CN106083836B - H based on quinazoline structure2S donor compound and application thereof - Google Patents

H based on quinazoline structure2S donor compound and application thereof Download PDF

Info

Publication number
CN106083836B
CN106083836B CN201610477893.8A CN201610477893A CN106083836B CN 106083836 B CN106083836 B CN 106083836B CN 201610477893 A CN201610477893 A CN 201610477893A CN 106083836 B CN106083836 B CN 106083836B
Authority
CN
China
Prior art keywords
reaction
compound
acid
qhs
quinazoline
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201610477893.8A
Other languages
Chinese (zh)
Other versions
CN106083836A (en
Inventor
郑友广
高彩云
苏军
刘毅
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.)
Xuzhou Medical College
Original Assignee
Xuzhou Medical College
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 Xuzhou Medical College filed Critical Xuzhou Medical College
Priority to CN201610477893.8A priority Critical patent/CN106083836B/en
Publication of CN106083836A publication Critical patent/CN106083836A/en
Application granted granted Critical
Publication of CN106083836B publication Critical patent/CN106083836B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a quinazoline structure-based H2An S donor compound and application thereof, which is a compound shown in a formula (I) or pharmaceutically acceptable salt thereof, wherein Ar is substituted or unsubstituted phenyl, and substituent groups of Ar are selected from: halogen, nitro, C1‑4Alkyl radical, C1‑4Haloalkyl, C1‑4Alkoxy radical, C1‑4One or more of halogenated alkoxy; x is C1‑4Alkoxy, B-NH-or A-CH2A CO-NH-group, Y being H, B-CnH2nO-or A-CnH2nO-group, A or B independently of one another being H2An S donor group, n is an integer of 1 to 5, and X is C1‑3Y is not H in the case of alkoxy. The invention relates to a series of H based on 4-anilino quinazoline structure2S donor compound by H2The S and the 4-anilino quinazoline derivative have synergistic effect, so that the antitumor activity of the medicament is improved.

Description

H based on quinazoline structure2S donor compound and application thereof
Technical Field
The invention belongs to the field of pharmaceutical compounds, and particularly relates to a quinazoline structure-based H2S donor compound and its preparation method and application.
Background
As is well known, hydrogen sulfide (H)2S) is a gas with the smell of rotten eggs, H, for centuries2S is considered to be a highly toxic air pollutant and inhalation of large quantities can lead to poisoning. However, recently such gas molecules have been considered as a further important gas transmitter following nitric oxide and carbon monoxide. Endogenous H2S production, associated with at least three enzymes: cystathionine beta-synthase (CBS), cystathionine gamma-lyase (CSE) and 3-mercaptopyruvate thiotransferase (MPST). In different tissues or organs, these enzymes convert cysteine or cysteine derivatives into H2S, and H2S has a variety of biological effects including vasodilation, anti-inflammatory, anti-cancer and cardioprotection, among others.
It is reported that H2S results in the hydrolysis of the disulfide bonds of the protein molecules (i.e., -S-SH formation), and thus, H2S may alter the biological functions of various proteins and enzymes. At the same time, H2S also reacts with S-nitrosothiols to produce mercaptonitrous acid (HSNO), and small amounts of S-nitrosothiols are free to pass through cell membranes, promoting nitrosation of proteins. The results of the above studies indicate that H can be regulated2Levels of S may have greater utility in the treatment of disease.
It has now been found that exogenous H2S induces tumor cell apoptosis by activating MAP kinase and caspase-3, thereby inhibiting tumor growth. In liver cancer, exogenous H2S blocks STAT3 signal channel, blocks cell cycle, induces apoptosis, and inhibits tumor growth. In human colon cancer, exogenous H2S inhibits the proliferation of tumors by inhibiting the NF-kB signaling pathway and the activity of Trx/TrxR. In breast cancer, exogenous H2S induces apoptosis in tumor cells by cleaving PARP and activating caspase-7. In gastric cancer, exogenous H2S inhibits tumor proliferation by regulating and controlling related apoptosis proteins Bax, CytC and caspase-3. It can be seen that exogenous H2S has an inhibiting effect on tumors, which provides a possible new way for treating tumors.
In the field of hydrogen sulfide, H2The S donor is an important research tool. Meanwhile, the results of the study confirmed that H2S has an inhibitory effect in many tumors. In these H2Among the S donors, the most commonly used are the sulfide salts, including sodium sulfide (Na)2S) and sodium hydrosulfide (NaHS). These inorganic salts have excellent water solubility, are nontoxic to cells at specific concentrations, and can rapidly increase H2The concentration of S. However, they spontaneously release H in aqueous solution2S, it is therefore difficult to precisely control H2The concentration of S. In addition, due to H2The concentration of S gas in the aqueous solution is rapidly reduced by volatilization of S gas, so that the use of sodium sulfide and sodium hydrosulfide is greatly limited.
In view of these disadvantages, synthesis H2The S donor has received considerable attention, and has been reported in the literature as H2An S donor compound. E.g. H2The S donor, GYY4137, is a derivative of Lawesson' S reagent. GYY4137 has been shown to release H both in vitro and in vivo2S, and can pass through slowly released H2S, the STAT3 signal channel is blocked, so that the inhibition effect on the liver cancer cells is generated. Other H reported so far2The S donor mainly comprises DATS, Z-ajoene, S-propylcysteine sulfoxide, ADT-OH, ACS-81, ACS-60 and the like. Above H2The S donor can be combined with a parent compound with biological activity to obtain H with better activity2S donor compounds such as HS-sublindac, HS-ibuprofen, HS-naproxen, HS-aspirin and HS-ASA, etc. After the compound enters the body, H is slowly released under the action of in-vivo enzyme2S and the parent compound or the structural analogue thereof exert the synergistic effect of the S and the parent compound, thereby showing better treatment effect or lower toxic and side effect than the parent compound. Researches find that the compounds have inhibiting effect on lung cancer, colon cancer, breast cancer and pancreatic cancer.
Figure BDA0001030122650000021
Quinazolines are the most studied structures in medicinal chemistry. The quinazoline alkaloid has potential antimalarial, antitumor, antibacterial, anti-inflammatory, antihypertensive and other activities. Over the last 15 years, the FDA has approved several anti-cancer drugs of 4-anilinoquinazoline derivatives, such as gefitinib, erlotinib, and lapatinib, among others. These 4-anilinoquinazoline compounds are all capable of inhibiting the activity of EGFR kinase. In addition, some kinase inhibitors using 4-anilinoquinazoline as a parent nucleus have been increasingly studied, and some of them have entered clinical research.
The Epidermal Growth Factor Receptor (EGFR) is a member of receptor tyrosine kinase families, mainly comprises subtypes of HER1 (namely EGFR), HER2, HER3, HER4 and the like, the high expression of the EGFR is closely related to epithelial cell tumors, the high expression tumor cells have strong invasiveness, are easy to transfer and have poor curative effect, and patients are poor after being cured.
Gefitinib (Gefitinib) has a 4-anilinoquinazoline structure, is a small molecule inhibitor targeting EGFR developed by Astra ZenecaAnd (3) preparing. Non-small cell lung cancer, which is not operable or relapsed, was first marketed in japan in 2002, and was approved as a three-line monotherapy for advanced non-small cell lung cancer (NSCLC) in the united states and australia in 2003. Experimental study shows that the compound can inhibit IC of EGFR in vitro500.037. mu.M; erlotinib (Erlotinib) a 4-anilinoquinazoline class of EGFR small molecule inhibitors developed by OSI. US FDA approval to market for locally advanced or metastatic non-small cell lung cancer (NSCLC) at 11 months 2004, which inhibits the IC of EGFR500.002. mu.M; lapatinib (Lapatinib), developed by the company glatiramer as an EGFR/HER2 dual inhibitor, is a 4-anilinoquinazoline compound that was approved by the FDA in the united states for sale in 2007 for the treatment of advanced or metastatic breast cancer. Research shows that in some cancer cell lines (A549, PC3, MCF7 and the like), lapatinib shows high antiproliferative activity, and IC of the lapatinib inhibits EGFR50It was 0.010. mu.M. In addition, some Lapatinib analogues have inhibition effect on EGFR and HER-2 in preclinical research stage, such as compound 1, and IC thereof50Values were less than 0.05 and 0.03nM, respectively.
Figure BDA0001030122650000031
Aurora kinase is an important serine/threonine kinase that plays an important regulatory role in cell mitosis. Aurora kinase expression is abnormal, interferes with mitosis, and may lead to gene instability and tumor growth.
AZD1152 is a 4-anilinoquinazoline Aurora B selective inhibitor developed by Astra Zeneca, Inc. that inhibits the IC of Aurora B500.37nM, and inhibition of Aurora A IC501368 nM. The study shows that AZD1152 can induce cycle arrest and apoptosis of human acute lymphocyte and leukemia cell, and the clinical phase II evaluation of the AZD for treating blood tumor, malignant solid tumor and advanced solid tumor. ZM447439 is the first reported Aurora A and Aurora B inhibitor, and is the first 4-anilinoquinazoline with Aurora A/B kinase inhibitory activity screened from 250000 compounds by Astra ZenecaLeads, IC's of Aurora A and B inhibition50110 and 130nM, respectively. Since 2011, quinazoline Aurora kinase inhibitors have been reported in a number of patents. Among them, a series of quinazoline derivatives are designed and synthesized in a patent (WO2011144059), and show a dual-inhibition effect on Aurora B and EGFR, for example, when the compound 3 is at 200 μ M, the inhibition rates on Aurora B and EGFR reach 96% and 98%. The compound 4 designed and synthesized in the Chinese patent CN10409855 has high homology with AZD1152 structure, has better Aurora kinase inhibitory activity and inhibits the IC of Aurora A50IC for inhibition of Aurora B at 82nM50It was 0.15 nM.
Figure BDA0001030122650000041
European patent EP2708532 describes a series of derivatives related to 4-anilinoquinazoline, which primarily inhibit HDAC-1 enzymes. Wherein IC of Compound 550The value was 178 nM. The US20120094997 patent found that when a hydroxamic acid substituent is present at the C-6 position of the quinazoline, the selectivity for HDAC-6 enzyme is higher than that for HDAC-1 enzyme.
Figure BDA0001030122650000042
In conclusion, the 4-anilinoquinazoline derivatives as EGFR inhibitors, Aurora kinase inhibitors, histone deacetylase inhibitors and the like show good antitumor activity.
In recent years, the incidence of tumors has increased year by year, and the research and development of novel safe, efficient and low-toxicity antitumor agents is imminent.
Disclosure of Invention
The invention aims to design and synthesize a series of H based on 4-anilino quinazoline structure based on the prior art2S donor compound by H2The S and the 4-anilino quinazoline derivative have synergistic effect, so that the antitumor activity of the medicament is improved.
Another object of the present invention is to provideFor one kind of the above-mentioned H based on quinazoline structure2A method for preparing an S donor compound.
The third purpose of the invention is to provide the H based on the quinazoline structure2The S donor compound is applied to the anti-tumor aspect.
The object of the invention can be achieved by the following measures:
quinazoline structure-based H2An S donor compound which is a compound represented by the formula (I) or a pharmaceutically acceptable salt thereof,
Figure BDA0001030122650000051
wherein the content of the first and second substances,
ar is substituted or unsubstituted phenyl, and the substituents are selected from: halogen, nitro, C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy radical, C1-4One or more of halogenated alkoxy;
x is C1-4Alkoxy, B-NH-or A-CH2A group of CO-NH-or a group of NH-,
y is H, B-CnH2nO-or A-CnH2nA group of O-is selected from the group consisting of,
a or B is independently H2An S donor group, a N-substituted phenyl group,
n is an integer of 1 to 5,
and X is C1-3Y is not H in the case of alkoxy.
In a preferred embodiment, Ar is a substituted or unsubstituted phenyl group, wherein the substituents are selected from the group consisting of: fluorine, chlorine, bromine, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C1-3One or more of alkoxy.
In a more preferred embodiment, Ar is a substituted or unsubstituted phenyl group, the substituents of which are selected from the group consisting of: one or more of fluorine, chlorine, bromine, nitro, methyl, ethyl, methoxy, ethoxy and trifluoromethyl.
In the invention, A or B is respectively H2S donor group, in a preferred embodiment, A is
Figure BDA0001030122650000052
B is
Figure BDA0001030122650000053
N in the present invention may be 1,2, 3,4 or 5, preferably 2, 3 or 4, most preferably 3.
In a preferred embodiment, X is C1-3Alkoxy, Y is B-CnH2nO-or A-CnH2nAn O-group.
In another preferred embodiment, X is B-NH-or A-CH2A CO-NH-group and Y is H.
In one embodiment, the compound of the present invention may be selected from compounds represented by formula (II) or formula (III),
Figure BDA0001030122650000054
wherein the content of the first and second substances,
R3is a group A or a group B,
R4is A-CH2CO-or B groups.
The invention relates to a compound or a pharmaceutically acceptable salt thereof, wherein the compound can be selected from the following compounds:
Figure BDA0001030122650000061
the invention provides a synthetic route of a compound shown in a formula (II), which comprises the following steps:
Figure BDA0001030122650000062
the invention provides a synthetic route of a compound shown in a formula (III), which comprises the following steps:
Figure BDA0001030122650000071
the invention provides a pharmaceutical composition, which takes the compound or the pharmaceutically acceptable salt thereof as an active ingredient or a main active ingredient, and is assisted by pharmaceutically acceptable auxiliary materials.
Unless otherwise indicated, the following terms used in the specification and claims have the meanings discussed below:
"alkyl" in the present invention means a saturated aliphatic group of 1 to 20 carbon atoms, including straight and branched chain groups (the numerical range mentioned in this application, e.g. "1 to 20" means that the group, in this case alkyl, may contain 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms). Alkyl group having 1 to 4 carbon atoms (C)1-4Alkyl) is referred to as lower alkyl. When a lower alkyl group has no substituent, it is referred to as unsubstituted lower alkyl. More preferably, the alkyl group is a medium size alkyl group having 1 to 10 carbon atoms, such as methyl, ethyl, propyl, 2-propyl, n-butyl, isobutyl, tert-butyl, pentyl, and the like. Preferably, the alkyl group is a lower alkyl group having 1 to 4 carbon atoms, such as methyl, ethyl, propyl, 2-propyl, n-butyl, isobutyl, tert-butyl, or the like. Alkyl groups may be substituted or unsubstituted.
"halogen" in the present invention means fluorine, chlorine, bromine or iodine, preferably fluorine or chlorine.
The "nitro group" in the present invention means "-NO2"group
"haloalkyl" in the context of the present invention denotes halogen-substituted alkyl, preferably halogen-substituted lower alkyl as defined above, which is substituted by one or more halogen atoms which may be the same or different (e.g. C)1-4Haloalkyl), e.g. -CH2Cl、-CF3、-CH2CF3、-CH2CCl3And the like.
"alkoxy" in the present invention means-O- (unsubstituted alkyl) and-O- (unsubstituted cycloalkyl). Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy, and the like.
"haloalkoxy" in the present invention means an alkoxy group substituted with one or more of the same or different halogen atoms. Representative examples include, but are not limited to, trifluoromethoxy, chloromethoxy, and the like.
By "pharmaceutically acceptable salts" in the context of this invention is meant those salts which retain the biological effectiveness and properties of the parent compound. Such salts include:
(1) salts with acids are obtained by reaction of the free base of the parent compound with inorganic acids including hydrochloric acid, hydrobromic acid, nitric acid, phosphoric acid, metaphosphoric acid, sulfuric acid, sulfurous acid, perchloric acid and the like, or with organic acids including acetic acid, trifluoroacetic acid, propionic acid, acrylic acid, caproic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, oxalic acid, (D) or (L) malic acid, fumaric acid, maleic acid, benzoic acid, hydroxybenzoic acid, γ -hydroxybutyric acid, methoxybenzoic acid, phthalic acid, methanesulfonic acid, ethanesulfonic acid, naphthalene-1-sulfonic acid, naphthalene-2-sulfonic acid, p-toluenesulfonic acid, salicylic acid, tartaric acid, citric acid, lactic acid, cinnamic acid, dodecylsulfuric acid, gluconic acid, glutamic acid, aspartic acid, stearic acid, mandelic acid, succinic acid or malonic acid and the like.
(2) The acidic proton present in the parent compound is replaced by a metal ion such as an alkali metal ion, an alkaline earth metal ion or an aluminum ion, or is complexed with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, quinine, or the like.
"pharmaceutical composition" in the present invention refers to one or more of the compounds of the present invention, or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof, in admixture with another chemical ingredient, such as a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to facilitate the administration process to an animal.
The compound or the pharmaceutically acceptable salt thereof can be applied to the preparation of anti-cancer drugs, in particular to the anti-liver cancer or liver cancer drugs.
The invention relates to a series of H based on 4-anilino quinazoline structure2S donor compound by H2The S and the 4-anilino quinazoline derivative have synergistic effect, so that the antitumor activity of the medicament is improved. Researching the in vitro anti-HepG 2 and A-549 cell proliferation activity of the compound; on the basis of the detection, the in-vitro release H of the compound is detected2S level; the molecular level EGFR inhibitory activity of the target compound is detected so as to discover a lead compound with higher comprehensive evaluation and provide a new way for the research and development of novel antitumor drugs.
Drawings
FIG. 1 shows the QHS-6 qualitative release H of the compound of the present invention2S level.
In the figure, the left panel A is: the fluorescent probe NIR-HS (10 μ M) was incubated with compound QHS-6(0, 5, 10,50,100,200 μ M) in PBS buffer (PBS: acetone ═ 2:3) for 3h (37 ℃) and the fluorescence intensity was measured. The right panel B is: the fluorescence intensity of compound QHS-6(0, 5, 10,50,100, 200. mu.M) was measured at 723 nm.
FIG. 2 shows the QHS-13 qualitative release H of the compound of the present invention2S level.
In the figure, the left panel A is: the fluorescent probe NIR-HS (10 μ M) was incubated with compound QHS-13(0, 5, 10,50,100,200 μ M) in PBS buffer (PBS: acetone ═ 2:3) for 3h (37 ℃) and the fluorescence intensity was measured. The right panel B is: the fluorescence intensity of compound QHS-13(0, 5, 10,50,100, 200. mu.M) was measured at 723 nm.
FIG. 3 shows qualitative release of H from LHS-1 of the compound of the invention2S level.
In the figure, the left panel A is: the fluorescent probe NIR-HS (10 μ M) was incubated with compound LHS-1(0, 5, 10,50,100,200 μ M) in PBS buffer (PBS: acetone ═ 2:3) for 3h (37 ℃) and the fluorescence intensity was measured. The right panel B is: the fluorescence intensity of compound LHS-1(0, 5, 10,50,100, 200. mu.M) was measured at 723 nm.
FIG. 4 is Na2S standard curve.
In the figure, the left panel A is: fluorescent probes NIR-HS (10. mu.M) and Na2S (0,2,4,6, 8. mu.M) was incubated in PBS buffer (PBS: acetone ═ 2:3) for 3h (37 ℃ C.), and the fluorescence intensity was measured. The right panel B is: na (Na)2S standard curve.
FIG. 5 is Compound QHS-6, QHS-13 and LHS-1 release H quantitatively2S level.
Detailed Description
Some aspects and embodiments of the invention are further illustrated by the following specific examples. The compounds encompassed by the present invention can be synthesized by known conventional techniques. These compounds can be conveniently synthesized from readily available starting materials. The following is a general synthetic scheme for the compounds synthesized in the present invention. The schemes disclosed herein are illustrative and are not meant to limit the ability of those skilled in the art to synthesize compounds using other possible methods. Various methods are conventional in the art. In addition, different synthetic steps may be applied to synthesize the target compound in different schemes. All documents cited herein are incorporated by reference herein.
The following representative examples are intended to aid in the illustration of the present invention and are not intended to, and should not be construed to, limit the scope of the present invention. Indeed, the entire contents of the documents in this invention, including examples in accordance with the scientific literature and patents cited herein, as well as various modifications and numerous further variations thereof, will be readily apparent to those skilled in the art, except to those shown and described herein. It should also be understood that the citation of these references is helpful in setting forth the disclosure herein. The following examples contain important supplementary information, examples and guidance, and are adaptable to various variations and the like in the present invention.
Synthetic examples
The compounds of the present invention are primarily directed to two classes: QHS series and LHS series. Wherein, the QHS series comprise ether bond QHS-a (QHS-1, QHS-2, QHS-3, QHS-4, QHS-5 and QHS-6) and ester bond QHS-b (QHS-13 and QHS-15), and the LHS series comprise LHS-a (FHS-1, LHS-1 and XHS-1) and LHS-b (FHS-2, LHS-2 and XHS-2). Specific compounds are shown in tables 1 and 2.
TABLE 1 QHS series of compounds
Figure BDA0001030122650000091
Figure BDA0001030122650000092
TABLE 2 LHS series of compounds
Figure BDA0001030122650000101
Figure BDA0001030122650000102
Overall synthetic route and steps of QHS series compounds
4-hydroxy-3-methoxybenzoic acid is used as a raw material, and a series of reactions such as esterification, nitration, reduction, ring closure, chlorination and the like are carried out to synthesize an intermediate (7a,7b,7c,7d,7e,7f), and then H is connected2And 8 QHS series compounds are finally obtained by an S donor, and the specific synthetic route is as follows.
Figure BDA0001030122650000103
Synthesis of methyl 4-hydroxy-3-methoxybenzoate (1)
Figure BDA0001030122650000111
To a 500mL three-necked flask were added 4-hydroxy-3-methoxybenzoic acid (20.0g, 0.19mol), 250mL of methanol, and 0.8mL of concentrated sulfuric acid, and the mixture was heated to reflux for about 48 hours, after which the reaction was stopped. The methanol is distilled off, water is added to the residue, saturated K2CO3Neutralizing the solution, extracting with ethyl acetate, drying the organic layer with anhydrous sodium sulfate, filtering, concentrating the mother liquor under reduced pressure to obtain brown oily substance, adding petroleum ether: ethyl acetate (8:1) was recrystallized to give (1) as a white solid (19.6g, 90.5%) Mp:62-63 ℃.
Synthesis of methyl 3-methoxy-4- (3-chloropropyloxy) benzoate (2)
Figure BDA0001030122650000112
4.22g (23mmol) of methyl 4-hydroxy-3-methoxybenzoate (1), 5.06g (32.4mmol) of 1-bromo-3-chloropropane are dissolved in 20ml of DMF, 10.24g (74.2mmol) of potassium carbonate are added, and the reaction is carried out at 100 ℃ for 12 hours. After the reaction solution was cooled to room temperature, the reaction solution was poured into 500mL of ice water and vigorously stirred for 30 min. Suction filtration gave 5.85g of off-white solid. Recrystallization from ethyl acetate gave 4.85g of the white target product in 80.7% yield, Mp: 103-.
Synthesis of methyl 2-nitro-5-methoxy-4- (3-chloropropyloxy) benzoate (3)
Figure BDA0001030122650000113
9.3g (12.8mmol) of methyl 3-methoxy-4- (3-chloropropyloxy) benzoate (2) are dissolved in 30mL of acetic acid and 3mL of acetic anhydride at 0 to 5 ℃, concentrated nitric acid (8.45mL, 66%) is added dropwise, and the mixture is stirred at room temperature for 6 hours. The reaction solution was poured into 500mL of ice water, and extracted with ethyl acetate. The organic layers were combined, washed with a saturated aqueous sodium bicarbonate solution and brine in this order, and dried over anhydrous sodium sulfate. The solvent was evaporated to dryness to give a yellow oil (13.0 g). Recrystallization from ethyl acetate/petroleum ether gave 8.13g of a bright yellow solid in 75% yield, Mp 57-59 ℃. Synthesis of methyl 2-amino-5-methoxy-4- (3-chloropropyloxy) benzoate (4)
Figure BDA0001030122650000114
After 5.0g (89.3mmol) of reduced iron powder was added to 40mL of acetic acid and stirred at 50 ℃ for 15min under nitrogen protection, 9.0g (25mmol) of methyl 2-nitro-5-methoxy-4- (3-chloropropyloxy) benzoate (3) in methanol (30mL) was slowly added, and the reaction mixture was reacted at 50-60 ℃ for 30 min. The iron mud in the reaction solution is filtered while the solution is hot, the filtrate is decompressed and distilled to remove the solvent until no distillate exists, the residue is poured into ice water, and ethyl acetate is used for extraction. The organic layer was washed with saturated potassium carbonate and brine in this order, and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give a brown solid, which was recrystallized from ethyl acetate/petroleum ether to give 6.34g of a bright brown solid in 77% yield, Mp:67-69 ℃.
Synthesis of 6-methoxy-7- (3-chloropropoxy) quinazolin-4 (3H) -one (5)
Figure BDA0001030122650000121
1.7g (6mmol) of methyl 2-amino-5-methoxy-4- (3-chloropropyloxy) benzoate (4) are dissolved in 20mL of absolute ethanol, 0.97g (9mmol) of formamidine acetate are added and the mixture is refluxed for 24 h. Cooling, standing in a refrigerator, filtering, washing with cold ethanol to obtain 1.52g of white powdery solid, with the yield of 94.4 percent and the Mp of 113 and 115 ℃.
Synthesis of 4-chloro-6-methoxy-7- (3-chloropropoxy) quinazoline (6)
Figure BDA0001030122650000122
1.0g (3.15mmol) of 6-methoxy-7- (3-chloropropoxy) quinazolin-4 (3H) -one (5) are added in portions to 8mL of thionyl chloride, and then 0.62mL of DMMF is slowly added dropwise. The reaction was refluxed for 4 h. The excess thionyl chloride was evaporated under reduced pressure to give a yellow solid which was dissolved in chloroform, washed with a saturated sodium carbonate solution and water in this order, and dried over anhydrous sodium sulfate. The organic solvent was evaporated under reduced pressure to obtain a white solid, which was recrystallized from ethyl acetate to obtain 0.91g of a white solid with a yield of 86%, Mp: 146-.
Synthesis of 4- (3-chloro-4-fluoroanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7a)
Figure BDA0001030122650000123
2.0g (7mmol) of 4-chloro-6-methoxy-7- (3-chloropropoxy) quinazoline (6) and 2.1g (13.7mmol) of 3-chloro-4-fluoroaniline were added to 10mL of isopropanol and reacted for 3 hours under reflux. The reaction was cooled to room temperature, cooled in a refrigerator overnight, filtered with suction, and the filter cake was washed with cold isopropanol. Vacuum drying at 60-70 deg.C to obtain yellow solid 2.73g, yield 93%, Mp: 170-.
Synthesis of 4- (4-bromoanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7b)
Figure BDA0001030122650000124
2.0g (7mmol) of 4-chloro-6-methoxy-7- (3-chloropropoxy) quinazoline (6) and 2.36g (13.7mmol) of 4-bromoaniline are added to 10mL of isopropanol and reacted under reflux for 3 h. The reaction was cooled to room temperature, cooled in a refrigerator overnight, filtered with suction, and the filter cake was washed with cold isopropanol. Vacuum drying at 60-70 deg.c to obtain yellow solid in 1.42g yield of 48% Mp 165-167 deg.c.
Synthesis of 4- (4-nitroanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7c)
Figure BDA0001030122650000131
2.0g (7mmol) of 4-chloro-6-methoxy-7- (3-chloropropoxy) quinazoline (6) and 1.91g (13.7mmol) of 4-nitroaniline were added to 10mL of isopropanol and reacted under reflux for 3 h. The reaction was cooled to room temperature, cooled in a refrigerator overnight, filtered with suction, and the filter cake was washed with cold isopropanol. Vacuum drying at 60-70 deg.C to obtain yellow solid 1.41g, yield 53%, Mp: 168-.
Synthesis of 4- (3-bromoanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7d)
Figure BDA0001030122650000132
2.0g (7mmol) of 4-chloro-6-methoxy-7- (3-chloropropoxy) quinazoline (6) and 2.36g (13.7mmol) of 3-bromoaniline are added to 10mL of isopropanol and reacted under reflux for 3 h. The reaction was cooled to room temperature, cooled in a refrigerator overnight, filtered with suction, and the filter cake was washed with cold isopropanol. Vacuum drying at 60-70 deg.c to obtain yellow solid in 0.92g yield of 31% and Mp: 157-.
Synthesis of 4- (3-trifluoromethylanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7e)
Figure BDA0001030122650000133
2.0g (7mmol) of 4-chloro-6-methoxy-7- (3-chloropropoxy) quinazoline (6) and 2.21g (13.7mmol) of 3-trifluoromethylaniline were added to 10mL of isopropanol and reacted under reflux for 3 hours. The reaction was cooled to room temperature, cooled in a refrigerator overnight, filtered with suction, and the filter cake was washed with cold isopropanol. Vacuum drying at 60-70 deg.C to obtain yellow solid 1.24g, yield 43%, Mp: 167-.
Synthesis of 4- (3, 4-dimethoxyanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7f)
Figure BDA0001030122650000134
2.0g (7mmol) of 4-chloro-6-methoxy-7- (3-chloropropoxy) quinazoline (6) and 2.0g (13.7mmol) of 3, 4-dimethoxyaniline are added to 10mL of isopropanol and reacted for 3h under reflux. The reaction was cooled to room temperature, cooled in a refrigerator overnight, filtered with suction, and the filter cake was washed with cold isopropanol. Vacuum drying at 60-70 deg.C to obtain yellow solid 1.41g, yield 50%, Mp 173-.
Synthesis of 5-p-hydroxyphenyl-3H-1, 2-dithiole-3-thione (ADT-OH)
Figure BDA0001030122650000141
To a single-neck flask was added 5-p-methoxyphenyl-3H-1, 2-dithiole-3-thione (0.24g, 1mmol) and pyridine hydrochloride
(1.20g, 10mmol) was mixed well. Carrying out melt reaction for 0.5h at 210 ℃, monitoring the reaction by TLC, cooling to room temperature after the reaction is finished, adding 1mol/L diluted hydrochloric acid, carrying out suction filtration to obtain a tan solid, and carrying out vacuum drying. The yield was 81%, Mp: 185-.
Synthesis of 3-allyldithiopropionic acid (ACS-81)
Figure BDA0001030122650000142
To a three-necked flask was added diallyl disulfide (5.85g, 40mmol), 3-mercaptopropionic acid (0.85g, 8mmol), methanol: diethyl ether (2: 1V/V) mixed solvent, then 10mol/L NaOH (0.24g, 6mmol) is added, nitrogen is introduced for 30min, and reaction is carried out for 24h at 25 ℃. The reaction was monitored by TLC, after completion of the reaction, the reaction solution was evaporated to dryness, 1mol/L HCl was added, extraction was performed with diethyl ether, and the solvent was evaporated under reduced pressure to give 1.2g of an off-white oil. The yield thereof was found to be 84%.
Synthesis example 1: synthesis of QHS-1
Figure BDA0001030122650000143
Adding 4- (3-chloro-4-fluoroanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7a) (0.395g, 1mmol), anhydrous DMF10mL, potassium carbonate (0.276g, 2mmol), and catalytic amount of potassium iodide into a single-neck flask, stirring for 10min, adding ADT-OH (0.226g, 1mmol), reacting at 80 ℃, monitoring the reaction by TLC, cooling to room temperature after the reaction is finished, pouring the reaction solution into ice water, stirring for 30min, performing suction filtration, performing vacuum drying, and separating dichloromethane through silica gel column chromatography: methanol (120:1) gave 0.12g of an orange solid in 20% yield, Mp: 104-.
1H NMR(400MHz,CDCl3)δ8.66(s,1H),7.91(s,1H),7.64-7.59(m,2H),7.31(d,J=16.0Hz,5H), 7.22-7.18(m,1H),7.03(d,J=8.0Hz,2H),4.36(d,J=36.0Hz,4H),4.04(s,3H),2.46(s,2H).
13C NMR(101MHz,DMSO)δ215.24,174.23,172.49,162.39,156.54,154.05,152.95,152.48,149.58, 134.63,129.49,124.21,124.01,122.87,122.80,119.36,119.17,117.10,116.89,116.01,109.24,108.14, 102.40,65.59,65.27,56.78,28.70.
LC-MSm/z:585.8[M-H]-.
Synthesis example 2: synthesis of QHS-2
Figure BDA0001030122650000151
Adding 4- (4-bromoanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7b) (0.423g and 1mmol), anhydrous DMF10mL, potassium carbonate (0.276g and 2mmol), a catalytic amount of potassium iodide into a single-neck flask, stirring for 10min, adding ADT-OH (0.226g and 1mmol), reacting at 80 ℃, monitoring the reaction by TLC, cooling to room temperature after the reaction is finished, pouring the reaction liquid into ice water, stirring for 30min, performing suction filtration, performing vacuum drying, and separating dichloromethane by silica gel column chromatography: ethyl acetate (2:1) was added to 1mL of methanol to give 0.1g of an orange solid in 16% yield, Mp: 113-.
1H NMR(400MHz,CDCl3)δ8.66(s,1H),7.63(dd,J=8.0,12.0Hz,3H),7.53(d,J=8.0Hz,2H),7.39(s, 1H),7.29(d,J=12.0Hz,3H),7.07-7.01(m,3H),4.39-4.29(m,4H),4.03(s,3H),2.47-2.44(m,2H).
13C NMR(101MHz,CDCl3)δ195.70,172.99,169.69,167.96,162.15,154.11,149.86,134.67,132.03, 128.60,124.31,123.42,121.81,116.97,116.23,115.52,100.68,65.34,64.69,56.38,29.70.
LC-MSm/z:611.8[M-H]-.
Synthetic example 3: synthesis of QHS-3
Figure BDA0001030122650000152
Adding 4- (4-nitroanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7c) (0.389g, 1mmol), anhydrous DMF10mL, potassium carbonate (0.276g, 2mmol), a catalytic amount of potassium iodide into a single-neck flask, stirring for 10min, adding ADT-OH (0.226g, 1mmol), reacting at 80 ℃, monitoring the reaction by TLC, cooling to room temperature after the reaction is finished, pouring the reaction solution into ice water, stirring for 30min, performing suction filtration, drying in vacuum, and separating dichloromethane by silica gel column chromatography: ethyl acetate (1:2) was added to 1mL of methanol to give 0.11g of an orange solid in 16% yield, Mp: 108-.
1H NMR(400MHz,DMSO)δ10.05(s,1H),8.65(s,1H),8.30(d,J=8.0Hz,2H),8.20(d,J=12.0Hz,2H), 7.92-7.87(m,2H),7.81-7.76(m,1H),7.31(s,1H),7.13(dd,J=4.0Hz,2H),4.34(dd,J=4.0,20.0Hz,4H), 4.01(s,3H),2.33-2.30(m,2H).
13C NMR(101MHz,DMSO)δ215.26,174.23,162.39,162.00,156.16,154.47,152.59,149.92,142.12, 134.65,129.50,128.73,125.07,124.22,121.15,116.56,116.03,109.85,108.03,102.52,65.74,65.26,56.89, 28.71.
LC-MSm/z:579.8[M+H]+.
Synthetic example 4: synthesis of QHS-4
Figure BDA0001030122650000161
Adding 4- (3-bromoanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7d) (0.423g, 1mmol), anhydrous DMF10mL, potassium carbonate (0.276g, 2mmol), a catalytic amount of potassium iodide into a single-neck flask, stirring for 10min, adding ADT-OH (0.226g, 1mmol), reacting at 80 ℃, monitoring the reaction by TLC, cooling to room temperature after the reaction is finished, pouring the reaction liquid into ice water, stirring for 30min, performing suction filtration, performing vacuum drying, and separating dichloromethane through silica gel column chromatography: ethyl acetate (2:1) was added to 1mL of methanol to give 0.12g of an orange solid in 20% yield, Mp: 104-.
1H NMR(400MHz,DMSO)δ9.53(s,1H),8.53(s,1H),8.16(s,1H),7.90-7.75(m,5H),7.34(d,J=8.0Hz, 1H),7.27(d,J=16.0Hz,2H),7.12(d,J=8.0Hz,2H),4.30(d,J=20.0Hz,4H),3.98(s,3H),2.30(s,2H).
13C NMR(101MHz,DMSO)δ215.26,174.24,162.40,156.47,154.03,153.05,149.59,147.32,141.73, 134.64,130.83,129.50,128.73,126.10,124.49,124.22,121.65,121.06,116.03,115.84,109.44,108.34, 102.42,65.60,65.28,56.79,28.72.
LC-MSm/z:611.7[M-H]-.
Synthesis example 5: synthesis of QHS-5
Figure BDA0001030122650000162
Adding the synthesized (7e) (0.411g and 1mmol) of 4- (3-trifluoromethylanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline, anhydrous DMF10mL, potassium carbonate (0.276g and 2mmol) and a catalytic amount of potassium iodide into a single-neck flask, stirring for 10min, adding ADT-OH (0.226g and 1mmol), reacting at 80 ℃, monitoring the reaction by TLC, cooling to room temperature after the reaction is finished, pouring the reaction liquid into ice water, stirring for 30min, performing suction filtration, drying in vacuum, and separating dichloromethane through silica gel column chromatography: ethyl acetate (2:1) was added to 1mL of methanol to give 0.12g of an orange solid in a yield of 20%, Mp: 109-.
1H NMR(400MHz,DMSO)δ9.67(s,1H),8.54(s,1H),8.23(d,J=8.0Hz,2H),7.87(d,J=8.0Hz,3H), 7.75(s,1H),7.63(t,J=8.0Hz,1H),7.44(d,J=8.0Hz,1H),7.26(s,1H),7.13(d,J=12.0Hz,2H), 4.35-4.28(m,4H),4.00(d,J=5.0Hz,3H),2.32-2.29(m,2H).
13C NMR(101MHz,DMSO)δ215.24,174.22,170.79,162.40,156.51,154.07,153.01,149.62,147.42, 140.93,134.63,130.03,129.48,126.10,125.85,124.21,123.39,119.75,118.27,116.01,109.45,108.37, 102.40,65.58,65.28,56.79,28.72.
LC-MSm/z:602.8[M+H]+.
Synthetic example 6: synthesis of QHS-6
Figure BDA0001030122650000171
Adding synthesized (7f) (0.404g and 1mmol) of 4- (3, 4-dimethoxyanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline, anhydrous DMF10mL, potassium carbonate (0.276g and 2mmol) and catalytic amount of potassium iodide into a single-neck flask, stirring for 10min, adding ADT-OH (0.226g and 1mmol), reacting at 80 ℃, monitoring the reaction by TLC, cooling to room temperature after the reaction is finished, pouring the reaction liquid into ice water, stirring for 30min, performing suction filtration, performing vacuum drying, and separating dichloromethane through silica gel column chromatography: ethyl acetate (2:1) was added to 1mL of methanol to give 0.1g of an orange solid in a yield of 17%, Mp: 116-.
1H NMR(400MHz,DMSO)δ9.39(s,1H),8.42(s,1H),7.90-7.83(m,3H),7.75(s,1H),7.38(s,1H),7.32 (d,J=8.0Hz,1H),7.21(s,1H),7.13(d,J=8.0Hz,2H),6.99(d,J=12.0Hz,1H),4.32-4.29(m,4H),3.97 (s,3H),3.79(d,J=8.0Hz,6H),2.32-2.29(m,2H).
13C NMR(101MHz,DMSO)δ215.25,174.24,162.40,157.10,153.73,149.31,148.91,145.88,134.64, 133.02,129.50,128.72,116.54,116.02,115.83,115.62,112.26,109.24,108.57,108.14,102.66,65.49,65.29, 56.78,56.23,56.07,28.75.
LC-MSm/z:594.9[M+H]+.
Synthetic example 7: synthesis of QHS-13
Figure BDA0001030122650000172
Adding 4- (4-bromoanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7b) (0.423g and 1mmol), anhydrous DMF10mL, potassium carbonate (0.276g and 2mmol), a catalytic amount of potassium iodide into a single-neck flask, stirring for 10min, adding ACS81(0.356g and 2mmol), reacting at 70 ℃, monitoring the reaction by TLC, cooling to room temperature after the reaction is finished, pouring the reaction liquid into ice water, stirring for 30min, extracting with ethyl acetate, combining organic layers, washing with saturated saline, drying with anhydrous sodium sulfate, evaporating the solvent under reduced pressure, drying under vacuum, and separating petroleum ether by silica gel column chromatography: ethyl acetate (1:2) to give 0.09g of an off-white solid in a yield of 16%, Mp: 105-.
1H NMR(400MHz,DMSO)δ9.53(s,1H),8.49(s,1H),7.84(d,J=12.0Hz,3H),7.57(d,J=12.0Hz,2H), 7.22(d,J=8.0Hz,1H),5.84-5.69(m,1H),5.21-5.15(m,1H),5.11-5.05(m,1H),4.33-4.24(m,4H),3.98(s, 3H),3.16(d,J=8.0Hz,2H),2.67-2.61(m,2H),2.51(s,2H),2.20-2.14(m,2H).
13C NMR(101MHz,DMSO)δ171.98,168.85,156.54,153.93,153.16,147.44,139.45,134.98,131.67, 128.77,124.43,117.57,115.32,109.43,102.46,65.69,61.67,56.77,34.49,34.07,28.31.
LC-MSm/z:563.8[M-H]-.
Synthesis example 8: synthesis of QHS-15
Figure BDA0001030122650000181
Adding 4- (3-bromoanilino) -6-methoxy-7- (3-chloropropoxy) quinazoline (7d) (0.423g and 1mmol), anhydrous DMF10mL, potassium carbonate (0.276g and 2mmol), a catalytic amount of potassium iodide into a single-neck flask, stirring for 10min, adding ACS81(0.356g and 2mmol), reacting at 70 ℃, monitoring the reaction by TLC, cooling to room temperature after the reaction is finished, pouring the reaction liquid into ice water, stirring for 30min, extracting with ethyl acetate, combining organic layers, washing with saturated saline, drying with anhydrous sodium sulfate, evaporating the solvent under reduced pressure, drying under vacuum, and separating petroleum ether by silica gel column chromatography: ethyl acetate (1:2) to give 0.11g of an off-white solid in a yield of 20%, Mp: 101-.
1H NMR(400MHz,DMSO)δ9.54(s,1H),8.53(s,1H),8.16(s,1H),7.90(d,J=8.0Hz,1H),7.84(s,1H), 7.36(t,J=8.0Hz 1H),7.29(d,J=8.0Hz,1H),7.24-7.22(m,1H),5.79-5.69(m,1H),5.14-5.05(m,2H), 4.33-4.23(m,4H),3.98(s,3H),3.16(d,J=4.0Hz,2H),2.63(t,J=4.0Hz,2H),2.52-2.51(m,2H), 2.17-2.11(m,2H).
13C NMR(101MHz,DMSO)δ171.98,165.97,156.44,153.99,153.11,149.55,147.51,134.97,130.82, 128.77,126.04,124.44,121.65,121.01,117.56,109.43,102.37,65.71,61.50,56.77,34.48,34.07,28.14.
LC-MSm/z:565.1[M+H]+.
Integral synthesis route and steps of LHS series compound
2-amino-5-nitrobenzoic acid is taken as a raw material, and an intermediate (4a,4b,4c) is synthesized by ring closure, chlorination, nitration and reduction reaction and then connected with H2S donor to obtain 6 LHS series compounds, and the specific synthetic route is as follows.
Figure BDA0001030122650000191
Synthesis (1) of 6-nitro-4 (3H) -quinazolinone
Figure BDA0001030122650000192
2-amino-5-nitrobenzoic acid (3.64g, 20mmol), formamidine acetate (4.16g, 40mmol) and 50mL of ethylene glycol monomethyl ether were added to a 100mL three-necked flask and reacted under reflux. TLC monitoring, after the reaction, the reaction solution was poured into a beaker, placed in a freezer at-20 ℃ overnight, filtered to obtain 3.0g of yellow solid with yield of 78.5% and Mp of 280 plus 283 ℃.
Synthesis of 4- (3-trifluoromethylanilino) -6-nitroquinazoline (3a)
Figure BDA0001030122650000193
6-Nitro-4 (3H) -quinazolinone (1) (3.8g, 20mmol) was added to a 250ml three-necked flask, SOCl was added240mL, slowly dripping 4mL of DMF, heating to 80 ℃, carrying out reflux reaction for 1h, and then supplementing 20mL of SOCl2And 2ml of DMF, and the reaction was continued under reflux. TLC monitoring, after the reaction, the reaction solution was transferred to a 250mL eggplant-shaped bottle and SOCl was removed by reduced pressure rotary evaporation2Then, petroleum ether was added thereto, the solid was washed, and the obtained solid was put into a 250mL single-neck eggplant-shaped bottle containing 3-trifluoromethylaniline (6.64g, 40mmol) and 60mL of isopropyl alcohol, and the mixture was refluxed at an elevated temperature. TLC monitoring, after the reaction is finished, suction filtration is carried out, and the filter cake is washed by isopropanol to obtain yellow solid with the yield of 60%.
Synthesis of 4- (3-bromoanilino) -6-nitroquinazoline (3b)
Figure BDA0001030122650000201
6-Nitro-4 (3H) -quinazolinone (1) (3.8g, 20mmol) was added to a 250mL three-necked flask, and thionyl chloride (SOCl) was added2) 40mL, slowly adding 4mL of N, N-Dimethylformamide (DMF) dropwise, heating to 80 ℃, carrying out reflux reaction for 1h, and supplementing 20mL of SOCl2And 2mL of DMF was added to the mixture,the reaction was continued under reflux. TLC monitoring, after the reaction, the reaction solution was transferred to a 250mL eggplant-shaped bottle and SOCl was distilled off under reduced pressure2Adding petroleum ether, washing the solid, adding the obtained solid into a 250ml single-opening eggplant-shaped bottle weighed to contain 3-bromoaniline (6.88g, 40mmol) and 60ml of isopropanol, heating, carrying out reflux reaction, monitoring by TLC, after the reaction is finished, carrying out suction filtration, and washing a filter cake by using the isopropanol to obtain a yellow solid with the yield of 90%.
Synthesis of 4- (3-chloro-4-fluoroanilino) -6-nitroquinazoline (3c)
Figure BDA0001030122650000202
6-Nitro-4 (3H) -quinazolinone (1) (3.8g, 20mmol) was added to a 250mL three-necked flask, and thionyl chloride (SOCl) was added2) 40mL, slowly adding 4mL of N, N-Dimethylformamide (DMF) dropwise, heating to 80 ℃, carrying out reflux reaction for 1h, and supplementing 20mL of SOCl2And 2mL of DMF, and the reaction was continued at reflux. TLC monitoring, after the reaction, the reaction solution was transferred to a 250mL eggplant-shaped bottle and SOCl was removed by reduced pressure rotary evaporation2Adding petroleum ether, washing the solid, adding the obtained solid into a 250mL single-mouth eggplant-shaped bottle which is called to contain 3-chloro-4-fluoroaniline (5.87g, 40mmol) and 60mL of isopropanol, heating, carrying out reflux reaction, monitoring by TLC, after the reaction is finished, carrying out suction filtration, and washing a filter cake by using the isopropanol to obtain a yellow solid with the yield of 50%.
Synthesis of 4- (3-trifluoromethylanilino) -6-aminoquinazoline (4a)
Figure BDA0001030122650000203
4- (3-trifluoromethylanilino) -6-nitroquinazoline (3a) (4.3g), 150mL of ethanol and 50mL of acetic acid are added into a 500mL three-necked flask, mechanically stirred, heated, and then iron powder (4.5g) activated by hydrochloric acid is added into the flask in portions and refluxed. And monitoring by TLC, filtering while the solution is hot after the reaction is finished, carrying out reduced pressure spin-drying on the filtrate, adding distilled water, stirring, pouring into a 500mL beaker, and carrying out suction filtration to obtain a light green solid with the yield of 60%.
Synthesis of 4- (3-bromoanilino) -6-aminoquinazoline (4b)
Figure BDA0001030122650000211
4- (3-bromoanilino) -6-nitroquinazoline (3b) (5.0g), 150mL of ethanol and 50mL of acetic acid are added into a 500mL three-necked flask, mechanically stirred, heated, added with iron powder activated by hydrochloric acid (5.0g) in portions into the flask, and refluxed for reaction. And monitoring by TLC, filtering while the solution is hot after the reaction is finished, carrying out reduced pressure spin-drying on the filtrate, adding distilled water, stirring, pouring into a 500mL beaker, and carrying out suction filtration to obtain a light green solid with the yield of 40%. Synthesis of 4- (3-chloro-4-fluoroanilino) -6-aminoquinazoline (4c)
Figure BDA0001030122650000212
4- (3-chloro-4-fluoroanilino) -6-nitroquinazoline (3c) (3.8g), 150mL of ethanol and 50mL of acetic acid were placed in a 500mL three-necked flask, mechanically stirred, warmed, and iron powder (4.5g) activated with hydrochloric acid was added in portions to the flask, and the reaction was refluxed. Monitoring by TLC, filtering while the solution is hot after the reaction is finished, carrying out rotary evaporation on the filtrate under reduced pressure until the filtrate is dry, adding distilled water, stirring, pouring into a 500mL beaker, and carrying out suction filtration to obtain a light green solid with the yield of 40%.
Synthesis of 5-p-carboxyethoxyphenyl-3H-1, 2-dithiole-3-thione (ACS-60)
Figure BDA0001030122650000213
Adding ADT-OH (0.45g, 2mmol), potassium carbonate (0.55g, 4mmol) and ethyl bromoacetate (0.66mL, 6mmol) into a three-neck flask, adding anhydrous DMF30mL, fully stirring for dissolving, reacting at 50 ℃ for 3h, monitoring by TLC, cooling to room temperature after the reaction is finished, extracting with ethyl acetate, washing with distilled water for 3 times, drying the ethyl acetate layer with anhydrous sodium sulfate, and concentrating under reduced pressure to obtain a tan oily substance. The oily substance (0.31g, 1mmol) obtained in the previous step is directly used for the next step of reaction, 50% sulfuric acid (2.2mL, 20mmol) and 20mL acetic acid are added, the reaction is carried out at 100 ℃ for 2h, the reaction is monitored by TLC, the reaction is cooled to room temperature after the reaction is finished, distilled water is added, and the deep green solid is obtained by suction filtration, and the yield is 49%.
Synthetic example 9: synthesis of FHS-1
Figure BDA0001030122650000221
Adding ACS-60(0.312g, 1.1mmol) and DMF10mL into a single-neck flask, stirring to dissolve, adding HATU (0.418g, 1.1mmol) and triethylamine (0.122g, 1.2mmol), stirring and activating at room temperature for 20min, slowly adding 4- (3-trifluoromethylanilino) -6-aminoquinazoline (0.30g, 1.1mmol) (4a), reacting at 25 ℃ for 24h, monitoring the reaction by TLC, pouring the reaction liquid into 100mL of ice water after the reaction is finished, stirring and filtering to obtain a gray solid, and performing silica gel column chromatography to separate a final light orange solid product, wherein the yield is 15%, and the Mp:108 and 109 ℃.
1H NMR(400MHz,DMSO)δ10.52(s,1H),10.05(s,1H),8.79(s,1H),8.60(s,1H),8.27(s,1H),8.20(d,J =8.0Hz,1H),7.96-7.91(m,3H),7.83(d,J=8.0Hz,1H),7.76(d,J=4.0Hz,1H),7.60(t,J=8.0Hz,1H), 7.43(s,1H),7.20(d,J=8.0Hz,2H),4.95(s,2H).
13C NMR(101MHz,DMSO)δ215.38,176.88,166.77,161.68,157.86,153.67,147.43,140.73,136.50, 134.86,129.92,129.87,129.55,129.45,129.02,128.71,128.10,128.07,126.16,124.84,124.55,118.66, 116.28,115.86,67.54.
LC-MSm/z:571.1[M+H]+.
Synthetic example 10: synthesis of FHS-2
Figure BDA0001030122650000222
Adding ACS81(0.267g, 1.5mmol) and DMF10mL into a single-neck flask, stirring to dissolve, adding HATU (0.57g, 1.5mmol) and triethylamine (0.167g, 1.6mmol), stirring and activating at room temperature for 20min, slowly adding 4- (3-trifluoromethylanilino) -6-aminoquinazoline (0.252g, 0.8mmol) (4a), reacting at 25 ℃ for 24h, monitoring the reaction by TLC, pouring the reaction liquid into 100mL of ice water, stirring, extracting with ethyl acetate, washing with saline, drying with anhydrous sodium sulfate, evaporating the solvent under reduced pressure, drying under vacuum, and separating by silica gel column chromatography to obtain a final beige solid product 0.06g, with the yield of 16%, Mp:107 and 108 ℃.
1H NMR(400MHz,DMSO)δ10.37(s,1H),10.02(s,1H),8.76(s,1H),8.57 (s,1H),8.27-8.20(m,2H),7.82(dd,J=8.0Hz,2H),7.60(d,J=8.0Hz,1H),7.41(s,1H),5.90-5.80(m, 1H),5.19(dd,J=16.0Hz,2H),3.43(d,J=8.0Hz,2H),3.05(m,2H),2.51-2.50(m,2H).
13C NMR(101MHz,DMSO)δ169.88,157.82,149.53,147.12,140.82,137.33,134.03,129.91,128.96, 128.04,126.13,126.06,119.15,118.42,117.29,115.95,100.00,41.52,36.41,33.93.
LC-MSm/z:465.1[M+H]+.
Synthetic example 11: synthesis of XHS-1
Figure BDA0001030122650000231
Adding ACS-60(0.43g, 1.5mmol) and DMF10mL into a single-neck flask, stirring to dissolve, adding HATU (0.57g, 1.5mmol) and triethylamine (0.182g, 1.6mmol), stirring and activating at room temperature for 20min, slowly adding 4- (3-bromoanilino) -6-aminoquinazoline (0.472g, 1.5mmol) (4b), reacting at 25 ℃ for 24h, monitoring the reaction by TLC, pouring the reaction liquid into 100mL of ice water after the reaction is finished, stirring, performing suction filtration to obtain a gray solid, and performing silica gel column chromatography to separate a final light orange solid product, wherein the yield is 10%, and the Mp is 112 ion 113 ℃.
1H NMR(400MHz,DMSO)δ10.55(s,1H),9.90(s,1H),8.77(s,1H),8.57(s,1H),8.17(s,1H),7.97(d,J =12.0Hz,1H),7.87(d,J=8.0Hz,2H),7.79(d,J=8.0Hz,2H),7.72(s,1H),7.30(d,J=8.0Hz,1H),7.25 (s,1H),7.19(d,J=8.0Hz,2H),4.93(s,2H).
13C NMR(101MHz,DMSO)δ215.34,173.94,166.71,161.66,157.80,153.69,147.40,141.55,136.42, 134.83,130.63,129.39,128.68,126.34,125.56,124.84,121.59,121.33,116.26,116.07,115.88,67.54.
LC-MSm/z:583.0[M+H]+.
Synthetic example 12: synthesis of XHS-2
Figure BDA0001030122650000232
Adding ACS81(0.267g, 1.5mmol) and DMF10mL into a single-neck flask, stirring to dissolve, adding HATU (0.57g, 1.5mmol) and triethylamine (0.167g, 1.6mmol), stirring and activating at room temperature for 20min, slowly adding 4- (3-bromoanilino) -6-aminoquinazoline (0.252g, 0.8mmol) (4b), reacting at 25 ℃ for 24h, monitoring the reaction by TLC, pouring the reaction liquid into 100mL of ice water, stirring, extracting with ethyl acetate, washing with saline, drying with anhydrous sodium sulfate, evaporating the solvent under reduced pressure, drying under vacuum, and separating by silica gel column chromatography to obtain a final beige solid product 0.06g, with the yield of 16%, Mp:108 and 109 ℃.
1H NMR(400MHz,DMSO)δ10.35(s,1H),9.88(s,1H),8.71(s,1H),8.57(d,J=4.0Hz,H),8.17(t,J= 4.0Hz,1H),7.87(d,J=8.0Hz,2H),7.78(dd,J=4.0Hz,1H),7.32-7.25(m,2H),5.90-5.80(m,1H),5.18 (dd,J=16.0Hz,2H),3.43(d,J=8.0Hz,2H),3.04(t,J=8.0Hz,2H),2.51-2.50(m,2H).
13C NMR(101MHz,DMSO)δ169.91,157.78,153.47,147.06,141.61,137.26,134.02,130.73,128.90, 126.37,124.82,121.59,121.37,119.17,115.92,100.00,41.50,36.39,33.93.
LC-MSm/z:476.0[M+H]+.
Synthetic example 13: synthesis of LHS-1
Figure BDA0001030122650000241
Adding ACS-60(0.313g, 1.1mmol) and DMF10mL into a single-neck flask, stirring to dissolve, adding HATU (0.418g, 1.1mmol) and triethylamine (0.122g, 1.2mmol), stirring and activating at room temperature for 20min, slowly adding 4- (3-chloro-4-fluoroanilino) -6-aminoquinazoline (0.317g, 1.1mmol) (4c), reacting at 25 ℃ for 24h, monitoring the reaction by TLC, pouring the reaction liquid into 100mL of ice water after the reaction is finished, stirring, performing suction filtration to obtain a gray solid, and performing silica gel column chromatography to separate a final light orange solid product 0.095g, wherein the yield is 15%, and the Mp:115-116 ℃.
1H NMR(400MHz,CDCl3)δ8.83(s,1H),8.76(s,1H),8.53(s,1H),7.98(d,J=12.0Hz,2H),7.75-7.70 (m,3H),7.63(d,J=8.0Hz,1H),7.57(d,J=8.0Hz,1H),7.43(s,1H),7.22(d,J=8.0Hz,1H),7.19-7.16 (m,2H),4.79(s,2H).
13C NMR(101MHz,DMSO)δ215.28,172.40,166.73,161.61,136.80,134.79,129.29,128.60,128.19, 124.85,124.74,119.53,119.35,116.88,116.66,116.22,116.02,113.08,100.00,67.49.
LC-MSm/z:556.0[M+H]+.
Synthesis example 14: synthesis of LHS-2
Figure BDA0001030122650000242
Adding ACS81(0.196g, 1.1mmol) and DMF10mL into a single-neck flask, stirring to dissolve, adding HATU (0.418g, 1.1mmol) and triethylamine (0.122g, 1.2mmol), stirring and activating at room temperature for 20min, slowly adding 4- (3-chloro-4-fluoroanilino) -6-aminoquinazoline (0.317g, 1.1mmol) (4c), reacting at 25 ℃ for 24h, monitoring the reaction by TLC, pouring the reaction liquid into 100mL of ice water, stirring, extracting with ethyl acetate, washing with saline, drying with anhydrous sodium sulfate, evaporating the solvent under reduced pressure, drying under vacuum to obtain a beige solid, separating by silica gel column chromatography to obtain the final beige solid product of 0.084g, yield of 17%, Mp of 121 ℃.
1H NMR(400MHz,DMSO)δ10.35(s,1H),9.90(s,1H),8.71(s,1H),8.54(s,1H),8.13(dd,J=4.0Hz, 1H),7.86-7.75(m,3H),7.38(s,1H),5.90-5.80(m,1H),5.18(dd,J=16.0Hz,2H),3.43(d,J=8.0Hz,2H), 3.04(t,J=8.0Hz,2H),2.51-2.50(m,2H).
13C NMR(101MHz,DMSO)δ169.86,157.79,153.43,152.59,147.03,137.27,137.13,134.02,128.91, 124.26,123.12,119.32,119.14,116.95,115.82,112.01,41.53,36.40,33.95.
LC-MSm/z:450.1[M+H]+.
Study of biological Activity
Preliminary screening for in vitro Activity of Compounds
Proliferation inhibition experiment of adherent cells-SRB method: to examine the possible biological activity of the synthesized compounds, cells in logarithmic growth phase were plated in 96-well plates (200. mu.L/well) in certain numbers, incubated for 24h to adhere to the walls and then dosed. Each drug concentration is provided with 2 multiple wells, and corresponding zero setting wells and blank control are arranged. After 72h of drug action, the adherent cells were added with 50% TCA (50. mu.L/well), fixed at 4 ℃ for 1h, the fixative was poured off, washed 5 times with distilled water, and naturally dried. Add 100. mu.L of 4mg/mL SRB to each well, stain at room temperature for 15min, discard, wash with 1% glacial acetic acid for 5 times, and dry naturally. Finally, 150. mu.L of 10mM Tris solution was added to each well, shaken well, and OD was measured at 565nm using a variable wavelength microplate reader (VERSAmaxTM, Molecular Device). The cell growth inhibition rate was calculated by the formula.
Inhibition ratio (%) - (OD value)Control wellOD valueMedicine feeding hole) OD valueControl well×100%
Calculating half inhibitory concentration IC by LOGIT method according to each concentration inhibition rate50. The experiment was repeated 3 more times and the data are expressed as mean ± SD.
Preliminary screening of the compounds
Cells in logarithmic growth phase were plated in 96-well plates (200. mu.L/well) in a certain number, and cultured for 24h to adhere to the walls and then dosed with drugs. After 72 hours of drug action, the OD values were measured and the average value was calculated to calculate the cell proliferation inhibition rate (Table 3).
TABLE 3 growth inhibition ratio of each compound
Figure BDA0001030122650000251
Figure BDA0001030122650000261
a. The final concentration of the detection target compound was 20. mu.M. The positive control drugs MHS-1 and MHS-3 have concentrations of 45. mu.M and 55. mu.M, respectively.
b. The compound concentration was 10. mu.M. c. The concentration of the compound was 50. mu.M.
TABLE 4 IC of the Compounds on HepG2 cell line50Value of
Figure BDA0001030122650000262
(mean±SD,n=3)
Target compound release H2Determination of S level
By means of H2H released by fluorescent probe technology of S detection on target compound2And S, measuring.
Preparing a probe solution: NIR-HS was dissolved in acetone to prepare a 1mM stock solution, which was diluted to 200. mu.M solution for use, and the probe solution was stored in low temperature and protected from light.
Na2Preparing an S stock solution: 5mg EDTA was dissolved in 10mL double distilled water, and nitrogen was introduced into the solution for 15 min. Under the protection of nitrogen, 48mg of Na2S·9H2O dissolved in the solution to give 20mM Na2S stock solution, which is diluted into 100 mu M solution for standby and needs to be prepared for use.
Preparation of PBS: potassium dihydrogen phosphate (KH)2PO4)0.24g of disodium hydrogen phosphate (Na)2HPO4)1.44g, sodium chloride (NaCl)8.0g, potassium chloride (KCl)0.2g, and water to 1000 mL. pH 7.4, requires low temperature storage.
Preparation of QHS-6 Compound: QHS-6 was dissolved in acetone to prepare a 2.5mM stock solution, which was diluted to 1.25mM, 625. mu.M, 100. mu.M, and 50. mu.M solutions for future use, and stored at low temperature.
Formulation of QHS-13 Donor Compounds: QHS-13 was dissolved in acetone to prepare a 5mM stock solution, which was diluted to 1mM, 500. mu.M, 100. mu.M, and 50. mu.M solutions for future use, and stored at low temperature.
Preparation of LHS-1 donor Compound: LHS-1 was dissolved in acetone to prepare a 2.5mM stock solution, which was diluted to 1.25mM, 625. mu.M, 100. mu.M, 50. mu.M solutions for future use and stored at low temperature.
3.1.2.3 qualitative determination of H released by target Compound2S
H is to be2The S fluorescent probe NIR-HS (final concentration of 10 μ M) was incubated with the target compound (0 μ M,5 μ M, 10 μ M,50 μ M, 100 μ M and 200 μ M) in PBS buffer (PBS: acetone ═ 2:3) for 3h (37 ℃). Thereafter, the fluorescence emission spectrum (. lamda.) was measured with an F4600 fluorescence spectrophotometerex=670nm,λem723nm), at least three replicates per concentration were tested at least 3 times.
Quantitative determination of H released by target Compounds2S
(1) Establishment of a standard curve:
h is to be2NIR-HS (final concentration of 10. mu.M) and Na as S fluorescent probes2S (0 μ M,2 μ M,4 μ M,6 μ M,8 μ M) was incubated in PBS buffer (PBS: acetone ═ 2:3) for 3h (37 ℃). Thereafter, the fluorescence emission spectrum (. lamda.) was measured with an F4600 fluorescence spectrophotometerex=670nm,λem723nm) and based on the fluorescence intensity, a standard curve was made. There were at least three replicates per concentration, measured at least 3 times.
(2)H2Quantitative determination of S release:
h is to be2The S fluorescent probe NIR-HS (final concentration of 10 μ M) and the target compound (20 μ M) were incubated in PBS buffer (PBS: acetone ═ 2:3) for 0min, 5min, 30min, 60min, 90min, 120min, 150min, 180min (37 ℃) respectively. Thereafter, the fluorescence emission spectrum (. lamda.) was measuredex=670nm,λem=723nm),
According to Na2S Standard Curve, converted to H Release from each Donor Compound2The amount of S. At least three replicates were tested at each time point for at least 3 times.
H2Measurement of S level
1. Target compound release H2Qualitative determination of S
Selecting compounds with relatively good activity (QHS-6, QHS-13 and L) from QHS-a series, QHS-b series and LHS-a series of compoundsHS-1), determination of its H2And (4) releasing S.
The experimental results show that: after incubation of compound QHS-6(10,50,100, 200. mu.M) with fluorescent probe NIR-HS (10. mu.M), a significant fluorescence response was produced with a significant increase in fluorescence intensity (10. mu.M, 33.77 + -1.55; 50. mu.M, 50.15 + -2.10; 100. mu.M, 66.08 + -3.59; 200. mu.M, 97.27 + -3.27) compared to no donor (0. mu.M, 29.65 + -1.42) ((0. mu.M, 29.65 + -1.42))**P<0.01) (fig. 1), and the fluorescence intensity gradually increased with increasing compound concentration. This indicates that the compound QHS-6 is capable of releasing H in PBS buffer2And S. The results are shown in FIG. 1.
The experimental results show that: after incubation of compound QHS-13(10,50,100, 200. mu.M) with fluorescent probe NIR-HS (10. mu.M), a significant fluorescence response was generated with a significant increase in fluorescence intensity (10. mu.M, 48.41 + -5.95; 50. mu.M, 66.58 + -6.34; 100. mu.M, 84.95 + -5.48; 200. mu.M, 120.39 + -7.21) compared to the absence of donor (0. mu.M, 36.78 + -1.29) ((0. mu.M, 36.78 + -1.29))**P<0.01) (fig. 2), and the fluorescence intensity gradually increased with increasing compound concentration. This indicates that the compound QHS-13 is capable of releasing H in PBS buffer2S。
The experimental results show that: after incubation of compound LHS-1(10,50,100, 200. mu.M) with fluorescent probe NIR-HS (10. mu.M), a significant fluorescence response was produced with a significant increase in fluorescence intensity (10. mu.M, 41.10 + -2.65; 50. mu.M, 83.35 + -2.83; 100. mu.M, 116.11 + -12.59; 200. mu.M, 204.02 + -14.38) compared to no donor (0. mu.M, 29.04 + -1.08) ((0. mu.M, 29.04 + -1.08))*P<0.05,**P<0.01) (fig. 3), and the fluorescence intensity gradually increased with increasing compound concentration. This indicates that LHS-1, a compound capable of releasing H in PBS buffer2S。
2. Target compound release H2Quantitative determination of S
Fluorescence intensity and Na of probe NIR-HS2The standard curve of S concentration (0,2,4,6, 8. mu.M) is shown in FIG. 4, and shows a good linear relationship (0. mu.M, 1.0. + -. 0.0; 2. mu.M, 2.05. + -. 0.045; 4. mu.M, 3.45. + -. 0.24; 6. mu.M, 4.78. + -. 0.39; 8. mu.M, 5.78. + -. 0.23).
Incubating the compounds (QHS-6, QHS-13, LHS-1, 20 μ M) with fluorescent probe NIR-HS, detecting the fluorescence intensity every 30min, and then, according to a standard curve, measuring the fluorescence intensityConversion of the value into H2Concentration of S, i.e. H2S concentration-time curve.
The experimental results show that: the compounds QHS-6, QHS-13 and LHS-1 can release H2And S. H of compound QHS-132The S release is obviously more than that of the compounds QHS-6 and LHS-1. H of compound QHS-13 within 1H2S release speed is high, and H is obtained after 2H2The release of S tends to be smooth; within 2H, H of the compound LHS-12S is released slowly, and H is released after 2H2S, accelerating the speed; the compound QHS-6 releases H in 0-3H2The S speed was slower (table 5, fig. 5).
TABLE 5 liberated H of the test compounds2Change in S concentration
Figure BDA0001030122650000281
(mean±SD,n=3)。

Claims (3)

1. A compound or a pharmaceutically acceptable salt thereof,
Figure FDA0003081516560000011
2. a pharmaceutical composition comprising the compound of claim 1 or a pharmaceutically acceptable salt thereof as an active ingredient or a main active ingredient, together with pharmaceutically acceptable excipients.
3. Use of the compound of claim 1 or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating liver cancer.
CN201610477893.8A 2016-06-26 2016-06-26 H based on quinazoline structure2S donor compound and application thereof Active CN106083836B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201610477893.8A CN106083836B (en) 2016-06-26 2016-06-26 H based on quinazoline structure2S donor compound and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201610477893.8A CN106083836B (en) 2016-06-26 2016-06-26 H based on quinazoline structure2S donor compound and application thereof

Publications (2)

Publication Number Publication Date
CN106083836A CN106083836A (en) 2016-11-09
CN106083836B true CN106083836B (en) 2021-08-20

Family

ID=57252919

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201610477893.8A Active CN106083836B (en) 2016-06-26 2016-06-26 H based on quinazoline structure2S donor compound and application thereof

Country Status (1)

Country Link
CN (1) CN106083836B (en)

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI1274692T1 (en) * 2000-04-07 2006-12-31 Astrazeneca Ab Quinazoline compounds
CN101108846B (en) * 2007-08-10 2011-02-09 东南大学 4- fragrant amido quinazoline derivatives and method of manufacturing the same and application in pharmacy thereof
CN101148439B (en) * 2007-09-14 2011-06-22 东南大学 Preparing method for gefitinib
CN101857588B (en) * 2010-06-08 2013-11-27 东南大学 4-aromatic aminoquinazoline derivative and purpose thereof
CA2827887C (en) * 2011-08-15 2020-06-09 Research Foundation Of The City University Of New York No- and h2s-releasing compounds

Also Published As

Publication number Publication date
CN106083836A (en) 2016-11-09

Similar Documents

Publication Publication Date Title
TWI229667B (en) Quinoline derivative and quinazoline derivative
KR100832593B1 (en) Quinazoline derivatives as an signal trnasduction inhibitor and method for the preparation thereof
Zhang et al. Design and discovery of 4-anilinoquinazoline-acylamino derivatives as EGFR and VEGFR-2 dual TK inhibitors
EP3299369B1 (en) Pyrido-azaheterecydic compound and preparation method and use thereof
KR100658891B1 (en) Quinazoline derivatives inhibiting the growth of cancer cell and preparation thereof
Yin et al. Design, synthesis and biological evaluation of novel EGFR/HER2 dual inhibitors bearing a oxazolo [4, 5-g] quinazolin-2 (1H)-one scaffold
Zhou et al. Design, synthesis and pharmacological evaluation of 6, 7-disubstituted-4-phenoxyquinoline derivatives as potential antitumor agents
CN102838590A (en) Amino quinazoline derivative and application thereof in preparation of antineoplastic drugs
Yu et al. Design, synthesis and antitumor activity of 4-aminoquinazoline derivatives targeting VEGFR-2 tyrosine kinase
Zhai et al. Design, synthesis and biological evaluation of novel 4-phenoxy-6, 7-disubstituted quinolines possessing (thio) semicarbazones as c-Met kinase inhibitors
CA2719523C (en) Polymorphic forms of 4-phenylamino quinazoline derivatives, the preparation methods and uses thereof
WO2023005280A1 (en) Preparation and application of aminopyrimidine derivative selectively targeting cdk9
CN113773305B (en) Aminopyrimidine derivative and application thereof as EGFR tyrosine kinase inhibitor
Moghadam et al. 6, 7-disubstituted-4-anilinoquinazoline: design, synthesis and anticancer activity as a novel series of potent anticancer agents
JP5504460B2 (en) Salt form of 4-phenylaminoquinazoline derivative
CN114920704A (en) Phenyl piperazine quinazoline compound or pharmaceutically acceptable salt thereof, preparation method and application
WO2007055513A1 (en) Quinazoline derivatives as a signal transduction inhibitor and method for the preparation thereof
CN109988110B (en) 4-phenoxy quinoline sulfonylurea compound, intermediate for synthesizing the compound and its preparation method and use
CN106083836B (en) H based on quinazoline structure2S donor compound and application thereof
CN103864680B (en) There is the 7-chloro-4-oxo-quinoline derivative of anti-tumor activity
CN106045980A (en) Quinazoline derivative and preparation method thereof
Liu et al. Synthesis and anti-tumor activity evaluation of novel 7-fluoro-4-(1-piperazinyl) quinolines
CN113880814B (en) Pyrimidine amine compound and application thereof
CN107501283B (en) Preparation of substituted arylmethyl hetero-substituted anilino ethylene glycol ether cycloquinazoline and application of tumor treatment drug
CN112358469A (en) Angiogenesis inhibitor, preparation method and application thereof

Legal Events

Date Code Title Description
C06 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