WO2023143501A1 - Composé inhibiteur de la voie wnt - Google Patents

Composé inhibiteur de la voie wnt Download PDF

Info

Publication number
WO2023143501A1
WO2023143501A1 PCT/CN2023/073533 CN2023073533W WO2023143501A1 WO 2023143501 A1 WO2023143501 A1 WO 2023143501A1 CN 2023073533 W CN2023073533 W CN 2023073533W WO 2023143501 A1 WO2023143501 A1 WO 2023143501A1
Authority
WO
WIPO (PCT)
Prior art keywords
halo
compound
alkylene
alkyl
reaction
Prior art date
Application number
PCT/CN2023/073533
Other languages
English (en)
Chinese (zh)
Inventor
陈宇锋
武朋
刘灿丰
李非凡
孙钊
杨寒
吕萌
程万里
金超凡
陈凯旋
陈可可
王友平
朱晓利
何南海
Original Assignee
杭州阿诺生物医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 杭州阿诺生物医药科技有限公司 filed Critical 杭州阿诺生物医药科技有限公司
Priority to CN202380009996.XA priority Critical patent/CN116964053A/zh
Publication of WO2023143501A1 publication Critical patent/WO2023143501A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D475/00Heterocyclic compounds containing pteridine ring systems
    • C07D475/02Heterocyclic compounds containing pteridine ring systems with an oxygen atom directly attached in position 4
    • C07D475/04Heterocyclic compounds containing pteridine ring systems with an oxygen atom directly attached in position 4 with a nitrogen atom directly attached in position 2

Definitions

  • the invention relates to a heterocyclic compound, in particular to a highly active Wnt pathway inhibitor and its application.
  • Wnt/ ⁇ -catenin signal transduction pathway is a pathway conserved in biological evolution.
  • ⁇ -catenin is only a cytoskeleton protein that forms a complex with E-cadherin at the cell membrane to maintain the adhesion of the same type of cells and prevent cell movement.
  • Wnt signaling pathway is not activated, ⁇ -catenin in the cytoplasm is phosphorylated, and forms a ⁇ -catenin degradation complex with APC, Axin, and GSK3 ⁇ , thereby initiating the ubiquitin system to degrade ⁇ -catenin through the proteasome pathway, so that ⁇ -catenin in the cytoplasm was maintained at a low level.
  • Wnt protein When cells are stimulated by Wnt signal, Wnt protein binds to the specific receptor Frizzled protein on the cell membrane, and the activated Frizzled receptor recruits intracellular Dishevelled protein, which inhibits the degradation activity of the ⁇ -catenin degradation complex formed by GSK3 ⁇ and other proteins, stabilizing Free ⁇ -catenin protein in the cytoplasm.
  • the stably accumulated ⁇ -catenin in the cytoplasm enters the nucleus and binds to the LEF/TCF transcription factor family to initiate the transcription of downstream target genes (such as c-myc, c-jun, Cyclin D1, etc.).
  • Wnt/ ⁇ -catenin signaling pathway is closely related to the occurrence of various cancers (including colon cancer, gastric cancer, breast cancer, etc.).
  • abnormal activation of Wnt classic signaling pathway and nuclear accumulation of ⁇ -catenin protein widely exist in colorectal cancer, and the proliferation of cancers such as colon cancer can be inhibited by inhibiting the activity of Wnt signaling pathway.
  • APC mutations exist in more than 85% of colorectal cancers, and the mutated APC blocks the phosphorylation and degradation of ⁇ -catenin and induces the occurrence of colorectal cancer.
  • mutations of Axin and ⁇ -catenin itself can also cause the intracellular accumulation of ⁇ -catenin and activate the Wnt/ ⁇ -catenin pathway.
  • the inventors unexpectedly found that the compound of the formula (I) structure of the present invention or its pharmaceutically acceptable salt, isotopic derivatives, and stereoisomers are effective Wnt pathway inhibitors, and have excellent Wnt pathway inhibitory activity.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt, isotopic derivative, or stereoisomer thereof:
  • R 1 and R 2 each independently represent hydrogen, (C 1 -C 6 ) alkyl, halogenated (C 1 -C 6 ) alkyl, (C 3 -C 8 ) cycloalkyl, halogenated (C 3 -C 8 )cycloalkyl, 4-8 membered heterocycloalkyl, halogenated 4-8 membered heterocycloalkyl, -(C 1 -C 6 )alkylene OR a , -halogenated (C 1 - C 6 ) alkylene OR a , -(C 1 -C 6 ) alkylene SR a , -halogenated (C 1 -C 6 ) alkylene SR a , or R 1 , R 2 are connected to them
  • the carbon atoms together form a 3-8 membered ring which may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O and S;
  • R 3 represents (C 1 -C 6 ) alkyl, halo (C 1 -C 6 ) alkyl, (C 3 -C 8 ) cycloalkyl, halo (C 3 -C 8 ) cycloalkyl, or R 3 and R 1 or R 2 together form a 4-7 membered ring, which may optionally contain 0 or 1 heteroatoms selected from O and S;
  • X means CR 4 or N
  • R 4 each independently represent hydrogen, halogen, cyano, (C 1 -C 3 ) alkyl, halo (C 1 -C 3 ) alkyl;
  • L represents -O- or -(CR m R m ')-, wherein R m and R m ' each independently represent hydrogen, (C 1 -C 3 ) alkyl, (C 3 -C 8 ) cycloalkyl , or R m , R m 'and the carbon atom connected to it form a 3-5 membered ring, and the ring may contain 0 or 1 heteroatom selected from O and S;
  • Cy represents a 5-12 membered aromatic heterocycle, which optionally contains 1, 2, 3 or 4 heteroatoms, each of which is independently selected from N, O, S;
  • R 5 represents halogen, (C 1 -C 6 ) alkyl, halo (C 1 -C 6 ) alkyl, (C 3 -C 8 ) cycloalkyl, halo (C 3 -C 8 ) cycloalkyl , -OR a , -halogenated OR a , -SR a , -halogenated SR a ;
  • R 6 represents halogen, (C 1 -C 6 ) alkyl, halo (C 1 -C 6 ) alkyl, (C 3 -C 8 ) cycloalkyl, halo (C 3 -C 8 ) cycloalkane radical, (C 3 -C 8 )heterocycloalkyl, halo(C 3 -C 8 )heterocycloalkyl, (C 2 -C 6 )alkynyl, halo(C 2 -C 6 )alkynyl, -(C 1 -C 6 )alkylene OR a , -halogenated (C 1 -C 6 )alkylene OR a , -(C 3 -C 8 )cycloalkylene OR a , -halogenated (C 3 -C 8 )cycloalkylene OR a , -(C 1 -C 6 )alkylene SR a , -halogen
  • R 7 each independently represent hydrogen, halogen, (C 1 -C 3 ) alkyl, halo (C 1 -C 3 ) alkyl;
  • n and n independently represent 0, 1 or 2;
  • R a and R a ' each independently represent hydrogen, (C 1 -C 6 ) alkyl, halo (C 1 -C 6 ) alkyl, (C 3 -C 8 ) cycloalkyl, halo (C 3 -C 8 ) cycloalkyl.
  • R 6 when R 6 is attached to a carbon atom, R 6 represents halogen, (C 1 -C 6 ) alkyl, halo (C 1 -C 6 ) alkyl, (C 3 -C 8 ) cycloalkyl, Halo(C 3 -C 8 )cycloalkyl, (C 3 -C 8 )heterocycloalkyl, halo(C 3 -C 8 )heterocycloalkyl, (C 2 -C 6 )alkynyl, halo Substituted (C 2 -C 6 )alkynyl, -(C 1 -C 6 )alkylene OR a , -halogenated (C 1 -C 6 )alkylene OR a , -(C 3 -C 8 )alkylene OR a , -(C 3 -C 8 )alkylene Cycloalkyl OR a , -Halo(C 3 -
  • R 6 represents halogen, (C 1 -C 6 ) alkyl, halo (C 1 -C 6 ) alkyl, (C 3 -C 8 ) cycloalkyl, halo ( C 3 -C 8 )cycloalkyl, (C 3 -C 8 )heterocycloalkyl, halo(C 3 -C 8 )heterocycloalkyl, (C 2 -C 6 )alkynyl, halo(C 2 -C 6 )alkynyl, -(C 1 -C 6 )alkylene OR a , -halo(C 1 -C 6 )alkylene OR a , -(C 3 -C 8 )cycloalkylene OR a , -halogenated (C 3 -C 8 )cycloalkylene OR a , -(C 1 -C 6 )alkylene SR
  • R 6 represents halogen, (C 1 -C 6 ) alkyl, halo (C 1 -C 6 ) alkyl, (C 3 -C 8 ) cycloalkyl, halo (C 3 -C 8 ) cycloalkyl.
  • R 1 and R 2 each independently represent hydrogen, (C 1 -C 6 ) alkyl, halogenated (C 1 -C 6 ) alkyl, (C 3 -C 8 ) cycloalkane group, halo(C 3 -C 8 )cycloalkyl, -(C 1 -C 6 )alkylene OR a , -(C 1 -C 6 )alkylene SR a ; or R 1 , R 2 and Together with the carbon atoms to which they are attached, they form a 3-8 membered ring which may optionally contain 0, 1, 2 or 3 heteroatoms selected from N, O and S.
  • R 3 is C 1 -C 6 )alkyl or halo(C 1 -C 6 )alkyl.
  • R 3 is (C 1 -C 6 )alkyl.
  • Cy is a 5-6 membered monocyclic aromatic heterocycle, or Cy is a 9-10 membered bicyclic aromatic heterocycle.
  • Cy is pyridyl, pyrimidinyl, pyrazolyl, imidazolyl or pyrrolyl.
  • R6 and L are connected at the ortho position of Cy.
  • R 5 is located at the meta position of R 6 , and is located at the meta or para position of L.
  • X is CR 4 .
  • R4 is hydrogen
  • connection of Cy to R 5 and R 6 is as follows:
  • connection of Cy to R 5 and R 6 is as follows:
  • R is selected from:
  • the present invention also provides a class of compounds or pharmaceutically acceptable salts, isotopic derivatives, and stereoisomers thereof, wherein the compounds have the following structure:
  • the present invention also provides a pharmaceutical composition, which comprises the aforementioned compound or a pharmaceutically acceptable salt, isotope derivative, stereoisomer, and optionally a pharmaceutically acceptable carrier.
  • the present invention also provides the aforementioned compounds or their pharmaceutically acceptable salts, isotope derivatives, stereoisomers, or the aforementioned pharmaceutical compositions used in the prevention and/or treatment of cancer, tumors, and inflammatory diseases , use in medicines for autoimmune diseases or immune-mediated diseases.
  • the pharmaceutically acceptable salts of the present invention may be formed using, for example, the following inorganic or organic acids:
  • “Pharmaceutically acceptable salt” means a salt which, within the scope of reasonable medical judgment, is suitable for use in contact with humans and lower other animals tissue, without undue toxicity, irritation, allergic reaction, etc., can be called a reasonable benefit/risk ratio.
  • the salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or alone by reacting the free base or acid with a suitable reagent, as outlined below. For example, a free base function can be reacted with a suitable acid.
  • inorganic acid addition salts are amino acids with inorganic acids (e.g., hydrochloric, hydrobromic, phosphoric, sulfuric, and perchloric) or organic acids (e.g., acetic, oxalic, maleic, tartaric, lemon acid, succinic acid or malonic acid), or by using other methods known in the art such as ion exchange.
  • inorganic acids e.g., hydrochloric, hydrobromic, phosphoric, sulfuric, and perchloric
  • organic acids e.g., acetic, oxalic, maleic, tartaric, lemon acid, succinic acid or malonic acid
  • salts include adipate, sodium alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, Camphorsulfonate, Citrate, Cyclopentanepropionate, Digluconate, Lauryl Sulfate, Ethylate, Formate, Fumarate, Glucoheptonate, Glycerin Phosphate, gluconate, hernisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate Salt, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectate Salt, persulf
  • alkali or alkaline earth metal salts include those of sodium, lithium, potassium, calcium, magnesium, and the like.
  • Other pharmaceutically acceptable salts include, where appropriate, nontoxic ammonium salts, quaternary ammonium salts, and amine cations formed with counterions, for example, halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, lower Alkylsulfonates and arylsulfonates.
  • the pharmaceutically acceptable salts of the present invention can be prepared by conventional methods, for example, by dissolving the compound of the present invention in a water-miscible organic solvent (such as acetone, methanol, ethanol and acetonitrile), adding an excess of organic acid or inorganic Aqueous acid solution, so that the salt is precipitated from the resulting mixture, the solvent and remaining free acid are removed therefrom, and the precipitated salt is isolated.
  • a water-miscible organic solvent such as acetone, methanol, ethanol and acetonitrile
  • the precursors or metabolites described in the present invention may be precursors or metabolites known in the art, as long as the precursors or metabolites are transformed into compounds through in vivo metabolism.
  • prodrugs refer to those prodrugs of the compounds of the present invention which, within the scope of sound medical judgment, are suitable for use in contact with human and lower animal tissues without undue toxicity, irritation, allergic response, etc., Qualified as having a reasonable benefit/risk ratio and valid for its intended use.
  • prodrug refers to a compound that is rapidly transformed in vivo to yield the parent compound of the above formula, for example by in vivo metabolism, or N-demethylation of a compound of the invention.
  • Solvate as used herein means a physical association of a compound of the present invention with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In some cases, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid, solvates will be able to be isolated. Solvent molecules in solvates may exist in regular and/or disordered arrangements. Solvates may contain stoichiometric or non-stoichiometric amounts of solvent molecules. "Solvate” encompasses both solution-phase and isolatable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Solvation methods are well known in the art.
  • Steps of “Stereoisomerism” described in the present invention is divided into conformational isomerism and configurational isomerism, and configurational isomerism can also be divided into cis-trans isomerism and optical isomerism (i.e. optical isomerism), conformational isomerism refers to having Due to the rotation or twisting of carbon and carbon single bonds in organic molecules of a certain configuration, a stereoisomerism phenomenon in which each atom or atomic group of the molecule has a different arrangement in space, the common structures are alkanes and cycloalkanes. Such as the chair conformation and boat conformation that appear in the structure of cyclohexane.
  • Stepoisomer means when a compound of the present invention contains one or more asymmetric centers and is thus available as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and single Diastereomers.
  • the compound of the present invention has an asymmetric center, and each asymmetric center can produce two optical isomers, and the scope of the present invention includes all possible optical isomers and diastereoisomer mixtures and pure or partially pure compounds .
  • the compounds described herein may exist in tautomeric forms having different points of attachment of hydrogens by displacement of one or more double bonds. For example, a ketone and its enol form are keto-enol tautomers.
  • An “isotopic derivative” of the present invention refers to an isotopically labeled molecule of a compound herein.
  • Isotopes commonly used for isotope labeling are: Hydrogen isotopes, 2 H and 3 H; Carbon isotopes: 11 C, 13 C and 14 C; Chlorine isotopes: 35 Cl and 37 Cl; Fluorine isotopes: 18 F; Iodine isotopes: 123 I and 125 I; nitrogen isotopes: 13 N and 15 N; oxygen isotopes: 15 O, 17 O and 18 O and sulfur isotope 35 S.
  • These isotope-labeled compounds can be used to study the distribution of pharmaceutical molecules in tissues.
  • deuterium 3 H and carbon 13 C are more widely used because of their easy labeling and convenient detection.
  • substitution of some heavy isotopes, such as deuterium ( 2 H) can enhance the stability of metabolism, prolong the half-life and thus achieve the purpose of reducing the dose and provide therapeutic advantages.
  • Isotopically labeled compounds generally start from labeled starting materials using known synthetic techniques such as the synthesis of non-isotopic Labeled compounds to complete their synthesis.
  • the present invention also provides the use of the compound of the present invention in the preparation of medicaments for preventing and/or treating cancer, tumor, inflammatory disease, autoimmune disease or immune-mediated disease.
  • the present invention provides a pharmaceutical composition for preventing and/or treating cancer, tumor, inflammatory disease, autoimmune disease, neurodegenerative disease, attention-related disease or immune-mediated disease, which comprises the present invention compounds as active ingredients.
  • the pharmaceutical composition may optionally comprise a pharmaceutically acceptable carrier.
  • the present invention provides a method of preventing and/or treating cancer, tumor, inflammatory disease, autoimmune disease, neurodegenerative disease, attention-related disease or immune-mediated disease, which includes the need for
  • the mammal of the present invention is administered the compound of formula (I) or its pharmaceutically acceptable salt, isotope derivative, stereoisomer or the pharmaceutical composition of the present invention.
  • inflammatory, autoimmune, and immune-mediated diseases may include, but are not limited to, arthritis, rheumatoid arthritis, spondyloarthritis, gouty arthritis, osteoarthritis, juvenile arthritis , Other Arthritis Conditions, Lupus, Systemic Lupus Erythematosus (SLE), Skin Related Disorders, Psoriasis, Eczema, Dermatitis, Atopic Dermatitis, Pain, Pulmonary Disease, Lung Inflammation, Adult Respiratory Distress Syndrome (ARDS) , pulmonary sarcoidosis, chronic pulmonary inflammatory disease, chronic obstructive pulmonary disease (COPD), cardiovascular disease, atherosclerosis, myocardial infarction, congestive heart failure, myocardial ischemia-reperfusion injury, inflammatory bowel disease, Crohn's disease, ulcerative colitis, irritable bowel syndrome, asthma, Sjogren's syndrome, autoimmune thyroid disease, urticaria (rubella), multiple sclerosis, s
  • cancer or tumor may include, but are not limited to, skin cancer, bladder cancer, ovarian cancer, breast cancer, stomach cancer, pancreatic cancer, prostate cancer, colon cancer, lung cancer, bone cancer, brain cancer, neuroblastoma, rectal cancer , colon cancer, familial adenomatous polyposis carcinoma, hereditary nonpolyposis colorectal cancer, esophageal cancer, lip Carcinoma, laryngeal cancer, hypopharyngeal cancer, tongue cancer, salivary gland cancer, gastric cancer, adenocarcinoma, medullary thyroid cancer, papillary thyroid cancer, kidney cancer, renal parenchymal cancer, ovarian cancer, cervical cancer, uterine body cancer, endometrium Carcinoma, choriocarcinoma, pancreatic cancer, prostate cancer, testicular cancer, urinary cancer, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma
  • the compound of the present invention or a pharmaceutically acceptable salt thereof when administered in combination with another anticancer agent or immune checkpoint inhibitor for the treatment of cancer or tumors, the compound of the present invention or a pharmaceutically acceptable salt thereof can provide enhanced anticancer effects .
  • anticancer agents useful in the treatment of cancer or tumors may include, but are not limited to, inhibitors of cell signaling, chlorambucil, guanfalan, cyclophosphamide, ifosfamide, busulfan, carbamate, Mustin, lomustine, streptozotocin, cisplatin, carboplatin, oxaliplatin, dacarbazine, temozolomide, procarbazine, methotrexate, fluorouracil, cytarabine, gemcitabine, Mercaptopurine, fludarabine, vinblastine, vincristine, vinorelbine, paclitaxel, docetaxel, topotecan, irinotecan, etoposide, trabectedin, dactinomycin, doxorubicin , epirubicin, daunorubicin, mitoxantrone, bleomycin, mitomycin C, ixabepilone
  • therapeutic agents useful in the treatment of inflammatory, autoimmune, and immune-mediated diseases can include, but are not limited to, steroidal drugs (e.g., prednisone, prednisone, prednisone, methylphenidate, Cortisone, cortisone, hydroxycortisone, betamethasone, dexamethasone, etc.), methotrexate, leflunomide, anti-TNF ⁇ agents (eg, etanercept, infliximab, adalib monoclonal antibody, etc.), calcineurin inhibitors (eg, tacrolimus, piguanolimus, etc.), and antihistamines (eg, diphenhydramine, hydroxyzine, loratadine, ebazan Tin, ketotifen, cetirizine, levocetirizine, fexofenadine, etc.), and at least one or more therapeutic agents selected from them can be included in the pharmaceutical composition of the present invention
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can be administered as an active ingredient.
  • the dose of the active ingredient can be adjusted according to a number of relevant factors such as the condition of the subject to be treated, the type and severity of the disease, the rate of administration and the opinion of the physician. In some cases, amounts less than the above dosages may be appropriate. Amounts greater than the above doses may be used if no deleterious side effects are caused and such amounts may be administered in divided doses daily.
  • the present invention also provides a method for preventing and/or treating tumors, cancers, viral infections, organ transplant rejection, neurodegenerative diseases, attention-related diseases or autoimmune diseases, which comprises the following A compound of the invention or a compound or pharmaceutical composition of the invention is administered to a mammal in need thereof.
  • compositions of the present invention can be formulated into dosage forms for oral administration or parenteral administration (including intramuscular, intravenous and subcutaneous routes, intratumoral injection) according to any of conventional methods, such as tablets, granules, powders , capsules, syrups, emulsions, microemulsions, solutions or suspensions.
  • compositions of the present invention for oral administration can be prepared by mixing the active ingredient with carriers such as cellulose, calcium silicate, corn starch, lactose, sucrose, dextrose, calcium phosphate, stearic acid, stearic acid, Magnesium stearate, calcium stearate, gelatin, talc, surfactant, suspending agent, emulsifier and diluent.
  • carriers such as cellulose, calcium silicate, corn starch, lactose, sucrose, dextrose, calcium phosphate, stearic acid, stearic acid, Magnesium stearate, calcium stearate, gelatin, talc, surfactant, suspending agent, emulsifier and diluent.
  • Examples of carriers employed in the pharmaceutical composition for injection administration of the present invention may be water, saline solution, glucose solution, glucose-like solution, alcohol, glycol, ether (for example, polyethylene glycol 400 ), oils, fatty acids, fatty acid esters, glycerides, surfactants, suspending agents and emulsifiers.
  • the compounds of the present invention can be prepared in a variety of ways known to those skilled in the art of organic synthesis, using the methods described below as well as synthetic methods known in the art of synthetic organic chemistry or by variations thereof known to those skilled in the art Synthesis of compounds of the invention. Preferred methods include, but are not limited to, those described below. Reactions are performed in solvents or solvent mixtures appropriate to the kit materials used and to the transformations effected. Those skilled in the art of organic synthesis will appreciate that the functionality present on the molecule is consistent with the proposed transitions. This sometimes requires judgment to alter the order of synthetic steps or starting materials to obtain the desired compound of the invention.
  • Figure 1 shows the effect of compound 1 on the tumor volume of human colon cancer cell Colo205 xenograft tumor in nude mice.
  • a given chemical formula or name shall encompass all stereoisomers and optical isomers thereof and racemates in which such isomers exist. Unless otherwise indicated, all chiral (enantiomers and diastereoisomers) and racemic forms are within the scope of the invention.
  • the present invention describes cis- and trans- (or E- and Z-) geometric isomers of the compounds of the invention and which may be isolated as a mixture of isomers or as separated isomeric forms.
  • the compounds of the invention may be isolated in optically active or racemic forms.
  • the compounds of the invention may exist in various tautomeric forms in which the hydrogen atoms are transposed to other parts of the molecule and thus the chemical bonds between the atoms of the molecule are rearranged. It is to be understood that all tautomeric forms which may exist are included within the present invention.
  • substituents in the present invention are independent and not interrelated, for example (listed but not exhaustive), in one aspect, for R a (or R a ') in the substituent, its are independent of each other in the definitions of the different substituents. Specifically, when one definition is selected for R a (or R a ') in one substituent, it does not mean that the R a (or R a ') has the same definition in other substituents.
  • R a when the definition of R a (or R a ') is selected from hydrogen, it does not mean that in -C(O)-NR In a R a ', R a (or R a ') must be hydrogen.
  • R a (or R a ') when there is more than one R a (or R a ') in a certain substituent, these R a (or R a ') are also independently independent.
  • substituent is selected from, for example, the following substituents, such as alkyl, cycloalkyl, aryl, heterocyclyl, halogen, hydroxy, Alkoxy, oxo, alkanoyl, aryloxy, alkanoyloxy, amino, alkylamino, arylamino, arylalkylamino, disubstituted amino (wherein 2 amino substituents are selected from alkyl radical, aryl or arylalkyl), alkanoylamino, aroylamino, aralkanoylamino, substituted alkanoylamino, substituted arylamino, substituted aralkanoylamino, thio, alkylthio , arylthio, arylalkylthio, arylthiocarbonyl, arylalkylthiocarbonyl, alkylsul
  • substituents such as alkyl, cycloalkyl,
  • carbamoyl such as -CONH 2
  • substituted carbamoyl such as -CONHalkyl, -CONHaryl, -CONHarylalkyl or on nitrogen
  • heterocyclic group such as indolyl, imidazolyl, furan base, thienyl, thiazolyl, pyrrolidinyl, pyridyl, Pyrimidinyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, homopiperazinyl, etc. and substituted heterocyclic groups.
  • alkyl or "alkylene” as used herein is intended to include both branched and straight chain saturated aliphatic hydrocarbon groups having the indicated number of carbon atoms.
  • C 1 -C 6 alkyl means an alkyl group having 1 to 6 carbon atoms.
  • alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (such as n-propyl and isopropyl), butyl (such as n-butyl, isobutyl, t-butyl), and Pentyl (eg n-pentyl, isopentyl, neopentyl).
  • the alkyl group is preferably an alkyl group having 1 to 6, 1 to 4, more preferably 1 to 3 carbon atoms.
  • alkenyl denotes a straight or branched chain hydrocarbon group containing one or more double bonds and generally having a length of 2 to 20 carbon atoms.
  • C2-C6 alkenyl contains two to six carbon atoms.
  • Alkenyl groups include, but are not limited to, eg vinyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, and the like.
  • alkynyl denotes a straight or branched chain hydrocarbon group containing one or more triple bonds and generally having a length of 2 to 20 carbon atoms.
  • C2 - C6 alkynyl contains two to six carbon atoms.
  • Representative alkynyl groups include, but are not limited to, for example, ethynyl, 1-propynyl, 1-butynyl, and the like.
  • alkoxy refers to -O-alkyl.
  • C 1 -C 6 alkoxy (or alkyloxy) is intended to include C 1 , C 2 , C 3 , C 4 , C 5 , C 6 alkoxy.
  • alkoxy include, but are not limited to, methoxy, ethoxy, propoxy (eg, n-propoxy and isopropoxy), and tert-butoxy.
  • the alkoxy group is preferably an alkoxy group having 1 to 6, more preferably 1 to 4 carbon atoms.
  • alkylthio or “thiothio” denotes an alkyl group as defined above having the indicated number of carbon atoms attached through a sulfur bridge; eg methyl-S- and ethyl-S-.
  • aryl alone or as part of a larger moiety such as “aralkyl”, “arylalkoxy” or “aryloxyalkyl”, refers to a single group having a total of 5 to 12 ring members Cyclic, bicyclic or tricyclic ring systems, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members.
  • aryl refers to an aromatic ring system which includes, but is not limited to, phenyl, biphenyl, indanyl, 1-naphthyl, 2-naphthyl, and tetrahydronaphthalene base.
  • aralkyl or "arylalkyl” refers to an alkyl residue attached to an aryl ring, non-limiting examples of which include benzyl, phenethyl, and the like.
  • a fused aryl group can be attached to another group at a suitable position on the cycloalkyl ring or aromatic ring. Dashed lines drawn from ring systems indicate that bonds may be attached to any suitable ring atom.
  • cycloalkyl refers to a monocyclic or bicyclic cyclic alkyl group.
  • Monocyclic cyclic alkyl refers to C3-C8 cyclic alkyl, including but not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and norbornyl.
  • Branched cycloalkyl groups such as 1-methylcyclopropyl and 2-methylcyclopropyl are included within the definition of "cycloalkyl”.
  • bicyclic cyclic alkyl group Cycloalkyl groups including bridged rings, spiro rings or fused rings.
  • the cycloalkyl group is preferably a cycloalkyl group having 3 to 8, more preferably 3 to 6 carbon atoms.
  • cycloalkenyl refers to a monocyclic or bicyclic cyclic alkenyl group.
  • Monocyclic cyclic alkenyl refers to C 3 -C 8 cyclic alkenyl, including but not limited to cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl and norbornenyl.
  • Branched cycloalkenyl groups such as 1-methylcyclopropenyl and 2-methylcyclopropenyl are included within the definition of "cycloalkenyl”.
  • Bicyclic cyclic alkenyl groups include bridged, spiro, or fused cyclic alkenyl groups.
  • Halo or halogen includes fluoro, chloro, bromo and iodo.
  • Haloalkyl is intended to include branched and straight chain saturated aliphatic hydrocarbon groups having the indicated number of carbon atoms substituted with one or more halogens. Examples of haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, pentachloroethyl, 2,2,2-trifluoroethyl, heptafluoro Propyl and Heptachloropropyl.
  • haloalkyl also include "fluoroalkyl” intended to include branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms and substituted with 1 or more fluorine atoms.
  • fluoroalkyl intended to include branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms and substituted with 1 or more fluorine atoms.
  • halocycloalkyl is intended to include branched cycloalkyl groups having the indicated number of carbon atoms substituted with one or more halogens.
  • Haloalkoxy or "haloalkyloxy” means a haloalkyl group as defined above having the indicated number of carbon atoms attached through an oxygen bridge.
  • haloC 1 -C 6 alkoxy is intended to include C 1 , C 2 , C 3 , C 4 , C 5 , C 6 haloalkoxy.
  • haloalkoxy include, but are not limited to, trifluoromethoxy, 2,2,2-trifluoroethoxy, and pentafluoroethoxy.
  • haloalkylthio or “thiohaloalkoxy” denotes a haloalkyl group as defined above having the indicated number of carbon atoms attached through a sulfur bridge; for example trifluoromethyl-S- and pentafluoroethyl -S-.
  • -halo(C 1 -C 6 )alkylene OR a , -halo(C 1 -C 6 )alkylene SR a , -halo(C 1 -C 6 )alkylene NR a R a ′, -Halo(C 1 -C 6 )alkylene CN, -Halo(C 3 -C 8 )cycloalkylene OR a , -Halo(C 3 -C 8 )cycloalkane
  • the group SR a , -halogenated (C 3 -C 8 )cycloalkylene NR a R a ′, -halogenated (C 3 -C 8 )cycloalkylene CN, etc.
  • C x1 -C x2 is used when referring to some substituent groups, which means that the number of carbon atoms in the substituent groups may be from x 1 to x 2 .
  • C 0 -C 8 means that the group contains 0, 1, 2, 3, 4, 5, 6, 7 or 8 carbon atoms
  • C 1 -C 8 means that the group contains 1, 2, 3 , 4, 5, 6, 7 or 8 carbon atoms
  • C 2 -C 8 means that the group contains 2, 3, 4, 5, 6, 7 or 8 carbon atoms
  • C 3 -C 8 means that the The group contains 3, 4, 5, 6, 7 or 8 carbon atoms
  • C 4 -C 8 means that the group contains 4, 5, 6, 7 or 8 carbon atoms
  • C 0 -C 6 means that the group contains 0, 1, 2, 3, 4, 5 or 6 carbon atoms
  • C 1 -C 6 means that the group contains 1, 2, 3, 4, 5 or 6 carbon atoms
  • C 2 -C 6 means that the group contains 2, 3, 4, 5 or 6 carbon atoms
  • x 1 -x 2 -membered ring is used when referring to cyclic groups (such as aryl, heteroaryl, cycloalkyl and heterocycloalkyl), which means that the group's The number of ring atoms may be x1 to x2 .
  • the 3-12 membered cyclic group may be a 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 membered ring, and the number of ring atoms may be 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12; 3-6-membered ring means that the cyclic group can be 3, 4, 5 or 6-membered ring, and the number of ring atoms can be 3, 4, 5 or 6 ; 3-8 membered ring means that the cyclic group can be 3, 4, 5, 6, 7 or 8-membered ring, and the number of ring atoms can be 3, 4, 5, 6, 7 or 8; 3-9 A membered ring means that the cyclic group can be a 3, 4, 5, 6, 7, 8 or 9-membered ring, and the number of ring atoms can be 3, 4, 5, 6, 7, 8 or 9; 4-7 A membered ring means that the cyclic group can be a 4, 5, 6 or 7-membered ring, and the number of ring atoms can be 4, 5, 6 or 7;
  • the ring atoms may be carbon atoms or heteroatoms, for example selected from N, O and S.
  • the heterocyclic ring may contain 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more ring heteroatoms, for example selected from N, O and S of heteroatoms.
  • the one or more halogens may each be independently selected from fluorine, chlorine, bromine and iodine.
  • heteroaryl means a specified number of stable monocyclic or polycyclic aromatic groups containing heteroatoms, preferably 3-membered, 4-membered, 5-membered, 6-membered, or 7-membered aromatic monocyclic or aromatic bicyclic Or 7-membered, 8-membered, 9-membered, 10-membered, 11-membered, 12-membered aromatic polycyclic heterocycles, which are fully unsaturated or partially unsaturated, and which contain carbon atoms and 1, 2, 3 or 4 heteroatoms independently selected from N, O and S; and include any of the following polycyclic groups, wherein any heterocycle defined above is fused to a benzene ring.
  • the heteroaryl group herein is preferably a 5- to 12-membered heteroaryl group. Nitrogen and sulfur heteroatoms can be optionally oxidized. The nitrogen atom is substituted or unsubstituted (ie N or NR, where R is H or another substituent if defined). A heterocycle can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. The heterocyclyl groups described herein may be substituted on carbon or nitrogen atoms if the resulting compound is stable. The nitrogen in the heterocycle can optionally be quaternized. Preferably, when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to each other.
  • heterocycle is intended to include heteroaryl.
  • heteroaryl groups include, but are not limited to, acridinyl, azetidinyl, aziocinyl, benzimidazolyl, benzofuryl, benzothiofuranyl, benzothienyl, benzooxa Azolyl, benzoxazolyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolyl, 2H, 6H-1,5,2-dithiazinyl, dihydrofuro[2, 3-b] Tetrahydrofu
  • heteroaryl may also include biaryl structures formed by the above-defined “aryl” and a monocyclic “heteroaryl”, such as but not limited to "-phenylbipyridyl-", “- "Phenyl bipyrimidyl”, “-pyridyl biphenyl”, “-pyridyl bipyrimidyl-”, “-pyrimidyl biphenyl-”; wherein the present invention also includes condensed rings containing, for example, the above-mentioned heterocycles and Spiro compound.
  • heterocycloalkyl refers to a monocyclic heterocycloalkyl system, or a bicyclic heterocycloalkyl system, and also includes spiroheterocycle or bridged heterocycloalkyl.
  • monocyclic heterocycloalkyl means It is a 3-8 membered, saturated or unsaturated but not aromatic cyclic alkyl system containing at least one heteroatom selected from O, N, S and P.
  • the bicyclic heterocycloalkyl system refers to a heterocycloalkyl fused to a phenyl group, or a cycloalkyl group, or a cycloalkenyl group, or a heterocycloalkyl group, or a heteroaryl group formed two-ring system.
  • bridged cycloalkyl refers to polycyclic compounds sharing two or more carbon atoms. Can be divided into bicyclic bridged ring hydrocarbons and polycyclic bridged ring hydrocarbons. The former is composed of two alicyclic rings sharing more than two carbon atoms; the latter is a bridged ring hydrocarbon composed of more than three rings.
  • spirocycloalkyl refers to polycyclic hydrocarbons in which monocyclic rings share one carbon atom (called a spiro atom).
  • bridged ring heterogroup refers to a polycyclic compound sharing two or more atoms, and the ring contains at least one heteroatom selected from O, N and S atoms. It can be divided into bicyclic bridged heterocycles and polycyclic bridged heterocycles.
  • heterospirocyclyl refers to a polycyclic hydrocarbon that shares one carbon atom (called a spiro atom) between monocyclic rings, and the ring contains at least one heteroatom selected from O, N and S atoms.
  • substituted means that at least one hydrogen atom is replaced by a non-hydrogen group, provided that normal valences are maintained and that the substitution results in a stable compound.
  • nitrogen atoms e.g. amines
  • these nitrogen atoms can be converted to N-oxides by treatment with oxidizing agents (e.g. mCPBA and/or hydrogen peroxide) to obtain other compounds of the invention .
  • oxidizing agents e.g. mCPBA and/or hydrogen peroxide
  • both shown and claimed nitrogen atoms are considered to cover both the shown nitrogen and its N-oxides to obtain the derivatives of the present invention.
  • any variable occurs more than one time in any composition or formula of a compound, its definition on each occurrence is independent of its definition at every other occurrence.
  • a group is shown to be substituted with 0-3 R, then said group may be optionally substituted with up to three R groups, and R at each occurrence is independently selected from the definition of R.
  • combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • patient refers to an organism being treated by the methods of the present invention.
  • organisms preferably include, but are not limited to, mammals (eg, murine, ape, monkey, equine, bovine, porcine, canine, feline, etc.) and most preferably refer to humans.
  • the term "effective amount” means an amount that will elicit, for example, the amount sought by a researcher or clinician.
  • therapeutically effective amount means an amount which results in improved treatment, cure, prevention or alleviation of a disease, disorder or side effect, or a reduction in the or the rate of disease progression.
  • An effective amount may be given in one or more administrations, applications or doses and is not intended to be limited by a particular formulation or route of administration. The term also includes within its scope amounts effective to enhance normal physiological function.
  • treating includes any effect that results in amelioration of a condition, disease, disorder, etc., such as alleviation, reduction, regulation, amelioration or elimination, or amelioration of the symptoms thereof.
  • pharmaceutically acceptable refers to those compounds, substances, compositions and/or dosage forms: within the scope of sound medical judgment, they are suitable for use in contact with human and animal tissues without excessive toxicity, irritation sex, allergic reactions, and/or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier means a pharmaceutical substance, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g. lubricant, talc, magnesium stearate, calcium stearate or zinc stearate or stearic acid) or solvent-encapsulated substances involved in the carrying or transport of a subject compound from one organ or body part to another.
  • manufacturing aid e.g. lubricant, talc, magnesium stearate, calcium stearate or zinc stearate or stearic acid
  • solvent-encapsulated substances involved in the carrying or transport of a subject compound from one organ or body part to another.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • composition means a composition comprising a compound of the present invention together with at least one other pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier means a medium generally accepted in the art for the delivery of biologically active agents to animals, particularly mammals, including (i.e.) adjuvants, excipients or vehicles, such as diluents, preservatives , fillers, flow regulators, disintegrants, wetting agents, emulsifiers, suspending agents, sweeteners, flavoring agents, fragrances, antibacterial agents, antifungal agents, lubricants and dispersants, depending on Mode of administration and nature of dosage form.
  • acceptable means that a formulation ingredient or active ingredient does not have an undue adverse effect on health for the general purpose of treatment.
  • cancer refers to an abnormal growth of cells that cannot be controlled and, under certain conditions, is capable of metastasizing (spreading). Cancers of this type include, but are not limited to, solid tumors (eg, bladder, bowel, brain, chest, uterus, heart, kidney, lung, lymphoid tissue (lymphoma), ovary, pancreas or other endocrine organs (eg, thyroid), prostate , skin (melanoma), or blood cancer (such as non-leukemic leukemia).
  • solid tumors eg, bladder, bowel, brain, chest, uterus, heart, kidney, lung, lymphoid tissue (lymphoma), ovary, pancreas or other endocrine organs (eg, thyroid), prostate , skin (melanoma), or blood cancer (such as non-leukemic leukemia).
  • administration in combination refers to the administration of several selected therapeutic agents to a patient, in the same or different modes of administration at the same or different times.
  • enhancing or “capable of enhancing”, as used herein, means that the desired result can be enhanced in potency or sustained time has been increased or extended.
  • capable of potentiating refers to the ability of the drug to increase or prolong its potency or duration in the system.
  • potency value refers to the ability to maximize the enhancement of another therapeutic drug in an ideal system.
  • immune disease refers to a disease or condition of an adverse or deleterious reaction to an endogenous or exogenous antigen.
  • the result is usually dysfunction of the cells, or destruction thereof and dysfunction, or destruction of organs or tissues that may produce immune symptoms.
  • subject or “patient” includes mammals and non-mammals.
  • Mammals include, but are not limited to, mammals: humans, non-human primates such as orangutans, apes, and monkeys; agricultural animals such as cattle, horses, goats, sheep, and pigs; domestic animals such as rabbits and dogs; experimental animals include rodents, Such as rats, mice and guinea pigs.
  • Non-mammalian animals include, but are not limited to, birds, fish, and the like.
  • the selected mammal is a human.
  • treatment includes alleviating, suppressing or ameliorating the symptoms or conditions of a disease; inhibiting the development of complications; ameliorating or preventing the underlying metabolic syndrome; inhibiting the development of diseases or symptoms, Such as controlling the development of a disease or condition; alleviating a disease or a symptom; causing a disease or a symptom to regress; alleviating a complication caused by a disease or a symptom, or preventing and/or treating a sign caused by a disease or a symptom.
  • a certain compound or pharmaceutical composition after administration, can improve a certain disease, symptom or situation, especially improve its severity, delay the onset, slow down the progression of the disease, or reduce the duration of the disease. Circumstances that may be attributable to or related to the administration, whether fixed or episodic, continuous or intermittent.
  • Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ocular, pulmonary, transdermal, vaginal, ear canal , nasal administration and topical administration.
  • parenteral administration includes intramuscular injection, subcutaneous injection, intravenous injection, intramedullary injection, intraventricular injection, intraperitoneal injection, intralymphatic injection, and intranasal injection.
  • the compounds described herein are administered locally rather than systemically.
  • the depot formulation is administered by implantation (eg, subcutaneously or intramuscularly) or by intramuscular injection.
  • the drug is administered via a targeted drug delivery system.
  • liposomes coated with organ-specific antibodies are selectively directed to and taken up by specific organs.
  • the raw materials and reagents used in the present invention are known products, which can be synthesized according to methods known in the art, or can be obtained by purchasing commercially available products. All commercially available reagents were used without further purification.
  • Room temperature means 20-30°C.
  • the nitrogen atmosphere refers to a nitrogen balloon of about 1 L connected to the reaction flask.
  • the hydrogenation reaction is usually vacuumized and filled with hydrogen, and the operation is repeated 3 times.
  • the hydrogen atmosphere means that the reaction bottle is connected with a hydrogen balloon of about 1L.
  • microwave reaction use Initiator+ microwave reactor.
  • NMR nuclear magnetic resonance
  • MS mass spectroscopy
  • LC-MS Thermo liquid mass spectrometer
  • UltiMate 3000+MSQ PLUS Thermo liquid mass spectrometer
  • a Thermo high pressure liquid chromatograph UltiMate 3000
  • Reverse-phase preparative chromatography Thermo (UltiMate 3000) reverse-phase preparative chromatography was used.
  • Fast column chromatography uses Agel (FS-9200T) automatic column passing machine, and silica gel prepacked column uses Santai Prepacked columns.
  • Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plates are used for thin-layer chromatography silica gel plates, and the specifications used for thin-layer chromatography separation and purification products are 0.4mm to 0.5mm.
  • the first step the compound Int-1a (5.0g, 28.09mmol), Int-1b (5.61g, 36.51mmol) and sodium bicarbonate (7.08g, 84.26mmol) were dissolved in ethanol (50mL) and water (5mL) , the reaction solution was heated to reflux overnight. After the reaction was monitored by LCMS, it was cooled to room temperature, suction filtered, the filter cake was washed with water, and then dried to obtain off-white solid Int-1 (5.0 g, yield 78%).
  • ESI-MS m/z: 227.4 [M+H] + .
  • the first step sodium hydrogen (1.59g, 39.64mmol, content 60%) was added to a two-necked flask filled with anhydrous tetrahydrofuran (10mL), placed in an ice-water bath, compound Int-2a (5.0g, 26.43mmol) It was dissolved in anhydrous tetrahydrofuran (30 mL), and slowly added dropwise to the reaction solution. After 30 minutes, the dropwise addition of deuteroiodomethane (4.02 g, 27.75 mmol) was started, and after the dropwise addition was completed, the mixture was slowly raised to room temperature and stirred overnight.
  • the third step the compound Int-1a (2.0g, 11.23mmol), Int-2c (3.17g, 20.22mmol) and sodium bicarbonate (2.83g, 33.70mmol) were dissolved in ethanol (20mL) and water (2mL) , the reaction solution was refluxed overnight. After the reaction was monitored by LCMS, it was cooled to room temperature, suction filtered, the filter cake was washed with water, and then dried to obtain off-white solid Int-2 (1.4 g, yield 54%).
  • the first step the compound Int-3a (10.17g, 55.26mmol) was dissolved in ethanol (50mL), at room temperature Hydrazine salt Int-3b (5.0 g, 46.05 mmol) was added under conditions, and the reaction was refluxed overnight.
  • the third step Dissolve compound Int-3e (1.1g, 3.74mmol) in methanol (10mL), add ammonia water (1mL) and Raney nickel (3mL suspension) to the reaction system in turn, replace hydrogen by hydrogen balloon and The reaction was carried out under atmosphere and room temperature for 16 hours, and the reaction was monitored by LCMS to complete.
  • ESI-MS m/z
  • the third step Dissolve the compound Int-4c (897mg, 3.18mmol) in methanol (30mL), add ammonia water (3mL) and Raney nickel (3mL suspension) to the reaction system in turn, replace the hydrogen with a hydrogen balloon and in a hydrogen atmosphere 1.
  • the reaction was carried out at room temperature for 3 hours, and the reaction was monitored by LCMS to complete.
  • ESI-MS m/z: 328.3 [M+CH 3 CN+H] + .
  • the first step the second step: the compound Int-5a (3.0g, 19.73mmol), Int-3d (2.41g, 19.73mmol) and cesium carbonate (12.85g, 39.45mmol) were added in acetonitrile (30mL), room temperature Stirred for 16 hours, LCMS monitored the completion of the reaction. Acetonitrile was distilled off under reduced pressure, and the residue was extracted by adding water and ethyl acetate. The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated by filtration.
  • the first step sodium hydrogen (153mg, 4.0mmol, content 60%) was added to anhydrous tetrahydrofuran (5mL)
  • compound Int-2a 500 mg, 2.09 mmol
  • anhydrous tetrahydrofuran (10 mL) was dissolved in anhydrous tetrahydrofuran (10 mL), and slowly added dropwise to the reaction solution.
  • iodomethane 593 mg, 4.18 mmol
  • Step 2 Dissolve Int-6b (500mg, 1.97mmol) in methanol (50mL), place in an ice-water bath, slowly add thionyl chloride (704mg, 5.92mmol) dropwise, and rise to room temperature after the addition is complete, Then rise to 60°C and react overnight. After the reaction was monitored by LCMS, the reaction solution was concentrated to obtain Int-6c (300 mg, yield 60%) as a yellow oil.
  • the third step the compound Int-1a (250mg, 1.40mmol), Int-6c (343mg, 1.69mmol) and sodium bicarbonate (353mg, 4.21mmol) were dissolved in ethanol (20mL) and water (2mL), the reaction solution Reflux overnight. After the reaction was monitored by LCMS, it was cooled to room temperature, filtered with suction, the filter cake was washed with water, and then dried to obtain an off-white solid Int-6 (170 mg, yield 43%).
  • ESI-MS (m/z): 277.2 [M+H] + .
  • the first step compound Int-5b (400mg, 1.57mmol) and cesium carbonate (1.03g, 3.15mmol) were added to N,N-dimethylformamide (8mL), then 1,1 - Difluoro-2-iodoethane (0.56 mL, 6.28 mmol). After the reaction solution was stirred at room temperature for 24 hours, LCMS monitored that only a small amount of raw materials remained in the reaction solution, adding water to quench the reaction, extracting with ethyl acetate, combining the organic phases and washing with saturated brine, drying over anhydrous sodium sulfate, and concentrating under reduced pressure The organic phase yielded a mixture of Int-7a and Int-7a'. ESI-MS (m/z): 319.1 [M+H] + .
  • the second step Dissolve the product of the first step in methanol (50mL), add ammonia water (5mL) and Raney nickel suspension (5mL) to the reaction system in turn, replace the hydrogen with a hydrogen balloon and react in a hydrogen atmosphere at room temperature After 2 hours, LCMS monitored the reaction to be complete.
  • ESI-MS (m/z): 323.3 [M+H] + .
  • Step 1 Add compound Int-8a (2.0g, 12.26mmol), Na 2 CO 3 (2.6g, 24.52mmol) into water (60mL), stir to dissolve, add iodine (3.11g, 12.26mmol) into the reaction solution and stirred at room temperature for 3 hours.
  • LCMS monitored the disappearance of the raw materials, adjusted the pH of the reaction solution to 5-6 with dilute hydrochloric acid, and extracted with ethyl acetate.
  • Step 2 Dissolve compound Int-8b (0.5g, 1.73mmol), benzyl bromide (337mg, 1.98mmol) in DMF (5mL), add K 2 CO 3 (364mg, 2.64mmol), and stir at 50°C for 2 hours , LCMS monitors the end of translation.
  • ESI-MS (m/z): 380.2 [M+H] + .
  • the third step compound Int-8c (500mg, 1.32mmol), cyclopropylboronic acid (566mg, 6.59mmol), tricyclohexylphosphine (74mg, 263umol), palladium acetate (29mg, 131umol), potassium phosphate (559mg, 2.63 mmol) was added to dioxane (30 mL) and water (3 mL), and the reaction system was replaced with nitrogen, then stirred at 90° C. for 17 hours under the protection of nitrogen atmosphere. LCMS monitored the reaction to be complete.
  • ESI-MS (m/z): 204.5 [M+H] + .
  • Step 5 Compounds Int-8e (172mg, 0.85mmol), Int-3d (144mg, 1.18mmol), Cs 2 CO 3 (0.64g, 2.0mmol) were added to DMF (5mL), stirred at 20°C for 17 hours , LCMS monitored the reaction to be complete. The reaction solution was added into water (100 mL), stirred for 30 minutes, and suction filtered to obtain a yellow solid Int-8f (170 mg, yield 65%). ESI-MS (m/z): 306.2 [M+H] + .
  • Step 6 Add compound Int-8f (90mg, 295umol), Raney nickel (0.5mL, aqueous suspension) into methanol (5mL), add ammonia water (0.1ml), and replace the reaction system with hydrogen in a hydrogen atmosphere Stir at room temperature for 2 hours, and the reaction is complete as monitored by LCMS.
  • the reaction solution was suction-filtered with celite, and the filtrate was concentrated to obtain a yellow solid Int-8 (85 mg), which was directly used in the next reaction without purification.
  • ESI-MS (m/z): 310.3 [M+H] + .
  • Second step dissolve compound Int-9b (2.0g, 9.61mmol) and compound Int-3d (1.17g, 9.61mmol) in N,N-dimethylformamide (10ml), add cesium carbonate (6.26g , 19.21 mmol). The reaction was stirred at room temperature for 4 hours. LCMS monitored the completion of the reaction. The reaction solution was diluted with water, the resulting suspension was filtered, and the filter cake was dried by a vacuum rotary evaporator to obtain a brown solid compound Int-9c (1.78 g, yield 59%). ESI-MS (m/z): 311.1 [M+H] + .
  • the first step under a nitrogen atmosphere, compound Int-5b (500mg, 1.97mmol) and cesium carbonate (1.28g, 3.93mmol) were added to N,N-dimethylformamide (10mL), and then added to the reaction solution 2,2,2-Trifluoroethyl trifluoromethanesulfonate (0.58 mL, 3.93 mmol) was added dropwise.
  • the second step Dissolve the product of the first step in methanol (60mL), add ammonia water (6mL) and Raney nickel suspension (6mL) to the reaction system in turn, replace the hydrogen with a hydrogen balloon and react in a hydrogen atmosphere at room temperature After 2 hours, LCMS monitored the reaction to be complete.
  • ESI-MS (m/z): 382.3 [M+CH 3 CN+H] + .
  • the first step compound Int-5b (400mg, 1.57mmol) and cesium carbonate (1.03g, 3.15mmol) were added to N,N-dimethylformamide (8mL), and then 1-fluoro - 2-iodoethane (548 mg, 3.15 mmol). After the reaction solution was stirred at room temperature for 16 hours, LCMS monitored the completion of the reaction, adding water to quench the reaction, extracting with ethyl acetate, combining the organic phases and washing with saturated brine, drying over anhydrous sodium sulfate, and concentrating the organic phase under reduced pressure to obtain the compound Int- Mixture of 11a and Int-11a'.
  • ESI-MS (m/z): 301.2 [M+H] + .
  • the second step Dissolve the product of the first step in methanol (50mL), add ammonia water (5mL) and Raney nickel suspension (5mL) to the reaction system in turn, replace the hydrogen with a hydrogen balloon and react in a hydrogen atmosphere at room temperature After 2 hours, LCMS monitored the reaction to be complete.
  • ESI-MS (m/z): 305.1 [M+H] + .
  • the first step Dissolve compound Int-3d (120mg, 0.1mmol) and compound Int-12a (200mg, 0.1mmol) in N,N-dimethylformamide (5mL), add cesium carbonate (383mg, 1.2mmol ). The reaction was stirred at room temperature for 4 hours. LCMS monitored the complete reaction of starting material. The reaction solution was diluted with water, the obtained suspension was filtered, and the filter cake was dried under reduced pressure to obtain a white solid Int-12b (265 mg, yield 88%). ESI-MS (m/z): 307.1 [M+H] + .
  • the first step compound Int-13a (700mg, 3.54mmol), Int-3d (433mg, 3.54mmol) and cesium carbonate (2.31g, 7.09mmol) were added in N,N-dimethylformamide (10mL) , reacted at 50° C. for 16 hours, and monitored the completion of the reaction by LCMS. N,N-dimethylformamide was distilled off under reduced pressure, then added with water and ethyl acetate for extraction, the combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, and the organic phase was concentrated under reduced pressure to obtain a white solid Int-13b ( 711 mg, yield 67%).
  • Step 2 Dissolve compound Int-13b (355mg, 1.18mmol) in methanol (35mL), add ammonia (3.5mL) () and Raney nickel (3.5mL suspension) to the reaction system in turn, and replace the hydrogen with a hydrogen balloon
  • the reaction was carried out under hydrogen atmosphere and room temperature for 3 hours, and the reaction was monitored by LCMS.
  • ESI-MS (m/z): 304.2 [M+H] + .
  • the third step Dissolve the compound Int-14c (250mg, 0.78mmol) in methanol (5mL), add ammonia water (1mL) and Raney nickel (3mL suspension) to the reaction system in turn, replace the hydrogen with a hydrogen balloon and in a hydrogen atmosphere 1.
  • the reaction was carried out at room temperature for 16 hours, and the reaction was monitored by LCMS.
  • ESI-MS (m/z): 325.3 [M+H] + .
  • Step 3 Dissolve compound Int-15d (213mg, 0.67mmol) in methanol (22mL), add ammonia water (2.2mL) and Raney nickel (2.2mL, aqueous suspension) to the reaction system in turn, and replace by hydrogen balloon Hydrogen was reacted for 16 hours under a hydrogen atmosphere at room temperature, and the reaction was completed by LCMS monitoring.
  • the reaction solution was diluted with methanol, filtered with suction, and the filtrate was concentrated to obtain the crude product Int-15.
  • ESI-MS (m/z): 325.2 [M+H] + .
  • Step 1 Compounds Int-16a (1.00 g, 11.89 mmol), Int-16b (3.38 g, 23.78 mmol) and sodium ethoxide (1.62 g, 23.78 mmol) were sequentially added to dimethyl sulfoxide (10 mL). The reaction solution was stirred at 60° C. for 16 hours. After the reaction was completed, 6N hydrochloric acid was added to quench the reaction, extracted with ethyl acetate, the combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, and the organic phase was concentrated under reduced pressure to obtain the crude product Int-16c.
  • Step 2 Compound Int-16c (1.94g), hydrazine hydrate (673mg, 10.75mmol, 80%) and acetic acid (5mL) were added to methanol (35mL), and the reaction solution was refluxed under nitrogen atmosphere for 16 hours. After the reaction was monitored by LCMS, the solvent was distilled off under reduced pressure, then diluted with water and sodium carbonate (solid) was added to pH 9, extracted with ethyl acetate, the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, and reduced pressure Concentration of the organic phase afforded Int-16d as a brown solid (1.73 g, 82% for two steps). ESI-MS (m/z): 177.5 [M+H] + .
  • the third step Compounds Int-16d (200 mg, 1.14 mmol), Int-15b (336 mg, 1.70 mmol) and potassium carbonate (314 mg, 2.27 mmol) were added into acetonitrile (10 mL). The reaction solution was stirred at 80°C for 16 hours.
  • Step 4 Dissolve compound Int-16e (228mg, 0.78mmol) in methanol (23mL), add ammonia water (2.3mL) and Raney nickel (2.3mL, aqueous suspension) to the reaction system in turn, and replace by hydrogen balloon hydrogen and reacted for 3 hours under a hydrogen atmosphere at room temperature, and LCMS monitored the completion of the reaction.
  • the reaction solution was diluted with methanol, filtered with suction, and the filtrate was concentrated to obtain the crude product Int-16.
  • ESI-MS (m/z): 297.3 [M+H] + .
  • Step 1 Dissolve compound Int-16e' (80mg, 0.27mmol) in methanol (8mL), add ammonia (0.8mL) and Raney nickel (0.8mL, aqueous suspension) to the reaction system in turn, and pass through a hydrogen balloon The hydrogen gas was replaced and reacted for 3 hours under a hydrogen atmosphere at room temperature, and the reaction was completed by LCMS monitoring. The reaction solution was diluted with methanol, filtered with suction, and the filtrate was concentrated to obtain the crude product Int-17. ESI-MS (m/z): 297.2 [M+H] + .
  • the first step Dissolve compound Int-18a (2.03g, 10mmol) in anhydrous dimethyl sulfoxide (10mL), add potassium hydroxide (1.68g, 30mmol) and deuteroiodomethane (2.17g, 15mmol) .
  • the reaction was stirred at room temperature for 12 hours.
  • LCMS detects that the reaction is complete.
  • the reaction solution was quenched with water, extracted with dichloromethane, the combined organic phases were washed with saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain a crude product of compound Int-18b (2.3 g).
  • ESI-MS (m/z): 238.3 [M+H] + .
  • Step 3 Compounds Int-18c (450 mg, 3 mmol) and Int-18d (539 mg, 3 mmol) were dissolved in tetrahydrofuran (3 mL), and N,N-diisopropylethylamine (1 g, 7.78 mmol) was added. The reaction was stirred at room temperature for 4 hours. LCMS detects that the reaction is complete.
  • Step 4 Compound Int-18e (400 mg, 1.30 mmol) was dissolved in methanol (5 mL), potassium carbonate (269 mg, 1.94 mmol) was added, followed by sodium dithionite (677 mg, 3.89 mmol). The reaction was stirred at room temperature for 30 minutes. Aqueous hydrochloric acid (2 mL, 4M) was then added. The reaction was stirred at room temperature for 8 hours. LCMS detects that the reaction is complete.
  • Embodiment 1 is prepared by the following steps:
  • the first step Dissolve compound Int-1 (1.0g, 4.41mmol) and compound Int-3 (1.71g, 5.74mmol) in n-butanol (6mL), add p-toluenesulfonic acid monohydrate (83mg, 0.44 mmol), reaction solution Stir at 160° C. for 4 hours under microwave conditions. After the reaction liquid was cooled to room temperature, the reaction liquid was concentrated under reduced pressure, and the residue was directly purified by reverse preparative HPLC to obtain white solid 1 (987 mg, yield 45%).
  • Embodiment 2 is prepared by the following steps:
  • the first step Dissolve compound Int-2 (50mg, 0.22mmol) and compound Int-3 (84mg, 0.28mmol) in n-butanol (2mL), add p-toluenesulfonic acid monohydrate (4mg, 0.02mmol) , the reaction solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain a white solid 2 (36 mg, yield 34%).
  • the first step Dissolve compound Int-5b (500mg, 1.97mmol) in N,N-dimethylformamide (10mL), then add cesium carbonate (1.28g, 3.93mmol) and deuterated iodoethane (633mg , 3.93 mmol). The reaction was stirred at room temperature for 4 hours. LCMS monitored the completion of the reaction. The reaction solution was diluted with saturated aqueous sodium chloride and extracted with ethyl acetate. The organic phase was washed with saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a mixture of compounds 3a and 3a'. ESI-MS (m/z): 288.9 [M+H] + .
  • Step 2 Dissolve the products 3a and 3a' obtained in the previous step in methanol (15mL) and ammonia water (1mL), add Raney Nickel (1mL, aqueous suspension), and stir the reaction system at room temperature for 12 hours after replacing hydrogen .
  • the reaction solution was filtered through celite, and the filtrate was concentrated.
  • Step 3 Dissolve compound 3b (60mg, 0.21mmol) in n-butanol (3mL), add compound Int-1 (31mg, 0.14mmol) and p-toluenesulfonic acid monohydrate (3mg, 0.02mmol), The reaction solution Stir at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain a white solid 3 (29 mg, yield 29%).
  • Step 4 Starting from compound 3b' (60 mg, 0.21 mmol), a white solid 3' (20 mg, yield 18%) was obtained by a method similar to the third step.
  • the first step Dissolve compound Int-5b (200mg, 0.79mmol) in N,N-dimethylformamide (5mL), then add cesium carbonate (513mg, 1.57mmol) and 2-iodopropane (268mg, 1.57 mmol). The reaction was stirred at room temperature for 4 hours. LCMS monitored the completion of the reaction. The reaction solution was diluted with saturated aqueous sodium chloride and extracted with ethyl acetate. The organic phase was washed with saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated to obtain a mixture of compounds 4a and 4a'. ESI-MS (m/z): 297.2 [M+H] + .
  • the third step Dissolve compound 4b (62mg, 0.21mmol) in n-butanol (3mL), add compound Int-1 (31mg, 0.14mmol) and p-toluenesulfonic acid monohydrate (3mg, 0.02mmol), react liquid in Stir at 160°C for 3 hours under microwave conditions. After the reaction liquid was cooled to room temperature, the reaction liquid was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain white solid 4 (13 mg, yield 13%).
  • Step 4 Starting from compound 4b' (60 mg, 0.21 mmol), a white solid 4' (5 mg, yield 5%) can be obtained by a method similar to the third step.
  • Embodiment 5 is prepared by the following steps:
  • the first step Dissolve compound Int-9 (100mg, 0.32mmol) and compound Int-1 (48mg, 0.21mmol) in n-butanol (3mL), add p-toluenesulfonic acid monohydrate (6mg, 0.03mmol) , the reaction solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain white solid 5 (20 mg, yield 13%).
  • Embodiment 6 is prepared by the following steps:
  • the first step intermediate Int-1 (37mg, 0.16mmol), intermediate Int-11 (55mg, 0.18mmol) and trifluoroacetic acid (2mg, 0.02mmol) were dissolved in n-butanol (2mL), and the The reaction was carried out at 160° C. for 4 hours. LCMS monitored the completion of the reaction. The reaction solution was purified by reverse-phase preparative HPLC to obtain a white solid 6 (21 mg, yield 26%).
  • Embodiment 7 is prepared by the following steps:
  • the first step intermediate Int-1 (39mg, 0.17mmol), intermediate Int-10 (65mg, 0.19mmol) and trifluoroacetic acid (2mg, 0.02mmol) were dissolved in n-butanol (2mL), and the The reaction was carried out at 160° C. for 4 hours. LCMS monitored the completion of the reaction. The reaction solution was purified by reverse-phase preparative HPLC to obtain 7 (24 mg, yield 26%) as a white solid.
  • Embodiment 8 is prepared by the following steps:
  • the first step intermediate Int-1 (38mg, 0.17mmol), intermediate Int-7 (60mg, 0.19mmol) and trifluoroacetic acid (2mg, 0.02mmol) were dissolved in n-butanol (2mL), and the The reaction was carried out at 160° C. for 4 hours. LCMS monitored the completion of the reaction. The reaction solution was purified by reverse-phase preparative HPLC to obtain 8 (24 mg, yield 27%) as a white solid.
  • Embodiment 9 is prepared by the following steps:
  • the first step intermediate Int-1 (34mg, 0.15mmol), intermediate Int-8 (51mg, 0.16mmol) and trifluoroacetic acid (2mg, 0.02mmol) were dissolved in n-butanol (2mL), and the The reaction was carried out at 160° C. for 4 hours. LCMS monitored the completion of the reaction. The reaction solution was purified by reverse-phase preparative HPLC to obtain white solid 9 (16 mg, yield 21%).
  • Example 10 was prepared by the following steps:
  • the first step intermediate Int-2 (50mg, 0.22mmol), intermediate Int-13 (85mg, 0.28mmol) and p-toluenesulfonic acid monohydrate (4mg, 0.02mmol) were dissolved in n-butanol (2mL) In the microwave, the reaction was carried out at 160°C for 3 hours. LCMS monitored the completion of the reaction. The reaction solution was purified by reverse-phase preparative HPLC to obtain white solid 10 (23 mg, yield 21%).
  • Example 11 was prepared by the following steps:
  • the first step Dissolve compound Int-12 (100mg, 0.34mmol) and compound Int-1 (75mg, 0.34mmol) in n-butanol (3mL), add p-toluenesulfonic acid monohydrate (6mg, 0.03mmol) ,The reaction solution Stir at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain white solid 11 (80 mg, yield 48%).
  • Example 12 was prepared by the following steps:
  • the first step Dissolve compound Int-1 (50mg, 0.22mmol) and compound Int-14 (93mg, 0.29mmol) in n-butanol (2mL), add p-toluenesulfonic acid monohydrate (4mg, 0.02mmol) , the reaction solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain white solid 12 (36 mg, yield 30%).
  • Example 13 was prepared by the following steps:
  • the first step intermediate Int-1 (40mg, 0.18mmol), intermediate Int-15 (69mg, 0.21mmol) and trifluoroacetic acid (2mg, 0.02mmol) were dissolved in n-butanol (2mL), and the The reaction was carried out at 160° C. for 3 hours. LCMS monitored the completion of the reaction. The reaction solution was purified by reverse-phase preparative HPLC to obtain white solid 13 (37 mg, yield 41%).
  • Example 14 was prepared by the following steps:
  • the first step Dissolve compound Int-1a (500mg, 2.81mmol) and compound 14a (602mg, 3.65mmol) in ethanol (20mL) and water (2mL), then add sodium bicarbonate (707mg, 8.34mmol), and The temperature of the reaction liquid was raised to 80° C. and stirred for 16 hours. After the reaction solution was cooled to room temperature, water (12 mL) was added to the reaction solution, and after filtration, off-white solid 14b (385 mg, yield 57%) was obtained, ESI-MS (m/z): 239.4 [M+H] + .
  • Second step Dissolve compound 14b (42mg, 0.17mmol) and compound Int-3 (68mg, 0.23mmol) in n-butanol (2mL), add p-toluenesulfonic acid monohydrate (3mg, 0.01mmol), react The solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain white solid 14 (15 mg, yield 17%).
  • Example 15 was prepared by the following steps:
  • the first step Ethyl formate (3.17g, 42.73mmol) and solid sodium ethoxide (3.49g, 50.50mmol) were sequentially added into dry tetrahydrofuran (140mL). After compound 15a (7 g, 38.85 mmol) was added at a temperature of 5-10° C., the reaction solution was heated to 50° C. and kept stirring for 2 hours, and the disappearance of the raw material was monitored by HPLC. Tetrahydrofuran was distilled off under reduced pressure to obtain yellow oil 15b, which was directly used in the next step.
  • Step 2 add 150ml of absolute ethanol to the oil 15b obtained in the previous step, stir and dissolve at room temperature, add trifluoroacetamidine (4.35g, 33.01mmol, purity 85%) dropwise, and keep stirring at 30°C for 5 hours, Then the temperature was raised to 80° C. and stirring was continued for 2 hours, and the disappearance of the raw material was monitored by HPLC. After the reaction solution cooled down, about 100ml of ethanol was evaporated under reduced pressure, and the remaining residue was added to 300ml of ice water, adjusted to pH 3 with concentrated hydrochloric acid, stirred for 0.5 hours, filtered with suction, and the filter cake was dried to obtain yellow solid compound 15c (4.37g, yield 41%, purity 99%).
  • ESI-MS (m/z): 271.4 [M+H] + .
  • Step 3 Add compound 15c (4.0g, 14.80mmol) to 60ml of acetonitrile, dropwise add phosphorus oxychloride (6.81g, 44.41mmol), stir for 10 minutes after the addition, heat up to 80°C, keep stirring for 2 Hours, HPLC monitoring raw material conversion is complete. Acetonitrile was removed under reduced pressure, and the residue was added to 200 mL of ice water, stirred for 0.5 hours, and filtered with suction to obtain a yellow solid 15d (3.9 g, yield 86%, purity 95%).
  • the fourth step Compound 15d (2.0g, 6.93mmol), trimethylboroxane (2.61g, 20.79mmol), palladium acetate (155mg, 0.69mol), potassium phosphate (2.94g, 13.86mmol) in sequence
  • 1,4-dioxane 150 mL
  • water 15 mL
  • HPLC HPLC monitors that the conversion of the raw material is complete
  • the filtrate is concentrated.
  • ESI-MS (m/z): 269.1 [M+H] + .
  • Step 5 Compound 15e (1.0 g, 3.73 mmol) was added to 20 mL of methanol, 10% palladium on carbon (100 mg) was added, and the reaction system was replaced with hydrogen and stirred overnight at room temperature. The reaction liquid was filtered with celite, and the filtrate was concentrated to obtain compound 15f (630 mg, yield 92%, purity 97%). ESI-MS (m/z): 177.1 [MH] - .
  • Step 6 Add compound 15f (0.5g, 2.81mmol), Cs 2 CO 3 (1.83g, 5.61mmol), Int-3d (514mg, 4.21mmol) to N,N-dimethylformamide (10mL ), stirred overnight at 20°C, HPLC monitored the disappearance of the raw materials, the reaction solution was added to 50ml of ice water, stirred for 0.5 hours, and filtered with suction to obtain 15g of a yellow solid (510mg, yield 64%, purity 99%).
  • ESI-MS (m/z): 281.3 [M+H] + .
  • Step 7 Dissolve compound 15g (100mg, 0.36mmol) in methanol (5mL), add ammonia water (0.2mL), add RaneyNickel (0.5mL, water suspension), and stir the reaction system at room temperature after replacing hydrogen 2 hours.
  • the reaction liquid was filtered with celite, and the filtrate was concentrated to obtain compound 15h (95 mg, yield 93%, purity 90%), ESI-MS (m/z): 284.9 [M+H] + .
  • Step 8 Dissolve compound 15h (95 mg, 0.33 mmol) in n-butanol (3 mL), add compound Int-1 (79 mg, 0.35 mmol) and p-toluenesulfonic acid monohydrate (3 mg, 0.02 mmol), and react The solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction liquid was cooled to room temperature, it was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain white solid 15 (31 mg, yield 21%, purity 99%).
  • Example 16 was prepared by the following steps:
  • Example 17 was prepared by the following steps:
  • Example 18 was prepared by the following steps:
  • the first step compound 18a (1.01g, 5mmol) and potassium hydroxide (842mg, 15mmol) were dissolved in anhydrous dimethyl sulfoxide (10mL), then deuteroiodomethane (1.59g, 11mmol) was added dropwise into the reaction solution. The reaction solution was stirred at room temperature for 8 hours. LCMS monitored the completion of the reaction. The reaction solution was diluted with water, extracted with ethyl acetate, the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to obtain a crude compound 18b (1.0 g, yield 84%). ESI-MS (m/z): 236.3 [M+H] + .
  • Second step Compound 18b (1.0 g) was dissolved in dioxane hydrochloride solution (4M, 10 mL). The reaction was stirred at room temperature for 4 hours. LCMS monitored the completion of the reaction. The reaction solution was concentrated under reduced pressure to obtain a crude product of Compound 18c (720 mg). ESI-MS (m/z): 136.1 [M+H] + .
  • the third step Compound 18c (720 mg) and compound 18d (872 mg, 4.19 mmol) were dissolved in tetrahydrofuran (10 mL), and N,N-diisopropylpropylamine (1.62 g, 12.57 mmol) was added. The reaction solution was stirred at room temperature for 8 hours. LCMS monitored the completion of the reaction. The reaction solution was diluted with water, filtered, the filter cake was washed with water, and dried to obtain off-white solid 18d (993 mg, yield 77% in two steps). ESI-MS (m/z): 307.2 [M+H] + .
  • Step 4 Dissolve compound 18d (306 mg, 1 mmol) and potassium carbonate (207 mg, 1.5 mmol) in methanol (10 mL), dissolve sodium dithionite (871 mg, 5 mmol) in water and add dropwise to the reaction solution. The reaction was stirred at room temperature for 15 minutes. LCMS monitored the complete reaction of starting material. Dioxane hydrochloride solution (4M, 1.25 mL) was added to the reaction solution. The reaction was continued to stir at room temperature for 8 hours.
  • Step 5 Dissolve Int-3 (35mg, 0.12mmol) and compound 18e (28mg, 0.12mmol) in n-butanol (3mL), add p-toluenesulfonic acid monohydrate (2mg, 0.01mmol), and the reaction solution Stir at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain white solid 18 (10 mg, yield 17%).
  • the first step trifluoromethanesulfonic anhydride (1.25 g, 4.44 mmol) was added to compound 19a (400 mg, 3.70 mmol) and 4-dimethylaminopyridine (543 mg, 4.44 mmol) in dichloromethane under ice bath conditions solution (10 mL). After reacting at room temperature for 16 hours, the reaction was quenched by adding saturated aqueous ammonium chloride solution, extracted with dichloromethane, the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, and the organic phase was concentrated under reduced pressure to obtain the crude product of compound 19b , used directly in the next reaction.
  • Step 3 Dissolve the mixture of 19c and 19c' (283mg, 0.82mmol) in methanol (30mL), add ammonia water (3mL) and Raney nickel (3mL, aqueous suspension) to the reaction system in turn, and pass through the hydrogen balloon The hydrogen was replaced and reacted for 2 hours under a hydrogen atmosphere at room temperature, and the reaction was completed by LCMS monitoring. The reaction solution was diluted with methanol, filtered through celite, and the filtrate was concentrated to obtain a mixture of 19d and 19d', which was directly used in the next reaction. ESI-MS (m/z): 349.3 [M+H] + .
  • the fourth step Int-2 (80mg, 0.35mmol), the mixture of 19d and 19d' (146mg, crude product) and p-toluenesulfonic acid monohydrate (4mg, 0.03mmol) were dissolved in n-butanol (2mL), in React under the condition of microwave at 160° C. for 3 hours. LCMS monitored the completion of the reaction. The reaction solution was purified by reverse-phase preparative HPLC to obtain white solids 19 (28 mg, yield 15%) and 19' (17 mg, yield 9%), respectively.
  • Example 20 was prepared by the following steps:
  • the first step Dissolve compound Int-6 (50mg, 0.18mmol) and compound Int-3 (70mg, 0.23mmol) in n-butanol (6mL), add p-toluenesulfonic acid monohydrate (3mg, 18umol), The reaction solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain a white solid 20 (3.9 mg, yield 4%).
  • Example 21 was prepared by the following steps:
  • the first step Compound 15d (550mg, 1.91mmol), cyclopropylboronic acid (818mg, 9.53mmol), palladium acetate (42mg, 0.19mmol), tricyclohexylphosphine (106mg, 0.38mmol) and potassium phosphate (1.21g , 5.72mmol) was added to the mixed solution of 1,4-dioxane (50mL) and water (5mL). After the reaction system was replaced with nitrogen, it was reacted at 110°C under nitrogen atmosphere for 16 hours, and the reaction was monitored by LCMS.
  • the second step Dissolving compound 21a (530mg, 1.80mmol) in methanol (10mL), adding palladium carbon (10%, 21mg) to the reaction system, displacing hydrogen through a hydrogen balloon and reacting under hydrogen atmosphere and room temperature for 16 hours, LCMS monitored the completion of the reaction.
  • the reaction solution was diluted with methanol, suction filtered, and the filtrate was concentrated to obtain brown oily liquid 21b (330 mg, yield 89%).
  • Step 4 Dissolve compound 21c (350mg, 1.14mmol) in methanol (20mL), add ammonia water (3.5mL) and Raney nickel (3.5mL, aqueous suspension) to the reaction system in turn, replace the hydrogen with a hydrogen balloon and The reaction was carried out under hydrogen atmosphere and room temperature for 16 hours, and the reaction was monitored by LCMS to complete.
  • ESI-MS (m/z): 311.2 [M+H] + .
  • Step 5 Dissolve compound Int-1 (50mg, 0.22mmol) and compound 21d (82.13mg, 0.26mmol) in n-butanol (2mL), add p-toluenesulfonic acid monohydrate (4.19mg, 22umol), The reaction solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain a white solid 21 (48.18 mg, yield 43%).
  • Example 22 was prepared by the following steps:
  • the first step Dissolve compound Int-18 (50mg, 0.17mmol) and compound Int-3 (40.85mg, 0.17mmol) in n-butanol (2mL), add p-toluenesulfonic acid monohydrate (2.89mg, 17umol ), the reaction solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain a white solid 22 (20 mg, yield 23%).
  • Example 23 was prepared by the following steps:
  • Step 2 Add compound 23a (1.0g, 3.73mmol) to 20ml of methanol, add palladium carbon (10%, 100mg), replace hydrogen in the reaction system and stir at room temperature overnight, filter the reaction solution with diatomaceous earth, and concentrate the filtrate , to obtain compound 23b (677 mg, yield 94%, purity 97%).
  • Step 3 Add compound 23b (0.5g, 2.81mmol), Cs 2 CO 3 (1.83g, 5.61mmol), Int-3d (514mg, 4.21mmol) to N,N-dimethylformamide (10mL ), stirred overnight at 20°C, and the disappearance of raw materials was monitored by HPLC. The reaction solution was added to 50 ml of ice water, stirred for 0.5 hours, and filtered with suction to obtain a yellow solid 23c (530 mg, yield 64%, purity 99%). ESI-MS (m/z): 294.3 [M+H] + .
  • Step 4 Dissolve compound 23c (100mg, 0.36mmol) in methanol (5mL), add ammonia water (0.2mL), add Raney Nickel (0.5mL, aqueous suspension), and replace hydrogen in the reaction system at room temperature Stir for 2 hours.
  • the reaction liquid was filtered with celite, and the filtrate was concentrated to obtain compound 23d (97 mg, yield 93%, purity 90%), ESI-MS (m/z): 298.5 [M+H] + .
  • Step 5 Dissolve compound Int-1 (79.74mg, 0.35mmol) and compound 23d (104.93mg, 0.35mmol) in n-butanol (2mL), add p-toluenesulfonic acid monohydrate (6.68mg, 0.35umol ), the reaction solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by reverse preparative HPLC to obtain a white solid 23 (33 mg, yield 19%).
  • Example 25 was prepared by the following steps:
  • Step 1 Compound Int-8c (930 mg, 2.45 mmol) was dissolved in tetrahydrofuran (10 mL), and n-butyllithium (1.5 mL, 2M in hexane) was slowly added dropwise to the solution at -78°C. The reaction solution was further stirred at -78°C for 10 minutes. Then N,N-dimethylformamide (359mg, 4.91mmol) was slowly added dropwise to the reaction solution, and the reaction solution was stirred at -78°C for 1 hour. LCMS detects that the reaction is complete.
  • Step 3 Dissolve compound 25b (247mg, crude product) in dichloromethane (5mL), slowly add boron tribromide (308mg, 1.23mmol) into the solution at -78°C, and react at -78°C Stirring was continued for 30 minutes.
  • LCMS detects that the reaction is complete.
  • Step 4 Dissolve compound 25c (82mg, 0.39mmol) and compound Int-2 (47mg, 0.39mmol) in N,N-dimethylformamide (3mL), add cesium carbonate (251mg, 0.77mmol), The reaction solution was stirred at 50°C for 12 hours. LCMS detects that the reaction is complete. The reaction solution was diluted with water, and filtered under reduced pressure to obtain compound 25d (98 mg, yield 80%).
  • Step 6 Dissolve compound 25e (20mg, 0.06mmol) and Int-2 (15mg, 0.06mmol) in n-butanol (3mL), add p-toluenesulfonic acid monohydrate (2mg, 0.01mmol), and the reaction solution Stir at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the residue was purified by Prep-HPLC to obtain compound 25 (7 mg, yield 22%) as a white solid.
  • Example 26 was prepared by the following steps:
  • Step 1 Dissolve compound 26a (1.0g, 4.65mmol) in anhydrous tetrahydrofuran (20mL), under the protection of nitrogen, cool down to -78°C, and slowly add n-butyllithium (3.05mL, 4.88mmol) dropwise, After one hour, dropwise addition of triisopropyl borate (0.79 mL, 6.97 mmol) was started, and the reaction was completed after two hours by LCMS. Water was slowly added dropwise to quench the reaction.
  • Second step Compounds 26b and 26b' (400mg, 1.73mmol), Int-3d (232.48mg, 1.90mmol) and N,N-diisopropylethylamine (671.1mg, 5.19mmol) were added to acetonitrile (10mL ), reacted at 120° C. for 4 hours, and LCMS monitored the end of the reaction.
  • the third step Dissolve compound 26c (180mg, 0.54mmol) in methanol (30mL), add ammonia water (5mL) and Raney nickel (3.5mL, aqueous suspension) to the reaction system in turn, replace hydrogen by hydrogen balloon and in The reaction was carried out under hydrogen atmosphere and room temperature for 3 hours, and the reaction was monitored by LCMS to complete. The reaction solution was diluted with methanol, filtered with suction, and the filtrate was concentrated to obtain a brown oily liquid 26d (150 mg, yield 82%). ESI-MS (m/z): 338.5 [M+H] + .
  • Step 4 Dissolve compound 26d (74.39mg, 0.22mmol) in n-butanol (2mL), add compound Int-1 (50mg, 0.22mmol) and p-toluenesulfonic acid monohydrate (4.19mg, 22umol), The reaction solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, it was concentrated under reduced pressure, and the residue was purified by reverse-phase preparative HPLC to obtain 26 (38.57 mg, yield 33%) as a white solid.
  • Example 27 was prepared by the following steps:
  • the first step under ice-water bath conditions, di-tert-butyl carbonic anhydride (1.75g, 8.02mmol) was added dropwise to a dichloromethane solution of Int-13 (2.03g, 6.68mmol) and triethylamine (1.39mL, 10.02mmol). methane (40mL) solution. After the reaction solution was stirred at room temperature for 16 hours, water was added to quench the reaction.
  • Second step Compound 27a (300mg, 0.74mmol), zinc cyanide (174mg, 1.49mmol), tetrakistriphenylphosphine palladium (172mg, 0.15mol) and N,N-dimethylformamide (10mL) were added into a round bottom flask. After the reaction solution was stirred at 120° C. for 24 hours under nitrogen protection, the reaction was monitored by LCMS.
  • Step 3 Trifluoroacetic acid (0.5 mL) was added dropwise to a solution of compound 27b in dichloromethane (6 mL). After the reaction solution was stirred at room temperature for 5 hours, the reaction was monitored by LCMS to complete. Concentrated by vacuum distillation To the reaction solution, an aqueous sodium hydroxide solution was added to pH 8, extracted with ethyl acetate, the combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, and the organic phase was concentrated to obtain the crude product 27c.
  • ESI-MS (m/z): 295.3 [M+H] + .
  • Step 4 Dissolve compound Int-2 (45mg, 0.20mmol) in n-butanol (3mL), add compound 27c (64mg, 0.22mmol) and p-toluenesulfonic acid monohydrate (8mg, 44umol), and the reaction solution Stir at 160° C. for 3 hours under microwave conditions. LCMS monitored the completion of the reaction. The reaction solution was purified by reverse-phase preparative HPLC to obtain a white solid 27 (16 mg, yield 8%).
  • Example 28 was prepared by the following steps:
  • ESI-MS (m/z): 385.1 [M+H] + .
  • the second step 28b (400mg, 1.04mmol), n-butyl bis (1-adamantyl) phosphine (37mg, 103umol), 28c (492mg, 2.08mmol), TBAF (56mg, 259umol), palladium acetate (23mg , 103umol), cesium carbonate (1.02g, 3.12mmol), water (1mL) were added to n-butanol (7mL), and stirred at 110°C for 17 hours under nitrogen protection.
  • Step 3 Add compound 28d (273mg, 627umol) into dichloromethane (3ml), add trifluoroacetic acid (1ml), and stir at room temperature for 2 hours.
  • LCMS showed that the conversion of the raw material was complete. Distilled under reduced pressure, the residue was sequentially added to 10ml of ethyl acetate and 10ml of saturated aqueous sodium bicarbonate solution for liquid separation. The organic phase was washed with water, dried over sodium sulfate, and concentrated to obtain a brown oil, which was directly used in the next step.
  • ESI-MS (m/z): 336.0 [M+H] + .
  • Step 4 Add compound 28e, Int-1 (79mg, 351umol) and p-toluenesulfonic acid (6mg, 35umol) obtained in the previous step into n-butanol (2mL), stir in microwave at 160°C for 3 hours. LCMS Display ingredients disappear. The reaction solution was purified by reverse-phase preparative HPLC to obtain a white solid 28 (20 mg, yield 6% in two steps).
  • Example 30 was prepared by the following steps:
  • the first step Dissolve compound 30a (1.10g, 5mmol) in anhydrous tetrahydrofuran (10mL), cool the solution to 0°C, and slowly add sodium hydride (478mg, 60% dispersion in mineral oil) to the reaction under nitrogen atmosphere in the liquid. The reaction solution was stirred at 0°C for 10 minutes. Then methyl iodide (1.42 g, 10 mmol) was added dropwise to the reaction solution. The reaction solution was slowly warmed to room temperature and stirring was continued for 2 hours. LCMS detects that the reaction is complete.
  • the second step Dissolve compound 30b (1.17g) in methanol (10mL), slowly add thionyl chloride (1.19g, 10mmol) dropwise to the reaction solution at room temperature, then the reaction solution is heated to 70°C and continued Stir at this temperature for 2 hours. LCMS detects that the reaction is complete. After the reaction liquid was cooled to room temperature, the reaction liquid was concentrated under reduced pressure to obtain compound 30c (918 mg, crude product).
  • Step 4 Dissolve compound 30d (320 mg, 1 mmol) and compound Int-3 (300 mg, 1 mmol) in N, N-dimethylformamide (5 mL), add N, N-diisopropylethylamine ( 390 mg, 3 mmol). The reaction was stirred at room temperature for 4 hours. LCMS detects that the reaction is complete.
  • Step 5 Dissolve compound 30e (430 mg, 0.74 mmol) in methanol (5 mL), add 10% palladium on carbon (394 mg), and stir the reaction system at room temperature for 6 hours after replacing hydrogen. LCMS detects that the reaction is complete. The reaction solution was filtered with celite, the filtrate was concentrated under reduced pressure, and the residue was purified by Prep-HPLC to obtain compound 30 (15 mg, yield 4%) as a white solid.
  • Example 31 was prepared by the following steps:
  • Step 1 Compound 30 (10 mg, 0.02 mmol) was dissolved in dichloromethane (2 mL), and boron tribromide (10 mg, 0.04 mmol) was slowly added dropwise to the reaction solution at -78°C. The reaction continued to stir at -78°C for 30 minutes. LCMS detects that the reaction is complete. The reaction was quenched by adding saturated aqueous sodium bicarbonate solution, extracted with dichloromethane, the organic phases were combined and washed with saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the residue was purified by Prep-HPLC to obtain white solid compound 31 (8 mg , yield 80%).
  • Example 34 was prepared by the following steps:
  • the first step compound 27a (675mg, 1.67mmol), vinylboronic acid pinacol ester (1.29g, 8.36mmol), tetrakistriphenylphosphine palladium (194mg, 0.17mol), sodium carbonate (354mg, 3.34mmol) , water (3 mL) and 1,4-dioxane (17 mL) were added to a round bottom flask. After the reaction solution was stirred for 16 hours at 120° C. under nitrogen protection, the reaction was monitored by LCMS.
  • the second step sodium periodate (907mg, 4.24mmol) was added to 34a (559mg, 1.41mmol) in the mixed solution of tetrahydrofuran (12mL) and water (3mL), after the reaction solution was stirred at room temperature for one minute, to To this was added potassium osmate (41 mg, 0.14 mmol). After the reaction solution was stirred at 40° C. for 2 hours, the reaction was monitored by LCMS to complete. Add water and ethyl acetate for extraction, combine the organic phases and successively use thiosulfur NaCl aqueous solution and saturated brine were washed, dried over anhydrous sodium sulfate, and the organic phase was concentrated to obtain the crude product 34b.
  • ESI-MS (m/z): 398.3 [M+H] + .
  • Step 4 Thionyl chloride (0.13 mL, 1.80 mmol) was added dropwise to a solution of 34c (359 mg, 0.90 mmol) in dichloromethane (14 mL). After the reaction solution was stirred at room temperature for 2 hours, the reaction was monitored by LCMS to complete. The reaction solution was concentrated by distillation under reduced pressure to obtain the crude product 34d. ESI-MS (m/z): 418.2 [M+H] + .
  • Step 5 Trimethylsilyl cyanide (178 mg, 1.80 mmol) was added dropwise to a solution of cesium fluoride (119 mg, 1.80 mmol) in acetonitrile (8 mL) at 10°C. After the reaction solution was stirred for 30 minutes, cesium carbonate (586 mg, 1.80 mmol) and the above crude product 34d were added thereto, and the reaction solution was heated to 40° C. and continued to stir at this temperature for 16 hours. After the reaction was monitored by LCMS, water and ethyl acetate were added for extraction, the combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, and the organic phase was concentrated to obtain the crude product 34e. ESI-MS (m/z): 409.3 [M+H] + .
  • Step 6 Trifluoroacetic acid (2 mL) was added dropwise to a solution of the above crude product 34e in dichloromethane (8 mL). After the reaction solution was stirred at room temperature for 1.5 hours, the reaction was monitored by LCMS to complete. The reaction solution was concentrated by distillation under reduced pressure, aqueous sodium hydroxide solution was added to pH 8, extracted with ethyl acetate, the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, and the organic phase was concentrated to obtain the crude product 34f.
  • ESI-MS (m/z): 309.3 [M+H] + .
  • Step 7 Dissolve compound Int-1 (50mg, 0.22mmol) in n-butanol (2mL), add compound 34f (82mg, crude product) and p-toluenesulfonic acid monohydrate (8mg, 44umol), and the reaction solution is in Stir at 160°C for 3 hours under microwave conditions. LCMS monitored the completion of the reaction. The reaction solution was purified by reverse-phase preparative HPLC to obtain 34 (20 mg, 4% yield in four steps) as a white solid.
  • Example 35 was prepared by the following steps:
  • Step 1 Dissolve compound 35a (300mg, 1.82mmol) in anhydrous tetrahydrofuran (5mL), under the protection of nitrogen, cool down to -78°C, and slowly add n-butyl lithium (1.19mL, 1.91mmol) dropwise. After 1 hour, dropwise addition of triisopropyl borate (0.63 mL, 2.73 mmol) was started, and the reaction was completed after 2 hours by LCMS monitoring. Water was slowly added dropwise to quench the reaction.
  • Step 3 Dissolve compound 35c (270mg, 0.95mmol) in methanol (30mL), add ammonia water (3.5mL) and Raney nickel (3.5mL, aqueous suspension) to the reaction system in turn, replace the hydrogen with a hydrogen balloon and The reaction was carried out under a hydrogen atmosphere at room temperature for 3 hours, and the reaction was monitored by LCMS to complete. The reaction solution was diluted with methanol, filtered with suction, and the filtrate was concentrated to obtain a brown oily liquid 35d (270 mg, yield 98%). ESI-MS (m/z): 271.5 [M+H] + .
  • Step 4 Dissolve compound 35d (82.36mg, 0.29mmol) in n-butanol (2mL), add compound Int-1 (50mg, 0.22mmol) and p-toluenesulfonic acid monohydrate (4.19mg, 22umol), The reaction solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, it was concentrated under reduced pressure, and the residue was purified by reverse-phase preparative HPLC to obtain 35 (1.07 mg, yield 1%) as a white solid.
  • Example 36 was prepared by the following steps:
  • the second step compound 36b (300mg, 0.84mmol), cyclopropylboronic acid (108mg, 1.26mmol), palladium acetate (19mg, 84umol), tricyclohexylphosphine (47mg, 0.17mmol) and potassium phosphate (534mg, 2.52 mmol), water (2 mL) and toluene (10 mL) were added to a round bottom flask. After the reaction solution was stirred for 16 hours at 80° C. under nitrogen protection, the reaction was monitored by LCMS.
  • the third step the compound 36c (76mg, 0.28mmol) was dissolved in methanol (8mL), followed by reaction Ammonia water (0.8 mL) and Raney nickel (0.8 mL, aqueous suspension) were added to the system, hydrogen was replaced by a hydrogen balloon, and the reaction was carried out under hydrogen atmosphere at room temperature for 3 hours, and the reaction was completed by LCMS monitoring.
  • the reaction solution was diluted with methanol, filtered with suction, and the filtrate was concentrated to obtain a brown oily liquid 36d.
  • ESI-MS (m/z): 276.4 [M+H] + .
  • the fourth step the compound 36d (51mg) obtained in the previous step was dissolved in n-butanol (2mL), and compound Int-1 (45mg, 0.19mmol) and p-toluenesulfonic acid monohydrate (7mg, 37umol) were added to react The solution was stirred at 160° C. for 3 hours under microwave conditions. After the reaction solution was cooled to room temperature, it was concentrated under reduced pressure, and the residue was purified by reverse-phase preparative HPLC to obtain 36 as a white solid (10 mg, two-step reaction yield 7%).
  • Comparative Example 1 was obtained by referring to the synthesis method of compound 136 described in patent WO2019209757.
  • ESI-MS (m/z): 447.2[M+H] + ;
  • Comparative Example 2 was obtained by referring to the synthesis method of compound 54 described in patent WO2019209757.
  • ESI-MS (m/z): 409.8[M+H] + ;
  • Test Example 1 Construction of Colo205-LUC-TCF/LEF-M1 reporter cell line
  • the Colo205 cell line (Cell Bank of the Chinese Academy of Sciences, Cat#TCHu102) was purchased from the Cell Bank of the Chinese Academy of Sciences. After expansion and subculture, in the exponential growth phase of the cells, the method of transfection with lipo3000 liposomes was transfected with TCF/LEF transcription factors Driven luciferase reporter plasmid (Promega). The plasmid carries a resistance gene for resistance screening. Transfection was carried out in 10 cm culture dishes using conventional complete medium without resistance. After 2 days, the medium with resistance was replaced, and the culture was continued. After that, the resistant medium was replaced every 2 days, and the suspended cells were discarded. The original medium was centrifuged to remove cells and debris and retained as an adaptive medium.
  • the cells When the cells covered the culture dish, the cells were digested, counted, and passaged in a 96-well plate, so that the average number of cells contained in each well was 1.5/well, and the adaptive medium was used for passage. The rest of the cells were frozen. After subculture, culture for 4 hours to allow the cells to adhere to the wall, and then observe the number of cells in each well under a microscope. Wells with only 1 cell per well were labeled as monoclonal wells. Afterwards, normal culture was performed, and the culture medium was replaced every 2 days, and observed. There are holes where the monoclonal cells continue to grow in the early stage, and they are labeled twice, and can be replaced with normal resistant medium.
  • a monoclonal well When a monoclonal well is overgrown with a 96-well plate, it is digested and passaged to a 24-well culture plate. After the 24-well plate is overgrown, it is passaged to a 96-well plate and a 6-well plate.
  • the cells in a 96-well plate are at least 6 wells, of which 3 wells were added with known Wnt inhibitors, and the other 3 wells were not treated. After 24 hours, the cells in the 96-well plate were added with a fluorescence detection reagent to detect the fluorescence intensity. Cell lines with fluorescent expression when not treated and decreased fluorescent light after inhibition were selected and further cultured.
  • Colo205-LUC-TCF/LEF-M1 cell line is one of the above screened cell lines, its growth curve, cell shape, and cell growth state are similar to those of the original Colo205 cells, and the fluorescence signals of inhibitor treatment and no treatment
  • the ratio is the largest among all cell lines, and the ratio can reach 4-5 times when inhibited at 4 hours, which is completely suitable for the screening of Wnt inhibitors in the later stage.
  • Test Example 2 Detection of compound's inhibitory ability on Colo205-LUC-TCF/LEF-M1 reporter cell line
  • the Colo205-LUC-TCF/LEF-M1 cell line is a reporter tool cell stably transfected with the pGL4.49-LUC2-TCF/LEF vector.
  • the ⁇ -catenin Wnt pathway is continuously activated. After adding the inhibitor, the Wnt pathway is inhibited.
  • the expression of firefly luciferase regulated by the TCF/LEF cis-element decreased, and after adding the detection substrate, the detected light signal decreased accordingly, so as to detect the inhibitory effect of the compound.
  • a 96-well cell culture plate To a 96-well cell culture plate, add 100uL of the compound with a maximum concentration of 20uM to each well, and make a 3-fold serial dilution of the compound concentration. Then inoculate 10,000 colo205 cells stably transfected with the reporter gene and 100uL medium into each well, and make corresponding positive and negative control wells at the same time. Put the cells in a 5% CO2 cell incubator, culture at 37°C for 4 hours, remove the culture medium after 4 hours, add 100uL of reagent (Promega) containing the corresponding firefly luciferase substrate to each well, and measure the luciferase reporter gene activity. Luminescence intensities were read with SpectraMax in full wavelength mode.
  • Test Example 3 Proliferation Inhibitory Test of Compounds on Wnt Mutant Cell Lines (Colo205, H929, HepG2 and DU4475) and Non-Wnt Mutant Cell Lines (RKO)
  • the cell lines used in the experiment are Colo205, H929, HepG2 and DU4475 cell lines whose Wnt pathway is continuously activated, and whose proliferation is Wnt pathway-dependent; while the Wnt pathway is not activated under normal circumstances, and the proliferation is independent of Wnt pathway RKO
  • the cell line is used as a control cell line, and it is judged that the inhibitory effect of the compound of the present invention on Wnt-dependent proliferation is not caused by other non-specific toxicity.
  • Colo205, H929, DU4475, HepG2, and RKO cell lines cultured in their respective culture media were treated during the logarithmic growth phase, and the cells were collected to prepare a uniform cell suspension of known concentration, and then transferred to a 96-well cell culture plate Add cell suspension to each well so that each well contains 1000-4000 cells. Put it into a 5% CO2 incubator and incubate at 37°C for 20-24h. On the second day, the fully dissolved, 3-fold serially diluted compound was added to each cell culture well, so that the final maximum concentration in the cell culture well was 10 uM, and the culture was continued for 96 hours. In this test, Promega's cell viability detection test is used for detection.
  • the detection instrument is SpectraMax, full wavelength mode.
  • the wells only added with DMSO were used as positive control wells, and the wells that were not inoculated with cells were used as negative control wells.
  • the IC50 values of each compound for the proliferation inhibition of Wnt sustained activation or proliferation-dependent cells, and for Wnt-inactive or proliferation-independent cells were calculated.
  • the IC50 value of cell proliferation inhibition was used to evaluate the inhibitory effect of the compound on the Wnt pathway and the toxic effect on normal cells (Table 2).
  • Test Example 4 The tumor growth inhibition test of Compound 1 on the Colo205 mouse Xenograft model
  • mice Female BALB/c Nude nude mice were subcutaneously inoculated with human colon cancer cell Colo205, and the Colo205 BALB/c Nude nude mouse xenograft model was established. After the tumor grew to an average tumor volume of about 80 mm3, the tumor-bearing mice were randomly divided into 3 groups according to the tumor volume: solvent treatment control group, 3 mg/kg compound 1 group and 10 mg/kg control group 1. Compound 1 and Control Example 1 were administered orally, once a day, and the administration cycle was 14 days. The tumor volume was measured every other day, and the body weight and tumor volume were measured on Day 14 (Table 3 and Figure 1).

Landscapes

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

Abstract

L'invention concerne un composé représenté par la structure de formule (I) et ayant une excellente activité inhibitrice de la voie Wnt, ou un sel pharmaceutiquement acceptable, un dérivé isotopique, ou un stéréoisomère de celui-ci. L'invention concerne en outre un procédé de préparation du composé et son utilisation dans la prévention et/ou le traitement d'un cancer, d'une tumeur, d'une maladie inflammatoire, d'une maladie auto-immune ou d'une maladie à médiation immunitaire.
PCT/CN2023/073533 2022-01-29 2023-01-28 Composé inhibiteur de la voie wnt WO2023143501A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202380009996.XA CN116964053A (zh) 2022-01-29 2023-01-28 一种Wnt通路抑制剂化合物

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210114025.9 2022-01-29
CN202210114025 2022-01-29

Publications (1)

Publication Number Publication Date
WO2023143501A1 true WO2023143501A1 (fr) 2023-08-03

Family

ID=87470624

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/073533 WO2023143501A1 (fr) 2022-01-29 2023-01-28 Composé inhibiteur de la voie wnt

Country Status (3)

Country Link
CN (1) CN116964053A (fr)
TW (1) TW202334157A (fr)
WO (1) WO2023143501A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024022365A1 (fr) * 2022-07-28 2024-02-01 杭州阿诺生物医药科技有限公司 Composé inhibiteur de la voie wnt

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013093508A2 (fr) * 2011-12-22 2013-06-27 Oslo University Hospital Hf Inhibiteurs de la voie wnt
CN104379583A (zh) * 2012-06-15 2015-02-25 广州源生医药科技有限公司 作为wnt信号传导抑制剂的化合物、组合物及其应用
CN106458983A (zh) * 2014-03-20 2017-02-22 拜耳制药股份公司 新型化合物
CN109310690A (zh) * 2016-04-27 2019-02-05 萨穆梅德有限公司 异喹啉-3-基甲酰胺类及其制备和其用途
WO2020125759A1 (fr) * 2018-12-21 2020-06-25 汇瀚医疗科技有限公司 Composé en tant qu'inhibiteur de la voie de signalisation wnt et son utilisation médicale
CN112218865A (zh) * 2018-04-24 2021-01-12 沃泰克斯药物股份有限公司 喋啶酮化合物及其用途

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013093508A2 (fr) * 2011-12-22 2013-06-27 Oslo University Hospital Hf Inhibiteurs de la voie wnt
CN104379583A (zh) * 2012-06-15 2015-02-25 广州源生医药科技有限公司 作为wnt信号传导抑制剂的化合物、组合物及其应用
CN106458983A (zh) * 2014-03-20 2017-02-22 拜耳制药股份公司 新型化合物
CN109310690A (zh) * 2016-04-27 2019-02-05 萨穆梅德有限公司 异喹啉-3-基甲酰胺类及其制备和其用途
CN112218865A (zh) * 2018-04-24 2021-01-12 沃泰克斯药物股份有限公司 喋啶酮化合物及其用途
WO2020125759A1 (fr) * 2018-12-21 2020-06-25 汇瀚医疗科技有限公司 Composé en tant qu'inhibiteur de la voie de signalisation wnt et son utilisation médicale

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ASHIHARA EISHI, TAKADA TETSUYA, MAEKAWA TAIRA: "Targeting the canonical Wnt/β‐catenin pathway in hematological malignancies", CANCER SCIENCE, JAPANESE CANCER ASSOCIATION, TOKYO, JP, vol. 106, no. 6, 1 June 2015 (2015-06-01), JP , pages 665 - 671, XP093080992, ISSN: 1347-9032, DOI: 10.1111/cas.12655 *
LIU ZHIQING, WANG PINGYUAN, WOLD ERIC A., SONG QIAOLING, ZHAO CHENYANG, WANG CHANGYUN, ZHOU JIA: "Small-Molecule Inhibitors Targeting the Canonical WNT Signaling Pathway for the Treatment of Cancer", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 64, no. 8, 22 April 2021 (2021-04-22), US , pages 4257 - 4288, XP093080990, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.0c01799 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024022365A1 (fr) * 2022-07-28 2024-02-01 杭州阿诺生物医药科技有限公司 Composé inhibiteur de la voie wnt

Also Published As

Publication number Publication date
TW202334157A (zh) 2023-09-01
CN116964053A (zh) 2023-10-27

Similar Documents

Publication Publication Date Title
CN107011348B (zh) 作为btk活性的抑制剂的杂芳基吡啶酮和氮杂-吡啶酮化合物
TWI723511B (zh) 一種高活性sting蛋白激動劑化合物
WO2022089454A1 (fr) Composé inhibiteur de la voie wnt à haute activité
TWI716976B (zh) 高活性sting蛋白激動劑
WO2019158070A1 (fr) Antagoniste du récepteur a2a et/ou a2b
WO2019222521A1 (fr) Inhibiteurs de cdk pour le traitement de troubles néoplasiques
EP4153597A1 (fr) Composés inhibiteurs de kinase dépendant de la cycline pour le traitement d'affections médicales
WO2023143501A1 (fr) Composé inhibiteur de la voie wnt
WO2022089398A1 (fr) Inhibiteur de kinase hpk1 à haute activité
JP7436630B2 (ja) アデノシン受容体拮抗薬
WO2021129584A1 (fr) Composé antagoniste de pd-l1
WO2020169058A1 (fr) Composé antagoniste de pd-l1
WO2024022365A1 (fr) Composé inhibiteur de la voie wnt
WO2023143546A1 (fr) Composés inhibiteurs de la voie wnt
LU505117B1 (en) A pan-KRAS inhibitor compound
WO2023005894A1 (fr) Composé inhibiteur pour la voie wnt
WO2024060966A1 (fr) Composé inhibiteur de pan-kras
WO2023025225A1 (fr) Agent de dégradation de la cycline k
TWI810547B (zh) Pd-l1拮抗劑化合物
WO2024104364A1 (fr) Composé inhibiteur de pan-kras
TW202304906A (zh) 高活性hpk1激酶抑制劑
WO2020206034A1 (fr) Composés d'inhibition de cycle cellulaire pour le traitement de troubles médicaux
WO2022199561A1 (fr) Composé inhibiteur de la kinase hpk1
CN117396482A (zh) 一种Wnt通路抑制剂化合物

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 202380009996.X

Country of ref document: CN

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

Ref document number: 23746378

Country of ref document: EP

Kind code of ref document: A1