CN117466777B - Cationic lipid compound, preparation method and application thereof and mRNA delivery system - Google Patents

Cationic lipid compound, preparation method and application thereof and mRNA delivery system Download PDF

Info

Publication number
CN117466777B
CN117466777B CN202311819318.8A CN202311819318A CN117466777B CN 117466777 B CN117466777 B CN 117466777B CN 202311819318 A CN202311819318 A CN 202311819318A CN 117466777 B CN117466777 B CN 117466777B
Authority
CN
China
Prior art keywords
compound
cationic lipid
reaction
mrna
delivery system
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
CN202311819318.8A
Other languages
Chinese (zh)
Other versions
CN117466777A (en
Inventor
赵钊
苏焘
李红燕
李三朋
郭凤娟
何华美
孙榕
刘方润
林燕真
万季
潘有东
王弈
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.)
Shenzhen Xinhe Ruien Biomedical Technology Co ltd
Beijing Xinhe Ruien Biomedical Technology Co ltd
Shenzhen Neocura Biotechnology Corp
Original Assignee
Shenzhen Xinhe Ruien Biomedical Technology Co ltd
Beijing Xinhe Ruien Biomedical Technology Co ltd
Shenzhen Neocura Biotechnology Corp
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 Shenzhen Xinhe Ruien Biomedical Technology Co ltd, Beijing Xinhe Ruien Biomedical Technology Co ltd, Shenzhen Neocura Biotechnology Corp filed Critical Shenzhen Xinhe Ruien Biomedical Technology Co ltd
Priority to CN202311819318.8A priority Critical patent/CN117466777B/en
Publication of CN117466777A publication Critical patent/CN117466777A/en
Application granted granted Critical
Publication of CN117466777B publication Critical patent/CN117466777B/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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C227/00Preparation of compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C227/04Formation of amino groups in compounds containing carboxyl groups
    • C07C227/06Formation of amino groups in compounds containing carboxyl groups by addition or substitution reactions, without increasing the number of carbon atoms in the carbon skeleton of the acid
    • C07C227/08Formation of amino groups in compounds containing carboxyl groups by addition or substitution reactions, without increasing the number of carbon atoms in the carbon skeleton of the acid by reaction of ammonia or amines with acids containing functional groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C239/00Compounds containing nitrogen-to-halogen bonds; Hydroxylamino compounds or ethers or esters thereof
    • C07C239/08Hydroxylamino compounds or their ethers or esters
    • C07C239/20Hydroxylamino compounds or their ethers or esters having oxygen atoms of hydroxylamino groups etherified
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C259/00Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups
    • C07C259/04Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids
    • C07C259/06Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids having carbon atoms of hydroxamic groups bound to hydrogen atoms or to acyclic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C67/00Preparation of carboxylic acid esters
    • C07C67/08Preparation of carboxylic acid esters by reacting carboxylic acids or symmetrical anhydrides with the hydroxy or O-metal group of organic compounds
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Preparation (AREA)

Abstract

The invention relates to a cationic lipid compound, a preparation method and application thereof, and an mRNA delivery system, relates to the technical field of medical biology, and solves the problem that the development of the cationic lipid compound with high delivery efficiency and targeting property is needed at present. Wherein the cationic lipid compound has the structure shown below:the method comprises the steps of carrying out a first treatment on the surface of the The cationic lipid compound is used for preparing an mRNA delivery system, and has the characteristics of less liver accumulation, high safety, high delivery efficiency and strong spleen targeting.

Description

Cationic lipid compound, preparation method and application thereof and mRNA delivery system
Technical Field
The invention relates to the technical field of medical biology, in particular to a cationic lipid compound, a preparation method and application thereof, and an mRNA delivery system.
Background
mRNA (messenger ribonucleic acid) is a single-stranded ribonucleic acid which is polymerized by taking one strand of a DNA (deoxyribonucleic acid) double strand as a template (template) and taking 4 kinds of ribonucleoside triphosphates (A, U, G, C) as substrates under the catalysis of RNA polymerase (RNA polymerase) through phosphodiester bonds. mRNA is capable of carrying and transmitting genetic information stored in the nuclear DNA, and plays a key role in the conversion of genetic information into functional proteins. In the cytoplasm, the immature mRNA is modified into mature mRNA through capping, tailing, intron cutting and other steps, and the mature mRNA can accurately guide the synthesis process of protein in the cytoplasm. Relatively, mRNA is easy to transfect due to its much smaller molecular weight than DNA, and there is no oncogenic risk of integration into the host DNA to initiate insertion mutations. Therefore, mRNA is taken as a preventive and therapeutic drug, and has great advantages and potential in the prevention and treatment of various diseases.
mRNA nucleic acid medicine is one kind of preventing and treating strategy for preventing and treating diseases with functional protein or subunit activating host immune system to produce corresponding humoral immunity or cell immunity reaction and for treating diseases with expressed protein or subunit possessing function of treating diseases or regulating the expression of other genes. Compared with other methods, the method has the advantages that the method can directly activate the organism on the molecular level to generate functional antibodies or cellular immune responses aiming at specific pathogens, or can purposefully repair pathogenic genes or correct the expression of abnormal genes, thereby achieving the effects of preventing and treating various diseases. The mRNA nucleic acid medicine can achieve the effect that the traditional medicine cannot replace, for example, the monoclonal antibody medicine can only act on the cell surface, but the mRNA nucleic acid medicine can not only act on the extracellular protein of the cell membrane, but also act on the intracellular protein, even act in the nucleus, and has accurate targeting. Of the 7000 diseases faced by humans, about 1/3 of the diseases are clinically almost drug-free due to problems (deletion, reduction or overexpression) of functional genes, such as Hemophilia (Hemophilia), duchenne Muscular Dystrophy (DMD), cystic fibrosis (cytosticibrosis), and severe immunodeficiency Syndrome (SCID), etc., and mRNA nucleic acid drugs are very advantageous for this monogenic disease. In the age background of popularization of personalized medicine and accurate medicine. In theory, diseases caused by gene differences or abnormal gene expression of patients can be accurately and effectively treated by using mRNA nucleic acid medicaments.
mRNA nucleic acid drugs have great advantages and potential in controlling gene expression and preventing and treating malignant diseases. However, there are difficulties in the development, preparation and subsequent systemic administration of such drugs. Firstly, mRNA exists in a single-chain form, so that the mRNA is extremely unstable in vitro and under physiological conditions, and is easily degraded by RNA nuclease (RNAase) in air or blood, and is also easily cleared by mononuclear macrophages in tissues and organs such as liver, spleen and the like; secondly, due to the electronegativity of mRNA, it is difficult to penetrate the cell membrane into the cell interior; again, mRNA is difficult to escape from endosomes and into the cytoplasm to function. Furthermore, uracil ribonucleoside (U) in mRNA is prone to immunogenicity, which in some cases may increase the potential toxic side effects of mRNA drugs. Finally, the susceptibility to off-target effects is also an important challenge in the preparation and administration of mRNA nucleic acid-based drugs. Therefore, the development of an intracellular delivery system of an mRNA nucleic acid drug is a key point of being capable of large-scale clinical application.
To more safely and effectively exert the therapeutic capacity of RNA, scientists use Lipid Nanoparticles (LNPs) to encapsulate and deliver RNA to specific sites in the body. This RNA delivery strategy has shown great utility in delivering double-stranded small interfering RNAs (siRNAs, 21 to 23 nucleotides in length). For example, lipid C12-200 has been widely used in the manufacture of siRNA-LNP formulations for various therapeutic applications in vivo to inhibit protein expression. The advent of synthetic ionizable lipid materials (synthetic ionizable lipids) and lipid materials (lipid-materials) has not only greatly reduced the in vivo toxicity of LNPs, but also made it possible to deliver large molecular weight RNAs (e.g., mRNA) in vivo. These amine-containing ionizable lipids or lipid molecules are positively charged and, by means of electrostatic attraction, can efficiently bind to negatively charged mRNA and self-assemble to form LNP. LNP can improve blood circulation time of RNA and increase uptake rate of cells; in the cell, RNA is released into cytoplasm through endosome escape way to express specific protein and to play a certain therapeutic role.
The LNP has the following 4 main raw material components: 1) Ionizable lipids or lipid molecules such as DLin-KC2-DMA, DLin-MC3-DMA, L319. This is the core component that enables in vivo delivery of mRNA. 2) Phospholipid molecules, a phospholipid, provide structure for the LNP bilayer and may also assist endosomal escape; 3) Cholesterol, enhancing LNP stability, promoting membrane fusion; 4) Polyethylene glycols such as DMG-PEG2000, which control and reduce the particle size of the LNP and "protect" the LNP from non-specific endocytosis of immune cells.
In vivo, changes in the raw materials and components of LNP can have profound effects on the physicochemical stability and efficacy of mRNA action of mRNA-LNP formulations. Existing LNP component protocols do not fully exploit the efficacy of mRNA-LNP formulations, requiring continuous structural adjustments to the ionizable lipid molecules and design optimization for specific RNAs. Therefore, there is a need to develop a targeted cationic lipid compound with high delivery efficiency.
Disclosure of Invention
In view of the above, the present invention provides a cationic lipid compound, a preparation method and application thereof, and an mRNA delivery system, and is mainly aimed at developing a cationic lipid compound with less liver accumulation, high delivery efficiency, and strong spleen targeting.
In order to achieve the above purpose, the present invention mainly provides the following technical solutions:
in one aspect, embodiments of the present invention provide a cationic lipid compound, wherein the cationic lipid compound has the structure shown below:
preferably, the cationic lipid compound is prepared according to the following equation:
wherein compound F, compound J, K 2 CO 3 Putting KI and 1,4-dioxane into a reaction vessel, and reacting at 125-135 ℃; and after the reaction is finished, sequentially performing extraction and purification treatment to obtain the cationic lipid compound.
Preferably, the compound F is prepared as follows:
wherein, compound E is dissolved in tetrahydrofuran THF, isobutyl chloroformate IBCF and N-methylmorpholine NMM are added, and the reaction solution is dried after stirring at room temperature; then, adding the compound D into the mixture to react; after the reaction is finished, extraction and purification treatment are carried out to obtain the compound F.
Preferably, the compound D is prepared as follows:
wherein, compound C, hydrochloric acid and 1,4-dioxane are added into a reaction vessel to react at 45-55 ℃; after the reaction is finished, the pH value is regulated to 8-9, and extraction and purification are carried out to obtain the compound D.
Preferably, the preparation equation of the compound C is as follows:
wherein, compound A, compound B and 1, 8-diazabicyclo (5.4.0) undec-7-ene DBU are added into a reaction vessel, tetrahydrofuran THF is then added, and the mixture is heated and stirred at the temperature of 70-80 ℃ for reaction; after the reaction is finished, extracting and purifying to obtain the compound C.
Preferably, the compound J is prepared as follows:
wherein, an intermediate and a compound I, K are added into a reaction vessel 2 CO 3 Adding tetrahydrofuran THF into the KI, heating and stirring at 70-80 ℃ to react; and (3) extracting and purifying after the reaction is finished to obtain the compound J.
Preferably, the intermediate is prepared as follows:
adding a compound H into a reaction vessel, dissolving in tetrahydrofuran THF, adding carbodiimide hydrochloride EDCI into the reaction vessel, stirring at room temperature, adding p-dimethylaminopyridine DMAP, stirring at room temperature again, adding a compound G into the reaction vessel, and stirring for reaction; after the reaction is finished, extracting and purifying are carried out to obtain an intermediate.
In a further aspect, the use of a cationic lipid compound of any of the above claims in the preparation of an mRNA delivery system.
In yet another aspect, embodiments of the present invention provide an mRNA delivery system, wherein the mRNA delivery system comprises the cationic lipid compound described above.
Preferably, the mRNA delivery system is an mRNA-LNP delivery system.
Compared with the prior art, the cationic lipid compound, the preparation method and application thereof and the mRNA delivery system have at least the following beneficial effects:
in one aspect, embodiments of the present invention develop a novel cationic lipid compound having the structure shown below:
the cationic lipid compound is used for preparing an mRNA delivery system, and has the characteristics of less liver accumulation, high safety, high delivery efficiency and strong spleen targeting.
On the other hand, the embodiment of the invention provides an mRNA delivery system (mRNA-LNP delivery system), and the mRNA delivery system comprises the cationic lipid compound, so that the LNP of the mRNA delivery system has the characteristics of less liver accumulation, high safety, high delivery efficiency and strong spleen targeting.
The foregoing description is only an overview of the present invention, and is intended to provide a better understanding of the present invention, as it is embodied in the following description, with reference to the preferred embodiments of the present invention and the accompanying drawings.
Drawings
FIG. 1 is the in vivo delivery efficiency of tail intravenous injection of XH152 lipid nanoparticle formulations of the present invention and controls; wherein, (a) graph shows the quantitative distribution result of luciferase in liver after intravenous injection of lipid nanoparticle preparation in tail of mice for 6 hours; (b) The figure shows the quantitative distribution results of luciferase in spleen after 6 hours of intravenous lipid nanoparticle preparation in tail of mice; (c) The figures are comparison graphs of spleen targeting 6 hours after intravenous lipid nanoparticle formulation at the tail of mice.
FIG. 2 is the in vivo delivery efficiency by intramuscular injection of XH152 lipid nanoparticle formulations of the invention and controls; wherein, (a) graph shows the quantitative result of distribution of luciferase in liver after 6 hours of intramuscular injection of lipid nanoparticle preparation into mice; (b) The figure shows the quantitative results of luciferase distribution in spleen after 6 hours of intramuscular injection of lipid nanoparticle formulation into mice.
Detailed Description
In order to further describe the technical means and effects adopted for achieving the preset aim of the invention, the following detailed description refers to the specific implementation, structure, characteristics and effects according to the application of the invention with reference to the accompanying drawings and preferred embodiments. In the following description, different "an embodiment" or "an embodiment" do not necessarily refer to the same embodiment. Furthermore, the particular features, structures, or characteristics of one or more embodiments may be combined in any suitable manner.
The invention mainly develops a cationic lipid compound with less liver accumulation, high delivery efficiency, good safety and strong spleen targeting (the compound of the invention is named as a compound XH 152); wherein the cationic lipid compound has the following structure:
compound XH152
The synthesis method of the cationic lipid compound comprises the following steps:
1. synthesis of Compound C
In which Compound A (34.81 g,180.2 mmol), compound B (20.00 g,150.2 mmol), 1, 8-diazabicyclo (5.4.0) undec-7-ene DBU (45.73 g,300.4 mmol) and THF (200 mL) were added to a 500mL Schlenk tube, followed by heating and stirring at 70℃overnight, and TLC monitored the reaction. TLC (petroleum ether PE: ethyl acetate ea=3:1) iodination showed complete reaction of starting material, with a new main point being generated, stopping the reaction. Extraction with ethyl acetate and water, separation, collection of the organic phase, drying, spin-drying, and then addition of silica gel to carry out stirring, gave compound C (23 g, 62%) as a colourless oil by column chromatography (petroleum ether PE: ethyl acetate ea=10:1).
2. Synthesis of Compound D
To a 25mL single vial was added compound C (20 g,82.173 mmol), aqueous hydrochloric acid (10 mL): 1,4-dioxane (5 mL) =2:1, reacted at 50 ℃ for 4h, tlc monitored the reaction. After completion of the reaction, the reaction mixture was adjusted to pH 8-9 with saturated aqueous potassium carbonate, extracted three times with ethyl acetate, concentrated under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate=5:1) to give colorless oily liquid D (9.0 g, 75%).
Compound D has a molecular weight of 145, lc-MS (M/z) [ m+h ] =146.18, rt= 1.054 min.
3. Synthesis of Compound F
To a 50mL single flask was added compound E (5.00 g,22.4 mmol), dissolved in THF (20 mL), followed by isobutyl chloroformate IBCF (3.37 g,24.65 mmol) and N-methylmorpholine NMM (2.89 g,24.65 mmol), stirred at room temperature for 15min, the reaction mixture was dried by spin-drying, followed by compound D (3.26 g,22.41 mmol) and THF (20 mL), stirred at room temperature overnight, and TLC monitored the reaction. The TLC (dichloromethane DCM: methanol meoh=50:1) iodometric showed complete reaction of starting material with a new starting point generated and stopped. Extraction with ethyl acetate and water, separation, collection of the organic phase, drying, spin-drying, then addition of silica gel and stirring, gave compound F (2.9 g, 37%) as a colourless oil by column chromatography (DCM: meoh=500:1 to 100:1).
Wherein, the molecular weight of the compound F is 350.34, LC-MS (M/z) [ M+H ] =351.82, rt= 2.766min.
4. Synthetic intermediates
In a 1000mL single flask, compound H (60.00G, 268.9 mmol) was added, dissolved in THF (600 mL), EDCI (77.33G, 403.388) was added, stirred at room temperature for 0.5H, DMAP (65.71G, 537.9 mmol) was added, stirred at room temperature for 15min, G (68.97G, 268.9 mmol) was added, stirred at room temperature overnight, and the reaction monitored by thin layer chromatography TLC. Among them, TLC (petroleum ether PE: ethyl acetate EA=30:1) iodine color shows that the starting material has reacted completely, a new main point is generated, and the reaction is stopped. Extraction with ethyl acetate and water, separation, collection of the organic phase, drying, spin-drying, and then addition of silica gel to sample, gave a colorless oily liquid intermediate (42 g, 34%) by column chromatography (petroleum ether).
5. Synthesis of Compound J
Wherein, the intermediate (20.00 g,43.33 mmol), compound I (3.25 g,43.33 mmol), K, was added to a 250mL Schlenk tube 2 CO 3 (11.98 g,86.66 mmol), KI (7.19 g,43.33 mmol), and THF (100 mL) were added and stirred overnight at 70deg.C with TLC monitoring. The TLC (dichloromethane: methanol=20:1) iodine color showed complete reaction of the starting material, with a new main spot being generated, stopping the reaction. Extraction with ethyl acetate and water, separation, collection of the organic phase, drying, spin-drying, and then addition of silica gel to sample, gave compound J as a pale yellow oil (5.7 g, 29%) by column chromatography (dichloromethane: methanol=500:1 to 100:1).
Compound J has a molecular weight of 454.78, lc-MS (M/z) [ m+h ] =456.68, rt=3.327 min.
6. Synthesis of cationic lipid Compound XH152 of the present invention
Wherein, compound F (2.80 g,7.99 mmol) and compound J (4.37 g,9.59 mmol) were charged into a 50mL pressure-resistant tube, K 2 CO 3 (2.21 g,16.0 mmol), KI (1.59 g,9.59 mmol), 1,4-dioxane (30 mL) was added and stirred overnight at 130℃with heating, and TLC monitoring of the reaction. The TLC (dichloromethane: methanol=20:1) iodine color showed complete reaction of the starting material, with a new main spot being generated, stopping the reaction. Extraction with ethyl acetate and water, separation, collection of the organic phase, drying, spin-drying, and then addition of silica gel to sample, and column chromatography (dichloromethane: methanol=500:1 to 100:1) gave compound XH152 (2.00 g, 35%). The crude product was prepared in high performance liquid phase to give the final product (599 mg) as a pale yellow oil with a purity of 97.8%.
The nuclear magnetic data of the compound XH152 are as follows: 1 H NMR (400 MHz, CDCl 3 ) δ 4.85 (t,J= 6.2 Hz, 1H), 3.94 – 3.69 (m, 4H),3.48 (s, 1H), 3.32 – 3.14 (m, 1H), 2.81 (d,J= 6.6 Hz, 2H), 2.59 (s, 3H), 2.27 (t,J= 7.5 Hz, 2H), 2.07 (s, 2H), 1.78 (p,J= 5.8 Hz, 2H), 1.71 – 1.44 (m, 14H), 1.41 – 1.16 (m, 47H), 0.87 (t,J= 6.6 Hz, 9H). C 44 H 88 N 2 O 5 Exact Mass: 724.67, found [M+H] + : 725.75.
the technical effect of the use of the cationic lipid compound XH152 of the present invention in an mRNA-LNP delivery system is described below by way of example.
Example 1
And (3) preparing and detecting the lipid nanoparticle preparation.
1. Preparation steps
The cationic lipid compound XH152 of the present invention is mixed with distearoyl phosphatidylcholine DSPC, cholesterol and DMG-PEG2000 at a ratio of 50:10:38.5:1.5 in ethanol to prepare an ethanol lipid solution.
N1-methyl-pseudouridine modified luciferase mRNA was diluted with 50mM citrate buffer (pH=4.0) to give an aqueous mRNA solution.
By microfluidic device 1:3 volume ratio of ethanol lipid solution and mRNA aqueous solution to prepare lipid nanoparticles, 1X PBS dialysis was performed for 18 hours to remove ethanol and complete the citrate buffer exchange procedure. Finally, the lipid nanoparticle solution was subjected to sterile filtration (0.2 μm) and ultrafiltration concentration steps to obtain a lipid nanoparticle preparation encapsulating luciferase mRNA, named: XH152 lipid nanoparticle formulations, abbreviated XH152 LNP.
In addition, lipid nanoparticle formulations of compound SM102, compound XH106, and compound XH160 were prepared as controls using the same methods as described above, and were named: SM102 lipid nanoparticle formulation (abbreviated as SM102 LNP), XH106 lipid nanoparticle formulation (abbreviated as XH106 LNP), XH160 lipid nanoparticle formulation (abbreviated as XH160 LNP).
SM102 was a cationic lipid used in the Moderna covd-19 vaccine, used as a control. In addition, the structural formula of the compound XH160 is as follows
The structural formula of compound XH106 is as follows:
the preparation methods of the compound XH106 and the compound XH160 are basically the same as those of the cationic lipid compound XH152 of the present invention.
2. Detection step
Lipid nanoparticle particle size, polydispersity index (PDI) and potential (Zeta) were determined using a Litesizer ™ (Anton Paar, austria). Wherein, the particle size and the potential were measured in 0.1% PBS. Lipid nanoparticle formulation encapsulation efficiency (EE%) was measured by the RiboGreen method. The test results are shown in table 1:
as can be seen from table 1: the cationic lipid compound XH152 of the present invention can form lipid nanoparticles as cationic lipids.
Example 2
The lipid nanoparticle formulation prepared in example 1 was subjected to in vivo transfection experiments.
BALB/c mice were randomly divided into eight groups of 3. Each group was injected with SM102 lipid nanoparticle formulation or XH106 lipid nanoparticle formulation or XH160 lipid nanoparticle formulation or XH152 lipid nanoparticle formulation in a single tail vein (0.3 mg/kg dose) or muscle (0.15 mg/kg dose). Specifically, a first set of single tail veins (0.3 mg/kg dose) were injected with XH152 lipid nanoparticle formulations. A second set of single tail veins (0.3 mg/kg dose) were injected with XH106 lipid nanoparticle formulations. A third set of single tail veins (0.3 mg/kg dose) were injected with XH160 lipid nanoparticle formulations. A fourth set of single tail vein (0.3 mg/kg dose) was injected with SM102 lipid nanoparticle formulation. A fifth group of single muscles (0.15 mg/kg dose) was injected with the XH152 lipid nanoparticle formulation. A sixth group of single muscles (0.15 mg/kg dose) was injected with XH106 lipid nanoparticle formulation. A seventh group of single muscles (0.15 mg/kg dose) was injected with XH160 lipid nanoparticle formulation. An eighth group of single muscles (0.15 mg/kg dose) was injected with SM102 lipid nanoparticle formulation.
After 6h of injection, the luciferase substrate was injected intraperitoneally, and after 10min, organs such as liver, spleen and the like were dissected and taken, and the signals were observed and quantified using a small animal living imaging system.
The results of tail vein injection are shown in fig. 1. Referring to fig. 1 (a), the XH152 lipid nanoparticle formulation was significantly lower in the liver site than the SM102 lipid nanoparticle formulation, XH106 lipid nanoparticle formulation. Referring to the graph (b) in fig. 1, the expression of the XH152 lipid nanoparticle formulation of the present invention was significantly higher at the spleen site than that of the SM102 lipid nanoparticle formulation, the XH106 lipid nanoparticle formulation, and the XH160 lipid nanoparticle formulation. Referring to fig. 1 (c), the expression of spleen targeting of the XH152 lipid nanoparticle formulation of the present invention was significantly higher than that of the SM102 lipid nanoparticle formulation, XH106 lipid nanoparticle formulation.
The intramuscular injection results are shown in FIG. 2. Among them, referring to the graph (a) in fig. 2, the XH152 lipid nanoparticle formulation of the present invention has lower expression at the liver site than the SM102 lipid nanoparticle formulation. Referring to fig. 2 (b), the expression of the XH152 lipid nanoparticle formulation of the present invention was higher than that of the SM102 lipid nanoparticle formulation, the XH106 lipid nanoparticle formulation, and the XH160 lipid nanoparticle formulation at the spleen site.
From this, the cationic lipid compound XH152 of the present invention has the characteristics of less liver accumulation, high delivery efficiency and strong spleen targeting.
The above description is only of the preferred embodiments of the present invention, and is not intended to limit the present invention in any way, but any simple modification, equivalent variation and modification made to the above embodiments according to the technical substance of the present invention still fall within the scope of the technical solution of the present invention.

Claims (10)

1. A cationic lipid compound, characterized in that the cationic lipid compound has the structure shown below:
2. the method for preparing a cationic lipid compound according to claim 1, wherein the cationic lipid compound is prepared by the following formula:
wherein compound F, compound J, K 2 CO 3 Putting KI and 1,4-dioxane into a reaction vessel, and reacting at 125-135 ℃; and after the reaction is finished, sequentially performing extraction and purification treatment to obtain the cationic lipid compound.
3. The method for preparing a cationic lipid compound according to claim 2, wherein the preparation equation of the compound F is as follows:
wherein, compound E is dissolved in tetrahydrofuran THF, isobutyl chloroformate IBCF and N-methylmorpholine NMM are added, and the reaction solution is dried after stirring at room temperature; then, adding the compound D into the mixture to react; after the reaction is finished, extraction and purification treatment are carried out to obtain the compound F.
4. A method for preparing a cationic lipid compound according to claim 3, wherein the preparation equation of the compound D is as follows:
wherein, compound C, hydrochloric acid and 1,4-dioxane are added into a reaction vessel to react at 45-55 ℃; after the reaction is finished, the pH value is regulated to 8-9, and extraction and purification are carried out to obtain the compound D.
5. The method for producing a cationic lipid compound according to claim 4, wherein the formula for producing the compound C is as follows:
wherein, compound A, compound B and 1, 8-diazabicyclo (5.4.0) undec-7-ene DBU are added into a reaction vessel, tetrahydrofuran THF is then added, and the mixture is heated and stirred at the temperature of 70-80 ℃ for reaction; after the reaction is finished, extracting and purifying to obtain the compound C.
6. The method for preparing a cationic lipid compound according to claim 2, wherein the preparation equation of the compound J is as follows:
wherein, an intermediate and a compound I, K are added into a reaction vessel 2 CO 3 Adding tetrahydrofuran THF into the KI, heating and stirring at 70-80 ℃ to react; and (3) extracting and purifying after the reaction is finished to obtain the compound J.
7. The method for producing a cationic lipid compound according to claim 6, wherein the intermediate is produced by the following formula:
adding a compound H into a reaction vessel, dissolving in tetrahydrofuran THF, adding carbodiimide hydrochloride EDCI into the reaction vessel, stirring at room temperature, adding p-dimethylaminopyridine DMAP, stirring at room temperature again, adding a compound G into the reaction vessel, and stirring for reaction; after the reaction is finished, extracting and purifying are carried out to obtain an intermediate.
8. Use of the cationic lipid compound of claim 1 in the preparation of an mRNA delivery system.
9. An mRNA delivery system comprising the cationic lipid compound of claim 1.
10. The mRNA delivery system of claim 9, wherein the mRNA delivery system is an mRNA-LNP delivery system.
CN202311819318.8A 2023-12-27 2023-12-27 Cationic lipid compound, preparation method and application thereof and mRNA delivery system Active CN117466777B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311819318.8A CN117466777B (en) 2023-12-27 2023-12-27 Cationic lipid compound, preparation method and application thereof and mRNA delivery system

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311819318.8A CN117466777B (en) 2023-12-27 2023-12-27 Cationic lipid compound, preparation method and application thereof and mRNA delivery system

Publications (2)

Publication Number Publication Date
CN117466777A CN117466777A (en) 2024-01-30
CN117466777B true CN117466777B (en) 2024-03-19

Family

ID=89626070

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311819318.8A Active CN117466777B (en) 2023-12-27 2023-12-27 Cationic lipid compound, preparation method and application thereof and mRNA delivery system

Country Status (1)

Country Link
CN (1) CN117466777B (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113372226A (en) * 2021-06-08 2021-09-10 深圳市人民医院 Lipid molecule, lipid nanoparticle, preparation method and application thereof
WO2023098679A1 (en) * 2021-11-30 2023-06-08 石药集团巨石生物制药有限公司 Novel coronavirus mrna vaccine against mutant strains
CN116283624A (en) * 2022-03-25 2023-06-23 深圳市新合生物医疗科技有限公司 Cationic lipid compound, preparation method and application thereof and mRNA delivery system
CN116554046A (en) * 2023-04-17 2023-08-08 武汉滨会生物科技股份有限公司 Ionizable lipid compound and lipid nanoparticle thereof
WO2023182756A1 (en) * 2022-03-21 2023-09-28 주식회사 메디치바이오 Novel ionizable lipid and lipid nanoparticle composition using same
WO2023186167A1 (en) * 2022-04-02 2023-10-05 科镁信(上海)生物医药科技有限公司 Cationic lipid, liposome, lipid nanoparticle, and use

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113372226A (en) * 2021-06-08 2021-09-10 深圳市人民医院 Lipid molecule, lipid nanoparticle, preparation method and application thereof
WO2023098679A1 (en) * 2021-11-30 2023-06-08 石药集团巨石生物制药有限公司 Novel coronavirus mrna vaccine against mutant strains
WO2023182756A1 (en) * 2022-03-21 2023-09-28 주식회사 메디치바이오 Novel ionizable lipid and lipid nanoparticle composition using same
CN116283624A (en) * 2022-03-25 2023-06-23 深圳市新合生物医疗科技有限公司 Cationic lipid compound, preparation method and application thereof and mRNA delivery system
CN116354836A (en) * 2022-03-25 2023-06-30 深圳市新合生物医疗科技有限公司 Cationic lipid compound, preparation method and application thereof and mRNA delivery system
WO2023186167A1 (en) * 2022-04-02 2023-10-05 科镁信(上海)生物医药科技有限公司 Cationic lipid, liposome, lipid nanoparticle, and use
CN116554046A (en) * 2023-04-17 2023-08-08 武汉滨会生物科技股份有限公司 Ionizable lipid compound and lipid nanoparticle thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Min Li et al..《Adv. Mater.》.2021,第33卷(第34期),1-13. *

Also Published As

Publication number Publication date
CN117466777A (en) 2024-01-30

Similar Documents

Publication Publication Date Title
CN110337429B (en) Ionizable cationic lipids for RNA delivery
US9211343B2 (en) siRNA conjugate and preparation method thereof
JP6557722B2 (en) Biodegradable lipids for delivery of nucleic acids
WO2022166213A1 (en) Ionizable lipid molecule, preparation method therefor, and application thereof in preparation of lipid nanoparticle
CN102245559B (en) Aminoalcohol lipidoids and uses thereof
CN113993839B (en) Ionizable lipid molecule, preparation method thereof and application thereof in preparation of lipid nanoparticles
CN114507195B (en) Lipid compound, composition containing lipid compound and application of lipid compound
US10030243B2 (en) Nanoparticle type oligonucleotide structure having high efficiency and method for preparing same
WO2010080724A1 (en) Novel lipid nanoparticles and novel components for delivery of nucleic acids
CN114890907B (en) Cationic lipid compound and preparation method and application thereof
WO2010021865A1 (en) Novel lipid nanoparticles and novel components for delivery of nucleic acids
CN105916839B (en) cationic lipid
CA2969664A1 (en) Cationic lipid
CN114746401A (en) Ionizable lipids for nucleic acid delivery
CN116063205B (en) Lipid compound containing alkylated carbamate bond and application thereof
CN116354836A (en) Cationic lipid compound, preparation method and application thereof and mRNA delivery system
CN117466777B (en) Cationic lipid compound, preparation method and application thereof and mRNA delivery system
CN117466768B (en) Cationic lipid compound, preparation method and application thereof and mRNA delivery system
JP6774965B2 (en) Compounds as cationic lipids
CN115521220A (en) Long-chain alkyl ester amine compound, preparation method thereof and application thereof in nucleic acid delivery
CN117886711A (en) Cationic lipid compound, preparation method and application thereof, and LNP composition
JP7043411B2 (en) Compounds as cationic lipids
CN117229160B (en) Triester cationic lipid compounds, compositions containing same and uses thereof
Fenton Design, synthesis, and biological evaluation of diketopiperazine based ionizable lipids for the in vivo delivery of messenger RNA
CN116063245A (en) mRNA liposome nano particle with degradable center 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
GR01 Patent grant
GR01 Patent grant