CN111875583B - Triazole derivative and preparation method and application thereof - Google Patents

Triazole derivative and preparation method and application thereof Download PDF

Info

Publication number
CN111875583B
CN111875583B CN202010800210.4A CN202010800210A CN111875583B CN 111875583 B CN111875583 B CN 111875583B CN 202010800210 A CN202010800210 A CN 202010800210A CN 111875583 B CN111875583 B CN 111875583B
Authority
CN
China
Prior art keywords
compound
cancer
membered
alkyl
pharmaceutically acceptable
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.)
Expired - Fee Related
Application number
CN202010800210.4A
Other languages
Chinese (zh)
Other versions
CN111875583A (en
Inventor
沈竞康
熊兵
陈丹琦
王昕�
张乃霞
黄敏
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Institute of Materia Medica of CAS
Original Assignee
Shanghai Institute of Materia Medica of CAS
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 Institute of Materia Medica of CAS filed Critical Shanghai Institute of Materia Medica of CAS
Priority to CN202010800210.4A priority Critical patent/CN111875583B/en
Publication of CN111875583A publication Critical patent/CN111875583A/en
Application granted granted Critical
Publication of CN111875583B publication Critical patent/CN111875583B/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • C07D249/101,2,4-Triazoles; Hydrogenated 1,2,4-triazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D249/12Oxygen or sulfur atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Abstract

The invention relates to a PDE inhibitor, in particular to a PDE delta inhibitor. In particular to a triazole derivative shown in a general formula (I) and a preparation method and application thereof. The series of compounds have good PDE delta inhibitory activity, are suitable for preparing medicaments for treating or preventing KRas-PDE delta mediated diseases, are particularly suitable for preparing KRas-PDE delta mediated antitumor medicaments, and have the same definition of each substituent in the general formula (I) as that in the specification.

Description

Triazole derivative and preparation method and application thereof
The application is a divisional application of a patent application with application number of 2017109648846, wherein the application is applied for 10 months and 17 days in 2017, and the name of the invention is triazole derivative and a preparation method and application thereof.
Technical Field
The invention relates to a PDE inhibitor, in particular to a triazole derivative and a preparation method and application thereof.
Background
Malignant tumors are a serious threat to human health and life, and are a major cause of morbidity and mortality worldwide. According to the published data of the world health organization, about 1400 million new cancer cases and 820 ten cancer death cases exist in the world in 2012, and in the next 20 years, the new cancer cases are predicted to rise from 1400 million in 2012 to 2400 million each year. The incidence rate of cancer in China is close to the world level, but the mortality rate is higher than the world level. The number of cancer diseases in the middle of 2015 is 429.2 ten thousand, which accounts for about one fifth of the global disease; the number of cancer deaths is 281.4 ten thousand, accounting for about one fourth of the cancer deaths worldwide in the year.
The generation and development of malignant tumor are closely related to the mutation and over-expression of oncogene, the inactivation and loss of cancer suppressor gene and the dysfunction of signal channel, and the like, and are oncogene-dependent molecular network diseases with multifactorial action, polygene participation, multistage development and multilevel relation. About 30% of tumors have been found to show the mutational activation of Ras gene and the overexpression of Ras protein. Ras proteins encoded by the Ras gene mediate mainly cell proliferation, growth, differentiation and apoptosis. When Ras signaling becomes very active, normal tissue cells are transformed into cancer cells, resulting in the development of tumors. Ras protein is involved in cell signaling, and is upstream of activated receptor tyrosine kinase (PTK), and downstream of it is mainly cascade amplification. The Ras-GTP active form forms a complex with downstream effector molecules such as Raf, PI3K and other effector factors through protein-protein interaction, activates downstream signal pathways, completes signal conduction and plays a biological function. Although the research and development of antitumor drugs targeting Ras has been carried out for nearly 20 years, no antitumor drug targeting Ras has been marketed.
PDE delta has been considered to be a catalytically inactive subunit of PDE 6. Recent research shows that after the Ras protein is subjected to farnesylation, three amino acid residues are removed by exonuclease and modification after methylation, the farnesyl binding protein PDE delta is combined with farnesyl on K-Ras4B, and then the K-Ras4B is transferred to the cell membrane for positioning. The K-Ras4B for completing the cell membrane localization is stimulated by various extracellular signal factors, and is combined with downstream effectors to complete signal transduction and regulate the proliferation and differentiation of cells. Based on the above findings, Anchal Chandra et al suggest that inhibition or degradation of PDE delta prevents the RAS protein from migrating to the cell membrane and that RAS protein can be prevented from exerting biological effects. Three classes of PDE δ inhibitors have been reported, being benzimidazoles, pyridazinopyrazoles and sulfonamides, respectively. The first PDE delta inhibition was Deltarasin reported by Gunther et al, McFarland molecular physiology research, Germany, 2013. The molecular activity of Deltarasin is IC5038nM, cell Activity IC505-20 μ M. Bjorn et al, McFlex molecular physiology research institute in 2016, Germany, reported a second PDE delta inhibitor Deltazinone. The molecular activity of the compound is IC508nM, but cell activity and Deltarasin were comparable to IC505-20 μ M. Pablo and the like of a Mackemann molecular physiology research institute in Germany in 2017 screen a compound library (200000) through an Alfa screening technology, find that one class of sulfonamide compounds have a good inhibitory effect on PDE delta, and find and report a third class of PDE delta inhibitor Deltasamide through structure optimization. IC of Deltasamide 1 therein50203pM, Deltasamide 2 IC50385pM, the cell activity was 0.75-1 μ M. The three inhibitors have good molecular activity, but poor cell activity, single molecular skeleton and poor drug forming property. Therefore, the development of PDE delta inhibitors with novel structure and better activity is needed.
Disclosure of Invention
Based on the above, the triazole derivative with better PDE delta inhibitory activity at a cellular level and the preparation method and the application thereof are provided.
Triazole derivatives with a structure shown as a formula (I) or pharmaceutically acceptable salts or stereoisomers thereof or prodrug molecules thereof:
Figure BDA0002627095020000011
wherein X is H, S or CH2
R1Is substituted or unsubstituted phenyl or substituted or unsubstituted benzyl;
R2selected from hydrogen atoms, substituted or unsubstituted C1-C6Alkyl or
Figure BDA0002627095020000021
Ring A is selected from 3-10 membered cycloalkyl, 4-10 membered heterocyclyl, 5-10 membered aryl or 5-10 membered heteroaryl; y is absent, or NH or O;
when Y is absent, R4Selected from hydrogen atom, halogen, hydroxyl, cyano, nitro, trifluoromethyl, C1-C4Alkyl, 3-10 membered cycloalkyl, -NH2、-NH(C1-C3Alkyl group), N (C)1-C3Alkyl radical)2Or C1-C4An alkoxy group;
when Y is NH or O, R4Selected from:
Figure BDA0002627095020000022
ring B is selected from 3-10 membered cycloalkyl, 4-10 membered heterocyclyl, 5-10 membered aryl or 5-10 membered heteroaryl;
R5or R6Each independently selected from hydrogen atom, C1-C4Alkyl radical, C1-C4Alkoxy, halogen, trifluoromethyl, cyano, nitro or hydroxy;
R7or R8Independently selected from hydrogen atom, C1-C4Alkyl, 3-to 10-membered cycloalkyl, 4-to 10-membered heterocyclyl, 5-to 10-membered aryl, 5-to 10-membered heteroaryl or
Figure BDA0002627095020000023
Said C is1-C4Each of alkyl, 3-10 membered cycloalkyl, 4-10 membered heterocyclyl, 5-10 membered aryl, 5-10 membered heteroaryl independently optionally further substituted with one or more substituents selected from C1-C4Alkyl, halogen, trifluoromethyl, hydroxy, cyano, nitro, 3-to 10-membered cycloalkyl, 4-to 10-membered heterocyclyl, 5-to 10-membered aryl, 5-to 10-membered heteroaryl, -NR11R12、-CONR11R12、-NR11C(O)R12、-OR11、-C(O)R12、-OC(O)R12OR-C (O) OR12Substituted with the substituent(s);
the R is7And R8Optionally together with the nitrogen atom to which it is attached, a 3-8 membered heterocyclyl containing 1-3 heteroatoms selected from N, O, S or a 5-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O and S;
when X is H, R3In the absence of the presence of the agent,
when X is S or CH2When R is3Is composed of
Figure BDA0002627095020000024
Ring C is selected from 5-10 membered aryl or 5-10 membered heteroaryl;
n is an integer of 1 to 3;
R13absent or selected from hydrogen atoms, C1-C4Alkyl, halogen, trifluoromethyl, cyano, nitro, hydroxy, 3-to 10-membered cycloalkyl, 4-to 10-membered heterocyclyl, 5-to 10-membered aryl, 5-to 10-membered heteroaryl, -NR11R12、-CONR11R12、-NR11C(O)R12、-OR11、-C(O)R12、-OC(O)R12OR-C (O) OR12Substituted with the substituent(s);
R11or R12Each independently selected from hydrogen atom, C1-C4Alkyl, 3-10 membered cycloalkyl, 5-10 membered aryl or 5-10 membered heteroaryl; said C is1-C4Alkyl, 3-10 membered cycloalkyl, 5-10 membered aryl or 5-10 membered heteroEach aryl group is independently further optionally substituted by one or more groups selected from halogen, hydroxy, cyano, nitro, trifluoromethyl, C1-C4Alkyl, 3-10 membered cycloalkyl or C1-C4Alkoxy radical,
Figure BDA0002627095020000025
Substituted with the substituent(s);
n is an integer of 0 to 4;
R9or R10Each independently selected from hydrogen atom or C1-C4An alkyl group;
the R is9And R10Optionally together with the nitrogen atom to which it is attached, form a 3-8 membered heterocyclic group containing 1-3 heteroatoms selected from N, O, S or a 5-10 membered heteroaryl group containing 1-3 heteroatoms selected from N, O and S.
In one embodiment, the compound is a compound of formula (II):
Figure BDA0002627095020000031
X、R1、R2and R13As defined above.
In one embodiment, the compound is a compound represented by the general formula (III)
Figure BDA0002627095020000032
Wherein R is1、R5、R6、R7And R8Is as defined above.
In one embodiment, X in the compound of formula (III) is S or CH2
R5Or R6Each independently selected from hydrogen atom, C1-C4An alkyl group;
R7or R8Independently selected from the group consisting of:
Figure BDA0002627095020000033
or R7And R8Together with the nitrogen atom to which they are attached, form the following group:
Figure BDA0002627095020000034
R14or R15Independently selected from: hydrogen atom, halogen, phenyl, trifluoromethyl, C1-C4Alkyl radical, C1-C3Alkoxy, pyrrolidinyl, morpholinyl, or-NR16R17、-CONR16R17、-C(O)R16or-COOR17
R16Or R17Each independently selected from hydrogen atom, C1-C4Alkyl or
Figure BDA0002627095020000035
Wherein n is an integer of 1 to 4; r18Or R19Independently selected from a hydrogen atom or C1-C4An alkyl group;
the R is16And R17Together with the nitrogen atom to which they are attached, form a 3-8 membered heterocyclic group containing 1-3 heteroatoms selected from N, O, S or a 5-10 membered heteroaryl group containing 1-3 heteroatoms selected from N, O and S.
In one embodiment, the compound is a compound represented by the general formula (IV)
Figure BDA0002627095020000041
R1Is phenyl or benzyl;
R14is-CONR16R17or-COOR17
R16、R17As defined above.
In one of themIn the examples, in the compounds represented by the general formula (IV), the R group16Or R17Each independently selected from the group consisting of:
Figure BDA0002627095020000042
in one embodiment, the compound is a compound represented by the general formula (V)
Figure BDA0002627095020000043
R2Is C1-C6An alkyl group;
R1、R11and R12As defined above.
In one embodiment, in the compound of formula (V), R1Is phenyl or benzyl;
R2is methyl, ethyl, propyl or 2-methylpropyl;
R11or R12Each independently selected from:
Figure BDA0002627095020000044
or R11And R12Together with the nitrogen atom to which they are attached, form the following group:
Figure BDA0002627095020000045
R14or R15As defined above.
In one embodiment, the compound is a compound represented by the general formula (VI)
Figure BDA0002627095020000051
R1Is phenyl or benzyl;
R14selected from halogen, phenyl, cyclohexyl or dimethylamino.
In one embodiment, the compound is selected from:
Figure BDA0002627095020000052
Figure BDA0002627095020000061
Figure BDA0002627095020000071
Figure BDA0002627095020000081
Figure BDA0002627095020000091
a pharmaceutical composition comprises a therapeutically effective amount of the triazole derivative or pharmaceutically acceptable salt or stereoisomer thereof or prodrug molecule thereof, and a pharmaceutically acceptable excipient.
The triazole derivative or a pharmaceutically acceptable salt or stereoisomer thereof or a prodrug molecule thereof according to any of the embodiments, or the use of the pharmaceutical composition in preparing a medicament for inhibiting PDE δ.
The triazole derivative or a pharmaceutically acceptable salt or stereoisomer thereof or a prodrug molecule thereof according to any embodiment, or the use of the pharmaceutical composition in preparing a medicament for treating or preventing KRas-PDE delta mediated diseases.
In one embodiment, the KRas-PDE mediated disease is a tumor.
In one embodiment, the tumor is squamous cell carcinoma, prostate cancer, renal cell carcinoma, kaposi's sarcoma, non-small cell lung cancer, lymphatic cancer, thyroid cancer, breast cancer, head and neck cancer, uterine cancer, esophageal cancer, melanoma, bladder cancer, genitourinary cancer, gastrointestinal cancer, glial cancer, colorectal cancer, or ovarian cancer.
The series of compounds have a brand new structure and have a larger difference with the structure of the traditional PDE delta inhibitor. The series of compounds have good PDE delta inhibitory activity, and can inhibit the PDE delta mediated positioning of KRAS on cell membranes as PDE delta inhibitors, thereby inhibiting the activation of downstream signaling pathways and generating biological activity. The application of the compound is suitable for preparing medicaments for treating or preventing KRas-PDE delta mediated diseases, and is particularly suitable for preparing KRas-PDE delta mediated antitumor medicaments.
Detailed Description
Definitions and general terms
Unless stated to the contrary, the following terms used in the specification and claims have the following meanings.
The term "optionally substituted with one or more substituents" in the present invention means substituted with one or more substituents, or unsubstituted. In particular, "optional" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not. For example, "a heterocyclic group optionally substituted with an alkyl" means that an alkyl may, but need not, be present, and the description includes the case where the heterocyclic group is substituted with an alkyl and the heterocyclic group is not substituted with an alkyl.
"alkyl" refers to a saturated aliphatic hydrocarbon group, including straight and branched chain groups. C1-C6Alkyl refers to an alkyl group containing 1 to 6 carbon atoms. Non-limiting examples include: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, tert-butyl, 2-dimethylpropyl, 1, 2-dimethylpropylCyclopropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl. C1-C4Alkyl refers to an alkyl group containing 1 to 4 carbon atoms. In one embodiment, C1-C4The alkyl group is methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl or sec-butyl. Alkyl groups may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment.
"cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbyl substituent. 3-10 membered cycloalkyl is meant to include 3 to 10 carbon atoms. In one embodiment, the 3-10 membered monocyclic cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like. Polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups. Cycloalkyl groups may be optionally substituted with one or more substituents.
"spirocycloalkyl" refers to polycyclic groups that share a single carbon atom (called a spiro atom) between single rings, which may contain one or more double bonds, but none of the rings have a completely conjugated pi-electron system. The spirocycloalkyl group is classified into a mono-spirocycloalkyl group, a di-spirocycloalkyl group or a multi-spirocycloalkyl group, preferably a mono-spirocycloalkyl group and a di-spirocycloalkyl group, according to the number of spiro atoms shared between rings. More preferably 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered.
"fused cyclic alkyl" refers to an all-carbon polycyclic group in which each ring in the system shares an adjacent pair of carbon atoms with other rings in the system, wherein one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system. They may be classified into bicyclic, tricyclic, tetracyclic or polycyclic fused ring alkyls according to the number of constituent rings, preferably bicyclic or tricyclic, more preferably 5-membered/5-membered, 5-membered/6-membered or 6-membered/6-membered bicycloalkyl.
"bridged cycloalkyl" groups in which any two rings share two all-carbon polycyclic groups of carbon atoms not directly attached, these may contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system. They may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl groups, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic, depending on the number of constituent rings.
The cycloalkyl ring of the above "cycloalkyl", "spirocycloalkyl", "fused ring alkyl", or "bridged cycloalkyl" groups may be fused to an aryl, heteroaryl, or heterocycloalkyl ring, where the rings attached to the parent structure are cycloalkyl, and in one embodiment, indanyl, tetrahydronaphthyl, and the like.
"Heterocyclyl" means a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent in which one or more ring atoms are selected from nitrogen, oxygen, or S (O)m(wherein m is an integer of 0 to 2), preferably a nitrogen or oxygen heteroatom; but not the ring moiety of-O-, -O-S-or-S-, the remaining ring atoms being carbon. 4-10 membered heterocyclyl is a ring containing 4 to 10 ring atoms, of which 1-3 are heteroatoms; preferably, the heterocyclyl ring contains 5 to 6 ring atoms of which 1-2 are heteroatoms. In one embodiment, the monocyclic heterocyclyl is dihydrofuranyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, or homopiperazinyl, and the like.
Polycyclic heterocyclic groups include spiro, fused and bridged heterocyclic groups. "Spiroheterocyclyl" polycyclic heterocyclic groups sharing one atom (referred to as a spiro atom) between monocyclic rings, wherein one or more ring atoms are selected from nitrogen, oxygen, or S (O)m(wherein m is an integer of 0 to 2) and the remaining ring atoms are carbon atoms. These may contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system.
"fused heterocyclyl" refers to polycyclic heterocyclic groups in which each ring in the system shares an adjacent pair of atoms with other rings in the system, and one or more rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system, wherein one or more ring atoms are selected from nitrogen, oxygen, or S (O)m(wherein m is an integer of 0 to 2) and the remaining ring atoms are carbon atoms.
"bridged heterocyclyl" means a 5 to 14 membered polycyclic heterocyclic group in which any two rings share two atoms which are not directly attached, which may contain one or more double bonds, but none of the rings have a completely conjugated pi-electron system in which one or more ring atoms are selected from nitrogen, oxygen or S (O)m(wherein m is an integer of 0 to 2) and the remaining ring atoms are carbon atoms.
The heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring to which the parent structure is attached is heterocyclyl, in one embodiment:
Figure BDA0002627095020000111
and the like.
The heterocyclyl group may be optionally substituted with one or more substituents.
"aryl" refers to an all-carbon monocyclic or fused polycyclic (i.e., rings which share adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 10 membered, more preferably phenyl and naphthyl, most preferably phenyl. The aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, and the aryl group may be substituted or unsubstituted.
A 5-10 membered "heteroaryl" refers to a heteroaromatic system containing 1 to 4 heteroatoms, 5 to 10 ring atoms, wherein the heteroatoms include oxygen, sulfur, and nitrogen. Heteroaryl is preferably 5-or 6-membered, for example furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl and the like. The heteroaryl ring may be fused to an aryl, heterocyclyl or cycloalkyl ring, the ring to which the parent structure is attached being a heteroaryl ring, in one embodiment:
Figure BDA0002627095020000112
Figure BDA0002627095020000113
and the like.
Heteroaryl groups may be optionally substituted or unsubstituted.
-NH(C1-C3Alkyl) means that an alkyl group containing 1 to 3 carbon atoms replaces one hydrogen on the amine group. In one embodiment, -NH (C)1-C3Alkyl) is:
Figure BDA0002627095020000114
-N(C1-C3alkyl radical)2Refers to the substitution of two hydrogens on the amine group with an alkyl group containing 1 to 3 carbon atoms. In one embodiment, -N (C)1-C3Alkyl radical)2Comprises the following steps:
Figure BDA0002627095020000115
C1-C4alkoxy means-O- (C)1-C4Alkyl) and-O- (unsubstituted C)1-C4Cycloalkyl groups). Wherein the alkyl and cycloalkyl groups are as defined above. In one embodiment, C1-C4Alkoxy is methoxy, ethoxy, propoxy, butoxy, cyclopropyloxy or cyclobutyloxy. Alkoxy groups may be optionally substituted or unsubstituted.
"hydroxy" refers to an-OH group.
"oxo" refers to ═ O, where the oxo group is typically, but not always, attached to a carbon atom (e.g., it may also be attached to a sulfur atom).
"halogen" means fluorine, chlorine, bromine or iodine.
"cyano" means-CN.
"nitro" means-NO2
"trifluoromethyl" means-CF3
The compounds of the present invention may exist in unsolvated forms as well as solvated forms containing pharmaceutically acceptable solvents such as water, ethanol, and the like, i.e., both solvated and unsolvated forms.
"pharmaceutical composition" means a mixture containing one or more compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, in admixture with other chemical components, as well as other components. Such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of the active ingredient and exert biological activity.
Excipients, which may be present in the composition, may be one or more buffers, stabilizers, anti-adherents, surfactants, wetting agents, lubricants, emulsifiers, binders, suspending agents, disintegrants, fillers, adsorbents, coating (enteric or slow release) preservatives, antioxidants, opacifying agents, glidants, processing aids, colorants, sweeteners, fragrances, flavoring agents and other known additives.
"pharmaceutically acceptable salt", i.e., "pharmaceutically acceptable salt", refers to an organic or inorganic salt of a pharmaceutically acceptable compound.
When the compound is acidic or includes sufficiently acidic bioisosteres, the appropriate "pharmaceutically acceptable salt" refers to a salt prepared from a pharmaceutically acceptable non-toxic base including inorganic and organic bases. The salts are derived from inorganic bases containing aluminum, ammonium, calcium, copper, iron, lithium, magnesium, manganese, potassium, sodium, octyl, and the like. Particular embodiments include ammonium, calcium, magnesium, potassium, and sodium salts. Salts are derived from pharmaceutically acceptable organic non-toxic bases including salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins such as arginine, betaine, caffeine, choline, N, N.sup.1-dibenzylethylenediamine, ethylenediamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, reduced glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, meglumine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
When the compound is basic or includes sufficiently basic bioisosteres, salts can be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, sulfuric, succinic, tartaric, p-toluenesulfonic acid and the like. Particular embodiments include citric acid, hydrobromic acid, hydrochloric acid, phosphoric acid, sulfuric acid, maleic acid, tartaric acid. Other exemplary salts include, but are not limited to, sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, sulfate, phosphate, acid phosphate, isonicotinic acid, lactic acid, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, fumarate, maleate, gentisate, gluconate, glucuronate, gluconate, formate, benzoate, glutamate, methylsulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate (e.g., 1, 1' -methylene-bis- (2-hydroxy-3-naphthoate)).
In addition, the pharmaceutical preparation containing the compound may be tablets, capsules, oral liquids, pills, granules, powders, ointments, patches, suppositories, buccal tablets, eye drops, eye ointments, ear drops, sprays, aerosols, inhalants, injections, and the like.
The term "therapeutically effective amount" refers to the amount of an effective compound or pharmaceutical agent that is the minimum amount necessary to ameliorate, cure or treat one or more symptoms of a disease or disorder.
In addition, the compounds and pharmaceutical compositions of the present invention may be administered alone or in combination with other agents. For combination therapy with more than one active agent, when the active agents are in separate dosage formulations, the active agents may be administered separately or in combination. In addition, administration of one agent may be performed before, simultaneously with, or after administration of another agent. When administered in combination with other agents, the "effective amount" of the second agent will depend on the type of drug used.
The compounds or pharmaceutical compositions of the present invention may also be included in a kit.
It should be noted that the reagent of the present invention is not specified as a specific source, and is a conventional reagent purchased in the market.
Explanation of abbreviations:
EDC 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride
DMF dimethyl formamide
DMAP 4-dimethylaminopyridine
DMSO dimethyl sulfoxide
CDCl3Deuterated chloroform
HATU 2- (7-benzotriazol oxide) -N, N, N ', N' -tetramethyluronium hexafluorophosphate
DIEA N, N-diisopropylethylamine
SEMCl 2- (trimethylsilyl) ethoxymethyl chloride
DIPEA N, N-diisopropylethylamine
HOBt 1-hydroxybenzotriazole
Synthesis of the Compounds of the invention
(1) Route one
Figure BDA0002627095020000131
R1、R2、R3As defined in claim 1;
the method comprises the following specific steps: refluxing the raw material (I-a) and hydrazine hydrate to obtain a compound (I-b), and condensing the compound (I-b) and phenyl isothiocyanate to form a ring so as to obtain a compound (I-c);
carrying out nucleophilic substitution reaction on the compound (I-c) to obtain a compound with a general formula (I); the compound (I-c) is subjected to sulfydryl removal by hydrogen peroxide to obtain R3A compound of the general formula (I) in which X is a hydrogen atom.
(2) Route two
Figure BDA0002627095020000132
The method comprises the following specific steps: reacting the compound (I-d) with methyl bromoacetate to obtain a compound (I-e), hydrolyzing the compound (I-e) by using an alkaline aqueous solution to obtain acid (I-f), and carrying out condensation reaction on the acid (I-f) and amine to obtain a compound (I-g);
wherein R is1、R3、R5、R6、R7Or R8As defined in claim 1;
(3) route three
Figure BDA0002627095020000141
The method comprises the following specific steps: cyclizing the compound (I-h) and p-nitrophenyl hydrazide (I-I) to obtain a compound (I-j); reducing the nitro group of the compound (I-j) by iron powder to obtain a compound (I-k); the compound (I-k) is reacted with an acid, an isocyanate, a sulfonyl chloride or the like to produce a compound (I-l).
(4) Route four
Figure BDA0002627095020000142
The method comprises the following specific steps: cyclizing the compound (III-a) and the p-hydroxybenzene hydrazide (III-b) protected by SEM to obtain a compound (III-c), removing the SEM from the compound (III-c) to obtain a compound (III-d), condensing the compound (III-d) and alpha-substituted methyl bromoacetate to obtain a compound (III-e), hydrolyzing the compound (III-e) by using an alkaline aqueous solution to obtain a compound (III-f), and carrying out condensation reaction on the compound (III-f) and various amines to obtain a compound (III-g);
wherein R is1、R5、R6、R7Or R8As defined in claim 3.
(5) Route five
Figure BDA0002627095020000143
The method comprises the following specific steps: carrying out nucleophilic substitution reaction on the compound (V-a) and 4- (bromomethyl) ethyl benzoate to obtain a compound (V-b), hydrolyzing the compound (V-b) into a compound (V-c) by using an alkaline aqueous solution, and carrying out condensation reaction on the compound (V-c) and various amines to obtain a compound with a general formula (V);
wherein R is1、R2、R11Or R12As defined in claim 6;
(6) route six
Figure BDA0002627095020000151
The method comprises the following specific steps: condensing the compound (V-c) and 5-aminopyridine-3-carboxylic acid methyl ester to obtain a compound (V-d), hydrolyzing the compound (V-d) by using an alkaline aqueous solution to generate a compound (V-e), and then carrying out condensation reaction on the compound (V-e) and various amines to obtain a compound (V-f);
wherein R is1、R2、R15Or R16As defined in claim 7.
The present invention will be described below by referring to specific compounds.
Synthesis of 4-benzyl-3-benzylmercapto-5-isobutyl-4H-1, 2, 4-triazole (Compound 1)
Figure BDA0002627095020000152
Step 1: synthesis of 3-methylbutyrylhydrazine (1-2)
Figure BDA0002627095020000153
Methyl isovalerate 1-1(11.4ml, 0.08mmol) was added portionwise to a solution of hydrazine monohydrate (10.7ml, 0.34mmol) in ethanol (50ml), heated under reflux under argon overnight. The solvent was evaporated under reduced pressure and taken up three times with toluene. And (5) storing under the protection of argon.
1H NMR(400MHz,DMSO-d6)δ8.91(s,1H),4.21(s,2H),1.97(m,1H),1.91–1.86(m,2H),0.86(d,J=6.5Hz,6H).+ESI:117[M+H]+
Step 2: synthesis of 4-benzyl-5-isobutyl-4H-1, 2, 4-triazole-3-mercapto (1-3)
Figure BDA0002627095020000154
Intermediate 1-2(0.58g, 0.5mmol) and benzyl isothiocyanate (0.745g, 0.5mmol) were dissolved in ethanol (8ml), heated under reflux for 2 hours to form a large amount of white solid, which was further dissolved after further heating, and potassium carbonate (0.69g, 0.5mmol) was added and further refluxed for 1 hour. After completion of the reaction, the reaction mixture was neutralized with a 1N hydrochloric acid solution, filtered, washed with water and then with dichloromethane, and dried to obtain 1 to 3(850mg, yield 69%) as a white solid.
1H NMR(400MHz,CDCl3)δ12.14(s,1H),7.44–7.23(m,5H),5.30(s,2H),2.38(d,J=7.2Hz,2H),1.97(dt,J=13.6,6.8Hz,1H),0.91(d,J=6.6Hz,6H).+ESI:248[M+H]+
And step 3: synthesis of 4-benzyl-3-benzylmercapto-5-isobutyl-4H-1, 2, 4-triazole (Compound 1)
Figure BDA0002627095020000161
Intermediate 1-3(50mg, 0.2mmol), bromobenzyl (24ul, 0.2mmol) and cesium carbonate (73mg, 0.22mmol) were dissolved in acetonitrile (2ml) and stirred at room temperature overnight. After completion of the reaction, water was added, followed by extraction with ethyl acetate, washing with saturated brine, drying over anhydrous sodium sulfate, evaporation to dryness, and purification by column chromatography (dichloromethane: methanol: 100:4) to give viscous compound 1(57mg, yield 86%).
1H NMR(400MHz,CDCl3)δ7.35–7.21(m,8H),6.89(dd,J=6.7,2.8Hz,2H),4.80(s,2H),4.34(s,2H),2.46(d,J=7.3Hz,2H),2.03(m,1H),0.93(d,J=6.7Hz,6H).+ESI:338[M+H]+
Compounds 2-7 were synthesized using the same synthetic procedure as Compound 1.
Figure BDA0002627095020000162
Synthesis of 4-benzyl-3-isobutyl-4H-1, 2, 4-triazole (Compound 8)
Figure BDA0002627095020000171
Dissolving intermediate 1-3(400mg, 1.6mmol) in dichloromethane (2.5ml), adding acetic acid (0.12ml), heating to 35 deg.C, and adding dropwise H2O2After the solution is added, the temperature is reduced to room temperature and the solution is stirred for 0.5 hour. After the reaction is finished, adjusting the pH of the reaction solution to be more than 10 by using 10% sodium hydroxide, extracting by using ethyl acetate, washing by using saturated salt water, drying by using anhydrous sodium sulfate, concentrating under reduced pressure, separating and purifying by using column chromatography, and eluting by using dichloromethane: methanol (15: 1) gave compound 8(248mg, yield 71%).
1H NMR(400MHz,CDCl3)δ8.07(s,1H),7.44–7.33(m,3H),7.19–6.98(m,2H),5.10(s,2H),2.58(d,J=7.3Hz,2H),2.13(dt,J=13.6,6.8Hz,1H),0.97(dd,J=6.6,0.6Hz,6H).+ESI:216[M+H]+
Synthesis of 4- (((4-benzyl-5-isobutyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) -N-phenylbenzamide (Compound 9)
Figure BDA0002627095020000172
Step 1: synthesis of ethyl 4- (((4-benzyl-5-isobutyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) benzoate (Compound 9-1)
Figure BDA0002627095020000173
Intermediate 1-3(0.12g, 0.48mmol), ethyl 4- (bromomethyl) benzoate (0.118g, 0.48mmol) and cesium carbonate (0.174g, 0.53mmol) were dissolved in acetonitrile (3ml) and stirred at room temperature for 48 hours. Water was added, followed by extraction with ethyl acetate, washing with saturated brine, drying over anhydrous sodium sulfate, and evaporation to dryness to give colorless oily compound 9-1(200mg, yield 82%).
Step 2: synthesis of 4- (((4-benzyl-5-isobutyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) benzoic acid (Compound 9-2)
Figure BDA0002627095020000174
Intermediate 9-1(0.2g, 0.49mmol) was dissolved in a mixed solvent of methanol and water (0.7ml, 3:1), and lithium hydroxide (0.15g, 0.36mmol) was added thereto, followed by heating to 60 ℃ for 5 hours and further stirring at room temperature overnight. Water dilution, then ethyl acetate extraction 2 times, aqueous phase with 1NHCl to adjust pH to 5-6, extraction, evaporation to dryness to obtain light yellow solid of compound 9-2(0.16g, yield 89%).
And step 3: synthesis of 4- (((4-benzyl-5-isobutyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) -N-phenylbenzamide (Compound 9)
Figure BDA0002627095020000181
Intermediate 9-2(40mg, 0.1mmol), aniline (19ul, 0.2mmol) and EDC (19.1mg, 0.1mmol) were dissolved in DMF (1ml), DMAP (1.2mg, 0.01mmol) was added with stirring and stirred at room temperature overnight. After completion of the reaction, the solvent was evaporated to dryness and isolated and purified on a thick plate to obtain compound 9(38mg, yield 84%) as a solid.
1H NMR(400MHz,DMSO-d6)δ10.45(s,1H),δ7.87(d,J=8.0Hz,2H),7.77(d,J=8.1Hz,2H),7.43(d,J=8.1Hz,2H),7.40–7.24(m,5H),7.11(t,J=7.5Hz,1H),6.95(d,J=7.1Hz,2H),5.06(s,2H),4.39(s,2H),2.48(d,J=7.0Hz,2H),1.87(dt,J=13.5,6.8Hz,1H),0.84(d,J=6.6Hz,6H).+ESI:457[M+H]+
Compounds 10-25 were synthesized using the same synthetic procedure as for compound 9
Figure BDA0002627095020000182
Figure BDA0002627095020000191
Figure BDA0002627095020000201
Synthesis of 5- (((4-benzyl-5-isobutyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) benzamido) -N- (2- (dimethylamino) ethyl) nicotinamide (Compound 26)
Figure BDA0002627095020000202
Step 1: synthesis of methyl 5- (((4-benzyl-5-isobutyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) benzamido) nicotinate (Compound 26-1)
Figure BDA0002627095020000203
Intermediate 9-2(200mg, 0.5mmol) and methyl 5-aminopyridine-3-carboxylate (80mg, 0.5mmol) and EDC (100mg, 0.5mmol) were dissolved in DMF (4.5ml) and DMAP (6.4mg, 0.05mmol) was added with stirring and stirred at room temperature overnight. Adding water, extracting with ethyl acetate, washing with saturated salt water, drying with anhydrous sodium sulfate, separating and purifying by column chromatography to obtain oily compound 26-1, and directly reacting in the next step.
1H NMR(400MHz,DMSO-d6)δ10.51(s,1H),9.05(s,1H),8.78(s,1H),8.64(s,1H),7.94(d,J=8.3Hz,2H),7.45(d,J=8.2Hz,2H),7.37–7.24(m,3H),7.03–6.86(m,2H),5.05(s,2H),4.39(s,2H),4.30(s,3H),2.48(d,J=7.2Hz,2H),1.96–1.82(m,1H),0.83(d,J=6.6Hz,6H).+ESI:516[M+H]+
Step 2: synthesis of 5- (((4-benzyl-5-isobutyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) benzamido) nicotinic acid (Compound 26-2)
Figure BDA0002627095020000204
Intermediate 26-1 was dissolved in a mixed solvent (4ml, MeOH: H)2To O ═ 3:1), lithium hydroxide (0.8g) was added, and the mixture was heated to 60 ℃ to react for 5 hours, and then stirred at room temperature overnight. Adjusting pH to 5-6 with 1N HCl, extracting, evaporating to dryness, separating and purifying by column chromatography to obtain yellow solid compound 26-2(0.16g, yield 85%).
1H NMR(400MHz,DMSO-d6)δ10.54(s,1H),9.05(s,1H),8.78(s,1H),8.64(s,1H),7.94(d,J=8.3Hz,2H),7.45(d,J=8.2Hz,2H),7.37–7.24(m,3H),7.03–6.86(m,2H),5.05(s,2H),4.39(s,2H),2.48(d,J=7.2Hz,2H),1.96–1.82(m,1H),0.83(d,J=6.6Hz,6H).+ESI:502[M+H]+
5- (((4-benzyl-5-isobutyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) benzamido) -N- (2- (dimethylamino) ethyl) nicotinamide (Compound 26)
Figure BDA0002627095020000211
Compound 26-2(40mg, 0.08mmol), N-dimethylethylenediamine (7mg, 0.08mmol) and HATU (32mg, 0.08mmol) were dissolved in DMF (1ml), and DIEA (60. mu.l, 0.4mmol) was added thereto with stirring and stirred at room temperature overnight. Water was added thereto, followed by extraction with ethyl acetate, washing with saturated brine, drying over anhydrous sodium sulfate, and purification by column chromatography to give compound 26 as an oil (24mg, yield 53%).
1H NMR(400MHz,DMSO-d6)δ10.65(s,1H),9.07(d,J=2.4Hz,1H),8.97(d,J=5.5Hz,1H),8.83(d,J=2.0Hz,1H),8.67(t,J=2.2Hz,1H),8.02–7.87(m,2H),7.52–7.42(m,2H),7.39–7.23(m,3H),7.01–6.90(m,2H),5.07(s,2H),4.40(s,2H),3.19–2.98(m,4H),2.72(s,6H),2.48(d,J=7.2Hz,2H),1.94–1.82(m,1H),0.84(d,J=6.6Hz,6H).+ESI:572[M+H]+
Compounds 27-30 were prepared using the same route and synthesis as for compound 9.
Figure BDA0002627095020000212
Synthesis of N-cyclohexyl-4- (((5-isobutyl-4-phenyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) benzamide (Compound 31)
Figure BDA0002627095020000213
Step 1: synthesis of 5-isobutyl-4-phenyl-3-mercapto-4H-1, 2, 4-triazole (Compound 31-1)
Figure BDA0002627095020000221
Dissolving intermediate 1-2(0.58g, 5.0mmol) and phenyl isothiocyanate (0.596g, 5.0mmol) in ethanol (10ml), heating and refluxing for 2 hours, cooling, filtering, drying the solid, adding the dried solid into 1N NaOH, and refluxing for 2 hours. After completion of the reaction, the reaction mixture was neutralized with a 1N hydrochloric acid solution, filtered, washed with water, and dried to obtain compound 31-1 as a white solid (0.83g, yield 71%).
1H NMR(400MHz,CDCl3)δ11.60–11.50(m,1H),7.63–7.54(m,3H),7.38–7.30(m,2H),2.39(d,J=7.2Hz,2H),1.90(m,1H),0.90(d,J=6.6Hz,6H).+ESI:234[M+H]+
Step 2: synthesis of ethyl 4- (((5-isobutyl-4-phenyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) benzoate (Compound 31-2)
Figure BDA0002627095020000222
Intermediate 31-1(0.4g, 1.8mmol), ethyl 4- (bromomethyl) benzoate (0.444g, 1.8mmol) and cesium carbonate (0.596g, 1.98mmol) were dissolved in acetonitrile (5ml) and stirred at room temperature for 12 hours. Adding water, extracting with ethyl acetate, washing with saturated salt water, drying with anhydrous sodium sulfate, evaporating to dryness, and directly reacting in the next step.
And step 3: synthesis of 4- (((5-isobutyl-4-phenyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) benzoic acid (Compound 31-3)
Figure BDA0002627095020000223
Intermediate 31-2 was dissolved in a mixed solvent of methanol and water (5ml, 3:1), and lithium hydroxide (1.0g) was added thereto, and the mixture was heated to 60 ℃ to react for 5 hours and then stirred at room temperature overnight. Diluting with water, extracting with ethyl acetate for 2 times, adjusting pH of the water phase to 3 with 1N HCl, extracting, and evaporating to obtain light yellow solid 31-3.
And 4, step 4: synthesis of N-cyclohexyl-4- (((5-isobutyl-4-phenyl-4H-1, 2, 4-triazol-3-yl) mercapto) methyl) benzamide (Compound 31)
Figure BDA0002627095020000224
Intermediate 31-3(46mg, 0.13mmol), cyclohexylamine (24mg, 0.13mmol) and EDC (25mg, 0.5mmol) were dissolved in DMF (1ml), DMAP (1.6mg, 0.013mmol) was added with stirring and stirred at room temperature overnight. Water was added thereto, followed by extraction with ethyl acetate, washing with saturated brine, drying over anhydrous sodium sulfate, and purification by column chromatography (dichloromethane: methanol: 100:4) to give compound 31 as a solid (46mg, yield 79%).
1H NMR(400MHz,DMSO-d6)δ8.15(d,J=7.9Hz,1H),7.79–7.71(m,2H),7.58-7,54(m,3H),7.39–7.31(m,2H),7.31–7.23(m,2H),4.33(s,2H),2.41(d,J=7.2Hz,2H),1.87–1.69(m,4H),1.61(d,J=13.1Hz,1H),1.44–1.20(m,6H),1.18–1.06(m,1H)0.79(d,J=6.6Hz,6H).+ESI:449[M+H]+
Synthesis of 3-isobutyl-4-phenyl-4H-1, 2, 4-triazole (Compound 32)
Figure BDA0002627095020000231
Dissolving intermediate 31-1(100mg, 0.43mmol) in dichloromethane (6ml), adding acetic acid (0.7ml), heating to 35 deg.C, and adding dropwise H2O2(0.12ml), after the dropwise addition, the temperature was lowered to room temperature and the mixture was stirred for 0.5 hour. For reactionsWashing with 1N sodium hydroxide, extraction with ethyl acetate, washing with saturated brine, drying over anhydrous sodium sulfate, concentration under reduced pressure, and separation and purification on a thick plate yielded compound 32 as a solid (40mg, yield 47%).1H NMR(400MHz,Chloroform-d)δ8.21(s,1H),7.62–7.52(m,3H),7.33–7.25(m,2H),2.63(d,J=7.3Hz,2H),2.06(m,1H),0.91(d,J=6.6Hz,6H).+ESI:202[M+H]+
Synthesis of 2- (4- (4-benzyl-5- ((4-methoxybenzyl) mercapto) -4H-1,2, 4-triazol-3-yl) phenoxy) -N- (4-fluorophenyl) acetamide (Compound 33)
Figure BDA0002627095020000232
Step 1: synthesis of methyl 4- ((2- (trimethylsilyl) ethoxy) methoxy) benzoate (Compound 33-2)
Figure BDA0002627095020000233
Methyl paraben 33-1(2.28g, 15mmol), SEMCl (3.89ml, 22mmol) and cesium carbonate (7.16g, 22mmol) were mixed in acetonitrile (45ml) and stirred at room temperature overnight. Acetonitrile was distilled off, and the residue was dissolved in ethyl acetate, washed with water three times, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated and separated and purified by column chromatography (petroleum ether: ethyl acetate (95:5)) to give 33-2(3.258g, yield 77%) as a colorless oily compound.
1H NMR(400MHz,CDCl3)δ8.05–7.94(m,2H),7.11–7.03(m,2H),5.29(s,2H),3.90(s,3H),3.86–3.66(m,2H),1.07–0.74(m,2H).0.01(s,9H).+ESI:283[M+H]+
Step 2: synthesis of 4- ((2- (trimethylsilyl) ethoxy) methoxy) benzoyl hydrazine (compound 33-3)
Figure BDA0002627095020000241
Compound 33-2(3.258g, 12mmol) was dissolved in methanol (15ml), hydrazine monohydrate (2.89g, 90mmol) was added, refluxed overnight, and after completion of the reaction, distilled under reduced pressure and taken up with toluene to give compound 33-3(1.94g) as an oil which solidified upon standing. The reaction mixture is directly put into the next step without purification.
And step 3: synthesis of 4-benzyl-5- (4- ((2- (trimethoxy silicon) ethoxy) methoxy) phenyl) -4H-1,2, 4-triazole-3-mercapto group (compound 33-4)
Figure BDA0002627095020000242
Compound 33-3(1.94g, 7mmol) and benzyl isothiocyanate (0.9ml, 7mmol) were dissolved in absolute ethanol (15ml), and 5.13ml of an ethanol solution of 21% sodium ethoxide was added thereto, followed by reflux overnight. After the reaction was completed, most of the solvent was distilled off, water was added thereto for ice bath, and neutralization was carried out with 1N HCI to precipitate a solid. Filtration, washing of the solid with water, and drying gave compound 33-4(1.484g, yield 51%) as a pale yellow solid.
And 4, step 4: synthesis of 4-benzyl-3- ((4-methoxybenzyl) mercapto) -5- (4- ((2- (trimethoxy silicon) ethoxy) methoxy) phenyl) -4H-1,2, 4-triazole (Compound 33-5)
Figure BDA0002627095020000243
Compound 33-4(1.484g, 4.4mmol), p-methoxybenzyl bromide (0.885g, 4.4mmol) and cesium carbonate (1.579g, 4.85mmol) were mixed in 30ml of acetonitrile, stirred at room temperature overnight, the acetonitrile was evaporated, the residue was dissolved in ethyl acetate, washed with water, dried, and subjected to column chromatography for concentration to give compound 33-5(1.15g, yield 49%) as a beige solid.
1H NMR(400MHz,CDCl3)δ7.46–7.38(m,2H),7.33–7.27(m,3H),7.27–7.22(m,2H),7.13–7.04(m,2H),6.96–6.88(m,2H),6.88–6.77(m,2H),5.25(s,2H),5.00(s,2H),4.40(s,2H),3.86–3.67(m,5H),1.07–0.87(m,2H),0.03-0.01(m,9H).+ESI:534[M+H]+
And 5: synthesis of 4- (4-benzyl-5- ((4-methoxybenzyl) mercapto) -4H-1,2, 4-triazol-3-yl) phenol (Compound 33-6)
Figure BDA0002627095020000244
Compound 33-5(900mg, 1.69mmol) was dissolved in 120ml dichloromethane: adding 4mL of dioxane solution of HCI into a mixed solvent of ethanol (1: 3), standing overnight, adding 4mL of dioxane solution of HCI again, standing overnight, adding a small amount of raw material, adding 4mL of dioxane solution of HCI again, reacting completely, evaporating reaction liquid, extracting with ethyl acetate and water, and purifying by a column to obtain a solid compound 33-6(650mg, yield 96%).
1H NMR(400MHz,CDCl3)δ7.46–7.38(m,2H),7.33–7.27(m,3H),7.27–7.22(m,2H),7.13–7.04(m,2H),6.96–6.88(m,2H),6.88–6.77(m,2H),5.25(s,2H),5.00(s,2H),3.89(s,3H).+ESI:404[M+H]+
Step 6: synthesis of methyl 2- (4- (4-benzyl-5- ((4-methoxybenzyl) mercapto) -4H-1,2, 4-triazol-3-yl) phenoxy) acetate (Compound 33-7)
Figure BDA0002627095020000251
Compound 33-6(285mg, 0.707mmol), methyl bromoacetate (98ul, 0.707mmol) and potassium carbonate (147mg, 1.065mmol) were mixed in DMF (4ml) and heated at 50 ℃ overnight. Extracting with ethyl acetate/water, and purifying with column. To give 33-7(318mg, yield 95%) as a yellow viscous compound.
1H NMR(400MHz,CDCl3)δ7.48–7.35(m,2H),7.31–7.25(m,3H),7.23–7.19(m,2H),7.11–7.02(m,2H),6.86–6.84(m,2H),6.82–6.75(m,2H),5.10(s,2H),4.86(s,2H),4.57(s,2H),3.89(s,3H),3.79(s,3H).+ESI:476[M+H]+
And 7: synthesis of 2- (4- (4-benzyl-5- ((4-methoxybenzyl) mercapto) -4H-1,2, 4-triazol-3-yl) phenoxy) acetic acid (Compound 33-8)
Figure BDA0002627095020000252
Dissolving compound 33-7(318mg, 0.669mmol) in 7ml ethanol, adding 1N NaOH, stirring overnight at room temperature, diluting with water, adjusting pH of the reaction solution to 4 with 1N HCl, extracting with ethyl acetate, evaporating to dryness, purifying with column chromatography, dichloromethane: methanol (15: 1) gave compound 33-8 as a white solid (120mg, yield 39%).
1H NMR(400MHz,CDCl3)δ7.46–7.38(m,2H),7.33–7.27(m,3H),7.27–7.22(m,2H),7.13–7.04(m,2H),6.96–6.88(m,2H),6.88–6.77(m,2H),5.10(s,2H),4.86(s,2H),4.57(s,2H),3.89(s,3H).+ESI:462[M+H]+
And 8: synthesis of 2- (4- (4-benzyl-5- ((4-methoxybenzyl) mercapto) -4H-1,2, 4-triazol-3-yl) phenoxy) -N- (4-fluorophenyl) acetamide (Compound 33)
Figure BDA0002627095020000253
Compound 33-8(50mg, 0.108mmol), 4-fluoroaniline (11.99mg, 0.108mmol), EDC (33mg, 0.172mmol) and HOBT (23mg,0.172mmol) were dissolved in DMF (1Ml), and DIPEA (72. mu.M, 0.384mmol) was added with stirring and reacted at room temperature overnight. Extraction with ethyl acetate, washing of the organic phase with brine, drying over anhydrous sodium sulfate, and purification by thin layer chromatography gave compound 33 as a pale yellow solid (30mg, yield 50%).
1H NMR(400MHz,DMSO-d6)δ10.19(s,1H),7.77–7.54(m,2H),7.49(d,J=9.2Hz,2H),7.31–7.21(m,5H),7.21–7.14(m,2H),7.13–7.06(m,2H),6.89-6.85(m,4H),5.14(s,2H),4.76(s,2H),4.33(s,2H),3.73(s,3H).+ESI:555[M+H]+
Compounds 34-60 were prepared using the same route and synthesis as compound 33.
Figure BDA0002627095020000261
Figure BDA0002627095020000271
Figure BDA0002627095020000281
Figure BDA0002627095020000291
Synthesis of 2- (4- (4-benzyl-5- ((4-methoxybenzyl) mercapto) -4H-1,2, 4-triazol-3-yl) phenoxy) -N- (5- (morpholine-4-carbonyl) pyridin-3-yl) acetamide (Compound 61)
Figure BDA0002627095020000292
Step 1: synthesis of 5- (2- (4- (4-benzyl-5- ((4-methoxybenzyl) mercapto) -4H-1,2, 4-triazol-3-yl) phenoxy) acetamide) nicotinic acid (Compound 61-1)
Figure BDA0002627095020000293
Compound 39(10mg) was dissolved in 1mL of ethanol, 1mL of 1N sodium hydroxide was added, and the reaction was carried out at room temperature for 30 min. 5mL of water was added, the pH was adjusted to 5-6 with 1N HCl, and the mixture was filtered under suction to give 61-1(8mg, yield 82%) as a yellow solid.
1H NMR(400MHz,DMSO-d6)δ11.12(s,1H),9.11(d,J=2.4Hz,1H),8.83(d,J=1.7Hz,1H),8.76(t,J=2.1Hz,1H),7.53(dd,J=12.9,8.7Hz,2H),7.32–7.20(m,5H),7.19–7.10(m,2H),6.91(dd,J=7.1,2.4Hz,2H),6.88–6.79(m,2H),5.18(s,2H),4.90(s,2H),4.37(s,2H),3.71(s,3H).
Step 2: synthesis of 2- (4- (4-benzyl-5- ((4-methoxybenzyl) mercapto) -4H-1,2, 4-triazol-3-yl) phenoxy) -N- (5- (morpholine-4-carbonyl) pyridin-3-yl) acetamide (Compound 61)
Figure BDA0002627095020000294
Compound 61-1(35mg), morpholine (7.8mg), HOBt (13.2mg) and EDC (18.5mg) were dissolved in 1mL of DMF, and DIEA 30. mu.L was added thereto, followed by stirring at room temperature overnight. The reaction mixture was diluted with water, extracted 3 times with ethyl acetate, and the organic phases were combined and washed with brine. The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified using a preparative plate (dichloromethane: methanol 15: 1) to give compound 61 as a brown solid (10mg, 25% yield).
1H NMR(400MHz,CDCl3)δ9.17(s,1H),8.78(d,J=2.5Hz,1H),8.43(d,J=1.9Hz,1H),8.34(t,J=2.1Hz,1H),7.45–7.36(m,2H),7.28(dd,J=3.1,1.5Hz,2H),7.26–7.17(m,2H),7.01–6.92(m,2H),6.93–6.84(m,2H),6.85–6.76(m,2H),4.99(s,2H),4.66(s,2H),4.37(s,2H),3.78(brs,4H),3.66(brs,2H),3.52–3.45(m,2H).
Compounds 62-65 were prepared using the same route and synthesis as compound 61.
Figure BDA0002627095020000301
Synthesis of methyl 5- (2- (4- (4-benzyl-5- (4-methoxyphenylethyl) -4H-1,2, 4-triazol-3-yl) phenoxy) acetamido) nicotinate (Compound 66)
Figure BDA0002627095020000302
Step 1: synthesis of N-benzyl-3- (4-methoxyphenyl) propionamide (Compound 66-2)
Figure BDA0002627095020000311
66-1(500mg, 2.78mmol) of 3- (4-methoxyphenyl) propionic acid, benzylamine (300mg, 2.78mmol), EDC (848mg, 4.44mmol) and HOBT (594mg,4.44mmol) were dissolved in DMF (10ml), and DIPEA (3ml, 7.84mmol) was added with stirring and reacted at room temperature overnight. Extraction with ethyl acetate, washing of the organic phase with brine, drying over anhydrous sodium sulfate, and purification by thin layer chromatography gave compound 66-2 as a white solid (650mg, yield 83%).
1H NMR(400MHz,CDCl3)δ7.44–7.25(m,3H),7.23–7.06(m,4H),6.98–6.69(m,2H),4.42(d,J=5.7Hz,2H),3.81(s,3H),2.96(t,J=7.5Hz,2H),2.51(t,J=7.5Hz,2H).+ESI:270[M+H]+
Step 2: synthesis of N-benzyl-3- (4-methoxyphenyl) propanethioamide (compound 66-3)
Figure BDA0002627095020000312
Compound 66-2(1.425g, 5.3mmol) and Lawson's reagent (1.26g, 2.7mmol) were dissolved in toluene (10ml) and stirred overnight at 110 ℃. After completion of the reaction, the solvent was evaporated in a fume hood, ethyl acetate and water were added for extraction, the organic phase was washed with brine, dried over anhydrous sodium sulfate, and separated and purified by thin layer chromatography to obtain 66-3(700mg, yield 47%) as a pale yellow solid.
1H NMR(400MHz,CDCl3)δ7.44–7.06(m,7H),6.94–6.59(m,2H),4.73(d,J=5.1Hz,2H),3.80(s,3H),3.10(t,J=7.2Hz,2H),2.95(t,J=7.2Hz,2H).+ESI:286[M+H]+
And step 3: synthesis of 4-benzyl-3- (4-methoxyphenylethyl) -5- (4- ((2- (trimethylsilyl) ethoxy) methoxy) phenyl) 4H-1,2, 4-triazole (Compound 66-4)
Figure BDA0002627095020000313
Compound 33-3(791.61mg, 2.8mmol) and compound 66-3(1.09g, 3.5mmol) were dissolved in dry tetrahydrofuran (8ml), acetic acid (2.40ml) was added with stirring, stirred for ten minutes in ice bath, mercury acetate (800mg, 3.07mmol) was added, and reacted in ice bath for two hours at room temperature overnight. Celite filtration, concentration of the filtrate, extraction of the residue with ethyl acetate and water, washing of the organic phase with saturated brine, drying over anhydrous sodium sulfate, concentration, and purification by column chromatography gave compound 66-4 as a pale yellow solid (578mg, yield 55%).
1H NMR(400MHz,CDCl3)δ7.45–7.39(m,2H),7.38–7.31(m,3H),7.10–7.05(m,2H),7.01(dd,J=8.7,2.4Hz,2H),6.91(dd,J=7.3,2.1Hz,2H),6.84–6.78(m,2H),5.25(s,2H),4.93(s,2H),3.80(d,J=0.5Hz,3H),3.78–3.73(m,2H),3.03(dd,J=9.3,6.4Hz,2H),2.86(dd,J=9.0,6.6Hz,2H),0.98–0.92(m,2H),0.02–0.00(s,9H).+ESI:516[M+H]+
And 4, step 4: synthesis of 4- (4-benzyl-5- (4-methoxyphenylethyl) -4H-1,2, 4-triazol-3-yl) phenol (Compound 66-5)
Figure BDA0002627095020000321
Compound 66-4(378mg, 0.73mmol) was dissolved in 12ml dichloromethane: to a mixed solvent of ethanol (1: 3), 3ml of HCl in dioxane was added, and the reaction was completed overnight. The reaction mixture was evaporated to dryness, extracted with ethyl acetate and water, and purified by column chromatography to obtain compound 66-5(271mg, yield 96%) as a solid.
And 5: synthesis of methyl 2- (4- (4-benzyl-5- (4-methoxyphenylethyl) -4H-1,2, 4-triazol-3-yl) phenoxy) acetate (Compound 66-6)
Figure BDA0002627095020000322
Compound 66-5(271mg, 0.7mmol), methyl bromoacetate (66ul, 0.7mmol) and potassium carbonate (193mg, 2.5mmol) were mixed in DMF (8ml) and heated at 50 ℃ overnight. Extracting with ethyl acetate/water, and purifying with column. To give 66-6(304mg, yield 95%) as a yellow viscous compound.
1H NMR(400MHz,DMSO-d6)δ7.61–7.54(m,2H),7.33-7.30(m,3H),7.11–7.06(m,2H),7.03–6.98(m,2H),6.92(d,J=7.3Hz,2H),6.85–6.80(m,2H),5.23(s,2H),4.72(s,2H),3.82(s,3H),3.71(s,3H),2.88(d,J=6.4Hz,2H),2.86–2.81(m,2H).+ESI:458[M+H]+
Step 6: synthesis of methyl 2- (4- (4-benzyl-5- (4-methoxyphenylethyl) -4H-1,2, 4-triazol-3-yl) phenoxy) acetate (Compound 66-7)
Figure BDA0002627095020000323
Compound 66-6(304mg, 0.66mmol) was dissolved in 7ml ethanol and 1N NaOH was added overnight at room temperature. Water was added to dilute the reaction mixture, and 1N HCl was added to adjust the pH of the reaction mixture to 4, whereby white flocs were precipitated, followed by filtration and drying to obtain 66-7(256mg, yield 87%) as a white solid.
1H NMR(400MHz,DMSO-d6)δ7.50–7.44(m,2H),7.30(dq,J=14.0,6.9Hz,3H),7.11–7.06(m,2H),7.03–6.98(m,2H),6.92(d,J=7.3Hz,2H),6.85–6.80(m,2H),5.23(s,2H),4.72(s,2H),3.71(s,3H),2.88(d,J=6.4Hz,2H),2.86–2.81(m,2H).+ESI:444[M+H]+
And 7: synthesis of methyl 5- (2- (4- (4-benzyl-5- (4-methoxyphenylethyl) -4H-1,2, 4-triazol-3-yl) phenoxy) acetamido) nicotinate (Compound 66)
Figure BDA0002627095020000324
Compound 66-7(50mg, 0.113mmol), methyl 5-aminopyridine-3-carboxylate (11.99mg, 0.113mmol), EDC (33mg, 0.172mmol) and HOBT (23mg,0.172mmol) were dissolved in DMF (1ml), and DIPEA (72. mu.M, 0.384mmol) was added with stirring and reacted at room temperature overnight. Extraction with ethyl acetate, washing of the organic phase with brine, drying over anhydrous sodium sulfate, and purification by thin layer chromatography gave compound 66(30mg, yield 50%) as a pale yellow solid.
1H NMR(400MHz,CDCl3)δ9.04–9.01(m,1H),8.93(s,1H),8.68(d,J=2.2Hz,1H),7.58–7.41(m,2H),7.40–7.30(m,3H),7.09–6.95(m,4H),6.90(s,2H),6.86–6.74(m,2H),4.95(s,2H),4.76–4.59(m,2H),4.02–3.93(m,3H),3.84–3.71(m,3H),3.06(t,J=7.8Hz,2H),2.95–2.81(m,2H).HPLC tR=3.13min purity:100%LCMS:578[M+H]+
Compounds 67-76 were prepared using the same route and synthesis as compound 66.
Figure BDA0002627095020000331
Figure BDA0002627095020000341
Synthesis of N- (4- (-benzyl-5- (4-methoxyphenethyl) -4H-1,2, 4-triazol-3-yl) phenyl) pyridine-3-sulfonamide (Compound 78)
Figure BDA0002627095020000342
Step 1: synthesis of 4-Nitrobenzohydrazide (Compound 78-2)
Figure BDA0002627095020000343
Ethyl p-nitrobenzoate 78-1(2g, 10mmol) was dissolved in methanol (12ml), hydrazine monohydrate (2.4g, 75mmol) was added, refluxed overnight, distilled under reduced pressure and toluene was taken dry, and the residue was taken up with ethyl acetate: the solvent mixture of ethanol (2: 1) was washed with stirring and filtered to obtain 78-2(1.53g, 83% yield) as a pale yellow solid. The reaction mixture is directly put into the next step without purification.
Step 2: synthesis of 4-benzyl-3- (4-methoxyphenylethyl) -5- (4-nitrophenyl) -4H-1,2, 4-triazole (Compound 78-3)
Figure BDA0002627095020000351
78-2(1.53g, 8.5mmol) and 66-3(4.82g, 17mmol) were dissolved in dry tetrahydrofuran (25ml), acetic acid (7.4ml) was added with stirring, stirring was carried out for ten minutes in ice bath, mercuric acetate (3g, 9.4mmol) was added, and reaction was carried out in ice bath for two hoursAfter that time, the mixture was stirred at room temperature overnight. Filtration through celite, concentration of the filtrate, extraction of the residue with ethyl acetate and water, washing of the organic phase with saturated brine, drying over anhydrous sodium sulfate, concentration, and purification by column chromatography gave 78-3(1.9g, 57% yield) as a crude product.+ESI:415[M+H]+
And step 3: synthesis of 4 (-4-benzyl-5- (4-methoxyphenylethyl) -4H-1,2, 4-triazol-3-yl) aniline (Compound 78-4)
Figure BDA0002627095020000352
Compound 78-3(1.86g, 4.5mmol) and iron powder (2.5g, 4.5mmol) were suspended in 71mL ethanol, 2.5M NH was added418.6mL of Cl solution. The reaction was carried out at 80 ℃ for 1 hour. The reaction solution was filtered, the filter residue was washed with ethyl acetate, the filtrate was concentrated, the residue was extracted with ethyl acetate/water, the organic phases were combined and washed with saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and evaporated to dryness. With dichloromethane: purification by column chromatography using methanol (20: 1) gave compound 78-4 as an off-white solid (700mg, yield 41%).
1H NMR(400MHz,DMSO-d6)δ7.37–7.22(m,3H),7.21–7.13(m,2H),7.11–7.02(m,2H),6.92(dt,J=6.7,1.4Hz,2H),6.85–6.77(m,2H),6.63–6.54(m,2H),5.49(s,2H),5.18(s,2H),3.70(s,3H),2.91–2.74(m,4H).+ESI:385[M+H]+
And 4, step 4: synthesis of N- (4- (4-benzyl-5- (4-methoxyphenethyl) -4H-1,2, 4-triazol-3-yl) phenyl) pyridine-3-sulfonamide (Compound 78)
Figure BDA0002627095020000353
Compound 78-4(40mg, 0.1mmol) was dissolved in 0.5mL pyridine, 3-pyridinesulfonyl chloride (13. mu.L, 0.1mmol) was added, and the mixture was heated at 60 ℃ overnight. The reaction mixture was diluted with ethyl acetate, washed with water, and the organic layer was washed with saturated sodium chloride, dried over anhydrous sodium sulfate, filtered, and evaporated to dryness. The residue was purified by column chromatography, dichloromethane: methanol (15: 1) to give compound 78 as a white solid (47mg, yield 88%)。1H NMR(400MHz,DMSO-d6)δ10.83(s,1H),8.93(dd,J=2.4,0.8Hz,1H),8.80(dd,J=4.9,1.6Hz,1H),8.15(ddd,J=8.1,2.4,1.6Hz,1H),7.61(ddd,J=8.1,4.8,0.8Hz,1H),7.48–7.39(m,2H),7.32–7.15(m,5H),7.13–7.04(m,2H),6.88–6.77(m,4H),5.20(s,2H),3.71(s,3H),2.87(q,J=3.4Hz,4H).+ESI:526[M+H]+
Synthesis of 1- (4- (4-benzyl-5- (4-methoxyphenylethyl) -4H-1,2, 4-triazol-3-yl) phenyl) -3- (3-methoxyphenyl) urea (Compound 79)
Figure BDA0002627095020000361
Compound 78-4(40mg, 0.1mmol) was dissolved in 1mL of anhydrous tetrahydrofuran under heating, and m-methoxyphenyl isocyanate (14. mu.L, 0.1mmol) was added thereto, followed by stirring at room temperature overnight. After completion of the reaction, filtration was performed, and the solid was washed with tetrahydrofuran and dried to obtain compound 79(38mg, yield 72%) as a white solid.
1H NMR(400MHz,DMSO-d6)δ8.90(s,1H),8.77(s,1H),7.60–7.51(m,2H),7.50–7.43(m,2H),7.37–7.22(m,3H),7.23–7.14(m,2H),7.13–7.04(m,2H),6.93(dt,J=7.8,1.6Hz,3H),6.86–6.78(m,2H),6.56(ddd,J=8.2,2.6,0.9Hz,1H),5.26(s,2H),3.73(s,3H),3.71(s,3H),2.94–2.79(m,4H).+ESI:534[M+H]+
Biological evaluation
The present invention is further described below by referring to examples of enzyme activity test, but these examples are not intended to limit the scope of the present invention.
The experimental method of the present invention in which the specific conditions are not specified in the enzyme activity test examples is generally performed under the conventional conditions or under the conditions recommended by commercial manufacturers. Reagents of specific sources are not indicated, and conventional reagents are purchased in the market.
The binding activity of compounds to PDE protein domains was tested using the Fluorescence Anisotropy assay (fluoroscence Anisotropy). The principle of the fluorescence anisotropy testing method is as follows: by detecting the change of molecular weight before and after the interaction between the small molecules marked by the fluorescein and other molecules, the fluorescence polarization values in the horizontal direction and the vertical direction are calculated for correlation analysis. If the binding equilibrium between the fluorescently labeled small molecules and the large molecules is established, the fluorescence is excited and moves slowly, and the measured fluorescence polarization value is increased. If the combination between the fluorescence labeling micromolecule and the macromolecule is replaced by other ligands, the rotation or overturning speed of the fluorescence labeling micromolecule in a free state is increased, the emitted light is depolarized relative to an excitation light plane, the measured polarized light value is reduced, and the fluorescence anisotropy of the sample is calculated.
The fluorogenic substrate was Atorvastatin (Atorvastatin) linked to a fluorescent molecule at a working concentration of 25 nM. The working concentration of PDE protein was 20nM, the total reaction system was 40. mu.l, the buffer was PBS (containing 0.05% CHAPS), the primary screening concentration of the compound was 1. mu.M, and the IC50 was determined for compounds with an inhibition greater than 50% under these conditions. The final DMSO concentration was chosen to be two thousandths, taking into account the solubility of the compound and the effect of DMSO on the assay. All measurements were performed under these conditions. All components were mixed and reacted for 6h at room temperature in the absence of light or after overnight reaction at 4 ℃ the anisotpy value was determined.
The test uses a kang ning (corning) all-black, low-binding treated 384-well plate (product number is CLS3575), the test instrument is a BioTek synergy2 detector, the Excitation wavelength (Excitation) is 485nm, and the emission wavelength (emission) is 530 nm. Blank values were read from wells with buffer only.
Numerical value processing: the inhibition ratio was (C-F)/(C-B). times.100%
Wherein: c: antipyropy value of complete binding of fluorescent substrate to protein
B: background value of fluorogenic substrate Anisotropyp
F: anistropy values at the corresponding concentrations of the compounds
The concentration of the compound and the corresponding inhibition rate were plotted as an S-curve. To give IC of the corresponding compound50
Figure BDA0002627095020000362
Figure BDA0002627095020000371
Figure BDA0002627095020000381
The enzyme activity test example shows that the series of compounds have better PDE delta inhibitory activity at a cellular level. The application of the compound is suitable for preparing medicaments for treating or preventing KRas-PDE delta mediated diseases, and is particularly suitable for preparing KRas-PDE delta mediated antitumor medicaments.
The technical features of the embodiments described above may be arbitrarily combined, and for the sake of brevity, all possible combinations of the technical features in the embodiments described above are not described, but should be considered as being within the scope of the present specification as long as there is no contradiction between the combinations of the technical features.
The above-mentioned embodiments only express several embodiments of the present invention, and the description thereof is more specific and detailed, but not construed as limiting the scope of the invention. It should be noted that, for a person skilled in the art, several variations and modifications can be made without departing from the inventive concept, which falls within the scope of the present invention. Therefore, the protection scope of the present patent shall be subject to the appended claims.

Claims (9)

1. Triazole derivatives having the structure of formula (V) or a pharmaceutically acceptable salt thereof:
Figure FDA0002969559430000011
R1is phenyl or benzyl;
R2is C1-C6An alkyl group;
R11or R12Each independently selected from hydrogen atom, C1-C4Alkyl, aryl, heteroaryl, and heteroaryl,3-10 membered cycloalkyl, 5-10 membered aryl or 5-10 membered heteroaryl; said C is1-C4Alkyl, 3-10 membered cycloalkyl, 5-10 membered aryl or 5-10 membered heteroaryl are each independently further optionally substituted by one or more groups selected from halogen, hydroxy, cyano, nitro, trifluoromethyl, C1-C4Alkyl, 3-10 membered cycloalkyl or C1-C4Alkoxy radical,
Figure FDA0002969559430000012
Substituted with the substituent(s);
n is an integer of 0 to 4;
R9or R10Each independently selected from hydrogen atom or C1-C4An alkyl group;
the R is9And R10Optionally together with the nitrogen atom to which it is attached, a 3-8 membered heterocyclyl containing 1-3 heteroatoms selected from N, O, S or a 5-10 membered heteroaryl containing 1-3 heteroatoms selected from N, O and S;
and the triazole derivative with the structure of the formula (V) does not comprise
Figure FDA0002969559430000013
2. Triazole derivatives having the structure of formula (V) or a pharmaceutically acceptable salt thereof:
Figure FDA0002969559430000014
R1is phenyl or benzyl;
R2is methyl, ethyl, propyl or 2-methylpropyl;
R11or R12Each independently selected from:
a hydrogen atom,
Figure FDA0002969559430000015
Or R11And R12To be connected withThe nitrogen atoms together form the following group:
Figure FDA0002969559430000021
R14or R15Each independently selected from: hydrogen atom, halogen, phenyl, trifluoromethyl, C1-C4Alkyl radical, C1-C3Alkoxy, pyrrolidinyl, morpholinyl, or-NR16R17、-CONR16R17、-C(O)R16or-COOR17
R16Or R17Each independently selected from hydrogen atom, C1-C4Alkyl or
Figure FDA0002969559430000022
Wherein n is an integer of 1 to 4; r18Or R19Each independently selected from a hydrogen atom or C1-C4An alkyl group;
the R is16And R17Together with the nitrogen atom to which it is attached, form a 3-8 membered heterocyclic group containing 1-3 heteroatoms selected from N, O, S or a 5-10 membered heteroaryl group containing 1-3 heteroatoms selected from N, O and S;
and the triazole derivative with the structure of the formula (V) does not comprise
Figure FDA0002969559430000023
3. The triazole derivative or a pharmaceutically acceptable salt thereof according to claim 1 or 2, wherein the compound is a compound represented by general formula (VI)
Figure FDA0002969559430000024
R1Is phenyl or benzyl;
R14selected from halogenAn element, a phenyl group, a cyclohexyl group or a dimethylamino group.
4. The triazole derivative of the general formula or the pharmaceutically acceptable salt thereof according to claim 2, wherein the compound is selected from any of the following compounds:
Figure FDA0002969559430000025
Figure FDA0002969559430000031
Figure FDA0002969559430000041
5. a pharmaceutical composition, which comprises a therapeutically effective amount of the triazole derivative or pharmaceutically acceptable salt thereof according to any one of claims 1 to 4, and a pharmaceutically acceptable excipient.
6. Use of the triazole derivatives or pharmaceutically acceptable salts thereof according to any one of claims 1 to 4 and the pharmaceutical composition according to claim 5 in preparation of a medicament for inhibiting PDE δ.
7. Use of the triazole derivatives or pharmaceutically acceptable salts thereof according to any one of claims 1 to 4 and the pharmaceutical composition according to claim 5 in the preparation of a medicament for treating or preventing KRas-PDE delta mediated diseases.
8. Use according to claim 7, wherein the KRas-PDE δ mediated disease is a tumor.
9. The use of claim 8, wherein the tumor is squamous cell cancer, prostate cancer, renal cell carcinoma, kaposi's sarcoma, non-small cell lung cancer, lymphoma, thyroid cancer, breast cancer, head and neck cancer, uterine cancer, esophageal cancer, melanoma, bladder cancer, genitourinary cancer, gastrointestinal cancer, glial cancer, colorectal cancer, or ovarian cancer.
CN202010800210.4A 2017-10-17 2017-10-17 Triazole derivative and preparation method and application thereof Expired - Fee Related CN111875583B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010800210.4A CN111875583B (en) 2017-10-17 2017-10-17 Triazole derivative and preparation method and application thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710964884.6A CN109666022B (en) 2017-10-17 2017-10-17 Triazole derivative and preparation method and application thereof
CN202010800210.4A CN111875583B (en) 2017-10-17 2017-10-17 Triazole derivative and preparation method and application thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN201710964884.6A Division CN109666022B (en) 2017-10-17 2017-10-17 Triazole derivative and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN111875583A CN111875583A (en) 2020-11-03
CN111875583B true CN111875583B (en) 2021-06-15

Family

ID=66139790

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202010800210.4A Expired - Fee Related CN111875583B (en) 2017-10-17 2017-10-17 Triazole derivative and preparation method and application thereof
CN201710964884.6A Active CN109666022B (en) 2017-10-17 2017-10-17 Triazole derivative and preparation method and application thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN201710964884.6A Active CN109666022B (en) 2017-10-17 2017-10-17 Triazole derivative and preparation method and application thereof

Country Status (1)

Country Link
CN (2) CN111875583B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023069721A2 (en) * 2021-10-22 2023-04-27 Carmot Therapeutics, Inc. Oxazole, oxadiazole, and indole derivatives for the inhibition of usp28
CN115160285B (en) * 2022-08-25 2022-11-29 云南大学 Thioamides ALDH 2 Agonist, preparation method and application

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013178572A1 (en) * 2012-05-30 2013-12-05 F. Hoffmann-La Roche Ag Triazolo compounds as pde10 inhibitors
CN103864754A (en) * 2012-12-10 2014-06-18 中国科学院上海药物研究所 Five-membered azole heterocycle compound, preparation method, drug composition and use thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MY133406A (en) * 2001-11-28 2007-11-30 Ipsen Pharma Sas 5-sulphanyl-4h-1,2,4-triazole derivatives and their use as a medicament
CN1934095A (en) * 2004-03-08 2007-03-21 惠氏公司 Ion channel modulators
WO2005090320A2 (en) * 2004-03-12 2005-09-29 Wyeth Triazole derivatives and method of using the same to treat hiv infections
CA2586612C (en) * 2004-11-18 2016-10-11 Synta Pharmaceuticals Corp. Triazole compounds that modulate hsp90 activity

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013178572A1 (en) * 2012-05-30 2013-12-05 F. Hoffmann-La Roche Ag Triazolo compounds as pde10 inhibitors
CN103864754A (en) * 2012-12-10 2014-06-18 中国科学院上海药物研究所 Five-membered azole heterocycle compound, preparation method, drug composition and use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SYNTHESIS AND ANTITUMOR ACTIVITY OF 2-N- AND 3-S-SUBSTITUTED 5-[2-(4-)BENZYLOXYPHENYL]- 1,2,4-TRIAZOLES AND ACYLHYDRAZIDES;L. A. Grigoryan等;《Pharmaceutical Chemistry Journal》;20120930;第46卷(第9期);第531-535页 *

Also Published As

Publication number Publication date
CN109666022B (en) 2021-06-15
CN111875583A (en) 2020-11-03
CN109666022A (en) 2019-04-23

Similar Documents

Publication Publication Date Title
US11008305B2 (en) DNA-PK inhibitors
ES2671354T3 (en) DNA-PK inhibitors
CN113544128A (en) KRAS-G12C inhibitors
CN105315285B (en) 2,4 2 substitution 7H pyrrolo-es [2,3 d] pyrimidine derivatives, its preparation method and purposes pharmaceutically
CN112724145A (en) Pyrazine derivatives for inhibiting SHP2 activity
AU2018269275B2 (en) Five- and six-membered aza-aromatic compound, preparation method therefor, pharmaceutical composition, and application
JP2017531679A (en) Indole carboxamides useful as kinase inhibitors
KR20200115583A (en) 2H-indazole derivatives as CDK4 and CDK6 inhibitors and their therapeutic use
PT3013814T (en) Substituted tetrahydrocarbazole and carbazole carboxamide compounds useful as kinase inhibitors
AU2019218187A1 (en) Dioxinoquinoline compounds, preparation method and uses thereof
CN113454081A (en) Imidazopyridinyl compounds and their use for the treatment of proliferative diseases
EP3856735B1 (en) Fused bicyclic heterocycles as therapeutic agents
AU2019383103B2 (en) Aromatic ring-linked dioxane-quinazoline or -quinoline compounds, compositions and use thereof
CN111875583B (en) Triazole derivative and preparation method and application thereof
WO2004014873A1 (en) 4-substituted quinazoline-8-carboxyamide derivative and pharmaceutically acceptable addition salt thereof
JP4878285B2 (en) Indole derivatives with apoptosis-inducing action
JP2024514015A (en) Alkynylphenylbenzamide compounds and their uses
US11407760B2 (en) Dioxinoquinoline compounds, preparation method and uses thereof
TW202202500A (en) Pb2 inhibitor, and preparation method therefor and use thereof
CN111320621B (en) Indolizine compound and preparation method and application thereof
JP7110335B2 (en) Pyridoquinazoline derivatives useful as protein kinase inhibitors
KR20140044790A (en) Piperazinedione compounds
CN107793360B (en) Indoleamine 2, 3-dioxygenase inhibitor and application thereof
CN115515589A (en) ULK1/2 inhibitors and methods of use thereof
CN111065635B (en) Novel pyrimidine derivatives as MTH1 inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CF01 Termination of patent right due to non-payment of annual fee

Granted publication date: 20210615

Termination date: 20211017

CF01 Termination of patent right due to non-payment of annual fee