WO2017186147A1 - 作为新型二价iap拮抗剂的苯并咪唑联吲哚化合物 - Google Patents

作为新型二价iap拮抗剂的苯并咪唑联吲哚化合物 Download PDF

Info

Publication number
WO2017186147A1
WO2017186147A1 PCT/CN2017/082227 CN2017082227W WO2017186147A1 WO 2017186147 A1 WO2017186147 A1 WO 2017186147A1 CN 2017082227 W CN2017082227 W CN 2017082227W WO 2017186147 A1 WO2017186147 A1 WO 2017186147A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
fluoro
pharmaceutically acceptable
acceptable salt
compound according
Prior art date
Application number
PCT/CN2017/082227
Other languages
English (en)
French (fr)
Inventor
孙飞
丁照中
蔡哲
钱文远
胡国平
黎健
陈曙辉
Original Assignee
南京明德新药研发股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 南京明德新药研发股份有限公司 filed Critical 南京明德新药研发股份有限公司
Priority to RU2018141411A priority Critical patent/RU2018141411A/ru
Priority to CA3022424A priority patent/CA3022424A1/en
Priority to EP17788799.9A priority patent/EP3450430A4/en
Priority to AU2017258145A priority patent/AU2017258145A1/en
Priority to CN201780023651.4A priority patent/CN109071508B/zh
Priority to KR1020187032899A priority patent/KR20180137518A/ko
Priority to JP2018555921A priority patent/JP2019514900A/ja
Priority to US16/096,848 priority patent/US10508103B2/en
Publication of WO2017186147A1 publication Critical patent/WO2017186147A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to a novel class of divalent benzimidazole hydrazine compounds as IAP antagonists, and specifically discloses a compound of the formula (I) or a pharmaceutically acceptable salt thereof.
  • Apoptosis refers to the orderly death of cells that are controlled by genes to maintain homeostasis. It plays an important role in the evolution of organisms, the stability of the internal environment, and the development of multiple systems. Apoptosis signaling is divided into intrinsic (mediated by death receptor-ligand interaction) and extrinsic (mediated by cellular stress and mitochondrial permeability). Both pathways eventually converge on caspase. Once the apoptotic signal is activated, caspase cleaves a large number of substrates associated with cell death, causing cell death.
  • IAPs apoptosis proteins
  • NAIP neuronal apoptosis inhibitor protein
  • IAP1 and c-IAP2 X-linked inhibitor of apoptosis
  • XIAP X-linked inhibitor of apoptosis
  • Survivin Survivin
  • melanoma-IAP ML-IAP/Livin
  • testicular specific inhibitor of apoptosis hILP
  • the baculovirus IAP repeat of ubiquitin ligase, etc. has found 8 human IAPs family protein members to date.
  • cIAP1, cIAP2, and XIAP were well studied. They all have three structural functional regions, called BIR1, BIR2 and BIR3, which play a role in blocking apoptosis mainly by inhibiting the activities of enzymes such as Caspase 3, 7, and 9.
  • Smac termed second mitochondria-derived activator of caspases
  • Smac is a protein that exists in mitochondria and regulates apoptosis. Its pro-apoptotic effect is through reversal of inhibitors of apoptosis (IAPs), especially X-linked inhibitor of apoptosis (XIAP).
  • IAPs inhibitors of apoptosis
  • XIAP X-linked inhibitor of apoptosis
  • the Smac protein directly binds to various IAP proteins through the N-terminal tetrapeptide, blocks the inhibition of apoptosis of the IAP protein in the cell, and can effectively promote apoptosis.
  • IAP inhibitors also known as Smac mimetics
  • Birinapant, LCL-161, AT-406, etc. have entered the clinical phase I or phase II study.
  • new IAP antagonists with better activity, selectivity and safety are still in great demand.
  • the present invention provides a compound of the formula (I) or a pharmaceutically acceptable salt thereof,
  • R 1 and R 2 are each independently selected from
  • R 3 , R 4 and R 5 are each independently selected from the group consisting of 1, 2 or 3 R substituted: C 1-6 alkyl, C 1-6 heteroalkyl, C 3-12 cycloalkyl, 3 ⁇ 12-membered heterocycloalkyl, 5- to 12-membered aryl or heteroaryl, 5- to 12-membered aralkyl or heteroaralkyl;
  • R 6a and R 6b are each independently selected from H, F, Cl, Br, I, OH, CN, NH 2 , COOH, or selected from: C 1-6 alkane optionally substituted by 1, 2 or 3 R , C 1-6 heteroalkyl, C 3-6 cycloalkyl, 3-6 -membered heterocycloalkyl, 5- to 6-membered aryl or heteroaryl, 5- to 6-membered aralkyl or heteroaralkyl ;
  • R 6a and R 6b are joined together to form a 3 to 6 membered ring optionally substituted by 1, 2 or 3 R;
  • R 7a and R 7b are each independently selected from H, F, Cl, Br, I, OH, CN, NH 2 , COOH, or from a C 1-6 alkane optionally substituted by 1, 2 or 3 R; , C 1-6 heteroalkyl, C 3-6 cycloalkyl, 3-6 -membered heterocycloalkyl, 5- to 6-membered aryl or heteroaryl, 5- to 6-membered aralkyl or heteroaralkyl ;
  • R 7a and R 7b are joined together to form a 3 to 6 membered ring optionally substituted by 1, 2 or 3 R;
  • Ring A and Ring B are each independently selected from: a 5- to 6-membered aryl or heteroaryl group, a 5- to 6-membered aralkyl group or a heteroarylalkyl group;
  • R 8 and R 9 are each independently selected from halogen, hydroxy or selected from C 1 1-6 alkyl, C 1-6 heteroalkyl, C 3-6 cycloalkane optionally substituted by 1, 2 or 3 R. a 3- to 6-membered heterocycloalkyl group;
  • n are each independently selected from: 0, 1, 2 or 3;
  • R is selected from F, Cl, Br, I, CN, OH, NH 2 , COOH, or selected from C 1 1-6 alkyl, C 1-6 heteroalkyl optionally substituted by 1, 2 or 3 R' a group, a C 3-6 cycloalkyl group, a 3 to 6 membered heterocycloalkyl group, a phenyl group and a 5- to 6-membered heteroaryl group;
  • R' is selected from the group consisting of F, Cl, Br, I, OH, CN, NH 2 , COOH, Me, Et, CF 3 , CHF 2 , CH 2 F, NHCH 3 , N(CH 3 ) 2 ;
  • the number of heteroatoms or heteroatoms is independently selected from 1, 2 or 3.
  • the above R is selected from the group consisting of F, Cl, Br, I, CN, OH, NH 2 , COOH, or selected from the group consisting of 1, 2 or 3 R' substitutions: C 1-3 Alkyl, C 1-3 alkoxy, C 1-3 alkylthio, C 1-3 alkylamino and N,N-di(C 1-2 alkyl)amino.
  • R is selected from the group consisting of F, Cl, Br, I, CN, OH, NH 2 , COOH, Me, Et, CF 3 , CHF 2 , CH 2 F, NHCH 3 , N (CH 3 ) ) 2 ,
  • R 3 and R 4 above are each independently selected from the group consisting of 1, 2 or 3 R: C 1-4 alkyl, C 1-4 heteroalkyl, C 3 - 6 -cycloalkyl, 3- to 6-membered heterocycloalkyl, 5- to 6-membered aryl or heteroaryl, 5- to 6-membered aralkyl or heteroaralkyl.
  • R 3 and R 4 are each independently selected from: C 1-3 alkyl, C 1-3 heteroalkyl, phenyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazine Base, furanyl, imidazolyl, oxazolyl, isoxazolyl, thienyl and pyrazolyl.
  • R 3 and R 4 above are each independently selected from Me.
  • R 5 above is selected from C 1 1-4 alkyl, C 1-4 heteroalkyl, C 3-6 cycloalkyl, 3 optionally substituted by 1, 2 or 3 R ⁇ 6-membered heterocycloalkyl, 5- to 6-membered aryl or heteroaryl, 5- to 6-membered aralkyl or heteroaralkyl.
  • the above R 5 is selected from the group consisting of: C 1-4 alkyl, C 3-6 cycloalkyl, 3- to 6-membered heterocycloalkyl, phenyl, pyridyl, pyrimidinyl, pyrazinyl , pyridazinyl, furyl, imidazolyl, oxazolyl, isoxazolyl, thienyl and pyrazolyl.
  • R 5 is selected from
  • the structural unit From:
  • the structural unit From:
  • R 6a and R 6b are bonded together to form a 3- to 6-membered cycloalkyl group optionally substituted by 1, 2 or 3 R.
  • R 7a and R 7b are bonded together to form a 3- to 6-membered cycloalkyl group optionally substituted by 1, 2 or 3 R.
  • R 7a and R 7b are linked together, the structural unit Selected from
  • the structural unit From:
  • the structural unit From:
  • the structural unit From:
  • the structural unit From:
  • the structural unit From:
  • the present invention also provides the compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of
  • R 3 , R 4 , R 5 , R 6a , R 7a , R 8 and R 9 are as defined above.
  • the invention also provides a compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of
  • the above compound, or a pharmaceutically acceptable salt thereof is selected from the group consisting of
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound described above, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the present invention also provides the use of the above compound or a pharmaceutically acceptable salt thereof or the above pharmaceutical composition for the preparation of a medicament for treating a disease caused by an IAP disorder.
  • the above-described disease caused by the IAP disorder is selected from a tumor or a hepatitis B virus infection.
  • the above R is selected from the group consisting of F, Cl, Br, I, CN, OH, NH 2 , COOH, or selected from the group consisting of 1, 2 or 3 R' substitutions: C 1-3 Alkyl, C 1-3 alkoxy, C 1-3 alkylthio, C 1-3 alkylamino and N,N-di(C 1-2 alkyl)amino, the other variables are as defined above.
  • R is selected from the group consisting of F, Cl, Br, I, CN, OH, NH 2 , COOH, Me, Et, CF 3 , CHF 2 , CH 2 F, NHCH 3 , N (CH 3 ) ) 2 , Other variables are as defined above.
  • R 3 and R 4 above are each independently selected from the group consisting of 1, 2 or 3 R: C 1-4 alkyl, C 1-4 heteroalkyl, C 3 - 6 -cycloalkyl, 3- to 6-membered heterocycloalkyl, 5- to 6-membered aryl or heteroaryl, 5- to 6-membered aralkyl or heteroaralkyl, the other variables are as defined above.
  • R 3 and R 4 are each independently selected from: C 1-3 alkyl, C 1-3 heteroalkyl, phenyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazine Base, furanyl, imidazolyl, oxazolyl, isoxazolyl, thienyl and pyrazolyl, other variables are as defined above.
  • R 3 and R 4 above are each independently selected from Me, and other variables are as defined above.
  • R 5 above is selected from C 1 1-4 alkyl, C 1-4 heteroalkyl, C 3-6 cycloalkyl, 3 optionally substituted by 1, 2 or 3 R ⁇ 6-membered heterocycloalkyl, 5- to 6-membered aryl or heteroaryl, 5- to 6-membered aralkyl or heteroaralkyl, and other variables are as defined above.
  • the above R 5 is selected from the group consisting of: C 1-4 alkyl, C 3-6 cycloalkyl, 3- to 6-membered heterocycloalkyl, phenyl, pyridyl, pyrimidinyl, pyrazinyl , pyridazinyl, furyl, imidazolyl, oxazolyl, isoxazolyl, thienyl and pyrazolyl, other variables are as defined above.
  • R 5 is selected from Other variables are as defined above.
  • the structural unit Selected from Other variables are as defined above.
  • the structural unit Selected from Other variables are as defined above.
  • R 6a and R 6b are bonded together to form a 3- to 6-membered cycloalkyl group optionally substituted by 1, 2 or 3 R, the other variables being as defined above.
  • R 6a and R 6b are linked together, the structural unit Selected from Other variables are as defined above.
  • R 6a and R 6b are linked together, the structural unit Selected from Other variables are as defined above.
  • R 7a and R 7b are bonded together to form a 3- to 6-membered cycloalkyl group optionally substituted by 1, 2 or 3 R, the other variables being as defined above.
  • R 7a and R 7b are linked together, the structural unit Selected from Other variables are as defined above.
  • the structural unit Selected from Other variables are as defined above.
  • the structural unit Selected from Other variables are as defined above.
  • the structural unit Selected from Other variables are as defined above.
  • the compound of the present invention has a mother core structure designed to be a benzimidazole, which improves the solubility of the entire molecule.
  • the benzimidazole-linked nucleus is also innovative in that it has fewer hydrogen bond donors on its mother nucleus and has lower XIAP binding, resulting in higher cIAP/XIAP selectivity and higher selectivity.
  • the compounds will have lower toxicity and better tolerance in animals and humans.
  • C 1-12 is selected from C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , C 10 , C 11 and C 12 ;
  • C 3-12 is selected from C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , C 10 , C 11 and C 12 .
  • C 1-12 alkyl or heteroalkyl, C 3-12 cycloalkyl or heterocycloalkyl, C 1-12 alkyl or heteroalkyl substituted by C 3-12 cycloalkyl or heterocycloalkyl includes, but is not limited to:
  • pharmaceutically acceptable as used herein is intended to mean that those compounds, materials, compositions and/or dosage forms are within the scope of sound medical judgment and are suitable for use in contact with human and animal tissues. Without excessive toxicity, irritation, allergic reactions or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt refers to a salt of a compound of the invention prepared from a compound having a particular substituent found in the present invention and a relatively non-toxic acid or base.
  • a base addition salt can be obtained by contacting a neutral amount of such a compound with a sufficient amount of a base in a neat solution or a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia or magnesium salts or similar salts.
  • an acid addition salt can be obtained by contacting a neutral form of such a compound with a sufficient amount of an acid in a neat solution or a suitable inert solvent.
  • pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, hydrogencarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, Hydrogen sulfate, hydroiodic acid, phosphorous acid, etc.; and an organic acid salt, such as acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, Similar acids such as fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, and me
  • the salt is contacted with a base or acid in a conventional manner, and the parent compound is separated, thereby regenerating the neutral form of the compound.
  • the parent form of the compound differs from the form of its various salts by certain physical properties, such as differences in solubility in polar solvents.
  • a "pharmaceutically acceptable salt” is a derivative of a compound of the invention wherein the parent compound is modified by salt formation with an acid or with a base.
  • pharmaceutically acceptable salts include, but are not limited to, inorganic or organic acid salts of bases such as amines, alkali metal or organic salts of acid groups such as carboxylic acids, and the like.
  • Pharmaceutically acceptable salts include the conventional non-toxic salts or quaternary ammonium salts of the parent compound, for example salts formed from non-toxic inorganic or organic acids.
  • non-toxic salts include, but are not limited to, those derived from inorganic acids and organic acids selected from the group consisting of 2-acetoxybenzoic acid, 2-hydroxyethanesulfonic acid, acetic acid, ascorbic acid, Benzenesulfonic acid, benzoic acid, hydrogencarbonate, carbonic acid, citric acid, edetic acid, ethane disulfonic acid, ethanesulfonic acid, fumaric acid, glucoheptose, gluconic acid, glutamic acid, glycolic acid, Hydrobromic acid, hydrochloric acid, hydroiodide, hydroxyl, hydroxynaphthalene, isethionethane, lactic acid, lactose, dodecylsulfonic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, nitric acid, oxalic acid, Pamoic acid, pantothenic acid, phenylacetic acid, phen
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound containing an acid group or a base by conventional chemical methods.
  • such salts are prepared by reacting these compounds in water or an organic solvent or a mixture of the two via a free acid or base form with a stoichiometric amount of a suitable base or acid.
  • a nonaqueous medium such as ether, ethyl acetate, ethanol, isopropanol or acetonitrile is preferred.
  • the compounds provided herein also exist in the form of prodrugs.
  • Prodrugs of the compounds described herein are readily chemically altered under physiological conditions to convert to the compounds of the invention.
  • prodrugs can be converted to the compounds of the invention by chemical or biochemical methods in an in vivo setting.
  • Certain compounds of the invention may exist in unsolvated or solvated forms, including hydrated forms.
  • the solvated forms are equivalent to the unsolvated forms and are included within the scope of the invention.
  • Certain compounds of the invention may have asymmetric carbon atoms (optical centers) or double bonds. Racemate, diastereomer, geometry Isomers and individual isomers are included within the scope of the invention.
  • the compounds of the invention may exist in specific geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including the cis and trans isomers, the (-)- and (+)-p-enantiomers, the (R)- and (S)-enantiomers, and the diastereomeric a conformation, a (D)-isomer, a (L)-isomer, and a racemic mixture thereof, and other mixtures, such as enantiomerically or diastereomeric enriched mixtures, all of which belong to It is within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in the substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the invention.
  • optically active (R)- and (S)-isomers as well as the D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If an enantiomer of a compound of the invention is desired, it can be prepared by asymmetric synthesis or by derivatization with a chiral auxiliary wherein the resulting mixture of diastereomers is separated and the auxiliary group cleaved to provide pure The desired enantiomer.
  • a diastereomeric salt is formed with a suitable optically active acid or base, followed by conventional methods well known in the art.
  • the diastereomers are resolved and the pure enantiomer is recovered.
  • the separation of enantiomers and diastereomers is generally accomplished by the use of chromatography using a chiral stationary phase, optionally in combination with chemical derivatization (eg, formation of an amino group from an amine). Formate).
  • the compounds of the present invention may contain unnatural proportions of atomic isotopes on one or more of the atoms that make up the compound.
  • radiolabeled compounds can be used, such as tritium (3 H), iodine -125 (125 I) or C-14 (14 C). Alterations of all isotopic compositions of the compounds of the invention, whether radioactive or not, are included within the scope of the invention.
  • pharmaceutically acceptable carrier refers to any formulation or carrier medium that is capable of delivering an effective amount of an active substance of the present invention, does not interfere with the biological activity of the active substance, and has no toxic side effects to the host or patient, including water, oil, Vegetables and minerals, cream bases, lotion bases, ointment bases, etc. These bases include suspending agents, tackifiers, transdermal enhancers and the like. Their formulations are well known to those skilled in the cosmetic or topical pharmaceutical arts. For additional information on vectors, reference is made to Remington: The Science and Practice of Pharmacy, 21st Ed., Lippincott, Williams & Wilkins (2005), the contents of which are hereby incorporated by reference.
  • excipient generally refers to the carrier, diluent and/or vehicle required to formulate an effective pharmaceutical composition.
  • an "effective amount” or “therapeutically effective amount” with respect to a pharmaceutical or pharmacologically active agent refers to a sufficient amount of a drug or agent that is non-toxic but that achieves the desired effect.
  • an "effective amount” of an active substance in a composition refers to the amount required to achieve the desired effect when used in combination with another active substance in the composition. The determination of the effective amount will vary from person to person, depending on the age and general condition of the recipient, and also on the particular active substance, and a suitable effective amount in a case can be determined by one skilled in the art based on routine experimentation.
  • active ingredient refers to a chemical entity that is effective in treating a target disorder, disease or condition.
  • substituted means that any one or more hydrogen atoms on a particular atom are replaced by a substituent, including variants of heavy hydrogen and hydrogen, as long as the valence of the particular atom is normal and the substituted compound is stable.
  • optionally substituted means that it may or may not be substituted. Unless otherwise specified, the kind and number of substituents may be arbitrary on the basis of chemically achievable.
  • any variable eg, R
  • its definition in each case is independent.
  • the group may optionally be substituted with at most two R, and each case has an independent option.
  • combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
  • linking group When the number of one linking group is 0, such as -(CRR) 0 -, it indicates that the linking group is a single bond.
  • one of the variables When one of the variables is selected from a single bond, it means that the two groups to which it is attached are directly linked. For example, when L represents a single bond in A-L-Z, the structure is actually A-Z.
  • substituents When a bond of a substituent can be cross-linked to two atoms on a ring, the substituent can be bonded to any atom on the ring.
  • substituents do not indicate which atom is attached to a compound included in the chemical structural formula including but not specifically mentioned, such a substituent may be bonded through any atomic phase thereof.
  • Combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds. For example, a structural unit It is indicated that it can be substituted at any position on the cyclohexyl or cyclohexadiene.
  • halo or halogen
  • haloalkyl is intended to include both monohaloalkyl and polyhaloalkyl.
  • halo(C 1 -C 4 )alkyl is intended to include, but is not limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like. Wait.
  • haloalkyl groups include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl.
  • Alkoxy represents the above alkyl group having a specified number of carbon atoms attached through an oxygen bridge.
  • the C 1-6 alkoxy group includes a C 1 , C 2 , C 3 , C 4 , C 5 and C 6 alkoxy groups.
  • alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentyloxy and S- Pentyloxy.
  • Cycloalkyl includes saturated cyclic groups such as cyclopropyl, cyclobutyl or cyclopentyl.
  • the 3-7 cycloalkyl group includes C 3 , C 4 , C 5 , C 6 and C 7 cycloalkyl groups.
  • Alkenyl includes hydrocarbon chains in a straight or branched configuration wherein one or more carbon-carbon double bonds, such as vinyl and propylene groups, are present at any stable site on the chain.
  • halo or halogen refers to fluoro, chloro, bromo and iodo.
  • hetero denotes a hetero atom or a hetero atomic group (ie, a radical containing a hetero atom), including atoms other than carbon (C) and hydrogen (H), and radicals containing such heteroatoms, including, for example, oxygen (O).
  • ring means substituted or unsubstituted cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, cycloalkynyl, heterocycloalkynyl, aryl or heteroaryl. So-called rings include single rings, interlocking rings, spiral rings, parallel rings or bridge rings. The number of atoms on the ring is usually defined as the number of elements of the ring. For example, "5 to 7-membered ring” means 5 to 7 atoms arranged in a circle. Unless otherwise specified, the ring optionally contains from 1 to 3 heteroatoms.
  • 5- to 7-membered ring includes, for example, phenyl, pyridine, and piperidinyl; on the other hand, the term “5- to 7-membered heterocycloalkyl ring” includes pyridyl and piperidinyl, but does not include phenyl.
  • ring also includes a ring system containing at least one ring, wherein each "ring” is independently Combined with the above definition.
  • heterocycle or “heterocyclyl” means a stable monocyclic, bicyclic or tricyclic ring containing a hetero atom or a heteroatom group which may be saturated, partially unsaturated or unsaturated ( Aromatic) which comprise a carbon atom and 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S, wherein any of the above heterocycles may be fused to a phenyl ring to form a bicyclic ring.
  • the nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O)p, p is 1 or 2).
  • the nitrogen atom can be substituted or unsubstituted (i.e., N or NR, wherein R is H or other substituents as already defined herein).
  • the heterocyclic ring can be attached to the side groups of any hetero atom or carbon atom to form a stable structure. If the resulting compound is stable, the heterocycles described herein can undergo substitutions at the carbon or nitrogen sites.
  • the nitrogen atom in the heterocycle is optionally quaternized.
  • a preferred embodiment is that when the total number of S and O atoms in the heterocycle exceeds 1, these heteroatoms are not adjacent to each other. Another preferred embodiment is that the total number of S and O atoms in the heterocycle does not exceed one.
  • aromatic heterocyclic group or "heteroaryl” as used herein means a stable 5, 6, or 7 membered monocyclic or bicyclic or aromatic ring of a 7, 8, 9 or 10 membered bicyclic heterocyclic group, It contains carbon atoms and 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S.
  • the nitrogen atom can be substituted or unsubstituted (i.e., N or NR, wherein R is H or other substituents as already defined herein).
  • the nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O)p, p is 1 or 2).
  • bridged rings are also included in the definition of heterocycles.
  • a bridged ring is formed when one or more atoms (ie, C, O, N, or S) join two non-adjacent carbon or nitrogen atoms.
  • Preferred bridged rings include, but are not limited to, one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms, and one carbon-nitrogen group. It is worth noting that a bridge always converts a single ring into a three ring. In the bridged ring, a substituent on the ring can also be present on the bridge.
  • heterocyclic compounds include, but are not limited to, acridinyl, octanoyl, benzimidazolyl, benzofuranyl, benzofuranylfuranyl, benzindenylphenyl, benzoxazolyl, benzimidin Oxazolinyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolyl, oxazolyl, 4aH-carbazolyl, Porphyrin, chroman, chromene, porphyrin-decahydroquinolinyl, 2H, 6H-1,5,2-dithiazinyl, dihydrofuro[2,3-b] Tetrahydrofuranyl, furyl, furfuryl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-carbazolyl, nonenyl,
  • hydrocarbyl or its subordinate concept (such as alkyl, alkenyl, alkynyl, phenyl, etc.) by itself or as part of another substituent means straight-chain, branched or cyclic
  • the hydrocarbon radical or a combination thereof may be fully saturated, mono- or polyunsaturated, may be monosubstituted, disubstituted or polysubstituted, and may be monovalent (such as methyl), divalent (such as methylene) or polyvalent (methine), may include a divalent or polyvalent radical having the specified number of carbon atoms (e.g., C 1 -C 10 represents 1 to 10 carbons).
  • Hydrocarbyl includes, but is not limited to, aliphatic hydrocarbyl groups including chain and cyclic, including but not limited to alkyl, alkenyl, alkynyl groups including, but not limited to, 6-12 members.
  • An aromatic hydrocarbon group such as benzene, naphthalene or the like.
  • hydrocarbyl means a straight or branched chain radical or a combination thereof, which may be fully saturated, unitary or polyunsaturated, and may include divalent and multivalent radicals.
  • saturated hydrocarbon radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, isobutyl, cyclohexyl, (cyclohexyl).
  • a homolog or isomer of a methyl group, a cyclopropylmethyl group, and an atomic group such as n-pentyl, n-hexyl, n-heptyl, n-octyl.
  • the unsaturated alkyl group has one or more double or triple bonds, and examples thereof include, but are not limited to, a vinyl group, a 2-propenyl group, a butenyl group, a crotyl group, a 2-isopentenyl group, and a 2-(butadienyl group). ), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and higher homologs and Structure.
  • heterohydrocarbyl or its subordinate concept (such as heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, etc.), by itself or in combination with another term, means a stable straight chain, branched chain. Or a cyclic hydrocarbon radical or a combination thereof having a number of carbon atoms and at least one heteroatom.
  • heteroalkyl by itself or in conjunction with another term refers to a stable straight chain, branched hydrocarbon radical or combination thereof, having a number of carbon atoms and at least one heteroatom.
  • the heteroatoms are selected from the group consisting of B, O, N, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen heteroatoms are optionally quaternized.
  • the heteroatom or heteroatom group can be located at any internal position of the heterohydrocarbyl group (including where the hydrocarbyl group is attached to the rest of the molecule).
  • Up to two heteroatoms may be consecutive, for example, -CH 2 -NH-OCH 3.
  • alkoxy alkylamino and “alkylthio” (or thioalkoxy) are customary expressions and refer to those alkane which are attached to the remainder of the molecule through an oxygen atom, an amino group or a sulfur atom, respectively.
  • Base group alkoxy
  • cycloalkyl refers to any heterocyclic alkynyl group, etc., by itself or in combination with other terms, denotes a cyclized “hydrocarbyl group” or “heterohydrocarbyl group”, respectively.
  • a hetero atom may occupy a position at which the hetero ring is attached to the rest of the molecule.
  • cycloalkyl groups include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like.
  • heterocyclic groups include 1-(1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothiophen-2-yl, tetrahydrothiophen-3-yl, 1-piperazinyl and 2-piperazinyl.
  • aryl denotes a polyunsaturated, aromatic hydrocarbon substituent which may be monosubstituted, disubstituted or polysubstituted, may be monovalent, divalent or polyvalent, it may be monocyclic or Polycyclic (such as 1 to 3 rings; at least one of which is aromatic), which are fused together or covalently linked.
  • heteroaryl refers to an aryl (or ring) containing one to four heteroatoms. In an illustrative example, the heteroatoms are selected from the group consisting of B, N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom is optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule through a heteroatom.
  • aryl or heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyridyl Azyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxan Azyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thiophene , 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-benzothiazolyl, 5-
  • aryl when used in conjunction with other terms (eg, aryloxy, arylthio, aralkyl) includes aryl as defined above Base and heteroaryl ring.
  • aralkyl is intended to include those radicals to which an aryl group is attached to an alkyl group (eg, benzyl, phenethyl, pyridylmethyl, and the like), including wherein the carbon atom (eg, methylene) has been, for example, oxygen.
  • alkyl groups substituted by an atom such as phenoxymethyl, 2-pyridyloxymethyl 3-(1-naphthyloxy)propyl and the like.
  • leaving group refers to a functional group or atom which may be substituted by another functional group or atom by a substitution reaction (for example, an affinity substitution reaction).
  • substituent groups include triflate; chlorine, bromine, iodine; sulfonate groups such as mesylate, tosylate, p-bromobenzenesulfonate, p-toluenesulfonic acid Esters and the like; acyloxy groups such as acetoxy, trifluoroacetoxy and the like.
  • protecting group includes, but is not limited to, "amino protecting group", “hydroxy protecting group” or “thiol protecting group”.
  • amino protecting group refers to a protecting group suitable for preventing side reactions at the amino nitrogen position.
  • Representative amino protecting groups include, but are not limited to, formyl; acyl, such as alkanoyl (e.g., acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl, e.g., tert-butoxycarbonyl (Boc) Arylmethoxycarbonyl, such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethoxycarbonyl (Fmoc); arylmethyl, such as benzyl (Bn), trityl (Tr), 1, 1-di -(4'-methoxyphenyl)methyl; silyl groups such as trimethylsilyl (TMS) and tert-
  • hydroxy protecting group refers to a protecting group suitable for use in preventing hydroxy side reactions.
  • Representative hydroxy protecting groups include, but are not limited to, alkyl groups such as methyl, ethyl and t-butyl groups; acyl groups such as alkanoyl groups (e.g., acetyl); arylmethyl groups such as benzyl (Bn), Oxybenzyl (PMB), 9-fluorenylmethyl (Fm) and diphenylmethyl (diphenylmethyl, DPM); silyl groups such as trimethylsilyl (TMS) and tert-butyl Dimethylsilyl (TBS) and the like.
  • alkyl groups such as methyl, ethyl and t-butyl groups
  • acyl groups such as alkanoyl groups (e.g., acetyl)
  • arylmethyl groups such as benzyl (Bn), Oxybenzyl (PMB), 9-fluoreny
  • the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments set forth below, combinations thereof with other chemical synthetic methods, and those well known to those skilled in the art. Equivalent alternatives, preferred embodiments include, but are not limited to, embodiments of the invention.
  • the present invention uses the following abbreviations:
  • Pd/C Pd/C catalyst HATU 2-(7-azobenzotriazole)-N,N,N',N'-tetramethyluron hexafluorophosphate TEMPO Tetramethylpiperidine oxynitride DIAD Diisopropyl azodicarboxylate NMM N-methylmorpholine DCM Dichloromethane THF Tetrahydrofuran Boc Tert-butoxycarbonyl Oxone Potassium persulfate composite salt Cbz Benzyloxycarbonyl DMF N,N-dimethylformamide LiBH4 Lithium borohydride TFA Trifluoroacetate EDCI 1-ethyl-(3-dimethylaminopropyl)carbonyldiimide hydrochloride Aq water EDC N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride m-CPBA 3-chloroperoxybenzoic acid Eq Equivalent, equivalent
  • Solvents used in the present invention are commercially available and require no further purification.
  • the reaction is generally carried out under an inert nitrogen atmosphere in an anhydrous solvent.
  • Proton nuclear magnetic resonance data was recorded on a Bruker Avance III 400 (400 MHz) spectrometer with chemical shifts expressed in ppm (ppm) at the high field of tetramethylsilane. Mass spectra were measured on an Agilent 1200 Series Plus 6110 (&1956A).
  • LC/MS or Shimadzu MS contains a DAD: SPD-M20A (LC) and Shimadzu Micromass 2020 detector.
  • the mass spectrometer is equipped with an electrospray ionization source (ESI) operating in either positive or negative mode.
  • ESI electrospray ionization source
  • High performance liquid chromatography was performed using a Shimadzu LC20AB system equipped with a Shimadzu SIL-20A autosampler and a Shimadzu DAD: SPD-M20A detector using a Xtimate C18 (3 m packing, size 2.1 x 300 mm) column.
  • 0-60AB_6 min method Apply a linear gradient, start elution with 100% A (A is 0.0675% TFA in water), and end the elution with 60% B (B is 0.0625% TFA in MeCN solution). The whole process is 4.2 minutes, then eluted with 60% B for 1 minute. The column was equilibrated for 0.8 minutes to reach 100:0 with a total run time of 6 minutes.
  • 10-80AB_6 min method Apply a linear gradient, start elution with 90% A (A is 0.0675% TFA in water), and end the elution with 80% B (B in 0.0625% TFA in acetonitrile). 4.2 minutes, then eluted with 80% B for 1 minute.
  • the column was equilibrated for 0.8 minutes to 90:10 with a total run time of 6 minutes.
  • the column temperature was 50 ° C and the flow rate was 0.8 mL/min.
  • the diode array detector has a scanning wavelength of 200-400 nm.
  • TLC Thin layer chromatography
  • a common solvent for flash column chromatography or thin layer chromatography is a mixture of dichloromethane/methanol, ethyl acetate/methanol and hexane/ethyl acetate.
  • Step A N-Boc-L-prolinol (50 g, 248.43 mmol), phthalimide (43.86 g, 298.12 mmol) and triphenylphosphine (65.16 g, 248.43 mmol) Dissolved in tetrahydrofuran (1 L), then DIAD (48.31 mL, 248.43 mmol) was added dropwise at 0 ° C under nitrogen. Then it was stirred at room temperature for 16 hours, quenched with water (200 ml), stirred for 10 min, and extracted with ethyl acetate (250 ml).
  • Step B tert-Butyl (2S)-2-[(1,3-dioxoisoindol-2-yl)methyl]pyrrolidin-1-carboxylate (51 g, 154.37 mmol) Dissolved in ethanol (350 ml), then hydrazine hydrate (22.07 ml, 385.93 mmol, 85% purity) was added dropwise at 80 °C. It was then stirred at this temperature for 16 hours.
  • Step C Mixing tert-butyl(2S)-2-(aminomethyl)pyrrolidine-1-carboxylate (15 g, 74.9 mmol), potassium carbonate (20.7 g, 149.8 mmol) in acetonitrile (300) In ML), 1,4-difluoro-2-nitrobenzene (13.11 ml, 82.39 mmol) was added at room temperature. The mixture was warmed to 80.degree. C., and stirred for 1 hr.
  • EtOAc EtOAc
  • Step D Dissolving tert-butyl(2S)-2-[(4-fluoro-2-nitro-anilino)methyl]pyrrolidine-1-carboxylate (11 g, crude) in methanol (100 mL) And ethyl acetate (100 ml), the mixture was added with wet palladium on carbon (200 mg) under nitrogen. The reaction was carried out for 10 hours under a hydrogen atmosphere of 50 psi. The reaction mixture was concentrated by filtration to give t-butyl(2S)-2-[(2-amino-4-fluoro-anilinyl)methyl]pyrrolidine-1-carboxylate (9 g, crude).
  • Step A To a solution of N-Cbz-L-valine (50.00 g, 200.59 mmol) in 500 ml of toluene was added DMF (146.61 mg, 2.01 mmol), then oxalyl chloride was added dropwise at 10 to 20 degrees Celsius ( 30.55 g, 240.71 mmol). After stirring at 10 to 30 ° C for 16 hours, the solvent was concentrated under reduced pressure. The crude product N-Cbz-L-prolyl chloride (53.70 g, 200.59 mmol) was used directly in the next step.
  • Step B To a solution of 6-fluoroindole (40.66 g, 300.88 mmol) in 300 ml of toluene was added dropwise ethylmagnesium bromide (3 mol/L, 106.98 ml) at -4 to 5 °C. After the addition was completed, the reaction liquid was stirred at -4 to 5 ° C for 30 minutes, and then a solution of N-Cbz-L-prolyl chloride (53.70 g, 200.59 mmol) in 200 ml of toluene was added dropwise at 0 to 10 °C. The reaction solution was stirred at 20 to 30 ° C for 2 hours.
  • ethylmagnesium bromide 3 mol/L, 106.98 ml
  • Acetic acid was added to the reaction solution at a temperature of not more than 30 ° C to adjust the pH to 3, and then quenched by adding 1 liter of water, and 1 liter of ethyl acetate was added for extraction. After separation, the organic phase was dried over anhydrous sodium sulfate. Filtration and concentration of the filtrate under reduced pressure gave a crude product.
  • Step C To a benzyl (2S)-2-(6-fluoro-1H-indole-3-carbonyl)pyrrolidine-1-carboxylate (58.00 g, 158.31 mmol) at 5 to 15 °C LiBH 4 (2 mol/L, 158.31 ml) was added dropwise to a solution of 600 ml of tetrahydrofuran, and stirred at this temperature for 2.5 hours. The reaction solution was then cooled to 5 ° C, and methanesulfonic acid (27.39 g, 284.96 mmol) was added dropwise over 30 minutes.
  • Step D A mixture of DMF (7.78 g, 106.41 mmol) and phosphorus oxychloride (17.68 g, 115.31 mmol) was stirred at 0 ° C for 30 min. Then (S)-benzyl 2-((6-fluoro-1H-3-indolyl)methyl)pyrrolidine-1 -carboxylate (12.50 g, 35.47 mmol) in 100 ml of 1 was added at this temperature. , 2-dichloroethane solution.
  • Step A Benzyl (2S)-2-[(6-fluoro-2-formyl-1H-indol-3-yl)methyl]pyrrolidin-1-carboxylate (7 g, crude) And tert-butyl(2S)-2-[(2-amino-4-fluoro-anilino)methyl]pyrrolidine-1-carboxylate (6.26 g, 20.24 mmol) was dissolved in DMF (30 mL) Water (1 ml) was then added to Oxone (8.4 g, 55.2 mmol) in one portion at 25 °C.
  • Step B Benzyl (2S)-2-[[2-[1-[[(2S)-1-tert-butoxycarbonylpyrrolidin-2-yl]methyl]-5-fluoro-benzo Imidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl]pyrrolidin-1-carboxylate (1 g, 1.49 mmol) in methanol (30 mL) and ethyl acetate (30 ml), the mixture was added with wet palladium on carbon (100 mg) under nitrogen. Hydrogenation was carried out at 40 psi for 3 hours.
  • Step C tert-Butyl (2S)-2-[[5-fluoro-2-[6-fluoro-3-[[(2S)-pyrrolidin-2-yl]methyl]-1H-indole- 2-yl]benzimidazole-1-yl]methyl]pyrrolidine-1-carboxylate (610 mg, 1.14 mmol) dissolved in dioxane hydrochloride (3 mL, 4 mol/L) at room temperature Stir for half an hour.
  • Step D To a stirred solution of N-Boc-L-proline (743.53 mg, 3.42 mmol) in DMF (2 mL), N-methylmorpholine (691.87 mg, 6.84 mmol) and HATU (1.52) Grams, 3.99 mmol), stirred at room temperature for 10 minutes, then 5-fluoro-2-[6-fluoro-3-[[(2S)-pyrrolidin-2-yl]methyl]]-1H-indole 2-yl]-1-[[(2S)-pyrrolidin-2-ylmethyl]benzimidazole (579.6 mg, hydrochloride) was dissolved in DMF (2 mL).
  • Step E tert-Butyl N-[(1S)-1-[(2S)-2-[[2-[1-[[(2S)-1-[(2S)-2-)) Carbonylamino)-3-methyl-butyryl]pyrrolidin-2-yl]methyl]-5-fluoro-benzoimidazole-2-yl]-6-fluoro-1H-indol-3-yl] Methyl]pyrrolidine-1-carbonyl]-2-methyl-propyl]carbamate (950 mg, 1.14 mmol) dissolved in dioxane hydrochloride (2 mL, 4 mol/L) at room temperature Stir for half an hour.
  • Step G tert-butyl N-[(1S)-2-[[(1S)-1-[(2S)-2-[[2-[1-[[(2S)-1-[(2S) )-2-[[(2S)-2-[tert-butoxycarbonyl(methyl)amino]propionyl]amino]-3-methyl-butyryl]pyrrolidine-2-yl]methyl ]-5-fluoro-benzoimidazole-2-yl]-6-fluoro-1H-indol-3-yl]methyl]pyrrolidine-1-carbonyl]-2-methyl-propyl]amino] 1-methyl-2-oxo-ethyl]-N-methyl-carbamate (910 mg, 906.18 ⁇ mol) was dissolved in dichloromethane (2 mL). Millimol), stirred at room temperature for half an hour. The reaction mixture was concentrated under reduced vacuo.
  • Step A Benzyl (2S)-2-[[2-[1-[[(2S)-1-tert-butoxycarbonylpyrrolidin-2-yl]methyl]-5-fluoro-benzo Imidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl]pyrrolidine-1-carboxylate (3 g, 4.48 mmol) was dissolved in dioxane hydrochloride (10 mL, 4 mol / liter), stirring at room temperature for half an hour. The reaction mixture was concentrated under reduced vacuo.
  • Step B To a stirred solution of N-Boc-L-proline (385.46 mg, 1.65 mmol) in DMF (1 mL), N-methylmorpholine (333.77 mg, 3.3 mmol) and HATU (690.08) Mg, 1.81 mmol), stirred at room temperature for 30 minutes, then benzyl (2S)-2-[[6-fluoro 2-[5-fluoro-1-[[(2S)-pyrrolidin-2-yl] Methyl]benzimidazol-2-yl]-1H-indol-3-yl]methyl]pyrrolidine-1-carboxylate (500 mg, crude) was dissolved in DMF (1 mL).
  • Step C Benzyl (2S)-2-[[2-[1-[[(2S)-1-[(2S)-2-(tert-butoxycarbonylamino)-3-methyl-) Butyryl]pyrrolidin-2-yl]methyl]-5-fluoro-benzoimidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl]pyrrolidine-1-carboxylic acid
  • the ester (570 mg, 741.33 micromoles) was dissolved in MeOH (15 mL) and ethyl acetate (15 mL). Hydrogenation was carried out at 45 psi for 16 hours.
  • Step D To a stirred solution of N-Boc-L-n-butyric acid (243.34 mg, 1.2 mmol) in DMF (1 mL), N-methylmorpholine (302.77 mg, 2.99 mmol) 569.06 mg, 1.5 mmol), then tert-butyl N-[(1S)-1-[(2S)-2-[[5-fluoro-2-[6-fluoro-3-[[(2S)-) Pyrrolidin-2-yl]methyl]-1H-indol-2-yl]benzimidazol-1-yl]methyl]pyrrolidin-1-carbonyl]-2-methyl-propyl]carbamic acid The ester (380 mg, crude) was dissolved in DMF (1 mL).
  • Step E tert-Butyl N-[(1S)-1-[(2S)-2-[[2-[3-[[(2S)-1-[(2S)-2-(tero-butoxy) Carbonylamino)butyryl]pyrrolidin-2-yl]methyl]-6-fluoro-1H-indol-2-yl]-5-fluoro-benzoimidazol-1-yl]methyl]pyrrolidine- 1-Carbonyl-2-methyl-propyl]carbamate (380 mg, 463.43 ⁇ mol) was dissolved in dioxane hydrochloride (2 mL, 4 mol/L) and stirred at room temperature for half an hour.
  • Step F To a stirred solution of N-Boc-N-methyl-L-alanine (281.68 mg, 1.39 mmol) in DMF (1 mL), N-methylmorpholine (233.65 mg, 2.31 m Mole) and HATU (439.15 mg, 1.15 mmol), then (2S)-2-amino-1-[(2S)-2-[[2-[3-[[(2S)-1-[( 2S)-2-aminobutyryl]pyrrolidin-2-yl]methyl]-6-fluoro-1H-indol-2-yl]-5-fluoro-benzoimidazol-1-yl]methyl] Pyrrolidin-1-yl]-3-methyl-butan-1-one (320 mg, 461.98 ⁇ mol, hydrochloride) was dissolved in DMF (1 mL).
  • Step G tert-Butyl N-[(1S)-2-[[(1S)-1-[(2S)-2-[[2-[3-[[(2S)-1-[(2S) -2-[[(2S)-2-[tert-butoxycarbonyl(methyl)amino]pyrrolidine]amino]butyryl]pyrrolidin-2-yl]methyl]-6-fluoro-1H- Indole-2-yl]-5-fluoro-benzoimidazol-1-yl]methyl]pyrrolidin-1-carbonyl]-2-methyl-propyl]amino]-1-methyl-2- Oxy-ethyl]-N-methyl-carbamate (450 mg, 454.46 ⁇ mol) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL, 27.01 mmol). hour. The reaction mixture was concentrated under reduced vacuo.
  • Examples 3 to 12 can be obtained by referring to the preparation reaction scheme of Example 2.
  • Step A Dilute N-Boc-trans-4-hydroxy-L-valine methyl ester (262.00 g, 1.07 mol) in 2.5 liters of dichloromethane at -78 ° C under nitrogen atmosphere. DAST (258.28 g, 1.60 mol) was added. After stirring at -78 ° C for 3 hours, the temperature was raised to 10 to 20 ° C and stirred for another 15 hours. The reaction solution was poured into 3 liters of saturated sodium hydrogencarbonate solution at 0 ° C and quenched, and extracted twice with 6 liters of dichloromethane.
  • Step B Add LiBH 4 (16.00 g) to a solution of N-Boc-cis-4-fluoro-L-valine methyl ester (80.00 g, 323.55 mmol) in 1 liter of tetrahydrofuran at 0 to 5 °C. , 734.62 mmol). After the reaction mixture was stirred at 10 to 20 ° C for 16 hours, it was quenched by adding 1500 ml of aq. The combined organic phases were concentrated under reduced pressure to give a crude material.
  • Step C Adding phthalic acid to a solution of N-Bco-cis-4-fluoro-L-prolinol (66.00 g, 301.03 mmol, crude) in 700 ml of tetrahydrofuran at 10 to 20 ° C Amine (46.50 g, 316.08 mmol) and triphenylphosphine (82.90 g, 316.08 mmol). DIAD (63.91 g, 316.08 mmol) was then added dropwise at 0-10 degrees Celsius. After the reaction mixture was stirred at 10 to 20 ° C for 16 hours, the solvent was removed under reduced pressure.
  • Step D To tert-butyl(2S,4S)-2-[(1,3-dioxo-2-isoindolyl)methyl]-4-fluoro-pyrrolidine-1-carboxylate (133.00 g , 381.78 mmol, crude) in 1 liter of ethanol solution was added hydrazine hydrate (48.75 g, 954.44 mmol). After reacting at 60 ° C for 2 hours, cool to room temperature, add 1 liter of dichloromethane to dilute, filter, filter The cake was washed with EtOAc (EtOAc m. 2S,4S)-2-(Aminomethyl)-4-fluoro-pyrrolidine-1-carboxylate (112.00 g, crude).
  • Step E To a solution of (2S,4S)-2-(aminomethyl)-4-fluoro-pyrrolidine-1-carboxylate (66.00 g, crude) in 1 liter of acetonitrile at 10 to 20 °C Add 1,4-difluoro-2-nitro-benzene (45.70 g, 287.26 mmol) and add potassium carbonate (3.58 g, 604.76 mmol). After reacting at 80 ° C for 2 hours, it was cooled to 10 to 20 ° C. The reaction solution was filtered, and the filtrate was concentrated to give a crude material. The crude product was dissolved in 500 ml of methyl tert-butyl ether.
  • Step F To a tert-butyl(2S,4S)-4-fluoro-2-[(4-fluoro-2-nitro-benzylamino)methyl]pyrrolidine-1-carboxylate under nitrogen atmosphere (20.00 Pd/C (10%, 2 g) was added to a mixed solution of 200 ml of methanol and 1 liter of ethyl acetate in gram, 55.97 mmol. The system was replaced three times with hydrogen and stirred for 4 hours at 25 to 30 degrees Celsius under a hydrogen atmosphere at 40 psi.
  • Step A N-Cbz-trans-4-hydroxy-L-proline methyl ester (21.00 g, 57.14 mmol) was dissolved in anhydrous dichloromethane (90.00 mL), then at -78. DAST (21.65 g, 134.29 mmol) was added dropwise. After stirring at -78 °C for 3 hours, it was stirred at room temperature (25 ° C) for 15 hours. The reaction was quenched with saturated sodium bicarbonate (600 mL). . The organic layer was washed with brine (600 ml ⁇ 1), dried over anhydrous sodium sulfate. The residue was purified by silica gel column chromatography (EtOAc /EtOAcEtOAcEtOAcEtOAcEtOAcEtOAc %).
  • Step B N-Cbz-cis-4-fluoro-L-proline methyl ester (14.00 g, 49.03 mmol) was dissolved in tetrahydrofuran (70.00 ml), then lithium hydroxide solution (1 mol) was added at 25 degrees. / liter, 69.13 ml). The reaction was stirred at 25 ° C for 14 hours. After adjusting to pH 3 with 1 mol/L hydrochloric acid solution, 200 ml of water was added, followed by extraction with ethyl acetate (200 ml ⁇ 4).
  • Step C N-Cbz-cis-4-fluoro-L-proline (5.00 g, 18.39 mmol) was dissolved in toluene (25.00 mL) then DMF (13.44 mg, 183.91 Molar, 14.15 microliters) and oxalyl chloride (2.80 grams, 22.07 mmol, 1.93 ml). After stirring at 25 °C for 1 hour, it was concentrated under reduced pressure at 45 °m toield of Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound Compound
  • Step D 6-Fluoro-1H-indole (5.01 g, 37.10 mmol) was dissolved in toluene (60.00 ml), and ethyl bromide was added dropwise in an ice/acetone bath (-4 degrees) under nitrogen. 3 mol/L, 12.55 ml), and the reaction solution was stirred at -4 ° for 1 hour. Then, benzyl N-Cbz-cis-4-fluoro-L-prolyl chloride (5.30 g, crude) was dissolved in toluene and added dropwise to the above reaction mixture at -4. The mixture was stirred at -4 degrees for 2 hours and stirred at 25 degrees for 14 hours.
  • Step E Dissolving benzyl (2S,4S)-4-fluoro-2-(6-fluoro-1H-indole-3-carbonyl)pyrrolidine-1-carboxylate (4.40 g, 11.45 mmol) Tetrahydrofuran (44.00 ml) was then slowly added dropwise at 25 °C with a solution of lithium borohydride (2 mol/L, 11.45 ml) (20 min drop). After the mixture was stirred at 25 ° C for 16 hours, methanesulfonic acid (2.04 g, 21.18 mmol, 1.51 ml) was added dropwise at 0 ° and stirring was continued for 2 hours.
  • Step F Phosphorus oxychloride (534.02 mg, 3.48 mmol) was added dropwise to DMF (254.56 mg, 3.48 mmol) under a nitrogen atmosphere, and the mixture was stirred at 0 ° for one hour. Then benzyl (2R,4S)-4-fluoro-2-[(6-fluoro-1H-indol-3-yl)methyl]pyrrolidine-1-carboxylate (1.00 g, 2.32 mmol) After dissolving in 1,2-dichloroethane (5.00 ml), the above mixture was added dropwise at 0 °C.
  • Step A Benzyl (2R,4S)-4-fluoro-2-[(6-fluoro-2-formyl-1H-indol-3-yl)methyl]pyrrolidine-1-carboxylate ( 1.00 g, 2.52 mmol, tert-butyl(2S,4S)-2-[(2-amino-4-fluoro-aniline)methyl]-4-fluoro-pyrrolidine-1-carboxylate (800.00 mg , 2.39 mmol), dissolved in a mixed solvent of DMF (12.00 ml) and water (1.00 ml), then added Oxone (1.09 g, 7.18 mmol) at 25 ° C, and the mixture was stirred at 25 ° C for 1 hour.
  • Step B Benzyl (2R,4S)-2-[[2-[1-[[(2S,4S)-1-tert-butoxycarbonyl-4-fluoro-pyrrolidin-2-yl]methyl] -5-fluoro-benzoimidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl]-4-fluoro-pyrrolidine-1-carboxylate (1.40 g, 1.69 mmol) ) A mixture of ethyl acetate (50.00 ml) and methanol (10.00 ml) was added, then palladium on carbon (200.00 mg, 10%) was added in the next portion under nitrogen atmosphere.
  • Step C tert-Butyl (2S,4S)-4-fluoro-2-[[5-fluoro-2-[6-fluoro-3-[[(2R,4S)-4-fluoropyrrolidine-2- Methyl]methyl]-1H-indol-2-yl]benzimidazol-1-yl]methyl]pyrrolidine-1-carboxylate (200.00 mg, 314.90 ⁇ mol) is soluble in 1,4-dioxane Six rings (4.00 ml), then dropwise hydrogen chloride / 1,4-dioxane (4 mol / liter, 3.60 ml) at 25 ° C, the mixture was stirred at 25 ° C for 30 minutes, and the solvent was distilled off under reduced pressure to give 5- Fluor-2-[6-fluoro-3-[[(2R,4S)-4-fluoropyrrolidin-2-yl]methyl]-1H-indol-2-yl]-1-[[(2R, 4S)
  • Step D N-Boc-L-n-butyric acid (158.65 mg, 780.65 ⁇ mol) was dissolved in DMF (3.00 mL), and N-methylmorpholine (126.34 mg, 1.25 mmol, 137.33 ⁇ L) and HATU (124.67 mg, 327.87 micromolar). The mixture was stirred at 25 degrees for 30 minutes.
  • Step E tert-Butyl-N-[(1S)-1-[(2R,4S)-2-[[2-[1-[[(2S,4S)-1-[(2S)-2-) (tert-Butoxycarbonylamino)butyryl]-4-fluoro-pyrrolidin-2-yl]methyl]-5-fluoro-benzoimidazol-2-yl]-6-fluoro-1H-indole-3 -yl]methyl]-4-fluoro-pyrrolidine-1-carbonyl]-n-propyl]carbamate (110.00 mg, 127.65 ⁇ mol) dissolved in 1,4-dioxane (1.00 mL), then The mixture of hydrogen chloride / 1,4-dioxane (4 mol / liter, 1.00 ml) was added dropwise at 25 °C for 30 minutes, and the solvent was distilled off under reduced pressure at 45 ° to give a pale yellow solid (2S)-2 -amin
  • Step F N-Boc-N-methyl-L-alanine (69.46 mg, 341.79 ⁇ mol) was dissolved in DMF (2.00 mL), and NMM (92.19 mg, 911.44 micromoles, 100.21 ⁇ l) was added at 25 degrees.
  • Step G tert-Butyl-N-[(1S)-2-[[(1S)-1-[(2R,4S)-2-[[2-[1-[[(2S,4S)-1 -[(2S)-2-[[(2S)-2-[tert-butoxycarbonyl(methyl)amino]propionyl]amino]butyryl]-4-fluoro-pyrrolidin-2-yl]methyl ]-5-fluoro-benzoimidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl]-4-fluoro-pyrrolidine-1-carbonyl]propyl]amino]-1 -Methyl-2-oxo-ethyl]-N-methyl-carbamate (90.00 mg, 79.14 ⁇ mol) was dissolved in dichloromethane (1.00 mL) then trifluoroacetic acid (1.54 g) , 13.51 mmol, 1.00
  • Examples 14 to 17 can be obtained by referring to the preparation reaction scheme of Example 13.
  • Step A Benzyl (2R,4S)-2-[[2-[1-[[(2S,4S)-1-tert-butoxycarbonyl-4-fluoro-pyrrolidin-2-yl]methyl ]-5-fluoro-benzoimidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl]-4-fluoro-pyrrolidine-1-carboxylate (400.00 mg, 510.10 micro) Ethyl acetate was dissolved in ethyl acetate (1.00 mL) then HCl / EtOAc (4 mol / liter, 1.00 mL).
  • Step B N-Boc-L proline (206.03 mg, 948.30 ⁇ mol) was dissolved in DMF (2.00 mL), and N-methylmorpholine (239.80 mg, 2.37 mmol, 260.65 ⁇ l) was added dropwise at 20 °C. And HATU (396.63 mg, 1.04 mmol). The mixture was stirred at 20 degrees for 30 minutes.
  • Step C Benzyl (2R,4S)-2-[[2-[1-[[(2S,4S)-1-[(2S)-2-(tert-butoxycarbonylamino)-3-) -butyryl]-4-fluoro-pyrrolidin-2-yl]methyl]-5-fluoro-benzoimidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl] 4-fluoro-pyrrolidine-1-carboxylate (400.00 mg, 455.73 ⁇ mol) was dissolved in ethyl acetate (30.00 mg) and methanol (5.00 mg).
  • Step D N-Boc-L-n-butyric acid (160.95 mg, 791.96 ⁇ mol) was dissolved in DMF (1.00 ml), and N-methylmorpholine (120.16 mg, 1.19 mmol, 130.61 ⁇ m) was added dropwise at 20 °C. l) and HATU (376.41 mg, 989.95 micromolar).
  • Step E tert-Butyl N-[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-) tert-Butoxycarbonylamino)butyryl]-4-fluoro-pyrrolidin-2-yl]methyl]-6-fluoro-1H-indol-2-yl]-5-fluoro-benzimidazole-1- Methyl]methyl]-4-fluoro-pyrrolidine-1-carbonyl]-2-methyl-propyl]carbamate (320.00 mg, 321.51 ⁇ mol) was dissolved in 1,4-dioxane (2.00) HCl), then HCl/1,4-dioxane (4 mol/L, 2.00 mL) was added dropwise at 25 °C.
  • Step F N-Boc-N-methyl-L-alanine (228.43 mg, 1.12 mmol) was dissolved in DMF (1.00 mL), and N-methylmorpholine (194.90 mg, 1.93) was added dropwise at 25 degrees. Millimol, 211.85 microliters) and HATU (451.80 mg, 1.19 mmol), and the mixture was stirred at 25 degrees for 30 minutes.
  • Step G tert-Butyl-N-[(1S)-2-[[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R)4S)-1 -[(2S)-2-[[(2S)-2-[tert-butoxycarbonyl(methyl)amino]propionyl]amino]butyryl]-4-fluoro-pyrrolidine-2-]methyl] -6-fluoro-1H-indol-2-yl]-5-fluoro-benzoimidazol-1-yl]methyl]-4-fluoro-pyrrolidine-1-carbonyl]-2-methyl-propyl Amino]-1-methyl-2-oxo-ethyl]-N-methyl-carbamate (260.00 mg, 222.97 ⁇ mol) was dissolved in dichloromethane (1.00 mL) and then dropped at 25 ° Trifluoroacetic acid (1.54 g, 13.51 mmol, 1.00
  • Examples 19 to 36 can be obtained by referring to the preparation reaction scheme of Example 18.
  • Step A 30 of dissolved (S)-benzyl-2-((6-fluoro-1hydro-3-indolyl)methyl)pyrrolidine-1-carboxylate (5 g, 14.19 mmol) DMAP (34.67 mg, 283.77 ⁇ mol) was added to a solution of methylene chloride, and then BOC anhydride (3.25 g, 14.90 mmol) dissolved in 20 ml of dichloromethane was added dropwise. After the addition was completed, the reaction solution was stirred at 10 to 20 ° C for 16 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure.
  • Step B Dissolved S)-tert-butyl 3-((1-((benzyloxy))))-pyrrolidine)methyl)-6-fluoro-1H-indole at -70 ° C under a nitrogen atmosphere LDA (2 mol / liter, 13.79 ml) was added dropwise to a solution of hydrazine-l-carboxylate (6.5 g, 13.79 mmol) in 120 ml of tetrahydrofuran, and the mixture was stirred at this temperature for 15 minutes. Then dry ice (13.79 mmol) was added.
  • LDA 2 mol / liter, 13.79 ml
  • Step C (S)-3-((1-((benzyloxy)carbonyl)) 2-pyrrolidine)methyl)-1-(tert-butoxycarbonyl)-dissolved at 5-20 ° C
  • 6-fluoro-1H-indole-2-carboxylic acid 2.5 g, 5.03 mmol
  • trifluoroacetic acid 135.06 ⁇ RTIgt
  • the resulting reaction solution was stirred at 5 to 20 ° C for 16 hours.
  • the reaction was concentrated under reduced pressure to remove dichloromethane and most of trifluoroacetic acid. The residue was diluted with dichloromethane and washed with aq.
  • Step D Dissolving (S)-3-((1-((benzyloxy)carbonyl)) 2-pyrrolidine)methyl)-1-(tert-butoxycarbonyl)-6-fluoro-1H-indole -2 carboxylic acid (1.5 g, 3.78 mmol) in 15 ml of dichloromethane was added pyridine (61.95 mmol, 5 ml), tert-butyl (2S,4S)-2-[(2-amino-4) -Fluoro-anilino)methyl]-4-fluoro-1pyrrolidine-carboxylate (1.11 g, 3.40 mmol) and EDCI (1.09 g, 5.67 mmol).
  • reaction solution was stirred at 10 to 25 ° C for 16 hours. After completion of the reaction, 50 ml of a 1 mol/L hydrochloric acid solution was added to the reaction mixture to quench the reaction, followed by extraction with 50 ml of dichloromethane. The organic layer was washed with 50 ml of saturated brine and dried over anhydrous sodium sulfate.
  • Step E (2S,4S)-tert-Butyl 2-(((2-(3-((()))))))))) Fluoro-1H-indole-2-carboxamide)-4-fluorophenyl)amino)methyl)-4-fluoropyrrolidine-1-carboxylate (500 mg, 708.46 ⁇ mol) dissolved in 5 ml of acetic acid The solution was then stirred at 85 degrees C for 4 hours. After the reaction was completed, the reaction mixture was concentrated under reduced pressure to remove solvent.
  • Step F (2S,4S)-tert-butyl 2-((2-(3-((S)-1-((benzyloxy))))-2-pyrrolidine) was dissolved in a nitrogen atmosphere.
  • -6-fluoro-1Hero-2 ⁇ )-5-fluoro-1H-1-benzimidazole)methyl)-4-fluoropyrrolidine-1-carboxylate (300.00 mg, 436.21 ⁇ mol)
  • a solution of 5 ml of methanol was added 0.1 g of palladium on carbon (10%). After the reaction system was replaced with 3 hydrogen to hydrogen atmosphere, it was stirred for 16 hours under a hydrogen atmosphere (15 psi) of 30 to 35 ° C.
  • Step G To a solution of N-Boc-L-n-butyric acid (122.13 mg, 514.8 ⁇ mol) in 1 ml of DMF was added HATU (195.74 mg, 514.8 ⁇ mol) and N-methylmorpholine (1.03 mol, 113.20 microliters). The resulting reaction solution was stirred at 10 to 20 ° C for 15 minutes.
  • Step H To 2S,4S)-tert-butyl 2-((2-(3-((())))))))) 2-pyrrolidine)methyl)-6-fluoro-1H-2-indole-5-fluoro-1H-1-benzimidazole)methyl)-4-fluoropyrrolidine-1-carboxylate TPA (13.15 mmol, 1 mL) was added to a solution of (300 mg, crude) m. The reaction solution was stirred at 10 to 20 ° C for 2 hours. After completion of the reaction, the reaction solution was concentrated under reduced pressure to remove solvent and most of trifluoroacetic acid.
  • Step I NMM (953.28 micromoles, 104.80 microliters) and HATU (181.23 mg, 476.64 micromoles) were added to a solution of N-Boc-L proline (103.55 mg, 476.64 micromoles) in 1 ml of DMF.
  • Step J To-tert-butyl N-[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2S)-1-[(2S)-2-(benzyloxy) Carbonylamine) tert-butoxy]2-pyrrolidine]methyl]-6-fluoro-1H-2-indyl]-5-fluoro-1-benzimidazole]methyl]-4-fluoro-pyrrolidine HBr/AcOH (229.36 micromolar, 1.00 ml) was added to a solution of 1-carbonylcarbonyl-2-methyl-propylpropyl]carboxylate (200.00 mg, 229.36 micromoles, crude) in 1 ml of dichloromethane.
  • the reaction solution was stirred at 10 to 20 ° C for 1 hour, and then concentrated under reduced pressure to remove solvent.
  • the crude product was diluted with 30 ml of water and extracted with 30 ml of methyl t-butyl ether.
  • the aqueous phase was adjusted to pH -8, and then extracted with 50 ml of ethyl acetate.
  • Step K To a solution of N-Bco-N-methyl-L-alanine (95.60 mg, 470.40 ⁇ mol) in 1 mL of DMF was added N-methylmorpholine (103.43 ⁇ L, 940.80 ⁇ mol) and HATU (178.86 mg, 470.40 micromolar). The reaction was stirred at 25 ° C for 15 minutes.
  • EtOAc EtOAc EtOAc: )-2-[[2-[3-[[(2S)-1-[(2S)-2-[[(2S)-2-[tert-butoxycarbonyl(methyl)amine]propanoyl]amine] Butoxy]2-pyrrolidine]methyl]-6-fluoro-1H-2-indole-5-fluoro-1-benzopyrazole]methyl]-4-fluoro-pyrrolidine-1-carbonyl ]-2-Methyl-propyl]amine]-1-methyl-2-oxo-ethyl]-N-methyl-carbamate (80.00 mg, 78.56 ⁇ mol, 50.10%).
  • Step L To-tert-butyl N-[(1S)-2-[[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2S)-1-[( 2S)-2-[[(2S)-2-[tert-butoxycarbonyl(methyl)amine]propionyl]amine]butoxy]2-pyrrolidine]methyl]-6-fluoro-1H-2- ⁇ ]-5-fluoro-1-benzopyrazole]methyl]-4-fluoro-pyrrolidine-1-carbonyl]-2-methyl-propyl]amine]-1-methyl-2-oxo To a solution of 2-ethyl]-N-methyl-carbamate (80.00 mg, 78.56 ⁇ mol) in 2 mL of dichloromethane was added 1 mL of trifluoroacetic acid.
  • Example 37 (38.22 mg, 43.39 micromoles, 54.68%, hydrochloride).
  • Step A tert-Butyl-(2S)-2-[(2-amino-4-fluoro-anilino)methyl]pyrrolidine-1-carboxylate (4.13 g, 13.36 mmol) was dissolved in DMF ( Benzyl (2R,4S)-4-fluoro-2-[(6-fluoro-2-formyl-1H-indole-) was added at 0 °C in a mixed solvent of 30.00 ml) and H 2 O (2.00 ml). 3-yl)methyl]pyrrolidine-1-carboxylate (3.80 g, 6.68 mmol).
  • Oxone (3.05 g, 20.03 mmol) was added at 0 °C, and the mixture was stirred at 15 °C for 16 hours, then diluted with ethyl acetate (300 mL) and water (300 mL).
  • Step B Benzyl (2R,4S)-2-[[2-[1-[[(2S)-1-tert-butoxycarbonylpyrrolidin-2-yl]methyl]-5-fluoro-benzene And imidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl]-4-fluoro-pyrrolidine-1-carboxylate (600.00 mg, 706.65 ⁇ mol) is dissolved in 1,4 - Dioxane (6.00 ml), HCl / 1,4-dioxane (4 mol / liter, 6.00 ml) was added dropwise at 15 °C.
  • Step C N-Boc-L proline (140.99 mg, 648.95 ⁇ mol) was dissolved in DMF (2.00 mL), and N-methylmorpholine (131.28 mg, 1.30 mmol, 142.70 ⁇ l) was added at 15 °C and HATU (263.20 mg, 692.21 micromolar).
  • Step D Benzyl (2R,4S)-2-[[2-[1-[[(2S)-1-[(2S)-2-(tert-butoxycarbonylamino)-3-methyl-) Butyryl]pyrrolidin-2-yl]methyl]-5-fluoro-benzoimidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl]-4-fluoro-pyrrolidine
  • 1-carboxylic acid ester (335.00 mg, 355.71 ⁇ mol) was dissolved in a mixed solvent of ethyl acetate (20.00 mL) and methanol (4.00 mL), then palladium/carbon (100.00 mg, 10%) .
  • Step E N-Boc-L-n-butyric acid (129.88 mg, 639.07 micromoles) was dissolved in DMF (3.00 ml), and N-methylmorpholine (129.28 mg, 1.28 mmol, 140.52 ⁇ l) was added at 18 degrees. ) and HATU (259.19 mg, 681.67 micromolar).
  • Step F tert-butyl N-[(1S)-1-[(2S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-) Butoxycarbonylamino)butyryl]-4-fluoro-pyrrolidin-2-yl]methyl]-6-fluoro-1H-indol-2-yl]-5-fluoro-benzimidazol-1-yl] Methyl]pyrrolidine-1-carbonyl]-2-methyl-propyl]carbamate (346.00 mg, 412.90 ⁇ mol) was dissolved in dichloromethane (3.00 mL), then added dropwise trifluoroacetic acid (4.62 g) , 40.52 mmol, 3.00 ml), the mixture was stirred at 18 ° for 30 minutes, and then the solvent was distilled off under reduced pressure at 45 ° to give (2S)-2-amino-1-[(2S)-2-[[2-[3- [[(2R
  • Step G N-Boc-N-methyl-L-alanine (209.50 mg, 1.03 mmol) was dissolved in DMF (3.00 mL), and N-methylmorpholine (250.25 mg, 2.47 m) was added at 18 degrees.
  • Step H tert-Butyl N-[(1S)-2-[[(1S)-1-[(2S)-2-[[2-[3-[[(2R,4S)-1-[( 2S)-2-[[(2S)-2-[tert-Butoxycarbonyl(methyl)amino]propionyl]amino]butyryl]-4-fluoro-pyrrolidin-2-yl]methyl]-6 -fluoro-1H-indol-2-yl]-5-fluoro-benzoimidazol-1-yl]methyl]pyrrolidine-1-carbonyl]-2-methyl-propyl]amino]-1-methyl Benzyl-2-oxo-ethyl]-N-methyl-carbamate (200.00 mg, 168.62 ⁇ mol) was dissolved in dichloromethane (3.00 mL), trifluoroacetic acid (4.62 g, 40.52 mmol, 3.00 mL).
  • Examples 40, 41, and 42 were prepared according to the preparation procedure of Example 39.
  • Step A To a solution of N-Boc-trans-4-hydroxy-L-valine methyl ester (114.00 g, 464.79 mmol) in 1.2 liters of dichloromethane was added trichloroisocyanuric acid (113.42 g, 488.03 Millol), TEMPO (1.46 g, 9.30 mmol) was added at 0 °C. The reaction solution was stirred at 10 to 20 ° C for half an hour.
  • Step B DAST (119.40 g, 740.78 m) was added dropwise to a solution of N-Boc-4-oxo-L-valine methyl ester (106.00 g, 435.75 mmol) in 500 ml of dichloromethane at 0 ° C. A solution of methylene chloride in 500 ml of dichloromethane was added, and then ethanol (4.02 g, 87.15 mmol) was added. Stir for 18 hours at 0 to 20 degrees Celsius. Pour the reaction solution into saturated carbonic acid that is slowly poured into ice.
  • Step C Add lithium chloride (17.58) to a stirred solution of N-Boc-4,4-difluoro-L-valine methyl ester (50.00 g, 188.50 mmol) in 500 ml of tetrahydrofuran at 10 to 20 °C. Gram, 414.70 mmol) and sodium borohydride (17.83 g, 471.25 mmol). After the reaction solution was cooled to 0 ° C, 1 liter of ethanol was added dropwise. The reaction solution was stirred at 0 ° C for 1 hour, and then at 10 to 20 ° C for 17 hours.
  • Step D Add phthalimide (28.65 g) to a solution of N-Boc-4,4-difluoro-L-prolinol (44 g, crude) in 440 ml of tetrahydrofuran at 10-20 ° C. , 194.74 mmol) and triphenylphosphine (51.08 g, 194.74 mmol). Then DIAD (39.38 g, 194.74 mmol) was added dropwise. After the addition was completed, the reaction was carried out for 16 hours at 10 to 20 °C. The reaction solution was concentrated under reduced pressure to remove solvent, and then, 500 ml of water. The combined organic layers were washed with EtOAc EtOAc m.
  • Step E To-tert-butyl(2S)-2-[(1,3-dioxo-2-isoindolyl)methyl]-4,4-difluoro-pyrrolidine-1-carboxylate (69.00 To a solution of 1 liter of ethanol in gram, 188.34 mmol) was added hydrazine hydrate (24.05 g, 470.85 mmol). The reaction solution was reacted at 60 ° C for 2 hours. After cooling to room temperature, it was diluted with 1000 ml of dichloromethane, filtered, and the filter cake was washed with dichloromethane. The organic phases were combined and concentrated under reduced pressure to give a crude material.
  • Step F To a tert-butyl(2S)-2-(aminomethyl)-4,4-difluoro-pyrrolidine-1-carboxylate (44.50 g, 188.35 mmol) at 10 to 20 °C To a solution of 500 ml of acetonitrile was added 1,4-difluoro-2-nitro-benzene (29.96 g, 188.35 mmol) and potassium carbonate (52.06 g, 376.70 mmol). After reacting at 80 ° C for 2 hours, it was cooled to 10 to 20 ° C. The reaction solution was filtered, and the filtrate was concentrated to give a crude material.
  • 1,4-difluoro-2-nitro-benzene 29.96 g, 188.35 mmol
  • potassium carbonate 52.06 g, 376.70 mmol
  • Step G To a tert-butyl (2S)-4,4-difluoro-2-[(4-fluoro-2-nitro-anilino)methyl]pyrrolidine-1-carboxylate under a nitrogen atmosphere (10.00 g, 26.64 mmol) in 300 ml of methanol was added Pd/C (10%, 1 g). The reaction system was replaced with hydrogen three times and then reacted at 30 to 40 psi, 25 to 30 degrees for 2 hours.
  • Step A To a solution of N-Cbz-cis-4-hydroxy-L-proline methyl ester (150.00 g, 537.08 mmol) in dichloromethane (1.5 L) was added trichloroisocyanuramide with stirring. Acid (131.06 g, 563.93 mmol), followed by careful addition of TEMPO (8.45 g, 53.71 mmol) at 0 °C. The reaction was stirred at 15 ° C for 30 minutes and filtered. The filtrate was quenched with aqueous sodium thiosulfate (300 mL) then EtOAc (EtOAc) The combined organic layers were washed with EtOAc EtOAc. The residue was purified by silica gel column chromatography (EtOAc:EtOAc:EtOAc:EtOAc 324.59 mmol, 60.44%).
  • Step B Add DAST to a stirred solution of N-Cbz-4-oxo-L-valine methyl ester (75.00 g, 270.49 mmol) in anhydrous dichloromethane (650 mL) at 0 ° C. (74.12 g, 459.83 mmol, 60.75 ml) in dichloromethane.
  • Ethanol (249.23 mg, 5.41 mmol, 315.48 ⁇ l) was then added to the reaction mixture.
  • the reaction was stirred at 15 ° C for 12 hours and then quenched with water (300 mL). It was diluted with water (150 ml) and extracted with ethyl acetate (500 mL). The combined organic layers were washed with EtOAc EtOAc EtOAc m.
  • Step C To a solution of N-Cbz-4,4-difluoro-L-valine methyl ester (78 g, crude) in tetrahydrofuran (500 mL), EtOAc (EtOAc) A milliliter of aqueous solution (100 ml). The reaction solution was stirred at 15 ° C for 2 hours, then concentrated under reduced pressure to remove tetrahydrofuran, and then the aqueous solution was washed with dichloromethane (200 ml). The aqueous phase was adjusted to pH 1 with hydrochloric acid (1M, ca. 40 mL) and extracted with dichloromethane (400 mL).
  • EtOAc EtOAc
  • Step D To a stirred solution of oxalyl chloride (26.70 g, 210.35 mmol, 18.41 ml) in dry toluene (50 ml), DMF (768.72 mg, 10.52 mmol, 809.18 uL, 0.10 eq) . After the dropwise addition was completed, the mixture was stirred for 30 minutes. Then, N-Cbz-4,4-difluoro-L-proline was added to the reaction solution at 0 ° C, and the reaction mixture was stirred at 25 ° C for 5 hours. The reaction solution was dried to give the product N-Cbz-4,4-difluoro-L-prolyl chloride, which was dissolved in toluene and used directly in the next step.
  • Step E Add Ethyl Grignard reagent (3M) to a stirred solvent of 6-fluoroindole (21.32 g, 157.76 mmol) in toluene (100 ml) and chlorobenzene (80 ml) at 0 °C. , 54.34 ml) and control added in 30 minutes. After the addition, the reaction solution was stirred at this temperature for 30 minutes, and then a toluene solution of N-Cbz-4,4-difluoro-L-prolyl chloride was added to the reaction solution at 0 °C. The reaction mixture was stirred at 25 ° C for 5 hours.
  • Step F To (S)-benzyl 4,4-difluoro-2-(6-fluoro-1H-indole-3-carbonyl)pyrrole-1-carboxylate (30 g, 64.87 m) with stirring. A solution of tetrahydrofuran (200 ml) was added to a solution of lithium borohydride (2M, 64.87 mL). The reaction solution was stirred at 15 ° C for 4 hours. Then methanesulfonic acid (11.53 g, 120.00 mmol, 8.54 ml) was added thereto and stirred at 15 ° C for 12 hours.
  • Step G Phosphorus oxychloride (8.15 g, 51.50 mmol, 3.96 ml) was added to stirred DMF (3.76 g, 51.50 mmol, 3.96 mL). The reaction solution was reacted at 0 ° C for 1 hour, and dissolved in 1,2-dichloroethane (20 ml) (R)-benzyl 4,4-difluoro-2-((6-fluoro-1H-indole) -3-yl)methyl)pyrrole-1-carboxylate (10 g, 25.75 mmol) Into the reaction solution, and keep the dropping temperature at 0 degrees Celsius. The reaction solution was reacted at 15 ° C for 11 hours.
  • Step A Benzyl (2R)-4,4-difluoro-2-[(6-fluoro-2-formyl-1H-3-indole)methyl]pyrrolidine-1 at 10 to 20 °C Addition of (S)-tert-butyl 2-(((2-amino-4-fluorophenyl)amino)methyl)-carboxylate (9.04 g, 8.69 mmol) in 20 mL of DMF and 0.2 mL of aqueous 4,4-difluoropyrrolidine-1-carboxylate (3.00 g, 8.69 mmol) and potassium hydrogen persulfate (2.64 g, 17.38 mmol).
  • Step B to (R)-benzyl (2R)-2-[[2-[1-[[(2S)-1-tert-butanoyl-4,4-difluoro-2) at 10 to 20 degrees Celsius -pyrrolidine]methyl]-5-fluoro-2-benzimidazole]-6-fluoro-1H-3-indole]methyl]-4,4-difluoro-pyrrolidine-1-carboxylate ( To a solution of 600.00 mg, 808.93 micromoles of 5 ml of dichloromethane was added HBr/AcOH (89.74 mg, 808.93 micromoles). The reaction solution was stirred at 10 to 20 ° C for 1 hour.
  • Step C To a solution of N-Boc-L-n-butyric acid (227.73 mg, 1.12 mmol) in 2 mL of DMF was added N-methylmorpholine (226.69 mg, 2.24 mmol) and HATU (426.08 mg, 1.12 Millimoles). The reaction solution was stirred at 10 to 20 ° C for 30 minutes. Then add 1-(((S)-4,4-difluoro-2-pyrrolidine)methyl)-2-(3-(((R)-4,4-difluoro-2-pyrrolidine)) 6-fluoro-1H-2-indole-5-fluoro-1H-benzimidazole (250.00 mg, 373.52 mmol).
  • Step D tert-Butyl N-[(1S)-1-[(2R)-2-[[2-[1-[[(2S)-1-[(2S)-2-(tert-butoxy) Carbonylamino)butyryl]-4,4-difluoro-2-pyrrolidine]methyl]-5-fluoro-2-benzimidazole]-6-fluoro-1H-3-indole]methyl] Methyl-4,4-difluoro-pyrrolidin-1-carbonyl]propyl]carboxylate (100 mg, 113.91 ⁇ mol) dissolved in HCl / dioxane (113.91 ⁇ mol, 5.00 mL) The reaction was stirred at 20 ° C for 5 hours.
  • Step E To a solution of N-Boc-N-methyl-L-alanine (69.86 mg, 343.74 micromoles) in 2 ml of DMF was added N-methylmorpholine (69.54 mg, 687.48 micromoles) and HATU ( 130.70 mg, 343.74 micromolar).
  • Examples 44 to 47 were prepared according to the preparation procedure of Example 43.
  • Step A L-pyroglutamine (15 g, 130.29 mmol), benzaldehyde (15.90 g, 149.83 mmol, 15.14 ml) and p-toluenesulfonic acid monohydrate (1.24 g, 6.51 m) with stirring.
  • a solution of the toluene (250 ml) was refluxed for 5 hours. After cooling to room temperature, the reaction mixture was dried with EtOAc EtOAc m. The combined organic phases were washed with EtOAc EtOAc m.
  • Step B (3R,7aS)-3-phenyltetrahydropyrrole[1,2-c]oxazol-5(3H)-one (14 g, 68.88 mmol) in anhydrous at -78 °C A solution of tetrahydrofuran (28 ml) was added dropwise to a solution of potassium hexamethyldisilazide (1 M, 68.88 mL). After the addition was completed, the reaction solution was stirred at -78 ° C for 30 minutes.
  • Step C To (3R,7aS)-3-phenyl-6-phenylselenoyltetrahydropyrrole-[1,2-c]oxazol-5(3H)-one at 0 ° C (19 g, 53.03 A solution of ethyl acetate (190 ml) was added EtOAc (30%,EtOAc. The reaction solution was stirred at 0 ° C for 30 minutes. Then methanesulfonic acid (11.53 g, 120.00 mmol, 8.54 ml) was added thereto and stirred at 15 ° C for 12 hours. The reaction mixture was poured with water (100 ml The combined organic phases were washed with EtOAc EtOAc m.
  • Step D Sodium hydrogen (3.05 g, 76.33 mmol, 60% purity) was added to dimethyl sulfoxide (160 mL) at 15 °C. Then, trimethylsulfoxonium iodide (18.90 g, 85.88 mmol, 11.31 ml) was added thereto in portions, and stirred at this temperature for 30 minutes. The reaction solution was warmed to 55 ° C and stirred for 30 minutes. Further, (3R,7aS)-3-phenyl-3,7a-dihydro-1H-pyrrole[1,2-c]oxazol-5-one (6.40 g, 31.81 mmol) was added dropwise to the reaction mixture.
  • Step E (3R, 5aR, 6aS, 6bS)-3-phenyltetrahydro-1H-cyclopropane[3,4]pyrrole[1,2-c]oxazole-5(3H) at 0 degrees Celsius a solution of the ketone (5 g, 23.23 mmol) in anhydrous tetrahydrofuran (30 ml) was added dropwise to lithium tetrahydroaluminum (1.5 g, 39.49 m Molar) in anhydrous tetrahydrofuran (30 ml) in suspension. The reaction solution was warmed to reflux and stirred for 1 hour.
  • Step F Palladium on carbon (788.08 mg, 442.74 ⁇ mol, 5% purity) was added to ((1S,2S,5R)-3-benzyl-3-azabicyclo[3.1.0] at 15 °C.
  • the reaction solution was filtered and concentrated under reduced pressure.
  • the product (1S, 2S, 5R)-tert-butyl 2-(hydroxymethyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate (3.8 g, 17.82 mmol, 80.50%. ).
  • Step G A solution of DIAD (3.96 g, 19.60 mmol, 3.81 ml) in anhydrous tetrahydrofuran (5 mL) was added dropwise (1S,2S,5R)-tert-butyl 2-(hydroxyl) under nitrogen at 0 °C. Methyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate (3.80 g, 17.82 mmol), phthalimide (2.88 g, 19.60 mmol) and triphenyl A solution of phosphine (5.14 g, 19.60 mmol) in anhydrous tetrahydrofuran (40 mL). The reaction solution was warmed to 15 ° C and stirred for 11 hours.
  • Step H Add hydrazine hydrate (39.63 mmol, 2.27 mL, 85% purity) to (1S,2S,5R)-tert-butyl 2-((1,3-dicarbonyl) under nitrogen at 78 °C.
  • the reaction solution was stirred at 78 ° C for 2 hours.
  • the reaction solution was filtered and concentrated under reduced pressure.
  • the crude product (1S, 2S, 5R)-tert-butyl 2-(aminomethyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate (3.7 g) was obtained.
  • Step I 1,4-Difluoro-2-nitro-benzene (2.77 g, 17.43 mmol, 1.89 mL) was added to (1S,2S,5R)-tert-butyl 2- under nitrogen at 15 °C. (Aminomethyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate (3.7 g), potassium carbonate in acetonitrile (30 mL). The reaction solution was warmed to 82 ° C and stirred for 2 hours. The reaction solution was added with water (100 ml) and then extracted with ethyl acetate Take (200 ml x 1). The combined organic phases were washed with EtOAc EtOAc m.
  • Step J Palladium on carbon (826.59 mg, 779.80 ⁇ mol, 10% purity) was added to (1S, 2S, 5R)-tert-butyl 2-(((4-fluoro-2-nitro) at 25 ° C under nitrogen. Ethylphenyl)amine)methyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate (5.3 g, 15.05 mmol) in ethyl acetate (150 mL) and methanol (150 ML) mixed solution. Stir for 2 hours at 25 ° C under a hydrogen atmosphere (30 psi). The reaction solution was filtered and concentrated under reduced pressure.
  • Step K to (1S,2S,5R)-tert-butyl 2-(((2-amino-4-fluorophenyl)amine))methyl)-3-azabicyclo[3. 0] Hexane-3-carboxylate (1 g, 3.11 mmol) and (2R,4S)-benzyl 4-fluoro-2-((6-fluoro-2-formyl-1H-indole-3- To a mixed solution of methyl)pyrrole-1-carboxylate (2.48, 3.11 mmol) in DMF (12 mL) and water (600 ⁇ RTIgt; ⁇ /RTI> ⁇ RTIgt; ⁇ /RTI> liter), Oxone (1.42 g, 9.33 mmol) was added.
  • Step L to (1S,2S,5R)-tert-butyl 2-((2-(3-(((()))))) Fluoropyrrol-2-yl)methyl)-6-fluoro-1H-indol-2-yl)-5-fluoro-1H-benzo[d]imidazol-1-yl)methyl)-3-aza
  • a bicyclo[3.1.0]hexane-3-carboxylate (1 g, 1.17 mmol)
  • hydrobromic acid acetic acid 5 ml, concentration 35%) was added. Stir at 15 degrees Celsius for 1 hour.
  • Step M HATU (808.64 mg, 2.13 mmol) was added to N-Boc-L n-butyric acid (432.21 mg, 2.13 mmol), N-methylmorpholine (286.82 mg, 2.84 m) at 15 °C. Moore, 311.76 ⁇ l) in a stirred solution of DMF (5 mL).
  • reaction solution was directly purified by a reverse phase column (eluent: acetonitrile and aqueous solution of trifluoroacetic acid) to afford product t-butyl N-[(1S)-1-[(2R,4S)-2-[[2 -[1-[[(1S,2S,5R)-3-[(2S)-2-(tert-Butoxycarbonylamino)butanoyl]-3-azabicyclo[3.1.0]hexane-2- Methyl]-5-fluoro-benzoimidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl]-4-fluoro-pyrrole-1-carbonyl]propyl]amino Formate (390 mg, 452.54 micromoles, 63.84%).
  • Step N at 15 ° C, to tert-butyl N-[(1S)-1-[(2R,4S)-2-[[2-[1-[[(1S,2S,5R)-3-[ (2S)-2-(tert-Butoxycarbonylamino)butyryl]-3-azabicyclo[3.1.0]hexane-2-yl]methyl]-5-fluoro-benzimidazol-2-yl -6-Fluoro-1H-indol-3-yl]methyl]-4-fluoro-pyrrole-1-carbonyl]propyl]carbamate (390 mg, 452.54 ⁇ mol) in dichloromethane (2 To the solution in milliliters, trifluoroacetic acid (2 mL) was added.
  • Step O HATU (532.20 mg, 1.4 mmol) was added to N-Boc-N-methyl-L-alanine (432.21 mg, 1.4 mmol), N-methylmorpholine at 15 °C. 283.15 mg, 2.80 mmol) in a stirred solution of DMF (5 mL).
  • reaction solution was directly purified by a reverse phase column (eluent: acetonitrile and aqueous solution of trifluoroacetic acid) to give the product t-butyl N-[(1S)-2-[[(1S)-1-[(2R, 4S)-2-[[2-[1-[[(1S,2S,5R)-3-[(2S)-2-[[(2S)-2-[tert-butoxycarbonyl(methyl)amino]] Propionyl]amino]butyryl]-3-azabicyclo[3.1.0]hexane-2-yl]methyl]-5-fluoro-benzimidazol-2-yl]-6-fluoro-1H- Indole-3-yl]methyl]-4-fluoro-pyrrole-1-carbonyl]propyl]amino]-1-methyl-2-oxo-ethyl]-N-methyl-carbamate ( 350 mg, 347.86 micromoles, 7
  • Step P at 15 ° C, to tert-butyl N-[(1S)-2-[[(1S)-1-[(2R,4S)-2-[[2-[1-[[(1S, 2S,5R)-3-[(2S)-2-[[(2S)-2-[tert-butoxycarbonyl(methyl)amino]propanoyl]amino]butyryl]-3-azabicyclo[3.1 .0]Hex-2-yl]methyl]-5-fluoro-benzoimidazol-2-yl]-6-fluoro-1H-indol-3-yl]methyl]-4-fluoro-pyrrole- 1-carbonyl]propyl]amino]-1-methyl-2-oxo-ethyl]-N-methyl-carbamate (350 mg, 347.86 ⁇ mol) in dichloromethane (2 mL) Add trifluoroacetic acid (2 mL). Stir at 15 degrees Celsius for 12 hours
  • Example 49 was prepared according to the preparation procedure of Example 48.
  • Step A DMF (13.68) was added to a stirred solution of N-Cbz-cis-4-fluoro-L-proline (5.00 g, 18.71 mmol) in dry toluene (50 mL). Mg, 187.10 micromolar, 14.40 microliters). After the addition is complete, stir 15 minutes. Then, oxalyl chloride (3.56 g, 28.07 mmol, 2.46 ml) was added to the reaction solution at 15 ° C, and the reaction mixture was stirred at 15 ° C for 2 hours. The reaction solution was spun to remove excess oxalyl chloride, dissolved in toluene, and then used in the next step.
  • Step B Ethyl Grignard reagent (3M, 9.68 ml) was added dropwise to a mixed solvent of toluene (3.29 g, 28.10 mmol) in toluene (25 ml) and chlorobenzene (25 ml) at 0 ° C. ) and control added in 30 minutes. After the addition, the reaction solution was stirred at this temperature for 30 minutes, and then N-Cbz-cis-4-fluoro-L-prolyl chloride (5.35 g, 18.73 mmol) was added to the reaction mixture at 0 °C. The reaction mixture was stirred at 25 ° C for 2 hours.
  • Step C To a solution of (2S,4S)-benzyl 4-fluoro-2-(1H-indole-3-carbonyl)pyrrole-1-carboxylate (3.80 g, 7.68 mmol) in THF. A solution of 36 ml) was added to a solution of lithium borohydride (2M, 7.68 ml). The reaction solution was stirred at 15 ° C for 4 hours. Then methanesulfonic acid (1.36 g, 14.20 mmol, 1.01 ml) was added thereto and stirred at 15 ° C for 12 hours. The reaction mixture was quenched with EtOAc EtOAc (EtOAc) The combined organic layers were washed with EtOAcq.
  • EtOAc EtOAc EtOAc
  • Step D Phosphorus oxychloride (1.66 g, 10.83 mmol, 1.01 mL) was added to stirred DMF (672.00 mg, 9.19 mmol, 707.36 liters). The reaction solution is reacted at 0 ° C for 1 hour, and is dissolved in 1,2-dichloroethane (20 ml) (2R,4S)-4-fluoro-2-(1H-indol-3-ylmethyl)pyrrole 1-carboxylic acid ester (1.80 g, 4.60 mmol) was added dropwise to the reaction solution while maintaining the dropping temperature at 0 °C. The reaction solution was reacted at 15 ° C for 11 hours.
  • Step E To (2R,4S)-benzyl 4-fluoro-2-((2-formyl-1H-indol-3-yl)methyl)pyrrole-1-carboxylate (at 15 ° C) 1.50 g, 1.42 mmol) and (2S,4S)-tert-butyl 2-((2-amino-4-fluorophenyl)amine)methyl)-4-fluoropyrrole-1-carboxylate (464.71 mg To a mixed solution of DMF (15 ml) and water (1 ml), Oxone (648.03 mg, 4.26 mmol) was added. Stir at 15 degrees Celsius for 12 hours.
  • reaction solution was respectively saturated aqueous sodium hydrogencarbonate (20 ml) and saturated aqueous sodium sulfate (20 ml) was quenched and then extracted with ethyl acetate (20 mL). The combined organic phases were washed with EtOAcq.
  • Step F to (2R,4S)-benzyl 2-((2-(1-((2S,4S)-1-(tert-butoxycarbonyl)-4-fluoropyrrole-2-) at 15 °C Methyl)-5-fluoro-1H-benzo[d]imidazol-2-yl)-1H-indol-3-yl)methyl)-4-fluoropyrrole-1-carboxylate (450.00 mg Trifluoroacetic acid (3 ml) was added to dichloromethane (3 mL). Stir at 15 degrees Celsius for 12 hours. It was then concentrated under reduced pressure.
  • Step G HATU (808.64 mg, 2.13 mmol) was added to (S)-2-((tert-butoxycarbonyl)amine)-3-isovaleric acid (232.23 mg, 1.07 mmol) at 15 °C.
  • reaction solution was directly purified by a reverse phase column (eluent: acetonitrile and aqueous solution of trifluoroacetic acid) to give the product (2R,4S)-benzyl 2-((2-(1-(((2S))) 1-((S)-2-((tert-butoxycarbonyl)amine)-3-isovaleryl)-4-fluoropyrrol-2-yl)methyl)-5-fluoro-1H-benzo[d Imidazol-2-yl)-1H-indol-3-yl)methyl)-4-fluoropyrrole-1-carboxylate (250.00 mg, 292.29 micromoles, 41.02%).
  • Step H Palladium on carbon (26.12 mg, 319.71 ⁇ mol, 10% purity) was added to (2R,4S)-benzyl 2-((2-(1-(((2S,4S) )-1-((S)-2-((tert-butoxycarbonyl)amine)-3-isovaleryl)-4-fluoropyrrol-2-yl)methyl)-5-fluoro-1H-benzo[ d]imidazol-2-yl)-1H-indol-3-yl)methyl)-4-fluoropyrrole-1-carboxylate (250.00 mg, 292.29 ⁇ mol) in ethyl acetate (5 mL) and methanol (5 ml) in a mixed solution.
  • Step I HATU (142.42 mg, 374.57 micromoles) was added to N-Boc-L n-butyric acid (76.13 mg, 374.57 micromoles), nitromethylmorpholine (75.77 mg, 749.13 micromolar) at 15 degrees Celsius. , 82.36 ⁇ l) in a stirred solution of DMF (3 mL).
  • reaction solution was directly purified by a reverse phase column (eluent: acetonitrile and aqueous solution of trifluoroacetic acid) to give the product t-butyl N-[(1S)-1-[(2S,4S)-2-[[2 -[3-[[(2R,4S)-1-[(2S)-2-(tert-butoxycarbonylamino)butanoyl]-4-fluoropyrrol-2-yl]methyl]-1H-indole- 2-yl]-5-fluoro-benzoimidazol-1-yl]methyl]-4-fluoropyrrole-1-carbonyl]-2-methyl-propyl]carbamate (100.00 mg, 119.34 micromolar , 47.79%).
  • Step J at 15 ° C, to tert-butyl N-[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S) )-2-(tert-Butoxycarbonylamino)butyryl]-4-fluoropyrrol-2-yl]methyl]-1H-indol-2-yl]-5-fluoro-benzimidazol-1-yl] Methyl]-4-fluoropyrrole-1-carbonyl]-2-methyl-propyl]carbamate (100.00 mg, 119.34 ⁇ mol) in dichloromethane (2 mL). ML). Stir at 15 degrees Celsius for 0.5 hours.
  • Step K HATU (158.10 mg, 415.81 micromoles) was added to N-Boc-N-methyl-L-alanine (84.51 mg, 415.81 micromoles), N-methylmorpholine at 15 degrees C. 84.12 mg, 831.62 micromoles, 91.43 microliters) of a stirred solution of DMF (5 ml).
  • reaction solution was directly purified by a reverse phase column (eluent: acetonitrile and an aqueous solution of trifluoroacetic acid) to give the product t-butyl N-[(1S)-2-[[(1S)-1-[(2S, 4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-[[(2S)-2-[tert-butoxycarbonyl(methyl)amino]propanoyl) Amino]butyryl]-4-fluoro-pyrrol-2-yl]methyl]-1H-indol-2-yl]-5-fluoro-benzoimidazol-1-yl]methyl]-4-fluoro -pyrrole-1-carbonyl]-2-methyl-propanyl]amino]-1-methyl-2-oxo-ethyl]-N-methyl-carbamate (65.00 mg, 64.47 ⁇ mol, 46.51 %).
  • Step L at 15 ° C, to tert-butyl N-[(1S)-2-[[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R, 4S)-1-[(2S)-2-[[(2S)-2-[tert-Butoxycarbonyl(methyl)amino]propanoyl]amino]butyryl]-4-fluoro-pyrrol-2-yl] Methyl]-1H-indol-2-yl]-5-fluoro-benzoimidazol-1-yl]methyl]-4-fluoro-pyrrole-1-carbonyl]-2-methyl-propan]amino] To a solution of 1-methyl-2-oxo-ethyl]-N-methyl-carbamate (65.00 mg, 64.47 ⁇ mol) in dichloromethane (2 mL) ).
  • the compounds of the present invention have high cIAP1 inhibitory activity and moderate XIAP inhibitory activity, and have good selectivity.
  • test compound is added to the corresponding DMSO to form a 10 mM mother liquor;
  • the first plate is used to detect the activity of the compound; the second plate is used to detect the toxicity of the compound to the cells; the third plate is used for the first plate to change the liquid on the second day:
  • the inducer TNF- ⁇ was mixed with a medium containing 0.1% FBS and added to a third plate and mixed, and the mixture was transferred to the first plate from which the supernatant was discarded.
  • the NF- ⁇ B Luciferase Reporter Hela cell density was adjusted to 2.0 ⁇ 10 5 cells/ml with the medium.
  • the cells were added to two Greeper 96-well black cell culture plates containing the compound, 100 ul (2.0 ⁇ 10 4 cells/well) per well, one plate for detecting compound activity; the other plate for detecting compound cells. toxicity.
  • the plated cell plates were incubated for 24 hours in a 37 ° C, 5% CO 2 cell incubator.
  • TNF- ⁇ 100 ug/ml was diluted to 20 ng/ml with a 0.1% FBS culture solution, and 100 ul/well was added to the third compound plate.
  • Example EC50(nM) Example EC50(nM) Example EC50(nM) Example EC50(nM) 1 ⁇ 10 18 ⁇ 10 35 ⁇ 10 2 10-100 19 ⁇ 10 36 ⁇ 10 3 ⁇ 10 20 ⁇ 10 37 10-100 4 10-100 twenty one 10-100 38 ⁇ 10 5 10-100 twenty two ⁇ 10 39 10-100 6 10-100 twenty three ⁇ 10 40 ⁇ 10 7 10-100 twenty four 10-100 41 10-100
  • Representative compounds of the invention have TNF- ⁇ -induced NF- ⁇ B inhibitory activity comparable to or even higher than Birinapant.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

本发明公开了一类作为IAP拮抗剂的新型二价苯并咪唑联吲哚化合物,具体公开了式(I)所示化合物或其药学上可接受的盐。

Description

作为新型二价IAP拮抗剂的苯并咪唑联吲哚化合物 技术领域
本发明涉及一类作为IAP拮抗剂的新型二价苯并咪唑联吲哚化合物,具体公开了式(I)所示化合物或其药学上可接受的盐。
背景技术
细胞凋亡(apoptosis)指为维持内环境稳定,由基因控制的细胞自主的有序的死亡。它在生物体的进化、内环境的稳定以及多个系统的发育中起着重要的作用。细胞凋亡信号传导分为内在的(由死亡受体-配体相互作用所介导)和外在的(由细胞应激和线粒体通透所介导)。两种途径最终汇合于半胱氨酸天冬氨酸酶(Caspase)。一旦凋亡信号被激活,半胱氨酸天冬氨酸酶就会裂解大量与细胞死亡相关的底物,造成细胞死亡。
凋亡抑制因子凋亡抑制蛋白(inhibitor of apoptosis proteins,IAPs)是一类高度保守的内源性抗细胞凋亡因子家族,主要通过抑制Caspase活性和参与调解核因子NF-κB的作用而抑制细胞凋亡。Roy等于1995年首次从脊髓性肌萎缩症的研究中发现IAP蛋白是神经元性凋亡抑制蛋白(neuronal apoptosis inhibitor protein,NAIP),随后人们陆续发现了细胞凋亡抑制蛋白(cellular IAP、c-IAP1和c-IAP2)、X染色体连锁凋亡抑制因子(XIAP)、Survivin、黑色素瘤凋亡抑制蛋白(melanoma-IAP,ML-IAP/Livin)、睾丸特异凋亡抑制蛋白(hILP)、以及含泛素连接酶的杆状病毒IAP重复序列等,至今已发现8个人类IAPs家族蛋白成员。在8个IAP家族中,cIAP1,cIAP2,XIAP被研究的比较充分。它们都具有三个结构功能区域,分别称为BIR1、BIR2和BIR3,这些区域主要通过抑制Caspase 3、7、9等酶的活性而发挥阻断凋亡的作用。
Smac,全称为second mitochondria-derived activator of caspases,是存在于线粒体并调节细胞凋亡的蛋白质,其促凋亡作用是通过逆转凋亡抑制蛋白(IAPs)尤其是X连锁凋亡抑制蛋白(XIAP)的作用实现的。当细胞受到凋亡刺激时,线粒体释放Smac蛋白到细胞质中,后者与IAPs结合,使其丧失抑制半胱氨酸天冬氨酸蛋白酶(caspase)活性的作用,从而促进细胞凋亡。Smac蛋白通过N端四肽直接与多种IAP蛋白相结合,阻断细胞内IAP蛋白的细胞凋亡抑制作用,能够有效的促进细胞凋亡。大量的文献已经报道多种IAP抑制剂,也即Smac模拟物,在体内和体外模型中具有抑制癌细胞增殖,促进感染的细胞发生凋亡。其中Birinapant,LCL-161,AT-406等已进入临床I期或II期研究。但具有更好的活性,选择性和安全性的新的IAP拮抗剂仍然有很大需求。
Figure PCTCN2017082227-appb-000001
背景研发资料参考以下文献:
Nat.Rev.Drug Discov.2012,11,p109-124;Pharmacology&Therapeutics,2014,144,p82–95;J.Med.Chem.2014,57,p3666-3677;Proc.Natl.Sci.Acad.USA 2015,112,p5797–5802;CONDON,Stephen,M.等,WO/2006/091972.
发明内容
本发明提供式(I)所示化合物或其药学上可接受的盐,
Figure PCTCN2017082227-appb-000002
其中,
R1和R2分别独立地选自
Figure PCTCN2017082227-appb-000003
R3、R4和R5分别独立地选自任选被1、2或3个R取代的:C1-6烷基、C1-6杂烷基、C3-12环烷基、3~12元杂环烷基、5~12元芳基或杂芳基、5~12元芳烷基或杂芳烷基;
R6a和R6b分别独立地选自H、F、Cl、Br、I、OH、CN、NH2、COOH,或选自任选被1、2或3个R取代的:C1-6烷基、C1-6杂烷基、C3-6环烷基、3~6元杂环烷基、5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基;
任选地,R6a和R6b连接在一起,形成一个任选被1、2或3个R取代的3~6元环;
R7a和R7b分别独立地选自H、F、Cl、Br、I、OH、CN、NH2、COOH,或选自任选被1、2或3个R取代的:C1-6烷基、C1-6杂烷基、C3-6环烷基、3~6元杂环烷基、5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基;
任选地,R7a和R7b连接在一起,形成一个任选被1、2或3个R取代的3~6元环;
环A和环B分别独立地选自:5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基;
R8和R9分别独立地选自卤素、羟基或选自任选被1、2或3个R取代的:C1-6烷基、C1-6杂烷基、C3-6环烷基、3~6元杂环烷基;
m和n分别独立地选自:0、1、2或3;
R选自F、Cl、Br、I、CN、OH、NH2、COOH,或选自任选被1、2或3个R’取代的:C1-6烷基、C1-6杂烷基、C3-6环烷基、3~6元杂环烷基、苯基和5~6元杂芳基;
R’选自F、Cl、Br、I、OH、CN、NH2、COOH、Me、Et、CF3、CHF2、CH2F、NHCH3、N(CH3)2
“杂”表示杂原子或杂原子团,选自-C(=O)N(R)-、-N(R)-、-C(=NR)-、-S(=O)2N(R)-、-S(=O)N(R)-、-O-、-S-、=O、=S、-O-N=、-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)-、-S(=O)2-、-N(R)C(=O)N(R)-;
以上任何一种情况下,杂原子或杂原子团的数目分别独立地选自1、2或3。
在本发明的一些方案中,上述R选自F、Cl、Br、I、CN、OH、NH2、COOH,或选自任选被1、2或3个R’取代的:C1-3烷基、C1-3烷氧基、C1-3烷硫基、C1-3烷氨基和N,N-二(C1-2烷基)氨基。
在本发明的一些方案中,上述R选自F、Cl、Br、I、CN、OH、NH2、COOH、Me、Et、CF3、CHF2、CH2F、NHCH3、N(CH3)2
Figure PCTCN2017082227-appb-000004
在本发明的一些方案中,上述R3和R4分别独立地选自任选被1、2或3个R取代的:C1-4烷基、C1-4杂烷基、C3-6环烷基、3~6元杂环烷基、5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基。
在本发明的一些方案中,上述R3和R4分别独立地选自:C1-3烷基、C1-3杂烷基、苯基、吡啶基、嘧啶基、吡嗪基、哒嗪基、呋喃基、咪唑基、恶唑基、异恶唑基、噻吩基和吡唑基。
在本发明的一些方案中,上述R3和R4分别独立地选自Me。
在本发明的一些方案中,上述R5选自任选被1、2或3个R取代的:C1-4烷基、C1-4杂烷基、C3-6环烷基、3~6元杂环烷基、5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基。
在本发明的一些方案中,上述R5选自:C1-4烷基、C3-6环烷基、3~6元杂环烷基、苯基、吡啶基、嘧啶基、吡嗪基、哒嗪基、呋喃基、咪唑基、恶唑基、异恶唑基、噻吩基和吡唑基。
在本发明的一些方案中,上述R5选自
Figure PCTCN2017082227-appb-000005
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000006
选自
Figure PCTCN2017082227-appb-000007
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000008
选自:
Figure PCTCN2017082227-appb-000009
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000010
选自
Figure PCTCN2017082227-appb-000011
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000012
选自:
Figure PCTCN2017082227-appb-000013
在本发明的一些方案中,上述R6a和R6b连接在一起,形成一个任选被1、2或3个R取代的3~6 元环烷基。
在本发明的一些方案中,上述R6a和R6b连接在一起,结构单元
Figure PCTCN2017082227-appb-000014
选自
Figure PCTCN2017082227-appb-000015
在本发明的一些方案中,上述R6a和R6b连接在一起,结构单元
Figure PCTCN2017082227-appb-000016
选自
Figure PCTCN2017082227-appb-000017
在本发明的一些方案中,上述R7a和R7b连接在一起,形成一个任选被1、2或3个R取代的3~6元环烷基。
在本发明的一些方案中,上述R7a和R7b连接在一起,结构单元
Figure PCTCN2017082227-appb-000018
选自
Figure PCTCN2017082227-appb-000019
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000020
选自
Figure PCTCN2017082227-appb-000021
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000022
选自:
Figure PCTCN2017082227-appb-000023
Figure PCTCN2017082227-appb-000024
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000025
选自:
Figure PCTCN2017082227-appb-000026
Figure PCTCN2017082227-appb-000027
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000028
选自:
Figure PCTCN2017082227-appb-000029
Figure PCTCN2017082227-appb-000030
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000031
选自
Figure PCTCN2017082227-appb-000032
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000033
选自:
Figure PCTCN2017082227-appb-000034
Figure PCTCN2017082227-appb-000035
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000036
选自:
Figure PCTCN2017082227-appb-000037
Figure PCTCN2017082227-appb-000038
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000039
选自
Figure PCTCN2017082227-appb-000040
Figure PCTCN2017082227-appb-000041
本发明还提供述化合物或其药学上可接受的盐,其中,化合物选自
Figure PCTCN2017082227-appb-000042
R3、R4、R5、R6a、R7a、R8、R9如上述所定义。
本发明还提供化合物或其药学上可接受的盐,其选自:
Figure PCTCN2017082227-appb-000043
Figure PCTCN2017082227-appb-000044
Figure PCTCN2017082227-appb-000045
Figure PCTCN2017082227-appb-000046
Figure PCTCN2017082227-appb-000047
Figure PCTCN2017082227-appb-000048
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其选自:
Figure PCTCN2017082227-appb-000049
Figure PCTCN2017082227-appb-000050
Figure PCTCN2017082227-appb-000051
Figure PCTCN2017082227-appb-000052
Figure PCTCN2017082227-appb-000053
Figure PCTCN2017082227-appb-000054
本发明还提供一种药物组合物,其含有治疗有效量的上述的化合物或其药学上可接受的盐和药学上可接受的载体。
本发明还提供上述化合物或其药学上可接受的盐或上述药物组合物在制备治疗因IAP紊乱所致疾病的药物中的应用。
在本发明的一些方案中,上述因IAP紊乱所致疾病选自肿瘤或乙型肝炎病毒感染。
在本发明的一些方案中,上述R选自F、Cl、Br、I、CN、OH、NH2、COOH,或选自任选被1、2或3个R’取代的:C1-3烷基、C1-3烷氧基、C1-3烷硫基、C1-3烷氨基和N,N-二(C1-2烷基)氨基,其他变量如上述所定义。
在本发明的一些方案中,上述R选自F、Cl、Br、I、CN、OH、NH2、COOH、Me、Et、CF3、CHF2、 CH2F、NHCH3、N(CH3)2
Figure PCTCN2017082227-appb-000055
其他变量如上述所定义。
在本发明的一些方案中,上述R3和R4分别独立地选自任选被1、2或3个R取代的:C1-4烷基、C1-4杂烷基、C3-6环烷基、3~6元杂环烷基、5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基,其他变量如上述所定义。
在本发明的一些方案中,上述R3和R4分别独立地选自:C1-3烷基、C1-3杂烷基、苯基、吡啶基、嘧啶基、吡嗪基、哒嗪基、呋喃基、咪唑基、恶唑基、异恶唑基、噻吩基和吡唑基,其他变量如上述所定义。
在本发明的一些方案中,上述R3和R4分别独立地选自Me,其他变量如上述所定义。
在本发明的一些方案中,上述R5选自任选被1、2或3个R取代的:C1-4烷基、C1-4杂烷基、C3-6环烷基、3~6元杂环烷基、5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基,其他变量如上述所定义。
在本发明的一些方案中,上述R5选自:C1-4烷基、C3-6环烷基、3~6元杂环烷基、苯基、吡啶基、嘧啶基、吡嗪基、哒嗪基、呋喃基、咪唑基、恶唑基、异恶唑基、噻吩基和吡唑基,其他变量如上述所定义。
在本发明的一些方案中,上述R5选自
Figure PCTCN2017082227-appb-000056
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000057
选自
Figure PCTCN2017082227-appb-000058
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000059
选自:
Figure PCTCN2017082227-appb-000060
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000061
选自
Figure PCTCN2017082227-appb-000062
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000063
选自:
Figure PCTCN2017082227-appb-000064
其他变量如上述所定义。
在本发明的一些方案中,上述R6a和R6b连接在一起,形成一个任选被1、2或3个R取代的3~6元环烷基,其他变量如上述所定义。
在本发明的一些方案中,上述R6a和R6b连接在一起,结构单元
Figure PCTCN2017082227-appb-000065
选自
Figure PCTCN2017082227-appb-000066
其他变量如上述所定义。
在本发明的一些方案中,上述R6a和R6b连接在一起,结构单元
Figure PCTCN2017082227-appb-000067
选自
Figure PCTCN2017082227-appb-000068
其他变量如上述所定义。
在本发明的一些方案中,上述R7a和R7b连接在一起,形成一个任选被1、2或3个R取代的3~6元环烷基,其他变量如上述所定义。
在本发明的一些方案中,上述R7a和R7b连接在一起,结构单元
Figure PCTCN2017082227-appb-000069
选自
Figure PCTCN2017082227-appb-000070
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000071
选自
Figure PCTCN2017082227-appb-000072
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000073
选自:
Figure PCTCN2017082227-appb-000074
Figure PCTCN2017082227-appb-000075
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000076
选自:
Figure PCTCN2017082227-appb-000077
Figure PCTCN2017082227-appb-000078
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000079
选自:
Figure PCTCN2017082227-appb-000080
Figure PCTCN2017082227-appb-000081
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000082
选自
Figure PCTCN2017082227-appb-000083
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000084
选自:
Figure PCTCN2017082227-appb-000085
Figure PCTCN2017082227-appb-000086
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000087
选自:
Figure PCTCN2017082227-appb-000088
Figure PCTCN2017082227-appb-000089
其他变量如上述所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2017082227-appb-000090
选自
Figure PCTCN2017082227-appb-000091
Figure PCTCN2017082227-appb-000092
其他变量如上述所定义。
本发明还有一些方案是由上述变量任意组合而来。
技术效果
本发明化合物将母核结构设计为苯并咪唑联吲哚,提高了分子整体的溶解性。苯并咪唑联吲哚母核的创新之处还在于,其母核上的氢键供体少,具有更低的XIAP结合力,从而产生更高的cIAP/XIAP的选择性,更高选择性的化合物在动物和人体内将具有更低的毒性以及更好的耐受性。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。
C1-12选自C1、C2、C3、C4、C5、C6、C7、C8、C9、C10、C11和C12;C3-12选自C3、C4、C5、C6、C7、C8、C9、C10、C11和C12
C1-12烷基或杂烷基、C3-12环烃基或杂环烃基、被C3-12环烃基或杂环烃基取代的C1-12烷基或杂烷基包括但不限于:
C1-12烷基、C1-12烷氨基、N,N-二(C1-12烷基)氨基、C1-12烷氧基、C1-12烷酰基、C1-12烷氧羰基、C1-12烷基磺酰基、C1-12烷基亚磺酰基、C3-12环烷基、C3-12环烷氨基、C3-12杂环烷氨基、C3-12环烷氧基、C3-12环烷基酰基、C3-12环烷基氧羰基、C3-12环烷基磺酰基、C3-12环烷基亚磺酰基、5~12元芳基或杂芳基、5~12元芳烷基或杂芳烷基;
甲基、乙基、正丙基、异丙基、-CH2C(CH3)(CH3)(OH)、环丙基、环丁基、丙基亚甲基、环丙酰基、苄氧基、三氟甲基、氨甲基、羟甲基、甲氧基、甲酰基、甲氧羰基、甲磺酰基、甲基亚磺酰基、乙氧基、乙酰基、乙磺酰基、乙氧羰基、二甲基氨基、二乙基氨基、二甲基氨基羰基、二乙基氨基羰基;
N(CH3)2,NH(CH3),-CH2CF3,-CH2CH2CF3,-CH2CH2F,-CH2CH2S(=O)2CH3,-CH2CH2CN,-CH2CH(OH)(CH3)2,-CH2CH(F)(CH3)2,-CH2CH2F,-CH2CF3,-CH2CH2CF3,-CH2CH2NH2,-CH2CH2OH,-CH2CH2OCH3,-CH2CH2CH2OCH3,-CH2CH2N(CH3)2,-S(=O)2CH3,-CH2CH2S(=O)2CH3;和
苯基、噻唑基、联苯基、萘基、环戊基、呋喃基、3-吡咯啉基、吡咯烷基、1,3-氧五环基、吡唑基、2-吡唑啉基、吡唑烷基、咪唑基、恶唑基、噻唑基、1,2,3-唑基、1,2,3-三唑基、1,2,4-三唑基、1,3,4-噻二唑基、4H-吡喃基、吡啶基、哌啶基、1,4-二氧六环基、吗啉基、哒嗪基、嘧啶基、吡嗪基、哌嗪基、1,3,5-三噻烷基、1,3,5-三嗪基、苯并呋喃基、苯并噻吩基、吲哚基、苯并咪唑基、苯并噻唑基、嘌呤基、喹啉基、异喹啉基、噌啉基或喹喔啉基;
这里所采用的术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物的中性形式接触的方式获得碱加成盐。药学上可接受的碱加成盐包括钠、钾、钙、铵、有机氨或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物的中性形式接触的方式获得酸加成盐。药学上可接受的酸加成盐的实例包括无机酸盐,所述无机酸包括例如盐酸、氢溴酸、硝酸、碳酸,碳酸氢根,磷酸、磷酸一氢根、磷酸二氢根、硫酸、硫酸氢根、氢碘酸、亚磷酸等;以及有机酸盐,所述有机酸包括如乙酸、丙酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、酒石酸和甲磺酸等类似的酸;还包括氨基酸(如精氨酸等)的盐,以及如葡糖醛酸等有机酸的盐(参见Berge et al.,"Pharmaceutical Salts",Journal of Pharmaceutical Science 66:1-19(1977))。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
优选地,以常规方式使盐与碱或酸接触,再分离母体化合物,由此再生化合物的中性形式。化合物的母体形式与其各种盐的形式的不同之处在于某些物理性质,例如在极性溶剂中的溶解度不同。
本文所用的“药学上可接受的盐”属于本发明化合物的衍生物,其中,通过与酸成盐或与碱成盐的方式修饰所述母体化合物。药学上可接受的盐的实例包括但不限于:碱基比如胺的无机酸或有机酸盐、酸根比如羧酸的碱金属或有机盐等等。药学上可接受的盐包括常规的无毒性的盐或母体化合物的季铵盐,例如无毒的无机酸或有机酸所形成的盐。常规的无毒性的盐包括但不限于那些衍生自无机酸和有机酸的盐,所述的无机酸或有机酸选自2-乙酰氧基苯甲酸、2-羟基乙磺酸、乙酸、抗坏血酸、苯磺酸、苯甲酸、碳酸氢根、碳酸、柠檬酸、依地酸、乙烷二磺酸、乙烷磺酸、富马酸、葡庚糖、葡糖酸、谷氨酸、乙醇酸、氢溴酸、盐酸、氢碘酸盐、羟基、羟萘、羟乙磺酸、乳酸、乳糖、十二烷基磺酸、马来酸、苹果酸、扁桃酸、甲烷磺酸、硝酸、草酸、双羟萘酸、泛酸、苯乙酸、磷酸、多聚半乳糖醛、丙酸、水杨酸、硬脂酸、亚乙酸、琥珀酸、氨基磺酸、对氨基苯磺酸、硫酸、单宁、酒石酸和对甲苯磺酸。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。一般地,优选醚、乙酸乙酯、乙醇、异丙醇或乙腈等非水介质。
除了盐的形式,本发明所提供的化合物还存在前药形式。本文所描述的化合物的前药容易地在生理条件下发生化学变化从而转化成本发明的化合物。此外,前体药物可以在体内环境中通过化学或生化方法被转换到本发明的化合物。
本发明的某些化合物可以以非溶剂化形式或者溶剂化形式存在,包括水合物形式。一般而言,溶剂化形式与非溶剂化的形式相当,都包含在本发明的范围之内。
本发明的某些化合物可以具有不对称碳原子(光学中心)或双键。外消旋体、非对映异构体、几何 异构体和单个的异构体都包括在本发明的范围之内。
本文中消旋体、ambiscalemic and scalemic或者对映体纯的化合物的图示法来自Maehr,J.Chem.Ed.1985,62:114-120。1985年,62:114-120。除非另有说明,用楔形键和虚线键表示一个立体中心的绝对构型。当本文所述化合物含有烯属双键或其它几何不对称中心,除非另有规定,它们包括E、Z几何异构体。同样地,所有的互变异构形式均包括在本发明的范围之内。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和反式异构体、(-)-和(+)-对对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。
本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚(3H),碘-125(125I)或C-14(14C)。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。
术语“药学上可接受的载体”是指能够递送本发明有效量活性物质、不干扰活性物质的生物活性并且对宿主或者患者无毒副作用的任何制剂或载体介质代表性的载体包括水、油、蔬菜和矿物质、膏基、洗剂基质、软膏基质等。这些基质包括悬浮剂、增粘剂、透皮促进剂等。它们的制剂为化妆品领域或局部药物领域的技术人员所周知。关于载体的其他信息,可以参考Remington:The Science and Practice of Pharmacy,21st Ed.,Lippincott,Williams&Wilkins(2005),该文献的内容通过引用的方式并入本文。
术语“赋形剂”通常是指配制有效的药物组合物所需要载体、稀释剂和/或介质。
针对药物或药理学活性剂而言,术语“有效量”或“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。对于本发明中的口服剂型,组合物中一种活性物质的“有效量”是指与该组合物中另一种活性物质联用时为了达到预期效果所需要的用量。有效量的确定因人而异,取决于受体的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
术语“活性成分”、“治疗剂”,“活性物质”或“活性剂”是指一种化学实体,它可以有效地治疗目标紊乱、疾病或病症。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧代(即=O)时,意味着两个氢原子被取代。氧取代不会发生在芳香基上。术语“任选被取代的”是指可以被取代,也可以不被取代, 除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当一个连接基团的数量为0时,比如-(CRR)0-,表示该连接基团为单键。
当其中一个变量选自单键时,表示其连接的两个基团直接相连,比如A-L-Z中L代表单键时表示该结构实际上是A-Z。
当一个取代基为空缺时,表示该取代基是不存在的,比如A-X中X为空缺时表示该结构实际上是A。
当一个取代基的键可以交叉连接到一个环上的两个原子时,这种取代基可以与这个环上的任意原子相键合。当所列举的取代基中没有指明其通过哪一个原子连接到化学结构通式中包括但未具体提及的化合物时,这种取代基可以通过其任何原子相键合。取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。例如,结构单元
Figure PCTCN2017082227-appb-000093
表示其可在环己基或者环己二烯上的任意一个位置发生取代。
除非另有规定,术语“卤代素”或“卤素”本身或作为另一取代基的一部分表示氟、氯、溴或碘原子。此外,术语“卤代烷基”意在包括单卤代烷基和多卤代烷基。例如,术语“卤代(C1-C4)烷基”意在包括但不仅限于三氟甲基、2,2,2-三氟乙基、4-氯丁基和3-溴丙基等等。
卤代烷基的实例包括但不仅限于:三氟甲基、三氯甲基、五氟乙基,和五氯乙基。“烷氧基”代表通过氧桥连接的具有特定数目碳原子的上述烷基。C1-6烷氧基包括C1、C2、C3、C4、C5和C6的烷氧基。烷氧基的例子包括但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、仲丁氧基、叔丁氧基、正戊氧基和S-戊氧基。“环烷基”包括饱和环基,如环丙基、环丁基或环戊基。3-7环烷基包括C3、C4、C5、C6和C7环烷基。“链烯基”包括直链或支链构型的烃链,其中该链上任何的稳定位点上存在一个或多个碳-碳双键,例如乙烯基和丙烯基。
术语“卤”或“卤素”是指氟、氯、溴和碘。
除非另有规定,术语“杂”表示杂原子或杂原子团(即含有杂原子的原子团),包括碳(C)和氢(H)以外的原子以及含有这些杂原子的原子团,例如包括氧(O)、氮(N)、硫(S)、硅(Si)、锗(Ge)、铝(Al)、硼(B)、-O-、-S-、=O、=S、-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)、-S(=O)2-,以及任选被取代的-C(=O)N(H)-、-N(H)-、-C(=NH)-、-S(=O)2N(H)-或-S(=O)N(H)-。
除非另有规定,“环”表示被取代或未被取代的环烷基、杂环烷基、环烯基、杂环烯基、环炔基、杂环炔基、芳基或杂芳基。所谓的环包括单环、联环、螺环、并环或桥环。环上原子的数目通常被定义为环的元数,例如,“5~7元环”是指环绕排列5~7个原子。除非另有规定,该环任选地包含1~3个杂原子。因此,“5~7元环”包括例如苯基、吡啶和哌啶基;另一方面,术语“5~7元杂环烷基环”包括吡啶基和哌啶基,但不包括苯基。术语“环”还包括含有至少一个环的环系,其中的每一个“环”均独立地符 合上述定义。
除非另有规定,术语“杂环”或“杂环基”意指稳定的含杂原子或杂原子团的单环、双环或三环,它们可以是饱和的、部分不饱和的或不饱和的(芳族的),它们包含碳原子和1、2、3或4个独立地选自N、O和S的环杂原子,其中上述任意杂环可以稠合到一个苯环上形成双环。氮和硫杂原子可任选被氧化(即NO和S(O)p,p是1或2)。氮原子可以是被取代的或未取代的(即N或NR,其中R是H或本文已经定义过的其他取代基)。该杂环可以附着到任何杂原子或碳原子的侧基上从而形成稳定的结构。如果产生的化合物是稳定的,本文所述的杂环可以发生碳位或氮位上的取代。杂环中的氮原子任选地被季铵化。一个优选方案是,当杂环中S及O原子的总数超过1时,这些杂原子彼此不相邻。另一个优选方案是,杂环中S及O原子的总数不超过1。如本文所用,术语“芳族杂环基团”或“杂芳基”意指稳定的5、6、7元单环或双环或7、8、9或10元双环杂环基的芳香环,它包含碳原子和1、2、3或4个独立地选自N、O和S的环杂原子。氮原子可以是被取代的或未取代的(即N或NR,其中R是H或本文已经定义过的其他取代基)。氮和硫杂原子可任选被氧化(即NO和S(O)p,p是1或2)。值得注意的是,芳香杂环上S和O原子的总数不超过1。桥环也包含在杂环的定义中。当一个或多个原子(即C、O、N或S)连接两个不相邻的碳原子或氮原子时形成桥环。优选的桥环包括但不限于:一个碳原子、两个碳原子、一个氮原子、两个氮原子和一个碳-氮基。值得注意的是,一个桥总是将单环转换成三环。桥环中,环上的取代基也可以出现在桥上。
杂环化合物的实例包括但不限于:吖啶基、吖辛因基、苯并咪唑基、苯并呋喃基、苯并巯基呋喃基、苯并巯基苯基、苯并恶唑基、苯并恶唑啉基、苯并噻唑基、苯并三唑基、苯并四唑基、苯并异恶唑基、苯并异噻唑基、苯并咪唑啉基、咔唑基、4aH-咔唑基、咔啉基、苯并二氢吡喃基、色烯、噌啉基十氢喹啉基、2H,6H-1,5,2-二噻嗪基、二氢呋喃并[2,3-b]四氢呋喃基、呋喃基、呋咱基、咪唑烷基、咪唑啉基、咪唑基、1H-吲唑基、吲哚烯基、二氢吲哚基、中氮茚基、吲哚基、3H-吲哚基、异苯并呋喃基、异吲哚基、异二氢吲哚基、异喹啉基、异噻唑基、异恶唑基、亚甲二氧基苯基、吗啉基、萘啶基,八氢异喹啉基、恶二唑基、1,2,3-恶二唑基、1,2,4-恶二唑基、1,2,5-恶二唑基、1,3,4-恶二唑基、恶唑烷基、恶唑基、羟吲哚基、嘧啶基、菲啶基、菲咯啉基、吩嗪、吩噻嗪、苯并黄嘌呤基、酚恶嗪基、酞嗪基、哌嗪基、哌啶基、哌啶酮基、4-哌啶酮基、胡椒基、蝶啶基、嘌呤基、吡喃基、吡嗪基、吡唑烷基、吡唑啉基、吡唑基、哒嗪基、吡啶并恶唑、吡啶并咪唑、吡啶并噻唑、吡啶基、吡咯烷基、吡咯啉基、2H-吡咯基、吡咯基、喹唑啉基、喹啉基、4H-喹嗪基、喹喔啉基、奎宁环基、四氢呋喃基、四氢异喹啉基、四氢喹啉基、四唑基,6H-1,2,5-噻二嗪基、1,2,3-噻二唑基、1,2,4-噻二唑基、1,2,5-噻二唑基、1,3,4-噻二唑基、噻蒽基、噻唑基、异噻唑基噻吩基、噻吩并恶唑基、噻吩并噻唑基、噻吩并咪唑基、噻吩基、三嗪基、1,2,3-三唑基、1,2,4-三唑基、1,2,5-三唑基、1,3,4-三唑基和呫吨基。还包括稠环和螺环化合物。
除非另有规定,术语“烃基”或者其下位概念(比如烷基、烯基、炔基、苯基等等)本身或者作为另一取代基的一部分表示直链的、支链的或环状的烃原子团或其组合,可以是完全饱和的、单元或多元不饱和的,可以是单取代、二取代或多取代的,可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基),可以包括二价或多价原子团,具有指定数量的碳原子(如C1-C10表示1至10个碳)。“烃基”包括但不限于脂肪烃基和芳香烃基,所述脂肪烃基包括链状和环状,具体包括但不限于烷基、烯基、炔 基,所述芳香烃基包括但不限于6-12元的芳香烃基,例如苯、萘等。在一些实施例中,术语“烃基”表示直链的或支链的原子团或它们的组合,可以是完全饱和的、单元或多元不饱和的,可以包括二价和多价原子团。饱和烃原子团的实例包括但不限于甲基、乙基、正丙基、异丙基、正丁基、叔丁基、异丁基、仲丁基、异丁基、环己基、(环己基)甲基、环丙基甲基,以及正戊基、正己基、正庚基、正辛基等原子团的同系物或异构体。不饱和烷基具有一个或多个双键或三键,其实例包括但不限于乙烯基、2-丙烯基、丁烯基、巴豆基、2-异戊烯基、2-(丁二烯基)、2,4-戊二烯基、3-(1,4-戊二烯基)、乙炔基、1-和3-丙炔基,3-丁炔基,以及更高级的同系物和异构体。
除非另有规定,术语“杂烃基”或者其下位概念(比如杂烷基、杂烯基、杂炔基、杂芳基等等)本身或者与另一术语联合表示稳定的直链的、支链的或环状的烃原子团或其组合,有一定数目的碳原子和至少一个杂原子组成。在一些实施例中,术语“杂烷基”本身或者与另一术语联合表示稳定的直链的、支链的烃原子团或其组合物,有一定数目的碳原子和至少一个杂原子组成。在一个典型实施例中,杂原子选自B、O、N和S,其中氮和硫原子任选地被氧化,氮杂原子任选地被季铵化。杂原子或杂原子团可以位于杂烃基的任何内部位置(包括该烃基附着于分子其余部分的位置)。实例包括但不限于-CH2-CH2-O-CH3、-CH2-CH2-NH-CH3、-CH2-CH2-N(CH3)-CH3、-CH2-S-CH2-CH3、-CH2-CH2、-S(O)-CH3、-CH2-CH2-S(O)2-CH3、-CH=CH-O-CH3、-CH2-CH=N-OCH3和–CH=CH-N(CH3)-CH3。至多两个杂原子可以是连续的,例如-CH2-NH-OCH3
术语“烷氧基”、“烷氨基”和“烷硫基”(或硫代烷氧基)属于惯用表达,是指分别通过一个氧原子、氨基或硫原子连接到分子的其余部分的那些烷基基团。
除非另有规定,术语“环烃基”、“杂环烃基”或者其下位概念(比如芳基、杂芳基、环烷基、杂环烷基、环烯基、杂环烯基、环炔基、杂环炔基等等)本身或与其他术语联合分别表示环化的“烃基”、“杂烃基”。此外,就杂烃基或杂环烃基(比如杂烷基、杂环烷基)而言,杂原子可以占据该杂环附着于分子其余部分的位置。环烃基的实例包括但不限于环戊基、环己基、1-环己烯基、3-环己烯基、环庚基等。杂环基的非限制性实例包括1-(1,2,5,6-四氢吡啶基)、1-哌啶基、2-哌啶基,3-哌啶基、4-吗啉基、3-吗啉基、四氢呋喃-2-基、四氢呋喃吲哚-3-基、四氢噻吩-2-基、四氢噻吩-3-基,1-哌嗪基和2-哌嗪基。
除非另有规定,术语“芳基”表示多不饱和的芳族烃取代基,可以是单取代、二取代或多取代的,可以是一价、二价或者多价,它可以是单环或多环(比如1至3个环;其中至少一个环是芳族的),它们稠合在一起或共价连接。术语“杂芳基”是指含有一至四个杂原子的芳基(或环)。在一个示范性实例中,杂原子选自B、N、O和S,其中氮和硫原子任选地被氧化,氮原子任选地被季铵化。杂芳基可通过杂原子连接到分子的其余部分。芳基或杂芳基的非限制性实施例包括苯基、1-萘基、2-萘基、4-联苯基、1-吡咯基、2-吡咯基、3-吡咯基、3-吡唑基、2-咪唑基、4-咪唑基、吡嗪基、2-恶唑基、4-恶唑基、2-苯基-4-恶唑基、5-恶唑基、3-异恶唑基、4-异恶唑基、5-异恶唑基、2-噻唑基、4-噻唑基、5-噻唑基、2-呋喃基、3-呋喃基、2-噻吩基、3-噻吩基、2-吡啶基、3-吡啶基、4-吡啶基、2-嘧啶基、4-嘧啶基、5-苯并噻唑基、嘌呤基、2-苯并咪唑基、5-吲哚基、1-异喹啉基、5-异喹啉基、2-喹喔啉基、5-喹喔啉基、3-喹啉基和6-喹啉基。上述任意一个芳基和杂芳基环系的取代基选自下文所述的可接受的取代基。
除非另有规定,芳基在与其他术语联合使用时(例如芳氧基、芳硫基、芳烷基)包括如上定义的芳 基和杂芳基环。因此,术语“芳烷基”意在包括芳基附着于烷基的那些原子团(例如苄基、苯乙基、吡啶基甲基等),包括其中碳原子(如亚甲基)已经被例如氧原子代替的那些烷基,例如苯氧基甲基、2-吡啶氧甲基3-(1-萘氧基)丙基等。
术语“离去基团”是指可以被另一种官能团或原子通过取代反应(例如亲和取代反应)所取代的官能团或原子。例如,代表性的离去基团包括三氟甲磺酸酯;氯、溴、碘;磺酸酯基,如甲磺酸酯、甲苯磺酸酯、对溴苯磺酸酯、对甲苯磺酸酯等;酰氧基,如乙酰氧基、三氟乙酰氧基等等。
术语“保护基”包括但不限于“氨基保护基”、“羟基保护基”或“巯基保护基”。术语“氨基保护基”是指适合用于阻止氨基氮位上副反应的保护基团。代表性的氨基保护基包括但不限于:甲酰基;酰基,例如链烷酰基(如乙酰基、三氯乙酰基或三氟乙酰基);烷氧基羰基,如叔丁氧基羰基(Boc);芳基甲氧羰基,如苄氧羰基(Cbz)和9-芴甲氧羰基(Fmoc);芳基甲基,如苄基(Bn)、三苯甲基(Tr)、1,1-二-(4'-甲氧基苯基)甲基;甲硅烷基,如三甲基甲硅烷基(TMS)和叔丁基二甲基甲硅烷基(TBS)等等。术语“羟基保护基”是指适合用于阻止羟基副反应的保护基。代表性羟基保护基包括但不限于:烷基,如甲基、乙基和叔丁基;酰基,例如链烷酰基(如乙酰基);芳基甲基,如苄基(Bn),对甲氧基苄基(PMB)、9-芴基甲基(Fm)和二苯基甲基(二苯甲基,DPM);甲硅烷基,如三甲基甲硅烷基(TMS)和叔丁基二甲基甲硅烷基(TBS)等等。
本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明采用下述缩略词:
缩略词清单
Pd/C Pd/C催化剂
HATU 2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯
TEMPO 四甲基哌啶氮氧化物
DIAD 偶氮二甲酸二异丙酯
NMM N-甲基吗啉
DCM 二氯甲烷
THF 四氢呋喃
Boc 叔丁氧羰基
Oxone 过硫酸氢钾复合盐
Cbz 苄氧羰基
DMF N,N-二甲基甲酰胺
LiBH4 硼氢化锂
TFA 三氟乙酸
EDCI 1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐
aq
EDC N-(3-二甲基氨基丙基)-N'-乙基碳二亚胺盐酸盐
m-CPBA 3-氯过氧苯甲酸
eq 当量、等量
CDI 羰基二咪唑
DCM 二氯甲烷
PE 石油醚
DMSO 二甲亚砜
EtOAc 乙酸乙酯
EtOH 乙醇
MeOH 甲醇
CBz 苄氧羰基,是一种胺保护基团
HOAc 乙酸
NaCNBH3 氰基硼氢化钠
r.t. 室温
O/N 过夜
Boc2O 二-叔丁基二碳酸酯
TFA 三氟乙酸
DIPEA 二异丙基乙基胺
SOCl2 氯化亚砜
CS2 二硫化碳
TsOH 对甲苯磺酸
NFSI N-氟-N-(苯磺酰基)苯磺酰胺
NCS 1-氯吡咯烷-2,5-二酮
n-Bu4NF 氟化四丁基铵
iPrOH 2-丙醇
mp 熔点
LDA 二异丙基胺基锂
化合物经手工或者
Figure PCTCN2017082227-appb-000094
软件命名,市售化合物采用供应商目录名称。
本发明所使用的溶剂可经市售获得且不需要进一步纯化。反应一般是在惰性氮气下、无水溶剂中进行的。质子核磁共振数据记录在Bruker Avance III 400(400MHz)分光仪上,化学位移以四甲基硅烷高场处的(ppm)表示。质谱是在安捷伦1200系列加6110(&1956A)上测定。LC/MS或Shimadzu MS包含一个DAD:SPD-M20A(LC)和Shimadzu Micromass 2020检测器。质谱仪配备有一个正或负模式下操作的电喷雾离子源(ESI)。
用配有Shimadzu SIL-20A自动进样器和日本岛津DAD:SPD-M20A探测器的岛津LC20AB系统进行高效液相色谱分析,采用Xtimate C18(3m填料,规格为2.1x 300mm)色谱柱。0-60AB_6分钟的方法:应用线性梯度,以100%A(A为0.0675%TFA的水溶液)开始洗脱,并以60%B(B为0.0625%TFA的MeCN溶液)结束洗脱,整个过程为4.2分钟,然后以60%B洗脱1分钟。将色谱柱再平衡0.8分钟达到100:0,总运行时间为6分钟。10-80AB_6分钟的方法:应用线性梯度,以90%A(A为0.0675%TFA的水溶液)开始洗脱,并以80%B(B为0.0625%TFA的乙腈溶液)结束洗脱,整个过程为4.2分钟,然后以80%B洗脱1分钟。将色谱柱再平衡0.8分钟达到90:10,总运行时间为6分钟。柱温为50℃,流速为0.8mL/min。二极管阵列检测器扫描波长为200-400nm。
在Sanpont-group的硅胶GF254上进行薄层色谱分析(TLC),常用紫外光灯照射检出斑点,在某些情况下也采用其他方法检视斑点,在这些情况下,用碘(10g硅胶中加入约1g碘并彻底混合而成)、香草醛(溶解大约1g香草醛于100mL 10%H2SO4中制得)、茚三酮(从Aldrich购得)或特殊显色剂(彻底混合25g(NH4)6Mo7O24·4H2O、5g(NH4)2Ce(IV)(NO3)6、450mL H2O和50mL浓H2SO4而制得)展开薄层板,检视化合物。采用Still,W.C.;Kahn,M.;and Mitra,M.Journal of Organic Chemistry, 1978,43,2923-2925.中所公开技术的类似方法,在Silicycle的40-63μm(230-400目)硅胶上进行快速柱色谱。快速柱色谱或薄层色谱的常用溶剂是二氯甲烷/甲醇、乙酸乙酯/甲醇和己烷/乙酸乙酯的混合物。
在使用Gilson UV/VIS-156检测器的Gilson-281Prep LC 322系统上进行制备色谱分析,所采用的色谱柱为Agella Venusil ASB Prep C18(5m填料,规格为150x 21.2mm)、Phenomenex Gemini C18(5m填料,规格为150x 30mm)、Boston Symmetrix C18(5m填料,规格为150x 30mm)或Phenomenex Synergi C18(4m填料,规格为150x 30mm)。在流速约为25mL/min时,用低梯度的乙腈/水(水中含有0.05%HCl、0.25%HCOOH或0.5%NH3·H2O)洗脱化合物,总运行时间为8-15分钟。
具体实施方式
下面通过实施例对本发明进行详细描述,但并不意味着对本发明任何不利限制。本文已经详细地描述了本发明,其中也公开了其具体实施例方式,对本领域的技术人员而言,在不脱离本发明精神和范围的情况下针对本发明具体实施方式进行各种变化和改进将是显而易见的。
实施例1
Figure PCTCN2017082227-appb-000095
反应流程:中间体1-5的制备
Figure PCTCN2017082227-appb-000096
步骤A:将N-Boc-L-脯氨醇(50克,248.43毫摩尔),邻苯二甲酰亚胺(43.86克,298.12毫摩尔)和三苯基膦(65.16克,248.43毫摩尔)溶于四氢呋喃(1升)中,然后在氮气保护下,0摄氏度逐滴滴加DIAD(48.31毫升,248.43毫摩尔)。然后升至室温搅拌16小时,加水(200毫升)淬灭,搅拌10分钟,并用乙酸乙酯(250毫升×2)萃取。将合并的有机层用盐水洗涤,无水硫酸钠干燥,并减压浓缩。残余物用硅胶柱纯化(洗脱剂:石油醚/乙酸乙酯=50/1至30/1),得到叔丁基(2S)-2-[(1,3-二氧代异吲哚-2-基)甲基]吡咯烷-1-羧酸酯(51克,62.14%)。
步骤B:将叔丁基(2S)-2-[(1,3-二氧代异吲哚-2-基)甲基]吡咯烷-1-羧酸酯(51克,154.37毫摩尔) 溶于乙醇(350毫升)中,然后在80摄氏度滴加水合肼(22.07毫升,385.93毫摩尔,85%纯度)。然后在该温度下搅拌16小时。冷却至室温,过滤,滤液减压浓缩,得到叔丁基(2S)-2-(氨甲基)吡咯烷-1-羧酸酯(29克,93.8%)。
步骤C:将叔丁基(2S)-2-(氨甲基)吡咯烷-1-羧酸酯(15克,74.9毫摩尔),碳酸钾(20.7克,149.8毫摩尔)混于乙腈(300毫升)中,室温加入1,4-二氟-2-硝基苯(13.11毫升,82.39毫摩尔)。混合物在氮气保护下,升温至80摄氏度,搅拌1小时,加水(200毫升)淬灭,并用乙酸乙酯(250毫升×2)萃取。将合并的有机层用盐水洗涤,无水硫酸钠干燥,并减压浓缩。残余物用硅胶柱纯化(洗脱剂:石油醚/乙酸乙酯=50/1至30/1),得到叔丁基(2S)-2-[(4-氟-2-硝基-苯胺基)甲基]吡咯烷-1-羧酸酯(28克,粗品),无需进一步纯化。
步骤D:将叔丁基(2S)-2-[(4-氟-2-硝基-苯胺基)甲基]吡咯烷-1-羧酸酯(11克,粗品)溶于甲醇(100毫升)和乙酸乙酯(100毫升),混合物在氮气保护下加入湿钯碳(200毫克)。在50psi的氢气气氛条件下反应10小时。反应液过滤浓缩,得到叔丁基(2S)-2-[(2-氨基-4-氟-苯胺基)甲基]吡咯烷-1-羧酸酯(9克,粗品)。
反应流程:中间体1-10的制备
Figure PCTCN2017082227-appb-000097
步骤A:往N-Cbz-L-脯氨酸(50.00克,200.59毫摩尔)的500毫升甲苯溶液中加入DMF(146.61毫克,2.01毫摩尔),然后在10到20摄氏度下滴加草酰氯(30.55克,240.71毫摩尔)。在10到30摄氏度下搅拌16个小时后,减压浓缩除去溶剂。粗产品N-Cbz-L-脯氨酰氯(53.70克,200.59毫摩尔)直接用于下一步。
步骤B:在-4到5摄氏度下,往6-氟吲哚(40.66克,300.88毫摩尔)的300毫升甲苯溶液中滴加乙基溴化镁(3摩尔/升,106.98毫升)。加完之后,反应液在-4到5摄氏度下搅拌30分钟后,在0到10度下滴加N-Cbz-L-脯氨酰氯(53.70克,200.59毫摩尔)的200毫升甲苯溶液。反应液在20到30摄氏度下搅拌2个小时。在不超过30摄氏度下往反应液里加入醋酸调节pH到3,然后加入1升水淬灭,加入1升的乙酸乙酯萃取,分液后,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩得到粗产品。粗产品经过柱层析(洗脱剂:乙酸乙酯/石油醚=1/1)得到橘黄色的固体产物苄基(2S)-2-(6-氟-1H-吲哚-3-羰基)吡咯烷-1-羧酸酯(47.00克,125.72毫摩尔,62.67%)。
1HNMR(DMSO,400MHz):δ12.06(br.s.,1H),8.45(d,J=12.0Hz,1H),8.17(ddd,J=5.6,8.7,17.5 Hz,1H),7.41-7.25(m,3H),7.13-6.99(m,3H),5.26-5.15(m,1H),5.11-5.02(m,1H),5.00-4.87(m,1H),3.57-3.45(m,2H),2.45-2.27(m,1H),1.95-1.79(m,3H)。
步骤C:在5到15摄氏度下,往苄基(2S)-2-(6-氟-1H-吲哚-3-羰基)吡咯烷-1-羧酸酯(58.00克,158.31毫摩尔)的600毫升四氢呋喃溶液中滴加LiBH4(2摩尔/升,158.31毫升),在此温度下搅拌2.5个小时。然后反应液冷却到5摄氏度,在30分钟内滴加甲烷磺酸(27.39克,284.96毫摩尔)。反应液在10到30摄氏度下搅拌16.5小时后,小心地加入100毫升冰水淬灭反应。然后用浓盐酸将反应液的pH调节到1。减压除去四氢呋喃,水溶液用100毫升乙酸乙酯萃取2次,有机相减压浓缩得到粗产品。粗产品经过柱层析(洗脱剂:乙酸乙酯/石油醚=3/1)得到产物(S)-苄基2-((6-氟-1H-3-吲哚)甲基)吡咯烷-1-羧酸酯(49.00克,139.05毫摩尔,87.83%)。
1HNMR(DMSO,400MHz):δ10.98-10.84(m,1H),7.69-7.04(m,8H),6.89-6.52(m,1H),5.22-5.09(m,2H),4.09-3.93(m,1H),3.36-3.27(m,2H),3.18-2.96(m,1H),2.72-2.56(m,1H),1.88-1.62(m,4H)。
步骤D:DMF(7.78克,106.41毫摩尔)和三氯氧磷(17.68克,115.31毫摩尔)的混合物在0摄氏度下搅拌30分钟。然后在此温度下加入(S)-苄基2-((6-氟-1H-3-吲哚)甲基)吡咯烷-1–羧酸酯(12.50克,35.47毫摩尔)的100毫升1,2-二氯乙烷溶液。反应液在20到30摄氏度下搅拌5.5个小时后,加入200毫升的饱和碳酸钠水溶液淬灭反应,用500毫升乙酸乙酯萃取2次,合并的有机相用500毫升的饱和食盐水洗涤,无水硫酸钠干燥后,过滤,滤液减压浓缩得到苄基(2S)-2-[(6-氟-2-甲酰基-1H-3-吲哚)甲基]吡咯烷-1-羧酸酯(17.00克,粗品)。
反应流程:实施例1的制备
Figure PCTCN2017082227-appb-000098
步骤A:将苄基(2S)-2-[(6-氟-2-甲酰基-1H-吲哚-3-基)甲基]吡咯烷-1-羧酸酯(7克,粗产品)和叔丁基(2S)-2-[(2-氨基-4-氟-苯胺基)甲基]吡咯烷-1-羧酸酯(6.26克,20.24毫摩尔)溶于DMF(30毫升)和水(1毫升),然后在25摄氏度向其中一次性加入Oxone(8.4克,55.2毫摩尔)。室温下搅拌1小时,加饱和亚硫酸钠水溶液(100毫升)淬灭,加水(250毫升)稀释,并用乙酸乙酯(250毫升×2)萃取。将合并的有机层用盐水洗涤,无水硫酸钠干燥,并减压浓缩。残余物用硅胶柱纯化(洗脱剂:石油醚/乙酸乙酯=50/1至30/1),得到苄基(2S)-2-[[2-[1-[[(2S)-1-叔-丁氧基羰基吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸酯(15克,粗产品)。
MS(ESI)m/z:670.5[M+H+]。
步骤B:将苄基(2S)-2-[[2-[1-[[(2S)-1-叔-丁氧基羰基吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸酯(1克,1.49毫摩尔)溶于甲醇(30毫升)和乙酸乙酯(30毫升),混合物在氮气保护下加入湿钯碳(100毫克)。在40psi下氢化3小时。反应液过滤浓缩,得到叔丁基(2S)-2-[[5-氟-2-[6-氟-3-[[(2S)-吡咯烷-2-基]甲基]-1H-吲哚-2-基]苯并咪唑-1-yl]甲基]吡咯烷-1-羧酸酯(750毫克,93.97%)。
步骤C:将叔丁基(2S)-2-[[5-氟-2-[6-氟-3-[[(2S)-吡咯烷-2-基]甲基]-1H-吲哚-2-基]苯并咪唑-1-yl]甲基]吡咯烷-1-羧酸酯(610毫克,1.14毫摩尔)溶于盐酸二氧六环(3毫升,4摩尔/升),室温下搅拌半小时。反应液减压浓缩,得到5-氟-2-[6-氟-3-[[(2S)-吡咯烷-2-基]甲基l]-1H-吲哚-2-基]-1-[[(2S)-吡咯烷-2-基甲基]苯并咪唑(579.6毫克,100%,盐酸盐)。
MS(ESI)m/z:436.2[M+H+]。
步骤D:向正在搅拌的N-Boc-L缬氨酸(743.53毫克,3.42毫摩尔)的DMF(2毫升)溶液中加入N-甲基吗啉(691.87毫克,6.84毫摩尔)和HATU(1.52克,3.99毫摩尔),室温下搅拌10分钟,然后将5-氟-2-[6-氟-3-[[(2S)-吡咯烷-2-基]甲基l]-1H-吲哚-2-基]-1-[[(2S)-吡咯烷-2-基甲基]苯并咪唑(579.6毫克,盐酸盐)溶于DMF(2毫升)加入上述溶液。室温下搅拌2小时,加水(100毫升)稀释,并用乙酸乙酯(100毫升×2)萃取。将合并的有机层用盐水洗涤,无水硫酸钠干燥,并减压浓缩。残余物用硅胶柱纯化(洗脱剂:乙酸乙酯/石油醚=3/1),得到叔丁基N-[(1S)-1-[(2S)-2-[[2-[1-[[(2S)-1-[(2S)-2-(叔-丁氧羰基胺基)-3-甲基-丁酰基]吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-yl]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(950毫克,76.94%)。
1HNMR(DMSO,400MHz):δ8.14-8.06(m,0.5H),8.00-7.85(m,1H),7.58-7.45(m,1H),7.32-7.19(m,1H),7.05-6.91(m,2H),6.84-6.76(m,0.5H),4.31(br.s.,1H),4.08-3.94(m,2H),3.60-3.47(m,1H),3.27(d,J=12.0Hz,1H),3.08-2.80(m,1H),2.70(s,11H),2.00(s,2H),1.48-1.43(m,2H),1.41-1.31(m,17H),1.30-1.14(m,5H),0.93-0.82(m,12H)。
MS(ESI)m/z:834.3[M+H+]。
步骤E:将叔丁基N-[(1S)-1-[(2S)-2-[[2-[1-[[(2S)-1-[(2S)-2-(叔-丁氧羰基胺基)-3-甲基-丁酰基]吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-yl]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(950毫克,1.14毫摩尔)溶于盐酸二氧六环(2毫升,4摩尔/升),室温下搅拌半小时。反应液减压浓缩,得到(2S)-2-胺基-1-[(2S)-2-[[2-[1-[[(2S)-1-[(2S)-2-胺基-3-甲基-丁酰基]吡咯烷-2-基]甲 基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-基]-3-甲基-丁烷-1-酮(800毫克,粗产品),直接用于下一步。
MS(ESI)m/z:643.3[M+H+]。
步骤F:向正在搅拌的N-Boc-N-甲基-L-丙氨酸(688.95毫克,3.39毫摩尔)的DMF(2毫升)溶液中加入N-甲基吗啉(685.8毫克,6.78毫摩尔)和HATU(1.5克,3.96毫摩尔),室温下搅拌10分钟,然后将(2S)-2-胺基-1-[(2S)-2-[[2-[1-[[(2S)-1-[(2S)-2-胺基-3-甲基-丁酰基]吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-基]-3-甲基-丁烷-1-酮(800毫克,粗产品)溶于DMF(2毫升)加入上述溶液。室温下搅拌2小时,加水(100毫升)稀释,并用乙酸乙酯(100毫升×2)萃取。将合并的有机层用盐水洗涤,无水硫酸钠干燥,并减压浓缩。残余物用硅胶柱纯化(洗脱剂:石油醚/乙酸乙酯=2:1至1:1),得到叔-丁基N-[(1S)-2-[[(1S)-1-[(2S)-2-[[2-[1-[[(2S)-1-[(2S)-2-[[(2S)-2-[叔-丁氧基羰基(甲基)胺基]丙酰基]胺基]-3-甲基-丁酰基]吡咯烷-2-yl]甲基]-5-氟-苯并咪唑-2-yl]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]胺基]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(910毫克,76.94%)。
1HNMR(DMSO,400MHz):δ11.81(s,1H),7.96(s,2H),7.55(dd,J=2.4,8.0Hz,1H),7.26(d,J=8.0Hz,2H),7.05-6.97(m,1H),4.49-4.20(m,4H),2.90(s,6H),2.74-2.73(m,8H),2.70(s,12H),2.34(s,1H),2.02-1.89(m,1H),1.40(d,J=3.8Hz,24H),1.26-1.19(m,6H),1.01-0.78(m,6H)。
MS(ESI)m/z:1004.4[M+H+]。
步骤G:将叔-丁基N-[(1S)-2-[[(1S)-1-[(2S)-2-[[2-[1-[[(2S)-1-[(2S)-2-[[(2S)-2-[叔-丁氧基羰基(甲基)胺基]丙酰基]胺基]-3-甲基-丁酰基]吡咯烷-2-yl]甲基]-5-氟-苯并咪唑-2-yl]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]胺基]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(910毫克,906.18微摩尔)溶于二氯甲烷(2毫升),加入三氟乙酸(2毫升,27.01毫摩尔),室温下搅拌半小时。反应液减压浓缩,残余物经制备HPLC纯化,得到实施例1(233毫克,盐酸盐)。
1HNMR(MeOD,400MHz):δ8.20(dd,J=4.0,8.0Hz,1H),8.01-7.91(m,2H),7.59-7.49(m,2H),7.12(dt,J=4.0,12.0Hz,1H),5.04-4.97(m,1H),4.87-4.79(m,1H),4.58(dd,J=4.0,8.0Hz,2H),4.54-4.48(m,1H),4.34(br.s.,1H),4.09-3.76(m,6H),3.59(d,J=12.0Hz,1H),3.29-3.18(m,1H),2.71(d,J=12.0Hz,6H),2.26(d,J=8.0Hz,3H),2.06(d,J=8.0Hz,6H),1.82-1.73(m,1H),1.54(dd,J=8.0,8.0Hz,6H),1.13(dd,J=4.0,8.0Hz,6H),0.96(t,J=8.0Hz,6H)。
MS(ESI)m/z:804.5[M+H+]。
实施例2
Figure PCTCN2017082227-appb-000099
反应流程:实施例2的制备
Figure PCTCN2017082227-appb-000100
步骤A:将苄基(2S)-2-[[2-[1-[[(2S)-1-叔-丁氧基羰基吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸酯(3克,4.48毫摩尔)溶于盐酸二氧六环(10毫升,4摩尔/升),室温下搅拌半小时。反应液减压浓缩,残余物用乙酸乙酯(50毫升)打浆,得到产物苄基(2S)-2-[[6-氟2-[5-氟-1-[[(2S)-吡咯烷-2-基]甲基]苯并咪唑-2-基]-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸酯(1.7克,粗品),直接用下一步。
1HNMR(DMSO,400MHz):δ11.95(d,J=19.3Hz,1H),9.71-9.21(m,2H),8.10(br.s.,1H),7.88(dd,J=5.4,8.4Hz,1H),7.67-7.57(m,1H),7.41-7.29(m,7H),7.08-6.67(m,1H),5.00(s,2H),4.26(br.s.,4H),3.33-3.15(m,4H),3.02(br.s.,1H),1.99(s,1H),1.75-1.46(m,8H)
步骤B:向正在搅拌的N-Boc-L缬氨酸(385.46毫克,1.65毫摩尔)的DMF(1毫升)溶液中加入N-甲基吗啉(333.77毫克,3.3毫摩尔)和HATU(690.08毫克,1.81毫摩尔),室温下搅拌30分钟,然后将苄基(2S)-2-[[6-氟2-[5-氟-1-[[(2S)-吡咯烷-2-基]甲基]苯并咪唑-2-基]-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸盐(500毫克,粗品)溶于DMF(1毫升)加入上述溶液。室温下搅拌2小时,加水(100毫升)稀释,并用乙酸乙酯(100毫升×2)萃取。将合并的有机层用盐水洗涤,无水硫酸钠干燥,并减压浓缩。残余物用硅胶柱纯化(洗脱剂:石油醚/乙酸乙酯=5/1至2/1),得到苄基(2S)-2-[[2-[1-[[(2S)-1-[(2S)-2-(叔-丁氧基羰基胺基)-3-甲基-丁酰基]吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸酯(570毫克,89.86%)。
1HNMR(DMSO,400MHz):δ11.77-11.98(m,1H),7.48-7.55(m,1H),7.36(s,7H),7.20-7.28(m,2H),6.92(s,1H),5.03(s,2H),4.34-4.39(m,1H),4.03(d,J=7.2Hz,1H),2.69(s,4H),2.31-2.35(m,1H),1.99(s,2H),1.45(s,2H),1.36-1.40 (m,6H),1.36(s,9H),1.18(t,J=7.1Hz,2H),0.96(d,J=6.9Hz,2H),0.88ppm(s,6H)。
MS(ESI)m/z:769.4[M+H+]。
步骤C:将苄基(2S)-2-[[2-[1-[[(2S)-1-[(2S)-2-(叔-丁氧基羰基胺基)-3-甲基-丁酰基]吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸酯(570毫克,741.33微摩尔)溶于甲醇(15毫升)和乙酸乙酯(15毫升),混合物在氮气保护下加入湿钯碳(50毫克,10%)。在45psi下氢化16小时。反应液过滤浓缩,得到叔丁基N-[(1S)-1-[(2S)-2-[[5-氟-2-[6-氟-3-[[(2S)-吡咯烷-2-基]甲基]-1H-吲哚-2-基]苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(380毫克,粗品),直接用于下一步。
MS(ESI)m/z:635.4[M+H+]。
步骤D:向正在搅拌的N-Boc-L正丁氨酸(243.34毫克,1.2毫摩尔)的DMF(1毫升)溶液中加入N-甲基吗啉(302.77毫克,2.99毫摩尔)和HATU(569.06毫克,1.5毫摩尔),然后将叔丁基N-[(1S)-1-[(2S)-2-[[5-氟-2-[6-氟-3-[[(2S)-吡咯烷-2-基]甲基]-1H-吲哚-2-基]苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(380毫克,粗品)溶于DMF(1毫升)加入上述溶液。室温下搅拌1小时,加水(50毫升)稀释,并用乙酸乙酯(50毫升×2)萃取。将合并的有机层用盐水洗涤,无水硫酸钠干燥,并减压浓缩。残余物用硅胶柱纯化(洗脱剂:石油醚/乙酸乙酯=4/1至3/1),得到叔丁基N-[(1S)-1-[(2S)-2-[[2-[3-[[(2S)-1-[(2S)-2-(叔-丁氧羰基胺基)丁酰基]吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(380毫克,77.41%)。
MS(ESI)m/z:820.5[M+H+]。
步骤E:将叔丁基N-[(1S)-1-[(2S)-2-[[2-[3-[[(2S)-1-[(2S)-2-(叔-丁氧羰基胺基)丁酰基]吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(380毫克,463.43微摩尔)溶于盐酸二氧六环(2毫升,4摩尔/升),室温下搅拌半小时。反应液减压浓缩,得到粗品(2S)-2-胺基-1-[(2S)-2-[[2-[3-[[(2S)-1-[(2S)-2-胺基丁酰基]吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-基]-3-甲基-丁-1-酮(320毫克,盐酸盐),直接用于下一步。
MS(ESI)m/z:620.3[M+H+]。
步骤F:向正在搅拌的N-Boc-N-甲基-L-丙氨酸(281.68毫克,1.39毫摩尔)的DMF(1毫升)溶液中加入N-甲基吗啉(233.65毫克,2.31毫摩尔)和HATU(439.15毫克,1.15毫摩尔),然后将(2S)-2-胺基-1-[(2S)-2-[[2-[3-[[(2S)-1-[(2S)-2-胺基丁酰基]吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-基]-3-甲基-丁-1-酮(320毫克,461.98微摩尔,盐酸盐)溶于DMF(1毫升)加入上述溶液。室温下搅拌1小时,加水(50毫升)稀释,并用乙酸乙酯(50毫升×2)萃取。将合并的有机层用盐水洗涤,无水硫酸钠干燥,并减压浓缩。残余物用硅胶柱纯化(洗脱剂:石油醚/乙酸乙酯=2/1至1/2),得到叔丁基N-[(1S)-2-[[(1S)-1-[(2S)-2-[[2-[3-[[(2S)-1-[(2S)-2-[[(2S)-2-[叔-丁氧羰基(甲基)胺基]吡咯烷]胺基]丁酰基]吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]胺基]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(450毫克,98.37%)。
MS(ESI)m/z:990.3[M+H+]。
步骤G:将叔丁基N-[(1S)-2-[[(1S)-1-[(2S)-2-[[2-[3-[[(2S)-1-[(2S)-2-[[(2S)-2-[叔-丁氧羰基(甲基)胺基]吡咯烷]胺基]丁酰基]吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]胺基]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(450毫克,454.46微摩尔)溶于二氯甲烷(2毫升),加入三氟乙酸(2毫升,27.01毫摩尔),室温下搅拌半小时。反应液减压浓缩,残余物经制备制备HPLC纯化,得到实施例2(117毫克,28.84%,盐酸盐)。
1HNMR(MeOD,400MHz):δ8.71(d,J=8.0Hz,1H),8.21(dd,J=4.0,8.0Hz,1H),7.99-7.88(m,2H),7.58-7.50(m,2H),7.12(dt,J=4.0,8.0Hz,1H),5.26-5.13(m,1H)4.88-4.76(m,1H),4.67(dd,J=8.0,8.0Hz,1H),4.60-4.45(m,2H),4.33(br.s.,1H),4.08-3.97(m,2H),3.91-3.78(m,4H),3.54(d,J=12.0Hz,1H),3.27-3.16(m,1H),2.70(d,J=12.0Hz,6H),2.41-2.20(m,2H),2.17-1.67(m,9H),1.63-1.50(m,6H),1.12(t,J=8.0Hz,3H),0.96(dd,J=8.0,16.0Hz,6H)。
MS(ESI)m/z:790.5[M+H+]。
实施例3至12均可参照实施例2的制备反应流程制得。
实施例3
Figure PCTCN2017082227-appb-000101
1HNMR(MeOD,400MHz):δ12.49(s,1H),8.92(d,J=6.7Hz,0.5H),8.51(d,J=7.7Hz,1H),8.25-8.16(m,1H),7.97-7.88(m,2H),7.56-7.47(m,2H),7.14-7.06(m,1H),5.00-4.95(m,1H),4.70-4.64(m,1H),4.56(d,J=6.8Hz,1H),4.50(br.s.,1H),4.33(br.s.,1H),4.04(d,J=6.8Hz,2H),3.93-3.76(m,4H),3.51(d,J=13.9Hz,1H),3.26-3.16(m,1H),2.71(s,3H),2.66(s,3H),2.41-2.18(m,2H),2.17-1.71(m,8H),1.59(d,J=7.0Hz,3H),1.46(d,J=6.8Hz,3H),1.11(t,J=7.3Hz,3H),1.00(s,9H)。
MS(ESI)m/z:804.5[M+H+]。
实施例4
Figure PCTCN2017082227-appb-000102
1HNMR(MeOD,400MHz):δ12.41(s,1H),8.89(d,J=7.0Hz,1H),8.64(d,J=7.7Hz,1H),8.18(dd,J=4.0,9.1Hz,1H),7.99-7.84(m,2H),7.57-7.44(m,2H),7.10(dt,J=2.1,9.2Hz,1H),5.00-4.92(m,1H),4.69-4.63(m,1H),4.54-4.43(m,2H),4.31(br.s.,1H),4.04-3.76(m,6H),3.52(d,J=13.2Hz,1H),3.18(dd,J=11.2,14.1Hz,1H),2.71(s,3H),2.66(s,3H),2.38-2.17(m,2H),2.10-1.62(m,14H),1.57(d,J=7.0Hz,3H),1.48(d,J=6.8Hz,3H),1.38-0.91(m,9H)。
MS(ESI)m/z:830.5[M+H+]。
实施例5
Figure PCTCN2017082227-appb-000103
1HNMR(MeOD,400MHz):δ12.33(s,1H),8.72-8.61(m,2H),8.15(dd,J=3.9,9.0Hz,1H),7.99-7.90(m,2H),7.55-7.44(m,2H),7.09(dt,J=2.0,9.2Hz,1H),4.76-4.69(m,1H),4.56-4.42(m,2H),4.31(br.s.,1H),4.10(q,J=6.5Hz,1H),3.96-3.84(m,3H),3.75(d,J=7.2Hz,2H),3.54(d,J=13.9Hz,1H),3.26-3.12(m,1H),2.68(d,J=13.9Hz,6H),2.38-2.19(m,2H),2.07-1.59(m,7H),1.51(t,J=6.8Hz,8H),1.17(s,9H),1.03-0.83(m,3H)。
MS(ESI)m/z:804.2[M+H+]。
实施例6
Figure PCTCN2017082227-appb-000104
1HNMR(MeOD,400MHz):δ12.35(s,1H),8.82(d,J=7.7Hz,1H),8.71(d,J=6.8Hz,1H),8.22-8.10(m,1H),8.00-7.85(m,2H),7.57-7.43(m,2H),7.09(t,J=8.3Hz,1H),4.85-4.79(m,1H),4.61-4.46(m,3H),4.31(br.s.,1H),4.05-3.73(m,6H),3.61-3.49(m,1H),3.25-3.12(m,1H),2.68(d,J=10.0Hz,6H),2.28(br.s.,2H),2.08-1.62(m,14H),1.61-1.41(m,8H),1.41-1.01(m,6H),0.94(t,J=7.2Hz,3H)。
MS(ESI)m/z:830.3[M+H+]。
实施例7
Figure PCTCN2017082227-appb-000105
1HNMR(MeOD,400MHz):δ12.45(s,1H),8.91(d,J=7.1Hz,1H),8.67(d,J=7.7Hz,1H),8.20(dd,J=3.8,9.0Hz,1H),7.97-7.86(m,2H),7.56-7.46(m,2H),7.10(dt,J=2.1,9.2Hz,1H),5.00(d,J=4.6Hz,1H),4.83-4.78(m,1H),4.71(dd,J=5.4,8.3Hz,1H),4.58-4.46(m,2H),4.31(br.s.,1H),4.01(quin,J=6.9Hz,2H),3.90-3.74(m,4H),3.50(d,J=13.4Hz,1H),3.20(dd,J=11.2,13.9Hz,1H), 2.69(d,J=12.8Hz,6H),2.39-2.18(m,2H),2.10-1.75(m,9H),1.61-1.45(m,8H),1.05-0.86(m,9H)。
MS(ESI)m/z:804.5[M+H+]。
实施例8
Figure PCTCN2017082227-appb-000106
1HNMR(MeOD,400MHz):δ8.94(d,J=7.1Hz,1H),8.54(d,J=7.8Hz,1H),8.21(dd,J=3.9,9.2Hz,1H),7.97-7.87(m,2H),7.56-7.46(m,2H),7.10(dt,J=2.0,9.1Hz,1H),5.03-4.92(m,2H),4.76-4.71(m,1H),4.61-4.46(m,2H),4.32(br.s.,1H),4.09-3.96(m,2H),3.93-3.74(m,4H),3.50(d,J=13.8Hz,1H),3.21(dd,J=11.4,13.9Hz,1H),2.68(d,J=19.2Hz,6H),2.43-2.15(m,2H),2.11-1.75(m,8H),1.61-1.45(m,8H),1.07-0.91(m,12H)。
MS(ESI)m/z:818.5[M+H+]。
实施例9
Figure PCTCN2017082227-appb-000107
1HNMR(MeOD,400MHz):δ12.41(s,1H),8.91(d,J=6.8Hz,1H),8.65(d,J=7.6Hz,1H),8.20(dd,J=3.8,9.0Hz,1H),7.98-7.85(m,2H),7.57-7.45(m,2H),7.09(dt,J=1.8,9.1Hz,1H),4.84(dd,J=8.3,14.8Hz,2H),4.74-4.68(m,1H),4.54-4.42(m,2H),4.30(br.s.,1H),4.05-3.93(m,2H),3.90-3.73(m,4H),3.49(d,J=13.3Hz,1H),3.19(dd,J=11.2,13.7Hz,1H),2.73-2.61(m,6H),2.41-2.16(m,2H),2.06-1.67(m,12H),1.60-1.36(m,10H),1.20(br.s.,3H),1.11-0.97(m,5H)。
MS(ESI)m/z:844.5[M+H+]。
实施例10
Figure PCTCN2017082227-appb-000108
1HNMR(MeOD,400MHz):δ8.16(dd,J=3.8,9.1Hz,1H),8.01-7.87(m,2H),7.56-7.40(m,2H), 7.09(dt,J=2.0,9.2Hz,1H),4.88(d,J=7.0Hz,2H),4.55(d,J=8.0Hz,1H),4.47(d,J=7.2Hz,2H),4.31(br.s.,1H),4.05(q,J=6.9Hz,1H),3.97-3.88(m,2H),3.87-3.81(m,1H),3.78-3.69(m,2H),3.55(d,J=13.3Hz,1H),3.17(dd,J=11.1,14.0Hz,1H),2.75-2.62(m,6H),2.39-2.17(m,3H),2.02(d,J=3.0Hz,2H),1.93-1.78(m,4H),1.58-1.45(m,6H),1.35-1.19(m,4H),1.16-1.00(m,6H),0.90(t,J=6.4Hz,3H)。
MS(ESI)m/z:804.2[M+H+]。
实施例11
Figure PCTCN2017082227-appb-000109
1HNMR(MeOD,400MHz):δ8.73-8.54(m,1H),8.15(dd,J=3.8,9.0Hz,1H),8.01-7.88(m,2H),7.60-7.42(m,2H),7.09(dt,J=2.0,9.2Hz,1H),4.74-4.70(m,1H),4.47(br.s.,2H),4.30(br.s.,1H),4.15-4.06(m,1H),3.96-3.85(m,3H),3.80-3.68(m,2H),3.54(d,J=13.8Hz,1H),3.18(dd,J=11.3,13.9Hz,1H),2.73-2.63(m,6H),2.30(d,J=5.4Hz,2H),2.16-1.66(m,7H),1.56-1.46(m,6H),1.34-1.22(m,4H),1.17(s,9H),1.03-0.82(m,4H)。
MS(ESI)m/z:818.3[M+H+]。
实施例12
Figure PCTCN2017082227-appb-000110
1HNMR(MeOD,400MHz):δ8.81(d,J=7.5Hz,1H),8.67(d,J=7.2Hz,1H),8.16(dd,J=3.7,8.8Hz,1H),7.96(dd,J=5.1,8.8Hz,1H),7.92-7.85(m,1H),7.58-7.43(m,2H),7.09(dt,J=1.9,9.1Hz,1H),4.62-4.46(m,3H),4.30(br.s.,1H),4.05-3.71(m,6H),3.55(d,J=13.7Hz,1H),3.21-3.11(m,1H),2.67(d,J=14.6Hz,6H),2.28(br.s.,2H),2.06-1.59(m,13H),1.58-1.46(m,6H),1.44-1.13(m,10H),0.97-0.85(m,3H)。
MS(ESI)m/z:844.3[M+H+]。
实施例13
Figure PCTCN2017082227-appb-000111
反应流程:中间体13-7的制备
Figure PCTCN2017082227-appb-000112
步骤A:氮气氛围下,在-78摄氏度下往搅拌着的N-Boc-反式-4-羟基-L-脯氨酸甲酯(262.00克,1.07摩尔)的2.5升二氯甲烷溶液中滴加DAST(258.28克,1.60摩尔)。在-78摄氏度下搅拌3小时后,升温到10到20摄氏度,再搅拌15个小时。将反应液倒入到0摄氏度的3升饱和碳酸氢钠溶液中进行淬灭,用6升的二氯甲烷萃取2次。有机相用3升的饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产品。粗产品经过柱层析(洗脱剂,石油醚/乙酸乙酯=50:1至10:1)得到N-Boc-顺式-4-氟-L-脯氨酸甲酯(53.00克,214.35毫摩尔,20.07%)。
1HNMR(CDCl3,400MHz):δ5.30-5.11(m,1H),4.58-4.39(m,1H),3.94-3.55(m,5H),2.56-2.24(m,2H),1.53-1.40(m,9H)。
步骤B:在0到5摄氏度下往N-Boc-顺式-4-氟-L-脯氨酸甲酯(80.00克,323.55毫摩尔)的1升四氢呋喃溶液中分批加入LiBH4(16.00克,734.62毫摩尔)。反应液在10到20摄氏度下搅拌16个小时后,加入1500毫升饱和碳酸氢钠水溶液淬灭后,用3000毫升乙酸乙酯萃取两次。合并的有机相减压浓缩得到粗产品。粗产品溶于500毫升二氯甲烷,用500毫升饱和食盐水洗涤后,用无水硫酸钠干燥,过滤,滤液减压浓缩得到N-Boc-顺式-4-氟-L-脯氨醇(66.00克,粗品)。
1HNMR(CDCl3,400MHz):δ5.20-4.98(m,1H),4.18(br.s.,1H),4.14-4.04(m,1H),3.83-3.74(m,1H),3.69-3.60(m,1H),3.59-3.48(m,2H),2.28-2.07(m,1H),2.02-1.87(m,1H),1.41(s,9H)。
步骤C:在10到20摄氏度下,往N-Bco-顺式-4-氟-L-脯氨醇(66.00克,301.03毫摩尔,粗品)的700毫升四氢呋喃溶液中加入邻苯二甲酰亚胺(46.50克,316.08毫摩尔)和三苯基膦(82.90克,316.08毫摩尔)。然后在0~10摄氏度下滴加DIAD(63.91克,316.08毫摩尔)。反应液在10~20摄氏度下搅拌16个小时后,减压除去溶剂。往残留物里加入500毫升水,用1000毫升二氯甲烷萃取2次。合并的有 机相用500毫升饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产品。粗产品柱层析(洗脱剂:石油醚/乙酸乙酯=10/1至5/1)得到叔丁基(2S,4S)-2-[(1,3-二氧-2-异吲哚啉)甲基]-4-氟-吡咯烷-1-羧酸酯(133.00克,粗品)。
1HNMR(CDCl3,400MHz):δ7.93-7.64(m,4H),4.55-4.31(m,1H),4.10-4.01(m,1H),3.87-3.55(m,3H),2.32-2.07(m,2H),1.27(d,J=6.3Hz,9H)。
MS(ESI)m/z:249.1[M+H+-100]。
步骤D:往叔丁基(2S,4S)-2-[(1,3-二氧-2-异吲哚啉)甲基]-4-氟-吡咯烷-1-羧酸酯(133.00克,381.78毫摩尔,粗品)的1升乙醇溶液中加入水合肼(48.75克,954.44毫摩尔。在60摄氏度下反应2个小时后冷去到室温,加入1升的二氯甲烷稀释,过滤,滤饼用二氯甲烷洗涤。合并的有机相减压浓缩得到残余物。残余物用200毫升二氯甲烷稀释,过滤,滤饼用二氯甲烷洗涤,合并的有机相减压浓缩得到叔丁基(2S,4S)-2-(胺基甲基)-4-氟-吡咯烷-1-羧酸酯(112.00克,粗品)。
1HNMR(CDCl3,400MHz):δ5.27-5.08(m,1H),3.95-3.77(m,1H),3.59(d,J=10.6Hz,1H),2.98(br.s.,1H),2.73(dd,J=7.8,12.2Hz,1H),2.55-2.41(m,2H),2.19(br.s.,1H),1.45(s,9H)。
步骤E:在10到20摄氏度下,往(2S,4S)-2-(胺基甲基)-4-氟-吡咯烷-1-羧酸酯(66.00克,粗品)的1升乙腈溶液中加入1,4-二氟-2-硝基-苯(45.70克,287.26毫摩尔),加入碳酸钾(3.58克,604.76毫摩尔)。在80摄氏度下反应2个小时后,冷却到10到20摄氏度。将反应液过滤,滤液浓缩得到粗产品。将粗产品溶于500毫升甲基叔丁基醚中,搅拌16个小时候,析出黄色固体,然后过滤,滤饼干燥得到叔丁基(2S,4S)-4-氟-2-[(4-氟2-硝基-苄胺基)甲基]吡咯烷-1-羧酸酯(68.00克,190.29毫摩尔,62.93%)。
1HNMR(CDCl3,400MHz):δ8.35-8.11(m,1H),7.89(d,J=9.0Hz,1H),7.35-7.16(m,2H),5.40-5.17(m,1H),4.33-4.16(m,1H),3.88-3.52(m,3H),3.45-3.30(m,1H),2.37-2.06(m,2H),1.59-1.48(m,9H)。
MS(ESI)m/z:380.1[M+Na]。
步骤F:在氮气氛围下往叔丁基(2S,4S)-4-氟-2-[(4-氟2-硝基-苄胺基)甲基]吡咯烷-1-羧酸酯(20.00克,55.97毫摩尔)的200毫升甲醇和1升的乙酸乙酯混合溶液中加入Pd/C(10%,2g)。体系用氢气置换三次,在25到30摄氏度,压力为40Psi的氢气氛围下搅拌4个小时。反应液过滤,滤液减压浓缩得到叔丁基(2S,4S)-2-[(2-胺基-4-氟-苄胺基)甲基]-4-氟-吡咯烷-1-羧酸酯(18.00克,53.33毫摩尔,95.29%)。
MS(ESI)m/z:328.1[M+H+]。
1HNMR(CDCl3,400MHz):δ6.48(br.s.,2H),6.41-6.30(m,2H),5.27-5.05(m,1H),4.35-4.21(m,1H),3.72-3.27(m,5H),3.10(dd,J=6.5,12.0Hz,1H),2.31-2.01(m,2H),1.41(br.s.,9H)。
反应流程:中间体13-14的制备
Figure PCTCN2017082227-appb-000113
步骤A:将N-Cbz-反式-4-羟基-L-脯氨酸甲酯(21.00克,57.14毫摩尔)溶于无水二氯甲烷(90.00毫升),然后在-78度,氮气保护下滴加DAST(21.65克,134.29毫摩尔)。-78度下搅拌3小时后,升至室温(25度)搅拌15小时.反应在0度下用饱和碳酸氢钠溶液(600毫升)淬灭后,用二氯甲烷萃取(600毫升×2)。有机相用饱和食盐水洗(600毫升×1),无水硫酸钠干燥,过滤,滤液45度下减压浓缩。所得残渣通过硅胶柱色谱法纯化(石油醚/乙酸乙酯=1/0至4/1)得到N-苄氧羰基-顺式-4-氟-L-脯氨酸甲酯(14.00克,85.80%)。
1HNMR(CDCl3,400MHz):δ7.43-7.27(m,5H),5.31-5.07(m,3H),4.65-4.51(m,1H),3.97-3.62(m,5H),2.51(d,J=18.4Hz,2H)。
步骤B:将N-Cbz-顺式-4-氟-L-脯氨酸甲酯(14.00克,49.03毫摩尔)溶于四氢呋喃(70.00毫升),然后25度下加入氢氧化锂溶液(1摩尔/升,69.13毫升)。反应25度下搅拌14小时。用1摩尔/升盐酸溶液调至pH等于3,加入200毫升水后,用乙酸乙酯萃取(200毫升×4)。有机相用无水硫酸钠干燥,过滤,滤液45度下减压浓缩得到N-Cbz-顺式-4-氟-L-脯氨酸(13.00克,97.53%)。
1HNMR(DMSO,400MHz):δ13.33-12.02(m,1H),7.49-7.12(m,5H),5.43-5.19(m,1H),5.17-5.00(m,2H),4.56-4.31(m,1H),3.85-3.50(m,2H),2.49-2.20(m,2H)。
步骤C:将N-Cbz-顺式-4-氟-L-脯氨酸(5.00克,18.39毫摩尔)溶于甲苯中(25.00毫升),然后在25度下加入DMF(13.44毫克,183.91微摩尔,14.15微升)和草酰氯(2.80克,22.07毫摩尔,1.93毫升)。反应25度下搅拌1小时后,45度下减压浓缩得黄色的油状化合物N-Cbz-顺式-4-氟-L-脯氨酰氯(5.30克,粗品),直接用于下一步。
步骤D:将6-氟-1H-吲哚(5.01克,37.10毫摩尔)溶于甲苯中(60.00毫升),氮气保护下,冰/丙酮浴(-4度)滴加乙基溴化镁(3摩尔/升,12.55毫升),反应液-4度搅拌1小时。然后将苄基N-Cbz-顺式-4-氟-L-脯氨酰氯(5.30克,粗品)溶于甲苯中于-4度滴入上述反应液中。混合液-4度继续搅拌2小时,升至25度搅拌14小时。反应液用醋酸(1毫升)淬灭后,用乙酸乙酯(300毫升)和水(300毫升)稀释,有机相分离后用饱和食盐水洗(200毫升×2),无水硫酸钠干燥,过滤,滤液45度下减压浓缩,所得残渣通过硅胶柱色谱法纯化(石油醚/乙酸乙酯=1/0至1/1)得到苄基(2S,4S)-4-氟-2-(6-氟-1H-吲哚-3-羰基)吡咯烷-1-羧酸酯(4.40克,11.45毫摩尔,61.71%)。
1HNMR(DMSO,400MHz):δ12.17-12.01(m,1H),8.56-8.31(m,1H),8.23-8.06(m,1H),7.46-6.98 (m,7H),5.45-5.21(m,2H),5.16-4.93(m,2H),3.96-3.64(m,2H),2.90-2.60(m,1H),2.42-2.20(m,1H)。
步骤E:将苄基(2S,4S)-4-氟-2-(6-氟-1H-吲哚-3-羰基)吡咯烷-1-羧酸酯(4.40克,11.45毫摩尔)溶于四氢呋喃(44.00毫升),然后在25度下缓慢滴加硼氢化锂溶液(2摩尔/升,11.45毫升)(20分钟滴完)。混合液25度下搅拌16小时后,0度下滴入甲烷磺酸(2.04克,21.18毫摩尔,1.51毫升)后继续搅拌2小时。之后0度下加入200毫升的水淬灭,混合液用乙酸乙酯萃取(200毫升×2),合并有机相后于45度减压浓缩。所得残渣通过硅胶柱色谱法纯化(石油醚/乙酸乙酯=3/1至1/1)得到苄基(2R,4S)-4-氟-2-[(6-氟-1H-吲哚-3-基)甲基]吡咯烷-1-羧酸酯(2.00克,4.64毫摩尔,40.56%)。
1HNMR(DMSO,400MHz):δ11.05-10.81(m,1H),7.75-6.50(m,9H),5.16(s,3H),4.21-4.00(m,1H),3.81-3.57(m,2H),3.18(d,J=5.3Hz,1H),2.81-2.65(m,1H),2.09(s,2H)。
步骤F:将三氯氧磷(534.02毫克,3.48毫摩尔)于0度,氮气保护下滴入DMF(254.56毫克,3.48毫摩尔),混合液0度下搅拌1小时。然后将苄基(2R,4S)-4-氟-2-[(6-氟-1H-吲哚-3-基)甲基]吡咯烷-1-羧酸酯(1.00克,2.32毫摩尔)溶于1,2-二氯乙烷(5.00毫升)后于0度下滴入上述混合液。反应液25度下搅拌18小时后,0度下倒入饱和的碳酸钠溶液(100毫升)中,混合液用乙酸乙酯(150毫升×2)萃取,有机相并用饱和食盐水(100毫升×2)洗,无水硫酸钠干燥,过滤,滤液45度下减压浓缩得到粗品苄基(2R,4S)-4-氟-2-[(6-氟-2-甲酰基-1H-吲哚-3-基)甲基]吡咯烷-1-羧酸酯(1.20克,粗品)。
MS ESI:399.0[M+H+]。
反应流程:实施例13的制备
Figure PCTCN2017082227-appb-000114
步骤A:将苄基(2R,4S)-4-氟-2-[(6-氟-2-甲酰基-1H-吲哚-3-基)甲基]吡咯烷-1-羧酸酯(1.00克,2.52毫摩尔)、叔丁基(2S,4S)-2-[(2-氨基-4-氟-苯胺)甲基]-4-氟-吡咯烷-1-羧酸酯(800.00毫克,2.39毫摩尔)溶于DMF(12.00毫升)和水(1.00毫升)的混合溶剂中,然后25度下一次加入Oxone(1.09克,7.18毫摩尔),混合液25度下搅拌1小时后,用饱和亚硫酸钠溶液(300毫升)和乙酸乙酯(300毫升)稀释,有机相分离后用饱和食盐水(200毫升×3)洗,无水硫酸钠干燥,过滤,滤液45度下减压浓缩。所得残渣通过硅胶柱色谱法纯化(石油醚/乙酸乙酯=1/0至3/1)得棕色固体苄基(2R,4S)-2-[[2-[1-[[(2S,4S)-1-叔丁氧羰基-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羧酸酯(1.40克,1.69毫摩尔,70.55%)。
1HNMR(DMSO,400MHz):δ11.89-11.22(m,1H),8.15-6.01(m,11H),5.12(br.s.,4H),4.71-4.43(m,1H),4.40-4.09(m,3H),3.76-3.38(m,4H),3.03-2.63(m,4H),1.94-1.68(m,2H),1.62-1.19(m,9H)。
步骤B:将苄基(2R,4S)-2-[[2-[1-[[(2S,4S)-1-叔丁氧羰基-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羧酸酯(1.40克,1.69毫摩尔)溶于乙酸乙酯(50.00毫升)和甲醇(10.00毫升)混合液,然后25度氮气保护下一次加入钯/碳(200.00毫克,10%)。所得悬浊液真空下脱气并用氢气置换几次后,25度氢气氛下(40psi)搅拌6小时。混合液用硅藻土过滤,甲醇(约200毫升)洗,滤液减压浓缩得叔丁基(2S,4S)-4-氟-2-[[5-氟-2-[6-氟-3-[[(2R,4S)-4-氟吡咯烷-2-基]甲基]-1H- 吲哚-2-基]苯并咪唑-1-基]甲基]吡咯烷-1-羧酸酯(900.00毫克,1.42毫摩尔,84.21%)。
MS(ESI)m/z:572.4[M+H+]。
步骤C:将叔丁基(2S,4S)-4-氟-2-[[5-氟-2-[6-氟-3-[[(2R,4S)-4-氟吡咯烷-2-基]甲基]-1H-吲哚-2-基]苯并咪唑-1-基]甲基]吡咯烷-1-羧酸酯(200.00毫克,314.90微摩尔)溶于1,4-二氧六环(4.00毫升),然后25度下滴入氯化氢/1,4-二氧六环(4摩尔/升,3.60毫升),混合液25度下搅拌30分钟,减压蒸馏掉溶剂得5-氟-2-[6-氟-3-[[(2R,4S)-4-氟吡咯烷-2-基]甲基]-1H-吲哚-2-基]-1-[[(2R,4S)-4-氟吡咯烷-2-基]甲基]苯并咪唑(175.00毫克,314.06微摩尔,99.73%,盐酸盐)。
MS ESI m/z:472.3[M+H+]。
步骤D:将N-Boc-L正丁氨酸(158.65毫克,780.65微摩尔)溶于DMF(3.00毫升)中,加入N-甲基吗啉(126.34毫克,1.25毫摩尔,137.33微升)和HATU(124.67毫克,327.87微摩尔)。混合液在25度下搅拌30分钟。然后加入5-氟-2-[6-氟-3-[[(2R,4S)-4-氟吡咯烷-2-基]甲基]-1H-吲哚-2-基]-1-[[(2R,4S)-4-氟吡咯烷-2-基]甲基]苯并咪唑(85.25毫克,156.13微摩尔,盐酸盐),混合液25度继续搅拌30分钟后用乙酸乙酯(100毫升)和水(100毫升)稀释,有机相分离后用饱和食盐水洗(100毫升×3),减压浓缩,所得残渣通过硅胶薄层色谱纯化(石油醚/乙酸乙酯=1/1)得淡黄色固体叔丁基-N-[(1S)-1-[(2R,4S)-2-[[2-[1-[[(2S,4S)-1-[(2S)-2-(叔丁氧羰基氨基)丁酰基]-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羰基]正丙基]氨基甲酸酯(110.00毫克,127.65微摩尔,81.76%)。
MS(ESI)m/z:842.1[M+H+]。
步骤E:将叔丁基-N-[(1S)-1-[(2R,4S)-2-[[2-[1-[[(2S,4S)-1-[(2S)-2-(叔-丁氧羰基氨基)丁酰基]-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羰基]正丙级]氨基甲酸酯(110.00毫克,127.65微摩尔)溶于1,4-二氧六环(1.00毫升),然后25度下滴入氯化氢/1,4-二氧六环(4摩尔/升,1.00毫升)混合液25度下搅拌30分钟,45度下减压蒸馏掉溶剂得淡黄色固体(2S)-2-氨基-1-[(2R,4S)-2-[[2-[1-[[(2S,4S)-1-[(2S)-2-氨基丁酰基]-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-基]丁烷-1-酮(95.00毫克,116.32微摩尔,91.12%,盐酸盐)。
步骤F:将N-Boc-N-甲基-L-丙氨酸(69.46毫克,341.79微摩尔)溶于DMF(2.00毫升),25度下加入NMM(92.19毫克,911.44微摩尔,100.21微升)和HATU(134.29毫克,353.18微摩尔),混合液25度下搅拌30分钟,然后将(2S)-2-氨基-1-[(2R,4S)-2-[[2-[1-[[(2S,4S)-1-[(2S)-2-氨基丁酰基]-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-基]丁烷-1-酮(93.05毫克,113.93微摩尔)溶于DMF(2.00毫升)中加入上述混合液中。混合液25度下继续搅拌1小时后,用乙酸乙酯(100毫升)和水(100毫升)稀释,有机相分离后,用饱和盐水洗(100毫升×3),45度减压浓缩,所得残渣通过硅胶薄层色谱纯化(石油醚/乙酸乙酯=1/1)得黄色的油状化合物叔丁基-N-[(1S)-2-[[(1S)-1-[(2R,4S)-2-[[2-[1-[[(2S,4S)-1-[(2S)-2-[[(2S)-2-[叔-丁氧羰基(甲基)氨基]丙酰基]氨基]丁酰基]-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羰基]丙基]氨基]-1-氨基-2-氧-乙基]-N-甲基-氨基甲酸酯(90.00毫克,79.14微摩尔,69.46%)。
MS(ESI)m/z:1012.2[M+H+]。
步骤G:将叔丁基-N-[(1S)-2-[[(1S)-1-[(2R,4S)-2-[[2-[1-[[(2S,4S)-1-[(2S)-2-[[(2S)-2-[叔-丁氧羰基(甲基)氨基]丙酰基]氨基]丁酰基]-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羰基]丙基]氨基]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(90.00毫克,79.14微摩尔)溶于二氯甲烷(1.00毫升),然后25度下加入三氟乙酸(1.54克,13.51毫摩尔,1.00毫升)。混合液25度下搅拌30分钟后,减压蒸馏,所得残渣通过制备HPLC纯化得到实施例13(40.00毫克,48.77微摩尔,61.63%)。
1HNMR(MeOD,400MHz):δ8.99-8.81(m,1H),8.75-8.51(m,1H),8.14-8.04(m,1H),7.93(dd,J=4.0,8.0Hz,1H),7.81(dd,J=4.0,8.0Hz,1H),7.54(td,J=4.0,8.0Hz,1H),7.43(dd,J=4.0,8.0Hz,1H),7.10(td,J=4.0,8.0Hz,1H),5.56-5.40(m,1H),5.39-5.28(m,1H),4.99-4.92(m,1H),4.83-4.76(m,1H),4.74-4.60(m,1H),4.58-4.45(m,2H),4.42-4.25(m,1H),4.23-3.75(m,6H),3.68-3.50(m,1H),3.25-3.08(m,1H),2.72-2.55(m,6H),2.27-1.98(m,4H),1.98-1.75(m,2H),1.53(dd,J=7.0,13.6Hz,7H),1.42-1.28(m,1H),1.10(t,J=7.3Hz,3H),0.96-0.76(m,3H)。
MS(ESI)m/z:812.5[M+H+]。
实施例14至17均可参照实施例13的制备反应流程制得。
实施例14
Figure PCTCN2017082227-appb-000115
1HNMR(MeOD,400MHz):δ8.10(dd,J=4.0,12.0Hz,1H),7.93(dd,J=4.0,12.0Hz,1H),7.82(dd,J=4.0,12.0Hz,1H),7.56(td,J=4.0,8.0Hz,1H),7.43(dd,J=4.0,12.0Hz,1H),7.11(td,J=4.0,8.0Hz,1H),5.47(d,J=12.0Hz,1H),5.33(d,J=12.0Hz,1H),4.91-4.77(m,2H),4.73-4.58(m,1H),4.52-4.46(m,1H),4.28-3.73(m,8H),3.70-3.59(m,1H),3.27-3.13(m,1H),2.66(d,J=16.0Hz,6H),2.24-1.88(m,4H),1.59-1.39(m,6H),1.38-1.25(m,1H),0.93-0.79(m,1H),0.77-0.12(m,8H)。
MS(ESI)m/z:836.4[M+H+]。
实施例15
Figure PCTCN2017082227-appb-000116
1HNMR(MeOD,400MHz):δ8.84(d,J=8.0Hz,1H),8.68(d,J=8.0Hz,1H),8.05(dd,J=4.0,8.0 Hz,1H),7.94(dd,J=4.0,8.0Hz,1H),7.80(dd,J=4.0,8.0Hz,1H),7.55(dt,J=2.4,8.0Hz,1H),7.42(dd,J=2.0,8.0Hz,1H),7.11(dt,J=2.0,8.0Hz,1H),5.57-5.28(m,2H),4.82-4.64(m,3H),4.60-4.47(m,2H),4.43-4.35(m,1H),4.22-3.85(m,6H),3.63(dd,J=4.0,16.0Hz,1H),3.21-3.11(m,1H),2.71-2.62(m,6H),2.25-1.90(m,5H),1.83-1.66(m,2H),1.52(dd,J=4.0,16.0Hz,6H),1.38-1.16(m,5H),1.03(t,J=8.0Hz,3H),0.93-0.89(m,3H)。
MS(ESI)m/z:840.3[M+H+]。
实施例16
Figure PCTCN2017082227-appb-000117
1HNMR(MeOD,400MHz):δ9.01-8.78(m,1H),8.74-8.54(m,1H),8.14-8.11(m,1H),8.06-7.78(m,2H),7.57-7.52(m,1H),7.45(d,J=8.0Hz,1H),7.12-7.08(m,1H),5.56-5.42(m,1H),5.40-5.28(m,1H),5.18-4.95(m,3H),4.79-4.64(m,1H),4.61-4.27(m,3H),4.27-3.84(m,6H),3.70-3.50(m,1H),2.67(d,J=16.0Hz,6H),2.32-1.78(m,6H),1.64-1.36(m,6H),1.11(d,J=4.0Hz,6H),0.91(d,J=4.0,6H)。
MS(ESI)m/z:840.5[M+H+]。
实施例17
Figure PCTCN2017082227-appb-000118
1HNMR(MeOD,400MHz):δ8.86-8.68(m,1H),8.14(d,J=4.0Hz,1H),7.93(dd,J=4.0,8.0Hz,1H),7.86(d,J=8.0Hz,1H),7.58(t,J=8.0Hz,1H),7.45(d,J=8.0Hz,1H),7.13(t,J=8.0Hz,1H),5.53-5.33(m,2H),4.80-4.60(m,4H),4.58-4.48(m,2H),4.26-4.07(m,3H),4.06-3.88(m,3H),3.63-3.51(m,1H),3.29-3.09(m,1H),2.95-2.79(m,1H),2.69(d,J=12.0Hz,6H),2.62-2.50(m,1H),1.90(d,J=4.0Hz,16.0H),1.52(dd,J=4.0,12.0Hz,6H)。
MS(ESI)m/z:864.3[M+H+]
实施例18
Figure PCTCN2017082227-appb-000119
反应流程:实施例18的制备
Figure PCTCN2017082227-appb-000120
步骤A:将苄基(2R,4S)-2-[[2-[1-[[(2S,4S)-1-叔-丁氧羰基-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羧酸酯(400.00毫克,510.10微摩尔)溶于乙酸乙酯(1.00毫升),然后于20度加入HCl/EtOAc(4摩尔/升,1.00毫升)。混合液于20度搅拌30分钟后,减压蒸馏除去溶剂得棕色固体苄基(2R,4S)-4-氟-2-[[6-氟-2-[5-氟-1-[[(2S,4S)-4-氟吡咯烷-2-基]甲基]苯并咪唑-2-基]-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸酯(332.00毫克,粗品,盐酸盐)。
步骤B:将N-Boc-L缬氨酸(206.03毫克,948.30微摩尔)溶于DMF(2.00毫升),20度滴入N-甲基吗啉(239.80毫克,2.37毫摩尔,260.65微升)和HATU(396.63毫克,1.04毫摩尔)。混合液20度搅拌30分钟。然后将苄基(2R,4S)-4-氟-2-[[6-氟-2-[5-氟-1-[[(2S,4S)-4-氟吡咯烷-2-基]甲基]苯并咪唑-2-基]-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸酯(304.44毫克,粗品,盐酸盐)溶于DMF(1.00毫升)20度下 加入上述混合液,20度继续搅拌1小时。反应液用水(200毫升)和乙酸乙酯(200毫升)稀释,有机相分离后用饱和食盐水(100毫升×3)洗,减压浓缩。所得残渣通过硅胶柱色谱纯化(石油醚/乙酸乙酯=3/1至2/3)得苄基(2R,4S)-2-[[2-[1-[[(2S,4S)-1-[(2S)-2-(叔-丁氧羰氨基)-3-甲基-丁酰基]-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羧酸酯(400.00毫克,455.73微摩尔,96.12%)。
1HNMR(DMSO,400MHz):δ12.05-11.67(m,1H),8.08-7.47(m,1H),7.46-7.36(m,1H),7.27(br.s.,5H),7.01(br.s.,3H),6.53(d,J=9.2Hz,1H),5.28-4.90(m,3H),4.62-4.09(m,2H),3.98-3.79(m,2H),3.78-3.68(m,2H),3.66-3.47(m,2H),2.96-2.80(m,1H),2.70(s,7H),1.92(s,4H),1.39(s,9H),0.85-0.80(m,6H)。
MS(ESI)m/z:805.5[M+H+]。
步骤C:将苄基(2R,4S)-2-[[2-[1-[[(2S,4S)-1-[(2S)-2-(叔-丁氧羰氨基)-3-甲基-丁酰基]-4-氟-吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羧酸酯(400.00毫克,455.73微摩尔)溶于乙酸乙酯(30.00毫克)和甲醇(5.00毫克),25度氮气保护下加入钯/碳(100.00毫克,10%),所得悬浊液真空下脱气并用氢气置换几次后,25度氢气氛下(40psi)搅拌3小时。混合液用硅藻土过滤,甲醇(约200毫升)洗,滤液减压浓缩得叔丁基N-[(1S)-1-[(2S,4S)-4-氟-2-[[5-氟-2-[6-氟-3-[[(2R,4S)-4-氟吡咯烷-2-基]甲基]-1H-吲哚-2-基]苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(320.00毫克,395.98微摩尔,86.89%)。
MS(ESI)m/z:671.5[M+H+]。
步骤D:将N-Boc-L正丁氨酸(160.95毫克,791.96微摩尔)溶于DMF(1.00毫升),20度下滴入N-甲基吗啉(120.16毫克,1.19毫摩尔,130.61微升)和HATU(376.41毫克,989.95微摩尔)。混合液20度搅拌30分钟后,将叔-丁基N-[(1S)-1-[(2S,4S)-4-氟-2-[[5-氟-2-[6-氟-3-[[(2R,4S)-4-氟吡咯烷-2-基]甲基]-1H-吲哚-2-基]苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(320.00毫克,395.98微摩尔)溶DMF(1.00毫升)后20度加入上述混合液,20度继续搅拌1小时。混合液用水(150毫升)和乙酸乙酯(200毫升)稀释,有机相分离后用饱和食盐水洗(100毫升×3),45度减压浓缩,所得残渣通过硅胶柱色谱纯化(石油醚/乙酸乙酯=3/1至2/1)得到叔-丁基N-[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-(叔-丁氧羰氨基)丁酰基]-4-氟-吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]-4-氟-吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(320.00毫克,321.51微摩尔,81.19%)
MS(ESI)m/z:856.4[M+H+]。
步骤E:将叔丁基N-[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-(叔-丁氧羰氨基)丁酰基]-4-氟-吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]-4-氟-吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(320.00毫克,321.51微摩尔)溶于1,4-二氧六环(2.00毫升),然后25度下滴加HCl/1,4-二氧六环(4摩尔/升,2.00毫升)。混合液25度下搅拌30分钟。减压蒸馏得到2S)-2-氨基-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-氨基丁酰基]-4-氟-吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]-4-氟-吡咯烷-1-基]-3-甲基-丁烷-1-酮(260.00毫克,321.14微摩尔,99.89%,盐酸盐)
MS(ESI)m/z:656.5[M+H+]。
步骤F:将N-Boc-N-甲基-L-丙氨酸(228.43毫克,1.12毫摩尔)溶于DMF(1.00毫升),25度下滴入N-甲基吗啉(194.90毫克,1.93毫摩尔,211.85微升)和HATU(451.80毫克,1.19毫摩尔),混合液25度下搅拌30分钟。然后将(2S)-2-氨基-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-氨基丁酰基]-4-氟-吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]-4-氟-吡咯烷-1-基]-3-甲基-丁烷-1-酮(260.00毫克,321.14微摩尔,盐酸盐)溶入DMF(1.00毫升)后加入上述混合液。混合液25度继续搅拌1小时后,用水(150毫升)和乙酸乙酯(200毫升)稀释,有机相分离后用饱和食盐水洗(100毫升×3),45度下减压蒸馏,所得残渣通过硅胶柱色谱纯化(石油醚/乙酸乙酯=3/1至2/3)得黄色固体叔-丁基-N-[(1S)-2-[[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-[[(2S)-2-[叔-丁氧羰基(甲基)氨基]丙酰基]氨基]丁酰基]-4-氟-吡咯烷-2-]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]-4-氟-吡咯烷-1-羰基]-2-甲基-丙基]氨基]-1-甲基-2-氧-乙基]-氮-甲基-氨基甲酸酯(260.00毫克,222.97微摩尔,69.43%)。
MS(ESI)m/z:1026.3[M+H+]。
步骤G:将叔丁基-N-[(1S)-2-[[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-[[(2S)-2-[叔-丁氧羰基(甲基)氨基]丙酰基]氨基]丁酰基]-4-氟-吡咯烷-2-]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]-4-氟-吡咯烷-1-羰基]-2-甲基-丙基]氨基]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(260.00毫克,222.97微摩尔)溶于二氯甲烷(1.00毫升),然后25度下滴入三氟乙酸(1.54克,13.51毫摩尔,1.00毫升)。混合液25度搅拌30分钟后,减压蒸馏除去溶剂,所得残渣通过制备HPLC纯化得实施例18(85.00毫克,94.00微摩尔,42.16%,盐酸盐)。
1HNMR(MeOD,400MHz):δ8.30-8.05(m,1H),7.98-7.76(m,2H),7.62-7.36(m,2H),7.18-6.99(m,1H),5.53-5.43(m,1H),5.38-5.30(m,1H),5.08-4.96(m,3H),4.76-4.67(m,1H),4.64-4.44(m,2H),4.41-4.25(m,1H),4.23-3.86(m,6H),3.70-3.52(m,1H),2.67(d,J=12.8Hz,6H),2.29-1.73(m,7H),1.60-1.42(m,6H),1.15-1.03(m,3H),0.98-0.85(m,6H)。
MS(ESI)m/z:826.5[M+H+]。
实施例19至36均可参照实施例18的制备反应流程制得。
实施例19
Figure PCTCN2017082227-appb-000121
1HNMR(MeOD,400MHz):δ9.01-8.87(m,1H),8.62-8.46(m,1H),8.15(dd,J=4.0,12.0Hz,1H),7.92(dd,J=4.0,8.0Hz,1H),7.87(dd,J=4.0,8.0Hz,1H),7.56(td,J=4.0,8.0Hz,1H),7.48(dd,J=4.0,12.0Hz,1H),7.12(td,J=4.0,8.0Hz,1H),5.52(d,J=16.0,1H),5.38(d,J=16.0,1H),5.08-4.97(m,1H), 4.76-4.63(m,1H),4.61-4.50(m,2H),4.48-4.42(m,1H),4.24-3.94(m,6H),3.70-3.55(m,1H),2.74-2.63(m,6H),2.32-1.78(m,7H),1.58(d,J=8.0Hz,3H),1.46(d,J=8.0Hz,3H),1.13(t,J=8.0Hz,3H),1.01(s,9H),0.98-0.93(m,1H)。
MS(ESI)m/z:840.3[M+H+]。
实施例20
Figure PCTCN2017082227-appb-000122
1HNMR(MeOD,400MHz):δ8.97-8.84(m,1H),8.74-8.59(m,1H),8.12(dd,J=4.0,8.0Hz,1H)),7.93(dd,J=4.0,12.0Hz,1H),7.85(dd,J=4.0,12.0Hz,1H),7.57(td,J=4.0,8.0Hz,1H),7.46(dd,J=4.0,12.0Hz,1H),7.12(td,J=4.0,8.0Hz,1H),5.50(d,J=16.0,1H),5.37(d,J=16.0,1H),5.09-5.00(m,1H),4.77-4.62(m,1H),4.61-4.46(m,2H),4.39-4.31(m,1H),4.24-3.91(m,6H),3.69-3.56(m,1H),3.29-3.23(m,1H),2.69(d,J=16.0Hz,6H),2.27-1.82(m,6H),1.80-1.62(m,5H),1.60-1.44(m,7H),1.43-1.33(m,1H),1.31-0.96(m,9H)。
MS(ESI)m/z:866.3[M+H+]。
实施例21
Figure PCTCN2017082227-appb-000123
1HNMR(MeOD,400MHz):δ8.97-8.89(m,1H),8.86-8.79(m,1H),8.10(dd,J=4.0,8.0Hz,1H),7.94(dd,J=4.0,8.0Hz,1H),7.84(dd,J=4.0,8.0Hz,1H),7.57(td,J=4.0,8.0Hz,1H),7.43(dd,J=4.0,8.0Hz,1H),7.12(td,J=4.0,8.0Hz,1H),5.52(d,J=16.0,1H),5.37(d,J=16.0,1H),5.02-4.95(m,1H),4.73-4.64(m,1H),4.59-4.48(m,1H),4.47-4.39(m,1H),4.27-3.97(m,5H),3.95-3.89(m.,2H),3.71-3.62(m,1H),3.30-3.20(m,1H),2.68(d,J=16.0Hz,6H),2.28-1.99(m,5H),1.53-1.50(m,6H),1.13(d,J=4.0Hz,6H),1.03-0.82(m,2H),0.64-0.52(m,1H),0.48-0.30(m,3H).
MS(ESI)m/z:838.4[M+H+]。
实施例22
Figure PCTCN2017082227-appb-000124
1HNMR(MeOD,400MHz):δ8.93-8.81(m,1H),8.69-8.58(m,1H),8.07(dd,J=4.0,8.0Hz,1H),7.92(dd,J=4.0,8.0Hz,1H),7.87(dd,J=4.0,8.0Hz,1H),7.54(td,J=4.0,8.0Hz,1H),7.42(dd,J=4.0,8.0Hz,1H),7.10(td,J=4.0,8.0Hz,1H),5.49(d,J=16.0,1H),5.36(d,J=16.0,1H),5.04-4.95(m,1H),4.71-4.46(m,1H),4.23-3.94(m,5H),3.92-3.82(m,2H),3.66-3.54(m,1H),2.66(d,J=16Hz,6H),2.25-1.92(m,4H),1.59-1.52(m,1H),1.52-1.43(m,6H),1.17(s,9H),1.05-0.98(m,1H),0.92-0.82(m,1H),0.60-0.50(m,1H),0.41-0.27(m,3H).
MS(ESI)m/z:852.4[M+H+]。
实施例23
Figure PCTCN2017082227-appb-000125
1HNMR(MeOD,400MHz):δ8.99-8.87(m,1H),8.86-8.75(m,1H),8.10(dd,J=4.0,12.0Hz,1H),7.92(dd,J=4.0,8.0Hz,1H),7.83(dd,J=4.0,8.0Hz,1H),7.55(td,J=4.0,12.0Hz,1H),7.42(dd,J=4.0,8.0Hz,1H),7.10(td,J=4.0,8.0Hz,1H),5.48(d,J=16.0,1H),5.34(d,J=16.0,1H),4.75-4.62(m,1H),4.57-4.46(m,1H),4.45-4.39(m,1H),4.30-3.79(m,8H),3.70-3.59(m,1H),3.29-3.19(m,1H),2.66(d,J=16Hz,6H),2.25-1.63(m,11H),1.51-1.48(m,7H),1.34-1.15(m,5H),0.99-0.82(m,1H),0.61-0.48(m,1H),0.46-0.27(m,3H).
MS(ESI)m/z:878.4[M+H+]。
实施例24
Figure PCTCN2017082227-appb-000126
1HNMR(MeOD,400MHz):δ8.09(d,J=8.0Hz,1H),7.94(dd,J=4.0,8.0Hz,1H),7.85(d,J=8.0Hz,1H),7.56(t,J=8.0Hz,1H),7.44(d,J=8.0Hz,1H),7.12(t,J=8.0Hz,1H),5.49(d,J=20.0Hz,1H),5.36(d,J=16.0Hz,1H),4.75-4.65(m,1H),4.63-4.33(m,4H),4.01(m,7H),3.66(d,J=8.0Hz,1H),3.29-3.13(m,1H),2.68(d,J=12.0Hz,6H),2.33-1.92(m,6H),1.52(d,J=8.0Hz,6H),1.43-1.33(m,1H)1.13(d,J=4.0Hz,6H),0.92(t,J=4.0Hz,3H)。
MS(ESI)m/z:826.4[M+H+]。
实施例25
Figure PCTCN2017082227-appb-000127
1HNMR(MeOD,400MHz):δ8.93-8.49(m,1H),7.92(d,J=8.0Hz,1H),7.76(dd,J=4.0,8.0Hz,1H),7.66(d,J=8.0Hz,1H),7.39(t,J=8.0Hz,1H),7.27(d,J=8.0Hz,1H),6.94(t,J=8.0Hz,1H),5.37-5.12(m,2H),4.59-4.47(m,1H),4.41-4.27(m,1H),4.24-4.15(m,1H),4.08-3.72(m,7H),3.48(d,J=12.0Hz,1H),3.06(br.s.,1H),2.51(d,J=12.0Hz,6H),2.07-1.85(m,4H),1.36(dd,J=4.0,12.0Hz,10H),0.73(t,J=8.0Hz,3H),0.61-0.14(m,4H)。
MS(ESI)m/z:824.3[M+H+]。
实施例26
Figure PCTCN2017082227-appb-000128
1HNMR(MeOD,400MHz):δ8.56(br.s.,1H),7.97(d,J=8.0Hz,1H),7.86-7.76(m,2H),7.44(br.s.,1H),7.34(d,J=8.0Hz,1H),7.01(t,J=8.0Hz,1H),5.40(d,J=8.0Hz,1H),5.26(d,J=12.0Hz, 1H),,4.49(br.s.,4H),4.31-4.16(m,1H),4.14-3.73(m,6H),3.51(d,J=12.0Hz,1H),2.56(d,J=12.0Hz,6H),2.16-1.84(m,4H),1.40(d,J=8.0Hz,9H),1.07(s,8H),0.94-0.87(m,1H),0.77(t,J=8.0Hz,3H)。
MS(ESI)m/z:840.3[M+H+]。
实施例27
Figure PCTCN2017082227-appb-000129
1HNMR(MeOD,400MHz):δ8.88-8.72(m,1H),8.10(dd,J=4.0,8.0Hz,1H),7.95(dd,J=4.0,8.0Hz,1H),7.87-7.82(m,1H),7.61-7.52(m,1H),7.44(dd,J=4.0,12.0Hz,1H),7.12(d,J=4.0Hz,1H),5.54-5.30(m,2H),5.01-4.92(m,1H),4.85-4.69(m,2H),4.42(td,J=4.0,4.0Hz,3H),4.01(br.s.,6H),3.66(d,J=12.0Hz,1H),3.29-3.18(m,1H),2.70-2.64(m,6H),2.19-1.70(m,10H),1.52(dd,J=8.0,12.0Hz,6H),1.47-1.18(m,7H),0.93ppm(t,J=8.0Hz,3H)。
MS(ESI)m/z:866.4[M+H+]。
实施例28
Figure PCTCN2017082227-appb-000130
1HNMR(MeOD,400MHz):δ8.59-8.44(d,J=8.0Hz,1H),8.04-7.88(m,1H),7.73(dd,J=4.0,8.0Hz,1H),7.64(d,J=8.0Hz,1H),7.40-7.32(m,1H),7.26(d,J=8.0Hz,1H),6.91(t,J=8.0Hz,1H),5.35-5.08(m,2H),4.88-4.76(m,1H),4.61-4.43(m,1H),4.39-4.13(m,2H),4.06-3.67(m,8H),3.44(d,J=12.0Hz,1H),3.09-3.02(m,1H),2.51-2.45(m,6H),2.02-1.73(m,5H),1.42-1.26(m,6H),1.21-1.05(m,1H),0.81-0.62(m,6H),0.60-0.37(m,3H),0.36-0.25(m,1H)。
MS(ESI)m/z:838.3[M+H+]。
实施例29
Figure PCTCN2017082227-appb-000131
1HNMR(MeOD,400MHz):δ8.04(dd,J=4.0,8.0Hz,1H),7.83(dd,J=4.0,8.0Hz,1H),7.69(d,J=4.0Hz,1H),7.40(t,J=8.0Hz,1H),7.35-7.28(m,1H),6.99(t,J=8.0Hz,1H),5.33(br.s.,1H),5.20(br.s.,1H),4.35(m,4H),4.18-4.09(m,1H),4.06-3.81(m,7H),3.60-3.45(m,1H),3.17-3.07(m,1H),2.57(d,J=20.0Hz,6H),2.09-1.85(m,4H),1.94(s,3H),1.45(d,J=8.0Hz,3H),1.29-1.18(m,1H),0.92(s,9H),0.69-0.49(m,3H),0.45-0.36(m,1H)。
MS(ESI)m/z:852.4[M+H+]。
实施例30
Figure PCTCN2017082227-appb-000132
1HNMR(MeOD,400MHz):δ8.58(d,J=8.0Hz,1H),8.01(d,J=4.0Hz,1H),7.82(dd,J=4.0,8.0Hz,1H),7.73(d,J=8.0Hz,1H),7.45(t,J=8.0Hz,1H),7.35(d,J=8.0Hz,1H),7.01(t,J=8.0Hz,1H),5.47-5.20(m,2H),4.87(br.s.,2H),4.66-4.34(m,2H),4.29-3.78(m,8H),3.58-3.44(m,1H),3.19-3.11(m,1H),2.57(d,J=20.0Hz,6H),2.18-1.77(m,4H),1.69-1.48(m,5H),1.47-1.35(m,6H),1.34-1.15(m,3H),1.14-0.87(m,4H),0.56(br.s.,4H)。
MS(ESI)m/z:878.4[M+H+]。
实施例31
Figure PCTCN2017082227-appb-000133
1HNMR(MeOD,400MHz):δ8.17-8.06(m,1H),7.94(dd,J=4.0,8.0Hz,1H),7.84(d,J=4.0Hz, 1H),7.57(t,J=8.0Hz,1H),7.44(d,J=8.0Hz,1H),7.16-7.08(m,1H),5.49(br.s.,1H),5.36(d,J=4.0Hz,1H),4.78-4.63(m,2H),4.53(br.s.,2H),4.22-3.85(m,8H),3.65(d,J=12.0Hz,1H),3.29-3.10(m,1H),2.94-2.76(m,1H),2.68(d,J=16.0Hz,6H),2.25-2.03(m,4H),2.03-1.75(m,3H),1.53(dd,J=4.0,16.0Hz,6H),1.11(t,J=8.0Hz,3H),0.94-0.83(m,1H),0.62-0.53(m,1H),0.43(q,J=8.0Hz,3H)。
MS(ESI)m/z:824.4[M+H+]。
实施例32
Figure PCTCN2017082227-appb-000134
1HNMR(MeOD,400MHz):δ8.90(d,J=8.0Hz,1H),8.72(d,J=8.0Hz,1H),8.15(dd,J=4.0,12.0Hz,1H),7.93(dd,J=4.0,8.0Hz,1H),7.85(d,J=8.0Hz,1H),7.61-7.52(m,1H),7.49-7.42(m,1H),7.12(dt,J=8.0,12.0Hz,1H),5.54-5.32(m,2H),4.79(m,2H),4.65-4.48(m,3H),4.40(t,J=8.0Hz,1H),4.24-3.90(m,7H),3.62(d,J=16.0Hz,1H),2.69(d,J=12.0Hz,6H),2.19-1.97(m,5H),1.84-1.76(m,2H),1.57-1.48(m,8H),1.03(t,J=8.0Hz,3H),0.97(d,J=8.0Hz,3H),0.92(d,J=8.0Hz,3H)。
MS(ESI)m/z:840.2[M+H+]。
实施例33
Figure PCTCN2017082227-appb-000135
1HNMR(MeOD,400MHz):δ8.94(d,J=8.0Hz,1H),8.58(d,J=8.0Hz,1H),8.16(dd,J=4.0,8.0Hz,1H),7.93(dd,J=8.0,12.0Hz,1H),7.87(d,J=8.0Hz,1H),7.55(t,J=12.0Hz,1H),7.48(d,J=8.0Hz,1H),7.12(t,J=8.0Hz,1H),5.58-5.31(m,2H),4.78-4.52(m,5H),4.47(d,J=8.0Hz,1H),4.28-3.89(m,7H),3.62(d,J=12.0Hz,1H),2.69(d,J=20.0Hz,6H),2.28-1.95(m,4H),1.83(d,J=8.0Hz,2H),1.67-1.40(m,8H),1.12-0.91(m,12H)。
MS(ESI)m/z:854.3[M+H+]。
实施例34
Figure PCTCN2017082227-appb-000136
1HNMR(MeOD,400MHz):δ8.13-8.05(m,1H),7.95(dd,J=4.0,8.0Hz,1H),7.75(d,J=4.0Hz,1H),7.49(t,J=8.0Hz,1H),7.39(d,J=12.0Hz,1H),7.09(t,J=12.0Hz,1H),5.46-5.26(m,2H),4.70(m,J=6.8Hz,3H),4.58(br s,1H),4.36(br d,J=8.0Hz,1H),4.11-3.84(m,7H),3.60(br d,J=13.2Hz,1H),3.22-3.12(m,1H),2.68(d,J=16.0Hz,6H),2.17-1.92(m,6H),1.83-1.66(m,9H),1.59-1.46(m,11H),1.22(d,J=8.0Hz,4H),1.03(t,J=8.0Hz,3H)。
MS(ESI)m/z:880.4[M+H+]。
实施例35
Figure PCTCN2017082227-appb-000137
1HNMR(MeOD,400MHz):δ8.64(d,J=5.77Hz,2H),8.05(dd,J=3.95,8.97Hz,1H),7.94(dd,J=5.14,8.91Hz,1H),7.87(d,J=7.28Hz,1H),7.56(t,J=8.41Hz,1H),7.44(d,J=7.78Hz,1H),7.12(t,J=8.16Hz,1H),5.44-5.58(m,1H),5.31-5.43(m,1H),4.57-4.68(m,3H),4.53(br.s.,1H),4.37(br.s.,1H),3.83-4.24(m,6H),3.62(d,J=13.55Hz,1H),3.18-3.30(m,2H),2.67(d,J=17.57Hz,6H),1.94-2.27(m,4H),1.49(d,J=6.65Hz,6H),1.30(d,J=7.15Hz,3H),1.18(s,9H),0.99(s,1H),0.90(t,J=5.52Hz,3H)。
MS(ESI)m/z:854.4[M+H+]。
实施例36
Figure PCTCN2017082227-appb-000138
1HNMR(MeOD,400MHz):δ8.79(d,J=7.28Hz,1H),8.69(d,J=6.65Hz,1H),8.06(dd,J=3.64,8.91Hz,1H),7.93(dd,J=5.08,8.85Hz,1H),7.83(d,J=6.27Hz,1H),7.55(t,J=8.47Hz,1H),7.42(d,J=7.53Hz,1H),7.10(t,J=9.10Hz,1H),5.27-5.55(m,2H),4.50(br.s.,1H),4.40(d,J=7.28Hz,2H),4.01-4.25(m,3H),3.82-4.01(m,4H),3.64(d,J=13.93Hz,1H),3.11-3.23(m,1H),2.65(d,J=11.92Hz,6H),2.00-2.28(m,4H),1.79(br.s.,7H),1.48(t,J=6.21Hz,6H),1.09-1.40(m,10H),0.90(t,J=6.09Hz,3H)。
MS(ESI)m/z:880.4[M+H+]。
实施例37
Figure PCTCN2017082227-appb-000139
反应流程:实施例37的制备
Figure PCTCN2017082227-appb-000140
Figure PCTCN2017082227-appb-000141
步骤A:往溶解有(S)-苄基-2-((6-氟-1氢-3-吲哚)甲基)吡咯烷-1-羧酸酯(5克,14.19毫摩尔)的30毫升二氯甲烷溶液中加入DMAP(34.67毫克,283.77微摩尔),然后滴加溶解在20毫升二氯甲烷的BOC酸酐(3.25克,14.90毫摩尔)。加完后,反应液在10~20摄氏度下搅拌16个小时。反应完成后,反应液直接减压浓缩。得到的残余物经柱层析纯化(洗脱剂:石油醚/乙酸乙酯=30:1到20:1)得到(S)-叔丁基3-((1-((苄氧基)酰基)2-吡咯烷)甲基)-6-氟-1H-吲哚-1-羧酸酯(6.5克,97.18%)。
1HNMR(CDCl3,400MHz):δ7.80-7.60(m,1H),7.40-7.20(m,6H),7.08-6.84(m,1H),6.61(t,J=8.2Hz,1H),5.13(s,2H),4.17-3.98(m,1H),3.47-3.29(m,2H),2.57-2.40(m,1H),1.85-1.65(m,4H),1.59(s,9H)。
MS(ESI)m/z:475.2[M+Na+]。
步骤B:在-70摄氏度氮气氛围下,往溶解有S)-叔丁基3-((1-((苄氧基)酰基)2-吡咯烷)甲基)-6-氟-1H-吲哚-1-羧酸盐(6.5克,13.79毫摩尔)的120毫升四氢呋喃溶液中滴加LDA(2摩尔/升,13.79毫升),滴加完后在这个温度下搅拌15分钟。然后加入干冰(13.79毫摩尔)。搅拌下,反应液经过105分钟升到20摄氏度后,用50毫升水淬灭,再用100毫升的乙酸乙酯萃取。有机相用100毫升饱和食盐水洗涤,无水硫酸钠干燥,过滤之后,滤液经过减压浓缩得到残余物。残余物经过柱层析(洗脱剂:二氯甲烷/甲醇=100:1到33:1)得到化合物(S)-3-((1-((苄氧基)羰基)2-吡咯烷)甲基)-1-(叔丁氧基羰基)-6-氟-1H-吲哚-2-羧酸(3.9克,44.09%)。
1HNMR(DMSO,400MHz):δ13.56(br.s.,1H),7.51-7.19(m,8H),5.21-4.97(m,2H),4.11-3.98(m,1H),3.00-2.79(m,1H),1.89-1.61(m,5H),1.56(d,J=6.1Hz,9H)。
MS(ESI)m/z:497.3[M+H+]。
步骤C:在5-20摄氏度下,往溶解有(S)-3-((1-((苄氧基)羰基)2-吡咯烷)甲基)-1-(叔丁氧基羰基)-6-氟-1H-吲哚-2-羧酸(2.5克,5.03毫摩尔)的20毫升二氯甲烷溶液中加入三氟乙酸(135.06毫摩尔,10毫升)。得到的反应液在5到20摄氏度下搅拌16个小时。反应完后,反应减压浓缩除去二氯甲烷及大部分三氟乙酸。残余物用二氯甲烷稀释,再用10%的氢氧化钠水溶液洗涤。有机相经减压浓缩得到的粗产物(S)-3-((1-((苄氧基)羰基)2-吡咯烷)甲基)-1-(叔丁氧羰基)-6-氟-1H-吲哚-2-羧酸(1.5克,75.23%),直接用于下一步。
MS(ESI)m/z:397.3[M+H+]。
步骤D:往溶解有(S)-3-((1-((苄氧基)羰基)2-吡咯烷)甲基)-1-(叔丁氧羰基)-6-氟-1H-吲哚-2羧酸(1.5克,3.78毫摩尔)的15毫升二氯甲烷溶液中加入吡啶(61.95毫摩尔,5毫升),叔丁基(2S,4S)-2-[(2-胺基-4-氟-苯胺基)甲基]-4-氟-1吡咯烷-羧酸酯(1.11克,3.40毫摩尔)和EDCI(1.09克,5.67毫摩尔)。反应液在10到25摄氏度下搅拌反应16个小时。反应完成后,往反应液中加入1摩尔/升的盐酸溶液50毫升淬灭反应,再用50毫升二氯甲烷萃取。分液,有机相经50毫升饱和食盐水洗涤,无水硫酸钠干燥,过滤后滤液减压浓缩得到粗产物。粗产品经过柱层析(洗脱剂,石油醚/乙酸乙酯10:1到5:1)得到产物(2S,4S)-叔丁基2-(((2-(3-(((S)-1-((丁氧基)羰基)2-吡咯烷)甲基)-6-氟-1H-吲哚-2-甲酰胺)-4-氟苯基)胺基)甲基)-4-氟吡咯烷-1-羧酸盐(800.00毫克,20.39%)。
MS(ESI)m/z:706.2,[M+H+]。
步骤E:将(2S,4S)-叔丁基2-(((2-(3-(((S)-1-((丁氧基)羰基)2-吡咯烷)甲基)-6-氟-1H-吲哚-2-甲酰胺)-4-氟苯基)胺基)甲基)-4-氟吡咯烷-1-羧酸盐(500毫克,708.46微摩尔)溶于5毫升醋酸溶液,然后再85摄氏度下搅拌4个小时。待反应完成后,反应液减压浓缩除去溶剂。粗产品经过柱层析(洗脱剂,乙酸乙酯/石油醚10%到20%)得到产物(2S,4S)-叔丁基2-((2-(3-(((S)-1-((苄氧基)羰基)2-吡咯烷)甲基)-6-氟-1氢-2吲哚)-5-氟-1H-1-苯并咪唑)甲基)-4-氟吡咯烷-1-羧酸(300.00毫克,56.03%)。
MS(ESI)m/z:688.1[M+H+]。
步骤F:在氮气氛围下往溶解有(2S,4S)-叔丁基2-((2-(3-(((S)-1-((苄氧基)羰基)2-吡咯烷)甲基)-6-氟-1氢-2吲哚)-5-氟-1H-1-苯并咪唑)甲基)-4-氟吡咯烷-1-羧酸化物(300.00毫克,436.21微摩尔)的5毫升甲醇溶液中加入0.1克钯碳(10%)。反应体系置换3次氢气成氢氛围后,在30到35摄氏度氢气(15psi)氛围下搅拌16个小时。反应液冷却到室温后经垫硅藻土过滤,滤液减压浓缩得到粗产物(2S,4S)-叔丁基4-氟-2-((5-氟-2-(6-氟-3-((S)-2-吡咯烷)甲基)-1H-2-吲哚)-1H-1-苯并咪唑)甲基)吡咯烷-1-羧酸盐(190.00毫克,322.60微摩尔,73.96%)。
MS(ESI)m/z:554.3[M+H+]。
步骤G:在溶解了N-Boc-L正丁氨酸(122.13毫克,514.8微摩尔)的1毫升DMF溶液中加入HATU(195.74毫克,514.8微摩尔)和N-甲基吗啉(1.03摩尔,113.20微升)。得到的反应液在10到20摄氏度下搅拌15分钟。然后加入溶解于2毫升DMF的(2S,4S)-叔丁基4-氟-2-((5-氟-2-(6-氟-3-((S)-2-吡咯烷)甲基)-1H-2-吲哚)-1H-1-苯并咪唑)甲基)吡咯烷-1-羧酸盐(粗产物,190毫克,343.20微 摩尔)。然后反应液在10到20摄氏度下搅拌16个小时。反应完成后,加入30毫升水,用50毫升乙酸乙酯萃取。分液后有机相用50毫升饱和食盐水洗涤,无水硫酸钠干燥,再过滤,滤液减压浓缩得到粗产物。(2S,4S)-叔丁基2-((2-(3-(((S)-1-((S)-2-(((苄氧基)羰基)胺基)丁氧基)2-吡咯烷)甲基)-6-氟-1H-2-吲哚)-5-氟-1H-1-苯并咪唑)甲基)-4-氟吡咯烷-1-羧酸酯(300.00毫克)。
MS(ESI)m/z:773.2[M+H+]。
步骤H:往2S,4S)-叔丁基2-((2-(3-(((S)-1-((S)-2-(((苄氧基)羰基)胺基)丁氧基)2-吡咯烷)甲基)-6-氟-1H-2-吲哚)-5-氟-1H-1-苯并咪唑)甲基)-4-氟吡咯烷-1-羧酸酯(300毫克,粗品)的2毫升二氯甲烷溶液中加入TFA(13.15毫摩尔,1毫升)。反应液在10到20摄氏度度下搅拌2个小时。反应完后,反应液减压浓缩除去溶剂和大部分三氟乙酸。粗产物苄基((S)-1-((S)-2-((6-氟-2-(5-氟-1-(((2S,4S)-4-氟-2-吡咯烷)甲基)-1H-2-苯并咪唑)-1H-3-吲哚)甲基)1-吡咯烷)-1-2-氧络丁基)氨基甲酸酯(500.00毫克,三氟乙酸盐)未经过进一步纯化直接用于下一步。
MS(ESI)m/z:673.3[M+H+]。
步骤I:在N-Boc-L缬氨酸(103.55毫克,476.64微摩尔)的1毫升DMF溶液中加入NMM(953.28微摩尔,104.80微升)和HATU(181.23毫克,476.64微摩尔)。反应液在10到20摄氏度下搅拌15分钟后加入苄基((S)-1-((S)-2-((6-氟-2-(5-氟-1-(((2S,4S)-4-氟-2-吡咯烷)甲基)-1H-2-苯并咪唑)-1H-3-吲哚)甲基)1-吡咯烷)-1-2-氧络丁基)氨基甲酸酯(250.00毫克,317.76微摩尔,粗品,三氟乙酸盐)的1毫升DMF溶液。然后反应液在10到20摄氏度下反应4小时。往反应体系中加入30毫升水,再用50毫升乙酸乙酯萃取,分液。有机相用50毫升饱和食盐水洗涤后用无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产物。粗产物经过薄层色谱制备(展开剂,石油醚/乙酸乙酯=1:1)得到产物叔丁基N-[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2S)-1-[(2S)-2-(苄氧基羰基胺)叔丁氧基]2-吡咯烷]methyl]-6-氟-1H-2-吲哚l]-5-氟-1-苯并咪唑]甲基]-4-氟-吡咯烷-1-羰基]-2-甲基-丙丙基]氨基甲酸酯(200.00毫克,粗产物)。
MS(ESI)m/z:872.4[M+H+]。
步骤J:往叔丁基N-[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2S)-1-[(2S)-2-(苄氧基羰基胺)叔丁氧基]2-吡咯烷]methyl]-6-氟-1H-2-吲哚l]-5-氟-1-苯并咪唑]甲基]-4-氟-吡咯烷-1-羰基]-2-甲基-丙丙基]羧酸盐(200.00毫克,229.36微摩尔,粗产物)的1毫升二氯甲烷溶液中加入HBr/AcOH(229.36微摩尔,1.00毫升).反应液在10到20摄氏度下搅拌1个小时之后减压浓缩除去溶剂。粗产物用30毫升水稀释,再用30毫升甲基叔丁基醚萃取,分液。水相调节Ph到8~9,再用50毫升乙酸乙酯萃取,分液,有机相用50毫升饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产品(2S)-2-胺基-1-[(2S,4S)-2-[[2-[3-[[(2S)-1-[(2S)-2-叔丁氧胺基]2-吡咯烷]甲基]-6-氟-1H-2-吲哚]-5-氟-1-苯并咪唑]甲基]-4-氟-1-吡咯烷]-3-甲基-1-丁酮(100.00毫克,粗品)。
MS(ESI)m/z:638.2,[M+H+]。
步骤K:往N-Bco-N-甲基-L-丙氨酸(95.60毫克,470.40微摩尔)的1毫升DMF溶液中加入N-甲基吗啉(103.43微升,940.80微摩尔)and HATU(178.86毫克,470.40微摩尔)。反应液在25摄氏度下搅拌15分钟。然后将(2S)-2-胺基-1-[(2S,4S)-2-[[2-[3-[[(2S)-1-[(2S)-2-叔丁氧胺基]2-吡咯烷]甲基]-6-氟-1H-2-吲哚]-5-氟-1-苯并咪唑]甲基]-4-氟-1-吡咯烷]-3-甲基-1-丁酮(粗品)溶于1毫升的DMF中加入 反应液。反应液在25摄氏度下搅拌16个小时后,加入20毫升水,再用60毫升乙酸乙酯萃取两次。合并的有机相用50毫升饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产品。粗产品经过薄层色谱纯化(展开剂,石油醚/乙酸乙酯=1:2)得到白色固体叔丁基N-[(1S)-2-[[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2S)-1-[(2S)-2-[[(2S)-2-[叔丁氧羰基(甲基)胺]丙酰基]胺]丁氧基]2-吡咯烷]甲基]-6-氟-1H-2-吲哚]-5-氟-1-苯并吡唑]甲基]-4-氟-吡咯烷-1-羰基]-2-甲基-丙基]胺]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(80.00毫克,78.56微摩尔,50.10%)。
MS(ESI)m/z:1008.5,[M+H+]。
步骤L:往叔丁基N-[(1S)-2-[[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2S)-1-[(2S)-2-[[(2S)-2-[叔丁氧羰基(甲基)胺]丙酰基]胺]丁氧基]2-吡咯烷]甲基]-6-氟-1H-2-吲哚]-5-氟-1-苯并吡唑]甲基]-4-氟-吡咯烷-1-羰基]-2-甲基-丙基]胺]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(80.00毫克,78.56微摩尔)的2毫升二氯甲烷溶液中加入1毫升三氟乙酸。在10到20摄氏度下反应了1个小时后,减压浓缩除去溶剂和三氟乙酸。得到的粗产品经过制备HPLC得到实施例37(38.22毫克,43.39微摩尔,54.68%,盐酸盐)。
1HNMR(MeOD,400MHz):δ12.34(s,1H),8.93(d,J=6.8Hz,1H),8.58(d,J=7.2Hz,1H),8.12(dd,J=3.6,9.0Hz,1H),7.99-7.90(m,2H),7.59-7.47(m,2H),7.16-7.07(m,1H),5.56-5.35(m,1H),5.07-5.01(m,1H),4.96(d,J=8.8Hz,1H),4.69(d,J=6.8Hz,2H),4.50-4.43(m,1H),4.35(br.s.,1H),4.20-3.97(m,4H),3.88(dd,J=7.8,16.2Hz,2H),3.52(d,J=13.9Hz,1H),3.26-3.15(m,1H),2.73(s,3H),2.67(s,3H),2.35(br.s.,1H),2.20-1.79(m,7H),1.60(d,J=6.8Hz,3H),1.47(d,J=6.6Hz,3H),1.12(t,J=7.2Hz,3H),1.03(s,9H).
MS(ESI)m/z:808.4,[M+H+].
实施实例38
Figure PCTCN2017082227-appb-000142
根据实施实例37的制备过程制备。
1HNMR(MeOD,400MHz):δ8.88(d,J=7.0Hz,1H),8.73(d,J=7.1Hz,1H),8.11(dd,J=3.9,9.1Hz,1H),7.99-7.86(m,2H),7.59-7.44(m,2H),7.11(dt,J=2.1,9.2Hz,1H),5.56-5.35(m,1H),5.10-4.99(m,1H),4.77(br.s.,1H),4.66(d,J=5.1Hz,1H),4.45-4.38(m,1H),4.33(br.s.,1H),4.19-3.94(m,4H),3.92-3.78(m,2H),3.53(d,J=14.2Hz,1H),3.24-3.12(m,1H),2.76-2.60(m,7H),2.31(br.s.,1H),2.24-1.76(m,9H),1.58(d,J=6.8Hz,3H),1.51(d,J=7.0Hz,3H),1.10(t,J=7.3Hz,3H),0.97(dd,J=6.7,19.2Hz,6H)。
MS(ESI)m/z:822.3[M+H+]。
实施实例39
Figure PCTCN2017082227-appb-000143
反应流程:实施例39的制备
Figure PCTCN2017082227-appb-000144
步骤A:将叔丁基-(2S)-2-[(2-氨基-4-氟-苯胺基)甲基]吡咯烷-1-羧酸酯(4.13克,13.36毫摩尔)溶于DMF(30.00毫升)和H2O(2.00毫升)的混合溶剂中,0度下加入苄基(2R,4S)-4-氟-2-[(6-氟-2-甲酰基-1H-吲哚-3-基)甲基]吡咯烷-1-羧酸酯(3.80克,6.68毫摩尔)。混合液搅拌30分钟后,0度下 加入Oxone(3.05克,20.03毫摩尔),混合液15度下继续搅拌16小时后,用乙酸乙酯(300毫升)和水(300毫升)稀释,有机相分离,饱和食盐水洗,无水硫酸钠干燥,过滤,滤液,减压浓缩。所得残渣通过硅胶柱色谱纯化(石油醚/乙酸乙酯=10/1至2/1)得苄基(2R,4S)-2-[[2-[1-[[(2S)-1-叔丁氧羰基吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羧酸酯(2.30克,2.71毫摩尔,40.55%)。
1H NMR(400MHz,DMSO-d6):δ11.52-11.78(m,1H),7.58-7.86(m,1H),7.45-7.54(m,1H),7.26-7.44(m,6H),7.11-7.26(m,2H),6.63-7.05(m,1H),4.90-5.21(m,4H),4.30-4.58(m,1H),3.89-4.26(m,3H),3.37-3.77(m,3H),2.64-3.15(m,3H),2.01-2.30(m,1H),2.00(s,2H),1.41-1.61(m,2H),1.13-1.40(m,9H)。
MS(ESI)m/z:688.3[M+H+]。
步骤B:将苄基(2R,4S)-2-[[2-[1-[[(2S)-1-叔-丁氧羰基吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羧酸酯(600.00毫克,706.65微摩尔)溶于1,4-二氧六环(6.00毫升),15度下滴加HCl/1,4-二氧六环(4摩尔/升,6.00毫升)。混合液15度搅拌1小时后,45度减压蒸馏除去溶剂得苄基(2R,4S)-4-氟-2-[[6-氟-2-[5-氟-1-[[(2S)-吡咯烷-2-基]甲基]苯并咪唑-2-基]-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸酯(540.00毫克,粗品,盐酸盐)。
步骤C:将N-Boc-L缬氨酸(140.99毫克,648.95微摩尔)溶于DMF(2.00毫升),15度加入N-甲基吗啉(131.28毫克,1.30毫摩尔,142.70微升)和HATU(263.20毫克,692.21微摩尔)。混合液15度搅拌30分钟,将苄基(2R,4S)-4-氟-2-[[6-氟-2-[5-氟-1-[[(2S)-吡咯烷-2-基]甲基]苯并咪唑-2-基]-1H-吲哚-3-基]甲基]吡咯烷-1-羧酸酯(270.00毫克,粗品,盐酸盐)溶于DMF(2.00毫升)后加入上述混合液,15度继续搅拌1小时后,混合液倒入水中(100毫升)析出固体,继续搅拌10分钟后,过滤,干燥后得苄基(2R,4S)-2-[[2-[1-[[(2S)-1-[(2S)-2-(叔-丁氧羰氨基)-3-甲基-丁酰基]吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羧酸酯(335.00毫克,357.61微摩尔,82.66%)
MS(ESI)m/z:787.3[M+H+]。
步骤D:将苄基(2R,4S)-2-[[2-[1-[[(2S)-1-[(2S)-2-(叔-丁氧羰氨基)-3-甲基-丁酰基]吡咯烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯烷-1-羧酸酯(335.00毫克,357.61微摩尔)溶于乙酸乙酯(20.00毫升)和甲醇(4.00毫升)混合溶剂中,然后15度氮气保护下加入钯/碳(100.00毫克,10%)。所得悬浊液真空下脱气并用氢气置换几次后,25度氢气氛下(15psi)搅拌24小时。混合液用硅藻土过滤,甲醇(约100毫升)洗涤,滤液减压浓缩得叔-丁基N-[(1S)-1-[(2S)-2-[[5-氟-2-[6-氟-3-[[(2R,4S)-4-氟吡咯烷-2-基]甲基]-1H-吲哚-2-基]苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(309.00毫克,粗品)
MS(ESI)m/z:653.3[M+H+]。
步骤E:将N-Boc-L正丁氨酸(129.88毫克,639.07微摩尔)溶于DMF(3.00毫升),18度下加入N-甲基吗啉(129.28毫克,1.28毫摩尔,140.52微升)和HATU(259.19毫克,681.67微摩尔)。混合液18度搅拌30分钟,然后将叔-丁基N-[(1S)-1-[(2S)-2-[[5-氟-2-[6-氟-3-[[(2R,4S)-4-氟吡咯烷-2-基]甲基]-1H-吲哚-2-基]苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(309.00毫克,粗品) 溶于DMF(3.00毫升)后加入上述混合液,混合液18度继续搅拌1小时后,倒入水中(100毫升)析出固体,继续搅拌10分钟,过滤,干燥后得到叔-丁基N-[(1S)-1-[(2S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-(叔-丁氧羰氨基)丁酰基]-4-氟-吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(346.00毫克,334.45微摩尔,78.50%)
MS(ESI)m/z:838.4[M+H+]。
步骤F:将叔丁基N-[(1S)-1-[(2S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-(叔-丁氧羰氨基)丁酰基]-4-氟-吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基甲酸酯(346.00毫克,412.90微摩尔)溶于二氯甲烷(3.00毫升)18度滴入三氟乙酸(4.62克,40.52毫摩尔,3.00毫升),混合液18度搅拌30分钟后,45度减压蒸馏除去溶剂得(2S)-2-氨基-1-[(2S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-氨基丁酰基]-4-氟-吡咯烷-2-yl]甲基]-6-氟-1氢-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-基]-3-甲基-丁烷-1-酮(357.00毫克,412.34微摩尔,99.87%,三氟乙酸盐)。
步骤G:将N-Boc-N-甲基-L-丙氨酸(209.50毫克,1.03毫摩尔)溶于DMF(3.00毫升),18度下加入N-甲基吗啉(250.25毫克,2.47毫摩尔,272.01微升)和HATU(407.64毫克,1.07毫摩尔),混合液18度搅拌30分钟后,将(2S)-2-氨基-1-[(2S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-氨基丁酰基]-4-氟-吡咯烷-2-yl]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-基]-3-甲基-丁烷-1-酮(357.00毫克,412.34微摩尔,三氟乙酸盐)溶于DMF(3.00毫升)后加入上述混合液,18度继续搅拌1小时后,倒入水中(100毫升)析出固体,混合液继续搅拌10分钟,过滤,干燥后得淡黄色固体,淡黄色固体通过硅胶薄层色谱纯化(石油醚/乙酸乙酯=1/3)得到叔-丁基N-[(1S)-2-[[(1S)-1-[(2S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-[[(2S)-2-[叔-丁氧羰基(甲基)氨基]丙酰基]氨基]丁酰基]-4-氟-吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(200.00毫克,168.62微摩尔,40.89%)
MS(ESI)m/z:1008.5[M+H+]。
步骤H:将叔丁基N-[(1S)-2-[[(1S)-1-[(2S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-[[(2S)-2-[叔-丁氧羰基(甲基)氨基]丙酰基]氨基]丁酰基]-4-氟-吡咯烷-2-基]甲基]-6-氟-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]吡咯烷-1-羰基]-2-甲基-丙基]氨基]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(200.00毫克,168.62微摩尔)溶于二氯甲烷(3.00毫升)中,18度下滴入三氟乙酸(4.62克,40.52毫摩尔,3.00毫升)。混合液18度搅拌30分钟后,减压蒸馏除去溶剂,所得残渣通过制备HPLC纯化得到实施例39(120.00毫克,136.23微摩尔,80.79%,盐酸盐)。
1H NMR(MeOD,400MHz):δ12.34(s,1H),8.93(d,J=6.8Hz,1H),8.58(d,J=7.2Hz,1H),8.12(dd,J=3.6,9.0Hz,1H),7.99-7.90(m,2H),7.59-7.47(m,2H),7.16-7.07(m,1H),5.56-5.35(m,1H),5.07-5.01(m,1H),4.96(d,J=8.8Hz,1H),4.69(d,J=6.8Hz,2H),4.50-4.43(m,1H),4.35(br.s.,1H),4.20-3.97(m,4H),3.88(dd,J=7.8,16.2Hz,2H),3.52(d,J=13.9Hz,1H),3.26-3.15(m,1H),2.73(s,3H),2.67(s,3H),2.35(br.s.,1H),2.20-1.79(m,7H),1.60(d,J=6.8Hz,3H),1.47(d,J=6.6Hz,3H),1.12(t,J=7.2Hz,3H),1.03(s,9H)。
MS(ESI)m/z:808.3[M+H+]。
实施实例40、41、42根据实施实例39的制备过程制备。
实施例40
Figure PCTCN2017082227-appb-000145
1HNMR(MeOD,400MHz):δ8.88(d,J=7.0Hz,1H),8.73(d,J=7.1Hz,1H),8.11(dd,J=3.9,9.1Hz,1H),7.99-7.86(m,2H),7.59-7.44(m,2H),7.11(dt,J=2.1,9.2Hz,1H),5.56-5.35(m,1H),5.10-4.99(m,1H),4.77(br.s.,1H),4.66(d,J=5.1Hz,1H),4.45-4.38(m,1H),4.33(br.s.,1H),4.19-3.94(m,4H),3.92-3.78(m,2H),3.53(d,J=14.2Hz,1H),3.24-3.12(m,1H),2.76-2.60(m,7H),2.31(br.s.,1H),2.24-1.76(m,9H),1.58(d,J=6.8Hz,3H),1.51(d,J=7.0Hz,3H),1.10(t,J=7.3Hz,3H),0.97(dd,J=6.7,19.2Hz,6H)。
MS(ESI)m/z:822.3[M+H+]。
实施例41
Figure PCTCN2017082227-appb-000146
1H NMR(MeOD,400MHz):δ8.20(dd,J=9.03,3.89Hz,1H),7.95(dd,J=8.91,5.14Hz,1H),7.86(dd,J=7.78,2.01Hz,1H),7.51-7.59(m,1H),7.47(dd,J=9.41,2.13Hz,1H),7.12(td,J=9.16,2.13Hz,1H),5.29-5.56(m,1H),4.74(dd,J=14.81,7.65Hz,2H),4.40-4.61(m,4H),4.08-4.26(m,2H),3.89-4.04(m,2H),3.58-3.80(m,3H),3.21-3.30(m,1H),2.63-2.74(m,6H),2.16-2.28(m,2H),2.11(d,J=18.32Hz,1H),1.96-2.06(m,2H),1.83-1.90(m,1H),1.68-1.79(m,1H),1.49-1.59(m,7H),1.42(dd,J=14.81,7.40Hz,1H),1.08-1.20(m,6H),0.87-0.96(m,3H)。
MS(ESI)m/z:808[M+H+]。
实施例42
Figure PCTCN2017082227-appb-000147
1H NMR(MeOD,400MHz):δ8.20(dd,J=9.16,4.02Hz,1H)7.94(dd,J=8.91,5.14Hz,1H),7.89(dd,J=7.84,2.20Hz,1H),7.55(td,J=9.16,2.13Hz,1H),7.48(dd,J=9.29,2.13Hz,1H),7.13(td,J=9.16,2.13Hz,1H),5.32-5.53(m,1H),4.78(dd,J=14.74,7.84Hz,2H),4.40-4.66(m,5H),4.04-4.24(m,3H),3.94(q,J=7.03Hz,1H),3.69-3.81(m,2H),3.64(d,J=13.18Hz,1H),2.61-2.76(m,7H),2.10-2.25(m,2H),2.02(m,1H),1.81-1.93(m,1H),1.68-1.78(m,1H),1.48-1.60(m,7H),1.39-1.46(m,1H),1.20(s,9H),0.90(t,J=7.34Hz,3H)。
MS(ESI)m/z:822[M+H+]。
实施例43
Figure PCTCN2017082227-appb-000148
反应流程:中间体43-8的制备
Figure PCTCN2017082227-appb-000149
步骤A:往N-Boc-反式-4-羟基-L-脯氨酸甲酯(114.00克,464.79毫摩尔)的1.2升二氯甲烷溶液中加入三氯异氰脲酸(113.42克,488.03毫摩尔),在0摄氏度下加入TEMPO(1.46克,9.30毫摩尔)。反应液在10到20摄氏度搅拌半个小时。反应液垫硅藻土过滤,二氯甲烷相用1000毫升的饱和碳酸钾溶液洗涤两次,再用800毫升的饱和食盐水洗涤,无水硫酸钠干燥后,过滤,滤液减压浓缩得到N-Boc-4-氧代-L-脯氨酸甲酯(111.00克,456.30毫摩尔,98.17%)。
1H NMR(CDCl3,400MHz):δ4.81-4.64(m,1H),3.85(d,J=7.8Hz,2H),3.73(s,3H),2.99-2.84(m,1H),2.55(d,J=20.0Hz,1H),1.43(d,J=8.0Hz,9H).
步骤B:在0摄氏度下,往N-Boc-4-氧代-L-脯氨酸甲酯(106.00克,435.75毫摩尔)的500毫升二氯甲烷溶液中滴加DAST(119.40克,740.78毫摩尔)的500毫升二氯甲烷溶液,滴加完后,加入乙醇(4.02克,87.15毫摩尔)。在0到20摄氏度下搅拌18个小时。将反应液倒入缓慢倒入冰的饱和的碳酸 氢钠溶液中,待二氧化碳气体完全溢出后,用2000毫升的二氯甲烷萃取2次,二氯甲烷用无水硫酸钠干燥,过滤,滤液浓缩得到粗产品。粗产品经柱层析纯化(洗脱剂:石油醚/乙酸乙酯=50:1到10:1)得到N-Boc-4,4-二氟-L-脯氨酸甲酯(70.00克,263.90毫摩尔,60.56%)。
1H NMR(CDCl3,400MHz):δ4.57-4.40(m,1H),3.89-3.72(m,5H),2.78-2.60(m,1H),2.45(m,1H),1.44(d,J=20.0Hz,9H)。
步骤C:在10到20摄氏度,往搅拌的N-Boc-4,4-二氟-L-脯氨酸甲酯(50.00克,188.50毫摩尔)的500毫升四氢呋喃溶液中加入氯化锂(17.58克,414.70毫摩尔)和硼氢化钠(17.83克,471.25毫摩尔)。反应液冷却到0摄氏度后滴加1升的乙醇。反应液在0摄氏度搅拌1个小时,再在10到20摄氏度下搅拌17小时后,反应液冷却到0摄氏度,加入10%的硫酸氢钠水溶液调节Ph到3。减压浓缩除去溶剂后,加入500毫升水,用1500毫升的二氯甲烷萃取三次,有机相用无水硫酸钠干燥后过滤,浓缩得到产物N-Boc-4,4-二氟-L-脯氨醇(44.00克,粗品)。
1H NMR(CDCl3,400MHz):δ4.19-3.95(m,2H),3.84-3.40(m,4H),2.42(dq,J=9.0,13.1Hz,1H),2.11(br.s.,1H),1.46-1.37(m,9H)。
步骤D:在10~20摄氏度下,往N-Boc-4,4-二氟-L-脯氨醇(44克,粗品)的440毫升四氢呋喃溶液中加入邻苯二甲酰亚胺(28.65克,194.74毫摩尔)和三苯基膦(51.08克,194.74毫摩尔)。然后滴加DIAD(39.38克,194.74毫摩尔)。滴加完后,在10到20摄氏度下反应16个小时。反应液减压浓缩除去溶剂,加入500毫升水,再用1000毫升二氯甲烷萃取2次。合并的有机相用500毫升饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液浓缩得到粗产物。粗产品经过柱层析纯化(洗脱剂:石油醚/乙酸乙酯=20:1到10:1)得到叔丁基(2S)-2-[(1,3-二氧-2-异吲哚啉)甲基]-4,4-二氟-吡咯烷-1-羧酸酯(65.00克,170.32毫摩尔,91.83%)。
1H NMR(CDCl3,400MHz):δ7.88-7.62(m,4H),4.62-4.37(m,1H),4.01-3.62(m,4H),2.61-2.42(m,1H),2.28(d,J=16.0Hz,1H),1.35-1.17(m,9H)。
步骤E:往叔丁基(2S)-2-[(1,3-二氧-2-异吲哚啉)甲基]-4,4-二氟-吡咯烷-1-羧酸酯(69.00克,188.34毫摩尔)的1升乙醇溶液中加入水合肼(24.05克,470.85毫摩尔)。反应液在60摄氏度下反应2个小时。冷却到室温后用1000毫升的二氯甲烷稀释,过滤,滤饼用二氯甲烷洗涤。有机相合并后减压浓缩得到粗产品。粗产品经200毫升二氯甲烷稀释,过滤,滤饼用二氯甲烷洗涤,有机相合并浓缩得到叔丁基(2S)-2-(氨甲基)-4,4-二氟-吡咯烷-1-羧酸酯(50.00克,粗品)。
1H NMR(DMSO,400MHz):δ3.90-3.68(m,2H),3.45(m,4H),2.83-2.56(m,2H),1.41(s,9H)。
步骤F:在10到20摄氏度下,往叔丁基(2S)-2-(氨甲基)-4,4-二氟-吡咯烷-1-羧酸酯(44.50克,188.35毫摩尔)的500毫升乙腈溶液中加入1,4-二氟-2-硝基-苯(29.96克,188.35毫摩尔)和碳酸钾(52.06克,376.70毫摩尔)。在80摄氏度下反应2小时后,冷却到10到20摄氏度。反应液过滤,滤液浓缩得到粗产品。粗产品经过柱层析(洗脱剂,石油醚/乙酸乙酯=20:1到10:1)得到叔丁基(2S)-4,4-二氟-2-[(4-氟-2-硝基-苯胺基)甲基]吡咯烷-1-羧酸酯(31.00克,78.30毫摩尔,41.57%)。
1H NMR(CDCl3,400MHz):δ8.21(br.s.,1H),7.92(d,J=8.0Hz,1H),7.36-7.30(m,1H),7.24 (br.s.,1H),4.37(br.s.,1H),3.88-3.65(m,3H),3.41(td,m,1H),2.65-2.47(m,1H),2.36(br.s.,1H),1.52(br.s.,9H)。
步骤G:在氮气氛围下,往叔丁基(2S)-4,4-二氟-2-[(4-氟-2-硝基-苯胺基)甲基]吡咯烷-1-羧酸酯(10.00克,26.64毫摩尔)的300毫升甲醇溶液中加入Pd/C(10%,1克)。反应体系用氢气置换3次后在30到40Psi,25到30度下反应2小时。反应液经过滤后,减压浓缩得到叔丁基(2S)-2-[(2-胺基-4-氟-苯胺基)甲基]-4,4-二氟-吡咯烷-1-羧酸酯(9.00克,17.98毫摩尔,67.50%)。
反应流程:中间体43-16的制备
Figure PCTCN2017082227-appb-000150
步骤A:在搅拌下,向N-Cbz-顺式-4-羟基-L-脯氨酸甲酯(150.00克,537.08毫摩尔)的二氯甲烷(1.5升)溶液中加入三氯异氰脲酸(131.06克,563.93毫摩尔),接着在0摄氏度小心加入TEMPO(8.45克,53.71毫摩尔)。将反应物在15摄氏度搅拌30分钟,过滤。滤液用硫代硫酸钠水溶液淬灭(300毫升),然后加水(500毫升)稀释,用二氯甲烷萃取(200毫升×2)。将合并的有机层用盐水(300毫升)洗涤,用无水硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=2/1到1/0)到N-Cbz-4-氧代-L-脯氨酸甲酯(90.00克,324.59毫摩尔,60.44%)。
1H NMR(CDCl3,400MHz):δ7.15-7.47(m,5H)5.07-5.29(m,2H)4.86(dd,J=18.63,10.48Hz,1H)3.88-4.05(m,2H)3.53-3.86(m,3H)2.88-3.03(m,1H)2.61(dd,J=18.89,2.57Hz,1H)。
步骤B:在0摄氏度下,向正在搅拌的N-Cbz-4-氧代-L-脯氨酸甲酯(75.00克,270.49毫摩尔)的无水二氯甲烷(650毫升)溶液中加入DAST(74.12克,459.83毫摩尔,60.75毫升)的二氯甲烷溶液。接着向反应液中加入乙醇(249.23毫克,5.41毫摩尔,315.48微升)。反应液在15摄氏度搅拌12小时,然后用水(300毫升)淬灭。加水(150毫升)稀释,并用乙酸乙酯萃取(500毫升x3)。合并有机相用饱和食盐水洗,用无水硫酸钠干燥,并减压浓缩,得到N-Cbz-4,4-二氟-L-脯氨酸甲酯(78克,粗品)。
1H NMR(CDCl3,400MHz):δ7.29-7.45(m,5H)5.05-5.29(m,2H)4.54-4.70(m,1H)3.91(t,J=12.92Hz,2H)3.59-3.84(m,3H)2.64-2.85(m,1H)2.52(qd,J=13.18,4.89Hz,1H)。
步骤C:在搅拌下,向N-Cbz-4,4-二氟-L-脯氨酸甲酯(78克,粗品)的四氢呋喃(500毫升)溶液加入一水合氢氧化锂(32.81克,781.89毫摩尔)的水溶液(100毫升)。将反应液在15摄氏度搅拌2小时,然后减压浓缩除去四氢呋喃,接着将水溶液用二氯甲烷洗(200毫升x2)。水相用盐酸(1M,大约40毫升)调pH到1,用二氯甲烷萃取(400毫升x3)。有机相用饱和食盐洗(300毫升x1),用无水硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:二氯甲烷/甲醇=1/0至20:1),得到N-Cbz-4,4-二氟-L-脯氨酸(50.00克,152.50毫摩尔,58.51%)。
1H NMR(CDCl3,400MHz):δ8.77(br.s.,1H)7.29-7.46(m,5H)5.09-5.30(m,2H)4.55-4.71(m,1H)3.78-4.04(m,2H)2.50-2.93(m,2H)。
步骤D:在0摄氏度下,向正在搅拌的草酰氯(26.70克,210.35毫摩尔,18.41毫升)的无水甲苯(50毫升)溶液中加入DMF(768.72mg,10.52mmol,809.18uL,0.10eq)。滴加完毕,搅拌30分钟。然后N-Cbz-4,4-二氟-L-脯氨酸在0摄氏度加入到反应液中,将反应混合物在25摄氏度下搅拌5小时。反应液旋干得到产物N-Cbz-4,4-二氟-L-脯氨酰氯,用甲苯溶解,直接用于下一步。
步骤E:在0摄氏度下,向正在搅拌的6-氟吲哚(21.32克,157.76毫摩尔)的甲苯(100毫升)、氯苯(80毫升)混合溶剂中滴加乙基格氏试剂(3M,54.34毫升)并控制在30分钟加完。加完后,反应液在此温度下搅拌30分钟,然后将N-Cbz-4,4-二氟-L-脯氨酰氯的甲苯溶液在0摄氏度下加入到反应液中。将反应混合物在25摄氏度下搅拌5小时。反应液用氯化铵水溶液(300毫升)在25摄氏度下淬灭,然后加水(100毫升)稀释,用乙酸乙酯萃取(200毫升x2)。将合并的有机层用盐水(50毫升x1)洗涤,用无水硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:二氯甲烷/乙酸乙酯等于1:0到10:1),得到(S)-苄基4,4-二氟-2-(6-氟-1H吲哚-3-羰基)吡咯-1-羧酸酯(30.00克,64.87毫摩尔,61.68%),为浅黄色固体。
1H NMR(DMSO,400MHz):δ12.20(d,J=9.29Hz,1H)8.52(dd,J=16.12,2.20Hz,1H)8.16(ddd,J=16.91,8.75,5.71Hz,1H)7.28-7.46(m,4H)7.00-7.18(m,3H)5.40-5.54(m,1H)4.92-5.17(m,2H)3.80-4.13(m,2H)2.93-3.18(m,1H)2.38-2.49(m,1H)
步骤F:在搅拌下,向(S)-苄基4,4-二氟-2-(6-氟-1H-吲哚-3-羰基)吡咯-1-羧酸酯(30克,64.87毫摩尔)的四氢呋喃(200毫升)溶液,加入硼氢化锂溶液(2M,64.87毫升)。将反应液在15摄氏度搅拌4小时。然后向其中加入甲磺酸(11.53克,120.00毫摩尔,8.54毫升)并在15摄氏度搅拌12小时。反应液用氯化铵水溶液(200毫升)淬灭,然后用乙酸乙酯萃取(200毫升x2)。将合并的有机相用盐水(50毫升)洗,硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=1:0/7:1),得到产品(R)-苄基4,4-二氟-2-((6-氟-1H-吲哚-3-基)甲基)吡咯-1-羧酸酯(17克,40.27毫摩尔,62.08%)。
1H NMR(CDCl3,400MHz):δ8.13(s,1H)7.36-7.54(m,6H)6.83-7.10(m,3H)5.24(s,2H)4.30-4.52(m,1H)3.65-4.07(m,2H)3.22-3.52(m,1H)2.75-3.00(m,1H)2.16-2.44(m,2H)。
步骤G:在0摄氏度,氮气保护下,向搅拌的DMF(3.76克,51.50毫摩尔,3.96毫升)中加入三氯氧磷(8.15克,51.50毫摩尔,3.96毫升)。反应液在0摄氏度反应1小时,将溶在1,2-二氯乙烷(20毫升)中(R)-苄基4,4-二氟-2-((6-氟-1H-吲哚-3-基)甲基)吡咯-1-羧酸脂(10克,25.75毫摩尔)滴加 到反应液中,且保持滴加温度为0摄氏度。反应液在15摄氏度反应11小时。反应液用饱和碳酸钠水溶液(100毫升)在0摄氏度淬灭,然后用二氯甲烷萃取(60毫升x3)。将合并的有机相用盐水(60毫升x1)洗,硫酸钠干燥,并减压浓缩。得9.2克(R)-苄基4,4-二氟-2-((6-氟-2-甲酰-1H-吲哚-3-基)甲基)吡咯-1羧酸脂。
反应流程:实施例43的制备
Figure PCTCN2017082227-appb-000151
步骤A:在10到20摄氏度下往苄基(2R)-4,4-二氟-2-[(6-氟-2-甲酰基-1H-3-吲哚)甲基]吡咯烷-1-羧酸酯(9.04克,8.69毫摩尔)的20毫升DMF和0.2毫升水溶液中加入(S)-叔丁基2-(((2-胺基-4-氟苯基)胺基)甲基)-4,4-二氟吡咯烷-1-羧酸酯(3.00克,8.69毫摩尔)和过硫酸氢钾(2.64克,17.38毫摩尔。在10到20摄氏度下搅拌16小时后,加入100毫升水淬灭,用300毫升乙酸乙酯萃取三次。将合并的有机相用300毫升饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产品。粗产品柱层析(洗脱剂,石油醚/乙酸乙酯等于10:1)得到(R)-苄基(2R)-2-[[2-[1-[[(2S)-1-叔丁酰基酰基-4,4-二氟-2-吡咯烷]甲基]-5-氟-2-苯并咪唑]-6-氟-1H-3-吲哚]甲基]-4,4-二氟-吡咯烷-1-羧酸酯(2克,2.56毫摩尔,29.48%)。
步骤B:在10到20摄氏度下往(R)-苄基(2R)-2-[[2-[1-[[(2S)-1-叔丁酰基酰基-4,4-二氟-2-吡咯烷]甲基]-5-氟-2-苯并咪唑]-6-氟-1H-3-吲哚]甲基]-4,4-二氟-吡咯烷-1-羧酸酯(600.00毫克,808.93微摩尔)的5毫升二氯甲烷溶液中加入HBr/AcOH(89.74毫克,808.93微摩尔)。反应液在10到20摄氏度下搅拌1个小时。反应液减压浓缩除去溶剂和乙酸。粗产品用30毫升甲基叔丁基醚洗涤三次后得到1-[[(2R)-4,4-二氟-2-吡咯烷]甲基]-2-[3-[[(2R)-4,4-二氟-2-吡咯烷]甲基]-6-氟-1H-2-吲哚]-5-氟-苯并咪唑(500毫克,672.34毫摩尔,83.12%)。
MS(ESI)m/z:508.2[M+H+]。
步骤C:往N-Boc-L正丁氨酸(227.73毫克,1.12毫摩尔)的2毫升的DMF溶液中加入N-甲基吗啉(226.69毫克,2.24毫摩尔)和HATU(426.08毫克,1.12毫摩尔)。反应液在10到20摄氏度下搅拌30分钟。然后加入1-(((S)-4,4-二氟-2-吡咯烷)甲基)-2-(3-(((R)-4,4-二氟-2-吡咯烷)甲基)-6-氟-1H-2-吲哚)-5-氟-1H-苯并咪唑(250.00毫克,373.52毫摩尔)。在10到20摄氏度下反应16个小时后,加入30毫升水淬灭,用50毫升乙酸乙酯萃取。有机相用50毫升饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗产品。粗产品经薄层制备色谱纯化(展开剂:石油醚/乙酸乙酯=1:1)得到叔丁基N-[(1S)-1-[(2R)-2-[[2-[1-[[(2S)-1-[(2S)-2-(叔丁氧基羰基胺基)书丁酰基]-4,4-二氟-2-吡咯烷]甲基]-5-氟-2-苯并咪唑]-6-氟-1H-3-吲哚]甲基]-4,4-二氟-吡咯烷-1-羰基]丙基]羧酸甲酯(100.00毫克,100.24微摩尔,26.84%)。
MS(ESI)m/z:878.3[M+H+]。
步骤D:将叔丁基N-[(1S)-1-[(2R)-2-[[2-[1-[[(2S)-1-[(2S)-2-(叔丁氧基羰基胺基)书丁酰基]-4,4-二氟-2-吡咯烷]甲基]-5-氟-2-苯并咪唑]-6-氟-1H-3-吲哚]甲基]-4,4-二氟-吡咯烷-1-羰基]丙基]羧酸甲酯(100毫克,113.91微摩尔)溶于HCl/二氧六环(113.91微摩尔,5.00毫升)中,在10到20摄氏度下搅拌反应5小时。反应液减压浓缩得到产物(2S)-2-胺基-1-[(2R)-2-[[2-[1-[[(2S)-1-[(2S)-2-胺基丁酰基]-4,4-二氟-2-吡咯烷]甲基]-5-氟-2-苯并咪唑]-6-氟-1H-3-吲哚]甲基]-4,4-二氟-1-吡咯烷]1-丁酮(85.00毫克,98.52微摩尔,86.49%,盐酸盐)。
MS(ESI)m/z:678.3,[M+H+]。
步骤E:往N-Boc-N-甲基-L-丙氨酸(69.86毫克,343.74微摩尔)的2毫升DMF溶液中加入N-甲基吗啉(69.54毫克,687.48微摩尔)和HATU(130.70毫克,343.74微摩尔)。反应液在10到20摄氏度下搅拌30分钟后,加入(2S)-2-胺基-1-[(2R)-2-[[2-[1-[[(2S)-1-[(2S)-2-胺基丁酰基]-4,4-二氟-2-吡咯烷]甲基]-5-氟-2-苯并咪唑]-6-氟-1H-3-吲哚]甲基]-4,4-二氟-1-吡咯烷]1-丁酮(85.00毫克,114.58微摩尔,盐酸盐)。在10到20摄氏度下反应1.5小时后,在反应体系中加入30毫升水,用50毫升乙酸乙酯萃取,有机相用50毫升饱和食盐水洗涤,再用无水硫酸钠干燥后过滤,滤液减压浓缩得到粗产品。粗产品用薄层色谱(展开剂:石油醚/乙酸乙酯=1:1)纯化后得到叔丁基N-[(1S)-2-[[(1S)-1-[(2R)-2-[[2-[1-[[(2S)-1-[(2S)-2-[[(2S)-2-[叔丁氧基羰基(甲基)胺基]丙酰基]胺基]丁酰基]-4,4-二氟-2-吡咯烷]甲基]-5-氟-2-苯并咪唑]-6-氟1H-3-吲哚]甲基]-4,4-二氟-吡咯烷-1-羰基]丙基]胺基]-1-甲基-2-氧-乙基]-N-甲基-羧酸甲酯(100.00毫克,87.78微摩尔,76.61%)。
MS(ESI)m/z:1048.4,[M+H+]。
步骤F:往叔丁基N-[(1S)-2-[[(1S)-1-[(2R)-2-[[2-[1-[[(2S)-1-[(2S)-2-[[(2S)-2-[叔丁氧基羰基(甲基)胺基]丙酰基]胺基]丁酰基]-4,4-二氟-2-吡咯烷]甲基]-5-氟-2-苯并咪唑]-6-氟1H-3-吲哚]甲基]-4,4-二氟-吡咯烷-1-羰基]丙基]胺基]-1-甲基-2-氧-乙基]-N-甲基-羧酸甲酯(100.00毫克,95.41微摩尔)的4毫升二氯甲烷溶液中加入TFA(3.00克,26.29毫摩尔)。反应液在10到20摄氏度下搅拌2个小时后,减压浓缩除去溶剂。粗产品经过制备HPLC得到实施例43(30.00毫克,32.58微摩尔,34.15%,盐酸盐)。
1HNMR(MeOD,400MHz):δ8.83(d,J=6.0Hz,1H),8.68(d,J=6.4Hz,1H),8.18(d,J=5.6Hz,1H),7.96(dd,J=5.0,8.8Hz,1H),7.78(d,J=6.8Hz,1H),7.58(t,J=8.7Hz,1H),7.40(d,J=8.4Hz,1H),7.12(t,J=8.4Hz,1H),4.85-4.06(m,10H),3.94(dd,J=6.8,16.9Hz,2H),3.67(d,J=12.5Hz,1H),3.10(t,J=11.2Hz,1H),2.74-2.14(m,11H),1.84-1.68(m,2H),1.60-1.44(m,7H),1.43-1.21(m,2H),1.04(t,J=6.4Hz,3H),0.90-0.78(m,3H)。
MS(ESI)m/z:870.3[M+Na+]。
实施实例44至47根据实施实例43的制备过程制备。
实施例44
Figure PCTCN2017082227-appb-000152
1HNMR(MeOD,400MHz):δ8.89(d,J=5.0Hz,1H),8.73(d,J=5.5Hz,1H),8.03(d,J=5.9Hz,1H),7.81(dd,J=5.1,8.6Hz,1H),7.62(d,J=6.7Hz,1H),7.42(t,J=8.2Hz,1H),7.24(d,J=9.0Hz,1H),6.96(t,J=8.8Hz,1H),4.84(br.s.,1H),4.60-4.36(m,2H),4.22-3.89(m,5H),3.87-3.69(m,3H),3.65-3.46(m,2H),2.98(br.s.,1H),2.57-2.01(m,11H),1.33(dd,J=6.5,19.4Hz,6H),1.24-1.01(m,2H),0.66-0.10(m,10H)。
MS(ESI)m/z:872.3[M+H+]。
实施例45
Figure PCTCN2017082227-appb-000153
1HNMR(MeOD,400MHz):δ8.82(d,J=6.9Hz,0.5H),8.66(d,J=7.5Hz,0.5H),8.23(d,J=5.5Hz,1H),7.94(dd,J=5.1,8.8Hz,1H),7.80(d,J=6.7Hz,1H),7.57(t,J=9.0Hz,1H),7.44-7.36(m, 1H),7.12(t,J=8.3Hz,1H),5.12-5.01(m,1H),4.83(dd,J=7.7,14.2Hz,2H),4.66(br.s.,2H),4.55(br.s.,1H),4.45-4.06(m,5H),4.01-3.90(m,2H),3.67(d,J=13.4Hz,1H),3.21-3.09(m,1H),2.70-2.61(m,6H),2.47(d,J=13.9Hz,1H),2.42-2.26(m,2H),2.24-2.08(m,2H),1.91(dd,J=6.5,13.1Hz,1H),1.45(dd,J=6.8,16.6Hz,6H),1.05(t,J=6.8Hz,6H),0.86-0.75(m,6H)。
MS(ESI)m/z:876.3[M+H+]。
实施例46
Figure PCTCN2017082227-appb-000154
1HNMR(MeOD,400MHz):δ8.71(d,J=4.0Hz,1H),8.55(d,J=8.0Hz,1H),8.13(d,J=4.0Hz,1H),7.84(dd,J=8.0,4.0Hz,1H),7.68(d,J=8.0Hz,1H),7.47(t,J=8.0Hz,1H),7.30(d,J=8.0Hz,1H),7.01(t,J=8.0Hz,1H),4.94(d,J=12.0Hz,1H),4.30-4.64(m,3H),3.97-4.29(m,5H),3.83(d,J=4.0Hz,2H),3.55(d,J=12.0Hz,1H),3.03(br.s.,1H),2.50-2.61(m,6H),1.98-2.49(m,5H),1.81(d,J=4.0Hz,1H),1.56-1.76(m,2H),1.28-1.43(m,6H),0.93(t,J=8.0Hz,3H),0.72ppm(dd,J=12.0,8.0Hz,6H)。
MS(ESI)m/z:862.2[M+H+]。
实施例47
Figure PCTCN2017082227-appb-000155
1HNMR(MeOD,400MHz):δ8.79(d,J=8.0Hz,1H),8.67(d,J=8.0Hz,1H),8.18(d,J=8.0Hz,1H),7.98(dd,J=4.0,8.0Hz,1H),7.80(d,J=8.0Hz,1H),7.59(t,J=8.0Hz,1H),7.41(dd,J=4.0,8.0Hz,1H),7.13(t,J=8.0Hz,1H),5.06-4.94(m,2H),4.81(br.s.,1H),4.70(br.s.,1H),4.58(br.s.,1H),4.46-4.08(m,6H),4.02-3.86(m,2H),3.70(d,J=16.0Hz,1H),3.18-3.07(m,1H),2.67(d,J=12.0Hz,6H),2.52(d,J=12.0Hz,1H),2.43-2.08(m,4H),1.53-1.42(m,6H),1.40-1.27(m,2H),1.06(t,J=8.0Hz,6H),0.83(t,J=8.0Hz,3H)。
MS(ESI)m/z:862.2[M+H+]。
实施例48
Figure PCTCN2017082227-appb-000156
反应流程:实施例48的制备
Figure PCTCN2017082227-appb-000157
步骤A:在搅拌下,将L-焦谷氨醇(15克,130.29毫摩尔)、苯甲醛(15.90克,149.83毫摩尔,15.14毫升)和对甲苯磺酸一水合物(1.24克,6.51毫摩尔)的甲苯(250毫升)溶液回流反应5小时。然后冷却到室温,反应液用饱和碳酸钠水溶液(300毫升)淬灭,然后用乙酸乙酯萃取(200毫升x3)。将合并的有机相用盐水(100毫升x1)洗,硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=2/1到2/3),得到产品(3R,7aS)-3-苯基四氢吡咯[1,2-c]噁唑-5(3H)-酮(11克,47.09毫摩尔,36.14%)。
1HNMR(CDCl3,400MHz):δ7.52-7.29(m,5H),6.34(s,1H),4.27-4.21(m,1H),4.20-4.10(m,1H),3.49(t,J=8.03Hz,1H),2.82(dt,J=17.38,9.57Hz,1H),2.56(ddd,J=17.35,9.94,3.70Hz,1H),2.44-2.33(m,1H),2.04-1.87(m,1H)。
步骤B:在-78摄氏度下,将(3R,7aS)-3-苯基四氢吡咯[1,2-c]噁唑-5(3H)-酮(14克,68.88毫摩尔)的无水四氢呋喃(28毫升)溶液滴加到六甲基二硅基胺基钾溶液(1M,68.88毫升)中。加完后,将反应液在-78摄氏度搅拌30分钟。然后向其中加入三甲基氯硅烷(9.73克,89.54毫摩尔,11.31毫升)的无水四氢呋喃(20毫升)溶液,并在1小时内在升温到0摄氏度,并在0摄氏度搅拌3小时。再向反应液中加入苯硒基溴(18.04克,76.46毫摩尔)的无水四氢呋喃(20毫升)溶液,并维持温度0摄氏度,并在17摄氏度搅拌7.5小时。反应液用饱和碳酸氢钠水溶液(250毫升)淬灭,然后用乙酸乙酯萃取(250毫升x3)。将合并的有机相用盐水(100毫升x1)洗,硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=4:1),得到产品(3R,7aS)-3-苯基-6-苯硒基四氢吡咯-[1,2-c]噁唑-5(3H)-酮(20克)。
步骤C:在0摄氏度下,向(3R,7aS)-3-苯基-6-苯硒基四氢吡咯-[1,2-c]恶唑-5(3H)-酮(19克,53.03毫摩尔)的乙酸乙酯(190毫升)溶液,加入过氧化氢水溶液(30%,23.90毫升)。将反应液在0摄氏度搅拌30分钟。然后向其中加入甲磺酸(11.53克,120.00毫摩尔,8.54毫升)并在15摄氏度搅拌12小时。反应液加入水(100毫升),然后用乙酸乙酯萃取(200毫升x1)。将合并的有机相用盐水(100毫升x1)洗,硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:石油醚/乙酸乙酯=4:1到0:1),得到产品(3R,7aS)-3-苯基-1,7a-二氢吡咯[1,2-c]恶唑-5(3H)-酮(7.4克,36.78毫摩尔,69.35%)。
1HNMR(CDCl3,400MHz):δ7.40-7.32(m,2H),7.25-7.13(m,3H),7.09(dd,J=5.93,1.65Hz,1H),6.04-5.96(m,2H),4.48-4.40(m,1H),4.13-4.06(m,1H),3.25(t,J=8.31Hz,1H)。
步骤D:在15摄氏度下,将钠氢(3.05克,76.33毫摩尔,60%纯度)加到二甲基亚砜(160毫升)中。然后向其中分批加入三甲基碘化亚砜(18.90克,85.88毫摩尔,11.31毫升),并在此温度搅拌30分钟。将反应液升温到55摄氏度并搅拌30分钟。再向反应液中滴加(3R,7aS)-3-苯基-3,7a-二氢-1H-吡咯[1,2-c]噁唑-5-酮(6.40克,31.81毫摩尔)的二甲基亚砜(64毫升)溶液,并在55摄氏度搅拌2小时。冷却到4摄氏度,反应液用水(500毫升)淬灭,然后用甲基叔丁基醚萃取(200毫升x2)。将合并的有机相用盐水(100毫升x1)洗,硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:乙酸乙酯/石油醚=20/1到10/1),得到产品(3R,5aR,6aS,6bS)-3-苯基四氢-1H-环丙烷[3,4]吡咯[1,2-c]恶唑-5(3H)-酮(4.7克,20.09毫摩尔,63.15%)。
1HNMR(CDCl3,400MHz):δ7.42-7.29(m,5H),6.39-6.30(m,1H),4.23(dd,J=7.78,6.15Hz,1H),3.91(dd,J=9.16,6.27Hz,1H),3.48(dd,J=9.47,7.97Hz,1H),2.21-1.99(m,2H),1.38-1.30(m,1H),1.20-1.13(m,1H)。
步骤E:在0摄氏度下,将(3R,5aR,6aS,6bS)-3-苯基四氢-1H-环丙烷[3,4]吡咯[1,2-c]噁唑-5(3H)-酮(5克,23.23毫摩尔)的无水四氢呋喃(30毫升)溶液滴加到四氢铝锂(1.5克,39.49毫 摩尔)的无水四氢呋喃(30毫升)悬浊液中。将反应液升温到回流并搅拌1小时。冷却到0摄氏度,非常缓慢滴入水(1.5毫升),然后将15%氢氧化钠水溶液(1.5毫升)、四氢呋喃(15毫升)和水(5毫升)分别加入并剧烈搅拌0.5小时。然后加硫酸钠干燥,过滤并减压浓缩。得到产品((1S,2S,5R)-3-苄基-3-氮杂双环[3.1.0]己烷-2-基)甲醇(4.5克,22.14毫摩尔,95.31%)。
1HNMR(CDCl3,400MHz):δ7.42-7.29(m,5H),6.39-6.30(m,1H),4.23(dd,J=7.78,6.15Hz,1H),3.91(dd,J=9.16,6.27Hz,1H),3.48(dd,J=9.47,7.97Hz,1H),2.21-1.99(m,2H),1.38-1.30(m,1H),1.20-1.13(m,1H)。
步骤F:在15摄氏度下,将钯碳(788.08毫克,442.74微摩尔,5%纯度)加到((1S,2S,5R)-3-苄基-3-氮杂双环[3.1.0]己烷-2-基)甲醇(4.5克22.14毫摩尔)、Boc酸酐(7.25克,33.21毫摩尔,7.63毫升)的乙酸乙酯(150毫升)溶液中。在氢气的氛围(15psi)下搅拌12小时。将反应液过滤并减压浓缩。得到产品(1S,2S,5R)-叔丁基2-(羟甲基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(3.8克,17.82毫摩尔,80.50%)。
1HNMR(CDCl3,400MHz):δ4.12-3.30(m,6H),1.48-1.43(m,9H),1.41-1.21(m,2H),0.77-0.67(m,1H),0.17(dt,J=8.16,3.95Hz,1H)。
步骤G:在0摄氏度氮气保护下,将DIAD(3.96克,19.60毫摩尔,3.81毫升)的无水四氢呋喃(5毫升)溶液滴加到(1S,2S,5R)-叔丁基2-(羟甲基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(3.80克,17.82毫摩尔)、邻苯二甲酰亚胺(2.88克,19.60毫摩尔)和三苯基膦(5.14克,19.60毫摩尔)的无水四氢呋喃(40毫升)溶液中。将反应液升温到15摄氏度并搅拌11小时。反应液用水(300毫升)淬灭,然后用乙酸乙酯萃取(300毫升x4)。将合并的有机相用盐水(200毫升x1)洗,硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:乙酸乙酯/石油醚=9.1%到25%),得到产品(1S,2S,5R)-叔丁基2-((1,3-二羰基异吲哚-2-基)基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(6克,15.85毫摩尔,88.97%)。
1HNMR(CDCl3,400MHz):δ7.90-7.81(m,2H),7.76-7.63(m,2H),4.37-4.16(m,1H),3.92-3.74(m,2H),3.73-3.47(m,1H),3.35(dd,J=10.98,4.20Hz,1H),1.50-1.35(m,2H),1.22(s,9H),0.65(td,J=7.69,5.33Hz,1H),0.23-0.05(m,1H)。
MS(ESI):m/z 744.1[2M+Na+]。
步骤H:在78摄氏度下氮气保护下,将水合肼(39.63毫摩尔,2.27毫升,85%纯度)加到(1S,2S,5R)-叔丁基2-((1,3-二羰基异吲哚-2-基)基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(6克,15.85毫摩尔)的乙醇(40毫升)溶液中。反应液在78摄氏度下搅拌2小时。将反应液过滤并减压浓缩。得到粗品(1S,2S,5R)-叔丁基2-(氨甲基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(3.7克)。
1HNMR(CDCl3,400MHz):δ3.83-3.26(m,3H),2.87-2.72(m,2H),1.44(d,J=2.20Hz,9H),1.41-1.38(m,1H),1.36-1.29(m,1H),0.73-0.62(m,1H),0.12(quin,J=4.46Hz,1H)。
步骤I:在15摄氏度氮气保护下,将1,4-二氟-2-硝基-苯(2.77克,17.43毫摩尔,1.89毫升)加到(1S,2S,5R)-叔丁基2-(氨甲基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(3.7克)、碳酸钾的乙腈(30毫升)溶液中。将反应液升温到82摄氏度并搅拌2小时。反应液加入水(100毫升),然后用乙酸乙酯萃 取(200毫升x1)。将合并的有机相用盐水(100毫升x1)洗,硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:石油醚乙酸乙酯/=20/1到10/1),得到产品(1S,2S,5R)-叔丁基2-(((4-氟-2-硝基苯基)胺)甲基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(5.3克,15.05毫摩尔,86.37%)。
1HNMR(CDCl3,400MHz):δ8.38-8.14(m,1H),7.90(td,J=9.76,2.95Hz,1H),7.14-6.92(m,1H),4.28-4.09(m,1H),3.62-3.34(m,4H),1.65-1.56(m,1H),1.47(d,J=14.81Hz,10H),0.82-0.72(m,1H),0.26-0.16(m,1H)。
步骤J:在25摄氏度氮气保护下,将钯碳(826.59毫克,779.80微摩尔,10%纯度)加到(1S,2S,5R)-叔丁基2-(((4-氟-2-硝基苯基)胺)甲基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(5.3克,,15.05毫摩尔)的乙酸乙酯(150毫升)和甲醇(150毫升)混合溶液中。在25摄氏度氢气的氛围(30psi)下搅拌2小时。将反应液过滤并减压浓缩。得到产品(1S,2S,5R)-叔丁基2-(((2-胺-4-氟苯基)胺)甲基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(4.5克,14.00毫摩尔,93.03%)。
1HNMR(CDCl3,400MHz):δ6.70-6.37(m,3H),3.79-3.07(m,7H),1.48-1.38(m,11H),0.76-0.68(m,1H),0.24-0.14(m,1H)。
步骤K:在25摄氏度下,向(1S,2S,5R)-叔丁基2-(((2-胺-4-氟苯基)胺)甲基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(1g,3.11毫摩尔)和(2R,4S)-苄基4-氟-2-((6-氟-2-甲酰基-1H-吲哚-3-基)甲基)吡咯-1-羧酸酯(2.48,3.11毫摩尔)的DMF(12毫升)和水(600微升)的混合溶液中,加入Oxone(1.42克,9.33毫摩尔)。在25摄氏度下搅拌2小时。反应液分别用饱和碳酸氢钠水溶液(20毫升)和饱和硫代硫酸钠水溶液(20毫升)淬灭,然后用乙酸乙酯萃取(20毫升x3)。将合并的有机相用盐水(20毫升x1)洗,硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:二氯甲烷/乙酸乙酯等于30:1到11:1),得到产品(1S,2S,5R)-叔丁基2-((2-(3-(((2R,4S)-1-((苄氧基)羰基)-4-氟吡咯-2-基)甲基)-6-氟-1H-吲哚-2-基)-5-氟-1H-苯并[d]咪唑-1-基)甲基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(1克,1.17毫摩尔,37.68%)。
1HNMR(CDCl3,400MHz):δ7.47-7.29(m,8H),7.08(d,J=8.93Hz,4H),5.36-4.81(m,4H),3.83-3.35(m,5H),1.93-1.57(m,2H),1.40-1.22(m,8H),1.09-0.85(m,5H),0.55-0.38(m,1H),-0.04(d,J=15.16Hz,1H)。
步骤L:在15摄氏度下,向(1S,2S,5R)-叔丁基2-((2-(3-(((2R,4S)-1-((苄氧基)羰基)-4-氟吡咯-2-基)甲基)-6-氟-1H-吲哚-2-基)-5-氟-1H-苯并[d]咪唑-1-基)甲基)-3-氮杂二环[3.1.0]己烷-3-羧酸酯(1g,1.17毫摩尔)中,加入氢溴酸乙酸溶液(5毫升,浓度35%)。在15摄氏度下搅拌1小时。反应液加入水(50毫升),然后用甲基叔丁基醚洗(20毫升x3)。水相用碳酸钠调pH到10,然后用二氯甲烷萃取(50毫升x4)。将合并的有机相用盐水(50毫升x1)洗,硫酸钠干燥,并减压浓缩。得到1-((1S,2S,5R)-3-氮杂二环[3.1.0]己烷-2-基甲基)-5-氟-2-(6-氟-3-(((2R,4S)-4-氟吡咯-2-基)甲基)-1H-吲哚-2-基)-1H-苯并[d]咪唑(660毫克,粗品)。
1HNMR(DMSO,400MHz):δ14.74(br.s.,1H),7.89(ddd,J=17.82,8.85,5.08Hz,2H),7.66(dd,J=9.41,2.38Hz,1H),7.38-7.25(m,2H),7.06(td,J=9.22,2.26Hz,1H),5.67-5.46(m,1H),4.43(d,J=14.68Hz,1H),4.20-3.95(m,4H),3.52-3.33(m,5H), 3.08(dd,J=11.42,3.01Hz,1H),2.31-2.16(m,1H),1.74-1.64(m,1H),1.61-1.53(m,1H),0.62(td,J=7.65,4.64Hz,1H),0.37(q,J=4.02Hz,1H)。
步骤M:在15摄氏度下,将HATU(808.64毫克,2.13毫摩尔)加到N-Boc-L正丁氨酸(432.21毫克,2.13毫摩尔)、N-甲基吗啉(286.82毫克,2.84毫摩尔,311.76微升)的DMF(5毫升)搅拌的溶液中。然后将1-((1S,2S,5R)-3-氮杂二环[3.1.0]己烷-2-基甲基)-5-氟-2-(6-氟-3-(((2R,4S)-4-氟吡咯-2-基)甲基)-1H-吲哚-2-基)-1H-苯并[d]咪唑(330毫克,粗品)加入到反应液中。反应液在15摄氏度下搅拌2小时。反应液直接用反相柱纯化(洗脱剂:乙腈和1‰三氟乙酸的水溶液),得到产品叔丁基N-[(1S)-1-[(2R,4S)-2-[[2-[1-[[(1S,2S,5R)-3-[(2S)-2-(叔丁氧羰基氨基)丁酰基]-3-氮杂二环[3.1.0]己烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯-1-羰基]丙基]氨基甲酸酯(390毫克,452.54微摩尔,63.84%)。
1HNMR(CDCl3,400MHz):δ7.56-7.38(m,3H),7.22(d,J=9.16Hz,2H),7.15-6.95(m,5H),5.33(d,J=8.66Hz,1H),4.74-4.52(m,4H),4.41-4.06(m,7H),4.01-3.71(m,8H),3.66-3.55(m,3H),1.82(dt,J=13.43,6.71Hz,4H),1.73-1.59(m,6H),1.47-1.41(m,30H),1.04-0.96(m,5H),0.93-0.79(m,3H),0.73-0.52(m,6H),0.43(t,J=7.28Hz,1H),0.14-0.07(m,1H)。
步骤N:在15摄氏度下,向叔丁基N-[(1S)-1-[(2R,4S)-2-[[2-[1-[[(1S,2S,5R)-3-[(2S)-2-(叔丁氧羰基氨基)丁酰基]-3-氮杂二环[3.1.0]己烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯-1-羰基]丙基]氨基甲酸酯(390毫克,452.54微摩尔)的二氯甲烷(2毫升)溶液中,加入三氟乙酸(2毫升)。在15摄氏度下搅拌12小时。然后减压浓缩。得到粗品(S)-2-氨基-1-((2R,4S)-2-((2-(1-(((1S,2S,5R)-3-((S)-2-氨基丁酰基)-3-氮杂二环[3.1.0]己烷-2-基)甲基)-5-氟-1H-苯并[d]咪唑-2-基)-6-氟-1H-吲哚-3-基)甲基)-4-氟吡咯-1-基)丁基-1-酮(三氟乙酸盐,403毫克)。
步骤O:在15摄氏度下,将HATU(532.20毫克,1.4毫摩尔)加到N-Boc-N-甲基-L-丙氨酸(432.21毫克,1.4毫摩尔)、N-甲基吗啉(283.15毫克,2.80毫摩尔)的DMF(5毫升)搅拌的溶液中。然后将(S)-2-氨基-1-((2R,4S)-2-((2-(1-(((1S,2S,5R)-3-((S)-2-氨基丁酰基)-3-氮杂二环[3.1.0]己烷-2-基)甲基)-5-氟-1H-苯并[d]咪唑-2-基)-6-氟-1H-吲哚-3-基)甲基)-4-氟吡咯-1-基)丁基-1-酮(403毫克)加入到反应液中。反应液在15摄氏度下搅拌0.5小时。反应液直接用反相柱纯化(洗脱剂:乙腈和1‰三氟乙酸的水溶液),得到产品叔丁基N-[(1S)-2-[[(1S)-1-[(2R,4S)-2-[[2-[1-[[(1S,2S,5R)-3-[(2S)-2-[[(2S)-2-[叔丁氧羰基(甲基)氨基]丙酰基]氨基]丁酰基]-3-氮杂二环[3.1.0]己烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯-1-羰基]丙基]氨基]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(350毫克,347.86微摩尔,74.56%)。
1HNMR(CDCl3,400MHz):δ8.07-7.97(m,1H),7.43(m,1H),7.30(d,J=8.91Hz,1H),7.10(d,J=8.28Hz,1H),7.04-6.96(m,2H),6.94-6.85(m,2H),5.14-4.90(m,2H),4.60-4.39(m,7H),3.89-3.69(m,5H),3.67-3.46(m,4H),2.78-2.65(m,11H),1.84-1.52(m,8H),1.50-1.35(m,35H),1.11-0.97(m,3H),0.96-0.67(m,7H),0.60-0.42(m,5H),0.06--0.06(m,1H)。
步骤P:在15摄氏度下,向叔丁基N-[(1S)-2-[[(1S)-1-[(2R,4S)-2-[[2-[1-[[(1S,2S,5R)-3-[(2S)-2-[[(2S)-2-[叔丁氧羰基(甲基)氨基]丙酰基]氨基]丁酰基]-3-氮杂二环[3.1.0]己烷-2-基]甲基]-5-氟-苯并咪唑-2-基]-6-氟-1H-吲哚-3-基]甲基]-4-氟-吡咯-1-羰基]丙基]氨基]-1-甲基-2-氧-乙基]-N-甲基-氨基甲酸酯(350毫克,347.86微摩尔)的二氯甲烷(2毫升)溶液中,加入三氟乙酸(2毫升)。在15摄氏度下搅拌12小时。然后减压浓缩,得到粗品,粗品用制备HPLC(HCl)纯化,得到实施例48(盐酸盐,170毫克,193.43微摩尔,55.61%)。
1HNMR(MeOD,400MHz):δ8.13(dd,J=9.05,3.79Hz,1H),7.85(dd,J=8.80,5.14Hz,1H),7.75(dd,J=7.70,1.96Hz,1H),7.46(t,J=9.11Hz,1H),7.37(dd,J=9.29,1.83Hz,1H),7.04(td,J=9.17,1.96Hz,1H),5.47-5.22(m,1H),4.89(m,J=6.80Hz,1H),4.79-4.70(m,2H),4.46(d,J=6.72Hz,2H),4.31(t,J=6.79Hz,1H),4.19-4.06(m,1H),4.04-3.66(m,6H),3.56(d,J=11.74Hz,1H),3.20-3.09(m,1H),2.63(s,3H),2.55(s,3H),2.14-1.60(m,5H),1.49-1.42(m,3H),1.38(d,J=6.97Hz,4H),1.13(dd,J=17.73,7.46Hz,1H),1.03(t,J=7.27Hz,3H),0.71-0.62(m,3H),0.60(d,J=5.62Hz,1H),0.00(d,J=4.40Hz,1H)。
MS(ESI)m/z:806[M+H+]。
实施实例49根据实施实例48的制备过程制备。
实施例49
Figure PCTCN2017082227-appb-000158
1H NMR(MeOD,400MHz):δ8.24(dd,J=9.03,3.76Hz,1H),7.98-7.82(m,2H),7.63-7.44(m,2H),7.14(td,J=9.13,2.07Hz,1H),5.60-5.35(m,1H),5.03-4.96(m,1H),4.60-4.51(m,1H),4.49-4.39(m,2H),4.30-4.10(m,4H),4.08-3.98(m,2H),3.95-3.79(m,3H),3.71-3.60(m,1H),2.71(s,3H),2.65(s,3H),2.31-2.18(m,2H),2.09-1.96(m,1H),1.90-1.72(m,2H),1.53(d,J=6.78Hz,3H),1.45(d,J=6.90Hz,4H),1.14(dd,J=6.46,4.20Hz,6H),0.78(d,J=6.65Hz,3H),0.69(d,J=6.65Hz,4H),0.12-0.02(m,1H)。
MS(ESI)m/z:834[M+H+]。
实施例50
Figure PCTCN2017082227-appb-000159
反应流程:实施例50的制备
Figure PCTCN2017082227-appb-000160
步骤A:在15摄氏度下,向正在搅拌的N-Cbz-顺式-4-氟-L-脯氨酸(5.00克,18.71毫摩尔)的无水甲苯(50毫升)溶液中加入DMF(13.68毫克,187.10微摩尔,14.40微升)。滴加完毕,搅拌 15分钟。然后草酰氯(3.56克,28.07毫摩尔,2.46毫升)在15摄氏度加入到反应液中,将反应混合物在15摄氏度下搅拌2小时。反应液旋干除去多余的草酰氯,用甲苯溶解,然后用于下一步。
步骤B:在0摄氏度下,向正在搅拌的吲哚(3.29克,28.10毫摩尔)的甲苯(25毫升)、氯苯(25毫升)混合溶剂中滴加乙基格氏试剂(3M,9.68毫升)并控制在30分钟加完。加完后,反应液在此温度下搅拌30分钟,然后将N-Cbz-顺式-4-氟-L-脯氨酰氯(5.35克,18.73毫摩尔)在0摄氏度下加入到反应液中。将反应混合物在25摄氏度下搅拌2小时。反应液用氯化铵水溶液(300毫升)在25摄氏度下淬灭,然后加水(100毫升)稀释,用乙酸乙酯萃取(200毫升x2)。将合并的有机层用盐水(200毫升x1)洗涤,用无水硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:乙酸乙酯/二氯甲烷等于0%到10%),得到(2S,4S)-苄基4-氟-2-(1H-吲哚-3-羰基)吡咯-1-羧酸酯(3.80克,7.68毫摩尔,40.98%)。
1HNMR(DMSO,400MHz):δ12.07(s,1H),8.43(dd,J=7.64,3.00Hz,1H),8.30-8.17(m,1H),7.55(d,J=8.07Hz,1H),7.50-7.34(m,3H),7.33-7.11(m,5H),5.47-5.25(m,2H),5.17(d,J=2.81Hz,1H),5.06(s,1H),4.00-3.72(m,2H),2.98-2.69(m,1H),2.47-2.26(m,1H)。
MS(ESI)m/z:367.0[M+H+]
步骤C:在搅拌下,向(2S,4S)-苄基4-氟-2-(1H-吲哚-3-羰基)吡咯-1-羧酸酯(3.80克,7.68毫摩尔)的四氢呋喃(36毫升)溶液,加入硼氢化锂溶液(2M,7.68毫升)。将反应液在15摄氏度搅拌4小时。然后向其中加入甲磺酸(1.36克,14.20毫摩尔,1.01毫升)并在15摄氏度搅拌12小时。反应液用氯化铵水溶液(200毫升)淬灭,然后用乙酸乙酯萃取(200毫升x2)。将合并的有机相用盐水(50毫升)洗,硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:乙酸乙酯/石油醚=0%到14%),得到产品(2R,4S)-4-氟-2-(1H-吲哚-3-基甲基)吡咯-1-羧酸酯(1.80克,4.60毫摩尔,59.86%)。
1HNMR(DMSO,400MHz):δ10.86(d,J=8.31Hz,1H),7.82-6.64(m,10H),5.42-5.22(m,1H),5.22-5.14(m,2H),4.16(m,J=8.30Hz,1H),3.82-3.59(m,2H),3.22(d,J=11.62Hz,1H),2.80-2.69(m,1H),2.13-1.92(m,2H)。
MS(ESI)m/z:375.0[M+Na+]
步骤D:在0摄氏度,氮气保护下,向搅拌的DMF(672.00毫克,9.19毫摩尔,707.36微升)中加入三氯氧磷(1.66克,10.83毫摩尔,1.01毫升)。反应液在0摄氏度反应1小时,将溶在1,2-二氯乙烷(20毫升)中(2R,4S)-4-氟-2-(1H-吲哚-3-基甲基)吡咯-1-羧酸酯(1.80克,4.60毫摩尔)滴加到反应液中,且保持滴加温度为0摄氏度。反应液在15摄氏度反应11小时。反应液用饱和碳酸氢钠水溶液(100毫升)在0摄氏度淬灭,然后加二氯甲烷(50毫升)稀释,然后用二氯甲烷萃取(100毫升x2)。将合并的有机相用盐水(100毫升x1)洗,硫酸钠干燥,并减压浓缩。得到粗品(2R,4S)-4-氟-2-[(2-甲酰基-1H-吲哚-3-基)甲基]吡咯-1-羧酸酯(1.5克)。
步骤E:在15摄氏度下,向(2R,4S)-苄基4-氟-2-((2-甲酰基-1H-吲哚-3-基)甲基)吡咯-1-羧酸酯(1.50克,1.42毫摩尔)和(2S,4S)-叔丁基2-(((2-胺-4-氟苯)胺)甲基)-4-氟吡咯-1-羧酸酯(464.71毫克,1.42毫摩尔)的DMF(15毫升)和水(1毫升)的混合溶液中,加入Oxone(648.03毫克,4.26毫摩尔)。在15摄氏度下搅拌12小时。反应液分别用饱和碳酸氢钠水溶液(20毫升)和饱和硫代硫酸钠水溶液 (20毫升)淬灭,然后用乙酸乙酯萃取(20毫升x3)。将合并的有机相用盐水(20毫升x1)洗,硫酸钠干燥,并减压浓缩。将残余物用硅胶柱层析纯化(洗脱剂:乙酸乙酯/石油醚等于3.2%到50%),得到产品(2R,4S)-苄基2-((2-(1-(((2S,4S)-1-(叔丁氧羰基)-4-氟吡咯-2-基)甲基)-5-氟-1H-苯并[d]咪唑-2-基)-1H-吲哚-3-基)甲基)-4-氟吡咯-1-羧酸酯(450.00毫克,615.05微摩尔,43.31%)。
1HNMR(CDCl3,400MHz):δ7.58-7.30(m,9H),7.23(d,J=7.65Hz,1H),7.07(td,J=9.16,2.38Hz,1H),5.28-4.89(m,4H),4.68-4.22(m,3H),3.84-3.42(m,5H),3.17(br.s.,1H),2.20(d,J=13.80Hz,1H),1.94-1.66(m,3H),1.28(s,9H)。
MS(ESI)m/z:688.2[M+H+]
步骤F:在15摄氏度下,向(2R,4S)-苄基2-((2-(1-(((2S,4S)-1-(叔丁氧羰基)-4-氟吡咯-2-基)甲基)-5-氟-1H-苯并[d]咪唑-2-基)-1H-吲哚-3-基)甲基)-4-氟吡咯-1-羧酸酯(450.00毫克,615.05微摩尔)的二氯甲烷(3毫升)中,加入三氟乙酸(3毫升)。在15摄氏度下搅拌12小时。然后减压浓缩。得到粗品(2R,4S)-苄基4-氟-2-((2-(5-氟-1-(((2S,4S)-4-氟吡咯-2-基)甲基)-1H-苯并[d]咪唑-2-基)-1H-吲哚-3-基)甲基)吡咯-1-羧酸酯(500毫克,三氟乙酸盐)。
步骤G:在15摄氏度下,将HATU(808.64毫克,2.13毫摩尔)加到(S)-2-((叔丁氧羰基)胺)-3-异戊酸(232.23毫克,1.07毫摩尔)、N-甲基吗啉(216.24毫克,2.14毫摩尔,235.04微升)的DMF(3毫升)搅拌的溶液中。然后将(2R,4S)-苄基4-氟-2-((2-(5-氟-1-(((2S,4S)-4-氟吡咯-2-基)甲基)-1H-苯并[d]咪唑-2-基)-1H-吲哚-3-基)甲基)吡咯-1-羧酸酯(500毫克,三氟乙酸盐)加入到反应液中。反应液在15摄氏度下搅拌0.5小时。反应液直接用反相柱纯化(洗脱剂:乙腈和1‰三氟乙酸的水溶液),得到产品(2R,4S)-苄基2-((2-(1-(((2S,4S)-1-((S)-2-((叔丁氧羰基)胺)-3-异戊酰基)-4-氟吡咯-2-基)甲基)-5-氟-1H-苯并[d]咪唑-2-基)-1H-吲哚-3-基)甲基)-4-氟吡咯-1-羧酸酯(250.00毫克,292.29微摩尔,41.02%)。
1HNMR(CDCl3,400MHz):δ7.63-6.79(m,12H),5.18-4.80(m,5H),4.70-4.21(m,3H),4.04-3.39(m,6H),3.10-2.85(m,1H),1.81-1.47(m,4H),1.34(s,9H),0.84-0.67(m,4H),0.54(dd,J=18.13,6.34Hz,3H)。
MS(ESI)m/z:787.2[M+H+]。
步骤H:在15摄氏度氮气保护下,将钯碳(26.12毫克,317.71微摩尔,10%纯度)加到(2R,4S)-苄基2-((2-(1-(((2S,4S)-1-((S)-2-((叔丁氧羰基)胺)-3-异戊酰基)-4-氟吡咯-2-基)甲基)-5-氟-1H-苯并[d]咪唑-2-基)-1H-吲哚-3-基)甲基)-4-氟吡咯-1-羧酸酯(250.00毫克,292.29微摩尔)的乙酸乙酯(5毫升)和甲醇(5毫升)混合溶液中。在15摄氏度氢气的氛围(30psi)下搅拌12小时。将反应液过滤并减压浓缩。得到产品叔丁基((S)-1-((2S,4S)-4-氟-2-((5-氟-2-(3-(((2R,4S)-4-氟吡咯-2-基)甲基)-1H-吲哚-2-基)-1H-苯并[d]咪唑-1-基)甲基)吡咯-1-基)-3-甲基-1-氧代丁烷-2-基)氨基甲酸酯(163.00毫克,249.71微摩尔,78.60%)。
MS(ESI)m/z:653.3[M+H+]。
步骤I:在15摄氏度下,将HATU(142.42毫克,374.57微摩尔)加到N-Boc-L正丁氨酸(76.13毫克,374.57微摩尔)、氮甲基吗啡啉(75.77毫克,749.13微摩尔,82.36微升)的DMF(3毫升)搅拌的溶液中。然后将叔丁基((S)-1-((2S,4S)-4-氟-2-((5-氟-2-(3-(((2R,4S)-4-氟吡咯-2-基)甲基)-1H-吲哚-2-基)-1H-苯并[d]咪唑-1-基)甲基)吡咯-1-基)-3-甲基-1-氧代丁烷-2-基)氨基甲酸酯(163.00毫克,249.71 微摩尔)加入到反应液中。反应液在15摄氏度下搅拌0.5小时。反应液直接用反相柱纯化(洗脱剂:乙腈和1‰三氟乙酸的水溶液),得到产品叔丁基N-[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-(叔丁氧羰基氨基)丁酰基]-4-氟吡咯-2-基]甲基]-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]-4-氟吡咯-1-羰基]-2-甲基-丙基]氨基甲酸酯(100.00毫克,119.34微摩尔,47.79%)。
MS(ESI)m/z:838.3[M+H+]。
步骤J:在15摄氏度下,向叔丁基N-[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-(叔丁氧羰基氨基)丁酰基]-4-氟吡咯-2-基]甲基]-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]-4-氟吡咯-1-羰基]-2-甲基-丙基]氨基甲酸酯(100.00毫克,119.34微摩尔)的二氯甲烷(2毫升)中,加入三氟乙酸(2毫升)。在15摄氏度下搅拌0.5小时。然后减压浓缩。得到粗品(S)-2-氨-1-((2S,4S)-2-((2-(3-(((2R,4S)-1-((S)-2-氨基丁酰基)-4-氟吡咯-2-基)甲基)-1H-吲哚-2-基)-5-氟-1H-苯并[d]咪唑-1-基)甲基)-4-氟吡咯-1-基)-3-异戊烷-1-羰基(120毫克,三氟乙酸盐)。
MS(ESI)m/z:638.2[M+H+]。
步骤K:在15摄氏度下,将HATU(158.10毫克,415.81微摩尔)加到N-Boc-N-甲基-L-丙氨酸(84.51毫克,415.81微摩尔)、N-甲基吗啉(84.12毫克,831.62微摩尔,91.43微升)的DMF(5毫升)搅拌的溶液中。然后将(S)-2-氨-1-((2S,4S)-2-((2-(3-(((2R,4S)-1-((S)-2-氨基丁酰基)-4-氟吡咯-2-基)甲基)-1H-吲哚-2-基)-5-氟-1H-苯并[d]咪唑-1-基)甲基)-4-氟吡咯-1-基)-3-异戊烷-1-羰基(120毫克,三氟乙酸盐)加入到反应液中。反应液在15摄氏度下搅拌12小时。反应液直接用反相柱纯化(洗脱剂:乙腈和1‰三氟乙酸的水溶液),得到产品叔丁基N-[(1S)-2-[[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-[[(2S)-2-[叔丁氧羰基(甲基)氨基]丙酰基]氨基]丁酰基]-4-氟-吡咯-2-基]甲基]-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]-4-氟-吡咯-1-羰基]-2-甲基-丙烷]氨基]-1-甲基-2-氧代-乙基]-N-甲基-氨基甲酸酯(65.00毫克,64.47微摩尔,46.51%)。
MS(ESI)m/z:1008.5[M+H+]。
步骤L:在15摄氏度下,向叔丁基N-[(1S)-2-[[(1S)-1-[(2S,4S)-2-[[2-[3-[[(2R,4S)-1-[(2S)-2-[[(2S)-2-[叔丁氧羰基(甲基)氨基]丙酰基]氨基]丁酰基]-4-氟-吡咯-2-基]甲基]-1H-吲哚-2-基]-5-氟-苯并咪唑-1-基]甲基]-4-氟-吡咯-1-羰基]-2-甲基-丙烷]氨基]-1-甲基-2-氧代-乙基]-N-甲基-氨基甲酸酯(65.00毫克,64.47微摩尔)的二氯甲烷(2毫升)溶液中,加入三氟乙酸(2毫升)。在15摄氏度下搅拌0.5小时。然后减压浓缩,得到粗品,粗品用反相柱纯化(洗脱剂:乙腈和1‰盐酸的水溶液),得到实施例50(36.00毫克,40.46微摩尔,收率62.76%,盐酸盐)。
1HNMR(MeOD,400MHz):δ8.17(dd,J=9.10,4.08Hz,1H),7.94(d,J=8.28Hz,1H),7.85(dd,J=7.84,2.20Hz,1H),7.75(d,J=8.28Hz,1H),7.56(td,J=9.29,2.38Hz,1H),7.47(t,J=7.59Hz,1H),7.31(t,J=7.65Hz,1H),5.29-5.56(m,2H),5.05-4.72(m,4H),4.61-4.51(m,2H),4.42-4.33(m,1H),4.16-3.97(m,5H),3.64(d,J=11.67Hz,1H),3.32-3.23(m,1H),2.71(s,3H),2.67(s,3H),2.21-1.84(m,7H),1.56(d,J=6.90Hz,3H),1.49(d,J=6.90Hz,3H),1.12(t,J=7.40Hz,3H),0.94(d,J=6.78Hz,3H),0.89(d,J=6.65Hz,3H)。
MS(ESI)m/z:808.3[M+H+]。
实施例51
Figure PCTCN2017082227-appb-000161
根据实施实例50的制备过程制备
1HNMR(MeOD,400MHz):δ8.15(br dd,J=9.17,3.79Hz,1H),7.96-7.84(m,2H),7.79(d,J=1.10Hz,1H),7.57(br t,J=8.93Hz,1H),7.30(dd,J=8.62,1.28Hz,1H),5.59-5.29(m,2H),5.02(m,1H),4.85-4.69(m,2H),4.61-4.47(m,2H),4.41-4.35(m,1H),4.23-3.94(m,6H),3.63(d,J=13.82Hz,1H),3.26(m,1H),2.69(d,J=12.72Hz,6H),2.21-1.81(m,7H),1.54(dd,J=16.14,6.85Hz,6H),1.11(t,J=7.27Hz,3H),0.94(dd,J=18.89,6.66Hz,6H)。
MS(ESI)m/z:842.3[M+H+]。
生物实验实例1:化合物对cIAP1-BIR3,cIAP2-BIR3,XIAP-BIR3蛋白的亲和力测试
1)表中列举的化合物对cIAP1-BIR3,cIAP2-BIR3,XIAP-BIR3蛋白的亲和力测试参考Nikolovska-Colesks,Z.et.Al.(Analytical Biochemistry,2004,332:261-273),得到IC50值。概括的讲,在10-dose IC50mode中,不同浓度的IAP拮抗剂采用3倍连续稀释,以荧光标记的ARPFAQ-K(5-FAM)-NH2肽为探针,测试化合物对蛋白的BIR3区域。
2)测试条件:cIAP1-BIR3:20nM;cIAP2-BIR3:60nM;cIAP1-BIR3:30nM5nM探针在100mM磷酸钾,pH 7.5,0.1mg/ml BSA,0.005%Triton X100and 0.5%DMSO
3)测试流程:
用ECHO(LabCyte),将化合物加入到一定浓度的蛋白中,接着15min预孵化,探针加入进去最终浓度为5nM;60min孵化后测试FP并且计算mP。实验结果见表1。
表1
Figure PCTCN2017082227-appb-000162
Figure PCTCN2017082227-appb-000163
结论:本发明化合物具有较高的cIAP1抑制活性和中等的XIAP抑制活性,具有较好的选择性。
生物实验实例2:TNF-α诱导的NF-κB reporter实验
第一天:
一、化合物稀释:
1)将测试化合物加相应DMSO配成10mM母液;
2)取32ul参考化合物Birinapant(10mM)到128ul DMSO中,配成2mM溶液;
3)利用Echo将2mM的参考化合物Birinapant和10mM的测试化合物,各取0.5ul到Greiner的96孔黑色细胞培养板中,使参考化合物的起始浓度为10uM,测试化合物的起始浓度为50uM,之后以5倍的梯度连续稀释9个点;每块板重复3份。
4)以终浓度为10uM的参考化合物WXFL2012A001(Birinapant)作为HPE,以终浓度为0.5%的DMSO为ZPE。化合物排版图如下表2:
表2
Figure PCTCN2017082227-appb-000164
Figure PCTCN2017082227-appb-000165
5)3个重复板的用途:第一块板用于检测化合物活性;第二块板用于检测化合物对细胞的毒性;第三块板用于第一块板第二天换液时使用:将诱导剂TNF-α与含0.1%FBS的培养基混合加入第三块板并混匀,再将此混合物转到弃去上清的第一块板中。
二、孵育细胞
1)将NF-κB Luciferase Reporter Hela细胞培养瓶中的培养基吸掉,用10ml PBS清洗细胞一次。
2)加3ml 0.25%的胰酶到该T150细胞培养瓶中,放置于37℃,5%CO2的细胞培养箱中消化细胞3分钟,然后加入10ml的培养基终止消化,并用电动移液枪吹打至细胞分散成单个细胞。
3)用细胞计数仪Countstar计算细胞密度。
4)将NF-κB Luciferase Reporter Hela细胞密度用培养基调整至2.0×105cells/ml。
5)将细胞加入含化合物的2块Greiner的96孔黑色细胞培养板中,每孔100ul(2.0×104细胞/孔),一块板用于检测化合物活性;另一块板用于检测化合物的细胞毒性。
6)将铺好的细胞板在37℃,5%CO2的细胞培养箱中孵育24小时。
第二天:
一、TNF-α诱导
1)用0.1%FBS的培养液将TNF-α(100ug/ml)稀释至20ng/ml,取100ul/孔加入到第三块化合物板中。
2)将化合物处理24小时后的第一块96孔黑色细胞培养板(化合物活性检测板)中的培养基弃去,替换成第三块板中含化合物和20ng/ml TNF-α的新鲜培养液。
3)放置于37℃,5%CO2的细胞培养箱中共孵育6小时。
4)6小时后,第一块化合物活性检测板按Bright-Glo(Promega)说明书方法,用Envision读板仪检测各孔荧光素酶信号。
5)第二块化合物毒性检测板按ATPlite 1Step(Perkin Elmer)说明书方法,用Envision读板仪检测各孔荧光素酶信号。
6)利用软件分析,得到化合物的EC50。实验结果见表3。
表3
实施例 EC50(nM) 实施例 EC50(nM) 实施例 EC50(nM)
1 <10 18 <10 35 <10
2 10-100 19 <10 36 <10
3 <10 20 <10 37 10-100
4 10-100 21 10-100 38 <10
5 10-100 22 <10 39 10-100
6 10-100 23 <10 40 <10
7 10-100 24 10-100 41 10-100
8 <10 25 10-100 42 10-100
9 10-100 26 <10 43 >100
10 10-100 27 <10 44 >100
11 10-100 28 <10 45 10-100
12 10-100 29 <10 46 10-100
13 >100 30 <10 47 10-100
14 10-100 31 10-100 48 10-100
15 10-100 32 <10 49 <10
16 <10 33 <10 50 10-100
17 <10 34 <10 51 <10
实施例化合物与对照品IAP抑制剂Birinapant头对头测试实验结果如表4所示:
表4
Figure PCTCN2017082227-appb-000166
结论:本发明的代表性化合物具有与Birinapant相当甚至更高的TNF-α诱导的NF-κB抑制活性。

Claims (32)

  1. 式(I)所示化合物或其药学上可接受的盐,
    Figure PCTCN2017082227-appb-100001
    其中,
    R1和R2分别独立地选自
    Figure PCTCN2017082227-appb-100002
    R3、R4和R5分别独立地选自任选被1、2或3个R取代的:C1-6烷基、C1-6杂烷基、C3-12环烷基、3~12元杂环烷基、5~12元芳基或杂芳基、5~12元芳烷基或杂芳烷基;
    R6a和R6b分别独立地选自H、F、Cl、Br、I、OH、CN、NH2、COOH,或选自任选被1、2或3个R取代的:C1-6烷基、C1-6杂烷基、C3-6环烷基、3~6元杂环烷基、5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基;
    任选地,R6a和R6b连接在一起,形成一个任选被1、2或3个R取代的3~6元环;
    R7a和R7b分别独立地选自H、F、Cl、Br、I、OH、CN、NH2、COOH,或选自任选被1、2或3个R取代的:C1-6烷基、C1-6杂烷基、C3-6环烷基、3~6元杂环烷基、5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基;
    任选地,R7a和R7b连接在一起,形成一个任选被1、2或3个R取代的3~6元环;
    环A和环B分别独立地选自:5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基;
    R8和R9分别独立地选自卤素、羟基或选自任选被1、2或3个R取代的:C1-6烷基、C1-6杂烷基、C3-6环烷基、3~6元杂环烷基;
    m和n分别独立地选自:0、1、2或3;
    R选自F、Cl、Br、I、CN、OH、NH2、COOH,或选自任选被1、2或3个R’取代的:C1-6烷基、C1-6杂烷基、C3-6环烷基、3~6元杂环烷基、苯基和5~6元杂芳基;
    R’选自F、Cl、Br、I、OH、CN、NH2、COOH、Me、Et、CF3、CHF2、CH2F、NHCH3、N(CH3)2
    “杂”表示杂原子或杂原子团,选自-C(=O)N(R)-、-N(R)-、-C(=NR)-、-S(=O)2N(R)-、-S(=O)N(R)-、-O-、-S-、=O、=S、-O-N=、-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)-、-S(=O)2-、-N(R)C(=O)N(R)-;
    以上任何一种情况下,杂原子或杂原子团的数目分别独立地选自1、2或3。
  2. 根据权利要求1所述化合物或其药学上可接受的盐,其中,R选自F、Cl、Br、I、CN、OH、NH2、COOH,或选自任选被1、2或3个R’取代的:C1-3烷基、C1-3烷氧基、C1-3烷硫基、C1-3烷氨基和N,N-二(C1-2烷基)氨基。
  3. 根据权利要求2所述化合物或其药学上可接受的盐,其中,R选自F、Cl、Br、I、CN、OH、NH2、COOH、Me、Et、CF3、CHF2、CH2F、NHCH3、N(CH3)2
    Figure PCTCN2017082227-appb-100003
  4. 根据权利要求1~3任意一项所述化合物或其药学上可接受的盐,其中,R3和R4分别独立地选自任选被1、2或3个R取代的:C1-4烷基、C1-4杂烷基、C3-6环烷基、3~6元杂环烷基、5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基。
  5. 根据权利要求4所述化合物或其药学上可接受的盐,其中,R3和R4分别独立地选自:C1-3烷基、C1-3杂烷基、苯基、吡啶基、嘧啶基、吡嗪基、哒嗪基、呋喃基、咪唑基、恶唑基、异恶唑基、噻吩基和吡唑基。
  6. 根据权利要求5所述化合物或其药学上可接受的盐,其中,R3和R4分别独立地选自Me。
  7. 根据权利要求1~3任意一项所述化合物或其药学上可接受的盐,其中,R5选自任选被1、2或3个R取代的:C1-4烷基、C1-4杂烷基、C3-6环烷基、3~6元杂环烷基、5~6元芳基或杂芳基、5~6元芳烷基或杂芳烷基。
  8. 根据权利要求7所述化合物或其药学上可接受的盐,其中,R5选自:C1-4烷基、C3-6环烷基、3~6元杂环烷基、苯基、吡啶基、嘧啶基、吡嗪基、哒嗪基、呋喃基、咪唑基、恶唑基、异恶唑基、噻吩基和吡唑基。
  9. 根据权利要求8所述化合物或其药学上可接受的盐,其中,R5选自
    Figure PCTCN2017082227-appb-100004
    Figure PCTCN2017082227-appb-100005
  10. 根据权利要求1~3任意一项所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100006
    选自
    Figure PCTCN2017082227-appb-100007
  11. 根据权利要求10所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100008
    选自:
    Figure PCTCN2017082227-appb-100009
    Figure PCTCN2017082227-appb-100010
  12. 根据权利要求1~3任意一项所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100011
    选自
    Figure PCTCN2017082227-appb-100012
  13. 根据权利要求12所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100013
    选自:
    Figure PCTCN2017082227-appb-100014
    Figure PCTCN2017082227-appb-100015
  14. 根据权利要求1~3任意一项所述化合物或其药学上可接受的盐,其中,R6a和R6b连接在一起,形成一个任选被1、2或3个R取代的3~6元环烷基。
  15. 根据权利要求14所述化合物或其药学上可接受的盐,其中,R6a和R6b连接在一起,结构单元
    Figure PCTCN2017082227-appb-100016
    选自
    Figure PCTCN2017082227-appb-100017
  16. 根据权利要求15所述化合物或其药学上可接受的盐,其中,R6a和R6b连接在一起,结构单元
    Figure PCTCN2017082227-appb-100018
    选自
    Figure PCTCN2017082227-appb-100019
  17. 根据权利要求1~3任意一项所述化合物或其药学上可接受的盐,其中,R7a和R7b连接在一起,形成一个任选被1、2或3个R取代的3~6元环烷基。
  18. 根据权利要求17所述化合物或其药学上可接受的盐,其中,R7a和R7b连接在一起,结构单元
    Figure PCTCN2017082227-appb-100020
    选自
    Figure PCTCN2017082227-appb-100021
  19. 根据权利要求1~3任意一项所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100022
    选自
    Figure PCTCN2017082227-appb-100023
  20. 根据权利要求19所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100024
    选自:
    Figure PCTCN2017082227-appb-100025
    Figure PCTCN2017082227-appb-100026
  21. 根据权利要求20所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100027
    选自:
    Figure PCTCN2017082227-appb-100028
    Figure PCTCN2017082227-appb-100029
  22. 根据权利要求21所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100030
    选自:
    Figure PCTCN2017082227-appb-100031
    Figure PCTCN2017082227-appb-100032
  23. 根据权利要求1~3任意一项所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100033
    选自
    Figure PCTCN2017082227-appb-100034
  24. 根据权利要求23所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100035
    选自:
    Figure PCTCN2017082227-appb-100036
  25. 根据权利要求24所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100037
    选自:
    Figure PCTCN2017082227-appb-100038
  26. 根据权利要求25所述化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2017082227-appb-100039
    选自
    Figure PCTCN2017082227-appb-100040
    Figure PCTCN2017082227-appb-100041
  27. 根据权利要求1~26任意一项所述化合物或其药学上可接受的盐,其中,化合物选自
    Figure PCTCN2017082227-appb-100042
    R3、R4、R5、R6a、R7a、R8、R9如权利要求1~26所定义。
  28. 化合物或其药学上可接受的盐,其选自:
    Figure PCTCN2017082227-appb-100043
    Figure PCTCN2017082227-appb-100044
    Figure PCTCN2017082227-appb-100045
    Figure PCTCN2017082227-appb-100046
    Figure PCTCN2017082227-appb-100047
    Figure PCTCN2017082227-appb-100048
  29. 根据权利要求28所述的化合物或其药学上可接受的盐,其选自:
    Figure PCTCN2017082227-appb-100049
    Figure PCTCN2017082227-appb-100050
    Figure PCTCN2017082227-appb-100051
    Figure PCTCN2017082227-appb-100052
    Figure PCTCN2017082227-appb-100053
    Figure PCTCN2017082227-appb-100054
  30. 一种药物组合物,其含有治疗有效量的根据权利要求1~29任意一项所述的化合物或其药学上可接受的盐和药学上可接受的载体。
  31. 根据权利要求1~29任意一项所述的化合物或其药学上可接受的盐或根据权利要求30所述的药物组合物在制备治疗因IAP紊乱所致疾病的药物中的应用。
  32. 根据权利要求31所述的应用,其中,所述因IAP紊乱所致疾病选自肿瘤或乙型肝炎病毒感染。
PCT/CN2017/082227 2016-04-27 2017-04-27 作为新型二价iap拮抗剂的苯并咪唑联吲哚化合物 WO2017186147A1 (zh)

Priority Applications (8)

Application Number Priority Date Filing Date Title
RU2018141411A RU2018141411A (ru) 2016-04-27 2017-04-27 Бензимидазол-связанное индольное соединение, выполняющее функцию нового двухвалентного антагониста iap
CA3022424A CA3022424A1 (en) 2016-04-27 2017-04-27 Benzimidazole-linked indole compound acting as novel divalent iap antagonist
EP17788799.9A EP3450430A4 (en) 2016-04-27 2017-04-27 BENZIMIDAZOLE-RELATED INDOLE COMPOUND ACTING AS NEW DIVALENT IAP ANTAGONIST
AU2017258145A AU2017258145A1 (en) 2016-04-27 2017-04-27 Benzimidazole-linked indole compound acting as novel divalent IAP antagonist
CN201780023651.4A CN109071508B (zh) 2016-04-27 2017-04-27 作为新型二价iap拮抗剂的苯并咪唑联吲哚化合物
KR1020187032899A KR20180137518A (ko) 2016-04-27 2017-04-27 신규 2가 iap 길항제로서의 벤즈이미다졸-인돌 화합물
JP2018555921A JP2019514900A (ja) 2016-04-27 2017-04-27 新規な二価のiapアンタゴニストとしてのベンズイミダゾール結合インドール化合物
US16/096,848 US10508103B2 (en) 2016-04-27 2017-04-27 Benzimidazole-linked indole compound acting as novel divalent IAP antagonist

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610271073 2016-04-27
CN201610271073.3 2016-04-27

Publications (1)

Publication Number Publication Date
WO2017186147A1 true WO2017186147A1 (zh) 2017-11-02

Family

ID=60160707

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/082227 WO2017186147A1 (zh) 2016-04-27 2017-04-27 作为新型二价iap拮抗剂的苯并咪唑联吲哚化合物

Country Status (9)

Country Link
US (1) US10508103B2 (zh)
EP (1) EP3450430A4 (zh)
JP (1) JP2019514900A (zh)
KR (1) KR20180137518A (zh)
CN (1) CN109071508B (zh)
AU (1) AU2017258145A1 (zh)
CA (1) CA3022424A1 (zh)
RU (1) RU2018141411A (zh)
WO (1) WO2017186147A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112521372A (zh) * 2019-09-18 2021-03-19 南京华威医药科技集团有限公司 一种细胞凋亡蛋白抑制剂及其制备方法和用途

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008014252A2 (en) * 2006-07-24 2008-01-31 Tetralogic Pharmaceuticals Corporation Iap inhibitors
CN101516904A (zh) * 2006-07-24 2009-08-26 泰特拉洛吉克药业公司 二聚的iap拮抗剂
CN102471275A (zh) * 2009-07-02 2012-05-23 泰特拉洛吉克药业公司 Smac模拟物
US20140303090A1 (en) * 2013-04-08 2014-10-09 Tetralogic Pharmaceuticals Corporation Smac Mimetic Therapy

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2598995C (en) 2005-02-25 2014-07-15 Stephen M. Condon Dimeric iap inhibitors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008014252A2 (en) * 2006-07-24 2008-01-31 Tetralogic Pharmaceuticals Corporation Iap inhibitors
CN101516904A (zh) * 2006-07-24 2009-08-26 泰特拉洛吉克药业公司 二聚的iap拮抗剂
CN102471275A (zh) * 2009-07-02 2012-05-23 泰特拉洛吉克药业公司 Smac模拟物
US20140303090A1 (en) * 2013-04-08 2014-10-09 Tetralogic Pharmaceuticals Corporation Smac Mimetic Therapy

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3450430A4 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112521372A (zh) * 2019-09-18 2021-03-19 南京华威医药科技集团有限公司 一种细胞凋亡蛋白抑制剂及其制备方法和用途
WO2021051827A1 (zh) * 2019-09-18 2021-03-25 南京华威医药科技集团有限公司 一种细胞凋亡蛋白抑制剂及其制备方法和用途

Also Published As

Publication number Publication date
CN109071508A (zh) 2018-12-21
EP3450430A1 (en) 2019-03-06
JP2019514900A (ja) 2019-06-06
CA3022424A1 (en) 2017-11-02
AU2017258145A1 (en) 2018-12-13
CN109071508B (zh) 2021-09-03
KR20180137518A (ko) 2018-12-27
US10508103B2 (en) 2019-12-17
EP3450430A4 (en) 2019-12-11
US20190135794A1 (en) 2019-05-09
RU2018141411A (ru) 2020-05-27

Similar Documents

Publication Publication Date Title
CN109952299B (zh) 用于制备蛋白降解靶向嵌合体的氟代羟脯氨酸衍生物
CN107438611B (zh) 稠环化合物、其药物组合物及应用
WO2015124063A1 (zh) 丙肝病毒抑制剂及其制药用途
ES2956090T3 (es) Inhibidores de bencimidazol de enzimas PAD
EP3333157A1 (en) Vinyl compounds as fgfr and vegfr inhibitors
JP7483678B2 (ja) Pad4阻害剤としての置換ベンズイミダゾール
WO2007069635A1 (ja) Vla-4阻害薬
CN110461855B (zh) 氮杂环丁烷衍生物
EP3171941B1 (en) N-methyl-d-aspartate receptor modulators and methods of making and using same
MX2014004705A (es) Procedimientos para la preparacion de derivados de bencimidazol novedosos.
CN109661391B (zh) S1p1激动剂及其应用
EP4100405A1 (en) Macrocyclic pad4 inhibitors useful as immunosuppressant
JP7257397B2 (ja) Iap阻害剤として有用なsmac模倣物及びその用途
CN109071508B (zh) 作为新型二价iap拮抗剂的苯并咪唑联吲哚化合物
WO2011090317A2 (en) Imidazopyrazinone derivatives with apoptosis inducing activity on cells
EP3498715B1 (en) Anti-hcmv virus compound
EA038176B1 (ru) Производные пирроло-[1,2,5]бензотиадиазепина и их применение для лечения заболевания, вызванного вирусом гепатита в

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2018555921

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3022424

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20187032899

Country of ref document: KR

Kind code of ref document: A

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

Ref document number: 17788799

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017788799

Country of ref document: EP

Effective date: 20181127

ENP Entry into the national phase

Ref document number: 2017258145

Country of ref document: AU

Date of ref document: 20170427

Kind code of ref document: A