Coumarin derivatives and analogs, and preparation method and application thereof

文档序号:823938 发布日期:2021-03-30 浏览:35次 中文

阅读说明:本技术 香豆素衍生物和类似物及其制备方法和用途 (Coumarin derivatives and analogs, and preparation method and application thereof ) 是由 陈俐娟 于 2020-09-15 设计创作,主要内容包括:本发明涉及香豆素衍生物和类似物及其制备方法和用途,属于化学医药领域。本发明提供了式Ⅰ所示的化合物或其药学上可接受的盐。本发明还提供了上述化合物的制备方法和用途。生物学实验表明,这类化合物具有较强的体外抗纤维化效果,在TGF-β诱导的NRK-49F细胞上能明显减少细胞间胶原纤维的沉积,对HUVEC细胞的迁移有抑制性。同时,这类结构的化合物在在四氯化碳诱导的肝纤维化、博来霉素诱导的肺纤维化小鼠模型上都具有一定的疗效,毒性较小,为临床治疗包括肝纤维化、肺纤维化、肾纤维化在内的组织纤维化疾病提供了新的选择。(The invention relates to coumarin derivatives and analogs, and a preparation method and application thereof, and belongs to the field of chemical medicine. The invention provides a compound shown as a formula I or a pharmaceutically acceptable salt thereof. The invention also provides a preparation method and application of the compound. Biological experiments show that the compound has stronger in-vitro anti-fibrosis effect, can obviously reduce the deposition of intercellular collagen fibers on the NRK-49F cells induced by TGF-beta, and has inhibition on the migration of HUVEC cells. Meanwhile, the compounds with the structure have certain curative effect on mouse models of hepatic fibrosis induced by carbon tetrachloride and pulmonary fibrosis induced by bleomycin, have low toxicity, and provide a new choice for clinically treating tissue fibrosis diseases including hepatic fibrosis, pulmonary fibrosis and renal fibrosis.)

1. A compound of formula I or a pharmaceutically acceptable salt thereof:

x is selected from O or NH;

R1、R2、R3、R4、R5、R6independently selected from H, nitro, C1~C6Alkoxy or

Y is selected from O, S or NR12;R7、R8、R9、R10、R11Independently selected from H, halogen orR12Selected from H, C1~C6An alkyl group; r13Selected from NR14R15The number of the hetero atoms is 1-2; r14、R15Independently selected from H, C1~C6Alkyl, substituted or unsubstituted 5-6 membered heterocycle, heteroatom is N or O, the number of heteroatoms is 1-2.

2. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein: r13Selected from NR14R15Unsubstituted 5-membered heterocycle, substituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, substituted 6-membered heterocycle; r14、R15Independently selected from H, C1~C6Alkyl, unsubstituted 5-membered heterocycle, substituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, substituted 6-membered heterocycle;

preferably, R13Selected from NR14R15Unsubstituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, C1~C4Alkyl-substituted 6-membered heterocycle, tert-butoxycarbonyl-substituted 6-membered heterocycle, -COR16Substituted 6-membered heterocyclic ring, -COOR17Substituted 6-membered heterocycles, -NR18R19A substituted 6-membered heterocyclic ring; r14、R15Independently selected from H, C1~C6Alkyl, unsubstituted 6-membered heterocycle, tert-butoxycarbonyl substituted 6-membered heterocycle, -COOR17A substituted 6-membered heterocyclic ring; r16Is selected from C1~C3Alkyl radical, C3~C6A cycloalkyl group; r17、R18、R19Independently selected from C1~C4An alkyl group;

further preferably, R13Selected from NR14R15Unsubstituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, C1~C4Alkyl-substituted 6-membered heterocycle, tert-butoxycarbonyl-substituted 6-membered heterocycle, -COR16Substituted 6-membered heterocyclic ring, -COOR17Substituted 6-membered heterocycles, -NR18R19A substituted 6-membered heterocyclic ring; r14、R15Independently selected from H, C1~C6Alkyl, unsubstituted 6-membered heterocycle, tert-butoxycarbonyl substituted 6-membered heterocycle, -COOR17A substituted 6-membered heterocyclic ring; r16Is selected from C1~C3Alkyl radical, C3~C6A cycloalkyl group; r17、R18、R19Independently selected from C1~C4An alkyl group; the 5-membered heterocycle has 1 heteroatom, and the heteroatom is N;

more preferably, R13Is selected from

Most preferably, R7、R8、R10、R11Independently selected from H or halogen; r9Is selected from

3. The compound according to claim 2, or a pharmaceutically acceptable salt thereof, wherein: when X is selected from O, the formula II:

wherein Y is selected from O or NR12;R1、R3、R4、R5、R6Independently selected from H, nitro, C1~C6An alkoxy group; r7、R8、R10、R11Independently selected from H or halogen; r12Selected from H or methyl.

4. A compound according to claim 3, characterized in that: r1、R3、R4、R5、R6Independently selected from H, nitro, C1~C3An alkoxy group; preferably, R1、R3、R4、R5、R6Independently selected from H, nitro or methoxy; more preferably, R1、R3、R6Are all selected from H, R4、R5Independently selected from H, nitro or methoxy;

and/or, R7、R8、R10、R11Independently selected from H or F; preferably, R7、R8、R11Are all selected from H, R10Selected from H or F;

and/or, R13Is selected from

5. The compound of claim 4, or a pharmaceutically acceptable salt thereof, having the formula:

6. the compound according to claim 2, or a pharmaceutically acceptable salt thereof, wherein: when X is selected from NH, Y is selected from O, S or NR12,R2、R4、R5Independently selected from H orAnd R is2、R4、R5One of which isR1、R3、R6Independently selected from H, nitro, C1~C6An alkoxy group; r7、R8、R10、R11Independently selected from H or halogen; r12Selected from H or methyl.

7. The compound according to claim 6, or a pharmaceutically acceptable salt thereof, wherein: r2Is selected fromWhen Y is selected from O, S or NR12;R4Or R5Is selected fromWhen, Y is selected from O;

and/or, R1、R3、R6Independently selected from H, nitro, C1~C3An alkoxy group; preferably, R1、R3、R6Are all selected from H;

and/or, R7、R8、R10、R11Independently selected from H or F; preferably, R7Selected from H or F, R8、R10、R11Are all selected from H;

and/or, R13Is selected from

8. The compound of claim 7, or a pharmaceutically acceptable salt thereof, having the formula:

9. a pharmaceutical composition comprising the compound of any one of claims 1 to 8 or a pharmaceutically acceptable salt thereof as an active ingredient, in combination with pharmaceutically acceptable auxiliary ingredients.

10. Use of a compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 9, in the manufacture of a medicament for the treatment of a fibrotic disease; preferably, the medicament for treating fibrotic diseases satisfies at least one of the following conditions:

the medicament is a medicament for inhibiting the expression of COL1A1, alpha-SMA and p-Smad3 protein;

the drug is a drug that inhibits the TGF-beta/Smad 3 pathway;

the fibrotic disease includes pulmonary fibrosis, hepatic fibrosis or renal fibrosis.

Technical Field

The invention relates to coumarin derivatives and analogs, and a preparation method and application thereof, and belongs to the field of chemical medicine.

Background

Fibrosis refers to the continuous repair of tissues or organs after the tissues or organs are damaged, and excessive fibrous connective tissues are formed due to excessive repair. It is manifested as a reactive, benign or pathological condition. The reactive process leading to injury, called scarring, if the fibrosis that occurs is derived from a single cell line, is called fibroma. Ideally, fibrosis occurs and deposits in connective tissue, gradually affecting the structure and function of normal tissues and organs. Fibrosis is a pathological condition that can be described by the use of over-deposited collagen. We usually define it as an excess of Extracellular matrix (ECM) deposition, fibrosis ultimately leads to scarring and thickening of the tissue, which is essentially an excessive injury repair response that ultimately affects normal organ function.

Renal fibrosis can be caused by different kidney diseases, eventually progressing to end stage renal disease. Although the pathogenesis of renal fibrosis has not been fully elucidated, it is usually manifested by hyperproliferation of renal resident cells (glomerular epithelial cells, mesangial cells and endothelial cells), repair of renal defects, abnormal activation of renal interstitial fibroblasts and excessive deposition of extracellular matrix, eventually leading to glomerulosclerosis and tubulointerstitial fibrosis.

Liver fibrosis occurs in different chronic lesions, including various viral hepatitis, alcohol abuse, drug causes, metabolic diseases, iron or copper overload, autoimmune attack on epithelial cells or congenital abnormalities of liver cells or bile ducts. Typically, the injury occurs months to years before scarring occurs. Liver fibrosis is reversible, while cirrhosis is the terminal result of fibrosis, usually irreversible.

Pulmonary fibrosis is a fibrotic pulmonary disease characterized by interstitial pneumonia, which is a long-term, progressive disease caused by unknown causes, and is manifested by symptoms such as dyspnea and shortness of breath. Of these, Idiopathic Pulmonary Fibrosis (IPF) is the most common and severe form of pulmonary fibrosis, and its main features are dry cough, dyspnea, decreased lung function, etc. The incidence of the disease is about 6.8 to 8.8 per 10 million persons, and the total prevalence is 14 to 27.9 per 10 million persons, as shown by data from 2012 in the united states. Through clinical research on patients, the IPF patients have the following characteristics: the incidence is high in men; the incidence of IPF increases with age, particularly after 50 years of age; IPF is more common in smokers, and is not improved even after smoking cessation; under CT the lungs of IPF patients will assume a honeycomb-like morphology. Although IPF is not generally prevalent, it is the most common and serious type of idiopathic interstitial pneumonia, and patients have a reduced quality of life, a low survival time and survival rate, and are difficult to diagnose accurately, which is an obstacle to the treatment of such diseases.

In recent years, there has been a shift in the study of the pathogenesis of IPF, from the inflammatory drive to epithelial diseases. It was previously classified as a chronic inflammatory disease, associated with the interaction of monocytes, fibroblasts and cytokines. IPF is a form of abnormal wound healing that involves fibrosis due to hyperproliferation of the interpulmonary spaces and alveolar spaces and myofibroblasts. That is, inflammation is not the primary cause of IPF, but it plays an important role in the pathogenesis and progression of the disease. Specific pathogenesis includes: first, the lungs are repeatedly attacked by micro-injury, such as smoking and viral infection on Alveolar Epithelial Cells (AECs). These injuries result in epithelial cell death and promote the wound healing process. Under normal conditions, apoptosis of epithelial cells initiates the wound healing process, increasing vascular permeability to proteins (fibrinogen and fibrin), and the formation of a wound clot. Damaged cells are removed by the action of inflammation, followed by the growth of fibroblasts to form a new extracellular matrix (ECM). After re-epithelialization, cells such as differentiated myofibroblasts, which participate in the wound healing process, undergo apoptosis. However, in IPF, the abnormal wound healing response leads to fibroblast proliferation and acute ECM deposition. This ultimately affects the balance between the fiber mediator and the anti-fibrotic mediator. Levels of active transforming growth factor (TGF-. beta.) are elevated in patients with IPF. When this abnormal process continues, repeated lung remodeling eventually leads to the formation of cellulite cysts and destruction of the lung structure, ultimately resulting in the development and loss of function of pulmonary fibrosis.

Cell-mediated pathways associated with fibrosis include: extracellular factors related to fibrosis are mainly growth factors and cytokines. These factors act on adjacent or distant cells through specific signaling pathways. They bind to specific receptors on the cell membrane, facilitating intracellular conduction of extracellular signals, ultimately leading to a pre-fibrotic cellular response. ② intracellular influencing factors, mainly involving a plurality of tyrosine kinases, which can regulate a plurality of intracellular different signal pathways through phosphorylation and dephosphorylation.

The only drugs currently approved by the FDA for IPF treatment are pirfenidone marketed in 2008 and nintedanib marketed in 2014. Nintedanib is a triple kinase inhibitor comprising: vascular Endothelial Growth Factor (VEGF); platelet growth factor (PDGF); fibroblast Growth Factor (FGFR). PDGFs can induce fibroblast chemotaxis and are the strongest stimulators of fibroblast proliferation. Therefore, they play an important role in the expansion of myofibroblasts and play a role in collagen synthesis. PDGF inhibitors like imatinib are reported to alter the fibrotic response, thereby reducing pulmonary fibrosis in animal studies. FGF channels are important signaling pathways that control angiogenesis, morphogenesis, and airway remodeling. VEGFR-2 antagonists may attenuate histopathological fibrosis and collagen deposition by modulating angiogenesis and inflammation. Thus, signaling of PDGF, FGF and VEGF has become a potential therapeutic target for IPF. Nintedanib inhibited significant anti-fibrotic effects by inhibiting the proliferation of primary human lung fibroblasts produced by IPF patients. However, nintedanib has poor selectivity and has certain inhibitory activity on other kinases, FLT3 and the like.

TGF-. beta.1-4 is a family of multifunctional cytokines that bind to TGF-. beta.receptors, consisting of TGF-. beta.R 1 and R2. TGF-beta R2, when bound to TGF-beta, phosphorylates TGF-beta R1 kinase, induces cascade amplification of signaling, recruits and activates S-mad protein. They are transcribed with nuclear downstream regulatory protein effectors, leading to differentiation, chemotaxis, proliferation and activation of target cells. TGF-. beta.1, as an inactive peptide, requires hydrolysis by proteases, including MMPs, to be activated. The key roles of TGF-beta 1 are: regulating the inflammatory process, the production of ECM, apoptosis and differentiation of T cells. During fibrosis, TGF- β signaling promotes differentiation of quiescent fibroblasts into myofibroblasts and secretion of ECM, the S-mad3 pathway upon which it depends is particularly important. Therefore, inhibition of TGF- β 1 binding to its receptor and related S-mad3 pathway signaling is another goal for anti-fibrosis. The anti-fibrosis property of pirfenidone is attributed to its ability to reduce the expression of cytokines such as TGF-beta, which ultimately leads to the inhibition of fibrosis.

Disclosure of Invention

The invention aims to provide coumarin derivatives and analogues or pharmaceutically acceptable salts thereof, wherein the structures of the coumarin derivatives and analogues are shown as a formula I:

x is selected from O or NH;

R1、R2、R3、R4、R5、R6independently selected from H, nitro, C1~C6Alkoxy or

Y is selected from O, S or NR12;R7、R8、R9、R10、R11Independently selected from H, halogen orR12Selected from H, C1~C6An alkyl group; r13Selected from NR14R15The number of the hetero atoms is 1-2; r14、R15Independently selected from H, C1~C6Alkyl, substituted or unsubstituted 5-6 membered heterocycle, heteroatom is N or O, the number of heteroatoms is 1-2.

As a preferred embodiment of the present invention, the above compound, R13Selected from NR14R15Unsubstituted 5-membered heterocycle, substituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, substituted 6-membered heterocycle; r14、R15Independently selected from H, C1~C6Alkyl, unsubstituted 5-membered heterocycle, substituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, substituted 6-membered heterocycle;

preferably, R13Selected from NR14R15Unsubstituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, C1~C4Alkyl-substituted 6-membered heterocycle, tert-butoxycarbonyl-substituted 6-membered heterocycle, -COR16Substituted 6-membered heterocyclic ring, -COOR17Substituted 6-membered heterocycles, -NR18R19A substituted 6-membered heterocyclic ring; r14、R15Independently selected from H, C1~C6Alkyl, unsubstituted 6-membered heterocycle, tert-butoxycarbonyl substituted 6-membered heterocycle, -COOR17A substituted 6-membered heterocyclic ring; r16Is selected from C1~C3Alkyl radical, C3~C6A cycloalkyl group; r17、R18、R19Independently selected from C1~C4An alkyl group;

further preferably, R13Selected from NR14R15Unsubstituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, C1~C4Alkyl-substituted 6-membered heterocycle, tert-butoxycarbonyl-substituted 6-membered heterocycle, -COR16Substituted 6-membered heterocyclic ring, -COOR17Substituted 6-membered heterocycles, -NR18R19A substituted 6-membered heterocyclic ring; r14、R15Independently selected from H, C1~C6Alkyl, unsubstituted 6-membered heterocycle, tert-butoxycarbonyl substituted 6-membered heterocycle, -COOR17A substituted 6-membered heterocyclic ring; r16Is selected from C1~C3Alkyl radical, C3~C6A cycloalkyl group; r17、R18、R19Independently selected from C1~C4An alkyl group; the 5-membered heterocycle has 1 heteroatom, and the heteroatom is N;

more preferably, R13Is selected from

Most preferably, R7、R8、R10、R11Independently selected from H or halogen; r9Is selected from

In a preferred embodiment of the present invention, when X is selected from O, the compound has a structural formula shown in formula ii below:

wherein Y is selected from O or NR12;R1、R3、R4、R5、R6Independently selected from H, nitro, C1~C6An alkoxy group; r7、R8、R10、R11Independently selected from H or halogen; r12Selected from H or methyl.

4. A compound according to claim 3, characterized in that: r1、R3、R4、R5、R6Independently selected from H, nitro, C1~C3An alkoxy group; preferably, R1、R3、R4、R5、R6Independently selected from H, nitro or methoxy; more preferably, R1、R3、R6Are all selected from H, R4、R5Independently selected from H, nitro or methoxy;

and/or, R7、R8、R10、R11Independently selected from H or F; preferably, R7、R8、R11Are all selected from H, R10Selected from H or F;

and/or, R13Is selected from

The compound has the following structural formula:

in a preferred embodiment of the present invention, in the above compound, when X is NH, Y is O, S or NR12,R2、R4、R5Independently selected from H orAnd R is2、R4、R5One of which isR1、R3、R6Independently selected from H, nitro, C1~C6An alkoxy group; r7、R8、R10、R11Independently selected from H or halogen; r12Selected from H or methyl.

Preferably, the above compound, R2Is selected fromWhen Y is selected from O, S or NR12;R4Or R5Is selected fromWhen, Y is selected from O;

and/or, R1、R3、R6Independently selected from H, nitro, C1~C3An alkoxy group; preferably, R1、R3、R6Are all selected from H;

and/or, R7、R8、R10、R11Independently selected from H or F; preferably, R7Selected from H or F, R8、R10、R11Are all selected from H;

and/or, R13Is selected from

The compound has the following structural formula:

the invention also provides a preparation method of the compound, which mainly adopts the following synthetic routes:

route (i):

reaction reagents and reaction conditions: (a) tetrabutylammonium bromide (TABA), P2O5Toluene (TOL), 90. + -. 5 ℃; (b) et (Et)3N, EtOH,70 +/-5 ℃, 2-5 h; (c) chloroacetyl chloride, Et3N, DMF, 0. + -. 5 ℃; (d) DMF, KI, room temperature.

Route (ii):

reaction reagents and reaction conditions: (a) TABA, P2O5,TOL,90±5℃;(b)K2CO3Acetone (ACE),65 ± 5 ℃; (c) fe, HCl, MeOH, H2O, room temperature; (d) chloroacetyl chloride, Et3N, DMF, 0. + -. 5 ℃; (e) DMF, KI, room temperature.

Route (iii):

reaction reagents and reaction conditions: (a) TABA, P2O5,TOL,90±5℃;(b)hydrobromic acid,1,4-dioxane,90±5℃;(c)aniline,Xantphos,Pd2(dba)3t-BuOK, dioxane, 130. + -. 5 ℃; (d) trifluoroacetic acid, rt, stirring overnight; (e) chloroacetyl chloride, Et3N,DMF,0-25℃;(f)RNH,Et3N, DMF, rt, stirred overnight.

Route (iv):

reaction reagents and reaction conditions: (a) POCl3,100±5℃;(b)hydrochloric acid,1,4-dioxane,90±5℃;(c)4-aminothiopenenol,K2CO3DMF, 130. + -. 5 ℃; (d) chloroacetyl chloride, Et3N,DMF,0±5℃,4h,0-25℃;(e)RNH2,Et3N, DMF, rt, stirred overnight.

Route (v):

reaction reagents and reaction conditions: (a) k2CO3,DMF,100±5℃;(b)Fe,HCl,MeOH/H2O-9/1, 85 ± 5 ℃; (c) chloroacetyl chloride, Et3N,DMF,0±5℃;(d)RNH2,Et3N, DMF, stirred at room temperature.

Route (six):

reaction reagents and reaction conditions: (a) k2CO3DMF, room temperature; (b) fe, HCl, MeOH/H2O-9/1, 85 ± 5 ℃; (c) chloroacetyl chloride, Et3N,DMF,0±5℃;(d)RNH2,Et3N, DMF, stirred at room temperature.

The method for synthesizing the compound 29-1 or the compound 29-2 is the same as that of the compound 29, except that the starting compound 10 is replaced with the compound 10-1 or the compound 10-2.

In all the above synthetic schemes, R13Selected from NR14R15The number of the hetero atoms is 1-2; r14、R15Independently selected from H, C1~C6Alkyl, substituted or unsubstituted 5-6 membered heterocycle, heteroatom is N or O, the number of heteroatoms is 1-2.

Preferably, R13Selected from NR14R15Unsubstituted 5-membered heterocycle, substituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, substituted 6-membered heterocycle; r14、R15Independently selected from H, C1~C6Alkyl, unsubstituted 5-membered heterocycle, substituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, substituted 6-membered heterocycle.

Further preferably, R13Selected from NR14R15Unsubstituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, C1~C4Alkyl-substituted 6-membered heterocycle, tert-butoxycarbonyl-substituted 6-membered heterocycle, -COR16Substituted 6-membered heterocyclic ring, -COOR17Substituted 6-membered heterocycles, -NR18R19A substituted 6-membered heterocyclic ring; r14、R15Independently selected from H, C1~C6Alkyl, unsubstituted 6-membered heterocycle, tert-butoxycarbonyl substituted 6-membered heterocycle, -COOR17A substituted 6-membered heterocyclic ring; r16Is selected from C1~C3Alkyl radical, C3~C6A cycloalkyl group; r17、R18、R19Independently selected from C1~C4An alkyl group.

More preferably, R13Selected from NR14R15Unsubstituted 5-membered heterocycle, unsubstituted 6-membered heterocycle, C1~C4Alkyl-substituted 6-membered heterocycle, tert-butoxycarbonyl-substituted 6-membered heterocycle, -COR16Substituted 6-membered heterocyclic ring, -COOR17Substituted 6-membered heterocycles, -NR18R19A substituted 6-membered heterocyclic ring; r14、R15Independently selected from H, C1~C6Alkyl, unsubstituted 6-membered heterocycle, tert-butoxycarbonyl substituted 6-membered heterocycle, -COOR17A substituted 6-membered heterocyclic ring; r16Is selected from C1~C3Alkyl radical, C3~C6A cycloalkyl group; r17、R18、R19Independently selected from C1~C4An alkyl group; the 5-membered heterocycle has 1 heteroatom, and the number of the heteroatoms is N.

Most preferably, R13Is selected from

The invention also provides a pharmaceutical composition which is prepared by taking the compound, the pharmaceutically acceptable salt or the pharmaceutically acceptable hydrate as an active ingredient and adding pharmaceutically acceptable auxiliary ingredients.

The invention also provides the application of the compound or the pharmaceutically acceptable salt thereof or the pharmaceutical composition in preparing a medicament for treating the fibrotic disease.

Further, the medicine is used for inhibiting the expression of COL1A1, alpha-SMA and p-Smad3 protein.

Further, the above use, the medicament is a medicament for inhibiting TGF-beta/Smad 3 pathway.

Further, in the above use, the fibrotic disease is pulmonary fibrosis, hepatic fibrosis or renal fibrosis.

Definition of terms:

the compounds and derivatives provided by the present invention may be named according to the IUPAC (international union of pure and applied chemistry) or CAS (chemical abstracts service, Columbus, OH) naming system.

The term "alkyl" is a radical of a straight or branched chain saturated hydrocarbon group. C1~C6Examples of alkyl groups include, but are not limited to, methyl (C)1) Ethyl (C)2) N-propyl (C)3) Isopropyl (C)3) N-butyl (C)4) Tert-butyl (C)4) Sec-butyl (C)4) Isobutyl (C)4) N-pentyl group (C)5) 3-pentyl radical (C)5) Pentyl group (C)5) Neopentyl (C)5) 3-methyl-2-butyl (C)5) Tert-amyl (C)5) And n-hexyl (C)6)。

The term "cycloalkyl" refers to a saturated cyclic hydrocarbon group, with or without heteroatoms, which may be a single ring structure or two or more rings, wherein the heteroatoms are selected from phosphorus, sulfur, oxygen and/or nitrogen.

The term "halogen" refers to fluorine (F), chlorine (Cl), bromine (Br), iodine (I).

The term "pharmaceutically acceptable" means that the carrier, cargo, diluent, adjuvant, and/or salt formed is generally chemically or physically compatible with the other ingredients comprising a pharmaceutical dosage form and physiologically compatible with the recipient.

The term "pharmaceutically acceptable salts" refers to acid and/or base salts of the compounds of the present invention with inorganic and/or organic acids and bases, and also includes zwitterionic salts (inner salts), and also includes quaternary ammonium salts, such as alkylammonium salts. These salts can be obtained directly in the final isolation and purification of the compounds. The compound may be obtained by appropriately (e.g., equivalent) mixing the above compound with a certain amount of an acid or a base. These salts may form precipitates in the solution which are collected by filtration, or they may be recovered after evaporation of the solvent, or they may be prepared by reaction in an aqueous medium followed by lyophilization. The salt in the invention can be hydrochloride, sulfate, citrate, benzene sulfonate, hydrobromide, hydrofluoride, phosphate, acetate, propionate, succinate, oxalate, malate, succinate, fumarate, maleate, tartrate or trifluoroacetate of the compound.

The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, parenteral (intravenous, intramuscular, or subcutaneous), and topical administration.

Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or solubilizers, for example, starch, lactose, sucrose, glucose, mannitol, and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.

Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.

In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.

Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.

Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.

Dosage forms for topical administration of the compounds of the present invention include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.

The pharmaceutically acceptable auxiliary components of the invention refer to substances contained in the dosage form in addition to the active ingredients, such as cyclodextrin, arginine or meglumine. The cyclodextrin is selected from alpha-cyclodextrin, beta-cyclodextrin, gamma-cyclodextrin and (C)1-4Alkyl) -alpha-cyclodextrin, (C)1-4Alkyl) -beta-cyclodextrin, (C)1-4Alkyl) -gamma-cyclodextrin, (hydroxy-C)1-4Alkyl radical) -alpha-cyclodextrin, (hydroxy-C)1-4Alkyl) -beta-cyclodextrin, (hydroxy-C)1-4Alkyl) -gamma-cyclodextrin, (carboxy-C)1-4Alkyl) -alpha-cyclodextrin, (carboxy-C)1-4Alkyl) -beta-cyclodextrin, (carboxy-C)1-4Alkyl) -gamma-cyclodextrin, saccharide ethers of alpha-cyclodextrin, saccharide ethers of beta-cyclodextrin, saccharide ethers of gamma-cyclodextrin, sulfobutyl ethers of alpha-cyclodextrin, sulfobutyl ethers of beta-cyclodextrin and sulfobutyl ethers of gamma-cyclodextrin. The auxiliary components also comprise a pharmaceutically acceptable carrier, adjuvant or vehicle. Can be used in pharmaceutically acceptable pharmaceutical composition, such as ion exchanger, aluminum oxide, aluminum stearate, and lecithin; buffer substances include phosphate, glycine, arginine, sorbic acid, and the like.

The invention provides a coumarin derivative with a novel structure. Biological experiments show that the compound has a strong in-vitro anti-fibrosis effect, can obviously reduce the deposition of intercellular collagen fibers on the NRK-49F cells induced by TGF-beta, and has an inhibitory effect on the migration of HUVEC cells. Meanwhile, the compounds with the structure have certain curative effect on mouse models of hepatic fibrosis induced by carbon tetrachloride and pulmonary fibrosis induced by bleomycin, have low toxicity, and provide a new choice for clinically treating fibrotic diseases including hepatic fibrosis and pulmonary fibrosis.

Drawings

FIG. 1 shows scratch test of compounds 21a, 25k, 29f, 29i, and 29 j;

FIG. 2 is a graph showing the inhibitory effect of Compound 9d on COL1A1, α -SMA, and p-Smad3 protein expression;

FIG. 3 is a graph of the inhibitory effect of Compound 29f on COL1A1, α -SMA, and p-Smad3 protein expression;

figure 4 is the in vivo effect of compound 9d in pulmonary fibrosis mice;

figure 5 is the in vivo effect of compound 29f in pulmonary fibrosis mice;

figure 6 is a graph of the in vivo effect of compound 9d in mice model of acute liver injury.

Detailed Description

The scheme of the invention will be explained with reference to the examples. It will be appreciated by those skilled in the art that the following examples are illustrative of the invention only and should not be taken as limiting the scope of the invention. The examples, where specific techniques or conditions are not indicated, are to be construed according to the techniques or conditions described in the literature in the art or according to the product specifications. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.

The abbreviations for the starting materials or reagents used in the examples are now set forth in the following tables.

Reagent Abbreviations Reagent Abbreviations
Tetrabutylammonium bromide TBAB Methylene dichloride DCM
Ethyl acetate EtOAc Tris (dibenzylidene-BASE acetone) dipalladium (0) Pd2(dba)3
Methanol MeOH 4, 5-bis-diphenylphosphino-9, 9-dimethylxanthene Xantphos
Ethanol EtOH Acetone (II) ACE
Isopropanol (I-propanol) IPA Toluene TOL
N, N-dimethylformamide DMF Reduced iron powder Fe
Phosphorus pentoxide P2O5 Water (W) H2O
Hydrochloric acid solution HCl Potassium iodide KI
Aqueous hydrobromic acid solution HBr Potassium tert-butoxide tBu-OK
Potassium carbonate K2CO3 Palladium on carbon (10%) Pd/C
Nitrogen gas N2 Nitrogen gas N2
Petroleum ether PE Hydrogen gas H2
Methane acyl chloride MsCl Phosphorus oxychloride POCl3
Triethylamine Et3N

Example 1

4-Hydroxycoumarin (4.86g, 3mol) was added to the reaction flask, 400ml of toluene was added as solvent, tetrabutylammonium bromide (TBAB, 14.49g,4.5mol) was added with stirring at room temperature, and the temperature was then slowly raised from room temperature to 90 ℃. At 90 ℃ adding P2O5(17.04g, 12mol) in three portions in one hourThe addition was continued for about 6-8 hours at 90 ℃ and the reaction was checked on a dot-plate. And (3) cooling the reaction liquid to room temperature, carrying out rotary evaporation to obtain a black viscous liquid, adding 1.2L of ice water and 400ml of ethyl acetate for extraction, combining three organic phases, and carrying out rotary drying. Adding n-heptane at room temperature, heating to 90 deg.C for dissolving completely, vacuum filtering to obtain yellowish liquid, cooling to room temperature to obtain yellow crystal, vacuum filtering, and vacuum drying to obtain yellow product with yield of 58%.

1H NMR(400MHz,DMSO-d6)δ:6.9(s,1H),7.29(m,1H),7.4(m,2H),7.80(m,1H).MS(ESI),m/z:224.94[M+H]+.

The following compound 2-1 was synthesized with reference to example 1, except that the starting material 4-hydroxycoumarin in example 1 was replaced with 4-hydroxy-7-methoxycoumarin.

1H NMR(400MHz,DMSO)δ7.72–7.67(m,1H),7.02(dd,J=7.5,2.2Hz,2H),6.87(s,1H),3.88(s,3H).MS(ESI),m/z:254.95[M+H]+.

The following compound 2-2 was synthesized with reference to example 1, except that the starting material 4-hydroxycoumarin in example 1 was replaced with 4-hydroxy-6-nitrocoumarin.

1H NMR(400MHz,DMSO-d6)δ:8.63(d,J=2.9Hz,1H),8.35(d,J=2.9Hz,1H),8.33(d,J=2.9Hz,1H),7.43(d,J=9.0Hz,1H).MS(ESI),m/z:269.93[M+H]+.

Example 2

4-Bromocoumarin (1g,3.93mmol) and p-diphenylamine (0.43g,3.93mmol) were placed at 50mAdding a proper amount of ethanol into an L-dry clean round-bottom flask as a reaction solvent, and then adding Et3Dropping N (1.1mL,7.88mmol) into a reaction bottle, lifting the reaction bottle into a heating reactor, stirring at 65 ℃ for 3h, lifting the reaction to room temperature, continuing stirring for two hours to precipitate a large amount of solid, filtering, and recrystallizing with ethanol to obtain the target product with the yield of 57 percent as a white powdery solid.

1H NMR(400MHz,DMSO-d6)δ:1H NMR(400MHz,DMSO)δ8.96(s,1H),8.11(d,J=9.0Hz,1H),6.96(m,3H),6.90(d,J=2.5Hz,1H),6.64(m,2H),5.21(s,2H),4.87(s,1H),3.86(s,3H).MS(ESI),m/z:253.09[M+H]+.

The following synthesis of compound 3-1 refers to example 2 except that the starting compound 2 in example 2 is replaced with compound 2-1.

1H NMR(400MHz,DMSO-d6)δ:1H NMR(400MHz,DMSO)δ8.96(s,1H),8.11(d,J=9.0Hz,1H),6.96(m,3H),6.90(d,J=2.5Hz,1H),6.64(m,2H),5.21(s,2H),4.87(s,1H),3.86(s,3H).MS(ESI),m/z:283.10[M+H]+.

The following synthesis of compound 3-2 refers to example 2 except that the starting compound 2 in example 2 is replaced with compound 2-2.

1H NMR(400MHz,DMSO-d6)δ:9.52(s,1H),9.28(d,J=2.1Hz,1H),8.45(dd,J=9.1,2.2Hz,1H),7.57(d,J=9.1Hz,1H),7.00(d,J=8.4Hz,2H),6.67(d,J=8.4Hz,2H),5.27(s,2H),5.12(s,1H).MS(ESI),m/z:298.08[M+H]+.

Synthesis of the following Compounds 3-3 reference example 2 except that the starting p-diphenylamine in example 2 was replaced with 2-fluoro-1, 4-phenylenediamine.

1H NMR(400MHz,DMSO-d6)δ:9.11(s,1H),8.19(d,J=7.8Hz,1H),7.64(t,J=7.6Hz,1H),7.37(dd,J=10.9,8.3Hz,2H),7.04(d,J=12.0Hz,1H),6.87(m,2H),5.28(s,2H),5.07(s,1H).MS(ESI),m/z:271.08[M+H]+.

Example 3

Placing 4-bromocoumarin (5g, 0.022mol), p-hydroxyphenol (4.65g, 0.033mol) and potassium carbonate (7.7g, 0.055mol) in a round-bottomed flask with a proper size, adding 300ml of acetone as a solvent, heating to 65 ℃, stirring, monitoring the reaction by TLC, removing the acetone by rotary evaporation, extracting by an EA/H2O system, combining EA layers, washing the EA layers by 10% NaOH for three times, drying by anhydrous Na2SO4, and carrying out rotary evaporation to obtain a solid, and recrystallizing by ethanol to finally obtain the target product with yield of 72% as a yellow powdery solid. 1H NMR (400MHz, DMSO-d6) δ 8.39(M,2H),7.90(d, J ═ 8.8Hz,1H),7.65(M,2H),7.11(d, J ═ 2.4Hz,1H),7.05(dd, J ═ 8.8,2.4Hz,1H),5.35(s,1H),3.90(s,3H) ms (esi), M/z 284.05[ M + H (esi) ], M/z]+.

The following compounds were synthesized with reference to example 3, except that the starting compound 2 in example 3 was replaced with compound 2-1.

1H NMR(400MHz,DMSO-d6)δ:8.39(m,2H),7.90(d,J=8.8Hz,1H),7.65(m,2H),7.11(d,J=2.4Hz,1H),7.05(dd,J=8.8,2.4Hz,1H),5.35(s,1H),3.90(s,3H).MS(ESI),m/z:314.06[M+H]+.

Example 4

Intermediate 6(3g, 0.01mol) was fully dispersed in 200ml of aqueous 10% methanol solution, then reduced iron powder was added at room temperature, concentrated hydrochloric acid (5ml) was added dropwise when the temperature was raised to 85 ℃, reflux was maintained at 85 ℃, after completion of the reaction monitoring by spotting, yellow filtrate was obtained by suction filtration while hot, and the solvent was removed by rotary evaporation. Then adding 50ml of water, pulping for 10 minutes, filtering to obtain a solid, and drying in vacuum to obtain a yellow solid.

1H NMR(400MHz,DMSO-d6)δ:8.01(d,J=7.0Hz,1H),7.74(dd,J=11.4,4.2Hz,1H),7.45(dd,J=12.7,7.9Hz,2H),6.98(d,J=8.7Hz,2H),6.70(m,2H),5.26(s,2H),5.18(s,1H).MS(ESI),m/z:254.07[M+H]+.

The following compounds were synthesized in accordance with example 4, and the compounds were substituted with the corresponding starting materials according to the structures of the compounds.

1H NMR(400MHz,DMSO-d6)δ:7.90(d,J=8.8Hz,1H),7.02(m,2H),7.01(m,2H),6.61(m,2H),5.24(s,2H),5.01(s,1H),3.95(m,3H).MS(ESI),m/z:284.09[M+H]+.

1H NMR(400MHz,DMSO)δ11.60(s,1H),7.95(d,J=7.7Hz,1H),7.60(t,J=7.4Hz,1H),7.36(q,J=8.8Hz,5H),7.25(t,J=7.3Hz,1H),5.33(s,1H).MS(ESI),m/z:253.09[M+H]+.

1H NMR(400MHz,DMSO)δ11.59(s,1H),7.97(d,J=7.3Hz,1H),7.60(s,1H),7.44–7.10(m,3H),6.67(dd,J=41.1,9.7Hz,2H),5.84(s,2H),5.33(s,1H).MS(ESI),m/z:271.08[M+H]+.

1H NMR(400MHz,DMSO)δ11.87(s,1H),7.88(d,J=9.5Hz,1H),7.45–7.35(m,2H),7.31(d,J=6.8Hz,2H),7.25–7.12(m,2H),6.52(d,J=9.4Hz,1H).MS(ESI),m/z:271.08[M+H]+.

1H NMR(400MHz,DMSO)δ11.59(s,1H),7.86(d,J=9.5Hz,1H),7.67(d,J=8.5Hz,1H),7.33(dd,J=15.3,8.0Hz,2H),7.14(d,J=7.4Hz,1H),6.91–6.77(m,2H),6.38(d,J=9.4Hz,1H).MS(ESI),m/z:271.08[M+H]+.

Example 5

4-hydroxy-2-quinolinone (8.05g, 5mol) was added to a reaction flask, 600ml of toluene was added as a solvent, tetrabutylammonium bromide (TBAB, 24.15g,7.5mol) was added with stirring at room temperature, and then the temperature was slowly raised to 90 ℃ from room temperature. At 90 ℃ adding P2O5(28.40g, 20mol) was added in five portions over an hour, and stirring was continued for about 6-8 hours at 90 ℃ with spotting to detect completion of the reaction. And (3) cooling the reaction liquid to room temperature, carrying out rotary evaporation to obtain a black viscous liquid, adding a proper amount of petroleum ether to fully dissolve the black viscous liquid, and carrying out suction filtration to obtain a light yellow filtrate and a brown solid. Adding a proper amount of petroleum ether to fully dissolve filter residues, combining the three filtrates, and carrying out spin drying and vacuum drying to obtain a yellow product with the yield of 40%.

1H NMR(400MHz,DMSO)δ7.82(dd,J=8.1,1.0Hz,1H),7.61(ddd,J=8.4,7.3,1.3Hz,1H),7.36(d,J=8.2Hz,1H),7.33–7.28(m,1H),7.03(d,J=1.5Hz,1H).MS(ESI),m/z:285.91[M+H]+.

Example 6

4-hydroxy-2-quinolinone (8.05g, 5mol) was added to a reaction flask, 200ml of phosphorus oxychloride was slowly added dropwise with stirring at room temperature, and a large amount of heat and gas were released during the dropwise addition. After the dropwise addition, the reaction solution is brown viscous, and then the temperature is slowly raised to 100 ℃ for reaction for about 6 hours. And (3) after the reaction is completely detected by a spot plate, slowly pouring the reaction liquid into a large amount of ice water to be fully cooled after the reaction liquid is cooled to room temperature, adding ethyl acetate for extraction, combining three organic phases, and spin-drying. Adding a proper amount of 60-80 mesh silica gel to mix the sample, and purifying the mixture by a quick column chromatography machine to obtain a light yellow product, wherein the yield is 53%.

1H NMR(400MHz,DMSO)δ7.78(dd,J=8.2,1.1Hz,1H),7.63(ddd,J=8.3,7.2,1.3Hz,1H),7.33(d,J=8.2Hz,1H),7.30–7.26(m,1H),7.08(d,J=1.5Hz,1H).MS(ESI),m/z:197.98[M+H]+.

Example 7

2, 4-dibromoquinoline (2.5g,8.77mmol) is dissolved in 60ml of 1, 4-dioxane solvent, the temperature is raised to 90 ℃, then 10ml of aqueous solution containing 40% HBr is added dropwise, the reaction is kept at 90 ℃ for about 6h, and after the reaction is monitored by a dot-plate, the solvent is removed by rotary evaporation. Then the appropriate amount of saturated NaHCO was added3In the aqueous solution, a large amount of bubbles are generated, simultaneously, a white solid is precipitated, and a white solid product is obtained by suction filtration and drying, wherein the yield is 95%.

1H NMR(400MHz,DMSO)δ12.06(s,1H),7.82(dd,J=8.1,1.0Hz,1H),7.61(ddd,J=8.4,7.3,1.3Hz,1H),7.36(d,J=8.2Hz,1H),7.33–7.28(m,1H),7.03(d,J=1.5Hz,1H).MS(ESI),m/z:223.96[M+H]+.

Synthesis of the following compounds with reference to example 7, HBr was replaced by concentrated HCl.

1H NMR(400MHz,DMSO)δ12.05(s,1H),7.82(dd,J=8.2,1.1Hz,1H),7.63(ddd,J=8.3,7.2,1.3Hz,1H),7.40(d,J=8.2Hz,1H),7.36–7.29(m,1H),7.11(d,J=1.5Hz,1H).MS(ESI),m/z:180.01[M+H]+.

Example 8

4-bromo-2-quinolinone (2.2g,9.8mmol), N-Boc-p-phenylenediamine (2.45g, 11.78mmol), potassium tert-butoxide (2.75g, 24.55mmol), Pd2(dba)3(0.90g, 0.99mmol) and Xantphos (1.41g, 2.45mmol) were dispersed well in 100ml of 1, 4-dioxane, heated from room temperature to 100 ℃ under nitrogen, and the temperature was maintained for an additional 12 hours, and the completion of the reaction was monitored on a dot-and-dash basis. And (3) carrying out suction filtration on the reaction solution to obtain brown filtrate, adding silica gel to mix with the sample, and purifying by using a rapid column chromatography to obtain a brown product with the yield of 60%.

1H NMR(400MHz,DMSO)δ10.95(s,1H),9.41(s,1H),8.52(s,1H),8.10(d,J=7.9Hz,1H),7.57–7.45(m,3H),7.29–7.23(m,1H),7.23–7.13(m,3H),5.46(d,J=0.9Hz,1H),1.49(s,9H).MS(ESI),m/z:423.18[M+H]+.MS(ESI),m/z:352.16[M+H]+.

Example 9

The compound (1.5g,4.27mmol) was placed in a 50ml round bottom flask and 20ml of trifluoroacetic acid (TFA) was added dropwise at room temperature followed by stirring at room temperature for about 4 hours and the reaction was monitored by dot-on-plate for completion. Then the trifluoroacetic acid is removed by rotary evaporation, and saturated NaHCO is added3And (3) dispersing the brown solid in water until no air bubbles are generated in the water solution, performing suction filtration and drying to obtain the brown solid with the yield of 93%.

1H NMR(400MHz,DMSO)δ10.82(s,1H),8.32(s,1H),8.09(d,J=7.8Hz,1H),7.46(dd,J=11.3,4.1Hz,1H),7.28–7.21(m,1H),7.18–7.08(m,1H),6.94(d,J=8.6Hz,2H),6.67–6.60(m,2H),5.25(s,1H),5.14(s,2H).MS(ESI),m/z:252.10[M+H]+.

Example 10

4-Aminobenzenethiol (7.0g, 0.06mol) and K2CO3(12.42g, 0.09mol) was placed in a round bottom flask, followed by addition of DMF (200 ml). After heating the reaction mixture to 130 ℃ 4-chloro-2-quinolinone (5.37g, 0.03mol) was added and stirred for a further 6 hours. After the reaction was monitored by TLC, the mixture was cooled to room temperature and 400ml of water was added to form a suspension mixture. The solid was obtained by filtration and after purification on silica gel column, a brown solid was obtained in 58% yield.

1H NMR(400MHz,DMSO)δ11.53(s,1H),7.82(d,J=7.6Hz,1H),7.59–7.52(m,1H),7.33(d,J=8.1Hz,1H),7.23(t,J=7.7Hz,3H),6.72(d,J=8.5Hz,2H),5.74(s,2H),5.56(s,1H).MS(ESI),m/z:269.06[M+H]+.

Example 11

4-hydroxy-2-quinolinone (9.66g, 0.06mol) and K2CO3(12.42g, 0.09mol) was placed in a round bottom flask, then DMF (200ml) was added and heated to 100 ℃. 1-fluoro-4-nitrobenzene (4.23g, 0.03mol) was dissolved in a further 20ml of DMF and the solution was added dropwise to the mixture prepared over a half hour. The mixture was kept stirring at 100 ℃ for about 6 hours, and after monitoring the reaction by TLC, the mixture was cooled to room temperature and 400ml of water was added to form a suspension mixture. The solids were then collected by a filter. After drying by vacuum oven, a pale yellow solid was obtained in 72% yield).

1H NMR(400MHz,DMSO)δ11.75(s,1H),8.37(d,J=7.4Hz,2H),7.87(d,J=6.6Hz,1H),7.62(s,1H),7.55(d,J=6.8Hz,2H),7.47–7.16(m,2H),5.72(s,1H).MS(ESI),m/z:283.06[M+H]+.

Example 12

4-hydroxy-2-quinolinone (9.66g, 0.06mol) and K2CO3(12.42g, 0.09mol) was placed in a round bottom flask and dispersed in DMF (200 ml). 3, 4-difluoro-nitrobenzene (4.77g, 0.03mol) was dissolved in a further 20ml of DMF and the solution was added dropwise to the previous mixture at room temperature with stirring. The mixture was kept stirring at room temperature for about 6 hours and after monitoring the reaction by TLC, 400ml of water was added to form a suspension mixture. The solids were then collected by a filter. After drying by vacuum oven, a pale yellow solid was obtained in 68% yield).

1H NMR(400MHz,DMSO)δ11.76(s,1H),8.45(dd,J=10.4,2.7Hz,1H),8.22(ddd,J=9.0,2.6,1.3Hz,1H),7.93(dd,J=8.0,1.0Hz,1H),7.81–7.72(m,1H),7.68–7.58(m,1H),7.40(d,J=8.2Hz,1H),7.32–7.23(m,1H),5.68(s,1H).MS(ESI),m/z:301.05[M+H]+.

Synthesis of the following Compounds reference example 12 is made, depending on the structure of the compound, to the corresponding starting materials

1H NMR(400MHz,DMSO)δ11.91(s,1H),8.34(dd,J=10.8,2.7Hz,1H),8.09–8.02(m,1H),7.88(d,J=9.6Hz,1H),7.56(d,J=1.2Hz,1H),7.43(d,J=2.3Hz,2H),7.13(t,J=8.7Hz,1H),6.56(d,J=9.6Hz,1H).MS(ESI),m/z:301.05[M+H]+.

1H NMR(400MHz,DMSO)δ11.73(s,1H),8.40(dd,J=10.7,2.7Hz,1H),8.13(ddd,J=9.1,2.6,1.3Hz,1H),7.92(d,J=9.6Hz,1H),7.76(d,J=8.5Hz,1H),7.39(t,J=8.6Hz,1H),7.04–6.97(m,2H),6.46(d,J=9.5Hz,1H).MS(ESI),m/z:301.05[M+H]+.

Example 13

Placing NaH (1.45g,60.2mmol) and 7.5mL diethyl carbonate into a round-bottom flask with proper size, moving the reaction bottle into a low-temperature reactor, stirring at 0 ℃, dissolving paeonol (2g,12.04mmol) into 10mL diethyl carbonate, placing the solution into a 25mL constant-pressure liquid funnel, slowly dripping the solution into the reaction bottle, keeping the temperature at 0 ℃ for continuous stirring for twenty minutes, moving the reaction bottle into a heating reactor, stirring at 100 ℃ for 3 hours, monitoring the reaction by TLC, stopping heating after the reaction is finished, placing the reaction bottle at room temperature, carefully dripping water to quench the reaction, washing off excessive diethyl carbonate by using a large amount of diethyl ether (3X 25mL), discarding an ether layer, combining water layers, acidifying the water layer to pH 3 by using 2N HCl (note that a large amount of emulsion is generated during the acidification process and a large amount of white solid is separated out, and the acidification treatment is carried out under the condition of stirring), filtration, washing of the filter cake with copious amounts of water, vacuum drying, to give the desired product in 69% yield as a white powdery solid.

1H NMR(400MHz,DMSO-d6)δ:12.37(s,1H),7.72(d,J=8.6Hz,1H),6.93(m,2H),5.47(s,1H),3.86(s,3H).MS(ESI),m/z:193.04[M+H]+.

Example 14

Putting sodium nitrate (0.52g,6.17mmol) into a round-bottom flask with a proper size, adding 20mL of concentrated sulfuric acid into a reaction bottle, lifting the reaction bottle into a low-temperature reactor, stirring at 0 ℃ for ten minutes, adding a compound 4-hydroxycoumarin (1g,6.17mmol) into the reaction bottle, keeping the temperature, continuing stirring for 1h, monitoring the reaction by TLC, after the reaction is finished, slowly adding crushed ice into the reaction bottle under low-temperature stirring until no excessive solid is separated out, filtering, washing a filter cake for a plurality of times by water, and drying in vacuum to obtain a target product, wherein the yield is 42%, and a white powdery solid is obtained.

1H NMR(400MHz,DMSO-d6)δ:8.53(d,J=2.6Hz,1H),8.45(dd,J=9.1,2.7Hz,1H),7.63(dd,J=17.8,8.2Hz,1H),5.71(s,1H).MS(ESI),m/z:208.02[M+H]+.

Example 15

Dissolving compound 3(2.82g,1.0mol) in 10mL of anhydrous DMF solution, placing in a low temperature reactor, stirring at 0 deg.C, and dropping Et3N (1.81ml,1.3mol), continuously stirring for ten minutes while keeping the temperature after dripping, then dripping chloroacetyl chloride (960ul,1.2mol), continuously stirring for 2 hours while keeping the temperature after dripping, extracting the reaction to room temperature, stirring for 2 hours at normal temperature, monitoring the reaction by TLC, after the reaction is finished, adding water to quench the reaction, simultaneously precipitating a large amount of solid, filtering, washing the filter cake with water, and drying in vacuum to obtain the target product, wherein the yield is 85 percent, and the white powdery solid is obtained.

1H NMR(400MHz,DMSO-d6)δ:9.06(s,1H),8.21(d,J=7.5Hz,1H),7.63(t,J=7.3Hz,1H),7.35(m,2H),6.99(d,J=8.5Hz,2H),6.66(d,J=8.5Hz,2H),5.23(s,2H),5.00(s,1H).MS(ESI),m/z:329.06[M+H]+.

The following compounds were synthesized in accordance with example 15, and the compounds were substituted with the corresponding starting materials according to the structures of the compounds.

1H NMR(400MHz,DMSO-d6)δ:10.42(s,1H),9.19(s,1H),8.14(d,J=9.0Hz,1H),7.70(d,J=8.8Hz,2H),7.33(d,J=8.8Hz,2H),6.99(dd,J=8.9,2.5Hz,1H),6.94(d,J=2.5Hz,1H),5.11(s,1H),4.27(s,2H),3.87(s,3H).MS(ESI),m/z:329.07[M+H]+.

1H NMR(400MHz,DMSO-d6)δ:10.17(s,1H),9.33(s,1H),8.21(d,J=7.6Hz,1H),7.96(t,J=8.7Hz,1H),7.67(t,J=7.4Hz,1H),7.39(m,3H),7.24(d,J=8.8Hz,1H),5.41(s,1H),4.37(s,2H).MS(ESI),m/z:347.05[M+H]+.

1H NMR(400MHz,DMSO-d6)δ:10.45(s,1H),9.74(s,1H),9.31(d,J=2.5Hz,1H),8.48(dd,J=9.1,2.6Hz,1H),7.73(d,J=8.8Hz,2H),7.61(d,J=9.1Hz,1H),7.37(d,J=8.8Hz,2H),5.35(s,1H),4.28(s,2H).MS(ESI),m/z:374.05[M+H]+.

1H NMR(400MHz,DMSO-d6)δ:10.59(s,1H),8.04(d,J=7.8Hz,1H),7.76(dd,J=16.1,8.5Hz,3H),7.47(m,2H),7.35(m,2H),5.22(s,1H),4.31(s,2H).MS(ESI),m/z:330.05[M+H]+.

1H NMR(400MHz,DMSO-d6)δ:10.49(s,1H),7.94(d,J=8.8Hz,1H),7.75(d,J=8.8Hz,2H),7.33(d,J=8.8Hz,2H),7.05(m,2H),5.05(s,1H),4.29(s,2H),3.89(s,3H).MS(ESI),m/z:360.06[M+H]+.

1H NMR(400MHz,DMSO)δ11.02(s,1H),10.89(s,1H),8.71(s,1H),8.19(d,J=8.1Hz,1H),7.72(d,J=8.6Hz,2H),7.50(t,J=7.6Hz,2H),7.33–7.27(m,3H),7.16(t,J=7.5Hz,1H),5.58(s,1H),4.35(s,2H).MS(ESI),m/z:328.07[M+H]+.

1H NMR(400MHz,DMSO)δ11.65(s,1H),10.76(s,1H),7.84(dd,J=8.3,3.2Hz,3H),7.67–7.54(m,3H),7.36(d,J=8.2Hz,1H),7.26(t,J=7.5Hz,1H),5.58(s,1H),4.34(s,2H).MS(ESI),m/z:345.04[M+H]+.

1H NMR(400MHz,DMSO)δ11.55(s,1H),10.47(s,1H),7.97(d,J=8.1Hz,1H),7.74(d,J=8.6Hz,2H),7.60(t,J=7.7Hz,1H),7.35(d,J=8.2Hz,1H),7.26(dd,J=14.6,8.0Hz,3H),5.32(s,1H),4.29(s,2H).MS(ESI),m/z:329.06[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),10.69(s,1H),7.92(dd,J=56.5,9.6Hz,2H),7.62(s,1H),7.47(s,2H),7.37(d,J=7.3Hz,1H),7.27(s,1H),5.35(s,1H),4.31(s,2H).MS(ESI),m/z:347.05[M+H]+.

1H NMR(400MHz,DMSO)δ11.80(s,1H),11.31(s,1H),7.83(dd,J=17.0,5.7Hz,2H),7.46(d,J=8.7Hz,1H),7.37(d,J=8.9Hz,1H),7.25(dd,J=8.9,2.5Hz,1H),7.18(dd,J=15.0,5.7Hz,2H),6.48(d,J=9.5Hz,1H),4.36(s,2H).MS(ESI),m/z:347.05[M+H]+.

1H NMR(400MHz,DMSO)δ11.50(s,1H),11.01(s,1H),7.84(s,2H),7.64(d,J=8.3Hz,1H),7.45(s,1H),7.33(d,J=8.3Hz,1H),6.80(dd,J=40.5,13.0Hz,2H),6.36(d,J=8.9Hz,1H),4.34(s,2H).MS(ESI),m/z:347.05[M+H]+.

Example 16

Compound 4(100mg, 0.3mmol) and KI (126mg, 0.75mmol) were dispersed in 5ml of anhydrous DMF and pyrrolidine (21mg, 0.45mmol) was added dropwise with stirring at room temperature, followed by stirring at room temperature overnight. Detecting the reaction completely by using a plate on the next day, adding 15ml of water while stirring, separating out a large amount of white solid, performing suction filtration to obtain a solid, and drying at 50 ℃ to obtain a white powder solid.

1H NMR(400MHz,DMSO-d6)δ:10.44(s,1H),9.28(s,1H),8.24(d,J=7.7Hz,1H),7.68(m,3H),7.38(m,4H),5.24(s,1H),4.28(s,2H).MS(ESI),m/z:329.06[M+H]+.

The following compounds were synthesized in accordance with example 16, by substituting the corresponding starting materials according to the structures of the compounds.

1H NMR(400MHz,DMSO)δ9.79(s,1H),9.27(s,1H),8.24(d,J=7.1Hz,1H),7.75(d,J=8.8Hz,2H),7.65(m,1H),7.39(dd,J=13.1,7.8Hz,2H),7.32(d,J=8.7Hz,2H),5.21(s,1H),3.09(s,2H),2.49(s,4H),1.58(d,J=4.8Hz,4H),1.41(s,2H).MS(ESI),m/z:378.19[M+H]+

1H NMR(400MHz,DMSO)δ9.87(s,1H),9.27(s,1H),8.24(d,J=7.3Hz,1H),7.75(d,J=8.8Hz,2H),7.66(m,1H),7.39(dd,J=13.1,7.8Hz,2H),7.32(d,J=8.8Hz,2H),5.21(s,1H),3.65(m,4H),3.15(s,2H),2.53(m,4H).MS(ESI),m/z:380.15[M+H]+

1H NMR(400MHz,DMSO)δ9.92(s,1H),9.41(d,J=6.5Hz,1H),8.32(d,J=8.0Hz,1H),7.77(dd,J=14.6,8.7Hz,2H),7.66(t,J=7.3Hz,1H),7.35(dd,J=12.7,10.5,5.9Hz,4H),5.21(s,1H),3.15(s,2H),2.55(s,4H),2.42(s,4H),2.20(s,3H).MS(ESI),m/z:393.18[M+H]+

1H NMR(400MHz,DMSO)δ9.78(s,1H),9.27(s,1H),8.24(d,J=7.2Hz,1H),7.76(d,J=8.8Hz,2H),7.70–7.63(m,1H),7.39(dd,J=13.1,7.8Hz,2H),7.32(d,J=8.8Hz,2H),5.22(s,1H),3.10(s,2H),2.79(t,J=8.8Hz,2H),2.08(td,J=10.7,3.7Hz,1H),1.85–1.52(m,6H),0.85(d,J=6.3Hz,4H).MS(ESI),m/z:392.19[M+H]+

1H NMR(400MHz,DMSO)δ9.84(s,1H),9.24(s,1H),8.18(d,J=8.9Hz,1H),7.81(d,J=8.7Hz,2H),7.29(d,J=8.7Hz,2H),6.99(dd,J=9.0,2.4Hz,1H),6.93(d,J=2.4Hz,1H),5.08(s,1H),4.01(d,J=5.9Hz,2H),3.87(s,3H),2.48(m,4H),1.24(m,4H).MS(ESI,m/z):394.10[M+H]+.

1H NMR(400MHz,DMSO)δ9.77(s,1H),9.17(s,1H),8.14(d,J=9.0Hz,1H),7.74(d,J=8.8Hz,2H),7.29(d,J=8.8Hz,2H),6.99(dd,J=8.9,2.5Hz,1H),6.94(d,J=2.5Hz,1H),5.08(s,1H),3.87(s,3H),3.08(s,2H),2.47(d,J=5.2Hz,4H),1.57(dd,J=10.9,5.6Hz,4H),1.41(d,J=4.9Hz,2H).MS(ESI,m/z):408.01[M+H]+.

1H NMR(400MHz,DMSO)δ9.85(s,1H),9.18(s,1H),8.14(d,J=9.0Hz,1H),7.74(d,J=8.8Hz,2H),7.30(d,J=8.8Hz,2H),6.99(dd,J=8.9,2.5Hz,1H),6.94(d,J=2.5Hz,1H),5.08(s,1H),3.87(s,3H),3.65(m,4H),3.15(s,2H),2.53(d,J=4.6Hz,4H).MS(ESI,m/z):410.10[M+H]+.

1H NMR(400MHz,DMSO)δ9.80(s,1H),9.17(s,1H),8.14(d,J=9.0Hz,1H),7.73(d,J=8.8Hz,2H),7.29(d,J=8.8Hz,2H),6.99(dd,J=9.0,2.5Hz,1H),6.94(d,J=2.5Hz,1H),5.08(s,1H),3.87(s,3H),3.13(s,2H),2.37(d,J=24.4Hz,8H),2.19(s,3H).MS(ESI,m/z):423.20[M+H]+.

1H NMR(400MHz,DMSO)δ9.77(s,1H),9.17(s,1H),8.14(d,J=9.0Hz,1H),7.74(d,J=8.8Hz,2H),7.29(d,J=8.8Hz,2H),6.99(dd,J=8.9,2.5Hz,1H),6.94(d,J=2.5Hz,1H),5.08(s,1H),3.87(s,3H),3.09(s,2H),2.79(s,2H),2.07(d,J=10.7,3.7Hz,2H),1.69(m,5H),0.85(d,J=6.3Hz,3H).MS(ESI,m/z):422.20[M+H]+.

1H NMR(400MHz,DMSO)δ9.81(s,1H),9.74(s,1H),9.32(d,J=2.5Hz,1H),8.48(dd,J=9.1,2.5Hz,1H),7.77(d,J=8.8Hz,2H),7.62(t,J=9.9Hz,1H),7.33(d,J=8.8Hz,2H),5.34(d,J=7.2Hz,1H),3.10(s,2H),2.79(t,J=8.7Hz,2H),2.08(td,J=10.7,3.6Hz,1H),1.67(m,6H),0.86(d,J=6.3Hz,3H).MS(ESI),m/z:437.17[M+H]+

1H NMR(400MHz,DMSO)δ9.85(s,1H),8.36(s,1H),8.30(d,J=9.8Hz,1H),7.52(d,J=8.7Hz,2H),7.37(t,J=8.2Hz,1H),7.08(d,J=8.7Hz,2H),6.89(d,J=8.2Hz,1H),6.77(d,J=8.1Hz,1H),3.31(s,2H),2.05(m,4H),1.22(m,4H),0.85(d,J=6.9Hz,2H).MS(ESI),m/z:423.16[M+H]+

1H NMR(400MHz,DMSO)δ9.83(s,1H),9.72(s,1H),9.31(d,J=2.2Hz,1H),8.47(dd,J=9.1,2.4Hz,1H),7.76(d,J=8.7Hz,2H),7.58(t,J=16.5Hz,1H),7.33(d,J=8.7Hz,2H),5.33(s,1H),3.12(d,J=15.0Hz,2H),2.54(s,4H),2.40(s,4H),2.19(s,3H).MS(ESI),m/z:438.17[M+H]+

1H NMR(400MHz,DMSO)δ9.89(s,1H),9.73(s,1H),9.32(d,J=2.5Hz,1H),8.48(dd,J=9.1,2.6Hz,1H),7.77(d,J=8.8Hz,2H),7.61(d,J=9.1Hz,1H),7.33(d,J=8.8Hz,2H),5.33(s,1H),3.65(m,4H),3.16(s,2H),2.51(s,4H).MS(ESI),m/z:425.14[M+H]+

1H NMR(400MHz,DMSO)δ9.84(s,1H),9.72(s,1H),9.31(d,J=2.5Hz,1H),8.48(dd,J=9.1,2.5Hz,1H),7.78(d,J=8.8Hz,2H),7.61(d,J=9.1Hz,1H),7.32(d,J=8.8Hz,2H),5.32(s,1H),3.27(d,J=8.6Hz,2H),2.64(d,J=26.1Hz,4H),1.76(t,J=3.4Hz,4H).MS(ESI),m/z:409.14[M+H]+

1H NMR(400MHz,DMSO)δ9.68(s,1H),9.32(s,1H),8.21(d,J=7.2Hz,1H),8.12(t,J=8.8Hz,1H),7.67(t,J=7.8Hz,1H),7.38(m,3H),7.23(d,J=8.7Hz,1H),5.36(s,1H),3.13(s,2H),2.50(s,4H),1.58(m,4H),1.43(d,J=5.0Hz,2H).MS(ESI),m/z:382.15[M+H]+

1H NMR(400MHz,DMSO)δ9.68(s,1H),9.32(s,1H),8.21(d,J=7.2Hz,1H),8.12(t,J=8.8Hz,1H),7.67(t,J=7.8Hz,1H),7.38(m,3H),7.23(d,J=8.7Hz,1H),5.36(s,1H),3.13(s,2H),2.50(s,4H),1.58(m,4H),1.43(d,J=5.0Hz,2H).MS(ESI),m/z:396.16[M+H]+

1H NMR(400MHz,DMSO)δ9.66(s,1H),9.33(s,1H),8.21(d,J=7.6Hz,1H),8.04(t,J=8.7Hz,1H),7.67(t,J=7.5Hz,1H),7.39(m,3H),7.23(d,J=8.4Hz,1H),5.37(s,1H),3.65(s,4H),3.20(s,2H),2.56(s,4H).MS(ESI),m/z:398.14[M+H]+

1H NMR(400MHz,DMSO)δ9.64(s,1H),9.32(s,1H),8.21(d,J=7.2Hz,1H),8.11(t,J=8.8Hz,1H),7.67(t,J=7.8Hz,1H),7.38(m,3H),7.23(d,J=8.7Hz,1H),5.36(s,1H),3.17(d,J=10.5Hz,2H),2.57(s,4H),2.39(s,4H),2.19(s,3H).MS(ESI),m/z:411.18[M+H]+

1H NMR(400MHz,DMSO)δ9.66(s,1H),9.32(s,1H),8.16(m,2H),7.67(t,J=7.6Hz,1H),7.39(dt,J=20.4,10.1Hz,3H),7.22(d,J=8.4Hz,1H),5.36(s,1H),3.13(s,2H),2.80(d,J=9.0Hz,2H),2.01(m,3H),1.61(d,J=45.4Hz,4H),0.87(d,J=6.1Hz,3H).MS(ESI),m/z:410.18[M+H]+

1H NMR(400MHz,DMSO)δ8.11(d,J=8.3Hz,2H),7.58(m,2H),7.30(m,4H),5.12(s,1H),3.66(t,J=6.3Hz,4H),3.31(s,2H),1.95(m,4H).MS(ESI),m/z:365.14[M+H]+.

1H NMR(400MHz,DMSO)δ9.86(s,1H),8.05(dd,J=7.9,1.3Hz,1H),7.77(m,3H),7.48(dd,J=13.7,7.6Hz,2H),7.38(d,J=8.8Hz,2H),5.20(s,1H),4.13(s,2H),3.45(s,2H),3.07(s,3H),1.77(s,5H).13CNMR(101MHz,DMSO):δ168.26,166.50,162.78,154.20,151.61,133.90,126.92,123.02,121.20,117.08,115.21,98.09,63.46,51.10,25.12,24.54.MS(ESI),m/z:379.16[M+H]+

1H NMR(400MHz,DMSO)δ9.94(s,1H),8.04(dd,J=7.9,1.3Hz,1H),7.82(d,J=9.0Hz,2H),7.76(m,1H),7.47(dd,J=14.0,7.5Hz,2H),7.32(d,J=9.0Hz,2H),5.20(s,1H),3.65(m,4H),3.16(s,2H),2.53(d,J=4.6Hz,4H).MS(ESI),m/z:381.14[M+H]+.

1H NMR(400MHz,DMSO)δ9.89(s,1H),8.04(m,1H),7.77(m,3H),7.47(dd,J=14.2,7.6Hz,2H),7.31(d,J=8.9Hz,2H),5.21(s,1H),3.14(s,2H),2.60(d,J=58.5Hz,4H),2.39(s,4H),2.18(s,3H).MS(ESI),m/z:394.17[M+H]+.

1H NMR(400MHz,DMSO)δ9.94(s,1H),8.03(dd,J=7.9,1.3Hz,1H),7.72(d,J=9.0Hz,2H),7.65(m,1H),7.47(dd,J=14.0,7.5Hz,2H),7.32(d,J=9.0Hz,2H),5.20(s,1H),3.16(s,2H),2.80(d,J=9.0Hz,2H),2.53(d,J=4.6Hz,4H),2.01(m,3H),1.61(d,J=45.4Hz,4H),0.87(d,J=6.1Hz,3H).MS(ESI),m/z:393.17[M+H]+.

1H NMR(400MHz,DMSO)δ:9.93(s,1H),7.94(d,J=8.8Hz,1H),7.80(d,J=8.9Hz,2H),7.29(d,J=8.9Hz,2H),7.05(m,2H),5.04(s,1H),3.89(s,3H),3.27(d,J=8.6Hz,2H),2.64(d,J=26.1Hz,4H),1.76(t,J=3.4Hz,4H).MS(ESI),m/z:395.15[M+H]+.

1H NMR(400MHz,DMSO)δ:9.02(s,1H),7.88(d,J=8.8Hz,1H),7.76(d,J=8.9Hz,2H),7.49(d,J=8.9Hz,2H),7.05(m,2H),5.02(s,1H),3.89(s,3H),3.45(s,2H),3.07(s,3H),1.77(s,5H).MS(ESI),m/z:409.17[M+H]+.

1H NMR(400MHz,DMSO)δ:9.93(s,1H),7.94(d,J=8.8Hz,1H),7.80(d,J=8.9Hz,2H),7.29(d,J=8.9Hz,2H),7.05(m,2H),5.04(s,1H),3.89(s,3H),3.62(m,8H),3.14(d,J=15.3Hz,2H).MS(ESI),m/z:411.15[M+H]+.

1H NMR(400MHz,DMSO-d6)δ:10.03(s,1H),7.98(d,J=8.8Hz,1H),7.72(d,J=8.9Hz,2H),7.39(d,J=8.9Hz,2H),7.05(m,2H),5.02(s,1H),3.89(s,3H),2.60(d,J=58.5Hz,4H),2.39(s,4H),2.18(s,3H).MS(ESI),m/z:424.18[M+H]+.

1H NMR(400MHz,DMSO)δ:9.93(s,1H),7.94(d,J=8.8Hz,1H),7.80(d,J=8.9Hz,2H),7.29(d,J=8.9Hz,2H),7.05(m,2H),5.04(s,1H),3.89(s,3H),3.62(m,9H),3.14(d,J=15.3Hz,3H).MS(ESI),m/z:423.18[M+H]+.

1H NMR(400MHz,DMSO)δ10.96(s,1H),9.74(s,1H),8.55(s,1H),8.10(d,J=7.8Hz,1H),7.70(d,J=8.8Hz,2H),7.49(t,J=7.3Hz,1H),7.26(t,J=8.8Hz,3H),7.17(t,J=7.6Hz,1H),5.54(s,1H),3.25(s,2H),2.60(d,J=5.3Hz,4H),1.80–1.72(m,4H).MS(ESI),m/z:363.18[M+H]+.

1H NMR(400MHz,DMSO)δ10.96(s,1H),9.72(s,1H),8.55(s,1H),8.10(d,J=7.9Hz,1H),7.68(d,J=8.8Hz,2H),7.49(t,J=7.2Hz,1H),7.26(dd,J=8.1,6.2Hz,3H),7.17(t,J=7.6Hz,1H),5.54(d,J=1.3Hz,1H),3.12(s,2H),2.51(s,4H),2.36(d,J=24.3Hz,4H),2.18(s,3H).MS(ESI),m/z:392.22[M+H]+.

1H NMR(400MHz,DMSO)δ10.96(s,1H),9.87(s,1H),8.55(s,1H),8.10(d,J=7.8Hz,1H),7.69(d,J=8.8Hz,2H),7.49(t,J=7.2Hz,1H),7.26(dd,J=7.9,5.2Hz,3H),7.17(t,J=7.6Hz,1H),5.54(s,1H),3.84(dt,J=11.4,3.4Hz,2H),3.33(s,2H),3.28(dd,J=11.5,2.1Hz,2H),2.68–2.58(m,1H),1.77(d,J=12.5Hz,2H),1.31(ddd,J=14.5,11.5,3.7Hz,2H).MS(ESI),m/z:393.19[M+H]+.

1H NMR(400MHz,DMSO)δ10.97(s,1H),9.71(s,1H),8.56(s,1H),8.11(d,J=7.8Hz,1H),7.69(d,J=8.8Hz,2H),7.49(dd,J=11.3,4.1Hz,1H),7.27(t,J=7.6Hz,3H),7.20–7.13(m,1H),5.55(s,1H),3.07(s,2H),2.50(td,J=3.9,2.1Hz,4H),1.62–1.53(m,4H),1.45–1.37(m,2H).MS(ESI),m/z:377.20[M+H]+.

1H NMR(400MHz,DMSO)δ10.98(s,1H),9.82(s,1H),8.57(s,1H),8.11(d,J=7.7Hz,1H),7.70(d,J=8.8Hz,2H),7.55–7.44(m,1H),7.27(dd,J=7.9,5.2Hz,3H),7.20–7.13(m,1H),5.55(s,1H),3.50(dd,J=10.1,5.8Hz,4H),3.18(s,2H),2.58–2.52(m,2H),2.50–2.46(m,2H),2.00(s,3H).MS(ESI),m/z:420.20[M+H]+.

1H NMR(400MHz,DMSO)δ11.00(s,1H),9.93(s,1H),8.65(s,1H),8.16(d,J=8.1Hz,1H),7.71(d,J=8.8Hz,2H),7.50(t,J=7.7Hz,1H),7.28(dd,J=11.4,8.6Hz,3H),7.17(t,J=7.3Hz,1H),5.56(s,1H),3.39(s,4H),3.19(s,2H),2.51–2.47(m,4H),1.41(s,9H).MS(ESI),m/z:478.24[M+H]+.

1H NMR(400MHz,MeOD)δ8.13–8.08(m,1H),7.69(d,J=8.8Hz,2H),7.58(t,J=7.7Hz,1H),7.41–7.26(m,4H),5.86(s,1H),3.49(s,2H),2.88(q,J=7.2Hz,4H),1.20(t,J=7.2Hz,6H).MS(ESI),m/z:365.20[M+H]+.

1H NMR(400MHz,DMSO)δ10.98(s,1H),9.71(s,1H),8.57(s,1H),8.12(d,J=8.0Hz,1H),7.70(d,J=8.7Hz,2H),7.50(t,J=7.4Hz,1H),7.27(t,J=8.1Hz,3H),7.18(t,J=7.5Hz,1H),5.56(s,1H),3.10(s,2H),2.85(d,J=11.4Hz,2H),2.14(t,J=10.7Hz,2H),1.60(d,J=11.4Hz,2H),1.33–1.21(m,3H),0.92(d,J=6.0Hz,3H).MS(ESI),m/z:391.21[M+H]+.

1H NMR(400MHz,DMSO)δ10.97(s,1H),9.74(s,1H),8.56(s,1H),8.11(d,J=8.0Hz,1H),7.69(d,J=8.5Hz,2H),7.50(t,J=7.4Hz,1H),7.27(t,J=7.4Hz,3H),7.18(t,J=7.4Hz,1H),5.55(s,1H),3.13(s,2H),2.52(s,4H),2.45(s,4H),2.34(dd,J=14.1,7.0Hz,2H),1.00(t,J=7.1Hz,3H).MS(ESI),m/z:406.22[M+H]+.

1H NMR(400MHz,MeOD)δ8.08(d,J=7.9Hz,1H),7.68(d,J=8.8Hz,2H),7.57(t,J=7.2Hz,1H),7.40–7.26(m,5H),5.86(s,1H),3.82–3.74(m,4H),3.20(s,2H),2.65–2.58(m,4H).MS(ESI),m/z:379.18[M+H]+.

1H NMR(400MHz,DMSO)δ10.96(s,1H),9.78(s,1H),8.57(s,1H),8.12(d,J=8.0Hz,1H),7.70(d,J=8.7Hz,2H),7.50(t,J=7.6Hz,1H),7.27(t,J=7.6Hz,3H),7.18(t,J=7.6Hz,1H),5.55(s,1H),3.16(s,2H),3.02–2.95(m,2H),2.87–2.77(m,1H),2.57(s,6H),2.22(t,J=11.2Hz,2H),1.89(d,J=11.7Hz,2H),1.73–1.60(m,2H).MS(ESI),m/z:420.23[M+H]+.

1H NMR(400MHz,DMSO)δ10.98(s,1H),10.35(s,1H),8.58(s,1H),8.11(d,J=8.1Hz,1H),7.66(d,J=8.7Hz,2H),7.51(t,J=7.6Hz,1H),7.29(t,J=9.0Hz,3H),7.18(t,J=7.6Hz,1H),5.58(s,1H),3.80(s,2H),2.94(q,J=7.1Hz,2H),1.18(dd,J=9.0,5.4Hz,3H).MS(ESI),m/z:337.16[M+H]+.

1H NMR(400MHz,DMSO)δ11.70(s,1H),10.53(s,1H),7.96(dd,J=19.8,5.3Hz,2H),7.62(d,J=7.1Hz,2H),7.43(t,J=8.7Hz,2H),7.27(s,1H),5.34(s,1H),3.44(s,2H),2.70(s,4H),1.78(s,4H).MS(ESI),m/z:382.16[M+H]+.

1H NMR(400MHz,DMSO)δ11.67(s,1H),10.28(s,1H),7.96(dd,J=19.0,10.6Hz,2H),7.60(dd,J=16.8,8.6Hz,2H),7.42(dd,J=16.5,8.4Hz,2H),7.27(t,J=7.5Hz,1H),5.34(s,1H),3.19(s,2H),2.55(s,4H),2.44(s,4H),2.21(s,3H).MS(ESI),m/z:411.18[M+H]+.

1H NMR(400MHz,DMSO)δ11.61(s,1H),10.26(s,1H),7.97(dd,J=9.0,6.3Hz,2H),7.62(dd,J=12.0,4.8Hz,2H),7.41(q,J=8.9Hz,2H),7.27(t,J=8.0Hz,1H),5.34(s,1H),3.13(s,2H),2.49–2.42(m,4H),1.61–1.52(m,4H),1.40(d,J=4.8Hz,2H).MS(ESI),m/z:396.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.69(s,1H),10.46(s,1H),7.97(t,J=10.6Hz,2H),7.62(s,2H),7.47–7.39(m,2H),7.27(t,J=7.5Hz,1H),5.34(s,1H),3.50(d,J=3.8Hz,4H),3.26(s,2H),2.56(s,2H),2.50–2.46(m,2H),2.00(s,3H).MS(ESI),m/z:439.18[M+H]+.

1H NMR(400MHz,DMSO)δ11.70(s,1H),10.42(s,1H),8.02–7.92(m,2H),7.66–7.57(m,2H),7.43(t,J=9.0Hz,2H),7.30–7.23(m,1H),5.34(s,1H),3.36(s,4H),3.23(s,2H),2.63(s,6H),1.06(d,J=5.1Hz,3H).MS(ESI),m/z:425.20[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),10.08(s,1H),7.95(dd,J=30.0,10.2Hz,2H),7.58(dd,J=20.2,7.5Hz,2H),7.49–7.19(m,3H),5.34(s,1H),3.40(s,4H),3.20(s,2H),2.50(s,4H),1.41(s,9H).MS(ESI),m/z:497.22[M+H]+.

1H NMR(400MHz,DMSO)δ11.67(s,1H),10.39(s,1H),8.02–7.92(m,2H),7.66–7.57(m,2H),7.43(dd,J=17.8,8.7Hz,2H),7.27(dd,J=11.2,4.1Hz,1H),5.34(s,1H),3.24(s,2H),3.00(s,3H),2.66(s,6H),2.24(t,J=11.4Hz,2H),2.00(d,J=10.9Hz,2H),1.77(dd,J=11.9,3.2Hz,2H).MS(ESI),m/z:439.21[M+H]+.

1H NMR(400MHz,DMSO)δ11.67(s,1H),10.31(s,1H),7.97(ddd,J=15.4,10.6,1.6Hz,2H),7.66–7.55(m,2H),7.42(dd,J=18.4,9.1Hz,2H),7.31–7.21(m,1H),5.34(s,1H),3.70–3.58(m,4H),3.20(s,2H),2.57–2.52(m,4H).MS(ESI),m/z:398.15[M+H]+.

1H NMR(400MHz,DMSO)δ11.67(s,1H),10.32(s,1H),7.97(ddd,J=15.5,10.6,1.6Hz,2H),7.67–7.58(m,2H),7.48–7.37(m,2H),7.27(dd,J=11.3,4.0Hz,1H),5.35(s,1H),2.70(d,J=5.8Hz,4H),1.06(t,J=7.1Hz,6H).MS(ESI),m/z:384.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.66(s,1H),10.36(s,1H),8.04–7.89(m,2H),7.61(dd,J=14.2,7.3Hz,2H),7.42(dd,J=20.1,8.7Hz,2H),7.27(t,J=7.6Hz,1H),5.34(s,1H),3.17(d,J=4.2Hz,2H),2.92(s,2H),2.26(s,2H),1.61(d,J=11.8Hz,2H),1.30(dd,J=21.9,10.5Hz,3H),0.92(d,J=6.0Hz,3H).MS(ESI),m/z:410.19[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),10.16(s,1H),7.99(dd,J=8.0,1.0Hz,1H),7.93(dd,J=13.1,2.3Hz,1H),7.65–7.59(m,1H),7.54(dd,J=8.9,1.4Hz,1H),7.44(t,J=8.9Hz,1H),7.37(d,J=8.1Hz,1H),7.27(dd,J=11.6,4.5Hz,1H),5.34(s,1H),3.84(dt,J=11.4,3.4Hz,2H),3.33(s,2H),3.28(dd,J=11.5,2.0Hz,3H),2.64(ddd,J=14.3,10.2,4.0Hz,1H),1.77(dd,J=12.5,1.7Hz,2H),1.35–1.25(m,2H).MS(ESI),m/z:412.19[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),7.99(d,J=7.3Hz,1H),7.92(dd,J=13.1,2.3Hz,1H),7.66–7.59(m,1H),7.54(dd,J=8.9,1.4Hz,1H),7.44(t,J=8.9Hz,1H),7.37(d,J=8.2Hz,1H),7.27(t,J=7.4Hz,1H),5.34(s,1H),3.33(s,3H),2.60(q,J=7.1Hz,2H),1.06(t,J=7.1Hz,3H).MS(ESI),m/z:356.14[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),9.99(s,1H),7.90–7.82(m,3H),7.59(ddd,J=10.9,6.2,1.6Hz,3H),7.35(d,J=7.7Hz,1H),7.29–7.24(m,1H),5.58(s,1H),3.12(s,2H),2.50–2.45(m,4H),1.58(dt,J=11.0,5.7Hz,4H),1.42(d,J=5.0Hz,2H).MS(ESI),m/z:394.16[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),9.98(s,1H),7.86(dd,J=10.6,8.0Hz,3H),7.62–7.55(m,3H),7.35(d,J=7.7Hz,1H),7.29–7.23(m,1H),5.57(d,J=1.1Hz,1H),3.13(s,2H),2.85(d,J=11.6Hz,2H),2.14(t,J=10.4Hz,2H),1.60(d,J=10.2Hz,2H),1.36–1.21(m,3H),0.92(d,J=6.1Hz,3H).MS(ESI),m/z:408.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.62(s,1H),10.04(s,1H),7.87(dd,J=15.0,8.2Hz,3H),7.58(t,J=7.8Hz,3H),7.35(d,J=8.0Hz,1H),7.27(d,J=8.0Hz,1H),5.57(s,1H),3.30(s,2H),2.61(s,4H),1.77(dd,J=6.5,3.2Hz,4H).MS(ESI),m/z:380.14[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),10.09(s,1H),7.86(dd,J=14.6,8.3Hz,3H),7.64–7.55(m,3H),7.35(d,J=8.2Hz,1H),7.25(dd,J=11.3,4.1Hz,1H),5.57(s,1H),3.51(dd,J=10.1,6.2Hz,4H),3.23(s,2H),2.59–2.53(m,2H),2.49(d,J=9.3Hz,2H),2.00(s,3H).MS(ESI),m/z:437.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.67(s,1H),10.19(s,1H),7.86(dd,J=18.9,8.2Hz,3H),7.58(t,J=8.3Hz,3H),7.38(d,J=8.2Hz,1H),7.25(t,J=7.6Hz,1H),5.57(s,1H),3.17(d,J=7.6Hz,2H),2.54(s,4H),2.38(s,4H),2.18(s,3H).MS(ESI),m/z:409.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.59(s,1H),10.03(s,1H),7.82(dd,J=11.4,8.6Hz,3H),7.55(dd,J=12.6,8.0Hz,3H),7.31(d,J=8.2Hz,1H),7.22(t,J=7.6Hz,1H),5.53(s,1H),3.73–3.53(m,4H),3.15(s,2H),2.47(s,4H).MS(ESI),m/z:396.14[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),9.95(s,1H),7.87(dd,J=18.0,8.4Hz,3H),7.63–7.54(m,3H),7.35(d,J=8.2Hz,1H),7.30–7.21(m,1H),5.58(s,1H),3.21(s,2H),2.63(q,J=7.1Hz,4H),1.04(t,J=7.1Hz,6H).MS(ESI),m/z:382.16[M+H]+.

1H NMR(400MHz,DMSO)δ11.64(s,1H),10.06(s,1H),7.87(dd,J=12.9,8.5Hz,3H),7.64–7.54(m,3H),7.36(d,J=8.2Hz,1H),7.26(t,J=7.5Hz,1H),5.57(s,1H),3.18(s,2H),2.96(d,J=11.2Hz,2H),2.52(s,1H),2.37(s,6H),2.19(t,J=11.2Hz,2H),1.82(d,J=11.0Hz,2H),1.66–1.52(m,2H).MS(ESI),m/z:437.20[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),10.07(s,1H),7.86(dd,J=10.9,8.7Hz,3H),7.59(dd,J=12.5,8.0Hz,3H),7.35(d,J=8.2Hz,1H),7.26(t,J=7.6Hz,1H),5.57(s,1H),3.40(m,4H),3.21(s,2H),2.49(m,4H),1.41(s,9H).MS(ESI),m/z:495.21[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),10.02(s,1H),7.86(t,J=8.5Hz,3H),7.65–7.52(m,3H),7.35(d,J=8.1Hz,1H),7.26(t,J=7.6Hz,1H),5.57(s,1H),3.17(s,2H),2.55(s,4H),2.44(s,4H),2.34(q,J=7.1Hz,2H),1.00(t,J=7.2Hz,3H).MS(ESI),m/z:423.18[M+H]+.

1H NMR(400MHz,DMSO)δ11.62(s,1H),10.03(s,1H),7.86(t,J=9.1Hz,3H),7.67–7.54(m,3H),7.35(d,J=8.1Hz,1H),7.25(dd,J=11.3,4.1Hz,1H),5.57(s,1H),3.33(s,2H),2.60(q,J=7.1Hz,2H),1.06(t,J=7.1Hz,3H).MS(ESI),m/z:354.13[M+H]+.

1H NMR(400MHz,DMSO)δ11.63(s,1H),10.16(s,1H),7.85(dd,J=13.4,4.8Hz,3H),7.68–7.51(m,3H),7.35(d,J=7.8Hz,1H),7.30–7.21(m,1H),5.57(s,1H),3.84(dt,J=11.4,3.4Hz,2H),3.38(s,2H),3.28(m,3H),2.64(ddd,J=14.3,10.2,4.0Hz,1H),1.82–1.74(m,2H),1.36–1.23(m,2H).MS(ESI),m/z:410.15[M+H]+.

1H NMR(400MHz,DMSO)δ11.54(s,1H),9.83(s,1H),7.98(d,J=7.9Hz,1H),7.80(d,J=8.9Hz,2H),7.60(t,J=7.7Hz,1H),7.36(d,J=8.2Hz,1H),7.25(t,J=7.3Hz,3H),5.32(s,1H),3.10(s,2H),2.48(s,4H),1.64–1.54(m,4H),1.42(d,J=4.8Hz,2H).MS(ESI),m/z:378.18[M+H]+.

1H NMR(400MHz,DMSO)δ11.55(s,1H),9.82(s,1H),7.97(d,J=7.8Hz,1H),7.80(d,J=8.8Hz,2H),7.60(t,J=7.4Hz,1H),7.36(d,J=8.2Hz,1H),7.25(t,J=7.1Hz,3H),5.32(s,1H),3.11(s,2H),2.85(d,J=11.4Hz,2H),2.13(t,J=11.0Hz,2H),1.59(d,J=11.1Hz,2H),1.36–1.21(m,3H),0.91(d,J=5.9Hz,3H).MS(ESI),m/z:392.20[M+H]+.

1H NMR(400MHz,DMSO)δ11.54(s,1H),9.89(s,1H),7.97(d,J=7.3Hz,1H),7.78(d,J=8.9Hz,2H),7.64–7.56(m,1H),7.36(d,J=8.2Hz,1H),7.26(dd,J=7.5,4.9Hz,3H),5.31(s,1H),3.19(s,2H),2.63(s,8H),2.35(s,3H).MS(ESI),m/z:393.19[M+H]+.

1H NMR(400MHz,DMSO)δ11.55(s,1H),9.96(s,1H),7.97(d,J=7.8Hz,1H),7.78(d,J=8.9Hz,2H),7.61(t,J=7.7Hz,1H),7.36(d,J=8.2Hz,1H),7.26(t,J=7.9Hz,3H),5.30(s,1H),3.31(s,2H),3.02(m,6H),2.83(m,4H),1.20(t,J=7.1Hz,3H).MS(ESI),m/z:421.20[M+H]+.

1H NMR(400MHz,DMSO)δ11.54(s,1H),9.87(s,1H),7.98(d,J=7.9Hz,1H),7.81(d,J=8.8Hz,2H),7.60(t,J=7.6Hz,1H),7.36(d,J=8.2Hz,1H),7.25(t,J=8.8Hz,3H),5.31(s,1H),3.27(s,2H),2.61(s,4H),1.76(s,4H).MS(ESI),m/z:364.16[M+H]+.

1H NMR(400MHz,DMSO)δ11.54(s,1H),9.90(s,1H),7.97(d,J=8.5Hz,1H),7.79(d,J=8.9Hz,2H),7.62–7.56(m,1H),7.35(d,J=8.2Hz,1H),7.25(dd,J=8.1,4.1Hz,3H),5.30(s,1H),3.39(m,4H),3.18(s,2H),2.49(m,4H),1.40(s,9H).MS(ESI),m/z:479.23[M+H]+.

1H NMR(400MHz,DMSO)δ11.54(s,1H),9.84(s,1H),7.97(d,J=8.0Hz,1H),7.79(d,J=8.8Hz,2H),7.60(t,J=7.6Hz,1H),7.35(d,J=8.2Hz,1H),7.25(t,J=7.2Hz,3H),5.31(s,1H),3.12(s,2H),2.92(d,J=11.5Hz,2H),2.51(m,1H),2.26(s,6H),2.16(t,J=11.2Hz,3H),1.76(d,J=11.7Hz,2H),1.54(dd,J=20.4,11.4Hz,2H).MS(ESI),m/z:421.22[M+H]+.

1H NMR(400MHz,DMSO)δ11.54(s,1H),9.98(s,1H),7.97(d,J=7.5Hz,1H),7.78(d,J=8.9Hz,2H),7.59(t,J=7.2Hz,1H),7.35(d,J=8.2Hz,1H),7.25(dd,J=7.8,5.5Hz,3H),5.31(s,1H),3.84(d,J=11.3Hz,2H),3.34(d,J=6.9Hz,2H),3.30–3.24(m,2H),2.64(m,J=10.3,5.1Hz,1H),1.77(d,J=11.4Hz,2H),1.31(dd,J=19.3,11.2Hz,2H).MS(ESI),m/z:407.21[M+H]+.

1H NMR(400MHz,DMSO)δ11.53(s,1H),9.89(s,1H),7.97(d,J=8.0Hz,1H),7.78(d,J=8.9Hz,2H),7.59(t,J=7.7Hz,1H),7.35(d,J=8.2Hz,1H),7.25(dd,J=7.6,5.1Hz,3H),5.30(s,1H),3.69–3.61(t,4H),3.15(s,2H),2.53(t,J=4.4Hz,4H).MS(ESI),m/z:380.16[M+H]+.

1H NMR(400MHz,DMSO)δ11.54(s,1H),9.92(s,1H),7.97(d,J=7.7Hz,1H),7.79(d,J=8.9Hz,2H),7.60(t,J=7.2Hz,1H),7.35(d,J=8.2Hz,1H),7.24(dd,J=7.9,4.3Hz,3H),5.31(s,1H),3.51(d,J=4.1Hz,4H),3.20(s,2H),2.57–2.53(m,2H),2.48(d,J=4.8Hz,2H),2.00(s,3H).MS(ESI),m/z:447.19[M+H]+.

1H NMR(400MHz,DMSO)δ11.54(s,1H),9.79(s,1H),7.97(d,J=7.3Hz,1H),7.80(d,J=8.9Hz,2H),7.62–7.57(m,1H),7.35(d,J=8.2Hz,1H),7.29–7.19(m,3H),5.31(s,1H),3.18(s,2H),2.62(q,J=7.1Hz,4H),1.04(t,J=7.1Hz,6H).MS(ESI),m/z:366.18[M+H]+.

1H NMR(400MHz,DMSO)δ11.56(s,1H),10.45(s,1H),7.97(d,J=7.3Hz,1H),7.76(d,J=8.9Hz,2H),7.68–7.55(m,1H),7.36(d,J=8.2Hz,1H),7.32–7.21(m,3H),5.31(s,1H),3.77(s,2H),2.91(q,J=7.2Hz,2H),1.18(t,J=7.2Hz,3H).MS(ESI),m/z:338.15[M+H]+.

1H NMR(400MHz,DMSO)δ10.04(s,1H),7.98(d,J=8.0Hz,1H),7.91(d,J=13.0Hz,1H),7.65–7.51(m,2H),7.47–7.33(m,2H),7.26(t,J=7.6Hz,1H),5.34(s,1H),3.13(s,2H),2.79(s,4H),2.46(s,4H).MS(ESI),m/z:397.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.68(s,1H),10.32(s,1H),7.96(t,J=12.0Hz,2H),7.59(d,J=7.9Hz,2H),7.41(d,J=6.6Hz,2H),7.26(t,J=7.5Hz,1H),5.33(s,1H),3.15(s,2H),2.50(m,8H),1.00(s,9H).MS(ESI),m/z:453.23[M+H]+.

1H NMR(400MHz,DMSO)δ11.70(s,1H),10.53(s,1H),8.02–7.94(m,2H),7.62(t,J=8.0Hz,2H),7.44(t,J=8.7Hz,2H),7.27(t,J=7.4Hz,1H),5.34(s,1H),3.74(s,2H),3.53(s,2H),3.27(s,2H),2.59(s,4H),1.98(ms,1H),0.71(m,J=8.5Hz,4H).MS(ESI),m/z:465.19[M+H]+.

1H NMR(400MHz,DMSO)δ11.74(s,1H),9.97(s,1H),7.84(d,J=8.7Hz,2H),7.51–7.03(m,5H),6.49(d,J=9.0Hz,1H),3.11(s,2H),2.83(d,J=8.2Hz,2H),2.13(s,2H),1.58(d,J=10.7Hz,2H),1.46–1.16(m,3H),0.91(s,3H).MS(ESI),m/z:410.19[M+H]+.

1H NMR(400MHz,DMSO)δ11.73(s,1H),9.98(s,1H),7.83(t,J=10.4Hz,2H),7.42(d,J=8.6Hz,1H),7.35–7.22(m,2H),7.17(dd,J=17.8,8.7Hz,2H),6.49(d,J=9.5Hz,1H),3.38(s,4H),3.17(s,2H),2.48(d,J=4.9Hz,4H),1.40(s,9H).MS(ESI),m/z:497.23[M+H]+.

1H NMR(400MHz,DMSO)δ11.73(s,1H),9.99(s,1H),7.88–7.79(m,2H),7.43(d,J=8.9Hz,1H),7.32(d,J=8.9Hz,1H),7.26(dd,J=8.9,2.6Hz,1H),7.22–7.13(m,2H),6.50(d,J=9.6Hz,1H),3.74(s,2H),3.54(s,2H),3.20(s,2H),2.56(s,2H),2.50(s,2H),1.97(m,J=12.6,7.7,4.8Hz,1H),0.77–0.69(m,4H).MS(ESI),m/z:465.19[M+H]+.

1H NMR(400MHz,DMSO)δ11.72(s,1H),9.93(s,1H),7.82(dd,J=18.7,5.9Hz,2H),7.41(d,J=9.0Hz,1H),7.31(d,J=8.9Hz,1H),7.25(dd,J=8.9,2.6Hz,1H),7.21–7.11(m,2H),6.49(d,J=9.6Hz,1H),3.14(s,2H),2.54(m,4H),2.42(m,4H),2.25(s,3H).MS(ESI),m/z:411.10[M+H]+.

1H NMR(400MHz,DMSO)δ11.72(s,1H),9.87(s,1H),7.88–7.78(m,2H),7.43(d,J=8.9Hz,1H),7.36–7.23(m,2H),7.22–7.11(m,2H),6.49(d,J=9.6Hz,1H),3.07(s,2H),2.45(s,4H),1.62–1.51(m,4H),1.40(d,J=4.9Hz,2H).MS(ESI),m/z:396.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.72(s,1H),9.96(s,1H),7.82(dd,J=16.8,5.9Hz,2H),7.42(d,J=8.8Hz,1H),7.35–7.22(m,2H),7.22–7.11(m,2H),6.49(d,J=9.6Hz,1H),3.70–3.59(m,4H),3.14(s,2H),2.50(d,J=1.1Hz,4H).MS(ESI),m/z:398.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.73(s,1H),9.98(s,1H),7.82(dd,J=16.0,5.8Hz,2H),7.43(d,J=8.9Hz,1H),7.35–7.23(m,2H),7.23–7.13(m,2H),6.50(d,J=9.6Hz,1H),3.50(d,J=4.3Hz,4H),3.18(s,2H),2.53(d,J=4.6Hz,2H),2.48–2.44(m,2H),2.00(s,3H).MS(ESI),m/z:439.18[M+H]+.

1H NMR(400MHz,DMSO)δ11.72(s,1H),9.85(s,1H),7.89–7.79(m,2H),7.46(d,J=8.9Hz,1H),7.31(d,J=8.9Hz,1H),7.25(dd,J=8.9,2.6Hz,1H),7.22–7.11(m,2H),6.49(d,J=9.6Hz,1H),3.17(s,2H),2.60(q,J=7.1Hz,4H),1.02(t,J=7.1Hz,6H).MS(ESI),m/z:384.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.72(s,1H),9.95(s,1H),7.90–7.77(m,2H),7.44(d,J=8.8Hz,1H),7.36–7.22(m,2H),7.22–7.11(m,2H),6.49(d,J=9.6Hz,1H),3.27(s,2H),2.60(s,4H),1.76(s,4H).MS(ESI),m/z:382.15[M+H]+.

1H NMR(400MHz,DMSO)δ11.73(s,1H),9.95(s,1H),7.82(t,J=12.2Hz,2H),7.48–7.05(m,5H),6.50(d,J=9.4Hz,1H),3.18(s,2H),2.97(d,J=10.6Hz,3H),2.77–2.54(m,6H),2.23(t,J=11.2Hz,2H),1.90(d,J=9.1Hz,2H),1.66(d,J=10.5Hz,2H).MS(ESI),m/z:439.21[M+H]+.

1H NMR(400MHz,DMSO)δ11.74(s,1H),10.72(s,1H),7.90–7.69(m,2H),7.36–7.15(m,5H),6.51(d,J=9.5Hz,1H),3.31(s,2H),3.03(dd,J=14.4,7.1Hz,2H),1.25–1.14(t,3H).MS(ESI),m/z:356.14[M+H]+.

1H NMR(400MHz,DMSO)δ11.73(s,1H),10.00(s,1H),7.82(t,J=12.0Hz,2H),7.47–7.08(m,5H),6.50(d,J=9.6Hz,1H),3.48(s,2H),3.32(s,2H),3.09(s,4H),2.70(s,2H),1.34(s,9H).MS(ESI),m/z:453.23[M+H]+.

1H NMR(400MHz,DMSO)δ11.48(s,1H),9.94(s,1H),7.87(dd,J=17.8,5.9Hz,2H),7.64(d,J=8.7Hz,1H),7.51(d,J=8.8Hz,1H),7.29(t,J=9.0Hz,1H),6.85(dd,J=8.6,2.4Hz,1H),6.76(d,J=2.1Hz,1H),6.36(d,J=9.5Hz,1H),3.11(s,2H),2.82(d,J=11.4Hz,2H),2.12(t,J=11.2Hz,2H),1.59(d,J=11.2Hz,2H),1.42–1.19(m,3H),0.91(d,J=6.0Hz,3H).MS(ESI),m/z:410.19[M+H]+.

1H NMR(400MHz,DMSO)δ11.49(s,1H),10.04(s,1H),7.91–7.80(m,2H),7.64(d,J=8.7Hz,1H),7.50(d,J=8.8Hz,1H),7.30(t,J=9.0Hz,1H),6.84(dd,J=8.6,2.4Hz,1H),6.76(d,J=2.1Hz,1H),6.36(d,J=9.5Hz,1H),3.70–3.61(m,4H),2.51(s,4H).MS(ESI),m/z:498.15[M+H]+.

1H NMR(400MHz,DMSO)δ11.49(s,1H),10.07(s,1H),7.92–7.78(m,2H),7.65(d,J=8.6Hz,1H),7.46(d,J=8.6Hz,1H),7.31(t,J=9.0Hz,1H),6.91–6.70(m,2H),6.36(d,J=9.5Hz,1H),3.31(s,2H),3.17(s,4H),2.77-2.45(d,J=20.0Hz,7H).MS(ESI),m/z:411.18[M+H]+.

1H NMR(400MHz,DMSO)δ11.48(s,1H),10.05(s,1H),7.91–7.81(m,2H),7.63(t,J=9.9Hz,1H),7.49(d,J=8.8Hz,1H),7.30(t,J=9.0Hz,1H),6.83(dt,J=16.9,8.5Hz,1H),6.76(d,J=2.2Hz,1H),6.36(d,J=9.5Hz,1H),3.58–3.45(m,4H),3.20(s,2H),2.53(dd,J=8.6,3.8Hz,2H),2.49–2.44(m,2H),2.01(d,J=10.8Hz,3H).MS(ESI),m/z:439.18[M+H]+.

1H NMR(400MHz,DMSO)δ11.48(s,1H),10.03(s,1H),7.86(dd,J=11.5,5.6Hz,2H),7.64(d,J=8.6Hz,1H),7.49(d,J=8.9Hz,1H),7.30(t,J=9.0Hz,1H),6.84(dd,J=8.6,2.1Hz,1H),6.76(s,1H),6.36(d,J=9.5Hz,1H),3.37(d,J=13.9Hz,4H),3.19(s,2H),2.48(s,3H),1.40(s,9H).MS(ESI),m/z:497.22[M+H]+.

1H NMR(400MHz,DMSO)δ11.49(s,1H),9.98(d,J=24.3Hz,1H),7.92–7.74(m,2H),7.62(t,J=13.5Hz,1H),7.51(d,J=8.6Hz,1H),7.28(t,J=8.9Hz,1H),6.84(d,J=6.8Hz,1H),6.76(s,1H),6.35(d,J=9.4Hz,1H),3.27(s,2H),2.59(s,4H),1.76(s,4H).MS(ESI),m/z:482.16[M+H]+.

1H NMR(400MHz,DMSO)δ11.48(s,1H),9.95(s,1H),7.86(dd,J=19.2,5.9Hz,2H),7.64(d,J=8.7Hz,1H),7.50(dd,J=8.8,1.0Hz,1H),7.29(t,J=9.0Hz,1H),6.84(dd,J=8.6,2.4Hz,1H),6.76(d,J=2.1Hz,1H),6.36(d,J=9.5Hz,1H),3.09(s,2H),2.46(s,4H),1.64–1.50(m,4H),1.40(d,J=4.2Hz,2H).MS(ESI),m/z:396.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.50(s,1H),10.05(s,1H),7.89–7.80(m,2H),7.65(d,J=8.7Hz,1H),7.48(d,J=8.8Hz,1H),7.31(t,J=9.0Hz,1H),6.83(dd,J=8.6,2.4Hz,1H),6.77(d,J=2.1Hz,1H),6.36(d,J=9.5Hz,1H),3.59–3.39(m,2H),3.32(s,2H),3.11(d,J=39.2Hz,4H),2.70(s,2H),1.32(s,9H).MS(ESI),m/z:453.23[M+H]+.

1H NMR(400MHz,DMSO)δ11.48(s,1H),10.06(s,1H),7.91–7.81(m,2H),7.64(d,J=8.7Hz,1H),7.50(d,J=8.8Hz,1H),7.30(t,J=9.0Hz,1H),6.84(dd,J=8.6,2.3Hz,1H),6.76(d,J=2.1Hz,1H),6.36(d,J=9.5Hz,1H),3.74(s,2H),3.54(s,2H),3.21(s,2H),2.57(s,2H),2.50–2.45(m,2H),2.03–1.91(m,1H),0.78–0.64(m,4H).MS(ESI),m/z:465.19[M+H]+.

1H NMR(400MHz,DMSO)δ11.48(s,1H),10.02(s,1H),7.94–7.76(m,2H),7.65(d,J=8.6Hz,1H),7.48(d,J=8.6Hz,1H),7.30(t,J=9.0Hz,1H),6.84(dd,J=8.6,2.2Hz,1H),6.76(s,1H),6.36(d,J=9.5Hz,1H),3.20(s,2H),2.99(d,J=11.3Hz,3H),2.71(d,J=12.9Hz,6H),2.24(m,J=11.3Hz,2H),1.92(d,J=11.0Hz,2H),1.68(m,J=20.6,10.2Hz,2H).MS(ESI),m/z:439.21[M+H]+.

1H NMR(400MHz,DMSO)δ11.53–11.42(m,1H),9.98–9.87(m,1H),7.95–7.76(m,2H),7.68–7.61(m,1H),7.58–7.50(m,1H),7.32–7.24(m,1H),6.89–6.83(m,1H),6.79–6.71(m,1H),6.39–6.31(m,1H),3.18(s,2H),2.67–2.56(m,4H),1.04(d,J=7.1Hz,6H).MS(ESI),m/z:384.17[M+H]+.

1H NMR(400MHz,DMSO)δ11.50(s,1H),7.83(dd,J=17.3,12.1Hz,2H),7.65(d,J=8.7Hz,1H),7.42(d,J=8.7Hz,1H),7.33(t,J=8.9Hz,1H),6.84(dd,J=8.6,2.0Hz,1H),6.77(s,1H),6.36(d,J=9.5Hz,1H),3.73(s,2H),2.87(q,J=7.3Hz,2H),1.16(t,J=7.3Hz,3H).MS(ESI),m/z:354.14[M+H]+.

1H NMR(400MHz,MeOD)δ8.11(d,J=7.3Hz,1H),7.77(d,J=8.9Hz,2H),7.67–7.60(m,1H),7.39(d,J=8.2Hz,1H),7.34(t,J=7.7Hz,1H),7.23(d,J=9.0Hz,2H),5.57(s,1H),3.30(s,2H),3.16(s,4H),2.86(s,4H),1.32(s,9H).MS(ESI),m/z:379.16[M+H]+.

1H NMR(400MHz,DMSO)δ11.54(s,1H),9.98(s,1H),7.97(d,J=7.5Hz,1H),7.78(d,J=8.9Hz,2H),7.59(t,J=7.2Hz,1H),7.35(d,J=8.2Hz,1H),7.25(dd,J=7.8,5.5Hz,3H),5.31(s,1H),3.84(d,J=11.3Hz,2H),3.34(d,J=6.9Hz,2H),3.30–3.24(m,2H),2.64(m,J=10.3,5.1Hz,1H),1.77(d,J=11.4Hz,2H),1.31(dd,J=19.3,11.2Hz,2H).MS(ESI),m/z:394.17[M+H]+.

Pharmacodynamic test section

The following representative experiments, without limitation, were used to analyze the biological activity of the compounds of the present invention

1. In vitro Activity

Excess extracellular matrix (ECM) is composed mainly of collagen and is a pathological marker of fibrotic diseases, and therefore detection of collagen synthesis is an effective index for evaluating fibrotic diseases. According to our previous studies and other reports, rat fibroblasts induced by TGF-. beta.s (NRK-49F) produce a number of extracellular collagens with characteristics similar to fibrosis in vivo. It has therefore been considered as an effective, convenient in vitro anti-fibrotic screening model.

1.1 anti-fibrotic Activity

NRK-49 cells were cultured in 1640 medium containing 10% FBS, 50U/ml penicillin and 50. mu.g/ml streptomycin and cultured in a 5% CO2 incubator at 37 ℃ for passaging. NRK-49F cells are spread on a 96-well plate (104 cells/well), cultured for 3d in a 1640+ 5% FBS culture medium, the cell supernatant is removed, cultured for 2d in a 1640+ 1% ITS culture medium, the cell supernatant is removed, and cultured for 2d in a 1640+ 1% ITS culture medium containing TGF-beta (5ng/ml) and 10 mu M of a compound to be screened. Removing cell supernatant, adding 100 μ l/well 4% paraformaldehyde for fixing for 30min, washing with PBS twice, incubating at room temperature for 4h with 100 μ l/well 0.1% PSR staining solution, removing staining solution, washing with 100 μ l/well 0.1% acetic acid for 3 times, air drying, and taking pictures under microscope. Adding 100 μ l/well 0.1M NaOH, shaking at room temperature for 30min to dissolve precipitate, measuring OD value of each well at 540nm wavelength of microplate reader, and screening out compounds with collagen deposition inhibiting effect. Total collagen accumulation inhibition ═ 100% (administration a value-control a value)/(model a value-control a value). All assays were repeated three times. Nintedanib as a positive drug.

1.2 cytotoxicity

NRK-49F cells were cultured at 0.5X 105Individual cells/ml were plated in 96-well plates and incubated overnight in 100. mu.l suspension for experimentsThe group was added with the drug at a concentration of 10. mu.M. After 72 hours, 20. mu.l of 5% (m/v) MTT solution was added to each well and incubated for 4 hours in an incubator. Then 150. mu.l DMSO was added to each well. Finally, the absorbance (a value) of each well was measured on a microplate reader. Survival rate-dosing value-zero a value)/(blank a value-zero a value) × 100%. All assays were repeated three times. Nintedanib as a positive drug.

1.3 collagen deposition inhibition and cytotoxicity test results

TABLE 1 collagen inhibition and cytotoxicity of NH-4 substituted coumarin parent nucleus compounds

TABLE 2 collagen inhibition and cytotoxicity of O-4 substituted coumarin parent nucleus compounds

TABLE 3 collagen inhibition and cytotoxicity of NH-4 and S-4 substituted 2-quinolinone core Compounds

TABLE 4 collagen inhibition and cytotoxicity of O-4 substituted 2-quinolinone core Compounds

TABLE 5 collagen inhibition and cytotoxicity of O-6-and O-7-substituted 2-quinolinone core Compounds

1.4 scratch test and results thereof

The occurrence and the exacerbation of inflammation are often accompanied by phenomena such as vascular proliferation and the like caused by the migration of fibroblasts, immune cells and the like and the healing of tissues. We selected several compounds with good collagen deposition inhibition and lower toxicity on TGF- β induced NRK-49F cells, followed by co-incubation with HUVEC cells, and investigated their effect on human venous endothelial cell migration capacity after 0h, 12h, 24 h.

The experiment comprises the following specific steps: 1. firstly, a marker pen is used at the back of the six-hole plate, 3 transverse lines are homogenized by a ruler and transversely penetrate through the holes; 2. cells grown in log phase were plated with complete medium at 5 × 10 per well6Individual HUVEC cells; 3. the next day after the cells were confluent, they were removed with sterilized 200uL tipsThe scratch of a transverse line at the back is measured to be vertical, and the gun head is vertical and cannot be inclined; 4. washing the cells with PBS for 3 times, washing off the scraped cells, and changing to a serum-free culture medium; 5. the blank group is not treated, the induction group induces cell migration by using TGF-beta of 10ng/ml, the administration group is treated by adding TGF-beta of 10ng/ml and adding a compound with gradient concentration, and a photograph represents 0 h; 6. put in 37 ℃ 5% CO2The incubator is used for culturing, and the samples are photographed for 12 and 24 hours.

As shown in fig. 1, compounds 21a, 25k, 29f were most inhibitory to migration of HUVEC cell de1, suggesting that these compounds may be inhibitory to excessive tissue healing and repair during fibrosis.

1.5 inhibition of COL1A1, alpha-SMA, and p-Smad3 protein expression by Compounds

Collagen type I α 1(COL1a1), α -smooth muscle actin (α -SMA), is overexpressed in fibrotic diseases, which is generally considered to be a fibrotic marker. In order to further study the anti-fibrotic activity of compounds with higher collagen inhibition rates, their ability to inhibit COL1a1, α -SMA protein expression in vitro was studied. TGF- β binds to its receptor and recruits downstream Smad3 protein, causing differentiation of resting fibroblasts into myofibroblasts secreting ECM. Thus, the TGF-. beta./Smad 3 pathway has been the target of many attempts at fibrosis.

Protein lysates were harvested using RIPA buffer with 1mM phenylmethylsulfonyl fluoride (PMSF) and protease inhibitor cocktail, and protein concentrations were determined by bicinchoninic acid (BCA) kit. Each sample of the same protein (30-40. mu.g) was separated on a 10% -12% SDS/PAGE gel at 80V for 20 minutes and then converted to 120V for 1 hour. Proteins were transferred to PVDF membranes for 1 hour at 80-100V, membranes were blocked in 5% (wt/vol) dry milk in PBS containing 1% Tween 20, and then incubated overnight at 4 ℃ with the indicated primary antibody. After incubation with HRP-conjugated secondary antibody, immunoreactive bands were detected with SuperLumia ECL Plus HRP substrate kit solution (K22030, abbkinene). The primary antibody used was: α -SMA (251411, ZENBIO), collagen I (14695-1-AP, protein technology), p-smad3(AF3363, Affinity), GAPDH (AB0037, Abways), using secondary antibodies: goat Anti-Rabbit IgG (H + L) HRP (AB0101, Abways), Goat Anti-Mouse IgG (H + L) HRP (AB0102, Abways).

The inhibitory activity of compounds 9d and 29f on COL1A1, α -SMA, and p-Smad3 protein expression can be seen in FIGS. 2 and 3.

2. Pharmacokinetic experiments

The oral bioavailability of Nintedanib is 12%, and the low bioavailability limits its further clinical use. The oral bioavailability of compounds 9d and 29f was 39.88% and 41.55%, respectively, showing good druggability. The specific data are shown in tables 6 and 7.

TABLE 6 drug metabolism parameters of Compound 9d

TABLE 7 drug metabolism parameters of Compound 29f

The experimental scheme is as follows: the pharmacokinetics of the test compounds were examined by oral intravenous administration in SD rats.

Sample preparation: about 15mg of the test compound is weighed, and 2% Tween80, 2% ethanol and normal saline are added to prepare 1mg/ml of 9d or 29f compound solution for administration.

Collecting samples: 5 SD rats (Chengdou Shuo laboratory animals Co., Ltd., license number: SCXK 2015-030), male and female half, respectively according to 5mg/kg intravenous administration, 10mg/kg oral administration, 0min before administration and 5min, 15min, 30min, 1h, 2h, 4h, 6h, 8h, 10h and 24h plasma after administration. Collecting about 0.2ml blood, centrifuging 3500r 9d or 29f compound of the collected blood for 15min, collecting supernatant plasma, and freezing at-20 deg.C.

And (3) sample determination: preparing corresponding instruments and equipment; preparing a standard solution, accurately sucking a certain amount of stock solution (1mg/ml) of a compound to be detected by using a pipette, diluting the stock solution into an EP (EP) tube by using methanol, and preparing the stock solution into a solution with the concentration respectively as follows: 19.53, 39.06, 78.125, 156.25, 312.5, 625, 1250, 2500, 5000 and 10000ng/ml of standard solution of the compound to be tested; and (3) establishing a standard curve, namely taking 45 mu l of blank plasma in each EP tube, respectively adding 5 mu l of 9d or 29f compound standard solution with each concentration, uniformly mixing by vortex, adding 200 mu l of methanol precipitation protein containing 12.5ng/ml internal standard SAHA, vortex for 3min, centrifuging (13000rpm for 15min), and taking 2 mu l of supernatant for sample injection. Obtaining the solution to be tested with the theoretical concentrations of the compounds to be tested of 1.95, 3.91, 7.81, 15.63, 31.25, 62.5, 125, 250, 500 and 1000ng/ml respectively; preparing a plasma sample, thawing the collected plasma sample at room temperature, and mixing uniformly by vortex. Adding 50 μ l plasma into each EP tube, adding 200 μ l methanol precipitation protein containing 12.5ng/ml internal standard SAHA, vortexing for 3min, centrifuging (13000rpm, 15min), and sampling 2 μ l for analysis; searching liquid phase conditions; finally, the data is collated and the relevant PK value is calculated.

3. In vivo activity on bleomycin-induced pulmonary fibrosis model in mice

Pulmonary fibrosis is a long-term progressive lung disease with excessive deposition of the extracellular matrix, which is composed of collagen, fibronectin and other inflammatory cells. To explore the anti-fibrotic efficacy of compounds 9d, 29f in vivo, a bleomycin-induced pulmonary fibrosis model was used. Upon injection of bleomycin as a positive drug, Nintedanib (Nintedanib) was orally administered at a dose of 50 mg/kg/day, respectively. The compounds were also administered orally at doses of 50 and 100 mg/kg/day, respectively.

Modeling:

preparing a reagent: a. preparing chloral hydrate: chloral hydrate is prepared into 3% solution, the weight of each mouse is 20g according to the proportion of 1ml/100g, and about 200 mul is injected; b. and (3) bleomycin preparation: a bottle of the Bleomycin is 15U, 5ml of normal saline is added firstly to prepare 3U/ml of liquid, and then the liquid is diluted to 1U/ml of working solution for use. Each mouse weighed about 20g at a rate of 3.5mg/kg, and was injected with about 75. mu.l.

Modeling: after anaesthetizing, the mice were attached to a dissecting wax plate with an adhesive tape, hairs between the larynx and the chest were shaved off, the skin of the larynx was cut open, muscles around the trachea were bluntly separated with forceps, the trachea was picked out, 75 μ l of the prepared bleomycin solution was injected into the trachea, the mice were turned upside down to uniformly distribute the bleomycin in the lung, and the muscle layer and the trachea layer were sutured.

Hydroxyproline is one of the biomarkers for collagen and is therefore measured on days 14 and 28 of dosing. The content of collagen in the right lung of the mouse is measured by using a conventional hydroxyproline determination kit (Nanjing institute of bioengineering, A030-2). Briefly, the right lung was dried and acid hydrolyzed, and the residue was filtered and adjusted to pH 6.5-8.0. Hydroxyproline analysis was performed using chloramine-T spectrophotometric absorbance.

3.1 in vivo Effect of Compound 9d in pulmonary fibrosis mice

The hydroxyproline content at day 14 and day 28 of the model group was significantly increased compared to that of the sham-operated group, which means that the bleomycin-induced pulmonary fibrosis model was successfully established. The Nintedanib group was distinguished from the sham group on day 28, and chronic dosing may reduce the degree of fibrosis. At a dose of 50 mg/kg/day, the in vivo effect of inhibiting fibrosis between compound 9d and the Nintedanib group was not significant. However, compound 9d at a dose of 100 mg/kg/day showed better therapeutic effect due to its lower hydroxyproline content (fig. 4A). In addition, the survival rate of 9d was about 90% for both doses (50 mg/kg/day and 100 mg/kg/day), which was superior to the Nintedanib and model groups, indicating that 9d had lower toxicity and better efficacy.

Lung tissue sections from the sham group showed clear lung architecture, ordered arrangement of bronchial ciliated epithelium, and little edema in the bronchial and perivascular interstitial spaces. In contrast, alveolar structures in the model group were destroyed, and degeneration and necrosis of bronchial epithelial cells were clearly observed (fig. 4C ↓). There was a lot of inflammatory cell infiltration in the alveolar interstitium, mainly lymphocytes and neutrophils (fig. 4D right ↓). Drug treatment (Nintedanib group, 9d group) delayed the reduction of alveolar number and destruction of alveolar structure. The inflammatory cell infiltration was also reduced in the drug-treated group near the pulmonary artery, especially in the 9d-100mg/kg group (fig. 4).

3.2 in vivo Effect of Compound 29f in pulmonary fibrosis mice

The hydroxyproline content at day 14 and day 28 of the model group was significantly increased compared to that of the sham-operated group, which means that the bleomycin-induced pulmonary fibrosis model was successfully established.

The sections of the sham group showed clear lung architecture, the ciliated bronchial epithelium was well-aligned, and there was little edema in the bronchial and perivascular interstitial spaces. In contrast, alveolar structures in the model group were destroyed, and degeneration and necrosis of bronchial epithelial cells were clearly observed. There is a lot of inflammatory cell infiltration in the alveolar interstitium, mainly lymphocytes and neutrophils. Drug treatment (Nintedanib group, 29f group) delayed the reduction of alveolar number and destruction of alveolar structure. Inflammatory cell infiltration was also reduced in the drug-treated group near the pulmonary artery, especially at 100mg/kg in the 29f group (fig. 5).

4. In vivo Activity on carbon tetrachloride-induced liver fibrosis model

Since fibrosis has a common pathological mechanism, in order to explore the effect of compounds on hepatic fibrosis occurring in the early stage of a mouse model with acute hepatic injury in vivo, the mouse is induced by CCl4 to obtain the model with acute hepatic injury. The levels of Aspartate transaminase (AST) and Alanine transaminase (ALT) are criteria for evaluating the degree of liver damage, and higher levels of these are indicative of higher degrees of liver damage. We assessed the severity of liver damage by detecting changes in both levels; and further performing Hematoxylin-eosin staining (H & E) on the liver tissue section to directly observe the liver injury condition.

Establishing an acute liver injury model:

on the first day, each mouse was intraperitoneally injected with a CCl4-Olive Oil solution in an amount of 1ml/kg, on the second to third days, each mouse was intraperitoneally injected with a compound in an amount of 5mg/kg, on the fourth day, the mice were sacrificed, peripheral blood and liver of the mobile phone, and the next experiment was performed.

Formal experiments: according to CCl4Olive Oil 1:1.5 compounding CCl44ml, at a dose of 1ml/kg, 25. mu.l per mouse (25g) were intraperitoneally injected, 5mg of Nintedanib and compound, respectively, at 30% PEG400+ 0.5% Tween80+ 5% glycol propylene + 64.5% H2O preparation protocol, grinding with mortar, preparing 1mg/ml solution in CCl424h after injection, mice were given the compound, and each mouse (25g) was injected intraperitoneally with 125 μ l for two consecutive days.

And (3) killing: on the third day, after the abdominal cavity administration of the mouse is completed, the mouse is fasted for 16 hours, the mouse is killed on the next day, blood is taken from the eye socket and all livers are taken out, the subsequent experiment is reserved, in the administration period, after the peripheral blood of the mouse is placed in a refrigerator at 4 ℃ overnight, the mouse is centrifuged for 15min under the conditions of 4 ℃ and 3500rpm, the supernatant is taken out after centrifugation, the supernatant is frozen and stored in the refrigerator at-80 ℃ for peripheral blood ALT detection, after the liver of the mouse is taken out, PBS is washed once, water is absorbed on paper, half of the mouse is placed in 4% paraformaldehyde, and after the mouse is fixed for more than 48 hours, the subsequent dehydration work and wax block embedding are carried out for H & E staining and immunohistochemical experiments.

4.1 in vivo Effect of Compound 9d in pulmonary fibrosis mice

Compound 9d and the positive drug Nintedanib stably sustained decreased ALT levels compared to the model group. Meanwhile, as reflected by H & E pathological sections, both compound 9d and Nintedanib can significantly inhibit liver tissue necrosis, delay the progress of hepatic fibrosis, and protect the liver from damage (FIG. 6).

5 in vitro and in vivo Activity summary of Compounds

The invention relates to preparation and activity of 4-substituted coumarin, 4-substituted-2-quinolinone, 6-substituted-2-quinolinone and 7-substituted-2-quinolinone derivatives. The compounds have certain inhibition on TGF-beta induced collagen deposition of NRK-49F cells and simultaneously have low cytotoxicity. It was also observed by western blot analysis that compounds with high collagen inhibition also inhibited TGF-beta induced expression of COL1a1, alpha-SMA and p-Smad3 proteins in vitro, indicating that compounds reduced extracellular matrix ECM deposition by inhibiting the TGF-beta/Smad 3 signaling pathway. The compound is in CCl4Can obviously reduce ALT level and delay the damage of liver tissue structure on an induced acute liver injury mouse model. More importantly, the compounds reduce bleomycin-induced pulmonary fibrosis in mice by reducing hydroxyproline content and inflammatory cell infiltration in lung tissue, which may also be associated with their inhibition of the TGF- β pathway and anti-inflammation. Given their good pharmacokinetic profile, anti-fibrotic effects and in vivo toxicity, this class may be potential active candidate compounds for the treatment of tissue fibrotic diseases including pulmonary fibrosis, hepatic fibrosis, renal fibrosis.

70页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:一种高良姜素衍生物及其制备方法和应用

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!

技术分类