CN111848690A - Anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and preparation method thereof - Google Patents

Anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and preparation method thereof Download PDF

Info

Publication number
CN111848690A
CN111848690A CN202010827724.9A CN202010827724A CN111848690A CN 111848690 A CN111848690 A CN 111848690A CN 202010827724 A CN202010827724 A CN 202010827724A CN 111848690 A CN111848690 A CN 111848690A
Authority
CN
China
Prior art keywords
platinum complex
tetravalent platinum
anticancer
immune escape
precipitate
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
CN202010827724.9A
Other languages
Chinese (zh)
Inventor
王晓勇
金素星
郭子建
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.)
Nanjing University
Original Assignee
Nanjing University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanjing University filed Critical Nanjing University
Priority to CN202010827724.9A priority Critical patent/CN111848690A/en
Publication of CN111848690A publication Critical patent/CN111848690A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F15/00Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic System
    • C07F15/0006Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic System compounds of the platinum group
    • C07F15/0086Platinum compounds
    • C07F15/0093Platinum compounds without a metal-carbon linkage
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The invention provides an anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and a preparation method thereof, and particularly relates to the field of biological medicine, wherein the molecular structure of the anticancer tetravalent platinum complex is as follows:
Figure DDA0002636832890000011
the synthesis method comprises the following steps: s1, mixing the divalent platinum complex with hydrogen peroxide H2O2Reacting to obtain a tetravalent platinum complex; s2 reaction of tetravalent platinum complex obtained in S1 with
Figure DDA0002636832890000012
Mixing O-benzotriazole-N, N, N ', N' -tetramethylurea tetrafluoroborate (TBTU) and Triethylamine (TEA) in a N, N-Dimethylformamide (DMF) solution, and stirring and mixing uniformly; s3, after the reaction is finished, collecting filtrate, concentrating to 0.5-5 mL, dropwise adding a mixture of ethanol and water, centrifuging and collecting precipitate; and dissolving the precipitate in methanol, washing the precipitate with ether for 3 times, and vacuum drying to obtain the target product. The tetravalent platinum complex has strong toxic activity on tumor cells, inhibits inflammation and immune escape through a plurality of action targets, and shows excellent anti-tumor effect on living bodies.

Description

Anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and preparation method thereof
Technical Field
The invention belongs to the field of biological medicines, and particularly relates to an anti-cancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and a preparation method thereof.
Background
Platinum antineoplastic drugs have become the most widely used metal drugs in clinic, and the first generation of platinum antineoplastic drugs represented by Cisplatin (CDDP) has played a leading role in the treatment of various solid tumors such as head and neck cancer, malignant pleural mesothelioma and the like since being approved by FDA for the treatment of testicular cancer and ovarian cancer. However, severe side effects can occur during treatment due to the strong cytotoxicity of cisplatin. In order to overcome the defects and reduce the systemic toxicity of cisplatin, the second-generation platinum antitumor drugs carboplatin (1989), nedaplatin (Japan), the third-generation platinum antitumor drugs oxaliplatin (2002), leplatin (China) and heptaplatin (Korea) were developed. Compared with cisplatin, carboplatin has great improvement on the aspect of relieving renal toxicity, the action mechanism of oxaliplatin is changed due to structural change, cross drug resistance with cisplatin is avoided, and toxic and side effects are reduced, so that the anticancer spectrum of platinum drugs is widened. Statistically, over 50% of the current anti-tumor chemotherapy regimens in clinical tumor treatment are based on or include platinum drugs. However, platinum antineoplastic drugs still face some challenges, such as high systemic toxicity, drug resistance, low selectivity and the like, and although the toxic and side effects of the second and third-generation platinum drugs are reduced, the problems are not solved fundamentally because the platinum antineoplastic drugs are analogues of cisplatin. Therefore, it is necessary to develop a novel platinum anticancer complex to overcome the defects of the conventional platinum drugs.
Inflammation is closely related to angiogenesis, immune tolerance, metastasis, invasion and drug resistance of tumors. The non-steroidal anti-inflammatory drug is an important bridge for connecting cancer and inflammation, and can reduce the risk and the death rate of the cancer; inhibition of cyclooxygenase (COX-2) and blocking of the prostaglandin cascade are the major causes of cancer inhibition. In addition, programmed death ligand 1(PD-L1) plays a key role in tumor evasion immune surveillance, and increased expression of PD-L1 on the surface of cancer cells and resultant T cell suppression contribute to immune escape of cancer cells. It has been found that COX-2 and prostaglandin (PGE2) have effects of promoting tumor inflammation and immunosuppression, and COX-2 can regulate the expression of PD-L1 in various cancers, so that inhibition of COX-2 can also affect PD-L1, thereby possibly promoting the immune response of cancer cells. Meanwhile, the inhibition of inflammation and immune escape provides a new idea for designing anti-tumor drugs and also provides an opportunity for obtaining novel multi-specific platinum drugs.
Disclosure of Invention
The invention aims to provide an anti-cancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and a preparation method thereof, the complex has excellent cytotoxic activity on tumor cells, plays an anti-tumor function through multi-specificity action from the aspects of inhibiting inflammation (inflammatory factors and related biological enzyme activity) and improving immune check point blocking treatment, and shows excellent anti-tumor effect and better anti-inflammatory and immunoregulation functions on a living body.
The invention provides the following technical scheme:
an anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape, which has the following molecular structure:
Figure BDA0002636832870000021
it is preferable that
Figure BDA0002636832870000031
The structure of (1) is as follows:
Figure BDA0002636832870000032
Figure BDA0002636832870000033
a preparation method of an anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape comprises the following steps:
s1, mixing the divalent platinum complex with hydrogen peroxide H2O2Reacting to obtain a tetravalent platinum complex;
wherein the divalent platinum complex is
Figure BDA0002636832870000034
Figure BDA0002636832870000035
S2 reaction of tetravalent platinum complex obtained in S1 with
Figure BDA0002636832870000041
Mixing O-benzotriazole-N, N, N ', N' -tetramethylurea tetrafluoroborate (TBTU) and Triethylamine (TEA) in a N, N-Dimethylformamide (DMF) solution, and stirring and mixing uniformly;
s3, after the reaction is finished, collecting filtrate, concentrating to 0.5-5 mL, dropwise adding a mixture of ethanol and water, centrifuging and collecting precipitate; and dissolving the precipitate in methanol, washing the precipitate with ether for 1-10 times, and vacuum drying to obtain the target product.
Preferably, in the step S2, the mixture is stirred and mixed uniformly under the protection of nitrogen.
Preferably, in the step S3, the solution is concentrated to 1mL by rotary evaporation; the precipitate was washed 3 times with diethyl ether.
The invention has the beneficial effects that:
the tetravalent platinum complex has excellent antitumor activity in vivo and in vitro, treats cancer from two aspects of inhibiting inflammation and blocking immune check points, has a plurality of action targets participating in an antitumor process, and has an action mechanism different from that of the traditional platinum drugs. The derivatives also comprise stereoisomers or pharmaceutically acceptable salts thereof, and preparations prepared by taking the derivatives as active ingredients or main active ingredients and auxiliary pharmaceutically acceptable auxiliary materials.
Drawings
The accompanying drawings, which are included to provide a further understanding of the invention and are incorporated in and constitute a part of this specification, illustrate embodiments of the invention and together with the description serve to explain the principles of the invention and not to limit the invention. In the drawings:
FIG. 1 shows the preparation of the compound of example 1 of the present invention1H-NMR spectrum (DMSO-d)6,400MHz);
FIG. 2 shows the preparation of the compound of example 1 of the present invention13C-NMR spectrum (DMSO-d)6,101MHz);
FIG. 3 shows the preparation of the compound of example 1 of the present invention195Pt-NMR Spectrum (DMSO-d)6,86MHz);
FIG. 4 is a graph showing the relative amounts of the compounds prepared in example 1 of the present invention and the inflammatory factors IL-6 and IL-1. beta. in mouse macrophage RAW264.7 after stimulation with LPS;
FIG. 5 shows prostaglandin E of breast cancer cells MCF-7 by the compound prepared in example 1 of the present invention2Inhibitory effects of secretion;
FIG. 6 shows the inhibitory effect of the compound prepared in example 1 of the present invention on cyclooxygenase COX-2 in breast cancer cells MCF-7;
FIG. 7 shows the inhibitory effect of the compound prepared in example 1 on PD-L1 in breast cancer cells MDA-MB-231;
FIG. 8 is a graph showing the inhibition of Balb/C nude mice carrying MDA-MB-231 transplanted breast cancer by the compound prepared in example 1 and cisplatin according to the present invention;
FIG. 9 is a graph showing the effect of the compound prepared in example 1 of the present invention and cisplatin on the volume of mouse MDA-MB-231 transplanted breast cancer;
FIG. 10 is a graph showing the effect of the compound prepared in example 1 of the present invention and cisplatin on body weight during MDA-MB-231 transplantation breast cancer tumor-bearing mice treatment;
FIG. 11 is an image of an immunohistochemical section of the compound prepared in example 1 of the present invention and cisplatin on inflammatory and immune-related proteins in mouse MDA-MB-231 transplanted breast cancer tissues;
FIG. 12 is a schematic diagram of the mechanism of action of the compound prepared in example 1 of the present invention.
Detailed Description
Example 1
Synthesis of tetravalent platinum complexes
Figure BDA0002636832870000061
S1, dropwise adding hydrogen peroxide (30 wt%, 20mL) into a cisplatin (CDDP, 400mg,1.33mmol) aqueous solution (12mL), refluxing and stirring at 60 ℃ for 4h, after the reaction is finished, cooling the bright yellow solution at room temperature overnight to obtain pale yellow crystals, filtering, washing with ice water, and drying in vacuum to obtain a tetravalent Oxoplatin yellow powdery product;
s2, mixing oxyplatin (100mg, 0.30mmol) with
Figure BDA0002636832870000062
(NPX, 207.23mg, 0.90mmol), TEA (125. mu.L, 0.90mmol) and TBTU (288.99mg, 0.90mmol) were mixed in DMF (15mL) and stirred for 48h, protected from light.
S3, after the reaction is finished, adding a mixture of ethanol and water into the mixture, and precipitating a yellow crude product. The precipitate was dissolved in methanol, washed several times with ether and dried in vacuo to give a pale yellow powder with a yield of about 65%.
EXAMPLE 2 cytotoxic Activity assay of Compounds
The compound prepared in the embodiment 1 of the invention is subjected to cytotoxic activity test, breast cancer cells (MFC-7, MDA-MB-231, MDA-MB-435), ovarian cancer cells (Caov-3) and cervical cancer cells (HeLa) are respectively used as models, the compound synthesized in the embodiment 1 and cisplatin are used as the substances to be detected, after the substances to be detected act on the cells, the survival condition of the cells is observed, and the survival rate of the cells is tested by a thiazole blue (MTT) method, and the specific operation steps are as follows:
s1, collecting the cells in the logarithmic phase, adjusting the concentration of the cell suspension, and adding the cells into a 96-well plate, wherein the number of the cells in each well is about 5000;
s2, placing the experimental cells at 37 ℃ and 5% CO2Culturing for 18h in the cell culture box;
s3, preparing compounds or cisplatin solutions with different concentration gradients by using a complete culture medium containing 10% FBS respectively, adding the compounds or cisplatin solutions into the 96-well plate, setting 3 parallel multiple wells for each concentration, and incubating the cells for 48 hours by using the medicine;
s4, adding MTT solution (5mg/mL, 20 mu L) into each well, continuously incubating for 4h, absorbing the culture medium, adding DMSO (150 mu L) into each well, and shaking the shaking table at a low speed for 10 min;
s5, detecting the absorbance value (O.D.) of each hole at 570nm by using a microplate reader;
s6, setting the zero setting wells (medium, MTT, DMSO) and the control group (cell, medium, DMSO, MTT) at the same time, and calculating the cell survival rate according to the following formula:
survival (%) - (o.d.(sample)-O.D.(blank))/(O.D.(control)-O.D.(blank))]×100%
Wherein, O.D.(sample)Is the absorbance value of the sample group cells; and (D) O.D.(blank)The absorbance value of the withered holes is obtained; and (D) O.D.(control)The absorbance value of the control group cells;
s7, making cell survival rate-concentration curve, calculating half Inhibitory Concentration (IC) of compound50) The results are shown in Table 1.
Table 1 shows the compounds prepared in example 1 of the present invention and the IC of cisplatin on various cells50Value (μ M,48h)
Name of drug MCF-7 MDA-MB-435 MDA-MB-231 Caov-3 HeLa
Complex 0.17±0.04 0.34±0.09 0.16±0.01 0.45±0.06 0.22±0.01
CDDP 4.00±1.00 8.34±0.09 29.98±1.10 24.00±2.50 4.50±0.50
EXAMPLE 3 detection of anti-inflammatory Capacity of Compounds
The compound prepared in the embodiment 1 of the invention is subjected to anti-inflammatory capability detection, a mouse macrophage RAW264.7 and a human breast cancer cell MCF-7 are selected as models, the compound synthesized in the embodiment 1 and cisplatin are used as objects to be detected, after the objects to be detected act on cells, an ELISA kit is used for detecting the relative content of inflammatory factors IL-6 and IL-1 beta in the supernatant of the RAW264.7 cells, and an immunoblotting test is used for detecting the change of COX-2 protein in the MCF-7 cells and the relative content of PGE2 in the cell supernatant, and the specific operation steps are as follows:
s1, collecting the logarithmic phase cells, adjusting the concentration of the cell suspension to 2X 10 per well5The density of individual cells was seeded in 6-well plates;
s2, placing the experimental cells at 37 ℃ and 5% CO2Culturing for 18h in the cell culture box;
s3, stimulating RAW264.7 cells with lipopolysaccharide (LPS, 100ng/mL) for 6 h; compound or cisplatin (0.4. mu.M) was then added to the cell culture medium and incubation continued for 18 h. The contents of inflammatory factors IL-6 and IL-1 beta in the supernatant were measured according to the protocol provided by the ELISA kit, and the results are shown in FIG. 4;
s4, for the MCF-7 cells, after the cells grow adherent to the walls for 24 hours, adding 2mL of complete culture medium containing the compound or the cisplatin into each hole again, and continuing to culture for 36 hours respectively;
s5, collecting cell supernatant after finishing the culture, carrying out ELISA test, and detecting the content of PGE2, wherein the result is shown in figure 5;
s6, digesting and collecting adherent cells by pancreatin, and centrifuging to obtain cell sediment;
s7, using the prepared RIPA cell lysate to lyse the cells, collecting the supernatant in a clean EP tube, determining the protein concentration in the tube, and adjusting the protein concentration of the sample to be the same according to the protein concentration. The expression quantity of the target protein in the collected protein is determined by immunoblotting test, namely, a sample protein is subjected to an electrophoresis experiment by using 10% SDS-PAGE gel according to the concentration of 40 mu g per hole, the electrophoresis is stopped after the target protein is properly separated, the protein is transferred onto a polyvinylidene fluoride (PVDF) membrane, the membrane is rotated for 1.5h under the conditions of 100V and 200mA, the protein membrane is placed in 5% skimmed milk powder which is prepared in advance for sealing for 0.5h after the membrane is transferred, then primary antibody solution is prepared according to the dilution ratio of COX-2 antibody and is incubated with the PVDF membrane containing the detected protein at 4 ℃ for overnight, after the incubation is finished, the primary antibody is washed by using PBST (PBS + Tween-20), and then secondary antibody is continuously incubated by using goat anti-rabbit HRP marked by peroxidase. After 1h, the secondary antibody was washed away, the PVDF membrane was further washed three times with PBST, and the result of exposure and photographing by using a chemiluminescence detection system was shown in FIG. 6.
EXAMPLE 4 testing of the immunomodulatory Capacity of Compounds
The compound prepared in the embodiment 1 of the invention is subjected to immunoregulatory capacity detection, a human breast cancer cell MDA-MB-231 is selected as a model, the compound synthesized in the embodiment 1 and cisplatin are used as a substance to be detected, and after the substance to be detected acts on cells, the change of PD-L1 protein in the cells is detected by an immunofluorescence assay, and the specific operation steps are as follows:
s1, collecting the logarithmic phase cells, adjusting the concentration of the cell suspension, and paving the cells in a 20mm confocal small dish;
s2, placing the experimental cellsAt 37 ℃ with 5% CO2Culturing for 18h in the cell culture box;
s3, adding 500 mu L of complete culture medium containing the compound or the cisplatin into each hole again, and continuing to culture for 36h respectively;
s4, the medium was removed and washed three times with pre-cooled PBS. Fixing the cells with 4% paraformaldehyde for 20min (4 deg.C), allowing PBS solution containing 1 ‰ Triton X-100 to permeate for 20min, and sealing with 5% skimmed milk powder solution at 4 deg.C for 30 min;
s5, the cells were washed three times with PBS and incubated overnight at 4 ℃ with diluted PD-L1 primary antibody solution. Followed by three washes with PBS for 5min each; incubating with a secondary antibody FITC-conjugated anti-rabbitIgG (Invitrogen) for 1h at room temperature;
s6, continuously washing the cells for three times by using PBS, wherein each time lasts for 5 min; the cells were then incubated with Hoechst3342 for 10min, washed 3 times with PBS, and finally observed for expression of PD-L1 protein on the cells by fluorescence microscopy, as shown in FIG. 7.
In vivo tumor inhibition assay for the Compounds of example 5
Subcutaneously planting human breast cancer cell MDA-MB-231(5 multiplied by 10) into Balb/c nude mice6cells/mouse), after the tumor formation, randomly grouping the mice, and each group comprises 5 mice;
the mice were administered tail vein with cisplatin and compound at a dose of 1.5mg Pt/kg every three days while tumor volume was measured and mouse weight was weighed, and the results are shown in fig. 8, 9 and 10.
EXAMPLE 6 immunohistochemical detection of Compounds
Tumor tissues of the mice in example 5 were respectively excised, fixed in 4% paraformaldehyde, washed, dehydrated, waxed, embedded, and subjected to immunohistochemical experiments after antigen retrieval, and the inflammatory and immune-related proteins COX-2 and PD-L1 were respectively detected in the tumor tissues treated with the compound or cisplatin, and the results are shown in fig. 11.
The tetravalent platinum complex has the characteristics of dynamic inertia, low reaction activity, small toxic and side effects and the like. Currently, 4 tetravalent platinum complexes have entered clinical trials, and the most representative tetravalent platinum prodrug, Satraplatin, has entered phase III clinical trials. In the process of generating and developing tumors, angiogenesis, immune tolerance, metastasis, invasiveness, drug resistance and the like are accompanied by inflammatory processes; in the inflammatory microenvironment of tumor cells, a number of proinflammatory cytokines are also involved in the immune escape of tumors, where cyclooxygenase, a key enzyme involved in the inflammatory response, regulates the expression of PD-L1 in many types of cancers. The tetravalent platinum complex can inhibit the growth of tumor cells from two aspects of inflammation inhibition and immune escape, and plays an anti-tumor effect by intervening the biological functions of a plurality of potential targets, and the action mechanism of the tetravalent platinum complex is shown in figure 12. The derivative also comprises a stereoisomer or pharmaceutically acceptable salt thereof, and other dosage forms prepared by taking the derivative as an active ingredient or a main active ingredient and adding pharmaceutically acceptable auxiliary materials.
Although the present invention has been described in detail with reference to the foregoing embodiments, those skilled in the art will understand that various changes, modifications and substitutions can be made without departing from the spirit and scope of the invention as defined by the appended claims. Any modification, equivalent replacement, or improvement made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (5)

1. An anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape is characterized in that the molecular structure is as follows:
Figure FDA0002636832860000011
2. an anti-cancer tetravalent platinum complex according to claim 1, capable of inhibiting inflammation and immune escape, characterized in that:
Figure FDA0002636832860000012
the structure of (1) is as follows:
Figure FDA0002636832860000013
Figure FDA0002636832860000014
3. a preparation method of an anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape is characterized by comprising the following steps: s1, mixing the divalent platinum complex with hydrogen peroxide H2O2Reacting to obtain a tetravalent platinum complex;
wherein the divalent platinum complex is
Figure FDA0002636832860000015
Figure FDA0002636832860000021
S2 reaction of tetravalent platinum complex obtained in S1 with
Figure FDA0002636832860000022
Mixing O-benzotriazole-N, N, N ', N' -tetramethylurea tetrafluoroborate (TBTU) and Triethylamine (TEA) in a N, N-Dimethylformamide (DMF) solution, and stirring and mixing uniformly;
s3, after the reaction is finished, collecting filtrate, concentrating to 0.5-5 mL, dropwise adding a mixture of ethanol and water, centrifuging and collecting precipitate; and dissolving the precipitate in methanol, washing the precipitate with ether for 1-10 times, and vacuum drying to obtain the target product.
4. The method of claim 3, wherein the preparation of the tetravalent platinum complex for anticancer capable of suppressing inflammation and immune escape comprises: in step S2, stirring and mixing under nitrogen protection.
5. The method of claim 3, wherein the preparation of the tetravalent platinum complex for anticancer capable of suppressing inflammation and immune escape comprises: in the step S3, concentrating to 1mL by rotary evaporation; the precipitate was washed 3 times with diethyl ether.
CN202010827724.9A 2020-08-17 2020-08-17 Anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and preparation method thereof Pending CN111848690A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010827724.9A CN111848690A (en) 2020-08-17 2020-08-17 Anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and preparation method thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010827724.9A CN111848690A (en) 2020-08-17 2020-08-17 Anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and preparation method thereof

Publications (1)

Publication Number Publication Date
CN111848690A true CN111848690A (en) 2020-10-30

Family

ID=72968767

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010827724.9A Pending CN111848690A (en) 2020-08-17 2020-08-17 Anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and preparation method thereof

Country Status (1)

Country Link
CN (1) CN111848690A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113336801A (en) * 2021-06-08 2021-09-03 中国人民解放军空军军医大学 Tetravalent platinum complex containing BET inhibitor and application
CN113416216A (en) * 2021-08-23 2021-09-21 江苏南创化学与生命健康研究院有限公司 High-activity Pt complex and preparation method and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
金素星: "靶向能量代谢和炎症因子的铂类抗癌前药设计及性能研究", 《中国博士学位论文全文数据库-医药卫生科技辑》 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113336801A (en) * 2021-06-08 2021-09-03 中国人民解放军空军军医大学 Tetravalent platinum complex containing BET inhibitor and application
CN113336801B (en) * 2021-06-08 2023-08-22 中国人民解放军空军军医大学 Tetravalent platinum complexes containing BET inhibitors and use thereof
CN113416216A (en) * 2021-08-23 2021-09-21 江苏南创化学与生命健康研究院有限公司 High-activity Pt complex and preparation method and application thereof
CN113416216B (en) * 2021-08-23 2021-10-29 江苏南创化学与生命健康研究院有限公司 High-activity Pt complex and preparation method and application thereof

Similar Documents

Publication Publication Date Title
CN111205331B (en) Anti-tumor tetravalent platinum complex with anti-drug resistance function and preparation method thereof
CN111039990B (en) Low-toxicity iridium complex and synthesis method and application thereof
CN110950914B (en) Iridium complex and synthesis method and application thereof
CN111848690A (en) Anticancer tetravalent platinum complex capable of inhibiting inflammation and immune escape and preparation method thereof
CN107629089A (en) Tacrine platinum (II) complex of high activity and its synthetic method and application
CN113024584A (en) 8-hydroxyquinoline complex for treating lung cancer and preparation method thereof
CN107602367B (en) 4 '-hydroxy-3' -methoxychalcone derivative, preparation method thereof and application thereof in tumor resistance
CN112010839A (en) Crystalline forms of a targeted silk/threonine kinase inhibitor
CN111377975B (en) Novel mitochondrion-targeted iridium complex and preparation method and application thereof
CN110305166B (en) Ruthenium (II) complex with curcumin derivative as ligand and preparation method and application thereof
CN110041375A (en) Compound, preparation method and its application in preparation of anti-tumor drugs with asymmetric monosubstituted naphthalimide tetravalence platinum structure
CN109675053A (en) Targeting preparation of Podophyllotoxin and its derivatives and preparation method thereof
CN107573318A (en) A kind of new gossypol Schiff bases derivative and its synthetic method for having antitumor activity
CN114605475B (en) Oral Pt (IV) anticancer prodrugs containing 3-bromopyruvate ligand axially
CN111072725A (en) Compound with naproxen tetravalent platinum structure, preparation method and application thereof in preparation of antitumor drugs
CN102827116A (en) Alpha-aryl-gamma-methylene butene lactone compounds, and synthesis method and application thereof
CN108017608B (en) Flavone derivatives, preparation method and application thereof
CN110078770B (en) Compound with quinolinone tetravalent platinum structure, preparation method and application thereof in preparation of antitumor drugs
CN113735781A (en) Copper complex and preparation method and application thereof
CN109867708B (en) Preparation method and application of glycyrrhetinic acid series derivatives (TNGA-X) with anti-tumor effect
CN111393318A (en) Synthesis of novel sildenafil acid amide derivative and application of novel sildenafil acid amide derivative in antitumor drugs
CN114891044B (en) Tetravalent platinum complex with anti-tumor activity and preparation method and application thereof
CN103833766B (en) Shore, Agra dimethylamine fumarate and the purposes in prepared by medicine thereof
CN115340577B (en) Synthesis method of platinum (II) complex and application of platinum (II) complex in antitumor drugs
CN113683645B (en) Binuclear Pt (II) complex with anti-tumor activity 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
RJ01 Rejection of invention patent application after publication

Application publication date: 20201030

RJ01 Rejection of invention patent application after publication