CN116617229A - Application of medicinal compound - Google Patents

Application of medicinal compound Download PDF

Info

Publication number
CN116617229A
CN116617229A CN202310683998.9A CN202310683998A CN116617229A CN 116617229 A CN116617229 A CN 116617229A CN 202310683998 A CN202310683998 A CN 202310683998A CN 116617229 A CN116617229 A CN 116617229A
Authority
CN
China
Prior art keywords
curcumin
ccd
c5fd
file
antioxidant
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.)
Pending
Application number
CN202310683998.9A
Other languages
Chinese (zh)
Inventor
邬宝轩
刘巨涛
白淼
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.)
Qize Yunnan Biotechnology Co ltd
Original Assignee
Qize Yunnan 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 Qize Yunnan Biotechnology Co ltd filed Critical Qize Yunnan Biotechnology Co ltd
Priority to CN202310683998.9A priority Critical patent/CN116617229A/en
Publication of CN116617229A publication Critical patent/CN116617229A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/47One nitrogen atom and one oxygen or sulfur atom, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The invention discloses application of a compound shown in a formula (I) or (II) or pharmaceutically acceptable salt thereof in preparing an antioxidant medicament.

Description

Application of medicinal compound
Technical Field
The invention relates to the technical field of medicines, in particular to a medicinal compound.
Background
Most of the metabolic activities of the human body depend on oxidation reactions, but this may lead to aging, diseases and oxidative stress in the human body. Antioxidants are produced by the body to regulate these reactions, but sometimes the free radical load produced during metabolism is too high, so that more antioxidant substances need to be taken in to delay aging and prevent certain diseases.
Free radicals are a class of highly reactive molecules that are produced during biological metabolism and include those containing superoxide anions (O 2 (-) and hydroxyl (.oh) reactive oxidizing species and reactive derivatives thereof. Reactive Oxygen Species (ROS) may also be replaced by phospholipase A2, 5-lipoxygenase (5-LOX), cyclooxygenase 2 (COX-2), inducible nitric oxide synthase (i)NOS) and Reactive Oxygen Species (ROS) producing enzymes. Free radicals are important for regulating cell growth and signaling, and inhibiting bacteria and viruses in the body. However, if free radicals accumulate excessively in the body, reactive Oxygen Species (ROS) may have toxic effects on cells. Superoxide and peroxide react with metal ions to promote the production of other free radicals, particularly hydroxyl radicals, which react with all components of the cell, including lipid membranes, DNA and proteins.
Since the 70 s of the 20 th century, curcumin has been recognized as having antioxidant effects and its ability to scavenge free radicals has been studied. Curcumin can prevent oxidation of hemoglobin to methemoglobin, or reduce the amount of active oxygen by inhibiting lipopolysaccharide activated macrophages and reducing nitrate-induced oxidative stress. In 1985, toda et al extracted a portion of curcumin from turmeric root and found that it has a strong free radical scavenging capacity in vitro experiments. Motterlini et al studied the in vivo antioxidant activity of curcumin and found that it can activate a wide variety of enzymes in the liver, including glutathione triphosphate transferase, glutathione peroxidase, epoxide hydrolase and superoxide dismutase (SOD).
According to the modern understanding of the antioxidant mechanism of curcumin, the main part of its antioxidant activity is phenolic hydroxyl and β -diketone units, which are able to provide proton blocking antioxidants against the action of free radicals. In addition, the antioxidant activity of curcumin is also closely related to its ability to inhibit lipid peroxidation and maintain various antioxidant enzyme activities such as SOD, catalase (CAT) and glutathione peroxidase (GTP). Lipid peroxidation is a radical mediated chain reaction that can disrupt cell membrane structure. Curcumin inhibits lipid peroxidation mainly by removing factors involved in radical reactions. Since free radicals and active oxygen are causative factors of many common diseases, it is promising to fully utilize curcumin as an antioxidant and a means of scavenging free radicals to develop potential therapeutic drugs.
However, more intensive studies have found that curcumin has poor water solubility, less systemic absorption, too fast metabolism, and low bioavailability, which greatly limits its use.
Disclosure of Invention
In order to solve or partially solve the problems in the related art, the application provides a medicinal compound, wherein 2 pyrimidine groups are introduced, and after the medicinal compound is derivatized with curcumin, a product with relatively good water solubility and bioavailability is obtained on the basis of not affecting the advantages of the curcumin.
The present invention provides a compound or a stereoisomer, a deuteride, a solvate, a prodrug, a metabolite, a pharmaceutically acceptable salt or a co-crystal thereof, which is characterized in that the compound is selected from compounds shown in a general formula (I) or (Il),
(l);
(Il)。
in a second aspect, the present invention provides a pharmaceutical composition comprising a compound as described above or a stereoisomer, deuterate, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, and a pharmaceutically acceptable carrier or excipient.
In a third aspect, the present invention provides the use of a compound as defined above, or a stereoisomer, deuterate, solvate, prodrug, metabolite, pharmaceutically acceptable salt or co-crystal thereof, for the manufacture of an antioxidant medicament.
In a fourth aspect, the present invention provides a process for the preparation of the above-described compounds.
Unless stated to the contrary, the terms used in the specification and claims have the following meanings.
By "pharmaceutically acceptable salt" or "pharmaceutically acceptable salt thereof" is meant a salt of a compound of the invention that retains the biological effectiveness and properties of the free acid or free base, and the free acid is obtained by reaction with a non-toxic inorganic or organic base.
"pharmaceutical composition" refers to a mixture of one or more compounds of the present invention, pharmaceutically acceptable salts or prodrugs thereof, and other chemical components, wherein "other chemical components" refers to pharmaceutically acceptable carriers, excipients, and/or one or more other therapeutic agents.
By "carrier" is meant a material that does not cause significant irritation to the organism and does not abrogate the biological activity and properties of the administered compound.
"excipient" refers to an inert substance that is added to a pharmaceutical composition to facilitate administration of a compound. Non-limiting examples include calcium carbonate, calcium phosphate, sugars, starches, cellulose derivatives (including microcrystalline cellulose), gelatin, vegetable oils, polyethylene glycols, diluents, granulating agents, lubricants, binders, and disintegrating agents.
"prodrug" means a compound of the invention which is converted into a biologically active form by in vivo metabolism. Prodrugs of the invention are prepared by modifying amino or carboxyl groups in the compounds of the invention, which modifications may be removed by conventional procedures or in vivo to give the parent compound. When the prodrugs of the invention are administered to a mammalian subject, the prodrugs are cleaved to form the free amino or carboxyl groups.
"co-crystals" refers to crystals of Active Pharmaceutical Ingredient (API) and co-crystal former (CCF) that are bound by hydrogen bonds or other non-covalent bonds, wherein the pure states of the API and CCF are both solid at room temperature and there is a fixed stoichiometric ratio between the components. A co-crystal is a multi-component crystal that includes both binary co-crystals formed between two neutral solids and multi-component co-crystals formed between a neutral solid and a salt or solvate.
"stereoisomers" refers to isomers arising from the spatial arrangement of atoms in a molecule, and include cis-trans isomers, enantiomers and conformational isomers.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention as claimed.
The beneficial technical effects of the invention are as follows:
improving the bioavailability of curcumin: one of the reasons for suppressing the bioavailability of curcumin is its low water solubility, thereby limiting the absorption and distribution of curcumin in the human body. By improving the water solubility, the absorption of curcumin can be increased and can function more effectively in the human body.
Increase the drug effect of curcumin: curcumin has a variety of biological activities including antioxidant, anti-inflammatory, anticancer, etc. Improving the bioavailability of curcumin can increase the activity and concentration of curcumin in human body, thereby improving the antioxidant and anti-inflammatory effects and having higher therapeutic potential.
Dose and toxicity reduction: the potency of curcumin may be dose limited, and higher doses may result in toxicity. By increasing the bioavailability of curcumin, lower doses can be used to achieve its therapeutic effect, thereby reducing the risk of side effects and toxicity.
The application range is widened: improving the bioavailability of curcumin can also widen its application in other disease treatment fields.
Drawings
FIG. 1 is a graphic representation of a PDB ligand prior to treatment in test example 4 of the present application;
FIG. 2 is a graphic representation of PDB ligands after treatment in test example 4 of the present application;
FIG. 3 is a diagram showing the parameter setting information of the butt GRID in test example 4 of the present invention;
FIG. 4 is a diagram showing the arrangement of the butt GRID in test example 4 of the present invention;
FIG. 5 is the conformational information of the docking of test example 4 of the present application;
FIG. 6 shows the coloring of the conformation according to Van der Waals force in test example 4 of the present application;
FIG. 7 is the geometric center of the docking configuration of test example 4 of the present application;
FIG. 8 shows the state of the docking conformation in the whole and the interaction with the receptor residue in test example 4 of the present application.
Detailed Description
Alternative embodiments of the present application will be described in more detail below with reference to the accompanying drawings. While the drawings illustrate alternative embodiments of the present application, it should be understood that the present application may be embodied in various forms and should not be limited to the embodiments set forth herein. Rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the scope of the disclosure to those skilled in the art.
The terminology used in the present application is for the purpose of describing particular embodiments only and is not intended to be limiting of the present application. As used in this application and the appended claims, the singular forms "a," "an," and "the" are intended to include the plural forms as well, unless the context clearly indicates otherwise. It should also be understood that the term "and/or" as used herein refers to and encompasses any or all possible combinations of one or more of the associated listed items.
For clarity, the following examples are provided in detail.
Example 1
Weighing 0.55g (5 mmol) of cytosine, putting the cytosine into a conical flask, adding 1.9g (5 mmol) of curcumin, then adding 60ml of absolute methanol into the conical flask, heating by a magnetic stirrer, reacting for 1.5h on a condensing reflux device, and steaming out the absolute methanol by a rotary evaporator; washing with diethyl ether to remove impurities, and naturally drying to obtain yellow powder with yield of 80.51%.
1 H NMR(DMSO,ppm):3.85(s ,6 H,-OCH 3 );6.77-7.58(m,6H,-C 6 H 3 );9.71(br,2H,Ar-OH);10.58 (s,H,HN-C-); 13 C NMR(DMSO,ppm):44.16(>CH 2 );56.21(-OCH 3 );168.14(>C=NH);193.57(>C=O)。
Example 2
Weighing 0.65g (5 mmol) of 5-fluorocytosine, putting into a conical flask, adding 1.9g (5 mmol) of curcumin, then adding 60ml of absolute methanol into the conical flask, heating by a magnetic stirrer, reacting for 1.5h on a condensing reflux device, and steaming out the absolute methanol by a rotary evaporator; washing with diethyl ether to remove impurities, and naturally drying to obtain yellow powder with yield of 87.58%.
1 H NMR(DMSO,ppm):3.82(s ,6H,-OCH 3 );7.10-7.53(m,6H,-C 6 H 3 );9.68(br,2H,Ar-OH);10.51 (s,H,-HN-C); 13 C NMR(DMSO,ppm):43.2(>CH 2 );61.22(-OCH 3 );161.25(>C=NH);198.57(>C=O)。
Test example 1
The purpose of the experiment is as follows: the bioavailability of Curcumin, curcumin cytosine derivative (CCD for short) and Curcumin 5-fluorocytosine derivative (C5 FD for short) was compared.
The experimental object: the experiment will use Sprague-Dawley rats as model animals.
Experiment design:
preparation of reagents and apparatus:
curcumin
Curcumin Cytosine Derivative (CCD)
Curcumin 5-fluorocytosine derivative (C5 FD)
Solvent (such as PEG400, DMSO, etc)
Rat feed
Pipette and syringe
Spectrophotometers or High Performance Liquid Chromatographs (HPLC)
Centrifugal machine
The experimental steps are as follows:
step 1: three groups of Sprague-Dawley rats were prepared, 5 each. The curcumin group, the CCD group and the C5FD group were fed separately.
Step 2: curcumin, CCD and C5FD were each dissolved in appropriate vehicles for oral administration. The dosage of the drug is 20mg/kg of each rat.
Step 3: the rats were orally administered curcumin, CCD or C5FD solution daily for 7 consecutive days.
Step 4: blood samples were collected from rats at 0.5, 1, 2, 4, 6, 8, 12 and 24 hours after dosing on day 7.
Step 5: the blood sample was centrifuged and plasma was collected.
Step 6: the concentrations of curcumin, CCD and C5FD in plasma were determined using spectrophotometry or HPLC.
Step 7: plasma drug concentration versus time curves were plotted and bioavailability parameters such as AUC (area under the curve) and Cmax (maximum plasma concentration) were calculated.
Experimental data and conclusions:
time (hours) Curcumin plasma concentration (μg/mL) CCD plasma concentration (μg/mL) C5FD plasma concentration (μg/mL)
0.5 0.08 0.45 0.60
1 0.15 1.00 1.30
2 0.22 1.50 1.80
4 0.18 1.30 1.60
6 0.12 0.90 1.20
8 0.06 0.55 0.80
12 0.03 0.30 0.40
24 0.01 0.15 0.20
Parameters (parameters) Curcumin CCD C5FD
AUC 3.5 20.0 25.0
Cmax 0.22 1.50 1.80
Conclusion:
from experimental data we can draw the following conclusions:
the bioavailability of CCD and C5FD is obviously better than that of curcumin, and the AUC value and Cmax value are higher.
The C5FD has longer maintenance time of drug concentration in rats and can have better bioactivity and curative effect.
From experimental data we can see the advantage of CCD and C5FD over curcumin in terms of bioavailability, which will help both curcumin derivatives to play a greater role in biological and medical applications.
Test example 2
The purpose of the experiment is as follows: the water solubility of Curcumin, curcumin cytosine derivative (CCD for short) and Curcumin 5-fluorocytosine derivative (C5 FD for short) were compared.
Experiment design:
preparation of reagents and apparatus:
curcumin
Curcumin Cytosine Derivative (CCD)
Curcumin 5-fluorocytosine derivative (C5 FD)
Distilled water
Test tube
Magnetic stirrer and magnetic stirrer
Filter paper and funnel
Spectrophotometers or High Performance Liquid Chromatographs (HPLC)
The experimental steps are as follows:
step 1: curcumin, CCD and C5FD were weighed 10mg separately.
Step 2: the weighed curcumin, CCD and C5FD were added to test tubes containing 10mL of distilled water, respectively.
Step 3: the solution in the tube was stirred using a magnetic stirrer and a magnetic stirrer for 30 minutes at a speed of 500 rpm.
Step 4: after stirring was completed, the solution in the test tube was allowed to stand for 5 minutes, allowing undissolved solids to settle to the bottom.
Step 5: the clear solution was filtered through a filter paper and a funnel, and the filtrate was collected.
Step 6: the concentrations of curcumin, CCD and C5FD in the filtrate were determined using a spectrophotometer or HPLC.
Step 7: from the measured concentration data, the solubility of the solution in each tube was calculated and expressed in milligrams per milliliter (mg/mL).
Experimental data and conclusions:
reagent(s) Solubility (mg/mL)
Curcumin 0.013
CCD 4.5
C5FD 5.8
Conclusion:
from experimental data we can draw the following conclusions:
CCD and C5FD are obviously better than curcumin in water solubility, and have higher solubility.
The water solubility of C5FD is slightly higher than that of CCD, which is likely to be beneficial for further biological application and drug development.
From experimental data we can see the advantage of CCD and C5FD over curcumin in terms of water solubility, which will help both curcumin derivatives to play a greater role in biological and medical applications.
Test example 3
The purpose of the experiment is as follows: the antioxidant properties of Curcumin, curcumin cytosine derivative (CCD for short) and Curcumin 5-fluorocytosine derivative (C5 FD for short) were compared.
Experiment design: antioxidant capacity was measured using DPPH radical scavenging method.
The experimental steps are as follows:
preparation of reagents and apparatus:
curcumin
Curcumin Cytosine Derivative (CCD)
Curcumin 5-fluorocytosine derivative (C5 FD)
0.1 mM DPPH solution (methanol solvent)
Methanol
Test tube
Spectrophotometer
The experimental steps are as follows:
step 1: curcumin, CCD and C5FD were dissolved in methanol to prepare sample solutions with concentrations of 0.1, 0.5, 1.0, 2.0 and 4.0. 4.0 mM, respectively.
Step 2: 3mL of 0.1 mM DPPH solution was added to each tube containing 1mL of sample solution.
Step 3: the solution in the tube was reacted at room temperature for 30 minutes in the dark.
Step 4: after the end of the reaction, the absorbance of each cuvette solution at 517nm was measured using a spectrophotometer.
Step 5: DPPH radical scavenging was calculated for each sample.
Experimental data and conclusions:
sample of 0.1 mM 0.5 mM 1.0 mM 2.0 mM 4.0 mM
Curcumin 15% 40% 60% 75% 85%
CCD 20% 45% 65% 80% 90%
C5FD 18% 43% 63% 78% 88%
From experimental data, we can observe that curcumin, CCD and C5FD all exhibit a certain antioxidant capacity at different concentrations. Meanwhile, we can see that the DPPH free radical clearance of CCD and C5FD at various concentrations is slightly higher than that of curcumin, which indicates that the oxidation resistance of the CCD and the C5FD is slightly better than that of curcumin.
Test example 4
1 pretreatment of ligands
Pretreatment of ligand: running Autodock software, opening a PDB file of the Ligand pyrimidyl curcumin derivative, clicking the PDB file of the Ligand in the Ligand-Input-Open, automatically adding hydrogen atoms by Autodock, calculating point charges, adding atom types, and clicking in a dialog box for determination. After pretreatment. The Ligand molecules are stored in Ligand-Output-Saveaspdbqt as a file of pdbqt suffix for later use. The PDB file graph before ligand treatment and the PDBQT file graph after ligand treatment are shown in figures 1 and 2.
2 setting of butt GRID
The Autodock software is opened, the pdbqt file of the G-quadruplex is run inside the Grid-macromolecules-Open, and the determination is performed by clicking in a dialog box. The pdbqt file of cytosine curcumin derivatives was then opened inside Grid-SetmapTypes-OpenLigand.
And opening Grid-GridBox-GridOptions, and adjusting the dimensions of the docking points, the X plane, the Y plane and the Z plane in the dialog box to be 64, 72, 78. The X plane, the Y plane and the Z plane respectively, wherein the central coordinates of the X plane, the Y plane and the Z plane are 0.056,1.000-1.806 respectively. After the end, click File-ClosesavingCurrent in GridOption dialog, store the lattice point and close the dialog. Next, click on Grid-Output-Savegpf, stored as gpf file for later use. The information and the image of the setting are as shown in fig. 3 and 4.
3 docking and results
After Grid setting is completed, an autoprid calculation is performed in Run-runautogorid.
Three spaces in the pop-up dialog require, in order from top to bottom, auto grid computing software, gpf files automatically generated when starting to set up grid points, and glg files generated simultaneously with gpf files. After that, clicking on the counth will automatically start to calculate autogrid.
The pdqt file of the previously processed receptor molecule is run in the locking-macromolecules-setrig and then the pdqt file of the previously processed ligand molecule is opened in the locking-ligand-open and the Accept is clicked in a pop-up dialog.
Opening the dock-search parameters-genetics generic parameters, then popping up the Docking parameters, using the default parameters of the system, clicking Accept in this test. Opening click on dock-dock parameters-setdock parameters directly clicks on Accept after parameter adjustment is completed.
And then storing the dpf file generated by the dock, and opening the dock-Output-Lamarckian Ga storage file.
The calculation of autodock is started inside Run-RunAutodock. The three blanks in the pop-up dialog box require from top to bottom that an autodock running file, a dpf file generated in the dock, and a dlg file generated together when the dpf file is generated are added. After that, clicking on the counth starts to calculate autoprid.
The dlg file stored while running Docking in Analyze-Docking-Open is then determined by clicking in the pop-up dialog box.
And loading the butted result and the molecular conformation into a graphic window by analysis-formats-Load, and then clicking the conformation sequence number of the corresponding molecule in the list in a pop-up dialog box, wherein the butted data of the molecular conformation is displayed in a display window at the top. At this time, a double click is selected to load the molecular conformation into the molecular display window for subsequent observation and analysis.
The analysis-formats-Play will pop up a dialog box controlling the Play. When the third button is clicked, the following dialog box appears, if the ShowInfo is hooked, the related information of the current sub-conformation can be displayed, and the drop-down menu of Colorby is hooked into vdw, the conformation of coloring according to the Van der Waals force can be obtained.
A Receptor rigid molecule is added to the Analyze-macromolecules-Open, so that the situation of the Ligand in the Receptor molecule can be seen.
All molecular conformational results generated by docking in Analyze-docks-ShowsSphere are shown in pellet form. And obtaining the result of the molecular docking experiment. The following are provided: FIG. 5 is a graph showing the conformational information of the docking, FIG. 6 is an image of the molecular conformation stained according to Van der Waals forces, FIG. 7 is an image of the conformational center of the molecular docking, and FIG. 8 is a graph showing the characterization of the docking conformation and the effect of interactions with receptor residues under computer modeling. From the above results, it was found that the 5-fluorocytosine curcumin derivative had 1 active site for docking with the G-quadruplex.
G-quadruplexes, which are a square formed by the interactive binding of 4 guanines as a basis, are transient structures, present in large numbers in the cell to be divided, and occur in the chromosome nucleus and chromosome terminal (which can protect the chromosome from damage). Because cancer cells divide very rapidly, defects often occur at the end of the chromosome, and the quadruple helix DNA molecule may be present only in cancer cells.
The results of test example 4 therefore demonstrate that the compounds of the present invention have:
anticancer potential: because g-quadruplexes are ubiquitous in cancer cells, compounds that specifically interact with this structure may have anticancer potential.
Specificity: the specific interaction of the compound with the g-quadruplex may make it more targeted in the cell and produce fewer non-specific side effects. This makes it possible to adapt the medicament more easily to clinical applications and reduces the risk of treatment.
Treatment strategy: based on the specific conditions of presence of g-quadruplexes, the discovery of this compound may drive relevant therapeutic strategies and approaches. This may also provide new directions and ideas for cancer treatment and clinical research.
The foregoing description of the embodiments of the present application is illustrative, not exhaustive, and not limited to the embodiments disclosed. Many modifications and variations will be apparent to those of ordinary skill in the art without departing from the scope and spirit of the various embodiments described. The terminology used herein was chosen in order to best explain the principles of the embodiments, the practical application, or the improvement of technology in the marketplace, or to enable others of ordinary skill in the art to understand the embodiments disclosed herein.

Claims (1)

1. Application of compound shown in formula (I) or (II) or pharmaceutically acceptable salt thereof in preparation of antioxidant drugs
(l);
(Il)。
CN202310683998.9A 2023-04-11 2023-04-11 Application of medicinal compound Pending CN116617229A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310683998.9A CN116617229A (en) 2023-04-11 2023-04-11 Application of medicinal compound

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202310683998.9A CN116617229A (en) 2023-04-11 2023-04-11 Application of medicinal compound
CN202310381929.2A CN116143705B (en) 2023-04-11 2023-04-11 A pharmaceutical compound

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN202310381929.2A Division CN116143705B (en) 2023-04-11 2023-04-11 A pharmaceutical compound

Publications (1)

Publication Number Publication Date
CN116617229A true CN116617229A (en) 2023-08-22

Family

ID=86362080

Family Applications (3)

Application Number Title Priority Date Filing Date
CN202310683998.9A Pending CN116617229A (en) 2023-04-11 2023-04-11 Application of medicinal compound
CN202310381929.2A Active CN116143705B (en) 2023-04-11 2023-04-11 A pharmaceutical compound
CN202310684003.0A Pending CN116891436A (en) 2023-04-11 2023-04-11 Preparation method of medicinal compound

Family Applications After (2)

Application Number Title Priority Date Filing Date
CN202310381929.2A Active CN116143705B (en) 2023-04-11 2023-04-11 A pharmaceutical compound
CN202310684003.0A Pending CN116891436A (en) 2023-04-11 2023-04-11 Preparation method of medicinal compound

Country Status (1)

Country Link
CN (3) CN116617229A (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116444522B (en) * 2023-06-14 2023-08-25 齐泽(云南)生物科技有限公司 Curcumin adenine derivative compound
CN116496277B (en) * 2023-06-14 2023-08-25 齐泽(云南)生物科技有限公司 Curcumin 2, 6-diaminopurine derivative compound

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102005039579B4 (en) * 2005-08-19 2022-06-30 Magforce Ag Method for introducing therapeutic substances into cells
CN101570515B (en) * 2009-06-12 2011-10-19 乐山师范学院 Curcumin substituted uracil derivative, preparation method and use thereof
WO2011142795A1 (en) * 2010-03-25 2011-11-17 Wayne State University Novel analogs of curcumin and methods of use
WO2011132171A1 (en) * 2010-04-23 2011-10-27 Piramal Life Sciences Limited Nitric oxide releasing prodrugs of therapeutic agents
WO2012145420A1 (en) * 2011-04-18 2012-10-26 Rutgers, The State University Of New Jersey Composition, synthetic process and use of new antioxidant inflammation modulators
AU2016284816B2 (en) * 2015-06-25 2021-10-07 Syn-Nat Products Enterprise LLC Pharmaceutical co-crystal composition and use thereof
CN110028415A (en) * 2018-01-11 2019-07-19 南京诺瑞特医药科技有限公司 Curcumin derivate and application thereof
EP3581172B1 (en) * 2018-06-14 2023-12-20 TFLL Pharmaceutical Food Supplements and Cosmetic Products - Industry Foreign Trade A pharmaceutical composition with enhanced bioavailability containing a medical herbal extract
CN113773315A (en) * 2020-06-10 2021-12-10 无锡佰翱得生物科学有限公司 Highly selective deuterated inhibitors of cyclin dependent kinase 2(CDK2)
CN112209887B (en) * 2020-09-30 2021-09-03 天津理工大学 Eutectic crystal of 5-fluorouracil and kaempferol and preparation method thereof

Also Published As

Publication number Publication date
CN116143705B (en) 2023-06-30
CN116143705A (en) 2023-05-23
CN116891436A (en) 2023-10-17

Similar Documents

Publication Publication Date Title
CN116143705B (en) A pharmaceutical compound
CN116082251B (en) A pharmaceutical compound
CN111148729A (en) Crystal form of benvitimod, application and preparation method thereof
RU2468015C2 (en) Polymorphic forms of deferasirox (icl670a)
US10358424B2 (en) Sodium salt of uric acid transporter inhibitor and crystalline form thereof
CN112898371B (en) Panaxatriol compounds, preparation method and medical application thereof
JP2000319290A (en) New platinum (iv) complex and its production
WO2010009739A1 (en) Copper(i)chloride complex of nicotinic acid and pharmaceutical compositions containing the same.
CN116444522B (en) Curcumin adenine derivative compound
CN105001195A (en) New crystal form of R(+)-thioctic acid-L-lysinate and preparation method thereof
CN116496277B (en) Curcumin 2, 6-diaminopurine derivative compound
JP5203644B2 (en) Compounds derived from Benicus mushroom for use in the treatment of autoimmune diseases
CN116836165A (en) Curcumin 2-chloroadenine derivative compound
CN116987082A (en) Curcumin guanine derivative compound
CN112500344B (en) Crystalline form of roxasistat and preparation method thereof
BE1001251A4 (en) Pharmaceutical composition containing organic complex zinc and method for preparing the active product.
CN108125963A (en) Polaprezinc rectally preparation and the application for preparing ulcerative colitis drug
CN108553649B (en) Novel sorafenib-gamma-cyclodextrin inclusion compound with pipeline structure, preparation method and application
CN112159448A (en) Ethinylestradiol pharmaceutical co-crystal and preparation method and application thereof
CN102351881B (en) Stable levofloxacin hydrochloride compound
EP2271343A1 (en) Salts of 2-substituted quinolines
CN106478616B (en) Crystalline form of GPR40 agonist and preparation method thereof
CN111574435B (en) 4,4' -dipyridyl methylpyrazine derivative eutectic crystal
BE1028852B1 (en) 5α-Hydroxy-6β-[2-(1H-imidazol-4-yl)ethylamino]cholestan-3β-ol PRODRUG AND PHARMACEUTICAL COMPOSITIONS COMPRISING THEM FOR USE IN THE TREATMENT OF CANCER
WO2017076307A1 (en) Taccalonolide compound cyclodextrin inclusion compound, 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