CN115448923A - Pyrimido-cyclic compounds, process for their preparation and their use - Google Patents

Pyrimido-cyclic compounds, process for their preparation and their use Download PDF

Info

Publication number
CN115448923A
CN115448923A CN202210427468.3A CN202210427468A CN115448923A CN 115448923 A CN115448923 A CN 115448923A CN 202210427468 A CN202210427468 A CN 202210427468A CN 115448923 A CN115448923 A CN 115448923A
Authority
CN
China
Prior art keywords
compound
substituted
azaspiro
decan
pyrimidin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202210427468.3A
Other languages
Chinese (zh)
Other versions
CN115448923B (en
Inventor
邹斌
张睿
付贤磊
马世超
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Blueray Biopharma Co ltd
Original Assignee
Qingyu Pharmaceutical R & D Shanghai Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Qingyu Pharmaceutical R & D Shanghai Co ltd filed Critical Qingyu Pharmaceutical R & D Shanghai Co ltd
Priority to CN202210427468.3A priority Critical patent/CN115448923B/en
Publication of CN115448923A publication Critical patent/CN115448923A/en
Application granted granted Critical
Publication of CN115448923B publication Critical patent/CN115448923B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • C07D487/04Ortho-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
    • A61K31/495Heterocyclic 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
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/96Spiro-condensed ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/08Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms
    • C07D211/18Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D211/26Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms with hydrocarbon radicals, substituted by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/70Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/73Unsubstituted amino or imino radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/36Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/20Spiro-condensed ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/02Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D223/06Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D223/12Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/36Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/78Benzo [b] furans; Hydrogenated benzo [b] furans
    • C07D307/79Benzo [b] furans; Hydrogenated benzo [b] furans with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to carbon atoms of the hetero ring
    • 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
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Abstract

The invention discloses a pyrimido-cyclic compound and a preparation method and application thereof. The pyrimido-cyclic compound has a structure shown in a formula (II), and can be used for preparing a medicament for treating diseases or symptoms related to abnormal SHP2 activity.

Description

Pyrimido-cyclic compounds, process for their preparation and their use
Technical Field
Pyrimido-cyclic compounds, pharmaceutically acceptable salts thereof, and solvates thereof are disclosed. The invention also provides a preparation method of the compounds, a composition containing the compounds and application of the compounds in preparing medicaments for treating diseases or symptoms related to the abnormal activity of SHP2.
Background
The tyrosine phosphatase SHP2 consists of two N-terminal Src homology 2 domains (N-SH 2 and C-SH 2) and a protein tyrosine phosphatase catalytic domain (PTP). In a basic state, N-SH2 can be combined with PTP to form a ring structure, so that the combination of PTP and a substrate is blocked, and the enzyme catalytic activity is inhibited; when tyrosine of the upstream receptor protein is phosphorylated, N-SH2 is combined with the tyrosine, and the PTP catalytic domain is released so as to exert phosphatase activity.
At the cellular level, SHP2 is involved in multiple tumor cell signaling pathways, such as RTK/Ras/MAPK, JAK/STAT, and PI3K/Akt, among others, through a functional role downstream of the cytoplasm of many receptor tyrosine kinases. Through the regulation of these kinases and signaling pathways, SHP2 is closely associated with many important vital cell activities, such as cell proliferation, migration, differentiation, death, cytokine regulation, tumorigenesis, etc.
At the same time, SHP2 is also involved in apoptosis receptor 1 (PD 1) mediated immune system suppression. After PD-1 of T cells is combined with PD-L1, a large amount of SHP2 can be recruited in the cells. SHP2 is capable of dephosphorylating an antigen receptor pathway protein within a T cell, thereby inhibiting activation of the T cell. Thus, inhibition of SHP2 activity could reverse immunosuppression in the tumor microenvironment.
SHP2 is an important member of the protein tyrosine phosphatase family and is associated with a variety of diseases in humans, such as Noonan Syndrome (Noonan Syndrome), leopard Syndrome (Leopard Syndrome), juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myelogenous leukemia, breast cancer, esophageal cancer, lung cancer, colon cancer, head cancer, neuroblastoma, squamous cell carcinoma of the head and neck, gastric cancer, anaplastic large-cell lymphoma, and glioblastoma, among others.
A series of patents published recently, such as WO2018/013597A1, WO2017/210134A1, WO2017/211303A1, WO 2017/216706A1, WO 2016/203406A1, WO 2016/203405A1, WO 2016/203404A1, WO2015/107495A1, WO2015/107494A1, and WO2015/107493A1, etc., indicate that SHP2 is of increasing interest as a novel druggable target. Surrounding the development of the SHP2 inhibitor, two strategies are provided for the development of an inhibitor of PTP catalytic domain of SHP2 and the development of an allosteric inhibitor of non-catalytic domain; due to the problems of selectivity and poor druggability of inhibitors of the PTP catalytic domain, more research is currently being directed towards the development of allosteric inhibitors. All of the above patents disclose allosteric inhibition, but most of them have low inhibitory activity against tumor cells, and for example, the compound SHP099 (6- (4-amino-4-methylpiperidin-1-yl) -3- (2,3-dichlorophenyl) pyrazine-2-amine) disclosed in WO2015/107493A1 has been awaited to further develop an SHP2 inhibitor having a novel structure, good biological activity and high drug potency.
Disclosure of Invention
The pyrimido-cyclic compound provided by the invention is a brand-new SHP2 inhibitor, shows good inhibitory activity on tumor cells, has good pharmacy and has wide drug development prospect. And the preparation method of the compound is simple and is beneficial to industrial production.
In a first aspect, the present invention provides a pyrimido ring compound represented by formula (I), a pharmaceutically acceptable salt thereof or a solvate thereof, or a solvate of the salt.
Figure BDA0003608939860000021
Wherein
Z 1 、Z 2 Simultaneously is C or one of them is N;
x is independently S or absent;
y is independently C or N;
n is independently 0, 1 or 2;
R 1 independently 0 to 4R 1a Substituted phenyl, 0 to 4R 1a Substituted containing 1-4 azaaryl groups, 0 to 4R 1a Substituted naphthyl, 0 to 4R 1a Substituted azanaphthalene aryl containing 1-4, 0 to 4R 1a Substituted or unsubstituted benzoheterocycles, 0 to 4R 1a Substituted or unsubstituted with 1-4 azaaromatic rings, 0 to 4R 1a Substituted containing 1-4N, NR 1b Hetero aromatic ring of hetero atom such as O or S (O) m, R 1c Substituted or unsubstituted C 1-8 Alkyl, R 1c Substituted or unsubstituted C 1-8 A haloalkyl group; it is composed ofWherein m is selected from 0, 1 and 2;
R 1a independently of one another is halogen, R 1a1 Substituted or unsubstituted C 1-4 Alkoxy radical, R 1a1 Substituted or unsubstituted C 1-4 Alkyl, trifluoromethyl, C (= O) OR 1a2 、NR 1a2 R 1a3 、NHC(=O)R 1a4 、R 1a1 Substituted or unsubstituted C 3-8 A cycloalkyl group; r is 1a1 Independently is halogen or C 1-4 An alkyl group; r 1a2 、R 1a3 Independently of one another is hydrogen, C 1-4 An alkyl group; r 1a4 Independently is C 1-4 Alkyl, substituted or unsubstituted alkenyl, amide, C 3-12 Mono-or poly-heterocyclic;
R 1b independently of each other is hydrogen, R 1a1 Substituted or unsubstituted C 1-4 An alkyl group;
R 1c independently hydrogen, -C (= O) OR 1a2 、R 1a1 Substituted or unsubstituted C 1-4 An alkyl group;
R 2a 、R 2b 、R 3a and R 3b Independently of each other is hydrogen, R 1a1 Substituted or unsubstituted C 1-4 An alkyl group;
when Y = N, R 4 Independently of each other is hydrogen, R 1a1 Substituted or unsubstituted C 1-4 An alkyl group; r is 5 Is absent;
when Y = C, R 4 、R 5 Independently of one another hydrogen, aryl, C 1-4 Alkyl radical, C 1-4 Alkoxy, -O-C 1-4 Alkyl, amino, C 1-4 Alkyl substituted amino, -O-C 1-4 Alkyl-substituted amino, or R 4 And R 5 Together with Y form 0 to 3R 4a A substituted 3-to 7-membered saturated or partially unsaturated spirocyclic ring, which ring may optionally contain 1 to 3 heteroatoms or groups independently selected from N, C (= O) and/or O;
R 4a independently of one another hydrogen, halogen, R 1a1 Substituted or unsubstituted C 1-4 Alkoxy radical, R 1a1 Substituted or unsubstituted C 1-4 Alkyl, hydroxy, amino, C 1-4 An alkylamino group.
In one preferred embodiment, the pyrimido-cyclic compound of the present invention has the structure of formula (II)
Figure BDA0003608939860000022
In another preferred embodiment, R 1 Selected from the following structures:
Figure BDA0003608939860000031
wherein o is 0, 1,2,3 or 4; ring a is a heteroaryl group containing 1 to 4N atoms; ring B is heteroaryl containing 1 to 4 heteroatoms such as N, S, O, etc.; g is independently a heteroatom or group such as C, C (= O), N, S or O; r is 1aa 、R 1ab Independently is R 1a ;R 1ac Independently is R 1c Substituted or unsubstituted C 1-8 Alkyl radical, R 1c Substituted or unsubstituted C 1-8 An alkyl halide;
in another preferred embodiment, R 2a 、R 2b 、R 3a And R 3b Independently hydrogen or methyl;
in another preferred embodiment, R is when Y = N 4 Independently hydrogen, methyl; r 5 Is absent;
in another preferred embodiment, R when Y = C 4 、R 5 Independently hydrogen, methyl, ethyl, phenyl, amino, methylamino or ethylamino; in another preferred embodiment, R when Y = C 4 And R 5 The ring formed with Y is selected from the following structures:
Figure BDA0003608939860000032
wherein p is 0, 1,2 or 3; r 4a As defined above;
in another preferred embodiment, R when Y = C 4 And R 5 The ring formed with Y is selected from the following configurations:
Figure BDA0003608939860000033
wherein, p and R 4a As defined above;
in another preferred embodiment, the compound is selected from any one of the following compounds:
Figure BDA0003608939860000034
Figure BDA0003608939860000041
Figure BDA0003608939860000051
Figure BDA0003608939860000061
Figure BDA0003608939860000071
Figure BDA0003608939860000081
Figure BDA0003608939860000091
Figure BDA0003608939860000101
Figure BDA0003608939860000111
Figure BDA0003608939860000121
in another aspect, the present invention provides an isotopically labeled compound of a pyrimido ring compound represented by formula (I), a pharmaceutically acceptable salt thereof, or a solvate thereof. The atom capable of being isotopically labeled in the compound represented by the formula (I) includes, but is not limited to, hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, chlorine, iodine and the like. Each of which can be isotopically substituted 2 H、 3 H、 11 C、 13 C、 14 C、 15 N、 18 F、 31 P、 32 P、 35 S、 36 Cl and 125 i, and the like.
In another aspect, the present invention provides a process for preparing pyrimido compounds represented by formula (I) and intermediates thereof, which comprises the following steps:
the invention provides a preparation method of a formula (I), which comprises the following steps:
halogenated intermediate compounds
Figure BDA0003608939860000131
After coupling reaction with boric acid, thiol or sodium sulfide (F), formula (I) is obtained, and the reaction equation is as follows:
Figure BDA0003608939860000132
wherein, W 1 Represents halogen, preferably Br, I; z 1 、Z 2 、X、Y、n、R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined above.
In one preferred embodiment, the preparation method of formula (II) comprises the following steps:
the halogenated intermediate compound A and boric acid, mercaptan or sodium (F) are subjected to coupling reaction to obtain a compound shown in a formula (II), wherein the reaction equation is as follows:
Figure BDA0003608939860000133
wherein, W 1 Represents halogen, preferably Br, I; x, Y, n, R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined above.
The present invention also provides a process for preparing a compound A,
Figure BDA0003608939860000134
wherein, W 1 Represents halogen, preferably Br, I; r is 2a 、R 2b 、R 3a 、R 3b 、R 4 、R 5 Y and n are as defined above.
The invention also provides a preparation method of the compound A, which comprises the following steps:
the halogenated intermediate E is substituted by the intermediate amine C under the alkaline condition to obtain an intermediate compound A, and the reaction equation is as follows:
Figure BDA0003608939860000135
wherein, W 1 Represents halogen, preferably Br, I; w is a group of 2 Represents halogen, preferably Cl, br, I; y, n, R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined above.
The present invention also provides a compound of formula C-1,
Figure BDA0003608939860000141
wherein U is independently C or O; q is selected from 0, 1 or 2; pg is selected from protecting groups Boc, ac, S (= O) t Bu;n、R 2a 、R 2b 、R 3a 、R 3b And R 4a Is as defined above.
The invention also provides a preparation method of the compound C-1, which comprises the following steps:
carrying out reductive amination on the spirocyclic ketone compound C-1a to obtain an intermediate C-1b; c-1b is selectively deprotected to obtain C-1, the reaction equation is as follows:
Figure BDA0003608939860000142
wherein Pg1 is selected from protecting groups Boc, benzoyl and benzyl; pg, U, q, n, R 2a 、R 2b 、R 3a 、R 3b And R 4a Is as defined above.
The present invention also provides a compound of formula C-2,
Figure BDA0003608939860000143
wherein R is 6 Independently is C 1-8 An alkyl group, a substituted or unsubstituted aryl group, a substituted or unsubstituted alkenyl group; u, q, pg, n, R 2a 、R 2b 、R 3a 、R 3b And R 4a Is as defined above.
The invention also provides a preparation method of the compound C-2, which comprises the following steps:
spirocyclic ketone compounds C-1a and R 6 The substituted nucleophilic reagent is added to obtain a hydroxyl compound C-2a; converting the compound C-2a into an amino compound C-2b, and then selectively removing a protecting group Pg1 to obtain C-2, wherein the reaction equation is as follows:
Figure BDA0003608939860000144
wherein R is 6 、U、q、Pg1、Pg、n、R 2a 、R 2b 、R 3a 、R 3b And R 4a Is as defined above.
The present invention also provides a compound of formula C-3,
Figure BDA0003608939860000151
wherein R is 6 、Pg、n、R 2a 、R 2b 、R 3a And R 3b Is as defined above.
The invention also provides a preparation method of the compound C-3, which comprises the following steps:
ester group ortho-dehydrogenation of Compound C-3a with R 6 The substituted electrophilic reagent is substituted to obtain a compound C-3b; hydrolyzing the ester group with the compound C-3b to obtain acid C-3C; rearranging the acid C-3C to obtain amine C-3d, and then selectively removing the protecting group Pg1 to obtain C-3, wherein the reaction equation is as follows:
Figure BDA0003608939860000152
wherein R is 6 、Pg1、Pg、n、R 2a 、R 2b 、R 3a And R 3b Is as defined above.
The present invention also provides a compound of formula C-4,
Figure BDA0003608939860000153
wherein R is 6 、Pg、n、R 2a 、R 2b 、R 3a And R 3b Is as defined above.
The invention also provides a preparation method of the compound C-4, which comprises the following steps:
reducing cyano compound C-4a and protecting amino to obtain intermediate C-4b, then selectively removing protecting group Pg1 to obtain C-4, wherein the reaction equation is as follows:
Figure BDA0003608939860000154
wherein Pg1, pg, R 6 、n、R 2a 、R 2b 、R 3a And R 3b Is as defined above.
The present invention also provides a compound of formula (I),
Figure BDA0003608939860000161
wherein, W 1 Represents halogen, preferably Br, I; w 2 Represents halogen, preferably Cl, br, I;
the invention also provides a preparation method of the compound E, which comprises the following steps:
halogenating the hydroxyl intermediate B-3 to obtain a double-halogen substituted compound E, wherein the reaction equation is as follows:
Figure BDA0003608939860000162
wherein, W 1 Represents halogen, preferably Br, I; w 2 Represents halogen, preferably Cl, br, I;
the invention also provides compounds F-1, F-2,
Figure BDA0003608939860000163
wherein, V is independently C or N; r 1a Is as defined above.
The invention also provides a preparation method of the compound F-1, which comprises the following steps:
halogenated compound F-1a and methyl mercaptopropionate are subjected to catalytic coupling to obtain intermediate F-1b, and then corresponding sodium sulfide compound F-1 is obtained under alkaline condition;
the invention also provides a preparation method of the compound F-2, which comprises the following steps:
the halogenated compound F-1a is condensed with mercaptan to obtain an intermediate F-2b, and then the thiophenol compound F-2 is obtained under the acidic condition.
The reaction equations for preparing F-1 and F-2 are as follows:
Figure BDA0003608939860000164
wherein, W 3 Is halogen, preferably Br, I; v, R 1a Is as defined above.
The present invention also provides another process for preparing a pyrimido-cyclic compound represented by formula (I), comprising the steps of:
intermediates
Figure BDA0003608939860000171
Substitution with amine C gives formula (I), the reaction equation is as follows:
Figure BDA0003608939860000172
wherein, W 2 Represents halogen, preferably Cl, br, I; z is a linear or branched member 1 、Z 2 、X、Y、n、R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined above.
In one preferred embodiment, the preparation method of formula (II) comprises the steps of:
substitution of the halogenated intermediate compound B with an amine C gives the formula (II) as follows:
Figure BDA0003608939860000173
wherein, W 2 Represents halogen, preferably Cl, br, I; x, Y, n, R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined above.
The present invention also provides a compound B which is,
Figure BDA0003608939860000174
wherein, W 2 Represents halogen, preferably Cl, br, I; r is 1 And X is as defined above;
the invention also provides a preparation method of the compound B, which comprises the following steps:
substituting dichloro pyrimidine compound B-1 with amine to obtain intermediate B-2; condensing, cyclizing and hydrolyzing the intermediate B-2 under the condition of strong acid to obtain a halogenated intermediate B-3; halogenated intermediate B-3 is subjected to catalytic coupling to obtain intermediate B-4, and then converted into intermediate B, wherein the reaction equation is as follows:
Figure BDA0003608939860000175
wherein W 1 Represents halogen, preferably Br, I; w is a group of 2 Represents halogen, preferably Cl, br, I; r 1 And X is as defined above.
In another preferred embodiment, the preparation method of the compound II-A comprises the following steps:
coupling the sodium sulfur intermediate compound D with a halide to obtain a compound of formula (II-A), wherein the reaction equation is as follows:
Figure BDA0003608939860000181
wherein, Y, n, R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined above.
The present invention also provides a compound D which is,
Figure BDA0003608939860000182
wherein, Y, n, R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined above.
The present invention also provides a process for the preparation of compound D comprising the steps of:
the intermediate compound A and methyl mercaptopropionate obtain an intermediate D-1 under the condition of catalytic coupling, and then obtain a corresponding sodium sulfide compound D under the alkaline condition, wherein the reaction equation is as follows:
Figure BDA0003608939860000183
wherein, W 1 Represents halogen, preferably Br, I; y, n, R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined above.
In another preferred embodiment, the preparation method of the compound II-B comprises the following steps:
removing the amino protecting group of the intermediate II-B1 under acidic or basic conditions to obtain a compound II-B, wherein the reaction equation is as follows:
Figure BDA0003608939860000191
wherein Pg is selected from protecting groups Boc, ac, S (= O) t Bu;R 4Pg 、R 5Pg Together with the linking carbon, is selected from the following structures:
Figure BDA0003608939860000192
R 4 、R 5 together with a linking carbon, is selected from the following structures:
Figure BDA0003608939860000193
X、n、R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 、R 5 and R 4a As defined above; p is 0, 1,2 or 3.
In another preferred embodiment, the process for the preparation of compounds II-C comprises the following steps:
aminoacylation of intermediate II-C1 yields compound II-C, the reaction equation being as follows:
Figure BDA0003608939860000194
wherein, X, Y, n, R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 、R 5 、R 1a And R 1a4 Is as defined above.
The solvent involved in the present invention is selected from: dichloromethane, chloroform, 1, 2-dichloroethane, dioxane, DMF, acetonitrile, DMSO, NMP, THF or combinations thereof.
The base to which the present invention relates includes organic bases and inorganic bases.
The organic base to which the present invention relates is preferably: TEA, DIPEA, or a combination thereof.
The inorganic base according to the present invention is preferably: sodium hydride, potassium carbonate, sodium carbonate, cesium carbonate, potassium tert-butoxide, sodium tert-butoxide, liHMDS, LDA, butyllithium or combinations thereof.
Isotopically labeled compounds of pyrimido ring compounds of formula (I) described in this invention can be prepared by analogous synthetic procedures to those for unlabeled compounds, except that the unlabeled starting materials and/or reagents are replaced with isotopically labeled starting materials and/or reagents.
In another aspect, the present invention also provides a pharmaceutical composition comprising a pyrimido-cyclic compound of formula (I), a pharmaceutically acceptable salt thereof or a solvate thereof, or an isotopically labeled compound of a pyrimido-cyclic compound of formula (I), a pharmaceutically acceptable salt thereof or a solvate thereof, and a pharmaceutically acceptable excipient. The pharmaceutically acceptable excipients are preferably selected from diluents, absorbents, wetting agents, binders, disintegrants, lubricants.
In another aspect, the invention also provides the use of the pyrimido-cyclic compound shown in formula (I), the pharmaceutically acceptable salt thereof or the solvate thereof for preparing a medicament for treating diseases or disorders related to abnormal SHP2 activity. Preferably, the disease or disorder includes, but is not limited to, noonan syndrome, leopard syndrome, juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myelogenous leukemia, breast cancer, esophageal cancer, lung cancer, colon cancer, head cancer, neuroblastoma, squamous cell carcinoma of the head and neck, gastric cancer, anaplastic large-cell lymphoma or glioblastoma.
In another aspect, the present invention provides a pharmaceutical preparation comprising the pyrimido-cyclic compound of formula (I), a pharmaceutically acceptable salt thereof or a solvate thereof, which can be administered in a suitable manner, such as in the form of a suspension, syrup, emulsion, solution, etc., in the form of tablets, capsules (e.g., sustained release or timed release capsules), pills, powders, granules (e.g., small particles), elixirs, tinctures, suspensions (e.g., nanosuspensions, microsuspensions) and spray-dried dispersions, and can be administered orally, sublingually, by injection including subcutaneous injection, intravenous injection, intramuscular injection, intrasternal injection, infusion, etc., nasally (e.g., nasal inhalation), topically (e.g., creams and ointments), rectally (e.g., suppositories), etc. The compounds disclosed herein may be administered alone or in combination with a suitable pharmaceutical carrier.
In another aspect, the invention provides a pharmaceutical formulation as described in the previous aspect formulated in an appropriate dosage to facilitate and control the amount of drug administered. The dosage regimen of the compounds disclosed herein will vary with such factors as the pharmacodynamics and mode of administration, the subject, sex, age, health, weight, condition, other concurrent conditions, frequency of administration, liver and kidney function, and the effect desired, etc. The compounds disclosed herein may be administered in a single dose per day, or may be administered in a total dose divided into multiple doses (e.g., two to four times per day).
In another aspect, the present invention also provides a pyrimido compound represented by formula (I), a pharmaceutically acceptable salt thereof, or a solvate thereof, for use in combination with another drug selected from the group consisting of: anticancer drugs, tumor immunity drugs, antiallergic drugs, antiemetics, analgesics, cytoprotective drugs, and the like, which have a better effect when combined together. It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
The invention has the following advantages:
1. the pyrimido-cyclic compound disclosed by the invention is a novel allosteric inhibitor, and can be combined with a non-catalytic region of SHP2 to lock a basic state with weak activity of SHP2, so that the aim of inhibiting the activity of the SHP2 is fulfilled. The pyrimido-cyclic compound disclosed by the invention overcomes the defects of poor general selectivity and druggability and the like of a PTP catalytic domain inhibitor, shows good biological activity and druggability, and has a good drug development prospect.
2. The present invention showed superior activity to that of SHP099 (6- (4-amino-4-methylpiperidin-1-yl) -3- (2, 3-dichlorophenyl) pyrazine-2-amine) disclosed in WO2015/107493A1 and the literature (Nature 2016,535, 148-152) in the evaluation systems such as SHP2 enzyme activity inhibition assay, phosphoprotein kinase (p-ERK) cell assay, MOLM-13 cell proliferation assay and the like under the same conditions.
Description of the terms
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. As used herein, the term "about" when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value. For example, as used herein, the expression "about 100" includes 99 and 101 and all values in between (e.g., 99.1, 99.2, 99.3, 99.4, etc.).
As used herein, the term "comprising" or "includes" can be open, semi-closed, and closed. In other words, the term also includes "consisting essentially of or" consisting of 823030A ".
Radical definition
Can be found in the literature (including Carey a)Definition of standardized chemical terms is found in nd Sundberg, "ADVANCED ORGANIC CHEMISTRY 4TH ED." vols. A (2000) and B (2001), plenum Press, new York). Unless otherwise indicated, conventional methods within the skill of the art are employed, such as mass spectrometry, NMR, IR and UV/VIS spectroscopy, and pharmacological methods. Unless a specific definition is set forth, the terminology used herein in the pertinent description of analytical chemistry, organic synthetic chemistry, and pharmaceutical and medicinal chemistry is known in the art. Standard techniques can be used in chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients. For example, the reaction and purification can be carried out using the instructions of the kit from the manufacturer, or according to the methods known in the art or the instructions of the present invention. The techniques and methods described above can generally be practiced according to conventional methods well known in the art, as described in various general and more specific documents referred to and discussed in this specification. In the present specification, groups and substituents thereof may be selected by one skilled in the art to provide stable moieties and compounds. When a substituent is described by a general formula written from left to right, the substituent also includes chemically equivalent substituents obtained when the formula is written from right to left. For example, -CH 2 O-is equivalent to OCH 2 -。
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including but not limited to patents, patent applications, articles, books, operating manuals, and treatises, are hereby incorporated by reference in their entirety.
Certain chemical groups defined herein are preceded by a shorthand notation to indicate the total number of carbon atoms present in the group. E.g. C 1-6 Alkyl refers to an alkyl group as defined below having a total of 1 to 6 carbon atoms. The total number of carbon atoms in the shorthand notation does not include carbons that may be present in a substituent of the group.
In addition to the foregoing, when used in the specification and claims of this application, the following terms take the meanings indicated below, unless otherwise specifically indicated.
In the present application, the term "halogen" refers to fluorine, chlorine, bromine or iodine.
"hydroxy" means an-OH group. Alkoxy "refers to an alkyl group as defined below substituted with a hydroxyl (-OH) group.
"carbonyl" means a-C (= O) -group. "cyano" means-CN.
"amino" refers to-NH 2.
"substituted amino" refers to amino substituted with one or two alkyl, alkylcarbonyl, aralkyl, heteroaralkyl groups as defined below, for example, monoalkylamino, dialkylamino, alkylamido, aralkylamino, heteroaralkylamino.
"carboxyl" means-COOH.
In this application, the term "alkyl" as a group or as part of another group (e.g., as used in halo-substituted alkyl and the like groups) refers to a fully saturated straight or branched hydrocarbon chain radical consisting only of carbon and hydrogen atoms, having, for example, 1 to 12 (preferably 1 to 8, more preferably 1 to 6) carbon atoms, and attached to the remainder of the molecule by a single bond, including, for example, but not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2-dimethylpropyl, n-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl, decyl and the like. For the purposes of the present invention, the term "alkyl" refers to alkyl groups containing from 1 to 6 carbon atoms.
In the present application, the term "alkenyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting of only carbon atoms and hydrogen atoms, containing at least one double bond, having, for example, 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms, and attached to the rest of the molecule by a single bond, such as, but not limited to, vinyl, propenyl, allyl, but-1-enyl, but-2-enyl, pent-1, 4-dienyl, and the like.
In the present application, the term "cycloalkyl" as a group or as part of another group means a stable non-aromatic monocyclic or polycyclic hydrocarbon group consisting of only carbon and hydrogen atoms, which may include fused, bridged or spiro ring systems, having from 3 to 15 carbon atoms, preferably from 3 to 10 carbon atoms, more preferably from 3 to 8 carbon atoms, and which is saturated or unsaturated and may be attached to the rest of the molecule by a single bond via any suitable carbon atom. Unless otherwise specifically indicated in the specification, carbon atoms in the cyclic hydrocarbon group may be optionally oxidized. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, 1H-indenyl, 2, 3-indanyl, 1,2,3, 4-tetrahydro-naphthyl, 5,6,7, 8-tetrahydro-naphthyl, 8, 9-dihydro-7H-benzocyclohepten-6-yl, 6,7,8, 9-tetrahydro-5H-benzocycloheptenyl, 5,6,7,8,9, 10-hexahydro-benzocyclooctenyl, fluorenyl, bicyclo [2.2.1] heptyl, 7 dimethyl-bicyclo [2.2.1] heptyl, bicyclo [2.2.1] heptenyl, bicyclo [2.2.2] octyl, bicyclo [3.1.1] heptyl, bicyclo [3.2.1] octyl, bicyclo [2.2.2] octenyl, bicyclo [3.2.1] octenyl, adamantyl, octahydro-4-methano-octahydro-1-indenyl, and the like.
In this application, the term "heterocyclyl" as a group or part of another group means a stable 3-to 20-membered non-aromatic cyclic group consisting of 2 to 14 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, phosphorus, oxygen, and sulfur. Unless otherwise specifically indicated in the specification, a heterocyclic group may be a monocyclic, bicyclic, tricyclic or higher ring system, which may include fused ring systems, bridged ring systems or spiro ring systems; wherein the nitrogen, carbon or sulfur atom in the heterocyclic group thereof may be optionally oxidized; the nitrogen atoms may optionally be quaternized; and the heterocyclic group may be partially or fully saturated. The heterocyclic group may be attached to the rest of the molecule via a carbon atom or a heteroatom and by a single bond. In heterocyclic groups containing fused rings, one or more of the rings may be aryl or heteroaryl as defined below, provided that the point of attachment to the rest of the molecule is a non-aromatic ring atom. For the purposes of the present invention, heterocyclyl is preferably a stable 4-to 11-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 4-to 8-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heterocyclyl groups include, but are not limited to: pyrrolidinyl, morpholinyl, piperazinyl, homopiperazinyl, piperidinyl, thiomorpholinyl, 2, 7-diaza-spiro [3.5] nonan-7-yl, 2-oxa-6-aza-spiro [3.3] heptan-6-yl, 2, 5-diaza-bicyclo [2.2.1] heptan-2-yl, azetidinyl, pyranyl, tetrahydropyranyl, thiopyranyl, tetrahydrofuranyl, oxazinyl, dioxolanyl, tetrahydroisoquinolinyl, decahydroisoquinolinyl, imidazolinyl, imidazolidinyl, quinolizinyl, thiazolidinyl, isothiazolidinyl, isoxazolidinyl, indolinyl, octahydroindolyl, octahydroisoindolyl, pyrrolidinyl, pyrazolidinyl, phthalimidyl, and the like.
In this application, the term "aryl" as a group or as part of another group means a conjugated hydrocarbon ring system group having 6 to 18 carbon atoms, preferably having 6 to 10 carbon atoms. For the purposes of the present invention, an aryl group may be a monocyclic, bicyclic, tricyclic or higher polycyclic ring system and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the aryl group is attached to the remainder of the molecule by a single bond via an atom on the aromatic ring. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthryl, phenanthryl, fluorenyl, 2, 3-dihydro-1H isoindolyl, 2-benzoxazolinone, 2H-1, 4-benzoxazin-3 (4H) -on-7-yl, and the like.
In the present application, the term "arylalkyl" refers to an alkyl group as defined above substituted with an aryl group as defined above.
In this application, the term "heteroaryl" as a group or part of another group means a 5-to 16-membered conjugated ring system group having 1 to 15 carbon atoms (preferably having 1 to 10 carbon atoms) and 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur in the ring. Unless otherwise specifically indicated in the specification, a heteroaryl group may be a monocyclic, bicyclic, tricyclic or higher ring system, and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the heteroaryl group is attached to the rest of the molecule by a single bond via an atom on the aromatic ring. The nitrogen, carbon or sulfur atoms in the heteroaryl group may be optionally oxidized; the nitrogen atoms may optionally be quaternized. For the purposes of the present invention, heteroaryl is preferably a stable 5-to 12-membered aromatic group containing 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 5-to 10-membered aromatic group containing 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur or a 5-to 6-membered aromatic group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. <xnotran> , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , ,4,5,6,7- [ b ] , , [1,2,4] [4,3-b ] , [1,2,4] [4,3-a ] , [1,2,4] [4,3-c ] , [1,2,4] [4,3-a ] , [1,2-a ] , [1,2-b ] , [1,2-a ] . </xnotran>
In the present application, the term "heteroarylalkyl" refers to an alkyl group as defined above substituted with a heteroaryl group as defined above. In this application, "optionally" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "optionally substituted aryl" means that the aryl group is substituted or unsubstituted, and the description includes both substituted and unsubstituted aryl groups.
The "optionally" substituents described in the claims and the description section of the present invention are selected from alkyl, alkenyl, alkynyl, halogen, haloalkyl, haloalkenyl, haloalkynyl, cyano, nitro, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl.
The terms "moiety," "structural moiety," "chemical moiety," "group," "chemical group" as used herein refer to a specific fragment or functional group in a molecule. Chemical moieties are generally considered to be chemical entities that are embedded in or attached to a molecule.
When the compounds of the present invention contain olefinic double bonds, the compounds of the present invention are intended to include both E-and Z-geometric isomers unless otherwise specified.
"tautomer" refers to an isomer formed by the transfer of a proton from one atom of a molecule to another atom of the same molecule.
All tautomeric forms of the compounds of the invention are also intended to be included within the scope of the invention. The compounds of the present invention or pharmaceutically acceptable salts thereof may contain one or more chiral carbon atoms and may therefore give rise to enantiomers, diastereomers and other stereoisomeric forms. Each chiral carbon atom may be defined as (R) -or (S) -, based on stereochemistry. The present invention is intended to include all possible isomers, as well as racemates and optically pure forms thereof. The compounds of the invention may be prepared by selecting as starting materials or intermediates racemates, diastereomers or enantiomers. Optically active isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, e.g., crystallization and chiral chromatography.
Substituents of the invention or representing symbols, e.g. Z 1 、Z 2 、X、Y、U、V、W 1 、W 2 、W 3 、n、o、p、q、R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 、R 5 Pg1, pg, etc., and the same symbols represent the same definitions in different places, unless otherwise specified.
Conventional techniques for preparing/separating individual isomers include Chiral synthesis from optically pure precursors as appropriate, or resolution of the racemates (or racemates of salts or derivatives) using, for example, chiral high performance liquid chromatography, as described, for example, in Gerald Gubitz and Martin G.Schmid (eds.), chiral Separations, methods and Protocols, methods in Molecular Biology, vol.243,2004; m. stalup, chiral Separations, annu. Rev. Anal. Chem.3:341-63,2010; fumiss et al (EDs.), VOGEL' S ENCYCOPEDIA OF PRACTICAL ORGANIC CHEMISTRY 5.Sup.TH ED., longman Scientific and Technical Ltd., essex,1991,809-816; heller, acc, chem, res, 1990,23,128.
In the present application, the term "pharmaceutically acceptable salts" includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
"pharmaceutically acceptable acid addition salts" refers to salts with inorganic or organic acids which retain the biological effectiveness of the free base without other side effects. Inorganic acid salts include, but are not limited to, hydrochloride, hydrobromide, sulfate, nitrate, phosphate, and the like; organic acid salts include, but are not limited to, formates, acetates, 2,2 dichloroacetates, trifluoroacetates, propionates, caproates, caprylates, caprates, undecenates, glycolates, gluconates, lactates, sebacates, adipates, glutarates, malonates, oxalates, maleates, succinates, fumarates, tartrates, citrates, palmitates, stearates, oleates, cinnamates, laurates, malates, glutamates, pyroglutamates, aspartates, benzoates, methanesulfonates, benzenesulfonates, p-toluenesulfonates, alginates, ascorbates, salicylates, 4-aminosalicylates, napadisylates, and the like. These salts can be prepared by methods known in the art.
"pharmaceutically acceptable base addition salts" refers to salts with inorganic or organic bases which maintain the biological effectiveness of the free acid without other side effects. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, the following: primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purine, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. These salts can be prepared by methods known in the art.
In the present application, a "pharmaceutical composition" refers to a formulation of a compound of the present invention with a vehicle generally accepted in the art for delivery of biologically active compounds to a mammal (e.g., a human). The medium includes a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of active ingredients and exert biological activity.
The term "pharmaceutically acceptable" as used herein refers to a substance (e.g., carrier or diluent) that does not affect the biological activity or properties of the compounds of the present invention and is relatively non-toxic, i.e., the substance can be administered to an individual without causing an adverse biological response or interacting in an adverse manner with any of the components contained in the composition. As used herein, "pharmaceutically acceptable excipient" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersing agent, suspending agent, stabilizing agent, isotonic agent, solvent, or emulsifying agent that is approved by the relevant governmental agency of authorities for use in humans or livestock.
The "tumor" of the present invention includes, but is not limited to, brain tumors including neuroblastoma, glioma, glioblastoma and astrocytoma, sarcoma, melanoma, articular chondroma, cholangioma, leukemia, gastrointestinal stromal tumor, diffuse large B-cell lymphoma, lymphoid cancer such as follicular lymphoma, histiocytic lymphoma, non-small cell lung cancer, pancreatic cancer, squamous cell lung cancer, lung adenocarcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, cervical cancer, ovarian cancer, intestinal cancer, nasopharyngeal cancer, brain cancer, bone cancer, esophageal cancer, melanoma, renal cancer, oral cancer, multiple myeloma, mesothelioma, malignant rhabdoid tumor, endometrial cancer, head and neck cancer, thyroid cancer, parathyroid tumor, uterine tumor, and soft tissue sarcoma.
The terms "preventing," "prevention," and "prevention" as used herein include reducing the likelihood of occurrence or worsening of a disease or disorder in a patient.
The term "treatment" and other similar synonyms as used herein include the following meanings:
(i) Preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is predisposed to the disease or condition but has not yet been diagnosed as having the disease or condition;
(ii) Inhibiting the disease or disorder, i.e., arresting its development;
(iii) Alleviating the disease or condition, i.e., causing a regression of the state of the disease or condition; or
(iv) Alleviating the symptoms caused by the disease or disorder.
The terms "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein, refer to an amount of at least one agent or compound sufficient to alleviate one or more symptoms of the disease or condition being treated to some extent upon administration. The result may be a reduction and/or alleviation of signs, symptoms, or causes, or any other desired change in a biological system. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is clinically necessary to provide a significant disorder-relieving effect. An effective amount suitable in any individual case can be determined using techniques such as a dose escalation assay. The terms "administering," "administration," "administering," and the like, as used herein, refer to a method of delivering a compound or composition to a desired site for a biological effect. These methods include, but are not limited to, oral routes, via the duodenal route, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. Administration techniques useful for The compounds and methods described herein are well known to those skilled in The art, for example, in Goodman and Gilman, the pharmaceutical Basis of Therapeutics, current ed.; pergamon; and Remington's, pharmaceutical Sciences (current edition), mack Publishing Co., easton, pa. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
The terms "drug combination," "administering other therapy," "administering other therapeutic agent," and the like, as used herein, refer to a drug treatment obtained by mixing or combining more than one active ingredient, including fixed and unfixed combinations of active ingredients. The term "fixed combination" refers to the simultaneous administration of at least one compound described herein and at least one co-agent to a patient in the form of a single entity or a single dosage form. The term "non-fixed combination" refers to the simultaneous administration, concomitant administration, or sequential administration at variable intervals of at least one compound described herein and at least one synergistic formulation to a patient as separate entities. These also apply to cocktail therapy, for example the administration of three or more active ingredients. It will also be appreciated by those skilled in the art that in the processes described below, the functional groups of the intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxyl, amino, mercapto, and carboxylic acid. Suitable hydroxyl protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g.t-butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butyloxycarbonyl, benzyloxycarbonyl and the like. Suitable thiol protecting groups include-C (O) -R "(where R" is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like. Suitable carboxyl protecting groups include alkyl, aryl or aralkyl esters. Protecting groups may be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protecting Groups is described in detail in Greene, T.W. and P.G.M.Wuts, protective Groups in organic Synthesis, (1999), 4th Ed., wiley. The protecting group may also be a polymeric resin.
Detailed Description
The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention. The experimental methods without specifying specific conditions in the following examples were selected according to the conventional methods and conditions, or according to the commercial instructions.
The starting materials used in the following examples are commercially available from chemical vendors such as Aldrich, TCI, alfa Aesar, bidey, annelgie, etc., or can be synthesized by known methods.
In the following examples, the ice bath refers to-5 ℃ to 0 ℃, the room temperature refers to 10 ℃ to 30 ℃, and the reflux temperature refers to the solvent reflux temperature under normal pressure. The reaction is carried out overnight in a period of 8 to 15 hours. In the following examples, the operation temperature is not limited and is carried out at room temperature.
In the following examples, the separation and purification of intermediates and final products are by normal phase or reverse phase chromatographic column separation or other suitable methods. The normal phase flash chromatographic column uses ethyl acetate and n-hexane or methanol and dichloromethane and the like as mobile phases. Reverse phase preparative High Pressure Liquid Chromatography (HPLC) was carried out using a C18 column with UV 214nm and 254nm detection, with mobile phases A (water and 0.1% formic acid), B (acetonitrile) or mobile phases A (water and 0.1% ammonium bicarbonate), B (acetonitrile).
In each example: LCMS instrument: pump Agilent 1260 UV detector: agilent 1260 DAD Mass Spectrometer API 3000
A chromatographic column: waters sunfire C18, 4.6X 50mm,5um
Mobile phase: A-H2O (0.1%; b-acetonitrile NMR
The instrument comprises the following steps: bruker Ascend 400M ( 1 H NMR:400MHz; 13 C NMR:100MHz)。
Example 1: preparation of intermediate 5-chloro-8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidine (B1)
The method comprises the following steps: 2-chloro-N- (2, 2-dimethoxyethyl) -5-iodopyrimidin-4-amine
Figure BDA0003608939860000261
To a dry 2L flask were added 2, 4-dichloro-5-iodopyrimidine (110g, 400mmol) and 2, 2-dimethoxyethylamine (84g, 800mmol) and absolute ethanol (1.2L) in this order. After triethylamine (109mL, 800mmol) was slowly added dropwise thereto under nitrogen at 0 ℃ the mixture was stirred at room temperature for 10 hours. After completion of the reaction, concentration was performed in vacuo, and the resulting concentrate was added with 1L of water and extracted with methylene chloride (3X 300 mL), washed with saturated brine and the organic layer was mixed, dried over anhydrous sodium sulfate, filtered and concentrated, and the resulting brown black solid was washed with anhydrous ethanol (3X 50 mL) to give 2-chloro-N- (2, 2-dimethoxyethyl) -5-iodopyrimidin-4-amine (110 g, yield: 78%) as a brown solid.
1 H NMR(400MHz,DMSO-d6)δ8.35(s,1H),4.58(t,J=5.4Hz,1H),3.47(t,J=5.6Hz,2H),3.29(s,6H)ppm;LC-MS:m/z 344.1[M+H] +
Step two: 8-iodoimidazo [1,2-c ] pyrimidin-5-ol
Figure BDA0003608939860000262
To a dry 2L flask were added 2-chloro-N- (2, 2-dimethoxyethyl) -5-iodopyrimidin-4-amine (110g, 317 mmol) and 800mL of concentrated sulfuric acid in that order. The mixture was heated to 65 ℃ under nitrogen and the reaction stirred for 2 hours. After completion of the reaction, the reaction solution was cooled to room temperature, and the mixture was slowly poured into ice water, then the pH was adjusted to about 6.0 with a 4M NaOH solution and extracted with ethyl acetate (3X 300 mL), the organic layers were mixed and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give 8-iodoimidazo [1,2-c ] pyrimidin-5-ol (70 g, 84.5% yield).
1 H NMR(400MHz,DMSO-d6)δ11.80(s,1H),7.92(d,J=1.5Hz,1H),7.60(d,J=3.9Hz,1H),7.40(d,J=1.5Hz,1H);LC-MS:m/z 262.1[M+H] +
Step three: 8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-ol
Figure BDA0003608939860000263
Sequentially adding 8-iodoimidazo [1,2-c ] into a dry 250mL three-neck flask]Pyrimidin-5-ol (2.61g, 10mmol), cuprous iodide (190mg, 1mmol), 1, 10-phenanthroline (360mg, 2mmol), 2, 3-dichlorothiophenol (2.15g, 12mmol), potassium phosphate (4.2g, 20mmol), and 50mL of dioxane solvent. The mixture was heated under nitrogen for 3 hours. After the reaction is finished, saturated NH is added 4 Cl solution (200 mL). It was extracted with ethyl acetate (3x200 mL). The combined organic phases are washed with Na 2 SO 4 Drying, filtering, concentrating the filtrate under reduced pressure, and purifying the resulting residue by silica gel chromatography (gradient 0 to 10% methanol: ethyl acetate) to give 8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c as a pale yellow solid]Pyrimidin-5-ol (2.3 g, yield: 74%).
LC-MS:m/z 312.1[M+H] + .
Step four: 5-chloro-8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidine (B1)
Figure BDA0003608939860000271
To a dry 100mL single-neck flask were added 8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-ol (2.3 g,7.3 mmol) and phosphorus oxychloride (30 mL) in that order, then N, N-diisopropylethylamine (1 mL) was added slowly dropwise under nitrogen, after which the mixture was heated to 120 ℃ and stirred for 4 hours. After completion of the reaction, the mixture was cooled to room temperature and the reaction was concentrated in vacuo and quenched with saturated sodium bicarbonate solution, extracted with ethyl acetate (3 × 30 mL), washed with saturated brine and the organic layer was mixed, dried over anhydrous sodium sulfate, filtered and concentrated, and the resulting residue was purified by silica gel chromatography (gradient 0 to 10% methanol: ethyl acetate) to give 5-chloro-8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidine B1 (660 mg, yield: 27.4%) as a white solid.
1 H NMR(400MHz,DMSO-d6)δ8.18(d,J=1.4Hz,1H),8.07(s,1H),7.76(d,J=1.4Hz,1H),7.52(dd,J=8.0,1.3Hz,1H),7.17(t,J=8.0Hz,1H),7.00(dd,J=8.1,1.2Hz,1H)ppm;LC-MS:m/z 330.1[M+H] + .
Example 2: preparation of intermediate 5-chloro-8-iodoimidazo [1,2-c ] pyrimidine (E1)
Figure BDA0003608939860000272
To a dry 250mL single-neck flask were added 8-iodoimidazo [1,2-c ] pyrimidin-5-ol (5 g, 19.1mmol) and phosphorus oxychloride (50 mL) in that order, N-diisopropylethylamine (1 mL) was slowly added dropwise under nitrogen, after which the mixture was heated to 120 ℃ and stirred for 4 hours. After completion of the reaction, the reaction solution was cooled to room temperature and concentrated in vacuo, then quenched by addition of saturated sodium bicarbonate solution, extracted with ethyl acetate (3 × 100 mL), the organic layers were mixed and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated, and the resulting residue was purified by silica gel chromatography (0 to 10% gradient of methanol: ethyl acetate) to give 5-chloro-8-iodoimidazo [1,2-c ] pyrimidine E1 (1.6 g, yield: 29.8%).
1 H NMR(400MHz,Methanol-d4)δ8.28(s,1H),8.16(d,J=1.6Hz,1H),7.81(d,J=1.6Hz,1H)ppm;LC-MS:m/z 280.1[M+H] + .
Example 3: preparation of intermediate (R) -2-methyl-N- ((R) -8-azaspiro [4.5] decan-1-yl) propane-2-sulfinamide (C-1A)
The method comprises the following steps: (R) -1- ((R) -1, 1-Dimethylethylsulfonamido) -8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000273
1-carbonyl-8-azaspiro [4.5] was added to a dry 100mL single-neck bottle in sequence]Decane-8-carboxylic acid tert-butyl esterButyl ester (2.53g, 10mmol), tetraethoxytitanium (6.84g, 30mmol) and 50mL of tetrahydrofuran were stirred under heating reflux for 4 hours. After cooling to room temperature, methanol (10 mL) was added followed by lithium borohydride (0.65g, 30mmol). The resulting mixture was stirred at room temperature for 3 hours. Methanol was added slowly to quench excess borohydride, followed by addition of brine. The resulting mixture was stirred for 15 minutes and then filtered through celite. The aqueous mixture was extracted with ethyl acetate (3 × 50 mL). The organic phase was washed with MgSO 4 Dry, filter, and remove volatiles under reduced pressure. The residue obtained is purified by chromatography on silica gel (gradient 0 to 50% ethyl acetate: petroleum ether) to give (R) -1- ((R) -1, 1-dimethylethylsulfonamido) -8-azaspiro [4.5] as a white solid]Decane-8-carboxylic acid tert-butyl ester (2.86 g, yield: 80%).
LC-MS:m/z 359.1[M+H] + .
Step two: (R) -2-methyl-N- ((R) -8-azaspiro [4.5] decan-1-yl) propane-2-sulfinamide (C-1A)
Figure BDA0003608939860000281
Reacting (R) -1- ((R) -1, 1-dimethylethylsulfonamido) -8-azaspiro [ 4.5%]A solution of tert-butyl decane-8-carboxylate (2.86g, 8mmol) and concentrated sulfuric acid (2.0mL, 32mmol) in dioxane (50 mL) was stirred at room temperature for reaction for 2 hours. Adding Na 2 CO 3 The aqueous solution was saturated until pH 11, and the aqueous mixture was extracted with DCM (3X 50 mL). The combined organic phases were washed with brine, washed with Na 2 SO 4 Drying, filtering and removing volatiles under reduced pressure to give (R) -2-methyl-N- ((R) -8-azaspiro [4.5] as a white solid]Decan-1-yl) propane-2-sulfinamide C-1A (1.86 g, yield: 90%)
1 H NMR(400MHz,DMSO-d 6 )δ4.82(d,J=7.5Hz,1H),3.04(d,J=7.6Hz,1H),2.81(ddd,J=12.1,8.0,4.0Hz,2H),2.60-2.51(m,2H),1.92-1.14(m,10H),1.12(s,9H)ppm;LC-MS:m/z 259.1[M+H] + .
Following the synthetic procedure of example 1, similar starting materials were reacted to give the following intermediates C-1B, C-1C, C-1D, C-1E, C-1F, C-1G.
Figure BDA0003608939860000282
Figure BDA0003608939860000291
Example 4: preparation of intermediate (R) -2-methyl-N- ((S) -2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (C-1H)
The method comprises the following steps: 4- (2- (benzyloxy) -1-hydroxyethyl) piperidine-1, 4-dicarboxylic acid 1-tert-butyl-4-methyl ester
Figure BDA0003608939860000292
To a dry 500mL three-necked flask, 1-tert-butyl-4-methylpiperidine-1, 4-dicarboxylate (45g, 180mmol) and tetrahydrofuran (400 mL) were added in this order under nitrogen, and the solution was cooled to-78 ℃ and LiHMDS (261mL, 261mmol) was added dropwise. After the addition was complete, the temperature was raised to room temperature and stirred at room temperature for 3 hours. It was then cooled again to-78 ℃ and a solution of benzyloxyacetaldehyde (46g, 300mmol) in tetrahydrofuran (50 mL) was slowly added dropwise. The reaction was slowly warmed to room temperature and stirred for 2.5 hours. After the reaction is finished, adding saturated NH 4 The reaction was quenched with Cl solution (200 mL). It was extracted with ethyl acetate (3 × 200 mL). The combined organic phases are washed with Na 2 SO 4 Drying, filtration, concentration of the filtrate under reduced pressure and chromatography of the resulting residue on silica gel (0 to 50% gradient of ethyl acetate/petroleum ether) afforded 4- (2- (benzyloxy) -1-hydroxyethyl) piperidine-1, 4-dicarboxylic acid 1-tert-butyl-4-methyl ester (52 g, yield: 73.3%).
1 H NMR(400MHz,CDCl 3 )δ7.36-7.30(m,5H),4.50(s,2H),3.97(s,2H),3.73-3.65(m,2H),3.62(s,3H),3.59-3.48(m,3H),2.88(d,J=6.2Hz,1H),2.23(dd,J=13.7,2.7Hz,1H),2.04-1.88(m,2H),1.74(d,J=14.7Hz,1H),1.56(d,J=4.2Hz,1H),1.44(s,9H)ppm;LC-MS:m/z 294.1[M+H] + .
Step two: 4- (2- (benzyloxy) -1-hydroxyethyl) -4- (hydroxymethyl) piperidine-1-carboxylic acid tert-butyl ester
Figure BDA0003608939860000293
To a dry 500mL three-necked flask were added a solution of 4- (2- (benzyloxy) -1-hydroxyethyl) piperidine-1, 4-dicarboxylic acid-1-tert-butyl-4-methyl ester (51.4g, 130mmol) and tetrahydrofuran (500 mL) in this order, and then LiBH was added to the solution 4 (11.44g, 520mmol) and stirred at room temperature for 6 hours. After the reaction is complete, saturated NaHCO is used 3 The reaction was quenched (200 mL). Extract with ethyl acetate (3 × 200 mL). The combined organic phases were washed with Na 2 S0 4 Drying, filtration, concentration of the filtrate under reduced pressure and chromatography of the resulting residue on silica gel (0 to 50% gradient of ethyl acetate/petroleum ether) afforded 4- (2- (benzyloxy) -1-hydroxyethyl) -4- (hydroxymethyl) piperidine-1-carboxylic acid tert-butyl ester (27 g, yield: 57%).
LC-MS:m/z 266.1[M+H] + .
Step three: 4- (1, 2-dihydroxyethyl) -4- (hydroxymethyl) piperidine-1-carboxylic acid tert-butyl ester
Figure BDA0003608939860000301
To a dry 500mL single-neck flask were added tert-butyl 4- (2- (benzyloxy) -1-hydroxyethyl) -4- (hydroxymethyl) piperidine-1-carboxylate (27g, 74mmol), methanol (270 mL) and Pd/C (20 g) in that order, followed by replacement three times with a hydrogen balloon and stirring at room temperature for 12 hours. The reaction mixture was filtered and concentrated to give tert-butyl 4- (1, 2-dihydroxyethyl) -4- (hydroxymethyl) piperidine-1-carboxylate (18.9 g, yield: 93%).
LC-MS:m/z 176.1[M+H] + .
Step four: 4-hydroxy-2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000302
To a dry 500mL one-necked flask were added 4- (1, 2-dihydroxyethyl) -4- (hydroxymethyl) piperidine-1-carboxylic acid tert-butyl ester (18.9g, 69mmol), triphenylphosphine (25.2g, 86.25mmol) and tetrahydrofuran (350 mL) in this order, the reaction was cooled to 0 ℃ and DEAD (12.46mL, 86mmol) was added, followed by warming to room temperature and stirring for 5 hours. After completion of the reaction, the reaction was quenched by addition of saturated water (200 mL). It was extracted with ethyl acetate (3 × 200 mL). The organic phases were combined and washed with Na 2 SO 4 Drying, filtering, concentrating the filtrate under reduced pressure and purifying the resulting residue by silica gel chromatography (0 to 2% gradient methanol/dichloromethane) to give 4-hydroxy-2-oxa-8-azaspiro [4.5]]Tert-butyl decane-8-carboxylate (13.2 g, yield: 74%).
1 H NMR(400MHz,CDCl 3 )δ4.04(dd,J=10.0,4.6Hz,1H),3.98-3.90(m,1H),3.71-3.63(m,2H),3.64-3.49(m,3H),3.20(dt,J=13.4,6.3Hz,1H),3.07(ddd,J=13.2,9.2,3.5Hz,1H),1.95(d,J=5.2Hz,1H),1.74-1.66(m,1H),1.53-1.46(m,1H),1.39(s,9H),1.27-1.11(m,1H)ppm;LC-MS:m/z 202.1[M-56+H] + .
Step five: 4-carbonyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000303
To a dry 500mL single neck flask was added 4-hydroxy-2-oxa-8-azaspiro [4.5] in sequence]Tert-butyl decane-8-carboxylate (13.2g, 51mmol), dichloromethane (280 mL) and Dess-Martin oxidant (32.2g, 76.5 mmol) were stirred for 5h under ice bath. After the reaction is finished, adding NaHCO 3 :Na 2 S 2 O 3 (1). The combined organic phases are washed with Na 2 SO 4 Drying, and concentrating the filtrate under reduced pressure. The residue obtained is purified by chromatography on silica gel (0 to 40% gradient of ethyl acetate/petroleum ether) to give 4-carbonyl-2-oxa-8-azaspiro [4.5] as a colourless solid]Tert-butyl decane-8-carboxylate (12 g, yield: 92.1%).
1 H NMR(400MHz,CDCl 3 )δ4.05(d,J=13.6Hz,4H),3.87(d,J=12.9Hz,2H),3.09(ddd,J=13.5,9.8,3.5Hz,2H),1.73(ddd,J=13.9,9.8,4.3Hz,2H),1.53(d,J=15.1Hz,2H),1.46(s,9H)ppm;LC-MS:m/z200.0[M-56+H] +
Step six: (S) -4- ((R) -1, 1-Dimethylethylsulfonamido) -2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000304
Using the same synthesis as for the intermediate C-1A of example 3, tert-butyl 4-carbonyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylate was reductively aminated to give (S) -4- ((R) -1-methylethylsulfonamido) -2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester as a white solid.
1 H NMR(400MHz,CDCl 3 )δ4.14(dd,J=9.3,6.2Hz,1H),3.90(d,J=13.8Hz,2H),3.77(s,2H),3.70(dd,J=9.2,5.3Hz,1H),3.63(q,J=6.1Hz,1H),3.27(d,J=6.4Hz,1H),2.90(t,J=12.4Hz,2H),1.71(dt,J=16.6,7.9Hz,2H),1.51(s,2H),1.45(s,9H),1.22(s,9H)ppm;LC-MS:m/z 361.1[M-100] +
Step seven: (R) -2-methyl-N- ((S) -2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (C-1H)
Figure BDA0003608939860000311
Using the same synthesis as step two of intermediate C-1A of example 3, (S) -4- ((R) -1-methylethylsulfonamido) -2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester was stripped of the Boc protecting group to give (R) -2-methyl-N- ((S) -2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfonamido C-1H as a white solid.
1 H NMR(400MHz,DMSO-d 6 )δ5.30(s,1H),5.23(d,J=8.9Hz,1H),3.93(dd,J=8.6,7.2Hz,1H),3.69(d,J=8.6Hz,1H),3.58(d,J=8.6Hz,1H),3.46(dd,J=8.5,7.0Hz,2H),2.89-2.73(m,2H),2.48-2.42(m,1H),1.69-1.50(m,2H),1.39-1.21(m,3H),1.12(s,9H)ppm;LC-MS:m/z 261.1[M+H] + .
Example 5: preparation of (R) -2-methyl-N- ((R) -1-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide Synthesis (C-1I)
The method comprises the following steps: 3-methoxyprop-1-yne
Figure BDA0003608939860000312
To a stirred solution of prop-2-yn-1-ol (50g, 892.8 mmol) in water (40 mL) was added 50% aqueous NaOH solution (98.2 g), and the reaction mixture was heated to 70 ℃. Dimethyl sulfate (67.4 g,535.7 mmol) was slowly added to the reaction mixture at below 70 ℃. The reaction mixture was stirred at 60 ℃ for 2h. The product was distilled from the reaction mass at 60 ℃ and collected in a receiver flask cooled at-70 ℃. The distillate was dried with calcium chloride overnight and redistilled to give 3-methoxyprop-1-yne (30 g, yield: 48%) as a colorless liquid.
1 H NMR(CDCl 3, 400MHz)δ4.10(d,J=2.0Hz,2H),3.39(s,3H),2.43(t,J=2.0Hz,1H)ppm.
Step two: 1-methoxy-1, 2-propadiene
Figure BDA0003608939860000313
A suspension of potassium tert-butoxide (3.9g, 35.7mmol) and 3-methoxyprop-1-yne (50g, 714.2mmol) was stirred at 70 ℃ for 2h. The product was distilled from the reaction mass at 50 ℃ and collected in a receiver cooled at-70 ℃ to give 1-methoxypropane-1, 2-diene (35 g, yield: 70%) as a colorless liquid. The compound was dried with KOH and kept at 0 ℃ for storage.
1 H NMR(CDCl 3, 400MHz)δ6.76(t,J=8.0Hz,1H),5.48(d,J=6.0Hz,2H),3.41(s,3H)ppm。
Step three: 4-hydroxy-4- (1-methoxypropan-1, 2-dien-1-yl) piperidine-1-carboxylic acid tert-butyl ester
Figure BDA0003608939860000314
To a stirred solution of 1-methoxypropane-1, 2-diene (0.527g, 7.5 mmol) in THF (10 mL) at-78 deg.C was added n-butyllithium (2.5M in THF) (2.8mL, 7.0 mmol) slowly dropwise and the reaction was stirred at this temperature for an additional 30 minutes. A solution of tert-butyl 4-carbonylpiperidine-1-carboxylate (1.0 g,5.0 mmol) in THF (5 mL) was then added to the reaction mixture and stirring was continued at-78 deg.C for 4 hours. The reaction mixture was saturated NaHCO 3 The aqueous solution was quenched and extracted with ethyl acetate (3X 10 mL). The combined organic layers were washed with brine (50 mL) and dried over anhydrous sodium sulfate. Filtration and concentration under reduced pressure gave tert-butyl 4-hydroxy-4- (1-methoxypropan-1, 2-dien-1-yl) piperidine-1-carboxylate (1.0 g, yield: 90%) as a brown gum
Step four: 4-methoxy-1-oxa-8-azaspiro [4.5] dec-3-ene-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000321
To a stirred solution of tert-butyl 4-hydroxy-4- (1-methoxyprop-1, 2-dien-1-yl) piperidine-1-carboxylate (6.0 g,22.3 mmol) in tert-butanol (60 mL) were added potassium tert-butoxide (12.5g, 111.5 mmol) and dicyclohexyl-18-crown-6 (0.42g, 1.1mmol). The reaction mixture was stirred at reflux for 9 hours. The reaction mixture was cooled to 10 ℃, neutralized with 5% hcl (pH = 7.0), and extracted with ethyl acetate (3 × 150 mL). The combined organic layers were washed with brine (200 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give tert-butyl 4-methoxy-1-oxa-8-azaspiro [4.5] dec-3-ene-8-carboxylate (4.0 g), which was used in the next step without purification.
1 H NMR(CDCl 3, 400MHz)δ4.56(s,2H),3.97(s,1H),3.69(s,3H),3.08(s,1H),1.79-1.74(m,1H),1.52(s,9H),1.45-1.26(m,1H)ppm;LCMS:m/z 214[M-55] + .
Step five: 4-carbonyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000322
Reacting 4-methoxy-1-oxa-8-azaspiro [4.5]]Tert-butyl dec-3-ene-8-carboxylate (38.0 g,141.3 mmol) and p-TSA. H 2 A mixture of O (29.6 g,155.4 mmol) and acetone (400 mL) was stirred at room temperature for 1 hour, and the reaction mixture was saturated with NaHCO 3 The aqueous solution was quenched, extracted with ethyl acetate (3X 500 mL), and the combined organic phases were washed with brine (1000 mL), dried over anhydrous sodium sulfate and filtered, and concentrated under reduced pressure to give 4-carbonyl-1-oxa-8-azaspiro [4.5] as a brown gum]Tert-butyl decane-8-carboxylate (25 g), which was used directly in the next reaction.
LCMS:m/z 278[M+Na] + .
Step six and step seven: synthesis of (R) -2-methyl-N- ((R) -1-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (C-1I)
Figure BDA0003608939860000323
Following the same synthetic procedure as for intermediate C-1A in example 3, the ketone intermediate 4-carbonyl-1-oxa-8-azaspiro [4.5]]The decane-8-carboxylic acid tert-butyl ester is subjected to reductive amination and Boc protective group removal to obtain (R) -2-methyl-N- ((R) -1-oxa-8-azaspiro [ 4.5)]Synthesis of decan-4-yl) propane-2-sulfinamide (C-1I). LCMS m/z 261[ alpha ], [ M ] +Na ]] + .
Example 6: preparation of (R) -2-methyl-N- ((3S, 4S) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (C-1J)
The method comprises the following steps: (S) -2- ((tert-butyldimethylsilyl) oxy) propionic acid ethyl ester
Figure BDA0003608939860000331
To a solution of ethyl (S) -2-hydroxypropionate (30g, 254mmol) in dichloromethane (300 mL) was added imidazole (2.75g, 304.9mmol) and cooled to 0 ℃. To the solution was added tert-butyldimethylsilyl chloride (46.0 g,304.9 mmol) in portions, and stirred at room temperature for 16 hours. After completion of the reaction as judged by TLC analysis, the reaction mixture was quenched with water and extracted with dichloromethane (2 × 100 mL). The combined organic layers were dried over anhydrous sodium sulfate. Filtration and concentration under reduced pressure gave ethyl (S) -2- ((tert-butyldimethylsilyl) oxy) propionate (50 g, 84% yield) as a colorless liquid.
1 H NMR(400MHz,CDCl 3 )δ4.32-4.27(m,1H),4.21-4.12(m,2H),1.37(d,J=6.8Hz,3H),1.27(d,J=7.2Hz,3H),0.90(s,9H),0.08(s,6H)ppm.
Step two: (S) -2- ((tert-butyldimethylsilyl) oxy) propanal
Figure BDA0003608939860000332
To a solution of ethyl (S) -2- ((tert-butyldimethylsilyl) oxy) propionate (25g, 107.6 mmol) in diethyl ether (500 mL) at-78 deg.C was added diisobutylaluminum hydride (1M in hexane) (129mL, 129.1 mmol) slowly dropwise and stirred at-78 deg.C for 1 hour. After completion of the reaction was confirmed by TLC analysis, the reaction mixture was warmed to-40 ℃ and quenched with saturated aqueous Rochelle salt (1L), then diethyl ether (500 mL) was added. The resulting mixture was stirred at room temperature for 2 hours. Then extracted with ether (200 mL). The organic layer was washed with saturated brine (250 mL) and Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give (S) -2- ((tert-butyldimethylsilyl) oxy) propanal (19 g, yield: 94%).
1 H NMR(400MHz,CDCl 3 )δ9.61(s,1H),4.12-4.06(m,1H),1.27(d,J=6.8Hz,3H),0.91(s,9H),0.10(s,6H)ppm.
Step three: 4- ((2S) -2- ((tert-butyldimethylsilyl) oxy) -1-hydroxypropyl) piperidine-1, 4-dicarboxylic acid 1- (tert-butyl)
Figure BDA0003608939860000333
To a stirred solution of 1- (tert-butyl) -4-ethylpiperidine-1, 4-dicarboxylate (30g, 116.6 mmol) in THF (250 mL) at 0 deg.C was added lithium diisopropylamide (2M in THF) (93.3 mL,186.6 mmol) and stirring continued at 0 deg.C for 30 min. Then a solution of (S) -2- ((tert-butyldimethylsilyl) oxy) propanal (22g, 116.6 mmol) in THF (50 mL) was added. The resulting reaction mixture was stirred at 0 ℃ for 1 hour and then kept at room temperature for 1 hour. After the reaction was judged to be complete by TLC analysis, the reaction mixture was quenched with saturated NH 4 The Cl solution was quenched and extracted with ethyl acetate (2X 250 mL). The combined organic layers were washed with water (150 mL), brine (150 mL) and dried over anhydrous sodium sulfate. Filtered and concentrated under reduced pressure. The crude product was purified by column chromatography on silica gel (60-120 mesh) using a solvent gradient mixture of 25% ethyl acetate in petroleum ether as eluent to give 1- (tert-butyl) 4- ((2S) -2- ((tert-butyldimethylsilyl) oxy) -1-hydroxypropyl) piperidine-1, 4-dicarboxylic acid 1- (tert-butyl) (17 g, yield: 32%) as a pale red oil.
1 H NMR(400MHz,CDCl 3 )δ4.29-4.09(m,2H),3.96-3.94(m,2H),3.86-3.80(m,1H),3.56-3.54(m,1H),2.86-2.76(m,2H),2.46(d,J=5.2Hz,1H),2.16-2.13(m,1H),2.13-2.04(m,1H),1.77-1.60(m,2H),1.46(s,9H),1.29-1.24(m,3H),1.12(d,J=4Hz,3H),0.89(s,9H),0.05(s,6H)ppm;LCMS:m/z 346[M-100] + .
Step four: ((2S) -2- ((tert-butyldimethylsilyl) oxy) -1-hydroxypropyl) -4- (hydroxymethyl) piperidine-1-carboxylic acid tert-butyl ester
Figure BDA0003608939860000341
To the stirred solution was added a solution of 4- ((2S) -2- ((tert-butyldimethylsilyl) oxy) -1-hydroxypropyl) piperidine-1, 4-dicarboxylic acid 1- (tert-butyl) (5g, 11.21mmol) in THF (50 mL) and LiBH was added portionwise 4 (0.73g, 33.65mmol) and stirred at room temperature for 16 hours. After the reaction is finished, the reaction kettle is used for reaction,the reaction mixture was washed with saturated NaHCO at 0 deg.C 3 The solution was quenched and stirred at room temperature for 15 minutes. The precipitated solid was filtered off and the aqueous phase was extracted with ethyl acetate (2X 50 mL). The combined organic layers were dried over anhydrous sodium sulfate. The crude product obtained by filtration and concentration under reduced pressure was purified by column chromatography on silica gel (100-200 mesh) using a gradient mixture of 25% ethyl acetate in petroleum ether as eluent to give tert-butyl ((2S) -2- ((tert-butyldimethylsilyl) oxy) -1-hydroxypropyl) -4- (hydroxymethyl) piperidine-1-carboxylate (3 g, yield: 66%).
1 H NMR(400MHz,CDCl 3 )δ4.55(t,J=4.8Hz,1H),4.43(d,J=6.4Hz,1H),3.52-3.47(m,5H),3.31-3.28(m,1H),3.05-3.01(m,2H),1.58-1.49(m,2H),1.42-1.38(m,11H),1.11(d,J=6.4Hz,3H),0.85(m,9H),0.04(s,6H)ppm;LC-MS:m/z 404.3[M+H] +
Step five: 4- ((2S) -1, 2-dihydroxypropyl) -4- (hydroxymethyl) piperidine-1-carboxylic acid tert-butyl ester
Figure BDA0003608939860000342
To a solution of tert-butyl ((2S) -2- ((tert-butyldimethylsilyl) oxy) -1-hydroxypropyl) -4- (hydroxymethyl) piperidine-1-carboxylate (25g, 61.93mmol) in THF (500 mL) was added tetrabutylammonium fluoride (1M in THF) (93mL, 92.89mmol), and the resulting reaction mixture was stirred at room temperature for 2 hours. After completion of the reaction as judged by TLC analysis, the reaction mixture was taken up with saturated NaHCO 3 The solution was quenched and extracted with ethyl acetate (2X 500 mL). The combined organic phases were dried over anhydrous sodium sulfate. The crude product obtained was purified by column chromatography on silica gel (60-120 mesh) using a solvent gradient mixture of 70-90% ethyl acetate in petroleum ether as an eluent, to give tert-butyl 4- ((2S) -1, 2-dihydroxypropyl) -4- (hydroxymethyl) piperidine-1-carboxylate (12 g, yield: 67%) as a colorless liquid.
1 H NMR(400MHz,DMSO-d 6 )δ4.72(t,J=4.8Hz,1H),4.61(d,J=5.2Hz,1H),4.50(d,J=7.2Hz,1H),3.72-3.68(m,1H),3.53-3.44(m,4H),3.11-2.98(m,3H),1.68-1.53(m,2H),1.42-1.35(m,11H),1.10(d,J=6.4Hz,3H)ppm;LC-MS:m/z 290.1[M+H] +
Step six: (3S) -4-hydroxy-3-methyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000343
To a stirred suspension of NaH (60% in mineral oil) (1.45g, 60.5mmol) in THF (30 mL) at 0 deg.C was added a solution of 4- ((2S) -1, 2-dihydroxypropyl) -4- (hydroxymethyl) piperidine-1-carboxylic acid tert-butyl ester (5g, 17.3mmol) and p-toluenesulfonyl chloride (3.29g, 17.3mmol) in THF (20 mL) and the resulting reaction mixture was reacted at 0 deg.C for 3 hours. After the reaction was complete, the reaction mixture was saturated with NH at-20 deg.C 4 The Cl solution (250 mL) was quenched and extracted with ethyl acetate (2X 50 mL). The combined organic layers were dried over anhydrous sodium sulfate. The crude product obtained by filtration and concentration under reduced pressure is purified by column chromatography on silica gel (100-200 mesh) using a solvent gradient mixture of 40% ethyl acetate in petroleum ether as eluent to give (3S) -4-hydroxy-3-methyl-2-oxa-8-azaspiro [ 4.5%]Tert-butyl decane-8-carboxylate (2.1 g, yield: 44%).
1 H NMR(400MHz,CDCl 3 )δ3.83-3.62(m,5H),3.43(d,J=6.0,1H),3.07-2.97(m,2H),1.72-1.55(m,3H),1.51-1.42(m,11H),1.33(d,J=6.4Hz,3H)ppm;LC-MS:m/z 172.2[M-100] +
Step seven: (S) -tert-butyl-3-methyl-4-carbonyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid ester
Figure BDA0003608939860000351
Tert-butyl (3S) -4-hydroxy-3-methyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylate (2.1g, 7.74mmol) was added to a solution of tetrahydrofuran (50 mL) and stirring was maintained for 1 hour. After the reaction was completed, the solvent was distilled off under reduced pressure. The resulting residual product was purified by column chromatography on silica gel (100-200 mesh) using a solvent gradient mixture of 30% ethyl acetate in petroleum ether as an eluent, followed by flash chromatography using 0.1% formic acid and acetonitrile as eluents to give (S) -tert-butyl-3-methyl-4-carbonyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid ester (1.2 g, yield: 57%).
1 H NMR(400MHz,CDCl 3 )δ4.20(d,J=9.5Hz,1H),3.94-3.90(m,4H),3.16-3.10(m,1H),3.03-2.97(m,1H),1.81-1.75(m,1H),1.67-1.62(m,1H),1.61-1.57(m,1H),1.42-1.45(m,10H),1.32(d,J=6.0Hz,3H)ppm;LC-MS:m/z 214.1[M-55] +
Step eight: (3S, 4S) -4- ((R) -tert-butylsulfinyl) amino) -3-methyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000352
(S) -tert-butyl-3-methyl-4-carbonyl-2-oxa-8-azaspiro [4.5]A stirred solution of decane-8-carboxylic acid ester (1.2g, 4.46mmol) in THF (15 mL) was charged with (R) -2-methylpropane-2-sulfinamide (1.07g, 8.91mmol) and tetraethyltitanate (4.07g, 17.84mmol), respectively. The resulting reaction mixture was stirred at 90 ℃ for 20 hours. The reaction mixture was cooled to-4 ℃ and MeOH (2 mL) was added followed by the addition of LiBH in portions 4 (282mg, 12.99mmol) and stirring was maintained at the same temperature for 1 hour. After completion of the reaction, the reaction mixture was quenched with a saturated saline solution at 0 ℃ and stirred at room temperature for 15 minutes. Filtration and the solution extracted with ethyl acetate (2X 50 mL). The combined organic layers were dried over anhydrous sodium sulfate. The crude product obtained was filtered and concentrated under reduced pressure and purified by GRACE flash chromatography using 0.1% formic acid and acetonitrile as eluent to give (3S, 4S) -4- ((R) -tert-butylsulfinyl) amino) -3-methyl-2-oxa-8-azaspiro [4.5]Decane-8-carboxylic acid tert-butyl ester (1.2 g, yield: 72%).
1 H NMR(400MHz,CDCl 3 )δ4.20-4.15(m,1H),3.90-3.84(m,2H),3.63-3.59(m,1H),3.49-3.43(m,1H),3.31-3.29(m,1H),2.95-2.81(m,2H),1.90-1.71(m,2H),1.49-1.40(m,11H),1.25(s,9H),1.19(d,J=6.5Hz,3H)ppm;LC-MS:m/z 375.2[M+H] +
Step nine: (R) -2-methyl-N- ((3S, 4S) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (C-1J)
Figure BDA0003608939860000353
To a solution of tert-butyl (3S, 4S) -4- ((R) -tert-butylsulfinyl) amino) -3-methyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylate (1.1 g,2.936 mmol) in dichloromethane (10 mL) was added trifluoroacetic acid (1.12mL, 14.68mmol) dropwise and stirred at room temperature for 6 hours. After completion of the reaction, the crude product obtained by concentrating the reaction mixture under reduced pressure was purified by chromatography using 0.1% formic acid and acetonitrile to give (R) -2-methyl-N- ((3S, 4S) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide C-1J (850 mg, yield: 72%).
1 H NMR(400MHz,DMSO-d 6 )δ8.40(brs,D 2 O Exchangeable,1H),8.30(brs,D 2 O Exchangeable,1H),5.28(d,J=12.0Hz,1H),4.13-4.09(m,1H),3.77(d,J=9.0Hz,1H),3.50-3.45(m,2H),3.29-3.26(m,1H),3.19-3.15(m,1H),2.94-2.85(m,2H),1.87-1.80(m,2H),1.69-1.59(m,2H),1.17(s,9H),1.08(d,J=6.0Hz,3H)ppm;LC-MS:m/z 275.2[M+H] +
Example 7: preparation of intermediate (R) -2-methyl-N- ((1R) -2-methyl-8-azaspiro [4.5] decan-1-yl) propane-2-sulfinamide (C-1K)
The method comprises the following steps: 2-methyl-1-carbonyl-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000361
1-carbonyl-8-azaspiro [4.5] is added into a 100mL dry single-neck flask in sequence under the protection of nitrogen at 0 DEG C]Tert-butyl decane-8-carboxylate (1g, 3.95mmol) and dried tetrahydrofuran (15 mL), liHMDS (3.95ml, 3.95mmol) was slowly added dropwise thereto, and after stirring at that temperature for 1 hour, iodomethane (0.25mL, 3.95mmol) was added thereto,and stirring was continued for 2 hours. After completion of the reaction, the reaction was quenched with saturated sodium bicarbonate solution, extracted with ethyl acetate (3X 20 mL), and the combined organic phases were Na 2 SO 4 Drying, concentrating the filtrate under reduced pressure and purifying by column silica gel chromatography (0 to 20% gradient of ethyl acetate/petroleum ether) to give 2-methyl-1-carbonyl-8-azaspiro [4.5]]Decane-8-carboxylic acid tert-butyl ester. LCMS m/z 368.0[ m ] +H] + .
Step two and step three: (R) -2-methyl-N- ((1R) -2-methyl-8-azaspiro [4.5] decan-1-yl) propane-2-sulfinamide (C-1K)
Figure BDA0003608939860000362
Following the same synthetic procedure as intermediate C-1A in example 3, tert-butyl ketone intermediate 2-methyl-1-carbonyl-8-azaspiro [4.5] decane-8-carboxylate was reductively aminated and the Boc protecting group was removed to give (R) -2-methyl-N- ((1R) -2-methyl-8-azaspiro [4.5] decan-1-yl) propane-2-sulfinamide C-1K.
1 H NMR(400MHz,CDCl 3 )δ5.07-4.97(m,1H),3.32-3.20(m,1H),3.01-2.84(m,2H),2.81-2.61(m,2H),2.20-2.11(m,1H),2.02-1.34(m,8H),1.25-1.20(m,9H),1.06-0.99(m,3H)ppm;LCMS:m/z 273.0[M+H] + Example 8: (R) -N- ((R) -3, 3-dimethyl-1-oxa-8-azaspiro [ 4.5)]Preparation of decan-4-yl) -2-methylpropane-2-sulfinamide (C-1L)
The method comprises the following steps: 3, 3-dimethyl-4-carbonyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000363
4-carbonyl-1-oxa-8-azaspiro [4.5] at-78 ℃ under the protection of nitrogen]To a solution of tert-butyl decane-8-carboxylate (5g, 19.6 mmol) in THF (50 mL) was added LiHMDS (1M in THF; 19.6mL,19.6 mmol) slowly dropwise and the mixture was stirred at-78 ℃ for 2h. Then, after the reaction mixture was warmed to room temperature, methyl iodide (1.22mL, 19.6 mmol) was added thereto in portions. Subjecting the obtained reaction mixture toStirring was continued at room temperature for 2 hours. The reaction mixture was diluted with ethyl acetate (150 mL) and saturated NaHCO 3 The solution was quenched (150 mL) and then extracted with ethyl acetate (2X 300 mL). The combined organic phases were dried over anhydrous sodium sulfate. Filtered and concentrated under reduced pressure. The residue obtained is purified by chromatography on silica gel (gradient 0 to 15% ethyl acetate/petroleum ether) to give 3, 3-dimethyl-4-carbonyl-1-oxa-8-azaspiro [4.5]]Decane-8-carboxylic acid tert-butyl ester (1 g, yield: 18%) and 3-methyl-4-carbonyl-1-oxa-8-azaspiro [4.5]]Decane-8-carboxylic acid tert-butyl ester (0.7 g, yield: 14%)
3, 3-dimethyl-4-carbonyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester:
1 HNMR(400MHz,CDCl 3 )δ3.94(brs,2H),3.89(s,2H),3.16-3.10(m,2H),1.70-1.61(m,4H),1.48(s,9H),1.14(s,6H)ppm;LCMS:m/z 306[M+Na] + .
3-methyl-4-carbonyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester:
1 H-NMR(400MHz,CDCl 3 )δ4.38(t,J=8.8Hz,1H),3.96(brs,2H),3.67(t,J=9.6Hz,1H),3.14-3.09(m,2H),2.65-2.58(m,1H),1.75-1.46(m,13H),1.15(d,J=7.2Hz,3H)ppm;LCMS:m/z 292[M+Na] + .
step two: (S) -4- ((tert-butylsulfinyl) imino) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000371
To a stirred solution of tert-butyl 3, 3-dimethyl-4-carbonyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylate (2.0g, 7.1 mmol) in THF (5.0 mL) was added (R) -2-methylpropane-2-sulfinamide (2.56g, 21.2mmol) and tetraethyltitanate (8.05g, 35.3mmol). The resulting reaction mixture was stirred at 90 ℃ for 48 hours. The reaction mixture was quenched with methanol (10 mL) and diluted with ethyl acetate (50 mL), then filtered over celite, and the crude product obtained by concentration was separated by reverse phase flash chromatography using 0.1% formic acid and acetonitrile as eluent to give tert-butyl (S) -4- ((tert-butylsulfinyl) imino) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylate (1.5 g, yield: 55%).
1 HNMR(400MHz,CDCl 3 )δ4.10-3.88(brs,2H),3.80-3.72(m,2H),3.04(brs,2H),1.70-1.59(m,5H),1.46-1.41(m,14H),1.24-1.14(m,9H)ppm;LCMS:m/z 331[M-55] + .
Step three: (R) -4- (((R) -tert-butylsulfinyl) amino) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester and (S) -4- (((R) -tert-butylsulfinyl) amino) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester)
Figure BDA0003608939860000372
To (S) -4- ((tert-butylsulfinyl) imino) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5]Tert-butyl decane-8-carboxylate (1.5g, 3.88mmol) to a mixed solvent of MeOH: THF (3 4 (886mg, 23.3mmol) was then stirred under reflux for 16 hours. After cooling to room temperature, part of the solvent was concentrated to 10mL under reduced pressure, and the mixture was poured into ice-water (100 mL) and stirred for 10 minutes. The resulting solid precipitate was removed by filtration, extracted with ethyl acetate (3X 50 mL), and the combined organic phases were concentrated by drying. The obtained crude product is purified and separated to obtain (R) -4- (((R) -tert-butylsulfinyl) amino) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5]]Decane-8-carboxylic acid tert-butyl ester (350 mg, yield: 23%) and (S) -4- (((R) -tert-butylsulfinyl) amino) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5]Tert-butyl decane-8-carboxylate (350 mg, yield: 23%) and a mixture of the two (400 mg, yield: 28%).
(S) -4- (((R) -tert-butylsulfinyl) amino) -3, 3-dimethyl-1-oxa-8-azaspiro [ 4.5%]Decane-8-carboxylic acid tert-butyl ester: 1 H NMR(400MHz,CDCl 3 )δ3.94(brs,2H),3.61(d,J=9.2Hz,1H),3.54(d,J=9.2Hz,1H),3.22(d,J=10Hz,1H),3.07-3.00(m,3H),1.54-1.49(m,4H),1.45(s,9H),1.24(s,9H),1.21(s,3H),1.03(s,3H)ppm;LCMS:m/z 411[M+Na] + ;[α] 25 D =+19.62(c 0.25,MeOH);retention time:1.835min
(R) -4- (((R) -tert-butylsulfinyl) amino) -3, 3-dimethyl-1-oxa-8-azaspiro [ 4.5%]Decane-8-carboxylic acid tert-butyl ester: 1 HNMR(400MHz,CDCl 3 )δ4.03-3.93(m,2H),3.62-3.58(m,1H),3.50-3.47(m,1H),3.30(brs,1H),3.11-2.96(m,3H),1.91-1.76(m,2H),1.54-1.56(m,merged in DMSO,2H)1.43(s,9H),1.25(s,9H),1.03(s,3H),0.99(s,3H)ppm;LCMS:m/z 411[M+Na] + ;[α] 25 D =-43.56(c 0.25,MeOH);retention time:2.009min.
step four: (R) -N- ((R) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decan-4-yl) -2-methylpropane-2-sulfinamide (C-1L)
Figure BDA0003608939860000381
Using the same synthetic procedure as in step nine of Synthesis example 6, intermediate C-1J, (R) -4- (((R) -tert-butylsulfinyl) amino) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester was stripped of the Boc protecting group to give (R) -N- ((R) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decan-4-yl) -2-methylpropane-2-sulfinamide C-1L.
1 H-NMR(400MHz,DMSO-d 6 )δ4.92(d,D 2 O Exchangeable,J=11.5Hz,1H),3.50(d,J=9.0Hz,1H),3.43(d,J=9.0Hz,1H),3.20-3.14(m,2H),3.09(d,J=12.0,1H),3.00-2.89(m,2H),1.92-1.86(m,1H),1.82-1.80(m,2H),1.73-1.70(m,1H),1.19(s,9H),0.97(s,3H),0.94(s,3H)ppm;LCMS:m/z 289[M+H] +
Example 9: preparation of (R) -N- ((S) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decan-4-yl) -2-methylpropane-2-sulfinamide (C-1M)
Figure BDA0003608939860000382
Using the same synthetic procedure as in step nine of Synthesis example 6, intermediate C-1J, (S) -4- (((R) -tert-butylsulfinyl) amino) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester was stripped of the Boc protecting group to give (R) -N- ((S) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decan-4-yl) -2-methylpropane-2-sulfinamide C-1M.
1 HNMR(400MHz,DMSO-d 6 )δ8.40(brs,D 2 O Exchangeable,1H),8.40(brs,D 2 O Exchangeable,1H),5.27(d,J=11.0Hz,1H),4.12-4.10(m,1H),3.77(d,J=8.5Hz,1H),3.47-3.44(m,2H),3.28-3.24(m,1H),3.17-3.15(m,1H),2.95-2.87(m,2H),1.86-1.82(m,2H),1.69-1.59(m,2H),1.17(s,9H),1.08(d,J=6.0Hz,3H)ppm;LCMS:m/z 289[M+H] + .
Example 10: preparation of intermediate (R) -2-methyl-N- ((1R) -3-methyl-8-aza-spiro [4.5] decan-1-yl) propane-2-sulfinamide (C-1N)
The method comprises the following steps: 4-allyl-4-formylpiperidine-1-carboxylic acid tert-butyl ester
Figure BDA0003608939860000391
To a dry 1L flask were added tert-butyl 4-formylpiperidine-1-carboxylate (35.0 g, 164mmol), lithium tert-butoxide (15.77g, 197mmol) and allyl bromide (11.54mL, 189mmol) in that order and the mixture was stirred at 0 ℃ for 1 hour. After the reaction is completed, the mixture is poured into a container containing saturated NH 4 Aqueous Cl solution H2O (1,500mL) was added to the separation funnel using Et 2 O (5X 50 mL). The combined organic phases were washed with MgSO 4 Drying, filtering, and concentrating the filtrate under reduced pressure. The resulting residue was purified by silica gel chromatography (0 to 25% gradient of ethyl acetate/petroleum ether) to give tert-butyl 4-allyl-4-formylpiperidine-1-carboxylate (24 g, yield: 48%) as a colorless oil.
1 H NMR(400MHz,CDCl 3 )δ9.52(s,1H),5.53-5.76(m,1H),4.96-5.19(m,2H),3.80(br.s.,2H),2.97(t,J=11.49Hz,2H),2.26(d,J=7.33Hz,2H),1.95(dt,J=13.71,3.13Hz,2H),1.38-1.58(m,11H)ppm.
Step two: 4-allyl-4- (1-hydroxyallyl) piperidine-1-carboxylic acid tert-butyl ester (C-1N-C)
Figure BDA0003608939860000392
To a dry 1L three-necked flask was added 4-allyl-4-formylpiperidine-1-carboxylic acid tert-butyl ester (24g, 95mmol) and THF (300 mL) in that order, the solution was cooled to-78 deg.C and vinyl magnesium bromide (1M in THF, 118mL, 118mmol) was slowly added dropwise under nitrogen. The resulting solution was allowed to slowly warm to room temperature over a period of 1 hour. After the reaction is completed, the mixture is poured into a reactor containing saturated NH 4 Aqueous Cl (250 mL) was extracted with EtOAc (4 × 50 mL) in a separatory funnel. The combined organic phases were washed with MgSO 4 Drying, filtration and concentration of the filtrate under reduced pressure gave tert-butyl 4-allyl-4- (1-hydroxyallyl) piperidine-1-carboxylate (26.7 g), which was used in the next step without further purification.
1 H NMR(400MHz,CDCl 3 )δ6.05-5.83(m,2H),5.32-5.21(m,2H),5.12(s,1H),5.08(d,J=3.5Hz,1H),4.05-3.97(m,1H),3.71(s,2H),3.12(ddd,J=13.8,10.4,3.6Hz,2H),2.33(dd,J=14.3,7.8Hz,1H),2.20(dd,J=14.3,7.2Hz,1H),1.60(q,J=4.3Hz,2H),1.57-1.50(m,2H),1.45(s,9H)ppm.
Step three: 4-Enopropionyl-4-allylpiperidine-1-carboxylic acid tert-butyl ester
Figure BDA0003608939860000393
To a dry 1L three-necked flask were added 4-allyl-4- (1-hydroxyallyl) piperidine-1-carboxylic acid tert-butyl ester (26.7 g, 95mmol), dess-Martin oxidant (44.3 g, 105mmol), and anhydrous dichloromethane (380 mL) in this order, and the mixture was stirred at room temperature for 1 hour. After the reaction is finished, the mixture is poured into a reactor containing NaHCO 3 :Na 2 SO 3 Saturated aqueous solution (1, 300ml) in a separatory funnel, then extracted with DCM (4 × 50 mL). The combined organic phases were washed with MgSO 4 Drying, filtering, and concentrating the filtrate under reduced pressure to obtain white solid. The white solid was suspended in petroleum ether (250 mL) and sonicated for 20 min. The white suspension was passed through a pad of celiteFiltration and removal under reduced pressure, and concentration of the filtrate under reduced pressure gave 4-levulinyl-4-allylpiperidine-1-carboxylic acid tert-butyl ester (25 g, two-step yield: 94%) as a yellow oil.
1 H NMR(400MHz,CDCl 3 )δ6.80(dd,J=16.8,10.3Hz,1H),6.39(dd,J=16.8,1.9Hz,1H),5.70(dd,J=10.3,1.9Hz,1H),5.55(ddt,J=17.5,10.2,7.4Hz,1H),5.09-4.98(m,2H),3.77(s,2H),2.94(s,2H),2.31(d,J=7.4Hz,2H),2.08(d,J=13.8Hz,2H),1.47-1.41(m,11H)ppm。
Step four: 1-carbonyl-8-azaspiro [4.5] decan-2-ene-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000401
To a dry 1L three-necked flask was added sequentially a solution of tert-butyl 4-levulinyl-4-allylpiperidine-1-carboxylate (25g, 89.6 mmol), toluene (degassed, 850 mL), and Grubbs' secondary catalyst (2.02g, 2.38mmol) in toluene (degassed, 100 mL). The resulting mixture was stirred at 85 ℃ under nitrogen for 45 minutes. After the reaction was complete, the solvent was removed under reduced pressure and the resulting residue was purified by silica gel chromatography (0 to 40% gradient of ethyl acetate/petroleum ether) to give tert-butyl 1-carbonyl-8-azaspiro [4.5] dec-2-ene-8-carboxylate (19g, 83mmol) as a brown solid. A solution of this compound and DDQ (565mg, 2.49mmol) in toluene (540 mL) was stirred at room temperature for 15 minutes. The resulting bright red solution was filtered through a pad of celite. Charcoal (100 g) was added and the resulting suspension was stirred at room temperature for 2 hours. The mixture was filtered through a celite pad, and the residue obtained by concentrating the filtrate under reduced pressure was purified by silica gel chromatography (0 to 40% gradient of ethyl acetate/petroleum ether) to give tert-butyl 1-carbonyl-8-azaspiro [4.5] dec-2-ene-8-carboxylate (12 g, yield: 53.3%) as a white solid.
Step five: 3-methyl-1-carbonyl-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000402
CuI (3.8g, 20mmol) and anhydrous tetrahydrofuran (100 mL) were sequentially added to a nitrogen-protected 250mL dry three-necked flask, the solution was cooled to-20 ℃, meLi (1.6M THF solution, 25mL, 40mmol) was slowly added dropwise to the solution, and after the addition was completed, the reaction solution was reacted at-20 ℃ until the solution was clear. Then slowly dropwise adding 1-carbonyl-8-azaspiro [4.5] at the temperature]A solution of tert-butyl decane-2-ene-8-carboxylate (2.51g, 10 mmol) in tetrahydrofuran (20 mL). After the reaction is finished, the mixture is poured into a reactor containing saturated NH 4 Aqueous Cl was extracted with ethyl acetate (3 × 15 mL) in a separatory funnel. The combined organic phases were washed with MgSO 4 Drying, filtration and concentration of the filtrate under reduced pressure purification by silica gel chromatography (0 to 50% gradient ethyl acetate/petroleum ether) afforded 3-methyl-1-carbonyl-8-azaspiro [4.5]]Tert-butyl dec-2-ene-8-carboxylate (1.6 g, yield: 60%).
1 H NMR(400MHz,CDCl 3 )δ3.92(s,1H),3.81(s,1H),3.55(d,J=5.0Hz,1H),3.13-3.04(m,1H),2.96(t,J=10.9Hz,1H),2.56-2.46(m,1H),2.31-2.21(m,2H),1.94-1.75(m,2H),1.62-1.49(m,1H),1.45(s,9H),1.41-1.35(m,2H),1.15(d,J=6.0Hz,3H),0.90(t,J=6.9Hz,3H)ppm.
Step six and step seven: (R) -2-methyl-N- ((1R) -3-methyl-8-aza-spiro [4.5] decan-1-yl) propane-2-sulfinamide (C-1N)
Figure BDA0003608939860000403
Using the same synthetic method as for step eight and nine for the synthesis of intermediate C-1J, the ketone intermediate 3-methyl-1-carbonyl-8-azaspiro [4.5] dec-2-ene-8-carboxylic acid tert-butyl ester was reductively aminated and the Boc protecting group was removed to give (R) -2-methyl-N- ((1R) -3-methyl-8-aza-spiro [4.5] dec-1-yl) propane-2-sulfinamide C-1N.
1 H NMR(400MHz,CDCl 31 H NMR(400MHz,DMSO-d 6 )δ3.04-2.95(m,1H),2.75(s,2H),2.62-2.53(m,2H),1.93-1.57(m,5H),1.52-1.27(m,13H),0.96(d,J=6.5Hz,3H)ppm;LCMS:m/z 273[M+H] + .
Example 11: preparation of intermediate (R) -2-methyl-N- ((1R, 3R) -3-methyl-8-azaspiro [4.5] decan-1-yl) propane-2-sulfinamide (C-1O)
The method comprises the following steps: (R) -3- ((tert-butyldimethylsilyl) oxy) -1-carbonyl-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000411
To a dry 100mL three-necked flask, cuCl (19mg, 0.19mmol),(s) -TolBlNAP (129mg, 0.19mmol), sodium tert-butoxide (18mg, 0.19mmol), and THF (9 mL) were added in this order, and the mixture was stirred at room temperature for 30 minutes. Adding B 2 Pin 2 (1.78g, 7.01mmol) in THF (2.5 mL) and the resulting mixture stirred at room temperature for 10 minutes. Then adding 1-carbonyl-8-azaspiro [4.5]]A solution of tert-butyl decan-2-ene-8-carboxylate (1.60g, 6.37mmol) in THF (9 mL) was added MeOH (0.53mL, 12.74mmol). The resulting mixture was stirred at room temperature for 16 hours. After the reaction is finished, H is added 2 O (20 mL), followed by sodium perborate (4.84g, 32mmol) and the resulting mixture stirred vigorously at room temperature for l hours. The resulting green suspension was filtered through a pad of celite and poured into a container containing NaHCO 3 Saturated aqueous solution: na (Na) 2 SO 3 The separatory funnel was saturated with aqueous solution (40 mL) and extracted with EtOAc (4X 40 mL). The combined organic phases were washed with MgSO 4 Drying, filtering and concentrating the filtrate under reduced pressure to obtain crude product of (R) -3-hydroxy-1-carbonyl-8-azaspiro [4.5]Decane-8-carboxylic acid tert-butyl ester.
Crude (R) -3-hydroxy-1-carbonyl-8-azaspiro [4.5] was added to a 100mL single-neck flask in sequence]Tert-butyl decane-8-carboxylate (theoretical amount, 6.37 mmol), imidazole (650mg, 9.56mmol), TBSCl (1.20g, 7.96mmol) and DMF (16 mL), and the mixture was stirred at room temperature for reaction for 16 hours. After the reaction is complete, the reaction mixture is poured into a container containing saturated NH 4 Aqueous Cl (30 mL) was separated from the funnel and extracted with ethyl acetate (5 × 50 mL). The combined organic phases were washed with MgSO 4 The residue obtained is dried, filtered and concentrated under reduced pressure is purified by chromatography on silica gel (0 to 40% gradient ethyl acetate/stone)Oily ether) to give (R) -3- ((tert-butyldimethylsilyl) oxy) -1-carbonyl-8-azaspiro [4.5]Tert-butyl decane-8-carboxylate (1.26 g, yield: 51.6%) as a colorless oil.
LCMS:m/z 328[M-56+H] + .
Step two (1R, 3R) -3- ((tert-butyldimethylsilyl) oxy) -1- ((R) -1, 1-dimethylethylsulfenamino) -8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000412
Using the same synthetic procedure as for step eight of the synthesis of intermediate C-1J, the reductive amination of the ketone intermediate, tert-butyl (R) -3- ((tert-butyldimethylsilyl) oxy) -1-carbonyl-8-azaspiro [4.5] decane-8-carboxylate, gave a white solid, tert-butyl (1R, 3R) -3- ((tert-butyldimethylsilyl) oxy) -1- ((R) -1, 1-dimethylethylsulfonamido) -8-azaspiro [4.5] decane-8-carboxylate (1.24 g, yield: 77%).
1 H NMR(400MHz,CDCl 3 )δ4.23(s,1H),3.84(d,J=13.6Hz,2H),3.24(s,1H),2.77(td,J=12.7,12.0,3.0Hz,2H),2.27(d,J=8.8Hz,1H),1.72-1.54(m,5H),1.38(s,9H),1.19(d,J=2.5Hz,3H),1.14(s,9H),0.80(s,9H),-0.03(s,6H)ppm;LCMS:m/z 488.9[M+H] + .
Step three: (R) -N- ((1R, 3R) -3-hydroxy-8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide (C-1O)
Figure BDA0003608939860000413
(1R) -3- ((tert-butyldimethylsilyl) oxy) -1- ((R) -1, 1-dimethylethylsulfinylamino) -8-azaspiro [4.5] under an ice bath]Concentrated sulfuric acid (0.023ml, 0.4mmol) was added to a solution of t-butyl decane-8-carboxylate (49mg, 0.1mmol) in 1, 4-dioxane (1 mL). The reaction was stirred at room temperature for 2 hours. The reaction was adjusted to PH =12 with sodium hydroxide solution and then extracted with DCM (4 × 10 mL). The combined organic phases are dried over anhydrous sodium sulfateFiltration and removal of volatiles under reduced pressure gave (R) -N- ((1R, 3R) -3-hydroxy-8-azaspiro [ 4.5%]Decan-1-yl) -2-methylpropane-2-sulfinamide C-1O (20 mg, yield: 70%). LC-MS m/z 275[ m ] +H] +
Example 12: preparation of intermediate (R) -tert-butyl 1- ((tert-butoxycarbonyl) amino) -3, 3-difluoro-8-azaspiro [4.5] decane-8-carboxylate (C-1P)
The method comprises the following steps: (1R, 3R) -1-amino) -3-hydroxy-8-azaspiro [4.5] decane
Figure BDA0003608939860000421
To a solution of tert-butyl (1R, 3R) -3- ((tert-butyldimethylsilyl) oxy) -1- ((R) -1, 1-dimethylethylsulfonamido) -8-azaspiro [4.5] decane-8-carboxylate (100mg, 0.2mmol) in methanol (5 mL) at room temperature was slowly added a solution of 1, 4-dioxane hydrochloride (4M, 2mmol,0.5 mL) and the reaction mixture was heated at 40 ℃ for 1 hour. Concentration under reduced pressure gave (1R, 3R) -1-amino) -3-hydroxy-8-azaspiro [4.5] decane which was used directly in the next reaction.
LC-MS:m/z 171.0
Step two: (1R, 3R) -1- ((tert-butoxycarbonyl) amino) -3-hydroxy-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester (C-1P-b)
Figure BDA0003608939860000422
The above (1R, 3R) -1-amino) -3-hydroxy-8-azaspiro [4.5]Decane (0.2 mmol) was dissolved in tetrahydrofuran solution (10 mL) and (Boc) was added 2 O (109mg, 0.5mmol) and DIPEA (516mg, 4.0mmol) were added thereto, and the reaction mixture was stirred at room temperature for 16 hours. After completion of the reaction, the reaction was quenched by addition of saturated ammonium chloride and then extracted with diethyl ether (5 × 50 mL), the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a crude product which was purified by silica gel chromatography (0 to 50% gradient of ethyl acetate/petroleum ether) to give (1r, 3r) -1- ((tert-butoxycarbonyl) amino) -3-hydroxy-8-azaspiro [4.5] o]Decane-8-carboxylic acid tert-butyl ester C-1P-b (60mg, two-step yield: 80%).
1 H NMR(400MHz,CDCl 3 )δ5.11(s,1H),4.36(s,1H),3.86-3.62(m,3H),2.94(t,J=11.7Hz,2H),2.20-2.08(m,1H),1.81(d,J=8.5Hz,1H),1.64-1.55(m,3H),1.49-1.42(m,3H),1.37(d,J=4.9Hz,18H)ppm;LC-MS:m/z 393.2[M+H] + .
Step three: (R) -1- ((tert-butoxycarbonyl) amino) -3-carbonyl-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000423
Tert-butyl (1R, 3R) -1- ((tert-butoxycarbonyl) amino) -3-hydroxy-8-azaspiro [4.5] decane-8-carboxylate (60mg, 0.16mmol) was dissolved in dichloromethane (5 mL) at 0 ℃ and Dess-Martin (76mg, 0.18mmol) was slowly added thereto at 0 ℃ and the reaction was allowed to stand at 0 ℃ for further reaction for 2 hours. After the reaction was complete, quench was added water (4 mL) and then extracted with dichloromethane (5 × 50 mL), the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a crude product which was purified by silica gel chromatography (0 to 80% gradient of ethyl acetate/petroleum ether) to give (R) -tert-butyl 1- ((tert-butoxycarbonyl) amino) -3-carbonyl-8-azaspiro [4.5] decane-8-carboxylate (30 mg, yield: 50%).
1 H NMR(400MHz,CDCl 3 )δ4.46(d,J=9.4Hz,1H),4.07(d,J=7.0Hz,1H),3.92(s,2H),2.73(t,J=12.7Hz,2H),2.63(dd,J=19.0,8.1Hz,1H),2.41(d,J=18.3Hz,1H),2.14-2.01(m,2H),1.68(td,J=12.9,4.6Hz,1H),1.59(d,J=4.8Hz,1H),1.39(d,J=2.5Hz,18H),1.26(d,J=2.9Hz,1H)ppm;LC-MS:m/z 369.1[M+H] + .
Step four: (R) -tert-butyl 1- ((tert-butoxycarbonyl) amino) -3, 3-difluoro-8-azaspiro [4.5] decane-8-carboxylate
Figure BDA0003608939860000431
Tert-butyl (R) -1- ((tert-butoxycarbonyl) amino) -3-carbonyl 8-azaspiro [4.5] decane-8-carboxylate (80mg, 0.22mmol) was dissolved in dichloromethane (10 mL), followed by addition of DeoxoFluor (162. Mu.L, 0.88 mmol), and the reaction was further heated and stirred at 50 ℃ for 48 hours. After completion of the reaction, the reaction solution was quenched with a saturated sodium bicarbonate solution at 0 ℃ and extracted with ethyl acetate (3X 20 mL), and the combined organic phases were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl (R) -1- ((tert-butoxycarbonyl) amino) -3, 3-difluoro-8-azaspiro [4.5] decane-8-carboxylate (46 mg, yield: 54%)
1 H NMR(400MHz,CDCl 3 )δ4.49(d,J=9.9Hz,1H),3.92(dd,J=24.5,15.9Hz,3H),2.87-2.68(m,2H),2.58-2.42(m,1H),2.21(td,J=17.7,16.8,9.6Hz,1H),2.06-1.86(m,2H),1.60(t,J=6.6Hz,1H),1.38(d,J=4.0Hz,21H)ppm;LC-MS:m/z 391.1[M+H] +
Step five: (R) -1-amino) -3, 3-difluoro-8-azaspiro [4.5] decane (C-1P)
Figure BDA0003608939860000432
To (R) -1- ((tert-butoxycarbonyl) amino) -3, 3-difluoro-8-azaspiro [4.5] at room temperature]1, 4-dioxane hydrochloride (4M, 1.2mmol,0.3 mL) was slowly added to a solution of t-butyl decane-8-carboxylate (46mg, 0.12mmol) in methanol (5 mL), and the reaction mixture was reacted at room temperature for 1 hour. Concentrating under reduced pressure to obtain (R) -1-amino) -3, 3-difluoro-8-azaspiro [4.5]Decane (C-1P). LC-MS m/z 191.1[ m ] +H] + .
Example 13: preparation of intermediate 1-methyl-8-azaspiro [4.5] decan-1-amine (C-2A)
The method comprises the following steps: 1-hydroxy-1-methyl-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0003608939860000433
A tetrahydrofuran solution of methyl magnesium bromide (1M, 2.2mmol,2.2 mL) was slowly added dropwise to a solution of tert-butyl 1-carbonyl-8-azaspiro [4.5] decane-8-carboxylate (500mg, 2.0 mmol) in toluene (20 mL) at 0 ℃ and the reaction mixture was further stirred at 0 ℃ for 1 hour. After the reaction was complete, it was quenched by addition of saturated aqueous ammonium chloride solution and then extracted with ethyl acetate (3 × 20 mL), the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a crude product which was purified by silica gel chromatography (0 to 50% gradient of ethyl acetate/petroleum ether) to give tert-butyl 1-hydroxy-1-methyl-8-azaspiro [4.5] decane-8-carboxylate (410 mg, yield: 77%).
1 H NMR(400MHz,CDCl 3 )δ4.16-4.04(m,2H),3.12-2.56(m,2H),1.98-1.49(m,8H),1.49(s,9H),1.42-1.29(m,2H),1.16(s,3H)ppm;LC-MS:m/z 270.2[M+H] + .
Step two: n- (1-methyl-8-azaspiro [4.5] decan-1-yl) acetamide
Figure BDA0003608939860000441
Tert-butyl 1-hydroxy-1-methyl-8-azaspiro [4.5] decane-8-carboxylate (410mg, 1.52mmol) was dissolved in acetonitrile (1.82 mL), concentrated sulfuric acid (1.56 mL) was added at 0 ℃, and the reaction solution was stirred at room temperature for 16 hours. After completion of the reaction, the reaction solution was poured into ice water, followed by basification with aqueous NaOH (50%) solution to pH =12, followed by extraction with ethyl acetate (3 × 20 mL), and the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a crude product of N- (1-methyl-8-azaspiro [4.5] decan-1-yl) acetamide (260 mg, yield: 81%) which was directly used in the next reaction.
LC-MS:m/z 211.0[M+H] + .
Step three: 1-methyl-8-azaspiro [4.5] decan-1-amine (C-2A)
Figure BDA0003608939860000442
The crude N- (1-methyl-8-azaspiro [4.5] decan-1-yl) acetamide (260mg, 1.23mmol) was dissolved in 6M hydrochloric acid solution (5 mL) at room temperature, and the reaction mixture was reacted at 120 ℃ for 4 hours under microwave heating. After completion, the reaction solution was concentrated under reduced pressure and then lyophilized to give a crude product, 1-methyl-8-azaspiro [4.5] decan-1-amine C-2A (200 mg, yield: 97%), which was used in the next reaction without purification.
LC-MS:m/z 169.1[M+H] + .
Example 14: preparation of intermediate tert-butyl (4-ethylpiperidin-4-yl) carbamate (C-3A)
The method comprises the following steps: 1-benzyl-4-ethylpiperidine-4-carboxylic acid methyl ester
Figure BDA0003608939860000443
To a solution of methyl 1-benzylpiperidine-4-carboxylate (1g, 4.3mmol) in tetrahydrofuran (10 mL) was slowly added dropwise a solution of LDA in tetrahydrofuran (1M, 5.1mmol, 5.1mL) under nitrogen at-78 deg.C, the reaction mixture was stirred at-78 deg.C for 1 hour, iodoethane (795mg, 5.1mmol) in tetrahydrofuran (1 mL) was then added slowly, and the reaction mixture was reacted at-78 deg.C for 4 hours. After completion of the reaction, saturated aqueous ammonium chloride solution was added to quench, followed by extraction with ethyl acetate (3 × 30 mL), and the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a crude product which was purified by silica gel chromatography (0 to 50% gradient of ethyl acetate/petroleum ether) to give methyl 1-benzyl-4-ethylpiperidine-4-carboxylate (800 mg, yield: 71%).
LC-MS:m/z 262.2[M+H] + .
Step two: 1-benzyl-4-ethylpiperidine-4-carboxylic acid
Figure BDA0003608939860000444
Methyl 1-benzyl-4-ethylpiperidine-4-carboxylate (800mg, 3.23mmol) was dissolved in an aqueous ethanol solution (ethanol: water =4:1, 20ml), and then NaOH (517mg, 12.9mmol) was added, and the reaction solution was stirred at room temperature for 16 hours. After completion of the reaction, the reaction solution was concentrated under reduced pressure, then acidified with 1N hydrochloric acid to pH =3, then extracted with ethyl acetate (3x30 mL), and the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 1-benzyl-4-ethylpiperidine-4-carboxylic acid (720 mg, yield: 90%) which was used directly in the next reaction.
LC-MS:m/z 248.2[M+H] + .
Step three: 1-methyl-8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000451
1-benzyl-4-ethylpiperidine-4-carboxylic acid (250mg, 1.0 mmol) was dissolved in tert-butanol (5 mL) at room temperature, and then triethylamine (306mg, 3.0 mmol) and diphenylphosphorylazide (330mg, 2.0 mmol) were added, followed by heating and refluxing of the reaction solution for 8 hours. After the reaction was completed, water was added to quench the reaction, followed by extraction with ethyl acetate (3X 30 mL), the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a crude product which was purified by silica gel chromatography (gradient of 0 to 70% ethyl acetate/petroleum ether) to give 1-methyl-8-azaspiro [4.5] decan-1-amine (160 mg, yield: 50%).
LC-MS:m/z 319.2[M+H] + .
Step four: (4-ethylpiperidin-4-yl) carbamic acid tert-butyl ester
Figure BDA0003608939860000452
1-methyl-8-azaspiro [4.5] decan-1-amine (160mg, 0.5mmol) was dissolved in 10mL of methanol at room temperature, and Pd/C (1695g, 10%) was added to the solution to react for 16 hours under a hydrogen atmosphere. Celite was filtered, then washed with ethyl acetate, and concentrated under reduced pressure to give tert-butyl (4-ethylpiperidin-4-yl) carbamate (110 mg, yield: 95%). The product can be used directly in the next reaction.
LC-MS:m/z 229.2[M+H] + .
Step five: (4-ethylpiperidin-4-yl) carbamic acid tert-butyl ester (C-3A)
Figure BDA0003608939860000453
(4-ethylpiperidin-4-yl) carbamic acid tert-butyl ester (110mg, 0.5mmol) was dissolved in methanol (3 mL) at room temperature, and then 1, 4-dioxane solution (4N, 5mmol, 1.1Ml) hydrochloride was added to the reaction solution, stirred at room temperature for 2 hours, and concentrated under reduced pressure to give a product, tert-butyl (4-ethylpiperidin-4-yl) carbamate (C-3A) (50 mg, yield: 95%), which was used directly in the next reaction.
LC-MS:m/z 128.2[M+H] + .
Example 15: preparation of intermediate t-butyl ((4-methylpiperidin-4-yl) methyl) carbamate (C-4A)
The method comprises the following steps: 1-benzoyl-4-methylpiperidine-4-carbonitrile
Figure BDA0003608939860000454
4-methylpiperidine-4-carbonitrile (496mg, 4mmol), DCM (10 mL) and triethylamine (611mg, 6mmol) were sequentially added to a 100mL dry single-neck flask under nitrogen, followed by slow addition of benzoyl chloride (670mg, 4.8mmol) at room temperature. The mixture was stirred at room temperature for a further 1 hour and the reaction was monitored by TLC until the starting material was reacted. After quenching the reaction with 1N HCl solution, dichloromethane (3X 20 mL) was extracted and the combined organic phases were Na 2 SO 4 Drying, the filtrate was concentrated under reduced pressure and purified by column silica gel chromatography (0 to 40% gradient of ethyl acetate/petroleum ether) to give 1-benzoyl-4-methylpiperidine-4-carbonitrile (650 mg, yield: 70.72%).
LC-MS:m/z 229[M+H] + .
Step two: 1-benzoyl- ((4-methylpiperidin-4-yl) methyl) carbamic acid tert-butyl ester
Figure BDA0003608939860000461
To a 100mL dry flask, 1-benzoyl-4-methylpiperidine-4-carbonitrile (650mg, 2.85mmol), nickel chloride hexahydrate (135mg, 0.67mmol), di-tert-butyl dicarbonate (1.86g, 8.54mmol) and methyl methacrylate were added in this order under nitrogen at 0 deg.CAlcohol (12 mL), and to this was added sodium borohydride (754mg, 20mmol). The reaction was then stirred at room temperature for 12 hours and monitored by TLC until the starting material was reacted. After completion of the reaction, the reaction was concentrated and extracted with dichloromethane (3X 20 mL), and the combined organic phases were extracted with Na 2 SO 4 Drying, concentration of the filtrate under reduced pressure and purification by column silica gel chromatography (0 to 40% gradient of ethyl acetate/petroleum ether) gave tert-butyl 1-benzoyl- ((4-methylpiperidin-4-yl) methyl) carbamate (620 mg, yield: 65.50%)
LC-MS:m/z 333[M+H] + .
Step three: ((4-methylpiperidin-4-yl) methyl) carbamic acid tert-butyl ester (C-4A)
Figure BDA0003608939860000462
To a 100mL one-neck flask were added tert-butyl ((1-benzoyl-4-methylpiperidin-4-yl) methyl) carbamate (620mg, 1.87mmol), ethanol (8 mL) and 7N NaOH (2 mL) in this order, the mixture was heated to 90 ℃ under nitrogen and stirred for 8 hours, after the mixture was cooled to room temperature, the mixture was filtered, diluted with water and extracted with ethyl acetate (3X 20 mL), and the combined organic phases were extracted with Na 2 SO 4 Drying, the filtrate was concentrated under reduced pressure and purified by column silica gel chromatography (0 to 80% gradient of ethyl acetate/petroleum ether) to give ((4-methylpiperidin-4-yl) methyl) carbamic acid tert-butyl ester C-4A (300 mg, yield: 70.5%).
1 H NMR(400MHz,DMSO-d 6 )δ3.97(q,J=7.0Hz,2H),2.80(d,J=6.4Hz,2H),2.65(d,J=30.3Hz,2H),1.38(s,9H),1.27(dd,J=16.2,7.0Hz,2H),1.10(d,J=12.8Hz,2H),0.81(s,3H)ppm;LC-MS:m/z 229[M+H] + .
EXAMPLE 16 preparation of intermediate tert-butyl ((4-phenylpiperidin-4-yl) methyl) carbamate (C-4B)
The method comprises the following steps: 4-cyano-4-phenylpiperidine-1-carboxylic acid tert-butyl ester
Figure BDA0003608939860000463
To a solution of tert-butyl (2-chloroethyl) carbamate (2g, 8.26mmol) and 2-phenylacetonitrile (968mg, 8.26mmol) in anhydrous DMF (20 mL) at 0 deg.C was added NaH (60% dispersed in mineral oil, 1.6g, 41.3mmol) in portions. The reaction mixture was heated at 60 ℃ for 16 hours. After completion of the reaction, the reaction mixture was quenched with ice water (30 mL) and then extracted with 3X50 mL). The combined organic layers were washed with saturated brine (2 × 50 mL), then dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column silica gel chromatography (0 to 40% gradient of ethyl acetate/petroleum ether) to give tert-butyl 4-cyano-4-phenylpiperidine-1-carboxylate (500 mg, yield: 21%).
LCMS:m/z 187.2[M-100] + .
Step two: 4- (aminomethyl) -4-phenylpiperidine-1-carboxylic acid tert-butyl ester
Figure BDA0003608939860000471
Tert-butyl 4-cyano-4-phenylpiperidine-1-carboxylate (0.5g, 1.75mmol) was dissolved in 20mL of methanol, palladium on carbon (50 mg) was added thereto, and the reaction mixture was reacted under hydrogen for 16 hours. After completion of the reaction, the reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl 4- (aminomethyl) -4-phenylpiperidine-1-carboxylate (0.4 g, yield: 80%).
1 H NMR(400MHz,CDCl 3 )δ7.38(t,J=7.6Hz,2H),7.30(d,J=7.5Hz,2H),7.24(d,J=7.2Hz,1H),3.75(d,J=7.8Hz,2H),3.04(t,J=11.2Hz,2H),2.58(brs,2H),2.21(d,J=13.9Hz,2H),1.76-1.61(m,2H),1.44(s,9H)ppm;LC-MS:m/z 191.0[M-100] + .
Step two: (4-phenylpiperidin-4-yl) methylamine (C-4B)
Figure BDA0003608939860000472
After 4- (aminomethyl) -4-phenylpiperidine-1-carboxylic acid tert-butyl ester (0.4 g, 1.37mmol) was dissolved in 10mL of methanol, 1, 4-dioxane solution (4M, 13.7 mmol) of hydrochloric acid was added thereto at room temperature, and the reaction mixture was reacted for 2 hours at room temperature. After completion of the reaction, concentration was carried out under reduced pressure to give (4-phenylpiperidin-4-yl) methylamine C-4B (0.25 g, yield: 95%) as a crude product which was used directly in the next reaction.
LC-MS:m/z 191.2[M+H] + .
EXAMPLE 17 preparation of intermediate sodium 6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridine-4-thiol (F-1A)
The method comprises the following steps: (6-Chloropyridin-2-yl) carbamic acid tert-butyl ester
Figure BDA0003608939860000473
6-chloropyridin-2-amine (8g, 62.2mmol) and THF (80 mL) were added to a dry 250mL three-necked flask under nitrogen, the mixture was stirred at 0 ℃ for 10 minutes, then NaHDMS (124.4 mL,1.0M in THF) was added, then a solution of di-tert-butyl dicarbonate (16.3g, 74.7 mmol) in tetrahydrofuran (50 mL) was slowly added while maintaining the system at 0 ℃ and the reaction was continued at 0 ℃ for 4 hours. After the reaction is finished, H is added 2 O (40 mL) and then extracted with EtOAc (3 × 100 mL). The combined organic phases were washed with MgSO 4 The residue obtained by drying, filtration and concentration under reduced pressure was purified by silica gel chromatography (gradient 0 to 10% ethyl acetate/petroleum ether) to give tert-butyl (6-chloropyridin-2-yl) carbamate (7 g, yield: 49%).
1 H NMR(400MHz,DMSO-d6)δ10.04(s,1H),7.79-7.58(m,2H),7.02(dd,J=5.5,2.9Hz,1H),1.38(s,9H)ppm;LCMS:m/z 288.1[M+H] + .
Step two: (5, 6-dichloropyridin-2-yl) carbamic acid tert-butyl ester
Figure BDA0003608939860000474
To a dry 100mL round bottom flask was added tert-butyl (6-chloropyridin-2-yl) carbamate (7 g,30.6 mmol) and N, N-dimethylformamide (50 mL), the mixture was stirred at room temperature for 10 minutes, then N-chlorosuccinimide (4.50g, 33.67mmol) was added and the mixture was stirred at room temperatureThe reaction was carried out at 100 ℃ for 4 hours. After the reaction is finished, the temperature of the reaction solution is reduced to room temperature, and H is added 2 O (50 mL) was then extracted with ethyl acetate (3 × 80 mL) and washed with saturated aqueous lithium chloride (2x40 mL). The organic phase was washed with MgSO 4 Drying, filtration and concentration under reduced pressure gave a residue which was purified by silica gel chromatography (0 to 5% gradient of ethyl acetate/petroleum ether) to give tert-butyl (5, 6-dichloropyridin-2-yl) carbamate (5.3 g, yield: 65.8%).
1 H NMR(400MHz,CDCl 3 )δ7.86(d,J=8.7Hz,1H),7.69(d,J=8.7Hz,1H),7.24(s,1H),1.51(s,9H);LCMS:m/z 207.1[M-55] + .
Step three: (5, 6-dichloro-4-iodopyridin-2-yl) carbamic acid tert-butyl ester
Figure BDA0003608939860000481
To a dry 100mL round bottom flask was added under nitrogen (5, 6-dichloropyridin-2-yl) carbamic acid tert-butyl ester (5.3g, 20.14mmol) and tetrahydrofuran (50 mL), n-butyllithium (44.3mmol, 2.5M in THF) was added slowly dropwise at-78 deg.C, and the reaction was stirred at this temperature for 1 hour. A solution of iodine (3.07g, 24.17mmol) in tetrahydrofuran (20 mL) was then added slowly dropwise, and the reaction was also continued at-78 deg.C for 3 hours. After the reaction is finished, H is added 2 O (50 mL), followed by extraction with EtOAc (3 × 80 mL). The combined organic phases were washed with MgSO 4 Dried, filtered and concentrated under reduced pressure, the residue obtained is purified by chromatography on silica gel (gradient 0 to 5% ethyl acetate/petroleum ether) to give tert-butyl (5, 6-dichloro-4-iodopyridin-2-yl) carbamate (4.3 g, yield: 55%).
1 H NMR(400MHz,DMSO-d6)δ10.44(s,1H),8.36(s,1H),1.46(s,9H)ppm;LCMS:m/z 334.1[M-55] + .
Step four: methyl 3- ((6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) propionate
Figure BDA0003608939860000482
To a dry 100mL round bottom flask was added, under nitrogen, (5, 6-dichloro-4-iodopyridin-2-yl) carbamic acid tert-butyl ester (3.2 g, 8.22mmol), palladium acetate (92mg, 0.41mmol), xantphos (285mg, 0.49mmol), DIPEA (2.12g, 16.46mmol) and 1, 4-dioxane (30 mL) in that order. The reaction mixture was heated and stirred at 100 ℃ for 2 hours. Filtration and concentration under reduced pressure gave a residue which was purified by silica gel chromatography (0 to 30% gradient of ethyl acetate/petroleum ether) to give methyl 3- ((6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) propanoate (3 g, yield: 96%).
1 H NMR(400MHz,DMSO-d6)δ10.25(s,1H),7.73(s,1H),3.64(s,3H),3.26(t,J=6.9Hz,2H),2.82(t,J=6.9Hz,2H),1.46(s,9H)ppm;LCMS:m/z 326.3[M-55] + .
Step five: sodium 6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridine-4-thiol (F-1A)
Figure BDA0003608939860000483
To a dry 100mL round bottom flask were added methyl 3- ((6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) propionate and tetrahydrofuran (30 mL) in that order, then a solution of sodium ethoxide in ethanol (21%, 6 mL) was added slowly dropwise at room temperature and the reaction stirred at room temperature for 1 hour. After concentration under reduced pressure, methylene chloride (10 mL) was added to precipitate a large amount of a brown solid, which was filtered, washed with methylene chloride, and dried to obtain sodium 6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-thiolate F-1A (2.1 g, yield: 84%).
1 H NMR(400MHz,DMSO-d6)δ9.05(s,1H),7.61(s,1H),1.41(s,9H)ppm;LCMS:m/z 262.2[M-55] + Using similar starting intermediates, the following intermediates F-1B, F-1C, F-1D, F-1E, F-1F, F-1G, F-1H, F-1I, F-1J, F-1K, F-1L, F-1M, F-1N, F-1O, F-1P were obtained according to the synthesis of example 17.
Figure BDA0003608939860000491
Figure BDA0003608939860000501
EXAMPLE 18 preparation of intermediate sodium 3-chloro-2-methylpyridine-4-thiolate (F-1Q)
The method comprises the following steps: 3- ((3-chloro-2-methylpyridin-4-yl) thio) propanoic acid methyl ester
The intermediate methyl 3- ((2, 3-dichloropyridin-4-yl) thio) propanoate obtained during the synthesis of intermediate F-1G was used in the following reaction.
Figure BDA0003608939860000502
To a dry 100mL round bottom flask was added methyl 3- ((2, 3-dichloropyridin-4-yl) thio) propanoate (500mg, 1.88mmol), pd (PPh) in that order under a nitrogen blanket 3 ) 4 (217mg, 0.188mmol), trimethylcyclotriboroxyloxane (354mg, 2.82mmol), potassium carbonate (389mg, 2.82mmol) and 1, 4-dioxane (10 mL). The reaction mixture was heated and stirred at 100 ℃ for 6 hours under nitrogen. The resulting residue was filtered and concentrated under reduced pressure to give a residue which was purified by silica gel chromatography (0 to 40% gradient of ethyl acetate/petroleum ether) to give methyl 3- ((3-chloro-2-methylpyridin-4-yl) thio) propionate (320 mg, yield: 69%).
Step two: 3-chloro-2-methylpyridine-4-thiol sodium (F-1Q)
Figure BDA0003608939860000503
To a dry 100mL round bottom flask were added methyl 3- ((3-chloro-2-methylpyridin-4-yl) thio) propionate (320mg, 1.30mmol) and tetrahydrofuran (10 mL) in that order, then a solution of sodium ethoxide in ethanol (21%, 2 mL) was added slowly dropwise at room temperature and the reaction stirred at room temperature for 1 hour. After concentration under reduced pressure, methylene chloride (10 mL) was added to precipitate a large amount of brown solid, which was filtered, washed with methylene chloride, and dried to obtain sodium 3-chloro-2-methylpyridin-4-thiolate F-1Q (200 mg, yield: 85%).
1 H NMR(400MHz,DMSO-d6)δ7.37(d,J=4.8Hz,1H),6.97(d,J=4.8Hz,1H),2.31(s,3H)ppm;LCMS:m/z 160.0[M+H] + .
Following the synthetic procedure of example 18, using similar intermediate starting materials, the reaction gave the following intermediate F-1R.
Figure BDA0003608939860000511
Example 19: preparation of intermediate sodium 6- ((tert-butoxycarbonyl) amino) -3-chloro-2-methylpyridine-4-thiol (F-1S)
The method comprises the following steps: 3- ((6- ((tert-Butoxycarbonyl) amino) -3-chloro-2-methylpyridin-4-yl) thio) propanoic acid methyl ester
The intermediate methyl 3- ((6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) propanoate obtained during the synthesis of intermediate F-1A was used in the following reaction.
Figure BDA0003608939860000512
To a dry 100mL round bottom flask was added methyl 3- ((6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) propanoate (600mg, 1.57mmol), [1,1' -bis (tert-butylphosphino) ferrocene dichloropalladium (103mg, 0.157mmol), trimethyltriboroxine (301mg, 2.4 mmol), potassium carbonate (331mg, 2.4 mmol), 1, 4-dioxane (10 mL) and water (1 mL) in that order under nitrogen. The reaction mixture was heated and stirred at 100 ℃ for 6 hours under nitrogen. The residue obtained was filtered and concentrated under reduced pressure and purified by silica gel chromatography (0 to 40% gradient of ethyl acetate/petroleum ether) to give methyl 3- ((6- ((tert-butoxycarbonyl) amino) -3-chloro-2-methylpyridin-4-yl) thio) propanoate (420 mg, yield: 74%).
1 H NMR(400MHz,DMSO-d6)δ9.90(s,1H),7.64(s,1H),3.64(s,3H),3.21(t,J=6.9Hz,2H),2.80(t,J=6.9Hz,2H),1.46(s,9H)ppm;LCMS:m/z 361.1[M+H] + .
Step two: sodium 6- ((tert-butoxycarbonyl) amino) -3-chloro-2-methylpyridine-4-thiol (F-1S)
Figure BDA0003608939860000513
To a dry 100mL round bottom flask were added methyl 3- ((6- ((tert-butoxycarbonyl) amino) -3-chloro-2-methylpyridin-4-yl) thio) propanoate (420mg, 1.17mmol) and tetrahydrofuran (10 mL) in this order, followed by slowly adding an ethanol solution of sodium ethoxide (21%, 2 mL) dropwise at room temperature, and the reaction was stirred at room temperature for 1 hour. Concentrated under reduced pressure, followed by addition of dichloromethane (10 mL) to precipitate a large amount of brown solid, which was filtered, washed with dichloromethane, and dried to give sodium 6- ((tert-butoxycarbonyl) amino) -3-chloro-2-methylpyridin-4-thiol F-1S (320 mg, yield: 92%).
1 H NMR(400MHz,DMSO-d 6 )δ9.87(s,1H),7.63(s,1H),3.64(s,3H),1.46(s,9H)ppm;LCMS:m/z275.0[M+H] + .
EXAMPLE 20 preparation of intermediate 3-amino-2-chlorobenzenethiol hydrochloride (F-2A)
The method comprises the following steps: 2-chloro-3-aminothiophenol tert-butyl ester
Figure BDA0003608939860000521
To a dry 100mL round-bottomed flask was added 2-chloro-3-fluoroaniline (5g, 34.3 mmol) and N-methylpyrrolidone (50 mL) in this order under a nitrogen blanket, followed by 2-methylpropane-2-thiol (8.66g, 96.04mmol) and cesium carbonate (22.36g, 68.6 mmol), and the reaction mixture was heated and stirred at 120 ℃ for 16 hours. After cooling to room temperature, the reaction solution was diluted with 60mL of ethyl acetate, washed with a saturated aqueous lithium chloride solution (30 mL), water (30 mL) and a saturated aqueous sodium chloride solution (30 mL) in this order, then dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl 2-chloro-3-aminophenethiolate (6.04 g, yield: 82%).
LCMS:m/z 216.1[M+H] + .
Step two: 3-amino-2-chlorobenzenethiol hydrochloride (F-2A)
Figure BDA0003608939860000522
To a dry 100mL round bottom flask was added tert-butyl 2-chloro-3-aminothiophenol (6.04g, 28mmol) and concentrated hydrochloric acid (50 mL) and the reaction mixture was heated with stirring at 45 ℃ for 8 hours. After naturally cooled to room temperature, the reaction solution was further cooled to 0 ℃ to precipitate a large amount of white solid, which was then filtered and washed with concentrated hydrochloric acid and petroleum ether in this order to give 3-amino-2-chlorobenzenethiol hydrochloride F-2A (4.9 g, yield: 90%).
LCMS:m/z 160.0[M+H] + .
EXAMPLE 21 preparation of the Compound 1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -4-methylpiperidin-4-amine
The method comprises the following steps: (1- (8-Iodoimidazo [1,2-c ] pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester
Figure BDA0003608939860000523
To a dry 50mL single-neck flask, 5-chloro-8-iodoimidazo [1,2-c ] was added sequentially under nitrogen]Pyrimidine E1 (50mg, 0.18mmol), (4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (77mg, 0.36mmol), DIEA (46mg, 0.36mmol) and NMP (5 mL), followed by stirring at 90 ℃ for 2 hours. After completion of the reaction, the obtained residue was poured into water (10 mL), and stirred at room temperature for 5 minutes. Then extracted with ethyl acetate (3x50 mL) and the combined organic phases are over MgSO 4 The residue obtained after drying, filtration and concentration under reduced pressure is purified by silica gel chromatography (0 to 80% gradient of ethyl acetate/petroleum ether) to give a pale yellow solid (1- (8-iodoimidazo [1,2-c ] s)]Pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (70 mg, yield: 43%)
1 H NMR(400MHz,DMSO-d 6 )δ8.02(s,1H),7.89(d,J=1.5Hz,1H),7.62(d,J=1.4Hz,1H),3.49(d,J=13.4Hz,2H),3.21(ddd,J=13.3,10.8,2.6Hz,2H),2.18(d,J=13.9Hz,2H),1.65(ddd,J=14.1,10.7,3.8Hz,2H),1.40(s,9H),1.28(s,3H)ppm;LCMS:m/z 458.1[M+H] + .
Step two: (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester
Figure BDA0003608939860000531
To a dry 50mL three-necked flask was added (1- (8-iodoimidazo [1,2-c ] in this order)]Pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (70mg, 0.15mmol), cuprous iodide (3mg, 0.015mmol), 1, 10-phenanthroline (6mg, 0.030mmol), 2, 3-dichlorothiophenol (32mg, 0.18mmol), potassium phosphate (66mg, 0.30mmol), and 5mL of dioxane. The mixture was heated under nitrogen for 3 hours. After the reaction is finished, saturated NH is added 4 Cl solution (10 mL). Then extracted with ethyl acetate (3x50 mL). The combined organic phases are treated with Na 2 SO 4 Drying, filtering, concentrating the filtrate under reduced pressure, and purifying the resulting residue by silica gel chromatography (0 to 60% gradient of ethyl acetate/petroleum ether) to give (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1, 2-c) as a pale yellow solid]Pyrimidin-5-) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (30 mg, yield: 39%).
LC-MS:m/z 508.1[M+H] + .
Step three: 1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -4-methylpiperidin-4-amine
Figure BDA0003608939860000532
To a dry 50mL round bottom flask were added tert-butyl (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-) -4-methylpiperidin-4-yl) carbamate (30mg, 0.059 mmol) and a solution of hydrochloric acid in 1, 4-dioxane (7M, 5mL) in this order, and reacted at room temperature for 1 hour. The reaction solution was concentrated under reduced pressure, and the obtained crude product was purified by high performance liquid chromatography to give the product 1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -4-methylpiperidine-4-amine (15 mg, yield: 62%).
1 H NMR(400MHz,DMSO-d6)δ8.02(s,1H),7.84(d,J=1.3Hz,1H),7.55(d,J=1.2Hz,1H),7.41(dd,J=8.0,1.2Hz,1H),7.12(t,J=8.0Hz,1H),6.69(dd,J=8.1,1.2Hz,1H),3.60(t,J=4.2Hz,4H),1.73-1.60(m,4H),1.19(s,3H)ppm;LC-MS:m/z 408.1[M+H] + .
Using the synthesis method of example 21, the following compounds can be synthesized:
example 22- ((2, 3-dichlorophenyl) thio) -5- (1, 8-diazaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidine
Figure BDA0003608939860000533
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.89(s,1H),7.57(s,1H),7.41(d,J=8.1Hz,1H),7.12(t,J=8.0Hz,1H),6.72(d,J=8.1Hz,1H),3.79(ddd,J=13.9,7.1,3.7Hz,2H),3.49(ddd,J=12.8,8.0,3.4Hz,2H),3.19(t,J=6.9Hz,2H),2.04(ddd,J=12.3,8.1,3.5Hz,2H),1.93(dt,J=20.5,6.5Hz,6H)ppm;LC-MS:m/z434.1[M+H] + .
EXAMPLE 23 preparation of the Compound 8- ((2, 3-dichlorophenyl) thio) -5- (piperidin-1-yl) imidazo [1,2-c ] pyrimidine
Figure BDA0003608939860000541
To a dry 50mL single neck flask was added 5-chloro-8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] in sequence]Pyrimidine B1 (50mg, 0.15mmol), piperidine (20mg, 0.23mmol), DIEA (39mg, 0.3mmol), and NMP (5 mL), followed by stirring at 90 ℃ for 2 hours. After completion of the reaction, the obtained residue was poured into water (100 mL), and stirred at room temperature for 5 minutes. Then extracted with ethyl acetate (3X 50 mL) and the combined organic phases were washed with MgSO 4 The residue obtained by drying, filtration and concentration under reduced pressure is purified by chromatography on silica gel (gradient 0 to 80% ethyl acetate/petroleum ether) and by high performance liquid chromatography to give the product ((1- (8- ((2, 3-bis)Chlorophenyl) thio) imidazo [1,2-c]Pyrimidin-5-yl) pyrrolidin-3-yl) methyl) carbamic acid tert-butyl ester (20 mg, yield: 35%)
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.83(d,J=1.4Hz,1H),7.55(d,J=1.4Hz,1H),7.41(dd,J=8.0,1.3Hz,1H),7.12(t,J=8.0Hz,1H),6.69(dd,J=8.1,1.3Hz,1H),3.50(d,J=5.6Hz,4H),1.79-1.63(m,6H)ppm;LC-MS:m/z 378.7[M+H] + .
Using the synthesis of example 23, the following compounds can be synthesized:
example 24:8- ((2, 3-dichlorophenyl) thio) -5- (3, 5-dimethylpiperazin-1-yl) imidazo [1,2-c ] pyrimidine
Figure BDA0003608939860000542
1 H NMR(400MHz,DMSO-d6)δ8.23(s,1H),8.03(s,1H),7.92(d,J=1.2Hz,1H),7.55(d,J=1.2Hz,1H),7.44-7.37(m,1H),7.12(t,J=8.1Hz,1H),6.69(dd,J=8.1,1.1Hz,1H),3.89(d,J=12.0Hz,2H),3.02(d,J=6.6Hz,2H),2.72-2.59(m,2H),1.05(t,J=6.0Hz,7H)ppm;LC-MS:m/z 407.7[M+H] + .
Example 25:8- ((2, 3-dichlorophenyl) thio) -5- (4-methylpiperazin-1-yl) imidazo [1,2-c ] pyrimidine
Figure BDA0003608939860000543
1 H NMR(400MHz,DMSO-d6)δ8.20(s,1H),8.04(s,1H),7.91(d,J=1.4Hz,1H),7.57(d,J=1.4Hz,1H),7.41(dd,J=8.0,1.2Hz,1H),7.12(t,J=8.0Hz,1H),6.70(dd,J=8.1,1.2Hz,1H),3.59-3.50(m,4H),2.59-2.53(m,4H),2.27(s,3H)ppm;LC-MS:m/z 393.8[M+H] + .
Example 26: (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -4-phenylpiperidin-4-yl) methylamine
Figure BDA0003608939860000551
1 H NMR(400MHz,DMSO-d6)δ8.00(s,1H),7.88(s,1H),7.54(s,1H),7.50-7.33(m,5H),7.28(t,J=6.8Hz,1H),7.10(t,J=8.0Hz,1H),6.67(d,J=8.1Hz,1H),3.77(d,J=12.5Hz,2H),3.31-3.22(m,2H),3.09(s,1H),2.82(s,1H),2.32(s,2H),2.06(t,J=10.3Hz,2H);LC-MS:m/z 484.7[M+H] + .
Example 27: (R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-difluoro-8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000552
1 H NMR(400MHz,DMSO-d 6 )δ8.03(s,1H),7.81(d,J=1.5Hz,1H),7.57(d,J=1.4Hz,1H),7.42(dd,J=8.0,1.3Hz,1H),7.12(t,J=8.1Hz,1H),6.69(dd,J=8.1,1.4Hz,1H),3.93(t,J=14.9Hz,2H),3.31(d,J=19.3Hz,2H),3.28-3.16(m,3H),2.252.06(m,2H),1.90(dd,J=32.1,11.8Hz,2H),1.61-1.45(m,2H)ppm;LC-MS:m/z 484.1[M+H] + .
Example 28: (R) -3- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-azaspiro [5.5] undecan-7-amine
Figure BDA0003608939860000553
1 H NMR(400MHz,DMSO-d6)δ8.02(s,1H),7.82(d,J=1.2Hz,1H),7.56(d,J=1.2Hz,1H),7.41(dd,J=8.0,1.2Hz,1H),7.12(t,J=8.1Hz,1H),6.68(dd,J=8.1,1.2Hz,1H),3.80(d,J=4.8Hz,2H),3.33(dd,J=29.0,11.6Hz,2H),2.77(d,J=5.2Hz,1H),2.04(dd,J=30.0,14.7Hz,2H),1.85-1.12(m,10H)ppm;LC-MS:m/z 461.7[M+H] + .
Example 29:1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -4-ethylpiperidin-4-amine
Figure BDA0003608939860000554
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.88(d,J=1.5Hz,1H),7.58(d,J=1.4Hz,1H),7.42(dd,J=8.0,1.3Hz,1H),7.12(t,J=8.1Hz,1H),6.70(dd,J=8.1,1.4Hz,1H),3.71-3.64(m,4H),1.90-1.73(m,4H),1.69(q,J=7.3Hz,2H),0.93(t,J=7.5Hz,3H)ppm;LC-MS:m/z 422.1[M+H] + .
Example 30:8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -1-methyl-8-aza-spiro [4.5] decan-1-amine
Figure BDA0003608939860000561
1 H NMR(400MHz,DMSO-d6)δ8.33(s,1H),7.83(s,1H),7.56(s,1H),7.41(d,J=8.1Hz,1H),7.17-7.08(m,1H),6.69(d,J=8.2Hz,1H),4.08-3.93(m,2H),3.18-3.04(m,2H),3.04-2.94(m,1H),2.45(s,1H),1.90-1.29(m,8H),0.95(t,J=6.3Hz,3H)ppm;LC-MS:m/z 462.1[M+H] + .
Example 31: preparation of the compound 1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) pyrrolidin-3-amine
The method comprises the following steps: (tert-butyl 1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) pyrrolidin-3-yl) carbamate
Figure BDA0003608939860000562
Using the same procedure as in example 23, B1 was substituted with the corresponding amine to give tert-butyl (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) pyrrolidin-3-yl) carbamate.
LC-MS:m/z 424.1[M+H] + .
Step two: the compound 1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) pyrrolidin-3-amine
Figure BDA0003608939860000563
Using the same procedure as in step three of example 21, (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) pyrrolidin-3-yl) carbamic acid tert-butyl ester is deblocked to give the compound 1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) pyrrolidin-3-amine.
1 H NMR(400MHz,DMSO-d6)δ8.18(s,1H),7.93(d,J=1.1Hz,1H),7.46(s,1H),7.40(d,J=8.0Hz,1H),7.13(t,J=8.0Hz,1H),6.63(d,J=8.1Hz,1H),4.14(d,J=9.3Hz,1H),4.05(d,J=5.9Hz,1H),3.82(d,J=11.2Hz,3H),2.24(d,J=7.3Hz,1H),2.02(s,1H)ppm;LC-MS:m/z 379.9[M+H] + .
Using the synthesis of example 31, the following compounds can be synthesized:
example 32:1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) piperidin-4-amine
Figure BDA0003608939860000571
1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),8.03(s,1H),7.86(s,1H),7.57(s,1H),7.42(d,J=7.6Hz,1H),7.12(t,J=8.0Hz,1H),6.69(d,J=8.1Hz,1H),4.00(d,J=13.0Hz,2H),3.13(d,J=11.5Hz,2H),2.02-1.86(m,3H),1.62(dd,J=21.0,9.8Hz,2H),1.23(s,1H);LC-MS:m/z 394.7[M+H] + .
Example 33: (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) pyrrolidin-3-yl) methylamine
Figure BDA0003608939860000572
1 H NMR(400MHz,DMSO-d6)δ8.17(d,J=1.6Hz,1H),7.91(s,1H),7.45(d,J=1.5Hz,1H),7.39(d,J=7.7Hz,1H),7.12(t,J=8.0Hz,1H),6.63(d,J=8.1Hz,1H),4.04(ddd,J=11.5,8.9,5.5Hz,2H),3.94-3.86(m,1H),3.72(dd,J=10.8,7.6Hz,2H),2.93(d,J=7.2Hz,2H),2.16(dq,J=11.9,6.2Hz,1H),1.79(dq,J=12.3,8.3Hz,1H)ppm;LC-MS:m/z 394.1[M+H] + .
Example 34: (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -4-methylpiperidin-4-yl) methylamine
Figure BDA0003608939860000573
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.83(d,J=1.5Hz,1H),7.57(d,J=1.4Hz,1H),7.42(dd,J=8.0,1.3Hz,1H),7.12(t,J=8.1Hz,1H),6.69(dd,J=8.1,1.3Hz,1H),3.68(dt,J=13.7,4.6Hz,2H),3.44(td,J=9.6,4.8Hz,2H),2.76(s,2H),1.70(ddd,J=13.3,9.2,3.7Hz,2H),1.62-1.49(m,2H),1.08(s,3H)ppm;LC-MS:m/z 422.1[M+H] + .
Example 35: (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) piperidin-4-yl) methylamine
Figure BDA0003608939860000574
1 H NMR(400MHz,DMSO-d6)δ8.30(s,1H),8.03(s,1H),7.83(s,1H),7.57(s,1H),7.42(d,J=7.7Hz,1H),7.12(t,J=8.0Hz,1H),6.69(d,J=8.0Hz,1H),4.05(d,J=13.2Hz,2H),3.07(t,J=12.3Hz,2H),2.75(d,J=5.9Hz,2H),1.88(d,J=11.8Hz,3H),1.53-1.35(m,2H)ppm;LC-MS:m/z 409.8[M+H] + .
Example 36:2- (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) piperidin-4-yl) ethan-1-amine
Figure BDA0003608939860000581
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.85(d,J=1.2Hz,1H),7.56(d,J=1.2Hz,1H),7.41(dd,J=8.0,1.2Hz,1H),7.12(t,J=8.1Hz,1H),6.69(dd,J=8.1,1.2Hz,1H),4.02(d,J=12.9Hz,2H),3.03(t,J=12.0Hz,2H),2.87-2.78(m,2H),1.82(d,J=12.5Hz,2H),1.70(s,1H),1.55(dd,J=14.7,6.9Hz,2H),1.40(dd,J=21.6,11.5Hz,2H)ppm;LC-MS:m/z 422.7[M+H] + .
Example 37: synthesis of compound (S) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
The method comprises the following steps: (R) -N- ((S) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-yl) -2-methylpropane-2-sulfinamide
Figure BDA0003608939860000582
Using the same procedure as in example 23, B1 was substituted with the corresponding amine to give (R) -N- ((S) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-yl) -2-methylpropane-2-sulfinamide.
LC-MS:m/z 554.1[M+H] + .
Step two: (S) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000583
To a 50mL single-neck flask were added in this order, (R) -N- ((S) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-yl) -2-methylpropane-2-sulfinamide (70mg, 0.13mmol) and methanol (0.5 mL) under a nitrogen atmosphere, a solution of 1, 4-dioxane hydrochloride (0.05mL, 4M) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1 hour. After completion of the reaction, it was cooled to room temperature, filtered, and the residue obtained by concentration under reduced pressure was purified by high performance liquid preparative chromatography to give (S) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine (10 mg, yield: 17%).
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.83(d,J=1.5Hz,1H),7.57(d,J=1.4Hz,1H),7.42(dd,J=8.0,1.4Hz,1H),7.12(t,J=8.0Hz,1H),6.69(dd,J=8.1,1.4Hz,1H),4.00(dd,J=8.8,6.4Hz,1H),3.87-3.71(m,2H),3.66(d,J=8.5Hz,1H),3.29-3.27(m,1H),3.17(t,J=5.8Hz,2H),1.92-1.76(m,2H),1.57(dd,J=12.5,5.8Hz,2H),1.24(d,J=3.2Hz,1H)ppm;LC-MS:m/z 450.1[M+H] + .
Following the synthetic procedure of example 37, the following compounds can be synthesized:
example 38: (R) -1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) azepan-4-amine
Figure BDA0003608939860000591
1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),7.95(s,2H),7.48(s,1H),7.40(d,J=7.9Hz,1H),7.12(t,J=8.0Hz,1H),6.66(d,J=8.0Hz,1H),4.03-3.89(m,2H),3.81-3.61(m,2H),3.20(s,1H),2.15(s,1H),1.95(d,J=14.1Hz,4H),1.56(d,J=10.7Hz,1H)ppm;LC-MS:m/z 409.8[M+H] + .
Example 39: (R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000592
1 H NMR(400MHz,DMSO-d6)δ8.18(s,1H),8.03(s,1H),7.84(s,1H),7.56(s,1H),7.41(d,J=7.9Hz,1H),7.12(t,J=8.1Hz,1H),6.70(d,J=8.1Hz,1H),3.94(t,J=13.7Hz,2H),3.54(d,J=8.7Hz,1H),3.45(d,J=8.7Hz,1H),3.32(d,J=12.2Hz,2H),2.68(s,1H),1.98-1.88(m,2H),1.65(s,2H),1.02(s,3H),0.95(s,3H);LC-MS:m/z 479.7[M+H] + .
Example 40: (S) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000593
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.84(s,1H),7.56(s,1H),7.41(d,J=7.9Hz,1H),7.12(t,J=8.0Hz,1H),6.69(d,J=8.1Hz,1H),3.94(t,J=14.1Hz,2H),3.54(d,J=8.7Hz,1H),3.45(d,J=8.7Hz,1H),3.35-3.25(m,2H),2.69(s,1H),1.98-1.85(m,2H),1.71-1.59(m,2H),1.03(s,3H),0.95(s,3H);LC-MS:m/z 479.7[M+H] + .
Example 41: (1R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-methyl-8-aza-spiro [4.5] decan-1-amine
Figure BDA0003608939860000601
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.81(dd,J=7.4,1.6Hz,1H),7.56(d,J=1.6Hz,1H),7.41(dd,J=8.0,1.4Hz,1H),7.12(t,J=8.0Hz,1H),6.68(dd,J=8.1,1.4Hz,1H),3.95(d,J=11.9Hz,2H),3.12(t,J=12.0Hz,2H),2.89(d,J=5.8Hz,1H),2.08-1.44(m,9H),1.00(dd,J=31.0,6.6Hz,3H)ppm;LC-MS:m/z462.1[M+H] + .
Example 42: (1R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-8-aza-spiro [4.5] decan-1-amine
Figure BDA0003608939860000602
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.80(s,1H),7.56(s,1H),7.41(d,J=8.0Hz,1H),7.12(t,J=8.1Hz,1H),6.69(d,J=8.1Hz,1H),3.99-3.80(m,2H),3.24-2.98(m,3H),2.27-1.14(m,9H),1.04-0.95(m,3H)ppm;LC-MS:m/z 462.1[M+H] + .
Example 43: (4R) -4-amino-8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-2-ol
Figure BDA0003608939860000603
1 H NMR(400MHz,DMSO-d6)δ8.37(s,1H),8.04(s,1H),7.81(s,1H),7.57(s,1H),7.42(d,J=7.9Hz,1H),7.12(t,J=8.0Hz,1H),6.68(d,J=8.0Hz,1H),4.15(d,J=6.5Hz,1H),3.91(d,J=14.3Hz,2H),3.22(s,2H),2.93(t,J=7.3Hz,1H),2.27-2.20(m,1H),1.89-1.66(m,5H),1.54(dt,J=13.4,6.7Hz,1H),1.35(d,J=12.9Hz,1H)ppm;LC-MS:m/z 465.7[M+H] + .
Example 44: (3S, 4S) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000604
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.84(s,1H),7.56(s,1H),7.41(d,J=7.9Hz,1H),7.12(t,J=8.0Hz,1H),6.69(d,J=8.0Hz,1H),4.15-4.06(m,1H),3.73(s,2H),3.56(d,J=8.5Hz,1H),3.42-3.27(m,3H),3.05(d,J=4.8Hz,1H),1.91(dt,J=39.6,9.8Hz,2H),1.67(dd,J=25.7,13.7Hz,2H),1.12(d,J=6.4Hz,3H)ppm;LC-MS:m/z 463.7[M+H] + .
Example 45: (R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -1-oxa-8-aza-spiro [4.5] decan-4-amine
Figure BDA0003608939860000611
1 H NMR(400MHz,DMSO-d6)δ7.95(s,1H),7.60(s,1H),7.48(s,1H),7.20(d,J=6.4Hz,1H),6.95(t,J=6.4Hz,1H),6.70(d,J=6.4Hz,1H),4.00-3.85(m,4H),3.46(t,J=8.8Hz,2H),3.20(t,J=5.2Hz,1H),2.39-2.35(m,1H),1.94-1.89(m,2H),1.80-1.74(m,2H),1.68-1.65(m,1H)ppm;LCMS:m/z 450.1[M+H] + .
Example 46: synthesis of the compound (R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -N-methyl-8-aza-spiro [4.5] decan-1-amine
The method comprises the following steps: (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide
Figure BDA0003608939860000612
Using the same procedure as in example 23, B1 was substituted with an amine to give (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide (70 mg, yield: 80%)
LC-MS:m/z 552.1[M+H] + .
Step two: (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -N, 2-dimethylpropane-2-sulfinamide
Figure BDA0003608939860000613
To a dry 50mL round bottom flask, add (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1, 2-c) in sequence]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-methylpropane-2-sulfinamide (70mg, 0.13mmol) and DMF (5 mL). NaH (10.4 mg, 0.26mmol) was then added slowly at 0 deg.C, the mixture was stirred at this temperature for 10 minutes, and CH was then added slowly while maintaining 0 deg.C 3 I (28mg, 0.20mmol), and then stirred at room temperature for 2 hours. After completion of the reaction, the reaction was quenched by addition of water (10 mL) and extracted with ethyl acetate (3X 10 mL) and the combined organic phases were MgSO 4 The residue obtained is dried, filtered and concentrated under reduced pressure to give a light yellow solid (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1, 2-c) by chromatography on silica gel (0 to 80% gradient of ethyl acetate/petroleum ether)]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -N, 2-dimethylpropane-2-sulfinamide (50 mg, yield: 49%)
LC-MS:m/z 566.1[M+H] + .
Step three: (R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -N-methyl-8-aza-spiro [4.5] decan-1-amine
Figure BDA0003608939860000621
Sulfinylation of (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -N, 2-dimethylpropane-2-sulfinamide was carried out in the same manner as in the second step of example 37 to give (R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -N-methyl-8-aza-spiro [4.5] decan-1-amine.
1 H NMR(400MHz,DMSO-d6)δ8.30(s,1H),8.03(s,1H),7.82(s,1H),7.55(s,1H),7.41(d,J=8.0Hz,1H),7.12(t,J=8.0Hz,1H),6.69(d,J=8.1Hz,1H),3.87(d,J=13.2Hz,2H),3.22(dd,J=22.1,10.8Hz,2H),2.56(t,J=7.3Hz,1H),2.35(s,3H),1.89(ddd,J=33.1,16.7,7.8Hz,4H),1.72-1.33(m,6H)ppm;LC-MS:m/z461.7[M+H] + .
Example 47: (R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
The method comprises the following steps: (R) -N- ((R) -8- (8-iodoimidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfonamide
Figure BDA0003608939860000622
To a dry 50mL single neck flask was added 5-chloro-8-iodoimidazo [1,2-c ] in sequence]Pyrimidine E1 (50mg, 0.18mmol), (R) -2-methyl-N- ((R) -8-azaspiro [ 4.5%]Decan-1-yl) propane-2-sulfinamide (C-1A) (93mg, 0.36mmol), DIEA (46mg, 0.36mmol) and NMP (5 mL), and the reaction was stirred at 90 ℃ for 2 hours. After completion of the reaction, the obtained residue was poured into water (10 mL), and stirred at room temperature for 5 minutes. Then extracted with ethyl acetate (3X 20 mL) and the combined organic phases were MgSO 4 The residue obtained is dried, filtered and concentrated under reduced pressure and purified by silica gel chromatography (0 to 80% gradient of ethyl acetate/petroleum ether) to give (R) -N- ((R) -8- (8-iodoimidazo [1, 2-c) as a pale yellow solid]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-methylpropane-2-sulfonamide (80 mg, yieldRate: 88%)
1 H NMR(400MHz,DMSO-d6)δ8.02(s,1H),7.79(d,J=1.4Hz,1H),7.64(d,J=1.4Hz,1H),4.97(d,J=8.0Hz,1H),3.77-3.65(m,2H),3.23-3.13(m,1H),3.04(dd,J=30.0,11.5Hz,2H),2.03-1.78(m,4H),1.67-1.31(m,6H),1.13(s,9H)ppm;LC-MS:m/z 502.1[M+H] + .
Step two: (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide
Figure BDA0003608939860000631
To a dry 50mL three-necked flask was added (R) -N- ((R) -8- (8-iodoimidazo [1, 2-c) in order]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-methylpropane-2-sulfonamide (80mg, 0.16mmol), cuprous iodide (3mg, 0.016 mmol), 1, 10-phenanthroline (6 mg, 0.032mmol), 2, 3-dichlorothiophenol (34mg, 0.192mmol), potassium phosphate (68mg, 0.32mmol), and 10mL of dioxane solution. The mixture was heated under nitrogen for 3 hours. After the reaction is finished, saturated NH is added 4 Cl solution (50 mL). It was extracted with ethyl acetate (3 × 20 mL). The combined organic phases are washed with Na 2 SO 4 Drying, filtering, concentrating the filtrate under reduced pressure, and purifying the resulting residue by silica gel chromatography (0 to 10% gradient of methanol/ethyl acetate) to give (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1, 2-c) as a pale yellow solid]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-methylpropane-2-sulfinamide (60 mg, yield: 68%).
1 H NMR(400MHz,DMSO-d6)δ8.02(s,1H),7.76(d,J=1.4Hz,1H),7.56(d,J=1.4Hz,1H),7.40(dd,J=8.0,1.3Hz,1H),7.11(t,J=8.0Hz,1H),6.68(dd,J=8.1,1.3Hz,1H),5.00(d,J=8.1Hz,1H),3.94(dd,J=12.8,3.4Hz,2H),3.26-3.14(m,3H),2.08-1.84(m,4H),1.69-1.36(m,6H),1.14-1.11(m,9H)ppm;LC-MS:m/z552.1[M+H] + .
Step three: (R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000632
To a dry 50mL round bottom flask were added sequentially (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide (example 17-2) (60mg, 0.11mmol) and a solution of hydrochloric acid in 1, 4-dioxane (7M, 5 mL) and reacted at room temperature for 1 hour. The reaction liquid was distilled under reduced pressure, and the obtained crude product was purified by reverse phase high performance liquid chromatography to give the product (R) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine (25 mg, yield: 51%).
1 H NMR(400MHz,CD 3 OD-d4)δ8.54(s,2H),8.04(s,1H),7.81(s,1H),7.58(s,1H),7.33(dd,J=8.0,1.3Hz,1H),7.05(t,J=8.0Hz,1H),6.68(dd,J=8.1,1.3Hz,1H),4.03(t,J=14.7Hz,2H),3.38(s,2H),2.27(d,J=5.4Hz,1H),1.97-1.63(m,9H)ppm;LC-MS:m/z 448.1[M+H] + .
Following the synthesis of example 47, the following compounds may be synthesized:
example 48: (R) -7- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -7-aza-spiro [3.5] nonan-1-amine
Figure BDA0003608939860000633
1 H NMR(400MHz,DMSO-d6)δ8.34(s,1H),8.02(s,1H),7.82(d,J=1.6Hz,1H),7.55(s,1H),7.41(d,J=7.8Hz,1H),7.12(t,J=8.1Hz,1H),6.69(d,J=7.9Hz,1H),3.91(d,J=13.2Hz,1H),3.84-3.73(m,1H),3.18-3.08(m,3H),2.13(q,J=8.0Hz,1H),1.83(td,J=10.7,8.5,4.2Hz,2H),1.80-1.69(m,3H)ppm;LC-MS:m/z 434.1[M+H] + .
Example 49:7- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -7-aza-spiro [3.5] nonan-2-amine
Figure BDA0003608939860000641
1 H NMR(400MHz,DMSO-d6)δ8.01(s,1H),7.83(d,J=1.6Hz,1H),7.56(d,J=1.5Hz,1H),7.41(dd,J=8.0,1.4Hz,1H),7.12(t,J=8.0Hz,1H),6.69(dd,J=8.1,1.4Hz,1H),3.50(d,J=10.2Hz,2H),3.41(s,2H),2.71-2.64(m,1H),2.36-2.30(m,1H),2.21(s,2H),1.81(d,J=26.6Hz,5H)ppm;LC-MS:m/z 434.1[M+H] + .
Example 50: (7R) -2- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-aza-spiro [4.4] nonan-7-ylamine
Figure BDA0003608939860000642
1 H NMR(400MHz,DMSO-d 6 )δ8.36(brs,1H),8.18(s,1H),7.89(d,J=2.4Hz,1H),7.43(s,1H),7.38(d,J=6.0Hz,1H),7.11(t,J=6.4Hz,1H),6.64(d,J=6.0Hz,1H),3.96-3.74(m,4H),3.53-3.51(m,1H),2.00-1.60(m,8H)ppm;LCMS:m/z 434.2[M+H] + .
Example 51: (R) -8- (8- ((6-amino-3-chloro-2-methyl-pyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-dimethyl 1-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000643
1 H NMR(400MHz,DMSO-d6)δ8.17(s,2H),8.08(s,1H),7.90(s,1H),7.65(s,1H),5.67(s,1H),
4.09-3.93(m,2H),3.61(dd,J=14.2,9.1Hz,4H),3.10(d,J=5.4Hz,1H),2.35(d,J=15.8Hz,3H),2.03-1.71(m,4H),1.16(s,3H),1.07(s,3H)ppm;LCMS:m/z 474.1[M+H] + .
Example 52: (S) -8- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000651
1 H NMR(DMSO-d6)δ8.43(s,1H),8.02(s,1H),7.79-7.80(d,1H),7.56-7.57(d,1H),7.40-7.42(d,1H),7.11-7.15(t,1H),6.68-6.71(d,1H),3.88(t,2H),3.24(t,2H),3.01(t,1H),1.60-1.90(m,5H),1.30-1.59(m,5H)ppm;LC-MS:m/z 448.1[M+H] + .
Example 53: (R) -8- (8- ((2-chlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000652
1 H NMR(400MHz,DMSO-d6)δ7.97(s,1H),7.80(d,J=1.5Hz,1H),7.57(d,J=1.4Hz,1H),7.49(dd,J=7.7,1.6Hz,1H),7.15(dtd,J=19.6,7.4,1.6Hz,2H),6.76(dd,J=7.7,1.7Hz,1H),3.93-3.80(m,2H),3.21(td,J=11.2,2.7Hz,2H),3.00(s,1H),2.05-1.75(m,4H),1.75-1.35(m,6H)ppm;LCMS:m/z 414.1[M+H] + .
Example 54: (R) -8- (8- ((3-chlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000653
1 H NMR(CD 3 OD-d4)δ7.96(s,1H),7.78(s,1H),7.58(s,1H),7.23-7.27(m,3H),7.14-7.16(m,1H),3.83(m,2H),3.24(m,2H),2.89(m,1H),1.77-1.83(m,4H),1.51-1.62(m,2H),1.354-1.46(m,4H)ppm;LCMS:m/z414.1[M+H] + .
Example 55: (R) -8- (8- ((4-chlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000654
1 H NMR(400MHz,DMSO-d6)δ7.91(s,1H),7.79-7.75(m,1H),7.58(d,J=1.2Hz,1H),7.33(d,J=8.5Hz,2H),7.23(d,J=8.5Hz,2H),3.91-3.71(m,2H),3.18(t,J=12.3Hz,2H),3.00(s,1H),2.05-1.66(m,5H),1.64-1.34(m,5H)ppm;LCMS:m/z 414.1[M+H] + .
Example 56: (R) -8- (8- ((2, 6-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000661
1 H NMR(400MHz,DMSO-d6)δ7.75(s,1H),7.66(s,1H),7.64(s,1H),7.60(s,1H),7.51(t,J=8.1Hz,1H),7.09(s,1H),3.64(s,2H),3.04(q,J=12.4Hz,2H),2.74(t,J=7.2Hz,1H),1.89-1.74(m,4H),1.52(s,2H),1.42-1.24(m,4H)ppm;LCMS:m/z 450.0[M+H] + .
Example 57: (R) -8- (8- ((2, 4-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000662
1 H NMR(400MHz,DMSO-d6)δ8.00(s,1H),7.81(d,J=1.6Hz,1H),7.69(d,J=2.3Hz,1H),7.56(d,J=1.4Hz,1H),7.20(dd,J=8.6,2.3Hz,1H),6.75(d,J=8.6Hz,1H),3.94-3.79(m,2H),3.20(q,J=11.9Hz,2H),2.77(t,J=7.3Hz,1H),1.90-1.77(m,4H),1.67-1.52(m,2H),1.37(td,J=27.0,26.1,12.9Hz,4H)ppm;LCMS:m/z 450.0[M+H] + .
Example 58: (R) -8- (8- ((2, 5-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000663
1 H NMR(CD 3 OD-d4)δ8.02(s,1H),7.82(s,1H),7.52-7.58(m,2H),7.22-7.25(m,1H),6.71(s,1H),3.88-3.89(m,2H),3.22-3.24(m,2H),2.81-2.83(m,1H),1.77-1.83(m,4H),1.51-1.62(m,2H),1.354-1.46(m,4H)ppm;LCMS:m/z 450.0[M+H] + .
Example 59: (R) -8- (8- ((2-isopropylphenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000664
1 H NMR(400MHz,DMSO-d6)δ7.76(s,1H),7.58(s,2H),7.37(d,J=7.7Hz,1H),7.24(s,1H),7.06(d,J=6.1Hz,2H),3.73(s,2H),3.52(s,1H),3.11(d,J=12.8Hz,2H),2.76(s,1H),1.83(d,J=31.7Hz,4H),1.58(d,J=42.0Hz,2H),1.36(s,4H),1.24(d,J=7.0Hz,6H)ppm;LCMS:m/z 423.2[M+H] + .
Example 60: (R) -8- (8- ((2-methoxyphenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000671
1 H NMR(400MHz,DMSO-d6)δ7.77(s,1H),7.73(s,1H),7.58(s,1H),7.19(t,J=7.9Hz,1H),7.05(d,J=8.2Hz,1H),6.82-6.69(m,2H),3.87(s,3H),3.77(d,J=12.4Hz,2H),3.17(s,2H),2.97(s,2H),1.97(s,1H),1.77-1.30(m,9H)ppm;LCMS:m/z 410.1[M+H] + .
Example 61: (R) -methyl 2- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) benzoate
Figure BDA0003608939860000672
1 H NMR(400MHz,DMSO-d6)δ8.38(s,1H),7.95(dd,J=7.8,1.6Hz,1H),7.78(d,J=1.5Hz,1H),7.53(d,J=1.4Hz,1H),7.30(td,J=7.7,1.6Hz,1H),7.21(t,J=7.5Hz,1H),6.77(d,J=8.1Hz,1H),3.91(s,3H),3.85(d,J=13.4Hz,2H),3.23(d,J=12.8Hz,2H),2.97(t,J=7.0Hz,1H),1.97(d,J=7.0Hz,2H),1.82-1.40(m,8H)ppm;LC-MS:m/z 438.1[M+H] + .
Example 62: (R) -8- (8- ((4-aminophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000673
1 H NMR(400MHz,DMSO-d6)δ7.71(s,1H),7.61(s,1H),7.24(d,J=7.5Hz,2H),7.13(s,1H),6.58(d,J=7.5Hz,2H),5.48(s,2H),3.60(s,2H),3.17(s,2H),3.04-3.01(m,1H),1.40(s,10H)ppm;LC-MS:m/z 396.2[M+H] + .
Example 63: (R) -8- (8- ((3-amino-2-chlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000681
1 H NMR(400MHz,DMSO-d6)δ8.31(s,2H),7.85(s,1H),7.78(s,1H),7.58(s,1H),6.80(t,J=7.9Hz,1H),6.59(d,J=8.0Hz,1H),5.94(d,J=7.8Hz,1H),5.50(s,2H),3.84(t,J=13.1Hz,2H),3.17(s,2H),3.04(s,1H),2.00(q,J=7.2Hz,2H),1.83-1.37(m,10H)ppm;LC-MS:m/z 429.1[M+H] + .
Example 64: (R) -8- (8- ((3- (trifluoromethyl) phenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000682
1 H NMR(400MHz,DMSO-d6)δ8.31(s,2H),8.00(s,1H),7.79(d,J=1.6Hz,1H),7.59-7.52(m,1H),7.47(dd,J=16.9,9.2Hz,5H),3.84(t,J=13.0Hz,2H),3.22(d,J=13.1Hz,2H),2.98(s,1H),1.79(d,J=13.8Hz,5H),1.60-1.36(m,5H)ppm;LC-MS:m/z 448.1[M+H] + .
Example 65: (R) -N- (4- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) phenyl) acetamide
Figure BDA0003608939860000683
1 H NMR(400MHz,DMSO-d6)δ10.04(d,J=5.3Hz,1H),8.35(s,1H),7.74(s,1H),7.61(d,J=13.9Hz,2H),7.54(d,J=8.5Hz,2H),7.31(d,J=8.4Hz,2H),3.73(t,J=12.0Hz,2H),3.12(s,2H),2.97(s,1H),2.02(s,3H),1.96(s,1H),1.74(dt,J=53.1,26.6Hz,5H),1.50(d,J=35.3Hz,4H)ppm;LC-MS:m/z 396.2[M+H] + .
Example 66: (R) -5- ((5- (1-amino-8-aza-spiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -1,3, 4-thiadiazol-2-amine
Figure BDA0003608939860000684
1 H NMR(400MHz,DMSO-d6)δ8.33(s,1H),7.96(s,1H),7.81(d,J=1.5Hz,1H),7.66(d,J=1.4Hz,1H),7.33(s,2H),3.83(td,J=11.2,9.7,4.9Hz,2H),3.20(s,2H),3.05(d,J=6.9Hz,1H),1.99(t,J=6.5Hz,1H),1.86-1.69(m,4H),1.62-1.38(m,5H)ppm;LC-MS:m/z 403.1[M+H] + .
Example 67: (R) -8- (8- ((1-methyl-1H-imidazol-2-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000691
1 H NMR(400MHz,DMSO-d6)δ7.76(d,J=1.5Hz,1H),7.60(dd,J=6.8,1.4Hz,1H),7.39(d,J=1.1Hz,1H),7.35(d,J=7.0Hz,1H),7.00(d,J=1.1Hz,1H),3.80(d,J=2.6Hz,3H),3.63(d,J=10.4Hz,2H),3.08(t,J=12.2Hz,2H),2.73(t,J=7.3Hz,1H),1.90-1.67(m,4H),1.65-1.51(m,2H),1.50-1.29(m,4H)ppm;LC-MS:m/z 384.1[M+H] + .
Example 68: (R) -8- (8- (naphthalen-1-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000692
1 H NMR(CD 3 OD-d4)δ8.35-8.40(m,2H),7.99-8.02(m,1H),7.89-7.91(m,1H),7.76(m,1H),7.57-7.64(m,4H),7.43-7.47(m,2H),3.73(m,2H),3.11(m,2H),2.95(m,1H),1.71-2.05(m,4H),1.35-1.68(m,6H)ppm;LC-MS:m/z 430.2[M+H] + .
Example 69: (R) -8- (8- (thiazol-2-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000693
1 H NMR(400MHz,DMSO-d6)δ8.35(s,1H),8.11(s,1H),7.82(s,1H),7.69(d,J=3.3Hz,1H),7.62(s,1H),7.58(d,J=3.3Hz,1H),3.89(d,J=4.1Hz,2H),3.25(t,J=12.2Hz,2H),3.07(s,1H),2.32-2.25(m,2H),2.06-1.69(m,5H),1.65-1.40(m,5H)ppm;LC-MS:m/z 387.1[M+H] + .
Example 70: (R) -8- (8- (oxazol-2-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000694
1 H NMR(CD 3 OD-d4)δ8.12(s,1H),8.04(s,1H),7.80(s,1H),7.60(s,1H),7.22(s,1H),3.86(m,2H),3.22(m,2H),2.98(m,1H),1.71-2.05(m,4H),1.35-1.68(m,6H)ppm;LC-MS:m/z 371.1[M+H] + .
Example 71: synthesis of Compound (R) -N- (3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -2-chlorophenyl) acrylamide
The method comprises the following steps: n- (2-chloro-3- ((5- ((R) -1, 1-dimethylethylidene) -8-azaspiro [4.5] decan-8-yl) imidazo [1,2d ] pyrimidin-8-yl) thio) phenyl) acrylamide
Figure BDA0003608939860000701
In the procedure of synthetic example 63, the obtained intermediate (R) -N- ((R) -8- (8- ((3-amino-2-chlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-cyclobutane-1-yl) -2-methylpropane-2-sulfinamide was used in the following reaction.
To a 100mL round bottom flask was added (R) -N- ((R) -8- (8- ((3-amino-2-chlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-cyclobutane-1-yl) -2-methylpropane-2-sulfinamide (60mg, 0.1mmol), triethylamine (22mg, 0.22mmol) and dichloromethane (5 mL) in this order at 0 deg.C, then acryloyl chloride (23mg, 0.22mmol) was slowly added dropwise to the reaction solution at 0 deg.C, and after completion of the addition, the reaction solution was reacted at room temperature for 2 hours. After completion of the reaction, the reaction was quenched with 10mL of water, extracted with ethyl acetate (3X 20 mL), and the organic phase was washed with water (20 mL X1) and saturated brine (20 mL X1) in succession. The organic phase was collected, dried over anhydrous sodium sulfate, filtered and the filtrate concentrated under reduced pressure. The crude product, N- (2-chloro-3- ((5- ((R) -1, 1-dimethylethyleneidene) -8-azaspiro [4.5] decan-8-yl) imidazo [1,2d ] pyrimidin-8-yl) thio) phenyl) acrylamide was obtained by column chromatography (petroleum ether: ethyl acetate = 1) (30 mg, yield: 46%).
LC-MS:m/z 587.2[M+H] + .
Step two: (R) -N- (3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -2-chlorophenyl) acrylamide
Figure BDA0003608939860000702
According to the same manner as in the second step of example 37, sulfinyl group was removed to give (R) -N- (3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -2-chlorophenyl) acrylamide.
1 H NMR(400MHz,DMSO-d6)δ9.86(s,1H),7.99(s,1H),7.80(s,1H),7.57(s,1H),7.49(s,1H),7.10(t,J=8.0Hz,1H),6.72-6.51(m,2H),6.29(d,J=16.9Hz,1H),5.81(d,J=10.3Hz,1H),3.87(dd,J=14.9,10.9Hz,2H),3.20(s,2H),3.01(t,J=6.8Hz,1H),2.05-1.34(m,10H)ppm;LC-MS:m/z 482.8[M+H] + .
Following the synthesis of example 71, the following compounds may be synthesized:
example 72: (R) -N1- (3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -2-chlorophenyl) -N2, N2-dimethyloxyalkanamide
Figure BDA0003608939860000711
1 H NMR(400MHz,DMSO-d 6 )δ8.00(s,1H),7.80(s,1H),7.57(s,1H),7.41(d,J=8.0Hz,1H),7.13(t,J=8.0Hz,1H),6.62(d,J=8.2Hz,1H),5.49(s,1H),3.86(d,J=12.7Hz,2H),3.81(s,3H),3.23(t,J=12.7Hz,2H),3.11(s,3H),3.02(d,J=6.8Hz,1H),2.05-1.75(m,5H),1.54(dd,J=41.2,13.2Hz,5H)ppm;LC-MS:m/z528.2[M+H] + .
Example 73: synthesis of the compound (R) -N- (3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -2-chlorophenyl) -2-hydroxy-4-carbonyl-4H-pyrido [1,2-a ] pyrimidine-3-carboxamide
The method comprises the following steps: 2-hydroxy-4-carbonyl-4H-pyrido [1,2-a ] pyrimidine-3-carboxylic acid ethyl ester
Figure BDA0003608939860000712
Triethyl methanetricarboxylate (4.64g, 20mmol) was added to a xylene (10 mL) solution of pyridin-2-amine (940mg, 10mmol) under nitrogen protection. The mixture was further stirred at 140 ℃ for 4 hours, TLC monitored until the starting materials reacted, the reaction mixture was filtered and washed with ethyl acetate to give ethyl 2-hydroxy-4-carbonyl-4H-pyrido [1,2-a ] pyrimidine-3-carboxylate (1.95 g, yield: 83%)
1 H NMR(400MHz,DMSO-d 6 )δ12.47(s,1H),8.93(dd,J=7.4,1.5Hz,1H),8.19(ddd,J=8.6,7.1,1.6Hz,1H),7.43-7.34(m,2H),4.15(q,J=7.1Hz,2H),1.23(t,J=7.1Hz,3H)ppm;LCMS:m/z 235[M+H] + .
Step two: n- (2-chloro-3- ((5- ((R) -1, 1-dimethylethylidene) -8-azaspiro [4.5] decan-8-yl) imidazo [1,2d ] pyrimidin-8-yl) thio) phenyl) -2-hydroxy-4-carbonyl-4H-pyrido [1,2-a ] pyrimidine-3-carboxamide
In the procedure of synthetic example 63, the obtained intermediate (R) -N- ((R) -8- (8- ((3-amino-2-chlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-cyclobutane-1-yl) -2-methylpropane-2-sulfinamide was used in the following reaction.
Figure BDA0003608939860000713
To a solution of ethyl 2-hydroxy-4-carbonyl-4H-pyrido [1,2-a ] pyrimidine-3-carboxylate (40mg, 0.1699 mmol) in DMF (2 mL) under nitrogen was added (R) -N- ((R) -8- (8- ((3-amino-2-chlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-cyclobutan-1-yl) -2-methylpropane-2-sulfinamide (60mg, 0.113mmol). The mixture was reacted at 160 ℃ for 1 hour by microwave. After the mixture was cooled to room temperature, the mixture was filtered, diluted with water and extracted with ethyl acetate (20 mL × 3), the organic layer was washed with saturated brine and mixed, dried over anhydrous sodium sulfate, filtered and concentrated to give N- (2-chloro-3- ((5- ((R) -1, 1-dimethylethylidene) -8-azaspiro [4.5] decan-8-yl) imidazo [1,2d ] pyrimidin-8-yl) thio) phenyl) -2-hydroxy-4-carbonyl-4H-pyrido [1,2-a ] pyrimidine-3-carboxamide (20 mg, yield 21.7%).
LCMS:m/z 721[M+H] + .
Step three: (R) -N- (3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -2-chlorophenyl) -2-hydroxy-4-carbonyl-4H-pyrido [1,2-a ] pyrimidine-3-carboxamide
Figure BDA0003608939860000721
According to the same manner as in the second step of example 37, sulfinyl group was removed to give (R) -N- (3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -2-chlorophenyl) -2-hydroxy-4-carbonyl-4H-pyrido [1,2-a ] pyrimidine-3-carboxamide.
1 H NMR(400MHz,DMSO-d6)δ8.68(s,1H),8.43(s,1H),7.94(s,1H),7.79(s,1H),7.63(d,J=34.6Hz,2H),7.03(d,J=8.2Hz,1H),6.61-6.51(m,1H),6.40(d,J=8.2Hz,1H),6.00(d,J=6.9Hz,1H),5.45(s,1H),3.94-3.80(m,2H),3.23(s,2H),3.05(s,1H),2.09-1.34(m,10H)ppm;LC-MS:m/z 617.1[M+H] + .
Example 74: synthesis of the compound (R) -N- (3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -2-chlorophenyl) -2-hydroxy-4-carbonyl-6, 7,8, 9-tetrahydro-4H-pyrido [1,2-a ] pyrimidine-3-carboxamide
The method comprises the following steps: 2-hydroxy-4-carbonyl-6, 7,8, 9-tetrahydro-4H-pyrido [1,2-a ] pyrimidine-3-carboxylic acid ethyl ester
Figure BDA0003608939860000722
To a 100mL round bottom flask were added ethyl 2-hydroxy-4-carbonyl-4H-pyrido [1,2-a ] pyrimidine-3-carboxylate (obtained in step one, example 69) (1g, 4.12mmol), methanol (25 mL) and 10% Pd/C (862 mg) in that order. The mixture was further stirred under hydrogen (balloon) at room temperature for 2.5 hours, TLC monitored until the starting materials reacted, and the reaction mixture was filtered through Celite and concentrated in vacuo to give ethyl 2-hydroxy-4-carbonyl-6, 7,8, 9-tetrahydro-4H-pyrido [1,2-a ] pyrimidine-3-carboxylate (700 mg, yield: 69.4%).
1 H NMR(400MHz,DMSO-d 6 )δ12.39(s,1H),4.09(q,J=7.1Hz,2H),3.66(t,J=6.0Hz,2H),2.77(t,J=6.4Hz,2H),1.88-1.80(m,2H),1.78-1.71(m,2H),1.19(t,J=7.1Hz,3H)ppm;LCMS:m/z[M+H] + .
(R) -N- (3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -2-chlorophenyl) -2-hydroxy-4-carbonyl-6, 7,8, 9-tetrahydro-4H-pyrido [1,2-a ] pyrimidine-3-carboxamide was obtained in the same manner as in example 73, step two and step three.
Figure BDA0003608939860000723
1 H NMR(400MHz,DMSO-d6)δ10.31(s,1H),8.24(s,1H),7.96(s,1H),7.85(s,1H),7.81-7.72(m,2H),7.58(s,1H),3.86(dd,J=30.5,16.1Hz,4H),3.29-3.15(m,4H),2.85-2.78(m,1H),2.11-1.61(m,14H);LC-MS:m/z 621.1[M+H] + .
Example 75: synthesis of (R) -8- (8- (2, 3-dichlorophenyl) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
The method comprises the following steps: (R) -N- ((R) -8- (8- (2, 3-dichlorophenyl) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide
Figure BDA0003608939860000731
(R) -N- ((R) -8- (8-iodoimidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfonamide (60mg, 0.12mmol), 1, 4-dioxane (2 mL), purified water (0.5 mL), (2, 3-dichlorophenyl) boronic acid (50mg, 0.24mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (9mg, 0.012mmol) and potassium carbonate (50mg, 0.36mmol) were added sequentially at room temperature in a 20mL stoppered tube. Nitrogen was bubbled for one minute, the tube was sealed and heated to 80 ℃ and the reaction was allowed to proceed for 6 hours. After completion of the reaction, 20mL of water was added to the reaction solution, and the mixture was extracted with ethyl acetate (50 mL. Times.3). The organic phase was washed successively with water (20 mL. Times.1) and saturated brine (20 mL. Times.1). The organic phase was collected, dried over anhydrous sodium sulfate, filtered and the filtrate concentrated under reduced pressure. The crude product (R) -N- ((R) -8- (8- (2, 3-dichlorophenyl) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide was obtained by column chromatography (petroleum ether: ethyl acetate = 1). (30 mg, yield: 48%) as a pale yellow solid.
LC-MS:m/z 520.1[M+H] + .
Step two: (R) -8- (8- (2, 3-dichlorophenyl) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000732
Following the same procedure as in step two of example 37, (R) -N- ((R) -8- (8- (2, 3-dichlorophenyl) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinylamide-sulfinyl group was removed to give (R) -8- (8- (2, 3-dichlorophenyl) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine.
1 H NMR(400MHz,DMSO-d6)δ8.35(s,1H),7.75(s,3H),7.60(s,1H),7.49(s,1H),3.79(s,2H),3.21-3.12(m,2H),2.93(s,1H),1.82(s,5H),1.46(s,7H)ppm;LCMS:m/z 416.1[M+H] + .
Example 76: synthesis of Compound (R) -8 (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
The method comprises the following steps: (R) -N- ((R) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide
Figure BDA0003608939860000733
To a 100mL single-neck flask, under nitrogen, was added the crude product (R) -N- ((R) -8- (8-iodoimidazo [1, 2-c) in sequence]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-methylpropane-2-sulfonamide (100mg, 0.20mmol), 2-amino-3-chloropyridine-4-thiol sodium (43mg, 0.26mmol), pd 2 (dba) 3 (20mg, 0.02mmol), xantphos (23mg, 0.040mmol), DIPEA (52mg, 0.40mmol) and 1, 4-dioxane solution (10 mL), and the mixture was heated to 100 ℃ under nitrogen and stirred for reaction for 6 hours. After completion of the reaction, it was cooled to room temperature, filtered and the residue obtained was concentrated under reduced pressure and purified by silica gel chromatography (0 to 30% gradient of ethyl acetate/methanol) to give (R) -N- ((R) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1, 2-c)]Pyrimidin-5-yl) -8-azaSpiro [4.5]]Decan-1-yl) -2-methylpropane-2-sulfinamide (40 mg, yield: 37%).
LC-MS:m/z 534.2[M+H] + .
Step two: (R) -8 (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000741
Following the same procedure as in step two of example 37, (R) -N- ((R) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide is desublimated to give (R) -8 (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine.
1 H NMR(400MHz,DMSO-d6)δ8.32(d,J=6.9Hz,1H),8.02(s,1H),7.81(s,1H),7.58(d,J=1.4Hz,1H),7.54(d,J=5.4Hz,1H),6.35(s,1H),5.78(d,J=5.4Hz,1H),3.89(t,J=12.5Hz,2H),3.29-3.17(m,3H),3.00(s,1H),1.97(s,1H),1.89-1.33(m,6H)ppm;LCMS:m/z 430.1[M+H] + .
Following the synthetic procedure of example 76, the following compounds can be synthesized:
example 77: (R) -8- (8- ((3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000742
1 H NMR(CD 3 OD-d4)δ8.54(s,1H),8.35(s,1H),8.15-8.16(m,1H),8.09(s,1H),7.83-7.84(m,1H),7.57-7.58(m,1H),6.67-6.68(m,1H),3.92(m,2H),3.26(m,2H),3.00(m,1H),1.71-2.05(m,4H),1.35-1.68(m,6H)ppm;LCMS:m/z 415.1[M+H] + .
Example 78: (R) -8- (8- ((3- (trifluoromethyl) pyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000743
1 H NMR(400MHz,DMSO-d6)δ8.82(s,1H),8.41(d,J=5.5Hz,1H),8.31(s,1H),8.10(s,1H),7.84(s,1H),7.57(s,1H),6.90(d,J=5.4Hz,1H),3.93(s,2H),3.28-3.20(m,2H),2.94(s,1H),1.90-1.32(m,10H);LCMS:m/z 449.1[M+H] + .
Example 79: (3S, 4S) -8- (8- ((2-amino-5-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000751
1 H NMR(400MHz,DMSO-d6)δ8.05(s,1H),7.87(s,1H),7.83(s,1H),7.62(d,J=1.3Hz,1H),5.91(s,2H),5.65(s,1H),4.13(d,J=5.2Hz,1H),3.77(d,J=8.5Hz,3H),3.60(d,J=8.6Hz,1H),3.40(s,1H),3.14(s,1H),1.81(dd,J=82.1,33.6Hz,5H),1.14(d,J=6.3Hz,3H)ppm;LC-MS:m/z 462.1[M+H] + .
EXAMPLE 80 (3S, 4S) -8- (8- ((6-amino-3-chloro-2-methyl-pyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000752
1 H NMR(400MHz,DMSO-d6)δ8.14(s,1H),8.04(s,1H),7.85(s,1H),7.63(s,1H),5.82(s,2H),5.51(s,1H),4.26-4.18(m,1H),3.88(t,J=13.0Hz,3H),3.70(d,J=8.9Hz,1H),3.42(d,J=4.3Hz,1H),3.27(s,2H),2.31(s,3H),1.99(d,J=10.0Hz,2H),1.80(d,J=13.5Hz,1H),1.69(d,J=12.8Hz,1H),1.23(d,J=6.4Hz,3H)ppm;LC-MS:m/z 459.8[M+H] + .
Example 81: (R) -8- (8- ((3-chloro-2-cyclopropylpyrimidin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000753
1 H NMR(400MHz,DMSO-d6)δ8.06(s,1H),7.97(d,J=5.3Hz,1H),7.82(d,J=1.5Hz,1H),7.57(d,J=1.4Hz,1H),6.40(d,J=5.3Hz,1H),3.91(t,J=13.1Hz,2H),3.25(t,J=11.7Hz,2H),3.02(t,J=6.6Hz,1H),2.03-1.65(m,5H),1.67-1.37(m,5H),1.04(ddt,J=8.0,5.6,2.4Hz,2H),0.97(dq,J=6.9,4.2,3.4Hz,2H)ppm;LCMS:m/z 455.1[M+H] + .
Example 82: (R) -8- (8- ((3-chloro-2-methyl-pyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000761
1 H NMR(400MHz,DMSO-d6)δ8.35-8.29(m,1H),8.07(s,1H),8.02(d,J=5.4Hz,1H),7.83(s,1H),7.57(d,J=1.4Hz,1H),6.50(d,J=5.3Hz,1H),3.91(t,J=12.9Hz,2H),3.26(t,J=12.3Hz,2H),3.01(s,1H),2.55(s,3H),2.02-1.77(m,4H),1.75-1.38(m,6H)ppm;LCMS:m/z 429.1[M+H] + .
Example 83: (R) -8- (8- ((2-amino-5-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000762
1 H NMR(CD 3 OD-d4)δ8.38(s,1H),8.04(s,1H),7.82-7.84(m,2H),7.61(m,1H),5.93(s,2H),5.66(m,1H),3.87-3.93(m,2H),3.21-3.28(m,2H),2.98(m,1H),1.71-2.05(m,4H),1.35-1.68(m,6H)ppm;LCMS:m/z430.1[M+H] + .
Example 84: (R) -8- (8- ((2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000763
1 H NMR(400MHz,DMSO-d6)δ8.10(s,1H),8.00(d,J=5.3Hz,1H),7.83(d,J=1.5Hz,1H),7.59(d,J=1.4Hz,1H),6.72(d,J=5.3Hz,1H),4.03-3.87(m,2H),3.27(d,J=13.6Hz,2H),3.14(t,J=6.3Hz,1H),2.05(q,J=6.4Hz,1H),1.87-1.42(m,9H)ppm;LCMS:m/z 449.1[M+H] + .
Example 85: (R) -8- (8- ((2-methylpyridin-3-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000764
1 H NMR(400MHz,DMSO-d6)δ8.25(dd,J=4.7,1.5Hz,1H),7.89(s,1H),7.79(d,J=1.4Hz,1H),7.56(d,J=1.4Hz,1H),7.23(dd,J=7.9,1.5Hz,1H),7.06(dd,J=7.9,4.7Hz,1H),3.89-3.70(m,2H),3.17(dd,J=22.2,10.5Hz,2H),2.80(t,J=7.2Hz,1H),2.60(s,3H),1.88-1.20(m,10H)ppm;LCMS:m/z 395.2[M+H] + .
Example 86: (R) -8- (8- ((2- (trifluoromethyl) pyridin-3-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000771
1 H NMR(400MHz,DMSO-d6)δ8.52(s,1H),8.35(s,1H),7.75(s,1H),7.21(d,J=8.2Hz,2H),6.52(d,J=8.4Hz,2H),5.39(s,2H),4.69(s,2H),2.87(s,1H),1.93-1.30(m,10H)ppm;LCMS:m/z 449.1[M+H] + .
Example 87: (R) -8- (8- ((2-Chloropyridin-3-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000772
1 H NMR(CD 3 OD-d4)δ8.38(s,1H),8.16-8.18(m,1H),8.06(s,1H),7.81(m,2H),7.23(m,1H),7.14-7.22(m,2H),3.88(m,2H),3.23(m,2H),2.97(m,1H),1.71-2.05(m,4H),1.35-1.68(m,6H)ppm;LCMS:m/z 415.1[M+H] + .
Example 88: (R) -8- (8- ((6-amino-2-chloropyridin-3-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000773
1 H NMR(400MHz,DMSO-d6)δ8.33(s,1H),7.74(d,J=1.4Hz,1H),7.61(d,J=1.4Hz,1H),7.51(d,J=8.5Hz,1H),7.40(s,1H),6.73(s,2H),6.38(d,J=8.4Hz,1H),3.68(d,J=12.5Hz,3H),3.10(s,2H),2.97(t,J=6.8Hz,1H),2.01-1.31(m,10H)ppm.LC-MS:m/z 431.1[M+H] + .
Example 89: (R) -8- (8- (benzo [ d ] thiazol-7-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000781
1 HNMR(CD 3 OD-d4)δ9.43(m,1H),8.35(s,3H),8.00-8.02(m,1H),7.88(s,1H),7.76(m,1H),7.56-7.58(m,1H),7.47-7.51(m,1H),7.38-7.40(m,1H),3.80(m,2H),3.17(m,2H),2.98(m,1H),1.71-2.05(m,4H),1.35-1.68(m,6H)ppm;LC-MS:m/z 437.1[M+H] + .
Example 90: (R) -8- (8- (phenylsulfanyl) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000782
1 H NMR(400MHz,DMSO-d6)δ8.31(s,1H),7.83(s,1H),7.76(s,1H),7.59(s,1H),7.25(tq,J=14.0,7.4Hz,5H),3.84-3.70(m,2H),3.16(d,J=12.4Hz,2H),3.01(t,J=6.8Hz,1H),1.97(dd,J=13.2,7.2Hz,1H),1.86-1.26(m,9H)ppm;LCMS:m/z 380.1[M+H] + .
Example 91: (R) -8- (8- ((1-methyl-1H-pyrrolo [2,3-b ] pyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000783
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.97(d,J=5.1Hz,1H),7.80(s,1H),7.61-7.49(m,2H),6.56-6.39(m,2H),3.84(d,J=22.6Hz,5H),3.22(t,J=12.5Hz,2H),2.98(t,J=6.9Hz,1H),2.03-1.33(m,10H)ppm;LC-MS:m/z 433.9[M+H] + .
Example 92: (R) -8- (8- ((2, 3-dihydrobenzofuran-5-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000784
1 H NMR(400MHz,DMSO-d6)δ7.72(s,1H),7.62(s,1H),7.46(s,1H),7.39(s,1H),7.27(d,J=8.2Hz,1H),6.77(d,J=8.3Hz,1H),4.54(t,J=8.7Hz,2H),3.69(t,J=11.9Hz,2H),3.11(dt,J=29.5,12.5Hz,6H),1.99(s,1H),1.87-1.36(m,9H)ppm;LC-MS:m/z 422.1[M+H] + .
Example 93: (R) -8- (8- (quinolin-4-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000791
1 H NMR(400MHz,DMSO-d6)δ8.52(d,J=4.8Hz,1H),8.26(dd,J=8.4,1.3Hz,1H),8.12(s,1H),8.04(dd,J=8.5,1.2Hz,1H),7.89-7.79(m,2H),7.73(ddd,J=8.2,6.8,1.3Hz,1H),7.56(d,J=1.4Hz,1H),6.82(d,J=4.8Hz,1H),3.91(dd,J=15.3,11.3Hz,2H),3.37-3.16(m,2H),3.05(t,J=6.5Hz,1H),2.10-1.65(m,5H),1.67-1.38(m,5H)ppm;LC-MS:m/z 431.2[M+H] + .
Example 94: (1R) -8- (8- ((2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000792
1 H NMR(400MHz,DMSO-d6)δ8.32(s,1H),8.09(d,J=6.5Hz,1H),8.00(d,J=5.3Hz,1H),7.84(s,1H),7.57(s,1H),6.71(d,J=5.3Hz,1H),4.03-3.85(m,2H),3.26-3.15(m,2H),2.98(d,J=21.9Hz,1H),2.14(dd,J=25.3,11.9Hz,2H),1.87(dd,J=41.5,14.7Hz,3H),1.43(dd,J=32.2,18.7Hz,2H),1.28(dd,J=18.2,8.7Hz,1H),1.12(d,J=12.5Hz,1H),1.05-0.93(m,3H)ppm;LC-MS:m/z 462.8[M+H] + .
Example 95: (1R) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000793
1 H NMR(400MHz,DMSO-d6)δ8.36(s,1H),8.02(s,1H),7.80(s,1H),7.63-7.49(m,2H),6.34(s,2H),5.78(d,J=5.4Hz,1H),3.98-3.83(m,2H),3.18(dd,J=27.6,14.3Hz,2H),3.06-2.94(m,1H),2.23-2.07(m,2H),1.96-1.89(m,1H),1.86-1.67(m,2H),1.51-1.33(m,2H),1.33-1.20(m,1H),1.21-1.09(m,1H),1.08-0.94(m,3H)ppm;LC-MS:m/z 443.8[M+H] + .
Example 96: (1R) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-8-aza-spiro [4.5] decan-1-amine
Figure BDA0003608939860000801
1 H NMR(400MHz,DMSO-d6)δ8.33(s,1H),8.06(s,1H),7.84(s,1H),7.61(s,1H),6.33(s,2H),5.62(s,1H),4.00-3.86(m,2H),3.17(d,J=13.0Hz,1H),3.07-2.95(m,1H),2.25-2.07(m,2H),1.94(dd,J=12.0,8.2Hz,2H),1.79(dd,J=28.2,13.0Hz,2H),1.45(dd,J=33.4,19.7Hz,2H),1.34-1.11(m,2H),1.01(dd,J=18.3,10.3Hz,3H);LCMS:m/z 447.8[M+H] + .
Example 97: (S) -8- (8- ((2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000802
1 H NMR(400MHz,DMSO-d6)δ8.19(s,1H),8.10(s,1H),8.00(d,J=5.3Hz,1H),7.86(s,1H),7.58(s,1H),6.72(d,J=5.3Hz,1H),4.02(dd,J=8.8,6.4Hz,1H),3.85(dd,J=17.4,13.7Hz,2H),3.72(dd,J=21.2,8.6Hz,2H),3.43(dd,J=9.0,4.8Hz,2H),3.25(dd,J=15.2,9.6Hz,2H),1.98-1.86(m,1H),1.85-1.73(m,1H),1.61(s,2H);LC-MS:m/z 451.7[M+H] + .
Example 98: (S) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000803
1 H NMR(400MHz,DMSO-d6)δ8.14(s,1H),8.04(s,1H),7.83(s,1H),7.60(s,1H),7.55(d,J=5.4Hz,1H),6.35(s,2H),5.78(d,J=5.4Hz,1H),4.10(dd,J=9.9,6.0Hz,1H),3.92-3.76(m,4H),3.69(dd,J=10.1,3.1Hz,1H),3.53(s,1H),3.24(d,J=10.8Hz,1H),1.99-1.83(m,2H),1.73(d,J=12.0Hz,2H),1.24(d,J=6.4Hz,1H)ppm;LC-MS:m/z 431.8[M+H] + .
Example 99: (3S, 4S) -8- (8- ((2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000811
1 H NMR(400MHz,DMSO-d6)δ8.17(s,1H),8.10(s,1H),8.00(d,J=5.3Hz,1H),7.87(s,1H),7.57(d,J=1.0Hz,1H),6.72(d,J=5.3Hz,1H),4.12(dt,J=12.1,6.1Hz,1H),3.85-3.67(m,3H),3.57(d,J=8.5Hz,1H),3.41(dd,J=26.4,9.0Hz,2H),3.06(d,J=4.9Hz,1H),1.99-1.81(m,2H),1.68(dd,J=27.9,14.6Hz,2H),1.13(d,J=6.4Hz,3H)ppm;LC-MS:m/z 464.8[M+H] + .
Example 100: (3S, 4S) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000812
1 H NMR(400MHz,DMSO-d6)δ8.21(s,1H),8.02(s,1H),7.84(s,1H),7.61-7.50(m,2H),6.34(s,2H),5.79(d,J=5.4Hz,1H),4.11(dt,J=12.3,6.3Hz,1H),3.73(d,J=8.5Hz,3H),3.56(d,J=8.5Hz,1H),3.38-3.26(m,2H),3.05(d,J=4.9Hz,1H),1.99-1.81(m,2H),1.67(dd,J=26.4,15.3Hz,2H),1.18-1.02(m,3H)ppm;LC-MS:m/z 445.8[M+H] + .
Example 101:4- ((5- (4-amino-4-methylpiperidin-1-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -3-chloropyridin-2-amine
Figure BDA0003608939860000813
1 H NMR(400MHz,DMSO-d6)δ8.00(s,1H),7.83(s,1H),7.54(d,J=5.8Hz,2H),6.29(s,2H),5.79(d,J=5.4Hz,1H),3.60(d,J=4.3Hz,4H),1.77-1.53(m,4H),1.19(s,3H)ppm;LC-MS:m/z 390.1[M+H] + .
Example 102: (R) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000814
1 H NMR(400MHz,DMSO-d6)δ8.05(s,1H),7.85(d,J=1.5Hz,1H),7.60(d,J=1.4Hz,1H),6.32(s,2H),5.62(s,1H),3.90(dd,J=13.2,8.5Hz,2H),3.23(q,J=11.9,11.0Hz,2H),2.77(t,J=7.3Hz,1H),1.90-1.77(m,4H),1.67-1.53(m,2H),1.45-1.28(m,4H)ppm;LCMS:m/z 466.1[M+H] + .
Example 103: (R) -8- (8- ((6-amino-3-chloro-2-methyl-pyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000821
1 H NMR(400MHz,DMSO-d6)δ7.88(s,4H),7.66(d,J=11.1Hz,1H),5.81(s,1H),3.96(t,J=13.9Hz,2H),3.37-3.23(m,3H),2.42(s,3H),2.14-2.05(m,1H),1.88-1.46(m,9H);LC-MS:m/z 443.8[M+H] + .
Example 104: (S) -8- (8- ((6-amino-3-chloro-2-methylpyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000822
1 H NMR(400MHz,DMSO-d6)δ8.17(d,J=4.9Hz,3H),7.91(s,1H),7.68(s,1H),5.81(s,1H),4.13(dd,J=10.4,5.8Hz,1H),3.90(d,J=9.0Hz,3H),3.80(d,J=9.0Hz,1H),3.74(dd,J=10.3,2.6Hz,1H),3.63(s,1H),3.40(dd,J=12.6,8.6Hz,1H),3.30(dd,J=12.6,9.4Hz,1H),2.42(s,3H),1.95-1.81(m,2H),1.75(d,J=10.3Hz,2H);LC-MS:m/z 445.8[M+H] + .
Example 105: (S) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000823
1 H NMR(400MHz,DMSO-d6)δ8.16(s,1H),8.07(s,1H),7.87(s,1H),7.63(s,1H),6.33(s,2H),5.62(s,1H),4.06(dd,J=9.6,6.2Hz,1H),3.93-3.74(m,4H),3.56(dd,J=9.5,4.0Hz,1H),3.26(s,3H),1.98-1.80(m,2H),1.67(s,2H)ppm;LC-MS:m/z 466.6[M+H] + .
Example 106: (3S, 4S) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000831
1 H NMR(400MHz,DMSO-d6)δ8.18(s,3H),8.08(s,1H),7.87(s,1H),7.65(s,1H),6.35(s,2H),5.64(s,1H),4.31-4.16(m,1H),4.03-3.83(m,3H),3.71(d,J=9.0Hz,1H),3.46(s,1H),3.24(d,J=11.2Hz,2H),2.03(t,J=12.3Hz,2H),1.75(dd,J=41.2,13.7Hz,2H),1.24(d,J=6.5Hz,3H);LCMS:m/z 479.7[M+H] + .
Example 107: (3S, 4S) -8- (8- ((3-chloro-2-methylpyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000832
1 H NMR(400MHz,DMSO-d6)δ8.06(s,1H),8.01(d,J=5.3Hz,1H),7.85(d,J=1.5Hz,1H),7.56(d,J=1.4Hz,1H),6.50(d,J=5.3Hz,1H),4.15-4.05(m,1H),3.74-3.66(m,5H),3.45-3.42(m,1H),3.03(d,J=4.9Hz,1H),2.55(s,3H),2.00-1.60(m,4H),1.12(d,J=6.4Hz,3H)ppm;LC-MS:m/z 445.2[M+H] + .
Example 108: (S) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000833
1 H NMR(400MHz,DMSO-d6)δ8.23(s,1H),8.06(s,1H),7.88(s,1H),7.61(s,1H),6.34(s,2H),5.64(s,1H),3.95(d,J=14.3Hz,2H),3.55(s,1H),3.44(s,1H),3.31(d,J=11.5Hz,2H),2.69(s,1H),1.94(d,J=12.4Hz,2H),1.66(t,J=11.9Hz,2H),1.06-0.99(m,3H),0.94(d,J=14.9Hz,3H);LC-MS:m/z 493.7[M+H] + .
Example 109: (S) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000841
1 H NMR(400MHz,DMSO-d6)δ8.15(s,2H),7.89(s,1H),7.64(s,1H),6.33(s,1H),5.63(s,1H),4.03(s,2H),3.59(d,J=14.4Hz,2H),3.38(s,2H),3.09(s,1H),1.83(s,4H),1.16(s,3H),1.07(s,3H);LC-MS:m/z493.7[M+H] + .
Example 110: (S) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000842
1 H NMR(400MHz,DMSO-d6)δ8.16(s,1H),8.02(s,1H),7.84(s,1H),7.61-7.51(m,2H),6.33(s,2H),5.80(d,J=5.4Hz,1H),4.00-3.88(m,2H),3.55(d,J=8.7Hz,1H),3.46(d,J=8.8Hz,1H),3.32-3.27(m,2H),2.73(s,1H),1.98-1.86(m,2H),1.68(s,2H),1.00(d,J=29.6Hz,6H);LC-MS:m/z 459.8[M+H] + .
Example 111: (R) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-dimethyl-1-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0003608939860000843
1 H NMR(400MHz,DMSO-d6)δ8.15(s,1H),7.84(s,1H),7.57(s,1H),7.55(d,J=5.4Hz,1H),6.33(s,2H),5.80(d,J=5.3Hz,1H),4.01-3.91(m,2H),3.56(d,J=8.8Hz,1H),3.48(d,J=8.9Hz,1H),3.32-3.26(m,2H),2.80(s,1H),1.97-1.88(m,2H),1.70(d,J=12.3Hz,2H),1.06(s,3H),0.99(s,3H);LC-MS:m/z 459.8[M+H] + .
EXAMPLE 112 Synthesis of (R) -8- (8- ((3-chloro-2-methoxypyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Step one methyl ((R) -1- ((R) -tert-butylsulfinyl) amino) -8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) propionate
Figure BDA0003608939860000851
To a 100mL single-neck flask, under nitrogen, was added (R) -N- ((R) -8- (8-iodoimidazo [1, 2-c) in sequence]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-methylpropane-2-sulfonamide (500mg, 1.0mmol), methyl mercaptopropionate (132mg, 1.1mmol), pd 2 (dba) 3 (46mg, 0.05mmol), xantphos (58mg, 0.10 mmol), DIPEA (258mg, 2.0 mmol) and 1, 4-dioxane solution (30 mL) were heated under nitrogen at 100 ℃ and the reaction was stirred for 4 hours. After completion of the reaction, it was cooled to room temperature, filtered and the residue obtained was concentrated under reduced pressure and purified by silica gel chromatography (0 to 30% gradient of ethyl acetate/petroleum ether) to give ((R) -1- ((R) -tert-butylsulfinyl) amino) -8-azaspiro [ 4.5%]Decan-8-yl) imidazo [1,2-c]Pyrimidin-8 yl) thio) propionic acid methyl ester (380 mg, yield: 77%).
LCMS:m/z 494.2[M+H] + .
Step two ((R) -1- ((R) -1, 1-dimethylethylsulfonamido) -8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidine-8-thiolate
Figure BDA0003608939860000852
To a dry 100mL round bottom flask were added methyl ((R) -1- ((R) -tert-butylsulfinyl) amino) -8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8 yl) thio) propanoate (380mg, 0.77mmol) and tetrahydrofuran (20 mL) in that order, and then a solution of sodium ethoxide in ethanol (21%, 3 mL) was slowly added dropwise at room temperature, and the reaction was stirred at room temperature for 1 hour. Concentration under reduced pressure gave the crude ((R) -1- ((R) -1, 1-dimethylethylsulfinylamino) -8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidine-8-thiolate (400 mg) without purification.
LC-MS:m/z 408.2[M+H] + .
Step three (R) -N- ((R) -8- (8- ((3-chloro-2-methoxypyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide)
Figure BDA0003608939860000853
To a 100mL single neck flask, under nitrogen, was added the crude ((R) -1- ((R) -1, 1-dimethylethylsulfonamido) -8-azaspiro [4.5] in sequence]Decan-8-yl) imidazo [1,2-c]Pyrimidine-8-thiolate (100mg, 0.23mmol), 3-chloro-4-iodo-2-methoxypyridine (71mg, 0.26mmol), pd 2 (dba) 3 (22mg, 0.0248 mmol), xantphos (28mg, 0.048 mmol), DIPEA (62mg, 0.48mmol) and 1, 4-dioxane solution (10 mL), and the mixture was heated to 100 ℃ under nitrogen and stirred for 4 hours. After completion of the reaction, it is cooled to room temperature, filtered and the residue obtained is concentrated under reduced pressure and purified by chromatography on silica gel (gradient 0 to 30% ethyl acetate/methanol) to give (R) -N- ((R) -8- (8- ((3-chloro-2-methoxypyridin-4-yl) thio) imidazo [1, 2-c)]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-methylpropane-2-sulfinamide (51 mg, yield: 40%).
LCMS:m/z 549.2[M+H] + .
Step four (R) -8- (8- ((3-chloro-2-methoxypyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000861
To a 100mL one-neck flask were added (R) -N- ((R) -8- (8- ((3-chloro-2-methoxypyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide (40mg, 0.09mmol) and methanol (2.3 mL) in this order under a nitrogen blanket, a solution of 1, 4-dioxane hydrochloride (0.23mL, 4M) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1 hour. After completion of the reaction, it was cooled to room temperature, filtered and the resulting residue was concentrated under reduced pressure to give (R) -8- (8- ((3-chloro-2-methoxypyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine (15 mg, yield: 37%) by high performance liquid preparative chromatography.
1 H NMR(400MHz,DMSO-d6)δ8.05(s,1H),7.83(d,J=1.6Hz,1H),7.77(d,J=5.5Hz,1H),7.56(d,J=1.5Hz,1H),6.28(d,J=5.5Hz,1H),3.93(s,3H),3.88(d,J=9.4Hz,2H),3.23(d,J=11.6Hz,2H),2.76(t,J=7.2Hz,1H),1.80(d,J=11.4Hz,4H),1.66-1.52(m,2H),1.44-1.29(m,4H).LCMS:m/z 445.1[M+H] + Following the synthesis of example 112, the following compound can be synthesized:
example 113 methyl (R) -3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) propionate
Figure BDA0003608939860000862
1 H NMR(400MHz,DMSO-d6)δ7.73(d,J=2.7Hz,2H),7.64(d,J=1.3Hz,1H),3.70(d,J=4.0Hz,2H),3.56(s,3H),3.26(t,J=7.0Hz,2H),3.15-3.04(m,3H),2.61(t,J=7.0Hz,2H),2.05-1.68(m,5H),1.66-1.37(m,5H)ppm;LC-MS:m/z 390.1[M+H] + .
Example 114: (R) -8- (8- ((3-chloro-2-fluoropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000863
1 H NMR(400MHz,DMSO-d6)δ8.10(s,1H),7.86-7.80(m,2H),7.58(d,J=1.4Hz,1H),6.66(d,J=5.4Hz,1H),3.95(d,J=12.7Hz,2H),3.28(s,2H),2.95(s,1H),1.97-1.80(m,4H),1.66(d,J=32.2Hz,2H),1.56-1.41(m,4H)ppm;LCMS:m/z 434.1[M+H] + .
Example 115: (R) -8- (8- ((3-chloro-2- (dimethylamino) pyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000871
1 H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.83(d,J=1.6Hz,1H),7.79(d,J=5.3Hz,1H),7.56(d,J=1.4Hz,1H),6.15(d,J=5.3Hz,1H),3.98-3.79(m,2H),3.23(d,J=11.6Hz,3H),2.90(s,6H),2.77(t,J=7.3Hz,1H),1.83-1.74(m,7H),1.69-1.51(m,3H),1.49-1.27(m,5H)ppm;LCMS:m/z 458.2[M+H] + .
Example 116: (R) -4- ((5- (1-amino-8-aza-spiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) indoline-2, 3-dione
Figure BDA0003608939860000872
1 H NMR(400MHz,DMSO-d6)δ8.28(s,1H),8.02(s,1H),7.81(s,1H),7.58(s,1H),7.24(t,J=8.0Hz,1H),6.58(d,J=7.7Hz,2H),6.21(d,J=8.3Hz,1H),3.90(s,3H),3.24(s,3H),2.97(s,1H),1.69(d,J=93.3Hz,9H)ppm;LC-MS:m/z 449.1[M+H] + .
EXAMPLE 117 Synthesis of the Compound (R) -8- (8- ((3-chloropyridazin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Step one (R) -N- ((R) -8- (8- ((3-chloropyridazin-4-yl) thio) imidazo [1,2-c ] pyrimidine-5-yl) -8-azaspiro [4.5] decyclobutane-1-yl) -2-methylpropane-2-sulfinamide
Figure BDA0003608939860000873
To a 100mL single-neck flask, crude ((R) -1- ((R) -1, 1-dimethylethylsulfonamido) -8-azaspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidine-8-thiolate (50mg, 0.12mmol), 3, 4-dichloropyridazine (19mg, 0.13mmol) and acetonitrile (3 mL) were added in this order under nitrogen, followed by DIPEA (31mg, 0.24mmol), and the reaction solution was stirred at 80 ℃ for 16 hours. After the reaction liquid was cooled, the obtained residue was condensed under reduced pressure to obtain (R) -N- ((R) -8- (8- ((3-chloropyridazin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-cyclobutan-1-yl) -2-methylpropane-2-sulfinamide (10 mg, yield: 16%) by silica gel chromatography (gradient of ethyl acetate/methanol from 0 to 30%).
LCMS:m/z 520.2[M+H] + .
Step two (R) -8- (8- ((3-chloropyridazin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000881
To a 50mL single-neck flask were added sequentially (R) -N- ((R) -8- (8- ((3-chloropyridazin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide (10mg, 0.02mmol) and methanol (0.5 mL) under a nitrogen blanket, a solution of 1, 4-dioxane hydrochloride (0.05mL, 4M) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1 hour. After completion of the reaction, it was cooled to room temperature, filtered, and the residue obtained by concentration under reduced pressure was purified by high performance liquid preparative chromatography to give (R) -8- (8- ((3-chloropyridazin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine (2 mg, yield: 24%).
1 H NMR(400MHz,DMSO-d6)δ8.79(d,J=5.4Hz,1H),8.36(s,2H),8.12(s,1H),7.84(s,1H),7.58(s,1H),7.01(d,J=5.4Hz,1H),3.88(d,J=9.4Hz,2H),3.23(d,J=11.6Hz,2H),2.76(t,J=7.2Hz,1H),1.70-1.32(m,9H);LCMS:m/z 416.1[M+H] + .
Following the synthetic procedure of example 117, the following compounds can be synthesized:
example 118 (R) -8- (8- ((2-Chloropyrimidin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000882
1 H NMR(400MHz,DMSO-d6)δ8.37(d,J=5.5Hz,1H),8.29(s,1H),8.09(s,1H),7.84(s,1H),7.61(d,J=1.2Hz,1H),7.06(d,J=5.5Hz,1H),3.93(t,J=12.9Hz,2H),3.25(s,2H),3.09(s,1H),2.03(s,1H),1.81(dd,J=19.3,13.3Hz,4H),1.65-1.46(m,5H)ppm;LCMS:m/z 416.1[M+H] + .
EXAMPLE 119 preparation of the Compound (R) -8- (8- (pyridin-2-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
The method comprises the following steps: (R) -2-methyl-N- (R) -8- (8- (pyridin-2-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-piperazin-1-yl) propane-2-sulfinamide
Figure BDA0003608939860000883
To a 100mL single-neck flask, under nitrogen, was added (R) -N- ((R) -8- (8-iodoimidazo [1, 2-c) in sequence]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-methylpropane-2-sulfonamide (50mg, 0.10mmol), pyridine-2-thiol (13mg, 0.12mmol), cu (OTf) 2 (4mg, 0.01mmol), BINAM (3mg, 0.01mmol) and 1, 4-dioxane (3 mL), followed by addition of cesium carbonate (65mg, 0.2mmol), the reaction was heated at 100 ℃ with stirring for 16h. After cooling the reaction mixture, the residue obtained by decompression was purified by silica gel chromatography (0 to 50% gradient of ethyl acetate/methanol) to give (R) -2-methyl-N- (R) -8- (8- (pyridin-2-ylthio) imidazo [1, 2-c)]Pyrimidin-5-yl) -8-azaspiro [4.5]Decapiperazin-1-yl) propane-2-sulfinamide (20 mg, yield: 41%).
LCMS:m/z 485.2[M+H] + .
Step two: (R) -8- (8- (pyridin-2-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000891
Using the same procedure as in step two of example 37, (R) -2-methyl-N- (R) -8- (8- (pyridin-2-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decanepiperazin-1-yl) propane-2-sulfinylamine was desulfulfinylated to give (R) -8- (8- (pyridin-2-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine. .
1 H NMR(400MHz,DMSO-d6)δ8.34(d,J=3.1Hz,2H),8.02(s,1H),7.78(s,1H),7.63-7.48(m,2H),7.11(dd,J=7.3,4.9Hz,1H),6.93(d,J=8.1Hz,1H),3.86(t,J=13.0Hz,2H),3.22(t,J=12.4Hz,2H),3.03(t,J=6.5Hz,1H),2.04-1.35(m,10H)ppm;LCMS:m/z 381.2[M+H] + .
Following the synthesis of example 119, the following compounds may be synthesized:
example 120 (R) -8- (8- (pyridazin-3-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000892
1 H NMR(400MHz,DMSO-d6)δ8.97(d,J=4.8Hz,1H),8.09(s,1H),7.80(s,1H),7.58(s,1H),7.48(dd,J=8.9,4.9Hz,1H),7.33(d,J=8.8Hz,1H),3.90(s,2H),3.25(d,J=12.2Hz,3H),3.18(s,1H),1.86-1.46(m,9H);LCMS:m/z 382.1[M+H] + .
Example 121 (R) -8- (8- (pyrazin-2-ylthio) imidazo [1,2-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000893
1 H NMR(400MHz,DMSO-d6)δ8.41(s,1H),8.36(s,1H),8.31(s,1H),8.07(s,1H),7.79(s,1H),7.58(s,1H),3.89(t,J=12.8Hz,2H),3.24(d,J=12.2Hz,2H),2.06-1.46(m,9H);LCMS:m/z 382.1[M+H] + .
Example 122: preparation of the compound 8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -1-methyl-8-azaspiro [4.5] decan-1-amine
The method comprises the following steps: 8- (8-Iodoimidazo [1,2-c ] pyrimidin-5-yl) -1-methylspiro [4.5] decan-1-amine
Figure BDA0003608939860000901
To a solution of 5-chloro-8-iodoimidazo [1,2-c ] pyrimidine (56mg, 0.2mmol) in anhydrous DMF (10 mL) at 0 ℃ was added 1-methyl-8-azaspiro [4.5] decan-1-amine (40mg, 0.24mmol) followed by diisopropylethylamine (51.6 mg,0.4 mmol) and the reaction was stirred at 0 ℃ for 1h. After the reaction is finished, the reaction solution is directly used for the next reaction.
LCMS:m/z 412.0[M+H] + .
Step two: (8- (8-Iodoimidazo [1,2-c ] pyrimidin-5-yl) -1-methylspiro [4.5] decan-1-yl) carbamic acid tert-butyl ester
Figure BDA0003608939860000902
Adding (Boc) to the solution obtained in the previous step 2 O (87mg, 0.4 mmol) was then added diisopropylethylamine (51.6 mg,0.4 mmol), and the reaction solution was stirred at room temperature for 4 hours. After completion of the reaction, water was added to quench the reaction, followed by extraction with ethyl acetate (3X 10 mL). The combined organic phases were dried over anhydrous sodium sulfate. Filtered and concentrated under reduced pressure. The residue obtained is purified by chromatography on silica gel (gradient 0 to 30% ethyl acetate/petroleum ether) to give (8- (8-iodoimidazo [1, 2-c)]Pyrimidin-5-yl) -1-methylspiro [4.5]Decan-1-yl) carbamic acid tert-butyl ester (15 mg, two-step yield: 7%)
LC-MS:m/z=512.0[M+H] + .
Step three: tert-butyl (8- (8- ((6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -methylspiro [4.5] decan-1-yl) carbamate.
Figure BDA0003608939860000903
Following the synthesis of step one of example 76, (8- (8-iodoimidazo [1, 2-c)]Pyrimidin-5-yl) -1-methylspiro [4.5]Coupling of tert-butyl decan-1-yl) carbamate with sodium 6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-thiolate gave (8- (8- ((6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c]Pyrimidin-5-yl) -methylspiro [4.5]Decan-1-yl) carbamic acid tert-butyl ester (4 mg, yield: 15%). LC-MS m/z 678.2[ m ] +H] + .
Step four: 4- ((5- (1-amino-1-methylspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) sulfanyl) -5, 6-dichloropyridin-2-amine.
Figure BDA0003608939860000904
Following the synthesis of example 21 step three, (8- (8- ((6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -methylspiro [4.5] decan-1-yl) carbamic acid tert-butyl ester was de-Boc protected to give 4- ((5- (1-amino-1-methylspiro [4.5] decan-8-yl) imidazo [1,2-c ] pyrimidin-8-yl) thio) -5, 6-dichloropyridin-2-amine (2 mg, yield: 60%)
1 HNMR(400MHz,DMSO-d6)δ8.10(s,1H),7.87(s,1H),7.64(s,1H),6.42(s,2H),5.63(s,1H),4.06(s,2H),3.18(t,J=12.4Hz,2H),1.96-1.37(m,9H),1.26(s,3H),0.98-0.84(m,1H)ppm;LC-MS:m/z=478.2
[M+H] +
Following the synthesis of example 122, the following compounds may be synthesized:
example 123 (R) -8- (8- ((6-amino-3-chloro-2-methyl-pyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-difluoro-8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000911
1 H NMR(400MHz,DMSO-d 6 )δ8.19(s,2H),8.07(s,1H),7.84(s,1H),7.65(s,1H),5.65(s,1H),3.99(t,J=15.1Hz,2H),3.62-3.55(m,3H),2.74-2.63(m,2H),2.44-2.33(m,5H),2.01(t,J=11.8Hz,1H),1.85-1.60(m,3H)ppm;LC-MS:m/z=480.2[M+H] + .
Example 124: preparation of the compound (R) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-difluoro-8-azaspiro [4.5] decan-1-amine
Step one and step two: 5, 6-dichloro-4- ((5-chloroimidazo [1,2-c ] pyrimidin-8-yl) thio) pyridin-2-amine
Figure BDA0003608939860000912
Following the synthesis procedure of steps three and four in example 1, 8-iodoimidazo [1,2-c ] pyrimidin-5-ol was coupled with sodium 6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridine-4-thiolate and halogenated to give 5, 6-dichloro-4- ((5-chloroimidazo [1,2-c ] pyrimidin-8-yl) thio) pyridin-2-amine.
LC-MS:m/z 347.9[M+H] +
Step three: (R) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-difluoro-8-azaspiro [4.5] decan-1-amine
Figure BDA0003608939860000913
Following the synthesis procedure of example 23, substitution of 5, 6-dichloro-4- ((5-chloroimidazo [1,2-c ] pyrimidin-8-yl) thio) pyridin-2-amine with (R) -3, 3-difluoro-8-azaspiro [4.5] decan-1-amine gave (R) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -3, 3-difluoro-8-azaspiro [4.5] decan-1-amine.
1 H NMR(400MHz,DMSO-d6)δ8.35(s,2H),8.06(s,1H),7.86(s,1H),7.61(s,1H),6.31(s,2H),5.62(s,1H),3.92(d,J=14.9Hz,2H),3.21(d,J=12.8Hz,3H),3.07(t,J=8.1Hz,2H),2.12-1.98(m,3H),1.91(d,J=12.4Hz,2H),1.47(t,J=16.6Hz,3H)ppm;LC-MS:m/z 501.7[M+H] + .
Examples 125 to 127 pharmacologically relevant examples
Example 125: SHP2 enzyme Activity inhibition assay
The compound powder was dissolved in DMSO to prepare a mother solution. In the experiments, compound stock solutions were diluted in DMSO in 3-fold gradients, with 10 different test concentrations set for the same compound. mu.L of each concentration point of compound was dispensed into assay plate (Corning, costar 3915) wells, and 2 replicates were placed at each concentration point. The protein is active protein SHP2 with 76 th amino acid mutation E76A The substrate used was DiFMUP (Invitrogen, E12020). SHP2 E76A The protein and substrate were diluted to 1.2nM and 20. Mu.M, respectively, with buffer (0.1M NaAc (pH 7.2), 0.02% Tween 20,0.1% BSA,1mM EDTA,5mM DTT). To the assay well, 50. Mu.L of enzyme solution was added followed by 50. Mu.L of substrate. The rate of accumulation of the product was calculated to characterize the enzyme activity by recording (Ex 358nm/Em 455 nm) the fluorescence signal every 1 minute on a Spectra max i3 (Molecular Devices) instrument. Nonlinear regression analysis was performed with GraphPad Prism 5, and a curve of enzyme activity as a function of compound concentration was fitted by the Y = Bottom + (Top-Bottom)/(1 +10^ ((LogIC 50-X) } HillSlope)) equation. Determination of IC of each Compound 50 The value is obtained.
Results
The following table shows the IC of some of the compounds of the invention 50 The value is obtained.
Letter A stands for IC 50 Less than 100nM;
letter B stands for IC 50 100nM to 1000nM;
Figure BDA0003608939860000921
Figure BDA0003608939860000931
Figure BDA0003608939860000941
example 126: phosphoprotein kinase (p-ERK) cell assay
The phosphorylation level of a compound inhibiting intracellular protein kinase (ERK) was examined by AlphaLISA method.
The first step is the treatment of the cells with the compound. The test compounds were first diluted 3-fold with 100% dmso, setting up a total of 9 different concentration gradients; then, 30000 cells per hole are used for inoculating MOLM13 cells to a 96-hole plate, and each hole volume is 100 mu L; subsequently, 0.5. Mu.L of DMSO or different concentrations of test compound were added to each well, each concentration was set at 2 replicates, and the final concentration of DMSO was controlled at 0.5%.
The second step lyses the cells. After 2 hours of cell treatment, the medium was removed and the cells were washed 3 times with phosphate buffered saline, 50 μ l of freshly prepared lysis buffer was added to each well, shaken and left at room temperature for 10 minutes.
The third step
Figure BDA0003608939860000943
Ultra TM p-ERK 1/2 (Thr 202/Tyr 204) kit (Perkin Elmer, ALSU-PERK-A10K)) detects phosphorylated extracellular signal-regulated kinase (p-ERK). Mu.l of the above lysate was applied to 384 well plates (Perkin Elmer, 6005350) and the samples were tested for the level of extracellular signal-regulated kinase phosphorylation according to the product instructions. Signals were read using an AlphaScreen detector on Spectra max i3 (Molecular Devices). The inhibition percentage (%) was obtained by calculating the following formula:
percent (%) inhibition = (1-p-ERK signal from compound treated cells/p-ERK signal from DMSO treated cells) = 100
Results
The following table shows the IC of some of the compounds of the invention 50 The value is obtained.
Letter A stands for IC 50 Less than 100nM;
letter B stands for IC 50 100nM to 1000nM;
Figure BDA0003608939860000942
Figure BDA0003608939860000951
example 127: MOLM-13 cell proliferation assay
MOLM-13 cells suspended in medium (RPMI-1640, containing 10% FBS and 1% Penicillin-Streptomyces, gibco) were seeded onto 384 well plates at 800 cells (40. Mu.L/well). The cells were immediately treated with the test compounds at concentrations of 50, 16.67,5.56,1.85,0.617,0.206,0.069,0.023,0.0076 μm, respectively. After 3 days, 5. Mu.L of CellTiter-Glo reagent (Promega, ZG 7572) was added to each well, and the mixture was left for 10 minutes at room temperature in the dark. Fluorescence signals were detected by Spectra max i3 (Molecular Devices). The relative growth rate of the treated cells was compared to the DMSO control.
As a result, the
The following table shows the IC of some of the compounds of the invention 50 The value is obtained.
Letter A stands for IC 50 Less than 100nM;
letter B stands for IC 50 100nM to 1000nM;
Figure BDA0003608939860000961
Figure BDA0003608939860000971
according to the same test method as SHP2 enzyme activity inhibition test described in example 125, phosphorylation protein kinase (p-ERK) cell test described in example 126 and MOLM-13 cell proliferation test described in example 127, the applicant conducted corresponding experiments with respect to WO2015/107493A1 or the compound SHP099 (6- (4-amino-4-methylpiperidin-1-yl) -3- (2, 3-dichlorophenyl) pyrazine-2-amine) disclosed in the literature (Nature 2016,535, 148-152), and the following table lists the comparative experimental data of the compound obtained in some examples of the present invention and SHP099, and by comparison, the pyrimido-cyclic compound of the present invention has more excellent activity.
Figure BDA0003608939860000972
Figure BDA0003608939860000981
The above embodiments are preferred embodiments of the present invention, but the present invention is not limited to the above embodiments, and any other changes, modifications, substitutions, combinations, and simplifications which do not depart from the spirit and principle of the present invention should be construed as equivalents thereof, and all such changes, modifications, substitutions, combinations, and simplifications are intended to be included in the scope of the present invention.

Claims (25)

1. A pyrimido-cyclic compound represented by formula (I), a pharmaceutically acceptable salt thereof or a solvate thereof,
Figure FDA0003608939850000011
wherein
Z 1 、Z 2 Simultaneously is C or one of them is N;
x is independently S or absent;
y is independently C or N;
n is independently 0, 1 or 2;
R 1 independently 0 to 4R 1a Substituted phenyl, 0 to 4R 1a Substituted heteroaryl groups containing 1-4 nitrogen heteroaryl groups, 0 to 4R 1a Substituted naphthyl, 0 to 4R 1a Substituted azanaphthalene aryl containing 1-4, 0 to 4R 1a Substituted or unsubstituted benzoheterocycles, 0 to 4R 1a Substituted or unsubstituted with 1-4 azaaromatic rings, 0 to 4R 1a Substituted containing 1-4N,NR 1b Hetero aromatic ring of hetero atom such as O or S (O) m, R 1c Substituted or unsubstituted C 1-8 Alkyl, R 1c Substituted or unsubstituted C 1-8 A haloalkyl group; wherein m is selected from 0, 1 and 2;
R 1a independently of one another is halogen, R 1a1 Substituted or unsubstituted C 1-4 Alkoxy radical, R 1a1 Substituted or unsubstituted C 1-4 Alkyl, trifluoromethyl, C (= O) OR 1a2 、NR 1a2 R 1a3 、NHC(=O)R 1a4 、R 1a1 Substituted or unsubstituted C 3-8 A cycloalkyl group; r 1a1 Independently halogen or C 1-4 An alkyl group; r 1a2 、R 1a3 Independently of one another is hydrogen, C 1-4 An alkyl group; r 1a4 Independently is C 1-4 Alkyl, substituted or unsubstituted alkenyl, amide, C 3-12 Mono-or poly-heterocyclic;
R 1b independently of each other is hydrogen, R 1a1 Substituted or unsubstituted C 1-4 An alkyl group;
R 1c independently hydrogen, -C (= O) OR 1a2 、R 1a1 Substituted or unsubstituted C 1-4 An alkyl group;
R 2a 、R 2b 、R 3a and R 3b Independently of each other is hydrogen, R 1a1 Substituted or unsubstituted C 1-4 An alkyl group;
when Y = N, R 4 Independently of each other is hydrogen, R 1a1 Substituted or unsubstituted C 1-4 An alkyl group; r is 5 Is absent;
when Y = C, R 4 、R 5 Independently of each other hydrogen, aryl, C 1-4 Alkyl radical, C 1-4 Alkoxy, -O-C 1-4 Alkyl, amino, C 1-4 Alkyl substituted amino, -O-C 1-4 Alkyl-substituted amino, or R 4 And R 5 Together with Y form 0 to 3R 4a A substituted 3-to 7-membered saturated or partially unsaturated spirocyclic ring, which ring may optionally contain 1 to 3 heteroatoms or groups independently selected from N, C (= O) and/or O;
R 4a independently hydrogen, halogen, R 1a1 Substituted or unsubstituted C 1-4 Alkoxy radicalBase, R 1a1 Substituted or unsubstituted C 1-4 Alkyl, hydroxy, amino, C 1-4 An alkylamino group.
2. The pyrimido compound, the pharmaceutically acceptable salt thereof, or the solvate thereof according to claim 1, wherein R 1 Selected from the following structures:
Figure FDA0003608939850000021
wherein o is 0, 1,2,3 or 4; ring a is a heteroaryl group containing 1 to 4N atoms; ring B is heteroaryl containing 1 to 4 heteroatoms such as N, S, O and the like; g is independently a heteroatom or group such as C, C (= O), N, S or O; r 1aa 、R 1ab Independently R as defined in claim 1 1a ;R 1ac Independently is R 1c Substituted or unsubstituted C 1-8 Alkyl radical, R 1c Substituted or unsubstituted C 1-8 An alkyl halide.
3. The pyrimido compound, the pharmaceutically acceptable salt thereof, or the solvate thereof according to claim 1, wherein R 2a 、R 2b 、R 3a And R 3b Independently hydrogen or methyl.
4. The pyrimido compound, the pharmaceutically acceptable salt thereof, or the solvate thereof according to claim 1, wherein R when Y = N 4 Independently hydrogen, methyl; r 5 Is absent.
5. The pyrimido ring compound, the pharmaceutically acceptable salt thereof, or the solvate thereof according to claim 1, wherein R is R when Y = C 4 、R 5 Independently hydrogen, methyl, ethyl, phenyl, amino, methylamino or ethylamino.
6. The pyrimido ring compound of claim 1, a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt thereofSolvate, wherein R when Y = C 4 And R 5 The ring formed with Y is selected from the following structures:
Figure FDA0003608939850000022
wherein p is 0, 1,2 or 3; r is 4a As defined in claim 1.
7. The pyrimido ring compound, the pharmaceutically acceptable salt thereof, or the solvate thereof according to claim 1, wherein R is R when Y = C 4 And R 5 The ring formed with Y is of the following configuration:
Figure FDA0003608939850000023
wherein p is 0, 1,2 or 3; r is 4a As defined in claim 1.
8. The pyrimido compound of claim 1, having the formula (II)
Figure FDA0003608939850000031
9. The pyrimido compound, the pharmaceutically acceptable salt thereof, or the solvate thereof according to claim 8 is selected from any one of the following compounds:
Figure FDA0003608939850000032
Figure FDA0003608939850000041
Figure FDA0003608939850000051
Figure FDA0003608939850000061
10. an isotopically labeled compound of the pyrimido compound, the pharmaceutically acceptable salt thereof or the solvate thereof according to any one of claims 1 to 9, wherein the isotope is selected from the group consisting of 2 H、 3 H、 11 C、 13 C、 14 C、 15 N、 18 F、 31 P、 32 P、 35 S、 36 Cl and 125 I。
11. a method for producing a pyrimido-cyclic compound represented by formula (II), which is method 1 or method 2:
the method 1 comprises the following steps:
coupling reaction of halogenated intermediate compound A with boric acid, thiol or sodium sulfide (F) gives formula (II), the reaction equation is as follows:
Figure FDA0003608939850000071
wherein, W 1 Represents halogen, preferably Br, I; x, Y, n, R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 As defined in claim 1;
the method 2 comprises the following steps:
substitution of the halogenated intermediate compound B with amine C affords formula (II) as follows:
Figure FDA0003608939850000072
wherein, W 2 Represents halogen, preferably Cl, br, I; x, Y, n, R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined in claim 1.
12. A process for the preparation of intermediate compounds A, B or D, wherein,
the preparation method of the compound A comprises the following steps:
the halogenated intermediate E is substituted by the intermediate amine C under the alkaline condition to obtain a halogenated intermediate compound A, and the reaction equation is as follows:
Figure FDA0003608939850000073
wherein, Y, n, R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 As defined in claim 1; w 1 Represents halogen, preferably Br, I;
the preparation method of the compound B comprises the following steps:
substituting dichloro pyrimidine compound B-1 with amine to obtain intermediate B-2; condensing, cyclizing and hydrolyzing the intermediate B-2 under the condition of strong acid to obtain a halogenated intermediate B-3; halogenated intermediate B-3 is subjected to catalytic coupling to obtain intermediate B-4, and then converted into halogenated intermediate B, wherein the reaction equation is as follows:
Figure FDA0003608939850000081
wherein, X and R 1 As defined in claim 1; w 1 Represents halogen, preferably Br, I; w is a group of 2 Represents halogen, preferably Cl, br, I;
the preparation method of the compound D comprises the following steps:
the intermediate compound A and methyl mercaptopropionate obtain an intermediate D-1 under the condition of catalytic coupling, and then obtain a corresponding sodium sulfide compound D under the alkaline condition, wherein the reaction equation is as follows:
Figure FDA0003608939850000082
wherein, Y, n, R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 As defined in claim 1; w 1 Represents halogen, preferably Br, I.
13. A process for producing a pyrimido-cyclic compound represented by formula (II-A), comprising the steps of:
coupling the sodium sulfur intermediate compound D with a halide to obtain a compound of formula (II-A), wherein the reaction equation is as follows:
Figure FDA0003608939850000083
wherein, Y, n, R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined in claim 1.
14. A process for producing a pyrimido-cyclic compound represented by formula (II-B), comprising the steps of:
removing the protecting group of the intermediate II-B1 under acidic or basic conditions to obtain a compound II-B, wherein the reaction equation is as follows:
Figure FDA0003608939850000091
wherein Pg is selected from protecting groups Boc, ac, S (= O) t Bu;R 4Pg 、R 5Pg Together with the attached carbon, is selected from the following structures:
Figure FDA0003608939850000092
R 4 、R 5 together with the attached carbon, is selected from the following structures:
Figure FDA0003608939850000093
X、n、R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 、R 5 as defined in claim 1; r is 4a As defined in claim 1; p is 0, 1,2 or 3.
15. A process for producing a pyrimido-cyclic compound represented by formula (II-C), comprising the steps of:
after aminoacylation of the intermediate II-C1, a compound II-C is obtained, and the reaction equation is as follows:
Figure FDA0003608939850000094
wherein, X, Y, n, R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 、R 5 、R 1a And R 1a4 Is as defined in claim 1.
16. An intermediate for preparing the pyrimido-ring compound of claim 8, said intermediate compound selected from the group consisting of:
Figure FDA0003608939850000095
Figure FDA0003608939850000101
wherein X, Y, n, R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Is as defined in claim 1; w 1 Represents halogen, preferably Br, I; w is a group of 2 Represents halogen, preferably Cl, br, I.
17. A method for producing a pyrimido-cyclic compound represented by formula (I), which is method 1 or method 2:
the method 1 comprises the following steps:
halogenated intermediate compounds
Figure FDA0003608939850000102
After the coupling reaction with boric acid, mercaptan or sodium sulfide (F), the formula (I) is obtained, and the reaction equation is as follows:
Figure FDA0003608939850000103
wherein, W 1 Represents halogen, preferably Br, I; z is a linear or branched member 1 、Z 2 、X、Y、n、R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 As defined in claim 1;
the method 2 comprises the following steps:
intermediate product
Figure FDA0003608939850000104
Substitution with amine C gives formula (I), the reaction equation is as follows:
Figure FDA0003608939850000105
wherein, W 2 Represents halogen, preferably Cl, br, I; z is a linear or branched member 1 、Z 2 、X、Y、n、R 1 、R 2a 、R 2b 、R 3a 、R 3b 、R 4 And R 5 Definition of (1)As claimed in claim 1.
18. Use of a pyrimido compound according to any of claims 1 to 9, a pharmaceutically acceptable salt thereof, a solvate thereof or an isotopically labeled compound according to claim 10 for the preparation of a medicament for the treatment of a disease or disorder associated with abnormal SHP2 activity.
19. A disease or disorder according to claim 18 selected from noonan syndrome, leopard syndrome, juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myelogenous leukemia, breast cancer, esophageal cancer, lung cancer, colon cancer, head cancer, neuroblastoma, squamous cell carcinoma of the head and neck, gastric cancer, anaplastic large-cell lymphoma or glioblastoma.
20. A pharmaceutical composition comprising a pyrimido-ring compound according to any one of claims 1 to 9, a pharmaceutically acceptable salt thereof or a solvate thereof, or an isotopically labeled compound of claim 10, and a pharmaceutically acceptable excipient.
21. A pharmaceutical formulation comprising a pyrimido compound of any of claims 1 to 9, a pharmaceutically acceptable salt thereof or a solvate thereof, or an isotopically labeled compound of claim 10, selected from the group consisting of tablets, capsules, pills, powders, granules, elixirs, tinctures, suspensions, syrups, emulsions, solutions.
22. A pharmaceutical formulation according to claim 21, administered in a manner selected from the group consisting of: oral, sublingual, subcutaneous, intravenous, intramuscular, intrasternal, nasal, topical or rectal administration, and/or said pharmaceutical formulation is administered in a single or multiple daily doses.
23. A pyrimido compound according to any of claims 1 to 9, a pharmaceutically acceptable salt thereof or a solvate thereof or an isotopically labeled compound according to claim 10, in combination with a further drug selected from the group consisting of: anticancer drugs, tumor immunity drugs, antiallergic drugs, antiemetic drugs, analgesics, cytoprotective drugs.
24. A compound selected from
Figure FDA0003608939850000111
Wherein U is independently C or O; q is selected from 0, 1 or 2; pg is selected from protecting groups Boc, ac, S (= O) t Bu;n、R 2a 、R 2b 、R 3a 、R 3b And R 4a As defined in claim 1; r 6 Independently is C 1-8 Alkyl, substituted or unsubstituted aryl, substituted or unsubstituted alkenyl.
25. A process for the preparation of a compound according to claim 24,
the preparation method of the compound C-1 comprises the following steps:
the spirocyclic ketone compound C-1a is subjected to reductive amination to obtain an intermediate C-1b; c-1b is obtained after selective deprotection, and the reaction equation is as follows:
Figure FDA0003608939850000112
wherein Pg1 is selected from protecting groups Boc, benzoyl and benzyl; pg, U, q, n, R 2a 、R 2b 、R 3a 、R 3b And R 4a As defined in claim 24;
the preparation method of the compound C-2 comprises the following steps:
spirocyclic ketone compounds C-1a and R 6 The hydroxyl compound C-2a is obtained by the addition of the substituted nucleophilic reagent; converting the compound C-2a into an amino compound C-2b, and then selectively removing a protecting group Pg1 to obtainC-2, the reaction equation is as follows:
Figure FDA0003608939850000121
wherein Pg1 is selected from protecting groups of Boc, benzoyl and benzyl; r is 6 、U、q、Pg、n、R 2a 、R 2b 、R 3a 、R 3b And R 4a As defined in claim 24;
the preparation method of the compound C-3 comprises the following steps:
ester group ortho-dehydrogenation of Compound C-3a with R 6 The substituted electrophile is substituted to obtain a compound C-3b; hydrolyzing the ester group by the compound C-3b to obtain acid C-3C; rearranging the acid C-3C to obtain amine C-3d, and then selectively removing the protecting group Pg1 to obtain C-3, wherein the reaction equation is as follows:
Figure FDA0003608939850000122
wherein Pg1 is selected from protecting groups Boc, benzoyl and benzyl; r 6 、Pg、n、R 2a 、R 2b 、R 3a And R 3b As defined in claim 24;
the preparation method of the compound C-4 comprises the following steps:
reducing cyano compound C-4a and protecting amino to obtain intermediate C-4b, then selectively removing protecting group Pg1 to obtain C-4, wherein the reaction equation is as follows:
Figure FDA0003608939850000123
wherein Pg1 is selected from protecting groups of Boc, benzoyl and benzyl; pg, R 6 、n、R 2a 、R 2b 、R 3a And R 3b Is as defined in claim 24.
CN202210427468.3A 2018-02-13 2018-02-13 Pyrimidine-fused ring compound, preparation method and application thereof Active CN115448923B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210427468.3A CN115448923B (en) 2018-02-13 2018-02-13 Pyrimidine-fused ring compound, preparation method and application thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210427468.3A CN115448923B (en) 2018-02-13 2018-02-13 Pyrimidine-fused ring compound, preparation method and application thereof
CN201810144135.3A CN110156786B (en) 2018-02-13 2018-02-13 Pyrimido-cyclic compounds, process for their preparation and their use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN201810144135.3A Division CN110156786B (en) 2018-02-13 2018-02-13 Pyrimido-cyclic compounds, process for their preparation and their use

Publications (2)

Publication Number Publication Date
CN115448923A true CN115448923A (en) 2022-12-09
CN115448923B CN115448923B (en) 2024-03-22

Family

ID=67635115

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201810144135.3A Active CN110156786B (en) 2018-02-13 2018-02-13 Pyrimido-cyclic compounds, process for their preparation and their use
CN202210427468.3A Active CN115448923B (en) 2018-02-13 2018-02-13 Pyrimidine-fused ring compound, preparation method and application thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN201810144135.3A Active CN110156786B (en) 2018-02-13 2018-02-13 Pyrimido-cyclic compounds, process for their preparation and their use

Country Status (1)

Country Link
CN (2) CN110156786B (en)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2019000548A (en) 2016-07-12 2019-10-30 Revolution Medicines Inc 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric shp2 inhibitors.
CA3051054A1 (en) 2017-01-23 2018-07-26 Revolution Medicines, Inc. Pyridine compounds as allosteric shp2 inhibitors
MX2019008695A (en) 2017-01-23 2019-09-11 Revolution Medicines Inc Bicyclic compounds as allosteric shp2 inhibitors.
RU2020115095A (en) 2017-10-12 2021-11-12 Революшн Медсинз, Инк. PYRIDINE, PYRAZINE AND TRIAZINE COMPOUNDS AS ALLOSTERIC SHP2 INHIBITORS
SG11202004090YA (en) 2017-12-15 2020-05-28 Revolution Medicines Inc Polycyclic compounds as allosteric shp2 inhibitors
CN114751903B (en) * 2018-11-07 2023-09-15 上海凌达生物医药有限公司 Nitrogen-containing fused heterocycle SHP2 inhibitor compound, preparation method and application
BR112021009880A2 (en) * 2018-11-30 2021-08-17 Tuojie Biotech (Shanghai) Co., Ltd. pyrimidine and derivative of five-membered nitrogen heterocycle, method of preparation thereof and medical uses thereof
CN112300160A (en) * 2019-08-01 2021-02-02 上海奕拓医药科技有限责任公司 Spiro aromatic ring compound, preparation and application thereof
CN112574212B (en) * 2019-09-30 2022-04-19 上海拓界生物医药科技有限公司 Pyrimido five-membered nitrogen heterocyclic derivative, preparation method and medical application thereof
EP4056563A4 (en) * 2019-11-08 2024-02-21 Nanjing Sanhome Pharmaceutical Co Ltd Compound as shp2 inhibitor and use thereof
WO2021121397A1 (en) * 2019-12-19 2021-06-24 首药控股(北京)股份有限公司 Substituted alkynyl heterocyclic compound
EP4165033A1 (en) * 2020-06-11 2023-04-19 Betta Pharmaceuticals Co., Ltd Shp2 inhibitors, compositions and uses thereof
CN112062699B (en) * 2020-11-13 2021-02-26 苏州开元民生科技股份有限公司 Preparation method of o-aminothiophenol
CN113372344B (en) * 2020-12-24 2022-11-01 上海药坦药物研究开发有限公司 Synthesis method of chloro-hexatomic nitrogen-containing heterocyclic imidazole compound
WO2022135568A1 (en) * 2020-12-25 2022-06-30 江苏恒瑞医药股份有限公司 Crystal form of pyrimido five-membered nitrogen heterocyclic derivative and preparation method therefor
WO2023109761A1 (en) * 2021-12-15 2023-06-22 贝达药业股份有限公司 Crystal of pyrazolopyrimidinone compound and salt thereof

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6921762B2 (en) * 2001-11-16 2005-07-26 Amgen Inc. Substituted indolizine-like compounds and methods of use
WO2006136830A1 (en) * 2005-06-21 2006-12-28 Astex Therapeutics Limited Aryl-alkylamines and heteroaryl-alkylamines as protein kinase inhibitors
WO2010114881A1 (en) * 2009-04-01 2010-10-07 Progenra Inc. Anti-neoplastic compounds, compositions and methods
CN102869666A (en) * 2010-03-10 2013-01-09 凯利普西斯公司 Heterocyclic inhibitors of histamine receptors for the treatment of disease
CN104350055A (en) * 2012-03-30 2015-02-11 新加坡科技研究局 Bicyclic heterocyclic derivatives as MNK1 and MNK2 modulators and uses thereof
CN105263929A (en) * 2013-03-14 2016-01-20 诺华股份有限公司 3-pyrimidin-4-yl-oxazolidin-2-ones as inhibitors of mutant idh
CN105899491A (en) * 2014-01-17 2016-08-24 诺华股份有限公司 1 -pyridazin-/triazin-3-yl-piper(-azine)/idine/pyrolidine derivatives and and compositions thereof for inhibiting the activity of SHP2
CN105916845A (en) * 2014-01-17 2016-08-31 诺华股份有限公司 N-azaspirocycloalkane substituted n-heteroaryl compounds and compositions for inhibiting the activity of shp2
WO2016203404A1 (en) * 2015-06-19 2016-12-22 Novartis Ag Compounds and compositions for inhibiting the activity of shp2
WO2018013597A1 (en) * 2016-07-12 2018-01-18 Revolution Medicines, Inc. 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric shp2 inhibitors
WO2020072656A1 (en) * 2018-10-03 2020-04-09 Gilead Sciences, Inc. Imidozopyrimidine derivatives
WO2020094018A1 (en) * 2018-11-06 2020-05-14 上海奕拓医药科技有限责任公司 Spiro aromatic ring compound and application thereof
CN111433205A (en) * 2017-12-15 2020-07-17 锐新医药公司 Polycyclic compounds as allosteric SHP2 inhibitors
AU2019386036A1 (en) * 2018-11-30 2021-05-27 Tuojie Biotech (Shanghai) Co., Ltd. Pyrimidine and five-membered nitrogen heterocycle derivative, preparation method therefor, and medical uses thereof

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6921762B2 (en) * 2001-11-16 2005-07-26 Amgen Inc. Substituted indolizine-like compounds and methods of use
WO2006136830A1 (en) * 2005-06-21 2006-12-28 Astex Therapeutics Limited Aryl-alkylamines and heteroaryl-alkylamines as protein kinase inhibitors
WO2010114881A1 (en) * 2009-04-01 2010-10-07 Progenra Inc. Anti-neoplastic compounds, compositions and methods
CN102869666A (en) * 2010-03-10 2013-01-09 凯利普西斯公司 Heterocyclic inhibitors of histamine receptors for the treatment of disease
CN104350055A (en) * 2012-03-30 2015-02-11 新加坡科技研究局 Bicyclic heterocyclic derivatives as MNK1 and MNK2 modulators and uses thereof
CN105263929A (en) * 2013-03-14 2016-01-20 诺华股份有限公司 3-pyrimidin-4-yl-oxazolidin-2-ones as inhibitors of mutant idh
CN105899491A (en) * 2014-01-17 2016-08-24 诺华股份有限公司 1 -pyridazin-/triazin-3-yl-piper(-azine)/idine/pyrolidine derivatives and and compositions thereof for inhibiting the activity of SHP2
CN105916845A (en) * 2014-01-17 2016-08-31 诺华股份有限公司 N-azaspirocycloalkane substituted n-heteroaryl compounds and compositions for inhibiting the activity of shp2
WO2016203404A1 (en) * 2015-06-19 2016-12-22 Novartis Ag Compounds and compositions for inhibiting the activity of shp2
WO2018013597A1 (en) * 2016-07-12 2018-01-18 Revolution Medicines, Inc. 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric shp2 inhibitors
CN111433205A (en) * 2017-12-15 2020-07-17 锐新医药公司 Polycyclic compounds as allosteric SHP2 inhibitors
WO2020072656A1 (en) * 2018-10-03 2020-04-09 Gilead Sciences, Inc. Imidozopyrimidine derivatives
WO2020094018A1 (en) * 2018-11-06 2020-05-14 上海奕拓医药科技有限责任公司 Spiro aromatic ring compound and application thereof
AU2019386036A1 (en) * 2018-11-30 2021-05-27 Tuojie Biotech (Shanghai) Co., Ltd. Pyrimidine and five-membered nitrogen heterocycle derivative, preparation method therefor, and medical uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
COLUMBUS,OHIO,US: "REGISTRY[Online]", 《REGISTRY》, pages 2167558 - 61 *
YING HUANG,等: "Discovery of First-in-Class, Potent, and Orally Bioavailable Embryonic Ectoderm Development (EED) Inhibitor with Robust Anticancer Efficacy", JOURNAL OF MEDICINAL CHEMISTRY, vol. 60, pages 2215 - 2226, XP002772151, DOI: 10.1021/acs.jmedchem.6b01576 *
柴茜;沈强;马兰萍;王昕;孟韬;李静雅;李佳;沈竞康;: "基于四氢喹啉酸骈环戊烯骨架的蛋白酪氨酸磷酸酶1B抑制剂的设计、合成及构效关系", 高等学校化学学报, no. 02, pages 306 - 315 *

Also Published As

Publication number Publication date
CN115448923B (en) 2024-03-22
CN110156786B (en) 2022-06-03
CN110156786A (en) 2019-08-23

Similar Documents

Publication Publication Date Title
CN110156786B (en) Pyrimido-cyclic compounds, process for their preparation and their use
JP7335882B2 (en) Pyrimidine-condensed ring compound, method for producing the same, and use
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
EP3712151B1 (en) (s)-1&#39;-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4&#39;-piperidine]-5-amine as shp2 inhibitor for the treatment of cancer
CN105732636B (en) Heteroaromatic compounds and their use in medicine
KR101864561B1 (en) JAK inhibitors
WO2020108590A1 (en) Pyrimidine and five-membered nitrogen heterocycle derivative, preparation method therefor, and medical uses thereof
WO2021169990A1 (en) Kras inhibitors for treating cancers
CN111050765B (en) Spiro compounds and methods of making and using the same
CN115557974A (en) KRAS G12D inhibitor and application thereof
CN110655520A (en) Pyrimido-cyclic compounds, process for their preparation and their use
WO2016192630A1 (en) Compound having kinase inhibiting activity, method of preparing same, and use of same
JP2023534803A (en) 7-(piperidin-1-yl)-4H-pyrimido[1,2-B]pyridazin-4-one derivatives as positive allosteric modulators of the muscarinic acetylcholine receptor M4
CN115867346A (en) Kinase inhibitors
CN113527299A (en) Nitrogen-containing condensed ring compounds, preparation method and application
CN111655689B (en) Pyrazolopyridinone compounds
CN109790160B (en) Pyrido five-membered aromatic ring compound, preparation method and application thereof
CN114524810A (en) Pyrimidine heterocyclic compound, preparation method and application
CN114805361A (en) Amino-substituted aromatic heterocyclic pyrazole compounds, preparation method and application
CN114644631A (en) KRAS G12C protein mutation inhibitor, preparation method thereof, pharmaceutical composition and application thereof
CN111566104B (en) Spiro aromatic ring compound and application thereof
CN110229151B (en) Indolizine compound, preparation method and application thereof
CN116600808A (en) Tetrahydronaphthyridine derivative serving as KRAS mutant G12C inhibitor, and preparation method and application thereof
CN114478585A (en) Nitrogen-containing fused heterocyclic compound and preparation method and application thereof
US20240034731A1 (en) Aza-quinazoline compounds and methods of use

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20240131

Address after: Building 4, No. 999 Cailun Road, Pudong New Area, Shanghai, March 2012

Applicant after: SHANGHAI BLUERAY BIOPHARMA Co.,Ltd.

Country or region after: China

Address before: 201203 Building 3, No. 576, Libing Road, Pudong New Area, Shanghai

Applicant before: Qingyu pharmaceutical R & D (Shanghai) Co.,Ltd.

Country or region before: China

GR01 Patent grant
GR01 Patent grant