CN112546025A - Preparation method of Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system - Google Patents

Preparation method of Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system Download PDF

Info

Publication number
CN112546025A
CN112546025A CN202011448169.5A CN202011448169A CN112546025A CN 112546025 A CN112546025 A CN 112546025A CN 202011448169 A CN202011448169 A CN 202011448169A CN 112546025 A CN112546025 A CN 112546025A
Authority
CN
China
Prior art keywords
dsp
ipi549
cmcs
delivery system
tumor
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
CN202011448169.5A
Other languages
Chinese (zh)
Other versions
CN112546025B (en
Inventor
张雪琼
杨梦佳
邱彤
詹斯文
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.)
Wuhan University of Technology WUT
Original Assignee
Wuhan University of Technology WUT
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 Wuhan University of Technology WUT filed Critical Wuhan University of Technology WUT
Priority to CN202011448169.5A priority Critical patent/CN112546025B/en
Publication of CN112546025A publication Critical patent/CN112546025A/en
Application granted granted Critical
Publication of CN112546025B publication Critical patent/CN112546025B/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
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0057Photodynamic therapy with a photosensitizer, i.e. agent able to produce reactive oxygen species upon exposure to light or radiation, e.g. UV or visible light; photocleavage of nucleic acids with an agent
    • A61K41/0071PDT with porphyrins having exactly 20 ring atoms, i.e. based on the non-expanded tetrapyrrolic ring system, e.g. bacteriochlorin, chlorin-e6, or phthalocyanines
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5161Polysaccharides, e.g. alginate, chitosan, cellulose derivatives; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y20/00Nanooptics, e.g. quantum optics or photonic crystals
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y40/00Manufacture or treatment of nanostructures
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery

Abstract

The invention discloses a Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system and a preparation method thereof, which can adjust action targets, control time, concentrate at tumor cells, effectively reduce the toxic and side effects of a photosensitizer, improve solubility and reduce adverse reactions of organisms. Dissolving an anti-tumor drug IPI-549 bonded with immunotherapy and an amphiphilic prodrug CMCS-DSP-IPI549 of a disulfide bond group in deionized water, adding an organic solution containing a hydrophobic photosensitizer chlorin (Ce6) by a dropping method, and carrying out ultrasonic disruption and dialysis self-assembly to obtain the Ce6@ CMCS-DSP-IPI549 anti-tumor nano delivery system. The nano delivery system synthesizes and encapsulates two medicines with different mechanisms of action in a nanoparticle, treats tumors by combining an immunotherapy and a phototherapy double channel, can release the two medicines in a tumor microenvironment and specific near-infrared illumination, and provides a new methodology and theoretical basis for clinical tumor treatment.

Description

Preparation method of Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system
Technical Field
The invention relates to the technical field of medicines, in particular to a preparation method of a Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system.
Background
Tumors are highly specific diseases in which each tumor is composed of cancer cells with different genetic backgrounds, and thus each tumor behaves differently in response to drugs. Once the cancer cells are shed from the organ tissues in vivo, the free cancer cells can be disseminated throughout the body along with blood or lymph fluid to form metastasis, which endangers life.
The traditional mode for treating tumors is chemotherapy, namely, an anti-tumor medicament is orally taken or injected intravenously, and is transmitted to a focus part through systemic circulation to kill tumor cells. However, most of the antitumor drugs have no selectivity to tumor cell tissues and normal cell tissues, and can cause unavoidable damage to normal cells, great damage to human bodies and great pain to patients. Secondly, because of the existence of biological barriers (such as blood brain barrier) in human bodies, only a small amount of drugs can reach a focus area, so that the bioavailability is low; in addition, long-term administration can cause drug resistance in the body, which makes tumor treatment more difficult. In order to solve the above problems, drug delivery systems using new formulations or new technologies are being developed vigorously.
Disclosure of Invention
Based on the defects of the prior art, the technical problem to be solved by the invention is to provide the Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system and the preparation method thereof. The core-shell nanoparticle prepared by the method has good stability, tumor targeting property, immunotherapy property and near-infrared light responsiveness, can improve immunosuppressive cells in a tumor microenvironment, and can regulate the action target and action time of near-infrared light to regulate and control the release of a photosensitizer.
In order to solve the technical problems, the invention provides a preparation method of a Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system, which is characterized by comprising the following steps:
(1) and (3) synthesis of a DSP-IPI549 intermediate:
dissolving a certain amount of DSP in DMSO, and stirring until the DSP is completely dissolved; dissolving IPI-549 in DMSO, slowly dripping into the DSP solution, reacting for 24h under magnetic stirring at room temperature in dark place, dialyzing with mixed solution of DMSO and deionized water and water, and freeze-drying to obtain lyophilized powder of DSP-IPI549 intermediate;
(2) synthesis of CMCS-DSP-IPI549 prodrug:
dissolving the DSP-IPI549 freeze-dried powder in DMSO to obtain a DSP-IPI549 solution; dissolving CMCS in deionized water, adjusting the pH value to 7.4, and performing ultrasonic treatment for 5min to obtain a CMCS solution; slowly dripping the DSP-IPI549 solution into the CMCS solution, reacting for 24 hours under the condition of magnetic stirring in the dark at room temperature, dialyzing with a mixed solution of DMSO and deionized water with a ratio of 7:3-3:7 and the deionized water, and freeze-drying to obtain CMCS-DSP-IPI549 prodrug freeze-dried powder;
(3) preparation of Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system
Dissolving Ce6 in DMSO to prepare a solution A, dissolving a CMCS-DSP-IPI549 prodrug in deionized water to prepare a solution B, dropwise adding the solution A into the solution B on a magnetic stirrer, stirring for 4 hours at room temperature in a dark place, performing low-temperature ultrasonic crushing to obtain a Ce6@ CMCS-DSP-IPI549 nanoparticle solution, dialyzing for 4 hours in deionized water, and freeze-drying to obtain a Ce6@ CMCS-DSP-IPI549 nanoparticle freeze-dried powder, namely the Ce6@ CMCS-DSP-IPI549 anti-tumor nano transfer system.
Preferably, the preparation method of the Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system provided by the invention further comprises part or all of the following technical characteristics:
as an improvement of the technical scheme, in the step (1), the molar ratio of the DSP to the IPI549 is 1: 1; the mixed solution of DMSO and deionized water is a mixed solution of DMSO and water in a volume ratio of 1: 9.
As an improvement of the above technical means, in the step (2), the molar mass of CMCS is 1X 104And the degree of substitution by carboxymethyl groups is 85%; the mass ratio of the CMCS to the DSP-IPI549 in the step (2) is 1: 1.3-3.3; the drug loading rate of the CMCS-DSP-IPI549 prodrug is 10.17-13.63%. Wherein the drug loading rate is determined by ultraviolet photometry
As an improvement of the technical scheme, in the step (3), the mass ratio of the CMCS-DSP-IPI549 prodrug to the photosensitizer Ce6 is 5:1-15: 1.
As an improvement of the technical scheme, in the step (1), the molecular weight cut-off of the dialysis bag is 500Da, and in the steps (2) to (3), the molecular weight cut-off of the dialysis bag is 3500 Da.
As an improvement of the technical scheme, in the step (3), the low-temperature ultrasonic crushing is performed by using an ultrasonic cell crusher, the probe performs ultrasonic operation at low temperature for 3.0s, the interval is 2.0s, and the pulse mode with the power of 90w is operated for 10 min.
As an improvement of the technical scheme, the particle size range of the Ce6@ CMCS-DSP-IPI549 nano-particles obtained in the step (3) is 141-396nm, the drug loading rate is 6.49-10.92%, and the encapsulation rate is 55.68-72.65%. Wherein the nanoparticles are measured by a dynamic light scattering instrument (DLS), and the drug loading rate and the encapsulation rate are measured by an ultraviolet photometry.
The Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system is prepared by any method.
An application of the Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system, which is characterized in that: two drugs with different mechanisms of action are synthesized and encapsulated in the same nanoparticle, and the immunotherapy and the phototherapy are combined to treat the tumor, so that the two drugs are released in the tumor microenvironment and specific near-infrared illumination, the solubility of the drugs is improved, the toxic and side effects are reduced, the targeting property is increased, and a new methodology and theoretical basis are provided for clinical tumor treatment
Preferably, the application of the Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system provided by the invention further comprises part or all of the following technical characteristics:
as an improvement of the technical scheme, the wavelength of the near-infrared light is 606 nm.
IPI-549 as an immunological clinical candidate tumor drug is discovered in the optimization process of an isoquinolone PI3K inhibitor, the selectivity of the IPI-549 is more than 100 times that of other lipid and protein kinases, and the IC50 is 16 nM. IPI-549 shows good pharmacokinetic properties and has inhibitory effect on PI 3K-gamma-mediated neutrophil migration, and is currently in phase I clinical trial (subjects are patients with advanced solid tumors). In animal experiments (mouse, rat, dog and monkey), IPI-549 was found to have excellent oral bioavailability, low clearance, short half-life (t 1/2 of mouse, rat, dog and monkey is 3.2, 4.4, 6.7, 4.3 hours, respectively) and easy distribution into each tissue (mean volume of distribution is 1.2L/kg). As a small molecule kinase inhibitor, IPI-549 is an ideal first choice medicament for combined treatment of tumors.
The nano medicine carrying system is prepared through physical embedding or chemical covalent coupling process to connect medicine and natural or synthetic polymer carrier. The system improves bioavailability of the medicine, reduces toxic and side effects of the medicine on organism, can specifically identify focus region, and controls medicine release. The system has targeting property, reduces the damage of the drug to normal cells and tissues, directionally conveys the drug to a tumor area, and reduces the drug toxicity.
Carboxymethyl chitosan (CMCS) is a polymer drug carrier with wide applications, and has been widely used in the field of pharmaceutical biology due to its characteristics of good biosafety and biocompatibility, multiple active groups, and easy biodegradation. The CMCS active group can react with the drug to form a covalent bond and can stably exist in blood circulation, so the CMCS is developed to be used as a carrier in a micromolecular antitumor drug delivery system, and the system can improve the water solubility of the micromolecular drug, prolong the half-life period, reduce the toxic and side effects and improve the targeting property.
Glutathione (GSH) is a sulfhydryl-containing polypeptide protein present in the human body, and its concentration is only micromolar (2-20. mu.M) in body fluid or outside cells, while intracellular GSH concentration generally reaches millimolar (2-10mM), resulting in 4-fold higher concentration than normal cells due to high hypoxia in cancer cells, and disulfide bonds are easily broken in this environment. The disulfide bond-containing 3, 3' -dithiodipropionic acid bis (N-hydroxysuccinimide ester) (DSP) can be applied to an antitumor drug nano-delivery system, can reduce the steric hindrance of an antitumor drug and a carrier, and can target-release the drug in tumor cells or tissues by utilizing the characteristic that the disulfide bond is easy to be cracked by high-concentration GSH.
Phototherapy is an emerging tumor treatment method, which can be used for local treatment at a specific target spot, and is characterized in that a photosensitive drug accumulated in tumor cells or tissues is excited by irradiation of an ultraviolet or near-infrared light source to generate local hyperthermia or Reactive Oxygen Species (ROS), so that the tumor cells are killed. The tissue penetrating power of ultraviolet light is weak and easy to damage organism, while the tissue penetrating power of near infrared light can reach deep tumor and almost has no damage to organism, so that a near infrared light source of 650-950nm is generally used. The light therapy has the advantages of small wound, low discomfort of the patient body, high acceptance, controllable action site and time, low toxic and side effects and the like. Light therapy is divided into photodynamic therapy (PDT) and photothermal therapy (PTT). Chlorin e6(Ce6) is a second generation photosensitizer used in PDT treatment, belonging to the chlorophyll derivatives, which have been approved for use by the FDA. The Ce6 excited has the advantages of strong penetrating power, high ROS yield, fluorescence imaging capability and the like.
The photosensitizer Ce6 is added into the aqueous solution containing the CMCS-DSP-IPI549 prodrug by a dropping method, after the mixture is stirred and mixed uniformly, the Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system is obtained by ultrasonic crushing and dialysis self-assembly, so that the immunosuppressive cells in a tumor microenvironment can be improved, the ROS yield can be increased, the action sites and time can be controlled, and a better antitumor treatment effect can be achieved by combining immunotherapy and light therapy of two different action mechanisms.
Compared with the prior art, the technical scheme of the invention has the following beneficial effects:
1. two drugs with different mechanisms of action are synthesized and encapsulated in the same nanoparticle, and the immunotherapy and photodynamic therapy dual-channel combined therapy is adopted to treat the tumor, so that the two drugs are released in the tumor microenvironment and specific near-infrared illumination, the solubility of the drugs is improved, the toxic and side effects are reduced, the targeting property is increased, and a new methodology and theoretical basis are provided for clinical immunotherapy and phototherapy.
2. The near infrared light has no damage to the organism, and the release of the photosensitizer Ce6 can be controlled by adjusting the action part and action time of the near infrared light through the outside world, and ROS is generated to kill tumor cells or tissues.
3. By utilizing the reduction sensitivity of the disulfide bond, the Ce6@ CMCS-DSP-IPI549 nanoparticle increases the targeting property of the photosensitizer Ce6, improves the bioavailability of the medicament, and reduces the toxic and side effects of the photosensitizer on normal cells or tissues.
The foregoing description is only an overview of the technical solutions of the present invention, and in order to make the technical means of the present invention more clearly understood, the present invention may be implemented in accordance with the contents of the description, and in order to make the above and other objects, features, and advantages of the present invention more clearly understood, the following detailed description is given in conjunction with the preferred embodiments.
Drawings
In order to more clearly illustrate the technical solutions of the embodiments of the present invention, the drawings of the embodiments will be briefly described below.
FIG. 1 is a transmission electron microscope image of Ce6@ CMCS-DSP-IPI549 nanoparticles prepared in example 1 of the present invention;
FIG. 2 is a release curve of Ce6@ CMCS-DSP-IPI549 nanoparticles prepared in example 1 of the present invention at different concentrations of GSH, wherein IPI-549(A), Ce6 (B);
FIG. 3 shows the absorbance at 426nm of the Ce6@ CMCS-DSP-IPI549 nanoparticle prepared in example 1 of the present invention and a singlet oxygen probe (DPBF).
Detailed Description
Other aspects, features and advantages of the present invention will become apparent from the following detailed description, which, when taken in conjunction with the drawings, illustrate by way of example the principles of the invention.
Example 1:
(1) and (3) synthesis of a DSP-IPI549 intermediate:
DSP 20mg was precisely weighed and dissolved in 4.2mL of DMSO, and stirred until completely dissolved. Accurately weighing IPI-54927 mg, dissolving in 5.6mL DMSO, slowly dropping into the DSP solution, magnetically stirring at room temperature in the dark for 24h, transferring into dialysis bag (molecular weight cutoff is 500Da), dialyzing with mixed solution of DMSO and deionized water at ratio of 1:9 for 48h, and dialyzing with deionized water for 24 h. And freeze-drying to obtain the DSP-IPI549 intermediate freeze-dried powder.
(2) Synthesis of CMCS-DSP-IPI549 prodrug:
precisely weighing 20mg of DSP-IPI549 freeze-dried powder, dissolving in 7mL of DMSO, precisely weighing 6mg of CMCS, dissolving in 70mL of deionized water, adjusting the pH value to 7.4, and performing ultrasonic treatment for 5min to obtain a CMCS solution. Slowly dripping the DSP-IPI549 solution into the CMCS solution, magnetically stirring the solution for reaction for 24 hours at room temperature in a dark place, transferring the solution into a dialysis bag (the molecular weight cutoff is 3500Da), dialyzing the solution for 12 hours by a mixed solution of DMSO with the weight ratio of 7:3/5:5/3:7 and deionized water, and dialyzing the solution for 24 hours by the deionized water. After freeze-drying, a lyophilized powder was obtained, which was dissolved in 40mL of DMSO, sonicated for 10min, and filtered again to remove the DMSO-dissolved reaction, and the procedure was repeated 3 times. Dissolving the filter cake in deionized water, dialyzing with pure water in a dialysis bag for 24h, and freeze-drying to obtain CMCS-DSP-IPI549 prodrug freeze-dried powder.
(3) Preparation of Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system
Weighing 1mg of Ce6, dissolving in 1mL of DMSO to prepare a 1mg/mL solution, dissolving 10mg of CMCS-DSP-IPI549 in 1mL of deionized water to prepare a 10mg/mL solution, dropwise adding the Ce6 solution into the aqueous solution on a magnetic stirrer, stirring for 4 hours at room temperature in a dark place, transferring the solution to an ultrasonic cell crusher, performing ultrasonic treatment at low temperature for 3.0 seconds, performing intermittent operation for 2.0 seconds and performing pulse operation at power of 90w for 10 minutes to obtain a Ce6@ CMCS-DSP-IPI549 nanoparticle solution, transferring the solution to a dialysis bag (molecular weight cutoff is 3500Da), dialyzing in deionized water for 4 hours, and performing freeze drying to obtain Ce6 CMCS-DSP-IPI549 nanoparticle freeze-dried powder.
The Ce6@ CMCS-DSP-IPI549 nanoparticle prepared in the embodiment has the drug loading rate of 8.05% and the encapsulation rate of 64.42%.
Example 2:
this example differs from example 1 in that: in this example (3), 2mg of Ce6 was weighed out and dissolved in 1mL of DMSO to prepare a 2mg/mL solution, and the other preparation raw materials and the preparation process of a Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system were the same as in example 1.
The drug loading rate of the Ce6@ CMCS-DSP-IPI549 nano-particles prepared in the embodiment is 10.92%, and the encapsulation rate is 55.68%.
Example 3:
this example differs from example 1 in that: in this example (3), 15mg of CMCS-DSP-IPI549 was weighed in 1mL of deionized water to prepare a solution of 15mg/mL, and the preparation process of the other raw materials and an anti-tumor nano-delivery system of Ce6@ CMCS-DSP-IPI549 was the same as in example 1.
The Ce6@ CMCS-DSP-IPI549 nanoparticle prepared in the embodiment has the drug loading rate of 6.49% and the encapsulation rate of 72.65%.
Characterization experiment of nanoparticles:
in order to prove that the Ce6@ CMCS-DSP-IPI549 prepared in the example 1 can be self-assembled in an aqueous solution to form core-shell nanoparticles, a transmission electron microscope is adopted for characterization (see figure 1), and the result shows that the Ce6@ CMCS-DSP-IPI549 nanoparticles are spherical, regular in shape and uniform in distribution; the average particle size of the nanoparticles is characterized by 218.8 +/-4.59 nm by using a dynamic light scattering instrument.
To demonstrate that the Ce6@ CMCS-DSP-IPI549 nano-delivery system prepared in example 1 has GSH responsiveness, 4 concentration gradients are established according to the concentration of GSH in normal cells and tumor cells, and release curves of the Ce6@ CMCS-DSP-IPI549 nano-delivery system at 267nm (IPI-549) and 402nm (Ce6) are prepared (see FIG. 2).
After irradiation of near infrared light 606nm, Ce6 can excite generated ROS to chemically react with a singlet oxygen probe (DPBF), so that the absorbance of the generated ROS at 426nm is reduced, and the yield of the ROS is reflected. To demonstrate that the Ce6@ CMCS-DSP-IPI549 nano-delivery system prepared in example 1 generates ROS under irradiation of near infrared light, the absorbance at 426nm was measured after irradiation of 606nm of near infrared light for various periods of time (see FIG. 3).
The invention discloses a Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system and a preparation method thereof, which can adjust action targets, control time, concentrate at tumor cells, effectively reduce the toxic and side effects of photosensitizers, improve solubility, reduce adverse reactions of organisms and provide new ideas and theoretical bases for the development of clinical antitumor drugs. Dissolving an anti-tumor drug IPI-549 bonded with immunotherapy and an amphiphilic prodrug CMCS-DSP-IPI549 of a disulfide bond group in deionized water, adding an organic solution containing a hydrophobic photosensitizer chlorin (Ce6) by a dropping method, and carrying out ultrasonic disruption and dialysis self-assembly to obtain the Ce6@ CMCS-DSP-IPI549 anti-tumor nano delivery system. The nano delivery system synthesizes and encapsulates two medicines with different mechanisms of action in a nanoparticle, treats tumors by combining an immunotherapy and a phototherapy double channel, can release the two medicines in a tumor microenvironment and specific near-infrared illumination, and provides a new methodology and theoretical basis for clinical tumor treatment.
The raw materials listed in the invention, the upper and lower limits and interval values of the raw materials of the invention, and the upper and lower limits and interval values of the process parameters (such as temperature, time and the like) can all realize the invention, and the examples are not listed.
While the foregoing is directed to the preferred embodiment of the present invention, other and further embodiments of the invention may be devised without departing from the basic scope thereof, and the scope thereof is determined by the claims that follow.

Claims (10)

1. A preparation method of a Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system is characterized by comprising the following steps:
(1) and (3) synthesis of a DSP-IPI549 intermediate:
dissolving a certain amount of DSP in DMSO, and stirring until the DSP is completely dissolved; dissolving IPI-549 in DMSO, slowly dripping into the DSP solution, reacting for 24h under magnetic stirring at room temperature in dark place, dialyzing with mixed solution of DMSO and deionized water and water, and freeze-drying to obtain lyophilized powder of DSP-IPI549 intermediate;
(2) synthesis of CMCS-DSP-IPI549 prodrug:
dissolving the DSP-IPI549 freeze-dried powder in DMSO to obtain a DSP-IPI549 solution; dissolving CMCS in deionized water, adjusting the pH value to 7.4, and performing ultrasonic treatment for 5min to obtain a CMCS solution; slowly dripping the DSP-IPI549 solution into the CMCS solution, reacting for 24 hours under the condition of magnetic stirring in the dark at room temperature, dialyzing with a mixed solution of DMSO and deionized water with a ratio of 7:3-3:7 and the deionized water, and freeze-drying to obtain CMCS-DSP-IPI549 prodrug freeze-dried powder;
(3) preparation of Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system
Dissolving Ce6 in DMSO to prepare a solution A, dissolving a CMCS-DSP-IPI549 prodrug in deionized water to prepare a solution B, dropwise adding the solution A into the solution B on a magnetic stirrer, stirring for 4 hours at room temperature in a dark place, performing low-temperature ultrasonic crushing to obtain a Ce6@ CMCS-DSP-IPI549 nanoparticle solution, dialyzing for 4 hours in deionized water, and freeze-drying to obtain a Ce6@ CMCS-DSP-IPI549 nanoparticle freeze-dried powder, namely the Ce6@ CMCS-DSP-IPI549 anti-tumor nano transfer system.
2. The method for preparing the Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system according to claim 1, wherein the method comprises the following steps: in the step (1), the molar ratio of the DSP to the IPI549 is 1: 1; the mixed solution of DMSO and deionized water is a mixed solution of DMSO and water in a volume ratio of 1: 9.
3. The method for preparing the Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system according to claim 1, wherein the method comprises the following steps: in the step (2), the molar mass of CMCS is 1X 104And the degree of substitution by carboxymethyl groups is 85%; the mass ratio of the CMCS to the DSP-IPI549 in the step (2) is 1: 1.3-3.3; the drug loading rate of the CMCS-DSP-IPI549 prodrug is 10.17-13.63%.
4. The method for preparing the Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system according to claim 1, wherein the method comprises the following steps: in the step (3), the mass ratio of the CMCS-DSP-IPI549 prodrug to the photosensitizer Ce6 is 5:1-15: 1.
5. The method for preparing the Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system according to claim 1, wherein the method comprises the following steps: in the step (1), the cut-off molecular weight of the dialysis bag is 500Da, and in the steps (2) to (3), the cut-off molecular weight of the dialysis bag is 3500 Da.
6. The method for preparing the Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system according to claim 1, wherein the method comprises the following steps: in the step (3), the low-temperature ultrasonic crushing is realized by using an ultrasonic cell crusher, and the probe operates for 10min in a pulse mode with the ultrasonic frequency of 3.0s at low temperature, the interval of 2.0s and the power of 90 w.
7. The method for preparing the Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system according to claim 1, wherein the method comprises the following steps: the particle size range of the Ce6@ CMCS-DSP-IPI549 nano-particles obtained in the step (3) is 141-396nm, the drug loading rate is 6.49-10.92%, and the encapsulation rate is 55.68-72.65%.
8. A Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system is characterized in that: the Ce6@ CMCS-DSP-IPI549 antitumor nano-delivery system is prepared by any method of claims 1-7.
9. Use of the Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system according to any one of claims 1 to 8, wherein: two drugs with different mechanisms of action are synthesized and encapsulated in the same nanoparticle, and the immunotherapy and the phototherapy are combined to treat the tumor, so that the two drugs are released in the tumor microenvironment and specific near-infrared illumination, the solubility of the drugs is improved, the toxic and side effects are reduced, the targeting property is increased, and a new methodology and theoretical basis are provided for clinical tumor treatment.
10. The use of the Ce6@ CMCS-DSP-IPI549 anti-tumor nano-delivery system of claim 9, wherein: the wavelength of the near infrared light is 606 nm.
CN202011448169.5A 2020-12-09 2020-12-09 Preparation method of Ce6@CMCS-DSP-IPI549 anti-tumor nano-delivery system Active CN112546025B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011448169.5A CN112546025B (en) 2020-12-09 2020-12-09 Preparation method of Ce6@CMCS-DSP-IPI549 anti-tumor nano-delivery system

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011448169.5A CN112546025B (en) 2020-12-09 2020-12-09 Preparation method of Ce6@CMCS-DSP-IPI549 anti-tumor nano-delivery system

Publications (2)

Publication Number Publication Date
CN112546025A true CN112546025A (en) 2021-03-26
CN112546025B CN112546025B (en) 2023-06-06

Family

ID=75061014

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011448169.5A Active CN112546025B (en) 2020-12-09 2020-12-09 Preparation method of Ce6@CMCS-DSP-IPI549 anti-tumor nano-delivery system

Country Status (1)

Country Link
CN (1) CN112546025B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114259476A (en) * 2021-12-29 2022-04-01 中南大学湘雅三医院 Nanometer preparation for regulating and controlling macrophage, and preparation method and application thereof
US11939333B2 (en) 2015-09-14 2024-03-26 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105708848A (en) * 2016-02-04 2016-06-29 武汉艾美博特医疗用品有限公司 Environmentally responsive tumor targeted combined administration transfer system
CN105749280A (en) * 2016-04-07 2016-07-13 沈阳大学 Preparation method and application of tumor-targeted nanometer drug delivery system for cooperative chemotherapy and photodynamic therapy
WO2017045192A1 (en) * 2015-09-18 2017-03-23 天津医科大学 Tumor-targeting nanomicelle capable of loading drug by using acousto-optical power, preparation method therefor, and applications thereof
CN110179982A (en) * 2019-06-20 2019-08-30 武汉理工大学 Multifunctional targeted drug delivery system with combined effect of chemotherapy and phototherapy and preparation method thereof
CN110859825A (en) * 2019-11-26 2020-03-06 武汉理工大学 Preparation method of targeted drug delivery nano-delivery system

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017045192A1 (en) * 2015-09-18 2017-03-23 天津医科大学 Tumor-targeting nanomicelle capable of loading drug by using acousto-optical power, preparation method therefor, and applications thereof
CN105708848A (en) * 2016-02-04 2016-06-29 武汉艾美博特医疗用品有限公司 Environmentally responsive tumor targeted combined administration transfer system
CN105749280A (en) * 2016-04-07 2016-07-13 沈阳大学 Preparation method and application of tumor-targeted nanometer drug delivery system for cooperative chemotherapy and photodynamic therapy
CN110179982A (en) * 2019-06-20 2019-08-30 武汉理工大学 Multifunctional targeted drug delivery system with combined effect of chemotherapy and phototherapy and preparation method thereof
CN110859825A (en) * 2019-11-26 2020-03-06 武汉理工大学 Preparation method of targeted drug delivery nano-delivery system

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CATHERINE A. EVANS等: "Discovery of a Selective Phosphoinositide-3-Kinase (PI3K)-γ Inhibitor (IPI-549) as an Immuno-Oncology Clinical Candidate", 《MEDICINAL CHEMISTRY LETTERS》 *
CATHERINE A. EVANS等: "Discovery of a Selective Phosphoinositide-3-Kinase (PI3K)-γ Inhibitor (IPI-549) as an Immuno-Oncology Clinical Candidate", 《MEDICINAL CHEMISTRY LETTERS》, vol. 7, no. 9, 22 July 2016 (2016-07-22) *
ZHANG XUEQIONG等: "Glutathione-dependent micelles based", 《JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION》 *
ZHANG XUEQIONG等: "Glutathione-dependent micelles based", 《JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION》, vol. 27, no. 18, 5 October 2016 (2016-10-05), pages 6 *
许斌等: "恶性肿瘤免疫检查点抑制剂耐药机制及临床应用", 中国肿瘤, vol. 26, no. 9, pages 707 - 713 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11939333B2 (en) 2015-09-14 2024-03-26 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
CN114259476A (en) * 2021-12-29 2022-04-01 中南大学湘雅三医院 Nanometer preparation for regulating and controlling macrophage, and preparation method and application thereof

Also Published As

Publication number Publication date
CN112546025B (en) 2023-06-06

Similar Documents

Publication Publication Date Title
Wang et al. Synthesis and modification of ZIF-8 and its application in drug delivery and tumor therapy
CN108452303A (en) It is a kind of to carry double medicine nanometer formulations and preparation method thereof
CN106139144A (en) A kind of hyaluronic acid decorated golden Nano carbon balls with synergistic antitumor characteristic and preparation method and application
EP1362598A1 (en) Active oxygen generator containing photosensitizer for ultrasonic therapy
CN111617246B (en) Self-assembled nanoparticles of pure photosensitizer and preparation and application thereof
Cao et al. Recent advances in phase change material based nanoplatforms for cancer therapy
CN112546025B (en) Preparation method of Ce6@CMCS-DSP-IPI549 anti-tumor nano-delivery system
CN113559064B (en) Novel self-oxygen-supply liposome nanoparticle and preparation method and application thereof
CN110665003A (en) Double-drug-loading carrier-free nanoparticle and preparation method thereof
CN108354913A (en) A kind of application of nano drug-carrying nanosystems in preparing the drug for treating intractable thyroid cancer
CN111840570A (en) Preparation method of nano-particles of sericin combined with photosensitizer
CN112933229B (en) Carrier-free self-assembly nanoparticle of IR820 and atovaquone and preparation method and application thereof
Yu et al. ATP-triggered drug release system based on ZIF-90 loaded porous poly (lactic-co-glycolic acid) microspheres
Lin et al. A review on reactive oxygen species (ROS)-inducing nanoparticles activated by uni-or multi-modal dynamic treatment for oncotherapy
CN108524470A (en) A kind of Nano medication system and its preparation method and application for treating triple negative breast cancer
CN110251672B (en) Nano diagnosis and treatment agent and preparation method and application thereof
CN109953974B (en) Preparation method of enzyme-reduction dual-responsiveness hyaluronic acid-polypropylene sulfide copolymer nanocapsule
CN106606783B (en) A kind of targeting is passed altogether to be released the drug of photosensitizer and chemotherapeutics and passs release system
WO2022016555A1 (en) Platinum-based drug-/photosensitizer-loaded protein nanoparticle, and preparation method therefor and application thereof
CN114848843B (en) Chemotherapeutic synergistic targeting combined treatment nano-drug and application thereof in tumor treatment
CN113633784B (en) Hybrid nano-assembly for heat shock protein inhibition sensitization photothermal therapy and preparation and application thereof
CN113827553B (en) Zinc phthalocyanine in-situ gel for intratumoral injection administration for photodynamic tumor treatment and preparation method thereof
CN112870356B (en) Series of medicines for photodynamic therapy of tumor and application thereof
CN107812189B (en) Hypocrellin nano preparation for actively targeting specific tumor cells and preparation method and application thereof
CN109806394B (en) Mesoporous silica drug delivery system and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant