[go: up one dir, main page]

WO2025229369A1 - Molécules d'acide nucléique synthétiques et utilisation associée - Google Patents

Molécules d'acide nucléique synthétiques et utilisation associée

Info

Publication number
WO2025229369A1
WO2025229369A1 PCT/IB2024/000666 IB2024000666W WO2025229369A1 WO 2025229369 A1 WO2025229369 A1 WO 2025229369A1 IB 2024000666 W IB2024000666 W IB 2024000666W WO 2025229369 A1 WO2025229369 A1 WO 2025229369A1
Authority
WO
WIPO (PCT)
Prior art keywords
utr
nucleic acid
protein
acid molecule
mrna
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
PCT/IB2024/000666
Other languages
English (en)
Korean (ko)
Inventor
권오성
이민형
신광수
강종설
박주리
윤소영
오찬희
황현하
정희윤
권성필
정문교
박옥현
강이고저
양세영
김호경
윤준
엄혜현
문지하
박언영
이성률
정남진
김원경
문승태
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.)
Samsung Biologics Co Ltd
Original Assignee
Samsung Biologics 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
Priority claimed from KR1020240154459A external-priority patent/KR20250160313A/ko
Application filed by Samsung Biologics Co Ltd filed Critical Samsung Biologics Co Ltd
Publication of WO2025229369A1 publication Critical patent/WO2025229369A1/fr
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression

Definitions

  • the present invention relates to synthetic nucleic acid molecules and uses thereof, and more particularly, to a polynucleotide comprising an isolated 3'-UTR and/or 5'-UTR among RNA regulatory elements that commonly enhance protein expression efficiency in various cells, a synthetic nucleic acid molecule comprising the polynucleotide, and a pharmaceutical composition, vaccine composition, or gene therapy composition comprising the same.
  • mRNA messenger ribonucleic acid
  • mRNA-based vaccines are emerging in relation to vaccine development.
  • mRNA can theoretically encode all types of proteins. This means that the efficiency of vaccine development can be optimized by modifying the mRNA base sequence, which is a convenient method compared to other types of vaccines.
  • the important part here is that since most mRNA vaccine production and purification processes are very similar even when using different antigens, it is possible that the facilities can be maintained or standardized and used to develop different types of mRNA vaccines, which could be considered advantageous in terms of production time and economic cost.
  • mRNA is RNA that participates in protein expression by transferring genetic information from DNA to ribosomes for translation.
  • Recombinant mRNA intended for the development of therapeutics or vaccines, is produced from linear DNA by promoters (including but not limited to T7 and SP6) and RNA polymerase. During in vitro transcription, 5' capping and poly A tailing may also occur. Similar to mature mRNA in the cytoplasm, this mRNA is composed of a 5' capping region, a 5'-untranslated region (UTR), the target gene to be expressed, a 3'-UTR, and poly A.
  • the untranslated region (5' or 3' untranslated region, UTR) is known to influence mRNA stability and translational activation, affecting its half-life and protein expression levels. Therefore, finding the optimal untranslated region sequence is crucial for efficient and stable protein expression in vivo.
  • a synthetic nucleic acid molecule has been developed and the present invention has been completed. Summary of the Invention An object of the present invention is to provide a synthetic nucleic acid molecule suitable for gene therapy and/or gene vaccination. An object of the present invention is to provide a pharmaceutical composition comprising the synthetic nucleic acid molecule. An object of the present invention is to provide a vaccine composition comprising the synthetic nucleic acid molecule. An object of the present invention is to provide a composition for gene therapy comprising the synthetic nucleic acid molecule.
  • the present invention provides a solinucleotide comprising a 3'-UTR (untranslated region) isolated from a gene encoding any one selected from the group consisting of CHMP2A (Charged multivesicular body protein 2a), NME2 (Nucleoside diphosphate kinase B), and TPM2 (P-Tropomyosin).
  • CHMP2A Charge multivesicular body protein 2a
  • NME2 Nucleoside diphosphate kinase B
  • TPM2 P-Tropomyosin
  • the present invention also provides a solinucleotide comprising a 5'-UTR (untranslated region) isolated from a gene encoding any one selected from the group consisting of VAMP 8 (Vesicle-associated membrane protein 8), C0X6B 1 (Cytochrome c oxidase subunit 6B1), and HBB (Hemoglobin subunit beta).
  • VAMP 8 Vehicle-associated membrane protein 8
  • C0X6B 1 Cytochrome c oxidase subunit 6B1
  • HBB Hemoglobin subunit beta
  • the present invention also provides a synthetic nucleic acid molecule comprising, in order from 5' to 3', a) a 5'-CAP structure; b) a 5'-untranslated region (UTR); c) one or more coding regions; d) a 3'-untranslated region (UTR) isolated from a gene encoding any one selected from the group consisting of CHMP2A (Charged multivesicular body protein 2a), NME2 (Nucleoside diphosphate kinase B), and TPM2 (P-Tropomyosin); and e) a poly(A) tail or a poly(A) tail-like sequence comprising 10 to 1000 adenines (A).
  • CHMP2A Charge multivesicular body protein 2a
  • NME2 Nucleoside diphosphate kinase B
  • TPM2 P-Tropomyosin
  • a poly(A) tail or a poly(A) tail-like sequence comprising 10 to 1000 aden
  • the present invention also provides a pharmaceutical composition comprising the synthetic nucleic acid molecule.
  • the present invention also provides a vaccine composition comprising the synthetic nucleic acid molecule.
  • the present invention also provides a composition for gene therapy comprising the synthetic nucleic acid molecule.
  • Figure 1 illustrates the increase in protein efficiency achieved through a typical mRNA construct and the mRNA construct according to the present invention.
  • Figure 2 illustrates the results of Glue (Gaussia Luciferase) activity assays. Media was collected at specific time points and Glue activity was measured using a plate reader. Since Glue activity is proportional to Glue protein expression, the area under the curve (AUC) representing Glue expression by time point was considered the level of total protein expression.
  • AUC area under the curve
  • the trend line is indicated by a red dotted line.
  • Control UTR pairs (Moderna and Curevac) are indicated by gray dots.
  • Figure 5 shows the results of measuring d2EGFP expression, calculating the area under the graph representing the fluorescence intensity over time measured by Incucyte. The calculated area represents the total d2EGFP protein expression. All values were corrected to the values of the Modema UTR pair.
  • the red horizontal dotted line represents the total protein expression of the Modema UTR pair. Data are expressed as the mean and standard deviation. All experiments were performed at least three times. was achieved. A Student's t-test result with a P value less than 0.05 was considered significant between the compared subjects (* P ⁇ 0.05 and ** P ⁇ 0.01).
  • Figure 6 is a representative fluorescence image of HFF-1 and 293T cells transfected with d2EGFP mRNA containing SB2, 4, 5, and Moderna UTRs every 12 hours, respectively. A 10x objective lens was used.
  • Figure 7 shows the results of analyzing the correlation between the expression of d2EGFP total protein in two different cell lines, HFF-1 and 293T. The trend line is indicated by a red dotted line. Control UTR pairs (Moderna and Curevac) are indicated by gray dots.
  • Figure 8 shows the results of analyzing the correlation between the expression of d2EGFP Glue of different UTR pairs in HFF-1 cells. The trend line is indicated by a red dotted line. Control UTR pairs (Moderna and Curevac) are indicated by gray dots.
  • Figure 9 shows the results of analyzing the correlation between the expression of d2EGFP Glue in different UTR pairs in 293T cells. The trend line is indicated by a red dotted line. Control UTR pairs (Moderna and Curevac) are indicated by gray dots.
  • Figure 10 shows the quantitative results of in-vivo protein expression efficiency verification of mRNA containing the final selected 5'-UTR x 3'-UTR combination.
  • Figure 11 shows the IVIS image of the results of in-vivo protein expression efficiency verification of mRNA containing the final selected 5'-UTR x 3'-UTR combination.
  • the inventors of the present invention have confirmed that protein expression at the cellular or animal level is equivalent or higher than that of UTR pairs known to be superior in the past through synthetic nucleic acid molecules comprising a 3'-UTR isolated from CHMP2A (Charged multivesicular body protein 2a), NME2 (Nucleoside diphosphate kinase B) or TPM2 (P-Tropomyosin) gene and/or a 5'-UTR isolated from VAMP 8 (Vesicle-associated membrane protein 8), C0X6B1 (Cytochrome c oxidase subunit 6B1) or HBB (Hemoglobin subunit beta) gene.
  • CHMP2A Charge multivesicular body protein 2a
  • NME2 Nucleoside diphosphate kinase B
  • TPM2 P-Tropomyosin gene
  • VAMP 8 Vesicle-associated membrane protein 8
  • C0X6B1 Cytochrome c oxidase subunit
  • the present invention applies ml *4 ⁇ (N ⁇ methylpseudouridine), a modified uridine mainly used in the production of mRNA drugs, to the sequences discovered. It is very suitable for practical use.
  • UTR sequence applied to the existing COVID-19 mRNA vaccine, it was a sequence discovered based on natural uridine.
  • actual vaccines used modified uridine to reduce side effects and increase protein expression.
  • the present invention by using modified uridine to discover UTR sequences, we sought to discover sequences more suitable for the development of mRNA vaccines/therapeutics applied to humans.
  • the present invention is very suitable for practical use with sequences discovered by applying mlsan, which is mainly used in the production of mRNA drugs.
  • the present invention provides a 3'-terminal protein isolated from a gene encoding any one selected from the group consisting of CHMP2A (Charged multivesicular body protein 2a), NME2 (Nucleoside diphosphate kinase B), and TPM2 (P-Tropomyosin).
  • UTR untranslated region
  • CHMP2A also known as Charged multivesicular body protein 2a, belongs to the chromatin-modifying protein/charged multivesicular body protein (CHMP) family. It is a component of ESCRT-III (endoplasmic reticulum sorting complex required for transport III), a complex involved in the degradation of surface receptor proteins and the formation of intracellular multivesicular bodies (MVBs).
  • NME2 is a nucleoside diphosphate kinase (NDPK, NDP kinase, (poly)nucleotide kinase, and nucleoside diphosphokinase), an enzyme that catalyzes the terminal phosphate exchange between different nucleoside diphosphates (NDPs) and triphosphates (NTPs) in a reversible manner, generating nucleotide triphosphates.
  • NDPs act as acceptors
  • NTPs act as donors of phosphate groups. They are involved in cell proliferation, differentiation and development, signal transduction, G protein-coupled receptors, endocytosis, and gene expression.
  • TPM2 is p-tropomyosin, also known as tropomyosin beta chain, a protein encoded by the TPM2 gene in humans.
  • p-tropomyosin is a striated muscle-specific coiled-coil dimer that functions to stabilize actin filaments and regulate muscle contraction.
  • Polynucleotide as used herein means, for example, a nucleic acid, preferably DNA or RNA.
  • Nucleotide “nucleotide sequence”, and “oligonucleotide” are used interchangeably. It may comprise a polymeric form of nucleotides of any length, deoxyribonucleotides or ribonucleotides, or analogs thereof.
  • a polynucleotide may have any three-dimensional structure and may perform any function, known or unknown.
  • a polynucleotide may comprise one or more modified nucleotides, for example, methylated nucleotides and nucleotide analogs. Modifications to the nucleotide structure may be possible before or after formation of the polymer.
  • a nucleic acid is a polymer comprising or consisting of nucleotide monomers covalently linked to each other by phosphodiester bonds of a sugar/phosphate backbone.
  • a "nucleic acid” includes modified nucleic acids, such as base-modified, sugar-modified, or backbone-modified DNA or RNA molecules.
  • the above DNA is an abbreviation for deoxyribonucleic acid. It is a polymer made of nucleic acid molecules, i.e. nucleotides. These nucleotides are usually deoxy-adenosine-monophosphate, deoxy-thymidine-monophosphate, deoxy-guanosine-monophosphate, and deoxy-cytidine-monophosphate monomers, and are made up of a sugar (deoxyribose), a base, and a phosphate, and are polymerized by a characteristic backbone structure.
  • nucleotides are usually deoxy-adenosine-monophosphate, deoxy-thymidine-monophosphate, deoxy-guanosine-monophosphate, and deoxy-cytidine-monophosphate monomers, and are made up of a sugar (deoxyribose), a base, and a phosphate, and are polymerized by a characteristic backbone structure.
  • the backbone structure is typically formed by the sugar moiety of the first nucleotide, i.e., the deoxyribose, and the phosphate moiety of the second, and the phosphodiester bond between adjacent monomers.
  • the specific order of the monomers i.e., the order of the bases linked to the sugar/phosphate backbone, is called the DNA sequence.
  • DNA can be single-stranded or double-stranded.
  • nucleotides of the first strand typically hybridize with nucleotides of the second strand, for example by A/T base pairing and G/C base pairing.
  • Such RNA includes, for example, mRNA.
  • RNA is usually an abbreviation for ribonucleic acid.
  • RNA can be obtained by transcription of a DNA sequence, for example, in cells. In the body, the DNA transcript can usually be processed into mRNA.
  • RNA transcribed from DNA undergoes splicing, 5'-capping, polyadenylation, and nuclear or mitochondrial mRNA is generated through various post-transcriptional modifications, such as excretion from lia and similar sites.
  • mRNA typically provides a nucleotide sequence that can be translated into the amino acid sequence of a specific peptide or protein.
  • mRNA typically contains a 5'-cap, a 5'-UTR, an open reading frame, a 3'-UTR, and a poly(A) sequence.
  • the 3'-UTR is a portion of an mRNA that is typically located between the protein coding region (i.e., open reading frame) of the mRNA and the poly(A) sequence.
  • the 3'-UTR of an mRNA is not translated into an amino acid sequence.
  • the 3'-UTR sequence is usually encoded by a gene, which is then transcribed into mRNA during gene expression.
  • the genomic sequence is first transcribed into a primary transcript containing optional introns. Mature mRNA is then generated through a process that includes steps such as 5' capping, splicing, and modification of the 3'-terminus, such as polyadenylation of the 3'-terminus, and optional endo- or exonuclease digestion.
  • the 3'-UTR comprises the nucleotides located immediately 3' to the stop codon of the protein coding region and immediately 5' to the poly(A) sequence.
  • the 3' untranslated region (UTR) may comprise any one of the base sequences selected from the group consisting of SEQ ID NOs: 1 to 3.
  • the present invention also relates to a solinucleotide comprising a 5'-UTR (untranslated region) isolated from a gene encoding any one selected from the group consisting of VAMP 8 (Vesicle-associated membrane protein 8), C0X6B1 (Cytochrome c oxidase subunit 6B1), and HBB (Hemoglobin subunit beta).
  • VAMP 8 Vehicle-associated membrane protein 8
  • C0X6B1 Cytochrome c oxidase subunit 6B1
  • HBB Hemoglobin subunit beta
  • VAMP8 is a vesicle-associated membrane protein 8, synaptobrevin/VAMP, syntaxin, 25kD synaptosome-associated protein SNAP25, a key component of a protein complex involved in the docking and/or fusion of synaptic vesicles with the presynaptic membrane.
  • the protein encoded by this gene is a member of the vesicle-associated membrane protein (VAMP)/synaptobrevin family, which is associated with the perinuclear vesicle structures of early intracellular compartments.
  • C0X6B1 is cytochrome c oxidase subunit 6B1.
  • Cytochrome c oxidase 6B1 is a subunit of the cytochrome c oxidase complex, also known as complex IV, the last enzyme of the mitochondrial electron transport chain. Mutations in the C0X6B1 gene are associated with severe infantile encephalopathy and mitochondrial complex IV deficiency (MT-C4D).
  • HBB is a globin protein encoded by the HBB gene that, along with alpha globin (HBA), constitutes hemoglobin A (HbA), the most common form of hemoglobin in adults. It is 147 amino acids long and has a molecular weight of 15,867 Da. It is encoded by the HBB gene on human chromosome 11, and mutations in the gene produce several variant proteins that are associated with genetic disorders such as sickle cell disease and beta-thalassemia, and beneficial traits such as genetic resistance to malaria.
  • HBA alpha globin
  • HbA hemoglobin A
  • the 5'-UTR is located 5' of the open reading frame.
  • the 5'-UTR starts at the transcription start site and ends at the nucleotide before the start codon of the open reading frame.
  • the 5'-UTR may contain elements that regulate gene expression, such as, for example, a ribosome binding site.
  • the 5'-UTR may be post-transcriptionally modified, for example, by the addition of a 5'-cap.
  • the 5'-UTR corresponds to a sequence of a mature mRNA located between the 5'-cap and the start codon.
  • the 5' untranslated region may include any one of the base sequences selected from the group consisting of SEQ ID NOs: 4 to 6. , there is.
  • the present invention further comprises a) a 5'-CAP structure; b) a 5'-UTR in the order of 5' to 3'.
  • the coding region (open reading frame) preferably comprises a start codon, i.e.
  • an open reading frame is a nucleotide sequence consisting of a number of nucleotides divisible by three, preferably beginning with a start codon (e.g. ATG or AUG) and preferably ending with a stop codon (e.g.
  • the open reading frame may be isolated or incorporated into a longer nucleic acid sequence, e.g. a vector or an mRNA.
  • An open reading frame may also be referred to as a 'polypeptide or protein coding region'.
  • 5'-CAP refers to a specially modified nucleotide at the 5' end of some primary transcripts, such as precursor mRNA.
  • the process known as mRNA capping is highly regulated and is essential for generating stable, mature mRNAs that can undergo translation during protein synthesis.
  • the starting point for capping with 7-methylguanylate is the unmodified 5' end of the RNA molecule, and the 5' end is
  • the present invention relates to a poly(A) tail or a poly(A) tail comprising 10 to 1000 adenines (A). contains a poly(A) tail-like sequence.
  • a promoter may additionally be located upstream of the 5' untranslated region (UTR).
  • the promoter may include elements necessary for transcription, such as an RNA polymerase promoter. It may include a phage RNA polymerase promoter such as SP6 or T7, preferably a T7 promoter encoding an mRNA sequence.
  • the length of the poly(A) sequence may vary within the range of 10 to 1000 adenine nucleotides.
  • the poly(A) sequence can have a length of from about 20 adenine nucleotides to about 400 adenine nucleotides, such as from about 20 adenine nucleotides to about 300 adenine nucleotides, preferably from about 40 to about 200 adenine nucleotides, more preferably from about 50 to about 100 adenine nucleotides, such as from about 60, 70, 80, 90 or 100 adenine nucleotides.
  • the poly(A) sequence can be located downstream of the 3' untranslated region (UTR).
  • the poly(A) sequences can be linked directly or via a linker, for example via a linker of 1-50, preferably 1-20 nucleotides, or via a stretch of nucleotides, such as 2, 4, 6, 8, 10, 20, etc. nucleotides.
  • the above poly(A) tail-like sequence may include one or more nucleotides other than adenine selected from the group consisting of uracil (U), cytosine (C) and guanine (G) inserted between a plurality of adenines or at the end of the poly(A) tail.
  • the 3' untranslated region (UTR) may include any one of the base sequences selected from the group consisting of SEQ ID NOs: 1 to 3.
  • the above 5' untranslated region may include any one of the base sequences selected from the group consisting of SEQ ID NOs: 4 to 6.
  • the coding region may encode, for example, a peptide or a protein.
  • the transcribable nucleic acid sequence or a transcript thereof may comprise an ORF encoding the peptide or protein.
  • the nucleic acid may express the encoded peptide or protein.
  • the nucleic acid may encode any one or more proteins selected from the group consisting of antigenic proteins, allergenic proteins, therapeutic proteins, and fragments, variants or derivatives of the proteins.
  • the nucleic acid may comprise one or more backbone-modified, sugar-modified or base-modified nucleic acids.
  • Nucleoside means a nucleobase linked to a sugar
  • nucleotide means a nucleoside having a phosphate group covalently linked to the sugar portion of the nucleoside.
  • Nucleobase means a heterocyclic moiety capable of binding to a base of another nucleic acid.
  • the nucleoside may include modifications.
  • the internucleoside linkage may comprise a modified internucleoside linkage or may comprise a modified sugar.
  • the internucleoside linkage may be a phosphorothioate, boranophosphate, or methyl phosphonate linkage.
  • the nucleoside may comprise a modified sugar.
  • the modification may comprise a substitution at the 2' carbon position of the sugar structure with 2'-0-methyl (2'-0-Me), 2'-0-methoxyethyl (2'MOE), 2'-0-methoxyethyl-5'methyl, 2'-0-aminoethyl, 5'-methyl, 2'-0-propyl, 2'-methylthioethyl, or 2'-fluoro.
  • the nucleoside may be, for example, a nucleoside linkage formed by a phosphorothioate, boranophosphate, or methyl phosphonate linkage; and a 2'-0-methyl (2'-0-Me), 2'-0-methoxyethyl (2'MOE), 2'-0-methoxyethyl-5'methyl, 2'-0-aminoethyl, 5'-methyl, 2'-0-propyl, 2'-methylthioethyl, or 2'-fluoro
  • the nucleoside may be characterized by including a modification selected from the group consisting of, for example, a modification of the sugar moiety, for example, a modification at the 2' carbon position of the sugar structure in the nucleotide, specifically,
  • Modification with 2'-Fluoro modification of nucleotide linkages to phosphorothioate, boranophosphate, or methyl phosphonate; modification to the form of PNA (peptide nucleic acid), LNA (locked nucleic acid), or UNA (unlocked nucleic acid); and may include a phosphate group.
  • “2’-O-methoxyethyl” (also 2’-M0E and 2’-O(CH2)2-OCH3) refers to the O-methoxy-ethyl modification at the 2’ position of the furosyl ring.
  • “2'-0-methoxyethyl nucleotide” means a nucleotide comprising a 2'-0-methoxyethyl modified sugar moiety.
  • Modified sugar refers to a substitution or change from a natural sugar.
  • 5-methylcytosine refers to cytosine modified with a methyl group attached at the 5' position. 5-methylcytosine is a modified nucleobase.
  • Modified internucleoside linkage means a substitution or any change from a naturally occurring internucleoside linkage.
  • Modified nucleobase means any nucleobase other than adenine, cytosine, guanine, thymine, or uracil.
  • Modified nucleotide means, independently, a nucleotide having a modified sugar moiety, a modified internucleoside linkage, or a modified nucleobase.
  • Modified nucleoside means, independently, a nucleoside having a modified sugar moiety or a modified nucleobase.
  • a “modified oligonucleotide” is a nucleotide having at least one modified nucleotide. .
  • the present invention also relates to a vector comprising the synthetic nucleic acid molecule.
  • the vector may be an expression vector, a cloning vector, or the like.
  • the vector may be used for the production of an expression product such as mRNA, or a peptide, a polypeptide, or a protein. It may contain a sequence necessary for the transcription of a sequence stretch of the vector, such as a promoter sequence, for example, an RNA promoter sequence.
  • the vector may be, for example, an RNA vector or a DNA vector.
  • the vector may be a viral vector or a plasmid vector.
  • the vector may be a circular molecule, and the vector may comprise a double-stranded molecule. Circular, double-stranded DNA molecules can be readily used to introduce an original artificial nucleic acid molecule.
  • the circular vector may be linearized, for example, by restriction enzyme digestion.
  • the vector may comprise sequences suitable for amplification of the vector, such as a cloning site, a selection marker such as an antibiotic resistance element, and an origin of replication.
  • the vector is suitable for transcription using a eukaryotic, prokaryotic, viral or phage transcription system, such as a eukaryotic, prokaryotic, viral or phage in vitro transcription system.
  • the vector is suitable for in vitro transcription using a phage based in vitro transcription system, such as a T7 RNA polymerase based in vitro transcription system.
  • the above vector can be used by microinjection, electroporation,
  • the vector can be delivered to cells by various methods known in the art, such as, but not limited to, DEAE-dextran treatment, lipofection, nanoparticle-mediated transfection, protein delivery domain-mediated introduction, and PEG-mediated transfection.
  • Known expression vectors such as plasmid vectors, cosmid vectors, and bacteriophage vectors can be used as the vector, and the vector can be easily prepared by those skilled in the art according to any known method using DNA recombinant technology.
  • the recombinant expression vector can contain the nucleic acid in a form suitable for expression of the nucleic acid in a host cell, and the recombinant expression vector can contain one or more regulatory elements operably linked to the nucleic acid sequence to be expressed.
  • operably linked means that the nucleotide sequence of interest is linked to a regulatory element in a manner that permits expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell).
  • the recombinant expression vector may contain a form suitable for mRNA synthesis, including a T7 promoter#, which means that it contains one or more regulatory elements that enable mRNA synthesis in vitro or in vitro, i.e., mRNA can be synthesized by T7 polymerase.
  • Regulatory elements may include promoters, enhancers, internal ribosome entry sites (IRES), and other expression modulating elements (e.g., transcription termination signals such as polyadenylation signals and poly-U sequences).
  • Regulatory elements are elements that direct induction or constant expression of a nucleotide sequence in many types of host cells and in certain host cells. It comprises elements (e.g., tissue-specific regulatory sequences) that direct the expression of a nucleotide sequence only in the nucleic acid.
  • the nucleic acid may be introduced in the form of RNA, e.g., mRNA.
  • the nucleic acid according to the present invention may be in the form of mRNA. This may result in transient protein expression.
  • the "human” may be selected from the group consisting of viral RNA, self-replicating RNA, and replicon RNA.
  • Self-replicating RNA (replicon RNA) extracted from the alphavirus genome can be a potent vaccine vector.
  • the replicon RNA encodes an enzyme complex (replicase) required for cytoplasmic replication of the replicon RNA in the first two-thirds.
  • the replicase recognizes internal RNA structures that act as subgenomic promoters for replicase-dependent subgenomic RNA synthesis.
  • the genes or antigens for vaccination are encoded in the subgenomic RNA, which is much shorter than the entire replicon.
  • both the genome and the subgenomic RNA resemble cellular mRNA. Both are flanked by UTRs, and both are capped and polyadenylated. The enzymes responsible for capping and polyadenylation are included in the replicase complex.
  • conserved sequence elements within the UTR overlap with the ORF of the replicase. (Conserved sequence elements: CSE) are required for the binding of replicase and can act as promoters for minus strand synthesis (3'CSE) or plus strand synthesis (5'CSE).
  • the present invention relates to a vaccine composition comprising the synthetic nucleic acid molecule.
  • the present invention relates to a gene comprising the synthetic nucleic acid molecule.
  • a therapeutic composition is provided.
  • the composition may include a delivery means for delivering mRNA expressed in the composition.
  • the expressed mRNA may be delivered via nanoparticles.
  • the composition may be delivered via gold nanoparticles.
  • the gold nanoparticles may have a modified surface. Specific examples of modifications are specifically exemplified in Acc Chem Res. 2019 June 18; 52(6): 1496-1506. and Pharmaceutics 2021, 13, 900., which may be incorporated herein by reference.
  • the mRNA may be delivered to cells by linking gold nanoparticles to form a cationic endosomal disruptive polymer complex (Nature Biomedical Engineering volume 1, pages 889-901 (2017)).
  • the cationic endosomal disruptive polymers • may be, for example, polyethylene glycol, poly(arginine), poly(lysine), poly(histidine), poly-[2 - ⁇ (2-aminoethyl)amino ⁇ -ethyl-aspartamide] (pAsp(DET)), a block co-polymer of poly(ethylene glycol) (PEG) and poly(arginine), a block co-polymer of PEG and poly(lysine), or a block co-polymer of PEG and poly ⁇ N-[N-(2-aminoethyl)-2 - aminoethyl]aspartamide ⁇ (PEG-pAsp(DET)).
  • a gold particle modified with arginine may be used on the surface.
  • the gold particle modified with arginine may be assembled with a nuclease or a polynucleotide encoding the same and/or a cleavage factor or a solinucleotide encoding the same. Through this, the gold particle fuses with the membrane of the target cell and moves into the cytoplasm. (ACS Nano. 2017, 11:2452-2458).
  • the expressed mRNA can be delivered via liposomes, lipid nanoparticles (LNPs), or various nanoparticles.
  • Liposomes or LNPs contain cationic lipids, non-cationic lipids, or neutral lipids, and may additionally contain other lipids such as polyethylene glycol (PEG) or cholesterol.
  • PEG polyethylene glycol
  • Such mRNA delivery systems are specifically described in U.S. Patent Publication Nos. 2018/0311176, 2019/0032051, 2021/0046192, International Patent Publication Nos. W02018/081480, W02020/097540, W02020/097548, W02021/007278, etc., and are incorporated herein by reference.
  • the cationic lipids are specifically exemplified in U.S. Patent Publication Nos.
  • N,N-dioleyl-N,N-dimethylammonium chloride DODAC
  • N,N-distearyl-N,N-dimethylammonium bromide DDAB
  • trimethylammonium chloride DOTAP
  • trimethylammonium chloride DOTMA
  • N,N-dimethyl-2,3-dioleyloxy)propylamine DODMA
  • 1,2-Dilinoleyloxy-N,N-dimethylaminopropane (DLinDMA), 1,2-Dilinolenyloxy-N,N-dimethylaminopropane (DLenDMA), 1,2-Dilinoleylcarbamoyloxy- 3 -dimethylaminopropane (DLin-C-DAP), 1,2-Dilinoley oxy-3 -
  • N,N-dimethylnonacosa- 11,20,2-trien- 10-amine 5-carboxyspermylglycine di octaol eoy 1 ami de (“DOGS”), dipalmitoylphosphatidylethanolamine 5-carboxyspermyl- amide (“DPPES”), l,2-dimyristyloxypropyl-3-dimethyl-hydroxy ethyl ammonium bromide (DMRIE), DMRIE-HP, Lipofectamine (DOSPA), 3b-(N— (N',N'- dimethylaminoethane)-carbamoyl)cholesterol (“DC-Choi”), N-( 1,2-dimyhstyloxyprop- 3-yl)-N,N-dimethyl-N-hydroxy ethyl ammonium bromide (“DMRIE”), l,2-Dioleoyl-3-dimethylammonium-propane (“DODAP”), DMD
  • the non-cationic lipid is specifically exemplified in U.S. Patent Publication Nos. 2018/0311176, 2019/0032051, etc., and may be, for example, phosphatidylglycerol, cardiolipin, diacylphosphatidylserine, diacylphosphatidic acid, N-dodecanoyl phosphatidylethanolamine, N-succinyl phosphatidylethanolamine, N-glutaryl phosphatidylethanolamine, or lysylphosphatidylglycerol.
  • the non-cationic lipids include, for example, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl-phosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoylphosphatidylethanolamine (POPE), dioleoyl-phosphatidylethanolamine 4-(-maleimidomethyl)-cyclohexane-l-carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPC), dip
  • the neutral lipid is specifically exemplified in U.S. Patent Publication Nos. 2018/0311176 and 2019/0032051, and may include, but is not limited to, diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide, sphingomyelin, dihydrosphingomyelin, cephalin, or cerebrosides.
  • PEG lipid may be included to prevent aggregation of particles generated during mRNA delivery.
  • PEG lipid is specifically exemplified in U.S. Patent Publication Nos.
  • 2018/0311176 and 2019/0032051 may include, but is not limited to, PEG-diacylglycerol (DAG), a PEG-dialkyloxypropyl (DAA), a PEG-phospholipid, a PEG-ceramide (Cer), or a mixture thereof.
  • DAG PEG-diacylglycerol
  • DAA PEG-dialkyloxypropyl
  • Cer PEG-ceramide
  • PLGA may be conjugated to a lipid-terminating PEG forming PLGA-D SPE-PEG
  • PEG lipid is selected from PEG-c-DOMG and 1,2-Dimyristoyl-sn-glycerol, methoxypolyethylene Glycol (PEG-DMG), 1,2-Distearoyl-sn-glycerol, methoxypolyethylene Glycol (PEG- DSG), PEG-c-DOMG, 1,2-Distearoyl-sn-glycerol, methoxypolyethylene glycol (PEG- DSG) 1,2-Dipalmitoyl-sn-glycerol, methoxypolyethylene glycol (PEG-DPG), PEG- lipid conjugates such as, eg, PEG coupled to di alkyl oxy propyl s (eg, PEG-DAA conjugates), PEG coupled to diacylglycerols (eg, PEG-DAG conjug
  • the PEG may be, but is not limited to, a PEG-dilauryloxypropyl (C12), a PEG-dimyristyloxypropyl (Cl 4), a PEG-dipalmityloxypropyl (Cl 6), a PEG-distearyloxypropyl (Cl 8), PEG-c-DOMG, PEG-DMG, Methoxypolyethyleneglycoloxy(2000)-N,N-ditetradecylacetamide (also known as ALC-0159) or mixtures thereof.
  • Peptides may be used for the mRNA delivery.
  • the peptide must have a cation to electrostatically interact with the anionic phosphate group of the nucleic acid, and may include a positively charged amino acid to electrostatically interact with the phosphate group.
  • Specific details of peptides usable for mRNA delivery are described in AIMS Biophysics, 7(4): 323-338, which may be incorporated herein by reference.
  • the peptide usable for mRNA delivery may include protamine.
  • Protamine is a small, cationic, arginine-rich nuclear protein that contributes to the stability of DNA during spermatogenesis in the testis, and protamine stabilizes mRNA molecules, enabling efficient delivery.
  • a protamine-mRNA complex is specifically described in U.S. Patent No.
  • Cell-penetrating peptides may also be promising cationic molecules for mRNA delivery.
  • Arginine-rich RALA peptide WEARL ARAL ARAL ARHL ARAL ARALRACEA
  • RALA LAH4
  • Amphiphilic CPPs such as (KKALLAHALHLLALLALHLAHALKKA) can be used to deliver mRNA molecules.
  • a peptide may be additionally included for mRNA delivery.
  • the peptide may provide nucleic acid packaging function and prevent DNA or RNA from being degraded intracellularly or extracellularly. Examples of such peptides are specifically described in U.S. Patent Publication No. 2021/0170046, which is incorporated herein by reference, but is not limited thereto.
  • the composition may additionally include one or more pharmaceutically acceptable carriers.
  • the pharmaceutically acceptable carriers must be compatible with the active ingredient of the present invention, and may be used as a mixture of saline solution, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol, and one or more of these components, and may be added as needed, and other conventional additives such as antioxidants, buffers, and bacteriostatic agents.
  • it can be formulated as an injectable formulation such as an aqueous solution, suspension, emulsion, etc. by additionally adding a diluent, a dispersant, a surfactant, a binder, and a lubricant.
  • a lyophilized formulation it is preferable to provide it by formulating it as a lyophilized formulation.
  • a method commonly known in the technical field to which the present invention pertains can be used, and a stabilizer for lyophilization may be added.
  • it can be preferably formulated according to each disease or ingredient using an appropriate method in the art or a method disclosed in Remington's pharmaceutical Science (Mack Publishing company, Easton PA).
  • the content and administration method of the active ingredient, etc. included in the composition of the present invention can be determined by a general expert in the technical field based on the symptoms and severity of the disease of a typical patient.
  • the composition of the present invention can be determined by the method.
  • composition of the present invention can be administered orally or parenterally.
  • the route of administration of the composition according to the present invention is not limited to these, but for example, it can be administered intrabronchially, buccally, intravenously, intramuscularly, intraarterially, intramedullaryly, intrathecally, intracardiacly, transdermally, subcutaneously, intraperitoneally, enterally, sublingually, or topically.
  • the dosage of the composition according to the present invention varies depending on the patient's weight, age, sex, health condition, diet, administration time, method, excretion rate, or disease severity, and can be easily determined by a person skilled in the art.
  • the composition of the present invention can be formulated into a suitable dosage form using a known technique for clinical administration.
  • the present invention will be described in more detail through examples. These examples are only for illustrating the present invention, and it will be apparent to those skilled in the art that the scope of the present invention is not to be construed as being limited by these examples.
  • the mRNA nucleic acid structure comprises a 5'-CAP, a 5' untranslated region (5'- It includes the 3' untranslated region (3'-UTR), poly(A) sequence, and the encoded gene sequence (CDS) between the 5'-UTR and 3'-UTR. (Fig. 1).
  • the UTR existing in HeLa, 293T, and Human fibroblast (HFF-1) were first screened.
  • Example 2 Confirmation of mRNA synthesis and protein translation efficiency containing the final selected 5'-UTR x 3'-UTR combination
  • a vector construct containing the CureVac sequence was constructed.
  • a backbone plasmid, pUC57-PolyA(50), containing a 50-mer polyA sequence was synthesized.
  • Eleven DNA inserts consisting of Glue CDS between different 5' and 3'-UTR pairs were synthesized and inserted into the pUC57-PolyA(50) backbone plasmid.
  • the Glue CDS region in the 11 plasmids described above was replaced with d2EGFP. All Glue plasmids and the synthesized d2EGFP DNA fragments were digested with NcoLHF (NEB) and Xbal (NEB).
  • Each digested DNA was separated by agarose gel electrophoresis, and bands of the expected size were isolated from the gel as recommended by the manufacturer, followed by DNA gel extraction.
  • the linearized DNA and inserts were ligated with T4 DNA ligase (NEB).
  • the reaction mixture was transformed into chemically competent cells (NEB). Some E. coli colonies were selected and sequenced by Sanger sequencing. The plasmids of the selected clones were used in further experiments.
  • IVT templates were amplified from plasmid DNA using specific primers (Table 2) and Q5 High-fidelity polymerase (NEB), with a primer annealing temperature of 58°C.
  • DNA was purified using the Monarch PCR & DNA Cleanup Kit according to the manufacturer's instructions.
  • RNA purity was measured by ultra-performance liquid chromatography (UPLC). RNA fragments in the samples were separated using an Acquity UPLC H-Class (Waters) and a DNApac reversed-phase column (Thermo Fisher Scientific). Diluted RNA samples and column wash deionized water were loaded onto the instrument's plate and sequentially injected for analysis, and RNA fragments were detected at 260 nm. The area of the major peak considered to represent intact mRNA was measured to calculate purity. Mobile phases A and B (0.1 M TEAA in water and 0.1 M TEAA in 25% acetonitrile, respectively) were consumed within 4 weeks. The column temperature and flow rate were 75 °C and 0.4 mL/min, respectively. Each mRNA was used in subsequent experiments after confirming that its purity was greater than 70%.
  • UPLC ultra-performance liquid chromatography
  • HFF-1, HeLa, and 293T cell lines were purchased from the American Type Culture Collection (ATCC). HFF-1 cells were cultured in DMEM supplemented with 15% heat-inactivated (HI)-FBS and IX penicillin/streptomycin (P/S, all purchased from Thermo Fisher Scientific). HeLa and 293T cells were cultured in DMEM supplemented with 10% HLFBS and IX P/S. For subculture, 0.05% trypsin/EDTA (Thermo Fisher Scientific)# Cells were dissociated using a 10 ⁇ g ml culture medium and then seeded onto plates containing serum-supplemented medium. The three cell lines were passaged every 2–3 days.
  • HFF-1 cells 12,500 cells/ cm2 , and for HeLa and 293T cells, 25,000 cells/ cm2 were seeded onto plates 24 hours prior to transfection.
  • Culture dishes were coated with Soly-D-lysine (Thermo Fisher Scientific) for 293T cells only.
  • mRNA was introduced into cells using Lipofectamine MessengerMax transfection reagent (Thermo Fisher Scientific) according to the manufacturer's instructions.
  • the medium was replaced with OptiMEM I Reduced Serum Medium (Thermo Fischer Scientific) before transfection and treated with the RNA-Lipofectamine mixture.
  • the medium was replaced with serum-containing medium 1 hour after transfection.
  • Glue assay For protein expression assays such as Gaussia luciferase assay (Glue assay), the medium from cells transfected with Glue mRNA was collected in 1.5 mL tubes and immediately frozen in a -80°C freezer. All samples were thawed and centrifuged immediately prior to analysis to remove cellular debris. After centrifugation, 50 pL of the sample was transferred to a new white plate (Greiner) and mixed with 50 pL of NanoDLR Stop & Gio reagent from the Nano-Gio Dual Luciferase Reporter Assay System (Promega) to quantify Glue activity. Glue activity was measured within 3 minutes at 480 nm for 50 ms using a Spectramax iD5 (Molecular Devices).
  • Glue activity was measured within 3 minutes at 480 nm for 50 ms using a Spectramax iD5 (Molecular Devices).
  • Imcucyte equipped with a 37°C CO2 incubator for GFP expression measurement Fluorescence images were acquired every 4 hours for 72 hours using a Satorius system. To exclude autofluorescence signals, the GFP expression profiles of lipofectamine control samples and GFP-transfected samples were compared. Fluorescence was corrected for the space occupied by cells analyzed by phase-contrast imaging. To determine total protein expression, Glue activity and GFP expression profiles were plotted over time, and the area under the curve was considered the total protein expression of mRNA. Each experiment was performed at least twice to reduce technical variation.
  • Gaussia luciferase is a type of luciferase enzyme with a relatively short intracellular protein half-life of approximately 30 minutes. Unlike FLuc, RLuc, and NanoLuc, it is a secreted enzyme. Glue is known to remain very stable for several days once secreted into the culture medium. Therefore, Glue can be easily collected from the culture medium without cell lysis, allowing continuous monitoring of Glue translation in the same transfection batch. The culture medium was replaced 3 hours before sampling to allow the secreted Glue to accumulate, and luminescence was measured after sampling at the specified time. The protein expression levels of the six UTR pair candidates and two conventional controls (Moderna and CureVac) were measured in HeLa cells.
  • Example 3 Testing the protein expression efficiency of mRNAs containing the final selected 5'-UTR x 3'-UTR combinations in various GOIs by changing the GOI to Green Florescence protein (GFP).
  • GFP Green Florescence protein
  • UTR pairs should be applicable to a wide range of therapeutics, it is desirable that the regulatory effect on translation persists for various coding sequences (CDS).
  • CDS coding sequences
  • the CDS was changed from Glue to d2EGFP.
  • the corresponding mRNAs were introduced into HFF-1 and 293T cell lines to compare the overall expression of d2EGFP across various UTR pairs.
  • phase contrast and GFP fluorescence images were acquired in live cells using Incucyte every 4 h. As expected, total protein expression was Compared to the control UTR pair of John, the fluorescence intensity was enhanced by the UTR pair of SB2, 4, and 5.
  • Example 4 Verification of the universality of the protein expression efficacy of mRNA containing the final selected 5'-UTR x 3'-UTR combination from different CDSs.
  • mRNA-loaded lipid nanoparticles were prepared.
  • 0159 Lipid mixture mixed in a molar ratio of 46.3:9.4:42.7:1.6 and Citrate Buffer mRNA diluted to (pH 4) was prepared into mRNA-LNP using microfluidics (Ignite, Precision NanoSystems Inc.) with an N/P ratio of 6. °], FRR was set to 3 (mRNA): 1 (lipid), and TRR was set to 12 ml/min.
  • FRR was set to 3 (mRNA): 1 (lipid)
  • TRR was set to 12 ml/min.
  • the final manufactured mRNA-LNP was diluted to 25 ng/ul based on mRNA by quantifying the mRNA concentration using the Quant-itTM RiboGreen RNA Assay Kit (Invitrogen). Empty LNP was prepared in the same manner by replacing it with Citrate Buffer (pH 4) without mRNA.
  • the manufactured mRNA-LNP was used in subsequent experiments when it met the criteria of size, PDI, Zeta potential, and EE. [Table 3]
  • mRNA-LNPs manufactured in Example 5 were intravenously administered to mice to verify the protein expression efficiency.
  • the animals used in the experiment were female BALB/c, obtained at 6 weeks of age, acclimatized for 1 week, and then the experiment was conducted at 7 weeks of age.
  • Each mRNA-LNP was intravenously administered to animals at a dose of 5 ug/head, and images were taken four times in total: 6 hours, 24 hours, 48 hours, and 72 hours after administration to confirm body distribution.
  • AUC area under the curve
  • RNA therapeutics are typically produced in vitro, a significantly different environment from cellular systems. In vitro-synthesized mRNA is translated upon introduction into the cytoplasm. Because exogenously administered mRNA is recognized as a non-self molecule in the cytoplasm, triggering an innate immune response, modified nucleosides, such as ml san, are incorporated into mRNA therapeutics to mitigate the innate immune response. Furthermore, these nucleoside modifications can affect the function of mRNA by affecting its structure and binding affinity for RNA-binding proteins. Furthermore, these mRNAs Naked mRNA exists in the absence of nuclear RNA-binding proteins.
  • IVTed mRNA in vitro transcribed mRNA
  • the present invention relates to the base sequences of 5'-UTR and 3'-UTR among RNA regulatory elements that commonly enhance protein expression efficiency in various cells, and 3'-UTR and/or 3'-UTR isolated from CHMP2A (Charged multivesicular body protein 2a), NME2 (Nucleoside diphosphate kinase B) or TPM2 (P-TYopomyosin) gene.
  • CHMP2A Charge multivesicular body protein 2a
  • NME2 Nucleoside diphosphate kinase B
  • TPM2 P-TYopomyosin
  • VAMP 8 Vehicle-associated membrane protein 8
  • COX6B1 Cytochrome c oxidase subunit 6B1
  • HBB Hemoglobin subunit beta

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • Plant Pathology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Mycology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des molécules d'acide nucléique synthétiques, une utilisation associée et : un polynucléotide comprenant un 3'-UTR isolé et/ou un 5'-UTR ; des molécules d'acide nucléique synthétiques comprenant le polynucléotide ; et une composition pharmaceutique, une composition vaccinale ou une composition de thérapie génique le comprenant.
PCT/IB2024/000666 2024-05-02 2024-11-05 Molécules d'acide nucléique synthétiques et utilisation associée Pending WO2025229369A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20240058580 2024-05-02
KR10-2024-0058580 2024-05-02
KR1020240154459A KR20250160313A (ko) 2024-05-02 2024-11-04 합성 핵산 분자 및 이의 용도
KR10-2024-0154459 2024-11-04

Publications (1)

Publication Number Publication Date
WO2025229369A1 true WO2025229369A1 (fr) 2025-11-06

Family

ID=97561294

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2024/000666 Pending WO2025229369A1 (fr) 2024-05-02 2024-11-05 Molécules d'acide nucléique synthétiques et utilisation associée

Country Status (1)

Country Link
WO (1) WO2025229369A1 (fr)

Similar Documents

Publication Publication Date Title
US12371699B2 (en) Nucleic acids encoding CRISPR-associated proteins and uses thereof
US20240409954A1 (en) Gene construct for expressing mrna
US20220340886A1 (en) Therapeutics for glycogen storage disease type iii
US11464836B2 (en) RNA for treatment or prophylaxis of a liver disease
US11939600B2 (en) Compositions and methods for treating phenylketonuria
WO2025229369A1 (fr) Molécules d'acide nucléique synthétiques et utilisation associée
KR20250160313A (ko) 합성 핵산 분자 및 이의 용도
US20220220189A1 (en) Compositions and methods for treatment of hemochromatosis
KR20250064736A (ko) 방광암 예방 또는 치료용 조성물
KR20250147284A (ko) 진세노사이드 또는 이의 유도체를 포함하는 지질나노입자 및 이의 용도
KR20240170771A (ko) 2차 또는 3차 구조를 갖는 폴리 a 테일을 포함하는 신규 핵산 구조체 및 이의 용도
KR20240165509A (ko) Rna/dna 하이브리드 구조를 포함하는 신규 핵산 구조체 및 이의 용도
WO2025211732A1 (fr) Nanoparticules lipidiques comprenant du ginsénoside ou un dérivé de celui-ci, et leur utilisation