CN115677665A - Biphenyl-containing derivative and medical application thereof - Google Patents

Biphenyl-containing derivative and medical application thereof Download PDF

Info

Publication number
CN115677665A
CN115677665A CN202110843500.1A CN202110843500A CN115677665A CN 115677665 A CN115677665 A CN 115677665A CN 202110843500 A CN202110843500 A CN 202110843500A CN 115677665 A CN115677665 A CN 115677665A
Authority
CN
China
Prior art keywords
compound
acid
arh
pharmaceutically acceptable
substituted
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
CN202110843500.1A
Other languages
Chinese (zh)
Other versions
CN115677665B (en
Inventor
徐云根
张宏波
朱启华
夏宇
吴昊哲
张易炜
于春秋
刘茈涵
李雯佳
孙雨情
张晶晶
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.)
China Pharmaceutical University
Original Assignee
China Pharmaceutical University
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 China Pharmaceutical University filed Critical China Pharmaceutical University
Priority to CN202110843500.1A priority Critical patent/CN115677665B/en
Priority claimed from CN202110843500.1A external-priority patent/CN115677665B/en
Publication of CN115677665A publication Critical patent/CN115677665A/en
Application granted granted Critical
Publication of CN115677665B publication Critical patent/CN115677665B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Abstract

The invention relates to the field of pharmaceutical chemistry, and discloses biphenyl derivatives with PD-1/PD-L1 inhibitory activity and medical application thereof. The invention also discloses a composition containing the biphenyl derivative with the PD-1/PD-L1 inhibitory activity or pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier, and application of the composition in preparation of a PD-1/PD-L1 inhibitor, wherein the composition can be used for treating various cancers or tumors related to the immune checkpoint PD-1/PD-L1 and has wide applicability.

Description

Biphenyl-containing derivative and medical application thereof
Technical Field
The invention belongs to the field of medicinal chemistry, and particularly relates to derivatives containing biphenyl structures and application of the compounds in preparation of medicaments for treating tumors.
Background
In recent years, tumor immunotherapy, which kills tumor cells by using the autoimmune system of human body, has become the focus of the tumor treatment field, unlike the traditional treatment means that directly aims at tumor cells. Activation of the immune checkpoint pathway inhibits activation of T cells, prevents over-activation of the human immune system, maintains immune tolerance of normal organisms, and avoids the occurrence of autoimmune diseases. Tumor cells express an excessive amount of negative immune checkpoint protein, which, upon binding to lymphocytes, over-activates the immune checkpoint pathway leading to the development of tumor immune escape. Among these immune checkpoints, over-activation of PD-1/PD-L1 plays a crucial role in tumor development. When the blocking agent is used for blocking the interaction of PD-1/PD-L1, the immune cells can be identified again and the tumor cells can be killed, so that the aim of treating the tumor is fulfilled. Currently, the PD-1/PD-L1 monoclonal antibody has achieved excellent results in Clinical practice, and shows good therapeutic effects in the treatment of tumors such as melanoma, colon cancer, non-small cell lung cancer, etc. (Clinical and Translational Oncology,2019,21, 702-712 the oncologist,2019,24 (Suppl 1): S31-S41; human Vaccines & immunotherapy, 2014,10 (11): 3111-6; journal of medical chemistry,2020,63, (22): 13825-13850.
In recent years, various PD-1/PD-L1 monoclonal antibody medicaments have been approved to be on the market, and the monoclonal antibody medicaments have made breakthrough progress in the clinical treatment of various tumors. Survival was significantly prolonged in many tumor patients, and partial patients were completely remitted. Although the clinical effect of the monoclonal antibody drug is remarkable, the long half-life period and the long time of binding with the target point can cause serious immune-related adverse reactions. The production process of the monoclonal antibody medicament is complex, the price is high, the storage and the transportation are inconvenient, and common patients can only expect the 'medicament' exclamation. Compared with monoclonal antibodies, small molecule drugs have the advantages of low price, oral administration, crossing biological barriers, convenient transportation and storage, good membrane permeability and non-immunogenicity, and the like. At present, no PD-1/PD-L1 small molecule inhibitor is on the market, so that the development of the inhibitor of the interaction of PD-1/PD-L1 protein and protein has great practical significance and potential application prospect.
Disclosure of Invention
The invention aims to: aiming at the prior art, the invention provides biphenyl derivatives with PD-1/PD-L1 inhibitory activity, a preparation method thereof and pharmaceutical application of the biphenyl derivatives as a PD-1/PD-L1 protein-protein interaction inhibitor.
The technical scheme is as follows: the invention discloses a biphenyl derivative shown as a general formula (I) or pharmaceutically acceptable salt thereof:
Figure BDA0003179708730000021
wherein:
x, Y and Z each independently represent: n or CH;
a represents substituted phenyl or aromatic heterocyclic radical, the aromatic heterocyclic radical is a five-membered or six-membered aromatic ring containing 1-3O, N or S atoms, and the substituent is H, F, cl, br, CN, NH 2 、OH、CF 3 、OCF 3 、C 1 ~C 4 Alkyl or C of 1 -C 4 Alkoxy group of (a);
m =0, 1 or 2; n =0, 1 or 2;
R 1 and R 2 Each independently represents NR 6 R 7 、OR 7 Or substituted quaternary, pentanary or hexanary heterocycloalkyl containing 1-2O or N atoms
R 6 Represents hydrogen or C 1 ~C 3 Alkyl groups of (a);
R 7 represents substituted C 1 ~C 6 The substituent is OH and NH 2 、COOH、CONH 2 、COOCH 3 、COOCH 2 CH 3 、C 1 ~C 4 The alkoxy group of (b) may be mono-or poly-substituted;
the substituted quaternary, quinary or hexahydric heterocycloalkyl containing 1-2O or N atoms is substituted tetrahydropyrrole-1-yl, substituted piperidine-1-yl, substituted morpholine-1-yl, substituted piperazine-1-yl or substituted azetidine-1-yl, wherein the substituent is OH, NH 2 、COOH、CONH 2 、COOCH 3 、COOCH 2 CH 3 、CF 3 、OCF 3 、C 1 ~C 4 Alkoxy group of (1), C 1 ~C 4 The alkyl group of (a), may be mono-or poly-substituted;
R 3 and R 4 Each independently represents H, F, cl, br, CN, CF 3 、C 1 ~C 3 Alkyl or cyclopropyl of (a);
R 5 represents H, F, cl, br, CN, CF 3 、OCH 3 、OCH 2 CH 3 、OCF 3 、C 1 ~C 4 Alkyl or cyclopropyl.
When m =1,a is a 1, 4-disubstituted 1,2, 3-triazazole ring, the compounds of the invention are preferably of the following general formula (II):
Figure BDA0003179708730000022
wherein: x, Y, Z, R 1 、R 2 、R 3 、R 4 、R 5 And n is as defined above.
n is preferably 0 or 1.
X is preferably N or CH.
Y and Z are preferably CH.
R 1 And R 2 Each preferably OH,
Figure BDA0003179708730000031
Wherein R is 8 Represents CH 3 、CH 2 CH 3 、CH 2 CH 2 OH, formyl, acetyl, cyclopropyl, and the like; r 9 And R 10 Each independently represents H, OH, COOH, CH 2 COOH、CH 2 NH 2 、CH 2 OH、CH 2 CH 2 OH、F、Cl、Br、CH 3 、CH 2 CH 3 Cyclopropyl and the like; r 11 Represents OH, NH 2 、NHCH 3 、NHCH 2 CH 3 、CH 3 、OCH 3 、OCH 2 CH 3 ;R 12 Represents CONH 2 、NHCOCH 3 、OH、CH 2 OH、CH 2 CH 2 OH、COOH、CH 2 COOH, etc.; r 13 Represents H, CH 3 、CH 2 CH 3 、CH 2 OH、CH 2 CH 2 OH and the like; r 14 And R 15 Each independently represents H, COOH, NH 2 、F、Cl、Br、CH 3 、CH 2 CH 3 、CH 2 OH、CH 2 CH 2 OH、CONH 2 Cyclopropyl and the like; w represents CH 2 、O、NH、N-CH 3 、N-CH 2 CH 3 、N-CH 2 CH 2 OH、N-COCH 3 Etc.; p represents 0 or 1.
R 3 And R 4 Each is preferably F, cl or CH 3
R 5 Preferably H, F, CH 3 Or OCH 3
Wherein:
x is more preferably N.
R 1 And R 2 More preferably-OH, -NHCH 2 CONH 2 、-NHCH 2 CH 2 OH、
Figure BDA0003179708730000032
Figure BDA0003179708730000033
R 3 And R 4 More preferably CH 3
R 5 More preferably OCH 3
The pharmaceutically acceptable salt of the compound is an acid addition salt of the compound shown in the general formula (I) or (II), wherein the acid for forming the salt is: 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, benzenesulfonic acid, p-toluenesulfonic acid or ferulic acid.
The compounds of the general formula (I) according to the invention can be prepared by the following process:
when X represents N or CH; y and Z represent CH; a represents a 1, 4-disubstituted 1,2, 3-triazazole ring; m =1; when n =0 or 1, the compound (I) can be prepared by the following method:
Figure BDA0003179708730000041
wherein: x, n, R 1 、R 2 、R 3 、R 4 And R 5 The definition of (2) is as before.
The compound V is prepared by the suzuki reaction of a compound III and a compound IV, and the used solvent is selected from toluene, N-dimethylformamide, N-dimethylacetamide, ethylene glycol dimethyl ether, ethylene glycol monomethyl ether, 1, 4-dioxane, tetrahydrofuran, methanol, ethanol, acetonitrile, acetone, water or a mixed solvent consisting of any two solvents, preferably a mixed solvent of 1, 4-dioxane and water; the base is selected from sodium ethoxide, potassium acetate, sodium hydroxide, potassium carbonate, sodium bicarbonate, potassium bicarbonate, sodium carbonate or tripotassium phosphate, preferably potassium carbonate; the catalyst is selected from Pd (PPh) 3 ) 4 、Pd(dppf)Cl 2 、Pd(PPh 3 ) 2 Cl 2 、Pd(OAc) 2 Or NiCl 2 (dppf), preferably Pd (PPh) 3 ) 4 (ii) a The reaction temperature is selected from 50 to 120 ℃, preferably 60 to 100 ℃.
The compound V reacts with sodium nitrite to obtain diazonium salt after being salified with sulfuric acid, and finally reacts with the diboron pinacol ester to prepare the compound VI, wherein the used solvent is selected from methanol, ethanol, tetrahydrofuran, 1, 4-dioxane, ethylene glycol dimethyl ether, acetonitrile, water or a mixed solvent consisting of the solvents, preferably the mixed solvent of methanol and water.
The compound VI and the compound VII are subjected to suzuki reaction to prepare a compound VIII, and the used solvent is selected from toluene, N-dimethylformamide, N-dimethylacetamide, ethylene glycol dimethyl ether, ethylene glycol monomethyl ether, 1, 4-dioxane, tetrahydrofuran, methanol, ethanol, acetonitrile, acetone, water or a mixed solvent consisting of any two solvents, preferably 1, 4-dioxane and water; the base is selected from sodium ethoxide, potassium acetate, sodium hydroxide, potassium carbonate, sodium bicarbonate, potassium bicarbonate, tripotassium phosphate, sodium carbonate or triethylamine, preferably potassium carbonate; the catalyst used was Pd (PPh) 3 ) 4 、Pd(dppf)Cl 2 、Pd(PPh 3 ) 2 Cl 2 、Pd(OAc) 2 Or NiCl 2 (dppf), preferably Pd (PPh) 3 ) 4 . The reaction temperature is selected from 50 to 120 ℃, preferably 60 to 100 ℃.
Preparing a target compound II by carrying out reductive amination reaction on a compound VIII and corresponding amine, wherein the used solvent is selected from dichloromethane, dichloroethane, trichloromethane, carbon tetrachloride, tetrahydrofuran, methanol, toluene, ethanol, acetonitrile, N-dimethylformamide or a mixed solvent consisting of any two or three solvents, preferably dichloromethane or a mixed solvent of dichloromethane and methanol; the reducing agent is selected from sodium borohydride, sodium cyanoborohydride and sodium triacetoxyborohydride, preferably sodium triacetoxyborohydride; the reaction temperature is selected from 0 to 80 ℃ and preferably from 25 to 40 ℃.
The application of the compound (including chiral isomer) with the general formula (I) and the hydrate, solvate or crystal thereof in the preparation of PD-1/PD-L1 inhibitor drugs is also in the protection scope of the invention.
Furthermore, the PD-1/PD-L1 inhibitor can be used for preparing medicaments for treating cancers such as non-small cell lung cancer, colon cancer, melanoma and the like.
Pharmacological experiments show that the biphenyl derivatives can generate good inhibition effect on the interaction of PD-1/PD-L1 in homogeneous phase time-resolved fluorescence experiments (HTRF). In a surface plasmon resonance experiment, the biphenyl derivatives have good affinity to human PD-L1. The biphenyl derivative can well inhibit the combination of PD-1/PD-L1, promote the recovery of T cell activity and promote the secretion of an immune factor INF-gamma, so that the biphenyl derivative can be used for immunotherapy of tumors. The biphenyl derivative has excellent activity, so that the development of the biphenyl inhibitor of PD-1/PD-L1 has great practical significance and potential application prospect.
Has the beneficial effects that: compared with the prior art, the invention has the following remarkable advantages: (1) The novel biphenyl derivative can obviously inhibit the interaction of PD-1/PD-L1, and particularly importantly, can obviously block the interaction of PD-L1 to CD3 + The inhibiting effect of T cells has the effect of promoting the T cells to secrete the immune factor INF-gamma. In the experiment of co-incubation of tumor cells and T cells, the biphenyl compound has higher effect on INF-gamma expression promotion than positive drug BMS-202, so that the biphenyl compound has the effect of enhancing the anti-tumor effect of the T cells; therefore, the biphenyl derivatives can be used as an immune checkpoint PD-1/PD-L1 inhibitor for preparing a medicament for tumor immunotherapy. (2) The synthetic route of the biphenyl derivatives is ingenious in design, simple and feasible, the raw materials are cheap and easy to obtain, the synthetic process is safe and environment-friendly, and the large-scale production is easy. (3) The medicine taking the compound as an active ingredient can be used for treating various cancers or tumors related to the immune checkpoint PD-1/PD-L1, and has wide applicability.
Drawings
FIG. 1 is an INF-gamma expression assay in co-culture of tumor cells and T cells.
Detailed Description
The present invention will be described in detail with reference to specific examples.
Example 1
N, N ' - ((((((((2, 2' -dimethyl- [1,1' -biphenyl)) s)]-3,3' -diyl) bis (2-methoxypyridine-6, 3-diyl))Bis (methylene)) bis (1H-1, 2, 3-triazol-1, 4-diyl)) bis (methylene)) bis (azenediyl)) bis (ethane-2, 1-diyl)) diethylamide (II-1: n =1,X = N,R 1 =R 2 =-NHCH 2 CH 2 NHCOCH 3 ,R 3 =R 4 =CH 3 ,R 5 =OCH 3 )
Synthesis of 2,2' -dimethyl- [1,1' -biphenyl ] -3,3' -diamine (V)
3-bromo-2-methylaniline (III) (10.00g, 53.8mmol) and Compound IV (12.50g, 53.8mmol) were dissolved in dioxane (150 mL), a solution of potassium carbonate (20.83g, 150.6 mmol) in water (15 mL) was added dropwise to the reaction mixture under nitrogen, and Pd (PPh) was added thereto 3 ) 4 (1.55g, 1.35mmol), warmed to 80 ℃ for 12 hours, TLC (petroleum ether: ethyl acetate =8 1) monitors the completion of the reaction of the starting materials, stops heating, and cools to room temperature. Insoluble matter was removed by suction filtration, and the filtrate was diluted with water (100 mL), extracted with ethyl acetate (120 mL. Times.3), washed with saturated sodium chloride solution (100 mL. Times.3), and dried over anhydrous sodium sulfate. And (3) carrying out suction filtration, carrying out reduced pressure evaporation on the filtrate to remove the solvent to obtain a crude product, and purifying by column chromatography (petroleum ether 100% -petroleum ether: ethyl acetate = 15). m.p.134-136 ℃. 1 H NMR(400MHz,Chloroform-d)δ7.07(t,J=7.7Hz,2H,ArH),6.72(d,J=7.9Hz,2H,ArH),6.63(d,J=7.6Hz,2H,ArH),3.37(br,4H,NH 2 ),1.90(s,6H,CH 3 )。
Synthesis of 2,2'- (2, 2' -dimethyl- [1,1 '-biphenyl ] -3,3' -diyl) bis (4, 5-tetramethyl-1, 3, 2-dioxaborane) (VI)
Compound V (4.00g, 18.9mmol) was dissolved in methanol (40 mL), and HCl solution (37.7mL, 3mol/L) and water (20 mL) were added successively, followed by stirring at room temperature for 30 minutes. After cooling to 0 ℃, slowly adding a sodium nitrite aqueous solution (18.86mL, 2.2mol/L) dropwise, keeping the temperature and stirring for 30 minutes continuously after dropwise adding, dissolving bis (pinacol) borate (19.20g, 75.6 mmol) in methanol (40 mL), then slowly adding dropwise into the reaction solution, returning to room temperature and stirring for 2 hours after dropwise adding, and monitoring by TLC (petroleum ether: ethyl acetate = 30) that the raw materials are completely reacted. Diluting with dichloromethane (50 mL), separating, extracting the aqueous phase with dichloromethane (50 mL. Times.2), combining the dichloromethane layers, and dissolving with saturated sodium chlorideThe solution was washed (50 mL. Times.3) and dried over anhydrous magnesium sulfate. Suction filtration, reduced pressure evaporation of the filtrate to remove the solvent to obtain a crude product, and purification by column chromatography (petroleum ether: ethyl acetate = 250). m.p.167-168 ℃. 1 H NMR(300MHz,Chloroform-d)δ7.77(dd,J=7.3,1.7Hz,2H,ArH),7.23(t,J=7.4Hz,2H,ArH),7.15(dd,J=7.5,1.7Hz,2H,ArH),2.23(s,6H,ArCH 3 ),1.38(s,24H,CH 3 )。
Synthesis of 1- ((6-chloro-2-methoxypyridin-3-yl) methyl) -1H-1,2, 3-triazole-4-carbaldehyde (VII-1)
Figure BDA0003179708730000061
Synthesis of 6-chloro-2-methoxypyridine-3-carbaldehyde (2)
6-chloro-2-methoxypyridine (2.00g, 13.9 mmol) was dissolved in anhydrous THF (10 mL) under nitrogen. The temperature was reduced to-10 ℃ and isopropyl magnesium chloride (6.95mL, 6.95mmol, 1.00mol/L) was slowly injected. After dropping, the reaction was carried out at-10 ℃ for 30 minutes. N-butyllithium (6.67mL, 16.7mmol, 2.50mol/L) was injected further at-10 ℃. After dropping was completed, stirring was continued at this temperature for 2 hours, and then anhydrous DMF (2.21mL, 27.8mmol) was added dropwise. After dropping, stirring was continued for 1.5 hours. TLC monitored the starting material reaction to completion and warmed to room temperature. The reaction mixture was slowly added to a mixture of acetic acid (5.00 g), concentrated hydrochloric acid (2.00 g), isopropyl alcohol (20.00 mL) and water (20.00 mL), and after quenching the reaction, the temperature was raised to 50 ℃ for 2 hours. Cooling to room temperature, evaporating to remove the organic solvent, cooling the filtrate, precipitating a solid, performing suction filtration to obtain a yellow solid, and performing vacuum drying to obtain 1.58g of compound 2, wherein the yield is 66.1%. m.p.62-64 ℃. 1 H NMR(300MHz,DMSO-d 6 )δ10.18(s,1H,CHO),8.13(d,J=7.9Hz,1H,ArH),7.28(d,J=7.9,1H,ArH),4.02(s,3H,OCH 3 ).
Synthesis of (6-chloro-2-methoxy) -3-pyridinemethanol (3)
Compound 2 (2.20g, 12.8mmol) was dissolved in anhydrous tetrahydrofuran (8.00 mL) and anhydrous methanol (3.00 mL). The temperature was reduced to 0 ℃ and sodium borohydride (0.59g, 15.4 mmol) was added slowly in portions. After the addition, the ice bath was removed, and the mixture was left at room temperature and stirred for 30min. TLC (Stone)Oil ether: ethyl acetate = 8) monitor the completion of the starting reaction, stop the reaction, and quench the reaction with water. The organic solvent was evaporated under reduced pressure, the residue was extracted with ethyl acetate (10 mL. Times.3), and the organic phases were combined, washed with a saturated sodium chloride solution (20 mL. Times.3), and dried over anhydrous sodium sulfate. The anhydrous sodium sulfate was removed by suction filtration, the solvent was distilled off to obtain a crude product, which was separated by column chromatography to obtain 1.64g of a white solid powder with a yield of 73.7%. m.p.58-61 ℃. 1 H NMR(300MHz,Chloroform-d)δ7.56(d,J=7.5,1H,ArH),6.92(d,J=7.6Hz,1H,ArH),4.63(s,2H,CH 2 ),4.00(s,3H,OCH 3 )。
Synthesis of 6-chloro-3- (chloromethyl) -2-methoxypyridine hydrochloride (4)
Compound 3 (11.80g, 68.0 mmol) was dissolved in dichloromethane (70 mL), thionyl chloride (6.01mL, 81.6 mmol) was slowly added dropwise, and the reaction was stirred overnight at room temperature from cloudy to clear. TLC (petroleum ether: ethyl acetate = 8): 1) monitored completion of the reaction of the starting materials, toluene (20 mL) was added to the reaction solution, the solvent was distilled off under reduced pressure, and after cooling, 15.34g of pale green crystals were obtained, with a yield of 98.8%. m.p.62-64 ℃. MS (ESI) M/z [ M + H ]]+.Calcd for C 7 H 8 Cl 2 NO:191.9;Found:192.0。
Synthesis of 3- (azidomethyl) -6-chloro-2-methoxypyridine (5)
Compound 4 (15.34g, 79.9mmol) and DIPEA (15.49g, 119.9mmol) were added to DMF (150 mL) and stirred at room temperature for 1 hour. CsF (14.57g, 95.9mmol) and TMSN were added 3 (12.6mL, 95.9 mmol), and the temperature is raised to 80 ℃ for reaction for 5 hours. TLC (petroleum ether: ethyl acetate = 30) monitored the completion of the starting material reaction. Stopping heating, and cooling to room temperature. The reaction mixture was poured into ice water (300 mL), extracted with ethyl acetate (150 mL. Times.3), and the organic layers were combined, washed with saturated sodium chloride (100 mL. Times.3), and dried over anhydrous sodium sulfate. The anhydrous sodium sulfate is removed by suction filtration, the solvent is removed by evaporation to obtain a crude product, and the crude product is separated by column chromatography to obtain 13.06g of colorless transparent oily liquid with the yield of 82.3 percent. MS (ESI) M/z [ M + H ]] + Calcd for C 7 H 8 ClN 4 O:199.0;Found:199.1。
Synthesis of 6-chloro-3- ((4- (diethoxymethyl) -1H-1,2, 3-triazol-1-yl) methyl) -2-methoxypyridine (6)
Compound 5 (12.50g, 62.9 mmol) and propionaldehyde diethyl acetal (9.67g, 75.5 mmol) were dissolved in acetonitrile (72 mL) and water (24 mL), and CuI (0.60g, 3.16mmol) was added. The temperature is raised to 30 ℃ for reaction for 12 hours. TLC (petroleum ether: ethyl acetate = 4) monitored the completion of the starting material reaction, stopped heating, and cooled to room temperature. Poured into ice water (50 mL), extracted with ethyl acetate (100 mL. Times.2), the organic layers were combined, washed with saturated aqueous sodium chloride (100 mL. Times.2), and dried over anhydrous sodium sulfate. Insoluble matter was removed by suction filtration, and the solvent was distilled off to give 15.53g of a colorless transparent oily liquid, which was used in the next reaction without purification.
Synthesis of 1- ((6-chloro-2-methoxypyridin-3-yl) methyl) -1H-1,2, 3-triazole-4-carbaldehyde (VI I-1)
Compound 6 (15.00g, 45.9mmol) was dissolved in dichloromethane (50 mL) and trifluoroacetic acid (6.82mL, 91.8mmol) was slowly added dropwise. After dropping, the reaction was carried out at room temperature for 1 hour. TLC (petroleum ether: ethyl acetate = 2). The pH was adjusted to alkaline with 2N NaOH solution, extracted with dichloromethane (50 mL. Times.3), and the organic layers were combined, washed with saturated NaCl solution (50 mL. Times.2), and dried over anhydrous sodium sulfate. The anhydrous sodium sulfate was removed by suction filtration, the solvent was distilled off to obtain a crude product, which was purified by column chromatography to obtain 11.14g of a white solid powder with a yield of 96.1%. m.p.128-130 ℃. 1 H NMR(300MHz,Chloroform-d)δ10.20(s,1H,CHO),8.22(s,1H,ArH),7.61(d,J=7.7Hz,1H,ArH),7.03(d,J=7.7Hz,1H,ArH),5.60(s,2H,CH 2 ),4.09(s,3H,OCH 3 ).
Synthesis of 1,1'- ((((2, 2' -dimethyl- [1,1 '-biphenyl ] -3,3' -diyl) bis (2-methoxypyridine-6, 3-diyl)) bis (methylene)) bis (1H-1, 2, 3-triazole-4-carbaldehyde) (VIII-1)
Mixing compound VI I-1 (1.42g, 3.27mmol), compound V I (2.06g, 8.17mmol), pd (PPh) 3 ) 4 (0.38g,0.33mmol)、K 2 CO 3 A water (2 mL) (1.27g, 9.17mmol) solution and dioxane (20 mL) are sequentially added into an eggplant-shaped bottle, nitrogen is used for protection, and the temperature is raised to 80 ℃ and stirring reaction is carried out for 12 hours. TLC (petroleum ether: ethyl acetate = 2). The insoluble matter was removed by suction filtration, extracted with ethyl acetate (50 mL. Times.3), and the organic layers were combined and washed with saturated sodium chloride solution (50 m)L × 2) and dried over anhydrous sodium sulfate. Insoluble materials were removed by suction filtration, and the solvent was evaporated under reduced pressure to give a crude product, which was purified by column chromatography (petroleum ether: ethyl acetate = 4. m.p.179-181 ℃. 1 H NMR(400MHz,DMSO-d 6 )δ10.05(s,2H,CHO),8.96(s,2H,ArH),7.70(d,J=7.6Hz,2H,ArH),7.45(dd,J=7.7,1.5Hz,2H,ArH),7.38(t,J=7.6Hz,2H,ArH),7.24–7.21(m,4H,ArH),5.72(s,4H,CH 2 ),3.92(s,6H,OCH 3 ),2.05(s,6H,CH 3 ).
Synthesis of N, N '- ((((((((((2, 2' -dimethyl- [1,1 '-biphenyl ] -3,3' -diyl) bis (2-methoxypyridin-6, 3-diyl)) bis (methylene)) bis (1H-1, 2, 3-triazole-1, 4-diyl)) bis (methylene)) bis (azepinyl)) bis (ethane-2, 1-diyl)) diethylamide (II-1)
Compound VIII-1 (0.20g, 0.33mmol), N-acetylethylenediamine (0.10g, 0.98mmol) and acetic acid (2 drops) were dissolved in methylene chloride (5 mL) and methanol (2 mL), and the reaction was stirred at room temperature for 1 hour. Cooling to 0 deg.C, adding NaBH (OAc) 3 (0.28g, 1.32mmol) was added to the reaction solution in portions, and stirred at room temperature for 4 hours. TLC (dichloromethane: methanol =10 = 1) monitors the completion of the raw material reaction, adjusts pH to 7 with saturated aqueous sodium bicarbonate solution, extracts with dichloromethane (10 mL × 3), combines organic layers, washes with saturated sodium chloride solution (10 mL × 2), and dries over anhydrous sodium sulfate. Filtering to remove insoluble substances, distilling under reduced pressure to remove the solvent to obtain a crude product, and purifying by silica gel column chromatography to obtain yellow solid powder 0.09g with a yield of 34.2%. m.p.86-88 ℃. 1 H NMR(300MHz,DMSO-d 6 )δ8.02(s,2H,ArH),7.83(s,2H,2CONH),7.54(d,J=7.6Hz,2H,ArH),7.46(dd,J=7.7,1.7Hz,2H,ArH),7.39(t,J=7.5Hz,2H,ArH),7.22(td,J=7.4,2.6Hz,4H,ArH),5.60(s,4H,2ArCH 2 ),3.95(s,6H,2OCH 3 ),3.78(s,4H,ArCH 2 NH),3.14(q,J=6.4Hz,4H,2CH 2 NHCH 2 ),2.59(t,J=6.5Hz,4H,2CONHCH 2 ),2.07(s,6H,2ArCH 3 ),1.80(s,6H,2COCH 3 ).HRMS(ESI):m/z[M+H] + Calcd for C 42 H 51 N 12 O 4 :787.4156;Found:787.4149。
Example 2
2,2'-(((((2, 2 '-dimethyl- [1,1' -biphenyl)]-3,3' -diyl) bis (2-methoxypyridin-6, 3-diyl)) bis (methylene)) bis (1H-1, 2, 3-triazol-1, 4-diyl)) bis (methylene)) bis (azepinyl)) bis (ethan-1-ol) (II-2: n =1,X = N,R 1 =R 2 =-NHCH 2 CH 2 OH,R 3 =R 4 =CH 3 ,R 5 =OCH 3 ) Synthesis of (2)
The same operation as that of Compound II-1 was carried out using Compound VII I-1 (0.20g, 0.33mmol) and ethanolamine (0.06g, 0.98mmol) as starting materials to obtain 0.08g of a yellow solid powder with a yield of 34.9%. m.p.112-114 ℃. 1 H NMR(300MHz,DMSO-d 6 )δ8.10(d,J=7.9Hz,2H,ArH),7.55(t,J=7.2Hz,2H,ArH),7.45(d,J=7.6Hz,2H,ArH),7.38-7.28(m,2H,ArH),7.25–7.09(m,4H,ArH),5.65–5.48(m,4H,ArCH 2 ),4.05(s,6H,OCH 3 ),3.90(s,4H,2NHCH 2 Ar),2.54–2.49(m,4H,CH 2 CH 2 OH),2.76–2.66(m,4H,CH 2 CH 2 OH),2.03(s,6H,2ArCH 3 ).HRMS(ESI):m/z[M+H] + Calcd for C 38 H 45 N 10 O 4 :705.3625;Found:705.3615。
Example 3
2,2' - ((((((2, 2' -dimethyl- [1,1' -biphenyl)) s)]-3,3' -diyl) bis (2-methoxypyridine-6, 3-diyl)) bis (methylene)) bis (1H-1, 2, 3-triazole-1, 4-diyl)) bis (methylene)) bis (azediyl)) diethylamide (II-3: n =1,X = N,R 1 =R 2 =-NHCH 2 CONH 2 ,R 3 =R 4 =CH 3 ,R 5 =OCH 3 )
Synthesis of compound II-1 was carried out in the same manner as for compound VII-1 (0.20g, 0.33mmol) and glycylamine hydrochloride (0.15g, 1.32mmol) to give 0.11g of a yellow solid powder, yield 46.3%. m.p.86-88 ℃. 1 H NMR(300MHz,DMSO-d 6 )δ8.08(s,2H,ArH),7.56(d,J=7.5Hz,2H,ArH),7.47(dd,J=7.7,1.7Hz,2H,ArH),7.40(t,J=7.6Hz,2H,ArH),7.34(br,2H,CONH 2 ),7.30–7.18(m,4H,ArH),7.09(s,2H,CONH 2 ),5.61(s,4H,2ArCH 2 ),3.97(s,6H,2OCH 3 ),3.77(s,4H,2ArCH 2 NH),3.10(s,4H,2COCH 2 ),2.65(s,2H,2NH),2.08(s,6H,2ArCH 3 )。HRMS(ESI):m/z[M+H] + Calcd for C 38 H 43 N 12 O 4 :731.3530;Found:731.3528。
Example 4
5,5' - (((((((2, 2' -dimethyl- [1,1' -biphenyl)) s)]-3,3' -diyl) bis (2-methoxypyridine-6, 3-diyl)) bis (methylene)) bis (1H-1, 2, 3-triazole-1, 4-diyl)) bis (methylene)) bis (azediyl)) bis (methylene)) bis (pyrrolidin-2-one) (II-4: n =1,X = N,R 1 =R 2 Is (pyrrolidin-2-one-5-yl) methylamino, R 3 =R 4 =CH 3 ,R 5 =-OCH 3 )
Synthesis of the same compound II-1 was carried out using the compound VII-1 (0.20g, 0.33mmol) and 5-aminomethyl-2-pyrrolidone (0.15g, 1.32mmol) as starting materials to give 0.12g of a yellow solid powder in a yield of 45.5%. m.p.111-114 ℃.1H NMR (300MHz, chloroform-d) delta 7.60 (s, 2H, arH), 7.51 (d, J =7.5Hz,2H, arH), 7.44-7.38 (m, 2H, arH), 7.32 (t, J =7.6Hz,2H, arH), 7.24-7.19 (m, 2H, arH), 7.05 (d, J =7.5Hz,2H, arH), 6.32 (br, 2H, NHCO), 5.54 (s, 4H, arCH 2 N),4.03(s,6H,OCH 3 ),3.94(s,4H,ArCH 2 NH),3.80-3.69(m,2H,2CH),2.86(dd,J=12.0,4.0Hz,2H,NHCH 2 ),2.61(dd,J=12.0,8.5Hz,2H,NHCH 2 ),2.33(t,J=8.0Hz,4H,NHCH 2 CH),2.25-2.17(m,2H,CH 2 ),2.17-2.12(m,2H,CH 2 ),2.11(s,6H,ArCH 3 ),2.08-2.00(m,2H,CH 2 ),1.80-1.69(m,2H,CH 2 ).HRMS(ESI):m/z[M+Na] + Calcd for C 44 H 50 N 12 O 4 Na:833.3976;Found:833.3993。
Example 5
2,2',2", 2'" - ((((((2, 2 '-dimethyl- [1,1' -biphenyl)) s)]-3,3' -diyl) bis (2-methoxypyridin-6, 3-diyl) bis (methylene)) bis (1H-1, 2, 3-triazol-1, 4-diyl)) bis (methylene)) bis (azatriyl)) tetrakis (ethane-1-ol) (II-5: n =1,X = N,R 1 =R 2 =-N(CH 2 CH 2 OH) 2 ,R 3 =R 4 =CH 3 ,R 5 =OCH 3 ) Is/are as followsSynthesis of
Synthesis of the compound II-1 was carried out in the same manner as in the case of the compound VII I-1 (0.20g, 0.33mmol) and diethanolamine (0.13mL, 0.98mmol) as raw materials to obtain 0.11g of a yellow solid powder with a yield of 42.6%. m.p.90-92 ℃. 1 H NMR(300MHz,DMSO-d 6 )δ8.09(s,2H,ArH),7.51(d,J=7.5Hz,2H,ArH),7.46(dd,J=7.7,1.6Hz,2H,ArH),7.38(t,J=7.5Hz,2H,ArH),7.22(td,J=7.5,2.4Hz,4H,ArH),5.60(s,4H,ArCH 2 ),4.43(s,4H,4OH),3.94(s,6H,2OCH 3 ),3.79(s,4H,2NHCH 2 ),3.39(s,4H,2CH 2 CH 2 OH),3.19(d,J=4.3Hz,4H,2CH 2 CH 2 OH),2.06(s,6H,2ArCH 3 )。HRMS(ESI):m/z[M+H] + Calcd for C 42 H 53 N 10 O 6 :793.4150;Found:793.4152。
Example 6
((((2, 2 '-dimethyl- [1,1' -biphenyl)]-3,3' -diyl) bis (2-methoxypyridine-6, 3-diyl)) bis (methylene)) bis (1H-1, 2, 3-triazole-1, 4-diyl)) dimethanol (II-6 1 =R 2 =-OH,R 3 =R 4 =CH 3 ,R 5 =OCH 3 ) Synthesis of (2)
Compound VII I-1 (0.20g, 0.33mmol) was dissolved in methanol (2 mL), and the temperature was decreased to 0 ℃. Reacting NaBH 4 (19mg, 0.51mmol) was added to the reaction solution, and the mixture was stirred for 30 minutes. TLC monitored the completion of the reaction of the starting materials and water was added to quench excess sodium borohydride. Methanol was evaporated under reduced pressure, and the residue was diluted with dichloromethane (20 mL), separated, and the dichloromethane layer was washed with a saturated sodium chloride solution (5 mL. Times.3) and water (5 mL. Times.3), and dried over anhydrous sodium sulfate. Filtering to remove insoluble substances, distilling under reduced pressure to remove solvent to obtain crude product, and performing column chromatography to obtain yellow solid powder 0.17g with yield of 84.5%. m.p.144-146 ℃. 1 H NMR(400MHz,DMSO-d 6 )δ8.03(s,2H,ArH),7.53(d,J=7.6Hz,2H,ArH),7.43(d,J=7.6Hz,2H,ArH),7.36(t,J=7.5Hz,2H,ArH),7.20(dd,J=11.1,6.9Hz,4H,ArH),5.58(s,4H,ArCH 2 N),5.21(t,J=5.7Hz,2H,OH),4.53(d,J=5.6Hz,4H,CH 2 ),3.93(s,6H,OCH 3 ),2.04(s,6H,ArCH 3 )。HRMS(ESI):m/z[M+H] + Calcd for C 34 H 35 N 8 O 4 :619.2781;Found:619.2777。
Example 7
N, N '- (((((3, 3"' -dimethoxy-2 ',2" -dimethyl- [1,1':3',1":3", 1' "-tetraphenyl]-4, 4' "-diyl) bis (1H-1, 2, 3-triazol-1, 4-diyl)) bis (methylene)) bis (azepinyl)) bis (ethane-2, 1-diyl)) diethylamide (II-7: n =0,X = CH,R 1 =R 2 =-NHCH 2 CH 2 NHCOCH 3 ,R 3 =R 4 =CH 3 ,R 5 =OCH 3 ) Synthesis of (2)
Synthesis of 1- (4-bromo-2-methoxyphenyl) -1H-1,2, 3-triazole-4-formaldehyde (VII-2)
Figure BDA0003179708730000101
Synthesis of 4-bromo-2-methoxyaniline (8)
Ortho-anisidine 7 (20.00g, 162.4 mmol) was dissolved in acetonitrile (100 mL). The temperature was lowered to 0 ℃ and N-bromosuccinimide (33.8g, 195.0 mmol) was slowly added to the reaction mixture in portions. After the addition was completed, the reaction was carried out at room temperature overnight. TLC (petroleum ether: ethyl acetate = 15) monitored completion of the raw material reaction, diluted with water, extracted with ethyl acetate (100 mL × 3), combined organic layers, washed with saturated sodium chloride (100 mL × 2), and dried over anhydrous sodium sulfate. The anhydrous sodium sulfate was removed by suction filtration, and the solvent was evaporated under reduced pressure to give a crude product, which was purified by column chromatography (petroleum ether: ethyl acetate = 100. The yield thereof was found to be 60.4%. m.p.60-61 ℃. 1 H NMR(300MHz,Chloroform-d)δ6.95-6.88(m,2H,ArH),6.63(dt,J=8.4,1.1Hz,1H,ArH),3.89(s,3H,OCH 3 ),3.81(s,2H,NH 2 )。
Synthesis of 1-azido-4-bromo-2-methoxybenzene (9)
Compound 8 (6.47g, 32.0 mmol) was dissolved in acetonitrile, and tert-butyl nitrite (3.96g, 38.4 mmol) was slowly added dropwise. After the completion of the dropwise addition, the temperature was lowered to 0 ℃ and trimethylsilyl azide (4.42g, 38.4 mmol) was slowly added dropwise, and after the completion of the dropwise addition, the reaction was carried out at room temperature for 2 hours. TLC (petroleum ether: ethyl acetate = 30). Evaporating the solvent to dryness and removing the residueThe resulting substance was dissolved in methylene chloride and purified by column chromatography to obtain 3.66g of a pale brown transparent oily liquid. The yield thereof was found to be 50.1%. 1 H NMR(300MHz,Chloroform-d)δ7.13(dd,J=8.4,1.6Hz,1H,ArH),7.08(d,J=1.5Hz,1H,ArH),6.93(d,J=8.3,1H,ArH),3.94(s,3H,OCH 3 )。
Synthesis of 1- (4-bromo-2-methoxyphenyl) -4- (diethoxymethyl) -1H-1,2, 3-triazole (10)
Compound 9 (3.88g, 17.0 mmol) was dissolved in acetonitrile (21 mL) and water (7 mL), and propargyl propionaldehyde diethyl acetal (2.61g, 20.4 mmol) was slowly added dropwise. CuI (0.16g, 0.85mmol) was added, and the reaction was allowed to proceed at 30 ℃ for 24 hours. TLC (petroleum ether: ethyl acetate = 4). The reaction mixture was poured into ice water (30 mL), extracted with ethyl acetate (20 mL. Times.2), washed with saturated aqueous sodium chloride (20 mL. Times.2), and the organic layers were combined and dried over anhydrous magnesium sulfate. Insoluble matter was removed by suction filtration, and the solvent was distilled off to leave 5.57g of a yellow oily liquid, which was used in the next reaction without purification.
Synthesis of 1- (4-bromo-2-methoxyphenyl) -1H-1,2, 3-triazole-4-formaldehyde (VII-2)
Compound 10 (5.57g, 15.6 mmol) was dissolved in dichloromethane (20 mL) and trifluoroacetic acid (2.32mL, 31.2mmol) was slowly added dropwise at room temperature. After dropping, the reaction was carried out at room temperature for 1 hour. TLC (petroleum ether: ethyl acetate = 4) the reaction was complete, pH was adjusted to 8 with 2N NaOH, dichloromethane (30 mL × 2) was extracted to no fluorescence, organic layers were combined, washed with saturated NaCl solution (20 mL × 2), dried over anhydrous magnesium sulfate. The anhydrous magnesium sulfate was removed by suction filtration, the solvent was distilled off to obtain a crude product, which was purified by column chromatography (petroleum ether: ethyl acetate = 8. m.p.137-138 ℃. 1 H NMR(300MHz,DMSO-d 6 )δ10.13(s,1H,CHO),9.26(s,1H,ArH),7.69(d,J=8.4Hz,1H,ArH),7.63(d,J=2.1Hz,1H,ArH),7.42(dd,J=8.4,2.0Hz,1H,ArH),3.92(s,3H,OCH 3 )。
1,1'- (3, 3"' -dimethoxy-2 ',2" -dimethyl- [1,1':3',1":3", 1' "-tetraphenyl ] -4,4" -diyl) bis (1H-1, 2, 3-triazole-4-carbaldehyde) (VIII-2) synthesis
Adding compound VI (0.20g, 0.46mmol) and compound VII-2 (0.32g, 1.15mmol) and K 3 PO 4 (0.10 g, 0.46mm) in water (0.5 mL) was dissolved in tetrahydrofuran, and Pd (dppf) Cl was added under nitrogen 2 (34mg, 0.046 mmol) and the reaction was allowed to warm to 65 ℃ for 12 hours. TLC (petroleum ether: ethyl acetate = 2) starting material reacted completely, heating was stopped, and cooling to room temperature. Insoluble matters are removed by suction filtration, and the solvent is evaporated under reduced pressure to obtain a crude product, which is purified by column chromatography (petroleum ether: ethyl acetate = 5. m.p.108-110 ℃. 1 H NMR(400MHz,Chloroform-d)δ10.28(s,2H,CHO),8.80(s,2H,ArH),7.96(d,J=8.1Hz,2H,ArH),7.37(d,J=7.4Hz,2H,ArH),7.33(d,J=1.6Hz,2H,ArH),7.18(dd,J=8.1,1.7Hz,2H,ArH),7.14(d,J=1.7Hz,2H,ArH),3.98(s,6H,OCH 3 ),2.06(s,6H,ArCH 3 )。
N, N ' - (((((3, 3"' -dimethoxy-2 ',2" -dimethyl- [1,1':3',1":3",1"' -tetraphenyl ] -4,4" ' -diyl) bis (1H-1, 2, 3-triazole-1, 4-diyl)) bis (methylene)) bis (azepindiyl)) bis (ethane-2, 1-diyl)) diethylamide (II-7) synthesis
The same procedures as for compound II-1 were repeated except that compound VIII-2 (0.20g, 0.34mmol) and N-acetylethylenediamine (0.14g, 1.37mmol) were used as starting materials to give 0.08g of a yellow solid powder in 30.9% yield. m.p.82-84 ℃. 1 H NMR(300MHz,DMSO-d 6 )δ8.39(s,2H,ArH),7.89(s,2H,CONH),7.71(d,J=8.1Hz,2H,ArH),7.46-7.32(m,6H,ArH),7.26(dd,J=7.2,1.9Hz,2H,ArH),7.18(dd,J=8.1,1.7Hz,2H,ArH),3.94(s,6H,OCH 3 ),3.92(s,4H,ArCH 2 ),3.26-3.19(m,6H,ArHCH 2 NHCH 2 ),2.70(t,J=6.6Hz,4H,CONHCH 2 ),2.06(s,6H,ArCH 3 ),1.83(s,6H,COCH 3 ).HRMS(ESI):m/z[M+H] + Calcd for C 42 H 49 N 10 O 4 :757.3938;Found:757.3940。
Example 8
2,2'- ((((3, 3"' -dimethoxy-2 ',2" -dimethyl- [1,1':3',1":3", 1' "-tetraphenyl]-4, 4' "-diyl) bis (1H-1, 2, 3-triazol-1, 4-diyl)) bis (methylene)) bis (azenediyl)) bis (ethan-1-ol) (II-8: n =0,X = CH,R 1 =R 2 =-NHCH 2 CH 2 OH,R 3 =R 4 =CH 3 ,R 5 =OCH 3 )
The same operation as that of Compound II-1 was carried out using compound VIII-2 (0.20g, 0.34mmol) and ethanolamine (81. Mu.L, 1.37 mmol) as starting materials to obtain 0.10g of a yellow solid powder in a yield of 43.3%. m.p.96-98 deg.C. 1 H NMR(300MHz,DMSO-d 6 )δ8.38(s,2H,ArH),7.69(d,J=8.1Hz,2H,ArH),7.43–7.30(m,6H,ArH),7.23(dd,J=7.1,2.1Hz,2H,ArH),7.16(dd,J=8.1,1.8Hz,2H,ArH),3.92(s,10H,OCH 3 /ArCH 2 NH),3.52(t,J=5.6Hz,4H,2CH 2 OH),2.71(t,J=5.7Hz,4H,2NHCH 2 CH 2 ),2.03(s,6H,2ArCH 3 )。HRMS(ESI):m/z[M+H] + Calcd for C 38 H 43 N 8 O 4 :675.3407;Found:675.3414。
Example 9
2,2'- ((((3, 3"' -dimethoxy-2 ',2" -dimethyl- [1,1':3',1":3", 1' "-tetraphenyl]4, 4' -diyl) bis (1H-1, 2, 3-triazole-1, 4-diyl)) bis (methylene)) bis (azepinyl)) bis (3-hydroxypropionic acid) (II-9: : n =0,X = CH,R 1 =R 2 =-NHCH(CH 2 OH)COOH,R 3 =R 4 =CH 3 ,R 5 =OCH 3 )
The same procedure as for compound II-1 was carried out using compound VIII-2 (0.20g, 0.34mmol) and serine methyl ester hydrochloride (0.21g, 1.37mmol) as starting materials to give 0.10g of a pale green viscous liquid. The resulting pale green liquid (0.10g, 0.13mmol) was dissolved in methanol (1 mL) and tetrahydrofuran (1 mL), and a solution of lithium hydroxide (30mg, 1.27mmol) in water (0.5 mL) was added dropwise, followed by stirring at room temperature overnight. TLC (dichloromethane: methanol =15 = 1) monitored the completion of the reaction of the starting materials, evaporated under reduced pressure to remove the organic solvent, adjusted the pH to about 6 with 0.1M HCl, precipitated a large amount of solid, filtered off with suction to give 50mg of a gray solid with a yield of 19.2%. m.p. > 250 ℃. 1 H NMR(300MHz,DMSO-d 6 )δ8.43(s,2H,ArH),7.69(d,J=8.1Hz,2H,ArH),7.37(q,J=7.3,6.4Hz,4H,ArH),7.32(s,2H,ArH),7.23(d,J=7.4Hz,2H,ArH),7.15(d,J=8.3Hz,2H,ArH),4.09(d,J=13.8Hz,2H,CH 2 OH),3.99(d,J=14.2Hz,2H,CH 2 OH),3.92(s,6H,OCH 3 ),3.62(d,J=5.5Hz,4H,NHCH 2 ),3.19(d,J=5.5Hz,2H,CH),2.01(s,6H,ArCH 3 )。
Example 10
((3, 3' "-dimethoxy-2 ',2" -dimethyl- [1,1':3',1":3", 1' "-tetraphenyl]-4, 4' -diyl) bis (1H-1, 2, 3-triazole-1, 4-diyl) dimethanol (II-10: n =0,X = CH,R 1 =R 2 =-OH,R 3 =R 4 =CH 3 ,R 5 =OCH 3 )
Compound VIII-2 (0.20g, 0.34mmol) was dissolved in methanol (5 mL) and cooled to 0 ℃. Reacting NaBH 4 (19mg, 0.51mmol) was added to the reaction solution, and the mixture was stirred for 30 minutes. TLC monitored the starting material reaction was complete and water quenched with excess sodium borohydride. Methanol was evaporated under reduced pressure, and the residue was diluted with methylene chloride (10 mL), washed with a saturated sodium chloride solution (5 mL. Times.3) and water (5 mL. Times.3), and dried over anhydrous sodium sulfate. The insoluble matter was removed by suction filtration, the solvent was distilled off under reduced pressure to give a crude product, which was purified by column chromatography to give 0.13g of yellow solid powder with a yield of 64.6%. m.p.130-132 ℃. 1 H NMR(300MHz,DMSO-d 6 )δ8.38(s,2H,ArH),7.71(d,J=8.1Hz,2H,ArH),7.42–7.36(m,4H,ArH),7.34(d,J=1.7Hz,2H,ArH),7.27(dd,J=7.1,2.0Hz,2H,ArH),7.19(dd,J=8.1,1.7Hz,2H,ArH),5.34(t,J=5.6Hz,2H,2OH),4.65(d,J=5.4Hz,4H,2ArCH 2 ),3.94(s,6H,2OCH 3 ),2.07(s,6H,2ArCH 3 )。HRMS(ESI):m/z[M+H] + Calcd for C 34 H 33 N 6 O 4 :589.2563;Found:589.2543。
Example 11
1,1'- (((3, 3"' -dimethoxy-2 ',2" -dimethyl- [1,1':3',1":3", 1' "-tetraphenyl]-4,4"' -diyl) bis (1H-1, 2, 3-triazole-1, 4-diyl)) bis (methylene)) bis (piperidine-2-carboxamide) (II-11: n =0,X = CH,R 1 =R 2 Is 2-carbamoylpiperidin-1-yl, R 3 =R 4 =CH 3 ,R 5 =OCH 3 ) Synthesis of (2)
The same procedure as for compound II-1 was repeated except that compound VIII-2 (0.20g, 0.34mmol) and 2-piperidinecarboxamide (0.12g, 1.03mmol) were used as starting materials to give 0.05g of a white solid in 18.1% yield. m.p.146-148 ℃. 1 H NMR(300MHz,DMSO-d 6 )δ8.41(s,2H,ArH),7.73(d,J=8.1Hz,2H,ArH),7.42(d,J=7.2Hz,2H,ArH),7.38(dd,J=7.7,1.9Hz,2H,ArH),7.34(d,J=1.7Hz,2H,ArH),7.31(s,2H,CONH 2 ),7.26(dd,J=7.1,1.9Hz,2H,ArH),7.19(dd,J=8.0,1.7Hz,2H,ArH),7.14(s,2H,CONH 2 ),3.94(s,6H,2OCH 3 ),3.87(d,J=14.1Hz,2H,ArCH 2 ),3.57(d,J=14.2Hz,2H,ArCH 2 ),3.35(s,2H,2CH),2.95(d,J=11.2Hz,2H,1/2Piperidine-CH 2 ),2.78–2.70(m,2H,Piperidine-CH 2 ),2.13(d,J=11.4Hz,2H,1/2Piperidine-CH 2 ),2.06(s,6H,ArCH 3 ),1.78(d,J=12.3Hz,2H,1/2Piperidine-CH 2 ),1.68(d,J=12.7Hz,2H,1/2Piperidine-CH 2 ),1.57(s,2H,1/2Piperidine-CH 2 ),1.51–1.39(m,2H,1/2Piperidine-CH 2 ),1.23(d,J=14.3Hz,2H,1/2Piperidine-CH 2 ).HRMS(ESI):m/z[M+H] + Calcd for C 46 H 53 N 10 O 4 :809.4251;Found:809.4263。
Example 12
2,2',2",2 '" - ((((3, 3' "-dimethoxy-2 ',2" -dimethyl- [1,1':3',1":3", 1' "-tetraphenyl)]-4, 4' "-diyl) bis (1H-1, 2, 3-triazol-1, 4-diyl)) bis (methylene)) bis (azatriyl)) tetrakis (ethan-1-ol) (II-12: n =0,X = CH,R 1 =R 2 =-N(CH 2 CH 2 OH),R 3 =R 4 =CH 3 ,R 5 =OCH 3 ) Synthesis of (2)
The same operation as that for compound II-1 was carried out using compound VIII-2 (0.20g, 0.34mmol) and diethanolamine (0.13mL, 1.37mmol) as starting materials to give 0.08g of a yellow-green solid in 30.7% yield. m.p.92-94 ℃. 1 H NMR(400MHz,DMSO-d 6 )δ8.38(s,2H,ArH),7.70(d,J=8.0Hz,2H,ArH),7.40(t,J=7.4Hz,2H,ArH),7.36(dd,J=7.7,1.9Hz,2H,ArH),7.31(d,J=1.9Hz,2H,ArH),7.24(dd,J=7.3,1.8Hz,2H,ArH),7.16(dd,J=8.1,1.8Hz,2H,ArH),4.40(s,4H,OH),3.92(s,6H,OCH 3 ),3.87(s,4H,2ArCH 2 ),3.50(t,J=6.1Hz,8H,4CH 2 OH),2.59(t,J=6.3Hz,8H,4NCH 2 CH 2 ),2.04(s,6H,2ArCH 3 ).HRMS(ESI):m/z[M+H] + Calcd for C 42 H 51 N 8 O 6 :763.3932;Found:763.3936。
Example 13
The pharmacological experiments and results of part of the compounds of the invention are as follows:
1. evaluation of inhibitory Activity against PD-1/PD-L1
Purpose of the experiment: the activity of the compound of formula (I) inhibiting the PD-1/PD-L1 interaction was tested using the PD-1/PD-L1 binding assay kit (CISBIO).
The experimental principle is as follows: HTRF (Homogeneous Time-Resolved Fluorescence) is a technique used to detect analytes in a pure liquid phase system. The energy transfer of two fluorescent groups is mainly utilized, and the two fluorescent groups are divided into an energy donor europium (Eu +) and an energy acceptor. When the donor is excited externally (e.g., by a flash lamp or laser), and within a sufficient distance from the acceptor, energy can be resonantly transferred to the acceptor, which is excited to a specific wavelength. Using HTRF technology, the assay enables simple, rapid characterization of compounds and antibody blockers in a high-throughput format. By using Eu as a label + (europium) (HTRF energy donor) and XL665 (HTRF energy acceptor) labeled anti-Tag 1 and anti-Tag 2, the interaction between PD-L1 and PD-1 can be detected. Labeling PD-L1 and PD-1 proteins, eu, respectively, with Tag1 and Tag2 + And XL665 bind to PD-L1 and PD-1, respectively, by means of antibodies to form a complex. Eu when PD-L1 and PD-1 are bonded close to each other + Excitation by an external laser triggers a fluorescence resonance energy transfer towards XL665, which in turn emits specifically at 665 nm. This specific signal is proportional to the degree of PD1/PD-L1 interaction. Thus, compounds or antibodies that prevent the PD-1/PD-L1 interaction will result in a decrease in HTRF signal.
Experimental materials: the kit is purchased from PD-1/PD-L1 binding assay kits of CISBIO company; 96-well plate: the CISBIO company was purchased.
Testing an instrument: perkin Elmer, type: and (6) EnVision.
Test compounds: a compound of formula (II). Dissolving by using DMSO, and diluting by using a diluent buffer; the DMSO concentration does not exceed 0.5%.
The experimental process comprises the following steps: PD1/PD-L1 binding assay kits were used. Setting a negative group, a positive group and an administration group, wherein each group comprises 2 multiple wells. Positive control group, add 2 μ L of solvent to 96-well plate; 4uL PD-L1 and 4uL PD-1 after dilution according to the instruction; negative control group, 6. Mu.L of solvent and 4. Mu.L of PD-L1 were added to 96-well plate; administration group 2. Mu.L of the compound of formula (I) to be tested (or positive compound BMS-202), 4. Mu.LPD-L1 and 4. Mu.L of PD-1 were added to a 96-well plate in this order. Centrifuge at 1000rpm for 1 min using a plate membrane seal and incubate for 15 min at room temperature. Then diluting the Buffer to obtain Anti-Tag-Eu3 + And Anti-tag-XL665 were mixed in equal volumes, then 10. Mu.L of the mixture was added to each well, centrifuged at 1000rpm for 1 minute on a plate, and incubated for 2 hours at room temperature. Remove the coversheet film, read the fluorescence intensity of 665nm and 615nm using EnVision, and calculate ratio = Signal 665nm/Signal 620nm 10 4 . IC of Compounds calculated Using Graphpad 50 . BMS-202 in WO2015034820 of BMS company was selected as the positive drug in this experiment, and the activity data is shown in Table 1.
A represents 0.50-10nM; b represents 10.01-100nM; c represents 100.01nM to 1. Mu.M.
TABLE 1 blocking of hPD-1/hPD-L1 by compounds at the protein level
Figure BDA0003179708730000151
Experimental results show that the compound has obvious inhibitory activity of PD-1/PD-L1 protein-protein interaction.
2. Experiment for blocking expression effect of PD-L1 in inhibiting secretion of INF-gamma by T cells
The experimental principle is as follows: the Hep3B-OS8-hPDL1 cells (Shanghai Ruizi chemical research, inc.) stably express hPD-L1 protein on the surface; CD3 + T cells (Shanghai Ruizi chemical research, ltd.) expressed PD-1 on the surface; when the two strains of cells are co-cultured, hPD-L1 on the surface of the Hep3B-OS8-hPDL1 cell interacts with PD-1 protein on the surface of a CD3+ T cell, so that the activation and proliferation of the CD3+ T cell and the expression of an immune factor INF-gamma are inhibited. When the compound blocks PD-1/PD-Upon L1 interaction, inhibition of CD3+ T cells is released, thereby promoting expression of INF-gamma.
The experimental process comprises the following steps: and (3) using an EDTA anticoagulation tube to contain whole blood, and separating PBMC by a density gradient centrifugation method. CD3+ T cells were further isolated from PBMC using the EasySepTM Human T Cell Isolation Kit and resuspended in RPMI-1640 complete medium at a concentration of 5X 10 5 The volume is/mL. Hep3B-OS8-hPDL1 cells treated with 10. Mu.g/mL mitomycin were incubated at 37 ℃ for 1.5h, washed 4 times with PBS, and cells were resuspended in RPMI-1640 complete medium at a concentration of 5X 10 5 The volume is/mL. Hep3B-OS8-hPDL1 (50. Mu.L/well) and T cells (100. Mu.L/well) were added to a 96-well round-bottom microplate. 4 XKeytruda (50. Mu.L/well), 4 Xtest compound (50. Mu.L/well) were prepared in RPMI-1640 complete medium and the prepared compounds and Keytruda were added to the corresponding wells (final concentration of Keytruda 5. Mu.g/mL) in a total volume of 200. Mu.L. 3 concentration gradients were set for each drug, double wells, keytruda and BMS-202 as positive controls. 37 ℃ C., 5% CO 2 Incubate for 72 hours in the incubator. After centrifugation at 350 Xg for 5 minutes, 150. Mu.L of the supernatant was collected and assayed for IFN-. Gamma.secretion by ELISA. And GrapdPad Prism6 for data processing.
The results are shown in FIG. 1, and the experimental results clearly show that the compound II-3 can relieve the CD3 by blocking the PD-1/PD-L1 interaction + T cell inhibition, promoting INF-gamma expression. The compounds II-1, II-3 and II-7 promote INF-gamma expression dose-dependently, and are significantly higher than BMS-202 and slightly lower than Keytruda (5. Mu.g/mL), thus having enhanced anti-tumor efficacy on T cells; therefore, the biphenyl compound can be used as an immune checkpoint PD-1/PD-L1 inhibitor to prepare a tumor immunotherapy drug.

Claims (10)

1. A compound of formula I or a pharmaceutically acceptable salt thereof:
Figure FDA0003179708720000011
wherein:
x, Y and Z each independently represent: n or CH;
a represents substituted phenyl or aromatic heterocyclic radical, the aromatic heterocyclic radical is a five-membered or six-membered aromatic ring containing 1-3O, N or S atoms, and the substituent is H, F, cl, br, CN, NH 2 、OH、CF 3 、OCF 3 、C 1 ~C 4 Alkyl or C of 1 -C 4 Alkoxy group of (a);
m =0, 1 or 2; n =0, 1 or 2;
R 1 and R 2 Each independently represents NR 6 R 7 、OR 7 Or substituted quaternary, quinary or hexahydric heterocycloalkyl containing 1 to 2O or N atoms; r is 6 Represents hydrogen or C 1 ~C 3 Alkyl groups of (a); r 7 Represents substituted C 1 ~C 6 The substituent is OH and NH 2 、COOH、CONH 2 、COOCH 3 、COOCH 2 CH 3 、C 1 ~C 4 The alkoxy group of (b) may be mono-or poly-substituted;
the substituted quaternary, quinary or hexahydric heterocycloalkyl containing 1-2O or N atoms is as follows: substituted tetrahydropyrrole-1-yl, substituted piperidine-1-yl, substituted morpholine-1-yl, substituted piperazine-1-yl or substituted azetidin-1-yl, wherein the substituents are OH, NH 2 、COOH、CONH 2 、COOCH 3 、COOCH 2 CH 3 、CF 3 、OCF 3 、C 1 ~C 4 Alkoxy group of (C) 1 ~C 4 The alkyl group of (b) may be mono-or poly-substituted;
R 3 and R 4 Each independently represents H, F, cl, br, CN, CF 3 、C 1 ~C 3 Alkyl or cyclopropyl of (a);
R 5 represents H, F, cl, br, CN, CF 3 、OCH 3 、OCH 2 CH 3 、OCF 3 、C 1 ~C 4 Alkyl or cyclopropyl.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, having the following general structural formula II when m =1 and a represents a 1, 4-disubstituted 1,2, 3-triazazole ring:
Figure FDA0003179708720000012
wherein: x, Y, Z, R 1 、R 2 、R 3 、R 4 、R 5 And n is as defined in claim 1.
3. A compound according to claim 2, wherein N represents 0 or 1, x represents N or CH, and Y and Z represent CH, or a pharmaceutically acceptable salt thereof.
4. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein R is 1 And R 2 Each represents: OH, OH,
Figure FDA0003179708720000021
Wherein R is 8 Represents CH 3 、CH 2 CH 3 、CH 2 CH 2 OH, formyl, acetyl, cyclopropyl, and the like; r 9 And R 10 Each independently represents H, OH, COOH, CH 2 COOH、CH 2 NH 2 、CH 2 OH、CH 2 CH 2 OH、F、Cl、Br、CH 3 、CH 2 CH 3 Cyclopropyl and the like; r 11 Represents OH, NH 2 、NHCH 3 、NHCH 2 CH 3 、CH 3 、OCH 3 、OCH 2 CH 3 ;R 12 Represents CONH 2 、NHCOCH 3 、OH、CH 2 OH、CH 2 CH 2 OH、COOH、CH 2 COOH, etc.; r 13 Represents H, CH 3 、CH 2 CH 3 、CH 2 OH、CH 2 CH 2 OH and the like; r is 14 And R 15 Each independently represents H, COOH, NH 2 、F、Cl、Br、CH 3 、CH 2 CH 3 、CH 2 OH、CH 2 CH 2 OH、CONH 2 Cyclopropyl and the like; w represents CH 2 、O、NH、N-CH 3 、N-CH 2 CH 3 、N-CH 2 CH 2 OH、N-COCH 3 Etc.; p represents 0 or 1.
5. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein R is 3 And R 4 Each independently of the other represents F, cl or CH 3
6. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein R is 5 Represents H, F, CH 3 Or OCH 3
7. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein n is preferably 0 or 1; x represents N or CH; y and Z represent CH; r is 1 And R 2 Each represents
Figure FDA0003179708720000022
Figure FDA0003179708720000023
R 3 And R 4 Each represents CH 3 ,R 5 Represents OCH 3
8. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the pharmaceutically acceptable salt is an acid addition salt of the compound of formula I with: 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, benzenesulfonic acid, p-toluenesulfonic acid or ferulic acid.
9. A pharmaceutical composition comprising a compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
10. Use of the compound or pharmaceutically acceptable salt thereof according to any one of claims 1 to 8 and the pharmaceutical composition according to claim 9 in preparation of antitumor drugs such as PD-1/PD-L1 inhibitors.
CN202110843500.1A 2021-07-26 Biphenyl-containing derivative and medical application thereof Active CN115677665B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110843500.1A CN115677665B (en) 2021-07-26 Biphenyl-containing derivative and medical application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110843500.1A CN115677665B (en) 2021-07-26 Biphenyl-containing derivative and medical application thereof

Publications (2)

Publication Number Publication Date
CN115677665A true CN115677665A (en) 2023-02-03
CN115677665B CN115677665B (en) 2024-04-19

Family

ID=

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018044963A1 (en) * 2016-09-01 2018-03-08 Bristol-Myers Squibb Company Biaryl compounds useful as immunomodulators
CN110799509A (en) * 2017-04-20 2020-02-14 吉利德科学公司 PD-1/PD-L1 inhibitors
CN111712494A (en) * 2018-02-13 2020-09-25 吉利德科学公司 PD-1/PD-L1 inhibitors
CN112041311A (en) * 2018-04-19 2020-12-04 吉利德科学公司 PD-1/PD-L1 inhibitors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018044963A1 (en) * 2016-09-01 2018-03-08 Bristol-Myers Squibb Company Biaryl compounds useful as immunomodulators
CN110799509A (en) * 2017-04-20 2020-02-14 吉利德科学公司 PD-1/PD-L1 inhibitors
CN111712494A (en) * 2018-02-13 2020-09-25 吉利德科学公司 PD-1/PD-L1 inhibitors
CN112041311A (en) * 2018-04-19 2020-12-04 吉利德科学公司 PD-1/PD-L1 inhibitors

Similar Documents

Publication Publication Date Title
US10662187B2 (en) Bruton's tyrosine kinase inhibitors
TWI787018B (en) Inhibitors of ret
RU2734760C2 (en) Anti-infectious compounds
JP7219902B2 (en) Bruton's Tyrosine Kinase Inhibitor
CA3133753A1 (en) Novel small molecule inhibitors of tead transcription factors
TW202112761A (en) SHP2 Phosphatase Allosteric Inhibitor
AU2013342203A1 (en) Therapeutic compounds and compositions and their use as PKM2 modulators
CN115716827A (en) Iminosulfones compound as bromodomain protein inhibitor, pharmaceutical composition and medical application thereof
WO2019141131A1 (en) Bromodomain inhibitor compound and use thereof
JP2022517280A (en) Bruton's tyrosine kinase inhibitor
CN110818609A (en) Preparation method and application of 3-acetyl indole BRPF1 inhibitor
CN115677665B (en) Biphenyl-containing derivative and medical application thereof
EP4332099A1 (en) Novel biphenyl derivative, preparation method therefor and medicinal use thereof
CN115677665A (en) Biphenyl-containing derivative and medical application thereof
AU2004303687A1 (en) Novel compounds
CN113698390B (en) Compounds useful as RET kinase inhibitors and uses thereof
JP4231405B2 (en) Bis (2-aryl-5-pyridyl) derivatives
CA3216045A1 (en) Compounds as pd1/pd-l1 inhibitors and methods thereof
CN115996727A (en) Process for preparing Rho related protein kinase inhibitors and intermediates in the process
US10793566B2 (en) Bruton's tyrosine kinase inhibitors
CN117551078A (en) Aromatic monocyclic derivative and preparation method and application thereof
CN116693514A (en) Aromatic ring substituted methoxy derivative and application thereof
JP2024516194A (en) Compounds as PD1/PD-L1 inhibitors and methods thereof
CN114437037A (en) Compound containing pyridine-bis-triazole as well as preparation method and application thereof
CN114573562A (en) Compound containing nicotinic acid and triazole as well as 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