CN117899048A - Targeting lipid nanoparticle and preparation and application thereof - Google Patents

Targeting lipid nanoparticle and preparation and application thereof Download PDF

Info

Publication number
CN117899048A
CN117899048A CN202410003683.XA CN202410003683A CN117899048A CN 117899048 A CN117899048 A CN 117899048A CN 202410003683 A CN202410003683 A CN 202410003683A CN 117899048 A CN117899048 A CN 117899048A
Authority
CN
China
Prior art keywords
sequence
lipid
seq
targeting
lipids
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
CN202410003683.XA
Other languages
Chinese (zh)
Inventor
章雪晴
高明珠
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.)
Shanghai Jiaotong University
Original Assignee
Shanghai Jiaotong University
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 Shanghai Jiaotong University filed Critical Shanghai Jiaotong University
Priority to CN202410003683.XA priority Critical patent/CN117899048A/en
Publication of CN117899048A publication Critical patent/CN117899048A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention provides a targeted lipid nanoparticle and preparation and application thereof, in particular to a targeted lipid nanoparticle composition, which comprises the following components: comprises an ionizable lipid and a targeting ingredient, wherein the ionizable lipid is AA3-Dlin. The nanoparticle composition has both targeting and synergistic effects. On one hand, the nanoparticle composition can specifically deliver mRNA encoding therapeutic protein to macrophages, so that the cells can translate active protein, and the problem that protein drugs cannot be directly delivered due to instability in vivo is solved. On the other hand, the nanoparticle composition can promote the polarization of macrophages from M1 type to M2 type, and more compositions are specifically ingested by the increased M2 type macrophages, so that a closed loop of a positive feedback targeting mechanism is formed, more compositions are recruited to be enriched at a lesion part, the transfer efficiency and the transfection efficiency are comprehensively improved, and a more efficient treatment effect is realized.

Description

Targeting lipid nanoparticle and preparation and application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to a targeting lipid nanoparticle, and preparation and application thereof.
Background
Macrophages are important immune cells in humans and originate from monocytes differentiated from hematopoietic stem cells in the bone marrow. Macrophages migrate and circulate within nearly every tissue and differentiate into tissue-specific macrophages, including osteoclasts (skeletal system), microglia (central nervous system), alveolar macrophages (lung), kupffer cell (liver), splenic macrophages (spleen), tissue cells (connective tissue), and the like. Macrophages have a variety of morphologies and phenotypes due to their distribution and action in a variety of different tissues and organs.
Macrophages can be generally classified into M1-phenotype macrophages and M2-phenotype macrophages. The M1 phenotype is a pro-inflammatory cell and is polarized by lipopolysaccharide alone or in combination with Th1 cytokines (e.g., IFN-gamma, GM-CSF) to produce pro-inflammatory cytokines such as interleukin-1β (IL-1β), IL-6, IL-12, IL-23 and TNF- α; the M2 phenotype has anti-inflammatory and immunoregulatory effects, and is polarized by Th2 cytokines IL-4 and IL-13 to produce anti-inflammatory cytokines such as IL-10 and TGF-beta. M1 and M2 phenotype macrophages have different functions and their equilibrium polarization plays an important role in the development and progression of cardiovascular and cerebrovascular diseases (e.g. atherosclerosis, stroke), autoimmune diseases (e.g. rheumatoid arthritis, multiple sclerosis, crohn's disease, inflammatory bowel disease), fibrosis, diabetes, tumors, etc.
Ischemic Stroke (IS) IS the second leading cause of death and disability in adults worldwide, and high mortality and long-term disability rates make IS one of the most serious public health problems. The current clinical treatment option is to dredge blood vessels using recombinant tissue plasminogen activator (rtPA) and intravascular thrombectomy (EVT) within 4.5 and 24 hours after symptoms appear. However, this recanalization therapy fails to alleviate the clinical problems of neuroinflammation and Blood Brain Barrier (BBB) damage following stroke, which may increase the risk of cerebral hemorrhagic transformation and death. Therefore, there is an urgent need to develop therapeutic strategies that can effectively reduce post-stroke neuroinflammation and BBB destruction to repair post-stroke brain tissue damage and neurological deficit.
There is growing evidence that the neuroimmune response is closely related to ischemic stroke, which is intimately involved in various stages of the pathological process from acute brain injury to tissue injury. In the early stages of stroke, the innate immune effector cells of the Central Nervous System (CNS), i.e., microglia, can respond rapidly (< 1 hour) after acute brain injury and promote brain repair by eliminating cell debris and producing anti-inflammatory mediators at the diseased site. In pathological progression, early aggregated protective microglia can rapidly switch from the M2 anti-inflammatory phenotype to the M1 pro-inflammatory phenotype, with concomitant upregulation of inflammatory substances such as cytokines, chemokines, and Reactive Oxygen Species (ROS). These inflammatory substances mediate neuroinflammation, BBB damage and neuronal death. This pathological process suggests that the use of a phenotypic conversion factor to drive the transition of microglial cells from the M1 pro-inflammatory phenotype to the M2 anti-inflammatory phenotype has therapeutic promise in reducing post-stroke neuroinflammation and BBB destruction.
IL-10 is an effective anti-inflammatory cytokine that promotes the polarization of microglial cells to the M2 phenotype through the Janus kinase 1-Stat3 pathway, thereby promoting neuroinflammatory regression, preventing BBB destruction, and restoring neural function after ischemic stroke. Although IL-10 has good neuroprotection, the clinical use of IL-10 has been limited due to its short plasma half-life (3 minutes), and the potential for systemic administration of high doses to trigger immunosuppression and increase the risk of infection.
Therefore, it is urgent to develop a strategy capable of locally expressing biologically active IL-10 in ischemic brain, promoting polarization of microglial cells to M2 phenotype, promoting regression of neuroinflammation, and preventing disease progression.
The rapid pooling of mRNA vaccines for the treatment of novel coronaviruses (COVID-19) highlights the unlimited potential of mRNA technology in the field of disease treatment. Wherein the lipid nanoparticle (Lipid Nanoparticle, LNP) is the mainstream delivery system of nucleic acid drugs by virtue of its unique structural and physicochemical properties.
The LNP system at present mainly comprises four components: ionizable lipids, structural lipids (e.g., cholesterol), helper lipids (e.g., DSPC), and polymer conjugated lipids (e.g., PEG-lipids). Because LNP accumulates in the liver, most current LNP delivery systems are liver-targeted, and there is still a lack of delivery vehicles that can deliver mRNA therapeutic drugs to brain injury sites, which hampers the clinical transformation of mRNA therapy-related brain diseases, including ischemic stroke.
Therefore, there IS an urgent need in the art to develop LNPs with macrophage targeting for the treatment of macrophage-related diseases, especially for the targeted delivery of mRNA therapeutic drugs to the brain microenvironment for application in the IS field, thereby improving post-IS exacerbated brain tissue damage and neurological dysfunction.
Disclosure of Invention
In view of the above problems, the present invention provides a novel targeted lipid nanoparticle, which can deliver a phenotype switching factor, such as an anti-inflammatory factor IL-10 (which can be used for treating IS), from mRNA to microglial cells by targeting the lipid nanoparticle, so that the cells transcribe and translate the active anti-inflammatory factor IL-10, and the problem that the directly delivered protein medicine IS unstable in vivo IS solved. Meanwhile, the nano particles have targeting and synergistic effects, can recruit more macrophage M1 type to M2 type polarization, form a closed loop of positive feedback mechanism, therefore, more nano particles are recruited to be enriched at the lesion site, the transfer efficiency and the transfection efficiency are comprehensively improved, and the more efficient and stable treatment effect is realized.
In a first aspect of the invention, there is provided a targeted lipid nanoparticle composition comprising: an ionizable lipid and a targeting ingredient; and the ionizable lipid is AA3-Dlin; wherein the method comprises the steps of
The targeting component is selected from any one or a combination of a plurality of ionizable lipid connected with a targeting group, structural lipid connected with the targeting group, auxiliary lipid connected with the targeting group, PEG-lipid connected with the targeting group and polymer connected with the targeting group;
the targeting group is selected from the group consisting of: mannose, mannose derivatives, galactose derivatives, dextran derivatives, peptide fragments or any one or a combination of a plurality thereof.
In another preferred embodiment, the derivative is a glycoside, sugar amine, glycan, PEG-modified sugar ring structure, isotope or other substituent substituted sugar ring structure; preferred derivatives are combinations of one or more of methyl-D-mannoside, 1-alpha formylmethyl-mannopyranoside, 4-aminophenyl-alpha-D-mannopyranoside, 4-nitrophenyl-alpha-D-mannopyranoside, 4-methylumbelliferone-alpha-D-mannopyranoside, mannose-6-phosphate, carbamoyl-D-mannose, N-acetamido-beta-1, 2-mannose, methyl 6-O- (aD-mannopyranosyl) -aD-mannopyranoside, methyl 3-O- (aD-mannopyranosyl) -aD-mannopyranoside, mannosamine, mannan, and the like.
In another preferred embodiment, the composition further comprises a combination of one or more of a helper lipid, a structural lipid, a PEG-lipid, a polymer.
In another preferred embodiment, the composition comprises the following components in mole percent: 20-65 ionizable lipids, 0-60 targeting group-linked ionizable lipids, 0-40 helper lipids and/or targeting group-linked helper lipids, 0-60 structural lipids and/or targeting group-linked structural lipids, 0-10 PEG-lipids and/or targeting group-linked PEG-lipids, and the molar ratio of the targeting components cannot be 0; when the targeting component is an ionizable lipid to which the targeting moiety is attached, at least any one of a helper lipid and/or a helper lipid to which the targeting moiety is attached, a structural lipid and/or a structural lipid to which the targeting moiety is attached, a PEG-lipid and/or a PEG-lipid to which the targeting moiety is attached is other than 0; preferred are: 20-65 ionizable lipids, 0-60 targeting group-linked ionizable lipids, 3-40 helper lipids and/or targeting group-linked helper lipids, 15-60 structural lipids and/or targeting group-linked structural lipids, 0.1-10 PEG-lipids and/or targeting group-linked PEG-lipids; or, the molar ratio of the ionizable lipid and/or the targeting group-linked ionizable lipid to the polymer and/or the targeting group-linked polymer is from 0.5:1 to 80:1, preferably from 20:1 to 80:1, and most preferably from 40:1 to 80:1.
In another preferred embodiment, the auxiliary material with the targeting function has a structure as shown in the following formula:
Wherein:
r is selected from any one or a combination of mannose, mannose derivatives, galactose derivatives, dextran derivatives; n is selected from integers from 22 to 220.
In another preferred embodiment, the composition further comprises: a drug carried; preferably, the composition further comprises pharmaceutically acceptable excipients.
In another preferred embodiment, the loaded medicament comprises: any one or a combination of nucleic acids, small molecules, proteins.
In another preferred embodiment, the drug is a nucleic acid encoding one or more proteins having phenotypic modulation, neurotrophic, inflammatory modulating effects; preferably, the nucleic acid encodes one or more phenotype-modulating associated factors, neurotrophic factors, or inflammatory mediators, preferably, the nucleic acid encodes an anti-inflammatory factor, and more preferably, the nucleic acid encodes IL-10.
In another preferred embodiment, the nucleic acid encodes an amino acid sequence of IL-10 selected from the group consisting of: a sequence as set forth in SEQ ID NO. 1 or 2, a sequence having at least 80% identity to the sequence set forth in SEQ ID NO. 1 or 2, or an amino acid sequence having 1 or more conservative substitutions as compared to the sequence set forth in SEQ ID NO. 1 or 2.
In another preferred embodiment, the nucleic acid is a messenger ribonucleic acid (IL-10 mRNA, mIL-10) encoding IL-10; and the mRNA encoding IL-10 comprises a sequence as set forth in SEQ ID NO. 6 or 7 or 8, a sequence having at least 80% identity to the sequence set forth in SEQ ID NO. 6 or 7 or 8, or a nucleic acid sequence having 1 or more conservative substitutions as compared to the sequence set forth in SEQ ID NO. 6 or 7 or 8; preferably, the method comprises the steps of,
MRNA encoding IL-10 comprises capping elements, 5 'UTRs, signal peptide sequences, open reading frames, 3' UTRs, and/or polyAs; more preferably, the capping structure comprises 3'-O-Me-m7G (5') ppp (5 ') G linked to a 5' utr;
More preferably, the 5' UTR comprises a sequence as shown at positions 1 to 47 in SEQ ID NO. 9 or 10, a sequence having at least 80% identity to the sequence shown at positions 1 to 47 in SEQ ID NO. 9 or 10, or a nucleic acid sequence having 1 or more conservative substitutions as compared to the sequence shown at positions 1 to 47 in SEQ ID NO. 9 or 10;
The signal peptide sequence comprises a sequence as shown in SEQ ID NO. 9 at positions 48 to 101, a sequence having at least 80% identity to the sequence shown in SEQ ID NO. 7 at positions 48 to 101, or a nucleic acid sequence having 1 or more conservative substitutions as compared to the sequence shown in SEQ ID NO. 7 at positions 48 to 101;
The open reading frame comprises a sequence as set forth in SEQ ID NO. 9 or 11 at positions 102 to 584, a sequence having at least 80% identity to the sequence set forth in SEQ ID NO. 9 at positions 102 to 584, or a nucleic acid sequence having 1 or more conservative substitutions as compared to the sequence set forth in SEQ ID NO. 9 at positions 102 to 584;
The 3' UTR comprises a sequence as shown in SEQ ID NO. 9 from 585 to 677, a sequence having at least 80% identity to the sequence shown in SEQ ID NO. 9 from 585 to 677, or a nucleic acid sequence having 1 or more conservative substitutions as compared to the sequence shown in SEQ ID NO. 9 from 585 to 677;
the nucleic acid sequence of polyA comprises 65-250A;
Optimally, the mRNA encoding IL-10 comprises a sequence as set forth in any of SEQ ID NOS: 9-12 having at least 80% identity to the sequence set forth in any of SEQ ID NOS: 9-12 or 1 or more conservatively substituted nucleic acid sequences as compared to the sequence set forth in any of SEQ ID NOS: 9-12.
In a second aspect of the invention there is provided the use of a lipid nanoparticle composition as described in the first aspect of the invention for the preparation of a macrophage targeted drug.
In another preferred example, the drug is any one or a combination of a plurality of inflammatory disease drugs, tumor drugs, infectious disease drugs, metabolic disease drugs, cardiovascular and cerebrovascular disease drugs, respiratory diseases, autoimmune diseases; preferably, the medicament is a medicament for the treatment of atherosclerosis and/or stroke.
It is understood that within the scope of the present invention, the above-described technical features of the present invention and technical features specifically described below (e.g., in the examples) may be combined with each other to constitute new or preferred technical solutions. And are limited to a space, and are not described in detail herein.
Drawings
FIG. 1 (A) stability of naked mRNA or LNP-coated mRNA in serum. (B and C) particle size (B) and turbidity (C) of the mll-10@lnps measured in the presence of serum at 37 ℃ for 24 hours (n=3). (D) haemolysis rate of ml10@lnps (n=3).
FIG. 2 mIL-10@LNPs in vitro targeting evaluation. (a and B) IL-4 activated microglial cells (a) or astrocytes (B) were incubated with Cy 5-labeled LNPs (Cy5 LNPs) for an average fluorescence intensity (MFI) of 4 hours (n=3) by flow cytometry analysis. (C) Confocal microscopy (CLSM) images (red) of IL-4 activated microglia incubated with Cy5 LNPs for 4 hours. Nuclei were stained with DAPI (blue) and microglia were immunostained with CD206 (green). White arrows indicate Cy5 LNPs co-located with CD 206.
FIG. 3mIL-10@MLNPs in vitro transfection efficiency and evaluation of anti-inflammatory effect. (A) Flow cytometry determined microglial in vitro transfection efficiency (n=3). (B-H) LPS stimulation for 4 hours, primary microglia were incubated with ml10@mlnps for 24 hours and the cells were analyzed for expression of IL-10 (B), TNF- α (C), CD206 (D), arg-1 (E), TGF- β (F) mRNA (n=3) by real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR).
FIG. 4 expression levels of IL-10, TNF-. Alpha., iNOS, IL-6 and TGF-. Beta.mRNA (n=3) in Lipopolysaccharide (LPS) -activated BV2 cells treated with mIL-10@MLNPs by RT-qPCR analysis.
FIG. 5 therapeutic effect of mIL-10@MLNPs in dMCAO mouse model. (A) A schematic of the treatment regimen for dMCAO mice of IS has been established. (B) Representative immunofluorescence images of endogenous IgG extravasation (green) and MAP2 (red) in brain sections from sham operated mice or dMCAO mice treated with PBS, MLNPs, or mll-10@mlnps. (C and D) quantitative determination of MFI (C) and infarct volume (D) of each group IgG (n=4). (E) The nervous system of the mice of the indicated group was overall assessed using Garcia (n=8).
Figure 6 in vivo safety assessment. (A and B) healthy C57BL/6 mice were given twice a week with a dose of mIL-10@MLNPs therapeutic agent of 0.3mg/kg mRNA per mouse, and histological, hematological and biochemical analysis of the major organ (A) and blood sample (B) were performed one week after the last administration.
Detailed Description
The compound is a lipid whose pH value influences the degree of protonation and thus the charging properties. Preferably, a class of lipids that is generally barely charged at normal neutral physiological pH conditions, but is capable of being positively charged at acidic pH to bind to negatively charged nucleic acids.
By "isomer" is meant a different compound having the same molecular formula, including but not limited to enantiomers, diastereomers, cis-trans isomers, and the like, as known in the art.
The connection mode of the targeting group in the targeting component is not limited, so long as the targeting component contains the targeting group. The means of attachment may be selected from physical and/or chemical attachment. The attachment means may be by intermolecular forces (e.g., van der Waals forces, hydrogen bonding, etc.) and/or chemical bonds (e.g., ionic bonds, covalent bonds, etc.). The connection mode can be direct connection or indirect connection through some fragments. The preferred attachment means is coupling by chemical bond, and further preferred the targeting component is selected from PEG-lipids attached by targeting groups.
In certain embodiments, the targeting component has a structure represented by the formula:
Wherein:
r is selected from any one or a combination of mannose, mannose derivatives, galactose derivatives, dextran derivatives; n is selected from integers from 22 to 220.
The derivatives of the invention are core structures which are the same as or basically similar to mannose, galactose and glucan in structure, and include but are not limited to: glycosides, sugar amines, glycans, PEG-modified sugar ring structures, isotopically or other substituent substituted sugar ring structures, and the like. Exemplary derivatives include, but are not limited to: methyl-D-mannoside, 1- α -formylmethyl-mannopyranoside, 4-aminophenyl- α -D-mannopyranoside, 4-nitrophenyl- α -D-mannopyranoside, 4-methylumbelliferone- α -D-mannopyranoside, mannose-6-phosphate, carbamoyl-D-mannose, N-acetamido- β -1, 2-mannose, methyl 6-O- (aD-mannopyranosyl) -aD-mannopyranoside, methyl 3-O- (aD-mannopyranosyl) -aD-mannopyranoside, mannosamine, mannosan, and the like.
The targeted lipid nanoparticles of the present invention may carry any therapeutic component known or to be developed in the art that can be delivered by conventional lipid nanoparticle technology, including but not limited to any one or a combination of nucleic acids, small molecules, proteins.
The "nucleic acid" according to the present invention may be a nucleotide polymer of any length. Including, but not limited to, single-stranded DNA, double-stranded DNA, plasmid DNA, short isomers, mRNA, tRNA, rRNA, long non-coding rnas (lncRNA), micronon-coding rnas (miRNA and siRNA), telomerase RNA (Telomerase RNA), small-molecule rnas (snRNA and sc small-molecule RNA), circular rnas (circRNA), synthetic mirnas (MIRNA MIMICS, miRNA agomir, miRNA antagomir), antisense oligonucleotides (ASO), ribozymes (ribozyme), asymmetric interfering rnas (aiRNA), dicer-substrate rnas (dsRNA), small hairpin rnas (shRNA), guide rnas (gRNA), small guide rnas (sgrnas), locked Nucleic Acids (LNA), peptide Nucleic Acids (PNA), morpholine antisense oligonucleotides, morpholino oligonucleotides, or combinations of one or more of the biospecific oligonucleotides.
In certain embodiments of the invention, the nucleic acid molecule is mRNA. The mRNA is a single-stranded ribonucleic acid transcribed from one strand of DNA as a template and carrying genetic information to direct protein synthesis. The mRNA may be monocistronic mRNA or polycistronic mRNA.
The cap structure of mRNA is that guanosine is attached to the 5' -end of mRNA via pyrophosphate to form a 5' -5' -triphosphate linkage. When the nitrogen atom at position 7 of G is methylated to form m7GPPPN, the Cap at this time is referred to as Cap0. Cap1 if the 2' -O position of the first nucleotide of the mRNA is also methylated to form m7 GPPPNm; if the first and second nucleotides of mRNA are both methylated at the 2' -O position to m7G-PPPNmNm, cap2 is obtained.
To increase mRNA stability and protein translation levels, the sequence of the coding region may be codon optimized during synthesis by adding cap analogs to achieve capping of cap0, cap1, cap2, etc., while natural nucleotides UTP, ATP, CTP, GTP are replaced in whole or in part with A variety of chemically modified nucleotides such as pseudouridine (ψ), N1-methyl-pseudouridine (N1M- ψ), 5-methyluridine (M5U), methoxyuridine (mo 5U), 2-thiouridine (s 2U), carboxymethyl uridine (cam 5U), 5-methoxyuridine (5 moU), N6-methyl adenosine (M6A), N1-methyl adenosine (M1A), methyl cytosine (M5C), methoxy cytosine (mo 5C), hydroxymethyl cytosine (hm 5C), etc., to achieve chemical modification of the bases. 64-250 poly (A) tails can also be added to the end of mRNA sequence by means of tailing enzyme, PCR tailing, chemical synthesis tailing and the like to achieve the purpose of stabilizing mRNA. In particular, a preferred mRNA for use in the present invention is an mRNA encoding IL-10 (IL-10 mRNA), which in a preferred embodiment of the present invention encodes an IL-10 amino acid sequence as set forth in Table 1A below, or a cDNA sequence as set forth in Table 2A:
TABLE 1 amino acid sequence of IL-10 (5 '. Fwdarw.3')
TABLE 2A cDNA sequence encoding IL-10 (i.e., ORF,5 '. Fwdarw.3')
In another preferred embodiment of the invention, the mRNA has the sequence shown in Table 3A or Table 4A below:
TABLE 3 mRNA corresponding to cDNA for IL-10 (5 '. Fwdarw.3')
TABLE 4A mRNA of IL-10 (5 '. Fwdarw.3')
/>
/>
As used herein, a "small molecule" refers to a compound that is not a protein or nucleic acid molecule. The small molecule may be a small molecule of a therapeutic and/or prophylactic agent, such as an antibiotic, anti-inflammatory, anti-cancer, anti-viral, immunosuppressant, analgesic, anti-fungal, antiparasitic, anticonvulsant, antidepressant, anxiolytic, antipsychotic, or the like.
"Protein" as used herein refers to a molecule or complex comprising one or more polypeptides having secondary, tertiary and/or quaternary structure. The secondary, tertiary and/or quaternary structure of a protein is typically stabilized using non-covalent bonds such as ionic bonds, hydrogen bonds, hydrophobic interactions and/or van der waals interactions. Additionally, or alternatively, the protein may include disulfide bonds, for example between thiol groups of cysteine residues. Exemplary proteins include, but are not limited to, antibodies, antigens or fragments thereof, fusion proteins, recombinant proteins, polypeptides, short peptides, enzymes, and the like.
The carried nucleic acid medicine can code one or more proteins with phenotype regulation effect, neurotrophic effect and inflammation regulation effect; preferably, one or more of a phenotype-modulating associated factor, a neurotrophic factor, an inflammatory factor, an anti-inflammatory factor, and more preferably IL-10.
By "phenotype-modulating factor" is meant a protein capable of causing a change in morphology, function and structure experienced by a cell to adapt to an environmental change (different stages of development or different disease states of the body), and illustratively preferably a factor capable of causing a transition from the M1 form to the M2 form in macrophages (including microglia and the like).
The "neurotrophic factor" is a class of proteins that play an important role in neuronal development, survival and apoptosis, including, but not limited to, nerve Growth Factor (NGF), brain derived growth factor (BDNF), neurotrophic factor 3 (NT-3), neurotrophic factor 4 (NT-4), vascular Endothelial Growth Factor (VEGF), basic fibroblast growth factor (FGF-2), insulin-like growth factor-1 (IGF-1), platelet Derived Growth Factor (PDGF), and the like.
The "inflammatory regulatory factor" refers to a protein having a biological function of regulating inflammatory reaction, and exemplified by IL-10 (interleukin-10), arg-1 (arginase-1), HILPDA (hypoxia-inducible lipid-related protein), IL-4 (interleukin-4), IL-6 (interleukin-6), IL-11 (interleukin-11), IL-13 (interleukin-13), TGF-beta (transforming growth factor-beta) and the like; preferably, the protein has a biological function of inhibiting inflammatory reaction, such as IL-10 (interleukin-10), arg-1 (arginase-1), etc.
The drug carried by the invention is nucleic acid for encoding interleukin (IL-10).
In some embodiments, the drug carried by the lipid nanoparticle composition is any one or more of an inflammatory disease drug, a tumor drug, an infectious disease drug, a metabolic disease drug, a cardiovascular disease drug, a respiratory disease, an autoimmune disease drug.
Inflammatory disease agents described herein include, but are not limited to, one or more of inflammatory bowel disease (inflammatory bowel disease, IBD), crohn's disease, ulcerative colitis, inflammatory colon cancer, chronic glomerular inflammation, chronic pyelonephritis, osteoporosis, psoriasis, sepsis.
Tumors described herein include, but are not limited to, solid tumors and hematological tumors. Hematological tumors include, but are not limited to, one or more combinations of acute myeloid leukemia (acute myelogenous leukemia, AML), acute lymphoblastic leukemia (acute lymphocytic leukemia, ALL), chronic lymphoblastic leukemia (chronic lymphocytic leukemia, CLL), multiple Myeloma (MM), hodgkin's lymphoma (hodgkin lymphoma, HL), non-hodgkin's lymphoma (non-hodgkin lymphoma, NHL). Solid tumors include, but are not limited to, one or more of adrenocortical tumors, bladder urothelial tumors, breast tumors, cervical tumors, biliary tumors, colon adenotumors, colon tumors, lymphoid tumors, esophageal tumors, glioma, squamous cell tumors, renal cell tumors, hepatoma, mesothelial cell tumors, ovarian tumors, pancreatic tumors, pheochromocytoma, paraganglioma, prostate tumors, rectal tumors, malignant sarcomas, melanoma, gastric tumors, testicular germ cell tumors, thyroid tumors, thymus tumors, endometrial tumors, myeloproliferative tumors, lung tumors, anal tumors, retinoblastoma.
The infectious diseases described herein include one or more combinations of viral infections, bacterial infections, fungal infections, parasitic infections. In some embodiments, viral infections include, but are not limited to, syncytial virus (SYNCYTIAL VIRUS, RSV) infections, atypical pneumonia virus (SARS coreavirus. SARS-COV) infections; bacterial and fungal infections including, but not limited to, salmonella infections (Salmonella typhimurium), candida albicans (candida albicans) infections; parasite infections include, but are not limited to, infection with blood sucking worm (Schistosoma mansoni) of Mannheimia, infection with trypanosoma cruzi (Trypanosoma cruzi), and infection with Leishmania (Leishmania).
Metabolic disorders described herein include, but are not limited to, one or more of insulin resistance, obesity, non-alcoholic fatty liver, cirrhosis, gout.
Cardiovascular and cerebrovascular diseases described in the invention include, but are not limited to, one or more of atherosclerosis, coronary heart disease, cerebral apoplexy, hypertension, myocardial infarction.
Respiratory diseases described herein include, but are not limited to, one or more of pneumonia, asthma (asthma, AA), chronic obstructive pulmonary disease, chronic bronchitis, and emphysema.
Autoimmune diseases described herein include, but are not limited to, one or more combinations of allergic asthma, autoimmune hepatitis (autoimmune hepatitis, AIH), rheumatoid arthritis.
The compositions of the present invention further comprise a combination of one or more of helper lipids, structural lipids, PEG-lipids, polymers.
The composition comprises the following components in percentage by mole: 20-65 ionizable lipids, 0-60 targeting group-linked ionizable lipids, 0-40 helper lipids and/or targeting group-linked helper lipids, 0-60 structural lipids and/or targeting group-linked structural lipids, 0-10 PEG-lipids and/or targeting group-linked PEG-lipids, and the molar ratio of the targeting components cannot be 0;
When the targeting component is an ionizable lipid to which the targeting moiety is attached, at least any one of a helper lipid and/or a helper lipid to which the targeting moiety is attached, a structural lipid and/or a structural lipid to which the targeting moiety is attached, a PEG-lipid and/or a PEG-lipid to which the targeting moiety is attached is other than 0;
preferred are: 20-65 ionizable lipids, 0-60 targeting group-linked ionizable lipids, 3-40 helper lipids and/or targeting group-linked helper lipids, 15-60 structural lipids and/or targeting group-linked structural lipids, 0.1-10 PEG-lipids and/or targeting group-linked PEG-lipids;
Or (b)
The composition comprises the following components in mole ratio: the molar ratio of the ionizable lipid and/or targeting group-linked ionizable lipid to the polymer and/or targeting group-linked polymer is from 0.5:1 to 80:1, preferably from 20:1 to 80:1, and most preferably from 40:1 to 80:1.
Structured lipids as described herein are meant to contain structures that stabilize the composition, including but not limited to sterols and derivatives thereof and non-sterols and derivatives thereof in combination.
In some embodiments, the structural lipids include, but are not limited to: sterols and derivatives thereof, non-sterols, sitosterols, ergosterols, cholestanones, cholestenone, campesterols, stigmasterols, brassicasterol, lycosyline, ursolic acid, fecal sterols, alpha-tocopherols or corticosteroids. Sterols as a preferred cholesterol and derivatives thereof; non-limiting examples of cholesterol derivatives include: such as 5α -cholesterol, 5α -fecal sterols, cholesteryl- (2 '-hydroxy) -ethyl ether, cholesteryl- (4' -hydroxy) -butyl ether and 6-ketocholestanol; nonpolar analogs such as 5 alpha-cholestane, cholestenone, 5 alpha-cholestenone and capric cholesterol ester; and mixtures thereof. In a preferred embodiment, the cholesterol derivative is a polar analogue, such as cholesteryl- (4' -hydroxy) -butyl ether. It is not intended to be exhaustive and any structural lipid may be used in the present invention.
In some embodiments, the structural lipid is a combination of one or more of cholesterol, sitosterol, ergosterol, corticosteroid, and derivatives thereof.
In some embodiments, the structural lipid is cholesterol.
The "helper lipids" of the present invention are not limited in kind, and preferably phospholipid lipids, including but not limited to: one or more combinations of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin, ceramide, phosphatidylserine, phosphatidylinositol, phosphatidic acid, phosphatidylglycerol, dimyristoyl phosphatidylglycerol.
In some embodiments, the helper lipid may be selected from: 1, 2-dioleoyl-sn-glycero 3-phosphoethanolamine (DOPE), 1, 2-dioleoyl-sn-glycero 3-phosphocholine (DOPC), 1, 2-dioleoyl-sn-glycero 3-phosphocholine (DLPC), 1, 2-dimyristoyl-sn-glycero phosphocholine (DMPC), 1, 2-dioleoyl-sn-glycero 3-phosphocholine (DPPC), 1, 2-distearoyl-sn-glycero 3-phosphocholine (DSPC), 1, 2-dioleoyl-sn-glycero 3-phosphocholine (DUPC), 1, 2-dioleoyl-sn-glycero 3-phosphocholine (POPC), 1, 2-dioleoyl-sn-glycero 3-phosphocholine (18:0 diether PC), dimyristoyl phosphoethanolamine (DMPE), distearoyl-phosphatidylethanolamine (DSPE), 1,2E), 1, 2-dioleoyl-sn-glycero 3-phosphocholine (DPPC), 1-palmitoyl-2-oleoyl-sn-glycero 3-phosphocholine (DPPC), 1, 2-di-linolenoyl-sn-glycero-3-phosphocholine, 1, 2-di-arachidonoyl-sn-glycero-3-phosphocholine, 1, 2-di-dodecanoyl-sn-glycero-3-phosphocholine, 1, 2-di-phytoyl-sn-glycero-3-phosphoethanolamine, 1, 2-di-stearoyl-sn-glycero-3-phosphoethanolamine, 1, 2-di-linolenoyl-sn-glycero-3-phosphoethanolamine, 1, 2-di-arachidonoyl-sn-glycero-3-phosphoethanolamine, 1, 2-di-dodecanoyl-sn-glycero-3-phosphoethanolamine, 1, 2-di-oleoyl-sn-glycero-3-phospho-rac- (1-glycero) sodium salt (DOPG), diacetyl-phosphoethanolamine (DEPE), stearoyl-phosphoethanolamine (SOPE), lysophosphatidylcholine, or combinations of one or more thereof.
In some embodiments, the phosphatidylcholine is a combination of one or more of DSPC, DPPC, DMPC, DOPC, POPC.
In some embodiments, the helper lipid is phosphatidylcholine, particularly DSPC.
In some embodiments, the helper lipid is phosphatidylethanolamine, particularly DOPE.
In some embodiments, the helper lipid is selected from one or more combinations of DOTAP ((1, 2-dioleoxypropyl) trimethylammonium chloride), DOTAP (1, 2-dioleoyl-3-dimethylammonium-propane), 18:1pa (1, 2-DI (cis-9-octadecenoyl) -SN-glycero 3-phosphate sodium salt), HS15 (polyethylene glycol (15) -hydroxystearate), GL67 (N4-argininocarbonamide).
The PEG-lipid of the present invention generally refers to a conjugate formed by linking PEG (polyethylene glycol) and a lipid molecule through a chemical bond. Including but not limited to PEG-modified phospholipids and derivatized lipids, exemplified by combinations of one or more of PEG-modified phosphatidylethanolamine, PEG-modified phosphatidic acid, PEG-modified ceramide, PEG-modified dialkylamine, PEG-modified diacylglycerol, PEG-modified dialkylglycerol.
In certain embodiments of the invention, the PEG in the PEG-lipid has a weight average molecular weight of 1000 to 10000, for example 1000 to 2000, 2000 to 4000, 4000 to 6000, 6000 to 8000, 8000 to 10000, preferably 2000.
In some embodiments, the PEG-lipid includes, but is not limited to, PEG-C-DMG, PEG-C-DOMG, PEG-DLPE, PEG-DMPE, PEG-DPPE, PEG-DOPE, PEG-DPPC, PEG-distearoyl phosphatidylethanolamine (PEG-DSPE), PEG-DS, chol (cholesterol) -PEG, 1, 2-dimyristoyl-rac-glycerol-3-methoxypolyethylene glycol (PEG-DMG), PEG-S-DMG, polyethylene glycol phosphatidylethanolamine, polyethylene glycol ceramide, polyethylene glycol dimethacrylate (PEG-DMA), PEG distearyl glycerol, PEG dipalmitoyl, PEG dioleyl, PEG distearyl, PEG diacyl Gan Xianan, PEG dipalmitoyl phosphatidylethanolamine, PEG-phosphatidylethyldimyristoxypropyl-3-amine, PEG oxypropyl-1, 2-distearoyloxypropyl-3-amine-N [ methoxy (PEG-DSA) ] (PEG-DSA), polyethylene glycol, methoxy-co-Acetyl (ALC), or a combination of one or more of lauric Acid and (ALC) of the group (ALC) of lauric Acid and (ALC) of the above.
In some embodiments, the PEG-lipid is PEG-DMG.
The kind of the "polymer" according to the present invention is not limited, and the polymer may include, but is not limited to, amphiphilic block copolymers, which are block copolymers composed of hydrophobic polymers and hydrophilic compounds, including, but not limited to, polylactic acid (PLA), polylactic acid-polyglycolic acid copolymer (PLGA), glycolide-lactide copolymer (PLCG), polycaprolactone (PCL), polyorthoester, polyanhydride (PAH), polyphosphazene, poly beta Polyaminoester (PBAE), poly (polyhydroxyb acid), lactide/glycolide copolymer (PLGA or PLG) (which includes lactide/glycolide copolymer, D-lactide/glycolide copolymer, L-lactide/glycolide copolymer and D, L-lactide/glycolide copolymer), polyglycolide (PGA), polyorthoester (POE), linear or branched polyethylene glycol (PEG), conjugates of poly (E hydroxy acid), polyacetin (polyaspirins), polyphosphazenes, D-lactide, D, L-lactide-caprolactone, D, L-lactide-glycolide-caprolactone, dextran, vinylpyrrolidone, polyvinyl alcohol (PVA), methacrylate, poly (N-isopropylenamide), SAIB (sucrose acetate isoparaffinate) hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl methylcellulose, carboxymethyl cellulose or salts thereof, carbopol, poly (hydroxyethyl methacrylate), poly (methoxyethyl methacrylate), poly (methoxyethoxy-ethyl methacrylate), polymethyl methacrylate (PMMA), methyl Methacrylate (MMA), PVA-g-PLGA, PEGT-PBT copolymers, PEO-PPO-PEO (pluronics), PEO-PPO-PAA copolymers, PLGA-PEO-PLGA, PEG-PLG, PLA-PLGA, poloxamer 407, PEG-PLGA-PEG triblock copolymers, PEG-PLA-PEG triblock copolymers, PEG-PCL-PEG triblock copolymers, or block copolymers thereof with polyethylene glycol (PEG), or combinations of one or more of the foregoing polymers or copolymers.
In some embodiments, the molar ratio of the ionizable lipid to the targeting component is as follows: 20-65 ionizable lipids, 0-60 targeting group-linked ionizable lipids, 3-40 helper lipids and/or targeting group-linked helper lipids, 20-60 structural lipids and/or targeting group-linked structural lipids, 0.1-10 PEG-lipids and/or targeting group-linked PEG-lipids.
Further preferably, the composition of the present invention further comprises pharmaceutically acceptable excipients. Typically, these materials are formulated in a nontoxic, inert and pharmaceutically acceptable aqueous carrier medium, wherein the pH is typically about 4 to 8, preferably about 5 to 7, although the pH may vary depending on the nature of the material being formulated and the condition being treated. The formulated pharmaceutical compositions may be administered by conventional routes including, but not limited to: intravenous injection, intravenous drip, subcutaneous injection, topical injection, intramuscular injection, intratumoral injection, intraperitoneal injection (e.g., intraperitoneal), intracranial injection, intracavity injection, inhalation administration, implantation administration, and the like.
By "pharmaceutically acceptable" is meant that the drug does not produce adverse, allergic or other untoward reactions when properly administered to an animal or human.
The "pharmaceutically acceptable excipients" should be compatible with the active ingredient, i.e. capable of being blended therewith without substantially reducing the efficacy of the drug in the usual manner. Specific examples of some substances which can be pharmaceutically acceptable excipients may be sugars, such as glucose, mannitol, sucrose, lactose, trehalose, maltose, etc.; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium methyl cellulose, ethyl cellulose, methyl cellulose, etc.; tragacanth powder; malt; gelatin; talc; solid lubricants such as stearic acid and magnesium stearate; calcium sulfate; vegetable oils such as peanut oil, cottonseed oil, sesame oil, olive oil, corn oil, and cocoa butter; alcohols such as ethanol, propylene glycol, glycerin, sorbitol, mannitol, polyethylene glycol and the like; alginic acid; emulsifying agents, such as Tween and the like; wetting agents such as sodium lauryl sulfate and the like; a surfactant; a lyoprotectant; a colorant; a flavoring agent; tabletting; a stabilizer; a diluent; an excipient; an antioxidant; a preservative; non-thermal raw water; isotonic saline solution; buffers, and the like, as well as combinations thereof. These substances are used as needed to increase the stability of the formulation or to help increase the activity or its bioavailability or to create an acceptable mouthfeel or odor in the case of oral administration.
Diluents are any pharmaceutically acceptable water-soluble excipients known to those skilled in the art, including: amino acids, monosaccharides, disaccharides, trisaccharides, tetrasaccharides, pentasaccharides, other oligosaccharides, mannitol, dextran, sodium chloride, sorbitol, polyethylene glycol, phosphates, or derivatives thereof, and the like.
The stabilizer can be any pharmaceutically acceptable auxiliary material known to those skilled in the art: tween-80, sodium dodecyl sulfate, sodium oleate, mannitol, mannose or sodium alginate, etc.
The preservative may be any pharmaceutically acceptable preservative known to those skilled in the art, such as: thiomerosal, and the like.
The lyoprotectant may be any pharmaceutically acceptable lyoprotectant known to those skilled in the art, such as: glucose, mannitol, sucrose, lactose, trehalose, maltose, and the like.
The composition of the present invention may be formulated into inhalable atomized formulations (e.g., dry powder formulations, aerosol formulations, inhalable aerosol droplet formulations, etc.), implantable gel formulations, microneedle formulations, and may also be formulated into injectable forms, for example, using physiological saline or aqueous solutions containing glucose and other adjuvants by conventional methods. The pharmaceutical compositions, such as injections, solutions are preferably manufactured under sterile conditions. The amount of active ingredient administered is a therapeutically effective amount, for example, from about 10 micrograms per kilogram of body weight to about 50 milligrams per kilogram of body weight per day.
Compared with the prior art, the invention has the beneficial effects that at least:
1. The targeting LNP prepared by the invention has targeting and synergistic effects, promotes more macrophage M1 type to M2 type polarization, forms a closed loop with positive feedback mechanism, recruits more nano particles to be enriched at lesion sites, comprehensively improves transfer efficiency and transfection efficiency, and realizes more efficient and stable treatment effect.
2. The mRNA-LNP targeting drug prepared by the invention has smaller size, can penetrate through the damaged blood brain barrier BBB and targets microglial cells of the central nervous system.
3. The targeting LNP prepared by the invention can efficiently deliver mRNA encoding IL-10, improve inflammation of pathological tissues and repair injury and functions of the pathological tissues.
4. The targeting LNP prepared by the invention can efficiently deliver mRNA encoding IL-10 for treating IS, can expand the treatment window duration of IS from 4-24 hours in clinic to at least 72 hours, enables more patients to receive thrombolysis or interventional therapy, and has very important clinical application value.
5. The targeting LNP prepared by the invention can efficiently deliver mRNA encoding IL-10 for treating AS, can relieve inflammation of AS vascular intima, reduce plaque necrosis core area, reduce fat deposition and increase plaque stability.
The invention will be further illustrated with reference to specific examples. It is to be understood that these examples are illustrative of the present invention and are not intended to limit the scope of the present invention.
It is also to be understood that the terminology used in the examples of the invention is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the invention; in the description and claims of the invention, the singular forms "a", "an" and "the" include plural referents unless the context clearly dictates otherwise. Where numerical ranges are provided in the examples, it is understood that unless otherwise stated herein, both endpoints of each numerical range and any number between the two endpoints are significant both in the numerical range. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
The experimental procedure, which does not address the specific conditions in the examples below, is generally followed by routine conditions, such as, for example, sambrook et al, molecular cloning: conditions described in the laboratory Manual (New York: cold Spring Harbor Laboratory Press, 1989) or as recommended by the manufacturer. Percentages and parts are weight percentages and parts unless otherwise indicated.
English abbreviations interpretation:
NPs refer to nanoparticles;
HNPs refers to nanoparticles in which the amphiphilic block copolymer is PLGA-PEG:
CALB (CANDIDA ANTARCTICA LIPASE B) is candida utilis lipase B;
HATU (2- (7-Azabenzotriazol-1-yl) -N, N '-tetramethyluronium hexafluorophosphate) refers to the polypeptide condensing reagent 2- (7-azabenzotriazol) -N, N' -tetramethylurea hexafluorophosphate;
DMF (N, N-Dimethylformamide) refers to N, N-Dimethylformamide;
DIEA (Diisopropylethylamine) refers to N, N-diisopropylethylamine;
MAN (4-Aminophenyl alpha-Dmannopyranoside) refers to 4-methoxyphenyl- α -D-mannopyranoside;
Mannose refers to mannose
LPS (Lipopolysaccharide) refers to lipopolysaccharide;
TNF-alpha (Tumor necrosis factor alpha) refers to tumor necrosis factor alpha;
dMCAO (DISTAL MIDDLE cerebral artery occlusion) refers to distal middle cerebral artery occlusion;
IVIS (Interactive Video Information System) refers to a living animal optical imaging system;
BBB refers to Blood-brain barrier;
PEG (Polyethylene glycol) is polyethylene glycol
IL-10 refers to the anti-inflammatory factor interleukin-10
BMDMs (Bone-marrow-derived macrophages) refers to primary Bone marrow-derived macrophages;
PBS (Phosphate Buffer Saline) refers to phosphate buffer solution;
Ldlr -/- refers to a transgenic mouse model of low density lipoprotein receptor knockout;
WD refers to western diet;
IS refers to ischemic stroke;
AS refers to atherosclerosis.
The general method comprises the following steps:
Preparation of modified mIL-10
In vitro transcription was performed using the mouse IL-10 plasmid as DNA template and T7 RNA polymerase, and anti-reverse cap 0 analogue (ARCA, APExBIO) and pseudouracil-5 '-triphosphate (ψTP, APExBIO) were added to the reaction instead of uridine-5' -triphosphate (UTP). RNA Clean & Concentrator purified RNA, nanoDrop 2000 phosphometer was quantified and stored in a-80℃refrigerator for use.
Synthesis of DMG-PEG-mannose (Mannose)
N 3 -PEG-COOH (50 mg,0.025 mmol) and HATU (48 mg,0.125 mmol) were dissolved in DMF (1.5 mL) and DIEA (44. Mu.L, 0.25 mmol) was added for reaction. Mannose MAN (8.1 mg,0.030 mmol) was added to the reaction solution and stirred at room temperature for 24 hours. The product was reacted with 3- (prop-2-yn-1-yloxy) propane-1, 2-dialkyl-dicetyl (41 mg,0.075 mmol) in the presence of CuI (5 mg,0.025 mmol) catalyst at room temperature for 24h. Acetic acid (50. Mu.L) was added to quench the reaction, and methanol (5 mL) was used to dilute the reaction. The product DMG-PEG-Mannose (white solid, 15.8 mg) was purified by Prep-HPLC. And characterized using nuclear magnetic resonance hydrogen spectroscopy.
1H NMR(400MHz,Chloroform-d)δ7.73(s,1H),7.52(d,J=8.8Hz,2H),7.01(d,J=8.8Hz,2H),4.17-3.22(m,126H),1.41-1.11(m,48H),0.85(t,J=6.7Hz,6H).
LCMS(ESI+):m/z 948.7±14.7[1/3M+1]+
MRNA LNPs Synthesis of (Targeted lipid nanoparticles)
AA3-DLin (commercially available source, CAS: 2832061-33-1), DOPE, cholesterol, and DMG-PEG-Mannose were dissolved in 42.7:34.2:21.4:1.7 molar ratio in ethanol phase, mRNA was dissolved in 25mM sodium acetate buffer (pH=5.0) as aqueous phase. The aqueous and ethanol organic phases were mixed in a mRNA/AA3-DLin weight ratio of 1:20 using a microfluidic system. The mixture was loaded into a 3500MWCO cassette and dialyzed against PBS (10 mm, ph=7.4) at 4 ℃ for 24 hours to form stable mRNA LNPs. The fluorescence-labeled LNP used in the examples, whose cholesterol component is labeled with Cy5, can be used for cellular uptake and biodistribution studies. The physicochemical properties of mRNA LNPs, including size, zeta potential and morphology, were examined using dynamic light scattering and transmission electron microscopy.
Isolation of primary microglia and Synthesis of M2 phenotype microglia
Cell source: new-born C57BL/6 mice
Brain tissue, except cerebellum, olfactory bulb and meninges, was minced and digested in 0.25% Trypsin-EDTA and 125U/mL DNase for 15 min at 37 ℃. Complete medium containing 10% FBS was then added to stop digestion, 400g, and centrifuged for 5 minutes. After centrifugation, the pellet was resuspended in complete medium and incubated in an incubator (5% CO 2, 37 ℃). After 14 days of culture, astrocytes reached 100% confluency and were firmly attached to the bottom of the flask, while the microglial subpopulations were loosely attached to the surface layer, which was isolated after shaking the flask at 200rpm for 4 hours. After 48 hours of primary microglial treatment with 20ng/mL IL-4, the expression of CD206 (a biomarker for M2 microglial cells) was examined by immunofluorescence to characterize differentiation.
Cellular uptake
Both microglia and astrocytes were treated with 20ng/mL IL-4 for 48 hours. Cells were incubated with Cy5 LNPs (1. Mu.g/mL mRNA) entrapped mIL-10 for 4 hours. Fluorescence signals were quantitatively analyzed using a flow cytometer and further observed using CLSM.
MRNA MLNPS transfection efficiency in vitro
Microglia cells 1X 10 5 were seeded on 24-well plates and transfected at 1. Mu.g/mL mRNA levels of NAKED MEGFP, MLNPs and mEGFP@M-HNPs for 24 hours. Transfection was performed using Lipofectamine3000 according to the manufacturer's instructions as a positive control. Cells were then collected to analyze MFI by flow cytometry.
In vitro anti-inflammatory activity evaluation.
Microglia and BV2 cells were seeded in 6-well plates, respectively, with 1X 10 6 cells per well. Cells were first stimulated with 500ng/mL LPS for 4 hours and then incubated with fresh medium or mIL-10@MLNPs (1. Mu.g/mL) for 24 hours. Total RNA was extracted using Trizol and IL-10, TNF- α, iNOS, IL-6, arg-1, TGF- β or CD206 gene levels were measured using RT-qPCR.
Construction of permanent MCAO (dMCAO) model
Mice were anesthetized under spontaneous breathing conditions with 2% isoflurane in a 30% O 2/68%N2 O mixture. The neck skin was incised, exposing the left common carotid artery and ligating. After the neck incision is sutured, a skin incision is made between the left eye and the left ear. The temporal muscle was dissected and a burr hole was made to expose the distal end of Middle Cerebral Artery Occlusion (MCAO). The dura was then dissected and coagulated with low intensity bipolar electrocautery directly outside the nasal fissures. Sham animals received anesthesia and were surgically exposed to the arteries, but without arterial occlusion. Mice were dosed with PBS, MLNPs and mIL-10@MLNPs, at 0.3mg/kg mRNA on days 3 and 7, respectively, after dMCAO. And sacrificed 7 days after the last injection to evaluate the effect of the treatment. Mice were sacrificed by high CO 2 asphyxiation, and the number of animals operated was determined according to laboratory preliminary experiments to minimize pain and use of the animals.
Immunofluorescent staining
Sections were blocked with 3% bsa for 30min, primary antibody incubated overnight at 4 ℃, followed by incubation with appropriate fluorescent-labeled secondary antibody for 60 min at Room Temperature (RT), DAPI counterstain for 5 min. Histological images were collected with a microscope or CLSM and quantitatively analyzed with ImageJ software. Immune positive cell counts are expressed as an average percentage of each field; three fields were randomly selected from three consecutive sections of each brain tissue cortex by a researcher for analysis.
Garcia score
The modified Garcia score is a complete sensorimotor assessment system consisting of five separate tests, one measuring sensory function and four measuring motor function. Each test score was from 0 to 3 points (highest score was 15 points), including physical proprioception, forelimb walking, limb symmetry, lateral rotation, and climbing. Researchers blinded to the experimental assignments in the experiments completed the preoperative and postoperative sensorimotor function assessment.
In vivo safety assessment
C56BL/6 Male mice were injected with PBS, MLNPs or mIL-10@MLNPs, respectively, twice a week for one week with a dose of 0.3mg/kg mRNA. One week after the last injection, blood and major organ samples were collected from each treatment group. Histological evaluation of H & E staining was performed on heart, liver, spleen, lung, kidney and brain. Meanwhile, blood samples were analyzed for hematology and biochemistry.
Statistical analysis
Statistically, normalization was determined using Shapiro-Wilk and/or Kolmogorov-Smirnov normalization tests. When a normal distribution is determined, the multiple sets of comparisons employ one-way analysis of variance (ANOVA). The Kruskal-Wall test calculates the P value of the non-normal distribution data. The differences between time points of the different groups were analyzed by two-factor analysis of variance and then by Bonferroni test. P <0.05 is considered statistically significant. All statistics in the graph were rated according to the following criteria: * P <0.05, < P <0.01, < P <0.001, < P < 0.0001. v8.2.1 all statistical analyses were performed using GRAPHPAD PRISM software.
Example 1 mRNA MLNPs evaluation of stability
Mu.g of free mIL-10, mIL-10@LNPs or mIL-10@MLNPs were incubated with 10% FBS (fetal bovine serum, v/v) at 37℃and 100rpm, respectively, for 24 hours. mRNA was extracted from LNPs using 50mg/mL heparin sodium, and samples were analyzed by electrophoresis using a 2% agarose gel and visualized by the ChemiDoc system. At predetermined time points, the particle size and aggregation state of mRNA LNPs were analyzed at 660nm using Dynamic Light Scattering (DLS) and a microplate reader, and repeated 3 times.
FIG. 1A shows that mRNA encapsulated in LNPs still shows excellent stability after 24 hours of exposure to 10% FBS, but the free form of mRNA has degraded rapidly. Fig. 1B shows no significant change in particle size over 24 hours. Turbidity measurement experiments demonstrated that no serum-induced LNPs aggregation occurred (see figure 1C). Both targeted MLNPs and non-targeted LNPs showed good haemocompatibility with less than 5% haemolysis (see figure 1D).
In short, MLNPs of the present invention has a stable nanostructure, which can protect the mRNA encapsulated therein from degradation.
Example 2 mIL-10@MLNP targeting evaluation
Microglia and astrocytes were each inoculated in 24-well plates (1X 10 5 cells per well) overnight and treated with 20ng/ml IL-4 for 48 hours. Cells were incubated with mIL-10@ Cy5 LNPs or mIL-10@ Cy5 MLNPs at an mRNA concentration of 1. Mu.g/ml for 4 hours. The fluorescent signal was quantitatively analyzed by a flow cytometer, and the fluorescent signal was visualized by CLSM.
The CLSM images and flow cytometry results showed enhanced uptake of ml10@ Cy5 MLNP by M2 microglia overexpressing CD206 (see fig. 2A), but this phenomenon of enhanced uptake was not observed in astrocytes not overexpressing CD206 (see fig. 2B).
More of the mIL-10@ Cy5 MLNP co-localizes with the M2 microglial surface expressed CD206 than does the non-targeted mIL-10@ Cy5 LNP (see FIG. 2C).
In conclusion, mIL-10@ Cy5 MLNPs can be specifically ingested by microglia, and has certain targeting property.
Example 3 in vitro evaluation of mRNA MLNPs transfection
The transfection efficiency of MRNA MLNPS in microglia was assessed using mRNA encoding the enhanced green fluorescent protein (mEGFP) as model mRNA. Microglia cells were seeded at a cell density of 1X 10 5 on 24 well plates and cells were transfected with free mEGFP, MLNPs and mEGFP@M-HNPs at an mRNA concentration of 1. Mu.g/ml for 24 hours. Positive control was lipofectamine 3000 (L3K) reagent and microglial cells were transfected according to the manufacturer's protocol. Cells were collected and flow cytometry analyzed for Mean Fluorescence Intensity (MFI).
The results showed that the MFI of microglia treated with mEGFP@MLNPs was higher than that of microglia treated with L3K (see FIG. 3A).
Example 4 evaluation of anti-inflammatory Effect of mIL-10@MLNPs in vitro
Transfection experiments were performed in primary microglial and BV2 cell lines extracted from mice to evaluate the effect of IL-10 expression and the anti-inflammatory effect mediated by mll-10@mlnps. Primary microglia and BV2 cell lines were seeded into 6-well plates at a density of 1 x 10 6 cells per well, respectively, and incubated overnight. Cells were stimulated with 500ng/ml LPS for 4 hours and incubated with fresh medium or mIL-10@MLNPs (1. Mu.g/ml) for 24 hours. Untreated microglia were used as negative controls. Trizol extracts total RNA and quantitative reverse transcription PCR (qRT-PCR) detects gene levels of inflammatory factors (IL-10, TNF-alpha, iNOS, IL-6, arg-1, TGF-beta, CD 206).
QRT-PCR results showed that the mIL-10@MLNPs treatment significantly increased the IL-10 gene expression level (see FIG. 3B and FIG. 4). The mll-10@mlnp treatment significantly inhibited TNF- α production in LPS-stimulated microglia and slightly increased the expression level of CD206, which may be the result of efficient production of bioactive IL-10 through MLNP-mediated mRNA delivery (see figures 3C and D). The reduced expression of iNOS and IL-6 in LPS-stimulated BV2 cells following mll-10@mlnp treatment further demonstrated this potent anti-inflammatory effect (see figure 4). Furthermore, MLNP treatment significantly enhanced the production of the protective and trophic factors Arg-1 and TGF- β in LPS-stimulated microglia, indicating a transition of microglia to the anti-inflammatory M2 phenotype (see figures 3E and F and figure 4).
EXAMPLE 5mIL-10@MLNP improvement of blood brain barrier injury and neurological deficit in the dMCAO mouse model
The dMCAO model is another commonly used ischemic stroke model, resulting in permanent cerebral cortex damage, similar to that observed in ischemic stroke patients who have not received reperfusion therapy. Therefore, dMCAO mouse models were developed to evaluate the therapeutic effect of mIL-10@MLNPs on neurological dysfunction caused by permanent cerebral ischemia. Mice were injected intravenously with the indicated therapeutic drug on days 3 and 7, respectively, following dMCAO surgery (fig. 5A). To assess BBB destruction associated with ischemia reperfusion injury following dMCAO, brain sections were stained to detect endogenous plasma IgG, and the green fluorescent areas shown indicate extravasation of blood components into brain tissue. The mll-10@mlnp treatment significantly improved BBB disruption, i.e. a decrease in green fluorescent signal corresponding to IgG (fig. 5B and C). Furthermore, microtubule-associated protein-2 (MAP-2) staining showed that mIL-10@MLNPs significantly reduced the mean infarct volume of the brain compared to the control group (FIGS. 5B and D). The neurological deficit of the permanently cerebral ischemic mice was then assessed using Garcia score. The results showed that the total nerve evaluation score was significantly higher in mice treated with mll-10@mlnp over 14 days post-stroke than in the control group (fig. 5E). These results demonstrate that mIL-10@MLNP can exert neuroprotection by improving sensorimotor nerve function after stroke.
EXAMPLE 6 in vivo safety assessment
Male C56BL/6 mice were injected intravenously with PBS, MLNPs or mIL-10@MLNPs twice a week for one week at an injection dose of 0.3mg/kg mRNA per mouse. Blood and major organ samples were collected from each treatment group 7 days after injection. Histological examination of heart, liver, spleen, lung, kidney and brain was performed using hematoxylin and eosin (H & E) staining. Blood samples were analyzed for hematological and biochemical indicators, including red blood cell count (RBC), white blood cell count (WBC), hemoglobin (HGB), hematocrit (HCT), mean red blood cell volume (MCV), mean red blood cell hemoglobin (MCH), mean red blood cell hemoglobin concentration (MCHC), lymphocytes, alanine Aminotransferase (ALT), aspartate Aminotransferase (AST), total Protein (TP), albumin (ALB), triglycerides (TG), low Density Lipoprotein (LDL), total Cholesterol (TC), creatinine (Crea), urea nitrogen (BUN), and glucose (Glu).
Major organ H & E staining of mice in the ml10@mlnp treated group showed no evidence of significant damage to heart, liver, spleen, lung, kidney and brain (fig. 6). Furthermore, there was no significant difference in the hematology and biochemistry indices of the 3 treatment groups (fig. 6). These evidence strongly suggest that MLNP vectors are safe and nontoxic.
EXAMPLE 7 therapeutic Effect of AS mice
C57BL/6 mice with 6-8 week old male low density lipoprotein receptor gene knockout (Ldlr-/-) were purchased from Nanjing Jieqin-Jieqin. The atherosclerosis mouse model was established by Western Diet (WD) induction for 12 weeks.
This experiment evaluates the protective effect of IL-10mRNA-LNP on late-medium AS by using WD-induced Ldlr-/-AS mouse model. The mice model was randomly divided into PBS group (blank control group) and IL-10 mRNA-LNPs group (treatment group), and the administration dose was such that each mouse was given nanoparticles containing 10. Mu.g mRNA, and the mice were given by intravenous injection twice a week for four weeks.
The results show that the brachiocephalic artery, aortic arch and aortic root of the placebo mice show higher ORO positive areas. In contrast, the plaque areas in the brachiocephalic artery, aortic arch and aortic root were significantly reduced in IL-10mRNA-LNP treated mice.
All documents mentioned in this disclosure are incorporated by reference in this disclosure as if each were individually incorporated by reference. Further, it will be appreciated that various changes and modifications may be made by those skilled in the art after reading the above teachings, and such equivalents are intended to fall within the scope of the application as defined in the appended claims.

Claims (12)

1. A targeted lipid nanoparticle composition comprising: an ionizable lipid and a targeting ingredient; and the ionizable lipid is AA3-Dlin; wherein the method comprises the steps of
The targeting component is selected from any one or a combination of a plurality of ionizable lipid connected with a targeting group, structural lipid connected with the targeting group, auxiliary lipid connected with the targeting group, PEG-lipid connected with the targeting group and polymer connected with the targeting group;
the targeting group is selected from the group consisting of: mannose, mannose derivatives, galactose derivatives, dextran derivatives, peptide fragments or any one or a combination of a plurality thereof.
2. The lipid nanoparticle composition of claim 1, wherein the derivative is a glycoside, a sugar amine, a glycan, a PEG-modified sugar ring structure, an isotopic or other substituent-substituted sugar ring structure; preferred derivatives are combinations of one or more of methyl-D-mannoside, 1-alpha formylmethyl-mannopyranoside, 4-aminophenyl-alpha-D-mannopyranoside, 4-nitrophenyl-alpha-D-mannopyranoside, 4-methylumbelliferone-alpha-D-mannopyranoside, mannose-6-phosphate, carbamoyl-D-mannose, N-acetamido-beta-1, 2-mannose, methyl 6-O- (aD-mannopyranosyl) -aD-mannopyranoside, methyl 3-O- (aD-mannopyranosyl) -aD-mannopyranoside, mannosamine, mannan, and the like.
3. The lipid nanoparticle composition of claim 1, wherein the composition further comprises a combination of one or more of a helper lipid, a structural lipid, a PEG-lipid, and a polymer.
4. A lipid nanoparticle composition according to claim 3, wherein the composition comprises the following components in mole percent: 20-65 ionizable lipids, 0-60 targeting group-linked ionizable lipids, 0-40 helper lipids and/or targeting group-linked helper lipids, 0-60 structural lipids and/or targeting group-linked structural lipids, 0-10 PEG-lipids and/or targeting group-linked PEG-lipids, and the molar ratio of the targeting components cannot be 0; when the targeting component is an ionizable lipid to which the targeting moiety is attached, at least any one of a helper lipid and/or a helper lipid to which the targeting moiety is attached, a structural lipid and/or a structural lipid to which the targeting moiety is attached, a PEG-lipid and/or a PEG-lipid to which the targeting moiety is attached is other than 0; preferred are: 20-65 ionizable lipids, 0-60 targeting group-linked ionizable lipids, 3-40 helper lipids and/or targeting group-linked helper lipids, 15-60 structural lipids and/or targeting group-linked structural lipids, 0.1-10 PEG-lipids and/or targeting group-linked PEG-lipids; or, the molar ratio of the ionizable lipid and/or the targeting group-linked ionizable lipid to the polymer and/or the targeting group-linked polymer is from 0.5:1 to 80:1, preferably from 20:1 to 80:1, and most preferably from 40:1 to 80:1.
5. The lipid nanoparticle composition of claim 1, wherein the targeting adjuvant has a structure according to the formula:
Wherein:
r is selected from any one or a combination of mannose, mannose derivatives, galactose derivatives, dextran derivatives; n is selected from integers from 22 to 220.
6. The lipid nanoparticle composition of claim 1, wherein the composition further comprises: a drug carried; preferably, the composition further comprises pharmaceutically acceptable excipients.
7. The lipid nanoparticle composition of claim 6, wherein the drug loaded comprises: any one or a combination of nucleic acids, small molecules, proteins.
8. The nanoparticle composition of claim 7, wherein the drug loaded is a nucleic acid encoding one or more proteins having phenotypic modulation, neurotrophic, inflammatory modulation; preferably, the nucleic acid encodes one or more phenotype-modulating associated factors, neurotrophic factors, or inflammatory mediators, preferably, the nucleic acid encodes an anti-inflammatory factor, and more preferably, the nucleic acid encodes IL-10.
9. The lipid nanoparticle composition of claim 8, wherein the nucleic acid encodes an amino acid sequence of IL-10 selected from the group consisting of seq id no: a sequence as set forth in SEQ ID NO. 1 or 2, a sequence having at least 80% identity to the sequence set forth in SEQ ID NO. 1 or 2, or an amino acid sequence having 1 or more conservative substitutions as compared to the sequence set forth in SEQ ID NO. 1 or 2.
10. The lipid nanoparticle composition of claim 8, wherein the nucleic acid is a messenger ribonucleic acid encoding IL-10 (IL-10 mrna, mll-10); and the mRNA encoding IL-10 comprises a sequence as set forth in SEQ ID NO. 6 or 7 or 8, a sequence having at least 80% identity to the sequence set forth in SEQ ID NO. 6 or 7 or 8, or a nucleic acid sequence having 1 or more conservative substitutions as compared to the sequence set forth in SEQ ID NO. 6 or 7 or 8; preferably, the method comprises the steps of,
MRNA encoding IL-10 comprises capping elements, 5 'UTRs, signal peptide sequences, open reading frames, 3' UTRs, and/or polyAs; more preferably, the capping structure comprises 3'-O-Me-m7G (5') ppp (5 ') G linked to a 5' utr;
More preferably, the 5' UTR comprises a sequence as shown at positions 1 to 47 in SEQ ID NO. 9 or 10, a sequence having at least 80% identity to the sequence shown at positions 1 to 47 in SEQ ID NO. 9 or 10, or a nucleic acid sequence having 1 or more conservative substitutions as compared to the sequence shown at positions 1 to 47 in SEQ ID NO. 9 or 10;
The signal peptide sequence comprises a sequence as shown in SEQ ID NO. 9 at positions 48 to 101, a sequence having at least 80% identity to the sequence shown in SEQ ID NO. 7 at positions 48 to 101, or a nucleic acid sequence having 1 or more conservative substitutions as compared to the sequence shown in SEQ ID NO. 7 at positions 48 to 101;
The open reading frame comprises a sequence as set forth in SEQ ID NO. 9 or 11 at positions 102 to 584, a sequence having at least 80% identity to the sequence set forth in SEQ ID NO. 9 at positions 102 to 584, or a nucleic acid sequence having 1 or more conservative substitutions as compared to the sequence set forth in SEQ ID NO. 9 at positions 102 to 584;
The 3' UTR comprises a sequence as shown in SEQ ID NO. 9 from 585 to 677, a sequence having at least 80% identity to the sequence shown in SEQ ID NO. 9 from 585 to 677, or a nucleic acid sequence having 1 or more conservative substitutions as compared to the sequence shown in SEQ ID NO. 9 from 585 to 677;
the nucleic acid sequence of polyA comprises 65-250A;
Optimally, the mRNA encoding IL-10 comprises a sequence as set forth in any of SEQ ID NOS: 9-12 having at least 80% identity to the sequence set forth in any of SEQ ID NOS: 9-12 or 1 or more conservatively substituted nucleic acid sequences as compared to the sequence set forth in any of SEQ ID NOS: 9-12.
11. Use of the lipid nanoparticle composition of claim 1 for the preparation of a macrophage-targeted drug.
12. The use according to claim 11, wherein the medicament is any one or a combination of more of an inflammatory disease medicament, a oncological medicament, an infectious disease medicament, a metabolic disease medicament, a cardiovascular disease medicament, a respiratory disease, an autoimmune disease; preferably, the medicament is a medicament for the treatment of atherosclerosis and/or stroke.
CN202410003683.XA 2024-01-02 2024-01-02 Targeting lipid nanoparticle and preparation and application thereof Pending CN117899048A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202410003683.XA CN117899048A (en) 2024-01-02 2024-01-02 Targeting lipid nanoparticle and preparation and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202410003683.XA CN117899048A (en) 2024-01-02 2024-01-02 Targeting lipid nanoparticle and preparation and application thereof

Publications (1)

Publication Number Publication Date
CN117899048A true CN117899048A (en) 2024-04-19

Family

ID=90694571

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202410003683.XA Pending CN117899048A (en) 2024-01-02 2024-01-02 Targeting lipid nanoparticle and preparation and application thereof

Country Status (1)

Country Link
CN (1) CN117899048A (en)

Similar Documents

Publication Publication Date Title
US10709664B2 (en) Nanolipogel comprising a polymeric matrix and a lipid shell
JP7256824B2 (en) Cryoprotectant for particulate formulations
JP2022095702A (en) Stabilized formulations of lipid nanoparticles
CN102231979B (en) Processes and compositions for liposomal and efficient delivery of gene silencing therapeutics
JP2023138984A (en) Lipid nanoparticles for delivering modified rna encoding vegf-a polypeptide
JP2020532528A (en) Method of producing lipid nanoparticles
US9080186B2 (en) Compositions comprising cationic amphiphiles and colipids for delivering therapeutic molecules
KR101948839B1 (en) Pharmaceutical Composition Containing Anionic Drug and Preparation Method of the Same
KR101870316B1 (en) Preparation Method of Polymeric Micelle Containing Anionic Drugs
JP2018529738A (en) Methods for therapeutic administration of messenger ribonucleic acid drugs
JP2021530547A (en) Dry powder formulation for messenger RNA
KR20150132131A (en) Biodegradable and clinically-compatible nanoparticles as drug delivery carriers
EA032345B1 (en) Method of treating cancer using coenzyme q10
CN117899048A (en) Targeting lipid nanoparticle and preparation and application thereof
CN114601922A (en) Nanoparticles containing 5-methylpyrimidine-2, 4(1H,3H) -diketone derivatives, preparation method and application thereof
WO2020017590A1 (en) LIPID PARTICLE CONTAINING A-TYPE CpG OLIGODEOXYNUCLEOTIDE
WO2007014150A2 (en) Method of administering liposomes containing oligonucleotides
CN115645369B (en) Lung-precisely-targeted liposome nanoparticle compound and preparation method and application thereof
US20230313191A1 (en) Rna comprising secretomes and methods of their use
CN114591386B (en) Uridine derivative-containing nanoparticle, nucleic acid nanocomposite and preparation method and application thereof
CN114605338B (en) Nanoparticle and nucleic acid nanocomposite containing uracil derivative, and preparation method and application thereof
CN116549667B (en) PAS modified lipid nanoparticle, pharmaceutical preparation containing PAS modified lipid nanoparticle, and preparation method and application of PAS modified lipid nanoparticle
EP4342993A1 (en) Hpv infectious disease vaccine
TW202340472A (en) Double-stranded oligodeoxynucleotides, composition comprising the same, and uses thereof
KR20220149400A (en) Nanoparticle composition for drug delivery by using cationic lipid having disulfide bond and preparation method thereof

Legal Events

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