[go: up one dir, main page]

WO2024255412A1 - Spiro heterocyclic compound and use thereof - Google Patents

Spiro heterocyclic compound and use thereof Download PDF

Info

Publication number
WO2024255412A1
WO2024255412A1 PCT/CN2024/087019 CN2024087019W WO2024255412A1 WO 2024255412 A1 WO2024255412 A1 WO 2024255412A1 CN 2024087019 W CN2024087019 W CN 2024087019W WO 2024255412 A1 WO2024255412 A1 WO 2024255412A1
Authority
WO
WIPO (PCT)
Prior art keywords
mmol
alkyl
mixture
substituted
deuterated
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/CN2024/087019
Other languages
French (fr)
Chinese (zh)
Inventor
卿远辉
何其捷
谭慧晨
吴少槟
贺耀南
邓心兰
翁运幄
温才雄
封巧
林明生
刘斌
朱俊玲
刘怡婷
钱长庚
蔡雄
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bebetter Med Inc
Original Assignee
Bebetter Med Inc
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 Bebetter Med Inc filed Critical Bebetter Med Inc
Priority to CN202480000825.5A priority Critical patent/CN118510780A/en
Publication of WO2024255412A1 publication Critical patent/WO2024255412A1/en
Anticipated expiration legal-status Critical
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/438The ring being spiro-condensed with carbocyclic or heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • 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
    • C07D471/12Heterocyclic 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 three hetero rings
    • C07D471/20Spiro-condensed systems

Definitions

  • the present invention relates to the technical field of chemical medicine, and in particular to a spiro heterocyclic compound and application thereof.
  • MCR Melanocortin receptors
  • GPCR G-protein coupled receptors
  • M1R pigmentation
  • M2R adrenal function
  • M1R/MC3R immune function
  • M5R sebaceous gland activity
  • MC4R is a family A G protein-coupled receptor (GPCR) regulated by endogenous MC4R agonists and inverse agonist/antagonist peptides. Its signaling has physiological effects in regulating energy homeostasis, cachexia, cardiovascular function, glycolipid homeostasis, reproductive and sexual function, brain inflammation and anxiety. Inhibition or antagonism of MC4R signaling leads to increased appetite and food intake (Endocr Rev. 2010 Aug; 31(4): 506-43).
  • GPCR G protein-coupled receptor
  • MC4R antagonists have been reported, which can reverse or prevent appetite and weight loss in preclinical models of cancer, chronic kidney disease, age-related cachexia or anorexia (Biochim. Biophys. Acta. Mol. Basis. Dis. 2017, 1863 (10), 2414–2435; PLoS One 2009, 4 (3), e4774; J. Med. Chem. 2004, 47 (7), 1602-1604).
  • TCMCB07 MC4R antagonist peptide
  • Cachexia is a devastating, multifactorial wasting syndrome characterized by loss of homeostatic control of energy and protein balance, involuntary weight loss, and loss of skeletal muscle mass. Cachexia is highly associated with mortality from multiple malignant cancers (e.g., pancreatic, esophageal, gastric, lung, liver, and colorectal cancers, which account for approximately half of cancer deaths worldwide) (Nat Rev Dis Primers. 2018 Jan 18;4:17105; http://www.who.int/mediacentre/factsheets/fs297/en/) and is also associated with further exacerbation of chronic non-malignant diseases.
  • malignant cancers e.g., pancreatic, esophageal, gastric, lung, liver, and colorectal cancers, which account for approximately half of cancer deaths worldwide
  • M4R melanocortin-4 receptor
  • the present invention provides a class of spiro heterocyclic compounds, which have the effect of antagonizing the activity of MC4 receptors and can be used to treat various diseases related to MC4 receptors.
  • the present invention includes the following technical solutions.
  • R 1 , R 2 , R 3 and R 4 are independently selected from the group consisting of H, deuterium, C 1 -C 6 alkyl, C 1 -C 6 deuterated alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, halogen-substituted C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkylmethyl, halogen, halogen-substituted C 1 -C 6 alkyl, hydroxy, hydroxy-substituted C 1 -C 6 alkyl, C 1 -C 6 alkoxy-substituted C 1 -C 6 alkyl, cyano, -OR, -N(R) 2 , -SR, -C(O)OR, -C(O)N(R) 2 , -C(O)R, -S(O)R, -S(
  • R is selected from the group consisting of: H, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkylmethyl, halogen-substituted C 1 -C 6 alkyl, hydroxy-substituted C 1 -C 6 alkyl, alkoxy-substituted C 1 -C 6 alkyl, amino-substituted C 1 -C 6 alkyl, alkylamino-substituted C 1 -C 6 alkyl;
  • R 5 and R 6 are independently selected from: H, deuterium, halogen
  • M is selected from: CH, N;
  • W is selected from: O, S, NR 7 ;
  • Q is selected from the group consisting of: CR 7 R 8 , NR 8 , C ⁇ O, O, S, or absent;
  • R7 and R8 are independently selected from the group consisting of: H, deuterium, halogen, C1 - C6 alkyl, C1 - C6 deuterated alkyl, C2 - C6 alkenyl, C2 - C6 Alkynyl, C 3 -C 8 cycloalkyl, halogen-substituted C 1 -C 6 alkyl, halogen-substituted C 3 -C 8 cycloalkyl, hydroxy, cyano, amino, C 1 -C 6 alkylamino; or R 8 and the substituents in A are linked to form a substituted or unsubstituted 5-7 membered carbocyclic or heterocyclic ring;
  • A is selected from: C 6 -C 10 membered aryl substituted or unsubstituted by R 9 and R 10 , 5-10 membered heteroaryl substituted or unsubstituted by R 9 and R 10, 7-10 membered benzoheterocyclyl substituted or unsubstituted by R 9 and R 10 ;
  • Each R9 and R10 is independently selected from the group consisting of H, deuterium, halogen, C1 - C6 alkyl , C1 - C6 deuterated alkyl, C2 - C6 alkenyl, C2 - C6 alkynyl, cyano , C3 - C8 cycloalkyl, C3 - C8 deuterated cycloalkyl, C3- C8 cycloalkylC1- C6 alkyl, C1 - C6 alkylsulfonyl, C1- C6 alkylthio, C1-C6 deuterated alkylthio, C3-C8 cycloalkylthio , halogen -substituted C1 - C6 alkyl, C1 - C6 alkoxy, C1 - C6 deuterated alkoxy, halogen-substituted C1 - C6 alkoxy, C3 - C8 cycloal
  • A1 is selected from: phenyl substituted or unsubstituted by R11 and R12 , 5-6 membered heteroaryl substituted or unsubstituted by R11 and R12 ;
  • Each of R 11 and R 12 is independently selected from the group consisting of: H, deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 deuterated alkyl, C 3 -C 8 cycloalkyl, halogen-substituted C 1 -C 6 alkyl, cyano, and C 1 -C 6 alkoxy.
  • A is selected from: naphthyl substituted or unsubstituted by R 9 and R 10 , 9-10 membered heteroaryl substituted or unsubstituted by R 9 and R 10 , 9-10 membered benzoheterocyclyl substituted or unsubstituted by R 9 and R 10 .
  • A is selected from:
  • the dotted line in the ring represents whether it is a C-C single bond or not;
  • X 1 , X 2 , and X 3 are independently selected from the group consisting of CR 9 , N.
  • A is selected from:
  • A is selected from: W is O.
  • each R 9 and R 10 is independently selected from the group consisting of H, deuterium, halogen, C 1 -C 3 alkyl, C 1 -C 3 deuterated alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, cyano, C 3 -C 6 cycloalkyl, C 3 -C 6 deuterated cycloalkyl, C 3 -C 6 cycloalkylmethyl, C 1 -C 3 alkylsulfonyl, C 1 -C 3 alkylthio, C 1 -C 3 deuterated alkylthio, C 3 -C 6 cycloalkylthio, halogen-substituted C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 1 -C 3 deuterated alkoxy, halogen-substituted C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyloxy
  • each R 9 and R 10 is independently selected from the group consisting of hydrogen, methoxy, fluorine, difluoromethyl, difluoromethoxy, cyano, methyl, ethynyl, carbamoyl, methylsulfonyl, cyclopropyloxy, methylthio, and chlorine.
  • A is selected from:
  • each R 11 and R 12 is independently selected from the group consisting of: H, deuterium, halogen, C 1 -C 3 alkyl, C 1 -C 3 deuterated alkyl, C 3 -C 6 cycloalkyl, halogen-substituted C 1 -C 3 alkyl, cyano, and C 1 -C 3 alkoxy.
  • A1 is selected from:
  • A1 is selected from:
  • W is selected from: O, S, NH, NCH 3 .
  • W is selected from: S, NR 7 ;
  • R7 in W is selected from the group consisting of: H, C1 - C6 alkyl, C1 - C6 deuterated alkyl, C3 - C8 cycloalkyl, halogen-substituted C1 - C6 alkyl, and halogen-substituted C3 - C8 cycloalkyl.
  • R7 and R8 are independently selected from: H, deuterium, halogen, C1 - C3 alkyl, C1 - C3 deuterated alkyl, C2 - C3 alkenyl, C2 -C3 alkynyl, C3 - C6 cycloalkyl, halogen-substituted C1-C3 alkyl, halogen-substituted C3-C6 cycloalkyl , hydroxyl , cyano, amino, C1 - C3 alkylamino; or R8 and the substituents in A are connected to form a substituted or unsubstituted 5-6 membered carbocyclic or heterocyclic ring.
  • R7 and R8 are independently selected from: H, deuterium, methyl, hydroxyl, cyano, vinyl, ethynyl, amino, deuterated methyl, or R8 and the substituent in A are connected to form a 5-6 membered carbocyclic or heterocyclic ring.
  • Q is selected from: -N(CH 3 )-,
  • R 4 is selected from: C 1 -C 6 alkyl, C 1 -C 6 deuterated alkyl, halogen-substituted C 1 -C 6 alkyl.
  • R 1 , R 2 , R 3 and R 4 are independently selected from the group consisting of: H, deuterium, C 1 -C 3 alkyl, C 1 -C 3 deuterated alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 6 cycloalkyl, halogen-substituted C 3 -C 6 cycloalkyl, C 3 -C 6 cycloalkylmethyl, halogen, halogen-substituted C 1 -C 3 alkyl, OH, hydroxy-substituted C 1 -C 3 alkyl, alkoxy-substituted C 1 -C 3 alkyl, and cyano.
  • R 1 is selected from: methyl, hydroxy, difluoromethyl, trifluoromethyl, cyano, chlorine, fluorine, vinyl, ethynyl, cyclopropyl, deuterated methyl.
  • R 2 is selected from: H, fluorine, methyl, chlorine, deuterium.
  • R 3 is selected from: H, fluorine.
  • R 4 is selected from: H, methyl, methoxymethyl, monofluoromethyl.
  • the spiro heterocyclic compound is selected from the following compounds:
  • the present invention also provides the use of the spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule, including the following technical solutions.
  • spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule in the preparation of a drug for preventing and/or treating diseases and/or symptoms associated with the MC4 receptor.
  • the diseases and/or symptoms associated with the MC4 receptor are selected from: diseases and/or symptoms associated with the MC4 receptor are selected from: cachexia (including cachexia associated with chronic diseases, cancer, acquired immune deficiency syndrome (AIDS), heart failure, chronic kidney disease (CKD) and the like); anorexia or anorexia nervosa (such as senile anorexia, anorexia associated with chemotherapy and radiotherapy); nausea; vomiting; weight loss (involuntary weight loss); sarcopenia; muscle atrophy; muscle weakness; weakness; bone diseases (such as bone loss); pain or neuropathic pain; anxiety (such as post-traumatic stress disorder, PTSD, etc.); sexual dysfunction; and inflammatory diseases (such as inflammatory diseases associated with cachexia, anorexia, sarcopenia, etc.), etc.
  • cachexia including cachexia associated with chronic diseases, cancer, acquired immune deficiency syndrome (AIDS), heart failure, chronic kidney disease (CKD) and the like
  • the present invention also provides a pharmaceutical composition for preventing and/or treating diseases and/or symptoms associated with MC4 receptor, including the following technical solutions.
  • a pharmaceutical composition for preventing and/or treating diseases and/or symptoms associated with MC4 receptors comprising an active ingredient and a pharmaceutically acceptable excipient and/or carrier, wherein the active ingredient comprises the spiroheterocyclic compound or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated product thereof or a prodrug molecule thereof.
  • the present invention also provides a method for preventing and/or treating diseases and/or symptoms associated with MC4 receptors, the method comprising: administering a safe and effective amount of the spiroheterocyclic compound of the present invention or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated substance thereof or a prodrug molecule thereof; and/or,
  • the prodrug molecule has the effect of antagonizing the activity of MC4 receptor and can be used to treat various diseases related to MC4 receptor, which has great application value.
  • FIG1 is a plasma drug-dose curve of Compound 100, Compound 104 and PF07258669 after oral administration to mice.
  • FIG2 is a drug-time curve of the brain tissue of Compound 100, Compound 104 and PF07258669 after oral administration to mice.
  • any variable e.g., R, etc.
  • its definition at each occurrence is independent of the definition at every other occurrence.
  • combinations of substituents and variables are permitted as long as such combinations render the compound stable.
  • Lines drawn from substituents into the ring system indicate that the indicated bond may be attached to any substitutable ring atom. If the ring system is polycyclic, it means that such bonds are attached only to any appropriate carbon atom of the adjacent ring. It is to be understood that one of ordinary skill in the art may select substituents and substitution patterns for the compounds of the invention to provide chemically stable compounds that can be readily synthesized from readily available raw materials by techniques in the art and the methods set forth below. If a substituent itself is substituted with more than one group, it is to be understood that these groups may be on the same carbon atom or on different carbon atoms as long as the structure is stable.
  • alkyl as used herein is intended to include branched and straight chain saturated aliphatic hydrocarbon groups having a specific number of carbon atoms.
  • C 1 -C 6 alkyl includes groups having 1, 2, 3, 4 , 5 or 6 carbon atoms in a straight or branched arrangement.
  • C 1 -C 6 alkyl specifically includes methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, isobutyl, pentyl, hexyl.
  • cycloalkyl refers to a monocyclic saturated aliphatic hydrocarbon group having a specific number of carbon atoms.
  • cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, etc.
  • alkoxy refers to a group in which an alkyl group is directly linked to oxygen, i.e., a group having an -O - alkyl structure, such as -OCH3 , -OCH2CH3 , -OCH2CH2CH3 , -O - CH2CH ( CH3 ) 2 , -OCH2CH2CH2CH3 , -O -CH( CH3 ) 2 , and the like.
  • heterocyclyl is a saturated or partially unsaturated monocyclic or polycyclic cyclic substituent (acyclic, bridged or spirocyclic) in which one or more ring atoms are selected from N, O or S(O)m (wherein m is an integer from 0 to 2) as a heteroatom, and the remaining ring atoms are carbon, for example: morpholinyl, piperidinyl, tetrahydropyrrolyl, pyrrolidinyl, dihydroimidazolyl, dihydroisoxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothioph
  • heteroaryl refers to an aromatic ring containing one or more heteroatoms selected from O, N or S.
  • Aryl includes, but is not limited to, quinolyl, pyrazolyl, pyrrolyl, thienyl, furanyl, pyridinyl, pyrimidinyl, pyrazinyl, triazolyl, imidazolyl, oxazolyl, isoxazolyl, pyridazinyl, benzofuranyl, benzothiophenyl, benzoxazole, indolyl, and the like; "heteroaryl” is also understood to include the N-oxide derivative of any heteroaryl containing nitrogen.
  • substituted refers to the replacement of hydrogen radicals in a particular structure with the radical of a specified substituent.
  • halo or halo as used herein refers to chlorine, fluorine, bromine and iodine.
  • alkyl, cycloalkyl, aryl, heteroaryl and heterocycloalkyl substituents may be unsubstituted or substituted.
  • C1-C6 alkyl may be substituted with one, two or three substituents selected from OH, halogen, alkoxy, dialkylamino or heterocyclic radicals such as morpholinyl, piperidinyl, etc.
  • the present invention includes free forms of compounds of formula I, as well as pharmaceutically acceptable salts and stereoisomers thereof.
  • Some specific exemplary compounds of the present invention are amine compounds in non-salt form, i.e., "free forms".
  • Pharmaceutically acceptable salts of the present invention can be synthesized from compounds of the present invention containing a basic or acidic moiety by conventional chemical methods. Typically, salts of basic compounds are prepared by ion exchange chromatography or by reacting a free base with a stoichiometric amount or excess of an inorganic or organic acid in the desired salt form in an appropriate solvent or a combination of multiple solvents. Similarly, salts of acidic compounds are formed by reacting with an appropriate inorganic or organic base.
  • the pharmaceutically acceptable salts of the compounds of this invention include conventional non-toxic salts of the compounds of this invention formed by reacting alkaline compounds of this invention with inorganic or organic acids.
  • conventional non-toxic salts include salts derived from inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, phosphoric acid, nitric acid, etc., and also include salts prepared from organic acids such as acetic acid, propionic acid, succinic acid, glycolic acid, stearic acid, lactic acid, malic acid, tartaric acid, citric acid, ascorbic acid, pamoic acid, maleic acid, hydroxymaleic acid, phenylacetic acid, glutamic acid, benzoic acid, salicylic acid, p-aminobenzenesulfonic acid, 2-acetoxy-benzoic acid, fumaric acid, toluenesulfonic acid, methanesulfonic acid, ethanedis
  • suitable "pharmaceutically acceptable salts” refer to salts prepared from pharmaceutically acceptable non-toxic bases including inorganic bases and organic bases.
  • Salts derived from inorganic bases include aluminum salts, ammonium salts, calcium salts, copper salts, iron salts, ferrous salts, lithium salts, magnesium salts, manganic salts, manganous salts, potassium salts, sodium salts, zinc salts, etc. Ammonium salts, calcium salts, magnesium salts, potassium salts and sodium salts are particularly preferred.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases including salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins such as arginine, betaine, caffeine, choline, N,N'-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, aminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucosamine, glucosamine, histidine, hydroxocobalamin, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, guaiac, polyamine resins, procaine, purine, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.
  • stereoisomers of the present invention are, i.e. (depending on their structure) as enantiomers, diastereomers, syn-/anti-isomers, cis-/trans-isomers, epimers and (E)-/(Z)-isomers.
  • the compounds of formula I can be used in the context of the present invention in the form of pure stereoisomers or in the form of any mixture of stereoisomers, in the latter case the racemates are preferred.
  • the present invention also provides a pharmaceutical composition for preventing and/or treating diseases and/or symptoms associated with MC4 receptors, which comprises an active ingredient within a safe and effective amount range and a pharmaceutically acceptable adjuvant.
  • active ingredient refers to the compound of formula I described in the present invention, or its stereoisomer, or its pharmaceutically acceptable salt, or its solvate, or its prodrug molecule, or its deuteride, or its tritiated product.
  • Safety and effective amount means that the amount of active ingredient is sufficient to significantly improve the condition without causing serious side effects.
  • a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, wherein the dosage during administration is a pharmaceutically effective dosage, and for a person weighing 60 kg, the daily dosage is usually 1 to 2000 mg, preferably 20 to 500 mg.
  • the specific dosage should also take into account factors such as the route of administration and the health status of the patient, which are all within the skill of a skilled physician.
  • “Pharmaceutically acceptable excipients” refer to: one or more compatible solid or liquid fillers or gel substances, which are suitable for human use and must be sufficiently pure and sufficiently low in toxicity.
  • composition means that the components in the composition can be blended with the active ingredient of the present invention and with each other without significantly reducing the efficacy of the active ingredient.
  • Examples of pharmaceutically acceptable excipients include cellulose and its derivatives (such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers (such as ), wetting agents (such as sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • cellulose and its derivatives such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.
  • gelatin such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.
  • the compound of formula I of the present invention can form a complex with a macromolecular compound or a polymer through a non-bonding reaction.
  • the compound of formula I of the present invention as a small molecule can also be connected to a macromolecular compound or a polymer through a chemical bond.
  • the macromolecular compound can be a biological macromolecule such as a high polysaccharide, protein, nucleic acid, polypeptide, etc.
  • compositions of the present invention there is no particular limitation on the administration of the active ingredient or pharmaceutical composition of the present invention.
  • Representative administration methods include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), etc.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active ingredient is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or Dicalcium phosphate, or in combination with:
  • fillers or extenders for example, starch, lactose, sucrose, glucose, mannitol and silicic acid;
  • binders for example, hydroxymethylcellulose, alginate, gelatin, polyvinyl pyrrolidone, sucrose and gum arabic;
  • humectants for example, glycerin
  • disintegrants for example, agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate;
  • a buffering solvent such as paraffin
  • absorption accelerators for example, quaternary ammonium compounds
  • adsorbents for example, kaolin
  • Lubricants for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate, or mixtures thereof.
  • the dosage form may also contain a buffering agent.
  • the solid dosage forms can also be prepared using coatings and shell materials, such as enteric coatings and other materials known in the art. They can contain opacifying agents, and the release of the active ingredient in such compositions can be delayed in a certain part of the digestive tract.
  • coatings and shell materials such as enteric coatings and other materials known in the art. They can contain opacifying agents, and the release of the active ingredient in such compositions can be delayed in a certain part of the digestive tract.
  • embedding components that can be used are polymeric substances and waxes.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • the liquid dosage form may contain inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butylene glycol, dimethylformamide and oils, in particular cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances.
  • the composition may also contain adjuvants, such as wetting agents, emulsifiers and suspending agents, sweeteners, flavoring agents and spices.
  • suspensions may contain suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methanol and agar, or mixtures of these substances.
  • suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methanol and agar, or mixtures of these substances.
  • compositions for parenteral injection may include physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
  • Step 1a Preparation of 1-benzyl-3-((trimethylsilyl)ethynyl)pyrrolidin-3-ol (Compound 0102-1): Under nitrogen protection, n-butyl lithium (25.11 ml, 62.78 mmol, 1.1 eq) was added dropwise to a solution of ethynyltrimethylsilane (6.73 g, 68.48 mmol, 1.2 eq) in tetrahydrofuran (50 ml) at -70°C, and the mixture was stirred at -70°C for 1.0 hour.
  • ethynyltrimethylsilane 6.73 g, 68.48 mmol, 1.2 eq
  • Step 1b Preparation of 1-benzyl-3-ethynylpyrrolidin-3-ol (Compound 0103-1): Under nitrogen protection, potassium carbonate (12.24 g, 88.72 mmol, 2.0 eq.) was added to a solution of 1-benzyl-3-((trimethylsilyl)ethynyl)pyrrolidin-3-ol (0102-1) (12.11 g, 44.36 mmol, 1.0 eq.) in methanol (100 ml), and the mixture was stirred at room temperature for 1.5 hours. The solvent was removed under reduced pressure, the residue was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (ethyl acetate) to obtain a yellow oily product 1-benzyl-3-ethynylpyrrolidin-3-ol (7.65 g, yield: 85.68%).
  • Step 1c Preparation of 1-benzyl-3-ethynylpyrrolidin-3-yl acetate (Compound 0104-1): Under nitrogen protection, acetyl chloride (5.08 g, 64.7 mmol, 1.0 equiv) was added to a solution of 1-benzyl-3-ethynylpyrrolidin-3-ol (0103-1) (7.65 g, 38.05 mmol, 1.0 equiv) and N,N-diisopropylethylamine (9.82 g, 76.1 mmol, 2.0 equiv) in dichloromethane (100 ml) at 0°C. The mixture was stirred at 0°C for 0.5 hours.
  • reaction solution was quenched with water and extracted with dichloromethane.
  • organic phase was dried and concentrated under reduced pressure.
  • Step 1d Preparation of 1-benzyl-3-ethynyl-N-(4-methoxybenzyl)pyrrolidin-3-amine (Compound 0105-1): Under nitrogen protection, a solution of 1-benzyl-3-ethynylpyrrolidin-3-yl acetate (0104-1) (8.92 g, 36.66 mmol, 1.0 eq.) in tetrahydrofuran (18 ml) was added to a solution of 4-methoxybenzylamine (10.06 g, 73.32 mmol, 2.0 eq.) and cuprous chloride (363 mg, 3.67 mmol, 0.1 eq.) in tetrahydrofuran (60 ml) at 70° C., and the mixture was stirred at 70° C.
  • Step 1e Preparation of 2-chloro-3-iodo-6-methylpyridine (Compound 0107-1): To a mixture of 2-chloro-6-methylpyridin-3-amine (0106-1) (25.0 g, 175.33 mmol, 1.0 eq) in 6M dilute hydrochloric acid (175 ml) was added dropwise a solution of sodium nitrite (14.52 g, 210.39 mmol, 1.2 eq) in water (50 ml) at 0°C. The mixture was stirred at 0°C for 0.5 h. A solution of potassium iodide (58.18 g, 350.46 mmol, 2.0 eq) in water (100 ml) was added dropwise.
  • Step 1f Preparation of 1-benzyl-3-((2-chloro-6-methylpyridin-3-yl)ethynyl)-N-(4-methoxybenzyl)pyrrolidin-3-amine
  • Compound 0108-1 To a mixture of 2-chloro-3-iodo-6-methylpyridine (0107-1) (35.78 g, 141.43 mmol, 1.0 eq), bistriphenylphosphine palladium dichloride (4.96 g, 7.1 mmol, 0.05 eq) and cuprous iodide (1.35 g, 7.1 mmol, 0.05 eq) in triethylamine (220 ml) was added 1-benzyl-3-ethynyl-N-(4-methoxybenzyl)pyrrolidin-3-amine (0105-1) (45.25 g, 141.43 mmol, 1.0 eq).
  • Step 1g Preparation of 1-benzyl-3-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-N-(4-methoxybenzyl)pyrrolidin-3-amine (Compound 0109-1): Platinum dioxide (4.17 g, 18.38 mmol, 0.15 eq) was added to a mixture of 1-benzyl-3-((2-chloro-6-methylpyridin-3-yl)ethynyl)-N-(4-methoxybenzyl)pyrrolidin-3-amine (0108-1) (54.66 g, 122.56 mmol, 1.0 eq) in methanol (300 ml).
  • Step 1h Preparation of 1-benzyl-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'H-spiro(pyrrolidine-3,2'-[1,8]naphthyridine (Compound 0110-1): To a mixture of 1-benzyl-3-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-N-(4-methoxybenzyl)pyrrolidin-3-amine (0109-1) (42.18 g, 93.94 mmol, 1.0 eq), palladium acetate (1.05 g, 4.70 mmol, 0.05 eq) and RuPhos (4.38 g, 1.0 mmol, 0.1 eq) in dioxane (420 ml) was added sodium tert-butoxide (18.04 g, 1 87.88 mmol, 2.0 eq.).
  • the mixture was heated to 115°C under a nitrogen atmosphere and reacted overnight.
  • the solvent was removed under reduced pressure.
  • the residue was diluted with water (200 ml).
  • the aqueous layer was extracted with ethyl acetate (150 ml ⁇ 3).
  • the combined organic layer was washed with saturated brine (100 ml ⁇ 1), dried over anhydrous sodium sulfate and concentrated.
  • Step 1i Preparation of 1-benzyl-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]
  • Compound 0111-1 To a mixture of 1-benzyl-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] (0110-1) (21.0 g, 50.85 mmol, 1.0 equiv) in dichloromethane (150 mL) was added dropwise trifluoroacetic acid (57.98 g, 508.5 mmol, 10.0 equiv) at 0°C.
  • Step 1j Preparation of tert-butyl 7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0112-1): Palladium hydroxide/carbon (10%, 3.73 g) was added to a mixture of 1-benzyl-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] (0111-1) (14.9 g, 50.85 mmol, 1.0 equiv) and di-tert-butyl dicarbonate (22.2 g, 101.7 mmol, 2.0 equiv) in tetrahydrofuran (150 ml).
  • Step 1k Preparation of (S)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0113-1) and (R)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0114-1): The enantiomers of 7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0112-1) were separated by supercritical fluid chromatography (chromatographic column: ChiralPak IC, 250 ⁇ 30 mm ID, 10 ⁇ m; mobile phase: A is carbon dioxide
  • the first eluting enantiomer is (R)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (compound 0114-1) (6.32 g, yield: 47%)
  • the second eluting enantiomer is (S)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (compound 0113-1) (6.29 g, yield: 47%).
  • Step 11 Preparation of (S)-6'-bromo-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester
  • Compound 0116-1 To a mixture of (S)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0113-1) (6.29 g, 20.76 mmol, 1.0 eq) in dichloromethane (60 mL) was added dibromohydantoin (2.97 g, 10.38 mmol, 0.5 eq) at 0°C.
  • Step 1m Preparation of (S)-(1-(tert-Butyloxycarbonyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (Compound 0118-1): to (S)-6'-bromo-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0116-1) (5.75 g, 15.05 mmol, 1.0 equiv), Xphos-Pd-G2 (237 mg, 0.30 mmol, 0.02 eq.), Xphos (215 mg, 0.45 mmol, 0.03 eq.), potassium acetate (3.69 g, 37.63 mmol, 2.5 eq.) and sodium tert
  • Step 1n Preparation of (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0120-1): To (S)-(1-(tert-butyloxycarbonyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (0118-1) (3.58 g, 10.
  • Step 1o Preparation of (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Intermediate 0122-1): A mixture of (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0120-1) (1.07 g, 3.08 mmol, 1.0 eq) in methanolic hydrogen chloride solution (4M solution, 10 ml) was stirred at room temperature for 3.5 hours.
  • methanolic hydrogen chloride solution (4M solution, 10 ml
  • Step 1p Preparation of 5-fluoro-4-iodo-2-methoxypyridine (Compound 0202-1): To a mixture of 2,5-difluoro-4-iodopyridine (Compound 0201-1) (25.0 g, 103.74 mmol, 1.0 eq.) in methanol (70 ml) was added sodium methoxide (5.4 M in methanol, 57.6 ml, 311.23 mmol, 3.0 eq.) at 0°C. The mixture was heated to 60°C under nitrogen atmosphere and stirred for 1.5 hours. The solvent was removed under reduced pressure.
  • Step 1q Preparation of 5-fluoro-2-methoxy-N-(4-methoxybenzyl)-N-methylpyridin-4-amine (compound 0203-1): To a mixture of 5-fluoro-4-iodo-2-methoxypyridine (0202-1) (1.0 g, 3.95 mmol, 1.0 eq), tris(dibenzylideneacetone)palladium (109 mg, 0.119 mmol, 0.03 eq), RuPhos (111 mg, 0.237 mmol, 0.06 eq) and sodium tert-butoxide (1.14 g, 11.85 mmol, 3.0 eq) in toluene (15 ml) was added 1-(4-methoxyphenyl)-N-methylmethanamine (0.72 g, 4.74 mmol, 1.2 eq).
  • Step 1s Preparation of (S)-N-(5-fluoro-2-methoxypyridin-4-yl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxamide
  • Compound 1 To a mixture of 5-fluoro-2-methoxy-N-methylpyridin-4-amine (0204-1) (20 mg, 0.13 mmol, 2.0 eq) and triethylamine (53 mg, 0.52 mmol, 8.0 eq) in dichloromethane (2.5 ml) was added triphosgene (19 mg, 0.065 mmol, 1.0 eq) at 0°C.
  • the mixture was warmed to room temperature and stirred for 1.5 hours.
  • the mixture was diluted with water (15 ml) and then extracted with ethyl acetate (10 ml x 2).
  • the combined organic layer was washed with saturated brine (15 ml x 1), dried over anhydrous sodium sulfate and concentrated.
  • Step 3a Preparation of 3,4-difluoro-N-methylaniline (Compound 0204-3):
  • Step 3b Preparation of (S)-N-(3,4-difluorophenyl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxamide
  • Compound 3 Under nitrogen protection, triphosgene (37.4 mg, 0.126 mmol, 0.9 equiv) was added to a solution of 3,4-difluoro-N-methylaniline (0204-3) (20 mg, 0.14 mmol, 1.0 equiv) and triethylamine (56 mg, 0.56 mmol, 4.0 equiv) in dichloromethane (5 ml), and the mixture was stirred at room temperature for 1.5 hours.
  • reaction solution was diluted with water and extracted with dichloromethane (8 ml), the organic phase was dried, 7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-1) (49.4 mg, 0.14 mmol, 1.0 eq.), N,N-diisopropylethylamine (63.21 mg, 0.49 mmol, 3.5 eq.) were added to the above solution, and the mixture was stirred at room temperature for 0.5 hours.
  • Step 4a Preparation of dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)malonate (compound 0403-7): To a mixture of 5-fluoro-4-iodo-2-methoxypyridine (0202-1) (22.06 g, 87.19 mmol, 1.0 eq), cuprous iodide (1.66 g, 8.72 mmol, 0.1 eq), 2-picolinic acid (2.15 g, 17.44 mmol, 0.2 eq) and cesium carbonate (85.27 g, 261.57 mmol, 3.0 eq) in tetrahydrofuran (130 ml) was added dibenzyl malonate (34.70 g, 12.21 mmol, 1.4 eq).
  • Step 4b Preparation of dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)-2-methylmalonate (Compound 0404-7): To a mixture of dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)malonate (0403-7) (35.70 g, 87.19 mmol, 1.0 eq) and potassium carbonate (36.10 g, 261.57 mmol, 3.0 eq) in acetonitrile (80 ml) was added iodomethane (24.75 g, 174.38 mmol, 2.0 eq). The mixture was stirred at room temperature overnight. The solvent was removed under reduced pressure.
  • Step 4c Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (Compound 0405-7): To a mixture of 2-(5-fluoro-2-methoxypyridin-4-yl)-2-methylmalonic acid dibenzyl ester (0404-7) (24.5 g, 59.32 mmol, 1.0 eq.) in ethyl acetate (100 mL) was added palladium/carbon (10%, 4.0 g). The mixture was stirred at 55° C. under hydrogen balloon pressure for two days. The mixture was filtered. The filtrate was concentrated under reduced pressure.
  • Step 4d Preparation of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (compound 0406-7) and (S)-2-(-5-fluoro-2-methylpyridin-4-yl)propanoic acid (0407-7): Separation of the enantiomers of 2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0405-7) (8.86 g, 44.52 mmol) was carried out by supercritical fluid chromatography (column: ChiralPak IG, 250 ⁇ 30 mm I.D., 10 ⁇ m; mobile phase: A is carbon dioxide, B is methanol; eluent: 10% B; flow rate: 60 ml/min; back pressure: 100 bar; column Temperature: 38°C).
  • the first eluting enantiomer was (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0406-7) (3.75 g, yield: 42%), and the second eluting enantiomer was (S)-2-(-5-fluoro-2-methylpyridin-4-yl)propanoic acid (0407-7) (3.86 g, yield: 44%).
  • Step 4e (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro Preparation of [pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (compound 0413-7): To a mixture of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (compound 0406-7) (66 mg, 0.33 mmol, 1.0 eq) in acetonitrile (3 ml) were added imidazole trifluoromethanesulfonate (108 mg, 0.50 mmol, 1.5 eq) and carbonyldiimidazole (64 mg, 0.40 mmol, 1.2 eq).
  • Step 4f Preparation of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione
  • Compound 7 To a mixture of (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (compound 0413-7) (80 mg, 0.18 mmol, 1.0 eq.) in toluene (5 ml) was added Lawesson's reagent (85 mg, 0.21 mmol, 1.2 eq.).
  • the mixture was heated at 125°C in a microwave reactor for 40 minutes.
  • the mixture was diluted with water (20 ml) and then extracted with ethyl acetate (15 ml x 3).
  • the combined organic layers were washed with saturated brine (15 ml x 1), dried over anhydrous sodium sulfate and concentrated.
  • Step 5a Preparation of dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)-2-(methyl-d3)malonate (Compound 0404-57): Under nitrogen protection, to a mixed solution of dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)malonate (0403-7) (500 mg, 1.22 mmol, 1.0 eq.), potassium carbonate (507 mg, 3.67 mmol, 3.0 eq.) in N,N-dimethylformamide (5 ml) was added deuterated iodomethane (355 mg, 2.44 mmol, 2.0 eq.), and the mixture was stirred at room temperature for three hours.
  • deuterated iodomethane 355 mg, 2.44 mmol, 2.0 eq.
  • Step 5b Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)propionic acid-3,3,3-d3 acid (Compound 0405-57): Under hydrogen protection, palladium carbon (102 mg, 20% mass fraction) was added to a mixture of 2-(5-fluoro-2-methoxypyridin-4-yl)-2-(methyl-d3)malonate (Compound 0404-57) (510 mg, 1.19 mmol, 1.0 equivalent) in ethyl acetate (5 ml). The mixture was stirred at 55°C overnight. The mixture was filtered and the filtrate was concentrated under reduced pressure.
  • Step 5c Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7′-methyl-6′-(pyrimidin-2-yl)-3′,4′-dihydro-1′H-spiro[pyrrolidine-3,2′-[1,8]naphthyridine]-1-yl)propan-1-one-3,3,3-d3 (Compound 57 (0412-57)): 2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid-3,3,3-d3.
  • Step 7a Preparation of tert-butyl 3-hydroxy-3-((trimethylsilyl)ethynyl)pyrrolidine-1-carboxylate (Compound 0102-34): Under nitrogen protection, n-butyl lithium (9.5 ml, 23.8 mmol, 1.1 eq) was added dropwise to a solution of ethynyltrimethylsilane (2.55 g, 25.9 mmol, 1.2 eq) in tetrahydrofuran (40 ml) at -70°C, and the mixture was stirred at -70°C for 1.0 hour.
  • Step 7b Preparation of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (Compound 0103-34): Under nitrogen protection, potassium carbonate (5.80 g, 42.3 mmol, 2.0 eq.) was added to a solution of tert-butyl 3-hydroxy-3-((trimethylsilyl)ethynyl)pyrrolidine-1-carboxylate (0102-34) (6.00 g, 21.2 mmol, 1.0 eq.) in methanol (100 ml), and the mixture was stirred at room temperature for 1.5 hours.
  • potassium carbonate 5.80 g, 42.3 mmol, 2.0 eq.
  • Step 7c Preparation of tert-butyl 3-acetoxy-3-ethynyl-pyrrolidine-1-carboxylate (Compound 0104-34): Under nitrogen protection, acetyl chloride (1.98 g, 25.2 mmol, 1.2 eq) was added to a solution of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (0103-34) (4.43 g, 21.0 mmol, 1.0 eq) and N,N-diisopropylethylamine (4.06 g, 31.5 mmol, 1.5 eq) in dichloromethane (100 ml) at 0°C, and the mixture was stirred at 0°C for 1 hour.
  • reaction solution was quenched with water and extracted with dichloromethane.
  • organic phase was dried and concentrated under reduced pressure.
  • Step 7d Preparation of tert-butyl 3-ethynyl-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0105-34): Under nitrogen protection, to a solution of 4-methoxybenzylamine (3.07 g, 22.3 mmol, 2.0 equiv.) and cuprous chloride (111 mg, 1.1 mmol, 0.1 equiv.) in tetrahydrofuran (30 ml) at 70°C, a solution of tert-butyl 3-acetoxy-3-ethynyl-pyrrolidine-1-carboxylate (0104-34) (2.83 g, 11.2 mmol, 1.0 equiv.) in tetrahydrofuran (10 ml) was added, and the mixture was stirred at 70°C for 1 hour.
  • reaction solution was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure.
  • Step 7e Preparation of 2-chloro-3-iodo-6-(trifluoromethyl)pyridine (Compound 0107-34): To a mixture of 2-chloro-6-(trifluoromethyl)pyridin-3-amine (0106-34) (1.5 g, 7.63 mmol, 1.0 eq) and 6M dilute hydrochloric acid (7.6 mL, 45.78 mmol, 6.0 eq) in acetonitrile (5 mL) was added dropwise a solution of sodium nitrite (0.63 g, 9.16 mmol, 1.2 eq) in water (503 mL) at 0°C. The mixture was stirred at 0°C for 35 minutes.
  • Step 7f Preparation of tert-butyl 3-((2-chloro-6-(trifluoromethyl)pyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate
  • Compound 0108-314 To a mixture of 2-chloro-3-iodo-6-(trifluoromethyl)pyridine (0107-34) (1.0 g, 3.26 mmol, 1.08 equiv), bistriphenylphosphine palladium dichloride (106 mg, 0.15 mmol, 0.05 equiv) and cuprous iodide (29 mg, 0.15 mmol, 0.05 equiv) in triethylamine (10 ml) was added tert-butyl 3-ethynyl-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (0105-34) (1.0 g, 3.03 mmol,
  • Step 7g Preparation of tert-butyl 3-(2-(2-chloro-6-(trifluoromethyl)pyridin-3-yl)ethyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0109-34): To a mixture of tert-butyl 3-((2-chloro-6-(trifluoromethyl)pyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (0108-34) (1.33 g, 2.61 mmol, 1.0 eq) in methanol (10 mL) was added platinum dioxide (118 mg, 0.52 mmol, 0.2 eq).
  • Step 7h Preparation of tert-butyl 1'-(4-methoxybenzyl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0110-34): To a mixture of tert-butyl 3-(2-(2-chloro-6-(trifluoromethyl)pyridin-3-yl)ethyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (0109-34) (1.29 g, 2.51 mmol, 1.0 equiv), palladium acetate (28 mg, 0.13 mmol, 0.05 equiv) and RuPhos (117 mg, 0.25 mmol, 0.1 equiv) in dioxane (12 ml) was added.
  • Step 7i Preparation of tert-butyl 7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate
  • Compound 0112-34 A mixture of tert-butyl 1'-(4-methoxybenzyl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0110-34) (724 mg, 1.52 mmol, 1.0 eq) in methanolic hydrogen chloride solution (4M solution, 7.5 mL) was heated to 70°C for 2 hours.
  • methanolic hydrogen chloride solution (4M solution, 7.5 mL
  • Step 7j Preparation of tert-butyl 6'-bromo-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0115-34): To a mixture of tert-butyl 7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0112-34) (543 mg, 1.52 mmol, 1.0 eq) in dichloromethane (10 mL) was added dibromohydantoin (261 mg, 0.91 mmol, 0.6 eq) at 0°C.
  • Step 7k Preparation of (1-(tert-butyloxycarbonyl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (Compound 0117-34): To tert-butyl 6'-bromo-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0115-34) (250 mg, 0.57 mmol, 1.0 equiv.
  • Xphos-Pd-G2 (9 mg, 0.011 mmol, 0.02 eq), Xphos (8 mg, 0.017 mmol, 0.03 eq), potassium acetate (140 mg, 1.43 mmol, 2.5 eq) and sodium tert-butoxide (1.4 mg, 0.014 mmol, 0.025 eq) were added to a mixture of methanol (6 ml) and ethylene glycol (0.6 ml). Tetrahydroxydiboron (129 mg, 1.43 mmol, 2.5 eq) was added. The mixture was heated to 53°C under a nitrogen atmosphere for 10 hours. The solvent was removed under reduced pressure. The residue was diluted with water (20 ml).
  • Step 71 Preparation of tert-butyl 6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0119-34): To (1-(tert-butyloxycarbonyl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (0117-34) (151 mg, 0.38 mmol, 1.0 eq.), Xphos-Pd-G2 (9 mg, 0.011 mmol, 0.03 eq.), Xphos (5.2 mg, 0.011 mmol, 0.03 eq.) and sodium carbonate (120 mg, 1.13 mmol, 3.0 eq.) were
  • Step 7m Preparation of 6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0121-34): A mixture of tert-butyl 6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0119-34) (49 mg, 0.113 mmol, 1.0 eq) in methanolic hydrogen chloride solution (4M solution, 1.5 mL) was stirred at room temperature for 3.5 hours.
  • methanolic hydrogen chloride solution 4M solution, 1.5 mL
  • Step 7n Preparation of (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one
  • Compound 34 To a mixture of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (Compound 0406-7) (22.5 mg, 0.113 mmol, 1.0 eq) in acetonitrile (3 mL) were added imidazole triflate (37 mg, 0.17 mmol, 1.5 eq) and carbonyldiimidazole (22 mg, 0.14 mmol, 1.2 eq).
  • Step 8a Preparation of tert-butyl 3-((2,6-difluoropyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0108-37): To a mixture of 2,6-difluoro-3-iodopyridine (422 mg, 1.75 mmol, 1.05 eq), bistriphenylphosphine palladium dichloride (59 mg, 0.084 mmol, 0.05 eq) and cuprous iodide (16 mg, 0.084 mmol, 0.05 eq) in triethylamine (8 ml) was added tert-butyl 3-ethynyl-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0105-34) (550 mg, 1.67 mmol, 1.0 eq).
  • Step 8b Preparation of tert-butyl 7'-fluoro-1'-(4-methoxybenzyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0110-37): To a mixture of tert-butyl 3-((2,6-difluoropyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (0108-37) (600 mg, 1.35 mmol, 1.0 eq) in methanol (10 mL) was added platinum dioxide (62 mg, 0.27 mmol, 0.2 eq).
  • Step 8c Preparation of tert-butyl 7'-fluoro-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0112-37): To a mixture of tert-butyl 7'-fluoro-1'-(4-methoxybenzyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0110-37) (315 mg, 0.74 mmol, 1.0 eq) in dichloromethane (6 mL) was added trifluoroacetic acid (1.5 mL).
  • Step 8d Preparation of tert-butyl 7'-fluoro-6'-iodo-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0115-37): To a mixture of tert-butyl 7'-fluoro-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0112-37) (227 mg, 0.74 mmol, 1.0 equiv) in acetonitrile (3.5 mL) at 0°C was added N-iodosuccinimide (166 mg, 0.74 mmol, 1.0 equiv).
  • Step 8e Preparation of tert-butyl 7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0119-37): To tert-butyl 7'-fluoro-6'-iodo-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0115-37) (240 mg, 0.55 mmol, 1.0 equiv), Xphos- Pd-G3 (23 mg, 0.028 mmol, 0.05 eq), Xphos (13.3 mg, 0.028 mmol, 0.05 eq), cuprous iodide (10.5 mg, 0.055 mmol, 0.1 eq) and cesium fluoride (167
  • the mixture was heated to 100°C in a microwave reactor for 50 minutes.
  • the mixture was diluted with water (30 ml) and then extracted with ethyl acetate (10 ml x 3).
  • the combined organic layer was washed with saturated brine (20 ml x 1), dried over anhydrous sodium sulfate and concentrated.
  • Step 8f Preparation of 7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0121-37): A mixture of tert-butyl 7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0119-37) (42 mg, 0.11 mmol, 1.0 eq) in ethyl acetate solution of hydrogen chloride (4M solution, 2.5 mL) was stirred at room temperature for 2.5 hours.
  • hydrogen chloride 4M solution, 2.5 mL
  • Step 8g Preparation of (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 37): To (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (Compound 0406-7) (22 mg, 0.
  • N,N-diisopropylethylamine (0.5 ml, 2.87 mmol, 18 eq) was added to a toluene (10 ml) solution of ethyl 2-cyano-2-(5-fluoro-2-methoxypyridin-4-yl)acetate (43 mg, 0.16 mmol, 1.0 eq) and 7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0122-1) (50 mg, 0.18 mmol, 1.0 eq), and the reaction was stirred at 120° C.
  • reaction mixture was diluted with water and extracted with ethyl acetate (10 ml ⁇ 3), and the combined organic phase was washed with saturated brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure at 45° C.
  • Step 10a Preparation of dimethyl 2-(2-methoxy-5-(methylthio)pyridin-4-yl)malonate (Compound 0403-31): To a mixture of 6-methoxy-4-methylpyridin-3-amine (1 g, 7.2 mmol, 1.0 eq) in dimethyl disulfide (10 ml) was added tert-butyl nitrite (5.96 g, 57.9 mmol, 8.0 eq) and the mixture was stirred at 50°C for half an hour. The reaction solution was diluted with water, extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure to give a crude oil.
  • tert-butyl nitrite 5.96 g, 57.9 mmol, 8.0 eq
  • Step 10b Preparation of dimethyl 2-(2-methoxy-5-(methylthio)pyridin-4-yl)-2-methylmalonate (Compound 0404-31): Under nitrogen protection, iodomethane (145 mg, 1.0 mol, 1.0 eq.) was added to a mixture of dimethyl 2-(2-methoxy-5-(methylthio)pyridin-4-yl)malonate (0403-31) (290 mg, 1.0 mmol, 1.0 eq.) and cesium carbonate (501 mg, 1.5 mmol, 1.5 eq.) in N,N-dimethylformamide (5 ml), and the mixture was stirred at 35°C overnight.
  • iodomethane 145 mg, 1.0 mol, 1.0 eq.
  • cesium carbonate 501 mg, 1.5 mmol, 1.5 eq.
  • the reaction solution was diluted with ammonium chloride.
  • the mixture was diluted with ethyl acetate and extracted with ethyl acetate.
  • the organic phase was dried and concentrated under reduced pressure to obtain a crude oil.
  • LCMS (ESI): m/z 260 [M+1] + .
  • Step 10d Preparation of 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 31, 0412-31): To 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)propanoic acid (0405-31) (50 mg, 0.2 mmol, 1.05 equiv), 7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one 2-(7-azabenzotriazole)-N,N,N',N'-te
  • the reaction solution was diluted with water, extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure to obtain a crude oil.
  • Step 12a Preparation of 4-fluoro-2-iodo-5-methylphenol: A mixture of 4-fluoro-3-methylphenol (5.0 g, 39.6 mmol, 1.0 eq.), p-toluenesulfonic acid monohydrate (7.5 g, 39.6 mmol, 1.0 eq.) and N-iodosuccinimide (9.8 g, 43.6 mmol, 1.1 eq.) in acetonitrile (50 ml) was stirred at room temperature for 2 hours. The reaction solution was quenched with sodium sulfite solution and extracted with ethyl acetate.
  • Step 12b Preparation of 4-fluoro-5-methyl-2-((trimethylsilyl)ethynyl)phenol: To a mixture of 4-fluoro-2-iodo-5-methylphenol (4.19 g, 16.7 mmol, 1.0 eq), bistriphenylphosphine palladium dichloride (292 mg, 0.42 mmol, 0.025 eq) and cuprous iodide (79.3 mg, 0.42 mmol, 0.025 eq) in triethylamine/tetrahydrofuran (3.6 ml/5 ml) was added ethynyltrimethylsilane (8.2 g, 83 mmol, 5.0 eq) under nitrogen, and the mixture was stirred at 90° C.
  • Step 12c Preparation of 5-fluoro-6-methylbenzofuran: A solution of 4-fluoro-5-methyl-2-((trimethylsilyl)ethynyl)phenol (crude), cuprous iodide (3.59 g, 18.9 mmol, 1.0 eq.) and N,N-diisopropylethylamine (4.9 g, 37.7 mmol, 2.0 eq.) in methanol (40 ml) was heated to 60°C and stirred for 3 hours. After cooling to room temperature, the reaction was diluted with water and extracted with ethyl acetate, and the organic layer was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure.
  • Step 12d Preparation of 6-(bromomethyl)-5-fluorobenzofuran: Under nitrogen protection, 5-fluoro-6-methylbenzofuran (1.5 g, A mixture of 1,4-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (3,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropan
  • Step 12e Preparation of 2-(5-fluorobenzofuran-6-yl)acetonitrile: Under nitrogen protection, a mixture of 6-(bromomethyl)-5-fluorobenzofuran (2 g, 8.7 mmol, 1.0 eq), trimethylsilyl nitrile (1.7 g, 17.4 mmol, 2.0 eq), tetrabutylammonium fluoride in tetrahydrofuran (17 ml, 17.4 mmol, 2.0 eq) and potassium carbonate (1.1 g, 8.7 mmol, 1.0 eq) in tetrahydrofuran (20 ml) was stirred at 60° C. overnight.
  • reaction solution was diluted with water and extracted with ethyl acetate, and the organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain 2-(5-fluorobenzofuran-6-yl)acetonitrile (crude product) as a yellow oil.
  • Step 12g Preparation of 2-(5-fluorobenzofuran-6-yl)propanoic acid: Under nitrogen, n-butyl lithium (1.38 ml, 2.06 mmol, 2.0 eq) was added to a solution of 2-(5-fluorobenzofuran-6-yl)acetic acid (210 mg, 1.03 mmol, 1.0 eq) in tetrahydrofuran (5 ml) at -70°C, and the mixture was stirred at -70°C for 30 minutes. Diisopropylamine (21.9 mg, 0.22 mmol, 0.2 eq) was then added and stirred for 15 minutes.
  • Step 12h Preparation of 2-(5-fluorobenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one
  • Compound 14 To 2-(5-fluorobenzofuran-6-yl)propanoic acid (20 mg, 0.096 mmol, 1.0 equiv), 7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro 2-(7-azabenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (47.5 mg, 0.12 mmol, 1.3 eq.) was added to a mixture of -1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine
  • Example 13 Preparation of 6-methoxy-4-(1-(S)-7'-methyl-6'(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-1-thiopropane-2-yl)nicotinonitrile (Compound 11) (Prepared according to Schemes 4 and 5)
  • Step 13a Preparation of methyl 2-(5-bromo-2-methoxypyridin-4-yl)acetate (Compound 0409-11): Under nitrogen, lithium diisopropylamide in tetrahydrofuran (0.9 mol, 67 ml, 60.00 mmol, 3.0 eq) was added dropwise to a solution of 5-bromo-2-methoxy-4-methylpyridine (0408-11) (4.04 g, 20.00 mmol, 1.0 eq) in tetrahydrofuran (40 ml) at -30°C.
  • Step 13b Preparation of methyl 2-(5-cyano-2-methoxypyridin-4-yl)propanoate (compound 0410-11): Under nitrogen, a solution of methyl 2-(5-bromo-2-methoxypyridin-4-yl)acetate (0409-11) (3.71 g, 14.27 mmol, 1.0 eq) in tetrahydrofuran (60 ml) at -70°C was added with lithium bis(trimethylsilyl)amide (1 mol per liter, 16 ml, 15.98 mmol, 1.12 eq) and stirred for 1 hour.
  • Step 13c Preparation of 2-(5-cyano-2-methoxypyridin-4-yl)propanoic acid (compound 0405-11): Under nitrogen protection, a mixture of methyl 2-(5-cyano-2-methoxypyridin-4-yl)propanoate (0410-11) (434 mg, 1.97 mmol, 1.0 equiv) and potassium trimethylsilanol (380 mg, 2.96 mmol, 1.5 equiv) in tetrahydrofuran (5 ml) was stirred at room temperature for 1 hour. The reaction mixture was quenched with a 1 mol/L hydrochloric acid solution and extracted with ethyl acetate.
  • Step 13d Preparation of 6-methoxy-4-(1-(S)-7'-methyl-6'(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)-1-oxopropan-2-yl)nicotinonitrile
  • Compound 0412-11 2-(5-cyano-2-methoxypyridin-4-yl)propanoic acid (0405-11) (20.6 mg, 0.10 mmol, 1.0 equiv), (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)-1-oxopropan-2-yl)nicotinonitrile (Compound 0412-11) A mixture of 2-(7-azabenzotriazo
  • Step 13e Preparation of 6-methoxy-4-(1-(S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)-1-thiopropane-2-yl)nicotinonitrile
  • Compound 11 6-methoxy-4-(1-(S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)-1-thiopropane-2-yl)nicotinonitrile
  • Compound 11 A mixture of 4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridin]-1-yl)-1-oxopropan-2
  • Step 15a Preparation of methyl 2-(5-fluoro-2-methoxypyridin-4-yl)propanoate (Compound 0410-67)
  • Step 15b Preparation of 2-deuterium-2-(5-fluoro-2-methoxypyridin-4-yl)propionic acid (Compound 0405-67): Under nitrogen protection, lithium bis(trimethylsilyl)amide (0.35 ml, 1M tetrahydrofuran solution, 0.35 mmol, 1.4 eq.) was added dropwise to a solution of methyl 2-(5-fluoro-2-methoxypyridin-4-yl)propanoate (0410-67) (50 mg, 0.23 mmol, 1.0 eq.) in tetrahydrofuran (5 ml) at -78°C, and the mixture was stirred at -78°C for 1.0 hour.
  • Step 15c Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one-2-deuterium
  • Compound 67 Under nitrogen protection, at 0°C, add (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl [1,8]naphthyridine] (0122-1) (50 mg, 0.14 mmol, 1.0 eq), 2-deuterium-2-(5-fluoro-2-methoxypyridin-4-yl) propionic acid (0405-67) (23 mg
  • Lawesson's reagent (47 mg, 0.12 mmol, 1.2 eq.) was added to a toluene (2 ml) solution of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one-2-deuterium (Compound 67) (45 mg, 0.10 mmol, 1.0 eq.), and the mixture was heated to 100°C for overnight reaction.
  • Step 17a Preparation of 2-chloro-6-(methyl-d3)pyridin-3-amine 2-chloro-6-(methyl-d3)pyridin-3-amine (Compound 0106-59):
  • tert-butyl lithium (1.3M, 51.69 ml, 67.20 mmol, 1.5 eq) was added dropwise to a solution of 6-bromo-2-chloro-N,N-bis(4-methoxybenzyl)pyridin-3-amine (19.98 g, 44.80 mmol, 1.0 eq) in tetrahydrofuran (200 ml) at -70°C and stirred for 1 hour.
  • Trifluoroacetic acid (20 ml) was added to a mixture of 2-chloro-N, N-bis(4-methoxybenzyl)-6-(methyl-d3)pyridine-3-amine (9.0 g, 23.32 mmol, 1.0 eq.) in dichloromethane (80 ml), and stirred overnight at room temperature. The solvent was removed under vacuum. The pH of the residue was adjusted to 13 with a 2M sodium hydroxide solution. The mixture was extracted with dichloromethane, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated.
  • Step 17b Preparation of 2-chloro-3-iodo-6-(methyl-d3)pyridine (Compound 0107-59): To a solution of 2-chloro-6-(methyl-d3)pyridin-3-amine (0106-59) (3.38 g, 23.32 mmol, 1.0 eq) in concentrated hydrochloric acid/water (25 ml/25 ml) was added 24 ml of sodium nitrite solution (2.41 g. 34.98 mmol, 1.5 eq) and stirred at 0°C for 30 minutes.
  • Methyl tert-butyl ether (15 ml) was then added to the reaction and 80 ml of potassium iodide solution (7.74 g, 46.64 mmol, 2.0 eq) was added dropwise at 0°C. The mixture was stirred at room temperature overnight. The reaction was quenched with sodium sulfite solution. The pH of the residue was adjusted to 7 with 2M sodium hydroxide solution. The mixture was extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated.
  • Step 17c Preparation of tert-butyl 3-hydroxy-3-((trimethylsilyl)ethynyl)pyrrolidine-1-carboxylate (Compound 0102-59): Under nitrogen protection, to a solution of ethynyltrimethylsilane (2.55 g, 25.9 mmol, 1.2 eq) in tetrahydrofuran (40 ml) was added dropwise n-butyllithium (9.5 ml, 23.8 mmol, 1.1 eq) at -70 °C, and the mixture was stirred at -70 °C for 1.0 hour.
  • n-butyllithium 9.5 ml, 23.8 mmol, 1.1 eq
  • Step 17d Preparation of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (Compound 0103-59): Under nitrogen protection, potassium carbonate (5.80 g, 42.3 mmol, 2.0 eq.) was added to a solution of tert-butyl 3-hydroxy-3-((trimethylsilyl)ethynyl)pyrrolidine-1-carboxylate (0102-59) (6.00 g, 21.2 mmol, 1.0 eq.) in methanol (100 ml), and the mixture was stirred at room temperature for 1.5 hours.
  • potassium carbonate 5.80 g, 42.3 mmol, 2.0 eq.
  • Step 17e Preparation of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (0104-59): Under nitrogen protection, acetyl chloride (1.98 g, 25.2 mmol, 1.2 eq) was added to a solution of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (0103-59) (4.43 g, 21.0 mmol, 1.0 eq) and N,N-diisopropylethylamine (4.06 g, 31.5 mmol, 1.5 eq) in dichloromethane (100 ml) at 0° C. The mixture was stirred at 0° C. for 1 hour.
  • reaction solution was quenched with water and extracted with dichloromethane, and the organic phase was dried and concentrated under reduced pressure.
  • Step 17f Preparation of tert-butyl 3-ethynyl-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0105-59): Under nitrogen protection, a solution of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (0104-59) (2.83 g, 11.2 mmol, 1.0 eq) in tetrahydrofuran (10 ml) was added to a solution of 4-methoxybenzylamine (3.07 g, 22.3 mmol, 2.0 eq) and cuprous chloride (111 mg, 1.1 mmol, 0.1 eq) in tetrahydrofuran (30 ml) at 70° C.
  • Step 17g Preparation of tert-butyl 3-((2-chloro-6-(methyl-d3)pyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0108-59): Under nitrogen protection, 2-chloro-3-iodo-6-(methyl-d3)pyridine (0107-59) (1.30 g, 5.07 mmol, 1.0 equiv), 3-ethynyl -3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylic acid tert-butyl ester (0105-59) (1.69 g, 5.07 mmol, 1.0 eq), cuprous iodide (96 mg, 0.51 mmol, 0.1 eq) and [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (356 mg, 0.51 mmol,
  • Step 17h Preparation of tert-butyl 3-(2-(2-chloro-6-(methyl-d3)pyridin-3-yl)ethyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate
  • Compound 0109-59 A mixture of tert-butyl 3-((2-chloro-6-(methyl-d3)pyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (0108-59) (1.90 g, 4.126 mmol, 1.0 eq), platinum dioxide (187 mg, 0.825 mmol, 0.2 eq) in methanol (20 ml) was stirred at room temperature overnight under a hydrogen atmosphere.
  • Step 17i Preparation of tert-butyl 1'-(4-methoxybenzyl)-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0110-59): Under nitrogen protection, tert-butyl 3-(2-(2-chloro-6-(methyl-d3)pyridin-3-yl)ethyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate A mixture of butyl ester (0109-59) (1.30 g, 2.81 mmol, 1.0 eq.), palladium acetate (63 mg, 0.28 mmol, 0.1 eq.), 2-dicyclohexylphosphine-2',6'-diisopropoxy-1,1'-biphenyl (262 mg, 0.56 mmol,
  • Step 17j Preparation of tert-butyl 7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0112-59): To a mixture of tert-butyl 1'-(4-methoxybenzyl)-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0110-59) (945 mg, 2.22 mmol, 1.0 eq) in dichloromethane (8 mL) was added trifluoroacetic acid (4 mL) and stirred at room temperature for 4 hours.
  • Step 17k Preparation of tert-butyl 6'-bromo-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0115-59): Under nitrogen protection, 1,3-dibromo-5,5-dimethylhydantoin (309 mg, 1.08 mmol, 0.6 eq) was added to a solution of tert-butyl 7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0112-59) (550 mg, 1.80 mmol, 1.0 eq) in dichloromethane (10 ml) and stirred at 0°C for 30 minutes.
  • the reaction was stirred with 2% thiocyanate. The mixture was quenched with sodium sulfate solution. The mixture was diluted with water and extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated.
  • Step 171 Preparation of (1-(tert-butyloxycarbonyl)-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (Compound 0117-59): Under nitrogen protection, 6'-bromo-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0115-59) (690 mg, 1.79 mmol, 1.0 equiv), tetrahydroxydiboron ( A mixture of 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (26 mg, 0.054 mmol, 0.03 eq.), potassium acetate (439 mg, 4.48
  • Step 17m Preparation of (7'-(methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0119-59): Under nitrogen protection, (1-(tert-butyloxycarbonyl)-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester 2-(2-(2-(2-(2-(2-nitro-3,2'-[1,8]naphthyridin]-6'-yl)boronic acid (0117-59) (360 mg, 1.03 mmol, 1.0 equiv), 2-bromopyrimidine (213 mg, 1.34 mmol, 1.3
  • Step 17o Preparation of (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(7'-(methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one
  • Compound 59 To a mixture of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0406-7) (40 mg, 0.20 mmol, 1.0 eq.) in acetonitrile (2 mL) was added pyridine trifluoromethanesulfonate (92 mg, 0.4 mmol, 2.0 eq) and N,N'-carbonyldiimidazole (36 mg, 0.22 mmol, 1.1 eq) were added and the mixture was stirred at room temperature for 1 hour.
  • Step 20a Preparation of 4-bromo-2,6-dimethoxybenzaldehyde: Sodium methoxide (5.4 M methanol solution, 5.87 mL, 31.67 mmol, 2.0 eq.) was added to a mixture of 4-bromo-2,6-difluorobenzaldehyde (3.5 g, 15.84 mmol, 1.0 eq.) in methanol (20 mL). The mixture was heated to 65°C under nitrogen atmosphere for 3.5 hours. The solvent was removed under reduced pressure. The residue was diluted with water (60 mL) and then extracted with ethyl acetate (30 mL ⁇ 3).
  • Step 20b Preparation of 4-bromo-2-hydroxy-6-methoxybenzaldehyde: To a mixture of aluminum chloride (3.54 g, 26.53 mmol, 1.0 eq.) and sodium iodide (3.98 g, 26.53 mmol, 2.0 eq.) in acetonitrile (16 ml) at 0°C was added dropwise a solution of 4-bromo-2,6-dimethoxybenzaldehyde (3.25 g, 13.27 mmol, 1.0 eq.) in dichloromethane (16 ml). The mixture was warmed to room temperature and then stirred for 4 hours.
  • Step 20c Preparation of 6-bromo-4-methoxybenzofuran: To a mixture of 4-bromo-2-hydroxy-6-methoxybenzaldehyde (2.87 g, 12.42 mmol, 1.0 eq.) and cesium carbonate (10.12 g, 31.05 mmol, 2.5 eq.) in N,N-dimethylformamide (20 ml) was added ethyl bromoacetate (2.49 g, 14.91 mmol, 1.2 eq.). The mixture was heated to 120°C under nitrogen atmosphere for 8 hours. The mixture was diluted with water (80 ml) and then extracted with ethyl acetate (20 ml x 3).
  • Step 20d Preparation of tert-butyl 2-(4-methoxybenzofuran-6-yl)propanoate: To a mixture of 6-bromo-4-methoxybenzofuran (100 mg, 0.44 mmol, 1.0 eq), Pd(dba) 2 (25 mg, 0.044 mmol, 0.1 eq) and X-Phos (42 mg, 0.088 mmol, 0.2 eq) in tetrahydrofuran (1 ml) was added (1-(tert-butyloxy)-1-oxopropan-2-yl)bromide.
  • 6-bromo-4-methoxybenzofuran 100 mg, 0.44 mmol, 1.0 eq
  • Pd(dba) 2 25 mg, 0.044 mmol, 0.1 eq
  • X-Phos 42 mg, 0.088 mmol, 0.2 eq
  • Step 20e Preparation of 2-(4-methoxybenzofuran-6-yl)propanoic acid: A mixture of tert-butyl 2-(4-methoxybenzofuran-6-yl)propanoate (70 mg, 0.254 mmol, 1.0 eq) in hydrogen chloride/dioxane solution (4M, 1 ml) was stirred at room temperature overnight. The solvent was removed under reduced pressure. The residue was dried under vacuum to give a pale yellow oil (2-(4-methoxybenzofuran-6-yl)propanoic acid (56 mg, crude).
  • LCMS (ESI): m/z 221 [M+1] + ; TLC: Rf 0.3 (dichloromethane:methanol 10:1).
  • Step 20f Preparation of 2-(4-methoxybenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one
  • Compound 87 2-(4-methoxybenzofuran-6-yl)propanoic acid (23 mg, 0.105 mmol, 1.0 equiv), 2-(7-azobenzotriazole)-N,N,N',N'- To a mixture of tetramethyluronium hexafluorophosphate (42 mg, 0.11 mmol, 1.05 eq) and N,N-diisopropylethylamine (68 mg, 0.525 mmol, 5.0 eq) in dichloromethane (2.5 ml) was added (S)-7'-methyl-6
  • the mixture was stirred at room temperature for 0.5 hours.
  • the mixture was diluted with water (20 ml) and then extracted with ethyl acetate (10 ml x 3).
  • the combined organic layer was washed with saturated brine (15 ml), dried over anhydrous sodium sulfate and concentrated.
  • Step 22a Preparation of 1-bromo-2-bromomethyl-4-methoxybenzene: Under nitrogen protection, a mixture of 1-bromo-4-methoxy-2-methylbenzene (2.5 g, 12.44 mmol, 1.0 eq.), N-bromosuccinimide (2.22 g, 12.44 mmol, 1.0 eq.) and azobisisobutyronitrile (409 mg, 2.49 mmol, 0.2 eq.) in 1,2-dichloroethane (25 ml) was stirred at 72°C overnight. Dichloromethane was added, and the mixture was washed with sodium thiosulfate solution and saturated brine. The organic phase was dried and concentrated under reduced pressure to obtain a yellow
  • Step 22b Preparation of 2-(2-bromo-5-methoxyphenyl)acetonitrile: Under nitrogen protection, a mixture of 1-bromo-2-bromomethyl-4-methoxybenzene (4.08 g, 12.44 mmol, 1.0 eq.), trimethylsilyl cyanide (1.85 g, 18.66 mmol, 1.5 eq.) and tetrabutylammonium fluoride (1M tetrahydrofuran solution, 18.7 ml, 18.66 mmol, 1.5 eq.) in acetonitrile (30 ml) was stirred at room temperature for 1 hour. The mixture was diluted with ethyl acetate, washed with water and saturated brine.
  • Step 22c Preparation of 2-(2-bromo-5-methoxyphenyl)acetic acid: To a solution of 2-(2-bromo-5-methoxyphenyl)acetonitrile (5.61 g, 12.44 mmol, 1.0 eq) in ethanol (50 ml) was added an aqueous solution (50 ml) of potassium hydroxide (2.09 g, 37.32 mmol, 3.0 eq). The mixture was stirred at 85°C overnight. The mixture was concentrated to 75 ml and washed with ethyl acetate. The pH value of the aqueous phase was adjusted to 2 with concentrated hydrochloric acid.
  • Step 22d Preparation of methyl 2-(2-bromo-5-methoxyphenyl)acetate: Under nitrogen protection, oxalyl chloride (2M dichloromethane solution, 7.56 ml, 15.12 mmol, 2.0 eq.) was added dropwise to a mixture of 2-(2-bromo-5-methoxyphenyl)acetic acid (1.86 g, 7.56 mmol, 1.0 eq.) and N,N-dimethylformamide (0.1 ml) in 10 ml dichloromethane. The mixture was stirred at room temperature for 2 hours. 2 ml of methanol was added dropwise and the mixture was stirred for 30 minutes.
  • oxalyl chloride 2M dichloromethane solution, 7.56 ml, 15.12 mmol, 2.0 eq.
  • 2-(2-bromo-5-methoxyphenyl)acetic acid (1.86 g, 7.56 mmol, 1.0 eq.
  • Step 22e Preparation of methyl 2-(2-bromo-5-methoxyphenyl)propanoate: To a solution of methyl 2-(2-bromo-5-methoxyphenyl)acetate (437 mg, 1.68 mmol, 1.0 eq) in tetrahydrofuran was added dropwise, under nitrogen, lithium bis-(trimethylsilyl)amide (1M in tetrahydrofuran, 1.71 ml, 1.71 mmol, 1.02 eq) at -70°C. The mixture was stirred for 1.5 hours.
  • Step 22f Preparation of methyl 2-(2-cyano-5-methoxyphenyl)propanoate: A mixture of methyl 2-(2-bromo-5-methoxyphenyl)propanoate (719 mg, 2.63 mmol, 1.0 eq.) and cuprous cyanide (1.42 g, 15.80 mmol, 6.0 eq.) in 8 ml of N-methylpyrrolidone was stirred and heated in a microwave at 180°C for 2 hours. The mixture was filtered through celite. The filtrate was diluted with ethyl acetate, washed with water and saturated brine.
  • Step 22g Preparation of methyl 2-(2-cyano-4-iodo-5-methoxyphenyl)propionate: To a solution of methyl 2-(2-cyano-5-methoxyphenyl)propionate (331 mg, 1.51 mmol, 1.0 eq.) in trifluoroacetic acid (5 ml) was added N-iodosuccinimide (340 mg, 1.51 mmol, 1.0 eq.) at room temperature and stirred for 6 hours. The mixture was concentrated and sodium carbonate solution was added. Ethyl acetate was added for extraction and washed with water and saturated brine.
  • Step 22h Preparation of 2-(2-cyano-5-hydroxy-4-iodophenyl)propionic acid: Under nitrogen protection, sodium hydride (145 mg, 3.63 mmol, 1.0 eq) was added to a solution of ethanethiol (225 mg, 3.63 mmol, 5.0 eq) in N,N-dimethylformamide (5 ml) at 0°C and stirred at room temperature for 30 minutes. A solution of methyl 2-(2-cyano-4-iodo-5-methoxyphenyl)propanoate (250 mg, 0.725 mmol, 1.0 eq) in N,N-dimethylformamide was added.
  • Step 22i Preparation of methyl 2-(2-cyano-5-hydroxy-4-iodophenyl)propanoate: Under nitrogen protection, 2-(2-cyano-5-hydroxy-4-iodophenyl)propanoic acid (160 mg, 0.505 mmol, 1.0 eq.) and N,N-dimethylformamide (0.05 ml) were added to 5% ethyl acetate. 1.52 mmol, 3.00 equiv.) was added dropwise to a mixture of 1.5 ml of dichloromethane and 2.0 ml of oxalyl chloride. The mixture was stirred at room temperature for 1.5 hours. 2 ml of methanol was added dropwise and the mixture was stirred for 15 minutes.
  • Step 22j Preparation of methyl 2-(5-cyanobenzofuran-6-yl)propanoate: Under nitrogen, a mixture of methyl 2-(2-cyano-5-hydroxy-4-iodophenyl)propanoate (130 mg, 0.393 mmol, 1.0 eq), trimethylsilylacetylene (192 mg, 0.393 mmol, 1.0 eq), cuprous iodide (38 mg, 0.197 mmol, 0.5 eq), dichlorobistriphenylphosphine palladium (28 US shares, 0.04 mmol, 0.1 eq) and N,N-diisopropylethylamine (253 mg, 1.96 mmol, 5.0 eq) in 15 ml of tetrahydrofuran was stirred at 66° C.
  • Step 22k Preparation of 2-(5-cyanobenzofuran-6-yl)propanoic acid: To a solution of methyl 2-(5-cyanobenzofuran-6-yl)propanoate (46 mg, 0.2 mmol, 1.0 eq) in tetrahydrofuran/water (2 ml/0.5 ml) was added lithium hydroxide (17 mg, 0.4 mmol, 2.0 eq). The mixture was stirred at room temperature for 1 hour. The mixture was acidified with dilute hydrochloric acid. Ethyl acetate was added for extraction, and the mixture was washed with water and saturated brine.
  • Step 221 Preparation of 6-(1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-1-oxopropan-2-yl)benzofuran-5-carbonitrile
  • Compound 77 (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122- A mixture of dichloromethane (2 ml) of 1-(4-nitrobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (42 mg, 0.11 mmol, 1.1 eq.) and 2-(7-azabenzotriazole)-N,
  • Step 23a Preparation of 1-(tert-butyl)2-methyl-4-hydroxy-4-(trimethylsilyl)ethynyl)pyrrolidine-1,2-dicarboxylate (Compound 0102-55): To a mixture of ethynyltrimethylsilane (4.85 g, 49.33 mmol, 1.2 eq) in tetrahydrofuran (60 mL) was added dropwise n-butyllithium (2.5 M in hexane, 17.3 mL, 43.16 mmol, 1.05 eq) at -78°C. The mixture was stirred at -78°C under nitrogen for 1 hour.
  • n-butyllithium 2.5 M in hexane, 17.3 mL, 43.16 mmol, 1.05 eq
  • Step 23b Preparation of 1-(tert-butyl)-2-methyl-4-ethynyl-4-hydroxypyrrolidine-1,2-dicarboxylate (Compound 0103-55): To a mixture of 1-(tert-butyl)-2-methyl-4-hydroxy-4-(trimethylsilyl)ethynyl)pyrrolidine-1,2-dicarboxylate (0102-55) (12.2 g, 35.78 mmol, 1.0 eq) in methanol (50 mL) was added potassium carbonate (7.4 g, 53.67 mmol, 1.5 eq). The mixture was stirred at room temperature for 3 hours. The solvent was removed under reduced pressure.
  • Step 23c Preparation of 1-(tert-butyl) 2-methyl 4-acetoxy-4-ethynylpyrrolidine-1,2-dicarboxylate (Compound 0104-55): To a mixture of 1-(tert-butyl) 2-methyl 4-ethynyl-4-hydroxypyrrolidine-1,2-dicarboxylate (0103-55) (5.61 g, 20.86 mmol, 1.0 eq) and N,N-diisopropylethylamine (9.2 mL, 52.14 mmol, 2.5 eq) in dichloromethane (50 mL) was added acetyl chloride (2.68 mL, 35.46 mmol, 1.7 eq) dropwise at 0°C.
  • Step 23d 1-(tert-butyl)2-methyl-4-ethynyl-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (compound Preparation of compound 0105-55): To a mixture of p-methoxybenzylamine (4.16 g, 30.35 mmol, 2.0 eq.) and cuprous chloride (0.15 g, 1.52 mmol, 0.1 eq.) in tetrahydrofuran (50 ml) was added dropwise a solution of 1-(tert-butyl) 2-methyl 4-acetoxy-4-ethynylpyrrolidine-1,2-dicarboxylate (0104-55) (4.72 g, 15.18 mmol, 1.0 eq.) in tetrahydrofuran (30 ml) at 70°C.
  • Step 23e Preparation of 1-(tert-butyl)2-methyl 4-((2-chloro-6-methylpyridin-3-yl)ethynyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate
  • Compound 0108-55 2-chloro-3-iodo-6-methylpyridine (0107-1) (1.41 g, 5.57 mmol, 1.05 equiv), bistriphenylphosphine palladium dichloride (0 1-(tert-butyl)-2-methyl-4-ethynyl-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (0105-55) (2.06 g, 5.31 mmol, 1.0 eq.) was added to a mixture of triethylamine (25 ml) with 1-(tert-butyl)-2-methyl-4-ethynyl-4-((4-meth)
  • the mixture was heated to 80°C under a nitrogen atmosphere for 3.5 hours.
  • the solvent was removed under reduced pressure.
  • the residue was diluted with water (50 ml) and then extracted with ethyl acetate (30 ml x 3).
  • the combined organic layer was washed with saturated brine (30 ml), dried over anhydrous sodium sulfate and concentrated.
  • Step 23f Preparation of 1-(tert-butyl)2-methyl 4-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate
  • Compound 0109-55 To a mixture of 1-(tert-butyl)2-methyl 4-((2-chloro-6-methylpyridin-3-yl)ethynyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (0108-55) (2.1 g, 4.09 mmol, 1.0 eq) in methanol (20 ml) was added platinum dioxide (0.186 g, 0.82 mmol, 0.2 eq).
  • Step 23g Preparation of tert-butyl 1'-(4-methoxybenzyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0110-55):
  • the mixture was heated to 100°C in a microwave reactor for 1 hour.
  • the mixture was diluted with water (40 ml).
  • Step 23h Preparation of tert-butyl 5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0112-55): To a mixture of tert-butyl 1'-(4-methoxybenzyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0110-55) (450 mg, 1.03 mmol, 1.0 equiv) in dichloromethane (6 mL) was added trifluoroacetic acid (1.5 mL).
  • Step 23i Preparation of tert-butyl 6'-bromo-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0115-55): To a mixture of tert-butyl 5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0112-55) (193 mg, 0.61 mmol, 1.0 equiv) in dichloromethane (5 mL) was added dibromohydantoin (96 mg, 0.34 mmol, 0.55 equiv) at 0°C.
  • Step 23j Preparation of (1-(tert-Butyloxycarbonyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (Compound 0117-55): tert-butyl 6'-bromo-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0115-55) (242 mg, 0.61 mmol, 1.0 equiv), Xphos-Pd-G 2 (9.6 mg, 0.012 mmol, 0.02 eq), 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (8.7 mg, 0.018 mmol, 0.03 eq), potassium
  • Step 23k Preparation of tert-butyl 5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0119-55): To (1-(tert-butyloxycarbonyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (0117-55) (165 mg, 0.457 mmol, 1.0 equiv.), Xphos-Pd-G2 (10.8 mg, 0.014 mmol, 0.03 equiv.), 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (6.7 mg, 0.014 mmol,
  • the mixture was heated to 55°C for 4 hours under a nitrogen atmosphere.
  • the mixture was diluted with water (20 ml) and then extracted with ethyl acetate (15 ml x 3).
  • the combined organic layer was washed with saturated brine (10 ml), dried over anhydrous sodium sulfate and concentrated.
  • Step 231 Preparation of 5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0121-55): A mixture of tert-butyl 5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0119-55) (109 mg, 0.276 mmol, 1.0 eq) in hydrogen chloride/methanol solution (4M solution, 2 ml) was stirred at room temperature for 3 hours.
  • Step 23m Preparation of (2R)-1-(5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)-2-(5-fluoro-2-methoxypyridin-4-yl)propan-1-one (Compound 55): to (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0406-7) (27.5 mg, 0.138 mmol, 1.0 equiv), benzotriazole-N,N,N',N'-tetramethyluronium hexafluoro Phosphate (63 mg, 0.166 mmol, 1.2 eq), 1-hydroxybenzotriazole (20 mg, 0.152 mmol, 1.1 eq) and N,N-diisopropylethylamine (89 mg,
  • Step 24a Preparation of (3S,5S)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0113-100):
  • Potassium carbonate (10.6 g, 76.98 mmol, 1.5 eq.) was added to a mixture of 1-(tert-butyl)2-methyl(2R)-4-hydroxy-4-((trimethylsilyl)ethynyl)pyrrolidine-1,2-dicarboxylate (17.5 g, 51.32 mmol, 1.0 eq.) in methanol (45 ml). The mixture was stirred at room temperature for 1 hour. The solvent was removed under reduced pressure. The residue was diluted with water (80 ml) and then extracted with ethyl acetate (50 ml ⁇ 4).
  • the mixture was warmed to room temperature and stirred for 3 hours.
  • the mixture was cooled to 0°C.
  • Water (10.5 ml) and 15% sodium hydroxide solution (2.65 ml) were added.
  • the mixture was warmed to room temperature and stirred for 10 minutes.
  • the mixture was diluted with ethyl acetate (50 ml) and then filtered.
  • the mixture was heated to 100° C. in a microwave reactor for 1 hour.
  • the mixture was diluted with water (20 ml) and tetrahydrofuran (20 ml).
  • Di-tert-butyl dicarbonate (4.67 g, 21.40 mmol, 1.3 eq) was added, and the mixture was stirred at room temperature for 1 hour.
  • the mixture was diluted with water (60 ml), and then extracted with ethyl acetate (40 ml ⁇ 3).
  • the combined organic layer was washed with saturated brine (50 ml), dried over anhydrous sodium sulfate and concentrated.
  • the mixture was warmed to room temperature and stirred for 6 hours.
  • the mixture was diluted with water (30 ml) and then extracted with ethyl acetate (15 ml x 4).
  • the combined organic layer was washed with saturated brine (20 ml), dried over anhydrous sodium sulfate and concentrated.
  • Step 24b Preparation of (3S,5S)-6'-bromo-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0116-100): To a mixture of (3S,5S)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0113-100) (548 mg, 1.73 mmol, 1.0 equiv) in dichloromethane (10 mL) at 0° C.
  • dibromohydantoin (272 mg, 0.95 mmol, 0.55 equiv). The mixture was stirred at 0° C. for 0.5 h. The reaction was quenched by the addition of saturated aqueous sodium sulfite (15 mL). The aqueous layer was extracted with ethyl acetate (15 ml x 3).
  • Step 24c Preparation of ((3S,5S)-1-(tert-butyloxycarbonyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (Compound 0118-100): to (3S,5S)-6'-bromo-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0116-100) (685 mg, 1.73 mmol, 1.0 equiv) , Xphos-Pd-G2 (27 mg, 0.035 mmol, 0.02 eq), 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (
  • Step 24d Preparation of (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0120-100): To ((3S,5S)-1-(tert-butyloxycarbonyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (0118-100) (299 mg, 0.
  • Step 24e Preparation of (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0122-100): A mixture of (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0120-100) (270 mg, 0.68 mmol, 1.0 eq) in hydrogen chloride/methanol solution (4M solution, 2 ml) was stirred at room temperature for 2 hours.
  • Step 24f Preparation of (R)-1-((3R,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)-2-(5-fluoro-2-methoxypyridin-4-yl)propan-1-one
  • Compound 100 to (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0406-7) (95 mg, 0.476 mmol, 1.0 equiv), 6-chlorobenzotriazole-1, To a mixture of 1,3,3-tetramethyluronium hexafluorophosphate (217 mg, 0.524 mmol, 1.1 eq) and N,N-diisopropylethylamine (196 mg, 1.523 mmol, 3.2 eq
  • Step 26a Preparation of (9H-fluoren-9-yl)methyl (3S,5R)-5-(hydroxymethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate: A solution of (3S,5R)-5-(hydroxymethyl)-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (500 mg, 1.10 mmol, 1.0 eq) in hydrogen chloride/dioxane (5 mL) was stirred at room temperature for one hour.
  • reaction was concentrated under vacuum. The residue was added with tetrahydrofuran (10 ml), 9-fluorenylmethyl chloroformate (424 mg, 1.66 mmol, 1.5 eq), triethylamine (335 mg, 3.30 mmol, 3.0 Equivalent). The mixture was then stirred at room temperature for two hours. The reaction solution was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure.
  • Step 26b Preparation of (9H-fluoren-9-yl)methyl (3S,5R)-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate: Under nitrogen protection, to a solution of (9H-fluoren-9-yl)methyl (3S,5R)-5-(hydroxymethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (380 mg, 0.83 mmol, 1.0 equiv) in dichloromethane (5 ml) at 0°C was added diethylaminosulfur trifluoride (269 mg, 1.67 mmol, 2.0 equiv), and the mixture was stirred at 0°C for 1.0 hour.
  • Step 26c Preparation of (9H-fluoren-9-yl)methyl (3S,5R)-6'-bromo-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'-pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate: Under nitrogen protection at 0°C, add (9H-fluoren-9-yl)methyl (3S,5R)-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'-pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate.
  • 1,3-Dibromo-5,5-dimethylimidazolidine-2,4-dione (87 mg, 0.303 mmol, 0.5 eq) was added dropwise to a solution of 4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (278 mg, 0.61 mmol, 1.0 eq) in dichloromethane (5 ml), and the mixture was stirred at 0°C for 1.0 hour. The reaction solution was quenched with sodium carbonate solution and extracted with dichloromethane, and the organic phase was dried and concentrated under reduced pressure.
  • Step 26d Preparation of tert-butyl (3S,5R)-6'-bromo-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate: A solution of (9H-fluoren-9-yl)methyl (3S,5R)-6'-bromo-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'-pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (230 mg, 0.43 mmol, 1.0 eq) in piperidine/dichloromethane (1 mL/4 mL) was stirred at room temperature for 1 hour.
  • Step 26e Preparation of (3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester: Under nitrogen protection, (3S,5R)-6'-bromo-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (150 mg, 0.36 mmol, 1.0 equiv), 2-(tributyltinyl)pyrimidine (268 mg, 0.72 mmol, 2.0 eq), cuprous iodide (7 mg, 0.036 mmol, 0.1 eq), methanesulfonic acid (2-dicy
  • Step 26f Preparation of (3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine] dihydrochloride: A mixture of (3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (64 mg, 0.15 mmol, 1.0 eq) in hydrogen chloride dioxane solution (4 M solution, 2.5 mL) was stirred at room temperature for 1.0 h, the solvent was removed under reduced pressure and dried under vacuum, and the residue was used directly in the next step without further purification.
  • LCMS (ESI): m/z 314
  • Step 26g Preparation of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)propan-1-one (Compound 103): To (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0406-7) (31 mg, 0.155 mmol, 1.0 equiv), (3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)propan-1-one was added.
  • 6-Chlorobenzotriazole-1,1,3,3-tetramethyluronium hexafluorophosphate (77 mg, 0.186 mmol, 1.2 eq.) was added to a solution of 5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine] dihydrochloride (crude product) and N,N-diisopropylethylamine (50 mg, 0.388 mmol, 2.5 eq.) in dichloromethane (3 ml), and the mixture was stirred at room temperature for 1.0 hour.
  • Step 27a Preparation of methyl 2-(5-chloro-2-methoxypyridin-4-yl)acetate (0409-10): Under nitrogen protection, to a solution of diisopropylamine (1.23 g, 12.22 mmol, 3.2 eq.) in tetrahydrofuran (5 ml) at -30°C was added dropwise n-butyllithium in n-hexyl After the mixture was stirred at -30°C for 1 hour, a tetrahydrofuran solution (1 ml) of 5-chloro-2-methoxy-4-methylpyridine (0408-10) (600 mg, 3.82 mmol, 0.2 eq) was added dropwise to the mixture.
  • Step 27b Preparation of methyl 2-(5-chloro-2-methoxypyridin-4-yl)propanoate (Compound 0410-10): To a solution of methyl 2-(5-chloro-2-methoxypyridin-4-yl)acetate (0409-10) (550 mg, 2.56 mmol, 1 eq) in tetrahydrofuran (5 ml) at 0°C was added lithium bis(trimethylsilyl)amide (1 M in tetrahydrofuran, 2.86 ml, 2.86 mmol, 1.2 eq).
  • Step 27c Preparation of 2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid (Compound 0405-10): To a solution of methyl 2-(5-chloro-2-methoxypyridin-4-yl)propanoate (0410-10) (320 mg, 1.4 mmol, 1 eq) in tetrahydrofuran/water (5/1 ml) was added lithium hydroxide monohydrate (12 mg, 0.28 mmol, 0.2 eq). The mixture was stirred at room temperature overnight and then concentrated under reduced pressure. The aqueous residue was poured into water and washed with methyl tert-butyl ether.
  • Step 27d Preparation of 2-(5-chloro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one
  • Compound 0412-10) 2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid (0405-10) (18 mg, 0.085 mmol, 1 eq), (S)-7'-methyl-6'-(pyrimidin-2 2-(7-azobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (36 mg, 0.094 mmol, 1.1 eq) was added to a solution of 1,2-dihydro- ...
  • Step 27e Preparation of 2-(5-chloro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (Compound 10): 2-(5-chloro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione A mixture of '-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthy
  • Example 28 Preparation of 2-(5-chloro-2-methoxypyridin-4-yl)-1-((3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 104):
  • reaction solution was diluted with water (15 ml) and extracted with ethyl acetate (5 ml x 3). The organic phase was washed with saturated brine (15 ml), dried over anhydrous sodium sulfate and concentrated under reduced pressure.
  • Step 29a Preparation of (R)-2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid: Separation of enantiomers of 2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid (0405-10) (12.5 g, 57.98 mmol) was carried out by supercritical fluid chromatography (chromatographic column: ChiralPak IG, 300 ⁇ 50 mm I.D., 10 ⁇ m; mobile phase: A is carbon dioxide, B is methanol; gradient: 15% B; flow rate: 200 ml/min; back pressure: 100 bar; column temperature: 38°C).
  • the first eluting enantiomer was (R)-2-(5-chloro-2-methoxypyridin-4-yl)propionic acid (5.29 g, yield: 42%), and the second eluting enantiomer was (S)-2-(5-chloro-2-methoxypyridin-4-yl)propionic acid (4.81 g, yield: 38%).
  • Analysis conditions Column: ChiralPak IG, 100 ⁇ 4.6 mm I.D., 3 ⁇ m; Mobile phase: A is carbon dioxide, B is methanol (0.05% diethylamine); Gradient: 5-40% B; Flow rate: 2.5 ml/min; Back pressure: 100 bar; Column temperature: 35°C.
  • Step 29b Preparation of (R)-2-(5-chloro-2-methoxypyridin-4-yl)-1-((3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one
  • Compound 105 At 0°C, (R)-2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid (154 mg, 0.712 mmol, 1.05 equiv), (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one were added.
  • the mixture was stirred at 0°C for 1.5 hours.
  • the mixture was diluted with water (30 ml) and then extracted with ethyl acetate (20 ml ⁇ 3).
  • the combined organic layer was washed with saturated brine (20 ml), dried over anhydrous sodium sulfate and concentrated.
  • the reference compound used in the present invention is PF-07258669 (Journal of Medicinal Chemistry, 66 (5), 3195-3211, 2023, WO 2021250541), and its structure is as follows:
  • TR-FRET time-resolved fluorescence resonance energy transfer
  • test compound was dissolved in DMSO to prepare a 10 mM stock solution
  • test compound was diluted 5-fold with DMSO, with 10 or 5 concentration gradients;
  • Complete culture medium DMEM + 10% fetal bovine serum + 1% penicillin-streptomycin + 200 ⁇ g/mL hygromycin
  • the cells were resuspended in the experimental buffer and inoculated into 384 cell culture plates at a density of 2000 per well and a volume of 15 ⁇ l per well.
  • reaction plate was centrifuged at 1000 g for 1 min at room temperature and allowed to stand at 25°C for 1 h. Data were collected using Envision, with excitation light at 340 nm and emission light at 665 nm and 615 nm.
  • Ratio Signal 665nm/Signal 620nm, and the inhibition rate at different concentrations was calculated.
  • the IC 50 of the compound was calculated using the GraphPad nonlinear fitting formula:
  • mice Female C57 mice weighing 18-20 g. The test compound was dissolved in 30% sulfobutyl ⁇ -cyclodextrin and administered orally at 10 mg/kg. Blood was collected from the heart of the mice at 15 minutes, 30 minutes, and 1, 2, 4, 7, and 24 hours after administration. The mice were then euthanized and brain tissue was collected. About 0.3 ml of whole blood and 50-100 mg of brain tissue were collected at each time point. The whole blood was placed in a centrifuge tube containing K 2 -EDTA and centrifuged (3000 g, 10 minutes, 4°C) to collect plasma. The plasma and brain tissue were stored at -40°C for later use.
  • the brain tissue weighed in advance was homogenized with 0.9% NaCl solution at a ratio of 1:3 (g/ml). 50 ⁇ L of plasma sample or brain tissue homogenate was vortexed with 135 ⁇ L of acetonitrile (containing internal standard 0.1 ⁇ g/mL) for protein precipitation, centrifuged, and the supernatant was taken for LC-MS/MS analysis.
  • the compounds 100 and 104 provided by the present invention are well absorbed after oral administration to mice, and the exposure to blood and brain is high.
  • the results are shown in Figures 1-2 and Table 2.
  • the spiro heterocyclic compound of the present invention has a T max of 0.25 hours in plasma, a C max of 1243.33-1593.33 ng/ml, and an AUC 0-24 of 1448.77-1649.36 hr*ng/mL, a T max of 0.25-0.50 hours in the brain, a C max of 413.20-512.00 ng/ml, and an AUC 0-24 of 435.13-633.25 hr*ng/mL.
  • the compound of the present invention has better pharmacokinetic properties in the brain and has greater therapeutic potential in treating MC4 receptor-related diseases and/or symptoms such as cachexia, anorexia, etc.
  • Cmax refers to the maximum plasma concentration
  • T1 /2 is the half-life
  • AUC0-24 refers to the area under the 0-24 hour time-concentration curve
  • AUC0 -inf refers to the area under the 0-Inf time-concentration curve.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurology (AREA)
  • Rheumatology (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Nutrition Science (AREA)
  • Hospice & Palliative Care (AREA)
  • Otolaryngology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed in the present invention are a spiro heterocyclic compound having a structure as shown in formula (I), or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a deuterated compound thereof or a prodrug molecule thereof, and the use thereof. The compound has the effect of antagonizing MC4 receptor activity, can be used for treating various diseases associated with MC4 receptors, and has great application value.

Description

螺杂环化合物及其应用Spiro heterocyclic compounds and their applications 技术领域Technical Field

本发明涉及化学医药技术领域,具体涉及一种螺杂环化合物及其应用。The present invention relates to the technical field of chemical medicine, and in particular to a spiro heterocyclic compound and application thereof.

背景技术Background Art

黑皮质素受体(Melanocortin Receptors,MCR)参与调节生物体多种生理行为,包括能量平衡、色素沉着、炎症、心脑血管功能、进食、皮脂腺脂肪分泌等。MCR是G-蛋白偶联受体(G-protein Coupled Receptors,GPCR)的一个亚家族,具有七次跨膜结构。已知五种黑皮质素受体亚型,分别为MC1R、MC2R、MC3R、MC4R以及MC5R。MC3R主要在中枢神经系统表达,调节能量平衡;MC4R除了广泛表达于整个中枢神经系统外,也表达于外周组织,如脂肪组织,MC4R参与调节摄食行为和能量消耗;其他成员主要在外周表达,并参与调节色素沉着(MC1R),肾上腺功能(MC2R),免疫功能(MC1R/MC3R)和皮脂腺活性(MC5R)(Endocr Rev.2010 Aug;31(4):506-43)。Melanocortin receptors (MCR) are involved in regulating a variety of physiological behaviors of organisms, including energy balance, pigmentation, inflammation, cardiovascular and cerebrovascular function, feeding, sebaceous gland fat secretion, etc. MCR is a subfamily of G-protein coupled receptors (GPCR) with seven transmembrane structures. Five subtypes of melanocortin receptors are known, namely MC1R, MC2R, MC3R, MC4R and MC5R. MC3R is mainly expressed in the central nervous system and regulates energy balance. MC4R is widely expressed throughout the central nervous system and is also expressed in peripheral tissues, such as adipose tissue, where it is involved in regulating feeding behavior and energy expenditure. Other members are mainly expressed in the periphery and are involved in regulating pigmentation (MC1R), adrenal function (MC2R), immune function (MC1R/MC3R) and sebaceous gland activity (MC5R) (Endocr Rev. 2010 Aug; 31(4):506-43).

MC4R是一种A家族G蛋白偶联受体(GPCR),受内源性MC4R激动剂和反向激动剂/拮抗剂多肽调节。其信号传导具有调节能量稳态、恶病质、心血管功能、糖脂稳态、生殖和性功能、脑部炎症和焦虑方面等生理作用。抑制或拮抗MC4R信号,会导致食欲和食物摄入量增加(Endocr Rev.2010 Aug;31(4):506-43)。MC4R is a family A G protein-coupled receptor (GPCR) regulated by endogenous MC4R agonists and inverse agonist/antagonist peptides. Its signaling has physiological effects in regulating energy homeostasis, cachexia, cardiovascular function, glycolipid homeostasis, reproductive and sexual function, brain inflammation and anxiety. Inhibition or antagonism of MC4R signaling leads to increased appetite and food intake (Endocr Rev. 2010 Aug; 31(4): 506-43).

到目前为止,已有多种小分子MC4R的拮抗剂被报道,这些拮抗剂在癌症、慢性肾病、与年龄相关的恶病质或厌食症等的临床前模型中可以逆转或预防食欲、体重减轻(Biochim.Biophys.Acta.Mol.Basis.Dis.2017,1863(10),2414–2435;PLoS One 2009,4(3),e4774;J.Med.Chem.2004,47(7),1602-1604)。最近的一项研究报道了一种MC4R拮抗肽(TCMCB07),在口服或皮下给药后对恶病质模型具有疗效,可使其食欲得到一定的改善,同时体重增加。现已进入临床1期(J Clin Invest.2020Sep 1;130(9):4921-4934)。另外辉瑞公司报道了一种小分子MC4R拮抗剂PF-07258669,对老年大鼠恶病质模型有很强的疗效,并已进入临床试验(J Med Chem.2023 Mar 9;66(5):3195-3211;patent WO2021250541A1)。So far, a variety of small molecule MC4R antagonists have been reported, which can reverse or prevent appetite and weight loss in preclinical models of cancer, chronic kidney disease, age-related cachexia or anorexia (Biochim. Biophys. Acta. Mol. Basis. Dis. 2017, 1863 (10), 2414–2435; PLoS One 2009, 4 (3), e4774; J. Med. Chem. 2004, 47 (7), 1602-1604). A recent study reported that a MC4R antagonist peptide (TCMCB07) was effective in cachexia models after oral or subcutaneous administration, which improved appetite to a certain extent and increased weight. It has now entered Phase 1 clinical trials (J Clin Invest. 2020 Sep 1; 130 (9): 4921-4934). In addition, Pfizer has reported a small molecule MC4R antagonist PF-07258669, which has a strong therapeutic effect on the cachexia model of elderly rats and has entered clinical trials (J Med Chem. 2023 Mar 9; 66(5): 3195-3211; patent WO2021250541A1).

恶病质是一种破坏性的多因素消耗综合征,其特征是失去对能量和蛋白质平衡的稳态控制,体重会不由自主地减轻,同时骨骼肌质量减少。恶病质与多种恶性癌症的死亡高度相关(例如:胰腺癌、食道癌、胃癌、肺癌、肝癌和肠癌等,这些恶性肿瘤约占全球癌症死亡人数的一半)(Nat Rev Dis Primers.2018 Jan 18;4:17105;http://www.who.int/mediacentre/factsheets/fs297/en/),还与慢性非恶性疾病的进一步恶化相关 (例如心力衰竭、肾病、慢性阻塞性肺病、神经系统疾病、艾滋病和类风湿性关节炎等)(Lancet Oncol.12,489–495(2011))。目前尚无有效手段治疗恶病质,另外观察到当慢性肾病(Chronic Kidney Disease,CKD)患者具有更多肌肉或脂肪、食欲更好、吃得更多时,寿命更长(J Cachexia Sarcopenia Muscle.2011;2(1):9–25)。Cachexia is a devastating, multifactorial wasting syndrome characterized by loss of homeostatic control of energy and protein balance, involuntary weight loss, and loss of skeletal muscle mass. Cachexia is highly associated with mortality from multiple malignant cancers (e.g., pancreatic, esophageal, gastric, lung, liver, and colorectal cancers, which account for approximately half of cancer deaths worldwide) (Nat Rev Dis Primers. 2018 Jan 18;4:17105; http://www.who.int/mediacentre/factsheets/fs297/en/) and is also associated with further exacerbation of chronic non-malignant diseases. (For example, heart failure, kidney disease, chronic obstructive pulmonary disease, nervous system diseases, AIDS and rheumatoid arthritis, etc.) (Lancet Oncol. 12, 489–495 (2011)). There is currently no effective treatment for cachexia. In addition, it has been observed that when patients with chronic kidney disease (CKD) have more muscle or fat, better appetite, and eat more, they live longer (J Cachexia Sarcopenia Muscle. 2011; 2(1): 9–25).

鉴于黑皮质素-4受体(MC4R)在上述疾病中的重要作用,开发高效安全的MC4R拮抗剂对于预防或治疗与MC4R相关的多种疾病具有较大的应用价值。In view of the important role of melanocortin-4 receptor (MC4R) in the above diseases, the development of efficient and safe MC4R antagonists has great application value for preventing or treating various diseases related to MC4R.

发明内容Summary of the invention

基于此,本发明提供了一类螺杂环化合物,该类化合物具有拮抗MC4受体活性的作用,可用于治疗与MC4受体相关的多种疾病。Based on this, the present invention provides a class of spiro heterocyclic compounds, which have the effect of antagonizing the activity of MC4 receptors and can be used to treat various diseases related to MC4 receptors.

本发明包括如下技术方案。The present invention includes the following technical solutions.

一种具有式(I)所示结构的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子:
A spiroheterocyclic compound having a structure represented by formula (I) or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated product thereof or a prodrug molecule thereof:

其中:in:

R1、R2、R3和R4分别独立选自:H,氘,C1-C6烷基,C1-C6氘代烷基,C2-C6烯基,C2-C6炔基,C3-C8环烷基,卤素取代的C3-C8环烷基,C3-C8环烷基甲基,卤素,卤素取代的C1-C6烷基,羟基,羟基取代的C1-C6烷基,C1-C6烷氧基取代的C1-C6烷基,氰基,-OR,-N(R)2,-SR,-C(O)OR,-C(O)N(R)2,-C(O)R,-S(O)R,-S(O)2R,-S(O)2N(R)2,-N(R)C(O)R;R 1 , R 2 , R 3 and R 4 are independently selected from the group consisting of H, deuterium, C 1 -C 6 alkyl, C 1 -C 6 deuterated alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, halogen-substituted C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkylmethyl, halogen, halogen-substituted C 1 -C 6 alkyl, hydroxy, hydroxy-substituted C 1 -C 6 alkyl, C 1 -C 6 alkoxy-substituted C 1 -C 6 alkyl, cyano, -OR, -N(R) 2 , -SR, -C(O)OR, -C(O)N(R) 2 , -C(O)R, -S(O)R, -S(O) 2 R, -S(O) 2 N(R) 2 , -N(R)C(O)R;

R选自:H,C1-C6烷基,C2-C6烯基,C2-C6炔基,C3-C8环烷基,C3-C8环烷基甲基,卤素取代的C1-C6烷基,羟基取代的C1-C6烷基,烷氧基取代的C1-C6烷基,氨基取代的C1-C6烷基,烷基胺基取代的C1-C6烷基;R is selected from the group consisting of: H, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkylmethyl, halogen-substituted C 1 -C 6 alkyl, hydroxy-substituted C 1 -C 6 alkyl, alkoxy-substituted C 1 -C 6 alkyl, amino-substituted C 1 -C 6 alkyl, alkylamino-substituted C 1 -C 6 alkyl;

R5和R6分别独立选自:H,氘,卤素;R 5 and R 6 are independently selected from: H, deuterium, halogen;

M选自:CH,N;M is selected from: CH, N;

W选自:O,S,NR7W is selected from: O, S, NR 7 ;

Q选自:CR7R8,NR8,C=O,O,S,或者不存在;Q is selected from the group consisting of: CR 7 R 8 , NR 8 , C═O, O, S, or absent;

R7和R8分别独立选自:H,氘,卤素,C1-C6烷基,C1-C6氘代烷基,C2-C6烯基,C2-C6 炔基,C3-C8环烷基,卤素取代的C1-C6烷基,卤素取代的C3-C8环烷基,羟基,氰基,氨基,C1-C6烷基胺基;或者R8和A中的取代基相连形成取代或者未取代的5-7元碳环或杂环; R7 and R8 are independently selected from the group consisting of: H, deuterium, halogen, C1 - C6 alkyl, C1 - C6 deuterated alkyl, C2 - C6 alkenyl, C2 - C6 Alkynyl, C 3 -C 8 cycloalkyl, halogen-substituted C 1 -C 6 alkyl, halogen-substituted C 3 -C 8 cycloalkyl, hydroxy, cyano, amino, C 1 -C 6 alkylamino; or R 8 and the substituents in A are linked to form a substituted or unsubstituted 5-7 membered carbocyclic or heterocyclic ring;

A选自:R9和R10取代或未取代的C6-C10元芳基,R9和R10取代或未取代的5-10元杂芳基,R9和R10取代或未取代的7-10元苯并杂环基;A is selected from: C 6 -C 10 membered aryl substituted or unsubstituted by R 9 and R 10 , 5-10 membered heteroaryl substituted or unsubstituted by R 9 and R 10, 7-10 membered benzoheterocyclyl substituted or unsubstituted by R 9 and R 10 ;

各R9和R10分别独立选自:H,氘,卤素,C1-C6烷基,C1-C6氘代烷基,C2-C6烯基,C2-C6炔基,氰基,C3-C8环烷基,C3-C8氘代环烷基,C3-C8环烷基C1-C6烷基,C1-C6烷基磺酰基,C1-C6烷硫基,C1-C6氘代烷硫基,C3-C8环烷基硫基,卤素取代的C1-C6烷基,C1-C6烷氧基,C1-C6氘代烷氧基,卤素取代的C1-C6烷氧基,C3-C8环烷基氧基,C3-C8氘代环烷基氧基,卤素取代的C3-C8环烷基氧基,氨基甲酰基;Each R9 and R10 is independently selected from the group consisting of H, deuterium, halogen, C1 - C6 alkyl , C1 - C6 deuterated alkyl, C2 - C6 alkenyl, C2 - C6 alkynyl, cyano , C3 - C8 cycloalkyl, C3 - C8 deuterated cycloalkyl, C3- C8 cycloalkylC1- C6 alkyl, C1 - C6 alkylsulfonyl, C1- C6 alkylthio, C1-C6 deuterated alkylthio, C3-C8 cycloalkylthio , halogen -substituted C1 - C6 alkyl, C1 - C6 alkoxy, C1 - C6 deuterated alkoxy, halogen-substituted C1 - C6 alkoxy, C3 - C8 cycloalkyloxy, C3-C8 deuterated cycloalkyloxy, halogen-substituted C3 -C6 8 -cycloalkyloxy, carbamoyl;

A1选自:R11和R12取代或未取代的苯基,R11和R12取代或未取代的5-6元杂芳基; A1 is selected from: phenyl substituted or unsubstituted by R11 and R12 , 5-6 membered heteroaryl substituted or unsubstituted by R11 and R12 ;

各R11和R12分别独立选自:H,氘,卤素,C1-C6烷基,C1-C6氘代烷基,C3-C8环烷基,卤素取代的C1-C6烷基,氰基,C1-C6烷氧基。Each of R 11 and R 12 is independently selected from the group consisting of: H, deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 deuterated alkyl, C 3 -C 8 cycloalkyl, halogen-substituted C 1 -C 6 alkyl, cyano, and C 1 -C 6 alkoxy.

在其中一些实施例中,A选自:R9和R10取代或未取代的萘基,R9和R10取代或未取代的9-10元杂芳基,R9和R10取代或未取代的9-10元苯并杂环基。In some embodiments, A is selected from: naphthyl substituted or unsubstituted by R 9 and R 10 , 9-10 membered heteroaryl substituted or unsubstituted by R 9 and R 10 , 9-10 membered benzoheterocyclyl substituted or unsubstituted by R 9 and R 10 .

在其中一些实施例中,A选自:
In some embodiments, A is selected from:

其中:环中虚线代表其是一个C-C单键或者没有;Among them: the dotted line in the ring represents whether it is a C-C single bond or not;

X1、X2、X3分别独立选自:CR9,N。X 1 , X 2 , and X 3 are independently selected from the group consisting of CR 9 , N.

在其中一些实施例中,A选自:
In some embodiments, A is selected from:

在其中一些实施例中,A选自:W为O。In some embodiments, A is selected from: W is O.

在其中一些实施例中,各R9和R10分别独立选自:H,氘,卤素,C1-C3烷基,C1-C3氘代烷基,C2-C3烯基,C2-C3炔基,氰基,C3-C6环烷基,C3-C6氘代环烷基,C3-C6环烷基甲基,C1-C3烷基磺酰基,C1-C3烷硫基,C1-C3氘代烷硫基,C3-C6环烷基硫基,卤素取代的C1-C3烷基,C1-C3烷氧基,C1-C3氘代烷氧基,卤素取代的C1-C3烷氧基,C3-C6环烷基氧基,C3-C6氘代环烷基氧基,卤素取代的C3-C6环烷基氧基,氨基甲酰基。In some embodiments, each R 9 and R 10 is independently selected from the group consisting of H, deuterium, halogen, C 1 -C 3 alkyl, C 1 -C 3 deuterated alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, cyano, C 3 -C 6 cycloalkyl, C 3 -C 6 deuterated cycloalkyl, C 3 -C 6 cycloalkylmethyl, C 1 -C 3 alkylsulfonyl, C 1 -C 3 alkylthio, C 1 -C 3 deuterated alkylthio, C 3 -C 6 cycloalkylthio, halogen-substituted C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 1 -C 3 deuterated alkoxy, halogen-substituted C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyloxy, C 3 -C 6 deuterated cycloalkyloxy, halogen-substituted C 3 -C 6 -cycloalkyloxy, carbamoyl.

在其中一些实施例中,各R9和R10分别独立选自:氢、甲氧基、氟、二氟甲基、二氟甲氧基、氰基、甲基、乙炔基、氨基甲酰基、甲磺酰基、环丙氧基、甲硫基、氯。In some embodiments, each R 9 and R 10 is independently selected from the group consisting of hydrogen, methoxy, fluorine, difluoromethyl, difluoromethoxy, cyano, methyl, ethynyl, carbamoyl, methylsulfonyl, cyclopropyloxy, methylthio, and chlorine.

在其中一些实施例中,A选自:

In some embodiments, A is selected from:

在其中一些实施例中,各R11和R12分别独立选自:H,氘,卤素,C1-C3烷基,C1-C3氘代烷基,C3-C6环烷基,卤素取代的C1-C3烷基,氰基,C1-C3烷氧基。In some embodiments, each R 11 and R 12 is independently selected from the group consisting of: H, deuterium, halogen, C 1 -C 3 alkyl, C 1 -C 3 deuterated alkyl, C 3 -C 6 cycloalkyl, halogen-substituted C 1 -C 3 alkyl, cyano, and C 1 -C 3 alkoxy.

在其中一些实施例中,A1选自:
In some embodiments, A1 is selected from:

在其中一些实施例中,A1选自:
In some embodiments, A1 is selected from:

在其中一些实施例中,W选自:O,S,NH、NCH3In some embodiments, W is selected from: O, S, NH, NCH 3 .

在其中一些实施例中,W选自:S,NR7In some embodiments, W is selected from: S, NR 7 ;

其中,W中的R7选自:H,C1-C6烷基,C1-C6氘代烷基,C3-C8环烷基,卤素取代的C1-C6烷基,卤素取代的C3-C8环烷基。Wherein, R7 in W is selected from the group consisting of: H, C1 - C6 alkyl, C1 - C6 deuterated alkyl, C3 - C8 cycloalkyl, halogen-substituted C1 - C6 alkyl, and halogen-substituted C3 - C8 cycloalkyl.

在其中一些实施例中,R7和R8分别独立选自:H,氘,卤素,C1-C3烷基,C1-C3氘代烷基,C2-C3烯基,C2-C3炔基,C3-C6环烷基,卤素取代的C1-C3烷基,卤素取代的C3-C6环烷基,羟基,氰基,氨基,C1-C3烷基胺基;或者R8和A中的取代基相连形成取代或者未取代的5-6元碳环或杂环。In some embodiments, R7 and R8 are independently selected from: H, deuterium, halogen, C1 - C3 alkyl, C1 - C3 deuterated alkyl, C2 - C3 alkenyl, C2 -C3 alkynyl, C3 - C6 cycloalkyl, halogen-substituted C1-C3 alkyl, halogen-substituted C3-C6 cycloalkyl , hydroxyl , cyano, amino, C1 - C3 alkylamino; or R8 and the substituents in A are connected to form a substituted or unsubstituted 5-6 membered carbocyclic or heterocyclic ring.

在其中一些实施例中,R7和R8分别独立选自:H、氘、甲基、羟基,氰基,乙烯基、乙炔基、氨基、氘代甲基,或者R8和A中的取代基相连形成5-6元碳环或杂环。 In some embodiments, R7 and R8 are independently selected from: H, deuterium, methyl, hydroxyl, cyano, vinyl, ethynyl, amino, deuterated methyl, or R8 and the substituent in A are connected to form a 5-6 membered carbocyclic or heterocyclic ring.

在其中一些实施例中,Q选自:-N(CH3)-, In some embodiments, Q is selected from: -N(CH 3 )-,

在其中一些实施例中,R4选自:C1-C6烷基,C1-C6氘代烷基,卤素取代的C1-C6烷基。In some embodiments, R 4 is selected from: C 1 -C 6 alkyl, C 1 -C 6 deuterated alkyl, halogen-substituted C 1 -C 6 alkyl.

在其中一些实施例中,R1、R2、R3和R4分别独立选自:H,氘,C1-C3烷基,C1-C3氘代烷基,C2-C4烯基,C2-C4炔基,C3-C6环烷基,卤素取代的C3-C6环烷基,C3-C6环烷基甲基,卤素,卤素取代的C1-C3烷基,OH,羟基取代的C1-C3烷基,烷氧基取代的C1-C3烷基,氰基。In some embodiments, R 1 , R 2 , R 3 and R 4 are independently selected from the group consisting of: H, deuterium, C 1 -C 3 alkyl, C 1 -C 3 deuterated alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 6 cycloalkyl, halogen-substituted C 3 -C 6 cycloalkyl, C 3 -C 6 cycloalkylmethyl, halogen, halogen-substituted C 1 -C 3 alkyl, OH, hydroxy-substituted C 1 -C 3 alkyl, alkoxy-substituted C 1 -C 3 alkyl, and cyano.

在其中一些实施例中,R1选自:甲基、羟基、二氟甲基、三氟甲基、氰基、氯、氟、乙烯基、乙炔基、环丙基、氘代甲基。In some embodiments, R 1 is selected from: methyl, hydroxy, difluoromethyl, trifluoromethyl, cyano, chlorine, fluorine, vinyl, ethynyl, cyclopropyl, deuterated methyl.

在其中一些实施例中,R2选自:H、氟、甲基、氯、氘。In some embodiments, R 2 is selected from: H, fluorine, methyl, chlorine, deuterium.

在其中一些实施例中,R3选自:H、氟。In some embodiments, R 3 is selected from: H, fluorine.

在其中一些实施例中,R4选自:H、甲基、甲氧基甲基、一氟甲基。In some embodiments, R 4 is selected from: H, methyl, methoxymethyl, monofluoromethyl.

在其中一些实施例中,所述的螺杂环化合物选自如下化合物:





In some embodiments, the spiro heterocyclic compound is selected from the following compounds:





本发明还提供了所述螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子的应用,包括如下技术方案。The present invention also provides the use of the spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule, including the following technical solutions.

所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子在制备MC4受体拮抗剂中的应用。The use of the spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule in the preparation of MC4 receptor antagonist.

所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子在制备用于预防和/或治疗与MC4受体相关的疾病和/或症状的药物中的应用。Use of the spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule in the preparation of a drug for preventing and/or treating diseases and/or symptoms associated with the MC4 receptor.

在其中一些实施例中,与MC4受体相关的疾病和/或症状选自:与MC4受体相关的疾病和/或症状选自:恶病质(包括与慢性疾病、癌症、获得性免疫缺陷综合征(AIDS)、心力衰竭、慢性肾脏病(CKD)等相关的恶病质);厌食症或神经性厌食症(如老年性厌食症、与化疗、放疗相关的厌食症);恶心;呕吐;体重减轻(非自愿的体重减轻);少肌症;肌肉萎缩;肌肉无力;虚弱;骨骼疾病(如骨质流失);疼痛或神经性疼痛;焦虑(如创伤后应激障碍、PTSD等);性功能障碍;和炎症性疾病(如与恶病质、厌食症、少肌症等相关的炎症性疾病)等。In some of these embodiments, the diseases and/or symptoms associated with the MC4 receptor are selected from: diseases and/or symptoms associated with the MC4 receptor are selected from: cachexia (including cachexia associated with chronic diseases, cancer, acquired immune deficiency syndrome (AIDS), heart failure, chronic kidney disease (CKD) and the like); anorexia or anorexia nervosa (such as senile anorexia, anorexia associated with chemotherapy and radiotherapy); nausea; vomiting; weight loss (involuntary weight loss); sarcopenia; muscle atrophy; muscle weakness; weakness; bone diseases (such as bone loss); pain or neuropathic pain; anxiety (such as post-traumatic stress disorder, PTSD, etc.); sexual dysfunction; and inflammatory diseases (such as inflammatory diseases associated with cachexia, anorexia, sarcopenia, etc.), etc.

本发明还提供了一种预防和/或治疗与MC4受体相关的疾病和/或症状的药物组合物,包括如下技术方案。The present invention also provides a pharmaceutical composition for preventing and/or treating diseases and/or symptoms associated with MC4 receptor, including the following technical solutions.

一种预防和/或治疗与MC4受体相关的疾病和/或症状的药物组合物,包括活性成分以及药学上可接受的辅料和/或载体,所述活性成分包括所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子。A pharmaceutical composition for preventing and/or treating diseases and/or symptoms associated with MC4 receptors, comprising an active ingredient and a pharmaceutically acceptable excipient and/or carrier, wherein the active ingredient comprises the spiroheterocyclic compound or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated product thereof or a prodrug molecule thereof.

本发明还提供了一种预防和/或治疗与MC4受体相关的疾病和/或症状的方法,所述方法包括:施用安全有效量本发明所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子;和/或,The present invention also provides a method for preventing and/or treating diseases and/or symptoms associated with MC4 receptors, the method comprising: administering a safe and effective amount of the spiroheterocyclic compound of the present invention or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated substance thereof or a prodrug molecule thereof; and/or,

施用安全有效量的本发明所述的药物组合物。Administer a safe and effective amount of the pharmaceutical composition of the present invention.

本发明具有以下有益效果:The present invention has the following beneficial effects:

本发明提供的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或 者其前药分子具有拮抗MC4受体活性的作用,可用于治疗与MC4受体相关的多种疾病,有较大的应用价值。The spiroheterocyclic compound provided by the present invention or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or The prodrug molecule has the effect of antagonizing the activity of MC4 receptor and can be used to treat various diseases related to MC4 receptor, which has great application value.

附图说明BRIEF DESCRIPTION OF THE DRAWINGS

图1为化合物100、化合物104及PF07258669口服给予小鼠后的血浆药时曲线图。FIG1 is a plasma drug-dose curve of Compound 100, Compound 104 and PF07258669 after oral administration to mice.

图2为化合物100、化合物104及PF07258669口服给予小鼠后的脑组织药时曲线图。FIG2 is a drug-time curve of the brain tissue of Compound 100, Compound 104 and PF07258669 after oral administration to mice.

具体实施方式DETAILED DESCRIPTION

本发明所述化合物中,当任何变量(例如R等)在任何组分中出现超过一次,则其每次出现的定义独立于其它每次出现的定义。同样,允许取代基及变量的组合,只要这种组合使化合物稳定。自取代基划入环系统的线表示所指的键可连接到任何能取代的环原子上。如果环系统为多环,其意味着这种键仅连接到邻近环的任何适当的碳原子上。要理解本领域普通技术人员可选择本发明化合物的取代基及取代型式而提供化学上稳定的并可通过本领域技术和下列提出的方法自可容易获得的原料容易的合成的化合物。如果取代基自身被超过一个基团取代,应理解这些基团可在相同碳原子上或不同碳原子上,只要使结构稳定。In the compounds described herein, when any variable (e.g., R, etc.) occurs more than once in any component, its definition at each occurrence is independent of the definition at every other occurrence. Likewise, combinations of substituents and variables are permitted as long as such combinations render the compound stable. Lines drawn from substituents into the ring system indicate that the indicated bond may be attached to any substitutable ring atom. If the ring system is polycyclic, it means that such bonds are attached only to any appropriate carbon atom of the adjacent ring. It is to be understood that one of ordinary skill in the art may select substituents and substitution patterns for the compounds of the invention to provide chemically stable compounds that can be readily synthesized from readily available raw materials by techniques in the art and the methods set forth below. If a substituent itself is substituted with more than one group, it is to be understood that these groups may be on the same carbon atom or on different carbon atoms as long as the structure is stable.

本文所用术语“烷基”意指包括具有特定碳原子数目的支链的和直链的饱和脂肪烃基。例如,“C1-C6烷基”中“C1-C6”的定义包括以直链或支链排列的具有1、2、3、4、5或6个碳原子的基团。例如,“C1-C6烷基”具体包括甲基、乙基、正丙基、异丙基、正丁基、叔丁基、异丁基、戊基、己基。The term "alkyl" as used herein is intended to include branched and straight chain saturated aliphatic hydrocarbon groups having a specific number of carbon atoms. For example, the definition of "C 1 -C 6 alkyl" includes groups having 1, 2, 3, 4 , 5 or 6 carbon atoms in a straight or branched arrangement. For example, "C 1 -C 6 alkyl " specifically includes methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, isobutyl, pentyl, hexyl.

术语“环烷基”指具有特定碳原子数目的单环饱和脂肪烃基。例如“环烷基”包括环丙基、环丁基、环戊基或环己基等。The term "cycloalkyl" refers to a monocyclic saturated aliphatic hydrocarbon group having a specific number of carbon atoms. For example, "cycloalkyl" includes cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, etc.

术语“烷氧基”指烷基与氧直接连接的基团,即具有-O-烷基结构的基团,如-OCH3、-OCH2CH3、-OCH2CH2CH3、-O-CH2CH(CH3)2、-OCH2CH2CH2CH3、-O-CH(CH3)2等。The term "alkoxy" refers to a group in which an alkyl group is directly linked to oxygen, i.e., a group having an -O - alkyl structure, such as -OCH3 , -OCH2CH3 , -OCH2CH2CH3 , -O - CH2CH ( CH3 ) 2 , -OCH2CH2CH2CH3 , -O -CH( CH3 ) 2 , and the like.

术语“杂环基”为饱和或部分不饱和的单环或多环环状取代基(并环、桥环或螺环),其中一个或多个环原子选自N、O或S(O)m(其中m是0-2的整数)的杂原子,其余环原子为碳,例如:吗啉基、哌啶基、四氢吡咯基、吡咯烷基、二氢咪唑基、二氢异噁唑基、二氢异噻唑基、二氢噁二唑基、二氢噁唑基、二氢吡嗪基、二氢吡唑基、二氢吡啶基、二氢嘧啶基、二氢吡咯基、二氢四唑基、二氢噻二唑基、二氢噻唑基、二氢噻吩基、二氢三唑基、二氢氮杂环丁烷基、四氢呋喃基、四氢噻吩基等,及其N-氧化物,杂环取代基的连接可通过碳原子或通过杂原子实现。The term "heterocyclyl" is a saturated or partially unsaturated monocyclic or polycyclic cyclic substituent (acyclic, bridged or spirocyclic) in which one or more ring atoms are selected from N, O or S(O)m (wherein m is an integer from 0 to 2) as a heteroatom, and the remaining ring atoms are carbon, for example: morpholinyl, piperidinyl, tetrahydropyrrolyl, pyrrolidinyl, dihydroimidazolyl, dihydroisoxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothiophenyl, dihydrotriazolyl, dihydroazetidinyl, tetrahydrofuranyl, tetrahydrothiophenyl and the like, and N-oxides thereof. The attachment of the heterocyclic substituent may be achieved through a carbon atom or through a heteroatom.

术语“杂芳基”指含有1个或多个选自O、N或S的杂原子的芳香环,本发明范围内的杂 芳基包括但不限于:喹啉基、吡唑基、吡咯基、噻吩基、呋喃基、吡啶基、嘧啶基、吡嗪基、三氮唑基、咪唑基、噁唑基、异噁唑基、哒嗪基、苯并呋喃基、苯并噻吩基、苯并恶唑、吲哚基等;“杂芳基”也理解为包括任何含有氮的杂芳基的N-氧化物衍生物。The term "heteroaryl" refers to an aromatic ring containing one or more heteroatoms selected from O, N or S. Aryl includes, but is not limited to, quinolyl, pyrazolyl, pyrrolyl, thienyl, furanyl, pyridinyl, pyrimidinyl, pyrazinyl, triazolyl, imidazolyl, oxazolyl, isoxazolyl, pyridazinyl, benzofuranyl, benzothiophenyl, benzoxazole, indolyl, and the like; "heteroaryl" is also understood to include the N-oxide derivative of any heteroaryl containing nitrogen.

术语“取代的”是指用指定取代基的基团置换特定结构中的氢基。The term "substituted" refers to the replacement of hydrogen radicals in a particular structure with the radical of a specified substituent.

正如本领域技术人员所理解的,本文中所用“卤素”(“halo”)或“卤”意指氯、氟、溴和碘。As will be appreciated by those skilled in the art, "halo" or "halo" as used herein refers to chlorine, fluorine, bromine and iodine.

除非另有定义,烷基、环烷基、芳基、杂芳基和杂环烷基取代基可为未被取代的或取代的。例如,C1-C6烷基可被一个、两个或三个选自OH、卤素、烷氧基、二烷基氨基或杂环基例如吗啉基、哌啶基等的取代基取代。Unless otherwise defined, alkyl, cycloalkyl, aryl, heteroaryl and heterocycloalkyl substituents may be unsubstituted or substituted. For example, C1-C6 alkyl may be substituted with one, two or three substituents selected from OH, halogen, alkoxy, dialkylamino or heterocyclic radicals such as morpholinyl, piperidinyl, etc.

本发明包括式I化合物的游离形式,也包括其药学上可接受的盐及立体异构体。本发明中一些特定的示例性化合物为非盐形式的胺类化合物,即“游离形式”。可通过常规化学方法自含有碱性部分或酸性部分的本发明化合物合成本发明的药学上可接受的盐。通常,通过离子交换色谱或通过游离碱和化学计算量或过量的所需盐形式的无机或有机酸在适当溶剂或多种溶剂的组合中反应制备碱性化合物的盐。类似的,通过和适当的无机或有机碱反应形成酸性化合物的盐。The present invention includes free forms of compounds of formula I, as well as pharmaceutically acceptable salts and stereoisomers thereof. Some specific exemplary compounds of the present invention are amine compounds in non-salt form, i.e., "free forms". Pharmaceutically acceptable salts of the present invention can be synthesized from compounds of the present invention containing a basic or acidic moiety by conventional chemical methods. Typically, salts of basic compounds are prepared by ion exchange chromatography or by reacting a free base with a stoichiometric amount or excess of an inorganic or organic acid in the desired salt form in an appropriate solvent or a combination of multiple solvents. Similarly, salts of acidic compounds are formed by reacting with an appropriate inorganic or organic base.

因此,本发明化合物的药学上可接受的盐包括通过碱性本发明化合物和无机或有机酸反应形成的本发明化合物的常规无毒盐。例如,常规的无毒盐包括得自无机酸例如盐酸、氢溴酸、硫酸、氨基磺酸、磷酸、硝酸等的盐,也包括自有机酸例如乙酸、丙酸、琥珀酸、乙醇酸、硬脂酸、乳酸、苹果酸、酒石酸、柠檬酸、抗坏血酸、扑酸、马来酸、羟基马来酸、苯乙酸、谷氨酸、苯甲酸、水杨酸、对氨基苯磺酸、2-乙酰氧基-苯甲酸、富马酸、甲苯磺酸、甲磺酸、乙烷二磺酸、草酸、羟乙基磺酸、三氟乙酸等制备的盐。Therefore, the pharmaceutically acceptable salts of the compounds of this invention include conventional non-toxic salts of the compounds of this invention formed by reacting alkaline compounds of this invention with inorganic or organic acids. For example, conventional non-toxic salts include salts derived from inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, phosphoric acid, nitric acid, etc., and also include salts prepared from organic acids such as acetic acid, propionic acid, succinic acid, glycolic acid, stearic acid, lactic acid, malic acid, tartaric acid, citric acid, ascorbic acid, pamoic acid, maleic acid, hydroxymaleic acid, phenylacetic acid, glutamic acid, benzoic acid, salicylic acid, p-aminobenzenesulfonic acid, 2-acetoxy-benzoic acid, fumaric acid, toluenesulfonic acid, methanesulfonic acid, ethanedisulfonic acid, oxalic acid, isethionic acid, trifluoroacetic acid, etc.

如果本发明化合物为酸性的,则适当的“药学上可接受的盐”指通过药学上可接受的无毒碱包括无机碱及有机碱制备的盐。得自无机碱的盐包括铝盐、铵盐、钙盐、铜盐、铁盐、亚铁盐、锂盐、镁盐、锰盐、亚锰盐、钾盐、钠盐、锌盐等。特别优选铵盐、钙盐、镁盐、钾盐和钠盐。得自药学上可接受的有机无毒碱的盐,所述碱包括伯胺、仲胺和叔胺的盐,取代的胺包括天然存在的取代胺、环状胺及碱性离子交换树脂例如精氨酸、甜菜碱、咖啡因、胆碱、N,N'-二苄基乙二胺、二乙胺、2-二乙基氨基乙醇、2-二甲基氨基乙醇、氨基乙醇、乙醇胺、乙二胺、N-乙基吗啉、N-乙基哌啶、葡萄糖胺、氨基葡萄糖、组氨酸、羟钴胺、异丙基胺、赖氨酸、甲基葡萄糖胺、吗啉、哌嗪,哌啶、呱咤、多胺树脂、普鲁卡因、嘌呤、可可碱、三乙胺、三甲胺、三丙胺、氨基丁三醇等。If the compound of the present invention is acidic, then suitable "pharmaceutically acceptable salts" refer to salts prepared from pharmaceutically acceptable non-toxic bases including inorganic bases and organic bases. Salts derived from inorganic bases include aluminum salts, ammonium salts, calcium salts, copper salts, iron salts, ferrous salts, lithium salts, magnesium salts, manganic salts, manganous salts, potassium salts, sodium salts, zinc salts, etc. Ammonium salts, calcium salts, magnesium salts, potassium salts and sodium salts are particularly preferred. Salts derived from pharmaceutically acceptable organic non-toxic bases, including salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins such as arginine, betaine, caffeine, choline, N,N'-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, aminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucosamine, glucosamine, histidine, hydroxocobalamin, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, guaiac, polyamine resins, procaine, purine, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.

Berg等,“Pharmaceutical Salts,”J.Pharm.Sci.’1977:66:1-19更详细描述了上文所述药学上可接受的盐及其它典型的药学上可接受的盐的制备。 Berg et al., "Pharmaceutical Salts," J. Pharm. Sci. '1977: 66: 1-19, describes in more detail the preparation of the above-mentioned pharmaceutically acceptable salts and other typical pharmaceutically acceptable salts.

本发明所述立体异构体,即(取决于其结构)作为对映体、非对映体、顺型/反型异构体(syn-/anti-isomer)、顺式/反式(cis-/trans-isomer)异构体、差向异构体以及(E)-/(Z)-异构体。式I化合物可以以纯立体异构体的形式或者以立体异构体的任何混合物的形式用于本发明的上下文中,在后一种情况中优选外消旋体。The stereoisomers of the present invention are, i.e. (depending on their structure) as enantiomers, diastereomers, syn-/anti-isomers, cis-/trans-isomers, epimers and (E)-/(Z)-isomers. The compounds of formula I can be used in the context of the present invention in the form of pure stereoisomers or in the form of any mixture of stereoisomers, in the latter case the racemates are preferred.

本发明所涉及的化合物及其药学可接受的盐的代谢产物,以及可以在体内转变为本发明所涉及的化合物及其药学可接受的盐的结构的前药,也包含在本发明的权利要求中。The metabolites of the compounds involved in the present invention and their pharmaceutically acceptable salts, as well as prodrugs that can be converted into the structures of the compounds involved in the present invention and their pharmaceutically acceptable salts in vivo, are also included in the claims of the present invention.

本发明还提供了一种预防和/或治疗与MC4受体相关的疾病和/或症状的药物组合物,它包含安全有效量范围内的活性成分,以及药学上可接受的辅料。The present invention also provides a pharmaceutical composition for preventing and/or treating diseases and/or symptoms associated with MC4 receptors, which comprises an active ingredient within a safe and effective amount range and a pharmaceutically acceptable adjuvant.

本发明所述的“活性成分”是指本发明所述的式I化合物、或者其立体异构体、或者其药学上可接受的盐、或者其溶剂合物、或者其前药分子、或者其氘化物、或者其氚化物。The "active ingredient" described in the present invention refers to the compound of formula I described in the present invention, or its stereoisomer, or its pharmaceutically acceptable salt, or its solvate, or its prodrug molecule, or its deuteride, or its tritiated product.

“安全有效量”指的是:活性成分的量足以明显改善病情,而不至于产生严重的副作用。"Safe and effective amount" means that the amount of active ingredient is sufficient to significantly improve the condition without causing serious side effects.

使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选20~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。When using the pharmaceutical composition, a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, wherein the dosage during administration is a pharmaceutically effective dosage, and for a person weighing 60 kg, the daily dosage is usually 1 to 2000 mg, preferably 20 to 500 mg. Of course, the specific dosage should also take into account factors such as the route of administration and the health status of the patient, which are all within the skill of a skilled physician.

“药学上可接受的辅料”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。"Pharmaceutically acceptable excipients" refer to: one or more compatible solid or liquid fillers or gel substances, which are suitable for human use and must be sufficiently pure and sufficiently low in toxicity.

“相容性”在此指的是组合物中各组分能和本发明的活性成分以及它们之间相互掺和,而不明显降低活性成分的药效。"Compatibility" herein means that the components in the composition can be blended with the active ingredient of the present invention and with each other without significantly reducing the efficacy of the active ingredient.

药学上可以接受的辅料部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。Examples of pharmaceutically acceptable excipients include cellulose and its derivatives (such as sodium carboxymethyl cellulose, sodium ethyl cellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid, magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers (such as ), wetting agents (such as sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.

在另一优选例中,本发明式I化合物可与大分子化合物或高分子通过非键合作用形成复合物。在另一优选例中,本发明式I化合物作为小分子还可通过化学键与大分子化合物或高分子相连接。所述大分子化合物可以是生物大分子如高聚糖、蛋白、核酸、多肽等。In another preferred embodiment, the compound of formula I of the present invention can form a complex with a macromolecular compound or a polymer through a non-bonding reaction. In another preferred embodiment, the compound of formula I of the present invention as a small molecule can also be connected to a macromolecular compound or a polymer through a chemical bond. The macromolecular compound can be a biological macromolecule such as a high polysaccharide, protein, nucleic acid, polypeptide, etc.

本发明的活性成分或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、瘤内、直肠、肠胃外(静脉内、肌肉内或皮下)等。There is no particular limitation on the administration of the active ingredient or pharmaceutical composition of the present invention. Representative administration methods include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), etc.

用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.

在这些固体剂型中,活性成分与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或 磷酸二钙,或与下述成分混合:In these solid dosage forms, the active ingredient is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or Dicalcium phosphate, or in combination with:

(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid;

(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和阿拉伯胶;(b) binders, for example, hydroxymethylcellulose, alginate, gelatin, polyvinyl pyrrolidone, sucrose and gum arabic;

(c)保湿剂,例如,甘油;(c) humectants, for example, glycerin;

(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;(d) disintegrants, for example, agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate;

(e)缓溶剂,例如石蜡;(e) a buffering solvent, such as paraffin;

(f)吸收加速剂,例如,季胺化合物;(f) absorption accelerators, for example, quaternary ammonium compounds;

(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(g) wetting agents, such as cetyl alcohol and glyceryl monostearate;

(h)吸附剂,例如,高岭土;和(h) adsorbents, for example, kaolin; and

(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。(i) Lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage form may also contain a buffering agent.

所述的固体剂型还可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性成分的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。The solid dosage forms can also be prepared using coatings and shell materials, such as enteric coatings and other materials known in the art. They can contain opacifying agents, and the release of the active ingredient in such compositions can be delayed in a certain part of the digestive tract. Examples of embedding components that can be used are polymeric substances and waxes.

用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性成分外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active ingredient, the liquid dosage form may contain inert diluents conventionally used in the art, such as water or other solvents, solubilizers and emulsifiers, for example, ethanol, isopropanol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butylene glycol, dimethylformamide and oils, in particular cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances. In addition to these inert diluents, the composition may also contain adjuvants, such as wetting agents, emulsifiers and suspending agents, sweeteners, flavoring agents and spices.

除了活性成分外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。In addition to the active ingredients, suspensions may contain suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methanol and agar, or mixtures of these substances.

用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。Compositions for parenteral injection may include physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.

合成方法:除在文献中已知的或在实验程序中例证的标准方法外,可采用如下合成方案(方案一-五)中的方法制备本发明化合物。结合下述的合成方案,能够对本发明中所述的化合物以及合成方法进行更好的理解。所述的合成方案描述了可以用于制备本发明中所述的化合物的方法,所述的方法仅仅是为说明目的的说明性方案描述,并不构成对本发明所具有的 范围的限制。
Synthetic Methods: In addition to standard methods known in the literature or exemplified in the experimental procedures, the methods in the following synthetic schemes (Schemes 1-5) can be used to prepare the compounds of the present invention. In conjunction with the following synthetic schemes, the compounds and synthetic methods described in the present invention can be better understood. The synthetic schemes described describe methods that can be used to prepare the compounds described in the present invention. The described methods are merely illustrative scheme descriptions for illustrative purposes and do not constitute an endorsement of the present invention. Limitation of scope.

方案一
Solution 1

方案二
Solution 2

方案三
Option 3

方案四
Option 4

方案五Option 5

实施例1:(S)-N-(5-氟-2-甲氧基吡啶-4-基)-N,7'-二甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酰胺(化合物1)的制备(按照方案一和二线路制备)Example 1: Preparation of (S)-N-(5-fluoro-2-methoxypyridin-4-yl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxamide (Compound 1) (Prepared according to Scheme 1 and 2)

步骤1a:1-苄基-3-((三甲基甲硅烷基)乙炔基)吡咯烷-3-醇(化合物0102-1)的制备:氮气保护下,在-70℃下,向乙炔基三甲基硅烷(6.73克,68.48毫摩尔,1.2当量)的四氢呋喃(50毫升)溶液中滴加正丁基锂(25.11毫升,62.78毫摩尔,1.1当量),混合物在-70℃下搅拌1.0小时。然后在-60℃下向反应液中滴加1-苄基吡咯烷-3-酮(化合物0101-1)(10.0克,57.07毫摩尔,1.0当量)的四氢呋喃(20毫升)溶液,撤去低温装置,并使混合物在室温下搅拌过夜。反应液用氯化铵溶液淬灭并用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=5/1至3/1)纯化,得到黄色油状产物1-苄基-3-((三甲基甲硅烷基)乙炔基)吡咯烷-3-醇(12.11克,收率:77.63%)。Step 1a: Preparation of 1-benzyl-3-((trimethylsilyl)ethynyl)pyrrolidin-3-ol (Compound 0102-1): Under nitrogen protection, n-butyl lithium (25.11 ml, 62.78 mmol, 1.1 eq) was added dropwise to a solution of ethynyltrimethylsilane (6.73 g, 68.48 mmol, 1.2 eq) in tetrahydrofuran (50 ml) at -70°C, and the mixture was stirred at -70°C for 1.0 hour. Then, a solution of 1-benzylpyrrolidin-3-one (Compound 0101-1) (10.0 g, 57.07 mmol, 1.0 eq) in tetrahydrofuran (20 ml) was added dropwise to the reaction solution at -60°C, the cryogenic device was removed, and the mixture was stirred at room temperature overnight. The reaction solution was quenched with ammonium chloride solution and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate=5/1 to 3/1) to give yellow oily product 1-benzyl-3-((trimethylsilyl)ethynyl)pyrrolidin-3-ol (12.11 g, yield: 77.63%).

步骤1b:1-苄基-3-乙炔基吡咯烷-3-醇(化合物0103-1)的制备:氮气保护下,向1-苄基-3-((三甲基甲硅烷基)乙炔基)吡咯烷-3-醇(0102-1)(12.11克,44.36毫摩尔,1.0当量)的甲醇(100毫升)溶液中加入碳酸钾(12.24克,88.72毫摩尔,2.0当量),混合物在室温下搅拌1.5小时。减压除去溶剂,残留物加水稀释并用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(乙酸乙酯)纯化,得到黄色油状产物1-苄基-3-乙炔基吡咯烷-3-醇(7.65克,收率:85.68%)。Step 1b: Preparation of 1-benzyl-3-ethynylpyrrolidin-3-ol (Compound 0103-1): Under nitrogen protection, potassium carbonate (12.24 g, 88.72 mmol, 2.0 eq.) was added to a solution of 1-benzyl-3-((trimethylsilyl)ethynyl)pyrrolidin-3-ol (0102-1) (12.11 g, 44.36 mmol, 1.0 eq.) in methanol (100 ml), and the mixture was stirred at room temperature for 1.5 hours. The solvent was removed under reduced pressure, the residue was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (ethyl acetate) to obtain a yellow oily product 1-benzyl-3-ethynylpyrrolidin-3-ol (7.65 g, yield: 85.68%).

步骤1c:1-苄基-3-乙炔基吡咯烷-3-基乙酸酯(化合物0104-1)的制备:氮气保护下,0℃下,向1-苄基-3-乙炔基吡咯烷-3-醇(0103-1)(7.65克,38.05毫摩尔,1.0当量)和N,N-二异丙基乙胺(9.82克,76.1毫摩尔,2.0当量)的二氯甲烷(100毫升)溶液中加入乙酰氯(5.08克,64.7 毫摩尔,1.7当量)的二氯甲烷(6毫升)溶液,混合物在0℃下搅拌0.5小时。反应液加水淬灭,并用二氯甲烷萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=4/1至1/1)纯化,得到黄色油状产物1-苄基-3-乙炔基吡咯烷-3-基乙酸酯(8.94克,收率:96.70%)。Step 1c: Preparation of 1-benzyl-3-ethynylpyrrolidin-3-yl acetate (Compound 0104-1): Under nitrogen protection, acetyl chloride (5.08 g, 64.7 mmol, 1.0 equiv) was added to a solution of 1-benzyl-3-ethynylpyrrolidin-3-ol (0103-1) (7.65 g, 38.05 mmol, 1.0 equiv) and N,N-diisopropylethylamine (9.82 g, 76.1 mmol, 2.0 equiv) in dichloromethane (100 ml) at 0°C. The mixture was stirred at 0°C for 0.5 hours. The reaction solution was quenched with water and extracted with dichloromethane. The organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate = 4/1 to 1/1) to obtain a yellow oily product 1-benzyl-3-ethynylpyrrolidin-3-yl acetate (8.94 g, yield: 96.70%).

步骤1d:1-苄基-3-乙炔基-N-(4-甲氧基苄基)吡咯烷-3-胺(化合物0105-1)的制备:氮气保护下,70℃下,向4-甲氧基苄胺(10.06克,73.32毫摩尔,2.0当量)和氯化亚铜(363毫克,3.67毫摩尔,0.1当量)的四氢呋喃(60毫升)溶液中加入1-苄基-3-乙炔基吡咯烷-3-基乙酸酯(0104-1)(8.92克,36.66毫摩尔,1.0当量)的四氢呋喃(18毫升)溶液,混合物在70℃下搅拌20分钟。反应液加水稀释并用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=4/1至1/1)纯化,得到黄色油状产物1-苄基-3-乙炔基-N-(4-甲氧基苄基)吡咯烷-3-胺(3.5克,收率:29.83%)。LCMS(ESI):m/z=321[M+1]+.Step 1d: Preparation of 1-benzyl-3-ethynyl-N-(4-methoxybenzyl)pyrrolidin-3-amine (Compound 0105-1): Under nitrogen protection, a solution of 1-benzyl-3-ethynylpyrrolidin-3-yl acetate (0104-1) (8.92 g, 36.66 mmol, 1.0 eq.) in tetrahydrofuran (18 ml) was added to a solution of 4-methoxybenzylamine (10.06 g, 73.32 mmol, 2.0 eq.) and cuprous chloride (363 mg, 3.67 mmol, 0.1 eq.) in tetrahydrofuran (60 ml) at 70° C., and the mixture was stirred at 70° C. for 20 minutes. The reaction solution was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate = 4/1 to 1/1) to obtain a yellow oily product 1-benzyl-3-ethynyl-N-(4-methoxybenzyl)pyrrolidin-3-amine (3.5 g, yield: 29.83%). LCMS (ESI): m/z = 321 [M+1] + .

步骤1e:2-氯-3-碘-6-甲基吡啶(化合物0107-1)的制备:在0℃下往2-氯-6-甲基吡啶-3-胺(0106-1)(25.0克,175.33毫摩尔,1.0当量)在6M稀盐酸(175毫升)的混合物中逐滴加入亚硝酸钠(14.52克,210.39毫摩尔,1.2当量)的水(50毫升)溶液。混合物在0℃下搅拌0.5小时。逐滴加入碘化钾(58.18克,350.46毫摩尔,2.0当量)的水(100毫升)溶液。混合物升到室温然后搅拌过夜。缓慢加入2M氢氧化钠溶液调节pH=8,然后水层用乙酸乙酯(100毫升×2)萃取。合并的有机层用饱和食盐水(100毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯=60∶1至10:1),得到淡黄色固体2-氯-3-碘-6-甲基吡啶(37.67克,收率:85%)。LCMS(ESI):m/z 254[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=20:1)。Step 1e: Preparation of 2-chloro-3-iodo-6-methylpyridine (Compound 0107-1): To a mixture of 2-chloro-6-methylpyridin-3-amine (0106-1) (25.0 g, 175.33 mmol, 1.0 eq) in 6M dilute hydrochloric acid (175 ml) was added dropwise a solution of sodium nitrite (14.52 g, 210.39 mmol, 1.2 eq) in water (50 ml) at 0°C. The mixture was stirred at 0°C for 0.5 h. A solution of potassium iodide (58.18 g, 350.46 mmol, 2.0 eq) in water (100 ml) was added dropwise. The mixture was warmed to room temperature and then stirred overnight. 2M sodium hydroxide solution was slowly added to adjust pH=8, and then the aqueous layer was extracted with ethyl acetate (100 ml×2). The combined organic layers were washed with saturated brine (100 ml×1), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate = 60: 1 to 10: 1) to give a pale yellow solid 2-chloro-3-iodo-6-methylpyridine (37.67 g, yield: 85%). LCMS (ESI): m/z 254 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 20: 1).

步骤1f:1-苄基-3-((2-氯-6-甲基吡啶-3-基)乙炔基)-N-(4-甲氧基苄基)吡咯烷-3-胺(化合物0108-1)的制备:往2-氯-3-碘-6-甲基吡啶(0107-1)(35.78克,141.43毫摩尔,1.0当量),双三苯基磷二氯化钯(4.96克,7.1毫摩尔,0.05当量)和碘化亚铜(1.35克,7.1毫摩尔,0.05当量)在三乙胺(220毫升)的混合物中加入1-苄基-3-乙炔基-N-(4-甲氧基苄基)吡咯烷-3-胺(0105-1)(45.25克,141.43毫摩尔,1.0当量)。氮气氛围下,混合物在80℃搅拌过夜。减压下除去溶剂。残余物在硅胶上进行柱色谱分离(石油醚:乙酸乙酯=10:1 to 1:2),得到棕色油状物1-苄基-3-((2-氯-6-甲基吡啶-3-基)乙炔基)-N-(4-甲氧基苄基)吡咯烷-3-胺(54.66克,收率:87%)。LCMS(ESI):m/z 446[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=2:1)。Step 1f: Preparation of 1-benzyl-3-((2-chloro-6-methylpyridin-3-yl)ethynyl)-N-(4-methoxybenzyl)pyrrolidin-3-amine (Compound 0108-1): To a mixture of 2-chloro-3-iodo-6-methylpyridine (0107-1) (35.78 g, 141.43 mmol, 1.0 eq), bistriphenylphosphine palladium dichloride (4.96 g, 7.1 mmol, 0.05 eq) and cuprous iodide (1.35 g, 7.1 mmol, 0.05 eq) in triethylamine (220 ml) was added 1-benzyl-3-ethynyl-N-(4-methoxybenzyl)pyrrolidin-3-amine (0105-1) (45.25 g, 141.43 mmol, 1.0 eq). The mixture was stirred at 80° C. overnight under nitrogen atmosphere. The solvent was removed under reduced pressure. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate = 10: 1 to 1: 2) to give a brown oily substance 1-benzyl-3-((2-chloro-6-methylpyridin-3-yl)ethynyl)-N-(4-methoxybenzyl)pyrrolidin-3-amine (54.66 g, yield: 87%). LCMS (ESI): m/z 446 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 2: 1).

步骤1g:1-苄基-3-(2-(2-氯-6-甲基吡啶-3-基)乙基)-N-(4-甲氧基苄基)吡咯烷-3-胺(化合物0109-1)的制备:往1-苄基-3-((2-氯-6-甲基吡啶-3-基)乙炔基)-N-(4-甲氧基苄基)吡咯烷-3-胺(0108-1)(54.66克,122.56毫摩尔,1.0当量)在甲醇(300毫升)的混合物中加入二氧化铂(4.17克,18.38毫摩尔,0.15当量)。混合物在氢气球压力下室温搅拌10小时。混合物过滤。滤液减压浓缩,得到棕色油状物1-苄基-3-(2-(2-氯-6-甲基吡啶-3-基)乙基)-N-(4-甲氧基苄基)吡咯烷-3- 胺(53.5克,粗品)。LCMS(ESI):m/z 450[M+1]+;TLC:Rf 0.3(石油醚:乙酸乙酯=2:1)。Step 1g: Preparation of 1-benzyl-3-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-N-(4-methoxybenzyl)pyrrolidin-3-amine (Compound 0109-1): Platinum dioxide (4.17 g, 18.38 mmol, 0.15 eq) was added to a mixture of 1-benzyl-3-((2-chloro-6-methylpyridin-3-yl)ethynyl)-N-(4-methoxybenzyl)pyrrolidin-3-amine (0108-1) (54.66 g, 122.56 mmol, 1.0 eq) in methanol (300 ml). The mixture was stirred at room temperature under hydrogen balloon pressure for 10 hours. The mixture was filtered. The filtrate was concentrated under reduced pressure to give a brown oily substance: 1-benzyl-3-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-N-(4-methoxybenzyl)pyrrolidine-3-yl Amine (53.5 g, crude product). LCMS (ESI): m/z 450 [M+1] + ; TLC: Rf 0.3 (petroleum ether:ethyl acetate=2:1).

步骤1h:1-苄基-1’-(4-甲氧基苄基)-7’-甲基-3',4'-二氢-1’H-螺(吡咯烷-3,2’-[1,8]萘啶(化合物0110-1)的制备:往1-苄基-3-(2-(2-氯-6-甲基吡啶-3-基)乙基)-N-(4-甲氧基苄基)吡咯烷-3-胺(0109-1)(42.18克,93.94毫摩尔,1.0当量),醋酸钯(1.05克,4.70毫摩尔,0.05当量)和RuPhos(4.38克,1.0毫摩尔,0.1当量)在二氧六环(420毫升)的混合物中加入叔丁醇钠(18.04克,187.88毫摩尔,2.0当量)。混合物在氮气氛围下加热至115℃反应过夜。减压下除去溶剂。残余物用水(200毫升)稀释。水层用乙酸乙酯(150毫升×3)萃取。合并的有机层用饱和食盐水(100毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯=20∶1至5:1),得到淡黄色固体1-苄基-1’-(4-甲氧基苄基)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶](21.0克,收率:54%)。LCMS(ESI):m/z 414[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=5:1)。Step 1h: Preparation of 1-benzyl-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'H-spiro(pyrrolidine-3,2'-[1,8]naphthyridine (Compound 0110-1): To a mixture of 1-benzyl-3-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-N-(4-methoxybenzyl)pyrrolidin-3-amine (0109-1) (42.18 g, 93.94 mmol, 1.0 eq), palladium acetate (1.05 g, 4.70 mmol, 0.05 eq) and RuPhos (4.38 g, 1.0 mmol, 0.1 eq) in dioxane (420 ml) was added sodium tert-butoxide (18.04 g, 1 87.88 mmol, 2.0 eq.). The mixture was heated to 115°C under a nitrogen atmosphere and reacted overnight. The solvent was removed under reduced pressure. The residue was diluted with water (200 ml). The aqueous layer was extracted with ethyl acetate (150 ml×3). The combined organic layer was washed with saturated brine (100 ml×1), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate = 20:1 to 5:1) to give a light yellow solid 1-benzyl-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] (21.0 g, yield: 54%). LCMS (ESI): m/z 414 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 5:1).

步骤1i:1-苄基-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶](化合物0111-1)的制备:在0℃下往1-苄基-1’-(4-甲氧基苄基)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶](0110-1)(21.0克,50.85毫摩尔,1.0当量)在二氯甲烷(150毫升)的混合物中逐滴加入三氟乙酸(57.98克,508.5毫摩尔,10.0当量)。将混合物升温至室温,并搅拌6小时。减压下除去溶剂。混合物用水(200毫升)稀释。加入碳酸钠固体调节pH=10,然后水层用二氯甲烷(100毫升×3)萃取。合并的有机层用饱和食盐水(100毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(二氯甲烷∶甲醇=80∶1至20:1),得到淡黄色油状物1-苄基-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶](14.9克,收率:100%)。LCMS(ESI):m/z 294[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=20:1)。Step 1i: Preparation of 1-benzyl-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] (Compound 0111-1): To a mixture of 1-benzyl-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] (0110-1) (21.0 g, 50.85 mmol, 1.0 equiv) in dichloromethane (150 mL) was added dropwise trifluoroacetic acid (57.98 g, 508.5 mmol, 10.0 equiv) at 0°C. The mixture was warmed to room temperature and stirred for 6 hours. The solvent was removed under reduced pressure. The mixture was diluted with water (200 mL). Solid sodium carbonate was added to adjust pH=10, and then the aqueous layer was extracted with dichloromethane (100 ml×3). The combined organic layer was washed with saturated brine (100 ml×1), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (dichloromethane: methanol=80:1 to 20:1) to obtain a pale yellow oily substance 1-benzyl-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] (14.9 g, yield: 100%). LCMS (ESI): m/z 294 [M+1] + ; TLC: Rf 0.5 (dichloromethane: methanol=20:1).

步骤1j:7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0112-1)的制备:往1-苄基-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶](0111-1)(14.9克,50.85毫摩尔,1.0当量)和二碳酸二叔丁酯(22.2克,101.7毫摩尔,2.0当量)在四氢呋喃(150毫升)的混合物中加入氢氧化钯/碳(10%,3.73克)。混合物在氢气球压力下加热到60℃反应40小时。混合物过滤。滤液减压浓缩。残余物在硅胶上进行柱色谱分离(二氯甲烷∶乙酸乙酯10∶1至1:1),得到淡黄色固体(S)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(13.5克,收率:87%)。LCMS(ESI):m/z 305[M+1]+;TLC:Rf 0.5(二氯甲烷:乙酸乙酯=1:1)。Step 1j: Preparation of tert-butyl 7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0112-1): Palladium hydroxide/carbon (10%, 3.73 g) was added to a mixture of 1-benzyl-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] (0111-1) (14.9 g, 50.85 mmol, 1.0 equiv) and di-tert-butyl dicarbonate (22.2 g, 101.7 mmol, 2.0 equiv) in tetrahydrofuran (150 ml). The mixture was heated to 60°C under hydrogen balloon pressure for 40 hours. The mixture was filtered. The filtrate was concentrated under reduced pressure. The residue was subjected to column chromatography on silica gel (dichloromethane: ethyl acetate 10: 1 to 1: 1) to give (S)-tert-butyl 7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (13.5 g, yield: 87%) as a pale yellow solid. LCMS (ESI): m/z 305 [M+1] + ; TLC: Rf 0.5 (dichloromethane: ethyl acetate = 1: 1).

步骤1k:(S)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0113-1)和(R)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0114-1)的制备:7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0112-1)对映体的分离通过超临界流体色谱法进行(色谱柱:ChiralPak IC,250×30mm I.D.,10μm;流动相:A为二氧化碳, B为乙醇;洗脱剂:30%B;流速:150毫升/分钟;背压:100巴;柱温:38℃)。第一个洗脱的对映体是(R)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0114-1)(6.32克,产率:47%),第二个洗脱的对映体是(S)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0113-1)(6.29克,产率:47%)。分析条件(色谱柱:ChiralPak IC,100×4.6mm I.D.,3μm;流动相:A为二氧化碳,B为乙醇(0.05%二乙胺);洗脱剂:40%B;流速:2.5毫升/分钟;背压:100巴;柱温:35℃)Step 1k: Preparation of (S)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0113-1) and (R)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0114-1): The enantiomers of 7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0112-1) were separated by supercritical fluid chromatography (chromatographic column: ChiralPak IC, 250×30 mm ID, 10 μm; mobile phase: A is carbon dioxide, B is ethanol; eluent: 30% B; flow rate: 150 ml/min; back pressure: 100 bar; column temperature: 38°C). The first eluting enantiomer is (R)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (compound 0114-1) (6.32 g, yield: 47%), and the second eluting enantiomer is (S)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (compound 0113-1) (6.29 g, yield: 47%). Analytical conditions (chromatographic column: ChiralPak IC, 100×4.6 mm ID, 3 μm; mobile phase: A is carbon dioxide, B is ethanol (0.05% diethylamine); eluent: 40% B; flow rate: 2.5 ml/min; back pressure: 100 bar; column temperature: 35°C)

步骤1l:(S)-6’-溴-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0116-1)的制备:在0℃下往(S)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0113-1)(6.29克,20.76毫摩尔,1.0当量)在二氯甲烷(60毫升)的混合物中加入二溴海因(2.97克,10.38毫摩尔,0.5当量)。混合物在0℃下搅拌1小时。加入饱和亚硫酸钠水溶液(50毫升)淬灭反应。水层用二氯甲烷(50毫升×3)萃取。合并的有机层用饱和食盐水(50毫升×1)洗涤,经无水硫酸钠干燥并浓缩,得到淡黄色固体(S)-6’-溴-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(7.74克,收率:98%)。LCMS(ESI):m/z 382[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。Step 11: Preparation of (S)-6'-bromo-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0116-1): To a mixture of (S)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0113-1) (6.29 g, 20.76 mmol, 1.0 eq) in dichloromethane (60 mL) was added dibromohydantoin (2.97 g, 10.38 mmol, 0.5 eq) at 0°C. The mixture was stirred at 0°C for 1 hour. Saturated aqueous sodium sulfite solution (50 mL) was added to quench the reaction. The aqueous layer was extracted with dichloromethane (50 mL×3). The combined organic layers were washed with saturated brine (50 mL x 1), dried over anhydrous sodium sulfate and concentrated to give a pale yellow solid (S)-6'-bromo-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (7.74 g, yield: 98%). LCMS (ESI): m/z 382 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=3:1).

步骤1m:(S)-(1-(叔丁氧羰基)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(化合物0118-1)的制备:往(S)-6’-溴-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0116-1)(5.75克,15.05毫摩尔,1.0当量),Xphos-Pd-G2(237毫克,0.30mmol,0.02当量),Xphos(215毫克,0.45mmol,0.03当量),乙酸钾(3.69克,37.63毫摩尔,2.5当量)和叔丁醇钠(36毫克,0.38毫摩尔,0.025当量)在甲醇(60毫升)和乙二醇(6毫升)的混合物中加入四羟基二硼(3.37克,37.63毫摩尔,2.5当量)。氮气氛围下,混合物在53℃搅拌过夜。减压下除去溶剂。残余物用水(50毫升)稀释。加入2M氢氧化钠溶液调节pH=12,然后混合物用甲基叔丁基醚(30毫升×3)洗涤。往水层加入2M稀盐酸溶液调节pH=8,然后用乙酸乙酯(50毫升×3)萃取。合并的有机层用饱和食盐水(30毫升×1)洗涤,经无水硫酸钠干燥并浓缩,得到白色固体(S)-(1-(叔丁氧羰基)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(3.58克,收率:69%)。LCMS(ESI):m/z 348[M+1]+;TLC:Rf 0.3(二氯甲烷:甲醇=10:1)。Step 1m: Preparation of (S)-(1-(tert-Butyloxycarbonyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (Compound 0118-1): to (S)-6'-bromo-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0116-1) (5.75 g, 15.05 mmol, 1.0 equiv), Xphos-Pd-G2 (237 mg, 0.30 mmol, 0.02 eq.), Xphos (215 mg, 0.45 mmol, 0.03 eq.), potassium acetate (3.69 g, 37.63 mmol, 2.5 eq.) and sodium tert-butoxide (36 mg, 0.38 mmol, 0.025 eq.) were added to a mixture of methanol (60 ml) and ethylene glycol (6 ml). Tetrahydroxydiboron (3.37 g, 37.63 mmol, 2.5 eq.) was added. Under nitrogen atmosphere, the mixture was stirred at 53°C overnight. The solvent was removed under reduced pressure. The residue was diluted with water (50 ml). 2M sodium hydroxide solution was added to adjust pH = 12, and then the mixture was washed with methyl tert-butyl ether (30 ml × 3). 2M dilute hydrochloric acid solution was added to the aqueous layer to adjust pH = 8, and then extracted with ethyl acetate (50 ml × 3). The combined organic layer was washed with saturated brine (30 mL x 1), dried over anhydrous sodium sulfate and concentrated to give a white solid (S)-(1-(tert-butyloxycarbonyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (3.58 g, yield: 69%). LCMS (ESI): m/z 348 [M+1] + ; TLC: Rf 0.3 (dichloromethane:methanol=10:1).

步骤1n:(S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0120-1)的制备:往(S)-(1-(叔丁氧羰基)-7’-甲基-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(0118-1)(3.58克,10.31毫摩尔,1.0当量),Xphos-Pd-G2(243毫克,0.31mmol,0.03当量),Xphos(147毫克,0.31毫摩尔,0.03当量)和碳酸钠(3.28克,30.93毫摩尔,3.0当量)在甲苯(25毫升),乙醇(25毫升)和水(10毫升)的混合物中加入2-溴嘧啶(1.80克,11.34毫摩尔,1.1当量)。氮气氛围下,混合物在55℃搅拌过夜。减压下除去溶剂。残余物用水(50 毫升)稀释然后用乙酸乙酯(30毫升×3)萃取。合并的有机层用饱和食盐水(20毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯=4∶1至1:1),得到白色固体(S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(2.94克,收率:75%)。LCMS(ESI):m/z 382[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=1:1)。Step 1n: Preparation of (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0120-1): To (S)-(1-(tert-butyloxycarbonyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (0118-1) (3.58 g, 10. 31 mmol, 1.0 eq.), Xphos-Pd-G2 (243 mg, 0.31 mmol, 0.03 eq.), Xphos (147 mg, 0.31 mmol, 0.03 eq.) and sodium carbonate (3.28 g, 30.93 mmol, 3.0 eq.) were added to a mixture of toluene (25 ml), ethanol (25 ml) and water (10 ml). 2-Bromopyrimidine (1.80 g, 11.34 mmol, 1.1 eq.) was added. The mixture was stirred at 55 °C overnight under nitrogen atmosphere. The solvent was removed under reduced pressure. The residue was washed with water (50 The mixture was diluted with ethyl acetate (30 ml × 3) and then extracted with ethyl acetate (30 ml × 3). The combined organic layer was washed with saturated brine (20 ml × 1), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate = 4: 1 to 1: 1) to give a white solid (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (2.94 g, yield: 75%). LCMS (ESI): m/z 382 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 1: 1).

步骤1o:(S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(中间体0122-1)的制备:(S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0120-1)(1.07克,3.08毫摩尔,1.0当量)在氯化氢甲醇溶液(4M溶液,10毫升)的混合物在室温下搅拌3.5小时。减压下除去溶剂。残余物在真空下干燥,得到淡黄色固体(S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(1.09克,粗品)。LCMS(ESI):m/z 282[M+1]+;TLC:Rf 0.3(二氯甲烷:甲醇=10:1)。Step 1o: Preparation of (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Intermediate 0122-1): A mixture of (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0120-1) (1.07 g, 3.08 mmol, 1.0 eq) in methanolic hydrogen chloride solution (4M solution, 10 ml) was stirred at room temperature for 3.5 hours. The solvent was removed under reduced pressure. The residue was dried under vacuum to give (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (1.09 g, crude) as a pale yellow solid. LCMS (ESI): m/z 282 [M+1] + ; TLC: Rf 0.3 (dichloromethane:methanol=10:1).

步骤1p:5-氟-4-碘-2-甲氧基吡啶(化合物0202-1)的制备:在0℃下往2,5-二氟-4-碘吡啶(化合物0201-1)(25.0克,103.74毫摩尔,1.0当量)在甲醇(70毫升)的混合物中加入甲醇钠(5.4M的甲醇溶液,57.6毫升,311.23毫摩尔,3.0当量)。混合物在氮气氛围下加热至60℃并搅拌1.5小时。减压下除去溶剂。残余物用饱和氯化铵溶液(150毫升)稀释然后用乙酸乙酯(70毫升×3)萃取。合并的有机层用饱和食盐水(100毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯=50:1),得到白色固体5-氟-4-碘-2-甲氧基吡啶(22.06克,收率:84%)。LCMS(ESI):m/z 254[M+1]+;TLC:Rf 0.6(石油醚:乙酸乙酯=20:1)。Step 1p: Preparation of 5-fluoro-4-iodo-2-methoxypyridine (Compound 0202-1): To a mixture of 2,5-difluoro-4-iodopyridine (Compound 0201-1) (25.0 g, 103.74 mmol, 1.0 eq.) in methanol (70 ml) was added sodium methoxide (5.4 M in methanol, 57.6 ml, 311.23 mmol, 3.0 eq.) at 0°C. The mixture was heated to 60°C under nitrogen atmosphere and stirred for 1.5 hours. The solvent was removed under reduced pressure. The residue was diluted with saturated ammonium chloride solution (150 ml) and then extracted with ethyl acetate (70 ml x 3). The combined organic layers were washed with saturated brine (100 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate = 50: 1) to give 5-fluoro-4-iodo-2-methoxypyridine (22.06 g, yield: 84%) as a white solid. LCMS (ESI): m/z 254 [M+1] + ; TLC: Rf 0.6 (petroleum ether: ethyl acetate = 20: 1).

步骤1q:5-氟-2-甲氧基-N-(4-甲氧基苄基)-N-甲基吡啶-4-胺(化合物0203-1)的制备:往5-氟-4-碘-2-甲氧基吡啶(0202-1)(1.0克,3.95毫摩尔,1.0当量),三(二亚苄基丙酮)钯(109毫克,0.119毫摩尔,0.03当量),RuPhos(111毫克,0.237毫摩尔,0.06当量)和叔丁醇钠(1.14克,11.85毫摩尔,3.0当量)在甲苯(15毫升)的混合物中加入1-(4-甲氧基苯基)-N-甲基甲胺(0.72克,4.74毫摩尔,1.2当量)。混合物在氮气氛围下加热至120℃并搅拌过夜。减压下除去溶剂。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯=30∶1至10:1),得到淡黄色油状物5-氟-2-甲氧基-N-(4-甲氧基苄基)-N-甲基吡啶-4-胺(630毫克,收率:58%)。LCMS(ESI):m/z 277[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=10:1)。Step 1q: Preparation of 5-fluoro-2-methoxy-N-(4-methoxybenzyl)-N-methylpyridin-4-amine (compound 0203-1): To a mixture of 5-fluoro-4-iodo-2-methoxypyridine (0202-1) (1.0 g, 3.95 mmol, 1.0 eq), tris(dibenzylideneacetone)palladium (109 mg, 0.119 mmol, 0.03 eq), RuPhos (111 mg, 0.237 mmol, 0.06 eq) and sodium tert-butoxide (1.14 g, 11.85 mmol, 3.0 eq) in toluene (15 ml) was added 1-(4-methoxyphenyl)-N-methylmethanamine (0.72 g, 4.74 mmol, 1.2 eq). The mixture was heated to 120° C. under nitrogen atmosphere and stirred overnight. The solvent was removed under reduced pressure. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate = 30: 1 to 10: 1) to give 5-fluoro-2-methoxy-N-(4-methoxybenzyl)-N-methylpyridin-4-amine (630 mg, yield: 58%) as a pale yellow oil. LCMS (ESI): m/z 277 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 10: 1).

步骤1r:5-氟-2-甲氧基-N-甲基吡啶-4-胺(化合物0204-1)的制备:往5-氟-2-甲氧基-N-(4-甲氧基苄基)-N-甲基吡啶-4-胺(0203-1)(630毫克,2.28毫摩尔,1.0当量)在二氯甲烷(12毫升)的混合物中加入三氟乙酸(3毫升)。混合物在室温下搅拌2小时。减压下除去溶剂。残余物用水(30毫升)稀释。加入碳酸氢钠固体调节pH=8,然后水层用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(20毫升×1)洗涤,经无水硫酸钠干燥并浓缩,得到淡黄色油状物5-氟 -2-甲氧基-N-甲基吡啶-4-胺(204毫克,收率:57%)。LCMS(ESI):m/z 157[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=8:1)。Step 1r: Preparation of 5-fluoro-2-methoxy-N-methylpyridin-4-amine (Compound 0204-1): To a mixture of 5-fluoro-2-methoxy-N-(4-methoxybenzyl)-N-methylpyridin-4-amine (0203-1) (630 mg, 2.28 mmol, 1.0 eq.) in dichloromethane (12 ml) was added trifluoroacetic acid (3 ml). The mixture was stirred at room temperature for 2 hours. The solvent was removed under reduced pressure. The residue was diluted with water (30 ml). Solid sodium bicarbonate was added to adjust pH=8, and then the aqueous layer was extracted with ethyl acetate (15 ml×3). The combined organic layers were washed with saturated brine (20 ml×1), dried over anhydrous sodium sulfate and concentrated to give 5-fluoro- -2-Methoxy-N-methylpyridin-4-amine (204 mg, yield: 57%). LCMS (ESI): m/z 157 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=8:1).

步骤1s:(S)-N-(5-氟-2-甲氧基吡啶-4-基)-N,7'-二甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酰胺(化合物1)的制备:在0℃下,往5-氟-2-甲氧基-N-甲基吡啶-4-胺(0204-1)(20毫克,0.13毫摩尔,2.0当量)和三乙胺(53毫克,0.52毫摩尔,8.0当量)在二氯甲烷(2.5毫升)的混合物中加入三光气(19毫克,0.065毫摩尔,1.0当量)。混合物升到室温并搅拌1.5小时。混合物用水(15毫升)稀释然后用乙酸乙酯(10毫升×2)萃取。合并的有机层用饱和食盐水(15毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残留物溶于二氯甲烷(2.5毫升),加入(S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-1)(23毫克,0.065毫摩尔,1.0当量)和N,N-二异丙基乙胺(54毫克,0.46毫摩尔,7.0当量)。混合物在室温下搅拌1小时。减压下除去溶剂。残余物通过制备薄层色谱(二氯甲烷:甲醇:氨水=80:4.5:0.8)纯化,得到白色固体(S)-N-(5-氟-2-甲氧基吡啶-4-基)-N,7'-二甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酰胺(15毫克,收率:50%)。LCMS(ESI):m/z 464[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=10:1)。1H NMR(500MHz,MeOD)δ8.81(d,J=4.9Hz,2H),8.00(d,J=2.8Hz,1H),7.87(s,1H),7.30(t,J=4.9Hz,1H),6.64(d,J=5.5Hz,1H),3.88(s,3H),3.55–3.37(m,2H),3.26(d,J=11.0Hz,2H),3.21(s,3H),2.90–2.65(m,2H),2.58(s,3H),2.01(dtd,J=20.0,13.0,7.3Hz,2H),1.92–1.74(m,2H).Step 1s: Preparation of (S)-N-(5-fluoro-2-methoxypyridin-4-yl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxamide (Compound 1): To a mixture of 5-fluoro-2-methoxy-N-methylpyridin-4-amine (0204-1) (20 mg, 0.13 mmol, 2.0 eq) and triethylamine (53 mg, 0.52 mmol, 8.0 eq) in dichloromethane (2.5 ml) was added triphosgene (19 mg, 0.065 mmol, 1.0 eq) at 0°C. The mixture was warmed to room temperature and stirred for 1.5 hours. The mixture was diluted with water (15 ml) and then extracted with ethyl acetate (10 ml x 2). The combined organic layer was washed with saturated brine (15 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was dissolved in dichloromethane (2.5 ml), and (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-1) (23 mg, 0.065 mmol, 1.0 eq) and N,N-diisopropylethylamine (54 mg, 0.46 mmol, 7.0 eq) were added. The mixture was stirred at room temperature for 1 hour. The solvent was removed under reduced pressure. The residue was purified by preparative thin layer chromatography (dichloromethane:methanol:aqueous ammonia=80:4.5:0.8) to give a white solid (S)-N-(5-fluoro-2-methoxypyridin-4-yl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxamide (15 mg, yield: 50%). LCMS (ESI): m/z 464 [M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=10:1). 1H NMR (500MHz, MeOD) δ8.81(d,J=4.9Hz,2H),8.00(d,J=2.8Hz,1H),7.87(s,1H),7.30(t,J=4.9Hz,1H),6.64(d,J=5.5Hz,1H),3.88(s,3H),3.5 5–3.37(m,2H),3.26(d,J=11.0Hz,2H),3.21(s,3H),2.90–2.65(m,2H) ,2.58(s,3H),2.01(dtd,J=20.0,13.0,7.3Hz,2H),1.92–1.74(m,2H).

实施例2:(S)-N-(5-氟-2-甲氧基吡啶-4-基)-N,7'-二甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-硫代甲酰胺(化合物2)的制备(按照方案三线路制备)Example 2: Preparation of (S)-N-(5-fluoro-2-methoxypyridin-4-yl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-thiocarboxamide (Compound 2) (Prepared according to Scheme 3)

在0℃下往5-氟-2-甲氧基-N-甲基吡啶-4-胺(0204-1)(51.5毫克,0.33毫摩尔,1.0当量)在四氢呋喃(1毫升)的混合物中加入双三甲基硅基胺基锂(1M的四氢呋喃溶液,0.99毫升,0.99毫摩尔,3.0当量)。混合物搅拌10分钟。加入硫光气(114毫克,0.99毫摩尔,3.0当量)。混合物升到室温并搅拌1小时。加入水(20毫升)淬灭反应。水层用二氯甲烷(10毫升×3)萃取。合并的有机层用饱和食盐水(10毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(石油醚∶乙酸乙酯=10∶1)纯化,得到淡黄色油状物(5-氟-2-甲氧基吡啶-4-基)(甲基)氨基硫代酰氯(33毫克,收率:43%)。LCMS(ESI):m/z 235[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=10:1)。往(5-氟-2-甲氧基吡啶-4-基)(甲基)氨基硫代酰氯(33毫克,0.14毫摩尔,1.0当量)和N,N-二异丙基乙胺(90毫克,0.70毫摩尔,5.0当量)在二氯甲烷(5毫升)的混合物中加入(S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-1)(50毫克,0.14毫摩尔,1.0当量)。混合物在室温下搅拌过夜。减压下除去溶剂。残余物通过制备薄层色谱(乙酸乙酯∶二氯甲烷:甲醇=20:20:1)纯化,得到白色固体(S)-N-(5-氟-2-甲氧 基吡啶-4-基)-N,7'-二甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-硫代甲酰胺(59毫克,收率:88%)。LCMS(ESI):m/z 480[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=15:1)。1H NMR(500MHz,MeOD)δ8.80(d,J=4.9Hz,2H),7.99(d,J=3.1Hz,1H),7.85(s,1H),7.30(t,J=4.9Hz,1H),6.50(d,J=5.6Hz,1H),3.88(s,3H),3.80–3.44(m,4H),3.36(d,J=11.3Hz,3H),2.92–2.69(m,2H),2.57(s,3H),2.19–2.00(m,2H),1.93–1.72(m,2H).To a mixture of 5-fluoro-2-methoxy-N-methylpyridin-4-amine (0204-1) (51.5 mg, 0.33 mmol, 1.0 eq.) in tetrahydrofuran (1 ml) was added lithium bis(trimethylsilyl)amide (1M in tetrahydrofuran, 0.99 ml, 0.99 mmol, 3.0 eq.) at 0°C. The mixture was stirred for 10 minutes. Thiophosgene (114 mg, 0.99 mmol, 3.0 eq.) was added. The mixture was warmed to room temperature and stirred for 1 hour. Water (20 ml) was added to quench the reaction. The aqueous layer was extracted with dichloromethane (10 ml x 3). The combined organic layers were washed with saturated brine (10 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (petroleum ether: ethyl acetate = 10: 1) to give a pale yellow oily substance (5-fluoro-2-methoxypyridin-4-yl)(methyl)aminothioyl chloride (33 mg, yield: 43%). LCMS (ESI): m/z 235 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 10: 1). To a mixture of (5-fluoro-2-methoxypyridin-4-yl)(methyl)aminothioyl chloride (33 mg, 0.14 mmol, 1.0 eq) and N,N-diisopropylethylamine (90 mg, 0.70 mmol, 5.0 eq) in dichloromethane (5 ml) was added (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-1) (50 mg, 0.14 mmol, 1.0 eq). The mixture was stirred at room temperature overnight. The solvent was removed under reduced pressure. The residue was purified by preparative thin layer chromatography (ethyl acetate: dichloromethane: methanol = 20:20:1) to give (S)-N-(5-fluoro-2-methoxypyridin-4-yl)(methyl)aminothioyl chloride (33 mg, 0.14 mmol, 1.0 eq) as a white solid. 4-(2-(4-pyridin-4-yl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carbothioamide (59 mg, yield: 88%). LCMS (ESI): m/z 480 [M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=15:1). 1H NMR(500MHz,MeOD)δ8.80(d,J=4.9Hz,2H),7.99(d,J=3.1Hz,1H),7.85(s,1H),7.30(t,J=4.9Hz,1H),6.50(d,J=5.6Hz,1H),3 .88(s,3H),3.80–3.44(m,4H),3.36(d,J=11.3Hz,3H),2.92–2.69(m,2H),2.57(s,3H),2.19–2.00(m,2H),1.93–1.72(m,2H).

实施例3:(S)-N-(3,4-二氟苯基)-N,7'-二甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酰胺(化合物3)的制备(按照方案二线路制备)Example 3: Preparation of (S)-N-(3,4-difluorophenyl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxamide (Compound 3) (Prepared according to Scheme 2)

步骤3a:3,4-二氟-N-甲基苯胺(化合物0204-3)的制备:
Step 3a: Preparation of 3,4-difluoro-N-methylaniline (Compound 0204-3):

氮气保护下,向3,4-二氟苯胺(1.0克,7.75毫摩尔,1.0当量),多聚甲醛(2.1克,23.25毫摩尔,3.0当量)的甲醇(10毫升)溶液中加入甲醇钠(5.4摩尔/升,7.17毫升,38.75毫摩尔,5.0当量),混合物在室温下搅拌过夜。反应液中加入硼氢化钠(879毫克,23.25毫摩尔,3.0当量),并于65℃搅拌2.0小时。混合物经过减压浓缩,残留物用硅胶柱(石油醚/乙酸乙酯=10/1至6/1)纯化,得到黄色油状产物3,4-二氟-N-甲基苯胺(600毫克,收率:54.15%)。LCMS(ESI):m/z=144[M+1]+.Under nitrogen protection, sodium methoxide (5.4 mol/L, 7.17 ml, 38.75 mmol, 5.0 eq.) was added to a solution of 3,4-difluoroaniline (1.0 g, 7.75 mmol, 1.0 eq.) and paraformaldehyde (2.1 g, 23.25 mmol, 3.0 eq.) in methanol (10 ml), and the mixture was stirred at room temperature overnight. Sodium borohydride (879 mg, 23.25 mmol, 3.0 eq.) was added to the reaction solution and stirred at 65°C for 2.0 hours. The mixture was concentrated under reduced pressure, and the residue was purified by silica gel column (petroleum ether/ethyl acetate = 10/1 to 6/1) to obtain a yellow oily product 3,4-difluoro-N-methylaniline (600 mg, yield: 54.15%). LCMS (ESI): m/z = 144 [M+1] + .

步骤3b:(S)-N-(3,4-二氟苯基)-N,7'-二甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酰胺(化合物3)的制备:氮气保护下,向3,4-二氟-N-甲基苯胺(0204-3)(20毫克,0.14毫摩尔,1.0当量),三乙胺(56毫克,0.56毫摩尔,4.0当量)的二氯甲烷(5毫升)溶液中加入三光气(37.4毫克,0.126毫摩尔,0.9当量),混合物在室温下搅拌1.5小时。反应液加水稀释并用二氯甲烷(8毫升)萃取,将有机相干燥,向上述溶液中加入7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-1)(49.4毫克,0.14毫摩尔,1.0当量),N,N-二异丙基乙胺(63.21毫克,0.49毫摩尔,3.5当量),混合物在室温下搅拌0.5小时。反应液加水稀释并用二氯甲烷萃取,将有机相干燥并减压浓缩。残留物用制备薄层色谱(二氯甲烷/甲醇=15/1,4%氨水)纯化,得到黄色固体产物(S)-N-(3,4-二氟苯基)-N,7'-二甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酰胺(30毫克,收率:62.94%)。LCMS(ESI):m/z=451[M+1]+.1H NMR(500MHz,MeOD)δ8.80(d,J=4.9Hz,2H),7.81(s,1H),7.33–7.25(m,2H),7.20(ddd,J=11.4,7.0,2.5Hz,1H),7.01(d,J=8.8Hz,1H),3.41–3.32(m,2H),3.23–3.10(m,5H),2.88–2.63(m,2H),2.56(s,3H),2.00–1.71(m,4H).Step 3b: Preparation of (S)-N-(3,4-difluorophenyl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxamide (Compound 3): Under nitrogen protection, triphosgene (37.4 mg, 0.126 mmol, 0.9 equiv) was added to a solution of 3,4-difluoro-N-methylaniline (0204-3) (20 mg, 0.14 mmol, 1.0 equiv) and triethylamine (56 mg, 0.56 mmol, 4.0 equiv) in dichloromethane (5 ml), and the mixture was stirred at room temperature for 1.5 hours. The reaction solution was diluted with water and extracted with dichloromethane (8 ml), the organic phase was dried, 7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-1) (49.4 mg, 0.14 mmol, 1.0 eq.), N,N-diisopropylethylamine (63.21 mg, 0.49 mmol, 3.5 eq.) were added to the above solution, and the mixture was stirred at room temperature for 0.5 hours. The reaction solution was diluted with water and extracted with dichloromethane, the organic phase was dried and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography (dichloromethane/methanol=15/1, 4% aqueous ammonia) to give a yellow solid product (S)-N-(3,4-difluorophenyl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxamide (30 mg, yield: 62.94%). LCMS (ESI): m/z=451[M+1] + . 1 H NMR(500MHz,MeOD)δ8.80(d,J=4.9Hz,2H),7.81(s,1H),7.33–7.25(m,2H),7.20(ddd,J=11.4,7.0,2.5Hz,1H), 7.01(d,J=8.8Hz,1H),3.41–3.32(m,2H),3.23–3.10(m,5H),2.88–2.63(m,2H),2.56(s,3H),2.00–1.71(m,4H).

实施例4:(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-硫酮(化合物7)的制备(按照方案四和五线路制备) Example 4: Preparation of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (Compound 7) (Prepared according to Schemes 4 and 5)

步骤4a:2-(5-氟-2-甲氧基吡啶-4-基)丙二酸二苄酯(化合物0403-7)的制备:往5-氟-4-碘-2-甲氧基吡啶(0202-1)(22.06克,87.19毫摩尔,1.0当量),碘化亚铜(1.66克,8.72毫摩尔,0.1当量),2-吡啶甲酸(2.15克,17.44毫摩尔,0.2当量)和碳酸铯(85.27克,261.57毫摩尔,3.0当量)在四氢呋喃(130毫升)的混合物中加入丙二酸二苄酯(34.70克,12.21毫摩尔,1.4当量)。混合物在氮气氛围下加热至70℃并搅拌过夜。混合物用水(200毫升)稀释然后用乙酸乙酯(100毫升×3)萃取。合并的有机层用饱和食盐水(100毫升×1)洗涤,经无水硫酸钠干燥并浓缩,得到棕色油状物2-(5-氟-2-甲氧基吡啶-4-基)丙二酸二苄酯(46.1克,粗品)。LCMS(ESI):m/z 410[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=10:1)。Step 4a: Preparation of dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)malonate (compound 0403-7): To a mixture of 5-fluoro-4-iodo-2-methoxypyridine (0202-1) (22.06 g, 87.19 mmol, 1.0 eq), cuprous iodide (1.66 g, 8.72 mmol, 0.1 eq), 2-picolinic acid (2.15 g, 17.44 mmol, 0.2 eq) and cesium carbonate (85.27 g, 261.57 mmol, 3.0 eq) in tetrahydrofuran (130 ml) was added dibenzyl malonate (34.70 g, 12.21 mmol, 1.4 eq). The mixture was heated to 70° C. under nitrogen atmosphere and stirred overnight. The mixture was diluted with water (200 ml) and then extracted with ethyl acetate (100 ml×3). The combined organic layer was washed with saturated brine (100 ml×1), dried over anhydrous sodium sulfate and concentrated to give dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)malonate (46.1 g, crude product) as a brown oil. LCMS (ESI): m/z 410 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=10:1).

步骤4b:2-(5-氟-2-甲氧基吡啶-4-基)-2-甲基丙二酸二苄酯(化合物0404-7)的制备:往2-(5-氟-2-甲氧基吡啶-4-基)丙二酸二苄酯(0403-7)(35.70克,87.19毫摩尔,1.0当量)和碳酸钾(36.10克,261.57毫摩尔,3.0当量)在乙腈(80毫升)的混合物中加入碘甲烷(24.75克,174.38毫摩尔,2.0当量)。混合物在室温下搅拌过夜。减压下除去溶剂。残余物用水(150毫升)稀释然后用乙酸乙酯(100毫升×2)萃取。合并的有机层用饱和食盐水(50毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物用石油醚(120毫升)稀释,然后在0℃下搅拌1小时。混合物过滤。固体用石油醚洗涤然后在真空下干燥,得到淡黄色固体2-(5-氟-2-甲氧基吡啶-4-基)-2-甲基丙二酸二苄酯(24.5克,收率:68%)。LCMS(ESI):m/z 414[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=10:1)。Step 4b: Preparation of dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)-2-methylmalonate (Compound 0404-7): To a mixture of dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)malonate (0403-7) (35.70 g, 87.19 mmol, 1.0 eq) and potassium carbonate (36.10 g, 261.57 mmol, 3.0 eq) in acetonitrile (80 ml) was added iodomethane (24.75 g, 174.38 mmol, 2.0 eq). The mixture was stirred at room temperature overnight. The solvent was removed under reduced pressure. The residue was diluted with water (150 ml) and then extracted with ethyl acetate (100 ml x 2). The combined organic layers were washed with saturated brine (50 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was diluted with petroleum ether (120 ml) and then stirred at 0°C for 1 hour. The mixture was filtered. The solid was washed with petroleum ether and then dried under vacuum to give a pale yellow solid 2-(5-fluoro-2-methoxypyridin-4-yl)-2-methylmalonic acid dibenzyl ester (24.5 g, yield: 68%). LCMS (ESI): m/z 414 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 10: 1).

步骤4c:2-(5-氟-2-甲氧基吡啶-4-基)丙酸(化合物0405-7)的制备:往2-(5-氟-2-甲氧基吡啶-4-基)-2-甲基丙二酸二苄酯(0404-7)(24.5克,59.32毫摩尔,1.0当量)在乙酸乙酯(100毫升)的混合物中加入钯/碳(10%,4.0克)。混合物在氢气球压力下55℃搅拌两天。混合物过滤。滤液减压浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯=6∶1至3:1),得到白色固体2-(5-氟-2-甲氧基吡啶-4-基)丙酸(8.86克,收率:75%)。LCMS(ESI):m/z 200[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=2:1)。Step 4c: Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (Compound 0405-7): To a mixture of 2-(5-fluoro-2-methoxypyridin-4-yl)-2-methylmalonic acid dibenzyl ester (0404-7) (24.5 g, 59.32 mmol, 1.0 eq.) in ethyl acetate (100 mL) was added palladium/carbon (10%, 4.0 g). The mixture was stirred at 55° C. under hydrogen balloon pressure for two days. The mixture was filtered. The filtrate was concentrated under reduced pressure. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate = 6: 1 to 3: 1) to give 2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (8.86 g, yield: 75%) as a white solid. LCMS (ESI): m/z 200 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 2: 1).

步骤4d:(R)-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(化合物0406-7)和(S)-2-(-5-氟-2-甲基吡啶-4-基丙酸(0407-7)的制备:2-(5-氟-2-甲氧基吡啶-4-基)丙酸(0405-7)(8.86克,44.52毫摩尔)对映体的分离通过超临界流体色谱法进行(色谱柱:ChiralPak IG,250×30mm I.D.,10μm;流动相:A为二氧化碳,B为甲醇;洗脱剂:10%B;流速:60毫升/分钟;背压:100巴;柱温:38℃)。第一个洗脱的对映体是(R)-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(0406-7)(3.75克,产率:42%),第二个洗脱的对映体是(S)-2-(-5-氟-2-甲基吡啶-4-基丙酸(0407-7)(3.86克,产率:44%)。分析条件(色谱柱:ChiralPak IG,100×4.6mm I.D.,5μm;流动相:A为二氧化碳,B为甲醇(0.05%二乙胺);洗脱剂:5%B;流速:2.5毫升/分钟;背压:100巴;柱温:35℃)Step 4d: Preparation of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (compound 0406-7) and (S)-2-(-5-fluoro-2-methylpyridin-4-yl)propanoic acid (0407-7): Separation of the enantiomers of 2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0405-7) (8.86 g, 44.52 mmol) was carried out by supercritical fluid chromatography (column: ChiralPak IG, 250×30 mm I.D., 10 μm; mobile phase: A is carbon dioxide, B is methanol; eluent: 10% B; flow rate: 60 ml/min; back pressure: 100 bar; column Temperature: 38°C). The first eluting enantiomer was (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0406-7) (3.75 g, yield: 42%), and the second eluting enantiomer was (S)-2-(-5-fluoro-2-methylpyridin-4-yl)propanoic acid (0407-7) (3.86 g, yield: 44%). Analysis conditions (chromatographic column: ChiralPak IG, 100×4.6 mm I.D., 5 μm; mobile phase: A is carbon dioxide, B is methanol (0.05% diethylamine); eluent: 5% B; flow rate: 2.5 ml/min; back pressure: 100 bar; column temperature: 35°C)

步骤4e:(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺 [吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(化合物0413-7)的制备:往(R)-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(化合物0406-7)(66毫克,0.33毫摩尔,1.0当量)在乙腈(3毫升)的混合物中加入咪唑三氟甲磺酸盐(108毫克,0.50毫摩尔,1.5当量)和羰基二咪唑(64毫克,0.40毫摩尔,1.2当量)。混合物在室温下搅拌2小时。加入(S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(化合物0122-1)(117毫克,0.33毫摩尔,1.0当量)和N,N-二异丙基乙胺(213毫克,1.65毫摩尔,5.0当量)。混合物在室温下搅拌3小时。混合物用水(20毫升)稀释然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(15毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(二氯甲烷:甲醇:氨水=80:4:0.8)纯化,得到白色固体(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(80毫克,收率:52%)。LCMS(ESI):m/z 463[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=10:1)。Step 4e: (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro Preparation of [pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (compound 0413-7): To a mixture of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (compound 0406-7) (66 mg, 0.33 mmol, 1.0 eq) in acetonitrile (3 ml) were added imidazole trifluoromethanesulfonate (108 mg, 0.50 mmol, 1.5 eq) and carbonyldiimidazole (64 mg, 0.40 mmol, 1.2 eq). The mixture was stirred at room temperature for 2 hours. (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0122-1) (117 mg, 0.33 mmol, 1.0 eq) and N,N-diisopropylethylamine (213 mg, 1.65 mmol, 5.0 eq) were added. The mixture was stirred at room temperature for 3 hours. The mixture was diluted with water (20 ml) and then extracted with ethyl acetate (15 ml x 3). The combined organic layer was washed with saturated brine (15 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (dichloromethane:methanol:aqueous ammonia=80:4:0.8) to give a white solid (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (80 mg, yield: 52%). LCMS (ESI): m/z 463 [M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=10:1).

步骤4f:(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-硫酮(化合物7)的制备:往(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(化合物0413-7)(80毫克,0.18毫摩尔,1.0当量)在甲苯(5毫升)的混合物中加入劳森试剂(85毫克,0.21毫摩尔,1.2当量)。混合物在微波反应器中在125℃加热40分钟。混合物用水(20毫升)稀释然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(15毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(二氯甲烷:甲醇:氨水=80:3.5:0.8)纯化,得到白色固体(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-硫酮(30毫克,收率:36%)。LCMS(ESI):m/z 479[M+1]+;TLC:Rf 0.6(二氯甲烷:甲醇=10:1)。1HNMR(500MHz,MeOD)δ8.81(t,J=4.4Hz,2H),7.88(dt,J=41.5,18.1Hz,2H),7.30(t,J=4.1Hz,1H),6.95–6.81(m,1H),4.54(dt,J=14.2,6.8Hz,1H),4.18–3.66(m,7H),2.97–2.76(m,2H),2.58(dd,J=9.6,4.7Hz,3H),2.32–2.06(m,2H),2.05–1.78(m,2H),1.63–1.51(m,3H).Step 4f: Preparation of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (Compound 7): To a mixture of (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (compound 0413-7) (80 mg, 0.18 mmol, 1.0 eq.) in toluene (5 ml) was added Lawesson's reagent (85 mg, 0.21 mmol, 1.2 eq.). The mixture was heated at 125°C in a microwave reactor for 40 minutes. The mixture was diluted with water (20 ml) and then extracted with ethyl acetate (15 ml x 3). The combined organic layers were washed with saturated brine (15 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (dichloromethane:methanol:aqueous ammonia=80:3.5:0.8) to give (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (30 mg, yield: 36%) as a white solid. LCMS (ESI): m/z 479 [M+1] + ; TLC: Rf 0.6 (dichloromethane:methanol=10:1). 1 HNMR(500MHz,MeOD)δ8.81(t,J=4.4Hz,2H),7.88(dt,J=41.5,18.1Hz,2H),7.30(t,J=4.1Hz,1H),6.95–6.81(m,1H),4.54(dt,J=14.2, 6.8Hz,1H),4.18–3.66(m,7H),2.97–2.76(m,2H),2.58(dd,J=9.6,4.7Hz,3H),2.32–2.06(m,2H),2.05–1.78(m,2H),1.63–1.51(m,3H).

实施例5:2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮-3,3,3-d3(化合物57)的制备(按照方案一和四线路制备)Example 5: Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one-3,3,3-d3 (Compound 57) (Prepared according to Schemes 1 and 4)

步骤5a:2-(5-氟-2-甲氧基吡啶-4-基)-2-(甲基-d3)丙二酸二苄基酯(化合物0404-57)的制备:氮气保护下,向2-(5-氟-2-甲氧基吡啶-4-基)丙二酸二苄基酯(0403-7)(500毫克,1.22毫摩尔,1.0当量),碳酸钾(507毫克,3.67毫摩尔,3.0当量)的N,N-二甲基甲酰胺(5毫升)混合溶液中加入氘代碘甲烷(355毫克,2.44毫摩尔,2.0当量),混合物在室温下搅拌三个小时。反应液用水稀释并用乙酸乙酯萃取。将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙 酯=3/1)纯化,得到白色油状产物2-(5-氟-2-甲氧基吡啶-4-基)-2-(甲基-d3)丙二酸二苄基酯(510毫克,收率:98.07%)。LCMS(ESI):m/z=427[M+1]+.Step 5a: Preparation of dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)-2-(methyl-d3)malonate (Compound 0404-57): Under nitrogen protection, to a mixed solution of dibenzyl 2-(5-fluoro-2-methoxypyridin-4-yl)malonate (0403-7) (500 mg, 1.22 mmol, 1.0 eq.), potassium carbonate (507 mg, 3.67 mmol, 3.0 eq.) in N,N-dimethylformamide (5 ml) was added deuterated iodomethane (355 mg, 2.44 mmol, 2.0 eq.), and the mixture was stirred at room temperature for three hours. The reaction solution was diluted with water and extracted with ethyl acetate. The organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate) Ester = 3/1) to obtain a white oily product, 2-(5-fluoro-2-methoxypyridin-4-yl)-2-(methyl-d3)malonic acid dibenzyl ester (510 mg, yield: 98.07%). LCMS (ESI): m/z = 427 [M+1] + .

步骤5b:2-(5-氟-2-甲氧基吡啶-4-基)丙酸-3,3,3-d3酸(化合物0405-57)的制备:氢气保护下,往2-(5-氟-2-甲氧基吡啶-4-基)-2-(甲基-d3)丙二酸二苄基酯(化合物0404-57)(510毫克,1.19毫摩尔,1.0当量)的乙酸乙酯(5毫升)混合物中加入钯碳(102毫克,20%质量分数)。混合物在55℃下搅拌过夜。过滤混合物并将滤液降压浓缩,残留物用硅胶柱(石油醚/乙酸乙酯=2/1)纯化,得到白色油状产物2-(5-氟-2-甲氧基吡啶-4-基)丙酸-3,3,3-d3酸(120毫克,收率:49.38%)。LCMS(ESI):m/z=203[M+1]+.Step 5b: Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)propionic acid-3,3,3-d3 acid (Compound 0405-57): Under hydrogen protection, palladium carbon (102 mg, 20% mass fraction) was added to a mixture of 2-(5-fluoro-2-methoxypyridin-4-yl)-2-(methyl-d3)malonate (Compound 0404-57) (510 mg, 1.19 mmol, 1.0 equivalent) in ethyl acetate (5 ml). The mixture was stirred at 55°C overnight. The mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate = 2/1) to obtain a white oily product 2-(5-fluoro-2-methoxypyridin-4-yl)propionic acid-3,3,3-d3 acid (120 mg, yield: 49.38%). LCMS (ESI): m/z=203[M+1] + .

步骤5c:2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮-3,3,3-d3(化合物57(0412-57))的制备:2-(5-氟-2-甲氧基吡啶-4-基)丙酸-3,3,3-d3酸(0405-57)(29毫克,0.143毫摩尔,1.0当量),7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-1)(50毫克,0.143毫摩尔,1.0当量),N,N-二异丙基乙胺(74毫克,0.571毫摩尔,4.0当量)的二氯甲烷(3毫升)溶液在室温下搅拌10分钟。然后混合物加入2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(66毫克,0.171毫摩尔,1.2当量)并在室温下搅拌一小时。反应液加水稀释并用二氯甲烷萃取,将有机相干燥并减压浓缩。残留物用制备薄层色谱(二氯甲烷/甲醇/氨水=80/4.5/0.8)纯化,得到黄色固体产物2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮-3,3,3-d3(40毫克,收率:60.61%)。LCMS(ESI):m/z=466[M+1]+.1HNMR(500MHz,MeOD)δ8.87–8.74(m,2H),8.02–7.80(m,2H),7.31(s,1H),6.82–6.67(m,1H),4.21(dd,J=42.9,6.0Hz,1H),3.93–3.82(m,3H),3.76–3.33(m,4H),2.98–2.75(m,2H),2.59(dd,J=12.9,4.1Hz,3H),2.21–1.76(m,4H).Step 5c: Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7′-methyl-6′-(pyrimidin-2-yl)-3′,4′-dihydro-1′H-spiro[pyrrolidine-3,2′-[1,8]naphthyridine]-1-yl)propan-1-one-3,3,3-d3 (Compound 57 (0412-57)): 2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid-3,3,3-d3. A solution of 7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-1) (50 mg, 0.143 mmol, 1.0 eq.) and N,N-diisopropylethylamine (74 mg, 0.571 mmol, 4.0 eq.) in dichloromethane (3 ml) was stirred at room temperature for 10 minutes. Then, 2-(7-azabenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (66 mg, 0.171 mmol, 1.2 eq.) was added to the mixture and stirred at room temperature for one hour. The reaction solution was diluted with water and extracted with dichloromethane, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography (dichloromethane/methanol/aqueous ammonia = 80/4.5/0.8) to give a yellow solid product 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one-3,3,3-d3 (40 mg, yield: 60.61%). LCMS (ESI): m/z=466[M+1] + . 1 HNMR(500MHz,MeOD)δ8.87–8.74(m,2H),8.02–7.80(m,2H),7.31(s,1H),6.82–6.67(m,1H),4.21(dd,J=42.9,6.0 Hz,1H),3.93–3.82(m,3H),3.76–3.33(m,4H),2.98–2.75(m,2H),2.59(dd,J=12.9,4.1Hz,3H),2.21–1.76(m,4H).

实施例6:2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-硫酮-3,3,3-d3(化合物58)的制备(按照方案五线路制备)Example 6: Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione-3,3,3-d3 (Compound 58) (Prepared according to Scheme 5)

2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮-3,3,3-d3(0412-57)(50毫克,0.107毫摩尔,1.0当量),劳森试剂(53毫克,0.129毫摩尔,1.2当量)的甲苯(2.5毫升)混合溶液在125℃下在微波反应器中搅拌一个小时。反应液用水稀释并用二氯甲烷萃取。将有机相干燥并减压浓缩。残留物用制备薄层色谱(二氯甲烷/甲醇/氨水=80/4.5/0.8)纯化,得到黄色固体产物2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-硫酮-3,3,3-d3(33毫克,收率:63.46%)。LCMS(ESI):m/z=484[M+1]+.1HNMR(500MHz,MeOD)δ8.82(t,J=4.2Hz,2H),7.99–7.76(m,2H),7.31(t,J=4.8Hz,1H),6.96–6.83(m,1H),4.52 (dd,J=48.3,6.5Hz,1H),4.22–3.90(m,3H),3.86(dd,J=15.1,5.9Hz,3H),3.82–3.62(m,1H),2.97–2.77(m,2H),2.59(dd,J=9.7,7.2Hz,3H),2.34–2.09(m,2H),2.07–1.77(m,2H).A mixed solution of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one-3,3,3-d3(0412-57) (50 mg, 0.107 mmol, 1.0 eq.), Lawesson's reagent (53 mg, 0.129 mmol, 1.2 eq.) in toluene (2.5 ml) was stirred at 125°C in a microwave reactor for one hour. The reaction solution was diluted with water and extracted with dichloromethane. The organic phase was dried and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography (dichloromethane/methanol/aqueous ammonia = 80/4.5/0.8) to give a yellow solid product 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione-3,3,3-d3 (33 mg, yield: 63.46%). LCMS (ESI): m/z=484[M+1] + . 1 HNMR (500MHz, MeOD) δ8.82(t,J=4.2Hz,2H),7.99–7.76(m,2H),7.31(t,J=4.8Hz,1H),6.96–6.83(m,1H),4.52 (dd,J=48.3,6.5Hz,1H),4.22–3.90(m,3H),3.86(dd,J=15.1,5.9Hz,3H),3.82–3.62(m,1 H),2.97–2.77(m,2H),2.59(dd,J=9.7,7.2Hz,3H),2.34–2.09(m,2H),2.07–1.77(m,2H).

实施例7:(2R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-(6’-(嘧啶-2-基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(化合物34)的制备(按照方案一和四线路制备)Example 7: Preparation of (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 34) (Prepared according to Schemes 1 and 4)

步骤7a:3-羟基-3-((三甲基硅基)乙炔基)吡咯烷-1-甲酸叔丁酯(化合物0102-34)的制备:氮气保护下,在-70℃下,向乙炔基三甲基硅烷(2.55克,25.9毫摩尔,1.2当量)的四氢呋喃(40毫升)溶液中滴加正丁基锂(9.5毫升,23.8毫摩尔,1.1当量),混合物在-70℃下搅拌1.0小时。然后在-60℃下向反应液中滴加1-叔丁氧碳基-3-吡咯烷酮(0101-34)(4.0克,21.6毫摩尔,1.0当量)的四氢呋喃(10毫升)溶液,撤去低温装置,并使混合物在室温下搅拌过夜。反应液用氯化铵溶液淬灭并用乙酸乙酯萃取,将有机相干燥并减压浓缩得到黄色油状粗产物3-羟基-3-((三甲基硅基)乙炔基)吡咯烷-1-甲酸叔丁酯(6.04克),粗产物不经过进一步纯化,直接用于下一步。Step 7a: Preparation of tert-butyl 3-hydroxy-3-((trimethylsilyl)ethynyl)pyrrolidine-1-carboxylate (Compound 0102-34): Under nitrogen protection, n-butyl lithium (9.5 ml, 23.8 mmol, 1.1 eq) was added dropwise to a solution of ethynyltrimethylsilane (2.55 g, 25.9 mmol, 1.2 eq) in tetrahydrofuran (40 ml) at -70°C, and the mixture was stirred at -70°C for 1.0 hour. Then, a solution of 1-tert-butyloxycarbonyl-3-pyrrolidone (0101-34) (4.0 g, 21.6 mmol, 1.0 eq) in tetrahydrofuran (10 ml) was added dropwise to the reaction solution at -60°C, the cryogenic device was removed, and the mixture was stirred at room temperature overnight. The reaction solution was quenched with ammonium chloride solution and extracted with ethyl acetate. The organic phase was dried and concentrated under reduced pressure to give a yellow oily crude product of tert-butyl 3-hydroxy-3-((trimethylsilyl)ethynyl)pyrrolidine-1-carboxylate (6.04 g). The crude product was used directly in the next step without further purification.

步骤7b:3-乙炔基-3-羟基吡咯烷-1-甲酸叔丁酯(化合物0103-34)的制备:氮气保护下,向3-羟基-3-((三甲基硅基)乙炔基)吡咯烷-1-甲酸叔丁酯(0102-34)(6.00克,21.2毫摩尔,1.0当量)的甲醇(100毫升)溶液中加入碳酸钾(5.80克,42.3毫摩尔,2.0当量),将混合物搅拌在室温下搅拌1.5小时。减压除去溶剂,残留物加水稀释并用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=10/1)纯化,得到黄色油状产物3-乙炔基-3-羟基吡咯烷-1-甲酸叔丁酯(4.43克,两步收率:97.1%)。Step 7b: Preparation of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (Compound 0103-34): Under nitrogen protection, potassium carbonate (5.80 g, 42.3 mmol, 2.0 eq.) was added to a solution of tert-butyl 3-hydroxy-3-((trimethylsilyl)ethynyl)pyrrolidine-1-carboxylate (0102-34) (6.00 g, 21.2 mmol, 1.0 eq.) in methanol (100 ml), and the mixture was stirred at room temperature for 1.5 hours. The solvent was removed under reduced pressure, the residue was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate=10/1) to obtain a yellow oily product, tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (4.43 g, two-step yield: 97.1%).

步骤7c:3-乙酰氧基-3-乙炔基-吡咯烷-1-甲酸叔丁酯(化合物0104-34)的制备:氮气保护下,0℃下,向3-乙炔基-3-羟基吡咯烷-1-甲酸叔丁酯(0103-34)(4.43克,21.0毫摩尔,1.0当量),N,N-二异丙基乙胺(4.06克,31.5毫摩尔,1.5当量)的二氯甲烷(100毫升)溶液中加入乙酰氯(1.98克,25.2毫摩尔,1.2当量),混合物在0℃下搅拌1小时。反应液加水淬灭,并用二氯甲烷萃取。将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=20/1至5/1)纯化,得到黄色油状产物3-乙酰氧基-3-乙炔基-吡咯烷-1-甲酸叔丁酯(3.07克,收率:57.8%)。Step 7c: Preparation of tert-butyl 3-acetoxy-3-ethynyl-pyrrolidine-1-carboxylate (Compound 0104-34): Under nitrogen protection, acetyl chloride (1.98 g, 25.2 mmol, 1.2 eq) was added to a solution of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (0103-34) (4.43 g, 21.0 mmol, 1.0 eq) and N,N-diisopropylethylamine (4.06 g, 31.5 mmol, 1.5 eq) in dichloromethane (100 ml) at 0°C, and the mixture was stirred at 0°C for 1 hour. The reaction solution was quenched with water and extracted with dichloromethane. The organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate=20/1 to 5/1) to give yellow oily product 3-acetoxy-3-ethynyl-pyrrolidine-1-carboxylic acid tert-butyl ester (3.07 g, yield: 57.8%).

步骤7d:3-乙炔基-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(化合物0105-34)的制备:氮气保护下,70℃下,向4-甲氧基苄胺(3.07克,22.3毫摩尔,2.0当量),氯化亚铜(111毫克,1.1毫摩尔,0.1当量)的四氢呋喃(30毫升)溶液中加入3-乙酰氧基-3-乙炔基-吡咯烷-1-甲酸叔丁酯(0104-34)(2.83克,11.2毫摩尔,1.0当量)的四氢呋喃(10毫升)溶液,混合物在70℃下搅拌1小时。反应液加水稀释并用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=10/1至4/1)纯化,得到黄色油状产物3-乙炔基 -3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(2.56克,收率:69.4%)。Step 7d: Preparation of tert-butyl 3-ethynyl-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0105-34): Under nitrogen protection, to a solution of 4-methoxybenzylamine (3.07 g, 22.3 mmol, 2.0 equiv.) and cuprous chloride (111 mg, 1.1 mmol, 0.1 equiv.) in tetrahydrofuran (30 ml) at 70°C, a solution of tert-butyl 3-acetoxy-3-ethynyl-pyrrolidine-1-carboxylate (0104-34) (2.83 g, 11.2 mmol, 1.0 equiv.) in tetrahydrofuran (10 ml) was added, and the mixture was stirred at 70°C for 1 hour. The reaction solution was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate = 10/1 to 4/1) to obtain a yellow oily product 3-ethynyl -tert-Butyl 3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (2.56 g, yield: 69.4%).

步骤7e:2-氯-3-碘-6-(三氟甲基)吡啶(化合物0107-34)的制备:在0℃下往2-氯-6-(三氟甲基)吡啶-3-胺(0106-34)(1.5克,7.63毫摩尔,1.0当量)和6M稀盐酸(7.6毫升,45.78毫摩尔,6.0当量)在乙腈(5毫升)的混合物中逐滴加入亚硝酸钠(0.63克,9.16毫摩尔,1.2当量)的水(503毫升)溶液。混合物在0℃下搅拌35分钟。加入碘化钾(2.53克,15.26毫摩尔,2.0当量)的水(5毫升)溶液。混合物升到室温然后搅拌过夜。缓慢加入2M氢氧化钠溶液调节pH=8,然后水层用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(15毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯=100:1),得到白色固体2-氯-3-碘-6-(三氟甲基)吡啶(1.77克,收率:76%)。LCMS(ESI):m/z 308[M+1]+;TLC:Rf 0.8(石油醚)。Step 7e: Preparation of 2-chloro-3-iodo-6-(trifluoromethyl)pyridine (Compound 0107-34): To a mixture of 2-chloro-6-(trifluoromethyl)pyridin-3-amine (0106-34) (1.5 g, 7.63 mmol, 1.0 eq) and 6M dilute hydrochloric acid (7.6 mL, 45.78 mmol, 6.0 eq) in acetonitrile (5 mL) was added dropwise a solution of sodium nitrite (0.63 g, 9.16 mmol, 1.2 eq) in water (503 mL) at 0°C. The mixture was stirred at 0°C for 35 minutes. A solution of potassium iodide (2.53 g, 15.26 mmol, 2.0 eq) in water (5 mL) was added. The mixture was warmed to room temperature and then stirred overnight. 2M sodium hydroxide solution was slowly added to adjust pH=8, and the aqueous layer was extracted with ethyl acetate (15 mL×3). The combined organic layer was washed with saturated brine (15 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate = 100: 1) to give a white solid 2-chloro-3-iodo-6-(trifluoromethyl)pyridine (1.77 g, yield: 76%). LCMS (ESI): m/z 308 [M+1] + ; TLC: Rf 0.8 (petroleum ether).

步骤7f:3-((2-氯-6-(三氟甲基)吡啶-3-基)乙炔基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(化合物0108-34)的制备:往2-氯-3-碘-6-(三氟甲基)吡啶(0107-34)(1.0克,3.26毫摩尔,1.08当量),双三苯基磷二氯化钯(106毫克,0.15毫摩尔,0.05当量)和碘化亚铜(29毫克,0.15毫摩尔,0.05当量)在三乙胺(10毫升)的混合物中加入3-乙炔基-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(0105-34)(1.0克,3.03毫摩尔,1.0当量)。混合物在氮气氛围下加热到80℃反应过夜。减压下除去溶剂。残余物用水(30毫升)稀释然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(20毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚:乙酸乙酯=5:1 to 2:1),得到淡黄色油状物3-((2-氯-6-(三氟甲基)吡啶-3-基)乙炔基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(1.36克,收率:88%)。LCMS(ESI):m/z 511[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。Step 7f: Preparation of tert-butyl 3-((2-chloro-6-(trifluoromethyl)pyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0108-34): To a mixture of 2-chloro-3-iodo-6-(trifluoromethyl)pyridine (0107-34) (1.0 g, 3.26 mmol, 1.08 equiv), bistriphenylphosphine palladium dichloride (106 mg, 0.15 mmol, 0.05 equiv) and cuprous iodide (29 mg, 0.15 mmol, 0.05 equiv) in triethylamine (10 ml) was added tert-butyl 3-ethynyl-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (0105-34) (1.0 g, 3.03 mmol, 1.0 equiv). The mixture was heated to 80°C under nitrogen atmosphere and reacted overnight. The solvent was removed under reduced pressure. The residue was diluted with water (30 ml) and then extracted with ethyl acetate (15 ml x 3). The combined organic layer was washed with saturated brine (20 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate = 5: 1 to 2: 1) to obtain a light yellow oily substance tert-butyl 3-((2-chloro-6-(trifluoromethyl)pyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (1.36 g, yield: 88%). LCMS (ESI): m/z 511 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 3: 1).

步骤7g:3-(2-(2-氯-6-(三氟甲基)吡啶-3-基)乙基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(化合物0109-34)的制备:往3-((2-氯-6-(三氟甲基)吡啶-3-基)乙炔基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(0108-34)(1.33克,2.61毫摩尔,1.0当量)在甲醇(10毫升)的混合物中加入二氧化铂(118毫克,0.52毫摩尔,0.2当量)。混合物在氢气球压力下室温搅拌2小时。混合物过滤。滤液减压浓缩,得到淡黄色油状物3-(2-(2-氯-6-(三氟甲基)吡啶-3-基)乙基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(1.29克,收率:96%)。LCMS(ESI):m/z 515[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。Step 7g: Preparation of tert-butyl 3-(2-(2-chloro-6-(trifluoromethyl)pyridin-3-yl)ethyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0109-34): To a mixture of tert-butyl 3-((2-chloro-6-(trifluoromethyl)pyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (0108-34) (1.33 g, 2.61 mmol, 1.0 eq) in methanol (10 mL) was added platinum dioxide (118 mg, 0.52 mmol, 0.2 eq). The mixture was stirred at room temperature under hydrogen balloon pressure for 2 hours. The mixture was filtered. The filtrate was concentrated under reduced pressure to give a light yellow oily substance, tert-butyl 3-(2-(2-chloro-6-(trifluoromethyl)pyridin-3-yl)ethyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (1.29 g, yield: 96%). LCMS (ESI): m/z 515 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=3:1).

步骤7h:1’-(4-甲氧基苄基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0110-34)的制备:往3-(2-(2-氯-6-(三氟甲基)吡啶-3-基)乙基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(0109-34)(1.29克,2.51毫摩尔,1.0当量),醋酸钯(28毫克,0.13毫摩尔,0.05当量)和RuPhos(117毫克,0.25毫摩尔,0.1当量)在二氧六环(12毫升)的混合物 中加入叔丁醇钠(0.48克,5.02毫摩尔,2.0当量)。混合物在微波反应器中加热到115℃反应1小时。减压下除去溶剂。残余物在硅胶上进行柱色谱分离(石油醚:乙酸乙酯=8:1 to 3:1),得到淡黄色油状物1’-(4-甲氧基苄基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(724毫克,收率:60%)。LCMS(ESI):m/z 478[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=5:1)。Step 7h: Preparation of tert-butyl 1'-(4-methoxybenzyl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0110-34): To a mixture of tert-butyl 3-(2-(2-chloro-6-(trifluoromethyl)pyridin-3-yl)ethyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (0109-34) (1.29 g, 2.51 mmol, 1.0 equiv), palladium acetate (28 mg, 0.13 mmol, 0.05 equiv) and RuPhos (117 mg, 0.25 mmol, 0.1 equiv) in dioxane (12 ml) was added. Sodium tert-butoxide (0.48 g, 5.02 mmol, 2.0 eq) was added to the mixture. The mixture was heated to 115°C in a microwave reactor for 1 hour. The solvent was removed under reduced pressure. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate = 8:1 to 3:1) to give a pale yellow oily substance 1'-(4-methoxybenzyl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (724 mg, yield: 60%). LCMS (ESI): m/z 478 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 5:1).

步骤7i:7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0112-34)的制备:1’-(4-甲氧基苄基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0110-34)(724毫克,1.52毫摩尔,1.0当量)在氯化氢甲醇溶液(4M溶液,7.5毫升)的混合物加热到70℃反应2小时。减压下除去溶剂。残余物溶于四氢呋喃(10毫升)和水(10毫升)。加入碳酸钠固体调节pH=9。加入二碳酸二叔丁酯(332毫克,1.52毫摩尔,1.0当量),然后将混合物在室温下搅拌1小时。混合物用水(20毫升)稀释,然后用乙酸乙酯(10毫升×3)萃取。合并的有机层用饱和食盐水(10毫升×1)洗涤,经无水硫酸钠干燥并浓缩,得到淡黄色油状物7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(610毫克,粗品)。LCMS(ESI):m/z 358[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=4:1)。Step 7i: Preparation of tert-butyl 7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0112-34): A mixture of tert-butyl 1'-(4-methoxybenzyl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0110-34) (724 mg, 1.52 mmol, 1.0 eq) in methanolic hydrogen chloride solution (4M solution, 7.5 mL) was heated to 70°C for 2 hours. The solvent was removed under reduced pressure. The residue was dissolved in tetrahydrofuran (10 mL) and water (10 mL). Solid sodium carbonate was added to adjust the pH to 9. Di-tert-butyl dicarbonate (332 mg, 1.52 mmol, 1.0 eq) was added, and the mixture was stirred at room temperature for 1 hour. The mixture was diluted with water (20 ml), and then extracted with ethyl acetate (10 ml×3). The combined organic layer was washed with saturated brine (10 ml×1), dried over anhydrous sodium sulfate and concentrated to give a pale yellow oily substance, tert-butyl 7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (610 mg, crude product). LCMS (ESI): m/z 358[M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=4:1).

步骤7j:6’-溴-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0115-34)的制备:在0℃下往7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0112-34)(543毫克,1.52毫摩尔,1.0当量)在二氯甲烷(10毫升)的混合物中加入二溴海因(261毫克,0.91毫摩尔,0.6当量)。混合物在0℃下搅拌1小时。加入饱和亚硫酸钠水溶液(20毫升)淬灭反应。水层用二氯甲烷(10毫升×3)萃取。合并的有机层用饱和食盐水(10毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚:乙酸乙酯=6:1),得到淡黄色油状物6’-溴-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(659毫克,收率:99%)。LCMS(ESI):m/z 436[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=4:1)。Step 7j: Preparation of tert-butyl 6'-bromo-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0115-34): To a mixture of tert-butyl 7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0112-34) (543 mg, 1.52 mmol, 1.0 eq) in dichloromethane (10 mL) was added dibromohydantoin (261 mg, 0.91 mmol, 0.6 eq) at 0°C. The mixture was stirred at 0°C for 1 hour. Saturated aqueous sodium sulfite solution (20 mL) was added to quench the reaction. The aqueous layer was extracted with dichloromethane (10 mL x 3). The combined organic layer was washed with saturated brine (10 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate = 6: 1) to give a pale yellow oily substance 6'-bromo-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (659 mg, yield: 99%). LCMS (ESI): m/z 436 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 4: 1).

步骤7k:(1-(叔丁氧羰基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(化合物0117-34)的制备:往6’-溴-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0115-34)(250毫克,0.57毫摩尔,1.0当量),Xphos-Pd-G2(9毫克,0.011毫摩尔,0.02当量),Xphos(8毫克,0.017毫摩尔,0.03当量),乙酸钾(140毫克,1.43毫摩尔,2.5当量)和叔丁醇钠(1.4毫克,0.014毫摩尔,0.025当量)在甲醇(6毫升)和乙二醇(0.6毫升)的混合物中加入四羟基二硼(129毫克,1.43毫摩尔,2.5当量)。混合物在氮气氛围下加热至53℃反应10小时。减压下除去溶剂。残余物用水(20毫升)稀释。加入2M氢氧化钠溶液调节pH=12,然后混合物用甲基叔丁基醚(8毫升×3)洗涤。往水层加入2M稀盐酸溶液调节 pH=7,然后用乙酸乙酯(10毫升×3)萃取。合并的有机层用饱和食盐水(10毫升×1)洗涤,经无水硫酸钠干燥并浓缩,得到白色固体(1-(叔丁氧羰基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(151毫克,收率:66%)。LCMS(ESI):m/z 402[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=10:1)。Step 7k: Preparation of (1-(tert-butyloxycarbonyl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (Compound 0117-34): To tert-butyl 6'-bromo-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0115-34) (250 mg, 0.57 mmol, 1.0 equiv. ), Xphos-Pd-G2 (9 mg, 0.011 mmol, 0.02 eq), Xphos (8 mg, 0.017 mmol, 0.03 eq), potassium acetate (140 mg, 1.43 mmol, 2.5 eq) and sodium tert-butoxide (1.4 mg, 0.014 mmol, 0.025 eq) were added to a mixture of methanol (6 ml) and ethylene glycol (0.6 ml). Tetrahydroxydiboron (129 mg, 1.43 mmol, 2.5 eq) was added. The mixture was heated to 53°C under a nitrogen atmosphere for 10 hours. The solvent was removed under reduced pressure. The residue was diluted with water (20 ml). 2M sodium hydroxide solution was added to adjust the pH to 12, and then the mixture was washed with methyl tert-butyl ether (8 ml×3). 2M dilute hydrochloric acid solution was added to the aqueous layer to adjust pH = 7, then extracted with ethyl acetate (10 ml x 3). The combined organic layer was washed with saturated brine (10 ml x 1), dried over anhydrous sodium sulfate and concentrated to give a white solid (1-(tert-butyloxycarbonyl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (151 mg, yield: 66%). LCMS (ESI): m/z 402 [M+1] + ; TLC: Rf 0.5 (dichloromethane: methanol = 10: 1).

步骤7l:6’-(嘧啶-2-基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0119-34)的制备:往(1-(叔丁氧羰基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(0117-34)(151毫克,0.38毫摩尔,1.0当量),Xphos-Pd-G2(9毫克,0.011毫摩尔,0.03当量),Xphos(5.2毫克,0.011毫摩尔,0.03当量)和碳酸钠(120毫克,1.13毫摩尔,3.0当量)在甲苯(2.5毫升),乙醇(2.5毫升)和水(1毫升)的混合物中加入2-溴嘧啶(66毫克,0.42毫摩尔,1.1当量)。混合物在氮气氛围下加热至55℃反应6小时。减压下除去溶剂。残余物在硅胶上进行柱色谱分离(二氯甲烷∶甲醇=100∶1至50:1),得到淡黄色油状物6’-(嘧啶-2-基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(115毫克,收率:70%)。LCMS(ESI):m/z 436[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=20:1)。Step 71: Preparation of tert-butyl 6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0119-34): To (1-(tert-butyloxycarbonyl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (0117-34) (151 mg, 0.38 mmol, 1.0 eq.), Xphos-Pd-G2 (9 mg, 0.011 mmol, 0.03 eq.), Xphos (5.2 mg, 0.011 mmol, 0.03 eq.) and sodium carbonate (120 mg, 1.13 mmol, 3.0 eq.) were added to a mixture of toluene (2.5 ml), ethanol (2.5 ml) and water (1 ml). The mixture was heated to 55° C. under a nitrogen atmosphere for 6 hours. The solvent was removed under reduced pressure. The residue was subjected to column chromatography on silica gel (dichloromethane: methanol = 100: 1 to 50: 1) to give pale yellow oily tert-butyl 6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (115 mg, yield: 70%). LCMS (ESI): m/z 436 [M+1] + ; TLC: Rf 0.5 (dichloromethane: methanol = 20: 1).

步骤7m:6’-(嘧啶-2-基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(化合物0121-34)的制备:6’-(嘧啶-2-基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0119-34)(49毫克,0.113毫摩尔,1.0当量)在氯化氢甲醇溶液(4M溶液,1.5毫升)的混合物在室温下搅拌3.5小时。减压下除去溶剂。残余物在真空下干燥,得到淡黄色油状物6’-(嘧啶-2-基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(46毫克,粗品)。LCMS(ESI):m/z 336[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=10:1)。Step 7m: Preparation of 6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0121-34): A mixture of tert-butyl 6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0119-34) (49 mg, 0.113 mmol, 1.0 eq) in methanolic hydrogen chloride solution (4M solution, 1.5 mL) was stirred at room temperature for 3.5 hours. The solvent was removed under reduced pressure. The residue was dried under vacuum to give 6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (46 mg, crude) as a pale yellow oil. LCMS (ESI): m/z 336 [M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=10:1).

步骤7n:(2R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-(6’-(嘧啶-2-基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(化合物34)的制备:往(R)-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(化合物0406-7)(22.5毫克,0.113毫摩尔,1.0当量)在乙腈(3毫升)的混合物中加入咪唑三氟甲磺酸盐(37毫克,0.17毫摩尔,1.5当量)和羰基二咪唑(22毫克,0.14毫摩尔,1.2当量)。混合物在室温下搅拌1.5小时。加入6’-(嘧啶-2-基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(化合物0121-34)(46毫克,0.113毫摩尔,1.0当量)和N,N-二异丙基乙胺(73毫克,0.57毫摩尔,5.0当量)。混合物在室温下搅拌过夜。混合物用水(30毫升)稀释然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(20毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(石油醚:乙酸乙酯=1:3)纯化,得到白色固体(2R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-(6’-(嘧啶-2-基)-7’-(三氟甲基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(16毫克,收率:28%)。LCMS(ESI):m/z 517[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=1:3)。1HNMR(500MHz,MeOD)δ8.83(s,2H),7.95(t,J=22.6Hz, 1H),7.62(d,J=10.2Hz,1H),7.43(d,J=2.1Hz,1H),6.81–6.70(m,1H),4.34–4.14(m,1H),3.86(d,J=10.8Hz,3H),3.78–3.38(m,4H),3.00–2.62(m,2H),2.00(dddd,J=47.1,41.6,24.0,15.9Hz,4H),1.46(dd,J=11.5,5.9Hz,3H).Step 7n: Preparation of (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 34): To a mixture of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (Compound 0406-7) (22.5 mg, 0.113 mmol, 1.0 eq) in acetonitrile (3 mL) were added imidazole triflate (37 mg, 0.17 mmol, 1.5 eq) and carbonyldiimidazole (22 mg, 0.14 mmol, 1.2 eq). The mixture was stirred at room temperature for 1.5 hours. 6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0121-34) (46 mg, 0.113 mmol, 1.0 eq) and N,N-diisopropylethylamine (73 mg, 0.57 mmol, 5.0 eq) were added. The mixture was stirred at room temperature overnight. The mixture was diluted with water (30 mL) and then extracted with ethyl acetate (15 mL×3). The combined organic layer was washed with saturated brine (20 mL×1), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (petroleum ether:ethyl acetate=1:3) to give a white solid (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(6'-(pyrimidin-2-yl)-7'-(trifluoromethyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (16 mg, yield: 28%). LCMS (ESI): m/z 517 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=1:3). 1 HNMR (500 MHz, MeOD) δ8.83 (s, 2H), 7.95 (t, J=22.6 Hz, 1H),7.62(d,J=10.2Hz,1H),7.43(d,J=2.1Hz,1H),6.81–6.70(m,1H),4.34–4.14(m,1H),3.86(d,J=10.8Hz,3H), 3.78–3.38(m,4H),3.00–2.62(m,2H),2.00(dddd,J=47.1,41.6,24.0,15.9Hz,4H),1.46(dd,J=11.5,5.9Hz,3H).

实施例8:(2R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-(7’-氟-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(化合物37)的制备(按照方案一和四线路制备)Example 8: Preparation of (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 37) (Prepared according to Schemes 1 and 4)

步骤8a:3-((2,6-二氟吡啶-3-基)乙炔基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(化合物0108-37)的制备:往2,6-二氟-3-碘吡啶(422毫克,1.75毫摩尔,1.05当量),双三苯基磷二氯化钯(59毫克,0.084毫摩尔,0.05当量)和碘化亚铜(16毫克,0.084毫摩尔,0.05当量)在三乙胺(8毫升)的混合物中加入3-乙炔基-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(化合物0105-34)(550毫克,1.67毫摩尔,1.0当量)。混合物在氮气氛围下加热到80℃反应3小时。减压下除去溶剂。残余物用水(30毫升)稀释然后用乙酸乙酯(20毫升×3)萃取。合并的有机层用饱和食盐水(20毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚:乙酸乙酯=5:1 to 1:1),得到淡黄色油状物3-((2,6-二氟吡啶-3-基)乙炔基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(600毫克,收率:81%)。LCMS(ESI):m/z 444[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。Step 8a: Preparation of tert-butyl 3-((2,6-difluoropyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0108-37): To a mixture of 2,6-difluoro-3-iodopyridine (422 mg, 1.75 mmol, 1.05 eq), bistriphenylphosphine palladium dichloride (59 mg, 0.084 mmol, 0.05 eq) and cuprous iodide (16 mg, 0.084 mmol, 0.05 eq) in triethylamine (8 ml) was added tert-butyl 3-ethynyl-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0105-34) (550 mg, 1.67 mmol, 1.0 eq). The mixture was heated to 80° C. under nitrogen atmosphere for 3 hours. The solvent was removed under reduced pressure. The residue was diluted with water (30 ml) and then extracted with ethyl acetate (20 ml x 3). The combined organic layer was washed with saturated brine (20 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate = 5: 1 to 1: 1) to obtain a light yellow oily substance tert-butyl 3-((2,6-difluoropyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (600 mg, yield: 81%). LCMS (ESI): m/z 444 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 3: 1).

步骤8b:7’-氟-1’-(4-甲氧基苄基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0110-37)的制备:往3-((2,6-二氟吡啶-3-基)乙炔基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(0108-37)(600毫克,1.35毫摩尔,1.0当量)在甲醇(10毫升)的混合物中加入二氧化铂(62毫克,0.27毫摩尔,0.2当量)。混合物在氢气球压力下室温搅拌2.5小时。混合物过滤。滤液减压浓缩。残余物在硅胶上进行柱色谱分离(石油醚:乙酸乙酯=5:1 to 3:1),得到淡黄色油状物7’-氟-1’-(4-甲氧基苄基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(315毫克,收率:55%)。LCMS(ESI):m/z 428[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。Step 8b: Preparation of tert-butyl 7'-fluoro-1'-(4-methoxybenzyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0110-37): To a mixture of tert-butyl 3-((2,6-difluoropyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (0108-37) (600 mg, 1.35 mmol, 1.0 eq) in methanol (10 mL) was added platinum dioxide (62 mg, 0.27 mmol, 0.2 eq). The mixture was stirred at room temperature under hydrogen balloon pressure for 2.5 hours. The mixture was filtered. The filtrate was concentrated under reduced pressure. The residue was separated by column chromatography on silica gel (petroleum ether:ethyl acetate=5:1 to 3:1) to give a pale yellow oily substance, tert-butyl 7'-fluoro-1'-(4-methoxybenzyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (315 mg, yield: 55%). LCMS (ESI): m/z 428 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=3:1).

步骤8c:7’-氟-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0112-37)的制备:往7’-氟-1’-(4-甲氧基苄基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0110-37)(315毫克,0.74毫摩尔,1.0当量)在二氯甲烷(6毫升)的混合物中加入三氟乙酸(1.5毫升)。混合物在室温下搅拌4.5小时。减压下除去溶剂。残余物溶于水(10毫升)和四氢呋喃(10毫升)。加入碳酸钠固体调节pH=9。加入二碳酸二叔丁酯(403毫克,1.84毫摩尔,2.5当量),然后将混合物在室温下搅拌1小时。混合物用水(30毫升)稀释,然后用乙酸乙酯(20毫升×3)萃取。合并的有机层用饱和食盐水(20毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯=5:1),得到淡黄色油状物7’-氟-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(227毫克,收率:100%)。LCMS(ESI):m/z 308[M+1]+; TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。Step 8c: Preparation of tert-butyl 7'-fluoro-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0112-37): To a mixture of tert-butyl 7'-fluoro-1'-(4-methoxybenzyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0110-37) (315 mg, 0.74 mmol, 1.0 eq) in dichloromethane (6 mL) was added trifluoroacetic acid (1.5 mL). The mixture was stirred at room temperature for 4.5 hours. The solvent was removed under reduced pressure. The residue was dissolved in water (10 mL) and tetrahydrofuran (10 mL). Solid sodium carbonate was added to adjust the pH to 9. Di-tert-butyl dicarbonate (403 mg, 1.84 mmol, 2.5 eq) was added, and the mixture was stirred at room temperature for 1 hour. The mixture was diluted with water (30 mL), and then extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with saturated brine (20 mL×1), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate = 5:1) to give a light yellow oily substance, tert-butyl 7'-fluoro-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (227 mg, yield: 100%). LCMS (ESI): m/z 308[M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=3:1).

步骤8d:7’-氟-6’-碘-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0115-37)的制备:在0℃下往7’-氟-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0112-37)(227毫克,0.74毫摩尔,1.0当量)在乙腈(3.5毫升)的混合物中加入N-碘代丁二酰亚胺(166毫克,0.74毫摩尔,1.0当量)。混合物在0℃下搅拌1小时。加入饱和亚硫酸钠水溶液(20毫升)淬灭反应。水层用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(20毫升×1)洗涤,经无水硫酸钠干燥并浓缩,得到淡黄色油状物7’-氟-6’-碘-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(277毫克,收率:87%)。LCMS(ESI):m/z 434[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。Step 8d: Preparation of tert-butyl 7'-fluoro-6'-iodo-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0115-37): To a mixture of tert-butyl 7'-fluoro-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0112-37) (227 mg, 0.74 mmol, 1.0 equiv) in acetonitrile (3.5 mL) at 0°C was added N-iodosuccinimide (166 mg, 0.74 mmol, 1.0 equiv). The mixture was stirred at 0°C for 1 hour. Saturated aqueous sodium sulfite solution (20 mL) was added to quench the reaction. The aqueous layer was extracted with ethyl acetate (15 mL x 3). The combined organic layer was washed with saturated brine (20 mL x 1), dried over anhydrous sodium sulfate and concentrated to give a pale yellow oily substance, tert-butyl 7'-fluoro-6'-iodo-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (277 mg, yield: 87%). LCMS (ESI): m/z 434 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=3:1).

步骤8e:7’-氟-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0119-37)的制备:往7’-氟-6’-碘-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0115-37)(240毫克,0.55毫摩尔,1.0当量),Xphos-Pd-G3(23毫克,0.028毫摩尔,0.05当量),Xphos(13.3毫克,0.028毫摩尔,0.05当量),碘化亚铜(10.5毫克,0.055毫摩尔,0.1当量)和氟化铯(167毫克,1.10毫摩尔,2.0当量)在N,N-二甲基甲酰胺(6毫升)的混合物中加入2-(三丁基锡基)嘧啶(406毫克,1.10毫摩尔,2.0当量)。混合物在微波反应器中加热到100℃反应50分钟。混合物用水(30毫升)稀释然后用乙酸乙酯(10毫升×3)萃取。合并的有机层用饱和食盐水(20毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(乙酸乙酯:石油醚=3:2)纯化,得到淡黄色油状物7’-氟-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(43毫克,收率:20%)。LCMS(ESI):m/z 386[M+1]+;TLC:Rf 0.5(乙酸乙酯:石油醚=3:2)。Step 8e: Preparation of tert-butyl 7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0119-37): To tert-butyl 7'-fluoro-6'-iodo-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0115-37) (240 mg, 0.55 mmol, 1.0 equiv), Xphos- Pd-G3 (23 mg, 0.028 mmol, 0.05 eq), Xphos (13.3 mg, 0.028 mmol, 0.05 eq), cuprous iodide (10.5 mg, 0.055 mmol, 0.1 eq) and cesium fluoride (167 mg, 1.10 mmol, 2.0 eq) were added to a mixture of N,N-dimethylformamide (6 ml) and 2-(tributyltinyl)pyrimidine (406 mg, 1.10 mmol, 2.0 eq). The mixture was heated to 100°C in a microwave reactor for 50 minutes. The mixture was diluted with water (30 ml) and then extracted with ethyl acetate (10 ml x 3). The combined organic layer was washed with saturated brine (20 ml x 1), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (ethyl acetate:petroleum ether=3:2) to give a pale yellow oily substance, tert-butyl 7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (43 mg, yield: 20%). LCMS (ESI): m/z 386 [M+1] + ; TLC: Rf 0.5 (ethyl acetate:petroleum ether=3:2).

步骤8f:7’-氟-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(化合物0121-37)的制备:7’-氟-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0119-37)(42毫克,0.11毫摩尔,1.0当量)在氯化氢乙酸乙酯溶液(4M溶液,2.5毫升)中的混合物在室温下搅拌2.5小时。减压下除去溶剂。残余物在真空下干燥,得到淡黄色固体7’-氟-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(39毫克,粗品)。LCMS(ESI):m/z 286[M+1]+;TLC:Rf 0.3(二氯甲烷:甲醇=10:1)。Step 8f: Preparation of 7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0121-37): A mixture of tert-butyl 7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0119-37) (42 mg, 0.11 mmol, 1.0 eq) in ethyl acetate solution of hydrogen chloride (4M solution, 2.5 mL) was stirred at room temperature for 2.5 hours. The solvent was removed under reduced pressure. The residue was dried under vacuum to give pale yellow solid 7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (39 mg, crude product). LCMS (ESI): m/z 286 [M+1] + ; TLC: Rf 0.3 (dichloromethane:methanol=10:1).

步骤8g:(2R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-(7’-氟-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(化合物37)的制备:往(R)-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(化合物0406-7)(22毫克,0.11mmol,1.0当量),2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(50毫克,0.13毫摩尔,1.2当量)和N,N-二异丙基乙胺(71毫克,0.55毫摩尔,5.0当量)在二氯甲烷(3毫升)的混合物中加入7’-氟-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷 -3,2’-[1,8]萘啶]二盐酸盐(化合物0121-37)(39毫克,0.11毫摩尔,1.0当量)。混合物在室温下搅拌1小时。混合物用水(30毫升)稀释然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(20毫升×1)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(二氯甲烷:乙酸乙酯:甲醇=10:10:1)纯化,得到白色固体(2R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-(7’-氟-6’-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(25毫克,收率:48%)。LCMS(ESI):m/z 476[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=15:1)。1HNMR(500MHz,MeOD)δ8.77(s,2H),8.13(d,J=9.2Hz,1H),7.91(d,J=64.6Hz,1H),7.28(s,1H),6.75(d,J=22.1Hz,1H),4.22(d,J=36.4Hz,1H),3.87(d,J=12.1Hz,3H),3.77–3.36(m,4H),2.72(d,J=114.3Hz,2H),2.16–1.71(m,4H),1.43(dd,J=13.0,6.6Hz,3H).Step 8g: Preparation of (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 37): To (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (Compound 0406-7) (22 mg, 0. 11 mmol, 1.0 equiv), 2-(7-azabenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (50 mg, 0.13 mmol, 1.2 equiv) and N,N-diisopropylethylamine (71 mg, 0.55 mmol, 5.0 equiv) were added to a mixture of dichloromethane (3 ml) with 7'-fluoro-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine -3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0121-37) (39 mg, 0.11 mmol, 1.0 eq). The mixture was stirred at room temperature for 1 hour. The mixture was diluted with water (30 ml) and then extracted with ethyl acetate (15 ml × 3). The combined organic layer was washed with saturated brine (20 ml × 1), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (dichloromethane: ethyl acetate: methanol = 10: 10: 1) to give a white solid (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(7'-fluoro-6'-(pyrimidine-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-one (25 mg, yield: 48%). LCMS (ESI): m/z 476 [M+1] + ; TLC: Rf 0.5 (dichloromethane: methanol = 15: 1). 1 H NMR (500 MHz, MeOD) δ 8.77 (s, 2H), 8.13 (d, J = 9.2 Hz, 1H), 7.91 (d, J = 64.6 Hz, 1H), 7.28 (s, 1H), 6.75 (d, J = 22.1 Hz, 1H), 4.22 (d, J = 36.4 Hz, 1H), 3.87 (d, J = 12.1 Hz, 3H), 3.77-3.36 (m, 4H), 2.72 (d, J = 114.3 Hz, 2H), 2.16-1.71 (m, 4H), 1.43 (dd, J = 13.0, 6.6 Hz, 3H).

实施例9:2-(5-氟-2-甲氧基吡啶-4-基)-3-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-基)-3-氧代丙腈(化合物41)的制备
Example 9: Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)-3-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-3-oxopropionitrile (Compound 41)

在氮气保护下,向2-氰基乙酸乙酯(335毫克,2.96毫摩尔,1.5当量)、吡啶甲酸(50毫克,0.40毫摩尔,0.2当量)、碘化亚铜(37.5毫克,0.20毫摩尔,0.1当量)和碳酸铯(730毫克,2.24毫摩尔,1.1当量)的四氢呋喃溶液(20毫升)中滴入5-氟-4-碘-2-甲氧基吡啶(0202-1)(500毫克,1.98毫摩尔,1.0当量)的四氢呋喃(5毫升)溶液,将反应在70℃搅拌过夜。反应完成后,将混合物过滤,滤饼用乙酸乙酯(20毫升×3)洗涤,合并的滤液,依次用饱和氯化铵水溶液(100毫升)和饱和食盐水洗涤,硫酸钠干燥,过滤,减压浓缩。残留物用硅胶柱(洗脱剂:10%至20%乙酸乙酯的石油醚溶液)纯化,得到白色固体产物2-氰基-2-(5-氟-2-甲氧基吡啶-4-基)乙酸乙酯(355毫克,收率:75.2%)。LCMS(ESI):m/z=239[M+1]+。氮气保护下,向2-氰基-2-(5-氟-2-甲氧基吡啶-4-基)乙酸乙酯(43毫克,0.16毫摩尔,1.0当量)和7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(化合物0122-1)(50毫克,0.18毫摩尔,1.0当量)的甲苯(10毫升)溶液中加入N,N-二异丙基乙胺(0.5毫升,2.87毫摩尔,18当量),将反应在120℃搅拌过夜。反应完成后,将反应混合物加水稀释,用乙酸乙酯(10毫升×3)萃取,合并的有机相用饱和食盐水洗涤,硫酸钠干燥,过滤,45℃减压浓缩。残留物通过制备型薄层色谱(洗脱剂:二氯甲烷/甲醇=10/1)纯化,得到白色固体产物2-(5-氟-2-甲氧基吡啶-4-基)-3-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-基)-3-氧代丙腈(15毫克,收率:17.6%)。LCMS(ESI):m/z=474.30[M+1]+1HNMR(500MHz,DMSO)δ8.88(d,J=4.6Hz,2H),8.38– 8.25(m,1H),7.97(d,J=6.3Hz,1H),7.48–7.31(m,2H),6.99–6.88(m,1H),5.92–5.80(m,1H),4.08–3.90(m,4H),3.83–3.69(m,2H),3.58(d,J=9.9Hz,2H),2.83(dd,J=16.2,6.3Hz,2H),2.65(d,J=8.4Hz,3H),2.15–2.04(m,2H),1.85(tdd,J=21.6,15.6,8.4Hz,2H).Under nitrogen protection, a solution of 5-fluoro-4-iodo-2-methoxypyridine (0202-1) (500 mg, 1.98 mmol, 1.0 eq) in tetrahydrofuran (5 ml) was added dropwise to a solution of ethyl 2-cyanoacetate (335 mg, 2.96 mmol, 1.5 eq), picolinic acid (50 mg, 0.40 mmol, 0.2 eq), cuprous iodide (37.5 mg, 0.20 mmol, 0.1 eq) and cesium carbonate (730 mg, 2.24 mmol, 1.1 eq) in tetrahydrofuran (20 ml), and the reaction was stirred at 70° C. overnight. After the reaction was completed, the mixture was filtered, the filter cake was washed with ethyl acetate (20 ml×3), and the combined filtrate was washed with saturated aqueous ammonium chloride (100 ml) and saturated brine in sequence, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column (eluent: 10% to 20% ethyl acetate in petroleum ether) to give ethyl 2-cyano-2-(5-fluoro-2-methoxypyridin-4-yl)acetate (355 mg, yield: 75.2%) as a white solid product. LCMS (ESI): m/z=239[M+1] + . Under nitrogen protection, N,N-diisopropylethylamine (0.5 ml, 2.87 mmol, 18 eq) was added to a toluene (10 ml) solution of ethyl 2-cyano-2-(5-fluoro-2-methoxypyridin-4-yl)acetate (43 mg, 0.16 mmol, 1.0 eq) and 7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0122-1) (50 mg, 0.18 mmol, 1.0 eq), and the reaction was stirred at 120° C. overnight. After the reaction was completed, the reaction mixture was diluted with water and extracted with ethyl acetate (10 ml×3), and the combined organic phase was washed with saturated brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure at 45° C. The residue was purified by preparative thin layer chromatography (eluent: dichloromethane/methanol=10/1) to give a white solid product 2-(5-fluoro-2-methoxypyridin-4-yl)-3-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-3-oxopropanenitrile (15 mg, yield: 17.6%). LCMS (ESI): m/z=474.30 [M+1] + . 1 HNMR (500 MHz, DMSO) δ8.88 (d, J=4.6 Hz, 2H), 8.38– 8.25(m,1H),7.97(d,J=6.3Hz,1H),7.48–7.31(m,2H),6.99–6.88(m,1H),5.92–5.80(m,1H),4.08–3.90(m,4H),3.83–3.69(m,2H), 3.58(d,J=9.9Hz,2H),2.83(dd,J=16.2,6.3Hz,2H),2.65(d,J=8.4Hz,3H),2.15–2.04(m,2H),1.85(tdd,J=21.6,15.6,8.4Hz,2H).

实施例10:2-(2-甲氧基-5-(甲磺酰基)吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物31)的制备(按照方案四线路制备)Example 10: Preparation of 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 31) (Prepared according to Scheme 4)

步骤10a:2-(2-甲氧基-5-(甲硫基)吡啶-4-基)丙二酸二甲酯(化合物0403-31)的制备:往6-甲氧基-4-甲基吡啶-3-胺(1克,7.2毫摩尔,1.0当量)的二甲基二硫醚(10毫升)混合物中加入亚硝酸叔丁酯(5.96克,57.9毫摩尔,8.0当量),并将混合物在50℃下搅拌半小时。反应液用水稀释,乙酸乙酯萃取,将有机相干燥并减压浓缩得到油状物粗品。残留物用硅胶柱层析(洗脱剂为:石油醚)纯化得到黄色油状物2-甲氧基-4-甲基-5-(甲硫基)吡啶(710毫克,产率59%)。MS(ES+):m/z=170(M+H)+。氮气保护下,将二异丙基氨基锂的四氢呋喃溶液(1.6M,6.7毫升,10.8毫摩尔,3.0当量)滴加到-30℃的2-甲氧基-4-甲基-5-(甲硫基)吡啶(610毫克,3.6毫摩尔,1.0当量)的四氢呋喃(5毫升)溶液中。将反应混合物在-30℃下搅拌1小时后,逐滴加入碳酸二甲酯(975毫克,10.8毫摩尔,3.0当量)。加完后,将反应混合物加热至25℃并搅拌1小时。反应液加水稀释后,用乙酸乙酯萃取。有机相用无水硫酸钠干燥并减压浓缩,残留物用硅胶柱层析(洗脱剂为:石油醚/乙酸乙酯=10/1)纯化得到黄色油状物2-(2-甲氧基-5-(甲硫基)吡啶-4-基)丙二酸二甲酯(290毫克,产率29%)。LCMS(ESI):m/z=286[M+1]+.Step 10a: Preparation of dimethyl 2-(2-methoxy-5-(methylthio)pyridin-4-yl)malonate (Compound 0403-31): To a mixture of 6-methoxy-4-methylpyridin-3-amine (1 g, 7.2 mmol, 1.0 eq) in dimethyl disulfide (10 ml) was added tert-butyl nitrite (5.96 g, 57.9 mmol, 8.0 eq) and the mixture was stirred at 50°C for half an hour. The reaction solution was diluted with water, extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure to give a crude oil. The residue was purified by silica gel column chromatography (eluent: petroleum ether) to give 2-methoxy-4-methyl-5-(methylthio)pyridine (710 mg, yield 59%) as a yellow oil. MS (ES + ): m/z=170 (M+H) + . Under nitrogen protection, a tetrahydrofuran solution of lithium diisopropylamide (1.6M, 6.7 ml, 10.8 mmol, 3.0 eq) was added dropwise to a tetrahydrofuran (5 ml) solution of 2-methoxy-4-methyl-5-(methylthio)pyridine (610 mg, 3.6 mmol, 1.0 eq) at -30°C. After the reaction mixture was stirred at -30°C for 1 hour, dimethyl carbonate (975 mg, 10.8 mmol, 3.0 eq) was added dropwise. After the addition was complete, the reaction mixture was heated to 25°C and stirred for 1 hour. The reaction solution was diluted with water and extracted with ethyl acetate. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 10/1) to obtain yellow oily 2-(2-methoxy-5-(methylthio)pyridin-4-yl)malonate (290 mg, yield 29%). LCMS (ESI): m/z = 286 [M+1] + .

步骤10b:2-(2-甲氧基-5-(甲硫基)吡啶-4-基)-2-甲基丙二酸二甲酯(化合物0404-31)的制备:氮气保护,向2-(2-甲氧基-5-(甲硫基)吡啶-4-基)丙二酸二甲酯(0403-31)(290毫克,1.0毫摩尔,1.0当量)和碳酸铯(501毫克,1.5毫摩尔,1.5当量)在N,N-二甲基甲酰胺(5毫升)中的混合物中加入碘甲烷(145毫克,1.0摩尔,1.0当量),并将混合物在35℃下搅拌过夜。冷却至室温后,反应液用水稀释,乙酸乙酯萃取。有机相用饱和食盐水洗涤,并用无水硫酸钠干燥并减压浓缩。残留物用硅胶柱层析(洗脱剂为:石油醚/乙酸乙酯=3/1)得到黄色油状物2-(2-甲氧基-5-(甲硫基)吡啶-4-基)-2-甲基丙二酸二甲酯(275毫克,产率95%)。LCMS(ESI):m/z=300[M+1]+.Step 10b: Preparation of dimethyl 2-(2-methoxy-5-(methylthio)pyridin-4-yl)-2-methylmalonate (Compound 0404-31): Under nitrogen protection, iodomethane (145 mg, 1.0 mol, 1.0 eq.) was added to a mixture of dimethyl 2-(2-methoxy-5-(methylthio)pyridin-4-yl)malonate (0403-31) (290 mg, 1.0 mmol, 1.0 eq.) and cesium carbonate (501 mg, 1.5 mmol, 1.5 eq.) in N,N-dimethylformamide (5 ml), and the mixture was stirred at 35°C overnight. After cooling to room temperature, the reaction solution was diluted with water and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 3/1) to give yellow oily product 2-(2-methoxy-5-(methylthio)pyridin-4-yl)-2-methylmalonic acid dimethyl ester (275 mg, yield 95%). LCMS (ESI): m/z = 300 [M+1] + .

步骤10c:2-(2-甲氧基-5-(甲基磺酰基)吡啶-4-基)丙酸(化合物0405-31)的制备:将2-(2-甲氧基-5-(甲硫基)吡啶-4-基)-2-甲基丙二酸二甲酯(0404-31)(275毫克,0.9毫摩尔,1.0当量)和氢氧化钠(183.8毫克,4.6毫摩尔,5.0当量)在乙醇/水=5/1(10毫升/2毫升)中的混合物在85℃下搅拌过夜。反应液用稀盐酸调pH至2~3,乙酸乙酯萃取,将有机相干燥并减压浓缩得到黄色油状物2-(5-氟-2-甲基苯并呋喃-6-基)丙酸(160毫克,产率76.9%)。LCMS(ESI):m/z=228[M+1]+。将2-(2-甲氧基-5-(甲硫基)吡啶-4-基)丙酸(160毫克,0.7毫摩尔,1.0当量)和过氧单磺酸钾(866.6毫克,1.4毫摩尔,2当量)在乙醇(5毫升)中的混合物在室温下搅拌过夜。反应溶液用氯化铵溶 液稀释,用乙酸乙酯萃取,有机相干燥并减压浓缩,得到粗品油状物。粗品用柱层析(洗脱液:二氯甲烷/甲醇=10/1)纯化,得到2-(2-甲氧基-5-(甲基磺酰基)吡啶-4-基)丙酸(160毫克,产率87.9%)。LCMS(ESI):m/z=260[M+1]+.Step 10c: Preparation of 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)propanoic acid (Compound 0405-31): A mixture of dimethyl 2-(2-methoxy-5-(methylthio)pyridin-4-yl)-2-methylmalonate (0404-31) (275 mg, 0.9 mmol, 1.0 eq) and sodium hydroxide (183.8 mg, 4.6 mmol, 5.0 eq) in ethanol/water = 5/1 (10 ml/2 ml) was stirred at 85°C overnight. The reaction solution was adjusted to pH 2-3 with dilute hydrochloric acid, extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure to give 2-(5-fluoro-2-methylbenzofuran-6-yl)propanoic acid (160 mg, yield 76.9%) as a yellow oil. LCMS (ESI): m/z = 228 [M+1] + . A mixture of 2-(2-methoxy-5-(methylthio)pyridin-4-yl)propanoic acid (160 mg, 0.7 mmol, 1.0 eq) and potassium peroxymonosulfonate (866.6 mg, 1.4 mmol, 2 eq) in ethanol (5 ml) was stirred at room temperature overnight. The reaction solution was diluted with ammonium chloride. The mixture was diluted with ethyl acetate and extracted with ethyl acetate. The organic phase was dried and concentrated under reduced pressure to obtain a crude oil. The crude product was purified by column chromatography (eluent: dichloromethane/methanol = 10/1) to obtain 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)propanoic acid (160 mg, yield 87.9%). LCMS (ESI): m/z = 260 [M+1] + .

步骤10d:2-(2-甲氧基-5-(甲磺酰基)吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物31,0412-31)制备:往2-(2-甲氧基-5-(甲基磺酰基)吡啶-4-基)丙酸(0405-31)(50毫克,0.2毫摩尔,1.05单量),7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-1)(65毫克,0.18毫摩尔,1.0单量)和N,N-二异丙基乙胺(94.5毫克,0.7毫摩尔,4.0单量)的N,N-二甲基甲酰胺(5毫升)混合物中加入2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(90.5毫克,0.24毫摩尔,1.3单量)。反应液在室温下搅拌两小时。将反应液加水稀释,乙酸乙酯萃取。有机相用饱和食盐水洗涤,无水硫酸钠干燥并减压浓缩。残留物用厚柱层析(洗脱剂为:石油醚/乙酸乙酯/氨水=1/10/1滴)得到黄色固体2-(2-甲氧基-5-(甲磺酰基)吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(44毫克,产率45.8%)。LCMS(ESI):m/z=523[M+1]+.1H NMR(500MHz,MeOD)δ8.81(s,2H),8.70(dd,J=28.5,23.2Hz,1H),7.92(d,J=60.3Hz,1H),7.34–7.22(m,1H),6.97(dd,J=15.9,7.4Hz,1H),4.02(s,3H),3.95–3.80(m,1H),3.76–3.39(m,4H),3.24–3.15(m,3H),2.84(d,J=15.8Hz,2H),2.58(d,J=5.9Hz,3H),2.23–1.78(m,4H),1.61–1.47(m,3H).Step 10d: Preparation of 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 31, 0412-31): To 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)propanoic acid (0405-31) (50 mg, 0.2 mmol, 1.05 equiv), 7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one 2-(7-azabenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (90.5 mg, 0.24 mmol, 1.3 molar amounts) was added to a mixture of (pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-1) (65 mg, 0.18 mmol, 1.0 molar amounts) and N,N-diisopropylethylamine (94.5 mg, 0.7 mmol, 4.0 molar amounts) in N,N-dimethylformamide (5 ml). The reaction solution was stirred at room temperature for two hours. The reaction solution was diluted with water and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by thick column chromatography (eluent: petroleum ether/ethyl acetate/ammonia water = 1/10/1 drop) to give a yellow solid 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (44 mg, 45.8% yield). LCMS (ESI): m/z=523[M+1] + . 1 H NMR(500MHz,MeOD)δ8.81(s,2H),8.70(dd,J=28.5,23.2Hz,1H),7.92(d,J=60.3Hz,1H),7.34–7.22(m,1H),6.97(dd,J=15.9,7.4Hz,1H),4.02(s, 3H),3.95–3.80(m,1H),3.76–3.39(m,4H),3.24–3.15(m,3H),2.84(d,J= 15.8Hz,2H),2.58(d,J=5.9Hz,3H),2.23–1.78(m,4H),1.61–1.47(m,3H).

实施例11:2-(2-甲氧基-5-(甲磺酰基)吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-硫酮(化合物70)的制备(按照方案五线路制备)Example 11: Preparation of 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (Compound 70) (Prepared according to Scheme 5)

将2-(2-甲氧基-5-(甲基磺酰基)吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3'-二氢-1'-吡咯烷-3,2'-[1,8]萘吡啶)-1-基)丙-1-酮(0412-31)(15毫克,0.03毫摩尔,1.0当量)和劳森试剂(17毫克,0.04毫摩尔,1.2当量)在甲苯(1毫升)中的混合物在125℃的微波反应器中搅拌1小时。反应溶液用水稀释,用乙酸乙酯萃取,有机相干燥并减压浓缩,得到粗品油状物。粗品用厚柱层析(洗脱液:二氯甲烷/甲醇=10/1)纯化,得到2-(2-甲氧基-5-(甲磺酰基)吡啶-4-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-硫酮(9毫克,产率60%)。LCMS(ESI):m/z=539[M+1]+.1H NMR(500MHz,CDCl3)δ8.76–8.63(m,2H),8.02–7.81(m,1H),7.17(s,1H),7.11–7.00(m,1H),6.90(dd,J=20.4,4.6Hz,1H),5.35–5.00(m,1H),4.46–3.97(m,2H),3.94(d,J=13.1Hz,3H),3.91–3.51(m,2H),3.04(dd,J=28.4,7.2Hz,3H),2.80(t,J=10.7Hz,2H),2.64(dd,J=16.3,9.1Hz,3H),2.37–2.07(m,2H),1.99(dd,J=32.6,16.3Hz,2H),1.55–1.46(m,3H).A mixture of 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3'-dihydro-1'-pyrrolidin-3,2'-[1,8]naphthyridine)-1-yl)propan-1-one (0412-31) (15 mg, 0.03 mmol, 1.0 eq) and Lawesson's reagent (17 mg, 0.04 mmol, 1.2 eq) in toluene (1 ml) was stirred in a microwave reactor at 125° C. for 1 hour. The reaction solution was diluted with water, extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure to obtain a crude oil. The crude product was purified by thick column chromatography (eluent: dichloromethane/methanol = 10/1) to give 2-(2-methoxy-5-(methylsulfonyl)pyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (9 mg, yield 60%). LCMS (ESI): m/z = 539 [M+1] + . 1 H NMR (500 MHz, CDCl 3 )δ8.76–8.63(m,2H),8.02–7.81(m,1H),7.17(s,1H),7.11–7.00(m,1H),6.90(dd,J =20.4,4.6Hz,1H),5.35–5.00(m,1H),4.46–3.97(m,2H),3.94(d,J=13.1Hz,3H),3.9 1–3.51(m,2H),3.04(dd,J=28.4,7.2Hz,3H),2.80(t,J=10.7Hz,2H),2.64(dd,J=16 .3,9.1Hz,3H),2.37–2.07(m,2H),1.99(dd,J=32.6,16.3Hz,2H),1.55–1.46(m,3H).

实施例12:2-(5-氟苯并呋喃-6-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷 -3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物14)的制备
Example 12: 2-(5-fluorobenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine Preparation of 3,2'-[1,8]naphthyridin-1-yl)propan-1-one (Compound 14)

步骤12a:4-氟-2-碘-5-甲基苯酚的制备:将4-氟-3-甲基苯酚(5.0克,39.6毫摩尔,1.0当量))、一水合对甲苯磺酸(7.5克,39.6毫摩尔,1.0当量)和N-碘代丁二酰亚胺(9.8克,43.6毫摩尔,1.1当量)的乙腈(50毫升)混合物在常温下搅拌2小时。反应液用亚硫酸钠溶液淬灭,并用乙酸乙酯萃取。有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩。残留物用硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=1/0至10/1)纯化,得到4-氟-2-碘-5-甲基苯酚(7.65克,收率:75.6%)为黄色油状物。Step 12a: Preparation of 4-fluoro-2-iodo-5-methylphenol: A mixture of 4-fluoro-3-methylphenol (5.0 g, 39.6 mmol, 1.0 eq.), p-toluenesulfonic acid monohydrate (7.5 g, 39.6 mmol, 1.0 eq.) and N-iodosuccinimide (9.8 g, 43.6 mmol, 1.1 eq.) in acetonitrile (50 ml) was stirred at room temperature for 2 hours. The reaction solution was quenched with sodium sulfite solution and extracted with ethyl acetate. The organic layer was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 1/0 to 10/1) to give 4-fluoro-2-iodo-5-methylphenol (7.65 g, yield: 75.6%) as a yellow oil.

步骤12b:4-氟-5-甲基-2-((三甲基甲硅烷基)乙炔基)苯酚的制备:氮气保护,往4-氟-2-碘-5-甲基苯酚(4.19克,16.7毫摩尔,1.0当量),双三苯基磷二氯化钯(292毫克,0.42毫摩尔,0.025当量)和碘化亚铜(79.3毫克,0.42毫摩尔,0.025当量)的三乙胺/四氢呋喃(3.6毫升/5毫升)混合物中加乙炔三甲基硅烷(8.2克,83毫摩尔,5.0当量),混合物在90℃下搅拌四小时。反应液冷却至室温,加水稀释,并用乙酸乙酯萃取。有机相用饱和食盐水洗涤,并用无水硫酸钠干燥,减压浓缩得黄色油状物4-氟-5-甲基-2-((三甲基甲硅烷基)乙炔基)苯酚(粗品)。Step 12b: Preparation of 4-fluoro-5-methyl-2-((trimethylsilyl)ethynyl)phenol: To a mixture of 4-fluoro-2-iodo-5-methylphenol (4.19 g, 16.7 mmol, 1.0 eq), bistriphenylphosphine palladium dichloride (292 mg, 0.42 mmol, 0.025 eq) and cuprous iodide (79.3 mg, 0.42 mmol, 0.025 eq) in triethylamine/tetrahydrofuran (3.6 ml/5 ml) was added ethynyltrimethylsilane (8.2 g, 83 mmol, 5.0 eq) under nitrogen, and the mixture was stirred at 90° C. for four hours. The reaction solution was cooled to room temperature, diluted with water, and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give 4-fluoro-5-methyl-2-((trimethylsilyl)ethynyl)phenol (crude product) as a yellow oil.

步骤12c:5-氟-6-甲基苯并呋喃的制备:将4-氟-5-甲基-2-((三甲基甲硅烷基)乙炔基)苯酚(粗品),碘化亚铜(3.59克,18.9毫摩尔,1.0当量)和N,N-二异丙基乙胺(4.9克,37.7毫摩尔,2.0当量)的甲醇(40毫升)溶液加热至60℃并搅拌3小时。冷却至室温后,反应加水稀释并用乙酸乙酯萃取,有机层用饱和食盐水洗涤,无水硫酸钠干燥并减压浓缩。残留物用四氢呋喃溶解,向溶液中加入四丁基氟化铵三水合物(6克,20.8摩尔,1.1当量),混合物在室温下搅拌1小时。反应液加水稀释并用乙酸乙酯萃取,有机层用饱和食盐水洗涤,无水硫酸钠干燥并减压浓缩。残留物用硅胶柱层析(洗脱剂:石油醚)纯化得到5-氟-6-甲基苯并呋喃(1.5克,收率:60%)为黄色油状物。Step 12c: Preparation of 5-fluoro-6-methylbenzofuran: A solution of 4-fluoro-5-methyl-2-((trimethylsilyl)ethynyl)phenol (crude), cuprous iodide (3.59 g, 18.9 mmol, 1.0 eq.) and N,N-diisopropylethylamine (4.9 g, 37.7 mmol, 2.0 eq.) in methanol (40 ml) was heated to 60°C and stirred for 3 hours. After cooling to room temperature, the reaction was diluted with water and extracted with ethyl acetate, and the organic layer was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was dissolved in tetrahydrofuran, and tetrabutylammonium fluoride trihydrate (6 g, 20.8 mol, 1.1 eq.) was added to the solution, and the mixture was stirred at room temperature for 1 hour. The reaction solution was diluted with water and extracted with ethyl acetate, and the organic layer was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether) to give 5-fluoro-6-methylbenzofuran (1.5 g, yield: 60%) as a yellow oil.

步骤12d:6-(溴甲基)-5-氟苯并呋喃的制备:氮气保护下,将5-氟-6-甲基苯并呋喃(1.5克, 9.99毫摩尔,1.0当量),偶氮二异丁腈(328.3毫克,1.99毫摩尔,0.2当量)和N-溴代琥珀酰亚胺(2.13克,11.99毫摩尔,1.2当量)的二氯乙烷(15毫升)混合物在72℃下搅拌过夜。反应液冷却至室温,加亚硫酸钠溶液淬灭并用乙酸乙酯萃取。有机相用无水硫酸钠干燥并减压浓缩。残留物用硅胶柱层析(洗脱剂:石油醚)得黄色油状化合物6-(溴甲基)-5-氟苯并呋喃(1.37克,产率59.5%)。Step 12d: Preparation of 6-(bromomethyl)-5-fluorobenzofuran: Under nitrogen protection, 5-fluoro-6-methylbenzofuran (1.5 g, A mixture of 1,4-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (3,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (2,6-dichloro-2-nitropropane (1,6-dichloro-2-nitropropane (

步骤12e:2-(5-氟苯并呋喃-6-基)乙腈的制备:氮气保护下,6-(溴甲基)-5-氟苯并呋喃(2克,8.7毫摩尔,1.0当量)、三甲基腈硅烷(1.7克,17.4毫摩尔,2.0当量)、四丁基氟化铵的四氢呋喃溶液(17毫升,17.4毫摩尔,2.0当量)和碳酸钾(1.1克,8.7毫摩尔,1.0当量)的四氢呋喃(20毫升)混合物在60℃搅拌过夜。将反应液加水稀释并用乙酸乙酯萃取,有机相用无水硫酸钠干燥并减压浓缩得到黄色油状物2-(5-氟苯并呋喃-6-基)乙腈(粗品)。Step 12e: Preparation of 2-(5-fluorobenzofuran-6-yl)acetonitrile: Under nitrogen protection, a mixture of 6-(bromomethyl)-5-fluorobenzofuran (2 g, 8.7 mmol, 1.0 eq), trimethylsilyl nitrile (1.7 g, 17.4 mmol, 2.0 eq), tetrabutylammonium fluoride in tetrahydrofuran (17 ml, 17.4 mmol, 2.0 eq) and potassium carbonate (1.1 g, 8.7 mmol, 1.0 eq) in tetrahydrofuran (20 ml) was stirred at 60° C. overnight. The reaction solution was diluted with water and extracted with ethyl acetate, and the organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain 2-(5-fluorobenzofuran-6-yl)acetonitrile (crude product) as a yellow oil.

步骤12f:2-(5-氟苯并呋喃-6-基)乙酸的制备:2-(5-氟苯并呋喃-6-基)乙腈(粗品)和氢氧化钾(2.4克,43.5毫摩尔,5当量)的乙醇/水=1/1(5毫升/5毫升)混合物在90℃搅拌过夜。反应液冷却至室温,加水稀释并用乙酸乙酯萃取,水相用稀硫酸溶液调节pH至2~3并用乙酸乙酯萃取,有机相用无水硫酸钠干燥并减压浓缩,残留物用硅胶柱层析(洗脱剂:乙酸乙酯)得到黄色固体2-(5-氟苯并呋喃-6-基)乙酸(210毫克,产率21%)。MS(ES+):m/z=193(M+H)+.Step 12f: Preparation of 2-(5-fluorobenzofuran-6-yl)acetic acid: A mixture of 2-(5-fluorobenzofuran-6-yl)acetonitrile (crude product) and potassium hydroxide (2.4 g, 43.5 mmol, 5 equivalents) in ethanol/water = 1/1 (5 ml/5 ml) was stirred at 90°C overnight. The reaction solution was cooled to room temperature, diluted with water and extracted with ethyl acetate. The aqueous phase was adjusted to pH 2-3 with dilute sulfuric acid solution and extracted with ethyl acetate. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was chromatographed on a silica gel column (eluent: ethyl acetate) to obtain 2-(5-fluorobenzofuran-6-yl)acetic acid (210 mg, yield 21%) as a yellow solid. MS (ES+): m/z = 193 (M+H)+.

步骤12g:2-(5-氟苯并呋喃-6-基)丙酸的制备:氮气保护下,-70℃下,往2-(5-氟苯并呋喃-6-基)乙酸(210毫克,1.03毫摩尔,1.0当量)的四氢呋喃(5毫升)溶液中加入正丁基锂(1.38毫升,2.06毫摩尔,2.0当量),混合物在-70℃下搅拌30分钟。然后加入二异丙基胺(21.9毫克,0.22毫摩尔,0.2当量),搅拌15分钟。然后在-30℃下加入碘甲烷(307毫克,2.2毫摩尔,2.0当量),混合物在室温下搅拌10分钟。反应液加水稀释并用乙酸乙酯萃取,有机相用无水硫酸钠干燥并减压浓缩得到黄色油状物2-(5-氟苯并呋喃-6-基)丙酸(20毫克,产率8.8%)。MS(ES+):m/z=209(M+H)+.Step 12g: Preparation of 2-(5-fluorobenzofuran-6-yl)propanoic acid: Under nitrogen, n-butyl lithium (1.38 ml, 2.06 mmol, 2.0 eq) was added to a solution of 2-(5-fluorobenzofuran-6-yl)acetic acid (210 mg, 1.03 mmol, 1.0 eq) in tetrahydrofuran (5 ml) at -70°C, and the mixture was stirred at -70°C for 30 minutes. Diisopropylamine (21.9 mg, 0.22 mmol, 0.2 eq) was then added and stirred for 15 minutes. Methyl iodide (307 mg, 2.2 mmol, 2.0 eq) was then added at -30°C, and the mixture was stirred at room temperature for 10 minutes. The reaction solution was diluted with water and extracted with ethyl acetate. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain a yellow oily substance, 2-(5-fluorobenzofuran-6-yl)propanoic acid (20 mg, yield 8.8%). MS (ES+): m/z=209 (M+H)+.

步骤12h:2-(5-氟苯并呋喃-6-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物14)的制备:往2-(5-氟苯并呋喃-6-基)丙酸(20毫克,0.096毫摩尔,1.0当量),7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-1)(34毫克,0.096毫摩尔,1.0单量)和N,N-二异丙基乙胺(50毫克,0.38毫摩尔,4.0当量)的N,N-二甲基甲酰胺(5毫升)混合物中加入2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(47.5毫克,0.12毫摩尔,1.3当量)。混合物在室温下搅拌两小时。反应液加水稀释并用乙酸乙酯萃取,有机相用无水硫酸钠干燥并减压浓缩,残留物用厚柱层析(洗脱剂为:石油醚/乙酸乙酯/氨水=1/100/1)得到黄色固体2-(5-氟苯并呋喃-6-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(22毫克,产率47.2%)。MS(ES+): m/z=486(M+H)+.1H NMR(500MHz,MeOD)δ8.80(s,2H),7.79(t,J=22.6Hz,2H),7.36(dd,J=38.6,33.6Hz,3H),6.79(d,J=44.9Hz,1H),4.31(d,J=53.9Hz,1H),3.69–3.43(m,4H),2.81(d,J=3.9Hz,2H),2.61–2.43(m,3H),1.99(dd,J=60.7,30.0Hz,4H),1.47(d,J=5.6Hz,3H).Step 12h: Preparation of 2-(5-fluorobenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 14): To 2-(5-fluorobenzofuran-6-yl)propanoic acid (20 mg, 0.096 mmol, 1.0 equiv), 7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro 2-(7-azabenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (47.5 mg, 0.12 mmol, 1.3 eq.) was added to a mixture of -1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-1) (34 mg, 0.096 mmol, 1.0 eq.) and N,N-diisopropylethylamine (50 mg, 0.38 mmol, 4.0 eq.) in N,N-dimethylformamide (5 ml). The mixture was stirred at room temperature for two hours. The reaction solution was diluted with water and extracted with ethyl acetate. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was chromatographed on a thick column (eluent: petroleum ether/ethyl acetate/ammonia water = 1/100/1) to give a yellow solid 2-(5-fluorobenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (22 mg, yield 47.2%). MS (ES+): m/z=486(M+H)+.1H NMR(500MHz,MeOD)δ8.80(s,2H),7.79(t,J=22.6Hz,2H),7.36(dd,J=38.6,33.6Hz,3H),6.79(d,J=44.9Hz,1H),4.31(d,J=53 .9Hz,1H),3.69–3.43(m,4H),2.81(d,J=3.9Hz,2H),2.61–2.43(m,3H),1.99(dd,J=60.7,30.0Hz,4H),1.47(d,J=5.6Hz,3H).

实施例13:6-甲氧基-4-(1-(S)-7'-甲基-6'(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-1-硫代丙烷-2-基)烟腈(化合物11)的制备(按照方案四和五线路制备)Example 13: Preparation of 6-methoxy-4-(1-(S)-7'-methyl-6'(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-1-thiopropane-2-yl)nicotinonitrile (Compound 11) (Prepared according to Schemes 4 and 5)

步骤13a:2-(5-溴-2-甲氧基吡啶-4-基)乙酸甲酯(化合物0409-11)的制备:在氮气保护下,将二异丙基氨基锂的四氢呋喃溶液(0.9摩尔每升,67毫升,60.00毫摩尔,3.0当量)滴加到-30℃的5-溴-2-甲氧基-4-甲基吡啶(0408-11)(4.04克,20.00毫摩尔,1.0当量)的四氢呋喃(40毫升)溶液中,将反应混合物在-30℃下搅拌1小时后,滴加加入碳酸二甲酯(5.40克,60.00毫摩尔,3.0当量),在加入结束时,将反应混合物升温至室温并搅拌1小时。反应加0.5摩尔每升的盐酸溶液(120毫升,60.00毫摩尔)淬灭,并用乙酸乙酯萃取。有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱(洗脱剂:石油醚至石油醚/乙酸乙酯=20/1)纯化,得到黄色油状产物2-(5-溴-2-甲氧基吡啶-4-基)乙酸甲酯(3.84克,收率:73.82%)。LCMS(ESI):m/z=260(M+H)+Step 13a: Preparation of methyl 2-(5-bromo-2-methoxypyridin-4-yl)acetate (Compound 0409-11): Under nitrogen, lithium diisopropylamide in tetrahydrofuran (0.9 mol, 67 ml, 60.00 mmol, 3.0 eq) was added dropwise to a solution of 5-bromo-2-methoxy-4-methylpyridine (0408-11) (4.04 g, 20.00 mmol, 1.0 eq) in tetrahydrofuran (40 ml) at -30°C. After the reaction mixture was stirred at -30°C for 1 hour, dimethyl carbonate (5.40 g, 60.00 mmol, 3.0 eq) was added dropwise. At the end of the addition, the reaction mixture was warmed to room temperature and stirred for 1 hour. The reaction was quenched with 0.5 mol hydrochloric acid solution (120 ml, 60.00 mmol) and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether to petroleum ether/ethyl acetate = 20/1) to obtain a yellow oily product, methyl 2-(5-bromo-2-methoxypyridin-4-yl)acetate (3.84 g, yield: 73.82%). LCMS (ESI): m/z = 260 (M+H) + .

步骤13b:2-(5-氰基-2-甲氧基吡啶-4-基)丙酸甲酯(化合物0410-11)的制备:在氮气保护下,在-70℃下的2-(5-溴-2-甲氧基吡啶-4-基)乙酸甲酯(0409-11)(3.71克,14.27毫摩尔,1.0当量)四氢呋喃(60毫升)溶液加入双三甲基硅基胺基锂(1摩尔每升,16毫升,15.98毫摩尔,1.12eq)并搅拌1小时。将碘甲烷(2.23克,15.70毫摩尔,1.1当量)的四氢呋喃(5毫升)溶液滴加到反应混合物中,在-70℃下搅拌2小时。反应用饱和氯化铵溶液淬灭,用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱(洗脱剂:石油醚至石油醚/乙酸乙酯=35/1)纯化,得到黄色油状产物2-(5-溴-2-甲氧基吡啶-4-基)丙酸甲酯(3.38克,收率:86.45%)。LCMS(ESI):m/z=274(M+H)+。将2-(5-溴-2-甲氧基吡啶-4-基)丙酸甲酯(1.37克,5.00毫摩尔,1.0当量)和氰化亚铜(2.68克,30.00毫摩尔,6.0当量)的N-甲基吡咯烷酮(15毫升)混合物在180℃下搅拌1.5小时。冷却至室温后,用水淬灭反应,用乙酸乙酯萃取。有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱(洗脱剂:石油醚/乙酸乙酯=30/1至10/1)纯化,得黄色油状产物2-(5-氰基-2-甲氧基吡啶-4-基)丙酸甲酯(434毫克,收率:39.45%)。LCMS(ESI):m/z=221(M+H)+Step 13b: Preparation of methyl 2-(5-cyano-2-methoxypyridin-4-yl)propanoate (compound 0410-11): Under nitrogen, a solution of methyl 2-(5-bromo-2-methoxypyridin-4-yl)acetate (0409-11) (3.71 g, 14.27 mmol, 1.0 eq) in tetrahydrofuran (60 ml) at -70°C was added with lithium bis(trimethylsilyl)amide (1 mol per liter, 16 ml, 15.98 mmol, 1.12 eq) and stirred for 1 hour. A solution of iodomethane (2.23 g, 15.70 mmol, 1.1 eq) in tetrahydrofuran (5 ml) was added dropwise to the reaction mixture and stirred at -70°C for 2 hours. The reaction was quenched with saturated ammonium chloride solution, extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether to petroleum ether/ethyl acetate = 35/1) to give a yellow oily product, methyl 2-(5-bromo-2-methoxypyridin-4-yl)propanoate (3.38 g, yield: 86.45%). LCMS (ESI): m/z = 274 (M+H) + . A mixture of methyl 2-(5-bromo-2-methoxypyridin-4-yl)propanoate (1.37 g, 5.00 mmol, 1.0 eq.) and cuprous cyanide (2.68 g, 30.00 mmol, 6.0 eq.) in N-methylpyrrolidone (15 ml) was stirred at 180° C. for 1.5 hours. After cooling to room temperature, the reaction was quenched with water and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 30/1 to 10/1) to give yellow oily product methyl 2-(5-cyano-2-methoxypyridin-4-yl)propanoate (434 mg, yield: 39.45%). LCMS (ESI): m/z = 221 (M+H) + .

步骤13c:2-(5-氰基-2-甲氧基吡啶-4-基)丙酸(化合物0405-11)的制备:在氮气保护下,将2-(5-氰基-2-甲氧基吡啶-4-基)丙酸甲酯(0410-11)(434毫克,1.97毫摩尔,1.0当量)和三甲基硅醇钾(380毫克,2.96毫摩尔,1.5当量)的四氢呋喃(5毫升)混合物在室温下搅拌1小时。 反应混合物用一摩尔每升的盐酸溶液淬灭并用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱纯化(洗脱剂:石油醚/乙酸乙酯=5/1至乙酸乙酯),得到黄色固体产物2-(5-氰基-2-甲氧基吡啶-4-基)丙酸(269毫克,收率:66.42%)。LCMS(ESI):m/z=207(M+H)+Step 13c: Preparation of 2-(5-cyano-2-methoxypyridin-4-yl)propanoic acid (compound 0405-11): Under nitrogen protection, a mixture of methyl 2-(5-cyano-2-methoxypyridin-4-yl)propanoate (0410-11) (434 mg, 1.97 mmol, 1.0 equiv) and potassium trimethylsilanol (380 mg, 2.96 mmol, 1.5 equiv) in tetrahydrofuran (5 ml) was stirred at room temperature for 1 hour. The reaction mixture was quenched with a 1 mol/L hydrochloric acid solution and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 5/1 to ethyl acetate) to obtain a yellow solid product 2-(5-cyano-2-methoxypyridin-4-yl)propanoic acid (269 mg, yield: 66.42%). LCMS (ESI): m/z = 207 (M+H) + .

步骤13d:6-甲氧基-4-(1-(S)-7'-甲基-6'(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-1-氧代丙烷-2-基)烟腈(化合物0412-11)的制备:将2-(5-氰基-2-甲氧基吡啶-4-基)丙酸(0405-11)(20.6毫克,0.10毫摩尔,1.0当量),(S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-1)(35毫克,0.10毫摩尔,1.0当量),N,N-二异丙基乙胺(77毫克,0.60毫摩尔,6.0当量)和2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(40毫克,0.105毫摩尔,1.05当量)的二氯甲烷(2毫升)混合物搅拌30分钟。混合物用水稀释并用二氯甲烷萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物经厚制备薄层色谱(洗脱剂:二氯甲烷/甲醇=20/1)纯化,得到白色固体产物6-甲氧基-4-(1-(S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-1-氧代丙烷-2-基)烟腈(45毫克,收率:95.74%)。LCMS(ESI):m/z=470(M+H)+Step 13d: Preparation of 6-methoxy-4-(1-(S)-7'-methyl-6'(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)-1-oxopropan-2-yl)nicotinonitrile (Compound 0412-11): 2-(5-cyano-2-methoxypyridin-4-yl)propanoic acid (0405-11) (20.6 mg, 0.10 mmol, 1.0 equiv), (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)-1-oxopropan-2-yl)nicotinonitrile (Compound 0412-11) A mixture of 2-(7-azabenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (40 mg, 0.105 mmol, 1.05 eq) in dichloromethane (2 ml) was stirred for 30 minutes. The mixture was diluted with water and extracted with dichloromethane, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (eluent: dichloromethane/methanol=20/1) to give a white solid product, 6-methoxy-4-(1-(S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridin]-1-yl)-1-oxopropan-2-yl)nicotinonitrile (45 mg, yield: 95.74%). LCMS (ESI): m/z=470 (M+H) + .

步骤13e:6-甲氧基-4-(1-(S)-7'-甲基-6'(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-1-硫代丙烷-2-基)烟腈(化合物11)的制备:将6-甲氧基-4-(1-(S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-1-氧代丙烷-2-基)烟腈(0412-11)(19.7毫克,0.042毫摩尔,1.0当量)和劳森试剂(20.4毫克,0.05毫摩尔,1.2当量)的甲苯(1.5毫升)混合物在微波反应器125℃下搅拌40分钟。将混合物用水稀释并用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物经厚制备薄层色谱(洗脱剂:二氯甲烷/甲醇=20/1)纯化,得到黄色固体产物6-甲氧基-4-(1-(S)-7'-甲基-6'(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-1-硫代丙烷-2-基)烟腈(13毫克,收率:63.73%)。LCMS(ESI):m/z=486(M+H)+1H NMR(500MHz,MeOD)δ8.72(t,J=5.4Hz,2H),8.32(dd,J=36.1,30.8Hz,1H),7.78(d,J=6.3Hz,1H),7.22(q,J=5.0Hz,1H),6.89(dd,J=26.3,11.2Hz,1H),4.49(dd,J=28.2,6.7Hz,1H),4.09–3.74(m,7H),2.85–2.69(m,2H),2.51–2.42(m,3H),2.31–2.00(m,2H),1.87(ddd,J=20.0,14.3,7.4Hz,2H),1.58–1.46(m,3H).Step 13e: Preparation of 6-methoxy-4-(1-(S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)-1-thiopropane-2-yl)nicotinonitrile (Compound 11): 6-methoxy-4-(1-(S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)-1-thiopropane-2-yl)nicotinonitrile (Compound 11) A mixture of 4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridin]-1-yl)-1-oxopropan-2-yl)nicotinonitrile (0412-11) (19.7 mg, 0.042 mmol, 1.0 eq) and Lawesson's reagent (20.4 mg, 0.05 mmol, 1.2 eq) in toluene (1.5 ml) was stirred in a microwave reactor at 125° C. for 40 minutes. The mixture was diluted with water and extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (eluent: dichloromethane/methanol=20/1) to give a yellow solid product, 6-methoxy-4-(1-(S)-7'-methyl-6'(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridin]-1-yl)-1-thiopropane-2-yl)nicotinonitrile (13 mg, yield: 63.73%). LCMS (ESI): m/z=486 (M+H) + . 1H NMR(500MHz,MeOD)δ8.72(t,J=5.4Hz,2H),8.32(dd,J=36.1,30.8Hz,1H),7.7 8(d,J=6.3Hz,1H),7.22(q,J=5.0Hz,1H),6.89(dd,J=26.3,11.2Hz,1H),4.49( dd,J=28.2,6.7Hz,1H),4.09–3.74(m,7H),2.85–2.69(m,2H),2.51–2.42(m,3 H),2.31–2.00(m,2H),1.87(ddd,J=20.0,14.3,7.4Hz,2H),1.58–1.46(m,3H).

实施例14:(S)-N-(3,4-二氟苯基)-N,7’-二甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-硫代甲酰胺(化合物4)的制备(按照方案三线路制备):Example 14: Preparation of (S)-N-(3,4-difluorophenyl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-thiocarboxamide (Compound 4) (prepared according to Scheme 3):

在氮气保护下,向3,4-二氟-N-甲基苯胺(0204-3)(14毫克,0.10毫摩尔,1.0当量),三乙胺(60.6毫克,0.60毫摩尔,6.0当量)的二氯甲烷(3毫升)混合物中滴加二氯硫化碳(11.5毫克,0.10毫摩尔,1.0当量),并将混合物在室温下搅拌一小时。混合物用1M的盐酸溶液淬灭, 用水稀释,并用二氯甲烷萃取。有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物(粗品)无需进一步纯化直接用于下一步。Under nitrogen protection, to a mixture of 3,4-difluoro-N-methylaniline (0204-3) (14 mg, 0.10 mmol, 1.0 eq.), triethylamine (60.6 mg, 0.60 mmol, 6.0 eq.) in dichloromethane (3 ml) was added dropwise carbon dichloride (11.5 mg, 0.10 mmol, 1.0 eq.), and the mixture was stirred at room temperature for one hour. The mixture was quenched with 1M hydrochloric acid solution, Dilute with water and extract with dichloromethane. Wash the organic phase with saturated brine, dry over anhydrous sodium sulfate and concentrate. The residue (crude product) is used directly in the next step without further purification.

在氮气保护下,向(S)-7'-甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶](0122-1)(31.7毫克,0.09毫摩尔,0.9当量)和三乙胺(60.6毫克,0.60毫摩尔,6.0当量)的二氯甲烷(2毫升)混合物中加入(3,4-二氟苯基)(甲基)硫代氨基酰氯(粗品),将混合物在室温下搅拌一小时。混合物用水稀释并用二氯甲烷萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物经过厚制备薄层色谱纯化(洗脱液:二氯甲烷/甲醇/氨水=80/3.5/0.8),得到黄色固体产物(S)-N-(3,4-二氟苯基)-N,7’-二甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-硫代甲酰胺(30毫克,收率:64.38%)。LCMS(ESI):467[M+H]+1H NMR(500MHz,MeOD)δ8.79(t,J=5.3Hz,2H),7.87–7.52(m,3H),7.50–7.36(m,1H),7.31–7.09(m,2H),6.77(dd,J=45.1,2.5Hz,1H),4.14–3.94(m,1H),3.93–3.37(m,4H),2.90–2.67(m,2H),2.53(dd,J=34.4,13.4Hz,4H),2.21–1.65(m,4H),1.50–1.40(m,3H).Under nitrogen protection, to a mixture of (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine](0122-1) (31.7 mg, 0.09 mmol, 0.9 eq) and triethylamine (60.6 mg, 0.60 mmol, 6.0 eq) in dichloromethane (2 ml) was added (3,4-difluorophenyl)(methyl)aminothioyl chloride (crude), and the mixture was stirred at room temperature for one hour. The mixture was diluted with water and extracted with dichloromethane, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by thick preparative thin layer chromatography (eluent: dichloromethane/methanol/aqueous ammonia = 80/3.5/0.8) to give a yellow solid product (S)-N-(3,4-difluorophenyl)-N,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carbothioamide (30 mg, yield: 64.38%). LCMS (ESI): 467 [M+H] + . 1H NMR(500MHz,MeOD)δ8.79(t,J=5.3Hz,2H),7.87–7.52(m,3H),7.50–7.36(m,1H),7.31–7.09(m,2H),6.77(dd,J=45.1,2.5Hz,1H ),4.14–3.94(m,1H),3.93–3.37(m,4H),2.90–2.67(m,2H),2.53(dd,J=34.4,13.4Hz,4H),2.21–1.65(m,4H),1.50–1.40(m,3H).

实施例15:2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮-2-氘(化合物67)的制备(按照方案四和五线路制备)Example 15: Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one-2-deuterium (Compound 67) (Prepared according to Schemes 4 and 5)

步骤15a:2-(5-氟-2-甲氧基吡啶-4-基)丙酸甲酯(化合物0410-67)的制备
Step 15a: Preparation of methyl 2-(5-fluoro-2-methoxypyridin-4-yl)propanoate (Compound 0410-67)

氮气保护下,在-78℃下,向2-(5-氟-2-甲氧基吡啶-4-基)丙酸(0405-7)(200毫克,1.0毫摩尔,1.0当量),N,N-二甲基甲酰胺(两滴)的二氯甲烷(5毫升)溶液中滴加草酰氯(1.5毫升,1M二氯甲烷溶液,1.5毫摩尔,1.5当量),混合物在-78℃下搅拌1.0小时。然后向反应液滴加甲醇(0.5毫升),并使混合物室温搅拌1小时。混合物用水和饱和食盐水洗。有机相用无水硫酸钠干燥并浓缩。残余物经硅胶柱层析(洗脱剂:石油醚/乙酸乙酯=4/1至1/2)纯化,得到黄色油状物2-(5-氟-2-甲氧基吡啶-4-基)丙酸甲酯(177毫克,收率:82.7%)。Under nitrogen protection, oxalyl chloride (1.5 ml, 1M dichloromethane solution, 1.5 mmol, 1.5 eq) was added dropwise to a solution of 2-(5-fluoro-2-methoxypyridin-4-yl)propionic acid (0405-7) (200 mg, 1.0 mmol, 1.0 eq) and N,N-dimethylformamide (two drops) in dichloromethane (5 ml) at -78°C, and the mixture was stirred at -78°C for 1.0 hour. Methanol (0.5 ml) was then added dropwise to the reaction solution, and the mixture was stirred at room temperature for 1 hour. The mixture was washed with water and saturated brine. The organic phase was dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 4/1 to 1/2) to give methyl 2-(5-fluoro-2-methoxypyridin-4-yl)propanoate (177 mg, yield: 82.7%) as a yellow oil.

步骤15b:2-氘-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(化合物0405-67)的制备:氮气保护下,在-78℃下,向2-(5-氟-2-甲氧基吡啶-4-基)丙酸甲酯(0410-67)(50毫克,0.23毫摩尔,1.0当量)的四氢呋喃(5毫升)溶液中滴加双三甲基硅基胺基锂(0.35毫升,1M四氢呋喃溶液,0.35毫摩尔,1.4当量),混合物在-78℃下搅拌1.0小时。然后向反应液滴加重水(0.2毫升),并使混合物室温搅拌过夜。混合物用甲基叔丁基醚稀释,水洗。水相用1M盐酸酸化,乙酸乙酯萃取,水和饱和食盐水洗。有机相用无水硫酸钠干燥并浓缩得到黄色油状物2-氘-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(50毫克,粗品)。 Step 15b: Preparation of 2-deuterium-2-(5-fluoro-2-methoxypyridin-4-yl)propionic acid (Compound 0405-67): Under nitrogen protection, lithium bis(trimethylsilyl)amide (0.35 ml, 1M tetrahydrofuran solution, 0.35 mmol, 1.4 eq.) was added dropwise to a solution of methyl 2-(5-fluoro-2-methoxypyridin-4-yl)propanoate (0410-67) (50 mg, 0.23 mmol, 1.0 eq.) in tetrahydrofuran (5 ml) at -78°C, and the mixture was stirred at -78°C for 1.0 hour. Heavy water (0.2 ml) was then added dropwise to the reaction solution, and the mixture was stirred at room temperature overnight. The mixture was diluted with methyl tert-butyl ether and washed with water. The aqueous phase was acidified with 1M hydrochloric acid, extracted with ethyl acetate, and washed with water and saturated brine. The organic phase was dried over anhydrous sodium sulfate and concentrated to give 2-deuterio-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (50 mg, crude) as a yellow oil.

步骤15c:2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮-2-氘(化合物67)的制备:氮气保护下,在0℃下,向(S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶](0122-1)(50毫克,0.14毫摩尔,1.0当量),2-氘-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(0405-67)(23毫克,0.14毫摩尔,1.00当量)和N,N-二异丙基乙胺(55毫克,0.42毫摩尔,1.05当量)的乙腈(5毫升)溶液中加入N'N-羰基二咪唑(25毫克,0.15毫摩尔,1.1当量)。混合物在室温搅拌过夜,然后用乙酸乙酯稀释,用水和饱和食盐水洗。有机相用无水硫酸钠干燥并浓缩。残余物经薄层制备色谱(洗脱剂:石油醚/乙酸乙酯=1/1)纯化,得到白色固体产物2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮-2-氘(47.6毫克,收率:72.6%),LCMS(ESI):m/z=454[M+1]+1H NMR(500MHz,MeOD)δ8.81(t,J=3.8Hz,2H),7.96(t,J=20.7Hz,1H),7.84(d,J=14.8Hz,1H),7.35–7.21(m,1H),6.83–6.67(m,1H),3.87(dd,J=10.5,4.2Hz,3H),3.76–3.34(m,4H),2.92–2.72(m,2H),2.57(d,J=12.7Hz,3H),2.19–1.74(m,4H),1.44(dt,J=12.5,6.4Hz,3H).Step 15c: Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one-2-deuterium (Compound 67): Under nitrogen protection, at 0°C, add (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl [1,8]naphthyridine] (0122-1) (50 mg, 0.14 mmol, 1.0 eq), 2-deuterium-2-(5-fluoro-2-methoxypyridin-4-yl) propionic acid (0405-67) (23 mg, 0.14 mmol, 1.00 eq) and N,N-diisopropylethylamine (55 mg, 0.42 mmol, 1.05 eq) in acetonitrile (5 ml) were added N'N-carbonyldiimidazole (25 mg, 0.15 mmol, 1.1 eq). The mixture was stirred at room temperature overnight, then diluted with ethyl acetate, washed with water and saturated brine. The organic phase was dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 1/1) to give a white solid product, 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one-2-deuterium (47.6 mg, yield: 72.6%), LCMS (ESI): m/z = 454 [M+1] + . 1H NMR (500MHz, MeOD) δ8.81(t,J=3.8Hz,2H),7.96(t,J=20.7Hz,1H),7.84(d,J=14.8Hz,1H),7.35–7.21(m,1H),6.83–6.67(m,1H),3.87( dd,J=10.5,4.2Hz,3H),3.76–3.34(m,4H),2.92–2.72(m,2H),2.57(d,J=12.7Hz,3H),2.19–1.74(m,4H),1.44(dt,J=12.5,6.4Hz,3H).

实施例16:2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-硫酮-2-氘(化合物75)的制备(按照方案五线路制备)Example 16: Preparation of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione-2-deuterium (Compound 75) (Prepared according to Scheme 5)

氮气保护下,向2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮-2-氘(化合物67)(45毫克,0.10毫摩尔,1.0当量)的甲苯(2毫升)溶液中加入劳森试剂(47毫克,0.12毫摩尔,1.2当量),混合物加热至100℃反应过夜。混合物用乙酸乙酯稀释,用水和饱和食盐水洗。有机相用无水硫酸钠干燥并浓缩。残余物经薄层制备色谱(洗脱剂:石油醚/乙酸乙酯=1/1)纯化,得到白色固体产物2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-硫酮-2-氘(26.0毫克,收率:55.7%),LCMS(ESI):m/z=481[M+1]+1HNMR(500MHz,MeOD)δ8.81(t,J=4.6Hz,2H),7.98–7.74(m,2H),7.30(t,J=3.9Hz,1H),6.97–6.80(m,1H),4.21–3.95(m,2H),3.95–3.63(m,5H),2.95–2.67(m,2H),2.57(dd,J=10.2,3.4Hz,3H),2.34–2.09(m,2H),2.08–1.85(m,2H),1.61–1.51(m,3H).Under nitrogen protection, Lawesson's reagent (47 mg, 0.12 mmol, 1.2 eq.) was added to a toluene (2 ml) solution of 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one-2-deuterium (Compound 67) (45 mg, 0.10 mmol, 1.0 eq.), and the mixture was heated to 100°C for overnight reaction. The mixture was diluted with ethyl acetate and washed with water and saturated brine. The organic phase was dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (eluent: petroleum ether/ethyl acetate = 1/1) to give a white solid product, 2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione-2-deuterium (26.0 mg, yield: 55.7%), LCMS (ESI): m/z = 481 [M+1] + . 1 HNMR(500MHz,MeOD)δ8.81(t,J=4.6Hz,2H),7.98–7.74(m,2H),7.30(t,J=3.9Hz,1H),6.97–6.80(m,1H),4.21–3.95(m,2H), 3.95–3.63(m,5H),2.95–2.67(m,2H),2.57(dd,J=10.2,3.4Hz,3H),2.34–2.09(m,2H),2.08–1.85(m,2H),1.61–1.51(m,3H).

实施例17:(2R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-(7'-(甲基-d3)-6'-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物59)的制备(按照方案一和四线路制备)Example 17: Preparation of (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(7'-(methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 59) (Prepared according to Schemes 1 and 4)

步骤17a:2-氯-6-(甲基-d3)吡啶-3-胺2-氯-6-(甲基-d3)吡啶-3-胺(化合物0106-59)的制备:
Step 17a: Preparation of 2-chloro-6-(methyl-d3)pyridin-3-amine 2-chloro-6-(methyl-d3)pyridin-3-amine (Compound 0106-59):

在氮气保护,0℃下,向6-溴-2-氯吡啶-3-胺(5.00克,24.10毫摩尔,1.0当量)的N,N-二甲基甲酰胺(25毫升)溶液中加入氢化钠(2.41克,60.25毫摩尔,2.5eq),反应液在0℃下搅拌一小时。在0℃下,往反应混合物中加入4-甲氧基氯苄(7.93克,50.61毫摩尔,2.1当量),并在室温下搅拌1小时。反应用氯化铵溶液淬灭,加水稀释,用乙酸乙酯萃取。有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱(洗脱液:石油醚/乙酸乙酯=5/1)纯化,得到黄色固体产物6-溴-2-氯-N,N-双(4-甲氧基苄基)吡啶-3-胺(11克,收率:100%)LCMS(ESI):m/z=447[M+H]+Under nitrogen protection, sodium hydride (2.41 g, 60.25 mmol, 2.5 eq) was added to a solution of 6-bromo-2-chloropyridin-3-amine (5.00 g, 24.10 mmol, 1.0 eq) in N,N-dimethylformamide (25 ml) at 0°C, and the reaction solution was stirred at 0°C for one hour. 4-Methoxybenzyl chloride (7.93 g, 50.61 mmol, 2.1 eq) was added to the reaction mixture at 0°C and stirred at room temperature for 1 hour. The reaction was quenched with ammonium chloride solution, diluted with water, and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 5/1) to give a yellow solid product 6-bromo-2-chloro-N,N-bis(4-methoxybenzyl)pyridin-3-amine (11 g, yield: 100%). LCMS (ESI): m/z = 447 [M+H] + .

在氮气保护,-70℃下,向6-溴-2-氯-N,N-双(4-甲氧基苄基)吡啶-3-胺(19.98克,44.80毫摩尔,1.0当量)的四氢呋喃(200毫升)溶液滴加叔丁基锂(1.3M,51.69毫升,67.20毫摩尔,1.5eq)并搅拌1小时。在-70℃下将干燥的碘甲烷(9.74克,67.20毫摩尔,1.5当量)的四氢呋喃(10毫升)溶液加入到反应混合物中,并在室温下搅拌过夜。反应加饱和氯化铵溶液淬灭,加水稀释,并用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱(洗脱剂:石油醚/乙酸乙酯=30/1至10/1)纯化,得到黄色油状产物2-氯-N,N-双(4-甲氧基苄基)-6-(甲基-d3)吡啶-3-胺(9.0克,收率:52.05%)。LCMS(ESI):m/z=386[M+H]+Under nitrogen protection, tert-butyl lithium (1.3M, 51.69 ml, 67.20 mmol, 1.5 eq) was added dropwise to a solution of 6-bromo-2-chloro-N,N-bis(4-methoxybenzyl)pyridin-3-amine (19.98 g, 44.80 mmol, 1.0 eq) in tetrahydrofuran (200 ml) at -70°C and stirred for 1 hour. A solution of dry iodomethane (9.74 g, 67.20 mmol, 1.5 eq) in tetrahydrofuran (10 ml) was added to the reaction mixture at -70°C and stirred overnight at room temperature. The reaction was quenched with saturated ammonium chloride solution, diluted with water, extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 30/1 to 10/1) to give a yellow oily product 2-chloro-N,N-bis(4-methoxybenzyl)-6-(methyl-d3)pyridin-3-amine (9.0 g, yield: 52.05%). LCMS (ESI): m/z = 386 [M+H] + .

向2-氯-N,N-双(4-甲氧基苄基)-6-(甲基-d3)吡啶-3-胺(9.0克,23.32毫摩尔,1.0当量)的二氯甲烷(80毫升)混合物中加入三氟乙酸(20毫升),并在室温下搅拌过夜。在真空下除去溶剂。用2M的氢氧化钠溶液调节残余物的pH至13。用二氯甲烷萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱(洗脱剂:石油醚/乙酸乙酯=5/1)纯化,得到黄色油状产物2-氯-6-(甲基-d3)吡啶-3-胺(3.38克,收率:100%)。LCMS(ESI):m/z=146[M+H]+Trifluoroacetic acid (20 ml) was added to a mixture of 2-chloro-N, N-bis(4-methoxybenzyl)-6-(methyl-d3)pyridine-3-amine (9.0 g, 23.32 mmol, 1.0 eq.) in dichloromethane (80 ml), and stirred overnight at room temperature. The solvent was removed under vacuum. The pH of the residue was adjusted to 13 with a 2M sodium hydroxide solution. The mixture was extracted with dichloromethane, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 5/1) to give a yellow oily product, 2-chloro-6-(methyl-d3)pyridine-3-amine (3.38 g, yield: 100%). LCMS (ESI): m/z = 146 [M+H] + .

步骤17b:2-氯-3-碘-6-(甲基-d3)吡啶(化合物0107-59)的制备:向2-氯-6-(甲基-d3)吡啶-3-胺(0106-59)(3.38克,23.32毫摩尔,1.0当量)的浓盐酸/水(25毫升/25毫升)溶液中加入亚硝酸钠溶液24毫升(2.41克.34.98毫摩尔,1.5当量),并在0℃下搅拌30分钟。然后向反应中加入甲基叔丁基醚(15毫升),并在0℃下滴加碘化钾溶液80毫升(7.74克,46.64毫摩尔,2.0当量)。将混合物在室温下搅拌过夜。反应用亚硫酸钠溶液淬灭。用2M的氢氧化钠溶液调节残余物的pH至7。混合物用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物经硅胶柱色谱(洗脱液:石油醚/乙酸乙酯=40/1)纯化,得到黄色固体产物2-氯-3-碘-6-(甲基-d3)吡啶(4.68克,收率:72.24%)。Step 17b: Preparation of 2-chloro-3-iodo-6-(methyl-d3)pyridine (Compound 0107-59): To a solution of 2-chloro-6-(methyl-d3)pyridin-3-amine (0106-59) (3.38 g, 23.32 mmol, 1.0 eq) in concentrated hydrochloric acid/water (25 ml/25 ml) was added 24 ml of sodium nitrite solution (2.41 g. 34.98 mmol, 1.5 eq) and stirred at 0°C for 30 minutes. Methyl tert-butyl ether (15 ml) was then added to the reaction and 80 ml of potassium iodide solution (7.74 g, 46.64 mmol, 2.0 eq) was added dropwise at 0°C. The mixture was stirred at room temperature overnight. The reaction was quenched with sodium sulfite solution. The pH of the residue was adjusted to 7 with 2M sodium hydroxide solution. The mixture was extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 40/1) to give a yellow solid product 2-chloro-3-iodo-6-(methyl-d3)pyridine (4.68 g, yield: 72.24%).

步骤17c:3-羟基-3-((三甲基硅基)乙炔基)吡咯烷-1-羧酸叔丁酯(化合物0102-59)的制备:氮气保护下,在-70℃下,向乙炔基三甲基硅烷(2.55克,25.9毫摩尔,1.2当量)的四氢呋喃(40毫升)溶液中滴加正丁基锂(9.5毫升,23.8毫摩尔,1.1当量),混合物在-70℃下搅拌1.0小时。 然后在-60℃下向反应液滴加1-叔丁氧碳基-3-吡咯烷酮(0101-59)(4.0克,21.6毫摩尔,1.0当量)的四氢呋喃(10毫升)溶液,撤去低温装置,并使混合物搅拌过夜。反应液用氯化铵溶液淬灭并用乙酸乙酯萃取,将有机相干燥并减压浓缩得到黄色油状粗产物3-羟基-3-((三甲基硅基)乙炔基)吡咯烷-1-羧酸叔丁酯(6.04克),粗产物不经过进一步纯化,直接用于下一步。Step 17c: Preparation of tert-butyl 3-hydroxy-3-((trimethylsilyl)ethynyl)pyrrolidine-1-carboxylate (Compound 0102-59): Under nitrogen protection, to a solution of ethynyltrimethylsilane (2.55 g, 25.9 mmol, 1.2 eq) in tetrahydrofuran (40 ml) was added dropwise n-butyllithium (9.5 ml, 23.8 mmol, 1.1 eq) at -70 °C, and the mixture was stirred at -70 °C for 1.0 hour. Then, a solution of 1-tert-butyloxycarbonyl-3-pyrrolidone (0101-59) (4.0 g, 21.6 mmol, 1.0 equivalent) in tetrahydrofuran (10 ml) was added dropwise to the reaction solution at -60°C, the cryogenic device was removed, and the mixture was stirred overnight. The reaction solution was quenched with ammonium chloride solution and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure to give a yellow oily crude product of tert-butyl 3-hydroxy-3-((trimethylsilyl)ethynyl)pyrrolidine-1-carboxylate (6.04 g), which was used directly in the next step without further purification.

步骤17d:3-乙炔基-3-羟基吡咯烷-1-羧酸叔丁酯(化合物0103-59)的制备:氮气保护下,向3-羟基-3-((三甲基硅基)乙炔基)吡咯烷-1-羧酸叔丁酯(0102-59)(6.00克,21.2毫摩尔,1.0当量)的甲醇(100毫升)溶液中加入碳酸钾(5.80克,42.3毫摩尔,2.0当量),混合物在室温下搅拌1.5小时。减压除去溶剂,残留物加水稀释并用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=10/1)纯化,得到黄色油状产物3-乙炔基-3-羟基吡咯烷-1-羧酸叔丁酯(4.43克,两步收率:97.1%)。Step 17d: Preparation of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (Compound 0103-59): Under nitrogen protection, potassium carbonate (5.80 g, 42.3 mmol, 2.0 eq.) was added to a solution of tert-butyl 3-hydroxy-3-((trimethylsilyl)ethynyl)pyrrolidine-1-carboxylate (0102-59) (6.00 g, 21.2 mmol, 1.0 eq.) in methanol (100 ml), and the mixture was stirred at room temperature for 1.5 hours. The solvent was removed under reduced pressure, the residue was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate=10/1) to obtain a yellow oily product, tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (4.43 g, two-step yield: 97.1%).

步骤17e:3-乙炔基-3-羟基吡咯烷-1-羧酸叔丁酯(0104-59)的制备:氮气保护下,0℃下,向3-乙炔基-3-羟基吡咯烷-1-羧酸叔丁酯(0103-59)(4.43克,21.0毫摩尔,1.0当量),N,N-二异丙基乙胺(4.06克,31.5毫摩尔,1.5当量)的二氯甲烷(100毫升)溶液中加入乙酰氯(1.98克,25.2毫摩尔,1.2当量),混合物在0℃下搅拌1小时。反应液加水淬灭,并用二氯甲烷萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=20/1至5/1)纯化,得到黄色油状产物3-乙炔基-3-羟基吡咯烷-1-羧酸叔丁酯(3.07克,收率:57.8%)。Step 17e: Preparation of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (0104-59): Under nitrogen protection, acetyl chloride (1.98 g, 25.2 mmol, 1.2 eq) was added to a solution of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (0103-59) (4.43 g, 21.0 mmol, 1.0 eq) and N,N-diisopropylethylamine (4.06 g, 31.5 mmol, 1.5 eq) in dichloromethane (100 ml) at 0° C. The mixture was stirred at 0° C. for 1 hour. The reaction solution was quenched with water and extracted with dichloromethane, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate=20/1 to 5/1) to give yellow oily product tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (3.07 g, yield: 57.8%).

步骤17f:3-乙炔基-3-((4-甲氧基苄基)氨基)吡咯烷-1-羧酸叔丁酯(化合物0105-59)的制备:氮气保护下,70℃下,向4-甲氧基苄胺(3.07克,22.3毫摩尔,2.0当量),氯化亚铜(111毫克,1.1毫摩尔,0.1当量)的四氢呋喃(30毫升)溶液中加入3-乙炔基-3-羟基吡咯烷-1-羧酸叔丁酯(0104-59)(2.83克,11.2毫摩尔,1.0当量)的四氢呋喃(10毫升)溶液,混合物在70℃下搅拌1小时。反应液加水稀释并用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=10/1至4/1)纯化,得到黄色油状产物3-乙炔基-3-((4-甲氧基苄基)氨基)吡咯烷-1-羧酸叔丁酯(2.56克,收率:69.4%)。Step 17f: Preparation of tert-butyl 3-ethynyl-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0105-59): Under nitrogen protection, a solution of tert-butyl 3-ethynyl-3-hydroxypyrrolidine-1-carboxylate (0104-59) (2.83 g, 11.2 mmol, 1.0 eq) in tetrahydrofuran (10 ml) was added to a solution of 4-methoxybenzylamine (3.07 g, 22.3 mmol, 2.0 eq) and cuprous chloride (111 mg, 1.1 mmol, 0.1 eq) in tetrahydrofuran (30 ml) at 70° C. The mixture was stirred at 70° C. for 1 hour. The reaction solution was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate=10/1 to 4/1) to give yellow oily product tert-butyl 3-ethynyl-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (2.56 g, yield: 69.4%).

步骤17g:3-((2-氯-6-(甲基-d3)吡啶-3-基)乙炔基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-羧酸叔丁酯(化合物0108-59)的制备:在氮气保护下,将2-氯-3-碘-6-(甲基-d3)吡啶(0107-59)(1.30克,5.07毫摩尔,1.0当量)、3-乙炔基-3-((4-甲氧基苄基)氨基)吡咯烷-1-羧酸叔丁酯(0105-59)(1.69克,5.07毫摩尔,1.0当量)、碘化亚铜(96毫克,0.51毫摩尔,0.1当量)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(356毫克,0.51毫摩尔,0.1当量)的混合物在90℃下搅拌4小时。将混合物用水稀释并用乙酸乙酯萃取。有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物经硅胶柱色谱(洗脱液:石油醚/乙酸乙酯=10/1至3/1)纯化,得到黄色油状产物3-((2-氯 -6-(甲基-d3)吡啶-3-基)乙炔基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-羧酸叔丁酯(1.90克,收率:81.90%)。LCMS(ESI):m/z=459[M+H]+Step 17g: Preparation of tert-butyl 3-((2-chloro-6-(methyl-d3)pyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0108-59): Under nitrogen protection, 2-chloro-3-iodo-6-(methyl-d3)pyridine (0107-59) (1.30 g, 5.07 mmol, 1.0 equiv), 3-ethynyl -3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylic acid tert-butyl ester (0105-59) (1.69 g, 5.07 mmol, 1.0 eq), cuprous iodide (96 mg, 0.51 mmol, 0.1 eq) and [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (356 mg, 0.51 mmol, 0.1 eq) were stirred at 90 ° C for 4 hours. The mixture was diluted with water and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 10/1 to 3/1) to give a yellow oily product 3-((2-chloro -6-(methyl-d3)pyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylic acid tert-butyl ester (1.90 g, yield: 81.90%). LCMS (ESI): m/z=459 [M+H] + .

步骤17h:3-(2-(2-氯-6-(甲基-d3)吡啶-3-基)乙基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-羧酸叔丁酯(化合物0109-59)的制备:在氢气氛围下,将3-((2-氯-6-(甲基-d3)吡啶-3-基)乙炔基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-羧酸叔丁酯(0108-59)(1.90克,4.126毫摩尔,1.0当量),二氧化铂(187毫克,0.825毫摩尔,0.2当量)的甲醇(20毫升)混合物在室温下搅拌过夜。将混合物通过硅藻土过滤,滤饼用甲醇洗涤,滤液浓缩。残余物经硅胶柱色谱(洗脱液:石油醚/乙酸乙酯=5/1至3/1)纯化,得到黄色油状产物3-(2-(2-氯-6-(甲基-d3)吡啶-3-基)乙基)-3-(((4-甲氧基苄基)氨基)吡咯烷-1-羧酸叔丁酯(1.30克,收率:68.42%)。LCMS(ESI):m/z=463[M+H]+Step 17h: Preparation of tert-butyl 3-(2-(2-chloro-6-(methyl-d3)pyridin-3-yl)ethyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (Compound 0109-59): A mixture of tert-butyl 3-((2-chloro-6-(methyl-d3)pyridin-3-yl)ethynyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (0108-59) (1.90 g, 4.126 mmol, 1.0 eq), platinum dioxide (187 mg, 0.825 mmol, 0.2 eq) in methanol (20 ml) was stirred at room temperature overnight under a hydrogen atmosphere. The mixture was filtered through celite, the filter cake was washed with methanol, and the filtrate was concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 5/1 to 3/1) to give yellow oily product tert-butyl 3-(2-(2-chloro-6-(methyl-d3)pyridin-3-yl)ethyl)-3-(((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (1.30 g, yield: 68.42%). LCMS (ESI): m/z = 463 [M+H] + .

步骤17i:1'-(4-甲氧基苄基)-7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(化合物0110-59)的制备:在氮气保护下,将3-(2-(2-氯-6-(甲基-d3)吡啶-3-基)乙基)-3-((4-甲氧基苄基)氨基)吡咯烷-1-羧酸叔丁酯(0109-59)(1.30克,2.81毫摩尔,1.0当量),醋酸钯(63毫克,0.28毫摩尔,0.1当量),2-二环己基磷-2',6'-二异丙氧基-1,1'-联苯(262毫克,0.56毫摩尔,0.2当量)和叔丁醇钠(540毫克,5.62毫摩尔,2.0当量)的混合物在微波仪器中搅拌反应1小时。将混合物用水稀释并用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱(洗脱液:石油醚/乙酸乙酯=5/1至3/1)纯化,得到黄色油状产物1'-(4-甲氧基苄基)-7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(945毫克,收率:78.95%)。LCMS(ESI):m/z=427M+H]+Step 17i: Preparation of tert-butyl 1'-(4-methoxybenzyl)-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0110-59): Under nitrogen protection, tert-butyl 3-(2-(2-chloro-6-(methyl-d3)pyridin-3-yl)ethyl)-3-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate A mixture of butyl ester (0109-59) (1.30 g, 2.81 mmol, 1.0 eq.), palladium acetate (63 mg, 0.28 mmol, 0.1 eq.), 2-dicyclohexylphosphine-2',6'-diisopropoxy-1,1'-biphenyl (262 mg, 0.56 mmol, 0.2 eq.) and sodium tert-butoxide (540 mg, 5.62 mmol, 2.0 eq.) was stirred in a microwave apparatus for 1 hour. The mixture was diluted with water and extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 5/1 to 3/1) to give a yellow oily product, 1'-(4-methoxybenzyl)-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (945 mg, yield: 78.95%). LCMS (ESI): m/z = 427M+H] + .

步骤17j:7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(化合物0112-59)的制备:往1'-(4-甲氧基苄基)-7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(0110-59)(945毫克,2.22毫摩尔,1.0当量)的二氯甲烷(8毫升)混合物中加入三氟乙酸(4毫升)并在室温下搅拌4小时。在真空下除去溶剂,用碳酸氢钠溶液调节残余物的pH至8。加入二碳酸二叔丁酯(485毫克,2.22毫摩尔,1.0当量)的四氢呋喃(5毫升)溶液,并在室温下搅拌3小时。将混合物加水稀释并用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物经硅胶柱色谱(洗脱液:二氯甲烷至二氯甲烷/甲醇=10/1)纯化,得到黄色油状产物7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(580毫克,收率:85.46%)。LCMS(ESI):m/z=307[M+H]+Step 17j: Preparation of tert-butyl 7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0112-59): To a mixture of tert-butyl 1'-(4-methoxybenzyl)-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0110-59) (945 mg, 2.22 mmol, 1.0 eq) in dichloromethane (8 mL) was added trifluoroacetic acid (4 mL) and stirred at room temperature for 4 hours. The solvent was removed under vacuum and the pH of the residue was adjusted to 8 with sodium bicarbonate solution. A solution of di-tert-butyl dicarbonate (485 mg, 2.22 mmol, 1.0 eq) in tetrahydrofuran (5 mL) was added and stirred at room temperature for 3 hours. The mixture was diluted with water and extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: dichloromethane to dichloromethane/methanol = 10/1) to obtain a yellow oily product 7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (580 mg, yield: 85.46%). LCMS (ESI): m/z = 307 [M+H] + .

步骤17k:6'-溴-7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(化合物0115-59)的制备:在氮气保护下,向7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(0112-59)(550毫克,1.80毫摩尔,1.0当量)的二氯甲烷(10毫升)溶液中加入1,3-二溴-5,5-二甲基海因(309毫克,1.08毫摩尔,0.6当量),并在0℃下搅拌30分钟。反应用亚 硫酸钠溶液淬灭。将混合物用水稀释并用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱(洗脱液:二氯甲烷/甲醇=50/1至30/1)纯化,得到黄色固体产物6'-溴-7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(720毫克,收率:100%)。(ESI):m/z=386[M+H]+Step 17k: Preparation of tert-butyl 6'-bromo-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0115-59): Under nitrogen protection, 1,3-dibromo-5,5-dimethylhydantoin (309 mg, 1.08 mmol, 0.6 eq) was added to a solution of tert-butyl 7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0112-59) (550 mg, 1.80 mmol, 1.0 eq) in dichloromethane (10 ml) and stirred at 0°C for 30 minutes. The reaction was stirred with 2% thiocyanate. The mixture was quenched with sodium sulfate solution. The mixture was diluted with water and extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol=50/1 to 30/1) to give a yellow solid product 6'-bromo-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (720 mg, yield: 100%). (ESI): m/z=386[M+H] + .

步骤17l:(1-(叔丁氧羰基)-7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-6'-基)硼酸(化合物0117-59)的制备:在氮气保护下,将6'-溴-7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(0115-59)(690毫克,1.79毫摩尔,1.0当量),四羟基二硼(402毫克,4.48毫摩尔,2.5当量),Xphos-Pd-G2(30毫克,0.036毫摩尔,0.02当量),2-二环己基磷-2’,4’,6’-三异丙基联苯(26毫克,0.054毫摩尔,0.03当量),乙酸钾(439毫克,4.48毫摩尔,2.5当量)和叔丁醇钠(4.3毫克,0.045毫摩尔,0.025当量)的甲醇/乙二醇(20毫升/1毫升)混合物在55℃下搅拌1小时。将混合物用水稀释并用二氯甲烷萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱(洗脱液:二氯甲烷/甲醇=30/1至10/1)纯化,得到黄色油状产物(1-(叔丁氧羰基)-7'-(甲基-d3)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-6'-基)硼酸(360毫克,收率:57.46%)。LCMS(ESI):m/z=351M+H]+Step 171: Preparation of (1-(tert-butyloxycarbonyl)-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (Compound 0117-59): Under nitrogen protection, 6'-bromo-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0115-59) (690 mg, 1.79 mmol, 1.0 equiv), tetrahydroxydiboron ( A mixture of 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (26 mg, 0.054 mmol, 0.03 eq.), potassium acetate (439 mg, 4.48 mmol, 2.5 eq.) and sodium tert-butoxide (4.3 mg, 0.045 mmol, 0.025 eq.) in methanol/ethylene glycol (20 ml/1 ml) was stirred at 55° C. for 1 hour. The mixture was diluted with water and extracted with dichloromethane, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol=30/1 to 10/1) to give a yellow oily product (1-(tert-butyloxycarbonyl)-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridin]-6'-yl)boronic acid (360 mg, yield: 57.46%). LCMS (ESI): m/z=351M+H] + .

步骤17m:(7'-(甲基-d3)-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(化合物0119-59)的制备:在氮气保护下,将(1-(叔丁氧羰基)-7'-(甲基-d3)-3',4'-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-6'-基)硼酸(0117-59)(360毫克,1.03毫摩尔,1.0当量),2-溴嘧啶(213毫克,1.34毫摩尔,1.3当量),Xphos-Pd-G2(30毫克,0.036毫摩尔,0.02当量),2-二环己基磷-2’,4’,6’-三异丙基联苯(15毫克,0.031毫摩尔,0.03当量)和碳酸钠(328毫克,3.09毫摩尔,3.0当量)的甲苯/乙醇/水(4毫升/2毫升/2毫升)混合物在60℃下搅拌1小时。将混合物用水稀释并用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物用硅胶柱色谱(洗脱液:二氯甲烷/甲醇=40/1至20/1)纯化,得到7'-(甲基-d3)-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(290毫克,收率:73.42%)。LCMS(ESI):m/z=385[M+H]+Step 17m: Preparation of (7'-(methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0119-59): Under nitrogen protection, (1-(tert-butyloxycarbonyl)-7'-(methyl-d3)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester 2-(2-(2-(2-(2-(2-nitro-3,2'-[1,8]naphthyridin]-6'-yl)boronic acid (0117-59) (360 mg, 1.03 mmol, 1.0 equiv), 2-bromopyrimidine (213 mg, 1.34 mmol, 1.3 equiv), Xphos-Pd-G2 (30 mg, 0.036 mmol, 0.02 equiv), 2-dicyclohexylphosphino-2',4',6'-triisopropylamine A mixture of propylbiphenyl (15 mg, 0.031 mmol, 0.03 eq) and sodium carbonate (328 mg, 3.09 mmol, 3.0 eq) in toluene/ethanol/water (4 ml/2 ml/2 ml) was stirred at 60°C for 1 hour. The mixture was diluted with water and extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol = 40/1 to 20/1) to give tert-butyl 7'-(methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (290 mg, yield: 73.42%). LCMS (ESI): m/z = 385 [M+H] + .

步骤17n:(甲基-d3)-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]二盐酸盐(化合物0121-59)的制备:将7'-(甲基-d3)-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-羧酸叔丁酯(77毫克,0.20毫摩尔,1.0当量)的氯化氢-甲醇(2毫升)溶液在室温下搅拌2小时。在真空下除去溶剂。残余物无需纯化用于下一步。(粗品)LCMS(ESI):m/z=285[M+H]+Step 17n: Preparation of (methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0121-59): A solution of tert-butyl 7'-(methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (77 mg, 0.20 mmol, 1.0 eq) in hydrogen chloride-methanol (2 mL) was stirred at room temperature for 2 hours. The solvent was removed under vacuum. The residue was used in the next step without purification. (Crude) LCMS (ESI): m/z=285[M+H] + .

步骤17o:(2R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-(7'-(甲基-d3)-6'-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物59)的制备:在(R)-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(0406-7)(40毫克,0.20毫摩尔,1.0当量)的乙腈(2毫升)混合物中加入吡啶三氟甲烷磺酸盐 (92毫克,0.4毫摩尔l,2.0当量)和N,N'-羰基二咪唑(36毫克,0.22毫摩尔,1.1当量),将混合物在室温下搅拌1小时。将(甲基-d3)-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]二盐酸盐(77毫克,0.60毫摩尔,3.0当量)加入反应混合物中,室温搅拌3小时。将混合物用水稀释并用乙酸乙酯萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残留物经厚制备薄层色谱(洗脱液:二氯甲烷/甲醇/氨水=80/3.5/0.8)纯化,得到白色固体产物(2R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-(7'-(甲基-d3)-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-基)丙-1-酮(化合物59)(40毫克,收率:43.01%)。LCMS(ESI):m/z=466[M+H]+1HNMR(500MHz,MeOD)δ8.71(t,J=3.8Hz,2H),7.81(dd,J=75.5,11.1Hz,2H),7.24–7.13(m,1H),6.72–6.52(m,1H),4.22–3.99(m,1H),3.77(t,J=7.6Hz,3H),3.64–3.25(m,4H),2.85–2.38(m,2H),2.10–1.62(m,5H),1.35(dt,J=12.9,6.8Hz,3H).Step 17o: Preparation of (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(7'-(methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 59): To a mixture of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0406-7) (40 mg, 0.20 mmol, 1.0 eq.) in acetonitrile (2 mL) was added pyridine trifluoromethanesulfonate (92 mg, 0.4 mmol, 2.0 eq) and N,N'-carbonyldiimidazole (36 mg, 0.22 mmol, 1.1 eq) were added and the mixture was stirred at room temperature for 1 hour. (Methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (77 mg, 0.60 mmol, 3.0 eq) was added to the reaction mixture and stirred at room temperature for 3 hours. The mixture was diluted with water and extracted with ethyl acetate, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (eluent: dichloromethane/methanol/aqueous ammonia=80/3.5/0.8) to give a white solid product (2R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-(7'-(methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 59) (40 mg, yield: 43.01%). LCMS (ESI): m/z=466[M+H] + . 1 HNMR(500MHz,MeOD)δ8.71(t,J=3.8Hz,2H),7.81(dd,J=75.5,11.1Hz,2H),7.24–7.13(m,1H),6.72–6.52(m,1H),4.22–3 .99(m,1H),3.77(t,J=7.6Hz,3H),3.64–3.25(m,4H),2.85–2.38(m,2H),2.10–1.62(m,5H),1.35(dt,J=12.9,6.8Hz,3H).

实施例18:(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7'-(甲基-d3)-6'-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物101)的制备:Example 18: Preparation of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-(methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 101):

将化合物59(31毫克)用手性高效液相色谱(色谱柱:ChiralPak AD-H,10×250mm)进行分离。第一个洗脱的对映异构体为白色固体产物(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((S)-7'-(甲基-d3)-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-基)丙-1-酮(化合物101)(19毫克,收率:62.50%)。LCMS(ESI):466[M+H]+1H NMR(500MHz,DMSO)δ8.80(d,J=2.6Hz,2H),8.11(d,J=5.7Hz,1H),7.89(d,J=11.6Hz,1H),7.23(d,J=50.3Hz,2H),6.72(dd,J=30.7,4.6Hz,1H),4.12(dd,J=26.8,6.9Hz,1H),3.83(s,3H),3.61–3.37(m,4H),2.85–2.59(m,2H),2.05–1.62(m,4H),1.33(t,J=6.7Hz,3H).Compound 59 (31 mg) was separated by chiral HPLC (chromatographic column: ChiralPak AD-H, 10×250 mm). The first eluting enantiomer was a white solid product (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((S)-7'-(methyl-d3)-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 101) (19 mg, yield: 62.50%). LCMS (ESI): 466 [M+H] + . 1H NMR (500MHz, DMSO) δ8.80(d,J=2.6Hz,2H),8.11(d,J=5.7Hz,1H),7.89(d,J=11.6Hz,1H),7.23(d,J=50.3Hz,2H),6.72(dd,J=30.7,4 .6Hz,1H),4.12(dd,J=26.8,6.9Hz,1H),3.83(s,3H),3.61–3.37(m,4H),2.85–2.59(m,2H),2.05–1.62(m,4H),1.33(t,J=6.7Hz,3H).

实施例19:2-(5-氟苯并呋喃-6-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-硫酮(化合物12)的制备(按照方案五线路制备):Example 19: Preparation of 2-(5-fluorobenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (Compound 12) (prepared according to Scheme 5 route):

将2-(5-氟苯并呋喃-6-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物14)(30毫克,0.064毫摩尔,1.0当量)和劳森试剂(49毫克,0.12毫摩尔,1.89当量)在甲苯(2毫升)中的混合物在125℃下微波加热搅拌1小时。反应溶液用水稀释,用乙酸乙酯萃取,有机相干燥并减压浓缩,得到粗品油状物。粗品用厚柱层析(洗脱液:二氯甲烷/甲醇=10/1)纯化,得到2-(5-氟苯并呋喃-6-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-硫酮(17毫克,产率54.8%)。LCMS(ESI):m/z=488[M+1]+.1H NMR(500MHz,MeOD)δ8.78–8.67(m,2H),7.85–7.62(m,2H),7.61–7.49(m,1H),7.31–6.97(m,2H),6.78–6.56(m,1H),4.56(d,J=7.2Hz,1H),3.93–3.61(m,4H),2.75(ddd,J=19.9,14.6,5.6Hz,2H),2.57–2.41(m,3H),2.15–1.83(m,4H),1.51(dd,J=12.8,6.5Hz,3H). A mixture of 2-(5-fluorobenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 14) (30 mg, 0.064 mmol, 1.0 eq) and Lawesson's reagent (49 mg, 0.12 mmol, 1.89 eq) in toluene (2 ml) was stirred and heated in a microwave at 125° C. for 1 hour. The reaction solution was diluted with water, extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure to obtain a crude oil. The crude product was purified by thick column chromatography (eluent: dichloromethane/methanol = 10/1) to give 2-(5-fluorobenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (17 mg, 54.8% yield). LCMS (ESI): m/z=488[M+1] + . 1 H NMR(500MHz,MeOD)δ8.78–8.67(m,2H),7.85–7.62(m,2H),7.61–7.49(m,1H),7.31–6.97(m,2H),6.78–6.56(m,1H),4.56(d,J=7.2H z,1H),3.93–3.61(m,4H),2.75(ddd,J=19.9,14.6,5.6Hz,2H),2.57–2.41(m,3H),2.15–1.83(m,4H),1.51(dd,J=12.8,6.5Hz,3H).

实施例20:2-(4-甲氧基苯并呋喃-6-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物87)的制备:
Example 20: Preparation of 2-(4-methoxybenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 87):

步骤20a:4-溴-2,6-二甲氧基苯甲醛的制备:往4-溴-2,6-二氟苯甲醛(3.5克,15.84毫摩尔,1.0当量)在甲醇(20毫升)的混合物中加入甲醇钠(5.4M的甲醇溶液,5.87毫升,31.67毫摩尔,2.0当量)。混合物在氮气氛围下加热至65℃反应3.5小时。减压下除去溶剂。残余物用水(60毫升)稀释然后用乙酸乙酯(30毫升×3)萃取。合并的有机层用饱和食盐水(30毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯8:1),得到白色固体4-溴-2,6-二甲氧基苯甲醛(2.89克,收率:74%)。LCMS(ESI):m/z 245[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=4:1)。Step 20a: Preparation of 4-bromo-2,6-dimethoxybenzaldehyde: Sodium methoxide (5.4 M methanol solution, 5.87 mL, 31.67 mmol, 2.0 eq.) was added to a mixture of 4-bromo-2,6-difluorobenzaldehyde (3.5 g, 15.84 mmol, 1.0 eq.) in methanol (20 mL). The mixture was heated to 65°C under nitrogen atmosphere for 3.5 hours. The solvent was removed under reduced pressure. The residue was diluted with water (60 mL) and then extracted with ethyl acetate (30 mL×3). The combined organic layers were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate 8:1) to give 4-bromo-2,6-dimethoxybenzaldehyde (2.89 g, yield: 74%) as a white solid. LCMS (ESI): m/z 245 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=4:1).

步骤20b:4-溴-2-羟基-6-甲氧基苯甲醛的制备:在0℃下,往三氯化铝(3.54克,26.53毫摩尔,1.0当量)和碘化钠(3.98克,26.53毫摩尔,2.0当量)在乙腈(16毫升)的混合物中逐滴加入4-溴-2,6-二甲氧基苯甲醛(3.25克,13.27毫摩尔,1.0当量)的二氯甲烷(16毫升)溶液。混合物升到室温然后搅拌4小时。加入1N稀盐酸(50毫升)淬灭反应。水层用乙酸乙酯(30毫升×3)萃取。合并的有机层用饱和食盐水(30毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯30:1),得到白色固体4-溴-2-羟基-6-甲氧基苯甲醛(2.87克,收率:94%)。LCMS(ESI):m/z 231[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=20:1)。Step 20b: Preparation of 4-bromo-2-hydroxy-6-methoxybenzaldehyde: To a mixture of aluminum chloride (3.54 g, 26.53 mmol, 1.0 eq.) and sodium iodide (3.98 g, 26.53 mmol, 2.0 eq.) in acetonitrile (16 ml) at 0°C was added dropwise a solution of 4-bromo-2,6-dimethoxybenzaldehyde (3.25 g, 13.27 mmol, 1.0 eq.) in dichloromethane (16 ml). The mixture was warmed to room temperature and then stirred for 4 hours. 1N dilute hydrochloric acid (50 ml) was added to quench the reaction. The aqueous layer was extracted with ethyl acetate (30 ml x 3). The combined organic layers were washed with saturated brine (30 ml), dried over anhydrous sodium sulfate and concentrated. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate 30:1) to give 4-bromo-2-hydroxy-6-methoxybenzaldehyde (2.87 g, yield: 94%) as a white solid. LCMS (ESI): m/z 231 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 20:1).

步骤20c:6-溴-4-甲氧基苯并呋喃的制备:往4-溴-2-羟基-6-甲氧基苯甲醛(2.87克,12.42毫摩尔,1.0当量)和碳酸铯(10.12克,31.05毫摩尔,2.5当量)在N,N-二甲基甲酰胺(20毫升)的混合物中加入溴乙酸乙酯(2.49克,14.91毫摩尔,1.2当量)。混合物在氮气氛围下加热至120℃反应8小时。混合物用水(80毫升)稀释然后用乙酸乙酯(20毫升×3)萃取。合并的有机层用饱和食盐水(20毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯50:1),得到白色固体6-溴-4-甲氧基苯并呋喃(0.492克,收率:17%)。LCMS(ESI):m/z 227[M+1]+;TLC:Rf 0.8(石油醚:乙酸乙酯=10:1)。Step 20c: Preparation of 6-bromo-4-methoxybenzofuran: To a mixture of 4-bromo-2-hydroxy-6-methoxybenzaldehyde (2.87 g, 12.42 mmol, 1.0 eq.) and cesium carbonate (10.12 g, 31.05 mmol, 2.5 eq.) in N,N-dimethylformamide (20 ml) was added ethyl bromoacetate (2.49 g, 14.91 mmol, 1.2 eq.). The mixture was heated to 120°C under nitrogen atmosphere for 8 hours. The mixture was diluted with water (80 ml) and then extracted with ethyl acetate (20 ml x 3). The combined organic layer was washed with saturated brine (20 ml), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate 50:1) to give 6-bromo-4-methoxybenzofuran (0.492 g, yield: 17%) as a white solid. LCMS (ESI): m/z 227 [M+1] + ; TLC: Rf 0.8 (petroleum ether:ethyl acetate=10:1).

步骤20d:2-(4-甲氧基苯并呋喃-6-基)丙酸叔丁酯的制备:往6-溴-4-甲氧基苯并呋喃(100毫克,0.44毫摩尔,1.0当量),Pd(dba)2(25毫克,0.044毫摩尔,0.1当量)和X-Phos(42毫克,0.088毫摩尔,0.2当量)在四氢呋喃(1毫升)的混合物中加入(1-(叔丁氧基)-1-氧代丙-2-基)溴化 锌(0.96M的四氢呋喃溶液,2毫升,1.92毫摩尔,4.4当量)。混合物在氮气氛围下加热至65℃反应1.5小时。混合物用饱和氯化铵溶液(20毫升)稀释然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(15毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(石油醚:乙酸乙酯=50:1)纯化,得到淡黄色油状物2-(4-甲氧基苯并呋喃-6-基)丙酸叔丁酯(79毫克,收率:65%)。LCMS(ESI):m/z 277[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=50:1)。Step 20d: Preparation of tert-butyl 2-(4-methoxybenzofuran-6-yl)propanoate: To a mixture of 6-bromo-4-methoxybenzofuran (100 mg, 0.44 mmol, 1.0 eq), Pd(dba) 2 (25 mg, 0.044 mmol, 0.1 eq) and X-Phos (42 mg, 0.088 mmol, 0.2 eq) in tetrahydrofuran (1 ml) was added (1-(tert-butyloxy)-1-oxopropan-2-yl)bromide. Zinc (0.96M tetrahydrofuran solution, 2 ml, 1.92 mmol, 4.4 eq). The mixture was heated to 65 ° C under a nitrogen atmosphere for 1.5 hours. The mixture was diluted with saturated ammonium chloride solution (20 ml) and then extracted with ethyl acetate (15 ml×3). The combined organic layer was washed with saturated brine (15 ml), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (petroleum ether: ethyl acetate = 50: 1) to give a light yellow oily substance tert-butyl 2-(4-methoxybenzofuran-6-yl) propionate (79 mg, yield: 65%). LCMS (ESI): m/z 277 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 50: 1).

步骤20e:2-(4-甲氧基苯并呋喃-6-基)丙酸的制备:将2-(4-甲氧基苯并呋喃-6-基)丙酸叔丁酯(70毫克,0.254毫摩尔,1.0当量)的氯化氢/二氧六环溶液(4M,1毫升)的混合物在室温下搅拌过夜。减压下除去溶剂。残余物在真空下干燥得到淡黄色油状物(2-(4-甲氧基苯并呋喃-6-基)丙酸(56毫克,粗品)。LCMS(ESI):m/z 221[M+1]+;TLC:Rf 0.3(二氯甲烷:甲醇=10:1)。Step 20e: Preparation of 2-(4-methoxybenzofuran-6-yl)propanoic acid: A mixture of tert-butyl 2-(4-methoxybenzofuran-6-yl)propanoate (70 mg, 0.254 mmol, 1.0 eq) in hydrogen chloride/dioxane solution (4M, 1 ml) was stirred at room temperature overnight. The solvent was removed under reduced pressure. The residue was dried under vacuum to give a pale yellow oil (2-(4-methoxybenzofuran-6-yl)propanoic acid (56 mg, crude). LCMS (ESI): m/z 221 [M+1] + ; TLC: Rf 0.3 (dichloromethane:methanol=10:1).

步骤20f:2-(4-甲氧基苯并呋喃-6-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物87)的制备:往2-(4-甲氧基苯并呋喃-6-基)丙酸(23毫克,0.105毫摩尔,1.0当量),2-(7-偶氮苯并三氮唑)-N,N,N’,N’-四甲基脲六氟磷酸酯(42毫克,0.11毫摩尔,1.05当量)和N,N-二异丙基乙胺(68毫克,0.525毫摩尔,5.0当量)在二氯甲烷(2.5毫升)的混合物中加入(S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-1)(37毫克,0.105毫摩尔,1.0当量)。混合物在室温下搅拌0.5小时。混合物用水(20毫升)稀释然后用乙酸乙酯(10毫升×3)萃取。合并的有机层用饱和食盐水(15毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(二氯甲烷:甲醇:氨水=80:3.5:0.8)纯化,得到两种非对映异构体的白色固体混合物2-(4-甲氧基苯并呋喃-6-基)-1-((S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物87)(34毫克,收率:67%),1H NMR(500MHz,MeOD)δ8.81(dd,J=7.3,4.8Hz,2H),7.82(d,J=72.9Hz,1H),7.57(d,J=56.2Hz,1H),7.30(d,J=3.9Hz,1H),7.07(dd,J=25.2,11.8Hz,1H),6.89–6.61(m,2H),4.03(dd,J=25.4,19.3Hz,1H),3.91(dd,J=30.5,17.0Hz,3H),3.75–3.36(m,4H),2.83(d,J=20.9Hz,2H),2.56(dd,J=33.0,14.8Hz,3H),2.23–1.71(m,4H),1.50–1.42(m,3H).Step 20f: Preparation of 2-(4-methoxybenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 87): 2-(4-methoxybenzofuran-6-yl)propanoic acid (23 mg, 0.105 mmol, 1.0 equiv), 2-(7-azobenzotriazole)-N,N,N',N'- To a mixture of tetramethyluronium hexafluorophosphate (42 mg, 0.11 mmol, 1.05 eq) and N,N-diisopropylethylamine (68 mg, 0.525 mmol, 5.0 eq) in dichloromethane (2.5 ml) was added (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-1) (37 mg, 0.105 mmol, 1.0 eq). The mixture was stirred at room temperature for 0.5 hours. The mixture was diluted with water (20 ml) and then extracted with ethyl acetate (10 ml x 3). The combined organic layer was washed with saturated brine (15 ml), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (dichloromethane:methanol:aqueous ammonia=80:3.5:0.8) to give a white solid mixture of two diastereoisomers 2-(4-methoxybenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine- 3,2 '-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 87) (34 mg, yield: 67%), NMR(500MHz,MeOD)δ8.81(dd,J=7.3,4.8Hz,2H),7.82(d,J=72.9Hz,1H),7.57(d,J=56 .2Hz,1H),7.30(d,J=3.9Hz,1H),7.07(dd,J=25.2,11.8Hz,1H),6.89–6.61(m,2H),4. 03(dd,J=25.4,19.3Hz,1H),3.91(dd,J=30.5,17.0Hz,3H),3.75–3.36(m,4H),2.83(d ,J=20.9Hz,2H),2.56(dd,J=33.0,14.8Hz,3H),2.23–1.71(m,4H),1.50–1.42(m,3H).

实施例21:(R)-2-(4-甲氧基苯并呋喃-6-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2-[1,8]萘啶]-1-基)丙烷-1-酮(化合物102)的制备:Example 21: Preparation of (R)-2-(4-methoxybenzofuran-6-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 102):

使用手性高效液相色谱(柱:ChiralPakAD-H,10×250mm)分离化合物87非对映异构体(20毫克)。第二个洗脱的对映体是白色固体(R)-2-(4-甲氧基苯并呋喃-6-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2-[1,8]萘啶]-1-基)丙烷-1-酮(化合物102)(5.6毫克,收率:28%)。LCMS(ESI):m/z 484[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=20:1)。1H NMR(500MHz,MeOD)δ8.79(d,J=4.8Hz,2H),7.75(d,J=70.3Hz,1H),7.63(d,J=1.7Hz,1H),7.29(dd,J=6.4,3.2Hz,1H),7.08(d,J=28.7Hz,1H),6.84(s,1H),6.72(d,J=17.3Hz,1H),4.51(s,4H), 4.12–3.86(m,4H),3.76–3.35(m,4H),2.85(d,J=6.3Hz,1H),2.59–2.49(m,3H),2.22–1.79(m,4H),1.47–1.39(m,3H).Compound 87 diastereomers (20 mg) were separated using chiral HPLC (column: ChiralPak AD-H, 10×250 mm). The second eluting enantiomer was a white solid (R)-2-(4-methoxybenzofuran-6-yl)-1-((S)-7′-methyl-6′-(pyrimidin-2-yl)-3′,4′-dihydro-1′H-spiro[pyrrolidine-3,2-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 102) (5.6 mg, yield: 28%). LCMS (ESI): m/z 484 [M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=20:1). 1H NMR (500MHz, MeOD) δ8.79(d,J=4.8Hz,2H),7.75(d,J=70.3Hz,1H),7.63(d,J=1.7Hz,1H),7.29( dd,J=6.4,3.2Hz,1H),7.08(d,J=28.7Hz,1H),6.84(s,1H),6.72(d,J=17.3Hz,1H),4.51(s,4H), 4.12–3.86(m,4H),3.76–3.35(m,4H),2.85(d,J=6.3Hz,1H),2.59–2.49(m,3H),2.22–1.79(m,4H),1.47–1.39(m,3H).

实施例22:6-(1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’-H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-1-氧代丙烷-2-基)苯并呋喃-5-腈(化合物77)的制备:
Example 22: Preparation of 6-(1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-1-oxopropane-2-yl)benzofuran-5-carbonitrile (Compound 77):

步骤22a:1-溴-2-溴甲基-4-甲氧基苯的制备:氮气保护下,将1-溴-4-甲氧基-2-甲基苯(2.5克,12.44毫摩尔,1.0当量),N-溴代琥珀酰亚胺(2.22克,12.44毫摩尔,1.0当量)和偶氮二异丁腈(409毫克,2.49毫摩尔,0.2当量)的1,2-二氯乙烷(25毫升)的混合物在72℃下搅拌过夜。加入二氯甲烷,用硫代硫酸钠溶液和饱和食盐水洗。有机相干燥并减压浓缩,得到黄色Step 22a: Preparation of 1-bromo-2-bromomethyl-4-methoxybenzene: Under nitrogen protection, a mixture of 1-bromo-4-methoxy-2-methylbenzene (2.5 g, 12.44 mmol, 1.0 eq.), N-bromosuccinimide (2.22 g, 12.44 mmol, 1.0 eq.) and azobisisobutyronitrile (409 mg, 2.49 mmol, 0.2 eq.) in 1,2-dichloroethane (25 ml) was stirred at 72°C overnight. Dichloromethane was added, and the mixture was washed with sodium thiosulfate solution and saturated brine. The organic phase was dried and concentrated under reduced pressure to obtain a yellow

油状产物1-溴-2-溴甲基-4-甲氧基苯(4.08克,粗品)。LCMS(ESI):m/z=279[M+1]+.Oily product 1-bromo-2-bromomethyl-4-methoxybenzene (4.08 g, crude product). LCMS (ESI): m/z=279 [M+1] + .

步骤22b:2-(2-溴-5-甲氧基苯基)乙腈的制备:氮气保护下,1-溴-2-溴甲基-4-甲氧基苯(4.08克,12.44毫摩尔,1.0当量),三甲基氰硅烷(1.85克,18.66毫摩尔,1.5当量)和四丁基氟化铵(1M四氢呋喃溶液,18.7毫升,18.66毫摩尔,1.5当量)在乙腈(30毫升)的混合物在室温下搅拌1小时。混合物用乙酸乙酯稀释,用水和饱和食盐水洗。有机相干燥并减压浓缩,得到棕色油状产物2-(2-溴-5-甲氧基苯基)乙腈(5.61克,粗品)。LCMS(ESI):m/z=226[M+1]+.Step 22b: Preparation of 2-(2-bromo-5-methoxyphenyl)acetonitrile: Under nitrogen protection, a mixture of 1-bromo-2-bromomethyl-4-methoxybenzene (4.08 g, 12.44 mmol, 1.0 eq.), trimethylsilyl cyanide (1.85 g, 18.66 mmol, 1.5 eq.) and tetrabutylammonium fluoride (1M tetrahydrofuran solution, 18.7 ml, 18.66 mmol, 1.5 eq.) in acetonitrile (30 ml) was stirred at room temperature for 1 hour. The mixture was diluted with ethyl acetate, washed with water and saturated brine. The organic phase was dried and concentrated under reduced pressure to give a brown oily product, 2-(2-bromo-5-methoxyphenyl)acetonitrile (5.61 g, crude product). LCMS (ESI): m/z=226[M+1] + .

步骤22c:2-(2-溴-5-甲氧基苯基)乙酸的制备:向2-(2-溴-5-甲氧基苯基)乙腈(5.61克,12.44毫摩尔,1.0当量)的乙醇(50毫升)溶液中加入氢氧化钾(2.09克,37.32毫摩尔,3.0当量)的水溶液(50毫升)。混合物在85℃下搅拌过夜。混合物浓缩至75毫升,用乙酸乙酯洗。用浓盐酸调水相pH值至2。加入乙酸乙酯萃取,用水和饱和食盐水洗。有机相干燥并减压浓缩,得到黄色油状产物2-(2-溴-5-甲氧基苯基)乙酸(1.86克,收率:60.7%)。LCMS(ESI):m/z=245[M+1]+.Step 22c: Preparation of 2-(2-bromo-5-methoxyphenyl)acetic acid: To a solution of 2-(2-bromo-5-methoxyphenyl)acetonitrile (5.61 g, 12.44 mmol, 1.0 eq) in ethanol (50 ml) was added an aqueous solution (50 ml) of potassium hydroxide (2.09 g, 37.32 mmol, 3.0 eq). The mixture was stirred at 85°C overnight. The mixture was concentrated to 75 ml and washed with ethyl acetate. The pH value of the aqueous phase was adjusted to 2 with concentrated hydrochloric acid. Ethyl acetate was added for extraction, and the mixture was washed with water and saturated brine. The organic phase was dried and concentrated under reduced pressure to give a yellow oily product, 2-(2-bromo-5-methoxyphenyl)acetic acid (1.86 g, yield: 60.7%). LCMS (ESI): m/z=245[M+1] + .

步骤22d:2-(2-溴-5-甲氧基苯基)乙酸甲酯的制备:在氮气保护下,向2-(2-溴-5-甲氧基苯基)乙酸(1.86克,7.56毫摩尔,1.0当量)和N,N-二甲基甲酰胺(0.1毫升)在10毫升二氯甲烷的混合物中滴加草酰氯(2M二氯甲烷溶液,7.56毫升,15.12毫摩尔,2.0当量)。混合物在室温下搅拌2小时。滴加2毫升甲醇,混合物搅拌30分钟。混合物用乙酸乙酯稀释,用水和饱和食盐水洗。有机相干燥并减压浓缩,残留物用硅胶柱层析色谱纯化(洗脱剂为:石油醚/乙酸 乙酯=100/1到50/1)得到黄色油状物2-(2-溴-5-甲氧基苯基)乙酸甲酯(1.8克,收率:91.3%)。MS(ES+):m/z=259(M+H)+.Step 22d: Preparation of methyl 2-(2-bromo-5-methoxyphenyl)acetate: Under nitrogen protection, oxalyl chloride (2M dichloromethane solution, 7.56 ml, 15.12 mmol, 2.0 eq.) was added dropwise to a mixture of 2-(2-bromo-5-methoxyphenyl)acetic acid (1.86 g, 7.56 mmol, 1.0 eq.) and N,N-dimethylformamide (0.1 ml) in 10 ml dichloromethane. The mixture was stirred at room temperature for 2 hours. 2 ml of methanol was added dropwise and the mixture was stirred for 30 minutes. The mixture was diluted with ethyl acetate and washed with water and saturated brine. The organic phase was dried and concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/acetic acid Ethyl ester = 100/1 to 50/1) to give methyl 2-(2-bromo-5-methoxyphenyl)acetate (1.8 g, yield: 91.3%) as a yellow oil. MS (ES + ): m/z = 259 (M+H) + .

步骤22e:2-(2-溴-5-甲氧基苯基)丙酸甲酯的制备:在氮气保护下,在-70℃下,向2-(2-溴-5-甲氧基苯基)乙酸甲酯(437毫克,1.68毫摩尔,1.0当量)的四氢呋喃溶液滴加双-(三甲基硅基)胺锂(1M四氢呋喃溶液,1.71毫升,1.71毫摩尔,1.02当量)。混合物搅拌1.5小时。滴加碘甲烷(429毫克,3.02毫摩尔,1.8当量)的四氢呋喃溶液,混合物在-70℃搅拌1小时。然后,混合物在室温下搅拌45分钟。加入氯化铵溶液淬灭反应。加入乙酸乙酯萃取,用水和饱和食盐水洗。有机相干燥并减压浓缩。残余物用硅胶柱层析色谱(洗脱剂:石油醚/乙酸乙酯=100/1至20/1)纯化,得到无色油状产物2-(2-溴-5-甲氧基苯基)丙酸甲酯(350毫克,收率:75.9%)。LCMS(ESI):m/z=273[M+H]+。Step 22e: Preparation of methyl 2-(2-bromo-5-methoxyphenyl)propanoate: To a solution of methyl 2-(2-bromo-5-methoxyphenyl)acetate (437 mg, 1.68 mmol, 1.0 eq) in tetrahydrofuran was added dropwise, under nitrogen, lithium bis-(trimethylsilyl)amide (1M in tetrahydrofuran, 1.71 ml, 1.71 mmol, 1.02 eq) at -70°C. The mixture was stirred for 1.5 hours. A solution of iodomethane (429 mg, 3.02 mmol, 1.8 eq) in tetrahydrofuran was added dropwise and the mixture was stirred at -70°C for 1 hour. The mixture was then stirred at room temperature for 45 minutes. The reaction was quenched by adding ammonium chloride solution. Ethyl acetate was added for extraction and washed with water and saturated brine. The organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100/1 to 20/1) to give a colorless oily product, methyl 2-(2-bromo-5-methoxyphenyl)propanoate (350 mg, yield: 75.9%). LCMS (ESI): m/z = 273 [M+H] +.

步骤22f:2-(2-氰基-5-甲氧基苯基)丙酸甲酯的制备:2-(2-溴-5-甲氧基苯基)丙酸甲酯(719毫克,2.63毫摩尔,1.0当量)和氰化亚铜(1.42克,15.80毫摩尔,6.0当量)在8毫升N-甲基吡咯烷酮的混合物在180℃下微波加热搅拌2小时。混合物经硅藻土过滤。滤液用乙酸乙酯稀释,用水和饱和食盐水洗。有机相干燥并减压浓缩,残留物用硅胶柱层析色谱纯化(洗脱剂为:石油醚/乙酸乙酯=100/1至10/1)得到黄色油状物2-(2-氰基-5-甲氧基苯基)丙酸甲酯(364毫克,收率:63.1%)。MS(ES+):m/z=220(M+H)+.Step 22f: Preparation of methyl 2-(2-cyano-5-methoxyphenyl)propanoate: A mixture of methyl 2-(2-bromo-5-methoxyphenyl)propanoate (719 mg, 2.63 mmol, 1.0 eq.) and cuprous cyanide (1.42 g, 15.80 mmol, 6.0 eq.) in 8 ml of N-methylpyrrolidone was stirred and heated in a microwave at 180°C for 2 hours. The mixture was filtered through celite. The filtrate was diluted with ethyl acetate, washed with water and saturated brine. The organic phase was dried and concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 100/1 to 10/1) to give methyl 2-(2-cyano-5-methoxyphenyl)propanoate (364 mg, yield: 63.1%) as a yellow oil. MS (ES + ): m/z = 220 (M+H) + .

步骤22g:2-(2-氰基-4-碘-5-甲氧基苯基)丙酸甲酯的制备:在室温下,向2-(2-氰基-5-甲氧基苯基)丙酸甲酯(331毫克,1.51毫摩尔,1.0当量)的三氟乙酸(5毫升)溶液中加入N-碘代丁二酰亚胺(340毫克,1.51毫摩尔,1.0当量),搅拌6小时。混合物浓缩,加入碳酸钠溶液。加入乙酸乙酯萃取,用水和饱和食盐水洗。有机相干燥并减压浓缩,残留物用硅胶柱层析色谱纯化(洗脱剂为:石油醚/乙酸乙酯=80/1至5/1)得到黄色油状产物2-(2-氰基-4-碘-5-甲氧基苯基)丙酸甲酯(355毫克,收率:68.0%)。LCMS(ESI):m/z=346[M+1]+.Step 22g: Preparation of methyl 2-(2-cyano-4-iodo-5-methoxyphenyl)propionate: To a solution of methyl 2-(2-cyano-5-methoxyphenyl)propionate (331 mg, 1.51 mmol, 1.0 eq.) in trifluoroacetic acid (5 ml) was added N-iodosuccinimide (340 mg, 1.51 mmol, 1.0 eq.) at room temperature and stirred for 6 hours. The mixture was concentrated and sodium carbonate solution was added. Ethyl acetate was added for extraction and washed with water and saturated brine. The organic phase was dried and concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 80/1 to 5/1) to give a yellow oily product methyl 2-(2-cyano-4-iodo-5-methoxyphenyl)propionate (355 mg, yield: 68.0%). LCMS (ESI): m/z = 346 [M+1] + .

步骤22h:2-(2-氰基-5-羟基-4-碘苯基)丙酸的制备:在氮气保护下,于0℃向乙硫醇(225毫克,3.63毫摩尔,5.0当量)的N,N-二甲基甲酰胺(5毫升)溶液中加入氢化钠(145毫克,3.63毫摩尔,1.0当量),在室温下搅拌30分钟。加入2-(2-氰基-4-碘-5-甲氧基苯基)丙酸甲酯(250毫克,0.725毫摩尔,1.0当量)的N,N-二甲基甲酰胺溶液。混合物在155℃搅拌1小时,加入稀盐酸淬灭反应。加入乙酸乙酯萃取,用水和饱和食盐水洗。有机相干燥并减压浓缩,残留物用硅胶柱层析色谱纯化(洗脱剂为:石油醚/乙酸乙酯=1/1至1/10)得到黄色油状产物2-(2-氰基-5-羟基-4-碘苯基)丙酸(173毫克,收率:75.1%)。LCMS(ESI):m/z=318[M+1]+.Step 22h: Preparation of 2-(2-cyano-5-hydroxy-4-iodophenyl)propionic acid: Under nitrogen protection, sodium hydride (145 mg, 3.63 mmol, 1.0 eq) was added to a solution of ethanethiol (225 mg, 3.63 mmol, 5.0 eq) in N,N-dimethylformamide (5 ml) at 0°C and stirred at room temperature for 30 minutes. A solution of methyl 2-(2-cyano-4-iodo-5-methoxyphenyl)propanoate (250 mg, 0.725 mmol, 1.0 eq) in N,N-dimethylformamide was added. The mixture was stirred at 155°C for 1 hour and diluted hydrochloric acid was added to quench the reaction. Ethyl acetate was added for extraction and washed with water and saturated brine. The organic phase was dried and concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 1/1 to 1/10) to obtain a yellow oily product 2-(2-cyano-5-hydroxy-4-iodophenyl)propanoic acid (173 mg, yield: 75.1%). LCMS (ESI): m/z = 318 [M+1] + .

步骤22i:2-(2-氰基-5-羟基-4-碘苯基)丙酸甲酯的制备:在氮气保护下,向2-(2-氰基-5-羟基-4-碘苯基)丙酸(160毫克,0.505毫摩尔,1.0当量)和N,N-二甲基甲酰胺(0.05毫升)在5 毫升二氯甲烷的混合物中滴加草酰氯(2M二氯甲烷溶液,0.76毫升,1.52毫摩尔,3.00当量)。混合物在室温下搅拌1.5小时。滴加2毫升甲醇,混合物搅拌15分钟。混合物用乙酸乙酯稀释,用水和饱和食盐水洗。有机相干燥并减压浓缩,残留物用硅胶柱层析色谱纯化(洗脱剂为:石油醚/乙酸乙酯=2/1到1/5)得到黄色油状物2-(2-氰基-5-羟基-4-碘苯基)丙酸甲酯(145毫克,收率:86.8%)。MS(ES+):m/z=332(M+H)+.Step 22i: Preparation of methyl 2-(2-cyano-5-hydroxy-4-iodophenyl)propanoate: Under nitrogen protection, 2-(2-cyano-5-hydroxy-4-iodophenyl)propanoic acid (160 mg, 0.505 mmol, 1.0 eq.) and N,N-dimethylformamide (0.05 ml) were added to 5% ethyl acetate. 1.52 mmol, 3.00 equiv.) was added dropwise to a mixture of 1.5 ml of dichloromethane and 2.0 ml of oxalyl chloride. The mixture was stirred at room temperature for 1.5 hours. 2 ml of methanol was added dropwise and the mixture was stirred for 15 minutes. The mixture was diluted with ethyl acetate and washed with water and saturated brine. The organic phase was dried and concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate = 2/1 to 1/5) to obtain a yellow oily substance 2-(2-cyano-5-hydroxy-4-iodophenyl) propionic acid methyl ester (145 mg, yield: 86.8%). MS (ES + ): m/z = 332 (M+H) + .

步骤22j:2-(5-氰基苯并呋喃-6-基)丙酸甲酯的制备:在氮气保护下,2-(2-氰基-5-羟基-4-碘苯基)丙酸甲酯(130毫克,0.393毫摩尔,1.0当量),三甲基硅乙炔(192毫克,0.393毫摩尔,1.0当量),碘化亚铜(38毫克,0.197毫摩尔,0.5当量),二氯二三苯基膦钯(28美股,0.04毫摩尔,0.1当量)和N,N-二异丙基乙胺(253毫克,1.96毫摩尔,5.0当量)在15毫升四氢呋喃的混合物于在66℃下搅拌1.5小时。混合物经硅藻土过滤。滤液减压浓缩至干。残余物溶解于10毫升甲醇中,然后加入碘化亚铜(75毫克,0.393毫摩尔,1.0当量)和N,N-二异丙基乙胺(253毫克,1.96毫摩尔,2.5当量)。混合物在72℃下搅拌2小时。混合物用二氯甲烷稀释,用氯化铵溶液和饱和食盐水洗。有机相干燥并减压浓缩至干。残留物溶解于12毫升四氢呋喃中,加入四丁基氟化铵(100毫克,0.32毫摩尔,0.8当量)。混合物用乙酸乙酯稀释,用水和饱和食盐水洗。有机相干燥并减压浓缩至干。残留物用厚制备薄层色谱(展开剂为:石油醚/乙酸乙酯=20/1)得到无色油状物2-(5-氰基苯并呋喃-6-基)丙酸甲酯(46毫克,收率:51.1%)。MS(ES+):m/z=230(M+H)+.Step 22j: Preparation of methyl 2-(5-cyanobenzofuran-6-yl)propanoate: Under nitrogen, a mixture of methyl 2-(2-cyano-5-hydroxy-4-iodophenyl)propanoate (130 mg, 0.393 mmol, 1.0 eq), trimethylsilylacetylene (192 mg, 0.393 mmol, 1.0 eq), cuprous iodide (38 mg, 0.197 mmol, 0.5 eq), dichlorobistriphenylphosphine palladium (28 US shares, 0.04 mmol, 0.1 eq) and N,N-diisopropylethylamine (253 mg, 1.96 mmol, 5.0 eq) in 15 ml of tetrahydrofuran was stirred at 66° C. for 1.5 hours. The mixture was filtered through celite. The filtrate was concentrated to dryness under reduced pressure. The residue was dissolved in 10 ml of methanol, and then cuprous iodide (75 mg, 0.393 mmol, 1.0 eq.) and N,N-diisopropylethylamine (253 mg, 1.96 mmol, 2.5 eq.) were added. The mixture was stirred at 72°C for 2 hours. The mixture was diluted with dichloromethane, washed with ammonium chloride solution and saturated brine. The organic phase was dried and concentrated to dryness under reduced pressure. The residue was dissolved in 12 ml of tetrahydrofuran, and tetrabutylammonium fluoride (100 mg, 0.32 mmol, 0.8 eq.) was added. The mixture was diluted with ethyl acetate, washed with water and saturated brine. The organic phase was dried and concentrated to dryness under reduced pressure. The residue was subjected to thick preparative thin layer chromatography (developing solvent: petroleum ether/ethyl acetate = 20/1) to obtain a colorless oily substance 2-(5-cyanobenzofuran-6-yl)propionic acid methyl ester (46 mg, yield: 51.1%). MS (ES + ): m/z = 230 (M + H) + .

步骤22k:2-(5-氰基苯并呋喃-6-基)丙酸的制备:向2-(5-氰基苯并呋喃-6-基)丙酸甲酯(46毫克,0.2毫摩尔,1.0当量)的四氢呋喃/水(2毫升/0.5毫升)溶液中加入氢氧化锂(17毫克,0.4毫摩尔,2.0当量)。混合物在室温下搅拌1小时。用稀盐酸酸化混合物。加入乙酸乙酯萃取,用水和饱和食盐水洗。有机相干燥并减压浓缩,得到黄色固体产物2-(5-氰基苯并呋喃-6-基)丙酸(30毫克,收率:69.7%)。LCMS(ESI):m/z=216[M+1]+.Step 22k: Preparation of 2-(5-cyanobenzofuran-6-yl)propanoic acid: To a solution of methyl 2-(5-cyanobenzofuran-6-yl)propanoate (46 mg, 0.2 mmol, 1.0 eq) in tetrahydrofuran/water (2 ml/0.5 ml) was added lithium hydroxide (17 mg, 0.4 mmol, 2.0 eq). The mixture was stirred at room temperature for 1 hour. The mixture was acidified with dilute hydrochloric acid. Ethyl acetate was added for extraction, and the mixture was washed with water and saturated brine. The organic phase was dried and concentrated under reduced pressure to give a yellow solid product, 2-(5-cyanobenzofuran-6-yl)propanoic acid (30 mg, yield: 69.7%). LCMS (ESI): m/z=216[M+1] + .

步骤22l:6-(1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’-H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-1-氧代丙烷-2-基)苯并呋喃-5-腈(化合物77)的制备:将(S)-7'-甲基-6'-(嘧啶-2-基)-3',4'-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]二盐酸盐(0122-1)(35毫克,0.09毫摩尔,0.9当量),2-(5-氰基苯并呋喃-6-基)丙酸(30毫克,0.1毫摩尔,1.0当量),N,N-二异丙基乙胺(40毫克,0.3毫摩尔,3.0当量)和2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(42毫克,0.11毫摩尔,1.1当量)的二氯甲烷(2毫升)混合物在室温下搅拌30分钟。混合物用二氯甲烷稀释,用水和饱和食盐水洗。有机相经无水硫酸钠干燥并减压浓缩。残余物经厚制备薄层色谱(展开剂:二氯甲烷/甲醇=20/1)纯化,得到黄色固体产物6-(1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’-H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-1-氧代丙烷-2-基)苯并呋喃-5-腈(38毫克,收率:88.3%)。LCMS(ESI): m/z=465[M+H]+1H NMR(500MHz,MeOD)δ8.81(dt,J=8.6,4.4Hz,2H),8.15–7.62(m,4H),7.33–7.12(m,1H),6.93(dd,J=37.9,1.3Hz,1H),4.57–4.30(m,1H),4.04–3.42(m,4H),2.92–2.61(m,2H),2.63–2.42(m,3H),2.20–1.69(m,4H),1.64–1.49(m,3H).Step 221: Preparation of 6-(1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-1-oxopropan-2-yl)benzofuran-5-carbonitrile (Compound 77): (S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122- A mixture of dichloromethane (2 ml) of 1-(4-nitrobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (42 mg, 0.11 mmol, 1.1 eq.) and 2-(7-azabenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (35 mg, 0.09 mmol, 0.9 eq.) was stirred at room temperature for 30 minutes. The mixture was diluted with dichloromethane and washed with water and saturated brine. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by thick preparative thin layer chromatography (developing solvent: dichloromethane/methanol = 20/1) to give a yellow solid product 6-(1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-1-oxopropane-2-yl)benzofuran-5-carbonitrile (38 mg, yield: 88.3%). LCMS (ESI): m/z=465[M+H] + . 1H NMR(500MHz,MeOD)δ8.81(dt,J=8.6,4.4Hz,2H),8.15–7.62(m,4H),7.33–7.12(m,1H),6.93(dd,J=37.9,1.3Hz,1H) ,4.57–4.30(m,1H),4.04–3.42(m,4H),2.92–2.61(m,2H),2.63–2.42(m,3H),2.20–1.69(m,4H),1.64–1.49(m,3H).

实施例23:(2R)-1-(5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’-H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-2-(5-氟-2-甲氧基吡啶-4-基)丙烷-1-酮(化合物55)的制备(按照方案一和四线路制备)Example 23: Preparation of (2R)-1-(5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-2-(5-fluoro-2-methoxypyridin-4-yl)propan-1-one (Compound 55) (Prepared according to Schemes 1 and 4)

步骤23a:1-(叔丁基)2-甲基4-羟基-4-(三甲基硅基)乙炔基)吡咯烷-1,2-二羧酸酯(化合物0102-55)的制备:在-78℃下,往乙炔基三甲基硅烷(4.85克,49.33毫摩尔,1.2当量)的四氢呋喃(60毫升)混合物中逐滴加入正丁基锂(2.5M的己烷溶液,17.3毫升,43.16毫摩尔,1.05当量)。混合物在-78℃氮气氛围下搅拌1小时。逐滴加入2-乙酰氧基-4-氧代吡咯烷-1-甲酸叔丁酯(0101-55)(10.0克,41.11毫摩尔,1.0当量)的四氢呋喃(40毫升)溶液。混合物升到室温然后搅拌过夜。加入饱和氯化铵溶液(60毫升)淬灭反应。水层用乙酸乙酯(50毫升×3)萃取。合并的有机层用饱和食盐水(50毫升)洗涤,经无水硫酸钠干燥并浓缩,得到棕色油状物1-(叔丁基)2-甲基4-羟基-4-(三甲基硅基)乙炔基)吡咯烷-1,2-二羧酸酯(12.2克,收率:87%)。LCMS(ESI):m/z 342[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=60:1)。Step 23a: Preparation of 1-(tert-butyl)2-methyl-4-hydroxy-4-(trimethylsilyl)ethynyl)pyrrolidine-1,2-dicarboxylate (Compound 0102-55): To a mixture of ethynyltrimethylsilane (4.85 g, 49.33 mmol, 1.2 eq) in tetrahydrofuran (60 mL) was added dropwise n-butyllithium (2.5 M in hexane, 17.3 mL, 43.16 mmol, 1.05 eq) at -78°C. The mixture was stirred at -78°C under nitrogen for 1 hour. A solution of tert-butyl 2-acetoxy-4-oxopyrrolidine-1-carboxylate (0101-55) (10.0 g, 41.11 mmol, 1.0 eq) in tetrahydrofuran (40 mL) was added dropwise. The mixture was warmed to room temperature and stirred overnight. Saturated ammonium chloride solution (60 ml) was added to quench the reaction. The aqueous layer was extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with saturated brine (50 ml), dried over anhydrous sodium sulfate and concentrated to give a brown oily substance 1-(tert-butyl) 2-methyl 4-hydroxy-4-(trimethylsilyl)ethynyl)pyrrolidine-1,2-dicarboxylate (12.2 g, yield: 87%). LCMS (ESI): m/z 342 [M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=60:1).

步骤23b:1-(叔丁基)2-甲基4-乙炔基-4-羟基吡咯烷-1,2-二羧酸酯(化合物0103-55)的制备:往1-(叔丁基)2-甲基4-羟基-4-(三甲基硅基)乙炔基)吡咯烷-1,2-二羧酸酯(0102-55)(12.2克,35.78毫摩尔,1.0当量)的甲醇(50毫升)混合物中加入碳酸钾(7.4克,53.67毫摩尔,1.5当量)。混合物在室温下搅拌3小时。减压下除去溶剂。残余物用水(60毫升)稀释然后用乙酸乙酯(50毫升×3)萃取。合并的有机层用饱和食盐水(50毫升)洗涤,经无水硫酸钠干燥并浓缩,得到棕色油状物1-(叔丁基)2-甲基4-乙炔基-4-羟基吡咯烷-1,2-二羧酸酯(5.61克,收率:58%)。LCMS(ESI):m/z 270[M+1]+;TLC:Rf 0.4(石油醚:乙酸乙酯=3:1)。Step 23b: Preparation of 1-(tert-butyl)-2-methyl-4-ethynyl-4-hydroxypyrrolidine-1,2-dicarboxylate (Compound 0103-55): To a mixture of 1-(tert-butyl)-2-methyl-4-hydroxy-4-(trimethylsilyl)ethynyl)pyrrolidine-1,2-dicarboxylate (0102-55) (12.2 g, 35.78 mmol, 1.0 eq) in methanol (50 mL) was added potassium carbonate (7.4 g, 53.67 mmol, 1.5 eq). The mixture was stirred at room temperature for 3 hours. The solvent was removed under reduced pressure. The residue was diluted with water (60 mL) and then extracted with ethyl acetate (50 mL x 3). The combined organic layers were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate and concentrated to give 1-(tert-butyl) 2-methyl 4-ethynyl-4-hydroxypyrrolidine-1,2-dicarboxylate (5.61 g, yield: 58%) as a brown oil. LCMS (ESI): m/z 270 [M+1] + ; TLC: Rf 0.4 (petroleum ether:ethyl acetate=3:1).

步骤23c:1-(叔丁基)2-甲基4-乙酰氧基-4-乙炔基吡咯烷-1,2-二羧酸酯(化合物0104-55)的制备:在0℃下,往1-(叔丁基)2-甲基4-乙炔基-4-羟基吡咯烷-1,2-二羧酸酯(0103-55)(5.61克,20.86毫摩尔,1.0当量)和N,N-二异丙基乙胺(9.2毫升,52.14毫摩尔,2.5当量)在二氯甲烷(50毫升)的混合物中逐滴加入乙酰氯(2.68毫升,35.46毫摩尔,1.7当量)。混合物升到室温然后搅拌1小时。混合物用水(50毫升)稀释然后用二氯甲烷(30毫升×3)萃取。合并的有机层用饱和食盐水(30毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯5:1),得到淡黄色油状物1-(叔丁基)2-甲基4-乙酰氧基-4-乙炔基吡咯烷-1,2-二羧酸酯(4.92克,收率:76%)。LCMS(ESI):m/z 312[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。Step 23c: Preparation of 1-(tert-butyl) 2-methyl 4-acetoxy-4-ethynylpyrrolidine-1,2-dicarboxylate (Compound 0104-55): To a mixture of 1-(tert-butyl) 2-methyl 4-ethynyl-4-hydroxypyrrolidine-1,2-dicarboxylate (0103-55) (5.61 g, 20.86 mmol, 1.0 eq) and N,N-diisopropylethylamine (9.2 mL, 52.14 mmol, 2.5 eq) in dichloromethane (50 mL) was added acetyl chloride (2.68 mL, 35.46 mmol, 1.7 eq) dropwise at 0°C. The mixture was warmed to room temperature and stirred for 1 hour. The mixture was diluted with water (50 mL) and then extracted with dichloromethane (30 mL x 3). The combined organic layers were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate and concentrated. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate 5: 1) to give 1-(tert-butyl) 2-methyl 4-acetoxy-4-ethynylpyrrolidine-1,2-dicarboxylate (4.92 g, yield: 76%) as a light yellow oil. LCMS (ESI): m/z 312 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 3: 1).

步骤23d:1-(叔丁基)2-甲基4-乙炔基-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(化合 物0105-55)的制备:在70℃下,往对甲氧基苄胺(4.16克,30.35毫摩尔,2.0当量)和氯化亚铜(0.15克,1.52毫摩尔,0.1当量)在四氢呋喃(50毫升)的混合物中逐滴加入1-(叔丁基)2-甲基4-乙酰氧基-4-乙炔基吡咯烷-1,2-二羧酸酯(0104-55)(4.72克,15.18毫摩尔,1.0当量)的四氢呋喃(30毫升)溶液。混合物在70℃的氮气氛围下搅拌1.5小时。混合物用水(50毫升)稀释然后用乙酸乙酯(30毫升×3)萃取。合并的有机层用饱和食盐水(30毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯8:1),得到淡黄色油状物1-(叔丁基)2-甲基4-乙炔基-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(2.06克,收率:35%)。LCMS(ESI):m/z 389[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=4:1)。Step 23d: 1-(tert-butyl)2-methyl-4-ethynyl-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (compound Preparation of compound 0105-55): To a mixture of p-methoxybenzylamine (4.16 g, 30.35 mmol, 2.0 eq.) and cuprous chloride (0.15 g, 1.52 mmol, 0.1 eq.) in tetrahydrofuran (50 ml) was added dropwise a solution of 1-(tert-butyl) 2-methyl 4-acetoxy-4-ethynylpyrrolidine-1,2-dicarboxylate (0104-55) (4.72 g, 15.18 mmol, 1.0 eq.) in tetrahydrofuran (30 ml) at 70°C. The mixture was stirred under a nitrogen atmosphere at 70°C for 1.5 hours. The mixture was diluted with water (50 ml) and then extracted with ethyl acetate (30 ml x 3). The combined organic layer was washed with saturated brine (30 ml), dried over anhydrous sodium sulfate and concentrated. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate 8: 1) to give 1-(tert-butyl) 2-methyl 4-ethynyl-4-((4-methoxybenzyl) amino) pyrrolidine-1,2-dicarboxylate (2.06 g, yield: 35%) as a pale yellow oil. LCMS (ESI): m/z 389 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 4: 1).

步骤23e:1-(叔丁基)2-甲基4-((2-氯-6-甲基吡啶-3-基)乙炔基)-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(化合物0108-55)的制备:往2-氯-3-碘-6-甲基吡啶(0107-1)(1.41克,5.57毫摩尔,1.05当量),双三苯基磷二氯化钯(0.187克,0.266毫摩尔,0.05当量)和碘化亚铜(51毫克,0.266毫摩尔,0.05当量)在三乙胺(25毫升)的混合物中加入1-(叔丁基)2-甲基4-乙炔基-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(0105-55)(2.06克,5.31毫摩尔,1.0当量)。混合物在氮气氛围下加热到80℃反应3.5小时。减压下除去溶剂。残余物用水(50毫升)稀释然后用乙酸乙酯(30毫升×3)萃取。合并的有机层用饱和食盐水(30毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚:乙酸乙酯6:1至2:1),得到淡黄色油状物1-(叔丁基)2-甲基4-((2-氯-6-甲基吡啶-3-基)乙炔基)-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(2.10克,收率:77%)。LCMS(ESI):m/z 514[M+1]+;TLC:Rf 0.4(石油醚:乙酸乙酯=3:1)。Step 23e: Preparation of 1-(tert-butyl)2-methyl 4-((2-chloro-6-methylpyridin-3-yl)ethynyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (Compound 0108-55): 2-chloro-3-iodo-6-methylpyridine (0107-1) (1.41 g, 5.57 mmol, 1.05 equiv), bistriphenylphosphine palladium dichloride (0 1-(tert-butyl)-2-methyl-4-ethynyl-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (0105-55) (2.06 g, 5.31 mmol, 1.0 eq.) was added to a mixture of triethylamine (25 ml) with 1-(tert-butyl)-2-methyl-4-ethynyl-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (0105-55) (2.06 g, 5.31 mmol, 1.0 eq.) and cuprous iodide (51 mg, 0.266 mmol, 0.05 eq.). The mixture was heated to 80°C under a nitrogen atmosphere for 3.5 hours. The solvent was removed under reduced pressure. The residue was diluted with water (50 ml) and then extracted with ethyl acetate (30 ml x 3). The combined organic layer was washed with saturated brine (30 ml), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether:ethyl acetate 6:1 to 2:1) to give a pale yellow oily substance 1-(tert-butyl)2-methyl 4-((2-chloro-6-methylpyridin-3-yl)ethynyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (2.10 g, yield: 77%). LCMS (ESI): m/z 514 [M+1] + ; TLC: Rf 0.4 (petroleum ether:ethyl acetate=3:1).

步骤23f:1-(叔丁基)2-甲基4-(2-(2-氯-6-甲基吡啶-3-基)乙基)-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(化合物0109-55)的制备:往1-(叔丁基)2-甲基4-((2-氯-6-甲基吡啶-3-基)乙炔基)-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(0108-55)(2.1克,4.09毫摩尔,1.0当量)在甲醇(20毫升)的混合物中加入二氧化铂(0.186克,0.82毫摩尔,0.2当量)。混合物在氢气球压力下室温搅拌40小时。混合物过滤。滤液减压浓缩,得到淡黄色油状物1-(叔丁基)2-甲基4-(2-(2-氯-6-甲基吡啶-3-基)乙基)-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(2.14克,粗品)。LCMS(ESI):m/z 518[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=2:1)。Step 23f: Preparation of 1-(tert-butyl)2-methyl 4-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (Compound 0109-55): To a mixture of 1-(tert-butyl)2-methyl 4-((2-chloro-6-methylpyridin-3-yl)ethynyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (0108-55) (2.1 g, 4.09 mmol, 1.0 eq) in methanol (20 ml) was added platinum dioxide (0.186 g, 0.82 mmol, 0.2 eq). The mixture was stirred at room temperature under hydrogen balloon pressure for 40 hours. The mixture was filtered. The filtrate was concentrated under reduced pressure to give 1-(tert-butyl)2-methyl 4-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (2.14 g, crude product) as a pale yellow oil. LCMS (ESI): m/z 518 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=2:1).

步骤23g:1’-(4-甲氧基苄基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0110-55)的制备:

Step 23g: Preparation of tert-butyl 1'-(4-methoxybenzyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0110-55):

往1-(叔丁基)2-甲基4-(2-(2-氯-6-甲基吡啶-3-基)乙基)-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(0109-55)(2.14克,4.14毫摩尔,1.0当量),醋酸钯(46毫克,0.21毫摩尔,0.05当量)和2-二环己基磷-2’,6’-二异丙氧基-1,1’-联苯(191毫克,0.41毫摩尔,0.1当量)在N-甲基吡咯烷酮(20毫升)的混合物中加入叔丁醇钠(0.795克,8.28毫摩尔,2.0当量)。混合物在微波反应器中加热至100℃反应1小时。混合物用水(40毫升)稀释。加入2M氢氧化钠溶液调节pH=12,然后混合物用甲基叔丁基醚(15毫升×2)洗涤。往水层加入1M稀盐酸溶液调节pH=7,然后用乙酸乙酯(20毫升×4)萃取。合并的有机层用饱和食盐水(30毫升)洗涤,经无水硫酸钠干燥并浓缩,得到棕色油状物1-(叔丁氧羰基)-1’-(4-甲氧基苄基)-7’-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-5-甲酸(2.13克,粗品)。LCMS(ESI):m/z 468[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=10:1)。To a mixture of 1-(tert-butyl)2-methyl 4-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (0109-55) (2.14 g, 4.14 mmol, 1.0 eq), palladium acetate (46 mg, 0.21 mmol, 0.05 eq) and 2-dicyclohexylphosphino-2',6'-diisopropoxy-1,1'-biphenyl (191 mg, 0.41 mmol, 0.1 eq) in N-methylpyrrolidone (20 ml) was added sodium tert-butoxide (0.795 g, 8.28 mmol, 2.0 eq). The mixture was heated to 100°C in a microwave reactor for 1 hour. The mixture was diluted with water (40 ml). 2M sodium hydroxide solution was added to adjust pH=12, and then the mixture was washed with methyl tert-butyl ether (15 ml×2). 1M dilute hydrochloric acid solution was added to the aqueous layer to adjust pH=7, and then extracted with ethyl acetate (20 ml×4). The combined organic layer was washed with saturated brine (30 ml), dried over anhydrous sodium sulfate and concentrated to give a brown oily substance 1-(tert-butyloxycarbonyl)-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-5-carboxylic acid (2.13 g, crude product). LCMS (ESI): m/z 468[M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=10:1).

往1-(叔丁氧羰基)-1’-(4-甲氧基苄基)-7’-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-5-甲酸(1.63克,3.49毫摩尔,1.0当量)在四氢呋喃(20毫升)的混合物中加入硼烷二甲硫醚络合物(10M的二甲硫醚溶液,0.87毫升,8.73毫摩尔,2.5当量)。混合物在氮气氛围下加热至65℃反应1.5小时。加入水(50毫升)淬灭反应。水层用乙酸乙酯(20毫升×3)萃取。合并的有机层用饱和食盐水(20毫升)洗涤,经无水硫酸钠干燥并浓缩,得到淡黄色油状物5-(羟甲基)-1’-(4-甲氧基苄基)-7’-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(1.39克,收率:88%)。LCMS(ESI):m/z 454[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=20:1)。Borane dimethyl sulfide complex (10 M dimethyl sulfide solution, 0.87 ml, 8.73 mmol, 2.5 eq) was added to a mixture of 1-(tert-butyloxycarbonyl)-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-5-carboxylic acid (1.63 g, 3.49 mmol, 1.0 eq) in tetrahydrofuran (20 ml). The mixture was heated to 65°C under a nitrogen atmosphere for 1.5 hours. Water (50 ml) was added to quench the reaction. The aqueous layer was extracted with ethyl acetate (20 ml x 3). The combined organic layers were washed with saturated brine (20 mL), dried over anhydrous sodium sulfate and concentrated to give a pale yellow oily substance, tert-butyl 5-(hydroxymethyl)-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (1.39 g, yield: 88%). LCMS (ESI): m/z 454 [M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=20:1).

在0℃下,往5-(羟甲基)-1’-(4-甲氧基苄基)-7’-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0.79克,1.74毫摩尔,1.0当量)和N,N-二异丙基乙胺(449毫克,3.48毫摩尔,2.0当量)在二氯甲烷(10毫升)的混合物中逐滴加入甲磺酰氯(0.2毫升,2.62毫摩尔,1.5当量)。混合物升到室温并搅拌22小时。混合物用水(30毫升)稀释然后用乙酸乙酯(30毫升×3)萃取。合并的有机层用饱和食盐水(30毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯3:1),得到淡黄色油状物1’-(4-甲氧基苄基)-7’-甲基-5-(((甲基磺酰基)氧基)甲基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0.545克,收率:59%)。LCMS(ESI):m/z 532[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=20:1)。To a mixture of tert-butyl 5-(hydroxymethyl)-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0.79 g, 1.74 mmol, 1.0 eq) and N,N-diisopropylethylamine (449 mg, 3.48 mmol, 2.0 eq) in dichloromethane (10 ml) was added methanesulfonyl chloride (0.2 ml, 2.62 mmol, 1.5 eq) dropwise at 0°C. The mixture was warmed to room temperature and stirred for 22 hours. The mixture was diluted with water (30 ml) and then extracted with ethyl acetate (30 ml x 3). The combined organic layer was washed with saturated brine (30 ml), dried over anhydrous sodium sulfate and concentrated. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate 3: 1) to give a pale yellow oily substance, tert-butyl 1'-(4-methoxybenzyl)-7'-methyl-5-(((methylsulfonyl)oxy)methyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0.545 g, yield: 59%). LCMS (ESI): m/z 532 [M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=20:1).

在0℃下,往1’-(4-甲氧基苄基)-7’-甲基-5-(((甲基磺酰基)氧基)甲基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0.545克,1.03毫摩尔,1.0当量)在四氢呋喃(6毫升)的混合物中逐滴加入三乙基硼氢化锂(1M的四氢呋喃溶液,3.09毫升,3.09毫摩尔,3.0当量)。 混合物升到室温然后搅拌6小时。加入水(30毫升)淬灭反应。水层用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(20毫升)洗涤,经无水硫酸钠干燥并浓缩,得到淡黄色油状物1’-(4-甲氧基苄基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(480毫克,粗品)。LCMS(ESI):m/z 438[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=5:1)。To a mixture of tert-butyl 1'-(4-methoxybenzyl)-7'-methyl-5-(((methylsulfonyl)oxy)methyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0.545 g, 1.03 mmol, 1.0 equiv) in tetrahydrofuran (6 mL) at 0°C was added lithium triethylborohydride (1 M in tetrahydrofuran, 3.09 mL, 3.09 mmol, 3.0 equiv) dropwise. The mixture was warmed to room temperature and then stirred for 6 hours. Water (30 ml) was added to quench the reaction. The aqueous layer was extracted with ethyl acetate (15 ml×3). The combined organic layer was washed with saturated brine (20 ml), dried over anhydrous sodium sulfate and concentrated to give a light yellow oily substance 1'-(4-methoxybenzyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (480 mg, crude product). LCMS (ESI): m/z 438[M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=5:1).

步骤23h:5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0112-55)的制备:往1’-(4-甲氧基苄基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0110-55)(450毫克,1.03毫摩尔,1.0当量)的二氯甲烷(6毫升)混合物中加入三氟乙酸(1.5毫升)。混合物在室温下搅拌2小时。减压下除去溶剂。残余物溶于水(10毫升)和四氢呋喃(10毫升)。加入碳酸钠固体调节pH=9。加入二碳酸二叔丁酯(449毫克,2.06毫摩尔,2.0当量),然后将混合物在室温下搅拌1小时。混合物用水(20毫升)稀释,然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(20毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯3:1),得到淡黄色油状物5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(193毫克,收率:59%)。LCMS(ESI):m/z 318[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=1:1)。Step 23h: Preparation of tert-butyl 5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0112-55): To a mixture of tert-butyl 1'-(4-methoxybenzyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0110-55) (450 mg, 1.03 mmol, 1.0 equiv) in dichloromethane (6 mL) was added trifluoroacetic acid (1.5 mL). The mixture was stirred at room temperature for 2 hours. The solvent was removed under reduced pressure. The residue was dissolved in water (10 mL) and tetrahydrofuran (10 mL). Solid sodium carbonate was added to adjust the pH to 9. Di-tert-butyl dicarbonate (449 mg, 2.06 mmol, 2.0 eq) was added, and the mixture was stirred at room temperature for 1 hour. The mixture was diluted with water (20 mL), and then extracted with ethyl acetate (15 mL×3). The combined organic layer was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate 3:1) to give a pale yellow oily substance tert-butyl 5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (193 mg, yield: 59%). LCMS (ESI): m/z 318 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 1:1).

步骤23i:6’-溴-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0115-55)的制备:在0℃下,往5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0112-55)(193毫克,0.61毫摩尔,1.0当量)的二氯甲烷(5毫升)混合物中加入二溴海因(96毫克,0.34毫摩尔,0.55当量)。混合物在0℃下搅拌20分钟。加入饱和亚硫酸钠水溶液(20毫升)淬灭反应。水层用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(20毫升)洗涤,经无水硫酸钠干燥并浓缩,得到淡黄色油状物6’-溴-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(255毫克,粗品)。LCMS(ESI):m/z 396[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。Step 23i: Preparation of tert-butyl 6'-bromo-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0115-55): To a mixture of tert-butyl 5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0112-55) (193 mg, 0.61 mmol, 1.0 equiv) in dichloromethane (5 mL) was added dibromohydantoin (96 mg, 0.34 mmol, 0.55 equiv) at 0°C. The mixture was stirred at 0°C for 20 minutes. Saturated aqueous sodium sulfite solution (20 mL) was added to quench the reaction. The aqueous layer was extracted with ethyl acetate (15 mL x 3). The combined organic layers were washed with saturated brine (20 mL), dried over anhydrous sodium sulfate and concentrated to give tert-butyl 6'-bromo-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (255 mg, crude) as a pale yellow oil. LCMS (ESI): m/z 396 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=3:1).

步骤23j:(1-(叔丁氧羰基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(化合物0117-55)的制备:往6’-溴-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0115-55)(242毫克,0.61毫摩尔,1.0当量),Xphos-Pd-G2(9.6毫克,0.012毫摩尔,0.02当量),2-二环己基膦-2’,4’,6’-三异丙基联苯(8.7毫克,0.018毫摩尔,0.03当量),乙酸钾(424毫克,4.32毫摩尔,2.5当量)和叔丁醇钠(4.2毫克,0.043毫摩尔,0.025当量)在甲醇(6毫升)和乙二醇(0.6毫升)的混合物中加入四羟基二硼(137毫克,1.532毫摩尔,2.5当量)。混合物在氮气氛围下加热至55℃反应1.5小时。减压下除去溶剂。残余物用水(20毫升)稀释。加入2M氢氧化钠溶液调节pH=12,然后混合物用甲基叔丁基醚(10毫升×2)洗涤。往水层加入2M稀盐酸溶液调节pH=8,然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水 (15毫升)洗涤,经无水硫酸钠干燥并浓缩,得到无色油状物(1-(叔丁氧羰基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(165毫克,收率:75%)。LCMS(ESI):m/z 362[M+1]+;TLC:Rf 0.3(二氯甲烷:甲醇=10:1)。Step 23j: Preparation of (1-(tert-Butyloxycarbonyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (Compound 0117-55): tert-butyl 6'-bromo-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0115-55) (242 mg, 0.61 mmol, 1.0 equiv), Xphos-Pd-G 2 (9.6 mg, 0.012 mmol, 0.02 eq), 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (8.7 mg, 0.018 mmol, 0.03 eq), potassium acetate (424 mg, 4.32 mmol, 2.5 eq) and sodium tert-butoxide (4.2 mg, 0.043 mmol, 0.025 eq) were added to a mixture of methanol (6 ml) and ethylene glycol (0.6 ml). Tetrahydroxydiboron (137 mg, 1.532 mmol, 2.5 eq) was added. The mixture was heated to 55° C. under nitrogen atmosphere for 1.5 hours. The solvent was removed under reduced pressure. The residue was diluted with water (20 ml). 2M sodium hydroxide solution was added to adjust pH=12, and then the mixture was washed with methyl tert-butyl ether (10 ml×2). 2M dilute hydrochloric acid solution was added to the aqueous layer to adjust the pH to 8, and then extracted with ethyl acetate (15 ml x 3). The combined organic layers were washed with saturated brine. (15 ml), dried over anhydrous sodium sulfate and concentrated to give a colorless oily substance (1-(tert-butyloxycarbonyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridin]-6'-yl)boronic acid (165 mg, yield: 75%). LCMS (ESI): m/z 362 [M+1] + ; TLC: Rf 0.3 (dichloromethane:methanol=10:1).

步骤23k:5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0119-55)的制备:往(1-(叔丁氧羰基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(0117-55)(165毫克,0.457毫摩尔,1.0当量),Xphos-Pd-G2(10.8毫克,0.014毫摩尔,0.03当量),2-二环己基膦-2’,4’,6’-三异丙基联苯(6.7毫克,0.014毫摩尔,0.03当量)和碳酸钠(145毫克,1.37毫摩尔,3.0当量)在甲苯(2.5毫升),乙醇(2.5毫升)和水(1毫升)的混合物中加入2-溴嘧啶(76毫克,0.480毫摩尔,1.05当量)。混合物在氮气氛围下加热至55℃反应4小时。混合物用水(20毫升)稀释然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(10毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(乙酸乙酯:石油醚=5:1)纯化,得到淡黄色固体5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(109毫克,收率:60%)。LCMS(ESI):m/z 396[M+1]+;TLC:Rf 0.5(乙酸乙酯:石油醚=5:1)。Step 23k: Preparation of tert-butyl 5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (Compound 0119-55): To (1-(tert-butyloxycarbonyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (0117-55) (165 mg, 0.457 mmol, 1.0 equiv.), Xphos-Pd-G2 (10.8 mg, 0.014 mmol, 0.03 equiv.), 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (6.7 mg, 0.014 mmol, 0.03 equiv.) and sodium carbonate (145 mg, 1.37 mmol, 3.0 equiv.) were added to a mixture of toluene (2.5 ml), ethanol (2.5 ml) and water (1 ml). The mixture was heated to 55°C for 4 hours under a nitrogen atmosphere. The mixture was diluted with water (20 ml) and then extracted with ethyl acetate (15 ml x 3). The combined organic layer was washed with saturated brine (10 ml), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (ethyl acetate:petroleum ether=5:1) to give a pale yellow solid tert-butyl 5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (109 mg, yield: 60%). LCMS (ESI): m/z 396 [M+1] + ; TLC: Rf 0.5 (ethyl acetate:petroleum ether=5:1).

步骤23l:5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(化合物0121-55)的制备:5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0119-55)(109毫克,0.276毫摩尔,1.0当量)在氯化氢/甲醇溶液(4M溶液,2毫升)的混合物在室温下搅拌3小时。减压下除去溶剂。残余物在真空下干燥,得到淡黄色固体5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(105毫克,粗品)。LCMS(ESI):m/z 296[M+1]+;TLC:Rf 0.3(二氯甲烷:甲醇=10:1)。Step 231: Preparation of 5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0121-55): A mixture of tert-butyl 5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0119-55) (109 mg, 0.276 mmol, 1.0 eq) in hydrogen chloride/methanol solution (4M solution, 2 ml) was stirred at room temperature for 3 hours. The solvent was removed under reduced pressure. The residue was dried under vacuum to give a pale yellow solid 5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (105 mg, crude product). LCMS (ESI): m/z 296 [M+1] + ; TLC: Rf 0.3 (dichloromethane:methanol=10:1).

步骤23m:(2R)-1-(5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’-H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-2-(5-氟-2-甲氧基吡啶-4-基)丙烷-1-酮(化合物55)的制备:往(R)-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(0406-7)(27.5毫克,0.138毫摩尔,1.0当量),苯并三氮唑-N,N,N’,N’-四甲基脲六氟磷酸盐(63毫克,0.166毫摩尔,1.2当量),1-羟基苯并三唑(20毫克,0.152毫摩尔,1.1当量)和N,N-二异丙基乙胺(89毫克,0.69毫摩尔,5.0当量)在二氯甲烷(3毫升)的混合物中加入5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0121-55)(51毫克,0.138毫摩尔,1.0当量)。混合物在室温下搅拌过夜。混合物用水(20毫升)稀释然后用乙酸乙酯(10毫升×3)萃取。合并的有机层用饱和食盐水(15毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(二氯甲烷:甲醇:氨水=80:3:0.8)纯化,得到白色固体(2R)-1-(5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’-H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-2-(5-氟-2-甲氧基吡啶-4-基)丙烷-1-酮(51毫克,收率:78%)。LCMS(ESI):m/z 477[M+1]+;TLC:Rf 0.5(二氯甲烷: 甲醇=20:1)。1H NMR(500MHz,MeOD)δ8.82(dt,J=9.0,5.0Hz,2H),7.88(ddd,J=127.8,64.9,35.9Hz,2H),7.34–7.24(m,1H),6.93–6.66(m,1H),4.43–4.06(m,2H),3.87(dd,J=19.4,11.6Hz,3H),3.75–3.45(m,2H),2.93–2.68(m,2H),2.63–2.53(m,3H),2.38–2.13(m,1H),2.01–1.61(m,3H),1.45–1.31(m,6H).Step 23m: Preparation of (2R)-1-(5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)-2-(5-fluoro-2-methoxypyridin-4-yl)propan-1-one (Compound 55): to (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0406-7) (27.5 mg, 0.138 mmol, 1.0 equiv), benzotriazole-N,N,N',N'-tetramethyluronium hexafluoro Phosphate (63 mg, 0.166 mmol, 1.2 eq), 1-hydroxybenzotriazole (20 mg, 0.152 mmol, 1.1 eq) and N,N-diisopropylethylamine (89 mg, 0.69 mmol, 5.0 eq) in dichloromethane (3 ml) was added 5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0121-55) (51 mg, 0.138 mmol, 1.0 eq). The mixture was stirred at room temperature overnight. The mixture was diluted with water (20 ml) and then extracted with ethyl acetate (10 ml x 3). The combined organic layer was washed with saturated brine (15 ml), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (dichloromethane:methanol:aqueous ammonia=80:3:0.8) to give a white solid (2R)-1-(5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-2-(5-fluoro-2-methoxypyridin-4-yl)propan-1-one (51 mg, yield: 78%). LCMS (ESI): m/z 477 [M+1] + ; TLC: Rf 0.5 (dichloromethane: Methanol=20:1). 1H NMR(500MHz,MeOD)δ8.82(dt,J=9.0,5.0Hz,2H),7.88(ddd,J=127.8,64.9,35.9Hz,2H),7.34–7.24(m,1H),6.93–6.66(m,1H),4.43–4.06(m,2H), 3.87(dd,J=19.4,11.6Hz,3H),3.75–3.45(m,2H),2.93–2.68(m,2H),2.6 3–2.53(m,3H),2.38–2.13(m,1H),2.01–1.61(m,3H),1.45–1.31(m,6H).

实施例24:(R)-1-((3R,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’-H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-2-(5-氟-2-甲氧基吡啶-4-基)丙烷-1-酮(化合物100)的制备(按照方案四和五线路制备)Example 24: Preparation of (R)-1-((3R,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-2-(5-fluoro-2-methoxypyridin-4-yl)propan-1-one (Compound 100) (Prepared according to Schemes 4 and 5)

步骤24a:(3S,5S)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0113-100)的制备:
Step 24a: Preparation of (3S,5S)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0113-100):

在-78℃下,往乙炔基三甲基硅烷(7.27克,73.99毫摩尔,1.2当量)在四氢呋喃(70毫升)的混合物中逐滴加入正丁基锂(2.5M的己烷溶液,25.90毫升,64.74毫摩尔,1.05当量)。混合物在-78℃的氮气氛围下搅拌1小时。逐滴加入(S)-2-乙酰氧基-4-氧代吡咯烷-1-甲酸叔丁酯(15.0克,61.66毫摩尔,1.0当量)的四氢呋喃(50毫升)溶液。混合物升到室温然后搅拌过夜。加入饱和氯化铵溶液(50毫升)淬灭反应。水层用乙酸乙酯(50毫升×3)萃取。合并的有机层用饱和食盐水(50毫升)洗涤,经无水硫酸钠干燥并浓缩,得到棕色固体1-(叔丁基)2-甲基(2R)-4-羟基-4-((三甲基甲硅烷基)乙炔基)吡咯烷-1,2-二羧酸酯(17.5克,收率:83%)。LCMS(ESI):m/z 342[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=60:1)。To a mixture of ethynyltrimethylsilane (7.27 g, 73.99 mmol, 1.2 eq) in tetrahydrofuran (70 ml) was added n-butyllithium (2.5 M in hexane, 25.90 ml, 64.74 mmol, 1.05 eq) dropwise at -78°C. The mixture was stirred at -78°C under a nitrogen atmosphere for 1 hour. A solution of (S)-tert-butyl 2-acetoxy-4-oxopyrrolidine-1-carboxylate (15.0 g, 61.66 mmol, 1.0 eq) in tetrahydrofuran (50 ml) was added dropwise. The mixture was warmed to room temperature and then stirred overnight. Saturated ammonium chloride solution (50 ml) was added to quench the reaction. The aqueous layer was extracted with ethyl acetate (50 ml x 3). The combined organic layers were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate and concentrated to give a brown solid 1-(tert-butyl) 2-methyl (2R)-4-hydroxy-4-((trimethylsilyl)ethynyl)pyrrolidine-1,2-dicarboxylate (17.5 g, yield: 83%). LCMS (ESI): m/z 342 [M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=60:1).

往1-(叔丁基)2-甲基(2R)-4-羟基-4-((三甲基甲硅烷基)乙炔基)吡咯烷-1,2-二羧酸酯(17.5克,51.32毫摩尔,1.0当量)在甲醇(45毫升)的混合物中加入碳酸钾(10.6克,76.98毫摩尔,1.5当量)。混合物在室温下搅拌1小时。减压下除去溶剂。残余物用水(80毫升)稀释然后用乙酸乙酯(50毫升×4)萃取。合并的有机层用饱和食盐水(50毫升)洗涤,经无水硫酸钠干燥并浓缩,得到棕色油状物1-(叔丁基)2-甲基(2R)-4-乙炔基-4-羟基吡咯烷-1,2-二羧酸酯(9.74克,收率:71%)。LCMS(ESI):m/z 270[M+1]+;TLC:Rf 0.4(石油醚:乙酸乙酯=3:1)。Potassium carbonate (10.6 g, 76.98 mmol, 1.5 eq.) was added to a mixture of 1-(tert-butyl)2-methyl(2R)-4-hydroxy-4-((trimethylsilyl)ethynyl)pyrrolidine-1,2-dicarboxylate (17.5 g, 51.32 mmol, 1.0 eq.) in methanol (45 ml). The mixture was stirred at room temperature for 1 hour. The solvent was removed under reduced pressure. The residue was diluted with water (80 ml) and then extracted with ethyl acetate (50 ml×4). The combined organic layer was washed with saturated brine (50 ml), dried over anhydrous sodium sulfate and concentrated to give 1-(tert-butyl)2-methyl(2R)-4-ethynyl-4-hydroxypyrrolidine-1,2-dicarboxylate (9.74 g, yield: 71%) as a brown oil. LCMS (ESI): m/z 270 [M+1] + ; TLC: Rf 0.4 (petroleum ether:ethyl acetate=3:1).

在0℃下,往1-(叔丁基)2-甲基(2R)-4-乙炔基-4-羟基吡咯烷-1,2-二羧酸酯(9.74克,36.21 毫摩尔,1.0当量)和N,N-二异丙基乙胺(11.68克,90.52毫摩尔,2.5当量)在二氯甲烷(100毫升)的混合物中逐滴加入乙酰氯(4.65毫升,61.56毫摩尔,1.7当量)。混合物升到室温然后搅拌1.5小时。混合物用水(60毫升)稀释然后用二氯甲烷(30毫升×3)萃取。合并的有机层用饱和食盐水(30毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯4:1),得到淡黄色油状物1-(叔丁基)2-甲基(2R)-4-乙酰氧基-4-乙炔基吡咯烷-1,2-二羧酸酯(9.50克,收率:84%)。LCMS(ESI):m/z 312[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。At 0°C, 1-(tert-butyl) 2-methyl (2R)-4-ethynyl-4-hydroxypyrrolidine-1,2-dicarboxylate (9.74 g, 36.21 To a mixture of 1,4-diisopropylethylamine (11.68 g, 90.52 mmol, 2.5 eq.) and diisopropylethylamine (11.68 g, 90.52 mmol, 2.5 eq.) in dichloromethane (100 ml) was added acetyl chloride (4.65 ml, 61.56 mmol, 1.7 eq.) dropwise. The mixture was warmed to room temperature and then stirred for 1.5 hours. The mixture was diluted with water (60 ml) and then extracted with dichloromethane (30 ml x 3). The combined organic layer was washed with saturated brine (30 ml), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate 4:1) to give 1-(tert-butyl) 2-methyl (2R)-4-acetoxy-4-ethynylpyrrolidine-1,2-dicarboxylate (9.50 g, yield: 84%) as a pale yellow oil. LCMS (ESI): m/z 312 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=3:1).

在70℃下,往对甲氧基苄胺(20.29克,147.9毫摩尔,2.0当量)和氯化亚铜(0.732克,7.40毫摩尔,0.1当量)在四氢呋喃(250毫升)的混合物中逐滴加入1-(叔丁基)2-甲基(2R)-4-乙酰氧基-4-乙炔基吡咯烷-1,2-二羧酸酯(23克,73.95毫摩尔,1.0当量)的四氢呋喃(100毫升)溶液。混合物在70℃的氮气氛围下搅拌1.5小时。混合物用水(150毫升)稀释然后用乙酸乙酯(100毫升×2)萃取。合并的有机层用饱和食盐水(100毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯8:1),得到淡黄色油状物1-(叔丁基)2-甲基(2R)-4-乙炔基-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(9.26克,收率:32%)。LCMS(ESI):m/z 389[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=4:1)。To a mixture of p-methoxybenzylamine (20.29 g, 147.9 mmol, 2.0 eq.) and cuprous chloride (0.732 g, 7.40 mmol, 0.1 eq.) in tetrahydrofuran (250 ml) was added dropwise a solution of 1-(tert-butyl) 2-methyl (2R)-4-acetoxy-4-ethynylpyrrolidine-1,2-dicarboxylate (23 g, 73.95 mmol, 1.0 eq.) in tetrahydrofuran (100 ml) at 70°C. The mixture was stirred under a nitrogen atmosphere at 70°C for 1.5 hours. The mixture was diluted with water (150 ml) and then extracted with ethyl acetate (100 ml x 2). The combined organic layer was washed with saturated brine (100 ml), dried over anhydrous sodium sulfate and concentrated. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate 8: 1) to give 1-(tert-butyl) 2-methyl (2R)-4-ethynyl-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (9.26 g, yield: 32%) as a light yellow oil. LCMS (ESI): m/z 389 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 4: 1).

往2-氯-3-碘-6-甲基吡啶(0107-1)(6.33克,25.06毫摩尔,1.05当量),双三苯基磷二氯化钯(0.838克,1.19毫摩尔,0.05当量)和碘化亚铜(0.227g,1.19毫摩尔,0.05当量)在三乙胺(100毫升)的混合物中加入1-(叔丁基)2-甲基(2R)-4-乙炔基-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(9.26克,23.87毫摩尔,1.0当量)。混合物在氮气氛围下加热到80℃反应8小时。减压下除去溶剂。残余物用水(50毫升)稀释然后用乙酸乙酯(50毫升×3)萃取。合并的有机层用饱和食盐水(50毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚:乙酸乙酯6:1 to 3:1),得到淡黄色油状物1-(叔丁基)2-甲基(2R)-4-((2-氯-6-甲基吡啶-3-基)乙炔基)-4-(4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(10.02克,收率:82%)。LCMS(ESI):m/z 514[M+1]+;TLC:Rf 0.4(石油醚:乙酸乙酯=3:1)。To a mixture of 2-chloro-3-iodo-6-methylpyridine (0107-1) (6.33 g, 25.06 mmol, 1.05 eq.), bistriphenylphosphine palladium dichloride (0.838 g, 1.19 mmol, 0.05 eq.) and cuprous iodide (0.227 g, 1.19 mmol, 0.05 eq.) in triethylamine (100 ml) was added 1-(tert-butyl) 2-methyl (2R)-4-ethynyl-4-((4-methoxybenzyl)amino) pyrrolidine-1,2-dicarboxylate (9.26 g, 23.87 mmol, 1.0 eq.). The mixture was heated to 80°C under nitrogen atmosphere for 8 hours. The solvent was removed under reduced pressure. The residue was diluted with water (50 ml) and then extracted with ethyl acetate (50 ml x 3). The combined organic layer was washed with saturated brine (50 ml), dried over anhydrous sodium sulfate and concentrated. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate 6:1 to 3:1) to obtain a light yellow oily substance 1-(tert-butyl) 2-methyl (2R)-4-((2-chloro-6-methylpyridin-3-yl)ethynyl)-4-(4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (10.02 g, yield: 82%). LCMS (ESI): m/z 514 [M+1] + ; TLC: Rf 0.4 (petroleum ether: ethyl acetate = 3:1).

往1-(叔丁基)2-甲基(2R)-4-((2-氯-6-甲基吡啶-3-基)乙炔基)-4-(4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(10.02克,19.53毫摩尔,1.0当量)在甲醇(80毫升)的混合物中加入二氧化铂(0.887克,39.06毫摩尔,0.2当量)。混合物在氢气球压力下室温搅拌40小时。混合物过滤。滤液减压浓缩,得到淡黄色油状物1-(叔丁基)2-甲基(2R)-4-(2-(2-氯-6-甲基吡啶-3-基)乙基)-4-((4-甲氧基苄基)氨基)吡咯烷-1,2-二羧酸酯(9.56克,粗品)。LCMS(ESI):m/z 518[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=2:1)。Platinum dioxide (0.887 g, 39.06 mmol, 0.2 eq.) was added to a mixture of 1-(tert-butyl)2-methyl (2R)-4-((2-chloro-6-methylpyridin-3-yl)ethynyl)-4-(4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (10.02 g, 19.53 mmol, 1.0 eq.) in methanol (80 ml). The mixture was stirred at room temperature for 40 hours under hydrogen balloon pressure. The mixture was filtered. The filtrate was concentrated under reduced pressure to give 1-(tert-butyl)2-methyl (2R)-4-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (9.56 g, crude) as a pale yellow oil. LCMS (ESI): m/z 518 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=2:1).

在0℃下,往1-(叔丁基)2-甲基(2R)-4-(2-(2-氯-6-甲基吡啶-3-基)乙基)-4-((4-甲氧基苄基) 氨基)吡咯烷-1,2-二羧酸酯(9.0克,17.41毫摩尔,1.0当量)在四氢呋喃(60毫升)的混合物中缓慢加入四氢铝锂(2.65克,69.63毫摩尔,4.0当量)。混合物升到室温并搅拌3小时。混合物冷却到0℃。加入水(10.5毫升)和15%氢氧化钠溶液(2.65毫升)。混合物升到室温并搅拌10分钟。混合物用乙酸乙酯(50毫升)稀释然后过滤。滤液用饱和食盐水(50毫升)洗涤,经无水硫酸钠干燥并浓缩,得到淡黄色油状物(2R)-4-(2-(2-氯-6-甲基吡啶-3-基)乙基)-2-(羟甲基)-4-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(8.05克,收率:95%)。LCMS(ESI):m/z 490[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=2:1)。At 0°C, 1-(tert-butyl)2-methyl(2R)-4-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-4-((4-methoxybenzyl) To a mixture of (2R)-4-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-2-(hydroxymethyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1,2-dicarboxylate (9.0 g, 17.41 mmol, 1.0 eq.) in tetrahydrofuran (60 ml) was slowly added lithium aluminum tetrahydride (2.65 g, 69.63 mmol, 4.0 eq.). The mixture was warmed to room temperature and stirred for 3 hours. The mixture was cooled to 0°C. Water (10.5 ml) and 15% sodium hydroxide solution (2.65 ml) were added. The mixture was warmed to room temperature and stirred for 10 minutes. The mixture was diluted with ethyl acetate (50 ml) and then filtered. The filtrate was washed with saturated brine (50 ml), dried over anhydrous sodium sulfate and concentrated to give a light yellow oily substance (2R)-4-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-2-(hydroxymethyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylic acid tert-butyl ester (8.05 g, yield: 95%). LCMS (ESI): m/z 490 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=2:1).

往(2R)-4-(2-(2-氯-6-甲基吡啶-3-基)乙基)-2-(羟甲基)-4-((4-甲氧基苄基)氨基)吡咯烷-1-甲酸叔丁酯(8.05克,16.46毫摩尔,1.0当量),醋酸钯(0.184克,0.82毫摩尔,0.05当量)和2-二环己基磷-2’,6’-二异丙氧基-1,1’-联苯(0.77g,1.65毫摩尔,0.1当量)在N-甲基吡咯烷酮(50毫升)的混合物中加入叔丁醇钠(3.16克,32.92毫摩尔,2.0当量)。混合物在微波反应器中加热至100℃反应1小时。混合物用水(20毫升)和四氢呋喃(20毫升)稀释。加入二碳酸二叔丁酯(4.67克,21.40毫摩尔,1.3当量),然后将混合物在室温下搅拌1小时。混合物用水(60毫升)稀释,然后用乙酸乙酯(40毫升×3)萃取。合并的有机层用饱和食盐水(50毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯8∶1至3:1),得到淡黄色油状物(3S,5R)-5-(羟甲基)-1’-(4-甲氧基苄基)-7’-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(833毫克,收率:11%)。LCMS(ESI):m/z 454[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=2:1)。To a mixture of tert-butyl (2R)-4-(2-(2-chloro-6-methylpyridin-3-yl)ethyl)-2-(hydroxymethyl)-4-((4-methoxybenzyl)amino)pyrrolidine-1-carboxylate (8.05 g, 16.46 mmol, 1.0 eq), palladium acetate (0.184 g, 0.82 mmol, 0.05 eq) and 2-dicyclohexylphosphino-2',6'-diisopropoxy-1,1'-biphenyl (0.77 g, 1.65 mmol, 0.1 eq) in N-methylpyrrolidone (50 ml) was added sodium tert-butoxide (3.16 g, 32.92 mmol, 2.0 eq). The mixture was heated to 100° C. in a microwave reactor for 1 hour. The mixture was diluted with water (20 ml) and tetrahydrofuran (20 ml). Di-tert-butyl dicarbonate (4.67 g, 21.40 mmol, 1.3 eq) was added, and the mixture was stirred at room temperature for 1 hour. The mixture was diluted with water (60 ml), and then extracted with ethyl acetate (40 ml×3). The combined organic layer was washed with saturated brine (50 ml), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (petroleum ether: ethyl acetate 8:1 to 3:1) to give a pale yellow oily substance (3S, 5R)-5-(hydroxymethyl)-1'-(4-methoxybenzyl)-7'-methyl-3', 4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (833 mg, yield: 11%). LCMS (ESI): m/z 454 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 2:1).

在0℃下,往(3S,5R)-5-(羟甲基)-1’-(4-甲氧基苄基)-7’-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(833毫克,1.84毫摩尔,1.0当量)和N,N-二异丙基乙胺(475毫克,3.68毫摩尔,2.0当量)在二氯甲烷(10毫升)的混合物中逐滴加入甲磺酰氯(0.21毫升,2.76毫摩尔,1.5当量)。混合物升到室温并搅拌6小时。混合物用水(30毫升)稀释然后用乙酸乙酯(15毫升×4)萃取。合并的有机层用饱和食盐水(20毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯3:1),得到淡黄色油状物(3S,5R)-1’-(4-甲氧基苄基)-7’-甲基-5-(((甲基磺酰基)氧基)甲基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0.92克,收率:94%)。LCMS(ESI):m/z 532[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=2:1)。To a mixture of (3S,5R)-5-(hydroxymethyl)-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (833 mg, 1.84 mmol, 1.0 eq) and N,N-diisopropylethylamine (475 mg, 3.68 mmol, 2.0 eq) in dichloromethane (10 ml) was added methanesulfonyl chloride (0.21 ml, 2.76 mmol, 1.5 eq) dropwise at 0°C. The mixture was warmed to room temperature and stirred for 6 hours. The mixture was diluted with water (30 ml) and then extracted with ethyl acetate (15 ml x 4). The combined organic layer was washed with saturated brine (20 ml), dried over anhydrous sodium sulfate and concentrated. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate 3: 1) to give a pale yellow oily substance (3S, 5R)-1'-(4-methoxybenzyl)-7'-methyl-5-(((methylsulfonyl)oxy)methyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0.92 g, yield: 94%). LCMS (ESI): m/z 532 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 2: 1).

在0℃下,往(3S,5R)-1’-(4-甲氧基苄基)-7’-甲基-5-(((甲基磺酰基)氧基)甲基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0.92克,1.73毫摩尔,1.0当量)在四氢呋喃(10毫升)的混合物中逐滴加入三乙基硼氢化锂(1M的四氢呋喃溶液,6.93毫升,6.93毫摩尔,4.0当量)。混合物升到室温然后搅拌过夜。加入水(30毫升)淬灭反应。水层用乙酸乙酯(15毫升 ×3)萃取。合并的有机层用饱和食盐水(20毫升)洗涤,经无水硫酸钠干燥并浓缩,得到淡黄色油状物(3S,5S)-1’-(4-甲氧基苄基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(800毫克,粗品)。LCMS(ESI):m/z 438[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=5:1)。To a mixture of tert-butyl (3S,5R)-1'-(4-methoxybenzyl)-7'-methyl-5-(((methylsulfonyl)oxy)methyl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (0.92 g, 1.73 mmol, 1.0 eq) in tetrahydrofuran (10 mL) was added dropwise lithium triethylborohydride (1 M in tetrahydrofuran, 6.93 mL, 6.93 mmol, 4.0 eq) at 0°C. The mixture was warmed to room temperature and then stirred overnight. Water (30 mL) was added to quench the reaction. The aqueous layer was washed with ethyl acetate (15 mL) ×3). The combined organic layer was washed with saturated brine (20 ml), dried over anhydrous sodium sulfate and concentrated to give a light yellow oily substance (3S,5S)-1'-(4-methoxybenzyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (800 mg, crude product). LCMS (ESI): m/z 438 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=5:1).

往(3S,5S)-1’-(4-甲氧基苄基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(756毫克,1.73毫摩尔,1.0当量)在二氯甲烷(12毫升)的混合物中加入三氟乙酸(3毫升)。混合物在室温下搅拌2小时。减压下除去溶剂。残余物溶于水(10毫升)和四氢呋喃(10毫升)。加入碳酸钠固体调节pH=9。加入二碳酸二叔丁酯(566毫克,2.60毫摩尔,1.5当量),然后将混合物在室温下搅拌1小时。混合物用水(20毫升)稀释,然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(20毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(石油醚∶乙酸乙酯3:1),得到淡黄色油状物(3S,5S)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(548毫克,收率:100%)。LCMS(ESI):m/z 318[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=1:1)。To a mixture of (3S,5S)-1'-(4-methoxybenzyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (756 mg, 1.73 mmol, 1.0 eq) in dichloromethane (12 ml) was added trifluoroacetic acid (3 ml). The mixture was stirred at room temperature for 2 hours. The solvent was removed under reduced pressure. The residue was dissolved in water (10 ml) and tetrahydrofuran (10 ml). Solid sodium carbonate was added to adjust pH=9. Di-tert-butyl dicarbonate (566 mg, 2.60 mmol, 1.5 eq) was added, and the mixture was stirred at room temperature for 1 hour. The mixture was diluted with water (20 ml), and then extracted with ethyl acetate (15 ml×3). The combined organic layer was washed with saturated brine (20 ml), dried over anhydrous sodium sulfate and concentrated. The residue was separated by column chromatography on silica gel (petroleum ether: ethyl acetate 3:1) to give a pale yellow oily substance (3S,5S)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (548 mg, yield: 100%). LCMS (ESI): m/z 318 [M+1] + ; TLC: Rf 0.5 (petroleum ether: ethyl acetate = 1:1).

步骤24b:(3S,5S)-6’-溴-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0116-100)的制备:在0℃下,往(3S,5S)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0113-100)(548毫克,1.73毫摩尔,1.0当量)在二氯甲烷(10毫升)的混合物中加入二溴海因(272毫克,0.95毫摩尔,0.55当量)。混合物在0℃下搅拌0.5小时。加入饱和亚硫酸钠水溶液(15毫升)淬灭反应。水层用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(20毫升)洗涤,经无水硫酸钠干燥并浓缩,得到淡黄色油状物(3S,5S)-6’-溴-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(754毫克,粗品)。LCMS(ESI):m/z 396[M+1]+;TLC:Rf 0.5(石油醚:乙酸乙酯=3:1)。Step 24b: Preparation of (3S,5S)-6'-bromo-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0116-100): To a mixture of (3S,5S)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0113-100) (548 mg, 1.73 mmol, 1.0 equiv) in dichloromethane (10 mL) at 0° C. was added dibromohydantoin (272 mg, 0.95 mmol, 0.55 equiv). The mixture was stirred at 0° C. for 0.5 h. The reaction was quenched by the addition of saturated aqueous sodium sulfite (15 mL). The aqueous layer was extracted with ethyl acetate (15 ml x 3). The combined organic layers were washed with saturated brine (20 ml), dried over anhydrous sodium sulfate and concentrated to give a pale yellow oily substance (3S,5S)-6'-bromo-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (754 mg, crude product). LCMS (ESI): m/z 396 [M+1] + ; TLC: Rf 0.5 (petroleum ether:ethyl acetate=3:1).

步骤24c:((3S,5S)-1-(叔丁氧羰基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(化合物0118-100)的制备:往(3S,5S)-6’-溴-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0116-100)(685毫克,1.73毫摩尔,1.0当量),Xphos-Pd-G2(27毫克,0.035毫摩尔,0.02当量),2-二环己基膦-2’,4’,6’-三异丙基联苯(25毫克,0.052毫摩尔,0.03当量),乙酸钾(424毫克,4.32毫摩尔,2.5当量)和叔丁醇钠(4.2毫克,0.043毫摩尔,0.025当量)在甲醇(10毫升)和乙二醇(1毫升)的混合物中加入四羟基二硼(388毫克,4.32毫摩尔,2.5当量)。混合物在氮气氛围下加热至55℃反应2小时。减压下除去溶剂。残余物用水(30毫升)稀释。加入2M氢氧化钠溶液调节pH=12,然后混合物用甲基叔丁基醚(10毫升×2)洗涤。往水层加入2M稀盐酸溶液调节pH=8,然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(15毫升)洗涤,经无水硫酸钠干燥并浓缩,得到白色固体((3S,5S)-1-(叔丁氧羰基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(299毫克,收率:48%)。 LCMS(ESI):m/z 362[M+1]+;TLC:Rf 0.3(二氯甲烷:甲醇=10:1)。Step 24c: Preparation of ((3S,5S)-1-(tert-butyloxycarbonyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (Compound 0118-100): to (3S,5S)-6'-bromo-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0116-100) (685 mg, 1.73 mmol, 1.0 equiv) , Xphos-Pd-G2 (27 mg, 0.035 mmol, 0.02 eq), 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (25 mg, 0.052 mmol, 0.03 eq), potassium acetate (424 mg, 4.32 mmol, 2.5 eq) and sodium tert-butoxide (4.2 mg, 0.043 mmol, 0.025 eq) were added to a mixture of methanol (10 ml) and ethylene glycol (1 ml). Tetrahydroxydiboron (388 mg, 4.32 mmol, 2.5 eq) was added. The mixture was heated to 55°C under a nitrogen atmosphere for 2 hours. The solvent was removed under reduced pressure. The residue was diluted with water (30 ml). 2M sodium hydroxide solution was added to adjust pH = 12, and then the mixture was washed with methyl tert-butyl ether (10 ml × 2). 2M dilute hydrochloric acid solution was added to the aqueous layer to adjust pH=8, and then extracted with ethyl acetate (15 ml×3). The combined organic layer was washed with saturated brine (15 ml), dried over anhydrous sodium sulfate and concentrated to obtain a white solid ((3S,5S)-1-(tert-butyloxycarbonyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (299 mg, yield: 48%). LCMS (ESI): m/z 362 [M+1] + ; TLC: Rf 0.3 (dichloromethane:methanol=10:1).

步骤24d:(3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(化合物0120-100)的制备:往((3S,5S)-1-(叔丁氧羰基)-5,7’-二甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-6’-基)硼酸(0118-100)(299毫克,0.828毫摩尔,1.0当量),Xphos-Pd-G2(19.5毫克,0.025毫摩尔,0.03当量),2-二环己基膦-2’,4’,6’-三异丙基联苯(11.9毫克,0.025毫摩尔,0.03当量)和碳酸钠(263毫克,2.48毫摩尔,3.0当量)在甲苯(5毫升),乙醇(5毫升)和水(2毫升)的混合物中加入2-溴嘧啶(151毫克,0.95毫摩尔,1.15当量)。混合物在氮气氛围下加热至55℃反应5小时。减压下除去溶剂。残余物用水(20毫升)稀释然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(15毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(二氯甲烷∶甲醇50∶1至25:1),得到淡黄色固体(3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(270毫克,收率:83%)。LCMS(ESI):m/z 396[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=20:1)。Step 24d: Preparation of (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (Compound 0120-100): To ((3S,5S)-1-(tert-butyloxycarbonyl)-5,7'-dimethyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-6'-yl)boronic acid (0118-100) (299 mg, 0. 828 mmol, 1.0 eq.), Xphos-Pd-G2 (19.5 mg, 0.025 mmol, 0.03 eq.), 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (11.9 mg, 0.025 mmol, 0.03 eq.) and sodium carbonate (263 mg, 2.48 mmol, 3.0 eq.) were added to a mixture of toluene (5 ml), ethanol (5 ml) and water (2 ml). The mixture was heated to 55°C for 5 hours under a nitrogen atmosphere. The solvent was removed under reduced pressure. The residue was diluted with water (20 ml) and then extracted with ethyl acetate (15 ml x 3). The combined organic layer was washed with saturated brine (15 ml), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (dichloromethane: methanol 50: 1 to 25: 1) to give a pale yellow solid (3S, 5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (270 mg, yield: 83%). LCMS (ESI): m/z 396 [M+1] + ; TLC: Rf 0.5 (dichloromethane: methanol = 20: 1).

步骤24e:(3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(化合物0122-100)的制备:将(3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-甲酸叔丁酯(0120-100)(270毫克,0.68毫摩尔,1.0当量)在氯化氢/甲醇溶液(4M溶液,2毫升)的混合物在室温下搅拌2小时。减压下除去溶剂。残余物在真空下干燥,得到淡黄色固体(3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(270毫克,粗品)。LCMS(ESI):m/z 296[M+1]+;TLC:Rf 0.3(二氯甲烷:甲醇=10:1)。Step 24e: Preparation of (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (Compound 0122-100): A mixture of (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (0120-100) (270 mg, 0.68 mmol, 1.0 eq) in hydrogen chloride/methanol solution (4M solution, 2 ml) was stirred at room temperature for 2 hours. The solvent was removed under reduced pressure. The residue was dried under vacuum to give a pale yellow solid (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (270 mg, crude product). LCMS (ESI): m/z 296 [M+1] + ; TLC: Rf 0.3 (dichloromethane:methanol=10:1).

步骤24f:(R)-1-((3R,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’-H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-2-(5-氟-2-甲氧基吡啶-4-基)丙烷-1-酮(化合物100)的制备:往(R)-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(0406-7)(95毫克,0.476毫摩尔,1.0当量),6-氯苯并三氮唑-1,1,3,3-四甲基脲六氟磷酸酯(217毫克,0.524毫摩尔,1.1当量)和N,N-二异丙基乙胺(196毫克,1.523毫摩尔,3.2当量)在二氯甲烷(7毫升)的混合物中加入(3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-100)(175毫克,0.476毫摩尔,1.0当量)。混合物在室温下搅拌1.5小时。混合物用水(30毫升)稀释然后用乙酸乙酯(15毫升×3)萃取。合并的有机层用饱和食盐水(15毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物通过制备薄层色谱(二氯甲烷:甲醇:氨水=80:2.5:0.8)纯化,得到白色固体(R)-1-((3R,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’-H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)-2-(5-氟-2-甲氧基吡啶-4-基)丙烷-1-酮(152毫克,收率:67%)。LCMS(ESI):m/z 477[M+1]+;TLC:Rf 0.5(二氯甲烷:甲醇=15:1)。1H NMR(500MHz,MeOD)δ8.81(d,J=4.9Hz,2H),7.93(d,J=31.5Hz,2H),7.31(t,J=4.9Hz, 1H),6.70(d,J=4.8Hz,1H),4.46–4.30(m,1H),4.16(d,J=6.9Hz,1H),3.88(d,J=9.9Hz,3H),3.73(d,J=10.8Hz,1H),3.24(d,J=11.0Hz,1H),2.90–2.68(m,2H),2.57(d,J=19.3Hz,3H),2.33(dd,J=12.7,8.2Hz,1H),1.89(dd,J=14.9,7.6Hz,2H),1.69(dd,J=13.2,8.7Hz,1H),1.40(d,J=6.9Hz,3H),1.31(d,J=6.1Hz,3H).Step 24f: Preparation of (R)-1-((3R,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)-2-(5-fluoro-2-methoxypyridin-4-yl)propan-1-one (Compound 100): to (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0406-7) (95 mg, 0.476 mmol, 1.0 equiv), 6-chlorobenzotriazole-1, To a mixture of 1,3,3-tetramethyluronium hexafluorophosphate (217 mg, 0.524 mmol, 1.1 eq) and N,N-diisopropylethylamine (196 mg, 1.523 mmol, 3.2 eq) in dichloromethane (7 ml) was added (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-100) (175 mg, 0.476 mmol, 1.0 eq). The mixture was stirred at room temperature for 1.5 hours. The mixture was diluted with water (30 ml) and then extracted with ethyl acetate (15 ml×3). The combined organic layer was washed with saturated brine (15 ml), dried over anhydrous sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography (dichloromethane:methanol:aqueous ammonia=80:2.5:0.8) to give a white solid (R)-1-((3R,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-2-(5-fluoro-2-methoxypyridin-4-yl)propan-1-one (152 mg, yield: 67%). LCMS (ESI): m/z 477 [M+1] + ; TLC: Rf 0.5 (dichloromethane:methanol=15:1). 1 H NMR (500MHz, MeOD) δ8.81 (d, J = 4.9 Hz, 2H), 7.93 (d, J = 31.5 Hz, 2H), 7.31 (t, J = 4.9 Hz, 1H),6.70(d,J=4.8Hz,1H),4.46–4.30(m,1H),4.16(d,J=6.9Hz,1H),3.88(d,J =9.9Hz,3H),3.73(d,J=10.8Hz,1H),3.24(d,J=11.0Hz,1H),2.90–2.68(m,2H), 2.57(d,J=19.3Hz,3H), 2.33(dd,J=12.7,8.2Hz,1H), 1.89(dd,J=14.9,7.6Hz, 2H), 1.69 (dd, J=13.2, 8.7Hz, 1H), 1.40 (d, J=6.9Hz, 3H), 1.31 (d, J=6.1Hz, 3H).

实施例25:4-(1-((3S,5S)-5,7'-二甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-基)-1-氧代丙烷-2-基)-6-甲氧基烟腈(化合物96)的制备(按照方案四线路制备):Example 25: Preparation of 4-(1-((3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-1-oxopropane-2-yl)-6-methoxynicotinonitrile (Compound 96) (prepared according to Scheme 4):

将(3S,5S)-5,7'-二甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-1λ 2-螺[吡咯烷-3,2'-[1,8]萘啶]二盐酸盐(0122-100)(88毫克,0.24毫摩尔,1.0当量),2-(5-氰基-2-甲氧基吡啶-4-基)丙酸(0405-11)(59毫克,0.29毫摩尔,1.2当量),N,N-二异丙基乙胺(186毫克,1.44毫摩尔,6.0当量)和2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(110毫克,0.29毫摩尔,1.2当量)的二氯甲烷(4毫升)混合物搅拌一小时。混合物用水稀释并用二氯甲烷萃取,有机相用饱和食盐水洗涤,用无水硫酸钠干燥并浓缩。残余物经厚制备薄层色谱(洗脱剂:二氯甲烷/甲醇/氨水=80/4.5/0.8)纯化,得到淡黄色固体产物4-(1-((3S,5S)-5,7'-二甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'H-螺[吡咯烷-3,2'-[1,8]萘啶]-1-基)-1-氧代丙烷-2-基)-6-甲氧基烟腈(20毫克,收率:17.24%)。LCMS(ESI):m/z=484[M+H]+1H NMR(500MHz,MeOD)δ8.82(dd,J=14.2,4.9Hz,2H),8.32(d,J=184.6Hz,1H),7.84(d,J=22.5Hz,1H),7.31(dt,J=9.7,4.9Hz,1H),6.89(d,J=31.8Hz,1H),4.45–4.32(m,1H),4.13(dd,J=77.8,6.8Hz,1H),3.98(d,J=6.6Hz,3H),3.71(dd,J=74.4,10.3Hz,1H),3.51(dd,J=40.4,10.8Hz,1H),2.91–2.82(m,2H),2.52(d,J=13.6Hz,3H),2.09–1.66(m,4H),1.49–1.34(m,6H).A mixture of (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3', 4' -dihydro- 1'H -1λ2-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-100) (88 mg, 0.24 mmol, 1.0 eq), 2-(5-cyano-2-methoxypyridin-4-yl)propanoic acid (0405-11) (59 mg, 0.29 mmol, 1.2 eq), N,N-diisopropylethylamine (186 mg, 1.44 mmol, 6.0 eq) and 2-(7-azabenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (110 mg, 0.29 mmol, 1.2 eq) in dichloromethane (4 ml) was stirred for one hour. The mixture was diluted with water and extracted with dichloromethane, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by thick preparative thin layer chromatography (eluent: dichloromethane/methanol/ammonia water=80/4.5/0.8) to obtain a light yellow solid product 4-(1-((3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)-1-oxopropane-2-yl)-6-methoxynicotinonitrile (20 mg, yield: 17.24%). LCMS (ESI): m/z=484[M+H] + . 1 H NMR(500MHz,MeOD)δ8.82(dd,J=14.2,4.9Hz,2H),8.32(d,J=184.6Hz,1H),7.84(d,J=22.5 Hz,1H),7.31(dt,J=9.7,4.9Hz,1H),6.89(d,J=31.8Hz,1H),4.45–4.32(m,1H),4.13(dd,J= 77.8,6.8Hz,1H),3.98(d,J=6.6Hz,3H),3.71(dd,J=74.4,10.3Hz,1H),3.51(dd,J=40.4,10 .8Hz,1H),2.91–2.82(m,2H),2.52(d,J=13.6Hz,3H),2.09–1.66(m,4H),1.49–1.34(m,6H).

实施例26:(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((3S,5R)-5-(氟甲基)-7'-甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'-吡咯烷-3,2'-[1,8]萘吡啶]-1-基)丙-1-酮(化合物103)的制备:
Example 26: Preparation of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 103):

步骤26a:(9H-芴-9-基)甲基(3S,5R)-5-(羟甲基)-7'-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-羧酸盐的制备:将(3S,5R)-5-(羟甲基)-1'-(4-甲氧基苄基)-7'-甲基-3’,4’-二氢-1'-H-吡咯并[3,2'-[1,8]萘啶]-1-羧酸叔丁酯(500毫克,1.10毫摩尔,1.0当量)的氯化氢/二氧六环(5毫升)溶液在室温下搅拌一小时。将反应液在真空下浓缩。残留物加入四氢呋喃(10毫升),氯甲酸-9-芴基甲酯(424毫克,1.66毫摩尔,1.5当量),三乙胺(335毫克,3.30毫摩尔,3.0 当量)。然后混合物在室温下搅拌两小时。反应液加水稀释并用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=10/1至3/1)纯化,得到黄色固体产物(9H-芴-9-基)甲基(3S,5R)-5-(羟甲基)-7'-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-羧酸盐(380毫克,收率:75.55%)。LCMS(ESI):m/z=456[M+1]+.Step 26a: Preparation of (9H-fluoren-9-yl)methyl (3S,5R)-5-(hydroxymethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate: A solution of (3S,5R)-5-(hydroxymethyl)-1'-(4-methoxybenzyl)-7'-methyl-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (500 mg, 1.10 mmol, 1.0 eq) in hydrogen chloride/dioxane (5 mL) was stirred at room temperature for one hour. The reaction was concentrated under vacuum. The residue was added with tetrahydrofuran (10 ml), 9-fluorenylmethyl chloroformate (424 mg, 1.66 mmol, 1.5 eq), triethylamine (335 mg, 3.30 mmol, 3.0 Equivalent). The mixture was then stirred at room temperature for two hours. The reaction solution was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate = 10/1 to 3/1) to obtain a yellow solid product (9H-fluoren-9-yl)methyl (3S, 5R)-5-(hydroxymethyl)-7'-methyl-3', 4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (380 mg, yield: 75.55%). LCMS (ESI): m/z = 456 [M+1] + .

步骤26b:(9H-芴-9-基)甲基(3S,5R)-5-(氟甲基)-7'-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-羧酸盐的制备:氮气保护下,在0℃下,向(9H-芴-9-基)甲基(3S,5R)-5-(羟甲基)-7'-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-羧酸盐(380毫克,0.83毫摩尔,1.0当量)的二氯甲烷(5毫升)溶液中加入二乙胺基三氟化硫(269毫克,1.67毫摩尔,2.0当量),混合物在0℃下搅拌1.0小时。反应液加水稀释并用二氯甲烷萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(二氯甲烷/甲醇=100/1至30/1)纯化,得到黄色固体产物(9H-芴-9-基)甲基(3S,5R)-5-(氟甲基)-7'-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-羧酸盐(278毫克,收率:72.97%)。LCMS(ESI):m/z=458[M+1]+.Step 26b: Preparation of (9H-fluoren-9-yl)methyl (3S,5R)-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate: Under nitrogen protection, to a solution of (9H-fluoren-9-yl)methyl (3S,5R)-5-(hydroxymethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (380 mg, 0.83 mmol, 1.0 equiv) in dichloromethane (5 ml) at 0°C was added diethylaminosulfur trifluoride (269 mg, 1.67 mmol, 2.0 equiv), and the mixture was stirred at 0°C for 1.0 hour. The reaction solution was diluted with water and extracted with dichloromethane, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (dichloromethane/methanol=100/1 to 30/1) to obtain a yellow solid product (9H-fluoren-9-yl)methyl (3S,5R)-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (278 mg, yield: 72.97%). LCMS (ESI): m/z=458[M+1] + .

步骤26c:(9H-芴-9-基)甲基(3S,5R)-6'-溴-5-(氟甲基)-7'-甲基-3’,4’-二氢-1'-吡咯烷-3,2'-[1,8]萘吡啶]-1-羧酸盐的制备:氮气保护下,在0℃下,向(9H-芴-9-基)甲基(3S,5R)-5-(氟甲基)-7'-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-羧酸盐(278毫克,0.61毫摩尔,1.0当量)的二氯甲烷(5毫升)溶液中滴加1,3-二溴-5,5-二甲基咪唑烷-2,4-二酮(87毫克,0.303毫摩尔,0.5当量),混合物在0℃下搅拌1.0小时。反应液用碳酸钠溶液淬灭并用二氯甲烷萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=5/1至2/1)纯化,得到黄色固体产物(9H-芴-9-基)甲基(3S,5R)-6'-溴-5-(氟甲基)-7'-甲基-3’,4’-二氢-1'-吡咯烷-3,2'-[1,8]萘吡啶]-1-羧酸盐(230毫克,收率:70.77%)。LCMS(ESI):m/z=537[M+1]+.Step 26c: Preparation of (9H-fluoren-9-yl)methyl (3S,5R)-6'-bromo-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'-pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate: Under nitrogen protection at 0°C, add (9H-fluoren-9-yl)methyl (3S,5R)-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'-pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate. 1,3-Dibromo-5,5-dimethylimidazolidine-2,4-dione (87 mg, 0.303 mmol, 0.5 eq) was added dropwise to a solution of 4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (278 mg, 0.61 mmol, 1.0 eq) in dichloromethane (5 ml), and the mixture was stirred at 0°C for 1.0 hour. The reaction solution was quenched with sodium carbonate solution and extracted with dichloromethane, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate=5/1 to 2/1) to obtain a yellow solid product (9H-fluoren-9-yl)methyl (3S,5R)-6'-bromo-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'-pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (230 mg, yield: 70.77%). LCMS (ESI): m/z=537[M+1] + .

步骤26d:(3S,5R)-6'-溴-5-(氟甲基)-7'-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-羧酸叔丁酯的制备:将(9H-芴-9-基)甲基(3S,5R)-6'-溴-5-(氟甲基)-7'-甲基-3’,4’-二氢-1'-吡咯烷-3,2'-[1,8]萘吡啶]-1-羧酸盐(230毫克,0.43毫摩尔,1.0当量)的哌啶/二氯甲烷(1毫升/4毫升)混合溶液在室温下搅拌1小时。将反应液在真空下浓缩。残留物加入二碳酸二叔丁酯(188毫克,0.86毫摩尔,2.0当量)和四氢呋喃(5毫升)。混合物在室温下搅拌一小时。反应液加水稀释并用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=5/1至2/1)纯化,得到黄色油状产物(3S,5R)-6'-溴-5-(氟甲基)-7'-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-羧酸叔丁酯(150毫克,收率:84.75%)。LCMS(ESI):m/z=414[M+1]+.Step 26d: Preparation of tert-butyl (3S,5R)-6'-bromo-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate: A solution of (9H-fluoren-9-yl)methyl (3S,5R)-6'-bromo-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'-pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylate (230 mg, 0.43 mmol, 1.0 eq) in piperidine/dichloromethane (1 mL/4 mL) was stirred at room temperature for 1 hour. The reaction solution was concentrated under vacuum. Di-tert-butyl dicarbonate (188 mg, 0.86 mmol, 2.0 eq) and tetrahydrofuran (5 mL) were added to the residue. The mixture was stirred at room temperature for one hour. The reaction solution was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate = 5/1 to 2/1) to obtain a yellow oily product (3S, 5R)-6'-bromo-5-(fluoromethyl)-7'-methyl-3', 4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (150 mg, yield: 84.75%). LCMS (ESI): m/z = 414 [M+1] + .

步骤26e:(3S,5R)-5-(氟甲基)-7'-甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'-H-吡咯并[3,2'-[1,8]萘啶]-1-羧酸叔丁酯的制备:氮气保护下,将(3S,5R)-6'-溴-5-(氟甲基)-7'-甲基-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-羧酸叔丁酯(150毫克,0.36毫摩尔,1.0当量),2-(三丁基锡基)嘧啶 (268毫克,0.72毫摩尔,2.0当量),碘化亚铜(7毫克,0.036毫摩尔,0.1当量),甲烷磺酸(2-二环己基膦基-2',4',6'-三-异丙基-1,1'-联苯基)(2'-氨基-1,1'-联苯-2-基)钯(II)(16毫克,0.018毫摩尔,0.05当量),2-二环己基磷-2’,4’,6’-三异丙基联苯(18毫克,0.036毫摩尔,0.1当量),氟化铯(111毫克,0.72毫摩尔,3.0当量)的N,N-二甲基甲酰胺(5毫升)混合物在100℃下微波加热搅拌1.5小时。反应液加水稀释并用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=5/1至2/1)纯化,得到黄色固体产物(3S,5R)-5-(氟甲基)-7'-甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'-H-吡咯并[3,2'-[1,8]萘啶]-1-羧酸叔丁酯(64毫克,收率:42.67%)。LCMS(ESI):m/z=414[M+1]+.Step 26e: Preparation of (3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester: Under nitrogen protection, (3S,5R)-6'-bromo-5-(fluoromethyl)-7'-methyl-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (150 mg, 0.36 mmol, 1.0 equiv), 2-(tributyltinyl)pyrimidine (268 mg, 0.72 mmol, 2.0 eq), cuprous iodide (7 mg, 0.036 mmol, 0.1 eq), methanesulfonic acid (2-dicyclohexylphosphino-2',4',6'-tri-isopropyl-1,1'-biphenyl)(2'-amino-1,1'-biphenyl-2-yl) palladium (II) (16 mg, 0.018 mmol, 0.05 eq), 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (18 mg, 0.036 mmol, 0.1 eq), cesium fluoride (111 mg, 0.72 mmol, 3.0 eq) in N,N-dimethylformamide (5 ml) was heated in a microwave at 100° C. for 1.5 hours. The reaction solution was diluted with water and extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate = 5/1 to 2/1) to obtain a yellow solid product (3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (64 mg, yield: 42.67%). LCMS (ESI): m/z = 414 [M+1] + .

步骤26f:(3S,5R)-5-(氟甲基)-7'-甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'-H-吡咯并[3,2'-[1,8]萘啶]二盐酸盐的制备:(3S,5R)-5-(氟甲基)-7'-甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'-H-吡咯并[3,2'-[1,8]萘啶]-1-羧酸叔丁酯(64毫克,0.15毫摩尔,1.0当量)在氯化氢二氧六环溶液(4M溶液,2.5毫升)的混合物在室温下搅拌1.0小时,减压除去溶剂并在真空下干燥,残留物不经过进一步纯化,直接用于下一步。LCMS(ESI):m/z=314[M+1]+Step 26f: Preparation of (3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine] dihydrochloride: A mixture of (3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine]-1-carboxylic acid tert-butyl ester (64 mg, 0.15 mmol, 1.0 eq) in hydrogen chloride dioxane solution (4 M solution, 2.5 mL) was stirred at room temperature for 1.0 h, the solvent was removed under reduced pressure and dried under vacuum, and the residue was used directly in the next step without further purification. LCMS (ESI): m/z=314[M+1] + .

步骤26g:(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((3S,5R)-5-(氟甲基)-7'-甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'-吡咯烷-3,2'-[1,8]萘吡啶]-1-基)丙-1-酮(化合物103)的制备:向(R)-2-(5-氟-2-甲氧基吡啶-4-基)丙酸(0406-7)(31毫克,0.155毫摩尔,1.0当量),(3S,5R)-5-(氟甲基)-7'-甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'-H-吡咯并[3,2'-[1,8]萘啶]二盐酸盐(粗品),N,N-二异丙基乙胺(50毫克,0.388毫摩尔,2.5当量)的二氯甲烷(3毫升)溶液中加入6-氯苯并三氮唑-1,1,3,3-四甲基脲六氟磷酸酯(77毫克,0.186毫摩尔,1.2当量),混合物在室温下搅拌1.0小时。反应液加水稀释并用二氯甲烷萃取,将有机相干燥并减压浓缩。残留物用制备薄层色谱(二氯甲烷/甲醇=20/1)纯化,得到黄色固体产物(R)-2-(5-氟-2-甲氧基吡啶-4-基)-1-((3S,5R)-5-(氟甲基)-7'-甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1'-吡咯烷-3,2'-[1,8]萘吡啶]-1-基)丙-1-酮(32毫克,收率42.11%)。LCMS(ESI):m/z=495[M+1]+.1H NMR(500MHz,MeOD)δ8.82(t,J=6.2Hz,2H),7.96(s,1H),7.86(s,1H),7.30(t,J=4.9Hz,1H),6.69(d,J=4.9Hz,1H),4.93(dd,J=9.3,3.2Hz,1H),4.61–4.37(m,2H),4.20(q,J=6.8Hz,1H),3.88(d,J=13.8Hz,3H),3.76(d,J=10.7Hz,1H),3.13(d,J=10.8Hz,1H),2.93–2.72(m,2H),2.56(d,J=16.9Hz,3H),2.31–2.15(m,1H),2.10(dd,J=13.3,9.0Hz,1H),1.90(t,J=6.4Hz,2H),1.41(d,J=6.9Hz,3H).Step 26g: Preparation of (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)propan-1-one (Compound 103): To (R)-2-(5-fluoro-2-methoxypyridin-4-yl)propanoic acid (0406-7) (31 mg, 0.155 mmol, 1.0 equiv), (3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)propan-1-one was added. 6-Chlorobenzotriazole-1,1,3,3-tetramethyluronium hexafluorophosphate (77 mg, 0.186 mmol, 1.2 eq.) was added to a solution of 5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-H-pyrrolo[3,2'-[1,8]naphthyridine] dihydrochloride (crude product) and N,N-diisopropylethylamine (50 mg, 0.388 mmol, 2.5 eq.) in dichloromethane (3 ml), and the mixture was stirred at room temperature for 1.0 hour. The reaction solution was diluted with water and extracted with dichloromethane, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography (dichloromethane/methanol=20/1) to give a yellow solid product (R)-2-(5-fluoro-2-methoxypyridin-4-yl)-1-((3S,5R)-5-(fluoromethyl)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'-pyrrolidin-3,2'-[1,8]naphthyridin]-1-yl)propan-1-one (32 mg, yield 42.11%). LCMS (ESI): m/z=495[M+1] + . 1 H NMR (500MHz, MeOD) δ8.82(t,J=6.2Hz,2H),7.96(s,1H),7.86(s,1H),7.30(t,J=4.9Hz,1H),6.69( d,J=4.9Hz,1H),4.93(dd,J=9.3,3.2Hz,1H),4.61–4.37(m,2H),4.20(q,J=6.8Hz,1H),3.88(d,J=1 3.8Hz,3H),3.76(d,J=10.7Hz,1H),3.13(d,J=10.8Hz,1H),2.93–2.72(m,2H),2.56(d,J=16.9Hz, 3H),2.31–2.15(m,1H),2.10(dd,J=13.3,9.0Hz,1H),1.90(t,J=6.4Hz,2H),1.41(d,J=6.9Hz,3H).

实施例27:2-(5-氯-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-硫酮(化合物10)的制备(按照方案四和五线路制备):Example 27: Preparation of 2-(5-chloro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (Compound 10) (prepared according to Schemes 4 and 5):

步骤27a:2-(5-氯-2-甲氧基吡啶-4-基)乙酸甲酯(0409-10)的制备:氮气保护下,向-30℃的二异丙胺(1.23克,12.22毫摩尔,3.2当量)的四氢呋喃(5毫升)溶液中滴加正丁基锂的正己 烷溶液(2.5摩尔,4.7毫升,11.84毫摩尔,3.1当量)。混合物在-30℃下搅拌反应1小时后,往混合物中滴加5-氯-2-甲氧基-4-甲基吡啶(0408-10)(600毫克,3.82毫摩尔,0.2当量)的四氢呋喃溶液(1毫升)。混合物在-30℃下搅拌反应1小时后,滴加碳酸二甲酯(1.03克,11.46毫摩尔,3当量)。加完后,混合物升温到室温并搅拌1小时。反应液用水淬灭并用乙酸乙酯萃取,有机相用饱和食盐水洗,硫酸钠干燥,过滤并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=10/1)纯化,得到2-(5-氯-2-甲氧基吡啶-4-基)乙酸甲酯(600毫克,收率:73.08%)为透明油状产物。LCMS(ESI):m/z=216[M+1]+.Step 27a: Preparation of methyl 2-(5-chloro-2-methoxypyridin-4-yl)acetate (0409-10): Under nitrogen protection, to a solution of diisopropylamine (1.23 g, 12.22 mmol, 3.2 eq.) in tetrahydrofuran (5 ml) at -30°C was added dropwise n-butyllithium in n-hexyl After the mixture was stirred at -30°C for 1 hour, a tetrahydrofuran solution (1 ml) of 5-chloro-2-methoxy-4-methylpyridine (0408-10) (600 mg, 3.82 mmol, 0.2 eq) was added dropwise to the mixture. After the mixture was stirred at -30°C for 1 hour, dimethyl carbonate (1.03 g, 11.46 mmol, 3 eq) was added dropwise. After the addition, the mixture was warmed to room temperature and stirred for 1 hour. The reaction solution was quenched with water and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried with sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate = 10/1) to obtain 2-(5-chloro-2-methoxypyridin-4-yl)acetic acid methyl ester (600 mg, yield: 73.08%) as a transparent oily product. LCMS (ESI): m/z=216[M+1] + .

步骤27b:2-(5-氯-2-甲氧基吡啶-4-基)丙酸甲酯(化合物0410-10)的制备:在0℃下,向2-(5-氯-2-甲氧基吡啶-4-基)乙酸甲酯(0409-10)(550毫克,2.56毫摩尔,1当量)的四氢呋喃(5毫升)溶液中滴加双(三甲基硅基)氨基锂(1M四氢呋喃溶液,2.86毫升,2.86毫摩尔,1.2当量)。将混合物在0℃下搅拌1小时,然后在0℃逐滴加入碘甲烷(327毫克,2.3毫摩尔,0.9eq)的四氢呋喃(1毫升)溶液。混合物在0℃下搅拌1小时。然后将混合物倒入饱和氯化铵水溶液中,并用乙酸乙酯萃取。有机相用饱和食盐水洗,硫酸钠干燥,过滤并减压浓缩。残留物用硅胶柱(石油醚/乙酸乙酯=10/1)纯化,得到2-(5-氯-2-甲氧基吡啶-4-基)丙酸甲酯(320毫克,收率:54.61%)为黄色油状产物。LCMS(ESI):m/z=230[M+1]+.Step 27b: Preparation of methyl 2-(5-chloro-2-methoxypyridin-4-yl)propanoate (Compound 0410-10): To a solution of methyl 2-(5-chloro-2-methoxypyridin-4-yl)acetate (0409-10) (550 mg, 2.56 mmol, 1 eq) in tetrahydrofuran (5 ml) at 0°C was added lithium bis(trimethylsilyl)amide (1 M in tetrahydrofuran, 2.86 ml, 2.86 mmol, 1.2 eq). The mixture was stirred at 0°C for 1 hour, and then a solution of iodomethane (327 mg, 2.3 mmol, 0.9 eq) in tetrahydrofuran (1 ml) was added dropwise at 0°C. The mixture was stirred at 0°C for 1 hour. The mixture was then poured into a saturated aqueous ammonium chloride solution and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate = 10/1) to obtain methyl 2-(5-chloro-2-methoxypyridin-4-yl)propanoate (320 mg, yield: 54.61%) as a yellow oily product. LCMS (ESI): m/z = 230 [M+1] + .

步骤27c:2-(5-氯-2-甲氧基吡啶-4-基)丙酸(化合物0405-10)的制备:向2-(5-氯-2-甲氧基吡啶-4-基)丙酸甲酯(0410-10)(320毫克,1.4毫摩尔,1当量)的四氢呋喃/水(5/1毫升)溶液中加入氢氧化锂一水合物(12毫克,0.28毫摩尔,0.2当量)。将混合物在室温下搅拌过夜,然后减压浓缩。将含水残留物倒入水中,并用甲基叔丁基醚洗涤。然后通过加入3M盐酸将水层的pH调节至4,并用乙酸乙酯萃取。有机相用饱和食盐水洗,硫酸钠干燥,过滤并减压浓缩。残留物用硅胶柱(洗脱剂:二氯甲烷/甲醇=20/1)纯化,得到2-(5-氯-2-甲氧基吡啶-4-基)丙酸(106毫克,收率:35.33%)为透明油状产物。LCMS(ESI):m/z=216[M+1]+.Step 27c: Preparation of 2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid (Compound 0405-10): To a solution of methyl 2-(5-chloro-2-methoxypyridin-4-yl)propanoate (0410-10) (320 mg, 1.4 mmol, 1 eq) in tetrahydrofuran/water (5/1 ml) was added lithium hydroxide monohydrate (12 mg, 0.28 mmol, 0.2 eq). The mixture was stirred at room temperature overnight and then concentrated under reduced pressure. The aqueous residue was poured into water and washed with methyl tert-butyl ether. The pH of the aqueous layer was then adjusted to 4 by the addition of 3M hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with saturated brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column (eluent: dichloromethane/methanol = 20/1) to obtain 2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid (106 mg, yield: 35.33%) as a transparent oily product. LCMS (ESI): m/z = 216 [M+1] + .

步骤27d:2-(5-氯-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(化合物0412-10)的制备:向2-(5-氯-2-甲氧基吡啶-4-基)丙酸(0405-10)(18毫克,0.085毫摩尔,1当量),(S)-7'-甲基-6'-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-1)(30毫克,0.085毫摩尔,1当量)N,N-二异丙基乙胺(22毫克,0.17毫摩尔,2当量)的二氯甲烷(5毫升)溶液中加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(36毫克,0.094毫摩尔,1.1当量),混合物在室温下搅拌1.0小时。反应液加水稀释并用二氯甲烷萃取,将有机相干燥并减压浓缩。残留物用制备薄层色谱(二氯甲烷/甲醇=20/1)纯化,得到2-(5-氯-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(12毫克,收率:30%)为黄色油状产物。 LCMS(ESI):m/z=479[M+1]+.Step 27d: Preparation of 2-(5-chloro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 0412-10): 2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid (0405-10) (18 mg, 0.085 mmol, 1 eq), (S)-7'-methyl-6'-(pyrimidin-2 2-(7-azobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (36 mg, 0.094 mmol, 1.1 eq) was added to a solution of 1,2-dihydro- ... The residue was purified by preparative thin layer chromatography (dichloromethane/methanol=20/1) to give 2-(5-chloro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (12 mg, yield: 30%) as a yellow oily product. LCMS (ESI): m/z=479[M+1] + .

步骤27e:2-(5-氯-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-硫酮(化合物10)的制备:将2-(5-氯-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(0412-10)(36毫克,0.075毫摩尔,1.0当量)和劳森试剂(36毫克,0.09毫摩尔,1.2当量)的混合物在125℃下微波加热搅拌45分钟。反应冷却至室温并用水淬灭。反应液用乙酸乙酯萃取,将有机相干燥并减压浓缩。残留物用制备薄层色谱(二氯甲烷/甲醇/氨水=80/4.5/0.8)纯化,得到2-(5-氯-2-甲氧基吡啶-4-基)-1-((S)-7’-甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-硫酮(21毫克,收率:56.3%)为黄色固体。LCMS(ESI):m/z=495[M+1]+.1H NMR(500MHz,MeOD)δ8.81(t,J=5.0Hz,2H),8.03(t,J=43.5Hz,1H),7.86(dd,J=13.1,9.3Hz,1H),7.32–7.23(m,1H),6.94(dd,J=15.6,13.9Hz,1H),4.57–4.37(m,1H),4.14–3.93(m,2H),3.91–3.85(m,3H),3.86–3.57(m,2H),2.96–2.72(m,2H),2.57(dd,J=9.6,4.8Hz,3H),2.31–2.08(m,2H),2.06–1.75(m,2H),1.54(ddd,J=15.7,9.2,6.9Hz,3H).Step 27e: Preparation of 2-(5-chloro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (Compound 10): 2-(5-chloro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione A mixture of '-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (0412-10) (36 mg, 0.075 mmol, 1.0 eq) and Lawesson's reagent (36 mg, 0.09 mmol, 1.2 eq) was stirred and heated in a microwave at 125°C for 45 minutes. The reaction was cooled to room temperature and quenched with water. The reaction solution was extracted with ethyl acetate, and the organic phase was dried and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography (dichloromethane/methanol/aqueous ammonia=80/4.5/0.8) to give 2-(5-chloro-2-methoxypyridin-4-yl)-1-((S)-7'-methyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propane-1-thione (21 mg, yield: 56.3%) as a yellow solid. LCMS (ESI): m/z=495[M+1] + . 1 H NMR(500MHz,MeOD)δ8.81(t,J=5.0Hz,2H),8.03(t,J=43.5Hz,1H),7.86(dd,J=13.1,9 .3Hz,1H),7.32–7.23(m,1H),6.94(dd,J=15.6,13.9Hz,1H),4.57–4.37(m,1H),4.14– 3.93(m,2H),3.91–3.85(m,3H),3.86–3.57(m,2H),2.96–2.72(m,2H),2.57(dd,J=9.6 ,4.8Hz,3H),2.31–2.08(m,2H),2.06–1.75(m,2H),1.54(ddd,J=15.7,9.2,6.9Hz,3H).

实施例28:2-(5-氯-2-甲氧基吡啶-4-基)-1-((3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(化合物104)的制备:Example 28: Preparation of 2-(5-chloro-2-methoxypyridin-4-yl)-1-((3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 104):

将(3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-100)(66毫克,0.181毫摩尔,1.0当量),2-(5-氯-2-甲氧基吡啶-4-基)丙酸(0405-10)(38毫克,0.181毫摩尔,1.0当量),N,N-二异丙基乙胺(94毫克,0.724毫摩尔,4.0当量),1-羟基苯并三唑(29毫克,0.217毫摩尔,1.2当量),苯并三氮唑-N,N,N',N'-四甲基脲六氟磷酸盐(82毫克,0.217毫摩尔,1.2当量)的二氯甲烷(2毫升)混合物在常温下搅拌2小时。反应液加水(15毫升)稀释,并用乙酸乙酯萃取(5毫升x3)。有机相用饱和食盐水洗涤(15毫升),再用无水硫酸钠干燥并减压浓缩。残留物用厚制备薄层色谱(二氯甲烷/甲醇=8/1)纯化,得到白色固体产物2-(5-氯-2-甲氧基吡啶-4-基)-1-((3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙-1-酮(48.3毫克,收率:97.3%)。LCMS(ESI):m/z=494[M+1]+.1HNMR(500MHz,MeOD)δ8.82(dd,J=6.9,5.0Hz,2H),8.18–7.70(m,2H),7.31(t,J=4.0Hz,1H),6.76(d,J=54.1Hz,1H),4.38(dd,J=14.4,7.8Hz,1H),4.16(dd,J=38.3,6.9Hz,1H),3.89(d,J=4.2Hz,3H),3.72–3.32(m,2H),2.91–2.69(m,2H),2.31(ddd,J=21.4,12.6,7.9Hz,1H),1.98–1.61(m,3H),1.38(d,J=6.6Hz,3H),1.34(dd,J=16.4,6.6Hz,3H).(3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-100) (66 mg, 0.181 mmol, 1.0 equiv), 2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid (0405-10) (38 mg, 0.181 mmol, 1.0 equiv) A mixture of 1-hydroxybenzotriazole (29 mg, 0.217 mmol, 1.2 eq.), N,N-diisopropylethylamine (94 mg, 0.724 mmol, 4.0 eq.), 1-hydroxybenzotriazole (29 mg, 0.217 mmol, 1.2 eq.), benzotriazole-N,N,N',N'-tetramethyluronium hexafluorophosphate (82 mg, 0.217 mmol, 1.2 eq.) in dichloromethane (2 ml) was stirred at room temperature for 2 hours. The reaction solution was diluted with water (15 ml) and extracted with ethyl acetate (5 ml x 3). The organic phase was washed with saturated brine (15 ml), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by thick preparative thin layer chromatography (dichloromethane/methanol=8/1) to give a white solid product 2-(5-chloro-2-methoxypyridin-4-yl)-1-((3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (48.3 mg, yield: 97.3%). LCMS (ESI): m/z=494[M+1] + .1 HNMR(500MHz,MeOD)δ8.82(dd,J=6.9,5.0Hz,2H),8.18–7.70(m,2H),7.31(t,J=4.0Hz ,1H),6.76(d,J=54.1Hz,1H),4.38(dd,J=14.4,7.8Hz,1H),4.16(dd,J=38.3,6.9Hz,1 H),3.89(d,J=4.2Hz,3H),3.72–3.32(m,2H),2.91–2.69(m,2H),2.31(ddd,J=21.4,12 .6,7.9Hz,1H),1.98–1.61(m,3H),1.38(d,J=6.6Hz,3H),1.34(dd,J=16.4,6.6Hz,3H).

实施例29:(R)-2-(5-氯-2-甲氧基吡啶-4-基)-1-((3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(化合物105)的制备(按照方案一和四线路制备) Example 29: Preparation of (R)-2-(5-chloro-2-methoxypyridin-4-yl)-1-((3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 105) (Prepared according to Schemes 1 and 4)

步骤29a:(R)-2-(5-氯-2-甲氧基吡啶-4-基)丙酸的制备:2-(5-氯-2-甲氧基吡啶-4-基)丙酸(0405-10)(12.5克,57.98毫摩尔)对映体的分离通过超临界流体色谱法进行(色谱柱:ChiralPak IG,300×50mm I.D.,10μm;流动相:A为二氧化碳,B为甲醇;梯度:15%B;流速:200毫升/分钟;背压:100巴;柱温:38℃)。第一个洗脱的对映体是(R)-2-(5-氯-2-甲氧基吡啶-4-基)丙酸(5.29克,产率:42%),第二个洗脱的对映体是(S)-2-(5-氯-2-甲氧基吡啶-4-基)丙酸(4.81g,产率:38%)。分析条件:色谱柱:ChiralPak IG,100×4.6mm I.D.,3μm;流动相:A为二氧化碳,B为甲醇(0.05%二乙胺);梯度:5-40%B;流速:2.5毫升/分钟;背压:100巴;柱温:35℃。Step 29a: Preparation of (R)-2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid: Separation of enantiomers of 2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid (0405-10) (12.5 g, 57.98 mmol) was carried out by supercritical fluid chromatography (chromatographic column: ChiralPak IG, 300×50 mm I.D., 10 μm; mobile phase: A is carbon dioxide, B is methanol; gradient: 15% B; flow rate: 200 ml/min; back pressure: 100 bar; column temperature: 38°C). The first eluting enantiomer was (R)-2-(5-chloro-2-methoxypyridin-4-yl)propionic acid (5.29 g, yield: 42%), and the second eluting enantiomer was (S)-2-(5-chloro-2-methoxypyridin-4-yl)propionic acid (4.81 g, yield: 38%). Analysis conditions: Column: ChiralPak IG, 100×4.6 mm I.D., 3 μm; Mobile phase: A is carbon dioxide, B is methanol (0.05% diethylamine); Gradient: 5-40% B; Flow rate: 2.5 ml/min; Back pressure: 100 bar; Column temperature: 35°C.

步骤29b:(R)-2-(5-氯-2-甲氧基吡啶-4-基)-1-((3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(化合物105)的制备:在0℃下,往(R)-2-(5-氯-2-甲氧基吡啶-4-基)丙酸(154毫克,0.712毫摩尔,1.05当量),(3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]二盐酸盐(0122-100)(247毫克,0.678毫摩尔,1.0当量),N-甲基咪唑(228毫克,2.78毫摩尔,4.1当量)在乙腈(2毫升)和二氯甲烷(2毫升)的混合物中加入N,N,N’,N’-四甲基氯甲脒六氟磷酸盐(247毫克,0.881毫摩尔,1.3当量)。混合物在0℃下搅拌1.5小时。混合物用水(30毫升)稀释然后用乙酸乙酯(20毫升×3)萃取。合并的有机层用饱和食盐水(20毫升)洗涤,经无水硫酸钠干燥并浓缩。残余物在硅胶上进行柱色谱分离(二氯甲烷∶甲醇30∶1),得到白色固体(R)-2-(5-氯-2-甲氧基吡啶-4-基)-1-((3S,5S)-5,7’-二甲基-6’-(嘧啶-2-基)-3’,4’-二氢-1’H-螺[吡咯烷-3,2’-[1,8]萘啶]-1-基)丙烷-1-酮(290毫克,收率:84%)。LCMS(ESI):m/z 493[M+1]+;TLC:Rf 0.5(乙酸乙酯:二氯甲烷=1:1)。1H NMR(500MHz,MeOD)δ8.82(d,J=4.9Hz,2H),8.12(s,1H),7.92(s,1H),7.31(t,J=4.9Hz,1H),6.71(s,1H),4.38(d,J=6.4Hz,1H),4.20(d,J=6.9Hz,1H),3.89(s,3H),3.69(d,J=10.9Hz,1H),3.22(d,J=11.0Hz,1H),2.81(ddd,J=24.1,14.6,7.9Hz,2H),2.61(s,3H),2.34(dd,J=13.2,7.8Hz,1H),1.90(dd,J=13.4,7.3Hz,2H),1.70(dd,J=13.3,8.6Hz,1H),1.38(d,J=6.8Hz,3H),1.33(d,J=6.2Hz,3H).Step 29b: Preparation of (R)-2-(5-chloro-2-methoxypyridin-4-yl)-1-((3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (Compound 105): At 0°C, (R)-2-(5-chloro-2-methoxypyridin-4-yl)propanoic acid (154 mg, 0.712 mmol, 1.05 equiv), (3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidin-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one were added. '-Dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine] dihydrochloride (0122-100) (247 mg, 0.678 mmol, 1.0 eq), N-methylimidazole (228 mg, 2.78 mmol, 4.1 eq) were added to a mixture of acetonitrile (2 ml) and dichloromethane (2 ml) with N,N,N',N'-tetramethylchloroformamidine hexafluorophosphate (247 mg, 0.881 mmol, 1.3 eq). The mixture was stirred at 0°C for 1.5 hours. The mixture was diluted with water (30 ml) and then extracted with ethyl acetate (20 ml×3). The combined organic layer was washed with saturated brine (20 ml), dried over anhydrous sodium sulfate and concentrated. The residue was subjected to column chromatography on silica gel (dichloromethane: methanol 30: 1) to give a white solid (R)-2-(5-chloro-2-methoxypyridin-4-yl)-1-((3S,5S)-5,7'-dimethyl-6'-(pyrimidin-2-yl)-3',4'-dihydro-1'H-spiro[pyrrolidine-3,2'-[1,8]naphthyridine]-1-yl)propan-1-one (290 mg, yield: 84% ) . LCMS (ESI): m/z 493 [M+1] + ; TLC: Rf 0.5 (ethyl acetate: dichloromethane = 1:1). NMR(500MHz,MeOD)δ8.82(d,J=4.9Hz,2H),8.12(s,1H),7.92(s,1H),7.31(t,J=4.9Hz,1H),6.71(s ,1H),4.38(d,J=6.4Hz,1H),4.20(d,J=6.9Hz,1H),3.89(s,3H),3.69(d,J=10.9Hz,1H),3.22(d,J= 11.0Hz,1H),2.81(ddd,J=24.1,14.6,7.9Hz,2H),2.61(s,3H),2.34(dd,J=13.2,7.8Hz,1H),1.90( dd,J=13.4,7.3Hz,2H),1.70(dd,J=13.3,8.6Hz,1H),1.38(d,J=6.8Hz,3H),1.33(d,J=6.2Hz,3H).

实施例30生物活性试验Example 30 Biological Activity Test

本发明所用的对照化合物为PF-07258669(Journal of Medicinal Chemistry,66(5),3195-3211,2023,WO 2021250541),其结构如下:
The reference compound used in the present invention is PF-07258669 (Journal of Medicinal Chemistry, 66 (5), 3195-3211, 2023, WO 2021250541), and its structure is as follows:

一、MC4R拮抗活性1. MC4R Antagonist Activity

利用时间分辨-荧光共振能量转移(TR-FRET)方法,检测待测化合物对人黑皮质素受体4(Melanocortin-4-receptor,MC4R)稳转在细胞株上的拮抗活性。The time-resolved fluorescence resonance energy transfer (TR-FRET) method was used to detect the antagonistic activity of the test compound on human melanocortin-4-receptor (MC4R) stably expressed in cell lines.

1实验材料与仪器
1 Experimental materials and instruments

2实验步骤2 Experimental steps

2.1待测化合物的配制与处理2.1 Preparation and handling of test compounds

2.1.1待测化合物储液的制备2.1.1 Preparation of stock solutions of test compounds

1)待测化合物溶解于DMSO,制备成10mM的储液;1) The test compound was dissolved in DMSO to prepare a 10 mM stock solution;

2)Melanotan I溶解于H2O,制备成1mM的储液。2) Melanotan I was dissolved in H 2 O to prepare a 1 mM stock solution.

2.1.2化合物储液的保存2.1.2 Storage of compound stock solutions

化合物溶解后,置于-20℃冰箱中保存。After the compound was dissolved, it was stored in a -20°C refrigerator.

2.1.3工作液的制备2.1.3 Preparation of working solution

1)待测化合物用DMSO进行5倍梯度稀释,10个或5个浓度梯度;1) The test compound was diluted 5-fold with DMSO, with 10 or 5 concentration gradients;

2)Melanotan I用Assay buffer配置成800pM待用。2) Configure Melanotan I assay buffer to 800 pM for use.

2.2待测化合物对MC4R的拮抗活性测定2.2 Determination of the antagonistic activity of the test compounds on MC4R

2.2.1细胞株、培养基及缓冲液2.2.1 Cell lines, culture media and buffers

1)细胞株:Flp-In-293-Human MC4R1) Cell line: Flp-In-293-Human MC4R

完全培养基:DMEM+10%胎牛血清+1%青霉素链霉素+200μg/mL潮霉素Complete culture medium: DMEM + 10% fetal bovine serum + 1% penicillin-streptomycin + 200 μg/mL hygromycin

2)实验缓冲液:HBSS+20mM HEPES+10%FBS+500μM IBMX 2) Experimental buffer: HBSS + 20 mM HEPES + 10% FBS + 500 μM IBMX

2.2.2细胞培养和种板2.2.2 Cell culture and plating

1)将Flp-In-293-MC4R培养于37℃,5%CO2环境下的完全培养基中;1) Flp-In-293-MC4R was cultured in complete medium at 37°C and 5% CO2 .

2)胰酶消化处理后将细胞分别重悬于实验缓冲液中,接种到384细胞培养板中,接种密度为2000每孔,接种体积为15μl每孔。2) After trypsin digestion, the cells were resuspended in the experimental buffer and inoculated into 384 cell culture plates at a density of 2000 per well and a volume of 15 μl per well.

2.2.3待测化合物对Human MC4R受体活性测定2.2.3 Determination of the activity of the test compounds on Human MC4R receptor

1)每孔加入2.5μl稀释好的8X待测化合物,37℃孵育10分钟。1) Add 2.5 μl of 8X diluted test compound to each well and incubate at 37°C for 10 minutes.

2)每孔加入2.5μl稀释好的8X Melanotan I(800pM),37℃孵育30分钟。2) Add 2.5 μl of diluted 8X Melanotan I (800 pM) to each well and incubate at 37°C for 30 minutes.

3)冻融Eu-cAMP tracer和Ulight-anti-cAMP,用lysis buffer将其稀释。3) Freeze-thaw Eu-cAMP tracer and Ulight-anti-cAMP, and dilute them with lysis buffer.

4)先加入10μl Eu-cAMP tracer至实验孔,再加入10μl Ulight-anti-cAMP至实验孔中。4) First add 10μl Eu-cAMP tracer to the experimental well, and then add 10μl Ulight-anti-cAMP to the experimental well.

5)将反应板于室温1000g离心1min,25℃静置1h后,利用Envision收集数据,激发光:340nm,发射光:665nm和615nm。5) The reaction plate was centrifuged at 1000 g for 1 min at room temperature and allowed to stand at 25°C for 1 h. Data were collected using Envision, with excitation light at 340 nm and emission light at 665 nm and 615 nm.

2.3数据分析2.3 Data Analysis

1)用公式Ratio=Signal 665nm/Signal 620nm计算每个单孔的受体和供体激发信号的比率,计算不同浓度的抑制率。利用GraphPad非线性拟合公式计算化合物IC501) The ratio of the acceptor and donor excitation signals in each single well was calculated using the formula Ratio = Signal 665nm/Signal 620nm, and the inhibition rate at different concentrations was calculated. The IC 50 of the compound was calculated using the GraphPad nonlinear fitting formula:

Y=Bottom﹢(Top﹣Bottom)/(1﹢10^((LogIC50﹣X)×HillSlope))Y=Bottom﹢(Top﹣Bottom)/(1﹢10^((LogIC 50 ﹣X)×HillSlope))

X:化合物浓度log值;Y:Inhibition%X: log value of compound concentration; Y: Inhibition%

3.实验结果3. Experimental results

计算出上述实验中各化合物的半数抑制浓度(IC50),结果如表1所示。The half maximal inhibitory concentration (IC 50 ) of each compound in the above experiment was calculated. The results are shown in Table 1.

表1.化合物对MC4R的拮抗活性

Table 1. Antagonistic activity of compounds on MC4R

二、药代动力学(PK)实验2. Pharmacokinetic (PK) Experiment

1.实验方法1. Experimental Methods

雌性C57小鼠,体重18-20克。待测化合物溶解在30%磺丁基β环糊精中,以10mg/kg灌胃给药。于给药后15分钟、30分钟和1、2、4、7及24小时进行麻醉心脏取血,然后安乐死取脑组织,每时间点约0.3ml全血及50-100mg脑组织,全血置于含K2-EDTA的离心管中,离心处理(3000g,10分钟,4℃)取血浆,血浆及脑组织-40℃保存待用;事先称重好的脑组织以1:3(g/ml)加入0.9%NaCl溶液进行匀浆处理。取50μL血浆样品或脑组织匀浆液用135μL乙腈(含内标0.1μg/mL)涡旋混合进行蛋白沉淀,离心,取上清进行LC-MS/MS分析。Female C57 mice weighing 18-20 g. The test compound was dissolved in 30% sulfobutyl β-cyclodextrin and administered orally at 10 mg/kg. Blood was collected from the heart of the mice at 15 minutes, 30 minutes, and 1, 2, 4, 7, and 24 hours after administration. The mice were then euthanized and brain tissue was collected. About 0.3 ml of whole blood and 50-100 mg of brain tissue were collected at each time point. The whole blood was placed in a centrifuge tube containing K 2 -EDTA and centrifuged (3000 g, 10 minutes, 4°C) to collect plasma. The plasma and brain tissue were stored at -40°C for later use. The brain tissue weighed in advance was homogenized with 0.9% NaCl solution at a ratio of 1:3 (g/ml). 50 μL of plasma sample or brain tissue homogenate was vortexed with 135 μL of acetonitrile (containing internal standard 0.1 μg/mL) for protein precipitation, centrifuged, and the supernatant was taken for LC-MS/MS analysis.

2.实验结果2. Experimental results

本发明提供的化合物100和化合物104在小鼠经口服给药后,吸收良好,血液和脑的暴露量较高。结果见图1-图2和表2,本发明的螺杂环化合物在血浆中的Tmax为0.25小时,Cmax为1243.33–1593.33ng/ml,AUC0-24为1448.77–1649.36hr*ng/mL,在脑中的Tmax为0.25-0.50小时,Cmax为413.20–512.00ng/ml,AUC0-24为435.13–633.25hr*ng/mL,与对照化合物PF07258669相比,本发明的化合物在脑中具有更好的药代动力学特性,在治疗MC4受体相关的疾病和/或症状如恶病质,厌食症等具有更大的治疗潜力。Cmax是指最大血药浓度,T1/2为半衰期,AUC0-24是指0-24小时时间-浓度曲线下面积,AUC0-inf是指0-Inf时间-浓度曲线下面积。The compounds 100 and 104 provided by the present invention are well absorbed after oral administration to mice, and the exposure to blood and brain is high. The results are shown in Figures 1-2 and Table 2. The spiro heterocyclic compound of the present invention has a T max of 0.25 hours in plasma, a C max of 1243.33-1593.33 ng/ml, and an AUC 0-24 of 1448.77-1649.36 hr*ng/mL, a T max of 0.25-0.50 hours in the brain, a C max of 413.20-512.00 ng/ml, and an AUC 0-24 of 435.13-633.25 hr*ng/mL. Compared with the control compound PF07258669, the compound of the present invention has better pharmacokinetic properties in the brain and has greater therapeutic potential in treating MC4 receptor-related diseases and/or symptoms such as cachexia, anorexia, etc. Cmax refers to the maximum plasma concentration, T1 /2 is the half-life, AUC0-24 refers to the area under the 0-24 hour time-concentration curve, and AUC0 -inf refers to the area under the 0-Inf time-concentration curve.

表2.小鼠灌胃给药(10mg/kg)药代动力学参数

Table 2. Pharmacokinetic parameters of oral administration in mice (10 mg/kg)

三、体外代谢稳定性试验3. In vitro metabolic stability test

1.实验方法1. Experimental Methods

将待测化合物用DMSO溶解后用乙腈稀释,离心管中加入2μL乙腈稀释待测化合物及300μL含人肝微粒体的磷酸钾缓冲液,混匀后,分别移取30μL至6个离心管中,37℃预热10分钟后,向其中加入15μL 6mM NADPH溶液(0点除外),混匀。分别在孵育30分钟和60分钟后加入135μL乙腈(含0.1μg/ml内标)溶液终止反应,0分钟样品先加入135μL乙腈(含0.1ug/ml内标)终止反应再加入15μL 6mM NADPH溶液(每个时间点均有两份样品)。混匀后,15000g离心10分钟,取上清进行LC-MS/MS分析。Dissolve the test compound in DMSO and dilute it with acetonitrile. Add 2 μL of acetonitrile to dilute the test compound and 300 μL of potassium phosphate buffer containing human liver microsomes to the centrifuge tube. After mixing, transfer 30 μL to 6 centrifuge tubes respectively. After preheating at 37°C for 10 minutes, add 15 μL of 6 mM NADPH solution (except 0 point) and mix. After incubation for 30 minutes and 60 minutes, add 135 μL of acetonitrile (containing 0.1 μg/ml internal standard) solution to terminate the reaction. For the 0-minute sample, first add 135 μL of acetonitrile (containing 0.1 ug/ml internal standard) to terminate the reaction and then add 15 μL of 6 mM NADPH solution (two samples for each time point). After mixing, centrifuge at 15000g for 10 minutes, and take the supernatant for LC-MS/MS analysis.

实验结果以半衰期(T1/2)=0.693/K(K是以受试化合物的相对剩余百分比的对数为纵坐标,孵育时间为横坐标作直线回归所获得的速率常数)来表征。The experimental results were characterized by half-life (T 1/2 )=0.693/K (K is the rate constant obtained by linear regression with the logarithm of the relative residual percentage of the test compound as the ordinate and the incubation time as the abscissa).

2.实验结果2. Experimental results

计算出上述实验中各化合物的半衰期(T1/2),结果见表3。结果表明,化合物100和化合物105在人肝微粒体中的代谢稳定性较PF07258669更好,代谢半衰期更长。提示本发明化合物在人体中药物消除半衰期会更长,从而减少给药频次。The half-life (T 1/2 ) of each compound in the above experiment was calculated, and the results are shown in Table 3. The results show that compound 100 and compound 105 have better metabolic stability in human liver microsomes than PF07258669 and longer metabolic half-life, suggesting that the drug elimination half-life of the compounds of the present invention in the human body will be longer, thereby reducing the frequency of administration.

表3化合物在人肝微粒体的代谢稳定性结果
Table 3 Metabolic stability results of compounds in human liver microsomes

以上所述实施例仅表达了本发明的几种实施方式,其描述较为具体和详细,但并不能因此而理解为对本发明专利范围的限制。应当指出的是,对于本领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干变形和改进,这些都属于本发明的保护范围。因此,本发明专利的保护范围应以所附权利要求为准。 The above-mentioned embodiments only express several implementation methods of the present invention, and the description thereof is relatively specific and detailed, but it cannot be understood as limiting the scope of the patent of the present invention. It should be pointed out that, for ordinary technicians in this field, several variations and improvements can be made without departing from the concept of the present invention, which all belong to the protection scope of the present invention. Therefore, the protection scope of the patent of the present invention shall be subject to the attached claims.

Claims (28)

一种具有式(I)所示结构的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子:
A spiroheterocyclic compound having a structure represented by formula (I) or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated product thereof or a prodrug molecule thereof:
其中:in: R1、R2、R3和R4分别独立选自:H,氘,C1-C6烷基,C1-C6氘代烷基,C2-C6烯基,C2-C6炔基,C3-C8环烷基,卤素取代的C3-C8环烷基,C3-C8环烷基甲基,卤素,卤素取代的C1-C6烷基,羟基,羟基取代的C1-C6烷基,C1-C6烷氧基取代的C1-C6烷基,氰基,-OR,-N(R)2,-SR,-C(O)OR,-C(O)N(R)2,-C(O)R,-S(O)R,-S(O)2R,-S(O)2N(R)2,-N(R)C(O)R;R 1 , R 2 , R 3 and R 4 are independently selected from the group consisting of H, deuterium, C 1 -C 6 alkyl, C 1 -C 6 deuterated alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, halogen-substituted C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkylmethyl, halogen, halogen-substituted C 1 -C 6 alkyl, hydroxy, hydroxy-substituted C 1 -C 6 alkyl, C 1 -C 6 alkoxy-substituted C 1 -C 6 alkyl, cyano, -OR, -N(R) 2 , -SR, -C(O)OR, -C(O)N(R) 2 , -C(O)R, -S(O)R, -S(O) 2 R, -S(O) 2 N(R) 2 , -N(R)C(O)R; R选自:H,C1-C6烷基,C2-C6烯基,C2-C6炔基,C3-C8环烷基,C3-C8环烷基甲基,卤素取代的C1-C6烷基,羟基取代的C1-C6烷基,烷氧基取代的C1-C6烷基,氨基取代的C1-C6烷基,烷基胺基取代的C1-C6烷基;R is selected from the group consisting of: H, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkylmethyl, halogen-substituted C 1 -C 6 alkyl, hydroxy-substituted C 1 -C 6 alkyl, alkoxy-substituted C 1 -C 6 alkyl, amino-substituted C 1 -C 6 alkyl, alkylamino-substituted C 1 -C 6 alkyl; R5和R6分别独立选自:H,氘,卤素;R 5 and R 6 are independently selected from: H, deuterium, halogen; M选自:CH,N;M is selected from: CH, N; W选自:O,S,NR7W is selected from: O, S, NR 7 ; Q选自:CR7R8,NR8,C=O,O,S,或者不存在;Q is selected from the group consisting of: CR 7 R 8 , NR 8 , C═O, O, S, or absent; R7和R8分别独立选自:H,氘,卤素,C1-C6烷基,C1-C6氘代烷基,C2-C6烯基,C2-C6炔基,C3-C8环烷基,卤素取代的C1-C6烷基,卤素取代的C3-C8环烷基,羟基,氰基,氨基,C1-C6烷基胺基;或者R8和A中的取代基相连形成取代或者未取代的5-7元碳环或杂环; R7 and R8 are independently selected from the group consisting of: H, deuterium, halogen, C1 - C6 alkyl, C1 - C6 deuterated alkyl, C2- C6 alkenyl, C2 - C6 alkynyl, C3 - C8 cycloalkyl, halogen-substituted C1 - C6 alkyl, halogen-substituted C3 - C8 cycloalkyl, hydroxyl, cyano, amino, C1 - C6 alkylamino; or R8 and the substituents in A are linked to form a substituted or unsubstituted 5-7 membered carbocyclic or heterocyclic ring; A选自:R9和R10取代或未取代的C6-C10元芳基,R9和R10取代或未取代的5-10元杂芳基,R9和R10取代或未取代的7-10元苯并杂环基;A is selected from: C 6 -C 10 membered aryl substituted or unsubstituted by R 9 and R 10 , 5-10 membered heteroaryl substituted or unsubstituted by R 9 and R 10, 7-10 membered benzoheterocyclyl substituted or unsubstituted by R 9 and R 10 ; 各R9和R10分别独立选自:H,氘,卤素,C1-C6烷基,C1-C6氘代烷基,C2-C6烯基,C2-C6炔基,氰基,C3-C8环烷基,C3-C8氘代环烷基,C3-C8环烷基C1-C6烷基,C1-C6烷基磺酰基,C1-C6烷硫基,C1-C6氘代烷硫基,C3-C8环烷基硫基,卤素取代的C1-C6烷基,C1-C6烷氧基,C1-C6氘代烷氧基,卤素取代的C1-C6烷氧基,C3-C8环烷基氧基,C3-C8氘代环烷基氧基,卤素取代的C3-C8环烷基氧基,氨基甲酰基;Each R9 and R10 is independently selected from the group consisting of H, deuterium, halogen, C1 - C6 alkyl , C1 - C6 deuterated alkyl, C2 - C6 alkenyl, C2 - C6 alkynyl, cyano , C3 - C8 cycloalkyl, C3 - C8 deuterated cycloalkyl, C3- C8 cycloalkylC1- C6 alkyl, C1 - C6 alkylsulfonyl, C1- C6 alkylthio, C1-C6 deuterated alkylthio, C3-C8 cycloalkylthio , halogen -substituted C1 - C6 alkyl, C1 - C6 alkoxy, C1 - C6 deuterated alkoxy, halogen-substituted C1 - C6 alkoxy, C3 - C8 cycloalkyloxy, C3-C8 deuterated cycloalkyloxy, halogen-substituted C3 -C6 8 -cycloalkyloxy, carbamoyl; A1选自:R11和R12取代或未取代的苯基,R11和R12取代或未取代的5-6元杂芳基; A1 is selected from: phenyl substituted or unsubstituted by R11 and R12 , 5-6 membered heteroaryl substituted or unsubstituted by R11 and R12 ; 各Rn和R12分别独立选自:H,氘,卤素,C1-C6烷基,C1-C6氘代烷基,C3-C8环烷基,卤素取代的C1-C6烷基,氰基,C1-C6烷氧基。Each of R n and R 12 is independently selected from the group consisting of: H, deuterium, halogen, C 1 -C 6 alkyl, C 1 -C 6 deuterated alkyl, C 3 -C 8 cycloalkyl, halogen-substituted C 1 -C 6 alkyl, cyano, and C 1 -C 6 alkoxy.
根据权利要求1所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,A选自:R9和R10取代或未取代的萘基,R9和R10取代或未取代的9-10元杂芳基,R9和R10取代或未取代的9-10元苯并杂环基。The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to claim 1, characterized in that A is selected from: naphthyl substituted or unsubstituted by R 9 and R 10 , 9-10 membered heteroaryl substituted or unsubstituted by R 9 and R 10 , 9-10 membered benzoheterocyclic group substituted or unsubstituted by R 9 and R 10 . 根据权利要求1所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,A选自:
The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to claim 1, characterized in that A is selected from:
其中:环中虚线代表其是一个C-C单键或者没有;Among them: the dotted line in the ring represents whether it is a C-C single bond or not; X1、X2、X3分别独立选自:CR9,N。X 1 , X 2 , and X 3 are independently selected from the group consisting of CR 9 , N.
根据权利要求3所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,A选自:
The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to claim 3, characterized in that A is selected from:
根据权利要求4所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,A选自:The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to claim 4, characterized in that A is selected from: W为O。 W is O. 根据权利要求1-5任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异 构体或者其氘代物或者其前药分子,其特征在于,各R9和R10分别独立选自:H,氘,卤素,C1-C3烷基,C1-C3氘代烷基,C2-C3烯基,C2-C3炔基,氰基,C3-C6环烷基,C3-C6氘代环烷基,C3-C6环烷基甲基,C1-C3烷基磺酰基,C1-C3烷硫基,C1-C3氘代烷硫基,C3-C6环烷基硫基,卤素取代的C1-C3烷基,C1-C3烷氧基,C1-C3氘代烷氧基,卤素取代的C1-C3烷氧基,C3-C6环烷基氧基,C3-C6氘代环烷基氧基,卤素取代的C3-C6环烷基氧基,氨基甲酰基。The spiroheterocyclic compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof The invention relates to a construct or a deuterated product thereof or a prodrug molecule thereof, characterized in that each R 9 and R 10 is independently selected from the group consisting of H, deuterium, halogen, C 1 -C 3 alkyl, C 1 -C 3 deuterated alkyl, C 2 -C 3 alkenyl, C 2 -C 3 alkynyl, cyano, C 3 -C 6 cycloalkyl, C 3 -C 6 deuterated cycloalkyl, C 3 -C 6 cycloalkylmethyl, C 1 -C 3 alkylsulfonyl, C 1 -C 3 alkylthio, C 1 -C 3 deuterated alkylthio, C 3 -C 6 cycloalkylthio, halogen-substituted C 1 -C 3 alkyl, C 1 -C 3 alkoxy, C 1 -C 3 deuterated alkoxy, halogen-substituted C 1 -C 3 alkoxy, C 3 -C 6 cycloalkyloxy, C 3 -C 6- deuterated cycloalkyloxy, halogen-substituted C 3 -C 6 cycloalkyloxy, carbamoyl. 根据权利要求6所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,各R9和Ri0分别独立选自:氢、甲氧基、氟、二氟甲基、二氟甲氧基、氰基、甲基、乙炔基、氨基甲酰基、甲磺酰基、环丙氧基、甲硫基、氯。The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to claim 6, characterized in that each R 9 and R i0 is independently selected from: hydrogen, methoxy, fluorine, difluoromethyl, difluoromethoxy, cyano, methyl, ethynyl, carbamoyl, methylsulfonyl, cyclopropyloxy, methylthio, and chlorine. 根据权利要求3所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,A选自:
The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to claim 3, characterized in that A is selected from:
根据权利要求1-5任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,各R11和R12分别独立选自:H,氘,卤素, C1-C3烷基,C1-C3氘代烷基,C3-C6环烷基,卤素取代的C1-C3烷基,氰基,C1-C3烷氧基。The spiroheterocyclic compound or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated product thereof or a prodrug molecule thereof according to any one of claims 1 to 5, characterized in that each R 11 and R 12 is independently selected from: H, deuterium, halogen, C 1 -C 3 alkyl, C 1 -C 3 deuterated alkyl, C 3 -C 6 cycloalkyl, halogen-substituted C 1 -C 3 alkyl, cyano, C 1 -C 3 alkoxy. 根据权利要求1-5任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,A1选自:
The spiroheterocyclic compound or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated product thereof or a prodrug molecule thereof according to any one of claims 1 to 5, characterized in that A 1 is selected from:
根据权利要求10所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,A1选自:
The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to claim 10, characterized in that A 1 is selected from:
根据权利要求1-5任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,W选自:S,NR7The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to any one of claims 1 to 5, characterized in that W is selected from: S, NR 7 ; 其中,W中的R7选自:H,C1-C6烷基,C1-C6氘代烷基,C3-C8环烷基,卤素取代的C1-C6烷基,卤素取代的C3-C8环烷基。Wherein, R7 in W is selected from the group consisting of: H, C1 - C6 alkyl, C1 - C6 deuterated alkyl, C3 - C8 cycloalkyl, halogen-substituted C1 - C6 alkyl, and halogen-substituted C3 - C8 cycloalkyl. 根据权利要求1-5任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,R7和R8分别独立选自:H,氘,卤素,C1-C3烷基,C1-C3氘代烷基,C2-C3烯基,C2-C3炔基,C3-C6环烷基,卤素取代的C1-C3烷基,卤素取代的C3-C6环烷基,羟基,氰基,氨基,C1-C3烷基胺基;或者R8和A中的取代基相连形成取代或者未取代的5-6元碳环或杂环。The spiroheterocyclic compound or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated product thereof or a prodrug molecule thereof according to any one of claims 1 to 5, characterized in that R7 and R8 are independently selected from: H, deuterium, halogen, C1 - C3 alkyl, C1- C3 deuterated alkyl, C2 - C3 alkenyl, C2-C3 alkynyl , C3 - C6 cycloalkyl, halogen-substituted C1 - C3 alkyl, halogen-substituted C3 -C6 cycloalkyl , hydroxyl, cyano, amino, C1 - C3 alkylamino; or R8 and the substituent in A are connected to form a substituted or unsubstituted 5-6-membered carbocyclic or heterocyclic ring. 根据权利要求13所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,R7和R8分别独立选自:H、氘、甲基、羟基,氰基,乙烯基、乙炔基、氨基、氘代甲基,或者R8和A中的取代基相连形成5-6元碳环或杂环。The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to claim 13, characterized in that R7 and R8 are independently selected from: H, deuterium, methyl, hydroxyl, cyano, vinyl, ethynyl, amino, deuterated methyl, or R8 and the substituent in A are connected to form a 5-6 membered carbocyclic or heterocyclic ring. 根据权利要求1-5任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,Q选自:-N(CH3)-, The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to any one of claims 1 to 5, characterized in that Q is selected from: -N(CH 3 )-, 根据权利要求1-5任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,R4选自:C1-C6烷基,C1-C6氘代烷基,卤素取代的C1-C6烷基。 The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to any one of claims 1 to 5, characterized in that R4 is selected from: C1 - C6 alkyl, C1 - C6 deuterated alkyl, halogen-substituted C1 - C6 alkyl. 根据权利要求1-5任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,R1、R2、R3和R4分别独立选自:H,氘,C1-C3烷基,C1-C3氘代烷基,C2-C4烯基,C2-C4炔基,C3-C6环烷基,卤素取代的C3-C6环烷基,C3-C6环烷基甲基,卤素,卤素取代的C1-C3烷基,OH,羟基取代的C1-C3烷基,烷氧基取代的C1-C3烷基,氰基。The spiroheterocyclic compound or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated product thereof or a prodrug molecule thereof according to any one of claims 1 to 5, characterized in that R 1 , R 2 , R 3 and R 4 are independently selected from the group consisting of: H, deuterium, C 1 -C 3 alkyl, C 1 -C 3 deuterated alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 6 cycloalkyl, halogen-substituted C 3 -C 6 cycloalkyl, C 3 -C 6 cycloalkylmethyl, halogen, halogen-substituted C 1 -C 3 alkyl, OH, hydroxy-substituted C 1 -C 3 alkyl, alkoxy-substituted C 1 -C 3 alkyl, and cyano. 根据权利要求17所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,R1选自:甲基、羟基、二氟甲基、三氟甲基、氰基、氯、氟、乙烯基、乙炔基、环丙基、氘代甲基。The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to claim 17, characterized in that R1 is selected from: methyl, hydroxyl, difluoromethyl, trifluoromethyl, cyano, chlorine, fluorine, vinyl, ethynyl, cyclopropyl, deuterated methyl. 根据权利要求17任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,R2选自:H、氟、甲基、氯、氘。The spiroheterocyclic compound or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a deuterated product thereof or a prodrug molecule thereof according to any one of claim 17, characterized in that R2 is selected from: H, fluorine, methyl, chlorine, deuterium. 根据权利要求17任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,R3选自:H、氟。The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to any one of claim 17, characterized in that R 3 is selected from: H, fluorine. 根据权利要求17任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,R4选自:H、甲基、甲氧基甲基、一氟甲基。The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to any one of claim 17, characterized in that R4 is selected from: H, methyl, methoxymethyl, monofluoromethyl. 根据权利要求1所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子,其特征在于,选自如下化合物:





The spiroheterocyclic compound or its pharmaceutically acceptable salt or its stereoisomer or its deuterated product or its prodrug molecule according to claim 1, characterized in that it is selected from the following compounds:





权利要求1-22任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子在制备MC4受体拮抗剂中的应用。Use of the spiroheterocyclic compound according to any one of claims 1 to 22, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, or a deuterated product thereof, or a prodrug molecule thereof in the preparation of an MC4 receptor antagonist. 权利要求1-22任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子在制备用于预防和/或治疗与MC4受体相关的疾病和/或症状的药物中的应用。Use of the spiroheterocyclic compound according to any one of claims 1 to 22, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, or a deuterated product thereof, or a prodrug molecule thereof, in the preparation of a medicament for preventing and/or treating diseases and/or symptoms associated with the MC4 receptor. 根据权利要求24所述的应用,其特征在于,与MC4受体相关的疾病和/或症状选自:恶病质,厌食症,恶心,呕吐,非自愿的体重减轻,少肌症,肌肉萎缩,肌肉无力,虚弱,骨骼疾病,疼痛或神经性疼痛,焦虑,性功能障碍,炎症性疾病。The use according to claim 24 is characterized in that the diseases and/or symptoms associated with the MC4 receptor are selected from: cachexia, anorexia, nausea, vomiting, involuntary weight loss, sarcopenia, muscle atrophy, muscle weakness, asthenia, bone diseases, pain or neuropathic pain, anxiety, sexual dysfunction, and inflammatory diseases. 根据权利要求25所述的应用,其特征在于,所述恶病质包括与慢性疾病,癌症,获得性免疫缺陷综合征,心力衰竭和/或慢性肾脏病相关的恶病质;所述厌食症包括神经性厌食症,老年性厌食症,与化疗、放疗相关的厌食症;所述骨骼疾病包括骨质流失;所述焦虑包括创伤后应激障碍、PTSD;所述炎症性疾病包括与恶病质、厌食症、少肌症相关的炎症性疾病。The use according to claim 25 is characterized in that the cachexia includes cachexia associated with chronic diseases, cancer, acquired immunodeficiency syndrome, heart failure and/or chronic kidney disease; the anorexia includes anorexia nervosa, senile anorexia, and anorexia associated with chemotherapy and radiotherapy; the bone disease includes bone loss; the anxiety includes post-traumatic stress disorder, PTSD; the inflammatory disease includes inflammatory diseases associated with cachexia, anorexia, and sarcopenia. 一种预防和/或治疗与MC4受体相关的疾病和/或症状的药物组合物,其特征在于,包括活性成分以及药学上可接受的辅料,所述活性成分包括有权利要求1-22任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子。A pharmaceutical composition for preventing and/or treating diseases and/or symptoms associated with MC4 receptors, characterized in that it comprises an active ingredient and a pharmaceutically acceptable excipient, wherein the active ingredient comprises a spiroheterocyclic compound according to any one of claims 1 to 22, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, or a deuterated product thereof, or a prodrug molecule thereof. 一种预防和/或治疗与MC4受体相关的疾病和/或症状的方法,其特征在于,所述方法包括:施用安全有效量的权利要求1-22任一项所述的螺杂环化合物或者其药学上可接受的盐或者其立体异构体或者其氘代物或者其前药分子;和/或,A method for preventing and/or treating diseases and/or symptoms associated with MC4 receptors, characterized in that the method comprises: administering a safe and effective amount of the spiroheterocyclic compound according to any one of claims 1 to 22, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, or a deuterated substance thereof, or a prodrug molecule thereof; and/or, 施用安全有效量的权利要求27所述的药物组合物。 Administer a safe and effective amount of the pharmaceutical composition of claim 27.
PCT/CN2024/087019 2023-06-14 2024-04-10 Spiro heterocyclic compound and use thereof Pending WO2024255412A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202480000825.5A CN118510780A (en) 2023-06-14 2024-04-10 Spiro heterocyclic compounds and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202310705063.6 2023-06-14
CN202310705063 2023-06-14

Publications (1)

Publication Number Publication Date
WO2024255412A1 true WO2024255412A1 (en) 2024-12-19

Family

ID=93851302

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2024/087019 Pending WO2024255412A1 (en) 2023-06-14 2024-04-10 Spiro heterocyclic compound and use thereof

Country Status (1)

Country Link
WO (1) WO2024255412A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023275715A1 (en) * 2021-06-30 2023-01-05 Pfizer Inc. Metabolites of selective androgen receptor modulators
CN116113635A (en) * 2020-06-09 2023-05-12 辉瑞公司 Spiro compounds as melanocortin 4 receptor antagonists and uses thereof
CN117586259A (en) * 2022-08-16 2024-02-23 长春金赛药业有限责任公司 Spirocyclic MC4R antagonist compound, pharmaceutical composition and application thereof in medicine
CN117736206A (en) * 2022-09-21 2024-03-22 长春金赛药业有限责任公司 Urea derivatives as MC4R antagonists, pharmaceutical compositions and pharmaceutical applications thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116113635A (en) * 2020-06-09 2023-05-12 辉瑞公司 Spiro compounds as melanocortin 4 receptor antagonists and uses thereof
WO2023275715A1 (en) * 2021-06-30 2023-01-05 Pfizer Inc. Metabolites of selective androgen receptor modulators
CN117586259A (en) * 2022-08-16 2024-02-23 长春金赛药业有限责任公司 Spirocyclic MC4R antagonist compound, pharmaceutical composition and application thereof in medicine
CN117736206A (en) * 2022-09-21 2024-03-22 长春金赛药业有限责任公司 Urea derivatives as MC4R antagonists, pharmaceutical compositions and pharmaceutical applications thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE Registry 5 July 2023 (2023-07-05), ANONYMOUS: " 1-Propanone, 2-[2-(difluoromethoxy)-6-methoxy-4-pyridinyl]-1-[(2S)-3,4- dihydro-7-methyl-6-(2-pyrimidinyl)spiro[1,8-naphthyridine-2(1H),3'- pyrrolidin]-1'-yl]-, (2R)- (CA INDEX NAME)", XP093248548, Database accession no. RN 2940996-64-3 *
DATABASE Registry 9 January 2022 (2022-01-09), ANONYMOUS: " 1-Propanone, 1-[(2S)-6-(6-cyclopropyl-3-pyridazinyl)-3,4-dihydro-7- methylspiro[1,8-naphthyridine-2(1H),3'-pyrrolidin]-1'-yl]-2-(5-fluoro-2- methoxy-4-pyridinyl)-, (2R)- ", XP093248551, Database accession no. RN 2755892-58-9 *
GARNSEY MICHELLE R, SMITH AARON C.; POLIVKOVA JANA; ARONS AUTUMN L.; BAI GUOYUN; BLAKEMORE CAROLINE; BOEHM MARKUS; BUZON LEANNE M.: "Discovery of the Potent and Selective MC4R Antagonist PF-07258669 for the Potential Treatment of Appetite Loss", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 66, no. 5, 9 March 2023 (2023-03-09), US , pages 3195 - 3211, XP093171846, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.2c02012 *

Similar Documents

Publication Publication Date Title
AU2017330733B2 (en) Pyrazolopyridine derivative having GLP-1 receptor agonist effect
JP6942853B2 (en) Diazanaphthalene compounds as JAK kinase inhibitors
CN112135824B (en) Heterocyclic compounds as immunomodulators
CN113493440B (en) Salt of nitrogen-containing heteroaromatic derivative and crystal form thereof
KR101675614B1 (en) N-containing heteroaryl derivatives as jak3 kinase inhibitors
WO2020108590A1 (en) Pyrimidine and five-membered nitrogen heterocycle derivative, preparation method therefor, and medical uses thereof
JP2019099571A (en) Pharmaceutical composition containing pyrazolopyridine derivative having GLP-1 receptor agonist activity
CN115504986A (en) Apoptosis signal-regulated kinase inhibitor and its preparation method and application
TW201833102A (en) Amine-substituted heterocyclic compound as an EHMT2 inhibitor and method of use thereof
WO2020078362A1 (en) Imidazole aromatic ring compound, preparation method and uses thereof
TW201625616A (en) Spiro urea compounds as RSV antiviral compounds
WO2019062657A1 (en) Nitrogen heterocyclic derivative, preparation method therefor, and pharmaceutical use thereof
CN118724878A (en) An AT2R antagonist
WO2022033567A1 (en) Benzimidazole derivatives, preparation method therefor and medical use thereof
CN112979654B (en) Heteroaryl fused ring compounds, preparation method and application thereof
JP6905993B2 (en) Substituted bicyclic heterocyclic compound
CN116655602A (en) PI3K alpha allosteric inhibitors
WO2022174765A1 (en) Fused ring compound as wee-1 inhibitor
WO2024255412A1 (en) Spiro heterocyclic compound and use thereof
TW202506118A (en) Heterocyclic alkynyl-substituted amide derivatives, preparation methods and applications thereof
KR20080108129A (en) Chromane derivatives
KR20240144093A (en) Carboline compounds and uses thereof
CN118510780A (en) Spiro heterocyclic compounds and uses thereof
WO2022156449A1 (en) Novel benzoazepine compound, and composition and use thereof
WO2022100738A1 (en) Crystal form of free base of inhibitor containing bicyclic ring derivative and preparation method and application of crystal form

Legal Events

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

Ref document number: 24822367

Country of ref document: EP

Kind code of ref document: A1