WO2018196768A1 - Heterocyclic compound serving as pd-l1 inhibitor - Google Patents

Heterocyclic compound serving as pd-l1 inhibitor Download PDF

Info

Publication number
WO2018196768A1
WO2018196768A1 PCT/CN2018/084352 CN2018084352W WO2018196768A1 WO 2018196768 A1 WO2018196768 A1 WO 2018196768A1 CN 2018084352 W CN2018084352 W CN 2018084352W WO 2018196768 A1 WO2018196768 A1 WO 2018196768A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
tert
amino
preparation
Prior art date
Application number
PCT/CN2018/084352
Other languages
French (fr)
Chinese (zh)
Inventor
王勇
赵立文
刘欣
尹伟
刘笑
于琪
Original Assignee
南京圣和药业股份有限公司
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 南京圣和药业股份有限公司 filed Critical 南京圣和药业股份有限公司
Publication of WO2018196768A1 publication Critical patent/WO2018196768A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic 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
    • A61K31/4245Oxadiazoles
    • 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
    • A61K31/433Thidiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/061,2,4-Oxadiazoles; Hydrogenated 1,2,4-oxadiazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/101,3,4-Oxadiazoles; Hydrogenated 1,3,4-oxadiazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D285/00Heterocyclic compounds containing rings having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by groups C07D275/00 - C07D283/00
    • C07D285/01Five-membered rings
    • C07D285/02Thiadiazoles; Hydrogenated thiadiazoles
    • C07D285/04Thiadiazoles; Hydrogenated thiadiazoles not condensed with other rings
    • C07D285/081,2,4-Thiadiazoles; Hydrogenated 1,2,4-thiadiazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the present invention belongs to the field of medical chemistry, and relates to a class of heterocyclic compounds as PD-L1 inhibitors and uses thereof.
  • the present invention relates to a compound represented by formula A or an isomer thereof, a pharmaceutically acceptable salt, Solvates or prodrugs, methods for their preparation, and pharmaceutical compositions containing these compounds and the use of such compounds or compositions for the treatment of cancer or infectious diseases.
  • PD-1 Programmed Cell Death-1
  • B7 ⁇ H1 ligand PD-L1
  • PD-1 is mainly expressed on the membrane surface of T cells, B cells, and natural killer cells (NK cells).
  • PD-L1 is mainly expressed on mature CD4T cells, CD8 T cells, B cells, monocytes, and dendrites.
  • Hematopoietic cells such as dendritic cells (DCs), macrophages, and some non-hematopoietic cells, such as membrane surfaces of endothelial cells, islet cells, mast cells, and the like.
  • DCs dendritic cells
  • macrophages macrophages
  • non-hematopoietic cells such as membrane surfaces of endothelial cells, islet cells, mast cells, and the like.
  • PD-L1 is highly expressed in various tumors, such as lung cancer, gastric cancer, multiple bone marrow, melanoma and breast cancer.
  • the expression of PD-L1 on the surface of tumor cells interacts with the ligand on the surface of T cells, which can induce apoptosis of T cells or reduce the reactivity of T cells, thereby inhibiting the tumor immune response and allowing tumor cells to escape immune attack. Therefore, blocking the antagonist of PD1-PDL1 signaling pathway can promote the activation of T cells, reverse the tumor immune microenvironment, and enhance the endogenous anti-tumor immune effect.
  • Targeting PD-1/PD-L1 inhibitors has broad application prospects in the field of tumor immunotherapy.
  • biomacromolecules also have some disadvantages such as immunogenicity and limitations of the route of administration. Therefore, there is still a need to develop targeted PD-1/PD-L1 inhibitors with better pharmacodynamics.
  • the inventors of the present invention have found that a class of small molecule drugs can specifically modulate and/or mediate the transduction of PD-L1 and its related protein kinases for the treatment of diseases associated with PD-1/PD-L1.
  • X and Y are each independently selected from N, O and S, and when X is N, Y is selected from O and S; when X is O or S, Y is N; when Y is N, X is selected from O and S; When Y is O or S, X is N;
  • R 1 is selected from the side chains of the amino acids Ser and Thr;
  • R 2 is selected from the group consisting of the amino acid Ser and Thr residues
  • R 3 is selected from the group consisting of H, —(CH 2 ) m C(O)OR 4 , —(CH 2 ) m C(O)N(R 5 )(R 6 ) and —(CH 2 ) m CN, wherein R 4 , R 5 , R 6 are each independently selected from H and alkyl, and m is 0, 1, 2, 3 or 4;
  • n 1, 2, 3 or 4.
  • a compound of Formula A, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof which is represented by Formula I or II a compound or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug:
  • Q is selected from O and S; and R 1 , R 2 and R 3 are as defined in Formula A.
  • a compound of Formula A, Formula I or II, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, according to the invention wherein:
  • R 1 is selected from the side chains of the amino acids Ser and Thr;
  • R 2 is selected from the group consisting of the amino acid Ser and Thr residues
  • R 3 is selected from the group consisting of H, —(CH 2 ) m C(O)OR 4 , —(CH 2 ) m C(O)N(R 5 )(R 6 ) and —(CH 2 ) m CN, wherein R 4 , R 5 and R 6 are each independently selected from H and C 1-6 alkyl, and m is 1, 2, 3 or 4;
  • n 1, 2, 3 or 4.
  • a compound of Formula A, Formula I or II, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, according to the invention wherein:
  • R 1 is selected from
  • R 2 is selected from
  • R 3 is selected from the group consisting of H, —(CH 2 ) m C(O)OR 4 , —(CH 2 ) m C(O)N(R 5 )(R 6 ) and —(CH 2 ) m CN, wherein R 4 , R 5 and R 6 are each independently selected from H and C 1-6 alkyl, and m is 1, 2, 3 or 4;
  • n 1, 2, 3 or 4.
  • a compound of Formula A, Formula I or II, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, according to the invention wherein:
  • R 1 is selected from
  • R 2 is selected from
  • R 3 is selected from H, And n is 1, 2, 3 or 4.
  • a compound of Formula A, Formula I or II, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, according to the invention wherein:
  • R 1 is selected from
  • R 2 is selected from with
  • a compound having the formula A or a stereoisomer thereof selected from A compound having the formula A or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, non-limiting examples of which include:
  • Another object of the present invention is to provide a compound of the formula B or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof,
  • X and Y are each independently selected from N, O and S, and when X is N, Y is selected from O and S; when X is O or S, Y is N; when Y is N, X is selected from O and S; When Y is O or S, X is N;
  • R 7 is selected from the side chain of the amino acids Asn, Gln, Asp, Glu;
  • R 8 is selected from the group consisting of the amino acid Ser and Thr residues
  • R 9 and R 10 are each independently selected from H, an alkyl group and an alkylhydroxy group, and R 9 and R 10 are not simultaneously H, and
  • R 9 or R 10 together with the amino group attached to its alpha carbon position form a nitrogen heterocycloalkyl or a nitrogen heterocycloalkyl group, optionally a one or more Substituted with a substituent selected from the group consisting of a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a halogen, and a nitro group; or
  • R 9 and R 10 together with the C atom to which they are attached form a cycloalkyl or oxacycloalkyl group, which is optionally selected from one or more selected from the group consisting of a hydroxyl group and a hydroxyalkyl group. Substituted by a substituent of a cyano group, an amino group, a halogen or a nitro group.
  • a compound of the formula B, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof is a compound of the formula III or IV.
  • Q is selected from O and S, and R 7 , R 8 , R 9 and R 10 are as defined in Formula B.
  • the compound of Formula B or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal, or prodrug thereof, according to the present invention, wherein:
  • R 7 is selected from the side chain of the amino acids Asn, Gln, Asp, Glu;
  • R 8 is selected from the group consisting of the amino acid Ser and Thr residues
  • R 9 and R 10 are each independently selected from the group consisting of H, C 1-6 alkyl and C 1-6 alkyl hydroxy, and R 9 and R 10 are not H at the same time, and
  • R 9 or R 10 together with the amino group attached to its alpha carbon position form an aza C 3-6 cycloalkyl or a nitrogenoxa C 3-6 cycloalkyl group, said aza C 3-6 cycloalkyl or nitrox a hetero C 3-6 cycloalkyl group optionally substituted with one or more substituents selected from the group consisting of hydroxyl, hydroxy C 1-6 alkyl, cyano, amino, halogen, nitro;
  • R 9 and R 10 together with the C atom to which they are attached form a C 3-6 cycloalkyl or oxa C 3-6 cycloalkyl group, said C 3-6 cycloalkyl or oxa C 3-6 cycloalkane
  • the group is optionally substituted with one or more substituents selected from the group consisting of a hydroxyl group, a hydroxy C 1-6 alkyl group, a cyano group, an amino group, a halogen, and a nitro group.
  • a compound of the formula B, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof wherein R 9 is H, R 10 is amino group formed on the ⁇ -carbon position with C 3-6 cycloalkyl aza, aza said C 3-6 cycloalkyl optionally substituted with one or more substituents selected from hydroxy, hydroxy C 1-6 alkyl, Substituted by a substituent of a cyano group, an amino group, a halogen or a nitro group.
  • a compound of the formula B, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof wherein R 9 is selected from C 1-3 An alkyl group, R 10 is selected from C 1-3 alkyl groups, and R 9 and R 10 together with the C atom to which they are attached form a C 3-6 cycloalkyl group, optionally a C 3-6 cycloalkyl group Or a plurality of substituents selected from the group consisting of a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a halogen, and a nitro group.
  • a compound of the formula B, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof wherein R 9 is selected from C 1-3 An alkyl group, R 10 is selected from a C 1-3 hydroxyalkyl group, and R 9 and R 10 together with the C atom to which they are attached form an oxa C 3-6 cycloalkyl group, said oxa C 3-6 cycloalkyl group It is optionally substituted with one or more substituents selected from the group consisting of a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a halogen, and a nitro group.
  • Another object of the present invention is to provide a compound of the formula C or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug,
  • X and Y are each independently selected from N, O and S, and when X is N, Y is selected from O and S; when X is O or S, Y is N; when Y is N, X is selected from O and S; When Y is O or S, X is N;
  • R 11 is selected from the side chains of the amino acids Ser and Thr;
  • R 12 is selected from the group consisting of amino acid Asn, Gln, Asp, Glu side chain;
  • R 15 is selected from the group consisting of H and alkyl
  • R 13 and R 14 are each independently selected from H and alkyl, and R 13 and R 14 are not H at the same time, and
  • R 13 , R 14 together with the C atom to which they are attached form a cycloalkyl group
  • R 13 or R 14 together with the imino group to which the ⁇ carbon position is bonded forms a nitrogen heterocycloalkyl group, or R 14 together with R 15 and the atom to which they are attached form an optionally substituted lactone cycloalkyl group.
  • cycloalkyl, azacycloalkyl or lactone cycloalkyl is optionally substituted with one or more substituents selected from the group consisting of hydroxyl, hydroxyalkyl, cyano, amino, halogen, nitro;
  • a compound of the formula C, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof is a compound of the formula V or VI.
  • Q is selected from O and S; and R 11 , R 12 , R 13 , R 14 and R 15 are as defined in Formula C.
  • the attached imino groups together form an aza C 3-6 cycloalkyl group
  • a compound of the formula C, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof wherein R 14 and R 15 and The attached atoms together form a lactone C 3-6 cycloalkyl group which is substituted by one or more selected from the group consisting of hydroxyl, oxo, hydroxyalkyl, cyano, amino, halogen, nitro Substituted by
  • Another object of the present invention is to provide a process for the preparation of a compound of the formula I, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, comprising the steps of:
  • a compound of the formula ia and a compound of the formula ib are subjected to a condensation reaction to form a compound of the formula ic;
  • Pg 1 represents a protecting group of R 1
  • Pg 2 represents a protecting group of R 2
  • R 1 and R 2 are each selected from the group consisting of the amino acid Ser and Thr side chains, in order to avoid the amino acid Ser or Thr side chain
  • a conventional hydroxy protecting group may be used to protect the amino acid Ser or Thr side chain
  • Pg 1 and Pg 2 are preferably tert-butyl ( t Bu);
  • Pg 3 represents a protecting group of R 3 or is absent; as described in Formula I, R 3 is selected from H, -(CH 2 ) m C(O)OR 4 , -(CH 2 ) m C(O)N ( R 5 )(R 6 ) and —(CH 2 ) m CN; when R 3 is a group containing a hydroxyl group or an amino group which is susceptible to change in further reaction, it can be protected by a conventional protecting group; Pg 4 Pg 5 represents an amino protecting group.
  • Another object of the present invention is to provide a process for the preparation of a compound of the formula II, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, comprising the steps of:
  • a compound of the formula ia and a compound of the formula iib are subjected to a condensation reaction to form a compound of the formula iic;
  • Pg 1 represents a protecting group for R 1
  • Pg 2 represents a protecting group for R 2
  • R 1 and R 2 are each selected from the group consisting of the amino acid Ser and Thr side chains, in order to avoid the amino acid Ser or Thr side chain Further reaction in the reaction, the conventional hydroxyl group can be used to protect the hydroxyl group
  • Pg 1 , Pg 2 is preferably tert-butyl ( t Bu);
  • Pg 3 represents the protection or absence of R 3 ; as described in Formula II, R 3 is selected from H, -(CH 2 ) m C(O)OR 4 , -(CH 2 ) m C(O)N(R 5 ) (R 6 ) and -(CH 2 ) m CN; when R 3 is a group which contains a hydroxyl group or an amino group and is susceptible to change in further reaction, it can be protected by a conventional protecting group;
  • Pg 4 and Pg 5 represent an amino protecting group.
  • Another object of the present invention is to provide a compound of Formula B, Formula C, Formula III, Formula IV, Formula V or Formula VI or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate thereof,
  • Formula III, Formula IV, Formula V or Formula VI is a compound of Formula B, Formula C, Formula III, Formula IV, Formula V or Formula VI.
  • Another object of the present invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising, for example, Formula A, Formula B, Formula C, Formula I, Formula II, Formula III, Formula IV, A compound of the formula V or formula VI, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, and a pharmaceutically acceptable carrier.
  • Another object of the present invention is to provide a compound of the formula A, formula B, formula C, formula I, formula II, formula III, formula IV, formula V, formula VI. Or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof or a pharmaceutical composition comprising the same for use in the manufacture of a medicament for the treatment of cancer or an infectious disease.
  • a compound of formula A, formula B, formula C, formula I, formula II, formula III, formula IV, formula V or formula VI Or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, or a pharmaceutical composition comprising the same, for use in the manufacture of a medicament for the treatment of a cancer or an infectious disease
  • the cancer comprises But not limited to melanoma, brain tumors (gliomas with malignant astroglia and oligodendroglioma), esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer (colon cancer) , rectal cancer, etc.), lung cancer (non-small cell lung cancer, small cell lung cancer, primary or metastatic squamous cell carcinoma, etc.), kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, Osteochondroma, osteoma, osteosarcoma, seminoma, testicular
  • a formula of formula A, formula B, formula C, formula I, formula II, formula III, formula IV, formula V or formula VI Use of a compound, or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, or a pharmaceutical composition comprising the same, in the manufacture of a medicament for the treatment of a cancer or an infectious disease, wherein the infection Sexual diseases include, but are not limited to, bacterial, viral, and fungal infections.
  • a compound of Formula A, Formula B, Formula C, Formula I, Formula II, Formula III, Formula IV, Formula V, or Formula VI It has a significant inhibitory effect on colon cancer.
  • Haldrogen and “carbon” in the compounds of the present invention include all isotopes thereof. Isotopes are understood to include those atoms having the same number of atoms but having different mass numbers, such as isotopes of hydrogen including lanthanum and cerium, and isotopes of carbon including 13 C and 14 C.
  • stereoisomer refers to a molecule having the same atomic composition and attachment in the same manner, but having a three-dimensional arrangement differently, including optical isomers, geometric isomers (also known as cis-trans isomers), "chiral” It is a molecule that has properties that cannot overlap with its mirror image; “non-chiral” refers to a molecule that can overlap with its mirror image.
  • Optical isomers are divided into enantiomers and diastereomers.
  • Enantiomer refers to two isomers of a compound that are not superimposable but are mirror images of each other.
  • Diastereomer refers to a stereoisomer that has two or more chiral neutralities and whose molecules are not mirror images of each other. Diastereomers have different physical properties such as melting point, boiling point, spectral properties and reactivity. The mixture of diastereomers can be separated by high resolution analytical procedures such as electrophoresis and chromatography, such as HPLC.
  • All stereoisomeric forms of the compounds of the invention include, but are not limited to, diastereomers, enantiomers, cis and trans isomers, and mixtures thereof, such as racemic mixtures.
  • Many organic compounds exist in optically active forms, that is, they have the ability to rotate planes of plane polarized light.
  • the prefix D, L or R, S is used to indicate the absolute configuration of the molecular chiral center.
  • the prefix D, L or (+), (-) is used to designate the sign of the plane-polarized light rotation of the compound, (-) or L means that the compound is left-handed, and the prefix (+) or D means that the compound is right-handed.
  • stereoisomers are the same, but their stereostructures are different.
  • a particular stereoisomer can be an enantiomer, and a mixture of isomers is often referred to as a mixture of enantiomers.
  • the 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may result in no stereoselectivity or stereospecificity during the chemical reaction.
  • racemic mixture and “racemate” refer to an equimolar mixture of two enantiomers that lack optical activity.
  • the compounds of the invention may be one of the possible isomers or mixtures thereof, such as racemates and mixtures of diastereomers (depending on the number of asymmetric carbon atoms) The form exists.
  • Optically active (R)- or (S)-isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
  • the resulting mixture of any stereoisomers can be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, for example, by chromatography, depending on the difference in physicochemical properties of the components. Method and / or step crystallization.
  • the "pharmaceutically acceptable salt” of the present invention means an acid and/or a basic salt of a compound with an inorganic and/or organic acid and a base form, and also a zwitterionic salt (internal salt).
  • the compound of the present invention contains an amino acid side chain and an amino acid residue, so that it can form an internal salt or a salt corresponding to other inorganic inorganic and/or organic acid and base forms.
  • Crystal as used in the present invention refers to various solid forms formed by the compounds of the present invention, including crystalline forms and amorphous forms.
  • the "prodrug” of the present invention refers to a compound which is converted into a compound of the present invention by a reaction with an enzyme, gastric acid or the like under physiological conditions of a living body, that is, a compound which is converted into the invention by oxidation, reduction, hydrolysis or the like of the enzyme and/or A compound which is converted into a compound of the invention by a hydrolysis reaction such as gastric acid or the like.
  • alkyl group of the present invention means a linear or branched saturated hydrocarbon group, preferably a C 1-8 alkyl group, more preferably a C 1-6 alkyl group.
  • alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropane 1,1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl -2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-Dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl
  • Cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic substituent comprising from 3 to 20 carbon atoms, preferably from 4 to 13 carbon atoms.
  • Non-limiting examples of monocycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene
  • the group, the cyclooctyl group and the like are preferably a cyclopentyl group or a cyclohexyl group.
  • Polycycloalkyl groups include spiro, fused, and bridged cycloalkyl groups.
  • the "azacycloalkylene group" of the present invention means a saturated ring comprising a nitrogen atom and a plurality of carbon atoms, and the azacycloalkylene group is preferably aza C 3-20 cycloalkyl group, more preferably aza C 4 -13 cycloalkyl. More preferably, it is an aza C 3-6 cycloalkyl group.
  • Non-limiting examples of azacycloalkyl groups include pyrrolidinyl and piperidinyl groups.
  • the "oxyheterocycloalkyl group” of the present invention means a saturated ring composed of one oxygen and a plurality of carbon atoms.
  • the oxacycloalkyl group is preferably an oxa C 3-20 cycloalkyl group, more preferably an oxa C 4-13 cycloalkyl group. More preferably, it is an oxa C 3-6 cycloalkyl group.
  • lactone cycloalkyl group of the present invention means an ester group A saturated ring composed of multiple carbon atoms.
  • the lactone cycloalkyl group is preferably a lactone C 3-20 cycloalkyl group, more preferably a lactone C 4-13 cycloalkyl group. More preferred is a lactone C 3-6 cycloalkyl group.
  • alkyl hydroxy group of the present invention means -alkyl-OH.
  • halogen of the present invention means fluorine, chlorine, bromine or iodine.
  • the "nitro group" of the present invention means -NO 2 .
  • the "cyano group" of the present invention means -CN.
  • the hydroxy "hydroxy group" of the present invention means -OH.
  • the "benzyl group” of the present invention means -CH 2 -phenyl group, abbreviated as "Bn”.
  • Tet-butoxycarbonyl group in the present invention means - (O) CO (t Bu ), abbreviated as “Boc.”
  • the "Fmoc-" of the present invention means a fluorenylmethoxycarbonyl group
  • L-amino acid of the present invention means that the ⁇ -carbon atom of the ⁇ -amino acid is left-handed; conversely, the "D-amino acid” refers to the ⁇ of the general structure CH(COOH)(NH 2 )-side chain - The carbon atom is right-handed.
  • the ⁇ -carbon atoms of other protein amino acids are asymmetric carbon atoms (ie, the four substituents bonded to the ⁇ -carbon atom are different), so the amino acids may have stereoisomers, ie, may be different Configuration (D-type and L-type two configurations).
  • Non-limiting examples of amino acids include alanine (Ala), arginine (Arg), asparagine (Asn), aspartic acid (Asp), cysteine (Cys), glutamine (Gln) , glutamic acid (Glu), glycine (Gly), histidine (His), isoleucine (Ile), leucine (Leu), lysine (Lys), methionine (Met), phenylalanine Acid (Phe), proline (Pro), serine (Ser), threonine (Thr), tryptophan (Trp), tyrosine (Tyr), valine (Val).
  • amino acid side chain of the present invention means a component of an amino acid, and the amino acid structural formula is represented by CH(COOH)(NH 2 )-R, and R represents an amino acid side chain, for example, the structure of alanine is CH (COOH).
  • (NH 2 )-CH 3 the amino acid side chain is -CH 3 ;
  • the structure of serine is CH(COOH)(NH 2 )-CH 2 OH, the amino acid side chain is -CH 2 OH;
  • the structure of threonine is CH(COOH)(NH 2 )-CH(OH)(CH 3 ), the amino acid side chain is -CH(OH)(CH 3 ).
  • amino acid residue of the present invention means that a part of the structure is absent compared to the structure of the parent amino acid, and is an incomplete amino acid.
  • a hydrogen atom on the amino group of the amino acid is replaced by a chemical bond to bond with another atom, or an amino acid.
  • -OH is replaced by a chemical bond to bond with other atoms.
  • Such as the structure of alanine Then its amino acid residue can be Can also be
  • the "protecting group" of the present invention is such that the nitrogen atom and the oxygen atom remain unchanged in the reaction of other parts of the molecule, and are protected by a group which is easy to remove.
  • the protecting group of the present invention includes an amino protecting group and a hydroxy protecting group, and the amino protecting group of the present invention means a group which prevents or prevents the amino group from participating in the next reaction until the protecting group is removed, and a non-limiting example includes a formyl group, an alkyl group.
  • the hydroxy protecting group of the present invention means a group which prevents or prevents the hydroxyl group from participating in the next reaction until the protecting group is removed.
  • hydroxy protecting group examples include acetyl, allyl, benzoyl, benzyl, ⁇ -methoxyethoxymethyl, methoxymethyl, dimethoxytrityl [di-( 4-methoxyphenyl)phenylmethyl], methoxytriphenyl[(4-methoxyphenyl)diphenylmethyl], p-methoxybenzyl ether, methylthiomethyl , trimethylacetyl, tetrahydropyranyl, triphenylmethyl, silicon (eg, trimethylsilyl, tert-butyldimethylsilyl, tert-butyldiphenylsilyl, triiso) Propylsilyloxymethyl and triisopropylsilyl).
  • alkyl groups such as methyl and t-butyl groups, as well as other ethers such as ethoxyethyl groups.
  • NMM N-methylmorpholine
  • HATU 2-(7-oxobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • TPP triphenylphosphine
  • EDC 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • TBDMSCl tert-butyldimethylchlorosilane
  • Fmoc-Asn(Trt)-OH N-fluorenylmethoxycarbonyl-L-asparagine.
  • Step 2 Preparation of benzyl 1-((tert-butoxycarbonyl)amino)-3-oxocyclobutane-1-carboxylate
  • Step 3 Preparation of benzyl 1-((tert-butoxycarbonyl)amino)-3-(2-ethoxy-2-oxoethylidene)cyclobutane-1-carboxylate
  • Step 4 Preparation of benzyl 1-amino-3-(2-ethoxy-2-oxoethylidene)cyclobutane-1-carboxylate
  • Step 5 1-(((9H- ⁇ -9-yl)methoxy)carbonyl)amino)-3-(2-ethoxy-2-oxoethylidene)cyclobutane-1-carboxylic acid Preparation of benzyl ester
  • Step 6 1-(((9H- ⁇ -9-yl)methoxy)carbonyl)amino)-3-(2-ethoxy-2-oxoethyl)cyclobutane-1-carboxylic acid preparation
  • Step 7 2-(3-(((9H- ⁇ -9-yl)methoxy)carbonyl)amino)-3-(2-(N-(tert-butoxy)-O-(tert-butyl) Preparation of ethyl-L-betathreonyl)nonyl-1-carbonyl)cyclobutyl)acetate
  • Step 8 2-(3-(((9H- ⁇ -9-yl)methoxy)carbonyl)amino)-3-(5-((1S,2S)-2-(tert-butoxy)- Preparation of ethyl 1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazol-2-yl)cyclobutyl)acetate
  • Step 9 2-(3-Amino-3-(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3 Of 4-(oxadiazol-2-yl)cyclobutyl)acetate
  • Step 10 Preparation of O-(tert-butyl)-N-((4-nitrophenoxy)carbonyl)-L-serine tert-butyl ester
  • Step 11 N-((1-(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxole
  • tert-butyl 2-oxazol-2-yl -3-(2-ethoxy-2-oxoethyl)cyclobutyl)carbamoyl-O-(tert-butyl)-L-serine
  • Step 12 N-((1-(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxole Preparation of oxazol-2-yl)-3-(carboxy)cyclobutyl)carbamoyl-O-(tert-butyl)-L-serine
  • Step 13 ((1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)-3-(carboxymethyl) Preparation of cyclobutyl)carbamoyl)-L-serine
  • Step 1 Preparation of ethyl 3-(2-amino-2-oxoethylidene)-1-((tert-butoxycarbonyl)amino)cyclobutane-1-carboxylate
  • Step 2 Preparation of ethyl 3-(2-amino-2-oxoethyl)-1-((tert-butoxycarbonyl)amino)cyclobutane-1-carboxylate
  • Step 3 Preparation of 3-(2-amino-2-oxoethyl)-1-((tert-butoxycarbonyl)amino)cyclobutanecarboxylic acid
  • Step 5 ((1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)-3-(2-amino-) Preparation of 2-oxoethyl)cyclobutyl)carbamoyl)-L-serine
  • the title compound was obtained according to the procedure of Steps 5, 7, 8, 9, 11, and 13 in Example 1, except that the starting material in the step 5 was 1-amino-3-(2-ethoxy-2-oxo Benzylene) benzyl cyclobutanecarboxylate (50 mg, 0.17 mmol) was dissolved in 1,4-dioxane to 3-(2-amino-2-oxoethyl)-1-carboxycyclobutylamine Hydrochloride.
  • the title compound was obtained in a similar manner to the method of the step 3 in Example 1, except that the starting material (triphenylphosphine) ethyl acetate was replaced with (triphenylphosphine) acetonitrile.
  • Step 2 Preparation of (1s,3s)-1-((tert-butoxycarbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylate
  • Step 3 Preparation of (1s,3s)-1-((tert-butoxycarbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylic acid
  • the title compound was obtained according to the procedure of Step 3 in Example 3, except that the starting material ethyl 3-(2-amino-2-oxoethyl)-1-((tert-butoxycarbonyl)amino)cyclobutane
  • the ethyl alkanoate was replaced with the ethyl ester of (1s, 3s)-1-((tert-butoxycarbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylate obtained in the above step 2.
  • Step 5 (((1s,3R)-1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)-3 Preparation of -(cyanomethyl)cyclobutyl)carbamoyl)-L-serine
  • the title compound was obtained according to the procedure of Steps 5, 7, 8, 9, 11, and 13 in Example 1, except that the starting material of the first step of Example 1 was 1-amino-3-(2-ethoxy-
  • the benzyl 2-oxoethylidenecyclobutanecarboxylate was replaced with the product of the above step 4 (1s, 3s)-1-cyano-3-(cyanomethyl)cyclobutylamine hydrochloride.
  • Step 1 1 - Preparation of ((benzyloxy)carbonyl)-3-oxocyclobutaneamine hydrochloride
  • Step 2 Preparation of benzyl 1-((((9H- ⁇ -9-yl)methoxy)carbonyl)amino)-3-oxocyclobutanecarboxylate
  • the title compound was obtained according to the procedure of Step 5 in Example 1, except that the starting material of the starting material of Example 1 was 1-amino-3-(2-ethoxy-2-oxoethylidene)cyclobutane.
  • the benzyl carbamate was replaced with the 1-((benzyloxy)carbonyl)-3-oxocyclobutaneamine hydrochloride obtained in the above step 1.
  • Step 3 Benzyl 1-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-(2-(tert-butoxy)-2-oxoethylidene)cyclobutane Preparation of alkacrylate
  • Step 4 1-(((9H-Indol-9-yl)methoxy)carbonyl)amino)-3-(2-(tert-butoxy)-2-oxoethyl)cyclobutanecarboxylic acid preparation
  • the title compound was obtained according to the procedure of Step 6 in Example 1, except that the starting material benzyl 1-((((9H- -9) yl)) oxy)) Ethoxy-2-oxoethylidene)cyclobutanecarboxylate is replaced by the benzyl 1-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)- 3-(2-(tert-Butoxy)-2-oxoethylidene)cyclobutanecarboxylate.
  • Step 5 (R,Z)-2-(3-(((9H- ⁇ -9-yl)methoxy)carbonyl)amino)-3-(4-amino-5-(tert-butoxymethyl) Of tert-butyl 9-dimethyl-7-oxo-2,8-dioxa-3,6-diazacyclo-3-ene-1-yl)cyclobutyl)acetate
  • Step 6 (R)-2-(3-((((9H- ⁇ -9-yl))methoxy)carbonyl)amino)-3-(3-(2-(tert-butoxy)-1) Preparation of tert-butyl (-(tert-butoxycarbonyl)amino)ethyl)-1,2,4-oxadiazol-5-yl)cyclobutyl)acetate
  • Step 7 ((1-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-(carboxymethyl)cyclobutane Preparation of carbamoyl)-L-pyrethine
  • Example 7 ((1-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-(2-amino-2) -Oxoethyl)cyclobutyl)carbamoyl)-L-pyrethine
  • Step 1 (R)-(1-(5-(1-(((9H- ⁇ -9-yl))methoxy)carbonyl)amino)-3-(3-(2-amino-2-oxo) Preparation of tert-butyl ester of ethyl)cyclobutyl)-1,2,4-oxadiazol-3-yl)-2-(tert-butoxy)ethyl)carbamate
  • the title compound was obtained according to the procedure of Steps 5 and 6 in Example 6 except that the starting material was 1-((((9H-in-9-yl)methoxy)carbonyl)amino)-3-(2-) Replacement of (tert-butoxy)-2-oxoethyl)cyclobutanecarboxylic acid with 3-(2-amino-2-oxoethyl)-1-((tert-butoxycarbonyl)amino)cyclobutane Formic acid.
  • Step 2 ((1-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-(2-amino-2-) Preparation of oxoethyl)cyclobutyl)carbamoyl)-L-pyrethine
  • Step 1 Preparation of (9H-fluoren-9-yl)methyl-((1r,3r)-3-(cyanomethyl)-1-(fluorocarbonyl)cyclobutyl)carbamate
  • Step 2 ((R)-1-(5-((1r,3R)-1-(((9H- ⁇ -9-yl)methoxy)carbonyl)amino)-3-(cyanomethyl) Of tert-butyl)-1,2,4-oxadiazol-3-yl)2-(tert-butoxy)ethyl)carbamic acid tert-butyl ester
  • the title compound was obtained according to the procedure of Steps 5 and 6 in Example 6 except that the starting material was 1-((((9H-in-9-yl)methoxy)carbonyl)amino)-3-(2-) Replacement of (tert-butoxy)-2-oxoethyl)cyclobutanecarboxylic acid with (9H-fluoren-9-yl)methyl(3-(cyanomethyl)-1-(fluorocarbonyl)cyclobutane Carbamate.
  • Step 3 (((1R,3S)-1-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-( Preparation of cyanomethyl)cyclobutyl)carbamoyl)-L-pyrethine
  • the title compound was obtained by the procedure of Steps 9, 11, and 13 in Example 1. The difference is that the starting material is 2-(3-(((9H- ⁇ -9-yl)methoxy)carbonyl)amino)-3-(5-((1S,2S)-2-(tert-butoxy) ) ethyl-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazol-2-yl)cyclobutyl)acetate was replaced by ((R)-1-(5) -((1r,3R)-1-(((9H- ⁇ -9-yl)methoxy)carbonyl)amino)-3-(cyanomethyl)cyclobutyl)-1,2,4- Tert-butyl ester of oxadiazol-3-yl)2-(tert-butoxy)ethyl)carbamate.
  • Step 7 (2S,4R)-2-(5-((6S,10S)-10-(tert-butoxymethyl)-13,13-dimethyl-3,8,11-trioxo-1, 1,1-triphenyl-12-oxy-2,7,9-azatetradecane-6-yl)-1,3,4-oxadiazol-2-yl)-4-((uncle Preparation of tert-butyl butyl dimethylsilyl)oxy)tetrahydropyrrole-1-carboxylate
  • Step 8 (((S)-4-Amino-1-(5-((2S,4R)-4-hydroxytetrahydropyrrole-2-yl)-1,3,4-oxadiazol-2-yl) Preparation of -4-oxobutyl)carbamoyl)-L-serine
  • Methyl (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylate (4.4 g, 30.3 mmol, 1 eq) was dissolved in tetrahydrofuran (40 mL), and aqueous sodium hydrogencarbonate (8.9 g, 106.1 Methyl acetate (3.5 eq, dissolved in 40 mL of water) and di-tert-butyl dicarbonate (6.28 g, 28.8 mmol, 0.95 eq) was added dropwise at 0 ° C, and then stirred at room temperature for 24 h. Water and ethyl acetate were added to the mixture, and the mixture was evaporated.
  • Step 3 (((S)-4-Amino-1-(5-((2S,3S)-3-hydroxytetrahydropyrrole-2-yl)-1,3,4-oxadiazol-2-yl) Preparation of -4-oxobutyl)carbamoyl)-L-serine
  • the preparation method is the same as the preparation method of Step 1 to Step 8 in Example 11, except that the starting material (2S, 4R)-N-(tert-butoxycarbonyl)-4-hydroxy-2-pyrrolecarboxylic acid A in Example 11 is prepared.
  • the ester was replaced with the product of the above step 2 (2S,3S)-N-(tert-butoxycarbonyl)-3-hydroxy-2-pyrrolecarboxylic acid methyl ester.
  • N-(tert-Butoxycarbonyl)-O-(tert-butyl)-L-threonine (5 g, 18.2 mmol, 1 eq) was dissolved in N,N-dimethylformamide (35 mL).
  • Methyl iodide (2.8 g, 20.0 mmol, 1.1 eq)
  • potassium carbonate (7.9 g, 54.5 mmol, 3 eq) was added and stirred at room temperature for about 3 h. After the reaction was completed, water and ethyl acetate were added to the mixture, and the mixture was evaporated. 99.0%.
  • N-(tert-Butoxycarbonyl)-O-(tert-butyl)-L-threonine methyl ester (5.2 g, 18.0 mmol) was dissolved in methanol, and hydrazine hydrate (15 mL) was added to the solution and stirred at room temperature. 24h. After the reaction was completed, the methanol was evaporated to dryness. EtOAc was evaporated. The yield was 100.0%.
  • Step 3 ((5S,10S,11R)-1-(9H- ⁇ -9-yl)-11,13,13-trimethyl-3,6,9-trioxo-5-(3-oxo Preparation of tert-butyl 3-(tritylmethylamino)propyl)-2,12-dioxa-4,7,8-triazatetradecane-10-yl)carbamate
  • Step 4 ((1S,2R)-1-(5-((S)-1-(((9H- ⁇ -9-yl)methoxy)carbonyl)amino)-4-oxo-4-()
  • Step 5 ((1S,2R)-1-(5-((S)-1-Amino-4-oxo-4-(tritylamino)butyl)-1,3,4-oxadiazole Preparation of tert-butyl 2-yl)-2-(tert-butoxy)propyl)carbamate
  • the preparation method is the same as the preparation method of the first step in the embodiment 12 except that (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid is replaced by (2S,4R)-4-hydroxytetrahydropyrrole-2. - Formic acid.
  • the preparation method is the same as the preparation method of the first step in the embodiment 11, except that the methyl (2S,4R)-N-(tert-butoxycarbonyl)-4-hydroxy-2-pyrrolecarboxylate is replaced with (2S, 4R). Methyl 4-hydroxytetrahydropyrrole-2-carboxylate.
  • the preparation method is the same as the preparation method of the step 8 in the embodiment 11, except that (2S,4R)-2-(5-((6S,10S)-10-(tert-butoxymethyl)-13,13-di Methyl-3,8,11-trioxo-1,1,1-triphenyl-12-oxy-2,7,9-azatetradecane-6-yl)-1,3,4 - Oxadiazol-2-yl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole-1-carboxylic acid tert-butyl ester was replaced by (1R,4S)-2-((( S)-1-(5-((1S,2R)-2-tert-butoxy-1-(tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazol-2-yl) -4-Oxo-4-(tritylamino)butyl)carbamoyl)-4-(
  • the preparation method is the same as the production method of Example 13, except that the 4-hydroxyproline in Step 7 is replaced with 3-hydroxyproline to prepare the target compound.
  • 1 H NMR 400 MHz, D 2 O: ⁇ 4.67-4.65 (d, 1H), 4.43-4.42 (d, 1H), 4.39-4.35 (m, 1H), 4.23-4.19 (m, 1H), 4.15 -4.13(t,1H),3.57-3.45(t,2H),3.05-3.02(m,2H),2.90-2.89(d,2H),2.46-2.43(m,1H),2.34-2.29(m, 1H), 1.35-1.33 (d, 3H).
  • ESI-MS m/z: 401.3 [M+H] + .
  • the preparation method was the same as the preparation method of Example 12 except that the (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid in the step 1 of Example 12 was replaced with 1-amino-3-(hydroxymethyl group).
  • the target compound can be obtained by replacing cyclobutane-1-carboxylic acid with O-(tert-butyl)-L-serine tert-butyl ester with O-(tert-butyl)-L-threonine tert-butyl ester.
  • the preparation method was the same as that of the production method of Example 12 except that (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid was replaced with 1-aminocyclopropane-1-carboxylic acid to obtain the target compound.
  • 1 H NMR 400 MHz, D 2 O
  • ESI-MS m/z: 357.2 [M+H] + .
  • the preparation method was the same as the preparation method of Example 12 except that (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid was replaced by 1-amino-3-(hydroxymethyl)cyclobutane-1- Formic acid can be used to prepare the target compound.
  • the preparation methods of steps 1 to 5 are the same as the preparation methods of steps 1 to 5 in Example 13, and the preparation methods of steps 6 to 8 are the same as the preparation methods of steps 6 to 8 in Example 11, except that O-(tert-butyl group) is used.
  • the L-serine tert-butyl ester is replaced with methyl 1-aminocyclopropane-1-carboxylate to obtain the target compound.
  • the preparation method is the same as the preparation method of Example 12, except that (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid is replaced by 1-amino-3-hydroxycyclobutane-1-carboxylic acid.
  • the target compound is obtained.
  • the preparation method is the same as the preparation method of Example 15, except that N 2 -(((9H-fluoren-9-yl)methoxy)carbonyl)-N 5 -trityl-L-glutamine is replaced with (S)-2-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(tert-butoxy)-5-oxopentanoic acid can be obtained to obtain the target compound.
  • the preparation method is the same as the preparation method of Example 17, except that N 2 -(((9H-fluoren-9-yl)methoxy)carbonyl)-N 5 -trityl-L-glutamine is replaced with (S)-2-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(tert-butoxy)-5-oxopentanoic acid can be obtained to obtain the target compound.
  • the preparation method of the steps 1 to 5 is the same as the preparation method of the steps 1 to 5 in the embodiment 13, except that N 2 -(((9H-fluoren-9-yl)methoxy)carbonyl)-N 5 -3 Benzyl-L-glutamine is replaced by 1-(((9H- ⁇ -9-yl)methoxy)carbonyl))amino)cyclopropane-1-carboxylic acid, and the preparation methods of steps 6-8 are the same In the preparation method of 6 to 8 in Example 11, the target compound can be obtained.
  • Methyl 4-aminotetrahydro-2H-pyran-4-carboxylate (6.50 g, 0.04 mol) was dissolved in 65 mL of tetrahydrofuran and 65 mL of water, sodium bicarbonate (12.10 g, 0.14 mol) was added, and the mixture was stirred and cooled to 0 ° C.
  • Di-tert-butyl dicarbonate (8.50 g, 0.038 mol) was added, and the reaction was allowed to warm up to room temperature for about 24 h. The reaction mixture was concentrated with EtOAc (EtOAc)EtOAc.
  • Step 4 (4-(2-(N 2 -((9H-indol-9-yl)methoxy)carbonyl)-N 5 -trityl-L-glutamyl)indole-1-carbonyl)
  • Step 4 (4-(2-(N 2 -((9H-indol-9-yl)methoxy)carbonyl)-N 5 -trityl-L-glutamyl)indole-1-carbonyl)
  • Step 7 N-(((S)-1-(5-(4-(di-tert-Butyl)-)-tetrahydro-2H-pyran-4-yl)-1,3,4-Ethyl Preparation of oxazol-2-yl)-4-carbonyl-4-(tritylamino)butyl)carbamoyl)-O-(tert-butyl)-L-threonine tert-butyl ester
  • Methyl 4-aminotetrahydro-2H-pyran-4-carboxylate (6.50 g, 0.04 mol) was dissolved in 65 mL of tetrahydrofuran and 65 mL of water, sodium bicarbonate (12.10 g, 0.14 mol) was added, and the mixture was stirred and cooled to 0 ° C. Add di-tert-butyl dicarbonate (8.50 g, 0.038 mol) and add to room temperature for about 24 h. The reaction mixture was concentrated with EtOAc (EtOAc)EtOAc.
  • Methyl 4-(tert-butoxycarbonyl)aminotetrahydro-2H-pyran-4-carboxylate (7.00 g, 27.02 mmol) was dissolved in 70 mL of methanol, then 40 mL water was added, then (1.63 g, 40. After reacting for 4 h, the reaction mixture was adjusted to pH 4-5 with 1N hydrochloric acid, and then extracted with ethyl acetate, washed with water and dried to give a white solid, 5.0 g, yield 75.7%.
  • Step 7 Tert-Butyl (S,Z)-4-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((((
  • Step 8 Tert-Butyl (S)-4-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-4-(3-(4-((tert-butoxycarbonyl)amino) Preparation of ethyl tetrahydro-2H-pyrano-pyridin-4-yl)-1,2,4-oxadiazol-5-yl)butanoate
  • Step 11 (S)-4-(3-(4-Aminotetrahydro-2H-pyran-4-yl)-1,2,4-oxadiazol-5-yl)-4-(3-(( S)-1-carboxy-2-hydroxyethyl)ureido)butyric acid
  • Step 1 Preparation of (2S,4R)-1-tert-butoxycarbonyl-2-cyano-4-(tert-butyldimethylsilyl)oxytetrahydropyrrole
  • the crude compound was purified by silica gel column chromatography (eluent: 0-5% ethyl acetate in petroleum ether). The mixture was cooled to 44.61 g of a white solid compound 1b (yield: 91.7%).
  • Tetrahydrofuran was distilled off under reduced pressure, and ethanol (200 mL) and sodium acetate (3.29 g, 40.05 mmol) were directly added thereto, and the mixture was stirred and refluxed for 2.5 hours. The reaction mixture was evaporated under reduced pressure. EtOAc was evaporated, evaporated, evaporated, evaporated, evaporated. Agent: 0-25% ethyl acetate in petroleum ether) gave 7.18 g of pale yellow solid 1d (yield: 29.2%).
  • Step 4 (2S,4R)-1-tert-Butoxycarbonyl-2-(5-((S)-1-amino-3-keto-3-(tritylamino)propyl)-1, Preparation of 2,4-oxadiazol-3-yl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole
  • Step 1 (2S,4R)-1-tert-Butoxycarbonyl-2-(5-((5S,9S)-9-((R)-1-(tert-butyl)ethyl)-12,12- Dimethyl-3,7,10-trione-1,1,1-triphenyl-11-oxa-2,6,8-triaza-5-yl)-1,2,4- Preparation of Oxadiazol-3-yl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole
  • Step 2 (2S,3R)-2-(3-((S)-3-Amino-1-(3-((2S,4R)-4-hydroxytetrahydropyrrole-2-yl)-1,2 Of 4-oxaoxazol-5-yl)-3-oxopropyl)ureido)-3-hydroxybutyric acid
  • Step 3 Preparation of O-(tert-butyl)-N-((4-nitrophenoxy)carbonyl)-L-threonine tert-butyl ester
  • Step 2 Preparation of (R)-(2-(tert-butoxy)-1-cyanoethyl)carbamic acid tert-butyl ester
  • the compound 4a (39 g, 149.81 mmol) was dissolved in pyridine (118.5 g, 1498.1 mmol), and the mixture was cooled in an ice bath, and trifluoroacetic anhydride (47.2 g, 224.72 mmol) was added dropwise at -5 ° C for 40 min, and the ice bath was removed.
  • the reaction mixture was stirred at room temperature for 3.5 hr, and the mixture was evaporated, evaporated, evaporated, evaporated Yield: 102.9%).
  • Step 3 Preparation of (R,Z)-(1-amino-3-(tert-butoxy)-1-(indolyl)propan-2-yl)carbamic acid tert-butyl ester
  • Step 4 tert-Butyl-((5S,Z)-9-amino-13,13-dimethyl-3,6-dione-1,1,1-triphenyl-7,12-dioxo
  • Step 5 ((R)-1-(5-((S)-1-(((9H- ⁇ -9-yl)methoxy)carbonyl)))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))))
  • the compound 4d (28.0 g, 32.79 mmol) was dissolved in ethanol (420 mL), and a solution of sodium acetate (4.04 g, 49.19 mmol) in water (60 mL) was added, and the mixture was stirred and refluxed for 2 hr. It was washed several times with saturated brine, distilled under reduced pressure, and purified by silica gel column chromatography (eluent: 0-30% ethyl acetate in petroleum ether). 6.6 g of milky white solid 4e was obtained (yield: 24.1%).
  • Step 6 ((R)-1-(5-((S)-1-Amino-3-keto-3-(tritylamino)propyl)-1,2,4-oxadiazole- Preparation of tert-butyl 3-yl)-2-(tert-butoxy)ethyl)carbamate
  • Example 29 The title compound was synthesized in a similar manner to the preparation of the compound 2b in Example 29, in which the H-Thr(O t Bu)-O t Bu in Example 29 was replaced with (S)-2-keto-3-aminotetrahydrofuran. .
  • Step 8 ((R)-2-(tert-Butoxy)-1-(5-((S)-3-keto-1-(3-((S)-2-ketotetrahydrofuran-3- Preparation of tert-butyl ester of ureido)-3-(tritylamino)propyl)-1,2,4-oxadiazol-3-yl)ethyl)carbamate
  • Step 9 (S)-3-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-(3-(( Preparation of S)-2-ketotetrahydrofuran-3-yl)ureido)propanamide
  • Step 5a 1-(3-((S)-1-(3-((R))-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)ethyl)-1,2, Preparation of methyl 4-oxadiazol-5-yl)-3-keto-3-(tritylamino)propyl)ureido)cyclopropane-1-carboxylate
  • Step 5b 1-(3-((S)-1-(3-((R))-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)ethyl)-1,2 Of 4-oxaoxazol-5-yl)-3-keto-3-(tritylamino)propyl)ureido)cyclopropane-1-carboxylic acid
  • the compound 5a (770 mg, 1.02 mmol) was dissolved in tetrahydrofuran (6 mL), and a solution of lithium hydroxide (85 mg, 2.04 mmol) in water (1.1 mL) was added at room temperature, and the mixture was stirred and refluxed for 3 hr.
  • Step 5c 1-(3-((S)-3-Amino-1-(3-((R)-1-amino-2-hydroxyethyl)-1,2,4-oxadiazole-5- Preparation of benzyl-3-propyloxy)ureido)cyclopropane-1-carboxylic acid
  • Example 32 The title compound was synthesized in a similar manner to the preparation of compound 5 in Example 32, in which Boc-L-Ser(tBu)-OH in Example 32 was replaced with Boc-D-Ser(tBu)-OH, using Fmoc-D -Asn(Trt)-OH replaces Fmoc-L-Asn(Trt)-OH in Example 5.
  • the crude product was purified by preparative liquid to give the title compound.
  • Step 2 Preparation of methyl 1-amino-3-(hydroxymethyl)cyclobutane-1-carboxylate
  • the compound 8a (4 g, 27.56 mmol) was dissolved in methanol (40 mL), and the mixture was cooled in an ice bath, and thionyl chloride (6.5 g, 55.13 mmol) was slowly added dropwise at 0 ° C. After the addition was completed, the mixture was heated to reflux for 4 h. The solvent was distilled off under reduced pressure to give a pale yellow oily liquid, which was taken directly to the next step.
  • Step 3 Preparation of methyl 1-((tert-butoxycarbonyl)amino)-3-(hydroxymethyl)cyclobutane-1-carboxylate
  • Step 4 Preparation of methyl 1-((tert-butoxycarbonyl)amino)-3-(tert-butyldimethylsilyloxy)cyclobutane-1-carboxylate
  • Step 5 Preparation of 1-((tert-Butoxycarbonyl)amino)-3-(tert-butyldimethylsilyloxy)cyclobutane-1-carboxylic acid
  • the compound 8d (1.6 g, 4.283 mmol) was dissolved in methanol (16 mL), and a solution of lithium hydroxide monohydrate (270 mg, 6.425 mmol) in water (3.2 mL) was added, and the mixture was stirred at room temperature for 3 h, and the solvent was evaporated under reduced pressure.
  • the ethyl ester and water were dissolved and separated, and the organic phase was extracted with water several times.
  • the aqueous phase was combined, the acid was combined with 1M citric acid solution, and then extracted with ethyl acetate.
  • the organic phase was combined, dried over anhydrous Na 2 SO 4 and evaporated. There was obtained 1.42 g of a colorless oily liquid 8e (yield: 92.2%).
  • Step 7 Preparation of tert-butyl (3-tert-butyldimethylsilylmethyl-1-cyanocyclobutyl)carbamate
  • Step 8 Preparation of (Z)-(3-tert-butyldimethylsilylmethyl-1-(N'-hydroxymethylindenyl)cyclobutyl)carbamic acid tert-butyl ester
  • Step 9 (Z)-(1-(N'-((N 2 -((9H- ⁇ -9-yl)methoxy)carbonyl)-N 4 -trityl-L-asparagine Preparation of tert-butyl ester of hydroxy)hydroxymethylindenyl)-3-(tert-butyldimethylsilylmethyl)cyclobutylcarbamate
  • Step 10 (S)-(1-(5-(1-(((9H- ⁇ -9-yl))methoxy)carbonyl)amino)-3-yl-3-yl-3-triphenylmethylamino Of propyl)-1,2,4-oxadiazol-3-yl)-3-(((tert-butyldimethylsilyl)oxy)methyl)cyclobutyl)carbamic acid tert-butyl ester preparation
  • Step 11 (S)-(1-(5-(1-Amino-3-keto-3-(tritylamino)propyl)-1,2,4-oxadiazol-3-yl) Preparation of tert-butyl 3-(((tert-butyldimethylsilyl)oxy)methyl)cyclobutyl)carbamate
  • Step 12 N-(((S)-1-(3-(1-((tert-Butyloxycarbonyl))amino)-3-(((tert-butyldimethylsilyl)oxy)methyl)) Butyl)-1,2,4-oxadiazol-5-yl)-3-keto-3-(tritylamino)propyl)carbamoyl)-O-(tert-butyl)-L -Preparation of threonine tert-butyl ester
  • Step 13 (2S,3R)-2-(3-((S)-3-Amino-1-(3-(1-amino-3-(hydroxymethyl)cyclobutyl)-1,2,4 -Oxadiazole-5-yl)-3-oxopropyl)ureido)-3-hydroxybutyric acid
  • Example 35 The title compound was synthesized in a similar manner to the preparation of the compound in Example 35. Wherein Fmoc-Asn(Trt)-OH in Example 8 was replaced with Fmoc-Asp(OtBu)-OH, and H-Thr (OtBu) in Example 35 was replaced with H-Ser(O t Bu)-O t Bu -OtBu.
  • Compound A The compound represented by the following formula (Compound A) was prepared according to the method disclosed in Example 2 of WO2015/033301 (PCT/IB2014/064281), and identified by hydrogen spectrum and mass spectrometry,
  • the pharmacokinetic characteristics of Compound A and the antitumor effect on the subcutaneous xenograft model of colon cancer CT26 cells were tested using the following Experimental Examples 1 and 2, and the results showed that the bioavailability (F) and inhibition of Compound A were observed.
  • the tumor rate is weaker than some of the compounds of the invention.
  • the experiment was carried out using the compound of the present invention prepared in the above examples and the compound of Example 2 ("Compound A") in WO2015/033301.
  • the oral drug was dissolved in physiological saline to prepare a clear solution of 0.5 mg/mL, and the intravenous drug was dissolved in physiological saline to prepare a 0.1 mg/mL clear solution.
  • mice Male BALB/c mice, 3 in each group, weighing 18-22 g, were provided by Shanghai Xipuer-Beikai Experimental Animal Co., Ltd.
  • test mice were given an environmental adaptation period of 2 to 4 days before the experiment, and were fasted for 8-12 hours before administration, and given water for 2 hours after administration, and fed after 4 hours.
  • Acetonitrile (chromatographically pure): produced by Spectrum;
  • mice were fasted but allowed to drink water for 12 hours, and the blank plasma was taken at 0 o'clock;
  • step 2) taking the mouse in step 1), administering intragastric administration (IG) to the test compound 10 mg/kg; intravenous (IV) administering the test compound 1 mg/kg;
  • Example 1 The pharmacokinetic experimental data are shown in Table 1. The results indicate that after oral administration of the compound of the above examples, there is a certain amount of exposure and a suitable half-life in the plasma of the animal, especially Example 4, Example 5
  • Example 8 The compounds of Example 8, Example 6 and Example 22 have very good half-life, area under the curve and bioavailability, have good drug-forming properties, and have good clinical application prospects.
  • mice Female BALB/c mice, 3 in each group, weighing 18-22 g, were provided by Shanghai Xipuer-Beikai Experimental Animal Co., Ltd. The test mice were given an environmental adaptation period of 2 to 4 days before the experiment, and were fasted for 8-12 hours before administration, and given water for 2 hours after administration, and fed after 4 hours.
  • Compound A was prepared according to the method disclosed in Example 2 of WO 2015/033301.
  • the animals were randomly divided into groups of 6 each, and each test group was orally administered with 20 mg/kg, one day. Once, continuous administration for 14 days. The changes in body weight of the experimental animals and whether the tumor growth was inhibited or delayed were examined. Tumor diameters were measured with vernier calipers three times a week.
  • T/C T RTV / C RTV X 100% (T RTV : treatment group RTV; C RTV : solvent control group RTV).
  • TGI [1 - (the average tumor volume at the end of a treatment group - the average tumor volume at the start of administration of the treatment group) / (the average of the solvent control group at the end of treatment) Tumor volume - mean tumor volume at the start of treatment in the solvent control group)] X 100%.
  • the compound of the present invention has no effect on the body weight of the mouse colon cancer CT26 cell subcutaneous homologous tumor in the BALB/C mouse model.
  • Table 2 shows the administration of Example 4, Example 5, Example 35 and Compound A.
  • the effect of body weight indicated that the body weight of the control group and each drug-administered group gradually increased during the administration period and was well tolerated.
  • the evaluation index of drug efficacy is shown in Table 3.
  • the average tumor volume of the tumor-bearing mice in the solvent control group reached 3672 mm 3 , and the average tumor volume of the tumor-bearing mice in the other drug-administered groups was average. It is smaller than the average tumor volume of the control group, wherein the compound of Example 4 has a T/C value of 41.5% on the 15th day and a TGI value of 52.4%, indicating that it has a significant inhibitory effect on CT26 colon cancer xenografts, and the effect is obvious.
  • the average tumor volume of the tumor-bearing mice in the solvent control group reached 1524 mm 3 , and the average tumor volume of the tumor-bearing mice in each of the other test groups was smaller than the average tumor volume of the control group.
  • the compound of Example 5 had a T/C value of 39.0% at day 14 and a TGI value of 63.7%, indicating that it had a significant inhibitory effect on CT26 colon cancer xenografts.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Hematology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to the field of medical chemistry, and relates to a heterocyclic compound serving as a PD-L1 inhibitor. Specifically, the present invention relates to a compound represented by formula A, or an isomer, pharmaceutically acceptable salt, solvate or prodrug thereof, preparation methods therefor, a pharmaceutical composition containing these compounds, and a use of these compounds or composition in treating cancers or infectious diseases.

Description

作为PD-L1抑制剂的杂环类化合物Heterocyclic compounds as PD-L1 inhibitors 技术领域Technical field
本发明属于医药化学领域,涉及一类作为PD-L1抑制剂的杂环类化合物及其应用,具体地,本发明涉及式A所示的化合物或其异构体、药学上可接受的盐、溶剂化物或前药,它们的制备方法以及含有这些化合物的药物组合物和这些化合物或组合物用于治疗癌症或者感染类疾病的用途。The present invention belongs to the field of medical chemistry, and relates to a class of heterocyclic compounds as PD-L1 inhibitors and uses thereof. In particular, the present invention relates to a compound represented by formula A or an isomer thereof, a pharmaceutically acceptable salt, Solvates or prodrugs, methods for their preparation, and pharmaceutical compositions containing these compounds and the use of such compounds or compositions for the treatment of cancer or infectious diseases.
背景技术Background technique
程序性死亡受体-1(Programmed Cell Death-1,PD-1)及其配体PD-Ll(B7·H1)属于CD28/B7超家族。PD-1主要表达在T细胞、B细胞、自然杀伤细胞(Natual Killer Cell,NK细胞)的膜表面,PD-L1主要表达于成熟的CD4T细胞、CD8T细胞、B细胞、单核细胞、树突状细胞(Dendritic Cells,DCs)、巨噬细胞等造血细胞及一些非造血细胞,如内皮细胞、胰岛细胞、肥大细胞等的膜表面。其中PD-L1在多种肿瘤中高表达,如肺癌、胃癌、多发性骨髓、黑色素瘤和乳腺癌等。肿瘤细胞表面上的PD-L1的表达与T细胞表面的配体相互作用,可诱导T细胞的凋亡或降低T细胞的反应活性,从而抑制肿瘤免疫应答,使肿瘤细胞逃避免疫攻击。因此阻断PD1-PDL1信号通路的拮抗剂,可促进T细胞的激活、逆转肿瘤免疫微环境、增强内源性抗肿瘤免疫效应。靶向PD-1/PD-L1抑制剂在肿瘤免疫治疗领域有着广阔的应用前景。Programmed Cell Death-1 (PD-1) and its ligand PD-L1 (B7·H1) belong to the CD28/B7 superfamily. PD-1 is mainly expressed on the membrane surface of T cells, B cells, and natural killer cells (NK cells). PD-L1 is mainly expressed on mature CD4T cells, CD8 T cells, B cells, monocytes, and dendrites. Hematopoietic cells such as dendritic cells (DCs), macrophages, and some non-hematopoietic cells, such as membrane surfaces of endothelial cells, islet cells, mast cells, and the like. Among them, PD-L1 is highly expressed in various tumors, such as lung cancer, gastric cancer, multiple bone marrow, melanoma and breast cancer. The expression of PD-L1 on the surface of tumor cells interacts with the ligand on the surface of T cells, which can induce apoptosis of T cells or reduce the reactivity of T cells, thereby inhibiting the tumor immune response and allowing tumor cells to escape immune attack. Therefore, blocking the antagonist of PD1-PDL1 signaling pathway can promote the activation of T cells, reverse the tumor immune microenvironment, and enhance the endogenous anti-tumor immune effect. Targeting PD-1/PD-L1 inhibitors has broad application prospects in the field of tumor immunotherapy.
目前抗PD-1/PD-L1抗体治疗在临床上已显示具有优势作用,然而生物大分子也具有一些缺点,例如免疫原性以及给药途径的限制。因此,仍需要开发具有更好药效的靶向PD-1/PD-L1抑制剂。本发明的发明人发现一类小分子药物能特异性地调控和/或调解PD-L1及其相关蛋白激酶的转导,从而用于治疗与PD-1/PD-L1相关的疾病。Currently, anti-PD-1/PD-L1 antibody therapy has been shown to have a clinical advantage, however, biomacromolecules also have some disadvantages such as immunogenicity and limitations of the route of administration. Therefore, there is still a need to develop targeted PD-1/PD-L1 inhibitors with better pharmacodynamics. The inventors of the present invention have found that a class of small molecule drugs can specifically modulate and/or mediate the transduction of PD-L1 and its related protein kinases for the treatment of diseases associated with PD-1/PD-L1.
发明内容Summary of the invention
本发明的一个目的在于提供一种通式A所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,It is an object of the present invention to provide a compound of the formula A or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof,
Figure PCTCN2018084352-appb-000001
Figure PCTCN2018084352-appb-000001
其中:among them:
X和Y各自独立地选自N、O和S,且X为N时,Y选自O和S;X为O或S时,Y为N;Y为N时,X选自O和S;Y为O或S时,X为N;X and Y are each independently selected from N, O and S, and when X is N, Y is selected from O and S; when X is O or S, Y is N; when Y is N, X is selected from O and S; When Y is O or S, X is N;
当X为N,Y选自O和S时或Y为O或S,X为N时,基团
Figure PCTCN2018084352-appb-000002
Figure PCTCN2018084352-appb-000003
当X为O或S,Y为N时或Y为N,X选自O和S时,基团
Figure PCTCN2018084352-appb-000004
Figure PCTCN2018084352-appb-000005
When X is N, Y is selected from O and S or Y is O or S, and X is N, the group
Figure PCTCN2018084352-appb-000002
for
Figure PCTCN2018084352-appb-000003
When X is O or S, Y is N or Y is N, and X is selected from O and S, the group
Figure PCTCN2018084352-appb-000004
for
Figure PCTCN2018084352-appb-000005
R 1选自氨基酸Ser和Thr的侧链; R 1 is selected from the side chains of the amino acids Ser and Thr;
R 2选自氨基酸Ser和Thr残基; R 2 is selected from the group consisting of the amino acid Ser and Thr residues;
R 3选自H、-(CH 2) mC(O)OR 4、-(CH 2) mC(O)N(R 5)(R 6)和-(CH 2) mCN,其中R 4、R 5、R 6各自独立地选自H和烷基,m为0、1、2、3或4;和 R 3 is selected from the group consisting of H, —(CH 2 ) m C(O)OR 4 , —(CH 2 ) m C(O)N(R 5 )(R 6 ) and —(CH 2 ) m CN, wherein R 4 , R 5 , R 6 are each independently selected from H and alkyl, and m is 0, 1, 2, 3 or 4;
n为1、2、3或4。n is 1, 2, 3 or 4.
在一些优选的实施方案中,一种通式A所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其为通式I或II所示的化合物或其立体异构体、药学上可接受的盐、溶剂化物、结晶或前药:In some preferred embodiments, a compound of Formula A, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, which is represented by Formula I or II a compound or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug:
Figure PCTCN2018084352-appb-000006
Figure PCTCN2018084352-appb-000006
其中:Q选自O和S;R 1、R 2、R 3的定义如通式A中所述。 Wherein: Q is selected from O and S; and R 1 , R 2 and R 3 are as defined in Formula A.
在一些实施方案中,根据本发明的通式A、通式I或II所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中:In some embodiments, a compound of Formula A, Formula I or II, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, according to the invention, wherein:
R 1选自氨基酸Ser和Thr的侧链; R 1 is selected from the side chains of the amino acids Ser and Thr;
R 2选自氨基酸Ser和Thr残基; R 2 is selected from the group consisting of the amino acid Ser and Thr residues;
R 3选自H、-(CH 2) mC(O)OR 4、-(CH 2) mC(O)N(R 5)(R 6)和-(CH 2) mCN,其中R 4、R 5、R 6各自独立地选自H和C 1-6烷基,m为1、2、3或4;和 R 3 is selected from the group consisting of H, —(CH 2 ) m C(O)OR 4 , —(CH 2 ) m C(O)N(R 5 )(R 6 ) and —(CH 2 ) m CN, wherein R 4 , R 5 and R 6 are each independently selected from H and C 1-6 alkyl, and m is 1, 2, 3 or 4;
n为1、2、3或4。n is 1, 2, 3 or 4.
在一些实施方案中,根据本发明的通式A、通式I或II所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中:In some embodiments, a compound of Formula A, Formula I or II, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, according to the invention, wherein:
R 1选自
Figure PCTCN2018084352-appb-000007
R 1 is selected from
Figure PCTCN2018084352-appb-000007
R 2选自
Figure PCTCN2018084352-appb-000008
R 2 is selected from
Figure PCTCN2018084352-appb-000008
R 3选自H、-(CH 2) mC(O)OR 4、-(CH 2) mC(O)N(R 5)(R 6)和-(CH 2) mCN,其中R 4、R 5、R 6各自独立地选自H和C 1-6烷基,m为1、2、3或4;和 R 3 is selected from the group consisting of H, —(CH 2 ) m C(O)OR 4 , —(CH 2 ) m C(O)N(R 5 )(R 6 ) and —(CH 2 ) m CN, wherein R 4 , R 5 and R 6 are each independently selected from H and C 1-6 alkyl, and m is 1, 2, 3 or 4;
n为1、2、3或4。n is 1, 2, 3 or 4.
在一些实施方案中,根据本发明的通式A、通式I或II所示的化合物或其立体异构体、药 学上可接受的盐、溶剂合物、结晶或前药,其中:In some embodiments, a compound of Formula A, Formula I or II, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, according to the invention, wherein:
R 1选自
Figure PCTCN2018084352-appb-000009
R 1 is selected from
Figure PCTCN2018084352-appb-000009
R 2选自
Figure PCTCN2018084352-appb-000010
R 2 is selected from
Figure PCTCN2018084352-appb-000010
R 3选自H、
Figure PCTCN2018084352-appb-000011
和n为1、2、3或4。
R 3 is selected from H,
Figure PCTCN2018084352-appb-000011
And n is 1, 2, 3 or 4.
在一些实施方案中,根据本发明的以上通式A、通式I或II所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中
Figure PCTCN2018084352-appb-000012
选自
Figure PCTCN2018084352-appb-000013
Figure PCTCN2018084352-appb-000014
In some embodiments, the compound of Formula A, Formula I or II above, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof thereof, according to the invention, wherein
Figure PCTCN2018084352-appb-000012
Selected from
Figure PCTCN2018084352-appb-000013
Figure PCTCN2018084352-appb-000014
在一些实施方案中,根据本发明的通式A、通式I或II所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中:In some embodiments, a compound of Formula A, Formula I or II, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, according to the invention, wherein:
R 1选自
Figure PCTCN2018084352-appb-000015
R 1 is selected from
Figure PCTCN2018084352-appb-000015
R 2选自
Figure PCTCN2018084352-appb-000016
R 2 is selected from
Figure PCTCN2018084352-appb-000016
with
Figure PCTCN2018084352-appb-000017
选自
Figure PCTCN2018084352-appb-000018
一种具有通式A所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其非限制性实例包括:
Figure PCTCN2018084352-appb-000017
Selected from
Figure PCTCN2018084352-appb-000018
A compound having the formula A or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, non-limiting examples of which include:
Figure PCTCN2018084352-appb-000019
Figure PCTCN2018084352-appb-000019
Figure PCTCN2018084352-appb-000020
Figure PCTCN2018084352-appb-000020
本发明的另一个目的在于提供一种通式B所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,Another object of the present invention is to provide a compound of the formula B or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof,
Figure PCTCN2018084352-appb-000021
Figure PCTCN2018084352-appb-000021
其中:among them:
X和Y各自独立地选自N、O和S,且X为N时,Y选自O和S;X为O或S时,Y为N;Y为N时,X选自O和S;Y为O或S时,X为N;X and Y are each independently selected from N, O and S, and when X is N, Y is selected from O and S; when X is O or S, Y is N; when Y is N, X is selected from O and S; When Y is O or S, X is N;
当X为N,Y选自O和S时或Y为O或S,X为N时,基团
Figure PCTCN2018084352-appb-000022
Figure PCTCN2018084352-appb-000023
当X为O或S,Y为N时或Y为N,X选自O和S时,基团
Figure PCTCN2018084352-appb-000024
Figure PCTCN2018084352-appb-000025
When X is N, Y is selected from O and S or Y is O or S, and X is N, the group
Figure PCTCN2018084352-appb-000022
for
Figure PCTCN2018084352-appb-000023
When X is O or S, Y is N or Y is N, and X is selected from O and S, the group
Figure PCTCN2018084352-appb-000024
for
Figure PCTCN2018084352-appb-000025
R 7选自氨基酸Asn、Gln、Asp、Glu的侧链; R 7 is selected from the side chain of the amino acids Asn, Gln, Asp, Glu;
R 8选自氨基酸Ser和Thr残基; R 8 is selected from the group consisting of the amino acid Ser and Thr residues;
R 9、R 10各自独立地选自H、烷基和烷基羟基,R 9、R 10不同时为H,且 R 9 and R 10 are each independently selected from H, an alkyl group and an alkylhydroxy group, and R 9 and R 10 are not simultaneously H, and
R 9或R 10与其α碳位上连接的氨基一起形成氮杂环烷基或氮氧杂环烷基,所述氮杂环烷基或氮氧杂环烷基任选地被一个或多个选自羟基、羟基烷基、氰基、氨基、卤素、硝基的取代基所取代;或 R 9 or R 10 together with the amino group attached to its alpha carbon position form a nitrogen heterocycloalkyl or a nitrogen heterocycloalkyl group, optionally a one or more Substituted with a substituent selected from the group consisting of a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a halogen, and a nitro group; or
R 9、R 10与它们所连接的C原子一起形成环烷基或氧杂环烷基,所述环烷基或氧杂环烷基任选地被一个或多个选自羟基、羟基烷基、氰基、氨基、卤素、硝基的取代基所取代。 R 9 and R 10 together with the C atom to which they are attached form a cycloalkyl or oxacycloalkyl group, which is optionally selected from one or more selected from the group consisting of a hydroxyl group and a hydroxyalkyl group. Substituted by a substituent of a cyano group, an amino group, a halogen or a nitro group.
在一个优选的实施方案中,一种通式B所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,为通式III或IV所示的化合物或其立体异构体、药学上可接受的盐、溶剂化物、结晶或前药:In a preferred embodiment, a compound of the formula B, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, is a compound of the formula III or IV. Or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof:
Figure PCTCN2018084352-appb-000026
Figure PCTCN2018084352-appb-000026
其中:Q选自O和S,R 7、R 8、R 9、R 10的定义如通式B中所述。 Wherein: Q is selected from O and S, and R 7 , R 8 , R 9 and R 10 are as defined in Formula B.
在一些实施方案中,根据本发明的提供的通式B所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中:In some embodiments, the compound of Formula B, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal, or prodrug thereof, according to the present invention, wherein:
R 7选自氨基酸Asn、Gln、Asp、Glu的侧链; R 7 is selected from the side chain of the amino acids Asn, Gln, Asp, Glu;
R 8选自氨基酸Ser和Thr残基; R 8 is selected from the group consisting of the amino acid Ser and Thr residues;
R 9、R 10各自独立地选自H、C 1-6烷基和C 1-6烷基羟基,R 9、R 10不同时为H,且 R 9 and R 10 are each independently selected from the group consisting of H, C 1-6 alkyl and C 1-6 alkyl hydroxy, and R 9 and R 10 are not H at the same time, and
R 9或R 10与其α碳位上连接的氨基一起形成氮杂C 3-6环烷基或氮氧杂C 3-6环烷基,所述氮杂C 3-6环烷基或氮氧杂C 3-6环烷基任选地被一个或多个选自羟基、羟基C 1-6烷基、氰基、氨基、卤素、硝基的取代基所取代;或 R 9 or R 10 together with the amino group attached to its alpha carbon position form an aza C 3-6 cycloalkyl or a nitrogenoxa C 3-6 cycloalkyl group, said aza C 3-6 cycloalkyl or nitrox a hetero C 3-6 cycloalkyl group optionally substituted with one or more substituents selected from the group consisting of hydroxyl, hydroxy C 1-6 alkyl, cyano, amino, halogen, nitro;
R 9、R 10与它们所连接的C原子一起形成C 3-6环烷基或氧杂C 3-6环烷基,所述C 3-6环烷基或氧杂C 3-6环烷基任选地被一个或多个选自羟基、羟基C 1-6烷基、氰基、氨基、卤素、硝基的取代基所取代。 R 9 and R 10 together with the C atom to which they are attached form a C 3-6 cycloalkyl or oxa C 3-6 cycloalkyl group, said C 3-6 cycloalkyl or oxa C 3-6 cycloalkane The group is optionally substituted with one or more substituents selected from the group consisting of a hydroxyl group, a hydroxy C 1-6 alkyl group, a cyano group, an amino group, a halogen, and a nitro group.
在另一个优选的实施方案中,一种通式B所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中R 9为H,R 10与其α碳位上连接的氨基一起形成氮杂C 3-6环烷基,所述氮杂C 3-6环烷基任选地被一个或多个选自羟基、羟基C 1-6烷基、氰基、氨基、卤素、硝基的取代基所取代。 In another preferred embodiment, a compound of the formula B, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein R 9 is H, R 10 is amino group formed on the α-carbon position with C 3-6 cycloalkyl aza, aza said C 3-6 cycloalkyl optionally substituted with one or more substituents selected from hydroxy, hydroxy C 1-6 alkyl, Substituted by a substituent of a cyano group, an amino group, a halogen or a nitro group.
在另一个优选的实施方案中,一种通式B所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中R 9选自C 1-3烷基,R 10选自C 1-3烷基,R 9、R 10与它们所连接的C原子一起形成C 3-6环烷基,所述C 3-6环烷基任选地被一个或多个选自羟基、羟基烷基、氰基、氨基、卤素、硝基的取代基所取代。 In another preferred embodiment, a compound of the formula B, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein R 9 is selected from C 1-3 An alkyl group, R 10 is selected from C 1-3 alkyl groups, and R 9 and R 10 together with the C atom to which they are attached form a C 3-6 cycloalkyl group, optionally a C 3-6 cycloalkyl group Or a plurality of substituents selected from the group consisting of a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a halogen, and a nitro group.
在另一个优选的实施方案中,一种通式B所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中R 9选自C 1-3烷基,R 10选自C 1-3羟基烷基,R 9、R 10与它们所连接的C原子一起形成氧杂C 3-6环烷基,所述氧杂C 3-6环烷基任选地被一个或多个选自羟基、羟基烷基、氰基、氨基、卤素、硝基的取代基所取代。 In another preferred embodiment, a compound of the formula B, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein R 9 is selected from C 1-3 An alkyl group, R 10 is selected from a C 1-3 hydroxyalkyl group, and R 9 and R 10 together with the C atom to which they are attached form an oxa C 3-6 cycloalkyl group, said oxa C 3-6 cycloalkyl group It is optionally substituted with one or more substituents selected from the group consisting of a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a halogen, and a nitro group.
一种具有通式B所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其非限制性实例包括:A compound having the formula B or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, non-limiting examples of which include:
Figure PCTCN2018084352-appb-000027
Figure PCTCN2018084352-appb-000027
Figure PCTCN2018084352-appb-000028
Figure PCTCN2018084352-appb-000028
本发明的另一个目的是提供一种具有通式C所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,Another object of the present invention is to provide a compound of the formula C or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug,
Figure PCTCN2018084352-appb-000029
Figure PCTCN2018084352-appb-000029
其中:among them:
X和Y各自独立地选自N、O和S,且X为N时,Y选自O和S;X为O或S时,Y为N;Y为N时,X选自O和S;Y为O或S时,X为N;X and Y are each independently selected from N, O and S, and when X is N, Y is selected from O and S; when X is O or S, Y is N; when Y is N, X is selected from O and S; When Y is O or S, X is N;
当X为N,Y选自O和S时或Y为O或S,X为N时,基团
Figure PCTCN2018084352-appb-000030
Figure PCTCN2018084352-appb-000031
当X为O或S,Y为N时或Y为N,X选自O和S时,基团
Figure PCTCN2018084352-appb-000032
Figure PCTCN2018084352-appb-000033
When X is N, Y is selected from O and S or Y is O or S, and X is N, the group
Figure PCTCN2018084352-appb-000030
for
Figure PCTCN2018084352-appb-000031
When X is O or S, Y is N or Y is N, and X is selected from O and S, the group
Figure PCTCN2018084352-appb-000032
for
Figure PCTCN2018084352-appb-000033
R 11选自氨基酸Ser和Thr的侧链; R 11 is selected from the side chains of the amino acids Ser and Thr;
R 12选自氨基酸Asn、Gln、Asp、Glu侧链; R 12 is selected from the group consisting of amino acid Asn, Gln, Asp, Glu side chain;
R 15选自H和烷基; R 15 is selected from the group consisting of H and alkyl;
R 13、R 14各自独立地选自H、烷基,R 13、R 14不同时为H,且 R 13 and R 14 are each independently selected from H and alkyl, and R 13 and R 14 are not H at the same time, and
R 13、R 14与它们所连接的C原子一起形成环烷基;或 R 13 , R 14 together with the C atom to which they are attached form a cycloalkyl group; or
R 13或R 14与其α碳位连接的亚氨基一起形成氮杂环烷基,或R 14与R 15及它们所连接的原子一起形成任选取代的内酯环烷基。 R 13 or R 14 together with the imino group to which the α carbon position is bonded forms a nitrogen heterocycloalkyl group, or R 14 together with R 15 and the atom to which they are attached form an optionally substituted lactone cycloalkyl group.
所述环烷基、氮杂环烷基或内酯环烷基任选地被一个或多个选自羟基、羟基烷基、氰基、氨基、卤素、硝基的取代基所取代;The cycloalkyl, azacycloalkyl or lactone cycloalkyl is optionally substituted with one or more substituents selected from the group consisting of hydroxyl, hydroxyalkyl, cyano, amino, halogen, nitro;
在一个优选的实施方案中,一种通式C所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,为通式V或VI所示的化合物或其立体异构体、药学上可接受的盐、溶剂化物、结晶或前药:In a preferred embodiment, a compound of the formula C, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, is a compound of the formula V or VI. Or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof:
Figure PCTCN2018084352-appb-000034
Figure PCTCN2018084352-appb-000034
其中:Q选自O和S;R 11、R 12、R 13、R 14、R 15的定义如通式C中所述。 Wherein: Q is selected from O and S; and R 11 , R 12 , R 13 , R 14 and R 15 are as defined in Formula C.
在一个优选的实施方案中,一种具有通式C所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中R 13、R 14与它们所连接的C原子一起形成C 3-6环烷基; In a preferred embodiment, a compound of the formula C, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein R 13 , R 14 and The attached C atoms together form a C 3-6 cycloalkyl group;
在另一个优选的实施方案中,一种具有通式C所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中R 14与其α碳位上连接的亚氨基一起形成氮杂C 3-6环烷基; In another preferred embodiment, a compound of the formula C, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein R 14 and its alpha carbon position The attached imino groups together form an aza C 3-6 cycloalkyl group;
在另一个优选的实施方案中,一种具有通式C所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中R 14与R 15及它们所连接的原子一起形成内酯C 3-6环烷基,所述内酯环烷基被一个或多个选自羟基、氧代、羟基烷基、氰基、氨基、卤素、硝基的取代基所取代; In another preferred embodiment, a compound of the formula C, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein R 14 and R 15 and The attached atoms together form a lactone C 3-6 cycloalkyl group which is substituted by one or more selected from the group consisting of hydroxyl, oxo, hydroxyalkyl, cyano, amino, halogen, nitro Substituted by
一种具有通式C所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其非限制性实例包括:A compound having the formula C or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, non-limiting examples of which include:
Figure PCTCN2018084352-appb-000035
Figure PCTCN2018084352-appb-000035
Figure PCTCN2018084352-appb-000036
Figure PCTCN2018084352-appb-000036
本发明的另一个目的是提供如通式I所示化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药的制备方法,所述方法包括以下步骤:Another object of the present invention is to provide a process for the preparation of a compound of the formula I, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, comprising the steps of:
Figure PCTCN2018084352-appb-000037
Figure PCTCN2018084352-appb-000037
(1)式ia化合物与式ib化合物经过缩合反应生成式ic化合物;(1) a compound of the formula ia and a compound of the formula ib are subjected to a condensation reaction to form a compound of the formula ic;
(2)式ic化合物环化得到式id化合物;(2) cyclizing a compound of the formula ic to give a compound of the formula id;
(3)式id化合物与胺类化合物反应,得到式ie化合物;(3) a compound of the formula id is reacted with an amine compound to give a compound of the formula IE;
(4)式ie化合物与式if化合物发生亲核取代反应,得到式ig化合物;(4) a nucleophilic substitution reaction of a compound of the formula IE with a compound of the formula i to give a compound of the formula ig;
(5)在酸性条件下,式ig化合物发生水解反应得到式I化合物;(5) under acidic conditions, a compound of formula ig is hydrolyzed to give a compound of formula I;
其中:Pg 1代表R 1的保护基;Pg 2代表R 2的保护基;如通式I所述,R 1和R 2各自选自氨基酸Ser和Thr侧链,为了避免氨基酸Ser或Thr侧链在反应过程中进一步的反应,可采取常规的羟基保护基保护氨基酸Ser或Thr侧链,Pg 1、Pg 2优选为叔丁基( tBu); Wherein: Pg 1 represents a protecting group of R 1 ; Pg 2 represents a protecting group of R 2 ; as described in Formula I, R 1 and R 2 are each selected from the group consisting of the amino acid Ser and Thr side chains, in order to avoid the amino acid Ser or Thr side chain In the further reaction during the reaction, a conventional hydroxy protecting group may be used to protect the amino acid Ser or Thr side chain, and Pg 1 and Pg 2 are preferably tert-butyl ( t Bu);
Pg 3代表R 3的保护基或不存在;如通式I所述,R 3选自H、-(CH 2) mC(O)OR 4、-(CH 2) mC(O)N(R 5)(R 6)和-(CH 2) mCN;当R 3为含有羟基或氨基等在进一步反应中易发生变化的基团时,可采用常规的保护基对其进行保护;Pg 4、Pg 5代表氨基保护基。 Pg 3 represents a protecting group of R 3 or is absent; as described in Formula I, R 3 is selected from H, -(CH 2 ) m C(O)OR 4 , -(CH 2 ) m C(O)N ( R 5 )(R 6 ) and —(CH 2 ) m CN; when R 3 is a group containing a hydroxyl group or an amino group which is susceptible to change in further reaction, it can be protected by a conventional protecting group; Pg 4 Pg 5 represents an amino protecting group.
本发明的另一个目的是提供如通式II所示化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药的制备方法,所述方法包括以下步骤:Another object of the present invention is to provide a process for the preparation of a compound of the formula II, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, comprising the steps of:
Figure PCTCN2018084352-appb-000038
Figure PCTCN2018084352-appb-000038
(1)式ia化合物与式iib化合物经过缩合反应生成式iic化合物;(1) a compound of the formula ia and a compound of the formula iib are subjected to a condensation reaction to form a compound of the formula iic;
(2)式iic化合物环化得到式iid化合物;(2) cyclization of a compound of formula iic to give a compound of formula iid;
(3)式iid化合物与胺类化合物反应,得到式iie化合物;(3) a compound of the formula iid is reacted with an amine compound to give a compound of the formula iie;
(4)式iie化合物与式if化合物经过亲核取代反应,得到式iig化合物;(4) a compound of the formula iie and a compound of the formula if subjected to nucleophilic substitution to give a compound of the formula iig;
(5)在酸性条件下,式iig化合物发生水解反应得到式II化合物;(5) under acidic conditions, a compound of formula iig is hydrolyzed to give a compound of formula II;
其中:Pg 1代表R 1的保护基;Pg 2代表R 2的保护基;如通式II所述,R 1和R 2各自选自氨基酸Ser和Thr侧链,为了避免氨基酸Ser或Thr侧链在反应过程中进一步的反应,可采取常规的羟基保护基保护羟基,Pg 1、Pg 2优选为叔丁基( tBu); Wherein: Pg 1 represents a protecting group for R 1 ; Pg 2 represents a protecting group for R 2 ; as described in Formula II, R 1 and R 2 are each selected from the group consisting of the amino acid Ser and Thr side chains, in order to avoid the amino acid Ser or Thr side chain Further reaction in the reaction, the conventional hydroxyl group can be used to protect the hydroxyl group, Pg 1 , Pg 2 is preferably tert-butyl ( t Bu);
Pg 3代表R 3的保护或不存在;如通式II所述,R 3选自H、-(CH 2) mC(O)OR 4、-(CH 2) mC(O)N(R 5)(R 6)和-(CH 2) mCN;当R 3为含有羟基或氨基等在进一步反应中易发生变化的基团,可采用常规的保护基对其进行保护; Pg 3 represents the protection or absence of R 3 ; as described in Formula II, R 3 is selected from H, -(CH 2 ) m C(O)OR 4 , -(CH 2 ) m C(O)N(R 5 ) (R 6 ) and -(CH 2 ) m CN; when R 3 is a group which contains a hydroxyl group or an amino group and is susceptible to change in further reaction, it can be protected by a conventional protecting group;
Pg 4、Pg 5代表氨基保护基。 Pg 4 and Pg 5 represent an amino protecting group.
本发明的另一个目的是提供如通式B、通式C、通式III、通式IV、通式V或通式VI或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药的制备方法,根据上述提供的通式I和通式II化合物的制备方法,本领域技术人员根据有机合成领域的常规知识采用常规的原料即可得到通式B、通式C、通式III、通式IV、通式V或通式VI。Another object of the present invention is to provide a compound of Formula B, Formula C, Formula III, Formula IV, Formula V or Formula VI or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate thereof, The preparation method of the crystallization or prodrug, according to the preparation method of the compound of the general formula I and the general formula II provided above, the person skilled in the art can obtain the general formula B, the general formula C, and the conventional raw material according to the conventional knowledge in the field of organic synthesis. Formula III, Formula IV, Formula V or Formula VI.
本发明的另一个目的是提供一种药物组合物,所述的药物组合物含有如通式A、通式B、通式C、通式I、通式II、通式III、通式IV、通式V或通式VI所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药以及药学上可接受的载体。Another object of the present invention is to provide a pharmaceutical composition comprising, for example, Formula A, Formula B, Formula C, Formula I, Formula II, Formula III, Formula IV, A compound of the formula V or formula VI, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, and a pharmaceutically acceptable carrier.
本发明的另一个目的是提供一种如通式A、通式B、通式C、通式I、通式II、通式III、通式 IV、通式V、通式VI所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药或包含其的药物组合物在制备用于治疗癌症或感染性疾病的药物中的应用。Another object of the present invention is to provide a compound of the formula A, formula B, formula C, formula I, formula II, formula III, formula IV, formula V, formula VI. Or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof or a pharmaceutical composition comprising the same for use in the manufacture of a medicament for the treatment of cancer or an infectious disease.
在一个优选的实施方案中,一种如通式A、通式B、通式C、通式I、通式II、通式III、通式IV、通式V或通式VI所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药或包含其的药物组合物在制备用于治疗癌症或感染性疾病的药物中的应用,其中所述癌症包括但不限于黑色素瘤、脑瘤(具有恶性的星形神经胶质和少突神经胶质细胞瘤成分的神经胶质瘤等)、食管癌、胃癌、肝癌、胰腺癌、结肠直肠癌(结肠癌、直肠癌等)、肺癌(非小细胞肺癌、小细胞肺癌、原发或转移性鳞状癌等)、肾癌、乳腺癌、卵巢癌、前列腺癌、皮肤癌、神经母细胞瘤、肉瘤、骨软骨瘤、骨瘤、骨肉瘤、精原细胞瘤、睾丸肿瘤、子宫癌(子宫颈癌、子宫内膜癌等)、头颈肿瘤(上颌骨癌、喉癌、咽癌、舌癌、口内癌等)、多发性骨髓瘤、恶性淋巴瘤(网状细胞肉瘤、淋巴肉瘤、霍奇金淋巴瘤等)、真性红细胞增多症、白血病(急性粒细胞白血病、慢性粒细胞白血病、急性淋巴细胞白血病、慢性淋巴细胞白血病等)、甲状腺肿瘤、输尿管肿瘤、膀胱肿瘤、胆囊癌、胆管癌、绒毛膜上皮癌或儿科肿瘤(尤因家族性肉瘤、维尔姆斯肉瘤、横纹肌肉瘤、血管肉瘤、胚胎睾丸癌、成神经细胞瘤、视网膜母细胞瘤、肝胚细胞瘤、肾母细胞瘤等)和所述癌症的组合。In a preferred embodiment, a compound of formula A, formula B, formula C, formula I, formula II, formula III, formula IV, formula V or formula VI Or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, or a pharmaceutical composition comprising the same, for use in the manufacture of a medicament for the treatment of a cancer or an infectious disease, wherein the cancer comprises But not limited to melanoma, brain tumors (gliomas with malignant astroglia and oligodendroglioma), esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, colorectal cancer (colon cancer) , rectal cancer, etc.), lung cancer (non-small cell lung cancer, small cell lung cancer, primary or metastatic squamous cell carcinoma, etc.), kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma, sarcoma, Osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer (cervical cancer, endometrial cancer, etc.), head and neck cancer (maxillary cancer, laryngeal cancer, pharyngeal cancer, tongue cancer, intraoral cancer) Etc., multiple myeloma, malignant lymphoma (reticulocyte sarcoma, lymphosarcoma) , Hodgkin's lymphoma, etc., polycythemia vera, leukemia (acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, etc.), thyroid neoplasms, ureteral tumors, bladder tumors, gallbladder cancer, Cholangiocarcinoma, chorionic epithelial cancer or pediatric tumor (Ewing familial sarcoma, Wilms' sarcoma, rhabdomyosarcoma, angiosarcoma, embryonic testicular cancer, neuroblastoma, retinoblastoma, hepatic blastoma, nephroblast Combination of tumors, etc.) and said cancer.
在另一个优选的实施方案中,一种如通式A、通式B、通式C、通式I、通式II、通式III、通式IV、通式V或通式VI所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药或包含其的药物组合物在制备用于治疗癌症或感染性疾病的药物中的应用,其中所述感染性疾病包括但不限于细菌、病毒和真菌感染。In another preferred embodiment, a formula of formula A, formula B, formula C, formula I, formula II, formula III, formula IV, formula V or formula VI Use of a compound, or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, or a pharmaceutical composition comprising the same, in the manufacture of a medicament for the treatment of a cancer or an infectious disease, wherein the infection Sexual diseases include, but are not limited to, bacterial, viral, and fungal infections.
在一个具体的实施方案中,一种如通式A、通式B、通式C、通式I、通式II、通式III、通式IV、通式V或通式VI所示的化合物对结肠癌具有显著的抑制作用。In a specific embodiment, a compound of Formula A, Formula B, Formula C, Formula I, Formula II, Formula III, Formula IV, Formula V, or Formula VI It has a significant inhibitory effect on colon cancer.
术语说明:Explanation of terms:
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。Terms used in the specification and claims have the following meanings unless stated to the contrary.
本发明化合物中的“氢”、“碳”包括其所有同位素。同位素应理解为包括具有相同原子数但具有不同质量数的那些原子,例如氢的同位素包括氚和氘,碳的同位素包括 13C和 14C。 "Hydrogen" and "carbon" in the compounds of the present invention include all isotopes thereof. Isotopes are understood to include those atoms having the same number of atoms but having different mass numbers, such as isotopes of hydrogen including lanthanum and cerium, and isotopes of carbon including 13 C and 14 C.
术语“立体异构体”是指原子组成及连接方式相同,而其三维空间排列不同的分子,它包括光学异构体、几何异构体(又叫顺反异构体),“手性”是具有与其镜像不能重叠性质的分子;而“非手性”是指与其镜像可以重叠的分子。光学异构体分为对映异构体和非对映异构体。“对映异构体”是指一个化合物的两个不能重叠但互成镜像关系的异构体。“非对映异构体”是指有两个或多个手性中性并且其分子不互为镜像的立体异构体。非对映异构体具有不同的物理性质,如熔点、沸点、光谱性质和反应性。非对映异构体混合物可通过高分辨分析操作如电泳和色谱,例如HPLC来分离。The term "stereoisomer" refers to a molecule having the same atomic composition and attachment in the same manner, but having a three-dimensional arrangement differently, including optical isomers, geometric isomers (also known as cis-trans isomers), "chiral" It is a molecule that has properties that cannot overlap with its mirror image; "non-chiral" refers to a molecule that can overlap with its mirror image. Optical isomers are divided into enantiomers and diastereomers. "Enantiomer" refers to two isomers of a compound that are not superimposable but are mirror images of each other. "Diastereomer" refers to a stereoisomer that has two or more chiral neutralities and whose molecules are not mirror images of each other. Diastereomers have different physical properties such as melting point, boiling point, spectral properties and reactivity. The mixture of diastereomers can be separated by high resolution analytical procedures such as electrophoresis and chromatography, such as HPLC.
本发明的化合物所有的立体异构形式,包括但不限于非对映体,对映异构体,顺反异构体,和它们的混合物,如外消旋混合物。很多有机化合物都以光学活性形式存在,即它们有能力旋转平面偏振光的平面。在描述光学活性化合物时,前缀D、L或R、S用来表示分子手性中心的绝对构型。前缀D、L或(+)、(-)用来命名化合物平面偏振光旋转的符号,(-)或L是指化合物是左 旋的,前缀(+)或D是指化合物是右旋的。这些立体异构体的化学结构是相同的,但是它们的立体结构不一样。特定的立体异构体可以是对映体,异构体的混合物通常称为对映异构体混合物。50:50的对映体混合物被称为外消旋混合物或外消旋体,这可能导致化学反应过程中没有立体选择性或立体定向性。术语“外消旋混合物”和“外消旋体”是指等摩尔的两个对映异构体的混合物,缺乏光学活性。All stereoisomeric forms of the compounds of the invention include, but are not limited to, diastereomers, enantiomers, cis and trans isomers, and mixtures thereof, such as racemic mixtures. Many organic compounds exist in optically active forms, that is, they have the ability to rotate planes of plane polarized light. In describing optically active compounds, the prefix D, L or R, S is used to indicate the absolute configuration of the molecular chiral center. The prefix D, L or (+), (-) is used to designate the sign of the plane-polarized light rotation of the compound, (-) or L means that the compound is left-handed, and the prefix (+) or D means that the compound is right-handed. The chemical structures of these stereoisomers are the same, but their stereostructures are different. A particular stereoisomer can be an enantiomer, and a mixture of isomers is often referred to as a mixture of enantiomers. The 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may result in no stereoselectivity or stereospecificity during the chemical reaction. The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomers that lack optical activity.
依据起始物料和方法的选择,本发明化合物可以以可能的异构体中的一个或它们的混合物,例如外消旋体和非对应异构体混合物(这取决于不对称碳原子的数量)的形式存在。光学活性的(R)-或(S)-异构体可使用手性合成子或手性试剂制备,或使用常规技术拆分。Depending on the choice of starting materials and methods, the compounds of the invention may be one of the possible isomers or mixtures thereof, such as racemates and mixtures of diastereomers (depending on the number of asymmetric carbon atoms) The form exists. Optically active (R)- or (S)-isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
所得的任何立体异构体的混合物可以依据组分物理化学性质上的差异被分离成纯的或基本纯的几何异构体,对映异构体,非对映异构体,例如,通过色谱法和/或分步结晶法。The resulting mixture of any stereoisomers can be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, for example, by chromatography, depending on the difference in physicochemical properties of the components. Method and / or step crystallization.
本发明的“药学上可接受的盐”是指化合物与无机和/或有机酸和碱形式的酸式和/或碱式盐,也包括两性离子盐(内盐)。本发明化合物含有氨基酸侧链、氨基酸残基,因此其可以形成内盐,也可以与其他的无机无机和/或有机酸和碱形式相应的盐。The "pharmaceutically acceptable salt" of the present invention means an acid and/or a basic salt of a compound with an inorganic and/or organic acid and a base form, and also a zwitterionic salt (internal salt). The compound of the present invention contains an amino acid side chain and an amino acid residue, so that it can form an internal salt or a salt corresponding to other inorganic inorganic and/or organic acid and base forms.
本发明的“溶剂合物”是指通过与溶剂分子配位形成固态或液态的配合物的本发明化合物的形式。水合物是溶剂合物的特殊形式,其中与水发生配位。在本发明范围内,溶剂合物优选是水合物。The "solvate" of the present invention means a form of the compound of the present invention which forms a solid or liquid complex by coordination with a solvent molecule. Hydrates are a special form of solvates in which coordination occurs with water. Within the scope of the invention, the solvate is preferably a hydrate.
本发明的“结晶”是指本发明所述的化合物形成的各种固体形态,包括晶型、无定形。"Crystalline" as used in the present invention refers to various solid forms formed by the compounds of the present invention, including crystalline forms and amorphous forms.
本发明的“前药”是指在生物体的生理条件下,由于与酶、胃酸等反应而转化成本发明的化合物,即通过酶的氧化、还原、水解等转化成本发明的化合物和/或通过胃酸等的水解反应等转化成本发明的化合物的化合物。The "prodrug" of the present invention refers to a compound which is converted into a compound of the present invention by a reaction with an enzyme, gastric acid or the like under physiological conditions of a living body, that is, a compound which is converted into the invention by oxidation, reduction, hydrolysis or the like of the enzyme and/or A compound which is converted into a compound of the invention by a hydrolysis reaction such as gastric acid or the like.
本发明的“烷基”是指直链或支链的饱和烃基,优选为C 1-8烷基,更优选C 1-6烷基。烷基的非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基。 The "alkyl group" of the present invention means a linear or branched saturated hydrocarbon group, preferably a C 1-8 alkyl group, more preferably a C 1-6 alkyl group. Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropane 1,1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl -2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-Dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-glycol Base, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2,3-dimethylpentyl, 2,4-dimethylpentyl, 2,2- Dimethylpentyl, 3,3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2,3-dimethylhexyl, 2,4-dimethyl Hexyl, 2,5-dimethylhexyl, 2,2-dimethylhexyl, 3,3-dimethylhexyl, 4,4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl.
本发明的“环烷基”是指饱和或者部分不饱和单环或多环取代基,其包括3-20个碳原子,优选4-13个碳原子。单环烷基的非限制性实施例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等,优选为环戊基、环己基。多环烷基包括螺环、稠环和桥环的环烷基。"Cycloalkyl" as used in the present invention refers to a saturated or partially unsaturated monocyclic or polycyclic substituent comprising from 3 to 20 carbon atoms, preferably from 4 to 13 carbon atoms. Non-limiting examples of monocycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene The group, the cyclooctyl group and the like are preferably a cyclopentyl group or a cyclohexyl group. Polycycloalkyl groups include spiro, fused, and bridged cycloalkyl groups.
本发明的“氮杂环烷基”是指包含一个氮原子和多个碳原子组成的饱和环,氮杂环烷基优选为氮杂C 3-20环烷基,更优选为氮杂C 4-13环烷基。更优选为氮杂C 3-6环烷基。氮杂环烷基基团非限制性实施例包括吡咯烷基和哌啶基基团。 The "azacycloalkylene group" of the present invention means a saturated ring comprising a nitrogen atom and a plurality of carbon atoms, and the azacycloalkylene group is preferably aza C 3-20 cycloalkyl group, more preferably aza C 4 -13 cycloalkyl. More preferably, it is an aza C 3-6 cycloalkyl group. Non-limiting examples of azacycloalkyl groups include pyrrolidinyl and piperidinyl groups.
本发明的“氧杂环烷基”是指由一个氧和多个碳原子组成的饱和环。氧杂环烷基优选为氧杂C 3-20环烷基,更优选为氧杂C 4-13环烷基。更优选为氧杂C 3-6环烷基。 The "oxyheterocycloalkyl group" of the present invention means a saturated ring composed of one oxygen and a plurality of carbon atoms. The oxacycloalkyl group is preferably an oxa C 3-20 cycloalkyl group, more preferably an oxa C 4-13 cycloalkyl group. More preferably, it is an oxa C 3-6 cycloalkyl group.
本发明的“内酯环烷基”是指由一个酯基
Figure PCTCN2018084352-appb-000039
和多个碳原子组成的饱和环。内酯环烷基优选为内酯C 3-20环烷基,更优选为内酯C 4-13环烷基。更优选为内酯C 3-6环烷基。
The "lactone cycloalkyl group" of the present invention means an ester group
Figure PCTCN2018084352-appb-000039
A saturated ring composed of multiple carbon atoms. The lactone cycloalkyl group is preferably a lactone C 3-20 cycloalkyl group, more preferably a lactone C 4-13 cycloalkyl group. More preferred is a lactone C 3-6 cycloalkyl group.
本发明的“烷基羟基”是指-烷基-OH。The "alkyl hydroxy group" of the present invention means -alkyl-OH.
本发明的“卤素”是指氟、氯、溴、碘。The "halogen" of the present invention means fluorine, chlorine, bromine or iodine.
本发明的“氨基”是指-NH 2、-NH(烷基)、-N(烷基)(烷基)。 The "amino group" of the present invention means -NH 2 , -NH(alkyl), -N(alkyl)(alkyl).
本发明的“硝基”是指-NO 2The "nitro group" of the present invention means -NO 2 .
本发明的“氰基”是指-CN。The "cyano group" of the present invention means -CN.
本发明的羟“羟基”是指-OH。The hydroxy "hydroxy group" of the present invention means -OH.
本发明的“苄基”是指-CH 2-苯基,简写为“Bn”。 The "benzyl group" of the present invention means -CH 2 -phenyl group, abbreviated as "Bn".
本发明的“叔丁氧羰基”是指-(O)CO( tBu),简写为“Boc”。 "Tert-butoxycarbonyl group" in the present invention means - (O) CO (t Bu ), abbreviated as "Boc."
本发明的“Fmoc-”是指芴甲氧羰基;The "Fmoc-" of the present invention means a fluorenylmethoxycarbonyl group;
本发明的“氨基酸”是指含有氨基的羧酸,氨基连在α-碳上的为α-氨基酸,结构通式表示为CH(COOH)(NH 2)-侧链。本发明的“L-氨基酸”指的是α-氨基酸的α-碳原子是左旋的;相反地,“D-氨基酸”指的是通式结构CH(COOH)(NH 2)-侧链的α-碳原子是右旋的。除甘氨酸外,其它蛋白质氨基酸的α-碳原子均为不对称碳原子(即与α-碳原子键合的四个取代基各不相同),因此氨基酸可以有立体异构体,即可以有不同的构型(D-型与L-型两种构型)。 The "amino acid" of the present invention means an amino group-containing carboxylic acid, and the amino group attached to the α-carbon is an α-amino acid, and the structural formula is represented by a CH(COOH)(NH 2 )- side chain. The "L-amino acid" of the present invention means that the α-carbon atom of the α-amino acid is left-handed; conversely, the "D-amino acid" refers to the α of the general structure CH(COOH)(NH 2 )-side chain - The carbon atom is right-handed. In addition to glycine, the α-carbon atoms of other protein amino acids are asymmetric carbon atoms (ie, the four substituents bonded to the α-carbon atom are different), so the amino acids may have stereoisomers, ie, may be different Configuration (D-type and L-type two configurations).
氨基酸的非限制性实例包括丙氨酸(Ala)、精氨酸(Arg)、天冬酰胺(Asn)、天冬氨酸(Asp)、半胱氨酸(Cys)、谷氨酰胺(Gln)、谷氨酸(Glu)、甘氨酸(Gly)、组氨酸(His)、异亮氨酸(Ile)、亮氨酸(Leu)、赖氨酸(Lys)、蛋氨酸(Met)、苯丙氨酸(Phe)、脯胺酸(Pro)、丝氨酸(Ser)、苏氨酸(Thr)、色氨酸(Trp)、酪氨酸(Tyr)、缬氨酸(Val)。Non-limiting examples of amino acids include alanine (Ala), arginine (Arg), asparagine (Asn), aspartic acid (Asp), cysteine (Cys), glutamine (Gln) , glutamic acid (Glu), glycine (Gly), histidine (His), isoleucine (Ile), leucine (Leu), lysine (Lys), methionine (Met), phenylalanine Acid (Phe), proline (Pro), serine (Ser), threonine (Thr), tryptophan (Trp), tyrosine (Tyr), valine (Val).
本发明的“氨基酸侧链”是指氨基酸的组成部分,氨基酸结构通式表示为CH(COOH)(NH 2)-R,R即代表氨基酸侧链,例如丙氨酸的结构为CH(COOH)(NH 2)-CH 3,氨基酸侧链即为-CH 3;丝氨酸的结构为CH(COOH)(NH 2)-CH 2OH,氨基酸侧链即为-CH 2OH;苏氨酸的结构为CH(COOH)(NH 2)-CH(OH)(CH 3),氨基酸侧链即为-CH(OH)(CH 3)。 The "amino acid side chain" of the present invention means a component of an amino acid, and the amino acid structural formula is represented by CH(COOH)(NH 2 )-R, and R represents an amino acid side chain, for example, the structure of alanine is CH (COOH). (NH 2 )-CH 3 , the amino acid side chain is -CH 3 ; the structure of serine is CH(COOH)(NH 2 )-CH 2 OH, the amino acid side chain is -CH 2 OH; the structure of threonine is CH(COOH)(NH 2 )-CH(OH)(CH 3 ), the amino acid side chain is -CH(OH)(CH 3 ).
本发明的“氨基酸残基”是指与母体氨基酸结构相比,缺少了一部分结构,是不完整的氨基酸,例如氨基酸氨基上的一个氢原子被一个化学键替代与其他原子连接成键,或氨基酸的-OH被化学键代替与其他原子连接成键。如丙氨酸的结构为
Figure PCTCN2018084352-appb-000040
那么其氨基酸残基可以为
Figure PCTCN2018084352-appb-000041
也可以为
Figure PCTCN2018084352-appb-000042
The "amino acid residue" of the present invention means that a part of the structure is absent compared to the structure of the parent amino acid, and is an incomplete amino acid. For example, a hydrogen atom on the amino group of the amino acid is replaced by a chemical bond to bond with another atom, or an amino acid. -OH is replaced by a chemical bond to bond with other atoms. Such as the structure of alanine
Figure PCTCN2018084352-appb-000040
Then its amino acid residue can be
Figure PCTCN2018084352-appb-000041
Can also be
Figure PCTCN2018084352-appb-000042
本发明的“保护基”是为了使分子其它部位进行反应时氮原子和氧原子保持不变,用易于脱去的基团对其进行保护。本发明的保护基包括氨基保护基和羟基保护基,本发明的氨基保护基 是指防止或阻止氨基参与下一步反应的基团,直到保护基被去除,非限制性实例包含甲酰基、烷基羰基、烷氧基羰基、苯甲酰基、芳烷基羰基、芳烷氧基羰基、三苯基甲基、邻苯二甲酰基、N,N-二甲基氨基亚甲基、取代的甲硅烷基、叔丁氧羰基、苄氧羰基等。本发明的羟基保护基是指防止或阻止羟基参与下一步反应的基团,直到保护基被去除。羟基保护基团的例子包括乙酰基、烯丙基、苯甲酰基、苄基、β-甲氧基乙氧基甲基、甲氧基甲基、二甲氧基三苯甲基[二-(4-甲氧基苯基)苯基甲基]、甲氧基三苯基[(4-甲氧基苯基)二苯基甲基]、对甲氧基苄基醚、甲基硫甲基、三甲基乙酰基、四氢吡喃基、三苯基甲基、硅基(例如:三甲基硅基、叔丁基二甲基硅基、叔丁基二苯基硅基、三异丙基硅氧基甲基和三异丙基硅基)。其他的例子包括烷基基团,如甲基和叔丁基,以及其他的醚,如乙氧基乙基。The "protecting group" of the present invention is such that the nitrogen atom and the oxygen atom remain unchanged in the reaction of other parts of the molecule, and are protected by a group which is easy to remove. The protecting group of the present invention includes an amino protecting group and a hydroxy protecting group, and the amino protecting group of the present invention means a group which prevents or prevents the amino group from participating in the next reaction until the protecting group is removed, and a non-limiting example includes a formyl group, an alkyl group. Carbonyl, alkoxycarbonyl, benzoyl, aralkylcarbonyl, aralkoxycarbonyl, triphenylmethyl, phthaloyl, N,N-dimethylaminomethylene, substituted monosilane Base, tert-butoxycarbonyl, benzyloxycarbonyl and the like. The hydroxy protecting group of the present invention means a group which prevents or prevents the hydroxyl group from participating in the next reaction until the protecting group is removed. Examples of the hydroxy protecting group include acetyl, allyl, benzoyl, benzyl, β-methoxyethoxymethyl, methoxymethyl, dimethoxytrityl [di-( 4-methoxyphenyl)phenylmethyl], methoxytriphenyl[(4-methoxyphenyl)diphenylmethyl], p-methoxybenzyl ether, methylthiomethyl , trimethylacetyl, tetrahydropyranyl, triphenylmethyl, silicon (eg, trimethylsilyl, tert-butyldimethylsilyl, tert-butyldiphenylsilyl, triiso) Propylsilyloxymethyl and triisopropylsilyl). Other examples include alkyl groups such as methyl and t-butyl groups, as well as other ethers such as ethoxyethyl groups.
具体实施方式:detailed description:
以下实施例中用到的有机溶剂简称如下:The organic solvents used in the following examples are abbreviated as follows:
NMM:N-甲基吗啡啉;NMM: N-methylmorpholine;
HATU:2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯;HATU: 2-(7-oxobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate;
TPP:三苯基膦;TPP: triphenylphosphine;
HOBT:1-羟基苯并三唑;HOBT: 1-hydroxybenzotriazole;
EDC:1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐;EDC: 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride;
DAST:二乙胺基三氟化硫;DAST: diethylaminosulfur trifluoride;
TBDMSCl:叔丁基二甲基氯硅烷;TBDMSCl: tert-butyldimethylchlorosilane;
Fmoc-Asn(Trt)-OH:N-芴甲氧羰基-L-天冬酰胺。Fmoc-Asn(Trt)-OH: N-fluorenylmethoxycarbonyl-L-asparagine.
实施例1:((1-(5-((1S,2S)-1-氨基-2-羟丙基)-1,3,4-噁二唑-2-基)-3-(羧甲基)环丁基)氨基甲酰基)-L-丝氨酸的制备Example 1: ((1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)-3-(carboxymethyl) Preparation of cyclobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000043
Figure PCTCN2018084352-appb-000043
步骤1:1–((叔丁氧基羰基)氨基)-3-氧代环丁烷-1-甲酸的制备Step 1:1 - Preparation of ((tert-Butoxycarbonyl)amino)-3-oxocyclobutane-1-carboxylic acid
Figure PCTCN2018084352-appb-000044
Figure PCTCN2018084352-appb-000044
将1–((叔丁氧羰基)氨基)-3-氧代环丁烷甲酸乙酯(50mg,0.19mmol)溶于甲醇(1mL)、水(0.5mL)的混合溶剂中,加入NaOH(9.3mg,0.23mmol),室温反应半小时,TLC板监测反应(碘熏、溴甲酚绿显色),反应完全。减压蒸馏除去甲醇,剩余液体用1M柠檬酸调pH至4。用EA和水分液,有机相用食盐水洗涤,无水Na 2SO 4干燥。减压蒸馏,硅胶柱分离纯化(0-50%EA in PE)得白色固体40mg,收率90%。 Ethyl 1-((tert-butoxycarbonyl)amino)-3-oxocyclobutanecarboxylate (50 mg, 0.19 mmol) was dissolved in methanol (1 mL), water (0.5 mL) Mg, 0.23 mmol), reacted at room temperature for half an hour, and the reaction was monitored by TLC plate (iodine, bromocresol green color development), and the reaction was complete. Methanol was distilled off under reduced pressure, and the remaining liquid was adjusted to pH 4 with 1M citric acid. , Brine and the organic phase was washed with EA and washed with water, dried over anhydrous Na 2 SO 4. The mixture was evaporated under reduced pressure, and purified by silica gel column (0-50% EA in PE) to yield 40 mg of white solid.
步骤2:1-((叔丁氧基羰基)氨基)-3-氧代环丁烷-1-甲酸苄酯的制备Step 2: Preparation of benzyl 1-((tert-butoxycarbonyl)amino)-3-oxocyclobutane-1-carboxylate
Figure PCTCN2018084352-appb-000045
Figure PCTCN2018084352-appb-000045
将1–((叔丁氧基羰基)氨基)-3-氧代环丁烷-1-甲酸(2g,8.73mmol)溶于DMF(20mL)中,加入K 2CO 3(1.81g,13.09mmol),BnCl(1.32g,10.48mmol)。置于65℃油浴上,加热反应2h,TLC板监测反应(碘熏显色),反应完全。用EA和水分液,有机相用食盐水洗涤,无水Na 2SO 4干燥。减压蒸馏,硅胶柱分离纯化(0-10%EA in PE)得淡白色固体2.1g,收率75.5%。 1 - ((tert-butoxycarbonyl) amino) -3-oxo-1 -carboxylic acid (2g, 8.73mmol) was dissolved in DMF (20mL) was added K 2 CO 3 (1.81g, 13.09mmol ), BnCl (1.32 g, 10.48 mmol). Placed on a 65 ° C oil bath, heated for 2 h, TLC plate monitoring reaction (iodine smoked color), the reaction is complete. , Brine and the organic phase was washed with EA and washed with water, dried over anhydrous Na 2 SO 4. The mixture was evaporated under reduced pressure and purified (yield: EtOAc (EtOAc)
步骤3:1-((叔丁氧基羰基)氨基)-3-(2-乙氧基-2-氧代亚乙基)环丁烷-1-甲酸苄酯的制备Step 3: Preparation of benzyl 1-((tert-butoxycarbonyl)amino)-3-(2-ethoxy-2-oxoethylidene)cyclobutane-1-carboxylate
Figure PCTCN2018084352-appb-000046
Figure PCTCN2018084352-appb-000046
将1-((叔丁氧基羰基)氨基)-3-氧代环丁烷-1-甲酸苄酯(1g,3.13mmol)溶于甲苯(15mL)中,加入(三苯基膦)乙酸乙酯(1.53g,4.38mmol),加热回流,反应4h,TLC板监测反应(碘熏显色),反应完全。用EA和水分液,有机相用食盐水洗涤,无水Na 2SO 4干燥。减压蒸馏,硅胶柱分离纯化(0-10%EA in PE)得淡黄色油状物0.8g,收率66%。 Benzyl 1-((tert-butoxycarbonyl)amino)-3-oxocyclobutane-1-carboxylate (1 g, 3.13 mmol) was dissolved in toluene (15 mL) and (triphenylphosphine)acetate The ester (1.53 g, 4.38 mmol) was heated under reflux for 4 h, and the reaction was monitored by TLC (yield of iodine) and the reaction was completed. , Brine and the organic phase was washed with EA and washed with water, dried over anhydrous Na 2 SO 4. The mixture was evaporated under reduced pressure, and purified (yield: EtOAc (EtOAc)
步骤4:1-氨基-3-(2-乙氧基-2-氧代亚乙基)环丁烷-1-甲酸苄酯的制备Step 4: Preparation of benzyl 1-amino-3-(2-ethoxy-2-oxoethylidene)cyclobutane-1-carboxylate
Figure PCTCN2018084352-appb-000047
Figure PCTCN2018084352-appb-000047
将1-((叔丁氧基羰基)氨基)-3-(2-乙氧基-2-氧代亚乙基)环丁烷-1-甲酸苄酯(40mg,0.1mmol)溶于二氯甲烷(1mL)、三氟乙酸(1mL)的混合溶剂中,室温反应2h,TLC板监测反应(碘熏显色),反应完全。减压蒸馏除去多余的三氟乙酸,加入20%的Na 2CO 3水溶液,搅拌15min,用EA分液,有机相用食盐水洗涤,无水Na 2SO 4干燥。减压蒸馏得淡黄色油状物30mg,收率100%。 Benzyl 1-((tert-butoxycarbonyl)amino)-3-(2-ethoxy-2-oxoethylidene)cyclobutane-1-carboxylate (40 mg, 0.1 mmol) was dissolved in dichloro In a mixed solvent of methane (1 mL) and trifluoroacetic acid (1 mL), the mixture was reacted at room temperature for 2 h, and the reaction was monitored by a TLC plate (yield of iodine), and the reaction was completed. Distillation under reduced pressure to remove excess trifluoroacetic acid was added 20% Na 2 CO 3 solution, stirred for 15min, with EA liquid separation, the organic phase was washed with brine, dried over anhydrous Na 2 SO 4. Distillation under reduced pressure gave a pale yellow oil (yield: 30%).
步骤5:1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-乙氧基-2-氧代亚乙基)环丁烷-1-甲酸苄酯的制备Step 5: 1-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(2-ethoxy-2-oxoethylidene)cyclobutane-1-carboxylic acid Preparation of benzyl ester
Figure PCTCN2018084352-appb-000048
Figure PCTCN2018084352-appb-000048
将1-氨基-3-(2-乙氧基-2-氧代亚乙基)环丁烷-1-甲酸苄酯(50mg,0.17mmol)溶于1,4-二氧六环(1mL)、水(0.2mL)的混合溶剂中,加入Na 2CO 3(22mg,0.21mmol,),置于冰浴,加入FmocCl(53.7mg,0.21mmol),加毕,逐渐升至室温。反应1h,TLC板监测反应(碘熏显色),反应完全。用EA和水分液,有机相用食盐水洗涤,无水Na 2SO 4干燥。减压蒸馏,硅胶柱分离纯化(0-10%EA in PE)得无色油状物64mg,收率72%。 Benzyl 1-amino-3-(2-ethoxy-2-oxoethylidene)cyclobutane-1-carboxylate (50 mg, 0.17 mmol) was dissolved in 1,4-dioxane (1 mL) To a mixed solvent of water (0.2 mL), Na 2 CO 3 (22 mg, 0.21 mmol) was added, and the mixture was placed in an ice bath, and FmocCl (53.7 mg, 0.21 mmol) was added thereto, and the mixture was gradually warmed to room temperature. After 1 h of reaction, the reaction was monitored by TLC plate (iodine smoked color development) and the reaction was completed. , Brine and the organic phase was washed with EA and washed with water, dried over anhydrous Na 2 SO 4. The mixture was evaporated under reduced pressure and purified (yield: EtOAc)
步骤6:1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-乙氧基-2-氧代乙基)环丁烷-1-甲酸的制备Step 6: 1-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(2-ethoxy-2-oxoethyl)cyclobutane-1-carboxylic acid preparation
Figure PCTCN2018084352-appb-000049
Figure PCTCN2018084352-appb-000049
将1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-乙氧基-2-氧代亚乙基)环丁烷-1-甲酸苄酯(800mg,1.56mmol)溶于甲醇(10mL)中,加入Pd/C(80mg,10%,w/w),氢化2h,TLC板监测反应,反应完全。用硅藻土过滤除去Pd/C,滤液减压蒸馏得无色油状物496mg,收率75%。Benzyl 1-((((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(2-ethoxy-2-oxoethylidene)cyclobutane-1-carboxylate (800 mg, 1.56 mmol) was dissolved in MeOH (10 mL). Pd / C (80 mg, 10%, w/w). The Pd/C was removed by filtration through Celite, and the filtrate was evaporated under reduced pressure to yield 496 g of colorless oil.
步骤7:2-(3-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-(N-(叔丁氧基)-O-(叔丁基)-L-别苏氨酸基)肼基-1-羰基)环丁基)乙酸乙酯的制备Step 7: 2-(3-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(2-(N-(tert-butoxy)-O-(tert-butyl) Preparation of ethyl-L-betathreonyl)nonyl-1-carbonyl)cyclobutyl)acetate
Figure PCTCN2018084352-appb-000050
Figure PCTCN2018084352-appb-000050
将1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-乙氧基-2-氧代乙基)环丁烷-1-甲酸(790mg,1.86mmol)溶于DMF(15mL)中,依次加入((2S,3S)-3-(叔丁氧基)-1-肼基-1-氧丁基-2-基)氨基甲酸叔丁酯(539mg,1.86mmol),NMM(188.6mg,4.65mmol),HATU(710mg,1.86mmol),室温反应2h,TLC板监测反应,反应完全。将反应液缓慢倒入冰水中,析出白色固体,抽滤,水洗得白色固体800mg,收率69%。1-((((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(2-ethoxy-2-oxoethyl)cyclobutane-1-carboxylic acid (790 mg, 1.86 mmol) was dissolved in DMF (15 mL) and ((2S,3S)-3-(tert-butoxy)-1-indol-1-yloxy-2-yl)carbamic acid tert-butyl ester ( 539 mg, 1.86 mmol), NMM (188.6 mg, 4.65 mmol), HATU (710 mg, 1.86 mmol), reacted at room temperature for 2 h, and the reaction was monitored by TLC plate. The reaction solution was slowly poured into ice water, and a white solid was precipitated, filtered, and washed with water to give a white solid.
步骤8:2-(3-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧基羰基)氨基)丙基)-1,3,4-噁二唑-2-基)环丁基)乙酸乙酯的制备Step 8: 2-(3-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(5-((1S,2S)-2-(tert-butoxy)- Preparation of ethyl 1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazol-2-yl)cyclobutyl)acetate
Figure PCTCN2018084352-appb-000051
Figure PCTCN2018084352-appb-000051
将2-(3-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-(N-(叔丁氧基)-O-(叔丁基)-L-别苏氨酸基)肼基-1-羰基)环丁基)乙酸乙酯(660mg,0.95mmol)溶于DMF(8mL)、THF(2mL)的混合溶剂中,置于冰浴,加入TPP(997mg,3.8mmol),I 2(966mg,3.8mmol),搅拌30min至I 2完全溶解,加入Et 3N(768mg,7.6mmol)。加毕,撤走冰浴,室温反应3h,TLC板监测反应,反应完全。用EA和水分液,有机相用饱和硫代硫酸钠、食盐水洗涤,无水Na 2SO 4干燥。减压蒸馏,硅胶柱分离纯化(0-25%EA in PE)得无色油状物418mg,收率65%。 2-(3-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(2-(N-(tert-butoxy)-O-(tert-butyl)- Ethyl L-(sub-threonyl) decyl-1-carbonyl)cyclobutyl)acetate (660 mg, 0.95 mmol) was dissolved in a mixed solvent of DMF (8 mL), THF (2 mL) TPP (997mg, 3.8mmol), I 2 (966mg, 3.8mmol), stirred for 30min until complete dissolution of I 2, was added Et 3 N (768mg, 7.6mmol) . After the addition, the ice bath was removed and reacted at room temperature for 3 h. The reaction was monitored by TLC plate and the reaction was complete. Solution with EA and water, the organic phase is washed with saturated sodium thiosulfate, brine, dried over anhydrous Na 2 SO 4. The mixture was evaporated under reduced pressure and purified by silica gel column (0-25% EA
步骤9:2-(3-氨基-3-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧基羰基)氨基)丙基)-1,3,4-噁二唑-2-基)环丁基)乙酸乙酯的制备Step 9: 2-(3-Amino-3-(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3 Of 4-(oxadiazol-2-yl)cyclobutyl)acetate
Figure PCTCN2018084352-appb-000052
Figure PCTCN2018084352-appb-000052
将2-(3-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧基羰基)氨基)丙基)-1,3,4-噁二唑-2-基)环丁基)乙酸乙酯(606mg,0.9mmol)溶于DCM(5mL)、加入Et 2NH(355 mg,4.85mmol),室温反应6h,反应完全。减压蒸馏,硅胶柱分离纯化(0-30%EA in PE)得无色油状物270mg,收率65%。 2-(3-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(5-((1S,2S)-2-(tert-butoxy)-1-) Ethyl ((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazol-2-yl)cyclobutyl)acetate (606 mg, 0.9 mmol) dissolved in DCM (5 mL) 2 NH (355 mg, 4.85 mmol), reacted at room temperature for 6 h, the reaction was complete. The mixture was evaporated under reduced pressure, and purified (yield: EtOAc)
步骤10:O-(叔丁基)-N-((4-硝基苯氧基)羰基)-L-丝氨酸叔丁酯的制备Step 10: Preparation of O-(tert-butyl)-N-((4-nitrophenoxy)carbonyl)-L-serine tert-butyl ester
Figure PCTCN2018084352-appb-000053
Figure PCTCN2018084352-appb-000053
将O-(叔丁基)-L-丝氨酸叔丁酯(10g,0.046mol,1.0eq)溶于DCM(200mL),冰浴降温,于0℃下依次滴加Et 3N(9.32g,0.092mol 2.0eq)和4-硝基氯甲酸苯酯(9.25g,0.046mol,1.0eq)的DCM(60mL)溶液,加毕,撤去冰浴,搅拌反应3h。TLC板监测反应(碘熏),反应完全。二氯甲烷与水萃取,有机层用饱和Na 2CO 3洗涤三次,用柠檬酸调节pH至4,饱和食盐水洗涤。无水Na 2SO 4干燥。减压蒸馏,用石油醚洗涤,产物溶于石油醚,过滤,将滤液进行减压蒸馏得无色油状物10.5g,收率59.6%。 The O- (tert-butyl) -L- serine t-butyl ester (10g, 0.046mol, 1.0eq) was dissolved in DCM (200mL), cooled ice bath, added dropwise successively at 0 ℃ Et 3 N (9.32g, 0.092 A solution of mol 2.0 eq) and phenyl 4-nitrochloroformate (9.25 g, 0.046 mol, 1.0 eq) in DCM (60 mL). The reaction was monitored by TLC plate (iodine) and the reaction was complete. The organic layer was washed three times with saturated Na 2 CO 3 , and then adjusted to pH 4 with EtOAc. Dry with anhydrous Na 2 SO 4 . Distilled under reduced pressure, washed with petroleum ether, and the product was dissolved in petroleum ether, filtered, and the filtrate was evaporated under reduced pressure to yield 10.5 g of colorless oil.
步骤11:N-((1-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧羰基)氨基)丙基)-1,3,4-噁二唑-2-基)-3-(2-乙氧基-2-氧代乙基)环丁基)氨基甲酰基-O-(叔丁基)-L-丝氨酸叔丁酯的制备Step 11: N-((1-(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxole Preparation of tert-butyl 2-oxazol-2-yl)-3-(2-ethoxy-2-oxoethyl)cyclobutyl)carbamoyl-O-(tert-butyl)-L-serine
Figure PCTCN2018084352-appb-000054
Figure PCTCN2018084352-appb-000054
将2-(3-氨基-3-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧基羰基)氨基)丙基)-1,3,4-噁二唑-2-基)环丁基)乙酸乙基酯(2.11g,4.64mmol)溶于DMF(20mL)、加入Et 3N(0.93g,9.28mmol),置于0℃,加入O-(叔丁基)-N-((4-硝基苯氧基)羰基)-L-丝氨酸叔丁酯(2.13g,5.57mmol)。加毕,升至室温,室温反应10h,TLC板监测反应(碘熏),反应完全。减压蒸馏,硅胶柱分离纯化(0-50%EA in PE)得无色油状物2.2g,收率68%。 2-(3-Amino-3-(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4 - oxadiazol-2-yl) cyclobutyl) acetic acid ethyl ester (2.11g, 4.64mmol) was dissolved in DMF (20mL), was added Et 3 N (0.93g, 9.28mmol) , was placed 0 ℃, was added O -(tert-Butyl)-N-((4-nitrophenoxy)carbonyl)-L-serine tert-butyl ester (2.13 g, 5.57 mmol). After the addition, the temperature was raised to room temperature, and reacted at room temperature for 10 hours. The reaction was monitored by TLC plate (iodine), and the reaction was completed. The mixture was evaporated under reduced pressure and purified by silica gel column (0-50% EA in
步骤12:N-((1-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧羰基)氨基)丙基)-1,3,4-噁二唑-2-基)-3-(羧基)环丁基)氨基甲酰基-O-(叔丁基)-L-丝氨酸的制备Step 12: N-((1-(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxole Preparation of oxazol-2-yl)-3-(carboxy)cyclobutyl)carbamoyl-O-(tert-butyl)-L-serine
Figure PCTCN2018084352-appb-000055
Figure PCTCN2018084352-appb-000055
将N-((1-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧羰基)氨基)丙基)-1,3,4-噁二唑-2-基)-3-(2-乙氧基-2-氧代乙基)环丁基)氨基甲酰基-O-(叔丁基)-L-丝氨酸叔丁酯(1g,1.43mmol)溶于THF(15mL)、水(5mL)的混合溶剂中,置于0℃,加入LiOH(240mg,5.73mmol)。加毕,升至室温,室温反应5h,TLC板监测反应(碘熏),反应完全。减压蒸馏除去多余的THF,加1M的柠檬酸酸化至pH为4,二氯甲烷萃取,无水Na 2SO 4干燥。减压蒸馏,硅胶柱分离纯化(0-5%MeOH in DCM)得无色油状物150mg,收率17%。 N-((1-(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazole 2-yl)-3-(2-ethoxy-2-oxoethyl)cyclobutyl)carbamoyl-O-(tert-butyl)-L-serine tert-butyl ester (1 g, 1.43 mmol) Dissolved in a mixed solvent of THF (15 mL) and water (5 mL), and placed at 0 ° C, and added LiOH (240 mg, 5.73 mmol), added to room temperature, reacted at room temperature for 5 h, and monitored by TLC plate (iodine). The reaction was completed. The excess THF was distilled off under reduced pressure, and then acidified to pH 4 with 1M citric acid, extracted with dichloromethane, dried over anhydrous Na 2 SO 4 , evaporated under reduced pressure and purified by silica gel column (0-5% MeOH in DCM) gave a colorless oil of 150 mg, yield 17%.
步骤13:((1-(5-((1S,2S)-1-氨基-2-羟丙基)-1,3,4-噁二唑-2-基)-3-(羧甲基)环丁基)氨基甲酰基)-L-丝氨酸的制备Step 13: ((1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)-3-(carboxymethyl) Preparation of cyclobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000056
Figure PCTCN2018084352-appb-000056
将N-((1-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧羰基)氨基)丙基)-1,3,4-噁二唑-2-基)-3-(羧基)环丁基)氨基甲酰基-O-(叔丁基)-L-丝氨酸(110mg,0.179mmol)溶于DCM(4mL),室温下加入TFA(1.2mL)、三异丙基硅烷(催化量),室温搅拌反应5h。反应液旋干、加DCM、水,分液,有机相用水萃取数次,合并水相,再用DCM洗涤数次,冻干、制备分离,得白色固体16mg;收率:22.2%。 1H NMR(500MHz,D 2O):δ4.73(d,J=6.4Hz,1H),4.49–4.39(m,1H),4.19(t,J=4.5Hz,1H),3.89(dd,J=10.5,4.5Hz,2H),3.11–2.97(m,1H),2.73(t,J=10.8Hz,2H),2.56(t,J=7.4Hz,4H),1.40(s,3H).ESI-MS m/z:402.2[M+H] + N-((1-(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazole 2-yl)-3-(carboxy)cyclobutyl)carbamoyl-O-(tert-butyl)-L-serine (110 mg, 0.179 mmol) was dissolved in DCM (4 mL). , triisopropylsilane (catalytic amount), stirring reaction at room temperature for 5 h. The reaction solution was spun dry, added DCM, water, liquid separation, the organic phase was extracted several times with water, the aqueous phase was combined, washed several times with DCM, lyophilized , preparative separation, 16mg white solid; yield:. 22.2% 1 H NMR ( 500MHz, D 2 O): δ4.73 (d, J = 6.4Hz, 1H), 4.49-4.39 (m, 1H), 4.19 (t, J = 4.5 Hz, 1H), 3.89 (dd, J = 10.5, 4.5 Hz, 2H), 3.11 - 2.97 (m, 1H), 2.73 (t, J = 10.8 Hz, 2H), 2.56 (t, J=7.4 Hz, 4H), 1.40 (s, 3H). ESI-MS m/z: 402.2 [M+H] +
实施例2:((1-(5-((1S,2S)-1-氨基-2-羟丙基)-1,3,4-噁二唑-2-基)-3-(2-乙氧基-2-氧乙基)环丁基)氨基甲酰基)-L-丝氨酸的制备Example 2: ((1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)-3-(2-B) Preparation of oxy-2-oxoethyl)cyclobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000057
Figure PCTCN2018084352-appb-000057
合成路线:synthetic route:
Figure PCTCN2018084352-appb-000058
Figure PCTCN2018084352-appb-000058
将N-((1-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧羰基)氨基)丙基)-1,3,4-噁二唑-2-基)-3-(2-乙氧基-2-氧代乙基)环丁基)氨基甲酰基-O-(叔丁基)-L-丝氨酸叔丁酯(200mg,0.28mmol)溶于DCM(2mL),室温下加入TFA(2.1mL)、三异丙基硅烷(催化量),室温搅拌反应5h。反应液旋干、加DCM、水,分液,有机相用水萃取数次,合并水相,再用DCM洗涤数次,冻干、制备分离,得白色固体60mg,收率48.8%。 1H NMR(400MHz,D 2O):δ4.51(dd,J=8.7,6.3Hz,1H),4.22(dd,J=12.7,6.4Hz,1H),4.05(q,J=7.1Hz,2H),3.97(dt,J=13.6,4.7Hz,1H),3.69(dd,J=8.5,4.7Hz,2H),2.87(ddd,J=17.7,14.4,8.4Hz,1H),2.61–2.51(m,3H),2.48–2.33(m,1H),2.25–2.13(m,1H),1.19(dd,J=6.4,2.7Hz,3H),1.14(t,J=7.1Hz,3H).ESI-MS m/z:430.2[M+H] + N-((1-(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazole 2-yl)-3-(2-ethoxy-2-oxoethyl)cyclobutyl)carbamoyl-O-(tert-butyl)-L-serine tert-butyl ester (200 mg, 0.28 mmol) Dissolve in DCM (2mL), add TFA (2.1mL), triisopropylsilane (catalytic amount) at room temperature, stir the reaction at room temperature for 5h. Dissolve the reaction solution, add DCM, water, separate the liquid, extract the organic phase several times with water The aqueous phase was combined and washed with DCM several times, lyophilized and purified to give a white solid (yield: 48 mg, yield: 48.8%) 1 H NMR (400 MHz, D 2 O): δ 4.51 (dd, J = 8.7, 6.3 Hz, 1H), 4.22 (dd, J = 12.7, 6.4 Hz, 1H), 4.05 (q, J = 7.1 Hz, 2H), 3.97 (dt, J = 13.6, 4.7 Hz, 1H), 3.69 (dd, J = 8.5, 4.7 Hz, 2H), 2.87 (ddd, J = 17.7, 14.4, 8.4 Hz, 1H), 2.61 - 2.51 (m, 3H), 2.48 - 2.33 (m, 1H), 2.25 - 2.13 (m, 1H) ), 1.19 (dd, J = 6.4, 2.7 Hz, 3H), 1.14 (t, J = 7.1 Hz, 3H). ESI-MS m/z: 430.2 [M+H] +
实施例3:((1-(5-((1S,2S)-1-氨基-2-羟丙基)-1,3,4-噁二唑-2-基)-3-(2-氨基-2-氧乙基)环丁基)氨基甲酰基)-L-丝氨酸的制备Example 3: ((1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)-3-(2-amino) Preparation of -2-oxoethyl)cyclobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000059
Figure PCTCN2018084352-appb-000059
步骤1:3-(2-氨基-2-氧代亚乙基)-1-((叔丁氧基羰基)氨基)环丁烷-1-甲酸乙酯的制备Step 1: Preparation of ethyl 3-(2-amino-2-oxoethylidene)-1-((tert-butoxycarbonyl)amino)cyclobutane-1-carboxylate
Figure PCTCN2018084352-appb-000060
Figure PCTCN2018084352-appb-000060
将NaH(676mg,0.016mol,1.1eq)溶于10mL DMF中,置于-20℃低温反应器中,加入氨基甲酰甲基膦酸二乙酯(3g,0.015mol)的DMF溶液,滴毕,搅拌1h,向其中逐滴滴加1–((叔丁氧羰基)氨基)-3-氧代环丁烷甲酸乙酯(3.56g,0.013mol),滴毕,继续反应4小时,TLC板监测反应(碘熏),反应完全。乙酸乙酯、水萃取,有机层用饱和食盐水洗涤两次,无水Na 2SO 4干燥。减压蒸馏,硅胶柱分离纯化(P/E=1/2)得无色油状物3.02g,收率73%。 NaH (676 mg, 0.016 mol, 1.1 eq) was dissolved in 10 mL of DMF, placed in a -20 ° C low temperature reactor, and a solution of diethyl carbamoylphosphonate (3 g, 0.015 mol) in DMF was added. After stirring for 1 h, ethyl 1 -((tert-butoxycarbonyl)amino)-3-oxocyclobutanecarboxylate (3.56 g, 0.013 mol) was added dropwise dropwise, and the reaction was continued for 4 hours, TLC plate. The reaction was monitored (iodine) and the reaction was complete. The organic layer was washed twice with saturated brine and dried over anhydrous Na 2 SO 4 . The mixture was evaporated under reduced pressure and purified (yield: EtOAc) (yield:
步骤2:3-(2-氨基-2-氧代乙基)-1-((叔丁氧羰基)氨基)环丁烷-1-甲酸乙酯的制备Step 2: Preparation of ethyl 3-(2-amino-2-oxoethyl)-1-((tert-butoxycarbonyl)amino)cyclobutane-1-carboxylate
Figure PCTCN2018084352-appb-000061
Figure PCTCN2018084352-appb-000061
按实施例1中的步骤6的方法制得标题化合物,不同的是将原料1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-乙氧基-2-氧代亚乙基)环丁烷-1-甲酸苄酯替换为3-(2-氨基-2-氧代亚乙基)-1-((叔丁氧基羰基)氨基)环丁烷-1-甲酸乙酯。The title compound was obtained according to the procedure of Step 6 in Example 1, except that the starting material was 1-((((())))))))))) Replacement of benzyl 2-oxoethylidene)cyclobutane-1-carboxylate with 3-(2-amino-2-oxoethylidene)-1-((tert-butoxycarbonyl)amino) ring Butane-1-carboxylate.
步骤3:3-(2-氨基-2-氧代乙基)-1-((叔丁氧基羰基)氨基)环丁烷甲酸的制备Step 3: Preparation of 3-(2-amino-2-oxoethyl)-1-((tert-butoxycarbonyl)amino)cyclobutanecarboxylic acid
Figure PCTCN2018084352-appb-000062
Figure PCTCN2018084352-appb-000062
将乙基3-(2-氨基-2-氧代乙基)-1-((叔丁氧羰基)氨基)环丁烷甲酸乙酯(3.03g,0.01mol)溶于THF(80mL)中,加入LiOH水溶液(508mg,0.012mol,),室温反应5h,减压蒸馏,除去多余的THF,用1M的柠檬酸调pH至2,冻干,硅胶柱分离纯化(0-10%MeOH in DCM),将得到的产物直接投入下一步反应。Ethyl ethyl 3-(2-amino-2-oxoethyl)-1-((tert-butoxycarbonyl)amino)cyclobutanecarboxylate (3.03 g, 0.01 mol) was dissolved in THF (EtOAc) Add LiOH aqueous solution (508 mg, 0.012 mol,), react at room temperature for 5 h, distill off under reduced pressure, remove excess THF, adjust pH to 2 with 1 M citric acid, freeze-dry, and purify on silica gel column (0-10% MeOH in DCM) The obtained product is directly put into the next reaction.
步骤4:3-(2-氨基-2-氧代乙基)-1-羧基环丁胺盐酸盐Step 4: 3-(2-Amino-2-oxoethyl)-1-carboxycyclobutylamine hydrochloride
Figure PCTCN2018084352-appb-000063
Figure PCTCN2018084352-appb-000063
将3-(2-氨基-2-氧代乙基)-1-((叔丁氧基羰基)氨基)环丁烷甲酸粗品(190mg,0.69mmol)溶于2mL的1.5M HCl乙醇溶液中,室温反应3h。减压蒸馏,得粗品132mg,收率91%,将得到的产物直接投下一步反应。The crude 3-(2-amino-2-oxoethyl)-1-((tert-butoxycarbonyl)amino)cyclobutanecarboxylic acid (190 mg, 0.69 mmol) was dissolved in 2 mL of 1.5M EtOAc The reaction was carried out for 3 h at room temperature. Distillation under reduced pressure gave a crude product (132 mg, yield: 91%).
步骤5:((1-(5-((1S,2S)-1-氨基-2-羟丙基)-1,3,4-噁二唑-2-基)-3-(2-氨基-2-氧乙基)环丁基)氨基甲酰基)-L-丝氨酸的制备Step 5: ((1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)-3-(2-amino-) Preparation of 2-oxoethyl)cyclobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000064
Figure PCTCN2018084352-appb-000064
按实施例1中的步骤5、7、8、9、11、13的方法制得标题化合物,不同的是将步骤5中的原料1-氨基-3-(2-乙氧基-2-氧代亚乙基)环丁烷甲酸苄酯(50mg,0.17mmol)溶于1,4-二氧六环替换为3-(2-氨基-2-氧代乙基)-1-羧基环丁胺盐酸盐。 1H NMR(400MHz,D 2O):δ4.21–4.11(m,2H),3.89(d,J=6.4Hz,1H),3.81(dd,J=6.0,2.4Hz,2H),2.97–2.72(m,2H),2.69(dd,J=11.6,5.1Hz,2H),2.58–2.29(m,2H),2.09(ddd,J=31.0,12.2,7.5Hz,1H),1.37(t,J=5.7Hz,3H).ESI-MS m/z:401.3[M+H] + The title compound was obtained according to the procedure of Steps 5, 7, 8, 9, 11, and 13 in Example 1, except that the starting material in the step 5 was 1-amino-3-(2-ethoxy-2-oxo Benzylene) benzyl cyclobutanecarboxylate (50 mg, 0.17 mmol) was dissolved in 1,4-dioxane to 3-(2-amino-2-oxoethyl)-1-carboxycyclobutylamine Hydrochloride. 1 H NMR (400 MHz, D 2 O): δ 4.21 - 4.11 (m, 2H), 3.89 (d, J = 6.4 Hz, 1H), 3.81 (dd, J = 6.0, 2.4 Hz, 2H), 2.97 - 2.72 (m, 2H), 2.69 (dd, J = 11.6, 5.1 Hz, 2H), 2.58 - 2.29 (m, 2H), 2.09 (ddd, J = 31.0, 12.2, 7.5 Hz, 1H), 1.37 (t, J=5.7 Hz, 3H). ESI-MS m/z: 401.3 [M+H] +
实施例4:(((1s,3R)-1-(5-((1S,2S)-1-氨基-2-羟丙基)-1,3,4-噁二唑-2-基)-3-(氰基甲基)环丁基)氨基甲酰基)-L-丝氨酸的制备Example 4: (((1s,3R)-1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)-) Preparation of 3-(cyanomethyl)cyclobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000065
Figure PCTCN2018084352-appb-000065
步骤1:1-((叔丁氧羰基)氨基)-3-(氰基亚甲基)环丁烷甲酸乙酯的制备Preparation of ethyl 1-((tert-butoxycarbonyl)amino)-3-(cyanomethylene)cyclobutanecarboxylate
Figure PCTCN2018084352-appb-000066
Figure PCTCN2018084352-appb-000066
制备方法类似与实施例1中的步骤3的方法制得标题化合物,不同的是将原料(三苯基膦)乙酸乙酯替换为(三苯基膦)乙腈。The title compound was obtained in a similar manner to the method of the step 3 in Example 1, except that the starting material (triphenylphosphine) ethyl acetate was replaced with (triphenylphosphine) acetonitrile.
步骤2:(1s,3s)-1-((叔丁氧基羰基)氨基)-3-(氰基甲基)环丁烷甲酸乙酯的制备Step 2: Preparation of (1s,3s)-1-((tert-butoxycarbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylate
Figure PCTCN2018084352-appb-000067
Figure PCTCN2018084352-appb-000067
制备方法实施例1中的步骤6的方法,不同的是将原料苄基1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-乙氧基-2-氧代亚乙基)环丁烷甲酸酯替换为上述步骤1所得物1-((叔丁氧羰基)氨基)-3-(氰基亚甲基)环丁烷甲酸乙酯,后处理时通过柱层析纯化制得标题化合物及其立体异构体。Preparation Method The method of the step 6 in Example 1, except that the starting material benzyl 1-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-(2-ethoxyl) -2-oxoethylidene)cyclobutanecarboxylate is replaced by ethyl 1-((tert-butoxycarbonyl)amino)-3-(cyanomethylene)cyclobutanecarboxylate obtained in the above step 1, The title compound and its stereoisomer were obtained by column chromatography after workup.
步骤3:(1s,3s)-1-((叔丁氧羰基)氨基)-3-(氰基甲基)环丁烷甲酸的制备Step 3: Preparation of (1s,3s)-1-((tert-butoxycarbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylic acid
Figure PCTCN2018084352-appb-000068
Figure PCTCN2018084352-appb-000068
按实施例3中的步骤3的方法制得标题化合物,不同的是将原料乙基3-(2-氨基-2-氧代乙基)-1-((叔丁氧羰基)氨基)环丁烷甲酸乙酯替换为上述步骤2所得物(1s,3s)-1-((叔丁氧基羰基)氨基)-3-(氰基甲基)环丁烷甲酸乙酯。The title compound was obtained according to the procedure of Step 3 in Example 3, except that the starting material ethyl 3-(2-amino-2-oxoethyl)-1-((tert-butoxycarbonyl)amino)cyclobutane The ethyl alkanoate was replaced with the ethyl ester of (1s, 3s)-1-((tert-butoxycarbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylate obtained in the above step 2.
步骤4:(1s,3s)-1-氰基-3-(氰基甲基)环丁胺盐酸盐的制备Step 4: Preparation of (1s,3s)-1-cyano-3-(cyanomethyl)cyclobutylamine hydrochloride
Figure PCTCN2018084352-appb-000069
Figure PCTCN2018084352-appb-000069
将(1s,3s)-1-((叔丁氧羰基)氨基)-3-(氰基甲基)环丁烷甲酸(1.4g,5.5mmol,1.0eq)溶于8mL1,4-二氧六环中,加入HCl的1,4-二氧六环溶液14mL,反应过夜,过滤得白色固体1.04g,收率100%。(1s,3s)-1-((tert-Butoxycarbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylic acid (1.4 g, 5.5 mmol, 1.0 eq) was dissolved in 8 mL of 1,4-dioxane To the ring, 14 mL of a HCl solution of 1,4-dioxane was added, and the mixture was reacted overnight, and filtered to give a white solid, 1.04 g, yield 100%.
步骤5:(((1s,3R)-1-(5-((1S,2S)-1-氨基-2-羟丙基)-1,3,4-噁二唑-2-基)-3-(氰基甲基)环丁基)氨基甲酰基)-L-丝氨酸的制备Step 5: (((1s,3R)-1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)-3 Preparation of -(cyanomethyl)cyclobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000070
Figure PCTCN2018084352-appb-000070
按实施例1中的步骤5、7、8、9、11、13的方法制得标题化合物,不同的是将实施例1步骤5中的原料1-氨基-3-(2-乙氧基-2-氧代亚乙基)环丁烷甲酸苄酯替换为上述步骤4所得物(1s,3s)-1-氰基-3-(氰基甲基)环丁胺盐酸盐。 1H NMR(400MHz,D 2O):δ4.75(d,J=6.5Hz,1H),4.37(p,J=6.4Hz,1H),4.30(t,J=4.3Hz,1H),3.94(dd,J=11.7,4.8Hz,1H),3.85(dd,J=11.6,4.0Hz,1H),2.99(ddt,J=16.9,8.1,4.6Hz,2H),2.86(tt,J=13.6,6.9Hz,1H),2.77(d,J=6.4Hz,2H),2.46–2.35(m,2H),1.33(d,J=6.5Hz,3H).ESI-MS m/z:383.1[M+H] + The title compound was obtained according to the procedure of Steps 5, 7, 8, 9, 11, and 13 in Example 1, except that the starting material of the first step of Example 1 was 1-amino-3-(2-ethoxy- The benzyl 2-oxoethylidenecyclobutanecarboxylate was replaced with the product of the above step 4 (1s, 3s)-1-cyano-3-(cyanomethyl)cyclobutylamine hydrochloride. 1 H NMR (400 MHz, D 2 O): δ 4.75 (d, J = 6.5 Hz, 1H), 4.37 (p, J = 6.4 Hz, 1H), 4.30 (t, J = 4.3 Hz, 1H), 3.94 (dd, J=11.7, 4.8 Hz, 1H), 3.85 (dd, J=11.6, 4.0 Hz, 1H), 2.99 (ddt, J=16.9, 8.1, 4.6 Hz, 2H), 2.86 (tt, J=13.6) , 6.9 Hz, 1H), 2.77 (d, J = 6.4 Hz, 2H), 2.46 - 2.35 (m, 2H), 1.33 (d, J = 6.5 Hz, 3H). ESI-MS m/z: 383.1 [M +H] +
实施例5:(((1r,3S)-1-(5-((1S,2S)-1-氨基-2-羟丙基)-1,3,4-噁二唑-2-基)-3-(氰基甲基)环丁基)氨基甲酰基)-L-丝氨酸的制备Example 5: (((1r,3S)-1-(5-((1S,2S)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)- Preparation of 3-(cyanomethyl)cyclobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000071
Figure PCTCN2018084352-appb-000071
用实施例4步骤2制得的(1s,3s)-1-((叔丁氧基羰基)氨基)-3-(氰基甲基)环丁烷甲酸乙酯的另一种构象异构体为原料,参照实施例4中步骤3-5的方法制得标题化合物。 1H NMR(400MHz,D 2O):δ4.75(d,J=6.5Hz,1H),4.37(p,J=6.4Hz,1H),4.30(t,J=4.3Hz,1H),3.94(dd,J=11.7,4.8Hz,1H),3.85(dd,J=11.6,4.0Hz,1H),2.99(ddt,J=16.9,8.1,4.6Hz,2H),2.86(tt,J=13.6,6.9Hz,1H),2.77(d,J=6.4Hz,2H),2.46–2.35(m,2H),1.33(d,J=6.5Hz,3H).ESI-MS m/z:383.1[M+H] +. Another conformational isomer of ethyl (1s,3s)-1-((tert-butoxycarbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylate prepared in Step 2 of Example 4 For the starting materials, the title compound was obtained by the procedure of Step 3-5 in Example 4. 1 H NMR (400 MHz, D 2 O): δ 4.75 (d, J = 6.5 Hz, 1H), 4.37 (p, J = 6.4 Hz, 1H), 4.30 (t, J = 4.3 Hz, 1H), 3.94 (dd, J=11.7, 4.8 Hz, 1H), 3.85 (dd, J=11.6, 4.0 Hz, 1H), 2.99 (ddt, J=16.9, 8.1, 4.6 Hz, 2H), 2.86 (tt, J=13.6) , 6.9 Hz, 1H), 2.77 (d, J = 6.4 Hz, 2H), 2.46 - 2.35 (m, 2H), 1.33 (d, J = 6.5 Hz, 3H). ESI-MS m/z: 383.1 [M +H] + .
实施例6:((1-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-(羧甲基)环丁基)氨基甲酰基)-L-别苏氨酸的制备Example 6: ((1-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-(carboxymethyl) ring Preparation of butyl)carbamoyl)-L-pyrethine
Figure PCTCN2018084352-appb-000072
Figure PCTCN2018084352-appb-000072
步骤1:1-((苄氧基)羰基)-3-氧代环丁烷胺盐酸盐的制备Step 1: 1 - Preparation of ((benzyloxy)carbonyl)-3-oxocyclobutaneamine hydrochloride
Figure PCTCN2018084352-appb-000073
Figure PCTCN2018084352-appb-000073
按实施例4中的步骤4的方法制得标题化合物,不同的是将原料(1s,3s)-1-((叔丁氧羰基)氨基)-3-(氰基甲基)环丁烷甲酸替换为1-((叔丁氧基羰基)氨基)-3-氧代环丁烷甲酸苄酯。The title compound was obtained according to the procedure of Step 4 in Example 4, except that starting material (1s, 3s)-1-((tert-butoxycarbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylic acid Replaced with benzyl 1-((tert-butoxycarbonyl)amino)-3-oxocyclobutanecarboxylate.
步骤2:1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-氧代环丁烷甲酸苄酯的制备Step 2: Preparation of benzyl 1-((((9H-芴-9-yl)methoxy)carbonyl)amino)-3-oxocyclobutanecarboxylate
Figure PCTCN2018084352-appb-000074
Figure PCTCN2018084352-appb-000074
按实施例1中的步骤5的方法制得标题化合物,不同的是将实施例1步骤5中的原料1-氨基-3-(2-乙氧基-2-氧代亚乙基)环丁烷甲酸苄酯替换为上述步骤1所得物1-((苄氧基)羰基)-3-氧代环丁烷胺盐酸盐。The title compound was obtained according to the procedure of Step 5 in Example 1, except that the starting material of the starting material of Example 1 was 1-amino-3-(2-ethoxy-2-oxoethylidene)cyclobutane. The benzyl carbamate was replaced with the 1-((benzyloxy)carbonyl)-3-oxocyclobutaneamine hydrochloride obtained in the above step 1.
步骤3:苄基1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-(叔丁氧基)-2-氧代亚乙基)环丁烷甲酸酯的制备Step 3: Benzyl 1-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-(2-(tert-butoxy)-2-oxoethylidene)cyclobutane Preparation of alkacrylate
Figure PCTCN2018084352-appb-000075
Figure PCTCN2018084352-appb-000075
将1-(((((9H-芴-9-基)甲氧基)羰基)氨基)-3-氧代环丁烷甲酸苄酯(300mg,0.68mmol)溶于甲苯(5mL)中,加入(叔丁氧基羰基亚甲基)三苯基磷(282mg,0.74mmol),置于50℃油浴,反应24h,减压蒸馏,EA和水分液,有机相用食盐水洗涤,无水Na 2SO 4干燥。减压蒸馏,硅胶柱分离纯化(0-10%EAin PE)得无色油状物263mg,收率71.8%。 Benzyl 1-(((((9H-芴-9-yl)methoxy)carbonyl)amino)-3-oxocyclobutanecarboxylate (300 mg, 0.68 mmol) was dissolved in toluene (5 mL) (tert-Butoxycarbonylmethylene)triphenylphosphorate (282 mg, 0.74 mmol), placed in an oil bath at 50 ° C, reacted for 24 h, distilled under reduced pressure, EA and aqueous solution, washed with brine, anhydrous Na 2 SO 4 was dried, distilled under reduced pressure, and purified by silica gel column (0-10% EAIN PE) to afford 263 mg of colorless oil.
步骤4:1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-(叔丁氧基)-2-氧代乙基)环丁烷甲酸的制备Step 4: 1-(((9H-Indol-9-yl)methoxy)carbonyl)amino)-3-(2-(tert-butoxy)-2-oxoethyl)cyclobutanecarboxylic acid preparation
Figure PCTCN2018084352-appb-000076
Figure PCTCN2018084352-appb-000076
按实施例1中的步骤6的方法制得标题化合物,不同的是将原料苄基1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-乙氧基-2-氧代亚乙基)环丁烷甲酸酯替换为上述步骤3所得物苄基1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-(叔丁氧基)-2-氧代亚乙基)环丁烷甲酸酯。The title compound was obtained according to the procedure of Step 6 in Example 1, except that the starting material benzyl 1-((((9H- -9) yl)) oxy)) Ethoxy-2-oxoethylidene)cyclobutanecarboxylate is replaced by the benzyl 1-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)- 3-(2-(tert-Butoxy)-2-oxoethylidene)cyclobutanecarboxylate.
步骤5:(R,Z)-2-(3-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(4-氨基-5-(叔丁氧基甲基)9-二甲基-7-氧代-2,8-二氧杂-3,6-二氮杂环戊烯-3-烯-1-基)环丁基)乙酸叔丁酯的制备Step 5: (R,Z)-2-(3-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(4-amino-5-(tert-butoxymethyl) Of tert-butyl 9-dimethyl-7-oxo-2,8-dioxa-3,6-diazacyclo-3-ene-1-yl)cyclobutyl)acetate
Figure PCTCN2018084352-appb-000077
Figure PCTCN2018084352-appb-000077
将1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-(叔丁氧基)-2-氧代乙基)环丁烷甲酸(1.51g,3.34 mmol)溶于DMF(20mL)中,置于冰浴,依次加入原料(2)(1.19g,4.35mmol),HOBT(540mg,4mmol),EDC(768mg,4.0mmol),室温反应过夜,将反应液缓慢倒入冰水中,固体析出、过滤、水洗,将粗品溶于乙酸乙酯,分液,有机相用无水Na 2SO 4干燥。减压蒸馏,得白色固体1.6g,收率67.5% 1-((((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(2-(tert-butoxy)-2-oxoethyl)cyclobutanecarboxylic acid (1.51 g) , 3.34 mmol), dissolved in DMF (20 mL), EtOAc (EtOAc m. the reaction mixture was slowly poured into ice water, the precipitated solid was filtered, washed with water, the crude product was dissolved in ethyl acetate, separated, the organic phase was dried over anhydrous Na 2 SO 4. Distillation under reduced pressure gave a white solid 1.6 g, yield 67.5%
步骤6:(R)-2-(3-(((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(3-(2-(叔丁氧基)-1-((叔丁氧羰基)氨基)乙基)-1,2,4-噁二唑-5-基)环丁基)乙酸叔丁酯的制备Step 6: (R)-2-(3-((((9H-芴-9-yl))methoxy)carbonyl)amino)-3-(3-(2-(tert-butoxy)-1) Preparation of tert-butyl (-(tert-butoxycarbonyl)amino)ethyl)-1,2,4-oxadiazol-5-yl)cyclobutyl)acetate
Figure PCTCN2018084352-appb-000078
Figure PCTCN2018084352-appb-000078
将(R,Z)-2-(3-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(4-氨基-5-(叔丁氧基甲基)9-二甲基-7-氧代-2,8-二氧杂-3,6-二氮杂环戊烯-3-烯-1-基)环丁基)乙酸叔丁酯(1.6g,2.27mmol)溶于乙醇(40mL)、水(4mL)的混合溶剂中,加入醋酸钠(280mg,3.4mmol),加热回流反应10h。减压蒸馏,乙酸乙酯、水萃取,饱和食盐水洗涤,有机层用无水硫酸钠干燥,减压蒸馏,硅胶柱分离纯化(P/E=4/1)得无色油状物802mg,收率52%。(R,Z)-2-(3-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(4-amino-5-(tert-butoxymethyl)) Tert-butyl 9-dimethyl-7-oxo-2,8-dioxa-3,6-diazacyclo-3-ene-1-yl)cyclobutyl)acetate (1.6 g, 2.27 mmol) was dissolved in a mixed solvent of ethanol (40 mL) and water (4 mL), and sodium acetate (280 mg, 3.4 mmol) was added, and the mixture was heated to reflux for 10 h. Distilled under reduced pressure, ethyl acetate and water were evaporated, washed with saturated brine, and dried over anhydrous sodium sulfate. The rate is 52%.
步骤7:((1-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-(羧甲基)环丁基)氨基甲酰基)-L-别苏氨酸的制备Step 7: ((1-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-(carboxymethyl)cyclobutane Preparation of carbamoyl)-L-pyrethine
Figure PCTCN2018084352-appb-000079
Figure PCTCN2018084352-appb-000079
按实施例1中的步骤9、11、13的方法制得标题化合物,不同的是将原料乙基2-(3-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧基羰基)氨基)丙基)-1,3,4-噁二唑-2-基)环丁基)乙酸酯替换为上述步骤6所得物(R)-2-(3-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(3-(2-(叔丁氧基)-1-((叔丁氧羰基)氨基)乙基)-1,2,4-噁二唑-5-基)环丁基)乙酸叔丁酯的制备。 1H NMR(400MHz,D 2O):δ4.73(dd,J=8.5,4.3Hz,1H),4.38–4.29(m,1H),4.17(dd,J=10.0,2.3Hz,1H),4.15–4.01(m,2H),2.95(dt,J=16.5,8.4Hz,1H),2.81(dt,J=15.8,8.0Hz,0.5H),2.69–2.47(m,5H),2.35–2.17(m,0.5H),1.17(t,J=6.9Hz,3H).ESI-MS m/z:402.2[M+H] + The title compound was obtained according to the procedure of Steps 9, 11, 13 in Example 1, except that the starting material ethyl 2-(3-(((((((()))))))) -3(5-((1S,2S)-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazole-2 -()-cyclobutyl)acetate is replaced by the above-mentioned step 6 (R)-2-(3-(((9H-芴-9-yl))methoxy)carbonyl)amino)-3-( Tert-butyl 3-(2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)ethyl)-1,2,4-oxadiazol-5-yl)cyclobutyl)acetate preparation. 1 H NMR (400 MHz, D 2 O): δ 4.73 (dd, J = 8.5, 4.3 Hz, 1H), 4.38 - 4.29 (m, 1H), 4.17 (dd, J = 10.0, 2.3 Hz, 1H), 4.15–4.01 (m, 2H), 2.95 (dt, J = 16.5, 8.4 Hz, 1H), 2.81 (dt, J = 15.8, 8.0 Hz, 0.5H), 2.69–2.47 (m, 5H), 2.35–2.17 (m, 0.5H), 1.17 (t, J = 6.9 Hz, 3H). ESI-MS m/z: 402.2 [M+H] +
实施例7:((1-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-(2-氨基-2-氧代乙基)环丁基)氨基甲酰基)-L-别苏氨酸的制备Example 7: ((1-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-(2-amino-2) -Oxoethyl)cyclobutyl)carbamoyl)-L-pyrethine
Figure PCTCN2018084352-appb-000080
Figure PCTCN2018084352-appb-000080
步骤1:(R)-(1-(5-(1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(3-(2-氨基-2-氧乙基)环丁基)-1,2,4-噁二唑-3-基)-2-(叔丁氧基)乙基)氨基甲酸叔丁酯的制备Step 1: (R)-(1-(5-(1-(((9H-芴-9-yl))methoxy)carbonyl)amino)-3-(3-(2-amino-2-oxo) Preparation of tert-butyl ester of ethyl)cyclobutyl)-1,2,4-oxadiazol-3-yl)-2-(tert-butoxy)ethyl)carbamate
Figure PCTCN2018084352-appb-000081
Figure PCTCN2018084352-appb-000081
按实施例6中的步骤5、6的方法制得标题化合物,不同的是将原料1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-(叔丁氧基)-2-氧代乙基)环丁烷甲酸替换为3-(2-氨基-2-氧代乙基)-1-((叔丁氧基羰基)氨基)环丁烷甲酸。The title compound was obtained according to the procedure of Steps 5 and 6 in Example 6 except that the starting material was 1-((((9H-in-9-yl)methoxy)carbonyl)amino)-3-(2-) Replacement of (tert-butoxy)-2-oxoethyl)cyclobutanecarboxylic acid with 3-(2-amino-2-oxoethyl)-1-((tert-butoxycarbonyl)amino)cyclobutane Formic acid.
步骤2:((1-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-(2-氨基-2-氧代乙基)环丁基)氨基甲酰基)-L-别苏氨酸的制备Step 2: ((1-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-(2-amino-2-) Preparation of oxoethyl)cyclobutyl)carbamoyl)-L-pyrethine
Figure PCTCN2018084352-appb-000082
Figure PCTCN2018084352-appb-000082
按实施例1中的步骤9、11、13的方法制得标题化合物。不同的是将原料2-(3-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧基羰基)氨基)丙基)-1,3,4-噁二唑-2-基)环丁基)乙酸乙酯替换为(R)-(1-(5-(1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(3-(2-氨基-2氧乙基)环丁基)-1,2,4-噁二唑-3-基)-2-(叔丁氧基)乙基)氨基甲酸叔丁酯。 1H NMR(400MHz,D 2O):δ4.41–4.32(m,1H),4.22(dt,J=9.5,3.3Hz,1H),4.18–4.03(m,2H),3.06–2.78(m,2H),2.68–2.49(m,4H),2.29(ddd,J=12.0,8.5,3.4Hz,1H),1.21(t,J=6.6Hz,3H).ESI-MS m/z:401.2[M+H] + The title compound was obtained by the procedure of Steps 9, 11, and 13 in Example 1. The difference is that the starting material is 2-(3-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(5-((1S,2S)-2-(tert-butoxy) -1 -((tert-Butoxycarbonyl)amino)propyl)-1,3,4-oxadiazol-2-yl)cyclobutyl)acetate was replaced by (R)-(1-(5) -(1-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(3-(2-amino-2-oxoethyl)cyclobutyl)-1,2,4 tert-Butyl oxazol-3-yl)-2-(tert-butoxy)ethyl)carbamate 1 H NMR (400 MHz, D 2 O): δ 4.41 - 4.32 (m, 1H), 4.22 (dt, J=9.5, 3.3 Hz, 1H), 4.18–4.03 (m, 2H), 3.06–2.78 (m, 2H), 2.68–2.49 (m, 4H), 2.29 (ddd, J=12.0, 8.5, 3.4 Hz, 1H), 1.21 (t, J = 6.6 Hz, 3H). ESI-MS m/z: 401.2 [M+H] +
实施例8:(((1r,3S)-1-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-(氰基甲基)环丁基)氨基甲酰基)-L-别苏氨酸的制备Example 8: (((1r,3S)-1-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3- Preparation of (cyanomethyl)cyclobutyl)carbamoyl)-L-pyrethine
Figure PCTCN2018084352-appb-000083
Figure PCTCN2018084352-appb-000083
步骤1:(9H-芴-9-基)甲基-((1r,3r)-3-(氰基甲基)-1-(氟代羰基)环丁基)氨基甲酸酯的制备Step 1: Preparation of (9H-fluoren-9-yl)methyl-((1r,3r)-3-(cyanomethyl)-1-(fluorocarbonyl)cyclobutyl)carbamate
Figure PCTCN2018084352-appb-000084
Figure PCTCN2018084352-appb-000084
将1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(氰基甲基)环丁烷甲酸(200mg,0.53mmol)溶于DCM(5mL)中,加入DAST(111mg,0.69mmol),室温反应1h,,加水淬灭反应,二氯甲烷、水萃取,有机层用饱和食盐水洗涤,Na 2SO 4干燥。减压蒸馏,将所得物直接投入下一步反应。 1-((((9H-Indol-9-yl)methoxy)carbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylic acid (200 mg, 0.53 mmol) was added DAST (111mg, 0.69mmol), stirred at rt for 1h ,, the reaction was quenched with water, dichloromethane and extracted with water, the organic layer was washed with saturated brine, dried Na 2 SO 4. The mixture was distilled under reduced pressure, and the resultant was directly poured into the next reaction.
步骤2:((R)-1-(5-((1r,3R)-1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(氰基甲基)环丁基)-1,2,4-噁二唑-3-基)2-(叔丁氧基)乙基)氨基甲酸叔丁酯的制备Step 2: ((R)-1-(5-((1r,3R)-1-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(cyanomethyl) Of tert-butyl)-1,2,4-oxadiazol-3-yl)2-(tert-butoxy)ethyl)carbamic acid tert-butyl ester
Figure PCTCN2018084352-appb-000085
Figure PCTCN2018084352-appb-000085
按实施例6中的步骤5、6的方法制得标题化合物,不同的是将原料1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(2-(叔丁氧基)-2-氧代乙基)环丁烷甲酸替换为(9H-芴-9-基)甲基(3-(氰基甲 基)-1-(氟代羰基)环丁基)氨基甲酸酯。The title compound was obtained according to the procedure of Steps 5 and 6 in Example 6 except that the starting material was 1-((((9H-in-9-yl)methoxy)carbonyl)amino)-3-(2-) Replacement of (tert-butoxy)-2-oxoethyl)cyclobutanecarboxylic acid with (9H-fluoren-9-yl)methyl(3-(cyanomethyl)-1-(fluorocarbonyl)cyclobutane Carbamate.
步骤3:(((1R,3S)-1-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-(氰基甲基)环丁基)氨基甲酰基)-L-别苏氨酸的制备Step 3: (((1R,3S)-1-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-( Preparation of cyanomethyl)cyclobutyl)carbamoyl)-L-pyrethine
Figure PCTCN2018084352-appb-000086
Figure PCTCN2018084352-appb-000086
按实施例1中的步骤9、11、13的方法制得标题化合物。不同的是将原料2-(3-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(5-((1S,2S)-2-(叔丁氧基)-1-((叔丁氧基羰基)氨基)丙基)-1,3,4-噁二唑-2-基)环丁基)乙酸乙酯替换为((R)-1-(5-((1r,3R)-1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(氰基甲基)环丁基)-1,2,4-噁二唑-3-基)2-(叔丁氧基)乙基)氨基甲酸叔丁酯。 1H NMR(400MHz,D 2O):δ4.65–4.56(m,1H),4.21(qd,J=6.4,2.7Hz,1H),4.12–4.03(m,1H),3.99–3.83(m,2H),2.93–2.73(m,1H),2.58(t,J=8.6Hz,2H),2.50(d,J=8.7Hz,3H),1.05(d,J=6.4Hz,3H).ESI-MS m/z:383.2[M+H] + The title compound was obtained by the procedure of Steps 9, 11, and 13 in Example 1. The difference is that the starting material is 2-(3-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(5-((1S,2S)-2-(tert-butoxy) ) ethyl-1-((tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazol-2-yl)cyclobutyl)acetate was replaced by ((R)-1-(5) -((1r,3R)-1-(((9H-芴-9-yl)methoxy)carbonyl)amino)-3-(cyanomethyl)cyclobutyl)-1,2,4- Tert-butyl ester of oxadiazol-3-yl)2-(tert-butoxy)ethyl)carbamate. 1 H NMR (400 MHz, D 2 O): δ 4.65 - 4.56 (m, 1H), 4.21. (qd, J = 6.4, 2.7 Hz, 1H), 4.12 - 4.03 (m, 1H), 3.99 - 3.83 (m) , 2H), 2.93 - 2.73 (m, 1H), 2.58 (t, J = 8.6 Hz, 2H), 2.50 (d, J = 8.7 Hz, 3H), 1.05 (d, J = 6.4 Hz, 3H). ESI -MS m/z: 383.2 [M+H] +
实施例9:(((1s,3R)-1-(5-((S)-1-氨基-2-羟乙基)-1,3,4-噁二唑-2-基)-3-(氰基甲基)环丁基)氨基甲酰基)-L-别苏氨酸的制备Example 9: (((1s,3R)-1-(5-((S)-1-Amino-2-hydroxyethyl)-1,3,4-oxadiazol-2-yl)-3- Preparation of (cyanomethyl)cyclobutyl)carbamoyl)-L-pyrethine
Figure PCTCN2018084352-appb-000087
Figure PCTCN2018084352-appb-000087
以(S)-(3-(叔丁氧基)-1-肼基-1-氧代丙-2-基)氨基甲酸叔丁酯和1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(氰基甲基)环丁烷甲酸为原料,按实施例1中的步骤7、8、9、11、13的方法制得标题化合物。Tert-butyl (S)-(3-(tert-butoxy)-1-indol-1-ylpropan-2-yl)carbamate and 1-((((9H-芴-9-yl))) The title compound was obtained by the procedure of Steps 7, 8, 9, 11, and 13 of Example 1, using methoxy)carbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylic acid as the starting material.
实施例10:(((1r,3S)-1-(5-((S)-1-氨基-2-羟乙基)-1,3,4-噁二唑-2-基)-3-(氰基甲基)环丁基)氨基甲酰基)-L-别苏氨酸的制备Example 10: (((1r,3S)-1-(5-((S)-1-Amino-2-hydroxyethyl)-1,3,4-oxadiazol-2-yl)-3- Preparation of (cyanomethyl)cyclobutyl)carbamoyl)-L-pyrethine
Figure PCTCN2018084352-appb-000088
Figure PCTCN2018084352-appb-000088
以1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-(氰基甲基)环丁烷甲酸的另一种构象异构体为原料,按实施例1中的步骤7、8、9、11、13的方法制得标题化合物。Taking another conformational isomer of 1-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-(cyanomethyl)cyclobutanecarboxylic acid as a raw material, according to the example The title compound was obtained by the method of Steps 7, 8, 9, 11, and 13 in 1.
实施例11 (((S)-4-氨基-1-(5-((2S,4R)-4-羟基四氢吡咯-2-基)-1,3,4-噁二唑-2-基)-4-氧代丁基)氨基甲酰基)-L-丝氨酸Example 11 (((S)-4-Amino-1-(5-((2S,4R)-4-hydroxytetrahydropyrrole-2-yl)-1,3,4-oxadiazol-2-yl) )-4-oxobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000089
Figure PCTCN2018084352-appb-000089
步骤1 (2S,4R)-N-(叔丁氧羰基)-4-((叔丁基二甲基硅基)氧基)-2-吡咯甲酸甲酯的制备Step 1 Preparation of (2S,4R)-N-(tert-Butoxycarbonyl)-4-((tert-butyldimethylsilyl)oxy)-2-pyrrolecarboxylic acid methyl ester
Figure PCTCN2018084352-appb-000090
Figure PCTCN2018084352-appb-000090
将(2S,4R)-N-(叔丁氧羰基)-4-羟基-2-吡咯甲酸甲酯(5g,20.4mmol,1eq)和咪唑(1.7g,24.5mmol,1.2eq)溶于N,N-二甲基甲酰胺(70mL)中,于0℃下向其溶液中加入叔丁基二甲基氯硅烷(3.7g,24.5mmol,1.2eq)室温下搅拌约24h。待反应完全后,向混合溶液中加入饱和氯化铵溶液和乙酸乙酯,有机相依次用饱和氯化铵溶液,水,饱和氯化钠溶液洗涤,无水硫酸钠干燥,抽滤,浓缩,得透明油状产物(6.0g),产率为81.2%。Methyl (2S,4R)-N-(tert-butoxycarbonyl)-4-hydroxy-2-pyrrolecarboxylate (5 g, 20.4 mmol, 1 eq) and imidazole (1.7 g, 24.5 mmol, 1.2 eq) were dissolved in N. To N-dimethylformamide (70 mL), tert-butyldimethylsilyl chloride (3.7 g, 24.5 mmol, 1.2 eq) was added to the solution at 0 ° C and stirred at room temperature for about 24 h. After the reaction is completed, a saturated ammonium chloride solution and ethyl acetate are added to the mixed solution, and the organic phase is washed successively with a saturated aqueous solution of ammonium chloride, water and a saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated. The product was obtained as a clear oil (6.0 g).
步骤2 (2S,4R)-4-((叔丁基二甲基硅基)氧基)-2-(肼基羰基)四氢吡咯-1-甲酸叔丁酯的制备Step 2 Preparation of (2S,4R)-4-((tert-butyldimethylsilyl)oxy)-2-(indolylcarbonyl)tetrahydropyrrole-1-carboxylic acid tert-butyl ester
Figure PCTCN2018084352-appb-000091
Figure PCTCN2018084352-appb-000091
将步骤1制得的(2S,4R)-N-(叔丁氧羰基)-4-((叔丁基二甲基硅基)氧基)-2-吡咯甲酸甲酯(6g,16.7mmol)溶于甲醇(36mL),向其溶液中加入水合肼(15mL),室温下搅拌24h。待反应完全后,将甲醇旋干,向混合物中加入水和乙酸乙酯,有机相用水洗,饱和氯化钠溶液洗涤,无水硫酸钠干燥,抽滤,浓缩,得纯白色固体(5.5g),产率为91.7%。Methyl (2S,4R)-N-(tert-butoxycarbonyl)-4-((tert-butyldimethylsilyl)oxy)-2-pyrrolecarboxylate (6 g, 16.7 mmol) obtained in Step 1 Dissolved in methanol (36 mL), and added hydrazine hydrate (15 mL). After the reaction is completed, the methanol is evaporated to dryness. Water and ethyl acetate are added to the mixture. The organic phase is washed with water, washed with saturated sodium chloride, dried over anhydrous sodium sulfate, filtered, The yield was 91.7%.
步骤3 (2S,4R)-2-(2-(N 2-(((9H-芴-9-基)甲氧基)羰基)-N 5-三苯甲基-L-谷氨酰胺酰基)肼基-1-羰基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸叔丁酯的制备 Step 3 (2S,4R)-2-(2-(N 2 -((9H-芴-9-yl)methoxy)carbonyl)-N 5 -trityl-L-glutaminyl) Preparation of tert-butyl ester of mercapto-1-carbonyl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole-1-carboxylate
Figure PCTCN2018084352-appb-000092
Figure PCTCN2018084352-appb-000092
将(2S,4R)-4-((叔丁基二甲基硅基)氧基)-2-(肼基羰基)四氢吡咯-1-甲酸叔丁酯(5.5g,15.3mmol,1eq),N 2-(((9H-芴-9-基)甲氧基)羰基)-N 5-三苯甲基-L-谷氨酰胺(9.4g,15.3mmol,1eq),1-乙基-(3-二甲基氨基丙基)碳化二亚胺盐酸盐(3.5g,18.4mmol,1.2eq),1-羟基苯并三氮唑(2.5g,18.4mmol,1.2eq)溶于N,N-二甲基甲酰胺(50mL)中,于0℃下缓慢滴入N-甲基吗啉(3.9g,38.2mmol,2.5eq),滴完后室温下搅拌反应12h。待反应完全后,将混合溶液投入冰水中搅拌,固体析出,抽滤,用二氯甲烷将固体溶解,分去水相,有机相用无水硫酸钠干燥,旋干,向其中加入石油醚洗涤,抽滤,固体干燥,得纯白色固体(14.1g),产率为96.8%。 (2S,4R)-4-((tert-Butyldimethylsilyl)oxy)-2-(indolylcarbonyl)tetrahydropyrrole-1-carboxylic acid tert-butyl ester (5.5 g, 15.3 mmol, 1 eq) , N 2 -(((9H-芴-9-yl)methoxy)carbonyl)-N 5 -trityl-L-glutamine (9.4 g, 15.3 mmol, 1 eq), 1-ethyl- (3-Dimethylaminopropyl)carbodiimide hydrochloride (3.5 g, 18.4 mmol, 1.2 eq), 1-hydroxybenzotriazole (2.5 g, 18.4 mmol, 1.2 eq) dissolved in N, N-methylmorpholine (3.9 g, 38.2 mmol, 2.5 eq) was slowly added dropwise to N-dimethylformamide (50 mL) at 0 ° C, and the mixture was stirred at room temperature for 12 h. After the reaction is completed, the mixed solution is poured into ice water, stirred, and the solid is precipitated, suction filtered, and the solid is dissolved with dichloromethane, the aqueous phase is separated, the organic phase is dried over anhydrous sodium sulfate, dried, and petroleum ether is added thereto. Filtration and solid drying gave a white solid (14.1 g).
步骤4 (2S,4R)-2-(5-((S)-1-((((9H-芴-9-基)甲氧基)羰基)氨基)-4-氧代-4-(三苯甲基氨基)丁基)-1,3,4-噁二唑-2-基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸叔丁酯的制备Step 4 (2S,4R)-2-(5-((S)-1-(((9H-芴-9-yl)methoxy)carbonyl)amino)-4-oxo-4-(3) Benzylamino)butyl)-1,3,4-oxadiazol-2-yl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole-1-carboxylic acid tert-butyl ester Preparation
Figure PCTCN2018084352-appb-000093
Figure PCTCN2018084352-appb-000093
将(2S,4R)-2-(2-(N 2-(((9H-芴-9-基)甲氧基)羰基)-N 5-三苯甲基-L-谷氨酰胺酰基)肼基-1-羰 基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸叔丁酯(14.1g,14.8mmol,1eq)溶于四氢呋喃(100mL)和N,N-二甲基甲酰胺(40mL)中,于0℃下加入三苯基膦(7.9g,30.1mmol,2eq)和碘(7.6g,30.1mmol,2eq)。当碘完全溶解后,加入三乙胺(5.9g,58.6mmol,4eq),加完后于室温下搅拌5h。待反应完全后,向混合溶液中加入水和乙酸乙酯,有机相用硫代硫酸钠溶液和饱和氯化钠溶液洗涤,无水硫酸钠干燥,抽滤,浓缩制砂,柱层析纯化(乙酸乙酯:石油醚=1:3)。得白色固体(4.8g),产率为34.7%。 (2S,4R)-2-(2-(N 2 -((9H-芴-9-yl)methoxy)carbonyl)-N 5 -trityl-L-glutaminyl) tert-Butyl benzyl-1-carbonyl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole-1-carboxylate (14.1 g, 14.8 mmol, 1 eq) dissolved in tetrahydrofuran (100 mL) and N Triphenylphosphine (7.9 g, 30.1 mmol, 2 eq) and iodine (7.6 g, 30.1 mmol, 2 eq) were added at 0 ° C in N-dimethylformamide (40 mL). After the iodine was completely dissolved, triethylamine (5.9 g, 58.6 mmol, 4 eq) was added, and the mixture was stirred at room temperature for 5 h. After the reaction is completed, water and ethyl acetate are added to the mixed solution, and the organic phase is washed with a sodium thiosulfate solution and a saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography. Ethyl acetate: petroleum ether = 1:3). A white solid (4.8 g) was obtained in a yield of 34.7%.
步骤5 (2S,4R)-2-(5-((S)-1-氨基-4-氧代-4-(三苯甲基氨基)丁基)-1,3,4-噁二唑-2-基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸叔丁酯的制备Step 5 (2S,4R)-2-(5-((S)-1-Amino-4-oxo-4-(tritylamino)butyl)-1,3,4-oxadiazole- Preparation of 2-butyryl-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole-1-carboxylic acid tert-butyl ester
Figure PCTCN2018084352-appb-000094
Figure PCTCN2018084352-appb-000094
将(2S,4R)-2-(5-((S)-1-((((9H-芴-9-基)甲氧基)羰基)氨基)-4-氧代-4-(三苯甲基氨基)丁基)-1,3,4-噁二唑-2-基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸叔丁酯(3g,3.2mmol)溶于二氯甲烷(15mL),加入二乙胺(15mL),室温下搅拌4h。待反应完全后,浓缩制砂,柱层析纯化(乙酸乙酯:石油醚=4:1)。得白色固体(1.7g),产率为74.2%。(2S,4R)-2-(5-((S)-1-(((9H-芴-9-yl)methoxy)carbonyl)amino)-4-oxo-4-(triphenyl) Methylamino)butyl)-1,3,4-oxadiazol-2-yl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole-1-carboxylic acid tert-butyl ester ( 3 g, 3.2 mmol) was dissolved in dichloromethane (15 mL). After the reaction was completed, the mixture was concentrated and purified by column chromatography (ethyl acetate: petroleum ether = 4:1). A white solid (1.7 g) was obtained in a yield of 74.2%.
步骤6 O-(叔丁基)-N-((4-硝基苯氧基)羰基)-L-丝氨酸叔丁酯的制备Step 6 Preparation of O-(tert-butyl)-N-((4-nitrophenoxy)carbonyl)-L-serine tert-butyl ester
Figure PCTCN2018084352-appb-000095
Figure PCTCN2018084352-appb-000095
将O-(叔丁基)-L丝氨酸叔丁酯(5g,23.01mmol,1eq)溶于二氯甲烷(25mL)中,加入三乙胺(4.7g,46.02mmol,2eq),于0℃下向其中缓慢滴加4-硝基苯基氯甲酸酯(5.1g,25.31mmol,1.1eq)溶于二氯甲烷(25mL)溶液,滴完后,室温反应3h。待反应完全后,加入柠檬酸和二氯甲烷,有机相用柠檬酸,饱和碳酸钠溶液,水依次洗涤,干燥,浓缩,加入石油醚洗涤,抽滤,滤液旋干得白色固体(2.2g),产率为25.0%。O-(tert-butyl)-L-serine tert-butyl ester (5 g, 23.01 mmol, 1 eq) was dissolved in dichloromethane (25 mL) and triethylamine (4.7 g, 46.02 mmol, 2 eq) 4-Nitrophenyl chloroformate (5.1 g, 25.31 mmol, 1.1 eq) was slowly added dropwise to a solution of dichloromethane (25 mL). After the reaction is completed, citric acid and dichloromethane are added, the organic phase is washed with citric acid, saturated sodium carbonate solution, water, dried, concentrated, washed with petroleum ether, suction filtered, and the filtrate is dried to give a white solid (2.2 g) The yield was 25.0%.
步骤7 (2S,4R)-2-(5-((6S,10S)-10-(叔丁氧甲基)-13,13-二甲基-3,8,11-三氧代-1,1,1-三苯基-12-氧基-2,7,9-氮杂十四烷-6-基)-1,3,4-噁二唑-2-基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸叔丁酯的制备Step 7 (2S,4R)-2-(5-((6S,10S)-10-(tert-butoxymethyl)-13,13-dimethyl-3,8,11-trioxo-1, 1,1-triphenyl-12-oxy-2,7,9-azatetradecane-6-yl)-1,3,4-oxadiazol-2-yl)-4-((uncle Preparation of tert-butyl butyl dimethylsilyl)oxy)tetrahydropyrrole-1-carboxylate
Figure PCTCN2018084352-appb-000096
Figure PCTCN2018084352-appb-000096
将(2S,4R)-2-(5-((S)-1-氨基-4-氧代-4-(三苯甲基氨基)丁基)-1,3,4-噁二唑-2-基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸叔丁酯(0.7g,0.98mmol,1eq)和三乙胺(0.2g,1.97mmol,2eq)溶于N,N-二甲基甲酰胺(15mL),于0℃下逐滴加入O-(叔丁基)-N-((4-硝基苯氧基)羰基)-L-丝氨酸叔丁酯(0.41g,1.08mmol,1.1eq),滴完后于室温下搅拌6h。待反应完全后,加入二氯甲烷 和水,有机相用水,饱和氯化钠溶液洗涤,无水硫酸钠干燥,抽滤,浓缩制砂,柱层析纯化(乙酸乙酯:石油醚=2:3)得白色固体(0.8g),产率为85.2%。(2S,4R)-2-(5-((S)-1-Amino-4-oxo-4-(tritylamino)butyl)-1,3,4-oxadiazole-2 -yl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole-1-carboxylic acid tert-butyl ester (0.7 g, 0.98 mmol, 1 eq) and triethylamine (0.2 g, 1.97 mmol, 2 eq) was dissolved in N,N-dimethylformamide (15 mL), and O-(tert-butyl)-N-((4-nitrophenoxy)carbonyl)-L-serine was added dropwise at 0 °C. tert-Butyl ester (0.41 g, 1.08 mmol, 1.1 eq) was stirred at room temperature for 6 h. After the reaction is completed, dichloromethane and water are added, and the organic phase is washed with water, saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, filtered, and purified by column chromatography (ethyl acetate: petroleum ether = 2: 3) A white solid (0.8 g) was obtained in a yield of 85.2%.
步骤8 (((S)-4-氨基-1-(5-((2S,4R)-4-羟基四氢吡咯-2-基)-1,3,4-噁二唑-2-基)-4-氧代丁基)氨基甲酰基)-L-丝氨酸的制备Step 8 (((S)-4-Amino-1-(5-((2S,4R)-4-hydroxytetrahydropyrrole-2-yl)-1,3,4-oxadiazol-2-yl) Preparation of -4-oxobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000097
Figure PCTCN2018084352-appb-000097
将(2S,4R)-2-(5-((6S,10S)-10-(叔丁氧甲基)-13,13-二甲基-3,8,11-三氧代-1,1,1-三苯基-12-氧基-2,7,9-氮杂十四烷-6-基)-1,3,4-噁二唑-2-基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸叔丁酯(800mg,0.84mmol)溶于二氯甲烷(16mL),加入三氟乙酸(16mL)和三异丙基硅烷(0.8mL),室温下搅拌6h。待反应完全后,旋干,加入水和二氯甲烷,水相用二氯甲烷洗涤,冻干,进行制备分离,冻干得白色固体(190mg),产率为58.6%。 1H NMR(400MHz,D 2O):δ5.35-5.31(t,1H),5.11-5.08(t,1H),4.87-4.73(m,1H),4.19-4.17(t,1H),3.89-3.83(d,2H),3.70-3.66(t,1H),3.53-3.50(t,1H),2.69-2.60(m,2H),2.56-2.52(t,2H),2.44-2.37(m,1H),2.30-2.23(m,1H).ESI-MS m/z:387.0[M+H] +. (2S,4R)-2-(5-((6S,10S)-10-(tert-butoxymethyl)-13,13-dimethyl-3,8,11-trioxo-1,1 ,1-triphenyl-12-oxy-2,7,9-azatetradecane-6-yl)-1,3,4-oxadiazol-2-yl)-4-((tert-butyl) tert-Butyl dimethylsilyl)oxy)tetrahydropyrrole-1-carboxylate (800 mg, 0.84 mmol) was dissolved in dichloromethane (16 mL), trifluoroacetic acid (16 mL) and triisopropylsilane (0.8) (mL), stir at room temperature for 6 h. After the reaction was completed, the mixture was evaporated to dryness, water and methylene chloride, and the aqueous phase was washed with methylene chloride, and lyophilized to give a white solid (190 mg). 1 H NMR (400 MHz, D 2 O): δ 5.35-5.31 (t, 1H), 5.11-5.08 (t, 1H), 4.87-4.73 (m, 1H), 4.19 - 4.17 (t, 1H), 3.89 -3.83(d,2H), 3.70-3.66(t,1H),3.53-3.50(t,1H),2.69-2.60(m,2H),2.56-2.52(t,2H),2.44-2.37(m, 1H), 2.30-2.23 (m, 1H). ESI-MS m/z: 387.0 [M+H] + .
实施例12 (((S)-4-氨基-1-(5-((2S,3S)-3-羟基四氢吡咯-2-基)-1,3,4-噁二唑-2-基)-4-氧代丁基)氨基甲酰基)-L-丝氨酸的制备Example 12 (((S)-4-Amino-1-(5-((2S,3S)-3-hydroxytetrahydropyrrole-2-yl)-1,3,4-oxadiazol-2-yl) Preparation of 4-oxobutyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000098
Figure PCTCN2018084352-appb-000098
步骤1 (2S,3S)-3-羟基四氢吡咯-2-甲酸甲酯的制备Step 1 Preparation of (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid methyl ester
Figure PCTCN2018084352-appb-000099
Figure PCTCN2018084352-appb-000099
将(2S,3S)-3-羟基四氢吡咯-2-甲酸(4g,30.5mmol,1eq)溶于甲醇(40mL),于0℃下滴加氯化亚砜(5.44g,45.8mmol,1.5eq),滴完后,混合溶液于65-70℃下回流3h,待反应完全后,旋干,得透明油状产物(4.4g),产率为99.3%。(2S,3S)-3-Hydroxytetrahydropyrrole-2-carboxylic acid (4 g, 30.5 mmol, 1 eq) was dissolved in methanol (40 mL), and chlorosulfoxide (5.44 g, 45.8 mmol, 1.5) was added dropwise at 0 °C. Eq), after the completion of the dropwise addition, the mixed solution was refluxed at 65-70 ° C for 3 h, and after the reaction was completed, it was dried to give a product (yield: 4.4 g).
步骤2 (2S,3S)-N-(叔丁氧羰基)-3-羟基-2-吡咯甲酸甲酯的制备Step 2 Preparation of (2S,3S)-N-(tert-Butoxycarbonyl)-3-hydroxy-2-pyrrolecarboxylic acid methyl ester
Figure PCTCN2018084352-appb-000100
Figure PCTCN2018084352-appb-000100
将(2S,3S)-3-羟基四氢吡咯-2-甲酸甲酯(4.4g,30.3mmol,1eq)溶于四氢呋喃(40mL)溶液中, 缓慢向其中加入碳酸氢钠水溶液(8.9g,106.1mmol,3.5eq,溶于40mL水),于0℃下滴加二碳酸二叔丁酯(6.28g,28.8mmol,0.95eq),滴完后,室温下搅拌24h。向混合物中加入水和乙酸乙酯,有机相用水洗,饱和氯化钠溶液洗涤,无水硫酸钠干燥,抽滤,浓缩,得白色固体(7.4g),产率为99.6%。Methyl (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylate (4.4 g, 30.3 mmol, 1 eq) was dissolved in tetrahydrofuran (40 mL), and aqueous sodium hydrogencarbonate (8.9 g, 106.1 Methyl acetate (3.5 eq, dissolved in 40 mL of water) and di-tert-butyl dicarbonate (6.28 g, 28.8 mmol, 0.95 eq) was added dropwise at 0 ° C, and then stirred at room temperature for 24 h. Water and ethyl acetate were added to the mixture, and the mixture was evaporated.
步骤3(((S)-4-氨基-1-(5-((2S,3S)-3-羟基四氢吡咯-2-基)-1,3,4-噁二唑-2-基)-4-氧代丁基)氨基甲酰基)-L-丝氨酸制备Step 3 (((S)-4-Amino-1-(5-((2S,3S)-3-hydroxytetrahydropyrrole-2-yl)-1,3,4-oxadiazol-2-yl) Preparation of -4-oxobutyl)carbamoyl)-L-serine
制备方法同实施例11中步骤1~步骤8的制备方法,不同的是将实施例11中的原料(2S,4R)-N-(叔丁氧羰基)-4-羟基-2-吡咯甲酸甲酯替换为上述步骤2所得物(2S,3S)-N-(叔丁氧羰基)-3-羟基-2-吡咯甲酸甲酯。 1H NMR(400MHz,D 2O):δ5.12-5.10(t,1H),5.09-5.06(t,1H),4.95-4.94(m,1H),4.20-4.19(t,1H),3.87-3.86(d,2H),3.78-3.73(t,2H),2.58-2.53(t,2H),2.53-2.48(m,1H),2.47-2.37(m,1H),2.29-2.22(m,2H).ESI-MS m/z:387.0[M+H] +. The preparation method is the same as the preparation method of Step 1 to Step 8 in Example 11, except that the starting material (2S, 4R)-N-(tert-butoxycarbonyl)-4-hydroxy-2-pyrrolecarboxylic acid A in Example 11 is prepared. The ester was replaced with the product of the above step 2 (2S,3S)-N-(tert-butoxycarbonyl)-3-hydroxy-2-pyrrolecarboxylic acid methyl ester. 1 H NMR (400 MHz, D 2 O): δ 5.12 - 5.10 (t, 1H), 5.09 - 5.06 (t, 1H), 4.95 - 4.94 (m, 1H), 4.20 - 4.19 (t, 1H), 3.87 -3.86(d,2H), 3.78-3.73(t,2H), 2.58-2.53(t,2H),2.53-2.48(m,1H), 2.47-2.37(m,1H), 2.29-2.22(m, 2H). ESI-MS m/z: 387.0 [M+H] + .
实施例13 (2S,4R)-1-(((S)-4-氨基-1-(5-((1S,2R)-1-氨基-2-羟基丙基)-1,3,4-噁二唑-2-基)-4-氧代丁基)氨基甲酰基)-4-羟基四氢吡咯-2-甲酸的制备Example 13 (2S,4R)-1-(((S)-4-amino-1-(5-((1S,2R)-1-amino-2-hydroxypropyl)-1,3,4- Preparation of oxadiazol-2-yl)-4-oxobutyl)carbamoyl)-4-hydroxytetrahydropyrrole-2-carboxylic acid
Figure PCTCN2018084352-appb-000101
Figure PCTCN2018084352-appb-000101
步骤1 N-(叔丁氧羰基)-O-(叔丁基)-L-苏氨酸甲酯的制备Step 1 Preparation of N-(tert-Butoxycarbonyl)-O-(tert-butyl)-L-threonine methyl ester
Figure PCTCN2018084352-appb-000102
Figure PCTCN2018084352-appb-000102
将N-(叔丁氧羰基)-O-(叔丁基)-L-苏氨酸(5g,18.2mmol,1eq)溶于N,N-二甲基甲酰胺(35mL)中,向其溶液中加入碘甲烷(2.8g,20.0mmol,1.1eq),碳酸钾(7.9g,54.5mmol,3eq),室温下搅拌约3h。反应完全后,向混合溶液中加入水和乙酸乙酯萃取,有机相用水,饱和氯化钠溶液洗涤,无水硫酸钠干燥,抽滤,浓缩得淡黄色油状产物(5.2g),产率为99.0%。N-(tert-Butoxycarbonyl)-O-(tert-butyl)-L-threonine (5 g, 18.2 mmol, 1 eq) was dissolved in N,N-dimethylformamide (35 mL). Methyl iodide (2.8 g, 20.0 mmol, 1.1 eq), potassium carbonate (7.9 g, 54.5 mmol, 3 eq) was added and stirred at room temperature for about 3 h. After the reaction was completed, water and ethyl acetate were added to the mixture, and the mixture was evaporated. 99.0%.
步骤2 ((2S,3R)-3-(叔丁氧基)-1-肼基-1-氧代丁-2-基)氨基甲酸叔丁酯的制备Step 2 Preparation of (2S,3R)-3-(tert-butoxy)-1-indolyl-1-oxobutan-2-yl)carbamic acid tert-butyl ester
Figure PCTCN2018084352-appb-000103
Figure PCTCN2018084352-appb-000103
将N-(叔丁氧羰基)-O-(叔丁基)-L-苏氨酸甲酯(5.2g,18.0mmol)溶于甲醇,向其溶液中加入水合肼(15mL),室温下搅拌24h。反应完全后,将甲醇旋干,向混合物中加入水和乙酸乙酯萃取,有机相用水,饱和氯化钠溶液洗涤,无水硫酸钠干燥,抽滤,浓缩,得浅黄色固体(5.2g),产率为100.0%。N-(tert-Butoxycarbonyl)-O-(tert-butyl)-L-threonine methyl ester (5.2 g, 18.0 mmol) was dissolved in methanol, and hydrazine hydrate (15 mL) was added to the solution and stirred at room temperature. 24h. After the reaction was completed, the methanol was evaporated to dryness. EtOAc was evaporated. The yield was 100.0%.
步骤3 ((5S,10S,11R)-1-(9H-芴-9-基)-11,13,13-三甲基-3,6,9-三氧代-5-(3-氧代-3-(三苯甲基氨基)丙基)-2,12-二氧杂-4,7,8-三氮杂十四烷-10-基)氨基甲酸叔丁酯的制备Step 3 ((5S,10S,11R)-1-(9H-芴-9-yl)-11,13,13-trimethyl-3,6,9-trioxo-5-(3-oxo Preparation of tert-butyl 3-(tritylmethylamino)propyl)-2,12-dioxa-4,7,8-triazatetradecane-10-yl)carbamate
Figure PCTCN2018084352-appb-000104
Figure PCTCN2018084352-appb-000104
将((2S,3R)-3-(叔丁氧基)-1-肼基-1-氧代丁-2-基)氨基甲酸叔丁酯(5.2g,18.0mmol,1eq),N2-(((9H-芴-9-基)甲氧基)羰基)-N5-三苯甲基-L-谷氨酰胺(11.0g,18.0mmol,1eq),1-乙基-(3-二甲基氨基丙基)碳化二亚胺盐酸盐(4.1g,21.6mmol,1.2eq),1-羟基苯并三氮唑(2.9g,21.6mmol,1.2eq)溶于N,N-二甲基甲酰胺(40mL)中,于0℃下缓慢滴入N-甲基吗啉(4.5g,44.9mmol,2.5eq),滴完后室温下搅拌反应12h。反应完全后,将混合溶液投入冰水中搅拌,固体析出,抽滤。用二氯甲烷将固体溶解,分去水相,有机相旋干,加入石油醚洗涤,抽滤,固体干燥,得纯白色固体(10.5g),产率为66.0%。tert-Butyl ((2S,3R)-3-(tert-butoxy)-1-indolyl-1-oxobutan-2-yl)carbamate (5.2 g, 18.0 mmol, 1 eq), N2- ( ((9H-芴-9-yl)methoxy)carbonyl)-N5-trityl-L-glutamine (11.0 g, 18.0 mmol, 1 eq), 1-ethyl-(3-dimethyl Aminopropyl)carbodiimide hydrochloride (4.1 g, 21.6 mmol, 1.2 eq), 1-hydroxybenzotriazole (2.9 g, 21.6 mmol, 1.2 eq) dissolved in N,N-dimethyl N-methylmorpholine (4.5 g, 44.9 mmol, 2.5 eq) was slowly added dropwise to the amide (40 mL) at 0 ° C, and the mixture was stirred at room temperature for 12 h. After the reaction was completed, the mixed solution was poured into ice water and stirred, and the solid was precipitated and suction filtered. The solid was dissolved in dichloromethane, the aqueous phase was separated, dried, evaporated, evaporated, evaporated, evaporated,
步骤4 ((1S,2R)-1-(5-((S)-1-((((9H-芴-9-基)甲氧基)羰基)氨基)-4-氧代-4-(三苯甲基氨基)丁基)-1,3,4-噁二唑-2-基)-2-(叔丁氧基)丙基)氨基甲酸叔丁酯的制备Step 4 ((1S,2R)-1-(5-((S)-1-(((9H-芴-9-yl)methoxy)carbonyl)amino)-4-oxo-4-() Preparation of tert-butyl ester of tritylamino)butyl)-1,3,4-oxadiazol-2-yl)-2-(tert-butoxy)propyl)carbamate
Figure PCTCN2018084352-appb-000105
Figure PCTCN2018084352-appb-000105
将((5S,10S,11R)-1-(9H-芴-9-基)-11,13,13-三甲基-3,6,9-三氧代-5-(3-氧代-3-(三苯甲基氨基)丙基)-2,12-二氧杂-4,7,8-三氮杂十四烷-10-基)氨基甲酸叔丁酯(10.5g,11.9mmol,1eq)溶于四氢呋喃(75mL)和N,N-二甲基甲酰胺(30mL)中,于0℃下加入三苯基膦(6.2g,23.8mmol,2eq)和碘(6.0g,23.8mmol,2eq)。当碘完全溶解后,加入三乙胺(4.8g,47.6mmol,4eq),加完后于室温下搅拌5h。反应完全后,向混合溶液中加入水和乙酸乙酯,有机相用饱和硫代硫酸钠溶液和饱和氯化钠溶液洗涤,无水硫酸钠干燥,抽滤,浓缩制砂,柱层析纯化得白色固体(3.9g),产率为38.0%。((5S,10S,11R)-1-(9H-芴-9-yl)-11,13,13-trimethyl-3,6,9-trioxo-5-(3-oxo- tert-Butyl 3-(tritylamino)propyl)-2,12-dioxa-4,7,8-triazatetradecane-10-yl)carbamate (10.5 g, 11.9 mmol, 1 eq) was dissolved in tetrahydrofuran (75 mL) and N,N-dimethylformamide (30 mL), and triphenylphosphine (6.2 g, 23.8 mmol, 2 eq) and iodine (6.0 g, 23.8 mmol, 2eq). After the iodine was completely dissolved, triethylamine (4.8 g, 47.6 mmol, 4 eq) was added, and the mixture was stirred at room temperature for 5 h. After the reaction is completed, water and ethyl acetate are added to the mixed solution, and the organic phase is washed with a saturated sodium thiosulfate solution and a saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography. White solid (3.9 g), yield 38.0%.
步骤5 ((1S,2R)-1-(5-((S)-1-氨基-4-氧代-4-(三苯甲基氨基)丁基)-1,3,4-噁二唑-2-基)-2-(叔丁氧基)丙基)氨基甲酸叔丁酯的制备Step 5 ((1S,2R)-1-(5-((S)-1-Amino-4-oxo-4-(tritylamino)butyl)-1,3,4-oxadiazole Preparation of tert-butyl 2-yl)-2-(tert-butoxy)propyl)carbamate
Figure PCTCN2018084352-appb-000106
Figure PCTCN2018084352-appb-000106
将((1S,2R)-1-(5-((S)-1-((((9H-芴-9-基)甲氧基)羰基)氨基)-4-氧代-4-(三苯甲基氨基)丁基)-1,3,4-噁二唑-2-基)-2-(叔丁氧基)丙基)氨基甲酸叔丁酯(3.9g,4.5mmol)溶于二氯甲烷(20mL),加入二乙胺(20mL),室温下搅拌4h。反应完全后,浓缩制砂,柱层析纯化(乙酸乙酯:石油醚=4:1)。得白色固体(1.5g),产率为51.7%。((1S,2R)-1-(5-((S)-1-(((9H-芴-9-yl)methoxy)carbonyl)amino)-4-oxo-4-(3) Benzylamino)butyl)-1,3,4-oxadiazol-2-yl)-2-(tert-butoxy)propyl)carbamic acid tert-butyl ester (3.9 g, 4.5 mmol) dissolved in two Methyl chloride (20 mL) was added diethylamine (20 mL). After completion of the reaction, the mixture was concentrated and purified by column chromatography (ethyl acetate: petroleum ether = 4:1). A white solid (1.5 g) was obtained in a 51.7% yield.
步骤6(2S,4R)-4-羟基四氢吡咯-2-甲酸甲酯的制备Preparation of Step 6(2S,4R)-4-Hydroxytetrahydropyrrole-2-carboxylic Acid Methyl Ester
Figure PCTCN2018084352-appb-000107
Figure PCTCN2018084352-appb-000107
制备方法同实施例12中步骤1的制备方法,不同的是将(2S,3S)-3-羟基四氢吡咯-2-甲酸替换成了(2S,4R)-4-羟基四氢吡咯-2-甲酸。The preparation method is the same as the preparation method of the first step in the embodiment 12 except that (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid is replaced by (2S,4R)-4-hydroxytetrahydropyrrole-2. - Formic acid.
步骤7 (2S,4R)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-2-甲酸甲酯的制备Step 7 Preparation of (2S,4R)-4-((tert-Butyldimethylsilyl)oxy)tetrahydropyrrole-2-carboxylic acid methyl ester
Figure PCTCN2018084352-appb-000108
Figure PCTCN2018084352-appb-000108
制备方法同实施例11中步骤1的制备方法,不同的是将(2S,4R)-N-(叔丁氧羰基)-4-羟基-2-吡咯甲酸甲酯替换成了(2S,4R)-4-羟基四氢吡咯-2-甲酸甲酯。The preparation method is the same as the preparation method of the first step in the embodiment 11, except that the methyl (2S,4R)-N-(tert-butoxycarbonyl)-4-hydroxy-2-pyrrolecarboxylate is replaced with (2S, 4R). Methyl 4-hydroxytetrahydropyrrole-2-carboxylate.
步骤8(2S,4R)-4-((叔丁基二甲基硅基)氧基)-1-(氯甲酰基)四氢吡咯-2-甲酸甲酯的制备Preparation of methyl (2S,4R)-4-((tert-butyldimethylsilyl)oxy)-1-(chloroformyl)tetrahydropyrrole-2-carboxylate
Figure PCTCN2018084352-appb-000109
Figure PCTCN2018084352-appb-000109
将(2S,4R)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-2-甲酸甲酯(1g,3.9mmol,1eq)溶于二氯甲烷(100mL),加入吡啶(0.4g,5.0mmol,1.3eq),于-5℃向溶液中滴加三光气的二氯甲烷溶液(0.5g,1.7mmol,0.44eq,溶于15mL的DCM),冰浴搅拌1h后升至室温搅拌反应18h。用柠檬酸和二氯甲烷萃取,有机相用饱和碳酸氢钠,水洗,浓缩制砂,柱层析纯化,得油状液体(600mg),产率为48.4%。Methyl (2S,4R)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole-2-carboxylate (1 g, 3.9 mmol, 1 eq) was dissolved in dichloromethane (100 mL) Pyridine (0.4 g, 5.0 mmol, 1.3 eq) was added dropwise to a solution of methylene chloride (0.5 g, 1.7 mmol, 0.44 eq, dissolved in 15 mL of DCM). The reaction was stirred at room temperature for 18 h. The mixture was extracted with citric acid and methylene chloride. EtOAc (EtOAc m.
步骤9 (1R,4S)-2-(((S)-1-(5-((1S,2R)-2-叔丁氧基-1-(叔丁氧羰基)氨基)丙基)-1,3,4-噁二唑-2-基)-4-氧基-4-(三苯甲基氨基)丁基)氨基甲酰基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸甲酯的制备Step 9 (1R,4S)-2-(((S)-1-(5-((1S,2R)-2-tert-butoxy-1-(tert-butoxycarbonyl)amino)propyl)-1 , 3,4-oxadiazol-2-yl)-4-oxy-4-(tritylamino)butyl)carbamoyl)-4-((tert-butyldimethylsilyl)oxy Preparation of methyl tetrahydropyrrole-1-carboxylate
Figure PCTCN2018084352-appb-000110
Figure PCTCN2018084352-appb-000110
将((1S,2R)-1-(5-((S)-1-氨基-4-氧代-4-(三苯甲基氨基)丁基)-1,3,4-噁二唑-2-基)-2-(叔丁氧基)丙基)氨基甲酸叔丁酯(700mg,1.1mmol,1eq)溶于二氧六环(10mL)中,向其溶液中加入三乙胺(331mg,3.3mmol,3eq)和(2S,4R)-4-((叔丁基二甲基硅基)氧基)-1-(氯甲酰基)四氢吡咯-2-甲酸甲酯(422mg,1.3mmol,1.2eq),升温至回流,搅拌12h。向混合溶液中加入柠檬酸和乙酸乙酯,有机相用饱和碳酸氢钠溶液,水,饱和氯化钠溶液洗涤,无水硫酸钠干燥,抽滤,浓缩制砂,柱层析纯化(EA:PE=3:2)。得白色固体(300mg),产率为29.7%。((1S,2R)-1-(5-((S)-1-Amino-4-oxo-4-(tritylamino)butyl)-1,3,4-oxadiazole- tert-Butyl 2-(ethyl)-2-(tert-butoxy)propyl)carbamate (700 mg, 1.1 mmol, 1 eq) was dissolved in dioxane (10 mL) and triethylamine (331 mg) , 3.3 mmol, 3 eq) and (2S,4R)-4-((tert-butyldimethylsilyl)oxy)-1-(chloroformyl)tetrahydropyrrole-2-carboxylic acid methyl ester (422 mg, 1.3 Methyl, 1.2 eq), warmed to reflux and stirred for 12 h. To the mixed solution, citric acid and ethyl acetate were added, and the organic phase was washed with saturated sodium bicarbonate solution, water, saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, filtered, and purified by column chromatography. PE=3:2). A white solid (300 mg) was obtained in a yield of 29.7%.
步骤10 (1R,4S)-2-(((S)-1-(5-((1S,2R)-2-叔丁氧基-1-(叔丁氧羰基)氨基)丙基)-1,3,4-噁二唑-2-基)-4-氧基-4-(三苯甲基氨基)丁基)氨基甲酰基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸的 制备Step 10 (1R,4S)-2-(((S)-1-(5-((1S,2R)-2-tert-butoxy-1-(tert-butoxycarbonyl)amino)propyl)-1 , 3,4-oxadiazol-2-yl)-4-oxy-4-(tritylamino)butyl)carbamoyl)-4-((tert-butyldimethylsilyl)oxy Preparation of tetrahydropyrrole-1-carboxylic acid
Figure PCTCN2018084352-appb-000111
Figure PCTCN2018084352-appb-000111
将(1R,4S)-2-(((S)-1-(5-((1S,2R)-2-叔丁氧基-1-(叔丁氧羰基)氨基)丙基)-1,3,4-噁二唑-2-基)-4-氧基-4-(三苯甲基氨基)丁基)氨基甲酰基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸甲酯(300mg,0.32mmol,1eq)溶于四氢呋喃(2mL),加入2M氢氧化锂溶液(4.5mg,0.64mmol,2eq溶于0.05mL水中),加热至回流(70℃)搅拌3h。用柠檬酸和二氯甲烷萃取,有机相用柠檬酸,水洗,浓缩制砂,柱层析纯化(EA:PE=9:1)得白色固体(250mg),产率为84.6%。(1R,4S)-2-(((S)-1-(5-((1S,2R)-2-tert-butoxy-1-(tert-butoxycarbonyl)amino)propyl)-1, 3,4-oxadiazol-2-yl)-4-oxy-4-(tritylamino)butyl)carbamoyl)-4-((tert-butyldimethylsilyl)oxy Methyl tetrahydropyrrole-1-carboxylate (300 mg, 0.32 mmol, 1 eq) was dissolved in tetrahydrofuran (2 mL). A 2M solution of lithium hydroxide (4.5 mg, 0.64 mmol, 2 eq dissolved in 0.05 mL water) Stir at 70 ° C for 3 h. The extract was extracted with citric acid and dichloromethane, and the organic phase was washed with EtOAc, EtOAc (EtOAc)
步骤11 (2S,4R)-1-(((S)-4-氨基-1-(5-((1S,2R)-1-氨基-2-羟基丙基)-1,3,4-噁二唑-2-基)-4-氧代丁基)氨基甲酰基)-4-羟基四氢吡咯-2-甲酸的制备Step 11 (2S,4R)-1-(((S)-4-Amino-1-(5-((1S,2R)-1-amino-2-hydroxypropyl)-1,3,4- oxox Preparation of oxazol-2-yl)-4-oxobutyl)carbamoyl)-4-hydroxytetrahydropyrrole-2-carboxylic acid
制备方法同实施例11中步骤8的制备方法,不同的是将(2S,4R)-2-(5-((6S,10S)-10-(叔丁氧甲基)-13,13-二甲基-3,8,11-三氧代-1,1,1-三苯基-12-氧基-2,7,9-氮杂十四烷-6-基)-1,3,4-噁二唑-2-基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸叔丁酯替换成了(1R,4S)-2-(((S)-1-(5-((1S,2R)-2-叔丁氧基-1-(叔丁氧羰基)氨基)丙基)-1,3,4-噁二唑-2-基)-4-氧基-4-(三苯甲基氨基)丁基)氨基甲酰基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯-1-甲酸,可制得目标化合物。 1H NMR(400MHz,D 2O:δ4.67-4.65(d,1H),4.39-4.35(m,1H),4.23-4.19(m,1H),4.15-4.13(t,1H),3.05-3.02(m,2H),2.90-2.89(d,2H),2.52-2.48(t,2H),2.46-2.43(m,1H),2.34-2.29(m,1H),2.12-2.10(t,1H),1.35-1.33(d,3H).ESI-MS m/z:401.2[M+H] +. The preparation method is the same as the preparation method of the step 8 in the embodiment 11, except that (2S,4R)-2-(5-((6S,10S)-10-(tert-butoxymethyl)-13,13-di Methyl-3,8,11-trioxo-1,1,1-triphenyl-12-oxy-2,7,9-azatetradecane-6-yl)-1,3,4 - Oxadiazol-2-yl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole-1-carboxylic acid tert-butyl ester was replaced by (1R,4S)-2-((( S)-1-(5-((1S,2R)-2-tert-butoxy-1-(tert-butoxycarbonyl)amino)propyl)-1,3,4-oxadiazol-2-yl) -4-Oxo-4-(tritylamino)butyl)carbamoyl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole-1-carboxylic acid can be obtained Target compound. 1 H NMR (400 MHz, D 2 O: δ 4.67-4.65 (d, 1H), 4.39 - 4.35 (m, 1H), 4.23-4.19 (m, 1H), 4.15 - 4.13 (t, 1H), 3.05- 3.02 (m, 2H), 2.90-2.89 (d, 2H), 2.52-2.48 (t, 2H), 2.46-2.43 (m, 1H), 2.34-2.29 (m, 1H), 2.12-2.10 (t, 1H) ), 1.35-1.33 (d, 3H). ESI-MS m/z: 401.2 [M+H] + .
实施例14 (2S,3S)-1-(((S)-4-氨基-1-(5-((1S,2R)-1-氨基-2-羟基丙基)-1,3,4-噁二唑-2-基)-4-氧代丁基)氨基甲酰基)-3-羟基四氢吡咯-2-甲酸的制备Example 14 (2S,3S)-1-(((S)-4-amino-1-(5-((1S,2R)-1-amino-2-hydroxypropyl)-1,3,4- Preparation of oxadiazol-2-yl)-4-oxobutyl)carbamoyl)-3-hydroxytetrahydropyrrole-2-carboxylic acid
Figure PCTCN2018084352-appb-000112
Figure PCTCN2018084352-appb-000112
制备方法同实施例13的制备方法,不同的是将步骤7中的4-羟基脯氨酸换成了3-羟基脯氨酸,可制得目标化合物。 1H NMR(400MHz,D 2O):δ4.67-4.65(d,1H),4.43-4.42(d,1H),4.39-4.35(m,1H),4.23-4.19(m,1H),4.15-4.13(t,1H),3.57-3.45(t,2H),3.05-3.02(m,2H),2.90-2.89(d,2H),2.46-2.43(m,1H),2.34-2.29(m,1H),1.35-1.33(d,3H).ESI-MS m/z:401.3[M+H] +. The preparation method is the same as the production method of Example 13, except that the 4-hydroxyproline in Step 7 is replaced with 3-hydroxyproline to prepare the target compound. 1 H NMR (400 MHz, D 2 O): δ 4.67-4.65 (d, 1H), 4.43-4.42 (d, 1H), 4.39-4.35 (m, 1H), 4.23-4.19 (m, 1H), 4.15 -4.13(t,1H),3.57-3.45(t,2H),3.05-3.02(m,2H),2.90-2.89(d,2H),2.46-2.43(m,1H),2.34-2.29(m, 1H), 1.35-1.33 (d, 3H). ESI-MS m/z: 401.3 [M+H] + .
实施例15 (2S,3R)-2-(3-((S)-4-氨基-1-(5-(1-氨基-3-(羟基甲基)环丁基)-1,3,4-噁二唑-2-基)-4-氧代丁基)脲基)-3-羟基丁酸的制备Example 15 (2S,3R)-2-(3-((S)-4-Amino-1-(5-(1-amino-3-(hydroxymethyl)cyclobutyl)-1,3,4 -Oxadiazol-2-yl)-4-oxobutyl)ureido)-3-hydroxybutyric acid
Figure PCTCN2018084352-appb-000113
Figure PCTCN2018084352-appb-000113
制备方法同实施例12的制备方法,不同的是将实施例12步骤1中的(2S,3S)-3-羟基四氢吡咯-2-甲酸替换成了1-氨基-3-(羟甲基)环丁烷-1-甲酸,以及将O-(叔丁基)-L-丝氨酸叔丁酯替换成O-(叔丁基)-L-苏氨酸叔丁酯,可制得目标化合物。 1H NMR(400MHz,D 2O):δ5.00-4.98(d,2H),4.17-4.15(d,1H),3.60-3.58(d,2H),2.76-2.73(d,1H),2.60–2.44(m,6H),2.24-2.22(d,1H),2.10-2.08(d,1H),1.10-1.08(d,3H).ESI-MS m/z:415.3[M+H]+. The preparation method was the same as the preparation method of Example 12 except that the (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid in the step 1 of Example 12 was replaced with 1-amino-3-(hydroxymethyl group). The target compound can be obtained by replacing cyclobutane-1-carboxylic acid with O-(tert-butyl)-L-serine tert-butyl ester with O-(tert-butyl)-L-threonine tert-butyl ester. 1 H NMR (400MHz, D 2 O): δ5.00-4.98 (d, 2H), 4.17-4.15 (d, 1H), 3.60-3.58 (d, 2H), 2.76-2.73 (d, 1H), 2.60 -2.44(m,6H),2.24-2.22(d,1H), 2.10-2.08(d,1H),1.10-1.08(d,3H).ESI-MS m/z:415.3[M+H]+.
实施例16 2-(3-((S)-4-氨基-1-(5-(1-氨基环丙基)-1,3,4-噁二唑-2-yl)-4-氧代丁基)脲基)-3-羟基丙酸的制备Example 16 2-(3-((S)-4-Amino-1-(5-(1-aminocyclopropyl)-1,3,4-oxadiazol-2-yl)-4-oxo Preparation of butyl)ureido)-3-hydroxypropionic acid
Figure PCTCN2018084352-appb-000114
Figure PCTCN2018084352-appb-000114
制备方法同实施例12的制备方法,不同的是将(2S,3S)-3-羟基四氢吡咯-2-甲酸替换成了1-氨基环丙烷-1-甲酸,可制得目标化合物。 1H NMR(400MHz,D 2O):δ5.11-5.08(t,1H),4.19-4.17(t,1H),3.89-3.83(d,2H),2.56-2.52(t,2H),2.44-2.37(m,1H),2.30-2.23(m,1H),1.63-1.59(t,2H),1.59-1.54(t,2H).ESI-MS m/z:357.2[M+H] +. The preparation method was the same as that of the production method of Example 12 except that (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid was replaced with 1-aminocyclopropane-1-carboxylic acid to obtain the target compound. 1 H NMR (400 MHz, D 2 O): δ 5.11-5.08 (t, 1H), 4.19-4.17 (t, 1H), 3.89-3.83 (d, 2H), 2.56-2.52 (t, 2H), 2.44 -2.37 (m, 1H), 2.30-2.23 (m, 1H), 1.63-1.59 (t, 2H), 1.59-1.54 (t, 2H). ESI-MS m/z: 357.2 [M+H] + .
实施例17 (S)-2-(3-((S)-4-氨基-1-(5-(1-氨基-3-(羟基)环丁基)-1,3,4-噁二唑-2-yl)-4-氧代丁基)脲基)-3-羟基丙酸的制备Example 17 (S)-2-(3-((S)-4-Amino-1-(5-(1-amino-3-(hydroxy)cyclobutyl)-1,3,4-oxadiazole) Preparation of -2-yl)-4-oxobutyl)ureido)-3-hydroxypropionic acid
Figure PCTCN2018084352-appb-000115
Figure PCTCN2018084352-appb-000115
制备方法同实施例12的制备方法,不同的是将(2S,3S)-3-羟基四氢吡咯-2-甲酸替换成了1-氨基-3-(羟甲基)环丁烷-1-甲酸,可制得目标化合物。 1H NMR(400MHz,D 2O):δ4.17-4.15(d,2H),3.89-3.83(d,2H),3.60-3.58(d,2H),2.76-2.73(d,1H),2.60–2.44(m,6H),2.24-2.22(m,1H),2.10-2.08(m,1H).ESI-MS m/z:415.3[M+H] +. The preparation method was the same as the preparation method of Example 12 except that (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid was replaced by 1-amino-3-(hydroxymethyl)cyclobutane-1- Formic acid can be used to prepare the target compound. 1 H NMR (400MHz, D 2 O): δ4.17-4.15 (d, 2H), 3.89-3.83 (d, 2H), 3.60-3.58 (d, 2H), 2.76-2.73 (d, 1H), 2.60 - 2.44 (m, 6H), 2.24 - 2.22 (m, 1H), 2.10 - 2.08 (m, 1H). ESI-MS m/z: 415.3 [M+H] + .
实施例18 1-(3-((S)-4-氨基-1-(5-((1S,2R)-1-氨基-2-羟基丙基)-1,3,4-噁二唑-2-基)-4-氧代丁基)脲基)环丙基甲酸甲酯的制备Example 18 1-(3-((S)-4-Amino-1-(5-((1S,2R)-1-amino-2-hydroxypropyl)-1,3,4-oxadiazole- Preparation of methyl 2-yl)-4-oxobutyl)ureido)cyclopropylcarboxylate
Figure PCTCN2018084352-appb-000116
Figure PCTCN2018084352-appb-000116
步骤1~5的制备方法同实施例13中步骤1~5的制备方法,步骤6~8的制备方法同实施例11 中步骤6~8的制备方法,不同的是将O-(叔丁基)-L-丝氨酸叔丁酯替换成了1-氨基环丙烷-1-甲酸甲酯,可制得目标化合物。 1H NMR(400MHz,D 2O):δ5.01-4.98(m,1H),4.61-4.59(d,1H),4.28-4.25(t,1H),3.59(s,3H),2.40-2.37(t,2H),2.30-2.27(m,1H),2.25-2.23(m,1H),1.47(t,2H),1.24-1.23(d,3H),1.13(t,2H).ESI-MS m/z:385.2[M+H] +. The preparation methods of steps 1 to 5 are the same as the preparation methods of steps 1 to 5 in Example 13, and the preparation methods of steps 6 to 8 are the same as the preparation methods of steps 6 to 8 in Example 11, except that O-(tert-butyl group) is used. The L-serine tert-butyl ester is replaced with methyl 1-aminocyclopropane-1-carboxylate to obtain the target compound. 1 H NMR (400 MHz, D 2 O): δ 5.01-4.98 (m, 1H), 4.61-4.59 (d, 1H), 4.28-4.25 (t, 1H), 3.59 (s, 3H), 2.40-2. (t, 2H), 2.30-2.27 (m, 1H), 2.25-2.23 (m, 1H), 1.47 (t, 2H), 1.24-1.23 (d, 3H), 1.13 (t, 2H). ESI-MS m/z: 385.2 [M+H] + .
实施例19 (((S)-4-氨基-1-(5-(1-氨基-3-羟基环丁基)-1,3,4-噁二唑-2-基)-4-氧代丁基)氨基甲酰基)-L-丝氨酸的制备Example 19 (((S)-4-Amino-1-(5-(1-amino-3-hydroxycyclobutyl)-1,3,4-oxadiazol-2-yl)-4-oxo Preparation of butyl)carbamoyl)-L-serine
Figure PCTCN2018084352-appb-000117
Figure PCTCN2018084352-appb-000117
制备方法同实施例12的制备方法,不同的是将(2S,3S)-3-羟基四氢吡咯-2-甲酸替换成了1-氨基-3-羟基环丁烷-1-甲酸,可制得目标化合物。 1H NMR(400MHz,D 2O):δ5.11-5.08(t,1H),4.59-4.52(m,1H),4.28-4.25(t,1H),3.89-3.83(d,2H),2.63-2.60(d,2H),2.40-2.37(t,2H),2.37-2.34(d,2H),2.30-2.27(m,1H),2.25-2.23(m,1H).ESI-MS m/z:387.2[M+H] +. The preparation method is the same as the preparation method of Example 12, except that (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid is replaced by 1-amino-3-hydroxycyclobutane-1-carboxylic acid. The target compound is obtained. 1 H NMR (400 MHz, D 2 O): δ 5.11-5.08 (t, 1H), 4.59-4.52 (m, 1H), 4.28-4.25 (t, 1H), 3.89-3.83 (d, 2H), 2.63 - 2.60 (d, 2H), 2.40-2.37 (t, 2H), 2.37-2.34 (d, 2H), 2.30-2.27 (m, 1H), 2.25-2.23 (m, 1H). ESI-MS m/z :387.2[M+H] + .
实施例20 (S)-4-(5-(1-氨基-3-(羟基甲基)环丁基)-1,3,4-噁二唑-2-基)-4-(3-((1S,2R)-1-羧基-2-羟基丙基)脲基)丁酸的制备Example 20 (S)-4-(5-(1-Amino-3-(hydroxymethyl)cyclobutyl)-1,3,4-oxadiazol-2-yl)-4-(3-( Preparation of (1S,2R)-1-carboxy-2-hydroxypropyl)ureido)butyric acid
Figure PCTCN2018084352-appb-000118
Figure PCTCN2018084352-appb-000118
制备方法同实施例15的制备方法,不同的是将N 2-(((9H-芴-9-基)甲氧基)羰基)-N 5-三苯甲基-L-谷氨酰胺替换成了(S)-2-((((9H-芴-9-基)甲氧基)羰基)氨基)-5-(叔丁氧基)-5-氧代戊酸,可制得目标化合物。 1H NMR(400MHz,D 2O):δ5.00-4.98(d,2H),4.17-4.15(d,1H),3.60-3.58(d,2H),2.76-2.73(d,1H),2.60–2.44(m,6H),2.24-2.22(d,1H),2.10-2.08(d,1H),1.10-1.08(d,3H).ESI-MSm/z:416.2[M+H] +. The preparation method is the same as the preparation method of Example 15, except that N 2 -(((9H-fluoren-9-yl)methoxy)carbonyl)-N 5 -trityl-L-glutamine is replaced with (S)-2-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(tert-butoxy)-5-oxopentanoic acid can be obtained to obtain the target compound. 1 H NMR (400MHz, D 2 O): δ5.00-4.98 (d, 2H), 4.17-4.15 (d, 1H), 3.60-3.58 (d, 2H), 2.76-2.73 (d, 1H), 2.60 - 2.44 (m, 6H), 2.24 - 2.22 (d, 1H), 2.10 - 2.08 (d, 1H), 1.10-1.08 (d, 3H). ESI-MS m/z: 416.2 [M+H] + .
实施例21 (S)-4-(5-(1-氨基-3-(羟基甲基)环丁基)-1,3,4-噁二唑-2-基)-4-(3-((S)-1-羧基-2-羟基乙基)脲基)丁酸的制备Example 21 (S)-4-(5-(1-Amino-3-(hydroxymethyl)cyclobutyl)-1,3,4-oxadiazol-2-yl)-4-(3-( Preparation of (S)-1-carboxy-2-hydroxyethyl)ureido)butyric acid
Figure PCTCN2018084352-appb-000119
Figure PCTCN2018084352-appb-000119
制备方法同实施例17的制备方法,不同的是将N 2-(((9H-芴-9-基)甲氧基)羰基)-N 5-三苯甲基-L-谷氨酰胺替换成了(S)-2-((((9H-芴-9-基)甲氧基)羰基)氨基)-5-(叔丁氧基)-5-氧代戊酸,可制得目标化合物。 1H NMR(400MHz,D 2O):δ4.17-4.15(d,2H),3.89-3.83(d,2H),3.60-3.58(d,2H),2.76-2.73(d,1H),2.60–2.44(m,6H),2.24-2.22(m,1H),2.10-2.08(m,1H).ESI-MS m/z:402.2[M+H] +. The preparation method is the same as the preparation method of Example 17, except that N 2 -(((9H-fluoren-9-yl)methoxy)carbonyl)-N 5 -trityl-L-glutamine is replaced with (S)-2-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-(tert-butoxy)-5-oxopentanoic acid can be obtained to obtain the target compound. 1 H NMR (400MHz, D 2 O): δ4.17-4.15 (d, 2H), 3.89-3.83 (d, 2H), 3.60-3.58 (d, 2H), 2.76-2.73 (d, 1H), 2.60 - 2.44 (m, 6H), 2.24 - 2.22 (m, 1H), 2.10 - 2.08 (m, 1H). ESI-MS m/z: 402.2 [M+H] + .
实施例22 ((1-(5-((1S,2R)-1-氨基-2-羟丙基)-1,3,4-二唑-2-基)环丙基)氨基甲酰基)-L-丝氨酸制备Example 22 ((1-(5-((1S,2R)-1-Amino-2-hydroxypropyl)-1,3,4-oxadiazol-2-yl)cyclopropyl)carbamoyl)- L-serine preparation
Figure PCTCN2018084352-appb-000120
Figure PCTCN2018084352-appb-000120
步骤1~5的制备方法同实施例13中步骤1~5的的制备方法,不同的是将N 2-(((9H-芴-9-基)甲氧基)羰基)-N 5-三苯甲基-L-谷氨酰胺替换成了1-((((9H-芴-9-基)甲氧基)羰基)氨基)环丙烷-1-甲酸,步骤6~8的制备方法同实施例11中6~8的制备方法,可制得目标化合物。 1H NMR(400MHz,D 2O):δ5.11-5.08(t,1H),5.01-4.98(m,1H),4.61-4.59(d,1H),3.89-3.83(d,2H),1.47(t,2H),1.24-1.23(d,3H),1.13(t,2H).ESI-MS m/z:330.2[M+H] +. The preparation method of the steps 1 to 5 is the same as the preparation method of the steps 1 to 5 in the embodiment 13, except that N 2 -(((9H-fluoren-9-yl)methoxy)carbonyl)-N 5 -3 Benzyl-L-glutamine is replaced by 1-(((9H-芴-9-yl)methoxy)carbonyl))amino)cyclopropane-1-carboxylic acid, and the preparation methods of steps 6-8 are the same In the preparation method of 6 to 8 in Example 11, the target compound can be obtained. 1 H NMR (400MHz, D 2 O): δ5.11-5.08 (t, 1H), 5.01-4.98 (m, 1H), 4.61-4.59 (d, 1H), 3.89-3.83 (d, 2H), 1.47 (t, 2H), 1.24-1.23 (d, 3H), 1.13 (t, 2H). ESI-MS m/z: 330.2 [M+H] + .
实施例23 (2S,3S)-2-(3-((S)-4-氨基-1-(5-(4-氨基四氢-2H-吡喃-4-基)-1,3,4-噁二唑-2-基)-4-氧代丁基)脲基)-3-羟基丁酸的制备Example 23 (2S,3S)-2-(3-((S)-4-Amino-1-(5-(4-aminotetrahydro-2H-pyran-4-yl)-1,3,4 -Oxadiazol-2-yl)-4-oxobutyl)ureido)-3-hydroxybutyric acid
Figure PCTCN2018084352-appb-000121
Figure PCTCN2018084352-appb-000121
步骤1 4-氨基四氢-2H-吡喃-4-甲酸甲酯的制备Step 1 Preparation of 4-aminotetrahydro-2H-pyran-4-carboxylic acid methyl ester
Figure PCTCN2018084352-appb-000122
Figure PCTCN2018084352-appb-000122
将4-氨基四氢-2H-吡喃-4-甲酸(5.00g,34.48mmol)溶于50mL甲醇中,搅拌冷却至-10℃,缓慢滴加二氯亚砜(13.96g,103.38mmol),加完升温回流反应3h。将反应液浓缩除去甲醇和二氯亚砜,得到粗产物6.5g,直接投入下一步反应。4-Aminotetrahydro-2H-pyran-4-carboxylic acid (5.00 g, 34.48 mmol) was dissolved in 50 mL of methanol, stirred and cooled to -10 ° C, and thionyl chloride (13.96 g, 103.38 mmol) was slowly added dropwise. The temperature was refluxed for 3 hours. The reaction solution was concentrated to remove methanol and dichloromethane, and 6.5 g of crude product was obtained.
步骤2 4-(叔丁氧羰基)氨基四氢-2H-吡喃-4-甲酸甲酯的制备Step 2 Preparation of methyl 4-(tert-butoxycarbonyl)aminotetrahydro-2H-pyran-4-carboxylate
Figure PCTCN2018084352-appb-000123
Figure PCTCN2018084352-appb-000123
将4-氨基四氢-2H-吡喃-4-甲酸甲酯(6.50g,0.04mol)溶解于65mL四氢呋喃和65mL水中,加入碳酸氢钠(12.10g,0.14mol),搅拌下降温至0℃,加入二碳酸二叔丁酯(8.50g,0.038mol),加完升至室温反应,约24h。将反应液浓缩残余物用乙酸乙酯萃取,饱和食盐水洗,无水硫酸钠干燥,浓缩得粗品,柱层析得到产物6.3g,收率60.8%。Methyl 4-aminotetrahydro-2H-pyran-4-carboxylate (6.50 g, 0.04 mol) was dissolved in 65 mL of tetrahydrofuran and 65 mL of water, sodium bicarbonate (12.10 g, 0.14 mol) was added, and the mixture was stirred and cooled to 0 ° C. Di-tert-butyl dicarbonate (8.50 g, 0.038 mol) was added, and the reaction was allowed to warm up to room temperature for about 24 h. The reaction mixture was concentrated with EtOAc (EtOAc)EtOAc.
步骤3 4-(叔丁氧羰基)氨基四氢-2H-吡喃-4-酰肼的制备Step 3 Preparation of 4-(tert-butoxycarbonyl)aminotetrahydro-2H-pyran-4-hydrazide
Figure PCTCN2018084352-appb-000124
Figure PCTCN2018084352-appb-000124
将4-(叔丁氧羰基)氨基四氢-2H-吡喃-4-甲酸甲酯(5.30g,0.02mmol,)溶解于53mL甲醇,再加入水合肼(13mL,0.2mmol),室温反应过夜,将反应液浓缩,残余物用二氯甲烷萃取,有机层用少量水洗,无水硫酸钠干燥,浓缩,柱层析得产品2.5g,收率47%。Methyl 4-(tert-butoxycarbonyl)aminotetrahydro-2H-pyran-4-carboxylate (5.30 g, 0.02 mmol) was dissolved in 53 mL of methanol, then hydrazine hydrate (13 mL, 0.2 mmol). The reaction mixture was concentrated, and the residue was crystallised from methylene chloride.
步骤4 (4-(2-(N 2-(((9H-芴-9-基)甲氧基)羰基)-N 5-三苯甲基-L-谷氨酰基)肼-1-羰基)四氢-2H吡喃-4-基)氨基甲酸叔丁酯的制备 Step 4 (4-(2-(N 2 -((9H-indol-9-yl)methoxy)carbonyl)-N 5 -trityl-L-glutamyl)indole-1-carbonyl) Preparation of tert-butyl tetrahydro-2H-pyran-4-yl)carbamate
Figure PCTCN2018084352-appb-000125
Figure PCTCN2018084352-appb-000125
将4-(叔丁氧羰基)氨基四氢-2H-吡喃-4-酰肼(1.40g,5.40mmol)溶于14mL DMF中,室温下加入N  2-((((9H-芴-9-基)甲氧基)羰基)-N 5-三苯甲基-L-谷氨酰胺(3.30g,5.40mmol),待溶解完,再加入NMM(1.36g,13.50mmol)和HATU(2.05g,5.40mmol),加完室温反应3h,将反应液加入60mL冰水中,析出固体,抽滤,水洗,干燥得到产物4.5g,收率97.8%。 4-(tert-Butoxycarbonyl)aminotetrahydro-2H-pyran-4-hydrazide (1.40 g, 5.40 mmol) was dissolved in 14 mL of DMF and N 2 -((((9H-芴-9) - yl) methoxy) carbonyl) -N 5 - trityl-glutamine -L- (3.30g, 5.40mmol), until complete dissolution, was added NMM (1.36g, 13.50mmol) and HATU (2.05g , 5.40 mmol), adding room temperature reaction for 3 h, the reaction liquid was added to 60 mL of ice water, and the solid was precipitated, suction filtered, washed with water, and dried to give a product 4.5 g, yield 97.8%.
步骤5 (S)-(4-(5-(1-((((9H-芴-9-基)甲氧基)羰基)氨基)-4-羰基-4-(三本甲基氨基)丁基)-1,3,4-噁二唑-2-基)四氢-2H-吡喃-4-基)氨基甲酸叔丁酯的制备Step 5 (S)-(4-(5-(1-(((9H-芴-9-yl))methoxy)carbonyl)amino)-4-carbonyl-4-(trimethylamino)butane Of tert-butyl-1,3,4-oxadiazol-2-yl)tetrahydro-2H-pyran-4-yl)carbamate
Figure PCTCN2018084352-appb-000126
Figure PCTCN2018084352-appb-000126
将(4-(2-(N 2-(((9-二硫代)甲氧基)羰基)-N 5-三苯甲基-L-谷氨酰基)肼-1-羰基)-四氢-2H吡喃-4)氨基甲酸叔丁酯(4.00g,4.59mmol)溶于40mL二氯甲烷中,室温下加入三苯基膦TPP(3.40g,9.18mmol),待溶解完,将反应液降温至-10℃,缓慢向反应液中加入碘(2.32g,9.18mmol),待碘完全溶解后加入三乙胺(3.72g,36.72mmol),加完升至室温反应2h。待反应完全后将反应浓缩,用乙酸乙酯萃取,有机层分别用硫代硫酸钠溶液,饱和食盐水洗涤,无水硫酸钠干燥,浓缩,柱层析,得到2.00g产物,产率51.2%。 (4-(2-(N 2 -((9-Dithio)methoxy)carbonyl)-N 5 -trityl-L-glutamyl)indole-1-carbonyl)-tetrahydro -2H pyran-4)-tert-butyl carbamate (4.00 g, 4.59 mmol) was dissolved in 40 mL of dichloromethane, and triphenylphosphine TPP (3.40 g, 9.18 mmol) was added at room temperature. The temperature was lowered to -10 ° C, and iodine (2.32 g, 9.18 mmol) was slowly added to the reaction mixture. After the iodine was completely dissolved, triethylamine (3.72 g, 36.72 mmol) was added, and the mixture was added to room temperature for 2 h. After the reaction was completed, the reaction was concentrated and evaporated with ethyl acetate. EtOAcjjjjjjjjjjjjjj .
步骤6 (S)-(4-(5-(1-氨基-4-羰基-4-三苯甲基氨基)丁基)-1,3,4-噁二唑-2-基)四氢-2H-吡喃-4-基)氨基甲酸叔丁酯的制备Step 6 (S)-(4-(5-(1-Amino-4-carbonyl-4-tritylamino)butyl)-1,3,4-oxadiazol-2-yl)tetrahydro- Preparation of tert-butyl 2H-pyran-4-yl)carbamate
Figure PCTCN2018084352-appb-000127
Figure PCTCN2018084352-appb-000127
将(S)-(4-(5-(1-((((9H-芴-9-基)甲氧基)羰基)氨基)-4-羰基-4-(三本甲基氨基)丁基)-1,3,4-噁二唑-2-基)四氢-2H-吡喃-4-基)氨基甲酸叔丁酯(2.0g,2.11mmol),溶于20mL二氯甲烷,再加入20mL乙二胺,加完室温反应6h,TLC监测至反应完全;将反应液浓缩,柱层析得到产物黄色固体1.0g,收率83.3%。(S)-(4-(5-(1-(((9H-芴-9-yl))methoxy)carbonyl)amino)-4-carbonyl-4-(trimethylamino)butyl -1,3,4-oxadiazol-2-yl)tetrahydro-2H-pyran-4-yl)carbamic acid tert-butyl ester (2.0 g, 2.11 mmol), dissolved in dichloromethane (20 mL) 20 mL of ethylenediamine was added to the reaction at room temperature for 6 h, and the reaction was completed by TLC. The reaction mixture was concentrated and purified by column chromatography.
步骤7 N-(((S)-1-(5-(4-((二碳酸二叔丁酯基)氨基)四氢-2H-吡喃-4-基)-1,3,4-噁二唑-2-基)-4-羰基-4-(三苯甲基氨基)丁基)氨甲酰基)-O-(叔丁基)-L-苏氨酸叔丁酯的制备Step 7 N-(((S)-1-(5-(4-(di-tert-Butyl)-)-tetrahydro-2H-pyran-4-yl)-1,3,4-Ethyl Preparation of oxazol-2-yl)-4-carbonyl-4-(tritylamino)butyl)carbamoyl)-O-(tert-butyl)-L-threonine tert-butyl ester
Figure PCTCN2018084352-appb-000128
Figure PCTCN2018084352-appb-000128
将(S)-(4-(5-(1-氨基-4-羰基-4-三苯甲基氨基)丁基)-1,3,4-噁二唑-2-基)四氢-2H-吡喃-4-基)氨基甲酸叔丁酯(300mg,0.49mmol),溶解于3mL DMF中,在加入三乙胺(100mg,0.98mmol),加完降温至0℃,再加入O-(叔丁基)-N-((4-硝基苯基)氨基甲酰基)-L-异亮氨酸叔丁酯(387mg, 0.53mmol),加完升至室温反应10h,将反应液倒入20mL水中,用乙酸乙酯萃取,有机层用1M柠檬酸洗,再用饱和碳酸氢钠洗,有机层用无水硫酸钠干燥,浓缩,柱层析得到产物白色固体340mg,收率81.0%。(S)-(4-(5-(1-Amino-4-carbonyl-4-tritylamino)butyl)-1,3,4-oxadiazol-2-yl)tetrahydro-2H tert-Butyl pyran-4-yl)carbamate (300 mg, 0.49 mmol), dissolved in 3 mL of DMF, added triethylamine (100 mg, 0.98 mmol), and then cooled to 0 ° C. tert-Butyl)-N-((4-nitrophenyl)carbamoyl)-L-isoleucine tert-butyl ester (387 mg, 0.53 mmol), added to room temperature for 10 h, poured into the reaction solution The mixture was extracted with EtOAc (EtOAc)EtOAc.
步骤8 (2S,3S)-2-(3-((S)-4-氨基-1-(5-(4-氨基四氢-2H-吡喃-4-基)-1,3,4-噁二唑-2-基)-4-氧代丁基)脲基)-3-羟基丁酸的制备Step 8 (2S,3S)-2-(3-((S)-4-Amino-1-(5-(4-aminotetrahydro-2H-pyran-4-yl)-1,3,4- Preparation of oxadiazol-2-yl)-4-oxobutyl)ureido)-3-hydroxybutyric acid
Figure PCTCN2018084352-appb-000129
Figure PCTCN2018084352-appb-000129
将N-(((S)-1-(5-(4-((二碳酸二叔丁酯基)氨基)四氢-2H-吡喃-4-基)-1,3,4-噁二唑-2-基)-4-羰基-4-(三苯甲基氨基)丁基)氨甲酰基)-O-(叔丁基)-L-苏氨酸叔丁酯(300mg,0.345mmol)溶解于5mL二氯甲烷中,室温下加入5mL三氟乙酸,滴加两滴三异丙基乙胺,室温反应5h。将反应液浓缩,向残余物中加入15mL水,用二氯甲烷萃取,水层冻干,过反相柱,得到产物。 1H NMR(400MHz,D 2O)δ4.97(dd,J=9.5,4.9Hz,1H),4.25(dd,J=12.2,9.7Hz,1H),4.18(d,J=13.8Hz,1H),3.93(d,J=12.0Hz,2H),3.49(td,J=11.6,5.9Hz,2H),2.56–2.02(m,8H),1.09(d,J=6.3Hz,3H).ESI-MS m/z:511.2[M+H] + N-(((S)-1-(5-(4-((di-tert-Butyl)))-tetrahydro-2H-pyran-4-yl)-1,3,4-oxan Zyridin-2-yl)-4-carbonyl-4-(tritylamino)butyl)carbamoyl)-O-(tert-butyl)-L-threonine tert-butyl ester (300 mg, 0.345 mmol) Dissolve in 5 mL of dichloromethane, add 5 mL of trifluoroacetic acid at room temperature, add two drops of triisopropylethylamine dropwise, and react at room temperature for 5 h. The reaction solution was concentrated, and 15 mL of water was added to the residue, and the mixture was extracted with methylene chloride. The aqueous layer was lyophilized and passed through a reverse phase column to give product. 1 H NMR (400 MHz, D 2 O) δ 4.97 (dd, J = 9.5, 4.9 Hz, 1H), 4.25 (dd, J = 12.2, 9.7 Hz, 1H), 4.18 (d, J = 13.8 Hz, 1H) ), 3.93 (d, J = 12.0 Hz, 2H), 3.49 (td, J = 11.6, 5.9 Hz, 2H), 2.56 - 2.02 (m, 8H), 1.09 (d, J = 6.3 Hz, 3H). ESI - MS m/z: 511.2 [M+H] + .
实施例24 (S)-2-(3-((S)-4-氨基-1-(5-(4-氨基四氢-2H-吡喃-4-基)-1,3,4-噁二唑-2-基)-4-氧代丁基)脲基)-3-羟基丙酸的制备Example 24 (S)-2-(3-((S)-4-Amino-1-(5-(4-aminotetrahydro-2H-pyran-4-yl)-1,3,4-oxa) Preparation of oxazol-2-yl)-4-oxobutyl)ureido)-3-hydroxypropionic acid
Figure PCTCN2018084352-appb-000130
Figure PCTCN2018084352-appb-000130
参照实施例23的合成路线,即可合成(((S)-4-氨基-1-(5-(4-氨基-2H-吡喃-4-基)-1,3,4-噁二唑-2-基)-4-丁基羰基)甲酰胺)-L-丝氨酸。 1H NMR(400MHz,D 2O)δ4.96(dd,J=9.7,4.9Hz,1H),4.24(t,J=4.2Hz,1H),3.92(d,J=12.3Hz,2H),3.83(dd,J=11.7,4.8Hz,1H),3.73(dd,J=11.6,3.8Hz,1H),3.48(td,J=11.1,5.8Hz,2H),2.49–2.34(m,4H),2.26(dt,J=13.0,7.0Hz,1H),2.18–2.04(m,3H).ESI-MS m/z:401.2[M+H] +. By the synthesis route of Example 23, (((S)-4-amino-1-(5-(4-amino-2H-pyran-4-yl)-1,3,4-oxadiazole) can be synthesized. -2-yl)-4-butylcarbonyl)carboxamide)-L-serine. 1 H NMR (400 MHz, D 2 O) δ 4.96 (dd, J = 9.7, 4.9 Hz, 1H), 4.24 (t, J = 4.2 Hz, 1H), 3.92 (d, J = 12.3 Hz, 2H), 3.83 (dd, J = 11.7, 4.8 Hz, 1H), 3.73 (dd, J = 11.6, 3.8 Hz, 1H), 3.48 (td, J = 11.1, 5.8 Hz, 2H), 2.49 - 2.34 (m, 4H) , 2.26 (dt, J = 13.0, 7.0 Hz, 1H), 2.18 - 2.04 (m, 3H). ESI-MS m/z: 401.2 [M+H] + .
实施例25 (S)-4-(5-(4-氨基四氢-2H-吡喃-4-基)-1,3,4-噁二唑-2-基)-4-(3-((1S,2S)-1-羧基-2-羟丙基)脲基)丁酸的制备Example 25 (S)-4-(5-(4-Aminotetrahydro-2H-pyran-4-yl)-1,3,4-oxadiazol-2-yl)-4-(3-( Preparation of (1S,2S)-1-carboxy-2-hydroxypropyl)ureido)butyric acid
Figure PCTCN2018084352-appb-000131
Figure PCTCN2018084352-appb-000131
参照实施例23的合成路线,即可合成(S)-4-(5-(4-氨基四氢-2H-吡喃-4-基)-1,3,4-噁二唑-2-基)-4-(3-((1S,2S)-1-羧基-2-羟丙基)脲基)丁酸。 1H NMR(400MHz,D 2O)δ4.94(d,J=7.0Hz,1H),4.19(d,J=5.7Hz,1H),4.07(s,1H),3.88(d,J=12.5Hz,2H),3.44(t,J=12.0Hz,2H),2.41(dd,J= 19.7,10.2Hz,3H),2.29–2.15(m,1H),2.08(t,J=10.8Hz,3H),1.03(d,J=6.2Hz,3H).ESI-MSm/z:416.2[M+H] +By the synthesis route of Example 23, (S)-4-(5-(4-aminotetrahydro-2H-pyran-4-yl)-1,3,4-oxadiazol-2-yl can be synthesized. -4-(3-((1S,2S)-1-carboxy-2-hydroxypropyl)ureido)butyric acid. 1 H NMR (400 MHz, D 2 O) δ 4.94 (d, J = 7.0 Hz, 1H), 4.19 (d, J = 5.7 Hz, 1H), 4.07 (s, 1H), 3.88 (d, J = 12.5) Hz, 2H), 3.44 (t, J = 12.0 Hz, 2H), 2.41 (dd, J = 19.7, 10.2 Hz, 3H), 2.29 - 2.15 (m, 1H), 2.08 (t, J = 10.8 Hz, 3H) ), 1.03 (d, J = 6.2 Hz, 3H). ESI-MS m/z: 416.2 [M+H] + .
实施例26 (S)-4-(5-(4-氨基四氢-2H-吡喃-4-基)-1,3,4-噁二唑-2-基)-4-(3-((S)-1-羧基-2-羟基乙基)脲基)丁酸的制备Example 26 (S)-4-(5-(4-Aminotetrahydro-2H-pyran-4-yl)-1,3,4-oxadiazol-2-yl)-4-(3-( Preparation of (S)-1-carboxy-2-hydroxyethyl)ureido)butyric acid
Figure PCTCN2018084352-appb-000132
Figure PCTCN2018084352-appb-000132
参照实施例23的合成路线,即可合成(S)-4-(5-(4-氨基四氢-2H-吡喃-4-基)-1,3,4-噁二唑-2-基)-4-(3-((1S,2S)-1-羧基-2-羟基乙基)脲基)丁酸。 1H NMR(400MHz,D 2O)δ5.03–4.93(m,1H),4.21(d,J=4.0Hz,1H),3.91(s,2H),3.79(dd,J=11.4,4.7Hz,1H),3.76–3.66(m,1H),3.46(d,J=9.3Hz,2H),2.53–2.33(m,4H),2.23(d,J=5.1Hz,1H),2.10(s,3H).ESI-MS m/z:402.2[M+H] +. By the synthesis route of Example 23, (S)-4-(5-(4-aminotetrahydro-2H-pyran-4-yl)-1,3,4-oxadiazol-2-yl can be synthesized. -4-(3-((1S,2S)-1-carboxy-2-hydroxyethyl)ureido)butyric acid. 1 H NMR (400 MHz, D 2 O) δ 5.03 - 4.93 (m, 1H), 4.21. (d, J = 4.0 Hz, 1H), 3.91 (s, 2H), 3.79 (dd, J = 11.4, 4.7 Hz , 1H), 3.76–3.66 (m, 1H), 3.46 (d, J=9.3 Hz, 2H), 2.53–2.33 (m, 4H), 2.23 (d, J=5.1 Hz, 1H), 2.10 (s, 3H). ESI-MS m/z: 402.2 [M+H] + .
实施例27 (S)-4-(3-(4-氨基四氢-2H-吡喃-4-基)-1,2,4-噁二唑-5-基)-4-(3-((S)-1-羧基-2-羟基乙基)脲基)丁酸的制备Example 27 (S)-4-(3-(4-Aminotetrahydro-2H-pyran-4-yl)-1,2,4-oxadiazol-5-yl)-4-(3-( Preparation of (S)-1-carboxy-2-hydroxyethyl)ureido)butyric acid
Figure PCTCN2018084352-appb-000133
Figure PCTCN2018084352-appb-000133
步骤1 4-氨基四氢-2H-吡喃-4-甲酸甲酯的制备Step 1 Preparation of 4-aminotetrahydro-2H-pyran-4-carboxylic acid methyl ester
Figure PCTCN2018084352-appb-000134
Figure PCTCN2018084352-appb-000134
将4-氨基四氢-2H-吡喃-4-甲酸(5.00g,34.48mmol)溶于50mLMeOH,搅拌冷却至-10℃,缓慢滴加二氯亚砜(13.96g,103.38mmolq),加完升温回流反应3h。将反应液浓缩除去甲醇和二氯亚砜,得到粗产物6.5g,未经进一步处理,直接投入下一步反应。4-Aminotetrahydro-2H-pyran-4-carboxylic acid (5.00 g, 34.48 mmol) was dissolved in 50 mL MeOH, stirred and cooled to -10 ° C, and thionyl chloride (13.96 g, 103.38 mmolq) was slowly added dropwise. The temperature was refluxed for 3 h. The reaction solution was concentrated to remove methanol and dichloromethane, and 6.5 g of crude product was obtained.
步骤2 4-(叔丁氧羰基)氨基四氢-2H-吡喃-4-甲酸甲酯的制备Step 2 Preparation of methyl 4-(tert-butoxycarbonyl)aminotetrahydro-2H-pyran-4-carboxylate
Figure PCTCN2018084352-appb-000135
Figure PCTCN2018084352-appb-000135
将4-氨基四氢-2H-吡喃-4-甲酸甲酯(6.50g,0.04mol)溶解于65mL四氢呋喃和65mL水中,加入碳酸氢钠(12.10g,0.14mol),搅拌下降温至0℃,加入二碳酸二叔丁酯(8.50g,0.038mol)加完升至室温反应,约24h。将反应液浓缩残余物用乙酸乙酯萃取,饱和食盐水洗,无水硫酸钠干燥,浓缩得粗品,柱层析得到产物6.3g,收率60.8%。Methyl 4-aminotetrahydro-2H-pyran-4-carboxylate (6.50 g, 0.04 mol) was dissolved in 65 mL of tetrahydrofuran and 65 mL of water, sodium bicarbonate (12.10 g, 0.14 mol) was added, and the mixture was stirred and cooled to 0 ° C. Add di-tert-butyl dicarbonate (8.50 g, 0.038 mol) and add to room temperature for about 24 h. The reaction mixture was concentrated with EtOAc (EtOAc)EtOAc.
步骤3 4-(叔丁氧羰基)氨基四氢-2H-吡喃-4-甲酸的制备Step 3 Preparation of 4-(tert-butoxycarbonyl)aminotetrahydro-2H-pyran-4-carboxylic acid
Figure PCTCN2018084352-appb-000136
Figure PCTCN2018084352-appb-000136
将4-(叔丁氧羰基)氨基四氢-2H-吡喃-4-甲酸甲酯(7.00g,27.02mmol)溶解于70mL甲醇,再加入40mL水,再加入(1.63g,40.53mmol)室温反应4h,将反应液用1N盐酸调至pH为4-5,再用乙酸乙酯萃取,水洗干燥,浓缩得产品白色固体5.0g,收率75.7%Methyl 4-(tert-butoxycarbonyl)aminotetrahydro-2H-pyran-4-carboxylate (7.00 g, 27.02 mmol) was dissolved in 70 mL of methanol, then 40 mL water was added, then (1.63 g, 40. After reacting for 4 h, the reaction mixture was adjusted to pH 4-5 with 1N hydrochloric acid, and then extracted with ethyl acetate, washed with water and dried to give a white solid, 5.0 g, yield 75.7%.
步骤4 4-(叔丁氧羰基)氨基四氢-2H-吡喃-4-甲酰胺的制备Step 4 Preparation of 4-(tert-Butoxycarbonyl)aminotetrahydro-2H-pyran-4-carboxamide
Figure PCTCN2018084352-appb-000137
Figure PCTCN2018084352-appb-000137
将4-(叔丁氧羰基)氨基四氢-2H-吡喃-4-甲酸(5.00g,20mmol)溶于50mL THF中,室温下加入NMM(2.50g,24mmol),将反应液降温至-10℃,缓慢滴加氯甲酸乙酯(2.70g,24mmol),低温反应2h,低温下缓慢滴加氨水(13mL,160mmol),加完低温反应3h,点板反应完全,将反应液浓缩,残余物用乙酸乙酯萃取,有机层分别用1N柠檬酸,饱和碳酸氢钠,饱和食盐水洗,无水硫酸钠干燥,浓缩得到产物白色固体4.2g,收率85.7%。4-(tert-Butoxycarbonyl)aminotetrahydro-2H-pyran-4-carboxylic acid (5.00 g, 20 mmol) was dissolved in 50 mL of THF. NMM (2.50 g, 24 mmol) was added at room temperature and the reaction was cooled to - Ethyl chloroformate (2.70 g, 24 mmol) was slowly added dropwise at 10 ° C, and the reaction was carried out at low temperature for 2 h. Ammonia water (13 mL, 160 mmol) was slowly added dropwise at low temperature. After adding low temperature reaction for 3 h, the reaction was complete and the reaction solution was concentrated. The extract was extracted with EtOAc. EtOAc (EtOAc)EtOAc.
步骤5 (4-氰基四氢-2H-吡喃-4-基)氨基甲酸叔丁酯的制备Step 5 Preparation of (4-cyanotetrahydro-2H-pyran-4-yl)carbamic acid tert-butyl ester
Figure PCTCN2018084352-appb-000138
Figure PCTCN2018084352-appb-000138
将4-(叔丁氧羰基)氨基四氢-2H-吡喃-4-甲酰胺(3.70g,15.14mmol)溶于吡啶(12.0g,151.4mmol)中,低温下滴加三氟乙酸酐(4.80g,22.71mmol)加完室温反应4h,将反应液导入饱和碳酸氢钠溶液中,用乙酸乙酯萃取,有机层用1M柠檬酸洗,饱和碳酸氢钠溶液洗,无水硫酸钠干燥,浓缩得粗品,用石油醚洗,得产品2.9g,收率78%。4-(tert-Butoxycarbonyl)aminotetrahydro-2H-pyran-4-carboxamide (3.70 g, 15.14 mmol) was dissolved in pyridine (12.0 g, 151.4 mmol), and trifluoroacetic anhydride was added dropwise at low temperature ( 4.80g, 22.71mmol) After the addition of room temperature for 4h, the reaction mixture was poured into a saturated aqueous solution of sodium bicarbonate, and extracted with ethyl acetate. The organic layer was washed with 1M EtOAc. The crude product was concentrated and washed with petroleum ether to give 2.9 g of product.
步骤6 E-(4-(N'-羟基甲脒基)四氢-2H-吡喃-4-基)氨基甲酸叔丁酯的制备Step 6 Preparation of E-(4-(N'-hydroxymethylindenyl)tetrahydro-2H-pyran-4-yl)carbamic acid tert-butyl ester
Figure PCTCN2018084352-appb-000139
Figure PCTCN2018084352-appb-000139
将(4-氰基四氢-2H-吡喃-4-基)氨基甲酸叔丁酯(2.50g,0.01mol),溶于25mL乙醇中,再加入5mL水,盐酸羟胺(1.20g,0.015mol),碳酸钾(2.30g,0.015mol),加完升温回流反应。将反应液浓缩,残余物用乙酸乙酯溶解,饱和食盐水洗,无水硫酸钠干燥,浓缩得产物白色固体2.50g,收率96.5%。(4-Cyanotetrahydro-2H-pyran-4-yl)carbamic acid tert-butyl ester (2.50 g, 0.01 mol), dissolved in 25 mL of ethanol, and then added 5 mL of water, hydroxylamine hydrochloride (1.20 g, 0.015 mol) ), potassium carbonate (2.30 g, 0.015 mol), and the temperature rise reflux reaction was added. The reaction mixture was concentrated, and the residue was evaporated, evaporated, evaporated, evaporated
步骤7 叔丁基(S,Z)-4-((((9H-芴-9-基)甲氧基)羰基)氨基)-5-(((((((叔丁氧基羰基)氨基)四氢-2H-吡喃-4-基)亚甲基)氨基)氧基)-5-氧代戊酸乙酯的制备Step 7 Tert-Butyl (S,Z)-4-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-5-((((((((((((((((((( Preparation of ethyl tetrahydro-2H-pyran-4-yl)methylene)amino)oxy)-5-oxopentanoate
Figure PCTCN2018084352-appb-000140
Figure PCTCN2018084352-appb-000140
将E-(4-(N'-羟基甲脒基)四氢-2H-吡喃-4-基)氨基甲酸叔丁酯(2.00g,7.72mmol),溶解于20mL DCM中,再加入NMM(1.6g,15.44mmol),(S)-2-(((((9H-芴-9-基)甲氧基)羰基)氨基)-5-(叔丁氧基)-5-氧代戊酸(3.6g,8.49mmol)加完室温反应,TLC监测至反应完全,将反应液浓缩,残余物柱层析得到产品白色固体2.68g,收率52%。E-(4-(N'-hydroxymethylindenyl)tetrahydro-2H-pyran-4-yl)carbamic acid tert-butyl ester (2.00 g, 7.72 mmol) was dissolved in 20 mL DCM, then NMM ( 1.6 g, 15.44 mmol), (S)-2-((((9H-indol-9-yl)methoxy)carbonyl)amino)-5-(tert-butoxy)-5-oxopentanoic acid (3.6 g, 8.49 mmol) was added to room temperature, and the reaction was completed by TLC. The reaction mixture was concentrated, and the residue was subjected to column chromatography.
步骤8 叔丁基(S)-4-((((9H-芴-9-基)甲氧基)羰基)氨基)-4-(3-(4-((叔丁氧基羰基)氨基)四氢 -2H-吡喃-吡啶-4-基)-1,2,4-噁二唑-5-基)丁酸乙酯的制备Step 8 Tert-Butyl (S)-4-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-4-(3-(4-((tert-butoxycarbonyl)amino) Preparation of ethyl tetrahydro-2H-pyrano-pyridin-4-yl)-1,2,4-oxadiazol-5-yl)butanoate
Figure PCTCN2018084352-appb-000141
Figure PCTCN2018084352-appb-000141
将叔丁基(S,Z)-4-((((9H-芴-9-基)甲氧基)羰基)氨基)-5-(((((((叔丁氧基羰基)氨基)四氢-2H-吡喃-4-基)亚甲基)氨基)氧基)-5-氧代戊酸乙酯(2.20g,3.29mmol)溶解于22mL乙醇和3mL水中,再加入醋酸钠,升温回流反应,TLC监测至反应完全,将反应液浓缩,残余物用乙酸乙酯溶解,再用饱和食盐水洗,无水硫酸钠干燥,柱层析得产物1.80g,收率84.5%。tert-Butyl (S,Z)-4-((((9H-芴-9-yl)methoxy)carbonyl)amino)-5-(((((((((((((((((((((((((((((((( Tetrahydro-2H-pyran-4-yl)methylene)amino)oxy)-5-oxopentanoate (2.20 g, 3.29 mmol) was dissolved in 22 mL of ethanol and 3 mL of water, then sodium acetate was added. The mixture was heated to reflux, and the mixture was evaporated to dryness. The residue was evaporated.
步骤9 叔丁基(S)-4-氨基-4-(3-(4-((叔丁氧基羰基)氨基)四氢-2H-吡喃-4-基)-1,2,4-噁二唑-5-基)丁酸盐的制备Step 9 tert-Butyl (S)-4-amino-4-(3-(4-((tert-butoxycarbonyl)amino)tetrahydro-2H-pyran-4-yl)-1,2,4- Preparation of Oxadiazole-5-yl) Butyrate
Figure PCTCN2018084352-appb-000142
Figure PCTCN2018084352-appb-000142
将叔丁基(S)-4-((((9H-芴-9-基)甲氧基)羰基)氨基)-4-(3-(4-((叔丁氧基羰基)氨基)四氢-2H-吡喃-吡啶-4-基)-1,2,4-噁二唑-5-基)丁酸乙酯(1.80g,2.77mmol),溶于18mL二氯甲烷,再加入18mL二乙胺,加完室温反应6h,TLC监测至反应完全。将反应液浓缩,柱层析得到产物黄色固体1.0g,收率84.7%。tert-Butyl (S)-4-(((9H-fluoren-9-yl)methoxy)carbonyl)amino)-4-(3-(4-((tert-butoxycarbonyl))amino) Hydrogen-2H-pyrano-pyridin-4-yl)-1,2,4-oxadiazol-5-yl)butyric acid ethyl ester (1.80 g, 2.77 mmol), dissolved in 18 mL dichloromethane, then 18 mL Diethylamine was added to room temperature for 6 h, and TLC was monitored until the reaction was completed. The reaction solution was concentrated and subjected to column chromatography to give a white solid.
步骤10 叔丁基(S)-4-(3-(4-((叔丁氧基羰基)氨基)四氢-2H-吡喃-4-基)-1,2,4-噁二唑-5-基)-4-(3-((S)-1,3-二叔丁氧基-1-氧代丙-2-基)脲基)丁酸乙酯的制备Step 10 tert-Butyl (S)-4-(3-(4-((tert-butoxycarbonyl)amino)tetrahydro-2H-pyran-4-yl)-1,2,4-oxadiazole- Preparation of 5-ethyl)-4-(3-((S)-1,3-di-tert-butoxy-1-oxopropan-2-yl)ureido)butyric acid ethyl ester
Figure PCTCN2018084352-appb-000143
Figure PCTCN2018084352-appb-000143
将叔丁基(S)-4-氨基-4-(3-(4-((叔丁氧基羰基)氨基)四氢-2H-吡喃-4-基)-1,2,4-噁二唑-5-基)丁酸盐(300mg,0.70mmol),溶解于3mL DMF中,在加入三乙胺(142mg,1.40mmol),加完降温至0℃,再加入O-(叔丁基)-N-((4-硝基苯基)氨基甲酰基)-L-丝氨酸叔丁酯(352mg,0.77mmol),加完升至室温反应10h,将反应液倒入20mL水中,用乙酸乙酯萃取,有机层用1M柠檬酸洗,再用饱和碳酸氢钠洗,有机层用无水硫酸钠干燥,浓缩,柱层析得到产物白色固体300mg,收率80%。tert-Butyl (S)-4-amino-4-(3-(4-((tert-butoxycarbonyl)amino)tetrahydro-2H-pyran-4-yl)-1,2,4-oxo Diazol-5-yl)butyrate (300 mg, 0.70 mmol), dissolved in 3 mL of DMF, added triethylamine (142 mg, 1.40 mmol), cooled to 0 ° C, then added O- (tert-butyl) -N-((4-Nitrophenyl)carbamoyl)-L-serine tert-butyl ester (352mg, 0.77mmol), added to room temperature for 10h, the reaction solution was poured into 20mL of water, with acetic acid The organic layer was washed with 1 M citric acid and washed with saturated sodium hydrogen sulfate.
步骤11 (S)-4-(3-(4-氨基四氢-2H-吡喃-4-基)-1,2,4-噁二唑-5-基)-4-(3-((S)-1-羧基-2-羟基乙基)脲基)丁酸Step 11 (S)-4-(3-(4-Aminotetrahydro-2H-pyran-4-yl)-1,2,4-oxadiazol-5-yl)-4-(3-(( S)-1-carboxy-2-hydroxyethyl)ureido)butyric acid
Figure PCTCN2018084352-appb-000144
Figure PCTCN2018084352-appb-000144
将叔丁基(S)-4-(3-(4-((叔丁氧基羰基)氨基)四氢-2H-吡喃-4-基)-1,2,4-噁二唑-5- 基)-4-(3-((S)-1,3-二叔丁氧基-1-氧代丙-2-基)脲基)丁酸乙酯(150mg,0.219mmol)溶解于5mL二氯甲烷中,室温下加入5mL三氟乙酸,滴加两滴三异丙基乙胺,室温反应5h,将反应液浓缩,向残余物中加入15mL水,用二氯甲烷萃取,水层冻干,得到产物。 1H NMR(400MHz,D 2O)δ4.94(dt,J=17.8,8.9Hz,1H),4.16(t,J=3.5Hz,1H),3.86–3.71(m,3H),3.65(dd,J=11.6,3.5Hz,1H),3.46(t,J=11.4Hz,2H),2.40(t,J=6.9Hz,2H),2.31(d,J=13.6Hz,2H),2.18(td,J=13.3,6.6Hz,1H),2.11–1.92(m,3H).ESI-MS m/z:402.2[M+H] + tert-Butyl (S)-4-(3-(4-((tert-butoxycarbonyl)amino)tetrahydro-2H-pyran-4-yl)-1,2,4-oxadiazole-5 Ethyl 4-(3-((S)-1,3-di-tert-butoxy-1-oxopropan-2-yl)ureido)butyrate (150 mg, 0.219 mmol) was dissolved in 5 mL In dichloromethane, 5 mL of trifluoroacetic acid was added at room temperature, and two drops of triisopropylethylamine were added dropwise, and the reaction was concentrated at room temperature for 5 h. The reaction mixture was concentrated, and 15 mL of water was added to the residue, and extracted with dichloromethane. Dry to give the product. 1 H NMR (400 MHz, D 2 O) δ 4.94 (dt, J = 17.8, 8.9 Hz, 1H), 4.16 (t, J = 3.5 Hz, 1H), 3.86 - 3.71 (m, 3H), 3.65 (dd , J = 11.6, 3.5 Hz, 1H), 3.46 (t, J = 11.4 Hz, 2H), 2.40 (t, J = 6.9 Hz, 2H), 2.31 (d, J = 13.6 Hz, 2H), 2.18 (td , J = 13.3, 6.6 Hz, 1H), 2.11 - 1.92 (m, 3H). ESI-MS m/z: 402.2 [M+H] + .
实施例28 (S)-3-氨基-3-(3-((2S,4R)-4-羟基四氢吡咯-2-基)-1,2,4-噁二唑-5-基)丙酰胺的制备Example 28 (S)-3-Amino-3-(3-((2S,4R)-4-hydroxytetrahydropyrrole-2-yl)-1,2,4-oxadiazol-5-yl)propane Preparation of amide
Figure PCTCN2018084352-appb-000145
Figure PCTCN2018084352-appb-000145
步骤1:(2S,4R)-1-叔丁氧羰基-2-氰基-4-(叔丁基二甲基硅基)氧基四氢吡咯的制备Step 1: Preparation of (2S,4R)-1-tert-butoxycarbonyl-2-cyano-4-(tert-butyldimethylsilyl)oxytetrahydropyrrole
Figure PCTCN2018084352-appb-000146
Figure PCTCN2018084352-appb-000146
将化合物1a(30g,141.34mmol)溶于DMF(300mL),加入咪唑(11.55g,169.61mmol),降温至-5℃后缓慢滴加TBDMSCl(25.56g,169.61mmol)的DMF(120mL)溶液,搅拌反应2h,升至室温反应13h,将反应液倒入氯化铵饱和溶液中淬灭,加乙酸乙酯萃取、分液,有机相饱和食盐水、水洗涤数次,无水硫酸钠干燥,减压蒸馏得黄色油状液体,静置冷却后为白色固体,将粗制化合物用硅胶柱色谱纯化(洗脱剂:石油醚中0-5%的乙酸乙酯)、收集产物组分,旋干、静置冷却得44.61g白色固体化合物1b(收率:91.7%)。Compound 1a (30 g, 141.34 mmol) was dissolved in DMF (300 mL), EtOAc (11.55 g, 169.61 mmol) was added, and then cooled to -5 ° C, then a solution of TBDMSCl (25.56 g, 169.61 mmol) in DMF (120 mL) was slowly added dropwise. The reaction was stirred for 2 h, and the mixture was stirred at room temperature for 13 h. The reaction mixture was poured into a saturated aqueous solution of ammonium chloride and then evaporated. Distillation under reduced pressure gave a yellow oily solid, which was crystallised to white solid. The crude compound was purified by silica gel column chromatography (eluent: 0-5% ethyl acetate in petroleum ether). The mixture was cooled to 44.61 g of a white solid compound 1b (yield: 91.7%).
步骤2:(2S,4R)-1-叔丁氧羰基-2-((Z)-N'-羟基甲脒基)-4-(叔丁基二甲基硅基)氧基四氢吡咯的制备Step 2: (2S,4R)-1-tert-Butoxycarbonyl-2-((Z)-N'-hydroxymethylindenyl)-4-(tert-butyldimethylsilyl)oxytetrahydropyrrole preparation
Figure PCTCN2018084352-appb-000147
Figure PCTCN2018084352-appb-000147
将化合物1b(20g,61.25mmol)溶于乙醇(200mL),依次加入碳酸钾(12.70g,91.88mmol)、盐酸羟胺(6.38g,91.88mmol),搅拌回流反应5h。反应液过滤,滤液旋干,用乙酸乙酯和水溶清、分液,有机相用饱和食盐水、水洗涤数次,无水Na 2SO 4干燥,减压蒸馏,向所得固体中加入乙酸乙酯,超声、过滤,滤饼用少量四氢呋喃溶解、加入大量石油醚析晶,过滤,滤饼为白色固体纯品,滤液减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-30%的乙酸乙酯)。所得固体与滤饼合并,共得9.6g产物(收率:43.6%)。 Compound 1b (20 g, 61.25 mmol) was dissolved in ethanol (200 mL), and then potassium carbonate (12.70 g, 91.88 mmol) and hydroxylamine hydrochloride (6.38 g, 91.88 mmol) were added, and the mixture was refluxed for 5 h. The reaction was filtered, the filtrate by rotary evaporation, ethyl acetate and aqueous cleaning liquid separation, and saturated brine, the organic phase was washed several times with water, dried over anhydrous Na 2 SO 4, evaporated under reduced pressure, ethyl acetate was added to the obtained solid Ester, ultrasonic, filtration, filter cake is dissolved in a small amount of tetrahydrofuran, a large amount of petroleum ether is added to crystallize, filtered, and the filter cake is pure white solid. The filtrate is distilled under reduced pressure and purified by silica gel column chromatography (eluent: petroleum ether 0- 30% ethyl acetate). The obtained solid was combined with a filter cake to give 9.6 g of product (yield: 43.6%).
步骤3:(2S,4R)-1-叔丁氧羰基-2-(5-((S)-1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-酮基-3-(三苯甲基氨基)丙基)-1,2,4-噁二唑-3-基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯的制备Step 3: (2S,4R)-1-tert-Butoxycarbonyl-2-(5-((S)-1-(((9H-fluoren-9-yl))methoxy)carbonyl)amino)-3 -keto-3-(tritylamino)propyl)-1,2,4-oxadiazol-3-yl)-4-((tert-butyldimethylsilyl)oxy)tetrahydrol Preparation of pyrrole
Figure PCTCN2018084352-appb-000148
Figure PCTCN2018084352-appb-000148
将化合物Fmoc-Asn(Trt)-OH(20.71g,34.71mmol)溶于二氯甲烷(200mL),冰浴降温,于-7℃下加入BAST(8.45g,38.18mmol),该温度下反应8h。减压蒸馏除去反应液,加入适量正己烷,振摇、过滤,重复数次,滤饼干燥,用四氢呋喃(200mL)溶解,加入化合物1c(9.6g,26.70mmol)、氮甲基吗啉(5.40g,53.40mmol),室温搅拌反应13h。减压蒸馏除去四氢呋喃,直接加入乙醇(200mL)、乙酸钠(3.29g,40.05mmol),搅拌回流反应2.5h。减压蒸馏除去反应液,加乙酸乙酯溶解,过滤,滤液依次用饱和碳酸氢钠、1M柠檬酸和饱和食盐水洗涤,无水硫酸钠干燥,减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-25%的乙酸乙酯),得7.18g淡黄色色固体1d(收率:29.2%)。The compound Fmoc-Asn(Trt)-OH (20.71 g, 34.71 mmol) was dissolved in dichloromethane (200 mL), cooled in an ice bath, and BAST (8.45 g, 38.18 mmol) was added at -7 ° C. . The reaction solution was evaporated under reduced pressure, and the mixture was evaporated, evaporated, filtered, and then filtered, and then filtered, and then filtered and dried with tetrahydrofuran (200 mL). Compound 1c (9.6 g, 26.70 mmol), nitrogen methyl morpholine (5.40) g, 53.40 mmol), the reaction was stirred at room temperature for 13 h. Tetrahydrofuran was distilled off under reduced pressure, and ethanol (200 mL) and sodium acetate (3.29 g, 40.05 mmol) were directly added thereto, and the mixture was stirred and refluxed for 2.5 hours. The reaction mixture was evaporated under reduced pressure. EtOAc was evaporated, evaporated, evaporated, evaporated, evaporated. Agent: 0-25% ethyl acetate in petroleum ether) gave 7.18 g of pale yellow solid 1d (yield: 29.2%).
步骤4:(2S,4R)-1-叔丁氧羰基-2-(5-((S)-1-氨基-3-酮基-3-(三苯甲基氨基)丙基)-1,2,4-噁二唑-3-基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯的制备Step 4: (2S,4R)-1-tert-Butoxycarbonyl-2-(5-((S)-1-amino-3-keto-3-(tritylamino)propyl)-1, Preparation of 2,4-oxadiazol-3-yl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole
Figure PCTCN2018084352-appb-000149
Figure PCTCN2018084352-appb-000149
将化合物1d(6.0g,6.52mmol)加入到二氯甲烷(60mL)中,滴加二乙胺(30mL),室温搅拌反应约1.5h,减压蒸馏除去溶剂,用硅胶柱色谱纯化(洗脱剂:石油醚中0-45%的乙酸乙酯),得2.8g黄色油状产物1e(收率:61.5%)。Compound 1d (6.0 g, 6.52 mmol) was added to dichloromethane (60 mL), and diethylamine (30 mL) was added dropwise, and the mixture was stirred at room temperature for about 1.5 h, and the solvent was evaporated under reduced pressure. Agent: 0-45% ethyl acetate in petroleum ether) 2.8 g of product 1e (yield: 61.5%) as a yellow oil.
步骤5:(S)-3-氨基-3-(3-((2S,4R)-4-羟基四氢吡咯-2-基)-1,2,4-噁二唑-5-基)丙酰胺的制备Step 5: (S)-3-Amino-3-(3-((2S,4R)-4-hydroxytetrahydropyrrol-2-yl)-1,2,4-oxadiazol-5-yl)-propyl Preparation of amide
Figure PCTCN2018084352-appb-000150
Figure PCTCN2018084352-appb-000150
将化合物1e(600mg,0.86mmol)溶于二氯甲烷(18mL),室温下加入三氟乙酸(18mL)和催化量的三异丙基硅烷,室温搅拌反应4.5h,减压蒸馏除去溶剂,制备液相纯化得化合物1。 1H NMR(300MHz,D 2O)δ5.05(dd,J=10.1,7.3Hz,1H),4.72(s,1H),4.62(d,J=6.4Hz,1H),3.51(dd,J=12.6,4.2Hz,1H),3.33(d,J=12.7Hz,1H),2.99–2.84(m,2H),2.54–2.31(m,2H).ESI-MS m/z:242.0[M+H] +. Compound 1e (600 mg, 0.86 mmol) was dissolved in dichloromethane (18 mL), trifluoroacetic acid (18 mL) and a catalytic amount of triisopropylsilane were added at room temperature, and the mixture was stirred at room temperature for 4.5 hr. The liquid phase was purified to give Compound 1. 1 H NMR (300MHz, D 2 O) δ5.05 (dd, J = 10.1,7.3Hz, 1H), 4.72 (s, 1H), 4.62 (d, J = 6.4Hz, 1H), 3.51 (dd, J =12.6, 4.2 Hz, 1H), 3.33 (d, J = 12.7 Hz, 1H), 2.99 - 2.84 (m, 2H), 2.54 - 2.31 (m, 2H). ESI-MS m/z: 242.0 [M+ H] + .
实施例29 (2S,3R)-2-(3-((S)-3-氨基-1-(3-((2S,4R)-4-羟基四氢吡咯-2-基)-1,2,4-噁二唑-5-基)-3-氧代丙基)脲基)-3-羟基丁酸的制备Example 29 (2S,3R)-2-(3-((S)-3-Amino-1-(3-((2S,4R)-4-hydroxytetrahydropyrrol-2-yl)-1,2 Of 4-oxaoxazol-5-yl)-3-oxopropyl)ureido)-3-hydroxybutyric acid
Figure PCTCN2018084352-appb-000151
Figure PCTCN2018084352-appb-000151
步骤1:(2S,4R)-1-叔丁氧羰基-2-(5-((5S,9S)-9-((R)-1-(叔丁基)乙基)-12,12-二甲基-3,7,10-三酮基-1,1,1-三苯基-11-氧杂-2,6,8-三氮杂-5-基)-1,2,4-噁二唑-3-基)-4-((叔丁基二甲基硅基)氧基)四氢吡咯的制备Step 1: (2S,4R)-1-tert-Butoxycarbonyl-2-(5-((5S,9S)-9-((R)-1-(tert-butyl)ethyl)-12,12- Dimethyl-3,7,10-trione-1,1,1-triphenyl-11-oxa-2,6,8-triaza-5-yl)-1,2,4- Preparation of Oxadiazol-3-yl)-4-((tert-butyldimethylsilyl)oxy)tetrahydropyrrole
Figure PCTCN2018084352-appb-000152
Figure PCTCN2018084352-appb-000152
将化合物1e(1.0g,1..43mmol)溶于四氢呋喃(10mL),室温下滴加化合物2b(0.68g,1.72mmol)和三乙胺(0.29g,2.86mmol)。室温搅拌24h,减压蒸馏除去溶剂后加入乙酸乙酯和水溶解、分液,有机相用饱和食盐水洗涤数次,无水硫酸钠干燥,减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-25%的乙酸乙酯),得700mg淡黄色结晶2a(收率:51.3%)。Compound 1e (1.0 g, 1..43 mmol) was dissolved in tetrahydrofuran (10 mL). Compound 2b (0.68 g, 1.72 mmol) and triethylamine (0.29 g, 2.86 mmol). After stirring at room temperature for 24 hours, the solvent was evaporated under reduced pressure, and ethyl acetate and water were evaporated and evaporated. The organic phase was washed several times with saturated brine, dried over anhydrous sodium sulfate and evaporated. : 0-25% ethyl acetate in petroleum ether to give 700 mg of pale yellow crystal 2a (yield: 51.3%).
步骤2:(2S,3R)-2-(3-((S)-3-氨基-1-(3-((2S,4R)-4-羟基四氢吡咯-2-基)-1,2,4-噁二唑-5-基)-3-氧代丙基)脲基)-3-羟基丁酸的制备Step 2: (2S,3R)-2-(3-((S)-3-Amino-1-(3-((2S,4R)-4-hydroxytetrahydropyrrole-2-yl)-1,2 Of 4-oxaoxazol-5-yl)-3-oxopropyl)ureido)-3-hydroxybutyric acid
Figure PCTCN2018084352-appb-000153
Figure PCTCN2018084352-appb-000153
将化合物2a(550mg,0.576mmol)溶于二氯甲烷(11.5mL),室温下加入三氟乙酸(11.5mL)、催化量的三异丙基硅烷,室温搅拌反应5h,减压蒸馏除去反应溶剂,加二氯甲烷和水溶解、分液,有机相用水萃取数次,合并水相,再用二氯甲烷洗涤数次,冻干,制备液相纯化得化合物2。 1H NMR(500MHz,D 2O)δ5.46(t,J=6.4Hz,1H),5.25–5.19(m,1H),4.85(s,1H),4.26(dt,J=17.5,8.7Hz,1H),4.02(d,J=17.0Hz,1H),3.67(dt,J=13.3,6.7Hz,1H),3.55–3.47(m,1H),3.20–3.11(m,2H),2.62(dd,J=14.1,7.1Hz,1H),2.50(tt,J=9.4,4.7Hz,1H),1.22(d,J=6.4Hz,3H).ESI-MS m/z:386.9[M+H] +. Compound 2a (550 mg, 0.576 mmol) was dissolved in dichloromethane (11.5 mL), trifluoroacetic acid (11.5 mL) was added at room temperature, and a catalytic amount of triisopropylsilane was stirred at room temperature for 5 h, and the reaction solvent was evaporated under reduced pressure. Dichloromethane and water were dissolved and separated, and the organic phase was extracted several times with water, and the aqueous phase was combined, washed several times with dichloromethane, lyophilized, and purified in liquid phase to obtain compound 2. 1 H NMR (500 MHz, D 2 O) δ 5.46 (t, J = 6.4 Hz, 1H), 5.25 - 5.19 (m, 1H), 4.85 (s, 1H), 4.26 (dt, J = 17.5, 8.7 Hz) , 1H), 4.02 (d, J = 17.0 Hz, 1H), 3.67 (dt, J = 13.3, 6.7 Hz, 1H), 3.55 - 3.47 (m, 1H), 3.20 - 3.11 (m, 2H), 2.62 ( Dd, J = 14.1, 7.1 Hz, 1H), 2.50 (tt, J = 9.4, 4.7 Hz, 1H), 1.22 (d, J = 6.4 Hz, 3H). ESI-MS m/z: 386.9 [M+H ] + .
步骤3:O-(叔丁基)-N-((4-硝基苯氧基)羰基)-L-苏氨酸叔丁酯的制备Step 3: Preparation of O-(tert-butyl)-N-((4-nitrophenoxy)carbonyl)-L-threonine tert-butyl ester
Figure PCTCN2018084352-appb-000154
Figure PCTCN2018084352-appb-000154
将H-Thr(O tBu)-O tBu(15g,64.84mmol)溶于二氯甲烷(150mL),冰浴降温,于-5℃下依次滴加三乙胺(9.84g,97.26mmol)和氯甲酸-4-硝基苯脂(14.38g,71.33mmol)的二氯甲烷(60mL)溶液,撤去冰浴,室温搅拌反应2h,减压蒸馏除去溶剂,加乙酸乙酯,有固体析出,过滤,滤液减压蒸馏除去部分溶剂,加入乙酸乙酯和石油醚的1:1混合溶剂,又有固体析出,过滤,滤液减压蒸馏、用硅胶柱色谱进一步纯化(洗脱剂:石油醚中0-5%的乙酸乙酯),得13.86g黄色油状液体2b(收率:53.9%)。 H-Thr(O t Bu)-O t Bu (15 g, 64.84 mmol) was dissolved in dichloromethane (150 mL), cooled in ice-cooling, and triethylamine (9.84 g, 97.26 mmol) was added dropwise at -5 °C. And a solution of 4-chlorophenyl chloroformate (14.38 g, 71.33 mmol) in dichloromethane (60 mL), EtOAc (EtOAc) Filtration, the filtrate was evaporated under reduced pressure to remove a portion of solvent, and a mixture of ethyl acetate and petroleum ether in 1:1 was added, and solids were separated and filtered. The filtrate was evaporated under reduced pressure and purified by silica gel column chromatography (eluent: petroleum ether) 0-5% ethyl acetate) gave 13.86 g of a yellow oily liquid 2b (yield: 53.9%).
实施例30 (S)-2-(3-((S)-3-氨基-1-(3-((2S,4R)-4-羟基四氢吡咯-2-基)-1,2,4-噁二唑-5-基)-3-氧 代丙基)脲基)-3-羟基丙酸的制备Example 30 (S)-2-(3-((S)-3-Amino-1-(3-((2S,4R)-4-hydroxytetrahydropyrrole-2-yl)-1,2,4 -Oxadiazole-5-yl)-3-oxopropyl)ureido)-3-hydroxypropionic acid
Figure PCTCN2018084352-appb-000155
Figure PCTCN2018084352-appb-000155
使用与实施例29制备类似的方法合成标题化合物,其中用H-Ser(O tBu)-O tBu替换实施例29中的H-Thr(OtBu)-OtBu。 1H NMR(300MHz,D 2O)δ5.36(t,J=6.1Hz,1H),5.19–5.09(m,1H),4.75(s,1H),4.06(d,J=4.3Hz,1H),3.75(d,J=6.1Hz,2H),3.59(dd,J=12.6,3.9Hz,1H),3.41(d,J=12.8Hz,1H),3.03(t,J=6.6Hz,2H),2.58–2.35(m,2H).ESI-MS m/z:372.9[M+H] +. The title compound was synthesized in a similar manner to the preparation of Example 29, in which H-Thr(OtBu)-OtBu in Example 29 was replaced with H-Ser(O t Bu)-O t Bu. 1 H NMR (300 MHz, D 2 O) δ 5.36 (t, J = 6.1 Hz, 1H), 5.19 - 5.09 (m, 1H), 4.75 (s, 1H), 4.06 (d, J = 4.3 Hz, 1H) ), 3.75 (d, J = 6.1 Hz, 2H), 3.59 (dd, J = 12.6, 3.9 Hz, 1H), 3.41 (d, J = 12.8 Hz, 1H), 3.03 (t, J = 6.6 Hz, 2H) ), 2.58 - 2.35 (m, 2H). ESI-MS m/z: 372.9 [M+H] + .
实施例31 (S)-3-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-(3-((S)-2-酮基四氢呋喃-3-基)脲基)丙酰胺的制备Example 31 (S)-3-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-(3-(( Preparation of S)-2-ketotetrahydrofuran-3-yl)ureido)propanamide
Figure PCTCN2018084352-appb-000156
Figure PCTCN2018084352-appb-000156
步骤1:(S)-2-(叔丁氧羰基)氨基-3-叔丁氧基-1-丙酰胺的制备Step 1: Preparation of (S)-2-(tert-butoxycarbonyl)amino-3-tert-butoxy-1-propanamide
Figure PCTCN2018084352-appb-000157
Figure PCTCN2018084352-appb-000157
将L-Boc-Ser(tBu)-OH(50g,191.45mmol)溶于四氢呋喃(500mL),加入N-甲基吗啉(23.2g,229.74mmol),降温至-15℃后缓慢滴加氯甲酸乙酯(24.8g,229.74mmol),-15℃搅拌1.5h;向反应液中缓慢滴加氨水(48mL,693.48mmol),升温至-5℃反应2.5h,减压蒸馏除去四氢呋喃,剩余液体用乙酸乙酯和水溶清、分液,有机相依次用1M柠檬酸、饱和碳酸氢钠和饱和食盐水洗涤,无水硫酸钠干燥,减压蒸馏得到乳白色结晶,加少量乙酸乙酯和大量石油醚重结晶得39.3g白色固体4a(收率:78.9%)。L-Boc-Ser(tBu)-OH (50g, 191.45mmol) was dissolved in tetrahydrofuran (500mL), N-methylmorpholine (23.2g, 229.74mmol) was added, and the temperature was lowered to -15 ° C, then the chloroformic acid was slowly added dropwise. Ethyl ester (24.8 g, 229.74 mmol), stirred at -15 ° C for 1.5 h; slowly added ammonia water (48 mL, 693.48 mmol) to the reaction solution, and the mixture was heated to -5 ° C for 2.5 h, and the tetrahydrofuran was distilled off under reduced pressure. Ethyl acetate and water were lysed and separated, and the organic phase was washed with 1M citric acid, saturated sodium hydrogen carbonate and brine, dried over anhydrous sodium sulfate and evaporated. Recrystallization gave 39.3 g of a white solid 4a (yield: 78.9%).
步骤2:(R)-(2-(叔丁氧基)-1-氰乙基)氨基甲酸叔丁酯的制备Step 2: Preparation of (R)-(2-(tert-butoxy)-1-cyanoethyl)carbamic acid tert-butyl ester
Figure PCTCN2018084352-appb-000158
Figure PCTCN2018084352-appb-000158
将化合物4a(39g,149.81mmol)溶于吡啶(118.5g,1498.1mmol),冰浴降温,于-5℃下滴加三氟乙酸酐(47.2g,224.72mmol),反应40min,撤去冰浴,室温搅拌反应3.5h,将反应液倒入乙酸乙酯中,并依次用饱和碳酸氢钠、1M柠檬酸和饱和食盐水洗涤,无水硫酸钠干燥,减压蒸馏得37.36g淡黄色结晶4b(收率:102.9%)。The compound 4a (39 g, 149.81 mmol) was dissolved in pyridine (118.5 g, 1498.1 mmol), and the mixture was cooled in an ice bath, and trifluoroacetic anhydride (47.2 g, 224.72 mmol) was added dropwise at -5 ° C for 40 min, and the ice bath was removed. The reaction mixture was stirred at room temperature for 3.5 hr, and the mixture was evaporated, evaporated, evaporated, evaporated Yield: 102.9%).
步骤3:(R,Z)-(1-氨基-3-(叔丁氧基)-1-(肟基)丙烷-2-基)氨基甲酸叔丁酯的制备Step 3: Preparation of (R,Z)-(1-amino-3-(tert-butoxy)-1-(indolyl)propan-2-yl)carbamic acid tert-butyl ester
Figure PCTCN2018084352-appb-000159
Figure PCTCN2018084352-appb-000159
将化合物4b(23g,184.88mmol)溶于乙醇(450mL)中,依次加入碳酸钾(38.34g,277.33mmol)、盐酸羟胺(19.28g,277.33mmol),搅拌、回流反应1.5h,减压蒸馏除去溶剂,加乙酸乙酯和水溶解、分液,有机相用饱和食盐水洗涤数次,无水硫酸钠干燥,减压蒸馏,得黄色油状液体,静置冷却后凝结为淡黄色固体,加PE打浆,过滤,滤饼干燥得38.67g白色固体4c(收率:76.0%)。The compound 4b (23 g, 184.88 mmol) was dissolved in ethanol (450 mL), and then potassium carbonate (38.34 g, 277.33 mmol), hydroxylamine hydrochloride (19.28 g, 277.33 mmol) was added, stirred, refluxed for 1.5 h, and evaporated under reduced pressure. Solvent, add ethyl acetate and water to dissolve, liquid separation, the organic phase is washed several times with saturated brine, dried over anhydrous sodium sulfate, and evaporated under reduced pressure to give a yellow oily liquid, which is cooled and then condensed to a pale yellow solid. It was beaten, filtered, and the filter cake was dried to give 38.67 g of white solid 4c (yield: 76.0%).
步骤4:叔丁基-((5S,Z)-9-氨基-13,13-二甲基-3,6-二酮基-1,1,1-三苯基-7,12-二氧杂-2,8-氮杂-8-烯-5,10-二基)二氨基甲酸(9H-芴-9-基)甲基酯的制备Step 4: tert-Butyl-((5S,Z)-9-amino-13,13-dimethyl-3,6-dione-1,1,1-triphenyl-7,12-dioxo Preparation of (hetero-2,8-aza-8-ene-5,10-diyl)dicarbamic acid (9H-fluoren-9-yl)methyl ester
Figure PCTCN2018084352-appb-000160
Figure PCTCN2018084352-appb-000160
将Fmoc-Asn(Trt)-OH(10g,36.32mmol)和化合物4c溶于DMF(200mL),冰浴降温,于-5℃下加入HOBt(5.90g,43.58mmol),和EDC(8.35g,43.58mmol),冰浴搅拌反应20min,升温,室温反应4h,将反应液倒入水中,有大量固体析出,过滤,滤饼用水洗涤数次,干燥得33.8g白色固体4d(收率:108.98%)。Fmoc-Asn(Trt)-OH (10 g, 36.32 mmol) and compound 4c were dissolved in DMF (200 mL), cooled in an ice-bath, and then, at -5 °C, HOBt (5.90 g, 43.58 mmol), and EDC (8.35 g, 43.58mmol), stirring in an ice bath for 20min, heating, room temperature reaction for 4h, the reaction liquid was poured into water, a large amount of solids were precipitated, filtered, and the filter cake was washed several times with water, and dried to obtain 33.8 g of white solid 4d (yield: 108.98%) ).
步骤5:((R)-1-(5-((S)-1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-酮基-3-(三苯甲基氨基)丙基)-1,2,4-噁二唑-3-基)-2-(叔丁氧基)乙基)氨基甲酸叔丁酯的制备Step 5: ((R)-1-(5-((S)-1-(((9H-芴-9-yl)methoxy)carbonyl)))))))) Preparation of tert-butyl ester of benzylamino)propyl)-1,2,4-oxadiazol-3-yl)-2-(tert-butoxy)ethyl)carbamate
Figure PCTCN2018084352-appb-000161
Figure PCTCN2018084352-appb-000161
将化合物4d(28.0g,32.79mmol)溶于乙醇(420mL),加入乙酸钠(4.04g,49.19mmol)的水(60mL)溶液,搅拌回流反应2h,减压蒸馏除去反应溶剂,乙酸乙酯溶解,用饱和食盐水洗涤数次,减压蒸馏,用硅胶柱色谱纯化(洗脱剂:石油醚中0-30%的乙酸乙酯)。得6.6g乳白色固体4e(收率:24.1%)。The compound 4d (28.0 g, 32.79 mmol) was dissolved in ethanol (420 mL), and a solution of sodium acetate (4.04 g, 49.19 mmol) in water (60 mL) was added, and the mixture was stirred and refluxed for 2 hr. It was washed several times with saturated brine, distilled under reduced pressure, and purified by silica gel column chromatography (eluent: 0-30% ethyl acetate in petroleum ether). 6.6 g of milky white solid 4e was obtained (yield: 24.1%).
步骤6:((R)-1-(5-((S)-1-氨基-3-酮基-3-(三苯甲基氨基)丙基)-1,2,4-噁二唑-3-基)-2-(叔丁氧基)乙基)氨基甲酸叔丁酯的制备Step 6: ((R)-1-(5-((S)-1-Amino-3-keto-3-(tritylamino)propyl)-1,2,4-oxadiazole- Preparation of tert-butyl 3-yl)-2-(tert-butoxy)ethyl)carbamate
Figure PCTCN2018084352-appb-000162
Figure PCTCN2018084352-appb-000162
将化合物4e(6.6g,7.89mmol)加入到二氯甲烷(66mL)中,滴加二乙胺(33mL),室温搅拌反 应3.5h。减压蒸馏除去溶剂、用硅胶柱色谱纯化(洗脱剂:石油醚中0-50%的乙酸乙酯),得3.78g淡黄色结晶4f(收率:78.1%)。Compound 4e (6.6 g, 7.89 mmol) was added to dichloromethane (66 mL). The solvent was evaporated under reduced pressure and purified by silica gel column chromatography (eluent: EtOAc: EtOAc
步骤7:(S)-(2-酮基四氢呋喃-3-基)氨基甲酸4-硝基苯酯的制备Step 7: Preparation of (S)-(2-ketotetrahydrofuran-3-yl)carbamic acid 4-nitrophenyl ester
Figure PCTCN2018084352-appb-000163
Figure PCTCN2018084352-appb-000163
使用与实施例29中化合物2b的制备类似的方法合成标题化合物,其中用(S)-2-酮基-3-氨基四氢呋喃替换实施例29中的H-Thr(O tBu)-O tBu。 The title compound was synthesized in a similar manner to the preparation of the compound 2b in Example 29, in which the H-Thr(O t Bu)-O t Bu in Example 29 was replaced with (S)-2-keto-3-aminotetrahydrofuran. .
步骤8:((R)-2-(叔丁氧基)-1-(5-((S)-3-酮基-1-(3-((S)-2-酮基四氢呋喃-3-基)脲基)-3-(三苯甲基氨基)丙基)-1,2,4-噁二唑-3-基)乙基)氨基甲酸叔丁酯的制备Step 8: ((R)-2-(tert-Butoxy)-1-(5-((S)-3-keto-1-(3-((S)-2-ketotetrahydrofuran-3- Preparation of tert-butyl ester of ureido)-3-(tritylamino)propyl)-1,2,4-oxadiazol-3-yl)ethyl)carbamate
Figure PCTCN2018084352-appb-000164
Figure PCTCN2018084352-appb-000164
将化合物4f(500mg,0.815mmol)和4g溶于四氢呋喃(5mL),加入三乙胺(9.84g,97.26mmol),室温搅拌反应22h。减压蒸馏除去溶剂,加乙酸乙酯溶解,用饱和食盐水洗涤数次,无水硫酸钠干燥、减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-60%的乙酸乙酯)得400mg白色泡沫状固体4h(收率:66.2%)。Compound 4f (500 mg, 0.815 mmol) and 4 g were dissolved in tetrahydrofuran (5 mL), triethylamine (9.84 g, 97.26 mmol). The solvent was evaporated under reduced pressure, and ethyl acetate was evaporated, evaporated, evaporated, evaporated, evaporated, evaporated, The ester gave 400 mg of a white foamy solid for 4 h (yield: 66.2%).
步骤9:(S)-3-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-(3-((S)-2-酮基四氢呋喃-3-基)脲基)丙酰胺的制备Step 9: (S)-3-(3-((R)-1-Amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-(3-(( Preparation of S)-2-ketotetrahydrofuran-3-yl)ureido)propanamide
Figure PCTCN2018084352-appb-000165
Figure PCTCN2018084352-appb-000165
将化合物4h(280mg,0.378mmol)溶于二氯甲烷(7mL),室温下加入三氟乙酸(3.88g,34.04mmol)、催化量的三异丙基硅烷,室温搅拌反应2.5h,减压蒸馏除去溶剂、加二氯甲烷和水溶解、分液,有机相用水萃取数次,合并水相,再用二氯甲烷洗涤数次,水相冻干、制备液相纯化得化合物4。 1H NMR(500MHz,D 2O)δ6.00–5.94(m,1H),4.13–4.03(m,2H),3.75(tt,J=11.1,5.4Hz,2H),3.48(ddd,J=15.8,6.3,3.4Hz,1H),3.34(ddd,J=15.1,10.1,5.7Hz,1H),2.19–2.02(m,2H),1.96(s,2H).ESI-MS m/z:343.3[M+H] +. Compound 4h (280mg, 0.378mmol) was dissolved in dichloromethane (7mL), trifluoroacetic acid (3.88g, 34.04mmol) was added at room temperature, catalytic amount of triisopropylsilane, stirred at room temperature for 2.5h, distilled under reduced pressure The solvent was removed, dichloromethane and water were added to dissolve, and the organic phase was extracted several times with water, the aqueous phase was combined, and the mixture was washed several times with dichloromethane, and the aqueous phase was lyophilized to obtain a liquid crystal. 1 H NMR (500 MHz, D 2 O) δ 6.00 - 5.94 (m, 1H), 4.13 - 4.03 (m, 2H), 3.75 (tt, J = 11.1, 5.4 Hz, 2H), 3.48 (ddd, J = 15.8, 6.3, 3.4 Hz, 1H), 3.34 (ddd, J = 15.1, 10.1, 5.7 Hz, 1H), 2.19 - 2.02 (m, 2H), 1.96 (s, 2H). ESI-MS m/z: 343.3 [M+H] + .
实施例32 1-(3-((S)-3-氨基-1-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-丙氧基)脲基)环丙烷-1-甲酸的制备Example 32 1-(3-((S))-1-amino-1-(3-((R)-1-amino-2-hydroxyethyl)-1,2,4-oxadiazole-5- Preparation of benzyl-3-propyloxy)ureido)cyclopropane-1-carboxylic acid
Figure PCTCN2018084352-appb-000166
Figure PCTCN2018084352-appb-000166
步骤5a 1-(3-((S)-1-(3-((R)-2-(叔丁氧基)-1-((叔丁氧羰基)氨基)乙基)-1,2,4-噁二唑-5-基)-3-酮基-3-(三苯甲基氨基)丙基)脲基)环丙烷-1-甲酸甲酯的制备Step 5a 1-(3-((S)-1-(3-((R))-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)ethyl)-1,2, Preparation of methyl 4-oxadiazol-5-yl)-3-keto-3-(tritylamino)propyl)ureido)cyclopropane-1-carboxylate
Figure PCTCN2018084352-appb-000167
Figure PCTCN2018084352-appb-000167
使用与实施例31中化合物4的制备类似的方法合成标题化合物,其中用1-氨基环丙烷-1-甲酸甲酯盐酸盐替换实施例31步骤4i中的(S)-2-酮基-3-氨基四氢呋喃。The title compound was synthesized in a similar manner to the preparation of compound 4 in Example 31, which was replaced with 1-aminocyclopropane-1-carboxylic acid methyl ester hydrochloride to replace the (S)-2-keto group in the step 4i of Example 31. 3-aminotetrahydrofuran.
步骤5b:1-(3-((S)-1-(3-((R)-2-(叔丁氧基)-1-((叔丁氧羰基)氨基)乙基)-1,2,4-噁二唑-5-基)-3-酮基-3-(三苯甲基氨基)丙基)脲基)环丙烷-1-甲酸的制备Step 5b: 1-(3-((S)-1-(3-((R))-2-(tert-butoxy)-1-((tert-butoxycarbonyl)amino)ethyl)-1,2 Of 4-oxaoxazol-5-yl)-3-keto-3-(tritylamino)propyl)ureido)cyclopropane-1-carboxylic acid
Figure PCTCN2018084352-appb-000168
Figure PCTCN2018084352-appb-000168
将化合物5a(770mg,1.02mmol)溶于四氢呋喃(6mL),室温下加入一水合氢氧化锂(85mg,2.04mmol)的水(1.1mL)溶液,搅拌回流反应3h,反应液减压蒸馏除去四氢呋喃,加乙酸乙酯和水溶解、分液,乙酸乙酯层依次用1M柠檬酸、饱和食盐水洗涤数次,无水硫酸钠干燥,减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-50%的乙酸乙酯,二氯甲烷中0-10%的甲醇),得180mg白色固体5b(收率:23.8%)。The compound 5a (770 mg, 1.02 mmol) was dissolved in tetrahydrofuran (6 mL), and a solution of lithium hydroxide (85 mg, 2.04 mmol) in water (1.1 mL) was added at room temperature, and the mixture was stirred and refluxed for 3 hr. Ethyl acetate and water were added and dissolved, and the ethyl acetate layer was washed successively with 1M citric acid and brine, dried over anhydrous sodium sulfate, evaporated under reduced pressure and purified by silica gel column chromatography (eluent: petroleum 0-50% ethyl acetate in ether, 0-10% methanol in dichloromethane) gave 180 mg of white solid 5b (yield: 23.8%).
步骤5c:1-(3-((S)-3-氨基-1-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-丙氧基)脲基)环丙烷-1-甲酸的制备Step 5c: 1-(3-((S)-3-Amino-1-(3-((R)-1-amino-2-hydroxyethyl)-1,2,4-oxadiazole-5- Preparation of benzyl-3-propyloxy)ureido)cyclopropane-1-carboxylic acid
Figure PCTCN2018084352-appb-000169
Figure PCTCN2018084352-appb-000169
将化合物5b(180mg,0.243mmol)溶于二氯甲烷(3mL),室温下依次加入三氟乙酸(1.66g,14.58mmol)和催化量的三异丙基硅烷,搅拌反应4h,反应液减压蒸馏,加二氯甲烷和水溶解、分液,有机相用水萃取数次,合并水相,再用二氯甲烷洗涤数次,水相冻干、制备液相纯化得化合物32。 1H NMR(500MHz,D 2O)δ5.51–5.43(m,1H),4.17–4.08(m,2H),3.12(d,J=6.4Hz,2H),1.39(s,2H),1.07(s,2H).ESI-MS m/z:343.2[M+H] +. Compound 5b (180 mg, 0.243 mmol) was dissolved in dichloromethane (3 mL), trifluoroacetic acid (1.66 g, 14.58 mmol) and a catalytic amount of triisopropylsilane were added sequentially at room temperature, and the reaction was stirred for 4 h. Distillation, adding dichloromethane and water to dissolve, liquid separation, the organic phase is extracted several times with water, the aqueous phase is combined, and then washed several times with dichloromethane, the aqueous phase is lyophilized, and the liquid phase is purified to obtain compound 32. 1 H NMR (500 MHz, D 2 O) δ 5.51 - 5.43 (m, 1H), 4.17 - 4.08 (m, 2H), 3.12 (d, J = 6.4 Hz, 2H), 1.39 (s, 2H), 1.07 (s, 2H). ESI-MS m/z: 343.2 [M+H] + .
实施例33 1-(3-((R)-3-氨基-1-(3-((S)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-丙氧基)脲基)环丙烷-1-甲酸的制备Example 33 1-(3-((R)-3-Amino-1-(3-((S)-1-amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-) Preparation of benzyl-3-propyloxy)ureido)cyclopropane-1-carboxylic acid
Figure PCTCN2018084352-appb-000170
Figure PCTCN2018084352-appb-000170
使用与实施例32中化合物5的制备类似的方法合成标题化合物,其中用Boc-D-Ser(tBu)-OH替换实施例32中的Boc-L-Ser(tBu)-OH,用Fmoc-D-Asn(Trt)-OH替换实施例5中的Fmoc-L-Asn(Trt)-OH。粗品通过制备液相纯化得标题化合物。 1H NMR(400MHz,D 2O):δ5.30(t,J=6.1Hz,1H),4.59(s,1H),3.94(d,J=4.7Hz,2H),2.95(d,J=6.3Hz,2H),1.22(s,2H),0.90(s,2H).ESI-MS m/z:343.1[M+H] + The title compound was synthesized in a similar manner to the preparation of compound 5 in Example 32, in which Boc-L-Ser(tBu)-OH in Example 32 was replaced with Boc-D-Ser(tBu)-OH, using Fmoc-D -Asn(Trt)-OH replaces Fmoc-L-Asn(Trt)-OH in Example 5. The crude product was purified by preparative liquid to give the title compound. 1 H NMR (400 MHz, D 2 O): δ 5.30 (t, J = 6.1 Hz, 1H), 4.59 (s, 1H), 3.94 (d, J = 4.7 Hz, 2H), 2.95 (d, J = 6.3 Hz, 2H), 1.22 (s, 2H), 0.90 (s, 2H). ESI-MS m/z: 343.1 [M+H] +
实施例34 1-(3-((S)-1-(3-((R)-1-氨基-2-羟乙基)-1,2,4-噁二唑-5-基)-3-羧丙基)脲基)环丙烷-1-甲酸的制备Example 34 1-(3-((S)-1-(3-((R))-1-amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3 -Carboxypropyl)ureido)cyclopropane-1-carboxylic acid preparation
Figure PCTCN2018084352-appb-000171
Figure PCTCN2018084352-appb-000171
使用与实施例32中化合物5的制备类似的方法合成标题化合物,其中用Fmoc-L-Glu(O tBu)-OH替换实施例32中的Fmoc-L-Asn(Trt)-OH。粗品通过制备液相纯化得标题化合物。 1H NMR(300MHz,D 2O):δ5.02(s,1H),4.05(s,2H),2.30(d,J=7.5Hz,2H),2.19(dd,J=19.8,6.4Hz,2H),1.32(s,2H),0.99(s,2H).ESI-MS m/z:343.1[M+H] + Use Example Preparation of compound 5 The title compound was synthesized analogous to 32 in which Fmoc-L-Asn (Trt) in Example 32 using Fmoc-L-Glu (O t Bu) -OH alternative embodiment -OH. The crude product was purified by preparative liquid to give the title compound. 1 H NMR (300MHz, D 2 O): δ5.02 (s, 1H), 4.05 (s, 2H), 2.30 (d, J = 7.5Hz, 2H), 2.19 (dd, J = 19.8,6.4Hz, 2H), 1.32 (s, 2H), 0.99 (s, 2H). ESI-MS m/z: 343.1 [M+H] +
实施例35 (2S,3R)-2-(3-((S)-3-氨基-1-(3-(1-氨基-3-(羟基甲基)环丁基)-1,2,4-噁二唑-5-基)-3-氧代丙基)脲基)-3-羟基丁酸的制备Example 35 (2S,3R)-2-(3-((S)-3-Amino-1-(3-(1-amino-3-(hydroxymethyl)cyclobutyl)-1,2,4 -Oxadiazole-5-yl)-3-oxopropyl)ureido)-3-hydroxybutyric acid
Figure PCTCN2018084352-appb-000172
Figure PCTCN2018084352-appb-000172
步骤1 (S)-(1-氟-1,4-二酮基-4-(三苯甲基氨基)丁-2-基)氨基甲酸((9H-芴-9-基)甲基)酯的制备Step 1 (S)-(1-Fluoro-1,4-dione-4-(tritylamino)butan-2-yl)carbamic acid ((9H-fluoren-9-yl)methyl)ester Preparation
Figure PCTCN2018084352-appb-000173
Figure PCTCN2018084352-appb-000173
将Fmoc-Asn(Trt)-OH(5.0g,8.380mmol)溶于二氯甲烷(84mL),室温下缓慢加入DAST(1.62g,10.056mmol),室温搅拌反应15min,将反应液倒入冰水中淬灭,分液,二氯甲烷层用水洗涤数次,无水硫酸钠干燥,减压蒸馏,得淡黄色油状液体,加入少量二氯甲烷溶解,大量正己烷重结晶,过滤,滤饼用少量正己烷洗涤、减压蒸馏,得4.5g白色固体8i(收率:89.8%)。Fmoc-Asn(Trt)-OH (5.0 g, 8.380 mmol) was dissolved in dichloromethane (84 mL), DAST (1.62 g, 10.056 mmol) was slowly added at room temperature, the reaction was stirred at room temperature for 15 min, and the reaction solution was poured into ice water. Quenching, liquid separation, dichloromethane layer washed several times with water, dried over anhydrous sodium sulfate, and evaporated under reduced pressure to give a pale yellow oily liquid, dissolved in a small amount of dichloromethane, recrystallized from a large amount of n-hexane, filtered, filter cake with a small amount The mixture was washed with n-hexane and distilled under reduced pressure to give 4.5 g of white solid (yield: 89.8%).
步骤2:1-氨基-3-(羟甲基)环丁烷-1-甲酸甲酯的制备Step 2: Preparation of methyl 1-amino-3-(hydroxymethyl)cyclobutane-1-carboxylate
Figure PCTCN2018084352-appb-000174
Figure PCTCN2018084352-appb-000174
将化合物8a(4g,27.56mmol)溶于甲醇(40mL)中,冰浴降温,于0℃下缓慢滴加二氯亚砜(6.5g,55.13mmol),滴加完毕后加热至回流反应4h。减压蒸馏除去溶剂得淡黄色油状液体,直接进行下一步反应。The compound 8a (4 g, 27.56 mmol) was dissolved in methanol (40 mL), and the mixture was cooled in an ice bath, and thionyl chloride (6.5 g, 55.13 mmol) was slowly added dropwise at 0 ° C. After the addition was completed, the mixture was heated to reflux for 4 h. The solvent was distilled off under reduced pressure to give a pale yellow oily liquid, which was taken directly to the next step.
步骤3:1-((叔丁氧羰基)氨基)-3-(羟甲基)环丁烷-1-甲酸甲酯的制备Step 3: Preparation of methyl 1-((tert-butoxycarbonyl)amino)-3-(hydroxymethyl)cyclobutane-1-carboxylate
Figure PCTCN2018084352-appb-000175
Figure PCTCN2018084352-appb-000175
将化合物8b(4g,25.14mmol)溶于四氢呋喃(20mL)中,室温下加入碳酸氢钠(8.3g,88.00mmol)的水(40mL)溶液,冰浴降温,于0℃下加入二碳酸二叔丁酯(5.7g,23.88mmol)的四氢呋喃(20mL)溶液,室温搅拌反应过夜,减压蒸馏除去反应溶剂,用乙酸乙酯和水溶解、分液,有机相依次用水、饱和食盐水洗涤数次,减压蒸馏、用硅胶柱色谱纯化得3.7g白色固体8c(收率:56.9%)。Compound 8b (4g, 25.14mmol) was dissolved in tetrahydrofuran (20mL), and a solution of sodium hydrogencarbonate (8.3g, 88.00mmol) in water (40mL) was added at room temperature, cooled in ice bath, and added to the di-dicarbonate at 0 °C A solution of butyl ester (5.7 g, 23.88 mmol) in tetrahydrofuran (20 mL) was stirred at room temperature overnight. The reaction solvent was evaporated under reduced pressure, and the mixture was dissolved with ethyl acetate and water, and the organic phase was washed several times with water and brine. It was distilled under reduced pressure and purified by silica gel column chromatography to yield 3.7 g of white solid (yield: 56.9%).
步骤4:1-((叔丁氧羰基)氨基)-3-(叔丁基二甲基硅氧甲基)环丁烷-1-甲酸甲酯的制备Step 4: Preparation of methyl 1-((tert-butoxycarbonyl)amino)-3-(tert-butyldimethylsilyloxy)cyclobutane-1-carboxylate
Figure PCTCN2018084352-appb-000176
Figure PCTCN2018084352-appb-000176
将化合物8c(700mg,2.70mmol)溶于DMF(7mL),冰浴降温,于-5℃下依次加入咪唑(460mg,6.75mmol),叔丁基二甲基氯硅烷(610mg,4.05mmol)的DMF(3.5mL)溶液。室温搅拌反应4h,反应液加乙酸乙酯稀释,依次用1M柠檬酸、饱和碳酸氢钠、水洗涤数次,无水硫酸钠干燥,减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-50%的乙酸乙酯)得900mg无色油状液体8d(收率:89.3%)。Compound 8c (700 mg, 2.70 mmol) was dissolved in DMF (7 mL) EtOAc (EtOAc) (EtOAcjjjjjjjjjjjjjj DMF (3.5 mL) solution. The reaction mixture was stirred at room temperature for 4 h, then diluted with EtOAc EtOAc EtOAc. 0-50% ethyl acetate in the ether) gave 900 mg of a colorless oily liquid (yield: 89.3%).
步骤5:1-((叔丁氧羰基)氨基)-3-(叔丁基二甲基硅氧甲基)环丁烷-1-甲酸的制备Step 5: Preparation of 1-((tert-Butoxycarbonyl)amino)-3-(tert-butyldimethylsilyloxy)cyclobutane-1-carboxylic acid
Figure PCTCN2018084352-appb-000177
Figure PCTCN2018084352-appb-000177
将化合物8d(1.6g,4.283mmol)溶于甲醇(16mL),加入一水合氢氧化锂(270mg,6.425mmol)的水(3.2mL)溶液,室温搅拌反应3h,减压蒸馏除去反应溶剂,乙酸乙酯和水溶解、分液,有机相用水萃取数次,合并水相,用1M柠檬酸溶液调酸,再用乙酸乙酯萃取,合并有机相,无水Na 2SO 4干燥,减压蒸馏,得1.42g无色油状液体8e(收率:92.2%)。 The compound 8d (1.6 g, 4.283 mmol) was dissolved in methanol (16 mL), and a solution of lithium hydroxide monohydrate (270 mg, 6.425 mmol) in water (3.2 mL) was added, and the mixture was stirred at room temperature for 3 h, and the solvent was evaporated under reduced pressure. The ethyl ester and water were dissolved and separated, and the organic phase was extracted with water several times. The aqueous phase was combined, the acid was combined with 1M citric acid solution, and then extracted with ethyl acetate. The organic phase was combined, dried over anhydrous Na 2 SO 4 and evaporated. There was obtained 1.42 g of a colorless oily liquid 8e (yield: 92.2%).
步骤6:1-((叔丁氧羰基)氨基)-3-(叔丁基二甲基硅氧甲基)环丁烷-1-甲酰胺的制备Step 6: Preparation of 1-((tert-Butoxycarbonyl)amino)-3-(tert-butyldimethylsilyloxy)cyclobutane-1-carboxamide
Figure PCTCN2018084352-appb-000178
Figure PCTCN2018084352-appb-000178
将化合物8e(2.6g,7.231mmol)溶于四氢呋喃(26mL),加入N-甲基吗啉(880mg,8.677mmol),有絮状固体生成;冰浴降温,于-5℃下加入氯甲酸乙酯(940mg,8.677mmol),有大量固体析出,冰浴搅拌反应0.5h;缓慢加入3.9mL 25%的氨水溶液,冰浴搅拌反应3h,减压蒸馏除去溶剂,剩余固体用乙酸乙酯和水溶清、分液。有机相依次用1M柠檬酸、饱和碳酸氢钠、水洗涤数次,无水硫酸钠干燥、减压蒸馏,得1.5g白色固体8f(收率:57.9%)。Compound 8e (2.6 g, 7.231 mmol) was dissolved in tetrahydrofuran (26 mL), N-methylmorpholine (880 mg, 8.673 mmol) was added to form a flocculent solid; ice bath was cooled, and chloroformic acid B was added at -5 °C. Ester (940mg, 8.677mmol), a large amount of solid precipitated, stirred for 0.5h in an ice bath; 3.9mL of 25% aqueous ammonia solution was slowly added, stirred for 3 hours in an ice bath, the solvent was distilled off under reduced pressure, and the remaining solid was dissolved with ethyl acetate and water. Clear and liquid. The organic phase was washed successively with 1 M citric acid, saturated sodium hydrogen carbonate and water, and dried over anhydrous sodium sulfate and evaporated under reduced pressure to give 1.5 g of white solid (yield: 57.9%).
步骤7:(3-叔丁基二甲基硅氧甲基-1-氰基环丁基)氨基甲酸叔丁酯的制备Step 7: Preparation of tert-butyl (3-tert-butyldimethylsilylmethyl-1-cyanocyclobutyl)carbamate
Figure PCTCN2018084352-appb-000179
Figure PCTCN2018084352-appb-000179
将化合物8f(1.58g,4.407mmol)溶于吡啶(16mL),冰浴降温,于-5℃下缓慢加入三氟乙酸酐(1.39g,6.611mmol),冰浴搅拌反应3h,将反应液倒入过量饱和碳酸氢钠溶液中淬灭,用乙酸乙酯萃取,有机相依次用1M柠檬酸、饱和食盐水洗涤数次,无水硫酸钠干燥,减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-15%的乙酸乙酯)得1.36g无色油状液体8g(收率:90.7%)。The compound 8f (1.58g, 4.407mmol) was dissolved in pyridine (16mL), cooled in an ice bath, trifluoroacetic anhydride (1.39g, 6.611mmol) was slowly added at -5 °C, and the reaction was stirred for 3 hours in an ice bath, and the reaction solution was poured. The mixture was quenched with EtOAc EtOAc (EtOAc)EtOAc. Agent: 0-15% ethyl acetate in petroleum ether) 1.36 g of a colorless oily liquid 8 g (yield: 90.7%).
步骤8:(Z)-(3-叔丁基二甲基硅氧甲基-1-(N'-羟基甲脒基)环丁基)氨基甲酸叔丁酯的制备Step 8: Preparation of (Z)-(3-tert-butyldimethylsilylmethyl-1-(N'-hydroxymethylindenyl)cyclobutyl)carbamic acid tert-butyl ester
Figure PCTCN2018084352-appb-000180
Figure PCTCN2018084352-appb-000180
将化合物8g(1.3g,3.817mmol)溶于乙醇(13mL),室温下依次加入碳酸钾(791mg,5.726mmol)的水(2.6mL)溶液、盐酸羟胺(398mg,5.726mmol),搅拌回流反应2h,减压蒸馏除去溶剂,加乙酸乙酯和水溶清、分液,有机相用饱和食盐水洗涤数次,无水硫酸钠干燥,减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-20%的乙酸乙酯)得700mg白色固体8h(收率:49.3%)。The compound 8g (1.3g, 3.818mmol) was dissolved in ethanol (13mL), and a solution of potassium carbonate (791mg, 5.726mmol) in water (2.6mL), hydroxylamine hydrochloride (398mg, 5.726mmol) The solvent was evaporated under reduced pressure, and the residue was evaporated. In the range of 0-20% ethyl acetate, 700 mg of white solid was obtained (yield: 49.3%).
步骤9:(Z)-(1-(N'-((N 2-(((9H-芴-9-基)甲氧基)羰基)-N 4-三苯甲基-L-天冬酰胺基)氧基)羟基甲脒基)-3-(叔丁基二甲基硅氧甲基)环丁基)氨基甲酸叔丁酯的制备 Step 9: (Z)-(1-(N'-((N 2 -((9H-芴-9-yl)methoxy)carbonyl)-N 4 -trityl-L-asparagine Preparation of tert-butyl ester of hydroxy)hydroxymethylindenyl)-3-(tert-butyldimethylsilylmethyl)cyclobutylcarbamate
Figure PCTCN2018084352-appb-000181
Figure PCTCN2018084352-appb-000181
将化合物8h(300mg,0.803mmol)溶于四氢呋喃(3mL)。室温下依次加入N-甲基吗啉(240mg,2.409mmol)和化合物8i(960mg,1.606mmol)的四氢呋喃(10mL)溶液,室温搅拌反应过夜,反应 液减压蒸馏除去四氢呋喃,加乙酸乙酯溶清,用饱和食盐水洗涤数次,无水硫酸钠干燥,减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-15%的乙酸乙酯)得350mg无色油状物8j(收率:55.8%)。Compound 8h (300 mg, 0.803 mmol) was dissolved in tetrahydrofuran (3 mL). A solution of N-methylmorpholine (240 mg, 2.409 mmol) and compound 8i (960 mg, 1.606 mmol) in tetrahydrofuran (10 mL) was added, and the mixture was stirred at room temperature overnight. The mixture was washed with saturated brine for several times, dried over anhydrous sodium sulfate and evaporated. Yield: 55.8%).
步骤10:(S)-(1-(5-(1-((((9H-芴-9-基)甲氧基)羰基)氨基)-3-酮基-3-(三苯甲基氨基)丙基)-1,2,4-噁二唑-3-基)-3-(((叔丁基二甲基硅基)氧基)甲基)环丁基)氨基甲酸叔丁酯的制备Step 10: (S)-(1-(5-(1-(((9H-芴-9-yl))methoxy)carbonyl)amino)-3-yl-3-yl-3-triphenylmethylamino Of propyl)-1,2,4-oxadiazol-3-yl)-3-(((tert-butyldimethylsilyl)oxy)methyl)cyclobutyl)carbamic acid tert-butyl ester preparation
Figure PCTCN2018084352-appb-000182
Figure PCTCN2018084352-appb-000182
将化合物8j(1g,1.050mmol)溶于 tBuOH(20mL)。室温下加入乙酸钠(129mg,1.575mmol)、搅拌回流反应3h,反应液减压蒸馏除去溶剂,加乙酸乙酯溶清,用饱和食盐水洗涤数次,无水硫酸钠干燥,减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-20%的乙酸乙酯)得520mg无色油状物8k(收率:53%)。 Compound 8j (1g, 1.050mmol) was dissolved in t BuOH (20mL). Sodium acetate (129 mg, 1.575 mmol) was added at room temperature, and the mixture was stirred and refluxed for 3 hr. The solvent was evaporated evaporated vacuo. Purification by silica gel column chromatography (EtOAc:EtOAc:EtOAc
步骤11:(S)-(1-(5-(1-氨基-3-酮基-3-(三苯甲基氨基)丙基)-1,2,4-噁二唑-3-基)-3-(((叔丁基二甲基硅基)氧基)甲基)环丁基)氨基甲酸叔丁酯的制备Step 11: (S)-(1-(5-(1-Amino-3-keto-3-(tritylamino)propyl)-1,2,4-oxadiazol-3-yl) Preparation of tert-butyl 3-(((tert-butyldimethylsilyl)oxy)methyl)cyclobutyl)carbamate
Figure PCTCN2018084352-appb-000183
Figure PCTCN2018084352-appb-000183
将化合物8k(500mg,0.535mmol)溶于二氯甲烷(5mL),室温下加入二乙胺(783mg,10.7mmol)、室温搅拌反应4h。反应的完成通过TLC分析确认。反应液减压蒸馏除去溶剂,用硅胶柱色谱纯化(洗脱剂:石油醚中0-40%的乙酸乙酯),得330mg无色油状物8l(收率:86.6%)。Compound 8k (500 mg, 0.535 mmol) was dissolved in dichloromethane (5 mL), and diethylamine (783 mg, 10.7 mmol) was added at room temperature, and the mixture was stirred at room temperature for 4 h. The completion of the reaction was confirmed by TLC analysis. The solvent was evaporated under reduced pressure and purified (EtOAcjjjjjjjjjjj
步骤12:N-(((S)-1-(3-(1-((叔丁氧羰基)氨基)-3-(((叔丁基二甲基硅基)氧基)甲基)环丁基)-1,2,4-噁二唑-5-基)-3-酮基-3-(三苯甲基氨基)丙基)氨甲酰基)-O-(叔丁基)-L-苏氨酸叔丁酯的制备Step 12: N-(((S)-1-(3-(1-((tert-Butyloxycarbonyl))amino)-3-(((tert-butyldimethylsilyl)oxy)methyl)) Butyl)-1,2,4-oxadiazol-5-yl)-3-keto-3-(tritylamino)propyl)carbamoyl)-O-(tert-butyl)-L -Preparation of threonine tert-butyl ester
Figure PCTCN2018084352-appb-000184
Figure PCTCN2018084352-appb-000184
将化合物8l(270mg,0.379mmol)溶于四氢呋喃(5mL),室温下依次加入化合物2b(180mg,0.455mmol)、三乙胺(77mg,0.758mmol)、室温搅拌反应过夜,反应液减压蒸馏除去溶剂,加乙酸乙酯溶清,依次用1M柠檬酸、饱和食盐水洗涤数次,无水硫酸钠干燥,减压蒸馏、用硅胶柱色谱纯化(洗脱剂:石油醚中0-15%的乙酸乙酯),得400mg白色固体8n(收率:95.1%)。The compound 8l (270 mg, 0.379 mmol) was dissolved in tetrahydrofuran (5 mL), and the compound 2b (180 mg, 0.45 mmol), triethylamine (77 mg, 0.758 mmol) was added, and the mixture was stirred at room temperature overnight. The solvent was dissolved in ethyl acetate, and washed successively with 1M citric acid and brine, dried over anhydrous sodium sulfate, and evaporated under reduced pressure and purified by silica gel column chromatography (eluent: 0-15% in petroleum ether) Ethyl acetate) gave 400 mg of white solid (yield: 95.1%).
步骤13:(2S,3R)-2-(3-((S)-3-氨基-1-(3-(1-氨基-3-(羟基甲基)环丁基)-1,2,4-噁二唑-5-基)-3- 氧代丙基)脲基)-3-羟基丁酸的制备Step 13: (2S,3R)-2-(3-((S)-3-Amino-1-(3-(1-amino-3-(hydroxymethyl)cyclobutyl)-1,2,4 -Oxadiazole-5-yl)-3-oxopropyl)ureido)-3-hydroxybutyric acid
Figure PCTCN2018084352-appb-000185
Figure PCTCN2018084352-appb-000185
将化合物8n(350mg,0.361mmol)溶于二氯甲烷(7mL),室温下加入三氟乙酸(2.06g,18.05mmol)和催化量的三异丙基硅烷,室温搅拌反应5h,反应液减压蒸馏除去溶剂,加二氯甲烷和水溶清、分液,有机相用水萃取数次,合并水相,再用二氯甲烷洗涤数次,水相冻干,制备液相纯化,得70mg白色固体8(收率:37.8%)。 1H NMR(400MHz,D 2O)δ5.31(dd,J=8.2,4.7Hz,1H),4.16–4.10(m,1H),3.89(d,J=3.0Hz,1H),3.61(d,J=6.8Hz,2H),2.99(d,J=6.4Hz,2H),2.77(dt,J=16.0,7.9Hz,1H),2.59–2.47(m,4H),1.08(d,J=6.4Hz,3H).ESI-MS m/z:401.2[M+H] +. Compound 8n (350 mg, 0.361 mmol) was dissolved in dichloromethane (7 mL), trifluoroacetic acid (2.06 g, 18.05 mmol) and a catalytic amount of triisopropylsilane were added at room temperature, and the reaction was stirred at room temperature for 5 h. The solvent was distilled off, dichloromethane and water were added to dissolve, and the organic phase was extracted several times with water, the aqueous phase was combined, and the mixture was washed several times with dichloromethane, and the aqueous phase was lyophilized to obtain 70 mg of white solid. (Yield: 37.8%). 1 H NMR (400 MHz, D 2 O) δ 5.31 (dd, J = 8.2, 4.7 Hz, 1H), 4.16 - 4.10 (m, 1H), 3.89 (d, J = 3.0 Hz, 1H), 3.61 (d) , J = 6.8 Hz, 2H), 2.99 (d, J = 6.4 Hz, 2H), 2.77 (dt, J = 16.0, 7.9 Hz, 1H), 2.59 - 2.47 (m, 4H), 1.08 (d, J = 6.4 Hz, 3H). ESI-MS m/z: 401.2 [M+H] + .
实施例36:(S)-4-(3-(1-氨基-3-(羟甲基)环丁基)-1,2,4-噁二唑-5-基)-4-(3-((1S,2R)-1-羧基-2-羟丙基)脲基)丁酸的制备Example 36: (S)-4-(3-(1-Amino-3-(hydroxymethyl)cyclobutyl)-1,2,4-oxadiazol-5-yl)-4-(3- Preparation of ((1S,2R)-1-carboxy-2-hydroxypropyl)ureido)butyric acid
Figure PCTCN2018084352-appb-000186
Figure PCTCN2018084352-appb-000186
使用与实施例35中化合物的制备相类似的方法合成标题化合物。其中用Fmoc-Glu(OtBu)-OH替换Fmoc-Asn(Trt)-OH。 1H NMR(400MHz,D 2O)δ5.02–4.97(m,1H),4.25(dd,J=6.3,2.8Hz,1H),4.15(d,J=2.6Hz,1H),3.60(d,J=6.7Hz,2H),2.76(dd,J=15.4,8.3Hz,1H),2.60–2.44(m,6H),2.24(dd,J=13.5,6.4Hz,1H),2.10(dd,J=13.9,6.8Hz,1H),1.34(s,1H),1.08(d,J=6.3Hz,3H).ESI-MS m/z:416.2[M+H] +. The title compound was synthesized in a similar manner to the preparation of the compound in Example 35. Wherein Fmoc-Asn(Trt)-OH was replaced with Fmoc-Glu(OtBu)-OH. 1 H NMR (400 MHz, D 2 O) δ 5.02 - 4.97 (m, 1H), 4.25 (dd, J = 6.3, 2.8 Hz, 1H), 4.15 (d, J = 2.6 Hz, 1H), 3.60 (d) , J = 6.7 Hz, 2H), 2.76 (dd, J = 15.4, 8.3 Hz, 1H), 2.60 - 2.44 (m, 6H), 2.24 (dd, J = 13.5, 6.4 Hz, 1H), 2.10 (dd, J = 13.9, 6.8 Hz, 1H), 1.34 (s, 1H), 1.08 (d, J = 6.3 Hz, 3H). ESI-MS m/z: 416.2 [M+H] + .
实施例37:(S)-3-(3-(1-氨基-3-(羟甲基)环丁基)-1,2,4-噁二唑-5-基)-3-(3-((S)-1-羧基-2-羟乙基)脲基)丙酸的制备Example 37: (S)-3-(3-(1-Amino-3-(hydroxymethyl)cyclobutyl)-1,2,4-oxadiazol-5-yl)-3-(3- Preparation of ((S)-1-carboxy-2-hydroxyethyl)ureido)propionic acid
Figure PCTCN2018084352-appb-000187
Figure PCTCN2018084352-appb-000187
使用与实施例35中化合物的制备相类似的方法合成标题化合物。其中用Fmoc-Asp(OtBu)-OH替换实施例8中的Fmoc-Asn(Trt)-OH,用H-Ser(O tBu)-O tBu替换实施例35中的H-Thr(OtBu)-OtBu。 1H NMR(400MHz,D 2O)δ5.35(t,J=6.0Hz,1H),4.28(t,J=4.0Hz,1H),3.86(dd,J=11.7,4.6Hz,1H),3.76(dd,J=11.7,3.6Hz,1H),3.61(d,J=6.7Hz,2H),3.13–3.08(m,2H),2.78(dt,J=15.8,8.0Hz,1H),2.56(p,J=13.8Hz,4H).ESI-MS m/z:388.1[M+H] +. The title compound was synthesized in a similar manner to the preparation of the compound in Example 35. Wherein Fmoc-Asn(Trt)-OH in Example 8 was replaced with Fmoc-Asp(OtBu)-OH, and H-Thr (OtBu) in Example 35 was replaced with H-Ser(O t Bu)-O t Bu -OtBu. 1 H NMR (400 MHz, D 2 O) δ 5.35 (t, J = 6.0 Hz, 1H), 4.28 (t, J = 4.0 Hz, 1H), 3.86 (dd, J = 11.7, 4.6 Hz, 1H), 3.76 (dd, J = 11.7, 3.6 Hz, 1H), 3.61 (d, J = 6.7 Hz, 2H), 3.13 - 3.08 (m, 2H), 2.78 (dt, J = 15.8, 8.0 Hz, 1H), 2.56 (p, J = 13.8 Hz, 4H). ESI-MS m/z: 388.1 [M+H] + .
实施例38 (2S,3S)-2-(3-((S)-4-氨基-1-(5-(1-氨基环丙基)-1,3,4-噁二唑-2-基)-4-氧代丁基)脲基)-3-羟基丁酸的制备Example 38 (2S,3S)-2-(3-((S)-4-Amino-1-(5-(1-aminocyclopropyl)-1,3,4-oxadiazol-2-yl) Preparation of 4-oxobutyl)ureido)-3-hydroxybutyric acid
Figure PCTCN2018084352-appb-000188
Figure PCTCN2018084352-appb-000188
制备方法同实施例12的制备方法,将实施例12步骤1中的(2S,3S)-3-羟基四氢吡咯-2-甲酸替换成了1-氨基环丙烷-1-甲酸,以及将O-(叔丁基)-L-丝氨酸叔丁酯替换成O-(叔丁基)-L-苏氨酸叔丁酯,可制得目标化合物。Preparation method The same as the preparation method of Example 12, the (2S,3S)-3-hydroxytetrahydropyrrole-2-carboxylic acid in the step 1 of Example 12 was replaced with 1-aminocyclopropane-1-carboxylic acid, and O was The (t-butyl)-L-serine tert-butyl ester is replaced with O-(tert-butyl)-L-threonine tert-butyl ester to obtain the target compound.
比较例1Comparative example 1
根据WO2015/033301(PCT/IB2014/064281)中实施例2公开的方法制备下式代表的化合物(化合物A),并通过氢谱和质谱鉴定,The compound represented by the following formula (Compound A) was prepared according to the method disclosed in Example 2 of WO2015/033301 (PCT/IB2014/064281), and identified by hydrogen spectrum and mass spectrometry,
Figure PCTCN2018084352-appb-000189
Figure PCTCN2018084352-appb-000189
使用以下实验例1和2的方法测试了化合物A的药代动力学特征以及在结肠癌CT26细胞皮下移植瘤模型上的抑瘤效果,实验结果显示,化合物A的生物利用度(F)和抑瘤率弱于本发明的一些化合物。The pharmacokinetic characteristics of Compound A and the antitumor effect on the subcutaneous xenograft model of colon cancer CT26 cells were tested using the following Experimental Examples 1 and 2, and the results showed that the bioavailability (F) and inhibition of Compound A were observed. The tumor rate is weaker than some of the compounds of the invention.
另外,本发明的发明人还根据WO2015/033301公开的方法合成并测试了WO2015/033301表3中Compound No.12,结果显示,Compound No.12的生物利用度和抑瘤率明显弱于本发明的化合物及化合物A。In addition, the inventors of the present invention also synthesized and tested Compound No. 12 in Table 3 of WO2015/033301 according to the method disclosed in WO2015/033301, and the results show that the bioavailability and tumor inhibition rate of Compound No. 12 are significantly weaker than the present invention. Compound and Compound A.
实验例1药物代谢实验Experimental Example 1 Drug Metabolism Experiment
1实验材料1 experimental material
1.1化合物1.1 compound
使用以上实施例制备的本发明的化合物和WO2015/033301中实施例2的化合物(“化合物A”)进行该实验。口服药物用生理盐水溶解,制成0.5mg/mL澄清溶液,静脉药物用生理盐水溶解,制成0.1mg/mL澄清溶液。The experiment was carried out using the compound of the present invention prepared in the above examples and the compound of Example 2 ("Compound A") in WO2015/033301. The oral drug was dissolved in physiological saline to prepare a clear solution of 0.5 mg/mL, and the intravenous drug was dissolved in physiological saline to prepare a 0.1 mg/mL clear solution.
1.2动物1.2 animals
雄性BALB/c小鼠,每组各3只,体重18-22g,上海西普尔-必凯实验动物有限公司提供。Male BALB/c mice, 3 in each group, weighing 18-22 g, were provided by Shanghai Xipuer-Beikai Experimental Animal Co., Ltd.
受试小鼠实验前给予2~4天的环境适应期,给药前禁食8-12h,给药2h后给水,4h后给食。The test mice were given an environmental adaptation period of 2 to 4 days before the experiment, and were fasted for 8-12 hours before administration, and given water for 2 hours after administration, and fed after 4 hours.
1.3试剂1.3 reagent
甲醇(色谱纯):Spectrum公司生产;Methanol (chromatographically pure): produced by Spectrum;
乙腈(色谱纯):Spectrum公司生产;Acetonitrile (chromatographically pure): produced by Spectrum;
其余试剂均为市售分析纯。The remaining reagents were all commercially available analytical grades.
1.4仪器1.4 Instrument
美国AB公司API 4500型三重四级杆液质联用仪,配有电喷雾离子源(ESI),LC-30AD双泵;SIL-30AC自动进样器;CTO-30AC柱温箱;DGU-20A3R脱气机;Analyst QS A01.01色谱工作站;Milli-Q超纯水器(Millipore Inc);Qilinbeier Vortex-5振荡器;HITACHI CF16R Ⅹ Ⅱ台式高速冷冻离心机。American AB company API 4500 triple quadrupole liquid-mass instrument with electrospray ion source (ESI), LC-30AD double pump; SIL-30AC autosampler; CTO-30AC column thermostat; DGU-20A3R Degasser; Analyst QS A01.01 chromatography workstation; Milli-Q ultrapure water (Millipore Inc); Qilinbeier Vortex-5 oscillator; HITACHI CF16R X II benchtop high speed refrigerated centrifuge.
2实验方法2 experimental methods
1)小鼠禁食但可自由饮水12小时后,采取0时刻空白血浆;1) The mice were fasted but allowed to drink water for 12 hours, and the blank plasma was taken at 0 o'clock;
2)取步骤1)中的小鼠,灌胃(intragastric administration,IG)给予待测化合物10mg/kg;静脉(IV)给予待测化合物1mg/kg;2) taking the mouse in step 1), administering intragastric administration (IG) to the test compound 10 mg/kg; intravenous (IV) administering the test compound 1 mg/kg;
3)于灌胃后5min,15min,30min,1h,2h,4h,8h,10h,24h,从眼底静脉丛连续取血置于分布有肝素的EP管中,8000rpm/min离心5min后取上层血浆,-20℃冻存,待LC-MS/MS分析;3) 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 8 h, 10 h, 24 h after intragastric administration, continuous blood was taken from the fundus venous plexus in an EP tube with heparin distributed, and centrifuged at 8000 rpm/min for 5 min. , frozen at -20 ° C, to be analyzed by LC-MS/MS;
4)根据步骤3)所得的血药浓度-时间数据,采用WinNonlin软件求算药代动力学参数;4) Calculate the pharmacokinetic parameters using WinNonlin software according to the plasma concentration-time data obtained in step 3);
3实验结果:3 experimental results:
药代动力学实验数据如表1中所示,结果表明口服给予小鼠以上实施例的化合物后,在动物血浆中皆有一定的暴露量和适宜的半衰期,尤其是实施例4、实施例5、实施例8、实施例6和实施例22的化合物,具有非常好的半衰期、曲线下面积以及生物利用度,成药性好,具有良好的临床应用前景。The pharmacokinetic experimental data are shown in Table 1. The results indicate that after oral administration of the compound of the above examples, there is a certain amount of exposure and a suitable half-life in the plasma of the animal, especially Example 4, Example 5 The compounds of Example 8, Example 6 and Example 22 have very good half-life, area under the curve and bioavailability, have good drug-forming properties, and have good clinical application prospects.
表1本发明实施例化合物的药代动力学数据Table 1 Pharmacokinetic data of the compounds of the examples of the invention
Figure PCTCN2018084352-appb-000190
Figure PCTCN2018084352-appb-000190
Figure PCTCN2018084352-appb-000191
Figure PCTCN2018084352-appb-000191
实验例2体内药效实验Experimental Example 2 In vivo pharmacodynamic experiment
1、实验材料1. Experimental materials
1.1化合物1.1 compound
使用根据本发明实施例制备的化合物进行该实验。阴性对照组给予生理盐水。采用口服给药,测试化合物用生理盐水溶解,制成2mg/mL澄清溶液。This experiment was carried out using the compounds prepared according to the examples of the present invention. The negative control group was given physiological saline. Orally administered, the test compound was dissolved in physiological saline to prepare a 2 mg/mL clear solution.
1.2动物1.2 animals
雌性BALB/c小鼠,每组各3只,体重18-22g,上海西普尔-必凯实验动物有限公司提供。受试小鼠实验前给予2~4天的环境适应期,给药前禁食8-12h,给药2h后给水,4h后给食。Female BALB/c mice, 3 in each group, weighing 18-22 g, were provided by Shanghai Xipuer-Beikai Experimental Animal Co., Ltd. The test mice were given an environmental adaptation period of 2 to 4 days before the experiment, and were fasted for 8-12 hours before administration, and given water for 2 hours after administration, and fed after 4 hours.
1.3阳性对照1.3 positive control
化合物A,根据WO2015/033301实施例2公开的方法制备。Compound A was prepared according to the method disclosed in Example 2 of WO 2015/033301.
2、实验方法2, experimental methods
接种细胞后待肿瘤生长至平均体积为40mm 3(第一轮实验)或50mm 3(第二轮实验)后,将动物随机分组,每组6只,各测试组口服给药20mg/kg,一天一次,连续给药14天。考察实验动物体重的变化及肿瘤生长是否被抑制或延缓。每周三次用游标卡尺测量肿瘤直径。 After the cells were inoculated and the tumors were grown to an average volume of 40 mm 3 (first round of experiments) or 50 mm 3 (second round of experiments), the animals were randomly divided into groups of 6 each, and each test group was orally administered with 20 mg/kg, one day. Once, continuous administration for 14 days. The changes in body weight of the experimental animals and whether the tumor growth was inhibited or delayed were examined. Tumor diameters were measured with vernier calipers three times a week.
肿瘤体积的计算公式为:V=0.5a×b 2,a和b分别表示肿瘤的长径和短径。 The calculation formula of tumor volume is: V = 0.5a × b 2 , and a and b represent the long diameter and short diameter of the tumor, respectively.
相对肿瘤增殖率T/C(%)的计算公式为:T/C=T RTV/C RTV X 100%(T RTV:治疗组RTV;C RTV:溶剂对照组RTV)。根据测量结果计算出相对肿瘤体积(relative tumor volume,RTV),RTV=V t/V 0,其中V 0为实验开始时的肿瘤体积,Vt每一次测量时的肿瘤体积。 The relative tumor growth rate T/C (%) was calculated as: T/C = T RTV / C RTV X 100% (T RTV : treatment group RTV; C RTV : solvent control group RTV). Relative tumor volume (RTV) was calculated based on the measurement results, RTV = V t /V 0 , where V 0 is the tumor volume at the start of the experiment, and the tumor volume at each measurement of Vt.
肿瘤生长抑制率TGI(%)的计算公式为:TGI=[1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]X 100%。The tumor growth inhibition rate TGI (%) was calculated as: TGI = [1 - (the average tumor volume at the end of a treatment group - the average tumor volume at the start of administration of the treatment group) / (the average of the solvent control group at the end of treatment) Tumor volume - mean tumor volume at the start of treatment in the solvent control group)] X 100%.
3、实验结论3. Experimental conclusion
3.1体重变化情况3.1 Weight changes
本发明的化合物对小鼠结肠癌CT26细胞皮下同系移植瘤在BALB/C小鼠模型的体重无影响,表2给出了实施例4、实施例5、实施例35以及化合物A给药后对体重的影响,表明对照组及各给药组动物体重在给药期间逐渐增加,具有较好的耐受性。The compound of the present invention has no effect on the body weight of the mouse colon cancer CT26 cell subcutaneous homologous tumor in the BALB/C mouse model. Table 2 shows the administration of Example 4, Example 5, Example 35 and Compound A. The effect of body weight indicated that the body weight of the control group and each drug-administered group gradually increased during the administration period and was well tolerated.
表2本发明的化合物给药后对小鼠体重的影响Table 2 Effect of the compound of the present invention on the body weight of mice after administration
Figure PCTCN2018084352-appb-000192
Figure PCTCN2018084352-appb-000192
3.2抗肿瘤药效评价指标3.2 Anti-tumor efficacy evaluation indicators
药效评价指标如表3所示,第一轮实验开始给药后第15天,溶剂对照组荷瘤鼠的平均瘤体积达到3672mm 3,其他各给药组荷瘤鼠的瘤体积平均值均小于对照组瘤体积平均值,其中实施例4的化合物在第15天时的T/C值为41.5%,TGI值为52.4%,表示其对CT26结肠癌细胞移植瘤具有显著的抑制作用,效果明显优于化合物A。第二轮实验开始给药后第14天,溶剂对照组荷瘤鼠的平均瘤体积达到1524mm 3,其他各受试物组荷瘤鼠的瘤体积平均值均小于对照组瘤 体积平均值,其中实施例5的化合物在第14天时的T/C值为39.0%,TGI值为63.7%,表示其对CT26结肠癌细胞移植瘤具有显著的抑制作用。 The evaluation index of drug efficacy is shown in Table 3. On the 15th day after the first round of experiment started, the average tumor volume of the tumor-bearing mice in the solvent control group reached 3672 mm 3 , and the average tumor volume of the tumor-bearing mice in the other drug-administered groups was average. It is smaller than the average tumor volume of the control group, wherein the compound of Example 4 has a T/C value of 41.5% on the 15th day and a TGI value of 52.4%, indicating that it has a significant inhibitory effect on CT26 colon cancer xenografts, and the effect is obvious. Better than compound A. On the 14th day after the start of the second round of experiment, the average tumor volume of the tumor-bearing mice in the solvent control group reached 1524 mm 3 , and the average tumor volume of the tumor-bearing mice in each of the other test groups was smaller than the average tumor volume of the control group. The compound of Example 5 had a T/C value of 39.0% at day 14 and a TGI value of 63.7%, indicating that it had a significant inhibitory effect on CT26 colon cancer xenografts.
表3抗肿瘤药效评价指标Table 3 anti-tumor efficacy evaluation indicators
Figure PCTCN2018084352-appb-000193
Figure PCTCN2018084352-appb-000193
尽管以上已经对本发明作了详细描述,但是本领域技术人员理解,在不偏离本发明的精神和范围的前提下可以对本发明进行各种修改和改变。本发明的权利范围并不限于上文所作的详细描述,而应归属于权利要求书。While the invention has been described hereinabove, it will be understood by those skilled in the art that various modifications and changes of the invention may be made without departing from the spirit and scope of the invention. The scope of the invention is not limited to the details described above, but rather to the claims.

Claims (10)

  1. 一种通式A所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,a compound of the formula A or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug,
    Figure PCTCN2018084352-appb-100001
    Figure PCTCN2018084352-appb-100001
    其中:among them:
    X和Y各自独立地选自N、O和S,且X为N时,Y选自O和S;X为O或S时,Y为N;Y为N时,X选自O和S;Y为O或S时,X为N;X and Y are each independently selected from N, O and S, and when X is N, Y is selected from O and S; when X is O or S, Y is N; when Y is N, X is selected from O and S; When Y is O or S, X is N;
    当X为N,Y选自O和S时或Y为O或S,X为N时,基团
    Figure PCTCN2018084352-appb-100002
    Figure PCTCN2018084352-appb-100003
    当X为O或S,Y为N时或Y为N,X选自O和S时,基团
    Figure PCTCN2018084352-appb-100004
    Figure PCTCN2018084352-appb-100005
    When X is N, Y is selected from O and S or Y is O or S, and X is N, the group
    Figure PCTCN2018084352-appb-100002
    for
    Figure PCTCN2018084352-appb-100003
    When X is O or S, Y is N or Y is N, and X is selected from O and S, the group
    Figure PCTCN2018084352-appb-100004
    for
    Figure PCTCN2018084352-appb-100005
    R 1选自氨基酸Ser和Thr的侧链; R 1 is selected from the side chains of the amino acids Ser and Thr;
    R 2选自氨基酸Ser和Thr残基; R 2 is selected from the group consisting of the amino acid Ser and Thr residues;
    R 3选自H、-(CH 2) mC(O)OR 4、-(CH 2) mC(O)N(R 5)(R 6)和-(CH 2) mCN,其中R 4、R 5、R 6各自独立地选自H和烷基,m为0、1、2、3或4;和 R 3 is selected from the group consisting of H, —(CH 2 ) m C(O)OR 4 , —(CH 2 ) m C(O)N(R 5 )(R 6 ) and —(CH 2 ) m CN, wherein R 4 , R 5 , R 6 are each independently selected from H and alkyl, and m is 0, 1, 2, 3 or 4;
    n为1、2、3或4。n is 1, 2, 3 or 4.
  2. 根据权利要求1所述的通式A所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中通式A具有以下通式II的结构,The compound of the formula A according to claim 1, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein the formula A has the structure of the following formula II,
    Figure PCTCN2018084352-appb-100006
    Figure PCTCN2018084352-appb-100006
    其中:Q选自O和S;R 1、R 2、R 3的定义如权利要求1中所述。 Wherein: Q is selected from O and S; and R 1 , R 2 and R 3 are as defined in claim 1.
  3. 根据权利要求1所述的通式A所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中通式A具有以下通式I的结构,The compound of the formula A according to claim 1, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein the formula A has the structure of the following formula I,
    Figure PCTCN2018084352-appb-100007
    Figure PCTCN2018084352-appb-100007
    其中:Q选自O和S;R 1、R 2、R 3的定义如权利要求1中所述。 Wherein: Q is selected from O and S; and R 1 , R 2 and R 3 are as defined in claim 1.
  4. 根据权利要求1-3之任一项所述的通式A所示的化合物或其立体异构体、药学上可接 受的盐、溶剂合物、结晶或前药,其中:A compound of the formula A according to any one of claims 1 to 3, or a stereoisomer thereof, a pharmaceutically acceptable salt, a solvate, a crystal or a prodrug thereof, wherein:
    R 1选自
    Figure PCTCN2018084352-appb-100008
    R 1 is selected from
    Figure PCTCN2018084352-appb-100008
    R 2选自
    Figure PCTCN2018084352-appb-100009
    R 2 is selected from
    Figure PCTCN2018084352-appb-100009
    R 3选自H、-(CH 2) mC(O)OR 4、-(CH 2) mC(O)N(R 5)(R 6)和-(CH 2) mCN,其中R 4、R 5、R 6各自独立地选自H和C 1-6烷基,m为1、2、3或4;和 R 3 is selected from the group consisting of H, —(CH 2 ) m C(O)OR 4 , —(CH 2 ) m C(O)N(R 5 )(R 6 ) and —(CH 2 ) m CN, wherein R 4 , R 5 and R 6 are each independently selected from H and C 1-6 alkyl, and m is 1, 2, 3 or 4;
    n为1、2、3或4。n is 1, 2, 3 or 4.
  5. 根据权利要求1-3之任一项所述的通式A所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中:A compound of the formula A according to any one of claims 1 to 3, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein:
    R 1选自
    Figure PCTCN2018084352-appb-100010
    R 1 is selected from
    Figure PCTCN2018084352-appb-100010
    R 2选自
    Figure PCTCN2018084352-appb-100011
    R 2 is selected from
    Figure PCTCN2018084352-appb-100011
    R 3选自H、
    Figure PCTCN2018084352-appb-100012
    R 3 is selected from H,
    Figure PCTCN2018084352-appb-100012
    with
    n为1、2、3或4。n is 1, 2, 3 or 4.
  6. 根据权利要求1所述的通式A所示的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药,其中所述化合物为选自以下的化合物:The compound of the formula A according to claim 1, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, wherein the compound is a compound selected from the group consisting of:
    Figure PCTCN2018084352-appb-100013
    Figure PCTCN2018084352-appb-100013
    Figure PCTCN2018084352-appb-100014
    Figure PCTCN2018084352-appb-100014
  7. 一种制备根据权利要求2所述的通式A所示化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药的制备方法,所述方法包括以下步骤:A process for the preparation of a compound of the formula A according to claim 2, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, comprising the steps of:
    Figure PCTCN2018084352-appb-100015
    Figure PCTCN2018084352-appb-100015
    (1)式ia化合物与式iib化合物经过缩合反应生成式iic化合物;(1) a compound of the formula ia and a compound of the formula iib are subjected to a condensation reaction to form a compound of the formula iic;
    (2)式iic化合物环化得到式iid化合物;(2) cyclization of a compound of formula iic to give a compound of formula iid;
    (3)式iid化合物与胺类化合物反应,得到式iie化合物;(3) a compound of the formula iid is reacted with an amine compound to give a compound of the formula iie;
    (4)式iie化合物与式if化合物经过亲核取代反应,得到式iig化合物;(4) a compound of the formula iie and a compound of the formula if subjected to nucleophilic substitution to give a compound of the formula iig;
    (5)在酸性条件下,式iig化合物发生水解反应得到式II化合物;(5) under acidic conditions, a compound of formula iig is hydrolyzed to give a compound of formula II;
    其中:Pg 1代表R 1的保护基;Pg 2代表R 2的保护基;Pg 3代表R 3的保护基或不存在;Pg 4、Pg 5代表氨基保护基。 Wherein: Pg 1 represents a protecting group of R 1 ; Pg 2 represents a protecting group of R 2 ; Pg 3 represents a protecting group of R 3 or is absent; and Pg 4 and Pg 5 represent an amino protecting group.
  8. 一种制备根据权利要求3所述的通式A所示化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药的制备方法,所述方法包括以下步骤:A process for the preparation of a compound of the formula A according to claim 3, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, comprising the steps of:
    Figure PCTCN2018084352-appb-100016
    Figure PCTCN2018084352-appb-100016
    (1)式ia化合物与式ib化合物经过缩合反应生成式ic化合物;(1) a compound of the formula ia and a compound of the formula ib are subjected to a condensation reaction to form a compound of the formula ic;
    (2)式ic化合物环化得到式id化合物;(2) cyclizing a compound of the formula ic to give a compound of the formula id;
    (3)式id化合物与胺类化合物反应,得到式ie化合物;(3) a compound of the formula id is reacted with an amine compound to give a compound of the formula IE;
    (4)式ie化合物与式if化合物发生亲核取代反应,得到式ig化合物;(4) a nucleophilic substitution reaction of a compound of the formula IE with a compound of the formula i to give a compound of the formula ig;
    (5)在酸性条件下,式ig化合物发生水解反应得到式I化合物;(5) under acidic conditions, a compound of formula ig is hydrolyzed to give a compound of formula I;
    其中:Pg 1代表R 1的保护基;Pg 2代表R 2的保护基;Pg 3代表R 3的保护基或不存在;Pg 4、Pg 5代表氨基保护基。 Wherein: Pg 1 represents a protecting group of R 1 ; Pg 2 represents a protecting group of R 2 ; Pg 3 represents a protecting group of R 3 or is absent; and Pg 4 and Pg 5 represent an amino protecting group.
  9. 一种药物组合物,其包含权利要求1-6任意一项所述的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药以及药学上可接受的载体。A pharmaceutical composition comprising a compound according to any one of claims 1 to 6, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, and a pharmaceutically acceptable carrier.
  10. 根据权利要求1-6任意一项所述的化合物或其立体异构体、药学上可接受的盐、溶剂合物、结晶或前药或根据权利要求9所述的药物组合物在制备用于治疗癌症或感染类疾病的药物中的应用。A compound according to any one of claims 1 to 6, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystal or prodrug thereof, or a pharmaceutical composition according to claim 9 Use in medicines for treating cancer or infectious diseases.
PCT/CN2018/084352 2017-04-26 2018-04-25 Heterocyclic compound serving as pd-l1 inhibitor WO2018196768A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710280707 2017-04-26
CN201710280707.6 2017-04-26

Publications (1)

Publication Number Publication Date
WO2018196768A1 true WO2018196768A1 (en) 2018-11-01

Family

ID=63920214

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/084352 WO2018196768A1 (en) 2017-04-26 2018-04-25 Heterocyclic compound serving as pd-l1 inhibitor

Country Status (3)

Country Link
CN (1) CN108794422B (en)
TW (1) TW201841896A (en)
WO (1) WO2018196768A1 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020083336A1 (en) * 2018-10-25 2020-04-30 南京圣和药业股份有限公司 1,3,4-oxadiazole-2-cyclobutyl compounds, preparation method therefor and application thereof
US10710986B2 (en) 2018-02-13 2020-07-14 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10774071B2 (en) 2018-07-13 2020-09-15 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10899735B2 (en) 2018-04-19 2021-01-26 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11059834B2 (en) 2017-08-08 2021-07-13 Chemocentryx, Inc. Macrocyclic immunomodulators
US11130740B2 (en) 2017-04-25 2021-09-28 Arbutus Biopharma Corporation Substituted 2,3-dihydro-1H-indene analogs and methods using same
US11135210B2 (en) 2018-02-22 2021-10-05 Chemocentryx, Inc. Indane-amines as PD-L1 antagonists
US11236085B2 (en) 2018-10-24 2022-02-01 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11266643B2 (en) 2019-05-15 2022-03-08 Chemocentryx, Inc. Triaryl compounds for treatment of PD-L1 diseases
US11426364B2 (en) 2016-06-27 2022-08-30 Chemocentryx, Inc. Immunomodulator compounds
US11485708B2 (en) 2019-06-20 2022-11-01 Chemocentryx, Inc. Compounds for treatment of PD-L1 diseases
US11708326B2 (en) 2017-07-28 2023-07-25 Chemocentryx, Inc. Immunomodulator compounds
US11713307B2 (en) 2019-10-16 2023-08-01 Chemocentryx, Inc. Heteroaryl-biphenyl amides for the treatment of PD-L1 diseases
US11866429B2 (en) 2019-10-16 2024-01-09 Chemocentryx, Inc. Heteroaryl-biphenyl amines for the treatment of PD-L1 diseases
US11872217B2 (en) 2019-07-10 2024-01-16 Chemocentryx, Inc. Indanes as PD-L1 inhibitors
US12083118B2 (en) 2018-03-29 2024-09-10 Arbutus Biopharma Corporation Substituted 1,1′-biphenyl compounds, analogues thereof, and methods using same

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015033301A1 (en) * 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
CN105814028A (en) * 2013-09-06 2016-07-27 奥瑞基尼探索技术有限公司 1,2,4-oxadiazole derivatives as immunomodulators
WO2016142833A1 (en) * 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited 1,2,4-oxadiazole and thiadiazole compounds as immunomodulators

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015033301A1 (en) * 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
CN105814028A (en) * 2013-09-06 2016-07-27 奥瑞基尼探索技术有限公司 1,2,4-oxadiazole derivatives as immunomodulators
WO2016142833A1 (en) * 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited 1,2,4-oxadiazole and thiadiazole compounds as immunomodulators

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11426364B2 (en) 2016-06-27 2022-08-30 Chemocentryx, Inc. Immunomodulator compounds
US11793771B2 (en) 2016-06-27 2023-10-24 Chemocentryx, Inc. Immunomodulator compounds
US11130740B2 (en) 2017-04-25 2021-09-28 Arbutus Biopharma Corporation Substituted 2,3-dihydro-1H-indene analogs and methods using same
US11708326B2 (en) 2017-07-28 2023-07-25 Chemocentryx, Inc. Immunomodulator compounds
US11059834B2 (en) 2017-08-08 2021-07-13 Chemocentryx, Inc. Macrocyclic immunomodulators
US11691985B2 (en) 2017-08-08 2023-07-04 Chemocentryx, Inc. Macrocyclic immunomodulators
US10710986B2 (en) 2018-02-13 2020-07-14 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11555029B2 (en) 2018-02-13 2023-01-17 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11759458B2 (en) 2018-02-22 2023-09-19 Chemocentryx, Inc. Indane-amines as PD-L1 antagonists
US11135210B2 (en) 2018-02-22 2021-10-05 Chemocentryx, Inc. Indane-amines as PD-L1 antagonists
US12083118B2 (en) 2018-03-29 2024-09-10 Arbutus Biopharma Corporation Substituted 1,1′-biphenyl compounds, analogues thereof, and methods using same
US10899735B2 (en) 2018-04-19 2021-01-26 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10774071B2 (en) 2018-07-13 2020-09-15 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11236085B2 (en) 2018-10-24 2022-02-01 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
WO2020083336A1 (en) * 2018-10-25 2020-04-30 南京圣和药业股份有限公司 1,3,4-oxadiazole-2-cyclobutyl compounds, preparation method therefor and application thereof
US11266643B2 (en) 2019-05-15 2022-03-08 Chemocentryx, Inc. Triaryl compounds for treatment of PD-L1 diseases
US11485708B2 (en) 2019-06-20 2022-11-01 Chemocentryx, Inc. Compounds for treatment of PD-L1 diseases
US11872217B2 (en) 2019-07-10 2024-01-16 Chemocentryx, Inc. Indanes as PD-L1 inhibitors
US11713307B2 (en) 2019-10-16 2023-08-01 Chemocentryx, Inc. Heteroaryl-biphenyl amides for the treatment of PD-L1 diseases
US11866429B2 (en) 2019-10-16 2024-01-09 Chemocentryx, Inc. Heteroaryl-biphenyl amines for the treatment of PD-L1 diseases

Also Published As

Publication number Publication date
TW201841896A (en) 2018-12-01
CN108794422B (en) 2022-07-01
CN108794422A (en) 2018-11-13

Similar Documents

Publication Publication Date Title
WO2018196768A1 (en) Heterocyclic compound serving as pd-l1 inhibitor
CN108863963B (en) Heterocyclic compounds as PD-L1 inhibitors
JP7032583B2 (en) Compositions and Methods of Inhibiting Arginase Activity
TWI820044B (en) Antibody-pyrrolobenzodiazepine derivative complex
ES2758506T3 (en) Cytotoxic peptides and conjugates of these
ES2657014T3 (en) Cytotoxic peptides and antibody drug conjugates thereof
BR112016017261B1 (en) Compound, pharmaceutical composition, use of a compound and process for making a compound
CN113614095B (en) Alkylboronic acids as arginase inhibitors
WO2023022231A1 (en) Reversible covalent binding inhibitor for treating or preventing viral infection
JP2022545542A (en) Pyrazole compounds, formulations thereof and methods of using the compounds and/or formulations
JP4927566B2 (en) PAR-2 agonist
JP4728248B2 (en) PAR-2 antagonist
JPH05505820A (en) new compound
JP6953435B2 (en) Dimer compound
JP2017528514A (en) New compounds
JP2017528514A5 (en)
US20230382886A1 (en) Conjugated tlr7 and nod2 agonists
JP7339285B2 (en) TLR7 peptide conjugate
WO2006104190A1 (en) Par-2 agonist
JPH08507552A (en) Novel 3- (substituted methyl) -4-oxa-1-azabicyclo [3.2.0] heptan-7-one derivatives useful as antitumor agents
JP2019532021A (en) Cyclic peptides as C5a receptor antagonists

Legal Events

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

Ref document number: 18791829

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18791829

Country of ref document: EP

Kind code of ref document: A1