CN109836378B - Sphingomyelin synthase inhibitor, preparation method and application thereof - Google Patents

Sphingomyelin synthase inhibitor, preparation method and application thereof Download PDF

Info

Publication number
CN109836378B
CN109836378B CN201811405438.2A CN201811405438A CN109836378B CN 109836378 B CN109836378 B CN 109836378B CN 201811405438 A CN201811405438 A CN 201811405438A CN 109836378 B CN109836378 B CN 109836378B
Authority
CN
China
Prior art keywords
phenyl
compound
nicotinic acid
acid carboxamide
benzyloxy
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.)
Active
Application number
CN201811405438.2A
Other languages
Chinese (zh)
Other versions
CN109836378A (en
Inventor
许忻
张瑱
李云飞
张立明
蒋青云
臧成旭
杨茂志
郁群
刘磊
刘强
张林丽
陈海继
奚丞昊
葛建
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.)
Zhejiang Huahai Pharmaceutical Co Ltd
Shanghai Synergy Pharmaceutical Sciences Co Ltd
Original Assignee
Zhejiang Huahai Pharmaceutical Co Ltd
Shanghai Synergy Pharmaceutical Sciences 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 Zhejiang Huahai Pharmaceutical Co Ltd, Shanghai Synergy Pharmaceutical Sciences Co Ltd filed Critical Zhejiang Huahai Pharmaceutical Co Ltd
Publication of CN109836378A publication Critical patent/CN109836378A/en
Application granted granted Critical
Publication of CN109836378B publication Critical patent/CN109836378B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Pyridine Compounds (AREA)

Abstract

The invention provides a compound shown in a general formula (I), and pharmaceutically acceptable salt or a stereoisomer thereof, wherein X, Y, M, Z, R and Ar are respectively defined in the specification. The invention also provides application of at least one compound shown in the general formula (I) in a therapeutically effective amount, pharmaceutically acceptable salts or stereoisomers thereof, the compound, the pharmaceutically acceptable salts or the stereoisomers thereof, or a pharmaceutical composition containing the compound in the general formula (I) in preparing a medicament for preventing and/or treating diseases caused by abnormal increase of sphingomyelin level. The compound shown in the general formula (I) and the pharmaceutically acceptable salt or stereoisomer thereof can selectively inhibit sphingomyelin synthase, and have remarkable in-vivo and in-vitro efficacy and high safety.

Description

Sphingomyelin synthase inhibitor, preparation method and application thereof
Technical Field
The invention relates to the technical field of medicinal chemistry, in particular to a sphingomyelin synthase inhibitor, a preparation method and application thereof.
Background
Atherosclerosis is the most common disease in cardiovascular system diseases, and the main pathological features of atherosclerosis are lipid deposition under the artery intima, and proliferation and migration of smooth muscle cells, formation of fibrous plaque, further causing vascular wall sclerosis, luminal stenosis and thrombosis, gradually developing and forming atherosclerotic plaque, wherein the atherosclerosis is often accompanied by hypertension, hypercholesterolemia, diabetes and the like, has great harm to the health of people, and is one of the main death reasons of the old.
The occurrence and the formation of atherosclerosis are a complicated pathological process, the age, smoking, drinking, hypertension, hyperglycemia, hyperlipidemia and the like are risk factors for the occurrence and the development of atherosclerosis, and with the deep understanding of people on the pathogenesis of atherosclerosis, medicaments for treating atherosclerosis are continuously developed and marketed, such as antiplatelet medicaments, vasodilators, thrombolytic medicaments, lipid-lowering medicaments, antioxidants, cyclooxygenase inhibitors and the like, however, the medicaments are clinically applied for preventing and treating atherosclerosis for a long time and still bring adverse reactions of some medicaments.
Sphingomyelin is a main lipid component in cell membrane and plasma proteins and has wide and important biological functions, epidemiological investigation shows that a large amount of sphingomyelin is accumulated in atherosclerotic plaques, clinical tests of large samples show that the sphingomyelin level is positively correlated with lipid risk score, the sphingomyelin level is higher than that of normal people in human familial hyperlipidemia patients, the sphingomyelin level in plasma is also obviously increased in animal models of atherosclerosis, and the sphingomyelin level and the sphingomyelin aggregation in plasma are one of risk factors of atherosclerosis. In recent years, numerous studies show that sphingomyelin plays a very important role in the development of atherosclerosis, and on one hand, sphingomyelin can induce atherosclerosis by inhibiting lipid metabolism pathways such as the lipolysis of triglyceride and the delay of the clearance of lipoprotein remnant, and on the other hand, sphingomyelin can regulate the inflammatory response associated with atherosclerosis by regulating the activation of NF-kappa B (nuclear factor-kappa B), thereby influencing the progression of atherosclerosis.
Sphingomyelin synthase (SMS) is a key enzyme in the last step of Sphingomyelin synthesis, and directly regulates the level of Sphingomyelin; numerous studies have revealed that lowering sphingomyelin levels by inhibiting SMS activity may be a new approach to the treatment of atherosclerosis. In recent years, research institutes and pharmaceutical enterprises at home and abroad have already carried out the development of SMS inhibitor drugs, but at present, no SMS inhibitor drug at home and abroad enters clinical research, and the development of SMS small-molecule inhibitors can help to find safer and more effective anti-atherosclerosis therapeutic drugs.
Disclosure of Invention
The invention aims to provide a sphingomyelin synthase inhibitor, a preparation method and application thereof. The specific scheme is as follows:
the invention firstly provides a compound shown in a general formula (I), and pharmaceutically acceptable salt or stereoisomer thereof.
Figure BDA0001877227330000021
Wherein X, Y and Z are each independently selected from a carbon atom or a nitrogen atom, and X, Y and
z is not simultaneously selected from nitrogen atoms;
m is selected from-NH-or-O-;
ar is selected from substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl; preferably, said substituted aryl or substituted heteroaryl is optionally substituted with one or more of the following substituents:
halogen, -C 1-10 Alkyl, -C 1-10 Alkoxy, halogen substituted C 1-10 Alkyl and halogen substituted C 1-10 An alkoxy group;
r is selected from hydrogen, deuterium, halogen, nitro, cyano and-C 1-6 Alkyl, -C 1-6 Alkoxy, substituted C 1-6 Alkyl, substituted C 1-6 An alkoxy group; preferably, substituted C 1-6 Alkyl, substituted C 1-6 Alkoxy is optionally substituted with one or more of the following substituents:
hydrogen, deuterium, halogen, nitro, cyano, -C 1-6 Alkyl and-C 1-6 An alkoxy group.
Some embodiments of the present invention relate to the aforementioned compounds, pharmaceutically acceptable salts or stereoisomers thereof, wherein, when Y and Z are carbon atoms, X is a nitrogen atom or a carbon atom; z is a nitrogen atom when X and Y are carbon atoms;
ar is selected from phenyl, pyridyl, substituted phenyl or substituted pyridyl, wherein said substituted phenyl or substituted pyridyl is optionally substituted with one or more of the following substituents:
halogen, -C 1-8 Alkyl, -C 1-8 Alkoxy, halogen substituted C 1-8 Alkyl, halogen substituted C 1-8 An alkoxy group;
r is hydrogen.
Some embodiments of the invention relate to the aforementioned compounds, pharmaceutically acceptable salts, or stereoisomers thereof, wherein Ar is
Figure BDA0001877227330000031
R 1 Selected from-F, -Cl, -Br, -C 1-8 Alkyl, -C 1-8 Alkoxy, halogen substituted C 1-8 Alkyl, halogen substituted C 1-8 An alkoxy group; r 2 Selected from-H, -F, -Cl, -Br and-C 1-3 An alkyl group.
Some embodiments of the present invention relate to the aforementioned compounds, pharmaceutically acceptable salts or stereoisomers thereof, wherein R is 1 is-F, -Cl, -CH 3 、-CH 2 CH 3 、-OCH 3 、-OCH 2 CH 3 、-O-(CH 2 ) n -CH 3 、-O-(CH 2 ) n -Cl; n is an integer of 2 to 6.
Some embodiments of the present invention relate to the aforementioned compounds, pharmaceutically acceptable salts or stereoisomers thereof, wherein X and Y are carbon atoms, Z is a nitrogen atom; r 2 is-H.
Some embodiments of the present invention relate to the aforementioned compounds, pharmaceutically acceptable salts or stereoisomers thereof, wherein Y and Z are carbon atoms and X is a nitrogen atom or a carbon atom.
Some embodiments of the invention relate to the aforementioned compound, a pharmaceutically acceptable salt, or a stereoisomer thereof, wherein Ar is selected from
Figure BDA0001877227330000041
Figure BDA0001877227330000042
In some embodiments of the present invention, the compound of formula (I) may be specifically:
n- [2- (2-chloro-5-fluorobenzyloxy) phenyl ] nicotinic acid carboxamide;
n- [2- (2, 6-dichlorobenzyloxy) phenyl ] nicotinic acid carboxamide;
n- [2- (2-methyl-5-fluorobenzyloxy) phenyl ] nicotinic acid carboxamide;
n- [2- (2-methoxybenzyloxy) phenyl ] nicotinic acid carboxamide;
n- {2- [ (2-ethyl) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2, 6-dimethyl) benzyloxy) ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2-ethoxy) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2-methoxy-5-chloro) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2, 5-dichloro) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2-methyl-5-chloro) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [2- (4-chlorobutoxy) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [2- (5-chloropentyloxy) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [2- (6-chlorohexyloxy) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- [2- (2-hexyloxybenzyloxy) phenyl ] nicotinic acid carboxamide;
n- [2- (2-heptyloxybenzyloxy) phenyl ] nicotinic acid carboxamide;
n- {2- [ (2-hexyloxy) -5-chlorobenzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2-heptyloxy) -5-chlorobenzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [2- (4-chlorobutoxy) benzyloxy ] phenyl } isonicotinic acid carboxamide;
n- {2- [2- (5-chloropentyloxy) benzyloxy ] phenyl } isonicotinic acid carboxamide;
n- {2- [ (5-chloro-2-heptyloxybenzyl) amino ] phenyl } nicotinamide;
n- {2- [ (5-chloro-2- (6-chlorohexyloxy) benzyl) amino ] phenyl } nicotinamide;
n- {2- [ (2-chloro-5-fluorobenzyl) amino ] phenyl } nicotinamide;
n- {2- [ (2, 6-dichlorobenzyl) amino ] phenyl } nicotinamide;
n- {2- [ (5-fluoro-2-methylbenzyl) amino ] phenyl } nicotinamide;
n- {2- [ (2- (4-chlorobutoxy) benzyl) amino ] phenyl } isonicotinamide;
n- {2- [ (2- (5-chloropentyloxy) benzyl) amino ] phenyl } isonicotinamide;
n- {2- [ (2-hexyloxybenzyl) amino ] phenyl } isonicotinamide;
n- {2- [ (5-chloro-2-heptyloxybenzyl) amino ] phenyl } benzamide;
n- {2- [ (6-chlorohexyloxybenzyl) amino ] phenyl } nicotinamide;
n- {2- [2- (6-chlorohexyloxy) benzyloxy ] phenyl } isonicotinic acid carboxamide;
n- {2- [2- (heptyloxy) benzyloxy ] phenyl } isonicotinic acid carboxamide.
The invention also provides a preparation method of the compound shown in the general formula (I), which comprises the following steps:
Figure BDA0001877227330000061
condensing the compound 1 and the compound 2 to obtain a compound 3;
removing benzyl protection from the compound 3 through catalytic hydrogenation to obtain a compound 4;
compound 4 and
Figure BDA0001877227330000062
the final product, a compound of formula (I), should be obtained;
wherein M is-O-; r 3 Selected from-OH, -F, -Cl or-Br.
In some implementations of the inventionIn the way that in the above-mentioned mode,
Figure BDA0001877227330000063
in particular substituted benzyl bromide; more specifically, the substituted benzyl bromide is optionally substituted by one or more of the following substituents: halogen, -C 1-8 Alkyl, -C 1-8 Alkoxy, halogen substituted C 1-8 Alkyl, halogen substituted C 1-8 An alkoxy group.
The invention also provides a preparation method of the compound shown in the general formula (I), which comprises the following steps:
Figure BDA0001877227330000064
condensing the compound 6 and the compound 2 to obtain a compound 7;
reducing nitro group of the compound 7 under the condition of catalytic hydrogenation to obtain a compound 8;
carrying out reductive amination on the compound 8 and Ar-CHO to obtain a compound shown in a general formula (I); wherein M is-NH-; r 3 Is selected from-OH, -F, -Cl or-Br.
In some embodiments of the invention, Ar-CHO may be specifically substituted benzaldehydes; more specifically, the substituted benzaldehyde is optionally substituted by one or more of the following substituents: halogen, -C 1-8 Alkyl, -C 1-8 Alkoxy, halogen substituted C 1-8 Alkyl, halogen substituted C 1-8 An alkoxy group.
X, Y, Z, R and Ar involved in the preparation method of the compound are as described above.
The invention also provides a pharmaceutical composition, which comprises at least one therapeutically effective amount of the compound shown in the general formula (I), pharmaceutically acceptable salt or stereoisomer thereof and optionally one or more pharmaceutically acceptable carriers and/or diluents.
The pharmaceutical compositions of the present invention may further comprise one or more pharmaceutically or physiologically acceptable carriers and/or diluents which will be appropriately formulated for administration. For example, the pharmaceutically or physiologically acceptable carrier can be saline, hot water under pressure, ringer's solution, buffered saline, dextrose, maltodextrin, glycerol, ethanol, and mixtures thereof. The pharmaceutical composition of the present invention may further include pharmaceutically or physiologically acceptable additives such as diluents, lubricants, binders, glidants, disintegrants, sweeteners, flavoring agents, wetting agents, dispersants, surfactants, solvents, coating agents, foaming agents, or aromatics.
Examples of diluents that may be used include, but are not limited to, lactose, sucrose, starch, kaolin, salt, mannitol, and dicalcium phosphate; examples of lubricants include, but are not limited to, talc, starch, magnesium or calcium stearate, lycopodium and stearic acid; examples of binders include, but are not limited to, microcrystalline cellulose, gum tragacanth, dextrose solutions, acacia mucilage, gelatin solutions, sucrose and starch pastes; examples of glidants include, but are not limited to, colloidal silicon dioxide; examples of disintegrants include, but are not limited to, croscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar, and carboxymethylcellulose; examples of sweetening agents include, but are not limited to, sucrose, lactose, mannitol, and artificial sweeteners, such as sodium cyclamate and saccharin, and any number of spray-dried flavoring agents; examples of flavoring agents include, but are not limited to, natural flavors extracted from plants, such as fruits, and better tasting compounds, such as, but not limited to, mint and methyl salicylate; examples of wetting agents include, but are not limited to, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether.
The pharmaceutically acceptable carrier optionally added to the pharmaceutical composition of the present invention may also be: water, alcohol, honey, mannitol, sorbitol, dextrin, lactose, caramel, gelatin, calcium sulfate, magnesium stearate, talcum powder, kaolin, glycerol, tween, agar-agar, calcium carbonate, calcium bicarbonate, surfactant, cyclodextrin and derivatives thereof, phospholipids, phosphates, starch and derivatives thereof, silicon derivatives, celluloses and derivatives thereof, pyrrolidones, polyethylene glycols, acrylics, phthalates, acrylic acid copolymers and trimellitates.
The pharmaceutical compositions provided by the present invention may be prepared in any form, such as granules, powders, tablets, coated tablets, capsules, pills, syrups, drops, solutions, suspensions and emulsions, or sustained release formulations of the active ingredient, wherein examples of capsules include hard or soft gelatin capsules, and granules and powders may be in non-effervescent or effervescent form.
The pharmaceutical compositions of the present invention may be administered by a variety of routes according to conventional methods, including oral, intravenous, intraarterial, intraperitoneal, intrapleural, transdermal, nasal, inhalation, rectal, ocular and subcutaneous introduction.
The invention also provides application of the compound, the pharmaceutically acceptable salt or the stereoisomer thereof, or the pharmaceutical composition in preparing a medicament for preventing and treating diseases caused by abnormal increase of sphingomyelin level, wherein the diseases include but are not limited to at least one of atherosclerosis, fatty liver and obesity.
The present invention also provides a method for preventing and/or treating atherosclerosis, fatty liver or obesity, which comprises administering to a subject in need thereof a therapeutically effective amount of the aforementioned compound, a pharmaceutically acceptable salt or stereoisomer thereof, or a pharmaceutical composition of the aforementioned.
The aforementioned compounds, pharmaceutically acceptable salts or stereoisomers thereof are generally provided in a dosage range of about 0.001mg/kg to 1000mg/kg per day, preferably about 0.01mg/kg to 100mg/kg, more preferably about 0.1 to 20mg/kg, and the pharmaceutical composition is in a dosage range calculated on the amount of the aforementioned compound it contains.
The term "halogen" refers to fluorine, chlorine, bromine and iodine, preferably fluorine and chlorine.
The term "alkyl" includes saturated aliphatic groups, including straight-chain alkyl groups (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, etc.), branched-chain alkyl groups (isopropyl, tert-butyl, isobutyl, etc.), cycloalkyl groups (cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl), alkyl-substituted cycloalkyl groups, and cycloalkyl-substituted alkyl groups.
In certain embodiments, the linear or branched alkyl group has 6 or fewer carbon atoms in the backbone (e.g., linear is C) 1-6 The branched chain is C 3-6 ) And more preferably 4 or less carbon atoms. Likewise, preferred cycloalkyl groups have 3 to 8 carbon atoms in their ring structure, more preferably 5 or 6 carbons in their ring structure.
The term "C 1-8 Alkyl "includes alkyl groups containing 1 to 8 carbon atoms.
The term "substituted alkyl" refers to an alkyl group in which a hydrogen on one or more carbons of the hydrocarbon backbone is replaced with a substituent. The substituents may include: alkenyl, alkynyl, halogen, hydroxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, hydroxycarbonyl, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxy, phosphate, phosphonate, cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl, and ureido), amidino, imino, mercapto, alkylthio, arylthio, hydroxythiocarbonyl, sulfate, alkylsulfinyl, sulfonic, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic group.
The term "aryl" includes 6-8 membered monocyclic aromatic groups, such as phenyl, cyclooctenyl; "aryl" also includes 8-14 membered polycyclic (bicyclic, tricyclic) aryl groups such as naphthalene, phenanthrene, and the like.
The term "heteroaryl" refers to an aromatic cyclic group in which at least one ring carbon atom is replaced by a heteroatom selected from O, S, N, and may contain 1 to 4 heteroatoms; the heteroaryl group may be monocyclic heteroaryl or polycyclic heteroaryl, and may be 5-14-membered heteroaryl, 5-12-membered heteroaryl, 5-10-membered heteroaryl, 5-8-membered heteroaryl, 5-6-membered heteroaryl, such as pyrrole, furan, thiophene, thiazole, isothiazole, imidazole, triazole, tetrazole, pyrazole, oxazole, isoxazole, pyridine, pyrazine, pyridazine and pyrimidine, benzoxazole, benzodiazole, benzothiazole, benzimidazole, benzothiophene, methylenedioxyphenyl, quinoline, isoquinoline, naphthyridine, indole, benzofuran, purine, benzofuran, deazapurine or indolizine. These heteroaryl groups are also referred to as "aryl heterocycles", "heteroaryls" or "heteroaromatics".
Typical heteroaryl groups include 2-or 3-thienyl; 2-or 3-furyl; 2-or 3-pyrrolyl; 2-, 4-or 5-imidazolyl; 3-, 4-or 5-pyrazolyl; 2-, 4-or 5-thiazolyl; 3-, 4-or 5-isothiazolyl; 2-, 4-or 5-oxazolyl; 3-, 4-or 5-isoxazolyl; 3-or 5-1, 2, 4-triazolyl; 4-or 5-1, 2, 3-triazolyl; a tetrazolyl group; 2-, 3-or 4-pyridyl; 3-or 4-pyridazinyl; 3-, 4-or 5-pyrazinyl; 2-pyrazinyl; 2-, 4-or 5-pyrimidinyl.
The term "heteroaryl" also includes groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic or heterocyclic rings, wherein the linking group or point is located on the heteroaromatic ring. Examples include, but are not limited to, 1-, 2-, 3-, 5-, 6-, 7-, or 8-indolizinyl; 1-, 3-, 4-, 5-, 6-or 7-isoindolyl; 2-, 3-, 4-, 5-, 6-, or 7-indolyl; 2-, 3-, 4-, 5-, 6-, or 7-indazolyl; 2-, 4-, 5-, 6-, 7-, or 8-purinyl; 1-, 2-, 3-, 4-, 6-, 7-, 8-, or 9-quinolizinyl; 2-, 3-, 4-, 5-, 6-, 7-or 8-quinolinyl; 1-, 3-, 4-, 5-, 6-, 7-or 8-isoquinolinyl; 1-, 4-, 5-, 6-, 7-or 8-phthalazinyl; 2-, 3-, 4-, 5-, or 6-naphthyridinyl; 2-, 3-, 5-, 6-, 7-, or 8-quinazolinyl; 3-, 4-, 5-, 6-, 7-or 8-cinnolinyl; 2-, 4-, 6-or 7-pteridinyl; 1-, 2-, 3-, 4-, 5-, 6-, 7-, or 8-4aH carbazolyl; 1-, 2-, 3-, 4-, 5-, 6-, 7-, or 8-carbazolyl; 1-, 3-, 4-, 5-, 6-, 7-, 8-, or 9-carboline group; 1-, 2-, 3-, 4-, 6-, 7-, 8-, 9-, or 10-phenanthridinyl; 1-, 2-, 3-, 4-, 5-, 6-, 7-, 8-, or 9-acridinyl; 1-, 2-, 4-, 5-, 6-, 7-, 8-, or 9-pyridyl; 2-, 3-, 4-, 5-, 6-, 8-, 9-, or 10-phenanthrolinyl; 1-, 2-, 3-, 4-, 6-, 7-, 8-, or 9-phenazinyl; 1-, 2-, 3-, 4-, 6-, 7-, 8-, 9-, or 10-phenothiazinyl; 1-, 2-, 3-, 4-, 6-, 7-, 8-, 9-, or 10-phenazinyl; 2-, 3-, 4-, 5-, 6-or 1-, 3-, 4-, 5-, 6-, 7-, 8-, 9-or 10-benzisoquinolinyl; 2-, 3-, 4-or thieno [2, 3-b ] furyl; 2-, 3-, 5-, 6-, 7-, 8-, 9-, 10-, or 11-7H-pyrazino [2, 3-c ] carbazolyl; 2-, 3-, 5-, 6-, or 7-2H-furo [3, 2-b ] -pyranyl; 2-, 3-, 4-, 5-, 7-, or 8-5H-pyrido [2, 3-d ] -o-oxazinyl; 1-, 3-or 5-1H-pyrazolo [4, 3-d ] -oxazolyl; 2-, 4-or 5-4H-imidazo [4, 5-d ] thiazolyl; 3-, 5-or 8-pyrazino [2, 3-d ] pyridazinyl; 2-, 3-, 5-or 6-imidazo [2, 1-b ] thiazolyl; 1-, 3-, 6-, 7-, 8-or 9-furo [3, 4-c ] cinnolinyl; 1-, 2-, 3-, 4-, 5-, 6-, 8-, 9-, 10-, or 11-4H-pyrido [2, 3-c ] carbazolyl; 2-, 3-, 6-or 7-imidazo [1, 2-b ] [1, 2, 4] triazinyl; 7-benzo [ b ] thienyl; 2-, 4-, 5-, 6-or 7-benzoxazolyl; 2-, 4-, 5-, 6-, or 7-benzimidazolyl; 2-, 3-, 4-, 5-, 6-or 7-benzothiazolyl; 1-, 2-, 4-, 5-, 6-, 7-, 8-, or 9-benzoxy; 2-, 4-, 5-, 6-, 7-, or 8-benzoxazine; 1-, 2-, 3-, 5-, 6-, 7-, 8-, 9-, 10-, or 11-1H-pyrrolo [1, 2-b ] [2] benzazepine. Typical fused heteroaryl groups include 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinolinyl; 1-, 3-, 4-, 5-, 6-, 7-or 8-isoquinolinyl; 2-, 3-, 4-, 5-, 6-, or 7-indolyl; 2-, 3-, 4-, 5-, 6-or 7-benzo [ b ] thienyl; 2-, 4-, 5-, 6-or 7-benzoxazolyl; 2-, 4-, 5-, 6-, or 7-benzimidazolyl; 2-, 4-, 5-, 6-or 7-benzothiazolyl.
The aromatic ring of an "aryl" or "heteroaryl" group may be substituted at one or more ring positions with a substituent as described above, such as halogen, hydroxy, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, hydroxycarbonyl, alkylcarbonyl, alkylaminocarbonyl, arylalkylaminocarbonyl, alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, arylalkylcarbonyl, alkenylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonate, cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl, and ureido), amidino, imino, mercapto, alkylthio, arylthio, hydroxythiocarbonyl, sulfate ester, Alkylsulfinyl, sulfonate, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic group, where the aryl group may also be fused or bridged with a non-aromatic alicyclic or heterocyclic ring to form a polycyclic ring (e.g., tetralin).
The term "alkoxy" includes substituted and unsubstituted alkyl groups covalently bonded to an oxygen atom. Examples of alkoxy groups include methoxy, ethoxy, isopropyloxy, propoxy, butoxy, and pentyloxy. Examples of substituted alkoxy groups include haloalkoxy groups. The alkoxy group may be substituted with: alkenyl, alkynyl, halogen, hydroxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, hydroxycarbonyl, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, phosphate, cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl, and urea), amidino, imino, mercapto, alkylthio, arylthio, hydroxythiocarbonyl, alkylsulfinyl, sulfonic, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or aromatic group.
Pharmaceutically acceptable salts of the compounds of the invention may be converted to the corresponding salts by reaction with the corresponding organic or inorganic acids, typically oxalic acid, tartaric acid, maleic acid, succinic acid, citric acid, in organic solvents such as acetonitrile, tetrahydrofuran, and typically nitric acid, hydrochloric acid, sulfuric acid, phosphoric acid.
The term "stereoisomer" includes possible enantiomers, diastereomers, racemates, cis-trans-isomers, tautomers, geometrical isomers, epimers and mixtures thereof of the compound of formula (I), all included in the scope of the present invention.
The compound shown in the general formula (I) and the pharmaceutically acceptable salt or stereoisomer thereof can selectively inhibit sphingomyelin synthase, have obvious in-vivo and in-vitro efficacy and high safety, and can be used for preventing and/or treating diseases caused by abnormal increase of sphingomyelin level.
Detailed Description
The technical solutions of the present invention will be described below with reference to specific embodiments, and the described embodiments are only a part of embodiments of the present invention, but not all embodiments. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
Example 1: n- [2- (2-chloro-5-fluorobenzyloxy) phenyl ] nicotinic acid carboxamide
Figure BDA0001877227330000121
First step synthesis of N- [ (2-benzyloxy) phenyl ] nicotinic acid carboxamide
Nicotinic acid (3.52g, 17.66mmol), dichloromethane (100ml), thionyl chloride (13.5g, 113.72mmol) were added to a dried eggplant-shaped flask, stirred at room temperature for 5 minutes, 2 drops of pyridine were added, heated under reflux for 3 hours, and the solvent and remaining thionyl chloride were distilled off under reduced pressure to obtain nicotinic acid formyl chloride (3g), which was used in the next reaction without purification.
2-Benzyloxyaniline (3.52g, 17.66mmol) was dissolved in dry dichloromethane (100ml), triethylamine (2.68g, 26.49mmol) was added and stirred well. Under ice-water bath conditions, a solution of nicotinic acid formyl chloride (3g, 21.19mmol) in methylene chloride was slowly added dropwise thereto. After dropping, the reaction was carried out at room temperature for 5 hours. Then the reaction system was washed twice with water, twice with saturated sodium chloride, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography to obtain 4.23g of N- [ (2-benzyloxy) phenyl ] nicotinic acid carboxamide with a yield of 78.6%.
Second step synthesis of N- [ (2-hydroxy) phenyl ] nicotinic acid carboxamide
To a solution of N- [ (2-benzyloxy) phenyl ] nicotinic acid carboxamide (3g, 9.86mmol) in methanol (80ml) was added 10% Pd/C (0.4g), followed by reaction at room temperature under a hydrogen pressure of 8MPa for 8 hours. Pd/C was removed by suction filtration, and the filtrate was concentrated to give N- [ (2-hydroxy) phenyl ] nicotinic acid-carboxamide (1.87g) as a product in 88.6% yield.
The third step is N- [2- (2-chloro-5-fluorobenzyloxy) phenyl ] nicotinic acid formamide
Reacting N- [ (2-hydroxy) phenyl group]Nicotinic acid formamide (0.1g,0.47mmol) was dissolved in DMF (10ml), potassium carbonate (0.13g,0.93mmol) was added, and 2-chloro-5-fluorobenzyl bromide (0.1g,0.47mmol) was further added, and the reaction was allowed to proceed at room temperature overnight. Adding water, extracting with ethyl acetate, washing with saturated saline, drying with anhydrous magnesium sulfate, concentrating, and performing column chromatography (PE/EA 0-50%) to obtain N-2- [ (2-chloro-5-fluorobenzyloxy) phenyl]Nicotinic acid carboxamide (0.13g), yield 78%. 1 H NMR(400MHz,Chloroform-d)δ9.03-8.99(m,1H).8.70(dd,J=4.9,1.7Hz,1H),8.63(s,1H),8.56(dd,J=8.0,1.7Hz,1H),8.22(dt,J=8.0,2.0Hz,1H),7.48-7.41(m,3H),7.35-7.31(m,1H),7.31-7.29(m,1H),7.27-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.91(s,1H),5.26(s,2H)MS:m/z(ESI)357[M+1] +
Example 2: n- [2- (2, 6-dichlorobenzyloxy) phenyl ] nicotinic acid carboxamide
Figure BDA0001877227330000141
By a synthetic method similar to example 1, intermediate N- [ (2-hydroxy) phenyl ] nicotinic acid carboxamide and 2, 6-dichlorobromobenzyl reacted to give the title compound 76mg in 78% yield as a pale yellow solid.
1 H NMR(400MHz,Chloroform-d)δ9.01–8.97(m,1H),8.74(dd,J=4.9,1.7Hz,1H),8.61(s,1H),8.53(dd,J=8.0,1.7Hz,1H),8.19(dt,J=8.0,2.0Hz,1H),7.45–7.38(m,3H),7.32–7.28(m,1H),7.28–7.25(m,1H),7.25–7.20(m,1H),7.16(td,J=7.7,1.7Hz,1H),7.10(td,J=7.7,1.6Hz,1H),5.42(s,2H).MS m/z(ESI):373.0[M+H] +
Example 3: n- [2- (2-methyl-5-fluorobenzyloxy) phenyl ] nicotinic acid carboxamide
Figure BDA0001877227330000142
Synthesis of N- [ (2-hydroxy) phenyl ] intermediate analogous to example 1]Nicotinic acid formamide reacts with 2-methyl-5-fluorobenzyl bromide to obtain 130mg of a target compound, the yield is 68%, and the appearance is white solid. 1 H NMR(400MHz,Chloroform-d)δ9.04-8.98(m,1H),8.70(dd,J=4.9,1.7Hz,1H),8.63(s,1H),8.56(dd,J=8.0,1.7Hz,1H),8.22(dt,J=8.0,2.0Hz,1H),7.48-7.41(m,3H),7.35-7.31(m,1H),7.31-7.29(m,1H),7.27-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.91(s,1H),5.26(s,2H),2.36(s,3H).MS:m/z(ESI)337[M+1] +
Example 4: n- [2- (2-methoxybenzyloxy) phenyl ] nicotinic acid carboxamide
Figure BDA0001877227330000151
Synthesis procedure analogous to example 1, intermediate N- [ (2-hydroxy) phenyl]Nicotinic acid formamide reacts with 2-methoxy benzyl bromide to obtain 95mg of a target compound, the yield is 55%, and the appearance is white-like solid. 1 H NMR(400MHz,Chloroform-d)δ9.03-8.97(m,1H),8.65(dd,J=4.9,1.7Hz,1H),8.60(s,1H),8.54(dd,J=8.0,1.7Hz,1H),8.26(dt,J=8.0,2.0Hz,1H),7.44-7.37(m,3H),7.33-7.30(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),6.91(d,1H),5.26(s,2H),3.90(s,3H).MS:m/z(ESI)335[M+1] +
Example 5: n- {2- [ (2-ethyl) benzyloxy ] phenyl } nicotinic acid carboxamide
Figure BDA0001877227330000152
Synthesis procedure analogous to example 1, intermediate N- [ (2-hydroxy) phenyl]Nicotinic acid formamide reacts with 2-ethyl benzyl bromide to obtain a target compound of 77mg with yield56% and the appearance is white solid. 1 H NMR(400MHz,Chloroform-d)δ9.03-8.97(m,1H),8.65(dd,J=4.9,1.7Hz,1H),8.60(s,1H),8.54(dd,J=8.0,1.7Hz,1H),8.26(dt,J=8.0,2.0Hz,1H),7.44-7.37(m,3H),7.33-7.30(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),6.91(d,1H),5.26(s,2H),2.71(m,2H),1.18(m,3H).MS:m/z(ESI)333[M+1] +
Example 6: n- {2- [ (2, 6-dimethyl) benzyloxy) ] phenyl } nicotinic acid carboxamide
Figure BDA0001877227330000161
Synthesis procedure analogous to example 1, intermediate N- [ (2-hydroxy) phenyl]Nicotinic acid formamide reacts with 2, 6-dimethyl benzyl bromide to obtain 108mg of a target compound, the yield is 70%, and the appearance is light yellow solid. 1 HNMR(400MHz,Chloroform-d)δ9.03-8.97(m,1H),8.65(dd,J=4.9,1.7Hz,1H),8.60(s,1H),8.54(dd,J=8.0,1.7Hz,1H),8.26(dt,J=8.0,2.0Hz,1H),7.44-7.37(m,3H),7.33-7.30(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),5.26(s,2H),2.29(m,3H),2.26(s,3H).MS:m/z(ESI)333[M+1] +
Example 7: n- {2- [ (2-ethoxy) benzyloxy ] phenyl } nicotinic acid amide
Figure BDA0001877227330000162
Synthesis of N- [ (2-hydroxy) phenyl ] intermediate analogous to example 1]Nicotinic acid formamide reacts with 2-ethoxy bromobenzyl to obtain 50mg of a target compound, the yield is 72%, and the appearance is light yellow solid. 1 HNMR(400MHz,Chloroform-d)δ9.05-8.99(m,1H),8.66(dd,J=4.9,1.7Hz,1H),8.61(s,1H),8.55(dd,J=8.0,1.7Hz,1H),8.27(dt,J=8.0,2.0Hz,1H),7.44-7.37(m,3H),7.33-7.30(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),6.91(d,1H),5.26(s,2H),4.10(m,2H),1.10(m,3H).MS:m/z(ESI)349[M+1] +
Example 8: n- {2- [ (2-methoxy-5-chloro) benzyloxy ] phenyl } nicotinic acid carboxamide
Figure BDA0001877227330000171
By a synthetic method similar to that of example 1, intermediate N- [ (2-hydroxy) phenyl ] nicotinic acid carboxamide and 2-methoxy-5-chlorobenzyl bromide reacted to obtain the objective compound 43mg, yield 83%, appearance as yellow solid.
1 H NMR(400MHz,DMSO-d 6 )δ9.28(s,1H),8.86(d,J=4.6Hz,1H),8.48(d,J=8.2Hz,1H),7.63(s,1H),7.20(d,J=8.8Hz,1H),7.12(s,1H),7.05(d,J=8.0Hz,1H),6.97(d,J=8.7Hz,2H),6.57(t,J=7.7Hz,1H),6.42(d,J=8.2Hz,1H),6.12(s,1H),3.80(d,J=1.8Hz,3H).MS m/z(ESI):369.0[M+H] +
Example 9: n- {2- [ (2, 5-dichloro) benzyloxy ] phenyl } nicotinic acid carboxamide
Figure BDA0001877227330000172
Synthesis procedure analogous to example 1, intermediate N- [ (2-hydroxy) phenyl]Nicotinic acid formamide reacts with 2, 5-dichlorobenzyl bromide to give the target compound 40mg in 53% yield as a grey solid. 1 H NMR(400MHz,Chloroform-d)δ9.09-9.01(m,1H),8.69(dd,J=4.8,1.8Hz,1H),8.64(s,1H),8.55(dd,J=8.0,1.7Hz,1H),8.27(dt,J=8.0,2.0Hz,1H),7.44-7.37(m,3H),7.33-7.30(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(s,1H),5.26(s,2H).MS:m/z(ESI)373[M+1] +
Example 10: n- {2- [ (2-methyl-5-chloro) benzyloxy ] phenyl } nicotinic acid carboxamide
Figure BDA0001877227330000181
Following a synthetic procedure analogous to example 1, intermediate N- [ (2-hydroxy) phenyl ] nicotinic acid carboxamide reacts with 2-methyl-5-chlorobenzyl bromide to give the title compound in 48mg, 79% yield, as a yellow solid in appearance.
1 H NMR(400MHz,DMSO-d 6 )δ9.27(dd,J=2.3,0.9Hz,1H),8.85(dd,J=4.9,1.7Hz,1H),8.47(dt,J=8.0,2.0Hz,1H),7.62(ddd,J=8.0,4.9,0.9Hz,1H),7.16(d,J=2.0Hz,1H),7.13(dd,J=6.7,4.7Hz,2H),7.05(dd,J=7.9,1.5Hz,1H),6.98(td,J=7.8,1.5Hz,1H),6.58(td,J=7.6,1.5Hz,1H),6.43(dd,J=8.2,1.4Hz,1H),6.20(t,J=6.0Hz,1H),4.23(d,J=6.0Hz,2H),2.25(s,3H).MS m/z(ESI):353.1[M+H] +
Example 11: n- {2- [2- (4-chlorobutoxy) benzyloxy ] phenyl } nicotinic acid carboxamide
Figure BDA0001877227330000182
Synthesis procedure analogous to example 1, intermediate N- [ (2-hydroxy) phenyl]Nicotinic acid formamide reacts with 2- (4-chlorobutoxy) bromobenzyl to give the title compound 120mg in 75% yield as a white solid. 1 H NMR(400MHz,Chloroform-d)δ9.04-8.99(m,1H),8.64(dd,J=4.9,1.7Hz,1H),8.60(s,1H),8.53(dd,J=8.0,1.7Hz,1H),8.25(dt,J=8.0,2.0Hz,1H),7.44-7.37(m,3H),7.33-7.30(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),6.91(d,1H),5.06(s,2H),4.10(m,2H),3.60(m,2H),1.78(m,2H),1.75(m,2H).MS:m/z(ESI)411[M+1] +
Example 12: n- {2- [2- (5-chloropentyloxy) benzyloxy ] phenyl } nicotinic acid carboxamide
Figure BDA0001877227330000191
Synthesis procedure analogous to example 1, intermediate N- [ (2-hydroxy) phenyl]Niacinecarboxamide and 2- (5-chloropentyloxy) bromobenzyl were reacted to give the title compound in 25mg, 43% yield, as a white solid in appearance. 1 HNMR(400MHz,Chloroform-d)δ9.04-8.99(m,1H),8.64(dd,J=4.9,1.7Hz,1H),8.60(s,1H),8.53(dd,J=8.0,1.7Hz,1H),8.25(dt,J=8.0,2.0Hz,1H),7.44-7.37(m,3H),7.33-7.30(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),6.91(d,1H),5.06(s,2H),4.10(m,2H),3.60(m,2H),1.78(m,2H),1.75(m,2H),1.29(m,2H).MS:m/z(ESI)425[M+1] +
Example 13: n- {2- [2- (6-chlorohexyloxy) benzyloxy ] phenyl } nicotinic acid carboxamide
Figure BDA0001877227330000192
Following a similar synthetic procedure to example 1, the intermediates N- [ (2-hydroxy) phenyl ] nicotinic acid carboxamide and (2- (6-chlorohexyloxy) bromobenzyl were reacted to give the title compound 44mg in 35% yield as a pale yellow solid.
1 H NMR(400MHz,DMSO-d6)δ9.70(s,1H),9.05(dd,J=2.3,0.8Hz,1H),8.76(dd,J=4.8,1.7Hz,1H),8.24(dt,J=8.0,1.9Hz,1H),7.77(dd,J=7.9,1.6Hz,1H),7.55(ddd,J=7.9,4.8,0.9Hz,1H),7.46(dd,J=7.5,1.7Hz,1H),7.28(ddd,J=8.2,7.4,1.8Hz,1H),7.22–7.12(m,2H),7.05–6.96(m,2H),6.90(td,J=7.5,1.0Hz,1H),5.17(s,2H),3.97(s,2H),3.59(t,J=6.6Hz,2H),1.67(s,4H),1.38(s,4H).MS m/z(ESI):439.2[M+H] +
Example 14: n- [2- (2-hexyloxybenzyloxy) phenyl ] nicotinic acid carboxamide
Figure BDA0001877227330000201
Synthesis of N- [ (2-hydroxy) phenyl ] intermediate analogous to example 1]Nicotinic acid formamide reacts with 2-hexyloxybenzyl bromide to give the target compound 42mg, in 88% yield, as a white solid in appearance. 1 H NMR(400MHz,Chloroform-d)δ9.04-8.99(m,1H),8.64(dd,J=4.9,1.7Hz,1H),8.60(s,1H),8.53(dd,J=8.0,1.7Hz,1H),8.25(dt,J=8.0,2.0Hz,1H),7.44-7.37(m,3H),7.33-7.30(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),6.91(d,1H),4.96(s,2H),4.60(m,2H),1.78(m,2H),1.47(m,2H),1.37(m,4H),0.89(m,2H).MS:m/z(ESI)405[M+1] +
Example 15: n- [2- (2-heptyloxybenzyloxy) phenyl ] nicotinic acid carboxamide
Figure BDA0001877227330000202
By a synthetic method similar to example 1, intermediate N- [ (2-hydroxy) phenyl ] nicotinic acid carboxamide and 2-heptyloxybromobenzyl were reacted to obtain the objective compound 82mg, a yield of 46%, and an appearance as a white solid.
1 H NMR(400MHz,Chloroform-d)δ9.01(d,J=2.3Hz,1H),8.75(dd,J=4.9,1.8Hz,2H),8.55–8.47(m,1H),8.19–8.11(m,1H),7.41(ddd,J=8.0,4.8,0.9Hz,1H),7.38–7.29(m,2H),7.14–7.01(m,3H),7.00–6.90(m,2H),5.23(s,2H),3.95(t,J=6.5Hz,2H),1.68(dd,J=8.2,6.3Hz,2H),1.41–1.13(m,8H),0.85(t,J=6.8Hz,3H).MS m/z(ESI):419.2[M+H] +
Example 16: n- {2- [ (2-hexyloxy) -5-chlorobenzyloxy ] phenyl } nicotinic acid carboxamide
Figure BDA0001877227330000211
Synthesis procedure analogous to example 1, intermediate N- [ (2-hydroxy) phenyl]Nicotinic acid formamide reacts with 2-hexyloxy-5-chlorobromobenzyl to obtain 18mg of a target compound, the yield is 38%, and the appearance is white solid. 1 H NMR(400MHz,Chloroform-d)δ9.04-8.99(m,1H),8.64(dd,J=4.9,1.7Hz,1H),8.60(s,1H),8.53(dd,J=8.0,1.7Hz,1H),8.25(dt,J=8.0,2.0Hz,1H),7.44-7.37(m,3H),7.33-7.30(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),5.03(s,2H),4.64(m,2H),1.79(m,2H),1.49(m,2H),1.38(m,4H),0.91(m,2H).MS:m/z(ESI)439[M+1] +
Example 17: n- {2- [ (2-heptyloxy) -5-chlorobenzyloxy ] phenyl } nicotinic acid amide
Figure BDA0001877227330000212
By a synthetic method similar to example 1, intermediate N- [ (2-hydroxy) phenyl ] nicotinic acid carboxamide and 2-heptyloxy-5-chlorobenzyl bromide were reacted to obtain the objective compound 59mg, yield 45%, appearance white solid.
1 H NMR(400MHz,DMSO-d 6 )δ9.97(s,1H),8.47–8.33(m,2H),7.64(d,J=7.9Hz,1H),7.42(s,1H),7.19(d,J=8.4Hz,2H),6.91(dt,J=18.9,8.4Hz,3H),6.67(d,J=8.2Hz,1H),6.55(t,J=7.7Hz,1H),5.13(d,J=15.4Hz,1H),4.66(d,J=15.3Hz,1H),1.55(s,2H),1.23(d,J=13.0Hz,8H),0.89–0.74(m,3H).MS m/z(ESI):453.1[M+H] +
Example 18: n- {2- [2- (4-chlorobutoxy) benzyloxy ] phenyl } isonicotinic acid carboxamide
Figure BDA0001877227330000221
First step synthesis of N- [ (2-benzyloxy) phenyl ] isonicotinic acid carboxamide
Isonicotinic acid (1.76g,8.83mmol), dichloromethane (30ml), and thionyl chloride (6.75g,56.86mmol) were added to a dried eggplant-shaped flask, and stirred at room temperature for 5 minutes, 2 drops of pyridine were added thereto, and the mixture was refluxed for 3 hours, and the solvent and the remaining thionyl chloride were distilled off under reduced pressure to obtain acid chloride (1.3 g). The crude product was used in the next reaction without purification.
2-Benzyloxyphenylamine (1g,5mmol) was dissolved in dry dichloromethane (30ml), triethylamine (0.76g,7.53mmol) was added and stirred well. Under ice-water bath conditions, a solution of isonicotinic acid formyl chloride (0.9g,6.52mmol) in dichloromethane was slowly added dropwise thereto. After dropping, the reaction was carried out at room temperature for 5 hours. Then the reaction system was washed twice with water, twice with saturated sodium chloride, dried over anhydrous magnesium sulfate, concentrated and subjected to column chromatography to obtain 1.15g of N- [ (2-benzyloxy) phenyl ] isonicotinic acid carboxamide with a yield of 75%.
Second step synthesis of N- [ (2-hydroxy) phenyl ] isonicotinic acid carboxamide
To a solution of N- [ (2-benzyloxy) phenyl ] isonicotinic acid carboxamide (1g,3.29mmol) in methanol (30ml) was added 10% Pd/C (0.1g), followed by reaction at room temperature under a hydrogen pressure of 8MPa for 8 hours. Pd/C was removed by suction filtration, and the filtrate was concentrated to give N- [ (2-hydroxy) phenyl ] isonicotinamide (0.57g) as a product in 81% yield.
The third step is the synthesis of N- {2- [2- (4-chlorobutoxy) benzyloxy ] phenyl } isonicotinic acid formamide
Reacting N- [ (2-hydroxy) phenyl group]Isonicotinic acid carboxamide (0.1g,0.47mmol) was dissolved in DMF (10ml), and potassium carbonate (0.13g,0.93mmol) was added, followed by addition of 2- (4-chlorobutoxy) benzyl bromide (0.15g,0.56mmol), followed by reaction at room temperature overnight. Adding water, extracting with ethyl acetate, washing with saturated salt water, drying with anhydrous magnesium sulfate, concentrating, and performing column chromatography (PE/EA 0-50%) to obtain N- {2- [2- (4-chlorobutoxy) benzyloxy group]Phenyl } isonicotinic acid carboxamide (0.67g), yield 35%. MS M/z (ESI)411[ M +1 ]] +
Example 19: n- {2- [2- (5-chloropentyloxy) benzyloxy ] phenyl } isonicotinic acid carboxamide
Figure BDA0001877227330000231
Synthesis analogous to example 18, intermediate N- [ (2-hydroxy) phenyl]Isonicotinic acid formamide reacted with 2- (5-chloropentyloxy) bromobenzyl to give 59mg of the title compound in 45% yield as a white solid in appearance. MS M/z (ESI)425[ M +1 ]] +
Example 20: n- {2- [ (5-chloro-2-heptyloxybenzyl) amino ] phenyl } nicotinamide
Figure BDA0001877227330000232
First step synthesis of N- (2-nitrophenyl) nicotinic acid formamide
The compound o-nitroaniline (15.0g, 0.1mol) and triethylamine (33.3g, 0.3mol) were dissolved in dichloromethane (150mL), the temperature was reduced to 0 ℃, and the compound nicotinic acid formyl chloride (18.4g, 0.1mol) was slowly added dropwise to the reaction system. After the completion of the dropwise addition, the reaction was carried out at 0 ℃ for 2 hours. TLC showed the reaction was complete, 500mL of water was added, the layers were separated, the organic phase was washed once with hydrochloric acid (1N), once with saturated brine, dried over anhydrous sodium sulfate, spun dry, and the crude compound N- (2-nitrophenyl) nicotinic acid carboxamide (6.0g, 25.0mmol) slurried with methyl tert-butyl ether was used in the next reaction in 22.7% yield.
Second step synthesis of N- (2-aminophenyl) nicotinic acid carboxamide
Raney nickel (1.2g), the compound N- (2-nitrophenyl) nicotinic acid carboxamide (6.0g, 25.0mmol) was added to methanol (60mL) and reacted at room temperature for 10 hours under a hydrogen pressure of 50 Psi. TLC showed the reaction was complete, the reaction was filtered with suction, and the filtrate was spin-dried to give the compound N- (2-aminophenyl) nicotinic acid carboxamide (4.0g, 19.0mmol) in 76.0% yield.
1 H NMR(400MHz,DMSO-d6,ppm):9.83(brs,1H),9.14(s,1H),8.74(d,J=4.8Hz,1H),8.33(d,J=7.6Hz,1H),7.44(dd,J=8Hz J=5.2Hz,2H),7.18(d,J=7.6Hz,1H),7.0-6.96(m,1H),6.79(dd,J=8Hz J=1.2Hz,1H),6.61(t,J=7.2Hz,1H),4.98(brs,2H)。
Step three, synthesizing N- {2- [ (5-chloro-2-heptyloxybenzyl) amino ] phenyl } nicotinamide
Methanol (5mL), the compound N- (2-aminophenyl) nicotinic acid carboxamide (0.50g, 2.3mmol) and 2-heptyloxy-5-chlorobenzaldehyde (1.2g, 4.6mmol) were added to a reaction flask and stirred at room temperature for 0.5 hour, after which NaBH was added to the solution 3 CN (0.7g, 11.5mmol) and 3 drops of acetic acid. The solution was stirred at 50 ℃ overnight. The reaction solution is directly spun to dry the scraper. Soaking with dichloromethane and methanol 10:1, filtering with silica gel scraper, and spin-drying to obtain compound N- {2- [ (5-chloro-2-heptyloxybenzyl) amino]Phenyl } nicotinamide (50.0mg, 0.1mmol), yield was 4.8%.
1 H NMR(400MHz,DMSO-d6,ppm):9.92(s,1H),9.20(s,1H),8.76(d,J=3.6Hz,1H),8.38(d,J=7.6Hz,1H),7.59(dd,J=7.6Hz J=4.8Hz,1H),7.30(d,J=2Hz,1H),7.22(dd,J=8.8Hz J=2.8Hz,1H),7.15(d,J=7.2Hz,1H),7.02-6.99(m,2H),6.62(d,J=7.6Hz,1H),6.40(d,J=8Hz,1H),5.83(brs,1H),4.28(s,2H),4.04(t,J=6Hz,2H),1.76-1.69(m,2H),1.45-1.37(m,2H),1.33-1.14(m,6H),0.87-0.80(m,3H)。MS:m/z(ESI)452[M+1] +
Example 21: n- {2- [ (5-chloro-2- (6-chlorohexyloxy) benzyl) amino ] phenyl } nicotinamide
Figure BDA0001877227330000241
By a synthetic method similar to example 20, intermediates N- (2-aminophenyl) nicotinic acid carboxamide and 2- (6-chlorohexyloxy) -5-chlorobenzaldehyde were reacted to obtain the objective compound 59mg, a yield of 45%, and an appearance of a white solid.
1 H NMR(400MHz,DMSO-d6,ppm):9.91(s,1H),9.19(s,1H),8.76(d,J=3.6Hz,1H),8.38(d,J=8.4Hz,1H),7.59(dd,J=7.6Hz J=4.8Hz,1H),7.29(d,J=7.2Hz,1H),7.19-7.12(m,2H),7.02-6.95(m,2H),6.86(t,J=7.2Hz,1H),6.59(t,J=7.2Hz,1H),6.48(d,J=8Hz,1H),5.72(brs,1H),4.30(s,2H),4.03(t,J=6Hz,2H),3.63(t,J=6.4Hz,2H),1.75-1.68(m,4H),1.50-1.36(m,4H)。MS:m/z(ESI)472[M+1] +
Example 22: n- {2- [ (2-chloro-5-fluorobenzyl) amino ] phenyl } nicotinamide
Figure BDA0001877227330000251
Following a synthetic procedure analogous to example 20, intermediate N- (2-aminophenyl) nicotinic acid carboxamide reacts with 2-chloro-5-fluorobenzaldehyde to give the title compound 33mg in 20% yield as an off-white solid. 1 H NMR(400MHz,DMSO-d6,ppm):9.91(s,1H),9.19(s,1H),8.76(d,J=3.6Hz,1H),8.38(d,J=8.4Hz,1H),7.59(dd,J=7.6Hz J=4.8Hz,1H),7.35(s,1H),7.19-7.12(m,2H),7.02-6.95(m,2H),6.86(t,J=7.2Hz,1H),6.59(t,J=7.2Hz,1H),6.48(d,J=8Hz,1H),4.30(s,2H)。MS:m/z(ESI)356[M+1] +
Example 23: n- {2- [ (2, 6-dichlorobenzyl) amino ] phenyl } nicotinamide
Figure BDA0001877227330000252
By a synthetic method similar to that of example 20, intermediate N- (2-aminophenyl) nicotinic acid carboxamide and 2, 5-dichlorobenzaldehyde reacted to give the objective compound 54mg, 26% yield, pale yellow solid in appearance. 1 H NMR(400MHz,DMSO-d6,ppm):9.87(s,1H),9.19(s,1H),8.75(d,J=3.6Hz,1H),8.38(d,J=8.4Hz,1H),7.59(dd,J=7.6Hz J=4.8Hz,1H),7.29(d,J=7.2Hz,1H),7.19-7.12(m,2H),7.02-6.95(m,2H),6.86(t,J=7.2Hz,1H),6.59(t,J=7.2Hz,1H),6.48(d,J=8Hz,1H),4.30(s,2H)。MS:m/z(ESI)372[M+1] +
Example 24: n- {2- [ (5-fluoro-2-methylbenzyl) amino ] phenyl } nicotinamide
Figure BDA0001877227330000261
By a synthetic method similar to example 20, intermediate N- (2-aminophenyl) nicotinic acid carboxamide and 2-methyl-5-fluorobenzaldehyde were reacted to obtain the objective compound 45mg, yield 22%, and appearance pale yellow solid. 1 H NMR(400MHz,DMSO-d6,ppm):9.87(s,1H),9.19(s,1H),8.75(d,J=3.6Hz,1H),8.38(d,J=8.4Hz,1H),7.59(dd,J=7.6Hz J=4.8Hz,1H),7.29(d,J=7.2Hz,1H),7.19-7.12(m,2H),7.02-6.95(m,2H),6.86(s,1H),6.59(t,J=7.2Hz,1H),6.48(d,J=8Hz,1H),4.30(s,2H),2.29(s,3H)。MS:m/z(ESI)336[M+1] +
Example 25: n- {2- [ (2- (4-chlorobutoxy) benzyl) amino ] phenyl } isonicotinamide
Figure BDA0001877227330000262
By a synthetic method similar to example 20, intermediate N- (2-aminophenyl) isonicotinic acid carboxamide and 4- (chlorobutoxy) benzaldehyde were reacted to obtain the objective compound 27mg, yield 18%, and appearance of pale yellow solid. 1 H NMR(400MHz,Chloroform-d)δ9.06-9.00(m,1H),8.65(dd,J=4.9,1.7Hz,1H),8.62(s,1H),8.55(dd,J=7.9,1.7Hz,1H),8.27(dt,J=7.9,2.0Hz,1H),7.45-7.38(m,3H),7.33-7.30(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),6.91(d,1H),4.45(s,2H),4.08(m,2H),3.63(m,2H),1.77(m,2H),1.74(m,2H).MS:m/z(ESI)410[M+1] +
Example 26: n- {2- [ (2- (5-chloropentyloxy) benzyl) amino ] phenyl } isonicotinamide
Figure BDA0001877227330000271
Following a similar synthetic procedure to example 20, the intermediates N- (2-aminophenyl) isonicotinic acid carboxamide and 2- (5-chloropentyloxy) benzaldehyde were reacted to give the title compound 57mg, 30% yield, as a pale yellow solid in appearance. 1 H NMR(400MHz,Chloroform-d)δ9.05-9.00(m,1H),8.64(dd,J=4.9,1.7Hz,1H),8.60(s,1H),8.53(dd,J=7.9,1.7Hz,1H),8.25(dt,J=7.9,2.0Hz,1H),7.43-7.36(m,3H),7.33-7.31(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),6.91(d,1H),4.45(s,2H),4.08(m,2H),3.63(m,2H),1.77(m,2H),1.74(m,2H),1.39(m,2H).MS:m/z(ESI)424[M+1] +
Example 27: n- {2- [ (2-hexyloxybenzyl) amino ] phenyl } isonicotinamide
Figure BDA0001877227330000272
By a synthetic method similar to that of example 20, intermediate N- (2-aminophenyl) isonicotinic acid carboxamide and 2- (hexyloxy) benzaldehyde reacted to obtain the objective compound 31mg, yield 38%, appearance yellow solid. 1 H NMR(400MHz,Chloroform-d)δ9.08-9.03(m,1H),8.67(dd,J=4.9,1.7Hz,1H),8.63(s,1H),8.56(dd,J=7.9,1.7Hz,1H),8.28(dt,J=7.9,2.0Hz,1H),7.43-7.36(m,3H),7.33-7.31(m,1H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),6.99(m,1H),6.91(d,1H),4.40(s,2H),4.06(m,2H),1.75(m,2H),1.46(m,2H),1.39(m,4H),0.88(t,3H).MS:m/z(ESI)404[M+1] +
Example 28: n- {2- [ (5-chloro-2-heptyloxybenzyl) amino ] phenyl } benzamide
Figure BDA0001877227330000281
First step synthesis of N- (2-nitrophenyl) benzamide
The compound o-nitroaniline (5.9g, 0.043mol) and triethylamine (6.5g, 0.064mol) were dissolved in dichloromethane (50mL), the temperature was reduced to 0 ℃, and a solution of the compound benzoyl chloride (6g, 0.043mol) in dichloromethane (15mL) was slowly added dropwise to the reaction system. After the completion of the dropwise addition, the reaction was carried out at 0 ℃ for 2 hours. TLC showed that the reaction was complete, 500mL of water was added, the solution was separated, and the organic phase was washed once with hydrochloric acid (1N), once with saturated brine, dried over anhydrous sodium sulfate, spin-dried, and subjected to column chromatography to give N- (2-nitrophenyl) benzamide (5.3g) in 51.2% yield.
Second step synthesis of N- (2-aminophenyl) benzamide
Raney nickel (1.2g), the compound N- (2-nitrophenyl) benzamide (5.0g, 20.6mmol) was added to methanol (60mL) and reacted at room temperature for 10 hours under a hydrogen pressure of 50 Psi. TLC showed the reaction was complete, the reaction was filtered with suction and the filtrate was spin-dried to give the compound N- (2-aminophenyl) benzamide (3.8g, 17.9mmol) in 86.74% yield.
Step three, synthesizing N- {2- [ (5-chloro-2-heptyloxybenzyl) amino ] phenyl } benzamide
Methanol (5mL), the compound N- (2-aminophenyl) benzamide (0.50g, 2.36mmol) and 2-heptyloxy-5-chlorobenzaldehyde (1.2g, 4.7mmol) were added to a reaction flask and stirred at room temperature for 0.5 hour, after which NaBH was added to the solution 3 CN (0.74g, 11.5mmol) and 3 drops of acetic acid. The solution was stirred at 50 ℃ overnight. The reaction solution is directly spun to dry the scraper.
Soaking with dichloromethane and methanol 10:1, filtering with silica gel scraper, and spin-drying to obtain compound N- {2- [ (5-chloro-2-heptyloxybenzyl) amino]Phenyl } benzamide (100mg) in 4.7% yield. 1 H NMR(400MHz,Chloroform-d)δ9.08-9.03(m,1H),8.67(dd,J=4.9,1.7Hz,1H),8.63(s,1H),8.56(dd,J=7.9,1.7Hz,1H),8.28(dt,J=7.9,2.0Hz,1H),7.43-7.36(m,3H),7.33-7.31(m,2H),7.29-7.27(m,1H),7.26-7.23(m,1H),7.19(td,J=7.6,1.6Hz,1H),7.01(s,1H),4.40(s,2H),4.09(m,2H),1.78(m,2H),1.48(m,2H),1.26(m,6H),,0.90(t,3H).MS:m/z(ESI)451[M+1] +
Example 29: n- {2- [ (6-chlorohexyloxybenzyl) amino ] phenyl } nicotinamide
Figure BDA0001877227330000291
Following a synthetic procedure analogous to example 20, the intermediates N- (2-aminophenyl) nicotinic acid carboxamide and 2- (6-chlorohexyloxy) benzaldehyde were reacted to give the title compound 33mg, 58% yield, as an externally white solid. 1 H NMR(400MHz,DMSO-d 6 )δ9.92(s,1H),9.19(d,J=2.2Hz,1H),8.76(dd,J=4.8,1.6Hz,1H),8.36(dt,J=8.2,1.9Hz,1H),7.56(dd,J=8.0,4.9Hz,1H),7.29(dd,J=7.6,1.7Hz,1H),7.22–7.09(m,2H),7.07–6.92(m,2H),6.90–6.78(m,1H),6.58(td,J=7.5,1.3Hz,1H),6.47(dd,J=8.2,1.3Hz,1H),5.72(t,J=6.3Hz,1H),4.31(d,J=6.1Hz,2H),4.01(t,J=6.3Hz,2H),3.62(t,J=6.6Hz,2H),1.72(p,J=6.9Hz,4H),1.45(td,J=9.7,8.4,5.2Hz,4H).MS m/z(ESI):438.2[M+H] +
Example 30: n- {2- [2- (6-chlorohexyloxy) benzyloxy ] phenyl } isonicotinic acid carboxamide
Figure BDA0001877227330000292
By a synthetic method similar to example 18, intermediate N- [ (2-hydroxy) phenyl ] isonicotinic acid carboxamide and 2- (6-chlorohexyloxy) bromobenzyl were reacted to obtain the objective compound 30mg in a yield of 29%, as an appearance white solid.
1 H NMR(400MHz,DMSO-d6)δ9.77(s,1H),8.80–8.74(m,2H),7.83–7.71(m,3H),7.45(dd,J=7.5,1.7Hz,1H),7.32–7.11(m,3H),7.05–6.96(m,2H),6.90(td,J=7.5,1.0Hz,1H),5.16(s,2H),3.97(t,J=6.3Hz,2H),3.59(t,J=6.6Hz,2H),1.66(dt,J=11.0,5.9Hz,4H),1.38(p,J=3.6Hz,4H).MS m/z(ESI):439.1[M+H] +
Example 31: n- {2- [2- (heptyloxy) benzyloxy ] phenyl } isonicotinic acid carboxamide
Figure BDA0001877227330000301
By a synthetic method similar to example 18, intermediate N- [ (2-hydroxy) phenyl ] isonicotinic acid carboxamide and 2-heptyloxybromobenzyl were reacted to obtain the objective compound 55mg, yield 40%, appearance white solid.
1 H NMR(400MHz,DMSO-d6)δ9.76(s,1H),8.82–8.72(m,2H),7.77(dd,J=17.0,6.5Hz,3H),7.44(d,J=7.5Hz,1H),7.27(t,J=7.8Hz,1H),7.22–7.10(m,2H),7.00(dd,J=10.4,7.9Hz,2H),6.90(t,J=7.5Hz,1H),5.15(s,2H),3.96(t,J=6.4Hz,2H),1.71–1.57(m,2H),1.28(d,J=52.1Hz,8H),0.83(t,J=6.5Hz,3H).MS m/z(ESI):419.2[M+H] +
Biological experimental example:
in each of the experimental examples described below, the target compounds prepared in examples 1 to 31 were referred to as example compounds 1 to 31, respectively.
Experimental example one inhibition of sphingomyelin synthase (SMS) Activity by Compounds of the invention
1. Purpose of the experiment
This experiment measured the half-inhibitory rate (IC50) of the inhibitory effect of the compounds provided by the present invention on sphingomyelin synthase (SMS) activity.
2. Experimental methods
The half inhibition rate of the compound provided by the invention under an optimized enzyme reaction system is determined by using an ultra-high performance liquid chromatography tandem mass spectrometry method in combination with an enzyme inhibitor activity test method.
2.1 chromatographic conditions A C18 column (Waters) was used and eluted with a gradient of 0.50mL/min using aqueous (A) -0.1% formic acid and acetonitrile (B) -0.1% formic acid as mobile phases.
2.2 the mass spectrum condition adopts an electrospray ionization source (ESI) positive ion mode, a multi-reaction detection mode (MRM) C6-ceramide and C6-sphingomyelin ion pair are 398.4/380.4 and 704.7/184 respectively, and a detection ion pair of the internal standard diclofenac is 296/215.
2.3 preparation of Standard Curve
A certain amount of C6-SM (C6-sphingomyelin, Sigma, cat #77238) and C6-Ceramide (C6-Ceramide, Santa Cruze, cat # sc-3527) were respectively weighed and dissolved in ethanol to obtain a 1mg/mL stock solution, and then diluted with 70% acetonitrile aqueous solution according to a gradient to obtain 60, 20, 6, 2, 0.6, 0.2, 0.06, 0.02 and 0.006. mu.g/mL working solutions, 10. mu.L of the working solution was added to 190. mu.L of SMS reaction solution, 800. mu.L of 50% acetonitrile methanol containing an internal standard (diclofenac, 100ng/mL) was added at a ratio of 1:4, 13000 rpm was carried out at room temperature, centrifugation was carried out for 15 minutes, and 200. mu.L of the centrifuged supernatant was taken to obtain a final concentration of 3000, 1000, 300, 100, 30, 10, 3, 1 and 0.3 ng/mL.
2.4 preparation of the Compounds
The stock solutions of the example compounds 1 to 31 and D609, dissolved in dimethyl sulfoxide to 10mM, were weighed and gradually diluted in a gradient to test concentrations of 1000uM, 333uM, 111uM, 33uM, 11uM, 3uM, 1uM, 0.3uM, 0.1uM and 0.03uM, each in duplicate wells.
2.5 enzymatic reactions
The SMS enzyme reaction consisted of 50nM Tris-HCL (Invitrogen, 10010023), 25nM KCL (Sigma, 746436) and 0.5nM EDTA (Sigma, EDS-100G).
Taking a proper amount of liver of a fresh male ICR mouse (20 g of Shanghai Semipril-BiKai laboratory animal Co., Ltd.), adding a proper amount of phosphate buffer solution (Invitrogen, 10010023) to prepare a tissue fluid of 300mg/mL, carrying out ice bath homogenization, carrying out 13000 revolutions per minute, centrifuging for 15 minutes to obtain a liver homogenate supernatant, taking a kit standard product (Sigma, 1002254738) and diluting the kit standard product to 1000 mu g/mL, 800 mu g/mL, 600 mu g/mL, 400 mu g/mL, 200 mu g/mL, 100 mu g/mL, 50 mu g/mL, 25 mu g/mL to obtain a detection standard curve, and carrying out detection on a fluid A and a fluid B by a reaction condition that the ratio of 1: mixing the materials in a volume ratio of 50 to obtain a protein quantitative working solution, sequentially adding 20 mu L of the protein quantitative working solution to corresponding vacant sites from low to high according to the concentration of a standard curve, sequentially adding 200 mu L of enzyme reaction working solution, and lightly shaking up; after reacting for 30min at 37 ℃, detecting the light absorption value at 562nm, and finally quantifying the protein stock solution to 400 mg/mL.
Adding 76 mu L of SMS enzyme reaction liquid into corresponding hole sites, respectively adding 2 mu L C6-Ceramide and C6-SM, then adding 10 mu L of 40 mu g/ml liver homogenate and 10 mu L D609 solution or compound solution, shaking up gently, reacting at 37 ℃ for 30min, adding 400 mu L of stop solution to stop enzyme reaction, centrifuging at 13000 r/min and room temperature for 15min to obtain supernatant, and analyzing the sample by LC/MS.
3. Results of the experiment
The compound provided by the invention has better effect of inhibiting the activity of sphingomyelin synthase, has 50 percent of inhibitory concentration (IC50) <50 mu M for the sphingomyelin synthase, and has better effect than D609 (see Table 1 below), wherein D609 is a sphingomyelin synthase inhibitor reported in the literature (Aimin Meng; Chiara Luberto; et al. Experimental Cell Research, 2004, 292, 385-392.).
TABLE 1 inhibitory Effect of example Compounds 1-31 and Positive control D609 on sphingomyelin synthase Activity
Figure BDA0001877227330000321
Figure BDA0001877227330000331
Experimental example II Effect of the Compound of the present invention on inhibition of sphingomyelin synthase (SMS) Activity
1. Purpose of experiment
This experiment examined the effect of the compounds of the invention on SMS activity in U937 cells.
1.1 cell culture
Human U937 cells (ATCC) were cultured normally, and the complete medium included L-glutamine-containing RPMI 1640 medium (corning, 21517002), 10% FBS (Ausbina, 0986180), 100U/mlP/S (Gibco, 1902422). In the experiment, cells are incubated with different concentrations of solvents (DMSO), D609 and the test compound for 2h and then subjected to lysis treatment.
1.2 preparation of the Compounds
Compounds were dissolved in DMSO and diluted in 3-fold gradient at concentrations of 810. mu.M, 270. mu.M, 90. mu.M, 30. mu.M, 10. mu.M, 3. mu.M, respectively.
1.3 reaction scheme
After the cells were rinsed 2 times with phosphate buffer, homogenization was performed with ice-bath lysate. The lysate comprises 25mM sucrose (Sigma, 57-50-1), 5mM HEPES (Sigma, 7365-45-9), 1mM phenylmethylsulfonyl fluoride (Sigma, 52332), 20. mu.g/mL chymotrypsin inhibitor, leupeptin, antipain and pepstatin (Sigma, 11206893001), and the supernatant obtained is the cell protein solution after centrifugation at 4 ℃ and 1000 Xg for 10 min. The supernatant was subjected to protein quantification by means of a Bio-Rad kit (Sigma, 1002254738). The protein concentration was 1mg/mL at the time of reaction.
The SMS enzyme reaction consisted of 50nM Tris-HCL (Invitrogen, 10010023), 25nM KCL (Sigma, 746436), 0.5nM EDTA (Gibco, 1947027).
The above SMS enzyme reaction mixture (76. mu.L) was added with 2. mu.L of 6-Cermide (Santa, sc-3572), 10. mu.L of the above cell protein solution and 10. mu.L of the compound solutions of different concentrations, and shaken well. Reacting at room temperature for 30min, adding 400 μ L of stop solution to stop reaction, centrifuging at 13000 rpm for 15min to obtain supernatant, and analyzing sample by LC/MS to determine SMS content.
1.4LC/MS reaction conditions
1.4.1LC/MS conditions
The column was a C18 column (Waters), mobile phase A was a 0.1% formic acid in water and B was a 0.1% formic acid in acetonitrile, the flow rate was 0.5mL/min and the autosampler temperature was 4 ℃.
Mobile phase gradient conditions were as follows:
time (minutes) A(%) B(%)
Initiation of 50 50
2.00 2 98
2.20 2 98
2.41 50 50
3.00 50 50
Mass spectrum conditions:
ionization mode Electrospray ion source
Scanning mode Positive ion
Scanning mode Multiple reaction monitoring
Air curtain gas (psi) 35
Spray mist (psi) 60
Auxiliary heating (psi) 60
Ionization voltage (V) 5500
Temperature (. degree.C.) 550.0
The ion pair information is as follows:
Figure BDA0001877227330000351
1.4.2 Standard Curve preparation
Respectively weighing appropriate amounts of C6-SM and C6-Ceramide, and dissolving in ethanol to obtain 1mg/mL stock solution; dilutions were made using a 70% acetonitrile water gradient to give 60, 20, 6, 2, 0.6, 0.2, 0.06, 0.02 and 0.006 μ g/mL of working solution. mu.L of the above working solution was added to 190. mu.L of the enzyme reaction working solution at a ratio of 1:4 to a volume of 800. mu.L of 50% acetonitrile methanol containing the internal standard, 13000 rpm, 15 minutes of centrifugation, 200. mu.L of supernatant to a 96 well plate, 4000 rpm for 15 minutes, at final concentrations of 3000, 1000, 300, 100, 30, 10, 3, 1 and 0.3 ng/mL.
2. Results of the experiment
The test compounds dose-dependently reduced SMS activity. EC of test Compound 50 The value is less than 50. mu.M, better than D609.
TABLE 2 inhibitory Activity of Compounds on SMS EC 50 Value of
Compound (I) EC 50 (uM)
EXAMPLES Compound 1 9
EXAMPLE Compound 4 21
EXAMPLE Compound 5 18
Example Compound 7 16
EXAMPLE Compound 16 35
EXAMPLE Compound 17 40
EXAMPLE Compound 18 32
Example Compound 20 20
Example Compound 24 45
Example Compound 29 15
D609 90
Third Experimental example Effect of the Compounds of the present invention on lipid metabolism
1. Experimental objects and methods
This experiment examined the effect of the compounds of the invention on the lipid metabolism of HepG2 cells.
a) Cell culture
HepG2 cells were purchased fromCell banks of the academy of sciences of the sea china (shanghai hanbo biotechnology). The cells were cultured in DMEM low-sugar medium (Gibco, 21885108) containing 10% fetal bovine serum (Ausbina, 0986180), 1% penicillin/streptomycin (Gibco, 1902422). Placing at 37 deg.C and 5% CO 2 Culturing in an incubator. The experiment was carried out at 1X 10 6 Density was seeded in 6-well plates. After the cells are attached to the wall for 24 hours, DMEM without FBS is replaced for culture, 0.5mM Free Fatty Acid (FFA) and compounds or PBS with different concentrations are added, and after 24 hours of action, detection is carried out by a TG kit.
Materials used for the experiments included: oil red (Sigma, 1320-06-5), TG kit (Nanjing Kangkui, A110-1).
b) Compound configuration
A certain amount of the compound was weighed and dissolved in dimethyl sulfoxide to prepare solutions having concentrations of 0.5mg/mL, 1mg/mL, 2mg/mL, 5mg/mL, and 10mg/mL, respectively, for experiments.
2. Results of the experiment
Phosphate buffer treated cells showed significant lipid deposition, while compound treatment significantly reduced lipid deposition, showing that the compounds of the present invention were able to dose-dependently reduce TG (triglyceride) levels in HepG2 cells.
TABLE 3 EC of Compounds on TG levels 50 Value of
Compound (I) EC 50 (uM)
EXAMPLES Compound 1 0.08
EXAMPLE Compound 4 0.06
EXAMPLES Compounds5 0.15
Example Compound 7 0.09
EXAMPLE Compound 17 0.19
Example Compound 20 0.21
Example Compound 29 0.16
D609 0.83
Experimental example four study on the pharmacological Effect of the Compound of the present invention on Apolipoprotein E Gene knock-out mice (ApoE-/-)
1. Objects and methods of the experiments
This experiment investigated the SM-lowering effect, lipid-lowering effect, and atherosclerosis-inhibiting effect of the compound of the present invention on apolipoprotein E gene-knock-out mice (ApoE-/-).
1.1 animals
At the age of 7-8 weeks, the apolipoprotein E gene knockout mice (ApoE-/-, nanjing model animal center) were randomly divided into a vehicle group, an example compound group, and a positive drug simvastatin group according to initial body weight and initial blood lipid level values, and the number of animals in each group was 8. Animals were given a cholesterol diet feed (0.15% cholesterol, D12079B, test diet).
1.2 Compounds
The compounds of the examples were administered at a dose of 15mg/kg and dissolved in 0.5% CMCNa (avastin) solution to form a homogeneous suspension. Simvastatin is administered at a dose of 40mg/kg in a volume of 10ml/kg by gavage twice a day or with vehicle at intervals of at least 6 hours. The blood fat is detected for 12 weeks by taking blood 1 time per week: total Cholesterol (TC), Triglycerides (TG), high density lipoprotein-cholesterol (HDL-C), low density lipoprotein-cholesterol (LDL-C). The end-point animals were euthanized and liver homogenates were taken and SM levels were determined as in example 1. Pathological changes of the aortic wall are observed.
2. Results of the experiment
Animals administered 15mg/kg of the example compound reduced total cholesterol values, low density lipoprotein values, reduced SM values and reduced plaque formation for 12 weeks; among them, the compounds of examples significantly reduced the SM value to a degree superior to that of the simvastatin group. The results show that the compound provided by the invention has obvious drug effects of reducing SM, reducing lipid and reducing atheromatous plaque formation.
TABLE 4 pharmacodynamic study of Compounds on lipid lowering and SM inhibition in ApoE-/-mice
Figure BDA0001877227330000381
Figure BDA0001877227330000391
Experimental example five study on drug effect of the compound of the invention on atherosclerotic rats
1. Experimental objects and methods
This experiment examined the lipid-lowering effect of the compounds of the present invention on atherosclerotic rats.
a) Laboratory animal
At 7-8 weeks of age, 50 male SD rats (shanghai sipel-bikeka experimental animal center) were randomly divided into a model group, an example compound group (15mg/kg) and a simvastatin group (20mg/kg), and the number of animals per group was 10. Animals were fed high fat diet and except for the model group, animals were dosed starting at week 4 and continuing for 6 weeks. High fat diet (TD.88137, Shanghai Saibo Biotechnology Co., Ltd.).
b) Preparation of the Compounds
The compound was dissolved in 0.5% CMCNa (alatin) to form a homogeneous suspension and administered by gavage in a volume of 2ml/kg 2 times daily.
1.3 Experimental procedures
Before the experiment begins, before the administration of the medicine at the 4 th week, blood is taken at the end of the experiment, and serum is separated to measure four items of blood fat: total Cholesterol (TC), Triglycerides (TG), high density lipoprotein-cholesterol (HDL-C), low density lipoprotein-cholesterol (LDL-C), observed for pathological changes in the aortic wall.
2. Results of the experiment
Compared with a model group, the compound group provided by the invention has the advantages that the triglyceride, the total cholesterol and the low-density lipoprotein-cholesterol of animals are obviously reduced, and the arterial plaque is reduced. The aortic intima does not thicken significantly under the light mirror, the arrangement of smooth muscle cells is regular, but mild edema can be seen. The results show that the compound provided by the invention has the function of regulating blood fat and has a remarkable effect on treating atherosclerosis.
TABLE 5 results of the drug effect of the compounds on the hypertriglyceridemia model rat
Figure BDA0001877227330000401
Experimental example six pharmacodynamic study of the compound of the invention on atherosclerotic rabbits
1. Objects and methods of the experiments
The purpose of this experiment was to test the lipid-lowering effect of the compounds provided by the present invention on atherosclerotic rabbits. Reference is made to the relevant literature (university of Shanxi medical school, 10 months 2003, 34 (5)).
High cholesterol feed (containing 0.5% cholesterol, 5% lard, 15% egg yolk powder, and basal feed) was custom made from shanghai sailpoise biotechnology limited. New Zealand rabbits (Shanghai laboratory animal center) were randomly divided into a normal control group, a model group, an example compound group (10mg/kg) and a simvastatin group (10mg/kg), and the number of animals per group was 8. The normal control group was fed with normal feed and injected with normal saline. Bovine serum albumin was injected and all other animals were fed high cholesterol diet. The rabbits were dosed with drug starting at week 13 and continuing for 26 weeks.
Before the experiment is started, before the administration of the medicine at the 13 th week, blood is taken at the end of the experiment, and serum is separated to measure four items of blood fat: total Cholesterol (TC), Triglyceride (TG), high density lipoprotein-cholesterol (HDL-C), low density lipoprotein-cholesterol (LDL-C), inflammatory factor index, and observing pathological changes of aortic wall.
2. Results of the experiment
Compared with a model group, the animal triglyceride and low density lipoprotein-cholesterol of the compound group of 15mg/kg are obviously reduced, and the total cholesterol content is obviously reduced. The aortic intima under the light microscope has no obvious thickening, and the arrangement of smooth muscle cells is more regular. The results show that the compound of the examples has the function of regulating blood lipid. Has good therapeutic effect on atherosclerosis.
TABLE 6 efficacy results of Compounds on atherosclerosis model rabbits
Figure BDA0001877227330000411
Figure BDA0001877227330000421
EXAMPLE seventhly, the inhibitory effect of the Compound of the present invention on the Rapid delayed rectifier Potassium channel Gene (hERG) Current
1. Purpose of the experiment
Preliminary cardiac safety of the compounds of the invention was investigated using electrophysiological manual patch clamp measurements of the effects of example compound 1, example compound 4, example compound 5, example compound 7, example compound 16, example compound 17, example compound 18, example compound 20, and example compound 29 on the hERG potassium channel.
2. Experimental protocol
2.1 preparation of the Compounds
Test compounds were dissolved in DMSO and formulated as 10, 3.3, 1.1, 0.37mM stock solutions. Following secondary dilutions using extracellular fluid, final test solutions were 30, 10, 3.3, 1.1, 0.37 μ M.
2.2 reagent preparation
Extracellular fluid: 130mM NaCl, 4mM KCl, 1.8mM CaCl 2 、1mM MgCl 2 10mM glucose and 10mM HEPES (pH 7.4).
Intracellular fluid: 130mM KCl, 1mM MgCl 2 5mM EGTA, 5mM MgATP and 10mM MgATP HEPES (pH 7.2)
Cell culture medium composition: DMEM (Gibco, 11330032), 15% fetal bovine serum (PAA, A15-101), 1% penicillin-streptomycin (Biowest, L0022-100)2.3 electrophysiological manual patch clamp system protocol
HEK293 cells overexpressing the hERG potassium channel (from Mohamed Boutjdir Boutzs laboratory, university of New York university) were cultured in DMEM/15% fetal bovine serum/1% penicillin-streptomycin medium at 37 ℃ in 5% CO 2 An incubator. During experiment, the cells are transferred into a cell bath embedded into an inverted microscope platform, extracellular fluid is perfused, and the experiment can be started after the cells are settled after stabilization for 5 minutes. Membrane currents were recorded using a HEKA EPC-10 patch clamp amplifier and PATCHMASTER acquisition system (HEKA instruments, D-67466Lambrcht, Pfalz, Germany). All experiments were performed at room temperature (22-24 ℃). A P-97 microelectrode drawing machine (Sun Instrument Company, One Digital Drive, Novato, CA 94949) was used to straighten the electrodes (BF150-110-10) during the experiments. The inner diameter of the electrode is 1-1.5mm, and the water inlet resistance after the electrode is filled with the internal liquid is 2-4M omega.
The experiment was performed in a whole-cell recording mode, and the current values were recorded according to the electrophysiological stimulation protocol below. The membrane voltage was first clamped at-80 mV, cells were given 2s, +20mV voltage stimulation, activation of hERG potassium channel, repolarization to-50 mV, lasting 5s, generation of an outward tail current, stimulation frequency once every 15 s. The current value is the peak value of the tail current. Channel currents were recorded in the experiment using a whole-cell recording mode. Extracellular fluid (approximately 2ml per minute) was first perfused and continuously recorded, waiting for the current to stabilize (current decay (Run-Down) less than 5% in 5 minutes) at which time the tail current peak was the control current value. And then, perfusing the extracellular fluid containing the drug to be detected and continuously recording until the experiment result of the drug on the hERG current is made to reach a stable state, wherein the tail current peak value is the current value after the drug is added. After reaching a stable state, perfusion testing can continue for other concentrations or drugs if the hERG current returns after the perfusion wash with extracellular fluid. And (3) using a solution containing the drug to be detected, performing perfusion and recording current values in the sequence of low concentration to high concentration. Data collection was performed using PATCHMASTER V2X60(HEKA instruments, D-67466 Lambrright, Pfallz, Germany), and analysis and statistics were performed using Origin 8.5(Origin Lab, Northampton, Mass.) software.
3. Results
The compounds of the invention on hERG current inhibition of IC50 is shown in the following table. As can be seen from the data in Table 7 below, the compounds of the present invention have a weak current inhibitory effect on hERG and are highly safe.
TABLE 7 IC50 values for the current inhibition of hERG by the compounds of the invention
Compound (I) IC50(uM)
EXAMPLES Compound 1 >30
EXAMPLE Compound 4 >30
EXAMPLE Compound 5 >30
EXAMPLE Compound 7 >30
EXAMPLE Compound 16 >30
EXAMPLE Compound 17 >30
EXAMPLE Compound 18 >30
Example Compound 20 >30
Example Compound 29 >30
Experimental example eight pharmacokinetic experiment of the Compound of the present invention
1. Purpose of experiment
Using 200-250g, 8-week-old male SD rats (shanghai sipel-bikeka center) as experimental animals, the drug concentrations in plasma at various times after intravenous and oral administration of example compound 1, example compound 4, example compound 5, example compound 7, example compound 16, example compound 17, example compound 18, example compound 20, example compound 24, and example compound 29 were determined using high performance liquid chromatography tandem mass spectrometry. The pharmacokinetic behavior of the compounds of the invention in rats was studied and their pharmacokinetic profile was evaluated.
2. Experimental protocol
2.1 Compounds for experiments
Example compound 1, example compound 4, example compound 5, example compound 7, example compound 16, example compound 17, example compound 18, example compound 20, example compound 29.
2.2 preparation of the Compounds
A certain amount of the compound was weighed and dissolved in dimethylsulfoxide/polyethylene glycol 400/water 10/10/80 to prepare a homogeneous solution.
2.3 Experimental procedures
Rats were administered the compound of the above example intravenously and blood was collected from the orbit at 0.2mL before and after administration at 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 12.0, 24.0 hours, placed in an anticoagulation tube, centrifuged at 6000 rpm for 10 minutes at 4 ℃ and plasma was isolated and stored at-80 ℃.
50 mu L of the plasma at different times is taken, 150 mu L of acetonitrile solution containing 200ng/mL tolbutamide (avastin, T129578) is added for mixing and shaking for 5 minutes, after centrifugation is carried out for 10 minutes at 12000 r/min, 100 mu L of supernatant is taken out and mixed with 200 mu L of deionized water, and then the mixture is subjected to liquid chromatography-tandem mass spectrometry sample injection analysis.
2.4 liquid chromatography conditions and analytical software
The liquid chromatography system is LC-20AD UFLC high performance liquid chromatography system (Shimadzu, LC-20 AD). The mass spectrometry system was equipped with an electrospray ionization source (ESI) for AB Sciex API4000 three-stage quadrupole mass spectrometry (Applied Biosystems, Canada). The software used to control the LC-MS and quantitative analysis was analysis 1.6(Applied Biosystems, Canada), and pharmacokinetic parameters were analyzed using WinNonlin (version 5.2, Pharsight, Mountain View, Calif.) non-compartmental model.
The liquid chromatography was performed using a Synergi Fusion-RP C18 column (50X 2.0mm, 4 μm inner diameter). The column temperature was maintained at room temperature. The composition and gradient of the mobile phase and the mass spectrometry conditions are respectively given in the following table:
liquid phase conditions of test compounds:
Figure BDA0001877227330000451
Figure BDA0001877227330000461
the mass spectrometry conditions for the test compound and internal standard are given in the following table:
Figure BDA0001877227330000462
2.5 preparation of Standard Curve and quality control samples
The test compound was dissolved in dimethyl sulfoxide to make a stock solution with a concentration of 1mg/mL, diluted with 50% acetonitrile to give a series of standard working solutions with concentrations of 10, 3, 1, 0.3, 0.1, 0.03 and 0.01. mu.g/mL, and a series of standard quality control solutions (8, 0.5 and 0.03. mu.g/mL). mu.L of the standard solution and 45. mu.L of the blank plasma matrix were mixed well to give standard solutions (1000, 500, 200, 100, 10, 5, 2 and 1ng/mL) and quality control standard solutions (800, 50 and 3ng/mL) at each concentration point of the standard curve.
Dissolving the internal standard tolbutamide solid powder in dimethyl sulfoxide to prepare 1mg/mL stock solution. The stock solution was diluted with 100% acetonitrile to give a 200ng/mL solution as protein precipitant.
2.6 results of the experiment
The pharmacokinetic parameters of the compounds of the invention are shown in table 8 below:
TABLE 8 pharmacokinetic parameters of the Compounds of the invention
Figure BDA0001877227330000463
Figure BDA0001877227330000471
And (4) conclusion: the compound has better pharmacokinetic absorption, longer half-life period in vivo and better pharmacokinetic characteristic.
EXAMPLE ninth acute toxicity test of Compound of the present invention
1. Purpose of experiment
Male ICR mice (shanghai sipel-bikeka center) of 20-22g, 8 weeks old, were orally administered with 10mg/kg, 30mg/kg, 90mg/kg, 270mg/kg, 810mg/kg, 2430mg/kg of the example compound 1 time for 14 consecutive days including clinical observation, body weight and pathological examination.
2. Experimental protocol
2.1 Compounds for experiments
Example compound 1, example compound 4, example compound 5, example compound 7, example compound 16, example compound 17, example compound 18, example compound 20, example compound 29.
2.2 preparation of the Compounds
A certain amount of the compound of the examples was weighed and dissolved in 0.5% CMCNa to prepare a homogeneous solution.
2.3 Experimental procedures
Mice were observed for acute toxicity following administration of the example compounds by the top-bottom method, 10 mice per dose group, and half each. The administration volume was 10mL/kg and 1 administration was performed.
2.4 results of the experiment
All animals were observed for 14 consecutive days after administration and showed no other abnormal manifestations. The weight of the animals in each group is slightly reduced on the 2 nd day after the administration, but the difference with the control group is not significant. After the 14-day observation period, the general anatomy examination is carried out after all animals are euthanized, no obvious abnormality is seen on the body surface, and no macroscopic lesions are seen in the thoracic cavity, abdominal cavity, pelvic cavity and cranial cavity.
Under the experimental conditions, the median lethal dose of the compound of the invention after oral administration is more than 2000 mg/kg. The safety is better.
Experimental example ten Long-term toxicity test of the Compound of the present invention
1. Purpose of experiment
180-200g of male SD rats (Shanghai Sphere-BikeKa center) aged 7-8 weeks are used as experimental animals to determine the possible toxic effects of the compounds provided by the invention after long-term administration to the SD rats.
2. Experimental protocol
2.1 Compounds for experiments
Example compound 1, example compound 4, example compound 5, example compound 7, example compound 16, example compound 17, example compound 18, example compound 20, example compound 29.
2.2 preparation of the Compounds
A certain amount of the compound was weighed and dissolved in 0.5% CMCNa to prepare a homogeneous solution.
2.3 Experimental procedures
SD rats were given varying doses of 30, 90, 270, 540mg/kg of the compound provided herein once a day for 15 days. Animals were 10 animals per group, male and female halves. The animals were observed daily conditions, body weight changes, diet, appearance, behavior, etc. daily. Animals were dissected at the end point, blood biochemical markers were measured, and organs were removed for histopathological examination.
2.4 results of the experiment
After administration and continuous observation for 15 days, all animals have no other abnormal manifestations, and after the 15-day observation period, all animals are euthanized and subjected to general anatomical examination, no obvious abnormality is seen on the body surface, and no macroscopic lesions are seen in the thoracic cavity, abdominal cavity, pelvic cavity and cranial cavity.
It can be seen that the compounds of the present invention are safe and the dosage at which no adverse effects are observed (NOAEL) is 540 mg/kg.
The sphingomyelin synthase inhibitors provided by the invention, the preparation method and the application thereof are described in detail above. The principles and embodiments of the present invention have been described herein using specific examples, which are presented only to assist in understanding the method and its central concept. It should be noted that it would be apparent to those skilled in the art that various changes and modifications can be made in the invention without departing from the principles of the invention, and such changes and modifications are intended to be covered by the appended claims.

Claims (7)

1. A compound of formula (I), a pharmaceutically acceptable salt or a stereoisomer thereof:
Figure FDA0003794381630000011
wherein X and Z are independently selected from-CH-or-N-, and X and Z are not-CH-or-N-;
y is-CH-;
m is selected from-O-;
r is hydrogen;
ar is
Figure FDA0003794381630000012
R 1 Selected from-F, -Cl, -Br, -C 1-8 Alkyl, -C 1-8 Alkoxy, halogen substituted C 1-8 Alkyl, halogen substituted C 1-8 An alkoxy group; r 2 Selected from-H, -F, -Cl, -Br, -C 1-3 An alkyl group.
2. The compound, pharmaceutically acceptable salt or stereoisomer thereof according to claim 1, wherein R is 1 is-F, -Cl, -CH 3 、-CH 2 CH 3 、-OCH 3 、-OCH 2 CH 3 、-O-(CH 2 ) n -CH 3 、-O-(CH 2 ) n -Cl; n is an integer of 2 to 6.
3. The compound, a pharmaceutically acceptable salt or a stereoisomer thereof according to any one of claims 1-2, wherein Ar is selected from
Figure FDA0003794381630000013
Figure FDA0003794381630000014
4. The compound, a pharmaceutically acceptable salt or a stereoisomer thereof according to claim 1, wherein the compound is:
n- [2- (2-chloro-5-fluorobenzyloxy) phenyl ] nicotinic acid carboxamide;
n- [2- (2, 6-dichlorobenzyloxy) phenyl ] nicotinic acid carboxamide;
n- [2- (2-methyl-5-fluorobenzyloxy) phenyl ] nicotinic acid carboxamide;
n- [2- (2-methoxybenzyloxy) phenyl ] nicotinic acid carboxamide;
n- {2- [ (2-ethyl) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2, 6-dimethyl) benzyloxy) ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2-ethoxy) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2-methoxy-5-chloro) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2, 5-dichloro) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2-methyl-5-chloro) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [2- (4-chlorobutoxy) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [2- (5-chloropentyloxy) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [2- (6-chlorohexyloxy) benzyloxy ] phenyl } nicotinic acid carboxamide;
n- [2- (2-hexyloxybenzyloxy) phenyl ] nicotinic acid carboxamide;
n- [2- (2-heptyloxybenzyloxy) phenyl ] nicotinic acid carboxamide;
n- {2- [ (2-hexyloxy) -5-chlorobenzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [ (2-heptyloxy) -5-chlorobenzyloxy ] phenyl } nicotinic acid carboxamide;
n- {2- [2- (4-chlorobutoxy) benzyloxy ] phenyl } isonicotinic acid carboxamide;
n- {2- [2- (5-chloropentyloxy) benzyloxy ] phenyl } isonicotinic acid carboxamide;
n- {2- [ (5-chloro-2-heptyloxybenzyl) amino ] phenyl } nicotinamide;
n- {2- [ (5-chloro-2- (6-chlorohexyloxy) benzyl) amino ] phenyl } nicotinamide;
n- {2- [ (2-chloro-5-fluorobenzyl) amino ] phenyl } nicotinamide;
n- {2- [ (2, 6-dichlorobenzyl) amino ] phenyl } nicotinamide;
n- {2- [ (5-fluoro-2-methylbenzyl) amino ] phenyl } nicotinamide;
n- {2- [ (2- (4-chlorobutoxy) benzyl) amino ] phenyl } isonicotinamide;
n- {2- [ (2- (5-chloropentyloxy) benzyl) amino ] phenyl } isonicotinamide;
n- {2- [ (2-hexyloxybenzyl) amino ] phenyl } isonicotinamide;
n- {2- [ (6-chlorohexyloxybenzyl) amino ] phenyl } nicotinamide
N- {2- [2- (6-chlorohexyloxy) benzyloxy ] phenyl } isonicotinic acid carboxamide
N- {2- [2- (heptyloxy) benzyloxy ] phenyl } isonicotinic acid carboxamide.
5. A process for the preparation of a compound of formula (I) according to any one of claims 1 to 4, comprising the following steps:
Figure FDA0003794381630000031
condensing the compound 1 and the compound 2 to obtain a compound 3;
removing benzyl protection from the compound 3 through catalytic hydrogenation to obtain a compound 4;
compound 4 with
Figure FDA0003794381630000032
The final product, a compound of formula (I), should be obtained;
wherein M is-O-; r is 3 Selected from-OH, -F, -Cl or-Br.
6. A pharmaceutical composition comprising at least one therapeutically effective amount of a compound of formula (I), a pharmaceutically acceptable salt or a stereoisomer thereof according to any one of claims 1 to 4; the pharmaceutical composition comprises one or more pharmaceutically or physiologically acceptable carriers and/or diluents.
7. Use of a compound of any one of claims 1 to 4, a pharmaceutically acceptable salt or stereoisomer thereof, or a pharmaceutical composition of claim 6, for the manufacture of a medicament for the prevention and/or treatment of a disease caused by an abnormal increase in sphingomyelin levels.
CN201811405438.2A 2017-11-25 2018-11-23 Sphingomyelin synthase inhibitor, preparation method and application thereof Active CN109836378B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201711197264 2017-11-25
CN2017111972640 2017-11-25

Publications (2)

Publication Number Publication Date
CN109836378A CN109836378A (en) 2019-06-04
CN109836378B true CN109836378B (en) 2022-09-30

Family

ID=66883134

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811405438.2A Active CN109836378B (en) 2017-11-25 2018-11-23 Sphingomyelin synthase inhibitor, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN109836378B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115184481B (en) * 2022-05-31 2024-04-05 河北圣雪大成制药有限责任公司 Method for detecting content of N-tertiary butyl glycyl chloride

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003043624A1 (en) * 2001-11-16 2003-05-30 Bristol-Myers Squibb Company Dual inhibitors of adipocyte fatty acid binding protein and keratinocyte fatty acid binding protein
WO2012014127A1 (en) * 2010-07-30 2012-02-02 Ranbaxy Laboratories Limited 5-lipoxygenase inhibitors
WO2017115914A1 (en) * 2015-12-29 2017-07-06 서울대학교산학협력단 PPARγ PHOSPHORYLATION INHIBITOR AND PHARMACEUTICAL COMPOSITION COMPRISING SAME

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003043624A1 (en) * 2001-11-16 2003-05-30 Bristol-Myers Squibb Company Dual inhibitors of adipocyte fatty acid binding protein and keratinocyte fatty acid binding protein
WO2012014127A1 (en) * 2010-07-30 2012-02-02 Ranbaxy Laboratories Limited 5-lipoxygenase inhibitors
WO2017115914A1 (en) * 2015-12-29 2017-07-06 서울대학교산학협력단 PPARγ PHOSPHORYLATION INHIBITOR AND PHARMACEUTICAL COMPOSITION COMPRISING SAME

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
2018114054382;陈翠翠;《STN检索报告》;20211210;第1-34页 *
芳酰胺及肉桂酰胺类衍生物的合成及扩血管活性;付焕建,林紫云等;《药学学报》;19990228;第34卷(第02期);第109-113页 *
降血脂药物研究进展;赵士魁等;《中国医药工业杂志》;20090710;第40卷(第07期);第536-542页 *
鞘磷脂合酶的生物学功能及与动脉粥样硬化的关系;卿艳等;《中国药理学通报》;20100520;第26卷(第05期);第573-576页 *

Also Published As

Publication number Publication date
CN109836378A (en) 2019-06-04

Similar Documents

Publication Publication Date Title
JP6861858B2 (en) SSAO inhibitor
RU2435756C2 (en) Selective androgen receptor modulators
EP3686198B1 (en) Mu-opioid receptor agonist and preparation method therefor and use thereof in field of medicine
CN108349911B (en) Nitric oxide releasing prodrug molecules
KR19990037018A (en) Amide derivatives
BG107889A (en) Tetralone derivatives as antitumor agents
EP3630759B1 (en) Compounds useful as ion channel inhibitors for the treatment of cancer
JP2015083610A (en) Compound and method for treatment of pain and other diseases
AU2008327709B2 (en) Inhibitors of 17beta-hydroxysteroid dehydrogenase
EP2300424B1 (en) Use of indole derivatives as nurr-1 activators for treating parkinson s disease
CN116262736A (en) Novel oxo-pyridine compound and preparation method and application thereof
CN109836378B (en) Sphingomyelin synthase inhibitor, preparation method and application thereof
US20210292315A1 (en) Selenium-containing isoxazolamine compound, preparation method therefor, and use thereof
US8530453B2 (en) Compounds and methods for the treatment of pain and other diseases
JP5330377B2 (en) 3,4-dihydroquinazoline derivatives
KR20230078737A (en) FXR small molecule agonists and methods for their preparation and uses
FR2641534A1 (en) NOVEL GLYCINE DERIVATIVES USEFUL AS ANTAGONISTIC THROMBOXANE A2 MEDICAMENTS AND PROCESS FOR THEIR PREPARATION
MX2011001053A (en) Phenanthrenone compounds, compositions and methods.
WO2001016091A1 (en) Biscyclopropanecarboxylic acid amide compounds and medicinal use thereof
KR100804827B1 (en) Thiazolidine derivatives, processes for the preparation thereof, and pharmaceutical compositions containing the same
EP0406502B1 (en) Process for the preparation of (-)2-(2-(2-aminoethoxy)ethoxy)methyl)-4-(2, 3-dichlorophenyl)-3-ethoxycarbonyl-5-methoxycarbonyl-6-methyl-1,4-dihydropyridine, and intermediate for its preparation
KR102514860B1 (en) Biphenyl pyrrolidine and biphenyl dihydroimidazole derivatives for inhibiting activity of 5-HT7 serotonin receptor and pharmaceutical composition comprising the same as an active ingredient
CN113651767A (en) Benzisoxazole heterocyclic compound and preparation method and application thereof
BE1009520A5 (en) ACID (-) - (3R) -3-METHYL-4- 4- [4- (4-PYRIDYL) -PIPERAZIN-1-YL] BUTYRIC PHENOXY.
KR101152630B1 (en) Novel oxazole-piperazine compounds having activity for T-type calcium channel

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