CN118290282A - Ionizable lipid compound, preparation method and application thereof - Google Patents

Ionizable lipid compound, preparation method and application thereof Download PDF

Info

Publication number
CN118290282A
CN118290282A CN202410324191.0A CN202410324191A CN118290282A CN 118290282 A CN118290282 A CN 118290282A CN 202410324191 A CN202410324191 A CN 202410324191A CN 118290282 A CN118290282 A CN 118290282A
Authority
CN
China
Prior art keywords
compound
pharmaceutically acceptable
lipid
tautomer
stereoisomer
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.)
Pending
Application number
CN202410324191.0A
Other languages
Chinese (zh)
Inventor
张�林
刘安东
杨柳
江天
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 Yetai Pharmaceutical Technology Co ltd
Beijing Yitai Pharmaceutical Technology Co ltd
Original Assignee
Hangzhou Yetai Pharmaceutical Technology Co ltd
Beijing Yitai Pharmaceutical 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 Yetai Pharmaceutical Technology Co ltd, Beijing Yitai Pharmaceutical Technology Co ltd filed Critical Hangzhou Yetai Pharmaceutical Technology Co ltd
Priority to CN202410324191.0A priority Critical patent/CN118290282A/en
Publication of CN118290282A publication Critical patent/CN118290282A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The specification provides compounds of formula (I), or pharmaceutically acceptable salts, isotopic variations, tautomers or stereoisomers thereof, wherein each group is defined in the specification. Also provided are lipid nanoparticles and pharmaceutical compositions comprising the compounds, and the use of the compounds, lipid nanoparticles, and compositions in delivery of nucleic acids, the compounds being useful for delivery of a variety of bioactive substances, and having the characteristics of high encapsulation efficiency and high delivery efficiency.

Description

Ionizable lipid compound, preparation method and application thereof
Technical Field
The invention belongs to the field of biological medicine, and in particular relates to an ionizable lipid compound, a preparation method and application thereof.
Background
The nucleic acid vaccine is prepared through introducing exogenous gene (DNA or RNA) encoding specific antigen protein directly into host cell, expressing the antigen protein in the host cell and inducing the host to produce immune response to the antigen protein for the purpose of preventing and treating diseases.
Moderna and BioNTech use Lipid Nanoparticles (LNP) as delivery systems, the LNP major components including Cationic Lipid molecules (Cationic Lipid), cholesterol, neutral lipids and polyethylene glycol conjugated lipids. The cationic lipid molecule is the core of the LNP delivery system, and the molecular structure plays a decisive role in the aspects of the delivery efficiency, targeting, preparation stability and the like of the whole liposome nano-particle.
Since specific delivery to achieve different kinds of nucleic acid substance delivery and different targets has different requirements for the delivery system, further development of new lipid molecules is required in order to meet different demands of gene therapy.
Disclosure of Invention
The invention aims to provide an ionizable lipid compound with a novel structure and application thereof, wherein the compound can be used for delivering various bioactive substances, has the characteristics of high encapsulation efficiency, high delivery efficiency and good biocompatibility, and provides more choices for delivering disease treatment or preventive agents.
In a first aspect of the invention, there is provided a compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer or stereoisomer thereof:
Wherein,
Each R 1、R2、R3 is independently selected from hydrogen or C 1-8 alkyl;
L 1 is selected from C 1-10 alkylene, C 2-10 alkenylene, or C 2-10 alkynylene;
G is phenylene;
Each R 4、R5 is independently selected from C 1-30 alkyl, C 2-30 alkenyl, or C 2-30 alkynyl.
In one embodiment, G is selected from
In one embodiment, the compound of formula (I) has the structure of formula (II):
Wherein R 1、R2、R3、R4、R5、L1 is as defined above.
In one embodiment, the total length of R 1, the total length of R 2, and the total length of R 3 are each independently 1,2, 3,4,5, 6, 7, or 8 carbon atoms.
In one embodiment, each R 1、R2、R3 is independently selected from hydrogen, methyl, ethyl, or propyl.
In one embodiment, R 1 is hydrogen; in another embodiment, R 1 is C 1 alkyl; in another embodiment, R 1 is C 2 alkyl; in another embodiment, R 1 is C 3 alkyl; in another embodiment, R 1 is C 4 alkyl; in another embodiment, R 1 is C 5 alkyl; in another embodiment, R 1 is C 6 alkyl; in another embodiment, R 1 is C 7 alkyl; in another embodiment, R 1 is C 8 alkyl.
In one embodiment, R 2 is hydrogen; in another embodiment, R 2 is C 1 alkyl; in another embodiment, R 2 is C 2 alkyl; in another embodiment, R 2 is C 3 alkyl; in another embodiment, R 2 is C 4 alkyl; in another embodiment, R 2 is C 5 alkyl; in another embodiment, R 2 is C 6 alkyl; in another embodiment, R 2 is C 7 alkyl; in another embodiment, R 2 is C 8 alkyl.
In one embodiment, R 3 is hydrogen; in another embodiment, R 3 is C 1 alkyl; in another embodiment, R 3 is C 2 alkyl; in another embodiment, R 3 is C 3 alkyl; in another embodiment, R 3 is C 4 alkyl; in another embodiment, R 3 is C 5 alkyl; in another embodiment, R 3 is C 6 alkyl; in another embodiment, R 3 is C 7 alkyl; in another embodiment, R 3 is C 8 alkyl.
In one embodiment, L 1 is selected from C 1-10 alkylene; in another embodiment, L 1 is selected from C 2-10 alkenylene; in another embodiment, L 1 is selected from C 2-10 alkynylene.
In one embodiment, the total length of L 1 is 1,2, 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms.
In one embodiment, L 1 is selected from C 1-6 alkylene; in another embodiment, L 1 is selected from C 1-4 alkylene; in another embodiment, L 1 is selected from C 1-2 alkylene.
In one embodiment, L 1 is selected from-CH 2-、-(CH2)2-、-(CH2)3 -or- (CH 2)4 -.
In one embodiment, L 1 is-CH 2 -; in another embodiment, L 1 is- (CH 2)2 -; in another embodiment, L 1 is- (CH 2)3 -, and in another embodiment, L 1 is- (CH 2)4 -.
In one embodiment, G is
In one embodiment, R 4 is selected from C 1-30 alkyl, C 2-30 alkenyl, or C 2-30 alkynyl.
In one embodiment, the total length of R 4 is 1,2, 3,4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 carbon atoms.
In one embodiment, R 4 is selected from- (CH 2)m1CH=CH(CH2)n1CH=CH(CH2)m2CH3;
m1 and m2 are each independently selected from 2, 3,4, 5, 6, 7, 8, 9, 10 or 11;
n1 is selected from 0,1,2 or 3.
In one embodiment, R 4 is selected from- (CH 2)7CH=CHCH2CH=CH(CH2)4CH3; in another embodiment, R 4 is selected from- (CH 2)8CH=CHCH2CH=CH(CH2)4CH3), in another embodiment R 4 is selected from- (CH 2)9CH=CHCH2CH=CH(CH2)4CH3), in another embodiment R 4 is selected from- (CH 2)7CH=CH(CH2)2CH=CH(CH2)4CH3), in another embodiment R 4 is selected from- (CH 2)8CH=CH(CH2)2CH=CH(CH2)4CH3), and in another embodiment R 4 is selected from- (CH 2)9CH=CH(CH2)2CH=CH(CH2)4CH3.
In one embodiment, R 5 is selected from C 1-30 alkyl, C 2-30 alkenyl, or C 2-30 alkynyl.
In one embodiment, the total length of R 5 is 1,2, 3,4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 carbon atoms.
In one embodiment, R 5 is selected from- (CH 2)m1CH=CH(CH2)n1CH=CH(CH2)m2CH3;
m1 and m2 are each independently selected from 6, 7, 8, 9, 10 or 11;
n1 is selected from 0,1,2 or 3.
In one embodiment, R 5 is selected from- (CH 2)7CH=CHCH2CH=CH(CH2)4CH3; in another embodiment, R 4 is selected from- (CH 2)8CH=CHCH2CH=CH(CH2)4CH3), in another embodiment R 5 is selected from- (CH 2)9CH=CHCH2CH=CH(CH2)4CH3), in another embodiment R 5 is selected from- (CH 2)7CH=CH(CH2)2CH=CH(CH2)4CH3), in another embodiment R 5 is selected from- (CH 2)8CH=CH(CH2)2CH=CH(CH2)4CH3), and in another embodiment R 5 is selected from- (CH 2)9CH=CH(CH2)2CH=CH(CH2)4CH3.
In a first aspect of the invention, the following compounds are provided:
in a second aspect of the invention, there is provided a process for the preparation of a compound of formula (II) above, or a pharmaceutically acceptable salt, isotopic variant, tautomer or stereoisomer thereof, comprising:
reacting a compound of formula (IIa) with a compound of formula (IIb) to obtain a compound of formula (II);
Wherein X 1 is selected from halogen; r 1、R2、R3、R4、R5、L1 are each as defined above.
In a third aspect of the present invention, there is provided a process for the preparation of compound 1, comprising:
Reacting linolenic alcohol (compound 1-1) with carbon tetrabromide in the presence of triphenylphosphine to obtain compound 1-2;
reacting p-toluenesulfonyl methyl isonitrile (TosMIC) with compound 1-2 to obtain compound 1-3; further, the reaction is carried out under strongly basic conditions; still further, the reaction is carried out in the presence of sodium hydride;
Hydrolyzing the compound 1-3 to obtain a compound 1-4; further, hydrolysis is performed under acidic conditions; further, hydrolysis is performed in the presence of hydrochloric acid;
Reducing the compound 1-4 to obtain a compound 1-5; further, reducing the compound 1-4 by sodium borohydride to obtain a compound 1-5;
Reacting the compound 1-5 with 4-fluorobenzoic acid to obtain a compound 1-6; further, the compound 1-5, 4-fluorobenzoic acid is reacted in the presence of EDCI and DMAP (4-dimethylaminopyridine) to obtain the compound 1-6;
Reacting the compound 1-6 with N, N-dimethyl ethylenediamine to obtain a compound 1; further, reacting the compounds 1-6 with N, N-dimethylethylenediamine in the presence of DIEA to obtain a compound shown in a formula 1;
in one embodiment, the method for preparing compound 1 comprises:
s1: reacting linolenic alcohol with carbon tetrabromide in the presence of triphenylphosphine to obtain a compound 1-2;
s2: reacting p-toluenesulfonyl methyl isonitrile with a compound 1-2 under a strong alkaline condition to obtain a compound 1-3;
s3: hydrolyzing the compound 1-3 under an acidic condition to obtain a compound 1-4;
S4: reducing the compound 1-4 by NaBH 4 to obtain a compound 1-5;
S5: reacting the compounds 1-5 and 4-fluorobenzoic acid in the presence of EDCI and DMAP to obtain compounds 1-6;
S6: reacting the compounds 1-6 with N, N-dimethylethylenediamine in the presence of DIEA to obtain the compound 1.
In a fourth aspect of the invention, there is provided a pharmaceutical composition comprising a compound as described in the preceding first aspect, or a pharmaceutically acceptable salt, isotopic variant, tautomer or stereoisomer thereof, and a pharmaceutically acceptable adjuvant.
In a fifth aspect of the invention, there is provided a lipid nanoparticle comprising a lipid component and optionally an active ingredient;
the lipid component comprises: ionizable cationic lipids, structural lipids, neutral lipids, and polymeric lipids;
Wherein the ionizable cationic lipid is a compound of the first aspect described above, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof.
In one embodiment, the neutral lipid is selected from one or more of DSPC, DMPC, DOPC, DPPC, POPC, DOPE, DMPE, POPE or DPPE.
In one embodiment, the structural lipid is selected from one or more of cholesterol, sitosterol, stigmasterol, rock-soap sterol, brassicasterol, ergosterol, lycorine, ursolic acid, alpha-tocopherol, stigmasterol, aveosterol, ergocalcitol, or campesterol.
In one embodiment, the polymeric lipid is a pegylated lipid.
In one embodiment, the pegylated lipid is selected from one or more of PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramide, PEG-modified dialkylamine, PEG-modified diacylglycerol, PEG-modified dialkylglycerol.
In one embodiment, the pegylated lipid is selected from one or more of DMPE-PEG1000、DPPE-PEG1000、DSPE-PEG1000、DOPE-PEG1000、DMG-PEG2000、Ceramide-PEG2000、DMPE-PEG2000、DPPE-PEG2000、DSPE-PEG2000、Azido-PEG2000、DSPE-PEG2000-Mannose、Ceramide-PEG5000、DSPE-PEG5000.
In a sixth aspect of the present invention, there is provided a method for preparing lipid nanoparticles, comprising:
(1) Sequentially dissolving and mixing an ionizable lipid compound, cholesterol, phospholipid and polyethylene glycol lipid (in mol percent) in ethanol according to 50%, 38.5%, 10% and 1.5%, respectively;
(2) The mRNA active ingredient was dissolved in 25mM sodium acetate solution (ph=4.5);
(3) Mixing the organic phase dissolved with the lipid mixture and the aqueous phase dissolved with the mRNA component by using an automatic high-throughput microfluidic system according to a flow rate ratio ranging from 1:1 to 1:4, wherein the mixing speed is 10-18mL/min;
(4) The lipid nanoparticles (N/P6) were diluted with phosphate buffer solution and then the nanoparticle solution was ultrafiltered to the original preparation volume using an ultrafiltration tube (available from Millipore) with a molecular weight cut-off of 30 kDa;
(5) The obtained nano particles are filtered and sterilized by a sterile filter membrane with the diameter of 0.2 mu m and then stored in a sealed glass bottle at low temperature.
In a seventh aspect of the present invention, there is provided the use of a compound according to the first aspect, or a pharmaceutically acceptable salt, isotopic variant, tautomer or stereoisomer thereof, or a pharmaceutical composition according to the fourth aspect, or a lipid nanoparticle according to the fifth aspect, for the manufacture of a medicament for the treatment, diagnosis or prophylaxis of a disease.
In an eighth aspect of the invention there is provided the use of a compound according to the first aspect of the invention, or a pharmaceutically acceptable salt, isotopic variant, tautomer or stereoisomer thereof, or a pharmaceutical composition according to the fourth aspect, or a lipid nanoparticle according to the fifth aspect, for the manufacture of a medicament for the delivery of a load selected from one or more of a therapeutic agent, a prophylactic agent or a diagnostic agent.
In a ninth aspect of the invention there is provided a method of treating, diagnosing or preventing a disease in a subject comprising administering to the subject an effective amount of a pharmaceutical composition according to the fourth aspect of the foregoing, or a lipid nanoparticle according to the fifth aspect.
In one embodiment, the therapeutic, prophylactic or diagnostic agent is selected from one or more of a nucleic acid, a small molecule compound, a polypeptide or a protein.
In one embodiment, the nucleic acid is selected from one or more of RNA or DNA.
In one embodiment, the RNA is selected from one or more of small interfering RNA (siRNA), short hairpin RNA (shRNA), antisense RNA (aRNA), messenger RNA (mRNA), modified messenger RNA (mmRNA), long non-coding RNA (lncRNA), microrna (miRNA), small activating RNA (saRNA), poly-coding nucleic acid (MCNA), poly-coding nucleic acid (PCNA), guide RNA (gRNA), CRISPRRNA (CRRNA), or ribozyme.
In one embodiment, the RNA is mRNA.
In one embodiment, the RNA is a modified mRNA.
Detailed Description
I. Definition of the definition
In the present invention, unless otherwise indicated, scientific and technical terms used herein have the meanings commonly understood by one of ordinary skill in the art. Also, the relative terms and laboratory procedures used herein are terms and conventional procedures that are widely used in the corresponding arts. Meanwhile, in order to better understand the present invention, definitions and explanations of related terms are provided below.
As used herein and unless otherwise indicated, the terms "comprising," "including," "having," "containing," and their grammatical equivalents are generally understood to be open-ended and not to be limiting, e.g., not to exclude other, unrecited elements or steps.
Compounds are named herein using standard nomenclature. Compounds having asymmetric centers, it is to be understood (unless otherwise indicated) that all optical isomers and mixtures thereof are encompassed. Furthermore, unless otherwise specified, all isomeric compounds encompassed by the present invention may occur with carbon-carbon double bonds in the form of Z and E. Compounds that exist in different tautomeric forms, one of the compounds is not limited to any particular tautomer, but is intended to encompass all tautomeric forms.
The compounds of the invention may include one or more asymmetric centers and thus may exist in a variety of stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms. For example, the compounds of the invention may be individual enantiomers, diastereomers, or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomers. The isomers may be separated from the mixtures by methods known to those skilled in the art, including: chiral High Pressure Liquid Chromatography (HPLC), formation and crystallization of chiral salts; alternatively, preferred isomers may be prepared by asymmetric synthesis.
The compounds of the present invention may exist in tautomeric forms. Tautomers are functional group isomers that result from the rapid movement of an atom in a molecule at two positions. Tautomers are a particular functional group isomer, and a pair of tautomers can be converted to each other, but usually take a relatively stable one as its predominant form. The most prominent examples are enol and keto tautomers.
The invention also includes isotopically-labeled compounds (isotopically-variant) which are identical to those recited in formula (I), formula (IA) or formula (IB), but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2H、3H、13C、11C、14C、15N、18O、17O、31P、32P、35S、18F and 36 Cl, respectively. The compounds of the invention, prodrugs thereof, and pharmaceutically acceptable salts of the compounds or prodrugs thereof, which contain the isotopes described above and/or other isotopes of other atoms, are within the scope of this invention. Certain isotopically-labeled compounds of the present invention, for example those into which radioactive isotopes (e.g., 3 H and 14 C) are introduced, are useful in drug and/or substrate tissue distribution assays. Tritium, i.e., 3 H, and carbon-14, i.e., 14 C isotopes are particularly preferred because they are easy to prepare and detect. Further, substitution with heavier isotopes, such as deuterium, i.e., 2 H, may be preferred in some circumstances because greater metabolic stability may afford therapeutic benefits such as increased in vivo half-life or reduced dosage requirements. Isotopically-labelled compounds of formula (I), formula (IA) or formula (IB) of the present invention and prodrugs thereof are typically obtained by substituting a readily available isotopically-labelled reagent for a non-isotopically-labelled reagent during the course of the preparation of the compound.
The term "substituted" or "substituted" means that any one or more hydrogen atoms on a particular atom is replaced with a substituent.
When numerical ranges are listed, it is intended to include each and every value and subrange within the range.
The term "alkyl" refers to a straight or branched saturated hydrocarbon moiety. In one embodiment, the alkyl group is a straight chain saturated hydrocarbon. "C 1-30 alkyl" refers to a straight or branched saturated hydrocarbon group having 1 to 30 carbon atoms. The term "C 1-30 alkyl" also includes heteroalkyl groups in which one or more (e.g., 1,2,3, or 4) carbon atoms are replaced with a heteroatom (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus). The alkyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
The term "alkenyl" refers to a straight or branched hydrocarbon moiety having one or more carbon-carbon double bonds. "C 2-30 alkenyl" refers to a straight or branched hydrocarbon group having 2 to 30 carbon atoms and at least one carbon-carbon double bond. In some embodiments, C 10-20 alkenyl, C 15-20 alkenyl, C 17-20 alkenyl, C 18 alkenyl, C 19 alkenyl, C 20 alkenyl are preferred. The term "C 2-30 alkenyl" also includes heteroalkenyl groups in which one or more (e.g., 1,2, 3, or 4) carbon atoms are replaced with heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus). The alkenyl group may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
The term "alkynyl" refers to a straight or branched hydrocarbon moiety having one or more carbon-carbon triple bonds. "C 2-30 alkynyl" refers to a straight or branched hydrocarbon group having 2 to 30 carbon atoms, at least one carbon-carbon triple bond, and optionally one or more carbon-carbon double bonds. In some embodiments, C 10-20 alkenyl, alkenyl C 15-20 alkynyl, C 17-20 alkynyl, C 18 alkynyl, C 19 alkynyl, C 20 alkynyl are preferred. The term "C 2-30 alkynyl" also includes heteroalkynyl groups in which one or more (e.g., 1,2,3, or 4) carbon atoms are replaced with heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus). Alkynyl groups may be optionally substituted with one or more substituents, for example, with 1 to 5 substituents, 1 to 3 substituents, or 1 substituent.
The term "alkylene" refers to a divalent group formed by removing another hydrogen of an alkyl group, which may be substituted or unsubstituted. "C 1-10 alkylene" refers to a divalent group formed by removing another hydrogen of a C 1-10 alkyl group, and may be substituted or unsubstituted. In some embodiments, methylene (-CH 2 -), ethylene (-CH 2CH2 -), propylene (-CH 2CH2CH2 -), butylene (-CH 2CH2CH2CH2 -) are preferred.
The term "alkenylene" refers to a divalent group formed by removal of another hydrogen of an alkenyl group, which may be substituted or unsubstituted. "C 2-10 alkenylene" refers to a divalent group formed by removal of another hydrogen of a C 2-10 alkenyl group, and may be substituted or unsubstituted.
The term "alkynylene" refers to a divalent group formed by the removal of another hydrogen of an alkynyl group, which may be substituted or unsubstituted. "C 2-10 alkynylene" refers to a divalent group formed by removal of another hydrogen of a C 2-10 alkynyl group, and may be substituted or unsubstituted.
The term "the total length of the variables a and B is x carbon atoms" means that the number of carbon atoms in the backbone of the group represented by the variable a and the number of carbon atoms in the backbone of the group represented by the variable B are x.
"Optionally substituted" means that the substituent may be designated, or may be unsubstituted.
"Nucleic acid" refers to a single-or double-stranded deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) molecule, and hybrid molecules thereof. Examples of nucleic acid molecules include, but are not limited to, messenger RNAs (mrnas), micrornas (mirnas), small interfering RNAs (sirnas), self-amplifying RNAs (sarnas), antisense oligonucleotides (ASOs), and the like. The nucleic acid may be further chemically modified, the chemical modification being selected from one of pseudouridine, N1-methyl-pseudouridine, 5-methoxyuridine, 5-methylcytosine, or a combination thereof. The mRNA molecules contain protein coding regions and may further contain expression control sequences, typical expression control sequences include, but are not limited to, 5 'caps (5' caps), 5 'untranslated regions (5' utrs), 3 'untranslated regions (3' utrs), polyadenylation sequences (polyas), miRNA binding sites.
"Cationic lipid" refers to a lipid molecule capable of being positively charged under physiological pH conditions.
"Neutral lipids" refers to lipid molecules that are uncharged at specific pH conditions, such as physiological pH conditions. Examples of neutral lipids include, but are not limited to, 1, 2-distearoyl-sn-glycero-3-phosphorylcholine (DSPC), 1, 2-dimyristoyl-sn-glycero-3-phosphorylcholine (DMPC), 1, 2-dioleoyl-sn-glycero-3-phosphorylcholine (DOPC), 1, 2-dipalmitoyl-sn-glycero-3-phosphorylcholine (DPPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphorylcholine (POPC), 1, 2-dioleoyl-sn-glycero-3-phosphorylethanolamine (DOPE), 1, 2-dimyristoyl-sn-glycero-3-phosphorylethanolamine (DMPE), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphorylethanolamine (DPPE), 1, 2-dipalmitoyl-sn-glycero-3-phosphorylethanolamine (DPPE).
"Structural lipids" refers to lipids, such as steroids, that are commonly used to enhance nanoparticle stability by filling in the interstices between the lipids. The steroid is a compound having a cyclopenta-polyhydrophenanthrene carbon skeleton, and in a preferred embodiment, the steroid is selected from cholesterol, sitosterol, stigmasterol, soapsterol, brassicasterol, ergosterol, lycorine, ursolic acid, alpha-tocopherol, stigmasterol, oat sterol, ergocalcitol, or campesterol.
"Polymer lipid" refers to a molecule that contains a polymer moiety and a lipid moiety. In some embodiments, the polymer lipid is a polyethylene glycol (PEG) lipid. Other lipids capable of reducing aggregation, such as products of coupling compounds having uncharged, hydrophilic, steric-blocking moieties to lipids, may also be used.
"Lipid nanoparticle" refers to particles having a nanoscale size that contain a lipid component.
The term "pharmaceutically acceptable" is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salt" refers to pharmaceutically acceptable acid addition salts or base addition salts, including salts of compounds with inorganic or organic acids, and salts of compounds with inorganic or organic bases.
The term "pharmaceutically acceptable excipients" refers to those excipients which do not significantly stimulate the organism and which do not impair the biological activity and properties of the active compound. Suitable adjuvants are well known to those skilled in the art. Pharmaceutically acceptable excipients for use in the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the co-formulated compounds. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of the invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (e.g., human serum albumin), buffer substances (e.g., phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (e.g., protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, and lanolin.
Typically, physiological saline may be used as a pharmaceutically acceptable carrier. Other suitable carriers include, for example, water, buffered water, 0.9% saline, 0.3% glycine, and the like, including glycoproteins for improved stability, e.g., albumin, lipoproteins, globulins, and the like. In compositions comprising saline or other carrier-containing salts, the carrier is preferably added after the lipid particles are formed. Thus, after formation of the lipid-nucleic acid composition, the composition may be diluted in a pharmaceutically acceptable carrier such as physiological saline.
The term "treating" relates to reversing, alleviating, inhibiting the progression or prevention of a disorder or condition to which the term applies, or one or more symptoms of such disorder or condition. The term "treatment" as used herein relates to the action of a verb treatment, the latter as just defined.
The term "subject" includes, but is not limited to: a human (i.e., male or female of any age group, e.g., pediatric subjects (e.g., infants, children, adolescents) or adult subjects (e.g., young adults, middle aged adults, or senior adults)) and/or a non-human animal, e.g., a mammal, e.g., a primate (e.g., cynomolgus monkey, rhesus monkey), cow, pig, horse, sheep, goat, rodent, cat, and/or dog. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human animal. The terms "human", "patient" and "subject" are used interchangeably herein.
The term "effective amount" refers to an amount sufficient to elicit a biological response of interest. As will be appreciated by those of ordinary skill in the art, the effective amount of the pharmaceutical composition, lipid nanoparticle of the present invention may vary depending on the following factors: for example, biological targets, pharmaceutical compositions, pharmacokinetics of lipid nanoparticles, the disease being treated, the mode of administration, and the age health and symptoms of the subject. The effective amount includes a therapeutically effective amount and a prophylactically effective amount.
As used herein, unless otherwise indicated, a "therapeutically effective amount" is an amount sufficient to provide a therapeutic benefit in the treatment of a disease, disorder or condition, or to delay or minimize one or more symptoms associated with a disease, disorder or condition. A therapeutically effective amount of a pharmaceutical composition, lipid nanoparticle refers to an amount of a therapeutic agent, alone or in combination with other therapies, that provides a therapeutic benefit in the treatment of a disease, disorder or condition. The term "therapeutically effective amount" may include an amount that improves overall treatment, reduces or avoids symptoms or causes of a disease or disorder, or enhances the therapeutic effect of other therapeutic agents.
As used herein, a "prophylactically effective amount" of a pharmaceutical composition, lipid nanoparticle, unless otherwise indicated, is an amount sufficient to prevent a disease, disorder, or condition, or to prevent one or more symptoms associated with a disease, disorder, or condition, or to prevent recurrence of a disease, disorder, or condition. A prophylactically effective amount of a pharmaceutical composition, lipid nanoparticle refers to an amount of therapeutic agent, alone or in combination with other agents, that provides a prophylactic benefit in the prevention of a disease, disorder or condition. The term "prophylactically effective amount" may include an amount that improves overall prophylaxis, or an amount that enhances the prophylactic effect of other prophylactic agents.
Examples II
The present invention will be further described in detail with reference to the following examples in order to make the technical solution of the present invention clearer and more specific. The following examples are presented only to illustrate specific embodiments of the invention so that those skilled in the art can understand the invention and are not intended to limit the scope of the invention. In the specific embodiment of the present invention, technical means, methods, and the like not specifically described are conventional technical means, methods, and the like in the art. Materials, reagents and the like used in the examples are commercially available unless otherwise specified.
TABLE 1
English or abbreviations Chinese character
THF Tetrahydrofuran (THF)
DCM Dichloromethane (dichloromethane)
MeOH Methanol
NMP N-methylpyrrolidone
EtOAc Acetic acid ethyl ester
CDCl3 Deuterated chloroform
TosMIC P-toluenesulfonylmethyl isonitrile
DMAP 4-Dimethylaminopyridine
EDCI 1- (3-Dimethylaminopropyl) -3-ethylcarbodiimide
NaBH4 Sodium borohydride
NaH Sodium hydride
Na2SO4 Sodium sulfate
EXAMPLE 1 Synthesis of Compound 1
In a 250mL round bottom flask, linolenic alcohol 1-1 (20 g,75.0mmol,1.0 eq.) carbon tetrabromide (37.3 g,112.5mmol,1.5 eq.) and triphenylphosphine (29.5 g,112.5mmol,1.5 eq.) were dissolved in dichloromethane (200 mL) at room temperature. The reaction system was stirred at room temperature overnight for reaction. The reaction was monitored by LCMS, after completion of the reaction, the reaction was quenched by addition of water (250 mL), extracted 3 times with dichloromethane (100 mL), the organic phases were combined, washed with brine (200 mL), dried over anhydrous Na 2SO4, filtered, and concentrated. The crude product was purified by a silica gel column to give 14g of oily compound 1-2.
TosMIC (3.85 g,19.7mmol,0.5 eq.) was dissolved in anhydrous DMF (30 mL) in ice bath, then sodium hydride (60%, 2.36g,59.1mmol,1.5 eq.) was added in portions to the reaction system, allowed to react for 0.5 hours after warming to room temperature, then Compound 1-2 (13.0 g,39.4mmol,1.0 eq.) was added, and the reaction was continued at room temperature for 12 hours after completion of the addition. After the reaction was completed, the reaction system was quenched by adding water (200 mL), extracted 3 times with ethyl acetate (70 mL), the organic phases were combined, washed 3 times with brine (200 mL), dried over anhydrous Na 2SO4, filtered, and concentrated. The crude product was purified by a silica gel column to give 6.9g of the oily compound 1-3.
Compounds 1-3 (4.9 g,7.1mmol,1.0 eq.) were dissolved in tetrahydrofuran (30 mL) in a 100mL round bottom flask at room temperature, hydrochloric acid (6M, 5.0 mL) was added to the reaction system, and the reaction system was stirred at room temperature for 2 hours. The reaction was adjusted to pH neutral with saturated aqueous sodium bicarbonate, extracted 3 times with ethyl acetate (30 mL), the organic phases were combined, washed with brine (100 mL), dried over anhydrous Na 2SO4, filtered, and concentrated. The crude product was purified by a silica gel column to give 3.0g of compounds 1-4.
Compounds 1-4 (1.5 g,2.8mmol,1.0 eq.) were dissolved in 10mL of methanol in a 25mL round bottom flask, sodium borohydride (158.9 mg,4.2mmol,1.5 eq.) was added and reacted for 3 hours at room temperature. After completion of the reaction, 15mL of water was added, quenched, extracted 3 times with ethyl acetate (20 mL), the organic phases were combined, washed with brine (50 mL), dried over anhydrous Na 2SO4, filtered, and concentrated to give 1.2g of the oily compound 1-5, which was used directly in the next reaction without purification.
In a 50mL round bottom flask, 4-fluorobenzoic acid (300 mg,2.1mmol,1.0 eq.) compounds 1-5 (1.1 g,2.1mmol,1.0 eq.), EDCI (1.2 g,6.3mmol,3.0 eq.), DMAP (256.6 mg,2.1mmol,1.0 eq.) are dissolved in 20mL dichloromethane. The reaction was carried out at room temperature overnight. LCMS monitors the reaction, after the reaction is completed, 20mL of water is added for quenching, dichloromethane (20 mL) is used for extraction for 3 times, the organic phases are combined, brine (50 mL) is used for washing, anhydrous Na 2SO4 is used for drying, filtering and concentrating are carried out, and the crude product is purified by a silica gel column to obtain 410mg of oily compound 1-6.
Compounds 1-6 (300 mg,0.46mmol,1.0 eq.) and N, N-dimethylethylenediamine (405.5 mg,4.6mmol,10.0 eq.) were dissolved in DIEA (5 mL) and NMP (5 mL) in a 20mL sealed tube and the reaction was warmed to 150℃and stirred for 6 hours. The reaction was cooled to room temperature, quenched by pouring into 30mL of water, extracted 3 times with ethyl acetate (30 mL), the organic phases were combined, washed with brine (50 mL), dried over anhydrous Na 2SO4, filtered, concentrated, and the crude product purified by prep-TLC to give 80.02mg of compound 1 as an oil.
1H NMR(400MHz,CDCl3):δppm 0.89(t,J=6.8Hz,6H),1.19-1.36(m,36H),1.55-1.64(m,4H),2.03-2.08(m,8H),2.74-2.79(m,4H),2.87(s,6H),3.29(t,J=5.2Hz,2H),3.62(t,J=5.2Hz,2H),5.08(p,J=6.4Hz,1H),5.32-5.42(m,8H),6.60(d,J=8.4Hz,2H),7.90(d,J=8.4Hz,2H),13.20(s,1H);ESI-MS m/z:719.7[M+H]+.
Pharmacological experiments
Experimental example 1 nanoparticle preparation
Materials used for lipid nanoparticle assembly are: (1) an ionizable lipid compound: the ionizable lipids synthesized as designed in the present invention or DLin-MC3-DMA (MC 3, purchased from AVT) as a control group; (2) structural lipids: such as Cholesterol (purchased from Sigma-Aldrich); (3) Phospholipids such as 1, 2-distearoyl-SN-glycero-3-phosphorylcholine (DSPC, available from AVT); (4) Polyethylene glycol lipid compounds such as dimyristoylglycerol-polyethylene glycol 2000 (DMG-PEG 2000 available from AVT); (5) The effective components of the nucleic acid fragment include Luciferase (Luciferase mRNA), siRNA, CRISPR Cas mRNA and the like. The component names and the structural formulas of the lipid nanoparticles are shown in Table 2.
TABLE 2
The preparation method of the lipid nanoparticle comprises the following steps: (1) Respectively dissolving and mixing 50%,38.5%,10% and 1.5% of ionizable lipid compound, cholesterol, phospholipid and polyethylene glycol lipid in ethanol; (2) The mRNA active ingredient was dissolved in 25mM sodium acetate solution (ph=4.5); (3) Mixing the organic phase dissolved with the lipid mixture and the aqueous phase dissolved with the mRNA component by using an automatic high-throughput microfluidic system according to a flow rate ratio ranging from 1:1 to 1:4, wherein the mixing speed is 10-18mL/min; (4) The lipid nanoparticles (N/P6) were diluted with phosphate buffer solution and then the nanoparticle solution was ultrafiltered to the original preparation volume using an ultrafiltration tube (available from Millipore) with a molecular weight cut-off of 30 kDa; (5) The obtained nano particles are filtered and sterilized by a sterile filter membrane with the diameter of 0.2 mu m and then stored in a sealed glass bottle at low temperature.
The preparation method of the lipid nanoparticle comprises a microfluidic mixing system, but is not limited to the method, and also comprises a T-type mixer, an ethanol injection method and the like.
Experimental example 2 characterization of physical Properties of lipid nanoparticles
Particle size and particle size dispersion coefficient (PDI) of the prepared lipid nanoparticles were measured using a Zetasizer Pro (available from Malvern Instruments Ltd) and DynaPro NanoStar (available from Wyatt) dynamic light scattering instrument. The extent of entrapment of the lipid nanoparticle to the RNA was characterized by the encapsulation efficiency (Encapsulation Efficiency%), a coefficient reflecting the extent of binding of the lipid nanoparticle to the RNA fragment. The coefficients were measured by the method of Quant-it TM RiboGreen RNA Assay (available from Invitrogen). Lipid nanoparticle samples were diluted in TE buffer (10 mM Tris-HCl,1mM EDTA,pH =7.5), and a portion of the sample solution was taken out and added to 0.5% Triton (Triton X-100) and allowed to stand at 37℃for 30 minutes. Adding inImmediately after the reaction, the fluorescence value was read by a Varioskan LUX multifunctional microplate reader (commercially available from Thermofisher) under the conditions of an absorption light band of 485nm and an emission light band of 528nm to obtain the entrapping rate value.
Experimental example 3 animal experiment
Assessment of the delivery efficacy and safety of nanoparticles loaded with luciferase mRNA (ex Trilink, L-7202) in mice. The test mice were SPF grade C57BL/6 mice, females, 6-8 weeks old, weighing 18-22g, purchased from Beijing Bei Fu Biotechnology Co. All animals are subjected to adaptive feeding for more than 7 days before the test, drinking water is fed freely during the test, the illumination is changed for 12/12h, the indoor temperature is 20-26 ℃, and the humidity is 40-70%. Mice were randomly grouped.
The lipid nanoparticle prepared above and having the luciferase mRNA contained therein was injected into mice intravenously at a single dose of 0.5mg/kg of mRNA, and the mice were subjected to in vivo bioluminescence detection using a small animal in vivo imaging system (IVIS LUMINA III, available from Perkinelmer) at 6 hours after administration. The specific operation steps of the detection are as follows: a15 mg/mL concentration of D-fluorescein solution was prepared in physiological saline, and each mouse was given the substrate by intraperitoneal injection. 10 minutes after substrate administration, mice were anesthetized with isoflurane at a concentration of 2.5% in an anesthetic box. The anesthetized mice are placed in IVIS for fluorescence imaging and data acquisition and data analysis are carried out on the parts with concentrated distribution of fluorescence.
The in vivo delivery efficiency of the lipid nanoparticle carrier is expressed as the average of the fluorescence intensity and total photon number of different animals within the same test group, as shown in table 3. The higher the values of fluorescence intensity and total photon number, the higher the in vivo delivery efficiency of the lipid nanoparticle for the mRNA fragment. Lipid nanoparticles comprising the ionizable lipids of the invention have good in vivo delivery efficiency.
TABLE 3 Table 3
Example 4 in vitro delivery efficiency and safety assessment
The effect of delivering nanoparticles loaded with luciferase mRNA (purchased from Trilink, L-7202) at the in vitro cellular level was evaluated as well as safety. The cells used in the test were human embryonic kidney cells 293 (HEK 293T cells) and cultured in DMEM Du modified Eagle medium (purchased from Thermofisher) containing 10% fetal bovine serum and 5% penicillin-streptomycin double antibody at room temperature of 37℃and carbon dioxide concentration of 5%. The cells were uniformly scattered and plated in a 48-well plate, and after 24 hours of stationary culture in an incubator, a lipid nanoparticle solution encapsulating fluorescein mRNA was added. After 24 hours incubation, cells were disrupted and the intensity and Relative Light Units (RLU) of each lipid nanoparticle-mediated luciferase expression was measured using luciferase assay reagents (available from Promega), with higher intensities of expression representing higher delivery efficiencies of the lipid material at the cellular level. Meanwhile, cytotoxicity was tested 24 hours later on a parallel lipid nanoparticle treated cell group using CCK-8 reagent (available from DOJINDO). The test was performed with the negative control of the cell group to which PBS alone was added. The method comprises the following specific steps: after CCK-8 solution is added into cells, the cells are kept stand in a cell incubator at 37 ℃ for 4 hours, and the light absorption value is read by a multifunctional enzyme-labeled instrument under the condition that the light absorption wave band is 450 nm. The ratio of absorbance read by the nanoparticle treated cells to that of the negative control was used as a characterization parameter for cell viability. The effect of nanoparticle delivery at the in vitro cellular level and toxicity data are shown in table 4. Lipid nanoparticles comprising the ionizable lipid compounds of the invention have good in vitro delivery efficiency and lower toxicity.
TABLE 4 Table 4
The foregoing descriptions of specific exemplary embodiments of the present invention are presented for purposes of illustration and description. It is not intended to limit the invention to the precise form disclosed, and obviously many modifications and variations are possible in light of the above teaching. The exemplary embodiments were chosen and described in order to explain the specific principles of the invention and its practical application to thereby enable one skilled in the art to make and utilize the invention in various exemplary embodiments and with various modifications as are suited to the particular use contemplated. It is intended that the scope of the invention be defined by the claims and their equivalents.

Claims (10)

1. A compound of formula (I), or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof:
Wherein,
Each R 1、R2、R3 is independently selected from hydrogen or C 1-8 alkyl;
L 1 is selected from C 1-10 alkylene, C 2-10 alkenylene, or C 2-10 alkynylene;
G is phenylene;
Each R 4、R5 is independently selected from C 1-30 alkyl, C 2-30 alkenyl, or C 2-30 alkynyl.
2. The compound of claim 1, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, having a structure represented by formula (II):
wherein R 1、R2、R3、R4、R5、L1 is each as defined in claim 1.
3. The compound of claim 1 or 2, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein each R 1、R2、R3 is independently selected from hydrogen, methyl, ethyl, or propyl; or, L 1 is selected from-CH 2-、-(CH2)2-、-(CH2)3 -or- (CH 2)4 -.
4. The compound of claim 1 or 2, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein each R 4、R5 is independently selected from- (CH 2)m1CH=CH(CH2)n1CH=CH(CH2)m2CH3;
m1 and m2 are each independently selected from 2, 3,4, 5, 6, 7, 8, 9, 10 or 11;
n1 is selected from 0,1,2 or 3.
5. The compound of claim 1, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, wherein the compound has the structure:
6. A process for the preparation of a compound of formula (II), or a pharmaceutically acceptable salt, isotopic variant, tautomer or stereoisomer thereof, according to claim 2, which comprises:
reacting a compound of formula (IIa) with a compound of formula (IIb) to obtain a compound of formula (II);
Wherein X 1 is selected from halogen; r 1、R2、R3、R4、R5、L1 is each as defined in claim 2.
7. A pharmaceutical composition comprising a compound of any one of claims 1-5, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof, and a pharmaceutically acceptable adjuvant.
8. A lipid nanoparticle comprising a lipid component and optionally an active ingredient;
the lipid component comprises: ionizable cationic lipids, structural lipids, neutral lipids, and polymeric lipids;
Wherein the ionizable cationic lipid is a compound of any one of claims 1-5, or a pharmaceutically acceptable salt, isotopic variant, tautomer, or stereoisomer thereof.
9. Use of a compound according to any one of claims 1-5, or a pharmaceutically acceptable salt, isotopic variant, tautomer or stereoisomer thereof, or a pharmaceutical composition according to claim 7, or a lipid nanoparticle according to claim 8, for the manufacture of a medicament for the treatment, diagnosis or prevention of a disease.
10. Use of a compound according to any one of claims 1-5, or a pharmaceutically acceptable salt, isotopic variant, tautomer or stereoisomer thereof, or a pharmaceutical composition according to claim 7, or a lipid nanoparticle according to claim 8, for the preparation of a medicament for delivering a load selected from one or more of a therapeutic, prophylactic or diagnostic agent.
CN202410324191.0A 2024-03-20 2024-03-20 Ionizable lipid compound, preparation method and application thereof Pending CN118290282A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202410324191.0A CN118290282A (en) 2024-03-20 2024-03-20 Ionizable lipid compound, preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202410324191.0A CN118290282A (en) 2024-03-20 2024-03-20 Ionizable lipid compound, preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN118290282A true CN118290282A (en) 2024-07-05

Family

ID=91685384

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202410324191.0A Pending CN118290282A (en) 2024-03-20 2024-03-20 Ionizable lipid compound, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN118290282A (en)

Similar Documents

Publication Publication Date Title
AU2010249881B2 (en) Compositions comprising cationic amphiphiles and colipids for delivering therapeutics molecules
CN115887674B (en) lipid nanoparticles
TWI828461B (en) Ionizable lipid compounds
JP2023505316A (en) nanomaterials
AU2013380825B2 (en) Lipidosome preparation, preparation method and application thereof
CN114989027A (en) Cationic lipid compounds and compositions for delivery of nucleic acids and uses
CN115850104B (en) Ionizable lipid compounds
CN118290282A (en) Ionizable lipid compound, preparation method and application thereof
CN118290339A (en) Ionizable lipid compounds and uses thereof
CN117924106A (en) Ionizable lipid compounds
WO2024149229A1 (en) Lipid nanoparticle compositions and uses thereof
CN116854606B (en) Cationic lipid compound, composition and application thereof
CN116082179B (en) Ionizable lipid based on endogenous dicarboxylic acid, preparation method and application thereof
EP4385523A1 (en) Lipid-based topical injection formulations
EP4332087A1 (en) Lipid nanoparticles
US20240245782A1 (en) Ionizable lipid compounds
CN116211831A (en) Lipid matrix local injection preparation
TWI847412B (en) Lipid nanoparticles
CN118105357A (en) Intrathecal delivery preparation and application thereof
TW202423452A (en) Lipid-based topical injection formulations
CN117534585A (en) Novel ionizable cationic lipid compound, 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