WO2023131345A1 - Gene treatment drug and method for x-linked adrenoleukodystrophy - Google Patents

Gene treatment drug and method for x-linked adrenoleukodystrophy Download PDF

Info

Publication number
WO2023131345A1
WO2023131345A1 PCT/CN2023/071616 CN2023071616W WO2023131345A1 WO 2023131345 A1 WO2023131345 A1 WO 2023131345A1 CN 2023071616 W CN2023071616 W CN 2023071616W WO 2023131345 A1 WO2023131345 A1 WO 2023131345A1
Authority
WO
WIPO (PCT)
Prior art keywords
target
vector
sequence
abcd1
expression
Prior art date
Application number
PCT/CN2023/071616
Other languages
French (fr)
Chinese (zh)
Inventor
吴小兵
Original Assignee
北京锦篮基因科技有限公司
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 北京锦篮基因科技有限公司 filed Critical 北京锦篮基因科技有限公司
Publication of WO2023131345A1 publication Critical patent/WO2023131345A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • 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
    • 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
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/861Adenoviral vectors
    • 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
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/864Parvoviral vectors, e.g. parvovirus, densovirus
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0656Adult fibroblasts
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0658Skeletal muscle cells, e.g. myocytes, myotubes, myoblasts
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/067Hepatocytes
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • C12N2510/00Genetically modified cells
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/34Vector systems having a special element relevant for transcription being a transcription initiation element
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/50Vector systems having a special element relevant for transcription regulating RNA stability, not being an intron, e.g. poly A signal

Definitions

  • the present invention relates to the field of gene therapy, and more specifically to nucleic acids, expression constructs, and pharmaceutical compositions comprising the nucleic acid or constructs for treating X-chromosome-linked adrenoleukodystrophy (X-ALD) and for treating X-ALD.
  • ALD method X-chromosome-linked adrenoleukodystrophy
  • X-linked adrenoleukodystrophy is a common metabolic genetic disorder characterized by progressive demyelination of the central nervous system (CNS), adrenal insufficiency, and very long-chain saturated fatty acids accumulation.
  • X-ALD The clinical types of X-ALD include Addison type, cerebral type (childrenal type, adolescent type, and adult type), adrenomyeloneuropathy type (AMN) with or without intracranial demyelination, asymptomatic or presymptomatic type , and the heterozygous type.
  • the disease primarily affects the adrenal cortex and nervous system.
  • the pathological changes of the nervous system are diverse, ranging from inflammatory demyelination to axonal lesions involving the ascending and descending fiber tracts of the spinal cord. (J. Berger, Pathophysiology of X-linked adrenoleukodystrophy, Biochimie 98(2014) 135e142)
  • X-ALD disease is caused by mutations in the ABCD1 gene (ALD).
  • the gene is located at Xq28, consists of 10 exons, and encodes the transmembrane ATP-binding cassette transporter (ATP-binding cassette transporter, ABCD1) on the peroxisome.
  • the ABCD1 protein is responsible for the transport of CoA-activated very long-chain fatty acids (VLCFAs) from the cytoplasm into the peroxisome for degradation. Mutations in the ABCD1 gene will lead to the incapacity of this transmembrane protein, which in turn will cause the accumulation of VLCFA in cells, resulting in neurotoxicity and adrenal toxicity.
  • gene therapy based on recombinant AAV viral vectors is considered to be a safer approach than lentiviruses because it does not depend on the integration of the viral genome in the host cell nucleus.
  • AAV virus also has the advantages of wide host cell range and long expression time in vivo, which make it one of the most promising gene therapy vectors.
  • Cardiotoxicity has been observed in both non-clinical and clinical studies of the marketed AAV gene therapy drug 'Zolgensma'. According to the analysis, this is related to the high-dose use of AAV9 vector and the expression of the target gene in heart tissue.
  • Yi Gong et al. have also pointed out that although the gene construct used ABCD1 could be successfully delivered to CNS cells by IV injection, but significant cardiac tissue toxicity was induced due to the expression level of the transgene in liver and cardiac tissue much higher than that in CNS.
  • the ABCD1 gene is a widely expressed housekeeping gene, and its expression can be detected in most tissues.
  • ABCD1 protein is abundantly expressed in the central nervous system, adrenal gland, testis and other parts, indicating that the metabolism of very long-chain fatty acids in these tissues is relatively strong.
  • the ABCD1 gene is deleted, the ABCD1 protein level in the central nervous system, adrenal gland, testis and other parts decreases, resulting in the blockage of the very long-chain fatty acid metabolic pathway, causing cell oxidative stress and cell death. Therefore, gene therapy for X-ALD needs to take into account both the central and peripheral regions.
  • the inventors have made in-depth research, and by optimizing and combining multiple gene elements, they have proposed a method that can effectively alleviate the main affected organs (central nervous system and adrenal gland) of X-ALD after systemic administration. ) disease burden, and at the same time have a new AAV virus vector with low drug peripheral tissue toxicity, and its use.
  • the expression construct of the present invention realizes high-efficiency expression of ABCD1 through the optimized promoter and ABCD1 encoding nucleic acid; at the same time, by including an optimized combination of miRNA target sequences in the 3'UTR, the toxicity of transgene overexpression to peripheral organs is reduced , especially myocardial tissue toxicity.
  • the invention provides a nucleic acid encoding ABCD1 comprising: SEQ ID No: 1, or at least 90%, 95%, 96%, 97%, 98%, 99%, 99.1% thereof , 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8% identical nucleotide sequences.
  • the expression level (preferably, the expression level in mammalian cells) is increased by at least 200% or more, such as at least 300%, 320%, 330%, 340%, 350%, 360% %, 370%, 380%, 390%, or 400%.
  • the ABCD1-encoding nucleic acid according to the invention comprises a Kozak sequence at the 5' end. According to the in vitro transient expression test described in Example 1, it can be determined that the expression efficiency of the encoding nucleic acid is improved compared with the reference nucleic acid.
  • the present invention provides an expression construct comprising the ABCD1 encoding nucleic acid according to the present invention, especially comprising the nucleotide sequence of SEQ ID NO:1.
  • the expression construct according to the present invention further comprises a promoter, such as a constitutive promoter, operably linked to the ABCD1 encoding nucleic acid.
  • a promoter such as a constitutive promoter
  • the promoter comprises a nucleotide sequence selected from SEQ ID No: 2-5, or nucleotides having at least 95%, 96%, 97%, 98%, 99% or 99.5% identity therewith Sequence, especially the nucleotide sequence of SEQ ID No:2.
  • the expression construct according to the present invention further comprises a 3'UTR operably linked to said ABCD1 encoding nucleic acid.
  • the 3'UTR may comprise at least 1 copy (eg, 1-8 copies, such as 2, 3, or 4 copies) of the target sequence of a myocyte-specific miRNA and/or at least 1 copy (eg, 1-8 copies, eg, 1 or 2 copies) of the target sequence of the hepatocyte-specific miRNA.
  • the 3'UTR comprises at least 1 copy (such as 3 copies) of a myocyte-specific miRNA target sequence and at least 1 (such as 2 copies) of a hepatocyte-specific miRNA target sequence.
  • the at least one myocyte-specific miRNA target sequence and the at least one hepatocyte-specific miRNA target sequence may be included in the 3'UTR in various arrangements, but preferably, the myocyte-specific miRNA target sequence is Hepatocyte-specific miRNA target sequences spaced apart.
  • the miRNA target sequences can be connected directly, or can be separated by a few nucleotides, such as 1-5 nucleotides.
  • the muscle cell-specific miRNA target sequence that can be used in the present invention can be selected from miRNA1 target sequence, miRNA206 target sequence and combinations thereof; for example, the miRNA1 target sequence shown in SEQ ID NO:6 and the miRNA1 target sequence shown in SEQ ID NO:7 miRNA206 target sequence.
  • the hepatocyte-specific miRNA target sequence that can be used in the present invention can be a miRNA122 target sequence, such as the miRNA122 target sequence shown in SEQ ID NO:8.
  • the 3'UTR comprises miRNA target sequences arranged as follows: miRNA1 target sequence-miRNA122 target sequence-miRNA1 target sequence-miRNA122 target sequence-miRNA206 target sequence; or miRNA206 target sequence-miRNA122 target sequence- miRNA1 target sequence - miRNA122 target sequence - miRNA122 target sequence - miRNA1 target sequence.
  • the expression construct according to the present invention comprises a 3'UTR operably linked to the ABCD1 encoding nucleic acid, said 3'UTR comprising the nucleotide sequence of SEQ ID NO:12.
  • the invention provides a vector comprising an expression construct according to the invention.
  • the vector is a plasmid or viral vector.
  • the vector is an adeno-associated viral (AAV) vector.
  • the invention also provides a host cell, preferably a mammalian cell, comprising an expression construct or vector according to the invention.
  • the present invention provides a recombinant adeno-associated virus (AAV) vector, wherein said recombinant AAV vector comprises in its genome an expression construct according to the present invention.
  • the recombinant AAV vector comprises in its genome: (a) 5' and 3' AAV inverted terminal repeat (ITR) sequences, and (b) an expression construct located between the 5' and 3' ITR, wherein said expression construct comprises the following elements functionally linked to each other in the direction of transcription: a promoter; a polynucleotide encoding human ABCD1; at least one miRNA target sequence; one or more terminators; and one or more polyA signal sequences .
  • the polyA signal sequence is selected from the group consisting of SV40 late polyA sequence, rabbit ⁇ -globin polyA sequence, and bovine growth hormone polyA sequence, more preferably bovine growth hormone polyA sequence, such as the nucleotide sequence of SEQ ID NO:13.
  • the recombinant AAV viral vector according to the present invention may be a ssAAV vector or a scAAV vector.
  • a recombinant AAV viral vector according to the invention comprises a wild-type AAV2 ITR sequence, or in the case of scAAV vectors, one of said ITRs is a wild-type AAV2 ITR sequence and the other of said ITRs lacks a functional AA2 ⁇ ITR sequence of terminal melting sites (trs) and optionally D sequence.
  • the recombinant AAV viral vector according to the present invention may comprise capsid proteins from any AAV serotype, such as AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9 serotypes, especially AAV9 serotypes .
  • the recombinant AAV vector is an AAV2/9 vector having AAV2 ITR sequence and AA9 capsid protein.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an expression construct according to the present invention, a vector according to the present invention or a recombinant AAV viral vector according to the present invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition may be formulated for different routes of administration, such as intraperitoneal, intramuscular, intraarterial, intravenous, intrathecal or intracerebroventricular administration, as desired.
  • the pharmaceutical composition is an intravenous injection formulation.
  • the formulation comprises a prophylactically or therapeutically effective amount of a recombinant AAV viral vector according to the invention.
  • the invention also relates to the use of an expression construct according to the invention or a vector according to the invention or a recombinant AAV viral vector according to the invention for expressing ABCD1 in a mammalian cell, cell line or cell population, or Use in the preparation of a medicament for expressing ABCD1 in mammalian cells, cell lines or cell groups.
  • the mammalian cells may be in vitro, ex vivo or in vivo.
  • the present invention also provides a method (such as an in vitro, in vivo, or ex vivo method) of expressing ABCD1 in mammalian cells, said method comprising using an expression construct according to the present invention or a method according to the present invention vector to transfect isolated mammalian cells, cell lines or cell populations.
  • the mammalian cell is a cell line, or a cell, such as a fibroblast, isolated from an ABCD1-deficient mammalian subject.
  • the impact of the construct, vector or recombinant AAV virus vector of the present invention on the expression and/or viability of the target protein of the cells can be monitored by testing the expression of the target protein and/or the growth rate of the cell in vitro or in isolated cells.
  • the present invention provides a method for treating or preventing X-chromosome-linked adrenoleukodystrophy (X-ALD) and/or improving symptoms associated with X-ALD, wherein the method comprises administering to a subject in need
  • the method comprises intraperitoneal, intramuscular, intraarterial, intravenous, intrathecal or intracerebroventricular administration, more preferably intravenous injection of a recombinant AAV viral vector according to the invention.
  • the recombinant AAV viral vector is an AAV9 vector.
  • the plasma and tissues of the subject especially the adrenal gland and cerebellum, the biochemical markers of X-ALD disease—very long chain fatty acid VLCFA (especially C26:0 fatty acid)——The content is reduced.
  • VLCFA very long chain fatty acid
  • autophagic activity stimulated by VLCFA accumulation in the subject's spinal cord is alleviated/improved by IV administration of a recombinant AAV viral vector of the invention.
  • the methods of the invention ameliorate/alleviate symptoms of progressive demyelination of the subject's CNS.
  • the present invention also provides an expression construct comprising a 3'UTR operably linked to a gene, wherein said 3'UTR comprises:
  • -miRNA1 target x2 ie, 2 copies of the miRNA1 target sequence
  • -miRNA206 target x1 ie, 1 copy of the miRNA206 target sequence
  • -miRNA122 target x2 ie, 2 copies of the miRNA122 target sequence
  • -miRNA1 target x2 i.e., 2 copies of the miRNA1 target sequence
  • -miRNA122 target x2 i.e., 2 copies of the miRNA122 target sequence
  • -miRNA206 target x1 i.e., 1 copy of the miRNA206 target sequence
  • SEQ ID NO:10 SEQ ID NO:10
  • -miRNA1 target x1 i.e., 1 copy of miRNA1 target sequence
  • -miRNA122 target x1 i.e., 1 copy of miRNA122 target sequence
  • -miRNA1 target x1 i.e., 1 copy of miRNA1 target sequence
  • miRNA122 target x1 i.e., 1 copy of miRNA122 target sequence
  • miRNA206 target x1 i.e., 1 copy of the miRNA206 target sequence
  • the expression construct according to the present invention can be used to selectively reduce the expression of a gene of interest operably linked thereto in muscle cells and liver cells, or to prepare a gene for selectively reducing the expression of a gene of interest in Use in drugs expressed in muscle cells and hepatocytes.
  • Figure 1 shows the schematic diagram of the expression construct used in the present invention ( Figure 1A) and the expression detection results of the optimized ABCD1 encoding nucleic acid ( Figure 1B).
  • ITR ITR sequence of AAV virus
  • CA CA promoter
  • CAR-Mut CAR promoter with mutation
  • hABCD1 natural human ABCD1 encoding nucleic acid
  • coABCD1 optimized ABCD1 encoding nucleic acid
  • EGFP enhanced green Fluorescent protein
  • polyA polyA signal sequence
  • 3'UTR 3' untranslated region.
  • the left panel of Figure 1B shows that the natural human ABCD1-encoding nucleic acid (hABCD1) and the optimized ABCD1-encoding nucleic acid (coABCD1) were transiently expressed in HEK293 cells, the cells were lysed after 48 hours, and the total cellular protein was extracted and separated by electrophoresis on 10% SDS-PAGE Protein bands (the upper left figure shows the ABCD1 protein band, and the lower left figure shows the actin band); the right figure in Figure 1B shows that the fluorescent color results of the bands separated by electrophoresis are relatively gray after grayscale scanning degree value.
  • Figure 2 shows a schematic diagram of the pscAAV-CAR-Gluc plasmid vector.
  • Figure 3 shows that in an assay based on cultured cells in vitro, compared with BHK-21 cells that were not transfected with the plasmid (i.e., blank control), the pscAAV-CAR-Gluc vector and the pscAAV-CAR-MutC-Gluc vector were transfected. Changes in Gluc levels measured in BHK-21 cells with vector, pscAAV-CAR-MutA-Gluc vector and pscAAV-CAR-MutG-Gluc vector. Among them, ** means p ⁇ 0.01.
  • Figure 4 shows that in the EGFP-based expression plasmid vector, the impact of the target sequence embedded in miRNA1, 122, and 206 on the expression of the target gene in muscle cells and liver cells was detected.
  • Figure 5 shows that in the expression plasmid vector based on the Gluc reporter gene, the effect of increasing the number of repetitions of the miRNA target sequence and changing the arrangement and combination of the miRNA target sequence on the expression of the target gene in muscle cells was detected.
  • Figure 6 shows that in the Gluc reporter gene-based expression plasmid vector, the effect of increasing the number of repeats of the miRNA target sequence and changing the arrangement and combination of the miRNA target sequence on the expression of the target gene in hepatocytes was detected.
  • Figure 7 shows a schematic diagram of the basic plasmid pRDAAV-CMV-EGFP used to construct the AAV plasmid vector.
  • Figure 8 shows that using fibroblasts from X-ALD patient 1 and patient 2, the impact of recombinant AAV9-coABCD1 virus transduction on the expression of ABCD1 in cells was determined in the cell slide immunofluorescence assay.
  • the red fluorescence shows the ABCD1 protein in the cytoplasm stained by the dylight549-labeled secondary antibody; the blue fluorescence shows the nuclei stained by dapi.
  • Figure 9 shows the effect of recombinant AAV9-coABCD1 virus transduction on cell growth and proliferation rate.
  • Figures 9A and 9B show the cell densities of patient 2 fibroblasts not inoculated with AAV9-coABCD1 and inoculated with AAV9-coABCD1 at 7 days in culture, respectively.
  • Figure 9C shows the cell density of normal human fibroblasts in culture for 7 days.
  • Figure 10 shows the changes in the content of very long chain fatty acids in various tissues after injection of AA9-coABCD1 or AAV9-coABCD1-miT recombinant virus compared to untreated C57BL6J wild-type mice (WT). Among them, * means p ⁇ 0.05.
  • Figure 11 shows the histopathological changes in the test animals after injection of AA9-coABCD1 or AAV9-coABCD1-miT recombinant virus.
  • Figure 11A shows that inoculation with the optimized recombinant virus rAAV9-coABCD1-miT significantly reduced the pathological changes in the hearts and livers of the tested mice.
  • Black arrows indicate vacuolar degeneration of the heart and nuclear pyknosis and focal necrosis of the liver.
  • Figure 11B shows the autopsy results of a dead mouse in the test animal group inoculated with rAAV9-coABCD1, the histopathological examination results of the heart tissue in the above figure, the black arrow indicates the occurrence of large-area vacuolar degeneration of the myocardium; Red arrows indicate extensive intracardiac thrombus formation; lower panel shows histopathological examination of liver tissue, showing extensive hepatocyte necrosis and nuclear pyknosis.
  • FIG. 12 shows the evaluation of the effect of administering AAV9-coABCD1-miT on the behavior of X-ALD model mice in the rope grabbing experiment. The results showed that intravenous administration of AAV9-coABCD1-miT effectively improved the exercise capacity of model mice.
  • Figure 13 shows that 8 weeks after the optimized drug AAV9-coABCD1-miT was administered to X-ALD model mice, the content of very long chain fatty acids in each tissue was detected, and compared with unadministered X-ALD model mice and wild animals. normal mice for comparison. Among them, * indicates p ⁇ 0.05; NS indicates that the difference is not significant.
  • Figure 14 shows the histopathological results of the adrenal glands of X-ALD model mice treated with AAV9-coABCD1-miT tail vein injection 8 weeks after administration.
  • the red fluorescence shows the ABCD1 protein in the immunofluorescence-labeled cells; the blue fluorescence shows that the nuclei are stained.
  • Figure 15 shows that after AAV9-coABCD1-miT administration, LC3 ⁇ was immunofluorescently labeled on spinal cord tissue sections to evaluate the autophagy and improvement in the nervous system of X-ALD model mice.
  • Figure 16 shows that after intravenous injection of optimized (AAV9-coABCD1-miRT) and unoptimized drugs (AAV9-coABCD1) into normal wild-type mice (2 mice in each group), the Western Blot method was used to determine the ABCD1 protein content.
  • Figure 16A shows the Western Blot detection results, wherein the upper figure shows the ABCD1 band, and the lower figure shows the ⁇ -actin band; wherein, lanes 1 and 2 are mice administered with AAV9-coABCD1 (RD48-1 and RD48 -2); lanes 3-4 are mice administered with AAV9-coABCD1-miRT (RD49-2 and RD49-4); lanes 5 and 6 are normal mouse controls without administration (N1 and N2).
  • Figure 16B shows the quantification results obtained by analyzing the gray value of the Western Blot detection result pictures using the software ImageJ.
  • the invention discloses a gene therapy construct, a pharmaceutical composition and a method for treating X-ALD, especially the construction, preparation and application of a recombinant AAV vector for delivering ABCD1.
  • the expression "and/or”, when used to connect multiple items, means any one of the listed related items, or any and all possible combinations of a plurality or all of the listed related items.
  • recombinant adeno-associated virus can be represented by the AAV virus serotype from which the capsid is derived alone, or by the AAV virus serotype from which the capsid and genomic ITR sequences are derived.
  • the identifier "/" is used for separation, followed by the serotype of origin of the capsid and before the identifier "/" by the serotype of origin of the ITR.
  • the number 9 in the expression recombinant AAV9 indicates that the recombinant adeno-associated virus has a capsid from the AAV9 serotype; while the number before the identifier "/" in the expression recombinant AAV2/9 indicates that the recombinant adeno-associated virus has The wild-type or variant ITR sequence from AAV2, while the number after the identifier "/" indicates that the recombinant adeno-associated virus has a capsid protein from AAV9.
  • sequence identity is used to describe the similarity in sequence structure between two amino acid sequences or polynucleotide sequences.
  • sequences can be aligned for optimal comparison purposes (e.g., the first and second amino acid sequences or the first and second amino acid sequences can be aligned for optimal alignment). Gaps may be introduced into one or both of the first and second nucleic acid sequences or non-homologous sequences may be discarded for comparison purposes).
  • the length of the reference sequence being aligned is at least 30%, preferably at least 40%, more preferably at least 50%, 60% and even more preferably at least 70%, 80%, 90%, 100%.
  • amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide at the corresponding position in the second sequence, then the first and second sequences are identical at that position.
  • the comparison of sequences and the calculation of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • the Needlema and Wunsch ((1970) J. Mol. Biol. 48:444-453) algorithm (available at http://www.gcg.com available), use the Blossum 62 matrix or the PAM250 matrix with gap weights of 16, 14, 12, 10, 8, 6 or 4 and length weights of 1, 2, 3, 4, 5 or 6 to determine the distance between two amino acid sequences. percent identity.
  • using the GAP program in the GCG software package (available at http://www.gcg.com), using the NWSgapdna.CMP matrix and gap weights of 40, 50, 60, 70 or 80 and Length weights of 1, 2, 3, 4, 5 or 6 determine the percent identity between two nucleotide sequences.
  • a particularly preferred parameter set (and one that should be used unless otherwise stated) is the Blossum 62 scoring matrix with a gap penalty of 12, a gap extension penalty of 4, and a frameshift gap penalty of 5.
  • the term "host cell” refers to a cell into which an exogenous polynucleotide has been introduced, including the progeny of such cells.
  • the host cell is any type of cell that can be used to produce a recombinant AAV vector of the invention, for example, mammalian cells (such as HEK 293 cells suitable for production of recombinant AAV by a three-plasmid packaging system) and insect cells ( For example sf9 cells suitable for the production of recombinant AAV by the baculovirus packaging system).
  • regulatory sequence refers to a nucleic acid sequence that induces, represses, or otherwise controls the transcription of a protein of an encoding nucleic acid sequence to which it is operably linked. Regulatory sequences can be, for example, initiation sequences, enhancer sequences, intron sequences, and promoter sequences, among others.
  • exogenous or heterologous are used interchangeably when describing a nucleic acid or protein to mean that the nucleic acid or protein does not naturally exist in the chromosomal or host cell location in which it is found.
  • An exogenous nucleic acid sequence also refers to a sequence that is derived from and inserted into the same host cell or subject but exists in a non-native state, eg, the sequence is present in a different copy number, or is under the control of a different regulatory element.
  • an "isolated" polynucleotide eg, isolated DNA or isolated RNA
  • an "isolated" nucleic acid is enriched at least about 10-fold, 100-fold, 1000-fold, 10,000-fold or more relative to the starting material.
  • an "isolated" polypeptide refers to a polypeptide that is at least partially separated from at least some other components of the native organism or virus in which it is contained. In some embodiments, an “isolated” polypeptide is enriched at least about 10-fold, 100-fold, 1000-fold, 10,000-fold or more relative to the starting material.
  • an “isolated” or “purified” viral vector means that the viral vector has been partially separated from at least some components of the starting material comprising it. In some embodiments, an “isolated” viral vector is enriched at least about 10-fold, 100-fold, 1000-fold, 10,000-fold or more relative to the starting material.
  • viral vector refers to a viral particle (such as an AAV viral particle) capable of serving as a delivery vehicle for a nucleic acid of interest.
  • a viral vector comprises a capsid and a viral genome (for example, viral DNA) packaged therein, and the target nucleic acid to be delivered is inserted into the viral genome.
  • recombinant AAV viral vectors in order to generate recombinant virus particles that can deliver the nucleic acid of interest to tissues or cells, it is usually only necessary to retain the inverted terminal repeat (ITR) cis element in the genome, while the rest required for viral packaging Sequences can be provided in trans.
  • ITR inverted terminal repeat
  • the recombinant AAV viral vectors of the present invention comprise a capsid and a recombinant viral genome packaged therein, wherein the recombinant viral genome comprises or consists of one or more exogenous genes located between two AAV ITR sequences.
  • Source nucleotide sequence composition The two ITR sequences located at the 5' and 3' ends of the recombinant viral genome (i.e., 5'ITR and 3'ITR) may be the same or different.
  • AAV "inverted terminal repeat” refers herein to a cis-acting element from the AAV viral genome that plays an important role in the integration, rescue, replication, and genome packaging of the AAV virus.
  • the ITR sequence of the natural AAV virus contains a Rep protein binding site (Rep binding site, RBS) and a terminal unzipping site trs (terminal resolution site), which can be recognized by the Rep protein and generate a nick at the trs.
  • the ITR sequence can also form a unique "T" letter-shaped secondary structure, which plays an important role in the life cycle of the AAV virus.
  • AAV2 The earliest isolated AAV virus, AAV2, has "inverted terminal repeats" (ITRs) with a palindrome-hairpin structure of 145 bp located at both ends of the genome. Later, different ITR sequences were found in various serotypes of AAV viruses, but they all formed hairpin structures and had Rep binding sites.
  • ITRs inverted terminal repeats
  • Traditional recombinant AAV viral vectors based on these wild-type ITR sequences are generally single-stranded AAV vectors (ssAAV), and the viral genome is packaged in the AAV capsid in a single-stranded form.
  • the genome carried by the recombinant AAV virus vector obtained by packaging can be self-complementary to form a double chain (Wang Z et al., Gene Ther. 2003; 10(26):2105-2111; McCarty DM et al., Gene Ther. 2003; 10(26):2112-2118).
  • the virus thus packaged is a double-stranded AAV virus, that is, scAAV (self-complementary AAV) virus.
  • the packaging capacity of the scAAV viral vector is smaller, only half of the packaging capacity of the ssAAV viral vector, about 2.2kb-2.5kb, but the transduction efficiency after infection of cells is higher.
  • ITR in relation to AAV encompasses wild-type ITRs and variant ITRs.
  • Wild-type ITRs can be from any native AAV virus, such as an AAV2 virus.
  • the wild-type ITR contains a Rep protein binding site (Rep binding site, RBS) and a terminal unzipping site trs (terminal resolution site), which can be recognized by the Rep protein and generate a nick at trs.
  • the wild-type ITR sequence can form a unique "T" letter-shaped secondary structure, which plays an important role in the life cycle of AAV virus.
  • a variant ITR is a non-native ITR sequence which may, for example, be derived from any wild-type AAV ITR sequence and which comprises a deletion, substitution, and/or addition of one or more nucleotides relative to the wild-type ITR, and/ Or truncated, but still functional, ie, can be used to generate ssAAV viral vectors or scAAV viral vectors.
  • a variant ITR is an AAV ITR sequence (also referred to herein as a ⁇ ITR) that has been deleted for a functional trs site and optionally a D region sequence.
  • wild-type ITRs are combined with ⁇ ITRs to generate self-complementary recombinant AAV viral vectors (scAAV).
  • scAAV self-complementary recombinant AAV viral vectors
  • two wild-type ITRs are used in combination to generate single-chain recombinant AAV viral vectors (ssAAV).
  • the AAV proteins VP1, VP2 and VP3 are capsid proteins that interact to form the AAV capsid.
  • Different serotypes of AAV viruses have different tissue infection tropisms, and foreign genes can be transferred to specific organs and tissues by selecting the source serotype of the recombinant AAV virus vector capsid (Wu Z et al., Mol Ther.2006; 14(3):316-327).
  • the recombinant AAV virus vector can have different targeting properties by selecting the source serotype of the capsid.
  • the capsid of the recombinant AAV virus is from an AAV serotype that targets neuronal cells.
  • the recombinant AAV viral vector comprises a capsid from AAV9.
  • the recombinant AAV viral vector comprises a capsid from AAV9 and an ITR from AAV2.
  • treatment refers to medical intervention intended to alter the natural course of a disease in the individual being treated. Desired therapeutic effects include, but are not limited to, preventing the onset or recurrence of the disease, alleviating symptoms, reducing any direct or indirect pathological consequences of the disease, preventing metastasis, reducing the rate of disease progression, ameliorating or palliation of the disease state, and remission or improved prognosis.
  • the recombinant AAV virus of the present invention after administration to an ABCD1-deficient subject or an X-ALD patient, preferably after systemic administration, reduces the number of affected tissues (especially, the adrenal gland and central nervous system) of the subject. Nervous system) VLCFA content.
  • the recombinant AAV virus of the present invention improves central nervous system damage and/or adrenal damage in a subject after administration to an ABCD1-deficient subject or an X-ALD patient, preferably after systemic administration. In some embodiments, the recombinant AAV virus of the invention improves the exercise capacity of the subject after administration to an ABCD1-deficient subject or an X-ALD patient, preferably after systemic administration.
  • prevention includes the inhibition of the occurrence or development of a disease or symptoms of a particular disease.
  • subjects predisposed to developing X-ALD disease are candidates for prophylactic regimens.
  • prevention refers to medical intervention performed before at least one symptom of a disease occurs. Therefore, in one embodiment, prevention includes administering the gene therapy drug of the present invention in subjects with ABCD1 gene deficiency before the symptoms of X-ALD disease occur, so as to delay the development of the disease or prevent the appearance of the disease.
  • the prevention includes using the gene therapy drug of the present invention to improve the abnormal phagocytosis stimulated by VLCFA in the nervous system, thereby preventing the occurrence of related spinal cord axonal lesions.
  • the gene construct according to the present invention has at least one or more optimized gene elements as follows: (1) optimized ABCD1 encoding nucleic acid (also referred to as coABCD1 for short); (2) optimized constitutive promoter; and (3) optimized combination of miRNA target sequences.
  • the ABCD1-encoding nucleic acid contained in the construct of the present invention may be any polynucleotide capable of encoding functional ABCD1 protein activity. However, in order to facilitate expression in mammalian cells, it is advantageous to optimize the nucleic acid sequence of the nucleic acid encoding the ABCD1 polypeptide.
  • the ABCD1 encoding nucleic acid used in the expression construct of the present invention comprises the polynucleotide sequence of SEQ ID NO: 1, or has at least about 95%, about 96%, about 97%, 98%, Polynucleotide sequences with 99% or greater nucleotide sequence identity.
  • the optimized ABCD1-encoding nucleic acid with respect to a reference nucleic acid (for example, a natural human ABCD1-encoding nucleic acid, such as a nucleic acid having the nucleotide sequence shown in SEQ ID NO: 15), is in an operably linked composition
  • Assays for determining protein expression levels are known in the art. Any such assay can be used by one skilled in the art to determine the degree of optimization achieved in terms of expression efficiency of an optimized ABCD1-encoding nucleic acid compared to a reference nucleic acid.
  • the optimized ABCD1-encoding nucleic acid according to the present invention may comprise a Kozak sequence located upstream of the start codon at the 5' end.
  • the Kozak sequence used in the present invention may be a consensus sequence defined as GCCRCC, wherein R is a purine (ie A or G), and wherein said sequence is located upstream of the start codon.
  • the construct of the invention comprises a Kozak sequence, wherein said Kozak sequence has a 5'-GCCACC-3' sequence. Other different Kozak sequences can also be used in the constructs of the invention.
  • the construct of the present invention may contain any promoter that can be used to promote the expression of an ABCD1-encoding nucleic acid in a mammalian cell of interest.
  • the mutant constitutive promoter CAR-Mut according to the invention is included.
  • the CAR-Mut constitutive promoter of the present invention can efficiently promote the expression of exogenous genes in various tissues, so it is especially suitable for use in the treatment method of the present invention, so as to take into account the peripheral and central therapeutic purposes of X-ALD.
  • the construct of the invention comprises a CAR-Mut promoter comprising a polynucleotide selected from the group consisting of:
  • the polynucleotide has a mutated nucleotide C or G or A at nucleotide 568 of SEQ ID NO:5 or the corresponding position, more preferably T is mutated to C.
  • the mutant promoter of the present invention increases the expression of a gene of interest functionally linked thereto, e.g., makes said gene of interest Gene expression is increased by 1%-70%, eg, at least 5%, 10%, 20%, 30%, 40%, or at least 50%, 60%.
  • the mutant promoter of the present invention increases the expression of the gene of interest functionally linked thereto in mammalian cells or tissues, for example, increases the expression of the gene of interest in mammals relative to the reference promoter. Expression in peripheral tissues and/or central nervous tissues, especially in the central nervous system.
  • the mammal is a human or a non-human mammal, eg, a mouse, a rat and a non-human primate.
  • the promoter comprises a nucleotide sequence selected from any one of SEQ ID NOs: 2 to 4, or differs therefrom by one or several nucleotide substitutions, deletions and/or additions and has Nucleotide sequences with equivalent promoter activity.
  • the promoter comprises or consists of the nucleotide sequence of SEQ ID NO:2.
  • any promoter functional assay known in the art such as the luciferase reporter gene expression assay in Example 1
  • the reference promoter such as SEQ ID NO: 5
  • the promoter to be tested under the same test conditions, compared with the reference promoter (such as SEQ ID NO: 5), if the promoter to be tested has the same or substantially the same activity, for example, the activity of the reference promoter ⁇ 10% , preferably ⁇ 5%, or more preferably ⁇ 1%, then the promoter to be tested can be considered to have equivalent promoter activity.
  • MicroRNA is an endogenous small non-coding RNA that can regulate cellular gene expression in a sequence-specific manner, usually by repressing translation of target mRNAs. Endogenous miRNAs can suppress the expression of transgenes in expression cassettes that contain their perfectly complementary target sequences. The level of repression is related to factors such as the promoter used to express the construct, the corresponding miRNA abundance in the target tissue cells, and the like. According to in-depth research, the inventors found that in the expression construct containing a constitutive promoter, embedding specific types, quantities and arrangements of miRNA target cells in the 3' UTR can increase the safety of gene therapy based on the expression construct sex.
  • the expression constructs of the invention further comprise one or more miRNA target sequences located in the 3'UTR operably linked to the coding nucleic acid sequence of interest.
  • miRNA target sequences located in the 3'UTR operably linked to the coding nucleic acid sequence of interest.
  • inclusion of miRNA target sequences in expression constructs will allow for the modulation (eg, inhibition) of expression of a gene of interest in cells and tissues producing the corresponding miRNA.
  • the expression constructs of the invention comprise one or more miRNA target sequences, so that the expression of ABCD1 can be downregulated in a cell type specific manner.
  • miRNA target sequences that can be used in the present invention include target sequences of muscle cell-specific miRNA and target sequences of hepatocyte-specific miRNA.
  • the muscle cell-specific miRNA target sequence that can be used in the present invention can be selected from miRNA1 target sequence, miRNA206 target sequence and combinations thereof; for example, the miRNA1 target sequence shown in SEQ ID NO:6 and the miRNA1 target sequence shown in SEQ ID NO:7 miRNA206 target sequence.
  • the liver cell-specific miRNA target sequence that can be used in the present invention can be a miRNA122 target sequence, such as the miRNA122 target sequence of SEQ ID NO:8.
  • the liver cell-specific miRNA target sequence contained in the expression construct according to the present invention can be at least one or more, and preferably 2-4, which can be connected in series or arranged at intervals with the muscle cell-specific miRNA target sequence .
  • the muscle cell-specific miRNA target sequence contained in the expression construct according to the present invention can be at least one or more, and preferably 2-4, which can be connected in series or combined with the liver cell-specific miRNA target sequence. Sequence spaced.
  • the miRNA target sequences can be connected directly, or can be separated by a few nucleotides, such as 1-5 nucleotides.
  • an expression construct according to the invention comprises a 3' UTR operably linked to said ABCD1 encoding nucleic acid.
  • the 3'UTR comprises at least 1 copy (eg, 1-8 copies, such as 2, 3, or 4 copies) of the target sequence of a myocyte-specific miRNA and/or at least 1 copy (eg, 1 - 8 copies, eg 1 or 2 copies) of the target sequence of the hepatocyte-specific miRNA.
  • the 3'UTR comprises at least 1 copy (such as 3 copies) of a myocyte-specific miRNA target sequence and at least 1 (such as 2 copies) of a hepatocyte-specific miRNA target sequence.
  • the at least one myocyte-specific miRNA target sequence and the at least one hepatocyte-specific miRNA target sequence may be present in various arrangements in the 3'UTR, but preferably, the myocyte-specific miRNA target sequence Spaced with hepatocyte-specific miRNA target sequences.
  • the 3'UTR comprises miRNA target sequences arranged as follows: miRNA1 target sequence-miRNA122 target sequence-miRNA1 target sequence-miRNA122 target sequence-miRNA206 target sequence; or miRNA206 target sequence-miRNA122 target sequence- miRNA1 target sequence - miRNA122 target sequence - miRNA1 target sequence.
  • the expression construct according to the present invention comprises a 3'UTR operably linked to the ABCD1 encoding nucleic acid, said 3'UTR comprising the nucleotide sequence of SEQ ID NO:12.
  • the invention provides expression constructs.
  • the expression construct of the present invention can be advantageously used for gene therapy of X-ALD disease.
  • the expression construct of the invention comprises the following elements functionally linked to each other in the direction of transcription:
  • -encoding ABCD1 nucleic acid preferably, according to the optimized ABCD1 encoding nucleic acid of the present invention, more preferably comprises the nucleic acid of nucleotide sequence shown in SEQ ID NO:1,
  • the expression construct also includes two ITR sequences.
  • the expression construct may comprise elements arranged as follows: 5'ITR-promoter-ABCD1 coding sequence-miRNA target sequence-polyA-3'ITR.
  • the 5'ITR and 3'ITR are the same.
  • the 5'ITR and the 3'ITR are different and one (preferably the 3'ITR) is a ⁇ ITR lacking a functional trs site.
  • the 5'ITR and 3'ITR in the expression construct are identical and both comprise or consist of the AAV2 ITR sequence.
  • the promoter used in the expression construct of the present invention may be the CAR-Mut promoter described in any of the above embodiments of the present invention.
  • the promoter comprises or consists of the nucleotide sequence of SEQ ID No:2.
  • the promoter comprises or consists of the nucleotide sequence of SEQ ID NO:3.
  • the promoter comprises or consists of the nucleotide sequence of SEQ ID NO:4.
  • the ABCD1-encoding nucleic acid used in the expression construct of the present invention may be the ABCD1-encoding nucleic acid described in any of the above embodiments of the present invention.
  • the encoding nucleic acid comprises or consists of the nucleotide sequence of SEQ ID NO:1.
  • the miRNA target sequence used in the expression construct of the present invention may be the miRNA target sequence described in any of the above embodiments of the present invention, in particular, at least one hepatocyte-specific miRNA target sequence according to the present invention and at least one muscle specific miRNA target sequence according to the present invention.
  • Cell-specific miRNA target sequence preferably, comprises the nucleotide sequence of SEQ ID NO:12.
  • Transcription terminators useful in the present invention include any nucleic acid sequence that can terminate translation of a nucleic acid.
  • the terminator can be "TAG", “TGA” or “TAA”, and the corresponding RNA sequence is “UAG”, “UGA” or “UAA”.
  • the expression construct of the invention further comprises at least one polyA tail located downstream of the ABCD1 encoding nucleic acid and the miRNA target sequence.
  • Any suitable polyA sequence may be used, including but not limited to hGHpolyA, BGHpolyA, SV40 late polyA sequence, rabbit ⁇ -globin polyA sequence, or any variant thereof.
  • polyA is BGHpolyA, such as polyA shown in SEQ ID NO: 13, or has at least 80%, 85%, 90%, 95%, 96%, 97% with SEQ ID NO: 13, A polyA polynucleotide sequence having 98% or 99% nucleotide sequence identity.
  • the invention also provides vectors comprising expression constructs of the invention.
  • the vector is a plasmid (eg, a plasmid used for recombinant viral particle production).
  • the vector is a viral vector, such as a recombinant AAV vector or a baculovirus vector.
  • the genome of the recombinant AAV vector is single-stranded (eg, single-stranded DNA).
  • the genome of the recombinant AAV vector is self-complementary.
  • the present invention also provides host cells, such as mammalian cells or insect cells, comprising the expression construct or vector of the present invention.
  • the cells can be used to produce recombinant AAV viruses.
  • the invention provides recombinant AAV vectors.
  • the recombinant AAV vectors of the present invention are particularly useful for X-ALD disease or ameliorating symptoms associated therewith.
  • the recombinant AAV vector comprises a capsid and nucleic acid located within the capsid, also referred to herein as the "genome of the recombinant AAV vector.”
  • the genome of the recombinant AAV vector contains multiple elements, including but not limited to two inverted terminal repeats (ITRs, i.e., 5'-ITR and 3'-ITR), and other elements located between the two ITRs, including the promoter , a heterologous gene, and a polyA tail.
  • ITRs inverted terminal repeats
  • at least one miRNA target sequence may also be included between the two ITRs.
  • adeno-associated virus includes, but is not limited to, AAV of any serotype, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 AAV, and AAV with AAV with artificially altered capsid proteins.
  • AAV adeno-associated virus
  • the genome sequences of various serotypes and artificial AAVs and their native inverted terminal repeat (ITR) sequences, Rep proteins and capsid cap proteins are known in the art. These sequences can be found in public databases such as GenBank or in the literature.
  • the present invention provides a recombinant AAV viral vector comprising a capsid, wherein the capsid is composed of a capsid protein capable of crossing the blood-brain barrier, such as AAV9, AAVPHP.B, AAVPHP.eB capsid protein.
  • the recombinant AAV vectors of the invention transduce cells of the central nervous system (CNS), including neuronal cells and glial cells, as well as peripheral non-neuronal cells.
  • recombinant AAV vectors are capable of targeting and transducing neuronal cells, astrocytes, and microglia after systemic administration.
  • the recombinant AAV vector is capable of targeting and transducing the peripheral organs and central nervous system of a subject following systemic administration.
  • the recombinant AAV vector is capable of targeting and transducing multiple tissues (e.g., brain, spinal cord, adrenal gland) of a subject following systemic administration, and preferably, is compatible with non-administered recombinant AAV vectors.
  • the recombinant AAV vector resulted in higher expression of the foreign gene of interest (the gene encoding ABCD1 in this application) and/or reduced VLCFA levels in the targeted and transduced tissues compared to control subjects.
  • the therapeutic efficacy of the recombinant AAV vector or gene drug of the present invention can be detected by decreasing the cellular level of saturated linear VLCFA (C24:0 and C26:0, especially C26:0).
  • the recombinant AAV vectors of the invention have a capsid from an AAV9 serotype (also referred to herein as an AAV9 vector); preferably, the recombinant AAV vector has a wild-type or capsid from AAV2 in its genome.
  • AAV9 serotype also referred to herein as an AAV9 vector
  • Variant ITR sequences also referred to herein as AAV2/9 vectors.
  • the two ITR sequences of the recombinant AAV vector of the present invention are full-length ITRs (for example, about 125-145 bp in length, and contain a functional Rep binding site (RBS) and a terminal melting site ( trs)).
  • full-length functional ITRs are used to produce single-chain recombinant AAV vectors (ssAAV).
  • one of the ITRs of the recombinant AAV vector is truncated.
  • truncated ITRs lack functional terminal melting sites trs and are used to produce self-complementary recombinant AAV vectors (scAAV vectors).
  • the present invention provides a recombinant adeno-associated virus (AAV) vector, wherein said recombinant AAV vector comprises in its genome: 5' and 3' AAV inverted terminal repeat (ITR) sequences, and located at Expression construct according to the invention between the 5' and 3'ITR.
  • AAV adeno-associated virus
  • the amount of ABCD1 gene expression or the amount of ABCD1 protein expression in the cells is increased compared with that before the administration , thereby reducing the level of VLCFA in the cell, eg by at least 1-3 fold.
  • the recombinant viral vector of the present invention after being administered to a subject, results in a decrease in VLCFA levels in the adrenal gland and CNS such as cerebellum, brain, spinal cord, for example, at least 0.5 times lower than before administration, for example At least 1-3 times. Determination of this fold reduction can be performed according to standard methods known in the art for the quantification of VLCFAs.
  • AAV vector packaging systems mainly include three-plasmid co-transfection system, adenovirus as helper virus system, Herpes simplex virus type 1 (HSV1) as helper virus packaging system, and baculovirus-based packaging system. system.
  • HSV1 Herpes simplex virus type 1
  • baculovirus-based packaging system baculovirus-based packaging system.
  • Each packaging system has its own characteristics, and those skilled in the art can make appropriate choices according to needs.
  • the three-plasmid co-transfection packaging system is the most widely used AAV vector packaging system because it does not require helper virus and has high safety. It is also the mainstream production system in the world.
  • the slight disadvantage is that the absence of an efficient large-scale transfection method limits the application of the three-plasmid transfection system in the large-scale preparation of AAV vectors.
  • the recombinant AAV viral vectors of the invention can be produced using any suitable method known in the art.
  • the recombinant AAV virus of the present invention is produced using a three-plasmid packaging system.
  • the recombinant AAV virus of the present invention is produced using a baculovirus packaging system.
  • the present invention provides a pharmaceutical composition comprising a recombinant AAV viral vector of the present invention.
  • the pharmaceutical composition of the present invention preferably comprises a pharmaceutically acceptable excipient, diluent or carrier.
  • the pharmaceutical compositions of the present invention may be formulated in any suitable preparation form.
  • Suitable pharmaceutically acceptable excipients, diluents or carriers for formulation are well known in the art and include, for example, phosphate buffered saline, water, emulsions, such as oil/water emulsions, various types of Wet agent, sterile solution, etc.
  • Preparations can be formulated by conventional methods, and administered to subjects in appropriate doses.
  • Administration of a suitably formulated composition can be achieved in different ways, eg. Administration is by intravenous, intraperitoneal, subcutaneous, intramuscular, topical or intradermal. The particular route of administration depends, inter alia, on the type of carrier included in the pharmaceutical composition. The dosage regimen will be determined by the attending physician and other clinical factors.
  • the dosage for any one patient will depend on many factors, including the patient's size, body surface area, age, sex, the particular active agent to be administered, the timing and route used, and the type and phase of the drug used. Infections or diseases, general health conditions, and combinations of other medications.
  • the pharmaceutical compositions of the present invention may include a second active agent.
  • the second active agent is a drug for treating or alleviating X-ALD, or a component capable of reducing side effects when the drug is administered.
  • compositions of the present invention may be administered by any suitable route, including systemic administration and topical administration.
  • the pharmaceutical composition of the present invention is for systemic administration, especially intravenous administration.
  • the present invention provides a pharmaceutical composition comprising a recombinant AAV vector of the present invention, wherein said pharmaceutical composition is an intravenous formulation, or a lyophilized stable formulation suitable for formulation as an intravenous formulation.
  • the pharmaceutical composition of the present invention is suitable for local administration, for example, directly in or near the organ or tissue to be treated in a subject.
  • the present invention provides a pharmaceutical composition comprising a recombinant AAV vector of the present invention, wherein said pharmaceutical composition is a formulation suitable for topical administration.
  • the present invention relates to methods of treating diseases using the recombinant AAV vectors of the present invention or pharmaceutical compositions comprising the same.
  • the disease is X-ALD.
  • the disease is a defect in the ABCD1 gene.
  • the method comprises: administering a recombinant AAV vector or pharmaceutical composition of the invention to a subject in need thereof.
  • the recombinant AAV vector or pharmaceutical composition can be administered by any suitable route, including but not limited to, intramuscular, subcutaneous, intraspinal, intracerebroventricular, intrathecal, intravenous, intradiaphragmatic, intrathoracic, intraperitoneal.
  • the recombinant AAV vector or pharmaceutical composition of the present invention is delivered to a subject by systemic administration, especially intravenous administration.
  • the treatment is therapeutic.
  • the treatment is prophylactic.
  • the subject is a mammal, wherein the mammal is especially a human, primate, dog, horse, cow, especially a human subject.
  • treatment includes any one or more of: (1) arresting or delaying the onset of the disease; (2) lessening the severity of the disease; (3) lessening the severity of the disease; or prevent the onset and/or worsening of at least one symptom of the disease; (4) improve disease-related neurodegeneration and/or behavior of the subject; and (5) prolong the survival of the subject.
  • treatment of X-ALD while encompassing, does not require complete elimination of the disease or symptoms associated therewith.
  • Subjects who can be treated/prevented by the method of the present invention include Addison type, cerebral type (children's brain type, adolescent brain type and adult brain type), adrenomyeloneuropathy type (AMN) with or without intracranial demyelination, Asymptomatic or presymptomatic, and heterozygous.
  • the subject is a patient with cerebral ALD.
  • the subject is an AMN patient.
  • the subject is an asymptomatic patient.
  • the subject is an individual exhibiting symptoms associated with the onset of ALD or AMN, such as accumulation of high levels of VLCFA in plasma.
  • the subject is an individual at risk for ALD or AMN disease due to family history; or an individual whose genetic testing includes one or more mutations associated with ALD or AMN in the ABCD1 gene.
  • the present invention provides a method for treating or preventing X-chromosome-linked adrenoleukodystrophy (X-ALD) and/or improving symptoms associated with X-ALD, wherein the method comprises providing A subject is administered the recombinant AAV viral vector according to the present invention or the pharmaceutical composition according to the present invention.
  • said method comprises intraperitoneal, intramuscular, intraarterial, intravenous, intrathecal or intraventricular administration, more preferably intravenous injection, of said recombinant AAV viral vector.
  • the X-ALD gene therapy drug of the present invention (for example, according to the recombinant AAV virus vector of the present invention or according to the pharmaceutical composition of the present invention) can break through the blood-brain barrier, thereby reducing the extremely long chain in the multi-organ organs of the whole body. fatty acid levels.
  • the adeno-associated virus AAV9 is used as a vector, the drug can have a wider biodistribution, especially to cover the central nervous system, improve the level of very long chain fatty acids in the central nervous system, and prevent white matter lesions.
  • the X-ALD gene therapy drug of the present invention (for example, according to the recombinant AAV viral vector of the present invention or according to the pharmaceutical composition of the present invention) has increased the expression level of the drug in vivo through the optimization of the promoter and the coding gene, and the viral vector Genomic stability is enhanced, resulting in longer-lasting reductions in tissue and blood VLCFA levels.
  • the X-ALD gene therapy drug of the present invention (for example, the recombinant AAV virus vector according to the present invention or the pharmaceutical composition according to the present invention) has reduced toxicity to peripheral tissues and organs.
  • the X-ALD gene therapy drug of the present invention can reduce the toxicity of transgene overexpression to peripheral organs, especially myocardial tissue toxicity.
  • the X-ALD gene therapy drug of the present invention after intravenous injection into the ABCD1 gene-deficient model mice, can Efficient, continuous and stable expression of ABCD1 protein, and the ABCD1 protein produced by expression can participate in the degradation of extremely long-chain fatty acids in cells, reduce its accumulation in cells, and maintain it at a normal level, thereby eliminating cells caused by extremely long-chain fatty acids Various disease symptoms caused by excessive accumulation of fatty acids, to achieve the purpose of treatment.
  • the X-ALD gene therapy medicine of the present invention after intravenous injection and treatment of X-ALD model mice, the extremely long The level of chain fatty acids decreased significantly and returned to normal levels, and the very long chain fatty acids in peripheral tissues and blood also changed significantly.
  • the medicine of the present invention can be used in the treatment of X-ALD by intrathecal injection.
  • the treatment effect of intrathecal injection will be better and more significant for the improvement of abnormal indicators of the central nervous system.
  • the biodistribution of the AAV vector genome will be more concentrated in the central nervous system, and the proportion of vector biodistribution in peripheral organs such as the heart and liver is low (lower exposure), so the safety of the drug will be improved. Further improvement.
  • the present invention realizes the improvement of disease symptoms and simultaneously achieves good drug safety through a uniquely designed and constructed gene expression construct. Below in conjunction with specific embodiment and accompanying drawing, the present invention will be further described:
  • Embodiment 1 ABCD1 gene coding sequence optimization
  • CDS ABCD1 coding sequence
  • the nucleotide sequence of the gene encoding hABCD1 (UniProtKB-P33897) was optimized, and the optimized sequence for improving the expression efficiency of hABCD1 was determined by in vitro expression in HEK293 cell line and detection of ABCD1 protein expression level by WESTERN BLOT (coABCD1).
  • the wild-type ABCD1 coding nucleic acid sequence (from the sequence of NCBI accession number NM_000033.3) and the optimized coABCD1 coding nucleic acid sequence (SEQ ID NO: 1) were synthesized by GenScript Biotechnology Co., Ltd. The synthesized sequence was cloned into the pUC57 simple vector (GenScript Biotechnology, Nanjing). After that, the synthetic wild-type coding sequence and the optimized coding sequence were respectively cloned into the pAAV vector plasmid through KpnI and EcoRI sites, and embedded between the CA promoter and polyA (as shown in Figure 1A). After sequencing and identification, they were frozen and stored in the library and the plasmids were extracted for in vitro transfection experiments.
  • plasmids were miniprepped and HEK293 cell line was transiently transfected using Lipofectamine 2000.
  • the medium was changed 6 hours after transfection, and the cells were collected 48 hours later, and the total protein of the cells was lysed and extracted.
  • the protein bands were separated by 10% SDS-PAGE gel electrophoresis and transferred to PVDF membrane by wet transfer method.
  • Anti-ABCD1 antibody (1:2000, Abcam, ab197013) was used to incubate overnight.
  • ECL-enhanced chemiluminescence reagent (Sanko, C500044) was used for ECL luminescent color development.
  • the color rendering results were processed and analyzed by grayscale scanning.
  • CA promoter composed of the enhancer sequence of human CMV virus and the basal promoter of chicken ⁇ -actin protein
  • the 3' end of the sequence was introduced into the human TATA box binding protein-related factor 1 gene (GenBank: NG_012771.2)
  • the intron sequence from position 62804 to position 62890 was named CAR promoter.
  • CAR-MutC with mutation T568C, the sequence is shown in SEQ ID NO: 2
  • CAR-MutA with mutation T568A, sequence shown in SEQ ID NO: 3
  • CAR-MutG with mutation T568G, sequence shown in SEQ ID NO: 4
  • the pscAAV-CAR-Gluc plasmid vector shown in Figure 2 was constructed, including:
  • Gluc the nucleotide sequence encoding the luciferase reporter gene
  • BGH polyA bovine growth hormone
  • the CAR promoter in the pscAAV vector was replaced with the CAR-Mut promoter sequence to obtain the pscAAV-CAR-Mut-Gluc vector.
  • the well-grown BHK-21 cells were passaged to 24-well plates, and when the density reached 60%, Lipofectamine2000 (Invitrogen, USA) was used to transfect pscAAV-CAR-Gluc, pscAAV-CAR-MutC-Gluc, pscAAV according to the manufacturer's instructions - 3 wells each for CAR-MutA-Gluc and pscAAV-CAR-MutG-Gluc. 48 hours after transfection, 100 ⁇ L of the supernatant was taken from each well, the Gluc level was detected with a Glomax96 microplate luminometer (Promega), and data analysis was performed using the detector software.
  • Lipofectamine2000 Invitrogen, USA
  • the target sequences of miRNA1, 122, and 206 were embedded, and whether the miRNA target sequences were effective in down-regulating the expression of the target gene in cell lines derived from different tissues expected effect.
  • plasmid vectors based on EGFP expression constructs.
  • the CMV promoter drives EGFP gene expression
  • the miRNA1 target sequence miRNA1 Target
  • miRNA206 target sequence miRNA206 Target
  • miRNA122 target sequence miRNA122 Target
  • the plasmids were transiently transfected in C2C12 cell line (myoblasts, preserved by Jinlan Laboratory) and Huh7 cell line (human liver cancer cells, preserved by Jinlan Laboratory). After 48 hours of transfection, the fluorescent signal of the cells was observed under a fluorescent microscope. The effects of different miRNA target sequences located in the 3'UTR on the expression of the target gene EGFP were compared.
  • the combination of miRNA1 and miRNA206 target sequences can significantly reduce the expression level of the target gene in skeletal muscle cells.
  • the miRNA122 target sequence can down-regulate the expression of target genes in hepatocytes.
  • the plasmid vector embedded with the miRNA target sequence had significantly down-regulated expression of the target gene in Huh7 cells and C2C12 cells, which indicated that the 3'UTR containing the miRNA target sequence had specific expression down-regulation ability.
  • the EGFP gene was replaced by the Gluc reporter gene, and the number of repeats of the miRNA target sequence was increased in the 3'UTR and its arrangement and combination were changed, thereby comparing miRNA 1, miRNA 206, and miRNA 122 targets The effect of different combinations of sequences on the expression of the target gene.
  • Transient transfection was performed on the C2C12 cell line (myoblast, preserved by Jinlan Laboratory) and the Huh7 cell line (human liver cancer cell, preserved by Jinlan Laboratory) with the expression plasmid containing the construct. After 48 hours of transfection, the fluorescent signal of the cells was observed.
  • the combination of the three miRNA target sequences also showed a better ability to down-regulate the target gene. Compared with the down-regulation ability of the single-copy miRNA combination, it increased the ability to inhibit the target gene by ⁇ 300%. The expression in hepatocytes was suppressed by more than 90%.
  • Embodiment 4 Construction of recombinant AAV virus
  • An AAV plasmid vector containing an optimized promoter CAR-MutC, an optimized target gene co-ABCD1, and an ABCD1 expression cassette inserted into the 3'UTR miRNA target sequence was constructed.
  • the constructed AAV plasmid vector contains:
  • the coABCD1 nucleic acid sequence was synthesized by GenScript Biotechnology Co., Ltd., and a KpnI restriction site and a Kozak sequence 5'-GCCACC-3' were added upstream of the synthetic sequence, and a taa stop codon and EcoRI enzyme were added downstream cut site.
  • the synthesized sequence was cloned into the pUC57 simple vector (GenScript Biotechnology, Nanjing) to obtain the pUC57-coABCD1 vector.
  • the pUC57-coABCD1 vector and the pRDAAV-CAR-Mut-EGFP vector were digested with KpnI and EcoRI respectively, the coABCD1 fragment and the pRDAAV-CAR-Mut-EGFP vector fragment with the EGFP reporter gene removed were recovered, and the two fragments were ligated and transformed into E.coli DH5 ⁇ competent cells (Qingke Xinye, Beijing) were screened and identified to obtain the pRDAAV-CAR-Mut-coABCD1 vector.
  • the artificially synthesized miRNA target fragment (comprising miRNA122, 206, 1 target sequence, sequence information see SEQ ID No.12) is cloned into between the EcoRI and SalI restriction sites of the pRD.AAV-CAR-Mut-coABCD1 vector
  • the pRD.AAV-CAR-Mut-coABCD1-miT vector was obtained.
  • Example 5 In vitro transduction and functional testing of recombinant AAV9 virus
  • Skin fibroblasts from two X-ALD patients were infected with the prepared recombinant AAV virus at a multiplicity of infection of 10000. Cells were fixed with formalin 72 hours after infection and immunofluorescent staining was performed. Cells were incubated overnight with Anti-ABCD1 antibody (diluted 1:100, Abcam), rinsed, washed with dylight549-labeled secondary antibody for 1 hour, counterstained with dapi, and mounted. Red fluorescence and blue fluorescence were observed under a fluorescence microscope.
  • Skin fibroblasts from X-ALD patients were infected with the prepared recombinant AAV virus at a MOI of 50000. Cell density was observed on day 7 after infection.
  • Example 6 A single tail vein injection of AAV9-coABCD1-miT and AAV9-coABCD1 interferes with C57BL6/J wild-type mice
  • the adeno-associated virus vector containing the ABCD1 expression construct before and after optimization was injected into the tail vein of C57BL6J wild-type mice at a dose of 1.5E+14vg/kg, with 4 mice in each group. Except for one mouse in the unoptimized group that died 3 weeks after administration, the remaining 7 mice were killed 4 weeks after injection, and samples were collected for histopathology and the content of very long chain fatty acids.
  • the histopathological changes in vivo and the changes of very long chain fatty acids before and after 3'UTR optimization are shown in Figures 10 and 11.
  • Necropsy was performed on mice in the non-optimized group that died 3 weeks after administration. Histopathological diagnosis revealed that the heart of the dead mouse had extensive vacuolar degeneration, obvious myocardial fibrosis, and thrombus in the cardiac cavity. Liver pathological examination revealed a large area of hepatocyte necrosis, which spread outwards mainly centered on the central vein. The above pathological changes are considered to be related to the toxicity of the test product. ( Figure 11B)
  • Example 7 AAV9-coABCD1-miT Tail Vein Injection Treats X-ALD Model Mice
  • a B6.129 mouse model in which the ABCD1 gene was knocked out was selected, and the model mouse was purchased from Jackson Lab. Due to the species differences of the mice, the deletion of ABCD1 did not cause serious physiological or behavioral effects in the model mice, but the model mice showed phenotypes such as decreased exercise ability at 10 weeks, and the fibroblasts and The levels of C26:0 in various tissues (adrenal gland, brain, etc.) and blood were significantly higher than those in wild-type mice, indicating that there is pathological accumulation of very long-chain fatty acids in the model mice. This process simulates the pathological process of X-ALD patients.
  • AAV9-coABCD1-miT was delivered by intravenous injection, and the model mice were treated reparatively, and compared with normal wild-type mice and untreated mice.
  • the optimized drug was administered intravenously to 8 X-ALD model mice of the same age at a dose of 5E+13vg/kg. There were 8 wild-type mice and X-ALD model mice of the same age without any intervention.
  • mice The locomotion of the mice was assessed using the grab rope method.
  • the test results can be seen from Figure 12, the average score of normal wild-type mice is 4.5 points; the average score of untreated X-ALD model mice is 0 points, and the mice that were put on the rope fell within 10s. It shows that under the development of the natural history of the diseased mice, the muscles will atrophy and become weak; the average score of the treated diseased mice is 3.7 points, these mice can grasp the rope for more than 30s, and some will try to climb On the rope, some can put the front paw and one hind paw on the rope. Compared with the untreated diseased mice, the treatment effect is very obvious, and the data results of the treatment group are not significantly different from those of the wild type . These results indicate that the optimized drug has a significantly superior therapeutic effect through intravenous administration.
  • the content of very long chain fatty acids was determined by HPLC-MS/MS. First, the experimental mice were dissected and weighed, and then each tissue was ground into powder by liquid nitrogen grinding, and the very long-chain fatty acids were extracted from the tissue through a series of rough extraction and fine extraction using the Matyash extraction method, and used The content of very long chain fatty acids was determined by HPLC-MS/MS, and the detection results are shown in Figure 13.
  • X-ALD model mice were treated by tail vein injection of AAV9-coABCD1-miT, and ABCD1 was immunofluorescently labeled on tissue sections 8 weeks after administration.
  • X-ALD activates autophagy due to the accumulation of very long-chain fatty acids.
  • Autophagy wraps and transports very long-chain fatty acids accumulated in various subcellular organelles to lysosomes. But very long-chain fatty acids must be degraded in peroxisomes, so autophagy does not improve the accumulation of very long-chain fatty acids.
  • the accumulation of very long-chain fatty acids continuously stimulates and activates autophagy, and thus, the autophagic flux is stagnant and ineffective.
  • X-ALD model mice were treated with AAV9-coABCD1-miT tail vein injection. Eight weeks after administration, the spinal cord tissue sections were immunofluorescently labeled with LC3 ⁇ (axonemal dynein light chain 2 ⁇ ). The results are shown in Figure 15. From the results shown in the figure, it can be seen that in the untreated group, the anterior and dorsal horns of the spinal cord showed strong autophagy protein signals of myelin sheath, indicating that autophagy activation was obvious. The autophagy signal in the anterior horn of the spinal cord decreased significantly in the treatment group.
  • the trend of decreasing autophagy signal can also be observed, although the decrease degree is not as good as that in the forefoot area of the spinal cord (this is because AAV9 is more tropistic to the anterior horn cells after intravenous injection), but it can be clearly compared
  • the autophagy activity in the spinal cord decreased after treatment. It shows that after treatment, the problem of autophagy pathway disorder can be improved.
  • Embodiment 8 Evaluation of the safety and optimization degree of drugs
  • the protein content of ABCD1 was determined by Western Blot method to explore the safety and optimization degree of the drug.
  • the optimized (AAV9-coABCD1-miRT) and unoptimized drug (AAV9-coABCD1) were intravenously injected into normal wild-type mice, and compared with untreated normal mice.

Abstract

The present invention provides a nucleic acid and an expression construct for treating X-linked adrenoleukodystrophy (X-ALD), a pharmaceutical composition including the nucleic acid or the construct, and a method for treating X-ALD.

Description

用于X染色体连锁肾上腺脑白质营养不良的基因治疗药物和方法Gene therapy drug and method for X chromosome-linked adrenoleukodystrophy 技术领域technical field
本发明涉及基因治疗领域,更具体地涉及用于治疗X染色体连锁肾上腺脑白质营养不良(X-ALD)的核酸、表达构建体、以及包含所述核酸或构建体的药物组合物和治疗X-ALD的方法。The present invention relates to the field of gene therapy, and more specifically to nucleic acids, expression constructs, and pharmaceutical compositions comprising the nucleic acid or constructs for treating X-chromosome-linked adrenoleukodystrophy (X-ALD) and for treating X-ALD. ALD method.
背景技术Background technique
X连锁肾上腺脑白质营养不良(X-adrenoleukodystrophy,X-ALD)是一种常见的代谢遗传疾病,表现为中枢神经系统(CNS)的进行性脱髓鞘、肾上腺功能不全、和极长链饱和脂肪酸的累积。X-linked adrenoleukodystrophy (X-ALD) is a common metabolic genetic disorder characterized by progressive demyelination of the central nervous system (CNS), adrenal insufficiency, and very long-chain saturated fatty acids accumulation.
X-ALD的临床类型包括Addison型,脑型(儿童脑型、青少年脑型和成人脑型),伴或不伴颅内脱髓鞘的肾上腺脊髓神经病型(AMN)、无症状或症状前期型、以及杂合子型。该疾病主要影响肾上腺皮质和神经系统。神经系统的病理改变具有多样性,可以是炎性脱髓鞘,也可以表现为累及脊髓上行和下行纤维束的轴索病变。(J.Berger,Pathophysiology of X-linked adrenoleukodystrophy,Biochimie 98(2014)135e142)The clinical types of X-ALD include Addison type, cerebral type (childrenal type, adolescent type, and adult type), adrenomyeloneuropathy type (AMN) with or without intracranial demyelination, asymptomatic or presymptomatic type , and the heterozygous type. The disease primarily affects the adrenal cortex and nervous system. The pathological changes of the nervous system are diverse, ranging from inflammatory demyelination to axonal lesions involving the ascending and descending fiber tracts of the spinal cord. (J. Berger, Pathophysiology of X-linked adrenoleukodystrophy, Biochimie 98(2014) 135e142)
X-ALD疾病由ABCD1基因(ALD)突变引起。该基因定位于Xq28,由10个外显子组成,编码过氧化物酶体上的跨膜ATP结合盒转运蛋白(ATP-binding cassette transporter,ABCD1)。ABCD1蛋白负责转运CoA活化的极长链脂肪酸(VLCFA)由胞质进入过氧化物酶体进行降解。ABCD1基因突变将导致该跨膜蛋白失能,进而引起VLCFA在细胞中蓄积,产生神经和肾上腺毒性。X-ALD disease is caused by mutations in the ABCD1 gene (ALD). The gene is located at Xq28, consists of 10 exons, and encodes the transmembrane ATP-binding cassette transporter (ATP-binding cassette transporter, ABCD1) on the peroxisome. The ABCD1 protein is responsible for the transport of CoA-activated very long-chain fatty acids (VLCFAs) from the cytoplasm into the peroxisome for degradation. Mutations in the ABCD1 gene will lead to the incapacity of this transmembrane protein, which in turn will cause the accumulation of VLCFA in cells, resulting in neurotoxicity and adrenal toxicity.
Nathalie Cartier等(Hematopoietic Stem Cell Gene Therapy with a Lentiviral Vector in X-Linked Adrenoleukodystrophy,Science 326,818(2009))报道了一种X-ALD的造血干细胞基因治疗方法。在该方法中,应用编码野生型ABCD1的慢病毒载体,转导来自患者的自体CD34+细胞进行遗传纠正,之后将经遗传纠正的细胞回输给患者。结果显示,由慢病毒介导的ABCD1在患者造血干细胞中的表达,带来了临床益处。然而,慢病毒基因治疗,在临床和临床前动物研究中都已经提示,存在因慢病毒基因插入宿主DNA中引入癌症的风险。Nathalie Cartier et al. (Hematopoietic Stem Cell Gene Therapy with a Lentiviral Vector in X-Linked Adrenoleukodystrophy, Science 326,818 (2009)) reported a hematopoietic stem cell gene therapy method for X-ALD. In this approach, autologous CD34+ cells from the patient are transduced with a lentiviral vector encoding wild-type ABCD1 for genetic correction, and the genetically corrected cells are then returned to the patient. The results showed that lentivirus-mediated expression of ABCD1 in patients' hematopoietic stem cells conferred clinical benefit. However, lentiviral gene therapy, both in clinical and preclinical animal studies, has suggested that there is a risk of introducing cancer due to lentiviral gene insertion into host DNA.
与慢病毒不同,基于重组AAV病毒载体的基因治疗,由于并不依赖于病毒基因组在宿主细胞核中的整合,而被认为是比慢病毒更安全的一种方式。此外,AAV病毒还存在宿主细胞范围广和在体内表达时间长等优势,这些都使其成为目前最有前景的基因治疗载体之一。Unlike lentiviruses, gene therapy based on recombinant AAV viral vectors is considered to be a safer approach than lentiviruses because it does not depend on the integration of the viral genome in the host cell nucleus. In addition, AAV virus also has the advantages of wide host cell range and long expression time in vivo, which make it one of the most promising gene therapy vectors.
Yi Gong等(Adeno-associated virus serotype 9-mediated gene therapy for X-linked Adrenoleukodystrophy(X-ALD),doi:10.1038/mt.2015.6)报道了一种应用AAV载体的X-ALD基因治疗方法。该方法使用重组腺相关病毒载体(rAAV)血清型9载体,将人ABCD1基因,通过ICV和IV途径,递送至小鼠的中枢神经系统(CNS)。然而,该文献的结果显示,在接受治疗后的模型小鼠体内,ABCD1蛋白和极长链脂肪酸水平与野生型小鼠相比,还存在不少的差距;例如,AAV9-ABCD1在ICV和IV注射后,虽然在小鼠的大脑和脊 髓中导致VLFA在总脂肪酸中的占比均降低,但不显著。推测原因为目的基因的转录与翻译能力低下所致。Yi Gong et al. (Adeno-associated virus serotype 9-mediated gene therapy for X-linked Adrenoleukodystrophy (X-ALD), doi:10.1038/mt.2015.6) reported a gene therapy method for X-ALD using AAV vector. The method uses a recombinant adeno-associated virus (rAAV) serotype 9 vector to deliver the human ABCD1 gene to the central nervous system (CNS) of mice through the ICV and IV routes. However, the results of this literature show that there are still many gaps in the levels of ABCD1 protein and very long-chain fatty acids in treated model mice compared with wild-type mice; After injection, although the proportion of VLFA in the total fatty acids in the brain and spinal cord of the mice was reduced, it was not significant. The reason is presumed to be the low transcription and translation ability of the target gene.
对于基因药物构建体,尽管可以通过提高剂量来达到所需的治疗效果,但是高剂量情况下的安全性风险是增加的。因此,这也导致剂量很难有较明显的提升空间。For gene drug constructs, although the desired therapeutic effect can be achieved by increasing the dosage, the safety risk in the case of high dosage is increased. Therefore, this also makes it difficult for the dose to have a more obvious room for improvement.
已上市的AAV基因治疗药物‘Zolgensma’,在非临床和临床研究中,均观察到了心脏毒性。据分析,这与大剂量使用AAV9载体和目的基因的心脏组织表达有关。此外,Yi Gong等在其后续的研究中(Intrathecal Delivery of rAAV9-ABCD1by Osmotic Pump in a Mouse Model of Adrenomyeloneuropathy,Molecular Therapy Volume 24,Supplement 1,May 2016),也已经指出,尽管其使用的基因构建体可以通过IV注射成功将ABCD1递送至CNS细胞,但由于转基因在肝脏和心脏组织中远高于CNS的表达水平,引发了明显的心脏组织毒性。Cardiotoxicity has been observed in both non-clinical and clinical studies of the marketed AAV gene therapy drug 'Zolgensma'. According to the analysis, this is related to the high-dose use of AAV9 vector and the expression of the target gene in heart tissue. In addition, in their follow-up study (Intrathecal Delivery of rAAV9-ABCD1 by Osmotic Pump in a Mouse Model of Adrenomyeloneuropathy, Molecular Therapy Volume 24, Supplement 1, May 2016), Yi Gong et al. have also pointed out that although the gene construct used ABCD1 could be successfully delivered to CNS cells by IV injection, but significant cardiac tissue toxicity was induced due to the expression level of the transgene in liver and cardiac tissue much higher than that in CNS.
鉴于目前在X-ALD治疗上存在的问题,本领域仍然需要提供新的X-ALD基因治疗手段。In view of the current problems in X-ALD treatment, there is still a need to provide new X-ALD gene therapy means in this field.
发明概述Summary of the invention
ABCD1基因是广泛表达的管家基因,绝大部分组织中均可以检测到其表达。ABCD1蛋白在中枢神经系统、肾上腺、睾丸等部位表达较为丰富,说明这些组织中极长链脂肪酸的代谢相对旺盛。而当ABCD1基因缺失时,中枢神经系统、肾上腺、睾丸等部位的ABCD1蛋白水平降低,导致极长链脂肪酸代谢路径阻塞,引起细胞氧化应激反应而导致细胞死亡。因此,X-ALD的基因治疗需兼顾中枢和外周。The ABCD1 gene is a widely expressed housekeeping gene, and its expression can be detected in most tissues. ABCD1 protein is abundantly expressed in the central nervous system, adrenal gland, testis and other parts, indicating that the metabolism of very long-chain fatty acids in these tissues is relatively strong. When the ABCD1 gene is deleted, the ABCD1 protein level in the central nervous system, adrenal gland, testis and other parts decreases, resulting in the blockage of the very long-chain fatty acid metabolic pathway, causing cell oxidative stress and cell death. Therefore, gene therapy for X-ALD needs to take into account both the central and peripheral regions.
一方面,鉴于中枢神经系统损伤通常都病情凶险,进一步降低中枢神经等组织中的极长链脂肪酸水平,是X-ALD基因治疗药物的开发方向。而另一方面,应当考虑到,基因治疗药物在不能使用特异性表达的情况下存在的相关毒性风险。因此,除了提升目的基因在靶器官中的表达之外,也需要在ABCD1蛋白耐受性差的组织脏器中实现特异性的表达降低,从而降低毒性风险。On the one hand, given that damage to the central nervous system is usually dangerous, further reducing the level of very long-chain fatty acids in the central nervous system and other tissues is the development direction of X-ALD gene therapy drugs. On the other hand, the risk of toxicity associated with gene therapy drugs that cannot be used for specific expression should be considered. Therefore, in addition to increasing the expression of the target gene in the target organ, it is also necessary to achieve a specific reduction in the expression of the ABCD1 protein in tissues and organs with poor tolerance, thereby reducing the risk of toxicity.
为了实现此目的,本发明人经过深入的研究,通过对多个基因元件进行优化和组合,提出了一种可以在全身给药后有效减轻X-ALD病的主要受累器官(中枢神经系统和肾上腺)的疾病负担,并同时具有低的药物外周组织毒性的新AAV病毒载体、及其用途。本发明的表达构建体,通过优化的启动子和ABCD1编码核酸,实现了ABCD1的高效表达;同时,通过在3’UTR中包含优化组合的miRNA靶序列,降低了转基因过表达对外周器官的毒性,尤其是心肌组织的毒性。In order to achieve this goal, the inventors have made in-depth research, and by optimizing and combining multiple gene elements, they have proposed a method that can effectively alleviate the main affected organs (central nervous system and adrenal gland) of X-ALD after systemic administration. ) disease burden, and at the same time have a new AAV virus vector with low drug peripheral tissue toxicity, and its use. The expression construct of the present invention realizes high-efficiency expression of ABCD1 through the optimized promoter and ABCD1 encoding nucleic acid; at the same time, by including an optimized combination of miRNA target sequences in the 3'UTR, the toxicity of transgene overexpression to peripheral organs is reduced , especially myocardial tissue toxicity.
因此,在一方面,本发明提供一种编码ABCD1的核酸,其包含:SEQ ID No:1,或与之具有至少90%、95%、96%、97%、98%、99%、99.1%、99.2%、99.3%、99.4%、99.5%、99.6%、99.7%、99.8%同一性的核苷酸序列。根据本发明的ABCD1编码核酸,在一些实施方案中,相比参比核酸(例如,天然人ABCD1编码核酸,例如,来自NCBI登录号NM_000033.3的核酸序列或SEQ ID NO:15的核酸序列),在组成型启 动子控制下,表达水平(优选地,在哺乳动物细胞中的表达水平)提升至少200%或更高,例如至少300%、320%、330%、340%、350%、360%、370%、380%、390%或400%。优选地,根据本发明的ABCD1编码核酸在5’端包含Kozak序列。可以根据实施例1所述的体外瞬时表达试验,确定所述编码核酸相比于参照核酸的表达效率提升。Accordingly, in one aspect, the invention provides a nucleic acid encoding ABCD1 comprising: SEQ ID No: 1, or at least 90%, 95%, 96%, 97%, 98%, 99%, 99.1% thereof , 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8% identical nucleotide sequences. ABCD1-encoding nucleic acid according to the present invention, in some embodiments, compared to reference nucleic acid (for example, natural human ABCD1-encoding nucleic acid, for example, from the nucleic acid sequence of NCBI accession number NM_000033.3 or the nucleic acid sequence of SEQ ID NO:15) , under the control of a constitutive promoter, the expression level (preferably, the expression level in mammalian cells) is increased by at least 200% or more, such as at least 300%, 320%, 330%, 340%, 350%, 360% %, 370%, 380%, 390%, or 400%. Preferably, the ABCD1-encoding nucleic acid according to the invention comprises a Kozak sequence at the 5' end. According to the in vitro transient expression test described in Example 1, it can be determined that the expression efficiency of the encoding nucleic acid is improved compared with the reference nucleic acid.
在再一方面,本发明提供了一种表达构建体,其包含根据本发明的ABCD1编码核酸,尤其是包含SEQ ID NO:1的核苷酸序列。In yet another aspect, the present invention provides an expression construct comprising the ABCD1 encoding nucleic acid according to the present invention, especially comprising the nucleotide sequence of SEQ ID NO:1.
在一些实施方案中,根据本发明的表达构建体,还包含与所述ABCD1编码核酸可操作性连接的启动子,例如组成型启动子。优选地,所述启动子包含选自SEQ ID No:2-5的核苷酸序列,或与其具有至少95%、96%、97%、98%、99%或99.5%同一性的核苷酸序列,尤其是SEQ ID No:2的核苷酸序列。In some embodiments, the expression construct according to the present invention further comprises a promoter, such as a constitutive promoter, operably linked to the ABCD1 encoding nucleic acid. Preferably, the promoter comprises a nucleotide sequence selected from SEQ ID No: 2-5, or nucleotides having at least 95%, 96%, 97%, 98%, 99% or 99.5% identity therewith Sequence, especially the nucleotide sequence of SEQ ID No:2.
在再一些实施方案中,根据本发明的表达构建体还包含与所述ABCD1编码核酸可操作连接的3’UTR。所述的3’UTR可以包含至少1个拷贝(例如,1-8个拷贝,例如2,3,或4个拷贝)的肌细胞特异性miRNA的靶序列和/或至少1个拷贝(例如,1-8个拷贝,例如,1个或2个拷贝)的肝细胞特异性miRNA的靶序列。优选地,所述3’UTR包含至少1个拷贝(如3个拷贝)的肌细胞特异性miRNA靶序列和至少1个(例如2个拷贝)的肝细胞特异性miRNA靶序列。所述至少一个肌细胞特异性miRNA靶序列,与所述至少一个肝细胞特异性miRNA靶序列,可以以各种排列方式包含在3’UTR中,但优选地,肌细胞特异性miRNA靶序列与肝细胞特异性miRNA靶序列间隔排列。miRNA靶序列之间可以直接连接,或可以间隔少数个核苷酸,例如1-5个核苷酸。可以用于本发明的肌细胞特异性miRNA靶序列,可以选自miRNA1靶序列、miRNA206靶序列及其组合;例如,SEQ ID NO:6所示的miRNA1靶序列和SEQ ID NO:7所示的miRNA206靶序列。可以用于本发明的肝细胞特异性miRNA靶序列,可以是miRNA122靶序列,例如SEQ ID NO:8所示的miRNA122靶序列。在一个优选的实施方案中,所述3’UTR包含如下排列的miRNA靶序列:miRNA1靶序列-miRNA122靶序列-miRNA1靶序列-miRNA122靶序列-miRNA206靶序列;或miRNA206靶序列-miRNA122靶序列-miRNA1靶序列-miRNA122靶序列-miRNA1靶序列。在一些更优选的实施方案中,根据本发明的表达构建体包含与ABCD1编码核酸可操作连接的3’UTR,所述3’UTR包含SEQ ID NO:12的核苷酸序列。In still other embodiments, the expression construct according to the present invention further comprises a 3'UTR operably linked to said ABCD1 encoding nucleic acid. The 3'UTR may comprise at least 1 copy (eg, 1-8 copies, such as 2, 3, or 4 copies) of the target sequence of a myocyte-specific miRNA and/or at least 1 copy (eg, 1-8 copies, eg, 1 or 2 copies) of the target sequence of the hepatocyte-specific miRNA. Preferably, the 3'UTR comprises at least 1 copy (such as 3 copies) of a myocyte-specific miRNA target sequence and at least 1 (such as 2 copies) of a hepatocyte-specific miRNA target sequence. The at least one myocyte-specific miRNA target sequence and the at least one hepatocyte-specific miRNA target sequence may be included in the 3'UTR in various arrangements, but preferably, the myocyte-specific miRNA target sequence is Hepatocyte-specific miRNA target sequences spaced apart. The miRNA target sequences can be connected directly, or can be separated by a few nucleotides, such as 1-5 nucleotides. The muscle cell-specific miRNA target sequence that can be used in the present invention can be selected from miRNA1 target sequence, miRNA206 target sequence and combinations thereof; for example, the miRNA1 target sequence shown in SEQ ID NO:6 and the miRNA1 target sequence shown in SEQ ID NO:7 miRNA206 target sequence. The hepatocyte-specific miRNA target sequence that can be used in the present invention can be a miRNA122 target sequence, such as the miRNA122 target sequence shown in SEQ ID NO:8. In a preferred embodiment, the 3'UTR comprises miRNA target sequences arranged as follows: miRNA1 target sequence-miRNA122 target sequence-miRNA1 target sequence-miRNA122 target sequence-miRNA206 target sequence; or miRNA206 target sequence-miRNA122 target sequence- miRNA1 target sequence - miRNA122 target sequence - miRNA1 target sequence. In some more preferred embodiments, the expression construct according to the present invention comprises a 3'UTR operably linked to the ABCD1 encoding nucleic acid, said 3'UTR comprising the nucleotide sequence of SEQ ID NO:12.
在再一方面,本发明提供了包含根据本发明的表达构建体的载体。在一些实施方案中,所述载体是质粒或病毒载体。在一些优选实施方案中,所述载体是腺相关病毒(AAV)载体。在再一方面,本发明也提供了包含根据本发明的表达构建体或载体的宿主细胞,优选哺乳动物细胞。In a further aspect, the invention provides a vector comprising an expression construct according to the invention. In some embodiments, the vector is a plasmid or viral vector. In some preferred embodiments, the vector is an adeno-associated viral (AAV) vector. In a further aspect, the invention also provides a host cell, preferably a mammalian cell, comprising an expression construct or vector according to the invention.
在再一方面,本发明提供了重组腺相关病毒(AAV)载体,其中所述重组AAV载体在其基因组中包含根据本发明的表达构建体。优选地,所述重组AAV载体在其基因组中包含:(a)5’和3’AAV反向末端重 复(ITR)序列,和(b)位于5’和3’ITR之间的表达构建体,其中所述表达构建体包含以转录方向彼此功能性连接的如下元件:启动子;编码人ABCD1的多核苷酸;至少一个miRNA靶序列;一个或多个终止子;和一个或多个polyA信号序列。优选地,polyA信号序列选自SV40晚期polyA序列、兔β-珠蛋白polyA序列、和牛生长激素polyA序列,更优选地牛生长激素polyA序列,例如SEQ ID NO:13的核苷酸序列。根据本发明的重组AAV病毒载体可以是ssAAV载体或scAAV载体。在一些实施方案中,根据本发明的重组AAV病毒载体包含野生型AAV2ITR序列,或在scAAV载体的情况下,所述ITR之一是野生型AAV2ITR序列,且所述ITR之另一是缺少功能性末端解链位点(trs)和任选地D序列的AA2ΔITR序列。根据本发明的重组AAV病毒载体可以包含来自任何AAV血清型的衣壳蛋白,例如AAV1,AAV2,AAV3,AAV4,AAV5,AAV6,AAV7,AAV8,AAV9血清型,尤其是AAV9血清型的衣壳蛋白。优选地,所述重组AAV载体是具有AAV2的ITR序列和AA9衣壳蛋白的AAV2/9载体。In a further aspect, the present invention provides a recombinant adeno-associated virus (AAV) vector, wherein said recombinant AAV vector comprises in its genome an expression construct according to the present invention. Preferably, the recombinant AAV vector comprises in its genome: (a) 5' and 3' AAV inverted terminal repeat (ITR) sequences, and (b) an expression construct located between the 5' and 3' ITR, Wherein said expression construct comprises the following elements functionally linked to each other in the direction of transcription: a promoter; a polynucleotide encoding human ABCD1; at least one miRNA target sequence; one or more terminators; and one or more polyA signal sequences . Preferably, the polyA signal sequence is selected from the group consisting of SV40 late polyA sequence, rabbit β-globin polyA sequence, and bovine growth hormone polyA sequence, more preferably bovine growth hormone polyA sequence, such as the nucleotide sequence of SEQ ID NO:13. The recombinant AAV viral vector according to the present invention may be a ssAAV vector or a scAAV vector. In some embodiments, a recombinant AAV viral vector according to the invention comprises a wild-type AAV2 ITR sequence, or in the case of scAAV vectors, one of said ITRs is a wild-type AAV2 ITR sequence and the other of said ITRs lacks a functional AA2ΔITR sequence of terminal melting sites (trs) and optionally D sequence. The recombinant AAV viral vector according to the present invention may comprise capsid proteins from any AAV serotype, such as AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9 serotypes, especially AAV9 serotypes . Preferably, the recombinant AAV vector is an AAV2/9 vector having AAV2 ITR sequence and AA9 capsid protein.
在再一方面,本发明也提供包含根据本发明的表达构建体、根据本发明的载体或根据本发明的重组AAV病毒载体和药物可接受载体的药物组合物。根据需要,所述药物组合物可以配制以适用于不同的给药途径,例如腹膜内、肌内、动脉内、静脉内给药、鞘内给药或脑室内给药。在一个优选的实施方案中,所述药物组合物为静脉注射制剂。在一个优选的实施方案中,所述制剂包含预防或治疗有效量的根据本发明的重组AAV病毒载体。In yet another aspect, the present invention also provides a pharmaceutical composition comprising an expression construct according to the present invention, a vector according to the present invention or a recombinant AAV viral vector according to the present invention and a pharmaceutically acceptable carrier. The pharmaceutical composition may be formulated for different routes of administration, such as intraperitoneal, intramuscular, intraarterial, intravenous, intrathecal or intracerebroventricular administration, as desired. In a preferred embodiment, the pharmaceutical composition is an intravenous injection formulation. In a preferred embodiment, the formulation comprises a prophylactically or therapeutically effective amount of a recombinant AAV viral vector according to the invention.
在再一方面,本发明也涉及根据本发明的表达构建体或根据本发明的载体或根据本发明的重组AAV病毒载体用于在哺乳动物细胞、细胞系或细胞群中表达ABCD1的用途,或在制备用于在哺乳动物细胞、细胞系或细胞群中表达ABCD1的药物中的应用。所述哺乳动物细胞可以是体外、离体或体内细胞。因此,再一方面,本发明也提供了一种在哺乳动物细胞中表达ABCD1的方法(例如体外、体内、或离体方法),所述方法包括用根据本发明的表达构建体或根据本发明的载体转染分离的哺乳动物细胞、细胞系或细胞群。在一些实施方案中,所述哺乳动物细胞是细胞系、或分离自ABCD1缺陷型哺乳动物受试者的细胞,例如成纤维细胞。可以通过在体外或离体细胞中测定目的蛋白表达和/或细胞生长增殖速度的试验,监测本发明构建体、载体或重组AAV病毒载体对细胞的目的蛋白表达和/或活力的影响。In a further aspect, the invention also relates to the use of an expression construct according to the invention or a vector according to the invention or a recombinant AAV viral vector according to the invention for expressing ABCD1 in a mammalian cell, cell line or cell population, or Use in the preparation of a medicament for expressing ABCD1 in mammalian cells, cell lines or cell groups. The mammalian cells may be in vitro, ex vivo or in vivo. Therefore, in another aspect, the present invention also provides a method (such as an in vitro, in vivo, or ex vivo method) of expressing ABCD1 in mammalian cells, said method comprising using an expression construct according to the present invention or a method according to the present invention vector to transfect isolated mammalian cells, cell lines or cell populations. In some embodiments, the mammalian cell is a cell line, or a cell, such as a fibroblast, isolated from an ABCD1-deficient mammalian subject. The impact of the construct, vector or recombinant AAV virus vector of the present invention on the expression and/or viability of the target protein of the cells can be monitored by testing the expression of the target protein and/or the growth rate of the cell in vitro or in isolated cells.
再一方面,本发明提供治疗或预防X染色体连锁肾上腺脑白质营养不良(X-ALD)和/或改善与X-ALD相关的症状的方法,其中所述方法包括向有需要的受试者施用根据本发明的重组AAV病毒载体或根据本发明的药物组合物。优选地,所述方法包括腹膜内、肌内、动脉内、静脉内、鞘内或脑室内给药,更优选地静脉内注射根据本发明的重组AAV病毒载体。优选地,所述重组AAV病毒载体是AAV9载体。在一些实施方案中,通过IV施用本发明重组AAV病毒载体,受试者的血浆和组织,尤其是肾上腺和小脑中,X-ALD疾病的生物化学标志物——极长链脂肪酸VLCFA(尤其是C26:0脂肪酸)——的含量降低。在一 些实施方案中,通过IV施用本发明重组AAV病毒载体,缓解/改善受试者脊髓中由VLCFA积累刺激的自噬活性。在一些实施方案中,本发明方法改善/缓解受试者CNS的进行性脱髓鞘症状。In yet another aspect, the present invention provides a method for treating or preventing X-chromosome-linked adrenoleukodystrophy (X-ALD) and/or improving symptoms associated with X-ALD, wherein the method comprises administering to a subject in need The recombinant AAV viral vector according to the present invention or the pharmaceutical composition according to the present invention. Preferably, the method comprises intraperitoneal, intramuscular, intraarterial, intravenous, intrathecal or intracerebroventricular administration, more preferably intravenous injection of a recombinant AAV viral vector according to the invention. Preferably, the recombinant AAV viral vector is an AAV9 vector. In some embodiments, by IV administration of the recombinant AAV virus vector of the present invention, the plasma and tissues of the subject, especially the adrenal gland and cerebellum, the biochemical markers of X-ALD disease—very long chain fatty acid VLCFA (especially C26:0 fatty acid)——The content is reduced. In some embodiments, autophagic activity stimulated by VLCFA accumulation in the subject's spinal cord is alleviated/improved by IV administration of a recombinant AAV viral vector of the invention. In some embodiments, the methods of the invention ameliorate/alleviate symptoms of progressive demyelination of the subject's CNS.
在再一方面,本发明也提供了包含与基因可操作性连接的3’UTR的表达构建体,其中所述3’UTR包含:In yet another aspect, the present invention also provides an expression construct comprising a 3'UTR operably linked to a gene, wherein said 3'UTR comprises:
-miRNA1 target x2(即,2个拷贝的miRNA1靶序列)-miRNA206 target x1(即,1个拷贝的miRNA206靶序列)-miRNA122 target x2(即,2个拷贝的miRNA122靶序列);例如SEQ ID NO:9-miRNA1 target x2 (ie, 2 copies of the miRNA1 target sequence) -miRNA206 target x1 (ie, 1 copy of the miRNA206 target sequence) -miRNA122 target x2 (ie, 2 copies of the miRNA122 target sequence); e.g. SEQ ID NO :9
-miRNA1 target x2(即,2个拷贝的miRNA1靶序列)-miRNA122 target x2(即,2个拷贝的miRNA122靶序列)-miRNA206 target x1(即,1个拷贝的miRNA206靶序列);例如,SEQ ID NO:10-miRNA1 target x2 (i.e., 2 copies of the miRNA1 target sequence) -miRNA122 target x2 (i.e., 2 copies of the miRNA122 target sequence) -miRNA206 target x1 (i.e., 1 copy of the miRNA206 target sequence); e.g., SEQ ID NO:10
-miRNA1 target x1(即,1个拷贝的miRNA1靶序列)-miRNA122 target x1(即,1个拷贝的miRNA122靶序列)-miRNA1 target x1(即,1个拷贝的miRNA1靶序列)-miRNA122 target x1(即,1个拷贝的miRNA122靶序列)-miRNA206 target x1(即,1个拷贝的miRNA206靶序列);例如SEQ ID NO:11。在一些实施方案中,根据本发明的表达构建体可以用于选择性地降低与之可操作性连接的目的基因在肌细胞和肝细胞中的表达,或用于制备选择性地降低目的基因在肌细胞和肝细胞中表达的药物中的用途。-miRNA1 target x1 (i.e., 1 copy of miRNA1 target sequence)-miRNA122 target x1 (i.e., 1 copy of miRNA122 target sequence)-miRNA1 target x1 (i.e., 1 copy of miRNA1 target sequence)-miRNA122 target x1 ( That is, 1 copy of the miRNA122 target sequence)-miRNA206 target x1 (ie, 1 copy of the miRNA206 target sequence); for example, SEQ ID NO: 11. In some embodiments, the expression construct according to the present invention can be used to selectively reduce the expression of a gene of interest operably linked thereto in muscle cells and liver cells, or to prepare a gene for selectively reducing the expression of a gene of interest in Use in drugs expressed in muscle cells and hepatocytes.
附图说明Description of drawings
图1显示,用于本发明的表达构建体示意图(图1A)和优化的ABCD1编码核酸的表达检测结果(图1B)。图1A中,ITR:AAV病毒的ITR序列;CA:CA启动子;CAR-Mut:具有突变的CAR启动子;hABCD1:天然人ABCD1编码核酸;coABCD1:优化的ABCD1编码核酸;EGFP:增强型绿色荧光蛋白;polyA:polyA信号序列;3’UTR:3’非翻译区。图1B左图显示,在HEK293细胞中瞬时表达天然人ABCD1编码核酸(hABCD1)和优化的ABCD1编码核酸(coABCD1),48小时后裂解细胞并提取细胞总蛋白,在10%SDS-PAGE上电泳分离蛋白条带(左图上方显示ABCD1蛋白条带,左图下方显示肌动蛋白条带);图1B右图显示,电泳分离的条带的荧光显色结果在经灰度扫描处理后的相对灰度值。Figure 1 shows the schematic diagram of the expression construct used in the present invention (Figure 1A) and the expression detection results of the optimized ABCD1 encoding nucleic acid (Figure 1B). In Fig. 1A, ITR: ITR sequence of AAV virus; CA: CA promoter; CAR-Mut: CAR promoter with mutation; hABCD1: natural human ABCD1 encoding nucleic acid; coABCD1: optimized ABCD1 encoding nucleic acid; EGFP: enhanced green Fluorescent protein; polyA: polyA signal sequence; 3'UTR: 3' untranslated region. The left panel of Figure 1B shows that the natural human ABCD1-encoding nucleic acid (hABCD1) and the optimized ABCD1-encoding nucleic acid (coABCD1) were transiently expressed in HEK293 cells, the cells were lysed after 48 hours, and the total cellular protein was extracted and separated by electrophoresis on 10% SDS-PAGE Protein bands (the upper left figure shows the ABCD1 protein band, and the lower left figure shows the actin band); the right figure in Figure 1B shows that the fluorescent color results of the bands separated by electrophoresis are relatively gray after grayscale scanning degree value.
图2显示,pscAAV-CAR-Gluc质粒载体示意图。Figure 2 shows a schematic diagram of the pscAAV-CAR-Gluc plasmid vector.
图3显示,在基于体外培养细胞的试验中,与未转染质粒的BHK-21细胞(即,空白对照)相比,在转染了pscAAV-CAR-Gluc载体以及pscAAV-CAR-MutC-Gluc载体、pscAAV-CAR-MutA-Gluc载体和pscAAV-CAR-MutG-Gluc载体的BHK-21细胞中,测定的Gluc水平变化。其中,**表示p<0.01。Figure 3 shows that in an assay based on cultured cells in vitro, compared with BHK-21 cells that were not transfected with the plasmid (i.e., blank control), the pscAAV-CAR-Gluc vector and the pscAAV-CAR-MutC-Gluc vector were transfected. Changes in Gluc levels measured in BHK-21 cells with vector, pscAAV-CAR-MutA-Gluc vector and pscAAV-CAR-MutG-Gluc vector. Among them, ** means p<0.01.
图4显示,在基于EGFP的表达质粒载体中,检测嵌入miRNA1、122、206靶序列对于目的基因在肌细胞和肝细胞中表达的影响。Figure 4 shows that in the EGFP-based expression plasmid vector, the impact of the target sequence embedded in miRNA1, 122, and 206 on the expression of the target gene in muscle cells and liver cells was detected.
图5显示,在基于Gluc报告基因的表达质粒载体中,检测增加miRNA靶序列的重复次数和改变miRNA 靶序列的排列组合方式,对目的基因在肌细胞中表达的影响。Figure 5 shows that in the expression plasmid vector based on the Gluc reporter gene, the effect of increasing the number of repetitions of the miRNA target sequence and changing the arrangement and combination of the miRNA target sequence on the expression of the target gene in muscle cells was detected.
图6显示,在基于Gluc报告基因的表达质粒载体中,检测增加miRNA靶序列的重复次数和改变miRNA靶序列的排列组合方式,对目的基因在肝细胞中表达的影响。Figure 6 shows that in the Gluc reporter gene-based expression plasmid vector, the effect of increasing the number of repeats of the miRNA target sequence and changing the arrangement and combination of the miRNA target sequence on the expression of the target gene in hepatocytes was detected.
图7显示,用于构建AAV质粒载体的基础质粒pRDAAV-CMV-EGFP的示意图。Figure 7 shows a schematic diagram of the basic plasmid pRDAAV-CMV-EGFP used to construct the AAV plasmid vector.
图8显示,采用来自X-ALD患者1和患者2的成纤维细胞,在细胞爬片免疫荧光检测试验中,测定重组AAV9-coABCD1病毒转导对细胞ABCD1表达的影响。红色荧光显示,被dylight549标记二抗染色的胞浆中ABCD1蛋白;蓝色荧光显示,dapi细胞核染色。Figure 8 shows that using fibroblasts from X-ALD patient 1 and patient 2, the impact of recombinant AAV9-coABCD1 virus transduction on the expression of ABCD1 in cells was determined in the cell slide immunofluorescence assay. The red fluorescence shows the ABCD1 protein in the cytoplasm stained by the dylight549-labeled secondary antibody; the blue fluorescence shows the nuclei stained by dapi.
图9显示,重组AAV9-coABCD1病毒转导对细胞生长增殖速度的影响。图9A和9B分别显示,未接种AAV9-coABCD1和接种了AAV9-coABCD1的患者2成纤维细胞在培养7天的细胞密度。图9C显示,正常人的成纤维细胞在培养7天的细胞密度。Figure 9 shows the effect of recombinant AAV9-coABCD1 virus transduction on cell growth and proliferation rate. Figures 9A and 9B show the cell densities of patient 2 fibroblasts not inoculated with AAV9-coABCD1 and inoculated with AAV9-coABCD1 at 7 days in culture, respectively. Figure 9C shows the cell density of normal human fibroblasts in culture for 7 days.
图10显示,相对于未给药的C57BL6J野生型小鼠(WT),注射AA9-coABCD1或AAV9-coABCD1-miT重组病毒后,在多种组织中的极长链脂肪酸含量改变情况。其中,*表示p<0.05。Figure 10 shows the changes in the content of very long chain fatty acids in various tissues after injection of AA9-coABCD1 or AAV9-coABCD1-miT recombinant virus compared to untreated C57BL6J wild-type mice (WT). Among them, * means p<0.05.
图11显示,注射AA9-coABCD1或AAV9-coABCD1-miT重组病毒后,供试动物的体内组织病理学改变情况。图11A显示,接种优化后的重组病毒rAAV9-coABCD1-miT,显著地减少了供试小鼠的心脏和肝脏的病理改变。黑箭头指示,心脏空泡样变性和肝脏的核固缩、灶性坏死。图11B显示,在接种rAAV9-coABCD1的供试动物组中出现的一例死亡小鼠的尸检结果,上图中心脏组织的组织病理学检测结果,黑箭头指示出现了心肌大面积空泡样变性;红箭头指示出现了心腔内大面积的血栓形成;下图显示肝脏组织的组织病理学检测结果,显示出广泛的肝细胞坏死和核固缩。Figure 11 shows the histopathological changes in the test animals after injection of AA9-coABCD1 or AAV9-coABCD1-miT recombinant virus. Figure 11A shows that inoculation with the optimized recombinant virus rAAV9-coABCD1-miT significantly reduced the pathological changes in the hearts and livers of the tested mice. Black arrows indicate vacuolar degeneration of the heart and nuclear pyknosis and focal necrosis of the liver. Figure 11B shows the autopsy results of a dead mouse in the test animal group inoculated with rAAV9-coABCD1, the histopathological examination results of the heart tissue in the above figure, the black arrow indicates the occurrence of large-area vacuolar degeneration of the myocardium; Red arrows indicate extensive intracardiac thrombus formation; lower panel shows histopathological examination of liver tissue, showing extensive hepatocyte necrosis and nuclear pyknosis.
图12显示,在抓绳实验中,评估施用AAV9-coABCD1-miT对于X-ALD模型小鼠行为的影响。结果显示,AAV9-coABCD1-miT的静脉内施用,有效改善了模型小鼠的运动能力。FIG. 12 shows the evaluation of the effect of administering AAV9-coABCD1-miT on the behavior of X-ALD model mice in the rope grabbing experiment. The results showed that intravenous administration of AAV9-coABCD1-miT effectively improved the exercise capacity of model mice.
图13显示,将优化后药物AAV9-coABCD1-miT施用给X-ALD模型小鼠后8周,检测各组织中的极长链脂肪酸含量,并与未给药的X-ALD模型小鼠和野生型正常小鼠进行比较。其中,*表示p<0.05;NS表示差异不显著。Figure 13 shows that 8 weeks after the optimized drug AAV9-coABCD1-miT was administered to X-ALD model mice, the content of very long chain fatty acids in each tissue was detected, and compared with unadministered X-ALD model mice and wild animals. normal mice for comparison. Among them, * indicates p<0.05; NS indicates that the difference is not significant.
图14显示,AAV9-coABCD1-miT尾静脉注射治疗X-ALD模型小鼠,给药后8周,肾上腺组织病理学检测结果。红色荧光显示,免疫荧光标记的细胞中ABCD1蛋白;蓝色荧光显示,细胞核染色。Figure 14 shows the histopathological results of the adrenal glands of X-ALD model mice treated with AAV9-coABCD1-miT tail vein injection 8 weeks after administration. The red fluorescence shows the ABCD1 protein in the immunofluorescence-labeled cells; the blue fluorescence shows that the nuclei are stained.
图15显示,在AAV9-coABCD1-miT给药后,在脊髓组织切片上免疫荧光标记LC3β,以评估X-ALD模型小鼠神经系统的自噬和改善情况。Figure 15 shows that after AAV9-coABCD1-miT administration, LC3β was immunofluorescently labeled on spinal cord tissue sections to evaluate the autophagy and improvement in the nervous system of X-ALD model mice.
图16显示,将优化后(AAV9-coABCD1-miRT)与未优化的药物(AAV9-coABCD1)静脉注射进正常野生型小鼠后(每组2只小鼠),利用Western Blot法测定心脏组织中的ABCD1蛋白含量。图16A显示, Western Blot检测结果,其中,上图显示ABCD1条带,下图显示β-肌动蛋白条带;其中,泳道1和2为AAV9-coABCD1给药的小鼠(RD48-1和RD48-2);泳道3-4为AAV9-coABCD1-miRT给药的小鼠(RD49-2和RD49-4);泳道5和6为未给药的正常小鼠对照(N1和N2)。图16B显示,利用软件ImageJ将Western Blot检测结果图片进行灰度值分析,得到的量化结果。Figure 16 shows that after intravenous injection of optimized (AAV9-coABCD1-miRT) and unoptimized drugs (AAV9-coABCD1) into normal wild-type mice (2 mice in each group), the Western Blot method was used to determine the ABCD1 protein content. Figure 16A shows the Western Blot detection results, wherein the upper figure shows the ABCD1 band, and the lower figure shows the β-actin band; wherein, lanes 1 and 2 are mice administered with AAV9-coABCD1 (RD48-1 and RD48 -2); lanes 3-4 are mice administered with AAV9-coABCD1-miRT (RD49-2 and RD49-4); lanes 5 and 6 are normal mouse controls without administration (N1 and N2). Figure 16B shows the quantification results obtained by analyzing the gray value of the Western Blot detection result pictures using the software ImageJ.
发明详述Detailed description of the invention
本发明公开了用于治疗X-ALD的基因治疗构建体、药物组合物和方法,尤其是用于递送ABCD1的重组AAV载体的构建、制备及应用。The invention discloses a gene therapy construct, a pharmaceutical composition and a method for treating X-ALD, especially the construction, preparation and application of a recombinant AAV vector for delivering ABCD1.
除非下文中另外定义,否则本说明书中所用的全部技术与科学术语具有如本发明所属领域的普通技术人员通常理解的相同含义。本文所提及的全部出版物、专利申请、专利和其他参考文献通过引用的方式完整地并入。此外,本文中所述的材料、方法和例子仅是说明性的并且不意在是限制性的。本发明的其他特征、目的和优点将从本说明书及附图并且从后附的权利要求书中显而易见。Unless defined otherwise hereinafter, all technical and scientific terms used in this specification have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In addition, the materials, methods, and examples described herein are illustrative only and not intended to be limiting. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the appended claims.
定义definition
术语“约”在与数字数值联合使用时意为涵盖具有比指定数字数值小5%的下限和比指定数字数值大5%的上限的范围内的数字数值。该术语也旨在涵盖在指定数字±1%,±0.5%,或±0.1%范围内的数值。The term "about" when used in conjunction with a numerical value is meant to encompass a numerical value within a range having a lower limit of 5% less and an upper limit of 5% greater than the stated numerical value. The term is also intended to cover values within ±1%, ±0.5%, or ±0.1% of the specified figure.
在本文中,术语“包含”或“包括”意指包括所述的要素、整数或步骤,但是不排除任意其他要素、整数或步骤。As used herein, the term "comprising" or "comprising" means including stated elements, integers or steps, but not excluding any other elements, integers or steps.
在本文中,表述“和/或”,用于连接多个项目时,表示所列相关项目中的任何一个、或所列相关项目中的多个或全部的任何和所有可能的组合。Herein, the expression "and/or", when used to connect multiple items, means any one of the listed related items, or any and all possible combinations of a plurality or all of the listed related items.
在本文中,重组腺相关病毒可以用衣壳的来源AAV病毒血清型单独来表示,或用衣壳和基因组ITR序列的来源AAV病毒血清型来表示。在后一情况下,在本文中,采用标识符“/”进行分隔,标识符“/”之后为衣壳的来源血清型,标识符“/”之前为ITR的来源血清型。因此,例如,表述重组AAV9中的数字9表示所述重组腺相关病毒具有来自AAV9血清型的衣壳;而表述重组AAV2/9中标识符“/”前的数字表示所述重组腺相关病毒具有来自AAV2的野生型或变体ITR序列,而标识符“/”后的数字表示所述重组腺相关病毒具有来自AAV9的衣壳蛋白。Herein, recombinant adeno-associated virus can be represented by the AAV virus serotype from which the capsid is derived alone, or by the AAV virus serotype from which the capsid and genomic ITR sequences are derived. In the latter case, in this paper, the identifier "/" is used for separation, followed by the serotype of origin of the capsid and before the identifier "/" by the serotype of origin of the ITR. Thus, for example, the number 9 in the expression recombinant AAV9 indicates that the recombinant adeno-associated virus has a capsid from the AAV9 serotype; while the number before the identifier "/" in the expression recombinant AAV2/9 indicates that the recombinant adeno-associated virus has The wild-type or variant ITR sequence from AAV2, while the number after the identifier "/" indicates that the recombinant adeno-associated virus has a capsid protein from AAV9.
术语“功能性连接”也称作“有效连接”或“可操作连接”,意指指定的各组分处于一种允许它们以预期的方式起作用的关系中。The term "functionally linked", also known as "operably linked" or "operably linked", means that the named components are in a relationship allowing them to function in their intended manner.
术语序列“同一性”用于描述两个氨基酸序列或多核苷酸序列之间的序列结构相似性。为确定两个氨基酸序列或两个核酸序列的同一性百分数,可以将所述序列出于最佳比较目的进行比对(例如,可以为了最 佳比对而在第一和第二氨基酸序列或第一和第二核酸序列之一或二者中引入空位或可以为比较目的而抛弃非同源序列)。在一个优选实施方案中,为比较目的,所比对的参考序列的长度是参考序列全长的至少30%、优选地至少40%、更优选地至少50%、60%和甚至更优选地至少70%、80%、90%、100%。随后比较在对应氨基酸位置或核苷酸位置处的氨基酸残基或核苷酸。当第一序列中的位置由第二序列中对应位置处的相同氨基酸残基或核苷酸占据时,则所述第一序列和第二序列在此位置处是相同的。The term sequence "identity" is used to describe the similarity in sequence structure between two amino acid sequences or polynucleotide sequences. To determine the percent identity of two amino acid sequences or two nucleic acid sequences, the sequences can be aligned for optimal comparison purposes (e.g., the first and second amino acid sequences or the first and second amino acid sequences can be aligned for optimal alignment). Gaps may be introduced into one or both of the first and second nucleic acid sequences or non-homologous sequences may be discarded for comparison purposes). In a preferred embodiment, for comparison purposes, the length of the reference sequence being aligned is at least 30%, preferably at least 40%, more preferably at least 50%, 60% and even more preferably at least 70%, 80%, 90%, 100%. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide at the corresponding position in the second sequence, then the first and second sequences are identical at that position.
可以利用数学算法实现两个序列间的序列比较和同一性百分数的计算。在一个优选实施方案中,使用已经集成至GCG软件包的GAP程序中的Needlema和Wunsch((1970)J.Mol.Biol.48:444-453)算法(在http://www.gcg.com可获得),使用Blossum 62矩阵或PAM250矩阵和空位权重16、14、12、10、8、6或4和长度权重1、2、3、4、5或6,确定两个氨基酸序列之间的同一性百分数。在又一个优选的实施方案中,使用GCG软件包中的GAP程序(在http://www.gcg.com可获得),使用NWSgapdna.CMP矩阵和空位权重40、50、60、70或80和长度权重1、2、3、4、5或6,确定两个核苷酸序列之间的同一性百分数。特别优选的参数集合(和除非另外说明否则应当使用的一个参数集合)是采用空位罚分12、空位延伸罚分4和移码空位罚分5的Blossum 62评分矩阵。The comparison of sequences and the calculation of percent identity between two sequences can be accomplished using a mathematical algorithm. In a preferred embodiment, the Needlema and Wunsch ((1970) J. Mol. Biol. 48:444-453) algorithm (available at http://www.gcg.com available), use the Blossum 62 matrix or the PAM250 matrix with gap weights of 16, 14, 12, 10, 8, 6 or 4 and length weights of 1, 2, 3, 4, 5 or 6 to determine the distance between two amino acid sequences. percent identity. In yet another preferred embodiment, using the GAP program in the GCG software package (available at http://www.gcg.com), using the NWSgapdna.CMP matrix and gap weights of 40, 50, 60, 70 or 80 and Length weights of 1, 2, 3, 4, 5 or 6 determine the percent identity between two nucleotide sequences. A particularly preferred parameter set (and one that should be used unless otherwise stated) is the Blossum 62 scoring matrix with a gap penalty of 12, a gap extension penalty of 4, and a frameshift gap penalty of 5.
还可以使用PAM120加权余数表、空位长度罚分12,空位罚分4,利用已经并入ALIGN程序(2.0版)的E.Meyers和W.Miller算法,((1989)CABIOS,4:11-17),确定两个氨基酸序列或核苷酸序列之间的同一性百分数。It is also possible to use a PAM120 weighted remainder table, a gap length penalty of 12, and a gap penalty of 4, using the E. Meyers and W. Miller algorithm that has been incorporated into the ALIGN program (version 2.0), ((1989) CABIOS, 4:11-17 ), to determine the percent identity between two amino acid sequences or nucleotide sequences.
术语“宿主细胞”指已经向其中引入外源多核苷酸的细胞,包括这类细胞的子代。在一些实施方案中,宿主细胞是可以用来产生本发明重组AAV载体的任何类型的细胞,例如,哺乳动物细胞(例如适用于通过三质粒包装系统生产重组AAV的HEK 293细胞)和昆虫细胞(例如适用于通过杆状病毒包装系统生产重组AAV的sf9细胞)。The term "host cell" refers to a cell into which an exogenous polynucleotide has been introduced, including the progeny of such cells. In some embodiments, the host cell is any type of cell that can be used to produce a recombinant AAV vector of the invention, for example, mammalian cells (such as HEK 293 cells suitable for production of recombinant AAV by a three-plasmid packaging system) and insect cells ( For example sf9 cells suitable for the production of recombinant AAV by the baculovirus packaging system).
术语“调控序列”或“表达控制序列”是指这样的核酸序列,其诱导、抑制或以其它方式控制与之有效连接的编码核酸序列的蛋白质转录。调控序列可以是例如起始序列、增强子序列、内含子序列和启动子序列等。The term "regulatory sequence" or "expression control sequence" refers to a nucleic acid sequence that induces, represses, or otherwise controls the transcription of a protein of an encoding nucleic acid sequence to which it is operably linked. Regulatory sequences can be, for example, initiation sequences, enhancer sequences, intron sequences, and promoter sequences, among others.
描述核酸或蛋白质时所用的术语“外源的”或“异源的”可互换使用,是指核酸或蛋白质并非天然存在于其所在的该染色体或宿主细胞的位置。外源核酸序列也指衍生自并插入相同宿主细胞或受试者但以非天然状态存在的序列,例如,所述序列以不同的拷贝数存在,或处于不同调控元件的控制下。The terms "exogenous" or "heterologous" are used interchangeably when describing a nucleic acid or protein to mean that the nucleic acid or protein does not naturally exist in the chromosomal or host cell location in which it is found. An exogenous nucleic acid sequence also refers to a sequence that is derived from and inserted into the same host cell or subject but exists in a non-native state, eg, the sequence is present in a different copy number, or is under the control of a different regulatory element.
在本文中,“分离的”多核苷酸(例如,分离的DNA或分离的RNA)是指,多核苷酸至少部分地从包含其的天然生物体或病毒的至少一些其他组分中分离出来。在一些实施方案中,“分离的”核酸相对于起始材料被富集了至少大约10倍、100倍、1000倍、10,000倍或更多。As used herein, an "isolated" polynucleotide (eg, isolated DNA or isolated RNA) means that the polynucleotide is at least partially separated from at least some other components of the native organism or virus in which it is comprised. In some embodiments, an "isolated" nucleic acid is enriched at least about 10-fold, 100-fold, 1000-fold, 10,000-fold or more relative to the starting material.
在本文中,“分离的”多肽是指多肽至少部分地从包含其的天然生物体或病毒的至少一些其他组分中分离出来。在一些实施方案中,“分离的”多肽相对于起始材料被富集了至少大约10倍、100倍、1000倍、10,000倍或更多。As used herein, an "isolated" polypeptide refers to a polypeptide that is at least partially separated from at least some other components of the native organism or virus in which it is contained. In some embodiments, an "isolated" polypeptide is enriched at least about 10-fold, 100-fold, 1000-fold, 10,000-fold or more relative to the starting material.
在本文中,“分离”或“纯化”的病毒载体是指,病毒载体从包含其的起始材料的至少一些组分中被部分地分离出来。在一些实施方案中,“分离的”病毒载体相对于起始材料被富集了至少大约10倍、100倍、1000倍、10,000倍或更多。As used herein, an "isolated" or "purified" viral vector means that the viral vector has been partially separated from at least some components of the starting material comprising it. In some embodiments, an "isolated" viral vector is enriched at least about 10-fold, 100-fold, 1000-fold, 10,000-fold or more relative to the starting material.
在本文中,术语“病毒载体”是指,能够作为目的核酸的运载工具的病毒颗粒(例如AAV病毒颗粒)。通常,病毒载体包含衣壳和包装在其中的病毒基因组(例如,病毒DNA),待递送的目的核酸插在病毒基因组中。在重组AAV病毒载体的情况下,为了产生可以将目的核酸递送至组织或细胞的重组病毒颗粒,通常仅需要在基因组中保留反向末端重复(ITR)顺式元件,而病毒包装所需的其余序列可以反式提供。因此,在一些实施方案中,本发明的重组AAV病毒载体包含衣壳和包装在其中的重组病毒基因组,其中所述重组病毒基因组包含或由位于两个AAV ITR序列之间的一个或多个外源核苷酸序列组成。位于重组病毒基因组5’和3’末端的两个ITR序列(即,5’ITR和3’ITR)可以相同或不同。As used herein, the term "viral vector" refers to a viral particle (such as an AAV viral particle) capable of serving as a delivery vehicle for a nucleic acid of interest. Generally, a viral vector comprises a capsid and a viral genome (for example, viral DNA) packaged therein, and the target nucleic acid to be delivered is inserted into the viral genome. In the case of recombinant AAV viral vectors, in order to generate recombinant virus particles that can deliver the nucleic acid of interest to tissues or cells, it is usually only necessary to retain the inverted terminal repeat (ITR) cis element in the genome, while the rest required for viral packaging Sequences can be provided in trans. Accordingly, in some embodiments, the recombinant AAV viral vectors of the present invention comprise a capsid and a recombinant viral genome packaged therein, wherein the recombinant viral genome comprises or consists of one or more exogenous genes located between two AAV ITR sequences. Source nucleotide sequence composition. The two ITR sequences located at the 5' and 3' ends of the recombinant viral genome (i.e., 5'ITR and 3'ITR) may be the same or different.
术语AAV“反向末端重复”(inverted terminal repeat,ITR)在本文中是指,来自AAV病毒基因组的顺式作用元件,在AAV病毒的整合、拯救、复制和基因组包装中发挥重要作用。天然AAV病毒的ITR序列中包含Rep蛋白结合位点(Rep binding site,RBS)和末端解链位点trs(terminal resolution site),能够被Rep蛋白结合识别并在trs处产生切口。该ITR序列还可形成独特的“T”字母型二级结构,在AAV病毒的生活周期中发挥重要作用。最早分离到的AAV病毒,AAV2,具有位于基因组两端、长度145bp的呈回文-发卡结构的“反向末端重复序列”(ITR)。之后,在各种血清型的AAV病毒中发现不尽相同的ITR序列,但是都能形成发卡结构和存在Rep结合位点。基于这些野生型ITR序列的传统重组AAV病毒载体一般为单链AAV载体(ssAAV),病毒基因组以单链形式包装在AAV衣壳中。与此类ssAAV不同,已经发现,通过改造ITR,删除AAV病毒的一侧ITR序列中的trs序列和任选地D序列,能够使包装得到的重组AAV病毒载体所携带的基因组自我互补,形成双链(Wang Z等人,Gene Ther.2003;10(26):2105-2111;McCarty DM等人,Gene Ther.2003;10(26):2112-2118)。由此包装得到的病毒为双链AAV病毒,即,scAAV(self-complementary AAV)病毒。scAAV病毒载体的包装容量更小,仅为ssAAV病毒载体包装容量的一半,约为2.2kb-2.5kb,但感染细胞后转导效率更高。The term AAV "inverted terminal repeat" (inverted terminal repeat, ITR) refers herein to a cis-acting element from the AAV viral genome that plays an important role in the integration, rescue, replication, and genome packaging of the AAV virus. The ITR sequence of the natural AAV virus contains a Rep protein binding site (Rep binding site, RBS) and a terminal unzipping site trs (terminal resolution site), which can be recognized by the Rep protein and generate a nick at the trs. The ITR sequence can also form a unique "T" letter-shaped secondary structure, which plays an important role in the life cycle of the AAV virus. The earliest isolated AAV virus, AAV2, has "inverted terminal repeats" (ITRs) with a palindrome-hairpin structure of 145 bp located at both ends of the genome. Later, different ITR sequences were found in various serotypes of AAV viruses, but they all formed hairpin structures and had Rep binding sites. Traditional recombinant AAV viral vectors based on these wild-type ITR sequences are generally single-stranded AAV vectors (ssAAV), and the viral genome is packaged in the AAV capsid in a single-stranded form. Different from this type of ssAAV, it has been found that by modifying the ITR and deleting the trs sequence and optionally the D sequence in the ITR sequence on one side of the AAV virus, the genome carried by the recombinant AAV virus vector obtained by packaging can be self-complementary to form a double chain (Wang Z et al., Gene Ther. 2003; 10(26):2105-2111; McCarty DM et al., Gene Ther. 2003; 10(26):2112-2118). The virus thus packaged is a double-stranded AAV virus, that is, scAAV (self-complementary AAV) virus. The packaging capacity of the scAAV viral vector is smaller, only half of the packaging capacity of the ssAAV viral vector, about 2.2kb-2.5kb, but the transduction efficiency after infection of cells is higher.
在本文中,与AAV相关的该术语ITR涵盖野生型ITR和变体ITR。野生型ITR可以来自任何天然AAV病毒,例如AAV2病毒。野生型ITR中包含Rep蛋白结合位点(Rep binding site,RBS)和末端解链位点trs(terminal resolution site),能够被Rep蛋白结合识别并在trs处产生切口。野生型ITR序列可形成独特 的“T”字母型二级结构,在AAV病毒的生活周期中发挥重要作用。在本文中,变体ITR是非天然的ITR序列,其可以例如来自任何野生型AAV ITR序列,并相对于野生型ITR包含一个或多个核苷酸的缺失、替代、和/或添加,和/或截短,但仍具有功能性,即,能够用于产生ssAAV病毒载体或scAAV病毒载体。在一些实施方案中,变体ITR是缺失了功能性trs位点和任选地D区序列的AAV ITR序列(在本文中,也称作ΔITR)。在一些实施方案中,野生型ITR与ΔITR组合用于产生自我互补型重组AAV病毒载体(scAAV)。在另一些实施方案中,两个野生型ITR组合用于产生单链重组AAV病毒载体(ssAAV)。As used herein, the term ITR in relation to AAV encompasses wild-type ITRs and variant ITRs. Wild-type ITRs can be from any native AAV virus, such as an AAV2 virus. The wild-type ITR contains a Rep protein binding site (Rep binding site, RBS) and a terminal unzipping site trs (terminal resolution site), which can be recognized by the Rep protein and generate a nick at trs. The wild-type ITR sequence can form a unique "T" letter-shaped secondary structure, which plays an important role in the life cycle of AAV virus. As used herein, a variant ITR is a non-native ITR sequence which may, for example, be derived from any wild-type AAV ITR sequence and which comprises a deletion, substitution, and/or addition of one or more nucleotides relative to the wild-type ITR, and/ Or truncated, but still functional, ie, can be used to generate ssAAV viral vectors or scAAV viral vectors. In some embodiments, a variant ITR is an AAV ITR sequence (also referred to herein as a ΔITR) that has been deleted for a functional trs site and optionally a D region sequence. In some embodiments, wild-type ITRs are combined with ΔITRs to generate self-complementary recombinant AAV viral vectors (scAAV). In other embodiments, two wild-type ITRs are used in combination to generate single-chain recombinant AAV viral vectors (ssAAV).
AAV蛋白VP1,VP2和VP3是衣壳蛋白,其相互作用以形成AAV衣壳。不同血清型的AAV病毒具有不同的组织感染嗜性,可以通过选择重组AAV病毒载体衣壳的来源血清型,将外源基因转运至特定的器官和组织(Wu Z等人,Mol Ther.2006;14(3):316-327)。在本发明中,重组AAV病毒载体可以通过选择衣壳的来源血清型,而具有不同的靶向性。在一些实施方案中,重组AAV病毒的衣壳来自对神经元细胞具有靶向性的AAV血清型。在一个实施方案中,重组AAV病毒载体包含来自AAV9的衣壳。在再一个实施方案中,重组AAV病毒载体包含来自AAV9的衣壳和来自AAV2的ITR。The AAV proteins VP1, VP2 and VP3 are capsid proteins that interact to form the AAV capsid. Different serotypes of AAV viruses have different tissue infection tropisms, and foreign genes can be transferred to specific organs and tissues by selecting the source serotype of the recombinant AAV virus vector capsid (Wu Z et al., Mol Ther.2006; 14(3):316-327). In the present invention, the recombinant AAV virus vector can have different targeting properties by selecting the source serotype of the capsid. In some embodiments, the capsid of the recombinant AAV virus is from an AAV serotype that targets neuronal cells. In one embodiment, the recombinant AAV viral vector comprises a capsid from AAV9. In yet another embodiment, the recombinant AAV viral vector comprises a capsid from AAV9 and an ITR from AAV2.
术语“治疗”指意欲改变正在接受治疗的个体中的疾病天然过程的医学介入。期望的治疗效果包括但不限于防止疾病出现或复发、减轻症状、减小疾病的任何直接或间接病理学后果、防止转移、降低病情进展速率、改善或缓和疾病状态,以及缓解或改善预后。在一些实施方案中,本发明的重组AAV病毒在施用给ABCD1缺陷性受试者或X-ALD患者后,优选在全身施用后,降低受试者的多个受累组织(尤其是,肾上腺和中枢神经系统)中的VLCFA含量。在一些实施方案中,本发明的重组AAV病毒在施用给ABCD1缺陷性受试者或X-ALD患者后,优选在全身施用后,改善受试者的中枢神经系统损伤和/或肾上腺损伤。在一些实施方案中,本发明的重组AAV病毒在施用给ABCD1缺陷性受试者或X-ALD患者后,优选在全身施用后,改善受试者的运动能力。The term "treatment" refers to medical intervention intended to alter the natural course of a disease in the individual being treated. Desired therapeutic effects include, but are not limited to, preventing the onset or recurrence of the disease, alleviating symptoms, reducing any direct or indirect pathological consequences of the disease, preventing metastasis, reducing the rate of disease progression, ameliorating or palliation of the disease state, and remission or improved prognosis. In some embodiments, the recombinant AAV virus of the present invention, after administration to an ABCD1-deficient subject or an X-ALD patient, preferably after systemic administration, reduces the number of affected tissues (especially, the adrenal gland and central nervous system) of the subject. Nervous system) VLCFA content. In some embodiments, the recombinant AAV virus of the present invention improves central nervous system damage and/or adrenal damage in a subject after administration to an ABCD1-deficient subject or an X-ALD patient, preferably after systemic administration. In some embodiments, the recombinant AAV virus of the invention improves the exercise capacity of the subject after administration to an ABCD1-deficient subject or an X-ALD patient, preferably after systemic administration.
在本文中,“预防”包括对疾病或特定疾病的症状的发生或发展的抑制。在一些实施方式中,具有X-ALD病发生倾向的受试者是预防性方案的候选者。通常,术语“预防”是指在疾病的至少一个症状发生前实施的医学干预。因此,在一个实施方案中,预防包括在具有ABCD1基因缺陷的受试者中于X-ALD病的症状发生前施用本发明的基因治疗药物,以延缓疾病发展或阻止疾病的出现。在一个实施方案中,预防包括通过使用本发明的基因治疗药物,改善神经系统中因VLCFA刺激的异常吞噬作用,从而防止与之相关的脊髓轴索病变发生。As used herein, "prevention" includes the inhibition of the occurrence or development of a disease or symptoms of a particular disease. In some embodiments, subjects predisposed to developing X-ALD disease are candidates for prophylactic regimens. In general, the term "prevention" refers to medical intervention performed before at least one symptom of a disease occurs. Therefore, in one embodiment, prevention includes administering the gene therapy drug of the present invention in subjects with ABCD1 gene deficiency before the symptoms of X-ALD disease occur, so as to delay the development of the disease or prevent the appearance of the disease. In one embodiment, the prevention includes using the gene therapy drug of the present invention to improve the abnormal phagocytosis stimulated by VLCFA in the nervous system, thereby preventing the occurrence of related spinal cord axonal lesions.
以下对本发明的各个方面进行描述。Various aspects of the invention are described below.
I.用于基因治疗的构建体I. Constructs for Gene Therapy
影响转基因在期望的靶细胞中表达的因素有多种,包括,启动子、基因编码核酸、5’UTR、3’UTR、以及转染的靶细胞中可获得的转录因子或调节因子(如细胞表达的miRNA)的类型、数量和活性。这些因素相互影响。因此,为了构建有效实现目的的基因构建体,通常需要针对性根据疾病特征,例如受累靶器官和组织细胞,综合考察基因构建体的元件组合。为此,本发明人通过对构建体的多个组成元件进行优化和组合,在深入研究的基础上,建立了有效且安全的X-ALD治疗用基因构建体。根据本发明的基因构建体具有如下至少一项或多项的优化基因元件:(1)优化的ABCD1编码核酸(也简称为coABCD1);(2)优化的组成型启动子;和(3)优化的miRNA靶序列组合。A variety of factors can affect the expression of a transgene in a desired target cell, including the promoter, gene-encoding nucleic acid, 5'UTR, 3'UTR, and transcription factors or regulators available in the transfected target cell (e.g., cell The type, quantity and activity of expressed miRNA). These factors influence each other. Therefore, in order to construct a gene construct that effectively achieves the purpose, it is usually necessary to comprehensively examine the combination of elements of the gene construct according to the characteristics of the disease, such as affected target organs and tissue cells. Therefore, the present inventors have established an effective and safe gene construct for X-ALD treatment by optimizing and combining multiple components of the construct and based on in-depth research. The gene construct according to the present invention has at least one or more optimized gene elements as follows: (1) optimized ABCD1 encoding nucleic acid (also referred to as coABCD1 for short); (2) optimized constitutive promoter; and (3) optimized combination of miRNA target sequences.
ABCD1编码核酸ABCD1 encoding nucleic acid
包含在本发明构建体中的ABCD1编码核酸可以是任何能够编码功能性ABCD1蛋白活性的多核苷酸。但为了有利于在哺乳动物细胞中的表达,有利的是,对编码ABCD1多肽的核酸,进行核酸序列优化。在一个实施方案中,用于本发明表达构建体中的ABCD1编码核酸包含SEQ ID NO:1的多核苷酸序列,或与之具有至少大约95%、大约96%,大约97%、98%、99%或更高核苷酸序列同一性的多核苷酸序列。有利的是,经优化的ABCD1编码核酸,相对于参比核酸(例如,天然人ABCD1编码核酸,例如具有SEQ ID NO:15所示的核苷酸序列的核酸),在可操作性连接的组成型启动子控制下,在宿主细胞(尤其是哺乳动物细胞)中具有增加的表达水平,例如,增加至少200%,例如,至少250%、至少300%、至少350%或至少400%。用于测定蛋白表达水平的测定试验是本领域已知的。本领域技术人员可以采用任何这样的测定试验,确定优化ABCD1编码核酸相比于参比核酸在表达效率上达到的优化程度。The ABCD1-encoding nucleic acid contained in the construct of the present invention may be any polynucleotide capable of encoding functional ABCD1 protein activity. However, in order to facilitate expression in mammalian cells, it is advantageous to optimize the nucleic acid sequence of the nucleic acid encoding the ABCD1 polypeptide. In one embodiment, the ABCD1 encoding nucleic acid used in the expression construct of the present invention comprises the polynucleotide sequence of SEQ ID NO: 1, or has at least about 95%, about 96%, about 97%, 98%, Polynucleotide sequences with 99% or greater nucleotide sequence identity. Advantageously, the optimized ABCD1-encoding nucleic acid, with respect to a reference nucleic acid (for example, a natural human ABCD1-encoding nucleic acid, such as a nucleic acid having the nucleotide sequence shown in SEQ ID NO: 15), is in an operably linked composition An increased expression level in a host cell, especially a mammalian cell, under the control of a promoter, eg, an increase of at least 200%, eg, at least 250%, at least 300%, at least 350%, or at least 400%. Assays for determining protein expression levels are known in the art. Any such assay can be used by one skilled in the art to determine the degree of optimization achieved in terms of expression efficiency of an optimized ABCD1-encoding nucleic acid compared to a reference nucleic acid.
在一个实施方案中,根据本发明的优化ABCD1编码核酸可以在5’端包含位于起始密码子上游的Kozak序列。用于本发明的Kozak序列可以是定义为GCCRCC的共有序列,其中R是嘌呤(即A或G),且其中所述序列位于起始密码子上游。在一个优选的实施方案中,本发明构建体包含Kozak序列,其中所述Kozak序列具有5’-GCCACC-3’序列。也可以在本发明构建体中使用其他不同的Kozak序列。In one embodiment, the optimized ABCD1-encoding nucleic acid according to the present invention may comprise a Kozak sequence located upstream of the start codon at the 5' end. The Kozak sequence used in the present invention may be a consensus sequence defined as GCCRCC, wherein R is a purine (ie A or G), and wherein said sequence is located upstream of the start codon. In a preferred embodiment, the construct of the invention comprises a Kozak sequence, wherein said Kozak sequence has a 5'-GCCACC-3' sequence. Other different Kozak sequences can also be used in the constructs of the invention.
启动子Promoter
本发明的构建体可以包含任何可以用于启动ABCD1编码核酸在目的哺乳动物细胞中表达的启动子。有利的是,包含根据本发明的突变组成型启动子CAR-Mut。本发明的CAR-Mut组成型启动子可以在多种组织中高效地启动外源基因表达,因而尤其适用在本发明治疗方法中应用,以兼顾X-ALD的外周和中枢治疗目的。The construct of the present invention may contain any promoter that can be used to promote the expression of an ABCD1-encoding nucleic acid in a mammalian cell of interest. Advantageously, the mutant constitutive promoter CAR-Mut according to the invention is included. The CAR-Mut constitutive promoter of the present invention can efficiently promote the expression of exogenous genes in various tissues, so it is especially suitable for use in the treatment method of the present invention, so as to take into account the peripheral and central therapeutic purposes of X-ALD.
在一个实施方案中,本发明构建体包含CAR-Mut启动子,其包含选自以下的多核苷酸:In one embodiment, the construct of the invention comprises a CAR-Mut promoter comprising a polynucleotide selected from the group consisting of:
(i)SEQ ID NO:5的多核苷酸,(i) the polynucleotide of SEQ ID NO:5,
(ii)与SEQ ID NO.5具有至少95%、96%、97%、98%、99%、99.5%同一性的多核苷酸,(ii) polynucleotides having at least 95%, 96%, 97%, 98%, 99%, 99.5% identity to SEQ ID NO.5,
(iii)在SEQ ID NO.5的多核苷酸中经取代、缺失或添加一个或几个核苷酸得到的多核苷酸,(iii) a polynucleotide obtained by substitution, deletion or addition of one or several nucleotides in the polynucleotide of SEQ ID NO.5,
且其中,所述多核苷酸在SEQ ID NO:5的核苷酸568位或相应位置上具有突变核苷酸C或G或A,更优选为T突变为C。And wherein, the polynucleotide has a mutated nucleotide C or G or A at nucleotide 568 of SEQ ID NO:5 or the corresponding position, more preferably T is mutated to C.
在一个优选实施方案中,本发明的突变启动子,相对于由不具有所述突变的相应多核苷酸组成的参照启动子,增加与其功能性连接的目的基因的表达,例如,使所述目的基因表达增加1%-70%,例如,至少5%,10%,20%,30%,40%,或至少50%,60%。In a preferred embodiment, the mutant promoter of the present invention, relative to a reference promoter consisting of a corresponding polynucleotide without said mutation, increases the expression of a gene of interest functionally linked thereto, e.g., makes said gene of interest Gene expression is increased by 1%-70%, eg, at least 5%, 10%, 20%, 30%, 40%, or at least 50%, 60%.
在再一优选实施方案中,本发明的突变启动子,相对于参照启动子,增加与之功能性连接的目的基因在哺乳动物细胞或组织中的表达,例如,增加所述目的基因在哺乳动物外周组织和/或中枢神经组织,尤其是中枢神经系统中的表达。优选地,所述哺乳动物为人或非人哺乳动物,例如,小鼠、大鼠和非人灵长类动物。In yet another preferred embodiment, the mutant promoter of the present invention increases the expression of the gene of interest functionally linked thereto in mammalian cells or tissues, for example, increases the expression of the gene of interest in mammals relative to the reference promoter. Expression in peripheral tissues and/or central nervous tissues, especially in the central nervous system. Preferably, the mammal is a human or a non-human mammal, eg, a mouse, a rat and a non-human primate.
在再一些实施方案中,所述启动子包含选自SEQ ID NOs:2至4之任一的核苷酸序列、或与之相差一个或几个核苷酸取代、缺失和/或添加且具有同等启动子活性的核苷酸序列。优选地,所述启动子包含或由SEQ ID NO:2的核苷酸序列组成。In some further embodiments, the promoter comprises a nucleotide sequence selected from any one of SEQ ID NOs: 2 to 4, or differs therefrom by one or several nucleotide substitutions, deletions and/or additions and has Nucleotide sequences with equivalent promoter activity. Preferably, the promoter comprises or consists of the nucleotide sequence of SEQ ID NO:2.
本领域技术人员可以采用本领域已知的任何启动子功能性测定试验(例如实施例1的荧光素酶报告基因表达测定试验),来确定任何两个启动子是否具有同等启动子活性。在一个实施方案中,在相同的测试条件下,与参照启动子(例如SEQ ID NO:5)相比,如果待测启动子具有相同或基本上相同的活性,例如参照启动子活性±10%、优选地±5%,或更优选±1%的活性,则可以认为待测启动子具有同等启动子活性。Those skilled in the art can use any promoter functional assay known in the art (such as the luciferase reporter gene expression assay in Example 1) to determine whether any two promoters have equivalent promoter activity. In one embodiment, under the same test conditions, compared with the reference promoter (such as SEQ ID NO: 5), if the promoter to be tested has the same or substantially the same activity, for example, the activity of the reference promoter ± 10% , preferably ±5%, or more preferably ±1%, then the promoter to be tested can be considered to have equivalent promoter activity.
miRNA靶序列miRNA target sequence
基因治疗在进入临床试验前,不仅需要对其功效进行优化,也需要谨慎地优化以证明其安全性。Before gene therapy enters clinical trials, it not only needs to be optimized for its efficacy, but also needs to be carefully optimized to prove its safety.
microRNA(miRNA)作为一种内源性的非编码小RNA,其可以以序列特异性方式,通常地通过阻抑靶mRNA翻译,来调节细胞基因表达。内源性miRNA可以对包含了与其完全互补的靶序列的表达盒中的转基因表达,产生抑制。阻抑水平与表达构建体所用的启动子、靶组织细胞中的相应miRNA丰度等因素相关。根据深入的研究,本发明人发现,在包含组成型启动子的表达构建体中,在3’UTR嵌入特定种类、数量和排列方式的miRNA靶细胞,可以增加基于该表达构建体的基因治疗安全性。MicroRNA (miRNA) is an endogenous small non-coding RNA that can regulate cellular gene expression in a sequence-specific manner, usually by repressing translation of target mRNAs. Endogenous miRNAs can suppress the expression of transgenes in expression cassettes that contain their perfectly complementary target sequences. The level of repression is related to factors such as the promoter used to express the construct, the corresponding miRNA abundance in the target tissue cells, and the like. According to in-depth research, the inventors found that in the expression construct containing a constitutive promoter, embedding specific types, quantities and arrangements of miRNA target cells in the 3' UTR can increase the safety of gene therapy based on the expression construct sex.
在一些实施方案中,因此,本发明的表达构建体还包含一个或多个miRNA靶序列,位于与目的编码核酸序列可操作连接的3’UTR中。不受任何具体理论的约束,包含miRNA靶序列在表达构建体中,将允许在产生相应miRNA的细胞和组织中调节(例如,抑制)目的基因的表达。由此,在一些实施方案中,本 发明表达构建体包含一个或多个miRNA靶序列,从而可以以细胞类型特异性方式下调ABCD1的表达。In some embodiments, therefore, the expression constructs of the invention further comprise one or more miRNA target sequences located in the 3'UTR operably linked to the coding nucleic acid sequence of interest. Without being bound by any particular theory, inclusion of miRNA target sequences in expression constructs will allow for the modulation (eg, inhibition) of expression of a gene of interest in cells and tissues producing the corresponding miRNA. Thus, in some embodiments, the expression constructs of the invention comprise one or more miRNA target sequences, so that the expression of ABCD1 can be downregulated in a cell type specific manner.
可用于本发明的miRNA靶序列包括,肌细胞特异性miRNA的靶序列和肝细胞特异性miRNA的靶序列。可以用于本发明的肌细胞特异性miRNA靶序列,可以选自miRNA1靶序列、miRNA206靶序列及其组合;例如,SEQ ID NO:6所示的miRNA1靶序列和SEQ ID NO:7所示的miRNA206靶序列。可以用于本发明的肝细胞特异性miRNA靶序列,可以是miRNA122靶序列,例如SEQ ID NO:8的miRNA122靶序列。miRNA target sequences that can be used in the present invention include target sequences of muscle cell-specific miRNA and target sequences of hepatocyte-specific miRNA. The muscle cell-specific miRNA target sequence that can be used in the present invention can be selected from miRNA1 target sequence, miRNA206 target sequence and combinations thereof; for example, the miRNA1 target sequence shown in SEQ ID NO:6 and the miRNA1 target sequence shown in SEQ ID NO:7 miRNA206 target sequence. The liver cell-specific miRNA target sequence that can be used in the present invention can be a miRNA122 target sequence, such as the miRNA122 target sequence of SEQ ID NO:8.
包含在根据本发明的表达构建体中的肝细胞特异性miRNA靶序列可以为至少一个或多个,且优选2-4个,其可以串联在一起、或与肌细胞特异性miRNA靶序列间隔排列。类似地,包含在根据本发明的表达构建体中的肌细胞特异性miRNA靶序列可以为至少一个或多个,且优选2-4个,其可以串联在一起、或与肝细胞特异性miRNA靶序列间隔排列。miRNA靶序列之间可以直接连接,或可以间隔少数个核苷酸,例如1-5个核苷酸。The liver cell-specific miRNA target sequence contained in the expression construct according to the present invention can be at least one or more, and preferably 2-4, which can be connected in series or arranged at intervals with the muscle cell-specific miRNA target sequence . Similarly, the muscle cell-specific miRNA target sequence contained in the expression construct according to the present invention can be at least one or more, and preferably 2-4, which can be connected in series or combined with the liver cell-specific miRNA target sequence. Sequence spaced. The miRNA target sequences can be connected directly, or can be separated by a few nucleotides, such as 1-5 nucleotides.
在一些实施方案中,根据本发明的表达构建体包含与所述ABCD1编码核酸可操作连接的3’UTR。所述的3’UTR包含至少1个拷贝(例如,1-8个拷贝,例如2,3,或4个拷贝)的肌细胞特异性miRNA的靶序列和/或至少1个拷贝(例如,1-8个拷贝,例如,1个或2个拷贝)的肝细胞特异性miRNA的靶序列。优选地,所述3’UTR包含至少1个拷贝(如3个拷贝)的肌细胞特异性miRNA靶序列和至少1个(例如2个拷贝的肝细胞特异性miRNA靶序列)。所述至少一个肌细胞特异性miRNA靶序列,与所述至少一个肝细胞特异性miRNA靶序列,在3’UTR中,可以以各种排列方式存在,但优选地,肌细胞特异性miRNA靶序列与肝细胞特异性miRNA靶序列间隔排列。在一个优选的实施方案中,所述3’UTR包含如下排列的miRNA靶序列:miRNA1靶序列-miRNA122靶序列-miRNA1靶序列-miRNA122靶序列-miRNA206靶序列;或miRNA206靶序列-miRNA122靶序列-miRNA1靶序列-miRNA122靶序列-miRNA1靶序列。在一些更优选的实施方案中,根据本发明的表达构建体包含与ABCD1编码核酸可操作连接的3’UTR,所述3’UTR包含SEQ ID NO:12的核苷酸序列。In some embodiments, an expression construct according to the invention comprises a 3' UTR operably linked to said ABCD1 encoding nucleic acid. The 3'UTR comprises at least 1 copy (eg, 1-8 copies, such as 2, 3, or 4 copies) of the target sequence of a myocyte-specific miRNA and/or at least 1 copy (eg, 1 - 8 copies, eg 1 or 2 copies) of the target sequence of the hepatocyte-specific miRNA. Preferably, the 3'UTR comprises at least 1 copy (such as 3 copies) of a myocyte-specific miRNA target sequence and at least 1 (such as 2 copies) of a hepatocyte-specific miRNA target sequence. The at least one myocyte-specific miRNA target sequence and the at least one hepatocyte-specific miRNA target sequence may be present in various arrangements in the 3'UTR, but preferably, the myocyte-specific miRNA target sequence Spaced with hepatocyte-specific miRNA target sequences. In a preferred embodiment, the 3'UTR comprises miRNA target sequences arranged as follows: miRNA1 target sequence-miRNA122 target sequence-miRNA1 target sequence-miRNA122 target sequence-miRNA206 target sequence; or miRNA206 target sequence-miRNA122 target sequence- miRNA1 target sequence - miRNA122 target sequence - miRNA1 target sequence. In some more preferred embodiments, the expression construct according to the present invention comprises a 3'UTR operably linked to the ABCD1 encoding nucleic acid, said 3'UTR comprising the nucleotide sequence of SEQ ID NO:12.
表达构建体expression construct
在一个方面,本发明提供了表达构建体。本发明的表达构建体可以有利地用于X-ALD疾病的基因治疗。In one aspect, the invention provides expression constructs. The expression construct of the present invention can be advantageously used for gene therapy of X-ALD disease.
在一个实施方案中,本发明的表达构建体包含以转录方向彼此功能性连接的如下元件:In one embodiment, the expression construct of the invention comprises the following elements functionally linked to each other in the direction of transcription:
-启动子,-Promoter,
-Kozak序列,-Kozak sequence,
-编码ABCD1的核酸,优选地,根据本发明的优化的ABCD1编码核酸,更优选地包含SEQ ID NO:1所示核苷酸序列的核酸,-encoding ABCD1 nucleic acid, preferably, according to the optimized ABCD1 encoding nucleic acid of the present invention, more preferably comprises the nucleic acid of nucleotide sequence shown in SEQ ID NO:1,
-至少一个miRNA靶序列;- at least one miRNA target sequence;
-一个或多个转录终止子,- one or more transcription terminators,
-polyA信号序列。- polyA signal sequence.
在一些实施方案中,表达构建体还包括两个ITR序列。例如,从5’末端到3’末端,表达构建体可以包含如下排列的元件:5’ITR-启动子-ABCD1编码序列-miRNA靶序列-polyA-3’ITR。在一些实施方案中,5’ITR和3’ITR相同。在另一实施方案中,5’ITR和3’ITR不同,且其一(优选3’ITR)为缺少功能性trs位点的ΔITR。在一个实施方案中,表达构建体中的5’ITR和3’ITR相同,均包含或由AAV2ITR序列组成。In some embodiments, the expression construct also includes two ITR sequences. For example, from the 5' end to the 3' end, the expression construct may comprise elements arranged as follows: 5'ITR-promoter-ABCD1 coding sequence-miRNA target sequence-polyA-3'ITR. In some embodiments, the 5'ITR and 3'ITR are the same. In another embodiment, the 5'ITR and the 3'ITR are different and one (preferably the 3'ITR) is a ΔITR lacking a functional trs site. In one embodiment, the 5'ITR and 3'ITR in the expression construct are identical and both comprise or consist of the AAV2 ITR sequence.
用于本发明表达构建体中的启动子可以是本发明上述任何实施方案中描述的CAR-Mut启动子。在一个优选的实施方案中,所述启动子包含或由SEQ ID No:2的核苷酸序列组成。在另一优选的实施方案中,所述启动子包含或由SEQ ID NO:3的核苷酸序列组成。在另一优选的实施方案中,所述启动子包含或由SEQ ID NO:4的核苷酸序列组成。The promoter used in the expression construct of the present invention may be the CAR-Mut promoter described in any of the above embodiments of the present invention. In a preferred embodiment, the promoter comprises or consists of the nucleotide sequence of SEQ ID No:2. In another preferred embodiment, the promoter comprises or consists of the nucleotide sequence of SEQ ID NO:3. In another preferred embodiment, the promoter comprises or consists of the nucleotide sequence of SEQ ID NO:4.
用于本发明表达构建体的ABCD1编码核酸可以是本发明上述任何实施方案中描述的ABCD1编码核酸。在一个优选的实施方案中,所述编码核酸包含或由SEQ ID NO:1的核苷酸序列组成。The ABCD1-encoding nucleic acid used in the expression construct of the present invention may be the ABCD1-encoding nucleic acid described in any of the above embodiments of the present invention. In a preferred embodiment, the encoding nucleic acid comprises or consists of the nucleotide sequence of SEQ ID NO:1.
用于本发明表达构建体的miRNA靶序列可以是本发明上述任何实施方案中描述的miRNA靶序列,尤其是,至少一个根据本发明的肝细胞特异性miRNA靶序列和至少一个根据本发明的肌细胞特异性miRNA靶序列,优选地,包含SEQ ID NO:12的核苷酸序列。The miRNA target sequence used in the expression construct of the present invention may be the miRNA target sequence described in any of the above embodiments of the present invention, in particular, at least one hepatocyte-specific miRNA target sequence according to the present invention and at least one muscle specific miRNA target sequence according to the present invention. Cell-specific miRNA target sequence, preferably, comprises the nucleotide sequence of SEQ ID NO:12.
可用于本发明的转录终止子,包括可以终止核酸翻译的任何核酸序列。例如,终止子可以是“TAG”,”TGA”或“TAA”,对应的RNA序列为“UAG”,“UGA”或“UAA”。Transcription terminators useful in the present invention include any nucleic acid sequence that can terminate translation of a nucleic acid. For example, the terminator can be "TAG", "TGA" or "TAA", and the corresponding RNA sequence is "UAG", "UGA" or "UAA".
在一些实施方案中,本发明表达构建体还包含至少一个polyA尾位于ABCD1编码核酸和miRNA靶序列的下游。任何合适的polyA序列均可以使用,包括但不限于hGHpolyA,BGHpolyA,SV40晚期polyA序列、兔β-珠蛋白polyA序列、或其任何变体。在一个优选的实施方案中,polyA是BGHpolyA,例如SEQ ID NO:13所示的polyA,或与SEQ ID NO:13具有至少80%,85%,90%,95%,96%,97%,98%或99%的核苷酸序列同一性的polyA多核苷酸序列。In some embodiments, the expression construct of the invention further comprises at least one polyA tail located downstream of the ABCD1 encoding nucleic acid and the miRNA target sequence. Any suitable polyA sequence may be used, including but not limited to hGHpolyA, BGHpolyA, SV40 late polyA sequence, rabbit β-globin polyA sequence, or any variant thereof. In a preferred embodiment, polyA is BGHpolyA, such as polyA shown in SEQ ID NO: 13, or has at least 80%, 85%, 90%, 95%, 96%, 97% with SEQ ID NO: 13, A polyA polynucleotide sequence having 98% or 99% nucleotide sequence identity.
在一些方面,本发明也提供包含本发明表达构建体的载体。在一些实施方案中,所述载体是质粒(例如用于重组病毒颗粒生产的质粒)。在另一些实施方案中,所述载体是病毒载体,例如重组AAV载体或杆状病毒载体。在一些实施方案中,重组AAV载体的基因组是单链的(例如单链DNA)。在一些实施方案中,重组AAV载体的基因组是自互补的。In some aspects, the invention also provides vectors comprising expression constructs of the invention. In some embodiments, the vector is a plasmid (eg, a plasmid used for recombinant viral particle production). In other embodiments, the vector is a viral vector, such as a recombinant AAV vector or a baculovirus vector. In some embodiments, the genome of the recombinant AAV vector is single-stranded (eg, single-stranded DNA). In some embodiments, the genome of the recombinant AAV vector is self-complementary.
在再一方面,本发明也提供了包含本发明的表达构建体或载体的宿主细胞,例如哺乳动物细胞或昆虫细胞。在一些实施方案中,所述细胞可以用于生产重组AAV病毒。In a further aspect, the present invention also provides host cells, such as mammalian cells or insect cells, comprising the expression construct or vector of the present invention. In some embodiments, the cells can be used to produce recombinant AAV viruses.
重组AAV载体recombinant AAV vector
在一个方面,本发明提供了重组AAV载体。本发明的重组AAV载体尤其可用于X-ALD疾病或改善与之相关的症状。在一个实施方案中,重组AAV载体包含衣壳和位于衣壳中的核酸,在本文中也称作“重组AAV载体的基因组”。重组AAV载体的基因组包含多个元件,包括但不限于两个反向末端重复(ITR,即,5’-ITR和3’-ITR),以及位于两个ITR之间的其它元件,包括启动子、异源基因、和polyA尾。优选地,两个ITR之间还可以包含至少一个miRNA靶序列。In one aspect, the invention provides recombinant AAV vectors. The recombinant AAV vectors of the present invention are particularly useful for X-ALD disease or ameliorating symptoms associated therewith. In one embodiment, the recombinant AAV vector comprises a capsid and nucleic acid located within the capsid, also referred to herein as the "genome of the recombinant AAV vector." The genome of the recombinant AAV vector contains multiple elements, including but not limited to two inverted terminal repeats (ITRs, i.e., 5'-ITR and 3'-ITR), and other elements located between the two ITRs, including the promoter , a heterologous gene, and a polyA tail. Preferably, at least one miRNA target sequence may also be included between the two ITRs.
在本文中,腺相关病毒(AAV)包括但不限于,任何血清型的AAV,例如1,2,3,4,5,6,7,8,9,10,11,12型AAV,及具有人工改变的衣壳蛋白的AAV。各种血清型和人工AAV的基因组序列及其天然反向末端重复(ITR)序列、Rep蛋白和衣壳cap蛋白是本领域已知的。这些序列可以在公开数据库例如GenBank或文献中找到。As used herein, adeno-associated virus (AAV) includes, but is not limited to, AAV of any serotype, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 AAV, and AAV with AAV with artificially altered capsid proteins. The genome sequences of various serotypes and artificial AAVs and their native inverted terminal repeat (ITR) sequences, Rep proteins and capsid cap proteins are known in the art. These sequences can be found in public databases such as GenBank or in the literature.
在一些实施方案中,本发明提供包含衣壳的重组AAV病毒载体,其中所述衣壳由能够跨血脑屏障的衣壳蛋白,例如AAV9、AAVPHP.B、AAVPHP.eB衣壳蛋白构成。在一些实施方案中,本发明的重组AAV载体转导中枢神经系统(CNS)的细胞,包括神经元细胞和神经胶质细胞,也转导外周非神经元细胞。在另一些实施方案中,重组AAV载体在全身给药后能靶向和转导神经元细胞、星形胶质细胞和小胶质细胞。在另一实施方案中,重组AAV载体在全身给药后能靶向和转导受试者的外周器官和中枢神经系统。在再一实施方案中,重组AAV载体在全身给药后能靶向和转导受试者的多数个组织(例如,脑、脊髓、肾上腺),且优选地,与未接受重组AAV载体施用的对照受试者相比,重组AAV载体在所述靶向和转导的组织中导致更高的目的外源基因(在本申请中ABCD1编码基因)的表达和/或VLCFA水平降低。在生物化学水平上,可以通过饱和直链VLCFA(C24:0和C26:0,尤其是C26:0)的细胞水平降低,检测本发明重组AAV载体或基因药物的治疗功效。In some embodiments, the present invention provides a recombinant AAV viral vector comprising a capsid, wherein the capsid is composed of a capsid protein capable of crossing the blood-brain barrier, such as AAV9, AAVPHP.B, AAVPHP.eB capsid protein. In some embodiments, the recombinant AAV vectors of the invention transduce cells of the central nervous system (CNS), including neuronal cells and glial cells, as well as peripheral non-neuronal cells. In other embodiments, recombinant AAV vectors are capable of targeting and transducing neuronal cells, astrocytes, and microglia after systemic administration. In another embodiment, the recombinant AAV vector is capable of targeting and transducing the peripheral organs and central nervous system of a subject following systemic administration. In yet another embodiment, the recombinant AAV vector is capable of targeting and transducing multiple tissues (e.g., brain, spinal cord, adrenal gland) of a subject following systemic administration, and preferably, is compatible with non-administered recombinant AAV vectors. The recombinant AAV vector resulted in higher expression of the foreign gene of interest (the gene encoding ABCD1 in this application) and/or reduced VLCFA levels in the targeted and transduced tissues compared to control subjects. At the biochemical level, the therapeutic efficacy of the recombinant AAV vector or gene drug of the present invention can be detected by decreasing the cellular level of saturated linear VLCFA (C24:0 and C26:0, especially C26:0).
在一些实施方案中,本发明的重组AAV载体具有来自AAV9血清型的衣壳(也本文中也称作AAV9载体);优选地,所述重组AAV载体在其基因组中具有来自AAV2的野生型或变体ITR序列(也在本文中也称作AAV2/9载体)。In some embodiments, the recombinant AAV vectors of the invention have a capsid from an AAV9 serotype (also referred to herein as an AAV9 vector); preferably, the recombinant AAV vector has a wild-type or capsid from AAV2 in its genome. Variant ITR sequences (also referred to herein as AAV2/9 vectors).
在一些实施方案中,本发明的重组AAV载体的两个ITR序列均是全长ITR(例如长度为约125-145bp,并含有功能性的Rep结合位点(RBS)和末端解链位点(trs))。在一些实施方案中,全长功能性ITR被用于生产单链重组AAV载体(ssAAV)。在再一些实施方案中,所述重组AAV载体的ITR之一是截短的。在一些 实施方案中,截短的ITR缺少功能性末端解链位点trs并被用于生产自我互补型重组AAV载体(scAAV载体)。In some embodiments, the two ITR sequences of the recombinant AAV vector of the present invention are full-length ITRs (for example, about 125-145 bp in length, and contain a functional Rep binding site (RBS) and a terminal melting site ( trs)). In some embodiments, full-length functional ITRs are used to produce single-chain recombinant AAV vectors (ssAAV). In still other embodiments, one of the ITRs of the recombinant AAV vector is truncated. In some embodiments, truncated ITRs lack functional terminal melting sites trs and are used to produce self-complementary recombinant AAV vectors (scAAV vectors).
因此,在一个方面,本发明提供了一种重组腺相关病毒(AAV)载体,其中所述重组AAV载体在其基因组中包含:5’和3’AAV反向末端重复(ITR)序列,和位于5’和3’ITR之间的根据本发明的表达构建体。Therefore, in one aspect, the present invention provides a recombinant adeno-associated virus (AAV) vector, wherein said recombinant AAV vector comprises in its genome: 5' and 3' AAV inverted terminal repeat (ITR) sequences, and located at Expression construct according to the invention between the 5' and 3'ITR.
在一些实施方案中,本发明重组AAV病毒载体在施用于哺乳动物靶细胞(包括体外、体内和体内细胞)后,造成细胞中ABCD1基因表达的量或ABCD1蛋白质表达的量,比未施用前增加,由此降低细胞中VLCFA的水平,例如降低至少1-3倍。在一些实施方案中,本发明重组病毒载体在施用于受试者后,导致肾上腺和CNS例如小脑、大脑、脊髓中的VLCFA水平降低,例如,与未施用前相比,降低至少0.5倍,例如至少1-3倍。可以根据本领域已知用于VLCFA定量的标准方法,进行该降低倍数的测定。In some embodiments, after the recombinant AAV viral vector of the present invention is administered to mammalian target cells (including cells in vitro, in vivo and in vivo), the amount of ABCD1 gene expression or the amount of ABCD1 protein expression in the cells is increased compared with that before the administration , thereby reducing the level of VLCFA in the cell, eg by at least 1-3 fold. In some embodiments, the recombinant viral vector of the present invention, after being administered to a subject, results in a decrease in VLCFA levels in the adrenal gland and CNS such as cerebellum, brain, spinal cord, for example, at least 0.5 times lower than before administration, for example At least 1-3 times. Determination of this fold reduction can be performed according to standard methods known in the art for the quantification of VLCFAs.
II.重组AAV载体的制备II. Preparation of Recombinant AAV Vectors
本领域中具有相对成熟的AAV载体包装系统,便于规模化生产AAV载体。There is a relatively mature AAV vector packaging system in the field, which facilitates large-scale production of AAV vectors.
目前常用的AAV载体包装系统主要包括三质粒共转染系统、腺病毒作为辅助病毒的系统、单纯疱疹病毒(Herpes simplex virus type 1,HSV1)作为辅助病毒的包装系统、以及基于杆状病毒的包装系统。每种包装系统都各具特点,本领域技术人员可以根据需要做出合适的选择。Currently commonly used AAV vector packaging systems mainly include three-plasmid co-transfection system, adenovirus as helper virus system, Herpes simplex virus type 1 (HSV1) as helper virus packaging system, and baculovirus-based packaging system. system. Each packaging system has its own characteristics, and those skilled in the art can make appropriate choices according to needs.
三质粒共转染包装系统因无需辅助病毒,安全性高,是应用最为广泛的AAV载体包装系统,也是目前国际上主流的生产系统。略显不足的是,高效大规模转染方法的缺失限制了三质粒转染系统在AAV载体大规模制备中的应用。The three-plasmid co-transfection packaging system is the most widely used AAV vector packaging system because it does not require helper virus and has high safety. It is also the mainstream production system in the world. The slight disadvantage is that the absence of an efficient large-scale transfection method limits the application of the three-plasmid transfection system in the large-scale preparation of AAV vectors.
本发明的重组AAV病毒载体可以使用本领域已知的任何合适的方法来生产。在一个实施方案中,本发明重组AAV病毒采用三质粒包装系统进行生产。在另一实施方案中,本发明重组AAV病毒采用杆状病毒包装系统进行生产。The recombinant AAV viral vectors of the invention can be produced using any suitable method known in the art. In one embodiment, the recombinant AAV virus of the present invention is produced using a three-plasmid packaging system. In another embodiment, the recombinant AAV virus of the present invention is produced using a baculovirus packaging system.
III.药物组合物III. Pharmaceutical composition
再一方面,本发明提供了包含本发明的重组AAV病毒载体的药物组合物。本发明的药物组合物优选地包含可药用赋形剂、稀释剂或载体。本发明的药物组合物可以配制为任何合适的制剂形式。In yet another aspect, the present invention provides a pharmaceutical composition comprising a recombinant AAV viral vector of the present invention. The pharmaceutical composition of the present invention preferably comprises a pharmaceutically acceptable excipient, diluent or carrier. The pharmaceutical compositions of the present invention may be formulated in any suitable preparation form.
用于配制的合适可药用赋形剂、稀释剂或载体的实例在本领域中是众所周知的,包括例如,磷酸盐缓冲盐溶液,水,乳液,例如油/水乳液,各种类型的润湿剂,无菌溶液等。制剂可以通过常规方法配制,并以合适的剂量向受试者给药。合适配制的组合物的施用可以通过不同的方式来实现,例如。通过静脉内,腹膜内,皮下,肌肉内,局部或皮内给药。具体施用途径尤其取决于药物组合物中包含的载体的类型。剂 量方案将由主治医师和其他临床因素决定。如医学领域众所周知的,任何一名患者的剂量取决于许多因素,包括患者的体型,体表面积,年龄,性别,所要施用的特定活性剂,所用的时间和途径,所用药物的种类和阶段。感染或疾病,一般健康状况、以及其他药物的联用。Examples of suitable pharmaceutically acceptable excipients, diluents or carriers for formulation are well known in the art and include, for example, phosphate buffered saline, water, emulsions, such as oil/water emulsions, various types of Wet agent, sterile solution, etc. Preparations can be formulated by conventional methods, and administered to subjects in appropriate doses. Administration of a suitably formulated composition can be achieved in different ways, eg. Administration is by intravenous, intraperitoneal, subcutaneous, intramuscular, topical or intradermal. The particular route of administration depends, inter alia, on the type of carrier included in the pharmaceutical composition. The dosage regimen will be determined by the attending physician and other clinical factors. As is well known in the medical arts, the dosage for any one patient will depend on many factors, including the patient's size, body surface area, age, sex, the particular active agent to be administered, the timing and route used, and the type and phase of the drug used. Infections or diseases, general health conditions, and combinations of other medications.
在一些实施方案中,本发明的药物组合物可以包含第二活性剂。在一些实施方案中,第二活性剂是用于治疗或缓解X-ALD的药物,或能够降低药物施用时副反应的组分。In some embodiments, the pharmaceutical compositions of the present invention may include a second active agent. In some embodiments, the second active agent is a drug for treating or alleviating X-ALD, or a component capable of reducing side effects when the drug is administered.
本发明的药物组合物可以通过任何合适途径给药,包括全身给药和局部给药。在一些实施方案中,本发明药物组合物用于全身给药,尤其是静脉注射给药。因此,在一个实施方案中,本发明提供了包含本发明重组AAV载体的药物组合物,其中所述药物组合物是静脉注射制剂,或适用于配制为静脉注射制剂的冻干稳定制剂。在另一些实施方案中,本发明药物组合物适用于局部给药,例如直接施用予受试者的待治疗器官或组织中或附近。因此,在一个实施方案中,本发明提供了包含本发明重组AAV载体的药物组合物,其中所述药物组合物是适用于局部给药的制剂。The pharmaceutical compositions of the present invention may be administered by any suitable route, including systemic administration and topical administration. In some embodiments, the pharmaceutical composition of the present invention is for systemic administration, especially intravenous administration. Accordingly, in one embodiment, the present invention provides a pharmaceutical composition comprising a recombinant AAV vector of the present invention, wherein said pharmaceutical composition is an intravenous formulation, or a lyophilized stable formulation suitable for formulation as an intravenous formulation. In other embodiments, the pharmaceutical composition of the present invention is suitable for local administration, for example, directly in or near the organ or tissue to be treated in a subject. Accordingly, in one embodiment, the present invention provides a pharmaceutical composition comprising a recombinant AAV vector of the present invention, wherein said pharmaceutical composition is a formulation suitable for topical administration.
IV.治疗方法IV. Treatment
在另一方面,本发明涉及使用本发明的重组AAV载体或包含其的药物组合物治疗疾病的方法。在一个的实施方案中,所述疾病是X-ALD。在另一实施方案中,所述疾病是ABCD1基因缺陷。在一个实施方案中,所述方法包括:将本发明的重组AAV载体或药物组合物施用给有需要的受试者。所述重组AAV载体或药物组合物可以通过任何适宜的途径施用,包括但不限于,肌内,皮下,脊髓内,脑室内,鞘内,静脉内,膈肌内,胸腔内,腹膜内。优选地,本发明的重组AAV载体或药物组合物通过全身给药方式,尤其是静脉内给药方式递送给受试者。在一些实施方案中,所述治疗是治疗性的。在另一些实施方案中,所述治疗是预防性的。在一些实施方案中,受试者是哺乳动物,其中所述哺乳动物尤其是人、灵长类、狗、马、牛,特别是人类受试者。In another aspect, the present invention relates to methods of treating diseases using the recombinant AAV vectors of the present invention or pharmaceutical compositions comprising the same. In one embodiment, the disease is X-ALD. In another embodiment, the disease is a defect in the ABCD1 gene. In one embodiment, the method comprises: administering a recombinant AAV vector or pharmaceutical composition of the invention to a subject in need thereof. The recombinant AAV vector or pharmaceutical composition can be administered by any suitable route, including but not limited to, intramuscular, subcutaneous, intraspinal, intracerebroventricular, intrathecal, intravenous, intradiaphragmatic, intrathoracic, intraperitoneal. Preferably, the recombinant AAV vector or pharmaceutical composition of the present invention is delivered to a subject by systemic administration, especially intravenous administration. In some embodiments, the treatment is therapeutic. In other embodiments, the treatment is prophylactic. In some embodiments, the subject is a mammal, wherein the mammal is especially a human, primate, dog, horse, cow, especially a human subject.
在涉及治疗受试者的方法中,在一些实施方案中,治疗包括以下之任一项或多项:(1)阻止或延迟疾病的发作;(2)减轻疾病的严重程度;(3)减轻或阻止疾病的至少一个症状的出现和/或恶化;(4)改善疾病相关的神经变性和/或受试者行为;和(5)延长受试者的生存期。应当理解,治疗X-ALD尽管涵盖,但并不要求,完全消除所述疾病或与其相关的症状。In methods involving treating a subject, in some embodiments, treatment includes any one or more of: (1) arresting or delaying the onset of the disease; (2) lessening the severity of the disease; (3) lessening the severity of the disease; or prevent the onset and/or worsening of at least one symptom of the disease; (4) improve disease-related neurodegeneration and/or behavior of the subject; and (5) prolong the survival of the subject. It should be understood that treatment of X-ALD, while encompassing, does not require complete elimination of the disease or symptoms associated therewith.
可以接受本发明方法治疗/预防的受试者包括Addison型,脑型(儿童脑型、青少年脑型和成人脑型),伴或不伴颅内脱髓鞘的肾上腺脊髓神经病型(AMN)、无症状或症状前期型、以及杂合子型。在一个实施方案中,受试者是脑型ALD患者。在另一些实施方案中,受试者是AMN患者。在再一些实施方案中,受试者是无症状患者。在另一些实施方案中,受试者是表现出ALD或AMN发病相关症状,例如在血浆中蓄积 高水平VLCFA的个体。在再一些实施方案中,受试者是由于家族史而具有ALD或AMN病发生风险的个体;或是遗传检测在ABCD1基因中包含一个或多个与ALD或AMN相关突变的个体。Subjects who can be treated/prevented by the method of the present invention include Addison type, cerebral type (children's brain type, adolescent brain type and adult brain type), adrenomyeloneuropathy type (AMN) with or without intracranial demyelination, Asymptomatic or presymptomatic, and heterozygous. In one embodiment, the subject is a patient with cerebral ALD. In other embodiments, the subject is an AMN patient. In yet other embodiments, the subject is an asymptomatic patient. In other embodiments, the subject is an individual exhibiting symptoms associated with the onset of ALD or AMN, such as accumulation of high levels of VLCFA in plasma. In yet other embodiments, the subject is an individual at risk for ALD or AMN disease due to family history; or an individual whose genetic testing includes one or more mutations associated with ALD or AMN in the ABCD1 gene.
因此,在一个方面,本发明提供了一种治疗或预防X染色体连锁肾上腺脑白质营养不良(X-ALD)和/或改善与X-ALD相关的症状的方法,其中所述方法包括向有需要的受试者施用根据本发明的重组AAV病毒载体或根据本发明的药物组合物。优选地,所述方法包括腹膜内、肌内、动脉内、静脉内、鞘内或脑室内给药,更优选地静脉内注射,所述的重组AAV病毒载体。Therefore, in one aspect, the present invention provides a method for treating or preventing X-chromosome-linked adrenoleukodystrophy (X-ALD) and/or improving symptoms associated with X-ALD, wherein the method comprises providing A subject is administered the recombinant AAV viral vector according to the present invention or the pharmaceutical composition according to the present invention. Preferably, said method comprises intraperitoneal, intramuscular, intraarterial, intravenous, intrathecal or intraventricular administration, more preferably intravenous injection, of said recombinant AAV viral vector.
本发明的有益技术效果Beneficial technical effect of the present invention
1.本发明的X-ALD基因治疗药物(例如,根据本发明的重组AAV病毒载体或根据本发明的药物组合物)能够突破血脑屏障,从而能降低全身多组织脏器中的极长链脂肪酸水平。使用腺相关病毒AAV9作为载体时,药物能够有更广泛的生物分布,尤其是能够覆盖中枢神经系统,改善中枢神经中的极长链脂肪酸水平,阻止脑白质病变。1. The X-ALD gene therapy drug of the present invention (for example, according to the recombinant AAV virus vector of the present invention or according to the pharmaceutical composition of the present invention) can break through the blood-brain barrier, thereby reducing the extremely long chain in the multi-organ organs of the whole body. fatty acid levels. When the adeno-associated virus AAV9 is used as a vector, the drug can have a wider biodistribution, especially to cover the central nervous system, improve the level of very long chain fatty acids in the central nervous system, and prevent white matter lesions.
2.本发明的X-ALD基因治疗药物(例如,根据本发明的重组AAV病毒载体或根据本发明的药物组合物)通过启动子和编码基因优化,增加了药物的体内表达水平,同时病毒载体基因组的稳定性得以提升,由此可以更长效地降低组织中和血液中极长链脂肪酸的水平。2. The X-ALD gene therapy drug of the present invention (for example, according to the recombinant AAV viral vector of the present invention or according to the pharmaceutical composition of the present invention) has increased the expression level of the drug in vivo through the optimization of the promoter and the coding gene, and the viral vector Genomic stability is enhanced, resulting in longer-lasting reductions in tissue and blood VLCFA levels.
3.本发明的X-ALD基因治疗药物(例如,根据本发明的重组AAV病毒载体或根据本发明的药物组合物)具有降低的外周组织器官毒性。通过选择组织特异性表达的miRNA的靶序列,本发明的X-ALD基因治疗药物可降低转基因过表达对外周器官的毒性,尤其是心肌组织毒性。3. The X-ALD gene therapy drug of the present invention (for example, the recombinant AAV virus vector according to the present invention or the pharmaceutical composition according to the present invention) has reduced toxicity to peripheral tissues and organs. By selecting the target sequence of tissue-specifically expressed miRNA, the X-ALD gene therapy drug of the present invention can reduce the toxicity of transgene overexpression to peripheral organs, especially myocardial tissue toxicity.
4.本发明的X-ALD基因治疗药物(例如,根据本发明的重组AAV病毒载体或根据本发明的药物组合物),经静脉注射至ABCD1基因缺陷模型小鼠体内后,能在小鼠体内高效持续稳定地表达ABCD1蛋白,而表达产生的ABCD1蛋白能参与细胞内的极长链脂肪酸的降解,降低其在细胞中的积累,并使其维持在正常水平,从而消除细胞中由极长链脂肪酸过多的积累导致的各种疾病症状,达到治疗目的。此外,尽管在静脉注射下,心脏、肝脏的药物暴露程度最大,但在使用本发明的X-ALD基因治疗药物达到治疗目的的同时,未见心脏、肝脏出现药物相关的毒性改变,说明本发明药物安全性较好。4. The X-ALD gene therapy drug of the present invention (for example, according to the recombinant AAV virus vector of the present invention or according to the pharmaceutical composition of the present invention), after intravenous injection into the ABCD1 gene-deficient model mice, can Efficient, continuous and stable expression of ABCD1 protein, and the ABCD1 protein produced by expression can participate in the degradation of extremely long-chain fatty acids in cells, reduce its accumulation in cells, and maintain it at a normal level, thereby eliminating cells caused by extremely long-chain fatty acids Various disease symptoms caused by excessive accumulation of fatty acids, to achieve the purpose of treatment. In addition, although the drug exposure of the heart and liver is the largest under intravenous injection, when the X-ALD gene therapy drug of the present invention is used to achieve the therapeutic purpose, no drug-related toxic changes have been seen in the heart and liver, indicating that the drug-related changes of the present invention Drug safety is better.
5.本发明的X-ALD基因治疗药物(例如,根据本发明的重组AAV病毒载体或根据本发明的药物组合物)在静脉注射和治疗X-ALD模型小鼠后,中枢神经系统的极长链脂肪酸水平下降显著,可恢复至正常水平,且外周组织、血液中的极长链脂肪酸也有明显的改变。这说明本发明药物可以采用鞘内注射的方式,来治疗X-ALD。一方面,对于改善中枢神经系统的指标异常,鞘内注射的治疗效果将更佳显著。与此同时,鞘内注射后AAV载体基因组的生物分布会更集中于中枢神经系统,心脏、肝脏等外周脏器的载体 生物分布比例偏低(暴露量更低),因此药物的安全性将得到更进一步的提升。5. The X-ALD gene therapy medicine of the present invention (for example, according to the recombinant AAV virus vector of the present invention or according to the pharmaceutical composition of the present invention) after intravenous injection and treatment of X-ALD model mice, the extremely long The level of chain fatty acids decreased significantly and returned to normal levels, and the very long chain fatty acids in peripheral tissues and blood also changed significantly. This shows that the medicine of the present invention can be used in the treatment of X-ALD by intrathecal injection. On the one hand, the treatment effect of intrathecal injection will be better and more significant for the improvement of abnormal indicators of the central nervous system. At the same time, after intrathecal injection, the biodistribution of the AAV vector genome will be more concentrated in the central nervous system, and the proportion of vector biodistribution in peripheral organs such as the heart and liver is low (lower exposure), so the safety of the drug will be improved. Further improvement.
实施例Example
本发明以X-ALD小鼠模型为例,通过独特设计构建的基因表达构建体,实现了对疾病症状的改善,并同时达成了良好的药物安全性。下面结合具体实施例及附图对本发明作进一步说明:Taking the X-ALD mouse model as an example, the present invention realizes the improvement of disease symptoms and simultaneously achieves good drug safety through a uniquely designed and constructed gene expression construct. Below in conjunction with specific embodiment and accompanying drawing, the present invention will be further described:
实施例1:ABCD1基因编码序列优化Embodiment 1: ABCD1 gene coding sequence optimization
天然的ABCD1编码序列(CDS)虽然可以有效表达目的基因,但是表达翻译的阻力仍在,存在多处反式作用元件和GC%分布不均匀等等限制因素。Although the natural ABCD1 coding sequence (CDS) can effectively express the target gene, the resistance to expression and translation still exists, there are many limiting factors such as trans-acting elements and uneven distribution of GC%.
在本实施例中,对hABCD1编码基因(UniProtKB-P33897)的核苷酸序列进行优化,并通过在HEK293细胞系中体外表达和WESTERN BLOT检测ABCD1蛋白表达水平,确定了改善hABCD1表达效率的优化序列(coABCD1)。In this example, the nucleotide sequence of the gene encoding hABCD1 (UniProtKB-P33897) was optimized, and the optimized sequence for improving the expression efficiency of hABCD1 was determined by in vitro expression in HEK293 cell line and detection of ABCD1 protein expression level by WESTERN BLOT (coABCD1).
由金斯瑞生物科技有限公司合成野生型ABCD1编码核酸序列(来自NCBI登录号NM_000033.3的序列)和优化后的coABCD1编码核酸序列(SEQ ID NO:1)。合成后的序列克隆至pUC57 simple载体(金斯瑞生物科技,南京)上。之后,通过KpnI和EcoRI位点,将合成的野生型编码序列和优化编码序列分别克隆至pAAV载体质粒中,嵌入至CA启动子和polyA之间(如图1A所示)。经测序鉴定后,冻存入库并提取质粒进行体外转染实验。简言之,小量提取质粒,使用Lipofectamine 2000瞬时转染HEK293细胞系。转染后6小时更换培养基,并于48小时后收集细胞,裂解提取细胞总蛋白。使用10%SDS-PAGE凝胶电泳分离蛋白条带并用湿转法转印蛋白至PVDF膜上。使用Anti-ABCD1抗体(1:2000,Abcam,ab197013)孵育过夜。经漂洗、二抗(HRP标记羊抗兔IgG,中杉金桥,ZB-2301)孵育以及再次漂洗后,采用ECL增强型化学发光试剂(生工,C500044)进行ECL发光显色。显色结果经灰度扫描处理分析。The wild-type ABCD1 coding nucleic acid sequence (from the sequence of NCBI accession number NM_000033.3) and the optimized coABCD1 coding nucleic acid sequence (SEQ ID NO: 1) were synthesized by GenScript Biotechnology Co., Ltd. The synthesized sequence was cloned into the pUC57 simple vector (GenScript Biotechnology, Nanjing). After that, the synthetic wild-type coding sequence and the optimized coding sequence were respectively cloned into the pAAV vector plasmid through KpnI and EcoRI sites, and embedded between the CA promoter and polyA (as shown in Figure 1A). After sequencing and identification, they were frozen and stored in the library and the plasmids were extracted for in vitro transfection experiments. Briefly, plasmids were miniprepped and HEK293 cell line was transiently transfected using Lipofectamine 2000. The medium was changed 6 hours after transfection, and the cells were collected 48 hours later, and the total protein of the cells was lysed and extracted. The protein bands were separated by 10% SDS-PAGE gel electrophoresis and transferred to PVDF membrane by wet transfer method. Anti-ABCD1 antibody (1:2000, Abcam, ab197013) was used to incubate overnight. After rinsing, incubation with secondary antibody (HRP-labeled goat anti-rabbit IgG, Zhongshan Jinqiao, ZB-2301) and rinsing again, ECL-enhanced chemiluminescence reagent (Sanko, C500044) was used for ECL luminescent color development. The color rendering results were processed and analyzed by grayscale scanning.
如图1B所示,基因编码序列优化后,表达水平提升约400%。证明序列优化有效地改变了ABCD1基因的表达效率。As shown in Figure 1B, after optimization of the gene coding sequence, the expression level increased by about 400%. It was proved that sequence optimization effectively changed the expression efficiency of ABCD1 gene.
实施例2:启动子优化Example 2: Promoter optimization
2.1.CAR-Mut启动子构建2.1. Construction of CAR-Mut promoter
在由人CMV病毒的增强子序列和鸡β-actin蛋白的基础启动子组成的CA启动子上,在序列3’端引入人TATA盒结合蛋白相关因子1基因(GenBank:NG_012771.2)中第62804位至62890位的内含子序列,得到命名为CAR启动子。对CAR启动子进行改造,将启动子末端第568位的T突变为非T核苷酸,获得CAR-Mut启动子,即,CAR-MutC(具有突变T568C,序列如SEQ ID NO:2所示),CAR-MutA(具有突变 T568A,序列见SEQ ID NO:3所示),和CAR-MutG(具有突变T568G,序列见SEQ ID NO:4所示)。On the CA promoter composed of the enhancer sequence of human CMV virus and the basal promoter of chicken β-actin protein, the 3' end of the sequence was introduced into the human TATA box binding protein-related factor 1 gene (GenBank: NG_012771.2) The intron sequence from position 62804 to position 62890 was named CAR promoter. Transform the CAR promoter, mutate the T at the 568th position at the end of the promoter to a non-T nucleotide, and obtain the CAR-Mut promoter, that is, CAR-MutC (with mutation T568C, the sequence is shown in SEQ ID NO: 2 ), CAR-MutA (with mutation T568A, sequence shown in SEQ ID NO: 3), and CAR-MutG (with mutation T568G, sequence shown in SEQ ID NO: 4).
2.2.CAR-Mut启动子体外活性的表征2.2. Characterization of in vitro activity of CAR-Mut promoter
为表征启动子CAR-Mut,构建了图2所示的pscAAV-CAR-Gluc质粒载体,包含:In order to characterize the promoter CAR-Mut, the pscAAV-CAR-Gluc plasmid vector shown in Figure 2 was constructed, including:
i)来自AAV2基因组(GenBank No.AF043303)5’端的ITR;i) ITR from the 5' end of the AAV2 genome (GenBank No.AF043303);
ii)CAR启动子,序列如SEQ ID NO:5所示;ii) CAR promoter, the sequence is as shown in SEQ ID NO:5;
iii)Gluc,编码荧光素酶报告基因的核苷酸序列;iii) Gluc, the nucleotide sequence encoding the luciferase reporter gene;
iv)牛生长激素的多聚核苷酸加尾信号,也缩写为BGH polyA;iv) the polynucleotide tailing signal of bovine growth hormone, also abbreviated as BGH polyA;
v)以AAV2基因组(GenBank No.AF043303)3’端ITR序列为基础,缺失该序列中的trs序列和D序列,得到的ΔITR。v) Based on the ITR sequence at the 3' end of the AAV2 genome (GenBank No.AF043303), the trs sequence and the D sequence in the sequence are deleted to obtain the ΔITR.
以pscAAV-CAR-Gluc质粒为基础,用CAR-Mut启动子序列替换pscAAV载体中的CAR启动子,得到pscAAV-CAR-Mut-Gluc载体。Based on the pscAAV-CAR-Gluc plasmid, the CAR promoter in the pscAAV vector was replaced with the CAR-Mut promoter sequence to obtain the pscAAV-CAR-Mut-Gluc vector.
将生长良好的BHK-21细胞传代至24孔板,待密度达到60%时,利用Lipofectamine2000(Invitrogen,美国),根据厂商说明书,转染pscAAV-CAR-Gluc、pscAAV-CAR-MutC-Gluc、pscAAV-CAR-MutA-Gluc和pscAAV-CAR-MutG-Gluc各3孔。转染48小时后每孔取上清液100μL,用Glomax96微孔板光度计(Promega)检测Gluc水平,并使用检测仪软件进行数据分析。The well-grown BHK-21 cells were passaged to 24-well plates, and when the density reached 60%, Lipofectamine2000 (Invitrogen, USA) was used to transfect pscAAV-CAR-Gluc, pscAAV-CAR-MutC-Gluc, pscAAV according to the manufacturer's instructions - 3 wells each for CAR-MutA-Gluc and pscAAV-CAR-MutG-Gluc. 48 hours after transfection, 100 μL of the supernatant was taken from each well, the Gluc level was detected with a Glomax96 microplate luminometer (Promega), and data analysis was performed using the detector software.
如图3所示,与空白(未转染质粒的BHK-21细胞)相比,在转染质粒pscAAV-CAR-Gluc与pscAAV-CAR-Mut-Gluc后,细胞Gluc表达水平极显著的增加,且转染pscAAV-CAR-MutC-Gluc后Gluc水平相比转染pscAAV-CAR-Gluc提升29.8%。pscAAV-CAR-MutA-Gluc、pscAAV-CAR-MutG-Gluc与pscAAV-CAR-MutC-Gluc无明显差异。As shown in Figure 3, compared with the blank (BHK-21 cells without transfection plasmid), after the transfection of plasmids pscAAV-CAR-Gluc and pscAAV-CAR-Mut-Gluc, the expression level of Gluc in the cells increased significantly, And the level of Gluc after transfection with pscAAV-CAR-MutC-Gluc was increased by 29.8% compared with transfection with pscAAV-CAR-Gluc. There was no significant difference between pscAAV-CAR-MutA-Gluc, pscAAV-CAR-MutG-Gluc and pscAAV-CAR-MutC-Gluc.
结果证实,CAR-MUT启动子具有增加的启动子功能。The results confirmed that the CAR-MUT promoter has increased promoter function.
实施例3:优化3’UTR序列Example 3: Optimizing the 3'UTR sequence
在基于EGFP的表达质粒载体中,嵌入miRNA1、122、206靶序列(分别为SEQ ID NO:6,7和8),在不同组织来源的细胞系中对比miRNA靶序列对下调目的基因表达是否有预期作用。In the EGFP-based expression plasmid vector, the target sequences of miRNA1, 122, and 206 (respectively SEQ ID NO: 6, 7, and 8) were embedded, and whether the miRNA target sequences were effective in down-regulating the expression of the target gene in cell lines derived from different tissues expected effect.
SEQ ID NO:6miRNA1 target_ATACATACTTCTTTACATTCCA;SEQ ID NO: 6miRNA1 target_ATACATACTTCTTTACATTCCA;
SEQ ID NO:7miRNA206 target_CCACACACTTCCTTACATTCCA;SEQ ID NO: 7miRNA206 target_CCACACACTTCCTTACATTCCA;
SEQ ID NO:8miRNA122 target_CAAACACCATTGTCACACTCCA;SEQ ID NO:8miRNA122 target_CAAACACCATTGTCACACTCCA;
构建基于EGFP表达构建体的质粒载体。在该载体质粒中,CMV启动子驱动EGFP基因表达,3’UTR 中嵌入miRNA1靶序列(miRNA1 Target)、miRNA206靶序列(miRNA206 Target)、和miRNA122靶序列(miRNA122 Target)。作为对照,构建不嵌入miRNA靶序列的相应EGFP表达构建体。Construction of plasmid vectors based on EGFP expression constructs. In this vector plasmid, the CMV promoter drives EGFP gene expression, and the miRNA1 target sequence (miRNA1 Target), miRNA206 target sequence (miRNA206 Target), and miRNA122 target sequence (miRNA122 Target) are embedded in the 3' UTR. As a control, a corresponding EGFP expression construct was constructed without embedding the miRNA target sequence.
分别在C2C12细胞系(成肌细胞,锦篮实验室保存)和Huh7细胞系(人肝癌细胞,锦篮实验室保存)中瞬时转染质粒。转染48小时后,荧光显微镜下观察细胞荧光信号。对比位于3’UTR的不同miRNA靶序列对目的基因EGFP表达的影响。The plasmids were transiently transfected in C2C12 cell line (myoblasts, preserved by Jinlan Laboratory) and Huh7 cell line (human liver cancer cells, preserved by Jinlan Laboratory). After 48 hours of transfection, the fluorescent signal of the cells was observed under a fluorescent microscope. The effects of different miRNA target sequences located in the 3'UTR on the expression of the target gene EGFP were compared.
结果显示在图4中。如图所示,miRNA1和miRNA206靶序列的组合能显著降低在骨骼肌细胞中目的基因的表达水平。而miRNA122靶序列能够在肝细胞中下调目的基因的表达。嵌入miRNA靶序列的质粒载体在Huh7细胞中和C2C12细胞中具有显著调低的目的基因表达,这说明包含所述miRNA靶序列的3’UTR具有特异性的表达下调能力。The results are shown in Figure 4. As shown, the combination of miRNA1 and miRNA206 target sequences can significantly reduce the expression level of the target gene in skeletal muscle cells. The miRNA122 target sequence can down-regulate the expression of target genes in hepatocytes. The plasmid vector embedded with the miRNA target sequence had significantly down-regulated expression of the target gene in Huh7 cells and C2C12 cells, which indicated that the 3'UTR containing the miRNA target sequence had specific expression down-regulation ability.
实施例4:3’UTR中miRNA靶序列的组合排列优化Example 4: Combinatorial alignment optimization of miRNA target sequences in the 3'UTR
在基于实施例3的构建体中,将EGFP基因替换为Gluc报告基因,并在3’UTR中增加miRNA靶序列的重复次数和改变其排列组合方式,从而对比miRNA 1、miRNA 206、miRNA 122靶序列的不同组合方式对目的基因表达的影响。In the construct based on Example 3, the EGFP gene was replaced by the Gluc reporter gene, and the number of repeats of the miRNA target sequence was increased in the 3'UTR and its arrangement and combination were changed, thereby comparing miRNA 1, miRNA 206, and miRNA 122 targets The effect of different combinations of sequences on the expression of the target gene.
构建不同的miRNA靶序列组合:(SEQ ID NO:9、10、或11)Construction of different miRNA target sequence combinations: (SEQ ID NO: 9, 10, or 11)
组合方式1(SEQ ID NO:9):Combination method 1 (SEQ ID NO:9):
Figure PCTCN2023071616-appb-000001
Figure PCTCN2023071616-appb-000001
组合方式2(SEQ ID NO:10):Combination mode 2 (SEQ ID NO: 10):
Figure PCTCN2023071616-appb-000002
Figure PCTCN2023071616-appb-000002
组合方式3(SEQ ID NO:11):Combination mode 3 (SEQ ID NO: 11):
Figure PCTCN2023071616-appb-000003
Figure PCTCN2023071616-appb-000003
用包含构建体的表达质粒,对C2C12细胞系(成肌细胞,锦篮实验室保存)和Huh7细胞系(人肝癌细胞,锦篮实验室保存),进行瞬时转染。转染48小时后,观察细胞荧光信号。Transient transfection was performed on the C2C12 cell line (myoblast, preserved by Jinlan Laboratory) and the Huh7 cell line (human liver cancer cell, preserved by Jinlan Laboratory) with the expression plasmid containing the construct. After 48 hours of transfection, the fluorescent signal of the cells was observed.
如图5所示,通过重复与组合三种miRNA靶序列,在C2C12(骨骼肌细胞)中目的基因表达水平呈现显著地下降,下降幅度均高于单拷贝miRNA组合,其中组合方式3的下调幅度最大,并显著高于其他两种组合(p<0.05)。As shown in Figure 5, by repeating and combining the three miRNA target sequences, the expression level of the target gene in C2C12 (skeletal muscle cells) showed a significant decrease, and the decline rate was higher than that of the single-copy miRNA combination, and the down-regulation rate of the combination mode 3 The largest and significantly higher than the other two combinations (p<0.05).
如图6所示,三种miRNA靶序列的组合同样表现出更优异的目的基因下调能力,相对于单拷贝的miRNA组合的下调能力,提升了~300%的目的基因抑制能力,将报告基因在肝细胞中的表达抑制了90%以上。As shown in Figure 6, the combination of the three miRNA target sequences also showed a better ability to down-regulate the target gene. Compared with the down-regulation ability of the single-copy miRNA combination, it increased the ability to inhibit the target gene by ~300%. The expression in hepatocytes was suppressed by more than 90%.
上述结果说明,增加miRNA靶序列的重复数和优化其排列顺序,可以进一步提升miRNA的基因表达沉默效率。The above results indicated that increasing the repeat number of miRNA target sequence and optimizing its sequence can further improve the gene expression silencing efficiency of miRNA.
实施例4.重组AAV病毒的构建 Embodiment 4. Construction of recombinant AAV virus
构建包含优化启动子CAR-MutC、优化目的基因co-ABCD1以及插入3’UTR miRNA靶序列的ABCD1表达盒的AAV质粒载体。构建的AAV质粒载体包含:An AAV plasmid vector containing an optimized promoter CAR-MutC, an optimized target gene co-ABCD1, and an ABCD1 expression cassette inserted into the 3'UTR miRNA target sequence was constructed. The constructed AAV plasmid vector contains:
i)来自AAV2基因组的ITR;i) ITRs from the AAV2 genome;
ii)CAR-MutC启动子;ii) CAR-MutC promoter;
iii)co-ABCD1编码核酸序列;iii) co-ABCD1 encoding nucleic acid sequence;
iv)3’UTR,包含miRNA靶序列;iv) 3'UTR, comprising the miRNA target sequence;
Figure PCTCN2023071616-appb-000004
Figure PCTCN2023071616-appb-000004
v)牛生长激素的多聚核苷酸加尾信号BGH polyA;v) polynucleotide tailing signal BGH polyA of bovine growth hormone;
Figure PCTCN2023071616-appb-000005
Figure PCTCN2023071616-appb-000005
vi)来自AAV2基因组的ITR。vi) ITRs from the AAV2 genome.
以pRDAAV-CMV-EGFP(图7)为基础,用CAR-MutC启动子(SEQ ID No.2)替换pRDAAV载体中的Based on pRDAAV-CMV-EGFP (Figure 7), replace the CAR-MutC promoter (SEQ ID No.2) in the pRDAAV vector
CMV启动子,得到pRDAAV-CAR-Mut-EGFP载体。接下来,将人工合成的coABCD1编码核苷酸序列(SEQ ID NO:1)克隆入pRDAAV-CAR-Mut-EGFP载体中以替代EGFP,得到pRDAAV-CAR-Mut-CMV promoter, resulting in pRDAAV-CAR-Mut-EGFP vector. Next, the artificially synthesized coABCD1 coding nucleotide sequence (SEQ ID NO: 1) was cloned into the pRDAAV-CAR-Mut-EGFP vector to replace EGFP to obtain pRDAAV-CAR-Mut-
coABCD1载体。coABCD1 vector.
具体而言,由金斯瑞生物科技有限公司合成coABCD1核酸序列,并在合成序列的上游加入KpnI酶切位点与Kozak序列5’-GCCACC-3’,在下游加入taa终止密码子及EcoRI酶切位点。合成后的序列克隆入pUC57 simple载体(金斯瑞生物科技,南京),得到pUC57-coABCD1载体。KpnI和EcoRI分别双酶切 消化pUC57-coABCD1载体和pRDAAV-CAR-Mut-EGFP载体,回收coABCD1片段和去除了EGFP报告基因的pRDAAV-CAR-Mut-EGFP载体片段,两片段连接后转化E.coli DH5α感受态细胞(擎科新业,北京),筛选、鉴定后得到pRDAAV-CAR-Mut-coABCD1载体。接着,将人工合成的miRNA靶片段(包含miRNA122、206、1靶序列,序列信息见SEQ ID No.12)克隆入pRD.AAV-CAR-Mut-coABCD1载体的EcoRI和SalI酶切位点之间得到pRD.AAV-CAR-Mut-coABCD1-miT载体。Specifically, the coABCD1 nucleic acid sequence was synthesized by GenScript Biotechnology Co., Ltd., and a KpnI restriction site and a Kozak sequence 5'-GCCACC-3' were added upstream of the synthetic sequence, and a taa stop codon and EcoRI enzyme were added downstream cut site. The synthesized sequence was cloned into the pUC57 simple vector (GenScript Biotechnology, Nanjing) to obtain the pUC57-coABCD1 vector. The pUC57-coABCD1 vector and the pRDAAV-CAR-Mut-EGFP vector were digested with KpnI and EcoRI respectively, the coABCD1 fragment and the pRDAAV-CAR-Mut-EGFP vector fragment with the EGFP reporter gene removed were recovered, and the two fragments were ligated and transformed into E.coli DH5α competent cells (Qingke Xinye, Beijing) were screened and identified to obtain the pRDAAV-CAR-Mut-coABCD1 vector. Next, the artificially synthesized miRNA target fragment (comprising miRNA122, 206, 1 target sequence, sequence information see SEQ ID No.12) is cloned into between the EcoRI and SalI restriction sites of the pRD.AAV-CAR-Mut-coABCD1 vector The pRD.AAV-CAR-Mut-coABCD1-miT vector was obtained.
采用构建的AAV质粒载体,pRDAAV-CAR-Mut-coABCD1载体和pRD.AAV-CAR-Mut-coABCD1-miT载体,通过三质粒共转染法,使用HEK293包装系统,包装产生重组AAV病毒,AAV9-coABCD1和AAV9-coABCD1-miT,并检测了病毒滴度。Using the constructed AAV plasmid vector, pRDAAV-CAR-Mut-coABCD1 vector and pRD.AAV-CAR-Mut-coABCD1-miT vector, through the three-plasmid co-transfection method, using the HEK293 packaging system, the recombinant AAV virus, AAV9- coABCD1 and AAV9-coABCD1-miT, and the virus titer was detected.
实施例5:重组AAV9病毒的体外转导和功能检测Example 5: In vitro transduction and functional testing of recombinant AAV9 virus
5.1重组AAV9病毒转导对细胞ABCD1表达的影响5.1 The effect of recombinant AAV9 virus transduction on the expression of ABCD1 in cells
以感染复数MOI 10000,使用制备的重组AAV病毒,感染来自两名X-ALD患者(patient 1和Patient2)的皮肤成纤维细胞。感染后72小时用福尔马林固定细胞,进行免疫荧光染色。使用Anti-ABCD1抗体(1:100稀释,Abcam)孵育细胞过夜,漂洗,dylight549标记的二抗孵育1小时后漂洗、复染dapi、封片。荧光显微镜下观察红色荧光和蓝色荧光。Skin fibroblasts from two X-ALD patients (patient 1 and patient 2) were infected with the prepared recombinant AAV virus at a multiplicity of infection of 10000. Cells were fixed with formalin 72 hours after infection and immunofluorescent staining was performed. Cells were incubated overnight with Anti-ABCD1 antibody (diluted 1:100, Abcam), rinsed, washed with dylight549-labeled secondary antibody for 1 hour, counterstained with dapi, and mounted. Red fluorescence and blue fluorescence were observed under a fluorescence microscope.
结果显示在图8中。在感染AAV9-coABCD1的细胞爬片中可以观察到ABCD1阳性的细胞,细胞的胞浆中有强烈红色荧光信号。未接种病毒的细胞爬片中没有发现ABCD1阳性的细胞。The results are shown in Figure 8. ABCD1-positive cells can be observed in the cell slides infected with AAV9-coABCD1, and there is a strong red fluorescent signal in the cytoplasm of the cells. No ABCD1-positive cells were found in the uninoculated cell slides.
结果说明,AAV9-coABCD1体外转导X-ALD患者的成纤维细胞增加了细胞中ABCD1的表达。The results showed that AAV9-coABCD1 in vitro transduction of fibroblasts from X-ALD patients increased the expression of ABCD1 in the cells.
5.2重组AAV9病毒转导对细胞生长增殖速度的影响5.2 Effect of recombinant AAV9 virus transduction on cell growth and proliferation rate
以感染复数MOI 50000,使用制备的重组AAV病毒,感染来自X-ALD患者的皮肤成纤维细胞。感染后第7天观察细胞密度。Skin fibroblasts from X-ALD patients were infected with the prepared recombinant AAV virus at a MOI of 50000. Cell density was observed on day 7 after infection.
结果显示在图9中。感染AAV9-coABCD1的细胞(图9B)与正常人的皮肤成纤维细胞(图9C)增殖速度近似,培养7天时细胞密度大约90%,两者无显著差别。未接种AAV9-coABCD1的患者成纤维细胞(图9A)生长增殖速度较低,细胞密度<50%。The results are shown in Figure 9. The proliferation rate of cells infected with AAV9-coABCD1 (Fig. 9B) was similar to that of normal human skin fibroblasts (Fig. 9C), and the cell density was about 90% when cultured for 7 days, and there was no significant difference between the two. Fibroblasts from patients not inoculated with AAV9-coABCD1 ( FIG. 9A ) grew and proliferated at a lower rate, and the cell density was <50%.
实施例6:单次尾静脉注射AAV9-coABCD1-miT和AAV9-coABCD1干预C57BL6/J野生型小鼠Example 6: A single tail vein injection of AAV9-coABCD1-miT and AAV9-coABCD1 interferes with C57BL6/J wild-type mice
本实施例将包含优化前后的ABCD1表达构建体的腺相关病毒载体,尾静脉注射到C57BL6J野生型小鼠,剂量为1.5E+14vg/kg,每组4只小鼠。除未优化组的一只小鼠于给药后3周死亡外,剩余的7只小鼠在注射后4周,剖杀取材,检测组织病理和极长链脂肪酸含量。3’UTR优化前后体内组织病理改变情况以 及极长链脂肪酸的改变情况显示在图10和11中。In this example, the adeno-associated virus vector containing the ABCD1 expression construct before and after optimization was injected into the tail vein of C57BL6J wild-type mice at a dose of 1.5E+14vg/kg, with 4 mice in each group. Except for one mouse in the unoptimized group that died 3 weeks after administration, the remaining 7 mice were killed 4 weeks after injection, and samples were collected for histopathology and the content of very long chain fatty acids. The histopathological changes in vivo and the changes of very long chain fatty acids before and after 3'UTR optimization are shown in Figures 10 and 11.
极长链脂肪酸含量检测结果表明,对于正常小鼠,在注射进行干预后,优先前后的ABCD1表达构建体均可以降低正常小鼠体内各组织和血液中的极长链脂肪酸水平(图10),说明病毒递送的ABCD1发挥了其生理功能作用。The detection results of the very long-chain fatty acid content showed that, for normal mice, after intervention by injection, the ABCD1 expression construct before and after priority could reduce the level of very long-chain fatty acids in various tissues and blood of normal mice (Figure 10), It shows that the ABCD1 delivered by the virus plays its physiological function.
对给药后第4周剩余的7只存活实验小鼠进行了组织病理学检测。结果表明,未优化试验组的小鼠在心肌和肝脏中均出现了比较明显的病理改变,以心肌细胞空泡样变性、单个肝细胞坏死、单核细胞浸润和广泛肝细胞水肿为主要病理改变。优化后试验组的小鼠未发现上述病理改变。(图11A)Histopathological examination was carried out on the remaining 7 surviving experimental mice in the 4th week after administration. The results showed that the mice in the unoptimized test group had obvious pathological changes in the myocardium and liver, with vacuolar degeneration of cardiomyocytes, necrosis of a single hepatocyte, mononuclear cell infiltration and extensive hepatocyte edema as the main pathological changes . The above-mentioned pathological changes were not found in the mice in the experimental group after optimization. (Figure 11A)
对未优化组于给药后3周死亡的小鼠,进行尸检。组织病理学诊断发现,该例死亡的小鼠心脏出现广泛的空泡样变性,心肌纤维化明显,心腔内存在血栓。肝脏病理检测,观察到大面积肝细胞坏死,主要以中央静脉为中心向外扩散。上述病理改变认为与供试品的毒性相关。(图11B)Necropsy was performed on mice in the non-optimized group that died 3 weeks after administration. Histopathological diagnosis revealed that the heart of the dead mouse had extensive vacuolar degeneration, obvious myocardial fibrosis, and thrombus in the cardiac cavity. Liver pathological examination revealed a large area of hepatocyte necrosis, which spread outwards mainly centered on the central vein. The above pathological changes are considered to be related to the toxicity of the test product. (Figure 11B)
病理结果表明,经优化后的AAV9-coABCD1-miT能够显著改善ABCD1基因表达带来的毒性。The pathological results showed that the optimized AAV9-coABCD1-miT could significantly improve the toxicity caused by ABCD1 gene expression.
实施例7:AAV9-coABCD1-miT尾静脉注射治疗X-ALD模型小鼠Example 7: AAV9-coABCD1-miT Tail Vein Injection Treats X-ALD Model Mice
本实施例选择了一种敲除了ABCD1基因的B6.129小鼠模型,该模型小鼠购自Jackson Lab。由于小鼠的种属差异,ABCD1缺失不在该模型小鼠中造成严重的生理或行为影响,但模型小鼠在10周即出现了运动能力下降等表型,且在小鼠的成纤维细胞和各组织(肾上腺、脑等)以及血液中C26:0含量水平显著高于野生型小鼠,表明模型小鼠体内极长链脂肪酸有病理性累积。这一过程模拟了X-ALD病人的病理进程。In this example, a B6.129 mouse model in which the ABCD1 gene was knocked out was selected, and the model mouse was purchased from Jackson Lab. Due to the species differences of the mice, the deletion of ABCD1 did not cause serious physiological or behavioral effects in the model mice, but the model mice showed phenotypes such as decreased exercise ability at 10 weeks, and the fibroblasts and The levels of C26:0 in various tissues (adrenal gland, brain, etc.) and blood were significantly higher than those in wild-type mice, indicating that there is pathological accumulation of very long-chain fatty acids in the model mice. This process simulates the pathological process of X-ALD patients.
针对这一模型,以AAV9-coABCD1-miT通过静脉注射的递送方式,进行模型小鼠的修复性治疗,且与正常野生型小鼠和未接受治疗的小鼠进行对照。简言之:For this model, AAV9-coABCD1-miT was delivered by intravenous injection, and the model mice were treated reparatively, and compared with normal wild-type mice and untreated mice. In short:
为确定优化后药物AAV9-coABCD1-miT的治疗效果,将优化后的药物通过静脉注射的方式施用到8只同龄X-ALD模型小鼠体内,给药剂量5E+13vg/kg。同龄的野生型小鼠组和X-ALD模型小鼠组,各8只,不做干预。In order to determine the therapeutic effect of the optimized drug AAV9-coABCD1-miT, the optimized drug was administered intravenously to 8 X-ALD model mice of the same age at a dose of 5E+13vg/kg. There were 8 wild-type mice and X-ALD model mice of the same age without any intervention.
在给药后8周,进行组织病理免疫荧光检测、极长链脂肪酸含量评估和运动评分。Eight weeks after administration, histopathological immunofluorescence detection, evaluation of very long chain fatty acid content and motor score were performed.
运动评估exercise assessment
小鼠的运动评估采用抓绳法。检测结果由图12可见,正常野生型小鼠的平均分是4.5分;未治疗的X-ALD模型小鼠的平均分为0分,被放到绳上的小鼠在10s内就掉下,表明患病小鼠在自然病史的发展下,肌肉会萎缩并变得无力;经过治疗的患病小鼠的平均分是3.7分,这些小鼠可抓住绳30s以上,且有的会试图爬上绳,有的能把前爪和一只后爪放上绳,相对于未治疗的患病小鼠来说,治疗效果是非常明显的, 并且治疗组的数据结果与野生型无显著性差异。这些结果表明,通过静脉给药,优化后药物具有明显优秀的治疗效果。The locomotion of the mice was assessed using the grab rope method. The test results can be seen from Figure 12, the average score of normal wild-type mice is 4.5 points; the average score of untreated X-ALD model mice is 0 points, and the mice that were put on the rope fell within 10s. It shows that under the development of the natural history of the diseased mice, the muscles will atrophy and become weak; the average score of the treated diseased mice is 3.7 points, these mice can grasp the rope for more than 30s, and some will try to climb On the rope, some can put the front paw and one hind paw on the rope. Compared with the untreated diseased mice, the treatment effect is very obvious, and the data results of the treatment group are not significantly different from those of the wild type . These results indicate that the optimized drug has a significantly superior therapeutic effect through intravenous administration.
极长链脂肪酸含量评估Assessment of Very Long Chain Fatty Acid Content
给药后8周,检测AAV9-coABCD1-miT药物治疗的模型动物在各个组织的极长链脂肪酸含量,并与正常健康小鼠和X-ALD模型小鼠作对比。Eight weeks after administration, the very long chain fatty acid content in various tissues of the AAV9-coABCD1-miT drug-treated model animals was detected, and compared with normal healthy mice and X-ALD model mice.
利用HPLC-MS/MS法测定极长链脂肪酸含量。首先对实验小鼠进行解剖、称重,接着将各个组织通过液氮研磨的方式研磨成粉,使用Matyash提取方法经过一系列粗提和精提将极长链脂肪酸从组织中提取出来,并利用HPLC-MS/MS法测定极长链脂肪酸含量,检测结果显示在图13中。The content of very long chain fatty acids was determined by HPLC-MS/MS. First, the experimental mice were dissected and weighed, and then each tissue was ground into powder by liquid nitrogen grinding, and the very long-chain fatty acids were extracted from the tissue through a series of rough extraction and fine extraction using the Matyash extraction method, and used The content of very long chain fatty acids was determined by HPLC-MS/MS, and the detection results are shown in Figure 13.
从图中可以看到,相对于未治疗的模型小鼠,给药小鼠体内各组织中极长链脂肪酸水平明显降低,且接近于野生型水平,尤其是受累部位的对比明显,如肾上腺、小脑。此结果表明,AAV9-coABCD1-miT药物治疗可以实现有效且优异的治疗效果。It can be seen from the figure that compared with untreated model mice, the levels of very long-chain fatty acids in various tissues in the treated mice were significantly reduced, and were close to the wild-type levels, especially the contrast of affected parts was obvious, such as the adrenal gland, cerebellum. This result indicates that AAV9-coABCD1-miT drug therapy can achieve effective and excellent therapeutic effect.
肾上腺组织病理学检测Adrenal histopathological examination
AAV9-coABCD1-miT尾静脉注射治疗X-ALD模型小鼠,给药后8周,在组织切片上免疫荧光标记ABCD1。X-ALD model mice were treated by tail vein injection of AAV9-coABCD1-miT, and ABCD1 was immunofluorescently labeled on tissue sections 8 weeks after administration.
肾上腺组织病理学检测结果显示在图14中。结果显示,未治疗组没有检测到ABCD1基因表达。治疗组的肾上腺细胞中广泛表达ABCD1,在胞浆内呈现红色斑点信号。The results of histopathological examination of the adrenal glands are shown in Figure 14. The results showed that no ABCD1 gene expression was detected in the untreated group. ABCD1 was widely expressed in the adrenal cells of the treatment group, showing red spot signal in the cytoplasm.
评估神经系统的自噬和改善情况,评价基因治疗后的疗效Assess the autophagy and improvement of the nervous system, and evaluate the efficacy after gene therapy
X-ALD由于极长链脂肪酸的累积,会激活自噬。通过自噬,将累积在各个亚细胞器中的极长链脂肪酸包裹并输送至溶酶体。但是极长链脂肪酸必须在过氧化物酶体中降解,因此自噬并不能改善极长链脂肪酸的累积。而极长链脂肪酸的累积会不断的刺激和激活自噬,因此,自噬流是淤塞的且无效的。在ABCD1缺陷型小鼠中,已经显示,异常吞噬作用损伤神经元投射,与脊髓轴索病变相关(Microglial dysfunction as a key pathological change in adrenomyeloneuropathy,doi:10.1002/ana.25085)。通过评价自噬现象的变化,可以间接评估细胞内极长链脂肪酸所引发的损伤是否有改善。X-ALD activates autophagy due to the accumulation of very long-chain fatty acids. Autophagy wraps and transports very long-chain fatty acids accumulated in various subcellular organelles to lysosomes. But very long-chain fatty acids must be degraded in peroxisomes, so autophagy does not improve the accumulation of very long-chain fatty acids. The accumulation of very long-chain fatty acids continuously stimulates and activates autophagy, and thus, the autophagic flux is stagnant and ineffective. In ABCD1-deficient mice, it has been shown that aberrant phagocytosis impairs neuronal projections and is associated with spinal cord axonopathy (Microglial dysfunction as a key pathological change in adrenomyeloneuropathy, doi:10.1002/ana.25085). By evaluating changes in autophagy, it is possible to indirectly assess whether the damage caused by intracellular very long-chain fatty acids has improved.
AAV9-coABCD1-miT尾静脉注射治疗X-ALD模型小鼠,给药后8周,脊髓组织切片免疫荧光标记LC3β(轴丝动力蛋白(axonemal dynein)轻链2β)。结果显示在图15中。从图所示结果可见:未治疗组中,脊髓前角、背角均呈现髓鞘强烈的自噬蛋白信号,表明自噬激活明显。治疗组中脊髓前角的自噬信号呈明显 下降。背角白质区的髓鞘中,也可观察到自噬信号下降的趋势,虽然下降程度不及脊髓前脚区(这是由于AAV9经静脉注射后更偏前角细胞嗜性),但是能明显比较出经治疗后脊髓中的自噬活跃程度在下降。说明治疗后能够改善自噬通路障碍的问题。X-ALD model mice were treated with AAV9-coABCD1-miT tail vein injection. Eight weeks after administration, the spinal cord tissue sections were immunofluorescently labeled with LC3β (axonemal dynein light chain 2β). The results are shown in Figure 15. From the results shown in the figure, it can be seen that in the untreated group, the anterior and dorsal horns of the spinal cord showed strong autophagy protein signals of myelin sheath, indicating that autophagy activation was obvious. The autophagy signal in the anterior horn of the spinal cord decreased significantly in the treatment group. In the myelin sheath of the dorsal horn white matter area, the trend of decreasing autophagy signal can also be observed, although the decrease degree is not as good as that in the forefoot area of the spinal cord (this is because AAV9 is more tropistic to the anterior horn cells after intravenous injection), but it can be clearly compared The autophagy activity in the spinal cord decreased after treatment. It shows that after treatment, the problem of autophagy pathway disorder can be improved.
实施例8:药物的安全性及优化程度评估Embodiment 8: Evaluation of the safety and optimization degree of drugs
利用Western Blot法测定ABCD1蛋白含量,以探究药物的安全性及优化程度。The protein content of ABCD1 was determined by Western Blot method to explore the safety and optimization degree of the drug.
将优化后(AAV9-coABCD1-miRT)与未优化的药物(AAV9-coABCD1)静脉注射进正常野生型小鼠体内,并与未给药的正常小鼠作比较。The optimized (AAV9-coABCD1-miRT) and unoptimized drug (AAV9-coABCD1) were intravenously injected into normal wild-type mice, and compared with untreated normal mice.
Western Blot检测结果显示在图16A中。利用软件ImageJ将图片进行灰度值分析,得到的量化结果显示在图16B中。从图示结果可见,注射了药物AAV9-coABCD1的两只小鼠,心肌组织中的ABCD1蛋白含量是相对较高的;而注射了药物AAV9-coABCD1-miRT的两只小鼠,心肌组织中的ABCD1蛋白含量相对较低,但仍较高于正常小鼠心肌组织中的ABCD1蛋白水平。这三种小鼠的平均灰度值比较为:C57/AAV9-hABCD1=1.58>C57/AAV9-coABCD1-miRT=0.60>C57/WT=0.15。The results of Western Blot detection are shown in Figure 16A. Using the software ImageJ to analyze the gray value of the picture, the quantification results obtained are shown in Figure 16B. From the results shown in the figure, it can be seen that the two mice injected with the drug AAV9-coABCD1 had relatively high levels of ABCD1 protein in the myocardial tissue; while the two mice injected with the drug AAV9-coABCD1-miRT had a relatively high level of ABCD1 protein in the myocardial tissue. The ABCD1 protein content was relatively low, but still higher than that in normal mouse myocardial tissue. The comparison of the average gray value of these three kinds of mice is: C57/AAV9-hABCD1=1.58>C57/AAV9-coABCD1-miRT=0.60>C57/WT=0.15.
结果表明,优化后的药物AAV9-coABCD1-miRT在一定程度上降低了药物在外周组织器官,主要是心肌组织,的毒性。The results showed that the optimized drug AAV9-coABCD1-miRT reduced the toxicity of the drug in peripheral tissues and organs, mainly myocardial tissue, to a certain extent.
Figure PCTCN2023071616-appb-000006
Figure PCTCN2023071616-appb-000006
Figure PCTCN2023071616-appb-000007
Figure PCTCN2023071616-appb-000007
Figure PCTCN2023071616-appb-000008
Figure PCTCN2023071616-appb-000008
Figure PCTCN2023071616-appb-000009
Figure PCTCN2023071616-appb-000009

Claims (24)

  1. 一种编码ABCD1的核酸,其包含:SEQ ID No:1,或与之具有至少90%、95%、96%、97%、98%、99%、99.5%同一性的核苷酸序列。A nucleic acid encoding ABCD1 comprising: SEQ ID No: 1, or a nucleotide sequence having at least 90%, 95%, 96%, 97%, 98%, 99%, 99.5% identity thereto.
  2. 权利要求1的核酸,其中,所述核酸,相比天然人ABCD1编码核酸,在组成型启动子控制下,其表达水平提升至少200%或更高,例如300%或400%。The nucleic acid of claim 1, wherein the expression level of said nucleic acid under the control of a constitutive promoter is increased by at least 200% or more, such as 300% or 400%, compared to the natural human ABCD1 encoding nucleic acid.
  3. 一种表达构建体,其包含权利要求1或2的ABCD1编码核酸。An expression construct comprising the ABCD1 encoding nucleic acid of claim 1 or 2.
  4. 权利要求3的表达构建体,其还包含与所述核酸可操作性连接的启动子,The expression construct of claim 3, which also comprises a promoter operably linked to the nucleic acid,
    优选所述启动子包含选自SEQ ID No:2-5的核苷酸序列,尤其是SEQ ID No:2的核苷酸序列。Preferably said promoter comprises a nucleotide sequence selected from SEQ ID No:2-5, especially the nucleotide sequence of SEQ ID No:2.
  5. 权利要求3或4的表达构建体,其还包含与所述ABCD1编码核酸可操作连接的3’UTR。The expression construct of claim 3 or 4, which further comprises a 3'UTR operably linked to said ABCD1-encoding nucleic acid.
  6. 权利要求5的表达构建体,其中,所述3’UTR包含1-8个拷贝(例如,2,3,或4个拷贝)的肌细胞特异性miRNA的靶序列和/或1-8个拷贝(例如,1个或2个拷贝)的肝细胞特异性miRNA的靶序列,The expression construct of claim 5, wherein the 3'UTR comprises 1-8 copies (for example, 2, 3, or 4 copies) of the target sequence of the myocyte-specific miRNA and/or 1-8 copies (e.g., 1 or 2 copies) of the target sequence of a hepatocyte-specific miRNA,
    优选地,所述3’UTR包含3个拷贝的肌细胞特异性miRNA靶序列和2个拷贝的肝细胞特异性miRNA靶序列,其中优选地,肌细胞特异性miRNA靶序列与肝细胞特异性miRNA靶序列间隔排列;Preferably, the 3'UTR comprises 3 copies of the muscle cell-specific miRNA target sequence and 2 copies of the liver cell-specific miRNA target sequence, wherein preferably, the muscle cell-specific miRNA target sequence and the liver cell-specific miRNA Target sequences spaced;
    更优选地,所述肌细胞特异性miRNA靶序列选自miRNA1靶序列、miRNA206靶序列及其组合;所述肝细胞特异性miRNA靶细胞为miRNA122靶序列;More preferably, the muscle cell-specific miRNA target sequence is selected from miRNA1 target sequence, miRNA206 target sequence and combinations thereof; the liver cell-specific miRNA target cell is miRNA122 target sequence;
    再优选地,所述3’UTR包含如下排列的miRNA靶序列:More preferably, the 3'UTR comprises miRNA target sequences arranged as follows:
    miRNA1 target-miRNA122 target-miRNA1 target-miRNA122 target-miRNA206 target;或miRNA1 target-miRNA122 target-miRNA1 target-miRNA122 target-miRNA206 target; or
    miRNA206 target-miRNA122 target-miRNA1 target-miRNA122 target-miRNA1 target。miRNA206 target-miRNA122 target-miRNA1 target-miRNA122 target-miRNA1 target.
  7. 权利要求6的表达构建体,其中,所述3’UTR包含SEQ ID NO:12的核苷酸序列。The expression construct of claim 6, wherein said 3'UTR comprises the nucleotide sequence of SEQ ID NO:12.
  8. 一种载体,其包含权利要求1-7任一项的表达构建体。A vector comprising the expression construct of any one of claims 1-7.
  9. 权利要求8的载体,其中所述载体是病毒载体。The vector of claim 8, wherein said vector is a viral vector.
  10. 权利要求8或9的载体,其中所述载体是腺相关病毒(AAV)载体。8. The vector of claim 8 or 9, wherein said vector is an adeno-associated viral (AAV) vector.
  11. 包含权利要求1-7任一项的表达构建体或权利要求8-10任一项的载体的宿主细胞,优选哺乳动物细胞。A host cell, preferably a mammalian cell, comprising an expression construct according to any one of claims 1-7 or a vector according to any one of claims 8-10.
  12. 一种重组腺相关病毒(AAV)载体,其中所述重组AAV载体在其基因组中包含位于5’和3’AAV反向末端重复序列之间的根据权利要求1-7任一项的表达构建体,A recombinant adeno-associated virus (AAV) vector, wherein said recombinant AAV vector comprises in its genome an expression construct according to any one of claims 1-7 between the 5' and 3' AAV inverted terminal repeats ,
    优选地,其中所述重组AAV载体在其基因组中包含:Preferably, wherein said recombinant AAV vector comprises in its genome:
    a.5’和3’AAV反向末端重复(ITR)序列,和a. 5' and 3' AAV inverted terminal repeat (ITR) sequences, and
    b.位于5’和3’ITR之间的表达构建体,其中所述表达构建体包含以转录方向彼此功能性连接的如下 元件:b. An expression construct positioned between the 5' and 3' ITR, wherein the expression construct comprises the following elements functionally linked to each other in the direction of transcription:
    -启动子,-Promoter,
    -编码人ABCD1的多核苷酸,- a polynucleotide encoding human ABCD1,
    -至少一个miRNA靶序列,- at least one miRNA target sequence,
    -一个或多个终止子;和- one or more terminators; and
    -一个或多个polyA信号序列,优选地选自SV40晚期polyA序列、兔β-珠蛋白polyA序列、和牛生长激素polyA序列,更优选地牛生长激素polyA序列,例如SEQ ID NO:13的核苷酸序列。- one or more polyA signal sequences, preferably selected from the group consisting of SV40 late polyA sequence, rabbit β-globin polyA sequence, and bovine growth hormone polyA sequence, more preferably bovine growth hormone polyA sequence, such as the nucleosides of SEQ ID NO: 13 acid sequence.
  13. 权利要求12的重组AAV病毒载体,其中所述ITR为野生型AAV2 ITR序列,或所述ITR之一是缺少功能性末端解链位点(trs)和任选地D序列的AA2 ΔITR序列。The recombinant AAV viral vector of claim 12, wherein said ITR is a wild-type AAV2 ITR sequence, or one of said ITRs is an AA2 ΔITR sequence lacking a functional terminal melting site (trs) and optionally a D sequence.
  14. 权利要求12-13任一项的重组AAV病毒载体,其中所述载体为ssAAV载体或scAAV载体,尤其是ssAAV载体。The recombinant AAV viral vector according to any one of claims 12-13, wherein said vector is a ssAAV vector or a scAAV vector, especially an ssAAV vector.
  15. 权利要求12-14任一项的重组AAV病毒载体,其中所述重组AAV载体包含来自AAV1,AAV2,AAV3,AAV4,AAV5,AAV6,AAV7,AAV8,AAV9血清型,尤其是AAV9血清型的衣壳蛋白,优选地,所述重组AAV载体是AAV2/9载体。The recombinant AAV viral vector according to any one of claims 12-14, wherein said recombinant AAV vector comprises a capsid from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9 serotype, especially AAV9 serotype protein, preferably, the recombinant AAV vector is an AAV2/9 vector.
  16. 一种药物组合物,其包含权利要求8-10的载体或权利要求12-15任一项的重组AAV病毒载体和药物可接受载体,优选地,所述药物组合物用于腹膜内、肌内、动脉内、静脉内、鞘内或脑室内给药,更优选地静脉内给药。A pharmaceutical composition comprising the carrier of claims 8-10 or the recombinant AAV virus vector of any one of claims 12-15 and a pharmaceutically acceptable carrier, preferably, the pharmaceutical composition is used for intraperitoneal, intramuscular , intraarterial, intravenous, intrathecal or intracerebroventricular administration, more preferably intravenous administration.
  17. 根据权利要求1-7任一项的表达构建体或根据权利要求8-15任一项的载体用于在哺乳动物细胞、细胞系或细胞群中表达ABCD1的用途,或在制备用于在哺乳动物细胞、细胞系或细胞群中表达ABCD1的药物中的应用。Use of the expression construct according to any one of claims 1-7 or the vector according to any one of claims 8-15 for expressing ABCD1 in mammalian cells, cell lines or cell groups, or in the preparation for use in breast-feeding Use in pharmaceuticals expressing ABCD1 in animal cells, cell lines or cell groups.
  18. 一种治疗或预防X染色体连锁肾上腺脑白质营养不良(X-ALD)和/或改善与X-ALD相关的症状的方法,其中所述方法包括向有需要的受试者施用权利要求12-15任一项的重组AAV病毒载体或权利要求16的药物组合物,A method for treating or preventing X-chromosome-linked adrenoleukodystrophy (X-ALD) and/or improving symptoms associated with X-ALD, wherein the method comprises administering claims 12-15 to a subject in need thereof any one of the recombinant AAV viral vectors or the pharmaceutical composition of claim 16,
    优选地,所述方法包括腹膜内、肌内、动脉内、静脉内、鞘内或脑室内给药,更优选地静脉内注射,所述的重组AAV病毒载体。Preferably, said method comprises intraperitoneal, intramuscular, intraarterial, intravenous, intrathecal or intraventricular administration, more preferably intravenous injection, of said recombinant AAV viral vector.
  19. 权利要求18的方法,其中所述重组AAV病毒载体是AAV9。18. The method of claim 18, wherein said recombinant AAV viral vector is AAV9.
  20. 权利要求18或19的方法,其中所述方法降低受试者的血浆和组织,尤其是肾上腺和小脑中,极长链脂肪酸VLCFA的含量。18. The method of claim 18 or 19, wherein said method reduces the level of very long chain fatty acids (VLCFA) in the subject's plasma and tissues, especially the adrenal gland and cerebellum.
  21. 权利要求18-20任一项的方法,其中所述方法降低脊髓中由VLCFA积累刺激的自噬活性。The method of any one of claims 18-20, wherein the method reduces autophagic activity stimulated by VLCFA accumulation in the spinal cord.
  22. 一种在哺乳动物细胞中表达ABCD1的方法(例如体外、体内、或离体方法),所述方法包括用根据权利要求1-7任一项的表达构建体或根据权利要求8-10任一项的载体转染分离的哺乳动物细胞、或细胞系或细胞群。A method (such as in vitro, in vivo, or ex vivo method) of expressing ABCD1 in mammalian cells, said method comprising using the expression construct according to any one of claims 1-7 or any one according to claims 8-10 Isolated mammalian cells, or cell lines or populations of cells are transfected with the vector of the present invention.
  23. 一种表达构建体,其包含与基因可操作性连接的3’UTR,其中所述3’UTR包含选自如下的串联miRNA靶序列:An expression construct comprising a 3'UTR operably linked to a gene, wherein the 3'UTR comprises a tandem miRNA target sequence selected from the group consisting of:
    -miRNA1 target x2-miRNA206 target x1-miRNA122 target x2;例如SEQ ID NO:9所示的序列;-miRNA1 target x2-miRNA206 target x1-miRNA122 target x2; for example the sequence shown in SEQ ID NO:9;
    -miRNA1 target x2-miRNA122 target x2-miRNA206 target x1;例如,SEQ ID NO:10所示的序列;或-miRNA1 target x2-miRNA122 target x2-miRNA206 target x1; for example, the sequence shown in SEQ ID NO: 10; or
    -miRNA1 target x1-miRNA122 target x1-miRNA1 target x1-miRNA122 target x1-miRNA206 target x1;例如SEQ ID NO:11所示的序列。-miRNA1 target x1-miRNA122 target x1-miRNA1 target x1-miRNA122 target x1-miRNA206 target x1; for example the sequence shown in SEQ ID NO:11.
  24. 权利要求23的表达构建体用于选择性地降低基因在肌细胞和肝细胞中的表达的用途。23. Use of the expression construct of claim 23 for selectively reducing gene expression in muscle cells and hepatocytes.
PCT/CN2023/071616 2022-01-10 2023-01-10 Gene treatment drug and method for x-linked adrenoleukodystrophy WO2023131345A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210020181.9A CN116445492A (en) 2022-01-10 2022-01-10 Gene therapy medicine and method for X chromosome linked adrenoleukodystrophy
CN202210020181.9 2022-01-10

Publications (1)

Publication Number Publication Date
WO2023131345A1 true WO2023131345A1 (en) 2023-07-13

Family

ID=87073312

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/071616 WO2023131345A1 (en) 2022-01-10 2023-01-10 Gene treatment drug and method for x-linked adrenoleukodystrophy

Country Status (2)

Country Link
CN (1) CN116445492A (en)
WO (1) WO2023131345A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108456697A (en) * 2018-03-29 2018-08-28 成都优娃生物科技有限公司 Using slow virus carrier EF1 α promoter Optimal Expression ABCD1 gene therapy adrenoleukodystrophy diseases
WO2021168405A1 (en) * 2020-02-21 2021-08-26 Combined Therapeutics, Inc. Compositions and methods for organ-protective expression and modulation of coding ribonucleic acids

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108456697A (en) * 2018-03-29 2018-08-28 成都优娃生物科技有限公司 Using slow virus carrier EF1 α promoter Optimal Expression ABCD1 gene therapy adrenoleukodystrophy diseases
WO2021168405A1 (en) * 2020-02-21 2021-08-26 Combined Therapeutics, Inc. Compositions and methods for organ-protective expression and modulation of coding ribonucleic acids

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE Nucleotide 14 July 2016 (2016-07-14), ANONYMOUS : "Synthetic construct Homo sapiens clone IMAGE:100008637; FLH168397.01L; RZPDo839G0591D ATP-binding cassette, sub-family D (ALD), member 1 (ABCD1) gene, encodes complete protein", XP093076546, retrieved from NCBI Database accession no. DQ894177.2 *
GONG, YI ET AL.: "Adenoassociated Virus Serotype 9-Mediated Gene Therapy for X-linked Adrenoleukodystrophy", MOL THER., vol. 23, no. 5, 31 May 2015 (2015-05-31), XP055381024, DOI: 10.1038/mt.2015.6 *
PILLAY SIRIKA, MEYER NANCY L, PUSCHNIK ANDREAS S, DAVULCO OMAR, DIEP JONATHAN, ISHIKAWA YOSHI, JAE LUCAS T, WOSEN JONATHAN E, NAGA: "295. Intrathecal Delivery of rAAV9-ABCD1 by Osmotic Pump in a Mouse Model of Adrenomyeloneuropathy", MOLECULAR THERAPY, vol. 24, no. Suppl. 1, 1 May 2016 (2016-05-01), pages S118 - S119, XP093076549 *

Also Published As

Publication number Publication date
CN116445492A (en) 2023-07-18

Similar Documents

Publication Publication Date Title
AU2019200949B2 (en) High-transduction-efficiency raav vectors, compositions, and methods of use
JP7389828B2 (en) Gene therapy for lysosomal diseases
TWI654301B (en) Modified Friedrich Reich (FRIEDREICH) dysmotility gene and vector for gene therapy
EP3113787B1 (en) Improved raav vectors and methods for transduction of photoreceptors and rpe cells
JP2022528416A (en) Recombinant adeno-associated virus and its use
US20190284244A1 (en) High-transduction-efficiency raav vectors, compositions, and methods of use
JP2020054367A (en) Adeno-associated virus vectors encoding modified g6pc and uses thereof
JP2022530824A (en) Composition useful for the treatment of Pompe disease
US20150045416A1 (en) Methods and Compositions for Gene Delivery
CN115029360A (en) Transgenic expression cassette for treating mucopolysaccharidosis type IIIA
WO2019091457A1 (en) Liver-specific transcriptional regulatory sequence and applications thereof
CN111601620A (en) Adeno-associated virus gene therapy for 21-hydroxylase deficiency
WO2023131345A1 (en) Gene treatment drug and method for x-linked adrenoleukodystrophy
WO2022198872A1 (en) Aav-mediated human lipoprotein lipase liver ectopic expression vector, and use thereof
JP2023545384A (en) Recombinant adeno-associated virus for central nervous system or muscle delivery
JP2023515442A (en) Muscle-specific nucleic acid regulatory elements and methods and uses thereof
CN115772520B (en) Gene therapy constructs, pharmaceutical compositions and methods for treating Pompe disease
WO2023184688A1 (en) FUNCTIONAL β-GALACTOSIDASE VARIANT, AAV-MEDIATED HUMAN β-GALACTOSIDASE EXPRESSION VECTOR AND USE THEREOF
CN115820740A (en) Recombinant adeno-associated virus vector for treating type II mucopolysaccharidosis and application thereof
WO2024079667A1 (en) Nucleic acid regulatory elements for gene expression in the central nervous system and methods of use
WO2023147304A1 (en) Aav capsids for improved heart transduction and detargeting of liver
JP2023522852A (en) Adeno-associated virus compositions for IDS gene transfer and methods of use thereof
CN115838725A (en) Promoter sequence of specific promoter in mammal heart and application thereof
Zentilin et al. 979. Chronic Expression of AZGP1 Following AAVrh. 10-Mediated Gene Transfer Limits Weight Gain in Mice on High Fat Diets

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23737207

Country of ref document: EP

Kind code of ref document: A1