CN110655520A - 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
CN110655520A
CN110655520A CN201810692211.4A CN201810692211A CN110655520A CN 110655520 A CN110655520 A CN 110655520A CN 201810692211 A CN201810692211 A CN 201810692211A CN 110655520 A CN110655520 A CN 110655520A
Authority
CN
China
Prior art keywords
substituted
compound
independently
pyrimido
azaspiro
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.)
Pending
Application number
CN201810692211.4A
Other languages
Chinese (zh)
Inventor
邹斌
付贤磊
付文成
刘兰震
徐帅杰
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.)
Qingyu Pharmaceutical R & D Shanghai Co ltd
Original Assignee
Shanghai Qing Yu Medical Science And Technology 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 Shanghai Qing Yu Medical Science And Technology Co Ltd filed Critical Shanghai Qing Yu Medical Science And Technology Co Ltd
Priority to CN201810692211.4A priority Critical patent/CN110655520A/en
Priority to SG11202007740TA priority patent/SG11202007740TA/en
Priority to JP2020543947A priority patent/JP7335882B2/en
Priority to US16/969,392 priority patent/US11498930B2/en
Priority to AU2019222026A priority patent/AU2019222026B2/en
Priority to MA051845A priority patent/MA51845A/en
Priority to KR1020207026395A priority patent/KR102614939B1/en
Priority to EP19754599.9A priority patent/EP3753941A4/en
Priority to PCT/CN2019/074685 priority patent/WO2019158019A1/en
Publication of CN110655520A publication Critical patent/CN110655520A/en
Pending legal-status Critical Current

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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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

Abstract

The invention discloses a pyrimido-cyclic compound, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, a solvate or an isotopically labeled compound thereof, a preparation method of the compound, a composition containing the compound and application of the compound in preparing a medicament for preventing and/or treating diseases or symptoms related to abnormal activity of SHP2The application in the aspect of things.

Description

Pyrimido-cyclic compounds, process for their preparation and their use
Technical Field
The present invention discloses pyrimido compounds, pharmaceutically acceptable salts, hydrates, prodrugs, stereoisomers, solvates, or isotopically labeled compounds thereof. The invention also provides a preparation method of the compound and an intermediate compound thereof, a composition containing the compound and application of the compound in preparing a medicament for preventing and/or treating diseases or symptoms related to the abnormal activity of SHP 2.
Background
The tyrosine phosphatase SHP2 consists of two N-terminal Src homology 2 domains (N-SH2 and C-SH2) and a protein tyrosine phosphatase catalytic domain (PTP). In the basal state, N-SH2 can be combined with PTP to form a ring structure, so that the combination of PTP and 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, the PTP catalytic domain is released, and phosphatase activity is exerted.
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 related to 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(PD1) mediated immune system suppression. After binding of PD-1 from T cells to PD-L1, a large amount of SHP2 was recruited into the cells. SHP2 is capable of dephosphorylating an antigen receptor pathway protein within T cells, thereby inhibiting activation of T cells. Thus, inhibition of the activity of SHP2 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 WO2015/107493 a1, etc., indicate that SHP2 is attracting increasing attention as a novel druggable target. Surrounding the development of the SHP2 inhibitor, there are two major strategies for the development of inhibitors of the PTP catalytic domain of SHP2 and the development of allosteric inhibitors of the 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 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 WO2015/107493 a1 is to be further developed into an SHP2 inhibitor having a novel structure, good biological activity and high pharmaceutical activity.
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 drug-forming property 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, a hydrate, a prodrug, a stereoisomer, or a solvate thereof,
Figure BDA0001712865010000031
wherein
Z1Is C, Z2Is N or Z1Is N, Z2Is C;
x is independently S or absent;
y is independently C or N;
n is independently 0, 1 or 2;
R1independently 0 to 4R1aSubstituted phenyl, 0-4R1aSubstituted containing 1-4 azaaryl groups, 0-4R1aSubstituted naphthyl, 0-4R1aSubstituted azanaphthalene aryl containing 1-4, 0-4R1aSubstituted or unsubstituted benzo-heterocycle, 0-4R1aSubstituted or unsubstituted aromatic ring containing 1-4 nitrogen heterocyclic ring, 0-4R1aSubstituted containing 1-4N, NR1bHetero-aromatic ring of hetero atoms O or S (O), R1cSubstituted or unsubstituted C1-8Alkyl radical, R1cSubstituted or unsubstituted C1-8A haloalkyl group; wherein m is selected from 0, 1 and 2;
R1aindependently is halogen, R1a1Substituted or unsubstituted C1-4Alkoxy radical, R1a1Substituted or unsubstituted C1-4Alkyl, trifluoromethyl, C (═ O) OR1a2、NR1a2R1a3、NHC(=O)R1a4、R1a1Substituted or unsubstituted C3-8A cycloalkyl group; r1a1Independently is halogen or C1-4An alkyl group; r1a2、R1a3Independently is hydrogen, C1-4An alkyl group; r1a4Independently is C1-4Alkyl, substituted or unsubstituted alkenyl, amide, C3-12Mono-or poly-heterocyclic;
R1bindependently is hydrogen, R1a1Substituted or unsubstituted C1-4An alkyl group;
R1cindependently hydrogen, -C (═ O) OR1a2、R1a1Substituted or unsubstituted C1-4An alkyl group;
R2a、R2b、R3aand R3bIndependently is hydrogen, R1a1Substituted or unsubstituted C1-4An alkyl group;
when Y is N, R4Independently is hydrogen, R1a1Substituted or unsubstituted C1-4An alkyl group; r5Is absent;
when Y is ═ C, R4、R5Independently are hydrogen, aryl, C1-4Alkyl radical, C1-4Alkoxy, -O-C1-4Alkyl, amino, C1-4Alkyl substituted amino, -O-C1-4Alkyl-substituted amino, or R4And R5Together with Y form 0-3R4aSubstituted 3 to 7 membered saturated or partially unsaturatedA saturated spirocyclic ring, which ring may optionally contain 1-3 independent heteroatoms or groups selected from N, C (═ O) and/or O;
R4aindependently are hydrogen, halogen, R1a1Substituted or unsubstituted C1-4Alkoxy radical, R1a1Substituted or unsubstituted C1-4Alkyl, hydroxy, amino, C1-4An alkylamino group.
Among the preferred examples, R1Selected from the following structures:
Figure BDA0001712865010000041
wherein o is 0, 1,2,3 or 4; ring a is a heteroaryl group containing 1-4N atoms; ring B is a heteroaryl group containing 1-4 heteroatoms of N, S, O; g is independently C, C (═ O), N, S, or an O heteroatom or group; r1aa、R1abIndependently is R1a;R1acIndependently is R1cSubstituted or unsubstituted C1-8Alkyl radical, R1cSubstituted or unsubstituted C1-8An alkyl halide;
in another preferred embodiment, R2a、R2b、R3aAnd R3bIndependently hydrogen or methyl;
in another preferred embodiment, when Y ═ N, R4Independently hydrogen, methyl; r5Is absent;
in another preferred embodiment, when Y ═ C, R4、R5Independently hydrogen, methyl, ethyl, phenyl, amino, methylamino or ethylamino;
in another preferred embodiment, when Y ═ C, R4And R5The ring formed with Y is selected from the following structures:
Figure BDA0001712865010000051
wherein p is 0, 1,2 or 3; r4aAs defined above;
in another preferred embodiment, when Y ═ C, R4And R5The ring formed with Y isThe following configurations:
wherein, p and R4aAs defined above;
in another preferred embodiment, the compound is selected from any one of the following compounds:
Figure BDA0001712865010000053
Figure BDA0001712865010000061
Figure BDA0001712865010000071
Figure BDA0001712865010000081
Figure BDA0001712865010000101
Figure BDA0001712865010000111
Figure BDA0001712865010000121
Figure BDA0001712865010000131
Figure BDA0001712865010000151
in a second aspect, the present invention provides a pyrimido ring compound represented by formula (I), a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, a solvate, or an isotopically labeled compound 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 substituted2H、3H、11C、13C、14C、15N、18F、31P、32P、35S、36Cl and125i, and the like.
The invention also provides a preparation method of the pyrimido-cyclic compound shown in the formula (I) and an intermediate compound thereof, which mainly comprises the following steps:
the invention provides a preparation method of a formula (I), which comprises the following steps:
halogenated intermediate compounds
Figure BDA0001712865010000161
The coupling reaction with F gives the formula (I), the reaction equation is as follows:
Figure BDA0001712865010000162
wherein F represents boric acid, thiol or sodium sulfide;
W1represents halogen, preferably Br, I; x, Y, Z1、Z2、n、R1、R2a、R2b、R3a、R3b、R4And R5Is as defined above.
The present invention also provides a process for the preparation of compound I-B, comprising the steps of:
removing the amino protecting group of intermediate I-B1 under acidic or basic conditions to obtain compound I-B, wherein the reaction equation is as follows:
Figure BDA0001712865010000171
wherein, X, Z1、Z2、n、p、R1、R2a、R2b、R3a、R3b、R4、R5And R4aAs defined above; pg is selected from Boc, Ac, S (═ O) as protecting grouptBu;R4Pg、R5PgTogether with the linking carbon, is selected from the following structures:
Figure BDA0001712865010000172
R4、R5together with the linking carbon, is selected from the following structures:
the present invention also provides a process for the preparation of compounds I-C comprising the steps of:
aminoacylation of intermediate I-C1 yields compound I-C according to the following reaction equation:
wherein, X, Y, n, R2a、R2b、R3a、R3b、R4、R5、R1aAnd R1a4Is as defined above.
The invention also provides a compound A which is a compound A,
Figure BDA0001712865010000181
wherein, W1、R2a、R2b、R3a、R3b、R4、R5Y, n are as defined above.
The present invention also provides a process for the preparation of compound a comprising the steps of:
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 BDA0001712865010000182
wherein, W2Represents halogen, preferably Cl, Br; w1、Y、n、R2a、R2b、R3a、R3b、R4And R5Is as defined above.
The invention also provides a compound C-1,
wherein U is independently C or O; q is selected from 0, 1 or 2; pg is selected from Boc, Ac, S (═ O) as protecting grouptBu;n、R2a、R2b、R3a、R3bAnd R4aIs as defined above.
The present invention also provides a process for the preparation of compound C-1, comprising the steps of:
the spirocyclic ketone compound C-1a is reduced and aminated to obtain an intermediate C-1b, and C-1b is selectively deprotected to obtain C-1, wherein the reaction equation is as follows:
Figure BDA0001712865010000191
wherein Pg1 is selected from protecting groups of Boc, benzoyl and benzyl; pg, U, q, n, R2a、R2b、R3a、R3bAnd R4aIs as defined above.
The invention also provides a compound C-2,
wherein R is6Independent of each otherIs C1-8Alkyl, substituted or substituted aryl, substituted or substituted alkenyl; u, q, Pg, n, R2a、R2b、R3a、R3bAnd R4aIs as defined above;
the present invention also provides a process for the preparation of compound C-2, comprising the steps of:
spirocyclic ketone compounds C-1a and R6The substituted nucleophilic reagent is added to obtain a hydroxyl compound C-2 a; compound C-2a is converted into amino compound C-2b by Ritter reaction, and then the protecting group Pg1 is selectively removed to obtain C-2, wherein the reaction equation is as follows:
Figure BDA0001712865010000193
wherein R is6、U、q、Pg1、Pg、n、R2a、R2b、R3a、R3bAnd R4aIs as defined above;
the invention also provides a compound C-3,
wherein R is6、Pg、n、R2a、R2b、R3aAnd R3bIs 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 R6The substituted electrophile is substituted to obtain a compound C-3 b; hydrolyzing the ester group by the compound C-3b to obtain acid C-3C; rearrangement of acid C-3C to obtain amine C-3d, selective removal of protecting group Pg1 to obtain C-3, the reaction equation is as follows:
Figure BDA0001712865010000202
wherein R is6、Pg1、Pg、n、R2a、R2b、R3aAnd R3bIs as defined inAs described above.
The invention also provides a compound C-4,
Figure BDA0001712865010000203
wherein R is6、Pg、n、R2a、R2b、R3aAnd R3bIs 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, and then selectively removing protecting group Pg1 to obtain C-4, wherein the reaction equation is as follows:
wherein Pg1, Pg, R6、n、R2a、R2b、R3aAnd R3bIs as defined above.
The present invention also provides a compound E,
Figure BDA0001712865010000212
wherein, W1Represents halogen, preferably Br, I; w2Represents halogen, preferably Cl, Br, I;
the invention also provides a preparation method of the compound E, which comprises the following steps:
substituting hydrazine for the 4-chloropyrimidine compound E-1 to obtain an intermediate E-2; condensing and cyclizing the intermediate E-2 to obtain a halogenated intermediate E, wherein the reaction equation is as follows:
Figure BDA0001712865010000213
wherein, W1、W2Is as defined above.
The present invention also provides a compound of formula (I),
Figure BDA0001712865010000214
wherein, V is independently C or N; r1aIs as defined above.
The present invention also provides a process for the preparation of compound F-1 comprising the steps of:
halogenated compound F-1a and methyl mercaptopropionate are subjected to catalytic coupling to obtain intermediate F-1b-1, then corresponding sodium sulfide compound F-1c is obtained under alkaline condition, and then F-1 is obtained under acidic condition; or a halogenated compound F-1a and sodium tert-butylsulfite are substituted to obtain an intermediate F-1b-2, and then F-1 is obtained under an acidic condition.
The reaction equation for the preparation of F-1 is as follows:
Figure BDA0001712865010000221
wherein, W3Is halogen, preferably Br, I; v, R1aIs as defined above.
The inert solvent involved in the present invention is selected from: dichloromethane, chloroform, 1, 2-dichloroethane, dioxane, DMF, acetonitrile, DMSO, NMP, THF, or a combination 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.
The invention also provides a pharmaceutical composition, which comprises the pyrimido-cyclic compound shown in the formula (I), pharmaceutically acceptable salts, hydrates, prodrugs, stereoisomers or solvates thereof or an isotopically labeled compound of the pyrimido-cyclic compound shown in the formula (I), pharmaceutically acceptable salts, hydrates, prodrugs, stereoisomers or solvates thereof, and pharmaceutically acceptable auxiliary materials. The pharmaceutically acceptable excipients are preferably selected from diluents, absorbents, wetting agents, binders, disintegrants, lubricants.
The invention also provides application of the pyrimido-cyclic compound shown in the formula (I), a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer or a solvate thereof, or an isotopically labeled compound of the pyrimido-cyclic compound shown in the formula (I), a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer or a solvate thereof in preparing a medicament for treating diseases or symptoms related to abnormal activity of SHP 2. 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.
The invention also provides a pharmaceutical preparation, which comprises the pyrimido-cyclic compound shown in the formula (I), pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof, or an isotopically labeled compound of the pyrimido-cyclic compound shown in the formula (I), pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof. Can be administered in a suitable manner, such as in the form of tablets, capsules (e.g., sustained release or timed release capsules), pills, powders, granules (e.g., small granules), elixirs, tinctures, suspensions (e.g., nanosuspensions, microsuspensions), and spray-dried dispersions, syrups, emulsions, solutions, and the like, and can be administered orally, sublingually, by injection including subcutaneous injection, intravenous injection, intramuscular injection, intrasternal injection, infusion, and the like, by nasal administration (e.g., nasal inhalation), by topical surfaces (e.g., creams and ointments), by rectal administration (e.g., suppositories), and the like. The compounds disclosed herein may be administered alone or in combination with a suitable pharmaceutical carrier.
The invention also provides that the pharmaceutical preparation according to the previous aspect can be formulated into an appropriate dosage of the drug to facilitate and control the dosage of the drug. 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).
The invention also provides a product of the pyrimido-cyclic compound shown in the formula (I), pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof, or an isotopically labeled compound of the pyrimido-cyclic compound shown in the formula (I), pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof, which is used in combination with other drugs, wherein the other drugs are selected from: anticancer drugs, tumor immunity drugs, antiallergic drugs, antiemetics, analgesics, cytoprotective drugs, etc., which have a better effect when used in combination.
The present invention also provides a method for using the pyrimido-cyclic compound represented by formula (I), a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, or an isotopically labeled compound of the pyrimido-cyclic compound represented by formula (I), a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, in combination with another drug selected from the group consisting of: anticancer drugs, tumor immunity drugs, antiallergic drugs, antiemetics, analgesics, cytoprotective drugs, etc., which have a better effect when used in combination.
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 pyrimido-cyclic compound 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 region inhibitor, shows good biological activity and druggability, and has good drug development prospect.
2. In the same evaluation system of SHP2 enzyme activity inhibition experiment, phosphoprotein kinase (p-ERK) cell experiment, MOLM-13 cell proliferation experiment and the like, the invention shows more excellent activity compared with the compound SHP099(6- (4-amino-4-methylpiperidin-1-yl) -3- (2, 3-dichlorophenyl) pyrazine-2-amine) disclosed in WO 2015/107493A1 and literature (Nature 2016,535, 148-.
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 ….
Radical definition
Definitions for the terms of the standardization sector can be found in the literature references including Carey and 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, it is used herein in the analytical chemistry, organic synthetic chemistry, and the related description of pharmaceutical and pharmaceutical chemistryAre 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, -CH2O-is equivalent to OCH2-。
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. C1-6Alkyl refers to an alkyl group as defined below having a total of 1,2,3,4, 5, or 6 carbon atoms. The total number of carbon atoms in the shorthand notation excludes carbons that may be present in a substituent of the group.
Herein, numerical ranges defined in substituents such as 0 to 4, 1-4, 1 to 3, 1-6, etc., indicate integers within the range such as 0, 1,2,3,4, 5, 6.
In addition to the foregoing, the following terms, when used in the specification and claims of this application, have 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" refers to a-C (═ O) -group. "cyano" means-CN.
"amino" refers to-NH 2.
"substituted amino" refers to an amino group substituted with one or two alkyl, alkylcarbonyl, aralkyl, heteroaralkyl groups as defined below, e.g., 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 3 to 15 carbon atoms, preferably having 3 to 10 carbon atoms, more preferably having 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, 7-methylene-1H-indenyl, octahydro-2, 5-methylene-pentalenyl 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 2-14 carbon atoms and 1-6 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 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 1-3 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 4-to 8-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1-3 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, anthracenyl, phenanthrenyl, fluorenyl, 2, 3-dihydro-1H isoindolyl, 2-benzoxazolinone, 2H-1, 4-benzoxazin-3 (4H) -one-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 1-15 carbon atoms (preferably 1-10 carbon atoms) and 1-6 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. Examples of heteroaryl groups include, but are not limited to, thienyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, benzopyrazolyl, indolyl, furyl, pyrrolyl, triazolyl, tetrazolyl, triazinyl, indolizinyl, isoindolyl, indazolyl, isoindolyl, purinyl, quinolyl, isoquinolyl, diazonaphthyl, naphthyridinyl, quinoxalinyl, pteridinyl, carbazolyl, carbolinyl, phenanthridinyl, phenanthrolinyl, acridinyl, phenazinyl, isothiazolyl, benzothiazolyl, benzothienyl, oxazolyl, cinnolinyl, quinazolinyl, thiophenyl, indolizinyl, orthophenanthrolidinyl, isoxazolyl, phenoxazinyl, phenothiazinyl, 4,5,6, 7-tetrahydrobenzo [ b ] thienyl, naphthopyridyl, pyridinyl, and the like, [1,2,4] triazolo [4,3-b ] pyridazine, [1,2,4] triazolo [4,3-a ] pyrazine, [1,2,4] triazolo [4,3-c ] pyrimidine, [1,2,4] triazolo [4,3-a ] pyridine, imidazo [1,2-b ] pyridazine, imidazo [1,2-a ] pyrazine and the like.
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.
Conventional techniques for preparing/isolating individual isomers include Chiral synthesis from suitable optically pure precursors, or resolution of racemates (or racemates of salts or derivatives) using, for example, Chiral high performance liquid chromatography, see, for example, GeraldG ü bitz and Martin G.Schmid (Eds.), Chiral Separations, Methods and Protocols, Methods in Molecular Biology, Vol.243, 2004; A.M.Stalcup, Chiral Separations, Annu.Rev.Anal.Chem.3: 341. 63, 2010; Fumis et al (Eds.), GEL' S CYENCLOP EDIA PRACTICA CHEMICAL TRY 5. CHEMISTH., TechtH., Longman Scientific and chemical Ltd., EsX, 1991, 809. 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 dichloroacetates, trifluoroacetates, propionates, caproates, caprylates, caprates, undecylenates, glycolates, gluconates, lactates, sebacates, adipates, glutarates, malonates, oxalates, maleates, succinates, fumarates, tartrates, citrates, palmitates, stearates, oleates, cinnamates, laurates, malates, glutamates, pyroglutamate, 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 regulatory agency for human or livestock use.
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.
As used herein, the term "treatment" and other similar synonyms include the following meanings:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease or condition, but has not 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., resolving 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 that is sufficient to alleviate one or more symptoms of the disease or disorder being treated to some extent after 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 remission effect of the condition. 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 capable of delivering a compound or composition to a desired site for biological action. 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 pharmacological 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 treatment", "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 hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g.tert-butyldimethylsilyl, tert-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 and 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 apparatus: 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% HCOOH); b-acetonitrile NMR
The instrument comprises the following steps: bruker Ascend 400M (1H NMR:400 MHz; 13C NMR:100 MHz).
Example 1: preparation of intermediate 5-chloro-8-iodo- [1,2,4] triazolo [4,3-c ] pyrimidine (E-1)
The method comprises the following steps: 2-chloro-4-hydrazino-5-iodopyrimidine
Figure BDA0001712865010000361
To a dry 100mL flask were added 2, 4-dichloro-5-iodopyrimidine (25g,91mmol) and absolute ethanol (20 mL). Hydrazine hydrate (13.66g,272.9mmol) was added slowly at 0 ℃. The mixture was stirred at room temperature for 2 hours, a large amount of solid appeared, filtered, then washed with ethanol, and dried under vacuum to give 2-chloro-4-hydrazino-5-iodopyrimidine (20g, yield: 81%) as a brown solid.
1H NMR(400MHz,CDCl3)δ8.29(s,1H),6.67(s,1H),4.08(s,2H)ppm;LC-MS:m/z 271.1[M+H]+
Step two: 5-chloro-8-iodo- [1,2,4] triazolo [4,3-c ] pyrimidine
Figure BDA0001712865010000362
To a dry 50mL stopcock were added 2-chloro-4-hydrazino-5-iodopyrimidine (10g,37mmol) and trimethyl orthoformate (3.92g,37mmol) in that order. The mixture was heated to 80 ℃ under nitrogen and the reaction stirred for 5 hours. After the reaction was completed, the reaction solution was cooled to room temperature, the mixture was slowly poured into a saturated sodium bicarbonate solution, and extracted with ethyl acetate (3 × 100mL), the organic layers were mixed and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give a product, which was then chromatographed using a silica gel column (ethyl acetate: petroleum ether ═ 1: 1) to give 5-chloro-8-iodoimidazo [1,2-c ] pyrimidine (5.7g, yield 55%).
1H NMR(400MHz,DMSO-d6)δ9.69(s,1H),8.28(s,1H)ppm;LC-MS:m/z 280.1[M+H]+
Example 2: 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 BDA0001712865010000371
1-oxo-8-azaspiro [4.5] was added to a dry 100mL single-necked flask in sequence]Tert-butyl decane-8-carboxylate (2.53g,10mmol), titanium tetraethoxide (6.84g,30mmol) and 50mL of tetrahydrofuran were stirred under reflux with heating for 4 hours. After cooling to room temperature, methanol (10mL) was added followed by lithium borohydride (0.65g,30 mmol). 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 combined phases were MgSO4Dry, 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]Tert-butyl decane-8-carboxylate (2.86g, 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 BDA0001712865010000372
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 (50mL) was stirred at room temperature for 2 hours. Adding Na2CO3The aqueous solution was saturated until pH 11 and the aqueous mixture was extracted with DCM (3 × 50 mL). The combined organic phases were washed with brine, washed with Na2SO4Drying, 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 (1.86g, yield: 90%)
1H NMR(400MHz,DMSO-d6)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 2, using similar starting materials, the following intermediate C-1B, C-1C, C-1D, C-1E, C-1F, C-1G, C-1H was obtained
Figure BDA0001712865010000381
Example 3: preparation of intermediate (R) -2-methyl-N- ((S) -2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (C-1I)
The method comprises the following steps: 4- (2- (benzyloxy) -1-hydroxyethyl) piperidine-1, 4-dicarboxylic acid 1-tert-butyl-4-methyl ester
Figure BDA0001712865010000401
To a dry 500mL three-necked flask, 1-tert-butyl-4-methylpiperidine-1, 4-dicarboxylate (45g,180mmol) and tetrahydrofuran (400mL) were added successively under nitrogen, and the solution was cooled to-78 deg.C, 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 (50mL) 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 NH4The reaction was quenched with Cl solution (200 mL). It was extracted with ethyl acetate (3 × 200 mL). The combined organic phases are washed with Na2SO4Drying, filtration, concentration of the filtrate under reduced pressure and purification of the resulting residue by silica gel chromatography (0 to 50% gradient of ethyl acetate/petroleum ether) gave 4- (2- (benzyloxy) -1-hydroxyethyl) piperidine-1, 4-dicarboxylic acid 1-tert-butyl-4-methyl ester (52g, yield: 73.3%).
1H NMR(400MHz,CDCl3)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 BDA0001712865010000402
To a dry 500mL three-necked flask was 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 (500mL) in this order, and then LiBH was added to the solution4(11.44g,520mmol)And stirred at room temperature for 6 hours. After the reaction was complete, saturated NaHCO was used3The reaction was quenched (200 mL). Extract with ethyl acetate (3 × 200 mL). The combined organic phases were washed with Na2SO4Drying, filtration, concentration of the filtrate under reduced pressure and purification of the resulting residue by silica gel chromatography (0 to 50% gradient of ethyl acetate/petroleum ether) gave 4- (2- (benzyloxy) -1-hydroxyethyl) -4- (hydroxymethyl) piperidine-1-carboxylic acid tert-butyl ester (27g, 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 BDA0001712865010000411
To a dry 500mL one-necked flask were added tert-butyl 4- (2- (benzyloxy) -1-hydroxyethyl) -4- (hydroxymethyl) piperidine-1-carboxylate (27g,74mmol), methanol (270mL) and Pd/C (20g) in this 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 (18.9g, 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
To a dry 500mL single-neck flask were added tert-butyl 4- (1, 2-dihydroxyethyl) -4- (hydroxymethyl) piperidine-1-carboxylate (18.9g, 69mmol), triphenylphosphine (25.2g,86.25mmol) and tetrahydrofuran (350mL) in that order, the reaction was cooled to 0 deg.C and DEAD (12.46mL,86mmol) was added, then warmed to room temperature and stirred for 5 hours. After completion of the reaction, water (200mL) was added to quench the reaction. It was extracted with ethyl acetate (3 × 200 mL). The organic phases were combined and washed with Na2SO4Drying, 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-hydroxyRadical-2-oxa-8-azaspiro [4.5]Tert-butyl decane-8-carboxylate (13.2g, yield: 74%).
1H NMR(400MHz,CDCl3)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-oxo-2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0001712865010000421
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 (280mL) and Dess-Martin oxidant (32.2g,76.5mmol) were stirred for 5h in an ice bath. Adding NaHCO after the reaction is finished3:Na2S2O3(1: 1) saturated solution (200mL), the organic phase was separated and the aqueous phase was extracted with DCM (3X 100 mL). The combined organic phases are washed with Na2SO4Drying, 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-oxo-2-oxa-8-azaspiro [4.5] as a colourless solid]Tert-butyl decane-8-carboxylate (12g, yield: 92.1%).
1H NMR(400MHz,CDCl3)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/z 200.0[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 BDA0001712865010000422
Following the synthesis of intermediate C-1A, step one of example 2, tert-butyl 4-oxo-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.
1H NMR(400MHz,CDCl3)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-1I)
Figure BDA0001712865010000431
Following the synthesis of intermediate C-1A, step two, in example 2, (S) -4- ((R) -1-methylethylsulfonamido) -2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester removed the Boc protecting group to give (R) -2-methyl-N- ((S) -2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfonamid-1I as a white solid.
1H NMR(400MHz,DMSO-d6)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 4: 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 BDA0001712865010000441
To a solution of ethyl (S) -2-hydroxypropionate (30g, 254mmol) in dichloromethane (300mL) was added imidazole (2.75g, 304.9mmol) and cooled to 0 ℃. To the solution was added tert-butyldimethylsilyl chloride (46.0g, 304.9mmol) 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 (50g, 84% yield) as a colorless liquid.
1H NMR(400MHz,CDCl3)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 BDA0001712865010000442
To a solution of ethyl (S) -2- ((tert-butyldimethylsilyl) oxy) propionate (25g, 107.6mmol) in diethyl ether (500mL) at-78 deg.C was slowly added dropwise diisobutylaluminum hydride (1M in hexane) (129mL, 129.1mmol) and stirred at-78 deg.C for 1 hour. After completion of the reaction was confirmed by TLC analysis, the temperature of the reaction mixture was slowly raised to-40 ℃ and the reaction was quenched with saturated aqueous solution of Rochelle salt (1L) and then diethyl ether (500mL) 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 (250mL) and Na2SO4Drying, filtration and concentration under reduced pressure gave (S) -2- ((tert-butyldimethylsilyl) oxy) propanal (19g, yield: 94%).
1H NMR(400MHz,CDCl3)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 BDA0001712865010000451
To a stirred mixture of 1- (tert-butyl) -4-ethylpiperidine-1 at 0 ℃,lithium diisopropylamide (2M in THF) (93.3mL, 186.6mmol) was added to a solution of 4-dicarboxylate (30g, 116.6mmol) in THF (250mL) and stirring continued at 0 deg.C for 30 min. Then a solution of (S) -2- ((tert-butyldimethylsilyl) oxy) propanal (22g, 116.6mmol) in THF (50mL) was added. The resulting reaction mixture was stirred at 0 ℃ for 1 hour and then kept at room temperature for 1 hour. After completion of the reaction as judged by TLC analysis, the reaction mixture was taken up with saturated NH4The Cl solution was quenched and extracted with ethyl acetate (2X 250 mL). The combined organic layers were washed with water (150mL), brine (150mL) 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) (17g, yield: 32%) as a pale red oil.
1H NMR(400MHz,CDCl3)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 BDA0001712865010000452
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 (50mL) and LiBH was added portionwise4(0.73g, 33.65mmol) and stirred at room temperature for 16 h. After the reaction was complete, the reaction mixture was quenched with saturated NaHCO at 0 deg.C3The 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. Filtering and concentrating under reduced pressureThe crude product 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 (3g, yield: 66%).
1H NMR(400MHz,CDCl3)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 BDA0001712865010000461
To a solution of tert-butyl ((2S) -2- ((tert-butyldimethylsilyl) oxy) -1-hydroxypropyl) -4- (hydroxymethyl) piperidine-1-carboxylate (25g, 61.93mmol) in THF (500mL) 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 NaHCO3The 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 filtered and concentrated under reduced pressure and 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 (12g, yield: 67%) as a colorless liquid.
1H NMR(400MHz,DMSO-d6)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 BDA0001712865010000471
To a stirred suspension of NaH (60% in mineral oil) (1.45g, 60.5mmol) in THF (30mL) at 0 deg.C was added a solution of tert-butyl 4- ((2S) -1, 2-dihydroxypropyl) -4- (hydroxymethyl) piperidine-1-carboxylate (5g,17.3mmol) and p-toluenesulfonyl chloride (3.29g, 17.3mmol) in THF (20mL), and the resulting reaction mixture was reacted at 0 deg.C for 3 hours. After completion of the reaction, the reaction mixture was saturated with NH at-20 deg.C4The Cl solution (250mL) 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 was 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 (2.1g, yield: 44%).
1H NMR(400MHz,CDCl3)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/z172.2[M-100]+
Step seven: (S) -tert-butyl-3-methyl-4-carbonyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylic acid ester
Figure BDA0001712865010000472
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 (50mL), followed by Dess-Martin oxidant (4.26g,10.06mmol)) 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 (1.2g, yield: 57%).
1H NMR(400MHz,CDCl3)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 BDA0001712865010000481
(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 (15mL) 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 (2mL) was added followed by the addition of LiBH in portions4(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 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%]Tert-butyl decane-8-carboxylate (1.2g, yield: 72%).
1H NMR(400MHz,CDCl3)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)
To a solution of tert-butyl (3S,4S) -4- ((R) -tert-butylsulfinyl) amino) -3-methyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylate (1.1g, 2.936mmol) in dichloromethane (10mL) 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 (850mg, yield: 72%).
1H NMR(400MHz,DMSO-d6)δ8.40(brs,D2O Exchangeable,1H),8.30(brs,D2O 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 5: preparation of (R) -2-methyl-N- ((3R,4S) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide and (R) -2-methyl-N- ((3R,4R) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (C-1K)
Step one (R) -tert-butyl 3-methyl-4-carbonyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylate
Figure BDA0001712865010000501
(R) -tert-butyl 3-methyl-4-carbonyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylate can be obtained from (R) -2- ((tert-butyldimethylsilyl) oxo) propanal by the method of example 4 above
Step two (3R,4S) -tert-butyl 4- ((R) -1, 1-dimethylethylsulfonamido) -3-methyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylate with (3R,4R) -tert-butyl 4- ((R) -1, 1-dimethylethylsulfonamido) -3-methyl-2-oxa-8-azaspiro [4.5] decane-8-carboxylate
Figure BDA0001712865010000502
(R) -tert-butyl-3-methyl-4-carbonyl-2-oxa-8-azaspiro [4.5]A stirred solution of decane-8-carboxylic acid ester (0.97g, 3.71mmol) in THF (5mL) was charged with (R) -2-methylpropane-2-sulfinamide (0.873g, 7.2mmol) and tetraethyltitanate (3.36mL, 14.74mmol), respectively. The resulting reaction mixture was stirred at 90 ℃ for 18 hours. The reaction mixture was cooled to-4 ℃ and MeOH (5mL) was added followed by the addition of LiBH in portions4(80mg, 3.71mmol) 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 is filtered and concentrated under reduced pressure to purify it by flash chromatography using 0.1% formic acid and acetonitrile as eluent to obtain (3R,4S) -tert-butyl 4- ((R) -1, 1-dimethylethylsulfonamido) -3-methyl-2-oxa-8-azaspiro [ 4.5%]Decane-8-carboxylic acid ester with (3R,4R) -tert-butyl 4- ((R) -1, 1-dimethylethylsulfonamido) -3-methyl-2-oxa-8-azaspiro [ 4.5%]Decane-8-carboxylic ester (0.5g, yield: 36%).
Step three (R) -2-methyl-N- ((3R,4S) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide and (R) -2-methyl-N- ((3R,4R) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (C-1K)
Figure BDA0001712865010000511
(R) -2-methyl-N- ((3R,4S) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide mixture and mixture with (R) -2-methyl-N- ((3R,4R) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide can be obtained according to the procedure of example 4 step nine the crude product is then purified by flash chromatography using 0.1% formic acid and acetonitrile as eluent to give (R) -2-methyl-N- ((3R,4S) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane Alkane-2-sulfinamide (119mg, 24% yield) and (R) -2-methyl-N- ((3R,4R) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (154mg, yield: 31%)
(R) -2-methyl-N- ((3R,4S) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide:
1H-NMR(400MHz,DMSO-d6):δ5.45(d,D2O Exchangeable,J=11.0Hz,1H),3.78(d,J=9.0Hz,1H),3.64-3.59(m,2H),3.27-3.25(m,1H),3.17-3.14(m,1H),2.88-2.76(m,3H),1.90-1.85(m,1H),1.82-1.76(m,1H),1.59-1.51(m,2H),1.18-1.17(m,12H)ppm;LC-MS:m/z275.2[M+H]+.
(R) -2-methyl-N- ((3R,4R) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide:
1H-NMR(400MHz,DMSO-d6):δ5.04(d,D2O Exchangeable,J=10.5Hz,1H),4.45-4.11(m,1H),3.48(d,J=8.5Hz,1H),3.50-3.46(m,1H),3.43(d,J=9.0Hz,1H),3.14-3.12(m,1H),3.04-3.02(m,1H),3.91-3.87(m,2H),1.73-1.68(m,2H),1.62-1.56(m,2H),1.17(s,9H),1.14(d,J=6.5Hz,3H)ppm;LC-MS:m/z 275.2[M+H]+.
example 6: synthesis of 2-methyl-N- ((S) -4-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (C-1L)
Step one (R, Z) -N- (8-benzoyl-2-oxa-8-azaspiro [4.5] decan-4-ylidene) -2-methylpropane-2-sulfinamide
(R, Z) -N- (8-benzoyl-2-oxa-8-azaspiro [4.5] decan-4-ylidene) -2-methylpropan-2-sulfinamide is obtained according to the procedure of example 3
LC-MS:m/z 363.2[M+H]+.
Step two N- ((S) -8-benzoyl-4-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) -2-methylpropane-2-sulfinamide
Figure BDA0001712865010000522
(R, Z) -N- (8-benzoyl-2-oxa-8-azaspiro [4.5] decan-4-ylidene) -2-methylpropane-2-sulfinamide (200mg,0.552mmol) was dissolved in toluene (5ml), the reaction system was cooled to 0 ℃ and a methylmagnesium bromide solution (1.1ml,3.32mmol) was slowly added dropwise, after which the reaction was stirred at room temperature for 2.5 hours. After the reaction was quenched with saturated ammonium chloride, extracted with ethyl acetate, dried over anhydrous sodium sulfate, and spin-dried under reduced pressure, the crude product was separated with a preparative plate (20% petroleum ether solution of ethyl acetate) to give N- ((S) -8-benzoyl-4-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) -2-methylpropane-2-sulfinamide (20mg, yield: 10%).
LC-MS:m/z 379.2[M+H]+.
Step three 2-methyl-N- ((S) -4-methyl-2-oxa-8-azaspiro [4.5] decan-4-yl) propane-2-sulfinamide (C-1L)
Figure BDA0001712865010000531
Reacting N- ((S) -8-benzoyl-4-methyl-2-oxa-8-azaspiro [4.5]Decan-4-yl) -2-methylpropane-2-sulfinamide (100mg,0.26mmol) was dissolved in tetrahydrofuran (5ml), and the reaction system was cooled to 0 ℃ followed by slow addition of 4N NaOH (0.65ml,2.6mmol), after which the reaction was stirred at room temperature for 2.5 hours. The reaction was extracted with ethyl acetate (5 × 10mL) and the combined organic phases were extracted with MgSO 44Drying, filtering, and concentrating the filtrate under reduced pressure to obtain 2-methyl-N- ((S) -4-methyl-2-oxa-8-azaspiro [ 4.5%]Decan-4-yl) propane-2-sulfinamide (50mg, yield: 70%) was used directly in the next reaction.
LC-MS:m/z 275.2[M+H]+.
Example 7: (R) -N, 2-dimethyl-N- ((R) -8-azaspiro [4.5] decan-1-yl) propane-2-sulfinamide (C-1M)
The method comprises the following steps: (R) -tert-butyl-1- ((R) -N, 2-dimethylpropan-2-ylsulfonamido) -8-azaspiro [4.5] decane-8-carboxylic acid ester
Figure BDA0001712865010000532
Reacting (R) -tert-butyl-1- ((R) -1, 1-dimethylethylsulfonamido with) -8-azaspiro [4.5]Decane-8-carboxylate (500mg,1.39mmol) was dissolved in tetrahydrofuran (10ml), and the reaction system was cooled to 0 ℃ followed by slow addition of NaH (54mg,2.23mmol), after which iodomethane (396mg, 2.79mmol) was added and the reaction stirred at room temperature overnight. The reaction was quenched with water and then extracted with ethyl acetate (3 × 10mL) and the combined organic phases were MgSO4Drying, filtering, concentrating the filtrate under reduced pressure to obtain crude product, separating the crude product with silica gel column (50% ethyl acetate in petroleum ether) to obtain (R) -tert-butyl-1- ((R) -N, 2-dimethylpropane-2-ylsulfonamido) -8-azaspiro [ 4.5%]Decane-8-carboxylic acid ester (400mg, yield: 80%).
LC-MS:m/z 373.2[M+H]+.
Step two: (R) -N, 2-dimethyl-N- ((R) -8-azaspiro [4.5] decan-1-yl) propane-2-sulfinamide (C-1M)
Figure BDA0001712865010000541
(R) -N, 2-dimethyl-N- ((R) -8-azaspiro [4.5] decan-1-yl) propane-2-sulfinamide can be obtained according to the procedure of step 9 of example 4.
LC-MS:m/z 273.2[M+H]+.
Example 8: 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
To a dry 1L flask was added tert-butyl 4-formylpiperidine-1-carboxylate (35.0g,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 h. After the reaction is completed, the mixture is poured into a reactor containing saturated NH4Aqueous Cl (50%, 500mL) in a separatory funnel using Et2O (5X 50 mL). The combined organic phases were washed with MgSO4Drying, filtering, and concentrating the filtrate under reduced pressure. Will be provided withThe residue obtained is purified by chromatography on silica gel (0 to 25% gradient of ethyl acetate/petroleum ether) to give tert-butyl 4-allyl-4-formylpiperidine-1-carboxylate (24g, yield: 48%) as a colourless oil.
1H NMR(400MHz,CDCl3)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 BDA0001712865010000551
To a 1L dry three-necked flask was added tert-butyl 4-allyl-4-formylpiperidine-1-carboxylate (24g,95mmol) and THF (300mL) 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 warm slowly to room temperature over 1 hour. After the reaction is completed, the mixture is poured into a reactor containing saturated NH4Aqueous Cl (250mL) was separated onto a separatory funnel and extracted with EtOAc (4 × 50 mL). The combined organic phases were washed with MgSO4Drying, filtration and concentration of the filtrate under reduced pressure gave tert-butyl 4-allyl-4- (1-hydroxyallyl) piperidine-1-carboxylate (26.7g), which was used in the next step without further purification.
1H NMR(400MHz,CDCl3)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
Adding 4-allyl-4- (1-hydroxy allyl) into a dry three-neck flask in sequence) Piperidine-1-carboxylic acid tert-butyl ester (26.7g,95mmol), Dess-Martin oxidant (44.3g,105mmol) and anhydrous dichloromethane (380mL), and the mixture was stirred at room temperature for 1 hour. After the reaction is finished, the mixture is poured into a reactor containing NaHCO3:Na2SO3Saturated aqueous (1:1,300mL) in a separatory funnel and then extracted with DCM (4X 50 mL). The combined organic phases were washed with MgSO4Drying, filtering, and concentrating the filtrate under reduced pressure to obtain white solid. The white solid was suspended in petroleum ether (250mL) and sonicated for 20 min. The white suspension was filtered through a celite pad and removed under reduced pressure, and the filtrate was concentrated under reduced pressure to give 4-levulinyl-4-allylpiperidine-1-carboxylic acid tert-butyl ester as yellow oil (25g, two-step yield: 94%).
1H NMR(400MHz,CDCl3)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-oxo-8-azaspiro [4.5] decan-2-ene-8-carboxylic acid tert-butyl ester
Figure BDA0001712865010000561
To a 1L dry three-necked flask was added sequentially tert-butyl 4-levulinyl-4-allylpiperidine-1-carboxylate (25g,89.6mmol), toluene (degassed, 850mL), and a solution of Grubbs II 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 (0-40% gradient of ethyl acetate/petroleum ether) to give tert-butyl 1-oxo-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 (540mL) was stirred at room temperature for 15 minutes. The resulting bright red solution was filtered through a pad of celite. Activated charcoal (100g) 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 (0-40% gradient of ethyl acetate/petroleum ether) to give tert-butyl 1-oxo-8-azaspiro [4.5] dec-2-ene-8-carboxylate (12g, yield: 53.3%) as a white solid.
Step five: 3-methyl-1-oxo-8-azaspiro [4.5] decane-8-carboxylic acid tert-butyl ester
Figure BDA0001712865010000571
CuI (3.8g,20mmol) and anhydrous tetrahydrofuran (100mL) were added sequentially to a nitrogen blanketed 250mL dry three-necked flask, the solution was cooled to-20 ℃ and MeLi (1.6M in THF, 25mL,40mmol) was slowly added dropwise to the solution, after which the reaction was allowed to react at-20 ℃ until the solution was clear. Then slowly dropwise adding 1-oxo-8-azaspiro [4.5] at the temperature]A solution of tert-butyl decan-2-ene-8-carboxylate (2.51g,10mmol) in tetrahydrofuran (20 mL). After the reaction is finished, the mixture is poured into a reactor containing saturated NH4Aqueous Cl was extracted with ethyl acetate (3 × 15mL) in a separatory funnel. The combined organic phases were washed with MgSO4Drying, filtering and concentrating the filtrate under reduced pressure and purification by silica gel chromatography (0 to 50% gradient of ethyl acetate/petroleum ether) to give 3-methyl-1-oxo-8-azaspiro [4.5]]Tert-butyl dec-2-ene-8-carboxylate (1.6g, yield: 60%).
1H NMR(400MHz,CDCl3)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 BDA0001712865010000572
According to the synthesis method of step eight and nine of the synthesis intermediate C-1J, the ketone intermediate 3-methyl-1-oxo-8-azaspiro [4.5] decane-2-ene-8-tert-butyl formate is subjected to reductive amination and Boc protecting group removal to obtain (R) -2-methyl-N- ((1R) -3-methyl-8-aza-spiro [4.5] decane-1-yl) propane-2-sulfinamide C-1N.
1H NMR(400MHz,CDCl31H NMR(400MHz,DMSO-d6)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 9: preparation of intermediate tert-butyl ((4-methylpiperidin-4-yl) methyl) carbamate (C-4A)
The method comprises the following steps: 1-benzoyl-4-methylpiperidine-4-carbonitrile
Figure BDA0001712865010000581
To a 100mL dry single-neck flask, under nitrogen, was added 4-methylpiperidine-4-carbonitrile (496mg,4mmol), DCM (10mL) and triethylamine (611mg,6mmol) in that order, followed by slow dropwise 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 20mL) was extracted and the combined organic phases were Na2SO4Drying, 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 (650mg, 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 BDA0001712865010000582
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 methanol (12mL) were added in that order under nitrogen at 0 deg.C, and sodium borohydride (754mg,20mmol) was added. The reaction was then stirred at room temperature for 12 hours and monitored by TLCThe reaction of the raw materials is finished. After completion of the reaction, the reaction was concentrated and extracted with dichloromethane (3X 20mL), and the combined organic phases were extracted with Na2SO4Drying, 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 (620mg, 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 BDA0001712865010000591
To a 100mL single-neck flask were added tert-butyl ((1-benzoyl-4-methylpiperidin-4-yl) methyl) carbamate (620mg,1.87mmol), ethanol (8mL) and 7N NaOH (2mL) in this order, the mixture was heated to 90 ℃ under nitrogen and stirred for 8 hours, the mixture was cooled to room temperature, filtered, diluted with water and extracted with ethyl acetate (3X 20mL), and the combined organic phases were Na-impregnated2SO4Drying, 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 (300mg, yield: 70.5%).
1H NMR(400MHz,DMSO-d6)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 10 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
To a solution of tert-butyl (2-chloroethyl) carbamate (2g,8.26mmol) and 2-phenylacetonitrile (968mg,8.26mmol) in anhydrous DMF (20mL) 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 (30mL) and then extracted with 3X50 mL). The combined organic layers were washed with saturated brine (2 × 50mL), 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 (500mg, yield: 21%).
LCMS:m/z 187.2[M-100]+.
Step two: 4- (aminomethyl) -4-phenylpiperidine-1-carboxylic acid tert-butyl ester
Figure BDA0001712865010000602
Tert-butyl 4-cyano-4-phenylpiperidine-1-carboxylate (0.5g,1.75mmol) was dissolved in 20mL of methanol, palladium on carbon (50mg) 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 (0.4g, yield: 80%).
1H NMR(400MHz,CDCl3)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)
Tert-butyl 4- (aminomethyl) -4-phenylpiperidine-1-carboxylate (0.4g,1.37mmol) was dissolved in 10mL of methanol, and 1, 4-dioxane hydrochloride (4M,13.7mmol) was added thereto at room temperature, and the reaction mixture was allowed to react at room temperature for 2 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give (4-phenylpiperidin-4-yl) methylamine C-4B (0.25g, yield: 95%) and the crude product was used directly in the next reaction.
LC-MS:m/z 191.2[M+H]+.
EXAMPLE 11 preparation of intermediate sodium 6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-thiolate (F-1A)
The method comprises the following steps: (6-Chloropyridin-2-yl) carbamic acid tert-butyl ester
Figure BDA0001712865010000611
6-chloropyridin-2-amine (8g,62.2mmol) and THF (80mL) were added under nitrogen to a dry 250mL three-necked flask, the mixture stirred at 0 ℃ for 10 minutes, then NaHDMS (124.4mL,1.0M in THF) was added, then a solution of di-tert-butyl dicarbonate (16.3g,74.7mmol) in tetrahydrofuran (50mL) was slowly added maintaining the system at 0 ℃ and the reaction continued at 0 ℃ for 4 hours. After the reaction is finished, H is added2O (40mL) and then extracted with EtOAc (3 × 100 mL). The combined organic phases were washed with MgSO4The residue obtained by drying, filtration and concentration under reduced pressure was purified by silica gel chromatography (0 to 10% gradient of ethyl acetate/petroleum ether) to give tert-butyl (6-chloropyridin-2-yl) carbamate (7g, yield: 49%).
1H 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
To a dry 100mL round bottom flask was added tert-butyl (6-chloropyridin-2-yl) carbamate (7g,30.6mmol) and N, N-dimethylformamide (50mL), the mixture was stirred at room temperature for 10 minutes, then N-chlorosuccinimide (4.50g,33.67mmol) was added and the mixture was reacted at 100 ℃ for 4 hours. After the reaction is finished, the temperature of the reaction solution is reduced to room temperature, and H is added2O (50mL) was then extracted with ethyl acetate (3x80mL) and washed with saturated aqueous lithium chloride (2x40 mL). The organic phase was washed with MgSO4Drying, filtering and concentrating under reduced pressureThe residue obtained is purified by chromatography on silica gel (gradient 0 to 5% ethyl acetate/petroleum ether) to give tert-butyl (5, 6-dichloropyridin-2-yl) carbamate (5.3g, yield: 65.8%).
1H NMR(400MHz,CDCl3)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 BDA0001712865010000621
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 (50mL), n-butyllithium (44.3mmol,2.5M inTHF) was added slowly dropwise at-78 deg.C and the reaction was stirred at this temperature for an additional 1 h. A solution of iodine (3.07g,24.17mmol) in tetrahydrofuran (20mL) was then added dropwise slowly and the reaction was continued at-78 deg.C for 3 hours. After the reaction is finished, H is added2O (50mL), followed by extraction with EtOAc (3 × 80 mL). The combined organic phases were washed with MgSO4Drying, 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-dichloro-4-iodopyridin-2-yl) carbamate (4.3g, yield: 55%).
1H 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: 3- ((6- ((tert-Butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) propionic acid methyl ester
Figure BDA0001712865010000631
To a dry 100mL round bottom flask was added tert-butyl (5, 6-dichloro-4-iodopyridin-2-yl) carbamate (3.2g,8.22mmol), palladium acetate (92mg, 0.41mmol), Xantphos (285mg,0.49mmol), DIPEA (2.12g,16.46mmol) and 1, 4-dioxane (30mL) in that order under nitrogen. 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 (0-30% gradient of ethyl acetate/petroleum ether) to give methyl 3- ((6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) propanoate (3g, yield: 96%).
1H 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: 6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridine-4-thiol (F-1A)
Figure BDA0001712865010000632
To a dry 100mL round bottom flask were added methyl 3- ((6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-yl) thio) propionate and tetrahydrofuran (30mL) in that order, and then a solution of sodium ethoxide in ethanol (21%, 6mL) was added slowly dropwise at room temperature, and the reaction was stirred at room temperature for 1 hour. Concentrated under reduced pressure, dichloromethane (10mL) was added, a large amount of brown solid precipitated, filtered, washed with dichloromethane and dried to give sodium 6- ((tert-butoxycarbonyl) amino) -2, 3-dichloropyridin-4-thiolate, which was acidified to pH 3 with 1N HCl, and the mixture was directly dried under reduced pressure to give crude F-1A (2.1g, ca.) which was used directly in the next reaction
1H NMR(400MHz,DMSO-d6)δ9.05(s,1H),7.61(s,1H),1.41(s,9H)ppm;LCMS:m/z 262.2[M-55]+.
Following the synthetic procedure of example 11, the following intermediate F-1B, F-1C, F-1D, F-1E, F-1F, F-1G, F-1H, F-1I, F-1J, F-1K was obtained by reaction with similar starting intermediates.
Figure BDA0001712865010000641
Figure BDA0001712865010000651
Figure BDA0001712865010000661
EXAMPLE 12 preparation of intermediate sodium 3-chloro-2-methylpyridin-4-thiolate (F-1L)
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 BDA0001712865010000662
To a dry 100mL round bottom flask under nitrogen was added methyl 3- ((2, 3-dichloropyridin-4-yl) thio) propionate (500mg,1.88mmol), Pd (PPh) in that order3)4(217mg,0.188mmol), trimethylcyclotriboroxane (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 (320mg, yield: 69%).
Step two: 3-chloro-2-methylpyridine-4-thiol sodium (F-1L)
To a dry 100mL round bottom flask were added methyl 3- ((3-chloro-2-methylpyridin-4-yl) thio) propionate (320mg,1.30mmol) and tetrahydrofuran (10mL) in that order, and then a solution of sodium ethoxide in ethanol (21%, 2mL) was slowly added dropwise at room temperature, and the reaction was stirred at room temperature for 1 hour. Concentrating under reduced pressure, adding dichloromethane (10mL), precipitating a large amount of brown solid, filtering, washing with dichloromethane, drying to obtain 3-chloro-2-methylpyridine-4-sodium mercaptide, acidifying sodium mercaptide with 1NHCl to pH 3, directly drying the mixture under reduced pressure to obtain crude F-1L (200mg) which is directly used for the next reaction
1H 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]+.
Example 13: preparation of intermediate sodium 6- ((tert-butoxycarbonyl) amino) -3-chloro-2-methylpyridin-4-thiolate (F-1M)
The method comprises the following steps: 3- ((6- ((tert-Butoxycarbonyl) amino) -3-chloro-2-methylpyridin-4-yl) thio) propionic 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.
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), trimethylcyclotriboroxane (301mg,2.4mmol), potassium carbonate (331mg,2.4mmol), 1, 4-dioxane (10mL) and water (1mL) in that order under nitrogen. 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 purify by silica gel chromatography (0 to 40% gradient of ethyl acetate/petroleum ether) to obtain methyl 3- ((6- ((tert-butoxycarbonyl) amino) -3-chloro-2-methylpyridin-4-yl) thio) propionate (420mg, yield: 74%).
1H 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-methylpyridin-4-thiolate (F-1M)
Figure BDA0001712865010000681
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 (10mL) in that order, followed by slow dropwise addition of a solution of sodium ethoxide in ethanol (21%, 2mL) at room temperature, and the reaction was stirred at room temperature for 1 hour. Concentrated under reduced pressure, then dichloromethane (10mL) was added, a large amount of brown solid precipitated, filtered, washed with dichloromethane and dried to give sodium 6- ((tert-butoxycarbonyl) amino) -3-chloro-2-methylpyridine-4-thiol which was then acidified to pH 3 with 1N HCl, the mixture was directly dried under reduced pressure and the crude F-1M (320mg) was used directly in the next reaction.
1H NMR(400MHz,DMSO-d6)δ9.87(s,1H),7.63(s,1H),3.64(s,3H),1.46(s,9H)ppm;LCMS:m/z 275.0.
EXAMPLE 14 preparation of intermediate 3-amino-2-chlorobenzenethiol hydrochloride (F-1N)
The method comprises the following steps: 2-chloro-3-aminothiophenol tert-butyl ester
Figure BDA0001712865010000682
To a dry 100mL round bottom flask was added 2-chloro-3-fluoroaniline (5g,34.3mmol) followed by N-methylpyrrolidinone (50mL) under nitrogen, then 2-methylpropane-2-thiol (8.66g, 96.04mmol) and cesium carbonate (22.36g,68.6mmol) and the reaction mixture was stirred at 120 ℃ for 16 h. After cooling to room temperature, the reaction solution was diluted with 60mL of ethyl acetate, washed with a saturated aqueous lithium chloride solution (30mL), water (30mL) and a saturated aqueous sodium chloride solution (30mL) in this order, then dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give tert-butyl 2-chloro-3-aminophenethiolate (6.04g, yield: 82%).
LCMS:m/z 216.1[M+H]+.
Step two: 3-amino-2-chlorobenzenethiol hydrochloride (F-1N)
To a dry 100mL round bottom flask were added tert-butyl 2-chloro-3-aminothiophenol (6.04g,28mmol) and concentrated hydrochloric acid (50mL), and the reaction mixture was stirred 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-1N (4.9g, yield: 90%).
LCMS:m/z 160.0[M+H]+.
EXAMPLE 15 preparation of the compound 1- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-amine
The method comprises the following steps: (1- (8-iodo- [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester
Figure BDA0001712865010000692
Under the protection of nitrogen, 5-chloro-8-iodo- [1,2,4] is added into a dry 50mL single-neck flask in sequence]Triazolo [4,3-c]Pyrimidine E1(50mg,0.18mmol), (4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (77mg,0.36mmol), DIEA (46mg,0.36mmol) and NMP (5mL), then the reaction was stirred at 90 ℃ for 2 hours. After completion of the reaction, the obtained residue was poured into water (10mL), and stirred at room temperature for 5 minutes. Then extracted with ethyl acetate (3 × 50mL) and the combined organic phases were extracted with MgSO4The residue obtained is dried, filtered and concentrated under reduced pressure, and purified by chromatography on silica gel (0 to 80% gradient of ethyl acetate/petroleum ether) to give a pale yellow solid (1- (8-iodo- [1,2, 4)]Triazolo [4,3-c]Pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (60mg, yield: 73%).
LCMS:m/z 459.1[M+H]+.
Step two: (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c ] pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester
Figure BDA0001712865010000701
A dry 50mL three-necked flask was charged with (1- (8-iodo- [1,2, 4)]Triazolo [4,3-c]Pyrimidin-5-yl) -4-methylpiperidin-4-yl) Tert-butyl carbamate (60mg,0.13mmol), cuprous iodide (3mg,0.013mmol), 1, 10-phenanthroline (5mg,0.026mmol), 2, 3-dichlorothiophenol (36mg,0.2mmol), potassium phosphate (60mg,0.26mmol), and 5mL dioxane. The mixture was heated under nitrogen for 3 hours. After the reaction is finished, saturated NH is added4Cl solution (10 mL). Then extracted with ethyl acetate (3 × 50 mL). The combined organic phases are washed with Na2SO4Drying, 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-yl) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (25mg, yield: 38%).
LC-MS:m/z 510.1[M+H]+.
Step three: 1- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-amine
Figure BDA0001712865010000711
To a dry 50mL round bottom flask was added (1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1, 2-c) in sequence]Pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester (25mg,0.049mmol) and a solution of hydrochloric acid in 1, 4-dioxane (7M,5mL) were reacted at room temperature for 1 hour. After the reaction is finished, saturated NaHCO is added3Solution (10 mL). Then extracted with ethyl acetate (3 × 50 mL). The combined organic phases are washed with Na2SO4Drying, filtering, concentrating the filtrate under reduced pressure, and purifying the obtained crude product by high performance liquid chromatography to obtain the product 1- (8- ((2, 3-dichlorophenyl) thio) imidazo [1,2-c]Pyrimidin-5-yl) -4-methylpiperidin-4-amine (10mg, yield: 49%).
1H NMR(400MHz,DMSO-d6)δ9.42(s,1H),8.00(s,1H),7.42(t,J=8.4Hz,1H),7.14(t,J=8.0Hz,1H),6.80(t,J=15.0Hz,1H),3.78(dd,J=35.6,5.8Hz,4H),1.75(s,4H),1.26(s,3H)ppm;LC-MS:m/z 410.1[M+H]+.
According to the synthesis method of example 15, the following compounds can be synthesized:
example 16 1- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) piperidin-4-amine
Figure BDA0001712865010000712
1H NMR(400MHz,DMSO-d6)δ9.45(s,1H),8.31(s,1H),8.02(s,1H),7.44(d,J=6.9Hz,1H),7.14(t,J=8.1Hz,1H),6.83(d,J=8.0Hz,1H),4.18(d,J=13.5Hz,2H),3.44(d,J=7.1Hz,1H),3.27(s,2H),1.99(d,J=10.9Hz,2H),1.68(d,J=9.9Hz,2H)ppm;LC-MS:m/z 395.0[M+H]+.
Example 17 (1- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-phenylpiperidin-4-yl) methylamine
Figure BDA0001712865010000721
1H NMR(400MHz,DMSO-d6)δ9.42(s,1H),7.99(s,1H),7.46(dt,J=22.1,7.5Hz,5H),7.33(t,J=6.8Hz,1H),7.13(t,J=8.0Hz,1H),6.81(d,J=8.0Hz,1H),3.97(d,J=14.4Hz,2H),3.45-3.37(m,2H),2.98(s,2H),2.38(d,J=14.8Hz,2H),2.06(t,J=10.4Hz,2H)ppm;LC-MS:m/z 485.1[M+H]+.
Example 18 (1- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) pyrrolidin-3-yl) methylamine
Figure BDA0001712865010000722
1H NMR(400MHz,DMSO-d6)δ9.49(s,1H),7.92(s,1H),7.86(brs,2H),8.42(d,J=8.0Hz,1H),7.14(t,J=8.0Hz,1H),6.74(d,J=8.0Hz,1H),4.16-3.93(m,4H),3.77-3.72(m,1H),3.01-2.97(m,1H),2.66-2.58(m,1H),2.25-2.18(m,2H),1.88-1.76(m,1H)ppm;LC-MS:m/z 395.1[M+H]+.
Example 19 (1- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-yl) methylamine
Figure BDA0001712865010000723
1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),7.98(s,1H),7.43(dd,J=8.0,1.3Hz,1H),7.14(t,J=8.0Hz,1H),6.82(dd,J=8.1,1.3Hz,1H),3.92-3.78(m,2H),3.64-3.55(m,2H),2.56(s,2H),1.67(ddd,J=13.2,9.4,3.7Hz,2H),1.53-1.41(m,2H),1.01(s,3H)ppm;LC-MS:m/z 423.1[M+H]+.
EXAMPLE 20 2- (1- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) piperidin-4-yl) ethan-1-amine
Figure BDA0001712865010000731
1H NMR(400MHz,DMSO-d6)δ9.39(s,1H),8.00(s,1H),7.44(d,J=8.0Hz,1H),7.14(t,J=8.0Hz,1H),6.83(d,J=8.1Hz,1H),4.20(d,J=13.3Hz,4H),3.20(d,J=12.5Hz,2H),2.83(t,J=7.7Hz,1H),1.82(d,J=12.7Hz,2H),1.59-1.49(m,2H),1.39-1.33(m,2H),1.23(s,2H)ppm;LC-MS:m/z 424.1[M+H]+.
Example 21- ((2, 3-dichlorophenyl) thio) -5- (3, 5-dimethylpiperazin-1-yl) - [1,2,4] triazolo [4,3-c ] pyrimidine
Figure BDA0001712865010000732
1H NMR(400MHz,DMSO-d6)δ9.52(s,1H),8.14(s,1H),8.03(s,1H),7.45(d,J=7.9Hz,1H),7.14(t,J=8.1Hz,1H),6.83(d,J=8.0Hz,1H),4.12(d,J=12.6Hz,2H),3.23(s,2H),2.94(s,2H),1.15(d,J=5.9Hz,6H)ppm;LC-MS:m/z 410.1[M+H]+.
Example 22 8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -1, 8-diazaspiro [4.5] decane
Figure BDA0001712865010000741
1H NMR(400MHz,DMSO-d6)δ9.47(s,1H),8.02(s,1H),7.43(d,J=8.0Hz,1H),7.14(t,J=8.0Hz,1H),6.85(d,J=8.1Hz,1H),3.97(ddd,J=14.1,6.6,3.9Hz,2H),3.62(ddd,J=13.3,8.9,3.3Hz,2H),3.30(t,J=6.7Hz,2H),2.10(ddd,J=13.1,8.8,3.7Hz,2H),2.00(p,J=6.3Hz,6H)ppm;LC-MS:m/z 435.1[M+H]+.
Example 23- ((5- (4-amino-4-methylpiperidin-1-yl) - [1,2,4] triazolo [4,3-c ] pyrimidin-8-yl) thio) -3-chloropyridin-2-amine
Figure BDA0001712865010000742
1H NMR(400MHz,CD3OD-d4)δ9.24(s,1H),7.94(s,1H),7.42-7.44(d,J=5.2Hz,1H),5.93-5.94(d,J=5.6Hz,1H),3.97-4.02(d,J=14Hz,2H),3.58-3.60(t,J=10.4Hz,2H),1.92-1.98(m,4H),1.44(s,3H)ppm;LC-MS:m/z 391.1[M+H]+.
Example 24 (R) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
The method comprises the following steps: (R) -N- ((R) -8- (8-iodo- [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide
Figure BDA0001712865010000743
To a dry 50mL single neck flask was added 5-chloro-8-iodo- [1,2,4] in sequence]Triazolo [4,3-c]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 (5mL), and the reaction was stirred at 90 ℃ for 2 hours. After completion of the reaction, the obtained residue was poured into water (10mL), and stirred at room temperature for 5 minutes. Then extracted with ethyl acetate (3X 20mL) and the combined organic phases were MgSO4The residue obtained is dried, filtered and concentrated under reduced pressure, purified by chromatography on silica gel (0 to 80% gradient of ethyl acetate/petroleum ether) to give (R) -N- ((R) -8- (8-iodo- [1,2, 4) as a pale yellow solid]Triazolo [4,3-c]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-carboxylic acid methyl esterMesityl-2-sulfinamide (80mg, yield: 88%).
LC-MS:m/z 503.1[M+H]+.
Step two: (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide
A dry 50mL three-necked flask was charged with (R) -N- ((R) -8- (8-iodo- [1,2, 4)]Triazolo [4,3-c]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-methylpropane-2-sulfinamide (80mg,0.16mmol), cuprous iodide (3mg,0.016mmol), 1, 10-phenanthroline (6mg,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 added4Cl solution (50 mL). It was extracted with ethyl acetate (3 × 20 mL). The combined organic phases are washed with Na2SO4Drying, 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) - [1,2,4] as a pale yellow solid]Triazolo [4,3-c]Pyrimidin-5-yl) -8-azaspiro [4.5]Decan-1-yl) -2-methylpropane-2-sulfinamide (60mg, yield: 68%).
LC-MS:m/z 553.1[M+H]+.
Step three: (R) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000761
To a dry 50mL round bottom flask were added sequentially (R) -N- ((R) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide (60mg,0.11mmol) and a solution of hydrochloric acid in 1, 4-dioxane (7M,5mL) and reacted at room temperature for 1 hour. The reaction solution 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) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine (20mg, yield: 46%).
1H NMR(400MHz,MeOH-d4)δ9.15(s,1H),7.90(s,1H),7.23(d,J=8.0Hz,J=1.6Hz,1H),6.97(t,J=8.0Hz,1H),6.74(d,J=8.0Hz,J=1.6Hz,1H),4.03-4.11(m,2H),3.32-3.40(m,2H),3.16(t,J=6.8Hz,1H),2.11-2.14(m,1H),1.51-1.84(m,9H)ppm;LC-MS:m/z 451.1[M+H]+.
Following the synthesis of example 24, the following compounds can be synthesized:
example 25 (S) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000771
1H NMR(400MHz,MeOD-d4)δ9.27(s,1H),8.55(s,2H),8.02(s,1H),7.35(dd,J=8.0,1.3Hz,1H),7.08(t,J=8.1Hz,1H),6.85(d,J=8.1Hz,1H),4.19(s,3H),3.51-3.48(m,2H),3.21(s,1H),3.15(s,1H),2.31-2.14(m,1H),2.03-1.78(m,7H),1.63(s,2H)ppm;LC-MS:m/z 449.1[M+H]+.
Example 26 (S) -7- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -7-azaspiro [3.5] nonan-1-amine
Figure BDA0001712865010000772
1H NMR(400MHz,MeOH-d4)δ9.15(s,1H),8.34(brs,2H),7.90(s,1H),7.23(dd,J=8.0Hz,J=1.6Hz,1H),6.96(t,J=8.0Hz,1H),6.74(dd,J=8.0Hz,J=1.6Hz,1H),4.11(d,J=12.0Hz,1H),4.00(d,J=13.2Hz,1H),3.44-3.40(m,1H),3.35-3.28(m,1H),2.33-2.29(m,1H),2.03-1.95(m,2H),1.91-1.87(m,2H),1.80-1.70(m,3H)ppm;LC-MS:m/z 435.1[M+H]+.
Example 27- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -7-aza-spiro [3.5] non-2-amine
1H NMR(400MHz,Methanol-d4)δ9.28(s,1H),8.01(s,1H),7.34(dd,J=8.0,1.4Hz,1H),7.08(t,J=8.0Hz,1H),6.84(dd,J=8.1,1.4Hz,1H),3.90-3.83(m,1H),3.76(t,J=5.7Hz,2H),3.72–3.66(m,2H),2.45(s,2H),2.05(t,J=10.6Hz,2H),1.95(t,J=5.7Hz,1H),1.91(t,J=5.7Hz,1H)ppm;LC-MS:m/z 435.1[M+H]+.
Example 28 (4R) -2- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) octahydrocyclopenta [ c ] pyrrol-4-amine
Figure BDA0001712865010000781
1H NMR(400MHz,DMSO-d6)δ9.55(s,1H),7.86(s,1H),7.39(d,J=7.9Hz,1H),7.13(d,J=8.1Hz,1H),6.73(d,J=8.1Hz,1H),4.10-4.04(m,2H),3.53(s,1H),3.16(d,J=4.8Hz,1H),2.79(s,1H),1.86-1.78(m,2H),1.55(d,J=8.5Hz,2H)ppm;LC-MS:m/z 421.1[M+H]+.
Example 29 (R) -3- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-azaspiro [5.5] undecan-7-amine
1H NMR(400MHz,DMSO-d6)δ9.37(s,1H),7.98(s,1H),7.43(d,J=7.2Hz,1H),7.14(t,J=8.0Hz,1H),6.81(d,J=8.1Hz,1H),3.99(d,J=13.5Hz,2H),3.59-3.51(m,2H),2.82(d,J=4.6Hz,1H),2.14-1.11(m,14H)ppm;LC-MS:m/z464.1[M+H]+.
Example 30 (R) -1- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) azepan-4-amine
1H NMR(400MHz,DMSO-d6)δ9.41(s,1H),7.93(s,1H),7.43(d,J=7.4Hz,1H),7.14(t,J=8.1Hz,1H),6.77(d,J=8.0Hz,1H),4.15-3.99(m,2H),3.84(ddd,J=52.8,16.3,7.7Hz,2H),3.21(s,3H),2.01(dd,J=56.3,34.4Hz,5H),1.65-1.47(m,1H)ppm;LC-MS:m/z410.1[M+H]+.
Example 31 (S) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
1H NMR(400MHz,DMSO-d6)δ9.39(s,1H),8.21(s,1H),8.00(s,1H),7.47-7.39(m,1H),7.14(t,J=8.0Hz,1H),6.82(d,J=8.1Hz,1H),4.00(dd,J=8.8,6.5Hz,2H),3.73(d,J=8.5Hz,1H),3.66(d,J=8.5Hz,1H),3.54-3.46(m,2H),3.39(dd,J=8.9,5.0Hz,2H),3.21-3.17(m,1H),1.92-1.75(m,2H),1.64-1.53(m,2H)ppm;LC-MS:m/z 450.7[M+H]+.
Example 32 (R) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -N-methyl-8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000793
1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),8.28(s,1H),8.00(s,1H),7.43(d,J=8.0Hz,1H),7.14(t,J=8.1Hz,1H),6.82(d,J=8.0Hz,1H),4.08(d,J=14.0Hz,2H),2.64(d,J=21.8Hz,1H),2.37(s,3H),2.06-1.14(m,12H)ppm;LC-MS:m/z 463.1[M+H]+.
Example 33 (1R) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000801
1H NMR(400MHz,DMSO-d6)δ9.38(d,J=3.5Hz,1H),8.37(s,1H),8.00(d,J=1.0Hz,1H),7.44(d,J=7.1Hz,1H),7.14(t,J=8.0Hz,1H),6.81(dd,J=8.1,1.1Hz,1H),4.15-4.04(m,2H),3.06-2.93(m,2H),2.15(dd,J=19.1,11.5Hz,2H),2.01-1.26(m,8H),1.06-0.99(m,3H)ppm;LC-MS:m/z 464.1[M+H]+.
Example 34 8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000802
1H NMR(CD3OD-d4)δ9.31(s,1H),8.49(s,1H),8.02(s,1H),7.34-7.37(m,1H),7.07-7.11(m,1H),6.86-6.88(m,1H),4.19-4.21(m,2H),4.10-4.12(m,2H),3.85-3.94(m,2H),3.37(m,1H),3.18(m,1H),2.04(m,2H),1.80(m,2H),1.38(s,3H)ppm;LC-MS:m/z 465.1[M+H]+.
Example 35 (3S,4S) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000811
1H NMR(400MHz,DMSO-d6)δ9.40(s,1H),8.18(s,1H),8.01(s,1H),7.44(d,J=7.0Hz,1H),7.14(t,J=8.0Hz,1H),6.82(d,J=7.2Hz,1H),4.12(s,1H),3.89(d,J=6.8Hz,2H),3.74(d,J=8.6Hz,1H),3.59-3.49(m,3H),3.08(d,J=4.8Hz,1H),1.96-1.81(m,2H),1.73(s,2H),1.13(d,J=6.4Hz,3H)ppm;LC-MS:m/z466.1[M+H]+.
Example 36 (3R,4R) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000812
1H NMR(400MHz,DMSO-d6)δ9.31(s,1H),7.95(s,1H),7.40(d,J=7.9Hz,1H),7.11(t,J=8.1Hz,1H),6.78(d,J=8.0Hz,1H),4.68-4.53(m,4H),3.17-3.08(m,1H),1.89(s,2H),1.71-1.61(m,4H),1.13(d,J=6.5Hz,3H);LC-MS:m/z 465.1[M+H]+.
Example 37: 1- (8- (2, 3-dichlorophenyl) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-amine
The method comprises the following steps: (1- (8- (2, 3-dichlorophenyl) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamic acid tert-butyl ester
Figure BDA0001712865010000821
Tert-butyl (1- (8-iodo- [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamate (55mg,0.12mmol), 1, 4-dioxane (2mL), purified water (0.5mL), (2, 3-dichlorophenyl) boronic acid (50mg, 0.24mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (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 carried out for 6 hours. After completion of the reaction, 20mL of water was added to the reaction solution and 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 tert-butyl (1- (8- (2, 3-dichlorophenyl) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamate (33mg, yield: 57%) was obtained as a pale yellow solid by chromatography (petroleum ether: ethyl acetate ═ 1: 1).
LC-MS:m/z 477.1[M+H]+.
Step two: 1- (8- (2, 3-dichlorophenyl) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-amine
Figure BDA0001712865010000822
Following the procedure of example 15, step three, tert-butyl (1- (8- (2, 3-dichlorophenyl) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-yl) carbamate was freed from tert-butoxycarbonyl to give 1- (8- (2, 3-dichlorophenyl) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -4-methylpiperidin-4-amine.
1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),7.79-7.72(m,2H),7.56-7.46(m,2H),3.68(t,J=5.3Hz,4H),1.76-1.51(m,4H),1.18(s,3H)ppm;LC-MS:m/z 377.1[M+H]+.
Example 38: synthesis of (R) -8- (8- (2, 3-dichlorophenyl) - [1,2,4] triazolo [4,3-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) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide
Figure BDA0001712865010000831
(R) -N- ((R) -8- (8-iodo- [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide (60mg,0.12mmol), 1, 4-dioxane (2mL), purified water (0.5mL), (2, 3-dichlorophenyl) boronic acid (50mg, 0.24mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (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 carried out for 6 hours. After completion of the reaction, 20mL of water was added to the reaction solution and 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) - [1,2,4] triazolo [4,3-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: 1). (30mg, yield: 48%) as a pale yellow solid.
LC-MS:m/z 521.1[M+H]+.
Step two: (R) -8- (8- (2, 3-dichlorophenyl) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000841
Following the procedure of example 24, step three, (R) -N- ((R) -8- (8- (2, 3-dichlorophenyl) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-yl) -2-methylpropane-2-sulfinamide is sulfinylated to give (R) -8- (8- (2, 3-dichlorophenyl) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine.
1H NMR(400MHz,DMSO-d6)δ9.35(s,1H),7.81-7.71(m,2H),7.58-7.45(m,2H),3.99(t,J=13.6Hz,2H),3.35(dd,J=22.9,10.6Hz,2H),3.06(t,J=6.2Hz,1H),2.09-1.35(m,10H)ppm;LC-MS:m/z 417.1[M+H]+.
Following the synthesis of example 24, the following compounds can be synthesized:
example 39 methyl (R) -3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) - [1,2,4] triazolo [4,3-c ] pyrimidin-8-yl) thio) propionate
Figure BDA0001712865010000842
1H NMR(400MHz,DMSO-d6)δ9.32(s,1H),8.34(s,1H),7.71(s,1H),3.88(t,J=13.3Hz,2H),3.57(s,3H),3.24(t,J=7.0Hz,4H),3.08(t,J=6.3Hz,1H),2.68-2.59(m,2H),2.03-1.39(m,10H)ppm;LC-MS:m/z 391.1[M+H]+.
EXAMPLE 40 (R) -8- (8- (Phenylthio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000851
1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),7.84(d,J=17.0Hz,1H),7.33-7.19(m,5H),3.98(dd,J=27.7,14.8Hz,2H),3.32(d,J=12.8Hz,2H),3.02(t,J=6.4Hz,1H),2.01-1.39(m,10H)ppm;LC-MS:m/z 381.1[M+H]+.
Example 41 (R) -8- (8- ((2-chlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000852
1H NMR(400MHz,DMSO-d6)δ9.37(s,1H),7.96(s,1H),7.50(dd,J=7.7,1.4Hz,1H),7.16(dtd,J=21.0,7.5,1.5Hz,2H),6.87(dd,J=7.8,1.6Hz,1H),4.07(dd,J=15.4,11.3Hz,2H),3.39(dtd,J=13.7,7.7,3.1Hz,2H),3.05(s,1H),2.00(q,J=7.9Hz,1H),1.92-1.36(m,9H)ppm;LC-MS:m/z 415.1[M+H]+.
Example 42 (R) -8- (8- ((4-chlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000853
1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),7.92(s,1H),7.34(d,J=8.6Hz,2H),7.27(d,J=8.6Hz,2H),4.12-3.92(m,2H),3.35(tt,J=13.8,3.4Hz,2H),3.04(d,J=7.0Hz,1H),1.99(t,J=5.3Hz,1H),1.92-1.33(m,9H)ppm;LC-MS:m/z415.1[M+H]+.
Example 43 (R) -8- (8- ((2, 4-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000861
1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),7.98(s,1H),7.70(d,J=2.2Hz,1H),7.20(dd,J=8.6,2.3Hz,1H),6.88(d,J=8.6Hz,1H),4.05(dd,J=12.3,6.8Hz,2H),2.76(t,J=7.3Hz,1H),1.94-1.75(m,4H),1.66-1.53(m,2H),1.45-1.27(m,4H)ppm;LC-MS:m/z 449.1[M+H]+.
Example 44 (R) -8- (8- ((2, 6-dichlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000862
1H NMR(400MHz,DMSO-d6)δ9.31(s,1H),7.62(d,J=8.1Hz,2H),7.48(dd,J=8.7,7.5Hz,1H),7.34(s,1H),3.82(s,2H),3.21(td,J=11.7,11.3,9.1Hz,2H),2.72(t,J=7.3Hz,1H),1.85(ddt,J=11.8,7.6,3.9Hz,1H),1.77(td,J=12.6,11.4,7.3Hz,3H),1.64-1.49(m,2H),1.42-1.25(m,4H)ppm;LC-MS:m/z 449.1[M+H]+.
Example 45 (R) -8- (8- ((2-isopropylphenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),7.71(s,1H),7.36(d,J=7.7Hz,1H),7.22(ddd,J=8.0,5.5,3.1Hz,1H),7.07-7.02(m,2H),3.95(dd,J=11.9,6.8Hz,2H),3.57-3.47(m,1H),3.32-3.21(m,2H),2.75(t,J=7.3Hz,1H),1.89-1.75(m,4H),1.651.50(m,2H),1.44-1.29(m,4H),1.27(d,J=6.8Hz,6H)ppm;LC-MS:m/z423.1[M+H]+.
EXAMPLE 46 (R) -8- (8- ((2-methoxyphenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000872
1H NMR(400MHz,DMSO-d6)δ9.30(s,1H),7.75(s,1H),7.17(ddd,J=8.5,6.1,2.8Hz,1H),7.03(d,J=8.2Hz,1H),6.80-6.74(m,2H),3.96(dd,J=12.5,8.4Hz,4H),3.86(s,3H),3.35-3.27(m,2H),2.75(t,J=7.2Hz,1H),1.90-1.76(m,2H),1.65-1.15(m,8H)ppm;LC-MS:m/z 411.1[M+H]+.
Example 47 methyl (R) -2- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) - [1,2,4] triazolo [4,3-c ] pyrimidin-8-yl) thio) benzoate
Figure BDA0001712865010000873
1H NMR(400MHz,DMSO-d6)δ9.35(s,1H),7.96(dd,J=8.2,5.3Hz,2H),7.33(t,J=7.7Hz,1H),7.24(t,J=7.4Hz,1H),6.90(d,J=8.0Hz,1H),4.04(dd,J=12.3,7.8Hz,2H),3.92(s,3H),3.39(d,J=10.5Hz,2H),2.78(t,J=7.2Hz,1H),1.89-1.29(m,10H);LC-MS:m/z 439.1[M+H]+.
Example 48 (R) -N- (4- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) - [1,2,4] triazolo [4,3-c ] pyrimidin-8-yl) thio) phenyl) acetamide
Figure BDA0001712865010000881
1H NMR(400MHz,DMSO-d6)δ10.03(s,1H),9.32(s,1H),8.33(s,1H),7.72(s,1H),7.53(d,J=8.5Hz,2H),7.31(d,J=8.5Hz,2H),3.93(t,J=11.8Hz,2H),3.29(s,2H),2.94(s,1H),2.02(s,3H),1.96(s,1H),1.77(t,J=11.4Hz,3H),1.67(s,1H),1.57(d,J=17.2Hz,2H),1.50-1.32(m,3H)ppm;LC-MS:m/z 396.2[M+H]+.
Example 49 (R) -8- (8- ((4-aminophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000882
1H NMR(400MHz,DMSO-d6)δ9.26(d,J=15.3Hz,1H),8.37(s,1H),7.23(dd,J=33.3,27.4Hz,3H),6.55(t,J=20.4Hz,2H),3.78(dd,J=35.2,22.6Hz,4H),3.26-3.16(m,2H),3.01(t,J=6.5Hz,1H),1.98(d,J=10.5Hz,1H),1.89-1.19(m,9H)ppm;LC-MS:m/z396.1[M+H]+.
EXAMPLE 50 (R) -8- (8- ((3-amino-2-chlorophenyl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
1H NMR(400MHz,DMSO-d6)δ9.35(s,1H),8.32(s,1H),7.87(s,1H),6.80(t,J=7.9Hz,1H),6.59(d,J=7.1Hz,1H),6.01(d,J=6.9Hz,1H),5.51(s,2H),4.03(t,J=12.0Hz,2H),3.39(s,2H),2.96(t,J=6.7Hz,1H),1.96-1.44(m,10H)ppm;LC-MS:m/z 430.1[M+H]+.
EXAMPLE 51 (R) -N- (3- ((5- (1-amino-8-azaspiro [4.5] decan-8-yl) - [1,2,4] triazolo [4,3-c ] pyrimidin-8-yl) thio) -2-chlorophenyl) acryloylamide
1H NMR(400MHz,DMSO-d6)δ9.86(s,1H),9.38(s,1H),8.34(s,1H),7.97(s,1H),7.53(d,J=7.7Hz,1H),7.12(t,J=8.0Hz,1H),6.71-6.59(m,2H),6.29(dd,J=17.1,1.8Hz,1H),5.83-5.77(m,1H),4.13-4.03(m,2H),3.53(s,2H),2.94(s,1H),1.92-1.40(m,10H)ppm;LC-MS:m/z 484.1[M+H]+.
EXAMPLE 52 (R) -8- (8- (pyridin-2-ylthio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000901
1H NMR(400MHz,DMSO-d6)δ9.36(s,1H),8.36(d,J=4.7Hz,1H),7.96(s,1H),7.59(t,J=7.7Hz,1H),7.13(d,J=2.3Hz,1H),7.03(d,J=8.0Hz,1H),4.10-3.90(m,2H),3.40(d,J=11.0Hz,2H),2.82(s,1H),1.59(ddd,J=23.4,10.6,4.0Hz,10H)ppm;LC-MS:m/z382.1[M+H]+.
Example 53 (R) -8- (8- ((3-Chloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000902
1H NMR(400MHz,DMSO-d6)δ9.41(s,1H),8.56(s,1H),8.28(s,1H),8.19(d,J=5.3Hz,1H),8.04(s,1H),6.84(d,J=5.3Hz,1H),4.11(t,J=12.3Hz,2H),3.43(s,2H),3.00(t,J=6.7Hz,1H),1.99-1.43(m,10H)ppm;LC-MS:m/z 416.1[M+H]+.
Example 54 (R) -8- (8- ((2, 3-dichloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000903
1HNMR(CD3OD-d4)δ9.42(s,1H),8.34(s,1H),8.03-8.05(m,2H),6.88-6.89(m,1H),4.11-4.12(m,2H),3.44-3.47(m,2H),2.89(m,1H),1.71-2.05(m,4H),1.35-1.68(m,6H)ppm;LC-MS:m/z 449.8[M+H]+.
Example 55 (R) -8- (8- ((3-chloro-2-methylpyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000911
1H NMR(400MHz,DMSO-d6)δ9.41(s,1H),8.31(s,1H),8.05(d,J=5.4Hz,1H),8.02(s,1H),6.67(d,J=5.3Hz,1H),4.10(t,J=12.9Hz,2H),3.46-3.40(m,2H),3.01(s,1H),1.99(s,1H),1.88-1.66(m,4H),1.65-1.39(m,5H)ppm;LC-MS:m/z 430.1[M+H]+.
Example 56 (R) -8- (8- ((3-chloro-2- (dimethylamino) pyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000912
1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),7.98(d,J=4.2Hz,1H),7.82(d,J=5.3Hz,1H),6.31(d,J=5.3Hz,1H),4.16-3.94(m,2H),3.41(dd,J=15.8,12.6Hz,2H),2.91(s,6H),2.84(dd,J=13.1,5.9Hz,1H),1.90-1.36(m,10H)ppm;LC-MS:m/z 459.1[M+H]+.
Example 57 (R) -8- (8- ((2-amino-5-chloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
1H NMR(400MHz,DMSO-d6)δ9.45(s,1H),8.37(s,1H),7.99(s,1H),6.56(s,1H),5.78(s,2H),5.59(s,1H),3.98(s,2H),3.72(s,1H),3.63(d,J=8.3Hz,1H),3.12(s,1H),1.67(d,J=79.0Hz,10H)ppm;LC-MS:m/z 432.1[M+H]+.
Example 58 (R) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000922
1H NMR(400MHz,DMSO-d6)δ9.46(s,1H),8.30(s,1H),8.03(s,1H),6.31(s,2H),5.71(s,1H),4.11(t,J=12.1Hz,2H),3.44(d,J=12.4Hz,2H),2.92(d,J=7.5Hz,1H),1.96-1.77(m,4H),1.70-1.64(m,2H),1.56-1.40(m,4H).
LC-MS:m/z 465.1[M+H]+.
Example 59 (R) -8- (8- ((2-methylpyridin-3-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000923
1H NMR(400MHz,DMSO-d6)δ9.35(s,1H),8.26(dd,J=4.7,1.5Hz,1H),7.91(d,J=3.0Hz,1H),7.31(dd,J=8.0,1.4Hz,1H),7.07(dd,J=7.9,4.7Hz,1H),4.05-3.94(m,2H),3.30(s,3H),2.80(t,J=7.3Hz,1H),2.61(s,3H),1.89-1.25(m,10H)ppm;LC-MS:m/z 449.8[M+H]+.
EXAMPLE 60 (R) -8- (8- ((2- (trifluoromethyl) pyridin-3-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000931
1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),8.49(d,J=4.5Hz,1H),8.36(s,1H),8.02(s,1H),7.57(d,J=8.3Hz,1H),7.46(dd,J=8.3,4.5Hz,1H),4.08(s,2H),2.94(s,2H),1.98(dd,J=14.4,7.7Hz,2H),1.88-1.64(m,4H),1.52(d,J=42.8Hz,6H)ppm;LC-MS:m/z450.1[M+H]+.
Example 61 (R) -8- (8- (naphthalen-1-ylthio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000932
1H NMR(400MHz,DMSO-d6)δ9.33(s,1H),8.39(d,J=8.2Hz,1H),8.27(d,J=11.7Hz,1H),8.00(d,J=7.7Hz,1H),7.88(d,J=7.9Hz,1H),7.73(s,1H),7.69-7.59(m,2H),7.47(d,J=7.1Hz,1H),7.44-7.38(m,1H),4.03-3.92(m,2H),3.28(s,2H),2.97(s,1H),1.68(dt,J=107.3,31.9Hz,10H)ppm;LC-MS:m/z 431.2[M+H]+.
Example 62 (R) -8- (8- (quinolin-4-ylthio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000941
1H NMR(400MHz,DMSO-d6)δ9.40(s,1H),8.54(d,J=4.7Hz,1H),8.26(d,J=8.4Hz,1H),8.11-7.99(m,2H),7.85(t,J=7.6Hz,1H),7.74(t,J=7.6Hz,1H),6.95(d,J=4.7Hz,1H),4.19-3.99(m,2H),3.43(dd,J=17.3,8.7Hz,2H),2.88(t,J=7.0Hz,1H),1.94-1.37(m,10H)ppm;LC-MS:m/z 432.2[M+H]+.
Example 63 (R) -8- (8- ((1-methyl-1H-imidazol-2-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000942
1H NMR(400MHz,DMSO-d6)δ9.33(s,1H),7.54(s,1H),7.37(d,J=1.2Hz,1H),6.97(d,J=1.2Hz,1H),3.85(d,J=2.4Hz,5H),3.29-3.20(m,2H),2.73(t,J=7.3Hz,1H),1.87-1.71(m,4H),1.63-1.49(m,2H),1.42-1.25(m,4H)ppm;LC-MS:m/z 385.2[M+H]+.
Example 64 (1R) -8- (8- ((2, 3-dichloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000943
1H NMR(400MHz,DMSO-d6)δ9.41(s,1H),8.25(s,1H),8.04(d,J=5.3Hz,1H),8.02(s,1H),6.67(d,J=5.3Hz,1H),4.02-3.96(m,2H),3.73(d,J=8.4Hz,1H),3.64(d,J=8.5Hz,1H),3.52(d,J=10.0Hz,2H),3.18-3.09(m,3H),2.56(s,3H),1.84(dd,J=40.8,9.5Hz,2H),1.57(d,J=12.5Hz,2H)ppm;LC-MS:m/z434.1[M+H]+.
Example 65 (1R) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000951
1H NMR(CD3OD-d4)δ9.37(s,1H),7.98(s,1H),7.57(s,1H),6.36(m,2H),5.93(m,1H),4.16(m,2H),3.44(m,2H),3.10(m,1H),2.42(m,1H),1.31-2.13(m,8H),1.01-1.05(m,3H)ppm;LC-MS:m/z 445.1M+H]+.
Example 66 (1R) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010000952
1H NMR(CD3OD-d4)δ9.45-9.46(m,1H),8.04(s,1H),6.32(m,2H),5.74(m,1H),4.14-4.17(m,2H),3.45(m,2H),3.14(m,2H),1.45-2.41(m,8H),1.02-1.05(m,3H)ppm;LC-MS:m/z 479.1M+H]+.
Example 67 (S) -8- (8- ((2, 3-dichloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000961
1H NMR(400MHz,DMSO-d6)δ9.43(s,1H),8.08-7.99(m,2H),6.88(d,J=5.3Hz,1H),4.08-3.95(m,3H),3.75-3.66(m,2H),3.51(d,J=13.6Hz,2H),3.24-3.15(m,2H),1.93-1.75(m,2H),1.60(d,J=13.1Hz,2H),1.32(s,2H)ppm;LC-MS:m/z 452.1[M+H]+.
Example 68 (S) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),8.28(s,1H),7.97(s,1H),7.57(d,J=5.4Hz,1H),6.34(s,2H),5.95(d,J=5.4Hz,1H),3.97(tt,J=13.8,5.4Hz,3H),3.72(d,J=8.5Hz,1H),3.64(d,J=8.4Hz,1H),3.49(dq,J=10.7,4.4,2.9Hz,2H),3.14(t,J=5.9Hz,2H),1.93-1.74(m,2H),1.57(dt,J=14.0,4.3Hz,2H)ppm;LC-MS:m/z 433.1[M+H]+.
Example 69 (S) -8- (8- ((3-chloro-2-methylpyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000971
1H NMR(400MHz,DMSO-d6)δ9.41(s,1H),8.25(s,1H),8.04(d,J=5.3Hz,1H),8.02(s,1H),6.67(d,J=5.3Hz,1H),4.02-3.96(m,2H),3.73(d,J=8.4Hz,1H),3.64(d,J=8.5Hz,1H),3.52(d,J=10.0Hz,2H),3.18-3.09(m,3H),2.56(s,3H),1.84(dd,J=40.8,9.5Hz,2H),1.57(d,J=12.5Hz,2H)ppm;LC-MS:m/z434.1M+H]+.
Example 70 (S) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000972
1H NMR(400MHz,DMSO-d6)δ9.47(s,1H),8.25(s,1H),8.04(s,1H),6.30(s,2H),5.71(s,1H),4.09-3.92(m,4H),3.73(d,J=8.4Hz,1H),3.65(d,J=8.4Hz,1H),3.49(s,2H),3.16(d,J=6.6Hz,1H),1.88(d,J=10.0Hz,2H),1.59(s,2H)ppm;LC-MS:m/z 468[M+H]+.
Example 71 (3S,4S) -8- (8- ((2, 3-dichloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000973
1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),8.09-7.93(m,2H),6.84(d,J=5.3Hz,1H),4.10(q,J=6.1Hz,2H),3.91(dd,J=13.8,5.5Hz,4H),3.56(m,1H),2.98(d,J=5.0Hz,1H),1.98-1.55(m,4H),1.10(d,J=6.3Hz,3H)ppm;LC-MS:m/z 466.1[M+H]+.
Example 72 (3S,4S) -8- (8- ((3-chloro-2-methylpyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000981
1H NMR(400MHz,DMSO-d6)δ9.41(s,1H),8.04(d,J=5.4Hz,1H),8.01(s,1H),6.67(d,J=5.3Hz,1H),4.12-4.06(m,1H),3.86(s,2H),3.69(d,J=8.4Hz,1H),3.60(dd,J=21.1,9.5Hz,2H),3.52(d,J=8.5Hz,1H),2.96(d,J=4.9Hz,1H),2.56(s,3H),1.88(d,J=48.6Hz,2H),1.64(s,2H),1.08(dd,J=15.3,6.5Hz,3H)ppm;LC-MS:m/z 446.1[M+H]+.
Example 73 (3S,4S) -8- (8- ((2-amino-5-chloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000982
1H NMR(400MHz,DMSO-d6)δ9.41(s,1H),7.99(s,1H),7.82(s,1H),5.83(s,2H),5.76(s,1H),4.14(p,J=6.3Hz,2H),3.96(ddd,J=15.1,10.0,5.1Hz,4H),3.60(s,1H),3.14(d,J=5.0Hz,1H),1.97-1.62(m,4H),1.13(t,J=5.9Hz,3H)ppm;LC-MS:m/z 447.1[M+H]+.
Example 74 (3S,4S) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000991
1H NMR(CD3OD-d4)δ9.39(s,1H),7.97(s,1H),7.57(s,1H),6.35(m,2H),5.95(m,1H),4.09-4.11(m,1H),3.86-3.90(m,2H),3.70-3.72(m,1H),3.54-3.62(m,3H),3.00-3.02(m,1H),1.93-1.95(m,1H),1.80-1.83(m,1H),1.60-1.70(m,2H),1.10-1.12(m,3H)ppm;LC-MS:m/z 447.1[M+H]+.
Example 75 (3S,4S) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010000992
1H NMR(400MHz,DMSO-d6)δ9.49(s,1H),8.06(s,1H),6.31(s,2H),5.71(s,1H),4.26-4.19(m,1H),4.06(s,2H),3.88(d,J=9.1Hz,1H),3.71(d,J=8.8Hz,1H),3.57-3.37(m,4H),1.91(s,2H),1.69(s,2H),1.21(d,J=6.4Hz,3H)ppm;LC-MS:m/z 481.1[M+H]+.
Example 76 (3R,4S) -8- (8- ((3-chloro-2-methylpyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010001001
1H NMR(400MHz,DMSO-d6)δ9.41(d,J=5.4Hz,1H),8.04(d,J=5.3Hz,1H),8.01(s,1H),6.67(d,J=5.2Hz,1H),4.12(dd,J=33.3,13.6Hz,2H),3.73(dd,J=23.6,8.8Hz,2H),3.39(dd,J=16.2,10.4Hz,2H),3.30(s,1H),2.54(d,J=16.4Hz,3H),2.42(d,J=8.2Hz,1H),1.93-1.81(m,2H),1.50(d,J=13.5Hz,2H),1.21(d,J=6.0Hz,3H)ppm;LC-MS:m/z446.1[M+H]+.
Example 77 (3R,4R) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) - [1,2,4] triazolo [4,3-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010001002
1H NMR(400MHz,DMSO-d6)δ9.35(s,1H),7.95(s,1H),7.55(d,J=5.4Hz,1H),6.27(s,2H),5.95(d,J=5.4Hz,1H),4.08(p,J=6.3Hz,2H),3.85-3.81(m,2H),3.55(s,2H),2.95(d,J=5.0Hz,1H),1.99-1.56(m,5H),1.09(d,J=6.4Hz,3H)ppm;LC-MS:m/z 447.1[M+H]+.
Example 78 1- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -4-methylpiperidin-4-amine
1H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.23(s,1H),7.40(dd,J=8.0,1.2Hz,1H),7.12(t,J=8.0Hz,1H),6.74(dd,J=8.1,1.2Hz,1H),4.54-4.30(m,2H),4.15-3.94(m,2H),1.76-1.48(m,4H),1.18(s,3H)ppm;LC-MS:m/z 409.1[M+H]+.
Example 79 (R) -1- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) azepine-4-amine
Figure BDA0001712865010001011
1H NMR(400MHz,DMSO-d6)δ8.47(s,1H),8.21(s,1H),7.41(d,J=7.5Hz,1H),7.13(t,J=8.1Hz,1H),6.74(d,J=7.9Hz,1H),4.15(dd,J=90.0,47.9Hz,4H),3.21(s,2H),2.22(s,1H),1.94(d,J=56.0Hz,5H),1.61(d,J=12.2Hz,1H)ppm;LC-MS:m/z 408.7[M+H]+.
EXAMPLE 80 (R) -3- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -3-azaspiro [5.5] undecan-7-amine
1H NMR(400MHz,DMSO-d6)δ8.49(s,1H),8.24(s,1H),7.42(d,J=7.1Hz,1H),7.12(t,J=8.0Hz,1H),6.73(d,J=8.0Hz,1H),4.84(s,2H),3.67-3.53(m,2H),2.89(d,J=4.3Hz,1H),1.98(dd,J=24.0,13.1Hz,2H),1.48(ddd,J=74.9,37.6,6.9Hz,12H)ppm;LC-MS:m/z 462.7[M+H]+.
Example 81 (3R,4S) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010001021
1H NMR(400MHz,DMSO-d6)δ8.48(d,J=6.0Hz,1H),8.23(d,J=6.3Hz,1H),7.41(d,J=8.0Hz,1H),7.12(t,J=8.1Hz,1H),6.74(d,J=8.1Hz,1H),4.95(dd,J=37.1,13.4Hz,2H),3.76(dd,J=26.2,8.9Hz,2H),3.44(d,J=7.8Hz,1H),2.42(d,J=8.1Hz,1H),1.82(t,J=11.6Hz,2H),1.50(d,J=13.6Hz,2H),1.28-1.17(m,5H)ppm;LC-MS:m/z 465.1[M+H]+.
Example 82 (S) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010001022
1H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.32(s,1H),8.24(s,1H),7.41(dd,J=8.0Hz,1.2Hz,1H),7.13(t,J=8.0Hz,1H),6.72(dd,J=8.0Hz,J=1.6Hz,1H),4.76(t,J=16.0Hz,2H),4.06(dd,J=9.6Hz,2.4Hz,1H),3.81(s,2H),3.67-3.54(m,3H),3.35(t,J=4.8Hz,1H),1.87-1.81(m,2H),1.68-1.65(m,2H)ppm;LC-MS:m/z 451.1[M+H]+.
Example 83 (3R,4R) -8- (8- ((2, 3-dichlorophenyl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010001023
1H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.23(s,1H),7.41(d,J=8.0Hz,1H),7.12(t,J=8.1Hz,1H),6.74(d,J=8.1Hz,1H),4.45(d,J=13.1Hz,2H),4.11-4.05(m,2H),3.96-3.80(m,2H),2.96(d,J=5.1Hz,1H),2.01-1.93(m,2H),1.78-1.56(m,4H),1.09(d,J=6.4Hz,3H);LC-MS:m/z 465.1[M+H]+.
EXAMPLE 84 (R) -8- (8- (Phenylthio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010001031
1H NMR(400MHz,DMSO-d6)δ8.49(s,1H),8.17(s,1H),7.29-7.24(m,2H),7.18(dd,J=12.1,7.2Hz,3H),4.86(t,J=11.7Hz,2H),3.44(t,J=12.5Hz,2H),3.01(t,J=6.7Hz,1H),1.97(dd,J=12.7,7.3Hz,1H),1.85-1.41(m,9H)ppm;LC-MS:m/z 381.2[M+H]+.
Example 85 (R) -8- (8- ((2-methoxyphenyl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
1H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.34(s,1H),8.10(s,1H),7.14(t,J=7.8Hz,1H),7.02(d,J=8.2Hz,1H),6.75(t,J=7.6Hz,1H),6.64(d,J=7.7Hz,2H),4.87(d,J=12.5Hz,3H),3.88(s,2H),2.93(d,J=7.2Hz,1H),2.02-1.92(m,2H),1.82-1.37(m,8H)ppm;LC-MS:m/z 411.1[M+H]+.
Example 86 (R) -8- (8- ((4-aminophenyl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010001041
1H 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;LC-MS:m/z 396.2[M+H]+.
Example 87 (R) -8- (8- ((2- (trifluoromethyl) phenyl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010001042
1H NMR(400MHz,DMSO-d6)δ8.47(d,J=10.2Hz,1H),8.22(d,J=11.4Hz,1H),7.57(s,1H),7.54-7.36(m,3H),4.97-4.72(m,2H),3.47(t,J=9.7Hz,2H),2.72(t,J=7.5Hz,1H),1.83-1.26(m,10H);LC-MS:m/z 449.1[M+H]+.
Example 88 (R) -8- (8- (pyridin-3-ylthio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010001043
1H NMR(400MHz,DMSO-d6)δ8.49(s,1H),8.36(s,1H),8.17(s,1H),7.27(d,J=7.6Hz,2H),7.23-7.13(m,3H),4.86(t,J=12.3Hz,2H),3.47(d,J=2.6Hz,2H),2.96(t,J=7.0Hz,1H),1.98(q,J=5.7,4.2Hz,1H),1.92-1.34(m,9H)ppm;LC-MS:m/z 382.2[M+H]+.
EXAMPLE 89 (R) -8- (8- ((3-Chloropyridin-4-yl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010001051
1H NMR(CD3OD-d4)δ8.50-8.54(m,2H),8.37(s,1H),8.26(m,1H),8.16-8.17(s,1H),6.79-6.80(m,1H),4.94(m,2H),3.48-3.53(m,2H),2.91(m,1H),1.71-2.05(m,4H),1.35-1.68(m,6H)ppm;LC-MS:m/z 416.1[M+H]+.
Example 90 (R) -8- (8- ((3- (trifluoromethyl) pyridin-4-yl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
1H NMR(400MHz,DMSO-d6)δ8.82(s,1H),8.49(s,1H),8.42(d,J=5.5Hz,1H),8.35(s,1H),8.28(s,1H),7.01(d,J=5.5Hz,1H),4.96(s,2H),3.51(t,J=11.8Hz,3H),2.96(t,J=7.0Hz,1H),2.02-1.35(m,10H)ppm;LC-MS:m/z 450.1[M+H]+.
Example 91 (R) -8- (8- ((2-Chloropyridin-3-yl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
1H NMR(CD3OD-d4)δ8.49(m,1H),8.36(s,1H),8.26(m,1H),8.16-8.18(s,1H),7.20-7.23(m,2H),4.96-4.94(m,2H),3.50-3.46(m,2H),2.96-2.90(m,1H),1.71-2.05(m,4H),1.35-1.68(m,6H)ppm.LC-MS:m/z 416.1[M+H]+.
Example 92 (R) -8- (8- ((2-methylpyridin-3-yl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
1H NMR(400MHz,DMSO-d6)δ8.50(s,1H),8.24-8.22(m,1H),8.21(s,1H),7.22(d,J=8.1Hz,1H),7.04(dd,J=7.9,4.5Hz,1H),3.52-3.44(m,2H),3.18-3.10(m,3H),2.59(s,3H),2.04(s,1H),1.86-1.35(m,9H)ppm.LC-MS:m/z 396.1[M+H]+.
Example 93 (R) -8- (8- ((6-amino-2-chloropyridin-3-yl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
1H NMR(400MHz,DMSO-d6)δ8.52(s,1H),7.98(s,1H),7.40(d,J=8.5Hz,1H),6.61(s,2H),6.33(d,J=8.5Hz,1H),4.78(s,2H),3.16(d,J=5.0Hz,1H),3.04(s,1H),2.67(s,2H),1.82-1.40(m,10H)ppm;LC-MS:m/z 431.2[M+H]+.
Example 94 (R) -8- (8- ((2-amino-3-chloropyridin-4-yl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010001071
1H NMR(CD3OD-d4)δ8.48(s,1H),8.20(s,1H),7.55(s,1H),6.36(s,2H),5.87(m,1H),4.90(m,2H),3.45-3.48(m,2H),2.74-2.76(m,1H),1.76-1.83(m,4H),1.51-1.63(m,2H),1.35-1.46(m,4H)ppm;LC-MS:m/z 431.1M+H]+.
Example 95 (R) -8- (8- ((6-amino-2, 3-dichloropyridin-4-yl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -8-azaspiro [4.5] decan-1-amine
Figure BDA0001712865010001072
1H NMR(400MHz,CD3OD-d4)δ8.25(s,1H),8.09(s,1H),5.64(s,1H),5.06(s,2H),3.45-3.37(m,2H),2.73(t,J=7.5Hz,1H),1.97-1.67(m,5H),1.54(dd,J=11.6,5.0Hz,2H),1.46-1.31(m,3H)ppm;LC-MS:m/z 467.1M+H]+.
EXAMPLE 96 (S) -8- (8- ((3-chloro-2-methylpyridin-4-yl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010001073
1H NMR(400MHz,DMSO-d6)δ8.50(s,1H),8.24(d,J=3.4Hz,2H),8.03(d,J=5.3Hz,1H),6.61(d,J=5.4Hz,1H),4.02-3.97(m,2H),3.78(s,2H),3.69(s,2H),3.17(s,3H),2.55(s,3H),1.80(d,J=27.7Hz,2H),1.59(s,2H)ppm;LC-MS:m/z434.1[M+H]+.
Example 97 (3S,4S) -8- (8- ((3-chloro-2-methylpyridin-4-yl) thio) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-yl) -3-methyl-2-oxa-8-azaspiro [4.5] decan-4-amine
Figure BDA0001712865010001081
1H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.23(s,1H),8.01(d,J=5.3Hz,1H),6.59(d,J=5.4Hz,1H),4.60(d,J=13.5Hz,2H),4.13(d,J=5.9Hz,1H),3.75(dd,J=21.8,9.9Hz,4H),3.14(d,J=19.8Hz,1H),2.54(s,3H),1.97-1.60(m,4H),1.13(d,J=6.4Hz,3H)ppm;LC-MS:m/z 446.1[M+H]+.
EXAMPLES 98-100 pharmacologically relevant examples
Example 98: 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 mutation of amino acid at position 76E76AThe substrate used was DiFMUP (Invitrogen, E12020). SHP2E76AThe protein and substrate were diluted to 1.2nM and 20. mu.M, respectively, with buffer (0.1M NaAc (pH7.2), 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 455nm) the fluorescence signal every 1 minute on a Spectra max i3(molecular devices) instrument. Nonlinear regression analysis was performed with GraphPad Prism 5 by Y ═ Bottom + (Top-Bottom)/(1+ 1)The 0^ ((LogIC 50-X). HillSlope) equation fitted a curve of enzyme activity as a function of compound concentration. Determination of IC of each Compound50The value is obtained. Results
The following table shows the IC of some of the compounds of the invention50The value is obtained.
Letter A stands for IC50Less than 100 nM;
letter B stands for IC50100nM to 1000 nM;
letter C stands for IC501000nM to 10000 nM;
Figure BDA0001712865010001091
Figure BDA0001712865010001101
Figure BDA0001712865010001111
Figure BDA0001712865010001121
example 99 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. Firstly, diluting a compound to be detected by 3-fold with 100% DMSO, and setting 9 different concentration gradients in total; then, 30000 cells per hole are inoculated into 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%.
Second step cell lysis. After 2 hours of cell treatment, the medium was removed, 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 BDA0001712865010001122
UltraTMp-ERK 1/2(Thr202/Tyr204) kit (Perkin Elmer, ALSU-PERK-A10K)) detects phosphorylated extracellular signal-regulated kinase (p-ERK). Mu.l of the lysate were transferred 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 invention50The value is obtained.
Letter A stands for IC50Less than 100 nM;
letter B stands for IC50100nM to 1000 nM;
letter C stands for IC501000nM to 10000 nM;
Figure BDA0001712865010001131
Figure BDA0001712865010001141
example 100 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 are immediately treated with the test compound 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, ZG7572) 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.
Results
The following table showsIC of a part of the compounds of the invention50The value is obtained.
Letter A stands for IC50Less than 100 nM;
letter B stands for IC50100nM to 1000 nM;
letter C stands for IC501000nM to 10000 nM;
Figure BDA0001712865010001161
according to the same test method as SHP2 enzyme activity inhibition test described in example 98, phosphoprotein kinase (p-ERK) cell test described in example 99 and MOLM-13 cell proliferation test described in example 100, the applicant carried out corresponding experiments with respect to SHP099(6- (4-amino-4-methylpiperidin-1-yl) -3- (2, 3-dichlorophenyl) pyrazin-2-amine) disclosed in WO 2015/107493A1 or the literature (Nature 2016,535, 148-152), and the data of comparative experiments between the compounds obtained in some examples of the present invention and SHP099 are shown in the following table, and it was found that the pyrimido-cyclic compounds of the present invention have superior activity by comparison.
Figure BDA0001712865010001171
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 (21)

1. A pyrimido ring compound represented by formula (I), a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer or a solvate thereof,
Figure FDA0001711221000000011
wherein
Z1Is C, Z2Is N or Z1Is N, Z2Is C;
x is independently S or absent;
y is independently C or N;
n is independently 0, 1 or 2;
R1independently 0 to 4R1aSubstituted phenyl, 0-4R1aSubstituted containing 1-4 azaaryl groups, 0-4R1aSubstituted naphthyl, 0-4R1aSubstituted azanaphthalene aryl containing 1-4, 0-4R1aSubstituted or unsubstituted benzo-heterocycle, 0-4R1aSubstituted or unsubstituted aromatic ring containing 1-4 nitrogen heterocyclic ring, 0-4R1aSubstituted containing 1-4N, NR1bHetero-aromatic ring of hetero atoms O or S (O), R1cSubstituted or unsubstituted C1-8Alkyl radical, R1cSubstituted or unsubstituted C1-8A haloalkyl group; wherein m is selected from 0, 1 and 2;
R1aindependently is halogen, R1a1Substituted or unsubstituted C1-4Alkoxy radical, R1a1Substituted or unsubstituted C1-4Alkyl, trifluoromethyl, C (═ O) OR1a2、NR1a2R1a3、NHC(=O)R1a4、R1a1Substituted or unsubstituted C3-8A cycloalkyl group; r1a1Independently is halogen or C1-4An alkyl group; r1a2、R1a3Independently is hydrogen, C1-4An alkyl group; r1a4Independently is C1-4Alkyl, substituted or unsubstituted alkenyl, amide, C3-12Mono-or poly-heterocyclic;
R1bindependently is hydrogen, R1a1Substituted or unsubstituted C1-4An alkyl group;
R1cindependently hydrogen, -C (═ O) OR1a2、R1a1Substituted or unsubstituted C1-4An alkyl group;
R2a、R2b、R3aand R3bIndependently isHydrogen, R1a1Substituted or unsubstituted C1-4An alkyl group;
when Y is N, R4Independently is hydrogen, R1a1Substituted or unsubstituted C1-4An alkyl group; r5Is absent;
when Y is ═ C, R4、R5Independently are hydrogen, aryl, C1-4Alkyl radical, C1-4Alkoxy, -O-C1-4Alkyl, amino, C1-4Alkyl substituted amino, -O-C1-4Alkyl-substituted amino, or R4And R5Together with Y form 0-3R4aA substituted 3 to 7 membered saturated or partially unsaturated spirocyclic ring, which ring may optionally contain 1-3 heteroatoms or groups independently selected from N, C (═ O) and/or O;
R4aindependently are hydrogen, halogen, R1a1Substituted or unsubstituted C1-4Alkoxy radical, R1a1Substituted or unsubstituted C1-4Alkyl, hydroxy, amino, C1-4An alkylamino group.
2. The pyrimido compound, the pharmaceutically acceptable salt thereof, the hydrate thereof, the prodrug thereof, the stereoisomer thereof, or the solvate thereof according to claim 1, wherein R1Selected from the following structures:
Figure FDA0001711221000000021
wherein o is 0, 1,2,3 or 4; ring a is a heteroaryl group containing 1-4N atoms; ring B is a heteroaryl group containing 1-4 heteroatoms of N, S, O; g is independently C, C (═ O), N, S, or an O heteroatom or group; r1aa、R1abIndependently R as defined in claim 11a;R1acIndependently is R1cSubstituted or unsubstituted C1-8Alkyl radical, R1cSubstituted or unsubstituted C1-8An alkyl halide.
3. The pyrimido compound of claim 1, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, orSolvates thereof wherein R2a、R2b、R3aAnd R3bIndependently hydrogen or methyl.
4. The pyrimido ring compound of claim 1, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, wherein R is when Y ═ N4Independently hydrogen, methyl; r5Is absent.
5. The pyrimido ring compound of claim 1, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, wherein when Y ═ C, R4、R5Independently hydrogen, methyl, ethyl, phenyl, amino, methylamino or ethylamino.
6. The pyrimido ring compound of claim 1, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, wherein when Y ═ C, R4And R5The ring formed with Y is selected from the following structures:
Figure FDA0001711221000000031
wherein p is 0, 1,2 or 3; r4aAs defined in claim 1.
7. The pyrimido ring compound of claim 1, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, wherein when Y ═ C, R4And R5The ring formed with Y is of the following configuration:
wherein p is 0, 1,2 or 3; r4aAs defined in claim 1.
8. The pyrimido compound, the pharmaceutically acceptable salt, the hydrate, the prodrug, the stereoisomer, or the solvate thereof according to claim 1, which is selected from any one of the following compounds:
Figure FDA0001711221000000041
Figure FDA0001711221000000051
Figure FDA0001711221000000071
9. a pyrimido compound according to any of claims 1 to 8, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, a solvate or an isotopically labeled compound thereof, wherein the isotope is selected from the group consisting of2H、3H、11C、13C、14C、15N、18F、31P、32P、35S、36Cl and125I。
10. a process for preparing a pyrimido-cyclic compound of formula (I) comprising the steps of:
the halogenated intermediate compound A and F are subjected to coupling reaction to obtain a formula (I), and the reaction equation is as follows:
Figure FDA0001711221000000072
wherein the content of the first and second substances,
f represents boric acid, thiol or sodium sulfide;
W1represents halogen, preferably Br,I;X、Y、n、Z1、Z2、R1、R2a、R2b、R3a、R3b、R4And R5Is as defined in claim 1.
11. A process for producing a pyrimido-cyclic compound represented by formula (I-B), which comprises the steps of:
removing the amino protecting group of intermediate I-B1 under acidic or basic conditions to obtain compound I-B, wherein the reaction equation is as follows:
Figure FDA0001711221000000073
wherein, X, Z1、Z2、n、R1、R2a、R2b、R3a、R3b、R4、R5As defined in claim 1; pg is selected from protecting groups Boc, Ac, S (═ O)tBu;R4Pg、R5PgTogether with the attached carbon, is selected from the following structures:
Figure FDA0001711221000000081
R4、R5together with the attached carbon, is selected from the following structures:
Figure FDA0001711221000000082
wherein p is 0, 1,2 or 3.
12. A method for producing a pyrimido-cyclic compound represented by formula (I-C), comprising the steps of:
aminoacylation of intermediate I-C1 yields compound I-C according to the following reaction equation:
Figure FDA0001711221000000083
wherein, X, Y, n, R2a、R2b、R3a、R3b、R4、R5、R1aAnd R1a4Is as defined in claim 1.
13. A process for the preparation of intermediate compound a comprising the steps of:
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 FDA0001711221000000091
wherein, W2Represents halogen, preferably Cl; y, n, R2a、R2b、R3a、R3b、R4And R5As defined in claim 1; w1Is as defined in claim 10.
14. An intermediate for the preparation of a pyrimido-cyclic compound of formula (I) according to any of claims 1 to 8, selected from:
Figure FDA0001711221000000092
wherein Y, n, R1、R2、R3、R4、R5And R1aAs defined in claim 1; w1As defined in claim 10; w2Is as defined in claim 13.
15. Use of a pyrimido compound according to any of claims 1 to 8, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer or a solvate thereof, or an isotopically labeled compound according to claim 9, for the preparation of a medicament for the prevention and/or treatment of a disease or disorder associated with abnormal activity of SHP 2.
16. Use according to claim 15, wherein the disease or disorder is 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.
17. A pharmaceutical composition comprising a pyrimido-cyclic compound according to any one of claims 1 to 8, a pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof, or an isotopically labeled compound of claim 9, and a pharmaceutically acceptable excipient.
18. A pharmaceutical formulation comprising a pyrimido compound according to any of claims 1 to 8, a pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof, or an isotopically labeled compound according to claim 9.
19. A pharmaceutical formulation according to claim 18, which is administered in a manner selected from the group consisting of: oral, sublingual, subcutaneous, intravenous, intramuscular, intrasternal, nasal, topical or rectal administration.
20. A pharmaceutical formulation according to claim 18, which is administered in a single dose or in multiple doses per day.
21. A pyrimido compound according to any of claims 1 to 8, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer or a solvate thereof, or an isotopically labeled compound according to claim 9, in combination with a further drug selected from the group consisting of: anticancer drugs, tumor immunity drugs, antiallergic drugs, antiemetic drugs, analgesic drugs, and cytoprotective drugs.
CN201810692211.4A 2018-02-13 2018-06-29 Pyrimido-cyclic compounds, process for their preparation and their use Pending CN110655520A (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CN201810692211.4A CN110655520A (en) 2018-06-29 2018-06-29 Pyrimido-cyclic compounds, process for their preparation and their use
SG11202007740TA SG11202007740TA (en) 2018-02-13 2019-02-03 Pyrimidine-fused cyclic compound, preparation method therefor and application thereof
JP2020543947A JP7335882B2 (en) 2018-02-13 2019-02-03 Pyrimidine-condensed ring compound, method for producing the same, and use
US16/969,392 US11498930B2 (en) 2018-02-13 2019-02-03 Pyrimidine-fused cyclic compound, preparation method therefor and application thereof
AU2019222026A AU2019222026B2 (en) 2018-02-13 2019-02-03 Pyrimidine-fused cyclic compound, preparation method therefor and application thereof
MA051845A MA51845A (en) 2018-02-13 2019-02-03 CYCLIC COMPOUND MERGED WITH A PYRIMIDINE, ITS PREPARATION PROCESS AND ITS APPLICATION
KR1020207026395A KR102614939B1 (en) 2018-02-13 2019-02-03 Pyrimidine-fused ring compounds and their production methods and uses
EP19754599.9A EP3753941A4 (en) 2018-02-13 2019-02-03 Pyrimidine-fused cyclic compound, preparation method therefor and application thereof
PCT/CN2019/074685 WO2019158019A1 (en) 2018-02-13 2019-02-03 Pyrimidine-fused cyclic compound, preparation method therefor and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201810692211.4A CN110655520A (en) 2018-06-29 2018-06-29 Pyrimido-cyclic compounds, process for their preparation and their use

Publications (1)

Publication Number Publication Date
CN110655520A true CN110655520A (en) 2020-01-07

Family

ID=69026461

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810692211.4A Pending CN110655520A (en) 2018-02-13 2018-06-29 Pyrimido-cyclic compounds, process for their preparation and their use

Country Status (1)

Country Link
CN (1) CN110655520A (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111153901A (en) * 2018-11-07 2020-05-15 如东凌达生物医药科技有限公司 Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
WO2021249449A1 (en) * 2020-06-11 2021-12-16 Betta Pharmaceuticals Co., Ltd Shp2 inhibitors, compositions and uses thereof
CN115210232A (en) * 2020-01-22 2022-10-18 南京明德新药研发有限公司 Pyrazolo heteroaromatic ring compound and application thereof
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 (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010144881A1 (en) * 2009-06-12 2010-12-16 The General Hospital Corporation Treatment of meningeal and neural diseases
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
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
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

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010144881A1 (en) * 2009-06-12 2010-12-16 The General Hospital Corporation Treatment of meningeal and neural diseases
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
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
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

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CA: "CAS RN:1416713-07-9;CAS RN:1353100-74-9", 《REGISTRY》 *
CA: "CAS RN:196614-16-1;CAS RN:13035-19-3;CAS RN:761355-17-3;CAS RN:1321603-78-4;CAS RN:952480-24-9", 《REGISTRY》 *
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》 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111153901A (en) * 2018-11-07 2020-05-15 如东凌达生物医药科技有限公司 Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
CN115210232A (en) * 2020-01-22 2022-10-18 南京明德新药研发有限公司 Pyrazolo heteroaromatic ring compound and application thereof
CN115210232B (en) * 2020-01-22 2024-03-01 上海齐鲁制药研究中心有限公司 Pyrazolo heteroaromatic ring compound and application thereof
WO2021249449A1 (en) * 2020-06-11 2021-12-16 Betta Pharmaceuticals Co., Ltd Shp2 inhibitors, compositions and uses thereof
CN115515946A (en) * 2020-06-11 2022-12-23 贝达药业股份有限公司 SHP2 inhibitor and composition and application thereof
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

Similar Documents

Publication Publication Date Title
JP7335882B2 (en) Pyrimidine-condensed ring compound, method for producing the same, and use
CN110156786B (en) Pyrimido-cyclic compounds, process for their preparation and their use
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
EP3712151B1 (en) (s)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine as shp2 inhibitor for the treatment of cancer
EP3035800A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
CN105566321B (en) Heteroaromatic compounds and their use in medicine
CN110655520A (en) Pyrimido-cyclic compounds, process for their preparation and their use
CN111050765B (en) Spiro compounds and methods of making and using the same
EP4072679A1 (en) Antagonists of the muscarinic acetylcholine receptor m4
WO2016192630A1 (en) Compound having kinase inhibiting activity, method of preparing same, and use of same
CN111518100A (en) Cyclopropenoarylbenzofuran substituted nitrogen heteroaryl compound and application thereof
TW202208379A (en) New macrocyclic lrrk2 kinase inhibitors
CN109790160B (en) Pyrido five-membered aromatic ring compound, preparation method and application thereof
CN112778336B (en) Nitrogen-containing condensed ring STING regulator compound, preparation method and application
WO2022017434A1 (en) Compound having kinase inhibitory activity
CN114805361B (en) Amino substituted aromatic heterocyclic pyrazole compound, preparation method and application
CN111057048B (en) Aminopyrazine/pyridine compound, preparation method and application
CN110229151B (en) Indolizine compound, preparation method and application thereof
CN111763217B (en) Thieno-nitrogen heterocyclic compounds, preparation method and application
WO2023212147A1 (en) Heterocyclic compounds as modulators of bcl6 as ligand directed degraders
WO2023137634A1 (en) Tricyclic compound, preparation therefor, pharmaceutical composition and use
CN114478585A (en) Nitrogen-containing fused heterocyclic compound and preparation method and application thereof
CN115703799A (en) Nitrogen heteroaryl compound, preparation method and application thereof
CN114685487A (en) Pyrimidine heterocyclic compound, preparation method and application

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

Effective date of registration: 20210608

Address after: 201203 1st floor, building 3, 576 libing Road, Pudong New Area, Shanghai

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

Address before: 201203 Room 106, building 2, no.230 Cailun Road, No.86 Faraday Road, China (Shanghai) pilot Free Trade Zone, Pudong New Area, Shanghai

Applicant before: SHANGHAI BLUERAY BIOPHARMA Co.,Ltd.

TA01 Transfer of patent application right
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20200107

WD01 Invention patent application deemed withdrawn after publication