CN108794422A - Heterocycle compound as PD-L1 inhibitor - Google Patents

Heterocycle compound as PD-L1 inhibitor Download PDF

Info

Publication number
CN108794422A
CN108794422A CN201810375602.3A CN201810375602A CN108794422A CN 108794422 A CN108794422 A CN 108794422A CN 201810375602 A CN201810375602 A CN 201810375602A CN 108794422 A CN108794422 A CN 108794422A
Authority
CN
China
Prior art keywords
amino
compounds
formula
bases
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN201810375602.3A
Other languages
Chinese (zh)
Other versions
CN108794422B (en
Inventor
王勇
赵立文
刘欣
尹伟
刘笑
于琪
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nanjing Sanhome Pharmaceutical Co Ltd
Original Assignee
Nanjing Sanhome Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanjing Sanhome Pharmaceutical Co Ltd filed Critical Nanjing Sanhome Pharmaceutical Co Ltd
Publication of CN108794422A publication Critical patent/CN108794422A/en
Application granted granted Critical
Publication of CN108794422B publication Critical patent/CN108794422B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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

Landscapes

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

Abstract

The invention belongs to medicinal chemistry arts, it is related to a kind of heterocycle compound as PD-L1 inhibitor and its application, specifically, pharmaceutical composition the present invention relates to compound shown in formula A or its isomers, pharmaceutically acceptable salt, solvate or prodrug, their preparation method and containing these compounds and these compound or compositions are for treating cancer or the purposes of infection class disease.

Description

Heterocycle compound as PD-L1 inhibitor
Technical field
The invention belongs to medicinal chemistry arts, are related to a kind of heterocycle compound as PD-L1 inhibitor and its application, In particular it relates to compound shown in formula A or its isomers, pharmaceutically acceptable salt, solvate or prodrug, it Preparation method and pharmaceutical composition containing these compounds and these compound or compositions for treating cancer or Person infects the purposes of class disease.
Background technology
Programmed death receptor -1 (Programmed Cell Death-1, PD-1) and its ligand PD-Ll (B7H1) belong to In CD28/B7 superfamilies.PD-1 is mainly expressed in T cell, B cell, natural killer cells (Natual Killer Cell, NK Cell) film surface, it is thin that PD-L1 is mainly expressed in ripe cd4 t cell, cd8 t cell, B cell, monocyte, dendron shape Hematopoietic cells and some non-hematopoietic cells such as born of the same parents (Dendritic Cells, DCs), macrophage, as endothelial cell, pancreas islet are thin The film surface of born of the same parents, mast cell etc..Wherein PD-L1 high expression in kinds of tumors, such as lung cancer, gastric cancer, multiple marrow, black Plain tumor and breast cancer etc..The expression of PD-L1 on tumor cell surface interacts with the ligand on T cell surface, and it is thin to can induce T The apoptosis of born of the same parents or the reactivity for reducing T cell make tumour cell escape immune attack to inhibit tumor immune response.Cause This blocks the antagonist of PD1-PDL1 signal paths, and the immune microenvironment of the activation of T cell, reversing tumor, enhancing can be promoted endogenous Property antineoplastic immune effect.Targeting PD-1/PD-L1 inhibitor has broad application prospects in immunotherapy of tumors field.
Current anti-PD-1/PD-L1 Antybody therapies clinically have been displayed with advantage function, however large biological molecule also has There are some disadvantages, such as the limitation of immunogenicity and administration route.Therefore, it is still necessary to develop the targeting with more preferable drug effect PD-1/PD-L1 inhibitor.The inventors found that small molecule drug can specifically regulate and control and/or reconcile PD-L1 And its transduction of related protein kinase, for treatment and the relevant diseases of PD-1/PD-L1.
Invention content
It is an object of the present invention to provide a kind of general formula A compounds represented or its stereoisomer, pharmaceutically may be used Salt, solvate, crystallization or the prodrug of receiving,
Wherein:
X and Y is each independently selected from N, O and S, and when X is N, and Y is selected from O and S;When X is O or S, Y N;When Y is N, X choosings From O and S;When Y is O or S, X N;
When X is N, and Y is selected from O and S or Y is O or S, when X is N, groupForWhen X be O or S, When Y is N or Y is N, when X is selected from O and S, groupFor
R1Side chain selected from amino acid Ser and Thr;
R2Selected from amino acid Ser and Thr residue;
R3Selected from H ,-(CH2)mC(O)OR4、-(CH2)mC(O)N(R5)(R6) and-(CH2)mCN, wherein R4、R5、R6Respectively solely On the spot it is selected from H and alkyl, m 0,1,2,3 or 4;With
N is 1,2,3 or 4.
In some preferred embodiments, a kind of general formula A compounds represented or its stereoisomer, can pharmaceutically connect Salt, solvate, crystallization or the prodrug received for general formula I or II compound represented or its stereoisomer, can pharmaceutically connect Salt, solvate, crystallization or the prodrug received:
Wherein:Q is selected from O and S;R1、R2、R3Definition as described in general formula A.
In some embodiments, general formula A according to the present invention, general formula I or II compound represented or its alloisomerism Body, pharmaceutically acceptable salt, solvate, crystallization or prodrug, wherein:
R1Side chain selected from amino acid Ser and Thr;
R2Selected from amino acid Ser and Thr residue;
R3Selected from H ,-(CH2)mC(O)OR4、-(CH2)mC(O)N(R5)(R6) and-(CH2)mCN, wherein R4、R5、R6Respectively solely On the spot it is selected from H and C1-6Alkyl, m 1,2,3 or 4;With
N is 1,2,3 or 4.
In some embodiments, general formula A according to the present invention, general formula I or II compound represented or its alloisomerism Body, pharmaceutically acceptable salt, solvate, crystallization or prodrug, wherein:
R1It is selected from
R2It is selected from
R3Selected from H ,-(CH2)mC(O)OR4、-(CH2)mC(O)N(R5)(R6) and-(CH2)mCN, wherein R4、R5、R6Respectively solely On the spot it is selected from H and C1-6Alkyl, m 1,2,3 or 4;With
N is 1,2,3 or 4.
In some embodiments, general formula A according to the present invention, general formula I or II compound represented or its alloisomerism Body, pharmaceutically acceptable salt, solvate, crystallization or prodrug, wherein:
R1It is selected from
R2It is selected from
R3Selected from H, With
N is 1,2,3 or 4.
In some embodiments, formula above A according to the present invention, general formula I or II compound represented or its alloisomerism Body, pharmaceutically acceptable salt, solvate, crystallization or prodrug, whereinIt is selected from
In some embodiments, general formula A according to the present invention, general formula I or II compound represented or its alloisomerism Body, pharmaceutically acceptable salt, solvate, crystallization or prodrug, wherein:
R1It is selected from
R2It is selected fromWith
It is selected from One kind having general formula A compounds represented or its stereoisomer, pharmaceutically acceptable salt, solvate, crystallization or prodrug, Its non-limiting examples includes:
It is another object of the present invention to provide a kind of Formula B compound represented or its stereoisomer, pharmaceutically Acceptable salt, solvate, crystallization or prodrug,
Wherein:
X and Y is each independently selected from N, O and S, and when X is N, and Y is selected from O and S;When X is O or S, Y N;When Y is N, X choosings From O and S;When Y is O or S, X N;
When X is N, and Y is selected from O and S or Y is O or S, when X is N, groupForWhen X be O or S, When Y is N or Y is N, when X is selected from O and S, groupFor
R7Side chain selected from amino acid Asn, Gln, Asp, Glu;
R8Selected from amino acid Ser and Thr residue;
R9、R10It is each independently selected from H, alkyl and alkyl hydroxy, R9、R10It is asynchronously H, and
R9Or R10With azacycloalkyl or nitrogen oxacycloalkyl, the azepine are formed together with the amino connected on its α carbon potential Naphthenic base or nitrogen oxacycloalkyl are optionally by one or more selected from hydroxyl, hydroxy alkyl, cyano, amino, halogen, nitro Substituent group is replaced;Or
R9、R10C atoms connected to them form naphthenic base or oxacycloalkyl, the naphthenic base or oxa- ring together Alkyl is optionally replaced by one or more substituent groups selected from hydroxyl, hydroxy alkyl, cyano, amino, halogen, nitro.
In a preferred embodiment, a kind of Formula B compound represented or its stereoisomer, can pharmaceutically connect Salt, solvate, crystallization or the prodrug received for general formula III or IV compounds represented or its stereoisomer, can pharmaceutically connect Salt, solvate, crystallization or the prodrug received:
Wherein:Q is selected from O and S, R7、R8、R9、R10Definition as described in Formula B.
In some embodiments, the Formula B compound represented or its stereoisomer, medicine of offer according to the present invention Acceptable salt, solvate, crystallization or prodrug on, wherein:
R7Side chain selected from amino acid Asn, Gln, Asp, Glu;
R8Selected from amino acid Ser and Thr residue;
R9、R10It is each independently selected from H, C1-6Alkyl and C1-6Alkyl hydroxy, R9、R10It is asynchronously H, and
R9Or R10Azepine C is formed with together with the amino connected on its α carbon potential3-6Naphthenic base or nitrogen oxa- C3-6Naphthenic base, institute State azepine C3-6Naphthenic base or nitrogen oxa- C3-6Naphthenic base is optionally by one or more selected from hydroxyl, hydroxyl C1-6Alkyl, cyano, Amino, halogen, nitro substituent group replaced;Or
R9、R10C atoms connected to them form C together3-6Naphthenic base or oxa- C3-6Naphthenic base, the C3-6Cycloalkanes Base or oxa- C3-6Naphthenic base is optionally by one or more selected from hydroxyl, hydroxyl C1-6Alkyl, cyano, amino, halogen, nitro Substituent group is replaced.
In another preferred embodiment, a kind of Formula B compound represented or its stereoisomer, pharmaceutically may be used Salt, solvate, crystallization or the prodrug of receiving, wherein R9For H, R10Azepine C is formed with together with the amino connected on its α carbon potential3-6 Naphthenic base, the azepine C3-6Naphthenic base is optionally by one or more selected from hydroxyl, hydroxyl C1-6Alkyl, cyano, amino, halogen Plain, nitro substituent group is replaced.
In another preferred embodiment, a kind of Formula B compound represented or its stereoisomer, pharmaceutically may be used Salt, solvate, crystallization or the prodrug of receiving, wherein R9Selected from C1-3Alkyl, R10Selected from C1-3Alkyl, R9、R10Connect with them The C atoms connect form C together3-6Naphthenic base, the C3-6Naphthenic base optionally by one or more selected from hydroxyl, hydroxy alkyl, Cyano, amino, halogen, nitro substituent group replaced.
In another preferred embodiment, a kind of Formula B compound represented or its stereoisomer, pharmaceutically may be used Salt, solvate, crystallization or the prodrug of receiving, wherein R9Selected from C1-3Alkyl, R10Selected from C1-3Hydroxy alkyl, R9、R10With them The C atoms connected form oxa- C together3-6Naphthenic base, the oxa- C3-6Naphthenic base is optionally selected from hydroxyl by one or more Base, hydroxy alkyl, cyano, amino, halogen, nitro substituent group replaced.
One kind having Formula B compound represented or its stereoisomer, pharmaceutically acceptable salt, solvate, knot Brilliant or prodrug, non-limiting examples include:
It is a further object to provide one kind having general formula C compounds represented or its stereoisomer, pharmacy Upper acceptable salt, solvate, crystallization or prodrug,
Wherein:
X and Y is each independently selected from N, O and S, and when X is N, and Y is selected from O and S;When X is O or S, Y N;When Y is N, X choosings From O and S;When Y is O or S, X N;
When X is N, and Y is selected from O and S or Y is O or S, when X is N, groupForWhen X be O or S, When Y is N or Y is N, when X is selected from O and S, groupFor
R11Side chain selected from amino acid Ser and Thr;
R12Selected from amino acid Asn, Gln, Asp, Glu side chain;
R15Selected from H and alkyl;
R13、R14It is each independently selected from H, alkyl, R13、R14It is asynchronously H, and
R13、R14C atoms connected to them form naphthenic base together;Or
R13Or R14Azacycloalkyl or R are formed with together with the imino group of its α carbon potential connection14With R15And they are connected Atom forms the lactone naphthenic base optionally replaced together.
The naphthenic base, azacycloalkyl or lactone naphthenic base optionally by one or more selected from hydroxyl, hydroxy alkyl, Cyano, amino, halogen, nitro substituent group replaced;
In a preferred embodiment, a kind of general formula C compounds represented or its stereoisomer, can pharmaceutically connect Salt, solvate, crystallization or the prodrug received are general formula V or VI compound represented or its stereoisomer, pharmaceutically acceptable Salt, solvate, crystallization or prodrug:
Wherein:Q is selected from O and S;R11、R12、R13、R14、R15Definition as described in general formula C.
In a preferred embodiment, a kind of that there is general formula C compounds represented or its stereoisomer, pharmaceutically Acceptable salt, solvate, crystallization or prodrug, wherein R13、R14C atoms connected to them form C together3-6Cycloalkanes Base;
In another preferred embodiment, a kind of that there is general formula C compounds represented or its stereoisomer, pharmacy Upper acceptable salt, solvate, crystallization or prodrug, wherein R14Azepine C is formed with together with the imino group connected on its α carbon potential3-6 Naphthenic base;
In another preferred embodiment, a kind of that there is general formula C compounds represented or its stereoisomer, pharmacy Upper acceptable salt, solvate, crystallization or prodrug, wherein R14With R15And the atom that they are connected forms lactone C together3-6 Naphthenic base, the lactone naphthenic base is by one or more selected from hydroxyl, oxo, hydroxy alkyl, cyano, amino, halogen, nitro Substituent group is replaced;
One kind having general formula C compounds represented or its stereoisomer, pharmaceutically acceptable salt, solvate, knot Brilliant or prodrug, non-limiting examples include:
It is a further object to provide the compound as shown in general formula I or its stereoisomers, pharmaceutically acceptable Salt, solvate, crystallization or prodrug preparation method, the described method comprises the following steps:
(1) formula ia compounds pass through condensation reaction production ic compounds with formula ib compounds;
(2) formula ic compounds are cyclized to obtain formula id compounds;
(3) formula id compounds are reacted with aminated compounds, obtain formula ie compounds;
(4) with formula if compounds nucleophilic substitution occurs for formula ie compounds, obtains formula ig compounds;
(5) in acid condition, formula ig compounds generation hydrolysis obtains compound of formula I;
Wherein:Pg1Represent R1Protecting group;Pg2Represent R2Protecting group;As described in general formula I, R1And R2It each is selected from amino Sour Ser and Thr side chains can take routine in order to avoid amino acid Ser or Thr side chain further react during the reaction Hydroxyl protection base protected amino acid Ser or Thr side chain, Pg1、Pg2Preferably tertiary butyl (tBu);
Pg3Represent R3Protecting group or be not present;As described in general formula I, R3Selected from H ,-(CH2)mC(O)OR4、-(CH2)mC(O) N(R5)(R6) and-(CH2)mCN;Work as R3It, can to contain hydroxyl or amino etc. in further reaction when labile group It is protected using conventional protecting group;Pg4、Pg5Represent amino protecting group.
It is a further object to provide the compound as shown in general formula II or its stereoisomers, pharmaceutically acceptable Salt, solvate, crystallization or prodrug preparation method, the described method comprises the following steps:
(1) formula ia compounds pass through condensation reaction production iic compounds with formula iib compounds;
(2) formula iic compounds are cyclized to obtain formula iid compounds;
(3) formula iid compounds are reacted with aminated compounds, obtain formula iie compounds;
(4) formula iie compounds pass through nucleophilic substitution with formula if compounds, obtain formula iig compounds;
(5) in acid condition, formula iig compounds generation hydrolysis obtains Formula II compound;
Wherein:Pg1Represent R1Protecting group;Pg2Represent R2Protecting group;As described in general formula II, R1And R2It each is selected from ammonia Base acid Ser and Thr side chain can be taken often in order to avoid amino acid Ser or Thr side chain further react during the reaction The hydroxyl protection base of rule protects hydroxyl, Pg1、Pg2Preferably tertiary butyl (tBu);
Pg3Represent R3Protection or be not present;As described in general formula II, R3Selected from H ,-(CH2)mC(O)OR4、-(CH2)mC(O)N (R5)(R6) and-(CH2)mCN;Work as R3To contain the labile group in further reaction such as hydroxyl or amino, can be used Conventional protecting group protects it;
Pg4、Pg5Represent amino protecting group.
It is a further object to provide such as Formula B, general formula C, general formula III, general formula IV, general formula V or general formula VI or The preparation method of its stereoisomer, pharmaceutically acceptable salt, solvate, crystallization or prodrug, according to the logical of above-mentioned offer The preparation method of Formulas I and Compounds of formula II, those skilled in the art are according to the Conventional wisdom of organic synthesis field using conventional Raw material can be obtained Formula B, general formula C, general formula III, general formula IV, general formula V or general formula VI.
It is a further object to provide a kind of pharmaceutical composition, the pharmaceutical composition contains such as general formula A, leads to Formula B, general formula C, general formula I, general formula II, general formula III, general formula IV, general formula V or general formula VI compounds represented or its alloisomerism Body, pharmaceutically acceptable salt, solvate, crystallization or prodrug and pharmaceutically acceptable carrier.
It is a further object to provide it is a kind of as general formula A, Formula B, general formula C, general formula I, general formula II, general formula III, General formula IV, general formula V, general formula VI compounds represented or its stereoisomer, pharmaceutically acceptable salt, solvate, crystallization Or prodrug or include application of its pharmaceutical composition in preparing the drug for treating cancer or infectious diseases.
In a preferred embodiment, it is a kind of as general formula A, Formula B, general formula C, general formula I, general formula II, general formula III, General formula IV, general formula V or general formula VI compounds represented or its stereoisomer, pharmaceutically acceptable salt, solvate, crystallization Or prodrug or include application of its pharmaceutical composition in preparing the drug for treating cancer or infectious diseases, wherein institute State cancer include but not limited to melanoma, brain tumor (have pernicious astroglia and Oligodendroglioma at Point glioma etc.), the cancer of the esophagus, gastric cancer, liver cancer, cancer of pancreas, colorectal cancer (colon and rectum carcinoma etc.), lung cancer it is (non- Small Cell Lung Cancer, Small Cell Lung Cancer, primary or metastatic carcinoma squamosum etc.), kidney, breast cancer, oophoroma, prostate cancer, skin Cancer, neuroblastoma, sarcoma, osteochondroma, osteoma, osteosarcoma, seminoma, orchioncus, uterine cancer (cervix Cancer, carcinoma of endometrium etc.), it is H/N tumors (cancer of maxilla, laryngocarcinoma, pharynx cancer, tongue cancer, cancer etc. in mouth), Huppert's disease, pernicious Lymthoma (reticulosarcoma, lymphosarcoma, Hodgkin lymphoma etc.), polycythemia vera, (acute grain is thin for leukaemia Born of the same parents' leukaemia, chronic myelocytic leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia etc.), goitre Tumor, tumor of ureter, tumor of bladder, gallbladder cancer, cholangiocarcinoma, chorioepithelioma or pediatric tumors are (especially because of familial sarcoma, dimension Er Musi sarcomas, rhabdomyosarcoma, angiosarcoma, embryonic testis cancer, neuroblastoma, retinoblastoma, liver embryo are thin Born of the same parents' tumor, nephroblastoma etc.) and the cancer combination.
In another preferred embodiment, a kind of such as general formula A, Formula B, general formula C, general formula I, general formula II, general formula III, general formula IV, general formula V or general formula VI compounds represented or its stereoisomer, pharmaceutically acceptable salt, solvate, Crystallization or prodrug include application of its pharmaceutical composition in preparing for the drug for the treatment of cancer or infectious diseases, Described in infectious diseases include but not limited to bacterium, virus and fungal infection.
In a specific embodiment, it is a kind of as general formula A, Formula B, general formula C, general formula I, general formula II, general formula III, General formula IV, general formula V or general formula VI compounds represented have significant inhibiting effect to colon cancer.
Term explanation:
Unless stated to the contrary, the term used in the specification and in the claims has following meanings.
" hydrogen ", " carbon " in the compounds of this invention include its all isotope.Isotope is understood to include with identical Atomicity but with those of different quality number atom, such as the isotope of hydrogen includes tritium and deuterium, the isotope of carbon includes13C With14C。
Term " stereoisomer " refer to atom composition and connection type it is identical, and its three-dimensional arrangement it is different point Son, it includes optical isomer, geometric isomer (being called cis-trans-isomer), and " chirality " is that have to be unable to plyability with its mirror image The molecule of matter;And " achirality " refer to its mirror image can be overlapped molecule.Optical isomer is divided into enantiomter and non-right Reflect isomers." enantiomter " refers to two isomers that cannot be overlapped but be mutually mirror of a compound.It is " non-right Reflect isomers " refer to there are two or multiple chiral centres and its molecule not stereoisomer of mirror image each other.Diastereo-isomerism Body has different physical properties, such as fusing point, boiling point, spectral quality and reactivity.Non-enantiomer mixture can pass through height Analysis operation such as electrophoresis and chromatography, such as HPLC are differentiated to detach.
All stereoisomeric forms in any ratio of the compound of the present invention, including but not limited to diastereomer, enantiomter, along anti- Isomers and their mixture, such as racemic mixture.Many organic compounds all exist with optical active forms, i.e., it The Plane of rotation polarised light of having the ability plane.When describing optically active compound, prefix D, L or R, S are used for indicating molecule The absolute configuration of chiral centre.The symbol that prefix D, L or (+), (-) are used for that compound linearly polarized light is named to rotate, (-) or L Refer to compound it is left-handed, it is dextrorotation that prefix (+) or D, which refer to compound,.The chemical constitution of these stereoisomers is phase With, but their stereochemical structure is different.Specific stereoisomer can be enantiomer, and the mixture of isomers is usual Referred to as enantiomeric mixture.50:50 mixture of enantiomers is referred to as racemic mixture or racemic modification, this may lead Cause there is no stereoselectivity or stereoselectivity in chemical reaction process.Term " racemic mixture " and " racemic modification " refer to The mixture of equimolar two enantiomters lacks optical activity.
According to the selection of starting material and method, the compounds of this invention can with one in possible isomers or they Mixture, such as the form of racemic modification and non-corresponding isomer mixture (this depends on the quantity of asymmetric carbon atom) deposits ?.Chiral synthon or chiral reagent can be used to prepare for optically active (R)-or (S)-isomers, or be torn open using routine techniques Point.
The mixture of any stereoisomer of gained can be separated into according to the difference in component physicochemical properties Pure or substantially pure geometric isomer, enantiomter, diastereoisomer, for example, passing through chromatography and/or fractional crystallization Method.
" pharmaceutically acceptable salt " of the present invention refers to the acid of compound and inorganic and/or organic bronsted lowry acids and bases bronsted lowry form And/or basic salt, also include amphoteric ion salt (inner salt).The compounds of this invention contains amino acid side chain, amino acid residue, therefore It can form inner salt, can also salt corresponding with other inorganics and/or organic bronsted lowry acids and bases bronsted lowry form.
" solvate " of the present invention refers to this hair by being coordinated the complex to form solid-state or liquid with solvent molecule The form of bright compound.Hydrate is the special shape of solvate, wherein being coordinated with water.Within the scope of the present invention, molten Agent closes object and is preferably hydrate.
" crystallization " of the present invention refers to the various solid forms that compound of the present invention is formed, including crystal form, nothing are calmly Shape.
" prodrug " of the present invention refers under the physiological condition of organism, due to being reacted and conversion cost with enzyme, hydrochloric acid in gastric juice etc. The compound of invention is converted to the compound of the present invention and/or by hydrochloric acid in gastric juice etc. by the oxidation of enzyme, reduction, hydrolysis etc. Hydrolysis etc. is converted to the compound of the compound of the present invention.
" alkyl " of the present invention refers to linear or branched saturated hydrocarbon base, preferably C1-8Alkyl, more preferable C1-6Alkyl.Alkane The non-limiting examples of base include methyl, ethyl, n-propyl, isopropyl, normal-butyl, isobutyl group, tertiary butyl, sec-butyl, positive penta Base, 1,1- dimethyl propyls, 1,2- dimethyl propyls, 2,2- dimethyl propyls, 1- ethyl propyls, 2- methyl butyls, 3- methyl Butyl, n-hexyl, 1- Ethyl-2-Methyls propyl, 1,1,2- thmethylpropyl, 1,1- dimethylbutyls, 1,2- dimethylbutyls, 2,2- dimethylbutyls, 1,3- dimethylbutyls, 2- ethyl-butyls, 2- methyl amyls, 3- methyl amyls, 4- methyl amyls, 2, 3- dimethylbutyls, n-heptyl, 2- methylhexyls, 3- methylhexyls, 4- methylhexyls, 5- methylhexyls, 2,3- dimethyl-pentens Base, 2,4- dimethyl amyl groups, 2,2- dimethyl amyl groups, 3,3- dimethyl amyl groups, 2- ethylpentyls, 3- ethylpentyls, n-octyl, 2,3- dimethylhexanyls, 2,4- dimethylhexanyls, 2,5- dimethylhexanyls, 2,2- dimethylhexanyls, 3,3- dimethylhexanyls, 4, 4- dimethylhexanyls, 2- ethylhexyls, 3- ethylhexyls, 4- ethylhexyls, 2- methyl -2- ethylpentyls, 2- methyl -3- ethyls Amyl.
" naphthenic base " of the present invention refers to the unsaturated monocycle of saturation or part or polycyclic substituent group comprising 3-20 carbon Atom, preferably 4-13 carbon atom.The non-limiting embodiment of monocycle alkyl includes cyclopropyl, cyclobutyl, cyclopenta, cyclopentene Base, cyclohexyl, cyclohexenyl group, cyclohexadienyl, suberyl, cycloheptatriene base, cyclooctyl etc., preferably cyclopenta, cyclohexyl. Multi-ring alkyl includes the naphthenic base of loop coil, condensed ring and bridged ring.
" azacycloalkyl " of the present invention refers to the saturated rings formed comprising a nitrogen-atoms and multiple carbon atoms, azacyclo- Alkyl is preferably azepine C3-20Naphthenic base, more preferably azepine C4-13Naphthenic base.More preferably azepine C3-6Naphthenic base.Azacycloalkyl Base group non-limiting embodiment includes pyrrolidinyl and piperidinyl group.
" oxacycloalkyl " of the present invention refers to the saturated rings being made of an oxygen and multiple carbon atoms.Oxacycloalkyl is excellent It is selected as oxa- C3-20Naphthenic base, more preferably oxa- C4-13Naphthenic base.More preferably oxa- C3-6Naphthenic base.
" the lactone naphthenic base " of the present invention refers to by an ester groupWith the saturated rings of multiple carbon atoms composition. Lactone naphthenic base is preferably lactone C3-20Naphthenic base, more preferably lactone C4-13Naphthenic base.More preferably lactone C3-6Naphthenic base.
" alkyl hydroxy " of the present invention refers to-alkyl-OH.
" halogen " of the present invention refers to fluorine, chlorine, bromine, iodine.
" amino " of the present invention refers to-NH2,-NH (alkyl) ,-N (alkyl) (alkyl).
" nitro " of the present invention refers to-NO2
" cyano " of the present invention refers to-CN.
The hydroxyl " hydroxyl " of the present invention refers to-OH.
" benzyl " of the present invention refers to-CH2Phenyl is abbreviated as " Bn ".
The present invention " tertbutyloxycarbonyl " refer to-(O) CO (tBu), it is abbreviated as " Boc ".
" Fmoc- " of the present invention refers to fluorenylmethyloxycarbonyl;
" amino acid " of the present invention refers to the carboxylic acid containing amino, and it is a-amino acid that amino, which is connected on α-carbon, and structure is logical Formula is expressed as CH (COOH) (NH2)-side chain." l-amino acid " of the present invention refers to that the alpha -carbon atom of a-amino acid is left-handed 's;On the contrary, " D- amino acid " refers to general formula structure CH (COOH) (NH2The alpha -carbon atom of)-side chain is dextrorotation.Except sweet ammonia Sour outer, the alpha -carbon atom of other oroteins amino acid is asymmetric carbon atom (four substituent groups being bonded with alpha -carbon atom It is different), therefore amino acid can have stereoisomer, you can to there is different configuration (D- types and two kinds of configurations of L-type).
The non-limiting examples of amino acid include alanine (Ala), arginine (Arg), asparagine (Asn), asparagus fern ammonia It is sour (Asp), cysteine (Cys), glutamine (Gln), glutamic acid (Glu), glycine (Gly), histidine (His), different bright Propylhomoserin (Ile), leucine (Leu), lysine (Lys), methionine (Met), phenylalanine (Phe), proline (Pro), silk ammonia Sour (Ser), threonine (Thr), tryptophan (Trp), tyrosine (Tyr), valine (Val).
" amino acid side chain " of the present invention refers to the component part of amino acid, and amino acid structure general formula is expressed as CH (COOH) (NH2)-R, R, that is, represented amino acid side chain, such as the structure of alanine is CH (COOH) (NH2)-CH3, amino acid side chain be- CH3;The structure of serine is CH (COOH) (NH2)-CH2OH, amino acid side chain are-CH2OH;The structure of threonine is CH (COOH)(NH2)-CH(OH)(CH3), amino acid side chain is-CH (OH) (CH3)。
" amino acid residue " of the present invention refers to having lacked a part of structure compared with parent amino acid structure, is endless Whole amino acid, such as a hydrogen atom on amino amino are substituted by a chemical bond and connect bonding with other atoms, or - the OH of amino acid is replaced connecting bonding with other atoms by chemical bond.As the structure of alanine isSo its ammonia Base acid residue can beOr
The present invention " protecting group " be in order to which nitrogen-atoms and oxygen atom remain unchanged when the other positions of molecule being made to be reacted, It is protected with the group for being easy to slough.The protecting group of the present invention includes amino protecting group and hydroxyl protection base, the present invention Amino protecting group refer to the group for preventing or preventing amino to participate in reaction in next step, it is non-limiting until protecting group is removed Example includes formoxyl, alkyl-carbonyl, alkoxy carbonyl, benzoyl, aromatic alkyl carbonyl, aromatic alkoxy carbonyl, triphenyl first Base, phthalyl, N, N- dimethylaminos methylene, substituted silicyl, tertbutyloxycarbonyl, benzyloxycarbonyl group etc..This The hydroxyl protection base of invention refers to the group for preventing or preventing hydroxyl to participate in reaction in next step, until protecting group is removed.Hydroxyl The example of blocking group include acetyl group, allyl, benzoyl, benzyl, 'beta '-methoxy ethoxyl methyl, methoxy, Dimethoxytrityl [two-(4- methoxyphenyls) phenyl methyls], methoxyl group triphenyl [(4- methoxyphenyls) diphenyl Methyl], to methoxy-benzyl ether, methylthiomethyl, pivaloyl group, THP trtrahydropyranyl, trityl group, silicon substrate (such as: Trimethyl silicon substrate, t-Butyldimethylsilyl, tert-butyl diphenyl silicon substrate, tri isopropyl siloxany methyl and triisopropyl silicon Base).Other examples include alkyl group, such as methyl and tertiary butyl, and other ethers, such as ethoxyethyl group.
Specific implementation mode:
The organic solvent used in following embodiment is referred to as follows:
NMM:N- methylmorpholines;
HATU:2- (7- aoxidizes benzotriazole)-N, N, N', N'- tetramethylurea hexafluorophosphoric acid esters;
TPP:Triphenylphosphine;
HOBT:I-hydroxybenzotriazole;
EDC:1- (3- dimethylamino-propyls) -3- ethyl-carbodiimide hydrochlorides;
DAST:Diethylin sulfur trifluoride;
TBDMSCl:Tert-butyl chloro-silicane;
Fmoc-Asn(Trt)-OH:N- fluorenylmethyloxycarbonyls-altheine.
Embodiment 1:((1- (5- ((1S, 2S) -1- amino -2- hydroxypropyls) -1,3,4- oxadiazole -2- bases) -3- (carboxylic first Base) cyclobutyl) carbamoyl) and-Serine preparation
Step 1:The preparation of 1-((tert-butoxycarbonyl) amino)-3- oxo cyclobutane-1- formic acid
1-((tertbutyloxycarbonyl) amino)-3- oxo cyclobutane formate ethyl esters (50mg, 0.19mmol) are dissolved in methanol NaOH (9.3mg, 0.23mmol) is added in the in the mixed solvent of (1mL), water (0.5mL), reacts at room temperature half an hour, the monitoring of TLC plates Reaction (iodine smoked, visualization Bromocresol green), the reaction was complete.Vacuum distillation removes methanol, remaining liq 1M citric acid tune pH to 4. With EA and moisture liquid, organic phase brine It, anhydrous Na2SO4It is dry.Vacuum distillation, silica gel column separating purification (0-50% EAin PE) obtain white solid 40mg, yield 90%.
Step 2:The preparation of 1- ((tert-butoxycarbonyl) amino) -3- oxo cyclobutane -1- benzyl formates
1-((tert-butoxycarbonyl) amino)-3- oxo cyclobutane-1- formic acid (2g, 8.73mmol) is dissolved in DMF In (20mL), K is added2CO3(1.81g, 13.09mmol), BnCl (1.32g, 10.48mmol).It is placed in 65 DEG C of oil baths, heats 2h is reacted, TLC plates monitoring reaction (the smoked colour developing of iodine), the reaction was complete.With EA and moisture liquid, organic phase brine It is anhydrous Na2SO4It is dry.Vacuum distillation, silica gel column separating purification (0-10%EAin PE) obtain pale white solid 2.1g, yield 75.5%.
Step 3:1- ((tert-butoxycarbonyl) amino) -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane -1- formic acid benzyls The preparation of ester
1- ((tert-butoxycarbonyl) amino) -3- oxo cyclobutane -1- benzyl formates (1g, 3.13mmol) are dissolved in toluene In (15mL), (triphenylphosphine) ethyl acetate (1.53g, 4.38mmol) is added, is heated to reflux, reacts 4h, the monitoring reaction of TLC plates (the smoked colour developing of iodine), the reaction was complete.With EA and moisture liquid, organic phase brine It, anhydrous Na2SO4It is dry.Vacuum distillation, silicon Rubber column gel column isolates and purifies (0-10%EAin PE) and obtains pale yellow oil 0.8g, yield 66%.
Step 4:The preparation of 1- amino -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane -1- benzyl formates
By 1- ((tert-butoxycarbonyl) amino) -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane -1- benzyl formates (40mg, 0.1mmol) is dissolved in the in the mixed solvent of dichloromethane (1mL), trifluoroacetic acid (1mL), reacts at room temperature 2h, TLC plates prison Reaction (the smoked colour developing of iodine) is surveyed, the reaction was complete.Vacuum distillation removes extra trifluoroacetic acid, and 20% Na is added2CO3Aqueous solution stirs 15min is mixed, with EA liquid separations, organic phase brine It, anhydrous Na2SO4It is dry.It is evaporated under reduced pressure to pale yellow oil 30mg, Yield 100%.
Step 5:1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (2- ethyoxyl -2- oxos ethylidene) ring The preparation of butane -1- benzyl formates
1- amino -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane -1- benzyl formates (50mg, 0.17mmol) is molten In the in the mixed solvent of Isosorbide-5-Nitrae-dioxane (1mL), water (0.2mL), Na is added2CO3(22mg, 0.21mmol), is placed in ice bath, FmocCl (53.7mg, 0.21mmol) is added, finishes, is gradually increased to room temperature.1h is reacted, (the smoked colour developing of iodine) is reacted in the monitoring of TLC plates, The reaction was complete.With EA and moisture liquid, organic phase brine It, anhydrous Na2SO4It is dry.Vacuum distillation, silica gel post separation are pure Change (0-10%EA in PE) and obtains colorless oil 64mg, yield 72%.
Step 6:1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (2- ethyoxyl -2- oxoethyls) ring fourth The preparation of alkane -1- formic acid
By 1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane - 1- benzyl formates (800mg, 1.56mmol) are dissolved in methanol (10mL), and Pd/C (80mg, 10%, w/w) is added, and hydrogenate 2h, TLC Plate monitoring reaction, the reaction was complete.It is filtered to remove Pd/C with diatomite, filtrate decompression distills to obtain colorless oil 496mg, yield 75%.
Step 7:2- (3- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (2- (N- (tert-butoxy)-O- (uncles Butyl)-L- allothreonines base) diazanyl -1- carbonyls) cyclobutyl) ethyl acetate preparation
By 1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (2- ethyoxyl -2- oxoethyls) cyclobutane -1- Formic acid (790mg, 1.86mmol) is dissolved in DMF (15mL), sequentially adds ((2S, 3S) -3- (tert-butoxy) -1- diazanyl -1- oxygen Butyl -2- bases) t-butyl carbamate (539mg, 1.86mmol), NMM (188.6mg, 4.65mmol), HATU (710mg, 1.86mmol), 2h is reacted at room temperature, the monitoring reaction of TLC plates, the reaction was complete.Reaction solution is poured slowly into ice water, it is solid that white is precipitated Body filters, washes to obtain white solid 800mg, yield 69%.
Step 8:2- (3- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (5- ((1S, 2S) -2- (tertiary fourth oxygen Base) -1- ((tert-butoxycarbonyl) amino) propyl) -1,3,4- oxadiazole -2- bases) cyclobutyl) ethyl acetate preparation
By 2- (3- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (2- (N- (tert-butoxy)-O- (tertiary fourths Base)-L- allothreonines base) diazanyl -1- carbonyls) cyclobutyl) ethyl acetate (660mg, 0.95mmol) is dissolved in DMF (8mL), THF The in the mixed solvent of (2mL) is placed in ice bath, and TPP (997mg, 3.8mmol), I is added2(966mg, 3.8mmol) stirs 30min To I2It is completely dissolved, Et is added3N (768mg, 7.6mmol).It finishes, withdraws ice bath, react at room temperature 3h, the monitoring of TLC plates is reacted, instead It should be complete.With EA and moisture liquid, organic phase saturated sodium thiosulfate, brine It, anhydrous Na2SO4It is dry.Vacuum distillation, Silica gel column separating purification (0-25%EAin PE) obtains colorless oil 418mg, yield 65%.
Step 9:2- (3- amino -3- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tert-butoxycarbonyl) amino) third Base) -1,3,4- oxadiazole -2- bases) cyclobutyl) and ethyl acetate preparation
By 2- (3- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tert-butoxycarbonyl) amino) propyl) -1,3,4- oxadiazole -2- bases) cyclobutyl) ethyl acetate (606mg, 0.9mmol) is molten In DCM (5mL), Et is added2NH (355mg, 4.85mmol) reacts at room temperature 6h, and the reaction was complete.Vacuum distillation, silica gel post separation It purifies (0-30%EAin PE) and obtains colorless oil 270mg, yield 65%.
Step 10:The preparation of O- (tertiary butyl)-N- ((4-nitrophenoxy) carbonyl)-L-serine tert-butyl ester
O- (tertiary butyl)-L-serine tert-butyl ester (10g, 0.046mol, 1.0eq) is dissolved in DCM (200mL), ice bath drop Et is added dropwise at 0 DEG C in temperature successively3N (9.32g, 0.092mol 2.0eq) and 4- nitros phenyl chloroformate (9.25g, 0.046mol, 1.0eq) DCM (60mL) solution, finish, remove ice bath, be stirred to react 3h.TLC plates monitoring reaction (iodine is smoked), The reaction was complete.Dichloromethane is extracted with water, organic layer saturation Na2CO3Three times, with lemon acid for adjusting pH to 4, saturation is eaten for washing Salt water washing.Anhydrous Na2SO4It is dry.Vacuum distillation, with petroleum ether, product is dissolved in petroleum ether, and filtering subtracts filtrate Pressure distills to obtain colorless oil 10.5g, yield 59.6%.
Step 11:N- ((1- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tertbutyloxycarbonyl) amino) propyl) -1,3, 4- oxadiazole -2- bases) -3- (2- ethyoxyl -2- oxoethyls) cyclobutyl) carbamoyl-O- (tertiary butyl)-Serine uncle The preparation of butyl ester
By 2- (3- amino -3- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tert-butoxycarbonyl) amino) propyl) -1, 3,4- oxadiazole -2- bases) cyclobutyl) ethyl acetate (2.11g, 4.64mmol) be dissolved in DMF (20mL), be added Et3N (0.93g, 9.28mmol) is placed in 0 DEG C, O- (tertiary butyl)-N- ((4-nitrophenoxy) carbonyl)-L-serine tert-butyl ester is added (2.13g, 5.57mmol).It finishes, is warmed to room temperature, react at room temperature 10h, TLC plates monitoring reaction (iodine is smoked), the reaction was complete.Decompression Distillation, silica gel column separating purification (0-50%EA in PE) obtain colorless oil 2.2g, yield 68%.
Step 12:N- ((1- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tertbutyloxycarbonyl) amino) propyl) -1,3, 4- oxadiazole -2- bases) -3- (carboxyl) cyclobutyl) carbamoyl-O- (tertiary butyl)-Serine preparation
By N- ((1- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tertbutyloxycarbonyl) amino) propyl) -1,3,4- Evil bis- Azoles -2- bases) -3- (2- ethyoxyl -2- oxoethyls) cyclobutyl) carbamoyl-O- (tertiary butyl)-L-serine tert-butyl ester (1g, 1.43mmol) is dissolved in the in the mixed solvent of THF (15mL), water (5mL), is placed in 0 DEG C, be added LiOH (240mg, 5.73mmol).It finishes, is warmed to room temperature, react at room temperature 5h, TLC plates monitoring reaction (iodine is smoked), the reaction was complete.Vacuum distillation removes Extra THF, add the acidified with citric acid of 1M to pH be 4, dichloromethane extraction, anhydrous Na2SO4It is dry.Vacuum distillation, silicagel column It isolates and purifies (0-5%MeOH in DCM) and obtains colorless oil 150mg, yield 17%.
Step 13:((1- (5- ((1S, 2S) -1- amino -2- hydroxypropyls) -1,3,4- oxadiazole -2- bases) -3- (carboxymethyl) Cyclobutyl) carbamoyl)-Serine preparation
By N- ((1- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tertbutyloxycarbonyl) amino) propyl) -1,3,4- Evil bis- Azoles -2- bases) -3- (carboxyl) cyclobutyl) carbamoyl-O- (tertiary butyl)-Serine (110mg, 0.179mmol) is dissolved in TFA (1.2mL), tri isopropyl silane (catalytic amount) is added in DCM (4mL) at room temperature, and reaction 5h is stirred at room temperature.Reaction solution is spin-dried for, DCM, water, liquid separation, organic phase water extracted several times is added to merge water phase, then washed for several times with DCM, freeze-drying, preparative separation obtain white Color solid 16mg;Yield:22.2%.1H NMR (500MHz, D2O):δ 4.73 (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]+
Embodiment 2:((1- (5- ((1S, 2S) -1- amino -2- hydroxypropyls) -1,3,4- oxadiazole -2- bases) -3- (2- ethoxies Base -2- oxygen ethyl) cyclobutyl) carbamoyl) and-Serine preparation
Synthetic route:
By N- ((1- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tertbutyloxycarbonyl) amino) propyl) -1,3,4- Evil bis- Azoles -2- bases) -3- (2- ethyoxyl -2- oxoethyls) cyclobutyl) carbamoyl-O- (tertiary butyl)-L-serine tert-butyl ester (200mg, 0.28mmol) is dissolved in DCM (2mL), TFA (2.1mL), tri isopropyl silane (catalytic amount) is added at room temperature, room temperature is stirred Mix reaction 5h.Reaction solution is spin-dried for plus DCM, water, liquid separation, and organic phase water extracted several times merge water phase, then wash number with DCM Secondary, freeze-drying, preparative separation obtain white solid 60mg, yield 48.8%.1H NMR(400MHz,D2O):δ 4.51 (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]+
Embodiment 3:((1- (5- ((1S, 2S) -1- amino -2- hydroxypropyls) -1,3,4- oxadiazole -2- bases) -3- (2- ammonia Base -2- oxygen ethyl) cyclobutyl) carbamoyl) and-Serine preparation
Step 1:3- (2- amino -2- oxos ethylidene) -1- ((tert-butoxycarbonyl) amino) cyclobutane -1- Ethyl formates Preparation
NaH (676mg, 0.016mol, 1.1eq) is dissolved in 10mLDMF, is placed in -20 DEG C of low-temp reaction devices, ammonia is added The DMF solution of base formyl methyl diethyl phosphonate (3g, 0.015mol), drop finish, and stir 1h, and 1-((tertiary fourth is added dropwise dropwise thereto Oxygen carbonyl) amino) -3- oxo cyclobutane formate ethyl esters (3.56g, 0.013mol), drop is complete, and the reaction was continued 4 hours, TLC plates prison Reaction (iodine is smoked) is surveyed, the reaction was complete.Ethyl acetate, water extraction, organic layer are washed twice with saturated salt solution, anhydrous Na2SO4It is dry It is dry.Vacuum distillation, silica gel column separating purification (P/E=1/2) obtain colorless oil 3.02g, yield 73%.
Step 2:The preparation of 3- (2- amino -2- oxoethyls) -1- ((tertbutyloxycarbonyl) amino) cyclobutane -1- Ethyl formates
Title compound is made by the method for the step 6 in embodiment 1, the difference is that by raw material 1- ((((9H- fluorenes -9- Base) methoxyl group) carbonyl) amino) -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane -1- benzyl formates replace with 3- (2- ammonia Base -2- oxos ethylidene) -1- ((tert-butoxycarbonyl) amino) cyclobutane -1- Ethyl formates.
Step 3:The preparation of 3- (2- amino -2- oxoethyls) -1- ((tert-butoxycarbonyl) amino) cyclobutane formate
By ethyl 3- (2- amino -2- oxoethyls) -1- ((tertbutyloxycarbonyl) amino) cyclobutane formate ethyl ester (3.03g, It 0.01mol) is dissolved in THF (80mL), LiOH aqueous solutions (508mg, 0.012mol) is added, react at room temperature 5h, be evaporated under reduced pressure, Extra THF is removed, with the citric acid tune pH to 2 of 1M, is lyophilized, silica gel column separating purification (0-10%MeOH in DCM), will To product direct plunge into next step react.
Step 4:3- (2- amino -2- oxoethyls) -1- carboxyl ring butylamine hydrochlorides
By 3- (2- amino -2- oxoethyls) -1- ((tert-butoxycarbonyl) amino) cyclobutane formate crude product (190mg, It 0.69mmol) is dissolved in the 1.5M HCl ethanol solutions of 2mL, reacts at room temperature 3h.Vacuum distillation, obtains crude product 132mg, yield 91%, obtained product is directly cast into single step reaction.
Step 5:((1- (5- ((1S, 2S) -1- amino -2- hydroxypropyls) -1,3,4- oxadiazole -2- bases) -3- (2- amino - 2- oxygen ethyl) cyclobutyl) carbamoyl) and-Serine preparation
By in embodiment 1 step 5,7,8,9,11,13 method be made title compound, unlike will be in step 5 Raw material 1- amino -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane formate benzyl ester (50mg, 0.17mmol) be dissolved in Isosorbide-5-Nitrae-two Six ring of oxygen replaces with 3- (2- amino -2- oxoethyls) -1- carboxyl ring butylamine hydrochlorides.1H NMR(400MHz,D2O):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]+
Embodiment 4:(((1s, 3R) -1- (5- ((1S, 2S) -1- amino -2- hydroxypropyls) -1,3,4- oxadiazole -2- bases) - 3- (cyano methyl) cyclobutyl) carbamoyl)-Serine preparation
Step 1:The preparation of 1- ((tertbutyloxycarbonyl) amino) -3- (cyanomethylene) cyclobutane formate ethyl ester
Title compound is made in the similar method with the step 3 in embodiment 1 of preparation method, the difference is that by raw material (three Phenylphosphine) ethyl acetate replaces with (triphenylphosphine) acetonitrile.
Step 2:The preparation of (1s, 3s) -1- ((tert-butoxycarbonyl) amino) -3- (cyano methyl) cyclobutane formate ethyl ester
The method of step 6 in preparation method embodiment 1, the difference is that by raw material benzyl 1- ((((9H- fluorenes -9- bases) first Oxygroup) carbonyl) amino) -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane formate ester replaces with 1 gains 1- of above-mentioned steps ((tertbutyloxycarbonyl) amino) -3- (cyanomethylene) cyclobutane formate ethyl ester passes through column chromatography purifying when post-processing and mark is made Inscribe compound and its stereoisomer.
Step 3:The preparation of (1s, 3s) -1- ((tertbutyloxycarbonyl) amino) -3- (cyano methyl) cyclobutane formate
Title compound is made by the method for the step 3 in embodiment 3, the difference is that by raw material ethyl 3- (2- amino -2- Oxoethyl) -1- ((tertbutyloxycarbonyl) amino) cyclobutane formate ethyl ester replaces with 2 gains of above-mentioned steps (1s, 3s) -1- ((tert-butoxycarbonyl) amino) -3- (cyano methyl) cyclobutane formate ethyl ester.
Step 4:The preparation of (1s, 3s) -1- cyano -3- (cyano methyl) ring butylamine hydrochloride
By (1s, 3s) -1- ((tertbutyloxycarbonyl) amino) -3- (cyano methyl) cyclobutane formate (1.4g, 5.5mmol, It 1.0eq) is dissolved in 8mL1, in 4- dioxane, Isosorbide-5-Nitrae-dioxane solution 14mL of HCl is added, reaction overnight, is filtered white Solid 1.04g, yield 100%.
Step 5:(((1s, 3R) -1- (5- ((1S, 2S) -1- amino -2- hydroxypropyls) -1,3,4- oxadiazole -2- bases) -3- (cyano methyl) cyclobutyl) carbamoyl)-Serine preparation
By in embodiment 1 step 5,7,8,9,11,13 method title compound is made, unlike by embodiment 1 Raw material 1- amino -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane formate benzyl ester in step 5 replaces with 4 institute of above-mentioned steps Obtain object (1s, 3s) -1- cyano -3- (cyano methyl) ring butylamine hydrochloride.1H NMR(400MHz,D2O):δ 4.75 (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]+
Embodiment 5:(((1r, 3S) -1- (5- ((1S, 2S) -1- amino -2- hydroxypropyls) -1,3,4- oxadiazole -2- bases) - 3- (cyano methyl) cyclobutyl) carbamoyl)-Serine preparation
(1s, 3s) -1- made from 4 step 2 of embodiment ((tert-butoxycarbonyl) amino) -3- (cyano methyl) cyclobutane Another rotamer of Ethyl formate is raw material, and title compound is made with reference to the method for step 3-5 in embodiment 4.1H NMR(400MHz,D2O):δ 4.75 (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]+.
Embodiment 6:((1- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazole -5- bases) -3- (carboxymethyl) ring Butyl) carbamoyl)-L- allothreonines preparation
Step 1:The preparation of 1- ((benzyloxy) carbonyl) -3- oxo cyclobutane amine hydrochlorates
Title compound is made by the method for the step 4 in embodiment 4, the difference is that by raw material (1s, 3s) -1- ((tertiary fourths Oxygen carbonyl) amino) -3- (cyano methyl) cyclobutane formate replaces with 1- ((tert-butoxycarbonyl) amino) -3- oxo cyclobutane Benzyl formate.
Step 2:The preparation of 1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- oxo cyclobutane formate benzyl esters
Title compound is made by the method for the step 5 in embodiment 1, the difference is that by the raw material in 1 step 5 of embodiment 1- amino -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane formate benzyl ester replaces with 1 gains 1- ((benzyloxies of above-mentioned steps Base) carbonyl) -3- oxo cyclobutane amine hydrochlorates.
Step 3:(2- (tert-butoxy) -2- oxos are sub- by benzyl 1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- Ethyl) cyclobutane formate ester preparation
By 1- (((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- oxo cyclobutane formates benzyl ester (300mg, It 0.68mmol) is dissolved in toluene (5mL), (tert-Butoxycarbonylmethylene) triphenyl phosphorus (282mg, 0.74mmol) is added, sets In 50 DEG C of oil baths, reaction for 24 hours, is evaporated under reduced pressure, EA and moisture liquid, organic phase brine It, anhydrous Na2SO4It is dry.Decompression Distillation, silica gel column separating purification (0-10%EAin PE) obtain colorless oil 263mg, yield 71.8%.
Step 4:1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (2- (tert-butoxy) -2- oxoethyls) The preparation of cyclobutane formate
Title compound is made by the method for the step 6 in embodiment 1, the difference is that by raw material benzyl 1- ((((9H- fluorenes- 9- yls) methoxyl group) carbonyl) amino) -3- (2- ethyoxyl -2- oxos ethylidene) cyclobutane formate ester replaces with 3 institute of above-mentioned steps Obtain object benzyl 1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (2- (tert-butoxy) -2- oxos ethylidene) ring fourth Alkane formic acid esters.
Step 5:(R, Z) -2- (3- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (4- amino -5- (tertiary fourth oxygen Ylmethyl) 9- dimethyl -7- oxo -2,8- dioxa -3,6- diazacyclo amylene -3- alkene -1- bases) cyclobutyl) the tertiary fourth of acetic acid The preparation of ester
By 1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (2- (tert-butoxy) -2- oxoethyls) ring fourth Alkane formic acid (1.51g, 3.34mmol) is dissolved in DMF (20mL), is placed in ice bath, sequentially add raw material (2) (1.19g, 4.35mmol), overnight, reaction solution is poured slowly into for HOBT (540mg, 4mmol), EDC (768mg, 4.0mmol), room temperature reaction In ice water, solid is precipitated, filtering, washes, and crude product is dissolved in ethyl acetate, liquid separation, organic phase anhydrous Na2SO4It is dry.Decompression Distillation, obtains white solid 1.6g, yield 67.5%
Step 6:(R) -2- (3- (((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (3- (2- (tert-butoxy) - 1- ((tertbutyloxycarbonyl) amino) ethyl) -1,2,4- oxadiazole -5- bases) cyclobutyl) and tert-butyl acetate preparation
By (R, Z) -2- (3- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (4- amino -5- (tert-butoxy first Base) 9- dimethyl -7- oxo -2,8- dioxa -3,6- diazacyclo amylene -3- alkene -1- bases) cyclobutyl) tert-butyl acetate (1.6g, 2.27mmol) is dissolved in the in the mixed solvent of ethyl alcohol (40mL), water (4mL), and sodium acetate (280mg, 3.4mmol) is added, Heating reflux reaction 10h.Vacuum distillation, ethyl acetate, water extraction, saturated common salt water washing, organic layer are dry with anhydrous sodium sulfate Dry, vacuum distillation, silica gel column separating purification (P/E=4/1) obtains colorless oil 802mg, yield 52%.
Step 7:((1- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazole -5- bases) -3- (carboxymethyl) ring fourth Base) carbamoyl)-L- allothreonines preparation
By in embodiment 1 step 9,11,13 method title compound is made, unlike by raw material ethyl 2- (3- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tert-butoxycarbonyl) Amino) propyl) -1,3,4- oxadiazole -2- bases) cyclobutyl) acetic acid esters replaces with above-mentioned steps 6 gains (R) -2- (3- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (3- (2- (tert-butoxy) -1- ((tertbutyloxycarbonyl) amino) second Base) -1,2,4- oxadiazole -5- bases) cyclobutyl) and tert-butyl acetate preparation.1H NMR(400MHz,D2O):δ 4.73 (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]+
Embodiment 7:((1- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazole -5- bases) -3- (2- amino -2- Oxoethyl) cyclobutyl) carbamoyl) and-L- allothreonines preparation
Step 1:(R)-(1- (5- (1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (3- (2- amino -2- oxygen Ethyl) cyclobutyl) -1,2,4- oxadiazole -3- bases) -2- (tert-butoxy) ethyl) t-butyl carbamate preparation
By in embodiment 6 step 5,6 method title compound is made, unlike by raw material 1- ((((9H- fluorenes -9- Base) methoxyl group) carbonyl) amino) -3- (2- (tert-butoxy) -2- oxoethyls) cyclobutane formate replaces with 3- (2- amino -2- Oxoethyl) -1- ((tert-butoxycarbonyl) amino) cyclobutane formate.
Step 2:((1- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazole -5- bases) -3- (2- amino -2- oxygen For ethyl) cyclobutyl) carbamoyl) and-L- allothreonines preparation
By in embodiment 1 step 9,11,13 method be made title compound.The difference is that by raw material 2- (3- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tert-butoxycarbonyl) Amino) propyl) -1,3,4- oxadiazole -2- bases) cyclobutyl) ethyl acetate replaces with (R)-(1- (5- (1- ((((9H- fluorenes -9- Base) methoxyl group) carbonyl) amino) -3- (3- (- 2 oxygen ethyl of 2- amino) cyclobutyl) -1,2,4- oxadiazole -3- bases) -2- (tertiary fourths Oxygroup) ethyl) t-butyl carbamate.1H NMR(400MHz,D2O):δ 4.41-4.32 (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]+
Embodiment 8:(((1r, 3S) -1- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazole -5- bases) -3- (cyanogen Ylmethyl) cyclobutyl) carbamoyl) and-L- allothreonines preparation
Step 1:(9H- fluorenes -9- bases) methyl-((1r, 3r) -3- (cyano methyl) -1- (fluoro carbonyl) cyclobutyl) amino The preparation of formic acid esters
By 1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (cyano methyl) cyclobutane formate (200mg, It 0.53mmol) is dissolved in DCM (5mL), DAST (111mg, 0.69mmol) is added, react at room temperature 1h, add water quenching to go out reaction, two Chloromethanes, water extraction, organic layer saturated common salt water washing, Na2SO4It is dry.Vacuum distillation, gains is direct plungeed into next Step reaction.
Step 2:((R) -1- (5- ((1r, 3R) -1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (cyano first Base) cyclobutyl) -1,2,4- oxadiazole -3- bases) 2- (tert-butoxy) ethyl) t-butyl carbamate preparation
By in embodiment 6 step 5,6 method title compound is made, unlike by raw material 1- ((((9H- fluorenes -9- Base) methoxyl group) carbonyl) amino) -3- (2- (tert-butoxy) -2- oxoethyls) cyclobutane formate replaces with (9H- fluorenes -9- bases) Methyl (3- (cyano methyl) -1- (fluoro carbonyl) cyclobutyl) carbamate.
Step 3:(((1R, 3S) -1- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazole -5- bases) -3- (cyano Methyl) cyclobutyl) carbamoyl) and-L- allothreonines preparation
By in embodiment 1 step 9,11,13 method be made title compound.The difference is that by raw material 2- (3- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (5- ((1S, 2S) -2- (tert-butoxy) -1- ((tert-butoxycarbonyl) Amino) propyl) -1,3,4- oxadiazole -2- bases) cyclobutyl) ethyl acetate replaces with ((R) -1- (5- ((1r, 3R) -1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (cyano methyl) cyclobutyl) -1,2,4- oxadiazole -3- bases) 2- (uncles Butoxy) ethyl) t-butyl carbamate.1H NMR(400MHz,D2O):δ 4.65-4.56 (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]+
Embodiment 9:(((1s, 3R) -1- (5- ((S) -1- amino -2- ethoxys) -1,3,4- oxadiazole -2- bases) -3- (cyanogen Ylmethyl) cyclobutyl) carbamoyl) and-L- allothreonines preparation
With (S)-(3- (tert-butoxy) -1- diazanyl -1- oxo propyl- 2- yls) t-butyl carbamate and 1- ((((9H- fluorenes - 9- yls) methoxyl group) carbonyl) amino) -3- (cyano methyl) cyclobutane formate be raw material, by embodiment 1 step 7,8,9, 11, title compound is made in 13 method.
Embodiment 10:(((1r, 3S) -1- (5- ((S) -1- amino -2- ethoxys) -1,3,4- oxadiazole -2- bases) -3- (cyano methyl) cyclobutyl) carbamoyl)-L- allothreonines preparation
With another structure of 1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- (cyano methyl) cyclobutane formate As isomers is raw material, by embodiment 1 step 7,8,9,11,13 the obtained title compound of method.
(((S) -4- amino -1- (5- ((2S, 4R) -4- hydroxy-pyrrolidine -2- bases) -1,3,4- oxadiazoles-of embodiment 11 2- yls) -4- oxos butyl) carbamoyl)-Serine
Step 1 (2S, 4R)-N- (tertbutyloxycarbonyl) -4- ((t-Butyldimethylsilyl) oxygroup) -2- pyrrole methyl formates Preparation
By (2S, 4R)-N- (tertbutyloxycarbonyl) -4- hydroxyl -2- pyrrole methyl formates (5g, 20.4mmol, 1eq) and imidazoles (1.7g, 24.5mmol, 1.2eq) is dissolved in n,N-Dimethylformamide (70mL), and tertiary butyl is added into its solution at 0 DEG C Stir about is for 24 hours at room temperature for dimethylchlorosilane (3.7g, 24.5mmol, 1.2eq).After complete reaction, add into mixed solution Entering saturated ammonium chloride solution and ethyl acetate, organic phase uses saturated ammonium chloride solution, water, saturated nacl aqueous solution to wash successively, Anhydrous sodium sulfate is dried, and is filtered, and concentration obtains product as clear oil (6.0g), yield 81.2%.
Step 2 (2S, 4R) -4- ((t-Butyldimethylsilyl) oxygroup) -2- (Hydrazinocarbonyl) nafoxidine -1- formic acid uncles The preparation of butyl ester
By (2S, 4R)-N- (tertbutyloxycarbonyl) -4- ((t-Butyldimethylsilyl) oxygroup) -2- pyrroles made from step 1 Methyl formate (6g, 16.7mmol) is dissolved in methanol (36mL), and hydrazine hydrate (15mL) is added into its solution, stirs at room temperature for 24 hours. After complete reaction, methanol is spin-dried for, water is added into mixture and ethyl acetate, organic phase are washed with water, saturated sodium-chloride is molten Liquid washs, anhydrous sodium sulfate drying, filters, and concentration obtains pure white solid (5.5g), yield 91.7%.
Step 3 (2S, 4R) -2- (2- (N2(((9H- fluorenes -9- bases) methoxyl group) carbonyl)-N5Trityl-L- glutamy Amine acyl group) diazanyl -1- carbonyls) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine -1- t-butyl formates preparation
By the tertiary fourth of (2S, 4R) -4- ((t-Butyldimethylsilyl) oxygroup) -2- (Hydrazinocarbonyl) nafoxidine -1- formic acid Ester (5.5g, 15.3mmol, 1eq), N2(((9H- fluorenes -9- bases) methoxyl group) carbonyl)-N5Trityl-L-Glutamine (9.4g, 15.3mmol, 1eq), 1- ethyls-(3- dimethylaminopropyls) carbodiimide hydrochloride (3.5g, 18.4mmol, 1.2eq), 1- hydroxy benzo triazoles (2.5g, 18.4mmol, 1.2eq) are dissolved in n,N-Dimethylformamide (50mL), in 0 It is slowly dropped into N-methylmorpholine (3.9g, 38.2mmol, 2.5eq) at DEG C, is stirred to react 12h after dripping off at room temperature.It waits having reacted Mixed solution is put into ice water and is stirred by Quan Hou, and solid is precipitated, and is filtered, is dissolved solid with dichloromethane, is divided and is gone water phase, has Machine is mutually dried with anhydrous sodium sulfate, is spin-dried for, and petroleum ether is added thereto, is filtered, and solid drying obtains pure white solid (14.1g), yield 96.8%.
Step 4 (2S, 4R) -2- (5- ((S) -1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -4- oxos -4- (tritylamino) butyl) -1,3,4- oxadiazole -2- bases) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine -1- The preparation of t-butyl formate
By (2S, 4R) -2- (2- (N2(((9H- fluorenes -9- bases) methoxyl group) carbonyl)-N5Trityl-L-Glutamine acyl Base) diazanyl -1- carbonyls) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine -1- t-butyl formates (14.1g, 14.8mmol, 1eq) it is dissolved in tetrahydrofuran (100mL) and n,N-Dimethylformamide (40mL), triphenyl is added at 0 DEG C Phosphine (7.9g, 30.1mmol, 2eq) and iodine (7.6g, 30.1mmol, 2eq).After iodine is completely dissolved, addition triethylamine (5.9g, 58.6mmol, 4eq), 5h is stirred after adding at room temperature.After complete reaction, water and ethyl acetate are added into mixed solution, Organic phase is washed with hypo solution and saturated nacl aqueous solution, and anhydrous sodium sulfate drying filters, concentrates sand processed, column layer Analysis purifying (ethyl acetate:Petroleum ether=1:3).Obtain white solid (4.8g), yield 34.7%.
Step 5 (2S, 4R) -2- (5- ((S) -1- amino -4- oxos -4- (tritylamino) butyl) -1,3,4- Evil bis- Azoles -2- bases) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine -1- t-butyl formates preparation
By (2S, 4R) -2- (5- ((S) -1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -4- oxo -4- (triphens Methylamino) butyl) -1,3,4- oxadiazole -2- bases) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine -1- formic acid uncles Butyl ester (3g, 3.2mmol) is dissolved in dichloromethane (15mL), and diethylamine (15mL) is added, stirs 4h at room temperature.Wait for that the reaction was complete Afterwards, sand processed is concentrated, column chromatography purifies (ethyl acetate:Petroleum ether=4:1).Obtain white solid (1.7g), yield 74.2%.
The preparation of step 6 O- (tertiary butyl)-N- ((4-nitrophenoxy) carbonyl)-L-serine tert-butyl ester
O- (tertiary butyl)-L-Ser tert-butyl ester (5g, 23.01mmol, 1eq) is dissolved in dichloromethane (25mL), is added Triethylamine (4.7g, 46.02mmol, 2eq), be slowly added dropwise thereto at 0 DEG C 4- chloroformate nitrophenyl esters (5.1g, 25.31mmol, 1.1eq) it is dissolved in dichloromethane (25mL) solution, after dripping off, react at room temperature 3h.After complete reaction, lemon is added Lemon acid and dichloromethane, organic phase citric acid, saturated sodium carbonate solution, water wash successively, dry, and petroleum ether is added in concentration Washing, filters, and filtrate is spin-dried for obtaining white solid (2.2g), yield 25.0%.
Step 7 (2S, 4R) -2- (5- (three oxygen of (6S, 10S) -10- (tertiary fourth oxygen methyl) -13,13- dimethyl -3,8,11- Generation -1,1,1- triphenyl -12- oxygroup -2,7,9- azepine the tetradecane -6- bases) -1,3,4- oxadiazole -2- bases) -4- ((tertiary butyls Dimethyl silicon substrate) oxygroup) nafoxidine -1- t-butyl formates preparation
By (2S, 4R) -2- (5- ((S) -1- amino -4- oxos -4- (tritylamino) butyl) -1,3,4- oxadiazoles - 2- yls) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine -1- t-butyl formates (0.7g, 0.98mmol, 1eq) and three Ethamine (0.2g, 1.97mmol, 2eq) is dissolved in n,N-Dimethylformamide (15mL), and O- (tertiary butyl)-is added dropwise at 0 DEG C N- ((4-nitrophenoxy) carbonyl)-L-serine tert-butyl ester (0.41g, 1.08mmol, 1.1eq), is stirred at room temperature after dripping off Mix 6h.After complete reaction, dichloromethane and water, organic phase water is added, saturated nacl aqueous solution washs, and anhydrous sodium sulfate is dry It is dry, it filters, concentrates sand processed, column chromatography purifies (ethyl acetate:Petroleum ether=2:3) white solid (0.8g) is obtained, yield is 85.2%.
Step 8 (((S) -4- amino -1- (5- ((2S, 4R) -4- hydroxy-pyrrolidine -2- bases) -1,3,4- oxadiazoles -2- Base) -4- oxos butyl) carbamoyl) and-Serine preparation
By (2S, 4R) -2- (5- ((6S, 10S) -10- (tertiary fourth oxygen methyl) -13,13- dimethyl -3,8,11- trioxy-s - 1,1,1- triphenyl -12- oxygroup -2,7,9- azepine the tetradecane -6- bases) -1,3,4- oxadiazole -2- bases) -4- ((tertiary butyl diformazans Base silicon substrate) oxygroup) nafoxidine -1- t-butyl formates (800mg, 0.84mmol) are dissolved in dichloromethane (16mL), trifluoro is added Acetic acid (16mL) and tri isopropyl silane (0.8mL), stir 6h at room temperature.After complete reaction, it is spin-dried for, water and dichloromethane is added Alkane, water phase are washed with dichloromethane, and freeze-drying carries out preparative separation, white solid (190mg), yield 58.6% is lyophilized to obtain.1H NMR (400MHz, D2O):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]+.
(((S) -4- amino -1- (5- ((2S, 3S) -3- hydroxy-pyrrolidine -2- bases) -1,3,4- oxadiazoles-of embodiment 12 2- yls) -4- oxos butyl) carbamoyl) and-Serine preparation
The preparation of step 1 (2S, 3S) -3- hydroxy-pyrrolidine -2- methyl formates
(2S, 3S) -3- hydroxy-pyrrolidine -2- formic acid (4g, 30.5mmol, 1eq) is dissolved in methanol (40mL), in 0 DEG C Lower dropwise addition thionyl chloride (5.44g, 45.8mmol, 1.5eq), after dripping off, mixed solution flows back 3h at 65-70 DEG C, waits reacting After completely, it is spin-dried for, obtains product as clear oil (4.4g), yield 99.3%.
The preparation of step 2 (2S, 3S)-N- (tertbutyloxycarbonyl) -3- hydroxyl -2- pyrrole methyl formates
(2S, 3S) -3- hydroxy-pyrrolidine -2- methyl formates (4.4g, 30.3mmol, 1eq) are dissolved in tetrahydrofuran In (40mL) solution, sodium bicarbonate aqueous solution (8.9g, 106.1mmol, 3.5eq are dissolved in 40mL water) is slowly added thereto, in Di-tert-butyl dicarbonate (6.28g, 28.8mmol, 0.95eq) is added dropwise at 0 DEG C, after dripping off, stirs at room temperature for 24 hours.To mixture Middle addition water and ethyl acetate, organic phase are washed with water, saturated nacl aqueous solution washing, and anhydrous sodium sulfate drying filters, concentration, Obtain white solid (7.4g), yield 99.6%.
Step 3 (((S) -4- amino -1- (5- ((2S, 3S) -3- hydroxy-pyrrolidine -2- bases) -1,3,4- oxadiazoles -2- Base) -4- oxos butyl) carbamoyl) preparation of-Serine
The preparation method for step 1~step 8 that the preparation method is the same as that of Example 1 in 1, unlike by the raw material in embodiment 11 (2S, 4R)-N- (tertbutyloxycarbonyl) -4- hydroxyl -2- pyrrole methyl formates replace with 2 gains of above-mentioned steps (2S, 3S)-N- (tertbutyloxycarbonyl) -3- hydroxyl -2- pyrrole methyl formates.1H NMR (400MHz, D2O):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]+.
Embodiment 13 (2S, 4R) -1- (((S) -4- amino -1- (5- ((1S, 2R) -1- amino -2- hydroxypropyls) -1,3, 4- oxadiazole -2- bases) -4- oxos butyl) carbamoyl) and -4- hydroxy-pyrrolidine -2- formic acid preparation
The preparation of step 1N- (tertbutyloxycarbonyl)-O- (tertiary butyl)-L-threonine methyl esters
N- (tertbutyloxycarbonyl)-O- (tertiary butyl)-L-threonine (5g, 18.2mmol, 1eq) is dissolved in N, N- dimethyl methyls In amide (35mL), the addition iodomethane (2.8g, 20.0mmol, 1.1eq) into its solution, potassium carbonate (7.9g, 54.5mmol, 3eq), stir about 3h at room temperature.After the reaction was complete, water and ethyl acetate extraction are added into mixed solution, organic phase water is satisfied It is washed with sodium chloride solution, anhydrous sodium sulfate drying filters, is concentrated to give faint yellow oil product (5.2g), yield 99.0%.
The preparation of step 2 ((2S, 3R) -3- (tert-butoxy) -1- diazanyl -1- oxo butyl- 2- yls) t-butyl carbamate
N- (tertbutyloxycarbonyl)-O- (tertiary butyl)-L-threonine methyl esters (5.2g, 18.0mmol) is dissolved in methanol, Xiang Qi Hydrazine hydrate (15mL) is added in solution, stirs at room temperature for 24 hours.After the reaction was complete, methanol is spin-dried for, into mixture be added water and Ethyl acetate extracts, organic phase water, saturated nacl aqueous solution washing, and anhydrous sodium sulfate drying filters, and concentration obtains light yellow Solid (5.2g), yield 100.0%.
Step 3 ((5S, 10S, 11R) -1- (9H- fluorenes -9- bases) -11,13,13- trimethyl -3,6,9- trioxy- -5- (3- Oxo -3- (tritylamino) propyl) the three azepine tetradecane -10- bases of -2,12- dioxas -4,7,8-) the tertiary fourth of carbamic acid The preparation of ester
By ((2S, 3R) -3- (tert-butoxy) -1- diazanyl -1- oxo butyl- 2- yls) t-butyl carbamate (5.2g, 18.0mmol, 1eq), N2- (((9H- fluorenes -9- bases) methoxyl group) carbonyl)-N5- trityls-L-Glutamine (11.0g, 18.0mmol, 1eq), 1- ethyls-(3- dimethylaminopropyls) carbodiimide hydrochloride (4.1g, 21.6mmol, 1.2eq), 1- hydroxy benzo triazoles (2.9g, 21.6mmol, 1.2eq) are dissolved in n,N-Dimethylformamide (40mL), at 0 DEG C slowly N-methylmorpholine (4.5g, 44.9mmol, 2.5eq) is instilled, is stirred to react 12h after dripping off at room temperature.After the reaction was complete, it will mix It is stirred in solution input ice water, solid is precipitated, and filters.Solid is dissolved with dichloromethane, divides and water phase, organic phase is gone to be spin-dried for, add Enter petroleum ether, filter, solid drying obtains pure white solid (10.5g), yield 66.0%.
Step 4 ((1S, 2R) -1- (5- ((S) -1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -4- oxos -4- (tritylamino) butyl) -1,3,4- oxadiazole -2- bases) -2- (tert-butoxy) propyl) and t-butyl carbamate preparation
By ((5S, 10S, 11R) -1- (9H- fluorenes -9- bases) -11,13,13- trimethyl -3,6,9- trioxy-s -5- (3- oxygen Generation -3- (tritylamino) propyl) the three azepine tetradecane -10- bases of -2,12- dioxas -4,7,8-) t-butyl carbamate (10.5g, 11.9mmol, 1eq) is dissolved in tetrahydrofuran (75mL) and n,N-Dimethylformamide (30mL), is added at 0 DEG C Triphenylphosphine (6.2g, 23.8mmol, 2eq) and iodine (6.0g, 23.8mmol, 2eq).After iodine is completely dissolved, triethylamine is added (4.8g, 47.6mmol, 4eq), stirs 5h at room temperature after adding.After the reaction was complete, water and acetic acid are added into mixed solution Ethyl ester, organic phase are washed with saturated sodium thiosulfate solution and saturated nacl aqueous solution, and anhydrous sodium sulfate drying filters, concentration Sand processed, column chromatography purify to obtain white solid (3.9g), yield 38.0%.
Step 5 ((1S, 2R) -1- (5- ((S) -1- amino -4- oxos -4- (tritylamino) butyl) -1,3,4- Evil Diazole -2- bases) -2- (tert-butoxy) propyl) t-butyl carbamate preparation
By ((1S, 2R) -1- (5- ((S) -1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -4- oxos -4- (three Benzylamino) butyl) -1,3,4- oxadiazole -2- bases) -2- (tert-butoxy) propyl) t-butyl carbamate (3.9g, It 4.5mmol) is dissolved in dichloromethane (20mL), diethylamine (20mL) is added, stirs 4h at room temperature.After the reaction was complete, sand processed is concentrated, Column chromatography purifies (ethyl acetate:Petroleum ether=4:1).Obtain white solid (1.5g), yield 51.7%.
The preparation of step 6 (2S, 4R) -4- hydroxy-pyrrolidine -2- methyl formates
The preparation method for step 1 that the preparation method is the same as that of Example 1 in 2, unlike by (2S, 3S) -3- hydroxy-pyrrolidines - 2- formic acid has been substituted for (2S, 4R) -4- hydroxy-pyrrolidine -2- formic acid.
The preparation of step 7 (2S, 4R) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine -2- methyl formates
The preparation method for step 1 that the preparation method is the same as that of Example 1 in 1, unlike by (2S, 4R)-N- (tertbutyloxycarbonyl)- 4- hydroxyl -2- pyrrole methyl formates have been substituted for (2S, 4R) -4- hydroxy-pyrrolidine -2- methyl formates.
Step 8 (2S, 4R) -4- ((t-Butyldimethylsilyl) oxygroup) -1- (chloroformyl) nafoxidine -2- formic acid first The preparation of ester
By (2S, 4R) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine -2- methyl formates (1g, 3.9mmol, It 1eq) is dissolved in dichloromethane (100mL), pyridine (0.4g, 5.0mmol, 1.3eq) is added, triphosgene is added dropwise into solution in -5 DEG C Dichloromethane solution (0.5g, 1.7mmol, 0.44eq are dissolved in the DCM of 15mL), it is anti-that stirring is warmed to room temperature after ice bath stirring 1h Answer 18h.It is extracted with citric acid and dichloromethane, organic phase saturated sodium bicarbonate, washing, concentrates sand processed, column chromatography purifying obtains Oily liquids (600mg), yield 48.4%.
Step 9 (1R, 4S) -2- (((S) -1- (5- ((1S, 2R) -2- tert-butoxies -1- (tertbutyloxycarbonyl) amino) third Base) -1,3,4- oxadiazole -2- bases) -4- oxygroups -4- (tritylamino) butyl) carbamoyl) -4- ((tertiary butyl diformazans Base silicon substrate) oxygroup) nafoxidine -1- methyl formates preparation
By ((1S, 2R) -1- (5- ((S) -1- amino -4- oxos -4- (tritylamino) butyl) -1,3,4- Evil bis- Azoles -2- bases) -2- (tert-butoxy) propyl) t-butyl carbamate (700mg, 1.1mmol, 1eq) is dissolved in dioxane (10mL) In, triethylamine (331mg, 3.3mmol, 3eq) and (2S, 4R) -4- ((t-Butyldimethylsilyl) oxygen is added into its solution Base) -1- (chloroformyl) nafoxidine -2- methyl formates (422mg, 1.3mmol, 1.2eq), it is warming up to reflux, stirs 12h. Citric acid is added into mixed solution and ethyl acetate, organic phase saturated sodium bicarbonate solution, water, saturated nacl aqueous solution are washed It washs, anhydrous sodium sulfate drying filters, and concentrates sand processed, and column chromatography purifies (EA:PE=3:2).Obtain white solid (300mg), yield It is 29.7%.
Step 10 (1R, 4S) -2- (((S) -1- (5- ((1S, 2R) -2- tert-butoxies -1- (tertbutyloxycarbonyl) amino) third Base) -1,3,4- oxadiazole -2- bases) -4- oxygroups -4- (tritylamino) butyl) carbamoyl) -4- ((tertiary butyl diformazans Base silicon substrate) oxygroup) nafoxidine -1- formic acid preparation
By (1R, 4S) -2- (((S) -1- (5- ((1S, 2R) -2- tert-butoxies -1- (tertbutyloxycarbonyl) amino) propyl) - 1,3,4- oxadiazole -2- bases) -4- oxygroups -4- (tritylamino) butyl) carbamoyl) -4- ((tert-butyldimethyl silyls Base) oxygroup) nafoxidine -1- methyl formates (300mg, 0.32mmol, 1eq) are dissolved in tetrahydrofuran (2mL), 2M hydroxides are added Lithium solution (4.5mg, 0.64mmol, 2eq are dissolved in 0.05mL water) is heated to (70 DEG C) stirring 3h of reflux.With citric acid and two Chloromethanes extracts, organic phase citric acid, washing, concentrates sand processed, and column chromatography purifies (EA:PE=9:1) white solid is obtained (250mg), yield 84.6%.
Step 11 (2S, 4R) -1- (((S) -4- amino -1- (5- ((1S, 2R) -1- amino -2- hydroxypropyls) -1,3,4- Oxadiazole -2- bases) -4- oxos butyl) carbamoyl) and -4- hydroxy-pyrrolidine -2- formic acid preparation
The preparation method for step 8 that the preparation method is the same as that of Example 1 in 1, unlike by (2S, 4R) -2- (5- ((6S, 10S) - 10- (tertiary fourth oxygen methyl) -13,13- dimethyl -3,8,11- trioxy- -1,1,1- triphenyl -12- oxygroup -2,7,9- azepines ten Four alkane -6- bases) -1,3,4- oxadiazole -2- bases) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine -1- t-butyl formates Be substituted for (1R, 4S) -2- (((S) -1- (5- ((1S, 2R) -2- tert-butoxies -1- (tertbutyloxycarbonyl) amino) propyl) -1, 3,4- oxadiazole -2- bases) -4- oxygroups -4- (tritylamino) butyl) carbamoyl) -4- ((tert-butyldimethyl silyls Base) oxygroup) nafoxidine -1- formic acid, target compound can be made.1H NMR (400MHz, D2O: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]+.
Embodiment 14 (2S, 3S) -1- (((S) -4- amino -1- (5- ((1S, 2R) -1- amino -2- hydroxypropyls) -1,3, 4- oxadiazole -2- bases) -4- oxos butyl) carbamoyl) and -3- hydroxy-pyrrolidine -2- formic acid preparation
The preparation method is the same as that of Example 13 preparation method, unlike changed the 4- hydroxy-prolines in step 7 into 3- Target compound can be made in hydroxy-proline.1H NMR (400MHz, D2O):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]+.
Embodiment 15 (2S, 3R) -2- (3- ((S) -4- amino -1- (5- (1- amino -3- (hydroxymethyl) cyclobutyl) -1, 3,4- oxadiazole -2- bases) -4- oxos butyl) urea groups) and -3-hydroxybutyrate preparation
The preparation method is the same as that of Example 12 preparation method, unlike by (2S, 3S) -3- hydroxyls in 12 step 1 of embodiment Base nafoxidine -2- formic acid has been substituted for 1- amino -3- (methylol) cyclobutane -1- formic acid, and by O- (tertiary butyl)-L- The propylhomoserin tert-butyl ester is substituted for O- (tertiary butyl)-L-threonine tert-butyl ester, and target compound can be made.1H NMR(400MHz,D2O): δ 5.00-4.98 (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]+.
16 2- of embodiment (3- ((S) -4- amino -1- (5- (1- amino cyclopropyl) -1,3,4- oxadiazoles -2-yl) -4- oxygen For butyl) urea groups) -3- hydracrylic acids preparation
The preparation method is the same as that of Example 12 preparation method, unlike by (2S, 3S) -3- hydroxy-pyrrolidine -2- formic acid It has been substituted for 1- amino-cyclopropane -1- formic acid, target compound can be made.1H NMR (400MHz, D2O):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]+.
Embodiment 17 (S) -2- (3- ((S) -4- amino -1- (5- (1- amino -3- (hydroxyl) cyclobutyl) -1,3,4- Evil bis- Azoles -2-yl) -4- oxos butyl) urea groups) and -3- hydracrylic acids preparation
The preparation method is the same as that of Example 12 preparation method, unlike by (2S, 3S) -3- hydroxy-pyrrolidine -2- formic acid It has been substituted for 1- amino -3- (methylol) cyclobutane -1- formic acid, target compound can be made.1H NMR(400MHz,D2O):δ 4.17-4.15 (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]+.
18 1- of embodiment (3- ((S) -4- amino -1- (5- ((1S, 2R) -1- amino -2- hydroxypropyls) -1,3,4- Evil bis- Azoles -2- bases) -4- oxos butyl) urea groups) and ethylene-acetic acid methyl esters preparation
The preparation method of the preparation method is the same as that of Example 1 for step 1~5 in 3 step 1~5, the preparation method of step 6~8 are same The preparation method of step 6~8 in embodiment 11, the difference is that O- (tertiary butyl)-L-serine tert-butyl ester has been substituted for 1- ammonia Basic ring propane -1- methyl formates, can be made target compound.1H NMR(400MHz,D2O):δ 5.01-4.98 (m, 1H), 4.61- 4.59 (d, 1H), 4.28-4.25 (t, 1H), 3.59 (s, 3H), 2.40-2.37 (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]+.
(((S) -4- amino -1- (5- (1- amino -3- hydroxycyclobutyls) -1,3,4- oxadiazole -2- the bases) -4- of embodiment 19 Oxo butyl) carbamoyl)-Serine preparation
The preparation method is the same as that of Example 12 preparation method, unlike by (2S, 3S) -3- hydroxy-pyrrolidine -2- formic acid It has been substituted for 1- amino -3- hydroxycyclobutane -1- formic acid, target compound can be made.1H NMR(400MHz,D2O):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]+.
Embodiment 20 (S) -4- (5- (1- amino -3- (hydroxymethyl) cyclobutyl) -1,3,4- oxadiazole -2- bases) -4- (3- ((1S, 2R) -1- carboxyl -2- hydroxypropyls) urea groups) butyric acid preparation
The preparation method is the same as that of Example 15 preparation method, unlike by N2(((9H- fluorenes -9- bases) methoxyl group) carbonyl) - N5Trityl-L-Glutamine has been substituted for (S) -2- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -5- (tertiary fourths Oxygroup) -5- oxopentanoic acids, target compound can be made.1H NMR(400MHz,D2O):δ 5.00-4.98 (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]+.
Embodiment 21 (S) -4- (5- (1- amino -3- (hydroxymethyl) cyclobutyl) -1,3,4- oxadiazole -2- bases) -4- (3- ((S) -1- carboxyl -2- hydroxyethyls) urea groups) butyric acid preparation
The preparation method is the same as that of Example 17 preparation method, unlike by N2(((9H- fluorenes -9- bases) methoxyl group) carbonyl) - N5Trityl-L-Glutamine has been substituted for (S) -2- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -5- (tertiary fourths Oxygroup) -5- oxopentanoic acids, target compound can be made.1H NMR(400MHz,D2O):δ 4.17-4.15 (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]+.
((1- (5- ((1S, 2R) -1- amino -2- hydroxypropyls) -1,3,4- of embodiment 22Diazole -2- bases) cyclopropyl) ammonia Base formoxyl) preparation of-Serine
The preparation method of the preparation method is the same as that of Example 1 for step 1~5 in 3 step 1~5, unlike by N2- (((9H- fluorenes -9- bases) methoxyl group) carbonyl)-N5Trityl-L-Glutamine has been substituted for 1- ((((9H- fluorenes -9- bases) first Oxygroup) carbonyl) amino) cyclopropane -1- formic acid, the preparation method is the same as that of Example 1 for step 6~8 in 1 6~8 preparation method, can Target compound is made.1H NMR(400MHz,D2O):δ 5.11-5.08 (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]+.
Embodiment 23 (2S, 3S) -2- (3- ((S) -4- amino -1- (5- (4- amino tetrahydrochysene -2H- pyrans -4- bases) -1,3, 4- oxadiazole -2- bases) -4- oxos butyl) urea groups) and -3-hydroxybutyrate preparation
The preparation of step 1 4- amino tetrahydrochysene -2H- pyrans -4- methyl formates
4- amino tetrahydrochysene -2H- pyrans -4- formic acid (5.00g, 34.48mmol) is dissolved in 50mL methanol, stirring cooling To -10 DEG C, thionyl chloride (13.96g, 103.38mmol) is slowly added dropwise, adds temperature rising reflux reaction 3h.Reaction solution concentration is removed Methanol and thionyl chloride are removed, crude product 6.5g is obtained, direct plunges into and reacts in next step.
The preparation of step 2 4- (tertbutyloxycarbonyl) amino tetrahydrochysene -2H- pyrans -4- methyl formates
By 4- amino tetrahydrochysene -2H- pyrans -4- methyl formates (6.50g, 0.04mol) be dissolved in 65mL tetrahydrofurans and In 65mL water, sodium bicarbonate (12.10g, 0.14mol) is added, 0 DEG C is cooled under stirring, di-tert-butyl dicarbonate is added (8.50g, 0.038mol) is added and is warmed to room temperature reaction, about for 24 hours.Reaction solution concentration residue is extracted with ethyl acetate, is saturated Salt is washed, and anhydrous sodium sulfate drying is concentrated to give crude product, column chromatography obtains product 6.3g, yield 60.8%.
The preparation of step 3 4- (tertbutyloxycarbonyl) amino tetrahydrochysene -2H- pyrans -4- hydrazides
4- (tertbutyloxycarbonyl) amino tetrahydrochysene -2H- pyrans -4- methyl formates (5.30g, 0.02mmol) are dissolved in 53mL methanol, adds hydrazine hydrate (13mL, 0.2mmol), and room temperature reaction overnight, reaction solution is concentrated, residue dichloromethane Alkane extracts, a small amount of washing of organic layer, anhydrous sodium sulfate drying, concentration, and column chromatography obtains product 2.5g, yield 47%.
Step 4 (4- (2- (N2(((9H- fluorenes -9- bases) methoxyl group) carbonyl)-N5Trityl-L- glutamyls) hydrazine -1- Carbonyl) tetrahydrochysene -2H pyrans -4- bases) t-butyl carbamate preparation
4- (tertbutyloxycarbonyl) amino tetrahydrochysene -2H- pyrans -4- hydrazides (1.40g, 5.40mmol) is dissolved in 14mL DMF In, N is added at room temperature2((((9H- fluorenes -9- bases) methoxyl group) carbonyl)-N5Trityl-L-Glutamine (3.30g, 5.40mmol), to be dissolved complete, NMM (1.36g, 13.50mmol) and HATU (2.05g, 5.40mmol) are added, room temperature is added 3h is reacted, reaction solution is added in 60mL ice water, solid is precipitated, is filtered, washing is dried to obtain product 4.5g, yield 97.8%.
Step 5 (S)-(4- (5- (1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -4- carbonyls -4- (three methyl Amino) butyl) -1,3,4- oxadiazole -2- bases) tetrahydrochysene -2H- pyrans -4- bases) t-butyl carbamate preparation
By (4- (2- (N2(((9- bis- is thio) methoxyl group) carbonyl)-N5Trityl-L- glutamyls) hydrazine -1- carbonyls Base)-tetrahydrochysene -2H pyrans -4) t-butyl carbamate (4.00g, 4.59mmol) is dissolved in 40mL dichloromethane, is added at room temperature Triphenylphosphine TPP (3.40g, 9.18mmol), it is to be dissolved complete, reaction solution is cooled to -10 DEG C, iodine is slowly added into reaction solution (2.32g, 9.18mmol) triethylamine (3.72g, 36.72mmol) is added after iodine is completely dissolved, adds and is warmed to room temperature reaction 2h.Reaction is concentrated after complete reaction, is extracted with ethyl acetate, organic layer uses hypo solution, saturated common salt respectively Water washing, anhydrous sodium sulfate drying, concentration, column chromatography obtain 2.00g products, yield 51.2%.
Step 6 (S)-(4- (5- (1- amino -4- carbonyl -4- tritylaminos) butyl) -1,3,4- oxadiazole -2- bases) Tetrahydrochysene -2H- pyrans -4- bases) t-butyl carbamate preparation
By (S)-(4- (5- (1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -4- carbonyls -4- (three methyl ammonia Base) butyl) -1,3,4- oxadiazole -2- bases) tetrahydrochysene -2H- pyrans -4- bases) t-butyl carbamate (2.0g, 2.11mmol), it is molten In 20mL dichloromethane, 20mL ethylenediamines are added, adds and reacts at room temperature 6h, TLC is monitored to the reaction was complete;Reaction solution is dense Contracting, column chromatography obtain product yellow solid 1.0g, yield 83.3%.
Step 7N- (((S) -1- (5- (4- ((di-tert-butyl dicarbonate base) amino) tetrahydrochysene -2H- pyrans -4- bases) -1,3, 4- oxadiazole -2- bases) -4- carbonyls -4- (tritylamino) butyl) carbamyl)-O- (the tertiary butyl)-tertiary fourth of L-threonine The preparation of ester
By (S)-(4- (5- (1- amino -4- carbonyl -4- tritylaminos) butyl) -1,3,4- oxadiazole -2- bases) four Hydrogen -2H- pyrans -4- bases) t-butyl carbamate (300mg, 0.49mmol), it is dissolved in 3mL DMF, triethylamine is being added (100mg, 0.98mmol) is added and is cooled to 0 DEG C, adds O- (tertiary butyl)-N- ((4- nitrobenzophenones) carbamoyl)-L- The isoleucine tert-butyl ester (387mg, 0.53mmol) adds and is warmed to room temperature reaction 10h, reaction solution is poured into 20mL water, second is used Acetoacetic ester extracts, organic layer 1M lemon pickling, then is washed with saturated sodium bicarbonate, and organic layer is dried with anhydrous sodium sulfate, concentration, Column chromatography obtains product as white solid 340mg, yield 81.0%.
Step 8 (2S, 3S) -2- (3- ((S) -4- amino -1- (5- (4- amino tetrahydrochysene -2H- pyrans -4- bases) -1,3,4- Evil Diazole -2- bases) -4- oxos butyl) urea groups) and -3-hydroxybutyrate preparation
By N- (((S) -1- (5- (4- ((di-tert-butyl dicarbonate base) amino) tetrahydrochysene -2H- pyrans -4- bases) -1,3,4- Evil Diazole -2- bases) -4- carbonyls -4- (tritylamino) butyl) carbamyl)-O- (the tertiary butyl)-L-threonine tert-butyl ester (300mg, 0.345mmol) is dissolved in 5mL dichloromethane, and 5mL trifluoroacetic acids are added at room temperature, and two drop triisopropyl second are added dropwise Amine reacts at room temperature 5h.Reaction solution is concentrated, 15mL water is added into residue, is extracted with dichloromethane, water layer freeze-drying, excessively instead Xiang Zhu obtains product.1H NMR(400MHz,D2O) δ 4.97 (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]+
Embodiment 24 (S) -2- (3- ((S) -4- amino -1- (5- (4- amino tetrahydrochysene -2H- pyrans -4- bases) -1,3,4- Evil Diazole -2- bases) -4- oxos butyl) urea groups) and -3- hydracrylic acids preparation
With reference to the synthetic route of embodiment 23, you can synthesis (((S) -4- amino -1- (5- (4- amino -2H- pyrans -4- Base) -1,3,4- oxadiazole -2- bases) -4- butyl carbonyl) formamide)-Serine.1H NMR(400MHz,D2O)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]+.
Embodiment 25 (S) -4- (5- (4- amino tetrahydrochysene -2H- pyrans -4- bases) -1,3,4- oxadiazole -2- bases) -4- (3- ((1S, 2S) -1- carboxyl -2- hydroxypropyls) urea groups) butyric acid preparation
With reference to the synthetic route of embodiment 23, you can synthesis (S) -4- (5- (4- amino tetrahydrochysene -2H- pyrans -4- bases) -1, 3,4- oxadiazole -2- bases) -4- (3- ((1S, 2S) -1- carboxyl -2- hydroxypropyls) urea groups) butyric acid.1H NMR(400MHz,D2O)δ 4.94 (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-MS m/z:416.2[M+H]+
Embodiment 26 (S) -4- (5- (4- amino tetrahydrochysene -2H- pyrans -4- bases) -1,3,4- oxadiazole -2- bases) -4- (3- ((S) -1- carboxyl -2- hydroxyethyls) urea groups) butyric acid preparation
With reference to the synthetic route of embodiment 23, you can synthesis (S) -4- (5- (4- amino tetrahydrochysene -2H- pyrans -4- bases) -1, 3,4- oxadiazole -2- bases) -4- (3- ((1S, 2S) -1- carboxyl -2- hydroxyethyls) urea groups) butyric acid.1H NMR(400MHz,D2O)δ 5.03-4.93 (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]+.
Embodiment 27 (S) -4- (3- (4- amino tetrahydrochysene -2H- pyrans -4- bases) -1,2,4- oxadiazole -5- bases) -4- (3- ((S) -1- carboxyl -2- hydroxyethyls) urea groups) butyric acid preparation
The preparation of step 1 4- amino tetrahydrochysene -2H- pyrans -4- methyl formates
4- amino tetrahydrochysene -2H- pyrans -4- formic acid (5.00g, 34.48mmol) is dissolved in 50mLMeOH, stirring is cooled to - 10 DEG C, thionyl chloride (13.96g, 103.38mmolq) is slowly added dropwise, adds temperature rising reflux reaction 3h.Reaction solution is concentrated and is removed Methanol and thionyl chloride obtain crude product 6.5g, not after further treatment, direct plunge into and react in next step.
The preparation of step 2 4- (tertbutyloxycarbonyl) amino tetrahydrochysene -2H- pyrans -4- methyl formates
By 4- amino tetrahydrochysene -2H- pyrans -4- methyl formates (6.50g, 0.04mol) be dissolved in 65mL tetrahydrofurans and In 65mL water, sodium bicarbonate (12.10g, 0.14mol) is added, 0 DEG C is cooled under stirring, di-tert-butyl dicarbonate is added (8.50g, 0.038mol), which is added, is warmed to room temperature reaction, about for 24 hours.Reaction solution concentration residue is extracted with ethyl acetate, is saturated Salt is washed, and anhydrous sodium sulfate drying is concentrated to give crude product, column chromatography obtains product 6.3g, yield 60.8%.
The preparation of step 3 4- (tertbutyloxycarbonyl) amino tetrahydrochysene -2H- pyrans -4- formic acid
4- (tertbutyloxycarbonyl) amino tetrahydrochysene -2H- pyrans -4- methyl formates (7.00g, 27.02mmol) are dissolved in 70mL methanol adds 40mL water, adds (1.63g, 40.53mmol) room temperature reaction 4h, reaction solution is adjusted to 1N hydrochloric acid PH is 4-5, then is extracted with ethyl acetate, washing and drying, is concentrated to give product white solid 5.0g, yield 75.7%
The preparation of step 4 4- (tertbutyloxycarbonyl) amino tetrahydrochysene -2H- pyrans -4- formamides
4- (tertbutyloxycarbonyl) amino tetrahydrochysene -2H- pyrans -4- formic acid (5.00g, 20mmol) is dissolved in 50mL THF, At room temperature be added NMM (2.50g, 24mmol), reaction solution is cooled to -10 DEG C, be slowly added dropwise ethyl chloroformate (2.70g, 24mmol), low-temp reaction 2h is slowly added dropwise ammonium hydroxide (13mL, 160mmol) under low temperature, adds low-temp reaction 3h, contact plate has been reacted Entirely, reaction solution is concentrated, residue is extracted with ethyl acetate, and organic layer uses 1N citric acids, saturated sodium bicarbonate, saturation food respectively Salt is washed, and anhydrous sodium sulfate drying is concentrated to give product as white solid 4.2g, yield 85.7%.
The preparation of step 5 (4- cyano tetrahydrochysene -2H- pyrans -4- bases) t-butyl carbamate
4- (tertbutyloxycarbonyl) amino tetrahydrochysene -2H- pyrans -4- formamides (3.70g, 15.14mmol) are dissolved in pyridine Trifluoroacetic anhydride (4.80g, 22.71mmol) is added dropwise in (12.0g, 151.4mmol), under low temperature and adds room temperature reaction 4h, it will be anti- It answers liquid to import in saturated sodium bicarbonate solution, is extracted with ethyl acetate, organic layer 1M lemon pickling, saturated sodium bicarbonate solution It washes, anhydrous sodium sulfate drying is concentrated to give crude product, is washed with petroleum ether, obtains product 2.9g, yield 78%.
The preparation of step 6E- (4- (N'- hydroxy formamidines base) tetrahydrochysene -2H- pyrans -4- bases) t-butyl carbamate
By (4- cyano tetrahydrochysene -2H- pyrans -4- bases) t-butyl carbamate (2.50g, 0.01mol), it is dissolved in 25mL ethyl alcohol In, 5mL water, hydroxylamine hydrochloride (1.20g, 0.015mol) are added, it is anti-to add temperature rising reflux for potassium carbonate (2.30g, 0.015mol) It answers.Reaction solution is concentrated, residue is dissolved with ethyl acetate, saturated common salt washing, and it is white to be concentrated to give product for anhydrous sodium sulfate drying Color solid 2.50g, yield 96.5%.
Step 7 tertiary butyl (S, Z) -4- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -5- (((((((tert-butoxies Carbonyl) amino) tetrahydrochysene -2H- pyrans -4- bases) methylene) amino) oxygroup) -5- oxopentanoic acid methyl esters preparation
By E- (4- (N'- hydroxy formamidines base) tetrahydrochysene -2H- pyrans -4- bases) t-butyl carbamate (2.00g, 7.72mmol), it is dissolved in 20mL DCM, adds NMM (1.6g, 15.44mmol), (S) -2- (((((9H- fluorenes -9- bases) first Oxygroup) carbonyl) amino) -5- (tert-butoxy) -5- oxopentanoic acids (3.6g, 8.49mmol) add room temperature reaction, TLC monitor to The reaction was complete, and reaction solution is concentrated, and residue column chromatography obtains product white solid 2.68g, yield 52%.
Step 8 tertiary butyl (S) -4- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -4- (3- (4- ((tert-butoxies Carbonyl) amino) tetrahydrochysene -2H- pyrans-pyridin-4-yl) -1,2,4- oxadiazole -5- bases) ethyl butyrate preparation
By tertiary butyl (S, Z) -4- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -5- (((((((tert-butoxy carbonyls Base) amino) tetrahydrochysene -2H- pyrans -4- bases) methylene) amino) oxygroup) -5- oxopentanoic acid methyl esters (2.20g, 3.29mmol) are molten Solution adds sodium acetate, temperature rising reflux reaction, TLC is monitored to the reaction was complete, and reaction solution is dense in 22mL ethyl alcohol and 3mL water Contracting, residue is dissolved with ethyl acetate, then is washed with saturated common salt, and anhydrous sodium sulfate drying, column chromatography obtains product 1.80g, receives Rate 84.5%.
Step 9 tertiary butyl (S) -4- amino -4- (3- (4- ((tert-butoxycarbonyl) amino) tetrahydrochysene -2H- pyrans -4- bases) - 1,2,4- oxadiazole -5- bases) butyrate preparation
By tertiary butyl (S) -4- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -4- (3- (4- ((tert-butoxy carbonyls Base) amino) tetrahydrochysene -2H- pyrans-pyridin-4-yl) -1,2,4- oxadiazole -5- bases) ethyl butyrate (1.80g, 2.77mmol), it is molten In 18mL dichloromethane, 18mL diethylamine is added, adds and reacts at room temperature 6h, TLC is monitored to the reaction was complete.Reaction solution is dense Contracting, column chromatography obtain product yellow solid 1.0g, yield 84.7%.
Step 10 tertiary butyl (S) -4- (3- (4- ((tert-butoxycarbonyl) amino) tetrahydrochysene -2H- pyrans -4- bases) -1,2,4- Oxadiazole -5- bases) -4- (3- (two tert-butoxy -1- oxo propyl- 2- yls of (S) -1,3-) urea groups) ethyl butyrate preparation
By tertiary butyl (S) -4- amino -4- (3- (4- ((tert-butoxycarbonyl) amino) tetrahydrochysene -2H- pyrans -4- bases) -1, 2,4- oxadiazole -5- bases) butyrate (300mg, 0.70mmol), be dissolved in 3mL DMF, be added triethylamine (142mg, 1.40mmol), it adds and is cooled to 0 DEG C, add O- (tertiary butyl)-N- ((4- nitrobenzophenones) carbamoyl)-Serine The tert-butyl ester (352mg, 0.77mmol) adds and is warmed to room temperature reaction 10h, reaction solution is poured into 20mL water, is extracted with ethyl acetate It takes, organic layer 1M lemon pickling, then is washed with saturated sodium bicarbonate, organic layer is dried with anhydrous sodium sulfate, and concentration, column chromatography obtains To product as white solid 300mg, yield 80%.
Step 11 (S) -4- (3- (4- amino tetrahydrochysene -2H- pyrans -4- bases) -1,2,4- oxadiazole -5- bases) -4- (3- ((S) -1- carboxyl -2- hydroxyethyls) urea groups) butyric acid
By tertiary butyl (S) -4- (3- (4- ((tert-butoxycarbonyl) amino) tetrahydrochysene -2H- pyrans -4- bases) -1,2,4- Evil bis- Azoles -5- bases) -4- (3- (bis- tert-butoxy -1- oxo propyl- 2- yls of (S) -1,3-) urea groups) ethyl butyrate (150mg, It 0.219mmol) is dissolved in 5mL dichloromethane, 5mL trifluoroacetic acids is added at room temperature, two drop triisopropyl ethamine, room temperature is added dropwise 5h is reacted, reaction solution is concentrated, 15mL water is added into residue, is extracted with dichloromethane, water layer freeze-drying obtains product.1H NMR(400MHz,D2O) δ 4.94 (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]+
Embodiment 28 (S) -3- amino -3- (3- ((2S, 4R) -4- hydroxy-pyrrolidine -2- bases) -1,2,4- oxadiazoles -5- Base) propionamide preparation
Step 1:(2S, 4R) -1- tertbutyloxycarbonyl -2- cyano -4- (t-Butyldimethylsilyl) oxygroup nafoxidine It prepares
Compound 1a (30g, 141.34mmol) is dissolved in DMF (300mL), imidazoles (11.55g, 169.61mmol) is added, DMF (120mL) solution of TBDMSCl (25.56g, 169.61mmol) is slowly added dropwise after being cooled to -5 DEG C, is stirred to react 2h, rises To room temperature reaction 13h, reaction solution is poured into ammonium chloride saturated solution and is quenched, adds ethyl acetate extraction, liquid separation, organic phase saturation For several times, anhydrous sodium sulfate drying is evaporated under reduced pressure to yellow oily liquid, is white solid after standing is cooling for saline solution, water washing, By crude compound silica gel chromatography (eluant, eluent:The ethyl acetate of 0-5% in petroleum ether), collect product component, rotation It does, stand cooling 44.61g compound as white solid 1b (yields:91.7%).
Step 2:(2S, 4R) -1- tertbutyloxycarbonyls -2- ((Z)-N'- hydroxy formamidines base) -4- (t-Butyldimethylsilyl) The preparation of oxygroup nafoxidine
Compound 1b (20g, 61.25mmol) is dissolved in ethyl alcohol (200mL), sequentially add potassium carbonate (12.70g, 91.88mmol), hydroxylamine hydrochloride (6.38g, 91.88mmol) is stirred at reflux reaction 5h.Reaction solution filters, and filtrate is spin-dried for, and uses second Acetoacetic ester and water-soluble clear, liquid separation, organic phase saturated salt solution, water washing for several times, anhydrous Na2SO4It is dry, vacuum distillation, to institute It obtains and ethyl acetate is added in solid, ultrasound, filtering, filter cake are dissolved with a small amount of tetrahydrofuran, bulk petroleum ether crystallization, mistake are added Filter, filter cake are white solid sterling, filtrate decompression distillation, with silica gel chromatography (eluant, eluent:The second of 0-30% in petroleum ether Acetoacetic ester).Obtained solid merges with filter cake, there are 9.6g product (yields:43.6%).
Step 3:(2S, 4R) -1- tertbutyloxycarbonyls -2- (5- ((S) -1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) ammonia Base) -3- ketone groups -3- (tritylamino) propyl) -1,2,4- oxadiazole -3- bases) -4- ((t-Butyldimethylsilyl) oxygen Base) nafoxidine preparation
Compound Fmoc-Asn (Trt)-OH (20.71g, 34.71mmol) is dissolved in dichloromethane (200mL), ice bath drop BAST (8.45g, 38.18mmol) is added at -7 DEG C, reacts 8h at this temperature for temperature.Vacuum distillation removes reaction solution, is added suitable N-hexane is measured, shaking, filtering are repeated several times, filtration cakes torrefaction, are dissolved with tetrahydrofuran (200mL), addition compound 1c (9.6g, 26.70mmol), reaction 13h is stirred at room temperature in N-methyl morpholine (5.40g, 53.40mmol).Vacuum distillation removes tetrahydrofuran, It is directly added into ethyl alcohol (200mL), sodium acetate (3.29g, 40.05mmol), is stirred at reflux reaction 2.5h.Vacuum distillation removes dereaction Liquid adds ethyl acetate to dissolve, and filtering, filtrate uses saturated sodium bicarbonate, 1M citric acids and saturated common salt water washing, anhydrous sulphur successively The drying of sour sodium, vacuum distillation, with silica gel chromatography (eluant, eluent:The ethyl acetate of 0-25% in petroleum ether), it is light to obtain 7.18g Yellow color solid 1d (yields:29.2%).
Step 4:(2S, 4R) -1- tertbutyloxycarbonyls -2- (5- ((S) -1- amino -3- ketone groups -3- (tritylamino) third Base) -1,2,4- oxadiazole -3- bases) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine preparation
Compound 1d (6.0g, 6.52mmol) is added in dichloromethane (60mL), diethylamine (30mL), room temperature is added dropwise It is stirred to react about 1.5h, vacuum distillation removes solvent, with silica gel chromatography (eluant, eluent:The acetic acid of 0-45% in petroleum ether Ethyl ester), obtain 2.8g yellow oil product 1e (yields:61.5%).
Step 5:(S) -3- amino -3- (3- ((2S, 4R) -4- hydroxy-pyrrolidine -2- bases) -1,2,4- oxadiazoles -5- Base) propionamide preparation
Compound 1e (600mg, 0.86mmol) is dissolved in dichloromethane (18mL), trifluoroacetic acid (18mL) is added at room temperature With the tri isopropyl silane of catalytic amount, reaction 4.5h is stirred at room temperature, vacuum distillation removes solvent, prepares liquid phase and purifies to obtain compound 1。1H NMR(300MHz,D2O) δ 5.05 (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]+.
Embodiment 29 (2S, 3R) -2- (3- ((S) -3- amino -1- (3- ((2S, 4R) -4- hydroxy-pyrrolidine -2- bases) - 1,2,4- oxadiazole -5- bases) -3- oxopropyls) urea groups) and -3-hydroxybutyrate preparation
Step 1:(2S, 4R) -1- tertbutyloxycarbonyls -2- (5- ((5S, 9S) -9- ((R) -1- (tertiary butyl) ethyl) -12, Three ketone group -1,1,1- triphenyl -11- oxa-s -2,6,8- of 12- dimethyl -3,7,10-, three azepine -5- bases) -1,2,4- oxadiazoles - 3- yls) -4- ((t-Butyldimethylsilyl) oxygroup) nafoxidine preparation
Compound 1e (1.0g, 1..43mmol) is dissolved in tetrahydrofuran (10mL), compound 2b is added dropwise at room temperature (0.68g, 1.72mmol) and triethylamine (0.29g, 2.86mmol).It is stirred at room temperature for 24 hours, second is added after removing solvent in vacuum distillation Acetoacetic ester and water dissolution, liquid separation, for several times with saturated common salt water washing, anhydrous sodium sulfate drying is evaporated under reduced pressure, uses silica gel organic phase Column chromatography purifies (eluant, eluent:The ethyl acetate of 0-25% in petroleum ether), obtain 700mg pale yellow crystals 2a (yields:51.3%).
Step 2:(2S, 3R) -2- (3- ((S) -3- amino -1- (3- ((2S, 4R) -4- hydroxy-pyrrolidine -2- bases) -1, 2,4- oxadiazole -5- bases) -3- oxopropyls) urea groups) and -3-hydroxybutyrate preparation
Compound 2a (550mg, 0.576mmol) is dissolved in dichloromethane (11.5mL), trifluoroacetic acid is added at room temperature The tri isopropyl silane of (11.5mL), catalytic amount, are stirred at room temperature reaction 5h, and vacuum distillation removes reaction dissolvent, adds methylene chloride With water dissolution, liquid separation, organic phase water extracted several times merge water phase, then are washed for several times with dichloromethane, and freeze-drying prepares liquid phase Purify to obtain compound 2.1H NMR(500MHz,D2O) δ 5.46 (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]+.
Step 3:The preparation of O- (tertiary butyl)-N- ((4-nitrophenoxy) carbonyl)-L-threonine tert-butyl ester
By H-Thr (OtBu)-OtBu (15g, 64.84mmol) is dissolved in dichloromethane (150mL), ice bath cooling, at -5 DEG C The dichloromethane of triethylamine (9.84g, 97.26mmol) and chloro-carbonic acid -4- nitrobenzenes fat (14.38g, 71.33mmol) are added dropwise successively Alkane (60mL) solution, removes ice bath, and reaction 2h is stirred at room temperature, and vacuum distillation removes solvent, adds ethyl acetate, there is solid precipitation, Partial solvent is distilled off in filtering, filtrate decompression, and the 1 of ethyl acetate and petroleum ether is added:1 mixed solvent, and have solid precipitation, (eluant, eluent is further purified with silica gel column chromatography in filtering, filtrate decompression distillation:The ethyl acetate of 0-5% in petroleum ether), it obtains 13.86g yellow oily liquid 2b (yields:53.9%).
Embodiment 30 (S) -2- (3- ((S) -3- amino -1- (3- ((2S, 4R) -4- hydroxy-pyrrolidine -2- bases) -1,2, 4- oxadiazole -5- bases) -3- oxopropyls) urea groups) and -3- hydracrylic acids preparation
Similar method synthesising title compound is prepared using with embodiment 29, wherein with H-Ser (OtBu)-OtBu is replaced H-Thr (OtBu)-OtBu in embodiment 29.1H NMR(300MHz,D2O) δ 5.36 (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]+.
Embodiment 31 (S) -3- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazole -5- bases) -3- (3- ((S) - 2- ketone group tetrahydrofuran -3- bases) urea groups) propionamide preparation
Step 1:(S) preparation of -2- (tertbutyloxycarbonyl) amino -3- tert-butoxy -1- propionamides
L-Boc-Ser (tBu)-OH (50g, 191.45mmol) is dissolved in tetrahydrofuran (500mL), N-methylmorpholine is added Ethyl chloroformate (24.8g, 229.74mmol) is slowly added dropwise in (23.2g, 229.74mmol) after being cooled to -15 DEG C, -15 DEG C are stirred Mix 1.5h;Ammonium hydroxide (48mL, 693.48mmol) is slowly added dropwise into reaction solution, is warming up to -5 DEG C of reaction 2.5h, vacuum distillation removes Tetrahydrofuran, remaining liq ethyl acetate and water-soluble clear, liquid separation, organic phase is gone to use 1M citric acids, saturated sodium bicarbonate successively With saturated common salt water washing, anhydrous sodium sulfate drying, vacuum distillation obtains creamy white crystals, adds a small amount of ethyl acetate and a large amount of stones Oily ether recrystallizes to obtain 39.3g white solid 4a (yields:78.9%).
Step 2:(R)-(2- (tert-butoxy) -1- cyanoethyls) preparation of t-butyl carbamate
Compound 4a (39g, 149.81mmol) is dissolved in pyridine (118.5g, 1498.1mmol), ice bath cooling, in -5 DEG C Lower dropwise addition trifluoroacetic anhydride (47.2g, 224.72mmol) reacts 40min, removes ice bath, and reaction 3.5h is stirred at room temperature, will react Liquid pours into ethyl acetate, and uses saturated sodium bicarbonate, 1M citric acids and saturated common salt water washing successively, and anhydrous sodium sulfate is dry It is dry, it is evaporated under reduced pressure to 37.36g pale yellow crystals 4b (yields:102.9%).
Step 3:The system of (R, Z)-(1- amino -3- (tert-butoxy) -1- (oximido) propane -2- bases) t-butyl carbamate It is standby
Compound 4b (23g, 184.88mmol) is dissolved in ethyl alcohol (450mL), sequentially add potassium carbonate (38.34g, 277.33mmol), hydroxylamine hydrochloride (19.28g, 277.33mmol), stirring, back flow reaction 1.5h, vacuum distillation remove solvent, add Ethyl acetate and water dissolution, liquid separation, for several times with saturated common salt water washing, anhydrous sodium sulfate drying, vacuum distillation obtains yellow organic phase Color oily liquids is condensed into faint yellow solid after standing cooling, PE is added to be beaten, and filters, and filtration cakes torrefaction obtains 38.67g white solids 4c (yields:76.0%).
Step 4:Tertiary butyl-(triphenyl -7 (5S, Z) -9- amino -13,13- dimethyl -3,6- diketo -1,1,1-, 12- dioxa -2,8- azepine -8- alkene -5,10- diyls) diamino acid (9H- fluorenes -9- bases) methyl ester preparation
Fmoc-Asn (Trt)-OH (10g, 36.32mmol) and compound 4c is dissolved in DMF (200mL), ice bath cools down, HOBt (5.90g, 43.58mmol) and EDC (8.35g, 43.58mmol) is added at -5 DEG C, ice bath stirring reacts 20min, rises Temperature reacts at room temperature 4h, reaction solution is poured into water, and has a large amount of solids to be precipitated, and filtering, filter cake is washed with water for several times, dry 33.8g white solid 4d (yields:108.98%).
Step 5:((R) -1- (5- ((S) -1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- ketone groups -3- (three Benzylamino) propyl) -1,2,4- oxadiazole -3- bases) -2- (tert-butoxy) ethyl) t-butyl carbamate preparation
Compound 4d (28.0g, 32.79mmol) is dissolved in ethyl alcohol (420mL), be added sodium acetate (4.04g, Water (60mL) solution 49.19mmol) is stirred at reflux reaction 2h, and vacuum distillation removes reaction dissolvent, and ethyl acetate dissolving is used Saturated common salt water washing for several times, is evaporated under reduced pressure, with silica gel chromatography (eluant, eluent:The acetic acid second of 0-30% in petroleum ether Ester).Obtain 6.6g Off-white solid 4e (yields:24.1%).
Step 6:((R) -1- (5- ((S) -1- amino -3- ketone groups -3- (tritylamino) propyl) -1,2,4- Evil bis- Azoles -3- bases) -2- (tert-butoxy) ethyl) t-butyl carbamate preparation
Compound 4e (6.6g, 7.89mmol) is added in dichloromethane (66mL), diethylamine (33mL), room temperature is added dropwise It is stirred to react 3.5h.Vacuum distillation removes solvent, with silica gel chromatography (eluant, eluent:The acetic acid second of 0-50% in petroleum ether Ester), obtain 3.78g pale yellow crystals 4f (yields:78.1%).
Step 7:(S)-(2- ketone group tetrahydrofuran -3- bases) preparation of carbamic acid 4- nitro phenyl esters
Similar method synthesising title compound is prepared using with compound 2b in embodiment 29, wherein with (S) -2- ketone H-Thr (O in base -3- amido tetrahydrofurans alternative embodiment 29tBu)-OtBu。
Step 8:((R) -2- (tert-butoxy) -1- (5- ((S) -3- ketone groups -1- (3- ((S) -2- ketone group tetrahydrofurans -3- Base) urea groups) -3- (tritylamino) propyl) -1,2,4- oxadiazole -3- bases) ethyl) and t-butyl carbamate preparation
Compound 4f (500mg, 0.815mmol) and 4g are dissolved in tetrahydrofuran (5mL), be added triethylamine (9.84g, 97.26mmol), reaction 22h is stirred at room temperature.Vacuum distillation removes solvent, adds ethyl acetate to dissolve, with saturated common salt water washing number It is secondary, anhydrous sodium sulfate is dry, vacuum distillation, with silica gel chromatography (eluant, eluent:The ethyl acetate of 0-60% in petroleum ether) Obtain 400mg white foam solid 4h (yields:66.2%).
Step 9:(S) -3- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazole -5- bases) -3- (3- ((S) -2- Ketone group tetrahydrofuran -3- bases) urea groups) propionamide preparation
Compound 4h (280mg, 0.378mmol) is dissolved in dichloromethane (7mL), trifluoroacetic acid is added at room temperature The tri isopropyl silane of (3.88g, 34.04mmol), catalytic amount, are stirred at room temperature reaction 2.5h, and vacuum distillation removes solvent plus two Chloromethanes and water dissolution, liquid separation, organic phase water extracted several times merge water phase, then are washed for several times with dichloromethane, and water phase is frozen Dry, preparation liquid phase purifies to obtain compound 4.1H NMR(500MHz,D2O)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]+.
32 1- of embodiment (3- ((S) -3- amino -1- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazoles -5- Base) -3- propoxyl group) urea groups) and cyclopropane -1- formic acid preparation
Step 5a 1- (3- ((S) -1- (3- ((R) -2- (tert-butoxy) -1- ((tertbutyloxycarbonyl) amino) ethyl) -1, 2,4- oxadiazole -5- bases) -3- ketone groups -3- (tritylamino) propyl) urea groups) and cyclopropane -1- methyl formates preparation
Similar method synthesising title compound is prepared using with compound 4 in embodiment 31, wherein with 1- amino rings (S) -2- ketone group -3- amido tetrahydrofurans in 31 step 4i of propane -1- methyl formate hydrochlorides alternative embodiment.
Step 5b:1- (3- ((S) -1- (3- ((R) -2- (tert-butoxy) -1- ((tertbutyloxycarbonyl) amino) ethyl) -1, 2,4- oxadiazole -5- bases) -3- ketone groups -3- (tritylamino) propyl) urea groups) and cyclopropane -1- formic acid preparation
Compound 5a (770mg, 1.02mmol) is dissolved in tetrahydrofuran (6mL), a hydronium(ion) lithia is added at room temperature Water (1.1mL) solution of (85mg, 2.04mmol), is stirred at reflux reaction 3h, and reaction solution vacuum distillation removes tetrahydrofuran, adds second Acetoacetic ester and water dissolution, liquid separation, ethyl acetate layer use 1M citric acids, saturated common salt water washing for several times successively, and anhydrous sodium sulfate is dry It is dry, vacuum distillation, with silica gel chromatography (eluant, eluent:The ethyl acetate of 0-50% in petroleum ether, 0-10% in dichloromethane Methanol), obtain 180mg white solid 5b (yields:23.8%).
Step 5c:1- (3- ((S) -3- amino -1- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazoles -5- Base) -3- propoxyl group) urea groups) and cyclopropane -1- formic acid preparation
Compound 5b (180mg, 0.243mmol) is dissolved in dichloromethane (3mL), sequentially adds trifluoroacetic acid at room temperature The tri isopropyl silane of (1.66g, 14.58mmol) and catalytic amount, are stirred to react 4h, and reaction solution vacuum distillation adds methylene chloride With water dissolution, liquid separation, organic phase water extracted several times merge water phase, then are washed for several times with dichloromethane, prepared by water phase freeze-drying Liquid phase purifies to obtain compound 32.1H NMR(500MHz,D2O)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]+.
33 1- of embodiment (3- ((R) -3- amino -1- (3- ((S) -1- amino -2- ethoxys) -1,2,4- oxadiazoles -5- Base) -3- propoxyl group) urea groups) and cyclopropane -1- formic acid preparation
Similar method synthesising title compound is prepared using with compound 5 in embodiment 32, wherein using Boc-D-Ser (tBu) Boc-L-Ser (tBu)-OH in-OH alternative embodiments 32, in Fmoc-D-Asn (Trt)-OH alternative embodiments 5 Fmoc-L-Asn(Trt)-OH.Crude product purifies to obtain title compound by preparing liquid phase.1H NMR(400MHz,D2O):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]+
34 1- of embodiment (3- ((S) -1- (3- ((R) -1- amino -2- ethoxys) -1,2,4- oxadiazole -5- bases) -3- carboxylics Propyl) urea groups) cyclopropane -1- formic acid preparation
Similar method synthesising title compound is prepared using with compound 5 in embodiment 32, wherein using Fmoc-L- Glu(OtBu) Fmoc-L-Asn (Trt)-OH in-OH alternative embodiments 32.Crude product purifies to obtain title compound by preparing liquid phase Object.1H NMR(300MHz,D2O):δ 5.02 (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]+
Embodiment 35 (2S, 3R) -2- (3- ((S) -3- amino -1- (3- (1- amino -3- (hydroxymethyl) cyclobutyl) -1, 2,4- oxadiazole -5- bases) -3- oxopropyls) urea groups) and -3-hydroxybutyrate preparation
Step 1 (S)-(fluoro- 1,4- diketos -4- (tritylamino) the butyl- 2- yls of 1-) carbamic acid ((9H- fluorenes -9- Base) methyl) ester preparation
Fmoc-Asn (Trt)-OH (5.0g, 8.380mmol) is dissolved in dichloromethane (84mL), is slowly added at room temperature DAST (1.62g, 10.056mmol) is stirred at room temperature reaction 15min, reaction solution is poured into ice water and is quenched, liquid separation, dichloromethane Layer is washed with water for several times, and anhydrous sodium sulfate drying, vacuum distillation obtains pale yellow oily liquid, and a small amount of dichloromethane dissolving is added, A large amount of n-hexane recrystallizations, filtering, filter cake are washed with a small amount of n-hexane, are evaporated under reduced pressure, and obtain 4.5g white solid 8i (yields: 89.8%).
Step 2:The preparation of 1- amino -3- (methylol) cyclobutane -1- methyl formates
Compound 8a (4g, 27.56mmol) is dissolved in methanol (40mL), dichloro is slowly added dropwise at 0 DEG C in ice bath cooling Sulfoxide (6.5g, 55.13mmol), is heated to back flow reaction 4h after being added dropwise.Vacuum distillation removes solvent and obtains faint yellow oily Liquid directly carries out next step reaction.
Step 3:The preparation of 1- ((tertbutyloxycarbonyl) amino) -3- (methylol) cyclobutane -1- methyl formates
Compound 8b (4g, 25.14mmol) is dissolved in tetrahydrofuran (20mL), at room temperature be added sodium bicarbonate (8.3g, Water (40mL) solution 88.00mmol), ice bath cooling, is added di-tert-butyl dicarbonate (5.7g, 23.88mmol) at 0 DEG C Reaction is stirred at room temperature overnight in tetrahydrofuran (20mL) solution, and vacuum distillation removes reaction dissolvent, with ethyl acetate and water dissolution, Liquid separation, for several times, vacuum distillation, to obtain with silica gel chromatography 3.7g whites solid for organic phase washed with water, saturated common salt water washing Body 8c (yields:56.9%).
Step 4:1- ((tertbutyloxycarbonyl) amino) -3- (tertiary butyl dimethyl Si methyl) cyclobutane -1- methyl formates Preparation
Compound 8c (700mg, 2.70mmol) is dissolved in DMF (7mL), ice bath cooling sequentially adds imidazoles at -5 DEG C (460mg, 6.75mmol), DMF (3.5mL) solution of tert-butyl chloro-silicane (610mg, 4.05mmol).It is stirred at room temperature 4h is reacted, reaction solution adds ethyl acetate to dilute, and uses 1M citric acids, saturated sodium bicarbonate, water washing for several times successively, anhydrous sodium sulfate It is dry, vacuum distillation, with silica gel chromatography (eluant, eluent:The ethyl acetate of 0-50% in petroleum ether) obtain 900mg colorless oils Shape liquid 8d (yields:89.3%).
Step 5:The system of 1- ((tertbutyloxycarbonyl) amino) -3- (tertiary butyl dimethyl Si methyl) cyclobutane -1- formic acid It is standby
Compound 8d (1.6g, 4.283mmol) is dissolved in methanol (16mL), be added a hydronium(ion) lithia (270mg, Water (3.2mL) solution 6.425mmol), is stirred at room temperature reaction 3h, and vacuum distillation removes reaction dissolvent, ethyl acetate and water-soluble Solution, liquid separation, organic phase water extracted several times merge water phase, sour with 1M citric acid solutions tune, then are extracted with ethyl acetate, and merge Organic phase, anhydrous Na2SO4Dry, vacuum distillation obtains 1.42g colourless oil liquid 8e (yields:92.2%).
Step 6:1- ((tertbutyloxycarbonyl) amino) -3- (tertiary butyl dimethyl Si methyl) cyclobutane -1- formamides It prepares
Compound 8e (2.6g, 7.231mmol) is dissolved in tetrahydrofuran (26mL), be added N-methylmorpholine (880mg, 8.677mmol), there is fluffy solid generation;Ice bath cools down, and ethyl chloroformate (940mg, 8.677mmol) is added at -5 DEG C, has A large amount of solids are precipitated, and ice bath stirring reacts 0.5h;It is slowly added to the ammonia spirit of 3.9mL 25%, ice bath stirring is reacted 3h, subtracted Solvent, remaining solid ethyl acetate and water-soluble clear, liquid separation is distilled off in pressure.Organic phase uses 1M citric acids, unsaturated carbonate successively For several times, anhydrous sodium sulfate is dry, is evaporated under reduced pressure, and obtains 1.5g white solid 8f (yields for hydrogen sodium, water washing:57.9%).
Step 7:The preparation of (3- tertiary butyl dimethyl Sis methyl-1-cyano cyclobutyl) t-butyl carbamate
Compound 8f (1.58g, 4.407mmol) is dissolved in pyridine (16mL), ice bath cooling is slowly added to three at -5 DEG C Fluoroacetic acid acid anhydride (1.39g, 6.611mmol), ice bath stirring react 3h, reaction solution are poured into excessive saturated sodium bicarbonate solution and is quenched It goes out, is extracted with ethyl acetate, organic phase uses 1M citric acids, saturated common salt water washing for several times successively, anhydrous sodium sulfate drying, decompression Distillation, with silica gel chromatography (eluant, eluent:The ethyl acetate of 0-15% in petroleum ether) obtain 1.36g colourless oil liquids 8g (yield:90.7%).
Step 8:(Z)-(3- tertiary butyl dimethyl Sis methyl-1-(N'- hydroxy formamidines base) cyclobutyl) carbamic acid uncle The preparation of butyl ester
Compound 8g (1.3g, 3.817mmol) is dissolved in ethyl alcohol (13mL), sequentially add at room temperature potassium carbonate (791mg, Water (2.6mL) solution, hydroxylamine hydrochloride (398mg, 5.726mmol) 5.726mmol), are stirred at reflux reaction 2h, and vacuum distillation removes Solvent is removed, adds ethyl acetate and water-soluble clear, liquid separation, organic phase for several times with saturated common salt water washing, depressurize by anhydrous sodium sulfate drying Distillation, with silica gel chromatography (eluant, eluent:The ethyl acetate of 0-20% in petroleum ether) obtain 700mg white solid 8h (yields: 49.3%).
Step 9:(Z)-(1-(N'-((N2(((9H- fluorenes -9- bases) methoxyl group) carbonyl)-N4Trityl-L- asparagus fern acyls Amido) oxygroup) hydroxy formamidine base) -3- (tertiary butyl dimethyl Si methyl) cyclobutyl) t-butyl carbamate preparation
Compound 8h (300mg, 0.803mmol) is dissolved in tetrahydrofuran (3mL).N-methylmorpholine is sequentially added at room temperature Tetrahydrofuran (10mL) solution of (240mg, 2.409mmol) and compound 8i (960mg, 1.606mmol), are stirred at room temperature reaction Overnight, reaction solution vacuum distillation removes tetrahydrofuran, adds ethyl acetate dissolved clarification, for several times with saturated common salt water washing, anhydrous slufuric acid Sodium is dried, vacuum distillation, with silica gel chromatography (eluant, eluent:The ethyl acetate of 0-15% in petroleum ether) 350mg it is colourless Grease 8j (yields:55.8%).
Step 10:(S)-(1- (5- (1- ((((9H- fluorenes -9- bases) methoxyl group) carbonyl) amino) -3- ketone groups -3- (triphen first Base amino) propyl) -1,2,4- oxadiazole -3- bases) -3- (((t-Butyldimethylsilyl) oxygroup) methyl) cyclobutyl) amino first The preparation of tert-butyl acrylate
Compound 8j (1g, 1.050mmol) is dissolved intBuOH(20mL).At room temperature be added sodium acetate (129mg, 1.575mmol), it is stirred at reflux reaction 3h, reaction solution vacuum distillation removes solvent, adds ethyl acetate dissolved clarification, use saturated salt solution Washing for several times, anhydrous sodium sulfate drying, vacuum distillation, with silica gel chromatography (eluant, eluent:The acetic acid of 0-20% in petroleum ether Ethyl ester) obtain 520mg colorless oil 8k (yields:53%).
Step 11:(S)-(1- (5- (1- amino -3- ketone groups -3- (tritylamino) propyl) -1,2,4- oxadiazoles -3- Base) -3- (((t-Butyldimethylsilyl) oxygroup) methyl) cyclobutyl) t-butyl carbamate preparation
Compound 8k (500mg, 0.535mmol) is dissolved in dichloromethane (5mL), at room temperature be added diethylamine (783mg, 10.7mmol), reaction 4h is stirred at room temperature.The completion of reaction is analyzed to identify by TLC.Reaction solution vacuum distillation removes solvent, uses Silica gel chromatography (eluant, eluent:The ethyl acetate of 0-40% in petroleum ether), obtain 330mg colorless oil 8l (yields: 86.6%).
Step 12:N- (((S) -1- (3- (1- ((tertbutyloxycarbonyl) amino) -3- (((t-Butyldimethylsilyl) oxygroup) Methyl) cyclobutyl) -1,2,4- oxadiazole -5- bases) -3- ketone groups -3- (tritylamino) propyl) carbamyl)-O- (tertiary fourths Base)-L-threonine the tert-butyl ester preparation
Compound 8l (270mg, 0.379mmol) is dissolved in tetrahydrofuran (5mL), sequentially adds compound 2b at room temperature Reaction is stirred at room temperature overnight in (180mg, 0.455mmol), triethylamine (77mg, 0.758mmol), and reaction solution vacuum distillation removes Solvent adds ethyl acetate dissolved clarification, uses 1M citric acids, saturated common salt water washing for several times successively, and anhydrous sodium sulfate drying, decompression is steamed It evaporates, with silica gel chromatography (eluant, eluent:The ethyl acetate of 0-15% in petroleum ether), obtain 400mg white solid 8n (yields: 95.1%).
Step 13:(2S, 3R) -2- (3- ((S) -3- amino -1- (3- (1- amino -3- (hydroxymethyl) cyclobutyl) -1,2, 4- oxadiazole -5- bases) -3- oxopropyls) urea groups) and -3-hydroxybutyrate preparation
Compound 8n (350mg, 0.361mmol) is dissolved in dichloromethane (7mL), trifluoroacetic acid is added at room temperature The tri isopropyl silane of (2.06g, 18.05mmol) and catalytic amount, are stirred at room temperature reaction 5h, and reaction solution vacuum distillation removes molten Agent adds methylene chloride and merges water phase with water-soluble clear, liquid separation, organic phase water extracted several times, then washed for several times with dichloromethane, Water phase is lyophilized, and prepares liquid phase purifying, obtains 8 (yield of 70mg white solids:37.8%).1H NMR(400MHz,D2O)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]+.
Embodiment 36:(S) -4- (3- (1- amino -3- (methylol) cyclobutyl) -1,2,4- oxadiazole -5- bases) -4- (3- ((1S, 2R) -1- carboxyl -2- hydroxypropyls) urea groups) butyric acid preparation
Similar method synthesising title compound is prepared using with compound in embodiment 35.Wherein use Fmoc-Glu (OtBu)-OH replaces Fmoc-Asn (Trt)-OH.1H NMR(400MHz,D2O) δ 5.02-4.97 (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]+.
Embodiment 37:(S) -3- (3- (1- amino -3- (methylol) cyclobutyl) -1,2,4- oxadiazole -5- bases) -3- (3- ((S) -1- carboxyl -2- ethoxys) urea groups) propionic acid preparation
Similar method synthesising title compound is prepared using with compound in embodiment 35.Wherein use Fmoc-Asp (OtBu) Fmoc-Asn (Trt)-OH in-OH alternative embodiments 8, with H-Ser (OtBu)-OtH- in Bu alternative embodiments 35 Thr(OtBu)-OtBu。1H NMR(400MHz,D2O) δ 5.35 (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]+.
Embodiment 38 (2S, 3S) -2- (3- ((S) -4- amino -1- (5- (1- amino cyclopropyl) -1,3,4- oxadiazoles -2- Base) -4- oxos butyl) urea groups) and -3-hydroxybutyrate preparation
The preparation method is the same as that of Example 12 preparation method, by (2S, 3S) -3- hydroxy tetrahydro pyrroles in 12 step 1 of embodiment It coughs up -2- formic acid and has been substituted for 1- amino-cyclopropane -1- formic acid, and O- (tertiary butyl)-L-serine tert-butyl ester is substituted for O- (tertiary butyl)-L-threonine tert-butyl ester, can be made target compound.
Comparative example 1
Following formula representative is prepared according to method disclosed in embodiment 2 in WO2015/033301 (PCT/IB2014/064281) Compound (compound A), and by hydrogen spectrum and Mass Spectrometric Identification,
The Pharmacokinetic Characteristics of compound A are tested using the method for following experimental example 1 and 2 and in colon cancer CT26 Tumor killing effect in cell subcutaneous transplantation knurl model, experimental result show that the bioavilability (F) and tumour inhibiting rate of compound A is weak In some compounds of the present invention.
In addition, the present inventor synthesizes also according to method disclosed in WO2015/033301 and tests WO2015/ Compound No.12 in 033301 table 3, the results show that the bioavilability of Compound No.12 and tumour inhibiting rate are markedly less than The compound of the present invention and compound A.
1 drug metabolism of experimental example is tested
1 experiment material
1.1 compound
The compound of embodiment 2 in the compound of the present invention and WO2015/033301 for being prepared using above example (" compound A ") carries out the experiment.Oral drugs physiological saline solution, is made 0.5mg/mL clear solutions, and intravenous pharmacy is used 0.1mg/mL clear solutions are made in physiological saline solution.
1.2 animal
Male BALB/c mouse, every group each 3, weight 18-22g, Shanghai western Poole-Bi Kai experimental animals Co., Ltd carries For.
Water supply after 2h, 4h is administered in the environment laundering period that 2~4 days are given before test mice experiment, fasting 8-12h before being administered Food is given afterwards.
1.3 reagent
Methanol (chromatographically pure):Spectrum companies produce;
Acetonitrile (chromatographically pure):Spectrum companies produce;
Remaining reagent is that commercially available analysis is pure.
1.4 instrument
The triple level four bars LC-MS instrument of 4500 types of American AB company API are furnished with electric spray ion source (ESI), LC- 30AD double pumps;SIL-30AC autosamplers;CTO-30AC column ovens;DGU-20A3R degassers;Analyst QS A01.01 Chromatographic work station;Milli-Q Superpure water machines (Millipore Inc);Qilinbeier Vortex-5 oscillators;HITACHI Ⅹ II table-type high-speed refrigerated centrifuges of CF16R.
2 experimental methods
1) mouse fasting but can free water after 12 hours, take 0 moment blank plasma;
2) mouse in step 1) is taken, gavage (intragastric administration, IG) give untested compound 10mg/kg;Vein (IV) give untested compound 1mg/kg;
3) 5min, 15min, 30min, 1h, 2h, 4h, 8h, 10h after gavage are set from eyeground vein clump continuous blood sampling for 24 hours In being distributed in the EP pipes of heparin, upper plasma is taken after 8000rpm/min centrifugations 5min, -20 DEG C freeze, and wait for LC-MS/MS
Analysis;
4) the blood concentration-time data obtained by step 3) are asked using WinNonlin softwares and calculate pharmacokinetics ginseng Number;
3 experimental results:
Pharmacokinetic studies data are as shown in table 1, the results showed that the oral compound for giving mouse above example Afterwards, all have in animal blood plasma certain exposed amount and suitable half-life period, especially embodiment 4, embodiment 5, embodiment 8, The compound of embodiment 6 and embodiment 22 has extraordinary half-life period, area under the curve and bioavilability, druggability It is good, there is good potential applicability in clinical practice.
The pharmacokinetic data of 1 compound of the embodiment of the present invention of table
Effect experiment in 2 body of experimental example
1, experiment material
1.1 compound
The experiment is carried out using the compound prepared according to embodiments of the present invention.Negative control group gives physiological saline.It adopts With oral medication, compound physiological saline solution is tested, 2mg/mL clear solutions are made.
1.2 animal
Female BAl BIc/c mouse, every group each 3, weight 18-22g, Shanghai western Poole-Bi Kai experimental animals Co., Ltd carries For.The environment laundering period that 2~4 days are given before test mice experiment, fasting 8-12h before being administered are administered water supply after 2h, are given after 4h Food.
1.3 positive control
Compound A is prepared according to method disclosed in WO2015/033301 embodiments 2.
2, experimental method
Wait for that tumour growth to average external volume is 40mm after inoculating cell3(first round experiment) or 50mm3(the second wheel experiment) Afterwards, animal is grouped at random, every group 6,20mg/kg, once a day, successive administration 14 days is administered orally in each test group.It investigates Whether the variation of experimental animal weight and tumour growth are suppressed or delay.Vernier caliper measurement diameter of tumor is used three-times-weekly.
The calculation formula of gross tumor volume is:V=0.5a × b2, a and b indicate the major diameter and minor axis of tumour respectively.
The calculation formula of Relative tumor proliferation rate T/C (%) is:T/C=TRTV/CRTV100% (T of XRTV:Treatment group RTV; CRTV:Solvent control group RTV).Relative tumour volume (relative tumor volume, RTV) is calculated according to measurement result, RTV=Vt/V0, wherein V0Gross tumor volume when starting for experiment, gross tumor volume when Vt is measured each time.
The calculation formula of inhibition rate of tumor growth TGI (%) is:TGI=[1- (certain processing group average knurls at the end of being administered Product-processing the group starts mean tumor volume when administration)/(mean tumor volume-solvent control group is opened when solvent control group treatment end Mean tumor volume when beginning to treat)] X 100%.
3, experiment conclusion
3.1 changes of weight situations
The compound of the present invention to homology transplantable tumor under mouse junction cancer CT26 cell skins BALB/C mice model body For weight without influence, table 2 gives the influence to weight after embodiment 4, embodiment 5, embodiment 35 and compound A are administered, and shows Control group and each administration group the weight of animals gradually increase during administration, have preferable tolerance.
Influence after the administration of 2 the compound of the present invention of table to mouse weight
3.2 antitumor drug effect evaluation indexes
Evaluating drug effect index is as shown in table 3, and first round experiment starts after administration the 15th day, and solvent control group mice with tumor is put down Equal knurl product reaches 3672mm3, the tumor bulk averaged value of other each administration group mice with tumor is respectively less than control group tumor bulk averaged value, It is 52.4% that wherein T/C value of the compound of embodiment 4 at the 15th day, which is 41.5%, TGI values, indicates it to CT26 colon cancers Cell transplantation tumor has significant inhibiting effect, with obvious effects to be better than compound A.Second wheel experiment starts after administration the 14th day, The mean tumor volume of solvent control group mice with tumor reaches 1524mm3, the tumor bulk averaged value of other each tested material group mice with tumor is equal Less than control group tumor bulk averaged value, wherein T/C value of the compound of embodiment 5 at the 14th day is that 39.0%, TGI values are 63.7%, indicate that it has significant inhibiting effect to CT26 colon cancer cell transplantable tumors.
3 antitumor drug effect evaluation index of table
Although being described in detail above to the present invention, however it is understood by skilled practitioners that without departing from this hair The present invention can be carry out various modifications and be changed under the premise of bright spirit and scope.The interest field of the present invention is not limited to It is described in detail made by above, and claims should be belonged to.

Claims (10)

1. a kind of general formula A compounds represented or its stereoisomer or pharmaceutically acceptable salt, solvate, crystallization or preceding Medicine,
Wherein:
X and Y is each independently selected from N, O and S, and when X is N, and Y is selected from O and S;When X is O or S, Y N;When Y is N, X is selected from O And S;When Y is O or S, X N;
When X is N, and Y is selected from O and S or Y is O or S, when X is N, groupForWhen X is O or S, Y N When or Y be N, X be selected from O and S when, groupFor
R1Side chain selected from amino acid Ser and Thr;
R2Selected from amino acid Ser and Thr residue;
R3Selected from H ,-(CH2)mC(O)OR4、-(CH2)mC(O)N(R5)(R6) and-(CH2)mCN, wherein R4、R5、R6Each independently Selected from H and alkyl, m 0,1,2,3 or 4;With
N is 1,2,3 or 4.
2. general formula A compounds represented according to claim 1 or its stereoisomer or pharmaceutically acceptable salt, molten Object, crystallization or prodrug are closed in agent, and formula of A has the structure of following general formula II,
Wherein:Q is selected from O and S;R1、R2、R3Definition as described in the appended claim 1.
3. general formula A compounds represented according to claim 1 or its stereoisomer or pharmaceutically acceptable salt, molten Object, crystallization or prodrug are closed in agent, and formula of A has the structure of following general formula I,
Wherein:Q is selected from O and S;R1、R2、R3Definition as described in the appended claim 1.
4. a kind of Formula B compound represented or its stereoisomer or pharmaceutically acceptable salt, solvate, crystallization or preceding Medicine,
Wherein:
X and Y is each independently selected from N, O and S, and when X is N, and Y is selected from O and S;When X is O or S, Y N;When Y is N, X is selected from O And S;When Y is O or S, X N;
When X is N, and Y is selected from O and S or Y is O or S, when X is N, groupForWhen X is O or S, Y N When or Y be N, X be selected from O and S when, groupFor
R7Side chain selected from amino acid Asn, Gln, Asp, Glu;
R8Selected from amino acid Ser and Thr residue;
R9、R10It is each independently selected from H, alkyl and alkyl hydroxy, R9、R10It is asynchronously H, and
R9Or R10With azacycloalkyl or nitrogen oxacycloalkyl, the azacycloalkyl are formed together with the amino connected on its α carbon potential Base or nitrogen oxacycloalkyl are optionally by one or more substitutions for being selected from hydroxyl, hydroxy alkyl, cyano, amino, halogen, nitro Base is replaced;Or
R9、R10C atoms connected to them form naphthenic base or oxacycloalkyl, the naphthenic base or oxacycloalkyl together Optionally replaced by one or more substituent groups selected from hydroxyl, hydroxy alkyl, cyano, amino, halogen, nitro.
5. a kind of general formula C compounds represented or its stereoisomer or pharmaceutically acceptable salt, solvate, crystallization or preceding Medicine,
Wherein:
X and Y is each independently selected from N, O and S, and when X is N, and Y is selected from O and S;When X is O or S, Y N;When Y is N, X is selected from O And S;When Y is O or S, X N;
When X is N, and Y is selected from O and S or Y is O or S, when X is N, groupForWhen X is O or S, Y N When or Y be N, X be selected from O and S when, groupFor
R11Side chain selected from amino acid Ser and Thr;
R12Selected from amino acid Asn, Gln, Asp, Glu side chain;
R15Selected from H and alkyl;
R13、R14It is each independently selected from H, alkyl, R13、R14It is asynchronously H, and
R13、R14C atoms connected to them form naphthenic base together;Or
R13Or R14Azacycloalkyl is formed with together with the imino group of its α carbon potential connection;Or R14With R15And their atoms for being connected The lactone naphthenic base optionally replaced is formed together, wherein the naphthenic base, azacycloalkyl or lactone naphthenic base are optionally by one A or multiple substituent groups selected from hydroxyl, hydroxy alkyl, cyano, amino, halogen, nitro are replaced.
6. compound or its stereoisomer or pharmaceutically acceptable salt according to claim 1-5 any one, molten Object, crystallization or prodrug are closed in agent, wherein the compound is compound selected from the following:
7. a kind of preparing compound or its stereoisomer or pharmaceutically acceptable shown in general formula A according to claim 2 Salt, solvate, crystallization or prodrug preparation method, the described method comprises the following steps:
(1) formula ia compounds pass through condensation reaction production iic compounds with formula iib compounds;
(2) formula iic compounds are cyclized to obtain formula iid compounds;
(3) formula iid compounds are reacted with aminated compounds, obtain formula iie compounds;
(4) formula iie compounds pass through nucleophilic substitution with formula if compounds, obtain formula iig compounds;
(5) in acid condition, formula iig compounds generation hydrolysis obtains Formula II compound;
Wherein:Pg1Represent R1Protecting group;Pg2Represent R2Protecting group;Pg3Represent R3Protecting group or be not present;Pg4、Pg5Generation Table amino protecting group.
8. a kind of preparing compound or its stereoisomer or pharmaceutically acceptable shown in general formula A according to claim 3 Salt, solvate, crystallization or prodrug preparation method, the described method comprises the following steps:
(1) formula ia compounds pass through condensation reaction production ic compounds with formula ib compounds;
(2) formula ic compounds are cyclized to obtain formula id compounds;
(3) formula id compounds are reacted with aminated compounds, obtain formula ie compounds;
(4) with formula if compounds nucleophilic substitution occurs for formula ie compounds, obtains formula ig compounds;
(5) in acid condition, formula ig compounds generation hydrolysis obtains compound of formula I;
Wherein:Pg1Represent R1Protecting group;Pg2Represent R2Protecting group;Pg3Represent R3Protecting group or be not present;Pg4、Pg5Generation Table amino protecting group.
9. a kind of pharmaceutical composition, it includes the compounds or its stereoisomer or medicine described in claim 1-6 any one Acceptable salt, solvate, crystallization or prodrug and pharmaceutically acceptable carrier on.
10. compound or its stereoisomer or pharmaceutically acceptable salt according to claim 1-6 any one, molten Agent is closed object, crystallization or prodrug or pharmaceutical composition according to claim 9 and is being prepared for treating cancer or infection class disease Application in the drug of disease.
CN201810375602.3A 2017-04-26 2018-04-25 Heterocyclic compounds as PD-L1 inhibitors Active CN108794422B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710280707 2017-04-26
CN2017102807076 2017-04-26

Publications (2)

Publication Number Publication Date
CN108794422A true CN108794422A (en) 2018-11-13
CN108794422B CN108794422B (en) 2022-07-01

Family

ID=63920214

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810375602.3A Active CN108794422B (en) 2017-04-26 2018-04-25 Heterocyclic compounds as PD-L1 inhibitors

Country Status (3)

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

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102401963B1 (en) 2016-06-27 2022-05-25 케모센트릭스, 인크. Immunomodulatory compounds
WO2018200571A1 (en) 2017-04-25 2018-11-01 Arbutus Biopharma Corporation Substituted 2,3-dihydro-1h-indene analogs and methods using same
JP7185681B2 (en) 2017-07-28 2022-12-07 ケモセントリックス,インコーポレイティド immunomodulatory compounds
AU2018313744C1 (en) 2017-08-08 2023-07-27 Chemocentryx, Inc. Macrocyclic immunomodulators
WO2019160882A1 (en) 2018-02-13 2019-08-22 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
WO2019165043A2 (en) 2018-02-22 2019-08-29 Chemocentryx, Inc. Indane-amines as pd-l1 antagonists
US10899735B2 (en) 2018-04-19 2021-01-26 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
JP7105359B2 (en) 2018-07-13 2022-07-22 ギリアード サイエンシーズ, インコーポレイテッド PD-1/PD-L1 inhibitor
US11236085B2 (en) 2018-10-24 2022-02-01 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
TW202024042A (en) * 2018-10-25 2020-07-01 大陸商南京聖和藥業股份有限公司 1,3,4-oxadiazole-2-cyclobutyl compounds, preparation method therefor and application thereof
MX2021013819A (en) 2019-05-15 2022-02-10 Chemocentryx Inc Triaryl compounds for treatment of pd-l1 diseases.
WO2020257549A2 (en) 2019-06-20 2020-12-24 Chemocentryx, Inc. Compounds for treatment of pd-l1 diseases
CN114206338A (en) 2019-07-10 2022-03-18 凯莫森特里克斯股份有限公司 Indanes as PD-L1 inhibitors
CN114555080A (en) 2019-10-16 2022-05-27 凯莫森特里克斯股份有限公司 Heteroaryl biphenylamines useful for the treatment of PD-L1 disease
EP4045036A4 (en) 2019-10-16 2023-11-15 ChemoCentryx, Inc. Heteroaryl-biphenyl amides for the treatment of pd-l1 diseases

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

Also Published As

Publication number Publication date
WO2018196768A1 (en) 2018-11-01
TW201841896A (en) 2018-12-01
CN108794422B (en) 2022-07-01

Similar Documents

Publication Publication Date Title
CN108794422A (en) Heterocycle compound as PD-L1 inhibitor
CN108863963B (en) Heterocyclic compounds as PD-L1 inhibitors
CN104185477B (en) Cytotoxic peptides and antibody drug conjugates thereof
JP3579751B2 (en) Human cancer-inhibiting pentapeptide amides and esters
CN103492370B (en) Substituted dicarbamylamine and diaminourea formonitrile HCN pyrimidine, a combination thereof thing, and the method treated with it
CN108137607A (en) Suitable for treating compound and composition with the relevant illnesss of NTRK
WO2022166890A1 (en) Substituted pyridazine phenol derivatives
CN105164117B (en) Dipeptides and tripeptide epoxy ketone protease inhibitors
CN106008569A (en) Boronates as arginase inhibitors, compositions and applications thereof
AU2022258377B2 (en) Ring-modified proline short peptide compound and use thereof
CA3063872A1 (en) Bis-octahydrophenanthrene carboxamides and protein conjugates thereof
CN105073730A (en) Monocyclic pyridine derivative
WO2003059866A1 (en) Geranyl compounds
CN107614504A (en) (6S, 9aS) N benzyls 6 [(4 hydroxy phenyl) methyl] 4,7 dioxies 8 ({ base of 6 [base of 3 (base of piperazine 1) azetidine 1] pyridine 2 } methyl) 2 (the third 2 base of alkene 1) octahydro 1H pyrazines simultaneously [2,1 c] [1,2,4] crystal of the benzamide compound of triazine 1
CN113614095A (en) Alkylboronic acids as arginase inhibitors
JPH06509324A (en) Novel peptides, their production and use
JPH05505820A (en) new compound
CN109071525A (en) dimeric compounds
CN104974221B (en) Dipeptide and tripeptide proteasome inhibitors and preparation method and pharmaceutical application thereof
CN118055929A (en) 2, 6-Piperidinedione compound and application thereof
JP4842254B2 (en) PAR-2 agonist
CA3076794A1 (en) Agents inhibiting tctp protein for the treatment of proliferative diseases, infectious diseases, allergies, inflammations and/or asthma
TW202110844A (en) Dispiro-diketopiperazine compounds
JP6601220B2 (en) Sugar amino acids and their uses
TW202342060A (en) Lonp1 inhibitor compounds, uses and methods

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant