柱规格:Boston Green C18 150mm*4.6mm 5um;流动相:A:0.05%TFA Water B:0.05%TFA Acétonitrile;梯度:B from 5%to 95%in 10.0min and hold 95%for 5.0min;流速:1.5mL/min;柱内温度:40℃.
method B:
柱规格:Boston Green ODS 150mm*4.6mm 5um;流动相:A:0.01M NH4HCO3Water B:Acétonitrile;梯度:B from 5%to 95%in 10.0min and hold 95%for 5.0min;流速:1.5mL/min;柱内温度:40℃.
实施例中无特殊说明时,SFC测试条件如下:
柱规格:3um,150mm*3mm;流动相:A:CO2,流动相B:Alcohols solvent;流速:1mL/min柱内温度:40℃
中间体实施例1:中间体IM-1的合成
步骤1:化合物IM-1-3的合成:
在干燥的单口瓶中加入底物IM-1-2(8.15g,35mmol)和THF(50mL),搅拌溶解后加入IM-1-1(6.3g,37mol)和DIPEA(15mL,75mol),加热至110℃反应,LC-MS监测。反应结束后,待体系恢复至室温,析出固体后滤出,烘箱烘干,即为粗产物IM-1-3(9.87g,76.6%产率),LCMS(ESI+)m/z:369.2[M+H]+
步骤2:化合物IM-1的合成:
在干燥的单口瓶中加入底物IM-1-3(9.16g,27.5mmol)和DCM(18mL),搅拌溶解后,缓慢加入TFA(18mL),加热至75℃反应,LC-MS监测。反应结束后有机溶剂减压浓缩,用乙醇溶解后,加入6M NaOH溶液室温搅拌,LC-MS监测。反应完成后,减压浓缩后有固体析出,滤出固体,用水洗涤三次,冷的乙醇洗涤三次,室温晾干,即为粗产物IM-1(5g,81.7%产率),LCMS(ESI+)m/z:223.1[M+H]+
实施例1:化合物1的合成
步骤1:化合物1-3的合成:
在干燥的单口瓶中加入底物1-1(1g,4.72mmol)和THF(10mL),搅拌溶解后在-40℃和氮气保护下缓慢滴加n-BuLi(2.5M,4.15mL),在-40℃下搅拌反应1小时后滴加1-2(2.66g,14.15mmol),滴加完成后恢复到室温反应12小时,LC-MS监测。反应结束后,加入4N HCl(5mL),旋干溶剂后加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,得到产物1-3(1.2g,crude),LCMS(ESI+)m/z:238/240[M+H]+
步骤2:化合物1-4的合成:
在干燥的单口瓶中加入底物1-3(700mg,2.94mmol)和DMF(3mL),搅拌溶解后加入碘乙烷(458.57mg,2.94mmol)和碳酸铯(1.05g,3.23mmol),在75℃下搅拌反应8小时,LC-MS监测。反应结束后,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物1-4(285mg,1.07mmol,36.42%产率),LCMS(ESI+)m/z:266[M+H]+
步骤3:化合物1-5的合成:
在干燥的单口瓶中加入底物1-4(155mg,582.41μmol)、B(pin)2(325.38mg,1.28mmol)、Pd(dppf)Cl2(42.73mg,58.24μmol)和碳酸钾(241.47mg,1.75mmol)用1,4-二氧六环(5mL),在氮气保护下,升温至110℃下搅拌反应12小时,LC-MS监测。反应结束后,旋干溶剂,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物1-5(80mg,255.43μmol,43.86%产率),LCMS(ESI+)m/z:314[M+H]+
步骤4:化合物1-6的合成:
在干燥的单口瓶中加入底物1-5(57mg,181.99μmol)用ACN(5mL)溶解,加入HCL(6M,1mL),升温至60℃,搅拌反应6小时,LC-MS监测。反应结束后,旋干溶剂,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物1-6(24mg,103.87μmol,57.07%产率),LCMS(ESI+)m/z:232[M+H]+
步骤5:化合物1-7的合成:
在干燥的单口瓶中加入底物1-6(24mg,103.87μmol)用DCM(3mL)溶解,加入IM-1 (26.93mg,121.18μmol),醋酸铜(4.52mg,24.86μmol)和吡啶(9.59mg,121.18μmol,9.76μL),室温搅拌反应72小时,LC-MS监测。反应结束后,旋干溶剂,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物1-7(19mg,46.63μmol,38.48%产率),LCMS(ESI+)m/z:408[M+H]+
步骤6:化合物1的合成:
在干燥的单口瓶中加入底物1-7(19mg,46.63μmol)用THF(1.5mL)溶解,加入m-CPBA(14.48mg,83.93μmol),室温搅拌反应0.5小时后加入DIPEA(30.13mg,233.14μmol,40.61μL),和1-8(10.70mg,55.95μmol),继续室温搅拌12小时,LC-MS监测。反应结束后,旋干溶剂,用反相柱层析纯化得到产物1(3.1mg,5.63μmol,12.07%产率)。1H NMR(600MHz,DMSO-d6)δ10.03(s,1H),8.81(s,1H),7.52(s,2H),7.21(s,2H),7.11–7.09(m,1H),6.83(d,J=6.0Hz,2H),5.70–5.67(m,1H),5.09(d,J=10.2Hz,1H),4.97(d,J=17.4Hz,1H),4.28(s,2H),3.82–3.78(m,2H),3.06(s,4H),2.47(s,4H),2.23(s,3H),1.69(d,J=3.6Hz,2H),1.58(d,J=3.0Hz,2H),1.15(t,J=6.6Hz,3H).LCMS(ESI+)m/z:551.2[M+H]+,HPLC method B:RT=5.65min,purity:100%。
实施例2化合物2的合成
步骤1:化合物2-2的合成:
在干燥的单口瓶中加入底物2-1(870mg,4.42mmol)和DMF(8mL),搅拌溶解后加入碘乙烷(1.38g,8.83mmol)和碳酸钾(1.22g,8.83mmol),在75℃下搅拌反应4小时,LC-MS监测。反应结束后,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物2-2(600mg,2.67mmol,60.37%产率),LCMS(ESI+)m/z:225[M+H]+
步骤2:化合物2-3的合成:
在干燥的单口瓶中加入底物2-2(600mg,2.67mmol)和1,4-二氧六环(10mL),搅拌溶解后加入三溴吡啶(3.41g,10.66mmol),在室温下搅拌反应16小时,LC-MS监测。反应结束后,旋干溶剂,加入水和DCM萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物2-3(230mg,576.62μmol,21.63%产率),LCMS(ESI+)m/z:399[M+H]+
步骤3:化合物2-4的合成:
在干燥的单口瓶中加入底物2-3(230mg,576.62μmol)和THF(1.5mL),搅拌溶解后加入饱和氯化铵溶液(1.5mL)和锌粉(753.99mg,11.53mmol),在室温下搅拌反应10分钟,LC-MS监测。反应结束后,过滤,旋干溶剂,用反相柱层析纯化得到产物2-4(44mg,182.51μmol,31.65%产率),LCMS(ESI+)m/z:241[M+H]+
步骤4:化合物2-5的合成:
在干燥的单口瓶中加入底物2-4(20mg,82.96μmol)和DMF(1mL),搅拌溶解后加入 NaH(11.95mg,497.75μmol),在0℃下搅拌反应0.5小时后,加入1,3-二碘丙烷(73.64mg,248.88μmol),在室温下搅拌反应0.5小时,LC-MS监测。反应结束后,加入水淬灭反应,用EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物2-5(10mg,35.57μmol,42.87%产率),LCMS(ESI+)m/z:281[M+H]+
步骤5:化合物2-6的合成:
在干燥的单口瓶中加入底物2-5(10mg,35.57μmol)和1,4-二氧六环(1mL),搅拌溶解后加入IM-1(10mg,44.99μmol)、CuI(15.58mg,81.80μmol)、K2CO3(7.91mg,57.26μmol)和DMEDA(14.42mg,163.60μmol),在氮气保护下升温至110℃,搅拌反应3小时,LC-MS监测。反应结束后,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物2-6(5mg,11.83μmol,28.93%产率),LCMS(ESI+)m/z:423[M+H]+
步骤6:化合物2的合成:
在干燥的单口瓶中加入底物2-6(5.0mg,11.86μmol)用THF(1mL)溶解,加入m-CPBA(3.68mg,21.35μmol),室温搅拌反应0.5小时后加入DIPEA(7.67mg,59.31μmol,10.33μL),和1-8(2.72mg,14.23μmol),继续室温搅拌8小时,LC-MS监测。反应结束后,旋干溶剂,用反相柱层析纯化得到产物2(2.1mg,3.71μmol,31.30%产率)。1H NMR(600MHz,CDCL3)δ8.77(s,1H),7.77(d,J=7.8Hz,1H),7.44(d,J=4.8Hz,2H),7.36(d,J=7.8Hz,1H),6.86(d,J=9.0Hz,2H),5.67–5.62(m,1H),5.00(d,J=9.6Hz,1H),4.92(d,J=16.8Hz,1H),4.65(d,J=5.4Hz,2H),3.75(q,J=14.4Hz,2H),3.40(s,4H),2.93–2.87(m,2H),2.64–2.61(m,4H),2.37–2.28(m,4H),2.23–2.18(m,1H),1.57(s,2H),1.21(t,J=7.2Hz,3H).LCMS(ESI+)m/z:566.2[M+H]+,HPLC method B:RT=6.03min,purity:85.9%。
实施例3:化合物3的合成
步骤1:化合物3-2的合成:
在干燥的单口瓶中加入底物NaH(166.08mg,4.15mmol,60%purity)和DMF(20mL),搅拌溶解后在0℃和氮气保护下加入1,2-二溴乙烷(780mg,4.15mmol)和3-1(200mg,1.19mmol)的DMF(10mL)溶液,在0℃下搅拌反应1小时后室温反应2小时,LC-MS监测。反应结束后,加入饱和氯化铵溶液和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物3-2(35mg,180.76μmol,15.24%产率),LCMS(ESI+)m/z:194.2[M+H]+
步骤2:化合物3-3的合成:
在干燥的单口瓶中加入底物3-2(33mg,170.43μmol)和THF(5mL),搅拌溶解后在0℃和氮气保护下滴加加入甲基溴化镁(2M,340.86μL),在0℃下搅拌反应1小时,LC-MS监测。反应结束后,加入饱和氯化铵溶液和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤 旋干,用反相柱层析纯化得到产物3-3(33mg,157.39μmol,92.35%产率),LCMS(ESI+)m/z:210.1[M+H]+
步骤3:化合物3-4的合成:
在干燥的单口瓶中加入底物3-3(33mg,157.39μmol)和苯甲醚(2mL),搅拌溶解后加入IM-1(34.98mg,157.39μmol)、CuI(59.95mg,314.78μmol)、K2CO3(54.38mg,393.47μmol)、N,N'-二甲基-1,2-环己二胺(89.55mg,629.56μmol)和NaI(47.18mg,314.78μmol),微波130℃,搅拌反应4小时,LC-MS监测。反应结束后,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物3-4(38mg,96.09μmol,61.05%产率),LCMS(ESI+)m/z:396.1[M+H]+
步骤4:化合物3的合成:
在干燥的单口瓶中加入底物3-4(38mg,96.09μmol)用THF(4mL)溶解,加入m-CPBA(35.11mg,172.96μmol,85%purity),室温搅拌反应1小时后加入DIPEA(124.18mg,960.86μmol,167.36μL),和1-8(22.05mg,115.30μmol),升温至50℃搅拌5小时,LC-MS监测。反应结束后,旋干溶剂,用反相柱层析纯化得到产物3(22.6mg,40.24μmol,41.88%产率)。1H NMR(600MHz,DMSO-d6)δ10.14(s,1H),8.82(s,1H),7.92(s,1H),7.70(d,J=8.4Hz,1H),7.59(s,2H),6.92(d,J=8.4Hz,2H),5.72–5.76(m,1H),5.02(s,1H),5.01(d,J=10.2Hz,1H),4.88(d,J=17.4Hz,1H),4.82–4.68(m,1H),4.66–4.52(m,1H),3.09(t,J=4.8Hz,4H),2.93–2.86(m,2H),2.46(t,J=4.8Hz,4H),2.23(s,3H),1.22(s,3H),0.94–0.91(m,1H),0.70–0.67(m,1H),0.60–0.56(m,1H),0.48–0.45(m,1H).LCMS(ESI+)m/z:539.3[M+H]+,HPLC method B:RT=6.92min,purity:95.9%。
化合物3通过SFC拆分得到以下两个化合物:3a RT=2.51min;3b RT=3.51min(如无特殊说明,化合物的手性此处只进行分子区分,不代表确切的绝对构型,下同)。
3a结构确证:1H NMR(400MHz,DMSO-d6)δ10.13(s,1H),8.83(s,1H),7.92(d,J=8.0Hz,1H),7.70(d,J=8.0Hz,1H),7.66–7.46(m,2H),6.92(d,J=8.8Hz,2H),5.74–5.67(m,1H),5.10–4.98(m,2H),4.90–4.86(m,1H),4.83–4.67(m,1H),4.60(d,J=14.8Hz,1H),3.11(t,J=5.2Hz,4H),2.90(d,J=2.4Hz,2H),2.50–2.44(m,4H),2.26(s,3H),1.22(s,3H),0.95–0.90(m,1H),0.71–0.66(m,1H),0.61–0.56(m,1H),0.48–0.45(m,Hz,1H).LCMS(ESI+)m/z:539.3[M+H]+,HPLC method B:RT=6.80min,purity:97.8%。
3b结构确证:1H NMR(400MHz,DMSO-d6)δ10.13(s,1H),8.82(s,1H),7.92(d,J=8.0Hz,1H),7.70(d,J=8.0Hz,1H),7.59(s,2H),7.02–6.83(m,2H),5.74–5.67(m,1H),5.06–4.97(m,2H),4.90–4.86(m,1H),4.80–4.66(m,1H),4.60(d,J=13.6Hz,1H),3.09(t,J=5.2Hz,4H),2.90(d,J=2.4Hz,2H),2.45(t,J=5.2Hz,4H),2.22(s,3H),1.22(s,3H),0.95–0.90(m,1H),0.71–0.66(m,1H),0.61–0.56(m,1H),0.48–0.45(m,Hz,1H).LCMS(ESI+)m/z:539.3[M+H]+,HPLC method B:RT=6.80min,purity:98.1%。
实施例4:化合物4的合成
步骤1:化合物4-2的合成:
在干燥的单口瓶中加入底物NaH(1.40g,34.96mmol,60%purity)和DMF(150mL),搅拌溶解后在0℃和氮气保护下加入1,2-二溴乙烷(6.57g,34.96mmol)和4-1(1.47g,9.99mmol)的DMF(30mL)溶液,在0℃下搅拌反应3小时,LC-MS监测。反应结束后,加入饱和氯化铵溶液和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物4-2(0.55g,3.18mmol,31.79%产率),LCMS(ESI+)m/z:174.0[M+H]+
步骤2:化合物4-3的合成:
在干燥的单口瓶中加入底物4-2(470mg,2.71mmol)和甲醇(20mL),搅拌溶解后在0℃下加入硼氢化钠(205.30mg,5.43mmol),在0℃下搅拌反应1小时,LC-MS监测。反应结束后,加入饱和碳酸氢钠溶液和DCM萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物4-3(470mg,2.68mmol,98.95%产率),LCMS(ESI+)m/z:176.2[M+H]+
步骤3:化合物4-4的合成:
在干燥的单口瓶中加入底物4-3(470mg,2.68mmol)和DCM(20mL),搅拌溶解后在0℃下加入DIPEA(1.39g,10.73mmol,1.87mL),在0℃下缓慢滴加加入乙酰氯(421.10mg,5.36mmol,381.43μL),0℃下搅拌反应1小时,LC-MS监测。反应结束后,旋干溶剂,用反相柱层析纯化得到产物4-4(540mg,2.49mmol,92.66%产率),LCMS(ESI+)m/z:218.2[M+H]+
步骤4:化合物4-6的合成:
在干燥的单口瓶中加入底物4-4(540mg,2.49mmol)和DCM(5mL),搅拌溶解后加入4-5(736.29mg,4.97mmol)和H2O2(845.31mg,7.46mmol,761.54μL,30%purity),升温至40℃下搅拌反应16小时,LC-MS监测。反应结束后,加入饱和碳酸氢钠溶液和DCM萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物4-6(560mg,2.40mmol,96.59%产率),LCMS(ESI+)m/z:234.0[M+H]+
步骤5:化合物4-7的合成:
在干燥的单口瓶中加入底物4-6(560mg,2.40mmol)和DCM(20mL),搅拌溶解后在0℃和氮气保护下滴加加入三乙胺(1.28g,12.65mmol,1.76mL)和POCl3(775.65mg,5.06mmol,471.52μL),0℃下搅拌反应3小时,LC-MS监测。反应结束后,加入饱和碳酸氢钠溶液和DCM萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干得到粗产物4-7(650mg,crude), LCMS(ESI+)m/z:252.0[M+H]+
步骤6:化合物4-8的合成:
在干燥的单口瓶中加入底物4-7(650mg,2.58mmol)和甲醇(20mL),搅拌溶解后加入碳酸钾(1.78g,12.91mmol),室温下搅拌反应2小时,LC-MS监测。反应结束后,过滤,滤液旋干,用反相柱层析纯化得到产物4-8(197mg,939.56μmol,36.38%产率),LCMS(ESI+)m/z:210.1[M+H]+
步骤7:化合物4-9的合成:
按照实施例3中步骤3的合成方法,将步骤3中的3-3替换为4-8(40mg,190.77μmol),合成方法相同,得到化合物4-9(39mg,98.62μmol,51.69%产率),LCMS(ESI+)m/z:396.0[M+H]+
步骤8:化合物4的合成:
按照实施例3中步骤4的合成方法,将步骤4中的3-4替换为4-9(39mg,98.62μmol),合成方法相同,得到化合物4(24.4mg,45.30μmol,45.93%产率)。1H NMR(600MHz,DMSO-d6)δ10.13(s,1H),8.81(s,1H),7.84(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.66–7.50(m,2H),6.92(d,J=8.4Hz,2H),5.70–5.64(m,1H),5.31(d,J=5.4Hz,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.78–4.54(m,2H),3.75(d,J=5.4Hz,1H),3.10(t,J=4.8Hz,4H),2.88–2.79(m,2H),2.46(t,J=4.8Hz,4H),2.28–2.19(m,4H),1.13–1.11(m,1H),0.69–0.66(m,1H),0.45–0.42(m,1H),0.41–0.31(m,2H).LCMS(ESI+)m/z:539.2[M+H]+,HPLC method B:RT=6.98min,purity:98.9%。
化合物4通过SFC拆分得到以下两个化合物:4a RT=3.12min;4b RT=5.44min.
4a结构确证:1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.81(s,1H),7.84(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.59(s,2H),6.92(d,J=9.2Hz,2H),5.74–5.61(m,1H),5.31(d,J=5.2Hz,1H),5.02–4.99(m,1H),4.91–4.87(m,1H),4.77–4.57(m,2H),3.75(d,J=5.2Hz,1H),3.10(t,J=5.2Hz,4H),2.94–2.75(m,2H),2.46(t,J=5.2Hz,4H),2.31–2.18(m,4H),1.13–1.10(m,1H),0.69–0.66(m,1H),0.45–0.42(m,1H),0.41–0.31(m,2H).LCMS(ESI+)m/z:539.2[M+H]+,HPLC method B:RT=6.91min,purity:98.7%。
4b结构确证:1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.81(s,1H),7.84(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.59(s,2H),6.97–6.87(m,2H),5.74–5.61(m,1H),5.31(d,J=5.6Hz,1H),5.02–4.99(m,1H),4.91–4.87(m,1H),4.76–4.56(m,2H),3.75(d,J=5.2Hz,1H),3.10(t,J=4.8Hz,4H),2.97–2.76(m,2H),2.46(t,J=5.2Hz,4H),2.30–2.18(m,4H),1.13–1.10(m,1H),0.69–0.66(m,1H),0.45–0.42(m,1H),0.41–0.31(m,2H).LCMS(ESI+)m/z:539.2[M+H]+,HPLC method B:RT=6.92min,purity:98.0%。
实施例5:化合物5的合成
步骤1:化合物5-1的合成:
在干燥的单口瓶中加入底物3-2(40.21mg,207.64μmol)和MeOH(1.2mL),搅拌溶解后在0℃下缓慢加入NaBD4(9.56mg,228.40μmol),在0℃下搅拌反应1小时,LC-MS监测。反应结束后,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物5-1(37mg,188.15μmol,90.62%产率),LCMS(ESI+)m/z:196[M+H]+
步骤2:化合物5-2的合成:
在干燥的单口瓶中加入底物5-1(37mg,188.15μmol)和苯甲醚(2mL),搅拌溶解后加入IM-1(50.18mg,225.78μmol)、CuI(71.67mg,376.30μmol)、K2CO3(65.01mg,470.38μmol)、N,N'-二甲基-1,2-环己二胺(107.05mg,752.60μmol)和NaI(56.40mg,376.30μmol),微波130℃,搅拌反应5小时,LC-MS监测。反应结束后,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物5-2(13mg,34.08μmol,18.11%yield),LCMS(ESI+)m/z:383[M+H]+
步骤3:化合物5的合成:
按照实施例3中步骤4的合成方法,将步骤4中的3-4替换为5-2(13mg,33.99μmol),合成方法相同,得到化合物5(8.5mg,15.28μmol,44.96%产率)。1H NMR(600MHz,DMSO-d6)δ10.14(s,1H),8.82(s,1H),7.94(s,1H),7.68(d,J=7.8Hz,1H),7.59(s,2H),6.91(d,J=8.4Hz,2H),5.69–5.67(m,1H),5.35(s,1H),5.02(d,J=10.2Hz,1H),4.90(d,J=16.8Hz,1H),4.63–4.49(m,2H),3.10–3.08(m,4H),3.07–3.04(m,1H),2.78(d,J=18.0Hz,1H),2.47–2.45(m,4H),2.22(s,3H),1.01–0.98(m,1H),0.69(t,J=7.8Hz,2H),0.53–0.49(m,1H).LCMS(ESI+)m/z:526.4[M+H]+,HPLC method B:RT=6.47min,purity:96.2%。
实施例6:化合物6的合成
步骤1:化合物6-2的合成:
在干燥的单口瓶中加入底物6-1(157mg,748.79μmol)和DCM(10mL),搅拌溶解后加入DMP(635.18mg,1.50mmol),在20℃下搅拌反应16小时,LC-MS监测。反应结束后,加入饱和碳酸氢钠溶液和DCM萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物6-2(141mg,679.01μmol,90.68%产率),LCMS(ESI+)m/z:208.2[M+H]+
步骤2:化合物6-3的合成:
按照实施例3中步骤2的合成方法,将步骤2中的3-2替换为6-2(141mg,679.01μmol),合成方法相同,得到化合物6-3(150mg,crude),LCMS(ESI+)m/z:224.2[M+H]+
步骤3:化合物6-4的合成:
按照实施例3中步骤3的合成方法,将步骤3中的3-3替换为6-3(95.39mg,429.15μmol),合成方法相同,得到化合物6-4(75mg,183.15μmol,51.21%产率),LCMS(ESI+)m/z:410.4[M+H]+
步骤4:化合物6的合成:
按照实施例3中步骤4的合成方法,将步骤4中的3-4替换为6-4(45mg,109.89μmol),合成方法相同,得到化合物6(28.5mg,51.56μmol,45.97%产率)。1H NMR(600MHz,DMSO-d6)δ10.12(s,1H),8.82(s,1H),7.79(s,1H),7.69(d,J=8.4Hz,1H),7.59(s,2H),6.93(d,J=8.4Hz,2H),5.70–5.64(m,1H),5.04–4.95(m,1H),4.86(d,J=17.4Hz,1H),4.81(d,J=15.6Hz,1H),4.69(s,1H),4.69–4.63(m,1H),3.10(t,J=4.8Hz,4H),2.90–2.86(m,1H),2.81–2.79(m,1H),2.47(s,4H),2.23(s,3H),2.01–1.96(m,1H),1.48–1.39(m,4H),0.86–0.85(m,1H),0.64–0.62(m,1H),0.34–0.31(m,1H),0.22–0.21(m,1H).LCMS(ESI+)m/z:553.2[M+H]+,HPLC method B:RT=7.91min,purity:99.7%。
化合物6通过SFC拆分得到以下两个化合物:6a RT=2.75min;6b RT=4.41min
6a结构确证:1H NMR(400MHz,DMSO-d6)δ10.11(s,1H),8.82(s,1H),7.79(d,J=8.4Hz,1H),7.69(d,J=8.4Hz,1H),7.58(d,J=8.4Hz,2H),6.99–6.85(m,2H),5.72–5.62(m,1H),5.00–4.98(m,1H),4.98–4.84(m,1H),4.79(s,1H),4.69(d,J=1.8Hz,1H),4.68–4.66(m,1H),3.10(t,J=4.8Hz,4H),2.89–2.78(m,2H),2.46(t,J=4.8Hz,4H),2.22(s,3H),1.99–1.97(m,1H),1.46–1.38(m,4H),0.87–0.85(m,1H),0.65–0.61(m,1H),0.33–0.30(m,1H),0.23–0.20(m,1H).LCMS(ESI+)m/z:553.2[M+H]+,HPLC method B:RT=7.91min,purity:97.9%。
6b结构确证:1H NMR(400MHz,DMSO-d6)δ10.11(s,1H),8.82(s,1H),7.79(d,J=8.4Hz,1H),7.69(d,J=8.4Hz,1H),7.67–7.49(m,2H),6.92(d,J=8.8Hz,2H),5.72–5.62(m,1H),5.03–4.94(m,1H),4.88–4.84(m,1H),4.79(s,1H),4.69(s,1H),4.67–4.65(m,1H),3.10(t,J=4.8Hz,4H),2.93–2.77(m,2H),2.46(t,J=4.8Hz,4H),2.22(s,3H),1.99–1.97(m,1H),1.47–1.36(m,4H),0.87–0.84(m,1H),0.65–0.62(m,1H),0.33–0.30(m,1H),0.22–0.20(m,1H).LCMS(ESI+)m/z:553.2[M+H]+,HPLC method B:RT=7.91min,purity:99.1%。
实施例7:化合物7的合成
步骤1:化合物7-3的合成:
在干燥的单口瓶中加入底物7-1(400mg,2.02mmol)和DMSO(5mL),搅拌溶解后加入7-2(313.14mg,2.22mmol)和碳酸钾(836.50mg,6.05mmol),在65℃下搅拌反应10小时,LC-MS监测。反应结束后,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物7-3(633mg,1.98mmol,98.24%产率),LCMS(ESI+)m/z:319[M+H]+
步骤2:化合物7-4的合成:
在干燥的单口瓶中加入底物7-3(633mg,1.98mmol)和甲醇(5mL),搅拌溶解后加入Pd/C(60mg,494.03μmol),在H2下室温搅拌反应12小时,LC-MS监测。反应结束后,用硅藻土过滤反应液,滤液旋干,得到粗产物7-4(459mg,crude),LCMS(ESI+)m/z:290[M+H]+
步骤3:化合物7-5的合成:
在干燥的单口瓶中加入底物7-4(150mg,518.36μmol)和THF(5mL),搅拌溶解后在0℃下缓慢加入LiAlH4(19.67mg,518.36μmol),后在65℃下搅拌反应4小时,LC-MS监测。反应结束后,在0℃下加入水淬灭反应,用10%的氢氧化钠溶液处理后,再加入水,过滤,滤液旋干,用反相柱层析纯化得到产物7-5(70mg,0.34mmol,66.43%产率),LCMS(ESI+)m/z:204[M+H]+
步骤4:化合物7的合成:
按照实施例3中步骤4的合成方法,将步骤4中的3-4替换为6-4(15mg,36.63μmol),将1-8替换为7-5(8.94mg,43.96μmol)合成方法相同,得到化合物7(10.4mg,16.21μmol,44.25%产率)。1H NMR(600MHz,DMSO-d6)δ10.06(s,1H),8.79(s,1H),7.76(d,J=8.4Hz,1H),7.71(d,J=8.4Hz,1H),7.66–7.47(m,2H),6.72(d,J=8.4Hz,2H),5.72–5.63(m,1H),4.99(d,J=10.2Hz,1H),4.86(d,J=17.4Hz,1H),4.80(s,1H),4.68(s,1H),4.64(d,J=15.6Hz,1H),3.75–3.54(m,2H),3.46(d,J=10.8Hz,2H),2.88–2.77(m,2H),2.50(s,3H),2.12–1.92(m,4H),1.57(s,1H),1.45–1.39(m,4H),0.87–0.85(m,1H),0.65–0.61(m,1H),0.33–0.29(m,1H),0.26–0.19(m,1H).LCMS(ESI+)m/z:565.2[M+H]+,HPLC method B:RT=8.49min,purity:88.0%。
化合物7通过SFC拆分得到以下两个化合物:7a RT=3.564min;7b RT=6.246min
化合物7a(8.3mg,13.52μmol);SFC保留时间t=3.564min。1H NMR(400MHz,DMSO-d6)δ10.06(s,1H),8.80(s,1H),7.76(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.59(s,2H),6.71(d,J=8.8Hz,2H),5.72–5.62(m,1H),5.00–4.98(m,1H),4.88–4.79(m,2H),4.68(s,1H),4.67–4.61(m,1H),4.80–4.77(m,1H),3.58(d,J=5.2Hz,2H),3.43(d,J=10.8Hz,2H),3.27(d,J=10.8Hz,2H),2.92–2.76(m,2H),2.46–2.41(m,1H),2.02–1.94(m,4H),1.53(d,J=8.0Hz,1H),1.46–1.42(m,1H),1.40(s,3H),0.87-0.84(m,1H),0.68–0.60(m,1H),0.34–0.30(m,1H),0.23–0.19(m,1H).LCMS(ESI)m/z:565.4[M+H]+,HPLC method B:RT=8.36min,purity>92.1%。
化合物7b(8.3mg,13.91μmol);SFC保留时间t=6.246min。1H NMR(400MHz,DMSO-d6)δ10.06(s,1H),8.79(s,1H),7.76(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.59(s,2H),6.71(d,J=8.8Hz,2H),5.72–5.62(m,1H),5.00–4.98(m,1H),4.88–4.79(m,2H),4.68(s,1H),4.67–4.61(m,1H),4.80–4.77(m,1H),3.58(d,J=5.2Hz,2H),3.43(d,J=10.8Hz,2H),3.27(d,J=10.8Hz,2H),2.92–2.76(m,2H),2.45–2.40(m,1H),2.02–1.95(m,4H),1.53(d,J=8.0Hz,1H),1.46–1.42(m,1H),1.40(s,3H),0.87–0.84(m,1H),0.68–0.60(m,1H),0.34–0.30(m,1H),0.23–0.19(m,1H).LCMS(ESI)m/z:565.4[M+H]+,HPLC method B:RT=8.30min,purity>95.2%.
实施例8:化合物8的合成
步骤1:化合物8的合成:
按照实施例3中步骤4的合成方法,将步骤4中的3-4替换为6-4(15mg,36.63μmol),将1-8替换为8-1(9.02mg,43.96μmol)合成方法相同,得到化合物8(7.4mg,12.14μmol,33.15%产率)。1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.84(s,1H),7.76(d,J=8.4Hz,1H),7.72(s,1H),7.70(s,1H),7.45–7.36(m,1H),6.99(d,J=8.4Hz,1H),5.72–5.63(m,1H),5.00–4.98(m,1H),,4.91–4.83(m,1H),4.83–4.75(m,1H),4.70(s,1H),4.67–4.61(m,1H),2.90–2.77(m,6H),2.52–2.45(m,4H),2.25(s,6H),2.01–1.97(m,1H),1.47–1.38(m,4H),0.87–0.84(m,1H),0.65–0.60(m,1H),0.36–0.29(m,1H),0.23–0.19(m,1H).LCMS(ESI+)m/z:567.2[M+H]+,HPLC method B:RT=8.83min,purity:93.0%。
实施例9:化合物9的合成
步骤1:化合物9-2的合成:
在干燥的单口瓶中加入底物9-1(3g,18.62mmol)和DMF(20mL),搅拌溶解后加入碘甲烷(8.7g,61.43mmol)、TBAHS(630mg,18.62mmol)和碳酸钾(8.48g,61.43mmol),在70℃下搅拌反应15小时,LC-MS监测。反应结束后,加入水淬灭反应,加入EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物9-2(3.4g,16.73mmol,89.87%产率),LCMS(ESI+)m/z:204[M+H]+
步骤2:化合物9-3的合成:
在干燥的单口瓶中加入底物9-2(3.4g,16.73mmol)和H2SO4(10mL),搅拌溶解后在0℃下,缓慢滴加加入HNO3(1.02g,16.24mmol),在0℃下搅拌反应,TLC监测。反应结束后,0℃下加入水淬灭反应,加入EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物9-3(3.9g,15.71mmol,96.76%产率)。
步骤3:化合物9-4的合成:
在干燥的单口瓶中加入底物9-3(270mg,1.09mmol)和THF(10mL),搅拌溶解后加入BMS(334.8mg,4.35mmol)),在70℃下搅拌反应24小时,LC-MS监测。反应结束后,加入饱和亚硫酸钠溶液淬灭反应,加入EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物9-4(200mg,907.99μmol,83.48%产率),LCMS(ESI+)m/z:220[M+H]+
步骤4:化合物9-5的合成:
在干燥的单口瓶中加入底物9-4(200mg,907.99μmol)和乙醇(10mL),搅拌溶解后加入Pd/C(110.28mg,907.99μmol),在H2下室温搅拌反应4小时,LC-MS监测。反应结束后,用硅藻土过滤反应液,滤液旋干,得到粗产物9-5(170mg,crude),LCMS(ESI+)m/z:191[M+H]+
步骤5:化合物9的合成:
按照实施例3中步骤4的合成方法,将步骤4中的1-8替换为9-5(8.66mg,45.51μmol)合成方法相同,得到化合物9(1.7mg,2.98μmol,7.84%产率)。1H NMR(400MHz,DMSO-d6)δ10.18(s,1H),8.87(s,1H),7.88(d,J=8.0Hz,1H),7.71(d,J=8.0Hz,1H),7.54(s,1H),7.42(d,J=8.4Hz,1H),7.29(d,J=8.4Hz,1H),5.75–5.65(m,1H),5.04(s,1H),5.04–5.00(m,1H),4.91–4.86(m,1H),4.80–4.67(m,1H),4.61–4.56(m,1H),3.45–3.37(m,2H),2.91(s,2H),2.43–2.28(m,5H),1.26–1.20(m,9H),0.95–0.90(m,1H),0.72–0.67(m,1H),0.61–0.56(m,1H),,0.49–0.45(m,1H).LCMS(ESI+)m/z:538.3[M+H]+,HPLC method B:RT=8.20min,purity:94.1%。
实施例10:化合物10的合成
步骤1:化合物10的合成:
按照实施例3中步骤4的合成方法,将步骤4中的1-8替换为8-1(9.34mg,45.51μmol)合成方法相同,得到化合物10(7.6mg,13.11μmol,34.55%产率)。1H NMR(400MHz,DMSO-d6)δ10.14(s,1H),8.85(s,1H),7.89(d,J=8.0Hz,1H),7.72(d,J=8.0Hz,1H),7.66(s,1H),7.47–7.39(m,1H),6.99(d,J=8.4Hz,1H),5.75–5.65(m,1H),5.03(s,1H),5.03–4.97(m,1H),4.90–4.86(m,1H),4.74(d,J=15.6Hz,1H),4.63–4.57(m,1H),2.89(d,J=2.4Hz,2H),2.83(t,J=4.8Hz,4H),2.58–2.51(m,4H),2.28(s,3H),2.24(s,3H),1.22(s,3H),0.95–0.90(m,1H),0.71–0.66(m,1H),0.60–0.56(m,1H),0.48–0.45(m,1H).LCMS(ESI+)m/z:553.4[M+H]+,HPLC method B:RT=7.62min,purity:95.3%。
实施例11:化合物11的合成
步骤1:化合物11的合成:
按照实施例3中步骤4的合成方法,将步骤4中的1-8替换为11-1(15.74mg,60.69μmol),合成方法相同,得到化合物11(5.33mg,8.16μmol,16.14%产率)。1H NMR(600MHz,DMSO-d6)δ10.12(s,1H),8.82(s,1H),7.91(s,1H),7.69(d,J=8.4Hz,1H),7.58(s,2H),6.91(d,J=8.4Hz,2H),5.72–5.66(m,1H),5.03–4.99(m,2H),4.87(d,J=17.4Hz,1H),4.76–4.73(m,1H),4.61–4.59(m 1H),3.09–3.07(m,4H),2.92–2.86(m,2H),2.43–2.39(m,4H),2.64(s,3H),1.55–1.49(m,8H),1.22(s,3H),0.94–0.91(m,1H),0.70–0.67(m,1H),0.59–0.56(m,1H),0.48–0.46(m,1H).LCMS(ESI+)m/z:607.3[M+H]+,HPLC method B:RT=8.06min,purity:93.4%。
化合物11通过SFC拆分得到以下两个化合物:11a RT=3.475min;11b RT=5.377min
化合物11a(12.86mg,21.19μmol);SFC保留时间t=3.475min。1H NMR(400MHz,DMSO-d6)δ10.11(s,1H),8.82(s,1H),7.91(d,J=7.2Hz,1H),7.70(d,J=8.0Hz,1H),7.56(s,2H),6.91(d,J=9.2Hz,2H),5.74–5.64(m,1H),5.02–4.99(m,2H),4.88(dd,J=17.2,1.2Hz,1H),4.72–4.58(m,2H),3.08(t,J=5.6Hz,4H),2.94–2.85(m,2H),2.29(s,4H),2.16(s,3H),1.53(t,J=5.6Hz,4H),1.46(t,J=5.6Hz,4H),1.22(s,3H),0.95–0.90(m,1H),0.71–0.66(m,1H),0.60–0.55(m,1H),0.48–0.43(m,1H).LCMS(ESI)m/z:607.3[M+H]+,HPLC method B:RT=8.35min,purity>94.6%。
化合物11b(12.1mg,19.94μmol);SFC保留时间t=5.377min。1H NMR(400MHz,DMSO-d6)δ10.11(s,1H),8.82(s,1H),7.91(d,J=7.2Hz,1H),7.69(d,J=8.0Hz,1H),7.56(s,2H),6.91(d,J=9.2Hz,2H),5.74–5.64(m,1H),5.02–4.99(m,2H),4.88(dd,J=17.2,1.2Hz,1H),4.72–4.58(m,2H),3.08(t,J=5.6Hz,4H),2.94–2.85(m,2H),2.29(s,4H),2.16(s,3H),1.53(t,J=5.6Hz,4H),1.46(t,J=5.6Hz,4H),1.22(s,3H),0.95–0.90(m,1H),0.71–0.66(m,1H),0.60–0.55(m,1H),0.48–0.43(m,1H).LCMS(ESI)m/z:607.3[M+H]+,HPLC method B:RT=7.89min,purity>97.2%。
实施例12:化合物12的合成
步骤1:化合物12的合成:
按照实施例3中步骤4的合成方法,将步骤4中的1-8替换为12-1(16.64mg,75.86μmol),合成方法相同,得到化合物12(9.38mg,15.64μmol,30.93%产率)。1H NMR(600MHz,DMSO-d6)δ10.14(s,1H),8.82(s,1H),7.92–7.90(m,1H),7.69(d,J=7.8Hz,1H),7.58(s,2H),6.92(d,J=9.0Hz,2H),5.72–5.66(m,1H),5.03–4.99(m,2H),4.87(d,J=17.4Hz,1H),4.73(s,1H),4.61–4.58(m,1H),3.68(d,J=12.0Hz,2H),2.92–2.86(m,2H),2.63(t,J=12.0Hz,2H),2.38–2.32(m,7H),1.91–1.87(m,2H),1.55–1.49(m,2H),1.22(s,3H),0.94–0.91(m,1H),0.70–0.67(m,1H),0.59–0.56(m,1H),0.48–0.46(m,1H).LCMS(ESI+)m/z:567.2[M+H]+,HPLC method B:RT=7.29min,purity:93.3%。
实施例13:化合物13的合成
步骤1:化合物13的合成:
按照实施例3中步骤4的合成方法,将步骤4中的1-8替换为7-5(19.76mg,97.21μmol),合成方法相同,得到化合物13(14mg,22.88μmol,47.08%产率)。1H NMR(600MHz,Chloroform-d)δ8.81(s,1H),7.69–7.67(m,2H),7.49(s,2H),6.72(d,J=9.0Hz,2H),5.74–5.68(m,1H),5.02(d,J=9.6Hz,1H),4.94(d,J=17.4Hz,1H),4.84–4.80(m,2H),4.03(s,2H),3.64–3.58(m,4H),2.95(d,J=6.6Hz,2H),2.32(s,3H),1.77(s,2H),1.41(s,3H),1.12–1.08(m,1H),0.91–0.87(m,1H),0.70–0.67(m,1H),0.60–0.56(m,1H).LCMS(ESI+)m/z:551.2[M+H]+,HPLC method B:RT=7.16min,purity:90.0%。
实施例14:化合物14的合成
步骤1:化合物14-3的合成:
在干燥的单口瓶中加入底物14-1(217.24mg,1mmol)用甲醇(3mL)溶解,加入14-2(200.32mg,2mmol),醋酸(3.00mg,50.00μmol),室温搅拌反应0.5小时,加入NaBH3CN(94.26mg,1.50mmol)后升温至50℃反应4小时,LC-MS监测。反应结束后,旋干溶剂,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物14-3(200mg,663.57μmol,66.36%产率),LCMS(ESI+)m/z:302[M+H]+
步骤2:化合物14-4的合成:
在干燥的单口瓶中加入底物14-3(200mg,663.57μmol)用DCM(1mL)溶解,加入TFA(756.61mg,6.64mmol),室温搅拌反应0.5小时,LC-MS监测。反应结束后,旋干溶剂得到粗产物14-4(100mg,496.81μmol,74.87%产率),LCMS(ESI+)m/z:202[M+H]+。
步骤3:化合物14-5的合成:
在干燥的单口瓶中加入底物14-4(140mg,695.54μmol)和DMSO(3mL),搅拌溶解后加入对氟硝基苯(98.14mg,695.54μmol)和碳酸钾(288.38mg,2.09mmol),在80℃下搅拌反应4小时,LC-MS监测。反应结束后,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物14-5(95mg,294.69μmol,42.37%产率),LCMS(ESI+)m/z:322[M+H]+
步骤3:化合物14-6的合成:
在干燥的单口瓶中加入底物14-5(95mg,294.69μmol)和甲醇(5mL),搅拌溶解后加入Pd/C(10.74mg,88.41μmol),在H2下室温搅拌反应12小时,LC-MS监测。反应结束后,用硅藻土过滤反应液,滤液旋干,得到粗产物14-6(73mg,crude),LCMS(ESI+)m/z:293[M+H]+
步骤4:化合物14的合成:
按照实施例3中步骤4的合成方法,将步骤4中的1-8替换为14-6(15mg,51.30μmol),合成方法相同,得到化合物14(4.1mg,5.72μmol,22.31%产率)。1H NMR(400MHz,DMSO-d6)δ10.13(s,1H),8.82(s,1H),7.91(s,1H),7.70(d,J=8.0Hz,1H),7.57(s,2H),6.92(d,J=8.8Hz,2H),5.75–7.64(m,1H),,5.13–4.97(m,3H),4.88(d,J=17.2Hz,1H),4.73(s,1H),4.60(d,J=15.2Hz,1H),3.88(t,J=12.4Hz,1H),3.75(d,J=12.4Hz,1H),2.90(d,J=2.4Hz,2H),2.78–2.69(m,1H),2.60(s,4H),2.42–2.22(m,6H),2.15(s,3H),1.94–1.81(m,6H),1.75(d,J=12.4Hz,1H),1.22(s,3H),0.95–0.84(m,6H),0.73–0.66(m,6H),0.61–0.53(m,1H),0.48–0.43(m,1H).LCMS(ESI+)m/z:640.3[M+H]+,HPLC method B:RT=6.63min,purity:89.4%。
实施例15:化合物15的合成
步骤1:化合物15-3的合成:
在干燥的单口瓶中加入底物15-1(181.83mg,1.42mmol)和DMF(10mL),搅拌溶解后加入15-2(200mg,1.29mmol)和碳酸钾(534.53mg,3.87mmol),在90℃下搅拌反应3小时,LC-MS监测。反应结束后,加入水和EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,得到粗产物15-3(100mg,crude),LCMS(ESI+)m/z:263[M+H]+
步骤2:化合物15-4的合成:
在干燥的单口瓶中加入底物15-3(100mg,379.75μmol)和甲醇(5mL),搅拌溶解后加入Pd/C(230.60mg,1.90mmol),在H2下室温搅拌反应0.5小时,LC-MS监测。反应结束后,用硅藻土过滤反应液,滤液旋干,得到粗产物15-4(109mg,crude),LCMS(ESI+)m/z:234[M+H]+
步骤3:化合物15的合成:
按照实施例3中步骤4的合成方法,将步骤4中的1-8替换为15-4(8.85mg,37.93μmol),合成方法相同,得到化合物15(1.75mg,2.59μmol,10.23%产率)。1H NMR(600MHz,DMSO-d6)δ10.12(s,1H),8.85(s,1H),7.88(d,J=8.4Hz,1H),7.71(d,J=8.0Hz,1H),7.65(s,1H),7.43–7.40(m,1H),6.97(d,J=8.8Hz,1H),5.74–5.66(m,1H),5.03(s,1H),5.03–5.00(m,1H),4.90–4.85(m,1H),4.77–4.71(m,1H),4.62–4.57(m,1H),3.03(d,J=12.0Hz,2H),2.89–2.87(m,2H),2.59–2.53(m,2H),2.24–2.18(m,10H),1.83(d,J=11.6Hz,2H),1.58–1.49(m,2H),1.22(s,3H),0.95–0.89(m,1H),0.71–0.66(m,1H),0.61–0.56(m,1H),0.48–0.44(m,1H).LCMS(ESI+)m/z:581.3[M+H]+,HPLC method B:RT=8.41min,purity:88.68%。
实施例16:化合物16的合成
步骤1:化合物16-1的合成:
在干燥的单口瓶中加入底物2-5(60mg,0.21mmol)和THF(1mL),搅拌溶解后加入硼烷的四氢呋喃络合物(1mL),在室温下搅拌反应5小时,LC-MS监测。反应结束后,加入水淬灭,用EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物16-1(40mg,0.15mmol,70.18%产率),LCMS(ESI+)m/z:267[M+H]+
步骤2:化合物16-2的合成:
按照实施例2中步骤5的合成方法,将步骤5中的2-5替换为16-1(40mg,0.15mmol),合成方法相同,得到化合物16-2(30mg,73.43μmol,48.83%产率),LCMS(ESI+)m/z:409[M+H]+
步骤3:化合物16的合成:
按照实施例3中步骤4的合成方法,将步骤4中的3-4替换为16-2(30mg,73.43μmol),合成方法相同,得到化合物16(6.31mg,11.44μmol,15.58%产率)。1H NMR(600MHz, DMSO-d6)δ10.05(s,1H),8.78(s,1H),7.71(s,1H),7.70–7.52(m,2H),7.98–7.82(m,3H),5.72–7.68(m,1H),5.05(d,J=10.2Hz,2H),4.93(d,J=16.8Hz,1H),4.51(s,2H),3.62(s,2H),3.31(d,J=14.4Hz,1H),3.08(s,4H),2.46(s,4H),2.45(s,2H),2.28(s,3H),2.20(t,J=14.4Hz,3H),2.03–1.99(m,2H),1.09(t,J=7.2Hz,3H).LCMS(ESI+)m/z:552.4[M+H]+,HPLC method B:RT=7.03min,purity:95.7%。
实施例17:化合物17的合成
步骤1:化合物17-1的合成:
在干燥的单口瓶中加入底物2-4(200mg,829.6μmol)和DMF(10mL),搅拌溶解后加入NaH(119.5mg,4.98mmol),在0℃下搅拌反应0.5小时后,加入1,3-二碘丙烷(736.4mg,2.48mmol),在室温下搅拌反应0.5小时,LC-MS监测。反应结束后,加入水淬灭反应,用EA萃取3次,合并有机相,无水硫酸钠处理后,过滤旋干,用反相柱层析纯化得到产物17-1(70mg,250μmol,29.87%产率),LCMS(ESI+)m/z:281[M+H]+
步骤2:化合物17-2的合成:
按照实施例2中步骤5的合成方法,将步骤5中的2-5替换为17-1(70mg,250μmol),合成方法相同,得到化合物17-2(25mg,59.17μmol,23.69%产率),LCMS(ESI+)m/z:423[M+H]+
步骤3:化合物17的合成:
按照实施例3中步骤4的合成方法,将步骤4中的3-4替换为17-2(25mg,59.17μmol),合成方法相同,得到化合物17(5mg,8.84μmol,14.94%产率)。1H NMR(600MHz,DMSO-d6)δ10.25(s,1H),8.85(s,1H),8.22(s,1H),7.73(s,1H),7.62(s,2H),6.92(d,J=8.6Hz,2H),5.75–5.65(m,1H),5.08–5.03(m,1H),4.95–4.89(m,1H),4.59(d,J=5.9Hz,2H),4.16(t,J=6.0Hz,2H),3.82(q,J=7.0Hz,2H),3.10(t,J=4.9Hz,4H),2.82(t,J=6.1Hz,2H),2.46(t,J=4.9Hz,4H),2.22(s,3H),2.20–2.15(m,2H),1.15(t,J=7.1Hz,3H).LCMS(ESI+)m/z:598.2[M+MeOH+H]+,HPLC method B:RT=7.23min,purity:80.0%。
实施例18:化合物18的合成
步骤1:化合物18-1的合成:
在干燥的三口瓶中加入底物3-2(100mg,516.45μmol),加入无水四氢呋喃(2mL)溶解,氮气保护下冷却至零度,然后缓慢滴加乙基溴化镁的四氢呋喃溶液(2.07mmol,1.04mL,2M),保持温度反应1个小时。LC-MS监测。向反应液中加入饱和氯化铵水溶液,用乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得化合物18-1(112.00mg,500.67μmol)。LCMS(ESI)m/z:224.2[M+H]+
步骤2:化合物18-2的合成:
在干燥的微波管中,加入底物18-1(112.00mg,500.67μmol)、底物IM-1(111.00mg,499.40μmol)、碘化亚铜(190.22mg,998.80μmol)、碘化钠(149.71mg,998.80μmol)、碳酸钾(172.29mg,1.25mmol)和反-(1R,2R)-N,N'-二甲基1,2-环己烷二胺(284.48mg,2.00mmol),然后加入苯甲醚(3mL),在氮气环境下,130度微波加热反应三小时。LC-MS监测。将反应液冷却至室温,过滤,用乙酸乙酯洗涤两次,减压浓缩,残余物用柱层析纯化得到化合物18-2(90mg,220.31μmol)。LCMS(ESI)m/z:410.2[M+H]+
步骤3:化合物18的合成:
在干燥的单口瓶中,加入底物18-2(16mg,39.07μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(12.14mg,70.33μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(50.40mg,390.72μmol)和底物4-(4-甲基哌嗪)苯胺(8.97mg,46.89μmol),45度反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物18(5.41mg,9.79μmol)。1H NMR(600MHz,DMSO-d6)δ10.16(s,1H),8.83(s,1H),7.90(s,1H),7.72–7.70(d,J=8.4Hz,1H),7.60(s,2H),6.95–6.93(d,J=8.4Hz,2H),5.72–5.64(m,1H),5.02–5.01(m,1H),4.95(s,1H),4.89–4.86(m,1H),4.84–4.71(m,1H),4.60–4.52(m,1H),3.26–3.05(m,4H),3.01(m,1H),2.74(m,1H),2.48–2.31(m,7H),1.78–1.73(m,1H),1.60–1.54(m,1H),0.96–0.95(m,1H),0.71–0.69(m,5H),0.41–0.39(m,1H).LCMS(ESI)m/z:553.3[M+H]+,HPLC method B:RT=7.26min,purity:>97.4%。
实施例19:化合物19的合成
步骤1:化合物19-2的合成:
在干燥的单口瓶中加入底物19-1(142.09mg,570.04μmol),然后加入甲醇(2mL)溶解,冷却至零度,加入硼氢化钠(86.26mg,2.28mmol)保持温度不变反应1个小时。LC-MS监测反应完全。将反应液减压浓缩,残余物加入水,用乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物19-2(130mg,517.35μmol)。LCMS(ESI)m/z:252.3[M+H]+
步骤2:化合物19-3的合成:
在干燥的单口瓶中加入底物19-2(125.64mg,0.5mmol),溶于甲醇(2mL)中,然后加入钯炭(20mg),在氢气环境下室温反应2个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物19-3(100mg,451.88μmol)。LCMS(ESI)m/z:222.3[M+H]+
步骤3:化合物19的合成:
在干燥的单口瓶中,加入底物3-4(10mg,25.29μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(9.60mg,55.63μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(32.68mg,252.86μmol)和底物19-3(33.57mg,151.72μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物19(4mg,6.57μmol)。1H NMR(600MHz,DMSO-d6)δ10.20(s,1H),8.85(s,1H),8.01(s,1H),7.88(d,J=7.8Hz,1H),7.79(d,J=7.8Hz,1H),7.45–7.48(m,1H),7.03(d,J=8.4Hz,1H),5.74–5.67(m,1H),5.12(s,1H),5.02–4.99(m,2H),4.87(d,J=16.8Hz,1H),4.75(s,1H),4.63–4.60(m,1H),4.57(d,J=4.2Hz,2H),2.88(d,J=5.4Hz,2H),2.82(t,J=4.8Hz,4H),2.46(s,4H),2.23(s,3H),1.21(s,3H),0.93–0.90(m,1H),0.70–0.66(m,1H),0.60–0.57(m,1H),0.47–0.44(m,1H).LCMS(ESI)m/z:569.3[M+H]+,HPLC method B:RT=6.37min,purity:>93.4%。
实施例20:化合物20的合成
步骤1:化合物20-2的合成:
在干燥的单口瓶中加入底物20-1(100mg,683.97μmol),溶于二甲基亚砜(2mL)中,然后加入对氟硝基苯(96.51mg,683.97μmol)和碳酸钾(94.53mg,683.97μmol),80℃下反应4个小时。LC-MS监测反应完全。将反应液冷却至室温,加水,然后用乙酸乙酯萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物20-2(120mg,448.94μmol)。LCMS(ESI)m/z:267.1[M+H]+
步骤2:化合物20-3的合成:
在干燥的单口瓶中加入底物20-2(110mg,411.52μmol),溶于甲醇(2mL)中,然后加入钯炭(15mg),在氢气环境下室温反应2个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物20-3(90mg,379.24μmol)。LCMS(ESI)m/z:238.2[M+H]+
步骤3:化合物20a&20b的合成:
在干燥的单口瓶中,加入底物3-4(10mg,25.29μmol),溶于四氢呋喃(0.5mL)中,然后加入间氯过氧苯甲酸(6.55mg,37.93μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(32.68mg,252.86μmol)和底物20-3(12.00mg,50.57μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物20a(1.0mg,1.47μmol)(20a和20b为非对映异构体,直接分离得到产物,结构为随机书写,绝对构型未确定,以保留时间为准,下同)。1H NMR(600MHz,DMSO-d6)δ10.13(s,1H),8.82(s,1H),7.92(s,1H),7.70(d,J=8.4Hz,1H),7.58(s,2H),6.92(d,J=8.4Hz,2H),5.73–5.66(m,1H),5.09–4.96(m,3H),4.88(d,J=17.4Hz,1H),4.74(d,J=12.6Hz,1H),4.60(s,1H),3.88(t,J=12.6Hz,1H),3.74(d,J=12.0Hz,1H),2.95–2.86(m,2H),2.85–2.76(m,1H),2.68(t,J=11.4Hz,1H),2.40–2.31(m,1H),2.28(s,6H),1.90–1.83(m,1H),1.76(d,J=12.6Hz,1H),1.22(s,3H),0.94–0.90(m,1H),0.70–0.64(m,1H),0.60–0.55(m,1H),0.49–0.42(m,1H).LCMS(ESI)m/z:567.2[M+H]+,HPLC method B:RT=8.69min,purity>88.7%。
化合物20b(1.0mg,1.50μmol)。1H NMR(600MHz,DMSO-d6)δ10.15(s,1H),8.83(s,1H),7.93(s,1H),7.69(d,J=8.4Hz,1H),7.60(s,2H),6.96(d,J=8.4Hz,2H),5.72–5.66(m,1H),5.04–4.85(m,3H),4.80–4.74(m,1H),4.73–4.64(m,1H),4.60(s,1H),3.87(s,1H),3.57(d,J=12.0Hz,1H),2.90(d,J=5.4Hz,2H),2.71–2.59(m,3H),2.30(s,6H),1.82(s,1H),1.60–1.54(m,1H),1.22(s,3H),0.94–0.90(m,1H),0.70–0.67(m,1H),0.62–0.55(m,1H),0.48–0.45(m, 1H).LCMS(ESI)m/z:567.2[M+H]+,HPLC method B:RT=8.70min,purity>90.2%。
实施例21:化合物21的合成
步骤1:化合物21-2的合成:
在干燥的三口瓶中加入底物21-1(80mg,385.25μmol),加入无水四氢呋喃(4.5mL)溶解,氮气保护下冷却至零度,然后缓慢滴加乙基溴化镁的四氢呋喃溶液(1.54mmol,0.77mL,2M),保持温度反应2个小时。LC-MS监测。向反应液中加入饱和氯化铵水溶液,用乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物21-2(68mg,286.05μmol)。LCMS(ESI)m/z:238.1[M+H]+
步骤2:化合物21-3的合成:
在干燥的微波管中,加入底物21-2(68mg,286.05μmol)、底物IM-1(73.86mg,332.29μmol)、碘化亚铜(108.95mg,572.09μmol)、碘化钠(85.75mg,572.09μmol)、碳酸钾(98.83mg,715.11μmol)和反-(1R,2R)-N,N'-二甲基1,2-环己烷二胺(162.75mg,1.14mmol),然后加入苯甲醚(3mL),在氮气环境下,130度微波加热反应四小时。LC-MS监测。将反应液冷却至室温,过滤,用乙酸乙酯洗涤两次,减压浓缩,残余物用柱层析纯化得到化合物21-3(57mg,134.58μmol)。LCMS(ESI)m/z:424.2[M+H]+
步骤3:化合物21的合成:
在干燥的单口瓶中,加入底物21-3(20mg,47.44μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(16.37mg,94.89μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(61.75mg,477.78μmol)和底物4-(4-甲基哌嗪)苯胺(40mg,209.13μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物21(12mg,20.54μmol)。1H NMR(400MHz,Chloroform-d)δ8.81(s,1H),7.64–7.58(m,2H),7.47(d,J=8.8Hz,2H),6.93(d,J=8.8Hz,2H),5.76–5.66(m,1H),5.08–5.05(m,1H),5.00–4.94(m,1H),4.75–4.61(m,2H),3.56(s,1H),3.28(t,J=4.8Hz,4H),3.04–2.95(m,1H),2.91–2.85(m,1H),2.73(t,J=4.8Hz,4H),2.50–2.43(m,4H),2.02–1.93(m,1H),1.80–1.71(m,1H),1.19–1.14(m,1H),1.02(t,J=7.6Hz,3H),0.75–0.71(m,1H),0.49–0.44(m,1H),0.07–0.01(m,1H).LCMS(ESI)m/z:567.3[M+H]+,HPLC method B:RT=8.54min,purity>97.0%。
实施例22:化合物22的合成
步骤1:化合物22-2的合成:
在干燥的单口瓶中加入底物22-1(90mg,410.51μmol),溶于1,2-二氯乙烷(5mL)中,加入醋酸(49.30mg,821.02μmol)和环丁酮(143.86mg,2.05mmo),室温搅拌半个小时,然后加入氰基硼氢化钠(435.02mg,2.05mmol),50℃加热反应2个小时。LC-MS监测反应完全。将反应液减压浓缩,加入饱和碳酸氢钠水溶液调节至pH=9,然后水相用二氯甲烷萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物22-2(93mg,340.25μmol)。LCMS(ESI)m/z:274.1[M+H]+
步骤2:化合物22-3的合成:
在干燥的单口瓶中加入底物22-2(93mg,340.25μmol),溶于甲醇(3mL)中,然后加入钯炭(10mg),在氢气环境下室温反应2个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物22-3(80mg,328.74μmol)。LCMS(ESI)m/z:244.2[M+H]+
步骤3:化合物22a&22b的合成:
在干燥的单口瓶中,加入底物22-3(18mg,43.96μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(15.17mg,87.91μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(28.40mg,219.78μmol)和底物21-3(16.04mg,65.93μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合22a(2.40mg,3.62μmol)。1H NMR(400MHz,DMSO-d6)δ10.08(s,1H),8.80(s,1H),7.88(d,J=8.4Hz,1H),7.72(d,J=8.0Hz,1H),7.64–7.55(m,2H),6.79–6.68(m,1H),5.74–5.64(m,1H),5.03–5.01(m,1H),4.95(s,1H),4.88(d,J=17.2Hz,1H),4.81–4.71(m,1H),4.59–4.53(m,1H),3.89–3.76(m,2H),3.48–3.42(m,4H),3.03–2.99(m,1H),2.75–2.71(m,1H),2.48(s,1H),2.19-2.12(m,2H),1.79–1.73(m,1H),1.61–1.52(m,2H),0.99–0.94(m,1H),0.85–0.79(m,1H),0.73–0.68(m,5H),0.41–0.35(m,3H),0.08–0.05(m,2H).LCMS(ESI)m/z:605.3[M+H]+,HPLC method B:RT=8.26min,purity>91.2%。
化合物22b(6.40mg,10.03μmol)。1H NMR(400MHz,DMSO-d6)δ10.08(s,1H),8.80(s, 1H),7.88(d,J=8.4Hz,1H),7.72(d,J=8.0Hz,1H),7.60(s,2H),6.69(d,J=7.2Hz,1H),5.74–5.64(m,1H),5.03–5.01(m,1H),4.95(s,1H),4.86(d,J=17.2Hz,1H),4.70–4.77(m,1H),4.59–4.53(m,1H),3.81–3.71(m,2H),3.19–3.08(m,4H),3.01(d,J=16.4Hz,2H),2.73(d,J=16.4Hz,1H),2.42–2.37(m,1H),2.03–1.91(m,2H),1.79–1.73(m,5H),1.61–1.52(m,2H),0.99–0.94(m,1H),0.72–0.68(m,5H),0.39–0.35(m,2H).LCMS(ESI)m/z:605.3[M+H]+,HPLC method B:RT=8.37min,purity>94.8%。
实施例23:化合物23的合成
步骤1:化合物23a&23b的合成:
在干燥的单口瓶中,加入底物6-4(30mg,73.26μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(22.76mg,131.87μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(94.68mg,732.59μmol)和底物23-1(30.08mg,146.52μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物23a(12.00mg,18.78μmol);SFC保留时间t=2.302min。1H NMR(400MHz,DMSO-d6)δ10.11(s,1H),8.83(s,1H),7.79–7.57(m,3H),7.41–7.38(m,1H),6.98(d,J=8.8Hz,1H),5.72–5.60(m,1H),5.01–4.96(m,1H),4.88–4.77(m,2H),4.69(s,1H),4.67–4.58(m,1H),2.92–2.74(m,6H),2.47(s,4H),2.24(s,3H),2.23(s,3H),2.03–1.92(m,1H),1.39(s,4H),0.86–0.82(m,1H),0.64–0.62(m,1H),0.30–0.29(m,1H),0.22–0.20(m,1H).LCMS(ESI)m/z:567.2[M+H]+,HPLC method B:RT=8.69min,purity>86.0%。
化合物23b(11.00mg,17.51μmol);SFC保留时间t=3.705min。1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.83(s,1H),7.79–7.57(m,3H),7.41–7.38(m,1H),6.98(d,J=8.8Hz,1H),5.72–5.60(m,1H),5.01–4.86(m,1H),4.89–4.74(m,2H),4.72–4.57(m,2H),2.94–2.77(m,6H),2.46(s,4H),2.24(s,3H),2.22(s,3H),2.03–1.92(m,1H),1.39(s,4H),0.86–0.82(m,1H),0.64–0.62(m,1H),0.31–0.29(m,1H),0.22–0.20(m,1H).LCMS(ESI)m/z:567.4[M+H]+,HPLC method B:RT=8.70min,purity>87.8%.
实施例24:化合物24的合成
在干燥的单口瓶中,加入底物18-2(10mg,24.42μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(8.43mg,48.84μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(31.56mg,244.20μmol)和底物24-1(21.76mg,122.10μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物24(4mg,6.92μmol)。1H NMR(400MHz,DMSO-d6)δ10.15(s,1H),8.83(s,1H),7.91(d,J=12.4Hz,1H),7.71(d,J=12.4Hz,1H),7.61(s,2H),6.93(d,J=13.2Hz,2H),5.74–5.64(m,1H),5.04–5.00(m,1H),4.95(s,1H),4.90–4.85(m,1H),4.77(s,1H),4.59–4.53(m,1H),3.76–3.73(m,4H),3.08–3.06(m,4H),3.01(d,J=25.2Hz,1H),2.74(d,J=24.4Hz,1H),1.81–1.72(m,1H),1.61–1.52(m,1H),0.99–0.94(m,1H),0.73–0.68(m,5H),0.40–0.35(m,1H).LCMS(ESI)m/z:540.2[M+H]+,HPLC method B:RT=7.49min,purity>93.2%。
实施例25:化合物25的合成
步骤1:化合物25-2的合成:
在干燥的单口瓶中加入底物25-1(332mg,1.56mmol),溶于二甲基亚砜(5mL)中,然后加入对氟硝基苯(242.73mg,1.72mmol)和碳酸钾(648.42mg,4.69mmol),80℃下反应12个小时。LC-MS监测反应完全。将反应液冷却至室温,加水,然后用乙酸乙酯萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物25-2(512mg,1.54mmol)。LCMS(ESI)m/z:278.1[M+H]+
步骤2:化合物25-3的合成:
在干燥的单口瓶中加入底物25-2(640mg,1.92mmol),溶于甲醇(10mL)中,然后加入钯炭(60mg),在氢气环境下室温反应12个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物25-3(570mg,1.88mmol)。LCMS(ESI)m/z:248.2[M+H]+
步骤3:化合物25-4的合成:
在干燥的三口瓶中加入底物25-3(200mg,659.20μmol),加入无水四氢呋喃(5mL)溶解,氮气保护下冷却至零度,然后缓慢滴加氢化铝锂的四氢呋喃溶液(3.3mL,3.30mmol,1M), 65度加热回流反应四个小时。LC-MS监测。向反应液中加入水,10%氢氧化钠水溶液,搅拌半个小时,用无水硫酸钠干燥,加硅藻土过滤,减压浓缩,残余物用柱层析纯化得化合物25-4(89mg,409.55μmol)。LCMS(ESI)m/z:218.2[M+H]+
步骤4:化合物25的合成:
在干燥的单口瓶中,加入底物18-2(10mg,24.42μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(7.59mg,43.96μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(31.56mg,244.20μmol)和底物25-4(10.60mg,48.78μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物25(2mg,2.98μmol)。1H NMR(400MHz,DMSO-d6)δ10.09(s,1H),8.81(s,1H),7.90(d,J=7.6Hz,1H),7.71(d,J=8.0Hz,1H),7.55(s,2H),6.78(d,J=8.8Hz,2H),5.73–5.62(m,1H),5.02(dd,J=10.0,1.6Hz,1H),4.95(s,1H),4.87(d,J=17.2Hz,1H),4.77(s,1H),4.61–4.49(m,1H),3.20(s,2H),3.01(d,J=16.4Hz,1H),2.76(dd,J=18.0,13.6Hz,3H),2.22(s,3H),1.98–1.90(m,2H),1.80–1.72(m,1H),1.66–1.53(m,3H),0.99–0.93(m,1H),0.70(t,J=7.2Hz,5H),0.42–0.33(m,1H).LCMS(ESI)m/z:579.4[M+H]+,HPLC method B:RT=7.93min,purity>86.1%。
实施例26:化合物26的合成
步骤1:化合物26-2的合成:
在干燥的单口瓶中加入底物26-1(200mg,857.39μmol),溶于甲醇(5mL)中,然后加入钯炭(20mg),在氢气环境下室温反应12个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物26-2(170mg,836.28μmol)。LCMS(ESI)m/z:204.1[M+H]+
步骤2:化合物26的合成:
在干燥的单口瓶中,加入底物18-2(10mg,24.42μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(9.27mg,53.72μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(15.78mg,122.10μmol)和底物26-2(10.92mg,53.72μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物26(2.03mg,3.59μmol)。1H NMR(400MHz,DMSO-d6)δ10.02(s,1H),8.79(s,1H),7.88(d,J=8.0Hz,1H),7.70(d,J=8.0Hz,1H),7.51–7.48(m,2H),6.55(d,J=8.8Hz,2H),5.73–5.64(m,1H),5.03–5.00(m,1H),4.95(s,1H),4.88(d,J=17.2Hz,1H),4.80–4.77(m 1H),4.58–4.52(m,1H),4.26(s,1H),3.41(s,1H),3.33–3.30(m,1H),3.12(d,J=9.2Hz,1H),3.01(d,J=16.4Hz,1H),2.78–2.72(m,2H),2.47–2.44(m,1H),2.23(s,3H),1.84(d,J=9.2Hz,1H),1.79–1.74(m,1H),1.61–1.52(m,1H),0.98–0.93(m,1H),0.72–0.68(m,5H),0.39– 0.34(m,1H).LCMS(ESI)m/z:565.3[M+H]+,HPLC method B:RT=7.65min,purity>95.9%。
实施例27:化合物27的合成
在干燥的单口瓶中,加入底物18-2(30mg,73.26μmo),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(25.28mg,146.52μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(94.68mg,732.59μmol)和底物11-1(38.01mg,146.52μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物27(15.00mg,22.64μmol)。1H NMR(400MHz,DMSO-d6)δ10.11(s,1H),8.82(s,1H),7.91(d,J=8.0Hz,1H),7.71(d,J=8.0Hz,1H),7.57(s,2H),6.91(d,J=9.2Hz,2H),5.74–5.63(m,1H),5.03–5.00(m,1H),4.95(s,1H),4.87(dd,J=17.3,1.7Hz,1H),4.77(s,1H),4.55(dd,J=16.0,6.7Hz,1H),3.08(t,J=5.7Hz,4H),3.01(d,J=16.6Hz,1H),2.74(d,J=16.6Hz,1H),2.28(t,J=5.4Hz,4H),2.15(s,3H),1.79–1.74(m,7.3Hz,1H),1.62–1.50(m,5H),1.46(t,J=5.6Hz,4H),0.97–0.93(m,1H),0.73–0.68(m,5H),0.40–0.36(m,1H).LCMS(ESI)m/z:621.2[M+H]+,HPLC method B:RT=8.72min,purity>93.7%。
实施例28:化合物28的合成
步骤1:化合物28-2的合成:
在干燥的单口瓶中加入底物28-1(500mg,2.36mmol),溶于N,N-二甲基甲酰胺(7mL)中,然后加入对氟硝基苯(302.12mg,2.14mmol)和碳酸钾(887.73mg,6.42mmol),90℃下反应12个小时。LC-MS监测反应完全。将反应液冷却至室温,加水,然后用乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物28-2(670mg,2.01mmol)。LCMS(ESI)m/z:278.1[M+H-56]+
步骤2:化合物28-3的合成:
在干燥的单口瓶中加入底物28-2(670mg,2.01mmol),溶于二氯甲烷(5mL)中,加入三氟乙酸(2.5mL),室温下反应2个小时。LC-MS监测反应完全。将反应液减压浓缩得到粗品化合物28-3(670mg,2.87mmol)。LCMS(ESI)m/z:234.2[M+H]+
步骤3:化合物28-4的合成:
在干燥的单口瓶中加入底物28-3(670mg,2.87mmol),溶于甲醇(8mL)中,加入醋酸(344.96mg,5.74mmol)和甲醛水溶液(862.42mg,28.72mmol,37%),室温搅拌半个小时,然后加入氰基硼氢化钠(360.99mg,5.74mmol),50℃下反应3个小时。LC-MS监测反应完全。将反应液减压浓缩,加入饱和碳酸氢钠水溶液调节至pH=9,然后水相用乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物28-4(520mg,2.10mmol)。LCMS(ESI)m/z:248.2[M+H]+
步骤4:化合物28-5的合成:
在干燥的单口瓶中加入底物28-4(520mg,2.10mmol),溶于甲醇(7mL)中,然后加入钯炭(50mg),在氢气环境下室温反应三个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物28-5(400mg,1.84mmol)。LCMS(ESI)m/z:218.1[M+H]+
步骤5:化合物28的合成:
在干燥的单口瓶中,加入底物18-2(15mg,36.63μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(13.91mg,80.59μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(47.34mg,366.30μmol)和底物28-5(17.51mg,80.59μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物28(8.09mg,11.21μmol)。1H NMR(400MHz,DMSO-d6)δ10.02(s,1H),8.80(s,1H),7.88(d,J=7.6Hz,1H),7.71(d,J=8.4Hz,1H),7.54(s,2H),6.63(d,J=8.8Hz,2H),5.74–5.64(m,1H),5.03–5.00(m,1H),4.95(s,1H),4.89–4.85(m,1H),4.80–4.77(m,1H),4.58–4.53(m,1H),3.31(s,2H),3.06–2.99(m,3H),2.89–2.84(m,2H),2.74(d,J=17.2Hz,1H),2.58–2.55(m,2H),2.40–2.37(m,2H),2.22(s,3H),1.79-1.74(m,1H),1.59–1.54(m,1H),0.98–0.94(m,1H),0.72–0.68(m,5H),0.39–0.31(m,1H).LCMS(ESI)m/z:579.4[M+H]+,HPLC method B:RT=8.17,purity>80.2%。
实施例29:化合物29的合成
步骤1:化合物29-2的合成:
在干燥的三口瓶中加入氢化钠(696.50mg,17.41mmol,60%),加入N,N-二甲基甲酰胺(50mL)溶解,氮气保护下冷却至零度,然后缓慢滴加底物29-1(1.05g,4.98mmol)和1,2-二溴乙烷(3.27g,17.41mmol)的N,N-二甲基甲酰胺(20mL)溶液,保持温度反应2个小时。LC-MS监测。向反应液中加入饱和氯化铵水溶液,然后用二氯甲烷萃取四次,有机相用水洗涤三次,饱和食盐水洗涤一次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物29-2(820mg,3.46mol)。LCMS(ESI)m/z:238.2[M+H]+
步骤2:化合物29-3的合成:
在干燥的三口瓶中加入底物29-2(800mg,3.46mol),加入四氢呋喃(10mL)溶解,氮气保护下冷却至零度,然后缓慢滴加乙基溴化镁的四氢呋喃溶液(2M,6.92mL),保持温度反应1个小时。LC-MS监测。向反应液中加入饱和氯化铵水溶液,用乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得化合物29-3(665mg,2.49mmol)。LCMS(ESI)m/z:268.1[M+H]+
步骤3:化合物29-4的合成:
在干燥的微波管中,加入底物29-3(30mg,112.29μmol)、底物IM-1(26.21mg,117.91μmol)、碘化亚铜(42.77mg,224.58μmol)、碘化钠(33.66mg,224.58μmol)、碳酸钾(38.80mg,280.73μmol)和反-(1R,2R)-N,N'-二甲基1,2-环己烷二胺(63.89mg,449.17μmol),然后加入苯甲醚(1mL),在氮气环境下,130度微波加热反应3.5个小时。LC-MS监测。将反应液冷却至室温,过滤,减压浓缩,残余物用柱层析纯化得到化合物29-4(8mg,19.58μmol)。LCMS(ESI)m/z:409.1[M+H]+
步骤4:化合物29的合成:
在干燥的单口瓶中,加入底物29-4(8mg,19.58μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(6.08mg,35.25μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(25.31mg,195.83μmol)和底物4-(4-甲基哌嗪)苯胺(7.49mg,39.17μmol),45度反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物29(2.9mg,4.38μmol)。1H NMR(400MHz,DMSO-d6)δ10.05(s,1H),8.80(s,1H),7.55(s,2H),7.41–7.29(m,2H),7.24(s,1H),6.85(d,J=9.2Hz,2H),5.74–5.63(m,1H),5.13–5.07(m,1H),4.94(d,J=17.2Hz,1H),4.86(s,1H),4.23(d,J=12.0Hz,1H),3.05(t,J=5.2Hz,4H),2.45(d,J=4.8Hz,4H),2.22(s,3H),1.67(dd,J=13.9,7.2Hz,1H),1.59–1.54(m,1H),1.23(s,3H),0.93(d,J=6.4Hz,1H),0.72(t,J=7.2Hz,3H),0.67(d,J=7.2Hz,2H),0.33(t,J=5.2Hz,1H).LCMS(ESI)m/z:552.3[M+H]+,HPLC method B:RT=7.57,purity> 83.3%。
实施例30:化合物30的合成
步骤1:化合物30-2的合成:
在干燥的单口瓶中加入底物30-1(230mg,2.01mmol),溶于二甲基亚砜(5mL)中,然后加入对氟硝基苯(298.42mg,2.11mmol)和碳酸钾(1.39g,10.07mmol),100℃下反应4个小时。LC-MS监测反应完全。将反应液冷却至室温,加水,然后用乙酸乙酯萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物30-2(450mg,1.91mmol)。LCMS(ESI)m/z:236.2[M+H]+
步骤2:化合物30-3的合成:
在干燥的单口瓶中加入底物30-2(450mg,1.91mmol),溶于甲醇(3mL)中,然后加入钯炭(40mg),在氢气环境下室温反应2个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物30-3(380mg,1.85mmol)。LCMS(ESI)m/z:206.1[M+H]+
步骤3:化合物30的合成:
在干燥的单口瓶中,加入底物18-2(20mg,48.84μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(15.17mg,87.91μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(63.12mg,488.40μmol)和底物30-3(20.05mg,97.68μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物30(4.9mg,8.32μmol)。1H NMR(400MHz,DMSO-d6)δ10.14(s,1H),8.83(s,1H),7.92(s,1H),7.71(d,J=8.0Hz,1H),7.58(s,2H),6.92(d,J=8.8Hz,2H),5.73–5.63(m,1H),5.02(dd,J=10.2,1.5Hz,1H),4.97(s,1H),4.87(d,J=17.2Hz,1H),4.78(s,1H),4.56(d,J=15.6Hz,1H),3.48(s,2H),3.02(d,J=16.4Hz,1H),2.84–2.69(m,4H),2.37–2.30(m,2H),2.21(s,3H),1.77(dd,J=13.6,7.2Hz,1H),1.56(dd,J=13.6,7.2Hz,1H),1.05(d,J=6.0Hz,3H),0.98–0.91(m,1H),0.70(t,J=7.2Hz,5H),0.42–0.31(m,1H).LCMS(ESI)m/z:567.3[M+H]+,HPLC method B:RT=7.57,purity>96.1%。
实施例31:化合物31的合成
步骤1:化合物31a&31b的合成:
在干燥的单口瓶中,加入底物18-2(20mg,48.84μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(15.18mg,87.91μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(63.00mg,488.40μmol)和底物31-1(12.09mg,58.61μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物31a(5.1mg,8.92μmol);SFC保留时间t=2.264min。1H NMR(400MHz,DMSO-d6)δ10.16(s,1H),8.83(s,1H),7.91(s,1H),7.72–7.70(m,1H),7.60(s,2H),6.94–6.92(d,J=8.8Hz,2H),5.74–5.64(m,1H),5.03–5.00(m,1H),4.97(s,1H),4.89–4.85(m,1H),4.82–4.72(m,1H),4.58–4.53(m,1H),3.74–3.66(m,2H),3.55–3.50(m,2H),3.05–2.98(m,1H),2.76–2.72(m,1H),2.24–2.18(m,2H),1.79–1.74(m,1H),1.59–1.53(m,1H),1.19–1.26(m,6H),0.98–0.93(m,1H),0.72–0.68(m,5H),0.41–0.32(m,1H).LCMS(ESI)m/z:568.4[M+H]+,HPLC method B:RT=8.29min,purity>99.3%。
化合物31b(5.1mg,17.60μmol);SFC保留时间t=3.730min。1H NMR(400MHz,DMSO-d6)δ10.16(s,1H),8.83(s,1H),8.00-7.88(m,1H),7.72–7.70(m,1H),7.66–7.55(m,2H),6.94–6.91(d,J=8.8Hz,2H),5.74–5.64(m,1H),5.03–5.00(m,1H),4.97(s,1H),4.89–4.85(m,1H),4.85–4.72(m,1H),4.63–4.50(m,1H),3.74–3.66(m,2H),3.55–3.50(m,2H),3.06–2.98(m,1H),2.79–2.71(m,1H),2.26–2.17(m,2H),1.82–1.72(m,1H),1.61–1.51(m,1H),1.19–1.12(m,6H),0.98–0.92(m,1H),0.74–0.66(m,5H),0.43–0.32(m,1H).LCMS(ESI)m/z:568.3[M+H]+,HPLC method B:RT=8.30min,purity>95.7%。
实施例32:化合物32的合成
步骤1:化合物32的合成:
在干燥的三口瓶中加入底物3-2(200mg,1.03mol),加入四氢呋喃(5mL)溶解,氮气保护下冷却至0℃,然后缓慢滴加(三氟甲基)三甲基硅烷(732.28mg,5.15mol)和四丁基氟化铵(1.03mL,1M),保持温度反应1个小时。LC-MS监测。将反应液减压浓缩,残余物用柱层析纯化得化合物32-1(220mg,834.47μmol)。LCMS(ESI)m/z:264.1[M+H]+
步骤2:化合物32-2的合成:
在干燥的微波管中,加入底物32-1(220mg,834.47μmol)、底物IM-1(194.75mg,876.19μmol)、碘化亚铜(318.05mg,1.67mmol)、碘化钠(250.32mg,1.67mmol)、碳酸钾(288.86mg,2.09mmol)和反-(1R,2R)-N,N'-二甲基1,2-环己烷二胺(475.08mg,3.34mmol),然后加入苯甲醚(4mL),在氮气环境下,130℃微波加热反应3.5个小时。LC-MS监测。将反应液冷却至室温,过滤,减压浓缩,残余物用柱层析纯化得到化合物32-2(150mg,333.74μmol)。LCMS(ESI)m/z:450.2[M+H]+
步骤3:化合物32的合成:
在干燥的单口瓶中,加入底物32-2(20mg,44.50μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(13.82mg,80.10μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(57.40mg,444.99μmol)和底物4-(4-甲基哌嗪)苯胺(1-8,10.21mg,53.40μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物:32(6.2mg,9.65μmol)。1H NMR(400MHz,DMSO-d6)δ10.21(s,1H),8.84(s,1H),8.14–8.00(m,1H),7.94–7.87(m,1H),7.67–7.50(m,2H),6.96–6.90(m,2H),6.80–6.60(m,1H),5.74–5.60(m,1H),5.03–4.94(m,1H),4.90–4.72(m,2H),4.63–4.50(m,1H),3.14–3.04(m,4H),2.78–2.69(m,1H),2.48–2.41(m,4H),2.22(s,3H),1.94–1.76(m,1H),1.09–0.99(m,2H),0.94–0.86(m,1H),0.52–0.44(m,1H).LCMS(ESI)m/z:593.3[M+H]+,HPLC method B:RT=7.79min,purity>92.3%。
实施例33:化合物33的合成
步骤1:化合物33-2的合成:
在干燥的单口瓶中加入底物33-1(160mg,668.77μmol),溶于甲醇(5mL)中,然后加入钯炭(20mg),在氢气环境下室温反应2个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物33-2(68mg,324.95μmol)。LCMS(ESI)m/z:210.2[M+H]+
步骤2:化合物33的合成:
在干燥的单口瓶中,加入底物18-2(20mg,36.63μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(16.55mg,65.93μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(45.56mg,352.52μmol)和底物33-2(30mg,143.36μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物33(6.37mg,11.16μmol)。1H NMR(400MHz,Chloroform-d)δ8.78(s,1H),7.81–7.70(m,3H),6.99–6.85(m,2H),5.75–5.65(m,1H),5.03(d,J=10.4Hz,1H),4.95–4.85(m,2H),4.70–4.64(m,1H),3.18–3.10(m,5H),2.80–2.76(m,5H),2.47(s,3H),1.99–1.90(m,1H),1.79–1.70(m,1H),1.19–1.14(m,1H),0.94–0.89(m,1H),0.85–0.79(m,4H),0.51–0.46(m,1H).LCMS(ESI)m/z:571.3[M+H]+,HPLC method B:RT=7.71min,purity>87.6%。
实施例34:化合物34的合成
步骤1:化合物34-2的合成:
在干燥的单口瓶中加入底物34-1(100.00g,904.08mmol),然后加入碘甲烷(256.65g,1.81mol)室温反应48个小时,有黄色固体产生。向反应液中加入甲基叔丁基醚,打浆、过滤、真空干燥得到化合物34-2(189.00g,748.40mmol)。
步骤2:化合物4-2的合成:
在干燥的三口瓶中加入底物4-1(30.00g,203.84mmol),然后加入四氢呋喃(750mL)和叔丁醇(1500mL)溶解,在氮气保护下冷却至零下40℃。向反应液中滴加叔丁醇钾(73.19g,652.29mmol)的四氢呋喃(650mL)溶液,保持温度不变反应四十分钟,然后加入三乙基苄基氯化铵(4.64g,20.38mmol)和底物34-2(77.22g,305.72mmol),在室温下反应18个小时,TLC监测反应完全。向反应液中缓慢加入饱和氯化铵溶液,减压浓缩除去有机相,水相用二氯甲烷萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析(石油醚:乙酸乙酯=1:1洗脱)纯化得到化合物4-2(11.00g,63.51mmol)。LCMS(ESI+)m/z:174.1[M+H]+
化合物4-3~4-8的合成请参考实施例4中的合成方法
步骤3:化合物34-3的合成:
在干燥的单口瓶中加入底物4-8(27.50g,131.12mmol),加入二氯甲烷(300mL)溶解,冰浴冷却下加入戴斯马丁氧化剂(83.42g,196.68mmol),室温反应两个小时,TLC监测反应完全。向反应液中加入饱和碳酸氢钠水溶液调节pH到8,然后用二氯甲烷萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析(石油醚:乙酸乙酯=2:1洗脱)纯化 得到化合物34-3(18.55g,89.33mmol)。LCMS(ESI+)m/z:208.1[M+H]+
步骤4:化合物34-4的合成:
在干燥的单口瓶中加入底物34-3(18.55g,89.33mmol),然后加入甲醇(270mL)溶解,冷却至零度,缓慢加入硼氘化钠(4.48g,107.20mmol)保持温度不变反应1个小时,TLC监测反应完全。将反应液减压浓缩,残余物加入水,用二氯甲烷萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物34-4(18.00g,85.44mmol)。LCMS(ESI+)m/z:211.0[M+H]+1H NMR(400MHz,DMSO-d6)δ7.63(d,J=8.0Hz,1H),7.32(d,J=8.0Hz,1H),5.35(s,1H),2.86–2.67(m,2H),2.22–2.15(m,1H),1.12–1.06(m,1H),0.73–0.62(m,1H),0.44–0.40(m,1H),0.38–0.30(m,2H)。
步骤5:化合物34-5的合成:
在干燥的密封管中加入底物34-4(5g,23.73mmol)、底物IM-1(5.55g,24.92mmol)、碘化亚铜(4.5g,23.73mmol)、碘化钠(7.14g,47.46mmol)、碳酸钾(8.2g,59.33mmol)和反-(1R,2R)-N,N'-二甲基1,2-环己烷二胺(6.76g,47.46mmol),然后加入苯甲醚(80mL),氮气环境下,在110℃反应18个小时,TLC监测反应完全。将反应液冷却至室温,过滤,用乙酸乙酯洗涤两次,滤液用氨水洗涤两次,饱和食盐水洗涤两次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析(石油醚:乙酸乙酯=1:1洗脱)纯化得到化合物34-5(4.60g,11.60mmol)。LCMS(ESI+)m/z:397.1[M+H]+
步骤6:化合物34&34a&34b的合成:
在干燥的单口瓶中,加入底物34-5(57mg,143.77μmol),溶于四氢呋喃(5mL)中,然后加入间氯过氧苯甲酸(44.66mg,258.78μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(46mg,177.34μmol)和底物7-5(43.84mg,215.66μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物34(4.2mg,6.40μmol)。1H NMR(400MHz,DMSO-d6)δ10.06(s,1H),8.78(s,1H),7.81(d,J=8.4Hz,1H),7.72(d,J=8.4Hz,1H),7.66–7.45(m,2H),6.71(d,J=8.8Hz,2H),5.73–5.64(m,1H),5.28(s,1H),5.00(dd,J=10.0,1.6Hz,1H),4.89(dd,J=17.2,1.6Hz,1H),4.74–4.56(m,2H),3.59(d,J=6.0Hz,2H),3.44(d,J=11.2Hz,2H),3.29–3.25(m,2H),2.88–2.80(m,2H),2.47–2.39(m,1H),2.29–2.22(m,1H),2.00(s,3H),1.54(d,J=8.4Hz,1H),1.16–1.08(m,1H),0.71–0.64(m,1H),0.45–0.40(m,1H),0.40–0.31(m,2H).LCMS(ESI)m/z:552.3[M+H]+,HPLC method B:RT=7.26min,purity>84.0%。
按照同样的方法合成34,再经过超临界液相色谱手型拆分得到化合物34a(10.1mg,18.31μmol);SFC保留时间t=4.312min。1H NMR(600MHz,DMSO-d6)δ10.07(s,1H),8.78(s,1H),7.81(d,J=8.4Hz,1H),7.72(d,J=8.4Hz,1H),7.61(s,2H),6.71(d,J=8.4Hz,2H),5.74–5.64(m,1H),5.29(s,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.70(s,1H),4.64–4.54(m,1H),3.59(d,J=6.0Hz,2H),3.44(d,J=11.4Hz,2H),3.28(d,J=10.8Hz,2H),2.92–2.81(m,2H),2.43(d,J=7.2Hz,1H),2.41–2.29(m,1H),2.00(s,3H),1.54(d,J=8.4Hz,1H),1.15–1.07(m,1H),0.72–0.63(m,1H),0.45–0.41(m,1H),0.40–0.31(m,2H).LCMS(ESI)m/z:552.3[M+H]+,HPLC method B:RT=7.29min,purity>94.5%。
化合物34b(10.5mg,19.03μmol);SFC保留时间t=7.619min。1H NMR(600MHz,DMSO-d6)δ10.07(s,1H),8.79(s,1H),7.81(d,J=8.4Hz,1H),7.72(d,J=8.4Hz,1H),7.61(s,2H),6.71(d,J=9.0Hz,2H),5.73–5.63(m,1H),5.40–5.18(m,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.76–4.54(m,2H),3.59(d,J=6.0Hz,2H),3.44(d,J=11.4Hz,2H),3.28(d,J=10.8Hz,2H),2.91–2.81(m,2H),2.44(d,J=7.2Hz,1H),2.41–2.30(m,1H),2.00(s,3H),1.54(d,J=8.4Hz,1H),1.14–1.08(m,1H),0.71–0.63(m,1H),0.45–0.41(m,1H), 0.40–0.31(m,2H).LCMS(ESI)m/z:552.3[M+H]+,HPLC method B:RT=7.38min,purity>84.4%。
实施例35:化合物35的合成
步骤1:化合物35a&35b的合成
在干燥的单口瓶中,加入底物6-4(40mg,97.68μmol),溶于四氢呋喃(4mL)中,然后加入间氯过氧苯甲酸(30.34mg,175.82μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(126.24mg,976.79μmol)和底物35-1(25.22mg,131.87μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化得到产物,再经过超临界液相色谱手型拆分得到化合物35a(10.0mg,14.69μmol);SFC保留时间t=3.112min。1H NMR(400MHz,DMSO-d6)δ10.05(s,1H),8.79(s,1H),7.79–7.67(m,2H),7.59(s,2H),6.69(d,J=8.8Hz,2H),5.74–5.62(m,1H),5.02–4.98(m,1H),4.90–4.73(m,2H),4.68(s,2H),3.71(d,J=6.0Hz,2H),3.42(d,J=11.2Hz,2H),3.24(d,J=11.2Hz,2H),2.93–2.75(m,2H),2.49–2.44(m,1H),2.35(q,J=6.4Hz,1H),2.04–1.93(m,1H),1.50(d,J=8.0Hz,1H),1.40(s,4H),0.88(d,J=6.0Hz,7H),0.66–0.58(m,1H),0.36–0.27(m,1H),0.25–0.17(m,1H).LCMS(ESI)m/z:593.2[M+H]+,HPLC method B:RT=8.55min,purity>85.5%。
化合物35b(10.0mg,14.21μmol);SFC保留时间t=4.854min。1H NMR(400MHz,DMSO-d6)δ10.05(s,1H),8.79(s,1H),7.87–7.67(m,2H),7.59(s,2H),6.69(d,J=8.8Hz,2H),5.74–5.64(m,1H),4.99(dd,J=10.4,1.6Hz,1H),4.91–4.75(m,2H),4.72–4.57(m,2H),3.71(d,J=6.0Hz,2H),3.42(d,J=11.2Hz,2H),3.24(d,J=11.2Hz,2H),2.93–2.77(m,2H),2.49–2.44(m,1H),2.35(q,J=6.4Hz,1H),2.05–1.91(m,1H),1.50(d,J=8.0Hz,1H),1.40(s,4H),0.88(d,J=6.0Hz,7H),0.65–0.58(m,1H),0.34–0.27(m,1H),0.23–0.17(m,1H).LCMS(ESI)m/z:593.2[M+H]+,HPLC method B:RT=8.59min,purity>84.0%。
实施例36:化合物36的合成
步骤1:化合物36a&36b的合成
在干燥的单口瓶中,加入底物21-3(45mg,106.25μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(33.00mg,191.25μmol),室温下反应1个小时,再向反应液中加 入N,N-二异丙基乙胺(137.32mg,1.06mmol)和底物35-1(49.16mg,212.50μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化得到产物,再经过超临界液相色谱手型拆分得到化合物36a(14.24mg,23.47μmol);SFC保留时间t=2.868min。1H NMR(400MHz,DMSO-d6)δ10.05(s,1H),8.78(s,1H),7.77(d,J=8.8Hz,1H),7.68(d,J=8.4Hz,1H),7.58(s,2H),6.69(d,J=8.8Hz,2H),5.74–7.63(m,1H),5.03–4.96(m,1H),4.85(d,J=17.2Hz,1H),4.75(d,J=16.0Hz,1H),4.61(d,J=15.6Hz,1H),4.40(s,1H),3.71(d,J=6.0Hz,2H),3.42(d,J=11.2Hz,2H),3.24(d,J=11.2Hz,2H),2.95–2.80(m,1H),2.83(dd,J=17.2,6.0Hz,1H),2.47(d,J=6.0Hz,1H),2.38–2.28(m,2H),1.95–1.88(m,1H),1.79–1.71(m,1H),1.50(d,J=8.0Hz,1H),1.17–1.11(m,1H),0.93(t,J=7.2Hz,3H),0.88(d,J=6.0Hz,7H),0.61(d,J=5.6Hz,1H),0.43–0.38(m,1H),0.00(s,1H).LCMS(ESI)m/z:607.2[M+H]+,HPLC method B:RT=9.29min,purity>89.7%。
化合物36b(13.16mg,21.69μmol);SFC保留时间t=3.896min。1H NMR(400MHz,DMSO-d6)δ10.05(s,1H),8.78(s,1H),7.77(d,J=8.4Hz,1H),7.68(d,J=8.2Hz,1H),7.59(s,2H),6.69(d,J=8.6Hz,2H),5.72–5.60(m,1H),5.00(dd,J=10.2,1.6Hz,1H),4.85(d,J=17.1Hz,1H),4.75(d,J=15.3Hz,1H),4.62(s,1H),4.40(s,1H),3.71(d,J=5.9Hz,2H),3.42(d,J=11.0Hz,2H),3.24(d,J=11.0Hz,2H),2.95–2.80(m,1H),2.83(dd,J=17.0,5.9Hz,1H),2.49–2.44(m,1H),2.38–2.28(m,2H),1.95–1.88(m,1H),1.79–1.71(m,1H),1.50(d,J=8.0Hz,1H),1.16–1.11(m,1H),0.93(t,J=7.4Hz,3H),0.88(d,J=6.0Hz,6H),0.85(d,J=5.8Hz,1H),0.65–0.58(m,1H),0.43–0.38(m,1H),-0.00(s,1H).LCMS(ESI)m/z:607.2[M+H]+,HPLC method B:RT=9.31min,purity>87.9%。
实施例37:化合物37的合成
步骤20:化合物37a&37b的合成:
在干燥的单口瓶中,加入底物34-5(4.10g,10.35mmol),溶于四氢呋喃(55mL)中,然后加入间氯过氧苯甲酸(3.79g,18.63mmol),室温下反应一个小时,再向反应液中加入N,N-二异丙基乙胺(5.33g,41.29mmol)和底物11-1(3.22g,12.41mmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化(乙腈/0.01%碳酸氢铵水溶液洗脱,60毫升/分钟)得到得到化合物,化合物再经超临界液相色谱手型拆分得到化合物37a(1.30g,2.14mmol),SFC保留时间t=5.043min。LCMS(ESI+)m/z:608.2[M+H]+,1H NMR(600MHz,DMSO-d6)δ10.12(s,1H),8.80(s,1H),7.83(s,1H),7.70(d,J=8.4Hz,1H),7.64–7.42(m,2H),6.91(d,J=9.0Hz,2H),5.70–5.64(m,1H),5.30(s,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.69–4.62(m,2H),3.11–3.07(m,4H),2.86–2.81(m,2H),2.27(q,J=12.6,9.0Hz,5H),2.15(s,3H),1.53(t,J=5.4Hz,4H),1.46(t,J=5.4Hz,4H),1.13–1.10(m,1H),0.69–0.65(m,1H),0.49–0.42(m,1H),0.39–0.32(m,2H),HPLC method B:RT=8.83min,purity>94.1%。
化合物37b(1.50g,2.47mmol),SFC保留时间t=9.666min。LCMS(ESI+)m/z:608.2[M+H]+,1H NMR(600MHz,DMSO-d6)δ10.11(s,1H),8.80(s,1H),7.83(s,1H),7.70(d,J=8.4Hz,1H),7.66–7.47(m,2H),6.91(d,J=8.4Hz,2H),5.70–5.64(m,1H),5.29(s,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.69(s,1H),4.62(d,J=15.0Hz,1H),3.09(t,J=5.4Hz,4H),2.88–2.79(m,2H),2.29(d,J=6.0Hz,4H),2.24(d,J=17.4Hz,1H),2.16(s,3H),1.54(t,J=5.4Hz,4H),1.47(t,J=5.4Hz,4H),1.13–1.09(m,1H),0.69–0.66(m,1H),0.46–0.42(m,1H),0.39–0.33(m,2H),HPLC method B:RT=8.09min,purity>96.5%。
实施例38:化合物38的合成
步骤1:化合物38a&38b的合成:
在干燥的单口瓶中,加入底物4-9(80mg,202.29μmol),溶于四氢呋喃(10mL)中,然后加入间氯过氧苯甲酸(61.60mg,303.43μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(260.58mg,2.02mmol)和底物11-1(78.71mg,303.43μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物38a(32.43mg,53.45μmol);SFC保留时间t=4.513min。1H NMR(600MHz,DMSO-d6)δ10.11(s,1H),8.80(s,1H),7.83(d,J=7.8Hz,1H),7.70(d,J=8.4Hz,1H),7.58(s,2H),6.91(d,J=8.4Hz,2H),5.67–5.65(m,1H),5.31(d,J=5.4Hz,1H),5.01–4.99(m,1H),4.93–4.85(m,1H),4.69(s,1H),4.64–4.64(m,1H),3.75(d,J=5.4Hz,1H),3.09–3.07(m,4H),2.87–2.80(m,2H),2.30–2.26(m,4H),2.24–2.22(m,1H),2.16(s,3H),1.54–1.52(m,4H),1.47–1.45(m,4H),1.14–1.10(m,1H),0.69–0.67(m,1H),0.46–0.41(m,1H),0.41–0.30(m,2H).LCMS(ESI)m/z:607.4[M+H]+,HPLC method B:RT=8.39min,purity>94.4%。
化合物38b(32.44mg,53.46μmol);SFC保留时间t=8.668min。1H NMR(600MHz,DMSO-d6)δ10.11(s,1H),8.80(s,1H),7.83(d,J=7.8Hz,1H),7.70(d,J=8.4Hz,1H),7.58(s,2H),6.91(d,J=8.4Hz,2H),5.67–5.55(m,1H),5.31(d,J=5.4Hz,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.69(s,1H),4.65–4.52(m,1H),3.75(d,J=5.4Hz,1H),3.09–3.07(m,4H),2.87–2.80(m,2H),2.84–2.77(m,1H),2.35–2.26(m,4H),2.26–2.22(m,1H),2.17(s,3H),1.54–1.52(m,4H),1.47–1.45(m,4H),1.14–1.10(m,1H),0.68–0.66(m,1H),0.46–0.42(m,1H),0.37–0.35(m,2H).LCMS(ESI)m/z:607.4[M+H]+,HPLC method B:RT=8.40min,purity>95.7%。
实施例39:化合物39的合成
步骤1:化合物39a&39b的合成
在干燥的单口瓶中,加入底物34-5(40mg,100.89μmol),溶于四氢呋喃(5mL)中,然后加入间氯过氧苯甲酸(36.87mg,181.60μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(130.39mg,1.01mmol)和底物35-1(46.68mg,201.77μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化得到产物,再经过超临界液相色谱手型拆分得到化合物39a(10.1mg,16.50μmol);SFC保留时间t=3.831min。1H NMR(600MHz,DMSO-d6)δ10.06(s,1H),8.78(s,1H),7.81(d,J=8.4Hz,1H),7.71(d,J=8.4Hz,1H),7.65–7.41(m,2H),6.69(d,J=8.4Hz,2H),5.70–5.60(m,1H),5.29(s,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.77–4.64(m,1H),4.62(d,J=15.0Hz,1H),3.71(d,J=6.0Hz,2H),3.42(d,J=11.4Hz,2H),3.25(d,J=11.4Hz,2H),2.85–2.81(m,2H),2.49–2.46(m,1H),2.35(q,J=6.8Hz,1H),2.26–2.23(m,1H),1.50(d,J=7.8Hz,1H),1.12–1.10(m,1H),0.88(d,J=6.0Hz,6H),0.69–0.66(m,1H),0.45–0.42(m,1H),0.39–0.34(m,2H).LCMS(ESI)m/z:580.2[M+H]+,HPLC method B:RT=7.43min,purity>94.7%。
化合物39b(10.4mg,17.49μmol);SFC保留时间t=6.371min。1H NMR(600MHz,DMSO-d6)δ10.06(s,1H),8.78(s,1H),7.81(d,J=8.4Hz,1H),7.71(d,J=8.4Hz,1H),7.67–7.38(m,2H),6.69(d,J=8.4Hz,2H),5.72–5.60(m,1H),5.29(s,1H),5.04–4.96(m,1H),4.89(d,J=17.4Hz,1H),4.77–4.64(m,1H),4.62(d,J=15.0Hz,1H),3.71(d,J=6.0Hz,2H),3.42(d,J=10.8Hz,2H),3.25(d,J=10.8Hz,2H),2.89–2.79(m,2H),2.48–2.44(m,1H),2.35(d,J=11.4Hz,1H),2.28–2.22(m,1H),1.50(d,J=7.8Hz,1H),1.13–1.10(m,1H),0.88(d,J=6.0Hz,6H),0.71–0.65(m,1H),0.45–0.42(m,1H),0.39–0.34(m,2H).LCMS(ESI)m/z:580.2[M+H]+,HPLC method B:RT=7.44min,purity>97.5%。
实施例40:化合物40的合成:
步骤1:化合物40-2的合成:
在干燥的单口瓶中加入底物40-1(180mg,1.58mmol),溶于二甲基亚砜(3mL)中,然后加入对氟硝基苯(233.54mg,1.66mmol)和碳酸钾(1.09g,7.88mmol),100℃下反应12 个小时。LC-MS监测反应完全。将反应液冷却至室温,加水,然后用乙酸乙酯萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物40-2(300mg,1.28mmol)。LCMS(ESI)m/z:236.2[M+H]+
步骤2:化合物40-3的合成:
在干燥的单口瓶中加入底物40-2(300mg,1.28mmol),溶于甲醇(5mL)中,然后加入钯炭(30mg),在氢气环境下室温反应2个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物40-3(230mg,1.12mmol)。LCMS(ESI)m/z:206.1[M+H]+
步骤3:化合物40的合成:
在干燥的单口瓶中,加入底物18-2(20mg,48.84μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(15.17mg,87.91μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(63.12mg,488.40μmol)和底物40-3(20.05mg,97.68μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物40(4mg,7.06μmol)。1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.82(s,1H),7.91(d,J=8.0Hz,1H),7.71(d,J=8.4Hz,1H),7.58(s,2H),6.92(d,J=8.8Hz,2H),5.75–5.64(m,1H),5.03–4.98(m,1H),4.95(s,1H),4.88(dd,J=17.2,1.6Hz,1H),4.78(d,J=16.4Hz,1H),4.56(dd,J=16.0,6.4Hz,1H),3.48(t,J=10.8Hz,2H),3.02(d,J=16.4Hz,1H),2.82(d,J=11.2Hz,1H),2.75(d,J=13.6Hz,1H),2.72–2.66(m,1H),2.38–2.30(m,1H),2.27(d,J=11.2Hz,1H),2.22(s,3H),2.13(d,J=8.8Hz,1H),1.80–1.75(m,1H),1.62–1.53(m,1H),1.06(d,J=6.0Hz,3H),0.99–0.92(m,1H),0.76–0.67(m,5H),0.42–0.33(m,1H).LCMS(ESI)m/z:567.3[M+H]+,HPLC method B:RT=7.59,purity>88.5%。
实施例41:化合物41的合成
步骤1:化合物41a&41b的合成
在干燥的单口瓶中,加入底物34-5(40mg,100.89μmol),溶于四氢呋喃(3mL)中,然后加入间氯过氧苯甲酸(36.87mg,181.60μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(130.39mg,1.01mmol)和底物41-1(47.08mg,201.77μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化得到产物,再经过超临界液相色谱手型拆分得到化合物41a(5.1mg,8.45μmol);SFC保留时间t=3.476min。1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.83(s,1H),7.84–7.78(m,1H),7.76–7.70(m,1H),7.70–7.60(m,1H),7.43–7.37(m,1H),7.00–6.94(m,1H),5.74–5.60(m,1H),5.29(s,1H),5.03–4.96(m,1H),4.93–4.83(m,1H),4.74–4.58(m,2H),3.10–3.01(m,2H),2.92–2.76(m,2H),2.62–2.53(m,2H),2.30–2.20(m,1H),1.90–1.80(m,2H),1.60–1.48(m,2H),1.15–1.14(m,1H),0.71–0.62(s,1H),0.47–0.31(m,3H)。LCMS(ESI)m/z:582.3[M+H]+,HPLC method B:RT=8.13min,purity>96.4%。
化合物41b(5.2mg,8.47μmol);SFC保留时间t=8.440min。1H NMR(400MHz, DMSO-d6)δ10.12(s,1H),8.83(s,1H),7.88–7.76(m,1H),7.76–7.69(m,1H),7.70–7.55(m,1H),7.44–7.31(m,1H),7.02–6.94(m,1H),5.74–5.60(m,1H),5.29(s,1H),5.03–4.96(m,1H),4.93-4.83(m,1H),4.77–4.56(m,2H),3.10–3.01(m,2H),2.92–2.76(m,2H),2.62–2.53(m,2H),2.30–2.20(m,1H),1.90–1.79(m,2H),1.60–1.48(m,2H),1.17–1.06(m,1H),0.71–0.62(s,1H),0.47–0.31(m,3H).LCMS(ESI)m/z:582.4[M+H]+,HPLC method B:RT=8.56min,purity>94.8%。
实施例42:化合物42的合成
步骤1:化合物42a&42b的合成:
在干燥的单口瓶中,加入底物3-4(30mg,75.86μmol),溶于四氢呋喃(1.5mL)中,然后加入间氯过氧苯甲酸(26.18mg,151.72μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(98.04mg,758.58μmol)和底物41-1(17.70mg,75.86μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物42a(4.0mg,6.39μmol);SFC保留时间t=2.339min。1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.85(s,1H),7.88(d,J=8.0Hz,1H),7.72(d,J=8.0Hz,1H),7.65(s,1H),7.43–7.40(m,1H),6.97(d,J=8.4Hz,1H),5.74–5.65(m,1H),5.03–4.99(m,2H),4.90–4.85(m,1H),4.74(d,J=16.4Hz,1H),4.62–4.57(m,1H),3.04(d,J=11.6Hz,2H),2.89(s,2H),2.60–2.49(m,2H),2.23(s,9H),1.84(d,J=12.0Hz,2H),1.58-1.50(m,2H),1.22(s,4H),0.95–0.90(m,1H),0.70–0.66(m,1H),0.61–0.56(m,1H),0.48–0.43(m,1H).LCMS(ESI)m/z:581.3[M+H]+,HPLC method B:RT=8.01min,purity>92.7%。
化合物42b(4.0mg,6.40μmol);SFC保留时间t=5.345min。1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.84(s,1H),7.88(d,J=8.0Hz,1H),7.72(d,J=8.0Hz,1H),7.65(s,1H),7.43–7.40(m,1H),6.97(d,J=8.8Hz,1H),5.74–5.65(m,1H),5.03–4.99(m,2H),4.74(d,J=16.0Hz,1H),4.62–4.57(m,1H),3.04(d,J=11.6Hz,2H),2.89(d,J=2.0Hz,2H),2.60–2.54(m,2H),2.23(d,J=4.0Hz,9H),1.84(d,J=12.4Hz,2H),1.57–1.49(m,2H),1.26–1.22(m,4H),0.95–0.90(s,1H),0.72–0.66(m,1H),0.60–0.56(m,1H),0.48–0.43(m,1H).LCMS(ESI)m/z:581.3[M+H]+,HPLC method B:RT=8.11min,purity>92.9%。
实施例43:化合物43的合成
步骤1:化合物43&43a&43b的合成:
在干燥的单口瓶中,加入底物6-4(105mg,256.41μmol),溶于四氢呋喃(5mL)中,然后加入间氯过氧苯甲酸(79.65mg,461.54μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(330.24mg,2.56mmol)和底物28-5(111.44mg,512.82μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物43(20.09mg,28.74μmol)。1H NMR(400MHz,DMSO-d6)δ10.05(s,1H),8.79(s,1H),7.77–7.68(m,2H),7.53(s,2H),6.63(d,J=8.8Hz,2H),5.72–5.62(m,1H),4.99(dd,J=10.2,1.6Hz,2H),4.88–4.79(m,2H),4.69–4.61(m,2H),3.31(s,2H),3.06–3.03(m,2H),2.93–2.76(m,4H),2.59–2.55(m,2H),2.40–2.37(m,2H),2.22(s,3H),2.02–1.95(m,1H),1.45–1.40(m,4H),1.87–0.84(m,1H),0.65–0.61(m,1H),0.34–0.30(m,1H),0.23–0.19(m,1H).LCMS(ESI)m/z:579.3[M+H]+,HPLC method B:RT=8.97min,purity>82.8%。
用同样方法合成化合物43,再经过超临界液相色谱手型拆分得到化合物43a(2.3mg,3.50μmol);SFC保留时间t=3.946min。1H NMR(600MHz,DMSO-d6)δ10.05(s,1H),8.79(s,1H),7.76(s,1H),7.69(d,J=8.4Hz,1H),7.54(s,2H),6.64(d,J=8.4Hz,2H),5.73–5.64(m,1H),4.99(dd,J=10.2,1.8Hz,1H),4.86(d,J=17.4Hz,1H),4.80(s,1H),4.68(s,1H),4.64(d,J=13.8Hz,1H),3.06–3.01(m,2H),2.90–2.84(m,3H),2.82–2.78(m,1H),2.59–2.55(m,2H),2.38(dd,J=9.0,3.0Hz,2H),2.22(s,3H),2.02–1.95(m,1H),1.45–1.42(m,1H),1.40(s,3H),0.88–0.84(m,1H),0.65–0.61(m,1H),0.34–0.30(m,1H),0.23–0.19(m,1H).LCMS(ESI)m/z:579.4[M+H]+,HPLC method B:RT=8.76min,purity>88.0%。
化合物43b(9.3mg,13.90μmol);SFC保留时间t=6.047min。1H NMR(600MHz,DMSO-d6)δ10.05(s,1H),8.79(s,1H),7.76(d,J=8.4Hz,1H),7.69(d,J=8.4Hz,1H),7.54(s,2H),6.63(d,J=8.4Hz,2H),5.72–5.62(m,1H),4.99(dd,J=10.2,1.8Hz,1H),4.86(d,J=17.4Hz,1H),4.80(s,1H),4.68(s,1H),4.67–4.59(m,1H),3.08–3.02(m,2H),2.90–2.84(m,3H),2.82–2.78(m,1H),2.57(t,J=7.8Hz,2H),2.42–2.37(m,2H),2.22(s,3H),2.02–1.95(m,1H),1.46–1.42(m,1H),1.40(s,3H),0.87–0.84(m,1H),0.66–0.61(m,1H),0.35–0.30(m,1H),0.24–0.19(m,1H).LCMS(ESI)m/z:579.4[M+H]+,HPLC method B:RT=8.81min,purity>82.1%。
实施例44:化合物44的合成
步骤1:化合物44&44a&44b的合成:
在干燥的单口瓶中,加入底物6-4(90mg,219.78μmol),溶于四氢呋喃(4mL)中,然后加入间氯过氧苯甲酸(68.27mg,395.06μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(282.51mg,2.19mmol)和底物11-1(114.02mg,439.56),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化 得到化合物44(21.5mg,34.63μmol)。1H NMR(400MHz,DMSO-d6)δ10.10(s,1H),8.81(s,1H),7.78(d,J=8.4Hz,1H),7.69(d,J=8.4Hz,1H),7.56(s,2H),6.92(d,J=8.8Hz,2H),5.73–5.62(m,1H),5.03–4.98(m,1H),4.92–4.75(m,2H),4.69–4.58(m,2H),3.09(t,J=5.6Hz,4H),2.92–2.76(m,2H),2.38–2.28(m,4H),2.19(s,3H),2.04–1.96(m,1H),1.54(t,J=5.6Hz,4H),1.47(t,J=5.6Hz,4H),1.45–1.42(m,1H),1.40(s,3H),0.88–0.82(m,1H),0.65–0.59(m,1H),0.36–0.29(m,1H),0.25–0.18(m,1H).LCMS(ESI)m/z:621.2[M+H]+,HPLC method B:RT=9.63min,purity>96.8%。
同样方法合成化合物44,再经过超临界液相色谱手型拆分得到化合物44a(1.6mg,2.23μmol);SFC保留时间t=3.819min。1H NMR(600MHz,DMSO-d6)δ10.10(s,1H),8.80(s,1H),7.82–7.73(m,1H),7.71–7.65(m,1H),7.63–7.48(m,2H),6.96–6.86(m,2H),5.72–5.62(m,1H),5.03–4.95(m,1H),4.91–4.74(m,2H),4.73–4.58(m,2H),3.29(s,1H),3.10–3.06(m,4H),2.93–2.84(m,1H),2.83–2.76(m,1H),2.36–2.25(m,3H),2.24–2.13(m,3H),2.03–1.95(m,1H),1.56–1.51(m,4H),1.50–1.45(m,4H),1.40(s,3H),1.25–1.21(m,1H),0.88–0.82(m,1H),0.66–0.58(m,1H),0.35–0.29(m,1H),0.24–0.18(m,1H)。LCMS(ESI)m/z:621.4[M+H]+,HPLC method B:RT=10.05min,purity>86.7%。
化合物44b(1.5mg,2.21μmol);SFC保留时间t=6.629min。1H NMR(600MHz,DMSO-d6)δ10.10(s,1H),8.80(s,1H),7.82–7.73(m,1H),7.71–7.65(m,1H),7.61–7.49(m,2H),6.96–6.86(m,2H),5.71–5.61(m,1H),5.03–4.95(m,1H),4.89–4.83(m,1H),4.83–4.74(m,1H),4.73–4.67(m,1H),4.67–4.59(m,1H),3.10–3.06(m,4H),2.92–2.85(m,1H),2.83–2.76(m,1H),2.32–2.27(m,3H),2.20–2.13(m,3H),2.02–1.95(m,1H),1.56–1.51(m,4H),1.50–1.45(m,4H),1.40(s,3H),1.26–1.17(m,2H),0.88–0.82(m,1H),0.66–0.58(m,1H),0.35–0.29(m,1H),0.24–0.18(m,1H)。LCMS(ESI)m/z:621.4[M+H]+,HPLC method B:RT=10.03min,purity>91.4%。
实施例45:化合物45的合成
步骤1:化合物45a&45b的合成:
在干燥的单口瓶中,加入底物6-4(40mg,97.68μmol),溶于四氢呋喃(4mL)中,然后加入间氯过氧苯甲酸(30.34mg,175.82μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(126.01mg,976.79μmol)和底物45-1(39.71mg,195.36μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物45a(8.52mg,14.66μmol);SFC保留时间t=3.602min。1H NMR(600MHz,DMSO-d6)δ10.00(s,1H),8.78(s,1H),7.79–7.62(m,2H),7.57–7.31(m,2H),6.56(d,J=7.8Hz,2H),5.72–5.60(m,1H),5.04–4.94(m,1H),4.91–4.72(m,2H),4.72–4.56(m,2H),4.27(s,1H),3.42–3.40(m,2H),3.16–3.10(m,1H),2.92–2.82(m,1H),2.82–2.74(m,2H),2.48–2.44(m,1H),2.24(s,3H),2.02–1.92(m,1H),1.90-1.81(m,1H),1.79–1.71(m,1H),1.47–1.37(m,4H),0.90–0.80(m,1H),0.66–0.58 (m,1H),0.36–0.26(m,1H),0.24–0.15(m,1H)。LCMS(ESI)m/z:565.4[M+H]+,HPLC Method B Rt=8.36min,purity>97.13%。
化合物45b(7.8mg,13.39μmol);SFC保留时间t=7.195min。1H NMR(600MHz,DMSO-d6)δ10.00(s,1H),8.78(s,1H),7.79–7.62(m,2H),7.57–7.15(m,2H),6.57–6.41(m,2H),5.72–5.60(m,1H),5.04–4.94(m,1H),4.91–4.72(m,2H),4.72–4.56(m,2H),4.27(s,1H),3.42–3.40(m,2H),3.16–3.10(m,1H),2.92–2.82(m,1H),2.82–2.74(m,2H),2.48–2.44(m,1H),2.25(s,3H),2.02–1.92(m,1H),1.90–1.81(m,1H),1.79–1.71(m,1H),1.47–1.37(m,4H),0.90–0.80(m,1H),0.66–0.58(m,1H),0.36–0.26(m,1H),0.24–0.15(m,1H)。LCMS(ESI)m/z:565.3[M+H]+,HPLC Method B Rt=8.317min,purity>96.94%。
实施例46:化合物46的合成
步骤1:化合物46-2的合成:
在干燥的单口瓶中加入底物46-1(400mg,2.02mmol),溶于二甲基亚砜(5mL)中,然后加入2-氟-5-硝基甲苯(310mg,2.00mmol)和碳酸钾(390.35mg,2.82mmol),120℃下反应6个小时。LC-MS监测反应完全。将反应液冷却至室温,加水,然后用乙酸乙酯萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物46-2(290mg,869.87μmol)。LCMS(ESI)m/z:278.2[M+H-56]+
步骤2:化合物46-3的合成:
在干燥的单口瓶中加入底物46-2(290mg,869.87μmol),溶于甲醇(5mL)中,然后加入钯炭(30mg),在氢气环境下室温反应2个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物46-3(240mg,791.03mmol)。LCMS(ESI)m/z:248.0[M+H-56]+
步骤3:化合物46-4的合成:
在干燥的三口瓶中加入底物46-3(120mg,395.52μmol),加入无水四氢呋喃(3mL)溶解,氮气保护下冷却至零度,然后缓慢滴加氢化铝锂的四氢呋喃溶液(1.98mL,1.98mmol,1M),65℃加热回流反应四个小时。LC-MS监测。向反应液中加入水,10%氢氧化钠水溶液,搅拌半个小时,用无水硫酸钠干燥,加硅藻土过滤,减压浓缩,残余物用柱层析纯化得化合物46-4(38mg,174.87μmol)。LCMS(ESI)m/z:218.2[M+H]+
步骤4:化合物46的合成:
在干燥的单口瓶中,加入底物3-4(15mg,37.93μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(7.59mg,68.27μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(48.93mg,379.30μmol)和底物46-4(16.49mg,75.86μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物46(2.0mg,3.10μmol)。1H NMR(600MHz,DMSO-d6)δ10.15(s,1H),8.85(s,1H),7.88(d,J=7.8Hz,1H),7.74–7.70(m,2H),7.48–7.42(m,1H),7.29(d,J=8.4Hz,1H),5.73–5.67(m,1H),5.03–5.00(m,2H),4.88(d,J=16.8Hz,1H),4.73–4.58(m,2H),3.52(d,J=10.8 Hz,4H),3.17(d,J=4.8Hz,1H),3.04–3.03(m,2H),2.92–2.86(m,2H),2.27(s,3H),2.20(s,3H),1.99–1.96(m,1H),1.22(s,3H),0.94–0.90(m,1H),0.70–0.67(m,1H),0.59–0.56(m,1H),0.47–0.44(m,1H).LCMS(ESI)m/z:565.3[M+H]+,HPLC method B:RT=7.66min,purity>87.6%。
实施例47:化合物47的合成
步骤1:化合物47-2的合成:
在干燥的单口瓶中加入底物47-1(300mg,1.15mmol),溶于甲醇(5mL)中,然后加入钯炭(30mg),在氢气环境下室温反应2个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物47-2(230mg,994.23μmol)。LCMS(ESI)m/z:232.2[M+H]+
步骤2:化合物47的合成:
在干燥的单口瓶中,加入底物6-4(15mg,36.63μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(11.38mg,65.93μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(47.34mg,366.30μmol)和底物47-2(16.95mg,73.26μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物47(4.0mg,6.75μmol)。1H NMR(400MHz,DMSO-d6)δ8.97–8.66(m,1H),7.79–7.45(m,3H),7.35(d,J=8.4Hz,1H),7.13(d,J=8.4Hz,1H),6.68–6.47(m,2H),5.68–5.59(m,2H),5.00(d,J=10.4Hz,1H),4.86(d,J=14.8Hz,1H),4.76–4.57(m,2H),4.16(s,3H),3.85(d,J=10.0Hz,1H),3.78–3.66(m,1H),3.59(s,3H),3.23(s,3H),2.85–2.71(m,2H),2.16(s,2H),1.99–1.89(m,3H),1.40(s,4H),0.85–0.83(m,1H),0.64–0.62(m,1H),0.34–0.31(m,1H),0.26–0.18(m,1H).LCMS(ESI)m/z:593.4[M+H]+,HPLC Method B RT=7.58min,purity>98.6%.LCMS(ESI)m/z:593.4[M+H]+,HPLC method B:RT=7.55min,purity>98.8%。
实施例48:化合物48的合成
步骤1:化合物48a&48b的合成
在干燥的单口瓶中,加入底物4-9(20mg,50.57μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(17.45mg,101.14μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(65.36mg,505.72μmol)和底物41-1(11.80mg,50.57μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化得到产物,再经过超临界液相色谱手型拆分得到化合物48a(4.93mg,7.62μmol)。1H NMR(400MHz,DMSO-d6)δ10.13(s,1H),8.83(s,1H),7.80(d,J=8.4Hz,1H),7.72(d,J=8.4Hz,1H),7.67–7.66(m,1H),7.42–7.39(m,1H),6.97(d,J=8.8Hz,1H),5.72–5.62(m,1H),5.31(d,J=5.6Hz,1H),5.01(dd,J=1.2,10.4Hz,1H),4.88(dd,J=10.4,1.2Hz,1H),4.72–4.64(m,2H),3.74(d,J=5.6Hz,1H),3.04(d,J=12.0Hz,2H),2.87–2.84(m,1H),2.57(t,J=10.8Hz,2H),2.28–2.18(m,11H),1.83(d,J=11.2Hz,2H),1.57–1.52(m,2H),1.15–1.09(m,1H),0.69–0.66(m,1H),0.45–0.42(m,1H),0.39–0.34(m,2H).LCMS(ESI)m/z:581.2[M+H]+,HPLC method B:RT=8.46min,purity>89.8%。
化合物48b(4.43mg,6.42μmol)。1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.83(s,1H),7.80(d,J=8.4Hz,1H),7.72(d,J=8.4Hz,1H),7.67–7.66(m,1H),7.42–7.39(m,1H),6.97(d,J=8.8Hz,1H),5.72–5.62(m,1H),5.31(d,J=5.6Hz,1H),5.01(dd,J=1.2,10.4Hz,1H),4.88(dd,J=10.4,1.2Hz,1H),4.72–4.60(m,2H),3.74(d,J=5.6Hz,1H),3.04(d,J=12.0Hz,2H),2.87–2.84(m,1H),2.57(t,J=10.8Hz,2H),2.28–2.18(m,11H),1.83(d,J=11.2Hz,2H),1.57–1.52(m,2H),1.15–1.09(m,1H),0.69–0.66(m,1H),0.45–0.42(m,1H),0.39–0.34(m,2H).LCMS(ESI)m/z:581.2[M+H]+,HPLC method B:RT=8.50min,purity>84.1%。
实施例49:化合物49的合成
步骤1:化合物49-2的合成:
在干燥的单口瓶中加入底物49-1(416mg,2.10mmol),溶于二甲基亚砜(5mL)中,然后加入2-氟5-硝基苯甲醚(342mg,2.00mmol)和碳酸钾(386.68mg,2.80mmol),110℃下反应4个小时。LC-MS监测反应完全。将反应液冷却至室温,加水,然后用乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物49-2(470 mg,1.35mmol)。LCMS(ESI)m/z:294.3[M+H-56]+
步骤2:化合物49-3的合成:
在干燥的单口瓶中加入底物49-2(200mg,572.44μmol),溶于二氯甲烷(3mL)中,加入三氟乙酸(3mL),室温下反应1个小时。LC-MS监测反应完全。将反应液减压浓缩得到粗品化合物49-3(140mg,561.65umol)。LCMS(ESI)m/z:250.1[M+H]+
步骤3:化合物49-4的合成:
在干燥的单口瓶中加入底物49-3(140mg,561.65μmol),溶于甲醇(5mL)中,加入醋酸(0.5mL)和甲醛水溶液(191.30mg,5.62mmol,37%),室温搅拌半个小时,然后加入氰基硼氢化钠(105.88mg,1.68mmol),50℃下反应6个小时。LC-MS监测反应完全。将反应液减压浓缩,加入饱和碳酸氢钠水溶液调节至pH=9,然后水相用乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物49-4(32mg,121.54μmol)。LCMS(ESI)m/z:264.2[M+H]+
步骤4:化合物49-5的合成:
在干燥的单口瓶中加入底物49-4(32mg,121.54μmol),溶于甲醇(3mL)中,然后加入钯炭(5mg),在氢气环境下室温反应2个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物49-5(25.0mg,107.15umol)。LCMS(ESI)m/z:234.1[M+H]+
步骤5:化合物49的合成:
在干燥的单口瓶中,加入底物4-9(20mg,50.57μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(15.54mg,90.03μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(138.49mg,1.07mmol)和底物49-5(25.0mg,107.15μmol),25℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物49(3.0mg,4.71μmol)。1H NMR(600MHz,Chloroform-d)δ8.83(s,1H),7.67–7.63(m,2H),7.17(s,1H),7.10–7.09(m,1H),7.00–6.98(m,1H),5.76–5.69(m,1H),5.06(d,J=10.2Hz,1H),4.98(dd,J=17.4,1.2Hz,1H),4.69–4.63(m,2H),4.28(s,1H),3.80(d,J=4.2Hz,2H),3.77(s,3H),3.75–3.72(m,2H),3.59(d,J=9.6Hz,2H),2.91–2.89(m,2H),2.76(s,1H),2.35(s,3H),2.04(s,1H),2.00–1.95(m,2H),1.63–1.59(m,1H),0.84–0.83(m,1H),0.68–0.66(m,1H),0.48–0.44(m,2H).LCMS(ESI)m/z:581.3[M+H]+,HPLC method B:RT=7.19min,purity>91.1%。
实施例50:化合物50的合成
步骤1:化合物50a&50b的合成
在干燥的单口瓶中,加入底物6-4(30mg,73.26μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(22.76mg,131.87μmol),室温下反应1个小时,再向反应液中加入 N,N-二异丙基乙胺(81.02mg,626.91μmol)和底物50-1(29mg,125.38μmol),25℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化得到产物,再经过超临界液相色谱手型拆分得到化合物50a(10.1mg,16.68μmol);SFC保留时间t=6.088min。1H NMR(600MHz,DMSO-d6)δ10.06(s,1H),8.79(s,1H),7.76(s,1H),7.70(d,J=7.8Hz,1H),7.58(s,2H),6.68(d,J=9.0Hz,2H),5.70–5.63(m,1H),4.99(d,J=10.2Hz,1H),4.87–4.80(m,2H),4.68(s,1H),4.65–4.63(m,2H),4.40(s,1H),3.70(d,J=9.6Hz,1H),3.60(d,J=9.6Hz,1H),3.49(d,J=10.8Hz,1H),3.31(s,1H),2.91-2.86(m,1H),2.81–2.78(m,1H),2.64–2.61(m,1H),2.01–1.96(m,1H),1.82(s,3H),1.58(d,J=8.4Hz,1H),1.45–1.40(m,4H),0.86–0.84(m,1H),0.64–0.61(m,1H),0.33–0.30(m,1H),0.22–0.19(m,1H).LCMS(ESI)m/z:593.3[M+H]+,HPLC method B:RT=7.16min,purity>97.9%。
化合物54b(9.5mg,15.44μmol);SFC保留时间t=9.232min。1H NMR(600MHz,DMSO-d6)δ10.06(s,1H),8.79(s,1H),7.76(s,1H),7.70(d,J=8.4Hz,1H),7.58(s,2H),6.68(d,J=8.4Hz,2H),5.70–5.63(m,1H),5.00–4.98(d,J=10.2Hz,1H),4.87–4.80(m,2H),4.68(s,1H),4.65–4.63(m,2H),4.40(s,1H),3.70(d,J=10.2Hz,1H),3.60(d,J=10.2Hz,1H),3.49(d,J=10.8Hz,1H),3.30(s,1H),2.91–2.86(m,1H),2.81–2.78(m,1H),2.64–2.61(m,1H),2.01–1.96(m,1H),1.82(s,3H),1.58(d,J=8.4Hz,1H),1.45–1.40(m,4H),0.86–0.84(m,1H),0.64–0.61(m,1H),0.33–0.30(m,1H),0.22–0.19(m,1H).LCMS(ESI)m/z:581.3[M+H]+,HPLC method B:RT=7.17min,purity>96.3%。
实施例51:化合物51的合成
步骤1:化合物51a&51b的合成
在干燥的单口瓶中,加入底物6-4(30mg,73.26μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(22.76mg,131.87μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(89.21mg,690.26μmol)和底物51-1(30mg,138.05μmol),25℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化得到产物,再经过超临界液相色谱手型拆分得到化合物51a(9.1mg,14.28μmol);SFC保留时间t=3.568min。1H NMR(400MHz,Chloroform-d)δ8.81(s,1H),7.66(d,J=12.0Hz,1H),7.57(d,J=12.0Hz,1H),7.46(s,2H),6.72(d,J=13.8Hz,2H),5.78–5.68(m,1H),5.07(dd,J=15.6,1.2Hz,1H),4.99(dd,J=25.2,1.8Hz,1H),4.78–4.61(m,2H),3.97(s,2H),3.59(d,J=16.8Hz,2H),3.43(d,J=16.8Hz,2H),3.02–2.93(m,1H),2.86–2.80(m,2H),2.54–2.52(m,2H),2.36–2.28(m,1H),1.54(s,3H),1.30–1.24(m,2H),1.10(t,J=10.8Hz,3H),1.03–0.98(m,1H),0.88–0.83(m,1H),0.44–0.39(m,1H),0.25–0.20(m,1H).LCMS(ESI)m/z:579.3[M+H]+,HPLC method B:RT=8.55min,purity>90.8%。
化合物51b(7.9mg,13.65μmol);SFC保留时间t=5.059min。1H NMR(400MHz,Chloroform-d)δ8.81(s,1H),7.65(d,J=12.0Hz,1H),7.57(d,J=12.0Hz,1H),7.49(s,2H),6.72(d,J=13.8Hz,2H),5.77–5.68(m,1H),5.07(dd,J=15.6,1.2Hz,1H),4.98(dd,J=25.2,1.8Hz,1H),4.78–4.61(m,2H),4.06(s,2H),3.62(d,J=16.8Hz,2H),3.48(d,J=16.8Hz,2H),3.34(s,1H),3.02–2.93(m,1H),2.86–2.80(m,2H),2.60–2.59(m,2H),2.36–2.28(m,1H),1.54(s,3H),1.30–1.24(m,2H),1.15(t,J=10.8Hz,3H),1.03–0.98(m,1H),0.88–0.83(m,1H),0.44–0.39(m,1H),0.25–0.20(m,1H).LCMS(ESI)m/z:579.3[M+H]+,HPLC method B:RT=8.40min,purity>95.4%。
实施例52:化合物52的合成
步骤1:化合物52a&52b的合成
在干燥的单口瓶中,加入底物6-4(30mg,73.26μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(31.61mg,183.15μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(94.68mg,732.59μmol)和底物41-1(34.19mg,146.52μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化得到产物,再经过超临界液相色谱手型拆分得到化合物52a(6.2mg,9.00μmol);SFC保留时间t=3.064min。1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.83(s,1H),7.75(d,J=8.4Hz,1H),7.72–7.65(m,2H),7.41–7.38(m,1H),6.97(d,J=8.8Hz,1H),5.72–5.62(m,1H),4.99(dd,J=10.4,1.2Hz,1H),4.88–4.83(m,2H),4.70(s,1H),4.66–4.61(m,1H),3.04(d,J=12.0Hz,1H),2.93–2.77(m,2H),2.59–2.54(m,2H),2.25–2.15(m,11H),2.02–1.95(m,1H),1.83(d,J=11.6Hz,2H),1.58–1.48(m,2H),1.46–1.40(m,4H),0.87–0.84(m,1H),0.65–0.60(m,1H),0.35–0.30(m,1H),0.23–0.19(m,1H).LCMS(ESI)m/z:595.4[M+H]+,HPLC method B:RT=10.14min,purity>86.3%。
化合物52b(6.56mg,9.89μmol);SFC保留时间t=5.193min。1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.83(s,1H),7.75(d,J=8.4Hz,1H),7.72–7.65(m,2H),7.41–7.38(m,1H),6.97(d,J=8.8Hz,1H),5.72–5.62(m,1H),4.99(dd,J=10.4,1.2Hz,1H),4.88–4.78(m,2H),4.70(s,1H),4.67–4.61(m,1H),3.04(d,J=12.0Hz,1H),2.93–2.77(m,2H),2.59–2.54(m,2H),2.25–2.15(m,11H),2.02–1.95(m,1H),1.83(d,J=11.6Hz,2H),1.58–1.48(m,2H),1.46–1.40(m,4H),0.87–0.84(m,1H),0.65–0.60(m,1H),0.35–0.30(m,1H),0.23–0.19(m,1H).LCMS(ESI)m/z:595.4[M+H]+,HPLC method B:RT=10.32min,purity>89.7%。
实施例53:化合物53的合成
步骤1:化合物53-1的合成:
在干燥的三口瓶中加入底物34-3(180mg,866.82μmol),加入四氢呋喃(5mL)溶解,氮气保护下冷却至零度,然后缓慢滴加(三氟甲基)三甲基硅烷(616.28mg,4.33mmol)和四丁基氟化铵(1M,866.82μL),保持温度反应1个小时。LC-MS监测。将反应液减压浓缩,残余物用柱层析纯化得化合物53-1(209mg,752.69μmol)。LCMS(ESI)m/z:278.1[M+H]+
步骤2:化合物53-2的合成:
在干燥的微波管中,加入底物53-1(209mg,752.69μmol)、底物IM-1(175.66mg,790.33μmol)、碘化亚铜(286.70mg,1.51mmol)、碘化钠(225.64mg,1.51mmol)、碳酸钾(260.07mg,1.88mmol)和反-(1R,2R)-N,N'-二甲基1,2-环己烷二胺(428.25mg,3.01mmol),然后加入苯甲醚(15mL),在氮气环境下,130度微波加热反应3个小时。LC-MS监测。将反应液冷却至室温,过滤,减压浓缩,残余物用柱层析纯化得到化合物53-2(210mg,453.10μmol)。LCMS(ESI)m/z:464.3[M+H]+
步骤3:化合物53a&53b的合成:
在干燥的单口瓶中,加入底物53-2(40mg,86.30μmol),溶于四氢呋喃(4mL)中,然后加入间氯过氧苯甲酸(31.54mg,155.35μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(111.54mg,863.05μmol)和底物4-(4-甲基哌嗪)苯胺(33.02mg,172.61μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物53a(12.6mg,18.88μmol);SFC保留时间t=2.141min。1H NMR(600MHz,Methanol-d4)δ10.17(s,1H),8.83(s,1H),8.06–7.92(m,1H),7.89(d,J=8.4Hz,1H),7.58(s,2H),6.94(d,J=8.4Hz,2H),6.03(s,1H),5.73–5.62(m,1H),4.97(d,J=10.2Hz,1H),4.88–4.73(m,2H),4.59(dd,J=16.2,7.2Hz,1H),3.11(t,J=4.8Hz,4H),3.05–2.99(m,1H),2.93(dd,J=17.2,6.0Hz,1H),2.47(t,J=4.8Hz,4H),2.23(s,3H),1.25–1.17(m,2H),1.05–1.09(m,1H),0.74–0.72(m,1H),0.60–0.52(m,1H),0.22–0.20(m,1H).LCMS(ESI)m/z:607.5[M+H]+,HPLC method B:RT=8.74min,purity>90.9%。
化合物53b(13.7mg,20.60μmol);SFC保留时间t=3.522min。1H NMR(600MHz,Methanol-d4)δ10.17(s,1H),8.83(s,1H),8.07–7.92(m,1H),7.89(d,J=8.4Hz,1H),7.58(s,2H),6.94(d,J=8.4Hz,2H),6.03(s,1H),5.73–5.62(m,1H),5.02–4.93(m,1H),4.86–4.73(m,2H),4.59(dd,J=16.2,7.2Hz,1H),3.11(t,J=4.8Hz,4H),3.05–2.99(m,1H),2.93(dd,J=17.2,6.0Hz,1H),2.46(t,J=4.8Hz,4H),2.23(s,3H),1.25–1.16(m,2H),1.05–1.09(m,1H), 0.74–0.72(m,1H),0.60–0.52(m,1H),0.22–0.20(m,1H).LCMS(ESI)m/z:607.1[M+H]+,HPLC method B:RT=8.77min,purity>91.2%。
实施例54:化合物54的合成
步骤1:化合物54a&54b的合成:
在干燥的单口瓶中,加入底物53-2(40mg,86.30μmol),溶于四氢呋喃(4mL)中,然后加入间氯过氧苯甲酸(31.54mg,155.35μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(111.54mg,863.05μmol)和底物12-1(37.86mg,172.61μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物54a(8.2mg,12.61μmol);SFC保留时间t=2.764min。1H NMR(600MHz,Methanol-d4)δ10.17(s,1H),8.83(s,1H),8.06–7.92(m,1H),7.89(d,J=8.4Hz,1H),7.58(s,2H),6.94(d,J=8.4Hz,2H),6.03(s,1H),5.73–5.62(m,1H),4.97(d,J=10.2Hz,1H),4.88–4.73(m,2H),4.59(dd,J=16.2,7.2Hz,1H),3.11(t,J=4.8Hz,4H),3.06–2.98(m,1H),2.93(dd,J=17.2,6.0Hz,1H),2.47(t,J=4.8Hz,4H),2.23(s,3H),1.25–1.17(m,2H),1.03–0.97(m,1H),0.70–0.75(m,1H),0.60–0.52(m,1H),0.23–0.18(m,1H).LCMS(ESI)m/z:635.4[M+H]+,HPLC method B:RT=9.66min,purity>97.6%。
化合物54b(10.0mg,14.98μmol);SFC保留时间t=4.157min。1H NMR(600MHz,Methanol-d4)δ10.17(s,1H),8.83(s,1H),8.07–7.92(m,1H),7.89(d,J=8.4Hz,1H),7.58(s,2H),6.94(d,J=8.4Hz,2H),6.03(s,1H),5.73–5.62(m,1H),5.02–4.93(m,1H),4.86–4.73(m,2H),4.59(dd,J=16.2,7.2Hz,1H),3.11(t,J=4.8Hz,4H),3.06–2.98(m,1H),2.93(dd,J=17.2,6.0Hz,1H),2.46(t,J=4.8Hz,4H),2.23(s,3H),1.25–1.16(m,2H),1.03–0.97(m,1H),0.70–0.75(m,1H),0.60–0.52(m,1H),0.23–0.18(m,1H).LCMS(ESI)m/z:635.4[M+H]+,HPLC method B:RT=9.75min,purity>95.1%。
实施例55:化合物55的合成
步骤1:化合物55的合成:
在干燥的单口瓶中,加入底物32-2(40mg,89.00μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(38.40mg,222.49μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(115.02mg,889.98μmol)和底物28-5(38.68mg,178.00μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物55(12.38mg,17.33μmol)。1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.82(s,1H),8.04–8.02(m,1H),7.91(d,J=8.0Hz,1H),7.53(s,2H),6.69(s,1H),6.54(d,J=8.4Hz,2H),5.72–5.62(m,1H),4.98(d,J=10.0Hz,1H),4.82(d,J=16.8Hz,2H),4.57(dd,J=7.2,16.0Hz,1H),3.30(s,2H),3.22–3.18(m,1H),3.05(d,J=8.8Hz,2H),2.87–2.86(m,2H),2.75–2.71(m,1H),2.56(t,J=8.8Hz,2H),2.40–2.22(m,2H),2.22(s,3H),1.04–0.99(m,2H),0.91–0.88(m,1H),0.49–0.47(m,1H).LCMS(ESI)m/z:619.3[M+H]+,HPLC method B:RT=8.79min,purity>86.6%。
实施例56:化合物56的合成
步骤1:化合物56a&56b的合成:
在干燥的单口瓶中,加入底物34-5(35mg,88.28μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(27.42mg,158.90μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(114.09mg,882.76μmol,)和底物30-3(36.25mg,176.55μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物56a(11.0mg,19.87μmol);SFC保留时间t=3.027min。1H NMR(400MHz,DMSO-d6)δ10.11(s,1H),8.81(s,1H),7.83(d,J=8.4Hz,1H),7.69(d,J=8.4Hz,1H),7.58(s,2H),6.92(d,J=9.2Hz,2H),5.74–5.62(m,1H),5.29(s,1H),5.02–4.97(m,1H),4.93–4.85(m,1H),4.68(s,1H),4.61(dd,J=15.9,5.7Hz,1H),3.52–3.43(m,2H),2.89–2.78(m,3H),2.72–2.65(m,1H),2.37–2.31(m,1H),2.30–2.22(m,2H),2.21(s,3H),2.17–2.10(m,J=9.7,6.3,2.9Hz,1H),1.14–1.08(m,1H),1.06(d,J=6.2Hz,3H),0.71–0.63(m,1H),0.46–0.41(m,1H),0.40–0.32(m,2H).LCMS(ESI)m/z:554.2[M+H]+,HPLC method B:RT=7.15min,purity>82.1%。
化合物56b(10.8mg,19.51μmol);SFC保留时间t=5.117min。1H NMR(400MHz,DMSO-d6)δ10.11(s,1H),8.81(s,1H),7.83(d,J=8.4Hz,1H),7.69(d,J=8.4Hz,1H),7.58(s,2H),7.01–6.79(m,2H),5.73–5.62(m,1H),5.29(s,1H),5.02–4.97(m,1H),4.94–4.85(m,1H),4.68(s,1H),4.61(dd,J=15.7,5.7Hz,1H),3.49(d,J=11.0Hz,2H),2.88–2.78(m,3H),2.72–2.64(m,1H),2.34(dd,J=11.6,9.9Hz,1H),2.30–2.23(m,2H),2.21(s,3H),2.17–2.10(m,J=9.7,6.3,2.9Hz,1H),1.15–1.08(m,1H),1.06(d,J=6.2Hz,3H),0.71–0.62(m,1H),0.47–0.41(m,1H),0.40–0.33(m,2H).LCMS(ESI)m/z:554.2[M+H]+,HPLC method B:RT=7.15min,purity>87.6%。
实施例57:化合物57的合成
步骤1:化合物57a&57b的合成:
在干燥的单口瓶中,加入底物34-5(35mg,88.28μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(15.23mg,88.28μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(114.09mg,882.76μmol,)和底物57-1(38.72mg,176.55μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物57a(12.5mg,22.02μmol);SFC保留时间t=3.149min。1H NMR(400MHz,DMSO-d6)δ10.11(s,1H),8.81(s,1H),7.83(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.64–7.49(m,2H),6.91(d,J=9.2Hz,2H),5.74–5.63(m,1H),5.29(s,1H),5.00(dd,J=10.2,1.6Hz,1H),4.89(dd,J=17.2,1.6Hz,1H),4.64(dd,J=17.2,1.2Hz,2H),3.09(t,J=4.8Hz,4H),2.90–2.81(m,2H),2.71–2.64(m,1H),2.58(t,J=5.2Hz,4H),2.29–2.21(m,1H),1.14–1.08(m,1H),1.02(s,3H),1.00(s,3H),0.70–0.64(m,1H),0.47–0.41(m,1H),0.39–0.30(m,2H).LCMS(ESI)m/z:568.2[M+H]+,HPLC method B:RT=7.64min,purity>86.5%。
化合物57b(12.4mg,21.84μmol);SFC保留时间t=4.956min。1H NMR(400MHz,DMSO-d6)δ10.11(s,1H),8.81(s,1H),7.83(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.58(s,2H),6.91(d,J=8.8Hz,2H),5.74–5.64(m,1H),5.29(s,1H),5.00(dd,J=10.2,1.6Hz,1H),4.89(dd,J=17.2,1.6Hz,1H),4.64(dd,J=17.2,1.6Hz,2H),3.09(t,J=4.8Hz,4H),2.89–2.79(m,2H),2.71–2.64(m,1H),2.58(t,J=5.0Hz,4H),2.29–2.21(m,1H),1.14–1.07(m,1H),1.02(s,3H),1.00(s,3H),0.70–0.64(m,1H),0.47–0.41(m,1H),0.39–0.31(m,2H).LCMS(ESI)m/z:568.2[M+H]+,HPLC method B:RT=7.63min,purity>88.6%。
实施例58:化合物58的合成
步骤1:化合物58a&58b的合成:
在干燥的单口瓶中,加入底物53-2(40mg,86.30μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(37.23mg,215.76μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(111.54mg,863.05μmol)和底物28-5(18.75mg,86.30μmol),45℃反应 过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物58a(7.25mg,9.60μmol);SFC保留时间t=3.125min。1H NMR(400MHz,DMSO-d6)δ10.09(s,1H),8.81(s,1H),7.94–7.88(m,2H),7.52(s,2H),6.64(d,J=8.4Hz,1H),6.02(s,1H),5.69–5.59(m,1H),4.96(d,J=9.6Hz,1H),4.80–4.76(m,2H),4.61–4.56(m,1H),3.30(s,2H),3.06(d,J=8.8Hz,2H),2.99–2.88(m,3H),2.57(t,J=8.4Hz,2H),2.46(s,1H),2.39(d,J=7.6Hz,2H),2.22(s,3H),1.23(s,1H),1.02–0.97(m,1H),0.74–0.71(m,1H),0.59–0.54(m,1H),0.23–0.19(m,1H).LCMS(ESI)m/z:633.3[M+H]+,HPLC method B:RT=10.11min,purity>83.8%。
化合物58b(6.7mg,8.65μmol);SFC保留时间t=4.825min。1H NMR(400MHz,DMSO-d6)δ10.09(s,1H),8.81(s,1H),7.94–7.88(m,2H),7.53–7.37(m,2H),6.64(d,J=8.4Hz,1H),6.02(s,1H),5.69–5.59(m,1H),4.96(d,J=9.6Hz,1H),4.80–4.76(m,2H),4.61–4.56(m,1H),3.30(s,2H),3.06(d,J=8.8Hz,2H),2.99–2.88(m,3H),2.59(t,J=8.4Hz,2H),2.47–2.41(m,3H),2.24(s,3H),1.26–1.23(m,2H),1.03–0.97(m,1H),0.74–0.71(m,1H),0.59–0.54(m,1H),0.23–0.19(m,1H).LCMS(ESI)m/z:633.2[M+H]+,HPLC method B:RT=10.14min,purity>81.7%。
实施例59:化合物59的合成
步骤1:化合物59-2的合成:
在干燥的单口瓶中加入底物59-1(66mg,301.04μmol),溶于丙酮(2mL)中,1-氟-2-碘乙烷(57mg,327.67μmol)和碳酸钾(58mg,419.68μmol),50℃加热反应6个小时。LC-MS监测反应完全。将反应液减压浓缩,加水,用乙酸乙酯取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物59-2(55mg,207.33μmol)。LCMS(ESI)m/z:266.2[M+H]+
步骤2:化合物59-3的合成:
在干燥的单口瓶中加入底物59-2(55mg,207.33μmol),溶于甲醇(5mL)中,然后加入钯炭(10mg),在氢气环境下室温反应2个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物59-3(30mg,127.50μmol)。LCMS(ESI)m/z:236.2[M+H]+
步骤3:化合物59a&59b的合成:
在干燥的单口瓶中,加入底物6-4(30mg,73.26μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(22.76mg,131.87μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(54.67mg,423.03μmol)和底物59-3(30mg,127.50μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物59a(10.8mg,16.54μmol);SFC保留时间t=3.435min。1H NMR(400MHz,Chloroform-d)δ8.81(s,1H),7.68–7.57(m,2H), 7.49–7.47(m,2H),6.73(d,J=8.8Hz,2H),5.77–5.68(m,1H),5.08–5.06(m,1H),5.01–4.96(m,1H),4.79–4.57(m,4H),4.03–4.02(m,2H),3.68(d,J=11.2Hz,2H),3.45(d,J=11.2Hz,2H),3.34(s,1H),3.02–2.94(m,1H),2.89–2.76(m,4H),2.36–2.28(m,1H),1.73(d,J=8.8Hz,1H),1.55(s,3H),1.29–1.24(m,2H),1.03–0.99(m,1H),0.88–0.84(m,1H),0.44–0.39(m,1H),0.25–0.20(m,1H).LCMS(ESI)m/z:597.2[M+H]+,HPLC method B:RT=7.95min,purity>91.4%。
化合物59b(13.6mg,22.22μmol);SFC保留时间t=4.794min。1H NMR(400MHz,Chloroform-d)δ8.81(s,1H),7.67(d,J=8.4Hz,1H),7.58(d,J=8.0Hz,1H),7.48–7.47(m,2H),6.73(d,J=8.8Hz,2H),5.77–5.68(m,1H),5.08–5.05(m,1H),5.01–4.96(m,1H),4.79–4.54(m,4H),3.98(d,J=4.0Hz,2H),3.67(d,J=11.2Hz,2H),3.43(d,J=11.2Hz,2H),3.34(s,1H),3.02–2.95(m,1H),2.85–2.73(m,4H),2.36–2.28(m,1H),1.70(d,J=8.8Hz,1H),1.54(s,3H),1.29–1.24(m,2H),1.03–0.99(m,1H),0.88–0.83(m,1H),0.44–0.39(m,1H),0.25–0.22(m,1H).LCMS(ESI)m/z:597.2[M+H]+,HPLC method B:RT=7.96min,purity>97.5%。
实施例60:化合物60的合成
步骤1:化合物60a&60b的合成:
在干燥的单口瓶中,加入底物6-4(30mg,73.26μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(22.76mg,131.87μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(54.67mg,423.03μmol)和底物60-1(35mg,147.48μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合60a(8.16mg,11.67μmol)。1H NMR(400MHz,Chloroform-d)δ8.81(s,1H),7.66-7.58(m,2H),7.46(d,J=8.8Hz,2H),6.93(d,J=8.8Hz,2H),5.77–5.67(m,1H),5.15–4.96(m,3H),4.80–4.63(m,2H),4.00–3.94(m,1H),3.76(d,J=12.0Hz,1H),3.35(s,1H),3.03–2.76(m,4H),2.48(s,6H),2.40–2.28(m,2H),2.22–2.12(m,1H),1.95–1.92(m,2H),1.54(s,3H),1.30–1.25(m,1H),1.02–1.00(m,1H),0.88–0.83(m,1H),0.44–0.39(m,1H),0.26–0.21(m,1H).LCMS(ESI)m/z:599.2[M+H]+,HPLC method B:RT=7.91min,purity>85.6%。
化合物60b(2mg,2.84μmol)。1H NMR(400MHz,Chloroform-d)δ8.82(s,1H),7.65–7.58(m,2H),7.48(d,J=8.8Hz,2H),6.92(d,J=8.8Hz,2H),5.77–5.67(m,1H),5.08–5.06(m,1H),5.00–4.96(m,1H),4.90–4.61(m,3H),3.96–3.90(m,1H),3.63–3.60(m,1H),3.30(s,1H),3.03–2.95(m,1H),2.88–2.70(m,4H),2.52(s,6H),2.32–2.29(m,1H),2.06(s,1H),1.80–1.70(m,1H),1.55(s,3H),1.31–1.25(m,2H),1.03–0.98(m,1H),0.88–0.83(m,1H),0.44–0.40(m,1H),0.26–0.21(m,1H).LCMS(ESI)m/z:599.2[M+H]+,HPLC method B:RT=8.26min,purity>85.1%。
实施例61:化合物61的合成
步骤1:化合物61-2的合成:
在干燥的单口瓶中加入底物61-1(0.5g,1.97mmol),溶于二甲基亚砜(10mL)中,然后加入对2-氟-5-硝基甲苯(304.93mg,1.97mmol)和碳酸钾(543.33mg,3.93mmol),80℃下反应16个小时。LC-MS监测反应完全。将反应液冷却至室温,加水,然后用乙酸乙酯萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物61-2(740mg,1.90mmol)。LCMS(ESI)m/z:390.2[M+H]+
步骤2:化合物61-3的合成:
在干燥的单口瓶中加入底物61-2(740mg,1.90mmol),溶于二氯甲烷(15mL)中,加入三氟乙酸(5mL),室温下反应2个小时。LC-MS监测反应完全。将反应液减压浓缩,残余物溶于水,并加入饱和碳酸氢钠水溶液调节至pH=9,然后水相用二氯甲烷萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物61-3(550mg,1.90mmol)。LCMS(ESI)m/z:290.2[M+H]+
步骤3:化合物61-4的合成:
在干燥的单口瓶中加入底物61-3(550mg,1.90mmol),溶于甲醇(15mL)中,加入醋酸(3mL)和甲醛水溶液(1.91g,20.73mmol,37%),室温搅拌1个小时,然后加入氰基硼氢化钠(260.60mg,4.15mmol)室温下反应18个小时。LC-MS监测反应完全。将反应液减压浓缩,加入饱和碳酸氢钠水溶液调节至pH=9,然后水相用二氯甲烷萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物61-4(400mg,1.32mmol)。LCMS(ESI)m/z:304.2[M+H]+
步骤4:化合物61-5的合成:
在干燥的单口瓶中加入底物61-4(400mg,1.32mmol),溶于甲醇(10mL)中,然后加入钯炭(50mg),在氢气环境下室温反应16个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物61-5(340mg,1.24mmol)。LCMS(ESI)m/z:274.4[M+H]+
步骤5:化合物61a&61b的合成:
在干燥的单口瓶中,加入底物34-5(33.35mg,84.12μmol),溶于四氢呋喃(3mL)中,然后加入间氯过氧苯甲酸(30.74mg,151.42μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(108.72mg,841.21μmol)和底物61-5(46mg,168.24μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物61a(8.1mg,11.75μmol);SFC保留时间t=3.216min。1H NMR(600MHz,DMSO-d6)δ10.13(s,1H),8.83(s,1H),7.81(d,J=8.4Hz,1H),7.72(d,J=8.4Hz,1H),7.65(s,1H),7.43–7.42(m,1H),7.01(d,J=8.4Hz,1H),5.70–5.64(m,1H),5.29(s,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.77–4.57(m,2H),2.90–2.82(m,2H),2.75(t,J=5.4Hz,4H),2.32(s,4H),2.26–2.22(m,4H),2.18(s,3H),1.55(t,J=5.4Hz,4H),1.51(t,J=5.4Hz,4H),1.16–1.09(m,1H),0.69–0.66(m,1H),0.44–0.42(m,1H),0.39–0.34(m,2H).LCMS(ESI)m/z:622.4[M+H]+,HPLC method B:RT=9.98min,purity>90.2%。
化合物61b(7.9mg,11.60μmol);SFC保留时间t=5.557min。1H NMR(600MHz,DMSO-d6)δ10.14(s,1H),8.83(s,1H),7.81(d,J=8.4Hz,1H),7.72(d,J=8.4Hz,1H),7.65(s,1H),7.43–7.42(m,1H),7.01(d,J=8.4Hz,1H),5.70–5.64(m,1H),5.30(s,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.73–4.58(m,2H),2.90–2.81(m,2H),2.75(t,J=5.4Hz,4H),2.34(s,4H),2.27–2.22(m,4H),2.19(s,3H),1.55(t,J=5.4Hz,4H),1.51(t,J=5.4Hz,4H),1.16–1.09(m,1H),0.69–0.66(m,1H),0.44–0.42(m,1H),0.39–0.34(m,2H).LCMS(ESI)m/z:622.4[M+H]+,HPLC method B:RT=10.05min,purity>91.3%。
实施例62:化合物62a和62b的合成
步骤1:化合物62-1的合成:
在干燥的单口瓶中加入底物61-1(0.5g,1.97mmol),溶于二甲基亚砜(10mL)中,然后加入3,4-二氟硝基苯(312.72mg,1.97mmol)和碳酸钾(326.00mg,2.36mmol),80℃下反应16个小时。LC-MS监测反应完全。将反应液冷却至室温,加水,然后用乙酸乙酯萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物62-1(730mg, 1.86mmol)。LCMS(ESI)m/z:394.2[M+H]+
步骤2:化合物62-2的合成:
在干燥的单口瓶中加入底物62-1(730mg,1.86mmol),溶于二氯甲烷(10mL)中,加入三氟乙酸(5mL),室温下反应2个小时。LC-MS监测反应完全。将反应液减压浓缩,残余物溶于水,并加入饱和碳酸氢钠水溶液调节至pH=9,然后水相用二氯甲烷萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物62-2(540mg,1.84mmol)。LCMS(ESI)m/z:294.1[M+H]+
步骤3:化合物62-3的合成:
在干燥的单口瓶中加入底物62-2(540mg,1.84mmol),溶于甲醇(15mL)中,加入醋酸(3mL)和甲醛水溶液(1.73g,18.75mmol,37%),室温搅拌1个小时,然后加入氰基硼氢化钠(235.66mg,3.75mmol)室温下反应18个小时。LC-MS监测反应完全。将反应液减压浓缩,加入饱和碳酸氢钠水溶液调节至pH=9,然后水相用二氯甲烷萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物62-3(500mg,1.63mmol)。LCMS(ESI)m/z:308.1[M+H]+
步骤4:化合物62-4的合成:
在干燥的单口瓶中加入底物62-3(500mg,1.63mmol),溶于甲醇(10mL)中,然后加入钯炭(60mg),在氢气环境下室温反应16个小时。LC-MS监测反应完全。将反应液用硅藻土过滤,用甲醇洗涤两次,滤液减压浓缩得到化合物62-4(450mg,1.62mmol)。LCMS(ESI)m/z:278.4[M+H]+
步骤5:化合物62a&62b的合成:
在干燥的单口瓶中,加入底物34-5(33.34mg,84.09μmol),溶于四氢呋喃(3mL)中,然后加入间氯过氧苯甲酸(30.73mg,151.36μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(108.68mg,840.91μmol)和底物62-4(46.65mg,168.18μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物62a(6.0mg,8.32μmol);SFC保留时间t=3.230min。1H NMR(600MHz,DMSO-d6)δ10.31(s,1H),8.87(s,1H),7.81(d,J=8.4Hz,1H),7.75–7.66(m,2H),7.39(d,J=8.4Hz,1H),7..04–7.01(m,1H),5.72–5.63(m,1H),5.30(s,1H),5.00(dd,J=10.2,1.8Hz,1H),4.89(dd,J=17.4,1.8Hz,1H),4.74–4.57(m,2H),2.92(t,J=5.4Hz,4H),2.88–2.80(m,2H),2.35–2.23(m,5H),2.18(s,3H),1.56(t,J=5.4Hz,4H),1.49(t,J=5.4Hz,4H),1.13–1.10(m,1H),0.68–0.66(m,1H),0.45–0.43(m,1H),0.39–0.33(m,2H).LCMS(ESI)m/z:626.2[M+H]+,HPLC method B:RT=9.55min,purity>86.8%。
化合物62b(6.2mg,8.76μmol);SFC保留时间t=5.103min。1H NMR(600MHz,DMSO-d6)δ10.31(s,1H),8.87(s,1H),7.81(d,J=8.4Hz,1H),7.75–7.66(m,2H),7.39(d,J=8.4Hz,1H),7.04–7.01(m,1H),5.72–5.63(m,1H),5.30(s,1H),5.00(dd,J=10.2,1.8Hz,1H),4.89(dd,J=17.4,1.8Hz,1H),4.74–4.58(m,2H),2.92(t,J=5.4Hz,4H),2.88–2.81(m,2H),2.33–2.23(m,5H),2.16(s,3H),1.56(t,J=5.4Hz,4H),1.49(t,J=5.4Hz,4H),1.13–1.10(m,1H),0.68–0.66(m,1H),0.45–0.43(m,1H),0.40–0.33(m,2H).LCMS(ESI)m/z:626.2[M+H]+,HPLC method B:RT=9.50min,purity>88.4%。
实施例63:化合物63a和63b的合成
步骤1:化合物63a&63b的合成:
在干燥的单口瓶中,加入底物34-5(33.33mg,83.99μmol),溶于四氢呋喃(3mL)中,然后加入间氯过氧苯甲酸(30.69mg,151.18μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(108.55mg,839.88μmol)和底物28-5(36.50mg,167.98μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物63a(6.9mg,10.11μmol);SFC保留时间t=3.357min。1H NMR(600MHz,DMSO-d6)δ10.06(s,1H),8.79(s,1H),7.81(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.55(s,2H),6.64(d,J=8.4Hz,2H),5.72–5.63(m,1H),5.28(s,1H),5.04–4.96(m,1H),4.89(d,J=17.4Hz,1H),4.82–4.54(m,2H),3.11–3.03(m,2H),2.92–2.79(m,4H),2.66–2.57(m,2H),2.53–2.50(m,2H),2.46–2.39(m,2H),2.29–2.21(m,4H),1.13–1.10(m,1H),0.69–0.66(m,1H),0.45–0.42(m,1H),0.39–0.34(m,2H).LCMS(ESI)m/z:566.4[M+H]+,HPLC method B:RT=7.83min,purity>82.9%。
化合物63b(6.8mg,10.53μmol);SFC保留时间t=5.596min。1H NMR(600MHz,DMSO-d6)δ10.06(s,1H),8.79(s,1H),7.81(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.55(s,2H),6.64(d,J=8.4Hz,2H),5.72–5.63(m,1H),5.29(s,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.76–4.58(m,2H),3.07(d,J=8.4Hz,2H),2.93–2.81(m,4H),2.62(d,J=7.2Hz,2H),2.52–2.59(m,2H),2.47–2.38(m,2H),2.29–2.21(m,4H),1.13–1.10(m,1H),0.69–0.66(m,1H),0.45–0.42(m,1H),0.39–0.34(m,2H).LCMS(ESI)m/z:566.4[M+H]+,HPLC method B:RT=7.78min,purity>87.6%。
实施例64:化合物64a&64b的合成
步骤1:化合物64a&64b的合成
在干燥的单口瓶中,加入底物21-3(40mg,94.44μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(29.34mg,170.00μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(122.06mg,944.44μmol)和底物7-5(38.40mg,188.89μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化得到产物,再经过超临界液相色谱手型拆分得到化合物64a(10.1mg,17.45μmol); SFC保留时间t=2.944min。1H NMR(600MHz,DMSO-d6)δ10.06(s,1H),8.79(s,1H),7.77(d,J=8.4Hz,1H),7.69(d,J=8.4Hz,1H),7.60(s,2H),6.71(d,J=8.4Hz,2H),5.72–5.63(m,1H),5.00(d,J=10.2Hz,1H),4.85(d,J=17.4Hz,1H),4.75(s,1H),4.61(d,J=16.2Hz,1H),4.40(s,1H),3.84(s,2H),3.64–3.55(m,2H),3.44(d,J=10.8Hz,2H),3.27(d,J=10.8Hz,2H),2.95–2.89(m,1H),2.86–2.80(m,1H),2.42(d,J=17.4Hz,1H),2.35–2.29(m,1H),2.00(s,3H),1.92(dd,J=14.4,7.2Hz,1H),1.75(dd,J=14.4,7.2Hz,1H),1.54(d,J=8.4Hz,1H),1.15–1.11(m,1H),0.93(t,J=7.2Hz,3H),0.87–0.85(m,1H),0.64–0.57(m,1H),0.42–0.36(m,1H),0.00(d,J=3.5Hz,1H).LCMS(ESI)m/z:579.4[M+H]+,HPLC method B:RT=9.34min,purity>86.7%。
化合物64b(8.7mg,15.03μmol);SFC保留时间t=4.409min。1H NMR(600MHz,DMSO-d6)δ10.06(s,1H),8.79(s,1H),7.77(s,1H),7.69(d,J=8.4Hz,1H),7.60(s,2H),6.71(d,J=8.4Hz,2H),5.72–5.63(m,1H),5.04–4.97(m,1H),4.85(d,J=17.4Hz,1H),4.74(s,1H),4.66–4.54(m,1H),4.40(s,1H),3.58(d,J=6.0Hz,2H),3.43(d,J=10.8Hz,2H),3.27(d,J=10.8Hz,2H),2.95–2.89(m,1H),2.87–2.81(m,1H),2.46–2.40(m,1H),2.35–2.29(m,1H),1.99(s,3H),1.96–1.90(m,1H),1.75(dd,J=14.4,7.2Hz,1H),1.53(d,J=8.4Hz,1H),1.15–1.11(m,1H),0.93(t,J=7.2Hz,3H),0.87–0.84(m,1H),0.64–0.57(m,1H),0.42–0.36(m,1H),0.00(s,1H).LCMS(ESI)m/z:579.4[M+H]+,HPLC method B:RT=9.25min,purity>91.1%。
实施例65:化合物65的合成
步骤1:化合物65a&65b的合成:
在干燥的单口瓶中,加入底物6-4(40mg,97.68μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(30.34mg,175.82μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(126.24mg,976.79μmol)和底物12-1(42.85mg,195.36μmol),50℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合65a(14.3mg,24.62μmol);SFC保留时间t=3.425min。1H NMR(600MHz,DMSO-d6)δ10.11(s,1H),8.81(s,1H),7.78(s,1H),7.69(d,J=8.4Hz,1H),7.56(s,2H),6.93(d,J=8.4Hz,2H),5.70–5.62(m,1H),4.99(dd,J=10.2,1.8Hz,1H),4.89–4.84(m,1H),4.83–4.73(m,1H),4.69(s,1H),4.64(dd,J=16.2,6.0Hz,1H),3.66(d,J=12.0Hz,2H),2.93–2.86(m,1H),2.83–2.77(m,1H),2.67–2.59(m,2H),2.19(s,6H),2.18(d,J=3.6Hz,1H),2.03–1.96(m,1H),1.83(d,J=12.0Hz,2H),1.52–1.45(m,2H),1.45–1.42(m,1H),1.40(s,3H),0.91–0.84(m,1H),0.68–0.60(m,1H),0.35–0.30(m,1H),0.23–0.18(m,1H).LCMS(ESI)m/z:581.4[M+H]+,HPLC method B:RT=8.79min,purity>93.5%。
化合物65b(15.6mg,26.86μmol);SFC保留时间t=5.024min。1H NMR(600MHz,DMSO-d6)δ10.11(s,1H),8.81(s,1H),7.78(d,J=7.8Hz,1H),7.69(d,J=8.4Hz,1H),7.64–7.41(m,2H),6.93(d,J=8.4Hz,2H),5.70–5.62(m,1H),4.99(d,J=10.2Hz,1H),4.86(d,J= 17.4Hz,1H),4.81(d,J=15.6Hz,1H),4.69(s,1H),4.64(dd,J=16.2,6.0Hz,1H),3.66(d,J=12.0Hz,2H),2.93–2.86(m,1H),2.83–2.77(m,1H),2.67–2.59(m,2H),2.20(s,6H),2.19–2.13(m,1H),1.96(m,1H),1.86–1.81(m,2H),1.48(dd,J=11.8,3.7Hz,2H),1.45–1.42(m,1H),1.41(s,3H),0.91–0.84(m,1H),0.67–0.60(m,1H),0.35–0.30(m,1H),0.23–0.18(m,1H).LCMS(ESI)m/z:581.4[M+H]+,HPLC method B:RT=8.86min,purity>93.6%。
实施例66:化合物66的合成
步骤1:化合物66a&66b的合成:
在干燥的单口瓶中,加入底物34-5(40mg,100.89μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(30.72mg,151.33μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(65.19mg,504.43μmol)和底物12-1(33.19mg,151.33μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物66a(12.12mg,21.35μmol);SFC保留时间t=3.579min。1H NMR(600MHz,DMSO-d6)δ10.12(s,1H),8.81(d,J=3.6Hz,1H),7.83(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.58(s,2H),6.98–6.90(m,2H),5.73–5.63(m,1H),5.30(s,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.69(s,1H),4.65–4.56(m,1H),3.66(d,J=12.0Hz,2H),3.04(s,1H),2.87–2.78(m,2H),2.65–2.59(m,2H),2.29–2.20(m,1H),2.19(s,6H),1.88–1.80(m,2H),1.53–1.44(m,2H),1.13–1.09(m,1H),0.71–0.62(m,1H),0.45–0.42(m,1H),0.39–0.32(m,2H).LCMS(ESI)m/z:568.2[M+H]+,HPLC method B:RT=7.48min,purity>86.1%。
化合物66b(12.56mg,22.12μmol);SFC保留时间t=5.858min。1H NMR(600MHz,DMSO-d6)δ10.12(s,1H),8.81(s,1H),7.83(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.58(s,2H),6.92(d,J=8.4Hz,2H),5.73–5.63(m,1H),5.29(s,1H),5.00(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.69(s,1H),4.65–4.54(m,1H),3.66(d,J=12.0Hz,2H),3.07(s,1H),2.87–2.79(m,2H),2.65–2.59(m,2H),2.29–2.20(m,1H),2.19(s,6H),1.82(s,2H),1.53–1.44(m,2H),1.13–1.10(m,1H),0.71–0.62(m,1H),0.45–0.42(m,1H),0.39–0.32(m,2H).LCMS(ESI)m/z:568.2[M+H]+,HPLC method B:RT=7.50min,purity>89.5%。
实施例67:化合物67的合成
步骤1:化合物67的合成:
在干燥的单口瓶中,加入底物4-9(20mg,50.57μmol),溶于四氢呋喃(2mL)中,然后加入间氯过氧苯甲酸(15.4mg,75.86μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(65.15mg,0.51mmol)和底物67-1(16.48mg,75.86μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到化合物67(6.8mg,12.02μmol)。1H NMR(600MHz,DMSO-d6)δ10.18(s,1H),8.85(s,1H),7.80(d,J=8.4Hz,1H),7.77–7.62(m,2H),7.46(d,J=8.4Hz,1H),7.34(d,J=9.0Hz,1H),5.71-5.65(m,1H),5.33(d,J=5.4Hz,1H),5.01(d,J=10.2Hz,1H),4.89(d,J=17.4Hz,1H),4.71–4.62(m,2H),3.75(d,J=4.8Hz,1H),3.65–3.65(m,2H),3.23–3.02(m,4H),2.86–2.80(m,2H),2.52(s,3H),2.33–2.21(m,6H),1.14–1.10(m,1H),0.70–0.66(m,1H),0.45–0.42(m,1H),0.39–0.34(m,2H).LCMS(ESI)m/z:565.2[M+H]+,HPLC method B:RT=8.22min,purity>87.8%。
实施例68:化合物68的合成
步骤1:化合物68a&68b的合成:
在干燥的单口瓶中,加入底物4-9(40mg,101.14μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(30.80mg,151.72μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(65.36mg,505.72μmol)和底物40-3(31.15mg,151.72μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物68a(10.48mg,518.96μmol);SFC保留时间t=3.121min。1H NMR(600MHz,DMSO-d6)δ10.12(s,1H),8.81(s,1H),7.83(d,J=8.4Hz,1H),7.69(d,J=8.4Hz,1H),7.64–7.46(m,2H),6.92(d,J=8.4Hz,2H),5.69–5.64(m,6.0Hz,1H),5.32(d,J=5.4Hz,1H),5.03–4.98(m,1H),4.91–4.85(m,1H),4.69(s,1H),4.63–4.60(m,5.4Hz,1H),3.75(d,J=5.4Hz,1H),3.47(d,J=13.2Hz,2H),2.88–2.80(m,3H),2.71–2.70(m,1H),2.35–2.33(m,1H),2.27–2.23(m,2H),2.22(s,3H),2.17–2.11(m,1H),1.13–1.11(m,1H),1.06(s,3H),0.70–0.66(m,1H),0.46–0.42(m,1H),0.39–0.35(m, 2H).LCMS(ESI)m/z:553.3[M+H]+,HPLC method B:RT=7.23min,purity>96.1%。
化合物68b(12.34mg,22.33μmol);SFC保留时间t=5.185min。1H NMR(600MHz,DMSO-d6)δ10.12(s,1H),8.81(s,1H),7.84(d,J=8.4Hz,1H),7.75–7.67(m,1H),7.59(s,2H),6.92(d,J=8.4Hz,2H),5.69–5.66(m,1H),5.31(d,J=5.4Hz,1H),5.05–4.96(m,1H),4.94–4.87(m,1H),4.69(s,1H),4.62–4.60(m,1H),3.75(d,J=5.4Hz,1H),3.48–3.45(m,2H),2.86–2.81(m,3H),2.74–2.69(m,1H),2.36–2.34(m,1H),2.29–2.23(m,2H),2.22(s,3H),2.15(s,1H),1.14–1.10(m,1H),1.06(s,3H),0.69–0.67(m,1H),0.44–0.42(m,1H),0.39–0.36(m,2H).LCMS(ESI)m/z:553.3[M+H]+,HPLC method B:RT=7.24min,purity>94.1%。
实施例69:化合物69的合成
步骤1:化合物69a&69b的合成:
在干燥的单口瓶中,加入底物21-3(40mg,94.44μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(28.76mg,141.67μmol),室温下反应1个小时,再向反应液中加入N,N-二异丙基乙胺(61.03mg,472.22μmol)和底物11-1(36.75mg,141.67μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱纯化得到产物,再经过超临界液相色谱手型拆分得到化合物69a(1.1mg,1.73μmol);SFC保留时间t=3.897min。1H NMR(600MHz,DMSO-d6)δ10.11(s,1H),8.81(d,J=4.2Hz,1H),7.79(s,1H),7.67(d,J=8.4Hz,1H),7.57(s,2H),6.92(d,J=9.0Hz,2H),5.74–5.62(m,1H),4.99(dd,J=10.2,1.8Hz,1H),4.85(d,J=17.4Hz,1H),4.74(s,1H),4.62(s,1H),4.40(d,J=4.2Hz,1H),3.09(s,4H),2.98–2.89(m,1H),2.84(dd,J=17.4,6.0Hz,1H),2.36–2.29(m,4H),2.18(s,3H),1.94–1.89(m,1H),1.75(dd,J=14.4,7.2Hz,1H),1.54(t,J=5.4Hz,4H),1.47(t,J=5.4Hz,4H),0.93(t,J=7.2Hz,3H),0.87–0.84(m,2H),0.78–0.73(m,2H),0.64–0.58(m,1H),0.43–0.37(m,1H).LCMS(ESI)m/z:635.4[M+H]+,HPLC method B:RT=7.07min,purity>83.2%。
化合物69b(1.0mg,1.58μmol);SFC保留时间t=5.463min。1H NMR(600MHz,DMSO-d6)δ10.11(s,1H),8.81(d,J=4.3Hz,1H),7.79(s,1H),7.67(d,J=8.2Hz,1H),7.57(s,2H),6.92(d,J=9.0Hz,2H),5.74–5.62(m,1H),4.99(dd,J=10.2,1.8Hz,1H),4.85(d,J=17.4Hz,1H),4.74(s,1H),4.61(s,1H),4.40(d,J=4.8Hz,1H),3.13–3.06(m,4H),2.98–2.89(m,1H),2.84(dd,J=16.8,6.0Hz,1H),2.38–2.29(m,4H),2.20(s,3H),1.93–1.89(m,1H),1.75(dd,J=14.4,7.2Hz,1H),1.54(t,J=5.4Hz,4H),1.48(t,J=5.4Hz,4H),0.93(d,J=7.2Hz,3H),0.87–0.84(m,2H),0.78–0.73(m,2H),0.64–0.58(m,1H),0.43–0.37(m,1H).LCMS(ESI)m/z:635.4[M+H]+,HPLC method B:RT=7.08min,purity>80.1%。
实施例70:化合物70的合成
步骤1:化合物70的合成:
在干燥的单口瓶中,加入底物34-5(68.41mg,172.55μmol),溶于四氢呋喃(3mL)中,然后加入间氯过氧苯甲酸(59.55mg,345.10μmol),室温下反应一个小时,再向反应液中加入N,N-二异丙基乙胺(111.50mg,862.75μmol)和底物70-1(65.67mg,345.10μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱(碱性)纯化得到化合物70(43.75mg,90.97μmol)。1H NMR(400MHz,DMSO-d6)δ10.23(s,1H),8.87(s,1H),7.82(d,J=8.4Hz,1H),7.75–7.63(m,3H),7.27–7.12(m,2H),5.74–5.61(m,1H),5.30(s,1H),5.02–4.99(m,1H),4.91–4.87(m,1H),4.64–4.51(m,2H),3.22(s,3H),2.93–2.78(m,2H),2.68–2.51(m,5H),2.31–2.20(m,1H),1.88(d,J=13.2Hz,2H),1.71–1.69(m,2H),1.13–1.10(m,1H),0.70–0.67(m,1H),0.47–0.40(m,1H),0.39–0.31(m,2H).LCMS(ESI)m/z:539.2[M+H]+,HPLC method B:RT=6.88min,purity>97.1%。
实施例71:化合物71的合成
步骤1:化合物71-2的合成:
向单口瓶中,加入底物71-1(2.5g,9.54mmol)、碳酸钾(5.27g,38.16mmol)和苄基三乙基氯化铵(217.34mg,0.95mmol),然后加入乙腈(30mL)溶解,待溶液变成黄色后,加入3-溴丙烷(1.58g,14.31mmol),80℃下搅拌48小时。LC-MS监测。反应结束后将反应液冷却至室温,过滤,然后用乙酸乙酯洗涤,向滤液中加入乙酸乙酯和水,然后用乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到粗产物,粗产物再用石油醚和二氯甲烷重结晶过滤真空干燥得到化合物71-2(2.00g,6.57mmol),LCMS(ESI)m/z:205.0[M+H-100]+
步骤2:化合物71-3的合成:
在干燥的单口瓶中加入底物71-2(2.00g,6.57mmol),溶于乙醇(40mL)中,然后加入水合肼(10mL),50℃下加热反应两个小时。LC-MS监测。反应结束后将反应液冷却至室温,过滤,用乙醇洗涤三次,滤液减压浓缩,残余物水和乙酸乙酯溶解,并用乙酸乙酯萃取四次, 有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物71-3(1.1g,6.31mmol),LCMS(ESI)m/z:160.2[M+H-56+41]+
步骤3:化合物71-5的合成:
在干燥的单口瓶中加入底物71-3(1.1g,6.31mmol)和底物4-氯-2-甲硫基嘧啶-5-羧酸乙酯(71-4,1.40g,6.01mmol),溶于四氢呋喃(20mL)中,然后加入N,N-二异丙基乙胺(1.94g,15.03mmol),80℃下加热反应16个小时。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩得到粗品,然后将粗品溶于乙酸乙酯中,用1M的稀盐酸洗涤五次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物71-5(2.15g,5.81mmol),LCMS(ESI)m/z:371.2[M+H]+
步骤4:化合物71-6的合成:
在干燥的单口瓶中加入底物71-5(2.15g,5.81mmol),溶于二氯甲烷(30mL)中,冰浴冷下然后加入三氟乙酸(10mL),45℃下反应两个小时。LC-MS监测反应完全。将反应液冰浴冷却至0度,缓慢加入40%氢氧化钠水溶液至pH=11,然后加入甲醇(30mL),室温反应6个小时LC-MS监测。反应结束减压浓缩除去有机相,然后加入3M的稀盐酸至pH=1,有黄色沉淀物产生,过滤,并用水洗涤两次,将固体真空干燥得到化合物71-6(785.00mg,3.50mmol)。LCMS(ESI)m/z:225.2[M+H]+
步骤5:化合物71-7的合成:
在干燥的密封管中加入底物4-8(183.6mg,875.66μmol)、底物71-6(206.21mg,919.44μmol)、碘化亚铜(166.77mg,875.66μmol)、碘化钠(262.50mg,1.75mmol)、碳酸钾(302.56mg,2.19mmol)和反-(1R,2R)-N,N'-二甲基1,2-环己烷二胺(249.11mg,1.75mmol),然后加入苯甲醚(4mL),氮气环境下,110℃反应18个小时。TLC监测反应完全。将反应液冷却至室温,过滤,用乙酸乙酯洗涤两次,滤液用氨水洗涤两次,饱和食盐水洗涤两次,有机相用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物71-7(132mg,332.08μmol)。LCMS(ESI+)m/z:398.1[M+H]+
步骤6:化合物77的合成:
在干燥的单口瓶中,加入底物71-7(132mg,332.08μmol),溶于四氢呋喃(5mL)中,然后加入间氯过氧苯甲酸(85.96mg,498.12μmol),室温下反应一个小时,再向反应液中加入N,N-二异丙基乙胺(214.59mg,1.66mmol)和底物11-1(172.28mg,664.16μmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱(碱性)纯化得到化合物71(75.15mg,123.45μmol)。1H NMR(400MHz,DMSO-d6)δ10.05(s,1H),8.74(s,1H),7.83(d,J=8.4Hz,1H),7.63–7.45(m,3H),6.88(d,J=9.2Hz,2H),5.24(d,J=5.2Hz,1H),4.23–4.11(m,1H),3.72(d,J=5.2Hz,1H),3.07(t,J=5.6Hz,4H),2.92–2.75(m,2H),2.35–2.20(m,5H),2.16(s,3H),1.53(t,J=5.6Hz,4H),1.46(t,J=5.6Hz,4H),1.31(d,J=7.2Hz,6H),1.14–1.06(m,1H),0.72–0.62(m,1H),0.46–0.41(m,1H),0.40–0.34(m,1H),0.33–0.27(m,1H).LCMS(ESI)m/z:609.3[M+H]+,HPLC method B:RT=7.55min,purity>95.5%。
实施例72:化合物72的合成
步骤1:化合物72-1的合成:
向单口瓶中,加入底物71-1(2.5g,9.54mmol)、碳酸钾(5.27g,38.16mmol)和苄基三乙基氯化铵(217.34mg,0.95mmol),然后加入乙腈(30mL)溶解,待溶液变成黄色后,加入碘甲烷(2.71g,19.08mmol),60℃下搅拌24小时。LC-MS监测。反应结束后将反应液冷却至室温,过滤,然后用乙酸乙酯洗涤,向滤液中加入乙酸乙酯和水,然后用乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到粗产物,粗产物再用石油醚和二氯甲烷重结晶过滤真空干燥得到化合物72-1(1.80g,6.51mmol),LCMS(ESI)m/z:177.0[M+H-100]+
步骤2:化合物72-2的合成:
在干燥的单口瓶中加入底物72-1(1.80g,6.51mmol),溶于乙醇(30mL)中,然后加入水合肼(8mL),50℃下加热反应两个小时。LC-MS监测。反应结束后将反应液冷却至室温,过滤,用乙醇洗涤三次,滤液减压浓缩,残余物水和乙酸乙酯溶解,并用乙酸乙酯萃取四次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物72-2(0.91g,6.20mmol),LCMS(ESI)m/z:132.2[M+H-56+41]+
步骤3:化合物72-3的合成:
在干燥的单口瓶中加入底物72-2(0.91g,6.20mmol)和底物4-氯-2-甲硫基嘧啶-5-羧酸乙酯(1.37g,5.90mmol),溶于四氢呋喃(20mL)中,然后加入N,N-二异丙基乙胺(1.90g,14.75mmol),80℃下加热反应16个小时。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩得到粗品,然后将粗品溶于乙酸乙酯中,用1M的稀盐酸洗涤五次,有机相用无水硫酸钠干燥,过滤,减压浓缩得到化合物72-3(1.96g,5.71mmol),LCMS(ESI)m/z:343.2[M+H]+
步骤4:化合物72-4的合成:
在干燥的单口瓶中加入底物72-3(1.96g,5.71mmol),溶于二氯甲烷(20mL)中,冰浴冷下然后加入三氟乙酸(80mL),45℃下反应两个小时。LC-MS监测反应完全。将反应液冰浴冷却至0度,缓慢加入40%氢氧化钠水溶液至pH=11,然后加入甲醇(20mL),室温反应6个小时LC-MS监测。反应结束减压浓缩除去有机相,然后加入3M的稀盐酸至pH=1,有黄色沉淀物产生,过滤,并用水洗涤两次,将固体真空干燥得到化合物72-4(647.00mg,3.30mmol)。LCMS(ESI)m/z:197.2[M+H]+
步骤5:化合物72-5的合成:
在干燥的密封管中加入底物4-8(101.76mg,485.34μmol)、底物72-4(100.00mg,509.61μmol)、碘化亚铜(97.05mg,509.61μmol)、碘化钠(153.00mg,1.02mmol)、碳酸钾(175.82mg,1.27mmol)和反-(1R,2R)-N,N'-二甲基1,2-环己烷二胺(144.73mg,1.02mmol),然后加入苯甲醚(2mL),氮气环境下,110℃反应18个小时。TLC监测反应完全。将反应液冷却至室温,过滤,用乙酸乙酯洗涤两次,滤液用氨水洗涤两次,饱和食盐水洗涤两次,有机相 用无水硫酸钠干燥,过滤,减压浓缩,残余物用柱层析纯化得到化合物72-5(36.00mg,97.44μmol)。LCMS(ESI+)m/z:370.1[M+H]+
步骤6:化合物72的合成:
在干燥的单口瓶中,加入底物72-5(36.00mg,97.44μmol),溶于四氢呋喃(1mL)中,然后加入间氯过氧苯甲酸(33.63mg,194.88μmol),室温下反应一个小时,再向反应液中加入N,N-二异丙基乙胺(125.70mg,974.40μmol)和底物11-1(50.55mg,194.88mmol),45℃反应过夜。LC-MS监测。反应结束后将反应液冷却至室温,减压浓缩,残余物用制备高效液相色谱(碱性)纯化得到化合物72(10.23mg,36.44μmol)。1H NMR(400MHz,DMSO-d6)δ10.07(s,1H),8.79(s,1H),7.86(d,J=8.4Hz,1H),7.71(d,J=8.4Hz,1H),7.57(s,2H),6.92(d,J=9.2Hz,2H),5.30(d,J=5.2Hz,1H),3.75(d,J=5.2Hz,1H),3.38(s,3H),3.09(t,J=5.6Hz,4H),2.92–2.81(m,2H),2.39–2.27(m,4H),2.19(s,3H),1.54(t,J=5.6Hz,4H),1.48(t,J=5.6Hz,4H),1.27–1.21(m,1H),1.18–1.10(m,1H),0.70–0.61(m,1H),0.47–0.40(m,1H),0.39–0.32(m,2H).LCMS(ESI)m/z:581.6[M+H]+,HPLC method B:RT=6.70min,purity>92.9%。
效果实验例:生物活性评价试验
无特殊说明时,该部分实施例中部分生物学评价实验用化合物AZD1775和ZnC3作为对照,AZD1775(CAS号:955365-80-7)和ZnC3(CAS号:2376146-48-2)的结构信息如下:
效果实验例1:通过TR-FRET方法评价化合物在Wee1蛋白与Tracer 178结合。
首先准备不同浓度梯度的化合物溶液。将化合物溶解于DMSO中,并将化合物进行4倍稀释,共10个剂量点,每个浓度设置2个平行重复,加入DMSO作为阳性对照(最大信号对照)和阴性对照(最小信号对照),同时确保每个反应孔中的DMSO最终含量为0.25%。
将配置于缓冲液(50mM HEPES pH 7.5,10mM MgCl2,1mM EGTA,0.01%Brij-35)WEE1(Thermo Fisher,Cat#PR7373A)蛋白最终反应浓度为15nM),不同浓度的化合物中,反应底物Tracer 178(Invitrogen,PV5593)和MAb Anti-GST-Eu crypate(Cisbio,61GSTKLA)加入384孔板(Corning,cat#3574)中,1000rpm离心1min,将384孔反应板置于恒温摇床孵育60min,25℃,300rpm。其中Tracer 178和MAb Anti-GST-Eu crypate配置于缓冲液(50mM HEPES pH 7.5,10mM MgCl2,1mM EGTA,0.01%Brij-35),且Tracer178最终反应浓度为50nM,MAb Anti-GST-Eu crypate的最终浓度为2nM,其中阴性对照(最小信号对照)使用等量的缓冲液来代替蛋白溶液。
孵育完成后,使用BMG PHERAStar读数(用337nm波长为激发光,在620nm和665nm波长值为发射光读荧光信号值)。计算荧光信号比值:665/620*1000为最终的酶活性信号值,根据阳性对照(最大信号对照)和阴性对照(最小信号对照)获得的读数TR-FRET信号进行标准化,以给出不同浓度化合物的抑制率。再通过GraphPad Prism 6以log(inhibitor)vs.response–Variable slope模式拟合计算得出化合物对酶活性抑制的IC50。拟合方程为: Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope)),其中Y代表已知的百分剩余酶活性,X代表Log后的已知化合物的浓度。
按照上述方法对实施例化合物进行Wee1抑制活性检测,试验结果见表1,其中测定各化合物的IC50按照说明分类如下:
“-”表示IC50测定值大于10μM;
“+”表示IC50测定值小于等于10μM大于1μM;
“++”表示IC50测定值小于等于1μM大于100nM;
“+++”表示IC50测定值小于等于100nM大于10nM;
“++++”表示IC50测定值小于等于10nM大于5nM;
“+++++”表示IC50测定值小于等于5nM。
表1本发明化合物对Wee1激酶的抑制活性
结论:本发明化合物具有良好的Wee1激酶抑制活性。
效果实验例2:化合物抗细胞增殖活性评价
A通过Cell Titer-Glo方法评价化合物对BxPC3、HT-29和OVCAR-3细胞抗增殖的效果。
配置不同浓度梯度的化合物溶液。取DMSO溶解到浓度为10mM测试化合物和10mM参考化合物AZD1775,将化合物进行系列稀释于培养基中,共9个剂量点,每个浓度设置2个平行重复。不添加化合物的细胞生长组作为阳性对照(最大信号对照),将培养基作为阴性对照(最小信号对照),同时确保每个反应孔中的DMSO最终含量为0.2%。移除384孔板中的培养基后,将配置好的25μL的不同浓度化合物转移进孔板中,化合物和细胞在细胞培养箱37℃,5%CO2孵育3天。
将384孔板从细胞培养箱中取出使其平衡1h至室温,再将25μL的Cell Titer-Glo检测试剂加入每个反应孔,摇床裂解2min后,于孵育10min后,用BMG PHERAStar读数(Luminescence)。根据发光信号计算抑制率。
来计算出不同浓度化合物对细胞的抑制率。通过GraphPad Prism 6以log(inhibitor)vs.response–Variable slope模式拟合计算得出化合物对细胞活性抑制的IC50。拟合方程为:Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope)),其中Y代表抑制率,X代表Log后的已知化合物的浓度。
按照上述方法对实施例化合物进行BxPC3、HT-29和OVCAR-3体外抗细胞增殖试验,试验结果见表2,其中测定各化合物的IC50按照说明分类如下:
“-”表示IC50测定值大于10μM;
“+”表示IC50测定值小于等于10μM大于5μM;
“++”表示IC50测定值小于等于5μM大于2μM;
“+++”表示IC50测定值小于等于2μM大于1μM;
“++++”表示IC50测定值小于等于1μM大于0.1μM;
“+++++”表示IC50测定值小于等于0.1μM。
表2本发明化合物对BxPC3、HT-29和OVCAR-3体外细胞增殖抑制活性

结论:本发明化合物对肿瘤细胞增殖具有良好的抑制作用,抗增殖活性数据部分化合物与对照化合物AZD1775相当或更优。
B通过Cell Titer-Glo方法评价化合物对正常细胞HUVEC和HK2的细胞抗增殖的效果。
测试方法同对肿瘤细胞的抗增殖活性评价。对两株正常细胞的测试结果如表3所示。
表3化合物对HUVEC和HK2细胞增殖抑制活性
结论:本发明化合物对正常细胞HUVEC增殖抑制活性较弱,对HK2细胞具有一定的抑制作 用。与对照化合物相比,本发明化合物大部分表现出相当或者更低的增殖抑制活性,具有更好的安全性。
效果实验例3:体外肝微粒体(小鼠和人)代谢稳定性评价
1.工作液的配制:
微粒体从-80℃冰箱取出,在37℃水浴锅中迅速融解,置于冰上待用。将供试品用DMSO稀释配制成10mM的储备液,然后用乙腈稀释成0.5mM的次级储备液。使用Buffer C将微粒体稀释为0.75mg/ml;再加入次级储备液至化合物终浓度1.5μM为工作液,依据n=2,5个时间点计算,每个化合物配制350μL,使用前置于冰上。用Buffer C将NADPH稀释为6mM的工作液,为启动液。配制含内标的乙腈溶液作为沉淀剂,内标选用Verapamil-HCl,浓度为4ng/ml。
2.实验过程:
取一块圆底孔板,记做反应板,将各化合物配置好的工作液按照复样数和时间点分装到孔板中(0h的样品同样加在反应板上),30μL/孔;反应板于37℃孵育10分钟。另取一块尖底孔板,记做沉淀板,每孔加入135μL沉淀剂;0h的样品在孵育10分钟后转移至沉淀板中,再加入15μL启动液,沉淀板在离心前置于冰上待用。
将稀释好的启动液足量加入至分装板中,方便排枪吸取操作。
反应在温孵震荡装置上进行,使用排枪吸取启动液15μL/样品,加入反应板中。稍震荡混匀以启动反应,使用计时器准确计时并记录;
到反应时间后,使用排枪将反应板中的所有溶液吸出加入到沉淀板中以终止该时间点反应。所有反应终止后,将沉淀板在摇板机上600rpm震荡十分钟沉淀蛋白。离心机最大转数在4℃离心15分钟,取上清80μL,加入320μL纯水,混匀后进行LC-MS分析。
3.测试结果如表4所示。
表4不同种属肝微粒体稳定性数据
结论:本发明化合物,在五种不同种属肝微粒体上均体现出良好的代谢稳定性。
效果实验例验例4:溶解度评价
将化合物置对应pH下的缓冲溶液中,恒温振摇24h,取上清液配制成约100μg/ml供试品溶液,采用反相高效液相色谱法,梯度洗脱,外标法计算溶解度。色谱条件:C18柱,流动相A:0.02M磷酸二氢钾:乙腈=90:10,流动相B:乙腈;V:1.0ml/min,T:35℃,λ:210nm。测试结果如表5所示。
表5化合物在不同pH条件下的溶解度

结论:在三个pH条件下,本发明化合物37b与对照化合物ZnC3的溶解度相近,总体上本发明化合物的溶解度明显优于对照化合物AZD1775。
效果实验例5:细胞渗透性评价
Caco-2细胞购买自美国模式组织细胞收藏中心(Rockville,MD)。细胞培养液为含10%灭活胎牛血清和1%非必须氨基酸的改良Eagle’s培养基(MEM)。细胞接种于聚碳酸脂滤膜(货号:3396)并置于37℃,5%CO2培养箱中培养。
细胞接种后培养21~28天可用于转运实验,并通过路西法黄的表观通透系数(Papp)来表征和验证细胞单层的致密性。实验中将化合物溶解于DMSO中制备10mM的储备液,并使用含有25mM HEPES(pH 7.4)的汉克斯平衡盐溶液(HBSS,Invitrogen,Cat#14025-092)进行稀释得到工作液。将10μM的待测化合物工作液加入Caco-2顶膜侧(apical side)和基底侧(basolateral side)并于37℃孵育90分钟,孵育结束后,将顶膜侧和基底侧的样品稀释,并通过LC-MS/MS检测顶膜侧和基底侧化合物的浓度,并通过标准曲线定量计算化合物的浓度。
测试结果如表6所示:
表6化合物在Caco2模型上的渗透性数据
结论:本发明化合物在Caco2模型上的透膜性与对照化合物相当。总体来看本发明化合物和对照化合物的细胞渗透性均不佳,且具有一定的外排属性。
效果实验例6:血浆蛋白结合率(PPB)评价
1.实验过程
样品准备:
将化合物用DMSO溶解为10mM的储备液,然后用PBS将化合物稀释为0.02mM的次级储备液,再使用空白血浆将上述0.02mM稀释至1μM,即为待孵育样品。
透析装置准备:
先在平衡透析板的白色孔中加入400μL空白PBS,红色孔中加入200μL配置好的血浆样品,用封口膜将透析板进行封口
回收率孔板准备:
准备两块96孔深孔板,标记为T0及T5,所有血浆样品以n=2分别加入两板。T0板直接加入300μL乙腈(Verapamil-HCl,4ng/mL),再补加50μL空白PBS混匀5min,放入4℃ 冰箱中静置至孵育实验结束。
实验操作:
透析装置和T5板置于微孔板恒温振荡器中共同孵育5h(37℃,使用300rpm或最小转速)。孵育结束后,加入300μL乙腈(Verapamil-HCl,4ng/mL),再补加50μL PBS溶液。透析孵育结束后,取一块新96孔深孔板。取50μL血浆孔样品加入96孔板对应位置,再加入300μL乙腈,补加50μL空白PBS;取50μL缓冲孔样品加入96孔板对应位置,再加入300μL乙腈,补加50μL空白血浆。T5板含血浆孔中加入300μL乙腈(Verapamil-HCl,4ng/mL),再补加50μL PBS溶液。振荡5min以充分沉淀蛋白,4℃,20000g离心10分钟。取200μL上清液加入200μL纯水中,混匀后进行LC-MS/MS分析。
2.数据处理和参数计算
●血浆蛋白结合率=[(Rpe-Rb)/Rpe]×100%
●回收率=[(Rpe+Rb)/R5h]×100%
●稳定性=(R5/R0)×100%
其中:
●Rpe=血浆侧供试品峰面积与内标比值
●Rb=缓冲液侧供试品峰面积与内标比值
●R5=孵箱稳定性样品峰面积与内标比值
●R0=冰箱稳定性样品峰面积与内标比值
3.测试结果如表7所示:
表7代表性化合物的血浆蛋白结合率(小鼠/大鼠/人)
结论:本发明化合物具有良好的血浆蛋白结合能力。
效果实验例7:化合物对细胞色素P450抑制评价
酶学实验通过细胞色素P450对底物的氧化产生的荧光,定量检测小分子抑制剂对CYP450各亚型酶活性的抑制情况。实验在384孔板(Corning,Cat#3575)中进行,使用的反应缓冲液为:142.86mM Potassium Phosphate,pH 7.4。实验所用Solution A成分为:26.13mM NADP+(Sigma-aldrich,Cat#N0505),65.77mM G6P(J&K,Cat#968161)及65.42mM  MgCl2(Sigma-aldrich,Cat#M2670)。实验所用的Solution B成分为:40U/mL G6PDH(Sigma-aldrich,Cat#G6378)。底物混合溶液成分为:0.05X Solution A,0.01X Solution B,50mM Potassium Phosphate,0.01mM BOMCC/0.01mM EOMCC/0.001mM DBOMF。对于CYP3A4和CYP2C9,反应体系分别为50μL或20μL,包括3nM CYP3A4或120nM CYP2C9,BOMCC底物混合溶液和不同浓度的待测化合物。对于CYP2C19,CYP2D6及CYP1A2,反应体系为20μL,包括12.5nM CYP2C19,80nM CYP2D6或1nM CYP1A2,EOMCC底物混合溶液和不同浓度的待测化合物。对于CYP2C8,反应体系为50μL,包括1.5nM CYP2C8,DBOMF底物混合溶液和不同浓度的待测化合物。化合物与酶预孵10分钟后,加入底物,并根据不同底物使用BMG PHERAStar读取不同波段的荧光信号(BOMCC/EOMCC Ex430nm/Em480nm,DBOMF Ex490nm/Em520nm),反应间隔30秒或更多(根据实际实验孔数设置),反应时长为30分钟。实验数据通过GraphPad Prism 6软件进行分析处理得到IC50值。
测试结果如表8所示:
表8化合物对7中CYP亚型的抑制活性
结论:本发明化合物均未表现出明显的细胞色素P450酶抑制,其中本发明化合物37b对所有亚型的抑制均大于10μM,优于对照化合物AZD1775。
效果实验例8:hERG钾离子通道抑制试验
实验步骤:
(一)实验材料:
A.CHO(中华仓鼠卵巢细胞)稳定转染细胞系培养
膜片钳实验所用细胞株为过表达hERG钾离子通道cDNA的第10代CHO细胞。CHO hERG细胞在37℃、5%CO2培养箱中用培养皿或培养瓶培养。电生理实验前24-48小时,将细胞滴于圆形玻片上在细胞培养液中培养,待细胞贴壁后用于实验。
细胞培养基(购自Invitrogen)成分:
·Ham’s F12培养基
·10%(v/v)热灭活FBS
·100μg/ml Hygromycin B(潮霉素)
·100μg/ml Geneticin(遗传霉素,G418)
B.化合物准备
化合物粉末溶解在细胞外液中,都经过常规的5到10分钟超声和振荡以保证化合物完全溶解。
用于电生理检测的化合物终浓度为5、20μM,DMSO的终浓度为0.1%。
(二)实验方案:
A.电生理记录实验过程
细胞膜电流记录使用HEKA EPC-10 USB膜片钳放大器(德国HEKA Elektronik)。
1)取表面有大量单个CHO hERG细胞均匀生长的盖玻片,放置于倒置显微镜上的连续记录池中,灌流细胞外液(大约每分钟1毫升)并持续记录,等待电流稳定。
2)使用标准的全细胞记录模式记录单个细胞的HERG通道电流。首先将膜电压钳制在-80mV,给予细胞持续5s,+20mV电压刺激,以激活hERG钾通道,再复极化至-50mV,持续5s,产生外向尾电流,持续灌注待电流稳定,此时尾电流峰值即为对照电流值。
3)接着灌流含待测药物的细胞外液并持续记录直到药物对hERG电流的抑制作用到达稳定状态,此时尾电流峰值即为加药后电流值。
4)再次用细胞外溶液灌注细胞,直到hERG电流回复或接近加药物之前的水平,则可以继续灌流测试其它浓度或药物。可在每个细胞上测试一种或多种化合物或药物浓度。
5)以Cisapride(C4740-10mg,Sigma)作为实验中的阳性对照以保证所使用的细胞反应正常。
(三)质量控制
试验数据需要满足以下标准:
电生理记录参数
a)封接电阻>500MΩ
b)接触电阻(Ra)<10MΩ
c)初始尾电流幅度>200pA
d)电流rundown(自发性减小)<2%/min
e)漏电流<200pA或者hERG电流峰值的10%(在90%的记录时间之内)
测试结果如表9所示:
表9化合物的hERG抑制活性
结论:部分本发明化合物(如4b,37b)的hERG抑制活性弱于对照化合物AZD1775,其心脏毒性风险相对更低。
效果实验例9:药物代谢动力学评价
(一)小鼠单次给药药代动力学评价
本实验旨在研究受试化合物单次静脉、单次口服给药后,在雄性ICR小鼠血浆中的药代动力学情况
1.试验目的
本试验的目的是为了获得受试化合物在ICR小鼠的药物代谢动力学情况(含静脉和口服)
2.规范依从性
本试验(非GLP研究)供试品-检测,DMPK动物试验,DMPK分析均在成都先导完成,所有试验均遵循本试验方案,及相关机构的相关SOP。
3.试验材料、仪器和设备
3.1试验材料
3.1.1供试品
以下供试品由成都先导提供,并确保其质量符合要求。
3.1.2试验系统
种属:SPF级雄性ICR小鼠
体重/周龄:30g左右
数量:6只
实验动物来源:维通利华
4.试验过程
4.1溶媒配制
静注溶媒:5%DMSO-10%Solutol-85%HPBCD(20%,W/V)
“灌胃溶媒/剂型:5%DMSO-10%Solutol-85%HPBCD(20%,W/V)”
4.2受试物给予(途径)
静脉注射:给药剂量1mg/kg,给药体积5mL/kg
灌胃:给药剂量10mg/kg,给药体积10mL/kg
给药前超声5min
4.3受试物配制(浓度)
静脉:0.2mg/ml
灌胃:1mg/ml
给药前禁食过夜,给药后四小时喂食。
4.4样品采集
给药后5分钟(仅IV)、15分钟、30分钟、1小时、2小时、4小时、8小时、24小时分别于每个时间点经眼眶静脉丛穿刺采血(40-50ul)至含预喷涂EDTA-K2的抗凝管中,血液样品1小时内以10000rpm离心20分钟(离心前后至于湿冰上保存)取上层液即血浆,于-20℃及以下冰箱冷冻保存,以备LC-MS/MS分析。
4.5生物药品分析方法及检测:用LC-MS/MS分析检测。
5数据处理
样品通过LC-MS/MS检测各时间点的药物浓度。采用Phoenix WinNonlin 5.2非房室模型计算药代动力学参数末端消除半衰期(t1/2)、药时曲线下面积(AUC)、表观分布容积(Vd)、清除率(CL)、平均驻留时间(MRT)、Cmax、生物利用度(F%)直接从血清浓度结果中获得。血药浓度及药代参数等采用均数±标准差(X±SD)。具体检测分析方法以方案修订的形式明确。
6试验结果如表10所示:
表10化合物AZD1775和37b在ICR小鼠上的药代动力学参数

结论:在小鼠体内药代动力学检测分析发现,本发明化合物37b与对照化合物AZD1775相比,其药代动力学性质更优,化合物37b的药物暴露更高、清除更慢。
(二)大鼠单次给药药代动力学评价
采用与小鼠体内药代动力学研究方法,评价药物在雄性SD大鼠中的药代动力学性质。结果如表11所示:
表11化合物AZD1775和37b在SD大鼠上的药代动力学参数
结果:本发明化合物37b和对照化合物AZD1775的最大血药浓度值相当,药物暴露情况本发明化合物37b静脉注射优势明显,表明化合物37b清除更慢,口服暴露水平化合物37b也优于对照化合物AZD1775。
(三)比格犬单次给药代动力学评价
采用小鼠药代动力学评价方法,开展雄性比格犬的药待动力学评价。基本信息如下:
比格犬:体重8-11公斤,购自Jiangsu Marshall biotechnology Co.LTD
给药剂量:2mg/kg(静脉注射);10mg/kg(口服)
溶媒:5%DMSO+5%Solutol+90%(20%HP-β-CD in saline)
测试结果如表12所示:
表12化合物37b和AZD1775在比格犬上的药代动力学参数
结论:在比格犬上,本发明化合物37b的药代动力学性质略优于对照化合物AZD1775。
效果实验例10:动物体内药效评价(CDX模型)
(一)化合物对人结肠癌HT-29细胞皮下异种植瘤雌性BALB/c裸鼠体内药效学评价
试验目的:评价化合物37b和AZD1775对异种荷瘤(HT-29)裸鼠的抑瘤药效评价
试验方法:选用BALB/c裸小鼠(维通利华提供),7-8周龄,体重19-22g
准备并培养好的HT-29细胞,计数,将5×106个HT-29细胞混于0.1mL PBS溶液中,皮下接种于小鼠右翼。待细胞成瘤,平均肿瘤体积达到120mm3时,开始进行分组并开始给药。给药剂量:AZD1775,60毫克/公斤,每日一次;37b设置3个剂量组分别为30毫克/公斤、60毫克/公斤和120毫克/公斤,给药频次每日一次。试验指标是考察肿瘤生长是否被抑制。肿瘤体积计算公式为:V=0.5a×b2,a和b分别表示肿瘤的长径和短径。
化合物的抑瘤效果用TGI(%)来评价。TGI(%)的计算:TGI(%)=[1-(某处理组给药结束时平均瘤子体积-该处理组开始给药时平均瘤子体积)/(溶媒对照组给药结束时平均瘤子体积-溶媒对照组开始给药时平均瘤子体积)]×100%。
在给药24天后,化合物37b和AZD1775的抑瘤效果如表13所示:
表13化合物AZD1775和37b在HT-29模型上的抑瘤效果
结论:在60毫克/公斤剂量下,本发明化合物37b与对照化合物AZD1775的抑瘤效果相当;在120毫克/公斤剂量下,本发明化合物37b具有显著的肿瘤生长抑制作用。
(二)化合物对人胰腺癌BxPC3细胞皮下异种植瘤雌性BALB/c裸鼠体内药效学评价试验目的:评价化合物37b和AZD1775对异种荷瘤(BxPC3)裸鼠的抑瘤药效评价
试验方法:选用BALB/c裸小鼠(维通利华提供),7-8周龄,体重19-22g
准备并培养好的BxPC3细胞,计数,将1×107个BxPC3细胞混于0.1mL PBS溶液中,皮下接种于小鼠右翼。待细胞成瘤,平均肿瘤体积达到190mm3时,开始进行分组并开始给药。给药剂量:AZD1775,60毫克/公斤,每日一次;37b设置3个剂量组分别为30毫克/公斤、60毫克/公斤和120毫克/公斤,给药频次每日一次。试验指标是考察肿瘤生长是否被抑制。肿瘤体积计算公式为:V=0.5a×b2,a和b分别表示肿瘤的长径和短径。
化合物的抑瘤效果用TGI(%)来评价。TGI(%)的计算:TGI(%)=[1-(某处理组给药结束时平均瘤子体积-该处理组开始给药时平均瘤子体积)/(溶媒对照组给药结束时平均瘤子体积-溶媒对照组开始给药时平均瘤子体积)]×100%。
在给药40天后,化合物37b和AZD1775的抑瘤效果如表14所示:
表14化合物AZD1775和37b在HT-29模型上的抑瘤效果
结论:在30毫克/公斤和30毫克/公斤剂量下,本发明化合物37b比对照化合物AZD1775抑瘤效果更佳;在120毫克/公斤剂量下,化合物37b具有显著的肿瘤生长抑制作用。
效果实验例11:化合物毒理评价
(一)大鼠单次给药毒性——最大耐受剂量评价
试验目的:化合物AZD1775和37b单次给药下,大鼠耐受性评价
试验动物:SD大鼠(维通利华提供),6-8周龄,180-220克。
试验方法:按预定剂量对大鼠进行单次口服给药,后续考察大鼠的生存情况(最长持续观察14天),试验方案及结果请见表15。
表15 AZD1775和37b大鼠单次给药最大耐受剂量试验方案
试验结果:在大鼠单次给药中,本发明化合物37b的最大耐受剂量高于对照化合物AZD1775,具有明显的安全性优势。
(二)大鼠连续14天给药毒性评价
试验目的:化合物AZD1775、ZnC3和37b在连续给药情况下,大鼠耐受性表现和化合物毒性评价
试验动物:SD大鼠(维通利华提供),6-8周龄,180-220克。
试验方法:按预定剂量对大鼠进行,每日一次口服给药,后续考察大鼠的生存、体重变化(最长持续给药14天),试验方案请见表16。
表16大鼠14天连续给药毒研究方案

其中毒性代谢动力学研究取样如下:首末次给药取样;取样时间点:15分钟,30分钟,1小时,2小时,4小时,8小时和24小时。
试验结果:
1.大鼠总体存活情况
按照上面的给药方案进行14天连续给药,发现部分剂量组大鼠由死亡现象,结果如表17所示。
表17连续给药14天大鼠存活情况统计
结果:本发明化合物37b组大鼠的生存率明显优于对照化合物AZD1775组和对照化合物ZnC3组。
2.毒性代谢动力学参数
三个化合物在大鼠上的毒性代谢动力学测试结果如表18(雌性大鼠)和表19(雄性大鼠)所示。
表18雌性大鼠毒性代谢动力学参数
表19雌性大鼠毒性代谢动力学参数
结论:从大鼠单次给药最大耐受剂量和大鼠连续14天给药毒性研究结果分析,本发明化合物37b在药物安全性上较对照化合物AZD1775和对照化合物ZnC3具有明显的优势。

Claims (17)

  1. 式I所示的化合物、或其立体异构体、或其药学上可接受的盐:
    其中,所述的R1选自-C1~6烷基、-C2~6烯基、-C2~6炔基、-C0~2亚烷基-CN、-C0~2亚烷基-(3~10元环烷基)、-C0~2亚烷基-(3~10元杂环烷基);
    R2选自
    所述的X选自O、NH或CH2
    所述的X1选自CH或N;
    R21、R22、R29分别独立地选自氢、氘、卤素、氰基、硝基、-OH、-C1~6烷基、卤素取代的C1~6烷基、-C0~2亚烷基-OH、-O(C1~6烷基)、-O(卤素取代的C1~6烷基)、-NH2、-C0~2亚烷基-NH(C1~6烷基)、-C0~2亚烷基-N(C1~6烷基)(C1~6烷基)、-C0~2亚烷基-(3~10元环烷基)、-C0~2亚烷基-(3~10元杂环烷基);
    所述的R23、R24与其直接相连的原子一起形成3~10元碳环、3~10元杂环;
    所述的R25、R26与其直接相连的原子一起形成3~10元碳环、3~10元杂环;
    所述的R27、R28与其直接相连的原子一起形成3~10元碳环、3~10元杂环;
    R3选自氢、氘、卤素、氰基、硝基、-C1~6烷基、卤素取代的C1~6烷基、-C0~2亚烷基-OH、-O(C1~6烷基)、-O(卤素取代的C1~6烷基)、-NH2、-C0~2亚烷基-NH(C1~6烷基)、-C0~2亚烷基-N(C1~6烷基)(C1~6烷基);
    所述的R4选自3~12元杂环烷基;所述的杂环烷基可进一步被一个、两个、三个或四个 独立的R41取代;
    所述的R41选自氢、卤素、氰基、硝基、-OH、-C1~6烷基、卤素取代的C1~6烷基、-C0~2亚烷基-OH、-O(C1~6烷基)、-O(卤素取代的C1~6烷基)、-NH2、-C0~2亚烷基-NH(C1~6烷基)、-C0~2亚烷基-N(C1~6烷基)(C1~6烷基)、-C(O)C1~6烷基、3~10元碳环、3~10元杂环;所述碳环、杂环可进一步被一个、两个、三个或四个独立的R31取代;
    或者,所述的R3、R4与其直接相连的原子一起形成3~10元碳环、3~10元杂环;所述的碳环、杂环烷基可进一步被一个、两个、三个或四个独立的R31取代;
    所述的R31选自氢、卤素、氰基、硝基、-OH、-C1~6烷基、卤素取代的C1~6烷基、-C0~2亚烷基-OH、-O(C1~6烷基)、-O(卤素取代的C1~6烷基)、-NH2、-C0~2亚烷基-NH(C1~6烷基)、-C0~2亚烷基-N(C1~6烷基)(C1~6烷基)。
  2. 根据权利要求1所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:所述的R1选自甲基、乙基、
  3. 根据权利要求1所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:R21、R22、R29分别独立地选自氢、氘、氰基、甲基、乙基、-OH、三氟甲基、环丙基、-CH2OH、-NH2
  4. 根据权利要求1所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:所述的R23、R24与其直接相连的原子一起形成环丙基、环丁基、环戊基;
    所述的R25、R26与其直接相连的原子一起形成环丙基、环丁基、环戊基;
    所述的R27、R28与其直接相连的原子一起形成环丙基、环丁基、环戊基。
  5. 根据权利要求1或3或4所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:所述的R2选自
  6. 根据权利要求1所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:所述的R3选自氢、氟、甲基、-CH2OH、甲氧基。
  7. 根据权利要求1所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:所述的R4选自含氮6元杂环、7元含氮桥环、8元含氮桥环、9元含氮杂螺环、11元含氮杂螺环。
  8. 根据权利要求7所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在 于:所述的R4选自 其中R41如权利要求1所述。
  9. 根据权利要求8所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:所述的R4选自
  10. 根据权利要求1所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:所述的R3、R4与其直接相连的原子一起形成6元含氮杂环。
  11. 根据权利要求10所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:所述的R3、R4与其直接相连的原子一起形成其中R31如权利要求1所述。
  12. 根据权利要求11所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:所述的R31选自甲基。
  13. 根据权利要求1所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:式I所述的化合物具体为:






























  14. 根据权利要求1所述的化合物、或其立体异构体、或其药学上可接受的盐,其特征在于:式I所述的化合物具体为:
  15. 权利要求1-14任一项所述的化合物、或其立体异构体、或其药学上可接受的盐在制 备治疗WEE1介导的疾病的药物中的用途。
  16. 权利要求15所述的用途,其特征在于:所述WEE1介导的疾病是与炎症、自身免疫性疾病、感染性疾病、癌症、癌前期综合征相关的疾病中的一种或几种。
  17. 一种药物组合物,其特征在于:它是以权利要求1~14任一项所述的化合物、或其立体异构体、或其药学上可接受的盐,加上药学上可接受的辅料制备而成的制剂。
PCT/CN2023/072296 2022-07-13 2023-01-16 Wee1抑制剂及其制备和用途 Ceased WO2024011883A1 (zh)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US18/867,250 US20250304587A1 (en) 2022-07-13 2023-01-16 Wee1 inhibitor, preparation therefor and use thereof
EP23838385.5A EP4570807A1 (en) 2022-07-13 2023-01-16 Wee1 inhibitor, preparation therefor and use thereof
KR1020247043576A KR20250034312A (ko) 2022-07-13 2023-01-16 Wee1 억제자, 그 제조방법 및 그 용도
CA3248185A CA3248185A1 (en) 2022-07-13 2023-01-16 WEE1 INHIBITOR, ITS PREPARATION AND USE
AU2023306869A AU2023306869A1 (en) 2022-07-13 2023-01-16 Wee1 inhibitor, preparation therefor and use thereof
JP2024568139A JP2025525290A (ja) 2022-07-13 2023-01-16 Wee1阻害剤およびその製造と用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210817976.2 2022-07-13
CN202210817976 2022-07-13

Publications (1)

Publication Number Publication Date
WO2024011883A1 true WO2024011883A1 (zh) 2024-01-18

Family

ID=89491409

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/072296 Ceased WO2024011883A1 (zh) 2022-07-13 2023-01-16 Wee1抑制剂及其制备和用途

Country Status (9)

Country Link
US (1) US20250304587A1 (zh)
EP (1) EP4570807A1 (zh)
JP (1) JP2025525290A (zh)
KR (1) KR20250034312A (zh)
CN (1) CN117402162A (zh)
AU (1) AU2023306869A1 (zh)
CA (1) CA3248185A1 (zh)
TW (1) TW202402273A (zh)
WO (1) WO2024011883A1 (zh)

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007126122A1 (en) 2006-04-27 2007-11-08 Banyu Pharmaceutical Co., Ltd. Dihydropyrazolopyrimidinone derivatives
WO2008133866A1 (en) 2007-04-25 2008-11-06 Merck & Co., Inc. Polymorph of dihydropyrazolopyrimidinone derivative as weel kinase.inhibitor
WO2013012681A1 (en) 2011-07-15 2013-01-24 Abbott Laboratories Tricyclic inhibitors of kinases useful for the treatment of proliferative diseases
WO2013126656A1 (en) 2012-02-23 2013-08-29 Abbvie Inc. Pyridopyrimidinone inhibitors of kinases
WO2014167347A1 (en) 2013-04-11 2014-10-16 Almac Discovery Limited 2-aminopyrido[4,3-d]pyrimidin-5-one derivatives and their use as wee-1 inhibitors
WO2015092431A1 (en) 2013-12-19 2015-06-25 Almac Discovery Limited Pyrimidopyrimidinones useful as wee-1 kinase inhibitors
WO2017075629A2 (en) * 2015-11-01 2017-05-04 The Regents Of The University Of Colorado, A Body Corporate Wee 1 kinase inhibitors and methods of making and using the same
WO2018011569A1 (en) 2016-07-12 2018-01-18 Almac Discovery Limited Wee-1 inhibiting pyrazolopyrimidinone compounds
WO2018011570A1 (en) 2016-07-12 2018-01-18 Almac Discovery Limited Wee-1 inhibiting pyridopyrimidinone compounds
WO2018090939A1 (zh) 2016-11-16 2018-05-24 上海瑛派药业有限公司 8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物
WO2018133829A1 (zh) 2017-01-23 2018-07-26 南京明德新药研发股份有限公司 作为Wee1抑制剂的1,2-二氢-3H-吡唑[3,4-d]嘧啶-3-酮衍生物
WO2018171633A1 (zh) 2017-03-23 2018-09-27 上海迪诺医药科技有限公司 吡唑[3,4-d]嘧啶-3-酮的大环衍生物、其药物组合物及应用
CN113387962A (zh) * 2020-03-12 2021-09-14 上海迪诺医药科技有限公司 吡唑并[3,4-d]嘧啶-3-酮衍生物、其药物组合物及应用
CN115403582A (zh) * 2021-05-28 2022-11-29 江苏天士力帝益药业有限公司 Wee1抑制剂及其用途

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007126122A1 (en) 2006-04-27 2007-11-08 Banyu Pharmaceutical Co., Ltd. Dihydropyrazolopyrimidinone derivatives
WO2008133866A1 (en) 2007-04-25 2008-11-06 Merck & Co., Inc. Polymorph of dihydropyrazolopyrimidinone derivative as weel kinase.inhibitor
WO2013012681A1 (en) 2011-07-15 2013-01-24 Abbott Laboratories Tricyclic inhibitors of kinases useful for the treatment of proliferative diseases
WO2013126656A1 (en) 2012-02-23 2013-08-29 Abbvie Inc. Pyridopyrimidinone inhibitors of kinases
WO2014167347A1 (en) 2013-04-11 2014-10-16 Almac Discovery Limited 2-aminopyrido[4,3-d]pyrimidin-5-one derivatives and their use as wee-1 inhibitors
WO2015092431A1 (en) 2013-12-19 2015-06-25 Almac Discovery Limited Pyrimidopyrimidinones useful as wee-1 kinase inhibitors
WO2017075629A2 (en) * 2015-11-01 2017-05-04 The Regents Of The University Of Colorado, A Body Corporate Wee 1 kinase inhibitors and methods of making and using the same
WO2018011569A1 (en) 2016-07-12 2018-01-18 Almac Discovery Limited Wee-1 inhibiting pyrazolopyrimidinone compounds
WO2018011570A1 (en) 2016-07-12 2018-01-18 Almac Discovery Limited Wee-1 inhibiting pyridopyrimidinone compounds
WO2018090939A1 (zh) 2016-11-16 2018-05-24 上海瑛派药业有限公司 8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物
WO2018133829A1 (zh) 2017-01-23 2018-07-26 南京明德新药研发股份有限公司 作为Wee1抑制剂的1,2-二氢-3H-吡唑[3,4-d]嘧啶-3-酮衍生物
CN110198943A (zh) * 2017-01-23 2019-09-03 石家庄智康弘仁新药开发有限公司 作为Wee1抑制剂的1,2-二氢-3H-吡唑[3,4-d]嘧啶-3-酮衍生物
WO2018171633A1 (zh) 2017-03-23 2018-09-27 上海迪诺医药科技有限公司 吡唑[3,4-d]嘧啶-3-酮的大环衍生物、其药物组合物及应用
CN113387962A (zh) * 2020-03-12 2021-09-14 上海迪诺医药科技有限公司 吡唑并[3,4-d]嘧啶-3-酮衍生物、其药物组合物及应用
CN115403582A (zh) * 2021-05-28 2022-11-29 江苏天士力帝益药业有限公司 Wee1抑制剂及其用途

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
CANCER BIOLOGY &THERAPY, vol. 9, no. 7, 2010, pages 523 - 525
CLINICAL CANCER RESEARCH, vol. 17, no. 13, 2011, pages 4200 - 4207
MOLECULAR & CELLULAR BIOLOGY, vol. 32, no. 20, 2012, pages 4226
MOLECULAR CANCER THERAPEUTICS, vol. 12, no. 12, 2013, pages 2675 - 2684
MOLECULAR CANCER, vol. 13, no. 1, 2014, pages 72
no. 2376146-48-2
ONCOTARGET, vol. 7, no. 31, 2016, pages 49902 - 49916
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 104, no. 10, 2007, pages 3753 - 3758

Also Published As

Publication number Publication date
AU2023306869A1 (en) 2024-11-21
JP2025525290A (ja) 2025-08-05
CA3248185A1 (en) 2025-07-09
KR20250034312A (ko) 2025-03-11
EP4570807A1 (en) 2025-06-18
CN117402162A (zh) 2024-01-16
US20250304587A1 (en) 2025-10-02
TW202402273A (zh) 2024-01-16

Similar Documents

Publication Publication Date Title
CN113631557B (zh) Jak激酶抑制剂及其制备方法和在医药领域的应用
TWI616436B (zh) 作為magl抑制劑的胺基甲酸1,1,1-三氟-3-羥基丙烷-2-基酯衍生物及胺基甲酸1,1,1-三氟-4-羥基丁烷-2-基酯衍生物
CN109219604B (zh) 四氢异喹啉雌激素受体调节剂及其用途
TWI663166B (zh) 新化合物
CN106478651B (zh) 取代的杂芳基化合物及其组合物和用途
EP3766882B1 (en) Phthalazine isoxazole alkoxy derivatives, preparation method thereof, pharmaceutical composition and use thereof
EP3640247B1 (en) Syk inhibitor and use method therefor
CN109970743A (zh) 为jak抑制剂的5-氯-2-二氟甲氧基苯基吡唑并嘧啶化合物
US20240383924A1 (en) Benzopyrimidine compounds and use thereof
CN111936465A (zh) 作为共价menin抑制剂的哌啶化合物
JP2022526926A (ja) イミダゾロニルキノリン化合物およびそれらの治療への使用
WO2021164746A1 (zh) 取代芳基类化合物
CN116670123A (zh) 芳香杂环类化合物、药物组合物及其应用
CN117510503A (zh) 一种酪氨酸苏氨酸激酶pkmyt1抑制剂
CN116462687B (zh) Wee1抑制剂及其制备和用途
WO2024153247A1 (zh) 作为ctps1抑制剂的化合物
WO2024011883A1 (zh) Wee1抑制剂及其制备和用途
CN115151257B (zh) 用于治疗与dux4表达相关的疾病的新颖化合物
AU2022340743A1 (en) Mixed lineage kinase inhibitors and methods of use
WO2021219100A1 (zh) 一类含有稠合三环结构的化合物
HK40088311A (zh) Wee1抑制剂及其制备和用途
HK40088311B (zh) Wee1抑制剂及其制备和用途
WO2025036490A1 (zh) 一种DNA聚合酶θ抑制剂及其用途
CN119431366A (zh) Irak4降解剂化合物及其应用
WO2024140933A1 (zh) 喜树碱衍生物、药物组合物及其制备方法和应用

Legal Events

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

Ref document number: 23838385

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: AU2023306869

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2024568139

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 18867250

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2023306869

Country of ref document: AU

Date of ref document: 20230116

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2023838385

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2023838385

Country of ref document: EP

Effective date: 20250213

WWP Wipo information: published in national office

Ref document number: 1020247043576

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2023838385

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 18867250

Country of ref document: US