CN113880845A - Pyrrolotriazine derivatives, and preparation method and application thereof - Google Patents

Pyrrolotriazine derivatives, and preparation method and application thereof Download PDF

Info

Publication number
CN113880845A
CN113880845A CN202111191478.3A CN202111191478A CN113880845A CN 113880845 A CN113880845 A CN 113880845A CN 202111191478 A CN202111191478 A CN 202111191478A CN 113880845 A CN113880845 A CN 113880845A
Authority
CN
China
Prior art keywords
compound
formula
compounds
general formula
tumor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
CN202111191478.3A
Other languages
Chinese (zh)
Inventor
刘兵
王思明
董俊军
黄桐堃
吴俊杰
蔡瀚
邓联武
陈轶
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Beike Huaxia Biomedical Technology Co ltd
Guangzhou Liushun Biotechnology Co ltd
Original Assignee
Beike Huaxia Biomedical Technology Co ltd
Guangzhou Liushun Biotechnology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beike Huaxia Biomedical Technology Co ltd, Guangzhou Liushun Biotechnology Co ltd filed Critical Beike Huaxia Biomedical Technology Co ltd
Priority to CN202111191478.3A priority Critical patent/CN113880845A/en
Publication of CN113880845A publication Critical patent/CN113880845A/en
Priority to CN202210128282.8A priority patent/CN114716440B/en
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention provides a pyrrolotriazine derivative, and a preparation method and application thereof. The pyrrolotriazine derivative disclosed by the invention is a compound with a general formula I, can effectively inhibit the activity of AXL and c-Met kinase, can effectively inhibit the in-vivo growth of tumor cells, and has good safety and tolerance. The preparation method of the compound is simple in process and convenient for industrial production.

Description

Pyrrolotriazine derivatives, and preparation method and application thereof
Technical Field
The invention belongs to the technical field of medicinal chemistry, and particularly relates to a pyrrolotriazine derivative, and a preparation method and application thereof.
Background
Cancer is one of the diseases that present in the world seriously jeopardize human health and life. With the continuous and intensive research on tumor molecular biology, intracellular signal transduction, cell cycle regulation and apoptosis induction, angiogenesis, interaction between extracellular matrix and cells, and the like of malignant tumors are gradually elucidated. Among them, Receptor Tyrosine Kinases (RTKs) are closely related to the development and progression of tumors. Its effects include activating downstream signal transduction molecules, promoting cell proliferation, migration, survival, etc. Thus, RTKs have become interesting molecular therapeutic targets for antitumor therapy.
c-Met is a type of disulfide-linked heterodimeric receptor tyrosine kinase that is expressed in both normal and malignant cells of the human body. Mutations in the c-Met receptor tyrosine kinase have been found in both hereditary and secondary kidney cancers, liver cancers, and a variety of other tumors. The c-Met-HGF/SF signaling pathway plays an important physiological role in embryonic development and tissue regeneration. In normal cells, the c-Met-HGF/SF signaling pathway is tightly regulated; in tumor cells, dysregulation occurs. Numerous studies have shown that c-Met in tumor tissues can functionally interact with a variety of signaling molecules, which has become a significant cause of tumor carcinogenesis and resistance to therapy.
AXL is a member of the TAM (TYRO3, AXL, MER) Receptor Tyrosine Kinase (RTK) family. This kinase family was originally identified as transforming genes expressed in cells from patients with chronic myeloid leukemia or chronic myeloproliferative diseases. Activation of AXL is performed by its cognate protein ligand binding of growth arrest-specific protein 6(Gas6), by homodimerization of its extracellular domain or crosstalk via Interleukin (IL) -15 receptor or HER 2. AXL signaling stimulates cellular responses, including activation of the PI3K-Akt, extracellular signal-regulated kinase (ERK), and P38 mitogen-activated protein kinase cascade, the NF- κ beta pathway, and Signal Transducer and Activator of Transcription (STAT) signaling. Human biological consequences of AXL signaling include invasion, migration, survival signaling, angiogenesis, resistance to chemotherapy and targeted drugs, cellular transformation and proliferation. In addition, AXL overexpression is one of the important causes of drug resistance of patients to tumor chemotherapeutic drugs or targeted drugs.
In the course of cancer treatment, tumor metastasis and drug resistance are two major difficulties affecting the efficacy of anticancer drugs, and are also the main causes of high cancer mortality. The upregulation of AXL expression is closely related to the pathological mechanism of tumor metastasis. Many studies have shown that inhibition of AXL kinase activity can effectively block the growth, migration and invasion of tumor cells. Therefore, it is possible to use AXL kinase inhibitors in patients with early stage cancer, especially those susceptible to cancer cell metastasis, to maximize the therapeutic effect of AXL kinase inhibitors. The mechanism by which resistance is conferred upon treatment of patients with receptor tyrosine kinase inhibitors is generally secondary mutations of the targeted kinase or compensatory upregulation of other receptor tyrosine kinases. Overexpression of AXL kinase is thought to be a major cause of drug resistance due to compensatory upregulation. The synergistic effect of a targeted drug in combination with an AXL kinase inhibitor results when it becomes resistant and has been demonstrated at the cellular level and in multiple tumor models in animals. In addition, tumor cells can also develop resistance through epithelial-mesenchymal transition mechanisms. Advanced cancer patients usually need second-line or even third-line medication to reduce the resistance to the first-line antitumor drug, in this case, the AXL inhibitor can be used in combination with the first-line drug to reduce the resistance of the patients, thereby achieving the effect of inhibiting the cancer progression. Therefore, there is an urgent need to develop a dual target inhibitor of AXL and c-Met.
Disclosure of Invention
The present invention is directed to solving at least one of the problems of the prior art described above. Therefore, the invention provides the pyrrolotriazine derivative in the first aspect, which has better inhibiting effect on c-Met and AXL targets.
In a second aspect of the invention, pharmaceutical compositions comprising the above-described pyrrolotriazine derivatives are provided.
In a third aspect of the present invention, a process for producing the above pyrrolotriazine derivative is provided.
In a fourth aspect of the invention, the use of the above-mentioned pyrrolotriazine derivatives is proposed.
According to a first aspect of the present invention, there is provided a compound of formula I or a pharmaceutically acceptable salt, isomer, solvate, crystal or prodrug thereof,
Figure BDA0003301367690000021
wherein: r1Selected from H or halogen;
R2is selected from
Figure BDA0003301367690000022
R3Selected from optionally substituted by one or more RbSubstituted heteroaryl; the heteroaryl is selected from pyrazolyl, pyridyl, imidazolyl, thienyl, furyl or thiazolyl;
R4selected from H, halogen or alkoxy;
Raselected from H, C1~C6Alkyl or C1~C6An alkoxy group;
Rbis selected from C1~C4Alkyl radical, C3~C6Cycloalkyl radical, C3~C6Heterocycloalkyl or-SO2Rc(ii) a Said C1~C4Alkyl radical, C3~C6Cycloalkyl radical, C3~C6The heterocycloalkyl radical may be substituted by one or more hydroxy, amino, halogen or alkoxy radicals, RcCan be C1~C4Alkyl radical, C3~C6Cycloalkyl or phenyl;
n is any integer from 1 to 5.
In some embodiments of the invention, R3Is selected from
Figure BDA0003301367690000031
In some preferred embodiments of the invention, RbIs selected from
Figure BDA0003301367690000032
Figure BDA0003301367690000033
In some more preferred embodiments of the invention, the compound of formula I is selected from the following compounds:
Figure BDA0003301367690000034
Figure BDA0003301367690000041
according to a second aspect of the present invention, there is provided a pharmaceutical composition comprising said compound of formula I or a pharmaceutically acceptable salt, isomer, solvate, crystal or prodrug thereof.
In some embodiments of the invention, the pharmaceutical composition further comprises one or more selected from the group consisting of: tyrosine protease inhibitors, EGFR inhibitors, VEGFR inhibitors, BCR-ABL inhibitors, c-KIT inhibitors, c-Met inhibitors, RAF inhibitors, MEK inhibitors, histone deacetylase inhibitors, VEGF antibodies, EGF antibodies, HIV protein kinase inhibitors, HMG-CoA reductase inhibitors, PD-1 inhibitors, PD-L1 inhibitors, and the like.
In some preferred embodiments of the present invention, the compound, isomer, solvate, crystal or prodrug of the present invention may be mixed with a pharmaceutically acceptable carrier, diluent or excipient to prepare a pharmaceutical formulation suitable for oral or parenteral administration. Methods of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, and oral routes. The formulations may be administered by any route, for example by infusion or bolus injection, by a route of absorption through epithelial or cutaneous mucosa (e.g. oral mucosa or rectum, etc.). Administration may be systemic or local. Examples of the formulation for oral administration include solid or liquid dosage forms, specifically, tablets, pills, granules, powders, capsules, syrups, emulsions, suspensions and the like. The formulations may be prepared by methods known in the art and include carriers, diluents or excipients conventionally used in the art of pharmaceutical formulation.
According to a third aspect of the invention, there is provided a process for the preparation of a compound of formula I according to the invention: the method comprises the following steps:
a compound of formula II
Figure BDA0003301367690000051
With compounds of the formula III
Figure BDA0003301367690000052
After substitution reaction, the compound is reacted with a compound of formula IV
Figure BDA0003301367690000053
Carrying out coupling reaction to obtain a compound shown in a general formula I; wherein R is1、R2、R3As defined above for the compounds of formula I.
In some embodiments of the invention, the compound of formula II is prepared by a process comprising: a compound of formula V
Figure BDA0003301367690000054
With compounds of the formula VI
Figure BDA0003301367690000055
The condensation reaction is carried out to obtain the product.
In some preferred embodiments of the invention, the compound of formula II
Figure BDA0003301367690000056
With compounds of the formula III
Figure BDA0003301367690000061
Is prepared by substitution reaction, and the product is a compound of a formula VII
Figure BDA0003301367690000062
In some more preferred embodiments of the invention, the catalyst of the coupling reaction is selected from pd (dppf) Cl2、Pd(OAc)2、Pd2(dba)3、Pd(PPh3)2Cl2、Pd(PPh3)4At least one of commercially available palladium catalysts such as Xphos-Pd-G3, Xphos-Pd-G2, Xphos-Pd-G1, RuPhos-Pd-G3 and SPhos-Pd-G2.
In some more preferred embodiments of the invention, the base of the coupling reaction is selected from at least one of sodium carbonate, potassium carbonate, cesium carbonate, potassium phosphate, potassium acetate, DBU, triethylamine, N-diisopropylethylamine, pyridine; cesium carbonate is preferred.
According to a fourth aspect of the present invention, there is provided the use of a compound of formula I, isomer, solvate, crystal or prodrug of the present invention or a pharmaceutical composition of the present invention in the manufacture of a medicament for the treatment or prevention of a tumor.
In some embodiments of the invention, the tumor comprises at least one of lung cancer, stomach cancer, liver cancer, kidney cancer, esophageal cancer, breast cancer, leukemia, prostate cancer, colorectal cancer, bone cancer, large intestine cancer, melanoma, lymphoma, blood cancer, brain tumor, pancreatic cancer, or skin cancer.
"solvate" in the present invention refers in the conventional sense to a complex formed by the combination of a solute (e.g., active compound, salt of active compound) and a solvent (e.g., water). Solvent means a solvent known or readily determined by one skilled in the art. In the case of water, the solvate is often referred to as a hydrate, e.g., a monohydrate, a dihydrate, a trihydrate, and the like.
The term "crystalline" as used herein refers to the various solid forms formed by the compounds of the present invention, including crystalline forms and amorphous forms.
"isomers" of the present invention include configurational isomers, conformers and enantiomers of the compounds. Configurational variant refers to cis-trans isomers in either the cis or trans configuration; conformational isomers refer to stereoisomers produced by rotation of a single bond.
The "prodrug" of the present invention refers to a compound which is converted into the present invention by reaction with an enzyme, gastric acid or the like under physiological conditions of an organism, that is, a compound which is converted into the present invention by oxidation, reduction, hydrolysis or the like by an enzyme and/or a compound which is converted into the present invention by hydrolysis reaction of gastric acid or the like.
The "pharmaceutically acceptable salt" of the present invention refers to a pharmaceutically acceptable salt of a compound of the present invention with an acid, including, but not limited to, phosphoric acid, sulfuric acid, hydrochloric acid, hydrobromic acid, citric acid, maleic acid, malonic acid, mandelic acid, succinic acid, fumaric acid, acetic acid, lactic acid, nitric acid, and the like.
The "pharmaceutical composition" of the present invention is intended to include a mixture of any one of the compounds described herein, including isomers, prodrugs, solvates, pharmaceutically acceptable salts, or chemically protected forms thereof, and one or more pharmaceutically acceptable carriers.
By "pharmaceutically acceptable carrier" herein is meant a carrier that does not cause significant irritation to an organism and does not interfere with the biological activity and properties of the administered compound, and includes solvents, diluents or other excipients, dispersants, surfactants, isotonicity agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like. Unless any conventional carrier medium is incompatible with the compounds of the present invention. Some examples of carriers that may be pharmaceutically acceptable include, but are not limited to, sugars such as lactose, glucose, and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, and cellulose acetate; malt, gelatin, and the like.
"excipient" in the context of the present invention refers to an inert substance added to a pharmaceutical composition to further facilitate administration of the compound. Excipients may include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, polyethylene glycols.
The term "use in the preparation of a medicament for treating or preventing a tumor" of the present invention means that the growth, development and/or metastasis of a tumor can be inhibited, and a therapeutically effective amount of a compound of the present invention is administered to a human or animal in need thereof to inhibit, slow or reverse the growth, migration or spread of a tumor in the subject.
The invention has the beneficial effects that:
1. the compounds of the present invention are effective in inhibiting the activity of AXL and c-Met kinases.
2. The compound can effectively inhibit the in-vivo growth of tumor cells and has good safety and tolerance.
3. The preparation method of the compound is simple in process and convenient for industrial production.
Drawings
The invention is further described with reference to the following figures and examples, in which:
FIG. 1 is a graph showing the growth of tumor volumes in mice of the compound group, the solvent control group and the positive control group in Experimental example 2 of the present invention.
FIG. 2 is a graph showing the change in body weight with treatment time of mice in the compound group, the solvent control group and the positive control group in test example 2 of the present invention.
Detailed Description
The concept and technical effects of the present invention will be clearly and completely described below in conjunction with the embodiments to fully understand the objects, features and effects of the present invention. It is obvious that the described embodiments are only a part of the embodiments of the present invention, and not all embodiments, and those skilled in the art can obtain other embodiments without inventive effort based on the embodiments of the present invention, and all embodiments are within the protection scope of the present invention.
Example 1
This example prepares an N- (4- (6- (1-methyl-1H-pyrazol-4-yl) pyrrolo [2,1-f ] [1,2,4] triazin-4-yl) oxy) phenyl) -N- (4-fluorophenyl) cyclopropane-1, 1-dimethylamide by:
Figure BDA0003301367690000081
6-bromo-4-chloropyrrolo [2,1-f ] [1,2,4] triazine (920mg,3.96mmol,1.0eq), N- (4-fluorophenyl) -N- (4-hydroxyphenyl) cyclopropane-1, 1-dimethylamide (1.31g,4.16mmol,1.05eq) and potassium carbonate (820mg,5.94mmol,1.5eq) were added to a reaction flask at room temperature, suspended in DMF (12mL), purged with nitrogen 3 times, transferred to an 80 ℃ oil bath and stirred for 6 hours.
The reaction solution was cooled to 20 ℃, slowly poured into a saturated ammonium chloride solution (60mL), extracted with ethyl acetate (20mL × 5), the organic phase was washed with a saturated saline solution (40mL), dried over sodium sulfate, concentrated under reduced pressure at 40 ℃, and purified by column chromatography to obtain 1.30g of a white solid.
LCMS(ESI):m/z 509/511[M+H]+
Figure BDA0003301367690000082
N- (4- ((6-bromopyrrolo [2, 1-f)][1,2,4]Triazin-4-yl) oxy) phenyl) -N- (4-fluorophenyl) cyclopropane-1, 1-dimethylamide (500mg,0.980mmol,1.0eq), 1-methyl-4-pyrazoleboronic acid pinacol ester (214mg,1.03mmol,1.05eq), cesium carbonate (957mg,2.94mmol,3.0eq), Pd (dppf) Cl2(41.5mg) was added to the reaction flask at ordinary temperature, and the mixture was dissolved in isobutanol (4mL) and water (2mL) to prepare a suspension, which was purged with nitrogen 3 times, transferred to an oil bath at 85 ℃ and stirred for 13 hours. TLC monitoring (developing solvent ratio petroleum ether: ethyl acetate 1: 1, R)f(product) ═ 0.25).
The reaction solution was cooled to 20 ℃, slowly poured into saturated EDTA-EDTA disodium salt buffer (20mL), extracted with ethyl acetate (15mL × 4), the organic phase was washed with saturated brine (30mL), dried over sodium sulfate, and concentrated at 40 ℃ under reduced pressure to give a crude product. Dissolving the crude product in a mixed solution of dimethylformamide (4mL) and acetonitrile (4mL), passing through a liquid chromatographic column, and carrying out reverse phase separation on 1mL of single needle; the mobile phase is 4 per mill ammonia water: acetonitrile, gradient elution: 0-30 min, 50-100%, the retention time of the target product is 18min, and 80% acetonitrile. Column model (DaCao C18-100-8um, ID 25.4mm 450 mm). The fractions were concentrated under reduced pressure at 40 ℃ and freeze-dried for 17.5 hours. 23mg of a white solid were obtained.
LCMS(ESI):m/z 511[M+H]+1H NMR(500MHz,DMSO-d6)δ10.12(s,1H),10.02(s,1H),8.30(d,J=1.6Hz,1H),8.12(s,1H),8.05(s,1H),7.90(s,1H),7.73–7.68(m,2H),7.62(dd,J=8.9,5.1Hz,2H),7.30–7.20(m,3H),7.14(t,J=8.9Hz,2H),3.90(q,J=5.5Hz,3H),1.46(s,4H).
Example 2
This example prepares an N- (4- (6- (1- (2-hydroxyethyl) -1H-pyrazol-4-yl) pyrrolo [2,1-f ] [1,2,4] triazin-4-yl) oxy) phenyl) -N- (4-fluorophenyl) cyclopropane-1, 1-dimethylamide by the following steps:
Figure BDA0003301367690000091
n- (4- ((6-bromopyrrolo [2, 1-f)][1,2,4]Triazin-4-yl) oxy) phenyl) -N- (4-fluorophenyl) cyclopropane-1, 1-dimethylamide (600mg,1.18mmol,1.0eq), 1- (hydroxyethyl) pyrazole-4-boronic acid pinacol ester (294mg,1.24mmol,1.05eq), cesium carbonate (1.15g,3.54mmol,3.0eq) and Pd (dppf) Cl2(100mg) was added to the reaction flask at room temperature, and the mixture was suspended in t-amyl alcohol (6mL) and water (1.8mL), purged with nitrogen 3 times, transferred to a 97 ℃ oil bath and stirred for 6.5 hours.
The reaction mixture was cooled to 20 ℃, slowly poured into a saturated ammonium chloride solution (50mL), extracted with ethyl acetate (15mL × 7), the organic phase was washed with a saturated saline solution (40mL), dried over sodium sulfate, concentrated under reduced pressure at 40 ℃, and purified by column chromatography to obtain 284mg of an off-white solid.
LCMS(ESI):m/z 542[M+H]+1H NMR(500MHz,DMSO-d6)δ10.14(s,1H),10.04(s,1H),8.32(d,J=1.6Hz,1H),8.14(s,1H),8.05(s,1H),7.91(s,1H),7.73–7.68(m,2H),7.63(dd,J=8.9,5.1Hz,2H),7.30–7.22(m,3H),7.14(t,J=8.9Hz,2H),4.92(t,J=5.2Hz,1H),4.16(t,J=5.6Hz,2H),3.77(q,J=5.5Hz,2H),1.48(s,4H).
Test example 1
This test example tests the inhibition of the enzymatic activity (IC) of the compounds on the kinases AXL and c-Met50)
In the experiment, a Mobility shift assay (Mobility shift assay) method is used, compounds are screened on AXL and c-Met kinase, initial concentration is 10000nM, 3-fold dilution is carried out, 10 concentrations are carried out, and multi-well detection is carried out.
Reagents and consumables are shown in table 1:
TABLE 1
Figure BDA0003301367690000092
Figure BDA0003301367690000101
The instrument comprises the following steps:
centrifuge (manufacturer: Eppendorf, model 5430)
Enzyme mark instrument (manufacturer: Perkin Elmer, model: Caliper EZ Reader II)
Echo 550 (manufacturer: Labcyte, model: Echo 550)
Enzyme-linked immunosorbent assay (manufacturer: Perkin Elmer, model: Envision)
The specific process is as follows:
1) preparing 1 Xkinase buffer solution;
2) preparation of compound concentration gradient: the test concentration of the tested compound is 10000nM, 3 times dilution, 10 concentrations, and multiple-hole detection; 100% DMSO solution diluted to 100-fold final concentration, 3-fold compound dilution, 10 concentrations in 384-mesh plates. Using a dispenser Echo 550 to the target plate 384-well plate transfer 250nL 100 times the final concentration of the compound.
3) A2.5 fold final concentration of kinase solution was prepared in 1 Xkinase buffer.
4) Add 10. mu.L of 2.5 fold final concentration kinase solution to the compound well and positive control well, respectively; add 10. mu.L of 1 Xkinase buffer to the negative control wells.
5) Centrifuge at 1000rpm for 30 seconds, shake the plate and incubate at room temperature for 10 minutes.
6) A mixed solution of ATP and kinase substrate at 5/3-fold final concentration was prepared using 1 Xkinase buffer.
7) The reaction was initiated by adding 15. mu.L of a mixed solution of ATP and substrate at 5/3-fold final concentration.
8) The 384 well plates were centrifuged at 1000rpm for 30 seconds, shaken and mixed and incubated at room temperature for the appropriate time.
9) Add 30. mu.L of termination detection solution to stop the kinase reaction, centrifuge at 1000rpm for 30 seconds, shake and mix.
10) The conversion was read with a microplate Reader (Caliper EZ Reader).
The dose-effect curves were fitted using the analysis software GraphPad Prism 5 log (inhibitor) vs. response-Variable slope to obtain the IC of each compound for enzyme activity50The value is obtained. The results are shown in Table 2.
TABLE 2
Figure BDA0003301367690000111
As is clear from the above table, the compounds of the present invention are effective in inhibiting the activity of AXL and c-Met kinases. The compound of example 2 exhibited slightly higher or comparable activity inhibitory potency compared to the positive controls cabozantinib, sertraline.
Test example 2
The experimental example tests the in vivo anti-tumor activity of the compound in the model mouse tumor cell ectopic transplantation experiment, and the concrete process is as follows:
selecting 12 female Balb/c nude mice with age of 6-8 weeks, inoculating human lung cancer cell line A549 tumor cell strain 5 x 10 in subcutaneous ectopic mode 626 days after the inoculation of the tumor cells, the tumor grows to 60mm3~250mm3At the time, mice were gavaged with the samples to be tested。
Mice were divided into a negative solvent control group, a compound of the invention group (compound of example 2) (20mg/kg) and a positive control group of sertraline (20mg/kg), 4 mice per group. All dose groups adopt equal-volume unequal-concentration oral administration and single-time intragastric administration every day, and the administration volume is 10 mL/kg. The negative solvent control group was administered the same volume of blank vehicle (DMSO: Solutol: water 1: 2: 7) once a day for 14 consecutive days.
After the start of the administration, the body weight and tumor size of the mice were measured twice a week. Tumor size calculation formula:
tumor volume (mm)3) 0.5 × (tumor major diameter × tumor minor diameter)2)。
The antitumor efficacy was evaluated based on the growth curve of the tumor under treatment (i.e., tumor volume per measurement versus its number of days of treatment) and relative tumor volume. Wherein the relative tumor inhibition ratio (TGI) is calculated according to the following formula:
relative tumor inhibition ratio TGI (%): TGI% (1-T/C) × 100%.
T/C% is the relative tumor proliferation rate, i.e., the percentage value of the relative tumor volume or tumor weight of the treated and control groups at a certain time point. T and C are the Relative Tumor Volume (RTV) or Tumor Weight (TW) at a particular time point in the treated and control groups, respectively. The calculation formula is as follows: T/C%RTV/CRTV×100%(TRTV: treatment group mean RTV; cRTV: vehicle control mean RTV; RTV-V0, V0 is the tumor volume of the animal at the time of the grouping, and Vt is the tumor volume of the animal after treatment). Or T/C% ═ TTW/CTW × 100% (TTW: average tumor weight at the end of the test in the treatment group and CTW: average tumor weight at the end of the test in the vehicle control group). The results are shown in FIGS. 1 and 2.
FIG. 1 shows the growth changes in tumor volume in mice of the compound group of the present invention, the solvent control group and the positive control group. As shown in the figure, the compound can effectively inhibit the growth of tumor cells in a model mouse, and the relative tumor inhibition rate (TGI) is 89.02% by volume; 63.95% by tumor weight, which is higher than 57.37% of the positive control seratrotinib.
FIG. 2 shows the body weight of mice in the compound group of the present invention, the solvent control group and the positive control group as a function of the treatment time. As shown in the figure, the body weight of the tumor-bearing mice has no obvious change in the experimental process, and the compound has good safety and tolerance.
The embodiments of the present invention have been described in detail, but the present invention is not limited to the embodiments, and various changes can be made without departing from the gist of the present invention within the knowledge of those skilled in the art. Furthermore, the embodiments of the present invention and the features of the embodiments may be combined with each other without conflict.

Claims (10)

1. A compound of formula I or a pharmaceutically acceptable salt, isomer, solvate, crystal or prodrug thereof,
Figure FDA0003301367680000011
wherein: r1Selected from H or halogen;
R2is selected from
Figure FDA0003301367680000012
R3Selected from optionally substituted by one or more RbSubstituted heteroaryl; the heteroaryl is selected from pyrazolyl, pyridyl, imidazolyl, thienyl, furyl or thiazolyl;
R4selected from H, halogen or alkoxy;
Raselected from H, C1~C6Alkyl or C1~C6An alkoxy group;
Rbis selected from C1~C4Alkyl radical, C3~C6Cycloalkyl radical, C3~C6Heterocycloalkyl or-SO2Rc(ii) a Said C1~C4Alkyl radical, C3~C6Cycloalkyl radical, C3~C6Heterocycloalkyl radical being substituted by oneOr substituted by more hydroxy, amino, halogen or alkoxy groups, RcCan be C1~C4Alkyl radical, C3~C6Cycloalkyl or phenyl;
n is any integer from 1 to 5.
2. Compounds of general formula I according to claim 1, characterized in that: r3Is selected from
Figure FDA0003301367680000013
3. Compounds of general formula I according to claim 1, characterized in that: rbIs selected from
Figure FDA0003301367680000014
Figure FDA0003301367680000015
4. Compounds of general formula I according to claim 1, characterized in that: the compound of the general formula I is selected from the following compounds:
Figure FDA0003301367680000021
Figure FDA0003301367680000031
5. a pharmaceutical composition comprising a compound of general formula I according to any one of claims 1 to 4 or a pharmaceutically acceptable salt, isomer, solvate, crystal or prodrug thereof.
6. A process for the preparation of a compound of formula I as claimed in any one of claims 1 to 4: the method is characterized in that: the method comprises the following steps:
a compound of formula II
Figure FDA0003301367680000032
With compounds of the formula III
Figure FDA0003301367680000033
After substitution reaction, the compound is reacted with a compound of formula IV
Figure FDA0003301367680000034
Carrying out coupling reaction to obtain a compound shown in a general formula I; wherein R is1、R2、R3As defined above for the compounds of formula I.
7. The method of claim 6, wherein: the preparation method of the compound of the formula II comprises the following steps: a compound of formula V
Figure FDA0003301367680000035
With compounds of the formula VI
Figure FDA0003301367680000036
The condensation reaction is carried out to obtain the product.
8. The method of claim 6, wherein: the compound of formula II
Figure FDA0003301367680000037
With compounds of the formula III
Figure FDA0003301367680000041
Substitution reaction takes place, the product being a compound of formula VII
Figure FDA0003301367680000042
9. The method of claim 6The method is characterized in that: the catalyst for the coupling reaction is selected from Pd (dppf) Cl2、Pd(OAc)2、Pd2(dba)3、Pd(PPh3)2Cl2、Pd(PPh3)4At least one of Xphos-Pd-G3, Xphos-Pd-G2, Xphos-Pd-G1, RuPhos-Pd-G3 or SPhos-Pd-G2.
10. Use of a compound of general formula I, an isomer, a solvate, a crystal or a prodrug of any of claims 1 to 4 or a pharmaceutical composition of claim 5 for the preparation of a medicament for the treatment or prevention of a tumor.
CN202111191478.3A 2021-10-13 2021-10-13 Pyrrolotriazine derivatives, and preparation method and application thereof Withdrawn CN113880845A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202111191478.3A CN113880845A (en) 2021-10-13 2021-10-13 Pyrrolotriazine derivatives, and preparation method and application thereof
CN202210128282.8A CN114716440B (en) 2021-10-13 2022-02-11 Pyrrolotriazine derivatives, and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111191478.3A CN113880845A (en) 2021-10-13 2021-10-13 Pyrrolotriazine derivatives, and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN113880845A true CN113880845A (en) 2022-01-04

Family

ID=79002623

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202111191478.3A Withdrawn CN113880845A (en) 2021-10-13 2021-10-13 Pyrrolotriazine derivatives, and preparation method and application thereof
CN202210128282.8A Active CN114716440B (en) 2021-10-13 2022-02-11 Pyrrolotriazine derivatives, and preparation method and application thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202210128282.8A Active CN114716440B (en) 2021-10-13 2022-02-11 Pyrrolotriazine derivatives, and preparation method and application thereof

Country Status (1)

Country Link
CN (2) CN113880845A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114573553A (en) * 2022-01-27 2022-06-03 广州六顺生物科技股份有限公司 Heteroaromatic ring derivative and preparation method and application thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7439246B2 (en) * 2004-06-28 2008-10-21 Bristol-Myers Squibb Company Fused heterocyclic kinase inhibitors
US7173031B2 (en) * 2004-06-28 2007-02-06 Bristol-Myers Squibb Company Pyrrolotriazine kinase inhibitors
CN101005843A (en) * 2004-06-28 2007-07-25 布里斯托尔-迈尔斯·斯奎布公司 Pyrrolotriazine kinase inhibitors

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114573553A (en) * 2022-01-27 2022-06-03 广州六顺生物科技股份有限公司 Heteroaromatic ring derivative and preparation method and application thereof
CN114573553B (en) * 2022-01-27 2023-11-10 广州六顺生物科技有限公司 Heteroaromatic ring derivative and preparation method and application thereof

Also Published As

Publication number Publication date
CN114716440A (en) 2022-07-08
CN114716440B (en) 2023-06-30

Similar Documents

Publication Publication Date Title
TW202043210A (en) Pyridazine derivative inhibitor, preparation method and application thereof
TWI772291B (en) Substituted pyrrolopyrimidine CDK inhibitors, pharmaceutical compositions containing the same and uses thereof
JP5689069B2 (en) Pyrazolopyridine PI3K inhibitor compounds and methods of use
EP3845527A1 (en) Multi-substituted pyridone derivatives and medical use thereof
CN105985342A (en) Pyrimido pyrimidine dione derivative as EGFR inhibitor and application thereof
CN108530444A (en) A kind of novel NAMPT and IDO double inhibitors and preparation method thereof and medical usage
EP3705480A1 (en) Class of amino-substituted nitrogen-containing fused ring compounds, preparation method therefor, and use thereof
CN116178347A (en) PRMT5-MTA inhibitors
JP2022502438A (en) FGFR4 inhibitor and its use
JP6909236B2 (en) Quinazoline derivative or salt thereof and a pharmaceutical composition containing it
JP6913955B2 (en) Molten quinoline compound as PI3K / mTOR inhibitor
CN113880845A (en) Pyrrolotriazine derivatives, and preparation method and application thereof
CN103664734B (en) Heterocycle hydroximic acid compound and medicinal compositions thereof and application
CN107428762B (en) Phthalazinone derivatives, preparation method and use thereof
CN115803326B (en) EGFR inhibitor, preparation method thereof and application thereof in pharmacy
WO2023143147A1 (en) Pyridazopyridone compounds, pharmaceutical composition thereof and use thereof
US10301325B2 (en) Quinoline derivative, and pharmaceutical composition, preparation method and use thereof
CN102688234A (en) Synthesis of indolone derivative as RSK2 inhibitor and application
RU2627692C1 (en) N-{3-[3-cyclopropyl-5-(2-fluoro-4-iodophenylamino)-6,8-dimethyl-2,4,7-trioxo-3,4,6,7-tetrahydro-2h-pyrido[4,3-d]pyrimidine-1-il]-phenyl}-cyclopropanecarboxamide dimethylsulfoxide solvate as mek1/2 inhibitor
CA2966709C (en) Cytidine derivative dimers and applications thereof
CN116096372A (en) EGFR inhibitor, preparation method and pharmaceutical application thereof
JP2023524230A (en) Histone deacetylase inhibitors for immunomodulation in the tumor microenvironment
CN105541792A (en) Polycyclic PI3K inhibitors
CN103965202B (en) Bicyclic-fused heterogeneous ring compound, Preparation Method And The Use
CN114573553B (en) Heteroaromatic ring derivative and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
WW01 Invention patent application withdrawn after publication

Application publication date: 20220104

WW01 Invention patent application withdrawn after publication