CN109438347A - A kind of cyano quinolines class IDO1 inhibitor, preparation method and application - Google Patents

A kind of cyano quinolines class IDO1 inhibitor, preparation method and application Download PDF

Info

Publication number
CN109438347A
CN109438347A CN201811557706.2A CN201811557706A CN109438347A CN 109438347 A CN109438347 A CN 109438347A CN 201811557706 A CN201811557706 A CN 201811557706A CN 109438347 A CN109438347 A CN 109438347A
Authority
CN
China
Prior art keywords
acid
compound
arh
pharmaceutically acceptable
acceptable salt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN201811557706.2A
Other languages
Chinese (zh)
Other versions
CN109438347B (en
Inventor
卞金磊
王举波
冯袭
李志裕
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.)
PHARMACEUTICAL CO Ltd
Original Assignee
PHARMACEUTICAL 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 PHARMACEUTICAL CO Ltd filed Critical PHARMACEUTICAL CO Ltd
Priority to CN201811557706.2A priority Critical patent/CN109438347B/en
Publication of CN109438347A publication Critical patent/CN109438347A/en
Application granted granted Critical
Publication of CN109438347B publication Critical patent/CN109438347B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/48Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • C07D215/54Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen attached in position 3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Psychiatry (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Hospice & Palliative Care (AREA)
  • Molecular Biology (AREA)
  • AIDS & HIV (AREA)
  • Transplantation (AREA)
  • Pain & Pain Management (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a kind of cyano quinolines class IDO1 inhibitor, preparation method and application, cyano quinolines class IDO1 inhibitor is compounds of formula I or its pharmaceutically acceptable salt.The preparation method is as follows: being obtained by mercaptan acid III and the chlorine atom in compound II through nucleophilic substitution reaction by the process of compound II prepare compound IV;By the process of compound IV prepare compound I, it is condensed to yield by substituted aniline V with carboxyl.The inhibitor is used to treat the immunosuppressive related disease of the mediation of indole amine 2,3-dioxygenase 1.

Description

A kind of cyano quinolines class IDO1 inhibitor, preparation method and application
Technical field
The present invention relates to field of medicaments, and in particular to a kind of derivative containing cyano quinolines, its is pharmaceutically acceptable Salt, its synthetic method and its application in medicine.
Background technique
Immunologic test point inhibitor (Immune checkpoint inhibitors) and Chimeric antigen receptor T cell The success of (CAR-T cell) therapy clinically shows that tumour immunotherapy has advantage efficiently, safe (Schadendorf D.et al.J.Clin.Oncol.,2015,33:1889-1894;Yang Y.J.Clin.Invest., 2015,125:3335-3337).However for such bio-pharmaceutical, clinically applies there are still certain defect, wrap Include response rate it is low, cannot using oral therapies, it is expensive (Mullard A.Nat.Rev.Drug Discov., 2015,14: 739;Kreamer KM, et al.J Adv Pract Oncol, 2014,5:418-431) etc..Therefore, research and development are for immune inspection The small-molecule drug for making an inventory for the treatment of can play supplement and potential synergistic effect to the clinical application of macromolecular bio-pharmaceutical.
Tryptophan (Trp) is a kind of essential amino acid of human body internal protein synthesis, and 95% or more Trp passes through dog urinary ammonia Sour approach (kynurenine pathway, KP) metabolism, is finally metabolized as kynurenic acid (kynurenic acid), nicotinoyl Amine adenine-dinucleotide (NAD) and xanthurenic acid (4,8-Er Qiangjikuilinjiasuan) (xanthurenic acid) isoreactivity metabolite, to be grown to cell Generate a series of important physiological actions (Routy JP, Routy B, Graziani GM, et al.Int.J.Tryptophan Res.,2016,9:67-77).Wherein, indoleamine 2,3-dioxygenase 1 (Indoleamine 2,3-dioxygenase 1, It IDO1) is the first rate-limiting enzyme (Mellor AL, the et al.Nat Rev for regulating and controlling tryptophan catabolism KP access outside liver Immunol,2004,4:762-774).IDO1 is a kind of monomeric protein comprising heme, by 403 amino acid residues Composition, including two fold α-helixstructure domain, big structure domain include catalytic pocket, substrate can in catalytic pocket with IDO1 The effects of hydrophobic (Rohrig UF, et al.J.Med.Chem., 2015,58:9421) occurs.IDO1 is in vivo under normal circumstances In low expression level, when interferon (IFN-α, IFN-β and IFN-γ), interleukin (IL-1 and IL-2), tumor necrosis factor (TNF) etc. when cytokine profiles induction IDO1 level increases, tryptophan will be extensively metabolized, to inhibit human body to parasitism Property, the pathogen such as viral, bacillary, fungoid immune response, human body will be in the immunosuppressive condition of morbid state (Banzola,I.et al.Front.Immunol.,2018,9:1051).IDO1 mainly passes through two aspect mediated immunity suppressions System: on the one hand, IDO1 exhausts local T rp, causes T cell Cycle Arrest;On the other hand, the resolution of Trp causes metabolism to produce Object (such as kynurenin, Kyn's) builds up, and activation aryl hydrocarbon receptor (AhR) etc. further activates regulatory T-cell (Treg ) and then depression effect T cell (Munn DH, et al.J Exp Med, 1999,189:1363-1372 cell;Mezrich JD, et al.J Immunol,2010,185:3190-3198)。
Studies have shown that IDO1 high expression in many tumours, in addition, IDO1 is again often associated with prognosis mala (Okamoto A,et al.Clin Cancer Res,2005,11:6030-6039).In fact, inhibit IDO1 activity for Oncotherapy has significant facilitation, and IDO1 inhibitor is clinically combined with PD-1, PD-L1 inhibitor to improve at present Curative effect (Marin-Acevedo JA, et al.J.Hematol.Oncol., 2018,11:39).
Disclosed selectivity IDO1 inhibitor patent application includes WO2010005958, WO2015173764, WO2016155545, WO2016073770 and WO2018184392 etc..
IDO1 inhibitor has a good application prospect as drug in pharmaceuticals industry, but not yet finds at present well IDO1 inhibitor is as marketed drug.In order to reach better oncotherapy effect, it is intended that develop high-efficiency low-toxicity of new generation Selective IDO1 inhibitor, show excellent effect and effect, excellent medicine generation absorbs activity.
Summary of the invention
The present invention is directed to find high structure novel, activity, Small side effects and resisting with good pharmacokinetic properties to swell Tumor candidate compound.These compounds are by being applied alone or being combined with other anti-tumor drugs, so that it is existing antitumor to reach raising Curative effect of medication and the effect for reducing dosage and toxicity.
The invention discloses compounds of formula I, its stereoisomer or its pharmaceutically acceptable salts.
Wherein:
R1Selected from hydrogen atom, cyano, amino, nitro, halogen, C1~C4Alkoxy, substituted or unsubstituted C1~C5Alkyl; The C wherein replaced1~C5In alkyl, substituent group is selected from one or more of halogen, amino, nitro, hydroxyl or cyano;
R2Selected from hydrogen atom, cyano, amino, nitro, halogen, C1~C4Alkoxy, substituted or unsubstituted C1~C5Alkyl, Substituted or unsubstituted aryl;The C wherein replaced1~C5In alkyl, substituent group is selected from halogen, amino, nitro, hydroxyl or cyano One or more of;
N is selected from 1,2,3,4.
In a preferred embodiment of the invention, in which:
R1Selected from hydrogen atom, fluorine atom, chlorine atom, bromine atom, methoxyl group, methyl;
R2Selected from hydrogen atom, fluorine atom, chlorine atom, bromine atom, phenyl;
N represents 1,2.
The pharmaceutically acceptable salt of the compound of logical formula (I) of the present invention refers to the compound and medicine of logical formula (I) On it is acceptable acid formed acid-addition salts, the acid include: hydrogen chloride, hydrogen bromide, sulfuric acid, carbonic acid, oxalic acid, citric acid, Succinic acid, tartaric acid, phosphoric acid, lactic acid, pyruvic acid, acetic acid, maleic acid, methanesulfonic acid, benzene sulfonic acid, p-methyl benzenesulfonic acid or ferulic acid.
Currently preferred part of compounds such as the following table 1:
1 preferred compound of table
Another object of the present invention is to provide the preparation methods of compound shown in logical formula (I):
Wherein: R1, R2, n is defined as above described.
By the process of compound II prepare compound IV, by the chlorine atom in mercaptan acid III and compound II through nucleophilic Substitution reaction obtains;Acid binding agent used in nucleophilic substitution is sodium carbonate, potassium carbonate, sodium hydroxide or triethylamine, preferably carbonic acid Potassium;Solvent for use is tetrahydrofuran, methylene chloride, n,N-Dimethylformamide, dimethyl sulfoxide, preferably dimethyl sulfoxide.
By the process of compound IV prepare compound I, dehydrating condensation is carried out with carboxyl by substituted aniline V and is obtained;It is used Condensing agent be dicyclohexylcarbodiimide (DCC), 1- (3- dimethylamino-propyl) -3- ethyl-carbodiimide hydrochloride (EDCI), N, N'- diisopropylcarbodiimide (DIC), preferably EDCI;Catalyst or activator are 4-dimethylaminopyridine (DMAP), 1- hydroxyl Base benzotriazole (HOBT), preferably HOBT;Solvent is methylene chloride, n,N-Dimethylformamide, dimethyl sulfoxide, preferably N, N- Dimethylformamide.
More specific method includes:
Wherein: R1, R2, n is defined as above described.
By the process of compound VI prepare compound VII, reaction reagent is phosphorus oxychloride, n,N-Dimethylformamide.
By the process of compound VII prepare compound II, reaction reagent is ammonium hydroxide, and oxidant is iodine or lead tetra-acetate or oxygen Gas, solvent are tetrahydrofuran.
The process of compound II prepare compound IV, reactant are mercaptan acid III, and acid binding agent is sodium carbonate, potassium carbonate, hydrogen Sodium oxide molybdena or triethylamine, solvent are tetrahydrofuran, methylene chloride, n,N-Dimethylformamide, dimethyl sulfoxide.
By the process of compound IV prepare compound I, reactant is substituted aniline V, and condensing agent DCC, EDCI, DIC are urged Agent or activator are DMAP, HOBT, and solvent is methylene chloride, n,N-Dimethylformamide, dimethyl sulfoxide.
The pharmaceutically acceptable salt of the compound of Formula I can by with equal chemical equivalents or excess acid (inorganic acid or Organic acid) in suitable solvent or solvent compositions react be made.The acid includes but is not limited to hydrogen chloride, hydrogen bromide, sulphur Acid, carbonic acid, oxalic acid, citric acid, succinic acid, tartaric acid, phosphoric acid, lactic acid, pyruvic acid, acetic acid, maleic acid, methanesulfonic acid, benzene sulfonic acid, P-methyl benzenesulfonic acid or ferulic acid.The solvent includes but is not limited to methanol, ethyl alcohol, methylene chloride, acetone, ethyl acetate, toluene Or tetrahydrofuran, or any several mixed solvents.
The present invention provides a kind of pharmaceutical compositions comprising the active component of medicine effective quantity and pharmaceutically acceptable Auxiliary material;The active component includes one of compound of Formula I and pharmaceutically acceptable salt or a variety of.The pharmaceutical composition In object, the auxiliary material includes pharmaceutically acceptable carrier, diluent and/or excipient.
Pharmaceutical composition can be made to various types of administration unit dosage forms according to therapeutic purposes, such as tablet, pill, powder Agent, liquid, suspension, lotion, granule, granule, capsule and injection (solution or suspension) etc., preferred tablet, capsule, liquid Body, suspension and injection (solution or suspension).
The modes such as oral, injection can be used in the administration mode of compound of the present invention clinically.
Generally, the compound of the present invention is for when treating, people to be 1-1000mg/ days with dosage range.It can also be according to agent The difference and disease severity of type, dosage exceed the range.
The present invention also provides compounds shown in general formula I to prepare the application in 1 inhibitor of indole amine 2,3-dioxygenase.
The present invention also provides compounds shown in general formula I in the immune suppression for treating the mediation of indole amine 2,3-dioxygenase 1 Application in the related disease of system.
The immunosuppressive related disease that indole amine 2,3-dioxygenase 1 of the present invention mediates includes cancer, virus Infection, neurodegenerative disease, cataract, organ-graft refection, depression or autoimmune disease.The wherein preferred lung of cancer Cancer, melanoma, head and neck cancer, clear-cell carcinoma, bladder transitional cell carcinoma.The preferred HIV infection of virus infection.Neurodegenerative disease preferred Ah The silent disease in Wurz sea.
Unless otherwise stated, the following term used in the specification and in the claims has meaning discussed below:
Term " alkyl " indicates the aliphatic group of the saturation of 1-20 carbon atom, including straight chain and branched group (this specification In the digital scope mentioned, such as " 1-5 " refers to the group, is at this time alkyl, can contain 1 carbon atom, 2 carbon atoms, 3 A carbon atom etc., until including 5 carbon atoms).Alkyl can be substituted or unsubstituted.It, should when being the alkyl replaced Substituent group is preferably one or more, and more preferable 1-3, most preferably 1 or 2 substituent group.
Term " alkoxy " expression-O-(unsubstituted alkyl) and-O-(unsubstituted naphthenic base).Representative example includes But be not limited to methoxyl group, ethyoxyl, propoxyl group, butoxy, cyclopropyl oxygroup, cyclobutoxy group, cyclopentyloxy, cyclohexyloxy etc..
Term " halogen " indicates fluorine, chlorine, bromine or iodine, preferably fluorine, chlorine, bromine.
Term " amino " expression-NH2Group.
Term " nitro " expression-NO2Group.
Term " hydroxyl " expression-OH group.
Term " cyano " expression-CN group.
Term " aryl " indicates the full carbon monocycle or fused polycycle group of 1 to 12 carbon atom, the π electricity with total conjugated Subsystem.The non-limiting example of aryl has phenyl, naphthalene and anthryl.Aryl can be substituted or unsubstituted.
Specific embodiment
The present invention is further illustrated below by the mode of embodiment, but does not therefore limit the present invention to the reality It applies among a range.
Embodiment 1
It is prepared by (VII-1) of the chloro- 3- quinoline aldehyde of step 1:2-
DMF (9.1g, 125mmol, 2.5equiv) is added in reaction flask, 10min is stirred under ice bath, is added dropwise POCl3(30.4g, 200mmol, 4equiv) and reacting liquid temperature is controlled no more than 5 DEG C, be added dropwise under rear ice bath and continue to stir Until reaction solution solidifies completely, antifebrin (6.8g, 50mmol, 1equiv) is added and is slowly risen after stirring 10min at room temperature Temperature reacts 3h to 75 DEG C.Excessive POCl is removed under reduced pressure after TLC detection fully reacting3, viscous fluid, which pours into, uses NaHCO in ice water3It adjusts PH value is saved to 5-6, filters to obtain VI-1 (6.8g, yield 71%).HRMS m/z[M+H]+calculated for C10H6ClNO: 192.0211,found:192.0213.1H NMR(300MHz,DMSO-d6): δ=10.39 (s, 1H ,-CHO), 9.01 (s, 1H ,- ), ArH 8.30 (d, J=8.2Hz, 1H ,-ArH), 8.07-7.97 (m, 2H ,-ArH), 7.77 (t, J=7.5Hz, 1H ,-ArH) ppm。
The preparation of step 2:2- chloroquinoline -3- formonitrile HCN (II-1)
VII-1 (3.8g, 20mmol, 1equiv) is dissolved in THF, be added 25% ammonium hydroxide (6.8mL, 100mmol, 5equiv) and I2(6.0g, 24mmol, 1.2equiv) reacts 3h at 25 DEG C.Reaction solution is poured into after TLC detection fully reacting It is saturated Na2S2O3In aqueous solution, there are a large amount of solids to be precipitated, filter to obtain V-2 (2.4g, yield 63%).HRMS m/z[M+H]+ calculated for C10H5ClN2:189.0214,found:189.0222.1H NMR(300MHz,DMSO-d6): δ=9.28 (s, 1H ,-ArH), 8.16 (d, J=8.3Hz, 1H ,-ArH), 8.10-8.02 (m, 2H ,-ArH), 7.83 (t, J=7.3Hz, 1H,–ArH)ppm。
Embodiment 2
The preparation of step 1:3- ((3- cyano quinolines -2- base) is thio) propionic acid (II-1)
Thioglycolic acid (1.1g, 12mmol, 1.2equiv) is dissolved in DMSO, stirs 10min at room temperature, is slowly divided It criticizes and K is added2CO3(6.9g, 50mmol, 5equiv) continuation stir 30min at room temperature, dropwise instill II-1 (1.9g, 10mmol, It is to slowly warm up to 60 DEG C after DMSO dilution 1equiv), reacts 3h.By bodies such as reaction solution additions after TLC detection fully reacting In ponding, with salt acid for adjusting pH value to 1-2, there are a large amount of grease to be precipitated, be extracted with ethyl acetate water phase 3 times, wash organic phase 2 It is secondary, it removes ethyl acetate under reduced pressure and obtains crude product, recrystallize to obtain IV-1 (2.1g, yield 86%).HRMS m/z[M+H]+ calculated for C12H8N2O2S:245.0379,found:245.0391.1H NMR(300MHz,DMSO-d6): δ= 12.88 (s, 1H ,-COOH), 9.01 (s, 1H ,-ArH), 8.02 (d, J=8.1Hz, 1H ,-ArH), 7.95-7.87 (m, 2H ,- ), ArH 7.66 (t, J=7.1Hz, 1H ,-ArH), 4.20 (s, 2H ,-CH2 -) ppm.
The preparation of step 2:2- ((3- cyano quinolines -2- base) is thio)-phenyl acetanilide,Phenacetylaniline (CPU-Q1)
By IV-1 (0.24g, 1mmol, 1equiv), EDCI (0.25g, 1.3mmol, 1.3equiv) and HOBT (0.04g, 0.3mmol, 0.3equiv) be dissolved in DMF, no more than 10 DEG C at react 30min.Addition aniline (0.10g, 1.1mmol, 1.1equiv) reaction solution color is deepened afterwards, and reaction is stayed overnight at room temperature.Reaction solution is poured into water after TLC detection fully reacting, is taken out Filter, filter cake column chromatograph to obtain CPU-Q1 (180mg, yield 56%).m.p.226–228℃.HRMS m/z[M+H]+calculated for C18H13N3OS:320.0852,found:320.0850.1H NMR(300MHz,DMSO-d6): δ=10.45 (s, 1H ,- NH -), 8.99 (s, 1H ,-ArH), 8.00 (d, J=8.2Hz, 1H ,-ArH), 7.88 (s, 2H ,-ArH), 7.62 (d, J= 8.0Hz, 3H ,-ArH), 7.31 (t, J=7.8Hz, 2H ,-ArH), 7.05 (t, J=7.3Hz, 1H ,-ArH), 4.33 (s, 2H ,- CH2–)ppm.13C NMR(75MHz,DMSO-d6): δ=165.83,157.22,147.64,144.14,138.99,133.64, 128.88,128.76,127.35,127.05,123.75,123.38,119.13,115.65,104.41,35.08ppm。
Embodiment 3
2- (3- cyano quinolines -2- base) sulfenyl)-N- (4- fluorophenyl) acetamide (CPU-Q2) preparation
Outside with 4- fluoroaniline (0.11g, 1.1mmol, 1.1equiv) substitution aniline, method identical with compound CPU-Q1 (with embodiment 2, step 2) synthesizes compound CPU-Q2 (120mg, yield 36%).m.p.250–252℃.HRMS m/z[M+H]+ calculated for C18H12FN3OS:338.0758,found:338.1H NMR(300MHz,DMSO-d6): δ=10.51 (s, 1H ,-NH -), 9.01 (s, 1H ,-ArH), 8.01 (d, J=9.0Hz, 1H ,-ArH), 7.93-7.85 (m, 2H ,-ArH), 7.66-7.61 (m, 3H ,-ArH), 7.16 (t, J=9.0Hz, 2H ,-ArH), 4.32 (s, 2H ,-CH2–)ppm。
Embodiment 4
The preparation of 2- ((3- cyano -6- methylquinoline -2- base) sulfenyl)-N- (4- fluorophenyl) acetamide (CPU-Q3)
Antifebrin, 4- fluoroaniline is replaced to replace outside aniline with 4- exalgine, using the identical preparation of CPU-Q1 Method synthesizes compound CPU-Q3 (130mg, yield 38%).m.p.247–250℃.HRMS m/z[M+H]+calculated for C19H14FN3OS:352.0914,found:352.0910.1H NMR(300MHz,DMSO-d6): δ=10.46 (s, 1H ,- NH -), 8.89 (s, 1H ,-ArH), 7.77 (d, J=6.0Hz, 3H ,-ArH), 7.63-7.59 (m, 2H ,-ArH), 7.14 (t, J= 9.0Hz,2H,–ArH),4.29(s,2H,–CH2–),2.51(s,3H,–CH3)ppm。
Embodiment 5
The preparation of 2- ((3- cyano -8- methylquinoline -2- base) sulfenyl)-N- (4- fluorophenyl) acetamide (CPU-Q4)
Antifebrin, 4- fluoroaniline is replaced to replace outside aniline with 2- exalgine, using the identical preparation of CPU-Q1 Method synthesizes compound CPU-Q4 (121mg, yield 35%).m.p.243–246℃.HRMS m/z[M+H]+calculated for C19H14FN3OS:352.4069,found:352.4064.1H NMR(300MHz,DMSO-d6): δ=10.43 (s, 1H ,- NH -), 8.95 (s, 1H ,-ArH), 7.82 (d, J=6.0Hz, 1H ,-ArH), 7.71 (d, J=6.0Hz, 1H ,-ArH), 7.62- 7.48 (m, 3H ,-ArH), 7.14 (t, J=9.0Hz, 2H ,-ArH), 4.31 (s, 2H ,-CH2–),2.56(s,3H,–CH3)ppm.
Embodiment 6
The preparation of 2- ((3- cyano -6- fluorine quinoline -2- base) sulfenyl)-N- (4- fluorophenyl) acetamide (CPU-Q5)
Antifebrin, 4- fluoroaniline is replaced to replace outside aniline with 4- fluoroacetanilide, using the identical preparation side CPU-Q1 Method synthesizes compound CPU-Q5 (142mg, yield 40%).m.p.218–220℃.HRMS m/z[M+H]+calculated for C18H11F2N3OS:356.0664,found:356.0662.1H NMR(300MHz,DMSO-d6): δ=10.57 (s, 1H ,-NH-), 9.09 (s, 1H ,-ArH), 8.11 (d, J=6.0Hz, 1H ,-ArH), 7.69-7.54 (m, 2H ,-ArH), 7.38-7.26 (m, 2H ,-ArH), 7.15 (t, J=9.0Hz, 2H ,-ArH), 4.35 (s, 2H ,-CH2–)ppm。
Embodiment 7
The preparation of 2- ((3- cyano -6- methoxy quinoline -2- base) sulfenyl)-N- (4- fluorophenyl) acetamide (CPU-Q6)
Antifebrin, 4- fluoroaniline is replaced to replace outside aniline with 4- p-methoxyacetanilide, using the identical system of CPU-Q1 Preparation Method synthesizes compound CPU-Q6 (126mg, yield 33%).m.p.232–235℃.HRMS m/z[M+H]+calculated for C19H14FN3O2S:368.0864,found:368.0863.1H NMR(300MHz,DMSO-d6): δ=10.44 (s, 1H ,- NH -), 8.85 (s, 1H ,-ArH), 7.79 (d, J=9.0Hz, 1H ,-ArH), 7.63-7.53 (m, 3H ,-ArH), 7.40 (d, J= 3.0Hz, 1H ,-ArH), 7.14 (t, J=9.0Hz, 2H ,-ArH), 4.27 (s, 2H ,-CH2–),3.88(s,3H,–CH3)ppm。
Embodiment 8
The preparation of N- (3- chlorphenyl) -2- ((3- cyano quinolines -2- base) is thio) acetamide (CPU-Q7)
Outside with 3- chloroaniline (0.14g, 1.1mmol, 1.1equiv) substitution aniline, method identical with compound CPU-Q1 (with embodiment 2, step 2) synthesizes compound CPU-Q7 (170mg, yield 48%).m.p.>250℃.HRMS m/z[M+H]+ calculated for C18H12ClN3OS:354.0462,found:354.0463.1H NMR(300MHz,DMSO-d6): δ= 9.67 (s, 1H ,-NH -), 9.20 (s, 1H ,-ArH), 8.11 (d, J=9.0Hz, 2H ,-ArH), 7.97 (t, J=2.1Hz, 1H ,- ), ArH 7.88 (t, J=7.7Hz, 1H ,-ArH), 7.72 (s, 2H ,-CH2–),7.69–7.64(m,2H,–ArH),7.38(t,J =8.1Hz, 1H ,-ArH), 7.15 (d, J=7.9Hz, 1H ,-ArH) ppm.13C NMR(75MHz,DMSO-d6): δ=163.94, 159.26,148.27,147.08,140.54,132.71,131.29,130.94,130.03,129.19,127.71,125.85, 125.69,124.59,122.98,120.30,119.20,95.28ppm。
Embodiment 9
The preparation of N- (3- bromophenyl) -2- ((3- cyano quinolines -2- base) is thio) acetamide (CPU-Q8)
Outside with 3- bromaniline (0.19g, 1.1mmol, 1.1equiv) substitution aniline, method identical with compound CPU-Q1 (with embodiment 2, step 2) synthesizes compound CPU-Q8 (160mg, yield 40%).m.p.190–192℃.HRMS m/z[M+H]+ calculated for C18H12BrN3OS:397.9957,found:397.9954.1H NMR(300MHz,DMSO-d6): δ= 10.63 (s, 1H ,-NH -), 9.01 (s, 1H ,-ArH), 8.03-7.83 (m, 4H ,-ArH), 7.64 (t, J=7.4Hz, 1H ,- ), ArH 7.53 (d, J=7.7Hz, 1H ,-ArH), 7.32-7.24 (m, 2H ,-ArH), 4.33 (s, 2H ,-CH2–)ppm.13C NMR (75MHz,DMSO-d6): δ=166.30,157.10,147.62,144.20,140.56,133.69,130.82,128.91, 127.31,127.10,126.00,123.77,121.56,121.42,117.86,115.63,104.37,35.06ppm。
Embodiment 10
The preparation of N- ([1,1 '-xenyl] -4- base) -2- ((3- cyano quinolines -2- base) is thio) acetamide (CPU-Q9)
Outside with 4- aminobphenyl (0.19g, 1.1mmol, 1.1equiv) substitution aniline, side identical with compound CPU-Q1 (with embodiment 2, step 2) synthesizes compound CPU-Q9 (180mg, yield 46%) to method.m.p.>250℃.HRMS m/z[M+H]+ calculated for C24H17N3OS:396.1165,found:396.1163.1H NMR(300MHz,DMSO-d6): δ= 10.60 (s, 1H ,-NH -), 9.03 (s, 1H ,-ArH), 8.02 (d, J=8.1Hz, 1H ,-ArH), 7.90 (s, 2H ,-ArH), 7.74-7.63 (m, 7H ,-ArH), 7.44 (t, J=7.6Hz, 2H ,-ArH), 7.33 (t, J=7.3Hz, 1H ,-ArH), 4.36 (s,2H,–CH2–)ppm.13C NMR(75MHz,DMSO-d6): δ=165.91,157.23,147.65,144.19,139.59, 138.47,135.03,133.69,128.91,128.86,127.36,127.08,126.98,126.21,123.76,119.46, 115.67,104.39,35.14ppm。
Embodiment 11
The preparation of 3- ((3- cyano quinolines -2- base) is thio)-N- Phenylpropionamide (CPU-Q10)
It is replaced outside thioglycolic acid with 3- mercaptopropionic acid, compound CPU- is synthesized using the identical preparation method of CPU-Q1 Q10 (180mg, yield 54%).m.p.226–228℃.HRMS m/z[M+H]+calculated for C19H15N3OS: 334.1009,found:334.1004.1H NMR(300MHz,DMSO-d6): δ=10.01 (s, 1H ,-NH -), 9.00 (s, 1H ,- ), ArH 8.03-7.90 (m, 3H ,-ArH), 7.69-7.58 (m, 3H ,-ArH), 7.30 (t, J=7.7Hz, 2H ,-ArH), 7.04 (t, J=7.6Hz, 1H ,-ArH), 3.65 (t, J=6.9Hz, 2H ,-S-CH2), 2.90 (t, J=6.9Hz, 2H ,-CH2- C= O–)ppm.13C NMR(75MHz,DMSO-d6): δ=169.24,157.56,147.82,144.15,139.01,133.56, 128.85,128.68,127.64,126.98,123.71,123.12,118.97,115.74,104.93,35.44, 25.30ppm。
Embodiment 12
The preparation of 3- ((3- cyano quinolines -2- base) is thio)-N- (4- fluorophenyl) propionamide (CPU-Q11)
Thioglycolic acid, 4- fluoroaniline is replaced to replace outside aniline with 3- mercaptopropionic acid, using the identical preparation side CPU-Q1 Method synthesizes compound CPU-Q11 (210mg, yield 60%).m.p.235–237℃.HRMS m/z[M+H]+calculated for C19H14FN3OS:352.0914,found:352.0912.1H NMR(300MHz,DMSO-d6): δ=10.06 (s, 1H ,- NH -), 8.97 (s, 1H ,-ArH), 8.03-7.90 (m, 3H ,-ArH), 7.68-7.58 (m, 3H ,-ArH), 7.13 (t, J= 8.7Hz, 2H ,-ArH), 3.65 (t, J=6.8Hz, 2H ,-S-CH2), 2.89 (t, J=6.8Hz, 2H ,-CH2- C=O -) ppm.13C NMR(75MHz,DMSO-d6): δ=169.14,157.88 (d, Ar-F, J=238.3Hz), 157.53,147.82, (144.10,135.39 d, J=2.3Hz), 133.54,128.83,127.63,126.97,123.71,120.74 (d, J= 7.8Hz), 115.72,115.54 (d, J=22.0Hz), 104.93,35.39,25.32ppm.
Embodiment 13
The preparation of N- (3- chlorphenyl) -3- ((3- cyano quinolines -2- base) is thio) propionamide (CPU-Q12)
Thioglycolic acid, 3- chloroaniline is replaced to replace outside aniline with 3- mercaptopropionic acid, using the identical preparation side CPU-Q1 Method synthesizes compound CPU-Q12 (200mg, yield 54%).m.p.200–202℃.HRMS m/z[M+H]+calculated for C19H14ClN3OS:368.0619,found:368.0615.1H NMR(300MHz,DMSO-d6): δ=10.22 (s, 1H ,- NH–),8.98(s,1H,–ArH),8.02–7.89(m,3H,–ArH),7.83(s,1H,–ArH),7.68–7.63(m,1H,– ), ArH 7.43 (d, J=8.3Hz, 1H ,-ArH), 7.33 (t, J=8.1Hz, 1H ,-ArH), 7.10 (d, J=8.0Hz, 1H ,- ), ArH 3.66 (t, J=6.7Hz, 2H ,-S-CH2), 2.92 (t, J=6.7Hz, 2H ,-CH2- C=O -) ppm.13C NMR (75MHz,DMSO-d6): δ=169.69,157.47,147.78,144.11,140.40,133.52,133.02,130.38, 128.83,127.62,126.96,123.69,122.83,118.44,117.32,115.72,104.91,35.55, 25.17ppm。
Embodiment 14
The preparation of N- (3- bromophenyl) -3- ((3- cyano quinolines -2- base) is thio) propionamide (CPU-Q13)
Thioglycolic acid, 3- bromaniline is replaced to replace outside aniline with 3- mercaptopropionic acid, using the identical preparation side CPU-Q1 Method synthesizes compound CPU-Q13 (200mg, yield 49%).m.p.216–218℃.HRMS m/z[M+H]+calculated for C19H14BrN3OS:412.0114,found:412.0111.1H NMR(300MHz,DMSO-d6): δ=10.20 (s, 1H ,- NH -), 8.99 (s, 1H ,-ArH), 8.03-7.90 (m, 4H ,-ArH), 7.66 (t, J=7.4Hz, 1H ,-ArH), 7.47 (d, J= 7.4Hz, 1H ,-ArH), 7.29-7.24 (m, 2H ,-ArH), 3.65 (t, J=6.6Hz, 2H ,-S-CH2), 2.91 (t, J= 6.7Hz,2H,–CH2- C=O -) ppm.13C NMR(75MHz,DMSO-d6): δ=169.68,157.47,147.79,144.14, 140.54,133.54,130.70,128.84,127.62,126.98,125.74,123.71,121.51,121.30,117.70, 115.72,104.92,35.54,25.17ppm。
Embodiment 15
The preparation of N- ([1,1 '-xenyl] -4- base) -3- ((3- cyano quinolines -2- base) is thio) propionamide (CPU-Q14)
Thioglycolic acid, 4- aminobphenyl is replaced to replace outside aniline with 3- mercaptopropionic acid, using the identical preparation of CPU-Q1 Method synthesizes compound CPU-Q14 (200mg, yield 49%).m.p.>250℃.HRMS m/z[M+H]+calculated for C25H19N3OS:410.1322,found:410.1324.1H NMR(300MHz,DMSO-d6): δ=10.11 (s, 1H ,-NH -), 8.98 (s, 1H ,-ArH), 8.03-7.90 (m, 3H ,-ArH), 7.71-7.63 (m, 7H ,-ArH), 7.44 (t, J=7.4Hz, 2H ,-ArH), 7.32 (t, J=7.4Hz, 1H ,-ArH), 3.67 (t, J=6.8Hz, 2H ,-S-CH2), 2.93 (t, J= 6.7Hz,2H,–CH2- C=O -) ppm.13C NMR(75MHz,DMSO-d6): δ=169.32,157.56,147.82,144.15, 139.64,138.49,134.77,133.56,128.85,127.65,126.97,126.88,126.18,123.71,119.40, 119.34,115.75,104.93,35.51,25.31ppm。
It is the pharmacology test and result of part of compounds of the present invention below:
One, inhibitory activity of the part of compounds of the present invention to hIDO1
Experimental method:
The 50mmol kaliumphosphate buffer that secure ph is 6.5, is added and reaches sodium ascorbate 25mmol, methylene blue 10 μM, 100 μ g/mL of catalase, 100 μM of L-Trp.The diluted compound of series of concentrations is added in 96 orifice plates, every hole adds The IDO1 enzyme for entering extraction guarantees that every hole final volume is 200 μ L, 37 DEG C of incubation 1h.Take 140 μ L supernatants that 96 new orifice plates are added In, 10 μ L, 30% trichloroacetic acid solution or 1N NaOH aqueous solution is added in 60 DEG C of hydrolysis 30min in every hole.After hydrolysis in 10000rpm is centrifuged 10min at 0 DEG C.When being hydrolyzed with trichloroacetic acid, takes in 100 μ L supernatants and new 96 hole transparent panel, add 100 μ L The paradime thylaminobenzaldehyde acetum of 2% (w/v), mixes 2min at room temperature, and the absorbance in every hole is surveyed at 480nm.With NaOH takes 100 μ L supernatants in white 96 orifice plates when hydrolyzing, the emitted luminescence intensity of 460nm is surveyed under 360nm exciting light.Use L- Kynurenin is that reference substance makes two standard curves.Prepare series of concentrations reference substance (200,100,50,25,12.5, 6.25,3.12,1.56 μm of ol), the paradime thylaminobenzaldehyde of 100 μ L and 2% isometric (w/v) are taken under (1) each concentration Acetum mixes, and surveys absorbance at 480nm.(2) with 100 μ L are taken in 96 hole blanks under each concentration, in 360nm excitation The emitted luminescence intensity of 460nm is surveyed under light.Nonlinear regression (Graphpad prism) generates IC to analyze data50Value.Using upper Method is stated, the IDO1 inhibitory activity of compound is measured, wherein IC50It is as follows, and with being currently in II phase clinical trial IDO1 inhibitor NLG0919 is control.
Experimental result is as shown in table 2.
Inhibitory activity IC of the part of compounds of the present invention of table 2 to hIDO150
As can be seen from Table 2, part of compounds of the present invention has good inhibitory activity to hIDO1.
Two, the measurement of part of compounds of the present invention IDO1 protease inhibiting activity intracellular to Hela
Experimental method:
With 1640 cell culture fluid of RPMI containing 10%FBS by Hela cell culture to logarithmic phase, simultaneously with pancreatin digestion It is 5 × 10 that concentration, which is made,4The cell suspension of a/mL, in the case where guaranteeing every 5000 cells in hole by cell inoculation transparent It is put in 37 DEG C of cell incubators and is cultivated for 24 hours in 96 orifice plates.It is adherent and grow form in 96 orifice plates to cell, discard former training Base is supported, IFN-γ human interferon (hole 20ng/) and the diluted compound of series of concentrations are added in 96 orifice plates and (guarantee every hole end body Product is 200 μ L) it is put in cell incubator and cultivates 48h.140 μ L culture solution supernatants in former 96 orifice plates are moved into 96 new orifice plates In, the trichloroacetic acid aqueous solution that 10 μ L 6.1N are added in every hole hydrolyzes 30min at 60 DEG C.96 orifice plates are put in after hydrolysis In centrifuge, 20min is centrifuged with 4000rpm at 0 DEG C.100mL supernatant is moved to 96 new hole transparent panels after centrifugation In, the paradime thylaminobenzaldehyde acetum of 100 μ L 2% (w/v) is added in every hole, 2min is mixed at room temperature, at 480nm Survey the absorbance in every hole.It is reference substance production standard curve with L- kynurenin.Prepare series of concentrations reference substance (200,100, 50,25,12.5,6.25,3.12,1.56 μM), take under each concentration 100 μ L and 2% isometric (w/v) to dimethylamino Benzaldehyde acetum mixes, and surveys absorbance at 480nm.It is raw that data are analyzed with nonlinear regression (Graphpad prism) At IC50Value.3 multiple holes of experimental setup.Using the above method, the IDO1 inhibitory activity of compound is measured, wherein IC50 It is as follows.It uses and is currently in the IDO1 inhibitor NLG0919 of II phase clinical trial as control.
Experimental result is as shown in table 3:
IDO1 inhibitory activity IC of the part of compounds of the present invention of table 3 based on Hela cell50
As shown in Table 3, IDO1 inhibitory activity IC of the part of compounds of the present invention based on Hela cell50It is substantially better than current place In the IDO1 inhibitor NLG0919 of II phase clinical trial.
The above is only a preferred embodiment of the present invention, it should be pointed out that: for the ordinary skill people of the art For member, various improvements and modifications may be made without departing from the principle of the present invention, these improvements and modifications are also answered It is considered as protection scope of the present invention.

Claims (10)

1. compounds of formula I or its pharmaceutically acceptable salt:
Wherein:
R1Selected from hydrogen atom, cyano, amino, nitro, halogen, alkoxy, substituted or unsubstituted C1~C5Alkyl;Wherein replace Base is selected from one or more of halogen, amino, nitro, hydroxyl or cyano;
R2Selected from hydrogen atom, cyano, amino, nitro, halogen, alkoxy, substituted or unsubstituted C1~C5Alkyl, substitution or not Substituted aryl;Wherein substituent group is selected from one or more of halogen, amino, nitro, hydroxyl or cyano;
N is selected from 1,2,3,4.
2. compounds of formula I or its pharmaceutically acceptable salt according to claim 1, which is characterized in that R1Selected from hydrogen original Son, fluorine atom, chlorine atom, bromine atom, methoxyl group, methyl;R2Selected from hydrogen atom, fluorine atom, chlorine atom, bromine atom, phenyl.
3. compounds of formula I or its pharmaceutically acceptable salt according to claim 1, which is characterized in that wherein n is represented 1、2。
4. compounds of formula I according to claim 1 or its pharmaceutically acceptable salt, which is characterized in that pharmaceutically Acceptable salt refers to compounds of formula I and the acid-addition salts that pharmaceutically acceptable acid is formed, and the acid includes: chlorination Hydrogen, hydrogen bromide, sulfuric acid, carbonic acid, oxalic acid, citric acid, succinic acid, tartaric acid, phosphoric acid, lactic acid, pyruvic acid, acetic acid, maleic acid, first Sulfonic acid, benzene sulfonic acid, p-methyl benzenesulfonic acid or ferulic acid.
5. compound described in general formula I or its pharmaceutically acceptable salt are most preferably following any structure:
6. the preparation method of compound shown in general formula I:
Wherein: R1、R2, n definition with described in claim 1;
By the process of compound II prepare compound IV, by the chlorine atom in mercaptan acid III and compound II through affine substitution Reaction obtains;By the process of compound IV prepare compound I, it is condensed to yield by substituted aniline V with carboxyl.
7. a kind of pharmaceutical composition, wherein the compound containing claim 1 or its pharmaceutically acceptable salt and pharmaceutically may be used The carrier of receiving.
8. compound described in claim 1 or its pharmaceutically acceptable salt or pharmaceutical composition as claimed in claim 6 are being made Application in standby 1 inhibitor of indole amine 2,3-dioxygenase.
9. compound described in claim 1 or its pharmaceutically acceptable salt or pharmaceutical composition as claimed in claim 6 are being made The application being ready for use in the drug for the immunosuppressive related disease that treatment indole amine 2,3-dioxygenase 1 mediates.
10. application according to claim 9, which is characterized in that the immune suppression that the indoleamine 2,3-dioxygenase 1 mediates The related disease of system includes cancer, virus infection, neurodegenerative disease, cataract, organ-graft refection, depression or itself exempts from Epidemic disease disease.
CN201811557706.2A 2018-12-19 2018-12-19 Cyanoquinoline IDO1 inhibitor, and preparation method and application thereof Active CN109438347B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811557706.2A CN109438347B (en) 2018-12-19 2018-12-19 Cyanoquinoline IDO1 inhibitor, and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811557706.2A CN109438347B (en) 2018-12-19 2018-12-19 Cyanoquinoline IDO1 inhibitor, and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN109438347A true CN109438347A (en) 2019-03-08
CN109438347B CN109438347B (en) 2022-09-06

Family

ID=65560311

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811557706.2A Active CN109438347B (en) 2018-12-19 2018-12-19 Cyanoquinoline IDO1 inhibitor, and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN109438347B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110092752A (en) * 2019-05-15 2019-08-06 三峡大学 A kind of quinoline fluorescent chemicals, preparation method and applications

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014207213A1 (en) * 2013-06-28 2014-12-31 Medizinische Universität Innsbruck Novel inhibitors of protein kinase c epsilon signaling
US20150140071A1 (en) * 2013-11-12 2015-05-21 Ayyappan K. Rajasekaran Kinase inhibitors
CN109574920A (en) * 2018-12-25 2019-04-05 药大制药有限公司 - 6 cyclopropyl pyridine class IDO1 inhibitor of 3- itrile group and application thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014207213A1 (en) * 2013-06-28 2014-12-31 Medizinische Universität Innsbruck Novel inhibitors of protein kinase c epsilon signaling
US20150140071A1 (en) * 2013-11-12 2015-05-21 Ayyappan K. Rajasekaran Kinase inhibitors
CN109574920A (en) * 2018-12-25 2019-04-05 药大制药有限公司 - 6 cyclopropyl pyridine class IDO1 inhibitor of 3- itrile group and application thereof

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
ACS.REGISTRY DATABASE: "D1", 《STNEXT》 *
ANNA KOHLMANN ET AL.: "Fragment Growing and Linking Lead to Novel Nanomolar Lactate Dehydrogenase Inhibitors", 《JOURNAL OF MEDICINAL CHEMISTRY》 *
ETIFY ABDEL-GHAFAR BAKHITE: "Synthesis and Reactions of Some New 3-amino-2-substituted Thieno[2,3-b]quinolines", 《CHEM.COMMUN》 *
JING LU ET AL.: "A Novel Anti-Tumor Inhibitor Identified by Virtual Screen with PLK1 Structure and Zebrafish Assay", 《PLOS ONE》 *
STEPHANIE M. STANFORD ET AL.: "Discovery of a Novel Series of Inhibitors of Lymphoid Tyrosine Phosphatase with Activity in Human T Cells", 《JOURNAL OF MEDICINAL CHEMISTRY》 *
叶发青 等: "《药物化学》", 30 June 2012, 浙江大学出版社 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110092752A (en) * 2019-05-15 2019-08-06 三峡大学 A kind of quinoline fluorescent chemicals, preparation method and applications
CN110092752B (en) * 2019-05-15 2022-07-15 三峡大学 Quinoline fluorescent compound, preparation method and application thereof

Also Published As

Publication number Publication date
CN109438347B (en) 2022-09-06

Similar Documents

Publication Publication Date Title
Altıntop et al. Design, synthesis, in vitro and in silico evaluation of a new series of oxadiazole-based anticancer agents as potential Akt and FAK inhibitors
JP6261340B2 (en) Use of hematopoietic growth factor mimics
WO2011043359A1 (en) Pharmaceutical product containing aromatic heterocyclic compound
Lai et al. 1-Arylsulfonyl indoline-benzamides as a new antitubulin agents, with inhibition of histone deacetylase
BG109688A (en) Carboxyaryl substituted diphenil ureas as raf kinase inhibitors
Xi et al. Novel 5-anilinoquinazoline-8-nitro derivatives as inhibitors of VEGFR-2 tyrosine kinase: synthesis, biological evaluation and molecular docking
Lv et al. Design, synthesis and biological evaluation of novel 4-alkynyl-quinoline derivatives as PI3K/mTOR dual inhibitors
KR20170095964A (en) Piperidine derivatives as hdac1/2 inhibitors
JP2020512399A (en) IDO inhibiting compounds, their preparation and their use
CA2754085A1 (en) Novel hydroxamate derivative, a production method for the same, and a pharmaceutical composition comprising the same
CN103467359A (en) Cinnamon amides histone deacetylase inhibitor with benzpyrole and preparation method and application of same
EP2272509A1 (en) New Uses of metabotropic glutamate receptors
EA039144B1 (en) New heteroaryl amide derivatives as selective inhibitors of histone deacetylases 1 and/or 2 (hdac1-2)
AU2014283774B2 (en) 1,3-diaminocyclopentane carboxamide derivatives
CN107428705B (en) Histone deacetylase inhibitor and preparation method and application thereof
CN107056755A (en) Five-ring heterocycles amide-type WNT pathway inhibitors
Abbhi et al. Design and synthesis of benzimidazole-based Rho kinase inhibitors for the treatment of glaucoma
CN102746213B (en) Cinnamamide histone deacetylase inhibitor, preparation method thereof, and application thereof
CN105308042A (en) Aryl sultam derivatives as rorc modulators
CN109438347A (en) A kind of cyano quinolines class IDO1 inhibitor, preparation method and application
CN102149678A (en) Novel ortho-aminoanilides for the treatment of cancer
CN103664734B (en) Heterocycle hydroximic acid compound and medicinal compositions thereof and application
CN105616408B (en) Purposes of pyrido [3,4-b] indole derivatives as IDO inhibitor
CN110317137B (en) 9, 10-anthraquinone compound or pharmaceutically acceptable salt thereof and pharmaceutical application thereof
CN113248481B (en) EZH2 covalent irreversible inhibitor, preparation method and application thereof

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