CN116410234A - Preparation and application of cisplatin prodrug self-assembled nanoparticles for overcoming cisplatin resistance - Google Patents

Preparation and application of cisplatin prodrug self-assembled nanoparticles for overcoming cisplatin resistance Download PDF

Info

Publication number
CN116410234A
CN116410234A CN202310397921.5A CN202310397921A CN116410234A CN 116410234 A CN116410234 A CN 116410234A CN 202310397921 A CN202310397921 A CN 202310397921A CN 116410234 A CN116410234 A CN 116410234A
Authority
CN
China
Prior art keywords
cisplatin
tpp
lnd
small molecule
drug
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
CN202310397921.5A
Other languages
Chinese (zh)
Other versions
CN116410234B (en
Inventor
路海滨
姜美旭
王凯
刘一霖
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.)
Jilin University
Original Assignee
Jilin 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 Jilin University filed Critical Jilin University
Priority to CN202310397921.5A priority Critical patent/CN116410234B/en
Publication of CN116410234A publication Critical patent/CN116410234A/en
Application granted granted Critical
Publication of CN116410234B publication Critical patent/CN116410234B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • A61K47/6939Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being a polysaccharide, e.g. starch, chitosan, chitin, cellulose or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention belongs to the technical field of medicines, and designs and synthesizes a cisplatin prodrug capable of overcoming cisplatin resistance, and on the basis, conjugated polysaccharide molecules are formed by constructing prodrug hyaluronic acid and cyclodextrin for encapsulation to prepare a small molecular prodrug self-assembled nano drug delivery system. The prodrug nano system can target a tumor cell CD44 receptor, a triphenyl phosphorus group contained in the structure targets cell mitochondria, and simultaneously disulfide bonds are broken to release Pt-TPP and LND under the high GSH environment of the tumor, and the Pt-TPP is activated by a biological reducer to release cisplatin, so that the cytotoxic effect of the cisplatin is exerted. The released LND can overcome the drug resistance of cells to cisplatin by inhibiting glycolysis to synergistically kill tumor cells with cisplatin. The invention realizes the specific effect of the medicine on tumor cells, overcomes the drug resistance of the tumor cells to cisplatin while improving the anti-tumor effect of the medicine, obviously improves the anti-tumor effect and has wide development prospect.

Description

Preparation and application of cisplatin prodrug self-assembled nanoparticles for overcoming cisplatin resistance
Technical Field
The invention belongs to the field of medical technology and pharmaceutical preparations, and mainly comprises synthesis of cisplatin prodrugs (LND-SS-Pt-TPP) for overcoming cisplatin resistance, assembly construction of an LND-SS-Pt-TPP/HA-CD nano targeting system, and research on anti-tumor effect of the system.
Background
Cancer is a serious threat to the health of all humans, so the development of anticancer drugs for the treatment of malignant tumors has been the focus of drug development. Cisplatin is one of the most potential and most widely used drugs for treating various solid cancers, such as testicular cancer, ovarian cancer, head and neck cancer, bladder cancer, lung cancer, cervical cancer and the like, but has a strong side effect and is easy to generate drug resistance due to poor targeting, so that clinical application of cisplatin is limited.
The invention synthesizes and constructs an LND-ss-Pt-TPP/HA-CD reduction stimulation response type self-assembled prodrug nano targeting delivery system with triple targeting function. The prodrug nanoparticle wrapped by the HA-CD macromolecule material can be used for actively targeting CD44 on the surface of a tumor and then targeting intracellular mitochondria through a triphenylphosphine group, so that the prodrug is delivered to the mitochondria in the tumor cell; disulfide bonds can be selectively degraded and broken by high-concentration GSH in tumor cells and mitochondria to release LND and tetravalent cisplatin, and simultaneously the tetravalent platinum is reduced to divalent platinum under the action of GSH and bad blood acid; release of LND can reduce expression of hexokinase 2 in tumor cells, destroy mitochondria and inhibit glycolysis to block energy supply, and can overcome cisplatin resistance while killing tumor cells. The cisplatin released simultaneously causes mitochondrial DNA damage, and the tumor cells are jointly killed under the synergistic effect.
Disclosure of Invention
The invention aims to synthesize a small molecular prodrug with targeting capability and resisting cisplatin drug resistance, and assemble the small molecular prodrug into a nano drug, so that the effects of high drug loading capacity, good stability and low toxic and side effects are realized, and the anti-tumor activity is further improved.
The invention realizes the aim through the following technical scheme:
the small molecule prodrug of the invention connects tetravalent platinum drugs conjugated with TPP groups with lonidamine through disulfide bonds, and the structural formula is as follows:
Figure BDA0004178254120000011
wherein m is an integer of 1 to 6; n is an integer of 1 to 6.
The synthesis method of the small molecule prodrug provided by the invention comprises the following steps: cisplatin is oxidized by hydrogen peroxide to form tetravalent platinum, which is then reacted with intermediate TPP-NHS to form TPP-Pt. The synthesized lonidamine is connected with TPP-Pt through disulfide side chains to obtain a small molecular prodrug LND-SS-Pt-TPP.
Specifically, the invention provides a synthesis method of a small molecular prodrug LND-SS-Pt-TPP, which comprises the following steps:
synthesis of lonidamine: adding 1H-indole-3-carboxylic acid methyl ester into sodium hydroxide aqueous solution, heating and dripping 2, 4-dichloro benzyl chloride, stirring and cooling to room temperature, regulating the reaction system to be acidic, and recrystallizing with glacial acetic acid to obtain lonidamine.
(1) TPP-Pt synthesis: and adding hydrogen peroxide into cisplatin, stirring at room temperature for reaction, filtering, washing, drying to obtain diaminodihydroxyplatinum (VI) chloride, adding EDCI and NHS, dissolving in acetonitrile, and stirring at room temperature to obtain an intermediate product TPP-NHS. Dissolving TPP-NHS in anhydrous DMSO, adding diaminoplatinum (VI) chloride, reacting for 72h, and then removing DMSO to obtain TPP-Pt; the carboxyalkyl triphenyl phosphorus bromide is one or more of (2-carboxyethyl) triphenyl phosphorus bromide, (3-carboxypropyl) triphenyl phosphorus bromide, (4-carboxybutyl) triphenyl phosphorus bromide or (5-carboxypentyl) triphenyl phosphorus bromide.
(2) LND-SS-Pt-TPP Synthesis: the lonidamine and the alkyl glycol containing disulfide bond are subjected to esterification reaction under the action of condensing agent, and then are subjected to esterification reaction with N, N' -disuccinimidyl carbonate to obtain activated ester, and the activated ester is esterified with TPP-Pt to obtain a final compound (LND-SS-Pt-TPP); the alkyl glycol containing disulfide bond is one or more of 2,2' -dithiodiethanol, dithiodimethanol, 3' -dithiodipropanol or 4,4' -dithiodibutanol.
The invention also provides a preparation method of conjugated polysaccharide molecules (HA-CD) constructed by HA and Cyclodextrin (CDs), and a method for forming nano-drugs by non-covalent bonding of LND-SS-Pt-TPP prodrugs and HA-CD.
The preparation method of HA-CD comprises the following steps: 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide (EDCI) and N-hydroxysuccinimide sulfonate sodium salt (NHSS) were added to sodium hyaluronate in PBS phosphate buffer and stirred at room temperature for 30min. Then, an amino-substituted cyclodextrin in PBS was added thereto and reacted at room temperature for 24 hours. Dialyzing and freeze-drying to obtain the HA-CD.
The preparation method of the LND-SS-Pt-TPP/HA-CD drug-loaded nano system comprises the following steps: dissolving LND-SS-Pt-TPP in an organic solvent, slowly adding the drug solution into the HA-CD aqueous solution under the condition of stirring, performing ultrasonic treatment, dialyzing to remove the organic solvent, and freeze-drying to obtain LND-SS-Pt-TPP/HA-CD drug-carrying nanoparticles; the organic solvent comprises one or more of methanol, ethanol, tetrahydrofuran, N-Dimethylformamide (DMF) or dimethyl sulfoxide (DMSO).
The invention constructs the LND-SS-Pt-TPP/HA-CD nano targeting drug delivery system. The drug small molecule targeting prodrug LND-SS-Pt-TPP wrapped by HA-CD recognizes high-expression GSH, specifically acts on tumor cells, disulfide bonds and GSH are subjected to action and are broken to release Pt-TPP and LND, and the drug HAs mitochondrial targeting capability due to the existence of TPP groups; the platinum (IV) complex is immediately transformed into cisplatin under the action of GSH to induce the formation of DNA cross-linking of tumor cells; lonidamine (LND) inhibits glycolytic enzyme hexokinase II (HKII) and blocks the glycolytic process of cells, thereby interfering the generation of ATP in tumor cells, overcoming cisplatin resistance and playing a role of synergistically killing the tumor cells with cisplatin. The invention realizes the specific effect of the medicine on tumor cells, has obviously better anti-tumor effect than the independent use of the anti-cancer medicine, and has bright development prospect.
The LND-SS-Pt-TPP/HA-CD nano targeting drug delivery system formed by the HA-CD entrapped small molecular prodrug HAs the advantages that (1) the drug containing disulfide bonds is sensitive to GSH and is enriched in tumor cells with high GSH content, so that the drug can specifically act on the tumor cells (2) with smaller particle size, the enrichment of nano drugs (3) is facilitated, the drug loading quantity (4) is obviously improved, the anti-tumor effect is improved, and the toxic and side effects are reduced.
The invention has the following effects: (1) The LND-SS-Pt-TPP small molecule prodrug is designed and synthesized, and the synthesis method is stable, convenient and feasible. (2) The HA and Cyclodextrin (CDs) are prepared to construct conjugated polysaccharide molecule HA-CD nano material, and small molecule prodrug is entrapped to form uniform and stable nanoparticle medicine, and the preparation method is simple and easy to implement. (3) The combined application of the two medicines with the synergistic effect is realized, the targeting effect is realized, the anticancer effect is obviously improved, and the application prospect is wide.
Drawings
Fig. 1: example 1 Synthesis route for Small molecule prodrug LND-SS-Pt-TPP
Fig. 2: example 1 Small molecule prodrug LND-SS-Pt-TPP 1 HNMR profile
Fig. 3: mass Spectrometry of Small molecule prodrug LND-SS-Pt-TPP in example 1
Fig. 4: example 3 particle size distribution Spectrum of LND-SS-Pt-TPP/HA-CD self-assembled nanoparticle Darling Wen Lijing Instrument particle size distribution Spectrum of (A) HA-CD (B) LND-SS-Pt-TPP/HA-CD
Fig. 5: in example 3, LND-SS-Pt-TPP/HA-CD self-assembled nanoparticle TME electron microscopy scan profile, (A) HA-CD; (B) LND-SS-Pt-TPP/HA-CD
Fig. 6: EXAMPLE 1 cleavage Mass Spectrometry of the Small molecule prodrug LND-SS-Pt-TPP in GSH solution
Fig. 7: in example 1, small molecule prodrug LND-SS-Pt-TPP was probed in GSH solution cleavage fluorescent probe color chart, (A) LND-SS-Pt-TPP; (B) is a probe; (C) After LND-SS-Pt-TPP is incubated by GSH, a probe is added
Fig. 8: schematic of in vitro simulated GSH-triggered disulfide bond cleavage release
Fig. 9: LND-SS-Pt-TPP/HA-CD self-assembled nanoparticle A549 cell uptake patterns in example 3
Detailed Description
The invention is further illustrated by way of examples which follow, but are not thereby limited to the scope of the examples described.
Example 1: synthesis of small molecule prodrug LND-SS-Pt-TPP (4- ((2, 5-Dioxypyrrolidin-1-yl) oxy) -4-oxobutyl) triphenylphosphine (2)
EDCI (211 mg,1.1 mmol) and NHS (127 mg,1.1 mmol) were added to TPP (329 mg,1.0 mmol) in a 50mL round bottom flask and dissolved in 15mL acetonitrile. Stirring at room temperature for 12h, and monitoring the reaction progress by thin layer chromatography. Acetonitrile was removed by rotary evaporation, water was added, extraction was performed three times with DCM, the organic layer was collected, dried over anhydrous sodium sulfate, and the organic phase was evaporated to dryness under reduced pressure to give compound 7 (361 mg, yield 81%).
1 H-NMR(400MHz,CDCl 3 ):7.81-7.72(m,9H),7.66-7.61(m,6H),4.04-3.96(m,2H),3.14(t,J=6.3Hz,2H),2.81-2.74(m,4H),2.06-2.00(m,2H)。
Synthesis of (4- ((2, 5-dioxopyrrolidin-1-yl) oxy) -4-oxobutyl) triphenylphosphine (4)
TPP-NHS (161 mg,0.36 mmol) was dissolved in 5mL of anhydrous DMSO in a 25mL round bottom flask, diaminodihydroxyplatinum (VI) chloride (100 mg,0.30 mmol) dissolved in 10mL of anhydrous DMSO was added dropwise, vigorously stirred at 30℃for 72h, excess diethyl ether was added to remove DMSO, the product was precipitated with methanol and washed twice again with methanol and diethyl ether, and then dried under vacuum as pale yellow solid compound 8 (122 mg, 61% yield).
Synthesis of lonidamine (6)
1H-indole-3-carboxylic acid methyl ester (706 mg,4.0 mmol) was added to 5.0mL of 30% aqueous sodium hydroxide solution in a 25mL round bottom flask, heated to 100℃and then 2, 4-dichlorobenzyl chloride (860 mg,4.4 mmol) was slowly added dropwise, followed by rapid stirring for 4 hours, then cooling to room temperature, adding 20% hydrochloric acid solution to adjust the reaction system to acidic (pH=1), stirring for 0.5H, then filtering, and recrystallizing the product with glacial acetic acid to give the target drug molecule lonidamine compound 2 (836 mg, 65% yield).
1 H-NMR(400MHz,CD 3 OD):8.28(d,J=8.2Hz,1H),7.66(d,J=8.6Hz,1H),7.60(d,J=2.1Hz,1H),7.57-7.52(m,1H),7.45-7.39(m,1H),7.30(dd,J=8.4;2.1Hz,1H),6.92(d,J=8.4Hz,1H),5.89(s,2H)。
Synthesis of 1- (2, 4-dichlorobenzyl) -1H-indazole-3-carboxylic acid 2- ((2-hydroxyethyl) disulfonyl) ethyl ester (7) LND (32 mg,0.10 mmol), EDCI (47 mg,0.25 mmol), DMAP (0.23 mg,0.01 mmol), 2' dithiodiethanol (51 mg,0.30 mmol) and 3mL anhydrous DCM were added to a 25mL round bottom flask and reacted at room temperature under nitrogen for 12 hours. After washing with water to remove excess 2-hydroxyethyl disulfide, the organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure. Silica gel column chromatography (methanol: dichloromethane 1:20) gave compound 3 (37 mg, yield 82%).
1 H-NMR(400MHz,CDCl 3 ):8.26(d,J=8.0Hz,1H),7.44-7.35(m,4H),7.09(dd,J=8.4;2.1Hz,1H),6.70(d,J=8.4Hz,1H),5.78(s,2H),4.75(t,J=6.8Hz,2H),3.90(t,J=5.8Hz,2H),3.90(t,J=6.8Hz,2H),2.93(t,J=5.8Hz,2H)。
Synthesis of 2- ((2- (2-) -4- (2, 5-dioxopyrrolidin-1-yl) oxy) carbonyl) oxyethyl) dithio-ethyl 1- (2, 4-dichlorobenzyl) -1H-indazole-3-carboxylate (8)
1- (2, 4-dichlorobenzyl) -1H-indazole-3-carboxylic acid 2- ((2-hydroxyethyl) disulfonyl) ethyl ester (912 mg,2.0 mmol) and triethylamine (242. Mu.L, 2.4 mmol) were dissolved in CH3CN 5mL in a 25mL round bottom flask, N' -succinimidyl carbonate (616 mg,2.4 mmol) was added, stirred at room temperature for 3H and the solvent was removed under reduced pressure, and the residue was isolated by silica gel column chromatography (ethyl acetate: petroleum ether=1:1) to give compound 4 (764 mg, yield 64%).
1 H-NMR(400MHz,CDCl 3 ):8.26(d,J=8.1Hz,1H),7.45-7.33(m,4H),7.10(dd,J=8.4,2.1Hz,1H),6.71(d,J=8.4Hz,1H),5.79(s,2H),4.75(t,J=6.6Hz,2H),4.59(t,J=6.8Hz,2H),3.18(t,J=6.6Hz,2H),3.06(t,J=6.8Hz,2H),2.82(s,4H)。
1 H-NMR(400MHz,DMSO-d 6 ):7.92-7.75(m,15H),6.12-5.86(m,6H),3.63-3.56(m,2H),2.50-2.39(m,2H),1.74-1.65(m,2H)。
Synthesis of LND-SS-Pt-TPP (9)
TPP-Pt (100 mg,0.17 mmol) and 100mg of 2- ((2- ((((2, 5-dioxapyrrolidin-1-yl) oxy) carbonyl) oxy) ethyl) disulfonyl) 1- (2, 4-dichlorobenzyl) -1H-indazole-3-carboxylic acid ethyl ester (100 mg,0.15 mmol) were added to a 10mL round bottom flask and reacted in 8mL DMSO for 72H at room temperature under dark conditions, extracted three times with dichloromethane, the organic layer was collected, dried over anhydrous sodium sulfate and the organic phase was evaporated to dryness under reduced pressure. Isolation by column chromatography on silica gel (methanol: dichloromethane=1:20) afforded compound 9 (96 mg, yield 56%).
1 H-NMR(400MHz,DMSO-d 6 ):8.16(d,J=8.8Hz,1H),7.89-7.74(m,16H),7.69(d,J=2.0Hz,1H),7.52(t,J=8.0Hz,1H),7.41-7.37(m,2H),6.97(d,J=8.4Hz,1H),6.59(s,2H),5.87(s,2H),4.60(t,J=6.0Hz,2H),4.09-3.95(m,2H),3.68-3.59(m,2H),3.16(t,J=6.0Hz,2H),2.90(t,J=6.4Hz,2H),2.38-2.27(m,2H),1.66(s,2H);ESI-MS[M+H] + :1147.08。
Example 2: synthesis of hyaluronic acid mono- (6-ethylenediamine-6-deoxy) -beta-cyclodextrin graft (HA-CD)
1-ethyl-3- (3-dimethylaminopropyl) carbodiimide (EDCI) (335.4 mg,1.75 mmol) and sodium N-hydroxysuccinimide sulfonate salt (NHSS) (380 mg,1.75 mmol) were added to sodium hyaluronate (Mw=46000) (200 mg, 4.34. Mu.M) in PBS phosphate buffer (0.1M, pH 7.2) 60mL in a 100mL round bottom flask, sonicated and stirred at room temperature for 30min. Then, 20mL of mono- (6-ethylenediamine-6-deoxy) - β -cyclodextrin (1.177 g,1.0 mmol) PBS was added and reacted at room temperature for 24 hours. The resulting solution was dialyzed against purified water for 5 days. After lyophilization, white flaky solid HA-CD was obtained.
Example 3: preparation of LND-SS-Pt-TPP/HA-CD nanoparticles
HA-CD 10.0mg was dissolved in 900. Mu.L of ultrapure water. LND-SS-Pt-TPP0.5mg, fully dissolved in 100. Mu.L DMSO, and the drug solution was slowly added to the HA-CD solution in portions with stirring, sonicated for 10min (ultrasound power 250W, working 2s, intermittent 3 s). Dialysis bag (molecular weight cut-off 3500) was dialyzed for 24h and stored in a refrigerator at 4℃for further use.
Table 1: particle size, particle size distribution, surface charge and drug loading of small molecule prodrug self-assembled nanoparticles
Figure BDA0004178254120000051
Figure BDA0004178254120000061
Example 4: CCK-8 method for detecting cell proliferation
Inoculating A549 cells and A549 cisplatin resistant cells in logarithmic growth phase into 96-well plates respectively, adjusting cell density, and culturing in incubator (37 ℃ C., 50mL/L CO 2) with volume of 100 μl each; after the cells are completely adhered, adding the test substances with different concentrations for continuous culture; after incubation for various times, 10. Mu.L of CCK-8 solution was added. After the culture plate is incubated in an incubator for 2 hours, the absorbance OD value at 450nm is measured by an enzyme-labeled instrument, and the cell proliferation inhibition rate IC of each group is calculated 50
TABLE 2CCK8 assay for inhibition of cell proliferation
Figure BDA0004178254120000062
Example 5: cell nucleus, cytoplasm and mitochondria platinum uptake assay
The test sample was co-cultured with the cells for 24 hours, the dead cells were washed off, the remaining adherent cells were washed once with PBS, and then the cells were digested with a membrane enzyme digest (Trypsin-EDTA solution), and collected by centrifugation. 1mL of mitochondrial isolation reagent was added, the cells were gently suspended, and after 15 minutes of ice bath placement, the cell suspension was transferred to a glass cell homogenizer of appropriate size, under homogenization 30. Centrifuging the homogenized suspension at 4deg.C under 1000r/min for 10min to obtain white precipitate as nucleus. Sucking out supernatant in the centrifuge tube, transferring into a new centrifuge tube, and centrifuging at 4deg.C for 10min under 10000r/min to obtain white precipitate on the wall of the centrifuge tube as mitochondria. Separating the supernatant after precipitation to obtain cytoplasm. And adding 20mL of concentrated nitric acid, 20mL of H2O2 and 50mL of concentrated hydrochloric acid into the three separated parts of each sample, carrying out high-temperature digestion until the solution is clarified, and determining the volume of purified water to 1mL, wherein the content of Pt element in each sample is determined by ICP-MS. The concentration of platinum in the mitochondria of LND-SS-Pt-TPP/HA-CD (LP/HA-CD) is far greater than that of cisplatin, and the nanosystem HAs good mitochondrial targeting capability.
TABLE 3 ICP-MS determination of Pt element content in organelles
Figure BDA0004178254120000071
Example 6: fracture mechanism validation
(1) 5mg of prodrug was precisely measured and added to a prepared PBS buffer solution of GSH 10mM at pH 7.2-7.4, and samples were taken at 0.5h, 1h and 2h respectively in a constant temperature shaker at 37℃and analyzed by MS method.
(2) Accurately measuring a proper amount of LND-SS-Pt-TPP in a sample bottle A, adding a PBS buffer solution to prepare a 1mM standard drug solution, accurately measuring a bivalent platinum fluorescent probe in a sample bottle B to prepare a 1mM standard drug solution, adding GSH into the sample bottle C to make GSH concentration be 10mM for the same concentration as the sample bottle A, and adding the bivalent platinum fluorescent probe. The sample bottle A, B, C was placed in a constant temperature shaker at 37℃and the phenomenon was observed after 4 hours of light-shielding reaction.
Experimental results show that LND-SS-Pt-TPP can be finally cracked into lonidamine and bivalent platinum under GSH condition, thereby exerting cell killing effect.
Example 7: self-assembled nanoparticle cell uptake studies
(1) Synthesizing coumarin-6 labeled nanoparticles:
HA-CD10mg was dissolved in 900. Mu.L of ultrapure water. LND-SS-Pt-TPP0.5mg, fully dissolved in 100. Mu. LDMSO, coumarin-6 (C-6) was added, and the drug solution was slowly added to the HA-CD solution in portions with stirring and sonication for 10min (sonication power 250W, working 2s, batch 3 s). The blank group is coumarin-6, and the rest methods are the same.
(2) Cell uptake assay:
after culturing A549 cells for 24 hours, the culture solution is discarded, and the C-6 and C-6 marked nanoparticles (with the same C-6 concentration) diluted by the fresh culture solution are respectively added and are respectively cultured for 3 hours in a CO2 incubator. After the completion of the incubation, the culture solution was aspirated, cell uptake was stopped by adding ice-cold PBS (pH 7.4), the cells were fixed with 4% paraformaldehyde by washing three times with PBS, nuclei were stained with DAPI as a site, and green fluorescence intensity of each cell was observed under a confocal laser microscope.
The result shows that the fluorescence intensity of the coumarin-6 marked LND-SS-Pt-TPP/HA-CD in the cells is stronger than that of coumarin-6, the uptake of the coumarin-6 marked LND-SS-Pt-TPP/HA-CD by the cells is increased, and the good cell targeting capability of the nano system is proved.

Claims (6)

1. A cisplatin-based small molecule prodrug compound characterized by having the structural formula:
Figure FDA0004178254110000011
wherein m is an integer of 1 to 6; n is an integer of 1 to 6.
2. A method of preparing a cisplatin-based small molecule prodrug compound as claimed in claim 1 comprising the steps of:
Figure FDA0004178254110000012
wherein m is an integer of 1 to 6; n is an integer of 1 to 6
(1) Adding hydrogen peroxide into cis-platinum, stirring at room temperature for reaction, filtering, washing, drying to obtain diamino dihydroxy platinum (1) chloride,
(2) Dissolving carboxyalkyl triphenylphosphine bromide, EDCI and NHS in acetonitrile, and stirring at room temperature to obtain an intermediate product TPP-NHS, wherein the carboxyalkyl triphenylphosphine bromide is one or more of (2-carboxyethyl) triphenylphosphine bromide, (3-carboxypropyl) triphenylphosphine bromide, (4-carboxybutyl) triphenylphosphine bromide or (5-carboxypentyl) triphenylphosphine bromide;
(3) Dissolving TPP-NHS in anhydrous DMSO, adding diaminodihydroxyplatinum (1) chloride, reacting for 72h, and then removing DMSO to obtain TPP-Pt;
(4) The lonidamine (2) and the alkyl glycol (3) containing disulfide bond are subjected to esterification reaction under the action of a condensing agent to obtain a compound (4), then the compound is subjected to esterification reaction with N, N' -disuccinimidyl carbonate to obtain an activated ester (5), and the activated ester is esterified with TPP-Pt to obtain a final compound (LND-SS-Pt-TPP); the alkyl glycol containing disulfide bond is one or more of 2,2' -dithiodiethanol, dithiodimethanol, 3' -dithiodipropanol or 4,4' -dithiodibutanol.
3. A small molecule prodrug self-assembled nanoparticle, characterized in that a hyaluronic acid-cyclodextrin (HA-CD) drug-carrying material formed by condensing hyaluronic acid and cyclodextrin is the nanoparticle formed by self-assembling cisplatin-based small molecule prodrug compound according to claim 1.
4. A method for preparing small molecule prodrug self-assembled nanoparticles according to claim 3, comprising the steps of: (1) HA-CD preparation: 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide (EDCI) and N-hydroxysuccinimide sulfonate sodium salt (NHSS) were added to sodium hyaluronate in PBS phosphate buffer and stirred at room temperature for 30min; then adding amino-substituted cyclodextrin, reacting for 24 hours at room temperature, dialyzing, and freeze-drying to obtain the HA-CD drug-loaded material;
(2) The preparation method of the LND-SS-Pt-TPP/HA-CD drug-loaded nano system comprises the following steps: dissolving LND-SS-Pt-TPP in an organic solvent, slowly adding the drug solution into an HA-CD aqueous solution under the condition of stirring, performing ultrasonic treatment, dialyzing to remove the organic solvent, and freeze-drying to obtain LND-SS-Pt-TPP/HA-CD drug-loaded nanoparticles; the organic solvent comprises one or more of methanol, ethanol, tetrahydrofuran, N-Dimethylformamide (DMF) or dimethyl sulfoxide (DMSO).
5. Use of a cisplatin-based small molecule prodrug compound as defined in claim 1 or a small molecule prodrug self-assembled nanoparticle as defined in claim 3 for the preparation of an antitumor drug.
6. Use of a cisplatin-based small molecule prodrug compound as defined in claim 1 or small molecule prodrug self-assembled nanoparticles as defined in claim 3 for the preparation of an injectable, oral or topical drug delivery system.
CN202310397921.5A 2023-04-14 2023-04-14 Preparation and application of cisplatin prodrug self-assembled nanoparticles for overcoming cisplatin resistance Active CN116410234B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310397921.5A CN116410234B (en) 2023-04-14 2023-04-14 Preparation and application of cisplatin prodrug self-assembled nanoparticles for overcoming cisplatin resistance

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310397921.5A CN116410234B (en) 2023-04-14 2023-04-14 Preparation and application of cisplatin prodrug self-assembled nanoparticles for overcoming cisplatin resistance

Publications (2)

Publication Number Publication Date
CN116410234A true CN116410234A (en) 2023-07-11
CN116410234B CN116410234B (en) 2024-04-19

Family

ID=87057745

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310397921.5A Active CN116410234B (en) 2023-04-14 2023-04-14 Preparation and application of cisplatin prodrug self-assembled nanoparticles for overcoming cisplatin resistance

Country Status (1)

Country Link
CN (1) CN116410234B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190345181A1 (en) * 2017-12-27 2019-11-14 Anhui University Tetravalent Platinum Compound-Bicyclic Double-bond-containing Amphiphilic Polymer Prodrug, Nano-micelle, Preparation Method and Application Thereof
CN110787146A (en) * 2019-09-25 2020-02-14 中国人民解放军第四军医大学 Preparation method and application of redox-responsive tumor-targeted cisplatin nano drug delivery system
CN110981918A (en) * 2019-12-20 2020-04-10 华中科技大学 Modified platinum compound and preparation method and application thereof
CN112961188A (en) * 2021-02-07 2021-06-15 山东省第二人民医院(山东省耳鼻喉医院、山东省耳鼻喉研究所) Tetravalent platinum prodrug platinum benzydate, preparation thereof, preparation method and application
CN114432269A (en) * 2022-01-28 2022-05-06 中国人民解放军空军军医大学 Preparation method and application of stimuli-responsive nano drug delivery system

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190345181A1 (en) * 2017-12-27 2019-11-14 Anhui University Tetravalent Platinum Compound-Bicyclic Double-bond-containing Amphiphilic Polymer Prodrug, Nano-micelle, Preparation Method and Application Thereof
CN110787146A (en) * 2019-09-25 2020-02-14 中国人民解放军第四军医大学 Preparation method and application of redox-responsive tumor-targeted cisplatin nano drug delivery system
CN110981918A (en) * 2019-12-20 2020-04-10 华中科技大学 Modified platinum compound and preparation method and application thereof
CN112961188A (en) * 2021-02-07 2021-06-15 山东省第二人民医院(山东省耳鼻喉医院、山东省耳鼻喉研究所) Tetravalent platinum prodrug platinum benzydate, preparation thereof, preparation method and application
CN114432269A (en) * 2022-01-28 2022-05-06 中国人民解放军空军军医大学 Preparation method and application of stimuli-responsive nano drug delivery system

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HUANG, YAXIN, ET AL.: ""The potential of lonidamine in combination with chemotherapy and physical therapy in cancer treatment"", 《CANCERS 》, vol. 12, no. 11, 11 November 2020 (2020-11-11), pages 3332 *
YULIA N. NOSOVA ET AL.: ""Enhancing the Cytotoxic Activity of Anticancer PtIV Complexes by Introduction of Lonidamine as an Axial Ligand"", 《EUR.J.INORG.CHEM》, 31 December 2017 (2017-12-31), pages 1785 - 1791 *

Also Published As

Publication number Publication date
CN116410234B (en) 2024-04-19

Similar Documents

Publication Publication Date Title
Zhang et al. Targeted delivery of a mannose‐conjugated BODIPY photosensitizer by nanomicelles for photodynamic breast cancer therapy
CN106177977B (en) Ternary conjugate of antitumor drug, synthesis and application
CN107674210B (en) Triphenylphosphine-chitosan stearic acid graft drug-loaded micelle, preparation and application thereof
KR102237234B1 (en) Integrated nanosystem for co-transporting genes/drugs with liver targeting and method for manufacturing the same
Yang et al. Scutellarin-cyclodextrin conjugates: synthesis, characterization and anticancer activity
EP1781707A1 (en) Hyaluronic acid butyric esters with a low degree of substitution, procedure for their preparation and use
JP7055881B2 (en) Preparation and use of new photosensitizer composite nano-multifunctional materials
CN105175656B (en) A kind of temperature and oxidant stimuli-responsive Micelle-like Nano-structure of Two preparation method and application
Wang et al. Intracellular GSH-activated galactoside photosensitizers for targeted photodynamic therapy and chemotherapy
CN109503455A (en) It is a kind of using tetraphenyl ethylene indole derivatives as the nanocomposite of photosensitizer and preparation method and the application in terms of oncotherapy
CN111888480A (en) Method for anchoring and modifying nano-drug on surface of living cell
CN108329404B (en) IR-780 iodide-chitosan stearic acid graft and preparation and application thereof
WO2023160354A1 (en) Novel acid-sensitive aptamer triptolide conjugate and application
Zhang et al. Near-infrared BODIPY-paclitaxel conjugates assembling organic nanoparticles for chemotherapy and bioimaging
US20090209485A1 (en) Modulating angiogenesis with nod factors such as glucosamine oligosaccharides
Jia et al. Small molecular target-based multifunctional upconversion nanocomposites for targeted and in-depth photodynamic and chemo-anticancer therapy
Dong et al. pH-responsive Mannose-modified ferrocene Metal-Organic frameworks with rare earth for Tumor-targeted synchronous Chemo/Chemodynamic therapy
Gang et al. Synthesis and biological evaluation of fluorescent hyaluronic acid modified amorphous calcium phosphate drug carriers for tumor-targeting
Tang et al. Rationally designed multifunctional nanoparticles as GSH-responsive anticancer drug delivery systems based on host-guest polymers derived from dextran and β-cyclodextrin
CN104926912B (en) Synthesis and purpose of glycyrrhetinic acid derivative
CN116410234B (en) Preparation and application of cisplatin prodrug self-assembled nanoparticles for overcoming cisplatin resistance
CN111848658B (en) Mitochondria-targeted fluoroborodipyrrole compound, preparation method and application of liposome-coated nano particles thereof
Zhu et al. Reduction responsive BODIPY decorated mesoporous silica nanoscale platforms for photodynamic therapy
CN108727437A (en) One curcuminoids metal aryl complex and its synthetic method and application
Assali et al. Self-assembly of diclofenac prodrug into nanomicelles for enhancing the anti-inflammatory activity

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