CN112961091B - Amino lipide compound and preparation method and application thereof - Google Patents

Amino lipide compound and preparation method and application thereof Download PDF

Info

Publication number
CN112961091B
CN112961091B CN202110178738.7A CN202110178738A CN112961091B CN 112961091 B CN112961091 B CN 112961091B CN 202110178738 A CN202110178738 A CN 202110178738A CN 112961091 B CN112961091 B CN 112961091B
Authority
CN
China
Prior art keywords
compound
amino
alkyl
lipid
nucleic acid
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
CN202110178738.7A
Other languages
Chinese (zh)
Other versions
CN112961091A (en
Inventor
李林鲜
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 Xinxin Biotechnology Co ltd
Original Assignee
Shenzhen Xinxin Biotechnology 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 Shenzhen Xinxin Biotechnology Co ltd filed Critical Shenzhen Xinxin Biotechnology Co ltd
Priority to CN202110178738.7A priority Critical patent/CN112961091B/en
Publication of CN112961091A publication Critical patent/CN112961091A/en
Priority to US18/264,397 priority patent/US20240124396A1/en
Priority to PCT/CN2022/074272 priority patent/WO2022166747A1/en
Application granted granted Critical
Publication of CN112961091B publication Critical patent/CN112961091B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones
    • C07D207/2732-Pyrrolidones with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to other ring carbon atoms
    • C07D207/277Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • 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
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/68Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D211/72Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D211/78Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen

Abstract

The invention relates to an amino lipide compound, a compound shown in a chemical formula I, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt thereof,
Figure DDA0003609621530000011
also disclosed are methods for the preparation of the compounds and their use as components for the delivery of therapeutic agents and in the preparation of medicaments.

Description

Amino lipide compound and preparation method and application thereof
Technical Field
The invention relates to an amino lipid compound, in particular to an amino lipid compound which can be used for delivering genes into cells, and a preparation method and application thereof.
Background
Gene therapy is the artificial delivery of genes with specific genetic information to target cells, and the expressed target proteins have the effects of regulating, treating and even curing diseases caused by congenital or acquired gene defects. Both nucleic acid and cell membrane have negative charge, so that naked nucleic acid is difficult to directly introduce into cell, and is easily degraded by nucleic acid degrading enzyme in cytoplasm, and can not reach the action of gene introduction and gene therapy, so that it can implement gene transfer by means of external force or carrier. Genetic vectors are generally classified into viral vectors and non-viral vectors. The viral vector has extremely high transfection efficiency in vivo and in vitro, but has a plurality of defects such as high toxicity, strong immune response, small gene capacity, poor targeting property, complex preparation process and the like. Non-viral vectors have gained increasing attention and application due to their ease of preparation, transport, and storage, safety, efficacy, lack of immunogenicity, and the like.
However, gene delivery currently faces two problems during therapy relative to gene delivery at the cellular level. First, free RNA is susceptible to nuclease digestion in plasma. A frequently used solution is to introduce into the nanoparticles phosphonate ester loaded with PEG chains, which extend at the outermost layer of the micelle during self-assembly. Because the PEG layer has the characteristics of electric neutrality, protein adsorption resistance, no functional group at the end group and the like, the PEG layer can reduce the cytotoxicity of the nano carrier in vivo and prolong the cycle time. Second, after endocytosis of the cell, the gene vector can be transported into endosomal/lysosomal vesicles, where the gene is readily degraded by enzymes or acidic materials that are abundant in the lysosome. Therefore, the ability of nucleic acids to escape from endosomes/lysosomes into the cytoplasm is an important link for gene delivery in non-viral vectors. In the endosomal/lysosomal pathway, the nanocomplex undergoes a process of acidity reduction from pH 5-6 in late endosomes to lysosomes at pH about 4.5, which are rich in lysosomal enzymes and highly susceptible to degradation of the nanocomplex, and commonly used non-viral vectors have very low efficiency of endosomal/lysosomal escape and inefficient gene delivery.
Disclosure of Invention
Aiming at the existing defects, the invention provides an amino lipid compound for delivering genes into cells, and a preparation method and application thereof.
The technical scheme adopted by the invention for solving the technical problems is as follows: an aminolipidic compound, a compound of formula I, or a stereoisomer, or a tautomer, or a pharmaceutically acceptable salt thereof:
Figure GDA0003609621520000021
wherein:
R1-R2independently of each other, selected from the following structures: the R is1Is one of N6, N7, N8, N9, N10, N11, N12, N13, N14, N15, N16, N18, N19, N20 selected from:
N6:CH3(CH2)5-;N7:CH3(CH2)6-;N8:CH3(CH2)7-;
N9:CH3(CH2)8-;N10:CH3(CH2)9-;N11:CH3(CH2)10-;
N12:CH3(CH2)11-;N13:CH3(CH2)12-;N14:CH3(CH2)13-;
N15:CH3(CH2)14-;N16:CH3(CH2)15-;N18:CH3(CH2)17-;
N19:
Figure GDA0003609621520000022
N20:
Figure GDA0003609621520000023
the R is2Is one selected from a6, a7, A8, a9, a10, a11, a12, a13, a14, a15, a16, a18, a19, a 20:
A6:CH3(CH2)4-;A7:CH3(CH2)5-;A8:CH3(CH2)6-;
A9:CH3(CH2)7-;A10:CH3(CH2)8-;A11:CH3(CH2)9-;
A12:CH3(CH2)10-;A13:CH3(CH2)11-;A14:CH3(CH2)12-;
A15:CH3(CH2)13-;A16:CH3(CH2)14-;A18:CH3(CH2)16-;
A19
Figure GDA0003609621520000031
A20:
Figure GDA0003609621520000032
-X-L-N(R3)(R4) Is any one of O1, O2, O3, O4, O5, O6, O7, O8, O9, O10, D1, D2, D3, D4, D5, D6, D7, D8, D9, D10 selected from the following:
O1:
Figure GDA0003609621520000033
O2:
Figure GDA0003609621520000034
O3:
Figure GDA0003609621520000035
O4:
Figure GDA0003609621520000036
O5:
Figure GDA0003609621520000037
O6:
Figure GDA0003609621520000038
O7:
Figure GDA0003609621520000039
O8:
Figure GDA00036096215200000310
O9:
Figure GDA00036096215200000311
O10:
Figure GDA00036096215200000312
D1:
Figure GDA00036096215200000313
D2:
Figure GDA00036096215200000314
D3:
Figure GDA00036096215200000315
D4:
Figure GDA00036096215200000316
D5:
Figure GDA00036096215200000317
D6:
Figure GDA00036096215200000318
D7:
Figure GDA00036096215200000319
D8:
Figure GDA00036096215200000320
D9:
Figure GDA0003609621520000041
D10:
Figure GDA0003609621520000042
n is 1 or 2.
A method for preparing amino lipid compound comprises the following steps:
s1, reacting compound NH2-R1And R2-CHO reacting in solvent under stirring, distilling to remove solvent, adding cyclic acid anhydride, heating to react, purifying to obtain compound (II) with structural formula as follows,
Figure GDA0003609621520000043
s2, reacting the compound (II) with
Figure GDA0003609621520000044
The alcohol or amine is reacted in the presence of a condensing agent to obtain the compound (I), the structural formula is as follows,
Figure GDA0003609621520000045
wherein:
R1-R2independently of each other, selected from the following structures: the R is1Is one of N6, N12, N13, N16, N19, N20 selected from:
N6:CH3(CH2)5-;N12:CH3(CH2)11-;N13:CH3(CH2)12-;N16:CH3(CH2)15-;
N19:
Figure GDA0003609621520000046
N20:
Figure GDA0003609621520000047
the R is2Is one selected from a9, a10, a12, a13, a16, a18, a19, a 20:
A9:CH3(CH2)7-;A10:CH3(CH2)8-;A12:CH3(CH2)10-;A13:CH3(CH2)11-;
A16:CH3(CH2)14-;A18:CH3(CH2)16-;
A19:
Figure GDA0003609621520000051
A20:
Figure GDA0003609621520000052
-X-L-N(R3)(R4) Is any one of O1, O2, O5, O6, O8, O9, O10, D1, D4, D5 and D6 which are selected from the following components:
O1:
Figure GDA0003609621520000053
O2:
Figure GDA0003609621520000054
O5:
Figure GDA0003609621520000055
O6:
Figure GDA0003609621520000056
O8:
Figure GDA0003609621520000057
O9:
Figure GDA0003609621520000058
O10:
Figure GDA0003609621520000059
D1:
Figure GDA00036096215200000510
D4:
Figure GDA00036096215200000511
D5:
Figure GDA00036096215200000512
D6:
Figure GDA00036096215200000513
n is 1 or 2.
Use of an aminolipidic compound as a medicament for any of the nucleic acid transfer of any of the foregoing compounds, wherein when n ═ 1, X is not O as a medicament for nucleic acid transfer
Preferably, the nucleic acid is any one of RNA, mRNA, antisense oligonucleotide, DNA, plasmid, rRNA, miRNA, tRNA, siRNA, and snRNA.
"substituted" as referred to above is optional, i.e. one or more hydrogen atoms attached to the atom or group are independently unsubstituted or substituted with one or more substituents independently selected from: deuterium (D), halogen, -OH, mercapto, cyano, -CD3、C1-C6Alkyl (preferably C)1-C3Alkyl group), C2-C6An alkenyl group,C2-C6Alkynyl, cycloalkyl (preferably C)3-C8Cycloalkyl), aryl, heterocyclyl (preferably 3-8 membered heterocyclyl), heteroaryl, aryl C1-C6Alkyl-, heteroaryl C1-C6Alkyl radical, C1-C6Haloalkyl, -OC1-C6Alkyl (preferably-OC)1-C3Alkyl), -OC2-C6Alkenyl, OC1-C6Alkyl phenyl, C1-C6alkyl-OH (preferably C)1-C4alkyl-OH), C1-C6alkyl-SH, C1-C6alkyl-O-C1-C6Alkyl, OC1-C6Haloalkyl, NH2、C1-C6alkyl-NH2(preferably C)1-C3alkyl-NH2)、-N(C1-C6Alkyl radical)2(preferably-N (C)1-C3Alkyl radical)2)、-NH(C1-C6Alkyl) (preferably-NH (C)1-C3Alkyl)), -N (C)1-C6Alkyl) (C1-C6Alkylphenyl), -NH (C)1-C6Alkylphenyl), nitro, -C (O) -OH, -C (O) OC1-C6Alkyl (preferably-C (O) OC1-C3Alkyl), -CONRiri (Ri and Rii are H, D or C)1-C6Alkyl, preferably C1-C3Alkyl), -NHC (O) (C)1-C6Alkyl), -NHC (O) (phenyl), -N (C)1-C6Alkyl radical of C (O) (C)1-C6Alkyl), -N (C)1-C6Alkyl group C (O) (phenyl), -C (O) C1-C6Alkyl, -C (O) heteroaryl (preferably-C (O) -5-7 membered heteroaryl), -C (O) C1-C6Alkylphenyl, -C (O) C1-C6Haloalkyl, -OC (O) C1-C6Alkyl (preferably-OC (O) C)1-C3Alkyl), -S (O)2-C1-C6Alkyl, -S (O) -C1-C6Alkyl, -S (O)2-phenyl, -S (O)2-C1-C6Haloalkyl, -S (O)2NH2、-S(O)2NH(C1-C6Alkyl), -S (O)2NH (phenyl), -NHS (O)2(C1-C6Alkyl), -NHS (O)2(phenyl) and-NHS (O)2(C1-C6Haloalkyl), wherein each of the alkyl, cycloalkyl, phenyl, aryl, heterocyclyl, and heteroaryl is optionally further substituted with one or more substituents selected from the group consisting of: halogen, -OH, -NH2Cycloalkyl, 3-8 membered heterocyclyl, C1-C4Alkyl radical, C1-C4Haloalkyl-, -OC1-C4Alkyl, -C1-C4alkyl-OH, -C1-C4alkyl-O-C1-C4Alkyl, -OC1-C4Haloalkyl, cyano, nitro, -C (O) -OH, -C (O) OC1-C6Alkyl, -CON (C)1-C6Alkyl radical)2、-CONH(C1-C6Alkyl), -CONH2、-NHC(O)(C1-C6Alkyl), -NH (C)1-C6Alkyl radical C (O) (C)1-C6Alkyl), -SO2(C1-C6Alkyl), -SO2(phenyl), -SO2(C1-C6Haloalkyl), -SO2NH2、-SO2NH(C1-C6Alkyl), -SO2NH (phenyl), -NHSO2(C1-C6Alkyl), -NHSO2(phenyl) and-NHSO2(C1-C6Haloalkyl). In this case, when one atom or group is substituted with a plurality of substituents, the plurality of substituents may be the same or different.
Wherein "alkyl" refers to the residue of aliphatic hydrocarbon after losing one hydrogen atom, and includes straight-chain or branched-chain, saturated or unsaturated alkyl, alkyl includes alkyl, alkenyl and alkynyl,
wherein "acyl" refers to hydrocarbyl-carbonyl, preferably said acyl is C4-C24An acyl group.
Wherein "alkoxy" means alkyl-oxy, preferably said alkoxy is C1-C10An alkoxy group.
Wherein "heterocycle" refers to a saturated or unsaturated cyclic group containing a heteroatom selected from N, O, S, which heterocycle may be optionally substituted with one or more substituents.
The invention has the beneficial effects that: the compound of the invention is an aminolipidic compound containing long apolar residues, the resulting compound all having hydrophobic character and, due to the amino group, also hydrophilic character, this amphoteric character can be used to form lipid particles, at the same time it has a 5-oxopyrrolidine-or 6-oxopiperidine group, the introduction of which significantly increases the membrane fusion to enhance the release of mRNA, thus promoting a synergistic improvement of mRNA delivery, being stable during the in vivo circulation, being rapidly degraded in endosomes/lysosomes, with significantly enhanced delivery efficiency. The preparation method of the amino lipid compound has the advantages of easily available raw materials, mild reaction conditions, good reaction selectivity, high reaction yield, low requirements on instruments and equipment and simple operation, and can be used as a medicament to remarkably improve the gene delivery efficiency.
Drawings
FIG. 1 is a body fluid antibody titer resulting from the delivery of OVA mRNA by subcutaneous administration of a representative amino lipid compound of an embodiment of the invention;
Detailed Description
To more clearly illustrate the objects, technical solutions and advantages of the embodiments of the present invention, the present invention will be further described in detail with reference to the accompanying drawings and embodiments, and it is obvious that the described embodiments are some, but not all embodiments of the present invention. All other embodiments, which can be derived by a person skilled in the art from the embodiments of the present invention without inventive step, are within the scope of the present invention.
Example 1: 1-dodecyl-5-oxo-2-undecylpyrrolidine-3-carboxylic acid
Figure GDA0003609621520000081
N-dodecylamine (1.85g,10mmol), n-dodecanal (1.84g,100mmol) and anhydrous methanol (100 mL) are sequentially added into a 250mL reaction flask, stirred at room temperature for reaction for 12 hours, the solvent is evaporated under reduced pressure, then xylene (150 mL) and succinic anhydride (1.00g,100mmol) are sequentially added, and the temperature is raised to 140 ℃ for reaction for 10 hours. After the solvent was evaporated to dryness under reduced pressure, 50mL of n-hexane was added, stirred, crystallized, filtered, washed with a small amount of n-hexane, and dried to give 1-dodecyl-5-oxo-2-undecylpyrrolidine-3-carboxylic acid (3.75g, 83%).
Example 2: synthesis of 1-hexadecyl-2-nonyl-6-oxopiperidine-3-carboxylic acid
Figure GDA0003609621520000082
Into a 250mL reaction flask were added n-hexadecylamine (2.42g,10mmol), n-decaldehyde (1.56g,100mmol) and anhydrous methanol (100 mL) in this order, and the mixture was stirred at room temperature for 12 hours, after the solvent was evaporated to dryness under reduced pressure, 150mL of xylene and glutaric anhydride (1.14g,100mmol) were added in this order, and the mixture was heated to 140 ℃ for 10 hours. After the solvent was evaporated to dryness under reduced pressure, 50mL of n-hexane was added, stirred, crystallized, filtered, washed with a small amount of n-hexane, and dried to obtain 1-hexadecyl-2-nonyl-6-oxopiperidine-3-carboxylic acid (3.51g, 71%).
Example 3: synthesis of ((Z) -octadecyl-9-en-1-yl) -5-oxo-2-undecylpyrrolidine-3-carboxylic acid
Figure GDA0003609621520000083
Oleylamine (2.68g,10mmol), n-decanal (1.56g,100mmol) and anhydrous methanol (100 mL) were added sequentially to a 250mL reaction flask, stirred at room temperature for 12 hours, the solvent was evaporated under reduced pressure, then xylene (150 mL) and succinic anhydride (1.00g,100mmol) were added sequentially, and the temperature was raised to 140 ℃ for reaction for 10 hours. After the solvent was evaporated under reduced pressure, 50mL of n-hexane was added, stirred, crystallized, filtered, washed with a small amount of n-hexane, and dried to give ((Z) -octadecyl-9-en-1-yl) -5-oxo-2-undecylpyrrolidine-3-carboxylic acid (4.54g, 85%).
Example 4: synthesis of 1-dodecyl-2- ((8Z, 11Z) -heptadecyl-8, 11-dien-1-yl) -5-oxopyrrolidine-3-carboxylic acid
Figure GDA0003609621520000091
N-dodecylamine (1.85g,10mmol), cis-9, 12-octadecadienal (2.64g,100mmol) and anhydrous methanol (100 mL) are sequentially added into a 250mL reaction flask, stirred at room temperature for reaction for 12 hours, after the solvent is evaporated to dryness under reduced pressure, xylene (150 mL) and succinic anhydride (1.00g,100mmol) are sequentially added, and the temperature is raised to 140 ℃ for reaction for 10 hours. After the solvent was evaporated to dryness under reduced pressure, 50mL of n-hexane was added, stirred, crystallized, filtered, washed with a small amount of n-hexane, and dried to give 1-dodecyl-2- ((8Z, 11Z) -heptadecyl-8, 11-dien-1-yl) -5-oxopyrrolidine-3-carboxylic acid (4.31g, 81%).
Example 5: synthesis of Compound N12A12C4O2
Figure GDA0003609621520000092
1-dodecyl-5-oxo-2-undecylpyrrolidine-3-carboxylic acid (903mg,2mmol), 3-dimethylamino-1-propanol (310mg,3mol), and 50mL of dichloromethane were sequentially added to a 250mL reaction flask, and after stirring and dissolving, dicyclohexylcarbodiimide (824mg,4mmol), 4-dimethylaminopyridine (5mg,0.04mmol) were added, and the mixture was reacted at room temperature for 2 hours, washed with water for 3 times, dried over anhydrous sodium sulfate, concentrated, and purified using a flash column chromatography system (dichloromethane: methanol 20: 1 to 5: 1) to obtain compound N12A12C4O2(1.03g, 96%).1H NMR(400MHz,DMSO-d6):δ4.15(m,2H);3.94(m,1H);3.18(m,2H);2.90(m,1H),2.73(m,1H),2.62(m,1H),2.34(t,2H),2.16(s,6H),1.67(m,2H),1.39-1.18(m,40H),0.89(m,6H).ESI-MS calculated for C33H65N2O3 +[M+H]+537.5,found 537.7
Example 6: synthesis of Compound N16A10C5O10
Figure GDA0003609621520000101
1-hexadecyl-2-nonyl-6-oxypiperidine-3-carboxylic acid (988mg,2mmol), N-hydroxyethylpiperidine (387mg,3mol), and 50mL of methylene chloride were sequentially charged into a 250mL reaction flask, and after stirring and dissolution, dicyclohexylcarbodiimide (824mg,4mmol), 4-dimethylaminopyridine (5mg,0.04mmol) were further added and reacted at room temperature for 2 hours, washed with water for 3 times, dried over anhydrous sodium sulfate, and after concentration, purified using a flash column chromatography system (methylene chloride: methanol 20: 1 to 5: 1) to obtain the compound N16A10C5O10(1.03g, 96%).1H NMR(400MHz,DMSO-d6):δ4.15(m,2H);3.94(m,1H);3.18(m,2H);2.90(m,1H),2.73(m,1H),2.62(m,1H),2.34(t,2H),2.16(s,6H),1.67(m,2H),1.39-1.18(m,40H),0.89(m,6H).ESI-MS calculated for C33H65N2O3 +[M+H]+606.0,found 606.3
Example 7: synthesis of Compound N12A12C4D1
Figure GDA0003609621520000102
1-dodecyl-5-oxo-2-undecylpyrrolidine-3-carboxylic acid (903mg,2mmol), N, N-dimethylethylenediamine (353mg,4mol), and 50mL of dichloromethane were sequentially added to a 250mL reaction flask, and after stirring and dissolving, O- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU,1.14g,3mmol), and N, N-diisopropylethylamine (516mg,4mmol) were added, and after reaction at room temperature for 2 hours, after completion of TLC detection reaction, dichloromethane 200mL was added, washing was performed 3 times, drying was performed using anhydrous sodium sulfate, concentration was performed, and purification was performed using a flash column chromatography system (dichloromethane: methanol ═ 20: 1 to 5: 1) to obtain compound N12A12C4D1(982mg, 94%).1H NMR(400MHz,DMSO-d6):δ4.17(m,2H);3.95(m,1H);3.19(m,2H);2.91(m,1H),2.72(m,1H),2.63(m,1H),2.34(t,2H),2.16(s,6H),1.67(m,2H),1.39-1.18(m,38H),0.89(m,6H).ESI-MS calculated for C32H64N3O2 +[M+H]+522.5,found 522.9
Example 8: synthesis of Compound N12A20C4O10
Figure GDA0003609621520000111
1-dodecyl-5-oxo-2-undecylpyrrolidine-3-carboxylic acid (903mg,2mmol), N-hydroxyethylpiperidine (387mg,3mol), and 50mL of dichloromethane were sequentially added to a 250mL reaction flask, and after stirring and dissolving, dicyclohexylcarbodiimide (824mg,4mmol), 4-dimethylaminopyridine (5mg,0.04mmol) were added, and the mixture was reacted at room temperature for 2 hours, washed with water for 3 times, dried over anhydrous sodium sulfate, concentrated, and purified using a flash column chromatography system (dichloromethane: methanol: 20: 1 to 5: 1) to obtain N12A20C4O10(1.03g, 96%).1H NMR(400MHz,DMSO-d6):δ4.15(m,2H);3.94(m,1H);3.18(m,2H);2.90(m,1H),2.73(m,1H),2.62(m,1H),2.34(t,2H),2.16(s,6H),1.67(m,2H),1.39-1.18(m,40H),0.89(m,6H).ESI-MS calculated for C33H65N2O3 +[M+H]+644.1,found 644.3
Example 9: evaluation of luciferase mRNA in vivo delivery Performance of lipid nanoparticles prepared from amino lipid Compound
The preparation method comprises the following steps: the mol ratio of the amino lipidic compound to DSPC, cholesterol and PEG2000-DMG is 50: 10: 38.5: 1.5 in absolute ethanol. Luciferase mrna (fluc mrna) was dissolved in sodium acetate solution (50mM, pH 4.0). Two micro-syringe pumps were used, the ratio of ethanol solution to sodium acetate solution (50mM, pH 4.0) was controlled to 1: 3, preparing a crude solution of lipid nanoparticles in a micro-flow channel chip, dialyzing with a dialysis cartridge (Fisher, MWCO 20,000) at 1 XPBS and a controlled temperature of 4 ℃ for 6h, and filtering with a 0.22 μm microporous membrane before use. The mass ratio of aminolipid compound to luciferase mrna (fluc mrna) was about 10: 1.
animal preparation: selecting male BALB/c mice of 6 weeks old, weighing about 20g, and feeding in SPF-level feeding room, wherein animal experiments are strictly carried out according to the guidelines of the national health institution and the ethical requirements of animals.
In vivo delivery: 3 mice per group were randomly selected and intramuscular injected with lipid nanoparticles at a dose of 0.5 mg/kg. After 6 hours, 200. mu.L of 10mg/mL D-fluorescein potassium salt was injected into each mouse via the tail vein, and after 10 minutes, the mice were placed under an in vivo imaging system (IVIS-200, Xenogen), and the total fluorescence intensity of each mouse was observed and recorded by photographing. Table 1 shows that intramuscular administration of representative amino lipid compounds delivered the expression intensity of Fluc mRNA, and DLin-MC3 was used as a control, and a number of the amino lipids were similar to the expression intensity of MC3 and were significantly better than the positive control.
Figure GDA0003609621520000121
Figure GDA0003609621520000131
Figure GDA0003609621520000141
Figure GDA0003609621520000151
TABLE 1
Example 10: in vivo delivery of ovalbumin mRNA and evaluation of immunological properties of lipid nanoparticles prepared from amino lipid compounds
The preparation method comprises the following steps: the mol ratio of the amino lipidic compound to DSPC, cholesterol and PEG2000-DMG is 50: 10: 38.5: 1.5 in absolute ethanol. Ovalbumin mrna (ova mrna) was dissolved in sodium acetate solution (50mM, pH 4.0). Two micro-syringe pumps were used, the ratio of ethanol solution to sodium acetate solution (50mM, pH 4.0) was controlled to 1: 3, preparing a crude solution of lipid nanoparticles in a micro-flow channel chip, dialyzing with a dialysis cartridge (Fisher, MWCO 20,000) at 1 XPBS and a controlled temperature of 4 ℃ for 6h, and filtering with a 0.22 μm microporous membrane before use. The mass ratio of aminolipid compound to ovalbumin mrna (ova mrna) was about 10: 1.
animal preparation: selecting male BALB/c mice of 6 weeks old, weighing about 20g, and feeding in SPF-grade feeding room, wherein animal experiments are strictly carried out according to the guidelines of the national health institution and the requirements of animal ethics.
In vivo delivery: 3 mice were randomly selected per group and injected subcutaneously with lipid nanoparticles (Day 0) at a dose of 0.5 mg/kg. After 7 days, the same amount was used for another boost (Day 7). Tail vein bleeds were taken on day 21 for serological analysis.
Enzyme linked immunosorbent assay (ELISA): flat bottom 96 well plates (Nunc) were pre-coated in 50mM carbonate buffer at a concentration of 0.5 μ g protein per well (pH 9.6) overnight at 4 ℃, then blocked with 5% glycine, antiserum obtained from immunized animals were diluted from 102 to 106 PBS-0.05% Tween (PBS-T), pH 7.4, and added to wells and incubated at room temperature for 1 hour at 37 ℃, horseradish peroxidase (HRP) conjugated goat anti-mouse IgG in PBS-T-1% BSA at 1: a dilution of 10,000 was labeled. After addition of the HRP substrate, absorbance at 450nm was measured in an optical density ELISA plate reader (Bio-Rad) at one wavelength. As shown in fig. 1, N12a12C4O2 was comparable to the IgG antibody titration generated by MC3, while the IgG antibody titration of N16a10C5O10, N12a12C4D1, N12a20C4O10 was significantly better than the MC3 control.
Thus, the amino lipid compound can be placed in an aqueous solution to produce nano-sized materials, i.e., lipid nanoparticles, i.e., liposomes used to encapsulate drugs in lipid bilayers or in the internal aqueous space of liposomes, where liposomes are vesicles composed of bilayers of amphiphilic molecules that encapsulate the aqueous compartments, such as lipid bilayers vesicles (liposomes), multilamellar vesicles, or micelles, where the lipid is placed in water to first form lipid vesicles and then a bilayer or series of bilayers, each separated by water molecules, can be formed by lipid vesicles in water by ultrasound, where the lipid bilayer is a thin film formed by two layers of lipid molecules, where micelles are aggregates of surfactant molecules dispersed in liquid colloids, where typical micelles in aqueous solutions form aggregates with the hydrophilic head regions when exposed to water, a hydrophobic single tail region in the center of the chelating micelle; lipid particles formed from lipid compounds in gene therapy introduce foreign genes into target cells to correct or compensate for diseases caused by defective and abnormal genes for therapeutic purposes, such as treatment of cancer and genetic diseases; the cancer is one or more of lung cancer, gastric cancer, liver cancer, esophageal cancer, colon cancer, pancreatic cancer, brain cancer, lymph cancer, blood cancer or prostate cancer, and the genetic disease is one or more of hemophilia, thalassemia and gaucher's disease; in vaccination, amino-lipid compounds are used to deliver antigens or nucleic acids encoding antigens that elicit immune responses against various antigens that are used to treat and/or prevent a variety of conditions, such as cancer, allergy, toxicity, and infection by pathogens (e.g., viruses, bacteria, fungi, and other pathogenic organisms); the nucleic acid in the nucleic acid transfer is any one of RNA, mRNA (messenger RNA), antisense oligonucleotide, DNA, plasmid, rRNA (ribosomal RNA), miRNA (microrna), tRNA (transfer RNA), siRNA (small inhibitory RNA), snRNA (small nuclear RNA), and can be used in gene therapy, gene vaccination, antisense therapy, or therapy by interfering RNA in a patient. The nucleic acid has a biological effect when introduced into a cell or host as a biologically active agent, for example, by stimulating an immune or inflammatory response, by exerting an enzymatic activity or by supplementing mutations or the like, the biologically active agent being in particular a nucleic acid, a peptide, a protein, an antibody and a small molecule, or a member selected from the group consisting of antineoplastic agents, antibiotics, immunomodulators, anti-inflammatory agents, agents acting on the central nervous system, polypeptides or polypeptides (polypeptoids); of course, the bioactive agent can also be an anti-tumor agent, an antibiotic, an immunomodulator, an anti-inflammatory agent, an agent acting on the central nervous system, an antigen or fragment thereof, a protein, a peptide, a polypeptide, a vaccine, a small molecule, or a mixture thereof; furthermore, one or more of a helper lipid, a sterol, a polyethylene glycol lipid may be added to the application of the amino-lipidic compound, such as the helper lipid being a non-cationic lipid, the sterol being cholesterol, the polyethylene glycol lipid being PEG2000-DMG ((1- (monomethoxypolyethylene glycol) -2,3 dimyristoyl glycerol), the non-cationic lipid may contain cationic functional groups (e.g., ammonium groups) but should contain anionic functional groups to at least neutralize the molecule, the totality of all functional groups in the lipid molecule should be non-cationic, liposomes consisting of a mixture of cationic amino lipids and non-cationic (neutral) phospholipids being most effective in delivering nucleic acids into cells, e.g., the non-cationic lipid is DOPE (dioleoylphosphatidylethanolamine) or DSPC (distearoylphosphatidylcholine), is a natural component in cell membranes that can be used to stabilize particles and aid in integration with cell membranes; polyethylene glycol lipids (PEG lipids) help protect the particles and their contents from degradation in vitro or in vivo, PEG forms a protective layer on the liposome surface and increases circulation time in vivo, and can be used in liposomal drug delivery (PEG-liposomes) which can be used to transfect multicellular tissues or organs, providing a novel therapeutic treatment to patients, which can be any mammal, preferably from humans, mice, rats, pigs, cats, dogs, horses, goats, cattle and monkeys, and/or others.
It will be understood that modifications and variations can be made by persons skilled in the art in light of the above teachings and all such modifications and variations are intended to be included within the scope of the invention as defined in the appended claims.

Claims (8)

1. An amino lipidic compound, characterized in that: a compound of formula I, or a stereoisomer thereof, or a tautomer thereof, or a pharmaceutically acceptable salt thereof:
Figure FDA0003614190980000011
wherein:
R1-R2independently of each other, selected from the following structures: the R is1Is selected from the following N6, N7, N8,One of N9, N10, N11, N12, N13, N14, N15, N16, N18, N19, N20:
N6:CH3(CH2)5-;N7:CH3(CH2)6-;N8:CH3(CH2)7-;
N9:CH3(CH2)8-;N10:CH3(CH2)9-;N11:CH3(CH2)10-;
N12:CH3(CH2)11-;N13:CH3(CH2)12-;N14:CH3(CH2)13-;
N15:CH3(CH2)14-;N16:CH3(CH2)15-;N18:CH3(CH2)17-;
N19:
Figure FDA0003614190980000012
N20:
Figure FDA0003614190980000013
the R is2Is one selected from a6, a7, A8, a9, a10, a11, a12, a13, a14, a15, a16, a18, a19, a 20:
A6:CH3(CH2)4-;A7:CH3(CH2)5-;A8:CH3(CH2)6-;
A9:CH3(CH2)7-;A10:CH3(CH2)8-;A11:CH3(CH2)9-;
A12:CH3(CH2)10-;A13:CH3(CH2)11-;A14:CH3(CH2)12-;
A15:CH3(CH2)13-;A16:CH3(CH2)14-;A18:CH3(CH2)16-;
A19:
Figure FDA0003614190980000021
A20:
Figure FDA0003614190980000022
-X-L-N(R3)(R4) Is any one of O1, O2, O3, O4, O5, O6, O7, O8, O9, O10, D1, D2, D3, D4, D5, D6, D7, D8, D9, D10 selected from the following:
O1:
Figure FDA0003614190980000023
O2:
Figure FDA0003614190980000024
O3:
Figure FDA0003614190980000025
O4:
Figure FDA0003614190980000026
O5:
Figure FDA0003614190980000027
O6:
Figure FDA0003614190980000028
O7:
Figure FDA0003614190980000029
O8:
Figure FDA00036141909800000210
O9:
Figure FDA00036141909800000211
O10:
Figure FDA00036141909800000212
D1:
Figure FDA00036141909800000213
D2:
Figure FDA00036141909800000214
D3:
Figure FDA00036141909800000215
D4:
Figure FDA00036141909800000216
D5:
Figure FDA00036141909800000217
D6:
Figure FDA00036141909800000218
D7:
Figure FDA00036141909800000219
D8:
Figure FDA00036141909800000220
D9:
Figure FDA00036141909800000221
D10:
Figure FDA00036141909800000222
n is 1 or 2.
2. The amino lipidic compound of claim 1, wherein said amino lipidic compound is selected from the group consisting of:
Figure FDA0003614190980000031
Figure FDA0003614190980000041
Figure FDA0003614190980000051
3. a method for preparing an amino lipid compound, comprising: the method comprises the following steps:
s1, reacting compound NH2-R1And R2-CHO is stirred in solvent for reaction, then cyclic acid anhydride is added after the solvent is removed by distillation, the compound (II) is obtained after temperature rise reaction and purification, the structural formula is as follows,
Figure FDA0003614190980000052
s2, reacting the compound (II) with
Figure FDA0003614190980000053
The alcohol or amine is reacted in the presence of a condensing agent to prepare the compound (I), the structural formula is as follows,
Figure FDA0003614190980000054
wherein:
R1-R2independently of each other, selected from the following structures: the R is1Is one of N6, N12, N13, N16, N19, N20 selected from:
N6:CH3(CH2)5-;N12:CH3(CH2)11-;N13:CH3(CH2)12-;N16:CH3(CH2)15-;
N19:
Figure FDA0003614190980000061
N20:
Figure FDA0003614190980000062
the R is2Is one selected from a9, a10, a12, a13, a16, a18, a19, a 20:
A9:CH3(CH2)7-;A10:CH3(CH2)8-;A12:CH3(CH2)10-;A13:CH3(CH2)11-;A16:CH3(CH2)14-;A18:CH3(CH2)16-;
A19:
Figure FDA0003614190980000063
A20:
Figure FDA0003614190980000064
-X-L-N(R3)(R4) Is any one selected from the group consisting of O2, O5, O6, O8, O9, O10, D1, D4, D5, and D6:
O2:
Figure FDA0003614190980000065
O5:
Figure FDA0003614190980000066
O6:
Figure FDA0003614190980000067
O8:
Figure FDA0003614190980000068
O9:
Figure FDA0003614190980000069
O10:
Figure FDA00036141909800000610
D1:
Figure FDA00036141909800000611
D4:
Figure FDA00036141909800000612
D5:
Figure FDA00036141909800000613
D6:
Figure FDA00036141909800000614
n is 1 or 2.
4. Use of the aminolipidated compound of claims 1-2 in the manufacture of a medicament for gene therapy, gene vaccination, antisense therapy, interfering RNA or nucleic acid transfer, wherein when n ═ 1, X is not O in the manufacture of a medicament for nucleic acid transfer.
5. Use of an aminolipid compound according to claim 4, characterized in that: the nucleic acid is RNA or DNA.
6. Use of an aminolipid compound according to claim 4, characterized in that: the nucleic acid is an antisense oligonucleotide.
7. Use of the amino lipid compound according to claim 5, characterized in that: the RNA is any one of mRNA, rRNA, miRNA, tRNA, siRNA and snRNA.
8. Use of an amino lipid compound according to claim 5, characterized in that: the DNA is a plasmid.
CN202110178738.7A 2021-02-07 2021-02-07 Amino lipide compound and preparation method and application thereof Active CN112961091B (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN202110178738.7A CN112961091B (en) 2021-02-07 2021-02-07 Amino lipide compound and preparation method and application thereof
US18/264,397 US20240124396A1 (en) 2021-02-07 2022-01-27 Amino lipid compound, preparation method therefor, and use thereof
PCT/CN2022/074272 WO2022166747A1 (en) 2021-02-07 2022-01-27 Amino lipid compound, preparation method therefor, and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110178738.7A CN112961091B (en) 2021-02-07 2021-02-07 Amino lipide compound and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN112961091A CN112961091A (en) 2021-06-15
CN112961091B true CN112961091B (en) 2022-06-17

Family

ID=76284554

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110178738.7A Active CN112961091B (en) 2021-02-07 2021-02-07 Amino lipide compound and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN112961091B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20240124396A1 (en) * 2021-02-07 2024-04-18 Shenzhen Shenxin Biotechnology Co., Ltd. Amino lipid compound, preparation method therefor, and use thereof
CN115925604A (en) * 2021-09-22 2023-04-07 广州谷森制药有限公司 Pyrrolidine novel lipid compound, preparation method, composition and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011154457A1 (en) * 2010-06-09 2011-12-15 L'oreal 2-pyrrolidone derivatives functionalized with an ester, acid or amide radical, cosmetic composition comprising them and use thereof for conditioning keratin materials
WO2011154458A1 (en) * 2010-06-09 2011-12-15 L'oreal Composition comprising at least one 2-pyrrolidone functionalized with an ester or amide radical, and at least one pigment or direct dye, for dyeing keratin materials
WO2015125892A1 (en) * 2014-02-19 2015-08-27 国立大学法人京都大学 Protein labeling compound
CN106795096A (en) * 2014-06-25 2017-05-31 爱康泰生治疗公司 Novel lipid and lipid nanoparticle preparation for delivering nucleic acid
CN108368028A (en) * 2015-10-28 2018-08-03 爱康泰生治疗公司 Novel lipid and lipid nanoparticle preparation for delivering nucleic acid

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9120744B2 (en) * 2007-04-16 2015-09-01 The Regents Of The University Of Michigan Plasminogen activator inhibitor-1 inhibitors and methods of use thereof to modulate lipid metabolism
US8642660B2 (en) * 2007-12-21 2014-02-04 The University Of Rochester Method for altering the lifespan of eukaryotic organisms
US10968176B2 (en) * 2014-09-14 2021-04-06 Nanosynthons Llc Pyrrolidone derivatives, oligomers and polymers

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011154457A1 (en) * 2010-06-09 2011-12-15 L'oreal 2-pyrrolidone derivatives functionalized with an ester, acid or amide radical, cosmetic composition comprising them and use thereof for conditioning keratin materials
WO2011154458A1 (en) * 2010-06-09 2011-12-15 L'oreal Composition comprising at least one 2-pyrrolidone functionalized with an ester or amide radical, and at least one pigment or direct dye, for dyeing keratin materials
WO2015125892A1 (en) * 2014-02-19 2015-08-27 国立大学法人京都大学 Protein labeling compound
CN106795096A (en) * 2014-06-25 2017-05-31 爱康泰生治疗公司 Novel lipid and lipid nanoparticle preparation for delivering nucleic acid
CN108368028A (en) * 2015-10-28 2018-08-03 爱康泰生治疗公司 Novel lipid and lipid nanoparticle preparation for delivering nucleic acid

Also Published As

Publication number Publication date
CN112961091A (en) 2021-06-15

Similar Documents

Publication Publication Date Title
US8192753B2 (en) pH-sensitive cationic lipids, and liposomes and nanocapsules containing the same
CN112891560B (en) mRNA delivery carrier and preparation method and application thereof
CN114073677B (en) Lipid nanoparticle
US20060084617A1 (en) Methods for delivery of nucleic acids
CN112961091B (en) Amino lipide compound and preparation method and application thereof
CN107406396B (en) Cationic lipid
US20230149562A1 (en) Carriers for efficient nucleic acid delivery
WO2023029928A1 (en) Amino lipid and application thereof
WO2023236976A1 (en) Lipid compound and preparation method therefor, and use thereof
JP7411824B2 (en) Aminolipid compounds, their preparation methods, and their uses
CN116574070A (en) Multi-tail type ionizable lipid, and preparation method and application thereof
WO2022166747A1 (en) Amino lipid compound, preparation method therefor, and use thereof
CA3214023A1 (en) Dendritic architectures as nonviral vectors in gene delivery
WO2021193397A1 (en) Novel cationic lipid having cystine skeleton
CN113968968A (en) Amino lipid compound, preparation method and application thereof
CN113403313B (en) sgRNA, plasmid and nano-composite for specifically recognizing human PLK1 locus and application
CN117915958A (en) Dendritic structures as non-viral vectors in gene delivery

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
CB02 Change of applicant information
CB02 Change of applicant information

Address after: 518000 floor 6, block B, Tsinghua information port, No. 1, songpingshan Xindong Road, songpingshan community, Xili street, Nanshan District, Shenzhen, Guangdong

Applicant after: Shenzhen Xinxin Biotechnology Co.,Ltd.

Address before: 518000 b515, building 7, Shenzhen Bay science and technology ecological park, 1819 Shahe West Road, high tech community, Yuehai street, Nanshan District, Shenzhen City, Guangdong Province

Applicant before: Shenzhen Xinxin Biotechnology Co.,Ltd.

GR01 Patent grant
GR01 Patent grant