WO2019144912A1 - β-内酰胺酶抑制剂及其用途 - Google Patents

β-内酰胺酶抑制剂及其用途 Download PDF

Info

Publication number
WO2019144912A1
WO2019144912A1 PCT/CN2019/073000 CN2019073000W WO2019144912A1 WO 2019144912 A1 WO2019144912 A1 WO 2019144912A1 CN 2019073000 W CN2019073000 W CN 2019073000W WO 2019144912 A1 WO2019144912 A1 WO 2019144912A1
Authority
WO
WIPO (PCT)
Prior art keywords
oxo
octane
cyclopropane
bicyclo
diazaspiro
Prior art date
Application number
PCT/CN2019/073000
Other languages
English (en)
French (fr)
Inventor
吴予川
黄少强
陈曦
胡永韩
刘霄
Original Assignee
苏州信诺维医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 苏州信诺维医药科技有限公司 filed Critical 苏州信诺维医药科技有限公司
Priority to CN201980010129.1A priority Critical patent/CN111670187B/zh
Priority to EP19743504.3A priority patent/EP3744722A4/en
Priority to JP2020540565A priority patent/JP7273420B2/ja
Priority to KR1020207024397A priority patent/KR20200113247A/ko
Priority to CA3089150A priority patent/CA3089150A1/en
Priority to AU2019211693A priority patent/AU2019211693B2/en
Publication of WO2019144912A1 publication Critical patent/WO2019144912A1/zh
Priority to US16/998,577 priority patent/US11078202B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • 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
    • A61K31/425Thiazoles
    • A61K31/429Thiazoles condensed with heterocyclic ring systems
    • A61K31/43Compounds containing 4-thia-1-azabicyclo [3.2.0] heptane ring systems, i.e. compounds containing a ring system of the formula, e.g. penicillins, penems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/468-Azabicyclo [3.2.1] octane; Derivatives thereof, e.g. atropine, cocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4995Pyrazines or piperazines forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems

Definitions

  • the present invention relates to a beta-lactamase inhibitor and a process for the preparation thereof.
  • the invention further relates to a pharmaceutical composition comprising a beta-lactamase inhibitor and to the use thereof.
  • Beta-lactam antibiotics are the first antibiotics cited in the clinic. Since the successful application of penicillin G as the first ⁇ -lactam antibiotic to the clinic, ⁇ -lactam antibiotics have developed rapidly. Different structure types of ⁇ -lactam antibiotics have been developed and applied in a large number of clinical applications, and have achieved good results. However, since bacterial cells can produce ⁇ -lactamase, the antibiotics are inactivated, causing the bacteria to develop resistance to ⁇ -lactam antibiotics.
  • the ⁇ -lactamase is an enzyme that catalyzes the hydrolysis of the ⁇ -lactam ring, which inactivates the antibacterial activity of the ⁇ -lactam antibiotic and allows the bacteria to be resistant to the ⁇ -lactam antibiotic.
  • the ⁇ -lactamase can be classified into A, B, C, and D depending on the amino acid sequence difference in the molecular structure.
  • Class A ⁇ -lactamases preferably hydrolyze penicillin antibiotics
  • Class B ⁇ -lactamases can hydrolyze various ⁇ -lactam antibiotics, including carbapenems
  • Class C ⁇ -lactamases are more effective.
  • Hydrolyzed cephalosporin antibiotics while D-type ⁇ -lactamases are more prone to hydrolyzing oxacillin and o-chloropenicillin.
  • the resistance of bacteria, especially Gram-negative bacteria, to ⁇ -lactam antibiotics is usually mediated by ⁇ -lactamase.
  • Inhibition of ⁇ -lactamase can delay or inhibit the degradation of ⁇ -lactam antibiotics and restore the sensitivity of bacteria producing ⁇ -lactam antibiotic resistance to ⁇ -lactam antibiotics.
  • ⁇ -lactamase can inactivate the hydrolysis activity of ⁇ -lactam antibiotics, thereby enhancing the bacteria against ⁇ -lactam.
  • the sensitivity of antibiotics reduces or overcomes the problem of drug resistance.
  • bacteriostatic beta-lactamase inhibitors such as those disclosed in WO2013149121A1, WO2014141132A1, US20130296290A1, WO2013030735A1, WO2015110963A1, WO2015150890A1, WO2015159265A1, WO2015173663, WO2015173665A1, WO2017055922A1 [bicyclo[3.2.1] ] Octane compounds.
  • beta-lactamase inhibitors available on the market, such as clavulanic acid, tazobactam, abivudan, Relebactam, and the like.
  • the present invention provides a compound of the formula (I) or an ester thereof, a stereoisomer thereof, and a pharmaceutically acceptable salt thereof:
  • W 1 is selected from the group consisting of an optionally substituted five- or six-membered heteroaryl ring containing O, N and/or S or -C(O)-; and, (i) when W 1 is selected from the group consisting of optionally substituted When a 5- or 6-membered heteroaryl ring of O, N and/or S is substituted, W 1 is optionally substituted by C 1 -C 12 alkyl,
  • W 2 is selected from:
  • R 1 is selected from Wherein R 2 , R 3 , R 4 , R 4 ', R 5 or R 6 are each independently selected from H, C 1 -C 12 alkyl, amino C 1 -C 12 alkyl, C 1 -C 12 alkylamine Base C 1 -C 12 alkyl, Ra and Rb are each independently selected from H, C 1 -C 12 alkyl, OH, -OC 1 -C 12 alkyl, -NH 2 , -NHC 1 -C 12 alkyl, -N(C 1 -C 12 Alkyl) 2 , -SH, -SC 1 -C 12 alkyl, -S(O)C 1 -C 12 alkyl, -S(O 2 )C 1 -C 12 alkyl, -SO 3 H;R 7 and R 7 ' are each independently selected from -NH 2 , -NHC 1 -C 12 alkyl, -N(C 1 -C 12 alkyl
  • R 1 ' is selected from H, C 1 -C 12 alkyl, C 3 -C 8 cycloalkyl, OH, -OC 1 -C 12 alkyl, -NH 2 , -NHC 1 -C 12 alkyl, -N (C 1 -C 12 alkyl) 2 , -SH, -SC 1 -C 12 alkyl, -S(O)C 1 -C 12 alkyl, -S(O 2 )C 1 -C 12 alkyl, -SO 3 H;
  • Z 2 is selected from CR 18 R 19 or NR 20 , and R 18 , R 19 and R 20 are each independently selected from H, NH 2 or
  • R 21 is selected from the group consisting of -NH 2 , -NHC 1 -C 12 alkyl, -N(C 1 -C 12 alkyl) 2 , -O C 1 -C 12 alkyl, -S C 1 -C 12 alkyl;
  • R 22 is selected from the group consisting of NH, -NHC 1 -C 12 alkyl, O, S;
  • t, u and w are each independently selected from 1, 2, 3, 4, 5 or 6, provided that t and u are not 0 at the same time;
  • W 1 when W 1 is selected from -C(O)-, W 2 is selected from -OR 26 , -NHR 27 , wherein R 26 and R 27 are each independently selected from H, C 1 -C 12 alkyl or Wherein Z 5 is selected from CR 28 R 29 or NR 30 , and R 28 , R 29 and R 30 are each independently selected from H, NH 2 or Wherein R 31 is selected from the group consisting of -NH 2 , -NHC 1 -C 12 alkyl, -N(C 1 -C 12 alkyl) 2 , -O C 1 -C 12 alkyl, -S C 1 -C 12 alkyl; R 32 is selected from the group consisting of NH, NC 1 -C 12 alkyl, O, S; x, y and z are each independently selected from 1, 2, 3, 4, 5 or 6, provided that x and y are not simultaneously 0;
  • W 3 is selected from the group consisting of -SO 3 M, -OSO 3 M, -OSO 2 NH 2 , -OPO 3 M, -OCR 33 R 34 CO 2 M, -OCR 35 R 36 SO 3 M, -OCR 37 R 38 PO 3 M; wherein M is selected from H or a pharmaceutically acceptable cation; and R 33 , R 34 , R 35 , R 36 , R 37 and R 38 are each independently selected from H, C 1 -C 12 alkyl, halogen.
  • the present invention provides a process for the preparation of a compound of the formula (I) or a pharmaceutically acceptable salt thereof.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the formula (I) or a pharmaceutically acceptable salt thereof and one or more ⁇ -lactam antibiotics.
  • the present invention provides a method of treating a disease caused by a bacterial infection comprising administering a compound of the formula (I) as described herein in combination with one or more ⁇ -lactam antibiotics A patient or subject in need.
  • alkyl refers to a straight or branched saturated hydrocarbon group having from 1 to 20 carbon atoms.
  • the alkyl group is an alkyl group having from 1 to 12 carbon atoms. More preferably, the alkyl group is an alkyl group having 1 to 6 carbon atoms. Most preferably, the alkyl group is an alkyl group having from 1 to 4 carbon atoms.
  • alkyl groups include, but are not limited to, methyl, ethyl, 1-propyl (n-propyl), 2-propyl (isopropyl), 1-butyl (n-butyl), 2-methyl 1-propyl (isobutyl), 2-butyl (sec-butyl), 2-methyl-2-propyl (tert-butyl), 1-pentyl (n-pentyl), 2-pentyl , 3-pentyl, 2-methyl-2-butyl, 3-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2- Methyl-3-pentyl, 2,3-dimethyl-2-butyl, 3,3-dimethyl-2-butyl, 1-heptyl, 1-octyl
  • amino refers to -NH 2.
  • halogen refers to fluoro, chloro, bromo, iodo.
  • aminoalkyl refers to an alkyl group in which one or more hydrogens are replaced by an amino group.
  • cycloalkyl refers to a monovalent saturated carbocyclic group.
  • the cycloalkyl group is a 3 to 8 membered monocyclic group. More preferably, the cycloalkyl group is a 3- to 6-membered monocyclic group.
  • Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • heteroaryl refers to a 5- or 6-membered, monovalent aromatic radical containing 1-3 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur.
  • heteroaryl groups include: furan ring, pyrrole ring, pyrazole ring, imidazole ring, triazole ring, thiophene ring, thiazole ring, isothiazole ring, oxazole ring, isoxazole ring, 1,2,4-oxine Diazole ring, 1,3,4-oxadiazole ring; pyridine ring, pyrimidinyl group, triazine ring, pyrazine ring, tetrazole ring, pyridazine ring, thiadiazole ring.
  • the heteroaryl group is optionally substituted independently with one or more substituents as described herein.
  • the term "optionally substituted” means that the given structure or group is unsubstituted or that the given structure or group is substituted with one or more specific substituents. Unless otherwise stated, the optional substitution can be substituted at any position of the substituted group.
  • ester refers to an ester of -OSO 3 - or COOH (if present) in the compound of formula (I) which can be esterified with an alcohol; as shown in formula (I) When a hydroxyl group is present in the compound, it can be esterified with an organic acid, an inorganic acid or the like to form an ester. The ester can be hydrolyzed under acidic or basic conditions to form the corresponding acid or alcohol.
  • stereoisomer refers to a compound that has the same chemical composition and connectivity, but whose atoms have different orientations in space, which orientation cannot be interchanged by single bond rotation.
  • Stepoisomers include “diastereomers” and “enantiomers.”
  • Diastereomer refers to a stereoisomer that has two or more centers of chirality and whose molecules are not mirror images of each other. Diastereomers have different physical properties such as melting point, boiling point, spectral properties and reactivity. The mixture of diastereomers can be separated under high resolution analytical procedures such as crystallization, electrophoresis and chromatography.
  • Enantiomer refers to two stereoisomers that are non-overlapping mirror images of each other.
  • salts refers to a pharmaceutically acceptable organic or inorganic salt of a compound of the invention.
  • exemplary salts include, but are not limited to, sulfates, citrates, acetates, oxalates, chlorides, bromides, iodides, nitrates, hydrogen sulfates, phosphates, acid phosphates, isoniazids Acid salt, lactate, salicylate, acid citrate, tartrate, oleate, citrate, pantothenate, hydrogen tartrate, ascorbate, succinate, maleate, dragon Cholate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate (methanesulfonate), B Alkane sulfonate, besylate, p-toluenesulfonate, and pamoate; or ammonium (eg, primary
  • the term "pharmaceutically acceptable” means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients of the mammal comprising the formulation and/or treatment therewith.
  • treatment refers to both therapeutic and prophylactic or preventative or preventive measures, wherein the goal is to prevent or slow (reduce) undesired pathological changes or conditions.
  • beneficial or desirable clinical outcomes include, but are not limited to, alleviation of symptoms, reduction in disease severity, delay or slowing of disease progression, improvement or mitigation of disease states, and relief (either in part or in whole), regardless of Is detectable or undetectable.
  • the term "therapeutically effective amount” means that the amount of a compound of the invention may be: (i) treating or preventing a disease or condition described herein, (ii) ameliorating or eliminating one or more diseases or conditions described herein. Or (iii) preventing or delaying the onset of one or more symptoms of the disease or condition described herein.
  • the invention claims a compound of formula (Ia) or an ester, solvate, stereoisomer thereof, and pharmaceutically acceptable salts thereof:
  • W 1 , W 2 and W 3 are as defined in the formula (I).
  • W 1 is selected from the group consisting of an optionally substituted five- or six-membered heteroaryl ring containing O, N and/or S;
  • W 2 is selected from:
  • R 1 is selected from Wherein R 2 , R 3 , R 4 , R 4 ', R 5 or R 6 are each independently selected from H, C 1 -C 12 alkyl, amino C 1 -C 12 alkyl, C 1 -C 12 alkylamine Base C 1 -C 12 alkyl, Ra and Rb are each independently selected from H, C 1 -C 12 alkyl, OH, -OC 1 -C 12 alkyl, -NH 2 , -NHC 1 -C 12 alkyl, -N(C 1 -C 12 Alkyl) 2 , -SH, -SC 1 -C 12 alkyl, -S(O)C 1 -C 12 alkyl, -S(O 2 )C 1 -C 12 alkyl, -SO 3 H;R 7 and R 7 ' are each independently selected from -NH 2 , -NHC 1 -C 12 alkyl, -N(C 1 -C 12 alkyl
  • R 1 ' is selected from H, C 1 -C 12 alkyl, C 3 -C 8 cycloalkyl, OH, -OC 1 -C 12 alkyl, -NH 2 , -NHC 1 -C 12 alkyl, -N (C 1 -C 12 alkyl) 2 , -SH, -SC 1 -C 12 alkyl, -S(O)C 1 -C 12 alkyl, -S(O 2 )C 1 -C 12 alkyl, -SO 3 H;
  • Z 2 is selected from CR 18 R 19 or NR 20 , and R 18 , R 19 and R 20 are each independently selected from H, NH 2 or
  • R 21 is selected from the group consisting of -NH 2 , -NHC 1 -C 12 alkyl, -N(C 1 -C 12 alkyl) 2 , -O C 1 -C 12 alkyl, -S C 1 -C 12 alkyl;
  • R 22 is selected from the group consisting of NH, -NHC 1 -C 12 alkyl, O, S;
  • t, u and w are each independently selected from 1, 2, 3, 4, 5 or 6, provided that t and u are not 0 at the same time;
  • Or NH;
  • R 25 is selected from H, amino C 1 -C 12 alkyl or C 1 -C 12 alkylamino C 1 -C 12 alkyl.
  • W 1 is selected from Wherein X is selected from O, S or NH, and Y and Z are each independently selected from CH or N, and W 1 is optionally substituted by C 1 -C 12 alkyl.
  • W 1 is selected from the group consisting of a furan ring, a pyrrole ring, a pyrazole ring, an imidazole ring, a triazole ring, a thiophene ring, a thiazole ring, an isothiazole ring, an oxazole ring, an isoxazole ring, 1,2,4- Oxadiazole ring, 1,3,4-oxadiazole ring; pyridine ring, pyrimidinyl group, triazine ring, pyrazine ring, tetrazole ring, pyridazine ring, thiadiazole ring.
  • W 1 is selected from the group consisting of
  • M is selected from the group consisting of H, sodium ion, potassium ion, calcium ion, magnesium ion, NH 4 + , N(C 1 -C 12 alkyl) 4 + .
  • W 1 is selected from the group consisting of -C(O)-;
  • W 2 is selected from the group consisting of -OR 26 , -NHR 27 , wherein R 26 and R 27 are each independently selected from H, C 1 -C 12 alkyl or Wherein Z 5 is selected from CR 28 R 29 or NR 30 , and R 28 , R 29 and R 30 are each independently selected from H, NH 2 or Wherein R 31 is selected from the group consisting of -NH 2 , -NHC 1 -C 12 alkyl, -N(C 1 -C 12 alkyl) 2 , -O C 1 -C 12 alkyl, -S C 1 -C 12 alkyl; R 32 is selected from the group consisting of NH, NC 1 -C 12 alkyl, O, S; x, y and z are each independently selected from 1, 2, 3, 4, 5 or 6, provided that x and y are not simultaneously 0.
  • Preferred compounds of the invention are as follows:
  • the invention provides a process for the preparation of a compound of formula (I) of the invention:
  • Step 1 The intermediate 1 is reacted with an alkyne under basic conditions to obtain an intermediate 2;
  • Step 2 Selective removal of the intermediate 2 to the protecting group P to obtain the intermediate 3;
  • Step 3 Intermediate 3 is reacted with SO 3 under basic conditions to obtain Intermediate 4;
  • Step 4 The protecting group P' in the intermediate 4 is removed to give the product 5.
  • P is a hydroxy protecting group which is common in the art
  • P' is a hydroxy or amino protecting group which is common in the art
  • B is as defined in the formula (I).
  • Hydroxy or amino protecting groups include, but are not limited to, benzyl, silane protecting groups, ester protecting groups, alkyl ether protecting groups, Boc, Fmoc, Cbz, etc. See, for example, Greene, TW and Wuts, PGM, Greene's Protective Groups in Organic Synthesis, 4th edition, John Wiley and Sons.
  • the base used in the reaction may be an inorganic base or an organic base, wherein the inorganic base may be selected from alkali metal or alkaline earth metal hydroxides (for example, sodium hydroxide, potassium hydroxide, barium hydroxide, lithium hydroxide).
  • alkali metal or alkaline earth metal hydroxides for example, sodium hydroxide, potassium hydroxide, barium hydroxide, lithium hydroxide.
  • alkali metal or alkaline earth metal carbonate or hydrogencarbonate such as potassium carbonate, sodium carbonate, lithium carbonate, calcium carbonate, magnesium carbonate, sodium hydrogencarbonate
  • alkali metal or alkaline earth metal Alkoxide such as sodium methoxide, sodium ethoxide, sodium t-butoxide, potassium t-butoxide, etc.
  • alkali metal or alkaline earth metal amide such as sodium amide, sodium hexamethyldisilazide
  • ammonia organic base can Selected from organic amines commonly found in the art, such as triethylamine, trimethylamine, pyridine, piperidine, 4-N,N-dimethylaminopyridine, morpholine, N-methylmorpholine, tetramethylethylenediamine , DBU, DBN, DABCO, etc.
  • the deprotection operation is a routine experimental operation in the art, see, for example, Greene, T. W. and Wuts, P. G. M., Greene's Protective Groups in Organic Synthesis, 4th edition, John Wiley and Sons.
  • step 3 the base used in the reaction is as defined in step 1.
  • P is a hydroxy protecting group which is common in the art
  • P' is a hydroxy or amino protecting group which is common in the art
  • B is as defined in the formula (I).
  • Hydroxy or amino protecting groups include, but are not limited to, benzyl, silane protecting groups, ester protecting groups, alkyl ether protecting groups, Boc, Fmoc, Cbz, etc. See, for example, Greene, TW and Wuts, PGM, Greene's Protective Groups in Organic Synthesis, 4th edition, John Wiley and Sons.
  • Step 1 Intermediate 1 is condensed with a hydrazide under basic conditions to obtain Intermediate 2; wherein the base used in the reaction may be an inorganic base or an organic base, which is defined as the base in step 1 of Scheme 1.
  • the condensing agent may be a condensing agent commonly used in the art for the condensation reaction between a carboxylic acid and an amine, such as a carbodiimide condensing agent (CDI, DCC, DIC, EDCI and DMAP, HOBt, HOAt, HOSu, NHPI).
  • the reaction temperature may be in the range of 0-100 ° C, preferably in The reaction is in the range of 0-70 ° C, more preferably 0-50 ° C, most preferably at room temperature.
  • Step 2 Intermediate 2 is obtained by a ring closure reaction to obtain Intermediate 3; wherein the ring closure reaction is carried out in the presence of a condensing agent, and the condensing agent used is preferably Burgess reagent; the reaction temperature may be in the range of 0-100 ° C, It is preferably in the range of 0 to 70 ° C, more preferably 0 to 50 ° C, most preferably at room temperature.
  • a condensing agent preferably Burgess reagent
  • the reaction temperature may be in the range of 0-100 ° C, It is preferably in the range of 0 to 70 ° C, more preferably 0 to 50 ° C, most preferably at room temperature.
  • Step 3 Selective removal of the intermediate 3 to the protecting group P to obtain the intermediate 4;
  • Step 4 Intermediate 4 is reacted with SO 3 under basic conditions to obtain Intermediate 5;
  • Step 5 removing the protecting group P' in the intermediate 5 to obtain the product 6;
  • the deprotection operation is a routine experimental operation in the art, see, for example, Greene, T. W. and Wuts, P. G. M., Greene's Protective Groups in Organic Synthesis, 4th edition, John Wiley and Sons.
  • step 4 the base used in the reaction is as defined in step 1 of Scheme 1.
  • the present invention provides a pharmaceutical composition of the compound of the formula (I) or a pharmaceutically acceptable salt thereof as an active ingredient.
  • the composition comprises a compound of the invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier can be either solid or liquid. Wherein the solid carrier can be one or more of those used as excipients, diluents, sweeteners, solubilizers, lubricants, binders, tablet disintegrating agents, stabilizers, preservatives or encapsulating materials. substance.
  • the liquid carrier can be a solvent or a liquid dispersion medium.
  • Suitable solid carriers include, but are not limited to, for example, cellulose, glucose, lactose, mannitol, magnesium stearate, magnesium carbonate, sodium carbonate, sodium saccharin, sucrose, dextrin, talc, starch, pectin, gelatin, tragacanth, Acacia gum, sodium alginate, parabens, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like.
  • Suitable liquid carriers include, but are not limited to, water, ethanol, polyols (eg, glycerol, propylene glycol, liquid polyethylene glycol, and the like), vegetable oils (eg, peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil). ), glycerides, agar, pyrogen-free water, isotonic saline, Ringer's solution, and mixtures thereof.
  • polyols eg, glycerol, propylene glycol, liquid polyethylene glycol, and the like
  • vegetable oils eg, peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil.
  • glycerides eg, agar, pyrogen-free water, isotonic saline, Ringer's solution, and mixtures thereof.
  • compositions of the invention are generally known.
  • the preparation of the pharmaceutical compositions of the present invention in a known manner includes conventional methods of mixing, granulating, tableting, coating, dissolving or lyophilizing.
  • the therapeutically effective amount of the compound of the present invention or a pharmaceutical composition comprising the same can be easily determined by routine experimentation, and the most effective and convenient route of administration can be determined by routine experimentation.
  • the pharmaceutical composition of the present invention can be administered to a patient or a subject in need of treatment by any suitable administration, including oral administration, parenteral (including subcutaneous, intramuscular, intravenous, and intradermal) administration.
  • parenteral including subcutaneous, intramuscular, intravenous, and intradermal
  • Inhalation spray administration topical administration, rectal administration, nasal administration, buccal administration, vaginal administration or administration via an implantable reservoir.
  • the provided pharmaceutical compositions are administered intravenously and/or intraperitoneally.
  • parenteral includes subcutaneous, intravenous, intramuscular, intraocular, intravitreal, intra-articular, intrasynovial, intrasternal, intrathecal, intrahepatic. , intraperitoneal, intralesional, and intracranial injection or infusion techniques.
  • these pharmaceutical compositions are administered orally, subcutaneously, intraperitoneally or intravenously. .
  • compositions suitable for oral administration for use in the present invention include solid forms such as pills, tablets, caplets, capsules (including immediate release formulations, time release formulations and sustained release formulations, respectively), granules and powders; and liquids Forms such as solutions, syrups, elixirs, emulsions and suspensions.
  • forms for ocular administration include sterile solutions or ocular delivery devices.
  • Forms useful for parenteral administration include sterile solutions, emulsions and suspensions.
  • the pharmaceutical composition of the present invention may additionally comprise one or more ⁇ -lactam antibiotics.
  • the ⁇ -lactam antibiotic may include a penicillin antibiotic, a cephalosporin antibiotic, a monolactam cyclic antibiotic, a carbapenem antibiotic, and a penemase inhibitor.
  • the ⁇ -lactam antibiotic may include penicillin, oxacillin, cloxacillin, dicloxacillin, flucloxacillin, ampicillin, piroxicillin, amoxicillin, carbenicillin, furazocillin, sulfonate.
  • a compound of the present invention or an ester thereof, a stereoisomer or a pharmaceutically acceptable salt thereof, and a pharmaceutical composition comprising the compound of the present invention or an ester thereof, a stereoisomer thereof or a pharmaceutically acceptable salt thereof, can be used for treatment and / or prevent diseases caused by bacterial infections. Therefore, in the use of the compound of the formula (I) of the present invention, an ester thereof, a stereoisomer thereof or a pharmaceutically acceptable salt thereof for the preparation of a medicament for the treatment and/or prevention of bacteria The disease caused by infection.
  • the present invention relates to a method of inhibiting bacteria or treating and/or preventing a disease caused by a bacterial infection, comprising administering to a patient or subject in need thereof a therapeutically and/or prophylactically effective amount of the formula (I) of the present invention.
  • the bacteria in the disease caused by bacterial interference are selected from the group consisting of Gram-positive bacteria and Gram-negative bacteria, wherein the Gram-positive bacteria are selected from the group consisting of: Staphylococcus aureus, Staphylococcus epidermidis, and lactose-free chains One or more of cocci, S. pneumoniae, Streptococcus pyogenes, Enterococcus or C.
  • Gram-negative bacteria selected from the group consisting of: Citrobacter faecalis, Citrobacter freundii, Enterobacter cloacae, pneumonia Lei Bojun, Escherichia coli, Proteus vulgaris, Salmonella, Serratia marcescens, Shigella, Pseudomonas aeruginosa, Moraxella muforma, Neisseria gonorrhoeae, Neisseria meningitidis, immobile Bacillus, Burkholder, Campylobacter, Helicobacter pylori, Vibrio cholerae, Klebsiella, Haemophilus influenzae, Mycobacterium avium, Mycobacterium abscessus, Mycobacterium kansii, Mycobacterium ulcerum, Chlamydia pneumoniae, Trachoma One or more of Chlamydia, ⁇ -hemolytic streptococcus, Acinetobacter baumannii,
  • the bacterium causes a disease selected from the group consisting of: respiratory tract infection (upper respiratory tract infection, lower respiratory tract infection, bronchitis, bronchitis, pneumonia, tuberculosis, pharyngitis), urinary tract infection (complex urinary tract infection, complicated urethra) Infection, non-concurrent urinary tract infection, cystitis, pyelonephritis), central nervous system infection (encephalitis, meningitis, brain abscess), ear infection (otitis, otitis media), pleural lung and bronchial infection, intra-abdominal infection , cardiovascular infection (blood infections such as sepsis or bacteremia, endocarditis, myocarditis, pericarditis), skin or soft tissue infections, bone and joint infections (arthritis, osteomyelitis), genital infections (genital ulcers, vaginitis) , cervicitis), one or more of eye infections (conjunctivitis,
  • the compounds or pharmaceutical compositions of the invention may be provided in a pack or dispenser device containing one or more unit dosage forms.
  • the package may comprise a metal or plastic foil or glass and a rubber stopper (eg, in a vial).
  • the package or dispensing device can be accompanied by a instructions for use of the drug.
  • the dosage will depend on various factors including the age, weight and condition of the patient as well as the route of administration. The precise dose administered is determined based on the judgment of the treating physician.
  • the actual dosage level and time course of administration of the active ingredient in the pharmaceutical compositions of the present invention can be varied to achieve the following amounts of active ingredient which are effective to achieve the desired condition for the particular patient, composition and mode of administration.
  • the response is treated without toxicity to the patient.
  • the agents or pharmaceutical compositions of the invention are administered in an amount sufficient to reduce or eliminate the symptoms associated with bacterial infection.
  • the preferred dosage of the agent is the maximum amount that the patient can withstand and does not produce serious or unacceptable side effects.
  • Exemplary dosage ranges include 0.01 mg to 250 mg/day, 0.01 mg to 100 mg/day, 1 mg to 100 mg/day, 10 mg to 100 mg/day, 1 mg to 10 mg/day, and 0.01 mg to 10 mg/day.
  • the preferred dosage of the agent is the maximum amount that the patient can withstand and does not produce serious or unacceptable side effects.
  • the agent is administered at a concentration of from about 10 micrograms to about 100 mg/kg body weight per day, from about 0.1 to about 10 mg/kg/day, or from about 1.0 mg to about 10 mg/kg body weight per day.
  • the therapeutically effective dose produces a serum concentration of the agent from about 0.1 ng/ml to about 50-100 mg/ml.
  • These pharmaceutical compositions should typically provide a dosage of from about 0.001 mg to about 2000 mg of compound per kilogram of body weight per day.
  • the dose for systemic administration to a human patient may range from 1-10 mg/kg, 20-80 mg/kg, 5-50 mg/kg, 75-150 mg/kg, 100-500 mg/kg, 250-750 mg/kg.
  • the pharmaceutical unit dosage form is prepared to provide a compound or combination of essential ingredients from about 1 mg to about 5000 mg (e.g., from about 100 mg to about 2500 mg) per unit dosage form.
  • Preferred unit dosage formulations are those containing the daily dose or unit, daily sub-dose, or suitable fraction thereof as discussed herein.
  • Steps 1-4 follow steps 2-5 in the synthesis of Compound D.
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Step 1-6 Steps 1-6 in the synthesis of Compound E
  • the crude product was purified by Prep-HPLC using the following conditions: column: XBridge Prep OBD C18 column 19*250 mm, 5 um; mobile phase A: water (0.1% FA), mobile phase B: ACN; flow rate: 20 mL/min; gradient: From 12% B to 23% B in 7 minutes; 220, 254 nm; Rt: 6.2 min; 8 mg (12%) of compound E was obtained.
  • ESI-MS (EI + , m/z): 402, 0.418 min.
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Step 1-5 follow steps 1-6 in the synthesis of Compound I
  • Step 6 follow step 8 in the synthesis of Compound I
  • Step 7-8 follow steps 1-2 in the synthesis of Compound E
  • ESI-MS EI + , m/z
  • Steps 10-12 Steps 3-5 in the synthesis of Compound E
  • Step 1-6 Steps 1-6 in the synthesis of Compound E
  • Step 1-6 Steps 1-6 in the synthesis of Compound E
  • Step 1-6 Steps 1-6 in the synthesis of Compound E
  • Step 1-3 follow steps 1-3 in the synthesis of Compound D
  • Step 4-8 Steps 1-5 in the synthesis of Compound E
  • Step 3-8 Steps 1-6 in the synthesis of Compound D
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Step 3-8 Steps 1-6 in the synthesis of Compound D
  • Step 1-3 follow steps 4-6 in the synthesis of Compound D
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Step 1-6 Steps 1-6 in the synthesis of Compound D
  • Steps 1-5 follow steps 2-6 in the synthesis of compound FF
  • Steps 1-5 follow steps 2-6 in the synthesis of compound FF
  • Steps 1-4 Steps 2, 4-6 in the synthesis of Compound D
  • Steps 1-4 Steps 2, 4-6 in the synthesis of Compound D
  • Steps 1-4 Steps 2, 4-6 in the synthesis of Compound D
  • Steps 5-9 Steps 1-5 in accordance with the synthesis of Compound D
  • Steps 10-11 Steps 8-9 in the synthesis of Compound I
  • Step 1-4 Steps 1-4 in the synthesis of compound DD
  • Steps 5-9 Steps 1-5 in the synthesis of Compound D
  • Steps 10-11 Steps 8-9 in the synthesis of Compound I
  • Step 2-5 follow steps 3-6 in the synthesis of compound FF
  • Step 1-5 Steps 1-5 in the synthesis of compound GG
  • Steps 1-4 follow steps 3-6 in the synthesis of compound FF
  • Step 1 Step 1 in the synthesis of compound GG
  • Step 2-5 follow steps 3-6 in the synthesis of compound FF
  • Step 1-5 Steps 1-5 in the synthesis of compound GG
  • Step 1 follow step 1 in the synthesis of compound UU
  • Step 2 follow step 1 in the synthesis of compound GG
  • Step 5-7 follow steps 3-5 in the synthesis of compound FF
  • TEA ⁇ HF (0.068 g, 0.56 mmol) was added to (1R,4S)-4-(5-((E)-8-((tert-butoxycarbonyl)amino)-2,2,3,3, 12,12-hexamethyl-10-oxo-4,11-dioxa-7,9-diaza-3-silatridec-8-8-en-5-yl)isoxazole-3- 6-oxo-5,7-diazaspiro[bicyclo[3.2.1]octane-2,1'-cyclopropane]-7-yl hydrogensulfate (0.102 g, 0.14 mmol) In a THF solution (2.000 mL).
  • Step 9 follow step 6 in the synthesis of compound LL
  • Steps 1-4 follow steps 6-9 in the synthesis of Compound I
  • Steps 1-4 follow steps 6-9 in the synthesis of Compound I
  • Step 1-5 Steps 1-5 in the synthesis of compound FF
  • Step 2-4 follow steps 4-6 in the synthesis of compound FF
  • Steps 1-4 follow steps 2-5 in the synthesis of compound FF
  • Steps 1-4 follow steps 2-5 in the synthesis of compound FF
  • Steps 1-4 follow steps 2-5 in the synthesis of compound FF
  • Step 3-6 follow steps 2-5 in the synthesis of compound FF
  • Step 2 follow step 2 in the synthesis of compound GG
  • Step 3-4 follow steps 1-2 in the synthesis of compound TT
  • Step 5-8 follow steps 2-5 in the synthesis of compound FF
  • Step 1 follow step 2 in the synthesis of compound TT
  • Step 3-6 Steps 1-4 in the synthesis of the compound PP
  • Step 4-7 Steps 1-4 in the synthesis of the compound PP
  • Step 2-6 follow steps 2-6 in the synthesis of compound VV
  • Step 1 Step 1 in the synthesis of compound GG
  • Step 2-3 follow steps 1-2 in the synthesis of compound TT
  • Step 4-7 Steps 1-4 in the synthesis of the compound PP
  • step 1
  • Step 2-7 Steps 2-7 in the synthesis of the compound WW
  • step 1
  • Step 3 follow step 2 in the synthesis of compound TT
  • Step 6 in accordance with step 1 in the synthesis of compound GG
  • Step 7-10 Steps 1-4 in the synthesis of the compound PP
  • Compound 5 was prepared according to steps 1-5 in Example 60.
  • Phenyl-I3-iodanediyl-bis(2,2,2-trifluoroacetate) (3.6 g, 8.37 mmol, 1.5 eq.) was added portionwise to compound 3 (1.48 g, 5.58 mmol, 1 eq.) And a solution of ter-2-butyl prop-2-yn-1-ylcarbamate (0.86 g, 5.58 mmol, 1 eq.) in methanol (20 mL) and water (4 mL). The resulting mixture was stirred at room temperature for 4 hours. The solvent was removed under reduced pressure.
  • DIPEA N,N-Diisopropylethylamine
  • TSA Trypticase soy agar
  • the bacterial strain used for the minimum inhibitory concentration (MIC) test was stored frozen in a -80 ° C low temperature refrigerator, and the use required to recover 2 days earlier. A small amount of frozen bacterial strain was scraped off with a sterile inoculating loop and streaked on a TSA solid medium plate, and placed in a normal atmospheric culture environment at 35 ⁇ 2 ° C for 20-24 hours.
  • the liquid medium was taken out from the refrigerator at 4 ° C and left to heat at room temperature.
  • the bacteria were diluted 300-fold with 1.1x CAMHB.
  • Test compound for line A-H highest final test concentration of 64 ⁇ g/ml or 32 ⁇ g/ml, diluted 2 times.
  • GC Growth control
  • SC Sterile control
  • Test compound dilution All test compounds were dissolved and diluted with dimethyl sulfoxide (DMSO). 170 ⁇ l of 3.2 mg/ml (50 ⁇ x 64 ⁇ g/ml) of the compound was transferred to the wells of column 1 of the dilution mother plate (A1-H1), and then 85 ⁇ l of DMSO was transferred to the wells of the other columns. Each compound was serially diluted 2 times (ie, 85 ⁇ l of the compound was aspirated from the wells of column 1 into the wells of column 2 and mixed, and 85 ⁇ l of the compound was aspirated from the wells of column 2 to column 3. Mix wells in the wells, then pipet 85 ⁇ l of the compound from the wells in column 3 into the wells of column 4 and mix, and so on to the wells in column 11).
  • DMSO dimethyl sulfoxide
  • Imipenem and ceftazidime were dissolved in 10 mM MOPS buffer. Transfer 110 ⁇ l of 50 ⁇ g/ml (12.5 ⁇ x 4 ⁇ g/ml) of imipenem or 110 ⁇ l of 100 ⁇ g/ml (12.5 ⁇ x 8 ⁇ g/ml) of ceftazidime to columns 1-11 of the addition plate, and then transfer 110 ⁇ l of MOPS buffer to Column 12.
  • cover 6 test plates with a sterile cover, centrifuge in a centrifuge at 1000 rpm for 30 seconds, mix at 800 rpm on a vibrating plate for 1 minute, and place in a normal incubator at 35 ⁇ 2 ° C for 16-20 hours. .
  • the inoculated bacteria were diluted from 10 -1 to 10 -7 with a liquid medium (such as 100 ⁇ l of bacterial inoculum + 900 ⁇ l of 1.1xCAMHB).
  • test plates were placed on a plate reading device, and the mirrors were adjusted to observe the growth of bacteria in each well. At the same time, each test board was photographed with QCount software.
  • the number of colonies of the bacterial inoculum at different dilutions on the TSA plates was counted and the bacterial inoculum was calculated.
  • test compound When the test compound was tested in combination with imipenem, ceftazidime and ampicillin, imipenem was fixed at a concentration of 4 ⁇ g/ml, ceftazidime was fixed at a concentration of 8 ⁇ g/ml, and ampicillin was fixed at a concentration of 8 ⁇ g/ml.
  • the bacterial inoculum in the assay plate was resuscitated from TSA and diluted in CAMHB, while a growth control (GC well) and a sterile control (SC well) were placed in the assay plate.
  • the test plates were observed and cultured at 35 ⁇ 2 ° C for 16-20 hours in a conventional incubator to observe and record the minimum inhibitory concentration of each compound combination against different bacteria. See Table 1-3 for the results.
  • A ⁇ 8 ⁇ g/ml
  • B 8-16 ⁇ g/ml
  • C 16-128 ⁇ g/ml
  • A ⁇ 8 ⁇ g/ml
  • B 8-16 ⁇ g/ml
  • C 16-128 ⁇ g/ml
  • A ⁇ 8 ⁇ g/ml
  • B 8-16 ⁇ g/ml
  • C 16-128 ⁇ g/ml

Landscapes

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

Abstract

提供了一种式(I)所示的β-内酰胺酶抑制剂或其酯、立体异构体、药学上可接受的盐及其制备方法,以及包含式(I)所示的β-内酰胺酶抑制剂或其酯、立体异构体、药学上可接受的盐的药物组合物;和治疗细菌感染导致的疾病的方法,包括向有需要的患者或受试者施用式(I)所示的β-内酰胺酶抑制剂或其酯、立体异构体、以及药学上可接受的盐。

Description

β-内酰胺酶抑制剂及其用途 技术领域
本发明涉及一种β-内酰胺酶抑制剂及其制备方法。本发明还涉及一种包含β-内酰胺酶抑制剂的药物组合物及其用途。
背景技术
β-内酰胺类抗生素是最早引用于临床的抗生素。自从青霉素G作为第一种β-内酰胺类抗生素成功应用到临床上以来,β-内酰胺类抗生素得到了快速的发展。不同结构类型的β-内酰胺类抗生素得到开发并在临床上得到大量的应用,取得了很好的效果。但是,由于细菌细胞可以产生β-内酰胺酶,使得抗生素失活,导致细菌对β-内酰胺类抗生素产生耐药性。β-内酰胺酶是催化β-内酰胺环水解的酶,其使得β-内酰胺抗生素的抗菌活性失活并允许细菌对β-内酰胺抗生素具有耐药性。β-内酰胺酶依据分子结构中的氨基酸序列差异可以分为A类、B类、C类和D类等。A类β-内酰胺酶优选地水解青霉素类抗生素,B类β-内酰胺酶可以水解各种β-内酰胺类抗生素,包括碳青霉烯类抗生素,C类β-内酰胺酶能更有效地水解头孢菌素类抗生素,而D类β-内酰胺酶则更倾向于水解苯唑青霉素和邻氯青霉素。细菌,特别是革兰氏阴性菌对于β-内酰胺类抗生素的耐药性通常都是通过β-内酰胺酶介导的。
对β-内酰胺酶的抑制可以延缓或抑制β-内酰胺类抗生素的降解并恢复产生β-内酰胺类抗生素耐药性的细菌对β-内酰胺类抗生素的敏感性。目前,临床上通过将β-内酰胺酶与β-内酰胺类抗生素联合应用,可以使得β-内酰胺酶对于β-内酰胺类抗生素的水解活性失活,从而增强了细菌对β-内酰胺类抗生素的敏感性,减少或克服了耐药性问题。现有技术公开了多种细菌对β-内酰胺酶抑制剂,例如WO2013149121A1、WO2014141132A1、 US20130296290A1、WO2013030735A1、WO2015110963A1、WO2015150890A1、WO2015159265A1、WO2015173663、WO2015173665A1、WO2017055922A1公开的二氮杂螺环[双环[3.2.1]辛烷类化合物。而且,市面上也有多种β-内酰胺酶抑制剂销售,例如克拉维酸、他唑巴坦、阿维巴坦、Relebactam等。但是,上述β-内酰胺酶抑制剂对β-内酰胺酶的抑制效果还不能令人十分满意。因此,目前存在对于新型β-内酰胺酶抑制剂的迫切需求,以期可以与β-内酰胺抗生素联合治疗由β-内酰胺抗生素具有抗性的细菌引起的感染。
发明内容
本发明提供一种式(I)所示的化合物或其酯、其立体异构体以及其药学上可接受的盐:
Figure PCTCN2019073000-appb-000001
其中,W 1选自任选取代的含有O、N和/或S的五元或6元杂芳环或-C(O)-;并且,(i)当W 1选自任选取代的含有O、N和/或S的五元或6元杂芳环时,W 1任选地被C 1-C 12烷基取代,
W 2选自:
a.H
b.
Figure PCTCN2019073000-appb-000002
其中R 1选自
Figure PCTCN2019073000-appb-000003
Figure PCTCN2019073000-appb-000004
Figure PCTCN2019073000-appb-000005
其中R 2、R 3、R 4、R 4’、R 5或R 6各自独立地选自H、C 1-C 12烷基、氨基C 1-C 12烷基、C 1-C 12烷 胺基C 1-C 12烷基、
Figure PCTCN2019073000-appb-000006
Ra和Rb各自独立地选自H、C 1-C 12烷基、OH、-OC 1-C 12烷基、-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-SH、-SC 1-C 12烷基、-S(O)C 1-C 12烷基、-S(O 2)C 1-C 12烷基、-SO 3H;R 7和R 7’各自独立地选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-O C 1-C 12烷基、-S C 1-C 12烷基;R 8和R 8’各自独立地选自NH、-NC 1-C 12烷基、O、S;R 9和R 9’各自独立地选自-NH-、-N(C 1-C 12烷基)-、-O-、-S-;Z 1选自CR 10R 11或NR 12,其中R 10、R 11和R 12各自独立地选自H、NH 2
Figure PCTCN2019073000-appb-000007
其中R 13和R 15各自独立地选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-O C 1-C 12烷基、-S C 1-C 12烷基;R 14和R 16各自独立地选自NH、-NHC 1-C 12烷基、O、S;R 17选自-NH-、-N(C 1-C 12烷基)-、-O-、-S-;i、j、k、l、m、n、p、q、r和s各自独立地选自0、1、2、3、4、5或6;条件是q和r不同时为0;
R 1’选自H、C 1-C 12烷基、C 3-C 8环烷基、OH、-OC 1-C 12烷基、-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-SH、-SC 1-C 12烷基、-S(O)C 1-C 12烷基、-S(O 2)C 1-C 12烷基、-SO 3H;
c.
Figure PCTCN2019073000-appb-000008
其中Z 2选自CR 18R 19或NR 20,R 18、R 19和R 20各自独立地选自H、NH 2
Figure PCTCN2019073000-appb-000009
其中R 21选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-O C 1-C 12烷基、-S C 1-C 12烷基;R 22选自NH、-NHC 1-C 12烷基、O、 S;t、u和w各自独立地选自1、2、3、4、5或6,条件是t和u不同时为0;
d.
Figure PCTCN2019073000-appb-000010
其中Z 3和Z 4各自独立地选自-NR 25-、-O-,R 23和R 24各自独立地选自H、C 1-C 12烷基,或者R 23和R 24一起形成=O或=NH;R 25选自H、氨基C 1-C 12烷基或C 1-C 12烷胺基C 1-C 12烷基;
(ii)当W 1选自-C(O)-时,W 2选自-OR 26、-NHR 27,其中R 26和R 27各自独立地选自H、C 1-C 12烷基或
Figure PCTCN2019073000-appb-000011
其中Z 5选自CR 28R 29或NR 30,R 28、R 29和R 30各自独立地选自H、NH 2
Figure PCTCN2019073000-appb-000012
其中R 31选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-O C 1-C 12烷基、-S C 1-C 12烷基;R 32选自NH、NC 1-C 12烷基、O、S;x、y和z各自独立地选自1、2、3、4、5或6,条件是x和y不同时为0;
W 3选自-SO 3M、-OSO 3M、-OSO 2NH 2、-OPO 3M、-OCR 33R 34CO 2M、-OCR 35R 36SO 3M、-OCR 37R 38PO 3M;其中,M选自H或药学上可接受的阳离子;R 33、R 34、R 35、R 36、R 37和R 38各自独立地选自H、C 1-C 12烷基、卤素。
另一方面,本发明提供一种式(I)所示的化合物或其药物上可接受的盐的制备方法。
再一方面,本发明提供一种药物组合物,其包含式(I)所示的化合物或其药学上可接受的盐以及一种或多种β-内酰胺类抗生素。
再一方面,本发明提供一种治疗由细菌感染导致的疾病的方法,其包括将本申请所述的式(I)所示的化合物与一种或多种β-内酰胺类抗生素联合施用于有需要的患者或者受试者。
具体实施方式
定义:
如本文所用,术语“烷基”是指直链的或支链的具有1-20个碳原子的饱和烃基。优选地,烷基是具有1至12个碳原子的烷基。更优选地,烷基是具有1-6个碳原子的烷基。最优选地,烷基是具有1-4个碳原子的烷基。烷基的实例包括,但不限于,甲基、乙基、1-丙基(正丙基)、2-丙基(异丙基)、1-丁基(正丁基)、2-甲基-1-丙基(异丁基)、2-丁基(仲丁基)、2-甲基-2-丙基(叔丁基)、1-戊基(正戊基)、2-戊基、3-戊基、2-甲基-2-丁基、3-甲基-2-丁基、3-甲基-1-丁基、2-甲基-1-丁基、1-己基、2-己基、3-己基、2-甲基-2-戊基、3-甲基-2-戊基、4-甲基-2-戊基、3-甲基-3-戊基、2-甲基-3-戊基、2,3-二甲基-2-丁基、3,3-二甲基-2-丁基、1-庚基、1-辛基、1-壬基、1-癸基等。
如本文所用,术语“氨基”是指-NH 2
如本文所用,术语“卤素”是指氟、氯、溴、碘。
如本文所用,术语“氨基烷基”是指其中一个或多个氢被氨基取代的烷基。
如本文所用,术语“环烷基”是指单价饱和碳环基团。优选地,环烷基是3至8元单环基团。更优选地,环烷基是3至6元单环基团。环烷基的实例包括但不限于环丙基、环丁基、环戊基、环己基等。
如本文所用,术语“杂芳基”是指5或6元环的一价芳族基团,其含有独立地选自选自氮、氧和硫的1-3个杂原子。杂芳基的实例包括:呋喃环、吡咯环、吡唑环、咪唑环、三唑环、噻吩环、噻唑环、异噻唑环、噁唑环、异噁唑环、1,2,4-噁二唑环、1,3,4-噁二唑环;吡啶环、嘧啶基、三嗪环、吡嗪环、四唑环、哒嗪环、噻二唑环。杂芳基任选独立地被本文描述的一个或多个取代基取代。
如本文所用,术语“任选取代的”是指所给结构或基团未被取代,或所给结构或基团被一个或多个具体取代基取代。除非其它方 面的说明,任选取代可以在被取代基团的任意位置进行取代。
如本文所用,术语“酯”是指式(I)所示的化合物中的-OSO 3-或COOH(如果存在的话)可与醇发生酯化反应而形成的酯;当式(I)所示的化合物中存在羟基的时候,其可与有机酸、无机酸等发生酯化反应而形成的酯。该酯在酸或碱性条件下可发生水解反应生成相应的酸或醇。
如本文所用,术语“立体异构体”是指具有相同的化学组成和连接性,但是其原子在空间具有不同取向的化合物,该取向不能通过单键旋转互换。“立体异构体”包括了“非对映异构体”和“对映异构体”。“非对映异构体”是指具有两个或多个手性中心且其分子并非彼此镜像的立体异构体。非对映异构体具有不同的物理性质,如熔点、沸点、光谱特性和反应性。非对映异构体混合物可在高分辨率分析程序(诸如结晶、电泳和色谱法)下进行分离。“对映异构体”是指彼此为非重叠镜像化合物的两种立体异构体。
如本文所用,术语“药学上可接受的盐”是指本发明的化合物的药学上可接受的有机或无机盐。示例性的盐包括但不限于:硫酸盐、柠檬酸盐、乙酸盐、草酸盐、氯化物、溴化物、碘化物、硝酸盐、硫酸氢盐、磷酸盐、酸式磷酸盐、异烟酸盐、乳酸盐、水杨酸盐、酸式柠檬酸盐、酒石酸盐、油酸盐、鞣酸盐、泛酸盐、酒石酸氢盐、抗坏血酸盐、琥珀酸盐、马来酸盐、龙胆酸盐、富马酸盐、葡糖酸盐、葡糖醛酸盐、糖酸盐、甲酸盐、苯甲酸盐、谷氨酸盐,甲烷磺酸盐(甲磺酸盐)、乙烷磺酸盐、苯磺酸盐、对甲苯磺酸盐、和双羟萘酸盐;或者铵盐(例如伯胺盐、仲胺盐、叔胺盐、季铵盐)、金属盐(例如钠盐、钾盐、钙盐、镁盐、锰盐、铁盐、锌盐、铜盐、锂盐、铝盐)。
如本文所用,术语“药学上可接受的”表示该物质或组合物与包含制剂和/或用其处理的哺乳动物的其他成分必须化学和/或毒理学上兼容。
如本文所用,术语“治疗”是指治疗性治疗和预防性或防范性 或阻止性措施,其中目的是预防或减缓(减轻)不期望的病理变化或病症。对于本发明的目的,有益或期望的临床结果包括但不限于:症状的减轻,疾病程度的降低,延缓或减慢疾病进展,改善或缓和疾病状态,以及缓解(无论是部分还是全部),无论是可检测的还是不可检测的。
如本文所用,术语“治疗有效量”是指本发明的化合物的量可以:(i)治疗或预防本文描述的疾病或病症,(ii)改善或消除本文描述的一种或多种疾病或病症,或者(iii)预防或延迟本文描述的疾病或病症的一种或多种症状的发作。
在一个实施方案中,本发明请求保护式(Ia)所示的化合物或其酯、溶剂合物、立体异构体以及其药学上可接受的盐:
Figure PCTCN2019073000-appb-000013
其中,W 1、W 2和W 3的定义如式(I)所述。
在一个优选的实施方案中,W 1选自任选取代的含有O、N和/或S的五元或6元杂芳环;
W 2选自:
a.H
b.
Figure PCTCN2019073000-appb-000014
其中R 1选自
Figure PCTCN2019073000-appb-000015
Figure PCTCN2019073000-appb-000016
Figure PCTCN2019073000-appb-000017
其中R 2、R 3、R 4、R 4’、R 5或R 6各自独立地选自H、C 1-C 12烷基、氨基C 1-C 12烷基、C 1-C 12烷胺基C 1-C 12烷基、
Figure PCTCN2019073000-appb-000018
Ra和Rb各自独立地选自H、C 1-C 12烷基、OH、-OC 1-C 12烷基、-NH 2、 -NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-SH、-SC 1-C 12烷基、-S(O)C 1-C 12烷基、-S(O 2)C 1-C 12烷基、-SO 3H;R 7和R 7’各自独立地选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-O C 1-C 12烷基、-S C 1-C 12烷基;R 8和R 8’各自独立地选自NH、-NC 1-C 12烷基、O、S;R 9和R 9’各自独立地选自-NH-、-N(C 1-C 12烷基)-、-O-、-S-;Z 1选自CR 10R 11或NR 12,其中R 10、R 11和R 12各自独立地选自H、NH 2
Figure PCTCN2019073000-appb-000019
Figure PCTCN2019073000-appb-000020
其中R 13和R 15各自独立地选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-O C 1-C 12烷基、-S C 1-C 12烷基;R 14和R 16各自独立地选自NH、-NHC 1-C 12烷基、O、S;R 17选自-NH-、-N(C 1-C 12烷基)-、-O-、-S-;i、j、k、l、m、n、p、q、r和s各自独立地选自0、1、2、3、4、5或6;条件是q和r不同时为0;
R 1’选自H、C 1-C 12烷基、C 3-C 8环烷基、OH、-OC 1-C 12烷基、-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-SH、-SC 1-C 12烷基、-S(O)C 1-C 12烷基、-S(O 2)C 1-C 12烷基、-SO 3H;
c.
Figure PCTCN2019073000-appb-000021
其中Z 2选自CR 18R 19或NR 20,R 18、R 19和R 20各自独立地选自H、NH 2
Figure PCTCN2019073000-appb-000022
其中R 21选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-O C 1-C 12烷基、-S C 1-C 12烷基;R 22选自NH、-NHC 1-C 12烷基、O、S;t、u和w各自独立地选自1、2、3、4、5或6,条件是t和u不同时为0;
d.
Figure PCTCN2019073000-appb-000023
其中Z 3和Z 4各自独立地选自-NR 25-、-O-,R 23和R 24各自独立地选自H、C 1-C 12烷基,或者R 23和R 24一起形成=O或=NH;R 25选自H、氨基C 1-C 12烷基或C 1-C 12烷胺基C 1-C 12烷基。
优选地,W 1选自
Figure PCTCN2019073000-appb-000024
其中,X选自O、S或NH,Y和Z各自独立地选自CH或N,并且W 1任选地被C 1-C 12烷基取代。
更优选地,W 1选自呋喃环、吡咯环、吡唑环、咪唑环、三唑环、噻吩环、噻唑环、异噻唑环、噁唑环、异噁唑环、1,2,4-噁二唑环、1,3,4-噁二唑环;吡啶环、嘧啶基、三嗪环、吡嗪环、四唑环、哒嗪环、噻二唑环。
最优选地,W 1选自选自
Figure PCTCN2019073000-appb-000025
M选自H、钠离子、钾离子、钙离子、镁离子、NH 4 +、N(C 1-C 12烷基) 4 +
在另一个优选的实施方案中,W 1选自选自-C(O)-;
W 2选自选自-OR 26、-NHR 27,其中R 26和R 27各自独立地选自H、C 1-C 12烷基或
Figure PCTCN2019073000-appb-000026
其中Z 5选自CR 28R 29或NR 30,R 28、R 29和R 30各自独立地选自H、NH 2
Figure PCTCN2019073000-appb-000027
其中R 31选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-O C 1-C 12烷基、-S C 1-C 12烷基;R 32选自NH、NC 1-C 12烷基、O、S;x、y和z各自独立地选自1、2、3、4、5或6,条件是x和y不同时为0。
本发明的优选的化合物如下所示:
(1R,4S)-4-(1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环 [3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(胍基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-氨基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-胍基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(3-氨基丙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸钠;
(1R,4S)-4-(5-(4-氨基丁基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-氨基-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-胍基-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((R)-2-胍基-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-2-胍基-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(1-(胍基甲基)环丙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((氮杂环丁烷-3-基氨基)甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(胍基甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((1-甲基胍基)甲基)异噁唑-3-y1)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7基硫酸;
(1R,4S)-4-(5-(氨基甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((甲基氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-胍基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-(1-甲基胍基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-胍基-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-2-胍基-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((R)-2-胍基-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-氨基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-(甲基氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-6-氧代-4-(5-(哌啶-4-基)异噁唑-3-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-6-氧代-4-(5-(哌啶-3-基)异噁唑-3-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-6-氧代-4-(5-(吡咯烷-3-基)异噁唑-3-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-6-氧代-4-(5-(2-(哌啶-4-基氨基)乙基)异噁唑-3-基)-5,7- 二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-((1-脒基哌啶-4-基)氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((1s,3R)-3-氨基环丁基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((1r,3S)-3-氨基环丁基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((1R,3S)-3-氨基环戊基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((1R,3R)-3-氨基环戊基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(((1r,3R)-3-氨基环丁基)甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(((1s,3S)-3-氨基环丁基)甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((1-氨基环丙基)甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((1-(甲基氨基)环丙基)甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(氮杂环丁烷-3-基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-(甲基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((氮杂环丁烷-3-基氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(氮杂环丁烷-3-基甲基)异噁唑-3-基)-6-氧代-5,7- 二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((氮杂环丁烷-3-基氧基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(((1-脒基氮杂环丁烷-3-基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(3-氨基环丁基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((1s,3S)-3-氨基环丁基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((1r,3R)-3-氨基环丁基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-((2-胍基乙基)氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-6-氧代-4-(哌啶-4-基氨基甲酰基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-6-氧代-4-(吡咯烷-3-基氨基甲酰基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(氮杂环丁烷-3-基氨基甲酰基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-2-((2-氨基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((R)-2-亚氨基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6- 氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
2-(((1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)乙酸;
2-(((1R,4S)-4-(5-(((叔丁氧羰基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)乙酸;
2-(((1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)-2,2-二氟乙酸;
2-(((1R,4S)-4-(5-(((叔丁氧羰基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)-2,2-二氟乙酸;
(1R,4S)-4-(5-((1s,3R)-3-胍基环丁基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((R)-1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-((2-氨基乙基)氨基)-1-羟基乙基)异噁唑-3-yl)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-2-((2-氨基乙基)氨基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((R)-2-((2-氨基乙基)氨基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫 酸;
(1R,4S)-4-(5-(2-((2-胍基乙基)氨基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-2-((2-胍基乙基)氨基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((R)-2-((2-胍基乙基)氨基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((R)-1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-((2-氨基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((R)-2-((2-氨基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-((2-胍基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-2-((2-胍基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((R)-2-((2-胍基乙基)氨基)-1-羟基乙基)-1,3,4-噁 二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-(氮杂环丁烷-3-基氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(1-羟基-2-(甲基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(1-羟基-2-(吡咯烷-3-基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-(二甲基氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(羟基(吡咯烷-2-基)甲基)-1,3,4-噁二唑-2-y1)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
2-羟基-N,N,N-三甲基-2-(5-((1R,4S)-6-氧代-7-(磺氧基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)-1,3,4-噁二唑-2-基)乙烷-1-铵;
(1R,4S)-4-(5-(1-羟基-2-(哌啶-4-基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(4-羟基哌啶-4-基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(1-脒基哌啶-4-基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-6-氧代-4-(5-(哌嗪-1-基)异噁唑-3-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-6-氧代-4-(5-(2-氧代哌嗪-1-基)异噁唑-3-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(L-脯氨酰基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-2-((2-(二甲基氨基)乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
2-(((2S)-2-羟基-2-(5-((1R,4S)-6-氧代-7-(磺氧基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)-1,3,4-噁二唑-2-基)乙基)氨基)-N,N,N-三甲基乙烷-1-铵;
(1R,4S)-6-氧代-4-(5-(2-氧代咪唑烷-4-基)-1,3,4-噁二唑-2-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-6-氧代-4-(5-(2-氧代噁唑烷-5-基)-1,3,4-噁二唑-2-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-6-氧代-4-(5-(2-氧代噁唑烷-4-基)-1,3,4-噁二唑-2-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-亚胺基-1-((甲基氨基)甲基)咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(1-((二甲基氨基)甲基)-2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
1-(2-亚胺基-4-(5-((1R,4S)-6-氧代-7-(磺氧基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)-1,3,4-噁二唑-2-基)咪唑烷-1-基)-N,N,N-三甲基甲烷铵;
(1R,4S)-4-(5-(2-亚胺基-1-((甲基氨基)甲基)-5-氧代咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(5-羟基-2-亚胺基-1-((甲基氨基)甲基)咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(3-((甲基氨基)甲基)-2-氧代噁唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(1-(氨基甲基)-2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(5-氨基-2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(2-亚胺基-5-(甲基氨基)咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-(5-(二甲基氨基)-2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((R)-2-氨基-1-羟乙基)-1,3,4-噁二唑-2-基-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-2-氨基-1-羟乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-1-羟基-2-(甲基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
(1R,4S)-4-(5-((S)-2-(二甲基氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
2-(((1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)乙酸;
2-(((1R,4S)-4-(5-(((叔丁氧羰基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)乙酸;
2-(((1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)-2,2-二氟乙酸;
2-(((1R,4S)-4-(5-(((叔丁氧羰基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)-2,2-二氟乙酸。
另一方面,本发明提供了制备本发明的式(I)所示的化合物的方法:
方案一
Figure PCTCN2019073000-appb-000028
步骤1:将中间体1与炔烃在碱性条件下关环反应得到中间体2;
步骤2:将中间体2选择性脱除保护基P得到中间体3;
步骤3:将中间体3在碱性条件下与SO 3反应得到中间体4;
步骤4:将中间体4中的保护基P’去除得到产物5。
其中,P为本领域常见的羟基保护基,P’为本领域常见的羟基或氨基保护基,B的定义如通式(I)中所定义的。羟基或氨基保护基包括但不限于苄基、硅烷保护基、酯基保护基、烷基醚基保护基、Boc、Fmoc、Cbz等,参见例如Greene,T.W.和Wuts,P.G.M.,Greene’s Protective Groups in Organic Synthesis,第4版,John Wiley and Sons。
在步骤1中,反应中所用的碱可以是无机碱或有机碱,其中无 机碱可以选自碱金属或碱土金属的氢氧化物(例如氢氧化钠、氢氧化钾、氢氧化钡、氢氧化锂、氢氧化钙、氢氧化镁)、碱金属或碱土金属的碳酸盐或碳酸氢盐(例如碳酸钾、碳酸钠、碳酸锂、碳酸钙、碳酸镁、碳酸氢钠)、碱金属或碱土金属的醇盐(例如甲醇钠、乙醇钠、叔丁醇钠、叔丁醇钾等)、碱金属或碱土金属的氨基化物(例如氨基钠、六甲基二硅氨基钠)、氨水,有机碱可以选自本领域常见的有机胺,例如三乙胺、三甲胺、吡啶、哌啶、4-N,N-二甲基氨基吡啶、吗啉、N-甲基吗啉、四甲基乙二胺、DBU、DBN、DABCO等。
在步骤2和4中,脱保护的操作是本领域常规的实验操作,参见例如Greene,T.W.和Wuts,P.G.M.,Greene’s Protective Groups in Organic Synthesis,第4版,John Wiley and Sons。
在步骤3中,反应中所用的碱如步骤1中所定义的。
方案二:
Figure PCTCN2019073000-appb-000029
其中,P为本领域常见的羟基保护基,P’为本领域常见的羟基或氨基保护基,B的定义如通式(I)中所定义的。羟基或氨基保护基 包括但不限于苄基、硅烷保护基、酯基保护基、烷基醚基保护基、Boc、Fmoc、Cbz等,参见例如Greene,T.W.和Wuts,P.G.M.,Greene’s Protective Groups in Organic Synthesis,第4版,John Wiley and Sons。
步骤1:将中间体1与酰肼在碱性条件下进行缩合反应得到中间体2;其中反应中所用的碱可以是无机碱或有机碱,其定义如方案一的步骤1中有关碱的定义;缩合剂可以为本领域常见的用于羧酸与胺之间的缩合反应的缩合剂,例如碳二亚胺类缩合剂(CDI、DCC、DIC、EDCI与DMAP、HOBt、HOAt、HOSu、NHPI、NHNI、PFPOH等的组合)、鎓盐类缩合剂(HATU、HBTU、HCTU、HAPyU、HBPyU、TBTU、TSTU、TNTU等)、有机膦类缩合剂(BOP、PyBOP、PyAOP、DPP-Cl、DPPA、DECP)、以及其它缩合剂(三苯基膦-多卤代甲烷、三苯基膦-六氯丙酮、三苯基膦-NBS);反应温度可以在0-100℃的范围内,优选在0-70℃的范围内,更优选在0-50℃,最优选在室温下反应。
步骤2:将中间体2通过关环反应得到中间体3;其中,关环反应是在缩合剂存在下进行的,使用的缩合剂优选Burgess试剂;反应温度可以在0-100℃的范围内,优选在0-70℃的范围内,更优选在0-50℃,最优选在室温下反应。
步骤3:将中间体3选择性脱除保护基P得到中间体4;
步骤4:将中间体4在碱性条件下与SO 3反应得到中间体5;
步骤5:将中间体5中的保护基P’去除得到产物6;
在步骤3和5中,脱保护的操作均是本领域常规的实验操作,参见例如Greene,T.W.和Wuts,P.G.M.,Greene’s Protective Groups in Organic Synthesis,第4版,John Wiley and Sons。
在步骤4中,反应中所用的碱如方案一中的步骤1中所定义的。
另一方面,本发明还提供了式(I)所示化合物或其药学上可接受的盐作为活性成分的药物组合物。该组合物包含了本发明的化 合物或其药学上可接受的盐以及药学上可接受的载体。药学上可接受的载体可以是固体或液体。其中,固体载体可以是用作赋形剂、稀释剂、甜味剂、增溶剂、润滑剂、粘合剂、片剂崩解剂、稳定剂、防腐剂或包封材料的一种或多种物质。液体载体可以是溶剂或液体分散介质。合适的固体载体包括但不限于例如纤维素、葡萄糖、乳糖、甘露醇、硬脂酸镁、碳酸镁、碳酸钠、糖精钠、蔗糖、糊精、滑石、淀粉、果胶、明胶、黄芪胶、阿拉伯胶、藻酸钠、对羟基苯甲酸酯、甲基纤维素、羧甲基纤维素钠、低熔点蜡、可可脂等。合适的液体载体包括但不限于水、乙醇、多元醇(例如甘油、丙二醇、液体聚乙二醇等)、植物油(例如花生油、棉籽油、红花油、芝麻油、橄榄油、玉米油和大豆油)、甘油酯、琼脂、无热原水、等渗盐水、林格溶液及其混合物。
制备本发明的药物组合物的方法一般是已知的。以已知的方法制备本发明的药物组合物包括常规的混合、制粒、压片、包衣、溶解或冻干方法。
本发明所述的化合物或包含其的药物组合物的治疗有效量可以通过常规实验容易地测定,最有效和方便的给药途径可以通过常规实验测定。
本发明的药物组合物可以以任何合适的给药方式施用于需要治疗的患者或者受试者,包括经口给药、肠胃外(包括皮下、肌内、静脉内以及真皮内)给药、通过吸入喷雾给药、局部给药、直肠给药、鼻腔给药、经颊给药、阴道给药或者经由植入型储器给药。在一些实施例中,提供的药物组合物是可以静脉内给药的和/或腹膜内给药的。
如在此使用的术语“肠胃外的”包括皮下的、静脉内的、肌内的、眼内的、玻璃体内的、关节内的、滑膜内的、胸骨内的、鞘内的、肝内的、腹膜内的、病灶内的以及颅内的注射或注入技术。优选地,将这些药物组合物经口给药、皮下给药、腹膜内给药或者静脉内给药。。
本发明所用适于经口施用的组合物包括固体形式,例如丸剂、片剂、囊片剂、胶囊剂(分别包括立即释放制剂、定时释放制剂和持续释放制剂)、颗粒剂和粉剂;和液体形式例如溶液剂、糖浆剂、酏剂、乳剂和混悬剂。用于眼施用的形式包括无菌溶液或眼递送装置。可用于肠胃外施用的形式包括无菌溶液剂、乳剂和混悬剂。
本发明的药物组合物还可以额外包含一种或多种β-内酰胺类抗生素。在本文中,β-内酰胺类抗生素可以包括青霉素类抗生素、头孢菌素类抗生素、单内酰胺环类抗生素、碳青霉烯类抗生素、青霉烯类酶抑制剂。具体而言,β-内酰胺类抗生素可以包括青霉素、苯唑西林、氯唑西林、双氯西林、氟氯西林、氨苄西林、匹氨西林、阿莫西林、羧苄西林、呋苄西林、磺苄西林、替卡西林、哌拉西林、美西林、头孢噻吩、头孢噻啶、头孢唑啉、头孢拉定、头孢呋辛、头孢克洛、头孢噻肟、头孢曲松、头孢他啶、头孢哌酮、头孢西丁、亚胺培南、氨曲南等。
本发明的化合物或其酯、立体异构体或其药学上可接受的盐以及包含本发明的化合物或其酯、立体异构体或其药学上可接受的盐的药物组合物可用于治疗和/或预防细菌感染导致的疾病。因此,在本发明式(I)所示的化合物、其酯、其立体异构体或其药学上可接受的盐用于制备药物中的用途中,所述药物用于治疗和/或预防细菌感染导致的疾病。此外,本发明还涉及一种抑制细菌或治疗和/或预防细菌感染导致的疾病的方法,包括向有需要的患者或受试者施用治疗和/或预防有效量的本发明的式(I)所示的化合物或其酯、立体异构体或其药学上可接受的盐。
在一个实施方案中,细菌干扰导致的疾病中的细菌选自革兰氏阳性菌和革兰氏阴性菌,其中,革兰氏阳性菌选自:金黄色葡萄球菌、表皮葡萄球菌、无乳链球菌、肺炎链球菌、化脓性链球菌、肠球菌或艰难梭菌中的一种或多种,革兰氏阴性菌选自:枸橼酸杆菌、弗氏柠檬酸杆菌、阴沟肠杆菌、肺炎克雷伯军、大肠杆菌、普通变形杆菌、沙门氏菌、粘质沙雷氏菌、志贺氏杆菌、铜绿假单胞菌、 黏膜炎莫拉菌、淋病奈瑟球菌、脑膜炎奈瑟球菌、不动杆菌、伯克氏菌、弯曲杆菌、幽门螺杆菌、霍乱弧菌、克雷伯杆菌、流感嗜血杆菌、鸟复合分支杆菌、脓肿分支杆菌、堪萨斯分支杆菌、溃疡分支杆菌、肺炎嗜衣原体、沙眼衣原体、β-溶血性链球菌、鲍曼不动杆菌、绿农假单胞菌、脆弱拟杆菌、蜡样芽胞杆菌和嗜麦芽窄食单胞菌中的一种或多种。细菌干扰导致的疾病中的细菌优选地选自革兰氏阴性菌。
在一个实施方案中,所述细菌导致的疾病选自:呼吸道感染(上呼吸道感染、下呼吸道感染、气管炎、支气管炎、肺炎、肺结核、咽炎)、尿道感染(复杂性尿道感染、并发性尿道感染、非并发性尿道感染、膀胱炎、肾盂肾炎)、中枢神经系统感染(脑炎、脑膜炎、脑脓肿、)、耳部感染(外耳炎、中耳炎)、胸膜肺和支气管感染、腹腔内感染、心血管感染(血液感染例如败血症或菌血症、心内膜炎、心肌炎、心包炎)、皮肤或软组织感染、骨与关节感染(关节炎、骨髓炎)、生殖器感染(生殖器溃疡、阴道炎、子宫颈炎)、眼部感染(结膜炎、角膜炎、眼内炎)、咽部感染(咽炎)、口腔感染(牙龈炎)中的一种或多种。
在需要时,本发明的化合物或药物组合物可以用含有一个或一个以上单位剂型的包装或分配装置提供。例如,所述包装可包含金属或塑料箔或玻璃和橡皮塞(例如在小瓶中)。所述包装或分配装置可附有药物使用说明书。
剂量取决于各种因素,包括患者的年龄、体重和状态以及给药途径。施用的精确剂量基于治疗医生的判断确定。本发明的药物组合物中的活性成分的给予的实际剂量水平和时程可以变化,以便获得以下量的活性成分,该量对于具体患者、组合物和给药方式而言可有效地达到希望的治疗应答,而对该患者没有毒性。通常,以足以减少或消除与细菌感染相关的症状的量给予本发明的药剂或药物组合物。
药剂的优选剂量是患者可承受的并且不产生严重或不可接 受的副作用的最大量。示例性剂量范围包括0.01mg至250mg/天、0.01mg至100mg/天、1mg至100mg/天、10mg至100mg/天、1mg至10mg/天以及0.01mg至10mg/天。药剂的优选剂量是患者可承受的并且不产生严重或不可接受的副作用的最大量。在实施例中,以约10微克至约100mg/千克体重/天、约0.1至约10mg/kg/天或约1.0mg至约10mg/kg体重/天的浓度给予该药剂。
在一个实施方案中,治疗有效剂量产生从约0.1ng/ml至约50-100mg/ml的药剂血清浓度。这些药物组合物典型地应该提供从约0.001mg至约2000mg的化合物/千克体重/天的剂量。例如,用于全身性给予人类患者的剂量的范围可以是1-10mg/kg、20-80mg/kg、5-50mg/kg、75-150mg/kg、100-500mg/kg、250-750mg/kg、500-1000mg/kg、1-10mg/kg、5-50mg/kg、25-75mg/kg、50-100mg/kg、100-250mg/kg、50-100mg/kg、250-500mg/kg、500-750mg/kg、750-1000mg/kg、1000-1500mg/kg、101500-2000mg/kg、5mg/kg、20mg/kg、50mg/kg、100mg/kg、500mg/kg、1000mg/kg、1500mg/kg、或2000mg/kg。制备药用单位剂型以提供每单位剂型从约1mg至约5000mg(例如从约100mg至约2500mg)的化合物或必要成分的组合。优选的单位剂量配制品是含有给予成分的如在此讨论的每日剂量或单位、每日亚剂量、或其适当部分的那些。
以下通过具体实施例的方式对本发明做进一步的说明,但这并非是对本发明的限制。本领域技术人员根据本发明的教导可以做出各种修改或调整,其并不背离本发明的精神和范围。
实施例
中间体的制备
(5S)-8-[(苄基氧基)氨基]-6-氮杂螺环[2.5]辛烷-5-羧酸苄基酯的制备步骤1:
Figure PCTCN2019073000-appb-000030
向3口圆底烧瓶中加入碳酸钾(552g,3.99mol,1.93当量)和(6S)-5-[(叔丁氧基)羰基]-5-氮杂螺环[2.4]庚烷-6-羧酸(500g,2.07mol,1.00当量)的N,N-二甲基甲酰胺溶液(1.5L)。随后在搅拌下逐滴加入苄基溴(354g,2.07mol,1.00当量)。将生成的溶液在室温下搅拌2h。然后用水(1L)淬灭反应混合物。用EA(1L*3)萃取混合物。有机层干燥并浓缩得到(6S)-5-氮杂螺环[2.4]庚烷-5,6-二羧酸6-苄基5-叔丁基二酯的粗产物750g(109%),其为黄色油状物。
步骤2:
Figure PCTCN2019073000-appb-000031
向20L圆底烧瓶中加入NaIO 4(1.2kg,5.63mol,2.70当量)在水(4L)和EA(4.5L)中的溶液和RuO 2(50g)。搅拌混合物,并逐滴加入(6S)-5-氮杂螺环[2.4]庚烷-5,6-二羧酸6-苄基5-叔丁基二酯(750g,2.26mol,1.00当量)的乙酸乙酯(1L)溶液。将生成的溶液在室温下搅拌4h。将反应混合物通过硅藻土过滤。分离溶液。水层用3x3L的乙酸乙酯萃取,合并有机层并用无水硫酸钠干燥,真空下浓缩。将粗的黄色油状物在己烷中结晶,得到560g(72%)的(6S)-4-氧代-5-氮杂螺环[2.4]庚烷-5,6-二羧酸6-苄基5-叔丁基二酯,其为灰白色固体。 1H NMR(300MHz,氯仿-d)δ0.79(dtdd,J=12.9,9.6,6.7,3.6Hz,2H),1.22(qt,J=6.6,2.8Hz,1H),1.31(ddt,J=8.3,5.3,3.1Hz,1H),1.47(s,9H),1.92(dd,J=13.3,3.2Hz,1H),2.55(dd,J=13.3,10.0Hz,1H),4.74(dd,J=10.0,3.2Hz,1H),5.24(d,J=2.1Hz,2H),7.38(d,J=2.2Hz,5H).
步骤3:
Figure PCTCN2019073000-appb-000032
向5L圆底烧瓶中加入三甲基碘化亚砜(415g,1.89mol,1.16当量) 的N,N-二甲基甲酰胺溶液(2.5L),并分批加入t-BuOK(192g,1.71mol,1.06当量)。反应在室温下搅拌2小时。然后逐滴加入(6S)-4-氧代-5-氮杂螺环[2.4]庚烷-5,6-二羧酸6-苄基5-叔丁基二酯(560g,1.62mol,1.00当量)的DMF溶液(500mL)。将生成的溶液在室温下搅拌2h。用NH 4Cl(水溶液)(2L)淬灭混合物,并用EA(2L×3)萃取。将溶剂浓缩,粗产物(500g,粗)不经进一步纯化用于下一步骤。
步骤4:
Figure PCTCN2019073000-appb-000033
向10L圆底烧瓶中加入(2S)-2-[[(叔丁氧基)羰基]氨基]-3-[1-[2-(二甲基亚磺酰亚基)乙酰基]环丙基]丙酸苄基酯(500g粗(来自步骤3),1.14mol,1.00当量)和O-苄基羟胺盐酸盐(260g,1.63mol,1.00当量(对于步骤3))的乙酸乙酯溶液(4L)。将生成的溶液在70℃搅拌过夜。将混合物过滤并浓缩滤液。通过硅胶柱(TLC:PE∶EA=5∶1)纯化粗产物得到240g(两步30%)的(2S)-3-(1-[1-[(苄基氧基)亚胺基]-2-氯乙基]环丙基)-2-[[(叔丁氧基)羰基]氨基]丙酸苄基酯,其为无色油状物。
步骤5:
Figure PCTCN2019073000-appb-000034
向10L圆底烧瓶中加入(2S)-3-(1-[1-[(苄基氧基)亚胺基]-2-氯乙基]环丙基)-2-[[(叔丁氧基)羰基]氨基]丙酸苄基酯(240g,479.03mmol,1.00当量)的乙酸乙酯溶液(2.5L),然后加入甲磺酸(137g,1.43mol,3.00当量)。反应在50℃下搅拌2小时。将混合物冷却至室温。逐滴 加入KHCO 3(240g,5.00当量)的水溶液(2L)。将生成的溶液在50℃的油浴中搅拌3小时。分离有机层并用EA萃取水层(1L*2)。除去溶剂得到180g(103%)的(5S)-8-[(苄基氧基)亚胺基]-6-氮杂螺环[2.5]辛烷-5-羧酸苄基酯的粗产物,其为黄色油状物,不经过进一步纯化用于下一步骤。1H NMR(300MHz,氯仿-d)δ7.36(d,J=11.5Hz,9H),5.20(s,2H),5.02(s,2H),4.34(d,J=16.0Hz,1H),3.83(dd,J=9.6,3.7Hz,1H),3.45(d,J=15.9Hz,1H),2.29(s,2H),2.06(dd,J=13.2,9.9Hz,1H),1.73(dd,J=13.4,3.7Hz,1H),1.42-1.26(m,1H),0.79(s,1H),0.71-0.58(m,1H),0.56-0.41(m,1H).
步骤6:
Figure PCTCN2019073000-appb-000035
向5L圆底烧瓶中加入(5S)-8-[(苄基氧基)亚胺基]-6-氮杂螺环[2.5]辛烷-5-羧酸苄基酯(180g,493.91mmol,1.00当量)的乙酸乙酯溶液(3L),在-20℃下加入硫酸(100mL,4.00当量)。在该温度下加入三乙酰氧基硼氢化钠(311g,1.46mol,3.00当量)。将生成的溶液在-20℃下搅拌5小时。反应完全。用浓NH 3.H 2O碱化混合物并添加水(1L)。混合物用EA(1L*3)萃取。将有机层干燥并浓缩。通过硅胶柱(TLC:PE∶EA=1∶1,Rf=0.35)纯化粗产物,得到109g(两步62%)的(5S)-8-[(苄基氧基)氨基]-6-氮杂螺环[2.5]辛烷-5-羧酸苄基酯,其为深红色油状物。
步骤7:
Figure PCTCN2019073000-appb-000036
向5L圆底烧瓶中加入(5S)-8-[(苄基氧基)氨基]-6-氮杂螺环[2.5]辛烷-5-羧酸苄基酯(107g,291.99mmol,1.00当量)和DIPEA(75g, 2.00当量)的二氯甲烷溶液(3L),然后在室温下加入三光气(34.5g,0.40当量)。将生成的溶液在室温下搅拌过夜。加入H 3PO 4(水溶液)(4当量)。将反应混合物在室温下继续搅拌4小时。分离有机层并浓缩。通过硅胶柱纯化粗产物得到40g(35%)期望的产物,其为黄色油状物。 1H NMR(400MHz,氯仿-d)δ-0.06-0.11(m,1H),0.30-0.45(m,2H),0.46-0.59(m,1H),1.48-1.63(m,1H),2.36(d,J=3.2Hz,1H),2.51(dd,J=15.2,8.0Hz,1H),2.98-3.16(m,2H),4.23(d,J=7.9Hz,1H),4.88(d,J=11.5Hz,1H),5.03(d,J=11.5Hz,1H),5.23(q,J=12.1Hz,2H),7.31-7.48(m,10H).
(E)-6-(苄基氧基)-7-氧代-1,6-二氮杂螺环[双环[3.2.1]辛烷-4,1′-环丙烷]-2-甲醛肟的制备
Figure PCTCN2019073000-appb-000037
步骤1:化合物2的合成
在0℃下向化合物1(10g,25.5mmol)的MeOH溶液(100mL)添加LiBH 4(19.1m L,4M,76.5mmol)的THF溶液。将溶液在室温下搅拌4小时直至反应完全。用Na 2HPO 4(10%水溶液)淬灭混合物,并用EA萃取。干燥有机层并除去溶剂得到粗产物。通过硅胶柱(PE∶EA=2∶1~EA∶MeOH=9.5∶0.5)纯化粗产物得到期望的产物(2.5g,34%),其为无色油状物。m/z(ES+),[M+H] +=289;ACID,HPLC RT=0.719min.
步骤2:化合物3的合成
在0℃下向化合物2(2.5g,8.7mmol)的DCM溶液(50mL)添加TEMPO(15mg)。在0℃下搅拌溶液并分批加入1,3,5-三氯-1,3,5-三 嗪-2,4,6-三酮(2.1g,8.7mmol)。将反应在0℃下搅拌1小时。然后将混合物通过硅藻土过滤,浓缩滤液得到粗产物,其直接用于下一步。m/z(ES+),[M+H]+=287;ACID,HPLC RT=0.764min.
步骤3:化合物4的合成
将来自步骤2的化合物3的溶液与NH 2OHHCl(786mg)溶解于MeOH(50mL)中,加入吡啶(2.7g,34.8mmol)。将反应混合物在室温下搅拌2小时。减压下除去溶剂,粗残余物通过硅胶柱(PE∶EA=2∶1)纯化得到期望的产物(1.2g,两步46%),其为无色油状物。m/z(ES+),[M+H]+=302;ACID,HPLC RT=0.706min.
实施例1化合物A的制备
Figure PCTCN2019073000-appb-000038
步骤1-4:按照化合物D的合成中的步骤2-5
ESI-MS(EI +,m/z):317,0.628min. 1H NMR(400MHz,D 2O)δ0.30-0.44(m,2H),0.48(tt,J=9.8,5.6Hz,2H),0.68(ddt,J=14.9,10.6,5.2Hz,3H),1.79(d,J=16.0Hz,2H),2.56(dd,J=16.0,7.7Hz,2H),3.09(d, J=12.2Hz,2H),3.27(dd,J=12.2,3.7Hz,2H),3.43(d,J=3.7Hz,2H),4.92(d,J=7.7Hz,2H),8.90(s,1H).
实施例2化合物B的合成
Figure PCTCN2019073000-appb-000039
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z):346,0.299min. 1H NMR(300MHz,D 2O)δ0.42(d,J=4.9Hz,1H),0.47-0.57(m,1H),0.71(ddd,J=17.0,10.3,6.1Hz,2H),1.80(d,J=16.0Hz,1H),2.59(dd,J=15.6,7.7Hz,1H),3.13(d,J=12.1Hz,1H),3.31(dd,J=12.3,3.7Hz,1H),3.47(d,J=3.7Hz,1H),4.53(s,2H),4.95(d,J=7.6Hz,1H).
实施例3化合物C的制备
Figure PCTCN2019073000-appb-000040
步骤1-6:按照化合物E的合成中的步骤1-6
ESI-MS(EI +,m/z):388,0.173min. 1H NMR(300MHz,D 2O)δ0.38(d,J=10.7Hz,1H),0.41-0.59(m,2H),0.70(dd,J=10.0,4.5Hz,1H),1.72(d,J=15.7Hz,1H),2.25(dd,J=15.8,7.9Hz,1H),3.36(dd,J=27.1,13.0Hz,3H),4.28(d,J=7.8Hz,1H),4.55(d,J=12.0Hz,1H).
实施例4化合物D的制备
Figure PCTCN2019073000-appb-000041
Figure PCTCN2019073000-appb-000042
步骤1:化合物1的合成
向50-mL密封管中加入3-(叔丁氧基羰基氨基)丙酸(1.89g,9.99mmol,1.00当量)的DCM溶液(20mL),然后加入CDI(1.94g,11.98mmol,1.20当量)。将得到的溶液在室温下搅拌1小时。然后在室温下加入NH 2NH 2.H 2O(1mL,10.00当量)继续反应30分钟。将得到的混合物在真空下浓缩。通过快速Prep-HPLC采用以下条件纯化粗产物:柱,C18硅胶;流动相,在20分钟内由ACN∶H 2O=5%增加至ACN∶H 2O=30%;检测器,UV 254nm。得到2g(99%)(3-肼基-3-氧代丙基)氨基甲酸叔丁酯,其为白色固体。
步骤2:化合物2的合成
向20-mL密封管中加入化合物1(240mg,0.79mmol,1.00当量)的THF溶液(10mL),3-肼基-3-氧代丙基氨基甲酸叔丁酯(400mg,1.97mmol,2.00当量),HATU(760mg,2.00mmol,2.00当量),DIEA(516mg,3.99mmol,3.00当量)。将得到的溶液在室温下搅拌120分钟。过滤掉固体。通过TLC采用乙酸乙酯/石油醚(1∶1)纯化残余物。得到240mg(62%)的化合物2,其为白色固体。ESI-MS(EI +,m/z):488,0.790min. 1H NMR(300MHz,甲醇-d 4)δ0.22(dt,J=9.4,4.6Hz,1H),0.35-0.47(m,1H),0.57(ddt,J=18.5,10.1,5.5Hz,2H),1.45(s,9H),1.77(d,J=15.0Hz,1H),2.46(t,J=6.7Hz,2H),3.37(d,J=4.0Hz,4H), 4.89-5.11(m,3H),7.21-7.61(m,5H).
步骤3:化合物3的合成
向20-mL密封管中加入化合物2(240mg,0.49mmol,1.00当量)的DCM溶液(5mL),DIEA(130mg,1.01mmol,2.00当量),最后加入Burgess试剂(238mg,85.00mmol,2.00当量)。将得到的溶液在室温下搅拌20小时。通过TLC采用乙酸乙酯/石油醚(1∶1)纯化残余物。得到150mg(65%)的化合物3,其为白色固体。 1H NMR(300MHz,甲醇-d 4)δ0.30(dd,J=9.7,5.1Hz,1H),0.40-0.55(m,1H),0.65(dt,J=10.9,5.4Hz,1H),0.76(dt,J=9.1,5.2Hz,1H),1.40(s,8H),1.77(d,J=15.4Hz,1H),2.61(dd,J=14.9,7.3Hz,1H),2.95-3.20(m,4H),3.42-3.55(m,2H),4.91-5.11(m,3H),7.27-7.62(m,5H).
步骤4:化合物4的合成
向50-mL圆底烧瓶中加入化合物3(150mg,0.32mmol,1.00当量)的THF溶液(15mL),然后加入Pd/C(150mg,1.00当量)。将得到的溶液在H 2下在室温下搅拌120分钟。过滤掉固体。将得到的混合物在真空下浓缩。得到150mg(124%)的化合物4,其为白色固体。ESI-MS(EI +,m/z):380,0.615min.
步骤5:化合物5的合成
向10-mL密封管中加入化合物4(150mg,0.40mmol,1.00当量)的DMF溶液(5mL),然后加入SO3.Py(300mg,1.90mmol,2.00当量)。将得到的溶液在室温下搅拌20小时。通过加入10mL的NaH 2PO 4。然后加入NBu 4HSO 4(100mg)。用乙酸乙酯萃取得到的溶液(2x20mL),合并有机层,用无水硫酸钠干燥,真空下浓缩。得到100mg(55%)的化合物5,其为白色固体。ESI-MS(EI +,m/z):460,0.725min.
步骤6:化合物D的合成
向20-mL密封管中加入化合物5(100mg,0.22mmol,1.00当量)的 DCM溶液(6mL),然后加入TFA(2mL,3.00当量)。将得到的溶液在0℃的冰水浴中搅拌120分钟。将得到的混合物在真空下浓缩。通过Prep-HPLC采用以下条件纯化粗产物:柱:XBridge Prep OBD C18柱19*250mm,5um;流动相A:水(0.1%FA),流动相B:ACN;流速:20mL/min;梯度:在7分钟内由2%B至20%B;220,254nm;Rt:6.38min。得到2.5mg(3%)的化合物D。ESI-MS(EI +,m/z):360,0.207min. 1H NMR(300MHz,D 2O)δ0.35-0.46(m,1H),0.51(td,J=7.2,3.7Hz,1H),0.71(tt,J=10.4,6.0Hz,2H),1.79(d,J=16.1Hz,1H),2.57(dd,J=15.7,8.2Hz,1H),3.15(d,J=12.1Hz,1H),3.24-3.40(m,3H),3.41-3.54(m,3H),4.91(d,J=7.6Hz,1H).
实施例5化合物E的合成
Figure PCTCN2019073000-appb-000043
步骤1:化合物2的合成
向20-mL密封管中加入化合物1(245mg,0.52mmol,1.00当量)的DCM溶液(6mL),在0℃下加入TFA(2mL,3.00当量)。将得到的溶 液在0℃的冰水浴中搅拌120分钟。将得到的混合物在真空下浓缩。得到200mg(104%)的化合物2,其为白色固体。ESI-MS(EI +,m/z):370,0.473min.
步骤2:化合物3的合成
在20-mL密封管中加入化合物2(200mg,0.54mmol,1.00当量)的甲醇溶液(5mL),然后加入TEA(350mg,3.46mmol,4.00当量),(1H-吡唑-1-基)甲烷二基亚基二氨基甲酸二叔丁酯(300mg,0.97mmol,1.10当量)。将得到的溶液在0℃的冰水浴中搅拌8小时。通过TLC采用乙酸乙酯/石油醚(1∶1)纯化残余物。得到200mg(60%)的化合物3,其为白色固体。ESI-MS(EI +,m/z):612,1.073min. 1H NMR(300MHz,甲醇-d 4)δ0.29(dd,J=9.6,4.9Hz,1H),0.39-0.55(m,1H),0.65(dt,J=10.8,5.3Hz,1H),0.71-0.85(m,1H),1.50(d,J=15.4Hz,18H),1.77(d,J=15.5Hz,1H),2.80(d,J=3.5Hz,1H),2.94-3.05(m,2H),3.20(t,J=6.4Hz,2H),3.75-3.94(m,2H),4.91-5.09(m,3H),7.32-7.57(m,5H).
步骤3:化合物4的合成
向50-mL圆底烧瓶中加入化合物3(200mg,0.33mmol,1.00当量)的THF溶液(10mL),然后加入Pd/C(200mg,2.00当量)。将产生的溶液在H 2(1atm)下在室温下搅拌120分钟。过滤掉固体。将产生的混合物在真空下浓缩。得到200mg(117%)的化合物4,其为白色固体。ESI-MS(EI +,m/z):522,0.865min.
步骤4:化合物5的合成
在20-mL密封管中加入化合物4(200mg,0.38mmol,1.00当量)的DMF溶液(5mL),然后加入SO3.Py(361mg,7.00当量)。将产生的溶液在室温下搅拌20小时。将产生的混合物在真空下浓缩。随后通过加入10mL的NaH 2PO 4淬灭反应。向反应混合物中加入NBu 4HSO 4(100mg)。用乙酸乙酯萃取产生的溶液(2x20mL),合并有 机层,用无水硫酸钠干燥,真空下浓缩。得到100mg(43%)化合物5,其为白色固体。ESI-MS(EI +,m/z):602,1.023min.
步骤5:化合物E的合成
在20-mL密封管中加入化合物5(100mg,0.17mmol,1.00当量)的DCM溶液(6ml),然后在0℃下加入TFA(2mL,3.00当量)。将产生的溶液在0℃的冰水浴中搅拌120分钟。将产生的混合物在真空下浓缩。通过Prep-HPLC采用以下条件纯化粗产物:柱:XBridge Prep OBD C18柱19*250mm,5um;流动相A:水(0.1%FA),流动相B:ACN;流速:20mL/min;梯度:在7分钟内由12%B至23%B;220,254nm;Rt:6.2min;得到8mg(12%)的化合物E。ESI-MS(EI +,m/z):402,0.418min. 1H NMR(300MHz,D 2O)δ0.34-0.47(m,1H),0.47-0.58(m,1H),0.70(ddt,J=14.7,10.4,5.9Hz,2H),1.77(d,J=16.1Hz,1H),2.57(dd,J=16.5,7.8Hz,1H),3.21(t,J=6.4Hz,2H),3.25-3.37(m,1H),3.46(d,J=3.8Hz,1H),3.64(t,J=6.4Hz,2H),4.90(d,J=7.7Hz,1H).
实施例6化合物F的制备
Figure PCTCN2019073000-appb-000044
Figure PCTCN2019073000-appb-000045
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z):374,0.240min. 1H NMR(300MHz,D 2O)δ0.41(s,1H),0.52(s,1H),0.71(t,J=8.6Hz,2H),1.78(d,J=15.9Hz,1H),2.14(p,J=7.4Hz,2H),2.57(dd,J=15.7,8.0Hz,1H),2.98-3.21(m,5H),3.30(d,J=12.0Hz,1H),3.46(d,J=3.8Hz,1H),4.89(d,J=7.5Hz,2H).
实施例7化合物G的制备
Figure PCTCN2019073000-appb-000046
Figure PCTCN2019073000-appb-000047
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z):388,0.273min. 1H NMR(300MHz,D 2O)δ0.42(d,J=4.9Hz,1H),0.47-0.58(m,1H),0.70(ddt,J=14.8,10.4,5.1Hz,2H),1.73(q,J=9.2,7.9Hz,2H),1.83(dd,J=14.9,7.9Hz,2H),2.56(dd,J=15.7,8.0Hz,1H),2.98(dt,J=10.3,7.4Hz,3H),3.10(d,J=12.1Hz,1H),3.30(dd,J=12.2,3.7Hz,1H),3.46(d,J=3.7Hz,1H),4.89(d,J=7.8Hz,2H).
实施例8化合物H的制备
Figure PCTCN2019073000-appb-000048
Figure PCTCN2019073000-appb-000049
步骤1-5:按照化合物I的合成中的步骤1-6
步骤6:按照化合物I的合成中的步骤8
步骤7-8:按照化合物E的合成中的步骤1-2
步骤9:向氮气保护的100-mL3口圆底烧瓶中加入化合物8(1.8g,2.87mmol,1.00当量),然后加入三苯基膦(1.507g,5.75mmol,2.00当量)的THF溶液(50mL),DIAD(1.159g,5.74mmol,2.00当量)的THF溶液(10mL)。将得到的溶液在室温下搅拌5小时。将得到的溶液在真空下浓缩。通过快速Prep-HPLC采用以下条件纯化粗产物:柱,C18硅胶;流动相,在30分钟内由ACN=0%增加至ACN=50%;检测器,UV 254nm。得到1.3g(74%)的化合物9,其为白色固体。ESI-MS(EI +,m/z):610,1.140min.
步骤10-12:按照化合物E的合成中的步骤3-5
ESI-MS(EI +,m/z):400,0.628min. 1H NMR(300MHz,D 2O)δ0.42(d,J=5.3Hz,1H),0.50(dt,J=7.9,5.0Hz,1H),0.60-0.78(m,2H), 1.75(d,J=16.1Hz,1H),2.56(dd,J=16.1,7.6Hz,1H),3.10(dd,J=12.1,3.9Hz,1H),3.29(dd,J=12.1,3.4Hz,1H),3.45(d,J=3.5Hz,1H),3.95(dt,J=10.3,5.0Hz,1H),4.14(t,J=10.3Hz,1H),4.92(d,J=7.5Hz,1H),5.47(dd,J=10.0,4.8Hz,1H).
实施例9化合物I的制备
Figure PCTCN2019073000-appb-000050
步骤1:化合物1的合成
向500-mL圆底烧瓶中加入3-氨基-2-羟基丙酸(5g,47.58mmol,1.00当量)的二噁烷溶液(200mL),然后加入NaOH(4g,100.01mmol,2.10当量)的水溶液(100mL)。随后,将得到的溶液在室温下搅拌5分钟。然后加入(Boc) 2O(12g,54.98mmol,1.14当量)。将得到的溶液在室温下继续搅拌5小时。用EA洗涤得到的混合物(2x200mL)。用HCl(1mol/L)调节水相的pH至1。将得到的溶液用乙酸乙酯萃取(3x200mL)并且合并有机层,用无水硫酸钠干燥,真空下浓缩。得到8.6g(88%) 的3-(叔丁氧基羰基氨基)-2-羟基丙酸,其为无色油状物。ESI-MS(EI +,m/z+Na):228,0.793min.
步骤2:化合物2的合成
向300-mL圆底烧瓶中加入3-(叔丁氧基羰基氨基)-2-羟基丙酸(8.6g,41.91mmol,1.00当量)的THF溶液(150mL),TEA(0mg,3.00当量),TBS-Cl(8.5g,56.40mmol,1.40当量)。将得到的溶液在室温下搅拌20小时。用HCl(1mol/L)调节pH至1。用乙酸乙酯萃取得到的溶液(2x300mL)并合并有机层,无水硫酸钠干燥,真空下浓缩。得到14g(105%)的3-(叔丁氧基羰基氨基)-2-(叔丁基二甲基硅氧基)丙酸,其为白色固体。ESI-MS(EI +,m/z+Na):342,1.324min.
步骤3:化合物3的合成
向100-mL圆底烧瓶中加入3-(叔丁氧基羰基氨基)-2-(叔丁基二甲基硅氧基)丙酸(7g,21.91mmol,1.00当量)的DCM溶液(50mL)和CDI(5.34g,32.96mmol,1.50当量)。将得到的溶液在室温下搅拌120分钟,然后加入肼(11g,220.00mmol,10.00当量)。将得到的溶液在室温下继续搅拌60分钟。将得到的混合物在真空下浓缩。通过快速Prep-HPLC采用以下条件纯化粗物质:柱,C18硅胶;流动相,在30分钟内由ACN增加至ACN=50%;检测器,UV 254nm。得到2.86g(39%)的2-(叔丁基二甲基硅氧基)-3-肼基-3-氧代丙基氨基甲酸叔丁酯,其为白色固体。ESI-MS(EI +,m/z+):334,1.081min.
步骤4:化合物4的合成
向20-mL密封管中加入化合物3(364mg,1.20mmol,1.00当量),2-(叔丁基二甲基硅氧基)-3-肼基-3-氧代丙基氨基甲酸叔丁酯(442mg,1.33mmol,1.10当量),HATU(912mg,2.40mmol,2.00当量),DIPEA(645mg,5.00mmol,4.00当量)的THF溶液(10mL)。将得到的溶液在室温下搅拌2小时。过滤掉固体。通过TLC乙酸乙酯/石油醚(1/1)纯化残余物。得到730mg(98%)的化合物4,其作为白色固体。ESI-MS (EI +,m/z+):618,1.361min.
步骤5:化合物5的合成
向20-mL密封管中加入化合物4(730mg,1.18mmol,1.00当量)的DCM溶液(10mL),然后加入DIEA(915mg,3.00当量),Burgess试剂(1126mg,3.00当量)。将得到的溶液在室温下搅拌20小时。通过TLC采用乙酸乙酯/石油醚(1/1)纯化残余物。得到450mg(63%)的化合物5,其为白色固体。ESI-MS(EI +,m/z+):600,1.441min.
步骤6:化合物6的合成
向50-mL圆底烧瓶中加入化合物5(900mg,1.50mmol,1.00当量)的THF溶液(20mL),然后加入Pd/C(100mg,0.10当量)。将得到的溶液在H 2下在室温下搅拌2小时。过滤掉固体。将得到的混合物在真空下浓缩。得到700mg(92%)的化合物6,其为白色固体。ESI-MS(EI +,m/z+):510,1.246min.
步骤7:化合物7的合成
向20-mL密封管中加入化合物6(700mg,1.37mmol,1.00当量)的DMF溶液(5mL),然后加入SO3.Py(1.1g,6.96mmol,5.00当量)。将得到的溶液在室温下搅拌20小时。通过快速Prep-HPLC采用以下条件纯化粗产物:柱,C18硅胶;流动相,在30分钟内由ACN增加至ACN=50%;检测器,UV 254nm。得到500mg(62%)化合物7,其为浅黄色固体。ESI-MS(EI +,m/z+Na):590,1.336min.
步骤8:化合物8的合成
向50-mL圆底烧瓶中加入化合物7(500mg,0.85mmol,1.00当量)的THF溶液(10mL),然后加入3HF.Et 3N(4mL,10.00当量)。将生成的溶液在室温下搅拌20小时。通过快速Prep-HPLC采用以下条件纯化粗产物:柱,C18硅胶;流动相,在30分钟内由ACN增加至ACN=50%;检测器,UV 254nm。得到300mg(74%)的化合物8,其为白色固体。ESI-MS(EI +,m/z+):476,0.861min.
步骤9:化合物I的合成
向50mL圆底烧瓶中加入化合物8(300mg,0.63mmol,1.00当量)的DCM溶液(10mL),然后加入TFA(3mL,10.00当量)。将生成的溶液在0℃下搅拌120分钟。将生成的混合物在真空下浓缩。通过Prep-HPLC采用以下条件纯化粗产物:柱:XBridge Prep OBD C18柱19*250mm,5um;流动相A:水(0.1%FA),流动相B:ACN;流速:20mL/min;梯度:在7分钟内由2%B至18%B;254,220nm;Rt:6.25min;得到100mg(42%)的化合物I。ESI-MS(EI +,m/z+):376,0.123min. 1H NMR(300MHz,D 2O)δ0.27-0.42(m,1H),0.48(tt,J=9.2,5.3Hz,1H),0.67(ddt,J=14.7,10.4,5.1Hz,2H),1.77(d,J=16.0Hz,1H),2.55(dd,J=15.9,7.7Hz,1H),3.11(d,J=12.1Hz,1H),3.28(dd,J=12.1,3.7Hz,1H),3.38-3.49(m,2H),3.54(dd,J=13.4,3.9Hz,1H),4.91(d,J=7.6Hz,1H),5.28(dd,J=8.4,3.9Hz,1H).
实施例10化合物J的制备
Figure PCTCN2019073000-appb-000051
步骤1-6:按照化合物E的合成中的步骤1-6
ESI-MS(EI +,m/z+):418,0.942min. 1H NMR(300MHz,D 2O)δ0.33-0.46(m,1H),0.50(td,J=7.3,4.0Hz,1H),0.69(ddt,J=14.6, 10.3,5.2Hz,2H),1.77(d,J=16.0Hz,1H),2.57(dd,J=15.9,7.9Hz,1H),3.08(d,J=12.1Hz,1H),3.30(dd,J=12.1,3.8Hz,1H),3.45(d,J=3.7Hz,1H),3.61-3.79(m,2H),4.92(d,J=7.6Hz,1H),5.15-5.29(m,1H).
实施例11化合物K的制备
Figure PCTCN2019073000-appb-000052
步骤1-6:按照化合物E的合成中的步骤1-6
ESI-MS(EI +,m/z+):418,0.716min. 1H NMR(300MHz,D 2O)δ0.41(d,J=5.1Hz,1H),0.45-0.58(m,1H),0.69(ddt,J=14.7,10.3,5.2Hz,1H),1.78(d,J=16.0Hz,1H),2.58(dd,J=16.1,7.8Hz,1H),3.08(d,J=12.1Hz,1H),3.30(dd,J=12.0,3.7Hz,1H),3.46(d,J=3.6Hz,1H),3.63-3.81(m,2H),4.92(d,J=7.5Hz,1H),5.17-5.25(m,1H).
实施例12化合物L的制备
Figure PCTCN2019073000-appb-000053
Figure PCTCN2019073000-appb-000054
步骤1-6:按照化合物E的合成中的步骤1-6
ESI-MS(EI +,m/z+):418,0.702min. 1H NMR(300MHz,D 2O)δ0.32-0.45(m,1H),0.45-0.57(m,1H),0.70(ddt,J=15.1,10.4,5.2Hz,2H),1.78(d,J=16.0Hz,1H),2.58(dd,J=15.8,7.7Hz,1H),3.09(d,J=12.1Hz,1H),3.31(dd,J=12.3,4.2Hz,1H),3.46(d,J=3.7Hz,1H),3.64-3.78(m,2H),4.93(d,J=7.6Hz,1H),5.20(t,J=5.2Hz,1H).
实施例13化合物M的制备
Figure PCTCN2019073000-appb-000055
步骤1-3:按照化合物D的合成中的步骤1-3
步骤4-8:按照化合物E的合成中的步骤1-5
ESI-MS(EI +,m/z+):428,0.323min. 1H NMR(400MHz,D 2O)δ0.37(dd,J=9.5,4.9Hz,1H),0.41-0.52(m,1H),0.60(dt,J=10.5,5.1Hz,1H),0.68(dt,J=9.6,5.1Hz,1H),1.15-1.25(m,2H),1.30-1.43(m,2H),1.68(d,J=16.1Hz,1H),2.50(dd,J=16.0,7.8Hz,1H),3.04(d,J=12.1Hz,1H),3.23(dd,J=12.2,3.5Hz,1H),3.40(d,J=3.7Hz,1H),3.55(s,2H),4.80(d,J=7.6Hz,1H).
实施例14化合物N的制备
Figure PCTCN2019073000-appb-000056
步骤3-8:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z+):401,0.223min. 1H NMR(300MHz,D 2O)δ0.42(d,J=4.9Hz,1H),0.45-0.58(m,1H),0.62-0.78(m,2H),1.78(d,J=15.9Hz,1H),2.57(dd,J=16.0,7.9Hz,1H),3.10(d,J=12.1Hz,1H),3.21-3.37(m,1H),3.46(d,J=3.7Hz,1H),3.81-4.01(m,2H),4.07(s,2H),4.10-4.29(m,2H),4.92(d,J=7.6Hz,1H).
实施例15化合物O的制备
Figure PCTCN2019073000-appb-000057
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z+):386,0.642min. 1H NMR(300MHz,D 2O)δ0.30-0.45(m,1H),0.48(td,J=7.5,6.8,3.6Hz,1H),0.68(ddt,J=17.5,10.3,6.0Hz,2H),1.76(d,J=16.0Hz,1H),2.54(dd,J=15.6,7.7Hz,3H),2.72-2.91(m,2H),3.09(d,J=12.1Hz,1H),3.27(dd,J=12.2,3.6Hz,1H),3.44(d,J=3.6Hz,1H),3.67(ddd,J=18.0,9.8,8.2Hz,1H),3.91(p, J=8.7Hz,1H),4.87(d,J=7.6Hz,1H).
实施例16化合物P的制备
Figure PCTCN2019073000-appb-000058
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z+):386,2.005min. 1H NMR(300MHz,D 2O)δ0.33-0.45(m,1H),0.45-0.55(m,1H),0.68(ddt,J=14.5,10.3,5.1Hz,2H),1.76(d,J=16.0Hz,1H),2.55(dd,J=16.3,8.0Hz,1H),2.72(dd,J=10.5,6.4Hz,4H),3.10(d,J=12.1Hz,1H),3.28(dd,J=12.2,3.9Hz,1H),3.44(d,J=3.7Hz,1H),3.84-3.99(m,1H),3.99-4.18(m,1H),4.88(d,J=7.6Hz,1H).
实施例17化合物Q的制备
Figure PCTCN2019073000-appb-000059
Figure PCTCN2019073000-appb-000060
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z+):400,682min. 1H NMR(300MHz,D 2O)δ0.40(t,J=7.2Hz,1H),0.49(tt,J=8.7,5.2Hz,1H),0.69(ddt,J=14.8,10.2,5.0Hz,2H),1.71-1.90(m,2H),1.90-2.14(m,2H),2.14-2.30(m,2H),2.50-2.62(m,1H),2.67(dd,J=13.5,7.7Hz,1H),3.10(d,J=12.1Hz,1H),3.28(dd,J=12.1,3.7Hz,1H),3.45(d,J=3.6Hz,1H),3.55(p,J=8.1Hz,1H),3.80(p,J=7.7Hz,1H),4.87(d,J=7.6Hz,1H).
实施例18化合物R的制备
Figure PCTCN2019073000-appb-000061
Figure PCTCN2019073000-appb-000062
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z+):400,0.695min. 1H NMR(300MHz,D 2O)δ0.40(t,J=7.3Hz,1H),0.48(td,J=9.4,8.6,5.2Hz,1H),0.69(ddt,J=17.5,10.1,6.0Hz,2H),1.70-1.89(m,2H),1.99(ddt,J=28.2,13.5,9.5Hz,2H),2.13-2.31(m,2H),2.56(dt,J=15.9,7.8Hz,1H),2.67(dd,J=13.6,7.6Hz,1H),3.10(d,J=12.1Hz,1H),3.28(dd,J=12.1,3.6Hz,1H),3.44(d,J=3.6Hz,1H),3.49-3.67(m,1H),3.79(p,J=7.6Hz,1H),4.87(d,J=7.6Hz,1H).
实施例19化合物S的制备
Figure PCTCN2019073000-appb-000063
步骤1:化合物1的合成
向250-mL圆底烧瓶中加入3-氧代环丁基氨基甲酸叔丁酯(7.4g,39.95mmol,1.00当量)的甲苯溶液(80mL),然后加入(三苯基亚膦基)乙酸乙酯(15.31g,43.95mmol,1.10当量)。将生成的溶液在100℃的油浴中搅拌120分钟。将反应混合物在真空下浓缩。通过快速Prep-HPLC采用以下条件纯化粗产物:柱,硅胶;流动相,在30分钟内由EA/PE=10%增加至EA/PE=60%;检测器,UV 254nm。得到6.2g(61%)的ethyl 2-(3-(叔丁氧基羰基氨基)环丁亚基)乙酸乙酯,其为无色油状物。ESI-MS(EI +,m/z 2n+1):511,0.906min.
步骤2:化合物2的合成
向250-mL圆底烧瓶中加入2-(3-(叔丁氧羰基氨基)环丁亚基)乙酸乙酯(6.2g,24.28mmol,1.00当量)的甲醇溶液(100mL),然后加入钯炭(1.8g,0.10当量)。将生成的溶液在H 2(1atm)下在室温下搅拌100分钟。过滤掉固体。将生成的混合物在真空下浓缩。得到6.2g(99%)2-(3-[[(叔丁氧基)羰基]氨基]环丁基)乙酸乙酯,其为白色固体。ESI-MS(EI +,m/z+Na):280,1.172min.
步骤3-8:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z+):400,0.668min. 1H NMR(300MHz,D 2O)δ0.30 -0.42(m,1H),0.42-0.53(m,1H),0.66(ddt,J=14.7,10.2,5.0Hz,2H),1.73(d,J=16.0Hz,1H),1.82-2.01(m,2H),2.55(dq,J=15.3,8.2Hz,4H),2.93-3.14(m,3H),3.26(dd,J=12.0,3.6Hz,1H),3.42(d,J=3.5Hz,1H),3.68(p,J=8.5Hz,1H),4.85(d,J=7.5Hz,1H).
实施例20化合物T的制备
Figure PCTCN2019073000-appb-000064
步骤1-3:按照化合物D的合成中的步骤4-6
ESI-MS(EI +,m/z+):400,0.654min. 1H NMR(300MHz,D 2O)δ0.39(d,J=4.8Hz,1H),0.42-0.53(m,1H),0.67(ddt,J=19.5,9.8,5.5Hz,2H),1.73(d,J=16.0Hz,1H),2.12-2.28(m,2H),2.28-2.40(m,2H),2.53(dd,J=15.9,7.7Hz,1H),2.87(s,1H),3.05-3.17(m,2H),3.26(dd,J=12.1,3.3Hz,1H),3.42(d,J=3.4Hz,1H),3.88(p,J=7.2Hz,1H),4.85(d,J=7.6Hz,1H).
实施例21化合物U的制备
Figure PCTCN2019073000-appb-000065
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z+):386,0.654min. 1H NMR(300MHz,D 2O)δ0.39 (t,J=7.3Hz,1H),0.43-0.53(m,1H),0.68(ddt,J=17.7,10.1,5.9Hz,2H),0.96-1.17(m,4H),1.76(d,J=16.0Hz,1H),2.55(dd,J=16.0,7.9Hz,1H),3.10-3.27(m,2H),3.32(s,2H),3.43(d,J=3.5Hz,1H),4.89(d,J=7.6Hz,1H).
实施例22化合物V的制备
Figure PCTCN2019073000-appb-000066
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z+):400,0.668min. 1H NMR(300MHz,D 2O)δ0.30-0.43(m,1H),0.43-0.55(m,1H),0.58-0.75(m,2H),1.07(h,J=3.8Hz,2H),1.10-1.24(m,2H),1.75(d,J=16.0Hz,1H),2.55(dd,J=16.0,7.8Hz,1H),2.71(s,3H),3.14(d,J=12.1Hz,1H),3.28(dd,J=12.3,3.6Hz,1H),3.40(s,2H),3.43(d,J=3.7Hz,1H),4.89(d,J=7.6Hz,1H).
实施例23化合物W的制备
Figure PCTCN2019073000-appb-000067
Figure PCTCN2019073000-appb-000068
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z+):386,0.636min. 1H NMR(300MHz,D 2O)δ0.33-0.44(m,1H),0.44-0.56(m,1H),0.59-0.79(m,2H),1.75(d,J=16.1Hz,1H),2.55(dd,J=15.9,7.8Hz,1H),3.09(d,J=12.1Hz,1H),3.21-3.36(m,3H),3.44(q,J=6.2Hz,2H),3.93-4.08(m,2H),4.16-4.36(m,2H),4.87(d,J=7.6Hz,1H).
实施例24化合物X的制备
Figure PCTCN2019073000-appb-000069
Figure PCTCN2019073000-appb-000070
步骤1-6:按照化合物D的合成中的步骤1-6
ESI-MS(EI +,m/z+):374,0.615min. 1H NMR(300MHz,D 2O)δ0.33-0.45(m,1H),0.49(td,J=7.3,3.6Hz,1H),0.68(ddt,J=14.6,10.3,5.1Hz,2H),1.76(d,J=16.0Hz,1H),2.55(dd,J=15.9,7.7Hz,1H),2.73(s,3H),3.13(d,J=12.1Hz,1H),3.27(dd,J=12.1,3.8Hz,1H),3.32-3.41(m,2H),3.41-3.54(m,3H),4.88(d,J=7.6Hz,1H).
实施例25化合物Y的制备
Figure PCTCN2019073000-appb-000071
步骤1-5:按照化合物FF的合成中的步骤2-6
ESI-MS(EI +,m/z+):385,0.761min. 1H NMR(300MHz,D 2O)δ0.33(s,1H),0.39-0.49(m,1H),0.49-0.61(m,1H),0.65(dd,J=9.5,4.2Hz,1H),1.64(d,J=15.7Hz,1H),2.46(dd,J=16.4,7.6Hz,1H),2.62(t,J= 7.4Hz,3H),3.05-3.22(m,2H),3.38(d,J=3.5Hz,1H),3.79(d,J=8.1Hz,1H),3.91-4.11(m,1H),4.73(s,1H),6.35(d,J=7.7Hz,1H),8.35(s,0H).
实施例26化合物Z的制备
Figure PCTCN2019073000-appb-000072
步骤1-5:按照化合物FF的合成中的步骤2-6
ESI-MS(EI +,m/z+):385,0.774min. 1H NMR(300MHz,D 2O)δ0.32(dt,J=9.3,4.7Hz,1H),0.37-0.48(m,1H),0.56(dt,J=10.2,5.0Hz,1H),0.66(dt,J=9.6,5.0Hz,1H),1.63(d,J=15.8Hz,1H),2.29-2.52(m,3H),2.64-2.84(m,2H),3.03-3.29(m,2H),3.37(d,J=3.4Hz,1H),3.53(ddd,J=18.0,9.9,8.1Hz,1H),3.73-3.96(m,1H),4.72(s,1H),6.32(s,1H).
实施例27化合物AA的制备
Figure PCTCN2019073000-appb-000073
Figure PCTCN2019073000-appb-000074
步骤1-4:按照化合物D的合成中的步骤2、4-6
ESI-MS(EI +,m/z+):375,0.716min. 1H NMR(400MHz,D 2O)δ0.29(dt,J=8.1,4.2Hz,1H),0.41(dtd,J=18.5,9.0,4.5Hz,2H),0.63(dt,J=10.2,4.7Hz,1H),1.59-1.77(m,3H),1.98-2.10(m,2H),2.14(dd,J=15.6,7.9Hz,1H),2.97-3.11(m,3H),3.27(dd,J=11.9,3.7Hz,1H),3.31-3.45(m,3H),3.95(tt,J=11.0,4.0Hz,1H),4.05(d,J=7.8Hz,1H).
实施例28化合物BB的制备
Figure PCTCN2019073000-appb-000075
步骤1-4:按照化合物D的合成中的步骤2、4-6
ESI-MS(EI +,m/z+):361,0.662min. 1H NMR(400MHz,D 2O)δ0.31(dd,J=9.8,4.6Hz,1H),0.42(dtd,J=18.5,9.0,4.5Hz,2H),0.64(dt,J=10.2,4.6Hz,1H),1.66(d,J=15.6Hz,1H),2.02(tt,J=16.0,8.5Hz,1H),2.14(dd,J=15.6,7.9Hz,1H),2.31(dp,J=13.3,6.7,6.0Hz,1H),3.08(dd,J=11.8,8.8Hz,1H),3.17-3.32(m,2H),3.34(dd,J=11.1,3.6Hz,2H),3.43(dt,J=12.1,7.5Hz,1H),3.52(dt,J=12.5,8.2Hz,1H),4.06(d,J=7.8Hz,1H),4.48(dq,J=12.1,6.8,6.1Hz,1H).
实施例29化合物CC的制备
Figure PCTCN2019073000-appb-000076
步骤1-4:按照化合物D的合成中的步骤2、4-6
ESI-MS(EI +,m/z+):347,0.572min. 1H NMR(400MHz,D 2O)δ0.29(dd,J=9.8,4.2Hz,1H),0.41(tp,J=9.1,4.3Hz,2H),0.60-0.70(m,1H),1.65(d,J=15.6Hz,1H),2.15(dd,J=15.7,7.9Hz,1H),3.07(d,J=11.9Hz,1H),3.29(dd,J=11.9,3.7Hz,1H),3.35(d,J=3.8Hz,1H),4.07(d,J=7.9Hz,1H),4.11-4.22(m,2H),4.28(dd,J=11.3,8.8Hz,2H),4.75(d,J=7.9Hz,1H).
实施例30化合物DD的制备
Figure PCTCN2019073000-appb-000077
步骤1:化合物1的合成
向500mL圆底烧瓶中加入(S)-3-氨基-2-羟基丙酸(20g,190.31mmol,1.00当量)的MeOH溶液(250mL)。在0℃下将SOCl 2(56g,470.71mmol,2.50当量)加入混合物中。将生成的溶液在65℃的油浴中搅拌15小时。将生成的混合物在真空下浓缩。得到22g(97%)的(S)-3-氨基-2-羟基丙酸甲酯,其为无色油状物。ESI-MS(EI +,m/z+):120,0.156min.
步骤2:化合物2的合成
向100mL圆底烧瓶中加入N-甲基-N-(2-氧代乙基)氨基甲酸叔丁酯(5.2g,30.02mmol,1.00当量)、(S)-3-氨基-2-羟基丙酸甲酯(4.3g,36.10mmol,1.20当量)的MeOH溶液(30mL)和三乙酰氧基硼氢化钠(12.72g,60.02mmol,2.00当量)。将生成的溶液在室温下搅拌2小时。将生成的混合物在真空下浓缩。通过快速Prep-HPLC采用以下条件纯化粗产物:柱,C18硅胶;流动相在60分钟内由ACN=10%增加至ACN=50%;检测器,UV 254nm。得到3g(36%)的(S)-3-(2-(叔丁氧羰基(甲基)氨基)乙基氨基)-2-羟基丙酸甲酯,其为无色油状物。(EI +,m/z+):277,0.381min。
步骤3:化合物3的合成
向100mL圆底烧瓶中加入(S)-3-(2-(叔丁氧基羰基(甲基)氨基)乙基氨基)-2-羟基丙酸甲酯(5g,18.09mmol,1.00当量),(Boc) 2O(4.8g,21.99mmol,1.20当量),TEA(2.7g,26.68mmol,1.50当量)的DCM溶液(20mL)。将生成的溶液在室温下搅拌15小时。将生成的混合物在真空下浓缩。通过快速Prep-HPLC采用以下条件纯化粗产物:柱,C18硅胶;流动相,在50分钟内由ACN=10%增加至ACN=60%;检测器,UV 254nm。得到2.5g(37%)的(S)-3-(叔丁氧基羰基(2-(叔丁氧基羰基(甲基)氨基)乙基)氨基)-2-羟基丙酸甲酯,其为无色油状物。(EI +,m/z+):377,0.879min.
步骤4:化合物4的合成
向100mL圆底烧瓶中加入(S)-3-(叔丁氧基羰基(2-(叔丁氧基羰基(甲基)氨基)乙基)氨基)-2-羟基丙酸甲酯(2.5g,6.64mmol,1.00当量),TEA(1g,9.88mmol,1.50当量),DMAP(100mg,0.10当量),TBSCl(1.2g,7.96mmol,1.20当量)的DCM溶液(30mL)。将生成的溶液在室温下搅拌15小时。将生成的混合物在真空下浓缩。通过快速Prep-HPLC采用以下条件纯化粗产物:柱,C18硅胶;流动相,50分钟内有ACN=10%增加至ACN=60%;检测器,UV 254nm。得到850mg(26%)的化合物4,其为白色固体。(EI +,m/z+):513,1.611min.
步骤5-9:按照化合物D的合成的步骤1-5
步骤10-11:按照化合物I的合成中的步骤8-9
(EI +,m/z+):433,0.548min. 1H NMR(300MHz,D 2O)δ0.39(d,J=5.1Hz,1H),0.46(d,J=14.0Hz,1H),0.54-0.76(m,2H),1.76(d,J=16.0Hz,1H),2.54(dd,J=15.9,7.8Hz,1H),2.72(s,3H),3.10(d,J=12.1Hz,1H),3.28(d,J=12.3Hz,1H),3.46(dt,J=18.5,11.3Hz,4H),3.59(d,J=8.1Hz,2H),4.90(d,J=7.4Hz,1H),5.24-5.42(m,1H).
实施例31化合物EE的制备
Figure PCTCN2019073000-appb-000078
步骤1-4:按照化合物DD的合成中步骤1-4
步骤5-9:按照化合物D的合成中的步骤1-5
步骤10-11:按照化合物I的合成中的步骤8-9
(EI +,m/z+):419,0.534min. 1H NMR(300MHz,D 2O)δ0.31-0.44(m,1H),0.44-0.54(m,1H),0.68(ddt,J=18.1,10.3,5.9Hz,2H),1.77(d,J=16.0Hz,1H),2.55(dd,J=16.1,7.8Hz,1H),3.10(d,J=12.1Hz, 1H),3.28(dd,J=12.1,3.7Hz,1H),3.35(dd,J=10.3,4.2Hz,2H),3.45(dd,J=7.8,5.0Hz,3H),3.52-3.69(m,2H),4.90(d,J=7.6Hz,1H),5.34(dd,J=8.0,4.3Hz,1H).
实施例32化合物FF的制备
Figure PCTCN2019073000-appb-000079
步骤1:化合物2的合成
将氯吡咯烷-2,5-二酮(0.146g,1.10mmol)添加至(E)-7-(苄基氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-甲醛肟(0.3g,1.00mmol)的DCM溶液(8mL)中,随后向其中加入吡啶(1滴)。将生成的混合物在室温下搅拌16小时。减压下除去溶剂得到(1R,4S,Z)-7-(苯基氧基)-N-羟基-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-碳酰亚胺基氯(0.330g,99%),其为棕色粘稠物。产物不经进一步纯化直接用于下一步。m/z(ES+),[M+H]+=336;ACID,HPLC tR=1.092min.
步骤2:化合物3的合成
将甲基(丙-2-炔-1-基)氨基甲酸叔丁酯(0.200g,1.18mmol)添加至(1R,4S,Z)-7-(苄基氧基)-N-羟基-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙基]-4-碳酰亚胺基氯(0.33g,0.98mmol)的DCM溶液(8mL)中,随后添加TEA(0.137mL,0.98mmol)。将生成的混合物在室温下搅拌16小时。减压下除去溶剂。通过快速C18快速色谱纯化粗产物,梯度洗脱(0至65%MeCN水溶液)。蒸发纯的级分至干提供((3-((1R,4S)-7-(苄基氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)异噁唑-5-基)甲基)(甲基)氨基甲酸叔丁酯(0.160g,34.7%),其为白色固体。m/z(ES+),[M+H]+=469;ACID,HPLC tR=1.305min. 1H NMR(300MHz,氯仿-d)δ1.48(s,10H),2.22(t,J=2.5Hz,1H),2.93(s,3H),4.05(s,2H).
步骤3:化合物4的合成
在室温下在氢气气氛下将((3-((1R,4S)-7-(苄基氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)异噁唑-5-基)甲基)(甲基)氨基甲酸叔丁酯(0.16g,0.34mmol)和Pd-C(0.036g,0.34mmol)的THF溶液(10mL)搅拌8小时。将固体过滤掉,减压下除去溶剂得到((3-((1R,4S)-7-羟基-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)异噁唑-5-基)甲基)(甲基)氨基甲酸叔丁酯(0.110g,85%),其为固体。m/z(ES+),[M+H]+=379;ACID,HPLC tR=1.047min.
步骤4:化合物5的合成
将吡啶化合物与三氧化硫(0.278g,1.74mmol)添加至((3-((1R,4S)-7-羟基-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)异噁唑-5-基)甲基)(甲基)氨基甲酸叔丁酯(0.11g,0.29mmol)的DMF溶液(2mL)中。将产生的混合物在50℃下搅拌4小时。减压下除去溶剂。将残余物再次溶解于NaH 2PO 4(1.5M,10mL)中,然后加入Bn 4NHSO 4(200mg)并搅拌20分钟。反应混合物用乙酸乙酯萃取, 蒸发有机层得到(1R,4S)-4-(5-((甲基氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸化四丁基铵(0.100g,57.4%)。m/z(ES+),[M+H]+=459;ACID,HPLC tR=1.038min.
步骤5:化合物FF的合成
在0℃下向(1R,4S)-4-(5-((甲基氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸四丁基铵(0.1g,0.17mmol)的DCM溶液(2mL)加入TFA(0.5ml,6.49mmol)。将产生的混合物在0℃下搅拌8小时。将溶剂在减压下除去。将残余物用乙醚稀释。通过离心(3次)收集沉淀物。通过制备型HPLC纯化固体:柱:Xselect CSH OBD柱30*150mm 5um n;流动相A:水(0.1%FA),流动相B:ACN;流速:60mL/min;梯度:在7分钟内由2%B至15%B;254;220nm;Rt:5.93min.将含有期望化合物的级分蒸发至干得到(1R,4S)-4-(5-((甲基氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环己烷]-7-基硫酸氢酯(10.00mg,16.74%)。m/z(ES+),[M+H]+=359;ACID,HPLC tR=0.682min. 1H NMR(400MHz,D 2O)δ0.33(d,J=9.7Hz,1H),0.41(dd,J=9.0,5.1Hz,1H),0.50-0.58(m,1H),0.64(dd,J=9.6,5.0Hz,1H),1.66(d,J=16.0Hz,1H),2.46(dd,J=16.2,7.4Hz,1H),2.69(s,3H),3.07(d,J=12.0Hz,1H),3.14-3.25(m,1H),3.37(d,J=3.7Hz,1H),4.39(s,2H),4.74(d,J=7.7Hz,1H),6.68(s,1H).
实施例33化合物GG的制备
Figure PCTCN2019073000-appb-000080
Figure PCTCN2019073000-appb-000081
步骤1:化合物1的合成
将(E)-(((叔丁氧基羰基)亚胺基)(1H-吡唑-1-基)甲基)氨基甲酸叔丁酯(1.976g,6.37mmol)加入N-甲基丙-2-炔-1-胺(0.4g,5.79mmol)和TEA(2.420mL,17.36mmol)的MeOH溶液(10mL)中。将生成的混合物在0℃下搅拌16小时。减压下去除溶剂。通过快速硅胶色谱纯化粗产物,1至10%的EtOAc的石油醚溶液梯度洗脱。将纯的级分蒸发至干得到产物1(0.500g,27.7%),其为白色固体。m/z(ES+),[M+H] +=312;ACID,HPLC tR=0.954min.
步骤2-5:按照化合物FF的合成中的步骤3-6
化合物GG:
m/z(ES+),[M+H]+=401;ACID,HPLC tR=1.114min.1H NMR(400MHz,D 2O)δ0.32(s,1H),0.43(s,1H),0.54(s,1H),0.61-0.71(m,0H),1.65(d,J=15.6Hz,0H),2.46(dd,J=15.9,7.7Hz,0H),3.03(s,1H),3.08(d,J=12.0Hz,1H),3.19(d,J=11.8Hz,0H),3.37(d,J=3.6Hz,0H),4.69(s,2H),4.73(s,1H),6.51(s,0H).
实施例34化合物HH的制备
Figure PCTCN2019073000-appb-000082
步骤1-5:按照化合物GG的合成中的步骤1-5
m/z(ES+),[M+H]+=298;ACID,HPLC RT=1.274min. 1H NMR(400MHz,氯仿-d)δ1.52(d,J=1.0Hz,18H),2.29(t,J=2.6Hz,1H),4.26(dd,J=4.9,2.6Hz,2H),8.49(s,1H),11.47(s,1H).
化合物HH:
m/z(ES+),[M+H]+=387;ACID,HPLC tR=0.724min.1H NMR(400MHz,氯仿-d)δ1.52(d,J=1.0Hz,18H),2.29(t,J=2.6Hz,1H),4.26(dd,J=4.9,2.6Hz,2H),8.49(s,1H),11.47(s,1H).
实施例35化合物II的制备
Figure PCTCN2019073000-appb-000083
Figure PCTCN2019073000-appb-000084
步骤1-4:按照化合物FF的合成中的步骤3-6
化合物II:
m/z(ES+),[M+H]+=345;ACID,HPLC tR=0.646min. 1H NMR(400MHz,D 2O)δ0.31(s,1H),0.43(s,1H),0.55(d,J=5.7Hz,1H),0.60-0.71(m,1H),1.65(d,J=16.0Hz,1H),2.45(dd,J=15.8,7.2Hz,1H),3.08(d,J=11.9Hz,1H),3.18(d,J=11.5Hz,1H),3.36(d,J=3.6Hz,1H),4.32(s,2H),4.74(s,1H),6.60(s,1H),8.33(s,0H).
实施例36化合物JJ的制备
Figure PCTCN2019073000-appb-000085
Figure PCTCN2019073000-appb-000086
步骤1:按照化合物GG的合成中的步骤1
m/z(ES+),[M+H]+=312;ACID,HPLC tR=1.178min. 1H NMR(400MHz,氯仿-d)δ1.52(d,J=1.7Hz,18H),2.07(t,J=2.6Hz,1H),2.49(td,J=6.6,2.6Hz,2H),3.63(q,J=6.5Hz,2H),8.64(s,1H),11.51(s,1H).
步骤2-5:按照化合物FF的合成中的步骤3-6
化合物JJ:
m/z(ES+),[M+H]+=401;ACID,HPLC tR=0.808min.1H NMR(400MHz,D 2O)δ0.32(dt,J=9.9,5.0Hz,1H),0.43(dt,J=9.1,5.3Hz,1H),0.55(dt,J=10.3,5.1Hz,1H),0.66(dt,J=10.0,5.2Hz,1H),1.63(d,J=15.8Hz,1H),2.46(dd,J=15.9,7.4Hz,1H),3.05(dd,J=13.4,7.0Hz,3H),3.19(dd,J=12.0,3.6Hz,1H),3.37(d,J=3.7Hz,1H),3.51(t,J=6.4Hz,2H),4.73(s,1H),6.34(s,1H).
实施例37化合物KK的制备
Figure PCTCN2019073000-appb-000087
步骤1-5:按照化合物GG的合成中的步骤1-5
化合物KK:
m/z(ES+),[M+H]+=415;ACID,HPLC tR=1.283min.1H NMR(400MHz,D 2O)δ0.32(dt,J=9.9,5.0Hz,1H),0.43(dt,J=9.1,5.3Hz,1H),0.55(dt,J=10.3,5.1Hz,1H),0.66(dt,J=10.0,5.2Hz,1H),1.63(d,J=15.8Hz,1H),2.46(dd,J=15.9,7.4Hz,1H),3.05(dd,J=13.4,7.0Hz,3H),3.19(dd,J=12.0,3.6Hz,1H),3.37(d,J=3.7Hz,1H),3.51(t,J=6.4Hz,2H),4.73(s,1H),6.34(s,1H).
实施例38化合物LL的制备
Figure PCTCN2019073000-appb-000088
步骤1:按照化合物UU的合成中的步骤1
步骤2:按照化合物GG的合成中的步骤1
m/z(ES+),[M+H]+=400;ACID,HPLC tR=1.276min. 1H NMR(400MHz,氯仿-d)δ0.20(s,9H),1.48(s,10H),3.20-3.36(m,1H),3.49(s,1H),4.46(dd,J=7.0,3.8Hz,1H),4.98(s,1H).
步骤3:化合物3的合成
将TBS-Cl(0.415g,2.75mmol)添加至化合物2(1g,2.50mmol)和TEA(0.698mL,5.01mmol)的DCM溶液(20mL)中。将生成的混合物在室温下搅拌4小时。减压下除去溶剂。通过快速硅胶色谱纯化粗产物,1至10%的EtOAc的石油醚溶液梯度洗脱。将纯的级分蒸发至 干得到粘稠状化合物3(1.100g,86%)。m/z(ES+),[M+H]+=514;ACID,HPLC tR=1.532min.
步骤4:化合物4的合成
将K 2CO 3(0.403g,2.92mmol)添加至化合物3(0.75g,1.46mmol)的MeOH溶液(20mL)中。将生成的混合物在室温下搅拌4小时。在减压下除去溶剂。将粗产物通过快速硅胶色谱纯化,0至10%EtOAc的石油醚溶液梯度洗脱。将纯的级分蒸发至干得到粘稠状化合物4(0.6g)。 1H NMR(300MHz,氯仿-d)δ0.13(dd,J=9.9,6.9Hz,6H),0.91(s,9H),1.49(d,J=5.2Hz,16H),2.39-2.49(m,1H),4.50(ddd,J=7.9,4.2,2.0Hz,1H).
步骤5-7:按照化合物FF的合成中的步骤3-5
步骤8:化合物8的合成
将TEA·HF(0.068g,0.56mmol)添加至(1R,4S)-4-(5-((E)-8-((叔丁氧基羰基)氨基)-2,2,3,3,12,12-六甲基-10-氧代-4,11-二氧杂-7,9-二氮杂-3-硅杂十三-8-烯-5-基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸氢酯(0.102g,0.14mmol)的THF溶液(2.000mL)中。将生成的混合物室温搅拌14小时。在减压下除去溶剂得到(1R,4S)-4-(5-(2-((E)-2,3-双(叔丁氧基羰基)胍基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸氢酯(0.080g,93%),其为粘稠物。m/z(ES+),[M+H]+=617;ACID,HPLC tR=1.044min.
步骤9:按照化合物LL的合成中的步骤6
m/z(ES+),[M+H]+=417;ACID,HPLC tR=0.716min. 1H NMR(400MHz,D 2O)δ0.30(s,1H),0.41(dt,J=9.1,5.3Hz,1H),0.49-0.58(m,1H),0.63(dd,J=10.0,4.9Hz,1H),1.63(d,J=15.9Hz,1H),2.45(dd,J=15.3,7.3Hz,1H),3.04(dd,J=12.0,5.3Hz,1H),3.18(d,J= 11.8Hz,1H),3.35(d,J=3.7Hz,1H),3.57(d,J=5.1Hz,1H),5.04(t,J=5.2Hz,1H),6.48(s,1H).
实施例39化合物MM的制备
Figure PCTCN2019073000-appb-000089
步骤1-4:按照化合物I的合成中的步骤6-9
化合物MM:
ESI-MS(EI +,m/z+):417,0.721min. 1H NMR(300MHz,D 2O)δ0.33(dd,J=9.6,4.5Hz,1H),0.43(dt,J=8.6,5.1Hz,1H),0.56(dt,J=10.2,5.0Hz,1H),0.66(dt,J=9.8,5.0Hz,1H),1.65(d,J=15.7Hz,1H),2.46(dd,J=16.1,7.9Hz,1H),3.05(d,J=12.0Hz,1H),3.20(dd,J=11.9,3.6Hz,1H),3.37(d,J=3.7Hz,1H),3.59(d,J=5.2Hz,2H),4.74(d,J=7.4Hz,1H),5.06(t,J=5.1Hz,1H),6.50(s,1H).
实施例40化合物NN的制备
Figure PCTCN2019073000-appb-000090
步骤1-4:按照化合物I的合成中的步骤6-9
ESI-MS(EI +,m/z+):417,0.721min. 1H NMR(300MHz,D 2O)δ0.33(dd,J=9.6,4.6Hz,1H),0.43(dt,J=8.6,5.1Hz,1H),0.56(dt,J=10.0,5.0Hz,1H),0.65(dd,J=9.6,4.4Hz,1H),1.65(d,J=15.8Hz,1H),2.47(dd,J=16.2,7.3Hz,1H),3.06(d,J=12.0Hz,1H),3.19(dd,J=12.0,3.6Hz,1H),3.37(d,J=3.6Hz,1H),3.59(dd,J=5.3,1.7Hz,2H),4.75(s,1H),5.06(t,J=5.2Hz,1H),6.50(s,1H).
实施例41化合物OO的制备
Figure PCTCN2019073000-appb-000091
步骤1-5:按照化合物FF的合成中的步骤1-5
m/z(ES+),[M+H]+=359;ACID,HPLC tR=0.710min. 1H NMR(400MHz,D 2O)δ0.32(dt,J=9.8,4.9Hz,1H),0.38-0.49(m,1H),0.55(dt,J=10.5,5.2Hz,1H),0.64(dt,J=9.8,5.2Hz,1H),1.63(d,J=15.8Hz,1H),2.44(dd,J=15.8,7.6Hz,1H),3.06-3.23(m,4H),3.24-3.41(m,3H),4.71(s,1H),6.39(s,1H).
实施例42化合物PP的制备
Figure PCTCN2019073000-appb-000092
步骤1:化合物1的合成
将碘苯双(2,2,2-三氟乙酸酯)(0.428g,1.00mmol)分批添加至(E)-7-(苄氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-甲醛肟(0.3g,1.00mmol)和丁-3-炔-1-基(甲基)氨基甲酸叔丁酯(0.182g,1.00mmol)的MeOH溶液(8mL)和水(2mL)中。将生成的混合物在室 温下搅拌4小时。在减压下除去溶剂。将粗产物通过快速C18快速色谱纯化,0至65%MeCN的水溶液梯度洗脱。将纯的级分蒸发至干得到(2-(3-((1R,4S)-7-(苄基氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)异噁唑-5-基)乙基)(甲基)氨基甲酸叔丁酯(0.180g,37.5%),其为白色固体。m/z(ES+),[M+H]+=483;ACID,HPLC tR=1.311min.
步骤2-4:按照化合物FF的合成中的步骤4-6
m/z(ES+),[M+H]+=373;ACID,HPLC tR=1.017min. 1H NMR(400MHz,D 2O)δ0.30(dt,J=9.8,5.1Hz,1H),0.40(dt,J=9.0,5.3Hz,1H),0.53(dt,J=10.2,5.1Hz,1H),0.62(dt,J=10.0,5.2Hz,1H),1.61(d,J=15.8Hz,1H),2.42(dd,J=15.8,7.6Hz,1H),2.65(s,3H),3.04-3.24(m,4H),3.33(t,J=6.7Hz,3H),4.68(s,1H),6.38(s,1H).
实施例43化合物QQ的制备
Figure PCTCN2019073000-appb-000093
步骤1-4:按照化合物FF的合成中的步骤2-5
m/z(ES+),[M+H]+=399;ACID,HPLC tR=0.792min. 1H NMR(400MHz,D 2O)δ0.33(dt,J=9.8,4.9Hz,1H),0.44(dt,J=9.1,5.3Hz,1H),0.57(dt,J=10.4,5.1Hz,1H),0.67(dt,J=9.9,5.2Hz,1H),1.65(d,J=15.8Hz,1H),1.81-2.00(m,2H),2.26(dd,J=14.8,3.4Hz,2H),2.46(dd,J=15.8,7.7Hz,1H),3.03-3.30(m,5H),3.35-3.51(m,3H),4.73(s,1H),6.35(s,1H).
实施例44化合物RR的制备
Figure PCTCN2019073000-appb-000094
步骤1-4:按照化合物FF的合成中的步骤2-5
m/z(ES+),[M+H]+=399;ACID,HPLC tR=0.806min. 1H NMR(400MHz,D 2O)δ0.31(dd,J=9.5,4.9Hz,1H),0.41(dt,J=9.1,5.4Hz,1H),0.54(dt,J=10.3,5.0Hz,1H),0.64(dt,J=9.8,5.2Hz,1H),1.63(d,J=15.8Hz,1H),1.68-1.83(m,2H),1.94(s,1H),2.16(s,1H),2.43(dd,J=15.9,7.5Hz,1H),2.91-3.05(m,1H),3.05-3.22(m,3H),3.26- 3.42(m,3H),3.59(d,J=12.5Hz,1H),6.37(s,1H).
实施例45化合物SS的制备
Figure PCTCN2019073000-appb-000095
步骤1-4:按照化合物FF的合成中的步骤2-5
m/z(ES+),[M+H]+=385;ACID,HPLC tR=0.764min. 1H NMR(400MHz,D 2O)δ0.31(dt,J=10.0,5.1Hz,1H),0.41(dt,J=9.0,5.3Hz,1H),0.54(dt,J=10.3,5.1Hz,1H),0.64(dt,J=9.8,5.2Hz,1H),1.63(d,J=15.8Hz,1H),2.11-2.26(m,1H),2.43(dd,J=13.7,7.3Hz,2H),3.05-3.20(m,2H),3.29-3.52(m,4H),3.68(dd,J=12.1,8.2Hz,1H),3.83(p,J=7.6Hz,1H),6.41(s,1H).
实施例46化合物TT的制备
Figure PCTCN2019073000-appb-000096
步骤1:化合物1的合成
将K 2CO 3(0.347g,2.51mmol)添加至4-氧代哌啶-1-甲酸叔丁酯(1g,5.02mmol)和丁-3-炔-1-胺盐酸盐(0.530g,5.02mmol)的MeOH溶液(30mL)中。将生成的混合物在室温下搅拌4小时。加入NaBH(OAc) 3(3.19g,15.06mmol)。将生成的混合物在室温下搅拌14小时。减压下除去溶剂得到4-(丁-3-炔-1-基氨基)哌啶-1-甲酸叔丁酯(0.800g,63.2%)。产物不经过进一步纯化直接用于下一步。m/z(ES+),[M+H]+=253;ACID,HPLC tR=0.812min.
步骤2:化合物2的合成
将BOC-酸酐(0.736mL,3.17mmol)添加至4-(丁-3-炔-1-基氨基)哌啶-1-甲酸叔丁酯(0.8g,3.17mmol)和TEA(0.442mL,3.17mmol)的DCM溶液(20mL)中。将生成的混合物在室温下搅拌6小时。用DCM(50mL)稀释反应混合物,并依次用水和饱和食盐水洗涤。将有机层用Na2SO4干燥,过滤并蒸发得到粗产物。将粗产物通过快速硅 胶色谱纯化,0至20%EtOAc的石油醚溶液梯度洗脱。将纯的级分蒸发至干得到4-(丁-3-炔-1-基(叔丁氧羰基)氨基)哌啶-1-甲酸叔丁酯(0.600g,53.7%),其为白色固体。m/z(ES+),[M+H]+=353;ACID,HPLC tR=1.383min. 1H NMR(300MHz,氯仿-d)δ1.49(d,J=3.1Hz,18H),1.64(d,J=24.2Hz,5H),2.41(t,J=6.0Hz,2H),2.74(t,J=12.5Hz,2H),3.27(s,2H),4.21(d,J=12.8Hz,2H).
步骤3-6:按照化合物FF的合成中的步骤2-5
m/z(ES+),[M+H]+=442;ACID,HPLC tR=0.672min. 1H NMR(400MHz,D 2O)δ0.36(dt,J=10.2,5.2Hz,1H),0.41-0.54(m,1H),0.59(dt,J=10.4,5.2Hz,1H),0.69(dt,J=9.8,5.2Hz,1H),1.67(d,J=15.8Hz,1H),1.83(qd,J=13.1,3.6Hz,2H),2.35(d,J=14.0Hz,2H),2.49(dd,J=16.2,7.4Hz,1H),3.06(td,J=13.4,2.8Hz,2H),3.16(t,J=12.2Hz,1H),3.20-3.30(m,2H),3.41(d,J=3.5Hz,1H),3.49(q,J=7.9,7.2Hz,2H),3.52-3.63(m,2H),4.74(d,J=7.3Hz,2H),6.44(s,1H).
实施例47化合物UU的制备
Figure PCTCN2019073000-appb-000097
Figure PCTCN2019073000-appb-000098
步骤1:化合物1的合成
将TFA(1.933mL,25.09mmol)添加至4-氧代哌啶-1-甲酸叔丁酯(5g,25.09mmol)的DCM溶液(50mL)中。将生成的混合物在室温下搅拌4小时。在减压下除去溶剂得到哌啶-4-酮盐酸盐(0.800g,23.51%),其为无色粘稠物。
步骤2:按照化合物GG的合成中的步骤2
步骤3-4:按照化合物TT的合成中的步骤1-2
1H NMR(400MHz,氯仿-d)δ1.49(d,J=7.8Hz,29H),1.75(s,3H),2.01(d,J=6.0Hz,1H),2.06(s,2H),2.40(s,2H),3.01(d,J=38.2Hz,2H),3.24(s,4H),4.21(s,2H).
步骤5-8:按照化合物FF的合成中的步骤2-5
化合物UU:
m/z(ES+),[M+H]+=484;ACID,HPLC tR=0.948min. 1H NMR(400MHz,D 2O)δ0.33(dd,J=9.6,4.8Hz,1H),0.43(dt,J=8.9,5.3Hz, 1H),0.56(dt,J=10.4,5.1Hz,1H),0.66(dt,J=9.7,5.2Hz,1H),1.54-1.72(m,3H),2.15(d,J=12.4Hz,2H),2.46(dd,J=15.8,7.5Hz,1H),2.98-3.29(m,6H),3.36-3.53(m,4H),3.89(d,J=14.3Hz,2H),6.40(s,1H),8.35(s,1H).
实施例48化合物VV的制备
Figure PCTCN2019073000-appb-000099
步骤1:按照化合物TT的合成中的步骤2
步骤2:
将NaH(1.469g,36.72mmol)添加至3-((叔丁氧羰基)氨基)氮杂换丁烷-1-甲酸叔丁酯(5g,18.36mmol)的DMF溶液(50mL)中。将生成的混合物在室温下搅拌1小时。加入3-溴丙-1-炔(2.402g,20.19mmol)。将生成的混合物在室温下搅拌3小时。用水(100mL)将反应混合物淬灭,用EtOAc萃取,有机层用Na2SO4干燥,过滤并蒸发以得到白色固体。将粗产物通过快速硅胶色谱纯化,0至20%EtOAc的石油醚溶液梯度洗脱。将纯的级分蒸发至干得到3-((叔丁氧基羰基)(丙 -2-炔-1-基)氨基)氮杂换丁烷-1-甲酸叔丁酯(0.800g,14.04%),其为白色固体。m/z(ES+),[M+H]+=311;ACID,HPLC tR=1.265min. 1H NMR(400MHz,氯仿-d)δ1.46(s,9H),1.50(s,8H),2.23(t,J=2.4Hz,1H),4.05-4.18(m,6H),4.42-4.81(m,1H).
步骤3-6:按照化合物PP的合成中的步骤1-4
化合物VV:
m/z(ES+),[M+H]+=400;ACID,HPLC tR=0.729min. 1H NMR(400MHz,D 2O)δ0.31(dt,J=10.1,5.1Hz,1H),0.41(dt,J=9.1,5.3Hz,1H),0.53(dt,J=10.3,5.1Hz,1H),0.63(dt,J=9.8,5.2Hz,1H),1.61(d,J=15.8Hz,1H),2.44(dd,J=15.7,7.7Hz,1H),3.05(d,J=12.0Hz,1H),3.16(dd,J=12.0,3.7Hz,1H),3.35(d,J=3.6Hz,1H),3.79(ddd,J=10.8,7.3,3.1Hz,2H),3.85-3.95(m,3H),4.06(dd,J=11.2,7.6Hz,2H),6.40(s,1H).
实施例49化合物WW的制备
Figure PCTCN2019073000-appb-000100
步骤1:化合物1的合成
在0℃下将LAH(0.828g,21.81mmol)分批添加至3-(2-甲氧基-2-氧代乙基)氮杂换丁烷-1-甲酸叔丁酯(5g,21.81mmol)的THF溶液(100mL)中。将反应混合物在室温下搅拌2小时。反应混合物用水(50mL)淬灭,用EtOAc萃取,有机层用Na2SO4干燥,过滤并蒸发以提供无色粘稠物。将粗产物通过快速硅胶色谱纯化,0至50%EtOAc的石油醚溶液梯度洗脱。将纯的级分蒸发至干得到3-(2-羟基乙基)氮杂换丁烷-1-甲酸叔丁酯(3.50g,80%),其为白色固体。m/z(ES+),[M+H-tBu]+=146;ACID,HPLC tR=1.079min.
步骤2:化合物2的合成
在0℃下将TEMPO(0.272g,1.74mmol)添加至3-(2-羟基乙基)氮杂换丁烷-1-甲酸叔丁酯(3.5g,17.39mmol)和1,3,5-三嗪烷-2,4,6-三酮(2.469g,19.13mmol)的DCM溶液(80mL)中。将生成的混合物在室温下搅拌2小时。使反应混合物通过硅藻土过滤。在减压下除去溶剂得到3-(2-氧代乙基)氮杂换丁烷-1-甲酸叔丁酯(3.40g,98%),其为浅黄色粘稠物。 1H NMR(400MHz,氯仿-d)δ1.44(s,9H),2.84(d,J=7.4Hz,2H),2.90-3.00(m,1H),3.58(dd,J=8.8,5.4Hz,2H),4.14(t,J=8.5Hz,2H),9.78(s,1H).
步骤3:化合物3的合成
在0℃下将K 2CO 3(4.16g,30.11mmol)添加至3-(2-氧代乙基)氮杂环丁烷-1-甲酸叔丁酯(3g,15.06mmol)的MeOH溶液(3mL)中。将生成的混合物在0℃下搅拌10分钟。将(1-重氮-2-氧代丙基)膦酸二甲酯(2.89g,15.06mmol)的MeOH溶液(3mL)滴加至上述混合物中。将生成的混合物在室温下搅拌16小时。将反应混合物倾入水(50mL)中,用EtOAc萃取,有机层用Na2SO4干燥,过滤并蒸发以提供粘稠物。将粗产物通过快速硅胶色谱纯化,0至15%EtOAc的石油醚溶液梯度 洗脱。将纯的级分蒸发至干得到3-(丙-2-炔-1-基)氮杂环丁烷-1-甲酸叔丁酯(1.900g,64.6%),其为白色固体。
步骤4-7:按照化合物PP的合成中的步骤1-4
化合物WW:
m/z(ES+),[M+H]+=385;ACID,HPLC tR=0.670min. 1H NMR(400MHz,D 2O)δ0.30(dt,J=9.8,5.0Hz,1H),0.40(dt,J=8.9,5.3Hz,1H),0.53(dt,J=10.4,5.1Hz,1H),0.63(dt,J=9.9,5.2Hz,1H),1.60(d,J=15.8Hz,1H),2.42(dd,J=15.9,7.4Hz,1H),3.03-3.19(m,4H),3.22-3.42(m,2H),3.90(dd,J=10.1,7.9Hz,2H),4.05-4.20(m,2H),4.68(d,J=7.7Hz,1H),6.26(s,1H).
实施例50化合物XX的制备
Figure PCTCN2019073000-appb-000101
步骤1:化合物1的合成
将NaBH4(0.442g,11.68mmol)添加至3-氧代氮杂环丁烷-1-甲酸叔丁酯(2g,11.68mmol)的THF溶液(40mL)中。将生成的混合物在室温下搅拌6小时。用EtOAc(50mL)稀释反应混合物,用依次用水和 饱和食盐水洗涤。有机层用Na2SO4干燥,过滤并蒸发以提供粗产物。将粗产物通过快速硅胶色谱纯化,0至50%EtOAc的石油醚溶液梯度洗脱。将纯的级分蒸发至干得到3-羟基氮杂环丁烷-1-甲酸叔丁酯(1.700g,84%),其为白色固体。m/z(ES+),[2M+H]+=347;ACID,HPLC tR=0.878min.
步骤2-6:按照化合物VV的合成中的步骤2-6
化合物2:
m/z(ES+),[2M+H]+=423;ACID,HPLC tR=1.118min. 1H NMR(400MHz,氯仿-d)δ1.45(s,9H),2.46(t,J=2.4Hz,1H),3.91(dd,J=10.3,4.4Hz,2H),4.04-4.24(m,4H),4.43(tt,J=6.5,4.4Hz,1H).
化合物XX:
m/z(ES+),[M+H]+=401;ACID,HPLC tR=0.772min. 1H NMR(400MHz,D 2O)δ0.31(dd,J=9.6,4.9Hz,1H),0.41(dt,J=9.1,5.3Hz,1H),0.54(dt,J=10.2,5.2Hz,1H),0.64(dt,J=9.8,5.1Hz,1H),1.63(d,J=15.8Hz,1H),2.44(dd,J=15.6,7.4Hz,1H),3.06(d,J=12.0Hz,1H),3.17(dd,J=12.0,3.6Hz,1H),3.35(d,J=3.6Hz,1H),3.94(dd,J=12.6,5.0Hz,2H),4.22(dd,J=12.5,6.6Hz,2H),4.56(p,J=5.9Hz,1H),4.65(s,2H),4.73(s,1H),6.53(s,1H).
实施例51化合物YY的制备
Figure PCTCN2019073000-appb-000102
Figure PCTCN2019073000-appb-000103
步骤1:按照化合物GG的合成中的步骤1
步骤2-3:按照化合物TT的合成中的步骤1-2
步骤4-7:按照化合物PP的合成中的步骤1-4
化合物YY:
m/z(ES+),[M+H]+=442;ACID,HPLC tR=0.769min. 1H NMR(400MHz,D 2O)δ0.32(dt,J=10.1,5.1Hz,1H),0.41(dt,J=9.1,5.3Hz,1H),0.55(dt,J=10.1,5.0Hz,1H),0.64(dt,J=9.9,5.2Hz,1H),1.63(d,J=15.8Hz,1H),2.44(dd,J=15.8,7.6Hz,1H),3.08(d,J=12.0Hz,1H),3.17(dd,J=12.0,3.6Hz,1H),3.36(d,J=3.6Hz,1H),3.83(ddq,J=13.6,9.3,4.8Hz,3H),3.97(s,2H),4.20-4.31(m,2H),4.72(s,1H),6.46(s,1H).
实施例52化合物ZZ的制备
Figure PCTCN2019073000-appb-000104
Figure PCTCN2019073000-appb-000105
步骤1:
将BH 3.THF(62.7mL,62.72mmol)滴加至3-((叔丁氧基羰基)氨基)换丁烷-1-羧酸(9g,41.81mmol)的THF溶液(40mL)中。将生成的混合物在室温下搅拌4小时。将反应混合物用水(50mL)淬灭,用EtOAc萃取,有机层用Na 2SO 4干燥,过滤并蒸发得到粘稠物。将粗产物通过快速硅胶色谱纯化,0至5%MeOH的DCM溶液梯度洗脱。将纯的级分蒸发至干得到(3-(羟基甲基)环丁基)氨基甲酸叔丁酯(7.10g,84%),其为粘稠物。 1H NMR(400MHz,氯仿-d)δ1.39(s,9H),1.53-1.68(m,1H),1.89-2.02(m,1H),2.12(dd,J=14.1,6.8Hz,2H),2.36(d,J=7.7Hz,1H),3.04(s,1H),3.50(d,J=5.6Hz,1H),3.60(d,J=7.2Hz,1H),3.86-4.06(m,1H),4.06-4.22(m,0H),5.00(s,1H).
步骤2-7:按照化合物WW的合成中的步骤2-7
化合物2:
1H NMR(400MHz,氯仿-d)δ1.36-1.55(m,9H),2.02-2.15(m, 2H),2.18(d,J=9.5Hz,1H),2.56(q,J=9.1,8.3Hz,1H),2.64-2.78(m,1H),2.92(p,J=7.8Hz,1H),3.05(s,0H),4.04-4.32(m,1H),4.73(s,1H),9.71(d,J=2.1Hz,1H),9.85(d,J=1.9Hz,0H).
化合物3:
1H NMR(400MHz,氯仿-d)δ1.45(d,J=2.5Hz,9H),1.97(qd,J=9.4,2.4Hz,1H),2.11-2.28(m,2H),2.43-2.51(m,1H),2.57-2.78(m,2H),2.94(d,J=3.7Hz,0H),4.04(d,J=7.5Hz,0H),4.43(s,0H),4.75(s,1H).
化合物ZZ:
异构体1:m/z(ES+),[M+H]+=385;ACID,HPLC tR=0.665min.异构体2:m/z(ES+),[M+H]+=385;ACID,HPLC tR=0.692min. 1H NMR(400MHz,D 2O)δ0.23-0.36(m,1H),0.41(dt,J=9.9,5.3Hz,1H),0.49-0.59(m,1H),0.63(dt,J=9.9,5.2Hz,1H),1.61(dd,J=15.8,4.2Hz,1H),2.28-2.50(m,2H),2.60(t,J=8.2Hz,2H),2.66-2.79(m,1H),3.04-3.22(m,2H),3.35(s,1H),3.51(p,J=9.8Hz,1H),3.69-3.89(m,1H),4.01(p,J=7.2Hz,1H),6.32(d,J=17.8Hz,1H).
实施例53化合物AAA的制备
Figure PCTCN2019073000-appb-000106
Figure PCTCN2019073000-appb-000107
步骤1:
将MsCl(6.67mL,85.60mmol)滴加至丁-3-炔-1-醇(5g,71.34mmol)和TEA(19.89mL,142.67mmol)的THF溶液中(40mL)。将生成的混合物在室温下搅拌4小时。减压下除去溶剂得到丁-3-炔-1-基甲磺酸酯(7.00g,66.2%),其为白色粘稠物。 1H NMR(400MHz,氯仿-d)δ2.09(t,J=2.7Hz,1H),2.67(td,J=6.7,2.7Hz,2H),3.07(s,3H),3.69(s,1H),4.31(t,J=6.7Hz,2H).
步骤2:
将2-氨基乙烷-1-醇(2.309g,37.79mmol)添加至丁-3-炔-1-基甲磺酸酯(2.8g,18.90mmol)和TEA(5.27mL,37.79mmol)的THF溶液(40mL)中。将生成的混合物在60℃搅拌14小时。减压下除去溶剂得到无色粘稠物。将粗产物通过快速硅胶色谱纯化,90%EtOAc的MeOH溶液洗脱。将纯的级分蒸发至干得到2-(丁-3-炔-1-基氨基)乙烷-1-醇(1.400g,65.5%),其为粘稠物。m/z(ES+),[M+H] +=114;ACID,HPLC tR=0.357min.
步骤3:按照化合物TT的合成中的步骤2
步骤4
将DEAD(9.46mL,59.78mmol)滴加至丁-3-炔-1-基(2-羟基乙基)氨基甲酸叔丁酯(8.5g,39.85mmol),异吲哚啉-1,3-二酮(6.45g,43.84mmol)和Ph 3P(15.68g,59.78mmol)的THF溶液(120mL)中。将生成的混合物在60℃搅拌14小时。在减压下除去溶剂得到无色粘稠物。将粗产物通过快速硅胶色谱纯化,0至15%EtOAc的石油醚溶液梯度洗脱。将纯的级分蒸发至干得到丁-3-炔-1-基(2-(1,3-二氧代异吲哚啉-2-基)乙基)氨基甲酸叔丁酯(10.50g,77%),其为白色粘稠物。 1H NMR(400MHz,氯仿-d)δ1.26(d,J=4.6Hz,9H),1.96(s,1H),2.44(ddq,J=18.8,6.8,4.7,3.5Hz,2H),3.38(dt,J=21.2,6.9Hz,2H),3.57(t,J=5.9Hz,2H),3.85(q,J=6.1,5.6Hz,2H),4.98(tp,J=12.5,6.3Hz,2H),7.64-7.80(m,3H),7.84(ddt,J=10.8,5.4,3.1Hz,2H).
步骤5
将肼(3.74g,116.82mmol)添加至丁-3-炔-1-基(2-(1,3-二氧代异吲哚啉-2-基)乙基)氨基甲酸叔丁酯(4g,11.68mmol)的THF溶液(50mL)中。将生成的混合物在室温下搅拌14小时。在减压下除去溶剂得到(2-氨基乙基)(丁-3-炔-1-基)氨基甲酸叔丁酯(2.100g,85%)。m/z(ES+),[M+H]+=213;ACID,HPLC tR=0.812min.
步骤6按照化合物GG的合成中的步骤1
1H NMR(400MHz,DMSO-d 6)δ1.33-1.43(m,18H),1.47(s,9H),2.39(td,J=7.3,2.5Hz,2H),2.81(s,1H),3.29(t,J=7.2Hz,2H),3.34-3.46(m,4H),8.38(s,1H),11.51(d,J=31.8Hz,1H).
步骤7-10:按照化合物PP的合成中的步骤1-4
化合物AAA:
m/z(ES+),[M+H] +=444;ACID,HPLC tR=0.898min. 1H NMR(300MHz,D 2O)δ0.34(dt,J=9.6,4.7Hz,1H),0.44(dt,J=8.6,5.1Hz,1H),0.58(dt,J=10.1,4.9Hz,1H),0.68(dt,J=9.7,4.9Hz,1H),1.66(d,J=15.8Hz,1H),2.47(dd,J=15.8,7.5Hz,1H),3.14-3.29(m,6H),3.38(t,J=4.9Hz,3H),3.51(t,J=6.0Hz,2H),4.74(s,1H),6.42(s,1H),8.37(s,1H).
实施例54化合物BBB的合成
Figure PCTCN2019073000-appb-000108
步骤1:化合物2的合成
向50-mL密封管中加入化合物1(300mg,1mmol,1.00当量)和乙炔基三甲基硅烷(150mg,1.5mmol,1.5当量)的MeOH∶H 2O=5∶1(20mL)溶液,在0℃下搅拌,分两批加入PIFA(645mg,1.5mmol,1.5当 量)。将反应混合物在室温下搅拌2小时。将粗残余物在Prep-TLC(PE∶EA=2∶1)上纯化得到期望的产物(160mg,40.5%),其为无色油状物。ESI-MS(EI +,m/z):398,1.115min.
步骤2:化合物3的合成
向50-mL密封管中加入化合物2(160mg,1mmol,1.00当量)和CsF(181mg,1.2mmol,3当量)的MeOH溶液(10mL),室温搅拌过夜。将反应混合物过滤并浓缩滤液。将粗产物在Prep-TLC上纯化得到期望的产物(100mg,76.9%),其为无色油状物。ESI-MS(EI +,m/z):326,0.954min.
步骤3:化合物4的合成
向50-mL圆底烧瓶中加入化合物3(100mg,0.3mmol,1.00当量)的THF溶液(15mL)中,然后加入Pd/C(10mg,催化基)。将产生的溶液在H2(1Atm)下在室温下搅拌120分钟。将固体过滤掉。将产生的混合物在真空下浓缩。得到100mg(粗)的化合物4,其为无色油状物。ESI-MS(EI +,m/z):236,0.855min.
步骤4:化合物BBB的合成
向50-mL密封管中加入化合物4(100mg,0.3mmol,1.00当量)的DMF溶液(5mL),然后加入SO 3.Py(270mg,1.7mmol,4.00当量)。将产生的溶液在室温下搅拌20小时。用Prep-HPLC(A相:水(10mmol/L NH 4HCO 3)B相:ACN)直接纯化反应得到铵盐(10.9mg)。ESI-MS(EI +,m/z):316,0.95min. 1H NMR(400MHz,D 2O)δ0.37(dd,J=9.6,4.7Hz,1H),0.42-0.56(m,1H),0.61(dt,J=10.5,5.2Hz,1H),0.69(dt,J=9.6,5.2Hz,1H),1.71(d,J=15.9Hz,1H),2.52(dd,J=15.9,7.8Hz,2H),3.09-3.25(m,2H),3.41(d,J=3.6Hz,2H),4.80(d,J=7.6Hz,2H),6.58(d,J=1.7Hz,1H),8.63(d,J=1.7Hz,1H).
实施例55化合物CCC的制备
Figure PCTCN2019073000-appb-000109
步骤1:化合物1的合成
向500-mL圆底烧瓶中加入L-丝氨酸(5g,47.58mmol,1.00当量)的二噁烷溶液(200mL),然后加入NaOH(4g,100.01mmol,2.10当量)的水溶液(100mL)。随后,将得到的溶液在室温下搅拌5分钟。然后加入(Boc) 2O(12g,54.98mmol,1.14当量)。将得到的溶液在室温下继续搅拌16小时。用EA萃取得到的混合物(2x200mL)。用HCl(1mol/L)调节水层pH至约1。将得到的溶液用乙酸乙酯萃取(3x200mL)并且合并有机层,用无水硫酸钠干燥,真空下浓缩。得到10.2g(粗)的 2(S)-2-[[(叔丁氧基)羰基]氨基]-3-羟基丙酸,其为无色油状物。ESI-MS(EI +,m/z+Na):228,0.779min.
步骤2:化合物2的合成
向500-mL圆底烧瓶中加入2(S)-2-[[(叔丁氧基)羰基]氨基]-3-羟基丙酸(10.2g,49.71mmol,1.00当量)的THF溶液(200mL),TEA(10g,98.82mmol,2.00当量),DMAP(6g,49.11mmol,1.00当量),TBS-Cl(10.5g,69.66mmol,1.40当量)。将得到的溶液在室温下搅拌16小时。反应完全后,用HCl(1mol/L)调节pH至1。用乙酸乙酯萃取得到的溶液(2x300mL)并合并有机层,无水硫酸钠干燥,真空下浓缩。得到16.3g(粗)的2(S)-2-[[(叔丁氧基)羰基]氨基]-3-[(叔丁基二甲基硅基)氧基]丙酸,其为无色油状物。ESI-MS(EI +,m/z+Na):320,0.553min.
步骤3:化合物3的合成
向200-mL圆底烧瓶中加入2(S)-2-[[(叔丁氧基)羰基]氨基]-3-[(叔丁基二甲基硅基)氧基]丙酸(16.3g,51.02mmol,1.00当量)的DCM溶液(100mL).室温下分批加入CDI(12.4g,76.54mmol,1.50当量)。将得到的溶液在室温下搅拌120分钟,然后加入肼(11g,220.00mmol,10.00当量)。将得到的溶液在室温下继续搅拌60分钟。将得到的混合物在真空下浓缩。通过快速制备HPLC采用以下条件纯化粗物质:柱,C18硅胶;流动相,在30分钟内由ACN增加至ACN=50%;检测器,UV 254nm。得到11g(65%)的N-[(1S)-2-[(叔丁基二甲基硅基)氧基]-1-(肼基羰基)乙基]氨基甲酸叔丁酯,其为白色固体。ESI-MS(EI +,m/z+):334,1.168min.1H NMR(DMSO-d6,400MHz):δ(ppm)0.84(s,9H),1.38(s,9H),3.50-3.81(m,2H),3.96-4.13(m,1H),4.20(d,J=3.6Hz,2H),6.58(d,J=8.8Hz,1H),9.10(s,1H)
步骤4:化合物4的合成
向200-mL圆底烧瓶中加入化合物3(3.5g,10.49mmol,1.00当量),(1R,4S)-7-(苄氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1’-环丙 烷]-4-羧酸(4.7g,15.55mmol,1.2当量),DIPEA(4.5g,34.88mmol,3.00当量),HATU(8.74g,23.00mmol,4.00当量)的THF溶液(100mL)。将得到的溶液在室温下搅拌2小时。过滤掉固体。通过硅胶柱用乙酸乙酯/石油醚(1/5至1/1)纯化残余物。得到7g(100%,纯度95.6%)的化合物4,其作为白色固体。ESI-MS(EI +,m/z+):618,1.404min.
步骤5:化合物5的合成
向200-mL圆底烧瓶中加入化合物4(7g,11.33mmol,1.00当量)的DCM溶液(100mL),然后加入DIEA(5.67g,43.95mmol,3.50当量),Burgess试剂(10.42g,43.97mmol,3.50当量)。将得到的溶液在室温下搅拌20小时。通过硅胶柱采用乙酸乙酯/石油醚(1/5至1/1)纯化残余物,得到6g(88%)的化合物5,其为白色固体。ESI-MS(EI +,m/z+):600,1.487min.
步骤6:化合物6的合成
向100-mL圆底烧瓶中加入化合物5(6g,10mmol,1.00当量)的DCM溶液(40mL),然后加入TFA(10mL,10.00当量)。将得到的溶液在0℃下在室温下搅拌120分钟。将得到的混合物在真空下浓缩。得到6g的化合物6,其为白色固体。ESI-MS(EI +,m/z):500,1.538min.
步骤7:化合物7的合成
在100-mL圆底烧瓶中加入化合物6(6g,10mmol,1.00当量)的THF溶液(60mL),然后加入3HF Et3N(15mL,10.00当量)。将得到的溶液在室温下搅拌2小时。粗产品通过快速制备HPLC用以下条件纯化:柱,C18硅胶;流动相,在30分钟内由ACN增加至ACN=50%;检测器,UV 254nm。得到6g(粗)化合物7,其为白色固体。ESI-MS(EI +,m/z+):386,0.878min.
步骤8:化合物8的合成
向100-mL密封管中加入化合物7的ACN溶液(100mL),TEA(6.3g,42.32mmol,4.00当量),(1H-吡唑-1-基)甲烷二基亚基二氨基甲酸二叔 丁酯(5.28g,17mmol,1.10当量)。将得到的溶液在40℃下搅拌16小时。通过硅胶柱采用乙酸乙酯/石油醚(1∶10至1∶1)纯化残余物。得到4.6g(三步73%)的化合物8,其为白色固体。ESI-MS(EI +,m/z):628,1.359min.1H NMR(甲醇-d4,400MHz):δ(ppm)0.30(p,J=5.4Hz,1H),0.48(dt,J=9.1,5.5Hz,1H),0.71(ddt,J=44.0,9.5,5.7Hz,2H),1.45(d,J=2.5Hz,9H),1.58(s,9H),1.76(d,J=15.4Hz,1H),2.51-2.68(m,1H),3.80-4.20(m,2H),4.77-4.86(m,1H),4.94-5.13(m,2H),5.50(dt,J=6.7,3.6Hz,1H),7.29-7.55(m,5H)
步骤9:化合物9的合成
向氮气保护的500-mL三口圆底烧瓶中加入化合物8(4.6g,7.33mmol,1.00当量),三苯基膦(3.15g,11mmol,1.50当量)的THF溶液(250mL),DIAD(2.5g,11mmol,1.50当量)的THF溶液(10mL)。将得到的溶液在室温下搅拌5小时。将得到的溶液在真空下浓缩。通过快速制备HPLC采用以下条件纯化粗产物:柱,C18硅胶;流动相,在50分钟内由ACN=0%增加至ACN=50%;检测器,UV 254nm。得到3.9g(74%)的化合物9,其为白色固体。ESI-MS(EI +,m/z):610,1.119min.
步骤10:化合物10的合成
向1000-mL圆底烧瓶中加入化合物9(3.5g,5.74mmol,1.00当量)的THF溶液(500mL),然后加入Pd/C(400mg)。将产生的溶液在H 2(1atm)下在室温下搅拌5小时。过滤掉固体。将产生的混合物在真空下浓缩。得到2.5g的化合物10粗品,其为白色固体。ESI-MS(EI +,m/z):520,0.964min.
步骤11:化合物11的合成
在250-mL密封管中加入化合物10(2.5g,4.8mmol,1.00当量)的DMF溶液(50mL),然后加入SO 3·Py(3.03g,19.2mmol,4.00当量)。将产生的溶液在室温下搅拌20小时。将产生的混合物在真空下浓缩。 随后通过加入10mL的NaH 2PO 4淬灭反应。向反应混合物中加入NBu 4HSO 4(100mg)。用乙酸乙酯萃取产生的溶液(2x20mL),合并有机层,用无水硫酸钠干燥,真空下浓缩。通过快速制备HPLC采用以下条件纯化残余物:柱,C18硅胶;流动相,在40分钟内由ACN=0%增加至ACN=50%;检测器,UV 254nm。得到2.0g(69.4%)化合物11。ESI-MS(EI +,m/z):600,1.014min.
步骤12:化合物CCC的合成
当用快速色谱纯化时,化合物11不稳定,容易在流动相中脱掉Boc保护基。在浓缩的时候会得到脱掉一个Boc的产物。将脱掉一个Boc的产物在温水中(pH=6-7)搅拌得到脱掉两个Boc保护基的产物CCC。通过制备HPLC采用以下条件纯化产物:柱:XSelect CSH OBD C18柱19*250mm,5um;流动相A:水(0.1%FA),流动相B:ACN;流速:25mL/min;梯度:在13分钟内由4%B至4%B;220/254nm;Rt:11.27,12.23min;得到43.4mg的化合物CCC。ESI-MS(EI +,m/z):400,0.650min. 1H NMR(400MHz,D 2O)δ(ppm)0.34-0.61(m,2H),0.63-0.92(m,1H),1.82(d,J=16.0Hz,1H),2.64(dd,J=16.1,7.8Hz,1H),3.18(d,J=12.2Hz,1H),3.36(dd,J=12.2,3.8Hz,1H),3.52(d,J=3.8Hz,1H),4.02(dd,J=10.5,4.8Hz,1H),4.21(t,J=10.3Hz,1H),4.99(d,J=7.6Hz,1H),5.54(dd,J=10.0,4.8Hz,1H).
实施例56化合物DDD的制备
Figure PCTCN2019073000-appb-000110
Figure PCTCN2019073000-appb-000111
步骤1:化合物1的合成
向500-mL圆底烧瓶中加入D-丝氨酸(5g,47.58mmol,1.00当量)的二噁烷溶液(200mL),然后加入NaOH(4g,100.01mmol,2.10当量)的水溶液(100mL)。随后,将得到的溶液在室温下搅拌5分钟。然后加入(Boc) 2O(12g,54.98mmol,1.14当量)。将得到的溶液在室温下继续搅拌16小时。用EA萃取得到的混合物(2x200mL)。用HCl(1mol/L)调节水层pH至约1。将得到的溶液用乙酸乙酯萃取(3x200mL)并且合并有机层,用无水硫酸钠干燥,真空下浓缩。得到10.2g(粗)的2(S)-2-[[(叔丁氧基)羰基]氨基]-3-羟基丙酸,其为无色油状物。ESI-MS(EI +,m/z+Na):228,0.787min.
步骤2:化合物2的合成
向500-mL圆底烧瓶中加入化合物1(10.2g,49.71mmol,1.00当量)的THF溶液(200mL),TEA(10g,98.82mmol,2.00当量),DMAP(6g, 49.11mmol,1.00当量),TBS-Cl(10.5g,69.66mmol,1.40当量)。将得到的溶液在室温下搅拌16小时。反应完全后,用HCl(1mol/L)调节pH至1。用乙酸乙酯萃取得到的溶液(2x300mL)并合并有机层,无水硫酸钠干燥,真空下浓缩。得到14.8g(93%)的2(R)-2-[[(叔丁氧基)羰基]氨基]-3-[(叔丁基二甲基硅基)氧基]丙酸,其为无色油状物。ESI-MS(EI +,m/z+Na):320,0.730min.
步骤3:化合物3的合成
向200-mL圆底烧瓶中加入2(R)-2-[[(叔丁氧基)羰基]氨基]-3-[(叔丁基二甲基硅基)氧基]丙酸(14.8g,46.3mmol,1.00当量)的DCM溶液(100mL)和CDI(10g,312.06mmol,6.00当量)。将得到的溶液在室温下搅拌120分钟,然后加入肼(11g,220.00mmol,10.00当量)。将得到的溶液在室温下继续搅拌60分钟。将得到的混合物在真空下浓缩。通过快速制备HPLC采用以下条件纯化粗物质:柱,C18硅胶;流动相,在30分钟内由ACN增加至ACN=50%;检测器,UV 254nm。得到10g(64.7%)的(R)-(3-((叔丁基二甲基硅基)氧基)-1-(肼基羰基)-1-氧代丙烷-2-基)氨基甲酸叔丁酯,其为白色固体。ESI-MS(EI +,m/z+):334,1.165min.1H NMR(DMSO-d6,400MHz):δ(ppm)0.84(s,9H),1.37(s,9H),3.65(ddd,J=35.5,10.0,6.1Hz,2H),4.04(q,J=6.9Hz,1H),4.20(d,J=3.7Hz,2H),6.58(d,J=8.8Hz,1H),9.10(s,1H).
步骤4:化合物4的合成
向200-mL圆底烧瓶中加入化合物3(3.5g,10.49mmol,1.00当量),(1R,4S)-7-(苄氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1’-环丙烷]-4-羧酸(4.7g,15.55mmol,1.2当量),DIPEA(4.5g,34.88mmol,3.00当量),HATU(8.74g,23.00mmol,4.00当量)的THF溶液(100mL)。将得到的溶液在室温下搅拌2小时。过滤掉固体。通过硅胶柱用乙酸乙酯/石油醚(1/5至1/1)纯化残余物。得到7.8g(粗)的化合物4,其为白色固体。ESI-MS(EI +,m/z+):618,1.402min.
步骤5:化合物5的合成
向200-mL圆底烧瓶中加入化合物4(7.8g,12.6mmol,1.00当量)的DCM溶液(100mL),然后加入DIEA(5.67g,43.95mmol,3.50当量),Burgess试剂(10.42g,43.97mmol,3.50当量)。将得到的溶液在室温下搅拌20小时。通过硅胶柱采用乙酸乙酯/石油醚(1/5至1/1)纯化残余物,得到6g(两步88%)的化合物5,其为白色固体。ESI-MS(EI +,m/z+Na):622,1.477min.1H NMR(氯仿-d,400MHz):δ(ppm)-0.16(s,6H),0.06-0.26(m,1H),0.50(ddt,J=36.1,9.3,5.7Hz,2H),0.79(d,J=2.0Hz,9H),1.45(d,J=3.4Hz,8H),2.67(dd,J=15.3,7.8Hz,1H),2.79(s,2H),2.81-2.99(m,2H),3.70-4.17(m,2H),4.64-4.94(m,2H),5.05(d,J=11.5Hz,2H),5.44(t,J=11.3Hz,1H),7.39(ddd,J=19.3,6.1,2.4Hz,5H).
步骤6:化合物6的合成
向100-mL圆底烧瓶中加入化合物5(6g,10mmol,1.00当量)的DCM溶液(40mL),然后加入TFA(10mL,10.00当量)。将得到的溶液在0℃下在室温下搅拌120分钟。将得到的混合物在真空下浓缩。得到6g化合物6粗品,不经纯化直接用于下一步。ESI-MS(EI +,m/z+Na):500,1.546min.
步骤7:化合物7的合成
在100-mL圆底烧瓶中加入化合物6(6g,10mmol,1.00当量)的THF溶液(60mL),然后加入3HF Et3N(15mL,10.00当量)。将得到的溶液在室温下搅拌2小时。粗产品通过快速制备HPLC用以下条件纯化:柱,C18硅胶;流动相,在40分钟内由ACN增加至ACN=50%;检测器,UV 254nm。得到3.8g化合物7,其为白色固体。ESI-MS(EI +,m/z+Na):386,0.878min.
步骤8:化合物8的合成
向100-mL圆底烧瓶中加入化合物7(3.8g,9.8mmol,1.00当量)的ACN溶液(100mL),TEA(6.3g,42.32mmol,4.30当量),(1H-吡唑-1- 基)甲烷二基亚基二氨基甲酸二叔丁酯(5.28g,17mmol,1.70当量)。将得到的溶液在40℃下搅拌16小时。通过硅胶柱采用乙酸乙酯/石油醚(1∶10至1∶1)纯化残余物。得到4.6g(75.4%)的化合物8,其为白色固体。ESI-MS(EI +,m/z+Na):628,1.356min.1H NMR(甲醇-d4,400MHz):δ(ppm)0.30(dq,J=10.0,5.2,4.2Hz,1H),0.48(dt,J=9.1,5.5Hz,1H),0.48-0.85(m,3H),1.46(d,J=2.5Hz,9H),1.58(s,18H),2.04(s,2H),2.47-2.71(m,1H),2.84(d,J=6.9Hz,2H),4.01(dt,J=11.5,3.5Hz,1H),4.05-4.23(m,2H),4.83(s,1H),4.94-5.12(m,2H),5.50(dt,J=7.1,3.6Hz,1H),7.21-7.55(m,5H),7.64(s,1H)
步骤9:化合物9的合成
向氮气保护的500-mL三口圆底烧瓶中加入化合物8(4.6g,7.33mmol,1.00当量),三苯基膦(3.15g,11mmol,1.50当量)的THF溶液(250mL),DIAD(2.5g,11mmol,1.50当量)的THF溶液(10mL)。将得到的溶液在室温下搅拌5小时。将得到的溶液在真空下浓缩。通过快速制备HPLC采用以下条件纯化粗产物:柱,C18硅胶;流动相,在30分钟内由ACN=0%增加至ACN=60%;检测器,UV 254nm。得到3.9g(87.6%)的化合物9,其为白色固体。ESI-MS(EI +,m/z+Na):610,1.120min.
步骤10:化合物10的合成
向1000-mL圆底烧瓶中加入化合物9(3.5g,5.74mmol,1.00当量)的THF溶液(500mL),然后加入Pd/C(300mg)。将产生的溶液在H 2(1atm)下在室温下搅拌5小时。过滤掉固体。将产生的混合物在真空下浓缩。得到2.5g的化合物10粗品,其为黄色固体。ESI-MS(EI +,m/z+Na):520,0.967min.
步骤11:化合物11的合成
在250-mL密封管中加入化合物10(2.5g,4.8mmol,1.00当量)的DMF溶液(50mL),然后加入SO3.Py(3.03g,19.2mmol,4.00当量)。 将产生的溶液在室温下搅拌20小时。将产生的混合物在真空下浓缩。随后通过加入10mL的NaH 2PO 4淬灭反应。向反应混合物中加入NBu 4HSO 4(100mg)。用乙酸乙酯萃取产生的溶液(2x20mL),合并有机层,用无水硫酸钠干燥,真空下浓缩。通过快速制备HPLC采用以下条件纯化残余物:柱,C18硅胶;流动相,在30分钟内由ACN=0%增加至ACN=80%;检测器,UV 254nm。得到2.0g(69.4%)化合物11。ESI-MS(EI +,m/z+Na):600,1.012min.
步骤12:化合物DDD的合成
当用快速色谱纯化时,化合物11不稳定,容易在流动相中脱掉Boc保护基。在浓缩的时候会得到脱掉一个Boc的产物。将脱掉一个Boc的产物在温水中(pH=6-7)搅拌得到脱掉两个Boc保护基的产物DDD。通过制备HPLC采用以下条件纯化产物:柱:XSelect CSH OBD C18柱19*250mm,5um;流动相A:水(0.1%FA),流动相B:ACN;流速:25mL/min;梯度:在13分钟内由4%B至4%B;220/254nm;Rt:11.27,12.23min;得到105mg的化合物DDD。ESI-MS(EI +,m/z):400,0.650min.HPLC:4.360,纯度:97%.1H NMR(D 2O,400MHz):δ(ppm)0.34-0.61(m,2H),0.63-0.92(m,1H),1.82(d,J=16.0Hz,1H),2.64(dd,J=16.1,7.8Hz,1H),3.18(d,J=12.2Hz,1H),3.36(dd,J=12.2,3.8Hz,1H),3.52(d,J=3.8Hz,1H),4.02(dd,J=10.5,4.8Hz,1H),4.21(t,J=10.3Hz,1H),4.99(d,J=7.6Hz,1H),5.54(dd,J=10.0,4.8Hz,1H).
实施例57化合物EEE的制备
Figure PCTCN2019073000-appb-000112
步骤1:化合物1的合成
向叔丁氧羰基甘氨酸甲酯(9g,47.6mmol,1当量)的乙醇溶液(50mL)中加入NH 2NH 2H 2O溶液(15mL)。将反应混合物在70℃下搅拌2小时。减压蒸去溶剂。粗产物不经进一步纯化用于下一步。M/z(ES+)[M+Na]+=212,酸,HPLC tR=0.509min.
步骤2:化合物2的合成
0℃下,向100mL圆底烧瓶中放入(1R,4S)-7-(苄氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-羧酸(2g,6.6mmol,1.00当量),(2-肼基-2-氧代乙基)氨基甲酸叔丁酯(2.5g粗品,13.2mmol,2.00当量),HATU(5.03g,13mmol,2.00当量)的THF溶液(300mL)。加入DIPEA(2.56g,20mmol,3.00当量)。将生成的溶液在0℃下搅拌2小时。过滤掉固体。浓缩滤液,并用硅胶柱色谱(0-60%梯度洗脱MeOH/DCM)纯化得到粗产品(3.3g,100%),其为浅黄色固体。m/z(ES+),[M+H]+=474;TFA,HPLC tR=1.131min.
步骤3:化合物3的合成
向100mL圆底烧瓶中放入化合物2(3.3g,7mmol,1.00当量),DIEA(2.69g,21mmol,3当量)的DCM溶液(50mL)。加入Burgess试剂(5g,21mmol,3当量)。将生成的溶液在室温下搅拌20小时。将生成的混合物在减压下浓缩。将粗产物通过硅胶柱色谱(0-60%梯度洗脱EA/PE)纯化得到产品化合物3(3.5g,粗),其为浅黄色固体。m/z(ES+),[M+H]+=456;TFA,HPLC tR=1.181min.
步骤4:化合物4的合成
向250mL圆底烧瓶中加入化合物3(3.3g,7mmol)的THF溶液(100mL)。加入Pd/C(600mg)。将生成的溶液在H2下(1atm)在室温下搅拌2小时。过滤掉固体。将滤液减压浓缩,生成2.8g粗产品,其为白色固体,直接用于下一步。m/z(ES+),[M+Na]+=388;TFA,HPLC tR=0.736min.
步骤5:化合物5的制备
向25mL圆底烧瓶中加入化合物4(219mg,0.6mmol)的DCM溶液(10mL)。在0℃下加入三乙胺(90mg,0.9mmol,1.5当量)和2-溴乙酸叔丁酯(350mg,1.8mmol,3当量)。将生成的溶液在0℃下搅拌过夜。粗产物经过硅胶柱色谱(0-60%梯度洗脱EA/PE)纯化得到产物化合物5(70mg,24.3%),其为浅黄色固体。m/z(ES+),[M+H]+=480;TFA,HPLC tR=1.200min.
步骤6:化合物EEE的合成
向25mL圆底烧瓶中放入化合物5(70mg,0.14mmol)的DCM溶液(10mL)。在0℃下加入TFA(5mL)。将生成的溶液在0℃下搅拌2小时。将粗产物经制备型HPLC(XBridge制备型C18 OBD柱,5μ silica,直径 19mm,长度150mm)纯化,使用水(含0.01%FA)和乙腈的渐小极性混合物作为洗脱剂。将含有期望的化合物的级分蒸发至干得到2-(((1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)乙酸(11.9mg,25.3%)。m/z(ES+),[M+H]+=324;TFA,HPLC tR=0.418min.1H NMR(300MHz,DMSO-d6)δ7.59(s,2H),4.82(d,J=7.5Hz,1H),4.63(d,J=16.4Hz,1H),4.31(d,J=32.3Hz,1H),4.11(d,J=6.2Hz,2H),3.03(d,J=10.9Hz,1H),2.85(d,J=11.7Hz,2H),1.71(d,J=15.3Hz,1H),1.27(d,J=6.8Hz,1H),0.81(d,J=6.2Hz,1H),0.65(s,1H),0.40(d,J=8.2Hz,2H).
实施例58化合物FFF的制备
Figure PCTCN2019073000-appb-000113
按照实施例60中的步骤1-5制备得到化合物5。
步骤6:化合物6的制备
在10mL圆底烧瓶中放入化合物5(128mg,0.35mmol)的DCM溶液(3mL)。在℃下加入三乙胺(53.3mg,0.53mmol,1.5当量)和2-溴乙酸叔丁酯(204.7mg,1.1mmol,3当量)。将生成的溶液在60℃下搅拌过夜。将粗产物经硅胶柱色谱(0-60%梯度洗脱EA/PE)纯化得到产物化 合物6(50mg,29.7%),其为浅白色固体。m/z(ES+),[M+H]+=479;酸,HPLC tR=1.122min.
步骤7:化合物FFF的合成
在0℃下在10mL圆底烧瓶中放入化合物6(50mg,0.11mmol)的DCM/TFA(8ml/4ml)溶液。将生成的溶液在0℃下搅拌2小时。将粗产物经制备型HPLC(XBridge制备型C18 OBD柱,5μ silica,直径19mm,长度150mm)纯化,使用水(含0.01%FA)和乙腈的渐小极性混合物作为洗脱剂。将含有期望的化合物的级分蒸发至干得到2-(((1R,4S)-4-(5-(((叔丁氧羰基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)乙酸(4.2mg,12.4%)。m/z(ES+),[M+H]+=323;酸,HPLC tR=0.549min.1H NMR(300MHz,DMSO-d6)δ6.41(s,1H),4.58(d,J=8.0Hz,1H),4.32(d,J=16.2Hz,1H),3.32(d,J=3.4Hz,2H),3.02-2.90(m,1H),2.79(d,J=11.6Hz,1H),2.39(dd,J=14.8,7.7Hz,1H),1.64(d,J=14.9Hz,1H),0.78(d,J=5.5Hz,1H),0.50(d,J=9.3Hz,1H),0.42-0.22(m,1H).
实施例59化合物GGG的制备
Figure PCTCN2019073000-appb-000114
步骤1:化合物1的制备
在100mL圆底烧瓶中放入化合物1(455mg,1mmol)的THF(20mL) 溶液。加入Pd/C(100mg)。将生成的溶液在H2下(1atm)在室温下搅拌2小时。过滤掉固体。将滤液减压浓缩,生成2.8g粗产品,其为白色固体,直接用于下一步。m/z(ES+),[M+Na]+=388;TFA,HPLC tR=0.736min.
步骤2:化合物2的制备
在10mL圆底烧瓶中放入化合物1(200mg,0.55mmol)的DMF(100mL)溶液。在室温下加入碳酸钾(151mg,1.1mmol,2当量)和2-溴-2,2-二氟乙酸乙酯(126mg,0.55mmol,1当量)。将生成的溶液在室温下搅拌4小时。粗产物经硅胶柱色谱(0-60%梯度洗脱EA/PE)纯化得到产品化合物2(74mg,26.3%),其为浅白色固体。m/z(ES+),[M+H]+=516;TFA,HPLC=1.273min
步骤3:化合物GGG的合成
在10mL圆底烧瓶中放入化合物2(74mg,0.14mmol)的DCM(10mL)溶液。在0℃下加入TFA(5mL)。将生成的溶液在0℃下搅拌2小时。将粗产物经制备型HPLC(XBridge制备型C18 OBD柱,5μ silica,直径19mm,长度150mm)纯化,使用水(含0.01%FA)和乙腈的渐小极性混合物作为洗脱剂。将含有期望的化合物的级分蒸发至干得到2-(((1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)-2,2-二氟乙酸(6.4mg,12.4%)。m/z(ES+),[M+H]+=360;TFA,HPLC tR=0.660min.1H NMR(300MHz,D2O)δ4.98(d,J=7.7Hz,2H),4.42(d,J=12.1Hz,2H),3.39(d,J=3.8Hz,1H),3.33-3.18(m,1H),3.15(d,J=12.2Hz,1H),2.61(d,J=7.7Hz,1H),1.80(d,J=16.0Hz,1H),0.84-0.59(m,2H),0.64-0.31(m,3H).
实施例60化合物HHH的制备
Figure PCTCN2019073000-appb-000115
步骤1:化合物1的合成
在0℃下将LiBH4/THF(1.13g,51mmol,4当量)添加至(1R,4S)-7-(苄氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-羧酸苄酯(5g,12.76mmol,1当量)的甲醇(50mL)溶液中。将混合物在0℃下搅拌4-5小时。在0℃下通过加入饱和NaH 2PO 4(50mL)小心地淬灭反应混合物。将混合物用水(20mL)稀释并用DCM萃取三次。浓缩合并的有机层并用硅胶柱色谱(100∶0至100∶1己烷/乙酸乙酯)纯化得到期望的产品化合物1(2.6g,71%)。m/z(ES+),[M+H]+=289;酸,HPLC tR=0.789min.
步骤2:化合物2的合成
在0℃下将TEMPO(14.08mg,9mmol,0.01当量)分批加入化合物1(2.6g,9mmol,1当量)和1,3,5-三氯-1,3,5-三嗪烷(2.08g,9mmol,1当量)的DCM(25mL)溶液中。将混合物在0℃下搅拌2小时,并经硅藻土过滤。将滤液用硫酸钠干燥并浓缩得到粗产物化合物2,其不经进一步纯化直接用于下一步。m/z(ES+),[M+H]+=287;酸,HPLC tR=0.931min.
步骤3:化合物3的合成
将粗产物化合物2(2.6g,8.74mmol,1当量),羟胺盐酸盐(0.69g,10.1mmol,1.16当量)和吡啶(2.76g,34.9mmol,4当量)的乙醇(25mL)溶液在室温下搅拌2小时。然后将反应混合物浓缩,并将残余物用DCM稀释,用水、饱和氯化钠洗涤,用Na2SO4干燥并浓缩。将残余物经硅胶柱色谱(100∶0至100∶1己烷/乙酸乙酯)纯化得到期望的产品化合物3(1.48g,54%)。m/z(ES+),[M+H]+=287;酸,HPLC tR=0.873min.
步骤4:化合物4的合成
将苯基-I3-碘烷二基-双(2,2,2-三氟乙酸酯)(3.6g,8.37mmol,1.5当量)分批加入化合物3(1.48g,5.58mmol,1当量)和丙-2-炔-1-基氨基甲酸叔丁酯(0.86g,5.58mmol,1当量)的甲醇(20mL)和水(4mL)的溶液中。将生成的混合物在室温下搅拌4小时。在减压下除去溶剂。将粗产物通过快速C18-快速色谱纯化,0至65%的乙腈水溶液梯度洗脱。将纯的级分蒸发至干得到化合物4(250mg,11%),其为白色固体。m/z(ES+),[M+H]+=455;酸,HPLC tR=1.288min.1H NMR(300MHz,氯仿-d)δ7.48-7.33(m,6H),6.32(s,1H),5.09(d,J=11.5Hz,1H),4.93(d,J=11.5Hz,2H),4.69(d,J=7.4Hz,1H),4.45(s,2H),2.96(dd,J=11.6,3.7Hz,1H),2.85(d,J=11.4Hz,1H),2.61(dd,J=15.1,7.5Hz,1H),2.40(d,J=3.7Hz,1H),1.79(d,J=15.1Hz,1H),1.48(s,9H),0.72(dt,J=10.2,5.4Hz,1H),0.51(ddd,J=20.9,9.9,5.3Hz,2H),0.17-0.03(m,1H).
步骤5:化合物5的合成
在100mL圆底烧瓶中放入化合物4(128mg,2.8mmol)的THF(6mL)溶液。添加Pd/C(20mg)。将生成的溶液在H2下(1atm)在室温下搅拌2小时。过滤掉固体。将滤液减压浓缩,形成128mg粗产品化合物5, 其为白色固体,直接用于下一步。m/z(ES+),[M+H]+=365;酸,HPLC tR=0.822min.
步骤6:化合物6的合成
在10mL圆底烧瓶中放入化合物5(150mg,0.41mmol)的DMF(5mL)溶液。在0℃下添加碳酸钾(113.6mg,0.82mmol,2当量)和2-溴-2,2-二氟乙酸叔丁酯(94.5mg,0.41mmol,1当量)。将生成的溶液在60℃下搅拌过夜。将粗产物经硅胶柱色谱(0-60%梯度洗脱EA/PE)纯化得到产品化合物6(50mg,23.69%),其为浅白色固体。m/z(ES+),[M+H] +=515;酸,HPLC tR=1.325min.
步骤7:化合物HHH的合成
在0℃下在10mL圆底烧瓶中放入化合物6(50mg,0.11mmol)的DCM/TFA(8ml/4ml)溶液。将生成的溶液在0℃下搅拌2小时。将粗产物通过制备型HPLC(XBridge制备型C18 OBD柱,5μ silica,直径19mm,长度150mm)纯化,使用水(含0.01%FA)和乙腈的渐小极性混合物作为洗脱剂。将含有期望的化合物的级分蒸发至干得到2-(((1R,4S)-4-(5-(((叔丁氧羰基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)-2,2-二氟乙酸(1.5mg,4.3%)。m/z(ES+),[M+H]+=359;酸,HPLC tR=0.868min.1H NMR(400MHz,氯仿-d)δ8.23-7.58(m,2H),6.61(s,1H),4.85-4.62(d,J=18.8Hz,1H),4.31-4.21(s,2H),3.29-3.21(m,1H),3.15-3.05(m,1H),2.88-2.78(m,1H),2.51-2.42(m,1H),1.72-1.60(d,J=6.6Hz,1H),0.70-0.61(m,1H),0.53-0.45(m,1H),0.43-0.39(m,1H),0.38-0.30(m,1H).
实施例61化合物III的合成
Figure PCTCN2019073000-appb-000116
步骤1:化合物1的合成
向500mL圆底烧瓶中加入(R)-3-氨基-2-羟基丙酸(3g,28.57mmol,1.00当量)的二噁烷(60mL)溶液,并在冰浴冷却下加入氢氧化钠(2.4g,60mmol,2.10当量)的水溶液(60mL)。将所得溶液在0℃下搅拌5分钟。然后在0℃下逐滴加入二碳酸二叔丁酯((Boc) 2O)(7.1g,32.57mmol,1.14当量)的二噁烷溶液(20mL)。将得到的溶液在室温下搅拌过夜。用乙酸乙酯(2×200mL)洗涤所得混合物。用HCl(1mol/L)将水相调节至PH~2。用乙酸乙酯(2×200mL)萃取所得溶液。合并有机层,用硫酸钠干燥,真空浓缩,得到5.2g(R)-3-((叔丁氧基羰基)氨基)-2-羟基丙酸粗品,为无色油状物。m/z(ES+),[M+Na] +=228;HPLC tR=0.701min.
步骤2:化合物2的合成
向250mL圆底烧瓶中加入(R)-3-((叔丁氧基羰基)氨基)-2-羟基丙酸(5.2g,25.36mmol,1.00当量)和叔丁基二甲基氯硅烷(TBSCl)(5.32g,35.5mol,1.40当量)的四氢呋喃溶液(80mL)。逐滴加入三乙胺(5.12g,50.72mol,2当量)。将得到的溶液在室温下搅拌20小时。用HCl(1mol/L) 将所得溶液调节至pH约为2。用乙酸乙酯(2×300mL)萃取所得溶液。合并有机层,用硫酸钠干燥,真空浓缩,得到9g(R)-3-((叔丁氧基羰基)氨基)-2-((叔丁基二甲基甲硅烷基)氧基)丙酸粗品,为白色固体。m/z(ES+),[M+H] +=320;HPLC tR=3.01min.
步骤3:化合物3的合成
向250mL圆底烧瓶中加入(R)-3-((叔丁氧基羰基)氨基)-2-((叔丁基二甲基甲硅烷基)氧基)丙酸(9g,28.21mmol,1.00当量)的二氯甲烷溶液(100mL)。在0℃下分批加入N’,N-羰基二咪唑(CDI)(7g,42.32mmol,1.50当量)。将所得溶液在室温下搅拌120分钟。然后在室温下滴加水合肼(NH 2NH 2.H 2O)(98%)(7g,140mmol,5当量)。将得到的溶液在室温下再搅拌60分钟。分离有机层并将其在真空下浓缩。通过柱色谱(石油醚/乙酸乙酯=1/5-1/1)纯化粗产物,得到3.7g(38.9%,3步)的(R)-(2-((叔丁基二甲基甲硅烷基)氧基)-3-肼基-3-氧代丙基)氨基甲酸叔丁酯,为白色固体。m/z(ES+),[M+H] +=334;HPLC tR=1.093min.
步骤4:化合物4的合成
在0℃下向100mL圆底烧瓶中加入(1R,4S)-7-(苄氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-羧酸(3g,10mmol,1.00当量),(R)-(2-((叔丁基二甲基甲硅烷基)氧基)-3-肼基-3-氧代丙基)氨基甲酸叔丁酯(3.7g,11.1mmol,1.10当量),2-(7-氧化苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸盐(HAUT)(5.7g,15mmol,1.5当量)的四氢呋喃溶液(50mL)。在0℃下滴加N,N-二异丙基乙胺(DIPEA)(3.22g,25mmol,2.5当量)。将所得溶液在0℃下搅拌2小时。滤出固体,浓缩滤液。通过柱色谱(石油醚/乙酸乙酯=1/5-1/1)纯化粗产物,得到4.5g粗产物4,为黄色固体。m/z(ES +),[M+H] +=618;HPLC tR=1.351min.1H NMR(氯仿-d,400MHz):δ(ppm)0.10(td,J=8.8,8.1,5.2Hz,1H),0.15(d,J=6.3Hz,6H),0.43(dt,J=8.9,6.3Hz,1H),0.54(td,J=9.2,4.5Hz,2H),0.93(s,9H),2.26-2.46(m,2H),2.81(s,6H),3.12(dd,J=11.7,3.9Hz, 1H),3.20-3.45(m,2H),3.59(dt,J=13.4,6.5Hz,1H),4.01-4.18(m,2H),4.29(t,J=4.7Hz,1H),4.85-5.10(m,2H),5.36(s,1H),7.34-7.49(m,5H),8.48(d,J=86.2Hz,2H).
步骤5:化合物5的合成
向100mL圆底烧瓶中加入中间产物4(4.5g,7.3mmol,1.00当量)和DIEA(3.8g,29mmol,4.00当量)在二氯甲烷中的溶液(40mL)。在室温下分批加入Burgess试剂(6.9g,29mmol,4.00当量)。将得到的溶液在室温下搅拌20小时。浓缩混合物,并通过柱色谱(石油醚/乙酸乙酯=1/5-1/1)纯化残余物,得到4.3g(73%,2步)产物5,为白色固体。m/z(ES+),[M+Na] +=622;HPLC tR=1.400min.1H NMR(甲醇-d4,300MHz):δ(ppm)0.06(s,3H),0.16(s,3H),0.31(dt,J=10.1,5.3Hz,1H),0.43-0.54(m,1H),0.66(dt,J=10.8,5.4Hz,1H),0.78(dt,J=9.5,5.3Hz,1H),0.90(s,9H),1.40(s,8H),1.75(d,J=15.4Hz,1H),2.44-2.74(m,1H),2.76-2.89(m,2H),3.03(d,J=2.4Hz,2H),3.46(d,J=6.4Hz,2H),4.84(d,J=7.6Hz,1H),4.93-5.15(m,3H),7.34-7.57(m,5H).
步骤6:化合物6的合成
向250mL圆底烧瓶中加入中间产物5(4g,6.67mmol,1.00当量)在四氢呋喃(100mL)中的溶液,再加入Pd/C(1g)。将所得溶液在室温下在H 2(1atm)下搅拌2小时。反应完成后,滤出固体。将得到的滤液真空浓缩,得到4g粗产物6,为白色固体。m/z(ES+),[M+Na] +=532;HPLC tR=1.238min.
步骤7:化合物7的合成
在室温下向50mL圆底烧瓶中加入中间产物6(4g,6.67mmol,1.00当量)在DMF(30mL)中的溶液,然后分批加入SO 3·Py(5.3g,33.5mmol,5.00当量)。将得到的溶液在室温下搅拌20小时。将所得混合物真空浓缩。向粗产物中加入40mL硫酸二氢钠饱和溶液。然后加入3克四 丁基硫酸氢铵。然后用乙酸乙酯(2×100mL)萃取溶液。合并有机层,用硫酸钠干燥,真空浓缩,得到5g粗产物7,为浅黄色固体。m/z(ES+),[M-H] +=588;HPLC tR=2.311min.1H NMR(甲醇-d4,400MHz):δ(ppm)0.07(d,J=2.7Hz,3H),0.16(s,3H),0.37(dt,J=10.3,5.5Hz,1H),0.49(dt,J=9.0,5.6Hz,1H),0.71-0.89(m,3H),0.91(s,9H),1.03(d,J=7.4Hz,9H),1.24(d,J=7.1Hz,3H),1.82(d,J=15.4Hz,1H),2.59-2.76(m,1H),3.35-3.63(m,3H),4.11(q,J=7.2Hz,2H),4.95-5.15(m,1H).
步骤8:化合物8的合成
向100mL圆底烧瓶中加入中间产物7(5g,8.5mmol,1.00当量)在四氢呋喃(40mL)中的溶液。在室温下滴加3HF·Et 3N(5mL)。将得到的溶液在室温下搅拌20小时。将反应混合物真空浓缩,通过快速柱色谱纯化粗产物(条件如下:C-18柱;流动相,乙腈在30分钟内由0%增加至100%;检测器:UV 220nm),得到3g粗产物8,为黄色油状物。m/z(ES+),[M-H] +=474;HPLC tR=1.261min.
步骤9:化合物III的合成
向500mL圆底烧瓶中加入中间产物8(3g,6.3mmol,1.00当量)在二氯甲烷(50mL)中的溶液,并在0℃下向其滴加三氟乙酸(15mL,10.00当量)。将所得溶液在0℃下搅拌120分钟。将得到的混合物在0℃下真空浓缩。加入乙醚以使产物悬浮为固体。通过离心沉淀收集产物。粗制产物通过制备型高效液相色谱纯化(条件如下:流动相A:水(无缓冲液),流动相B:乙腈;流速:40mL/min;梯度:7分钟内由0%B至15%B;254,220nm;tR=3.4min),得到(1R,4S)-4-(5-((R)-2-氨基-1-羟乙基)-1,3,4-噁二唑-2-基-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸(798.3mg,4步29.7%),为白色固体。m/z(ES+),[M+H] +=376;HPLC tR=0.683min.1H NMR(D 2O,400MHz):δ(ppm)0.38(p,J=5.0Hz,1H),0.42-0.55(m,1H),0.58-0.74(m,2H), 1.77(dd,J=16.1,4.3Hz,1H),2.29-2.66(m,1H),2.96-3.21(m,1H),3.28(dt,J=12.4,4.3Hz,1H),3.43(qd,J=8.3,7.4,4.3Hz,2H),3.47-3.66(m,1H),4.91(dd,J=7.9,4.2Hz,1H),5.18-5.37(m,1H).
实施例62化合物JJJ的合成
Figure PCTCN2019073000-appb-000117
步骤1:化合物1的合成
向5L圆底烧瓶中加入(S)-3-氨基-2-羟基丙酸(95g,904mmol,1.00当量)在二噁烷(1.9L)中的溶液,再在冰浴冷却下加入氢氧化钠溶液(76g,1.9mol,2.10当量)的水溶液(1.9L)。将所得溶液在0℃下搅拌5分钟。然后在0℃下滴加(Boc) 2O(225g,1.03mol,1.14当量)。将得到的溶液在室温下搅拌过夜。用乙酸乙酯(2×2L)洗涤所得混合物。用HCl(1mol/L)将水相的pH调节至2。用乙酸乙酯(2×3L)萃取所得溶液。合并有机层,用硫酸钠干燥,真空浓缩,得到195g(S)-3-((叔丁氧基羰基)氨基)-2-羟基丙酸粗品,为无色油状物。m/z(ES+),[M+Na] +=228;HPLC tR=0.692min.
步骤2:化合物2的合成
向5L圆底烧瓶中加入(S)-3-((叔丁氧基羰基)氨基)-2-羟基丙酸(190g,0.926mol,1.00当量)和TBSCl(190g,1.29mol,1.40当量)的四氢呋喃(3L)溶液。逐滴加入三乙胺(187g,1.85mol,2当量)。将得到的溶液在室温下搅拌20小时。用HCl(1mol/L)将所得溶液调节至pH~2。然后用乙酸乙酯(2×3L)萃取所得溶液。合并有机层,用硫酸钠干燥,真空浓缩,得到300g(S)-3-((叔丁氧基羰基)氨基)-2-((叔丁基二甲基甲硅烷基)氧基)丙酸粗品,为白色固体。m/z(ES+),[M+Na] +=342;HPLC tR=2.080min.
步骤3:化合物3的合成
向5L圆底烧瓶中加入(S)-3-((叔丁氧基羰基)氨基)-2-((叔丁基二甲基甲硅烷基)氧基)丙酸(300g,940mmol,1.00当量)的二氯甲烷(3L)溶液,再在0℃下分批加入CDI(228g,1.41mol,1.50当量)。将所得溶液在室温下搅拌120分钟,然后在室温下滴加NH 2NH 2·H 2O(98%)(235g,4.7mol,5当量)。将得到的溶液在室温下再搅拌60分钟。分离有机层,并将其在真空下浓缩。通过柱色谱(石油醚/乙酸乙酯=1/5-1/1)纯化粗产物,得到35g(11.6%)的(S)-(2-((叔丁基二甲基甲硅烷基)氧基)-3-肼基-3-氧代丙基)氨基甲酸叔丁酯,为白色固体。m/z(ES+),[M+H] +=334;HPLC tR=1.046min.1H NMR(DMSO-d 6,300MHz):δ(ppm)0.01(d,J=35.0Hz,6H),0.85(s,9H),1.36(s,9H),2.92-3.20(m,2H),3.96-4.14(m,2H),6.70(t,J=5.7Hz,1H),8.79(s,1H).
步骤4:化合物4的合成
向1L圆底烧瓶中加入(1R,4S)-7-(苄氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-羧酸(28.5g,94mmol,1.00当量),(S)-(2-((叔丁基二甲基甲硅烷基)氧基)-3-肼基-3-氧代丙基)氨基甲酸叔丁酯(35g,104mmol,1.10当量)和HATU(53.3g,141mmol,1.5当量)在干燥四氢呋喃(300mL)中的溶液。在0℃下滴加DIPEA(30g,235mmol,2.5当量)。将所得溶液在0℃下搅拌2小时,滤出固体。通 过柱色谱(石油醚/乙酸乙酯=1/5-1/1)纯化粗产物,得到60g粗产物4,为黄色固体。m/z(ES+),[M+H] +=618;HPLC tR=1.320min.1H NMR(氯仿-d,300MHz):δ(ppm)0.04-0.11(m,1H),0.17(d,6H),0.34-0.63(m,3H),0.94(s,9H),1.42(s,9H),2.24-2.38(m,2H),2.79(s,10H),3.12(dd,J=11.7,3.5Hz,1H),3.44(t,J=5.6Hz,2H),4.01-4.18(m,2H),4.30(t,J=4.9Hz,1H),4.83-5.10(m,3H),5.29(s,1H),7.30-7.45(m,5H),8.41(dd,J=55.5,3.8Hz,2H).
步骤5:化合物5的合成
向1L圆底烧瓶中加入产物4(60g,97mmol,1.00当量)和DIEA(52g,388mmol,4.00当量)在二氯甲烷(500mL)中的溶液。分批加入Burgess试剂(95g,388mmol,4.00当量)。将得到的溶液在室温下搅拌20小时。浓缩混合物,并通过柱色谱(石油醚/乙酸乙酯=1/5-1/1)纯化残余物,得到45g(79%,2步)产物5,为黄色固体。m/z(ES+),[M+H] +=600;HPLC tR=1.400min.1H NMR(甲醇-d4,300MHz):δ(ppm)0.06(s,3H),0.15(s,3H),0.27(dd,J=9.6,5.1Hz,1H),0.46(dt,J=9.3,5.3Hz,1H),0.58-0.80(m,2H),0.90(s,9H),1.38(s,8H),1.79(d,J=15.4Hz,1H),2.61(dt,J=18.4,9.3Hz,1H),2.89-3.20(m,2H),3.34(s,2H),3.54(dd,J=13.8,7.6Hz,1H),4.81(d,J=7.6Hz,1H),4.99(q,J=11.1Hz,3H),7.27-7.55(m,5H).
步骤6:化合物6的合成
向1L圆底烧瓶中加入中间产物5(45g,75mmol,1.00当量)的四氢呋喃(500mL)溶液,并加入Pd/C(10g,22.2%/m%)。将所得溶液在室温下在H 2(1atm)下搅拌2小时。反应完毕后,滤出固体。将得到的滤液真空浓缩,得到40g粗产物6,为白色固体。m/z(ES+),[M+H] +=510;HPLC tR=1.201min.
步骤7:化合物7的合成
向500mL圆底烧瓶中加入中间产物6(45g,75mmol,1.00当量)在DMF(200mL)中的溶液,然后分批加入SO 3·Py(60g,375mmol,5.00当量)。将得到的溶液在室温下搅拌20小时。将所得混合物真空浓缩。将粗产物溶于400mL NaH 2PO 4饱和溶液中,然后加入30g四丁基硫酸氢铵。用乙酸乙酯(2×500Ml)萃取所得溶液。合并有机层,用Na 2SO 4干燥,真空浓缩,得到60g粗产物7,为浅黄色固体。m/z(ES+),[M+H] +=590;HPLC tR=1.702min.1H NMR(甲醇-d4,300MHz):δ(ppm)0.06(s,3H),0.16(s,3H),0.28-0.60(m,2H),0.69-0.80(m,2H),0.91(s,9H),1.39(s,8H),1.87(td,J=7.7,7.2,3.9Hz,2H),2.82(s,3H),3.01(d,J=2.6Hz,1H),3.17(s,2H),3.62-3.83(m,1H),4.80(d,J=7.6Hz,1H),5.02(dd,J=7.5,6.0Hz,1H)
步骤8:化合物8的合成
向500mL圆底烧瓶中放入中间产物7(60g,101mmol,1.00当量)在四氢呋喃(300Ml)中的溶液,然后逐滴加入3HF·Et 3N(80mL,10.00当量)。将得到的溶液在室温下搅拌20小时。将反应混合物真空浓缩。通过快速柱色谱纯化粗产物(条件如下:C18柱;流动相,乙腈在30分钟内由0%增加至100%;检测器:UV 220nm),得到40g粗产物8,为黄色油状物。m/z(ES+),[M-H] +=474;HPLC tR=1.245min.1H NMR(甲醇-d4,300MHz):δ(ppm)0.41(t,J=6.3Hz,1H),0.50(dd,J=5.7,2.9,1.7Hz,1H),0.84(td,J=10.1,9.5,3.9Hz,2H),1.04(d,J=7.4Hz,10H),1.66(d,J=4.9Hz,2H),1.82(d,J=15.4Hz,2H),2.66(dd,J=15.4,7.7Hz,1H),3.17-3.25(m,3H),3.48(d,J=3.5Hz,1H),3.53(d,J=6.2Hz,2H),4.96(t,J=6.1Hz,1H).
步骤9:化合物JJJ的合成
向500mL圆底烧瓶中放入产物8(40g,84mmol,1.00当量)在二氯甲烷(200mL)中的溶液,然后在0℃下滴加三氟乙酸(TFA)(60mL)。将所得溶液在0℃下搅拌120分钟。将得到的混合物在0℃下真空浓 缩。加入乙醚使产物悬浮为固体。通过离心沉淀收集产物。粗产物通过制备型HPLC纯化(条件如下:流动相A:水(无缓冲液),流动相B:乙腈;流速:40mL/min;梯度:7分钟内由0%B至15%B;检测器:254,220nm;t R=3.4分钟),得到(1R,4S)-4-(5-((S)-2-氨基-1-羟乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸(5g,4步17.7%),为白色固体。m/z(ES+),[M+H] +=376;HPLC tR=0.838min.1H NMR(D 2O,400MHz):δ(ppm)0.38(p,J=5.0Hz,1H),0.42-0.55(m,1H),0.58-0.74(m,2H),1.77(dd,J=16.1,4.3Hz,1H),2.29-2.66(m,1H),2.96-3.21(m,1H),3.28(dt,J=12.4,4.3Hz,1H),3.43(qd,J=8.3,7.4,4.3Hz,2H),3.47-3.66(m,1H),4.91(dd,J=7.9,4.2Hz,1H),5.18-5.37(m,1H).
实施例63化合物KKK的合成
Figure PCTCN2019073000-appb-000118
步骤1:化合物1的合成
向250mL圆底烧瓶中加入(1R,4S)-4-(5-((S)-2-氨基-1-((叔丁基二甲基甲硅烷基)氧基)乙基)-1,3,4-噁二唑-2-基)-7-(苄氧基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-6-酮(4.0g,7.8mmol,1.0当量)的四氢呋喃(80mL)溶液,DIEA(4.0g,31.2mmol,4.0当量),碘甲烷(CH 3I)(1.7g, 11.7mmol,1.5当量)。将所得溶液在室温下搅拌2小时。将得到的混合物真空浓缩,加入乙酸乙酯(100mL)。将所得混合物用水(2×80mL),盐水(2×80mL)洗涤,用硫酸钠干燥并真空浓缩,得到4.0g化合物1粗品,为黄色油状物。m/z(ES+),[M+H] +=514;HPLC tR=1.469min.
步骤2:化合物2的合成
向500mL圆底烧瓶中加入(1R,4S)-7-(苄氧基)-4-(5-((S)-1-((叔丁基二甲基甲硅烷基)氧基)-2-(甲基氨基)乙基)-1,3,4-噁二唑-2-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-6-酮(4.0g,来自步骤1的粗品)在二氯甲烷(80mL)中的溶液,Boc 2O(7.0g,32mmol)和三乙胺(3.2g,32mmol)。将所得溶液在室温下搅拌2小时。将反应混合物真空浓缩。通过快速C-18柱色谱纯化粗产物(条件如下:流动相,乙腈在30分钟内由0%增加至100%;检测器:UV 220nm),得到叔丁基((2S)-2-(5-((1R,4S)-7-(苄氧基)-6-氧代-5,7-二氮杂螺环[双环][3.2.1]辛烷-2,1′-环丙烷基-4-基)-1,3,4-噁二唑-2-基)-2-((叔丁基二甲基甲硅烷基)氧基)乙基)(甲基)氨基甲酸酯(300mg),为黄色油状物。m/z(ES+),[M+Na] +=636;HPLC tR=3.092min.
步骤3:化合物3的合成
向25mL圆底烧瓶中加入((2S)-2-(5-((1R,4S)-7-(苄氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)-1,3,4-噁二唑-2-基)-2-((叔丁基二甲基硅烷基)氧基)乙基)(甲基)氨基甲酸叔丁酯(300mg,0.489mmol,1.0当量)在四氢呋喃(10mL)中的溶液和Pd/C(0.1g)。将所得溶液在室温下在H 2(大气压)下搅拌2小时,滤出固体,将滤液真空浓缩,得到200mg((2S)-2-((叔丁基二甲基甲硅烷基)氧基)-2-(5-((1R,4S)-7-羟基-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙基]-4-基)-1,3,4-噁二唑-2-基)乙基)(甲基)氨基甲酸叔丁基酯粗品,为白色固体。m/z(ES+),[M+Na] +=546;HPLC tR=3.026min.
步骤4:化合物4的合成
向25mL圆底烧瓶中加入((2S)-2-((叔丁基二甲基甲硅烷基)氧基)-2-(5-((1R,4S)-7-羟基-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)-1,3,4-噁二唑-2-基)乙基)(甲基)氨基甲酸叔丁基酯(200mg,0.38mmol,1.0当量)的DMF(5mL)溶液,然后分批加入SO 3·Py(112mg,1.9mmol,5.00当量)。将得到的溶液在室温下搅拌20小时。将所得混合物真空浓缩。向粗产物中加入10mL NaH 2PO 4饱和溶液。然后加入150mg硫酸氢四丁基铵。然后用2x10mL乙酸乙酯萃取溶液。合并有机层,用硫酸钠干燥,真空浓缩,得到100mg(1R,4S)-4-(5-((S)-2,2,3,3,7,10,10-七甲基-8-氧代-4,9-二氧杂-7-氮杂-3-硅杂十一烷-5-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[二环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸粗品,为浅黄色固体。m/z(ES+),[M-H] +=602;HPLC tR=2.027min.
步骤5:化合物5的合成
向25mL圆底烧瓶中加入(1R,4S)-4-(5-((S)-2,2,3,3,7,10,10-七甲基-8-氧代-4,9-二氧杂-7-氮杂-3-硅杂十一烷-5-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[二环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸(100mg,0.165mmol,1.0当量)的THF(2mL)溶液,然后加入3HF·Et 3N(0.5mL)。将得到的溶液在室温下搅拌20小时。将反应混合物真空浓缩。通过快速C-18柱纯化粗产物(条件如下:流动相,乙腈在30分钟内由0%增加至100%;检测器:UV 220nm),得到50mg(1R,4S)-4-(5-((S)-2-((叔丁氧基羰基)(甲基)氨基)-1-羟乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸粗品,为黄色油状物。m/z(ES+),[M-H] +=474;HPLC tR=1.261min.
步骤6:化合物KKK的合成
向7mL圆底烧瓶中加入化合物6(50mg,0.10mmol,1.00当量)在二氯甲烷(1mL)中的溶液,然后在0℃下滴加TFA(0.25mL,10.00当量)。将所得溶液在0℃下搅拌120分钟。将得到的混合物在0℃下真空浓 缩。加入乙醚以使产物悬浮为固体。通过离心沉淀收集产物。粗产物通过制备型HPLC纯化(条件如下:流动相A:水(0.1%甲酸),流动相B:乙腈;流速:60mL/min;梯度:7分钟内由20%B至31%B;检测器:254/220nm;tR=5.43分钟),得到(1R,4S)-4-(5-((S)-1-羟基-2-(甲基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸(20.6mg,四步10.84%),为白色固体。m/z(ES+),[M-H] +=390;HPLC tR=0.746min.1H NMR(D 2O,400MHz):δ(ppm)0.35-0.66(m,2H),0.68-0.85(m,2H),1.86(d,J=16.0Hz,1H),2.64(ddd,J=16.1,7.8,1.4Hz,1H),2.85(s,3H),3.19(d,J=12.2Hz,1H),3.36(dd,J=12.2,3.8Hz,1H),3.52(d,J=3.7Hz,1H),3.59-3.71(m,2H),4.99(d,J=7.6Hz,1H),5.43(dd,J=8.2,4.3Hz,1H).
实施例64化合物LLL的合成
Figure PCTCN2019073000-appb-000119
步骤1:化合物1的合成
向250mL圆底烧瓶中加入((2S)-2-(5-((1R,4S)-7-(苄氧基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)-1,3,4-噁二唑-2-基)-2-((叔丁基二甲基甲硅烷基)氧基)乙基)氨基甲酸叔丁酯(4.0g,8.3mmol,1.0当量)的二氯甲烷(40mL)溶液,加入三氟乙酸(20mL, 18.0当量)。将所得溶液在室温下搅拌2小时。将所得混合物真空浓缩,加入二氯甲烷(50mL),用碳酸氢钠(2×30mL),盐水(30mL)洗涤,用硫酸钠干燥并真空浓缩,得到4.0g(1R,4S)-4-(5-((S)-2-氨基-1-((叔丁基二甲基甲硅烷基)氧基)乙基)-1,3,4-噁二唑-2-基-7-(苄氧基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-6-酮粗品,为黄色固体。m/z(ES+),[M+H] +=500;HPLC tR=1.061min.
步骤2:化合物2的合成
向250mL圆底烧瓶中加入(1R,4S)-4-(5-((S)-2-氨基-1-((叔丁基二甲基甲硅烷基)氧基)乙基)-1,3,4-噁二唑-2-基)-7-(苄氧基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-6-酮(2.0g,3.9mmol,1.0当量)在四氢呋喃(40mL)中的溶液,加入DIEA(2.0g,15.6mmol,4.0当量),CH 3I(1.14g,7.8mmol,2.0当量)。将所得溶液在室温下搅拌2小时。将得到的混合物真空浓缩,加入乙酸乙酯(100mL)。将所得混合物用水(2×40mL),盐水(2×40mL)洗涤。将有机层用硫酸钠干燥并真空浓缩。通过TLC(乙酸乙酯)纯化粗产物,得到(1R,4S)-7-(苄氧基)-4-(5-((S)-1-((叔丁基二甲基甲硅烷基)氧基)-2-(二甲基氨基)乙基)-1,3,4-噁二唑-2-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-6-酮(380mg,两步21.6%),为黄色油状物。m/z(ES+),[M+H] +=528;HPLC tR=1.561min.
步骤3:化合物3的合成
向25mL圆底烧瓶中加入(1R,4S)-7-(苄氧基)-4-(5-((S)-1-((叔丁基二甲基甲硅烷基)氧基)-2-(二甲基氨基)乙基)-1,3,4-噁二唑-2-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-6-酮(380mg,0.489mmol,1当量)在四氢呋喃(10mL)中的溶液和Pd/C(0.1g)。将所得溶液在室温下在H 2(大气压)下搅拌2小时。滤出固体,将滤液真空浓缩,得到330mg(1R,4S)-4-(5-((S)-1-((叔丁基二甲基甲硅烷基)氧基)-2-(二甲基氨基)乙基)-1,3,4-噁二唑-2-基)-7-羟基-5,7-二氮杂螺环[双环[3.2.1]辛烷 -2,1′-环丙烷]-6-酮粗品,为白色固体。m/z(ES+),[M+Na] +=438;HPLC tR=0.821min.
步骤4:化合物4的合成
向25mL圆底烧瓶中加入(1R,4S)-4-(5-((S)-1-((叔丁基二甲基甲硅烷基)氧基)-2-(二甲基氨基)乙基)-1,3,4-噁二唑-2-基)-7-羟基-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-6-酮(330mg,0.76mmol,1.0当量)在DMF(10mL)中的溶液,然后分批加入SO 3·Py(221mg,3.8mmol,5.00当量)。将得到的溶液在室温下搅拌20小时。将所得混合物真空浓缩。向粗产物中加入10mL NaH 2PO 4饱和溶液,然后加入250mg四丁基硫酸氢钠,然后用2x15mL乙酸乙酯萃取溶液。合并有机层,用硫酸钠干燥,真空浓缩,得到100mg(1R,4S)-4-(5-((S)-1-((叔丁基二甲基甲硅烷基)氧基)-2-(二甲基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸粗品,为浅黄色固体。m/z(ES+),[M+H] +=518;HPLC tR=0.872min.
步骤5:化合物LLL的合成
向25mL圆底烧瓶中加入(1R,4S)-4-(5-((S)-1-((叔丁基二甲基甲硅烷基)氧基)-2-(二甲基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸(100mg,0.165mmol,1.0当量)在四氢呋喃(2mL)中的溶液,加入3HF·Et 3N(0.5mL)。将得到的溶液在室温下搅拌20小时。将反应混合物真空浓缩。粗产物通过制备型HPLC纯化,条件如下:流动相A:水(0.1%甲酸),流动相B:乙腈;流速:25mL/min;梯度:7分钟内由5%B至14%B;tR=5.28,7.24分钟,得到(1R,4S)-4-(5-((S)-2-(二甲基氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸(14.8mg,三步19%),为黄色油状物。m/z(ES+),[M+H] +=404;HPLC tR=1.448min.1H NMR(D 2O,400MHz):δ(ppm)0.32-0.65(m,2H),0.62-0.87(m,2H),1.85(d,J=16.0Hz,1H),2.43-2.77(m,1H), 3.04(s,6H),3.17(d,J=12.2Hz,1H),3.36(dd,J=12.2,3.8Hz,1H),3.52(d,J=3.7Hz,1H),3.63-3.90(m,2H),4.99(d,J=7.6Hz,1H),5.54(dd,J=10.0,3.9Hz,1H).
生物实验
1.材料
1.1.细菌菌株
Figure PCTCN2019073000-appb-000120
1.2.培养基
胰酶大豆琼脂(Trypticase soy agar,TSA)(BD BBL 211043)
离子校正的马-欣二氏肉汤(Cation-adjusted Mueller Hinton broth,CAMHB)(BD BBL 212322)
1.3.试剂和耗材
亚胺培南(Imipenem)(USP 1337809)
头孢他啶(Ceftazidime)(USP 1098129)
氨苄西林(Ampicillin)(USP 1033000)
3-(N-吗啉基)丙磺酸4-吗啉丙磺酸(MOPS)(Sigma M1254)
一次性平皿,100mm(VWR 25384-302)
96孔微量滴定板(Greiner 650162)
2.方法
2.1.细菌复苏
用于最小抑菌浓度(MIC)测试的细菌菌株冻存于-80℃低温冰箱,使用需要提前2天复苏。用无菌接种环刮取少许冻存的细菌菌株在TSA固体培养基平皿上划线接种,放入普通大气培养环境中35±2℃培养20-24小时。
用无菌接种环从上述培养皿中挑取5-10个形态相似的菌落,再次划线接种于合适的固体培养基平皿上。随后放入普通培养箱中35±2℃培养20-24小时。
2.2.接种细菌准备
将液体培养基从4℃冰箱取出放置室温加热。
从上述固体培养皿中挑取5-10个细菌单菌落重悬于500μl的0.9%NaCl中,用分光光度计调节OD600至0.1~0.15。
再用1.1x CAMHB将细菌稀释300倍。
2.3.试验板准备
试验用96孔板
A-H行测试化合物:最高最终测试浓度64μg/ml或32μg/ml,2倍倍比稀释。
生长对照(Growth control,GC):含有细菌接种物的1.1xCAMHB和化合物溶剂,无化合物。
无菌对照(Sterile control,SC):含有1.1xCAMHB和化合物溶剂,无化合物。
测试化合物稀释:所有测试化合物均用二甲亚砜(DMSO)溶解和稀释。转移170μl 3.2mg/ml(50倍x 64μg/ml)的化合物到稀释母板的第1列孔中(A1-H1),再转移85μl的DMSO至其他列的孔中。依次对每个化合物进行2倍倍比稀释(即从第1列的孔中吸取85μl化合物至第2列的孔中并混匀,再从第2列的孔中吸取85μl化合物到第3 列的孔中并混匀,再从第3列的孔中吸取85μl化合物到第4列的孔中并混匀,以此类推稀释至第11列的孔)。
添加物亚胺培南和头孢他啶的配制:亚胺培南和头孢他啶用10mM MOPS缓冲液溶解。转移110μl 50μg/ml(12.5倍x 4μg/ml)的亚胺培南或者110μl 100μg/ml(12.5倍x 8μg/ml)的头孢他啶到添加物板的1-11列,再转110μl MOPS缓冲液到第12列。
添加物氨苄西林的配制:氨苄西林用ddH 2O溶解。转移30μl 100μg/ml(12.5倍x 8μg/ml)的氨苄西林到添加物板的1-11列,再转30μl的ddH 2O到第12列。
从测试化合物稀释母板中各转移2μl的化合物到试验板的相应孔中,同时转移2μl的100%DMSO到无化合物孔中(GC和SC孔)。(1)从亚胺培南添加物板转移8μl的亚胺培南到试验板的相应孔中;(2)从头孢他啶添加物板转移8μl的头孢他啶到试验板的相应孔中;(3)从氨苄西林添加物板转移8μl的氨苄西林到试验板的相应孔中。
加入90μl相应的细菌接种物至试验板中(SC孔除外)。
加入90μl的1.1xCAMHB培养基至试验板的SC孔中。
体系加完后用无菌盖盖住6块试验板,放入离心机1000rpm离心30秒,再在振板机上800rpm振1分钟混匀后放入普通培养箱35±2℃培养16-20小时。
2.4.菌落计数
将接种细菌用液体培养基从10 -1稀释至10 -7(比如100μl的细菌接种物+900μl的1.1xCAMHB)。
将100μl上述细菌稀释液均匀涂布于TSA平皿中,每个稀释度2个重复。待培养基被TSA吸收10分钟后,反转平皿在培养箱中35±2℃培养24小时。
2.5.最小抑菌浓度记录及菌落数统计
打开化合物管理系统检查每块试验板的条形码和化合物排布是否正确。
将试验板置于读板设备上,调节反射镜观察记录每个孔中细菌生 长情况。同时用QCount软件对每块试验板拍照。
参照临床和实验室标准研究所指南记录每个化合物的最小抑菌浓度。
统计不同稀释度细菌接种物在TSA平皿的菌落数并计算细菌接种量。
3.结果
3.1.第一部分:本发明化合物对细菌的MIC检测
本研究参照临床和实验室标准研究所指南的微量培养基稀释法检测了本发明的化合物和抗生素(亚胺培南、头孢他啶和氨苄西林)对11株细菌的最小抑菌浓度。并且对测试化合物与亚胺培南、头孢他啶和氨苄西林联合用药的效果进行了测试。化合物在96孔板中从最高检测浓度64μg/ml进行两倍倍比稀释。当测试化合物与亚胺培南、头孢他啶和氨苄西林联合用药检测时,亚胺培南固定4μg/ml的浓度,头孢他啶固定8μg/ml的浓度,氨苄西林固定8μg/ml的浓度。试验板中的细菌接种物从TSA复苏并稀释于CAMHB中,同时在试验板中设置了生长对照(GC孔)和无菌对照(SC孔)。试验板在普通培养箱35±2℃培养16-20小时后观察并记录每个化合物组合对不同细菌的最小抑菌浓度,结果参见表1-3。
表1.与头孢他啶8μg/ml组合抑菌时测试化合物的最小浓度(μg/ml)
Figure PCTCN2019073000-appb-000121
Figure PCTCN2019073000-appb-000122
A:<8μg/ml;B:8-16μg/ml;C:16-128μg/ml
表2.与亚胺培南4μg/ml组合抑菌时测试化合物的最小浓度(μg/ml)
Figure PCTCN2019073000-appb-000123
Figure PCTCN2019073000-appb-000124
A:<8μg/ml;B:8-16μg/ml;C:16-128μg/ml
表3.与氨苄西林8μg/ml组合抑菌时测试化合物的最小浓度(μg/ml)
Figure PCTCN2019073000-appb-000125
A:<8μg/ml;B:8-16μg/ml;C:16-128μg/ml
结果表明,所有β-内酰胺酶抑制剂单独测试均无抗菌活性(MIC>64μg/ml),测试化合物与亚胺培南、头孢他啶或氨苄西林的组合均可以不同程度提高亚胺培南、头孢他啶或氨苄西林对含β-内酰胺酶的耐药菌株中的抑菌效果。

Claims (19)

  1. 一种通式(I)所述的化合物或其酯、立体异构体以及药学上可接受的盐:
    Figure PCTCN2019073000-appb-100001
    其中,W 1选自任选取代的含有O、N和/或S的五元或6元杂芳环或-C(O)-;并且,(i)当W 1选自任选取代的含有O、N和/或S的五元或6元杂芳环时,W 1任选地被C 1-C 12烷基取代,
    W 2选自:
    a.H
    b.
    Figure PCTCN2019073000-appb-100002
    其中R 1选自
    Figure PCTCN2019073000-appb-100003
    Figure PCTCN2019073000-appb-100004
    其中R 2、R 3、R 4、R 4’、R 5或R 6各自独立地选自H、C 1-C 12烷基、氨基C 1-C 12烷基、C 1-C 12烷胺基C 1-C 12烷基、
    Figure PCTCN2019073000-appb-100005
    Ra和Rb各自独立地选自H、C 1-C 12烷基、C 3-C 8环烷基、OH、-OC 1-C 12烷基、-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-SH、-SC 1-C 12烷基、-S(O)C 1-C 12烷基、-S(O 2)C 1-C 12烷基、-SO 3H;R 7和R 7’各自独立地选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-OC 1-C 12烷基、-SC 1-C 12烷基;R 8和R 8’各自独立地选自NH、-NC 1-C 12烷基、O、S;R 9和R 9’各自独立地选自-NH-、-N(C 1-C 12烷基)-、-O-、-S-;Z 1选自CR 10R 11或NR 12,R 10、R 11和R 12各自独立地选自H、NH 2
    Figure PCTCN2019073000-appb-100006
    其中R 13和R 15各自独立地选自-NH 2、-NHC 1-C 12烷基、 -N(C 1-C 12烷基) 2、-OC 1-C 12烷基、-SC 1-C 12烷基;R 14和R 16各自独立地选自NH、-NHC 1-C 12烷基、O、S;R 17选自-NH-、-N(C 1-C 12烷基)-、-O-、-S-;i、j、k、l、m、n、p、q、r和s各自独立地选自0、1、2、3、4、5或6;条件是q和r不同时为0;
    R 1’选自H、C 1-C 12烷基、C 3-C 8环烷基、OH、-OC 1-C 12烷基、-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-SH、-SC 1-C 12烷基、-S(O)C 1-C 12烷基、-S(O 2)C 1-C 12烷基、-SO 3H;
    c.
    Figure PCTCN2019073000-appb-100007
    其中Z 2选自CR 18R 19或NR 20,R 18、R 19和R 20各自独立地选自H、NH 2
    Figure PCTCN2019073000-appb-100008
    其中R 21选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-OC 1-C 12烷基、-SC 1-C 12烷基;R 22选自NH、-NHC 1-C 12烷基、O、S;t、u和w各自独立地选自1、2、3、4、5或6,条件是t和u不同时为0;
    d.
    Figure PCTCN2019073000-appb-100009
    其中Z 3和Z 4各自独立地选自-NR 25-、-O-,R 23和R 24各自独立地选自H、C 1-C 12烷基,或者R 23和R 24一起形成=O或=NH;R 25选自H、氨基C 1-C 12烷基或C 1-C 12烷胺基C 1-C 12烷基;
    (ii)当W 1选自-C(O)-时,
    W 2选自-OR 26、-NHR 27,其中R 26和R 27各自独立地选自H、C 1-C 12烷基或
    Figure PCTCN2019073000-appb-100010
    其中Z 5选自CR 28R 29或NR 30,R 28、R 29和R 30各自独立地选自H、NH 2
    Figure PCTCN2019073000-appb-100011
    其中R 31选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-OC 1-C 12烷基、 -SC 1-C 12烷基;R 32选自NH、NC 1-C 12烷基、O、S;x、y和z各自独立地选自1、2、3、4、5或6,条件是x和y不同时为0;
    W 3选自-SO 3M、-OSO 3M、-OSO 2NH 2、-OPO 3M、-OCR 33R 34CO 2M、-OCR 35R 36SO 3M、-OCR 37R 38PO 3M;其中,M选自H或药学上可接受的阳离子;R 33、R 34、R 35、R 36、R 37和R 38各自独立地选自H、C 1-C 12烷基、卤素。
  2. 根据权利要求1所述的化合物或其酯、立体异构体以及药学上可接受的盐,其中所所式(I)所示化合物为:
    Figure PCTCN2019073000-appb-100012
    其中,W 1、W 2和W 3如式(I)所定义。
  3. 根据权利要求2所述的化合物或其酯、立体异构体以及药学上可接受的盐,其中,
    W 1选自任选取代的含有O、N和/或S的五元或6元杂芳环;
    W 2选自:
    a.H
    b.
    Figure PCTCN2019073000-appb-100013
    其中R 1选自
    Figure PCTCN2019073000-appb-100014
    Figure PCTCN2019073000-appb-100015
    其中R 2、R 3、R 4、R 4’、R 5或R 6各自独立地选自H、C 1-C 12烷基、氨基C 1-C 12烷基、C 1-C 12烷胺基C 1-C 12烷基、
    Figure PCTCN2019073000-appb-100016
    Ra和Rb各自独立地选自H、C 1-C 12烷基、OH、-OC 1-C 12烷基、-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-SH、-SC 1-C 12烷基、-S(O)C 1-C 12烷基、-S(O 2)C 1-C 12烷基、-SO 3H;R 7和R 7’各自独立地选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-OC 1-C 12烷基、 -SC 1-C 12烷基;R 8和R 8’各自独立地选自NH、-NC 1-C 12烷基、O、S;R 9和R 9’各自独立地选自-NH-、-N(C 1-C 12烷基)-、-O-、-S-;Z 1选自CR 10R 11或NR 12,R 10、R 11和R 12各自独立地选自H、NH 2
    Figure PCTCN2019073000-appb-100017
    其中R 13和R 15各自独立地选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-OC 1-C 12烷基、-SC 1-C 12烷基;R 14和R 16各自独立地选自NH、-NHC 1-C 12烷基、O、S;R 17选自-NH-、-N(C 1-C 12烷基)-、-O-、-S-;i、j、k、l、m、n、p、q、r和s各自独立地选自0、1、2、3、4、5或6;条件是q和r不同时为0;
    R 1’选自H、C 1-C 12烷基、C 3-C 8环烷基、OH、-OC 1-C 12烷基、-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-SH、-SC 1-C 12烷基、-S(O)C 1-C 12烷基、-S(O 2)C 1-C 12烷基、-SO 3H;
    c.
    Figure PCTCN2019073000-appb-100018
    其中Z 2选自CR 18R 19或NR 20,R 18、R 19和R 20各自独立地选自H、NH 2
    Figure PCTCN2019073000-appb-100019
    其中R 21选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-OC 1-C 12烷基、-SC 1-C 12烷基;R 22选自NH、-NHC 1-C 12烷基、O、S;t、u和w各自独立地选自1、2、3、4、5或6,条件是t和u不同时为0;
    d.
    Figure PCTCN2019073000-appb-100020
    其中Z 3和Z 4各自独立地选自-NR 25-、-O-,R 23和R 24各自独立地选自H、C 1-C 12烷基,或者R 23和R 24一起形成=O或=NH;R 25选自H、氨基C 1-C 12烷基或C 1-C 12烷胺基C 1-C 12烷基。
  4. 根据权利要求2所述的化合物或其酯、立体异构体以及药学上可接受的盐,其中,
    W 1选自-C(O)-;
    W 2选自选自-OR 26、-NHR 27,其中R 26和R 27各自独立地选自H、C 1-C 12烷基或
    Figure PCTCN2019073000-appb-100021
    其中Z 5选自CR 28R 29或NR 30,R 28、R 29和R 30各自独立地选自H、NH 2
    Figure PCTCN2019073000-appb-100022
    其中R 31选自-NH 2、-NHC 1-C 12烷基、-N(C 1-C 12烷基) 2、-OC 1-C 12烷基、-SC 1-C 12烷基;R 32选自NH、NC 1-C 12烷基、O、S;x、y和z各自独立地选自1、2、3、4、5或6,条件是x和y不同时为0。
  5. 根据权利要求1-3中任一项所述的化合物或其酯、立体异构体以及药学上可接受的盐,其中,W 1选自:
    Figure PCTCN2019073000-appb-100023
    其中,X选自O、S或NH,Y和Z各自独立地选自CH或N,并且W 1任选地被C 1-C 12烷基取代。
  6. 根据权利要求1-3中任一项所述的化合物或其酯、立体异构体以及药学上可接受的盐,其中,W 1选自呋喃环、吡咯环、吡唑环、咪唑环、三唑环、噻吩环、噻唑环、异噻唑环、噁唑环、异噁唑环、1,2,4-噁二唑环、1,3,4-噁二唑环;吡啶环、嘧啶基、三嗪环、吡嗪环、四唑环、哒嗪环、噻二唑环;
    M选自H、钠离子、钾离子、钙离子、镁离子、NH 4 +、N(C 1-C 12烷基) 4 +
  7. 根据权利要求6所述的化合物或其药学上可接受的盐,其中W 1选自噁唑环、异噁唑环、1,2,4-噁二唑环、1,3,4-噁二唑环。
  8. 根据权利要求7所述的化合物或其药学上可接受的盐,其中W 1选自
    Figure PCTCN2019073000-appb-100024
  9. 根据权利要求1所述的化合物或其药学上可接受的盐,其选自:
    (1R,4S)-4-(1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1] 辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(胍基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-氨基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-胍基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(3-氨基丙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸钠;
    (1R,4S)-4-(5-(4-氨基丁基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-氨基-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-胍基-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((R)-2-胍基-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-2-胍基-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(1-(胍基甲基)环丙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((氮杂环丁烷-3-基氨基)甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(胍基甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((1-甲基胍基)甲基)异噁唑-3-yl)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7基硫酸;
    (1R,4S)-4-(5-(氨基甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((甲基氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-胍基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-(1-甲基胍基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-胍基-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-2-胍基-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((R)-2-胍基-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-氨基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-(甲基氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-6-氧代-4-(5-(哌啶-4-基)异噁唑-3-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-6-氧代-4-(5-(哌啶-3-基)异噁唑-3-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-6-氧代-4-(5-(吡咯烷-3-基)异噁唑-3-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-6-氧代-4-(5-(2-(哌啶-4-基氨基)乙基)异噁唑-3-基)-5,7- 二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-((1-脒基哌啶-4-基)氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((1s,3R)-3-氨基环丁基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((1r,3S)-3-氨基环丁基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((1R,3S)-3-氨基环戊基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((1R,3R)-3-氨基环戊基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(((1r,3R)-3-氨基环丁基)甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(((1s,3S)-3-氨基环丁基)甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((1-氨基环丙基)甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((1-(甲基氨基)环丙基)甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(氮杂环丁烷-3-基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-(甲基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((氮杂环丁烷-3-基氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(氮杂环丁烷-3-基甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((氮杂环丁烷-3-基氧基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(((1-脒基氮杂环丁烷-3-基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(3-氨基环丁基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((1s,3S)-3-氨基环丁基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((1r,3R)-3-氨基环丁基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-((2-胍基乙基)氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-6-氧代-4-(哌啶-4-基氨基甲酰基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-6-氧代-4-(吡咯烷-3-基氨基甲酰基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(氮杂环丁烷-3-基氨基甲酰基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-2-((2-氨基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((R)-2-亚氨基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    2-(((1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)乙酸;
    2-(((1R,4S)-4-(5-(((叔丁氧羰基)氨基)甲基)异噁唑-3-基)-6-氧代 -5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)乙酸;
    2-(((1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)-2,2-二氟乙酸;
    2-(((1R,4S)-4-(5-(((叔丁氧羰基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)-2,2-二氟乙酸;
    (1R,4S)-4-(5-((1s,3R)-3-胍基环丁基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((R)-1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-((2-氨基乙基)氨基)-1-羟基乙基)异噁唑-3-y1)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-2-((2-氨基乙基)氨基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((R)-2-((2-氨基乙基)氨基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-((2-胍基乙基)氨基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-2-((2-胍基乙基)氨基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((R)-2-((2-胍基乙基)氨基)-1-羟基乙基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)-1,3,4-噁 二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((R)-1-羟基-2-((2-(甲基氨基)乙基)氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-((2-氨基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((R)-2-((2-氨基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-((2-胍基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-2-((2-胍基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((R)-2-((2-胍基乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-(氮杂环丁烷-3-基氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(1-羟基-2-(甲基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(1-羟基-2-(吡咯烷-3-基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-(二甲基氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(羟基(吡咯烷-2-基)甲基)-1,3,4-噁二唑-2-yl)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    2-羟基-N,N,N-三甲基-2-(5-((1R,4S)-6-氧代-7-(磺氧基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)-1,3,4-噁二唑-2-基)乙烷-1-铵;
    (1R,4S)-4-(5-(1-羟基-2-(哌啶-4-基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(4-羟基哌啶-4-基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(1-脒基哌啶-4-基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-6-氧代-4-(5-(哌嗪-1-基)异噁唑-3-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-6-氧代-4-(5-(2-氧代哌嗪-1-基)异噁唑-3-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(L-脯氨酰基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-2-((2-(二甲基氨基)乙基)氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    2-(((2S)-2-羟基-2-(5-((1R,4S)-6-氧代-7-(磺氧基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)-1,3,4-噁二唑-2-基)乙基)氨基)-N,N,N-三甲基乙烷-1-铵;
    (1R,4S)-6-氧代-4-(5-(2-氧代咪唑烷-4-基)-1,3,4-噁二唑-2-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-6-氧代-4-(5-(2-氧代噁唑烷-5-基)-1,3,4-噁二唑-2-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-6-氧代-4-(5-(2-氧代噁唑烷-4-基)-1,3,4-噁二唑-2-基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-亚胺基-1-((甲基氨基)甲基)咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7- 基硫酸;
    (1R,4S)-4-(5-(1-((二甲基氨基)甲基)-2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    1-(2-亚胺基-4-(5-((1R,4S)-6-氧代-7-(磺氧基)-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-4-基)-1,3,4-噁二唑-2-基)咪唑烷-1-基)-N,N,N-三甲基甲烷铵;
    (1R,4S)-4-(5-(2-亚胺基-1-((甲基氨基)甲基)-5-氧代咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(5-羟基-2-亚胺基-1-((甲基氨基)甲基)咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(3-((甲基氨基)甲基)-2-氧代噁唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(1-(氨基甲基)-2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(5-氨基-2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(2-亚胺基-5-(甲基氨基)咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-(5-(二甲基氨基)-2-亚胺基咪唑烷-4-基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((R)-2-氨基-1-羟乙基)-1,3,4-噁二唑-2-基-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-2-氨基-1-羟乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-1-羟基-2-(甲基氨基)乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    (1R,4S)-4-(5-((S)-2-(二甲基氨基)-1-羟基乙基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基硫酸;
    2-(((1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)乙酸;
    2-(((1R,4S)-4-(5-(((叔丁氧羰基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)乙酸;
    2-(((1R,4S)-4-(5-(氨基甲基)-1,3,4-噁二唑-2-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)-2,2-二氟乙酸;
    2-(((1R,4S)-4-(5-(((叔丁氧羰基)氨基)甲基)异噁唑-3-基)-6-氧代-5,7-二氮杂螺环[双环[3.2.1]辛烷-2,1′-环丙烷]-7-基)氧基)-2,2-二氟乙酸。
  10. 根据权利要求1所述式(I)化合物的制备方法,其中,式(I)化合物中的W 1选自异噁唑环,所述方法包括:
    Figure PCTCN2019073000-appb-100025
    步骤1:将中间体1与炔烃在碱性条件下关环反应得到中间体2;
    步骤2:将中间体2选择性脱除保护基P得到中间体3;
    步骤3:将中间体3在碱性条件下与SO 3反应得到中间体4;
    步骤4:将中间体4中的保护基P’去除得到产物5;
    其中,P为羟基保护基,P’为羟基或氨基保护基,W 2为如式(I)所定义。
  11. 根据权利要求1所述式(I)化合物的制备方法,其中,式(I)化合物中的W 1选自1,3,4-噁二唑环,所述方法包括:
    Figure PCTCN2019073000-appb-100026
    步骤1:将中间体1与酰肼在碱性条件下进行缩合反应得到中间体2;
    步骤2:将中间体2通过关环反应得到中间体3;
    步骤3:将中间体3选择性脱除保护基P得到中间体4;
    步骤4:将中间体4在碱性条件下与SO 3反应得到中间体5;
    步骤5:将中间体5中的保护基P’去除得到产物6;
    其中,P为羟基保护基,P’为羟基或氨基保护基,W 2为如式(I)所定义。
  12. 一种药物组合物,其包含权利要求1-9中任一项所述的化 合物以及药学上可接受的赋形剂。
  13. 根据权利要求12所述的药物组合物,其还包括一种或多种β内酰胺类抗生素。
  14. 根据权利要求13所述的药物组合物,其中所述β内酰胺类抗生素选自青霉素类抗生素、头孢菌素类抗生素、单内酰胺环类抗生素、碳青霉烯类抗生素、青霉烯类酶抑制剂。
  15. 根据权利要求1-9中任一项所述的化合物在制备用于治疗细菌感染导致的疾病中的药物中的用途。
  16. 根据权利要求15所述的用途,其中所述药物进一步包含β内酰胺类抗生素。
  17. 根据权利要求15所述的用途,其中所述细菌选自枸橼酸杆菌、弗氏柠檬酸杆菌、阴沟肠杆菌、肺炎克雷伯氏菌、大肠杆菌、普通变形杆菌、沙门氏菌、粘质沙雷氏菌、志贺氏杆菌、铜绿假单胞菌、黏膜炎莫拉菌、淋病奈瑟球菌、脑膜炎奈瑟球菌、不动杆菌、伯克氏菌、弯曲杆菌、幽门螺杆菌、霍乱弧菌、克雷伯杆菌、流感嗜血杆菌、鸟复合分支杆菌、脓肿分支杆菌、堪萨斯分支杆菌、溃疡分支杆菌、肺炎嗜衣原体、沙眼衣原体、β-溶血性链球菌、鲍氏不动杆菌、绿农假单胞菌、绿浓杆菌、脆弱拟杆菌、蜡样芽胞杆菌和嗜麦芽窄食单胞菌中的一种或多种。
  18. 根据权利要求15所述的用途,其中所述疾病选自呼吸道感染、尿道感染、中枢神经系统感染、耳部感染、胸膜肺和支气管感染、腹腔内感染、心血管感染、皮肤或软组织感染、骨与关节感染、生殖器感染、眼部感染、咽部感染、口腔感染中的一种或多种。
  19. 根据群里要求18所述的用途,其中所述疾病选自上呼吸道感染、下呼吸道感染、气管炎、支气管炎、肺炎、肺结核、咽炎、复杂性尿道感染、并发性尿道感染、非并发性尿道感染、膀胱炎、肾盂肾炎、脑炎、脑膜炎、脑脓肿、外耳炎、中耳炎、血液感染例如败血症或菌血症、心内膜炎、心肌炎、心包炎、关节炎、骨髓炎、生殖器溃疡、阴道炎、子宫颈炎、结膜炎、角膜炎、眼内炎、咽炎、 牙龈炎中的一种或多种。
PCT/CN2019/073000 2018-01-25 2019-01-24 β-内酰胺酶抑制剂及其用途 WO2019144912A1 (zh)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CN201980010129.1A CN111670187B (zh) 2018-01-25 2019-01-24 β-内酰胺酶抑制剂及其用途
EP19743504.3A EP3744722A4 (en) 2018-01-25 2019-01-24 Ss-LACTAMASE INHIBITOR AND ITS USE
JP2020540565A JP7273420B2 (ja) 2018-01-25 2019-01-24 β-ラクタマーゼ阻害剤及びその用途
KR1020207024397A KR20200113247A (ko) 2018-01-25 2019-01-24 β-락타마제 억제제 및 그 용도
CA3089150A CA3089150A1 (en) 2018-01-25 2019-01-24 .beta.-lactamase inhibitor and use thereof
AU2019211693A AU2019211693B2 (en) 2018-01-25 2019-01-24 Beta-lactamase inhibitor and use thereof
US16/998,577 US11078202B2 (en) 2018-01-25 2020-08-20 β-lactamase inhibitor and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNPCT/CN2018/074185 2018-01-25
CN2018074185 2018-01-25

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/964,416 A-371-Of-International US20210046057A1 (en) 2018-01-25 2019-01-24 Beta-lactamase inhibitor and use thereof
US16/998,577 Continuation US11078202B2 (en) 2018-01-25 2020-08-20 β-lactamase inhibitor and use thereof

Publications (1)

Publication Number Publication Date
WO2019144912A1 true WO2019144912A1 (zh) 2019-08-01

Family

ID=67394549

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/073000 WO2019144912A1 (zh) 2018-01-25 2019-01-24 β-内酰胺酶抑制剂及其用途

Country Status (8)

Country Link
US (1) US11078202B2 (zh)
EP (1) EP3744722A4 (zh)
JP (1) JP7273420B2 (zh)
KR (1) KR20200113247A (zh)
CN (1) CN111670187B (zh)
AU (1) AU2019211693B2 (zh)
CA (1) CA3089150A1 (zh)
WO (1) WO2019144912A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023088375A1 (zh) 2021-11-17 2023-05-25 苏州信诺维医药科技股份有限公司 β-内酰胺酶抑制剂中间体及制备方法
CN116730941A (zh) * 2023-06-15 2023-09-12 广东省科学院微生物研究所(广东省微生物分析检测中心) 5-苯基-1,3,4-恶二唑类化合物及其制备方法和应用

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20200113247A (ko) 2018-01-25 2020-10-06 쑤저우 시노벤트 파마슈티칼즈 씨오., 엘티디. β-락타마제 억제제 및 그 용도

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1033000A (en) 1910-12-14 1912-07-16 Joseph Leslie Forster Pneumatic piano-player.
US1098129A (en) 1910-04-05 1914-05-26 John Thomas Simpson Aeroplane.
US1337809A (en) 1917-06-22 1920-04-20 Winton Alexander Protecting means for vessels
CN101918407A (zh) * 2008-01-18 2010-12-15 默沙东公司 β-内酰胺酶抑制剂
WO2013030735A1 (en) 2011-08-30 2013-03-07 Wockhardt Limited 1,6- diazabicyclo [3,2,1] octan- 7 - one derivatives and their use in the treatment of bacterial infections
WO2013149121A1 (en) 2012-03-30 2013-10-03 Cubist Pharmaceuticals, Inc. 1,3,4-oxadiazole and 1,3,4-thiadiazole beta-lactamase inhibitors
WO2014141132A1 (en) 2013-03-14 2014-09-18 Naeja Pharmaceutical Inc. NEW HETEROCYCLIC COMPOUNDS AND THEIR USE AS ANTIBACTERIAL AGENTS AND β-LACTAMASE INHIBITORS
CN104334555A (zh) * 2012-03-30 2015-02-04 丘比斯特药物股份有限公司 异噁唑β-内酰胺酶抑制剂
WO2015110963A1 (en) 2014-01-21 2015-07-30 Wockhardt Limited 2-(1,3,4-oxadiazol-2-yl)-7-oxo-1,6-diazabicyclo[3.2.1 ]octane derivatives and their use as antibacterial agents
WO2015150890A1 (en) 2014-03-29 2015-10-08 Wockhardt Limited A process for preparation of trans-sulfuric acid mono-{2-(5-(3-amino-propyl)-[1,3,4]oxadiazol-2-yl]-7-oxo-1,6-diazabicyclo [3.2.1] oct-6-yl}ester
WO2015159265A1 (en) 2014-04-18 2015-10-22 Wockhardt Limited Pharmaceutical compositions comprising antibacterial agents
WO2015173665A1 (en) 2014-05-14 2015-11-19 Wockhardt Limited A process for preparation of trans-sulfuric acid mono-{2-[5-(3-azetidinylamino)-methyl-[1,3,4]- oxadiazol-2-yl]-7-oxo-1,6-diazabicyclo[3.2.1] oct-6-yl} ester trifluoroacetate
WO2015173663A1 (en) 2014-05-14 2015-11-19 Wockhardt Limited A process for preparation of trans-sulfuric acid mono-{2-[5-(2-methylamino-ethyl)-[1,3,4]-oxadiazol-2-yl]-7-oxo-1,6-diaza-bicyclo [3.2.1]oct-6-yl} ester
WO2017055922A1 (en) 2015-10-02 2017-04-06 Legochem Biosciences, Inc. Compositions and methods for inhibiting beta-lactamase

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20200113247A (ko) 2018-01-25 2020-10-06 쑤저우 시노벤트 파마슈티칼즈 씨오., 엘티디. β-락타마제 억제제 및 그 용도

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1098129A (en) 1910-04-05 1914-05-26 John Thomas Simpson Aeroplane.
US1033000A (en) 1910-12-14 1912-07-16 Joseph Leslie Forster Pneumatic piano-player.
US1337809A (en) 1917-06-22 1920-04-20 Winton Alexander Protecting means for vessels
CN101918407A (zh) * 2008-01-18 2010-12-15 默沙东公司 β-内酰胺酶抑制剂
WO2013030735A1 (en) 2011-08-30 2013-03-07 Wockhardt Limited 1,6- diazabicyclo [3,2,1] octan- 7 - one derivatives and their use in the treatment of bacterial infections
CN103649088A (zh) * 2011-08-30 2014-03-19 沃克哈特有限公司 1,6-二氮杂双环[3,2,1]辛-7-酮衍生物及其在治疗细菌感染中的用途
CN104334555A (zh) * 2012-03-30 2015-02-04 丘比斯特药物股份有限公司 异噁唑β-内酰胺酶抑制剂
WO2013149121A1 (en) 2012-03-30 2013-10-03 Cubist Pharmaceuticals, Inc. 1,3,4-oxadiazole and 1,3,4-thiadiazole beta-lactamase inhibitors
US20130296290A1 (en) 2012-03-30 2013-11-07 Cubist Pharmaceuticals, Inc. 1,3,4-oxadiazole and 1,3,4-thiadiazole beta-lactamase inhibitors
WO2014141132A1 (en) 2013-03-14 2014-09-18 Naeja Pharmaceutical Inc. NEW HETEROCYCLIC COMPOUNDS AND THEIR USE AS ANTIBACTERIAL AGENTS AND β-LACTAMASE INHIBITORS
WO2015110963A1 (en) 2014-01-21 2015-07-30 Wockhardt Limited 2-(1,3,4-oxadiazol-2-yl)-7-oxo-1,6-diazabicyclo[3.2.1 ]octane derivatives and their use as antibacterial agents
WO2015150890A1 (en) 2014-03-29 2015-10-08 Wockhardt Limited A process for preparation of trans-sulfuric acid mono-{2-(5-(3-amino-propyl)-[1,3,4]oxadiazol-2-yl]-7-oxo-1,6-diazabicyclo [3.2.1] oct-6-yl}ester
WO2015159265A1 (en) 2014-04-18 2015-10-22 Wockhardt Limited Pharmaceutical compositions comprising antibacterial agents
WO2015173665A1 (en) 2014-05-14 2015-11-19 Wockhardt Limited A process for preparation of trans-sulfuric acid mono-{2-[5-(3-azetidinylamino)-methyl-[1,3,4]- oxadiazol-2-yl]-7-oxo-1,6-diazabicyclo[3.2.1] oct-6-yl} ester trifluoroacetate
WO2015173663A1 (en) 2014-05-14 2015-11-19 Wockhardt Limited A process for preparation of trans-sulfuric acid mono-{2-[5-(2-methylamino-ethyl)-[1,3,4]-oxadiazol-2-yl]-7-oxo-1,6-diaza-bicyclo [3.2.1]oct-6-yl} ester
WO2017055922A1 (en) 2015-10-02 2017-04-06 Legochem Biosciences, Inc. Compositions and methods for inhibiting beta-lactamase

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GREENE, T.W.WUTS, P.G.M.: "Greene's Protective Groups in Organic Synthesis,", JOHN WILEY AND SONS
See also references of EP3744722A4

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023088375A1 (zh) 2021-11-17 2023-05-25 苏州信诺维医药科技股份有限公司 β-内酰胺酶抑制剂中间体及制备方法
CN116730941A (zh) * 2023-06-15 2023-09-12 广东省科学院微生物研究所(广东省微生物分析检测中心) 5-苯基-1,3,4-恶二唑类化合物及其制备方法和应用

Also Published As

Publication number Publication date
CA3089150A1 (en) 2019-01-24
AU2019211693B2 (en) 2023-12-21
AU2019211693A1 (en) 2020-09-03
KR20200113247A (ko) 2020-10-06
CN111670187A (zh) 2020-09-15
US20210032260A1 (en) 2021-02-04
JP7273420B2 (ja) 2023-05-15
CN111670187B (zh) 2021-04-16
JP2021511351A (ja) 2021-05-06
EP3744722A4 (en) 2021-11-03
US11078202B2 (en) 2021-08-03
EP3744722A1 (en) 2020-12-02

Similar Documents

Publication Publication Date Title
TWI773687B (zh) β內醯胺酶抑制劑化合物
US11078202B2 (en) β-lactamase inhibitor and use thereof
WO2018177302A1 (zh) 含氮杂环取代的苯并噁嗪噁唑烷酮类化合物及其制备方法和用途
TW200530255A (en) 3-pyridiniummethylcefem compound
JP2022097520A (ja) 複素環式化合物及び細菌感染症の予防又は治療におけるそれらの使用
TW201731844A (zh) 羥基烷基噻二唑衍生物
JP5662940B2 (ja) 新規な抗微生物薬
WO2015192714A1 (zh) 吡咯并咪唑环衍生物及其在医药上的应用
CN109415365B (zh) 卡巴培南化合物
WO2023011574A1 (zh) 芳香乙炔类衍生物及其制备方法和用途
US20120058989A1 (en) Antibacterial fluoroquinolone analogs
RU2162088C2 (ru) 1-метилкарбапенем или его фармакологически приемлемые соли, композиция, способ предупреждения или лечения бактериальных инфекций
WO2017189866A1 (en) Polymyxin analogs useful as antibiotic potentiators
US20060247286A1 (en) Oxazolidinone antibiotics and derivatives thereof
KR20180022917A (ko) 설핀일 또는 설폰일을 갖는 3환성 화합물
WO1996001261A1 (fr) Nouveau derive de carbapeneme
US20210046057A1 (en) Beta-lactamase inhibitor and use thereof
TW202007689A (zh) 雜環化合物及它們在預防或治療細菌感染中的應用
KR20000048687A (ko) 피리도벤족사진 유도체
WO2024067813A1 (zh) 芳香乙炔类衍生物及其制备方法和其医药上的用途
JP2004203809A (ja) 新規ビフェニル誘導体
TW201840322A (zh) 羥烷基噻二唑衍生物之n-膦醯基氧基甲基前驅藥
TW202421162A (zh) 氧基取代化合物
JPH0343272B2 (zh)

Legal Events

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

Ref document number: 19743504

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3089150

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2020540565

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20207024397

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019211693

Country of ref document: AU

Date of ref document: 20190124

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019743504

Country of ref document: EP

Effective date: 20200825