CN106667914B - Composition of targeted liposome-cyclic dinucleotide, preparation method and application of targeted liposome-cyclic dinucleotide in resisting tumors - Google Patents

Composition of targeted liposome-cyclic dinucleotide, preparation method and application of targeted liposome-cyclic dinucleotide in resisting tumors Download PDF

Info

Publication number
CN106667914B
CN106667914B CN201710147574.5A CN201710147574A CN106667914B CN 106667914 B CN106667914 B CN 106667914B CN 201710147574 A CN201710147574 A CN 201710147574A CN 106667914 B CN106667914 B CN 106667914B
Authority
CN
China
Prior art keywords
liposome
cgamp
cyclic dinucleotide
targeted
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.)
Active
Application number
CN201710147574.5A
Other languages
Chinese (zh)
Other versions
CN106667914A (en
Inventor
谭瀛轩
向道凤
陆星
谭相石
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.)
Hangzhou star bioscience Co., Ltd.
Original Assignee
Hangzhou Xing'ao Biological Technology Co ltd
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 Hangzhou Xing'ao Biological Technology Co ltd filed Critical Hangzhou Xing'ao Biological Technology Co ltd
Priority to CN201710147574.5A priority Critical patent/CN106667914B/en
Publication of CN106667914A publication Critical patent/CN106667914A/en
Application granted granted Critical
Publication of CN106667914B publication Critical patent/CN106667914B/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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7084Compounds having two nucleosides or nucleotides, e.g. nicotinamide-adenine dinucleotide, flavine-adenine dinucleotide
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0002Galenical forms characterised by the drug release technique; Application systems commanded by energy

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Dispersion Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Molecular Biology (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the technical field of medicines, and particularly relates to a composition and a preparation method of target liposome-encapsulated cyclic dinucleotide cGAMP and application of the cyclic dinucleotide cGAMP in tumor resistance. The targeted liposome is composed of lecithin, cholesterol, polyethylene glycol and the like and linked targeting molecules thereof, and the cGAMP slow-release medicine coated by the targeted liposome can enhance the cell penetration effect, strengthen the immune response, effectively deliver the targeted medicine and enhance and inhibit the growth of various tumor cells. Therefore, the cGAMP encapsulated by the targeting liposome can be used for preparing anti-tumor targeting slow-release medicines and has important potential application in the field of targeting immunity and anti-tumor.

Description

Composition of targeted liposome-cyclic dinucleotide, preparation method and application of targeted liposome-cyclic dinucleotide in resisting tumors
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to a composition of targeted liposome-cyclodiphonucleotide, a preparation method and application thereof in preparing an anti-tumor medicine.
Background
Liposomes (liposomes) are an ultra-microspheroidal carrier formulation formed from lipid bilayers, which are typical representatives of drug delivery nanosomes. When amphiphilic molecules such as phospholipids are dispersed in an aqueous phase, the hydrophobic tails of the molecules cluster together and the hydrophilic heads are exposed to the aqueous phase, forming closed vesicles (vesicles) with a bilayer structure. A plurality of drugs with different polarities can be wrapped in the vesicle inner water phase and the bilayer membrane. Since the structure of liposomes is similar to that of biological membranes, they are also called artificial biological membranes. Liposomes, which consist of natural membrane components, have a bilayer structure of the liposome membrane which is in principle identical to that of natural cell membranes. In addition, liposomes can also be composed of artificially synthesized lipids to improve their chemical and biological properties.
Liposomes vary in size from tens of nanometers to tens of micrometers, and can be classified into unilamellar liposomes (unilamellar vesicles), Multilamellar Liposomes (MLV), and multivesicular liposomes (MVL) according to the type of structure. Among them, unilamellar liposomes are vesicles formed of a bilayer lipid membrane, and are classified into small unilamellar liposomes (SUV) and large unilamellar Liposomes (LUV).
The liposome as a drug carrier has the following advantages:
(1) targeting and lymph targeting; (2) the slow release effect is as follows: slow release, delay renal excretion and metabolism, thereby prolonging the action time; (3) the toxicity of the medicine is reduced; (4) improving the stability of the medicine.
The liposome has the properties of histocompatibility, cell affinity, targeting property, slow release and the like, and is widely applied to the research and development of antitumor drugs. Polyethylene glycol (PEG) and its derivatives are used for modification, so as to weaken aggregation of liposome during long-term storage, increase redispersibility, and improve stability of liposome. This is mainly due to the fact that PEG can produce a steric barrier layer with a thickness of 6nm, thereby weakening the action of plasma protein and the uptake action of endothelial reticulum, and prolonging the drug effect. And secondly, the PEG can improve the hydrophilicity of the membrane surface and reduce the affinity effect of the liposome and a mononuclear phagocyte system. The modified liposomes are referred to as long-acting liposomes or sterically stabilized liposomes. The characteristic that the surface of the liposome is easy to be modified to obtain a targeting effect is utilized, the molecular targeting liposome which targets certain specific targets on the surface of tumor cells can be prepared, the liver and kidney accumulation toxicity is reduced, the bioavailability is improved, and the liposome can play a drug effect at specific positions. The development of liposomes as drug carriers focuses mainly on three aspects: 1. the liposome can effectively reduce the side effect of the chemotherapeutic drugs and improve the therapeutic effect of the drugs; 2. the liposome is used as a carrier of protein-polypeptide vaccine and DNA vaccine, and can enhance the immune response of the organism as a vaccine carrier because the liposome is an effective immunologic adjuvant; 3. the liposome is used as a carrier of nucleic acid medicaments, and the adverse reaction of virus carriers can be avoided by using the liposome as the carrier for gene therapy.
Cancer is the second leading killer threatening human health and life worldwide, and its high incidence and high recurrence rate make the research of antitumor drugs a hot area. Most of the antitumor drugs have unobvious targeting property, and can generate side effects on normal tissues of a human body, cause irreversible damage and influence the treatment of cancers. The liposome drug delivery system has received wide attention due to its low toxicity and ability to target. At present, many liposome drugs are approved to be on the market or be clinically studied for cancer treatment at home and abroad.
The molecular targeting liposome is characterized in that a certain molecular ligand is connected to the surface of the liposome, and the ligand can specifically recognize a certain receptor on the surface of a tumor cell, so that a liposome drug can be enriched to a target cell, and the damage to normal cells is reduced. Appropriate targeting ligands can enhance the bioavailability of the drug. According to the type of ligand, molecular targeted liposomes can be classified into antibody targeted liposomes and non-antibody targeted liposomes.
Antibody-targeted liposomes, also known as Immunoliposomes (ILs), typically covalently bind monoclonal antibodies or antigen-binding fragments (Fab) of antibodies to the surface of the drug-loaded liposomes, and transport the drug to a specific site by virtue of the recognition of the antibody by an antigen or receptor on the surface of the target cell. As a novel drug carrier, the immunoliposome has many advantages, including obvious selective killing effect on tumor target cells, and the killing activity is stronger than that of free drugs, non-specific antibody liposomes, single monoclonal antibodies and the like; the medicine is specifically distributed in the body of a tumor-bearing animal, the concentration of the medicine on the tumor focus is increased, and the toxic and side effects of the medicine are small; long half life of in vivo circulation, large drug delivery amount, and the like. A variety of antibody-targeted liposomes are currently under investigation.
Due to the change of the microenvironment around the tumor cells, the surface of the tumor cells excessively express a plurality of receptors, while the surface of normal cells does not express or underexpresses the receptors, so that the targeting ligands of the receptors can be connected to the surface of the liposome to play the role of specific recognition and targeting. The invention discloses a non-antibody targeted liposome, which belongs to the non-antibody targeted liposome, and the common ligand of the non-antibody targeted liposome comprises folic acid, transferrin, polypeptide, saccharides and the like.
Microbial and viral DNA can induce an endogenous and powerful immune response in infected mammalian cells by stimulating interferon secretion. The immune response of the Endoplasmic Reticulum (ER) receptor protein (STING) to cytoplasmic DNA is an essential factor. Recent studies have shown that cyclic cGMP-AMP dinucleotide synthetase (cGAS) endogenously catalyzes cGAMP synthesis under activated conditions upon DNA binding. cGAMP is a cytosolic DNA sensor that acts as a second messenger to stimulate the induction of INF-beta by STING, mediating the activation of TBK1 and IRF-3, which in turn initiates transcription of the INF-beta gene. Recently, recombinant cGAS was reported to catalyze the cyclization of cGMP-AMP dinucleotide, gammp, under DNA binding conditions. The crystal structure of a complex where cGAS binds to double-stranded dsDNA has also been reported that cGAMP binds STING, activating the transcription factor IRF3 and producing interferon-beta.
Cyclic dinucleotide synthetase (cGAS) is an important cytoplasmic DNA receptor in the innate immune pathway. cGAMP, as a secondary messenger molecule, induces the production of interferon IFN- β and other cytokines through the STING protein pathway on the endoplasmic reticulum membrane, regulates the expression of downstream proteins, induces cell growth arrest and apoptosis, and produces antiviral effects. The STING pathway can regulate innate immune recognition of immunogenic tumors and promote anti-tumor effects of interferons. IFN-gamma plays an anti-tumor role through TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) in vivo and promotes tumor cell apoptosis. cGAMP is a key stimulator of cGAS-cGAMP-STING-IRF 3-mediated innate immune responses and is an endogenous activator of STING, and thus, cGAMP has the effect of promoting anti-tumor immune responses.
STING is a transmembrane protein of the endoplasmic reticulum, which has a hydrolase enzyme ENPP 1. No ENPP1 activity was found in the cytoplasm. Instead, it is found on the basolateral surface of the plasma membrane in hepatocytes and the rough endoplasmic reticulum portion of the liver. Its catalytic domain resides in the endoplasmic reticulum cavity and requires a high concentration of calcium ions for its activity. ENPP1 hydrolase can degrade 2 '3' -cGAMP. Experiments showed that 2 '3' -cGAMP is a good substrate for recombinant ENPP 1. Therefore, the cGAMP is wrapped by the liposome, so that the contact of the cGAMP and ENPP1 hydrolase is inhibited, the metabolic time of the cGAMP is prolonged, and the drug effect is improved. The liposome has the function of immunologic adjuvant, can target tumor cells, and improves the utilization rate of the medicament, so that the cGAMP contained in the targeted liposome has the advantages of preparing antitumor medicaments and potential application.
Disclosure of Invention
The invention aims to provide a composition of a targeted liposome-cyclic dinucleotide cGAMP, a preparation method and application thereof in preparing antitumor drugs. The cyclic dinucleotide cGAMP medicament wrapped by the targeted liposome has the advantages of targeted delivery, cell permeability enhancement, medicament metabolism period prolongation and medicament utilization rate improvement. Because the liposome has the function of immunologic adjuvant, the targeting liposome-cyclic dinucleotide cGAMP has better immunologic anti-tumor effect.
The targeted liposome-cyclic dinucleotide prepared by the invention comprises the following components: lecithin (lipoid EPCs), Cholesterol (CH), distearoylphosphatidylethanolamine-polyethylene glycol (DSPE-PEG 2000), and distearoylphosphatidylethanolamine-polyethylene glycol-Folic Acid (DSPE-PEG-Folic Acid), encapsulating a cyclic dinucleotide, cGAMP (or a derivative of cGAMP). Targeted liposome-cyclic dinucleotide cGAMP (or derivatives of cGAMP) is prepared by methods including reverse evaporation and ammonium sulfate gradient.
The experimental research of the invention shows that the target liposome-encapsulated cyclic dinucleotide cGAMP can inhibit the growth of various tumor cells, has obvious antitumor effect, has the antitumor effect obviously superior to that of the cGAMP which is independently used, and can be used for preparing antitumor drugs.
The invention also relates to an antitumor drug prepared by using the cyclic dinucleotide cGAMP wrapped by the targeting liposome. The tumors include but are not limited to lung cancer, gastric cancer, colorectal cancer, melanoma, ovarian cancer and the like.
The targeted liposome of the present invention includes, but is not limited to, targeted liposomes composed of lecithin (lipoid EPCs), Cholesterol (CH), distearoylphosphatidylethanolamine-polyethylene glycol (DSPE-PEG 2000), and distearoylphosphatidylethanolamine-polyethylene glycol-Folic Acid (DSPE-PEG-Folic Acid).
The Cyclic dinucleotide described in the present invention refers to 2 '3' -cGAMP or Cyclic [ G (2 ', 5') pA (3 ', 5') p ], but is not limited to 2 '3' -cGAMP, including derivatives of cGAMP.
The cyclic dinucleotide cGAMP slow-release antitumor drug wrapped by the targeting liposome can prolong the cGAMP metabolic cycle and enhance the antitumor treatment effect, and is superior to the antitumor treatment effect of cGAMP when being singly used.
Detailed Description
The present invention will be described in detail with reference to examples. In the present invention, the following examples are given to better illustrate the present invention and are not intended to limit the scope of the present invention.
Example 1: composition and preparation method of targeted liposome-cGAMP
And respectively preparing targeted liposomes encapsulating the cGAMP medicine by a reverse evaporation method and an ammonium sulfate gradient method, and performing property characterization on the targeted liposomes.
1. Liposome raw materials: lecithin (lipoid EPCs), Cholesterol (CH), distearoylphosphatidylethanolamine-polyethylene glycol (DSPE-PEG 2000), and distearoylphosphatidylethanolamine-polyethylene glycol-Folic Acid (DSPE-PEG-Folic Acid), all purchased from Sierrance Rexi Biotech, Inc.
2. Method for preparing liposome
(1) Reverse evaporation method
(A) Dissolving a phospholipid membrane material in chloroform serving as an organic solvent, wherein the phospholipid membrane material comprises the following components in percentage by weight:
EPC CH DSPE-PEG2000-FA = 10:10:1:0.01 (molar ratio); an organic phase solution was formed.
(B) The cGAMP drug was dissolved in ultrapure water to form an aqueous solution.
(C) Organic phase phospholipid solution: aqueous cGAMP solution =3:1 (V: V), cGAMP total phospholipid membrane mass =1:10, and aqueous drug solution is added to the phospholipid organic phase solution.
(D) The probe ultrasound, power 200W, 20 minutes (ultrasound 5 seconds, stop 3 seconds), form stable W/O emulsion.
(E) The organic solvent was removed by rotary evaporation under vacuum (water bath 45 ℃ C., rotation speed 90 rpm).
(F) And carrying out water bath ultrasound on the obtained liposome solution to reduce the particle size.
(G) Unencapsulated drug was removed by ultrafiltration using ultrafiltration tubes (MWCO =3000 Da).
(2) Ammonium sulfate gradient method
(A) Dissolving phospholipid membrane material in chloroform as organic solvent in the proportion of
EPC : CH : DSPE-PEG2000 : DSPE-PEG2000-FA = 10 : 10 : 1 : 0.01 (molar ratio);
(B) Carrying out rotary evaporation on the phospholipid solution to form a film (the water bath temperature is 35 ℃, the rotating speed is 90rpm, and the vacuum degree is 0.09 Mpa), and then carrying out rotary evaporation to remove the organic solvent;
(C) a120 mmol/L ammonium sulfate solution was added to the phospholipid membrane, and the mixture was shaken (120 rpm, 5 minutes) to form a blank liposome solution.
(D) The blank liposome solution was dialyzed overnight in ultrapure water.
(E) Dissolving cGAMP in ultrapure water, adding the solution into blank liposome solution, and incubating for 20 minutes at 65 ℃;
(F) reducing the particle size by water bath ultrasound;
(G) unencapsulated drug was removed by ultrafiltration using ultrafiltration tubes (MWCO =3000 Da).
3. Characterization of Targeted Liposomal-cGAMP
(1) Characterization of particle size
The particle size and particle size distribution (PDI) of the liposomes was measured using Dynamic Light Scattering (DLS). The basic principle is that tiny particles can randomly move (brownian motion) when being suspended in liquid, and when light passes through colloid, the particles can scatter the light, and light signals can be detected under a certain angle. Large particles move slowly, and the intensity of scattering light spots fluctuates slowly; the small particles move rapidly, the density of scattering light spots fluctuates rapidly, and finally the particle size and the distribution thereof are calculated through the fluctuation change of light intensity and the related function of the light intensity. PDI represents the uniformity of particle size and is a concept of variance. The prepared liposome has the particle size of about 100 nm and PDI = 0.426.
(2) Zeta potential
The Zeta potential is the potential difference between the continuous phase and the fluid stabilizing layer attached to the dispersed particles. Generally used to evaluate or predict the physical stability of a fine particle dispersion, the higher the absolute value of Zeta potential, the larger the electrostatic repulsion between particles, and the better the physical stability. Generally, the Zeta potential reaches 30mV in absolute value, so that the system is relatively stable. The Zeta potential absolute value of the liposome prepared by the invention is 29.5 mV, which is relatively stable.
(3) Encapsulation efficiency
Separating the target liposome from the free cGAMP medicine, measuring the amount of the free cGAMP medicine by using an ultraviolet spectrophotometer as a standard curve method, and calculating the liposome encapsulation efficiency: EE (%) = (1-Cf/Ct) × 100%. Wherein: EE-cGAMP liposome encapsulation efficiency; cf-free cGAMP content; ct total cGAMP content. According to calculation, the encapsulation rate of the liposome prepared by the reverse evaporation method is about 60 percent, and the encapsulation rate of the liposome prepared by the ammonium sulfate gradient method is more than 75 percent.
Example 2: preparation of cGAMP
cGAMP (cyclic-GMP-AMP) is synthesized catalytically by cyclic cGMP-AMP dinucleotide synthetase (cGAS) under activating conditions after binding DNA according to literature procedures. The purity is more than 98%. (Li P.W, et al., Immunity, 2013, 39(6), 1019-
Example 3: a tumor-bearing mouse model is adopted to detect the anti-tumor effect of the cGAMP slow-release medicine wrapped by the liposome, namely the inhibition effect on the growth of subcutaneous transplanted tumors of animals.
Animal(s) production
Species, strain, sex, weight, source, qualification certificate
BALB/C normal mice, C57/BL6 normal mice, male, 16-18g in weight, 6-8 weeks old, SPF grade, purchased from shanghai slaike laboratory animals llc [ laboratory animal quality certification no: SCXK (Shanghai) 2007 + 0005 ].
Feeding conditions
All mice were left to eat and drink water freely, and were bred at room temperature (23 + -2) deg.C in the laboratory animal center of the university of civil liberation military and military medical sciences of China. The feed and water are sterilized by high pressure, and the whole experimental feeding process is SPF grade.
Dose setting
I.v. mice, 1 dose group was set: 10mg/kg
Test control
Negative control: physiological saline solution
Positive control: cGAMP at a dose of 10mg/kg
Method of administration
The administration route is as follows: tail vein injection administration
Administration volume: 100 microliter/piece
The administration times are as follows: 1 time per day for 21 days
Number of animals per group: 10 pieces of
Tumor cell strain
Mouse colorectal cancer cell line CT26, mouse lung cancer Lewis tumor line LL/2, human ovarian cancer cell line SK-OV-3, human melanoma cell line A375 and human gastric cancer cell line MNK-45 are purchased from cell banks of Chinese academy of sciences.
The main steps of the test
1. Establishment and intervention of tumor model mouse
Culturing the cells, subculturing, collecting the cells at logarithmic phase of the cells to a concentration of (1.0X 10)7) Each milliliter of cell suspension, 0.2 ml of cell suspension (cell number 2.0X 10) was injected into the right anterior axillary region of the mouse6One tumor/one tumor), the tumors grow to about 5 mm in diameter after about 10 days, the tumors are successfully induced, and the tumors are randomly divided into 3 groups. Respectively is A: negative control group (i.v. saline group); b: cGAMP group (intravenous cGAMP)10mg/kg C: the targeted liposome-cGAMP group (intravenous liposome-cGAMP) was 10 mg/kg. The administration is 1 time per day for 21 days. After 21 days, mice were sacrificed and tumor weights were weighedAmount, tumor inhibition rate = [ 1-mean tumor weight in experimental group (B, C group)/mean tumor weight in group a)]×100%。
Preparing subcutaneous transplantation tumor models respectively: a mouse colorectal cancer cell line CT26, transplanted to a BalB/C common mouse; mouse lung cancer Lewis tumor strain LL/2 was transplanted into C57/BL6 mice, and the antitumor effect was observed.
2. Statistical analysis
Data are expressed in x ± s, processed using SPSS10.0 software, and the significance of tumor weight differences for each group was compared using a one-way ANOVA test, with significance level a = 0.05.
Results
After mice are inoculated with tumor cells subcutaneously, a subcutaneous tumor transplantation model is successfully prepared, the targeted liposome-cGAMP and the cGAMP can both obviously inhibit the tumor growth, the tumor weight average after 21 days of administration is obviously lower than that of a negative control group (P <0.05, P < 0.01), and the targeted liposome-cGAMP is superior to that of cGAMP in single administration, which indicates that the targeted liposome-cGAMP has better anti-tumor effect. Specific results table 1-table 5:
TABLE 1 Effect of Liposome-cGAMP on murine colorectal carcinoma cells CT26 subcutaneous transplantation tumor of BalB/C
(n=10,mean±SD)
Group average tumor weight (g) average tumor inhibition rate (%)
Negative control 2.266 + -0.244 (g) -
cGAMP group 0.748 ± 0.182 (g) × 67.0
liposome-cGAMP group 0.428 ± 0.154 (g) × 81.0
Note: p <0.05 vs negative control group; p <0.01 vs negative control group.
TABLE 2 Effect of Liposome-cGAMP on C57 mouse Lung carcinoma Lewis tumor strain LL-2 subcutaneous transplantation tumor
(n=10,mean±SD)
Group average tumor weight (g) average tumor inhibition rate (%)
Negative control 2.640 + -0.378 (g) -
cGAMP group 0.782 ± 0.245 (g) × 70.4
liposome-cGAMP group 0.505 ± 0.128 (g) × 80.8
Note: p <0.05 vs negative control group; p <0.01 vs negative control group.
TABLE 3 Effect of Liposome-cGAMP on human melanoma cell line A375 murine subcutaneous transplantation tumors
(n=10,mean±SD)
Group average tumor weight (g) average tumor inhibition rate (%)
Negative control 2.680 + -0.368 (g) -
cGAMP group 0.768 + -0.254 (g). 71.3
Liposome-cGAMP group 0.498. + -. 0.135 (g). 81.4
Note: p <0.05 vs negative control group; p <0.01 vs negative control group.
TABLE 4 Effect of Liposome-cGAMP on murine subcutaneous transplantable tumors of human gastric carcinoma cell line MNK-45
(n=10,mean±SD)
Group average tumor weight (g) average tumor inhibition rate (%)
Negative control group 2.668 + -0.382 (g) -
cGAMP group 0.786 + -0.265 (g). 70.5
liposome-cGAMP group 0.510 ± 0.128 (g) × 80.8
Note: p <0.05 vs negative control group; p <0.01 vs negative control group.
TABLE 5 Effect of Liposome-cGAMP on human ovarian cancer cell line SK-OV-3 murine subcutaneous transplantation tumor
(n=10,mean±SD)
Group average tumor weight (g) average tumor inhibition rate (%)
Negative control group 2.728 + -0.336 (g) -
cGAMP group 0.769 ± 0.258 (g) × 71.8
liposome-cGAMP group 0.509 ± 0.156 (g) × 81.3
Note: p <0.05 vs negative control group; p <0.01 vs negative control group.
Example 3 acute toxicity study of Liposome-cGAMP
Experimental Material
20 ICR mice (purchased from Shanghaisleke laboratory animals, Limited liability company [ laboratory animal quality certification number: SCXK (Shanghai) 2007-0005 ]), each half of male and female, the weight of the mice is 18-22 g, and the mice are fed with pellet feed and can freely eat and drink water.
Liposome-cGAMP was prepared from example 1 and formulated with physiological saline into a solution at a concentration of 200 mg/mL.
Experimental methods
ICR mice were injected with 2g/kg of liposome-cGAMP sustained release drug by single tail vein injection according to body weight, and the mice were observed for toxicity and death within 14 days after administration. As a result, it was found that the mice were normally active after a single tail vein injection administration. Within 14 days after administration, the mice did not die, and on day 15, all mice were sacrificed, dissected, and examined by naked eyes for each organ, and no obvious lesion was observed.
Results of the experiment
The result of the acute toxicity experiment shows that the maximum tolerance MTD of intravenous injection administration is not less than 2g/Kg, which indicates that the liposome-cGAMP medicament has low acute toxicity.

Claims (3)

1. A targeted liposome-cyclic dinucleotide, cGAMP, consisting of: a targeted liposome consisting of lecithin, cholesterol, distearoyl phosphatidyl ethanolamine-polyethylene glycol folic acid, and a liposome-encapsulated cyclic dinucleotide cGAMP;
the preparation method of the targeting liposome-cyclic dinucleotide cGAMP comprises the following steps:
the liposome membrane material is dissolved in chloroform which is an organic solvent, and the mole ratio of each component in the liposome membrane material is lecithin: cholesterol: distearoylphosphatidylethanolamine-polyethylene glycol: distearoyl phosphatidyl ethanolamine-polyethylene glycol-folic acid-10: 10:1:0.01, then carrying out rotary evaporation to form a membrane, carrying out vacuum pumping on the residual organic solvent, adding 120mmol/L ammonium sulfate solution into the organic solution of the liposome membrane material, shaking to form a blank liposome solution, dialyzing the blank liposome solution in ultrapure water overnight, adding a cGAMP aqueous solution into the blank liposome solution, incubating at 60-65 ℃, carrying out water bath ultrasound treatment, and then carrying out ultrafiltration by using an ultrafiltration tube to remove unencapsulated cGAMP;
the target application object of the liposome-cyclic dinucleotide cGAMP is at least one of lung cancer, gastric cancer, colorectal cancer, melanoma and ovarian cancer.
2. The sustained-release drug prepared by using the targeted liposome-cyclic dinucleotide cGAMP according to claim 1, wherein the sustained-release drug is an antitumor drug, an anti-neurodegenerative disease drug, an anti-cardiovascular and cerebrovascular drug, an anti-diabetic drug.
3. The sustained-release drug prepared from targeted liposome-cyclic dinucleotide cGAMP according to claim 2, characterized in that: the antitumor drug is prepared by unit preparations with different specifications and pharmaceutically acceptable carriers, and the prevention or treatment of tumors and directly related diseases thereof is carried out by one or more administration routes of oral administration or injection.
CN201710147574.5A 2017-03-13 2017-03-13 Composition of targeted liposome-cyclic dinucleotide, preparation method and application of targeted liposome-cyclic dinucleotide in resisting tumors Active CN106667914B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201710147574.5A CN106667914B (en) 2017-03-13 2017-03-13 Composition of targeted liposome-cyclic dinucleotide, preparation method and application of targeted liposome-cyclic dinucleotide in resisting tumors

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201710147574.5A CN106667914B (en) 2017-03-13 2017-03-13 Composition of targeted liposome-cyclic dinucleotide, preparation method and application of targeted liposome-cyclic dinucleotide in resisting tumors

Publications (2)

Publication Number Publication Date
CN106667914A CN106667914A (en) 2017-05-17
CN106667914B true CN106667914B (en) 2022-02-01

Family

ID=58828853

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710147574.5A Active CN106667914B (en) 2017-03-13 2017-03-13 Composition of targeted liposome-cyclic dinucleotide, preparation method and application of targeted liposome-cyclic dinucleotide in resisting tumors

Country Status (1)

Country Link
CN (1) CN106667914B (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JOP20170192A1 (en) 2016-12-01 2019-01-30 Takeda Pharmaceuticals Co Cyclic dinucleotide
WO2019092660A1 (en) 2017-11-10 2019-05-16 Takeda Pharmaceutical Company Limited Sting modulator compounds, and methods of making and using
CN110227166A (en) * 2019-06-13 2019-09-13 芜湖先声中人药业有限公司 The pharmaceutical carrier and preparation method thereof of sustained-release chemotherapy targeting preparation in a kind of Tumor Resection
CN113813375B (en) * 2020-06-19 2023-06-16 杭州星鳌生物科技有限公司 Composition of novel anti-novel coronavirus complex and application of novel anti-novel coronavirus complex in medicines for preventing and treating coronavirus infection diseases
MX2023005381A (en) 2020-11-09 2023-05-23 Takeda Pharmaceuticals Co Antibody drug conjugates.
CN114569627A (en) * 2020-12-02 2022-06-03 杭州星鳌生物科技有限公司 Preparation and composition of novel STING agonist compound and application of novel STING agonist compound in antitumor drugs
CN115252554B (en) * 2021-05-01 2023-08-25 杭州星鳌生物科技有限公司 Preparation, composition and application of novel 4 (3H) -quinazolinone analogue/cyclic dinucleotide cGAMP co-carrier liposome in antitumor drugs
CN113288871B (en) * 2021-05-28 2022-09-16 华中科技大学 Pharmaceutical composition liposome formulations for modulation of epigenetic and immune checkpoints
CN114903856A (en) * 2022-05-06 2022-08-16 四川大学华西医院 2 ', 3' -cGAMP-loaded liposome nano preparation and preparation method and application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101474155A (en) * 2009-01-24 2009-07-08 重庆医科大学 Lung-targeted medicine carrying precursor liposome for injection and method of use thereof
CN103989624A (en) * 2013-02-20 2014-08-20 中国科学院上海药物研究所 Irinotecan hydrochloride composition and preparation method thereof
WO2014179760A1 (en) * 2013-05-03 2014-11-06 The Regents Of The University Of California Cyclic di-nucleotide induction of type i interferon
CN104540945A (en) * 2012-04-30 2015-04-22 格伦·N·巴伯 Modulating immune responses

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101474155A (en) * 2009-01-24 2009-07-08 重庆医科大学 Lung-targeted medicine carrying precursor liposome for injection and method of use thereof
CN104540945A (en) * 2012-04-30 2015-04-22 格伦·N·巴伯 Modulating immune responses
CN103989624A (en) * 2013-02-20 2014-08-20 中国科学院上海药物研究所 Irinotecan hydrochloride composition and preparation method thereof
WO2014179760A1 (en) * 2013-05-03 2014-11-06 The Regents Of The University Of California Cyclic di-nucleotide induction of type i interferon

Also Published As

Publication number Publication date
CN106667914A (en) 2017-05-17

Similar Documents

Publication Publication Date Title
CN106667914B (en) Composition of targeted liposome-cyclic dinucleotide, preparation method and application of targeted liposome-cyclic dinucleotide in resisting tumors
CN106727331B (en) Composition of immunoliposome-cycloddinucleotide, preparation method and application of immunoliposome-cycloducleotide in resisting tumor
CN110522918B (en) Targeting element and preparation method and application thereof
WO2019233300A1 (en) Composition of anti-tumor compound medicine and use thereof in combating tumor
KR100794449B1 (en) Composition of cationic phospho lipid nanoparticles for effective delivery of nucleic acids
CN112826808B (en) Neutral/cation mixed lipid nano preparation of cyclic dinucleotide or analogue thereof and application thereof
Abdel‐Bar et al. Combinatory delivery of etoposide and siCD47 in a lipid polymer hybrid delays lung tumor growth in an experimental melanoma lung metastatic model
Pi et al. Exosomes: powerful weapon for cancer nano-immunoengineering
Dai et al. Nanomedicines modulating myeloid-derived suppressor cells for improving cancer immunotherapy
CN105012956A (en) Difunctional tumor targeted liposome drug-delivery system and preparation and application thereof
US20220287969A1 (en) Multilamellar rna nanoparticles
Cao et al. Extracellular vesicles as drug vectors for precise cancer treatment
EP4119127A1 (en) Composition of novel immune agonist compound and use thereof in medicine capable of resisting multiple diseases
WO2017162055A1 (en) Application of cyclic dinucleotide cgamp-liposome for resisting tumours
CN108653312B (en) Antitumor study of activator of endoplasmic reticulum receptor protein STING in combination with inhibitor of phosphodiesterase ENPP1
Hao et al. Lipid‐based nanoparticles as drug delivery systems for cancer immunotherapy
US20100209494A1 (en) Liposomes and Uses Thereof
CN104784118A (en) Acid-sensitive liposome drug delivery system with internalization effect, and preparation method and applications thereof
CN110898231B (en) Functionalized Lalotxel liposome and preparation method and application thereof
Nie et al. Nonviral vector system for cancer immunogene therapy
Jiang et al. Remodeling the tumor immune microenvironment via siRNA therapy for precision cancer treatment
Qu et al. Advanced nano-based strategies for mRNA tumor vaccine
US20220378936A1 (en) Delivery system complexes comprising a precipitate of an active agent and methods of use
Li et al. Nanoparticle technology for mRNA: Delivery strategy, clinical application and developmental landscape
CN114191539A (en) Exosome nano particle for composite co-transport of small molecule nucleic acid and active protein, and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20190424

Address after: 311200 C-806, Kaidi Road, Xiaoshan Economic and Technological Development Zone, Xiaoshan District, Hangzhou City, Zhejiang Province

Applicant after: Hangzhou star bioscience Co., Ltd.

Address before: 252000 Room 504, Unit 2, Building 15, Building 13 East Road, Dongchangfu District, Liaocheng City, Shandong Province

Applicant before: Liaocheng city run bio Pharmaceutical Technology Co., Ltd.

GR01 Patent grant
GR01 Patent grant