CN112266396A - Integrated prodrug based on bio-orthogonal chemistry, preparation method and medical application thereof - Google Patents

Integrated prodrug based on bio-orthogonal chemistry, preparation method and medical application thereof Download PDF

Info

Publication number
CN112266396A
CN112266396A CN202011077271.9A CN202011077271A CN112266396A CN 112266396 A CN112266396 A CN 112266396A CN 202011077271 A CN202011077271 A CN 202011077271A CN 112266396 A CN112266396 A CN 112266396A
Authority
CN
China
Prior art keywords
compound
cells
cisplatin
tumor
preparation
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.)
Granted
Application number
CN202011077271.9A
Other languages
Chinese (zh)
Other versions
CN112266396B (en
Inventor
黄张建
张奕华
孙涛
吴建兵
朱杰
朱明超
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.)
China Pharmaceutical University
Original Assignee
China Pharmaceutical 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 China Pharmaceutical University filed Critical China Pharmaceutical University
Priority to CN202011077271.9A priority Critical patent/CN112266396B/en
Publication of CN112266396A publication Critical patent/CN112266396A/en
Application granted granted Critical
Publication of CN112266396B publication Critical patent/CN112266396B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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 Table
    • C07F15/0006Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table 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
    • A61P35/00Antineoplastic agents

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (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)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses an integrated prodrug based on bioorthogonal chemistry, a preparation method thereof and application of the integrated prodrug as a novel integral bioorthogonal prodrug in antitumor drugs. The compound can be selectively activated in tumor cells, tetravalent platinum in the structure of the compound is reduced into cisplatin, meanwhile, the cisplatin is used as a novel bioorthogonal catalyst to catalyze part of NO donor fragments to release NO, and the NO donor fragments and the cisplatin cooperatively play an anti-tumor activity; the compound does not have the above process in normal cells, so that the compound has better safety.

Description

Integrated prodrug based on bio-orthogonal chemistry, preparation method and medical application thereof
Technical Field
The invention belongs to the technical field of pharmacy, and relates to a preparation method and application of a Nitric Oxide (NO) donor containing a tetravalent platinum complex, in particular to a novel micromolecule which has tumor selectivity and combines the tetravalent platinum complex and an NO donor azonium dialkoxide fragment, wherein the micromolecule and the NO donor azonium dialkoxide fragment are connected through a succinyl group.
Background
At present, the morbidity and mortality of malignant tumors always show a significant trend, and chemotherapy is once the standard therapy for tumor treatment. However, the traditional small molecule drugs lack targeting property, and can kill normal cells by mistake while killing tumor cells to cause serious toxic and side effects, so that the wide application in clinic is limited. Bio-orthogonal chemistry, an emerging and developing discipline, is widely used to label, track, manipulate and activate target molecules by means of bond generation and bond cleavage. In recent years, some bioorthogonal bond cleavage reactions catalyzed by metal catalysts have been successfully applied to tumor therapy. Compared with the traditional cytotoxic drugs or the antitumor drugs which rely on enzyme activation, the prodrug activation strategy which relies on the biological orthogonal bond breakage of the exogenous catalyst can better realize the aim of targeting tumors and reduce side effects and adverse reactions to the maximum extent.
Bioorthogonal metal catalysts
The most widely used bio-orthogonal chemical metal catalysts currently include gold, copper and palladium. The structure of the metal catalyst is difficult to modify, and the metal catalyst and the prodrug are required to be separately administrated, so that the curative effect of the compound is greatly limited, the selectivity and the targeting property in vivo are lacked, and the metal catalyst is easy to accumulate in vivo and difficult to metabolize. Therefore, to solve the above problems, the discovery of a new catalyst is crucial for bio-orthogonal chemistry. As platinum and palladium are in the same main group of the periodic table of elements, cisplatin can be found as a new bioorthogonal catalyst after a series of verification. However, cisplatin has strong cytotoxicity and cannot be directly administered, so that modification of cisplatin is required. The tetravalent platinum prodrug can be selectively reduced into cisplatin under the action of tumor cell reduction mediators, so that the tetravalent platinum prodrug plays a catalytic role. Meanwhile, cisplatin can be crosslinked with DNA in tumor cells to play a role in cytotoxicity, so that a tetravalent prodrug of cisplatin is selected as a precursor form of a catalyst.
Tetravalent platinum prodrugs
Tetravalent platinum prodrugs are a new class of molecules that may improve the pharmacological properties of divalent platinum anti-tumor drugs. The central platinum ion of the tetravalent platinum complex relative to the divalent platinum complex in planar configuration is d6The electronic configuration is increased by two axial orbitals, and a stable octahedral coordination configuration can be formed. Chemical ligand modification of the axial position of the tetravalent platinum complex may further influence and adjust the physical and chemical properties of the tetravalent platinum complex. Different ligand modifications to the axial position of tetravalent platinum can affect the fat solubility, reducibility, targeting property and other biological activities of the tetravalent platinum complex. The tetravalent platinum complex is reduced by transfer of two electrons, which is reduced to divalent platinum with antitumor activity, accompanied by release of two axial ligands.
NO donor type medicine
NO donor type drugs generally refer to prodrugs formed from NO donors and related drugs or certain active compounds through various linking groups. It has now been found that NO donors of various structural types, such as nitrosothiols, nitrates, NO-metal complexes (nitroprusside), furazan N-oxides, azonium dialkoxides, etc.; among them, azonium dialkoxides (diazeniumdiolates) have significant advantages in selective and targeted release of NO. On one hand, azonium dialkoxide is extremely unstable under physiological conditions, can automatically release 1-2 molecules of NO, and has half-life period from several seconds to several hours. On the other hand, O of azonium dialkoxide2The site (O attached to the azonium ion is called O1O attached to the nitrogen atom of the olefinic bond is O2) After alkylation, can be converted into prodrugs that are stable under physiological conditions; removing O by recognition of some specific enzymes in vivo or under the action of special physiological environment2The protective group is converted into unstable azo onium diol salt anions, thereby achieving the selective and targeted release of NO. To date, a variety of O's have been developed2The new policy is protected.
NO and cancer
NO is an organismThe inner water-soluble gas carrying free radicals has redox properties and plays an important role in physiology and pathology. Research has shown that NO is a potential anti-tumor agent that can modulate tumor-related processes including angiogenesis, apoptosis, invasion and metastasis, cell cycle, etc. The direct action mechanism of NO on tumor cells is not clear, and there are 3 main aspects reported at present: (1) NO reacts with intracellular superoxide anion to form peroxynitrite, which is protonated and decomposed into NO2And hydroxyl radicals, which can bind to various molecules of tumor cells, thereby causing tumor cell damage, such as lipid peroxidation, protein, amino acid crosslinking reaction; (2) NO is easy to form Fe-NO with Fe-S center-containing protein Fe, so that the degradation of Fe-S prosthetic group and aconitase on mitochondrial respiratory chain is caused, the synthesis of intracellular energy is prevented, and apoptosis is induced; (3) NO can directly act on RNA reductase to influence the replication of tumor cell DNA, and can also nitrify the DNA to inhibit the proliferation of tumor cells. Another major mechanism of NO against tumors is the killing of tumor cells by NO-mediated macrophages, and the specific mechanism is: t cell recognition antigen → T cell secretion of cytokines → cytokine stimulation to produce NO → NO killing of tumor cells. In addition, there are studies demonstrating that NK cytotoxicity is also NO-dependent. Therefore, designing and studying NO prodrugs becomes one of the important strategies for the innovation of antitumor drugs. The National Cancer Institute (NCI), some world-known pharmaceutical companies such as Merk, Pfizer, NicOx, Nitromed, etc. all invest in supporting NO donor-type drug development. In recent years, research and development of NO donor type drugs have also been greatly advanced, and NCI has placed JS-K, an anti-tumor drug, into rapid research and development plans.
Disclosure of Invention
Object of the Invention
The present invention provides an organic compound comprising a tetravalent platinum complex and an azonium dialkoxide fragment having an antitumor therapeutic effect. The compound can selectively release NO and cisplatin in tumor cells, thereby exerting a synergistic antitumor effect.
Technical scheme
An organic compound comprising a tetravalent platinum complex and an azonium dialkoxide fragment, which compound is based on a tetravalent platinum complex and a nitric oxide donor molecule, the nitric oxide donor molecule and the tetravalent platinum complex being connected by a chemical coupling process using a succinate bond as a single entity; the nitric oxide donor molecule is N-methylethanolamine azonium dialkoxide; the tetravalent platinum complex is cis-diamine trichlorohydroxyplatine and trans-diamine trichlorohydroxyplatine.
In a first aspect, the present invention provides a compound having N-methylethanolamine azonium dialkoxide as a skeleton, which is a compound represented by the general formula I:
Figure BDA0002717319900000031
wherein: r1Selected from methyl, allyl, propargyl; r2Selected from cis-diamine trichlorohydroxyplatine and trans-diamine trichlorohydroxyplatine.
Further, R1Selected from propargyl, methyl, R2Selected from cis-diamine trichlorohydroxyplatine and trans-diamine trichlorohydroxyplatine.
In some embodiments, the compound of formula I is selected from:
Figure BDA0002717319900000032
in a second aspect, a process for the preparation of said compounds, the synthetic route for the compounds of formula I is as follows:
Figure BDA0002717319900000041
NOgas: nitric oxide gas, anhydrousether: anhydrous diethyl ether; CH (CH)3ONa: sodium methoxide; CH (CH)3OH: methanol; DMF: n, N-dimethylformamide; THF: tetrahydrofuran;CHP: cis-diamminetrichlorohydroxyplatinum; FHP: trans-diamine trichlorohydroxyplatine; et (Et)3N: triethylamine; TBTU: 2- (1H-benzotriazol L-1-yl) -1,1,3, 3-tetramethyluronium tetrafluoroborate.
In a third aspect, the present invention also provides a pharmaceutical composition comprising a therapeutically effective amount of the compound and a pharmaceutically acceptable carrier.
In a fourth aspect, the compound or the solvate thereof is used for preparing a medicament for preventing and/or treating tumor diseases. The application of the pharmaceutical composition in preparing medicines for preventing and/or treating tumor diseases.
Has the advantages that: the invention integrates two fragments of NO donor azonium dialkoxide and tetravalent platinum complex together, and designs the integrated bioorthogonal chemical catalytic NO donor drug. In the specific reducing environment of tumor cells, the compound reduces tetravalent platinum fragments into cisplatin and releases O2Protected azonium dialkoxide fragments. Cisplatin plays an anti-tumor role on one hand, and on the other hand, cisplatin is used as a bioorthogonal catalyst to catalyze azonium dialkoxide O2The breakage of the protecting group further releases NO, thereby exerting the curative effect of synergy anti-tumor. The integrated prodrug molecule has higher tumor specificity, so that cisplatin and NO are specifically released in tumor cells, the toxic and side effects on normal cells caused by low targeting when the integrated prodrug molecule is used alone are avoided, and the problem that the traditional bioorthogonal reaction catalyst lacks selectivity is solved. Meanwhile, the design concept of the whole molecule also improves the traditional mode of separately feeding the catalyst and the prodrug of the bioorthogonal chemistry, and further improves the drug forming property of the whole bioorthogonal prodrug.
The invention synthesizes a new compound I for the first time5aThe compound I is verified by cell and zebra fish experiments5aCan be selectively activated in tumor cells, thereby exerting the anti-tumor curative effect. Compound I compared to Normal cells5aWill preferentially enter tumor cells, then tetravalent platinum is reduced to cisplatin, and the cisplatin catalytic compound releases NO, thereby exerting the antitumor activity in synergy with cisplatin. Andin comparison to the currently existing bioorthogonal prodrugs, Compound I5aAs an integral prodrug molecule, the compound avoids separate administration, enhances the targeting property of the compound, reduces the toxicity of a catalyst, and greatly improves the druggability of the integrated bioorthogonal chemical catalytic NO donor molecule.
Drawings
FIG. 1 is Compound I5aThe design concept of (1).
FIG. 2 is Compound I5aStability in PBS and in the presence of L- (+) -ascorbic acid.
FIG. 3 is I5aLevels of NO released in PBS and in the presence of L- (+) -ascorbic acid.
FIG. 4 is Compound I5aLevels of NO were released in a2780 and IOSE80 cells.
FIG. 5 shows the concentrations of Compound I5aAnd levels of NO released at different times from cell incubation.
FIG. 6 is the NO scavenger carboxy-PTIO vs. Compound I5aThe effect of the tumor cell proliferation inhibitory activity of (3).
FIG. 7 shows the comparison of A2780 and IOSE80 cells against Compound I5aAnd the amount of CDDP taken.
FIG. 8 is Compound I5aAnd CDDP Pt content on DNA in a2780 cells and IOSE80 cells, respectively.
FIG. 9 is a study of Compound I using a cisplatin Probe5aIn the case of cisplatin conversion in A2780 cells and IOSE80 cells.
FIG. 10 is a photograph of a further study of Compound I5aAntitumor proliferation inhibitory activity in zebra fish.
FIG. 11 shows further investigation of Compound I5aNO release in zebrafish.
Detailed Description
The invention will be further described with reference to specific embodiments, and the advantages and features of the invention will become apparent as the description proceeds. These examples are illustrative only and do not limit the scope of the present invention in any way. It will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention, and that such changes and modifications may be made without departing from the spirit and scope of the invention.
The synthetic route for the compounds of formula I is as follows:
Figure BDA0002717319900000051
CH3ONa: sodium methoxide; CH (CH)3OH: methanol; DMF: n, N-dimethylformamide; THF: tetrahydrofuran; CHP: cis-diamminetrichlorohydroxyplatinum; FHP: trans-diamine trichlorohydroxyplatine; et (Et)3N: triethylamine; TBTU: 2- (1H-benzotriazol L-1-yl) -1,1,3, 3-tetramethyluronium tetrafluoroborate.
In some embodiments, the intermediate compound I2To compound I4The synthesis of (a) is as follows:
Figure BDA0002717319900000061
compound I2The synthesis of (2): adding N-methyl-2-hydroxyethylamine (I)120mL), and 50.79mL of a methanol solution (5.4mol/L) of sodium methoxide were mixed and added to a polytetrafluoroethylene container, and 200mL of anhydrous ether was added thereto to prepare a reaction system N2After replacement, introducing Nitric Oxide (NO) gas to make the pressure reach 0.4-0.8MPa, and carrying out closed reaction in a greenhouse for 24 h. Post-treatment, discharging excessive unreacted NO gas to reduce pressure to normal pressure, opening the container, pouring the reaction solution into 1L anhydrous ether to separate out a large amount of white solid, filtering, washing the filter cake with ether for 3 times, drying in a vacuum drying oven at room temperature for 2h, and collecting a white product, namely the compound I2. The resulting compound I2The reaction was carried out without further purification.
Figure BDA0002717319900000062
Compound I3aThe synthesis of (2): take 1g toCompound I2And 5mL of DMF was added to a 100mL two-necked glass reaction flask, and the flask was placed in ice bath and subjected to N2And (3) after protection, slowly adding 757.8mg of bromopropyne into a reaction bottle, continuing to react for 0.5h in an ice bath after dripping, and then moving the reaction solution to room temperature to continue to react for 12 h. After-treatment, DMF is removed by rotary evaporation, the residue is prepared into sand, and column chromatography is carried out to obtain colorless oily substance I3a
Compound I3bThe synthesis of (2): 1g of the compound I are taken2And 5mL of DMF was added to a 100mL two-necked glass reaction flask, and the flask was placed in ice bath and subjected to N2And (3) protecting, slowly adding 949.8mg of methyl iodide into the reaction bottle, keeping the temperature in an ice bath for 0.5h after dripping, and then moving the reaction solution to room temperature to continue reacting for 12 h. After-treatment, DMF is firstly removed by rotary evaporation, and the residue is subjected to sand preparation and column chromatography to obtain a light yellow oily substance I3b
Figure BDA0002717319900000063
Compound I4aThe synthesis of (2): taking 700mg of compound I3aThe reaction mixture was dissolved in 10mL of anhydrous tetrahydrofuran, 122mg of DMAP was added thereto, the mixture was stirred at room temperature for 15mins, 608mg of succinic anhydride was added thereto, and the reaction mixture was refluxed overnight. Post-treatment, filtering the reaction solution, concentrating the filtrate, adding water, extracting with dichloromethane for 5 times, drying, and concentrating to obtain compound I4a
Compound I4bThe synthesis of (2): taking 602mg of compound I3bThe reaction mixture was dissolved in 10mL of anhydrous tetrahydrofuran, 122mg of DMAP was added thereto, the mixture was stirred at room temperature for 15mins, 608mg of succinic anhydride was added thereto, and the reaction mixture was refluxed overnight. Post-treatment, filtering the reaction solution, concentrating the filtrate, adding water, extracting with dichloromethane for 5 times, drying, and concentrating to obtain compound I4b
Figure BDA0002717319900000071
Example 1: compound I5a,I5bPreparation of
Figure BDA0002717319900000072
The synthetic route is as follows:
Figure BDA0002717319900000073
Figure BDA0002717319900000074
CH3ONa: sodium methoxide; CH (CH)3OH: methanol; DMF: n, N-dimethylformamide; THF: tetrahydrofuran; CHP: cis-diamminetrichlorohydroxyplatinum; FHP: trans-diamine trichlorohydroxyplatine; et (Et)3N: triethylamine; TBTU: 2- (1H-benzotriazol L-1-yl) -1,1,3, 3-tetramethyluronium tetrafluoroborate.
217mg of Compound I4In a single-neck reaction flask, 86.6mg of triethylamine, 265mg of TBTU and 5mL of DMF were added and dissolved, and the reaction mixture was stirred at room temperature for 30 mins. Then, 200mg of CHP or FHP was added to the reaction flask, and the reaction was continued overnight under exclusion of light. Post-treatment, firstly concentrating to remove DMF, preparing sand from residues, and carrying out column chromatography to obtain a target product I5aAnd I5b
Compound I5a: pale yellow solid, 97mg, yield 28%.1H NMR(ppm,300MHz,DMSO-d6)δ6.28-6.03(m,6H),4.82(d,J=2.4Hz,2H),4.16(t,J=5.4Hz,2H),3.58(t,J=2.4Hz,2H),2.98(s,3H),2.49-2.42(m,5H).13C NMR(ppm,75MHz,DMSO-d6)δ178.90,172.01,78.75,60.54,60.39,51.83,40.52,30.94,29.80.HRMS(ESI)calcd for C10H20Cl3N5O6Pt[M+H]+:607.0200;Found:607.0191,ppm error-1.5.
Compound I5bLight yellow solid, 89mg, yield 18%.1H NMR(ppm,300MHz,DMSO-d6)δ6.06(br,6H),4.83(d,J=2.4Hz,2H),4.17(t,J=5.1Hz,2H),3.60(t,J=4.2Hz,2H),3.00(s,3H),2.47-2.45(m,5H).ESI-MS m/z:629.1[M+Na]+
Example 2: compound I5cPreparation of
Figure BDA0002717319900000081
The synthetic route is as follows:
Figure BDA0002717319900000082
Figure BDA0002717319900000083
CH3ONa: sodium methoxide; CH (CH)3OH: methanol; CH (CH)3I: methyl iodide; DMF: n, N-dimethylformamide; THF: tetrahydrofuran; CHP: cis-diamminetrichlorohydroxyplatinum; et (Et)3N: triethylamine; TBTU: 2- (1H-benzotriazol L-1-yl) -1,1,3, 3-tetramethyluronium tetrafluoroborate.
200mg of compound I are taken4In a single-necked reaction flask, 86.6mg of triethylamine and 265mg of TBTU were added and dissolved in 5mL of DMF, and the reaction mixture was stirred at room temperature for 30 min. Then, 200mg of CHP was added to the reaction flask, and the reaction solution was allowed to react overnight under exclusion of light. Post-treatment, firstly concentrating to remove DMF, preparing sand from residues, and carrying out column chromatography to obtain a target product I of 134mg5cThe yield thereof was found to be 37%.1H NMR(ppm,500MHz,DMSO-d6)δ6.28-6.03(m,6H),4.15(t,J=5.0Hz,2H),3.90(s,3H),3.52(t,J=5.0Hz,2H),2.95(s,3H),2.52-2.44(m,4H).13C NMR(ppm,125MHz,DMSO-d6)δ182.01,175.12,63.69,63.57,55.06,43.86,34.06,32.93.HRMS(ESI)calcd for C8H20Cl3N5O6Pt[M+H]+:583.0200;Found:583.0191,ppm error-1.5.
FIG. 1 shows Compound I5aTransformation in tumor cells and Normal cells, respectivelyAnd (6) carrying out the process. From the above, Compound I5aCan be stable in normal cells and substantially free of cytotoxicity, while in tumor cells, Compound I5aThe tetravalent platinum can be reduced to cisplatin first, and the cisplatin catalytic compound releases NO, so that the cisplatin and the cisplatin can exert the antitumor activity synergistically.
FIG. 2 is Compound I5aStability in PBS and in the presence of L- (+) -ascorbic acid. As can be seen from FIG. 2, I5aRelatively stable in PBS at 24 deg.c; however, in the presence of L- (+) -ascorbic acid, the degradation can be faster, e.g., more than 50% in 2h, and substantially complete in 24 h.
FIG. 3 is I5aThe concentration of NO released in PBS and in the presence of L- (+) -ascorbic acid was measured. As can be seen from FIG. 3, I5aSubstantially NO production in the absence of L- (+) -ascorbic acid; when the L- (+) -ascorbic acid exists, more NO is generated; description of the invention I5aFirstly, cis-platinum and I are generated under the action of L- (+) -ascorbic acid4aCDDP further catalyzes I4aTo thereby release NO.
Table 1 shows Compound I5aThe proliferation inhibitory activity of human ovarian cancer cell A2780 and human ovarian normal cell IOSE80 was studied.
TABLE 1
Figure BDA0002717319900000091
As can be seen from table 1, CHP showed higher tumor cell selectivity compared to cisplatin CDDP. And the compound I4aIC for tumor cell A2780 and normal cell IOSE8050The larger values indicate that the growth inhibitory activity of the cells of 2 strains was weak. When CHP is pre-incubated with A2780 for 10h, I is added4aShowing a strong proliferation inhibitory activity (I)4a,IC50=89.46±4.87μMvs CHP+I4a,IC500.81 ± 0.07 μ M), indicating that CHP can be reduced to cisplatin, which catalyzes I, after entering tumor cells4aNO is produced. Importantly, I5aShows stronger proliferation inhibition activity on A2780 than CDDP and CHP (I)5a,IC50=0.23±0.01μM;CDDP,IC50=1.08±0.06μM;CHP,IC501.69 ± 0.06 μ M). At the same time, I5a(SF ═ 518) shows better selectivity for tumor cells than CDDP (SF ═ 7.8) and CHP (SF ═ 10.9).
FIG. 4 is Compound I5aLevels of NO were released in a2780 and IOSE 80. As can be seen in FIG. 4, I4aWhen incubated with cells alone, substantially NO was produced. When CHP and A2780 cells are added for pre-incubation for 10 hours, I is added4aAfter 8h incubation, more NO was produced, while essentially NO was produced in normal cells IOSE80, indicating that cisplatin-catalyzed O-propynyl cleavage can be selectively performed in tumor cells. I is5aThe strongest MFI value (25.33) was produced in tumor cells A2780, higher than I4a+ CHP (MFI 15.16) and JS-K (MFI 7.75).
FIG. 5 shows the concentrations of Compound I5aAnd levels of NO released at different times from cell incubation. As can be seen from FIG. 5, Compound I5aThere is essentially NO production in normal cells, while the level of NO released in tumor cells is dose and time dependent, i.e. as the concentration increases or the incubation time increases, the amount of NO released gradually increases.
FIG. 6 is the NO scavenger carboxy-PTIO vs. Compound I5aThe effect of the tumor cell proliferation inhibitory activity of (3). As can be seen from FIG. 6, when carboxy-PTIO was added, Compound I was reduced5aInhibitory Activity on tumor cells A2780 and with increasing Carboxy-PTIO concentrations on Compound I5aThe more significant the decrease in proliferation inhibitory activity of (a). The results show that the compounds I5aNO produced in tumor cells contributes to the activity of tumor cells.
FIG. 7 shows the comparison of A2780 and IOSE80 cells against Compound I5aAnd the amount of CDDP taken. From FIG. 7, it can be seen that both the whole cell and the nucleus of A2780 are of pair I5a(Whole cell, 763.53. + -. 14.81pg Pt/106cells; in the nucleus, 84.4. + -. 1.77pg Pt/106cells) are all significantly higher thanCDDP (Whole cell, 98.34. + -. 23.0pg Pt/106cells; nucleus, 24.78. + -. 1.55pg Pt/106cells)。
FIG. 8 is Compound I5aAnd CDDP Pt content on DNA in a2780 cells and IOSE80 cells, respectively. As can be seen from FIG. 8, in A2780 cells, Compound I5a(22.56pg Pt/. mu.gDNA) platinized DNA levels were significantly higher than cisplatin (5.18pg Pt/. mu.gDNA). And compounds I5aThe level of platinized DNA in A2780 tumor cells (22.56pg Pt/. mu.gDNA) was higher than its level in ovarian normal cells IOSE80 (5.67pg Pt/. mu.gDNA).
FIG. 9 is a study of Compound I using a cisplatin Probe5aIn the case of cisplatin conversion in A2780 cells and IOSE80 cells. As can be seen from the left panel of FIG. 9, at 0h, the fluorescence in A2780 cells and IOSE80 cells was weak, indicating that there was almost no cisplatin in the cells, but after 3h, the fluorescence in A2780 cells was strong, indicating more cisplatin content, while the fluorescence in IOSE80 cells was still weak, indicating less cisplatin content in the cells. The right graph is a data statistics graph of fluorescence intensity in confocal microscope pictures, and it can be seen that after 3h, the fluorescence intensity in a2780 cells is significantly higher than 0h, and the fluorescence intensity in a2780 cells is also significantly higher than that in IOSE80 cells. From the above, Compound I5aCisplatin production was significantly higher in A2780 tumor cells than in IOSE80 normal cells.
Example 3: investigation of Compound I5aAntitumor proliferation inhibitory activity in zebra fish.
Firstly, a certain amount of quilt CellTracker is implanted into the yolk part of the young zebra fish in a displacement wayTMA2780 cells marked by CM-Dil fluorescent dye are grown for 24 hours, and strong yellow fluorescence can be seen from the yellow parts of the zebra fish egg under a confocal microscope, so that a zebra fish experimental model of successful tumor cells is constructed. Then the zebra fish which are implanted with tumor cells are respectively mixed with the zebra fish containing I5aFeeding zebra fish culture solution containing 1 mu M of cisplatin, 1 mu M of cisplatin and no other compounds, observing yellow fluorescence distribution in zebra fish body under confocal microscope after 72h, and counting corresponding fluorescence intensity values, thereby obtaining fluorescent valueEvaluation of Compound I5aAnti-tumor proliferation inhibiting effect. FIG. 10 is a photograph of a further study of Compound I5aAntitumor proliferation inhibitory activity in zebra fish. As can be seen from FIG. 10, when zebrafish were compared to the control group with Compound I5aAfter incubation for 72h, the fluorescence distribution in the zebra fish bodies is obviously reduced, and the compound I is proved5aEffectively inhibit the growth of tumor cells in zebra fish bodies, and the effect is better than that of cisplatin. The right-hand fluorescence intensity histogram also validates this conclusion.
Example 4: investigation of Compound I5aNO release in zebrafish.
Firstly, a certain amount of quilt CellTracker is implanted into the yolk part of the young zebra fish in a displacement wayTMA2780 cells marked by CM-Dil fluorescent dye are grown for 24 hours, and strong yellow fluorescence can be seen from the yellow parts of the zebra fish egg under a confocal microscope, so that a zebra fish experimental model of successful tumor cells is constructed. Zebrafish were incubated with the DAF-FM DA probe for 40 minutes at 37 ℃ and then photographed under a confocal microscope to determine their own NO levels in the zebrafish. Then, the zebra fish is raised in a pure zebra fish culture solution for 24 hours, so that the zebra fish metabolizes the NO probe in the body completely. Then mixing zebra fish with the mixture containing I5a(10. mu.M) and a zebrafish culture broth without addition of other compounds, and after 24 hours, the zebrafish were incubated with a DAF-FM DA (5. mu.M) probe at 37 ℃ for 40 minutes, and then photographed under a confocal microscope, and the corresponding fluorescence intensity values were counted, thereby verifying the compound I5aWhether NO can be selectively released at the tumor cell part of the zebra fish. FIG. 11 shows further investigation of Compound I5aNO release in zebrafish. As can be seen from FIG. 11, when zebrafish were mixed with Compound I5aAfter incubation, the fluorescence intensity at the tumor site was significantly higher than that of the control group, thereby verifying that Compound I5aThe fluorescent protein can selectively release NO at a tumor site in zebra fish bodies, and the fluorescence statistics on the right side also verify the conclusion.
The above description is only of the preferred embodiments of the present invention, and it should be noted that: it will be apparent to those skilled in the art that various modifications and adaptations can be made without departing from the principles of the invention and these are intended to be within the scope of the invention.

Claims (7)

1. A compound of formula I or an optical isomer, enantiomer, diastereomer, racemate or racemic mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure FDA0002717319890000011
wherein: r1Selected from methyl, allyl, propargyl; r2Selected from cis-diamine trichlorohydroxyplatine and trans-diamine trichlorohydroxyplatine.
2. A compound of claim 1, wherein R is1Selected from methyl and propargyl.
3. The compound of claim 1, wherein the compound of formula I is selected from the group consisting of:
Figure FDA0002717319890000012
4. a process for the preparation of a compound according to any one of claims 1 to 3, characterized in that the compound of formula I is synthesized as follows:
Figure FDA0002717319890000021
CH3ONa: sodium methoxide; CH (CH)3OH: methanol; DMF: n, N-dimethylformamide; THF: tetrahydrofuran; CHP: cis-diamminetrichlorohydroxyplatinum; FHP: trans-diamine trichlorohydroxyplatine; et (Et)3N: triethylamine;TBTU: 2- (1H-benzotriazol L-1-yl) -1,1,3, 3-tetramethyluronium tetrafluoroborate.
5. A pharmaceutical composition comprising a therapeutically effective amount of a compound of any one of claims 1-3 and a pharmaceutically acceptable carrier.
6. Use of a compound according to any one of claims 1 to 3 or a solvate thereof for the manufacture of a medicament for the prophylaxis and/or treatment of a neoplastic disease.
7. Use of the pharmaceutical composition of claim 5 for the preparation of a medicament for the prevention and/or treatment of a tumor disease.
CN202011077271.9A 2020-10-10 2020-10-10 Integrated prodrug based on bio-orthogonal chemistry, preparation method and medical application thereof Active CN112266396B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011077271.9A CN112266396B (en) 2020-10-10 2020-10-10 Integrated prodrug based on bio-orthogonal chemistry, preparation method and medical application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011077271.9A CN112266396B (en) 2020-10-10 2020-10-10 Integrated prodrug based on bio-orthogonal chemistry, preparation method and medical application thereof

Publications (2)

Publication Number Publication Date
CN112266396A true CN112266396A (en) 2021-01-26
CN112266396B CN112266396B (en) 2021-08-20

Family

ID=74337809

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011077271.9A Active CN112266396B (en) 2020-10-10 2020-10-10 Integrated prodrug based on bio-orthogonal chemistry, preparation method and medical application thereof

Country Status (1)

Country Link
CN (1) CN112266396B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1234698C (en) * 2001-03-01 2006-01-04 国家研究委员会 Nitric oxide donors based on metallic centres
CN104829652A (en) * 2015-03-31 2015-08-12 华东理工大学 Near infrared light-controlled nitrogen monoxide release nanosystem, and preparation method and application thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1234698C (en) * 2001-03-01 2006-01-04 国家研究委员会 Nitric oxide donors based on metallic centres
CN104829652A (en) * 2015-03-31 2015-08-12 华东理工大学 Near infrared light-controlled nitrogen monoxide release nanosystem, and preparation method and application thereof

Also Published As

Publication number Publication date
CN112266396B (en) 2021-08-20

Similar Documents

Publication Publication Date Title
CN101801369B (en) Phosphaplatins and their use in the treatment of cancers resistant to cisplantin and carboplatin
TW200408644A (en) Platinum compounds
EA010834B1 (en) Pyridyl-substituted porphyrin compounds and methods of use thereof
US9365593B2 (en) Photolabile caged transition metal complexes and methods of using the same
CN106279321A (en) Gemcitabine ProTide weary oxygen activation prodrug and application thereof
US6894049B1 (en) Platinum complexes as antitumor agents
US20230075925A1 (en) Non-peripheral quaternary ammonium-modified zinc phthalocyanine and preparation method and use thereof
JP2017519004A (en) Texaphyrin-platinum (IV) conjugates and compositions for use in overcoming platinum resistance
US6413953B1 (en) Pt(IV) antitumor agent
KR100239787B1 (en) Trinuclear cationic platinum complexes having antitumour activity and pharmaceutical compositions containing them
Hashmi et al. Medicinal applications of vanadium complexes with Schiff bases
CN102627685B (en) Nitric oxide-donating glutathione compound, preparation method and medical purpose thereof
CN112175014B (en) Nitric oxide donor type tetravalent platinum derivative, preparation method and medical application thereof
CN112266396B (en) Integrated prodrug based on bio-orthogonal chemistry, preparation method and medical application thereof
CN114437128B (en) Choline phosphate modified taxol medicine and preparation method and application thereof
CN106924732A (en) A kind of ultrasonic therapy cancer target type haematoporphyrin injection and preparation method thereof
EP0897389B1 (en) Binuclear platinum complexes, method for preparing same and pharmaceutical compositions containing said complexes
CN114478561B (en) Epalrestat lycorine conjugate and preparation method and application thereof
JPH04502311A (en) Organometallic compounds, their preparation methods and pharmaceutical compounds containing them
CN101489540A (en) Ruthenium II compounds
CN110016048B (en) Derivative of mitochondrion targeting IR780 iodide, preparation method and application
CN109553558B (en) Selenium-containing compound and anti-tumor application thereof
AU2021362841A1 (en) Hexadecylammonium group-modified phthalocyanine, and preparation method therefor and application thereof as photodynamic drug
ES2272755T3 (en) ORGANOMETAL ANTITUMORAL AGENT.
CN105153238B (en) A kind of preparation method of Miboplatin

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
GR01 Patent grant
GR01 Patent grant