CN115300637B - Chalcogen hybrid bond bridged dimer prodrug, self-assembled nanoparticle thereof, preparation method and application - Google Patents

Chalcogen hybrid bond bridged dimer prodrug, self-assembled nanoparticle thereof, preparation method and application Download PDF

Info

Publication number
CN115300637B
CN115300637B CN202210776933.4A CN202210776933A CN115300637B CN 115300637 B CN115300637 B CN 115300637B CN 202210776933 A CN202210776933 A CN 202210776933A CN 115300637 B CN115300637 B CN 115300637B
Authority
CN
China
Prior art keywords
prodrug
self
dtx
preparation
hybrid
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.)
Active
Application number
CN202210776933.4A
Other languages
Chinese (zh)
Other versions
CN115300637A (en
Inventor
孙进
孙丙军
刘钿
何仲贵
罗聪
董付丹
李凌霄
左诗意
王硕
黄乐童
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.)
Shenyang Pharmaceutical University
Original Assignee
Shenyang 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 Shenyang Pharmaceutical University filed Critical Shenyang Pharmaceutical University
Priority to CN202210776933.4A priority Critical patent/CN115300637B/en
Publication of CN115300637A publication Critical patent/CN115300637A/en
Application granted granted Critical
Publication of CN115300637B publication Critical patent/CN115300637B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

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
    • A61K47/55Medicinal 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 the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • 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
    • A61K47/56Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • 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
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D305/00Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms
    • C07D305/14Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms condensed with carbocyclic rings or ring systems
    • 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
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

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

Abstract

Chalcogen hybrid bond bridged dimer prodrug, self-assembled nanoparticle thereof, preparation method and application thereof, belongs to the technical field of medicines, and relates to construction of redox double-sensitive sulfur tellurium sulfide and sulfur selenium sulfur hybrid bond bridged prodrug and self-assembled nanoparticle thereof, and application thereof in drug delivery. The preparation method is simple and easy to implement, and can effectively cope with tumor heterogeneous microenvironment by utilizing the redox double hypersensitivity of the chalcogenides, so that the intelligent response activation of the prodrug in tumor cells is realized, and the anti-tumor effect and safety of the prodrug are ensured. Provides a new strategy and selection for solving the limitation of tumor heterogeneous microenvironment on nano drug release, and meets the urgent need of high-efficiency chemotherapy preparation in clinic.

Description

Chalcogen hybrid bond bridged dimer prodrug, self-assembled nanoparticle thereof, preparation method and application
Technical Field
The invention belongs to the technical field of medicines, relates to a chalcogen hybrid bond bridged dimer prodrug, a preparation method and application of self-assembled nanoparticles thereof, and in particular relates to construction of a sulfur tellurium sulfur hybrid bond and sulfur selenium sulfur hybrid bond bridged redox double-sensitive docetaxel dimer prodrug and dimer prodrug self-assembled nanoparticles and application of the self-assembled nanoparticles in drug delivery.
Background
Cancer is a serious threat to human health. Drug therapy is one of the most common strategies for current cancer treatment, especially for advanced tumors, tumors that cannot be resected by surgery, and tumors that have metastasized and spread. The existing preparation technology in clinic has low delivery efficiency, and the anti-tumor drugs have the problems of poor curative effect and serious adverse reaction. Docetaxel, for example, is used clinically as a first-line chemotherapeutic agent for treating breast cancer, non-small cell lung cancer, prostate cancer, and the like. However, docetaxel has very low water solubility, and the commercially available injection Taxotere (Taxotere) uses tween 80 as a solubilizer, which causes severe allergic reaction. In addition, docetaxel has poor tumor selectivity, and the wide distribution in vivo after intravenous injection can cause serious toxic and side effects such as bone marrow suppression, allergic pneumonia and the like. These drawbacks greatly limit the clinical use of docetaxel. Therefore, there is a need to construct an anti-tumor drug delivery system that is highly efficient-low toxic.
In recent years, prodrug technology and nanotechnology have greatly enriched the delivery strategies of antitumor drugs, and a number of formulations have been successfully marketed, such as capecitabine, paclitaxel for injection (albumin binding), doxorubicin hydrochloride liposomes, and the like. The prodrug technology can effectively improve the adverse properties of the drug, but the small molecule prodrug still has the problems of poor pharmacokinetic behavior and low tumor targeting efficiency. The nanometer drug delivery system can effectively prolong the in vivo circulation time of the drug, and can improve the accumulation of the drug at the tumor part through active targeting or passive targeting. However, the traditional nano preparation is often loaded with the medicine in a physical embedding way, and has the defects of low medicine loading rate (generally less than 10%), poor safety of carrier materials and the like. In contrast, the small molecular prodrug self-assembled nanoparticle combines the advantages of the prodrug and the nanotechnology, the drug loading capacity of the preparation is high, adverse reactions caused by carrier materials can be avoided, the preparation process is simple, the reproducibility is good, and the application transformation potential is good.
Homodimer prodrug self-assembled nano-scale has ultra-high drug loading (greater than 50%), and is becoming a potential anticancer drug delivery technology. However, poor self-assembly capability and insufficient activation of targets become two key factors restricting the self-assembly of homodimeric prodrugs for nano-applications. Designing and developing self-assembled nano-sized dimeric prodrugs with good assembly stability and tumor selective bioactivity remains a significant challenge. In addition, tumor cells exhibit redox heterogeneity in their microenvironment due to irregular production of glutathione and reactive oxygen species. This heterogeneity exists in different tumors, even in different areas of the same tumor and in different stages of development. Prodrugs that release active agents only for one environmental stimulus may have limited efficacy. Therefore, there is a need to develop a novel drug delivery system with ultra-high dual sensitivity to tumor redox heterogeneity.
Disclosure of Invention
The invention aims to design and synthesize a dimer prodrug containing a chalcogenides bond bridge, prepare a self-assembled nano drug delivery system of the prodrug and application of the self-assembled nano drug delivery system in drug delivery. The influence of different chemical bridging on the stability, drug release, cytotoxicity, pharmacokinetics, tissue distribution and pharmacodynamics of the prodrug self-assembly nanoparticles is discussed, and the chemical bridging with the best effect is comprehensively screened, so that a new strategy and more choices are provided for developing an intelligent response type drug delivery system for a tumor microenvironment, and the urgent requirements of clinical high-efficiency chemotherapeutic agents are met.
In order to achieve the above object, the present invention provides a redox-sensitive chalcogenides bridged dimer prodrug represented by the general formula (I).
Wherein X is selected from Te, se, C, S, and Drug is a Drug containing hydroxyl, amino or carboxyl. The medicine containing hydroxyl, amino or carboxyl is selected from antitumor medicines, antimetabolites and anti-inflammatory medicines, and the antitumor medicines are selected from taxane, anthraquinone, nucleoside, camptothecine, platinum, vinblastine, poisins and artemisinin compounds; the antimetabolite is selected from pyrimidine, purine, tabine and folic acid; the anti-inflammatory agent is selected from halofantrine, griseofulvin, and perimycin A, and their derivatives. Further, X is selected from Te and Se; the drug containing hydroxyl, amino or carboxyl is docetaxel.
The invention selects Docetaxel (DTX) as a model drug, and prepares a redox double-sensitive docetaxel dimer prodrug containing a sulfur tellurium hybridization bond, a sulfur selenium hybridization bond, a trisulfide bond or a sulfur carbon hybridization bond bridge by connecting the drug through 3,3 '-tellurium dithiodipropionic acid, 3' -selenium dithiodipropionic acid, 3 '-trithiodipropionic acid or 3,3' -methylene dithiodipropionic acid.
The docetaxel dimer prodrug bridged by a sulfur tellurium sulfur hybrid bond has the structure that:
the docetaxel dimer prodrug bridged by a sulfur selenium sulfur hybrid bond has the structure that:
docetaxel dimer structures bridged with trisulfide and thiocarbon sulfide bonds are respectively as follows:
the invention provides a method for synthesizing docetaxel dimer prodrug containing sulfur tellurium hybridization bond, sulfur selenium hybridization bond, three sulfur bond and sulfur carbon sulfur hybridization bond bridging, which comprises the following steps:
respectively dissolving 3,3 '-tellurium-dithiodipropionic acid, 3' -selenium-dithiodipropionic acid, 3 '-trithiodipropionic acid and 3,3' -methylene dithiodipropionic acid in dichloromethane, and uniformly stirring; dissolving 4-Dimethylaminopyridine (DMAP), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (EDCI) and docetaxel in anhydrous dichloromethane, stirring, and mixing with the above solutions of 3,3 '-tellurium dithiodipropionic acid, 3' -selenious dithiodipropionic acid, 3 '-trithiodipropionic acid and 3,3' -methylenedithiodipropionic acid, respectively, N 2 Stirring at room temperature for 10-12h under the protection; adding EDCI and DMAP, N into the mixed solution 2 Under the protection, stirring is continued for 12-24 hours at room temperature, and the obtained product is separated and purified by a preparation liquid phase.
In the preparation method, the docetaxel can be replaced by other anticancer drugs containing active hydroxyl, amino or carboxyl. The anticancer drug containing active hydroxyl, amino or carboxyl is selected from other taxanes, anthraquinone, nucleoside, platinum, vinblastine, poisoside, artemisinin and camptothecins.
A pharmaceutical composition comprising the chalcogenides bridged dimer prodrug, and a pharmaceutically acceptable carrier and excipient.
The invention also provides application of the chalcogen hybrid bond bridge Lian Duo sitaxe dimer prodrug or a pharmaceutical composition containing the prodrug in preparation of antitumor drugs.
The invention also provides application of the chalcogen hybrid bridge Lian Duo sitaxe dimer prodrug or a pharmaceutical composition containing the prodrug in preparation of a drug delivery system.
The invention also provides application of the chalcogen hybrid bridge Lian Duo sitaxe dimer prodrug or a pharmaceutical composition containing the prodrug in preparing injection, oral administration or local administration systems.
The invention also provides self-assembled nanoparticles of the chalcogenides bridged dimer prodrug, which can be non-PEG dimer prodrug self-assembled nanoparticles or PEG modified dimer prodrug self-assembled nanoparticles. The preparation method is a nano precipitation method, including a high-speed stirring method and an ultrasonic method.
The preparation method of the chalcogen hybrid bond bridged dimer prodrug self-assembled nanoparticle comprises the following steps:
dissolving the chalcogenides bridged dimer prodrug and a PEG modifier (the self-assembled nano-particles of the non-PEGylated dimer prodrug are not added with the PEG modifier) into a solvent, slowly dripping the solution into water under stirring, spontaneously forming uniform nano-particles by the prodrug, and removing the solvent by a reduced pressure distillation method to obtain a nano-colloid solution without an organic solvent.
The PEG modifier is selected from TPGS, DSPE-PEG, PLGA-PEG and PE-PEG, and the preferred PEG modifier is DSPE-PEG. The molecular weight of the PEG is 1000-5000, preferably 1000, 2000 and 5000, more preferably the molecular weight of the PEG is 2000.
The solvent is selected from ethanol, dimethyl sulfoxide, N' -dimethylformamide, tetrahydrofuran and acetone.
The weight ratio of the chalcogen hybrid bond bridging dimer prodrug to the PEG modifier is 90:10-70:30, and under the condition of the range, the prodrug nanoparticles can exert better anti-tumor effect.
The invention also provides application of the chalcogen hybrid bond bridging dimer prodrug self-assembled nanoparticle in preparation of a drug delivery system.
The invention also provides application of the chalcogen hybrid bond bridging dimer prodrug self-assembled nanoparticle in preparation of antitumor drugs.
The invention also provides application of the self-assembled nano-particles of the chalcogenides bridged dimer prodrug in preparing injection administration, oral administration or local administration systems.
The technical problem solved by the invention is that a sulfur tellurium sulfur hybrid bond and a sulfur selenium sulfur hybrid bond are introduced into a prodrug and self-assembled nano particles, a redox double-sensitive dimer prodrug bridged by a sulfur hybrid bond is designed, and the dimer prodrug is used for constructing the self-assembled nano particles, so that the high drug loading, the good stability, the low toxic and side effect and the quick drug release of tumor site specificity are realized, and the treatment effect is improved. Meanwhile, dimer prodrugs containing trisulfide bonds and thiocarbon-sulfide hybrid bonds are used as a control, and differences of different chemical bridges in self-assembly, redox-sensitive response, anti-tumor activity and the like are examined, and influences on stability, drug release, cytotoxicity, pharmacokinetics, tissue distribution and pharmacodynamics of the prodrug self-assembly nanoparticles are also examined.
The invention has the advantages that:
(1) The dimer prodrug containing the chalcogen hybrid bond bridging and the control prodrug containing the trithio bond and the sulfur-carbon hybrid bond bridging are designed and synthesized, and the synthesis method is simple and easy to implement;
(2) The preparation method is simple and easy to implement, realizes high-efficiency entrapment of the medicine, has the ultrahigh drug loading rate of more than 67 percent and the stability of 10 percent FBS in phosphate buffer solution for 24 hours, and basically keeps the particle diameter unchanged in 35 days at 4 ℃;
(3) Differences of different chemical bridging in self-assembly, redox sensitive response capability, anti-tumor activity and the like are examined, and influences on stability, drug release, cytotoxicity, pharmacokinetics, tissue distribution and pharmacodynamics of prodrug self-assembly nanoparticles are examined. And the comprehensive experimental result shows that the sulfur tellurium-sulfur hybrid bond prodrug has higher redox double-hypersensitive characteristic, and can better cope with tumor redox heterogeneous microenvironment. Meanwhile, the prodrug nanoparticle with the sulfur tellurium-sulfur hybrid bond has the best assembling capability. The invention provides a new strategy and more choices for developing an intelligent response type drug delivery system for tumor microenvironment, and meets the urgent requirements of high-efficiency chemotherapeutics in clinic.
Drawings
FIG. 1 is a structural confirmation of the sulfur tellurium-sulfur hybrid bridge Lian Duo Sitazidime prodrug (DTX-STeS-DTX) of example 1 of the present invention.
A: DTX-STeS-DTX 1 H-NMR spectrum.
B: mass spectrum of DTX-stis-DTX.
C: high performance liquid chromatography purity profile for DTX-stis-DTX.
FIG. 2 is a structural confirmation of the S-Se-S hybrid bridge Lian Duo Sitazidime prodrug (DTX-SSeS-DTX) in example 2 of the present invention.
A: DTX-SSeS-DTX 1 H-NMR spectrum.
B: mass spectrum of DTX-SSeS-DTX.
C: high performance liquid chromatography purity profile for DTX-SSeS-DTX.
FIG. 3 is a structural confirmation of the disulfide bridge Lian Duo cetime dimer prodrug (DTX-SSS-DTX) in example 3 of the present invention.
A: DTX-SSS-DTX 1 H-NMR spectrum.
B: mass spectrum of DTX-SSS-DTX.
C: high performance liquid chromatography purity map for DTX-SSS-DTX.
FIG. 4 shows the structural confirmation of the thiocarbothioic bridge Lian Duo Sitazidime prodrug (DTX-SCS-DTX) in example 4 of the present invention.
A: DTX-SCS-DTX 1 H-NMR spectrum.
B: mass spectrum of DTX-SCS-DTX.
C: high performance liquid chromatography purity map for DTX-SCS-DTX.
Fig. 5 is a graph of the particle size of self-assembled nanoparticles of the chalcogen hybrid bridge Lian Duo docetaxel dimer prodrug prepared in example 5 of the present invention and a transmission electron microscope graph.
A: particle size diagram of self-assembled nanoparticles of chalcogen hybrid bridge Lian Duo cetioxel dimer prodrug.
B: transmission electron microscopy of self-assembled nanoparticles of chalcogen hybrid bridge Lian Duo cetime dimer prodrug.
FIG. 6 is a graph of particle size versus colloidal stability for non-PEGylated chalcogenides bridged Lian Duo cetime dimer prodrug self-assembled nanoparticles in example 6 of the present invention.
FIG. 7 is a graph of particle size versus colloidal stability of PEGylated chalcogenides bridged Lian Duo cetime dimer prodrug self-assembled nanoparticles in example 6 of the present invention.
A: the storage stability of the PEGylated chalcogen hybrid bridge Lian Duo cetime dimer prodrug self-assembled nanoparticles at 4 ℃.
B: stability of pegylated chalcogenides bridge Lian Duo cetracetam dimer prodrug self-assembled nanoparticles in PBS containing 10% fbs.
FIG. 8 is a graph showing in vitro release assays of self-assembled nanoparticles of a chalcogen hybrid bridge Lian Duo sitaxe dimer prodrug in example 7 of the present invention.
A: self-assembled nanoparticles of chalcogen hybrid bridge Lian Duo cetioxel dimer prodrug in 1mM H 2 O 2 In vitro release profile under conditions.
B: self-assembled nanoparticles of chalcogen hybrid bridge Lian Duo cetioxel dimer prodrug at 10mM H 2 O 2 In vitro release profile under conditions.
C: self-assembled nanoparticles of chalcogen hybrid bridge Lian Duo cetioxel dimer prodrug at 50mM H 2 O 2 In vitro release profile under conditions.
D: in vitro release assay of chalcogen hybrid bridge Lian Duo cetime dimer prodrug self-assembled nanoparticles under 0.01mM DTT conditions.
E: in vitro release assay of chalcogen hybrid bridge Lian Duo cetioxel dimer prodrug self-assembled nanoparticles under 0.1mM DTT conditions.
F: in vitro release assay of chalcogen hybrid bridge Lian Duo cetioxel dimer prodrug self-assembled nanoparticles under 1mM DTT conditions.
FIG. 9 is a graph showing cytotoxicity of self-assembled nanoparticles of a chalcogen hybrid bridge Lian Duo sitaxe dimer prodrug in example 8 of the present invention.
A: half inhibition concentration of chalcogen hybrid bridge Lian Duo cetioxel dimer prodrug self-assembled nanoparticles on 4T1 cells.
B: half inhibitory concentration of the chalcogen hybrid bridge Lian Duo cetioxel dimer prodrug self-assembled nanoparticles on B16F10 cells.
C: semi-inhibitory concentration of the chalcogen hybrid bridge Lian Duo cetioxel dimer prodrug self-assembled nanoparticles on Hepa1-6 cells.
D: half inhibition concentration of the chalcogen hybrid bridge Lian Duo cetioxel dimer prodrug self-assembled nanoparticles on 3T3 cells.
FIG. 10 is a graph of blood concentration versus time for self-assembled nanoparticles of a chalcogen hybrid bridge Lian Duo sitaxe dimer prodrug in example 9 of the present invention.
A: a blood concentration versus time profile of prodrugs of self-assembled nanoparticles of chalcogen hybrid bridge Lian Duo cilexetil dimer prodrug.
B: a blood concentration versus time profile of docetaxel of the chalcogen hybrid bridge Lian Duo docetaxel dimer prodrug self-assembled nanoparticle.
C: a blood concentration versus time profile of the prodrug of the chalcogen hybrid bridge Lian Duo docetaxel dimer prodrug self-assembled nanoparticle and the total amount of docetaxel.
FIG. 11 is an in vivo anti-tumor experimental graph of self-assembled nano-particles of the chalcogen hybrid bridge Lian Duo sitaxe dimer prodrug in example 10 of the present invention.
A: chalcogen hybrid bridge Lian Duo cetime dimer prodrug self-assembled nanoparticles affect the growth of Balb/C mouse breast cancer subcutaneous tumors.
B: the self-assembled nanoparticle of the chalcogen hybrid bridge Lian Duo cetioxel dimer has an effect on the body weight of tumor-bearing mice.
C: the self-assembled nanoparticle of the chalcogen hybrid bridge Lian Duo cetime dimer has an effect on the tumor bearing rate of Balb/C mice.
D: tumor photographs of Balb/C tumor-bearing mice after treatment with chalcogen hybrid bridge Lian Duo cetuximab dimer prodrug self-assembled nanoparticles.
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 Tie-S-Tx dimer prodrug (DTX-STeS-DTX) of Tie-S-bridge Lian Duo
0.25mmol of 3,3 '-tellurium-ene dithiodipropionic acid was dissolved in 10mL of methylene chloride, and 0.05mmol of 4-Dimethylaminopyridine (DMAP) and 1mmol of 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (EDCI) and 0.5mmol of docetaxel were dissolved in 20mL of anhydrous methylene chloride and mixed with a methylene chloride solution of 3,3' -tellurium-ene dithiodipropionic acid, and stirred at room temperature for 10-12 hours. Then adding 0.5mmol of 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride and 0.05mmol of 4-dimethylaminopyridine, stirring at room temperature for 12-24h, wherein the whole reaction process is N 2 Under protection, and the obtained product is separated and purified by a preparation liquid phase.
The structure of DTX-STeS-DTX obtained in example 1 was determined by mass spectrometry and nuclear magnetic resonance hydrogen spectrometry, and the results are shown in FIG. 1.
The results of nuclear magnetic resonance spectroscopy are as follows: DTX-STeS-DTX
1 H NMR(600MHz,CDCl 3 )d ppm 8.108(d,4H,Ar-H,J=7.61Hz),7.612(t,2H,Ar-H,J=7.37Hz),7.506(t,4H,Ar-H,J=7.72Hz),7.394(t,4H,Ar-H,J=7.53Hz),7.313(dd,6H,Ar-H,J=15.15,7.34Hz),6.227(s,2H,13-CH),5.686(d,2H,2-CH,J=6.90Hz),5.467(s,2H,3’-CH),5.408(s,2H,10-CH),5.212(s,2H,2’-CH),4.970(d,2H,5-CH,J=9.21Hz),4.328(d,2H,20-CH 2 -αH,J=8.55Hz),4.264(d,2H,7-CH,J=12.02Hz),4.197(d,2H,20-CH 2 -βH,J=8.48Hz),3.926(d,2H,3-CH,J=6.31Hz),3.289(s,4H,CH 2 2 CHSTeSCH 2 CH 2 ),2.829(m,2H, 2 CHCH 2 STeSCH 2 2 CH-αH),2.599(m,2H, 2 CHCH 2 STeSCH 2 CH 2 -βH),2.425(s,6H,-OAc),2.007(s,2H,6-CH 2 -αH),1.880-1.830(m,2H,6-CH 2 -βH),1.747(s,6H,19-CH 3 ),1.580(s,4H,14-CH 2 ),1.343(s,18H,-Boc),1.223(s,6H,16-CH 3 ),1.121(s,6H,17-CH 3 )。
The mass spectrum was found to be MS (ESI) M/z [ M+Na] + = 1919.59037. Purity results showed that DTX-STeS-DTX purity was 99.01%, meets the requirement of subsequent experiments.
Example 2: the preparation of the thioselenothioyl bridge Lian Duo sitaxe dimer prodrug (DTX-SSeS-DTX) was carried out by the method of example 1, and the 3,3 '-tellurium-free dithiodipropionic acid was changed to 3,3' -selenium-free dithiodipropionic acid to prepare the thioselenothioyl bridge Lian Duo sitaxe dimer prodrug.
The structure of DTX-SSeS-DTX obtained in example 2 was determined by mass spectrometry and hydrogen nuclear magnetic resonance spectrometry, and the results are shown in FIG. 2.
The results of nuclear magnetic resonance spectroscopy are as follows: DTX-SSeS-DTX
1 H NMR(600MHz,CDCl 3 )d ppm 8.109(d,4H,Ar-H,J=7.55Hz),7.612(t,2H,Ar-H,J=7.30Hz),7.506(t,4H,Ar-H,J=7.74Hz),7.393(t,4H,Ar-H,J=7.54Hz),7.305(dd,6H,Ar-H,J=15.57,7.36Hz),6.228(s,2H,13-CH),5.679(d,2H,2-CH,J=6.82Hz),5.470(s,2H,3’-CH),5.414(s,2H,10-CH),5.214(s,2H,2’-CH),4.974(d,2H,5-CH,J=9.21Hz),4.321(d,2H,20-CH 2 -αH,J=8.53Hz),4.248(s,2H,7-CH),4.191(d,2H,20-CH 2 -βH,J=8.51Hz),3.921(d,2H,3-CH,J=6.11Hz),3.111(s,4H,CH 2 2 CHSSeSCH 2 CH 2 ),2.934-2.856(m,2H, 2 CHCH 2 SSeSCH 2 2 CH-αH),2.643-2.564(m,2H, 2 CHCH 2 SSeSCH 2 CH 2 -βH),2.429(s,6H,-OAc),2.006(s,2H,6-CH 2 -αH),1.858(dd,2H,6-CH 2 -βH),1.746(s,6H,19-CH 3 ),1.611(s,4H,14-CH 2 ),1.343(s,18H,-Boc),1.232(d,6H,16-CH 3 ),1.121(s,6H,17-CH 3 )。
The mass spectrum was found to be MS (ESI) M/z [ M+Na] + = 1891.58113. Purity results showed that DTX-SSeS-DTX purity was 99.94% and met the requirements of the subsequent experiments.
Example 3: synthesis of Trisulfide bridge Lian Duo Cyclobetasol dimer prodrug (DTX-SSS-DTX)
Trisulfide bridge Lian Duo cetracetam dimer prodrug was prepared by the preparation method of example 1, changing 3,3 '-tellurium-free dithiodipropionic acid to 3,3' -trithiodipropionic acid.
The structure of DTX-SSS-DTX obtained in example 3 was determined by mass spectrometry and hydrogen nuclear magnetic resonance spectrometry, and the results are shown in FIG. 3.
The results of nuclear magnetic resonance spectroscopy are as follows: DTX-SSS-DTX
1 H NMR(600MHz,CDCl 3 )d ppm 8.109(d,4H,Ar-H,J=7.59Hz),7.612(t,2H,Ar-H,J=7.36Hz),7.506(t,4H,Ar-H,J=7.74Hz),7.393(t,4H,Ar-H,J=7.56Hz),7.304(dd,6H,Ar-H,J=16.65,7.32Hz),6.226(s,2H,13-CH),5.680(d,2H,2-CH,J=6.90Hz),5.468(d,2H,,3’-CH,J=0.90Hz),5.412(s,2H,10-CH),5.213(s,2H,2’-CH),4.973(d,2H,5-CH,J=9.01Hz),4.322(d,2H,20-CH 2 -αH,J=8.54Hz),4.285-4.222(m,2H,7-CH),4.190(d,2H,20-CH 2 -βH,J=8.51Hz),3.922(d,2H,3-CH,J=6.33Hz),3.016(d,4H,CH 2 2 CHSSSCH 2 CH 2 ),2.919(td,2H, 2 CHCH 2 SSSCH 2 2 CH-αH),2.636-2.566(m,2H, 2 CHCH 2 SSSCH 2 CH 2 -βH),2.427(s,6H,-OAc),2.006(s,2H,6-CH 2 -αH),1.884-1.816(m,2H,6-CH 2 -βH),1.746(s,6H,19-CH 3 ),1.599(s,4H,14-CH 2 ),1.344(s,18H,-Boc),1.253-1.204(m,6H,16-CH 3 ),1.121(s,6H,17-CH 3 )。
The mass spectrum was found to be MS (ESI) M/z [ M+Na] + =1843.63295,[M+K] + = 1859.60525. Purity results showed that DTX-SSS-DTX purity was 99.07% and met the requirements of the subsequent experiments.
Example 4: synthesis of thiocarbothioic bridge Lian Duo Cyclobetasol dimer prodrug (DTX-SCS-DTX)
Thiocarbamate Lian Duo cetime dimer prodrug was prepared by the preparation method of example 1, by changing 3,3 '-tellurite dithiodipropionic acid to 3,3' -methylenedithiodipropionic acid.
The structure of DTX-SCS-DTX obtained in example 4 was determined by mass spectrometry and nuclear magnetic resonance hydrogen spectrometry, and the results are shown in FIG. 4.
The results of nuclear magnetic resonance spectroscopy are as follows: DTX-SCS-DTX
1 H NMR(600MHz,CDCl 3 )d ppm 8.106(d,4H,Ar-H,J=7.49Hz),7.611(t,2H,Ar-H,J=7.18Hz),7.506(t,4H,Ar-H,J=7.68Hz),7.389(t,4H,Ar-H,J=7.54Hz),7.293(d,6H,Ar-H,J=8.00Hz),6.228(s,2H,13-CH),5.675(d,2H,2-CH,J=6.37Hz),5.477(d,2H,3’-CH,J=0.68Hz),5.407(s,2H,10-CH),5.219(s,2H,2’-CH),4.973(d,2H,5-CH,J=9.36Hz),4.319(d,2H,20-CH 2 -αH,J=8.55Hz),4.260(dd,2H,7-CH,J=10.84,6.74Hz),4.190(d,2H,20-CH 2 -βH,J=8.43Hz),3.914(d,2H,3-CH,J=4.94Hz),3.581(s,4H,CH 2 2 CHSCH 2 S 2 CHCH 2 ),2.797(s,2H,CH 2 CH 2 S 2 CHSCH 2 CH 2 ),2.797-2.716(m,2H, 2 CHCH 2 SCH 2 SCH 2 2 CH-αH)2.619-2.564(m,2H, 2 CHCH 2 SCH 2 SCH 2 2 CH-βH),2.426(s,6H,-OAc),2.001(d,2H,6-CH 2 -αH,J=5.26Hz),1.929(s,4H,14-CH 2 ),1.885-1.823(m,2H,6-CH 2 -βH),1.743(s,6H,19-CH 3 ),1.338(s,18H,-Boc),1.221(s,6H,16-CH 3 ),1.119(s,6H,17-CH 3 )。
The mass spectrum was found to be MS (ESI) M/z [ M+H] + =1803.69432,[M+Na] + =1825.67840,[M+K] + = 1841.65112. The purity result shows that the purity of DTX-SCS-DTX is 99.80 percent, which meets the requirement of the subsequent experiment.
Example 5: preparation of non-PEGylated/PEG modified chalcogenides bridged Lian Duo cetime dimer prodrug self-assembled nanoparticles
Non-pegylated chalcogenides bridge Lian Duo cetime dimer prodrug nanoparticles: 1mg of the chalcogen hybrid bridge Lian Duo sitaxe dimer prodrug is dissolved in 0.2mL of ethanol, the ethanol solution is slowly added dropwise to 2mL of deionized water under stirring, and nanoparticles spontaneously form. The results are shown in Table 1, where the particle sizes of DTX-STeS-DTX NPs and DTX-SSeS-DTX NPs are the smallest, followed by DTX-SSS-DTX NPs. Whereas the particle size of DTX-SCS-DTX NPs was the largest, indicating poor ability of the thiocarbamate bridge Lian Duo sitaxel dimer prodrug to assemble.
PEGylated chalcogenides bridge Lian Duo Sitazidime prodrug nanoparticles: precisely weighing DSPE-PEG 2k 2mg and 8mg of the chalcogen hybrid bridge Lian Duo cetuximab dimer prodrug were mixed, dissolved in 1mL of ethanol, and the ethanol solution was slowly dropped into 4mL of deionized water with stirring, spontaneously forming uniform nanoparticles DTX-STeS-DTX NPs, DTX-SSeS-DTX NPs, DTX-SSS-DTX NPs and DTX-SCS-DTX NPs. The ethanol in the nano-preparation is removed by dialysis with deionized water at 25 ℃. As shown in Table 2, the particle diameter of the nanoparticle was about 100nm, the particle diameter distribution was less than 0.2, the surface charge was between-12 mV and-20 mV, and the drug loading was about 70%. The particle size and morphology of the PEGylated chalcogenides bridge Lian Duo sitaxe dimer prodrug self-assembled nanoparticles were measured by transmission electron microscopy, and the result is shown in FIG. 5, wherein the transmission electron microscopy shows that the drug-loaded nanoparticles are uniformly spherical, and the average particle size is 100nm.
TABLE 1 particle size and particle size distribution of non-PEG modified dimer prodrug self-assembled nanoparticles
TABLE 2 particle size, particle size distribution, surface Charge and drug loading of PEG-modified dimer prodrug self-assembled nanoparticles
Example 6: colloidal stability assay of non-PEGylated/PEG-modified chalcogenides bridged Lian Duo cetime dimer prodrug self-assembled nanoparticles
(1) Assembly stability of non-PEGylated chalcogenides bridged Lian Duo cetracetam dimer prodrug self-assembled nanoparticles
The self-assembled nanoparticle of PEG-modified chalcogenides bridged Lian Duo cetracetam dimer prepared in example 5 was subjected to room temperature stability study, and the results are shown in fig. 6. DTX-SCS-DTX NPs and DTX-SSS-DTX NPs successively increase in particle size in the first 4 hours; DTX-SSeS-DTX NPs become larger in particle size on day 10; the DTX-STeS-DTX NPs are preferably stable in assembly, and the particle size is always kept around 200nm within 10 days. This suggests that subtle differences in chalcogen composition affect the self-assembly ability of the dimeric prodrug, and the introduction of sulfur selenium sulfur and sulfur tellurium sulfur hybrid bonds can effectively enhance the self-assembly stability of the dimeric prodrug.
(2) Colloidal stability of PEG modified chalcogenides bridge Lian Duo Caesalpoly prodrug self-assembled nanoparticles
Colloidal stability studies were performed on PEG-modified chalcogenides bridge Lian Duo cetime dimer prodrug self-assembled nanoparticles prepared in example 5; taking the particle size change as an index, the long-term storage stability of the prodrug self-assembled nanoparticle at 4 ℃ is examined. As shown in FIG. 7A, the prepared PEG modified chalcogenides bridge Lian Duo sitaxe prodrug self-assembled nanoparticle is placed at 4 ℃ for 35 days, the particle size is not changed obviously, and the PEG modified chalcogenides bridge Lian Duo sitaxe prodrug self-assembled nanoparticle has good long-term storage stability at 4 ℃.
The PEG-modified chalcogeno hybrid bridge Lian Duo cetime dimer prodrug self-assembled nanoparticle was taken out 1mL, added to 20mL of phosphate buffer (PBS, pH 7.4) containing 10% fbs, incubated at 37 ℃ for 24h, and its particle size change was determined by dynamic light scattering at predetermined time points (0, 2, 4, 8, 12 and 24 h). As a result, as shown in FIG. 7B, the DTX-STeS-DTX NPs were excellent in colloidal stability, and no significant change in particle size occurred within 24 hours. In contrast, DTX-SSS-PTX NPs and DTX-SCS-DTX NPs nanoparticles have poor colloidal stability, and the particle size of the nanoparticles increases with the prolongation of incubation time.
Example 7: in vitro release assay of PEG modified chalcogen hybrid bridge Lian Duo Caesalpolyte prodrug self-assembled nanoparticles
The in vitro release of the chalcogen hybrid bridge Lian Duo cetime dimer prodrug self-assembled nanoparticles was examined using Phosphate Buffer (PBS) at pH 7.4 containing 30% ethanol as the release medium. 1mL of PEG-modified chalcogenides bridge Lian Duo docetaxel dimer prodrug self-assembled nanoparticle prepared in example 5 (docetaxel content is 200 μg/mL) was added to 30mL of a release medium, and a certain concentration of hydrogen peroxide (H) was added to the release medium 2 O 2 1mM,10mM,50 mM) or dithiothreitol (DTT, 0.01mM,0.1mM,1mM). Sampling at 37 ℃ at a set time point, and measuring the concentration of the released docetaxel by high performance liquid chromatography to respectively examine the release conditions of the nanoparticles under the oxidation condition and the reduction condition. As shown in FIG. 8, all dimer self-assembled nanoparticles have oxidation response release characteristics with oxidation sensitivity order of DTX-STeS-DTX NPs>DTX-SSeS-DTX NPs>DTX-SSS-DTX NPs≡DTX-SCS-DTX NPs. DTX-STeS-DTX NPs, DTX-SSeS-DTX NPs, DTX-SSS-DTX NPs alone in the presence of DTT exhibit a reduction reactive drug release. The sequence of the reduction responsiveness is described below as STeS->-SSeS->SSS-. DTX-SCS-DTX NPs release minimum docetaxel. Since oxidation and reduction are opposite reactions, DTX-STeS-DTX NPs surprisingly exhibit ultra-high redox double responsiveness.
Example 8: cytotoxicity of PEG-modified chalcogen hybrid bridge Lian Duo Caesalpoly drug self-assembled nanoparticles
The self-assembled nanoparticle of PEG-modified chalcogenides bridged dimer was examined for three tumor cells and one normal cell by MTT method: toxicity of mouse breast cancer (4T 1) cells, mouse melanoma (B16F 10) cells, mouse liver cancer (Hepa 1-6) cells, and mouse fibroblasts (3T 3) cells. Firstly, cells with good forms are digested, the cells are diluted to 5000cells/mL by culture solution and are uniformly blown, 100 mu L of cell suspension is added into each well of a 96-well plate, and the cells are placed in an incubator for incubation for 24 hours to enable the cells to adhere to the wall. After cell attachment, taxotere or the chalcogen hybrid bridge Lian Duo sitaxel dimer prodrug nanoparticles prepared in example 5 are added. The preparation and dilution of the drug solution and nanoparticle preparation in this experiment were carried out using the culture solution of the corresponding cells and sterile filtration with 0.22 μm filter. 100 μl of each well of test solution was added, 3 wells in parallel per concentration. The control group, i.e. without adding the liquid medicine to be detected, is singly supplemented with 100 mu L of culture solution, and is placed in an incubator for incubation with cells. After 48h post-dosing, the 96-well plates were removed, 20 μl of 5mg/mL MTT solution was added to each well, incubated in an incubator for 4h, the medium was discarded, the 96-well plates were back-buckled on filter paper to sufficiently blot the residual liquid, and 200 μl of DMSO was added to each well and shaken on a shaker for 10min to dissolve the bluish violet crystals. A1 wells (containing only 200. Mu.L DMSO) were set as zeroed wells. Absorbance values after zeroing of each well were determined at 570nm using a microplate reader.
As shown in fig. 9, the four dimeric prodrug nanocomposites were all less cytotoxic than docetaxel solutions because of the process that the prodrug needs to undergo to activate to function in the cell. Cytotoxicity of the dimeric prodrug nanocomposites is closely related to their redox-activating ability. The anti-tumor activity sequence of the four dimer prodrug nano-assemblies is as follows: DTX-STS-DTX NPs > DTX-SSS-DTX NPs > DTX-SCS-DTX NPs. DTX-STeS-DTX NPs exhibit the strongest in vitro antitumor activity due to their dual hypersensitivity to redox and their ability to effectively cope with heterogeneous microenvironment of tumor cells.
Example 9: pharmacokinetic study of PEG-modified chalcogen hybrid bridge Lian Duo Caesalper dimer prodrug self-assembled nanoparticles
SD rats weighing 200-250g were randomly grouped and fasted for 12h before dosing, and were given free water. The PEGylated chalcogenides bridge Lian Duo sitaxetil dimer prodrug self-assembled nanoparticles prepared in example 5 were injected intravenously separately. The dosage of docetaxel was 4mg/kg. The orbit was bled at the prescribed time points and plasma was isolated. The drug concentration in the plasma was determined by liquid chromatography-mass spectrometry.
The experimental results are shown in figure 10, in which docetaxel in taxotere is rapidly cleared from the blood due to the short half-life. In contrast, the cycle time of docetaxel dimer prodrug self-assembled nanoparticles is significantly prolonged. Meanwhile, different chemical linkages have a significant effect on the pharmacokinetic behavior of the dimeric prodrug nanoparticles. DTX-SCS-DTX NPs have higher area under the curve and longer cycle time due to weaker chemosensitivity. The self-assembled nano-particles of the dimer prodrug with the hybrid bond and the trisulfide bond have strong colloid stability, and the in-vivo retention of the self-assembled nano-particles is prolonged. DTX-STeS-DTX NPs and DTX-SSeS-DTX NPs release more docetaxel than other nanoparticles, probably because of the higher redox responsiveness of chalcogen hybrid bonds.
Example 10: in-vivo anti-tumor experiment of PEG modified chalcogen hybrid bridge Lian Duo cetime dimer prodrug self-assembled nanoparticle
A4T 1 tumor-bearing BALB/c mouse is taken as a model, PEG modified chalcogenides bridge Lian Duo cetime dimer prodrug self-assembled nanoparticle is given to tail vein, and a taxotere and normal saline intravenous injection group is set as a control group. As a result, as shown in FIG. 11, the DTX-SCS-DTX NPs, DTX-SSS-DTX NPs and DTX-SSeS-DTX NPs groups slowed down the tumor growth to some extent compared to the physiological saline group. In contrast, taxotere and DTX-STeS-DTX NPs showed higher antitumor effect. This is due to the good colloidal stability of DTX-STeS-PTX NPs, improved pharmacokinetic behavior and higher area under the curve. Meanwhile, DTX-STeS-PTX NPs have a relatively fast drug release rate in tumor cells, so that cytotoxicity of the DTX-STeS-PTX NPs is improved. However, the taxotere group showed a significant weight loss, whereas none of the dimeric prodrug nanoparticle groups showed a significant weight loss, indicating that DTX-STeS-DTX NPs have better safety while having a comparable tumor-inhibiting effect with taxotere. In conclusion, stability, cytotoxicity, pharmacokinetic profile and drug release capacity of tumor sites of the nanoparticles affect the final anti-tumor effect, and the above results again demonstrate the advantages of the self-assembled nano-particles of the chalcogen hybrid bridge Lian Duo sitaxe dimer prodrug.

Claims (4)

1. The self-assembled nanoparticle of the chalcogenide hybrid bridged dimer prodrug is characterized in that the chalcogenide hybrid bridged dimer prodrug is a redox double-sensitive type sulfur tellurium sulfur hybrid bridged docetaxel dimer prodrug or a sulfur selenium sulfur hybrid bridged docetaxel dimer prodrug, and the structural formulas are as follows:
the preparation method of the chalcogen hybrid bond bridging dimer prodrug comprises the following steps:
respectively dissolving 3,3 '-tellurium-dithiodipropionic acid and 3,3' -selenium-dithiodipropionic acid in dichloromethane, and stirring uniformlyThe method comprises the steps of carrying out a first treatment on the surface of the Dissolving DMAP, EDCI and docetaxel in anhydrous dichloromethane, stirring, and mixing with the above 3,3 '-tellurium dithiodipropionic acid and 3,3' -selenium dithiodipropionic acid solution, respectively, N 2 Stirring at room temperature under the protection; adding EDCI and DMAP, N into the mixed solution 2 Under the protection, stirring is continued at room temperature, and the obtained product is separated and purified through a preparation liquid phase;
the self-assembled nanoparticle of the chalcogen hybrid bond bridged dimer prodrug is a PEG modified dimer self-assembled prodrug nanoparticle; the preparation method comprises the following steps: dissolving a chalcogenides bridged dimer prodrug and a PEG modifier into a solvent, slowly dripping the solution into water under stirring, spontaneously forming uniform nanoparticles by the prodrug, and removing the solvent by a reduced pressure distillation method to obtain a nano colloid solution without an organic solvent;
the PEG modifier is DSPE-PEG, and the molecular weight of the PEG is 2000; the solvent is selected from ethanol, dimethyl sulfoxide, N' -dimethylformamide, tetrahydrofuran and acetone; the weight ratio of the chalcogen hybrid bond bridging dimer prodrug to the PEG modifier is 90:10-70:30.
2. Use of self-assembled nanoparticles of a chalcogenide hybrid bond bridged dimer prodrug of claim 1 in the preparation of a drug delivery system.
3. Use of self-assembled nanoparticles of a chalcogenized bridged dimer prodrug of claim 1 in the preparation of an antitumor drug.
4. Use of self-assembled nanoparticles of a chalcogenide hybrid bridged dimer prodrug of claim 1 for the preparation of an injectable, oral or topical delivery system.
CN202210776933.4A 2022-07-04 2022-07-04 Chalcogen hybrid bond bridged dimer prodrug, self-assembled nanoparticle thereof, preparation method and application Active CN115300637B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210776933.4A CN115300637B (en) 2022-07-04 2022-07-04 Chalcogen hybrid bond bridged dimer prodrug, self-assembled nanoparticle thereof, preparation method and application

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210776933.4A CN115300637B (en) 2022-07-04 2022-07-04 Chalcogen hybrid bond bridged dimer prodrug, self-assembled nanoparticle thereof, preparation method and application

Publications (2)

Publication Number Publication Date
CN115300637A CN115300637A (en) 2022-11-08
CN115300637B true CN115300637B (en) 2024-03-29

Family

ID=83856963

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210776933.4A Active CN115300637B (en) 2022-07-04 2022-07-04 Chalcogen hybrid bond bridged dimer prodrug, self-assembled nanoparticle thereof, preparation method and application

Country Status (1)

Country Link
CN (1) CN115300637B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114163458A (en) * 2021-10-31 2022-03-11 南京碳硅人工智能生物医药技术研究院有限公司 Design synthesis and activity evaluation of ROS (reactive oxygen species) -responsive dimeric camptothecin prodrug

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108478803A (en) * 2018-04-08 2018-09-04 沈阳药科大学 The structure of redox hypersensitization disulfide bond bridging prodrug self-assembled nanometer grain
CN109350748A (en) * 2018-10-24 2019-02-19 沈阳药科大学 Redox sensitive key bridging small molecule prodrugs and its self-assembled nanometer grain
CN111484501A (en) * 2020-04-22 2020-08-04 沈阳药科大学 Hydroxycamptothecin linoleate micromolecule prodrug and construction of self-assembled nanoparticles thereof
CN111494640A (en) * 2020-05-11 2020-08-07 沈阳药科大学 Redox double-sensitive trithio bond bridged dimer prodrug and self-assembled nanoparticles thereof
CN113264906A (en) * 2020-02-15 2021-08-17 沈阳药科大学 Docetaxel dimer micromolecule prodrug and construction of self-assembled nanoparticles thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108478803A (en) * 2018-04-08 2018-09-04 沈阳药科大学 The structure of redox hypersensitization disulfide bond bridging prodrug self-assembled nanometer grain
CN109350748A (en) * 2018-10-24 2019-02-19 沈阳药科大学 Redox sensitive key bridging small molecule prodrugs and its self-assembled nanometer grain
CN113264906A (en) * 2020-02-15 2021-08-17 沈阳药科大学 Docetaxel dimer micromolecule prodrug and construction of self-assembled nanoparticles thereof
CN111484501A (en) * 2020-04-22 2020-08-04 沈阳药科大学 Hydroxycamptothecin linoleate micromolecule prodrug and construction of self-assembled nanoparticles thereof
CN111494640A (en) * 2020-05-11 2020-08-07 沈阳药科大学 Redox double-sensitive trithio bond bridged dimer prodrug and self-assembled nanoparticles thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Trisulfide bond–mediated doxorubicin dimeric prodrug nanoassemblies with high drug loading, high self-assembly stability, and high tumor selectivity;Yinxian Yang等;《SCIENCE ADVANCES》;第6卷;第1-13页 *
肿瘤氧化还原微环境智能响应型前药纳米组装体的研究进展;陈瑶等;《药学进展》;第45卷(第5期);第337-348页,尤其是第344页右栏第1段 *

Also Published As

Publication number Publication date
CN115300637A (en) 2022-11-08

Similar Documents

Publication Publication Date Title
CN111494640B (en) Redox double-sensitive trithio bond bridged dimer prodrug and self-assembled nanoparticles thereof
CN109350748B (en) Redox double-sensitive bond bridged micromolecule prodrug and self-assembled nanoparticle thereof
CN113264906B (en) Docetaxel dimer micromolecular prodrug and construction of self-assembled nanoparticle thereof
CN106046029B (en) A kind of reproducibility response amphipathic small molecules prodrug and preparation method thereof
CN112604002A (en) Disulfide-bond bridged docetaxel-fatty acid prodrug and self-assembled nanoparticles thereof
CN112089845B (en) Taxane drug-adriamycin prodrug self-assembly nanoparticles and application thereof
CN111484501A (en) Hydroxycamptothecin linoleate micromolecule prodrug and construction of self-assembled nanoparticles thereof
CN113398277A (en) Fatty acid/fatty alcohol-antitumor substance prodrug and preparation method of self-assembled nanoparticles thereof
CN111116521B (en) Solanesol modified taxol prodrug and preparation method and application thereof
CN115300637B (en) Chalcogen hybrid bond bridged dimer prodrug, self-assembled nanoparticle thereof, preparation method and application
CN111298132B (en) Tree-shaped molecule gemcitabine self-assembled nano prodrug and preparation method and application thereof
CN112494458B (en) Construction of triglyceride-like prodrug intravenous injection self-assembly nanoparticles
CN114796513B (en) Diselenide bridge Lian Duo cetime dimer prodrug and self-assembled nanoparticles thereof
CN109675048B (en) Anticancer prodrug liposome and artemisinin liposome nano-drug
CN115611926B (en) Non-sensitive bond bridged SN38 dimer prodrug, self-assembled nanoparticle thereof and application thereof
CN116120333B (en) Podophyllotoxin nano prodrug and preparation method and application thereof
CN110227164B (en) Hydrophobic anti-tumor drug containing ketocarbonyl and conjugate thereof, nano preparation containing conjugate, and preparation method and application thereof
CN115844891A (en) Disulfide bond bridged SN38 dimer prodrug, self-assembled nanoparticles thereof, preparation method and application
CN117700378A (en) Four-sulfur bond bridged dimer prodrug, self-assembled nanoparticle thereof and application thereof
CN114931650B (en) Quantum dot nano-drug carrier based on dihydrolipoic acid modification and preparation method and application thereof
WO2022227556A1 (en) Docetaxel-fatty alcohol small molecule prodrug and construction of self-assembled nanoparticles thereof
CN113082221B (en) Preparation method of Tyr/Ang dual-modified artesunate nano-targeting preparation and application of preparation in craniocerebral tumor treatment
WO2022227554A1 (en) Larotaxel-fatty alcohol small molecule prodrug and construction of self-assembling nanoparticle thereof
CN116327968A (en) Docetaxel prodrug antitumor preparation
CN117964611A (en) Paclitaxel prodrug, nanometer preparation, and preparation methods and applications 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
GR01 Patent grant
GR01 Patent grant