WO2024080304A1 - Fusion protein of serum albumin and physiologically active protein - Google Patents

Fusion protein of serum albumin and physiologically active protein Download PDF

Info

Publication number
WO2024080304A1
WO2024080304A1 PCT/JP2023/036884 JP2023036884W WO2024080304A1 WO 2024080304 A1 WO2024080304 A1 WO 2024080304A1 JP 2023036884 W JP2023036884 W JP 2023036884W WO 2024080304 A1 WO2024080304 A1 WO 2024080304A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
acid sequence
fusion protein
hsa
seq
Prior art date
Application number
PCT/JP2023/036884
Other languages
French (fr)
Japanese (ja)
Inventor
健一 高橋
泰功 菅野
貴士 小野内
Original Assignee
Jcrファーマ株式会社
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 Jcrファーマ株式会社 filed Critical Jcrファーマ株式会社
Publication of WO2024080304A1 publication Critical patent/WO2024080304A1/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
    • C07K14/76Albumins
    • C07K14/765Serum albumin, e.g. HSA
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/24Hydrolases (3) acting on glycosyl compounds (3.2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione

Definitions

  • the present invention relates to a fusion protein in which serum albumin (SA) is bound to a protein having physiological activity (biologically active protein) and a method for producing the same.
  • SA serum albumin
  • biologically active protein a protein having physiological activity
  • Such a fusion protein relates, for example, to one in which the C-terminus of SA is bound to the N-terminus of a biologically active protein.
  • Some biologically active proteins have low expression levels and/or low activity when expressed as a recombinant protein by introducing a gene encoding the biologically active protein into a host cell such as a mammalian cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in the culture medium.
  • the present invention relates to such a fusion protein of SA and a biologically active protein that can be efficiently produced as a highly active recombinant protein, and also relates to a method for producing the fusion protein.
  • a biologically active protein to be fused to SA, and any biologically active protein can be made into a fusion protein with SA.
  • the present invention relates in particular to a fusion protein in which serum albumin (SA) is bound to a lysosomal enzyme, for example, the C-terminus of SA is bound to the N-terminus of a lysosomal enzyme, or the C-terminus of a lysosomal enzyme is bound to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • Some lysosomal enzymes have low expression levels and/or low activity when expressed as recombinant proteins by introducing a gene encoding the lysosomal enzyme into a host cell such as a mammalian cell, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium.
  • the present invention relates to such a fusion protein of a lysosomal enzyme and SA that can be efficiently produced as a highly active recombinant protein, and also relates to a method for producing the fusion protein.
  • a physiologically active protein to be fused to the lysosomal enzyme, and any lysosomal enzyme can be made into a fusion protein with SA.
  • the present invention also relates to a fusion protein in which serum albumin (SA) and galactosylceramidase (GALC) are bound, for example, by binding the C-terminus of SA to the N-terminus of GALC, or the C-terminus of GALC to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • GALC galactosylceramidase
  • a gene encoding GALC is introduced into a host cell such as a mammalian cell and the protein is expressed as a recombinant protein, the expression level and/or activity of GALC may be low, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium.
  • the present invention relates to such a fusion protein of GALC and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing the fusion protein.
  • the present invention also relates to a fusion protein in which serum albumin (SA) and glucocerebrosidase (GBA) are bound, for example, by binding the C-terminus of SA to the N-terminus of GBA, or the C-terminus of GBA to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • GBA glucocerebrosidase
  • the present invention relates to a fusion protein in which serum albumin (SA) and a cytokine are bound, for example, by binding the C-terminus of SA to the N-terminus of a cytokine, or the C-terminus of a cytokine to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • Some cytokines have low expression levels and/or low activity when expressed as a recombinant protein by introducing a gene encoding the cytokine into a host cell such as a mammalian cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in the culture medium.
  • the present invention relates to such a fusion protein of cytokine and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing the fusion protein.
  • physiologically active protein to be fused with the cytokine, and any cytokine can be made into a fusion protein with SA.
  • the present invention relates in particular to a fusion protein in which serum albumin (SA) and an interleukin are bound, for example, by binding the C-terminus of SA to the N-terminus of an interleukin, or the C-terminus of an interleukin to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • Some interleukins have low expression levels and/or low activity when expressed as a recombinant protein by introducing a gene encoding the interleukin into a host cell such as a mammalian cell, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium.
  • the present invention relates to such a fusion protein of an interleukin and SA that can be efficiently produced as a highly active recombinant protein, and also relates to a method for producing the fusion protein.
  • a physiologically active protein to be fused with the interleukin, and all interleukins can be made into a fusion protein with SA.
  • the present invention relates in particular to a fusion protein in which serum albumin (SA) and interleukin 10 (IL-10) are bound, for example, by binding the C-terminus of SA to the N-terminus of IL-10, or the C-terminus of IL-10 to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • IL-10 interleukin 10
  • IL-10 is expressed as a recombinant protein by introducing a gene encoding IL-10 into a host cell such as a mammalian cell
  • the expression level and/or activity of IL-10 may be low, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium.
  • the present invention relates to such a fusion protein of IL-10 and SA that can be efficiently produced as a highly active recombinant protein, and also to a method for producing said fusion protein.
  • the present invention relates to a fusion protein in which serum albumin (SA) and a neurotrophic factor are bound, for example, by binding the C-terminus of SA to the N-terminus of a neurotrophic factor, or the C-terminus of a neurotrophic factor to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • Some neurotrophic factors have low expression levels and/or low activity when expressed as recombinant proteins by introducing a gene encoding the neurotrophic factor into a host cell such as a mammalian cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in the culture medium.
  • the present invention relates to such a fusion protein of a neurotrophic factor and SA that can be efficiently produced as a highly active recombinant protein, and also relates to a method for producing the fusion protein.
  • physiologically active protein to be fused to the neurotrophic factor, and any neurotrophic factor can be made into a fusion protein with SA.
  • the present invention relates in particular to a fusion protein in which serum albumin (SA) and brain-derived neurotrophic factor (BDNF) are bound, for example, by binding the C-terminus of SA to the N-terminus of BDNF, or the C-terminus of BDNF to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • BDNF brain-derived neurotrophic factor
  • BDNF brain-derived neurotrophic factor
  • SA serum albumin
  • BDNF brain-derived neurotrophic factor
  • the present invention also relates to a fusion protein in which serum albumin (SA) and nerve growth factor (NGF) are bound, for example, by binding the C-terminus of SA to the N-terminus of NGF, or the C-terminus of NGF to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • NGF nerve growth factor
  • the present invention relates to such a fusion protein of NGF and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing the fusion protein.
  • the present invention also relates to a fusion protein in which serum albumin (SA) and neurotrophin 3 (NT-3) are bound, for example, by binding the C-terminus of SA to the N-terminus of NT-3, or the C-terminus of NT-3 to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • NT-3 neurotrophin 3
  • NT-3 is expressed as a recombinant protein by introducing a gene encoding NT-3 into a host cell such as a mammalian cell, the expression level and/or activity may be low, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium.
  • the present invention relates to such a fusion protein of NT-3 and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing said fusion protein.
  • the present invention also relates to a fusion protein in which serum albumin (SA) and neurotrophin 4 (NT-4) are bound, for example, by binding the C-terminus of SA to the N-terminus of NT-4, or the C-terminus of NT-4 to the N-terminus of SA, either directly or via a linker.
  • SA serum albumin
  • NT-4 neurotrophin 4
  • the present invention relates to such a fusion protein of NT-4 and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing said fusion protein.
  • Krabbe disease a type of lysosomal disease, is also known as galactosylceramide lipidosis or globoid cell leukodystrophy. It is a genetic disease caused by a genetic abnormality that reduces the activity or deficiency of galactosylceramidase (galactocerebrosidase, GALC), which is necessary for the breakdown of sphingolipids in lysosomes.
  • GALC galactosylceramidase
  • GALC galactosylceramidase uses galactosylsphingosine, galactocerebroside, and other substrates as catalysts for the hydrolysis of the galactose ester bonds in the molecules of these substrates.
  • Krabbe disease is classified into infantile type, which begins at about 3 to 6 months of age and is characterized by irritability and regression, and often results in death within 2 to 3 years; late infantile type, which begins at about 6 months to 3 years of age and is characterized by irritability, delayed psychomotor development, and regression; juvenile type, which begins at about 3 to 10 years of age and progresses slowly, and is characterized by visual impairment, gait disturbance, ataxia, etc.; and adult type, which begins at about 10 years of age or older and is characterized by psychiatric symptoms, etc.
  • Gaucher disease a type of lysosomal disease, is a genetic disease caused by a genetic abnormality that results in a decrease in the activity or deficiency of glucocerebrosidase ( ⁇ -glucosidase, GBA), an enzyme required for the breakdown of the biological glycolipid glucocerebroside in the lysosome.
  • GBA glucocerebrosidase
  • GBA Glucocerebrosidase
  • Glucocerebroside accumulates in macrophages, particularly in the liver, spleen, and bones, causing anemia and thrombocytopenia due to decreased splenic function, as well as hepatosplenomegaly, bone pain, fractures, and central nervous system disorders. It is believed that the central nervous system disorders are caused by the accumulation of glucosylsphingosine, the lyso form of glucocerebroside, in the brain.
  • Gaucher disease is classified according to the presence or absence and severity of neurological symptoms into the mildest type I (non-neurological type), which occurs at ages ranging from infancy to adulthood and is characterized by slowly progressing symptoms such as enlargement of the liver and spleen, anemia, decreased platelets, and fractures without accompanying neurological symptoms; the most severe type II (acute neurological type), which begins in infancy and, in addition to the symptoms of type I, rapidly progresses with neurological symptoms such as psychomotor developmental delay, convulsions, and neck retroversion, resulting in death from oxygen deficiency by the age of two; and the progressive type III (subacute neurological type), which develops gradually during infancy and progresses more slowly than type II.
  • Lysosomal diseases other than Krabbe disease and Gaucher disease are also caused by genetic deficiencies in lysosomal enzymes.
  • enzyme replacement therapy is performed in which the genetically deficient enzyme is produced as a recombinant enzyme using genetic engineering technology and administered to the patient.
  • Non-Patent Document 1 The gene encoding human GALC (hGALC) was isolated in 1993 (Non-Patent Document 1). However, there are no drugs for use as enzyme replacement therapy for Krabbe disease that contain recombinant human GALC (rhGALC) produced using this gene as the active ingredient.
  • Non-Patent Document 2 The gene encoding human GBA (hGBA) was isolated in 1986 (Non-Patent Document 2). However, there are no drugs for use as enzyme replacement therapy for Gaucher disease that contain recombinant human GBA (rhGBA) produced using this gene as the active ingredient.
  • IL-10 a type of cytokine, is an anti-inflammatory cytokine produced by Th2 cells and can inhibit cytokine production by Th1 cells, suppressing immune responses. Due to its anti-inflammatory effect, IL-10 is expected to be effective against many inflammatory diseases, namely, neuropathic pain, multiple sclerosis, spinal cord injury, ALS, neuroinflammation, symptoms associated with arthritis and other joint diseases, and autoimmune diseases. In addition to its immunosuppressive effect, it has also been reported that IL-10 may have an anti-cancer effect (Non-Patent Document 3). The gene encoding human IL-10 was isolated in 1991 (Non-Patent Document 4). However, there are no pharmaceuticals for use as a therapeutic agent for inflammatory diseases or cancer that contain recombinant human IL-10 (rhIL-10) produced using this gene as an active ingredient.
  • rhIL-10 recombinant human IL-10
  • BDNF neurotrophic factor
  • TrkB a specific receptor on the surface of target cells and regulates the growth of nerve cells, such as the survival and growth of nerve cells and the enhancement of synaptic function. Due to its neurodevelopmental effect, BDNF is expected to be developed as a treatment for a variety of diseases, including neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, spinal degenerative diseases such as amyotrophic lateral sclerosis, as well as diabetic neuropathy, cerebral ischemic disease, developmental disorders such as Rett syndrome, schizophrenia, depression, and Rett syndrome.
  • neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease
  • spinal degenerative diseases such as amyotrophic lateral sclerosis, as well as diabetic neuropathy, cerebral ischemic disease, developmental disorders such as Rett syndrome, schizophrenia, depression, and Rett syndrome.
  • Non-Patent Documents 5 and 6 The gene encoding human BDNF was isolated in 1993 (Non-Patent Documents 5 and 6). However, there are no pharmaceuticals containing recombinant human BDNF (rhBDNF) produced using this gene as an active ingredient for use as a treatment for neurodegenerative diseases, etc.
  • rhBDNF recombinant human BDNF
  • NGF neurotrophic factor
  • a type of neurotrophic factor is a protein that promotes the survival and growth of sympathetic nerve cells and spinal sensory neurons in the peripheral nervous system, and promotes the survival and differentiation of cholinergic nerve cells in the central nervous system, especially in the basal forebrain.
  • NGF is expected to be developed as a treatment for neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, but it is also expected to be effective in preventing aging and degeneration of brain function, improving brain function, preventing and treating dementia, improving memory and learning ability by building neural networks, and enhancing the action of neurotransmitters.
  • Non-Patent Document 7 a gene encoding at least the ⁇ subunit was isolated in 1990 (Non-Patent Document 7).
  • rhNGF recombinant human NGF
  • NT-3 a type of neurotrophic factor, is known to be involved in promoting the survival and growth of nerve cells and glial cells and neurogenesis by transmitting signals through Trk receptors, particularly TrkC, and is attracting attention for its ability to promote neurotransmission and nerve repair, particularly the differentiation and regeneration of photoreceptor cells.
  • Trk receptors particularly TrkC
  • NT-3 is therefore expected to be developed as a treatment for neurodegenerative diseases.
  • the gene encoding human NT-3 was isolated in 1991 (Non-patent Document 8). However, there are no pharmaceuticals for use as a treatment for neurodegenerative diseases that contain recombinant human NT-3 (rhNT-3) as an active ingredient, which is produced using the gene encoding human NT-3.
  • NT-4 a type of neurotrophic factor, transmits signals through Trk receptors, particularly TrkB, and promotes the growth and survival of peripheral and central nervous system neurons, similar to NT-3.
  • TrkB Trk receptors
  • NT-4 is therefore expected to be developed as a treatment for neurodegenerative diseases.
  • the gene encoding human NT-4 was isolated in 1992 (Non-patent Document 9). However, there are no pharmaceuticals for use as a treatment for neurodegenerative diseases that contain recombinant human NT-4 (rhNT-4) as an active ingredient, which is produced using the gene encoding human NT-4.
  • Non-Patent Document 10 A method for producing growth hormone, which is rapidly degraded and loses its activity when administered to the body, as a fusion protein with serum albumin.
  • Growth hormone fused to serum albumin has increased stability in the body. Therefore, although growth hormone is normally administered subcutaneously every day, by making it into a fusion protein with serum albumin, the number of times it is administered can be reduced.
  • One of the objects of the present invention is to provide a physiologically active substance that is usually expressed in a low amount and/or has a low activity when expressed as a recombinant protein using a host cell such as a CHO cell, in the form of a fusion protein with serum albumin (SA).
  • SA serum albumin
  • Such a fusion protein can be efficiently produced as a highly active recombinant protein.
  • Also provided is a method for producing the fusion protein.
  • Another object of the present invention is to provide a lysosomal enzyme that is expressed in a low amount and/or has low activity when expressed as a recombinant protein using a host cell such as a CHO cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in the culture medium, in the form of a fusion protein with serum albumin.
  • a fusion protein can be efficiently produced as a highly active recombinant protein.
  • a method for producing the fusion protein is also provided.
  • the lysosomal enzyme include hGALC and hGBA.
  • a DNA sequence encoding a leader peptide is placed in frame on the 5' side of the gene encoding the recombinant protein. This allows the expressed recombinant protein to be secreted from cells.
  • the leader peptide is preferably an SA leader peptide when an SA is located at the N-terminus of the recombinant protein, a lysosomal enzyme leader peptide when a lysosomal enzyme is located at the N-terminus of the recombinant protein, a cytokine leader peptide when a cytokine is located at the N-terminus of the recombinant protein, or a neurotrophic factor leader peptide when a neurotrophic factor is located at the N-terminus of the recombinant protein.
  • the leader peptide may be a leader peptide of a heterologous protein, such as a growth hormone leader peptide, or an artificial leader peptide.
  • Another object of the present invention is to provide a cytokine that is expressed in a low amount and/or has low activity when expressed as a recombinant protein using a host cell such as a CHO cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in a culture medium, in the form of a fusion protein with serum albumin.
  • a fusion protein can be efficiently produced as a highly active recombinant protein.
  • a method for producing the fusion protein is also provided.
  • the cytokine is, for example, an interleukin, particularly hIL-10.
  • Another object of the present invention is to provide a neurotrophic factor that is expressed in a low amount and/or has low activity when expressed as a recombinant protein using a host cell such as a CHO cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulates in the culture medium, in the form of a fusion protein with serum albumin.
  • a fusion protein can be efficiently produced as a highly active recombinant protein.
  • a method for producing the fusion protein is also provided.
  • Examples of neurotrophic factors include hBDNF, hNGF, hNT-3, and hNT-4.
  • the inventors conducted extensive investigations and found that, as a result of the discovery, when a fusion protein in which HSA is bound directly or via a linker to the N-terminus or C-terminus of human lysosomal enzymes hGALC or hGBA, as described in detail in this specification, is expressed by culturing a host cell into which an expression vector incorporating a gene encoding the fusion protein has been introduced, the expression level of the recombinant fusion protein (converted to the expression level of the portion of the recombinant fusion protein corresponding to the wild-type human lysosomal enzyme) is significantly increased compared to the expression level of the recombinant wild-type human lysosomal enzyme when the recombinant fusion protein is expressed by culturing a host cell into which an expression vector incorporating a gene encoding the wild-type human lysosomal enzyme has been introduced, thereby completing the present invention
  • the present inventors conducted extensive investigations and found that, as a result of culturing and expressing a fusion protein in which HSA is bound directly or via a linker to the N-terminus or C-terminus of the human cytokine hIL-10, as described in detail herein, in a host cell into which an expression vector incorporating a gene encoding the fusion protein has been introduced, the activity of the recombinant fusion protein is significantly increased compared to the activity of recombinant wild-type hIL-10 in which the recombinant fusion protein is expressed in a host cell into which an expression vector incorporating a gene encoding wild-type hIL-10 has been introduced, thereby completing the present invention.
  • a fusion protein in which HSA is bound directly or via a linker to the N-terminus or C-terminus of human neurotrophic factors hBDNF, hNGF, hNT-3, or hNT-4, as described in detail in this specification is expressed by culturing a host cell into which an expression vector incorporating a gene encoding the fusion protein has been introduced, the activity of the recombinant fusion protein is significantly increased compared to the activity of recombinant wild-type human neurotrophic factor when expressed by culturing a host cell into which an expression vector incorporating a gene encoding a wild-type human neurotrophic factor has been introduced, thereby completing the present invention.
  • the present invention includes the following.
  • a fusion protein comprising a lysosomal enzyme and serum albumin (SA).
  • SA serum albumin
  • SA human serum albumin
  • the hGALC has 90% or more identity to wild-type hGALC having the amino acid sequence shown in SEQ ID NO:1
  • the HSA has 90% or more identity to wild-type HSA having the amino acid sequence shown in SEQ ID NO:3. 6.
  • the hGALC comprises the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1
  • the HSA comprises the amino acid sequence of wild-type human serum albumin shown in SEQ ID NO:3. 15.
  • the linker is a peptide chain consisting of 1 to 150 amino acids.
  • the linker comprises an amino acid sequence selected from the group consisting of the following (a) to (g): (a) Gly; (b) Ser; (c) GlySer; (d) GlyGlySer; (e) the amino acid sequence set forth in SEQ ID NO:9; (f) the amino acid sequence set forth in SEQ ID NO: 10; and (g) The amino acid sequence shown in SEQ ID NO:11. 21.
  • linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 10 times: (a) Gly; (b) Ser; (c) GlySer; (d) GlyGlySer; (e) the amino acid sequence set forth in SEQ ID NO:9; (f) the amino acid sequence set forth in SEQ ID NO: 10; and (g) The amino acid sequence shown in SEQ ID NO:11. 22.
  • linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 6 times: (a) Gly; (b) Ser; (c) GlySer; (d) GlyGlySer; (e) the amino acid sequence set forth in SEQ ID NO:9; (f) the amino acid sequence set forth in SEQ ID NO: 10; and (g) The amino acid sequence shown in SEQ ID NO:11. 23.
  • the fusion protein according to claim 19, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 3 to 5 times: (a) Gly; (b) Ser; (c) GlySer; (d) GlyGlySer; (e) the amino acid sequence set forth in SEQ ID NO:9; (f) the amino acid sequence set forth in SEQ ID NO: 10; and (g) The amino acid sequence shown in SEQ ID NO:11.
  • the linker comprises the amino acid sequence Gly Ser. 25.
  • the fusion protein according to 18 above which comprises an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:5.
  • the fusion protein according to claim 26 which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:5.
  • the fusion protein according to claim 26 which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:5.
  • the fusion protein according to claim 26 which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added relative to the amino acid sequence shown in SEQ ID NO:5. 30.
  • the fusion protein according to claim 18, which comprises the amino acid sequence shown in SEQ ID NO:5.
  • the fusion protein according to 17 above which comprises an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:7.
  • the fusion protein according to 17 above which comprises an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:7.
  • the fusion protein according to 32 above which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:7. 34.
  • the fusion protein according to 32 above which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:7.
  • fusion protein according to 32 above which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:7.
  • 36. The fusion protein according to claim 17, comprising the amino acid sequence shown in SEQ ID NO:7.
  • 37. The fusion protein according to any one of 4 to 36 above, which has a specific activity of 10% or more compared with the specific activity of normal wild-type hGALC. 38.
  • hGBA has 90% or more identity to wild-type hGBA having the amino acid sequence shown in SEQ ID NO:37
  • said HSA has 90% or more identity to wild-type HSA having the amino acid sequence shown in SEQ ID NO:3. 40.
  • hGBA comprises an amino acid sequence in which one amino acid has been substituted with respect to the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37.
  • amino acid substitution is within an amino acid family in which the side chain is an amino acid that can be hydroxylated.
  • HSA comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
  • the fusion protein according to claim 38 wherein said hGBA comprises the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, and said HSA comprises the amino acid sequence of wild-type HSA shown in SEQ ID NO:12. 50.
  • linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 10 times: (a) Gly; (b) Ser; (c) GlySer; (d) GlyGlySer; (e) the amino acid sequence set forth in SEQ ID NO:9; (f) the amino acid sequence set forth in SEQ ID NO: 10; and (g) The amino acid sequence shown in SEQ ID NO:11. 56.
  • linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 6 times: (a) Gly; (b) Ser; (c) GlySer; (d) GlyGlySer; (e) the amino acid sequence set forth in SEQ ID NO:9; (f) the amino acid sequence set forth in SEQ ID NO: 10; and (g) The amino acid sequence shown in SEQ ID NO:11. 57.
  • the fusion protein according to the above 53, wherein the linker consists of the amino acid sequence represented by Gly Ser. 59.
  • the fusion protein according to 52 above comprising an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:39. 60.
  • the fusion protein according to 52 above which comprises an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:39. 61.
  • the fusion protein according to 60 above which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:39. 62.
  • the fusion protein according to 60 above which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:39. 63.
  • the fusion protein according to 60 above which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:39.
  • 64. The fusion protein according to claim 51, comprising the amino acid sequence shown in SEQ ID NO:41.
  • 65 The fusion protein according to claim 51, comprising an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:41.
  • 66. The fusion protein according to the above item 51, comprising an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:41. 67.
  • the fusion protein according to 66 above which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:41. 68.
  • the fusion protein according to 66 above which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added relative to the amino acid sequence shown in SEQ ID NO:41. 69.
  • the fusion protein according to 66 above which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added relative to the amino acid sequence shown in SEQ ID NO:41. 70.
  • the fusion protein according to claim 51 comprising the amino acid sequence shown in SEQ ID NO:41. 71.
  • the fusion protein according to any one of 38 to 70 above which has a specific activity of 10% or more compared with the specific activity of normal wild-type hGBA.
  • 72. A DNA comprising a gene encoding the fusion protein according to any one of 1 to 71 above.
  • An expression vector comprising the DNA according to 72 above.
  • 74. A mammalian cell transformed with the expression vector according to 73 above.
  • 75. A method for producing a fusion protein, comprising the step of culturing the mammalian cell according to 74 above in a serum-free medium.
  • 76. A conjugate of the fusion protein according to any one of 1 to 170 above with an antibody.
  • 77. Conjugates of lysosomal enzymes, serum albumin and antibodies.
  • the conjugate according to claim 77 which is selected from the following (1) to (6): (1) A conjugate in which serum albumin is bound to the C-terminus of the lysosomal enzyme directly or via a linker, and the antibody is further bound to the C-terminus of the serum albumin directly or via a linker; (2) A conjugate in which an antibody is bound to the C-terminus of the lysosomal enzyme directly or via a linker, and the serum albumin is further bound to the C-terminus of the antibody directly or via a linker; (3) A conjugate in which the lysosomal enzyme is bound to the C-terminus of the serum albumin directly or via a linker, and the antibody is further bound to the C-terminus of the lysosomal enzyme directly or via a linker; (4) A conjugate in which the antibody is bound to the C-terminus of the serum albumin, either directly or via a linker, and the lysosomal enzyme is further bound to the C-terminus of
  • the receptor on a vascular endothelial cell is selected from the group consisting of an insulin receptor, a transferrin receptor, a leptin receptor, a lipoprotein receptor, and an IGF receptor.
  • the receptor on vascular endothelial cells is a transferrin receptor.
  • the antibody is any one of a Fab antibody, an F(ab') 2 antibody, an F(ab')2 antibody, a single domain antibody, a single chain antibody, a VHH, or an Fc antibody.
  • the fusion protein is bound to either the C-terminus or the N-terminus of the heavy chain of the antibody.
  • the linker sequence comprises an amino acid sequence selected from the group consisting of one glycine, one serine, the amino acid sequence Gly-Ser, the amino acid sequence Ser-Ser, the amino acid sequence Gly-Gly-Ser, the amino acid sequence of SEQ ID NO:9, the amino acid sequence of SEQ ID NO:10, the amino acid sequence of SEQ ID NO:11, and an amino acid sequence consisting of 1 to 10 consecutive amino acids of these amino acid sequences.
  • a DNA comprising a gene encoding the conjugate according to any one of 76 to 87 above.
  • An expression vector comprising the DNA according to 88 above.
  • a method for producing a conjugate of an antibody and a fusion protein of a physiologically active protein and SA comprising the step of culturing the mammalian cell according to 90 above in a serum-free medium.
  • a human lysosomal enzyme that is relatively difficult to express as an active recombinant protein can be provided in the form of a fusion protein with HSA. Since such a fusion protein can be efficiently produced as a highly active recombinant protein, it can be stably supplied to medical institutions as a drug used for enzyme replacement therapy for patients with lysosomal diseases in which the lysosomal enzyme is deficient.
  • a cytokine that is relatively difficult to express as an active recombinant protein can be provided in the form of a fusion protein with HSA.
  • a fusion protein can be efficiently produced as a recombinant protein, it can be stably supplied to medical institutions as a drug.
  • a human neurotrophic factor that is relatively difficult to express as an active recombinant protein can be provided in the form of a fusion protein with HSA. Since such a fusion protein can be efficiently produced as a recombinant protein, it can be stably supplied to medical institutions as a drug. Note that the effects of the present invention are not limited to these.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, HSA, a linker, and hGALC.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hGALC by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having HSA, a linker, and hGBA in that order from the N-terminus.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hGBA by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hGALC, a linker, and HSA.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of hGALC is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of HSA by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hGBA, a linker, and HSA.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of hGBA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of HSA by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, HSA, a linker, and hIL-10.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hIL-10 by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hIL-10, a linker, and HSA.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of hIL-10 is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of HSA by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, HSA, a linker, and hBDNF.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hBDNF by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having HSA, a linker, and hNGF in that order from the N-terminus.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hNGF by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, HSA, a linker, and hNT-3.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hNT-3 by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, HSA, a linker, and hNT-4.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hNT-4 by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hBDNF, a linker, and HSA.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of hBDNF and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hNGF, a linker, and HSA.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of hNGF and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hNT-3, a linker, and HSA.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of hNT-3 and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond.
  • the figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hNT-4, a linker, and HSA.
  • the linker is a peptide linker
  • the fusion protein is such that the C-terminus of hNT-4 and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond.
  • the vertical axis of the bar graph in the upper row (a) indicates the expression level of the enzyme contained in the culture supernatant in terms of enzyme activity ( ⁇ M/hour).
  • the black bars indicate the enzyme activity of wild-type hGALC
  • the white bars indicate the enzyme activity of HSA-hGALC
  • the diagonal line bars indicate the enzyme activity of hGALC-HSA.
  • the middle row (b) shows the results of an analysis of the culture supernatant by SDS-page
  • the lower row (c) shows the results of an analysis of the culture supernatant by Western blotting, showing the positions of the bands corresponding to wild-type hGALC and the fusion protein of HSA and hGALC. From the left, the analysis results are shown 6, 7, and 8 days after the start of culture in transient expression.
  • the figure shows the elution profiles of wild-type hGALC, HSA-hGALC, and hGALC-HSA obtained by transient expression in SE-HPLC analysis.
  • the vertical dashed line indicates the position of the peak derived from the buffer, and the vertical straight line indicates the positions of the peaks corresponding to the HSA-hGALC and hGALC-HSA monomers.
  • FIG. 1 Schematic diagram showing the structure of the pCI MCS-modified vector (plasmid) Schematic diagram showing the structure of Dual(+)pCI-neo vector (plasmid)
  • the figure shows the results of an experiment to confirm the expression levels of wild-type hGALC, Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab by transient expression.
  • the vertical axis shows the expression level of the enzyme contained in the culture supernatant in terms of enzyme activity ( ⁇ M/hour).
  • FIG. 1 shows the elution profiles of transiently expressed Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab in SE-HPLC analysis.
  • the vertical dashed line indicates the position of the peak derived from the buffer.
  • FIG. 1 Schematic diagram showing the structure of pEmIGS-hGBA vector (plasmid) Schematic diagram showing the structure of pCIneo-hGBA vector (plasmid) Schematic diagram showing the structure of pCIneo-HSA-hGBA vector (plasmid)
  • the figure shows the results of an experiment (activity measurement) to confirm the expression levels of wild-type hGBA, HSA-hGBA, and hGBA-HSA by transient expression.
  • the vertical axis shows the expression level of the enzyme contained in the culture supernatant in terms of enzyme activity ( ⁇ M/hour).
  • FIG. 1 shows the results of an experiment (SDS-PAGE) to confirm the expression levels of wild-type hGBA, HSA-hGBA, and hGBA-HSA by transient expression. The positions of the bands corresponding to wild-type hGBA and the fusion protein of HSA and hGBA are shown.
  • the figure shows the results of an experiment (ELISA) to confirm the expression levels of wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 by transient expression.
  • the vertical axis shows the expression level of each protein contained in the culture supernatant as a concentration (mol/L).
  • FIG. 1 shows the results of an experiment (SDS-PAGE) to confirm the expression levels of wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 by transient expression, in which the positions of the bands corresponding to wild-type mIL-10 and the fusion protein of MSA and mIL-10 are shown.
  • This figure shows the results of an experiment (ELISA) to confirm the expression levels of wild-type human neurotrophic factors and fusion proteins of HSA and human neurotrophic factors by transient expression.
  • the vertical axis shows the expression level of each protein contained in the culture supernatant as concentration (mol/L).
  • the black bars show the concentration of each wild-type human neurotrophic factor
  • the white bars show the concentration of each human neurotrophic factor-HSA fusion protein
  • the shaded bars show the concentration of each HSA-human neurotrophic factor fusion protein.
  • Lane (a) corresponds to wild-type human neurotrophic factor
  • lane (b) to human neurotrophic factor-HSA fusion protein
  • lane (c) to HSA-human neurotrophic factor fusion protein.
  • the positions of the bands corresponding to wild-type human neurotrophic factor and the fusion protein of HSA and human neurotrophic factor are shown.
  • There are four types of human neurotrophic factors and from the left in the figure, the results for hBDNF, hNGF, hNT-4, and hNT-4 are shown.
  • Lane (a) corresponds to wild-type human neurotrophic factor
  • lane (b) to human neurotrophic factor-HSA fusion protein
  • lane (c) to HSA-human neurotrophic factor fusion protein.
  • the positions of the bands corresponding to wild-type human neurotrophic factor and the fusion protein of HSA and human neurotrophic factor are shown.
  • the protein should be one that has a low expression level and/or low activity when the gene encoding the protein is introduced into a host cell and expressed as a recombinant protein.
  • examples of such proteins include lysosomal enzymes, cytokines, interleukins, and neurotrophic factors, or fusion proteins of these with antibodies. Interleukins belong to the cytokine category, and are a general term for cytokines distributed in particular from helper T cells.
  • Lysosomal enzymes include, in particular, galactosylceramidase (GALC) and glucocerebrosidase (GBA), or fusion proteins of these with antibodies or ligands.
  • GALC galactosylceramidase
  • GBA glucocerebrosidase
  • lysosomal enzymes are not limited to GALC and GBA.
  • Other lysosomal enzymes that can increase the expression level and/or activity when expressed as recombinant proteins by binding with SA, particularly when expressed so that the recombinant proteins are secreted from cells and accumulated in the culture medium, are also included in the lysosomal enzymes to be bound with SA. The same applies to fusion proteins of these other lysosomal enzymes with antibodies or ligands.
  • the present invention can be applied to lysosomal enzymes that are easy to produce as recombinant proteins. That is, the lysosomal enzyme is not particularly limited, and examples thereof include iduronate-2-sulfatase, ⁇ -L-iduronidase, ⁇ -galactosidase, GM2 activator protein, ⁇ -hexosaminidase A, ⁇ -hexosaminidase B, N-acetylglucosamine 1-phosphotransferase, ⁇ -mannosidase, ⁇ -mannosidase, saposin C, arylsulfatase A, ⁇ -L-fucosidase, aspartylglucosaminidase, ⁇ -N-acetylgalactosaminidase, acid sphingomyelinase, and the like.
  • It may be enzyme, ⁇ -galactosidase A, ⁇ -glucuronidase, heparan N-sulfatase, ⁇ -N-acetylglucosaminidase, acetyl-CoA ⁇ -glucosaminide N-acetyltransferase, N-acetylglucosamine 6-sulfate sulfatase, acid ceramidase, amylo-1,6-glucosidase, sialidase, palmitoyl protein thioesterase-1, tripeptidyl peptidase-1, hyaluronidase-1, acid ⁇ -glucosidase, CLN1, or CLN2.
  • Cytokines include interleukins, such as IL-10, and fusion proteins of IL-10 with antibodies or ligands.
  • the cytokines are not limited to IL-10.
  • the present invention can also be applied to cytokines that are easy to produce as recombinant proteins.
  • fusion proteins also include fusion proteins of cytokines other than IL-10 with SA, and further fusion proteins of these with antibodies.
  • the cytokine is not particularly limited, but may be, for example, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, and IL-19 to IL-36.
  • the neurotrophic factors include, in particular, BDNF, NGF, NT-3, and NT-4, and fusion proteins of these with antibodies.
  • the neurotrophic factors are not limited to these.
  • Other neurotrophic factors that can increase the expression level and/or activity when expressed as a recombinant protein by binding with SA, particularly when expressed so that the recombinant protein is secreted from cells and accumulated in the culture medium, are also included in the neurotrophic factors to be bound with SA.
  • the present invention can be applied to neurotrophic factors that are easy to produce as recombinant proteins.
  • the neurotrophic factors are not particularly limited, and may be, for example, glial cell line neurotrophic factor (GDNF) or NT-5.
  • the biological species of the protein to be fused with SA there are no particular limitations on the biological species of the protein to be fused with SA, but it is preferably derived from humans, such as human lysosomal enzymes, human cytokines, and human growth and nutritional factors.
  • Fusion proteins of serum albumin (SA) and lysosomal enzymes can be used as therapeutic agents in enzyme replacement therapy for lysosomal diseases.
  • SA serum albumin
  • lysosomal enzymes For example, glucocerebrosidase (GBA) fused with SA can be used as a therapeutic agent for Gaucher disease, and galactosylceramidase (GALC) can be used as a therapeutic agent for Krabbe disease.
  • human lysosomal enzyme when used, it includes not only normal wild-type human lysosomal enzymes, but also mutants of human lysosomal enzymes in which one or more amino acid residues have been substituted, deleted, and/or added (in this specification, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence of the wild-type human lysosomal enzyme, as long as the mutants have the function of a human lysosomal enzyme, such as having the enzymatic activity corresponding to the type of human lysosomal enzyme. The same applies to lysosomal enzymes of animal species other than humans.
  • a human lysosomal enzyme when a human lysosomal enzyme is said to have the function of a human lysosomal enzyme, it means that the human lysosomal enzyme has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of a normal wild-type human lysosomal enzyme is taken as 100%.
  • specific activity refers to the enzyme activity per mass of protein.
  • the specific activity of a fusion protein of a human lysosomal enzyme and another protein is calculated as the enzyme activity per mass of the portion of the fusion protein that corresponds to the human lysosomal enzyme.
  • the specific activity of the human lysosomal enzyme in the fusion protein is calculated by multiplying the enzyme activity of the human lysosomal enzyme per unit mass of the fusion protein ( ⁇ M/hour/mg protein) by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein that corresponds to the human lysosomal enzyme).
  • amino acid residues in the amino acid sequence of a wild-type human lysosomal enzyme are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • amino acid residues in the amino acid sequence of a wild-type human lysosomal enzyme are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • a mutant human lysosomal enzyme consisting of an amino acid sequence in which one amino acid residue is deleted from the N-terminus or C-terminus of a wild-type human lysosomal enzyme a mutant human lysosomal enzyme consisting of an amino acid sequence in which two amino acid residues are deleted from the N-terminus or C-terminus of a wild-type human lysosomal enzyme, etc. are also human lysosomal enzymes.
  • a mutation that combines the substitution and deletion of these amino acid residues can be added to the amino acid sequence of a wild-type human lysosomal enzyme. The same can be said for lysosomal enzymes of animal species other than humans.
  • amino acid residues When adding amino acid residues to the amino acid sequence of a wild-type human lysosomal enzyme, one or more amino acid residues are added into the amino acid sequence of the human lysosomal enzyme or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of a wild-type human lysosomal enzyme, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of a wild-type human lysosomal enzyme. The same can be said for lysosomal enzymes of animal species other than humans.
  • a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues can be added to the amino acid sequence of a wild-type human lysosomal enzyme.
  • a human lysosomal enzyme is obtained by deleting 1 to 10 amino acid residues from the amino acid sequence of a wild-type human lysosomal enzyme, substituting 1 to 10 amino acid residues with other amino acid residues, and adding 1 to 10 amino acid residues
  • a human lysosomal enzyme is obtained by deleting 1 to 5 amino acid residues from the amino acid sequence of a wild-type human lysosomal enzyme, substituting 1 to 5 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues
  • a human lysosomal enzyme is obtained by deleting 1 to 3 amino acid residues from the amino acid sequence of a wild-type human lysosomal enzyme, substituting 1 to 3 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues
  • Human lysosomal enzymes are also those in which 1 or 2 amino acid residues have been deleted from the amino acid sequence of a wild-type human lysosomal enzyme, 1 or 2 amino acid residues have been replaced with another amino acid residue, and 1 or 2 amino acid residues have been added. Human lysosomal enzymes are also those in which 1 amino acid residue has been deleted from the amino acid sequence of a wild-type human lysosomal enzyme, 1 amino acid residue has been replaced with another amino acid residue, and 1 amino acid residue has been added. The same can be said about lysosomal enzymes of animal species other than humans.
  • each mutation in a human lysosomal enzyme mutant compared to a normal wild-type human lysosomal enzyme can be easily confirmed by aligning the amino acid sequences of both human lysosomal enzymes. The same can be said for lysosomal enzymes of animal species other than humans.
  • the amino acid sequence of the human lysosomal enzyme mutant preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, to the amino acid sequence of a normal wild-type human lysosomal enzyme. The same can be said for lysosomal enzyme mutants of animal species other than humans.
  • the identity between the amino acid sequence of a wild-type human lysosomal enzyme and the amino acid sequence of a mutant human lysosomal enzyme can be easily calculated using well-known homology calculation algorithms. Examples of such algorithms include BLAST (Altschul S F. J Mol. Biol. 215. 403-10, (1990)), the similarity search method of Pearson and Lipman (Proc. Natl. Acad. Sci. USA. 85. 2444 (1988)), and the local homology algorithm of Smith and Waterman (Adv. Appl. Math. 2. 482-9 (1981)). The same can be said for the lysosomal enzymes of non-human animal species, GALC, GBA, MSA, and non-human animal species SA. These algorithms can also be applied throughout this specification as homology calculation algorithms between the wild-type amino acid sequence of another protein and the amino acid sequence of a mutant of that other protein.
  • human galactosylceramidase “human galactocerebrosidase”, or “hGALC”
  • hGALC human galactosylceramidase
  • mutants of hGALC in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence as shown in SEQ ID NO: 1, as long as they have the function of hGALC, such as the enzyme activity capable of decomposing sphingolipids such as galactocerebroside and/or galactosylsphingosine.
  • Wild-type hGALC is, for example, encoded by a gene having the base sequence as shown in SEQ ID NO: 2.
  • mouse galactosylceramidase when simply referring to "mouse galactosylceramidase”, “mouse galactocerebrosidase”, or “mGALC”, it includes, without distinction, not only the normal wild-type mGALC having the amino acid sequence shown in SEQ ID NO: 14, but also mGALC mutants in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition” of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 14, as long as they have the function of mGALC, such as having the enzymatic activity to degrade sphingolipids such as galactocerebroside and/or galactosylsphingosine.
  • Wild-type mGALC is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 15.
  • hGALC When hGALC is said to have the function of hGALC, it means that hGALC preferably retains a specific activity of 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, of the specific activity of normal wild-type hGALC taken as 100%.
  • specific activity refers to the enzyme activity per mass of protein.
  • the specific activity of a fusion protein of hGALC and another protein is calculated as the enzyme activity per mass of the portion of the fusion protein that corresponds to hGALC. The same can be said for mGALC.
  • the specific activity of hGALC in a fusion protein is calculated by multiplying the enzyme activity of hGALC per unit mass of the fusion protein ( ⁇ M/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hGALC).
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • hGALC mutants consisting of 642 amino acid residues with one amino acid residue deleted from the N-terminus or C-terminus of wild-type hGALC, and hGALC mutants consisting of 641 amino acid residues with two amino acid residues deleted from the N-terminus or C-terminus of wild-type hGALC are also hGALC. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hGALC. The same can be said for GALC of animal species other than humans.
  • amino acid residues When amino acid residues are added to the amino acid sequence of wild-type hGALC, one or more amino acid residues are added into the amino acid sequence of hGALC or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hGALC, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hGALC. The same can be said for GALC of animal species other than humans.
  • amino acid sequence of wild-type hGALC can be obtained by deleting 1 to 10 amino acid residues, substituting 1 to 10 amino acid residues with other amino acid residues, and adding 1 to 10 amino acid residues.
  • amino acid sequence of wild-type hGALC shown in SEQ ID NO:1 can be obtained by deleting 1 to 5 amino acid residues, substituting 1 to 5 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues.
  • the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1 can be obtained by deleting 1 to 3 amino acid residues, hGALC also includes those in which one to three amino acid residues have been replaced with other amino acid residues and one to three amino acid residues have been added, and those in which one or two amino acid residues have been deleted from the wild-type hGALC amino acid sequence shown in SEQ ID NO:1, one or two amino acid residues have been replaced with other amino acid residues, and one or two amino acid residues have been added, and those in which one amino acid residue has been deleted from the wild-type hGALC amino acid sequence shown in SEQ ID NO:1, one amino acid residue has been replaced with other amino acid residue, and one amino acid residue has been added.
  • the same can be said for GALCs of animal species other than humans.
  • each mutation in the hGALC mutant compared to the normal wild-type hGALC can be easily confirmed by aligning the amino acid sequences of both hGALCs. The same can be said for GALCs of animal species other than humans.
  • the amino acid sequence of the hGALC mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of the normal wild-type hGALC shown in SEQ ID NO: 1. The same can be said for GALC of animal species other than humans.
  • human glucocerebrosidase “human ⁇ -glucosidase,” or “hGBA”
  • hGBA human ⁇ -glucosidase
  • Wild-type hGBA is, for example, encoded by a gene having the base sequence as shown in SEQ ID NO: 38.
  • mouse glucocerebrosidase when simply referring to "mouse glucocerebrosidase”, “mouse ⁇ -glucosidase”, or “mGBA”, it includes, without distinction, not only the normal wild-type mGBA having the amino acid sequence shown in SEQ ID NO: 43, but also mGBA mutants in which one or more amino acid residues have been substituted, deleted, and/or added (in this specification, “addition” of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 43, as long as they have the function of mGBA, such as having the enzymatic activity to degrade glycolipids such as galactocerebroside and/or galactosylsphingosine.
  • Wild-type mGBA is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 44.
  • hGBA When hGBA is said to have the function of hGBA, it means that hGBA has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hGBA is taken as 100%.
  • specific activity refers to the enzyme activity per mass of protein.
  • the specific activity of a fusion protein of hGBA and another protein is calculated as the enzyme activity per mass of the portion of the fusion protein that corresponds to hGBA.
  • the specific activity of hGBA in the fusion protein is calculated by multiplying the enzyme activity of hGBA per unit mass of the fusion protein ( ⁇ M/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hGBA). The same can be said for GBA of animal species other than humans.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • hGBA mutants consisting of 496 amino acid residues with one amino acid residue deleted from the N-terminus or C-terminus of wild-type hGBA, and hGBA mutants consisting of 495 amino acid residues with two amino acid residues deleted from the N-terminus or C-terminus of wild-type hGBA are also hGBA. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hGBA. The same can be said for GBAs of animal species other than humans.
  • amino acid residues When amino acid residues are added to the amino acid sequence of wild-type hGBA, one or more amino acid residues are added into the amino acid sequence of hGBA or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hGBA, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hGBA. The same can be said for GBAs of animal species other than humans.
  • an hGBA is obtained by deleting 1 to 10 amino acid residues from the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, substituting 1 to 10 amino acid residues with other amino acid residues, and adding 1 to 10 amino acid residues.
  • An hGBA is also obtained by deleting 1 to 5 amino acid residues from the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, substituting 1 to 5 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues.
  • hGBA is also obtained by deleting 1 to 3 amino acid residues from the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37
  • hGBA also includes the amino acid sequence of wild-type hGBA shown in SEQ ID NO: 37 in which one or two amino acid residues have been deleted, one or two amino acid residues have been replaced with other amino acid residues, and one or two amino acid residues have been added.
  • hGBA also includes the amino acid sequence of wild-type hGBA shown in SEQ ID NO: 37 in which one amino acid residue has been deleted, one amino acid residue has been replaced with other amino acid residue, and one amino acid residue has been added. The same can be said for GBAs of animal species other than humans.
  • each mutation in the hGBA mutant compared to the normal wild-type hGBA can be easily confirmed by aligning the amino acid sequences of both hGBAs. The same can be said for GBAs of animal species other than humans.
  • the amino acid sequence of the hGBA mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type hGBA shown in SEQ ID NO: 37. The same is true for GBA of animal species other than humans.
  • SA serum albumin
  • HSA human serum albumin
  • human serum albumin or "HSA” includes, without distinction, wild-type human serum albumin consisting of 585 amino acids having the amino acid sequence shown in SEQ ID NO: 3, as well as mutants of HSA in which one or more amino acid residues have been substituted, deleted, and/or added to the amino acid sequence shown in SEQ ID NO: 3 (as used herein, “addition” of an amino acid residue means adding a residue to the end or inside of the sequence).
  • Wild-type HSA is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 4.
  • human serum albumin When human serum albumin is expressed as a recombinant protein using host cells such as CHO as a fusion protein bound to a wild-type human lysosomal enzyme, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of at least one type of wild-type human lysosomal enzyme as a human lysosomal enzyme can be increased compared to when the wild-type human lysosomal enzyme is expressed as a recombinant protein by a similar method.
  • human serum albumin when human serum albumin is expressed as a recombinant protein using host cells such as CHO as a fusion protein bound to wild-type hGALC or wild-type hGBA, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of the enzyme can be increased compared to when these wild-type enzymes are expressed as recombinant proteins by a similar method.
  • the human serum albumin here is preferably one that has the functions of human serum albumin, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited thereto.
  • mouse serum albumin or “MSA” includes, without distinction, wild-type mouse serum albumin having the amino acid sequence shown in SEQ ID NO: 16, as well as mutants of MSA in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 16.
  • Wild-type MSA is encoded, for example, by a gene having the base sequence shown in SEQ ID NO: 17.
  • MSA is preferably one that has the functions of MSA, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited to this.
  • serum albumin refers to a comprehensive concept including serum albumin of mammals other than humans, including mouse serum albumin (MSA) and bovine serum albumin (BSA).
  • MSA mouse serum albumin
  • BSA bovine serum albumin
  • serum albumin when serum albumin is expressed as a recombinant protein using host cells such as CHO as a fusion protein bound to wild-type hGALC or wild-type hGBA, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, it is possible to increase the expression amount of the enzyme, compared to when these wild-type enzymes are expressed as recombinant proteins by the same method.
  • Human serum albumin is known to have several wild-type variants. Human serum albumin Redhill is one of them. Human serum albumin Redhill differs from the above-mentioned normal human serum albumin amino acid sequence, which consists of 585 amino acids, in that the 320th amino acid residue from the N-terminus is threonine instead of alanine, and an arginine residue is added to the N-terminus, and consists of 586 amino acids as shown in SEQ ID NO: 12.
  • the above-mentioned change of alanine to threonine creates a sequence represented by Asn-Tyr-Thr in the amino acid sequence of human serum albumin Redhill, and the Asn (asparagine) residue in this sequence is N-linked glycosylated.
  • This human serum albumin Redhill is also a wild-type HSA.
  • HSA mutant human serum albumin (HSA-A320T) consisting of 585 amino acids as shown in SEQ ID NO:13, in which the 320th amino acid residue from the N-terminus of the wild-type HSA amino acid sequence as shown in SEQ ID NO:3 is replaced with threonine.
  • HSA mutant is one in which the 319th amino acid residue, tyrosine, is replaced with an amino acid other than proline while preserving the 318th amino acid residue from the N-terminus of the HSA mutant as shown in SEQ ID NO:13.
  • mutations that can be made to the wild-type HSA amino acid sequence that are permissible in one embodiment of the present invention, but the mutations can also be applied to human serum albumin Redhill, the HSA mutant as shown in SEQ ID NO:13, or serum albumin of animal species other than humans.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • HSA mutants in which one amino acid residue is deleted at the N-terminus or C-terminus of wild-type or HSA-A320T HSA, and HSA mutants in which two amino acid residues are deleted at the N-terminus or C-terminus of wild-type or HSA-A320T HSA are also HSA. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type or HSA-A320T HSA. The same can be said about SA in non-human animal species.
  • amino acid residues When amino acid residues are added to the amino acid sequence of wild-type HSA or HSA-A320T, one or more amino acid residues are added to the amino acid sequence of HSA or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • HSA mutants in which one amino acid residue is added to the N-terminus or C-terminus of wild-type or HSA-A320T HSA, and HSA mutants in which two amino acid residues are added to the N-terminus or C-terminus of wild-type or HSA-A320T HSA, etc. are also HSA.
  • mutations that combine the addition of these amino acid residues and the above-mentioned substitutions can also be added to the amino acid sequence of wild-type or HSA-A320T HSA
  • mutations that combine the addition of these amino acid residues and the above-mentioned deletions can also be added to the amino acid sequence of wild-type or HSA-A320T HSA. The same can be said for SA of animal species other than humans.
  • HSA can be obtained by deleting 1 to 10 amino acid residues from the amino acid sequence of wild-type HSA shown in SEQ ID NO:3, substituting 1 to 10 amino acid residues with other amino acid residues, and adding 1 to 10 amino acid residues
  • HSA can be obtained by deleting 1 to 5 amino acid residues from the amino acid sequence of wild-type HSA shown in SEQ ID NO:3, substituting 1 to 5 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues
  • HSA can be obtained by deleting 1 to 3 amino acid residues from the amino acid sequence of wild-type HSA shown in SEQ ID NO:3, HSA is also one in which 1 to 3 amino acid residues have been replaced with other amino acid residues and 1 to 3 amino acid residues have been added, HSA is also one in which 1 or 2 amino acid residue
  • the amino acid sequence of the HSA mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type HSA shown in SEQ ID NO: 3. The same is true for SA of animal species other than humans.
  • the expression amount of at least one type of wild-type human lysosomal enzyme as a human lysosomal enzyme in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, etc., compared to when the wild-type human lysosomal enzyme is expressed as a recombinant protein by a similar method.
  • the amount of hGALC expressed in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, compared to when wild-type hGALC is expressed as a recombinant protein by a similar method.
  • the amount of hGBA expressed in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, etc., compared to when wild-type hGBA is expressed as a recombinant protein by a similar method.
  • the term "conservative amino acid substitution” refers to a substitution of an amino acid within a family of amino acids that are related in their side chains and chemical properties. Substitutions within such an amino acid family are not expected to significantly alter the function of the original protein.
  • amino acid families include the following (1) to (12): (1) The acidic amino acids aspartic acid and glutamic acid, (2) the basic amino acids histidine, lysine, and arginine; (3) the aromatic amino acids phenylalanine, tyrosine, and tryptophan; (4) Serine and threonine, which are amino acids having a hydroxyl group (hydroxyamino acids), (5) the hydrophobic amino acids methionine, alanine, valine, leucine, and isoleucine, (6) The neutral hydrophilic amino acids cysteine, serine, threonine, asparagine, and glutamine, (7) Glycine and proline, which are amino acids that affect the orientation of peptide chains.
  • Asparagine and glutamine which are amide amino acids (polar amino acids), (9) The aliphatic amino acids alanine, leucine, isoleucine, and valine, (10) Amino acids with small side chains: alanine, glycine, serine, and threonine; (11) Alanine and glycine, which are amino acids with particularly small side chains. (12) The branched chain amino acids valine, leucine, and isoleucine, (13) Proline and serine, which are amino acids whose side chains can be hydroxylated. The same can be said for the lysosomal enzymes GALC, GBA, and SA of non-human animal species.
  • substitution of an amino acid in the amino acid sequence of wild-type human lysosomal enzymes and HSA with another amino acid is preferably a conservative amino acid substitution.
  • the above conservative amino acid substitutions apply to the wild-type forms of hGALC and hGBA.
  • the above wild-type or mutant human lysosomal enzymes such as hGALC or hGBA, whose constituent amino acids are modified with sugar chains are also human lysosomal enzymes.
  • the above wild-type or mutant human lysosomal enzymes whose constituent amino acids are modified with phosphate are also human lysosomal enzymes. Those modified with something other than sugar chains and phosphate are also human lysosomal enzymes.
  • the above wild-type or mutant human lysosomal enzymes whose constituent amino acid side chains have been converted by substitution reactions or the like are also human lysosomal enzymes. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for the lysosomal enzymes, GALC, and GBA of animal species other than humans.
  • human lysosomal enzymes modified with sugar chains such as hGALC or hGBA
  • human lysosomal enzymes modified with phosphate are included in human lysosomal enzymes with the original amino acid sequence.
  • Those modified with things other than sugar chains and phosphate are also included in human lysosomal enzymes with the original amino acid sequence.
  • Human lysosomal enzymes in which the side chains of the amino acids constituting the human lysosomal enzymes have been converted by substitution reactions or the like are also included in human lysosomal enzymes with the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for lysosomal enzymes of animal species other than humans, GALC, and GBA.
  • the above wild-type or mutant HSA in which the constituent amino acids are modified with sugar chains is also HSA.
  • the above wild-type or mutant HSA in which the constituent amino acids are modified with phosphate is also HSA.
  • HSA modified with something other than sugar chains and phosphate is also HSA.
  • the above wild-type or mutant HSA in which the side chains of the constituent amino acids have been converted by substitution reactions or the like is also HSA.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for SA of animal species other than humans.
  • HSA modified with sugar chains is included in HSA with the original amino acid sequence.
  • HSA modified with phosphate is included in HSA with the original amino acid sequence.
  • HSA modified with things other than sugar chains and phosphate is also included in HSA with the original amino acid sequence.
  • HSA with the original amino acid sequence also includes HSA with the original amino acid sequence when the side chains of the amino acids that make up HSA have been changed by substitution reactions, etc. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for SA of animal species other than humans.
  • the present invention relates to a fusion protein in which a polypeptide containing the amino acid sequence of a wild-type or mutant human lysosomal enzyme is bound to a polypeptide containing the amino acid sequence of a wild-type or mutant SA.
  • binding polypeptides refers to binding different polypeptides by covalent bond, either directly or indirectly via a linker.
  • the SA is preferably HSA.
  • the human lysosomal enzyme is, for example, hGALC or hGBA.
  • a common method for linking two different polypeptides is, for example, to create an expression vector incorporating a DNA fragment in which a gene encoding one polypeptide is linked in-frame downstream of a gene encoding the other polypeptide, and then to express the polypeptide as a recombinant protein by culturing a host cell transformed with this expression vector.
  • the resulting recombinant protein is a single-chain polypeptide in which the two polypeptides are peptide-linked, either directly or via different amino acid sequences.
  • SA-human lysosomal enzyme fusion protein or "SA-human lysosomal enzyme” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of a human lysosomal enzyme is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and which has the function of a human lysosomal enzyme.
  • animal species of the SA there is no particular limitation on the animal species of the SA, but it is preferably a mammalian SA such as a primate, mouse, or cow, more preferably a primate SA, and even more preferably HSA.
  • the human lysosomal enzyme in the SA-human lysosomal enzyme fusion protein has the function of a human lysosomal enzyme, it means that the human lysosomal enzyme preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of a normal wild-type human lysosomal enzyme is taken as 100%.
  • the specific activity of the human lysosomal enzyme in the SA-human lysosomal enzyme fusion protein is calculated by multiplying the enzymatic activity of the human lysosomal enzyme per unit mass of the fusion protein ( ⁇ M/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to the human lysosomal enzyme).
  • the term "HSA-human lysosomal enzyme fusion protein" or "HSA-human lysosomal enzyme” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of a human lysosomal enzyme is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of a human lysosomal enzyme.
  • the definition of the SA-human lysosomal enzyme fusion protein described above can be applied.
  • the SA-human lysosomal enzyme fusion protein it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • a fusion protein having an amino acid sequence in which the N-terminus of a lysosomal enzyme of a non-human animal species is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of SA of a non-human animal species, and which has the function of a human lysosomal enzyme is referred to as a "(non-human animal species) SA-(non-human animal species) lysosomal enzyme fusion protein.”
  • a fusion protein of mouse SA and mouse lysosomal enzyme is referred to as an "MSA-mouse lysosomal enzyme fusion protein" or "MSA-mouse lysosomal enzyme.”
  • mutations When mutations are added to an HSA-human lysosomal enzyme fusion protein, which is a fusion protein of wild-type HSA and wild-type human lysosomal enzyme, mutations can be added only to the HSA portion and not to the human lysosomal enzyme portion, mutations can be added only to the human lysosomal enzyme portion without mutations to the HSA portion, or mutations can be added to both the HSA portion and the human lysosomal enzyme portion.
  • the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of human lysosomal enzyme obtained by adding a mutation to the wild-type human lysosomal enzyme described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the human lysosomal enzyme portion is the amino acid sequence of human lysosomal enzyme obtained by adding a mutation to the wild-type human lysosomal enzyme described above.
  • the same can be said about fusion proteins between wild-type SA (including wild-type MSA) of non-human animal species and wild-type human lysosomal enzymes.
  • HSA-human lysosomal enzyme fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-human lysosomal enzyme fusion proteins.
  • HSA-human lysosomal enzyme fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-human lysosomal enzyme fusion proteins.
  • HSA-human lysosomal enzyme fusion proteins modified with anything other than sugar chains and phosphate are also HSA-human lysosomal enzyme fusion proteins.
  • HSA-human lysosomal enzyme fusion proteins in which the side chains of the amino acids constituting the protein are converted by substitution reactions or the like are also HSA-human lysosomal enzyme fusion proteins.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • the same can be said about fusion proteins of SA of a non-human animal species and a human lysosomal enzyme (SA-human lysosomal enzyme), and fusion proteins of SA of a non-human animal species and a lysosomal enzyme of a non-human animal species, such as MSA-mouse lysosomal enzyme fusion proteins.
  • HSA-human lysosomal enzyme fusion proteins modified with sugar chains are included in HSA-human lysosomal enzyme fusion proteins having the original amino acid sequence.
  • HSA-human lysosomal enzyme fusion proteins modified with phosphate are included in HSA-human lysosomal enzyme fusion proteins having the original amino acid sequence.
  • those modified with something other than sugar chains and phosphate are included in HSA-human lysosomal enzyme fusion proteins having the original amino acid sequence.
  • those in which the side chains of the amino acids constituting the HSA-human lysosomal enzyme fusion proteins have been converted by substitution reactions or the like are included in HSA-human lysosomal enzyme fusion proteins having the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • SA-human lysosomal enzyme fusion proteins of SA of animal species other than human and lysosomal enzyme of animal species other than human, such as MSA-mouse lysosomal enzyme fusion protein.
  • HSA-human lysosomal enzyme fusion proteins in which the human lysosomal enzyme that constitutes it is a precursor of human lysosomal enzyme, are also HSA-human lysosomal enzyme fusion proteins.
  • precursor refers to a type that is biosynthesized as an HSA-human lysosomal enzyme fusion protein, and after the biosynthesis, the part that functions as a human lysosomal enzyme is separated from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as a human lysosomal enzyme by itself.
  • the HSA-human lysosomal enzyme fusion protein may be biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the part containing HSA and the part containing the mature human lysosomal enzyme may be separated.
  • the obtained human lysosomal enzyme is not a fusion protein with HSA, but the HSA-human lysosomal enzyme fusion protein is synthesized once during the process of manufacturing the human lysosomal enzyme. Therefore, when human lysosomal enzyme is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-human lysosomal enzyme fusion protein.
  • SA-human lysosomal enzyme fusion proteins of SA of non-human animal species and human lysosomal enzyme
  • fusion proteins of SA of non-human animal species and lysosomal enzyme of non-human animal species such as MSA-mouse lysosomal enzyme fusion protein.
  • SA-hGALC fusion protein or "SA-hGALC” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hGALC is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of SA, and which has the function of hGALC.
  • SA animal species of the SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA.
  • hGALC in the SA-hGALC fusion protein has the function of hGALC
  • hGALC preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of normal wild-type hGALC is taken as 100%.
  • the specific activity of hGALC in the SA-hGALC fusion protein is calculated by multiplying the enzyme activity of hGALC per unit mass of the fusion protein ( ⁇ M/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hGALC).
  • the term "HSA-hGALC fusion protein" or "HSA-hGALC” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hGALC is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hGALC.
  • HSA-hGALC fusion protein has the function of hGALC
  • the definition of the SA-hGALC fusion protein above can be applied.
  • a fusion protein having an amino acid sequence in which the N-terminus of GALC of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of SA of a non-human animal species, and which has the function of GALC is designated as a "(non-human animal species) SA-(non-human animal species) GALC fusion protein.”
  • a fusion protein of mouse SA and mouse GALC is designated as an "MSA-mouse GALC fusion protein" or "MSA-mouse GALC.”
  • the definition of the SA-hGALC fusion protein described above can be applied.
  • the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited thereto.
  • a preferred HSA-hGALC fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 5.
  • the HSA-hGALC fusion protein shown in SEQ ID NO: 5 is obtained by linking wild-type hGALC to the C-terminus of wild-type HSA via the linker sequence Gly-Ser.
  • the HSA-hGALC fusion protein shown in SEQ ID NO: 5 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 6.
  • HSA-hGALC fusion proteins include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 5 have been replaced with other amino acid residues, deleted, or mutated by addition, so long as they have the function of hGALC. It is preferable that the HSA-hGALC fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • MSA-mGALC fusion protein refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of mGALC is linked, directly or via a linker, to the C-terminus of the amino acid sequence of MSA, and which has the function of mGALC.
  • a preferred MSA-mGALC fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 18.
  • the MSA-mGALC fusion protein shown in SEQ ID NO: 18 is a fusion protein in which wild-type mGALC is linked to the C-terminus of wild-type MSA via the linker sequence Gly-Ser.
  • the MSA-mGALC fusion protein shown in SEQ ID NO: 18 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 19.
  • MSA-mGALC fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 18 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of mGALC. It is preferable that the MSA-mGALC fusion protein has the functions of mouse serum albumin, such as binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • mutations When mutations are introduced into the HSA-hGALC fusion protein, which is a fusion protein of wild-type HSA and wild-type hGALC, mutations can be introduced only in the HSA portion and not in the hGALC portion, mutations can be introduced only in the hGALC portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hGALC portion.
  • mutations When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hGALC obtained by adding a mutation to the wild-type hGALC described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hGALC portion is the amino acid sequence of hGALC obtained by adding a mutation to the wild-type hGALC described above.
  • MSA-mGALC fusion proteins including those having the amino acid sequence shown in SEQ ID NO: 18.
  • the MSA-mGALC fusion protein having the amino acid sequence shown in SEQ ID NO: 18 is encoded by a gene having the base sequence shown in SEQ ID NO: 19, for example.
  • the same can be said about fusion proteins of wild-type SA (including wild-type MSA) of animal species other than humans and wild-type hGALC.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:5 or to the N-terminus or C-terminus of the amino acid sequence.
  • the HSA-hGALC fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hGALC portion, or to both portions.
  • SA-hGALC fusion proteins between SA of a non-human animal species and hGALC
  • GALC fusion proteins between SA of a non-human animal species and GALC of a non-human animal species, such as the fusion protein MSA-mouse GALC.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:5 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:5.
  • HSA-hGALC fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hGALC fusion proteins.
  • HSA-hGALC fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hGALC fusion proteins.
  • HSA-hGALC fusion proteins modified with anything other than sugar chains and phosphate are also HSA-hGALC fusion proteins.
  • HSA-hGALC fusion proteins in which the side chains of the amino acids constituting the protein are modified by substitution reactions or the like are also HSA-hGALC fusion proteins.
  • Such modifications include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • SA-hGALC fusion proteins of SA of non-human animal species and hGALC
  • fusion proteins of SA of non-human animal species and GALC of non-human animal species such as the fusion protein of MSA-mouse lysosomal enzyme.
  • HSA-hGALC fusion proteins modified with sugar chains are included in HSA-hGALC fusion proteins having the original amino acid sequence.
  • HSA-hGALC fusion proteins modified with phosphate are included in HSA-hGALC fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are included in HSA-hGALC fusion proteins having the original amino acid sequence.
  • those in which the side chains of the amino acids constituting the HSA-hGALC fusion protein have been changed by substitution reactions or the like are included in HSA-hGALC fusion proteins having the original amino acid sequence.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • fusion proteins of SA of a non-human animal species and hGALC SA-hGALC
  • fusion proteins of SA of a non-human animal species and GALC of a non-human animal species such as the fusion protein MSA-mouse GALC.
  • HSA-hGALC fusion proteins in which the constituent hGALC is a precursor of hGALC are also HSA-hGALC fusion proteins.
  • the term "precursor” here refers to a type of protein that is biosynthesized as an HSA-hGALC fusion protein, and in which the portion that functions as hGALC after the biosynthesis is cleaved from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hGALC by itself.
  • the HSA-hGALC fusion protein after the HSA-hGALC fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the portion containing HSA and the portion containing mature hGALC may be separated.
  • the obtained hGALC is not a fusion protein with HSA, but the HSA-hGALC fusion protein is synthesized once during the process of synthesizing the hGALC. Therefore, when hGALC is produced by such a method, the production method is included in the method for producing HSA-hGALC fusion protein.
  • SA-hGALC non-human animal species SA and hGALC
  • GALC MSA-mouse GALC
  • SA-hGBA fusion protein or "SA-hGBA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hGBA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of SA, and which has the function of hGBA.
  • SA animal species of the SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA.
  • hGBA in the SA-hGBA fusion protein has the function of hGBA
  • hGBA retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hGBA is taken as 100%.
  • the specific activity of hGBA in the SA-hGBA fusion protein is calculated by multiplying the enzyme activity of hGBA per unit mass of the fusion protein ( ⁇ M/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hGBA).
  • the term "HSA-hGBA fusion protein" or "HSA-hGBA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hGBA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hGBA.
  • HSA-hGBA fusion protein has the function of hGALC
  • the definition of the SA-hGBA fusion protein described above can be applied.
  • a fusion protein having an amino acid sequence in which the N-terminus of GBA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of SA of a non-human animal species, and which has the function of GBA is designated as a "(non-human animal species) SA-(non-human animal species) GBA fusion protein.”
  • a fusion protein of mouse SA and mouse GBA is designated as an "MSA-mouse GBA fusion protein" or "MSA-mouse GBA.”
  • a preferred HSA-hGBA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 39.
  • the HSA-hGBA fusion protein shown in SEQ ID NO: 39 is obtained by linking wild-type hGBA to the C-terminus of wild-type HSA via a linker sequence having the amino acid sequence shown in SEQ ID NO: 9.
  • the HSA-hGBA fusion protein shown in SEQ ID NO: 39 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 40.
  • HSA-hGBA fusion proteins include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 39 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hGBA. It is preferable that the HSA-hGBA fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • MSA-mGBA fusion protein refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of mGBA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of MSA, and which has the function of mGBA.
  • a preferred MSA-mGBA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 45.
  • the MSA-mGBA fusion protein shown in SEQ ID NO: 45 is a fusion protein in which wild-type mGBA is linked to the C-terminus of wild-type MSA via a linker sequence having the amino acid sequence shown in SEQ ID NO: 9.
  • the MSA-mGBA fusion protein shown in SEQ ID NO: 45 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 46.
  • MSA-mGBA fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 45 are replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of mGBA. It is preferable that the MSA-mGBA fusion protein has mouse serum albumin functions, such as binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • mutations When mutations are introduced into an HSA-hGBA fusion protein, which is a fusion protein of wild-type HSA and wild-type hGBA, mutations can be introduced only in the HSA portion and not in the hGBA portion, mutations can be introduced only in the hGBA portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hGBA portion.
  • the amino acid sequence of the portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above.
  • mutations are introduced only in the hGBA portion
  • the amino acid sequence of the portion is the amino acid sequence of hGBA obtained by mutating the wild-type hGBA described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above
  • the amino acid sequence of the hGBA portion is the amino acid sequence of hGBA obtained by mutating the wild-type hGBA described above.
  • the same can be said about MSA-mGBA fusion proteins, including those having the amino acid sequence shown in SEQ ID NO: 45.
  • the MSA-mGBA fusion protein having the amino acid sequence shown in SEQ ID NO: 45 is encoded by a gene having the base sequence shown in SEQ ID NO: 46, for example. The same can be said about fusion proteins of wild-type SA (including wild-type MSA) of animal species other than humans and wild-type hGBA.
  • the following is an example of a case where a mutation is made to an HSA-hGBA fusion protein having the amino acid sequence shown in SEQ ID NO:39.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:39 or to the N-terminus or C-terminus of the amino acid sequence.
  • the HSA-hGBA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hGBA portion, or to both portions.
  • SA-hGBA fusion proteins of SA of a non-human animal species and hGBA
  • GBA fusion protein of a non-human animal species
  • MSA-mouse GBA fusion protein MSA-mouse GBA
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:39 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:39.
  • HSA-hGBA fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hGBA fusion proteins.
  • HSA-hGBA fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hGBA fusion proteins.
  • HSA-hGBA fusion proteins modified with anything other than sugar chains and phosphate are also HSA-hGBA fusion proteins.
  • HSA-hGBA fusion proteins in which the side chains of the amino acids constituting the protein have been converted by substitution reactions or the like are also HSA-hGBA fusion proteins.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hGBA of non-human animal species (SA-hGBA), and fusion proteins of SA and GBA of non-human animal species, such as the fusion protein MSA-mouseGBA.
  • HSA-hGBA fusion proteins modified with sugar chains are included in HSA-hGBA fusion proteins having the original amino acid sequence.
  • HSA-hGBA fusion proteins modified with phosphate are included in HSA-hGBA fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are included in HSA-hGBA fusion proteins having the original amino acid sequence.
  • those in which the side chains of the amino acids constituting the HSA-hGBA fusion protein have been converted by substitution reactions or the like are included in HSA-hGBA fusion proteins having the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • SA-hGBA non-human animal species
  • GBA fusion protein MSA-mouse GBA
  • HSA-hGBA fusion proteins in which the hGBA that constitutes them is a precursor of hGBA are also HSA-hGBA fusion proteins.
  • precursor refers to a type that is biosynthesized as an HSA-hGBA fusion protein, and in which the portion that functions as hGBA after the biosynthesis is separated from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hGBA by itself.
  • the HSA-hGBA fusion protein after the HSA-hGBA fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the portion containing HSA and the portion containing mature hGBA may be separated.
  • the obtained hGBA is not a fusion protein with HSA, but the HSA-hGBA fusion protein is synthesized once during the process of synthesizing the hGBA. Therefore, when hGBA is produced by such a method, the production method is included in the method for producing HSA-hGBA fusion protein.
  • SA-hGBA fusion proteins of SA of a non-human animal species and hGBA
  • GBA fusion proteins of SA of a non-human animal species, such as the MSA-mouse GBA fusion protein.
  • human lysosomal enzyme-SA fusion protein or "human lysosomal enzyme-SA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of a human lysosomal enzyme, and which has the function of a human lysosomal enzyme.
  • the human lysosomal enzyme in a human lysosomal enzyme-SA fusion protein is said to have the function of a human lysosomal enzyme, it means that the human lysosomal enzyme preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of a normal wild-type human lysosomal enzyme is taken as 100%.
  • the specific activity of the human lysosomal enzyme in the human lysosomal enzyme-SA fusion protein is calculated by multiplying the enzymatic activity of the human lysosomal enzyme per unit mass of the fusion protein ( ⁇ M/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to the human lysosomal enzyme).
  • human lysosomal enzyme-HSA fusion protein or "human lysosomal enzyme-HSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of a human lysosomal enzyme, and which has the function of a human lysosomal enzyme.
  • human lysosomal enzyme-HSA fusion protein has the function of a human lysosomal enzyme
  • the definition of human lysosomal enzyme-SA above can be applied.
  • the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • the human lysosomal enzyme-HSA fusion protein it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • the human lysosomal enzyme-HSA fusion protein it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • the human lysosomal enzyme-HSA fusion protein it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • the human lysosomal enzyme-HSA fusion protein it is prefer
  • a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of a lysosomal enzyme of a non-human animal species, and which has the function of a human lysosomal enzyme is referred to as a "(non-human animal species) lysosomal enzyme-(non-human animal species) SA fusion protein.”
  • a fusion protein of mouse lysosomal enzyme and mouse SA is referred to as a "mouse lysosomal enzyme-MSA fusion protein" or "mouse lysosomal enzyme-MSA.”
  • a mutation When a mutation is introduced into a human lysosomal enzyme-HSA fusion protein, which is a fusion protein of wild-type human lysosomal enzyme and wild-type HSA, a mutation can be introduced only into the human lysosomal enzyme portion and not into the HSA portion, a mutation can be introduced only into the HSA portion without introducing a mutation into the human lysosomal enzyme portion, or a mutation can be introduced into both the human lysosomal enzyme portion and the HSA portion.
  • the amino acid sequence of the portion is the amino acid sequence of the human lysosomal enzyme obtained by introducing a mutation into the wild-type human lysosomal enzyme described above.
  • the amino acid sequence of the portion is the amino acid sequence of the HSA obtained by introducing a mutation into the wild-type HSA described above.
  • the amino acid sequence of the human lysosomal enzyme portion is the amino acid sequence of the human lysosomal enzyme obtained by introducing a mutation into the wild-type human lysosomal enzyme described above
  • the amino acid sequence of the HSA portion is the amino acid sequence of the HSA obtained by introducing a mutation into the wild-type HSA described above. The same can be said about fusion proteins between wild-type human lysosomal enzymes and wild-type SA (including wild-type MSA) of non-human animal species.
  • Human lysosomal enzyme-HSA fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also human lysosomal enzyme-HSA fusion proteins.
  • Human lysosomal enzyme-HSA fusion proteins in which the amino acids constituting the protein are modified with phosphate are also human lysosomal enzyme-HSA fusion proteins.
  • Human lysosomal enzyme-HSA fusion proteins modified with anything other than sugar chains and phosphate are also human lysosomal enzyme-HSA fusion proteins.
  • Human lysosomal enzyme-HSA fusion proteins in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like are also human lysosomal enzyme-HSA fusion proteins.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • the same can be said about fusion proteins of human lysosomal enzyme and SA of a species of animal other than human (human lysosomal enzyme-SA) and fusion proteins of lysosomal enzyme and SA of a species of animal other than human, such as mouse lysosomal enzyme-MSA.
  • a human lysosomal enzyme-HSA fusion protein modified with a sugar chain is included in the human lysosomal enzyme-HSA fusion protein having the original amino acid sequence.
  • a human lysosomal enzyme-HSA fusion protein modified with phosphate is included in the human lysosomal enzyme-HSA fusion protein having the original amino acid sequence.
  • a fusion protein modified with something other than sugar chains and phosphate is also included in the human lysosomal enzyme-HSA fusion protein having the original amino acid sequence.
  • a human lysosomal enzyme-HSA fusion protein in which the side chains of the amino acids constituting the human lysosomal enzyme-HSA fusion protein have been converted by a substitution reaction or the like is also included in the human lysosomal enzyme-HSA fusion protein having the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about a fusion protein of a human lysosomal enzyme and SA of an animal species other than human (human lysosomal enzyme-SA), and a fusion protein of a lysosomal enzyme of an animal species other than human and SA of an animal species other than human.
  • a human lysosomal enzyme-HSA fusion protein in which the human lysosomal enzyme that constitutes it is a precursor of a human lysosomal enzyme is also a human lysosomal enzyme-HSA fusion protein.
  • hGALC-SA fusion protein or "hGALC-SA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hGALC, and which has the function of hGALC.
  • hGALC in an hGALC-SA fusion protein is said to have the function of hGALC, it means that hGALC retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hGALC is taken as 100%.
  • the specific activity of hGALC in the hGALC-SA fusion protein is calculated by multiplying the enzyme activity of hGALC per unit mass of the fusion protein ( ⁇ M/hour/mg protein) by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hGALC).
  • hGALC-HSA fusion protein or "hGALC-HSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hGALC, and which has the function of hGALC.
  • the definition of the hGALC-SA fusion protein described above can be applied.
  • a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of GALC of a non-human animal species, and which has the function of GALC is designated as a "(non-human animal species) GALC-(non-human animal species) SA fusion protein.”
  • a fusion protein of mouse GALC and mouse SA is designated as a "mouse GALC-MSA fusion protein" or "mouse GALC-MSA.”
  • the definition of the hGALC-HSA fusion protein described above can be applied.
  • the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited thereto.
  • a preferred hGALC-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 7.
  • the hGALC-HSA fusion protein shown in SEQ ID NO: 7 is obtained by linking wild-type HSA to the C-terminus of wild-type hGALC via the linker sequence Gly-Ser.
  • the hGALC-HSA fusion protein shown in SEQ ID NO: 7 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 8.
  • the hGALC-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 7 have been replaced with other amino acid residues, deleted, or mutated by addition, so long as it has the function of hGALC.
  • the hGALC-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • mGALC-MSA fusion protein refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of MSA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of mGALC, and which has the function of mGALC.
  • a preferred mGALC-MSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 20.
  • the mGALC-MSA fusion protein shown in SEQ ID NO: 20 is a fusion protein in which wild-type MSA is linked to the C-terminus of wild-type mGALC via the linker sequence Gly-Ser.
  • the mGALC-MSA fusion protein shown in SEQ ID NO: 20 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 21.
  • mGALC-MSA fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 20 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of mGALC. It is preferable that the mGALC-MSA fusion protein has mouse serum albumin functions, such as binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • mutations When mutations are introduced into the hGALC-HSA fusion protein, which is a fusion protein of wild-type hGALC and wild-type HSA, mutations can be introduced only in the hGALC portion and not in the HSA portion, mutations can be introduced only in the HSA portion without mutations in the hGALC portion, or mutations can be introduced in both the hGALC portion and the HSA portion.
  • the amino acid sequence of the portion is the amino acid sequence of hGALC obtained by mutating the wild-type hGALC described above.
  • mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above.
  • the amino acid sequence of the hGALC portion is the amino acid sequence of hGALC obtained by mutating the wild-type hGALC described above
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above.
  • the same can be said about mGALC-MSA fusion proteins, including those having the amino acid sequence shown in SEQ ID NO: 20.
  • the mGALC-MSA fusion protein having the amino acid sequence shown in SEQ ID NO: 20 is encoded by a gene having the base sequence shown in SEQ ID NO: 21, for example. The same can be said about fusion proteins of wild-type hGALC and wild-type SA (including wild-type MSA) of animals other than humans.
  • mutations When mutations are introduced into the hGALC-HSA fusion protein, which is a fusion protein of wild-type hGALC and wild-type HSA, mutations can be introduced only in the HSA portion and not in the hGALC portion, mutations can be introduced only in the hGALC portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hGALC portion.
  • mutations When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hGALC obtained by adding a mutation to the wild-type hGALC described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hGALC portion is the amino acid sequence of hGALC obtained by adding a mutation to the wild-type hGALC described above.
  • mGALC-MSA fusion proteins including those having the amino acid sequence shown in SEQ ID NO: 20.
  • the mGALC-MSA fusion protein having the amino acid sequence shown in SEQ ID NO: 20 is encoded by a gene having the base sequence shown in SEQ ID NO: 21, for example.
  • the same can be said about a fusion protein of wild-type hGALC and wild-type SA (hGALC-SA) of an animal other than human.
  • the following is an example of a case where a mutation is made to an hGALC-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:7.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:7 or to the N-terminus or C-terminus of the amino acid sequence.
  • the hGALC-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hGALC portion, or to both portions.
  • SA-GALC non-human animal species
  • fusion proteins between SA and GALC of non-human animal species such as the fusion protein MSA-mouse GALC.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:7 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:7.
  • An hGALC-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also an hGALC-HSA fusion protein.
  • An hGALC-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also an hGALC-HSA fusion protein.
  • An hGALC-HSA fusion protein modified with something other than sugar chains and phosphate is also an hGALC-HSA fusion protein.
  • An hGALC-HSA fusion protein in which the side chains of the amino acids constituting the protein have been changed by a substitution reaction or the like is also an hGALC-HSA fusion protein.
  • Such a change includes, but is not limited to, the conversion of a cysteine residue to formylglycine.
  • the same can be said about a fusion protein of GALC and SA of a non-human animal species (GALC-SA), and a fusion protein of GALC of a non-human animal species and SA of a non-human animal species, for example, a fusion protein of mouse GALC and MSA.
  • hGALC-HSA fusion proteins modified with sugar chains are included in the hGALC-HSA fusion proteins having the original amino acid sequence.
  • hGALC-HSA fusion proteins modified with phosphate are included in the hGALC-HSA fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are included in the hGALC-HSA fusion proteins having the original amino acid sequence.
  • hGALC-HSA fusion proteins in which the side chains of the amino acids constituting the hGALC-HSA fusion protein have been changed by substitution reactions or the like are included in the hGALC-HSA fusion proteins having the original amino acid sequence.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • the same can be said for fusion proteins of GALC and SA of a non-human animal species (GALC-SA), and fusion proteins of GALC of a non-human animal species and SA of a non-human animal species.
  • an hGALC-HSA fusion protein in which the hGALC that constitutes it is a precursor of hGALC is also an HSA-hGALC fusion protein.
  • hGBA-SA fusion protein or "hGBA-SA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hGBA, and which has the function of hGBA.
  • animal species of the SA it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA.
  • hGBA in an hGBA-SA fusion protein has the function of hGBA
  • hGBA retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hGBA is taken as 100%.
  • the specific activity of hGBA in the hGBA-SA fusion protein is calculated by multiplying the enzyme activity of hGBA per unit mass of the fusion protein ( ⁇ M/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hGBA).
  • hGBA-HSA fusion protein or "hGBA-HSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hGBA, and which has the function of hGBA.
  • the definition of the SA-hGBA fusion protein described above can be applied.
  • a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of GBA of a non-human animal species, and which has the function of GBA is designated as a "(non-human animal species) GBA-(non-human animal species) SA fusion protein.”
  • a fusion protein of mouse GBA and mouse SA is designated as a "mouse GBA-MSA fusion protein" or "mouse GBA-MSA.”
  • the definition of the hGBA-HSA fusion protein described above can be applied.
  • the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited thereto.
  • a preferred hGBA-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 41.
  • the hGBA-HSA fusion protein shown in SEQ ID NO: 41 is obtained by linking wild-type HSA to the C-terminus of wild-type hGBA via a linker sequence having the amino acid sequence shown in SEQ ID NO: 9.
  • the hGBA-HSA fusion protein shown in SEQ ID NO: 41 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 42.
  • hGBA-HSA fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 41 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hGBA. It is preferable that the hGBA-HSA fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • mGBA-MSA fusion protein or "mGBA-MSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of MSA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of mGBA, and which has the function of mGBA.
  • a preferred mGBA-MSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 47.
  • the mGBA-MSA fusion protein shown in SEQ ID NO: 47 is a fusion protein in which wild-type MSA is linked to the C-terminus of wild-type mGBA via a linker sequence having the amino acid sequence shown in SEQ ID NO: 9.
  • the mGBA-MSA fusion protein shown in SEQ ID NO: 47 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 48.
  • mGBA-MSA fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 47 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of mGBA. It is preferable that the mGBA-MSA fusion protein has mouse serum albumin functions, such as binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • mutations When mutations are introduced into the hGBA-HSA fusion protein, which is a fusion protein of wild-type hGBA and wild-type HSA, mutations can be introduced only in the hGBA portion and not in the HSA portion, mutations can be introduced only in the HSA portion without mutations in the hGBA portion, or mutations can be introduced in both the hGBA portion and the HSA portion.
  • the amino acid sequence of the portion is the amino acid sequence of hGBA obtained by mutating the wild-type hGBA described above.
  • mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above.
  • the amino acid sequence of the hGBA portion is the amino acid sequence of hGBA obtained by mutating the wild-type hGBA described above
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above.
  • the same can be said about mGBA-MSA fusion proteins, including those having the amino acid sequence shown in SEQ ID NO: 47.
  • the mGBA-MSA fusion protein having the amino acid sequence shown in SEQ ID NO: 47 is encoded by a gene having the base sequence shown in SEQ ID NO: 48, for example.
  • the same can be said about fusion proteins of wild-type hGBA and wild-type SA (including wild-type MSA) of an animal species other than human.
  • the following is an example of a case where a mutation is made to an hGBA-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:41.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:41 or to the N-terminus or C-terminus of the amino acid sequence.
  • the hGBA-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hGBA portion, or to both portions.
  • fusion proteins of hGBA and SA of non-human animal species hGBA-SA
  • fusion proteins of GBA of non-human animal species and SA of non-human animal species such as mouse GBA-MSA fusion protein.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:41 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:41.
  • An hGBA-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also an hGBA-HSA fusion protein.
  • An hGBA-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also an hGBA-HSA fusion protein.
  • An hGBA-HSA fusion protein modified with something other than sugar chains and phosphate is also an hGBA-HSA fusion protein.
  • An hGBA-HSA fusion protein in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like is also an hGBA-HSA fusion protein.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • a fusion protein of hGBA and SA of a non-human animal species hGBA-SA
  • a fusion protein of GBA of a non-human animal species and SA of a non-human animal species such as mouse GBA-MSA fusion protein.
  • hGBA-HSA fusion proteins modified with sugar chains are included in the hGBA-HSA fusion proteins having the original amino acid sequence.
  • hGBA-HSA fusion proteins modified with phosphate are included in the hGBA-HSA fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are included in the hGBA-HSA fusion proteins having the original amino acid sequence.
  • those in which the side chains of the amino acids constituting the hGBA-HSA fusion proteins have been changed by substitution reactions or the like are included in the hGBA-HSA fusion proteins having the original amino acid sequence.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • hGBA-SA non-human animal species
  • GBA fusion proteins of non-human animal species and SA of non-human animal species, such as mouse GBA-MSA fusion protein.
  • an hGBA-HSA fusion protein in which the hGBA that constitutes it is a precursor of hGBA is also an HSA-hGALC fusion protein.
  • fusion protein of SA and lysosomal enzyme include both the above-mentioned “SA-lysosomal enzyme fusion protein” and “lysosomal enzyme-SA fusion protein”.
  • fusion protein of SA and GALC include both the above-mentioned “SA-GALC fusion protein” and "GALC-SA fusion protein”.
  • fusion protein of SA and GBA include both the above-mentioned “SA-GBA fusion protein” and "GBA-SA fusion protein”.
  • SA is HSA
  • lysosomal enzyme is human lysosomal enzyme
  • GALC is hGALC
  • GBA hGBA
  • SA and lysosomal enzymes taking as examples a fusion protein of HSA and hGALC and a fusion protein of HSA and hGBA, and the manufacturing method of the fusion protein, but these descriptions can also be applied to fusion proteins in which the SA is non-human, and the lysosomal enzyme is other than GALC and GBA.
  • they also apply to fusion proteins of SA of a non-human animal species and hGALC or hGBA, fusion proteins of SA of a non-human animal species and GALC or GBA of a non-human animal species, and fusion proteins of MSA and mGALC or mGBA.
  • the SA and the lysosomal enzyme are linked directly or via a linker.
  • linker refers to a structure used to link two types of proteins, or a portion that does not belong to either protein when two or more types of proteins are fused.
  • Linkers include peptide linkers and non-peptide linkers.
  • a peptide linker that constitutes part of a fusion protein can also be simply called a linker.
  • the amino acid sequence of a peptide linker can also be called a linker sequence.
  • the linker refers to a portion that does not belong to either the amino acid sequence of SA or the amino acid sequence of the lysosomal enzyme.
  • the linker is a peptide chain that is interposed between the SA and the lysosomal enzyme. Linkers have various functions.
  • the functions include a function between SA and lysosomal enzyme that binds SA and lysosomal enzyme, a function to reduce mutual interference between SA and lysosomal enzyme by increasing the distance between them within the fusion protein molecule, and a function to act as a hinge between SA and lysosomal enzyme that connects SA and lysosomal enzyme and gives flexibility to the three-dimensional structure of the fusion protein.
  • the linker exerts at least one of these functions. This is the same, for example, when the SA is HSA, and when the lysosomal enzyme is a human lysosomal enzyme, such as hGALC or hGBA.
  • the amino acid sequence of the linker is not particularly limited as long as it functions as a linker in the fusion protein molecule.
  • the length of the peptide linker is also not particularly limited as long as it functions as a linker in the fusion protein molecule.
  • the peptide linker is composed of one or more amino acids. When the peptide linker is composed of multiple amino acids, the number of amino acids is preferably 2 to 50, more preferably 5 to 30, and even more preferably 10 to 25.
  • Suitable examples of peptide linkers include those consisting of Gly-Ser, Gly-Gly-Ser, or amino acid sequences shown in SEQ ID NOs: 9 to 11 (collectively referred to as basic sequences), and those containing these.
  • the peptide linker is one that contains an amino acid sequence in which the basic sequence is repeated 2 to 10 times, one that contains an amino acid sequence in which the basic sequence is repeated 2 to 6 times, and one that contains an amino acid sequence in which the basic sequence is repeated 3 to 5 times.
  • One or more amino acids in these amino acid sequences may be deleted, replaced with other amino acids, added, etc. When deleting an amino acid, the number of amino acids to be deleted is preferably 1 or 2.
  • the number of amino acids to be substituted is preferably 1 or 2.
  • the number of amino acids to be added is preferably 1 or 2.
  • the desired amino acid sequence of the linker portion can be obtained by combining these deletions, substitutions, and additions of amino acids.
  • the peptide linker may be composed of one amino acid, and the amino acid constituting the linker is, for example, glycine or serine. This is the same, for example, when the SA is HSA, and when the lysosomal enzyme is, for example, a human lysosomal enzyme, such as hGALC or hGBA.
  • a fusion protein of HSA and hGALC can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hGALC is linked in-frame downstream or upstream of a gene encoding HSA, and culturing a host cell transformed by introducing this expression vector.
  • a fusion protein of HSA and hGBA can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hGBA is linked in-frame downstream or upstream of a gene encoding HSA, and culturing a host cell transformed by introducing this expression vector.
  • the fusion protein produced as a recombinant protein in this way consists of a single-chain polypeptide.
  • a fusion protein produced as a recombinant protein is called a recombinant fusion protein.
  • a gene encoding hGALC When preparing an HSA-hGALC fusion protein as a recombinant fusion protein, a gene encoding hGALC can be linked in-frame downstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hGALC at the C-terminus of the amino acid sequence of HSA. Conversely, a gene encoding hGALC can be linked in-frame upstream of a gene encoding HSA to obtain an hGALC-HSA fusion protein having the amino acid sequence of hGALC at the N-terminus of the amino acid sequence of HSA.
  • a gene encoding hGBA can be linked in-frame downstream of a gene encoding HSA to obtain an HSA-hGBA fusion protein having the amino acid sequence of hGBA at the C-terminus of the amino acid sequence of HSA.
  • a gene encoding hGBA can be linked in-frame upstream of a gene encoding HSA to obtain an hGBA-HSA fusion protein having the amino acid sequence of hGBA at the N-terminus of the amino acid sequence of HSA.
  • the fusion protein prepared as a recombinant fusion protein is a single-chain polypeptide.
  • single-chain polypeptide refers to a polypeptide that has one N-terminus and one C-terminus and has no branched peptide chains. As long as this condition is met, those that have intramolecular disulfide bonds and those that are modified with sugar chains, lipids, phospholipids, etc. are also single-chain polypeptides. In addition, when single-chain polypeptides form complexes such as dimers through non-covalent bonds, each peptide chain that forms the complex is understood to be a single-chain polypeptide, and the complex itself is understood to be an assembly of single-chain polypeptides.
  • FIG. 1 shows a schematic diagram of a single-chain polypeptide HSA-hGALC fusion protein having HSA, a linker, and hGALC in this order from the N-terminus.
  • FIG. 2 also shows a schematic diagram of a single-chain polypeptide HSA-hGBA fusion protein having HSA, a linker, and hGBA in this order from the N-terminus.
  • HSA-hGBA fusion protein the C-terminus of HSA and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of hGBA are bound by a peptide bond.
  • FIG. 3 shows a single-chain polypeptide hGALC-HSA fusion protein having hGALC, a linker, and HSA in this order from the N-terminus.
  • FIG. 4 shows a single-chain polypeptide hGBA-HSA fusion protein having hGBA, a linker, and HSA in this order from the N-terminus.
  • the C-terminus of hGBA and the N-terminus of the linker are bound by a peptide bond
  • the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond.
  • a fusion protein of HSA and hGALC refers to a fusion protein characterized in that when expressed in host cells as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hGALC in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, compared to when wild-type hGALC is expressed in host cells as a recombinant protein under the same conditions.
  • the fusion protein of HSA and hGBA refers to a fusion protein characterized in that when expressed in a host cell as a recombinant protein, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in the culture medium, the amount of hGBA expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, etc., in terms of concentration or enzyme activity, compared to when wild-type hGBA is expressed in a host cell as a recombinant protein under the same conditions.
  • "under the same conditions” means that the expression vector, host cell, culture conditions, etc. are the same.
  • the preferred host cells used in this case are mammalian cells such as CHO cells and NS/0 cells, and particularly CHO cells.
  • Preferred embodiments of the fusion protein of HSA and hGALC include the following (1) and (2): (1) A polypeptide having an amino acid sequence shown in SEQ ID NO:5, in which the N-terminus of the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO:3 via a linker represented by Gly-Ser; (2) Having the amino acid sequence shown in SEQ ID NO: 7, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hGALC shown in SEQ ID NO: 1 via a linker represented by Gly-Ser.
  • Preferred embodiments of the fusion protein of HSA and hGBA include the following (1) and (2): (1) having the amino acid sequence shown in SEQ ID NO: 39, in which the N-terminus of the amino acid sequence of wild-type hGBA shown in SEQ ID NO: 37 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via a linker having the amino acid sequence shown in SEQ ID NO: 9; (2) Having the amino acid sequence shown in SEQ ID NO: 41, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hGBA shown in SEQ ID NO: 37 via a linker having the amino acid sequence shown in SEQ ID NO: 9.
  • the fusion protein of HSA and hGALC is characterized in that when expressed in a host cell as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hGALC in the culture supernatant is increased in terms of concentration or enzyme activity compared to when wild-type hGALC is expressed in a host cell as a recombinant protein under the same conditions. Therefore, when the fusion protein of HSA and hGALC is produced as a recombinant protein, the production efficiency can be increased compared to wild-type hGALC, and production costs can be reduced.
  • the fusion protein of HSA and hGBA is characterized in that when expressed in a host cell as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hGBA in the culture supernatant is increased in terms of concentration or enzyme activity compared to when wild-type hGBA is expressed in a host cell as a recombinant protein under the same conditions. Therefore, when the fusion protein of HSA and hGBA is produced as a recombinant protein, the production efficiency can be increased compared to wild-type hGBA, and production costs can be reduced.
  • a fusion protein of HSA and hGALC expressed in a host cell as a recombinant protein with wild-type hGALC expressed in a host cell under the same conditions, it is preferable to compare the GALC enzyme activity of the expressed protein rather than the mass of the expressed protein.
  • the expression levels of a fusion protein of HSA and hGBA, a fusion protein of SA of a non-human animal species and hGALC or hGBA, and a fusion protein of SA of a non-human animal species and GALC or GBA of a non-human animal species for example, a fusion protein of MSA and mGALC or mGBA.
  • HSA/hGALC fusion protein and HSA/hGBA fusion protein can be produced as recombinant proteins by culturing host cells transformed with an expression vector incorporating a gene encoding the fusion protein.
  • the host cells used in this case are not particularly limited as long as they are capable of expressing a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA by introducing such an expression vector, and may be any eukaryotic cell such as a mammalian cell, yeast, plant cell, or insect cell, although mammalian cells are particularly preferred.
  • mammalian cells When mammalian cells are used as host cells, there is no particular limitation on the type of mammalian cell, but cells derived from humans, mice, or Chinese hamsters are preferred, and in particular CHO cells derived from Chinese hamster ovary cells or NS/0 cells derived from mouse myeloma are preferred.
  • the expression vector used to incorporate and express a DNA fragment containing a gene encoding a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA can be used without particular limitation as long as it brings about expression of the gene when introduced into a mammalian cell.
  • the gene incorporated in the expression vector is placed downstream of a DNA sequence (gene expression control site) that can regulate the frequency of gene transcription in mammalian cells.
  • a DNA sequence gene expression control site
  • gene expression control sites include a promoter derived from cytomegalovirus, an SV40 early promoter, a human elongation factor-1 ⁇ (EF-1 ⁇ ) promoter, and a human ubiquitin C promoter.
  • GGS glutamine synthetase
  • IVS internal ribosome entry site
  • the term "internal ribosome binding site” refers to a region (structure) present within an mRNA strand to which a ribosome can directly bind and initiate translation independent of a cap structure, or a region (structure) of a DNA strand that generates the region by being transcribed.
  • the term "gene encoding an internal ribosome binding site” refers to a region (structure) of a DNA strand that generates the region by being transcribed.
  • IRES internal ribosome entry sites
  • viruses such as Picornaviridae viruses (poliovirus, rhinovirus, mouse encephalomyocarditis virus, etc.), foot and mouth disease virus, hepatitis A virus, hepatitis C virus, coronavirus, bovine enteric virus, Theiler's murine encephalomyelitis virus, and Coxsackie B virus, as well as in the 5' untranslated regions of genes such as human immunoglobulin heavy chain binding protein, Drosophila antennapedia, and Drosophila ultravithorax.
  • viruses such as Picornaviridae viruses (poliovirus, rhinovirus, mouse encephalomyocarditis virus, etc.), foot and mouth disease virus, hepatitis A virus, hepatitis C virus, coronavirus, bovine enteric virus, Theiler's murine encephalomyelitis virus, and Coxsackie B virus, as well as in the 5' untranslated regions of genes such as human immuno
  • the IRES is a region of approximately 450 bp that exists in the 5' untranslated region of the mRNA.
  • the "5' untranslated region of the virus” refers to the 5' untranslated region of the viral mRNA, or the region (structure) of the DNA strand that produces said region by being transcribed.
  • an expression vector for expressing a target protein which comprises a first gene expression control site, a gene encoding the protein downstream of the first gene expression control site, an internal ribosome binding site further downstream, and a gene encoding glutamine synthetase further downstream, and further comprises a dihydrofolate reductase gene or a drug resistance gene downstream of the first gene expression control site or a second gene expression control site different from the first gene expression control site, can be suitably used for producing a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA.
  • the first gene expression control site or the second gene expression control site can be suitably used with a promoter derived from cytomegalovirus, an SV40 early promoter, a human elongation factor-1 ⁇ promoter (hEF-1 ⁇ promoter), or a human ubiquitin C promoter, with the hEF-1 ⁇ promoter being particularly suitable.
  • a promoter derived from cytomegalovirus an SV40 early promoter, a human elongation factor-1 ⁇ promoter (hEF-1 ⁇ promoter), or a human ubiquitin C promoter, with the hEF-1 ⁇ promoter being particularly suitable.
  • the internal ribosome binding site those derived from the 5' untranslated region of the genome of a virus selected from the group consisting of a virus of the picornavirus family (including mouse encephalomyocarditis virus), foot and mouth disease virus, hepatitis A virus, hepatitis C virus, coronavirus, bovine enteric virus, Theiler's murine encephalomyelitis virus, and Coxsackie B virus, or a gene selected from the group consisting of the human immunoglobulin heavy chain binding protein gene, the Drosophila antennapedia gene, and the Drosophila ultravithorax gene, are preferably used, but an internal ribosome binding site derived from the 5' untranslated region of the mouse encephalomyocarditis virus genome is particularly preferable.
  • a virus of the picornavirus family including mouse encephalomyocarditis virus
  • foot and mouth disease virus hepatitis A virus, hepatitis C virus, coronavirus,
  • the drug resistance gene preferably used is a puromycin or neomycin resistance gene, and more preferably a puromycin resistance gene.
  • an expression vector for expressing a target protein which contains a human elongation factor-1 ⁇ promoter, a gene encoding the protein downstream thereof, an internal ribosome binding site derived from the 5' untranslated region of the mouse encephalomyocarditis virus genome further downstream thereof, and a gene encoding glutamine synthetase further downstream thereof, and further contains another gene expression control site and a dihydrofolate reductase gene downstream thereof, in which the internal ribosome binding site is an internal ribosome binding site in which some of the multiple initiation codons contained in the wild-type internal ribosome binding site have been destroyed, can be suitably used for producing a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA.
  • expression vectors include the expression vectors described in WO2013/161958.
  • an expression vector for expressing a target protein which contains a human elongation factor-1 ⁇ promoter, a gene encoding the protein downstream thereof, an internal ribosome binding site derived from the 5' untranslated region of the mouse encephalomyocarditis virus genome further downstream thereof, and a gene encoding glutamine synthetase further downstream thereof, and further contains another gene expression control site and a drug resistance gene downstream thereof, in which the internal ribosome binding site is an internal ribosome binding site in which some of the multiple initiation codons contained in the wild-type internal ribosome binding site have been destroyed, can be suitably used for producing a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA.
  • expression vectors include pE-mIRES-GS-puro described in WO2012/063799 and pE-mIRES-GS-mNeo described in WO2013/161958.
  • the 3' end of the internal ribosome binding site derived from the 5' untranslated region of the wild-type murine encephalomyocarditis virus genome contains three initiation codons (ATG).
  • the above pE-mIRES-GS-puro and pE-mIRES-GS-mNeo are expression vectors that have an IRES in which some of the initiation codons have been destroyed.
  • the fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA can be expressed in cells or in a medium by culturing a host cell into which an expression vector incorporating a gene encoding the protein has been introduced.
  • the method of expressing the fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA when the host cell is a mammalian cell is described in detail below.
  • a serum-free medium used as a medium for producing recombinant proteins is preferably one containing, for example, 3 to 700 mg/L amino acids, 0.001 to 50 mg/L vitamins, 0.3 to 10 g/L monosaccharides, 0.1 to 10,000 mg/L inorganic salts, 0.001 to 0.1 mg/L trace elements, 0.1 to 50 mg/L nucleosides, 0.001 to 10 mg/L fatty acids, 0.01 to 1 mg/L biotin, 0.1 to 20 ⁇ g/L hydrocortisone, 0.1 to 20 mg/L insulin, 0.1 to 10 mg/L vitamin B12, 0.01 to 1 mg/L putrescine, 10 to 500 mg/L sodium pyruvate and a water-soluble iron compound. If desired, thy
  • DMEM/F12 medium (mixture of DMEM and F12) may be used as a basic medium, and these media are well known to those skilled in the art.
  • DMEM(HG)HAM Improved (R5) medium which contains sodium bicarbonate, L-glutamine, D-glucose, insulin, sodium selenite, diaminobutane, hydrocortisone, ferrous sulfate (II), asparagine, aspartic acid, serine, and polyvinyl alcohol, may be used.
  • serum-free media such as CD OptiCHO TM medium, CHO-S-SFM II medium, or CD CHO medium (Thermo Fisher Scientific), EX-CELL TM 302 medium, or EX-CELL TM 325-PF medium (SAFC Biosciences), may be used as a basic medium.
  • the fusion protein of HSA and hGALC is characterized in that when host cells into which an expression vector incorporating a gene encoding the fusion protein has been introduced are cultured in the above-mentioned serum-free medium to be expressed as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hGALC expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, compared to when wild-type hGALC is expressed as a recombinant protein in host cells under the same conditions.
  • the expression amount of hGBA in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, etc., in terms of concentration or enzyme activity, compared to when wild-type hGBA is expressed as a recombinant protein in a host cell under the same conditions.
  • the host cell used in this case is preferably a mammalian cell, particularly a normal cell used for producing recombinant proteins, such as a CHO cell or an NS/0 cell.
  • wild-type hGALC When wild-type hGALC is expressed as a recombinant using a host cell transformed with an expression vector incorporating a gene encoding it, it is difficult to efficiently produce it as a recombinant due to reasons such as a decrease in the viability of the host cell. However, by expressing hGALC as a fusion protein with HSA, the decrease in the viability of the host cell observed when wild-type hGALC is expressed is suppressed.
  • a fusion protein of HSA and hGALC When a fusion protein of HSA and hGALC is expressed as a recombinant protein by culturing mammalian cells into which an expression vector incorporating a gene encoding it has been introduced in a serum-free medium, especially when the recombinant protein is expressed so that it is secreted from the cells and accumulated in the culture medium, the expression amount increases compared to when wild-type hGALC is expressed as a recombinant protein under the same conditions. It is considered that the suppression of the decrease in viability contributes to this increase in the expression amount.
  • the host cell used for expressing the fusion protein is preferably a mammalian cell, particularly a normal cell used for producing recombinant proteins such as CHO cells and NS/0 cells. That is, one embodiment of the present invention is a recombinant fusion protein between SA and GALC, in particular a recombinant fusion protein between HSA and hGALC.
  • the decrease in the survival rate of host cells when hGALC and hGBA are expressed means that the number of dead cells increases.
  • the contents of dead cells leak out, becoming contaminants, and a process for removing the contaminants is required in the subsequent purification process of the expressed fusion protein.
  • the number of dead cells generated during culture can be reduced, and contaminants can be reduced, making it easier to purify the expressed fusion protein. This also increases the purification efficiency during purification. It also reduces the amount of contaminants contained in the purified fusion protein.
  • a host cell By culturing a host cell encoding a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA, it can be expressed in cells or in a medium. These fusion proteins can be separated from impurities and purified by methods such as column chromatography.
  • the purified fusion protein of HSA and hGALC or fusion protein of HSA and hGBA can be used as a pharmaceutical composition.
  • the fusion protein of HSA and hGALC can be used as a pharmaceutical composition for treating Krabbe disease (galactosylceramide lipidosis or globoid cell leukodystrophy).
  • the fusion protein of HSA and hGBA can be used as a pharmaceutical composition for treating Gaucher disease.
  • pharmaceutical composition refers to a composition that contains a fusion protein as an active ingredient as well as a pharma- ceutical acceptable excipient.
  • a pharmaceutical composition containing the fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA as an active ingredient can be administered intravenously, intramuscularly, intraperitoneally, subcutaneously, or intracerebroventricularly as an injection.
  • injections can be supplied as freeze-dried preparations or aqueous liquid preparations.
  • an aqueous liquid preparation it may be in the form of being filled in a vial, or it can be supplied as a prefilled preparation in which it is filled in a syringe in advance.
  • a freeze-dried preparation it is dissolved in an aqueous medium before use and reconstituted for use.
  • the ratio of monomer to total protein is preferably 70% or more, more preferably 80% or more, even more preferably 90% or more, for example 95% or more.
  • the same can be said for a solution obtained by dissolving a freeze-dried preparation in an aqueous medium and reconstituting it.
  • the fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA can be further bound to an antibody or a ligand.
  • the fusion protein of HSA and hGALC in one embodiment of the present invention can be bound to an antibody or a ligand capable of specifically binding to a receptor on cerebrovascular endothelial cells.
  • the fusion protein of HSA and hGBA in one embodiment of the present invention can be bound to an antibody or a ligand capable of specifically binding to a receptor on cerebrovascular endothelial cells.
  • the fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA can bind to a receptor on cerebrovascular endothelial cells by being bound to an antibody or a ligand capable of specifically binding to a receptor on cerebrovascular endothelial cells.
  • the fusion protein bound to the receptor on cerebrovascular endothelial cells can pass through the blood-brain barrier (BBB) and reach the tissues of the central nervous system (CNS). Therefore, by combining a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA with such an antibody or ligand, it is possible to pass through the blood-brain barrier (BBB) and exert its function in the central nervous system (CNS).
  • BBB blood-brain barrier
  • CNS central nervous system
  • the term "ligand" refers to a substance having affinity for a specific substance, particularly a protein having affinity for a specific substance.
  • such a ligand is a substance, particularly a protein, having affinity for a receptor present on cerebrovascular endothelial cells.
  • receptors present on cerebrovascular endothelial cells include insulin receptors, transferrin receptors, leptin receptors, lipoprotein receptors, and IGF receptors, particularly, but not limited to, transferrin receptors.
  • the receptors are preferably human-derived receptors.
  • Ligands for insulin receptors, transferrin receptors, leptin receptors, lipoprotein receptors, and IGF receptors are insulin, transferrin, leptin, lipoprotein, and IGF (IGF-1 and IGF-2), respectively. These ligands may be full-length or fragments thereof as long as they have affinity for the receptors, and may be wild-type or mutants in which substitutions, deletions, and/or additions have been introduced into the wild-type.
  • hGALC When hGALC is said to have the function of hGALC in a conjugate of an antibody and a fusion protein of HSA and hGALC, it means that hGALC has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more, when the specific activity of normal wild-type hGALC is taken as 100%.
  • the specific activity of hGALC in the conjugate is calculated by multiplying the enzyme activity of hGALC per unit mass of the conjugate ( ⁇ M/hour/mg protein) by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hGALC).
  • hGBA in a conjugate of an antibody and a fusion protein of HSA and hGBA is said to have the function of hGBA, it means that, when the specific activity of normal wild-type hGBA is taken as 100%, hGBA has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more.
  • the specific activity of hGBA in the conjugate is calculated by multiplying the enzyme activity of hGBA per unit mass of the conjugate ( ⁇ M/hour/mg protein) by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hGBA).
  • a conjugate of an antibody and a fusion protein of HSA and hGALC refers to a conjugate characterized in that when the conjugate is expressed as a recombinant protein in host cells, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hGALC in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, compared to when wild-type hGALC is expressed as a recombinant protein in host cells under the same conditions.
  • a conjugate of an antibody and a fusion protein of HSA and hGBA is characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hGBA expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, compared to when wild-type hGBA is expressed as a recombinant protein in a host cell under the same conditions, such as 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, etc.
  • cytokines particularly interleukins
  • IL-10 is one such interleukin.
  • One embodiment of the present invention is a recombinant protein in which such a cytokine that is difficult to produce as a recombinant protein is fused with SA.
  • SA a recombinant protein in which such a cytokine that is difficult to produce as a recombinant protein is fused with SA.
  • Such a recombinant protein can be mass-produced more easily than the corresponding wild-type cytokine using host cells into which an expression vector incorporating a gene encoding the cytokine is introduced.
  • the animal species of the cytokine to be bound to SA, but it is preferably a human cytokine.
  • the animal species of the SA to be bound to the cytokine, but it is preferably HSA.
  • human cytokine when used simply, it includes not only normal wild-type human cytokines, but also mutant human cytokines in which one or more amino acid residues have been substituted, deleted, and/or added to the amino acid sequence of a wild-type human cytokine (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence), without any particular distinction, so long as the mutant has the function of a human cytokine, such as having physiological activity corresponding to the type of human cytokine in question.
  • cytokines of animal species other than humans The same also applies to interleukins.
  • a human cytokine When a human cytokine is said to have the function of a human cytokine, it means that the human cytokine preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of a normal wild-type human cytokine is taken as 100%.
  • specific activity refers to the physiological activity per mass of the protein.
  • the specific activity of a fusion protein of a human cytokine and another protein is determined as the physiological activity per mass of the portion of the fusion protein that corresponds to the human cytokine.
  • the specific activity of the human cytokine in the fusion protein is calculated by multiplying the physiological activity of the human cytokine per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein that corresponds to the human cytokine).
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • a human cytokine mutant consisting of an amino acid sequence in which one amino acid residue is deleted from the N-terminus or C-terminus of a wild-type human cytokine a human cytokine mutant consisting of an amino acid sequence in which two amino acid residues are deleted from the N-terminus or C-terminus of a wild-type human cytokine, and the like are also human cytokines.
  • a mutation that combines the substitution and deletion of these amino acid residues can be added to the amino acid sequence of a wild-type human cytokine. The same applies to cytokines of animal species other than humans. The same applies to interleukins.
  • amino acid residues When adding amino acid residues to the amino acid sequence of a wild-type human cytokine, one or more amino acid residues are added into the amino acid sequence of the human cytokine or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of a wild-type human cytokine, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of a wild-type human cytokine.
  • cytokines of animal species other than humans The same applies to interleukins.
  • a combination of the three types of mutations mentioned above, namely substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of a wild-type human cytokine.
  • a human cytokine is one in which 1 to 10 amino acid residues are deleted from the amino acid sequence of a wild-type human cytokine, 1 to 10 amino acid residues are substituted with other amino acid residues, and 1 to 10 amino acid residues are added;
  • a human cytokine is one in which 1 to 5 amino acid residues are deleted from the amino acid sequence of a wild-type human cytokine, 1 to 5 amino acid residues are substituted with other amino acid residues, and 1 to 5 amino acid residues are added;
  • a human cytokine is one in which 1 to 3 amino acid residues are deleted from the amino acid sequence of a wild-type human cytokine, 1 to 3 amino acid residues are substituted with other amino acid residues, and 1 to 3 amino acid residues are added;
  • each mutation in a human cytokine mutant compared to a normal wild-type human cytokine can be easily confirmed by aligning the amino acid sequences of both human cytokines. The same is true for cytokines of non-human animal species, and also for interleukins.
  • the amino acid sequence of the human cytokine mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, to the amino acid sequence of a normal wild-type human cytokine.
  • identity preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, to the amino acid sequence of a normal wild-type human cytokine.
  • cytokines of animal species other than humans The same also applies to interleukins.
  • human interleukin 10 when simply referring to "human interleukin 10", “human IL-10”, or “hIL-10”, it includes not only the normal wild-type hIL-10 consisting of 160 amino acid residues as shown in SEQ ID NO:24, but also includes, without any particular distinction, mutants of hIL-10 in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, “addition” of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence as shown in SEQ ID NO:24, so long as they have the functions of hIL-10, such as having physiological activity as an inhibitory cytokine that calms the immune response.
  • Wild-type hIL-10 is encoded, for example, by a gene having the base sequence as shown in SEQ ID NO:49. The same applies to IL-10 of animal species other than humans.
  • hIL-10 when hIL-10 is said to have the function of hIL-10, it means that hIL-10 retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hIL-10 is taken as 100%.
  • specific activity refers to the physiological activity per mass of the protein.
  • the specific activity of a fusion protein of hIL-10 and another protein is calculated as the physiological activity per mass of the portion of the fusion protein that corresponds to hIL-10.
  • the specific activity of hIL-10 in the fusion protein is calculated by multiplying the physiological activity of hIL-10 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hIL-10).
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • a mutant hIL-10 consisting of 159 amino acid residues obtained by deleting one amino acid residue at the N-terminus or C-terminus of wild-type hIL-10, and a mutant hIL-10 consisting of 158 amino acid residues obtained by deleting two amino acid residues at the N-terminus or C-terminus of wild-type hIL-10, etc. are also hIL-10.
  • a mutation that combines the substitution and deletion of these amino acid residues can be added to the amino acid sequence of wild-type hIL-10. The same applies to IL-10 of animal species other than humans.
  • amino acid residues When amino acid residues are added to the amino acid sequence of wild-type hIL-10, one or more amino acid residues are added into the amino acid sequence of hIL-10 or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hIL-10, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hIL-10. The same applies to IL-10 from animal species other than humans.
  • a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues can be added to the amino acid sequence of wild-type hIL-10.
  • a wild-type hIL-10 amino acid sequence shown in SEQ ID NO:24 in which 1 to 10 amino acid residues have been deleted, 1 to 10 amino acid residues have been replaced with other amino acid residues, and 1 to 10 amino acid residues have been added is also hIL-10
  • a wild-type hIL-10 amino acid sequence shown in SEQ ID NO:24 in which 1 to 5 amino acid residues have been deleted, 1 to 5 amino acid residues have been replaced with other amino acid residues, and 1 to 5 amino acid residues have been added is also hIL-10
  • a wild-type hIL-10 amino acid sequence shown in SEQ ID NO:24 in which 1 to 3 amino acid residues have been deleted hIL-10 also includes the wild-type hIL-10 amino acid sequence shown in SEQ ID NO:24 in which one or two amino acid residues have been deleted, one
  • hIL-10 also includes the wild-type hIL-10 amino acid sequence shown in SEQ ID NO:24 in which one amino acid residue has been deleted, one amino acid residue has been replaced with other amino acid residue, and one amino acid residue has been added. The same applies to IL-10 from animal species other than humans.
  • each mutation in the hIL-10 mutant compared to normal wild-type hIL-10 can be easily confirmed by aligning the amino acid sequences of both hIL-10. The same is true for IL-10 from animal species other than humans.
  • the amino acid sequence of the hIL-10 mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type hIL-10 shown in SEQ ID NO: 24.
  • substitution of an amino acid in the amino acid sequence of a wild-type human cytokine with another amino acid is preferably conservative amino acid substitutions.
  • substitution of an amino acid in the amino acid sequence of a wild-type human cytokine with another amino acid is preferably conservative amino acid substitutions.
  • substitution of an amino acid in the amino acid sequence of a wild-type human cytokine with another amino acid are preferably conservative amino acid substitutions.
  • the above wild-type or mutant human cytokines in which the constituent amino acids are modified with sugar chains are also human cytokines.
  • the above wild-type or mutant human cytokines in which the constituent amino acids are modified with phosphate are also human cytokines. Those modified with something other than sugar chains and phosphate are also human cytokines.
  • the above wild-type or mutant human cytokines in which the side chains of the constituent amino acids have been converted by substitution reactions or the like are also human cytokines.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • human cytokines modified with sugar chains are included in human cytokines with the original amino acid sequence.
  • Human cytokines modified with phosphate are included in human cytokines with the original amino acid sequence.
  • Cytokines modified with substances other than sugar chains and phosphate are also included in human cytokines with the original amino acid sequence.
  • Cytokines in which the side chains of the amino acids that make up human cytokines have been changed by substitution reactions or the like are also included in human cytokines with the original amino acid sequence.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • a wild-type human cytokine is produced as a recombinant protein fused with HSA.
  • a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium
  • the amount of expression of the human cytokine in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when the wild-type human cytokine is expressed as a recombinant protein by a similar method. For example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 3 to 4 times, etc.
  • hIL-10 is produced as a recombinant protein fused with HSA.
  • a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium
  • the amount of expression of hIL-10 in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hIL-10 is expressed as a recombinant protein by a similar method. For example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 3 to 4 times, etc.
  • one embodiment of the present invention is a fusion protein in which a polypeptide containing the amino acid sequence of a cytokine is bound to a polypeptide containing the amino acid sequence of an SA.
  • binding polypeptides refers to binding different polypeptides by covalent bonding, either directly or indirectly via a linker.
  • the cytokine and SA are preferably of human origin.
  • Another embodiment of the present invention is a fusion protein in which a polypeptide containing the amino acid sequence of IL-10 is bound to a polypeptide containing the amino acid sequence of SA.
  • binding polypeptides refers to binding different polypeptides by covalent bonding, either directly or indirectly via a linker.
  • IL-10 and SA are preferably of human origin.
  • a common method for linking two different polypeptides is, for example, to create an expression vector incorporating a DNA fragment in which a gene encoding one polypeptide is linked in-frame downstream of a gene encoding the other polypeptide, and then to express the resulting protein as a recombinant protein by culturing a host cell transformed with this expression vector.
  • the resulting recombinant protein is a single-chain polypeptide in which the two polypeptides are peptide-linked, either directly or via different amino acid sequences.
  • SA-human cytokine fusion protein or "SA-human cytokine” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of a human cytokine is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and which has the function of a human cytokine.
  • the animal species of the SA is not particularly limited, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA.
  • a human cytokine in an SA-human cytokine fusion protein has the function of a human cytokine
  • the human cytokine has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of a normal wild-type human cytokine is taken as 100%.
  • the specific activity of the human cytokine in the SA-human cytokine fusion protein is calculated by multiplying the physiological activity of the human cytokine per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to the human cytokine).
  • the term "HSA-human cytokine fusion protein" or "HSA-human cytokine” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of a human cytokine is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of a human cytokine.
  • HSA-human cytokine fusion protein has the function of a human cytokine
  • the definition of the SA-human cytokine fusion protein described above can be applied.
  • the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • the HSA-human lysosomal enzyme fusion protein it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • the HSA-human lysosomal enzyme fusion protein is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • the HSA-human lysosomal enzyme fusion protein it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • the HSA-human lysosomal enzyme fusion protein is preferable that
  • mutations When mutations are added to an HSA-human cytokine fusion protein, which is a fusion protein of wild-type HSA and wild-type human cytokine, mutations can be added only to the HSA portion and not to the human cytokine portion, mutations can be added only to the human cytokine portion without mutations to the HSA portion, or mutations can be added to both the HSA portion and the human cytokine portion.
  • the amino acid sequence of the portion is the amino acid sequence of the wild-type HSA with a mutation added.
  • mutations are added only to the human cytokine portion, the amino acid sequence of the portion is the amino acid sequence of the wild-type human cytokine with a mutation added.
  • the amino acid sequence of the HSA portion is the amino acid sequence of the wild-type HSA with a mutation added
  • the amino acid sequence of the human cytokine portion is the amino acid sequence of the wild-type human cytokine with a mutation added. The same can be said for fusion proteins of wild-type SA of animal species other than humans and human cytokine.
  • HSA-human cytokine fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-human cytokine fusion proteins.
  • HSA-human cytokine fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-human cytokine fusion proteins.
  • HSA-human cytokine fusion proteins modified with something other than sugar chains and phosphate are also HSA-human cytokine fusion proteins.
  • HSA-human cytokine fusion proteins in which the side chains of the amino acids constituting the protein have been converted by substitution reactions or the like are also HSA-human cytokine fusion proteins.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA of animal species other than humans and human cytokines (SA-human cytokine).
  • HSA-human cytokine fusion proteins modified with sugar chains are included in HSA-human cytokine fusion proteins having the original amino acid sequence.
  • HSA-human cytokine fusion proteins modified with phosphate are included in HSA-human cytokine fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are also included in HSA-human cytokine fusion proteins having the original amino acid sequence.
  • those in which the side chains of the amino acids constituting the HSA-human cytokine fusion proteins have been converted by substitution reactions or the like are also included in HSA-human cytokine fusion proteins having the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • SA-human cytokines The same can be said for fusion proteins of SA of other animals and human cytokines (SA-human cytokines).
  • SA-human cytokines The same can be said for fusion proteins of SA of animal species other than humans and human cytokines (SA-human cytokines).
  • HSA-human cytokine fusion proteins in which the human cytokine that constitutes it is a precursor of the human cytokine are also HSA-human cytokine fusion proteins.
  • the term "precursor” here refers to a type that is biosynthesized as an HSA-human cytokine fusion protein, and after the biosynthesis, the part that functions as a human cytokine is separated from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as a human cytokine by itself.
  • the HSA-human cytokine fusion protein after the HSA-human cytokine fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the part containing HSA and the part containing the mature human cytokine may be separated.
  • the human cytokine obtained is not a fusion protein with HSA, but the HSA-human cytokine fusion protein is synthesized once during the process of manufacturing the cytokine. Therefore, when human cytokine is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-human cytokine.
  • SA-human cytokine a fusion protein of SA of an animal species other than human and a human cytokine
  • MSA-mouse cytokine fusion protein a fusion protein of SA of an animal species other than human and a human cytokine of an animal species other than human
  • SA-hIL-10 fusion protein or "SA-hIL-10” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hIL-10 is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and which has the function of hIL-10.
  • SA animal species of the SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA.
  • hIL-10 in the SA-hIL-10 fusion protein has the function of hIL-10
  • hIL-10 retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hIL-10 is taken as 100%.
  • the specific activity of hIL-10 in the SA-hIL-10 fusion protein is calculated by multiplying the physiological activity of hIL-10 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hIL-10).
  • the term "HSA-hIL-10 fusion protein" or "HSA-hIL-10” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hIL-10 is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hIL-10.
  • HSA-hIL-10 fusion protein has the function of hIL-10
  • the definition of the SA-hIL-10 fusion protein described above can be applied.
  • the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited to this.
  • a preferred HSA-hIL-10 fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 50.
  • the HSA-hIL-10 fusion protein shown in SEQ ID NO: 50 is a protein in which wild-type hIL-10 is directly bound to the C-terminus of wild-type HSA.
  • the HSA-hIL-10 fusion protein shown in SEQ ID NO: 50 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 51.
  • HSA-hIL-10 fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 50 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hIL-10. It is preferable that the HSA-hIL-10 fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:50 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:50.
  • mutations When mutations are added to an HSA-hIL-10 fusion protein, which is a fusion protein of wild-type HSA and wild-type hIL-10, mutations can be added only to the HSA portion and not to the hIL-10 portion, mutations can be added only to the hIL-10 portion without mutations to the HSA portion, or mutations can be added to both the HSA portion and the hIL-10 portion.
  • mutations When mutations are added only to the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hIL-10 obtained by adding a mutation to the wild-type hIL-10 described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hIL-10 portion is the amino acid sequence of hIL-10 obtained by adding a mutation to the wild-type hIL-10 described above.
  • the HSA-hIL-10 fusion protein having the amino acid sequence shown in SEQ ID NO: 50.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:50 or to the N-terminus or C-terminus of the amino acid sequence.
  • the HSA-hIL-10 fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA part, only to the hIL-10 part, or to both parts.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:50 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:50.
  • HSA-hIL-10 fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hIL-10 fusion proteins.
  • HSA-hIL-10 fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hIL-10 fusion proteins.
  • HSA-hIL-10 fusion proteins modified with anything other than sugar chains and phosphate are also HSA-hIL-10 fusion proteins.
  • HSA-hIL-10 fusion proteins in which the side chains of the amino acids constituting the protein are changed by substitution reactions, etc. Examples of such changes include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • SA-hIL-10 The same can be said for fusion proteins of SA and hIL-10 of other animals (SA-hIL-10).
  • SA-hIL-10 The same can be said for fusion proteins of SA and hIL-10 of animal species other than humans
  • HSA-hIL-10 fusion proteins modified with sugar chains are included in HSA-hIL-10 fusion proteins having the original amino acid sequence.
  • HSA-hIL-10 fusion proteins modified with phosphate are included in HSA-hIL-10 fusion proteins having the original amino acid sequence.
  • those modified with something other than sugar chains and phosphate are included in HSA-hIL-10 fusion proteins having the original amino acid sequence.
  • HSA-hIL-10 fusion proteins in which the side chains of the amino acids constituting the HSA-hIL-10 fusion proteins have been changed by substitution reactions or the like are included in HSA-hIL-10 fusion proteins having the original amino acid sequence.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hIL-10 of animal species other than humans (SA-hIL-10).
  • HSA-hIL-10 fusion proteins in which the constituent hIL-10 is a precursor of hIL-10 are also HSA-hIL-10 fusion proteins.
  • precursor refers to a type that is biosynthesized as an HSA-hIL-10 fusion protein, and after the biosynthesis, the part that functions as hIL-10 is cleaved from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hIL-10 by itself.
  • the HSA-hIL-10 fusion protein after the HSA-hIL-10 fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the part containing HSA and the part containing mature hIL-10 may be separated.
  • the obtained hIL-10 is not a fusion protein with HSA, but the HSA-hIL-10 fusion protein is synthesized once during the process of synthesizing the hIL-10. Therefore, when hIL-10 is produced by such a method, the production method is included in the method for producing HSA-hIL-10 fusion protein. The same can be said about the fusion protein of SA and hIL-10 from non-human animal species (SA-hIL-10).
  • human cytokine-SA fusion protein or "human cytokine-SA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of a human cytokine, and which has a function as a human cytokine.
  • a human cytokine in a human cytokine-SA fusion protein When a human cytokine in a human cytokine-SA fusion protein is said to have a function as a human cytokine, it means that the human cytokine retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of a normal wild-type human cytokine is taken as 100%.
  • the specific activity of the human cytokine in the human cytokine-SA fusion protein is calculated by multiplying the physiological activity of the human cytokine per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to the human cytokine).
  • human cytokine-HSA fusion protein or "human cytokine-HSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of a human cytokine, and which has the function of a human cytokine.
  • the definition of human cytokine-SA above can be adopted.
  • the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • the same is true for the human cytokine-HSA fusion protein.
  • mutations When mutations are introduced into a human cytokine-HSA fusion protein, which is a fusion protein of a wild-type human cytokine and a wild-type HSA, mutations can be introduced only into the human cytokine portion and not into the HSA portion, mutations can be introduced only into the HSA portion without mutations in the human cytokine portion, or mutations can be introduced into both the human cytokine portion and the HSA portion.
  • the amino acid sequence of the portion is the amino acid sequence of the human cytokine obtained by mutating the wild-type human cytokine described above.
  • the amino acid sequence of the portion is the amino acid sequence of the HSA obtained by mutating the wild-type HSA described above.
  • the amino acid sequence of the human cytokine portion is the amino acid sequence of the human cytokine obtained by mutating the wild-type human cytokine described above
  • the amino acid sequence of the HSA portion is the amino acid sequence of the HSA obtained by mutating the wild-type HSA described above. The same can be said about a fusion protein between a wild-type human cytokine and the wild-type SA of an animal species other than human.
  • a human cytokine-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also a human cytokine-HSA fusion protein.
  • a human cytokine-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also a human cytokine-HSA fusion protein.
  • a human cytokine-HSA fusion protein modified with something other than sugar chains and phosphate is also a human cytokine-HSA fusion protein.
  • a human cytokine-HSA fusion protein in which the side chains of the amino acids constituting the protein have been modified by a substitution reaction or the like is also a human cytokine-HSA fusion protein.
  • Such a conversion includes, but is not limited to, the conversion of a cysteine residue to formylglycine. The same can be said about a fusion protein of a human cytokine and SA of an animal species other than human (human cytokine-SA).
  • a human cytokine-HSA fusion protein modified with a sugar chain is included in the human cytokine-HSA fusion protein having the original amino acid sequence.
  • a human cytokine-HSA fusion protein modified with phosphate is included in the human cytokine-HSA fusion protein having the original amino acid sequence.
  • a human cytokine-HSA fusion protein modified with something other than a sugar chain or phosphate is also included in the human cytokine-HSA fusion protein having the original amino acid sequence.
  • a human cytokine-HSA fusion protein in which the side chains of the amino acids constituting the human cytokine-HSA fusion protein have been changed by a substitution reaction or the like is also included in the human cytokine-HSA fusion protein having the original amino acid sequence.
  • Such a change includes, but is not limited to, the change of a cysteine residue to formylglycine. The same can be said about a fusion protein of a human cytokine and SA of an animal species other than human (human cytokine-SA).
  • a human cytokine-HSA fusion protein in which the constituent human cytokine is a precursor of a human cytokine is also a human cytokine-HSA fusion protein.
  • the same can be said for a fusion protein of a human cytokine and SA of an animal species other than human (human cytokine-SA).
  • hIL-10-SA fusion protein or "hIL-10-SA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hIL-10, and which has the function of hIL-10.
  • hIL-10 in the hIL-10-SA fusion protein is said to have the function of hIL-10, it means that hIL-10 retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hIL-10 is taken as 100%.
  • the specific activity of hIL-10 in the hIL-10-SA fusion protein is calculated by multiplying the physiological activity of hIL-10 per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hIL-10).
  • hIL-10-HSA fusion protein or "hIL-10-HSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hIL-10, and which has the function of hIL-10.
  • the definition of hIL-10-SA above can be applied.
  • the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited to this.
  • a preferred hIL-10-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 52.
  • the hIL-10-HSA fusion protein shown in SEQ ID NO: 52 is a fusion protein in which wild-type HSA is directly bound to the C-terminus of wild-type hIL-10.
  • the hIL-10-HSA fusion protein shown in SEQ ID NO: 52 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 53.
  • the hIL-10-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 52 are replaced with other amino acid residues, deleted, or mutated by addition, as long as it has the function of hIL-10.
  • the hIL-10-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:52 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:52.
  • mutations When mutations are introduced into the hIL-10-HSA fusion protein, which is a fusion protein of wild-type hIL-10 and wild-type HSA, mutations can be introduced only in the HSA portion and not in the hIL-10 portion, mutations can be introduced only in the hIL-10 portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hIL-10 portion.
  • the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hIL-10 obtained by adding a mutation to the wild-type hIL-10 described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hIL-10 portion is the amino acid sequence of hIL-10 obtained by adding a mutation to the wild-type hIL-10 described above.
  • the hIL-10-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 52.
  • the same is true for the fusion protein of wild-type hIL-10 and wild-type SA (hIL-10-SA) of an animal species other than human.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:52 or to the N-terminus or C-terminus of the amino acid sequence.
  • the hIL-10-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hIL-10 portion, or both portions.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:52 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:52.
  • hIL-10-HSA fusion proteins in which the amino acids constituting the fusion protein are modified with sugar chains are also hIL-10-HSA fusion proteins.
  • hIL-10-HSA fusion proteins in which the amino acids constituting the fusion protein are modified with phosphate are also hIL-10-HSA fusion proteins.
  • hIL-10-HSA fusion proteins modified with anything other than sugar chains and phosphate are also hIL-10-HSA fusion proteins.
  • hIL-10-HSA fusion proteins in which the side chains of the amino acids constituting the fusion protein are changed by a substitution reaction or the like are also hIL-10-HSA fusion proteins.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of SA and hIL-10 of animal species other than humans (SA-hIL-10).
  • hIL-10-HSA fusion proteins modified with sugar chains are included in the hIL-10-HSA fusion proteins having the original amino acid sequence.
  • hIL-10-HSA fusion proteins modified with phosphate are included in the hIL-10-HSA fusion proteins having the original amino acid sequence.
  • those modified with something other than sugar chains and phosphate are also included in the hIL-10-HSA fusion proteins having the original amino acid sequence.
  • those in which the side chains of the amino acids constituting the hIL-10-HSA fusion protein have been changed by a substitution reaction or the like are also included in the hIL-10-HSA fusion proteins having the original amino acid sequence.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hIL-10 of animal species other than humans (SA-hIL-10).
  • a hIL-10-HSA fusion protein in which the constituent hIL-10 is a precursor of hIL-10 is also a hIL-10-HSA fusion protein.
  • SA and hIL-10 of a non-human animal species SA-hIL-10
  • SA-hIL-10 a fusion protein of SA and hIL-10 of a non-human animal species
  • SA-hIL-10 a fusion protein of SA and IL-10 of a non-human animal species
  • a fusion protein of SA and IL-10 of a non-human animal species such as a fusion protein of MSA-mouse IL-10.
  • the terms “HSA and human cytokine fusion protein”, “human cytokine and HSA fusion protein”, or “human serum albumin and human cytokine fusion protein” include both the above-mentioned “HSA-human cytokine fusion protein” and “human cytokine HSA fusion protein”.
  • the terms “HSA and hIL-10 fusion protein”, “hIL-10 and HSA fusion protein”, or “human serum albumin and human cytokine 10 fusion protein” include both the above-mentioned "HSA-hIL-10 fusion protein” and "hIL-10-HSA fusion protein”.
  • fusion protein of HSA and hIL-10 The manufacturing method and other details of the fusion protein of HSA and hIL-10 are described below, but these descriptions can also be applied when the cytokine is other than IL-10, when the SA is from an animal species other than human, or when the cytokine is from an animal species other than human. For example, they also apply to a fusion protein of SA of an animal species other than human and hIL-10, and a fusion protein of SA of an animal species other than human and IL-10 of an animal species other than human.
  • the SA and the cytokine are linked directly or via a linker.
  • linker refers to a portion that does not belong to either the amino acid sequence of the SA or the amino acid sequence of the cytokine.
  • the linker is a peptide chain that is interposed between the SA and the cytokine.
  • the linker has various functions.
  • These functions include a function between the SA and the cytokine to link the SA and the cytokine, a function to reduce mutual interference between the SA and the cytokine by increasing the distance between them within the fusion protein molecule, and a function between the SA and the cytokine to act as a hinge that connects the SA and the cytokine and gives flexibility to the three-dimensional structure of the fusion protein.
  • the linker exerts at least one of these functions. This is the same, for example, whether the SA is HSA or whether the cytokine is a human cytokine, such as hIL-10.
  • the amino acid sequence of the peptide linker is not particularly limited as long as it functions as a linker in the fusion protein molecule.
  • the length of the peptide linker is also not particularly limited as long as it functions as a linker in the fusion protein molecule.
  • the peptide linker is composed of one or more amino acids. When the peptide linker is composed of multiple amino acids, the number of amino acids is preferably 2 to 50, more preferably 5 to 30, and even more preferably 10 to 25.
  • Suitable examples of peptide linkers include those consisting of Gly-Ser, Gly-Gly-Ser, or amino acid sequences shown in SEQ ID NOs: 9 to 11 (collectively referred to as basic sequences), and those containing these.
  • the peptide linker is one that contains an amino acid sequence in which the basic sequence is repeated 2 to 10 times, one that contains an amino acid sequence in which the basic sequence is repeated 2 to 6 times, or one that contains an amino acid sequence in which the basic sequence is repeated 3 to 5 times.
  • One or more amino acids in these amino acid sequences may be deleted, replaced with other amino acids, added, etc. When deleting an amino acid, the number of amino acids to be deleted is preferably 1 or 2.
  • the number of amino acids to be replaced is preferably 1 or 2.
  • the number of amino acids to be added is preferably 1 or 2.
  • the amino acid sequence of the desired linker portion can be obtained by combining these deletions, substitutions, and additions of amino acids.
  • the peptide linker may be composed of one amino acid, and the amino acid that constitutes the linker is, for example, glycine or serine.
  • the fusion protein of HSA and hIL-10 can be produced as a recombinant protein by creating an expression vector incorporating a DNA fragment in which a gene encoding hIL-10 is linked in-frame to the downstream or upstream of a gene encoding HSA, and then culturing a host cell transformed with this expression vector.
  • the fusion protein produced as a recombinant protein in this way consists of a single polypeptide chain.
  • a gene encoding hIL-10 When producing a fusion protein as a recombinant fusion protein, a gene encoding hIL-10 can be linked in-frame downstream of a gene encoding HSA to obtain an HSA-hIL-10 fusion protein having the amino acid sequence of hIL-10 at the C-terminus of the amino acid sequence of HSA. Conversely, a gene encoding hIL-10 can be linked in-frame upstream of a gene encoding HSA to obtain an hIL-10-HSA fusion protein having the amino acid sequence of hIL-10 at the N-terminus of the amino acid sequence of HSA. In either case, the fusion protein produced as a recombinant fusion protein is a single-chain polypeptide.
  • FIG. 5 shows a schematic diagram of an HSA-hIL-10 fusion protein, a single-chain polypeptide having HSA, a linker, and hIL-10 in that order from the N-terminus.
  • the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond
  • the C-terminus of the linker is bound to the N-terminus of hIL-10 by a peptide bond.
  • FIG. 6 shows a schematic diagram of a hIL-10-HSA fusion protein, which is a single-chain polypeptide having hIL-10, a linker, and HSA in that order from the N-terminus.
  • the C-terminus of hIL-10 is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of HSA by a peptide bond.
  • the fusion protein of HSA and hIL-10 refers to a fusion protein characterized in that when expressed in a host cell as a recombinant protein, particularly when expressed so that the recombinant protein is secreted from the cells and accumulated in the culture medium, the amount of expression of hIL-10 in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hIL-10 is expressed in a host cell as a recombinant protein under the same conditions.
  • "under the same conditions” means that the expression vector, host cell, culture conditions, etc. are the same.
  • the preferred host cells used in this case are mammalian cells such as CHO cells and NS/0 cells, but particularly CHO cells.
  • Preferred embodiments of the fusion protein of HSA and hIL-10 include the following (1) to (4): (1) A substance having an amino acid sequence shown in SEQ ID NO:50, in which the N-terminus of the amino acid sequence of wild-type hIL-10 shown in SEQ ID NO:24 is directly linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO:3; (2) Having the amino acid sequence shown in SEQ ID NO:52, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO:3 is directly linked to the C-terminus of the amino acid sequence of wild-type hIL-10 shown in SEQ ID NO:24.
  • preferred embodiments of the fusion protein of HSA and hIL-10 include human serum albumin (HSA-A320T) consisting of 585 amino acids as shown in SEQ ID NO:13, in which the 320th amino acid residue from the N-terminus of the amino acid sequence of wild-type HSA as shown in SEQ ID NO:3 is replaced with threonine, and further include the following (5) to (8): (5) A peptide having an amino acid sequence shown in SEQ ID NO:56, in which the N-terminus of the amino acid sequence of wild-type hIL-10 shown in SEQ ID NO:24 is directly linked to the C-terminus of the amino acid sequence of human serum albumin (HSA-A320T) shown in SEQ ID NO:13; (6) The C-terminus of the amino acid sequence of wild-type hIL-10 shown in SEQ ID NO:24 is directly linked to the N-terminus of the amino acid sequence of human serum albumin (HSA-A320T) shown in SEQ ID NO:13, and the overall amino
  • the fusion protein of HSA and hIL-10 is characterized in that when expressed in a host cell as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hIL-10 expressed in the culture supernatant is increased in terms of concentration or physiological activity, compared to when wild-type hIL-10 is expressed in a host cell as a recombinant protein under the same conditions. Therefore, when produced as a recombinant protein, the fusion protein of HSA and hIL-10 can increase the production efficiency compared to wild-type hIL-10, and therefore can reduce production costs.
  • fusion protein of HSA and hIL-10 expressed in a host cell as a recombinant protein with wild-type hIL-10 expressed in a host cell under the same conditions as a recombinant protein
  • the comparison is not based on the mass of the expressed protein, but on the number of molecules of the expressed protein or the hIL-10 physiological activity.
  • fusion proteins of SA and hIL-10 from animal species other than human, and fusion proteins of SA and other cytokines.
  • the fusion protein of HSA and hIL-10 can be produced using an expression vector, host cell, medium, etc. that can be used to produce the above-mentioned fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA. The same can be said for fusion proteins of SA and cytokines in general. is preferred
  • the fusion protein of HSA and hIL-10 is characterized in that when host cells into which an expression vector incorporating a gene encoding this protein has been introduced are cultured in the above-mentioned serum-free medium to be expressed as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of expression of hIL-10 in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hIL-10 is expressed as a recombinant protein in host cells under the same conditions.
  • the expression vector, host cells, culture conditions, etc. are the same.
  • the host cells used in this case are preferably mammalian cells, particularly ordinary cells used for producing recombinant proteins such as CHO cells and NS/0 cells.
  • Wild-type hIL-10 tends to have a low expression level when expressed as a recombinant using host cells transformed with an expression vector incorporating a gene encoding it, making it difficult to efficiently mass-produce it as a recombinant.
  • the expression level of the recombinant can be increased.
  • the fusion protein of HSA and hIL-10 can be expressed more efficiently than when wild-type hIL-10 is expressed as a recombinant protein under the same conditions, making it suitable for mass production.
  • the host cells used to express the fusion protein are preferably mammalian cells, especially ordinary cells used for producing recombinant proteins such as CHO cells and NS/0 cells. That is, one embodiment of the present invention is a recombinant fusion protein of SA and IL-10, particularly a recombinant fusion protein of HSA and hIL-10.
  • the fusion protein of HSA and hIL-10 By culturing host cells encoding the fusion protein of HSA and hIL-10, it can be expressed in cells or in the medium.
  • the fusion protein of HSA and hIL-10 can be purified by separating it from impurities using methods such as column chromatography.
  • the purified fusion protein of HSA and hIL-10 can be used as a pharmaceutical composition.
  • the fusion protein of HSA and hIL-10 can be used as a pharmaceutical composition targeting inflammatory diseases or cancer.
  • a pharmaceutical composition containing a fusion protein of HSA and hIL-10 as an active ingredient can be administered intravenously, intramuscularly, intraperitoneally, subcutaneously, or intracerebroventricularly as an injection.
  • injections can be supplied as lyophilized preparations or aqueous liquid preparations.
  • an aqueous liquid preparation it may be in the form of a vial filled with the drug, or it can be supplied as a prefilled preparation in a syringe.
  • a lyophilized preparation it is dissolved in an aqueous medium and reconstituted before use.
  • the fusion protein of HSA and hIL-10 can further be conjugated with an antibody or a ligand.
  • the fusion protein of HSA and hIL-10 can be conjugated with an antibody or a ligand capable of specifically binding to a receptor on cerebrovascular endothelial cells.
  • the fusion protein of HSA and hIL-10 or the fusion protein of HSA and hGBA can bind to a receptor on cerebrovascular endothelial cells by being conjugated with an antibody or a ligand capable of specifically binding to a receptor on cerebrovascular endothelial cells.
  • the fusion protein bound to the receptor on cerebrovascular endothelial cells can pass through the blood-brain barrier (BBB) and reach the tissues of the central nervous system (CNS). Therefore, the fusion protein of HSA and hIL-10 can be conjugated with such an antibody or ligand to pass through the blood-brain barrier (BBB) and exert its function in the central nervous system (CNS).
  • BBB blood-brain barrier
  • CNS central nervous system
  • hIL-10 in a conjugate of an antibody and a fusion protein of HSA and hIL-10 is said to have the function of hIL-10, it means that, when the specific activity of normal wild-type hIL-10 is taken as 100%, hIL-10 has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more.
  • the specific activity of hIL-10 in the conjugate is calculated by multiplying the physiological activity of hIL-10 per unit mass of the conjugate by (the molecular weight of the conjugate/the molecular weight of the portion of the conjugate corresponding to hIL-10).
  • a conjugate of an antibody and a fusion protein of HSA and hIL-10 refers to a conjugate characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of expression of hIL-10 in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hIL-10 is expressed as a recombinant protein in a host cell under the same conditions.
  • neurotrophic factors are relatively difficult to produce in large quantities when expressed as recombinant proteins using host cells introduced with an expression vector incorporating a gene encoding the wild-type neurotrophic factor, due to limited expression levels.
  • neurotrophic factors include BDNF (brain-derived neurotrophic factor), NGF (nerve growth factor), NT-3 (neurotrophin-3), NT-4 (neurotrophin-4), and NT-5 (neurotrophin-5).
  • BDNF brain-derived neurotrophic factor
  • NGF nerve growth factor
  • NT-3 neurotrophin-3
  • NT-4 neurotrophin-4
  • NT-5 neurotrophin-5
  • the difference in structure between NT-4 and NT-5 is an interspecies variation, and since these are considered to be the same factor, they may be referred to as NT-4/5, etc., but in this specification, they will be referred to as NT-4.
  • One embodiment of the present invention is a recombinant protein in which such a neurotrophic factor, which is relatively difficult to produce as a recombinant protein, is fused with SA.
  • a recombinant protein can be produced in large quantities more easily using host cells introduced with an expression vector incorporating a gene encoding the recombinant protein, compared to the corresponding neurotrophic factor.
  • SA neurotrophic factor
  • there is no particular restriction on the animal species of the neurotrophic factor to be bound to SA but human neurotrophic factor is preferable.
  • HSA is preferred.
  • human neurotrophic factor when used, it includes not only normal wild-type human neurotrophic factor, but also mutant human neurotrophic factors in which one or more amino acid residues have been substituted, deleted, and/or added (in this specification, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence of the wild-type human neurotrophic factor, as long as the mutant has the function of a human neurotrophic factor, such as having physiological activity corresponding to the type of human neurotrophic factor. The same applies to neurotrophic factors of animal species other than humans.
  • a human neurotrophic factor has a function as a human neurotrophic factor
  • the human neurotrophic factor has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of a normal wild-type human neurotrophic factor is taken as 100%.
  • specific activity refers to the physiological activity per mass of the protein.
  • the specific activity of a fusion protein of a human neurotrophic factor and another protein is calculated as the physiological activity per mass of the portion of the fusion protein that corresponds to the human neurotrophic factor.
  • the specific activity of the human neurotrophic factor in the fusion protein is calculated by multiplying the physiological activity of the human neurotrophic factor per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein that corresponds to the human neurotrophic factor).
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • a human neurotrophic factor mutant consisting of an amino acid sequence in which one amino acid residue is deleted from the N-terminus or C-terminus of wild-type human neurotrophic factor, a human neurotrophic factor mutant consisting of an amino acid sequence in which two amino acid residues are deleted from the N-terminus or C-terminus of wild-type human neurotrophic factor, etc. are also human neurotrophic factors.
  • a mutation that combines the substitution and deletion of these amino acid residues can be added to the amino acid sequence of wild-type human neurotrophic factor. The same applies to neurotrophic factors of animal species other than humans.
  • amino acid residues When adding amino acid residues to the amino acid sequence of wild-type human neurotrophic factor, one or more amino acid residues are added into the amino acid sequence of human neurotrophic factor or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type human neurotrophic factor, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type human neurotrophic factor. The same applies to neurotrophic factors of animal species other than humans.
  • a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues can be added to the amino acid sequence of wild-type human neurotrophic factor.
  • a human neurotrophic factor obtained by deleting 1 to 10 amino acid residues from the amino acid sequence of wild-type human neurotrophic factor, substituting 1 to 10 amino acid residues with other amino acid residues, and adding 1 to 10 amino acid residues is also a human neurotrophic factor
  • a human neurotrophic factor obtained by deleting 1 to 5 amino acid residues from the amino acid sequence of wild-type human neurotrophic factor, substituting 1 to 5 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues is also a human neurotrophic factor
  • a human neurotrophic factor obtained by deleting 1 to 3 amino acid residues from the amino acid sequence of wild-type human neurotrophic factor, and adding 1 to 3 amino acid residues is also a human neurotrophic factor.
  • Human neurotrophic factors are those in which an amino acid residue has been replaced with another amino acid residue and one to three amino acid residues have been added. Human neurotrophic factors are also those in which one or two amino acid residues have been deleted from the amino acid sequence of wild-type human neurotrophic factors, one or two amino acid residues have been replaced with another amino acid residue, and one or two amino acid residues have been added. Human neurotrophic factors are also those in which one amino acid residue has been deleted from the amino acid sequence of wild-type human neurotrophic factors, one amino acid residue has been replaced with another amino acid residue, and one amino acid residue has been added. The same applies to neurotrophic factors of animal species other than humans.
  • each mutation in a human neurotrophic factor mutant compared to normal wild-type human neurotrophic factor can be easily confirmed by aligning the amino acid sequences of both human neurotrophic factors.
  • the amino acid sequence of the human neurotrophic factor mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of a normal wild-type human neurotrophic factor.
  • substitution of an amino acid in the amino acid sequence of wild-type human neurotrophic factor with another amino acid is preferably a conservative amino acid substitution.
  • the above wild-type or mutant human neurotrophic factors in which the constituent amino acids are modified with sugar chains are also human neurotrophic factors.
  • the above wild-type or mutant human neurotrophic factors in which the constituent amino acids are modified with phosphate are also human neurotrophic factors.
  • human neurotrophic factors modified with something other than sugar chains and phosphate are also human neurotrophic factors.
  • the above wild-type or mutant human neurotrophic factors in which the side chains of the constituent amino acids have been converted by a substitution reaction or the like are also human neurotrophic factors.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same applies to neurotrophic factors of animal species other than humans.
  • human neurotrophic factors modified with sugar chains are included in human neurotrophic factors with the original amino acid sequence.
  • Human neurotrophic factors modified with phosphate are included in human neurotrophic factors with the original amino acid sequence.
  • Human neurotrophic factors modified with things other than sugar chains and phosphate are also included in human neurotrophic factors with the original amino acid sequence.
  • Human neurotrophic factors in which the side chains of the amino acids that make up human neurotrophic factors have been converted by substitution reactions or the like are also included in human neurotrophic factors with the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same applies to neurotrophic factors of animal species other than humans.
  • human brain-derived neurotrophic factor when simply referring to "human brain-derived neurotrophic factor,” “human BDNF,” or “hBDNF,” it includes not only the normal wild-type hBDNF consisting of 119 amino acid residues as shown in SEQ ID NO: 60, but also includes, without any particular distinction, mutants of hBDNF in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, “addition” of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence as shown in SEQ ID NO: 60, as long as they have the functions of hBDNF, such as binding to the specific receptor TrkB on the surface of target cells and having physiological activity as a neurotrophic factor that promotes the development, growth, maintenance, and regeneration of nerve cells.
  • Wild-type hBDNF is, for example, encoded by a gene having the base sequence as shown in SEQ ID NO: 61. The same applies to BDNF of animal species other than humans.
  • hBDNF When hBDNF is said to have the function of hBDNF, it means that hBDNF preferably retains a specific activity of 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, of the specific activity of normal wild-type hBDNF taken as 100%.
  • specific activity refers to the physiological activity per mass of protein.
  • the specific activity of a fusion protein of hBDNF and another protein is determined as the physiological activity per mass of the portion of the fusion protein that corresponds to hBDNF.
  • the specific activity of hBDNF in the fusion protein is calculated by multiplying the physiological activity of hBDNF per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hBDNF). The same applies to BDNF of animal species other than humans.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • hBDNF mutants consisting of 118 amino acid residues obtained by deleting one amino acid residue at the N-terminus or C-terminus of wild-type hBDNF, and hBDNF mutants consisting of 117 amino acid residues obtained by deleting two amino acid residues at the N-terminus or C-terminus of wild-type hBDNF are also hBDNF. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hBDNF. The same applies to BDNF of animal species other than humans.
  • amino acid residues When amino acid residues are added to the amino acid sequence of wild-type hBDNF, one or more amino acid residues are added into the amino acid sequence of hBDNF or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hBDNF, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hBDNF.
  • a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues can be added to the amino acid sequence of wild-type hBDNF.
  • a wild-type hBDNF shown in SEQ ID NO:60 in which 1 to 10 amino acid residues have been deleted, 1 to 10 amino acid residues have been replaced with other amino acid residues, and 1 to 10 amino acid residues have been added is also hBDNF
  • a wild-type hBDNF shown in SEQ ID NO:60 in which 1 to 5 amino acid residues have been deleted, 1 to 5 amino acid residues have been replaced with other amino acid residues, and 1 to 5 amino acid residues have been added is also hBDNF
  • a wild-type hBDNF shown in SEQ ID NO:60 in which 1 to 3 amino acid residues have been deleted, hBDNF is also obtained by deleting one or two amino acid residues from the amino acid sequence of wild-type hBDNF shown in SEQ ID NO:60, substituting one or two amino acid residues
  • hBDNF is also obtained by deleting one amino acid residue from the amino acid sequence of wild-type hBDNF shown in SEQ ID NO:60, substituting one amino acid residue with other amino acid residues, and adding one amino acid residue.
  • SEQ ID NO:60 amino acid sequence of wild-type hBDNF shown in SEQ ID NO:60
  • substituting one amino acid residue with other amino acid residues substituting one amino acid residue with other amino acid residues.
  • adding one amino acid residue is also obtained by deleting one amino acid residue from the amino acid sequence of wild-type hBDNF shown in SEQ ID NO:60, substituting one amino acid residue with other amino acid residues, and adding one amino acid residue.
  • each mutation in the hBDNF mutant compared to normal wild-type hBDNF can be easily confirmed by aligning the amino acid sequences of both hBDNFs. The same is true for BDNFs of animal species other than humans.
  • the amino acid sequence of the hBDNF mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type hBDNF shown in SEQ ID NO:60.
  • human nerve growth factor when simply referring to "human nerve growth factor”, “human NGF” or “hNGF”, it includes, without distinction, not only the usual wild-type hNGF consisting of the 120 amino acid residues shown in SEQ ID NO: 62, but also mutants of hNGF in which one or more amino acid residues have been substituted, deleted and/or added (in this specification, “addition” of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 62, as long as they have the functions of hNGF, such as the physiological activity as a neurotrophic factor, such as the action of promoting the extension of nerve axons and the synthesis of neurotransmitters, the action of maintaining nerve cells, the action of repairing damaged cells, and the action of promoting the recovery of brain nerve functions and preventing aging.
  • Wild-type hNGF is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 63. The
  • hNGF When hNGF is said to have the function of hNGF, it means that hNGF preferably retains a specific activity of 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, of the specific activity of normal wild-type hNGF taken as 100%.
  • specific activity refers to the physiological activity per mass of protein.
  • the specific activity of a fusion protein of hNGF and another protein is determined as the physiological activity per mass of the portion of the fusion protein that corresponds to hNGF.
  • the specific activity of hNGF in the fusion protein is calculated by multiplying the physiological activity of hNGF per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hNGF). The same applies to NGFs of animal species other than humans.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • hNGF mutants consisting of 119 amino acid residues obtained by deleting one amino acid residue at the N-terminus or C-terminus of wild-type hNGF, and hNGF mutants consisting of 118 amino acid residues obtained by deleting two amino acid residues at the N-terminus or C-terminus of wild-type hNGF, are also hNGF. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hNGF. The same applies to NGFs of animal species other than humans.
  • amino acid residues When amino acid residues are added to the amino acid sequence of wild-type hNGF, one or more amino acid residues are added into the amino acid sequence of hNGF or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hNGF, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hNGF. The same applies to NGFs of animal species other than humans.
  • a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues can be added to the amino acid sequence of wild-type hNGF.
  • a wild-type hNGF shown in SEQ ID NO:62 in which 1 to 10 amino acid residues have been deleted, 1 to 10 amino acid residues have been replaced with other amino acid residues, and 1 to 10 amino acid residues have been added is also an hNGF;
  • a wild-type hNGF shown in SEQ ID NO:62 in which 1 to 5 amino acid residues have been deleted, 1 to 5 amino acid residues have been replaced with other amino acid residues, and 1 to 5 amino acid residues have been added is also an hNGF;
  • a wild-type hNGF shown in SEQ ID NO:62 in which 1 to 3 amino acid residues have been deleted, hNGF is also obtained by deleting one or two amino acid residues from the amino acid sequence of wild-type hNGF shown in SEQ ID NO:62, substituting one or two amino acid
  • hNGF is also obtained by deleting one amino acid residue from the amino acid sequence of wild-type hNGF shown in SEQ ID NO:62, substituting one amino acid residue with other amino acid residues, and adding one amino acid residue.
  • SEQ ID NO:62 the amino acid sequence of wild-type hNGF shown in SEQ ID NO:62
  • substituting one amino acid residue with other amino acid residues substituting one amino acid residue with other amino acid residues.
  • adding one amino acid residue is also obtained by deleting one amino acid residue from the amino acid sequence of wild-type hNGF shown in SEQ ID NO:62, substituting one amino acid residue with other amino acid residues, and adding one amino acid residue. The same applies to NGFs of animal species other than humans.
  • each mutation in the hNGF mutant compared to normal wild-type hNGF can be easily confirmed by aligning the amino acid sequences of both hNGFs. The same is true for NGFs of animal species other than humans.
  • the amino acid sequence of the hNGF mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type hNGF shown in SEQ ID NO:62.
  • human neurotrophin-3 when simply referring to "human neurotrophin-3", “human NT-3”, or “hNT-3”, it includes, without distinction, not only the usual wild-type hNT-3 consisting of the 119 amino acid residues shown in SEQ ID NO: 64, but also mutants of hNT-3 in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, “addition” of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 64, as long as they have the functions of hNT-3, such as having physiological activity as a nerve growth factor that acts via TrkC and is necessary for maintaining the survival and promoting differentiation of nerve cells.
  • Wild-type hNT-3 is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 65. The same applies to hNT-3 of animal species other than humans.
  • hNT-3 When hNT-3 is said to have the function of hNT-3, it means that hNT-3 preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of normal wild-type hNT-3 is taken as 100%.
  • specific activity refers to the physiological activity per mass of the protein.
  • the specific activity of a fusion protein of hNT-3 and another protein is determined as the physiological activity per mass of the portion of the fusion protein that corresponds to hNT-3.
  • the specific activity of hNT-3 in the fusion protein is calculated by multiplying the physiological activity of hNT-3 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hNT-3).
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • hNT-3 mutants consisting of 118 amino acid residues obtained by deleting one amino acid residue at the N-terminus or C-terminus of wild-type hNT-3, and hNT-3 mutants consisting of 117 amino acid residues obtained by deleting two amino acid residues at the N-terminus or C-terminus of wild-type hNT-3, etc. are also hNT-3. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hNT-3. The same applies to hNT-3 of animal species other than humans.
  • amino acid residues When amino acid residues are added to the amino acid sequence of wild-type hNT-3, one or more amino acid residues are added into the amino acid sequence of hNT-3 or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hNT-3, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hNT-3. The same applies to hNT-3 from animal species other than humans.
  • a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues can be added to the amino acid sequence of wild-type hNT-3.
  • a wild-type hNT-3 shown in SEQ ID NO:64 has 1 to 10 amino acid residues deleted, 1 to 10 amino acid residues replaced with other amino acid residues, and 1 to 10 amino acid residues added to it, which is also an hNT-3;
  • a wild-type hNT-3 shown in SEQ ID NO:64 has 1 to 5 amino acid residues deleted, 1 to 5 amino acid residues replaced with other amino acid residues, and 1 to 5 amino acid residues added to it, which is also an hNT-3;
  • a wild-type hNT-3 shown in SEQ ID NO:64 has 1 to 3 amino acid residues deleted, 1 to 10 amino acid residues replaced with other amino acid residues, and 1 to 10 amino acid residues added to it, which is also an hNT-3.
  • hNT-3 also includes a wild-type hNT-3 amino acid sequence shown in SEQ ID NO:64 in which one or two amino acid residues have been deleted, one or two amino acid residues have been replaced with other amino acid residues, and one or two amino acid residues have been added; and hNT-3 also includes a wild-type hNT-3 amino acid sequence shown in SEQ ID NO:64 in which one amino acid residue has been deleted, one amino acid residue has been replaced with other amino acid residue, and one amino acid residue has been added. The same applies to hNT-3 from animal species other than humans.
  • the amino acid sequence of the hNT-3 mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of the normal wild-type hNT-3 shown in SEQ ID NO:64.
  • human neurotrophin-4 when simply referring to "human neurotrophin-4,” “human NT-4,” or “hNT-4,” it includes, without distinction, not only the usual wild-type hNT-4 consisting of the 130 amino acid residues shown in SEQ ID NO: 66, but also mutants of hNT-4 in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, “addition” of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 66, as long as they have the functions of hNT-4, such as having physiological activity as a nerve growth factor that acts via TrkB and is necessary for maintaining the survival and promoting differentiation of nerve cells.
  • Wild-type hNT-4 is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 67. The same applies to hNT-4 of animal species other than humans.
  • hNT-4 When hNT-4 is said to have the function of hNT-4, it means that hNT-4 preferably retains a specific activity of 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, of the specific activity of normal wild-type hNT-4 taken as 100%.
  • specific activity refers to the physiological activity per mass of protein.
  • the specific activity of a fusion protein of hNT-4 with another protein is determined as the physiological activity per mass of the portion of the fusion protein that corresponds to hNT-4.
  • the specific activity of hNT-4 in the fusion protein is calculated by multiplying the physiological activity of hNT-4 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hNT-4). The same applies to hNT-4 from animal species other than humans.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • hNT-4 mutants consisting of 129 amino acid residues obtained by deleting one amino acid residue at the N-terminus or C-terminus of wild-type hNT-4, and hNT-4 mutants consisting of 128 amino acid residues obtained by deleting two amino acid residues at the N-terminus or C-terminus of wild-type hNT-4, etc. are also hNT-4. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hNT-4. The same applies to hNT-4 of animal species other than humans.
  • amino acid residues When amino acid residues are added to the amino acid sequence of wild-type hNT-4, one or more amino acid residues are added into the amino acid sequence of hNT-4 or to the N-terminus or C-terminus of the amino acid sequence.
  • the number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hNT-4, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hNT-4. The same applies to hNT-4 from animal species other than humans.
  • a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues can be added to the amino acid sequence of wild-type hNT-4.
  • a wild-type hNT-4 amino acid sequence shown in SEQ ID NO:66 in which 1 to 10 amino acid residues have been deleted, 1 to 10 amino acid residues have been replaced with other amino acid residues, and 1 to 10 amino acid residues have been added is also an hNT-4;
  • a wild-type hNT-4 amino acid sequence shown in SEQ ID NO:66 in which 1 to 5 amino acid residues have been deleted, 1 to 5 amino acid residues have been replaced with other amino acid residues, and 1 to 5 amino acid residues have been added is also an hNT-4;
  • a wild-type hNT-4 amino acid sequence shown in SEQ ID NO:66 in which 1 to 3 amino acid residues have been deleted, hNT-4 also includes a wild-type hNT-4 amino acid sequence shown in SEQ ID NO:66 in which one or two amino acid residues have been
  • hNT-4 mutant compared to normal wild-type hNT-4 can be easily confirmed by aligning the amino acid sequences of both hNT-4. The same is true for hNT-4 from animal species other than humans.
  • the amino acid sequence of the hNT-4 mutant preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type hNT-4 shown in SEQ ID NO:66.
  • substitutions of amino acids in the amino acid sequence of wild-type hBDNF or wild-type hNGF, wild-type hNT-3, and wild-type hNT-4 are, for example, conservative amino acid substitutions. The same is true for these neurotrophic factors of animal species other than humans.
  • the above wild-type or mutant hBDNF, hNGF, hNT-3, or hNT-4, in which the constituent amino acids are modified with sugar chains are also hBDNF, hNGF, hNT-3, or hNT-4, respectively.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • hBDNF, hNGF, hNT-3, and hNT-4 The same applies to these neurotrophic factors of animal species other than humans.
  • hBDNF, hNGF, hNT-3, or hNT-4 modified with sugar chains are included in hBDNF, hNGF, hNT-3, or hNT-4 having the original amino acid sequence, respectively.
  • these human neurotrophic factors modified with phosphate The same applies to those modified with things other than sugar chains and phosphate.
  • the same also applies to those in which the side chains of the amino acids that make up these human neurotrophic factors have been changed by substitution reactions, etc.
  • One such conversion is the conversion of a cysteine residue to formylglycine.
  • neurotrophic factors of animal species other than humans are examples of neurotrophic factors of animal species other than humans.
  • One embodiment of the present invention is a recombinant protein in which a wild-type neurotrophic factor is fused with HSA.
  • a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium
  • the amount of expression of human neurotrophic factor in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc., compared to when wild-type human neurotrophic factor is expressed as a recombinant protein by a similar method.
  • Another embodiment of the present invention is a recombinant protein in which wild-type hBDNF is fused with HSA.
  • a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium
  • the amount of hBDNF expressed in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc., compared to when wild-type hBDNF is expressed as a recombinant protein by a similar method.
  • Another embodiment of the present invention is a recombinant protein in which wild-type hNGF is fused with HSA.
  • a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium
  • the amount of hNGF expressed in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc., compared to when wild-type hNGF is expressed as a recombinant protein by a similar method.
  • Another embodiment of the present invention is a recombinant protein in which wild-type hNT-3 is fused with HSA.
  • a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium
  • the amount of expression of hNT-3 in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc., compared to when wild-type hNT-3 is expressed as a recombinant protein by a similar method.
  • Another embodiment of the present invention is a recombinant protein in which wild-type hNT-4 is fused with HSA.
  • a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium
  • the amount of hNT-4 expressed in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc., compared to when wild-type hNT-4 is expressed as a recombinant protein by a similar method.
  • One embodiment of the present invention relates to a fusion protein in which a polypeptide containing the amino acid sequence of a human neurotrophic factor is bound to a polypeptide containing the amino acid sequence of SA.
  • binding polypeptides refers to binding different polypeptides by covalent bonding, either directly or indirectly via a linker.
  • the neurotrophic factor is preferably human neurotrophic factor.
  • SA is preferably HSA.
  • the human neurotrophic factor is, for example, hBDNF, hNGF, hNT-3, or hNT-4. The same applies to neurotrophic factors of animal species other than humans.
  • a common method for linking two different polypeptides is, for example, to create an expression vector incorporating a DNA fragment in which a gene encoding one polypeptide is linked in-frame downstream of a gene encoding the other polypeptide, and then to express the polypeptide as a recombinant protein by culturing a host cell transformed with this expression vector.
  • the resulting recombinant protein is a single-chain polypeptide in which the two polypeptides are peptide-linked, either directly or via different amino acid sequences.
  • SA-human neurotrophic factor fusion protein or "SA-human neurotrophic factor” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of human neurotrophic factor is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and which has the function of a human neurotrophic factor.
  • the animal species of SA is not particularly limited, but preferably it is a mammalian SA, more preferably it is a primate SA, and even more preferably it is HSA.
  • the human neurotrophic factor in the SA-human neurotrophic factor fusion protein has the function of a human neurotrophic factor
  • the human neurotrophic factor preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of a normal wild-type human neurotrophic factor is taken as 100%.
  • the specific activity of human neurotrophic factor in the SA-human neurotrophic factor fusion protein is calculated by multiplying the physiological activity of human neurotrophic factor per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to human neurotrophic factor).
  • the term "HSA-human neurotrophic factor fusion protein" or "HSA-human neurotrophic factor” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of human neurotrophic factor is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of a human neurotrophic factor.
  • HSA-human neurotrophic factor fusion protein has the function of a human neurotrophic factor
  • the definition of the SA-neurotrophic factor fusion protein described above can be applied.
  • the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • the HSA-human neurotrophic factor fusion protein The same is true for the HSA-human neurotrophic factor fusion protein.
  • mutations When mutations are added to an HSA-human neurotrophic factor fusion protein, which is a fusion protein of wild-type HSA and wild-type human neurotrophic factor, mutations can be added only to the HSA portion without mutations in the human neurotrophic factor portion, mutations can be added only to the human neurotrophic factor portion without mutations in the HSA portion, or mutations can be added to both the HSA portion and the human neurotrophic factor portion.
  • the amino acid sequence of the portion is the amino acid sequence of the wild-type HSA with a mutation added.
  • mutations are added only to the human neurotrophic factor portion, the amino acid sequence of the portion is the amino acid sequence of the wild-type human neurotrophic factor with a mutation added.
  • the amino acid sequence of the HSA portion is the amino acid sequence of the wild-type HSA with a mutation added
  • the amino acid sequence of the human neurotrophic factor portion is the amino acid sequence of the wild-type human neurotrophic factor with a mutation added. The same can be said about fusion proteins between wild-type SA of non-human animal species and wild-type human neurotrophic factor.
  • HSA-human neurotrophic factor fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-human neurotrophic factor fusion proteins.
  • HSA-human neurotrophic factor fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-human neurotrophic factor fusion proteins.
  • HSA-human neurotrophic factor fusion proteins modified with something other than sugar chains and phosphate are also HSA-human neurotrophic factor fusion proteins.
  • HSA-human neurotrophic factor fusion proteins in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like are also HSA-human neurotrophic factor fusion proteins.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of SA of animal species other than humans and human neurotrophic factor (SA-human neurotrophic factor).
  • HSA-human neurotrophic factor fusion proteins modified with sugar chains are included in HSA-human neurotrophic factor fusion proteins having the original amino acid sequence.
  • HSA-human neurotrophic factor fusion proteins modified with phosphate are included in HSA-human neurotrophic factor fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are included in HSA-human neurotrophic factor fusion proteins having the original amino acid sequence.
  • HSA-human neurotrophic factor fusion proteins in which the side chains of the amino acids constituting the HSA-human neurotrophic factor fusion proteins have been converted by substitution reactions or the like are included in HSA-human neurotrophic factor fusion proteins having the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and human neurotrophic factor of animal species other than humans (SA-human neurotrophic factor).
  • HSA-human neurotrophic factor fusion protein in which the human neurotrophic factor constituting the protein is a precursor of human neurotrophic factor is also an HSA-human neurotrophic factor fusion protein.
  • precursor used here refers to a type of protein that is biosynthesized as an HSA-human neurotrophic factor fusion protein, and in which the portion that functions as a human neurotrophic factor after the biosynthesis is cleaved from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as a human neurotrophic factor by itself.
  • the HSA-human neurotrophic factor fusion protein may be cleaved at a specific site after biosynthesis by a hydrolase or the like, and the portion containing HSA and the portion containing mature human neurotrophic factor may be separated.
  • the human neurotrophic factor obtained is not a fusion protein with HSA, but the HSA-human neurotrophic factor fusion protein is synthesized once during the process of manufacturing the neurotrophic factor. Therefore, when human neurotrophic factor is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-human neurotrophic factor. The same can be said about fusion proteins of SA from non-human animal species and human neurotrophic factor (SA-human neurotrophic factor).
  • SA-hBDNF fusion protein or "SA-hBDNF” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hBDNF is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and which has the function of hBDNF.
  • the animal species of SA is not particularly limited, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA.
  • hBDNF in an SA-hBDNF fusion protein When hBDNF in an SA-hBDNF fusion protein is said to have the function of hBDNF, it means that hBDNF retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hBDNF is taken as 100%.
  • the specific activity of hBDNF in the SA-hBDNF fusion protein is calculated by multiplying the physiological activity of hBDNF per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hBDNF).
  • the term "HSA-hBDNF fusion protein" or "HSA-hBDNF” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hBDNF is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hBDNF.
  • HSA-hBDNF fusion protein has the function of hBDNF
  • the definition of the SA-hBDNF fusion protein described above can be applied.
  • a preferred HSA-hBDNF fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 68.
  • the HSA-hBDNF fusion protein shown in SEQ ID NO: 68 is obtained by linking wild-type hBDNF to the C-terminus of wild-type HSA via the linker sequence Gly-Ser.
  • the HSA-hBDNF fusion protein shown in SEQ ID NO: 68 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 69.
  • HSA-hBDNF fusion proteins include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 68 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hBDNF. It is preferable that the HSA-hBDNF fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • an HSA-hBDNF fusion protein which is a fusion protein of wild-type HSA and wild-type hBDNF
  • a mutation may be added only to the HSA portion and not to the hBDNF portion, or a mutation may be added only to the hBDNF portion without any mutation to the HSA portion, or a mutation may be added to both the HSA portion and the hBDNF portion.
  • the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hBDNF obtained by adding a mutation to the wild-type hBDNF described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hBDNF portion is the amino acid sequence of hBDNF obtained by adding a mutation to the wild-type hBDNF described above.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:68 or to the N-terminus or C-terminus of the amino acid sequence.
  • the HSA-hBDNF fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA part, only to the hIL-10 part, or to both parts.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:68 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:68.
  • HSA-hBDNF fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hBDNF fusion proteins.
  • HSA-hBDNF fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hBDNF fusion proteins.
  • HSA-hBDNF fusion proteins modified with something other than sugar chains and phosphate are also HSA-hBDNF fusion proteins.
  • HSA-hBDNF fusion proteins in which the side chains of the amino acids constituting the protein have been changed by a substitution reaction or the like are also HSA-hBDNF fusion proteins.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of SA and hBDNF of animal species other than humans (SA-hBDNF).
  • HSA-hBDNF fusion proteins modified with sugar chains are included in HSA-hBDNF fusion proteins with the original amino acid sequence.
  • HSA-hBDNF fusion proteins modified with phosphate are included in HSA-hBDNF fusion proteins with the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are also included in HSA-hBDNF fusion proteins with the original amino acid sequence.
  • those in which the side chains of the amino acids constituting the HSA-hBDNF fusion protein have been converted by substitution reactions or the like are also included in HSA-hBDNF fusion proteins with the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hBDNF of animal species other than humans (SA-hBDNF).
  • HSA-hBDNF fusion proteins in which the constituent hBDNF is a precursor of hBDNF are also HSA-hBDNF fusion proteins.
  • the term "precursor” refers to a type that is biosynthesized as an HSA-hBDNF fusion protein, and after the biosynthesis, the part that functions as hBDNF is separated from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hBDNF by itself.
  • the HSA-hBDNF fusion protein after the HSA-hBDNF fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the part containing HSA and the part containing mature hBDNF may be separated.
  • the obtained hBDNF is not a fusion protein with HSA, but the HSA-hBDNF fusion protein is synthesized once during the process of manufacturing hBDNF. Therefore, when hBDNF is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-hBDNF fusion protein. The same can be said for a fusion protein of SA and hBDNF of an animal species other than human (SA-hBDNF).
  • SA-hNGF fusion protein or "SA-hNGF” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNGF is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and having the function of hNGF.
  • the animal species of SA is not particularly limited, but preferably it is a mammalian SA, more preferably it is a primate SA, and even more preferably it is HSA.
  • hNGF in an SA-hNGF fusion protein has the function of hNGF
  • hNGF retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hNGF is taken as 100%.
  • the specific activity of hNGF in the SA-hNGF fusion protein is calculated by multiplying the physiological activity of hNGF per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hNGF).
  • the term "HSA-hNGF fusion protein" or “HSA-hNGF” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNGF is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hNGF.
  • HSA-hNGF fusion protein has the function of hNGF
  • the definition of the SA-hNGF fusion protein described above can be applied.
  • a preferred HSA-hNGF fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 70.
  • the HSA-hNGF fusion protein shown in SEQ ID NO: 70 is obtained by linking wild-type hNGF to the C-terminus of wild-type HSA via the linker sequence Gly-Ser.
  • the HSA-hNGF fusion protein shown in SEQ ID NO: 70 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 71.
  • HSA-hNGF fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 70 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hNGF. It is preferable that the HSA-hNGF fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • mutations When mutations are introduced into an HSA-hNGF fusion protein, which is a fusion protein of wild-type HSA and wild-type hNGF, mutations can be introduced only in the HSA portion and not in the hNGF portion, mutations can be introduced only in the hNGF portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNGF portion.
  • mutations When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hNGF obtained by adding a mutation to the wild-type hNGF described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hNGF portion is the amino acid sequence of hNGF obtained by adding a mutation to the wild-type hNGF described above.
  • the following is an example of a case where a mutation is made to an HSA-hNGF fusion protein having the amino acid sequence shown in SEQ ID NO:70.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:70 or to the N-terminus or C-terminus of the amino acid sequence.
  • the HSA-hNGF fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hNGF portion, or to both portions. The same applies to a fusion protein of SA and hNGF of an animal species other than human (SA-hNGF).
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:70 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:70.
  • HSA-hNGF fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hNGF fusion proteins.
  • HSA-hNGF fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hNGF fusion proteins.
  • HSA-hNGF fusion proteins modified with anything other than sugar chains and phosphate are also HSA-hNGF fusion proteins.
  • HSA-hNGF fusion proteins in which the side chains of the amino acids constituting the protein are modified by substitution reactions, etc. are also HSA-hNGF fusion proteins.
  • Such modifications include, but are not limited to, the conversion of cysteine residues to formylglycine. Mutations may be made only to the HSA portion, only to the hNGF portion, or to both portions. The same can be said about fusion proteins of SA and hNGF of animal species other than humans (SA-hNGF).
  • HSA-hNGF fusion proteins modified with sugar chains are included in HSA-hNGF fusion proteins having the original amino acid sequence.
  • HSA-hNGF fusion proteins modified with phosphate are included in HSA-hNGF fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are included in HSA-hNGF fusion proteins having the original amino acid sequence.
  • HSA-hNGF fusion proteins in which the side chains of the amino acids constituting the HSA-hNGF fusion protein have been changed by substitution reactions or the like are included in HSA-hNGF fusion proteins having the original amino acid sequence.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. Mutations may be made only to the HSA portion, only to the hNGF portion, or to both portions. The same can be said for fusion proteins of SA and hNGF of animal species other than humans (SA-hNGF).
  • HSA-hNGF fusion proteins in which the hNGF constituting the protein is a precursor of hNGF are also HSA-hNGF fusion proteins.
  • precursor here refers to a type of protein that is biosynthesized as an HSA-hNGF fusion protein, and after the biosynthesis, the portion that functions as hNGF is separated from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hNGF by itself.
  • the HSA-hNGF fusion protein after the HSA-hNGF fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the portion containing HSA and the portion containing mature hNGF may be separated.
  • the resulting hNGF is not a fusion protein with HSA, but the HSA-hNGF fusion protein is synthesized once during the process of manufacturing hNGF. Therefore, when hNGF is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-hNGF fusion protein.
  • the mutation may be added only to the HSA portion, only to the hNGF portion, or both portions. The same can be said about fusion proteins of SA and hNGF from non-human animal species (SA-hNGF).
  • SA-hNT-3 fusion protein or "SA-hNT-3” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNT-3 is linked, directly or via a linker, to the C-terminus of the amino acid sequence of SA, and which has the function of hNT-3.
  • SA animal species of SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA.
  • hNT-3 in the SA-hNT-3 fusion protein has the function of hNT-3
  • hNT-3 preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of normal wild-type hNT-3 is taken as 100%.
  • the specific activity of hNT-3 in the SA-hNT-3 fusion protein is calculated by multiplying the physiological activity of hNT-3 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hNT-3).
  • the term "HSA-hNT-3 fusion protein" or "HSA-hNT-3” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNT-3 is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hNT-3.
  • HSA-hNT-3 fusion protein has the function of hNT-3
  • the definition of the SA-hNT-3 fusion protein described above can be applied.
  • a preferred HSA-hNT-3 fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 72.
  • the HSA-hNT-3 fusion protein shown in SEQ ID NO: 72 is obtained by linking wild-type hNT-3 to the C-terminus of wild-type HSA via the linker sequence Gly-Ser.
  • the HSA-hNT-3 fusion protein shown in SEQ ID NO: 72 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 73.
  • HSA-hNT-3 fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 72 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hNT-3. It is preferable that the HSA-hNT-3 fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • mutations When mutations are introduced into the HSA-hNT-3 fusion protein, which is a fusion protein of wild-type HSA and wild-type hNT-3, mutations can be introduced only in the HSA portion and not in the hNT-3 portion, mutations can be introduced only in the hNT-3 portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNT-3 portion.
  • mutations When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hNT-3 obtained by adding a mutation to the wild-type hNT-3 described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hNT-3 portion is the amino acid sequence of hNT-3 obtained by adding a mutation to the wild-type hNT-3 described above.
  • the HSA-hNT-3 fusion protein having the amino acid sequence shown in SEQ ID NO: 72.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:72 or to the N-terminus or C-terminus of the amino acid sequence.
  • the HSA-hNT-3 fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hGALC portion, or to both portions.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:72 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:72.
  • HSA-hNT-3 fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hNT-3 fusion proteins.
  • HSA-hNT-3 fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hNT-3 fusion proteins.
  • HSA-hNT-3 fusion proteins modified with something other than sugar chains and phosphate are also HSA-hNT-3 fusion proteins.
  • HSA-hNT-3 fusion proteins in which the side chains of the amino acids constituting the protein have been changed by a substitution reaction or the like are also HSA-hNT-3 fusion proteins.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of SA and hNT-3 of animal species other than humans (SA-hNT-3).
  • HSA-hNT-3 fusion proteins modified with sugar chains are included in HSA-hNT-3 fusion proteins having the original amino acid sequence.
  • HSA-hNT-3 fusion proteins modified with phosphate are included in HSA-hNT-3 fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are also included in HSA-hNT-3 fusion proteins having the original amino acid sequence.
  • HSA-hNT-3 fusion proteins in which the side chains of the amino acids constituting the HSA-hNT-3 fusion proteins have been converted by substitution reactions or the like are also included in HSA-hNT-3 fusion proteins having the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hNT-3 of animal species other than humans (SA-hNT-3).
  • HSA-hNT-3 fusion proteins in which the hNT-3 constituting the protein is a precursor of hNT-3 are also HSA-hNT-3 fusion proteins.
  • the term "precursor” here refers to a type of protein that is biosynthesized as an HSA-hNT-3 fusion protein, and in which the portion that functions as hNT-3 after the biosynthesis is cleaved from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hNT-3 by itself.
  • the HSA-hNT-3 fusion protein after the HSA-hNT-3 fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the portion containing HSA and the portion containing mature hNT-3 may be separated.
  • the resulting hNT-3 is not a fusion protein with HSA, but the HSA-hNT-3 fusion protein is synthesized once during the process of synthesizing the hNT-3. Therefore, when hNT-3 is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-hNT-3 fusion protein. The same can be said about the fusion protein of SA and hNT-3 from non-human animal species (SA-hNT-3).
  • SA-hNT-4 fusion protein or "SA-hNT-4" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNT-4 is linked, directly or via a linker, to the C-terminus of the amino acid sequence of SA, and which has the function of hNT-4.
  • SA animal species of SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA.
  • hNT-4 in the SA-hNT-4 fusion protein has the function of hNT-4, it means that hNT-4 preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of normal wild-type hNT-4 is taken as 100%.
  • the specific activity of hNT-4 in the SA-hNT-4 fusion protein is calculated by multiplying the physiological activity of hNT-4 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hNT-4).
  • the term "HSA-hNT-4 fusion protein" or "HSA-hNT-4" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNT-4 is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hNT-4.
  • HSA-hNT-4 fusion protein has the function of hNT-4
  • the definition of the SA-hNT-4 fusion protein above can be applied.
  • a preferred HSA-hNT-4 fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 74.
  • the HSA-hNT-4 fusion protein shown in SEQ ID NO: 74 is obtained by linking wild-type hNT-4 to the C-terminus of wild-type HSA via the linker sequence Gly-Ser.
  • the HSA-hNT-4 fusion protein shown in SEQ ID NO: 74 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 75.
  • HSA-hNT-4 fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 74 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hNT-4. It is preferable that the HSA-hNT-4 fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • mutations When mutations are introduced into the HSA-hNT-4 fusion protein, which is a fusion protein of wild-type HSA and wild-type hNT-4, mutations can be introduced only in the HSA portion and not in the hNT-4 portion, mutations can be introduced only in the hNT-4 portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNT-4 portion.
  • mutations When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hNT-4 obtained by adding a mutation to the wild-type hNT-4 described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hNT-4 portion is the amino acid sequence of hNT-4 obtained by adding a mutation to the wild-type hNT-4 described above.
  • the HSA-hNT-4 fusion protein having the amino acid sequence shown in SEQ ID NO: 74.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:74 or to the N-terminus or C-terminus of the amino acid sequence.
  • the HSA-hNT-4 fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hNT-4 portion, or to both portions. The same can be said about the fusion protein (SA-hNT-4) of SA and hNT-3 from non-human animal species.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:74 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably shows 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:74.
  • HSA-hNT-4 fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hNT-4 fusion proteins.
  • HSA-hNT-4 fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hNT-4 fusion proteins.
  • HSA-hNT-4 fusion proteins modified with something other than sugar chains and phosphate are also HSA-hNT-4 fusion proteins.
  • HSA-hNT-4 fusion proteins in which the side chains of the amino acids constituting the protein have been changed by a substitution reaction or the like are also HSA-hNT-4 fusion proteins.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins (SA-hNT-4) of SA and hNT-3 of animal species other than humans.
  • HSA-hNT-4 fusion proteins modified with sugar chains are included in HSA-hNT-4 fusion proteins having the original amino acid sequence.
  • HSA-hNT-4 fusion proteins modified with phosphate are included in HSA-hNT-4 fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are also included in HSA-hNT-4 fusion proteins having the original amino acid sequence.
  • HSA-hNT-4 fusion proteins in which the side chains of the amino acids constituting the HSA-hNT-4 fusion protein have been converted by substitution reactions or the like are also included in HSA-hNT-4 fusion proteins having the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hNT-3 of animal species other than humans (SA-hNT-4).
  • HSA-hNT-4 fusion proteins in which the constituent hNT-4 is a precursor of hNT-4 are also HSA-hNT-4 fusion proteins.
  • precursor refers to a type of protein that is biosynthesized as an HSA-hNT-4 fusion protein, and in which the portion that functions as hNT-4 after the biosynthesis is cleaved from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hNT-4 by itself.
  • the HSA-hNT-4 fusion protein after the HSA-hNT-4 fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the portion containing HSA and the portion containing hNT-4 may be separated.
  • the resulting hNT-4 is not a fusion protein with HSA, but the HSA-hNT-4 fusion protein is synthesized once during the process of synthesizing the hNT-4. Therefore, when hNT-4 is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-hNT-4 fusion protein. The same can be said about the fusion protein of SA and hNT-4 from non-human animal species (SA-hNT-4).
  • human neurotrophic factor-SA fusion protein or “human neurotrophic factor-SA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked directly or via a linker to the C-terminus of the amino acid sequence of human neurotrophic factor, and which has the function of human neurotrophic factor.
  • the human neurotrophic factor in the human neurotrophic factor-SA fusion protein is said to have the function of human neurotrophic factor, it means that the human neurotrophic factor retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of a normal wild-type human neurotrophic factor is taken as 100%.
  • the specific activity of human neurotrophic factor in the human neurotrophic factor-SA fusion protein is calculated by multiplying the physiological activity of human neurotrophic factor per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to human neurotrophic factor).
  • human neurotrophic factor-HSA fusion protein or "human neurotrophic factor-HSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of human neurotrophic factor, and which has the function of a human neurotrophic factor.
  • the definition of the neurotrophic factor-HSA fusion protein described above can be applied.
  • the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • human neurotrophic factor-HSA fusion protein it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
  • human neurotrophic factor-HSA fusion protein The same is true for hBDNF-HSA fusion protein, hNGF-HSA fusion protein, hNT-3-HSA fusion protein, and hNT-4-HSA fusion protein.
  • a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of a neurotrophic factor of a non-human animal species, and which has a function as a neurotrophic factor is referred to as a "(non-human animal species) neurotrophic factor-(non-human animal species) SA fusion protein.”
  • a fusion protein of mouse neurotrophic factor and mouse SA is referred to as a "mouse neurotrophic factor-MSA fusion protein" or “mouse neurotrophic factor-MSA.”
  • the definition of the human neurotrophic factor-SA fusion protein described above can be applied.
  • the SA has a function as an SA, such as a function to bind and transport endogenous substances in the blood and
  • a mutation When a mutation is added to a human neurotrophic factor-HSA fusion protein, which is a fusion protein of wild-type human neurotrophic factor and wild-type HSA, a mutation can be added only to the human neurotrophic factor portion and not to the HSA portion, a mutation can be added only to the HSA portion without adding a mutation to the human neurotrophic factor portion, or a mutation can be added to both the human neurotrophic factor portion and the HSA portion.
  • the amino acid sequence of that portion is the amino acid sequence of the human neurotrophic factor obtained by adding a mutation to the wild-type human neurotrophic factor described above.
  • the amino acid sequence of that portion is the amino acid sequence of the HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the human neurotrophic factor portion is the amino acid sequence of the human neurotrophic factor obtained by adding a mutation to the wild-type human neurotrophic factor described above
  • the amino acid sequence of the HSA portion is the amino acid sequence of the HSA obtained by adding a mutation to the wild-type HSA described above. The same can be said about fusion proteins between wild-type human neurotrophic factor and wild-type SA of non-human animal species.
  • Human neurotrophic factor-HSA fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also human neurotrophic factor-HSA fusion proteins.
  • Human neurotrophic factor-HSA fusion proteins in which the amino acids constituting the protein are modified with phosphate are also human neurotrophic factor-HSA fusion proteins.
  • Human neurotrophic factor-HSA fusion proteins modified with something other than sugar chains and phosphate are also human neurotrophic factor-HSA fusion proteins.
  • Human neurotrophic factor-HSA fusion proteins in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like are also human neurotrophic factor-HSA fusion proteins.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of human neurotrophic factor and SA of animal species other than human (human neurotrophic factor-SA).
  • a human neurotrophic factor-HSA fusion protein modified by a sugar chain is included in the human neurotrophic factor-HSA fusion protein having the original amino acid sequence.
  • a human neurotrophic factor-HSA fusion protein modified by phosphate is included in the human neurotrophic factor-HSA fusion protein having the original amino acid sequence.
  • a human neurotrophic factor-HSA fusion protein modified by something other than a sugar chain or phosphate is also included in the human neurotrophic factor-HSA fusion protein having the original amino acid sequence.
  • a human neurotrophic factor-HSA fusion protein in which the side chains of the amino acids constituting the human neurotrophic factor-HSA fusion protein have been converted by a substitution reaction or the like is also included in the human neurotrophic factor-HSA fusion protein having the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about a fusion protein of human neurotrophic factor and SA of an animal species other than human (human neurotrophic factor-SA).
  • a human neurotrophic factor-HSA fusion protein in which the human neurotrophic factor that constitutes it is a precursor of human neurotrophic factor is also a human neurotrophic factor-HSA fusion protein.
  • the precursor of human neurotrophic factor does not exhibit the function of human neurotrophic factor, it is necessary that it is processed in vivo or in vitro and converted into one that exhibits the function of human neurotrophic factor.
  • hBDNF-SA fusion protein or "hBDNF-SA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hBDNF, and which has the function of hBDNF.
  • hBDNF in an hBDNF-SA fusion protein is said to have the function of hBDNF, it means that hBDNF retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hBDNF is taken as 100%.
  • the specific activity of hBDNF in an hBDNF-SA fusion protein is calculated by multiplying the physiological activity of hBDNF per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hBDNF).
  • hBDNF-HSA fusion protein or "hBDNF-HSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hBDNF, and which has the function of hBDNF.
  • the definition of the hBDNF-SA fusion protein described above can be applied.
  • a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of BDNF of a non-human animal species, and which has the function of BDNF is referred to as a "(non-human animal species) BDNF-(non-human animal species) SA fusion protein.”
  • a fusion protein of mouse BDNF and mouse SA is referred to as a "mouse BDNF-mouse SA fusion protein" or "mouse BDNF-MSA.”
  • a preferred hBDNF-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 76.
  • the hBDNF-HSA fusion protein shown in SEQ ID NO: 76 is obtained by linking wild-type HSA to the C-terminus of wild-type hBDNF via the linker sequence Gly-Ser.
  • the hBDNF-HSA fusion protein shown in SEQ ID NO: 76 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 77.
  • the hBDNF-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 76 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as it has the function of hBDNF.
  • the hBDNF-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:76 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:76.
  • mutations When mutations are added to an hBDNF-HSA fusion protein, which is a fusion protein of wild-type hBDNF and wild-type HSA, mutations can be added only to the HSA portion and not to the hBDNF portion, mutations can be added only to the hBDNF portion without mutations to the HSA portion, or mutations can be added to both the HSA portion and the hBDNF portion.
  • mutations When mutations are added only to the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hBDNF obtained by adding a mutation to the wild-type hBDNF described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hBDNF portion is the amino acid sequence of hBDNF obtained by adding a mutation to the wild-type hBDNF described above.
  • the following is an example of a case where a mutation is made to an hBDNF-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:76.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:76 or to the N-terminus or C-terminus of the amino acid sequence.
  • the hBDNF-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hBDNF portion, or to both portions. The same applies to a fusion protein of hBDNF and SA of an animal species other than human (hBDNF-SA).
  • An hBDNF-HSA fusion protein in which the constituent amino acids are modified with sugar chains is also an hBDNF-HSA fusion protein.
  • An hBDNF-HSA fusion protein in which the constituent amino acids are modified with phosphate is also an hBDNF-HSA fusion protein.
  • An hBDNF-HSA fusion protein modified with something other than sugar chains and phosphate is also an hBDNF-HSA fusion protein.
  • An hBDNF-HSA fusion protein in which the side chains of the constituent amino acids have been converted by a substitution reaction or the like is also an hBDNF-HSA fusion protein.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about a fusion protein of hBDNF and SA of an animal species other than human (hBDNF-SA).
  • hBDNF-HSA fusion proteins modified with sugar chains are included in hBDNF-HSA fusion proteins having the original amino acid sequence.
  • hBDNF-HSA fusion proteins modified with phosphate are included in hBDNF-HSA fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are also included in hBDNF-HSA fusion proteins having the original amino acid sequence.
  • hBDNF-HSA fusion proteins in which the side chains of the amino acids constituting the hBDNF-HSA fusion protein have been changed by substitution reactions or the like are also included in hBDNF-HSA fusion proteins having the original amino acid sequence.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of hBDNF and SA of animal species other than human (hBDNF-SA).
  • hBDNF-HSA fusion protein in which the hBDNF that constitutes it is a precursor of hBDNF is also an hBDNF-HSA fusion protein.
  • the hBDNF-HSA fusion protein in which hBDNF is a precursor does not exhibit the functions of hBDNF, it is necessary that it be processed in vivo or in vitro and converted into one that exhibits the functions of hBDNF.
  • hNGF-SA fusion protein or "hNGF-SA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked to the C-terminus of the amino acid sequence of hNGF, either directly or via a linker, and which has the function of hNGF.
  • hNGF in an hNGF-SA fusion protein is said to have the function of hNGF, it means that hNGF retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hNGF is taken as 100%.
  • the specific activity of hNGF in an hNGF-SA fusion protein is calculated by multiplying the physiological activity of hNGF per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hNGF).
  • hNGF-HSA fusion protein or "hNGF-HSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hNGF, and which has the function of hNGF.
  • the definition of the hNGF-SA fusion protein described above can be applied.
  • a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of NGF of a non-human animal species, and which has the function of NGF is referred to as a "(non-human animal species) NGF-(non-human animal species) SA fusion protein.”
  • a fusion protein of mouse NGF and mouse SA is referred to as a "mouse NGF-mouse SA fusion protein" or "mouse NGF-MSA.”
  • a preferred hNGF-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 78.
  • the hNGF-HSA fusion protein shown in SEQ ID NO: 78 is obtained by linking wild-type HSA to the C-terminus of wild-type hNGF via the linker sequence Gly-Ser.
  • the hNGF-HSA fusion protein shown in SEQ ID NO: 78 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 79.
  • the hNGF-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 78 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as it has the function of hNGF.
  • the hNGF-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:78 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:78.
  • mutations When mutations are introduced into an hNGF-HSA fusion protein, which is a fusion protein of wild-type hNGF and wild-type HSA, mutations can be introduced only in the HSA portion and not in the hNGF portion, mutations can be introduced only in the hNGF portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNGF portion.
  • mutations When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hNGF obtained by adding a mutation to the wild-type hNGF described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hNGF portion is the amino acid sequence of hNGF obtained by adding a mutation to the wild-type hNGF described above.
  • the following is an example of a case where a mutation is made to an hNGF-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:78.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:78 or to the N-terminus or C-terminus of the amino acid sequence.
  • the hNGF-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hNGF portion, or to both portions. The same can be said for fusion proteins of non-human animal species' NGF and HSA (NGF-SA), and fusion proteins of non-human animal species' NGF and non-human animal species' SA.
  • An hNGF-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also an hNGF-HSA fusion protein.
  • An hNGF-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also an hNGF-HSA fusion protein.
  • An hNGF-HSA fusion protein modified with something other than sugar chains and phosphate is also an hNGF-HSA fusion protein.
  • An hNGF-HSA fusion protein in which the side chains of the amino acids constituting the protein have been changed by a substitution reaction or the like is also an hNGF-HSA fusion protein.
  • Such a change includes, but is not limited to, the change of a cysteine residue to formylglycine. The same can be said about a fusion protein (hNGF-SA) between hNGF and SA of an animal species other than human.
  • hNGF-HSA fusion proteins modified with sugar chains are included in the hNGF-HSA fusion proteins having the original amino acid sequence.
  • hNGF-HSA fusion proteins modified with phosphate are included in the hNGF-HSA fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are included in the hNGF-HSA fusion proteins having the original amino acid sequence.
  • hNGF-HSA fusion proteins in which the side chains of the amino acids constituting the hNGF-HSA fusion proteins have been changed by substitution reactions or the like are included in the hNGF-HSA fusion proteins having the original amino acid sequence.
  • Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of hNGF and SA of animals other than humans (hNGF-SA).
  • hNGF-HSA fusion protein in which the hNGF that constitutes it is a precursor of hNGF is also an hNGF-HSA fusion protein.
  • the hNGF-HSA fusion protein, in which hNGF is a precursor does not function as hNGF, it is necessary that it be processed in vivo or in vitro and converted into one that functions as hNGF. The same can be said for a fusion protein of hNGF and SA of an animal species other than human (hNGF-SA).
  • hNT-3-SA fusion protein or "hNT-3-SA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hNT-3, and which has the function of hNT-3.
  • hNT-3 in an hNT-3-SA fusion protein is said to have the function of hNT-3, it means that hNT-3 retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hNT-3 is taken as 100%.
  • the specific activity of hNT-3 in the hNT-3-SA fusion protein is calculated by multiplying the physiological activity of hNT-3 per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hNT-3).
  • hNT-3-HSA fusion protein or "hNT-3-HSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hNT-3, and which has the function of hNT-3.
  • the definition of the hNT-3-SA fusion protein described above can be applied.
  • a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of NT-3 of a non-human animal species, and which has the function of NT-3 is referred to as a "(non-human animal species) NT-3-(non-human animal species) SA fusion protein.”
  • a fusion protein of mouse NT-3 and mouse SA is referred to as a "mouse NT-3-mouse SA fusion protein" or "mouse NT-3-MSA.”
  • a preferred hNT-3-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 80.
  • the hNT-3-HSA fusion protein shown in SEQ ID NO: 80 is obtained by linking wild-type HSA to the C-terminus of wild-type hNT-3 via the linker sequence Gly-Ser.
  • the hNT-3-HSA fusion protein shown in SEQ ID NO: 80 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 81.
  • the hNT-3-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 80 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as it has the function of hNT-3.
  • the hNT-3-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:80 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:80.
  • mutations When mutations are introduced into the hNT-3-HSA fusion protein, which is a fusion protein of wild-type hNT-3 and wild-type HSA, mutations can be introduced only in the HSA portion and not in the hNT-3 portion, mutations can be introduced only in the hNT-3 portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNT-3 portion.
  • mutations When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hNT-3 obtained by adding a mutation to the wild-type hNT-3 described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hNT-3 portion is the amino acid sequence of hNT-3 obtained by adding a mutation to the wild-type hNT-3 described above.
  • the hNT-3-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 80.
  • the same is true for the fusion protein of wild-type hNT-3 and wild-type SA (hNT-3-SA) of an animal species other than human.
  • the following is an example of a case where a mutation is made to an hNT-3-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:80.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:80 or to the N-terminus or C-terminus of the amino acid sequence.
  • the hNT-3-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hNT-3 portion, or to both portions. The same can be said about the fusion protein (hNT-3-SA) between hNT-3 and SA of a non-human animal species.
  • An hNT-3-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also an hNT-3-HSA fusion protein.
  • An hNT-3-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also an hNT-3-HSA fusion protein.
  • An hNT-3-HSA fusion protein modified with something other than sugar chains and phosphate is also an hNT-3-HSA fusion protein.
  • An hNT-3-HSA fusion protein in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like is also an hNT-3-HSA fusion protein.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about a fusion protein (hNT-3-SA) of hNT-3 and SA of an animal species other than human.
  • hNT-3-HSA fusion proteins modified with sugar chains are included in the hNT-3-HSA fusion proteins having the original amino acid sequence.
  • hNT-3-HSA fusion proteins modified with phosphate are included in the hNT-3-HSA fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are included in the hNT-3-HSA fusion proteins having the original amino acid sequence.
  • hNT-3-HSA fusion proteins in which the side chains of the amino acids constituting the hNT-3-HSA fusion proteins have been converted by substitution reactions or the like are included in the hNT-3-HSA fusion proteins having the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of hNT-3 and SA of animal species other than human (hNT-3-SA).
  • hNT-3-HSA fusion protein in which the hNT-3 that constitutes it is a precursor of hNT-3 is also an hNT-3-HSA fusion protein.
  • the hNT-3-HSA fusion protein, of which hNT-3 is a precursor does not function as hNT-3, it is necessary that it be processed in vivo or in vitro and converted into one that functions as hNT-3. The same can be said for a fusion protein of hNT-3 and SA of an animal species other than human (hNT-3-SA).
  • hNT-4-SA fusion protein or "hNT-4-SA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hNT-4, and which has the function of hNT-4.
  • hNT-4 in an hNT-4-SA fusion protein is said to have the function of hNT-4, it means that hNT-4 retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hNT-4 is taken as 100%.
  • the specific activity of hNT-4 in the hNT-4-SA fusion protein is calculated by multiplying the physiological activity of hNT-4 per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hNT-4).
  • hNT-4-HSA fusion protein or "hNT-4-HSA” refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hNT-4, and which has the function of hNT-4.
  • the definition of the hNT-4-SA fusion protein described above can be applied.
  • a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of NT-4 of a non-human animal species, and which has the function of NT-4 is referred to as a "(non-human animal species) NT-4-(non-human animal species) SA fusion protein.”
  • a fusion protein of mouse NT-4 and mouse SA is referred to as a "mouse NT-4-mouse SA fusion protein" or "mouse NT-4-MSA.”
  • a preferred hNT-4-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 82.
  • the hNT-4-HSA fusion protein shown in SEQ ID NO: 82 is obtained by linking wild-type HSA to the C-terminus of wild-type hNT-4 via the linker sequence Gly-Ser.
  • the hNT-4-HSA fusion protein shown in SEQ ID NO: 82 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 83.
  • the hNT-4-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 82 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as it has the function of hNT-4.
  • the hNT-4-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
  • the position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:82 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation.
  • the mutated amino acid sequence preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:82.
  • mutations When mutations are introduced into the hNT-4-HSA fusion protein, which is a fusion protein of wild-type hNT-4 and wild-type HSA, mutations can be introduced only in the HSA portion and not in the hNT-4 portion, mutations can be introduced only in the hNT-4 portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNT-4 portion.
  • mutations When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above.
  • the amino acid sequence of the portion is the amino acid sequence of hNT-4 obtained by adding a mutation to the wild-type hNT-4 described above.
  • the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above
  • the amino acid sequence of the hNT-4 portion is the amino acid sequence of hNT-4 obtained by adding a mutation to the wild-type hNT-4 described above.
  • the hNT-4-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 82.
  • the same is true for the fusion protein of wild-type hNT-4 and wild-type SA (hNT-4-SA) of an animal species other than human.
  • the following is an example of a case where a mutation is made to an hNT-4-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:82.
  • the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2.
  • an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:82 or to the N-terminus or C-terminus of the amino acid sequence.
  • the hNT-4-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues.
  • the mutation may be made only to the HSA portion, only to the hNT-4 portion, or to both portions. The same can be said about the fusion protein of hNT-4 and SA of a non-human animal species (hNT-4-SA).
  • An hNT-4-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also an hNT-4-HSA fusion protein.
  • An hNT-4-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also an hNT-4-HSA fusion protein.
  • An hNT-4-HSA fusion protein in which the amino acids constituting the protein are modified with something other than sugar chains and phosphate is also an hNT-4-HSA fusion protein.
  • An hNT-4-HSA fusion protein in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like is also an hNT-4-HSA fusion protein.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for a fusion protein of NT-4 of a non-human animal species and HSA (NT-4-SA), and a fusion protein of NT-4 of a non-human animal species and SA of a non-human animal species.
  • hNT-4-HSA fusion proteins modified with sugar chains are included in the hNT-4-HSA fusion proteins having the original amino acid sequence.
  • hNT-4-HSA fusion proteins modified with phosphate are included in the hNT-4-HSA fusion proteins having the original amino acid sequence.
  • those modified with things other than sugar chains and phosphate are included in the hNT-4-HSA fusion proteins having the original amino acid sequence.
  • those in which the side chains of the amino acids constituting the hNT-4-HSA fusion protein have been converted by a substitution reaction or the like are included in the hNT-4-HSA fusion protein having the original amino acid sequence.
  • Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine.
  • NT-4-SA non-human animal species and HSA
  • NT-4-SA fusion protein of NT-4 of a non-human animal species and SA of a non-human animal species.
  • hNT-4-HSA fusion protein in which the hNT-4 that constitutes it is a precursor of hNT-4 is also an hNT-4-HSA fusion protein.
  • the hNT-4-HSA fusion protein, of which hNT-4 is a precursor does not exhibit the function of hNT-4, it is necessary that it be processed in vivo or in vitro and converted into one that exhibits the function of hNT-4.
  • fusion protein of HSA and human neurotrophic factor include both the above-mentioned “HSA-human neurotrophic factor fusion protein” and “human neurotrophic factor-HSA fusion protein”. These also apply when SA is from an animal species other than human, or when nerve growth factor is from an animal species other than human.
  • a fusion protein of HSA and hBDNF we include both the above-mentioned “HSA-hBDNF fusion protein” and "hBDNF-HSA fusion protein.”
  • a fusion protein of HSA and hNGF we include both the above-mentioned “HSA-hNGF fusion protein” and "hNGF-HSA fusion protein.”
  • a fusion protein of HSA and hNT-3 we include both the above-mentioned “HSA-hNT-3 fusion protein” and "hNT-3-HSA fusion protein.”
  • SA is of an animal species other than human
  • NT-3 is of an animal species other than human.
  • a fusion protein of HSA and hNT-4 we include both the above-mentioned “HSA-hNT-4 fusion protein” and "hNT-4-HSA fusion protein.” These also apply when SA is from an animal species other than human, or when NT-4 is from an animal species other than human.
  • the SA and the neurotrophic factor are linked directly or via a linker.
  • linker refers to a portion that does not belong to either the amino acid sequence of SA or the amino acid sequence of a neurotrophic factor.
  • the linker is a peptide chain that is interposed between the SA and the neurotrophic factor.
  • the linker has various functions.
  • These functions include a function between the SA and the neurotrophic factor to bind the SA and the neurotrophic factor, a function to reduce mutual interference between the SA and the neurotrophic factor by increasing the distance between them in the fusion protein molecule, and a function between the SA and the neurotrophic factor to act as a hinge that connects the SA and the neurotrophic factor and gives flexibility to the three-dimensional structure of the fusion protein.
  • the linker exerts at least one of these functions.
  • fusion proteins of SA and neurotrophic factors are described in detail, taking as examples a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, and a fusion protein of HSA and hNT-4.
  • the matters described in detail here can be applied when SA is from an animal species other than human, or when BDNF, NGF, NT-3, or NT-4 is from an animal species other than human. They can also be applied to neurotrophic factors other than BDNF, NGF, NT-3, or NT-4.
  • a fusion protein of HSA and hBDNF can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hBDNF is linked in-frame downstream or upstream of a gene encoding HSA, and culturing a host cell transformed with the expression vector.
  • a fusion protein of HSA and hNGF can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hNGF is linked in-frame downstream or upstream of a gene encoding HSA, and culturing a host cell transformed with the expression vector.
  • a fusion protein of HSA and hNT-3 can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hNT-3 is linked in-frame downstream or upstream of a gene encoding HSA, and culturing a host cell transformed with the expression vector.
  • a fusion protein of HSA and hNT-4 can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hNT-4 is linked in-frame to the downstream or upstream of a gene encoding HSA, and culturing a host cell transformed with the expression vector.
  • the fusion protein produced as a recombinant protein in this way consists of a single polypeptide chain.
  • a gene encoding hBDNF When preparing a fusion protein as a recombinant fusion protein, a gene encoding hBDNF can be linked in-frame downstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hBDNF at the C-terminus of the amino acid sequence of HSA. Conversely, a gene encoding hBDNF can be linked in-frame upstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hBDNF at the N-terminus of the amino acid sequence of HSA.
  • a gene encoding hNGF When preparing a fusion protein as a recombinant fusion protein, a gene encoding hNGF can be linked in-frame downstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hNGF at the C-terminus of the amino acid sequence of HSA. Conversely, a gene encoding hNGF can be linked in-frame upstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hNGF at the N-terminus of the amino acid sequence of HSA.
  • a gene encoding hNT-3 When preparing a fusion protein as a recombinant fusion protein, a gene encoding hNT-3 can be linked in-frame downstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hNT-3 at the C-terminus of the amino acid sequence of HSA. Conversely, by linking a gene encoding hNT-3 in frame upstream of the gene encoding HSA, a fusion protein having the amino acid sequence of hNT-3 at the N-terminus of the amino acid sequence of HSA can be obtained.
  • a fusion protein produced as a recombinant fusion protein is a single-chain polypeptide.
  • the amino acid sequence of HSA when the amino acid sequence of HSA is located on the N-terminal side of the amino acid sequence of hBDNF, hNGF, hNT-3 or hNT-4, the C-terminus of HSA and the N-terminus of hBDNF, hNGF, hNT-3 or hNT-4 are linked directly by a peptide bond or via a linker.
  • the "linker” refers to a portion of the single-chain polypeptide that is present between the C-terminus of HSA and the N-terminus of hBDNF, hNGF, hNT-3 or hNT-4 and has an amino acid sequence that does not belong to HSA, hBDNF, hNGF, hNT-3 or hNT-4.
  • Figure 7 shows a schematic diagram of a single-chain polypeptide HSA-hBDNF fusion protein that has HSA, a linker and hBDNF in this order from the N-terminus.
  • FIG. 8 shows a schematic diagram of a single-chain polypeptide HSA-hNGF fusion protein having HSA, a linker, and hNGF in this order from the N-terminus.
  • the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond
  • the C-terminus of the linker is bound to the N-terminus of hNGF by a peptide bond.
  • FIG. 9 shows a schematic diagram of a single-chain polypeptide HSA-hNT-3 fusion protein having HSA, a linker, and hNT-3 in this order from the N-terminus.
  • the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond
  • the C-terminus of the linker is bound to the N-terminus of hNT-3 by a peptide bond ...
  • FIG. 10 shows a schematic diagram of a single-chain polypeptide HSA-hNT-4 fusion protein having HSA, a linker, and hNT-4 in this order from the N-terminus.
  • the HSA-hNT-4 fusion protein is composed of the C-terminus of HSA and the N-terminus of the linker bound by a peptide bond, and the C-terminus of the linker and the N-terminus of hNT-4 bound by a peptide bond.
  • the amino acid sequence of hBDNF, hNGF, hNT-3 or hNT-4 is located at the N-terminus of the amino acid sequence of HSA in the single-chain polypeptide of the fusion protein, the C-terminus of hBDNF, hNGF, hNT-3 or hNT-4 and the N-terminus of HSA are linked directly by a peptide bond or via a linker.
  • the "linker” refers to a portion of the single-chain polypeptide that is present between the C-terminus of hBDNF, hNGF, hNT-3 or hNT-4 and the N-terminus of HSA and has an amino acid sequence that does not belong to HSA, hBDNF, hNGF, hNT-3 or hNT-4.
  • Figure 11 shows a schematic diagram of a single-chain polypeptide hBDNF-HSA fusion protein having hBDNF, a linker and HSA in that order from the N-terminus.
  • FIG. 12 shows a schematic representation of a single-chain polypeptide hNGF-HSA fusion protein having hNGF, a linker, and HSA in this order from the N-terminus.
  • FIG. 13 shows a schematic representation of a single-chain polypeptide hNT-3-HSA fusion protein having hNT-3, a linker, and HSA in this order from the N-terminus.
  • FIG. 14 shows a schematic diagram of the hNT-4-HSA fusion protein, which is a single-chain polypeptide having, in order from the N-terminus, hNT-4, a linker, and HSA.
  • the C-terminus of hNT-4 and the N-terminus of the linker are bound by a peptide bond
  • the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond.
  • the amino acid sequence of the peptide linker is composed of one or more amino acids. When the peptide linker is composed of multiple amino acids, the number of amino acids is preferably 2 to 50, more preferably 5 to 30, and even more preferably 10 to 25. Suitable examples of peptide linkers include those consisting of Gly-Ser, Gly-Gly-Ser, or amino acid sequences shown in SEQ ID NOs: 9 to 11 (collectively referred to as basic sequences), and those containing these.
  • the peptide linker is one that contains an amino acid sequence in which the basic sequence is repeated 2 to 10 times, one that contains an amino acid sequence in which the basic sequence is repeated 2 to 6 times, or one that contains an amino acid sequence in which the basic sequence is repeated 3 to 5 times.
  • One or more amino acids in these amino acid sequences may be deleted, replaced with other amino acids, added, etc.
  • the number of amino acids to be deleted is preferably 1 or 2.
  • the number of amino acids to be replaced is preferably 1 or 2.
  • the number of amino acids to be added is preferably 1 or 2.
  • the amino acid sequence of the desired linker portion can be obtained by combining these deletions, substitutions, and additions of amino acids.
  • the peptide linker may be composed of one amino acid, and the amino acid that constitutes the linker is, for example, glycine or serine.
  • a fusion protein of HSA and hBDNF refers to a fusion protein characterized in that when expressed in a host cell as a recombinant protein, particularly when expressed so that the recombinant protein is secreted from the cells and accumulated in the culture medium, the amount of hBDNF expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hBDNF is expressed in a host cell as a recombinant protein under the same conditions, such as 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc.
  • the term "fusion protein of HSA and hNGF” refers to a fusion protein characterized in that, when expressed in a host cell as a recombinant protein, in particular when expressed so that the recombinant protein is secreted from the cells and accumulated in the culture medium, the amount of hNGF expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hNGF is expressed in a host cell as a recombinant protein under the same conditions, such as 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc.
  • a fusion protein of HSA and hNT-3 refers to a fusion protein characterized in that, when expressed in a host cell as a recombinant protein, particularly when expressed so that the recombinant protein is secreted from the cells and accumulated in the culture medium, the expression amount of hNT-3 in the culture supernatant is at least 1.1 times or more, 1.2 times or more, 1.5 times or more, 2 times or more, 2.5 times or more, or 3.5 times or more, in terms of concentration or physiological activity, compared to when wild-type hNT-3 is expressed in a host cell as a recombinant protein under the same conditions, (4)
  • a fusion protein of HSA and hNT-4 refers to a fusion protein characterized in that, when expressed in host cells as a recombinant protein, in particular when expressed so that the recombinant protein is secreted from the cells and accumulated in the culture medium, the expression amount of
  • "under the same conditions” means that the expression vector, host cells, culture conditions, etc. are the same.
  • the preferred host cells used in this case are mammalian cells such as CHO cells and NS/0 cells, and particularly CHO cells.
  • Preferred embodiments of the fusion protein of HSA and hBDNF include the following (1) to (4): (1) A peptide having an amino acid sequence shown in SEQ ID NO:68, in which the N-terminus of the amino acid sequence of wild-type hBDNF shown in SEQ ID NO:60 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO:3 via the linker sequence Gly-Ser; (2) Having the amino acid sequence shown in SEQ ID NO: 76, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hBDNF shown in SEQ ID NO: 60 via the linker sequence Gly-Ser.
  • Preferred embodiments of the fusion protein of HSA and hNGF include the following (1) to (4): (1) A peptide having an amino acid sequence shown in SEQ ID NO: 70, in which the N-terminus of the amino acid sequence of wild-type hNGF shown in SEQ ID NO: 62 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via the linker sequence Gly-Ser; (2) Having the amino acid sequence shown in SEQ ID NO: 78, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hNGF shown in SEQ ID NO: 62 via the linker sequence Gly-Ser.
  • preferred embodiments of the fusion protein of HSA and hNT-3 include the following (1) to (4): (1) A peptide having an amino acid sequence shown in SEQ ID NO: 72, in which the N-terminus of the amino acid sequence of wild-type hNT-3 shown in SEQ ID NO: 64 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via the linker sequence Gly-Ser; (2) Having the amino acid sequence shown in SEQ ID NO: 80, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hNT-3 shown in SEQ ID NO: 64 via the linker sequence Gly-Ser.
  • Preferred embodiments of the fusion protein of HSA and hNT-4 include the following (1) to (4): (1) A peptide having an amino acid sequence shown in SEQ ID NO: 74, in which the N-terminus of the amino acid sequence of wild-type hNT-4 shown in SEQ ID NO: 66 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via the linker sequence Gly-Ser; (2) Having the amino acid sequence shown in SEQ ID NO: 82, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hNT-4 shown in SEQ ID NO: 66 via the linker sequence Gly-Ser.
  • the fusion protein of HSA and hBDNF is characterized in that when expressed in a host cell as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hBDNF expressed in the culture supernatant is increased in terms of concentration or physiological activity compared to when wild-type hBDNF is expressed in a host cell as a recombinant protein under the same conditions. Therefore, when produced as a recombinant protein, the fusion protein of HSA and hBDNF can increase production efficiency compared to wild-type hBDNF, thereby reducing production costs.
  • neurotrophic factors such as hBDNF, hNGF, hNT-3, and hNT-4.
  • fusion protein of HSA and hBDNF expressed in a host cell when comparing the expression levels of a fusion protein of HSA and hBDNF expressed in a host cell as a recombinant protein with wild-type hBDNF expressed in a host cell under the same conditions, the comparison is not based on the mass of the expressed protein, but on the number of molecules of the expressed protein or on the hBDNF biological activity.
  • This rule also applies to fusion proteins of HSA and hNGF, fusion proteins of HSA and hNT-3, and fusion proteins of HSA and hNT-4. This rule also applies regardless of the animal species of the SA and nerve growth factor that make up the fusion protein.
  • HSA and hBDNF fusion protein, HSA and hNGF fusion protein, HSA and hNT-3 fusion protein, and HSA and hNT-4 fusion protein can be produced as recombinant proteins by culturing host cells transformed with an expression vector incorporating a gene encoding the fusion protein. In this case, they can be produced using an expression vector, host cells, culture medium, etc. that can be used to produce the above-mentioned HSA and hGALC fusion protein or HSA and hGBA fusion protein.
  • the protein By culturing host cells encoding a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, the protein can be expressed in cells or in the medium. These fusion proteins can be separated from impurities and purified by methods such as column chromatography.
  • the purified fusion protein of HSA and hBDNF, the fusion protein of HSA and hNGF, the fusion protein of HSA and hNT-3, or the fusion protein of HSA and hNT-4 can be used as a pharmaceutical composition.
  • the fusion protein of HSA and hBDNF can be used as a pharmaceutical composition for treating neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, spinal degenerative diseases such as amyotrophic lateral sclerosis, and other diseases such as diabetic neuropathy, cerebral ischemic disease, developmental disorders such as Rett syndrome, schizophrenia, depression, and Rett syndrome.
  • the fusion protein of HSA and hNGF can be used as a pharmaceutical composition for treating neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease.
  • the fusion protein of HSA and hNT-3 and the fusion protein of HSA and hNT-4 can be used as a pharmaceutical composition for treating neurodegenerative diseases.
  • a pharmaceutical composition containing as an active ingredient a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4 can be administered intravenously, intramuscularly, intraperitoneally, subcutaneously, or intracerebroventricularly as an injection.
  • injections can be supplied as lyophilized preparations or aqueous liquid preparations.
  • an aqueous liquid preparation it may be in the form of a vial filled with the preparation, or it can be supplied as a prefilled preparation in which the preparation is filled in a syringe beforehand.
  • a lyophilized preparation it is dissolved in an aqueous medium and reconstituted before use.
  • the fusion protein of HSA and hBDNF, the fusion protein of HSA and hNGF, the fusion protein of HSA and hNT-3, or the fusion protein of HSA and hNT-4 can be further bound to an antibody or a ligand to form a conjugate.
  • the fusion protein of HSA and hBDNF or a ligand, the fusion protein of HSA and hNGF, the fusion protein of HSA and hNT-3, and the fusion protein of HSA and hNT-4 can each be bound to an antibody or a ligand that can specifically bind to a receptor on cerebrovascular endothelial cells.
  • the fusion proteins of HSA and hBDNF, HSA and hNGF, HSA and hNT-3, and HSA and hNT-4 can bind to the receptor on cerebrovascular endothelial cells by being bound to an antibody or a ligand that can specifically bind to the receptor on cerebrovascular endothelial cells.
  • the fusion proteins bound to the receptor on cerebrovascular endothelial cells can pass through the blood-brain barrier (BBB) and reach the tissues of the central nervous system (CNS). Therefore, these fusion proteins can be conjugated with such antibodies or ligands to pass through the blood-brain barrier (BBB) and exert their functions in the central nervous system (CNS).
  • the SA is from an animal species other than human
  • BDNF, NGF, hNT-3, and hNT-4 are from an animal species other than human
  • the neurotrophic factors are neurotrophic factors other than BDNF, NGF, hNT-3, and hNT-4.
  • hBDNF When hBDNF is said to have the function of hBDNF in a conjugate of a fusion protein of HSA and hBDNF with an antibody or ligand, it means that hBDNF has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more, when the specific activity of normal wild-type hBDNF is taken as 100%.
  • the specific activity of hBDNF in the conjugate is calculated by multiplying the physiological activity of hBDNF per unit mass of the conjugate by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hBDNF).
  • hNGF when hNGF is said to have the function of hNGF in a conjugate of a fusion protein of HSA and hNGF with an antibody or ligand, it means that hNGF has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more, when the specific activity of normal wild-type hNGF is taken as 100%.
  • the specific activity of hNGF in the conjugate is calculated by multiplying the physiological activity of hNGF per unit mass of the conjugate by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hNGF).
  • hNT-3 in a conjugate of a fusion protein of HSA and hNT-3 with an antibody or ligand is said to have the function of hNT-3, it means that hNT-3 has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more, when the specific activity of normal wild-type hNT-3 is taken as 100%.
  • the specific activity of hNT-3 in the conjugate is calculated by multiplying the physiological activity of hNT-3 per unit mass of the conjugate by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hNT-3).
  • hNT-4 when hNT-4 is said to have the function of hNT-4 in a conjugate of a fusion protein of HSA and hNT-4 with an antibody or ligand, it means that hNT-4 has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more, when the specific activity of normal wild-type hNT-4 is taken as 100%.
  • the specific activity of hNT-4 in the conjugate is calculated by multiplying the physiological activity of hNT-4 per unit mass of the conjugate by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hNT-4).
  • a conjugate between a fusion protein of HSA and hBDNF and an antibody or ligand is characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hBDNF expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hBDNF is expressed as a recombinant protein in a host cell under the same conditions, such as 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc.
  • a conjugate between a fusion protein of HSA and hNGF and an antibody or ligand is characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hNGF expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hNGF is expressed as a recombinant protein in a host cell under the same conditions.
  • a conjugate between a fusion protein of HSA and hNT-3 and an antibody or ligand is characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hNT-3 in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hNT-3 is expressed as a recombinant protein in a host cell under the same conditions.
  • a conjugate between a fusion protein of HSA and hNT-4 and an antibody or ligand is characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hNT-4 in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hNT-4 is expressed as a recombinant protein in a host cell under the same conditions, and is 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc.
  • Some neurotrophic factors are difficult to produce in large quantities when expressed as recombinant proteins using host cells introduced with an expression vector incorporating a gene encoding the wild-type neurotrophic factor, because the expression level is limited.
  • Examples of such neurotrophic factors include BDNF, NGF, NT-3, and NT-4.
  • One embodiment of the present invention is a recombinant protein in which such a neurotrophic factor that is difficult to produce as a recombinant protein is fused with SA.
  • SA a recombinant protein
  • Such a recombinant protein can be produced in large quantities more easily using host cells introduced with an expression vector incorporating a gene encoding the recombinant protein, compared to the corresponding wild-type neurotrophic factor.
  • there is no particular restriction on the animal species of the neurotrophic factor to be bound to SA but human neurotrophic factor is preferred.
  • antibody primarily refers to human antibodies, mouse antibodies, humanized antibodies, antibodies derived from camelids (including alpacas), chimeric antibodies between human antibodies and antibodies from other mammals, and chimeric antibodies between mouse antibodies and antibodies from other mammals, but is not limited to these as long as it has the property of specifically binding to a specific antigen, and there is also no particular restriction on the animal species of the antibody.
  • human antibody refers to an antibody whose entire protein is encoded by a gene of human origin.
  • human antibodies also include antibodies encoded by genes in which mutations have been added to the original human gene without changing the original amino acid sequence for the purpose of increasing gene expression efficiency, etc.
  • antibodies produced by combining two or more genes encoding human antibodies and replacing a part of a human antibody with a part of another human antibody are also "human antibodies”.
  • Human antibodies have three complementarity determining regions (CDRs) in the light chain and three complementarity determining regions (CDRs) in the heavy chain. The three CDRs in the light chain are called CDR1, CDR2, and CDR3, starting from the N-terminus.
  • the three CDRs in the heavy chain are also called CDR1, CDR2, and CDR3, starting from the N-terminus.
  • Antibodies in which the antigen specificity, affinity, etc. of a human antibody have been modified by replacing the CDR of a certain human antibody with the CDR of another human antibody are also included in human antibodies.
  • the term "human antibody” also includes antibodies in which mutations such as substitution, deletion, and addition have been added to the amino acid sequence of the original antibody by modifying the gene of the original human antibody.
  • the number of amino acids to be replaced is preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3.
  • the number of amino acids to be deleted is preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3.
  • antibodies in which mutations that combine these amino acid substitutions and deletions have been added are also human antibodies.
  • amino acids When amino acids are added, preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3 amino acids are added to the amino acid sequence or the N-terminus or C-terminus of the original antibody.
  • Antibodies in which mutations that combine these amino acid additions, substitutions, and deletions have been added are also human antibodies.
  • the amino acid sequence of the mutated antibody preferably exhibits 80% or more identity to the amino acid sequence of the original antibody, more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
  • the mutations can be added to the variable region of the antibody.
  • the mutations can be added to either the CDR or framework region of the variable region, but are particularly added to the framework region.
  • the term "human-derived gene” includes not only the original human-derived gene, but also genes obtained by modifying it.
  • humanized antibody refers to an antibody in which the amino acid sequence of a part of the variable region (e.g., all or part of the CDR in particular) is derived from a mammal other than human, and the other regions are derived from humans.
  • a humanized antibody can be an antibody produced by replacing the three complementarity determining regions (CDRs) of the light chain and the three complementarity determining regions (CDRs) of the heavy chain that constitute a human antibody with the CDRs of another mammal.
  • the species of the other mammal from which the CDRs are grafted to the appropriate positions of the human antibody are derived is not particularly limited as long as it is a mammal other than human, but is preferably a mouse, rat, rabbit, horse, or a non-human primate, more preferably a mouse or rat, and even more preferably a mouse.
  • an antibody in which the amino acid sequence of the original humanized antibody is modified with the same mutations as those that can be added to the above-mentioned human antibody is also included in the "humanized antibody".
  • chimeric antibody refers to an antibody that is formed by linking fragments of two or more different antibodies derived from two or more different species.
  • a chimeric antibody between a human antibody and an antibody of another mammal is an antibody in which a part of a human antibody is replaced by a part of an antibody of a mammal other than human.
  • the antibody consists of an Fc region, a Fab region, and a hinge region, which are explained below.
  • a specific example of such a chimeric antibody is a chimeric antibody in which the Fc region is derived from a human antibody while the Fab region is derived from an antibody of another mammal.
  • the hinge region is derived from either a human antibody or an antibody of another mammal.
  • a chimeric antibody in which the Fc region is derived from another mammal while the Fab region is derived from a human antibody is included.
  • the hinge region is derived from either a human antibody or an antibody of another mammal.
  • an antibody can also be said to be composed of a variable region and a constant region.
  • chimeric antibodies include those in which the heavy chain constant region ( CH ) and the light chain constant region ( CL ) are derived from a human antibody, while the heavy chain variable region ( VH ) and the light chain variable region ( VL ) are derived from an antibody of another mammal, and conversely, those in which the heavy chain constant region ( CH ) and the light chain constant region ( CL ) are derived from an antibody of another mammal, while the heavy chain variable region ( VH ) and the light chain variable region ( VL ) are derived from a human antibody.
  • the species of the other mammal is not particularly limited as long as it is a mammal other than human, but is preferably a mouse, rat, rabbit, horse, or a non-human primate, such as a mouse.
  • an antibody has a basic structure consisting of a total of four polypeptide chains: two immunoglobulin light chains (or simply “light chains”) and two immunoglobulin heavy chains (or simply “heavy chains”).
  • an “antibody” in addition to those having this basic structure, (1) Consists of two polypeptide chains, one light chain and one heavy chain, (2) Those consisting of a Fab region in which the Fc region has been deleted from the basic structure of an antibody in the original sense, and those consisting of a Fab region and all or part of the hinge region (including Fab, F(ab') and F(ab') 2 ), (3) A single-chain antibody, which is composed of a light chain with a linker at its C-terminus and a heavy chain at its C-terminus.
  • a single-chain antibody which is composed of a heavy chain with a linker at its C-terminus and a light chain at its C-terminus.
  • Single domain antibodies described below, are also included in the term "antibody.”
  • the antibody in one embodiment of the present invention is an antibody derived from a camelid (including an alpaca). Some camelid antibodies are composed of two heavy chains linked by a disulfide bond. An antibody composed of two heavy chains is called a heavy chain antibody.
  • a VHH is an antibody composed of a single heavy chain including the variable region of the heavy chain constituting a heavy chain antibody, or an antibody composed of a single heavy chain lacking the constant region (CH) constituting a heavy chain antibody.
  • a VHH is also one of the antibodies in an embodiment of the present invention.
  • Another antibody in an embodiment of the present invention is an antibody composed of two light chains linked by a disulfide bond. An antibody composed of two light chains is called a light chain antibody.
  • an antibody in one embodiment of the present invention is also an antibody in which a mutation has been added to the amino acid sequence of an antibody of a camelid.
  • a mutation is added to the amino acid of an antibody of a camelid, the same mutation as that which can be added to an antibody described in this specification can be added.
  • the antibody in one embodiment of the present invention is a shark-derived antibody.
  • a shark antibody consists of two heavy chains linked by a disulfide bond. An antibody consisting of these two heavy chains is called a heavy chain antibody.
  • a VNAR is an antibody consisting of a single heavy chain that includes the variable region of the heavy chain that constitutes a heavy chain antibody, or an antibody consisting of a single heavy chain that lacks the constant region (CH) that constitutes a heavy chain antibody.
  • VNAR is also one of the antibodies in an embodiment of the present invention.
  • an antibody in one embodiment of the present invention is also an antibody in one embodiment of the present invention in which mutations have been added to the amino acid sequence of the shark antibody.
  • mutations are added to the amino acids of a shark antibody, the same mutations that can be added to the antibodies described in this specification can be added.
  • a humanized shark antibody is also one of the antibodies in one embodiment of the present invention.
  • An antibody has a basic structure consisting of four polypeptide chains, two light chains and two heavy chains, and has three complementarity determining regions (CDRs) in the variable region of the light chain (V L ) and three complementarity determining regions (CDRs) in the variable region of the heavy chain (V H ).
  • the three CDRs in the light chain are called CDR1, CDR2, and CDR3, starting from the N-terminus.
  • the three CDRs in the heavy chain are also called CDR1, CDR2, and CDR3, starting from the N-terminus.
  • these CDRs are incomplete or absent, they are included in the antibody as long as they have the property of specifically binding to a specific antigen.
  • the regions other than the CDRs in the variable regions of the light and heavy chains are called framework regions (FR).
  • the FRs are called FR1, FR2, FR3, and FR4, starting from the N-terminus.
  • CDRs and FRs are present in the following order from the N-terminus: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • the antibody also includes an antibody in which a mutation such as substitution, deletion, or addition has been added to the amino acid sequence of the original antibody.
  • a mutation such as substitution, deletion, or addition has been added to the amino acid sequence of the original antibody.
  • the number of amino acids to be replaced is preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3.
  • the number of amino acids to be deleted is preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3.
  • an antibody in which a mutation combining these amino acid substitutions and deletions has been added is also an antibody.
  • amino acid sequence of the mutated antibody preferably exhibits 80% or more identity with the amino acid sequence of the original antibody, more preferably 85% or more identity, and even more preferably 90% or more, 95% or more, or 98% or more identity.
  • the antibody also includes an antibody in which a mutation such as substitution, deletion, or addition has been added to the amino acid sequence of the variable region of the original antibody.
  • a mutation such as substitution, deletion, or addition has been added to the amino acid sequence of the variable region of the original antibody.
  • the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3.
  • the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3.
  • an antibody in which a mutation that combines the substitution and deletion of these amino acids has been added is also an antibody.
  • an amino acid preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3 amino acids are added to the amino acid sequence or the N-terminus or C-terminus of the original antibody.
  • An antibody in which a mutation has been added is also an antibody in which a mutation that combines the addition, substitution, and deletion of these amino acids has been added is also an antibody.
  • the amino acid sequence of the antibody in which the mutation has been added is preferably 80% or more identical to the amino acid sequence of the original antibody, more preferably 85% or more identical, and even more preferably 90% or more, 95% or more, or 98% or more identical.
  • mutations may be made in either the CDR or framework region of the variable region, but are particularly made in the framework region.
  • the antibody also includes an antibody in which a mutation such as substitution, deletion, or addition has been added to the amino acid sequence of the framework region of the variable region of the original antibody.
  • a mutation such as substitution, deletion, or addition has been added to the amino acid sequence of the framework region of the variable region of the original antibody.
  • the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3.
  • the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3.
  • an antibody in which a mutation that combines these amino acid substitutions and deletions has been added is also an antibody.
  • amino acid sequence of the mutated antibody preferably exhibits 80% or more identity to the amino acid sequence of the original antibody, more preferably 85% or more identity, and even more preferably 90% or more, 95% or more, or 98% or more identity.
  • the antibody also includes an antibody in which a mutation such as substitution, deletion, or addition has been added to the amino acid sequence in the CDR region of the variable region of the original antibody.
  • a mutation such as substitution, deletion, or addition has been added to the amino acid sequence in the CDR region of the variable region of the original antibody.
  • the number of amino acids to be replaced is preferably 1 to 5, more preferably 1 to 3, and even more preferably 1 or 2.
  • the number of amino acids to be deleted is preferably 1 to 5, more preferably 1 to 3, and even more preferably 1 or 2.
  • an antibody in which a mutation combining these amino acid substitutions and deletions has been added is also an antibody.
  • amino acid sequence of the mutated antibody preferably exhibits 80% or more identity to the amino acid sequence of the original antibody, more preferably 85% or more identity, and even more preferably 90% or more, 95% or more, or 98% or more identity.
  • Fab refers to a molecule in which one light chain including a variable region and a CL region (light chain constant region) and one heavy chain including a variable region and a CH1 region (part 1 of the heavy chain constant region) are bound by disulfide bonds between cysteine residues present in each.
  • the heavy chain may further include a part of the hinge region in addition to the variable region and the CH1 region (part 1 of the heavy chain constant region), but in this case, the hinge region lacks cysteine residues present in the hinge region that bind the heavy chains of the antibody.
  • the light chain and the heavy chain are bound by disulfide bonds formed between cysteine residues present in the light chain constant region ( CL region) and cysteine residues present in the heavy chain constant region ( CH1 region) or hinge region.
  • the heavy chain that forms Fab is called a Fab heavy chain.
  • Fab lacks the cysteine residues present in the hinge region that bind the heavy chains of an antibody, and therefore consists of one light chain and one heavy chain.
  • the light chain that constitutes Fab contains a variable region and a CL region.
  • the heavy chain that constitutes Fab may be composed of a variable region and a CH1 region, or may contain a part of the hinge region in addition to the variable region and the CH1 region.
  • the hinge region is selected so as not to contain a cysteine residue that binds the heavy chains, so that a disulfide bond is not formed between the two heavy chains at the hinge region.
  • the heavy chain contains the whole or part of the hinge region that contains the cysteine residue that binds the heavy chains, in addition to the variable region and the CH1 region.
  • F(ab') 2 refers to a molecule in which two F(ab') are bound by disulfide bonds between the cysteine residues present in the hinge regions of each other.
  • the heavy chain that forms F(ab') or F(ab') 2 is called a Fab' heavy chain.
  • polymers such as dimers and trimers formed by binding multiple antibodies directly or via a linker are also antibodies.
  • any one that contains a part of an antibody molecule and has the property of specifically binding to an antigen is included in the "antibody” as used in the present invention. That is, the term “light chain” includes one that is derived from a light chain and has all or part of the amino acid sequence of its variable region.
  • the term “heavy chain” includes one that is derived from a heavy chain and has all or part of the amino acid sequence of its variable region. Therefore, as long as it has all or part of the amino acid sequence of the variable region, for example, one that has deleted the Fc region is also a heavy chain.
  • Fc or Fc region herein refers to a region in an antibody molecule that includes a fragment consisting of the CH2 region (part 2 of the heavy chain constant region) and the CH3 region (part 3 of the heavy chain constant region).
  • the antibody in one embodiment of the present invention comprises: (7) Also included are scFab, scF(ab'), and scF(ab') 2 , which are single-chain antibodies formed by linking the light chain and the heavy chain constituting Fab, F(ab'), or F(ab') 2 shown in (2) above via a linker.
  • scFab, scF(ab'), and scF(ab') 2 may be formed by linking a linker to the C-terminus of the light chain and further linking a heavy chain to the C-terminus, or may be formed by linking a linker to the C-terminus of the heavy chain and further linking a light chain to the C-terminus.
  • antibodies also include scFv, which is a single-chain antibody formed by linking a light chain variable region and a heavy chain variable region via a linker.
  • scFv is a single-chain antibody formed by linking a light chain variable region and a heavy chain variable region via a linker.
  • it may be formed by linking a linker to the C-terminus of the light chain variable region and further linking a heavy chain variable region to the C-terminus, or may be formed by linking a linker to the C-terminus of the heavy chain variable region and further linking a light chain variable region to the C-terminus.
  • antibody includes full-length antibodies, as well as those listed in (1) to (7) above, and also includes antigen-binding fragments (antibody fragments) in which a portion of a full-length antibody is deleted, which is a broader concept including (1) to (7).
  • Antigen-binding fragments include heavy chain antibodies, light chain antibodies, VHHs, VNARs, and those in which a portion of these antibodies is deleted.
  • binding fragment refers to a fragment of an antibody that retains at least a part of the specific binding activity with an antigen.
  • binding fragments include Fab, Fab', F(ab') 2 , variable region (Fv), single-chain antibody (scFv) in which the heavy chain variable region ( VH ) and the light chain variable region ( VL ) are linked with an appropriate linker, diabody, which is a dimer of a polypeptide containing a heavy chain variable region ( VH ) and a light chain variable region ( VL ), minibody, which is a dimer of a scFv heavy chain (H chain) bound to a part of the constant region ( CH3 ), other low molecular weight antibodies, etc.
  • it is not limited to these molecules as long as it has the ability to bind to an antigen.
  • single-chain antibody refers to a protein that can specifically bind to a specific antigen, which is composed of an amino acid sequence containing all or part of the variable region of a light chain, to which a linker is attached at the C-terminus, and to which an amino acid sequence containing all or part of the variable region of a heavy chain is further attached at the C-terminus.
  • a protein that can specifically bind to a specific antigen which is composed of an amino acid sequence containing all or part of the variable region of a heavy chain, to which a linker is attached at the C-terminus, and to which an amino acid sequence containing all or part of the variable region of a light chain is further attached at the C-terminus
  • a single-chain antibody in which a light chain is attached to the C-terminus of a heavy chain via a linker, the heavy chain usually lacks an Fc region.
  • the variable region of the light chain has three complementarity determining regions (CDRs) that are involved in the antigen specificity of the antibody. Similarly, the variable region of the heavy chain also has three CDRs.
  • a single chain antibody contains all three CDRs of the heavy chain and all three CDRs of the light chain.
  • a single chain antibody can also be one in which one or more CDRs are deleted, so long as the antigen-specific affinity of the antibody is maintained.
  • the linker disposed between the light and heavy chains of the antibody is preferably a peptide chain composed of 2 to 50, more preferably 8 to 50, even more preferably 10 to 30, even more preferably 12 to 18 or 15 to 25, for example 15 or 25 amino acid residues.
  • amino acid sequence of such a linker is preferably composed of only glycine or glycine and serine, and includes, for example, the amino acid sequence Gly-Ser, the amino acid sequence Gly-Gly-Ser, the amino acid sequence Gly-Gly-Gly, the amino acid sequence Gly-Gly-Gly, the amino acid sequence Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 9), the amino acid sequence Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 10), the amino acid sequence Ser-Gly-Gly-Gly-Gly (SEQ ID NO: 11), or a sequence in which these amino acid sequences are repeated 2 to 10 times or 2 to 5 times.
  • a linker containing a total of 15 amino acids corresponding to three consecutive amino acid sequences Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 9) is preferred.
  • a single domain antibody refers to an antibody that has the property of specifically binding to an antigen through a single variable region.
  • Single domain antibodies include antibodies whose variable region consists only of the variable region of a heavy chain (heavy chain single domain antibodies) and antibodies whose variable region consists only of the variable region of a light chain (light chain single domain antibodies).
  • VHH and VNAR are types of single domain antibodies.
  • the term "human transferrin receptor” refers to a membrane protein having the amino acid sequence shown in SEQ ID NO: 22.
  • the antibody of the present invention specifically binds to the portion of the amino acid sequence shown in SEQ ID NO: 22 from the 89th cysteine residue from the N-terminus to the phenylalanine at the C-terminus (the extracellular domain of the transferrin receptor), but is not limited thereto.
  • the most common method for producing antibodies against a desired protein is to produce a recombinant protein using cells that have been introduced with an expression vector incorporating a gene that codes for the protein, and then immunize animals such as mice with this recombinant protein. After immunization, antibody-producing cells against the recombinant protein are extracted from the animal, and these are then fused with myeloma cells to produce hybridoma cells capable of producing antibodies against the recombinant protein.
  • mice for immunizing immune system cells obtained from an animal such as a mouse with a desired protein by in vitro immunization
  • cells that produce antibodies against the protein can be obtained.
  • in vitro immunization there is no particular limitation on the animal species from which the immune system cells are derived, but preferred are mice, rats, rabbits, guinea pigs, dogs, cats, horses, and primates including humans, more preferably mice, rats, and humans, and even more preferably mice and humans.
  • splenocytes prepared from mouse spleens can be used as mouse immune system cells.
  • human immune system cells cells prepared from human peripheral blood, bone marrow, spleen, etc. can be used.
  • human immune system cells are immunized by in vitro immunization, human antibodies against the recombinant protein can be obtained.
  • Immune system cells can be immunized by in vitro immunization, and then fused with myeloma cells to produce hybridoma cells capable of producing antibodies. It is also possible to extract mRNA from the immunized cells, synthesize cDNA, and use this cDNA as a template to amplify DNA fragments containing genes encoding the light and heavy chains of immunoglobulins through PCR reactions, which can then be used to artificially reconstruct antibody genes.
  • Hybridoma cells obtained as is by the above method also include cells that produce antibodies that recognize non-target proteins as antigens. Furthermore, not all hybridoma cells that produce antibodies against a desired protein necessarily produce antibodies that exhibit the desired properties, such as having high affinity for that protein.
  • artificially reconstructed antibody genes also include genes that code for antibodies that recognize untargeted proteins as antigens. Furthermore, not all genes that code for antibodies against a desired protein necessarily code for antibodies that exhibit the desired properties, such as having high affinity for that protein.
  • a step is required to select hybridoma cells that produce antibodies with the desired characteristics from the hybridoma cells obtained as described above. Furthermore, in the case of artificially reconstructed antibody genes, a step is required to select genes that code for antibodies with the desired characteristics from the antibody genes.
  • the method described in detail below is effective as a method for selecting hybridoma cells that produce antibodies that show high affinity for a desired protein (high affinity antibodies), or genes that code for high affinity antibodies.
  • the protein when selecting hybridoma cells that produce antibodies with high affinity to a desired protein, the protein is added to a plate and retained thereon, then the culture supernatant of the hybridoma cells is added, and antibodies that are not bound to the protein are removed from the plate, and the amount of antibody retained on the plate is measured.
  • the higher the affinity of the antibody contained in the culture supernatant of the hybridoma cells added to the plate for the protein the greater the amount of antibody retained on the plate. Therefore, the amount of antibody retained on the plate can be measured, and the hybridoma cells corresponding to the plate that retain more antibodies can be selected as cell lines that produce antibodies with relatively high affinity for the protein.
  • mRNA can be extracted to synthesize cDNA, and the cDNA can be used as a template to amplify a DNA fragment containing a gene encoding an antibody against the protein using PCR, thereby isolating a gene encoding a high-affinity antibody.
  • the artificially reconstructed antibody gene is first incorporated into an expression vector, and this expression vector is introduced into a host cell.
  • the cells used as the host cell are not particularly limited, regardless of whether they are prokaryotic or eukaryotic, as long as they can express the antibody gene by introducing an expression vector incorporating the artificially reconstructed antibody gene, but cells derived from mammals such as humans, mice, and Chinese hamsters are preferred, and CHO cells derived from Chinese hamster ovaries or NS/0 cells derived from mouse myeloma are particularly preferred.
  • the expression vector used to incorporate and express a gene encoding an antibody gene can be used without any particular limitation as long as it expresses the gene when introduced into a mammalian cell.
  • the gene incorporated into the expression vector is located downstream of a DNA sequence (gene expression control site) that can regulate the frequency of gene transcription in a mammalian cell.
  • gene expression control sites include a promoter derived from a cytomegalovirus, an SV40 early promoter, a human elongation factor-1 alpha (EF-1 ⁇ ) promoter, and a human ubiquitin C promoter.
  • Mammalian cells into which such an expression vector has been introduced will express the above-mentioned artificially reconstructed antibody incorporated in the expression vector.
  • a method is used in which the protein is added to a plate and retained thereon, the cell culture supernatant is brought into contact with the protein, and then antibodies that are not bound to the protein are removed from the plate, and the amount of antibody retained on the plate is measured. According to this method, the higher the affinity of the antibody contained in the cell culture supernatant for the protein, the greater the amount of antibody retained on the plate.
  • cells corresponding to the plate that retain a greater number of antibodies can be selected as cell lines that produce antibodies with relatively high affinity for the protein, and thus a gene encoding an antibody with high affinity for the protein can be selected. From the cell line thus selected, a DNA fragment containing a gene encoding an antibody against the protein can be amplified using PCR to isolate a gene encoding a high affinity antibody.
  • a method for producing a conjugate in which a fusion protein of SA and a lysosomal enzyme, a fusion protein of SA and a cytokine, or a fusion protein of SA and a neurotrophic factor is bound to an antibody is a method in which the fusion protein and the antibody are bound via a non-peptide linker or a peptide linker.
  • non-peptide linker polyethylene glycol, polypropylene glycol, a copolymer of ethylene glycol and propylene glycol, polyoxyethylated polyol, polyvinyl alcohol, polysaccharides, dextran, polyvinyl ether, biodegradable polymers, lipid polymers, chitins, and hyaluronic acid, or derivatives thereof, or combinations thereof, can be used.
  • the peptide linker is a peptide chain or a derivative thereof consisting of 1 to 50 amino acids bound by peptide bonds, and its N-terminus and C-terminus form covalent bonds with either the fusion protein or the antibody, respectively, thereby binding the fusion protein and the antibody.
  • the SA, lysosomal enzyme, cytokine, and neurotrophic factor may each be derived from a human or a non-human animal.
  • a conjugate of a fusion protein and an antibody is produced by producing the fusion protein and the antibody separately, then reacting them with a non-peptide linker or a peptide linker to bind the fusion protein to one end of the linker and the antibody to the other end.
  • the fusion protein and the antibody can each be produced as recombinant proteins.
  • a conjugate in which a fusion protein of HSA and a human lysosomal enzyme is bound to an antibody via a non-peptide linker or a peptide linker a conjugate in which a fusion protein of HSA and a human cytokine is bound to an antibody via a non-peptide linker or a peptide linker, or a conjugate in which a fusion protein of HSA and a human neurotrophic factor is bound to an antibody via a non-peptide linker or a peptide linker can be produced.
  • a fusion protein of HSA and hGALC a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4 can be obtained in which the fusion protein is bound to an antibody or a ligand via a non-peptide linker or a peptide linker.
  • a conjugate formed by binding a fusion protein of SA and a lysosomal enzyme, a fusion protein of SA and a cytokine, or a fusion protein of SA and a neurotrophic factor to an antibody or a ligand can be produced by binding the C-terminus or N-terminus of these fusion proteins to the N-terminus or C-terminus of an antibody or ligand via a peptide bond, either directly or via a linker.
  • Such conjugates can be produced as recombinant proteins by the production methods exemplified below.
  • an antibody and a fusion protein of HSA and hGALC can be produced by binding the N-terminus or C-terminus of the fusion protein to the C-terminus or N-terminus of the heavy or light chain of the antibody via a peptide bond, either directly or via a linker.
  • a conjugate formed by binding an antibody to a fusion protein of HSA and hGALC can be obtained as a conjugate protein by inserting a DNA fragment in which the cDNA encoding the fusion protein of HSA and hGALC is placed in frame at the 3'-terminus or 5'-terminus of the cDNA encoding the heavy or light chain of the antibody, either directly or via a DNA fragment encoding a linker, into an expression vector for mammalian cells, and culturing mammalian cells into which this expression vector has been introduced.
  • an expression vector for mammalian cells incorporating a cDNA fragment encoding the light chain of an antibody can also be introduced into the same host cell, and in the case where a DNA fragment encoding a fusion protein of HSA and hGALC is bound to a light chain, an expression vector for mammalian cells incorporating a cDNA fragment encoding the heavy chain of an antibody can also be introduced into the same host cell.
  • the conjugate of the antibody and the fusion protein of HSA and hGALC can be obtained as a recombinant protein by incorporating a DNA fragment encoding a single-chain antibody or VHH linked to the 5'-end or 3'-end of the cDNA encoding the fusion protein of HSA and hGALC directly or via a DNA fragment encoding a linker into an expression vector (for mammalian cells, eukaryotic cells such as yeast, or prokaryotic cells such as E. coli) and expressing the DNA fragment in the cells into which the expression vector has been introduced.
  • an expression vector for mammalian cells, eukaryotic cells such as yeast, or prokaryotic cells such as E. coli
  • Conjugates of the fusion protein of HSA and hGBA and an antibody conjugates of the fusion protein of HSA and hIL-10 and an antibody, conjugates of the fusion protein of HSA and hBDNF and an antibody, conjugates of the fusion protein of HSA and hNGF and an antibody, conjugates of the fusion protein of HSA and hNT-3 and an antibody, and conjugates of the fusion protein of HSA and hNT-4 and an antibody can also be obtained as recombinant proteins in the same manner as the conjugates of the fusion protein of HSA and hGALC and an antibody.
  • Antibody, HSA and hGALC can also be conjugated in a form in which an antibody is interposed between HSA and hGALC.
  • the N-terminus of the heavy or light chain of the antibody can be bound to the C-terminus of HSA via a linker or directly, and hGALC can be bound to the C-terminus via a peptide bond either via a linker or directly.
  • the N-terminus of the heavy or light chain of the antibody can be bound to the C-terminus of hGALC via a linker or directly, and HSA can be bound to the C-terminus via a linker or directly, respectively.
  • the antibody is a single-chain antibody or VHH
  • the antibody, HSA and hGALC can be made into a fusion protein in a form in which an antibody is interposed between HSA and hGALC.
  • the N-terminus of the single-chain antibody or VHH can be bound to the C-terminus of HSA via a linker or directly
  • hGALC can be bound to the C-terminus via a peptide bond either via a linker or directly.
  • the N-terminus of a single chain antibody or VHH can be bound to the C-terminus of hGALC via a linker or directly
  • HSA can be bound to the C-terminus of the N-terminus of the single chain antibody or VHH via a linker or directly, respectively, by peptide bonds.
  • Similar conjugates can also be produced for lysosomal enzymes other than hGALC, such as hGBA, cytokines, such as hIL-10, and neurotrophic factors, such as hBDNF, hNGF, hNT-3, and hNT-4.
  • conjugates in this manner that involve an antibody are also referred to as fusion proteins of an antibody, another human lysosomal enzyme, and HSA, fusion proteins of an antibody, another human cytokine, and HSA, and fusion proteins of an antibody, another human neurotrophic factor, and HSA.
  • the above method can also be applied to the production of conjugates of fusion proteins of other human lysosomal enzymes and HSA, fusion proteins of other human cytokines and HSA, or fusion proteins of other human neurotrophic factors and HSA, and antibodies. It can also be applied to the production of conjugates in which the SA is from an animal species other than human, and conjugates in which the lysosomal enzymes, cytokines, and neurotrophic factors are from an animal species other than human.
  • the expression vectors, host cells, media, etc. that can be used to produce the above-mentioned fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA can also be used to produce a conjugate of the above-mentioned fusion protein and antibody.
  • Preferred embodiments of the conjugates of antibody, HSA and hGALC include the following (1) to (4): (1) A conjugate in which a fusion protein of HSA and hGALC is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (2) A conjugate in which a fusion protein of HSA and hGALC is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (3) A conjugate in which a fusion protein of HSA and hGALC is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (4) A conjugate in which a fusion protein of HSA and hGALC is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hGALC is further bound
  • a preferred example of (1) is a conjugate A in which a Fab heavy chain having the amino acid sequence of SEQ ID NO:25 is connected to its C-terminus with a linker having an amino acid sequence in which SEQ ID NO:9 is repeated three times, and further connected to its C-terminus with HSA having the amino acid sequence of SEQ ID NO:3, and further connected to its C-terminus with a linker having the amino acid sequence of SEQ ID NO:9, and further connected to its C-terminus with hGALC having the amino acid sequence of SEQ ID NO:1, and a light chain of SEQ ID NO:23.
  • conjugate A has the amino acid sequence of SEQ ID NO:28.
  • a suitable example of (2) is a conjugate B in which a linker having the amino acid sequence of SEQ ID NO: 9 is attached to the C-terminus of HSA having the amino acid sequence of SEQ ID NO: 3, hGALC having the amino acid sequence of SEQ ID NO: 1 at its C-terminus, a linker having an amino acid sequence in which SEQ ID NO: 9 is repeated three times at its C-terminus, and a Fab heavy chain having the amino acid sequence of SEQ ID NO: 25 at its C-terminus is attached to the light chain of SEQ ID NO: 23.
  • conjugate B has the amino acid sequence of SEQ ID NO: 30.
  • a preferred example of (3) is a conjugate C in which a linker having an amino acid sequence of SEQ ID NO: 25, an amino acid sequence in which SEQ ID NO: 9 is repeated three times, and hGALC having the amino acid sequence of SEQ ID NO: 1 at its C-terminus, a linker having the amino acid sequence of SEQ ID NO: 9 at its C-terminus, and HSA having the amino acid sequence of SEQ ID NO: 3 at its C-terminus are bound to the light chain of SEQ ID NO: 23.
  • the conjugate C has the amino acid sequence of SEQ ID NO: 32.
  • a preferred example of (4) is a conjugate D in which hGALC having the amino acid sequence of SEQ ID NO:1 is connected to its C-terminus with a linker having the amino acid sequence of SEQ ID NO:9, and further connected to its C-terminus with HSA having the amino acid sequence of SEQ ID NO:3, and further connected to its C-terminus with a linker having an amino acid sequence in which SEQ ID NO:9 is repeated three times, and further connected to its C-terminus with a Fab heavy chain having the amino acid sequence of SEQ ID NO:25, and a light chain of SEQ ID NO:23.
  • conjugate C has the amino acid sequence of SEQ ID NO:34.
  • Preferred embodiments of the conjugates of antibody, HSA and hGBA include the following (1) to (4): (1) A conjugate in which a fusion protein of HSA and hGBA is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (2) A conjugate in which a fusion protein of HSA and hGBA is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (3) A conjugate in which a fusion protein of HSA and hGBA is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (4) A conjugate in which a fusion protein of HSA and hGBA is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hGBA is further bound
  • Preferred embodiments of the conjugate of an antibody, HSA and hIL-10 include the following (1) to (4): (1) A conjugate in which a fusion protein of HSA and hIL-10 is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (2) A conjugate in which a fusion protein of HSA and hIL-10 is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (3) A conjugate in which a fusion protein of HSA and hIL-10 is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (4) A conjugate in which a fusion protein of HSA and hIL-10 is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hIL-10 is further
  • Preferred embodiments of the conjugate of an antibody, HSA and hBDNF include the following (1) to (4): (1) A conjugate in which a fusion protein of HSA and hBDNF is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (2) A conjugate in which a fusion protein of HSA and hBDNF is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (3) A conjugate in which a fusion protein of HSA and hBDNF is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (4) A conjugate in which a fusion protein of HSA and hBDNF is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hBDNF is further
  • Preferred embodiments of the conjugate of antibody, HSA and hNGF include the following (1) to (4): (1) A conjugate in which a fusion protein of HSA and hNGF is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (2) A conjugate in which a fusion protein of HSA and hNGF is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (3) A conjugate in which a fusion protein of HSA and hNGF is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (4) A conjugate in which a fusion protein of HSA and hNGF is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hNGF is further bound
  • Preferred embodiments of the conjugate of an antibody, HSA and hNT-3 include the following (1) to (4): (1) A conjugate in which a fusion protein of HSA and hNT-3 is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (2) A conjugate in which a fusion protein of HSA and hNT-3 is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (3) A conjugate in which a fusion protein of HSA and hNT-3 is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (4) A conjugate in which a fusion protein of HSA and hNT-3 is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hNT-3 is further
  • Preferred embodiments of the conjugate of an antibody, HSA and hNT-4 include the following (1) to (4): (1) A conjugate in which a fusion protein of HSA and hNT-4 is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (2) A conjugate in which a fusion protein of HSA and hNT-4 is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain; (3) A conjugate in which a fusion protein of HSA and hNT-4 is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (4) A conjugate in which a fusion protein of HSA and hNT-4 is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain; (5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hNT-4 is further
  • Linkers used when binding antibodies to a fusion protein of SA and a lysosomal enzyme, a fusion protein of SA and a cytokine, or a fusion protein of SA and a neurotrophic factor are described in detail below. These linkers are particularly suitable as linkers when binding antibodies to a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4.
  • the linker disposed between the antibody or ligand and the fusion protein is preferably a peptide chain composed of 1 to 60 or 1 to 50, more preferably 1 to 17, even more preferably 1 to 10, and even more preferably 1 to 5 amino acids, but the number of amino acids constituting the linker can be appropriately adjusted to 1, 2, 3, 1 to 17, 1 to 10, 10 to 40, 20 to 34, 23 to 31, 25 to 29, 27, etc.
  • Such a linker is not limited in its amino acid sequence as long as the antibody linked thereto retains affinity for a receptor on cerebrovascular endothelial cells and the fusion protein linked by the linker can exert the physiological activity of the fusion protein under physiological conditions, but is preferably composed of glycine and serine.
  • amino acid sequence Gly-Ser amino acid sequence Ser-Ser, the amino acid sequence Gly-Gly-Ser, the amino acid sequence Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 9), the amino acid sequence Gly-Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 10), the amino acid sequence Ser-Gly-Gly-Gly-Gly (SEQ ID NO: 11), or those containing a sequence consisting of 1 to 10 or 2 to 5 consecutive amino acids of these amino acid sequences.
  • those containing the amino acid sequence Gly-Ser can be suitably used as a linker.
  • a linker containing a total of 27 amino acids consisting of the amino acid sequence Gly-Ser followed by five consecutive amino acid sequences of Gly-Gly-Gly-Gly-Ser SEQ ID NO: 9 can be preferably used as the linker.
  • a linker containing a total of 25 amino acids consisting of five consecutive amino acid sequences of Gly-Gly-Gly-Gly-Ser can also be preferably used as the linker.
  • each linker is named, starting from the N-terminus, as the first linker, the second linker, and so on.
  • antibodies to be bound to the fusion protein of SA and a lysosomal enzyme, the fusion protein of SA and a cytokine, or the fusion protein of SA and a neurotrophic factor include the following anti-human transferrin receptor antibodies (anti-hTfR antibodies).
  • a light chain variable region comprising the amino acid sequence of SEQ ID NO:96 as CDR1, the amino acid sequence of SEQ ID NO:97 as CDR2, and the amino acid sequence of SEQ ID NO:98 as CDR3, and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO:99 as CDR1, the amino acid sequence of SEQ ID NO:100 as CDR2, and the amino acid sequence of SEQ ID NO:101 as CDR3, respectively; and (2) a light chain variable region comprising the amino acid sequence of SEQ ID NO:96 as CDR1, the amino acid sequence of SEQ ID NO:97 as CDR2, and the amino acid sequence of SEQ ID NO:98 as CDR3, respectively, and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO:102 as CDR1, the amino acid sequence of SEQ ID NO:103 as CDR2, and the amino acid sequence of SEQ ID NO:104 as CDR3,
  • the present invention is not limited to these, and the above amino acid sequences may be appropriately mutated by substitution, deletion, addition
  • the anti-hTfR antibodies (1) and (2) above are particularly suitable as antibodies to be bound to a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4.
  • anti-hTfR antibodies (1) and (2) above with substitutions, additions, deletions, etc. can also be used as antibodies constituting the conjugate, so long as they retain their affinity for the human transferrin receptor.
  • the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2.
  • the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2. Mutations that combine these amino acid replacements and deletions can also be added.
  • amino acids are added to the amino acid sequence of the light chain of the anti-hTfR antibody of (1) and (2) above, preferably 1 to 10 amino acids, more preferably 1 to 5 amino acids, even more preferably 1 to 3 amino acids, and even more preferably 1 or 2 amino acids are added to the amino acid sequence of the light chain or to the N-terminus or C-terminus. Mutations that combine the addition, substitution, and deletion of these amino acids can also be added.
  • the amino acid sequence of the mutated light chain preferably has an identity of 80% or more, more preferably an identity of 90% or more, and even more preferably an identity of 95% or more, with the amino acid sequence of the original light chain.
  • the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2.
  • the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2. Mutations that combine these amino acid substitutions and deletions can also be added.
  • amino acids are added to the amino acid sequence of the heavy chain of the anti-hTfR antibody of (1) and (2) above, preferably 1 to 10 amino acids, more preferably 1 to 5 amino acids, even more preferably 1 to 3 amino acids, and even more preferably 1 or 2 amino acids are added to the amino acid sequence of the heavy chain or to the N-terminus or C-terminus. Mutations that combine the addition, substitution, and deletion of these amino acids can also be added.
  • the amino acid sequence of the mutated heavy chain preferably has an identity of 80% or more, more preferably an identity of 90% or more, and even more preferably an identity of 95% or more, with the amino acid sequence of the original heavy chain.
  • antibodies to be bound to the fusion protein of SA and a lysosomal enzyme, the fusion protein of SA and a cytokine, or the fusion protein of SA and a neurotrophic factor include the following anti-hTfR heavy chain antibody variable region (VHH) peptides (hTfR affinity peptides): (3) An hTfR affinity peptide comprising the amino acid sequence of SEQ ID NO: 105 or SEQ ID NO: 106 as CDR1, the amino acid sequence of SEQ ID NO: 107 or SEQ ID NO: 108 as CDR2, and the amino acid sequence of SEQ ID NO: 109 or SEQ ID NO: 110 as CDR3, and (4) an hTfR affinity peptide comprising the amino acid sequence of SEQ ID NO: 105 or SEQ ID NO: 106 as CDR1, the amino acid sequence of SEQ ID NO: 111 or SEQ ID NO: 112 as CDR2, and the amino acid sequence of SEQ ID NO: 109 or SEQ ID NO: 110
  • the hTfR affinity peptides (3) and (4) above are particularly suitable as antibodies to be bound to a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4.
  • hTfR affinity peptides (3) and (4) can also be used as antibodies that contain substitutions, additions, deletions, etc., as long as they retain their affinity for the human transferrin receptor.
  • the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2.
  • the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2. Mutations that combine these amino acid substitutions and deletions can also be added.
  • amino acids are added to the amino acid sequence of the hTfR affinity peptide in (3) and (4) above, preferably 1 to 10 amino acids are added to the amino acid sequence of the light chain or to the N-terminus or C-terminus, more preferably 1 to 5 amino acids, even more preferably 1 to 3 amino acids, and even more preferably 1 or 2 amino acids. Mutations that combine the addition, substitution, and deletion of these amino acids can also be added.
  • the amino acid sequence of the mutated light chain preferably has an identity of 80% or more, more preferably 90% or more, and even more preferably 95% or more with the amino acid sequence of the original hTfR affinity peptide.
  • the fusion protein of HSA and hGALC can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration into the blood to treat central nervous system diseases caused by GALC deficiency, such as Krabbe disease.
  • the conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the conjugate into the blood (including intravenous injection such as intravenous drip) to a patient for central nervous system diseases caused by GALC deficiency, such as Krabbe disease.
  • the conjugate administered into the blood can reach not only the brain, but also other organs and tissues that express GALC.
  • the drug can also be used to prevent the onset of the disease.
  • the fusion protein of HSA and hGBA can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration into the blood to treat central nervous system diseases caused by GBA deficiency, such as Gaucher disease.
  • the conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the conjugate into the blood (including intravenous injection such as intravenous drip) to a patient for central nervous system diseases caused by GBA deficiency, such as Gaucher disease.
  • the conjugate administered into the blood can reach not only the brain but also other organs and tissues.
  • the drug can also be used to prevent the onset of the disease.
  • the fusion protein of HSA and hIL-10 can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and exert its function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration to the blood for the treatment of a disease of the central nervous system.
  • the conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the disease of the central nervous system to a patient in the blood (including intravenous injection such as intravenous drip).
  • the conjugate administered to the blood can reach not only the brain, but also other organs and tissues in which a therapeutic effect can be exerted by IL-10.
  • the drug can also be used to prevent the onset of the disease.
  • the fusion protein of HSA and hBDNF can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and exert its function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration to the blood for the treatment of a disease of the central nervous system.
  • the conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the disease of the central nervous system to a patient in the blood (including intravenous injection such as intravenous drip).
  • the conjugate administered to the blood can reach not only the brain, but also other organs and tissues in which BDNF can exert a therapeutic effect.
  • the drug can also be used to prevent the onset of the disease.
  • the fusion protein of HSA and hNGF can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and exert its function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration to the blood for the treatment of diseases of the central nervous system.
  • the conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the conjugate to the blood of a patient (including intravenous injection such as intravenous drip infusion).
  • the conjugate administered to the blood can reach not only the brain, but also other organs and tissues in which NGF can exert a therapeutic effect.
  • the drug can also be used to prevent the onset of the disease.
  • the fusion protein of HSA and hNT-3 can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and exert its function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration to the blood for the treatment of diseases of the central nervous system.
  • the conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the drug to a patient in the blood (including intravenous injection such as intravenous drip infusion).
  • the conjugate administered to the blood can reach not only the brain, but also other organs and tissues in which NT-3 can exert a therapeutic effect.
  • the drug can also be used to prevent the onset of the disease.
  • the fusion protein of HSA and hNT-4 can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and exert its function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration to the blood for the treatment of a disease of the central nervous system.
  • the conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the disease of the central nervous system to a patient in the blood (including intravenous injection such as intravenous drip).
  • the conjugate administered to the blood can reach not only the brain, but also other organs and tissues in which NT-4 can exert a therapeutic effect.
  • the drug can also be used to prevent the onset of the disease.
  • HSA-hGALC fusion protein, HSA-hGBA fusion protein, HSA-hIL-10 fusion protein, HSA-hBDNF fusion protein, HSA-hNGF fusion protein, HSA-hNT-3 fusion protein, and HSA-hNT-4 fusion protein can be used as drugs that are administered into the blood and exert their medicinal effects in the central nervous system (CNS) by binding to antibodies or ligands for receptors on cerebrovascular endothelial cells.
  • CNS central nervous system
  • Such drugs are generally administered to patients by intravenous injection, subcutaneous injection, or intramuscular injection, but there are no particular limitations on the administration route.
  • hGALC has the function of hGALC in a conjugate of an antibody and a fusion protein of HSA and hGALC
  • hGALC has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more or 95% or more, when the specific activity of normal wild-type hGALC is taken as 100%.
  • hGALC has the function of hGALC in a conjugate of an antibody and a fusion protein of HSA and hGALC
  • hGALC has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more or 95% or more, when the specific activity of normal wild-type hGALC is taken as 100%.
  • the specific activity of hGALC in the conjugate is calculated by multiplying the enzyme activity of hGALC per unit mass of the conjugate ( ⁇ M/hour/mg protein) by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hGALC).
  • the same can be said for the conjugate of the fusion protein of HSA and hGBA and an antibody.
  • the same can be said for the conjugate of the fusion protein of HSA and hIL-10 and an antibody, the conjugate of the fusion protein of HSA and hBDNF and an antibody, the conjugate of the fusion protein of HSA and hNGF and an antibody, the conjugate of the fusion protein of HSA and hNT-3 and an antibody, and the conjugate of the fusion protein of HSA and hNT-4 and an antibody, although hIL-10, hBDNF, hNGF, hNT-3, and hNT-4 are not enzymes, so the specific activity is based on the physiological activity of each of the proteins hIL-10, hBDNF, hNGF, hNT-3, and hNT-4, rather than on the enzyme activity.
  • the fusion protein of SA and a lysosomal enzyme, the fusion protein of SA and a cytokine, or the fusion protein of SA and a neurotrophic factor may be a conjugate with a ligand.
  • the SA, the lysosomal enzyme, the cytokine, and the neurotrophic factor may all be derived from humans or from animals other than humans.
  • examples of the fusion protein of SA and a lysosomal enzyme include a fusion protein of HSA and hGALC and a fusion protein of HSA and hGBA
  • examples of the fusion protein of SA and a cytokine include a fusion protein of HSA and hIL-10
  • examples of the fusion protein of SA and a neurotrophic factor include a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, and a fusion protein of HSA and hNT-4.
  • the ligand is, for example, one that can specifically bind to a receptor on cerebrovascular endothelial cells, such as the insulin receptor, transferrin receptor, leptin receptor, lipoprotein receptor, and IGF receptor.
  • the corresponding ligands for the insulin receptor, transferrin receptor, leptin receptor, lipoprotein receptor, and IGF receptor are insulin, leptin, lipoprotein, and IGF (IGF-1, IGF-2), respectively.
  • the ligand may be a wild type or a mutant wild type, so long as it can specifically bind to the target receptor. Also, it may be a fragment of the ligand, so long as it can specifically bind to the target receptor.
  • ligands are particularly suitable as ligands to be bound to a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4.
  • the transferrin when the ligand is transferrin (Tf), the transferrin may be human Tf or Tf of an animal species other than human, but is preferably human Tf.
  • the transferrin may be wild-type, or may have a mutation introduced therein, so long as it has affinity for the transferrin receptor (TfR).
  • the mutation may be, for example, a substitution, addition, or deletion of one or two amino acids.
  • the transferrin may be full-length, or may be a fragment thereof, so long as it has affinity for the transferrin receptor (TfR). The same can be said for insulin, leptin, lipoproteins, and IGF (IGF-1, IGF-2).
  • a conjugate of a fusion protein of SA and a lysosomal enzyme, a fusion protein of SA and a cytokine, or a fusion protein of SA and a neurotrophic factor, and a ligand can be produced by the following method (1) or (2): (1) A fusion protein and a ligand are conjugated via a non-peptide linker or a peptide linker; (2) The fusion protein and the ligand are produced as a recombinant protein bound via a linker sequence or directly via a peptide bond.
  • non-peptide linker examples include polyethylene glycol, polypropylene glycol, copolymers of ethylene glycol and propylene glycol, polyoxyethylated polyol, polyvinyl alcohol, polysaccharides, dextran, polyvinyl ether, biodegradable polymers, lipid polymers, chitins, and hyaluronic acid, or derivatives or combinations of these.
  • the peptide linker is a peptide chain or derivatives consisting of 1 to 50 peptide-bonded amino acids, whose N-terminus and C-terminus form covalent bonds with either the fusion protein or the ligand, respectively, thereby binding the fusion protein and the ligand.
  • the fusion protein and the ligand are each produced as recombinant proteins.
  • the fusion protein and the ligand are produced as a recombinant protein in which the N-terminus of the ligand is bound to the C-terminus of the fusion protein via a linker sequence or directly, or the N-terminus of the fusion protein is bound to the C-terminus of the ligand via a linker sequence or directly.
  • the expression vectors, host cells, and media used to produce such recombinant proteins can be those used to produce the above-mentioned fusion protein of HSA and hGALC.
  • the fusion protein of SA and a lysosomal enzyme, the fusion protein of SA and a cytokine, or the fusion protein of SA and a neurotrophic factor can be a conjugate with another protein (A) as well as an antibody and a ligand.
  • A another protein
  • the SA, the lysosomal enzyme, the cytokine, and the neurotrophic factor may be derived from humans or animals other than humans.
  • examples of the fusion protein of SA and a lysosomal enzyme include a fusion protein of HSA and hGALC and a fusion protein of HSA and hGBA
  • examples of the fusion protein of SA and a cytokine include a fusion protein of HSA and hIL-10
  • examples of the fusion protein of SA and a neurotrophic factor include a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, and a fusion protein of HSA and hNT-4.
  • a conjugate of a fusion protein of SA and a lysosomal enzyme, a fusion protein of SA and a cytokine, or a fusion protein of SA and a neurotrophic factor, and a protein (A) can be produced by the following method (1) or (2): (1) The fusion protein and protein (A) are conjugated via a non-peptide linker or a peptide linker; (2) The fusion protein and protein (A) are produced as a recombinant protein by binding them via a linker sequence or directly via a peptide bond.
  • non-peptide linker examples include polyethylene glycol, polypropylene glycol, copolymers of ethylene glycol and propylene glycol, polyoxyethylated polyol, polyvinyl alcohol, polysaccharides, dextran, polyvinyl ether, biodegradable polymers, lipid polymers, chitins, and hyaluronic acid, or derivatives or combinations of these.
  • the peptide linker is a peptide chain or derivatives thereof consisting of 1 to 50 peptide-bonded amino acids, and its N-terminus and C-terminus form covalent bonds with either the fusion protein or protein (A), respectively, thereby binding the fusion protein and protein (A).
  • the fusion protein and protein (A) are each produced as recombinant proteins.
  • the fusion protein and the ligand are produced as a recombinant protein in which the N-terminus of the ligand is bound to the C-terminus of the fusion protein via a linker sequence or directly, or the N-terminus of the fusion protein is bound to the C-terminus of the ligand via a linker sequence or directly.
  • the expression vectors, host cells, and media used to produce such recombinant proteins can be those used to produce a fusion protein of HSA and hGALC.
  • the conjugate of the fusion protein of HSA and hGALC with an antibody, and the conjugate of the fusion protein of HSA and hGALC with a ligand or protein (A) can both be used as a pharmaceutical composition for the treatment of Krabbe disease (galactosylceramide lipidosis, or globoid cell leukodystrophy).
  • Krabbe disease galactosylceramide lipidosis, or globoid cell leukodystrophy
  • the conjugate of the fusion protein of HSA and hGBA with an antibody, and the conjugate of the fusion protein of HSA and hGBA with a ligand or protein (A) can both be used as a pharmaceutical composition for the treatment of Gaucher disease.
  • the conjugate of the fusion protein of HSA and hIL-10 with an antibody, and the conjugate of the fusion protein of HSA and hIL-10 with a ligand or protein (A) can both be used as pharmaceutical compositions for treating inflammatory diseases, neuropathic pain, multiple sclerosis, spinal cord injury, ALS, neuroinflammation, symptoms associated with arthritis and other joint diseases, and autoimmune diseases, etc.
  • the conjugate of the fusion protein of HSA and hBDNF with an antibody, and the conjugate of the fusion protein of HSA and hBDNF with a ligand or protein (A) can both be used as pharmaceutical compositions for treating neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, spinal degenerative diseases such as amyotrophic lateral sclerosis, diabetic neuropathy, cerebral ischemic disease, developmental disorders such as Rett syndrome, schizophrenia, depression, and Rett syndrome, etc.
  • neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease
  • spinal degenerative diseases such as amyotrophic lateral sclerosis, diabetic neuropathy, cerebral ischemic disease, developmental disorders such as Rett syndrome, schizophrenia, depression, and Rett syndrome, etc.
  • the conjugate of the fusion protein of HSA and hNGF with an antibody, and the conjugate of the fusion protein of HSA and hNGF with a ligand or protein (A) can both be used as pharmaceutical compositions for treating neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease.
  • the conjugate of the fusion protein of HSA and hNT-3 with an antibody, and the conjugate of the fusion protein of HSA and hNT-3 with a ligand or protein (A) can both be used as a pharmaceutical composition for treating diseases such as neurodegenerative disorders.
  • the conjugate of the fusion protein of HSA and hNT-4 with an antibody, and the conjugate of the fusion protein of HSA and hNT-4 with a ligand or protein (A) can both be used as a pharmaceutical composition for treating diseases such as neurodegenerative disorders.
  • a pharmaceutical composition containing as an active ingredient a conjugate of an antibody and a fusion protein of HSA and hGALC, or a conjugate of a fusion protein of HSA and hGALC and a ligand or protein (A), can be administered intravenously, intramuscularly, intraperitoneally, subcutaneously, or intracerebroventricularly as an injection. These injections can be supplied as freeze-dried preparations or aqueous liquid preparations. In the case of an aqueous liquid preparation, it may be in the form of a vial filled with the fusion protein, or it can be supplied as a prefilled preparation in a syringe.
  • the ratio of monomer to total protein is preferably 70% or more, more preferably 80% or more, even more preferably 90% or more, for example 95% or more. The same can be said for a solution obtained by dissolving a freeze-dried preparation in an aqueous medium and reconstituting it.
  • compositions comprising, as an active ingredient, a conjugate of a fusion protein of HSA and hGBA and an antibody, or a conjugate of a fusion protein of HSA and hGBA and a ligand or protein (A), a pharmaceutical composition comprising, as an active ingredient, a conjugate of a fusion protein of HSA and hIL-10 and an antibody, or a conjugate of a fusion protein of HSA and hIL-10 and a ligand or protein (A), a pharmaceutical composition comprising, as an active ingredient, a conjugate of a fusion protein of HSA and hBDNF and an antibody, or a conjugate of a fusion protein of HSA and hBDNF and a ligand or protein (A), It may also be applied to pharmaceutical compositions containing as active ingredients a conjugate of a fusion protein of SA and hNGF and an antibody, or a
  • the ratio of monomers to the total protein is preferably 70% or more, more preferably 80% or more, even more preferably 90% or more, for example 95% or more.
  • HSA-hGALC fusion proteins such as HSA-hGALC and hGALC-HSA, can be expressed as recombinant proteins to obtain homogeneous monomers. From the standpoint of production control, the method of producing hGALC as a fusion protein with HSA can be said to be an excellent method of producing hGALC.
  • the conjugate of the antibody and the fusion protein of HSA and hGALC is expressed as a recombinant protein, most of it is obtained as a monomer.
  • the ratio of monomer to total protein is preferably 70% or more, more preferably 80% or more, and even more preferably 90% or more, for example 95% or more.
  • the conjugate of the fusion protein of HSA and hGALC with an antibody is expressed as a recombinant protein, a homogeneous monomer can be obtained. Therefore, from the standpoint of production control, the method of producing hGALC as a fusion protein of HSA and an antibody can be said to be an excellent method of producing hGALC.
  • a fusion protein between SA and a lysosomal enzyme, a fusion protein between SA and a cytokine, or a fusion protein between SA and a neurotrophic factor, as well as a nucleic acid molecule containing a gene encoding the conjugate can be used in gene therapy.
  • a nucleic acid molecule containing a gene encoding a conjugate between any of these fusion proteins and an antibody, a nucleic acid molecule containing a gene encoding a conjugate between any of these fusion proteins and a ligand, and a nucleic acid molecule containing a gene encoding a conjugate between any of these fusion proteins and protein (A) can also be used in gene therapy.
  • the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4.
  • the antibody is, for example, an anti-transferrin receptor antibody.
  • the ligand is, for example, transferrin or a fragment thereof.
  • a nucleic acid molecule that can be used for gene therapy includes, for example, one having the following base sequence: (1) A base sequence including a first inverted terminal repeat (ITR) or a functional equivalent thereof, a base sequence including a gene expression control site downstream of the first inverted terminal repeat (ITR), a base sequence encoding a fusion protein further downstream, and a base sequence including a second inverted terminal repeat (ITR) or a functional equivalent thereof further downstream; (2) a nucleotide sequence containing a first long terminal repeat (LTR) or a functional equivalent thereof, a nucleotide sequence downstream thereof containing a gene expression control site, a nucleotide sequence further downstream thereof encoding a fusion protein, and a nucleotide sequence further downstream thereof containing a second long terminal repeat (LTR) or a functional equivalent thereof; (3) a base sequence containing a leader or a functional equivalent thereof, a base sequence downstream thereof containing a gene expression control site, a base
  • nucleic acid molecule refers primarily to either DNA, which is a polymer of deoxyribonucleotides formed by phosphodiester bonds, or RNA, which is a polymer of ribonucleotides formed by phosphodiester bonds.
  • the DNA may be single-stranded (single-stranded) or double-stranded with a complementary strand.
  • the DNA may be a (+) strand or a (-) strand.
  • the individual deoxyribonucleotides constituting the DNA may be of a naturally occurring type or may be modified from the natural type, so long as the gene encoding the protein contained in the DNA can be translated into mRNA in the cells of a mammal (particularly a human).
  • the individual deoxyribonucleotides constituting the DNA may be of a naturally occurring type or may be modified from the natural type, so long as the gene encoding the protein contained in the DNA can be translated into mRNA in the cells of a mammal (particularly a human) and the whole or part of the DNA can be replicated.
  • the RNA when the "nucleic acid molecule" is RNA, the RNA may be single-stranded (single-stranded) or double-stranded with a complementary strand. When the RNA is single-stranded, the RNA may be a (+) strand or a (-) strand.
  • the individual ribonucleotides constituting the RNA may be of a naturally occurring type or may be modified, so long as the gene encoding the protein contained in the RNA can be reverse transcribed into DNA in a mammalian (particularly human) cell.
  • the individual ribonucleotides constituting the RNA may be of a naturally occurring type or may be modified, so long as the gene encoding the protein contained in the RNA can be translated into a protein in a mammalian (particularly human) cell. Modification of ribonucleotides is performed, for example, to suppress degradation of RNA by RNase and to increase the stability of RNA in cells.
  • the term "inverted terminal repeat (ITR)" refers to a base sequence that exists at the end of the viral genome and in which the same sequence is repeated.
  • ITR those derived from adeno-associated virus and those derived from adenovirus can be preferably used.
  • the ITR of the adeno-associated virus is a region of approximately 145 bases in length and functions as a replication origin, etc.
  • there are two inverted terminal repeats (ITR) in the nucleic acid molecule which are called the first inverted terminal repeat (ITR) and the second inverted terminal repeat (ITR), respectively.
  • the ITR located on the 5' side is called the first inverted terminal repeat (ITR), and the ITR located on the 3' side is called the second inverted terminal repeat (ITR).
  • the inverted terminal repeat (ITR) may be derived from any virus as long as it has at least one of the functions of an original ITR, such as functioning as an origin of replication or inserting a gene into a host cell, and the ITR of the adeno-associated virus is one suitable one.
  • the serotype of AAV is not particularly limited and may be any of serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11.
  • the inverted terminal repeat (ITR) of AAV serotype 2 has a first ITR that contains the base sequence shown in SEQ ID NO: 113 and a second ITR that contains the base sequence shown in SEQ ID NO: 114.
  • the ITR is not limited to the wild-type ITR, and may be modified by substitution, deletion, addition, or the like, in the base sequence of the wild-type ITR.
  • the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3.
  • the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3.
  • a mutation combining these base substitutions and deletions can also be added.
  • a base When a base is added to the wild-type ITR, preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3 bases are added in the base sequence of the ITR or to the 5' end or 3' end. A mutation combining these base additions, substitutions, and deletions can also be added.
  • the nucleotide sequence of the mutated ITR preferably exhibits 80% or more identity to the nucleotide sequence of the wild-type ITR, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
  • a functional equivalent of an ITR is something that can be used functionally in place of an ITR.
  • an ITR artificially constructed based on an ITR is also a functional equivalent of an ITR as long as it can replace an ITR.
  • a functional equivalent of an AAV ITR is one that can be used functionally in place of an AAV ITR.
  • an ITR artificially constructed based on an AAV ITR is also a functional equivalent of an AAV ITR as long as it can replace an AAV ITR.
  • first AAV-ITR An example of a functional equivalent of the artificially constructed first AAV inverted terminal repeat (first AAV-ITR) is one having the base sequence shown in SEQ ID NO: 115 (functional equivalent of the first AAV-ITR).
  • This base sequence shown in SEQ ID NO: 115 to which substitutions, deletions, or mutations have been added is also included in the functional equivalent of the first AAV-ITR, so long as it can be used functionally in place of the ITR of the first AAV.
  • the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3.
  • the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3.
  • an ITR to which mutations that combine the substitution and deletion of these bases have been added is also a functional equivalent of the first AAV-ITR.
  • bases are added to the base sequence shown in SEQ ID NO:115, preferably 1 to 20 bases, more preferably 1 to 10 bases, even more preferably 1 to 5 bases, and even more preferably 1 to 3 bases are added to the base sequence or to the 5' end or 3' end.
  • ITRs with mutations that combine the addition, substitution, and deletion of these bases are also included in the functional equivalent of the first AAV-ITR.
  • the base sequence of the mutated ITR preferably shows 80% or more identity to the base sequence shown in SEQ ID NO:115, more preferably shows 85% or more identity, even more preferably shows 90% or more identity, even more preferably shows 95% or more identity, and even more preferably shows 98% or more identity.
  • an example of a functional equivalent of an artificially constructed second inverted terminal repeat is one having the base sequence shown in SEQ ID NO: 116 (functional equivalent of the second AAV-ITR).
  • This base sequence shown in SEQ ID NO: 116 to which substitutions, deletions, or mutations have been added is also included in the functional equivalent of the ITR of the second AAV, so long as it can be used functionally in place of the ITR of the second AAV.
  • the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3.
  • the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3.
  • an ITR to which mutations have been added that combine the substitution and deletion of these bases is also a functional equivalent of the ITR of the second AAV.
  • bases are added to the base sequence shown in SEQ ID NO:116, preferably 1 to 20 bases, more preferably 1 to 10 bases, even more preferably 1 to 5 bases, and even more preferably 1 to 3 bases are added to the base sequence or to the 5' end or 3' end.
  • ITRs with mutations that combine the addition, substitution, and deletion of these bases are also included in the functional equivalent of the ITR of the second AAV.
  • the base sequence of the mutated ITR preferably shows 85% or more identity with the base sequence shown in SEQ ID NO:116, more preferably shows 85% or more identity, even more preferably shows 90% or more identity, even more preferably shows 95% or more identity, and even more preferably shows 98% or more identity.
  • the term "long terminal repeat (LTR)” refers to a base sequence in which the same sequence is repeated hundreds to thousands of times, for example, at the end of a retrotransposon of a eukaryote, or a retrovirus genome, lentivirus genome, etc.
  • LTRs long terminal repeats
  • the LTR located on the 5' side is called the first long terminal repeat (LTR), and the LTR located on the 3' side is called the second long terminal repeat (LTR).
  • the long terminal repeat (LTR) may be derived from any virus as long as it has at least one of the functions of an original LTR, such as a function as a replication origin or gene insertion into a host cell, and preferred examples include retrovirus genomes and lentiviruses.
  • the LTR is not limited to a wild-type LTR, and may be a wild-type LTR with a modified base sequence, such as a substitution, deletion, or addition.
  • the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3.
  • the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. Mutations combining these base substitutions and deletions can also be added.
  • bases are added to the wild-type LTR preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3 bases are added in the LTR base sequence or at the 5' end or 3' end. Mutations combining these base additions, substitutions, and deletions can also be added.
  • the mutated LTR base sequence preferably exhibits 80% or more identity with the wild-type LTR base sequence, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
  • a functional equivalent of an LTR is something that can be used functionally in place of an LTR.
  • an artificially constructed LTR based on an LTR is also a functional equivalent of an LTR as long as it can replace an LTR.
  • a functional equivalent of a lentiviral LTR is one that can be used functionally in place of the lentiviral LTR.
  • an artificially constructed LTR based on a lentiviral LTR is also a functional equivalent of the lentiviral LTR as long as it can replace the lentiviral LTR.
  • a functional equivalent of a retroviral LTR is one that can be used functionally in place of a retroviral LTR.
  • an artificially constructed LTR based on a retroviral LTR is also a functional equivalent of a retroviral LTR as long as it can replace the retroviral LTR.
  • An example of a functional equivalent of the first LTR is one having the base sequence shown in SEQ ID NO: 117.
  • This base sequence shown in SEQ ID NO: 117 with substitutions, deletions, or mutations is also included in the functional equivalent of the first LTR, so long as it can be functionally used as the first LTR.
  • the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 20, even more preferably 1 to 5, and even more preferably 1 to 3.
  • the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3.
  • an LTR with a mutation that combines the substitution and deletion of these bases is also a functional equivalent of the first LTR.
  • bases are added to the base sequence shown in SEQ ID NO:117, preferably 1 to 20 bases, more preferably 1 to 10 bases, even more preferably 1 to 5 bases, and even more preferably 1 to 3 bases are added to the base sequence or to the 5' end or 3' end.
  • LTRs with mutations that combine the addition, substitution, and deletion of these bases are also included in the functional equivalent of the first LTR.
  • the base sequence of the mutated LTR preferably shows 80% or more identity with the base sequence shown in SEQ ID NO:117, more preferably shows 85% or more identity, even more preferably shows 90% or more identity, even more preferably shows 95% or more identity, and even more preferably shows 98% or more identity.
  • an example of a functional equivalent of the second LTR is one having the base sequence shown in SEQ ID NO: 118.
  • This base sequence shown in SEQ ID NO: 118 with substitutions, deletions, or mutations is also included in the functional equivalent of the second LTR as long as it can be functionally used as the second LTR.
  • the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3.
  • the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3.
  • an LTR with a mutation that combines the substitution and deletion of these bases is also a functional equivalent of the second LTR.
  • bases are added to the base sequence shown in SEQ ID NO:118, preferably 1 to 20 bases, more preferably 1 to 10 bases, even more preferably 1 to 5 bases, and even more preferably 1 to 3 bases are added to the base sequence or to the 5' or 3' end.
  • LTRs with mutations that combine the addition, substitution, and deletion of these bases are also included in the functional equivalent of the second LTR.
  • the base sequence of the mutated LTR preferably shows 80% or more identity with the base sequence shown in SEQ ID NO:118, more preferably shows 85% or more identity, even more preferably shows 90% or more identity, even more preferably shows 95% or more identity, and even more preferably shows 98% or more identity.
  • leader and trailer refer to partially complementary base sequences present at the ends of the viral genome.
  • Those derived from Sendai virus can be preferably used as leaders and trailers. Both the leader and trailer of Sendai virus are regions with a chain length of approximately 50 bases. Usually, the leader is located on the 5' side and the trailer is located on the 3' side.
  • Examples that are preferably used in one embodiment of the present invention include a leader derived from Sendai virus having the base sequence shown in SEQ ID NO: 119 and a trailer derived from Sendai virus having the base sequence shown in SEQ ID NO: 120.
  • Mutations in the leader and trailer derived from wild-type Sendai virus can also be suitably used in the present invention.
  • the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3.
  • the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. Mutations combining these base substitutions and deletions can also be added.
  • bases are added, preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3 bases are added in the base sequence of the leader and/or trailer or to the 5' end or 3' end.
  • the mutated base sequence preferably exhibits 80% or more identity to the wild-type base sequence, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
  • the base sequence that can be used as the base sequence containing the gene expression control site that controls the expression of the gene of the fusion protein is not particularly limited as long as it can express the fusion protein in the cells, tissues, or living body of a mammal (especially a human) into which the gene encoding the fusion protein is introduced.
  • cytomegalovirus-derived promoter (optionally containing an enhancer), SV40 early promoter, human elongation factor-1 ⁇ (EF-1 ⁇ ) promoter, human ubiquitin C promoter, retroviral Rous sarcoma virus LTR promoter, dihydrofolate reductase promoter, and ⁇ -actin promoter, phosphoglycerate kinase (PGK) promoter, mouse albumin promoter, human albumin promoter, and human ⁇ -1 antitrypsin promoter.
  • EF-1 ⁇ human elongation factor-1 ⁇
  • ubiquitin C promoter human ubiquitin C promoter
  • retroviral Rous sarcoma virus LTR promoter retroviral Rous sarcoma virus LTR promoter
  • dihydrofolate reductase promoter and ⁇ -actin promoter
  • PGK phosphoglycerate kinase
  • a synthetic promoter having the base sequence shown in SEQ ID NO: 121 containing a mouse albumin promoter downstream of a mouse ⁇ -fetoprotein enhancer can be preferably used as the gene expression control site.
  • a chicken ⁇ -actin/MVM chimeric intron having the base sequence shown in SEQ ID NO: 122 may be placed downstream of the mouse ⁇ -fetoprotein enhancer/mouse albumin promoter. By placing such an intron, the expression level of the protein controlled by the gene expression control site can be increased.
  • the base sequence from 1 to 219 is the mouse ⁇ -fetoprotein enhancer
  • the base sequence from 241 to 549 is the mouse albumin promoter.
  • the gene regulatory site may be a promoter of a gene that is expressed in an organ-specific or cell type-specific manner.
  • an organ-specific expression promoter or a cell type-specific expression promoter By using an organ-specific expression promoter or a cell type-specific expression promoter, the gene encoding the fusion protein incorporated into the nucleic acid molecule can be expressed specifically in a desired organ or cell.
  • the nucleic acid molecule contains a gene encoding a conjugate of a fusion protein and an antibody between a first inverted terminal repeat (ITR) and a second inverted terminal repeat (ITR).
  • the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4
  • the antibody is, for example, an anti-transferrin receptor antibody.
  • the following (1) to (4) are examples of the nucleic acid molecule: (1) A gene encoding a conjugate between a fusion protein and an antibody between a first inverted terminal repeat (ITR) and a second inverted terminal repeat (ITR), the gene including a base sequence encoding an internal ribosome binding site downstream of a gene encoding an antibody light chain, and a base sequence encoding a conjugate in which a fusion protein is bound directly or via a linker to the C-terminus or N-terminus of an antibody heavy chain downstream of the internal ribosome binding site; (2) A gene encoding a conjugate between a first inverted terminal repeat (ITR) and a second inverted terminal repeat (ITR), the gene including a base sequence encoding an internal ribosome binding site downstream of the gene encoding a conjugate in which the fusion protein is bound directly or via a linker to the C-terminus or N-terminus of the antibody heavy chain, and a base sequence encoding an antibody light chain downstream
  • the nucleic acid molecule may further include a base sequence including a gene expression control site between the first inverted terminal repeat (ITR) and the gene encoding the conjugate of the fusion protein and the antibody.
  • the base sequence encoding the internal ribosome binding site may be replaced with a base sequence including a gene expression control site.
  • the first gene expression control site and the second gene expression control site are referred to in order from the first inverted terminal repeat (ITR).
  • the base sequence encoding the internal ribosome binding site may be replaced with a base sequence encoding a 2A self-cleaving peptide.
  • the 2A peptide derived from the porcine teschovirus is a suitable example of a 2A self-cleaving peptide.
  • the nucleic acid molecule contains a gene encoding a conjugate of a fusion protein and an antibody between a first long terminal repeat (LTR) and a second long terminal repeat (LTR).
  • the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4
  • the antibody is, for example, an anti-transferrin receptor antibody.
  • the following (1) to (4) are examples of the nucleic acid molecule: (1) A gene encoding a conjugate of a fusion protein and an antibody is contained between a first long terminal repeat (LTR) and a second long terminal repeat (LTR), and the gene contains a base sequence encoding an internal ribosome binding site downstream of a gene encoding the light chain of the antibody, and a base sequence encoding a conjugate in which the fusion protein is bound directly or via a linker to the C-terminus or N-terminus of the heavy chain of the antibody downstream of the internal ribosome binding site; (2) A gene encoding a conjugate of a fusion protein and an antibody between the first long terminal repeat (LTR) and the second long terminal repeat (LTR), the gene including a base sequence encoding an internal ribosome binding site downstream of the gene encoding a conjugate in which the fusion protein is bound directly or via a linker to the C-terminus or N-terminus of the antibody heavy chain, and a base
  • the nucleic acid molecule may further include a base sequence including a gene expression control site between the first long terminal repeat (LTR) and the gene encoding the conjugate of the fusion protein and the antibody.
  • the base sequence encoding the internal ribosome binding site may be replaced with a base sequence including a gene expression control site.
  • the first gene expression control site and the second gene expression control site are referred to in order from the first inverted terminal repeat (ITR).
  • the base sequence encoding the internal ribosome binding site may be replaced with a base sequence encoding a 2A self-cleaving peptide.
  • the 2A peptide derived from the porcine teschovirus is a suitable example of the 2A self-cleaving peptide.
  • the expression of one peptide chain constituting the fusion protein is controlled by the gene expression control site, while the expression of the other peptide chain is controlled by the base sequence encoding the internal ribosome binding site.
  • the two peptide chains pair up within the cell to form a conjugate between the fusion protein and the antibody.
  • the nucleic acid molecule in one embodiment of the present invention can be used in an AAV vector system.
  • AAV inverted terminal repeats
  • the genes of the viral genome integrated into the genome of the host cell are hardly expressed, but when the cell is infected with a helper virus, AAV is excised from the host genome and replication of the infectious virus begins.
  • the helper virus is an adenovirus
  • the genes responsible for the helper function are E1A, E1B, E2A, VA1, and E4.
  • the host cell is a HEK293 cell, which is a human fetal kidney tissue-derived cell transformed with adenovirus E1A and E1B, the E1A and E1B genes are originally expressed in the host cell.
  • the wild-type AAV genome contains two genes, rep and cap.
  • the rep proteins (rep78, rep68, rep52, and rep40) produced by the rep gene are essential for capsid formation and mediate the integration of the viral genome into the chromosome.
  • the cap gene is responsible for the production of three capsid proteins (VP1, VP2, and VP3).
  • a recombinant AAV virion is a virion in which the region containing the rep gene and the cap gene has been replaced with a gene that codes for a foreign protein, and then packaged in a capsid.
  • rAAV virions are used as vectors to deliver foreign genes to cells for therapeutic purposes.
  • Plasmid 1 having a structure containing a base sequence including a first inverted terminal repeat (ITR) and a base sequence including a second inverted terminal repeat (ITR) derived from a virus such as AAV, and a gene encoding a desired protein arranged between these two ITRs;
  • a plasmid (plasmid 2) containing an AAV Rep gene having the function required to integrate the base sequence of the region (including the ITR sequence) sandwiched between the ITR sequences into the genome of a host cell, and a gene encoding an AAV capsid protein;
  • a plasmid (plasmid 3) containing the E2A region, E4 region, and VA1 RNA region of adenovirus.
  • the desired protein is a conjugate of a fusion protein and an antibody.
  • the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody.
  • these three types of plasmids are first introduced into host cells, such as HEK293 cells, in which the adenovirus E1a and E1b genes have been incorporated into the genome, by a general transfection method. Then, a region containing a base sequence containing a first inverted terminal repeat (ITR), a base sequence containing a second inverted terminal repeat (ITR), and a gene encoding a desired protein arranged between these two ITRs is replicated in the host cell, and the resulting single-stranded DNA is packaged into the capsid protein of AAV to form a recombinant AAV virion.
  • ITR inverted terminal repeat
  • ITR second inverted terminal repeat
  • This recombinant AAV virion has infectivity and can be used to introduce a foreign gene into a cell, tissue, or living body.
  • the foreign gene is a gene encoding a conjugate of a fusion protein and an antibody. In the cell or the like to which it is introduced, the conjugate is expressed from this gene.
  • the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody.
  • the Rep protein of adeno-associated virus is encoded by the AAV rep gene.
  • the Rep protein has the necessary function of integrating, for example, the AAV genome into the genome of a host cell via the ITRs present in the genome.
  • Rep68 and Rep78 are the translation products of two types of mRNA transcribed by alternative splicing from the same gene.
  • AAV Rep protein we mean one that includes at least the two types of proteins, Rep68 and Rep78.
  • the base sequence encoding the Rep protein of an adeno-associated virus refers to a base sequence encoding at least Rep68 and Rep78, or a base sequence with a mutation added thereto.
  • the Rep protein is preferably that of an AAV of serotype 2, but is not limited thereto, and may be any of serotypes 1, 3, 4, 5, 6, 7, 8, 9, 10, or 11.
  • a preferred example of a base sequence encoding the Rep protein of a wild-type AAV of serotype 2 is one having the base sequence shown in SEQ ID NO: 123.
  • Rep68 may be a mutant in which the amino acid sequence of wild-type Rep68 of any of AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 has been modified by substitution, deletion, addition, or other means, so long as it exerts its function. Rep68 with these mutations is also included in Rep68.
  • the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3.
  • the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3.
  • Rep68 with mutations that combine these amino acid substitutions and deletions is also Rep68.
  • amino acids are added, preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3 amino acids are added to the amino acid sequence or to the N-terminus or C-terminus of wild-type Rep68.
  • Rep68 with mutations that combine these amino acid additions, substitutions, and deletions is also included in Rep68.
  • the amino acid sequence of the mutated Rep68 preferably exhibits 80% or more identity to the amino acid sequence of wild-type Rep68, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
  • Rep78 may be a mutant in which the amino acid sequence of wild-type Rep78 of any of AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 has been modified by substitution, deletion, addition, or other means, so long as it exerts its function. Rep78 with these mutations is also included in Rep78.
  • the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3.
  • the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3.
  • Rep78 with mutations that combine these amino acid substitutions and deletions is also Rep78.
  • amino acids are added, preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3 amino acids are added to the amino acid sequence or to the N-terminus or C-terminus of wild-type Rep78.
  • Rep78 with mutations that combine these amino acid additions, substitutions, and deletions is also included in Rep78.
  • the amino acid sequence of the mutated Rep78 preferably exhibits 80% or more identity to the amino acid sequence of wild-type Rep78, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
  • the functional equivalent of the AAV Rep protein refers to a substance that can be used functionally in place of Rep68, and to a substance that can be used functionally in place of Rep78.
  • the functional equivalent of the Rep protein may be a mutant of the wild-type Rep protein.
  • the base sequence shown in SEQ ID NO:123 can be modified by substitution, deletion, addition, etc., so long as it encodes functional Rep68 and Rep78.
  • the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3.
  • the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3.
  • a mutation that combines the substitution and deletion of these bases can also be used as a base sequence encoding a Rep protein.
  • a base When a base is added, preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3 bases are added to the base sequence shown in SEQ ID NO:123 or to the 5' end or 3' end.
  • a mutation that combines the addition, substitution, and deletion of these bases can also be used as a nucleic acid molecule encoding a Rep protein.
  • the mutated base sequence preferably exhibits 80% or more identity, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity to the base sequence shown in SEQ ID NO: 123.
  • the start codon of the genes encoding the Rep proteins Rep68 and Rep78 is ACG.
  • the base sequence encoding the Cap protein of an adeno-associated virus refers to a base sequence that encodes at least VP1, a protein that constitutes the capsid of AAV, or a base sequence that includes a mutated base sequence thereof.
  • VP1 is preferably that of AAV serotype 8, but is not limited thereto, and may be any of serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11.
  • a suitable example of a base sequence of the Cap region of serotype 8 is one that includes the base sequence shown in SEQ ID NO: 124.
  • VP1 may be modified by substitution, deletion, addition, or other alteration to the amino acid sequence of wild-type VP1 of any of AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11, so long as it exerts its function. In one embodiment of the present invention, VP1 with these mutations is also included in VP1.
  • the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3.
  • the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3.
  • a mutation that combines the substitution and deletion of these bases can also be used as a base sequence encoding a Cap protein.
  • bases are added, preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3 bases are added to the base sequence shown in SEQ ID NO: 124 or to the 5' end or 3' end.
  • a mutation that combines the addition, substitution, and deletion of these bases can also be used as a nucleic acid molecule encoding a Cap protein.
  • the mutated base sequence preferably exhibits 80% or more identity to the base sequence shown in SEQ ID NO:124, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
  • the AAV vector can produce a recombinant AAV virion in which a nucleic acid molecule containing a foreign gene is packaged between the first and second ITRs.
  • the recombinant AAV virion has infectivity and can be used to introduce a foreign gene into cells, tissues, or living organisms.
  • the foreign gene is a gene encoding a conjugate of a fusion protein and an antibody.
  • the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody.
  • the fusion protein is expressed from this gene.
  • an adenovirus vector system which is a recombinant virion in which a nucleic acid molecule containing a foreign gene is packaged between the first and second ITRs and can be used to introduce a gene into cells, etc.
  • a recombinant adenovirus virion is obtained in which a nucleic acid molecule containing a foreign gene is packaged between the first and second ITRs.
  • the recombinant adenovirus virion has infectivity and can be used to introduce a foreign gene into a cell, tissue, or living organism.
  • Lentiviruses are viruses that belong to the Lentivirus genus in the Orthoretrovirinae subfamily of the Retroviridae family, and have a single-stranded (+) strand RNA genome (ssRNA).
  • the genome of lentiviruses contains essential genes, gag (a region that codes for structural proteins including capsid proteins), pol (a region that codes for a group of enzymes including reverse transcriptase), and env (a region that codes for envelope proteins required for binding to host cells), which are sandwiched between two LTRs (5'LTR and 3'LTR).
  • the genome of lentiviruses contains auxiliary genes, such as Rev (a region that codes for a protein that binds to the RRE (rev responsive element) present in the viral RNA and transports the viral RNA from the nucleus to the cytoplasm), tat (a region that codes for a protein that binds to the TAR in the 5'LTR and increases the promoter activity of the LTR), and others, such as vif, vpr, vpu, and nef.
  • Rev a region that codes for a protein that binds to the RRE (rev responsive element) present in the viral RNA and transports the viral RNA from the nucleus to the cytoplasm
  • tat a region that codes for a protein that binds to the TAR in the 5'LTR and increases the promoter activity of the LTR
  • others such as vif, vpr, vpu, and nef.
  • Lentiviruses are enveloped viruses that infect cells by fusing the envelope with the cell membrane. Lentiviruses are also RNA viruses, and reverse transcriptase is present in virions. After lentivirus infection, single-stranded plus-strand DNA is replicated from the (+) strand RNA genome by reverse transcriptase, and double-stranded DNA is then synthesized. Proteins that are components of virions are expressed from this double-stranded DNA, and the (+) strand RNA genome is packaged into this to cause virions to proliferate.
  • the lentivirus vector has been developed based on the genome of HIV-1, a type of lentivirus, but is not limited to this.
  • a first-generation lentiviral vector consists of three types of plasmids: a packaging plasmid, an Env plasmid, and a transfer plasmid.
  • the packaging plasmid has the gag and pol genes under the control of a CMV promoter, etc.
  • the Env plasmid has the env gene under the control of a CMV promoter.
  • the transfer plasmid has a 5'LTR, an RRE, a gene encoding a desired protein under the control of a CMV promoter, and a 3'LTR.
  • a recombinant virion is obtained in which a nucleic acid molecule containing a foreign gene between the first LTR and the second LTR is packaged in the capsid protein.
  • the packaging plasmid of the first-generation lentiviral vector also contains the auxiliary genes rev, tat, vif, vpr, vpu, and nef derived from the virus.
  • the desired protein in one embodiment of the present invention is a conjugate of a fusion protein and an antibody.
  • the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody.
  • the promoter controlling the gene encoding the desired protein is preferably a promoter other than the CMV promoter, such as an SV40 early promoter, a human elongation factor-1 ⁇ (EF-1 ⁇ ) promoter, a human ubiquitin C promoter, a Rous sarcoma virus LTR promoter of a retrovirus, a dihydrofolate reductase promoter, a ⁇ -actin promoter, a phosphoglycerate kinase (PGK) promoter, a mouse albumin promoter, a human albumin promoter, and a human ⁇ -1 antitrypsin promoter.
  • a promoter other than the CMV promoter such as an SV40 early promoter, a human elongation factor-1 ⁇ (EF-1 ⁇ ) promoter, a human ubiquitin C promoter, a Rous sarcoma virus LTR promoter of a retrovirus, a dihydrofolate reductase promoter, a ⁇ -
  • it can be a synthetic promoter having the base sequence shown in SEQ ID NO: 121, which includes a mouse albumin promoter downstream of a mouse alpha-fetoprotein enhancer (mouse alpha-fetoprotein enhancer/mouse albumin promoter), etc.
  • Second-generation lentiviral vectors like the first-generation, consist of three plasmids: a packaging plasmid, an Env plasmid (envelope plasmid), and a transfer plasmid.
  • a packaging plasmid an Env plasmid (envelope plasmid)
  • Env plasmid envelope plasmid
  • transfer plasmid a transfer plasmid.
  • the non-essential auxiliary genes vif, vpr, vpu, and nef have been deleted from the packaging plasmid.
  • Third-generation lentiviral vectors consist of four types of plasmids: packaging plasmid, Env plasmid (envelope plasmid), Rev plasmid, and transfer plasmid.
  • packaging plasmid envelope plasmid
  • Env plasmid envelope plasmid
  • Rev plasmid Rev plasmid
  • transfer plasmid transfer plasmid.
  • the rev that was in the packaging plasmid in the second generation has been separated to form the Rev plasmid.
  • tat has been deleted from the packaging plasmid.
  • the TAR in the 5'LTR of the transfer plasmid has been replaced with a CMV promoter.
  • the foreign gene is a gene encoding a conjugate of a fusion protein and an antibody. In cells, etc., into which the gene is introduced, the conjugate is expressed from this gene.
  • the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody.
  • Retroviruses have a single-stranded (+) strand RNA genome (ssRNA).
  • the viral genome encodes the gag (encoding structural proteins including capsid protein), pol (encoding a group of enzymes including reverse transcriptase), env (encoding envelope proteins required for binding to host cells) and packaging signal ( ⁇ ) genes, which are flanked by two long terminal repeats (LTR, 5'LTR and 3'LTR).
  • LTR long terminal repeats
  • 5'LTR and 3'LTR packaging signal
  • Retroviral vectors are mainly developed based on mouse leukemia viruses, and the viral genome is divided to lose the ability to self-replicate while maintaining its infectivity in order to eliminate pathogenicity and increase safety.
  • the first generation retroviral vector consists of a packaging plasmid (the viral genome excluding the packaging signal) and an introduction plasmid (the packaging signal, a part of gag, and a foreign gene flanked by 5'LTR and 3'LTR at both ends). Therefore, when homologous recombination occurs in the gag sequence portion that is common to the packaging plasmid and the introduction plasmid, a self-replicating retrovirus, i.e., an RC (replication competent) virus, appears.
  • RC replication competent
  • the 3' LTR of the first generation packaging plasmid is replaced with a polyA addition signal.
  • the third generation is composed of three plasmids, and the second generation packaging plasmid is further divided into a plasmid encoding gag/pol and a plasmid encoding env. This means that for an RC virus to emerge, homologous recombination must occur simultaneously at three sites, making the probability of this occurring extremely low, further increasing safety.
  • these packaging plasmids and transfer plasmids are first transferred into host cells by a general transfection method. Then, the region containing the 5'LTR and 3'LTR and the gene encoding the desired protein located between these two LTRs is replicated in the host cell, and the resulting single-stranded (+) strand RNA is packaged in the retroviral capsid protein to form a recombinant retroviral virion.
  • This recombinant retroviral virion has infectivity and can be used to transfer a foreign gene into a cell, tissue, or living body.
  • the foreign gene is a gene encoding a conjugate of a fusion protein and an antibody.
  • the conjugate is expressed from this gene.
  • the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4
  • the antibody is, for example, an anti-transferrin receptor antibody.
  • the nucleic acid molecule may be encapsulated in a liposome, lipid nanoparticle (LNP), or the like.
  • a liposome is a spherical vesicle with a lipid bilayer, and is composed mainly of phospholipids, particularly phosphatidylcholine.
  • the present invention is not limited to this, and liposomes may contain other lipids, such as egg yolk phosphatidylethanolamine, as long as they form a lipid bilayer. Since cell membranes are mainly composed of phospholipid bilayers, liposomes have the advantage of being highly biocompatible.
  • a lipid nanoparticle is a particle with a diameter of 10 nm to 1000 nm, typically less than about 200 nm, that is mainly composed of lipids, and can encapsulate hydrophobic (lipophilic) molecules and is mainly composed of biocompatible lipids, such as triglycerides, diglycerides, monoglycerides, fatty acids, and steroids. It is believed that when a gene encapsulated in liposomes or lipid nanoparticles is administered to a living body, it fuses directly with the cell membrane of the cell or is taken up by endocytosis, etc., and then migrates to the nucleus and is introduced into the cell.
  • the gene introduction method using liposomes or lipid nanoparticles is superior to gene introduction using a viral vector in that there is no limit to the size of the gene to be introduced and that it is highly safe.
  • the liposomes, lipid nanoparticles, etc. encapsulating the nucleic acid molecule of the present invention can be used to introduce a gene of a conjugate of a fusion protein and an antibody into a cell, tissue, or living body. In the cell, etc. into which the gene is introduced, the fusion protein is expressed from the gene.
  • liposomes, lipid nanoparticles, etc. includes not only the above-mentioned liposomes and lipid nanoparticles, but also polymer nanoparticles, micelles, emulsions, nanoemulsions, microspheres, nanospheres, microcapsules, nanocapsules, dendrimers, nanogels, metal nanoparticles, and any other nano-microparticles that can be used as a drug delivery system (DDS).
  • DDS drug delivery system
  • the behavior of a nucleic acid molecule introduced into a cell, tissue, or living body in the form of a plasmid, encapsulated in a recombinant virus virion, or encapsulated in a liposome, lipid nanoparticle, or the like is exemplified below as (1) to (6).
  • the behavior of the nucleic acid molecule is not limited to these.
  • the nucleic acid molecule is a single-stranded (+) strand RNA, and when introduced into a cell, a gene encoding a conjugate between a fusion protein and an antibody contained in the nucleic acid molecule is translated, thereby expressing the conjugate.
  • the nucleic acid molecule is a single-stranded (+) strand RNA, which, when introduced into a cell, is reverse transcribed to form a single-stranded (+) strand DNA, which is then transcribed and translated to express the conjugate.
  • the nucleic acid molecule is a single-stranded (+) strand RNA or (-) strand RNA, and when introduced into a cell, the nucleic acid molecule is reverse transcribed to form double-stranded DNA, which is then transcribed and translated to express the conjugate.
  • the nucleic acid molecule is a single-stranded (+) or (-) RNA, and when introduced into a cell, the nucleic acid molecule is reverse transcribed to form double-stranded DNA, which then undergoes random or homologous recombination with the genome of the host cell and is integrated into the genome, and the integrated DNA is transcribed and translated to express the conjugate.
  • the nucleic acid molecule is a single-stranded (+) strand DNA, and when introduced into a cell, the nucleic acid molecule is transcribed and translated to express the conjugate.
  • the nucleic acid molecule is double-stranded DNA, and when introduced into a cell, the nucleic acid molecule is transcribed and translated to express the conjugate.
  • Nucleic acid molecules in the form of plasmids, encapsulated in recombinant viral virions, or encapsulated in liposomes, lipid nanoparticles, etc. can be introduced into cells, tissues, or organisms.
  • the nucleic acid molecule When the nucleic acid molecule is to be introduced into a living body, the nucleic acid molecule is administered parenterally, such as subcutaneous injection, intramuscular injection, or intravenous injection, in the form of a plasmid, encapsulated in a recombinant virus virion, or encapsulated in a liposome or lipid nanoparticle.
  • parenterally such as subcutaneous injection, intramuscular injection, or intravenous injection, in the form of a plasmid, encapsulated in a recombinant virus virion, or encapsulated in a liposome or lipid nanoparticle.
  • nucleic acid molecule When the nucleic acid molecule is introduced into a cell, there is no particular limitation on the type of cell, but examples include mesenchymal stem cells, dental pulp-derived stem cells, hematopoietic stem cells, embryonic stem cells, endothelial stem cells, mammary stem cells, intestinal stem cells, hepatic stem cells, pancreatic stem cells, neural stem cells, and iPS cells.
  • Cells that have been transfected with the nucleic acid molecule will express the fusion protein-antibody conjugate, which can then be transplanted into a patient for therapeutic purposes.
  • Example 1 Preparation of wild-type hGALC expression plasmid, HSA-hGALC expression plasmid, and hGALC-HSA expression plasmid
  • pCI-neo vector Promega Corp.
  • restriction enzymes MluI and NotI were treated with restriction enzymes MluI and NotI, and gel extraction and purification were performed.
  • Each DNA fragment treated with a restriction enzyme was mixed with each vector treated with a restriction enzyme, and a ligation reaction was performed at 16°C for 30 to 60 minutes using Ligation high Ver.2 (Toyobo Co., Ltd.), and the DNA fragment was inserted into the pCI vector.
  • Escherichia coli E. coli DH5 ⁇ Competent Cells, Takara Bio
  • Escherichia coli E. coli DH5 ⁇ Competent Cells, Takara Bio
  • LB liquid medium LB Broth, Sigma-Aldrich
  • plasmid DNA was purified from the cells using FastGene Plasmid Mini Kit (Nihon Genetics).
  • the purified plasmid DNA was subjected to restriction enzyme treatment with MluI and NotI, and separated by agarose gel electrophoresis to confirm that the desired insert DNA had been inserted. Plasmids that were confirmed to contain wild-type hGALC, HSA-hGALC, and hGALC-HSA were purified using standard methods.
  • Example 2 Transient expression of wild-type hGALC, HSA-hGALC, and hGALC-HSA
  • plasmids were used in which the genes encoding wild-type hGALC, HSA-hGALC, and hGALC-HSA, respectively, were incorporated into the purified pCI-neo vector obtained in Example 1.
  • ExpiCHO cells were transformed with plasmids incorporating genes encoding wild-type hGALC, HSA-hGALC, and hGALC-HSA, respectively. After transformation, the cells were cultured for 8 days, and wild-type hGALC, HSA-hGALC, and hGALC-HSA were expressed in the culture supernatant, respectively. The culture solutions 6, 7, and 8 days after the start of culture were centrifuged to collect the culture supernatant.
  • Example 3 Confirmation of the expression levels of wild-type hGALC, HSA-hGALC, and hGALC-HSA by transient expression (SDS page electrophoresis) 10 ⁇ L of each culture supernatant obtained in Example 2 was mixed with 8 ⁇ L of 2 ⁇ Sample Buffer (Bio-Rad) and 2 ⁇ L of 2-Mercaptoethanol, and heated at 100° C. for 3 minutes to be heat-denatured under reducing conditions.
  • SDS page electrophoresis SDS page electrophoresis
  • Example 4 Confirmation of expression levels of wild-type hGALC, HSA-hGALC, and hGALC-HSA by transient expression (Western blotting) Electrophoresis was performed in the same manner as described in Example 3, and the nitrocellulose membrane and the gel after electrophoresis were sandwiched between blotting papers soaked in 25 mM Tris buffer/192 mM glycine buffer containing 20% methanol, and the proteins were transferred to the nitrocellulose membrane by applying electricity at 1.0 A and 25 V for 10 minutes in a blotting device.
  • the nitrocellulose membrane after transfer was immersed in PBST containing 5% skim milk and shaken for 1 hour, and then immersed in Anti-GALC antibody (Rabbit polyclonal to GALC, Abcam) diluted to 0.4 ⁇ g/mL and shaken for 1 hour. After washing the membrane with PBST, it was immersed in Anti-mouse IgG (H+L), HRP Conjugate (Promega) solution diluted to 0.4 ⁇ g/mL and shaken for 30 minutes, and washed again with PBST.
  • Anti-GALC antibody Rabbit polyclonal to GALC, Abcam
  • Example 5 Confirmation of expression levels of wild-type hGALC, HSA-hGALC, and hGALC-HSA by transient expression (enzyme activity measurement)
  • the culture supernatant obtained in Example 2 was diluted 100-fold with citrate buffer (pH 4.5) containing 0.5% Triton X-100 to prepare a sample solution.
  • the sample solution was further diluted appropriately as necessary and used for measurement.
  • a standard solution was prepared by serially diluting 4-MU (4-Methylumbelliferone, Sigma-Aldrich) from 75 to 4.39 ⁇ M with 100 mM citrate buffer (pH 4.2) containing 0.1% BSA.
  • a substrate solution was prepared by diluting 4-Methylumbelliferyl- ⁇ -D-galactopyranoside (Sigma-Aldrich), an artificial substrate for hGALC, to 1 mmol/L with citrate buffer (pH 4.5) containing 0.5% Triton X-100. 25 ⁇ L/well of the sample solution or standard solution was added to a microplate, and 25 ⁇ L of substrate solution was added to each well and mixed by stirring with a plate shaker. After the plate was left at 37°C for 1 hour, 150 ⁇ L of 200 mmol/L glycine-NaOH buffer (pH 10.6) was added to each well to stop the reaction.
  • the fluorescence intensity of each well was then measured using a fluorescence plate reader (Gemini XPS, Molecular Devices, Inc.) (excitation wavelength 365 nm, fluorescence wavelength 460 nm). This fluorescence intensity is proportional to the concentration of 4-MU (4-Methylumbelliferone) contained in the solution in each well.
  • a calibration curve was created based on the measurement results of the standard solution, and the measured values of each sample solution were interpolated to determine the enzyme activity ( ⁇ M/hour). The unit of enzyme activity indicates the amount of substrate decomposed per hour.
  • Electrophoretic images and Western blotting images obtained in Example 3 are shown in Figures 15(b) and (c), respectively. From the Western blotting images, bands corresponding to wild-type hGALC, HSA-hGALC, and hGALC-HSA on the electrophoretic images were identified. The expression levels (in terms of molecular number) of the parts of wild-type hGALC, HSA-hGALC corresponding to hGALC, and the parts of hGALC-HSA corresponding to hGALC, estimated from the density of the bands in the Western blotting images, were approximately correlated with the expression levels (in terms of enzyme activity) shown in Figure 15(a) and Table 1.
  • the relative values of the expression levels shown as the expression levels (in terms of enzyme activity) in Table 1 can be said to be the expression levels (in terms of molecular number) of the parts of wild-type hGALC, HSA-hGALC corresponding to hGALC, and hGALC-HSA corresponding to hGALC.
  • these results show that by making wild-type hGALC into a fusion protein with HSA, it is possible to express a larger number of molecules of hGALC, which is difficult to mass-produce as a recombinant in its wild form, without reducing its specific activity by making it into a fusion protein.
  • the specific activity of hGALC in the fusion protein is calculated by multiplying the enzyme activity of hGALC per unit mass of the fusion protein ( ⁇ M/hour/mg protein) by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hGALC).
  • Example 7 Cell viability, etc. in transient expression of wild-type hGALC, HSA-hGALC, and hGALC-HSA
  • the viable cell density and cell viability in the culture medium 6, 7, and 8 days after the start of culture of transformed ExpiCHO cells were measured using an EVE Automated Cell Counter (Nano EnTek).
  • Table 2 shows the viable cell density and cell viability in the culture medium 6, 7, and 8 days after the start of culture for each of the cells expressing wild-type hGALC, HSA-hGALC, and hGALC-HSA.
  • the relative viability ratios were 1.1 and 1.1 for HSA-hGALC and hGALC-HSA expressing cells, respectively, 6 days after the start of culture, 1.3 and 1.2 for HSA-hGALC and hGALC-HSA expressing cells, respectively, 7 days after the start of culture, and 1.5 and 1.2 for HSA-hGALC and hGALC-HSA expressing cells, respectively, 8 days after the start of culture.
  • Example 8 Summary The above results show that when producing hGALC as a recombinant protein, producing hGALC in the form of a fusion protein of HSA-hGALC or hGALC-HSA makes it possible to obtain about 1.5 to 3.6 times the number of molecules of hGALC without decreasing the specific activity of hGALC.
  • the method of producing recombinant hGALC by expressing hGALC in the form of a fusion protein of HSA-hGALC or hGALC-HSA can be said to be an extremely effective means for producing a large amount of recombinant hGALC.
  • hGALC in the form of a fusion protein of HSA-hGALC or hGALC-HSA, cell death during culture can be suppressed, making it easier to purify the expressed fusion protein. This also increases the purification efficiency during purification.
  • Example 9 SE-HPLC analysis of wild-type hGALC, HSA-hGALC, and hGALC-HSA obtained by transient expression
  • the culture supernatant of each protein was subjected to a purification process and then subjected to SE-HPLC analysis.
  • the wild-type hGALC, HSA-hGALC, and hGALC-HSA adsorbed on the strong anion exchange column were then eluted with a salt concentration gradient using 25 mM MES buffer (pH 6.5) containing 500 mM NaCl.
  • each eluate containing wild-type hGALC, HSA-hGALC, and hGALC-HSA obtained in the above purification process was loaded onto the column, and then 0.2 M sodium phosphate buffer solution containing 10% ethanol was passed through the column at a flow rate of 0.5 mL/min. During this time, the absorbance (measurement wavelength 215 nm) of the effluent from the column was measured to obtain an elution profile.
  • the elution profile is shown in Figure 16.
  • Wild-type hGALC has two main peaks, which are considered to correspond to the hGALC tetramer and the hGALC dimer, respectively, from the left side of the elution profile.
  • HSA-hGALC and hGALC-HSA both have one main peak, which is considered to be the peak of the HSA-hGALC or hGALC-HSA monomer, respectively. This shows that, compared to the case where wild-type hGALC is expressed in cells, when the fusion protein of HSA and hGALC is expressed, a molecule having hGALC activity is stably obtained as a monomer.
  • the mass ratio of the monomer to the total expression amount is 90% or more in each of HSA-hGALC and hGALC-HSA.
  • HSA-hGALC and hGALC-HSA can be obtained as homogeneous monomers, so from the viewpoint of production control, the method of producing hGALC as a fusion protein of hGALC with HSA can be said to be superior.
  • Example 10 Preparation of plasmids expressing conjugates of anti-hTfR antibody and fusion protein of HSA and hGALC Plasmids were prepared that express the four types of proteins shown below as conjugates of an anti-hTfR antibody (Fab) consisting of a heavy chain having the amino acid sequence shown in SEQ ID NO:25 and a light chain having the amino acid sequence shown in SEQ ID NO:23, and a fusion protein of HSA and hGALC.
  • Fab anti-hTfR antibody
  • a conjugate consisting of a fusion protein in which the N-terminus of HSA-hGALC is linked via a linker to the C-terminus of the heavy chain of an anti-hTfR antibody (Fab) and the light chain of an anti-hTfR antibody (Fab) (referred to as Fab-HSA-hGALC);
  • a conjugate consisting of a fusion protein in which the C-terminus of HSA-hGALC is linked via a linker to the N-terminus of the heavy chain of an anti-hTfR antibody (Fab) and the light chain of an anti-hTfR antibody (Fab) (HSA-hGALC-Fab)
  • a conjugate consisting of a fusion protein in which the N-terminus of hGALC-HSA is linked via a linker to the C-terminus of the heavy chain of an anti-hTfR antibody (Fab) and a light chain of an anti-hTfR antibody (Fab) (referred to as Fab-hGALC-HSA);
  • the pCI vector (Promega) was digested with NheI and NotI (Takara Bio), and the above DNA fragment was inserted into it by ligation.
  • the resulting plasmid was named pCI MCS-modified vector ( Figure 17).
  • the pCI MCS-modified vector was digested with BamHI and BglII (Takara Bio), and a DNA fragment containing the CMV immediate-early enhancer/promoter region, multicloning site, and polyA sequence was excised.
  • the pCI-neo vector (Promega) was digested with BamHI and treated with CIAP (Takara Bio), and the excised DNA fragment was inserted into it by ligation.
  • the resulting plasmid was named Dual(+)pCI-neo vector ( Figure 18).
  • the Dual(+)pCI-neo vector into which the gene encoding the anti-hTfR antibody Fab heavy chain-HSA-hGALC was inserted was digested with restriction enzymes AscI and AfeI (New England BioLabs), and a DNA fragment containing the base sequence shown in SEQ ID NO:36, including a gene encoding the light chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO:23, digested with AscI and AfeI, was inserted into the vector.
  • the resulting plasmid was used as a Fab-HSA-hGALC expression plasmid.
  • an HSA-hGALC-Fab expression plasmid was prepared using a DNA fragment containing the base sequence shown in SEQ ID NO: 31, which contains a gene encoding a conjugate in which the C-terminus of HSA-hGALC is linked via a linker to the N-terminus of the heavy chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 30, and a DNA fragment containing the base sequence shown in SEQ ID NO: 36, which contains a gene encoding the light chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 23.
  • Fab anti-hTfR antibody
  • a Fab-hGALC-HSA expression plasmid was prepared using a DNA fragment containing the base sequence shown in SEQ ID NO: 33, which contains a gene encoding a conjugate in which the N-terminus of hGALC-HSA is linked via a linker to the C-terminus of the heavy chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 32, and a DNA fragment containing the base sequence shown in SEQ ID NO: 36, which contains a gene encoding the light chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 23.
  • Fab anti-hTfR antibody
  • an hGALC-HSA-Fab expression plasmid was prepared using a DNA fragment containing the base sequence shown in SEQ ID NO: 35, which contains a gene encoding a conjugate in which the C-terminus of hGALC-HSA is linked via a linker to the N-terminus of the heavy chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 34, and a DNA fragment containing the base sequence shown in SEQ ID NO: 36, which contains a gene encoding the light chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 23.
  • SEQ ID NO: 35 which contains a gene encoding a conjugate in which the C-terminus of hGALC-HSA is linked via a linker to the N-terminus of the heavy chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 34
  • Fab anti-hTfR antibody
  • each conjugate expression plasmid prepared in Example 10 was used to transform ExpiCHO cells. After transformation, the cells were cultured for 8 days, and Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab were expressed in the culture supernatant. After 8 days from the start of culture, the culture solutions were centrifuged to collect the culture supernatants.
  • Example 12 Confirmation of expression levels of Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab by transient expression (enzyme activity measurement) Using the culture supernatant obtained in Example 11 as a sample solution, the expression levels of Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab by transient expression were confirmed by enzyme activity measurement in the same manner as described in Example 5.
  • FIG. 19 shows the results of enzyme activity measurement of wild-type hGALC, Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab.
  • the values obtained in Example 2 using the culture supernatant 8 days after the start of culturing cells expressing wild-type hGALC as the sample solution were used for the enzyme activity measurement results.
  • Example 13 SE-HPLC analysis of Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab obtained by transient expression
  • the culture supernatant of each protein was subjected to a purification process, followed by SE-HPLC analysis.
  • the Capture Select TM CH1-XL column is an affinity column in which a ligand having the property of specifically binding to the CH1 domain of an IgG antibody is immobilized on a carrier.
  • the column was then washed by supplying the same buffer in a volume 5 times the column volume.
  • the column was then further washed by supplying 25 mM MES buffer (pH 6.5) in a volume 3 times the column volume.
  • the bound proteins were then eluted with 5 column volumes of 20 mM acetate buffer (pH 4.0) and immediately neutralized in a container that had previously contained 250 mM MES buffer (pH 6.0) and 2 M NaCl solution.
  • each eluate containing Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab obtained in the above purification process was loaded onto the column, and 0.2 M sodium phosphate buffer solution containing 10% ethanol was further passed through the column at a flow rate of 0.5 mL/min. During this time, the absorbance (measurement wavelength 215 nm) of the effluent from the column was measured to obtain an elution profile.
  • these conjugates When expressed as recombinant proteins, these conjugates can be obtained as homogeneous monomers, so from the viewpoint of production control, the method of producing hGALC as a conjugate of HSA and an antibody (here, Fab) can be said to be excellent.
  • Examples 14 to 20 show the experimental results of a fusion protein of HSA and hGBA.
  • Example 14 Preparation of wild-type hGBA expression plasmid pE-neo7 vector and pCAGIPuro vector (Miyahara M. et.al., J. Biol. Chem. 275, 613-618(2000)) were treated with restriction enzymes BamHI and NotI (Takara Bio), respectively, and separated by agarose gel electrophoresis.
  • the pE-neo7 vector was prepared by the method described in International Publication WO2012/101998. After EtBr staining, the band containing the target DNA fragment was excised under UV irradiation, and DNA was extracted from the gel using QIAEX II Gel Extraction Kit (QIAGEN).
  • Ligation reaction was performed at 16°C for 60 minutes using Ligation high Ver.2 (Toyobo Co., Ltd.) to complete the pEI-puro vector.
  • the DNA fragment was mixed with the pEI-puro vector that had been treated with the restriction enzymes MluI and NotI, and a ligation reaction was carried out at 16°C for 60 minutes using Ligation high Ver.2 to complete the pEI-puro-hGBA vector.
  • the pEI-puro-hGBA vector and the pE-mIRES-GS vector were treated with restriction enzymes MluI and NotI, and then gel extracted and purified.
  • the pE-mIRES-GS vector was prepared by the method described in International Publication WO2013/161958.
  • the vectors treated with the restriction enzymes were mixed and ligated for 60 minutes at 16°C using Ligation high Ver.2 to complete the pEmIGS-hGBA vector ( Figure 21). ).
  • the pEmIGS-hGBA vector and the pCIneo vector (Promega) were treated with restriction enzymes MluI and NotI, and gel extracted and purified. Then, the vectors treated with the restriction enzymes were mixed and ligated at 16°C for 60 minutes using Ligation high Ver.2 to complete the wild-type hGBA expression plasmid, the pCIneo-hGBA vector ( Figure 22).
  • Example 15 Preparation of HSA-hGBA expression plasmid and hGBA-HSA expression plasmid
  • a DNA fragment having a base sequence shown in SEQ ID NO: 40 was synthesized, including a gene encoding HSA-hGBA shown in SEQ ID NO: 39, which is a fusion protein in which the C-terminus of wild-type HSA (SEQ ID NO: 3) and the N-terminus of wild-type hGBA (SEQ ID NO: 37) are linked via a linker sequence shown in SEQ ID NO: 9.
  • a ligation reaction was also carried out in the same manner with a DNA fragment having the base sequence shown in SEQ ID NO: 42, which contains a gene encoding hGBA-HSA shown in SEQ ID NO: 41, a fusion protein in which the C-terminus of wild-type hGBA (SEQ ID NO: 37) and the N-terminus of wild-type HSA (SEQ ID NO: 3) are linked via a linker sequence shown in SEQ ID NO: 9, to complete the pCIneo-hGBA-HSA vector, an hGBA-HSA expression plasmid.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Plant Pathology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

One embodiment of the present invention makes it possible to express, in the form of a fusion protein with a serum albumin (SA), a physiologically active protein that is expressed in a low amount and/or is less active when a host cell such as a CHO cell is normally used to express the physiologically active protein as a recombinant protein, thereby efficiently producing the physiologically active protein as a highly active recombinant protein. The fusion protein may be obtained by bonding the physiologically active protein to the amino-terminal side of the SA, or may be obtained by bonding the physiologically active protein to a carboxyl terminal. Further, said fusion protein can be a body coupled with a ligand or an antibody.

Description

血清アルブミンと生理活性を有する蛋白質との融合蛋白質Fusion protein of serum albumin and a biologically active protein
 本発明は,血清アルブミン(SA)と生理活性を有する蛋白質(生理活性蛋白質)とを結合させた融合蛋白質及びその製造方法に関する。かかる融合蛋白質は,例えば,SAのC末端と生理活性蛋白質のN末端を結合させたものに関する。生理活性蛋白質には,これを哺乳動物細胞等の宿主細胞に当該生理活性蛋白質をコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低いものがある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるSAと生理活性蛋白質との融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。但し,SAと融合させるべき生理活性蛋白質に特に制限なく,全ての生理活性蛋白質をSAとの融合蛋白質とさせることができる。 The present invention relates to a fusion protein in which serum albumin (SA) is bound to a protein having physiological activity (biologically active protein) and a method for producing the same. Such a fusion protein relates, for example, to one in which the C-terminus of SA is bound to the N-terminus of a biologically active protein. Some biologically active proteins have low expression levels and/or low activity when expressed as a recombinant protein by introducing a gene encoding the biologically active protein into a host cell such as a mammalian cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of SA and a biologically active protein that can be efficiently produced as a highly active recombinant protein, and also relates to a method for producing the fusion protein. However, there is no particular restriction on the biologically active protein to be fused to SA, and any biologically active protein can be made into a fusion protein with SA.
 本発明は,特に血清アルブミン(SA)とリソソーム酵素とを結合させた融合蛋白質に関し,例えば,SAのC末端とリソソーム酵素のN末端,もしくはリソソーム酵素のC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。リソソーム酵素には,これを哺乳動物細胞等の宿主細胞に当該リソソーム酵素をコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低いものがある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるリソソーム酵素とSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。但し,リソソーム酵素と融合させるべき生理活性蛋白質に特に制限なく,全てのリソソーム酵素をSAとの融合蛋白質とさせることができる。 The present invention relates in particular to a fusion protein in which serum albumin (SA) is bound to a lysosomal enzyme, for example, the C-terminus of SA is bound to the N-terminus of a lysosomal enzyme, or the C-terminus of a lysosomal enzyme is bound to the N-terminus of SA, either directly or via a linker. Some lysosomal enzymes have low expression levels and/or low activity when expressed as recombinant proteins by introducing a gene encoding the lysosomal enzyme into a host cell such as a mammalian cell, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of a lysosomal enzyme and SA that can be efficiently produced as a highly active recombinant protein, and also relates to a method for producing the fusion protein. However, there is no particular restriction on the physiologically active protein to be fused to the lysosomal enzyme, and any lysosomal enzyme can be made into a fusion protein with SA.
 本発明は,また特に血清アルブミン(SA)とガラクトシルセラミダーゼ(GALC)とを結合させた融合蛋白質に関し,例えば,SAのC末端とGALCのN末端,もしくはGALCのC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。GALCは,これを哺乳動物細胞等の宿主細胞にGALCをコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低い場合がある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるGALCとSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。 The present invention also relates to a fusion protein in which serum albumin (SA) and galactosylceramidase (GALC) are bound, for example, by binding the C-terminus of SA to the N-terminus of GALC, or the C-terminus of GALC to the N-terminus of SA, either directly or via a linker. When a gene encoding GALC is introduced into a host cell such as a mammalian cell and the protein is expressed as a recombinant protein, the expression level and/or activity of GALC may be low, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of GALC and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing the fusion protein.
 本発明は,また特に血清アルブミン(SA)とグルコセレブロシダーゼ(GBA)とを結合させた融合蛋白質に関し,例えば,SAのC末端とGBAのN末端,もしくはGBAのC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。GBAは,これを哺乳動物細胞等の宿主細胞にGBAをコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は活性が低い場合がある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるGBAとSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。 The present invention also relates to a fusion protein in which serum albumin (SA) and glucocerebrosidase (GBA) are bound, for example, by binding the C-terminus of SA to the N-terminus of GBA, or the C-terminus of GBA to the N-terminus of SA, either directly or via a linker. When a gene encoding GBA is introduced into a host cell such as a mammalian cell and expressed as a recombinant protein, GBA may have a low expression level and/or low activity, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of GBA and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing said fusion protein.
 本発明は,血清アルブミン(SA)とサイトカインとを結合させた融合蛋白質に関し,例えば,SAのC末端とサイトカインのN末端,もしくはサイトカインのC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。サイトカインには,これを哺乳動物細胞等の宿主細胞に当該サイトカインをコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低いものがある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるサイトカインとSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。但し,サイトカインと融合させるべき生理活性蛋白質に特に制限なく,全てのサイトカインをSAとの融合蛋白質とさせることができる。 The present invention relates to a fusion protein in which serum albumin (SA) and a cytokine are bound, for example, by binding the C-terminus of SA to the N-terminus of a cytokine, or the C-terminus of a cytokine to the N-terminus of SA, either directly or via a linker. Some cytokines have low expression levels and/or low activity when expressed as a recombinant protein by introducing a gene encoding the cytokine into a host cell such as a mammalian cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of cytokine and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing the fusion protein. However, there is no particular restriction on the physiologically active protein to be fused with the cytokine, and any cytokine can be made into a fusion protein with SA.
 本発明は,特に血清アルブミン(SA)とインターロイキンとを結合させた融合蛋白質に関し,例えば,SAのC末端とインターロイキンのN末端,もしくはインターロイキンのC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。インターロイキンには,これを哺乳動物細胞等の宿主細胞に当該インターロイキンをコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は活性が低いものがある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるインターロイキンとSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。但し,インターロイキンと融合させるべき生理活性蛋白質に特に制限なく,全てのインターロイキンをSAとの融合蛋白質とさせることができる。 The present invention relates in particular to a fusion protein in which serum albumin (SA) and an interleukin are bound, for example, by binding the C-terminus of SA to the N-terminus of an interleukin, or the C-terminus of an interleukin to the N-terminus of SA, either directly or via a linker. Some interleukins have low expression levels and/or low activity when expressed as a recombinant protein by introducing a gene encoding the interleukin into a host cell such as a mammalian cell, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of an interleukin and SA that can be efficiently produced as a highly active recombinant protein, and also relates to a method for producing the fusion protein. However, there is no particular restriction on the physiologically active protein to be fused with the interleukin, and all interleukins can be made into a fusion protein with SA.
 本発明は,特に血清アルブミン(SA)とインターロイキン10(IL-10)とを結合させた融合蛋白質に関し,例えば,SAのC末端とIL-10のN末端,もしくはIL-10のC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。IL-10は,これを哺乳動物細胞等の宿主細胞にIL-10をコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低い場合がある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるIL-10とSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。 The present invention relates in particular to a fusion protein in which serum albumin (SA) and interleukin 10 (IL-10) are bound, for example, by binding the C-terminus of SA to the N-terminus of IL-10, or the C-terminus of IL-10 to the N-terminus of SA, either directly or via a linker. When IL-10 is expressed as a recombinant protein by introducing a gene encoding IL-10 into a host cell such as a mammalian cell, the expression level and/or activity of IL-10 may be low, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of IL-10 and SA that can be efficiently produced as a highly active recombinant protein, and also to a method for producing said fusion protein.
 本発明は,血清アルブミン(SA)と神経栄養因子とを結合させた融合蛋白質に関し,例えば,SAのC末端と神経栄養因子のN末端,もしくは神経栄養因子のC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。神経栄養因子には,これを哺乳動物細胞等の宿主細胞に当該神経栄養因子をコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低いものがある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかる神経栄養因子とSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。但し,神経栄養因子と融合させるべき生理活性蛋白質に特に制限なく,全ての神経栄養因子をSAとの融合蛋白質とさせることができる。 The present invention relates to a fusion protein in which serum albumin (SA) and a neurotrophic factor are bound, for example, by binding the C-terminus of SA to the N-terminus of a neurotrophic factor, or the C-terminus of a neurotrophic factor to the N-terminus of SA, either directly or via a linker. Some neurotrophic factors have low expression levels and/or low activity when expressed as recombinant proteins by introducing a gene encoding the neurotrophic factor into a host cell such as a mammalian cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of a neurotrophic factor and SA that can be efficiently produced as a highly active recombinant protein, and also relates to a method for producing the fusion protein. However, there is no particular restriction on the physiologically active protein to be fused to the neurotrophic factor, and any neurotrophic factor can be made into a fusion protein with SA.
 本発明は,特に血清アルブミン(SA)と脳由来神経栄養因子(BDNF)とを結合させた融合蛋白質に関し,例えば,SAのC末端とBDNFのN末端,もしくはBDNFのC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。BDNFは,これを哺乳動物細胞等の宿主細胞にBDNFをコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低い場合がある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるBDNFとSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。 The present invention relates in particular to a fusion protein in which serum albumin (SA) and brain-derived neurotrophic factor (BDNF) are bound, for example, by binding the C-terminus of SA to the N-terminus of BDNF, or the C-terminus of BDNF to the N-terminus of SA, either directly or via a linker. When a gene encoding BDNF is introduced into a host cell such as a mammalian cell and BDNF is expressed as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium, BDNF may have a low expression level and/or low activity. The present invention relates to such a fusion protein of BDNF and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing said fusion protein.
 本発明は,また特に血清アルブミン(SA)と神経成長因子(NGF)とを結合させた融合蛋白質に関し,例えば,SAのC末端とNGFのN末端,もしくはNGFのC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。NGFは,これを哺乳動物細胞等の宿主細胞にNGFをコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低い場合がある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるNGFとSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。 The present invention also relates to a fusion protein in which serum albumin (SA) and nerve growth factor (NGF) are bound, for example, by binding the C-terminus of SA to the N-terminus of NGF, or the C-terminus of NGF to the N-terminus of SA, either directly or via a linker. When NGF is expressed as a recombinant protein by introducing a gene encoding NGF into a host cell such as a mammalian cell, the expression level and/or activity may be low, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of NGF and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing the fusion protein.
 本発明は,また特に血清アルブミン(SA)とニューロトロフィン3(NT-3)とを結合させた融合蛋白質に関し,例えば,SAのC末端とNT-3のN末端,もしくはNT-3のC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。NT-3は,これを哺乳動物細胞等の宿主細胞にNT-3をコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低い場合がある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるNT-3とSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。 The present invention also relates to a fusion protein in which serum albumin (SA) and neurotrophin 3 (NT-3) are bound, for example, by binding the C-terminus of SA to the N-terminus of NT-3, or the C-terminus of NT-3 to the N-terminus of SA, either directly or via a linker. When NT-3 is expressed as a recombinant protein by introducing a gene encoding NT-3 into a host cell such as a mammalian cell, the expression level and/or activity may be low, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of NT-3 and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing said fusion protein.
 本発明は,また特に血清アルブミン(SA)とニューロトロフィン4(NT-4)とを結合させた融合蛋白質に関し,例えば,SAのC末端とNT-4のN末端,もしくはNT-4のC末端とSAのN末端とを直接又はリンカーを介して結合させたものに関する。NT-4は,これを哺乳動物細胞等の宿主細胞にNT-4をコードする遺伝子を導入して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低い場合がある。本発明は,活性の高い組換え蛋白質として効率よく製造できる,かかるNT-4とSAとの融合蛋白質に関し,また当該融合蛋白質を製造するための方法に関する。 The present invention also relates to a fusion protein in which serum albumin (SA) and neurotrophin 4 (NT-4) are bound, for example, by binding the C-terminus of SA to the N-terminus of NT-4, or the C-terminus of NT-4 to the N-terminus of SA, either directly or via a linker. When NT-4 is expressed as a recombinant protein by introducing a gene encoding NT-4 into a host cell such as a mammalian cell, the expression level and/or activity may be low, particularly when the recombinant protein is expressed so as to be secreted from the cell and accumulated in the culture medium. The present invention relates to such a fusion protein of NT-4 and SA that can be efficiently produced as a highly active recombinant protein, and to a method for producing said fusion protein.
 リソソーム病の一種であるクラッベ病は,ガラクトシルセラミドリピドーシス,又はグロボイド細胞白質ジストロフィーとしても知られ,リソソーム内においてスフィンゴ脂質の分解に必要となるガラクトシルセラミダーゼ(ガラクトセレブロシダーゼ,GALC)の,遺伝子異常に伴う当該酵素の活性低下又は欠損を原因とする遺伝子疾患である。ガラクトシルセラミダーゼ(GALC)は,ガラクトシルスフィンゴシン,ガラクトセレブロシド等を基質とし,これら基質の分子中のガラクトースエステル結合を加水分解する反応を触媒する作用を持つ。クラッベ病の患者では,GALCの欠損により,基質であるガラクトシルスフィンゴシン等が体内に蓄積する。ガラクトシルスフィンゴシンは強い細胞毒性を持つことが知られており,その蓄積により,中枢神経・末梢神経の髄鞘又はミエリンが破壊される脱髄が引き起こされる。クラッベ病は進行性の疾患であり,重症例では,知的障害,麻痺,失明,難聴,仮性球麻痺が認められる。クラッベ病は発症の時期により,生後3ヶ月~6ヶ月程度で発症し易刺激性,退行等がみられ2~3年で死亡することが多い乳児型,生後6ヶ月~3歳程度で発症し易刺激性,精神運動発達遅延,退行がみられる後期乳児型,3歳~10歳程度で発症し視力障害,歩行障害,失調等がみられ緩やかに進行する若年型,及び10歳程度以降で発症し精神症状等がみられる成人型に分類される。 Krabbe disease, a type of lysosomal disease, is also known as galactosylceramide lipidosis or globoid cell leukodystrophy. It is a genetic disease caused by a genetic abnormality that reduces the activity or deficiency of galactosylceramidase (galactocerebrosidase, GALC), which is necessary for the breakdown of sphingolipids in lysosomes. Galactosylceramidase (GALC) uses galactosylsphingosine, galactocerebroside, and other substrates as catalysts for the hydrolysis of the galactose ester bonds in the molecules of these substrates. In patients with Krabbe disease, the substrate galactosylsphingosine and other substances accumulate in the body due to the deficiency of GALC. Galactosylsphingosine is known to have strong cytotoxicity, and its accumulation causes demyelination, which destroys the myelin sheath or myelin in the central and peripheral nerves. Krabbe disease is a progressive disease, and in severe cases, intellectual disability, paralysis, blindness, hearing loss, and pseudobulbar palsy are observed. Depending on the time of onset, Krabbe disease is classified into infantile type, which begins at about 3 to 6 months of age and is characterized by irritability and regression, and often results in death within 2 to 3 years; late infantile type, which begins at about 6 months to 3 years of age and is characterized by irritability, delayed psychomotor development, and regression; juvenile type, which begins at about 3 to 10 years of age and progresses slowly, and is characterized by visual impairment, gait disturbance, ataxia, etc.; and adult type, which begins at about 10 years of age or older and is characterized by psychiatric symptoms, etc.
 リソソーム病の一種であるゴーシェ病は,リソソーム内において生体糖脂質であるグルコセレブロシドの分解に必要となるグルコセレブロシダーゼ(β-グルコシダーゼ,GBA)の,遺伝子異常に伴う当該酵素の活性低下又は欠損を原因とする遺伝子疾患である。グルコセレブロシダーゼ(GBA)は,グルコセレブロシドを基質とし,当該基質の分子中の糖と脂質の脱水縮合部位を加水分解する反応を触媒する作用を持つ。ゴーシェ病の患者では,GBAの欠損により,基質であるグルコセレブロシド等が体内に蓄積する。グルコセレブロシドは,特に肝臓・脾臓・骨などのマクロファージに蓄積することにより,脾機能の低下に伴う貧血や血小板減少,あるいは肝脾腫,骨痛,骨折,中枢神経障害等が引き起こされる。中枢神経障害は,グルコセレブロシドのリゾ体であるグルコシルスフィンゴシンの脳内蓄積が影響していると考えられている。ゴーシェ病は神経症状の有無と重症度により,最も軽度であり発症年齢が幼児から成人にわたる,神経症状を伴わず肝臓・脾臓の肥大,貧血,血小板の減少,骨折等の症状が緩慢に経過するI型(非神経型),乳児期に発症しI型の症状に加えて精神運動発達遅滞,痙攣,項部後屈などの神経症状が急速に進行して2歳までに酸素欠乏によって死亡する最も重篤なII型(急性神経型),及び乳幼児期に徐々に発症しII型に比べ緩慢な経過を辿る進行性のIII型(亜急性神経型)に分類される。 Gaucher disease, a type of lysosomal disease, is a genetic disease caused by a genetic abnormality that results in a decrease in the activity or deficiency of glucocerebrosidase (β-glucosidase, GBA), an enzyme required for the breakdown of the biological glycolipid glucocerebroside in the lysosome. Glucocerebrosidase (GBA) catalyzes the hydrolysis of the dehydration condensation site of sugar and lipid in the substrate molecule using glucocerebroside as a substrate. In patients with Gaucher disease, the substrate glucocerebroside and other substances accumulate in the body due to a deficiency of GBA. Glucocerebroside accumulates in macrophages, particularly in the liver, spleen, and bones, causing anemia and thrombocytopenia due to decreased splenic function, as well as hepatosplenomegaly, bone pain, fractures, and central nervous system disorders. It is believed that the central nervous system disorders are caused by the accumulation of glucosylsphingosine, the lyso form of glucocerebroside, in the brain. Gaucher disease is classified according to the presence or absence and severity of neurological symptoms into the mildest type I (non-neurological type), which occurs at ages ranging from infancy to adulthood and is characterized by slowly progressing symptoms such as enlargement of the liver and spleen, anemia, decreased platelets, and fractures without accompanying neurological symptoms; the most severe type II (acute neurological type), which begins in infancy and, in addition to the symptoms of type I, rapidly progresses with neurological symptoms such as psychomotor developmental delay, convulsions, and neck retroversion, resulting in death from oxygen deficiency by the age of two; and the progressive type III (subacute neurological type), which develops gradually during infancy and progresses more slowly than type II.
 クラッベ病及びゴーシェ病以外のリソソーム病もリソソーム酵素の遺伝的欠損を原因とする。リソソーム病の中でもファブリー病,ハンター症候群等に関しては,遺伝的に欠損する酵素を遺伝子組み換え技術により組換え酵素として製造し,これを患者に投与する酵素補充療法が行われている。  Lysosomal diseases other than Krabbe disease and Gaucher disease are also caused by genetic deficiencies in lysosomal enzymes. For lysosomal diseases such as Fabry disease and Hunter syndrome, enzyme replacement therapy is performed in which the genetically deficient enzyme is produced as a recombinant enzyme using genetic engineering technology and administered to the patient.
 ヒトGALC(hGALC)をコードする遺伝子は,1993年に単離されている(非特許文献1)。しかし,この遺伝子を用いて製造された組換えヒトGALC(rhGALC)を有効成分として含む,クラッベ病の酵素補充療法として用いるための医薬品はない。 The gene encoding human GALC (hGALC) was isolated in 1993 (Non-Patent Document 1). However, there are no drugs for use as enzyme replacement therapy for Krabbe disease that contain recombinant human GALC (rhGALC) produced using this gene as the active ingredient.
 ヒトGBA(hGBA)をコードする遺伝子は,1986年に単離されている(非特許文献2)。しかし,この遺伝子を用いて製造された組換えヒトGBA(rhGBA)を有効成分として含む,ゴーシェ病の酵素補充療法として用いるための医薬品はない。 The gene encoding human GBA (hGBA) was isolated in 1986 (Non-Patent Document 2). However, there are no drugs for use as enzyme replacement therapy for Gaucher disease that contain recombinant human GBA (rhGBA) produced using this gene as the active ingredient.
 サイトカインの一種であるIL-10は,Th2細胞で産生される抗炎症性のサイトカインであり,Th1細胞のサイトカイン産生を阻害することができ,免疫応答を抑制する働きを持つ。当該抗炎症作用により,IL-10は,多くの炎症性疾患,すなわち,神経傷害性疼痛,多発性硬化症,脊髄傷害,ALS,神経炎症,関節炎および関節の他の疾患に関連する症状,及び自己免疫疾患等に効果があることが期待されている。また,免疫抑制作用を有する一方で,IL-10は抗癌作用を有する可能性も報告されている(非特許文献3)。ヒトIL-10をコードする遺伝子は,1991年に単離されている(非特許文献4)。しかし,この遺伝子を用いて製造された組換えヒトIL-10(rhIL-10)を有効成分として含む,炎症性疾患又は癌の治療薬として用いるための医薬品はない。 IL-10, a type of cytokine, is an anti-inflammatory cytokine produced by Th2 cells and can inhibit cytokine production by Th1 cells, suppressing immune responses. Due to its anti-inflammatory effect, IL-10 is expected to be effective against many inflammatory diseases, namely, neuropathic pain, multiple sclerosis, spinal cord injury, ALS, neuroinflammation, symptoms associated with arthritis and other joint diseases, and autoimmune diseases. In addition to its immunosuppressive effect, it has also been reported that IL-10 may have an anti-cancer effect (Non-Patent Document 3). The gene encoding human IL-10 was isolated in 1991 (Non-Patent Document 4). However, there are no pharmaceuticals for use as a therapeutic agent for inflammatory diseases or cancer that contain recombinant human IL-10 (rhIL-10) produced using this gene as an active ingredient.
 神経栄養因子の一種であるBDNFは,標的細胞表面上にある特異的受容体TrkBに結合し,神経細胞の生存・成長・シナプスの機能亢進などの神経細胞の成長を調節する機能を持つ神経系の液性蛋白質である。当該神経発達作用により,BDNFは,アルツハイマー病,パーキンソン病,ハンチントン病などの神経変性疾患,筋萎縮性側策硬化症などの脊髄変性疾患,この他,糖尿病性神経障害,脳虚血性疾患,Rett症候群などの発達障害,統合失調症,うつ病およびRett症候群等,多様な疾患の治療剤としての開発が期待されている。ヒトBDNFをコードする遺伝子は,1993年に単離されている(非特許文献5,6)。しかし,この遺伝子を用いて製造された組換えヒトBDNF(rhBDNF)を有効成分として含む,神経変性疾患等の治療薬として用いるための医薬品はない。 BDNF, a type of neurotrophic factor, is a humoral protein of the nervous system that binds to a specific receptor TrkB on the surface of target cells and regulates the growth of nerve cells, such as the survival and growth of nerve cells and the enhancement of synaptic function. Due to its neurodevelopmental effect, BDNF is expected to be developed as a treatment for a variety of diseases, including neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, spinal degenerative diseases such as amyotrophic lateral sclerosis, as well as diabetic neuropathy, cerebral ischemic disease, developmental disorders such as Rett syndrome, schizophrenia, depression, and Rett syndrome. The gene encoding human BDNF was isolated in 1993 (Non-Patent Documents 5 and 6). However, there are no pharmaceuticals containing recombinant human BDNF (rhBDNF) produced using this gene as an active ingredient for use as a treatment for neurodegenerative diseases, etc.
 神経栄養因子の一種であるNGFは,末梢神経系において交感神経細胞および脊髄感覚ニューロンの生存及び成長を促進し,中枢神経系において,また特に前脳基底核において,コリン作動性神経細胞の生存及び分化を促進する機能を有する蛋白質である。特に樹状突起の機能低下を防ぐ作用により,NGFは,アルツハイマー病,パーキンソン病,ハンチントン病などの神経変性疾患のための治療剤としての開発が期待されているが,脳機能の老化・変性の予防,脳機能の改善,認知症の予防及び治療,神経ネットワークの構築による記憶学習能の向上,及び神経伝達物質の作用増強等にも効果を有することが期待される。ヒトNGFを構成するα,β,及びγの3つのサブユニットのうち少なくともβサブユニットをコードする遺伝子は,1990年に単離されている(非特許文献7)。しかし,ヒトNGFをコードする遺伝子を用いて製造された組換えヒトNGF(rhNGF)を有効成分として含む,神経変性疾患等の治療薬として用いるための医薬品はない。 NGF, a type of neurotrophic factor, is a protein that promotes the survival and growth of sympathetic nerve cells and spinal sensory neurons in the peripheral nervous system, and promotes the survival and differentiation of cholinergic nerve cells in the central nervous system, especially in the basal forebrain. In particular, due to its effect of preventing the decline of dendrite function, NGF is expected to be developed as a treatment for neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, but it is also expected to be effective in preventing aging and degeneration of brain function, improving brain function, preventing and treating dementia, improving memory and learning ability by building neural networks, and enhancing the action of neurotransmitters. Of the three subunits α, β, and γ that make up human NGF, a gene encoding at least the β subunit was isolated in 1990 (Non-Patent Document 7). However, there are no pharmaceuticals for use as a treatment for neurodegenerative diseases, etc., that contain recombinant human NGF (rhNGF) as an active ingredient, which is manufactured using a gene encoding human NGF.
 神経栄養因子の一種であるNT-3は,Trkレセプター,特にTrkCを通したシグナル伝達を行い,神経細胞及びグリア細胞の生存と成長及び神経新生の促進に関与することが知られており,神経伝達の促進や神経の修復,特に視細胞の分化や再生を促進する作用が注目されている。従ってNT-3は,神経変性疾患のための治療剤としての開発が期待されている。ヒトNT-3をコードする遺伝子は,1991年に単離されている(非特許文献8)。しかし,ヒトNT-3をコードする遺伝子を用いて製造された組換えヒトNT-3(rhNT-3)を有効成分として含む,神経変性疾患等の治療薬として用いるための医薬品はない。 NT-3, a type of neurotrophic factor, is known to be involved in promoting the survival and growth of nerve cells and glial cells and neurogenesis by transmitting signals through Trk receptors, particularly TrkC, and is attracting attention for its ability to promote neurotransmission and nerve repair, particularly the differentiation and regeneration of photoreceptor cells. NT-3 is therefore expected to be developed as a treatment for neurodegenerative diseases. The gene encoding human NT-3 was isolated in 1991 (Non-patent Document 8). However, there are no pharmaceuticals for use as a treatment for neurodegenerative diseases that contain recombinant human NT-3 (rhNT-3) as an active ingredient, which is produced using the gene encoding human NT-3.
 神経栄養因子の一種であるNT-4は,Trkレセプター,特にTrkBを通したシグナル伝達を行い, NT-3と同様に抹消神経系ニューロンや中枢神経系ニューロンの成長と生存を促進する。従ってNT-4は,神経変性疾患のための治療剤としての開発が期待されている。ヒトNT-4をコードする遺伝子は,1992年に単離されている(非特許文献9)。しかし,ヒトNT-4をコードする遺伝子を用いて製造された組換えヒトNT-4(rhNT-4)を有効成分として含む,神経変性疾患等の治療薬として用いるための医薬品はない。 NT-4, a type of neurotrophic factor, transmits signals through Trk receptors, particularly TrkB, and promotes the growth and survival of peripheral and central nervous system neurons, similar to NT-3. NT-4 is therefore expected to be developed as a treatment for neurodegenerative diseases. The gene encoding human NT-4 was isolated in 1992 (Non-patent Document 9). However, there are no pharmaceuticals for use as a treatment for neurodegenerative diseases that contain recombinant human NT-4 (rhNT-4) as an active ingredient, which is produced using the gene encoding human NT-4.
 生体内に投与したときに,投与後に速やかに分解されて活性を消失する成長ホルモンを,血清アルブミンとの融合蛋白質として製造する方法が知られている(非特許文献10)。血清アルブミンと融合させた成長ホルモンは,生体内での安定性が増加する。従って,通常,成長ホルモンは,皮下に毎日投与されるべきところ,血清アルブミンとの融合蛋白質とすることで,その投与回数を減ずることができる。 A method is known for producing growth hormone, which is rapidly degraded and loses its activity when administered to the body, as a fusion protein with serum albumin (Non-Patent Document 10). Growth hormone fused to serum albumin has increased stability in the body. Therefore, although growth hormone is normally administered subcutaneously every day, by making it into a fusion protein with serum albumin, the number of times it is administered can be reduced.
 本発明の目的の一つは,通常ではCHO細胞等の宿主細胞を用いて組換え蛋白質として発現させたときに,発現量が低く,及び/又は,活性が低い生理活性物質を,血清アルブミン(SA)との融合蛋白質の形態で提供することである。かかる融合蛋白質は,活性の高い組換え蛋白質として効率よく製造できる。また,当該融合蛋白質の製造方法も提供される。
 本発明の他の目的の一つは,通常ではCHO細胞等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低いリソソーム酵素を,血清アルブミンとの融合蛋白質の形態で提供することである。かかる融合蛋白質は,活性の高い組換え蛋白質として効率よく製造できる。また,当該融合蛋白質の製造方法も提供される。ここで,リソソーム酵素としては,例えば,hGALC,hGBAが挙げられる。
One of the objects of the present invention is to provide a physiologically active substance that is usually expressed in a low amount and/or has a low activity when expressed as a recombinant protein using a host cell such as a CHO cell, in the form of a fusion protein with serum albumin (SA). Such a fusion protein can be efficiently produced as a highly active recombinant protein. Also provided is a method for producing the fusion protein.
Another object of the present invention is to provide a lysosomal enzyme that is expressed in a low amount and/or has low activity when expressed as a recombinant protein using a host cell such as a CHO cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in the culture medium, in the form of a fusion protein with serum albumin. Such a fusion protein can be efficiently produced as a highly active recombinant protein. A method for producing the fusion protein is also provided. Examples of the lysosomal enzyme include hGALC and hGBA.
 なお,組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させるときには,組換え蛋白質をコードする遺伝子の5’側に,リーダーペプチドをコードするDNA配列がインフレームで配置される。これにより,発現した組換え蛋白質が細胞から分泌されるようになる。かかるリーダーペプチドは,組換え蛋白質のN末端に,SAが位置する場合はSAのリーダーペプチド,リソソーム酵素が位置する場合はリソソーム酵素のリーダーペプチド,サイトカインが位置する場合はサイトカインのリーダーペプチド,神経栄養因子が位置する場合は神経栄養因子のリーダーペプチドであることが好ましい。但し,これらに替えて,リーダーペプチドを,成長ホルモンのリーダーペプチド等の異種蛋白質のリーダーペプチド,又は人工的なリーダーペプチドとすることもできる。 When expressing a recombinant protein so that it is secreted from cells and accumulated in the culture medium, a DNA sequence encoding a leader peptide is placed in frame on the 5' side of the gene encoding the recombinant protein. This allows the expressed recombinant protein to be secreted from cells. The leader peptide is preferably an SA leader peptide when an SA is located at the N-terminus of the recombinant protein, a lysosomal enzyme leader peptide when a lysosomal enzyme is located at the N-terminus of the recombinant protein, a cytokine leader peptide when a cytokine is located at the N-terminus of the recombinant protein, or a neurotrophic factor leader peptide when a neurotrophic factor is located at the N-terminus of the recombinant protein. However, instead of these, the leader peptide may be a leader peptide of a heterologous protein, such as a growth hormone leader peptide, or an artificial leader peptide.
 本発明の他の目的の一つは,通常ではCHO細胞等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低いサイトカインを,血清アルブミンとの融合蛋白質の形態で提供することである。かかる融合蛋白質は,活性の高い組換え蛋白質として効率よく製造できる。また,当該融合蛋白質の製造方法も提供される。ここで,サイトカインとしては,例えば,インターロイキンであり,特にhIL-10である。
 本発明の他の目的の一つは,通常ではCHO細胞等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量が低く,及び/又は,活性が低い神経栄養因子を,血清アルブミンとの融合蛋白質の形態で提供することである。かかる融合蛋白質は,活性の高い組換え蛋白質として効率よく製造できる。また,当該融合蛋白質の製造方法も提供される。ここで,神経栄養因子としては,例えば,hBDNF,hNGF,hNT-3,及びhNT-4が挙げられる。
Another object of the present invention is to provide a cytokine that is expressed in a low amount and/or has low activity when expressed as a recombinant protein using a host cell such as a CHO cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in a culture medium, in the form of a fusion protein with serum albumin. Such a fusion protein can be efficiently produced as a highly active recombinant protein. A method for producing the fusion protein is also provided. Here, the cytokine is, for example, an interleukin, particularly hIL-10.
Another object of the present invention is to provide a neurotrophic factor that is expressed in a low amount and/or has low activity when expressed as a recombinant protein using a host cell such as a CHO cell, particularly when expressed so that the recombinant protein is secreted from the cell and accumulates in the culture medium, in the form of a fusion protein with serum albumin. Such a fusion protein can be efficiently produced as a highly active recombinant protein. A method for producing the fusion protein is also provided. Examples of neurotrophic factors include hBDNF, hNGF, hNT-3, and hNT-4.
 上記目的に向けた研究において,本発明者らは,鋭意検討を重ねた結果,本明細書において詳述する,ヒトリソソーム酵素であるhGALC又はhGBAのN末端又はC末端に直接又はリンカーを介してHSAが結合した融合蛋白質を,当該融合蛋白質をコードする遺伝子を組み込んだ発現ベクターを導入した宿主細胞を培養して発現させたときの組換え融合蛋白質の発現量(当該組換え融合蛋白質の野生型ヒトリソソーム酵素に対応する部分の発現量に換算)が,野生型ヒトリソソーム酵素をコードする遺伝子を組み込んだ発現ベクターを導入した宿主細胞を培養して発現させたときの組換え野生型ヒトリソソーム酵素の発現量と比較して,格段に増加することを見出し,本発明を完成した。
 また上記目的に向けた研究において,本発明者らは,鋭意検討を重ねた結果,本明細書において詳述する,ヒトサイトカインであるhIL-10のN末端又はC末端に直接又はリンカーを介してHSAが結合した融合蛋白質を,当該融合蛋白質をコードする遺伝子を組み込んだ発現ベクターを導入した宿主細胞を培養して発現させたときの組換え融合蛋白質の活性が,野生型hIL-10をコードする遺伝子を組み込んだ発現ベクターを導入した宿主細胞を培養して発現させたときの組換え野生型hIL-10の活性と比較して,格段に増加することを見出し,本発明を完成した。
 また上記目的に向けた研究において,本発明者らは,鋭意検討を重ねた結果,本明細書において詳述する,ヒト神経栄養因子であるhBDNF,hNGF,hNT-3,又はhNT-4のN末端又はC末端に直接又はリンカーを介してHSAが結合した融合蛋白質を,当該融合蛋白質をコードする遺伝子を組み込んだ発現ベクターを導入した宿主細胞を培養して発現させたときの組換え融合蛋白質の活性が,野生型ヒト神経栄養因子をコードする遺伝子を組み込んだ発現ベクターを導入した宿主細胞を培養して発現させたときの組換え野生型ヒト神経栄養因子の活性と比較して,格段に増加することを見出し,本発明を完成した。
 すなわち,本発明は以下を含むものである。
1.リソソーム酵素と,血清アルブミン(SA)を含んでなる,融合蛋白質。
2.該リソソーム酵素が,ヒトリソソーム酵素である,上記1に記載の融合蛋白質。
3.該SAが,ヒト血清アルブミン(HSA)である,上記1又は2に記載の融合蛋白質。
4.該リソソーム酵素が,配列番号1で示されるアミノ酸配列を有する野生型ヒトガラクトシルセラミダーゼに対して80%以上の同一性を有するヒトガラクトシルセラミダーゼ(hGALC)であって,該SAが,配列番号3で示されるアミノ酸配列を有する野生型ヒト血清アルブミンに対して80%以上の同一性を有するヒト血清アルブミン(HSA)である,上記1乃至3の何れかに記載の融合蛋白質。
5.該hGALCが配列番号1で示されるアミノ酸配列を有する野生型hGALCに対して90%以上の同一性を有するものであり,該HSAが配列番号3で示されるアミノ酸配列を有する野生型HSAに対して90%以上の同一性を有するものである,上記4に記載の融合蛋白質。
6.該hGALCが,配列番号1で示される野生型hGALCのアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記4に記載の融合蛋白質。
7.該hGALCが,配列番号1で示される野生型hGALCのアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記4に記載の融合蛋白質。
8.該hGALCが,配列番号1で示される野生型hGALCのアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記4に記載の融合蛋白質。
9.該hGALCが,配列番号1で示される野生型hGALCのアミノ酸配列に対して1個のアミノ酸が置換されたアミノ酸配列を含んでなるものである,上記4に記載の融合蛋白質。
10.該アミノ酸の置換が,側鎖が水酸化反応され得るアミノ酸であるアミノ酸ファミリー内での置換である,上記9に記載の融合蛋白質。
11.該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記4乃至10の何れかに記載の融合蛋白質。
12.該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記4乃至10の何れかに記載の融合蛋白質。
13.該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記4乃至10の何れかに記載の融合蛋白質。
14.該hGALCが配列番号1で示される野生型hGALCのアミノ酸配列を含んでなるものであり,該HSAが配列番号3で示される野生型ヒト血清アルブミンのアミノ酸配列を含んでなるものである,上記4に記載の融合蛋白質。
15.該hGALCが配列番号1で示される野生型hGALCのアミノ酸配列を含んでなるものであり,該HSAが配列番号12で示される野生型ヒト血清アルブミンのアミノ酸配列を含んでなるものである,上記4に記載の融合蛋白質。
16.該hGALCが配列番号1で示される野生型hGALCのアミノ酸配列を含んでなるものであり,該HSAが配列番号13で示される野生型ヒト血清アルブミンのアミノ酸配列を含んでなるものである,上記4に記載の融合蛋白質。
17.該hGALCのC末端に,直接又はリンカーを介して,該HSAが結合したものである,上記4乃至16の何れかに記載の融合蛋白質。
18.該HSAのC末端に,直接又はリンカーを介して,該hGALCが結合したものである,上記4乃至16の何れかに記載の融合蛋白質。
19.該リンカーが,1~150個のアミノ酸からなるペプチド鎖である,上記17又は18に記載の融合蛋白質。
20.該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列からなるものである,上記19に記載の融合蛋白質:
 (a)Gly;
 (b)Ser;
 (c)Gly Ser;
 (d)Gly Gly Ser;
 (e)配列番号9で示されるアミノ酸配列;
 (f)配列番号10で示されるアミノ酸配列;及び,
 (g)配列番号11で示されるアミノ酸配列。
21.該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が2~10回反復したアミノ酸配列からなるものである,上記19に記載の融合蛋白質:
 (a)Gly;
 (b)Ser;
 (c)Gly Ser;
 (d)Gly Gly Ser;
 (e)配列番号9で示されるアミノ酸配列;
 (f)配列番号10で示されるアミノ酸配列;及び,
 (g)配列番号11で示されるアミノ酸配列。
22.該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が2~6回反復したアミノ酸配列からなるものである,上記19に記載の融合蛋白質:
 (a)Gly;
 (b)Ser;
 (c)Gly Ser;
 (d)Gly Gly Ser;
 (e)配列番号9で示されるアミノ酸配列;
 (f)配列番号10で示されるアミノ酸配列;及び,
 (g)配列番号11で示されるアミノ酸配列。
23.該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が3~5回反復したアミノ酸配列からなるものである,上記19に記載の融合蛋白質:
 (a)Gly;
 (b)Ser;
 (c)Gly Ser;
 (d)Gly Gly Ser;
 (e)配列番号9で示されるアミノ酸配列;
 (f)配列番号10で示されるアミノ酸配列;及び,
 (g)配列番号11で示されるアミノ酸配列。
24.該リンカーが,Gly Serで示されるアミノ酸配列からなるものである,上記19に記載の融合蛋白質。
25.配列番号5で示されるアミノ酸配列に対して80%以上の同一性を有するアミノ酸配列を含んでなる,上記18に記載の融合蛋白質。
26.配列番号5で示されるアミノ酸配列に対して90%以上の同一性を有するアミノ酸配列を含んでなる,上記18に記載の融合蛋白質。
27.該融合蛋白質が,配列番号5で示されるアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記26に記載の融合蛋白質。
28.該融合蛋白質が,配列番号5で示されるアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記26に記載の融合蛋白質。
29.該融合蛋白質が,配列番号5で示されるアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記26に記載の融合蛋白質。
30.配列番号5で示されるアミノ酸配列を含んでなる,上記18に記載の融合蛋白質。
31.配列番号7で示されるアミノ酸配列に対して80%以上の同一性を有するアミノ酸配列を含んでなる,上記17に記載の融合蛋白質。
32.配列番号7で示されるアミノ酸配列に対して90%以上の同一性を有するアミノ酸配列を含んでなる,上記17に記載の融合蛋白質。
33.該融合蛋白質が,配列番号7で示されるアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記32に記載の融合蛋白質。
34.該融合蛋白質が,配列番号7で示されるアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記32に記載の融合蛋白質。
35.該融合蛋白質が,配列番号7で示されるアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記32に記載の融合蛋白質。
36.配列番号7で示されるアミノ酸配列を含んでなる,上記17に記載の融合蛋白質。
37.通常の野生型のhGALCの比活性と比較して,10%以上の比活性を有するものである,上記4乃至36の何れかに記載の融合蛋白質。
38.該リソソーム酵素が,配列番号37で示されるアミノ酸配列を有する野生型ヒトグルコセレブロシダーゼに対して80%以上の同一性を有するヒトグルコセレブロシダーゼ(hGBA)であって,該SAが,配列番号3で示されるアミノ酸配列を有する野生型ヒト血清アルブミンに対して80%以上の同一性を有するヒト血清アルブミン(HSA)である,上記1乃至3の何れかに記載の融合蛋白質。
39.該hGBAが配列番号37で示されるアミノ酸配列を有する野生型hGBAに対して90%以上の同一性を有するものであり,該HSAが配列番号3で示されるアミノ酸配列を有する野生型HSAに対して90%以上の同一性を有するものである,上記38に記載の融合蛋白質。
40.該hGBAが,配列番号37で示される野生型hGBAのアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記38に記載の融合蛋白質。
41.該hGBAが,配列番号37で示される野生型hGBAのアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記38に記載の融合蛋白質。
42.該hGBAが,配列番号37で示される野生型hGBAのアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記38に記載の融合蛋白質。
43.該hGBAが,配列番号37で示される野生型hGBAのアミノ酸配列に対して1個のアミノ酸が置換されたアミノ酸配列を含んでなるものである,上記38に記載の融合蛋白質。
44.該アミノ酸の置換が,側鎖が水酸化反応され得るアミノ酸であるアミノ酸ファミリー内での置換である,上記43に記載の融合蛋白質。
45.該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記38乃至44の何れかに記載の融合蛋白質。
46.該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記38乃至44の何れかに記載の融合蛋白質。
47.該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記38乃至44の何れかに記載の融合蛋白質。
48.該hGBAが配列番号37で示される野生型hGBAのアミノ酸配列を含んでなるものであり,該HSAが配列番号3で示される野生型HSAのアミノ酸配列を含んでなるものである,上記38に記載の融合蛋白質。
49.該hGBAが配列番号37で示される野生型hGBAのアミノ酸配列を含んでなるものであり,該HSAが配列番号12で示される野生型HSAのアミノ酸配列を含んでなるものである,上記38に記載の融合蛋白質。
50.該hGBAが配列番号37で示される野生型hGBAのアミノ酸配列を含んでなるものであり,該HSAが配列番号13で示される野生型HSAのアミノ酸配列を含んでなるものである,上記38に記載の融合蛋白質。
51.該hGBAのC末端に,直接又はリンカーを介して,該HSAが結合したものである,上記38乃至50の何れかに記載の融合蛋白質。
52.該HSAのC末端に,直接又はリンカーを介して,該hGBAが結合したものである,上記38乃至50の何れかに記載の融合蛋白質。
53.該リンカーが,1~150個のアミノ酸からなるペプチド鎖である,上記51又は52に記載の融合蛋白質。
54.該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列からなるものである,上記53に記載の融合蛋白質:
 (a)Gly;
 (b)Ser;
 (c)Gly Ser;
 (d)Gly Gly Ser;
 (e)配列番号9で示されるアミノ酸配列;
 (f)配列番号10で示されるアミノ酸配列;及び,
 (g)配列番号11で示されるアミノ酸配列。
55.該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が2~10回反復したアミノ酸配列からなるものである,上記53に記載の融合蛋白質:
 (a)Gly;
 (b)Ser;
 (c)Gly Ser;
 (d)Gly Gly Ser;
 (e)配列番号9で示されるアミノ酸配列;
 (f)配列番号10で示されるアミノ酸配列;及び,
 (g)配列番号11で示されるアミノ酸配列。
56.該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が2~6回反復したアミノ酸配列からなるものである,上記53に記載の融合蛋白質:
 (a)Gly;
 (b)Ser;
 (c)Gly Ser;
 (d)Gly Gly Ser;
 (e)配列番号9で示されるアミノ酸配列;
 (f)配列番号10で示されるアミノ酸配列;及び,
 (g)配列番号11で示されるアミノ酸配列。
57.該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が3~5回反復したアミノ酸配列からなるものである,上記53に記載の融合蛋白質:
 (a)Gly;
 (b)Ser;
 (c)Gly Ser;
 (d)Gly Gly Ser;
 (e)配列番号9で示されるアミノ酸配列;
 (f)配列番号10で示されるアミノ酸配列;及び,
 (g)配列番号11で示されるアミノ酸配列。
58.該リンカーが,Gly Serで示されるアミノ酸配列からなるものである,上記53に記載の融合蛋白質。
59.配列番号39で示されるアミノ酸配列に対して80%以上の同一性を有するアミノ酸配列を含んでなる,上記52に記載の融合蛋白質。
60.配列番号39で示されるアミノ酸配列に対して90%以上の同一性を有するアミノ酸配列を含んでなる,上記52に記載の融合蛋白質。
61.該融合蛋白質が,配列番号39で示されるアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記60に記載の融合蛋白質。
62.該融合蛋白質が,配列番号39で示されるアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記60に記載の融合蛋白質。
63.該融合蛋白質が,配列番号39で示されるアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記60に記載の融合蛋白質。
64.配列番号41で示されるアミノ酸配列を含んでなる,上記51に記載の融合蛋白質。
65.配列番号41で示されるアミノ酸配列に対して80%以上の同一性を有するアミノ酸配列を含んでなる,上記51に記載の融合蛋白質。
66.配列番号41で示されるアミノ酸配列に対して90%以上の同一性を有するアミノ酸配列を含んでなる,上記51に記載の融合蛋白質。
67.該融合蛋白質が,配列番号41で示されるアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記66に記載の融合蛋白質。
68.該融合蛋白質が,配列番号41で示されるアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記66に記載の融合蛋白質。
69.該融合蛋白質が,配列番号41で示されるアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,上記66に記載の融合蛋白質。
70.配列番号41で示されるアミノ酸配列を含んでなる,上記51に記載の融合蛋白質。
71.通常の野生型のhGBAの比活性と比較して,10%以上の比活性を有するものである,上記38乃至70の何れかに記載の融合蛋白質。
72,上記1乃至71の何れかに記載の融合蛋白質をコードする遺伝子を含んでなるDNA。
73.上記72に記載のDNAを含んでなる発現ベクター。
74.上記73に記載の発現ベクターで形質転換された哺乳動物細胞。
75.上記74に記載の哺乳動物細胞を無血清培地で培養するステップを含む,融合蛋白質の製造方法。
76.上記1乃至170の何れかに記載の融合蛋白質と,抗体との結合体。
77.リソソーム酵素,血清アルブミン及び抗体との結合体。
78.以下の(1)~(6)から選択されるものである,上記77の結合体:
(1)該リソソーム酵素のC末端に,直接又はリンカーを介して,血清アルブミンが結合し,更にそのC末端に,直接又はリンカーを介して,該抗体が結合したものである結合体;
(2)該リソソーム酵素のC末端に,直接又はリンカーを介して,抗体が結合し,更にそのC末端に,直接又はリンカーを介して,該血清アルブミンが結合したものである結合体;
(3)該血清アルブミンのC末端に,直接又はリンカーを介して,該リソソーム酵素が結合し,更にそのC末端に,直接又はリンカーを介して,該抗体が結合したものである結合体;
(4)該血清アルブミンのC末端に,直接又はリンカーを介して,該抗体が結合し,更にそのC末端に,直接又はリンカーを介して,該リソソーム酵素が結合したものである結合体;
(5)該抗体のC末端に,直接又はリンカーを介して,該リソソーム酵素が結合し,更にそのC末端に,直接又はリンカーを介して,該血清アルブミンが結合したものである結合体;
(6)該抗体のC末端に,直接又はリンカーを介して,該血清アルブミンが結合し,更にそのC末端に,直接又はリンカーを介して,該リソソーム酵素が結合したものである結合体。
79.該抗体が,血管内皮細胞上に発現する受容体に対する抗体である,上記76乃至78の結合体。
80.該血管内皮細胞上の受容体が,インスリン受容体,トランスフェリン受容体,レプチン受容体,リポタンパク質受容体,及びIGF受容体からなる群から選択されるものである,上記79に記載の結合体。
81.該血管内皮細胞上の受容体が,トランスフェリン受容体である,上記79に記載の結合体。
82.該抗体が,Fab抗体,F(ab’)抗体,F(ab’)抗体,単一ドメイン抗体,一本鎖抗体,VHH,又はFc抗体の何れかである,上記76乃至81の何れかに記載の結合体。
83.該融合蛋白質が,該抗体の軽鎖のC末端側又はN末端側の何れかに結合しているものである,上記76乃至82の何れかに記載の結合体。
84.該融合蛋白質が,該抗体の重鎖のC末端側又はN末端側の何れかに結合しているものである,上記76乃至82の何れかに記載の結合体。
85.該融合蛋白質が,該抗体の軽鎖のC末端側若しくはN末端側の何れか,又は重鎖のC末端側若しくはN末端側の何れかに,リンカー配列を介して結合しているものである,上記76乃至82の何れかに記載の結合体。
86.該リンカー配列が,1~50個のアミノ酸残基からなるものである,上記78乃至85に記載の結合体。
87.該リンカー配列が,1個のグリシン,1個のセリン,アミノ酸配列Gly-Ser,アミノ酸配列Ser-Ser,アミノ酸配列Gly-Gly-Ser,配列番号9のアミノ酸配列,配列番号10のアミノ酸配列,配列番号11のアミノ酸配列,及びこれらのアミノ酸配列が1~10個連続してなるアミノ酸配列からなる群より選ばれるアミノ酸配列を含んでなるものである,上記86に記載の結合体。
88.上記76乃至87の何れかに記載の結合体をコードする遺伝子を含んでなるDNA。
89.上記88に記載のDNAを含んでなる発現ベクター。
90.上記89に記載の発現ベクターで形質転換された哺乳動物細胞。
91.上記90に記載の哺乳動物細胞を無血清培地で培養するステップを含む,生理活性を有する蛋白質とSAとの融合蛋白質と抗体との結合体の製造方法。
In research aimed at the above-mentioned object, the inventors conducted extensive investigations and found that, as a result of the discovery, when a fusion protein in which HSA is bound directly or via a linker to the N-terminus or C-terminus of human lysosomal enzymes hGALC or hGBA, as described in detail in this specification, is expressed by culturing a host cell into which an expression vector incorporating a gene encoding the fusion protein has been introduced, the expression level of the recombinant fusion protein (converted to the expression level of the portion of the recombinant fusion protein corresponding to the wild-type human lysosomal enzyme) is significantly increased compared to the expression level of the recombinant wild-type human lysosomal enzyme when the recombinant fusion protein is expressed by culturing a host cell into which an expression vector incorporating a gene encoding the wild-type human lysosomal enzyme has been introduced, thereby completing the present invention.
Furthermore, in the course of research aimed at the above-mentioned object, the present inventors conducted extensive investigations and found that, as a result of culturing and expressing a fusion protein in which HSA is bound directly or via a linker to the N-terminus or C-terminus of the human cytokine hIL-10, as described in detail herein, in a host cell into which an expression vector incorporating a gene encoding the fusion protein has been introduced, the activity of the recombinant fusion protein is significantly increased compared to the activity of recombinant wild-type hIL-10 in which the recombinant fusion protein is expressed in a host cell into which an expression vector incorporating a gene encoding wild-type hIL-10 has been introduced, thereby completing the present invention.
Furthermore, in research aimed at the above-mentioned object, the inventors have conducted extensive investigations and as a result have found that when a fusion protein in which HSA is bound directly or via a linker to the N-terminus or C-terminus of human neurotrophic factors hBDNF, hNGF, hNT-3, or hNT-4, as described in detail in this specification, is expressed by culturing a host cell into which an expression vector incorporating a gene encoding the fusion protein has been introduced, the activity of the recombinant fusion protein is significantly increased compared to the activity of recombinant wild-type human neurotrophic factor when expressed by culturing a host cell into which an expression vector incorporating a gene encoding a wild-type human neurotrophic factor has been introduced, thereby completing the present invention.
That is, the present invention includes the following.
1. A fusion protein comprising a lysosomal enzyme and serum albumin (SA).
2. The fusion protein according to claim 1, wherein the lysosomal enzyme is a human lysosomal enzyme.
3. The fusion protein according to 1 or 2 above, wherein the SA is human serum albumin (HSA).
4. The fusion protein according to any one of 1 to 3 above, wherein the lysosomal enzyme is human galactosylceramidase (hGALC) having 80% or more identity to wild-type human galactosylceramidase having the amino acid sequence shown in SEQ ID NO: 1, and the SA is human serum albumin (HSA) having 80% or more identity to wild-type human serum albumin having the amino acid sequence shown in SEQ ID NO: 3.
5. The fusion protein according to 4 above, wherein the hGALC has 90% or more identity to wild-type hGALC having the amino acid sequence shown in SEQ ID NO:1, and the HSA has 90% or more identity to wild-type HSA having the amino acid sequence shown in SEQ ID NO:3.
6. The fusion protein according to 4 above, wherein the hGALC comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1.
7. The fusion protein according to 4 above, wherein the hGALC comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1.
8. The fusion protein according to 4 above, wherein the hGALC comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added relative to the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1.
9. The fusion protein according to 4 above, wherein the hGALC comprises an amino acid sequence in which one amino acid has been substituted with respect to the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1.
10. The fusion protein according to 9 above, wherein the amino acid substitution is within an amino acid family in which the side chain is an amino acid that can be hydroxylated.
11. The fusion protein according to any one of 4 to 10 above, wherein the HSA comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
12. The fusion protein according to any one of 4 to 10 above, wherein the HSA comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
13. The fusion protein according to any one of 4 to 10 above, wherein the HSA comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
14. The fusion protein according to 4 above, wherein the hGALC comprises the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1, and the HSA comprises the amino acid sequence of wild-type human serum albumin shown in SEQ ID NO:3.
15. The fusion protein according to 4 above, wherein the hGALC comprises the amino acid sequence of wild-type hGALC shown in SEQ ID NO: 1, and the HSA comprises the amino acid sequence of wild-type human serum albumin shown in SEQ ID NO: 12.
16. The fusion protein according to 4 above, wherein the hGALC comprises the amino acid sequence of wild-type hGALC shown in SEQ ID NO: 1, and the HSA comprises the amino acid sequence of wild-type human serum albumin shown in SEQ ID NO: 13.
17. The fusion protein according to any one of 4 to 16 above, wherein the HSA is bound to the C-terminus of the hGALC directly or via a linker.
18. The fusion protein according to any one of 4 to 16 above, wherein the hGALC is bound to the C-terminus of the HSA directly or via a linker.
19. The fusion protein according to 17 or 18 above, wherein the linker is a peptide chain consisting of 1 to 150 amino acids.
20. The fusion protein according to claim 19, wherein the linker comprises an amino acid sequence selected from the group consisting of the following (a) to (g):
(a) Gly;
(b) Ser;
(c) GlySer;
(d) GlyGlySer;
(e) the amino acid sequence set forth in SEQ ID NO:9;
(f) the amino acid sequence set forth in SEQ ID NO: 10; and
(g) The amino acid sequence shown in SEQ ID NO:11.
21. The fusion protein according to claim 19, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 10 times:
(a) Gly;
(b) Ser;
(c) GlySer;
(d) GlyGlySer;
(e) the amino acid sequence set forth in SEQ ID NO:9;
(f) the amino acid sequence set forth in SEQ ID NO: 10; and
(g) The amino acid sequence shown in SEQ ID NO:11.
22. The fusion protein according to 19 above, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 6 times:
(a) Gly;
(b) Ser;
(c) GlySer;
(d) GlyGlySer;
(e) the amino acid sequence set forth in SEQ ID NO:9;
(f) the amino acid sequence set forth in SEQ ID NO: 10; and
(g) The amino acid sequence shown in SEQ ID NO:11.
23. The fusion protein according to claim 19, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 3 to 5 times:
(a) Gly;
(b) Ser;
(c) GlySer;
(d) GlyGlySer;
(e) the amino acid sequence set forth in SEQ ID NO:9;
(f) the amino acid sequence set forth in SEQ ID NO: 10; and
(g) The amino acid sequence shown in SEQ ID NO:11.
24. The fusion protein according to claim 19, wherein the linker comprises the amino acid sequence Gly Ser.
25. The fusion protein according to 18 above, which comprises an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:5.
26. The fusion protein according to 18 above, which comprises an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:5.
27. The fusion protein according to claim 26, which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:5.
28. The fusion protein according to claim 26, which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:5.
29. The fusion protein according to claim 26, which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added relative to the amino acid sequence shown in SEQ ID NO:5.
30. The fusion protein according to claim 18, which comprises the amino acid sequence shown in SEQ ID NO:5.
31. The fusion protein according to 17 above, which comprises an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:7.
32. The fusion protein according to 17 above, which comprises an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:7.
33. The fusion protein according to 32 above, which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:7.
34. The fusion protein according to 32 above, which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:7.
35. The fusion protein according to 32 above, which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:7.
36. The fusion protein according to claim 17, comprising the amino acid sequence shown in SEQ ID NO:7.
37. The fusion protein according to any one of 4 to 36 above, which has a specific activity of 10% or more compared with the specific activity of normal wild-type hGALC.
38. The fusion protein according to any one of 1 to 3 above, wherein the lysosomal enzyme is human glucocerebrosidase (hGBA) having 80% or more identity to wild-type human glucocerebrosidase having the amino acid sequence shown in SEQ ID NO: 37, and the SA is human serum albumin (HSA) having 80% or more identity to wild-type human serum albumin having the amino acid sequence shown in SEQ ID NO: 3.
39. The fusion protein according to claim 38, wherein said hGBA has 90% or more identity to wild-type hGBA having the amino acid sequence shown in SEQ ID NO:37, and said HSA has 90% or more identity to wild-type HSA having the amino acid sequence shown in SEQ ID NO:3.
40. The fusion protein according to 38 above, wherein the hGBA comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added relative to the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37.
41. The fusion protein according to 38 above, wherein the hGBA comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added relative to the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37.
42. The fusion protein according to 38 above, wherein the hGBA comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added relative to the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37.
43. The fusion protein according to 38 above, wherein said hGBA comprises an amino acid sequence in which one amino acid has been substituted with respect to the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37.
44. The fusion protein according to 43 above, wherein the amino acid substitution is within an amino acid family in which the side chain is an amino acid that can be hydroxylated.
45. The fusion protein according to any one of 38 to 44 above, wherein the HSA comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
46. The fusion protein according to any one of 38 to 44 above, wherein the HSA comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
47. The fusion protein according to any one of 38 to 44 above, wherein the HSA comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
48. The fusion protein according to claim 38, wherein said hGBA comprises the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, and said HSA comprises the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
49. The fusion protein according to claim 38, wherein said hGBA comprises the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, and said HSA comprises the amino acid sequence of wild-type HSA shown in SEQ ID NO:12.
50. The fusion protein according to claim 38, wherein said hGBA comprises the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, and said HSA comprises the amino acid sequence of wild-type HSA shown in SEQ ID NO:13.
51. The fusion protein according to any one of 38 to 50 above, wherein the HSA is bound to the C-terminus of the hGBA directly or via a linker.
52. The fusion protein according to any one of 38 to 50 above, wherein the hGBA is bound to the C-terminus of the HSA directly or via a linker.
53. The fusion protein according to 51 or 52 above, wherein the linker is a peptide chain consisting of 1 to 150 amino acids.
54. The fusion protein according to 53 above, wherein the linker comprises an amino acid sequence selected from the group consisting of the following (a) to (g):
(a) Gly;
(b) Ser;
(c) GlySer;
(d) GlyGlySer;
(e) the amino acid sequence set forth in SEQ ID NO:9;
(f) the amino acid sequence set forth in SEQ ID NO: 10; and
(g) The amino acid sequence shown in SEQ ID NO:11.
55. The fusion protein according to 53 above, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 10 times:
(a) Gly;
(b) Ser;
(c) GlySer;
(d) GlyGlySer;
(e) the amino acid sequence set forth in SEQ ID NO:9;
(f) the amino acid sequence set forth in SEQ ID NO: 10; and
(g) The amino acid sequence shown in SEQ ID NO:11.
56. The fusion protein according to 53 above, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 6 times:
(a) Gly;
(b) Ser;
(c) GlySer;
(d) GlyGlySer;
(e) the amino acid sequence set forth in SEQ ID NO:9;
(f) the amino acid sequence set forth in SEQ ID NO: 10; and
(g) The amino acid sequence shown in SEQ ID NO:11.
57. The fusion protein according to 53 above, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 3 to 5 times:
(a) Gly;
(b) Ser;
(c) GlySer;
(d) GlyGlySer;
(e) the amino acid sequence set forth in SEQ ID NO:9;
(f) the amino acid sequence set forth in SEQ ID NO: 10; and
(g) The amino acid sequence shown in SEQ ID NO:11.
58. The fusion protein according to the above 53, wherein the linker consists of the amino acid sequence represented by Gly Ser.
59. The fusion protein according to 52 above, comprising an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:39.
60. The fusion protein according to 52 above, which comprises an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:39.
61. The fusion protein according to 60 above, which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:39.
62. The fusion protein according to 60 above, which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:39.
63. The fusion protein according to 60 above, which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:39.
64. The fusion protein according to claim 51, comprising the amino acid sequence shown in SEQ ID NO:41.
65. The fusion protein according to claim 51, comprising an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:41.
66. The fusion protein according to the above item 51, comprising an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:41.
67. The fusion protein according to 66 above, which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:41.
68. The fusion protein according to 66 above, which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added relative to the amino acid sequence shown in SEQ ID NO:41.
69. The fusion protein according to 66 above, which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added relative to the amino acid sequence shown in SEQ ID NO:41.
70. The fusion protein according to claim 51, comprising the amino acid sequence shown in SEQ ID NO:41.
71. The fusion protein according to any one of 38 to 70 above, which has a specific activity of 10% or more compared with the specific activity of normal wild-type hGBA.
72. A DNA comprising a gene encoding the fusion protein according to any one of 1 to 71 above.
73. An expression vector comprising the DNA according to 72 above.
74. A mammalian cell transformed with the expression vector according to 73 above.
75. A method for producing a fusion protein, comprising the step of culturing the mammalian cell according to 74 above in a serum-free medium.
76. A conjugate of the fusion protein according to any one of 1 to 170 above with an antibody.
77. Conjugates of lysosomal enzymes, serum albumin and antibodies.
78. The conjugate according to claim 77, which is selected from the following (1) to (6):
(1) A conjugate in which serum albumin is bound to the C-terminus of the lysosomal enzyme directly or via a linker, and the antibody is further bound to the C-terminus of the serum albumin directly or via a linker;
(2) A conjugate in which an antibody is bound to the C-terminus of the lysosomal enzyme directly or via a linker, and the serum albumin is further bound to the C-terminus of the antibody directly or via a linker;
(3) A conjugate in which the lysosomal enzyme is bound to the C-terminus of the serum albumin directly or via a linker, and the antibody is further bound to the C-terminus of the lysosomal enzyme directly or via a linker;
(4) A conjugate in which the antibody is bound to the C-terminus of the serum albumin, either directly or via a linker, and the lysosomal enzyme is further bound to the C-terminus of the antibody, either directly or via a linker;
(5) A conjugate in which the lysosomal enzyme is bound to the C-terminus of the antibody, either directly or via a linker, and the serum albumin is further bound to the C-terminus of the lysosomal enzyme, either directly or via a linker;
(6) A conjugate in which the serum albumin is bound to the C-terminus of the antibody, either directly or via a linker, and the lysosomal enzyme is further bound to the C-terminus of the serum albumin, either directly or via a linker.
79. The conjugate according to any one of claims 76 to 78, wherein the antibody is an antibody against a receptor expressed on a vascular endothelial cell.
80. The conjugate according to claim 79, wherein the receptor on a vascular endothelial cell is selected from the group consisting of an insulin receptor, a transferrin receptor, a leptin receptor, a lipoprotein receptor, and an IGF receptor.
81. The conjugate according to claim 79, wherein the receptor on vascular endothelial cells is a transferrin receptor.
82. The conjugate according to any one of 76 to 81 above, wherein the antibody is any one of a Fab antibody, an F(ab') 2 antibody, an F(ab')2 antibody, a single domain antibody, a single chain antibody, a VHH, or an Fc antibody.
83. The conjugate according to any one of 76 to 82 above, wherein the fusion protein is bound to either the C-terminus or the N-terminus of the light chain of the antibody.
84. The conjugate according to any one of 76 to 82 above, wherein the fusion protein is bound to either the C-terminus or the N-terminus of the heavy chain of the antibody.
85. The conjugate according to any one of 76 to 82 above, wherein the fusion protein is bound to either the C-terminal or N-terminal side of the light chain, or to either the C-terminal or N-terminal side of the heavy chain of the antibody, via a linker sequence.
86. The conjugate according to any one of claims 78 to 85, wherein the linker sequence consists of 1 to 50 amino acid residues.
87. The conjugate according to claim 86, wherein the linker sequence comprises an amino acid sequence selected from the group consisting of one glycine, one serine, the amino acid sequence Gly-Ser, the amino acid sequence Ser-Ser, the amino acid sequence Gly-Gly-Ser, the amino acid sequence of SEQ ID NO:9, the amino acid sequence of SEQ ID NO:10, the amino acid sequence of SEQ ID NO:11, and an amino acid sequence consisting of 1 to 10 consecutive amino acids of these amino acid sequences.
88. A DNA comprising a gene encoding the conjugate according to any one of 76 to 87 above.
89. An expression vector comprising the DNA according to 88 above.
90. A mammalian cell transformed with the expression vector according to 89 above.
91. A method for producing a conjugate of an antibody and a fusion protein of a physiologically active protein and SA, comprising the step of culturing the mammalian cell according to 90 above in a serum-free medium.
 本発明によれば,例えば,活性のある組換え蛋白質として発現させることが比較的困難なヒトリソソーム酵素を,HSAとの融合蛋白質の形態で提供することができる。かかる融合蛋白質は,活性の高い組換え蛋白質として効率よく製造できるので,当該リソソーム酵素の欠損するリソソーム病の患者の酵素補充療法のために用いる薬剤として,医療機関に安定して供給することができる。また,本発明によれば,例えば,活性のある組換え蛋白質として発現させることが比較的困難なサイトカインを,HSAとの融合蛋白質の形態で提供することができる。かかる融合蛋白質は,組換え蛋白質として効率よく製造できるので,薬剤として,医療機関に安定して供給することができる。また,本発明によれば,例えば,活性のある組換え蛋白質として発現させることが比較的困難なヒト神経栄養因子を,HSAとの融合蛋白質の形態で提供することができる。かかる融合蛋白質は,組換え蛋白質として効率よく製造できるので,薬剤として,医療機関に安定して供給することができる。なお,本発明の効果は,これらに限定されるものではない。 According to the present invention, for example, a human lysosomal enzyme that is relatively difficult to express as an active recombinant protein can be provided in the form of a fusion protein with HSA. Since such a fusion protein can be efficiently produced as a highly active recombinant protein, it can be stably supplied to medical institutions as a drug used for enzyme replacement therapy for patients with lysosomal diseases in which the lysosomal enzyme is deficient. In addition, according to the present invention, for example, a cytokine that is relatively difficult to express as an active recombinant protein can be provided in the form of a fusion protein with HSA. Since such a fusion protein can be efficiently produced as a recombinant protein, it can be stably supplied to medical institutions as a drug. In addition, according to the present invention, for example, a human neurotrophic factor that is relatively difficult to express as an active recombinant protein can be provided in the form of a fusion protein with HSA. Since such a fusion protein can be efficiently produced as a recombinant protein, it can be stably supplied to medical institutions as a drug. Note that the effects of the present invention are not limited to these.
N末端側からHSA,リンカー及びhGALCを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とhGALCのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, HSA, a linker, and hGALC. The linker is a peptide linker, and the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hGALC by a peptide bond. N末端側からHSA,リンカー及びhGBAを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とhGBAのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having HSA, a linker, and hGBA in that order from the N-terminus. The linker is a peptide linker, and the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hGBA by a peptide bond. N末端側からhGALC,リンカー及びHSAを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はhGALCのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hGALC, a linker, and HSA. The linker is a peptide linker, and the fusion protein is such that the C-terminus of hGALC is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of HSA by a peptide bond. N末端側からhGBA,リンカー及びHSAを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はhGBAのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hGBA, a linker, and HSA. The linker is a peptide linker, and the fusion protein is such that the C-terminus of hGBA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of HSA by a peptide bond. N末端側からHSA,リンカー及びhIL-10を順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とhIL-10のN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, HSA, a linker, and hIL-10. The linker is a peptide linker, and the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hIL-10 by a peptide bond. N末端側からhIL-10,リンカー及びHSAを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はhIL-10のC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hIL-10, a linker, and HSA. The linker is a peptide linker, and the fusion protein is such that the C-terminus of hIL-10 is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of HSA by a peptide bond. N末端側からHSA,リンカー及びhBDNFを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とhBDNFのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, HSA, a linker, and hBDNF. The linker is a peptide linker, and the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hBDNF by a peptide bond. N末端側からHSA,リンカー及びhNGFを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とhNGFのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having HSA, a linker, and hNGF in that order from the N-terminus. The linker is a peptide linker, and the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hNGF by a peptide bond. N末端側からHSA,リンカー及びhNT-3を順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とhNT-3のN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, HSA, a linker, and hNT-3. The linker is a peptide linker, and the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hNT-3 by a peptide bond. N末端側からHSA,リンカー及びhNT-4を順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とhNT-4のN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, HSA, a linker, and hNT-4. The linker is a peptide linker, and the fusion protein is such that the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hNT-4 by a peptide bond. N末端側からhBDNF,リンカー及びHSAを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はhBDNFのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hBDNF, a linker, and HSA. The linker is a peptide linker, and the fusion protein is such that the C-terminus of hBDNF and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond. N末端側からhNGF,リンカー及びHSAを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はhNGFのC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hNGF, a linker, and HSA. The linker is a peptide linker, and the fusion protein is such that the C-terminus of hNGF and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond. N末端側からhNT-3,リンカー及びHSAを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はhNT-3のC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hNT-3, a linker, and HSA. The linker is a peptide linker, and the fusion protein is such that the C-terminus of hNT-3 and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond. N末端側からhNT-4,リンカー及びHSAを順に有する一本鎖ポリペプチドの融合蛋白質を模式的に示す。リンカーは,ペプチドリンカーであり,融合蛋白質はhNT-4のC末端と該リンカーのN末端がペプチド結合により結合し,かつ該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。The figure shows a schematic diagram of a single-chain polypeptide fusion protein having, in order from the N-terminus, hNT-4, a linker, and HSA. The linker is a peptide linker, and the fusion protein is such that the C-terminus of hNT-4 and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond. 一過性発現による野生型hGALC,HSA-hGALC,及びhGALC-HSAの発現量の確認実験の結果を示す図。上段(a)の棒グラフの縦軸は,培養上清中に含まれる酵素の発現量を酵素活性(μM/時間)で示す。黒棒は野生型hGALCの酵素活性を,白棒はHSA-hGALCの酵素活性を,斜線棒はhGALC-HSAの酵素活性を,それぞれ示す。中段(b)は,培養上清のSDS-pageによる分析結果であり,下段(c)は,培養上清のウエスタンブロッティングによる分析結果であり,野生型hGALC及びHSAとhGALCとの融合蛋白質に相当するバンドの位置がそれぞれ示されている。左から順に,一過性発現における培養開始6日後,7日後,8日後の分析結果を示す。Figure showing the results of an experiment to confirm the expression levels of wild-type hGALC, HSA-hGALC, and hGALC-HSA by transient expression. The vertical axis of the bar graph in the upper row (a) indicates the expression level of the enzyme contained in the culture supernatant in terms of enzyme activity (μM/hour). The black bars indicate the enzyme activity of wild-type hGALC, the white bars indicate the enzyme activity of HSA-hGALC, and the diagonal line bars indicate the enzyme activity of hGALC-HSA. The middle row (b) shows the results of an analysis of the culture supernatant by SDS-page, and the lower row (c) shows the results of an analysis of the culture supernatant by Western blotting, showing the positions of the bands corresponding to wild-type hGALC and the fusion protein of HSA and hGALC. From the left, the analysis results are shown 6, 7, and 8 days after the start of culture in transient expression. 一過性発現による野生型hGALC,HSA-hGALC,及びhGALC-HSAのSE-HPLC分析における溶出プロフィールを示す図。縦の破線はBuffer由来のピークの位置を示し,縦の直線はHSA-hGALC及びhGALC-HSAの単量体に相当するピークの位置を示す。The figure shows the elution profiles of wild-type hGALC, HSA-hGALC, and hGALC-HSA obtained by transient expression in SE-HPLC analysis. The vertical dashed line indicates the position of the peak derived from the buffer, and the vertical straight line indicates the positions of the peaks corresponding to the HSA-hGALC and hGALC-HSA monomers. pCI MCS-modifiedベクター(プラスミド)の構造を示す模式図Schematic diagram showing the structure of the pCI MCS-modified vector (plasmid) Dual(+)pCI-neoベクター(プラスミド)の構造を示す模式図Schematic diagram showing the structure of Dual(+)pCI-neo vector (plasmid) 一過性発現による野生型hGALC,Fab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabの発現量の確認実験の結果を示す図。縦軸は,培養上清中に含まれる酵素の発現量を酵素活性(μM/時間)で示す。The figure shows the results of an experiment to confirm the expression levels of wild-type hGALC, Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab by transient expression. The vertical axis shows the expression level of the enzyme contained in the culture supernatant in terms of enzyme activity (μM/hour). 一過性発現によるFab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-FabのSE-HPLC分析における溶出プロフィールを示す図。縦の破線はBuffer由来のピークの位置を示す。1 shows the elution profiles of transiently expressed Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab in SE-HPLC analysis. The vertical dashed line indicates the position of the peak derived from the buffer. pEmIGS-hGBAベクター(プラスミド)の構造を示す模式図Schematic diagram showing the structure of pEmIGS-hGBA vector (plasmid) pCIneo-hGBAベクター(プラスミド)の構造を示す模式図Schematic diagram showing the structure of pCIneo-hGBA vector (plasmid) pCIneo-HSA-hGBAベクター(プラスミド)の構造を示す模式図Schematic diagram showing the structure of pCIneo-HSA-hGBA vector (plasmid) 一過性発現による野生型hGBA,HSA-hGBA,及びhGBA-HSAの発現量の確認実験(活性測定)の結果を示す図。縦軸は,培養上清中に含まれる酵素の発現量を酵素活性(μM/時間)で示す。The figure shows the results of an experiment (activity measurement) to confirm the expression levels of wild-type hGBA, HSA-hGBA, and hGBA-HSA by transient expression. The vertical axis shows the expression level of the enzyme contained in the culture supernatant in terms of enzyme activity (μM/hour). 一過性発現による野生型hGBA,HSA-hGBA,及びhGBA-HSAの発現量の確認実験(SDS-PAGE)の結果を示す図。野生型hGBA,及びHSAとhGBAとの融合蛋白質に相当するバンドの位置がそれぞれ示されている。1 shows the results of an experiment (SDS-PAGE) to confirm the expression levels of wild-type hGBA, HSA-hGBA, and hGBA-HSA by transient expression. The positions of the bands corresponding to wild-type hGBA and the fusion protein of HSA and hGBA are shown. 一過性発現による野生型mIL-10,mIL-10-MSA,及びMSA-mIL-10の発現量の確認実験(ELISA)の結果を示す図。縦軸は,培養上清中に含まれる各蛋白質の発現量を濃度(mol/L)で示す。The figure shows the results of an experiment (ELISA) to confirm the expression levels of wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 by transient expression. The vertical axis shows the expression level of each protein contained in the culture supernatant as a concentration (mol/L). 一過性発現による野生型mIL-10,mIL-10-MSA,及びMSA-mIL-10の発現量の確認実験(SDS-PAGE)の結果を示す図。野生型mIL-10,及びMSAとmIL-10との融合蛋白質に相当するバンドの位置がそれぞれ示されている。1 shows the results of an experiment (SDS-PAGE) to confirm the expression levels of wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 by transient expression, in which the positions of the bands corresponding to wild-type mIL-10 and the fusion protein of MSA and mIL-10 are shown. 一過性発現による野生型ヒト神経栄養因子,及びHSAとヒト神経栄養因子との融合蛋白質の発現量の確認実験(ELISA)の結果を示す図。縦軸は,培養上清中に含まれる各蛋白質の発現量を濃度(mol/L)で示す。黒棒は各野生型ヒト神経栄養因子の濃度を,白棒は各ヒト神経栄養因子-HSA融合蛋白質の濃度を,斜線棒は各HSA-ヒト神経栄養因子融合蛋白質の濃度を,それぞれ示す。ヒト神経栄養因子は4種類あり,図中左からhBDNF,hNGF,hNT-4,及びhNT-4の結果が示されている。This figure shows the results of an experiment (ELISA) to confirm the expression levels of wild-type human neurotrophic factors and fusion proteins of HSA and human neurotrophic factors by transient expression. The vertical axis shows the expression level of each protein contained in the culture supernatant as concentration (mol/L). The black bars show the concentration of each wild-type human neurotrophic factor, the white bars show the concentration of each human neurotrophic factor-HSA fusion protein, and the shaded bars show the concentration of each HSA-human neurotrophic factor fusion protein. There are four types of human neurotrophic factors, and from the left in the figure, the results for hBDNF, hNGF, hNT-4, and hNT-4 are shown. 一過性発現による野生型ヒト神経栄養因子,及びHSAとヒト神経栄養因子との融合蛋白質の発現量の確認実験(SDS-PAGE)の結果を示す図。レーン(a)は野生型ヒト神経栄養因子に,レーン(b)はヒト神経栄養因子-HSA融合蛋白質に,レーン(c)はHSA-ヒト神経栄養因子融合蛋白質に,それぞれ対応する。野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質に相当するバンドの位置がそれぞれ示されている。ヒト神経栄養因子は4種類あり,図中左からhBDNF,hNGF,hNT-4,及びhNT-4の結果が示されている。Figure showing the results of an experiment (SDS-PAGE) to confirm the expression levels of wild-type human neurotrophic factor and the fusion protein of HSA and human neurotrophic factor by transient expression. Lane (a) corresponds to wild-type human neurotrophic factor, lane (b) to human neurotrophic factor-HSA fusion protein, and lane (c) to HSA-human neurotrophic factor fusion protein. The positions of the bands corresponding to wild-type human neurotrophic factor and the fusion protein of HSA and human neurotrophic factor are shown. There are four types of human neurotrophic factors, and from the left in the figure, the results for hBDNF, hNGF, hNT-4, and hNT-4 are shown. 一過性発現による野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質の発現量の確認実験(ウエスタンブロッティング)の結果を示す図。レーン(a)は野生型ヒト神経栄養因子に,レーン(b)はヒト神経栄養因子-HSA融合蛋白質に,レーン(c)はHSA-ヒト神経栄養因子融合蛋白質に,それぞれ対応する。野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質に相当するバンドの位置がそれぞれ示されている。ヒト神経栄養因子は4種類あり,図中左からhBDNF,hNGF,hNT-4,及びhNT-4の結果が示されている。Figure showing the results of an experiment (Western blotting) to confirm the expression levels of wild-type human neurotrophic factor and the fusion protein of HSA and human neurotrophic factor by transient expression. Lane (a) corresponds to wild-type human neurotrophic factor, lane (b) to human neurotrophic factor-HSA fusion protein, and lane (c) to HSA-human neurotrophic factor fusion protein. The positions of the bands corresponding to wild-type human neurotrophic factor and the fusion protein of HSA and human neurotrophic factor are shown. There are four types of human neurotrophic factors, and from the left in the figure, the results for hBDNF, hNGF, hNT-4, and hNT-4 are shown.
 本発明において,血清アルブミン(SA)と融合させるべき蛋白質の種類に特に制限はないが,当該蛋白質をコードする遺伝子を宿主細胞に組込んで組換え蛋白質として発現されたときに,発現量が低く,及び/又は,活性が低いものである。かかる蛋白質としては,例えば,リソソーム酵素,サイトカイン,インターロイキン,及び神経栄養因子,若しくはこれらと抗体の融合蛋白質が挙げられる。なお,インターロイキンはサイトカインに属するものであり,特にヘルパーT細胞から分布されるサイトカインの総称である。 In the present invention, there is no particular limit to the type of protein to be fused with serum albumin (SA), but the protein should be one that has a low expression level and/or low activity when the gene encoding the protein is introduced into a host cell and expressed as a recombinant protein. Examples of such proteins include lysosomal enzymes, cytokines, interleukins, and neurotrophic factors, or fusion proteins of these with antibodies. Interleukins belong to the cytokine category, and are a general term for cytokines distributed in particular from helper T cells.
 リソソーム酵素としては,特にガラクトシルセラミダーゼ(GALC)及びグルコセレブロシダーゼ(GBA),若しくはこれらと抗体又はリガンドの融合蛋白質が挙げられる。但し,リソソーム酵素については,GALCとGBAに限られない。SAと結合させることにより,組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量,及び/又は,活性を高めることができる他のリソソーム酵素も,SAと結合させるべきリソソーム酵素に含まれる。これら他のリソソーム酵素と抗体又はリガンドの融合蛋白質についても同様である。更には,本発明は,組換え蛋白質として製造が容易なリソソーム酵素にも応用が可能である。すなわち,リリソソーム酵素に特に限定はなく,例えば,イズロン酸-2-スルファターゼ,α-L-イズロニダーゼ,β-ガラクトシダーゼ,GM2活性化蛋白質,β-ヘキソサミニダーゼA,β-ヘキソサミニダーゼB,N-アセチルグルコサミン1-フォスフォトランスフェラーゼ,α-マンノシダーゼ,β-マンノシダーゼ,サポシンC,アリールスルファターゼA,α-L-フコシダーゼ,アスパルチルグルコサミニダーゼ,α-N-アセチルガラクトサミニダーゼ,酸性スフィンゴミエリナーゼ,α-ガラクトシダーゼA,β-グルクロニダーゼ,ヘパランN-スルファターゼ,α-N-アセチルグルコサミニダーゼ,アセチルCoAα-グルコサミニドN-アセチルトランスフェラーゼ,N-アセチルグルコサミン6-硫酸スルファターゼ,酸性セラミダーゼ,アミロ-1,6-グルコシダーゼ,シアリダーゼ,パルミトイル蛋白質チオエステラーゼ-1,トリペプチジルペプチダーゼ-1,ヒアルロニダーゼ-1,酸性α-グルコシダーゼ,CLN1及びCLN2であってもよい。 Lysosomal enzymes include, in particular, galactosylceramidase (GALC) and glucocerebrosidase (GBA), or fusion proteins of these with antibodies or ligands. However, lysosomal enzymes are not limited to GALC and GBA. Other lysosomal enzymes that can increase the expression level and/or activity when expressed as recombinant proteins by binding with SA, particularly when expressed so that the recombinant proteins are secreted from cells and accumulated in the culture medium, are also included in the lysosomal enzymes to be bound with SA. The same applies to fusion proteins of these other lysosomal enzymes with antibodies or ligands. Furthermore, the present invention can be applied to lysosomal enzymes that are easy to produce as recombinant proteins. That is, the lysosomal enzyme is not particularly limited, and examples thereof include iduronate-2-sulfatase, α-L-iduronidase, β-galactosidase, GM2 activator protein, β-hexosaminidase A, β-hexosaminidase B, N-acetylglucosamine 1-phosphotransferase, α-mannosidase, β-mannosidase, saposin C, arylsulfatase A, α-L-fucosidase, aspartylglucosaminidase, α-N-acetylgalactosaminidase, acid sphingomyelinase, and the like. It may be enzyme, α-galactosidase A, β-glucuronidase, heparan N-sulfatase, α-N-acetylglucosaminidase, acetyl-CoA α-glucosaminide N-acetyltransferase, N-acetylglucosamine 6-sulfate sulfatase, acid ceramidase, amylo-1,6-glucosidase, sialidase, palmitoyl protein thioesterase-1, tripeptidyl peptidase-1, hyaluronidase-1, acid α-glucosidase, CLN1, or CLN2.
 サイトカインとしては,特にインターロイキンであり,例えば,IL-10,及びこれと抗体又はリガンドの融合蛋白質が挙げられる。但し,サイトカインについては,IL-10に限られない。SAと結合させることにより,組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量,及び/又は,活性を高めることができる他のサイトカインも,SAと結合させるべきサイトカインに含まれる。これら他のサイトカインと抗体又はリガンドの融合蛋白質についても同様である。但し,本発明は,組換え蛋白質として製造が容易なサイトカインにも応用が可能である。つまり,融合蛋白質には,IL-10以外のサイトカインとSAとの融合蛋白質,更にはこれらと抗体の融合蛋白質も包含される。ここで,サイトカインに特に限定はないが,例えば,IL-1,IL-2,IL-3,IL-4,IL-5,IL-6,IL-7,IL-8,IL-9,IL-11,IL-12,IL-13,IL-14,IL-15,IL-16,IL-17,IL-18,及びIL-19~IL-36であってもよい。 Cytokines include interleukins, such as IL-10, and fusion proteins of IL-10 with antibodies or ligands. However, the cytokines are not limited to IL-10. Other cytokines that can increase the expression level and/or activity when expressed as recombinant proteins by binding with SA, particularly when expressed so that the recombinant proteins are secreted from cells and accumulate in the culture medium, are also included in the cytokines to be bound with SA. The same applies to fusion proteins of these other cytokines with antibodies or ligands. However, the present invention can also be applied to cytokines that are easy to produce as recombinant proteins. In other words, fusion proteins also include fusion proteins of cytokines other than IL-10 with SA, and further fusion proteins of these with antibodies. Here, the cytokine is not particularly limited, but may be, for example, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, and IL-19 to IL-36.
 神経栄養因子としては,特にBDNF,NGF,NT-3,及びNT-4及びこれらと抗体の融合蛋白質が挙げられる。但し,神経栄養因子については,これらに限られない。SAと結合させることにより,組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,発現量,及び/又は,活性を高めることができる他の神経栄養因子も,SAと結合させるべき神経栄養因子に含まれる。これら神経栄養因子と抗体の融合蛋白質についても同様である。更には,本発明は,組換え蛋白質として製造が容易な神経栄養因子にも応用が可能である。すなわち,神経栄養因子に特に限定はなく,例えば,グリア細胞株神経栄養因子(GDNF),NT-5であってもよい。 The neurotrophic factors include, in particular, BDNF, NGF, NT-3, and NT-4, and fusion proteins of these with antibodies. However, the neurotrophic factors are not limited to these. Other neurotrophic factors that can increase the expression level and/or activity when expressed as a recombinant protein by binding with SA, particularly when expressed so that the recombinant protein is secreted from cells and accumulated in the culture medium, are also included in the neurotrophic factors to be bound with SA. The same applies to fusion proteins of these neurotrophic factors and antibodies. Furthermore, the present invention can be applied to neurotrophic factors that are easy to produce as recombinant proteins. In other words, the neurotrophic factors are not particularly limited, and may be, for example, glial cell line neurotrophic factor (GDNF) or NT-5.
 SAと融合させる蛋白質の生物種に特に限定はないが,好ましくはヒトに由来するものであり,例えばヒトリソソーム酵素,ヒトサイトカイン,ヒト成長栄養因子である。 There are no particular limitations on the biological species of the protein to be fused with SA, but it is preferably derived from humans, such as human lysosomal enzymes, human cytokines, and human growth and nutritional factors.
 血清アルブミン(SA)とリソソーム酵素との融合蛋白質は,リソソーム病に対する酵素補充療法における治療剤として使用できる。例えば,SAと融合させたグルコセレブロシダーゼ(GBA)はゴーシェ病における治療剤として,ガラクトシルセラミダーゼ(GALC)はクラッベ病における治療剤として使用できる。 Fusion proteins of serum albumin (SA) and lysosomal enzymes can be used as therapeutic agents in enzyme replacement therapy for lysosomal diseases. For example, glucocerebrosidase (GBA) fused with SA can be used as a therapeutic agent for Gaucher disease, and galactosylceramidase (GALC) can be used as a therapeutic agent for Krabbe disease.
 本明細書において,単に「ヒトリソソーム酵素」というときは,通常の野生型のヒトリソソーム酵素に加え,当該ヒトリソソーム酵素の種類に対応する酵素活性を有する等のヒトリソソーム酵素としての機能を有するものである限り,野生型のヒトリソソーム酵素のアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するヒトリソソーム酵素の変異体も特に区別することなく包含する。ヒト以外の動物種のリソソーム酵素についても同様である。 In this specification, when the term "human lysosomal enzyme" is used, it includes not only normal wild-type human lysosomal enzymes, but also mutants of human lysosomal enzymes in which one or more amino acid residues have been substituted, deleted, and/or added (in this specification, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence of the wild-type human lysosomal enzyme, as long as the mutants have the function of a human lysosomal enzyme, such as having the enzymatic activity corresponding to the type of human lysosomal enzyme. The same applies to lysosomal enzymes of animal species other than humans.
 なお,ここで,ヒトリソソーム酵素がヒトリソソーム酵素としての機能を有するというときは,ヒトリソソーム酵素が,通常の野生型のヒトリソソーム酵素の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。ここで比活性とは蛋白質の質量当たりの酵素活性のことをいう。なお,ヒトリソソーム酵素とその他の蛋白質との融合蛋白質の比活性は,融合蛋白質の中でヒトリソソーム酵素に相当する部分の質量当たりの酵素活性として求められる。ヒト以外の動物種のリソソーム酵素についても同様のことがいえる。なお,ここで融合蛋白質におけるヒトリソソーム酵素の比活性は,当該融合蛋白質の単位質量当たりのヒトリソソーム酵素の酵素活性(μM/時間/mg蛋白質)に(当該融合蛋白質の分子量/当該融合蛋白質中のヒトリソソーム酵素に相当する部分の分子量)を乗じて算出される。 Here, when a human lysosomal enzyme is said to have the function of a human lysosomal enzyme, it means that the human lysosomal enzyme has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of a normal wild-type human lysosomal enzyme is taken as 100%. Here, specific activity refers to the enzyme activity per mass of protein. The specific activity of a fusion protein of a human lysosomal enzyme and another protein is calculated as the enzyme activity per mass of the portion of the fusion protein that corresponds to the human lysosomal enzyme. The same can be said for lysosomal enzymes of animal species other than humans. Here, the specific activity of the human lysosomal enzyme in the fusion protein is calculated by multiplying the enzyme activity of the human lysosomal enzyme per unit mass of the fusion protein (μM/hour/mg protein) by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein that corresponds to the human lysosomal enzyme).
 野生型のヒトリソソーム酵素のアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のヒトリソソーム酵素のアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のヒトリソソーム酵素のN末端又はC末端のアミノ酸残基を1個欠失させたアミノ酸配列からなるヒトリソソーム酵素の変異体,野生型のヒトリソソーム酵素のN末端又はC末端のアミノ酸残基を2個欠失させたアミノ酸配列からなるヒトリソソーム酵素の変異体等も,ヒトリソソーム酵素である。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型のヒトリソソーム酵素のアミノ酸配列に加えることもできる。ヒト以外の動物種のリソソーム酵素についても同様のことがいえる。 When amino acid residues in the amino acid sequence of a wild-type human lysosomal enzyme are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of a wild-type human lysosomal enzyme are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. A mutant human lysosomal enzyme consisting of an amino acid sequence in which one amino acid residue is deleted from the N-terminus or C-terminus of a wild-type human lysosomal enzyme, a mutant human lysosomal enzyme consisting of an amino acid sequence in which two amino acid residues are deleted from the N-terminus or C-terminus of a wild-type human lysosomal enzyme, etc. are also human lysosomal enzymes. In addition, a mutation that combines the substitution and deletion of these amino acid residues can be added to the amino acid sequence of a wild-type human lysosomal enzyme. The same can be said for lysosomal enzymes of animal species other than humans.
 野生型のヒトリソソーム酵素のアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はヒトリソソーム酵素のアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型のヒトリソソーム酵素のアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型のヒトリソソーム酵素のアミノ酸配列に加えることもできる。ヒト以外の動物種のリソソーム酵素についても同様のことがいえる。 When adding amino acid residues to the amino acid sequence of a wild-type human lysosomal enzyme, one or more amino acid residues are added into the amino acid sequence of the human lysosomal enzyme or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of a wild-type human lysosomal enzyme, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of a wild-type human lysosomal enzyme. The same can be said for lysosomal enzymes of animal species other than humans.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型のヒトリソソーム酵素のアミノ酸配列に加えることもできる。例えば,野生型のヒトリソソーム酵素のアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもヒトリソソーム酵素であり,野生型のヒトリソソーム酵素のアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもヒトリソソーム酵素であり,野生型のヒトリソソーム酵素のアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもヒトリソソーム酵素であり,野生型のヒトリソソーム酵素のアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもヒトリソソーム酵素であり,野生型のヒトリソソーム酵素のアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもヒトリソソーム酵素である。ヒト以外の動物種のリソソーム酵素についても同様のことがいえる。 Furthermore, a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of a wild-type human lysosomal enzyme. For example, a human lysosomal enzyme is obtained by deleting 1 to 10 amino acid residues from the amino acid sequence of a wild-type human lysosomal enzyme, substituting 1 to 10 amino acid residues with other amino acid residues, and adding 1 to 10 amino acid residues; a human lysosomal enzyme is obtained by deleting 1 to 5 amino acid residues from the amino acid sequence of a wild-type human lysosomal enzyme, substituting 1 to 5 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues; a human lysosomal enzyme is obtained by deleting 1 to 3 amino acid residues from the amino acid sequence of a wild-type human lysosomal enzyme, substituting 1 to 3 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues; Human lysosomal enzymes are those in which an amino acid residue has been replaced with another amino acid residue, and 1 to 3 amino acid residues have been added. Human lysosomal enzymes are also those in which 1 or 2 amino acid residues have been deleted from the amino acid sequence of a wild-type human lysosomal enzyme, 1 or 2 amino acid residues have been replaced with another amino acid residue, and 1 or 2 amino acid residues have been added. Human lysosomal enzymes are also those in which 1 amino acid residue has been deleted from the amino acid sequence of a wild-type human lysosomal enzyme, 1 amino acid residue has been replaced with another amino acid residue, and 1 amino acid residue has been added. The same can be said about lysosomal enzymes of animal species other than humans.
 ヒトリソソーム酵素変異体における通常の野生型ヒトリソソーム酵素と比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両ヒトリソソーム酵素のアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種のリソソーム酵素についても同様のことがいえる。 The location and type (deletion, substitution, and addition) of each mutation in a human lysosomal enzyme mutant compared to a normal wild-type human lysosomal enzyme can be easily confirmed by aligning the amino acid sequences of both human lysosomal enzymes. The same can be said for lysosomal enzymes of animal species other than humans.
 ヒトリソソーム酵素変異体のアミノ酸配列は,通常の野生型ヒトリソソーム酵素のアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。ヒト以外の動物種のリソソーム酵素変異体についても同様のことがいえる。 The amino acid sequence of the human lysosomal enzyme mutant preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, to the amino acid sequence of a normal wild-type human lysosomal enzyme. The same can be said for lysosomal enzyme mutants of animal species other than humans.
 野生型ヒトリソソーム酵素のアミノ酸配列とヒトリソソーム酵素変異体のアミノ酸配列との同一性,周知の相同性計算アルゴリズムを用いて容易に算出することができる。例えば,そのようなアルゴリズムとして,BLAST(Altschul SF. J Mol .Biol. 215. 403-10, (1990)),Pearson及びLipmanの類似性検索法(Proc. Natl. Acad. Sci. USA. 85. 2444 (1988)),Smith及びWatermanの局所相同性アルゴリズム(Adv. Appl. Math. 2. 482-9(1981))等がある。ヒト以外の動物種のリソソーム酵素, GALC, GBA,MSA及びヒト以外の動物種SAについても同様のことがいえる。これらアルゴリズムは,本明細書全体を通じて,他の蛋白質の野生型のアミノ酸配列と,当該他の蛋白質の変異体のアミノ酸配列の相同性計算アルゴリズムとしても適用することができる。 The identity between the amino acid sequence of a wild-type human lysosomal enzyme and the amino acid sequence of a mutant human lysosomal enzyme can be easily calculated using well-known homology calculation algorithms. Examples of such algorithms include BLAST (Altschul S F. J Mol. Biol. 215. 403-10, (1990)), the similarity search method of Pearson and Lipman (Proc. Natl. Acad. Sci. USA. 85. 2444 (1988)), and the local homology algorithm of Smith and Waterman (Adv. Appl. Math. 2. 482-9 (1981)). The same can be said for the lysosomal enzymes of non-human animal species, GALC, GBA, MSA, and non-human animal species SA. These algorithms can also be applied throughout this specification as homology calculation algorithms between the wild-type amino acid sequence of another protein and the amino acid sequence of a mutant of that other protein.
 本明細書において,単に「ヒトガラクトシルセラミダーゼ」,「ヒトガラクトセレブロシダーゼ」,又は「hGALC」というときは,配列番号1で示される643個のアミノ酸残基からなる通常の野生型のhGALCに加え,ガラクトセレブロシド及び/又はガラクトシルスフィンゴシン等のスフィンゴ脂質を分解することのできる酵素活性を有する等のhGALCとしての機能を有するものである限り,配列番号1で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するhGALCの変異体も特に区別することなく包含する。野生型のhGALCは,例えば,配列番号2で示される塩基配列を有する遺伝子にコードされる。 In this specification, when simply referring to "human galactosylceramidase", "human galactocerebrosidase", or "hGALC", it includes, without distinction, not only the normal wild-type hGALC consisting of 643 amino acid residues as shown in SEQ ID NO: 1, but also mutants of hGALC in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence as shown in SEQ ID NO: 1, as long as they have the function of hGALC, such as the enzyme activity capable of decomposing sphingolipids such as galactocerebroside and/or galactosylsphingosine. Wild-type hGALC is, for example, encoded by a gene having the base sequence as shown in SEQ ID NO: 2.
 本明細書において,単に「マウスガラクトシルセラミダーゼ」,「マウスガラクトセレブロシダーゼ」,又は「mGALC」というときは,配列番号14で示されるアミノ酸配列を有する通常の野生型のmGALCに加え,ガラクトセレブロシド及び/又はガラクトシルスフィンゴシン等のスフィンゴ脂質を分解することのできる酵素活性を有する等のmGALCとしての機能を有するものである限り,配列番号14で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するmGALCの変異体も特に区別することなく包含する。野生型のmGALCは,例えば,配列番号15で示される塩基配列を有する遺伝子にコードされる。 In this specification, when simply referring to "mouse galactosylceramidase", "mouse galactocerebrosidase", or "mGALC", it includes, without distinction, not only the normal wild-type mGALC having the amino acid sequence shown in SEQ ID NO: 14, but also mGALC mutants in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 14, as long as they have the function of mGALC, such as having the enzymatic activity to degrade sphingolipids such as galactocerebroside and/or galactosylsphingosine. Wild-type mGALC is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 15.
 なお,ここで,hGALCがhGALCとしての機能を有するというときは,hGALCが,通常の野生型のhGALCの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。ここで比活性とは蛋白質の質量当たりの酵素活性のことをいう。なお,hGALCとその他の蛋白質との融合蛋白質の比活性は,融合蛋白質の中でhGALCに相当する部分の質量当たりの酵素活性として求められる。mGALCについても同様のことがいえる。なお,ここで融合蛋白質におけるhGALCの比活性は,当該融合蛋白質の単位質量当たりのhGALCの酵素活性(μM/時間/mg蛋白質)に(当該融合蛋白質の分子量/当該融合蛋白質中のhGALCに相当する部分の分子量)を乗じて算出される。 When hGALC is said to have the function of hGALC, it means that hGALC preferably retains a specific activity of 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, of the specific activity of normal wild-type hGALC taken as 100%. Here, specific activity refers to the enzyme activity per mass of protein. The specific activity of a fusion protein of hGALC and another protein is calculated as the enzyme activity per mass of the portion of the fusion protein that corresponds to hGALC. The same can be said for mGALC. Here, the specific activity of hGALC in a fusion protein is calculated by multiplying the enzyme activity of hGALC per unit mass of the fusion protein (μM/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hGALC).
 野生型のhGALCのアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhGALCのアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhGALCのN末端又はC末端のアミノ酸残基を1個欠失させた642個のアミノ酸残基からなるhGALCの変異体,野生型のhGALCのN末端又はC末端のアミノ酸残基を2個欠失させた641個のアミノ酸残基からなるhGALCの変異体等も,hGALCである。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型のhGALCのアミノ酸配列に加えることもできる。ヒト以外の動物種のGALCについても同様のことがいえる。 When amino acid residues in the amino acid sequence of wild-type hGALC are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of wild-type hGALC are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. hGALC mutants consisting of 642 amino acid residues with one amino acid residue deleted from the N-terminus or C-terminus of wild-type hGALC, and hGALC mutants consisting of 641 amino acid residues with two amino acid residues deleted from the N-terminus or C-terminus of wild-type hGALC are also hGALC. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hGALC. The same can be said for GALC of animal species other than humans.
 野生型のhGALCのアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はhGALCのアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型のhGALCのアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型のhGALCのアミノ酸配列に加えることもできる。ヒト以外の動物種のGALCについても同様のことがいえる。 When amino acid residues are added to the amino acid sequence of wild-type hGALC, one or more amino acid residues are added into the amino acid sequence of hGALC or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hGALC, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hGALC. The same can be said for GALC of animal species other than humans.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型のhGALCのアミノ酸配列に加えることもできる。例えば,配列番号1で示される野生型のhGALCのアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもhGALCであり,配列番号1で示される野生型のhGALCのアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもhGALCであり,配列番号1で示される野生型のhGALCのアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもhGALCであり,配列番号1で示される野生型のhGALCのアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもhGALCであり,配列番号1で示される野生型のhGALCのアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもhGALCである。ヒト以外の動物種のGALCについても同様のことがいえる。 Furthermore, a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of wild-type hGALC. For example, the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1 can be obtained by deleting 1 to 10 amino acid residues, substituting 1 to 10 amino acid residues with other amino acid residues, and adding 1 to 10 amino acid residues. The amino acid sequence of wild-type hGALC shown in SEQ ID NO:1 can be obtained by deleting 1 to 5 amino acid residues, substituting 1 to 5 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues. The amino acid sequence of wild-type hGALC shown in SEQ ID NO:1 can be obtained by deleting 1 to 3 amino acid residues, hGALC also includes those in which one to three amino acid residues have been replaced with other amino acid residues and one to three amino acid residues have been added, and those in which one or two amino acid residues have been deleted from the wild-type hGALC amino acid sequence shown in SEQ ID NO:1, one or two amino acid residues have been replaced with other amino acid residues, and one or two amino acid residues have been added, and those in which one amino acid residue has been deleted from the wild-type hGALC amino acid sequence shown in SEQ ID NO:1, one amino acid residue has been replaced with other amino acid residue, and one amino acid residue has been added. The same can be said for GALCs of animal species other than humans.
 hGALC変異体における通常の野生型hGALCと比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両hGALCのアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種のGALCについても同様のことがいえる。 The location and type (deletion, substitution, and addition) of each mutation in the hGALC mutant compared to the normal wild-type hGALC can be easily confirmed by aligning the amino acid sequences of both hGALCs. The same can be said for GALCs of animal species other than humans.
 hGALC変異体のアミノ酸配列は,配列番号1で示される通常の野生型hGALCのアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。ヒト以外の動物種のGALCについても同様のことがいえる。 The amino acid sequence of the hGALC mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of the normal wild-type hGALC shown in SEQ ID NO: 1. The same can be said for GALC of animal species other than humans.
 本明細書において,単に「ヒトグルコセレブロシダーゼ」,「ヒトβ-グルコシダーゼ」,又は「hGBA」というときは,配列番号37で示される497個のアミノ酸残基からなる通常の野生型のhGBAに加え,ガラクトセレブロシド及び/又はガラクトシルスフィンゴシン等の糖脂質を分解することのできる酵素活性を有する等のhGBAとしての機能を有するものである限り,配列番号37で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するhGBAの変異体も特に区別することなく包含する。野生型のhGBAは,例えば,配列番号38で示される塩基配列を有する遺伝子にコードされる。 In this specification, when simply referring to "human glucocerebrosidase," "human β-glucosidase," or "hGBA," it includes, without distinction, not only the normal wild-type hGBA consisting of 497 amino acid residues as shown in SEQ ID NO: 37, but also mutants of hGBA in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence as shown in SEQ ID NO: 37, as long as they have the function of hGBA, such as having the enzymatic activity to degrade glycolipids such as galactocerebroside and/or galactosylsphingosine. Wild-type hGBA is, for example, encoded by a gene having the base sequence as shown in SEQ ID NO: 38.
 本明細書において,単に「マウスグルコセレブロシダーゼ」,「マウスβ-グルコシダーゼ」,又は「mGBA」というときは,配列番号43で示されるアミノ酸配列を有する通常の野生型のmGBAに加え,ガラクトセレブロシド及び/又はガラクトシルスフィンゴシン等の糖脂質を分解することのできる酵素活性を有する等のmGBAとしての機能を有するものである限り,配列番号43で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するmGBAの変異体も特に区別することなく包含する。野生型のmGBAは,例えば,配列番号44で示される塩基配列を有する遺伝子にコードされる。 In this specification, when simply referring to "mouse glucocerebrosidase", "mouse β-glucosidase", or "mGBA", it includes, without distinction, not only the normal wild-type mGBA having the amino acid sequence shown in SEQ ID NO: 43, but also mGBA mutants in which one or more amino acid residues have been substituted, deleted, and/or added (in this specification, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 43, as long as they have the function of mGBA, such as having the enzymatic activity to degrade glycolipids such as galactocerebroside and/or galactosylsphingosine. Wild-type mGBA is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 44.
 なお,ここで,hGBAがhGBAとしての機能を有するというときは,hGBAが,通常の野生型のhGBAの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。ここで比活性とは蛋白質の質量当たりの酵素活性のことをいう。なお,hGBAとその他の蛋白質との融合蛋白質の比活性は,融合蛋白質の中でhGBAに相当する部分の質量当たりの酵素活性として求められる。なお,ここで融合蛋白質におけるhGBAの比活性は,当該融合蛋白質の単位質量当たりのhGBAの酵素活性(μM/時間/mg蛋白質)に(当該融合蛋白質の分子量/当該融合蛋白質中のhGBAに相当する部分の分子量)を乗じて算出される。ヒト以外の動物種のGBAについても同様のことがいえる。 When hGBA is said to have the function of hGBA, it means that hGBA has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hGBA is taken as 100%. Here, specific activity refers to the enzyme activity per mass of protein. The specific activity of a fusion protein of hGBA and another protein is calculated as the enzyme activity per mass of the portion of the fusion protein that corresponds to hGBA. The specific activity of hGBA in the fusion protein is calculated by multiplying the enzyme activity of hGBA per unit mass of the fusion protein (μM/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hGBA). The same can be said for GBA of animal species other than humans.
 野生型のhGBAのアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhGBAのアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhGBAのN末端又はC末端のアミノ酸残基を1個欠失させた496個のアミノ酸残基からなるhGBAの変異体,野生型のhGBAのN末端又はC末端のアミノ酸残基を2個欠失させた495個のアミノ酸残基からなるhGBAの変異体等も,hGBAである。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型のhGBAのアミノ酸配列に加えることもできる。ヒト以外の動物種のGBAについても同様のことがいえる。 When amino acid residues in the amino acid sequence of wild-type hGBA are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of wild-type hGBA are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. hGBA mutants consisting of 496 amino acid residues with one amino acid residue deleted from the N-terminus or C-terminus of wild-type hGBA, and hGBA mutants consisting of 495 amino acid residues with two amino acid residues deleted from the N-terminus or C-terminus of wild-type hGBA are also hGBA. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hGBA. The same can be said for GBAs of animal species other than humans.
 野生型のhGBAのアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はhGBAのアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型のhGBAのアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型のhGBAのアミノ酸配列に加えることもできる。ヒト以外の動物種のGBAについても同様のことがいえる。 When amino acid residues are added to the amino acid sequence of wild-type hGBA, one or more amino acid residues are added into the amino acid sequence of hGBA or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hGBA, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hGBA. The same can be said for GBAs of animal species other than humans.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型のhGBAのアミノ酸配列に加えることもできる。例えば,配列番号37で示される野生型のhGBAのアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもhGBAであり,配列番号37で示される野生型のhGBAのアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもhGBAであり,配列番号37で示される野生型のhGBAのアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもhGBAであり,配列番号37で示される野生型のhGBAのアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもhGBAであり,配列番号37で示される野生型のhGBAのアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもhGBAである。ヒト以外の動物種のGBAについても同様のことがいえる。 Furthermore, a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of wild-type hGBA. For example, an hGBA is obtained by deleting 1 to 10 amino acid residues from the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, substituting 1 to 10 amino acid residues with other amino acid residues, and adding 1 to 10 amino acid residues. An hGBA is also obtained by deleting 1 to 5 amino acid residues from the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, substituting 1 to 5 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues. An hGBA is also obtained by deleting 1 to 3 amino acid residues from the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, hGBA also includes the amino acid sequence of wild-type hGBA shown in SEQ ID NO: 37 in which one or two amino acid residues have been deleted, one or two amino acid residues have been replaced with other amino acid residues, and one or two amino acid residues have been added. hGBA also includes the amino acid sequence of wild-type hGBA shown in SEQ ID NO: 37 in which one amino acid residue has been deleted, one amino acid residue has been replaced with other amino acid residue, and one amino acid residue has been added. The same can be said for GBAs of animal species other than humans.
 hGBA変異体における通常の野生型hGBAと比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両hGBAのアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種のGBAについても同様のことがいえる。 The location and type (deletion, substitution, and addition) of each mutation in the hGBA mutant compared to the normal wild-type hGBA can be easily confirmed by aligning the amino acid sequences of both hGBAs. The same can be said for GBAs of animal species other than humans.
 hGBA変異体のアミノ酸配列は,配列番号37で示される通常の野生型hGBAのアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。ヒト以外の動物種のGBAについても同様のことがいえる。 The amino acid sequence of the hGBA mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type hGBA shown in SEQ ID NO: 37. The same is true for GBA of animal species other than humans.
 本発明において,血清アルブミン(SA)の生物種に特に限定はないが,好ましくはヒト血清アルブミン(HSA)である。 In the present invention, there is no particular limitation on the biological species of serum albumin (SA), but human serum albumin (HSA) is preferred.
 本発明において,「ヒト血清アルブミン」又は「HSA」というときは,配列番号3で示されるアミノ酸配列を有する585個のアミノ酸からなる野生型のヒト血清アルブミンに加え,配列番号3で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するHSAの変異体も特に区別することなく包含する。野生型のHSAは,例えば,配列番号4で示される塩基配列を有する遺伝子にコードされる。 In the present invention, the term "human serum albumin" or "HSA" includes, without distinction, wild-type human serum albumin consisting of 585 amino acids having the amino acid sequence shown in SEQ ID NO: 3, as well as mutants of HSA in which one or more amino acid residues have been substituted, deleted, and/or added to the amino acid sequence shown in SEQ ID NO: 3 (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence). Wild-type HSA is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 4.
 ヒト血清アルブミンは,これを野生型ヒトリソソーム酵素と結合させた融合蛋白質としてCHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,少なくとも1種類の野生型ヒトリソソーム酵素について,当該野生型ヒトリソソーム酵素を同様の手法により組換え蛋白質として発現させたときと比較して,ヒトリソソーム酵素としての発現量を増加させることができるものである。特に,ヒト血清アルブミンは,これを野生型hGALC又は野生型hGBAと結合させた融合蛋白質としてCHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,これら野生型酵素を同様の手法により組換え蛋白質として発現させたときと比較して,酵素としての発現量を増加させることができるものである。なお,ここでヒト血清アルブミンは,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 When human serum albumin is expressed as a recombinant protein using host cells such as CHO as a fusion protein bound to a wild-type human lysosomal enzyme, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of at least one type of wild-type human lysosomal enzyme as a human lysosomal enzyme can be increased compared to when the wild-type human lysosomal enzyme is expressed as a recombinant protein by a similar method. In particular, when human serum albumin is expressed as a recombinant protein using host cells such as CHO as a fusion protein bound to wild-type hGALC or wild-type hGBA, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of the enzyme can be increased compared to when these wild-type enzymes are expressed as recombinant proteins by a similar method. Note that the human serum albumin here is preferably one that has the functions of human serum albumin, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited thereto.
 本発明において,「マウス血清アルブミン」又は「MSA」というときは,配列番号16で示されるアミノ酸配列を有する野生型のマウス血清アルブミンに加え,配列番号16で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するMSAの変異体も特に区別することなく包含する。野生型のMSAは,例えば,配列番号17で示される塩基配列を有する遺伝子にコードされる。また,MSAは,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のMSAとしての機能を有するものであることが好ましいが,これに限定されない。 In the present invention, the term "mouse serum albumin" or "MSA" includes, without distinction, wild-type mouse serum albumin having the amino acid sequence shown in SEQ ID NO: 16, as well as mutants of MSA in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 16. Wild-type MSA is encoded, for example, by a gene having the base sequence shown in SEQ ID NO: 17. In addition, MSA is preferably one that has the functions of MSA, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited to this.
 本発明において,「血清アルブミン」又は「SA」というときは,ヒト以外の他の哺乳動物の血清アルブミンを含む包括的な概念であり,マウス血清アルブミン(MSA)及びウシ血清アルブミン(BSA)もこれに含まれる。血清アルブミンは,これを野生型ヒトリソソーム酵素と結合させた融合蛋白質としてCHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,少なくとも1種類の野生型ヒトリソソーム酵素について,当該野生型ヒトリソソーム酵素を同様の手法により組換え蛋白質として発現させたときと比較して,ヒトリソソーム酵素としての発現量を増加させることができるものである。特に,血清アルブミンは,これを野生型hGALC又は野生型hGBAと結合させた融合蛋白質としてCHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,これら野生型酵素を同様の手法により組換え蛋白質として発現させたときと比較して,酵素としての発現量を増加させることができるものである。 In the present invention, "serum albumin" or "SA" refers to a comprehensive concept including serum albumin of mammals other than humans, including mouse serum albumin (MSA) and bovine serum albumin (BSA). When serum albumin is expressed as a recombinant protein using host cells such as CHO as a fusion protein bound to a wild-type human lysosomal enzyme, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, it is possible to increase the expression amount of at least one type of wild-type human lysosomal enzyme as a human lysosomal enzyme, compared to when the wild-type human lysosomal enzyme is expressed as a recombinant protein by the same method. In particular, when serum albumin is expressed as a recombinant protein using host cells such as CHO as a fusion protein bound to wild-type hGALC or wild-type hGBA, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, it is possible to increase the expression amount of the enzyme, compared to when these wild-type enzymes are expressed as recombinant proteins by the same method.
 ヒト血清アルブミン(HSA)には,野生型のバリアントが複数あることが知られている。ヒト血清アルブミンRedhillはその一つである。ヒト血清アルブミンRedhillは,585個のアミノ酸からなる上記の通常のヒト血清アルブミンのアミノ酸配列と比べて,そのN末端から320番目のアミノ酸残基がアラニンでなくトレオニンであり,且つそのN末端にアルギニン残基が一つ付加している点で異なり,配列番号12で示される586個のアミノ酸からなる。上記アラニンのトレオニンへの変化により,ヒト血清アルブミンRedhillではそのアミノ酸配列中にAsn-Tyr-Thrで表される配列が生じ,この配列中のAsn(アスパラギン)残基がN-結合型グリコシド化される。このヒト血清アルブミンRedhillも野生型のHSAである。 Human serum albumin (HSA) is known to have several wild-type variants. Human serum albumin Redhill is one of them. Human serum albumin Redhill differs from the above-mentioned normal human serum albumin amino acid sequence, which consists of 585 amino acids, in that the 320th amino acid residue from the N-terminus is threonine instead of alanine, and an arginine residue is added to the N-terminus, and consists of 586 amino acids as shown in SEQ ID NO: 12. The above-mentioned change of alanine to threonine creates a sequence represented by Asn-Tyr-Thr in the amino acid sequence of human serum albumin Redhill, and the Asn (asparagine) residue in this sequence is N-linked glycosylated. This human serum albumin Redhill is also a wild-type HSA.
 配列番号3で示される野生型のHSAのアミノ酸配列のN末端から320番目のアミノ酸残基であるアラニンをトレオニンで置換させた,配列番号13で示される585個のアミノ酸からなるヒト血清アルブミン(HSA-A320T)は,HSA変異体の好適な一例である。また,配列番号13で示されるHSA変異体のN末端からアミノ酸残基である318番目のアスパラギンを保存しつつ,319番目のアミノ酸残基であるチロシンをプロリン以外のアミノ酸に置換させたものも,HSA変異体の好適な一例である。以下,本発明の一実施形態において許容される野生型のHSAアミノ酸配列に加えられる変異について詳述するが,当該変異は,ヒト血清アルブミンRedhill,配列番号13で示されるHSA変異体,又はヒト以外の動物種の血清アルブミンへも適用し得る。 A preferred example of an HSA mutant is human serum albumin (HSA-A320T) consisting of 585 amino acids as shown in SEQ ID NO:13, in which the 320th amino acid residue from the N-terminus of the wild-type HSA amino acid sequence as shown in SEQ ID NO:3 is replaced with threonine. Another preferred example of an HSA mutant is one in which the 319th amino acid residue, tyrosine, is replaced with an amino acid other than proline while preserving the 318th amino acid residue from the N-terminus of the HSA mutant as shown in SEQ ID NO:13. The following describes in detail the mutations that can be made to the wild-type HSA amino acid sequence that are permissible in one embodiment of the present invention, but the mutations can also be applied to human serum albumin Redhill, the HSA mutant as shown in SEQ ID NO:13, or serum albumin of animal species other than humans.
 野生型のHSA又はHSA-A320Tのアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型又はHSA-A320TのHSAのアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型又はHSA-A320TのHSAのN末端又はC末端のアミノ酸残基を1個欠失させたHSAの変異体,野生型又はHSA-A320TのHSAのN末端又はC末端のアミノ酸残基を2個欠失させたHSAの変異体も,HSAである。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型又はHSA-A320TのHSAのアミノ酸配列に加えることもできる。ヒト以外の動物種のSAについても同様のことがいえる。  When amino acid residues in the amino acid sequence of wild-type HSA or HSA-A320T are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of wild-type or HSA-A320T HSA are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. HSA mutants in which one amino acid residue is deleted at the N-terminus or C-terminus of wild-type or HSA-A320T HSA, and HSA mutants in which two amino acid residues are deleted at the N-terminus or C-terminus of wild-type or HSA-A320T HSA are also HSA. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type or HSA-A320T HSA. The same can be said about SA in non-human animal species.
 野生型のHSA又はHSA-A320Tのアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はHSAのアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型又はHSA-A320TのHSAのN末端又はC末端にアミノ酸残基を1個付加させたHSAの変異体,野生型又はHSA-A320TのHSAのN末端又はC末端にアミノ酸残基を2個付加させたHSAの変異体等も,HSAである。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型又はHSA-A320TのHSAのアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型又はHSA-A320TのHSAのアミノ酸配列に加えることもできる。ヒト以外の動物種のSAについても同様のことがいえる。 When amino acid residues are added to the amino acid sequence of wild-type HSA or HSA-A320T, one or more amino acid residues are added to the amino acid sequence of HSA or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. HSA mutants in which one amino acid residue is added to the N-terminus or C-terminus of wild-type or HSA-A320T HSA, and HSA mutants in which two amino acid residues are added to the N-terminus or C-terminus of wild-type or HSA-A320T HSA, etc. are also HSA. In addition, mutations that combine the addition of these amino acid residues and the above-mentioned substitutions can also be added to the amino acid sequence of wild-type or HSA-A320T HSA, and mutations that combine the addition of these amino acid residues and the above-mentioned deletions can also be added to the amino acid sequence of wild-type or HSA-A320T HSA. The same can be said for SA of animal species other than humans.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型又はHSA-A320TのHSAのアミノ酸配列に加えることもできる。例えば,配列番号3で示される野生型のHSAのアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもHSAであり,配列番号3で示される野生型のHSAのアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもHSAであり,配列番号3で示される野生型のHSAのアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもHSAであり,配列番号3で示される野生型のHSAのアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもHSAであり,配列番号3で示される野生型のHSAのアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもHSAである。ヒト以外の動物種のSAについても同様のことがいえる。 Furthermore, a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of wild-type or HSA-A320T HSA. For example, HSA can be obtained by deleting 1 to 10 amino acid residues from the amino acid sequence of wild-type HSA shown in SEQ ID NO:3, substituting 1 to 10 amino acid residues with other amino acid residues, and adding 1 to 10 amino acid residues, and HSA can be obtained by deleting 1 to 5 amino acid residues from the amino acid sequence of wild-type HSA shown in SEQ ID NO:3, substituting 1 to 5 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues, and HSA can be obtained by deleting 1 to 3 amino acid residues from the amino acid sequence of wild-type HSA shown in SEQ ID NO:3, HSA is also one in which 1 to 3 amino acid residues have been replaced with other amino acid residues and 1 to 3 amino acid residues have been added, HSA is also one in which 1 or 2 amino acid residues have been deleted from the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3, 1 or 2 amino acid residues have been replaced with other amino acid residues, and 1 or 2 amino acid residues have been added, and HSA is also one in which 1 amino acid residue has been deleted from the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3, 1 amino acid residue has been replaced with other amino acid residue, and 1 amino acid residue has been added. The same can be said about SA of animal species other than humans.
 HSA変異体における通常の野生型HSAと比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両HSAのアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種のSAについても同様のことがいえる。 The location and type (deletion, substitution, and addition) of each mutation in an HSA mutant compared to normal wild-type HSA can be easily confirmed by aligning the amino acid sequences of both HSAs. The same can be said for SA of animal species other than humans.
 HSA変異体のアミノ酸配列は,配列番号3で示される通常の野生型HSAのアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。ヒト以外の動物種のSAについても同様のことがいえる。 The amino acid sequence of the HSA mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type HSA shown in SEQ ID NO: 3. The same is true for SA of animal species other than humans.
 なお,本発明の一実施形態におけるHSAは,これを野生型ヒトリソソーム酵素と結合させた融合蛋白質としてCHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型ヒトリソソーム酵素を同様の手法により組換え蛋白質として発現させたときと比較して,少なくとも1種類の野生型ヒトリソソーム酵素について,培養上清中のヒトリソソーム酵素としての発現量が,濃度あるいは酵素活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍等となるようにすることができるものである。 In one embodiment of the present invention, when HSA is combined with a wild-type human lysosomal enzyme to form a fusion protein and expressed as a recombinant protein using host cells such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of at least one type of wild-type human lysosomal enzyme as a human lysosomal enzyme in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, etc., compared to when the wild-type human lysosomal enzyme is expressed as a recombinant protein by a similar method.
 また,本発明の一実施形態におけるHSAは,これを野生型hGALCと結合させた融合蛋白質としてCHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型hGALCを同様の手法により組換え蛋白質として発現させたときと比較して,培養上清中のhGALCとしての発現量が,濃度あるいは酵素活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍等となるようにすることができるものである。 In one embodiment of the present invention, when HSA is combined with wild-type hGALC to form a fusion protein and expressed as a recombinant protein using host cells such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hGALC expressed in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, compared to when wild-type hGALC is expressed as a recombinant protein by a similar method.
 また,本発明の一実施形態におけるHSAは,これを野生型hGBAと結合させた融合蛋白質としてCHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型hGBAを同様の手法により組換え蛋白質として発現させたときと比較して,培養上清中のhGBAとしての発現量が,濃度あるいは酵素活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍等となるようにすることができるものである。 In one embodiment of the present invention, when HSA is combined with wild-type hGBA to form a fusion protein and expressed as a recombinant protein using host cells such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hGBA expressed in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, etc., compared to when wild-type hGBA is expressed as a recombinant protein by a similar method.
 本発明において「保存的アミノ酸置換」というときは,アミノ酸のそれらの側鎖及び化学的性質で関連性のあるアミノ酸ファミリー内でアミノ酸を置換することをいう。このようなアミノ酸ファミリー内での置換は,元の蛋白質の機能に大きな変化をもたらさないことが予測される。かかるアミノ酸ファミリーとしては,例えば以下の(1)~(12)で示されるものがある:
(1)酸性アミノ酸であるアスパラギン酸とグルタミン酸,
(2)塩基性アミノ酸であるヒスチジン,リシン,及びアルギニン,
(3)芳香族アミン酸であるフェニルアラニン,チロシン,及びトリプトファン,
(4)水酸基を有するアミノ酸(ヒドロキシアミノ酸)であるセリンとトレオニン,
(5)疎水性アミノ酸であるメチオニン,アラニン,バリン,ロイシン,及びイソロイシン,
(6)中性の親水性アミノ酸であるシステイン,セリン,トレオニン,アスパラギン,及びグルタミン,
(7)ペプチド鎖の配向に影響するアミノ酸であるグリシンとプロリン,
(8)アミド型アミノ酸(極性アミノ酸)であるアスパラギンとグルタミン,
(9)脂肪族アミノ酸である,アラニン,ロイシン,イソロイシン,及びバリン,
(10)側鎖の小さいアミノ酸であるアラニン,グリシン,セリン,及びトレオニン,
(11)側鎖の特に小さいアミノ酸であるアラニンとグリシン,
(12)分岐鎖を有するアミノ酸であるバリン,ロイシン,及びイソロイシン,
(13)側鎖が水酸化反応され得るアミノ酸であるプロリンとセリン。
 ヒト以外の動物種のリソソーム酵素,GALC,GBA,SAについても同様のことがいえる。
In the present invention, the term "conservative amino acid substitution" refers to a substitution of an amino acid within a family of amino acids that are related in their side chains and chemical properties. Substitutions within such an amino acid family are not expected to significantly alter the function of the original protein. Examples of such amino acid families include the following (1) to (12):
(1) The acidic amino acids aspartic acid and glutamic acid,
(2) the basic amino acids histidine, lysine, and arginine;
(3) the aromatic amino acids phenylalanine, tyrosine, and tryptophan;
(4) Serine and threonine, which are amino acids having a hydroxyl group (hydroxyamino acids),
(5) the hydrophobic amino acids methionine, alanine, valine, leucine, and isoleucine,
(6) The neutral hydrophilic amino acids cysteine, serine, threonine, asparagine, and glutamine,
(7) Glycine and proline, which are amino acids that affect the orientation of peptide chains.
(8) Asparagine and glutamine, which are amide amino acids (polar amino acids),
(9) The aliphatic amino acids alanine, leucine, isoleucine, and valine,
(10) Amino acids with small side chains: alanine, glycine, serine, and threonine;
(11) Alanine and glycine, which are amino acids with particularly small side chains.
(12) The branched chain amino acids valine, leucine, and isoleucine,
(13) Proline and serine, which are amino acids whose side chains can be hydroxylated.
The same can be said for the lysosomal enzymes GALC, GBA, and SA of non-human animal species.
 野生型ヒトリソソーム酵素及びHSAのアミノ酸配列中のアミノ酸の他のアミノ酸への置換は,好ましくは保存的アミノ酸置換である。ヒト以外の動物種のリソソーム酵素及びSAについても同様である。また,特に,上記の保存的アミノ酸置換は,hGALC及びhGBAの野生型に適用される。 The substitution of an amino acid in the amino acid sequence of wild-type human lysosomal enzymes and HSA with another amino acid is preferably a conservative amino acid substitution. The same applies to lysosomal enzymes and SA of animal species other than humans. In particular, the above conservative amino acid substitutions apply to the wild-type forms of hGALC and hGBA.
 上記の野生型又は変異型のヒトリソソーム酵素,例えばhGALC又はhGBAであって,これを構成するアミノ酸が糖鎖により修飾されたものもヒトリソソーム酵素である。また,上記の野生型又は変異型のヒトリソソーム酵素であって,これを構成するアミノ酸がリン酸により修飾されたものもヒトリソソーム酵素である。また,糖鎖及びリン酸以外のものにより修飾されたものもヒトリソソーム酵素である。また,上記の野生型又は変異型のヒトリソソーム酵素であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもヒトリソソーム酵素である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。また,ヒト以外の動物種のリソソーム酵素,GALC,及びGBAについても同様のことがいえる。  The above wild-type or mutant human lysosomal enzymes, such as hGALC or hGBA, whose constituent amino acids are modified with sugar chains are also human lysosomal enzymes. The above wild-type or mutant human lysosomal enzymes whose constituent amino acids are modified with phosphate are also human lysosomal enzymes. Those modified with something other than sugar chains and phosphate are also human lysosomal enzymes. The above wild-type or mutant human lysosomal enzymes whose constituent amino acid side chains have been converted by substitution reactions or the like are also human lysosomal enzymes. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for the lysosomal enzymes, GALC, and GBA of animal species other than humans.
 つまり,糖鎖により修飾されたヒトリソソーム酵素,例えばhGALC又はhGBAは,元のアミノ酸配列を有するヒトリソソーム酵素に含まれるものとする。また,リン酸により修飾されたヒトリソソーム酵素は,元のアミノ酸配列を有するヒトリソソーム酵素に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するヒトリソソーム酵素に含まれるものとする。また,ヒトリソソーム酵素を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するヒトリソソーム酵素に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のリソソーム酵素,GALC,及びGBAについても同様のことがいえる。 In other words, human lysosomal enzymes modified with sugar chains, such as hGALC or hGBA, are included in human lysosomal enzymes with the original amino acid sequence. Human lysosomal enzymes modified with phosphate are included in human lysosomal enzymes with the original amino acid sequence. Those modified with things other than sugar chains and phosphate are also included in human lysosomal enzymes with the original amino acid sequence. Human lysosomal enzymes in which the side chains of the amino acids constituting the human lysosomal enzymes have been converted by substitution reactions or the like are also included in human lysosomal enzymes with the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for lysosomal enzymes of animal species other than humans, GALC, and GBA.
 上記の野生型又は変異型のHSAであって,これを構成するアミノ酸が糖鎖により修飾されたものもHSAである。また,上記の野生型又は変異型のHSAであって,これを構成するアミノ酸がリン酸により修飾されたものもHSAである。また,糖鎖及びリン酸以外のものにより修飾されたものもHSAである。また,上記の野生型又は変異型のHSAであって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSAである。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAについても同様のことがいえる。  The above wild-type or mutant HSA in which the constituent amino acids are modified with sugar chains is also HSA. The above wild-type or mutant HSA in which the constituent amino acids are modified with phosphate is also HSA. HSA modified with something other than sugar chains and phosphate is also HSA. The above wild-type or mutant HSA in which the side chains of the constituent amino acids have been converted by substitution reactions or the like is also HSA. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for SA of animal species other than humans.
 つまり,糖鎖により修飾されたHSAは,元のアミノ酸配列を有するHSAに含まれるものとする。また,リン酸により修飾されたHSAは,元のアミノ酸配列を有するHSAに含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSAに含まれるものとする。また,HSAを構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSAに含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAについても同様のことがいえる。 In other words, HSA modified with sugar chains is included in HSA with the original amino acid sequence. HSA modified with phosphate is included in HSA with the original amino acid sequence. HSA modified with things other than sugar chains and phosphate is also included in HSA with the original amino acid sequence. HSA with the original amino acid sequence also includes HSA with the original amino acid sequence when the side chains of the amino acids that make up HSA have been changed by substitution reactions, etc. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for SA of animal species other than humans.
 本発明は,その一実施形態において,野生型又は変異型のヒトリソソーム酵素のアミノ酸配列を含むポリペプチドと,野生型又は変異型のSAのアミノ酸配列を含むポリペプチドとを結合させた,融合蛋白質に関する。ここで,「ポリペプチドを結合させる」というときは,直接又はリンカーを介して間接的に,共有結合により異なるポリペプチドを結合させることをいう。ここで,SAは好ましくはHSAである。また,ヒトリソソーム酵素は,例えばhGALC,又はhGBAである。 In one embodiment, the present invention relates to a fusion protein in which a polypeptide containing the amino acid sequence of a wild-type or mutant human lysosomal enzyme is bound to a polypeptide containing the amino acid sequence of a wild-type or mutant SA. Here, the term "binding polypeptides" refers to binding different polypeptides by covalent bond, either directly or indirectly via a linker. Here, the SA is preferably HSA. The human lysosomal enzyme is, for example, hGALC or hGBA.
 2つの異なるポリペプチドを結合させる方法としては,例えば,一方のポリペプチドをコードする遺伝子の下流に,インフレームで他方のポリペプチドをコードする遺伝子を結合させたDNA断片を組み込んだ発現ベクターを作製し,この発現ベクターを用いて形質転換させた宿主細胞を培養することにより,組換え蛋白質として発現させる方法が一般的である。得られた組換え蛋白質は,2つのポリペプチドが,直接又は別のアミノ酸配列を介してペプチド結合した,一本鎖ポリペプチドである。 A common method for linking two different polypeptides is, for example, to create an expression vector incorporating a DNA fragment in which a gene encoding one polypeptide is linked in-frame downstream of a gene encoding the other polypeptide, and then to express the polypeptide as a recombinant protein by culturing a host cell transformed with this expression vector. The resulting recombinant protein is a single-chain polypeptide in which the two polypeptides are peptide-linked, either directly or via different amino acid sequences.
 本発明の一実施形態において,「SA-ヒトリソソーム酵素融合蛋白質」又は「SA-ヒトリソソーム酵素」の語は,SAのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒトリソソーム酵素のアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒトリソソーム酵素としての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは霊長類,マウス,ウシ等の哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。SA-ヒトリソソーム酵素融合蛋白質において,ヒトリソソーム酵素がヒトリソソーム酵素としての機能を有するというときは,ヒトリソソーム酵素が,通常の野生型のヒトリソソーム酵素の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでSA-ヒトリソソーム酵素融合蛋白質におけるヒトリソソーム酵素の比活性は,当該融合蛋白質の単位質量当たりのヒトリソソーム酵素の酵素活性(μM/時間/mg蛋白質)に(当該融合蛋白質の分子量/当該融合蛋白質中のヒトリソソーム酵素に相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "SA-human lysosomal enzyme fusion protein" or "SA-human lysosomal enzyme" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of a human lysosomal enzyme is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and which has the function of a human lysosomal enzyme. Here, there is no particular limitation on the animal species of the SA, but it is preferably a mammalian SA such as a primate, mouse, or cow, more preferably a primate SA, and even more preferably HSA. When it is said that the human lysosomal enzyme in the SA-human lysosomal enzyme fusion protein has the function of a human lysosomal enzyme, it means that the human lysosomal enzyme preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of a normal wild-type human lysosomal enzyme is taken as 100%. Here, the specific activity of the human lysosomal enzyme in the SA-human lysosomal enzyme fusion protein is calculated by multiplying the enzymatic activity of the human lysosomal enzyme per unit mass of the fusion protein (μM/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to the human lysosomal enzyme).
 本発明の一実施形態において,「HSA-ヒトリソソーム酵素融合蛋白質」又は「HSA-ヒトリソソーム酵素」の語は,HSAのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒトリソソーム酵素のアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒトリソソーム酵素としての機能を有するものを示す。ここでHSA-ヒトリソソーム酵素融合蛋白質がヒトリソソーム酵素としての機能を有するというときは,上記のSA-ヒトリソソーム酵素融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "HSA-human lysosomal enzyme fusion protein" or "HSA-human lysosomal enzyme" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of a human lysosomal enzyme is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of a human lysosomal enzyme. When it is said here that the HSA-human lysosomal enzyme fusion protein has the function of a human lysosomal enzyme, the definition of the SA-human lysosomal enzyme fusion protein described above can be applied.
 SA-ヒトリソソーム酵素融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。HSA-ヒトリソソーム酵素融合蛋白質においても同様である。 In the SA-human lysosomal enzyme fusion protein, it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this. The same applies to the HSA-human lysosomal enzyme fusion protein.
 本発明の一実施形態において,ヒト以外の動物種のSAのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のリソソーム酵素のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒトリソソーム酵素としての機能を有するものは,「(ヒト以外の動物種)SA-(ヒト以外の動物種)リソソーム酵素融合蛋白質」と表記される。例えば,マウスSAとマウスリソソーム酵素の融合蛋白質は,「MSA-マウスリソソーム酵素融合蛋白質」又は「MSA-マウスリソソーム酵素」と表記される。これら融合蛋白質についても,リソソーム酵素としての機能を有するというときは,上記のSA-ヒトリソソーム酵素融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, a fusion protein having an amino acid sequence in which the N-terminus of a lysosomal enzyme of a non-human animal species is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of SA of a non-human animal species, and which has the function of a human lysosomal enzyme, is referred to as a "(non-human animal species) SA-(non-human animal species) lysosomal enzyme fusion protein." For example, a fusion protein of mouse SA and mouse lysosomal enzyme is referred to as an "MSA-mouse lysosomal enzyme fusion protein" or "MSA-mouse lysosomal enzyme." When these fusion proteins are said to have the function of a lysosomal enzyme, the definition of SA-human lysosomal enzyme fusion protein described above can be applied.
 野生型HSAと野生型ヒトリソソーム酵素との融合蛋白質であるHSA-ヒトリソソーム酵素融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えヒトリソソーム酵素部分には変異を加えないことも,HSA部分には変異を加えずにヒトリソソーム酵素部分にのみ変異を加えることも,また,HSA部分とヒトリソソーム酵素部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。ヒトリソソーム酵素部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型ヒトリソソーム酵素に変異が加えられたヒトリソソーム酵素のアミノ酸配列となる。HSA部分とヒトリソソーム酵素部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,ヒトリソソーム酵素部分のアミノ酸配列は上述した野生型ヒトリソソーム酵素に変異が加えられたヒトリソソーム酵素のアミノ酸配列となる。ヒト以外の動物種の野生型SA(野生型MSAを含む)と野生型ヒトリソソーム酵素との融合蛋白質についても同様のことがいえる。 When mutations are added to an HSA-human lysosomal enzyme fusion protein, which is a fusion protein of wild-type HSA and wild-type human lysosomal enzyme, mutations can be added only to the HSA portion and not to the human lysosomal enzyme portion, mutations can be added only to the human lysosomal enzyme portion without mutations to the HSA portion, or mutations can be added to both the HSA portion and the human lysosomal enzyme portion. When mutations are added only to the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are added only to the human lysosomal enzyme portion, the amino acid sequence of the portion is the amino acid sequence of human lysosomal enzyme obtained by adding a mutation to the wild-type human lysosomal enzyme described above. When mutations are added to both the HSA portion and the human lysosomal enzyme portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the human lysosomal enzyme portion is the amino acid sequence of human lysosomal enzyme obtained by adding a mutation to the wild-type human lysosomal enzyme described above. The same can be said about fusion proteins between wild-type SA (including wild-type MSA) of non-human animal species and wild-type human lysosomal enzymes.
 HSA-ヒトリソソーム酵素融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもHSA-ヒトリソソーム酵素融合蛋白質である。また,HSA-ヒトリソソーム酵素融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもHSA-ヒトリソソーム酵素融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもHSA-ヒトリソソーム酵素融合蛋白質である。また,HSA-ヒトリソソーム酵素融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSA-ヒトリソソーム酵素融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとヒトリソソーム酵素との融合蛋白質(SA-ヒトリソソーム酵素),及びヒト以外の動物種のSAとヒト以外の動物種のリソソーム酵素との融合蛋白質,例えばMSA-マウスリソソーム酵素融合蛋白質についても同様のことがいえる。  HSA-human lysosomal enzyme fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-human lysosomal enzyme fusion proteins. HSA-human lysosomal enzyme fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-human lysosomal enzyme fusion proteins. HSA-human lysosomal enzyme fusion proteins modified with anything other than sugar chains and phosphate are also HSA-human lysosomal enzyme fusion proteins. HSA-human lysosomal enzyme fusion proteins in which the side chains of the amino acids constituting the protein are converted by substitution reactions or the like are also HSA-human lysosomal enzyme fusion proteins. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of SA of a non-human animal species and a human lysosomal enzyme (SA-human lysosomal enzyme), and fusion proteins of SA of a non-human animal species and a lysosomal enzyme of a non-human animal species, such as MSA-mouse lysosomal enzyme fusion proteins.
 つまり,糖鎖により修飾されたHSA-ヒトリソソーム酵素融合蛋白質は,元のアミノ酸配列を有するHSA-ヒトリソソーム酵素融合蛋白質に含まれるものとする。また,リン酸により修飾されたHSA-ヒトリソソーム酵素融合蛋白質は,元のアミノ酸配列を有するHSA-ヒトリソソーム酵素融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSA-ヒトリソソーム酵素融合蛋白質に含まれるものとする。また,HSA-ヒトリソソーム酵素融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSA-ヒトリソソーム酵素融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとヒトリソソーム酵素との融合蛋白質(SA-ヒトリソソーム酵素),及びヒト以外の動物種のSAとヒト以外の動物種のリソソーム酵素との融合蛋白質,例えばMSA-マウスリソソーム酵素融合蛋白質についても同様のことがいえる。 In other words, HSA-human lysosomal enzyme fusion proteins modified with sugar chains are included in HSA-human lysosomal enzyme fusion proteins having the original amino acid sequence. Also, HSA-human lysosomal enzyme fusion proteins modified with phosphate are included in HSA-human lysosomal enzyme fusion proteins having the original amino acid sequence. Also, those modified with something other than sugar chains and phosphate are included in HSA-human lysosomal enzyme fusion proteins having the original amino acid sequence. Also, those in which the side chains of the amino acids constituting the HSA-human lysosomal enzyme fusion proteins have been converted by substitution reactions or the like are included in HSA-human lysosomal enzyme fusion proteins having the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of SA of animal species other than human and human lysosomal enzyme (SA-human lysosomal enzyme), and fusion proteins of SA of animal species other than human and lysosomal enzyme of animal species other than human, such as MSA-mouse lysosomal enzyme fusion protein.
 また,HSA-ヒトリソソーム酵素融合蛋白質であって,これを構成するヒトリソソーム酵素が,ヒトリソソーム酵素の前駆体であるものもHSA-ヒトリソソーム酵素融合蛋白質である。ここで前駆体というときは,HSA-ヒトリソソーム酵素融合蛋白質として生合成されたものであって,当該生合成後に,ヒトリソソーム酵素として機能する部分が,製造過程において又は生体に投与したときに当該生体内において,当該融合蛋白質から切り離されて,単独でもヒトリソソーム酵素としての機能を発揮することができるタイプのものをいう。この場合,HSA-ヒトリソソーム酵素融合蛋白質は生合成された後に,加水分解酵素等により,特定の箇所で切断され,HSAを含む部分と成熟型のヒトリソソーム酵素を含む部分が分離することがある。この場合,得られるヒトリソソーム酵素はHSAとの融合蛋白質ではないが,当該ヒトリソソーム酵素が製造される過程で,一旦,HSA-ヒトリソソーム酵素融合蛋白質が合成されることになる。従って,かかる方法によりヒトリソソーム酵素を製造する場合,当該製造方法は,HSA-ヒトリソソーム酵素融合蛋白質の製造方法に含まれる。ヒト以外の動物種のSAとヒトリソソーム酵素との融合蛋白質(SA-ヒトリソソーム酵素),及びヒト以外の動物種のSAとヒト以外の動物種のリソソーム酵素との融合蛋白質,例えばMSA-マウスリソソーム酵素融合蛋白質についても同様のことがいえる。  Also, HSA-human lysosomal enzyme fusion proteins, in which the human lysosomal enzyme that constitutes it is a precursor of human lysosomal enzyme, are also HSA-human lysosomal enzyme fusion proteins. Here, the term "precursor" refers to a type that is biosynthesized as an HSA-human lysosomal enzyme fusion protein, and after the biosynthesis, the part that functions as a human lysosomal enzyme is separated from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as a human lysosomal enzyme by itself. In this case, after the HSA-human lysosomal enzyme fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the part containing HSA and the part containing the mature human lysosomal enzyme may be separated. In this case, the obtained human lysosomal enzyme is not a fusion protein with HSA, but the HSA-human lysosomal enzyme fusion protein is synthesized once during the process of manufacturing the human lysosomal enzyme. Therefore, when human lysosomal enzyme is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-human lysosomal enzyme fusion protein. The same can be said for fusion proteins of SA of non-human animal species and human lysosomal enzyme (SA-human lysosomal enzyme), and fusion proteins of SA of non-human animal species and lysosomal enzyme of non-human animal species, such as MSA-mouse lysosomal enzyme fusion protein.
 本発明の一実施形態において,「SA-hGALC融合蛋白質」又は「SA-hGALC」の語は,SAのアミノ酸配列のC末端に,直接又はリンカーを介して,hGALCのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hGALCとしての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。SA-hGALC融合蛋白質において,hGALCがhGALCとしての機能を有するというときは,hGALCが,通常の野生型のhGALCの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでSA-hGALC融合蛋白質におけるhGALCの比活性は,当該融合蛋白質の単位質量当たりのhGALCの酵素活性(μM/時間/mg蛋白質)に(当該融合蛋白質の分子量/当該融合蛋白質中のhGALCに相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "SA-hGALC fusion protein" or "SA-hGALC" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hGALC is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of SA, and which has the function of hGALC. Here, there is no particular limitation on the animal species of the SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA. When it is said that hGALC in the SA-hGALC fusion protein has the function of hGALC, it means that hGALC preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of normal wild-type hGALC is taken as 100%. Here, the specific activity of hGALC in the SA-hGALC fusion protein is calculated by multiplying the enzyme activity of hGALC per unit mass of the fusion protein (μM/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hGALC).
 本発明の一実施形態において,「HSA-hGALC融合蛋白質」又は「HSA-hGALC」の語は,HSAのアミノ酸配列のC末端に,直接又はリンカーを介して,hGALCのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hGALCとしての機能を有するものを示す。ここでHSA-hGALC融合蛋白質がhGALCとしての機能を有するというときは,上記のSA-hGALC融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "HSA-hGALC fusion protein" or "HSA-hGALC" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hGALC is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hGALC. When it is said here that the HSA-hGALC fusion protein has the function of hGALC, the definition of the SA-hGALC fusion protein above can be applied.
 本発明の一実施形態において,ヒト以外の動物種のSAのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のGALCのN末端が結合されているアミノ酸配列を有する融合蛋白質であって,GALCとしての機能を有するものは,「(ヒト以外の動物種)SA-(ヒト以外の動物種)GALC融合蛋白質」と表記される。例えば,マウスSAとマウスGALCの融合蛋白質は,「MSA-マウスGALC融合蛋白質」又は「MSA-マウスGALC」と表記される。これら融合蛋白質についても,GALCとしての機能を有するというときは,上記のSA-hGALC融合蛋白質における定義を適用することができる。また,これら融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a fusion protein having an amino acid sequence in which the N-terminus of GALC of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of SA of a non-human animal species, and which has the function of GALC, is designated as a "(non-human animal species) SA-(non-human animal species) GALC fusion protein." For example, a fusion protein of mouse SA and mouse GALC is designated as an "MSA-mouse GALC fusion protein" or "MSA-mouse GALC." When these fusion proteins are said to have the function of GALC, the definition of the SA-hGALC fusion protein described above can be applied. In addition, in these fusion proteins, it is preferable that the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited thereto.
 本発明の一実施形態において好ましいHSA-hGALC融合蛋白質は,例えば配列番号5で示されるアミノ酸配列を有するものである。配列番号5で示されるHSA-hGALC融合蛋白質は,野生型HSAのC末端にリンカー配列Gly-Serを介して野生型hGALCが結合したものである。配列番号5で示されるHSA-hGALC融合蛋白質は,例えば,配列番号6で示される塩基配列を有する遺伝子にコードされる。また,配列番号5で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hGALCとしての機能を有するものである限りHSA-hGALC融合蛋白質に含まれる。HSA-hGALC融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred HSA-hGALC fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 5. The HSA-hGALC fusion protein shown in SEQ ID NO: 5 is obtained by linking wild-type hGALC to the C-terminus of wild-type HSA via the linker sequence Gly-Ser. The HSA-hGALC fusion protein shown in SEQ ID NO: 5 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 6. In addition, HSA-hGALC fusion proteins include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 5 have been replaced with other amino acid residues, deleted, or mutated by addition, so long as they have the function of hGALC. It is preferable that the HSA-hGALC fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 本発明の一実施形態において,「MSA-mGALC融合蛋白質」又は「MSA-mGALC」の語は,MSAのアミノ酸配列のC末端に,直接又はリンカーを介して,mGALCのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,mGALCとしての機能を有するものを示す。好ましいMSA-mGALC融合蛋白質は,例えば配列番号18で示されるアミノ酸配列を有するものである。配列番号18で示されるMSA-mGALC融合蛋白質は,野生型MSAのC末端にリンカー配列Gly-Serを介して野生型mGALCが結合したものである。配列番号18で示されるMSA-mGALC融合蛋白質は,例えば,配列番号19で示される塩基配列を有する遺伝子にコードされる。また,配列番号18で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,mGALCとしての機能を有するものである限りMSA-mGALC融合蛋白質に含まれる。MSA-mGALC融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のマウス血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, the term "MSA-mGALC fusion protein" or "MSA-mGALC" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of mGALC is linked, directly or via a linker, to the C-terminus of the amino acid sequence of MSA, and which has the function of mGALC. A preferred MSA-mGALC fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 18. The MSA-mGALC fusion protein shown in SEQ ID NO: 18 is a fusion protein in which wild-type mGALC is linked to the C-terminus of wild-type MSA via the linker sequence Gly-Ser. The MSA-mGALC fusion protein shown in SEQ ID NO: 18 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 19. In addition, MSA-mGALC fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 18 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of mGALC. It is preferable that the MSA-mGALC fusion protein has the functions of mouse serum albumin, such as binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
 野生型HSAと野生型hGALCとの融合蛋白質であるHSA-hGALC融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhGALC部分には変異を加えないことも,HSA部分には変異を加えずにhGALC部分にのみ変異を加えることも,また,HSA部分とhGALC部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hGALC部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hGALCに変異が加えられたhGALCのアミノ酸配列となる。HSA部分とhGALC部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hGALC部分のアミノ酸配列は上述した野生型hGALCに変異が加えられたhGALCのアミノ酸配列となる。配列番号5で示されるアミノ酸配列を有するHSA-hGALC融合蛋白質についても同様である。また,配列番号18で示されるアミノ酸配列を有するものを含む,MSA-mGALC融合蛋白質についても同様のことがいえる。配列番号18で示されるアミノ酸配列を有するMSA-mGALC融合蛋白質は,例えば配列番号19で示される塩基配列を有する遺伝子にコードされる。ヒト以外の動物種の野生型SA(野生型MSAを含む)と野生型hGALCとの融合蛋白質についても同様のことがいえる。 When mutations are introduced into the HSA-hGALC fusion protein, which is a fusion protein of wild-type HSA and wild-type hGALC, mutations can be introduced only in the HSA portion and not in the hGALC portion, mutations can be introduced only in the hGALC portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hGALC portion. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are introduced only in the hGALC portion, the amino acid sequence of the portion is the amino acid sequence of hGALC obtained by adding a mutation to the wild-type hGALC described above. When mutations are introduced in both the HSA portion and the hGALC portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hGALC portion is the amino acid sequence of hGALC obtained by adding a mutation to the wild-type hGALC described above. The same applies to the HSA-hGALC fusion protein having the amino acid sequence shown in SEQ ID NO:5. The same can be said about MSA-mGALC fusion proteins, including those having the amino acid sequence shown in SEQ ID NO: 18. The MSA-mGALC fusion protein having the amino acid sequence shown in SEQ ID NO: 18 is encoded by a gene having the base sequence shown in SEQ ID NO: 19, for example. The same can be said about fusion proteins of wild-type SA (including wild-type MSA) of animal species other than humans and wild-type hGALC.
 配列番号5で示されるアミノ酸配列を有するHSA-hGALC融合蛋白質に変異を加える場合について,以下例示する。配列番号5で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号5で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。HSA-hGALC融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hGALC部分にのみ加えても,又は両方の部分に加えてもよい。ヒト以外の動物種のSAとhGALCとの融合蛋白質(SA-hGALC),及びヒト以外の動物種のSAとヒト以外の動物種のGALCとの融合蛋白質,例えばMSA-マウスGALCとの融合蛋白質についても同様のことがいえる。 The following is an example of a case where a mutation is made to an HSA-hGALC fusion protein having the amino acid sequence shown in SEQ ID NO:5. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:5 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:5 or to the N-terminus or C-terminus of the amino acid sequence. The HSA-hGALC fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hGALC portion, or to both portions. The same can be said for fusion proteins between SA of a non-human animal species and hGALC (SA-hGALC), and fusion proteins between SA of a non-human animal species and GALC of a non-human animal species, such as the fusion protein MSA-mouse GALC.
 配列番号5で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号5で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:5 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:5.
 HSA-hGALC融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもHSA-hGALC融合蛋白質である。また,HSA-hGALC融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもHSA-hGALC融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもHSA-hGALC融合蛋白質である。また,HSA-hGALC融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSA-hGALC融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhGALCとの融合蛋白質(SA-hGALC),及びヒト以外の動物種のSAとヒト以外の動物種のGALCとの融合蛋白質,例えばMSA-マウスリソソーム酵素との融合蛋白質についても同様のことがいえる。  HSA-hGALC fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hGALC fusion proteins. HSA-hGALC fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hGALC fusion proteins. HSA-hGALC fusion proteins modified with anything other than sugar chains and phosphate are also HSA-hGALC fusion proteins. HSA-hGALC fusion proteins in which the side chains of the amino acids constituting the protein are modified by substitution reactions or the like are also HSA-hGALC fusion proteins. Such modifications include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA of non-human animal species and hGALC (SA-hGALC), and fusion proteins of SA of non-human animal species and GALC of non-human animal species, such as the fusion protein of MSA-mouse lysosomal enzyme.
 つまり,糖鎖により修飾されたHSA-hGALC融合蛋白質は,元のアミノ酸配列を有するHSA-hGALC融合蛋白質に含まれるものとする。また,リン酸により修飾されたHSA-hGALC融合蛋白質は,元のアミノ酸配列を有するHSA-hGALC融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSA-hGALC融合蛋白質に含まれるものとする。また,HSA-hGALC融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSA-hGALC融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhGALCとの融合蛋白質(SA-hGALC),及びヒト以外の動物種のSAとヒト以外の動物種のGALCとの融合蛋白質,例えばMSA-マウスGALCとの融合蛋白質についても同様のことがいえる。 In other words, HSA-hGALC fusion proteins modified with sugar chains are included in HSA-hGALC fusion proteins having the original amino acid sequence. Also, HSA-hGALC fusion proteins modified with phosphate are included in HSA-hGALC fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are included in HSA-hGALC fusion proteins having the original amino acid sequence. Also, those in which the side chains of the amino acids constituting the HSA-hGALC fusion protein have been changed by substitution reactions or the like are included in HSA-hGALC fusion proteins having the original amino acid sequence. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA of a non-human animal species and hGALC (SA-hGALC), and fusion proteins of SA of a non-human animal species and GALC of a non-human animal species, such as the fusion protein MSA-mouse GALC.
 また,HSA-hGALC融合蛋白質であって,これを構成するhGALCが,hGALCの前駆体であるものもHSA-hGALC融合蛋白質である。ここで前駆体というときは,HSA-hGALC融合蛋白質として生合成されたものであって,当該生合成後に,hGALCとして機能する部分が,製造過程において又は生体に投与したときに当該生体内において,当該融合蛋白質から切り離されて,単独でもhGALCとしての機能を発揮することができるタイプのものをいう。この場合,HSA-hGALC融合蛋白質は生合成された後に,加水分解酵素等により,特定の箇所で切断され,HSAを含む部分と成熟型のhGALCを含む部分が分離することがある。この場合,得られるhGALCはHSAとの融合蛋白質ではないが,当該hGALCが合成される過程で,一旦,HSA-hGALC融合蛋白質が合成されることになる。従って,かかる方法によりhGALCを製造する場合,当該製造方法は,HSA-hGALC融合蛋白質の製造方法に含まれる。ヒト以外の動物種のSAとhGALCとの融合蛋白質(SA-hGALC),及びヒト以外の動物種のSAとヒト以外の動物種のGALCとの融合蛋白質,例えばMSA-マウスGALC融合蛋白質についても同様のことがいえる。  Also, HSA-hGALC fusion proteins in which the constituent hGALC is a precursor of hGALC are also HSA-hGALC fusion proteins. The term "precursor" here refers to a type of protein that is biosynthesized as an HSA-hGALC fusion protein, and in which the portion that functions as hGALC after the biosynthesis is cleaved from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hGALC by itself. In this case, after the HSA-hGALC fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the portion containing HSA and the portion containing mature hGALC may be separated. In this case, the obtained hGALC is not a fusion protein with HSA, but the HSA-hGALC fusion protein is synthesized once during the process of synthesizing the hGALC. Therefore, when hGALC is produced by such a method, the production method is included in the method for producing HSA-hGALC fusion protein. The same can be said for fusion proteins of non-human animal species SA and hGALC (SA-hGALC), and fusion proteins of non-human animal species SA and non-human animal species GALC, such as MSA-mouse GALC fusion protein.
 本発明の一実施形態において,「SA-hGBA融合蛋白質」又は「SA-hGBA」の語は,SAのアミノ酸配列のC末端に,直接又はリンカーを介して,hGBAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hGBAとしての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。SA-hGBA融合蛋白質において,hGBAがhGBAとしての機能を有するというときは,hGBAが,通常の野生型のhGBAの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでSA-hGBA融合蛋白質におけるhGBAの比活性は,当該融合蛋白質の単位質量当たりのhGBAの酵素活性(μM/時間/mg蛋白質)に(当該融合蛋白質の分子量/当該融合蛋白質中のhGBAに相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "SA-hGBA fusion protein" or "SA-hGBA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hGBA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of SA, and which has the function of hGBA. Here, there is no particular limitation on the animal species of the SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA. When it is said that hGBA in the SA-hGBA fusion protein has the function of hGBA, it means that hGBA retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hGBA is taken as 100%. Here, the specific activity of hGBA in the SA-hGBA fusion protein is calculated by multiplying the enzyme activity of hGBA per unit mass of the fusion protein (μM/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hGBA).
 本発明の一実施形態において,「HSA-hGBA融合蛋白質」又は「HSA-hGBA」の語は,HSAのアミノ酸配列のC末端に,直接又はリンカーを介して,hGBAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hGBAとしての機能を有するものを示す。ここでHSA-hGBA融合蛋白質がhGALCとしての機能を有するというときは,上記のSA-hGBA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "HSA-hGBA fusion protein" or "HSA-hGBA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hGBA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hGBA. When it is said here that the HSA-hGBA fusion protein has the function of hGALC, the definition of the SA-hGBA fusion protein described above can be applied.
 本発明の一実施形態において,ヒト以外の動物種のSAのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のGBAのN末端が結合されているアミノ酸配列を有する融合蛋白質であって,GBAとしての機能を有するものは,「(ヒト以外の動物種)SA-(ヒト以外の動物種)GBA融合蛋白質」と表記される。例えば,マウスSAとマウスGBAの融合蛋白質は,「MSA-マウスGBA融合蛋白質」又は「MSA-マウスGBA」と表記される。これら融合蛋白質についても,GBAとしての機能を有するというときは,上記のSA-hGBA融合蛋白質における定義を適用することができる。また,これら融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a fusion protein having an amino acid sequence in which the N-terminus of GBA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of SA of a non-human animal species, and which has the function of GBA, is designated as a "(non-human animal species) SA-(non-human animal species) GBA fusion protein." For example, a fusion protein of mouse SA and mouse GBA is designated as an "MSA-mouse GBA fusion protein" or "MSA-mouse GBA." When these fusion proteins are said to have the function of GBA, the definition of the SA-hGBA fusion protein described above can be applied. In addition, in these fusion proteins, it is preferable that the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited thereto.
 本発明の一実施形態において好ましいHSA-hGBA融合蛋白質は,例えば配列番号39で示されるアミノ酸配列を有するものである。配列番号39で示されるHSA-hGBA融合蛋白質は,野生型HSAのC末端に配列番号9で示されるアミノ酸配列を有するリンカー配列を介して野生型hGBAが結合したものである。配列番号39で示されるHSA-hGBA融合蛋白質は,例えば,配列番号40で示される塩基配列を有する遺伝子にコードされる。また,配列番号39で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hGBAとしての機能を有するものである限りHSA-hGBA融合蛋白質に含まれる。HSA-hGBA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred HSA-hGBA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 39. The HSA-hGBA fusion protein shown in SEQ ID NO: 39 is obtained by linking wild-type hGBA to the C-terminus of wild-type HSA via a linker sequence having the amino acid sequence shown in SEQ ID NO: 9. The HSA-hGBA fusion protein shown in SEQ ID NO: 39 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 40. In addition, HSA-hGBA fusion proteins include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 39 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hGBA. It is preferable that the HSA-hGBA fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 本発明の一実施形態において,「MSA-mGBA融合蛋白質」又は「MSA-mGBA」の語は,MSAのアミノ酸配列のC末端に,直接又はリンカーを介して,mGBAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,mGBAとしての機能を有するものを示す。好ましいMSA-mGBA融合蛋白質は,例えば配列番号45で示されるアミノ酸配列を有するものである。配列番号45で示されるMSA-mGBA融合蛋白質は,野生型MSAのC末端に配列番号9で示されるアミノ酸配列を有するリンカー配列を介して野生型mGBAが結合したものである。配列番号45で示されるMSA-mGBA融合蛋白質は,例えば,配列番号46で示される塩基配列を有する遺伝子にコードされる。また,配列番号45で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,mGBAとしての機能を有するものである限りMSA-mGBA融合蛋白質に含まれる。MSA-mGBA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のマウス血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, the term "MSA-mGBA fusion protein" or "MSA-mGBA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of mGBA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of MSA, and which has the function of mGBA. A preferred MSA-mGBA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 45. The MSA-mGBA fusion protein shown in SEQ ID NO: 45 is a fusion protein in which wild-type mGBA is linked to the C-terminus of wild-type MSA via a linker sequence having the amino acid sequence shown in SEQ ID NO: 9. The MSA-mGBA fusion protein shown in SEQ ID NO: 45 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 46. In addition, MSA-mGBA fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 45 are replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of mGBA. It is preferable that the MSA-mGBA fusion protein has mouse serum albumin functions, such as binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
 野生型HSAと野生型hGBAとの融合蛋白質であるHSA-hGBA融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhGBA部分には変異を加えないことも,HSA部分には変異を加えずにhGBA部分にのみ変異を加えることも,また,HSA部分とhGBA部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hGBA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hGBAに変異が加えられたhGBAのアミノ酸配列となる。HSA部分とhGBA部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hGBA部分のアミノ酸配列は上述した野生型hGBAに変異が加えられたhGBAのアミノ酸配列となる。配列番号39で示されるアミノ酸配列を有するHSA-hGBA融合蛋白質についても同様である。また,配列番号45で示されるアミノ酸配列を有するものを含む,MSA-mGBA融合蛋白質についても同様のことがいえる。配列番号45で示されるアミノ酸配列を有するMSA-mGBA融合蛋白質は,例えば配列番号46で示される塩基配列を有する遺伝子にコードされる。ヒト以外の動物種の野生型SA(野生型MSAを含む)と野生型hGBAとの融合蛋白質についても同様のことがいえる。 When mutations are introduced into an HSA-hGBA fusion protein, which is a fusion protein of wild-type HSA and wild-type hGBA, mutations can be introduced only in the HSA portion and not in the hGBA portion, mutations can be introduced only in the hGBA portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hGBA portion. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above. When mutations are introduced only in the hGBA portion, the amino acid sequence of the portion is the amino acid sequence of hGBA obtained by mutating the wild-type hGBA described above. When mutations are introduced in both the HSA portion and the hGBA portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above, and the amino acid sequence of the hGBA portion is the amino acid sequence of hGBA obtained by mutating the wild-type hGBA described above. The same applies to the HSA-hGBA fusion protein having the amino acid sequence shown in SEQ ID NO: 39. The same can be said about MSA-mGBA fusion proteins, including those having the amino acid sequence shown in SEQ ID NO: 45. The MSA-mGBA fusion protein having the amino acid sequence shown in SEQ ID NO: 45 is encoded by a gene having the base sequence shown in SEQ ID NO: 46, for example. The same can be said about fusion proteins of wild-type SA (including wild-type MSA) of animal species other than humans and wild-type hGBA.
 配列番号39で示されるアミノ酸配列を有するHSA-hGBA融合蛋白質に変異を加える場合について,以下例示する。配列番号39で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号39で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。HSA-hGBA融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hGBA部分にのみ加えても,又は両方の部分に加えてもよい。ヒト以外の動物種のSAとhGBAとの融合蛋白質(SA-hGBA),及びヒト以外の動物種のSAとヒト以外の動物種のGBAとの融合蛋白質,例えばMSA-マウスGBAとの融合蛋白質についても同様のことがいえる。 The following is an example of a case where a mutation is made to an HSA-hGBA fusion protein having the amino acid sequence shown in SEQ ID NO:39. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:39 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:39 or to the N-terminus or C-terminus of the amino acid sequence. The HSA-hGBA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hGBA portion, or to both portions. The same can be said for fusion proteins of SA of a non-human animal species and hGBA (SA-hGBA), and fusion proteins of SA of a non-human animal species and GBA of a non-human animal species, such as the fusion protein MSA-mouse GBA.
 配列番号39で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号39で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:39 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:39.
 HSA-hGBA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもHSA-hGBA融合蛋白質である。また,HSA-hGBA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもHSA-hGBA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもHSA-hGBA融合蛋白質である。また,HSA-hGBA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSA-hGBA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhGBAとの融合蛋白質(SA-hGBA),及びヒト以外の動物種のSAとヒト以外の動物種のGBAとの融合蛋白質,例えばMSA-マウスGBAとの融合蛋白質についても同様のことがいえる。  HSA-hGBA fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hGBA fusion proteins. HSA-hGBA fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hGBA fusion proteins. HSA-hGBA fusion proteins modified with anything other than sugar chains and phosphate are also HSA-hGBA fusion proteins. HSA-hGBA fusion proteins in which the side chains of the amino acids constituting the protein have been converted by substitution reactions or the like are also HSA-hGBA fusion proteins. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hGBA of non-human animal species (SA-hGBA), and fusion proteins of SA and GBA of non-human animal species, such as the fusion protein MSA-mouseGBA.
 つまり,糖鎖により修飾されたHSA-hGBA融合蛋白質は,元のアミノ酸配列を有するHSA-hGBA融合蛋白質に含まれるものとする。また,リン酸により修飾されたHSA-hGBA融合蛋白質は,元のアミノ酸配列を有するHSA-hGBA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSA-hGBA融合蛋白質に含まれるものとする。また,HSA-hGBA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSA-hGBA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhGBAとの融合蛋白質(SA-hGBA),及びヒト以外の動物種のSAとヒト以外の動物種のGBAとの融合蛋白質,例えばMSA-マウスGBAとの融合蛋白質についても同様のことがいえる。 In other words, HSA-hGBA fusion proteins modified with sugar chains are included in HSA-hGBA fusion proteins having the original amino acid sequence. Also, HSA-hGBA fusion proteins modified with phosphate are included in HSA-hGBA fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are included in HSA-hGBA fusion proteins having the original amino acid sequence. Also, those in which the side chains of the amino acids constituting the HSA-hGBA fusion protein have been converted by substitution reactions or the like are included in HSA-hGBA fusion proteins having the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hGBA of non-human animal species (SA-hGBA), and fusion proteins of SA of non-human animal species and GBA of non-human animal species, such as the fusion protein MSA-mouse GBA.
 また,HSA-hGBA融合蛋白質であって,これを構成するhGBAが,hGBAの前駆体であるものもHSA-hGBA融合蛋白質である。ここで前駆体というときは,HSA- hGBA融合蛋白質として生合成されたものであって,当該生合成後に,hGBAとして機能する部分が,製造過程において又は生体に投与したときに当該生体内において,当該融合蛋白質から切り離されて,単独でもhGBAとしての機能を発揮することができるタイプのものをいう。この場合,HSA-hGBA融合蛋白質は生合成された後に,加水分解酵素等により,特定の箇所で切断され,HSAを含む部分と成熟型のhGBAを含む部分が分離することがある。この場合,得られるhGBAはHSAとの融合蛋白質ではないが,当該hGBAが合成される過程で,一旦,HSA-hGBA融合蛋白質が合成されることになる。従って,かかる方法によりhGBAを製造する場合,当該製造方法は,HSA-hGBA融合蛋白質の製造方法に含まれる。ヒト以外の動物種のSAとhGBAとの融合蛋白質(SA-hGBA),及びヒト以外の動物種のSAとヒト以外の動物種のGBAとの融合蛋白質,例えばMSA-マウスGBA融合蛋白質についても同様のことがいえる。 Also, HSA-hGBA fusion proteins in which the hGBA that constitutes them is a precursor of hGBA are also HSA-hGBA fusion proteins. Here, the term "precursor" refers to a type that is biosynthesized as an HSA-hGBA fusion protein, and in which the portion that functions as hGBA after the biosynthesis is separated from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hGBA by itself. In this case, after the HSA-hGBA fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the portion containing HSA and the portion containing mature hGBA may be separated. In this case, the obtained hGBA is not a fusion protein with HSA, but the HSA-hGBA fusion protein is synthesized once during the process of synthesizing the hGBA. Therefore, when hGBA is produced by such a method, the production method is included in the method for producing HSA-hGBA fusion protein. The same can be said for fusion proteins of SA of a non-human animal species and hGBA (SA-hGBA), and fusion proteins of SA of a non-human animal species and GBA of a non-human animal species, such as the MSA-mouse GBA fusion protein.
 本発明の一実施形態において,「ヒトリソソーム酵素-SA融合蛋白質」又は「ヒトリソソーム酵素-SA」の語は,ヒトリソソーム酵素のアミノ酸配列のC末端に,直接又はリンカーを介して,SAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒトリソソーム酵素としての機能を有するものを示す。ヒトリソソーム酵素-SA融合蛋白質において,ヒトリソソーム酵素がヒトリソソーム酵素としての機能を有するというときは,ヒトリソソーム酵素が,通常の野生型のヒトリソソーム酵素の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでヒトリソソーム酵素-SA融合蛋白質におけるヒトリソソーム酵素の比活性は,当該融合蛋白質の単位質量当たりのヒトリソソーム酵素の酵素活性(μM/時間/mg蛋白質)に(当該融合蛋白質の分子量/当該融合蛋白質中のヒトリソソーム酵素に相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "human lysosomal enzyme-SA fusion protein" or "human lysosomal enzyme-SA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of a human lysosomal enzyme, and which has the function of a human lysosomal enzyme. When the human lysosomal enzyme in a human lysosomal enzyme-SA fusion protein is said to have the function of a human lysosomal enzyme, it means that the human lysosomal enzyme preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of a normal wild-type human lysosomal enzyme is taken as 100%. Here, the specific activity of the human lysosomal enzyme in the human lysosomal enzyme-SA fusion protein is calculated by multiplying the enzymatic activity of the human lysosomal enzyme per unit mass of the fusion protein (μM/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to the human lysosomal enzyme).
 本発明の一実施形態において,「ヒトリソソーム酵素-HSA融合蛋白質」又は「ヒトリソソーム酵素-HSA」の語は,ヒトリソソーム酵素のアミノ酸配列のC末端に,直接又はリンカーを介して,HSAのアミノ酸配列のN末端が結合されている,アミノ酸配列を有する融合蛋白質であって,ヒトリソソーム酵素としての機能を有するものを示す。ここでヒトリソソーム酵素-HSA融合蛋白質がヒトリソソーム酵素としての機能を有するというときは,上記のヒトリソソーム酵素-SAにおける定義を適用することができる。 In one embodiment of the present invention, the term "human lysosomal enzyme-HSA fusion protein" or "human lysosomal enzyme-HSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of a human lysosomal enzyme, and which has the function of a human lysosomal enzyme. When it is said that the human lysosomal enzyme-HSA fusion protein has the function of a human lysosomal enzyme, the definition of human lysosomal enzyme-SA above can be applied.
 ヒトリソソーム酵素-SA融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。ヒトリソソーム酵素-HSA融合蛋白質においても同様である。 In the human lysosomal enzyme-SA fusion protein, it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this. The same is true for the human lysosomal enzyme-HSA fusion protein.
 本発明の一実施形態において,ヒト以外の動物種のリソソーム酵素のアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のSAのN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒトリソソーム酵素としての機能を有するものは,「(ヒト以外の動物種)リソソーム酵素-(ヒト以外の動物種)SA融合蛋白質」と表記される。例えば,マウスリソソーム酵素とマウスSAとの融合蛋白質は,「マウスリソソーム酵素-MSA融合蛋白質」又は「マウスリソソーム酵素-MSA」と表記される。これら融合蛋白質についても,リソソーム酵素としての機能を有するというときは,上記のヒトリソソーム酵素-SA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of a lysosomal enzyme of a non-human animal species, and which has the function of a human lysosomal enzyme, is referred to as a "(non-human animal species) lysosomal enzyme-(non-human animal species) SA fusion protein." For example, a fusion protein of mouse lysosomal enzyme and mouse SA is referred to as a "mouse lysosomal enzyme-MSA fusion protein" or "mouse lysosomal enzyme-MSA." When these fusion proteins are said to have the function of a lysosomal enzyme, the definition of the human lysosomal enzyme-SA fusion protein described above can be applied.
 野生型ヒトリソソーム酵素と野生型HSAとの融合蛋白質であるヒトリソソーム酵素-HSA融合蛋白質に変異を加える場合,ヒトリソソーム酵素部分にのみ変異を加えHSA部分には変異を加えないことも,ヒトリソソーム酵素部分には変異を加えずにHSA部分にのみ変異を加えることも,また,ヒトリソソーム酵素部分とHSA部分の何れにも変異を加えることもできる。ヒトリソソーム酵素部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型ヒトリソソーム酵素に変異が加えられたヒトリソソーム酵素のアミノ酸配列となる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。ヒトリソソーム酵素部分とHSA部分の何れにも変異を加える場合にあっては,ヒトリソソーム酵素部分のアミノ酸配列は上述した野生型ヒトリソソーム酵素に変異が加えられたヒトリソソーム酵素のアミノ酸配列となり,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。野生型ヒトリソソーム酵素とヒト以外の動物種の野生型SA(野生型MSAを含む)との融合蛋白質についても同様のことがいえる。 When a mutation is introduced into a human lysosomal enzyme-HSA fusion protein, which is a fusion protein of wild-type human lysosomal enzyme and wild-type HSA, a mutation can be introduced only into the human lysosomal enzyme portion and not into the HSA portion, a mutation can be introduced only into the HSA portion without introducing a mutation into the human lysosomal enzyme portion, or a mutation can be introduced into both the human lysosomal enzyme portion and the HSA portion. When a mutation is introduced only into the human lysosomal enzyme portion, the amino acid sequence of the portion is the amino acid sequence of the human lysosomal enzyme obtained by introducing a mutation into the wild-type human lysosomal enzyme described above. When a mutation is introduced only into the HSA portion, the amino acid sequence of the portion is the amino acid sequence of the HSA obtained by introducing a mutation into the wild-type HSA described above. When a mutation is introduced into both the human lysosomal enzyme portion and the HSA portion, the amino acid sequence of the human lysosomal enzyme portion is the amino acid sequence of the human lysosomal enzyme obtained by introducing a mutation into the wild-type human lysosomal enzyme described above, and the amino acid sequence of the HSA portion is the amino acid sequence of the HSA obtained by introducing a mutation into the wild-type HSA described above. The same can be said about fusion proteins between wild-type human lysosomal enzymes and wild-type SA (including wild-type MSA) of non-human animal species.
 ヒトリソソーム酵素-HSA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもヒトリソソーム酵素-HSA融合蛋白質である。また,ヒトリソソーム酵素-HSA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもヒトリソソーム酵素-HSA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもヒトリソソーム酵素-HSA融合蛋白質である。また,ヒトリソソーム酵素-HSA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもヒトリソソーム酵素-HSA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒトリソソーム酵素とヒト以外の動物種のSAとの融合蛋白質(ヒトリソソーム酵素-SA),及びヒト以外の動物種のリソソーム酵素とヒト以外の動物種のSAとの融合蛋白質,例えばマウスリソソーム酵素-MSAについても同様のことがいえる。  Human lysosomal enzyme-HSA fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also human lysosomal enzyme-HSA fusion proteins. Human lysosomal enzyme-HSA fusion proteins in which the amino acids constituting the protein are modified with phosphate are also human lysosomal enzyme-HSA fusion proteins. Human lysosomal enzyme-HSA fusion proteins modified with anything other than sugar chains and phosphate are also human lysosomal enzyme-HSA fusion proteins. Human lysosomal enzyme-HSA fusion proteins in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like are also human lysosomal enzyme-HSA fusion proteins. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of human lysosomal enzyme and SA of a species of animal other than human (human lysosomal enzyme-SA) and fusion proteins of lysosomal enzyme and SA of a species of animal other than human, such as mouse lysosomal enzyme-MSA.
 つまり,糖鎖により修飾されたヒトリソソーム酵素-HSA融合蛋白質は,元のアミノ酸配列を有するヒトリソソーム酵素-HSA融合蛋白質に含まれるものとする。また,リン酸により修飾されたヒトリソソーム酵素-HSA融合蛋白質は,元のアミノ酸配列を有するヒトリソソーム酵素-HSA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するヒトリソソーム酵素-HSA融合蛋白質に含まれるものとする。また,ヒトリソソーム酵素-HSA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するヒトリソソーム酵素-HSA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒトリソソーム酵素とヒト以外の動物種のSAとの融合蛋白質(ヒトリソソーム酵素-SA),及びヒト以外の動物種のリソソーム酵素とヒト以外の動物種のSAとの融合蛋白質についても同様のことがいえる。 In other words, a human lysosomal enzyme-HSA fusion protein modified with a sugar chain is included in the human lysosomal enzyme-HSA fusion protein having the original amino acid sequence. A human lysosomal enzyme-HSA fusion protein modified with phosphate is included in the human lysosomal enzyme-HSA fusion protein having the original amino acid sequence. A fusion protein modified with something other than sugar chains and phosphate is also included in the human lysosomal enzyme-HSA fusion protein having the original amino acid sequence. A human lysosomal enzyme-HSA fusion protein in which the side chains of the amino acids constituting the human lysosomal enzyme-HSA fusion protein have been converted by a substitution reaction or the like is also included in the human lysosomal enzyme-HSA fusion protein having the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about a fusion protein of a human lysosomal enzyme and SA of an animal species other than human (human lysosomal enzyme-SA), and a fusion protein of a lysosomal enzyme of an animal species other than human and SA of an animal species other than human.
 また,ヒトリソソーム酵素-HSA融合蛋白質であって,これを構成するヒトリソソーム酵素が,ヒトリソソーム酵素の前駆体であるものもヒトリソソーム酵素-HSA融合蛋白質である。 Furthermore, a human lysosomal enzyme-HSA fusion protein in which the human lysosomal enzyme that constitutes it is a precursor of a human lysosomal enzyme is also a human lysosomal enzyme-HSA fusion protein.
 本発明の一実施形態において,「hGALC-SA融合蛋白質」又は「hGALC-SA」の語は,hGALCのアミノ酸配列のC末端に,直接又はリンカーを介して,SAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hGALCとしての機能を有するものを示す。hGALC-SA融合蛋白質において,hGALCがhGALCとしての機能を有するというときは,hGALCが,通常の野生型のhGALCの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでhGALC-SA融合蛋白質におけるhGALCの比活性は,当該融合蛋白質の単位質量当たりのhGALCの酵素活性(μM/時間/mg蛋白質)に(当該融合蛋白質の分子量/当該融合蛋白質中のhGALCに相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "hGALC-SA fusion protein" or "hGALC-SA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hGALC, and which has the function of hGALC. When hGALC in an hGALC-SA fusion protein is said to have the function of hGALC, it means that hGALC retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hGALC is taken as 100%. Here, the specific activity of hGALC in the hGALC-SA fusion protein is calculated by multiplying the enzyme activity of hGALC per unit mass of the fusion protein (μM/hour/mg protein) by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hGALC).
 本発明の一実施形態において,「hGALC-HSA融合蛋白質」又は「hGALC-HSA」の語は,hGALCのアミノ酸配列のC末端に,直接又はリンカーを介して,HSAのアミノ酸配列のN末端が結合されている,アミノ酸配列を有する融合蛋白質であって,hGALCとしての機能を有するものを示す。ここでhGALC-HSA融合蛋白質がhGALCとしての機能を有するというときは,上記のhGALC-SA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "hGALC-HSA fusion protein" or "hGALC-HSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hGALC, and which has the function of hGALC. When it is said here that the hGALC-HSA fusion protein has the function of hGALC, the definition of the hGALC-SA fusion protein described above can be applied.
 本発明の一実施形態において,ヒト以外の動物種のGALCのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のSAのN末端が結合されているアミノ酸配列を有する融合蛋白質であって,GALCとしての機能を有するものは,「(ヒト以外の動物種)GALC-(ヒト以外の動物種)SA融合蛋白質」と表記される。例えば,マウスGALCとマウスSAの融合蛋白質は,「マウスGALC-MSA融合蛋白質」又は「マウスGALC-MSA」と表記される。これら融合蛋白質についても,GALCとしての機能を有するというときは,上記のhGALC-HSA融合蛋白質における定義を適用することができる。また,これら融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of GALC of a non-human animal species, and which has the function of GALC, is designated as a "(non-human animal species) GALC-(non-human animal species) SA fusion protein." For example, a fusion protein of mouse GALC and mouse SA is designated as a "mouse GALC-MSA fusion protein" or "mouse GALC-MSA." When these fusion proteins are said to have the function of GALC, the definition of the hGALC-HSA fusion protein described above can be applied. In addition, in these fusion proteins, it is preferable that the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited thereto.
 本発明の一実施形態において好ましいhGALC-HSA融合蛋白質は,例えば配列番号7で示されるアミノ酸配列を有するものである。配列番号7で示されるhGALC-HSA融合蛋白質は,野生型hGALCのC末端にリンカー配列Gly-Serを介して野生型HSAが結合したものである。配列番号7で示されるhGALC-HSA融合蛋白質は,例えば,配列番号8で示される塩基配列を有する遺伝子にコードされる。また,配列番号7で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hGALCとしての機能を有するものである限りhGALC-HSA融合蛋白質に含まれる。hGALC-HSA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred hGALC-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 7. The hGALC-HSA fusion protein shown in SEQ ID NO: 7 is obtained by linking wild-type HSA to the C-terminus of wild-type hGALC via the linker sequence Gly-Ser. The hGALC-HSA fusion protein shown in SEQ ID NO: 7 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 8. In addition, the hGALC-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 7 have been replaced with other amino acid residues, deleted, or mutated by addition, so long as it has the function of hGALC. The hGALC-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 本発明の一実施形態において,「mGALC-MSA融合蛋白質」又は「mGALC-MSA」の語は,mGALCのアミノ酸配列のC末端に,直接又はリンカーを介して,MSAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,mGALCとしての機能を有するものを示す。好ましいmGALC-MSA融合蛋白質は,例えば配列番号20で示されるアミノ酸配列を有するものである。配列番号20で示されるmGALC-MSA融合蛋白質は,野生型mGALCのC末端にリンカー配列Gly-Serを介して野生型MSAが結合したものである。配列番号20で示されるmGALC-MSA融合蛋白質は,例えば,配列番号21で示される塩基配列を有する遺伝子にコードされる。また,配列番号20で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,mGALCとしての機能を有するものである限りmGALC-MSA融合蛋白質に含まれる。mGALC-MSA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のマウス血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, the term "mGALC-MSA fusion protein" or "mGALC-MSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of MSA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of mGALC, and which has the function of mGALC. A preferred mGALC-MSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 20. The mGALC-MSA fusion protein shown in SEQ ID NO: 20 is a fusion protein in which wild-type MSA is linked to the C-terminus of wild-type mGALC via the linker sequence Gly-Ser. The mGALC-MSA fusion protein shown in SEQ ID NO: 20 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 21. In addition, mGALC-MSA fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 20 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of mGALC. It is preferable that the mGALC-MSA fusion protein has mouse serum albumin functions, such as binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
 野生型hGALCと野生型HSAとの融合蛋白質であるhGALC-HSA融合蛋白質に変異を加える場合,hGALC部分にのみ変異を加えHSA部分には変異を加えないことも,hGALC部分には変異を加えずにHSA部分にのみ変異を加えることも,また,hGALC部分とHSA部分の何れにも変異を加えることもできる。hGALC部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hGALCに変異が加えられたhGALCのアミノ酸配列となる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hGALC部分とHSA部分の何れにも変異を加える場合にあっては,hGALC部分のアミノ酸配列は上述した野生型hGALCに変異が加えられたhGALCのアミノ酸配列となり,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。配列番号7で示されるアミノ酸配列を有するhGALC-HSA融合蛋白質についても同様である。また,配列番号20で示されるアミノ酸配列を有するものを含む,mGALC-MSA融合蛋白質についても同様のことがいえる。配列番号20で示されるアミノ酸配列を有するmGALC-MSA融合蛋白質は,例えば配列番号21で示される塩基配列を有する遺伝子にコードされる。野生型hGALCとヒト以外の動物の野生型SA(野生型MSAを含む)との融合蛋白質についても同様のことがいえる。 When mutations are introduced into the hGALC-HSA fusion protein, which is a fusion protein of wild-type hGALC and wild-type HSA, mutations can be introduced only in the hGALC portion and not in the HSA portion, mutations can be introduced only in the HSA portion without mutations in the hGALC portion, or mutations can be introduced in both the hGALC portion and the HSA portion. When mutations are introduced only in the hGALC portion, the amino acid sequence of the portion is the amino acid sequence of hGALC obtained by mutating the wild-type hGALC described above. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above. When mutations are introduced both in the hGALC portion and the HSA portion, the amino acid sequence of the hGALC portion is the amino acid sequence of hGALC obtained by mutating the wild-type hGALC described above, and the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above. The same applies to the hGALC-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 7. The same can be said about mGALC-MSA fusion proteins, including those having the amino acid sequence shown in SEQ ID NO: 20. The mGALC-MSA fusion protein having the amino acid sequence shown in SEQ ID NO: 20 is encoded by a gene having the base sequence shown in SEQ ID NO: 21, for example. The same can be said about fusion proteins of wild-type hGALC and wild-type SA (including wild-type MSA) of animals other than humans.
 野生型hGALCと野生型HSAとの融合蛋白質であるhGALC-HSA融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhGALC部分には変異を加えないことも,HSA部分には変異を加えずにhGALC部分にのみ変異を加えることも,また,HSA部分とhGALC部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hGALC部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hGALCに変異が加えられたhGALCのアミノ酸配列となる。HSA部分とhGALC部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hGALC部分のアミノ酸配列は上述した野生型hGALCに変異が加えられたhGALCのアミノ酸配列となる。配列番号7で示されるアミノ酸配列を有するhGALC-HSA融合蛋白質についても同様である。また,配列番号20で示されるアミノ酸配列を有するものを含む,mGALC-MSA融合蛋白質についても同様のことがいえる。配列番号20で示されるアミノ酸配列を有するmGALC-MSA融合蛋白質は,例えば配列番号21で示される塩基配列を有する遺伝子にコードされる。野生型hGALCとヒト以外の動物の野生型SA(hGALC-SA)との融合蛋白質についても同様のことがいえる。 When mutations are introduced into the hGALC-HSA fusion protein, which is a fusion protein of wild-type hGALC and wild-type HSA, mutations can be introduced only in the HSA portion and not in the hGALC portion, mutations can be introduced only in the hGALC portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hGALC portion. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are introduced only in the hGALC portion, the amino acid sequence of the portion is the amino acid sequence of hGALC obtained by adding a mutation to the wild-type hGALC described above. When mutations are introduced in both the HSA portion and the hGALC portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hGALC portion is the amino acid sequence of hGALC obtained by adding a mutation to the wild-type hGALC described above. The same applies to the hGALC-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 7. The same can be said about mGALC-MSA fusion proteins, including those having the amino acid sequence shown in SEQ ID NO: 20. The mGALC-MSA fusion protein having the amino acid sequence shown in SEQ ID NO: 20 is encoded by a gene having the base sequence shown in SEQ ID NO: 21, for example. The same can be said about a fusion protein of wild-type hGALC and wild-type SA (hGALC-SA) of an animal other than human.
 配列番号7で示されるアミノ酸配列を有するhGALC-HSA融合蛋白質に変異を加える場合について,以下例示する。配列番号7で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号7で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。hGALC-HSA融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hGALC部分にのみ加えても,又は両方の部分に加えてもよい。ヒト以外の動物種のSAとGALCとの融合蛋白質(SA-GALC),及びヒト以外の動物種のSAとヒト以外の動物種のGALCとの融合蛋白質,例えばMSA-マウスGALCとの融合蛋白質についても同様のことがいえる。 The following is an example of a case where a mutation is made to an hGALC-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:7. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:7 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:7 or to the N-terminus or C-terminus of the amino acid sequence. The hGALC-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hGALC portion, or to both portions. The same can be said for fusion proteins between SA and GALC of non-human animal species (SA-GALC), and fusion proteins between SA and GALC of non-human animal species, such as the fusion protein MSA-mouse GALC.
 配列番号7で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号7で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:7 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:7.
 hGALC-HSA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもhGALC-HSA融合蛋白質である。また,hGALC-HSA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもhGALC-HSA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもhGALC-HSA融合蛋白質である。また,hGALC-HSA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもhGALC-HSA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のGALCとSAとの融合蛋白質(GALC-SA),及びヒト以外の動物種のGALCとヒト以外の動物種のSAとの融合蛋白質,例えばマウスGALCとMSAとの融合蛋白質についても同様のことがいえる。  An hGALC-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also an hGALC-HSA fusion protein. An hGALC-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also an hGALC-HSA fusion protein. An hGALC-HSA fusion protein modified with something other than sugar chains and phosphate is also an hGALC-HSA fusion protein. An hGALC-HSA fusion protein in which the side chains of the amino acids constituting the protein have been changed by a substitution reaction or the like is also an hGALC-HSA fusion protein. Such a change includes, but is not limited to, the conversion of a cysteine residue to formylglycine. The same can be said about a fusion protein of GALC and SA of a non-human animal species (GALC-SA), and a fusion protein of GALC of a non-human animal species and SA of a non-human animal species, for example, a fusion protein of mouse GALC and MSA.
 つまり,糖鎖により修飾されたhGALC-HSA融合蛋白質は,元のアミノ酸配列を有するhGALC-HSA融合蛋白質に含まれるものとする。また,リン酸により修飾されたhGALC-HSA融合蛋白質は,元のアミノ酸配列を有するhGALC-HSA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するhGALC-HSA融合蛋白質に含まれるものとする。また,hGALC-HSA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するhGALC-HSA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のGALCとSAとの融合蛋白質(GALC-SA),及びヒト以外の動物種のGALCとヒト以外の動物種のSAとの融合蛋白質についても同様のことがいえる。 In other words, hGALC-HSA fusion proteins modified with sugar chains are included in the hGALC-HSA fusion proteins having the original amino acid sequence. Also, hGALC-HSA fusion proteins modified with phosphate are included in the hGALC-HSA fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are included in the hGALC-HSA fusion proteins having the original amino acid sequence. Also, hGALC-HSA fusion proteins in which the side chains of the amino acids constituting the hGALC-HSA fusion protein have been changed by substitution reactions or the like are included in the hGALC-HSA fusion proteins having the original amino acid sequence. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of GALC and SA of a non-human animal species (GALC-SA), and fusion proteins of GALC of a non-human animal species and SA of a non-human animal species.
 また,hGALC-HSA融合蛋白質であって,これを構成するhGALCが,hGALCの前駆体であるものもHSA-hGALC融合蛋白質である。 In addition, an hGALC-HSA fusion protein in which the hGALC that constitutes it is a precursor of hGALC is also an HSA-hGALC fusion protein.
 本発明の一実施形態において,「hGBA-SA融合蛋白質」又は「hGBA-SA」の語は,hGBAのアミノ酸配列のC末端に,直接又はリンカーを介して,SAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hGBAとしての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。hGBA-SA融合蛋白質において,hGBAがhGBAとしての機能を有するというときは,hGBAが,通常の野生型のhGBAの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでhGBA-SA融合蛋白質におけるhGBAの比活性は,当該融合蛋白質の単位質量当たりのhGBAの酵素活性(μM/時間/mg蛋白質)に(当該融合蛋白質の分子量/当該融合蛋白質中のhGBAに相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "hGBA-SA fusion protein" or "hGBA-SA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hGBA, and which has the function of hGBA. Here, there is no particular limitation on the animal species of the SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA. When it is said that hGBA in an hGBA-SA fusion protein has the function of hGBA, it means that hGBA retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hGBA is taken as 100%. Here, the specific activity of hGBA in the hGBA-SA fusion protein is calculated by multiplying the enzyme activity of hGBA per unit mass of the fusion protein (μM/hour/mg protein) by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hGBA).
 本発明の一実施形態において,「hGBA-HSA融合蛋白質」又は「hGBA-HSA」の語は,hGBAのアミノ酸配列のC末端に,直接又はリンカーを介して,HSAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hGBAとしての機能を有するものを示す。ここでhGBA-HSA融合蛋白質がhGALCとしての機能を有するというときは,上記のSA-hGBA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "hGBA-HSA fusion protein" or "hGBA-HSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hGBA, and which has the function of hGBA. When it is said here that the hGBA-HSA fusion protein has the function of hGALC, the definition of the SA-hGBA fusion protein described above can be applied.
 本発明の一実施形態において,ヒト以外の動物種のGBAのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のSAのN末端が結合されているアミノ酸配列を有する融合蛋白質であって,GBAとしての機能を有するものは,「(ヒト以外の動物種)GBA-(ヒト以外の動物種)SA融合蛋白質」と表記される。例えば,マウスGBAとマウスSAの融合蛋白質は,「マウスGBA-MSA融合蛋白質」又は「マウスGBA-MSA」と表記される。これら融合蛋白質についても,GBAとしての機能を有するというときは,上記のhGBA-HSA融合蛋白質における定義を適用することができる。また,これら融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of GBA of a non-human animal species, and which has the function of GBA, is designated as a "(non-human animal species) GBA-(non-human animal species) SA fusion protein." For example, a fusion protein of mouse GBA and mouse SA is designated as a "mouse GBA-MSA fusion protein" or "mouse GBA-MSA." When these fusion proteins are said to have the function of GBA, the definition of the hGBA-HSA fusion protein described above can be applied. In addition, in these fusion proteins, it is preferable that the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited thereto.
 本発明の一実施形態において好ましいhGBA-HSA融合蛋白質は,例えば配列番号41で示されるアミノ酸配列を有するものである。配列番号41で示されるhGBA-HSA融合蛋白質は,野生型hGBAのC末端に配列番号9で示されるアミノ酸配列を有するリンカー配列を介して野生型HSAが結合したものである。配列番号41で示されるhGBA-HSA融合蛋白質は,例えば,配列番号42で示される塩基配列を有する遺伝子にコードされる。また,配列番号41で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hGBAとしての機能を有するものである限りhGBA-HSA融合蛋白質に含まれる。hGBA-HSA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred hGBA-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 41. The hGBA-HSA fusion protein shown in SEQ ID NO: 41 is obtained by linking wild-type HSA to the C-terminus of wild-type hGBA via a linker sequence having the amino acid sequence shown in SEQ ID NO: 9. The hGBA-HSA fusion protein shown in SEQ ID NO: 41 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 42. In addition, hGBA-HSA fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 41 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hGBA. It is preferable that the hGBA-HSA fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 本発明の一実施形態において,「mGBA-MSA融合蛋白質」又は「mGBA-MSA」の語は,mGBAのアミノ酸配列のC末端に,直接又はリンカーを介して,MSAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,mGBAとしての機能を有するものを示す。好ましいmGBA-MSA融合蛋白質は,例えば配列番号47で示されるアミノ酸配列を有するものである。配列番号47で示されるmGBA-MSA融合蛋白質は,野生型mGBAのC末端に配列番号9で示されるアミノ酸配列を有するリンカー配列を介して野生型MSAが結合したものである。配列番号47で示されるmGBA-MSA融合蛋白質は,例えば,配列番号48で示される塩基配列を有する遺伝子にコードされる。また,配列番号47で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,mGBAとしての機能を有するものである限りmGBA-MSA融合蛋白質に含まれる。mGBA-MSA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のマウス血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, the term "mGBA-MSA fusion protein" or "mGBA-MSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of MSA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of mGBA, and which has the function of mGBA. A preferred mGBA-MSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 47. The mGBA-MSA fusion protein shown in SEQ ID NO: 47 is a fusion protein in which wild-type MSA is linked to the C-terminus of wild-type mGBA via a linker sequence having the amino acid sequence shown in SEQ ID NO: 9. The mGBA-MSA fusion protein shown in SEQ ID NO: 47 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 48. In addition, mGBA-MSA fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 47 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of mGBA. It is preferable that the mGBA-MSA fusion protein has mouse serum albumin functions, such as binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this.
 野生型hGBAと野生型HSAとの融合蛋白質であるhGBA-HSA融合蛋白質に変異を加える場合,hGBA部分にのみ変異を加えHSA部分には変異を加えないことも,hGBA部分には変異を加えずにHSA部分にのみ変異を加えることも,また,hGBA部分とHSA部分の何れにも変異を加えることもできる。hGBA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hGBAに変異が加えられたhGBAのアミノ酸配列となる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hGBA部分とHSA部分の何れにも変異を加える場合にあっては,hGBA部分のアミノ酸配列は上述した野生型hGBAに変異が加えられたhGBAのアミノ酸配列となり,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。配列番号41で示されるアミノ酸配列を有するhGBA-HSA融合蛋白質についても同様である。また,配列番号47で示されるアミノ酸配列を有するものを含む, mGBA-MSA融合蛋白質についても同様のことがいえる。配列番号47で示されるアミノ酸配列を有するmGBA-MSA融合蛋白質は,例えば配列番号48で示される塩基配列を有する遺伝子にコードされる。野生型hGBAとヒト以外の動物種の野生型SA(野生型MSAを含む)との融合蛋白質についても同様のことがいえる。 When mutations are introduced into the hGBA-HSA fusion protein, which is a fusion protein of wild-type hGBA and wild-type HSA, mutations can be introduced only in the hGBA portion and not in the HSA portion, mutations can be introduced only in the HSA portion without mutations in the hGBA portion, or mutations can be introduced in both the hGBA portion and the HSA portion. When mutations are introduced only in the hGBA portion, the amino acid sequence of the portion is the amino acid sequence of hGBA obtained by mutating the wild-type hGBA described above. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above. When mutations are introduced both in the hGBA portion and the HSA portion, the amino acid sequence of the hGBA portion is the amino acid sequence of hGBA obtained by mutating the wild-type hGBA described above, and the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by mutating the wild-type HSA described above. The same applies to the hGBA-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 41. The same can be said about mGBA-MSA fusion proteins, including those having the amino acid sequence shown in SEQ ID NO: 47. The mGBA-MSA fusion protein having the amino acid sequence shown in SEQ ID NO: 47 is encoded by a gene having the base sequence shown in SEQ ID NO: 48, for example. The same can be said about fusion proteins of wild-type hGBA and wild-type SA (including wild-type MSA) of an animal species other than human.
 配列番号41で示されるアミノ酸配列を有するhGBA-HSA融合蛋白質に変異を加える場合について,以下例示する。配列番号41で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号41で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。hGBA-HSA融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hGBA部分にのみ加えても,又は両方の部分に加えてもよい。hGBAとヒト以外の動物種のSAとの融合蛋白質(hGBA-SA),及びヒト以外の動物種のGBAとヒト以外の動物種のSAとの融合蛋白質,例えばマウスGBA-MSA融合蛋白質についても同様のことがいえる。 The following is an example of a case where a mutation is made to an hGBA-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:41. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:41 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:41 or to the N-terminus or C-terminus of the amino acid sequence. The hGBA-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hGBA portion, or to both portions. The same can be said for fusion proteins of hGBA and SA of non-human animal species (hGBA-SA), and fusion proteins of GBA of non-human animal species and SA of non-human animal species, such as mouse GBA-MSA fusion protein.
 配列番号41で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号41で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:41 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:41.
 hGBA-HSA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもhGBA-HSA融合蛋白質である。また,hGBA-HSA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもhGBA-HSA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもhGBA-HSA融合蛋白質である。また,hGBA-HSA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもhGBA-HSA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。hGBAとヒト以外の動物種のSAとの融合蛋白質(hGBA-SA),及びヒト以外の動物種のGBAとヒト以外の動物種のSAとの融合蛋白質,例えばマウスGBA-MSA融合蛋白質についても同様のことがいえる。  An hGBA-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also an hGBA-HSA fusion protein. An hGBA-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also an hGBA-HSA fusion protein. An hGBA-HSA fusion protein modified with something other than sugar chains and phosphate is also an hGBA-HSA fusion protein. An hGBA-HSA fusion protein in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like is also an hGBA-HSA fusion protein. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about a fusion protein of hGBA and SA of a non-human animal species (hGBA-SA), and a fusion protein of GBA of a non-human animal species and SA of a non-human animal species, such as mouse GBA-MSA fusion protein.
 つまり,糖鎖により修飾されたhGBA-HSA融合蛋白質は,元のアミノ酸配列を有するhGBA-HSA融合蛋白質に含まれるものとする。また,リン酸により修飾されたhGBA-HSA融合蛋白質は,元のアミノ酸配列を有するhGBA-HSA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するhGBA-HSA融合蛋白質に含まれるものとする。また,hGBA-HSA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するhGBA-HSA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。hGBAとヒト以外の動物種のSAとの融合蛋白質(hGBA-SA),及びヒト以外の動物種のGBAとヒト以外の動物種のSAとの融合蛋白質,例えばマウスGBA-MSA融合蛋白質についても同様のことがいえる。 In other words, hGBA-HSA fusion proteins modified with sugar chains are included in the hGBA-HSA fusion proteins having the original amino acid sequence. Also, hGBA-HSA fusion proteins modified with phosphate are included in the hGBA-HSA fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are included in the hGBA-HSA fusion proteins having the original amino acid sequence. Also, those in which the side chains of the amino acids constituting the hGBA-HSA fusion proteins have been changed by substitution reactions or the like are included in the hGBA-HSA fusion proteins having the original amino acid sequence. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of hGBA and SA of non-human animal species (hGBA-SA) and fusion proteins of GBA of non-human animal species and SA of non-human animal species, such as mouse GBA-MSA fusion protein.
 また,hGBA-HSA融合蛋白質であって,これを構成するhGBAが,hGBAの前駆体であるものもHSA-hGALC融合蛋白質である。 In addition, an hGBA-HSA fusion protein in which the hGBA that constitutes it is a precursor of hGBA is also an HSA-hGALC fusion protein.
 本発明の一実施形態において,「SAとリソソーム酵素との融合蛋白質」,「リソソーム酵素とSAとの融合蛋白質」,又は「血清アルブミンとリソソーム酵素との融合蛋白質」,というときは,上記の「SA-リソソーム酵素融合蛋白質」及び「リソソーム酵素-SA融合蛋白質」のいずれをも含むものとする。また,「SAとGALCとの融合蛋白質」,「GALCとSAとの融合蛋白質」,又は「血清アルブミンとガラクトシルセラミダーゼとの融合蛋白質」,というときは,上記の「SA-GALC融合蛋白質」及び「GALC-SA融合蛋白質」のいずれをも含むものとする。また,「SAとGBAとの融合蛋白質」,「GBAとSAとの融合蛋白質」,又は「血清アルブミンとグルコセレブロシダーゼとの融合蛋白質」,というときは,上記の「SA-GBA融合蛋白質」及び「GBA-SA融合蛋白質」のいずれをも含むものとする。SAがHSAである場合,リソソーム酵素がヒトリソソーム酵素である場合,GALCがhGALCである場合,及びGBAがhGBAである場合も同様である。 In one embodiment of the present invention, the terms "fusion protein of SA and lysosomal enzyme", "fusion protein of lysosomal enzyme and SA", or "fusion protein of serum albumin and lysosomal enzyme" include both the above-mentioned "SA-lysosomal enzyme fusion protein" and "lysosomal enzyme-SA fusion protein". In addition, the terms "fusion protein of SA and GALC", "fusion protein of GALC and SA", or "fusion protein of serum albumin and galactosylceramidase" include both the above-mentioned "SA-GALC fusion protein" and "GALC-SA fusion protein". In addition, the terms "fusion protein of SA and GBA", "fusion protein of GBA and SA", or "fusion protein of serum albumin and glucocerebrosidase" include both the above-mentioned "SA-GBA fusion protein" and "GBA-SA fusion protein". The same applies when SA is HSA, when the lysosomal enzyme is human lysosomal enzyme, when GALC is hGALC, and when GBA is hGBA.
 以下,SAとリソソーム酵素について,HSAとhGALCとの融合蛋白質及びHSAとhGBAとの融合蛋白質を例にとり融合蛋白質の製造法等について詳述するが,これらの記載事項はSAがヒト以外のものである融合蛋白質,リソソーム酵素がGALC及びGBA以外のものである場合にも適用され得る。例えば,ヒト以外の動物種のSAとhGALC又はhGBAとの融合蛋白質,ヒト以外の動物種のSAとヒト以外の動物種のGALC又はGBAとの融合蛋白質,MSAとmGALC又はmGBAとの融合蛋白質についても適用される。  Below, we will provide a detailed description of SA and lysosomal enzymes, taking as examples a fusion protein of HSA and hGALC and a fusion protein of HSA and hGBA, and the manufacturing method of the fusion protein, but these descriptions can also be applied to fusion proteins in which the SA is non-human, and the lysosomal enzyme is other than GALC and GBA. For example, they also apply to fusion proteins of SA of a non-human animal species and hGALC or hGBA, fusion proteins of SA of a non-human animal species and GALC or GBA of a non-human animal species, and fusion proteins of MSA and mGALC or mGBA.
 本発明の一実施形態の融合蛋白質において,SAとリソソーム酵素とは,直接又はリンカーを介して結合される。ここで,「リンカー」というときは,2種類の蛋白質を結合させるときに用いられる構造体,又は2種類またはそれ以上の蛋白質を融合させたときに,いずれの蛋白質にも属さない部分のことをいう。リンカーにはペプチドリンカーと非ペプチドリンカーがある。融合蛋白質の一部を構成するペプチドリンカーを単にリンカーということもできる。また,ペプチドリンカーのアミノ酸配列をリンカー配列ということができる。SAとリソソーム酵素の融合蛋白質にあっては,SAのアミノ酸配列とリソソーム酵素のアミノ酸配列のいずれにも属さない部分のことをいう。つまり,リンカーはSAとリソソーム酵素との間に介在するペプチド鎖のことである。リンカーは,種々の機能を有する。その機能には,SAとリソソーム酵素との間にあってSAとリソソーム酵素とを結合する機能の他,SA及びリソソーム酵素の,融合蛋白質分子内での距離を離すことにより相互の干渉を低減させる機能,SAとリソソーム酵素との間にあってSAとリソソーム酵素とを連結するヒンジとなって,融合蛋白質の立体構造に柔軟性を与える機能等が含まれる。融合蛋白質の分子内において,リンカーは,これら機能の少なくとも一つを発揮する。このことは,例えばSAがHSAであっても,また例えばリソソーム酵素がヒトリソソーム酵素,例えばhGALC又はhGBAであっても,同様である。 In one embodiment of the fusion protein, the SA and the lysosomal enzyme are linked directly or via a linker. Here, the term "linker" refers to a structure used to link two types of proteins, or a portion that does not belong to either protein when two or more types of proteins are fused. Linkers include peptide linkers and non-peptide linkers. A peptide linker that constitutes part of a fusion protein can also be simply called a linker. The amino acid sequence of a peptide linker can also be called a linker sequence. In the case of a fusion protein of SA and a lysosomal enzyme, the linker refers to a portion that does not belong to either the amino acid sequence of SA or the amino acid sequence of the lysosomal enzyme. In other words, the linker is a peptide chain that is interposed between the SA and the lysosomal enzyme. Linkers have various functions. The functions include a function between SA and lysosomal enzyme that binds SA and lysosomal enzyme, a function to reduce mutual interference between SA and lysosomal enzyme by increasing the distance between them within the fusion protein molecule, and a function to act as a hinge between SA and lysosomal enzyme that connects SA and lysosomal enzyme and gives flexibility to the three-dimensional structure of the fusion protein. Within the fusion protein molecule, the linker exerts at least one of these functions. This is the same, for example, when the SA is HSA, and when the lysosomal enzyme is a human lysosomal enzyme, such as hGALC or hGBA.
 SAとリソソーム酵素との融合蛋白質において,リンカーのアミノ酸配列は,融合蛋白質分子の中にあって,リンカーとしての機能が発揮されるものである限り特に限定はない。また,ペプチドリンカーの長さにも,融合蛋白質分子の中にあって,リンカーとしての機能が発揮されるものである限り特に限定はない。ペプチドリンカーは一個又は複数個のアミノ酸から構成されるものである。ペプチドリンカーが複数個のアミノ酸から構成される場合,そのアミノ酸の個数は,好ましくは2~50個であり,より好ましくは5~30個であり,更に好ましくは10~25個である。ペプチドリンカーの好適な例として,Gly-Ser,Gly-Gly-Ser,又は配列番号9~11で示されるアミノ酸配列(これらをあわせて基本配列という)からなるもの,及びこれらを含むものが挙げられる。例えば,ペプチドリンカーは,基本配列が2~10回反復したアミノ酸配列を含むものであり,基本配列が2~6回反復したアミノ酸配列を含むものであり,基本配列が3~5回反復したアミノ酸配列を含むものである。これらのアミノ酸配列中の1個又は複数個のアミノ酸が,欠失,他のアミノ酸へ置換,付加等されたものであってもよい。アミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1又は2個である。アミノ酸を他のアミノ酸へ置換させる場合,置換させるアミノ酸の個数は,好ましくは1又は2個である。アミノ酸を付加する場合,付加されるアミノ酸の個数は,好ましくは1又は2個である。これらアミノ酸の欠失,置換,及び付加を組み合わせて,所望のリンカー部のアミノ酸配列とすることもできる。ペプチドリンカーは一つのアミノ酸からなるものであってもよく,リンカーを構成するアミノ酸は,例えばグリシン,セリンである。このことは,例えばSAがHSAであっても,また例えばリソソーム酵素がヒトリソソーム酵素,例えばhGALC又はhGBAであっても,同様である。 In the fusion protein of SA and a lysosomal enzyme, the amino acid sequence of the linker is not particularly limited as long as it functions as a linker in the fusion protein molecule. The length of the peptide linker is also not particularly limited as long as it functions as a linker in the fusion protein molecule. The peptide linker is composed of one or more amino acids. When the peptide linker is composed of multiple amino acids, the number of amino acids is preferably 2 to 50, more preferably 5 to 30, and even more preferably 10 to 25. Suitable examples of peptide linkers include those consisting of Gly-Ser, Gly-Gly-Ser, or amino acid sequences shown in SEQ ID NOs: 9 to 11 (collectively referred to as basic sequences), and those containing these. For example, the peptide linker is one that contains an amino acid sequence in which the basic sequence is repeated 2 to 10 times, one that contains an amino acid sequence in which the basic sequence is repeated 2 to 6 times, and one that contains an amino acid sequence in which the basic sequence is repeated 3 to 5 times. One or more amino acids in these amino acid sequences may be deleted, replaced with other amino acids, added, etc. When deleting an amino acid, the number of amino acids to be deleted is preferably 1 or 2. When substituting an amino acid with another amino acid, the number of amino acids to be substituted is preferably 1 or 2. When adding an amino acid, the number of amino acids to be added is preferably 1 or 2. The desired amino acid sequence of the linker portion can be obtained by combining these deletions, substitutions, and additions of amino acids. The peptide linker may be composed of one amino acid, and the amino acid constituting the linker is, for example, glycine or serine. This is the same, for example, when the SA is HSA, and when the lysosomal enzyme is, for example, a human lysosomal enzyme, such as hGALC or hGBA.
 HSAとhGALCとの融合蛋白質は,HSAをコードする遺伝子の下流又は上流に,インフレームでhGALCをコードする遺伝子を結合させたDNA断片を組み込んだ発現ベクターを作製し,この発現ベクターを導入して形質転換させた宿主細胞を培養することにより,組換え蛋白質として作製することができる。また,HSAとhGBAとの融合蛋白質は,HSAをコードする遺伝子の下流又は上流に,インフレームでhGBAをコードする遺伝子を結合させたDNA断片を組み込んだ発現ベクターを作製し,この発現ベクターを導入して形質転換させた宿主細胞を培養することにより,組換え蛋白質として作製することができる。このようにして組換え蛋白質として作製される融合蛋白質は,一本鎖ポリペプチドからなる。また,本発明において,組換え蛋白質として作製された融合蛋白質のことを,組換え融合蛋白質という。 A fusion protein of HSA and hGALC can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hGALC is linked in-frame downstream or upstream of a gene encoding HSA, and culturing a host cell transformed by introducing this expression vector. A fusion protein of HSA and hGBA can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hGBA is linked in-frame downstream or upstream of a gene encoding HSA, and culturing a host cell transformed by introducing this expression vector. The fusion protein produced as a recombinant protein in this way consists of a single-chain polypeptide. In the present invention, a fusion protein produced as a recombinant protein is called a recombinant fusion protein.
 組換え融合蛋白質としてHSA-hGALC融合蛋白質を作製する場合,HSAをコードする遺伝子の下流にインフレームでhGALCをコードする遺伝子を結合させることにより,HSAのアミノ酸配列のC末端にhGALCのアミノ酸配列を有する融合蛋白質を得ることができる。逆に,HSAをコードする遺伝子の上流にインフレームでhGALCをコードする遺伝子を結合させることにより,HSAのアミノ酸配列のN末端にhGALCのアミノ酸配列を有するhGALC-HSA融合蛋白質が得られる。また,HSAをコードする遺伝子の下流にインフレームでhGBAをコードする遺伝子を結合させることにより,HSAのアミノ酸配列のC末端にhGBAのアミノ酸配列を有するHSA-hGBA融合蛋白質を得ることができる。逆にHSAをコードする遺伝子の上流にインフレームでhGBAをコードする遺伝子を結合させることにより,HSAのアミノ酸配列のN末端にhGBAのアミノ酸配列を有するhGBA-HSA融合蛋白質が得られる。いずれの場合においても,組換え融合蛋白質として作製される融合蛋白質は,一本鎖ポリペプチドである。 When preparing an HSA-hGALC fusion protein as a recombinant fusion protein, a gene encoding hGALC can be linked in-frame downstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hGALC at the C-terminus of the amino acid sequence of HSA. Conversely, a gene encoding hGALC can be linked in-frame upstream of a gene encoding HSA to obtain an hGALC-HSA fusion protein having the amino acid sequence of hGALC at the N-terminus of the amino acid sequence of HSA. Also, a gene encoding hGBA can be linked in-frame downstream of a gene encoding HSA to obtain an HSA-hGBA fusion protein having the amino acid sequence of hGBA at the C-terminus of the amino acid sequence of HSA. Conversely, a gene encoding hGBA can be linked in-frame upstream of a gene encoding HSA to obtain an hGBA-HSA fusion protein having the amino acid sequence of hGBA at the N-terminus of the amino acid sequence of HSA. In either case, the fusion protein prepared as a recombinant fusion protein is a single-chain polypeptide.
 なお,本発明において,「一本鎖ポリペプチド」というときは,一つのN末端と一つのC末端を有するポリペプチドであって,枝分かれするペプチド鎖のないポリペプチドのことをいう。この条件を満たす限り,分子内ジスルフィド結合を有するもの,糖鎖,脂質,リン脂質等で修飾されたものも一本鎖ポリペプチドである。また,一本鎖ポリペプチドが非共有結合によりダイマー等の複合体を形成した場合にあっては,当該複合体を形成する個々のペプチド鎖は一本鎖ポリペプチドと理解され,当該複合体自体は,一本鎖ポリペプチドの集合体であると理解される。 In the present invention, the term "single-chain polypeptide" refers to a polypeptide that has one N-terminus and one C-terminus and has no branched peptide chains. As long as this condition is met, those that have intramolecular disulfide bonds and those that are modified with sugar chains, lipids, phospholipids, etc. are also single-chain polypeptides. In addition, when single-chain polypeptides form complexes such as dimers through non-covalent bonds, each peptide chain that forms the complex is understood to be a single-chain polypeptide, and the complex itself is understood to be an assembly of single-chain polypeptides.
 融合蛋白質の一本鎖ポリペプチド内で,HSAのアミノ酸配列がhGALC又はhGBAのアミノ酸配列のN末端側に位置する場合,HSAのC末端とhGALC又はhGBAのN末端が,ペプチド結合により直接,又はリンカーを介して結合される。図1にN末端側からHSA,リンカー及びhGALCを順に有する一本鎖ポリペプチドのHSA-hGALC融合蛋白質を模式的に示す。該HSA-hGALC融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とhGALCのN末端がペプチド結合により結合したものである。また,図2にN末端側からHSA,リンカー及びhGBAを順に有する一本鎖ポリペプチドのHSA-hGBA融合蛋白質を模式的に示す。該HSA-hGBA融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とhGBAのN末端がペプチド結合により結合したものである。 When the amino acid sequence of HSA is located on the N-terminal side of the amino acid sequence of hGALC or hGBA in the single-chain polypeptide of the fusion protein, the C-terminus of HSA and the N-terminus of hGALC or hGBA are bound directly by a peptide bond or via a linker. Figure 1 shows a schematic diagram of a single-chain polypeptide HSA-hGALC fusion protein having HSA, a linker, and hGALC in this order from the N-terminus. In this HSA-hGALC fusion protein, the C-terminus of HSA and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of hGALC are bound by a peptide bond. Figure 2 also shows a schematic diagram of a single-chain polypeptide HSA-hGBA fusion protein having HSA, a linker, and hGBA in this order from the N-terminus. In this HSA-hGBA fusion protein, the C-terminus of HSA and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of hGBA are bound by a peptide bond.
 また,融合蛋白質の一本鎖ポリペプチド内で,hGALC又はhGBAのアミノ酸配列がHSAのアミノ酸配列のN末端に位置する場合,hGALC又はhGBAのC末端とHSAのN末端が,ペプチド結合により直接,又はリンカーを介して結合される。図3にN末端側からhGALC,リンカー及びHSAを順に有する一本鎖ポリペプチドのhGALC-HSA融合蛋白質を模式的に示す。該hGALC-HSA融合蛋白質はhGALCのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。また,図4にN末端側からhGBA,リンカー及びHSAを順に有する一本鎖ポリペプチドのhGBA-HSA融合蛋白質を模式的に示す。該hGBA-HSA融合蛋白質はhGBAのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。 In addition, when the amino acid sequence of hGALC or hGBA is located at the N-terminus of the amino acid sequence of HSA in the single-chain polypeptide of the fusion protein, the C-terminus of hGALC or hGBA and the N-terminus of HSA are bound directly by a peptide bond or via a linker. Figure 3 shows a single-chain polypeptide hGALC-HSA fusion protein having hGALC, a linker, and HSA in this order from the N-terminus. In this hGALC-HSA fusion protein, the C-terminus of hGALC and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond. Figure 4 shows a single-chain polypeptide hGBA-HSA fusion protein having hGBA, a linker, and HSA in this order from the N-terminus. In this hGBA-HSA fusion protein, the C-terminus of hGBA and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond.
 本発明の一実施形態において,HSAとhGALCとの融合蛋白質というときは,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhGALCを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhGALCとしての発現量が,濃度あるいは酵素活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍等となることを特徴とする融合蛋白質である。また,本発明の一実施形態において,HSAとhGBAとの融合蛋白質というときは,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhGBAを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhGBAとしての発現量が,濃度あるいは酵素活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍等となることを特徴とする融合蛋白質である。ここで同一条件下とは,発現ベクター,宿主細胞,培養条件等が同一であることをいう。このとき用いられる好ましい宿主細胞は,CHO細胞,NS/0細胞等の哺乳動物細胞であるが,特にCHO細胞である。 In one embodiment of the present invention, a fusion protein of HSA and hGALC refers to a fusion protein characterized in that when expressed in host cells as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hGALC in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, compared to when wild-type hGALC is expressed in host cells as a recombinant protein under the same conditions. In one embodiment of the present invention, the fusion protein of HSA and hGBA refers to a fusion protein characterized in that when expressed in a host cell as a recombinant protein, particularly when expressed so that the recombinant protein is secreted from the cell and accumulated in the culture medium, the amount of hGBA expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, etc., in terms of concentration or enzyme activity, compared to when wild-type hGBA is expressed in a host cell as a recombinant protein under the same conditions. Here, "under the same conditions" means that the expression vector, host cell, culture conditions, etc. are the same. The preferred host cells used in this case are mammalian cells such as CHO cells and NS/0 cells, and particularly CHO cells.
 かかるHSAとhGALCとの融合蛋白質の好ましい実施形態として,以下の(1)及び(2)が挙げられる:
(1)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号1で示される野生型のhGALCのアミノ酸配列のN末端が,Gly-Serで表されるリンカーを介して結合したものである,配列番号5で示されるアミノ酸配列を有するもの,
(2)配列番号1で示される野生型のhGALCのアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端が,Gly-Serで表されるリンカーを介して結合したものである,配列番号7で示されるアミノ酸配列を有するもの。
Preferred embodiments of the fusion protein of HSA and hGALC include the following (1) and (2):
(1) A polypeptide having an amino acid sequence shown in SEQ ID NO:5, in which the N-terminus of the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO:3 via a linker represented by Gly-Ser;
(2) Having the amino acid sequence shown in SEQ ID NO: 7, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hGALC shown in SEQ ID NO: 1 via a linker represented by Gly-Ser.
 また,かかるHSAとhGBAとの融合蛋白質の好ましい実施形態として,以下の(1)及び(2)が挙げられる:
(1)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号37で示される野生型のhGBAのアミノ酸配列のN末端が,配列番号9で示されるアミノ酸配列を有するリンカーを介して結合したものである,配列番号39で示されるアミノ酸配列を有するもの,
(2)配列番号37で示される野生型のhGBAのアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端が,配列番号9で示されるアミノ酸配列を有するリンカーを介して結合したものである,配列番号41で示されるアミノ酸配列を有するもの。
Preferred embodiments of the fusion protein of HSA and hGBA include the following (1) and (2):
(1) having the amino acid sequence shown in SEQ ID NO: 39, in which the N-terminus of the amino acid sequence of wild-type hGBA shown in SEQ ID NO: 37 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via a linker having the amino acid sequence shown in SEQ ID NO: 9;
(2) Having the amino acid sequence shown in SEQ ID NO: 41, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hGBA shown in SEQ ID NO: 37 via a linker having the amino acid sequence shown in SEQ ID NO: 9.
 本発明の一実施形態におけるHSAとhGALCとの融合蛋白質は,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件下で野生型のhGALCを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhGALCとしての発現量が,濃度あるいは酵素活性換算で増加することを特徴とするものである。従って,HSAとhGALCとの融合蛋白質は,組換え蛋白質として製造したときに,野生型のhGALCと比較して生産効率を上昇させることができるので,生産コストを低減することができる。また,本発明の一実施形態におけるHSAとhGBAとの融合蛋白質は,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件下で野生型のhGBAを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhGBAとしての発現量が,濃度あるいは酵素活性換算で増加することを特徴とするものである。従って,HSAとhGBAとの融合蛋白質は,組換え蛋白質として製造したときに,野生型のhGBAと比較して生産効率を上昇させることができるので,生産コストを低減することができる。なお,組換え蛋白質を有効成分として含有する医薬品は,非常に高価であることが知られている。従って,同一条件で製造したときに得られる組換え蛋白質の量を数パーセント,例えば3~9%上昇させることにも,多大な経済的効果がある。同様のことは,ヒトリソソーム酵素,例えば,hGALC,hGBAについてもいえる。 In one embodiment of the present invention, the fusion protein of HSA and hGALC is characterized in that when expressed in a host cell as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hGALC in the culture supernatant is increased in terms of concentration or enzyme activity compared to when wild-type hGALC is expressed in a host cell as a recombinant protein under the same conditions. Therefore, when the fusion protein of HSA and hGALC is produced as a recombinant protein, the production efficiency can be increased compared to wild-type hGALC, and production costs can be reduced. In one embodiment of the present invention, the fusion protein of HSA and hGBA is characterized in that when expressed in a host cell as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hGBA in the culture supernatant is increased in terms of concentration or enzyme activity compared to when wild-type hGBA is expressed in a host cell as a recombinant protein under the same conditions. Therefore, when the fusion protein of HSA and hGBA is produced as a recombinant protein, the production efficiency can be increased compared to wild-type hGBA, and production costs can be reduced. It is known that pharmaceuticals that contain recombinant proteins as active ingredients are very expensive. Therefore, even increasing the amount of recombinant protein obtained by a few percent, for example 3-9%, when produced under the same conditions can have a significant economic effect. The same can be said about human lysosomal enzymes, such as hGALC and hGBA.
 なお,本明細書において,組換え蛋白質として宿主細胞で発現させたHSAとhGALCとの融合蛋白質と,同一条件下で組換え蛋白質として宿主細胞で発現させた野生型のhGALCの,発現量を比較するときは,好ましくは発現した蛋白質の質量で比較するのではなく,発現した蛋白質のGALC酵素活性で比較される。HSAとhGBAとの融合蛋白質,ヒト以外の動物種のSAとhGALC又はhGBAとの融合蛋白質,及びヒト以外の動物種のSAとヒト以外の動物種のGALC又はGBAとの融合蛋白質,例えば,MSAとmGALC又はmGBAと融合蛋白質の発現量についても同様とする。 In this specification, when comparing the expression levels of a fusion protein of HSA and hGALC expressed in a host cell as a recombinant protein with wild-type hGALC expressed in a host cell under the same conditions, it is preferable to compare the GALC enzyme activity of the expressed protein rather than the mass of the expressed protein. The same applies to the expression levels of a fusion protein of HSA and hGBA, a fusion protein of SA of a non-human animal species and hGALC or hGBA, and a fusion protein of SA of a non-human animal species and GALC or GBA of a non-human animal species, for example, a fusion protein of MSA and mGALC or mGBA.
 HSAとhGALCとの融合蛋白質及びHSAとhGBAとの融合蛋白質は,当該融合蛋白質をコードする遺伝子を組み込んだ発現ベクターを用いて形質転換させた宿主細胞を培養することにより,組換え蛋白質として製造することができる。 HSA/hGALC fusion protein and HSA/hGBA fusion protein can be produced as recombinant proteins by culturing host cells transformed with an expression vector incorporating a gene encoding the fusion protein.
 このとき用いられる宿主細胞は,かかる発現ベクターを導入することによりHSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質を発現させることができるものである限り特に制限はなく,哺乳動物細胞,酵母,植物細胞,昆虫細胞等の真核生物細胞の何れであってもよいが,哺乳動物細胞が特に好適である。 The host cells used in this case are not particularly limited as long as they are capable of expressing a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA by introducing such an expression vector, and may be any eukaryotic cell such as a mammalian cell, yeast, plant cell, or insect cell, although mammalian cells are particularly preferred.
 哺乳動物細胞を宿主細胞として使用する場合,該哺乳動物細胞の種類について特に限定はないが,ヒト,マウス,チャイニーズハムスター由来の細胞が好ましく,特にチャイニーズハムスター卵巣細胞由来のCHO細胞,又はマウス骨髄腫に由来するNS/0細胞が好ましい。またこのときHSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質をコードする遺伝子を含むDNA断片を組み込んで発現させるために用いる発現ベクターは,哺乳動物細胞内に導入したとき該遺伝子の発現をもたらすものであれば特に限定なく用いることができる。発現ベクターに組み込まれた該遺伝子は,哺乳動物細胞内で遺伝子の転写の頻度を調節することができるDNA配列(遺伝子発現制御部位)の下流に配置される。本発明において用いることのできる遺伝子発現制御部位としては,例えば,サイトメガロウイルス由来のプロモーター,SV40初期プロモーター,ヒト伸長因子-1α(EF-1α)プロモーター,ヒトユビキチンCプロモーター等が挙げられる。 When mammalian cells are used as host cells, there is no particular limitation on the type of mammalian cell, but cells derived from humans, mice, or Chinese hamsters are preferred, and in particular CHO cells derived from Chinese hamster ovary cells or NS/0 cells derived from mouse myeloma are preferred. In this case, the expression vector used to incorporate and express a DNA fragment containing a gene encoding a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA can be used without particular limitation as long as it brings about expression of the gene when introduced into a mammalian cell. The gene incorporated in the expression vector is placed downstream of a DNA sequence (gene expression control site) that can regulate the frequency of gene transcription in mammalian cells. Examples of gene expression control sites that can be used in the present invention include a promoter derived from cytomegalovirus, an SV40 early promoter, a human elongation factor-1α (EF-1α) promoter, and a human ubiquitin C promoter.
 目的の蛋白質をコードする遺伝子の下流側に内部リボソーム結合部位(IRES: internal ribosome entry site)を介して,選択マーカーとしてグルタミン合成酵素(GS)を配置した発現ベクターが知られている(国際特許公報WO2012/063799,WO2013/161958)。これら文献に記載された発現ベクターは,HSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質の製造に特に好適に使用することができる。 Expression vectors are known in which glutamine synthetase (GS) is placed as a selection marker downstream of a gene encoding a target protein via an internal ribosome entry site (IRES) (International Patent Publications WO2012/063799, WO2013/161958). The expression vectors described in these publications are particularly suitable for producing fusion proteins of HSA and hGALC or fusion proteins of HSA and hGBA.
 なお,本発明の一実施形態において,「内部リボソーム結合部位」とは,mRNA鎖の内部に存在する,リボソームが直接結合し且つキャップ構造非依存性に翻訳を開始し得る領域(構造),又は転写されることにより該領域を生じるDNA鎖の領域(構造)である。また,本発明において,「内部リボソーム結合部位をコードする遺伝子」とは,転写されることにより該領域を生じるDNA鎖の領域(構造)である。内部リボソーム結合部位は,一般に,IRES(internal ribosome entry site)と称され,ピコルナウイルス科のウイルス(ポリオウイルス,ライノウイルス,マウス脳心筋炎ウイルスなど),口蹄疫ウイルス,A型肝炎ウイルス,C型肝炎ウイルス,コロナウイルス,ウシ腸内ウイルス,サイラーのネズミ脳脊髄炎ウイルス,コクサッキーB型ウイルス等のウイルスの5'非翻訳領域,ヒト免疫グロブリン重鎖結合蛋白質,ショウジョウバエアンテナペディア,ショウジョウバエウルトラビトラックス等の遺伝子の5'非翻訳領域に見出されている。ピコルナウイルスの場合,そのIRESは,mRNAの5'非翻訳領域に存在する約450bpからなる領域である。ここで「ウイルスの5'非翻訳領域」とは,ウイルスのmRNAの5'非翻訳領域,又は転写されることにより該領域を生じるDNA鎖の領域(構造)である。 In one embodiment of the present invention, the term "internal ribosome binding site" refers to a region (structure) present within an mRNA strand to which a ribosome can directly bind and initiate translation independent of a cap structure, or a region (structure) of a DNA strand that generates the region by being transcribed. In addition, in the present invention, the term "gene encoding an internal ribosome binding site" refers to a region (structure) of a DNA strand that generates the region by being transcribed. Internal ribosome binding sites are generally called IRES (internal ribosome entry sites), and have been found in the 5' untranslated regions of viruses such as Picornaviridae viruses (poliovirus, rhinovirus, mouse encephalomyocarditis virus, etc.), foot and mouth disease virus, hepatitis A virus, hepatitis C virus, coronavirus, bovine enteric virus, Theiler's murine encephalomyelitis virus, and Coxsackie B virus, as well as in the 5' untranslated regions of genes such as human immunoglobulin heavy chain binding protein, Drosophila antennapedia, and Drosophila ultravithorax. In the case of picornaviruses, the IRES is a region of approximately 450 bp that exists in the 5' untranslated region of the mRNA. Here, the "5' untranslated region of the virus" refers to the 5' untranslated region of the viral mRNA, or the region (structure) of the DNA strand that produces said region by being transcribed.
 例えば,目的の蛋白質を発現させるための発現ベクターであって,第1の遺伝子発現制御部位,並びに,その下流に該蛋白質をコードする遺伝子,更に下流に内部リボソーム結合部位,及び更に下流にグルタミン合成酵素をコードする遺伝子を含み,且つ,上記第1の遺伝子発現制御部位の又はこれとは別の第2の遺伝子発現制御部位の下流にジヒドロ葉酸レダクターゼ遺伝子又は薬剤耐性遺伝子を更に含んでなる発現ベクターは,HSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質の製造に好適に使用できる。この発現ベクターにおいて,第1の遺伝子発現制御部位又は第2の遺伝子発現制御部位としては,サイトメガロウイルス由来のプロモーター,SV40初期プロモーター,ヒト伸長因子-1αプロモーター(hEF-1αプロモーター),ヒトユビキチンCプロモーターが好適に用いられるが,hEF-1αプロモーターが特に好適である。 For example, an expression vector for expressing a target protein, which comprises a first gene expression control site, a gene encoding the protein downstream of the first gene expression control site, an internal ribosome binding site further downstream, and a gene encoding glutamine synthetase further downstream, and further comprises a dihydrofolate reductase gene or a drug resistance gene downstream of the first gene expression control site or a second gene expression control site different from the first gene expression control site, can be suitably used for producing a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA. In this expression vector, the first gene expression control site or the second gene expression control site can be suitably used with a promoter derived from cytomegalovirus, an SV40 early promoter, a human elongation factor-1α promoter (hEF-1α promoter), or a human ubiquitin C promoter, with the hEF-1α promoter being particularly suitable.
 また,内部リボソーム結合部位としては,ピコルナウイルス科のウイルス(マウス脳心筋炎ウイルスを含む),口蹄疫ウイルス,A型肝炎ウイルス,C型肝炎ウイルス,コロナウイルス,ウシ腸内ウイルス,サイラーのネズミ脳脊髄炎ウイルス,コクサッキーB型ウイルスからなる群より選択されるウイルスのゲノム,又はヒト免疫グロブリン重鎖結合蛋白質遺伝子,ショウジョウバエアンテナペディア遺伝子,ショウジョウバエウルトラビトラックス遺伝子からなる群から選択される遺伝子の5’非翻訳領域に由来するものが好適に用いられるが,マウス脳心筋炎ウイルスゲノムの5’非翻訳領域に由来する内部リボソーム結合部位が特に好適である。マウス脳心筋炎ウイルスゲノムの5’非翻訳領域に由来する内部リボソーム結合部位を用いる場合,野生型のもの以外に,野生型の内部リボソーム結合部位に含まれる複数の開始コドンのうちの一部が破壊されたものも好適に使用できる。また,この発現ベクターにおいて,好適に用いられる薬剤耐性遺伝子は,好ましくはピューロマイシン又はネオマイシン耐性遺伝子であり,より好ましくはピューロマイシン耐性遺伝子である。 Furthermore, as the internal ribosome binding site, those derived from the 5' untranslated region of the genome of a virus selected from the group consisting of a virus of the picornavirus family (including mouse encephalomyocarditis virus), foot and mouth disease virus, hepatitis A virus, hepatitis C virus, coronavirus, bovine enteric virus, Theiler's murine encephalomyelitis virus, and Coxsackie B virus, or a gene selected from the group consisting of the human immunoglobulin heavy chain binding protein gene, the Drosophila antennapedia gene, and the Drosophila ultravithorax gene, are preferably used, but an internal ribosome binding site derived from the 5' untranslated region of the mouse encephalomyocarditis virus genome is particularly preferable. When using an internal ribosome binding site derived from the 5' untranslated region of the mouse encephalomyocarditis virus genome, in addition to the wild type, one in which some of the multiple initiation codons contained in the wild type internal ribosome binding site have been destroyed can also be preferably used. Furthermore, in this expression vector, the drug resistance gene preferably used is a puromycin or neomycin resistance gene, and more preferably a puromycin resistance gene.
 また,例えば,目的の蛋白質を発現させるための発現ベクターであって,ヒト伸長因子-1αプロモーター,その下流に該蛋白質をコードする遺伝子,更に下流にマウス脳心筋炎ウイルスゲノムの5’非翻訳領域に由来する内部リボソーム結合部位,及び更に下流にグルタミン合成酵素をコードする遺伝子を含み,且つ別の遺伝子発現制御部位及びその下流にジヒドロ葉酸レダクターゼ遺伝子を更に含む発現ベクターであって,該内部リボソーム結合部位が,野生型の内部リボソーム結合部位に含まれる複数の開始コドンのうちの一部が破壊されたものである発現ベクターは,HSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質の製造に好適に使用できる。このような発現ベクターとして,WO2013/161958に記載された発現ベクターが挙げられる。 Also, for example, an expression vector for expressing a target protein, which contains a human elongation factor-1α promoter, a gene encoding the protein downstream thereof, an internal ribosome binding site derived from the 5' untranslated region of the mouse encephalomyocarditis virus genome further downstream thereof, and a gene encoding glutamine synthetase further downstream thereof, and further contains another gene expression control site and a dihydrofolate reductase gene downstream thereof, in which the internal ribosome binding site is an internal ribosome binding site in which some of the multiple initiation codons contained in the wild-type internal ribosome binding site have been destroyed, can be suitably used for producing a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA. Examples of such expression vectors include the expression vectors described in WO2013/161958.
 また,例えば,目的の蛋白質を発現させるための発現ベクターであって,ヒト伸長因子-1αプロモーター,その下流に該蛋白質をコードする遺伝子,更に下流にマウス脳心筋炎ウイルスゲノムの5’非翻訳領域に由来する内部リボソーム結合部位,及び更に下流にグルタミン合成酵素をコードする遺伝子を含み,且つ別の遺伝子発現制御部位及びその下流に薬剤耐性遺伝子を更に含む発現ベクターであって,該内部リボソーム結合部位が,野生型の内部リボソーム結合部位に含まれる複数の開始コドンのうちの一部が破壊されたものである発現ベクターは,HSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質の製造に好適に使用できる。このような発現ベクターとして,WO2012/063799に記載されたpE-mIRES-GS-puro及びWO2013/161958に記載されたpE-mIRES-GS-mNeoが挙げられる。 Also, for example, an expression vector for expressing a target protein, which contains a human elongation factor-1α promoter, a gene encoding the protein downstream thereof, an internal ribosome binding site derived from the 5' untranslated region of the mouse encephalomyocarditis virus genome further downstream thereof, and a gene encoding glutamine synthetase further downstream thereof, and further contains another gene expression control site and a drug resistance gene downstream thereof, in which the internal ribosome binding site is an internal ribosome binding site in which some of the multiple initiation codons contained in the wild-type internal ribosome binding site have been destroyed, can be suitably used for producing a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA. Examples of such expression vectors include pE-mIRES-GS-puro described in WO2012/063799 and pE-mIRES-GS-mNeo described in WO2013/161958.
 野生型のマウス脳心筋炎ウイルスゲノムの5’非翻訳領域に由来する内部リボソーム結合部位の3’末端には,3つの開始コドン(ATG)が存在している。上記のpE-mIRES-GS-puro及びpE-mIRES-GS-mNeoは,開始コドンのうちの一部が破壊されたIRESを有する発現ベクターである。 The 3' end of the internal ribosome binding site derived from the 5' untranslated region of the wild-type murine encephalomyocarditis virus genome contains three initiation codons (ATG). The above pE-mIRES-GS-puro and pE-mIRES-GS-mNeo are expression vectors that have an IRES in which some of the initiation codons have been destroyed.
 HSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質は,これをコードする遺伝子を組み込んだ発現ベクターを導入した宿主細胞を培養することにより,細胞中又は培地中に発現させることができる。哺乳動物細胞が宿主細胞である場合のHSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質の発現方法について,以下に詳述する。  The fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA can be expressed in cells or in a medium by culturing a host cell into which an expression vector incorporating a gene encoding the protein has been introduced. The method of expressing the fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA when the host cell is a mammalian cell is described in detail below.
 哺乳動物細胞の培養のための培地としては,哺乳動物細胞を培養して増殖させることのできるものであれば,特に限定なく用いることができるが,好ましくは無血清培地が用いられる。本発明において,組換え蛋白質生産用培地として用いられる無血清培地としては,例えば,アミノ酸を3~700 mg/L,ビタミン類を0.001~50 mg/L,単糖類を0.3~10 g/L,無機塩を0.1~10000 mg/L,微量元素を0.001~0.1 mg/L,ヌクレオシドを0.1~50 mg/L,脂肪酸を0.001~10 mg/L,ビオチンを0.01~1 mg/L,ヒドロコルチゾンを0.1~20 μg/L,インシュリンを0.1~20 mg/L,ビタミンB12を0.1~10 mg/L,プトレッシンを0.01~1 mg/L,ピルビン酸ナトリウムを10~500 mg/L,及び水溶性鉄化合物を含有する培地が好適に用いられる。所望により,チミジン,ヒポキサンチン,慣用のpH指示薬及び抗生物質等を培地に添加してもよい。 As a medium for culturing mammalian cells, any medium that can be used for culturing and growing mammalian cells can be used without any particular limitations, but preferably a serum-free medium is used. In the present invention, a serum-free medium used as a medium for producing recombinant proteins is preferably one containing, for example, 3 to 700 mg/L amino acids, 0.001 to 50 mg/L vitamins, 0.3 to 10 g/L monosaccharides, 0.1 to 10,000 mg/L inorganic salts, 0.001 to 0.1 mg/L trace elements, 0.1 to 50 mg/L nucleosides, 0.001 to 10 mg/L fatty acids, 0.01 to 1 mg/L biotin, 0.1 to 20 μg/L hydrocortisone, 0.1 to 20 mg/L insulin, 0.1 to 10 mg/L vitamin B12, 0.01 to 1 mg/L putrescine, 10 to 500 mg/L sodium pyruvate and a water-soluble iron compound. If desired, thymidine, hypoxanthine, conventional pH indicators, antibiotics, etc. may be added to the medium.
 組換え蛋白質生産用培地として用いられる無血清培地として,DMEM/F12培地(DMEMとF12の混合培地)を基本培地として用いてもよく,これら各培地は当業者に周知である。更にまた,無血清培地として,炭酸水素ナトリウム,L-グルタミン,D-グルコース,インスリン,ナトリウムセレナイト,ジアミノブタン,ヒドロコルチゾン,硫酸鉄(II),アスパラギン,アスパラギン酸,セリン及びポリビニルアルコールを含むものである,DMEM(HG)HAM改良型(R5)培地を使用してもよい。更には市販の無血清培地,例えば,CD OptiCHOTM培地,CHO-S-SFM II培地又はCD CHO培地(Thermo Fisher Scientific社),EX-CELLTM302培地又はEX-CELLTM325-PF培地(SAFC Biosciences社)等を基本培地として使用することもできる。 As a serum-free medium used as a medium for recombinant protein production, DMEM/F12 medium (mixture of DMEM and F12) may be used as a basic medium, and these media are well known to those skilled in the art. Furthermore, as a serum-free medium, DMEM(HG)HAM Improved (R5) medium, which contains sodium bicarbonate, L-glutamine, D-glucose, insulin, sodium selenite, diaminobutane, hydrocortisone, ferrous sulfate (II), asparagine, aspartic acid, serine, and polyvinyl alcohol, may be used. Furthermore, commercially available serum-free media, such as CD OptiCHO TM medium, CHO-S-SFM II medium, or CD CHO medium (Thermo Fisher Scientific), EX-CELL TM 302 medium, or EX-CELL TM 325-PF medium (SAFC Biosciences), may be used as a basic medium.
 HSAとhGALCとの融合蛋白質は,これをコードする遺伝子を組み込んだ発現ベクターを導入した宿主細胞を上記の無血清培地で培養して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhGALCを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhGALCとしての発現量が,濃度あるいは酵素活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍等となることを特徴とする。 The fusion protein of HSA and hGALC is characterized in that when host cells into which an expression vector incorporating a gene encoding the fusion protein has been introduced are cultured in the above-mentioned serum-free medium to be expressed as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hGALC expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, compared to when wild-type hGALC is expressed as a recombinant protein in host cells under the same conditions.
 また,HSAとhGBAとの融合蛋白質は,これをコードする遺伝子を組み込んだ発現ベクターを導入した宿主細胞を上記の無血清培地で培養して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhGBAを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhGBAとしての発現量が,濃度あるいは酵素活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍等となることを特徴とする。ここで同一条件下とは,発現ベクター,宿主細胞,培養条件等が同一であることをいう。このとき用いられる宿主細胞は,好ましくは哺乳動物細胞,特に,CHO細胞,NS/0細胞等の組換え蛋白質の製造に用いられる通常の細胞である。  Furthermore, when a fusion protein of HSA and hGBA is expressed as a recombinant protein by culturing a host cell into which an expression vector incorporating a gene encoding the fusion protein has been introduced in the above-mentioned serum-free medium, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hGBA in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, etc., in terms of concentration or enzyme activity, compared to when wild-type hGBA is expressed as a recombinant protein in a host cell under the same conditions. Here, "under the same conditions" means that the expression vector, host cell, culture conditions, etc. are the same. The host cell used in this case is preferably a mammalian cell, particularly a normal cell used for producing recombinant proteins, such as a CHO cell or an NS/0 cell.
 野生型のhGALCは,これをコードする遺伝子を組込んだ発現ベクターで形質転換させた宿主細胞を用いて組換え体として発現させたとき,当該宿主細胞の生存率が低下等の理由により,組換え体として効率的に製造することが困難である。しかし,hGALCをHSAとの融合蛋白質として発現させることにより,野生型のhGALCを発現させたときに観察される宿主細胞の生存率の低下が抑制される。HSAとhGALCとの融合蛋白質は,これをコードする遺伝子を組み込んだ発現ベクターを導入した哺乳動物細胞を無血清培地で培養して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型hGALCを同一条件で組換え蛋白質として発現させたときと比較して,発現量が増加する。この発現量の増加には上記の生存率の低下の抑制が寄与しているものと考えられる。ヒト以外の動物種のSAとhGALCの融合蛋白質,ヒト以外の動物種のSAとヒト以外の動物種のGALCの融合蛋白質についても同様である。なお,当該融合蛋白質の発現に用いられる宿主細胞は,好ましくは哺乳動物細胞,特に,CHO細胞,NS/0細胞等の組換え蛋白質の製造に用いられる通常の細胞である。つまり,本発明の一実施形態は,SAとGALCとの組換え融合蛋白質,特にHSAとhGALCとの組換え融合蛋白質である。 When wild-type hGALC is expressed as a recombinant using a host cell transformed with an expression vector incorporating a gene encoding it, it is difficult to efficiently produce it as a recombinant due to reasons such as a decrease in the viability of the host cell. However, by expressing hGALC as a fusion protein with HSA, the decrease in the viability of the host cell observed when wild-type hGALC is expressed is suppressed. When a fusion protein of HSA and hGALC is expressed as a recombinant protein by culturing mammalian cells into which an expression vector incorporating a gene encoding it has been introduced in a serum-free medium, especially when the recombinant protein is expressed so that it is secreted from the cells and accumulated in the culture medium, the expression amount increases compared to when wild-type hGALC is expressed as a recombinant protein under the same conditions. It is considered that the suppression of the decrease in viability contributes to this increase in the expression amount. The same applies to a fusion protein of SA of an animal species other than human and hGALC, and a fusion protein of SA of an animal species other than human and GALC of an animal species other than human. The host cell used for expressing the fusion protein is preferably a mammalian cell, particularly a normal cell used for producing recombinant proteins such as CHO cells and NS/0 cells. That is, one embodiment of the present invention is a recombinant fusion protein between SA and GALC, in particular a recombinant fusion protein between HSA and hGALC.
 hGALC及びhGBAを発現させたときの宿主細胞の生存率の低下は,死細胞が増加することを意味する。死細胞からは,その内容物が流出するので,それらが夾雑物となり,発現した融合蛋白質の,その後の精製工程において,夾雑物を除去する工程が必要となる。つまり,hGALC及びhGBAは,HSAとの融合蛋白質の形態で発現させることにより,培養中に生じる死細胞数を減じて夾雑物を減少させることができるので,発現した融合蛋白質の精製を容易にすることができる。これにより精製時における精製効率を高めることもできる。また,精製後の融合蛋白質に含まれる夾雑物の量を減少させることもできる。 The decrease in the survival rate of host cells when hGALC and hGBA are expressed means that the number of dead cells increases. The contents of dead cells leak out, becoming contaminants, and a process for removing the contaminants is required in the subsequent purification process of the expressed fusion protein. In other words, by expressing hGALC and hGBA in the form of a fusion protein with HSA, the number of dead cells generated during culture can be reduced, and contaminants can be reduced, making it easier to purify the expressed fusion protein. This also increases the purification efficiency during purification. It also reduces the amount of contaminants contained in the purified fusion protein.
 HSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質をコードする宿主細胞を培養することにより,細胞内又は培地中に発現させることができる。これらの融合蛋白質は,カラムクロマトグラフィー等の方法により不純物から分離し,精製することができる。精製されたHSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質は,医薬組成物として使用することができる。特に,HSAとhGALCとの融合蛋白質は,クラッベ病(ガラクトシルセラミドリピドーシス,又はグロボイド細胞白質ジストロフィー)を対象疾患とする医薬組成物として使用することができる。また特に,HSAとhGBAとの融合蛋白質は,ゴーシェ病を対象疾患とする医薬組成物として使用することができる。なお,本明細書において医薬組成物というときは,有効成分としての融合蛋白質に加えて,薬学的に許容可能な賦形剤を含む組成物のことをいう。  By culturing a host cell encoding a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA, it can be expressed in cells or in a medium. These fusion proteins can be separated from impurities and purified by methods such as column chromatography. The purified fusion protein of HSA and hGALC or fusion protein of HSA and hGBA can be used as a pharmaceutical composition. In particular, the fusion protein of HSA and hGALC can be used as a pharmaceutical composition for treating Krabbe disease (galactosylceramide lipidosis or globoid cell leukodystrophy). In particular, the fusion protein of HSA and hGBA can be used as a pharmaceutical composition for treating Gaucher disease. In this specification, the term "pharmaceutical composition" refers to a composition that contains a fusion protein as an active ingredient as well as a pharma- ceutical acceptable excipient.
 HSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質を有効成分として含有してなる医薬組成物は,注射剤として静脈内,筋肉内,腹腔内,皮下又は脳室内に投与することができる。それらの注射剤は,凍結乾燥製剤又は水性液剤として供給することができる。水性液剤とする場合,バイアルに充填した形態としてもよく,注射器に予め充填したものであるプレフィルド型の製剤として供給することもできる。凍結乾燥製剤の場合,使用前に水性媒質に溶解し復元して使用する。水性液剤中に含まれるHSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質における,総蛋白質に占める単量体の比率(単量体の質量/総蛋白質の質量×100(%))は,好ましくは70%以上,より好ましくは80%以上,更に好ましくは90%以上,例えば95%以上であり95%以上である。凍結乾燥製剤を水性媒質に溶解し復元した溶液についても同じことがいえる。  A pharmaceutical composition containing the fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA as an active ingredient can be administered intravenously, intramuscularly, intraperitoneally, subcutaneously, or intracerebroventricularly as an injection. These injections can be supplied as freeze-dried preparations or aqueous liquid preparations. In the case of an aqueous liquid preparation, it may be in the form of being filled in a vial, or it can be supplied as a prefilled preparation in which it is filled in a syringe in advance. In the case of a freeze-dried preparation, it is dissolved in an aqueous medium before use and reconstituted for use. In the fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA contained in the aqueous liquid preparation, the ratio of monomer to total protein (monomer mass/total protein mass x 100 (%)) is preferably 70% or more, more preferably 80% or more, even more preferably 90% or more, for example 95% or more. The same can be said for a solution obtained by dissolving a freeze-dried preparation in an aqueous medium and reconstituting it.
 HSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質は,更に,抗体又はリガンドとの結合体とすることができる。例えば,本発明の一実施形態におけるHSAとhGALCとの融合蛋白質は,脳血管内皮細胞上の受容体と特異的に結合することのできる抗体又はリガンドとの結合体とすることができる。また例えば,本発明の一実施形態におけるHSAとhGBAとの融合蛋白質は,脳血管内皮細胞上の受容体と特異的に結合することのできる抗体又はリガンドとの結合体とすることができる。HSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質は,脳血管内皮細胞上の受容体と特異的に結合することのできる抗体又はリガンドとの結合体とすることにより,脳血管内皮細胞上の受容体に結合できるようになる。脳血管内皮細胞上の受容体に結合した融合蛋白質は,血液脳関門(BBB)を通過して,中枢神経系(CNS)の組織に到達できる。従って,HSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質は,かかる抗体又はリガンドとの結合体とすることで,血液脳関門(BBB)を通過させて,中枢神経系(CNS)において機能を発揮させるようにすることができる。 The fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA can be further bound to an antibody or a ligand. For example, the fusion protein of HSA and hGALC in one embodiment of the present invention can be bound to an antibody or a ligand capable of specifically binding to a receptor on cerebrovascular endothelial cells. For example, the fusion protein of HSA and hGBA in one embodiment of the present invention can be bound to an antibody or a ligand capable of specifically binding to a receptor on cerebrovascular endothelial cells. The fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA can bind to a receptor on cerebrovascular endothelial cells by being bound to an antibody or a ligand capable of specifically binding to a receptor on cerebrovascular endothelial cells. The fusion protein bound to the receptor on cerebrovascular endothelial cells can pass through the blood-brain barrier (BBB) and reach the tissues of the central nervous system (CNS). Therefore, by combining a fusion protein of HSA and hGALC or a fusion protein of HSA and hGBA with such an antibody or ligand, it is possible to pass through the blood-brain barrier (BBB) and exert its function in the central nervous system (CNS).
 本発明で「リガンド」というときは,特定の物質に親和性を有する物質,特に特定の物質に親和性を有す蛋白質のことをいう。本発明における一実施例において,かかるリガンドは,脳血管内皮細胞上に存在する受容体に対して親和性を有する物質,特に蛋白質である。脳血管内皮細胞上に存在する受容体としては,例えば,インスリン受容体,トランスフェリン受容体,レプチン受容体,リポ蛋白質受容体,及びIGF受容体であり,特にトランスフェリン受容体であるが,これらに限定されるものでない。また,当該受容体は好ましくはヒト由来の受容体である。インスリン受容体,トランスフェリン受容体,レプチン受容体,リポ蛋白質受容体,及びIGF受容体に対するリガンドは,それぞれ,インスリン,トランスフェリン,レプチン,リポ蛋白質,及びIGF(IGF-1及びIGF-2)である。これらリガンドは全長であっても,受容体に対する親和性を有する限り,その断片であってもよく,また野生型であっても,野生型に置換,欠失,又は/及び付加を導入した変異体であってもよい。 In the present invention, the term "ligand" refers to a substance having affinity for a specific substance, particularly a protein having affinity for a specific substance. In one embodiment of the present invention, such a ligand is a substance, particularly a protein, having affinity for a receptor present on cerebrovascular endothelial cells. Examples of receptors present on cerebrovascular endothelial cells include insulin receptors, transferrin receptors, leptin receptors, lipoprotein receptors, and IGF receptors, particularly, but not limited to, transferrin receptors. The receptors are preferably human-derived receptors. Ligands for insulin receptors, transferrin receptors, leptin receptors, lipoprotein receptors, and IGF receptors are insulin, transferrin, leptin, lipoprotein, and IGF (IGF-1 and IGF-2), respectively. These ligands may be full-length or fragments thereof as long as they have affinity for the receptors, and may be wild-type or mutants in which substitutions, deletions, and/or additions have been introduced into the wild-type.
 HSAとhGALCとの融合蛋白質と抗体との結合体において,hGALCがhGALCとしての機能を有するというときは,hGALCが,通常の野生型のhGALCの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性,例えば90%以上,95%以上の比活性を有することをいう。なお,ここで当該結合体におけるhGALCの比活性は,当該結合体の単位質量当たりのhGALCの酵素活性(μM/時間/mg蛋白質)に(当該結合体の分子量/当該結合体中のhGALCに相当する部分の分子量)を乗じて算出される。 When hGALC is said to have the function of hGALC in a conjugate of an antibody and a fusion protein of HSA and hGALC, it means that hGALC has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more, when the specific activity of normal wild-type hGALC is taken as 100%. Note that the specific activity of hGALC in the conjugate is calculated by multiplying the enzyme activity of hGALC per unit mass of the conjugate (μM/hour/mg protein) by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hGALC).
 また,HSAとhGBAとの融合蛋白質と抗体との結合体において,hGBAがhGBAとしての機能を有するというときは,hGBAが,通常の野生型のhGBAの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性,例えば90%以上,95%以上の比活性を有することをいう。なお,ここで当該結合体におけるhGBAの比活性は,当該結合体の単位質量当たりのhGBAの酵素活性(μM/時間/mg蛋白質)に(当該結合体の分子量/当該結合体中のhGBAに相当する部分の分子量)を乗じて算出される。 In addition, when hGBA in a conjugate of an antibody and a fusion protein of HSA and hGBA is said to have the function of hGBA, it means that, when the specific activity of normal wild-type hGBA is taken as 100%, hGBA has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more. Here, the specific activity of hGBA in the conjugate is calculated by multiplying the enzyme activity of hGBA per unit mass of the conjugate (μM/hour/mg protein) by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hGBA).
 本発明の一実施形態において,HSAとhGALCとの融合蛋白質と抗体との結合体というときは,当該結合体を組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhGALCを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhGALCとしての発現量が,濃度あるいは酵素活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍等となることを特徴とするものである。また,本発明の一実施形態において,HSAとhGBAとの融合蛋白質と抗体との結合体というときは,当該結合体を組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhGBAを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhGBAとしての発現量が,濃度あるいは酵素活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍等となることを特徴とするものである。 In one embodiment of the present invention, a conjugate of an antibody and a fusion protein of HSA and hGALC refers to a conjugate characterized in that when the conjugate is expressed as a recombinant protein in host cells, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hGALC in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, compared to when wild-type hGALC is expressed as a recombinant protein in host cells under the same conditions. Furthermore, in one embodiment of the present invention, a conjugate of an antibody and a fusion protein of HSA and hGBA is characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hGBA expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or enzyme activity, compared to when wild-type hGBA is expressed as a recombinant protein in a host cell under the same conditions, such as 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, etc.
 サイトカイン,特にインターロイキンの中には,野生型のものをコードする遺伝子を組込んだ発現ベクターを導入した宿主細胞を用いて組換え蛋白質として発現させた場合に,発現量が限定的であるため大量に製造することが困難なものがある。IL-10はそのようなインターロイキンの一つである。本発明の一実施形態は,このような組換え蛋白質として製造が困難なサイトカインを,SAとの融合蛋白質とした組換え蛋白質である。かかる組換え蛋白質は,これをコードする遺伝子を組込んだ発現ベクターを導入した宿主細胞を用いて,大量に製造することが,対応する野生型サイトカインと比較して,容易になる。ここで,SAと結合させるサイトカインの動物種に特に制限はないが,好ましくはヒトサイトカインである。また,サイトカインと結合させるSAの動物種に特に制限はないが,好ましくはHSAである。 Some cytokines, particularly interleukins, are difficult to mass-produce when expressed as recombinant proteins using host cells into which an expression vector incorporating a gene encoding the wild-type cytokine is introduced, because the expression level is limited. IL-10 is one such interleukin. One embodiment of the present invention is a recombinant protein in which such a cytokine that is difficult to produce as a recombinant protein is fused with SA. Such a recombinant protein can be mass-produced more easily than the corresponding wild-type cytokine using host cells into which an expression vector incorporating a gene encoding the cytokine is introduced. Here, there is no particular restriction on the animal species of the cytokine to be bound to SA, but it is preferably a human cytokine. There is also no particular restriction on the animal species of the SA to be bound to the cytokine, but it is preferably HSA.
 本明細書において,単に「ヒトサイトカイン」というときは,通常の野生型のヒトサイトカインに加え,当該ヒトサイトカインの種類に対応する生理活性を有する等のヒトサイトカインとしての機能を有するものである限り,野生型のヒトサイトカインのアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するヒトサイトカインの変異体も特に区別することなく包含する。ヒト以外の動物種のサイトカインについても同様である。また,インターロイキンについても同様である。 In this specification, when the term "human cytokine" is used simply, it includes not only normal wild-type human cytokines, but also mutant human cytokines in which one or more amino acid residues have been substituted, deleted, and/or added to the amino acid sequence of a wild-type human cytokine (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence), without any particular distinction, so long as the mutant has the function of a human cytokine, such as having physiological activity corresponding to the type of human cytokine in question. The same applies to cytokines of animal species other than humans. The same also applies to interleukins.
 なお,ここで,ヒトサイトカインがヒトサイトカインとしての機能を有するというときは,ヒトサイトカインが,通常の野生型のヒトサイトカインの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。ここで比活性とは蛋白質の質量当たりの生理活性のことをいう。なお,ヒトサイトカインとその他の蛋白質との融合蛋白質の比活性は,融合蛋白質の中でヒトサイトカインに相当する部分の質量当たりの生理活性として求められる。なお,ここで融合蛋白質におけるヒトサイトカインの比活性は,当該融合蛋白質の単位質量当たりのヒトサイトカインの生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のヒトサイトカインに相当する部分の分子量)を乗じて算出される。ヒト以外の動物種のサイトカインについても同様である。また,インターロイキンについても同様である。 When a human cytokine is said to have the function of a human cytokine, it means that the human cytokine preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of a normal wild-type human cytokine is taken as 100%. Here, specific activity refers to the physiological activity per mass of the protein. The specific activity of a fusion protein of a human cytokine and another protein is determined as the physiological activity per mass of the portion of the fusion protein that corresponds to the human cytokine. Here, the specific activity of the human cytokine in the fusion protein is calculated by multiplying the physiological activity of the human cytokine per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein that corresponds to the human cytokine). The same applies to cytokines of animal species other than humans. The same also applies to interleukins.
 野生型のヒトサイトカインのアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のヒトサイトカインのアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のヒトサイトカインのN末端又はC末端のアミノ酸残基を1個欠失させたアミノ酸配列からなるヒトサイトカインの変異体,野生型のヒトサイトカインのN末端又はC末端のアミノ酸残基を2個欠失させたアミノ酸配列からなるヒトサイトカインの変異体等も,ヒトサイトカインである。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型のヒトサイトカインのアミノ酸配列に加えることもできる。ヒト以外の動物種のサイトカインについても同様である。また,インターロイキンについても同様である。 When amino acid residues in the amino acid sequence of a wild-type human cytokine are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of a wild-type human cytokine are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. A human cytokine mutant consisting of an amino acid sequence in which one amino acid residue is deleted from the N-terminus or C-terminus of a wild-type human cytokine, a human cytokine mutant consisting of an amino acid sequence in which two amino acid residues are deleted from the N-terminus or C-terminus of a wild-type human cytokine, and the like are also human cytokines. In addition, a mutation that combines the substitution and deletion of these amino acid residues can be added to the amino acid sequence of a wild-type human cytokine. The same applies to cytokines of animal species other than humans. The same applies to interleukins.
 野生型のヒトサイトカインのアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はヒトサイトカインのアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型のヒトサイトカインのアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型のヒトサイトカインのアミノ酸配列に加えることもできる。ヒト以外の動物種のサイトカインについても同様である。また,インターロイキンについても同様である。 When adding amino acid residues to the amino acid sequence of a wild-type human cytokine, one or more amino acid residues are added into the amino acid sequence of the human cytokine or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of a wild-type human cytokine, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of a wild-type human cytokine. The same applies to cytokines of animal species other than humans. The same applies to interleukins.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型のヒトサイトカインのアミノ酸配列に加えることもできる。例えば,野生型のヒトサイトカインのアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもヒトサイトカインであり,野生型のヒトサイトカインのアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもヒトサイトカインであり,野生型のヒトサイトカインのアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもヒトサイトカインであり,野生型のヒトサイトカインのアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもヒトサイトカインであり,野生型のヒトサイトカインのアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもヒトサイトカインである。ヒト以外の動物種のサイトカインについても同様である。また,インターロイキンについても同様である。 Furthermore, a combination of the three types of mutations mentioned above, namely substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of a wild-type human cytokine. For example, a human cytokine is one in which 1 to 10 amino acid residues are deleted from the amino acid sequence of a wild-type human cytokine, 1 to 10 amino acid residues are substituted with other amino acid residues, and 1 to 10 amino acid residues are added; a human cytokine is one in which 1 to 5 amino acid residues are deleted from the amino acid sequence of a wild-type human cytokine, 1 to 5 amino acid residues are substituted with other amino acid residues, and 1 to 5 amino acid residues are added; a human cytokine is one in which 1 to 3 amino acid residues are deleted from the amino acid sequence of a wild-type human cytokine, 1 to 3 amino acid residues are substituted with other amino acid residues, and 1 to 3 amino acid residues are added; a human cytokine is one in which 1 or 2 amino acid residues are deleted from the amino acid sequence of a wild-type human cytokine, 1 or 2 amino acid residues are substituted with other amino acid residues, and 1 or 2 amino acid residues are added; and a human cytokine is one in which 1 amino acid residue is deleted from the amino acid sequence of a wild-type human cytokine, 1 amino acid residue is substituted with other amino acid residues, and 1 amino acid residue is added. The same is true for cytokines in non-human animal species, and also for interleukins.
 ヒトサイトカイン変異体における通常の野生型ヒトサイトカインと比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両ヒトサイトカインのアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種のサイトカインについても同様である。また,インターロイキンについても同様である。 The location and type (deletion, substitution, and addition) of each mutation in a human cytokine mutant compared to a normal wild-type human cytokine can be easily confirmed by aligning the amino acid sequences of both human cytokines. The same is true for cytokines of non-human animal species, and also for interleukins.
 ヒトサイトカイン変異体のアミノ酸配列は,通常の野生型ヒトサイトカインのアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。ヒト以外の動物種のサイトカインについても同様である。また,インターロイキンについても同様である。 The amino acid sequence of the human cytokine mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, to the amino acid sequence of a normal wild-type human cytokine. The same applies to cytokines of animal species other than humans. The same also applies to interleukins.
 本明細書において,単に「ヒトインターロイキン10」,「ヒトIL-10」,又は「hIL-10」というときは,配列番号24で示される160個のアミノ酸残基からなる通常の野生型のhIL-10に加え,免疫反応を鎮静化する抑制性サイトカインとしての生理活性を有する等のhIL-10としての機能を有するものである限り,配列番号24で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するhIL-10の変異体も特に区別することなく包含する。野生型のhIL-10は,例えば,配列番号49で示される塩基配列を有する遺伝子にコードされる。ヒト以外の動物種のIL-10についても同様である。 In this specification, when simply referring to "human interleukin 10", "human IL-10", or "hIL-10", it includes not only the normal wild-type hIL-10 consisting of 160 amino acid residues as shown in SEQ ID NO:24, but also includes, without any particular distinction, mutants of hIL-10 in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence as shown in SEQ ID NO:24, so long as they have the functions of hIL-10, such as having physiological activity as an inhibitory cytokine that calms the immune response. Wild-type hIL-10 is encoded, for example, by a gene having the base sequence as shown in SEQ ID NO:49. The same applies to IL-10 of animal species other than humans.
 なお,ここで,hIL-10がhIL-10としての機能を有するというときは,hIL-10が,通常の野生型のhIL-10の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。ここで比活性とは蛋白質の質量当たりの生理活性のことをいう。なお,hIL-10とその他の蛋白質との融合蛋白質の比活性は,融合蛋白質の中でhIL-10に相当する部分の質量当たりの生理活性として求められる。なお,ここで融合蛋白質におけるhIL-10の比活性は,当該融合蛋白質の単位質量当たりのhIL-10の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhIL-10に相当する部分の分子量)を乗じて算出される。ヒト以外の動物種のIL-10についても同様である。 Here, when hIL-10 is said to have the function of hIL-10, it means that hIL-10 retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hIL-10 is taken as 100%. Here, specific activity refers to the physiological activity per mass of the protein. The specific activity of a fusion protein of hIL-10 and another protein is calculated as the physiological activity per mass of the portion of the fusion protein that corresponds to hIL-10. Here, the specific activity of hIL-10 in the fusion protein is calculated by multiplying the physiological activity of hIL-10 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hIL-10). The same applies to IL-10 from animal species other than humans.
 野生型のhIL-10のアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhIL-10のアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhIL-10のN末端又はC末端のアミノ酸残基を1個欠失させた159個のアミノ酸残基からなるhIL-10の変異体,野生型のhIL-10のN末端又はC末端のアミノ酸残基を2個欠失させた158個のアミノ酸残基からなるhIL-10の変異体等も,hIL-10である。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型のhIL-10のアミノ酸配列に加えることもできる。ヒト以外の動物種のIL-10についても同様である。 When amino acid residues in the amino acid sequence of wild-type hIL-10 are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of wild-type hIL-10 are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. A mutant hIL-10 consisting of 159 amino acid residues obtained by deleting one amino acid residue at the N-terminus or C-terminus of wild-type hIL-10, and a mutant hIL-10 consisting of 158 amino acid residues obtained by deleting two amino acid residues at the N-terminus or C-terminus of wild-type hIL-10, etc. are also hIL-10. In addition, a mutation that combines the substitution and deletion of these amino acid residues can be added to the amino acid sequence of wild-type hIL-10. The same applies to IL-10 of animal species other than humans.
 野生型のhIL-10のアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はhIL-10のアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型のhIL-10のアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型のhIL-10のアミノ酸配列に加えることもできる。ヒト以外の動物種のIL-10についても同様である。 When amino acid residues are added to the amino acid sequence of wild-type hIL-10, one or more amino acid residues are added into the amino acid sequence of hIL-10 or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hIL-10, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hIL-10. The same applies to IL-10 from animal species other than humans.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型のhIL-10のアミノ酸配列に加えることもできる。例えば,配列番号24で示される野生型のhIL-10のアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもhIL-10であり,配列番号24で示される野生型のhIL-10のアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもhIL-10であり,配列番号24で示される野生型のhIL-10のアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもhIL-10であり,配列番号24で示される野生型のhIL-10のアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもhIL-10であり,配列番号24で示される野生型のhIL-10のアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもhIL-10である。ヒト以外の動物種のIL-10についても同様である。 Furthermore, a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of wild-type hIL-10. For example, a wild-type hIL-10 amino acid sequence shown in SEQ ID NO:24 in which 1 to 10 amino acid residues have been deleted, 1 to 10 amino acid residues have been replaced with other amino acid residues, and 1 to 10 amino acid residues have been added is also hIL-10, a wild-type hIL-10 amino acid sequence shown in SEQ ID NO:24 in which 1 to 5 amino acid residues have been deleted, 1 to 5 amino acid residues have been replaced with other amino acid residues, and 1 to 5 amino acid residues have been added is also hIL-10, and a wild-type hIL-10 amino acid sequence shown in SEQ ID NO:24 in which 1 to 3 amino acid residues have been deleted, hIL-10 also includes the wild-type hIL-10 amino acid sequence shown in SEQ ID NO:24 in which one or two amino acid residues have been deleted, one or two amino acid residues have been replaced with other amino acid residues, and one or two amino acid residues have been added. hIL-10 also includes the wild-type hIL-10 amino acid sequence shown in SEQ ID NO:24 in which one amino acid residue has been deleted, one amino acid residue has been replaced with other amino acid residue, and one amino acid residue has been added. The same applies to IL-10 from animal species other than humans.
 hIL-10変異体における通常の野生型hIL-10と比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両hIL-10のアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種のIL-10についても同様である。 The location and type (deletion, substitution, and addition) of each mutation in the hIL-10 mutant compared to normal wild-type hIL-10 can be easily confirmed by aligning the amino acid sequences of both hIL-10. The same is true for IL-10 from animal species other than humans.
 hIL-10変異体のアミノ酸配列は,配列番号24で示される通常の野生型hIL-10のアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。ヒト以外の動物種のIL-10についても同様である。 The amino acid sequence of the hIL-10 mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type hIL-10 shown in SEQ ID NO: 24. The same applies to IL-10 of animal species other than humans.
 野生型ヒトサイトカインのアミノ酸配列中のアミノ酸の他のアミノ酸への置換,野生型ヒトサイトカインのアミノ酸配列中のアミノ酸の他のアミノ酸への置換,及び野生型hIL-10のアミノ酸配列中のアミノ酸の他のアミノ酸への置換は,好ましくは保存的アミノ酸置換である。ヒト以外の動物種のサイトカイン,インターロイキン及びIL-10についても同様である。 The substitution of an amino acid in the amino acid sequence of a wild-type human cytokine with another amino acid, the substitution of an amino acid in the amino acid sequence of a wild-type human cytokine with another amino acid, and the substitution of an amino acid in the amino acid sequence of a wild-type hIL-10 with another amino acid are preferably conservative amino acid substitutions. The same applies to cytokines, interleukins, and IL-10 of animal species other than human.
 上記の野生型又は変異型のヒトサイトカインであって,これを構成するアミノ酸が糖鎖により修飾されたものもヒトサイトカインである。また,上記の野生型又は変異型のヒトサイトカインであって,これを構成するアミノ酸がリン酸により修飾されたものもヒトサイトカインである。また,糖鎖及びリン酸以外のものにより修飾されたものもヒトサイトカインである。また,上記の野生型又は変異型のヒトサイトカインであって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもヒトサイトカインである。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のサイトカインについても同様である。また,インターロイキン及びIL-10についても同様である。  The above wild-type or mutant human cytokines in which the constituent amino acids are modified with sugar chains are also human cytokines. The above wild-type or mutant human cytokines in which the constituent amino acids are modified with phosphate are also human cytokines. Those modified with something other than sugar chains and phosphate are also human cytokines. The above wild-type or mutant human cytokines in which the side chains of the constituent amino acids have been converted by substitution reactions or the like are also human cytokines. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same applies to cytokines of animal species other than humans. The same applies to interleukins and IL-10.
 つまり,糖鎖により修飾されたヒトサイトカインは,元のアミノ酸配列を有するヒトサイトカインに含まれるものとする。また,リン酸により修飾されたヒトサイトカインは,元のアミノ酸配列を有するヒトサイトカインに含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するヒトサイトカインに含まれるものとする。また,ヒトサイトカインを構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するヒトサイトカインに含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のサイトカインについても同様である。また,インターロイキン及びIL-10についても同様である。 In other words, human cytokines modified with sugar chains are included in human cytokines with the original amino acid sequence. Human cytokines modified with phosphate are included in human cytokines with the original amino acid sequence. Cytokines modified with substances other than sugar chains and phosphate are also included in human cytokines with the original amino acid sequence. Cytokines in which the side chains of the amino acids that make up human cytokines have been changed by substitution reactions or the like are also included in human cytokines with the original amino acid sequence. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same applies to cytokines of animal species other than humans. The same applies to interleukins and IL-10.
 本発明の一実施形態において,野生型のヒトサイトカインは,HSAと融合させた組換え蛋白質として製造される。かかる融合蛋白質は,CHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型ヒトサイトカインを同様の手法により組換え蛋白質として発現させたときと比較して,培養上清中のヒトサイトカインとしての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,3~4倍等となるようにすることができる。 In one embodiment of the present invention, a wild-type human cytokine is produced as a recombinant protein fused with HSA. When such a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of expression of the human cytokine in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when the wild-type human cytokine is expressed as a recombinant protein by a similar method. For example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 3 to 4 times, etc.
 また,本発明の一実施形態において,hIL-10が,HSAと融合させた組換え蛋白質として製造される。かかる融合蛋白質は,CHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型hIL-10を同様の手法により組換え蛋白質として発現させたときと比較して,培養上清中のhIL-10としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,3~4倍等となるようにすることができる。 In one embodiment of the present invention, hIL-10 is produced as a recombinant protein fused with HSA. When such a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of expression of hIL-10 in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hIL-10 is expressed as a recombinant protein by a similar method. For example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 3 to 4 times, etc.
 つまり,本発明の一実施形態は,サイトカインのアミノ酸配列を含むポリペプチドと,SAのアミノ酸配列を含むポリペプチドとを結合させた,融合蛋白質である。ここで,「ポリペプチドを結合させる」というときは,直接又はリンカーを介して間接的に,共有結合により異なるポリペプチドを結合させることをいう。ここで,サイトカイン及びSAは好ましくはヒト由来のものである。 In other words, one embodiment of the present invention is a fusion protein in which a polypeptide containing the amino acid sequence of a cytokine is bound to a polypeptide containing the amino acid sequence of an SA. Here, "binding polypeptides" refers to binding different polypeptides by covalent bonding, either directly or indirectly via a linker. Here, the cytokine and SA are preferably of human origin.
 また,本発明の一実施形態は,IL-10のアミノ酸配列を含むポリペプチドと,SAのアミノ酸配列を含むポリペプチドとを結合させた,融合蛋白質である。ここで,「ポリペプチドを結合させる」というときは,直接又はリンカーを介して間接的に,共有結合により異なるポリペプチドを結合させることをいう。ここで,IL-10及びSAは好ましくはヒト由来のものである。 Another embodiment of the present invention is a fusion protein in which a polypeptide containing the amino acid sequence of IL-10 is bound to a polypeptide containing the amino acid sequence of SA. Here, the term "binding polypeptides" refers to binding different polypeptides by covalent bonding, either directly or indirectly via a linker. Here, IL-10 and SA are preferably of human origin.
 2つの異なるポリペプチドを結合させる方法としては,例えば,一方のポリペプチドをコードする遺伝子の下流に,インフレームで他方のポリペプチドをコードする遺伝子を結合させたDNA断片を組み込んだ発現ベクターを作製し,この発現ベクターを用いて形質転換させた宿主細胞を培養することにより,組換え蛋白質として発現させる方法が一般的である。得られた組換え蛋白質は,2つのポリペプチドが,直接又は別のアミノ酸配列を介してペプチド結合した,一本鎖ポリペプチドである。 A common method for linking two different polypeptides is, for example, to create an expression vector incorporating a DNA fragment in which a gene encoding one polypeptide is linked in-frame downstream of a gene encoding the other polypeptide, and then to express the resulting protein as a recombinant protein by culturing a host cell transformed with this expression vector. The resulting recombinant protein is a single-chain polypeptide in which the two polypeptides are peptide-linked, either directly or via different amino acid sequences.
 本発明の一実施形態において,「SA-ヒトサイトカイン融合蛋白質」又は「SA-ヒトサイトカイン」の語は,SAのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒトサイトカインのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒトサイトカインとしての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。SA-ヒトサイトカイン融合蛋白質において,ヒトサイトカインがヒトサイトカインとしての機能を有するというときは,ヒトサイトカインが,通常の野生型のヒトサイトカインの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでSA-ヒトサイトカイン融合蛋白質におけるヒトサイトカインの比活性は,当該融合蛋白質の単位質量当たりのヒトサイトカインの生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のヒトサイトカインに相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "SA-human cytokine fusion protein" or "SA-human cytokine" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of a human cytokine is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and which has the function of a human cytokine. Here, the animal species of the SA is not particularly limited, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA. When it is said that a human cytokine in an SA-human cytokine fusion protein has the function of a human cytokine, it means that the human cytokine has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of a normal wild-type human cytokine is taken as 100%. Here, the specific activity of the human cytokine in the SA-human cytokine fusion protein is calculated by multiplying the physiological activity of the human cytokine per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to the human cytokine).
 本発明の一実施形態において,「HSA-ヒトサイトカイン融合蛋白質」又は「HSA-ヒトサイトカイン」の語は,HSAのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒトサイトカインのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒトサイトカインとしての機能を有するものを示す。ここでHSA-ヒトサイトカイン融合蛋白質がヒトサイトカインとしての機能を有するというときは,上記のSA-ヒトサイトカイン融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "HSA-human cytokine fusion protein" or "HSA-human cytokine" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of a human cytokine is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of a human cytokine. When it is said here that the HSA-human cytokine fusion protein has the function of a human cytokine, the definition of the SA-human cytokine fusion protein described above can be applied.
 SA-ヒトサイトカイン融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。HSA-ヒトリソソーム酵素融合蛋白質においても同様である。 In the SA-human cytokine fusion protein, it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this. The same is true for the HSA-human lysosomal enzyme fusion protein.
 野生型HSAと野生型ヒトサイトカインとの融合蛋白質であるHSA-ヒトサイトカイン融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えヒトサイトカイン部分には変異を加えないことも,HSA部分には変異を加えずにヒトサイトカイン部分にのみ変異を加えることも,また,HSA部分とヒトサイトカイン部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。ヒトサイトカイン部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型ヒトサイトカインに変異が加えられたヒトサイトカインのアミノ酸配列となる。HSA部分とヒトサイトカイン部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,ヒトサイトカイン部分のアミノ酸配列は上述した野生型ヒトサイトカインに変異が加えられたヒトサイトカインのアミノ酸配列となる。ヒト以外の動物種の野生型のSAとヒトサイトカインとの融合蛋白質についても同様のことがいえる。 When mutations are added to an HSA-human cytokine fusion protein, which is a fusion protein of wild-type HSA and wild-type human cytokine, mutations can be added only to the HSA portion and not to the human cytokine portion, mutations can be added only to the human cytokine portion without mutations to the HSA portion, or mutations can be added to both the HSA portion and the human cytokine portion. When mutations are added only to the HSA portion, the amino acid sequence of the portion is the amino acid sequence of the wild-type HSA with a mutation added. When mutations are added only to the human cytokine portion, the amino acid sequence of the portion is the amino acid sequence of the wild-type human cytokine with a mutation added. When mutations are added to both the HSA portion and the human cytokine portion, the amino acid sequence of the HSA portion is the amino acid sequence of the wild-type HSA with a mutation added, and the amino acid sequence of the human cytokine portion is the amino acid sequence of the wild-type human cytokine with a mutation added. The same can be said for fusion proteins of wild-type SA of animal species other than humans and human cytokine.
 HSA-ヒトサイトカイン融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもHSA-ヒトサイトカイン融合蛋白質である。また,HSA-ヒトサイトカイン融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもHSA-ヒトサイトカイン融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもHSA-ヒトサイトカイン融合蛋白質である。また,HSA-ヒトサイトカイン融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSA-ヒトサイトカイン融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとヒトサイトカインとの融合蛋白質(SA-ヒトサイトカイン)についても同様のことがいえる。  HSA-human cytokine fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-human cytokine fusion proteins. HSA-human cytokine fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-human cytokine fusion proteins. HSA-human cytokine fusion proteins modified with something other than sugar chains and phosphate are also HSA-human cytokine fusion proteins. HSA-human cytokine fusion proteins in which the side chains of the amino acids constituting the protein have been converted by substitution reactions or the like are also HSA-human cytokine fusion proteins. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA of animal species other than humans and human cytokines (SA-human cytokine).
 つまり,糖鎖により修飾されたHSA-ヒトサイトカイン融合蛋白質は,元のアミノ酸配列を有するHSA-ヒトサイトカイン融合蛋白質に含まれるものとする。また,リン酸により修飾されたHSA-ヒトサイトカイン融合蛋白質は,元のアミノ酸配列を有するHSA-ヒトサイトカイン融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSA-ヒトサイトカイン融合蛋白質に含まれるものとする。また,HSA-ヒトサイトカイン融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSA-ヒトサイトカイン融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。その他の動物のSAとヒトサイトカインとの融合蛋白質(SA-ヒトサイトカイン)についても同様のことがいえる。ヒト以外の動物種のSAとヒトサイトカインとの融合蛋白質(SA-ヒトサイトカイン)についても同様のことがいえる。 In other words, HSA-human cytokine fusion proteins modified with sugar chains are included in HSA-human cytokine fusion proteins having the original amino acid sequence. Also, HSA-human cytokine fusion proteins modified with phosphate are included in HSA-human cytokine fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are also included in HSA-human cytokine fusion proteins having the original amino acid sequence. Also, those in which the side chains of the amino acids constituting the HSA-human cytokine fusion proteins have been converted by substitution reactions or the like are also included in HSA-human cytokine fusion proteins having the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA of other animals and human cytokines (SA-human cytokines). The same can be said for fusion proteins of SA of animal species other than humans and human cytokines (SA-human cytokines).
 また,HSA-ヒトサイトカイン融合蛋白質であって,これを構成するヒトサイトカインが,ヒトサイトカインの前駆体であるものもHSA-ヒトサイトカイン融合蛋白質である。ここで前駆体というときは,HSA-ヒトサイトカイン融合蛋白質として生合成されたものであって,当該生合成後に,ヒトサイトカインとして機能する部分が,製造過程において又は生体に投与したときに当該生体内において,当該融合蛋白質から切り離されて,単独でもヒトサイトカインとしての機能を発揮することができるタイプのものをいう。この場合,HSA-ヒトサイトカイン融合蛋白質は生合成された後に,加水分解酵素等により,特定の箇所で切断され,HSAを含む部分と成熟型のヒトサイトカインを含む部分とが分離することがある。この場合,得られるヒトサイトカインはHSAとの融合蛋白質ではないが,当該サイトカインが製造される過程で,一旦,HSA-ヒトサイトカイン融合蛋白質が合成されることになる。従って,かかる方法によりヒトサイトカインを製造する場合,当該製造方法は,HSA-ヒトサイトカインの製造方法に含まれる。ヒト以外の動物種のSAとヒトサイトカインとの融合蛋白質(SA-ヒトサイトカイン),及びヒト以外の動物種のSAとヒト以外の動物種のサイトカインとの融合蛋白質,例えばMSA-マウスサイトカイン融合蛋白質についても同様のことがいえる。  Also, HSA-human cytokine fusion proteins in which the human cytokine that constitutes it is a precursor of the human cytokine are also HSA-human cytokine fusion proteins. The term "precursor" here refers to a type that is biosynthesized as an HSA-human cytokine fusion protein, and after the biosynthesis, the part that functions as a human cytokine is separated from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as a human cytokine by itself. In this case, after the HSA-human cytokine fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the part containing HSA and the part containing the mature human cytokine may be separated. In this case, the human cytokine obtained is not a fusion protein with HSA, but the HSA-human cytokine fusion protein is synthesized once during the process of manufacturing the cytokine. Therefore, when human cytokine is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-human cytokine. The same can be said for a fusion protein of SA of an animal species other than human and a human cytokine (SA-human cytokine), and a fusion protein of SA of an animal species other than human and a cytokine of an animal species other than human, such as MSA-mouse cytokine fusion protein.
 本発明の一実施形態において,「SA-hIL-10融合蛋白質」又は「SA-hIL-10」の語は,SAのアミノ酸配列のC末端に,直接又はリンカーを介して,hIL-10のアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hIL-10としての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。SA-hIL-10融合蛋白質において,hIL-10がhIL-10としての機能を有するというときは,hIL-10が,通常の野生型のhIL-10の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでSA-hIL-10融合蛋白質におけるhIL-10の比活性は,当該融合蛋白質の単位質量当たりのhIL-10の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhIL-10に相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "SA-hIL-10 fusion protein" or "SA-hIL-10" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hIL-10 is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and which has the function of hIL-10. Here, there is no particular limitation on the animal species of the SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA. When it is said that hIL-10 in the SA-hIL-10 fusion protein has the function of hIL-10, it means that hIL-10 retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hIL-10 is taken as 100%. Here, the specific activity of hIL-10 in the SA-hIL-10 fusion protein is calculated by multiplying the physiological activity of hIL-10 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hIL-10).
 本発明の一実施形態において,「HSA-hIL-10融合蛋白質」又は「HSA-hIL-10」の語は,HSAのアミノ酸配列のC末端に,直接又はリンカーを介して,hIL-10のアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hIL-10としての機能を有するものを示す。ここでHSA-hIL-10融合蛋白質がhIL-10としての機能を有するというときは,上記のSA-hIL-10融合蛋白質における定義を適用することができる。また,これら融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, the term "HSA-hIL-10 fusion protein" or "HSA-hIL-10" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hIL-10 is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hIL-10. When it is said that the HSA-hIL-10 fusion protein has the function of hIL-10, the definition of the SA-hIL-10 fusion protein described above can be applied. Furthermore, in these fusion proteins, it is preferable that the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited to this.
 本発明の一実施形態において好ましいHSA-hIL-10融合蛋白質は,例えば配列番号50で示されるアミノ酸配列を有するものである。配列番号50で示されるHSA-hIL-10融合蛋白質は,野生型HSAのC末端に直接野生型hIL-10が結合したものである。配列番号50で示されるHSA-hIL-10融合蛋白質は,例えば,配列番号51で示される塩基配列を有する遺伝子にコードされる。また,配列番号50で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hIL-10としての機能を有するものである限りHSA-hIL-10融合蛋白質に含まれる。HSA-hIL-10融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred HSA-hIL-10 fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 50. The HSA-hIL-10 fusion protein shown in SEQ ID NO: 50 is a protein in which wild-type hIL-10 is directly bound to the C-terminus of wild-type HSA. The HSA-hIL-10 fusion protein shown in SEQ ID NO: 50 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 51. In addition, HSA-hIL-10 fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 50 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hIL-10. It is preferable that the HSA-hIL-10 fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 配列番号50で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号50で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:50 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:50.
 野生型HSAと野生型hIL-10との融合蛋白質であるHSA-hIL-10融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhIL-10部分には変異を加えないことも,HSA部分には変異を加えずにhIL-10部分にのみ変異を加えることも,また,HSA部分とhIL-10部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hIL-10部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hIL-10に変異が加えられたhIL-10のアミノ酸配列となる。HSA部分とhIL-10部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hIL-10部分のアミノ酸配列は上述した野生型hIL-10に変異が加えられたhIL-10のアミノ酸配列となる。配列番号50で示されるアミノ酸配列を有するHSA-hIL-10融合蛋白質についても同様である。ヒト以外の動物種の野生型SAと野生型hIL-10との融合蛋白質についても同様のことがいえる。 When mutations are added to an HSA-hIL-10 fusion protein, which is a fusion protein of wild-type HSA and wild-type hIL-10, mutations can be added only to the HSA portion and not to the hIL-10 portion, mutations can be added only to the hIL-10 portion without mutations to the HSA portion, or mutations can be added to both the HSA portion and the hIL-10 portion. When mutations are added only to the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are added only to the hIL-10 portion, the amino acid sequence of the portion is the amino acid sequence of hIL-10 obtained by adding a mutation to the wild-type hIL-10 described above. When mutations are added to both the HSA portion and the hIL-10 portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hIL-10 portion is the amino acid sequence of hIL-10 obtained by adding a mutation to the wild-type hIL-10 described above. The same is true for the HSA-hIL-10 fusion protein having the amino acid sequence shown in SEQ ID NO: 50. The same is true for a fusion protein of wild-type SA and wild-type hIL-10 of an animal species other than human.
 配列番号50で示されるアミノ酸配列を有するHSA-hIL-10融合蛋白質に変異を加える場合について,以下例示する。配列番号50で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号50で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。HSA-hIL-10融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hIL-10部分にのみ加えても,又は両方の部分に加えてもよい。 The following is an example of a case where a mutation is made to an HSA-hIL-10 fusion protein having the amino acid sequence shown in SEQ ID NO:50. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:50 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:50 or to the N-terminus or C-terminus of the amino acid sequence. The HSA-hIL-10 fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA part, only to the hIL-10 part, or to both parts.
 配列番号50で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号50で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:50 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:50.
 HSA-hIL-10融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもHSA-hIL-10融合蛋白質である。また,HSA-hIL-10融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもHSA-hIL-10融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもHSA-hIL-10融合蛋白質である。また,HSA-hIL-10融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSA-hIL-10融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。その他の動物のSAとhIL-10との融合蛋白質(SA-hIL-10)についても同様のことがいえる。ヒト以外の動物種のSAとhIL-10との融合蛋白質(SA-hIL-10)についても同様のことがいえる。  HSA-hIL-10 fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hIL-10 fusion proteins. HSA-hIL-10 fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hIL-10 fusion proteins. HSA-hIL-10 fusion proteins modified with anything other than sugar chains and phosphate are also HSA-hIL-10 fusion proteins. HSA-hIL-10 fusion proteins in which the side chains of the amino acids constituting the protein are changed by substitution reactions, etc. Examples of such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hIL-10 of other animals (SA-hIL-10). The same can be said for fusion proteins of SA and hIL-10 of animal species other than humans (SA-hIL-10).
 つまり,糖鎖により修飾されたHSA-hIL-10融合蛋白質は,元のアミノ酸配列を有するHSA-hIL-10融合蛋白質に含まれるものとする。また,リン酸により修飾されたHSA-hIL-10融合蛋白質は,元のアミノ酸配列を有するHSA-hIL-10融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSA-hIL-10融合蛋白質に含まれるものとする。また,HSA-hIL-10融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSA-hIL-10融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhIL-10との融合蛋白質(SA-hIL-10)についても同様のことがいえる。 In other words, HSA-hIL-10 fusion proteins modified with sugar chains are included in HSA-hIL-10 fusion proteins having the original amino acid sequence. Also, HSA-hIL-10 fusion proteins modified with phosphate are included in HSA-hIL-10 fusion proteins having the original amino acid sequence. Also, those modified with something other than sugar chains and phosphate are included in HSA-hIL-10 fusion proteins having the original amino acid sequence. Also, HSA-hIL-10 fusion proteins in which the side chains of the amino acids constituting the HSA-hIL-10 fusion proteins have been changed by substitution reactions or the like are included in HSA-hIL-10 fusion proteins having the original amino acid sequence. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hIL-10 of animal species other than humans (SA-hIL-10).
 また,HSA-hIL-10融合蛋白質であって,これを構成するhIL-10が,hIL-10の前駆体であるものもHSA-hIL-10融合蛋白質である。ここで前駆体というときは,HSA-hIL-10融合蛋白質として生合成されたものであって,当該生合成後に,hIL-10として機能する部分が,製造過程において又は生体に投与したときに当該生体内において,当該融合蛋白質から切り離されて,単独でもhIL-10としての機能を発揮することができるタイプのものをいう。この場合,HSA-hIL-10融合蛋白質は生合成された後に,加水分解酵素等により,特定の箇所で切断され,HSAを含む部分と成熟型のhIL-10を含む部分が分離することがある。この場合,得られるhIL-10はHSAとの融合蛋白質ではないが,当該hIL-10が合成される過程で,一旦,HSA-hIL-10融合蛋白質が合成されることになる。従って,かかる方法によりhIL-10を製造する場合,当該製造方法は,HSA-hIL-10融合蛋白質の製造方法に含まれる。ヒト以外の動物種のSAとhIL-10との融合蛋白質(SA-hIL-10)についても同様のことがいえる。  Also, HSA-hIL-10 fusion proteins in which the constituent hIL-10 is a precursor of hIL-10 are also HSA-hIL-10 fusion proteins. Here, the term "precursor" refers to a type that is biosynthesized as an HSA-hIL-10 fusion protein, and after the biosynthesis, the part that functions as hIL-10 is cleaved from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hIL-10 by itself. In this case, after the HSA-hIL-10 fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the part containing HSA and the part containing mature hIL-10 may be separated. In this case, the obtained hIL-10 is not a fusion protein with HSA, but the HSA-hIL-10 fusion protein is synthesized once during the process of synthesizing the hIL-10. Therefore, when hIL-10 is produced by such a method, the production method is included in the method for producing HSA-hIL-10 fusion protein. The same can be said about the fusion protein of SA and hIL-10 from non-human animal species (SA-hIL-10).
 本発明の一実施形態において,「ヒトサイトカイン-SA融合蛋白質」又は「ヒトサイトカイン-SA」の語は,ヒトサイトカインのアミノ酸配列のC末端に,直接又はリンカーを介して,SAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒトサイトカインとしての機能を有するものを示す。ヒトサイトカイン-SA融合蛋白質において,ヒトサイトカインがヒトサイトカインとしての機能を有するというときは,ヒトサイトカインが,通常の野生型のヒトサイトカインの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでヒトサイトカイン-SA融合蛋白質におけるヒトサイトカインの比活性は,当該融合蛋白質の単位質量当たりのヒトサイトカインの生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のヒトサイトカインに相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "human cytokine-SA fusion protein" or "human cytokine-SA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of a human cytokine, and which has a function as a human cytokine. When a human cytokine in a human cytokine-SA fusion protein is said to have a function as a human cytokine, it means that the human cytokine retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of a normal wild-type human cytokine is taken as 100%. Here, the specific activity of the human cytokine in the human cytokine-SA fusion protein is calculated by multiplying the physiological activity of the human cytokine per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to the human cytokine).
 本発明の一実施形態において,「ヒトサイトカイン-HSA融合蛋白質」又は「ヒトサイトカイン-HSA」の語は,ヒトサイトカインのアミノ酸配列のC末端に,直接又はリンカーを介して,HSAのアミノ酸配列のN末端が結合されている,アミノ酸配列を有する融合蛋白質であって,ヒトサイトカインとしての機能を有するものを示す。ここでヒトサイトカイン-HSA融合蛋白質がヒトサイトカインとしての機能を有するというときは,上記のヒトサイトカイン-SAにおける定義を採用することができる。 In one embodiment of the present invention, the term "human cytokine-HSA fusion protein" or "human cytokine-HSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of a human cytokine, and which has the function of a human cytokine. When it is said that the human cytokine-HSA fusion protein has the function of a human cytokine, the definition of human cytokine-SA above can be adopted.
 ヒトサイトカイン-SA融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。ヒトサイトカイン-HSA融合蛋白質においても同様である。 In the human cytokine-SA fusion protein, it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this. The same is true for the human cytokine-HSA fusion protein.
 野生型ヒトサイトカインと野生型HSAとの融合蛋白質であるヒトサイトカイン-HSA融合蛋白質に変異を加える場合,ヒトサイトカイン部分にのみ変異を加えHSA部分には変異を加えないことも,ヒトサイトカイン部分には変異を加えずにHSA部分にのみ変異を加えることも,また,ヒトサイトカイン部分とHSA部分の何れにも変異を加えることもできる。ヒトサイトカイン部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型ヒトサイトカインに変異が加えられたヒトサイトカインのアミノ酸配列となる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。ヒトサイトカイン部分とHSA部分の何れにも変異を加える場合にあっては,ヒトサイトカイン部分のアミノ酸配列は上述した野生型ヒトサイトカインに変異が加えられたヒトサイトカインのアミノ酸配列となり,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。野生型ヒトサイトカインとそのヒト以外の動物種の野生型SAとの融合蛋白質についても同様のことがいえる。 When mutations are introduced into a human cytokine-HSA fusion protein, which is a fusion protein of a wild-type human cytokine and a wild-type HSA, mutations can be introduced only into the human cytokine portion and not into the HSA portion, mutations can be introduced only into the HSA portion without mutations in the human cytokine portion, or mutations can be introduced into both the human cytokine portion and the HSA portion. When mutations are introduced only into the human cytokine portion, the amino acid sequence of the portion is the amino acid sequence of the human cytokine obtained by mutating the wild-type human cytokine described above. When mutations are introduced only into the HSA portion, the amino acid sequence of the portion is the amino acid sequence of the HSA obtained by mutating the wild-type HSA described above. When mutations are introduced into both the human cytokine portion and the HSA portion, the amino acid sequence of the human cytokine portion is the amino acid sequence of the human cytokine obtained by mutating the wild-type human cytokine described above, and the amino acid sequence of the HSA portion is the amino acid sequence of the HSA obtained by mutating the wild-type HSA described above. The same can be said about a fusion protein between a wild-type human cytokine and the wild-type SA of an animal species other than human.
 ヒトサイトカイン-HSA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもヒトサイトカイン-HSA融合蛋白質である。また,ヒトサイトカイン-HSA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもヒトサイトカイン-HSA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもヒトサイトカイン-HSA融合蛋白質である。また,ヒトサイトカイン-HSA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもヒトサイトカイン-HSA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒトサイトカインとヒト以外の動物種のSAとの融合蛋白質(ヒトサイトカイン-SA)についても同様のことがいえる。  A human cytokine-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also a human cytokine-HSA fusion protein. A human cytokine-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also a human cytokine-HSA fusion protein. A human cytokine-HSA fusion protein modified with something other than sugar chains and phosphate is also a human cytokine-HSA fusion protein. A human cytokine-HSA fusion protein in which the side chains of the amino acids constituting the protein have been modified by a substitution reaction or the like is also a human cytokine-HSA fusion protein. Such a conversion includes, but is not limited to, the conversion of a cysteine residue to formylglycine. The same can be said about a fusion protein of a human cytokine and SA of an animal species other than human (human cytokine-SA).
 つまり,糖鎖により修飾されたヒトサイトカイン-HSA融合蛋白質は,元のアミノ酸配列を有するヒトサイトカイン-HSA融合蛋白質に含まれるものとする。また,リン酸により修飾されたヒトサイトカイン-HSA融合蛋白質は,元のアミノ酸配列を有するヒトサイトカイン-HSA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するヒトサイトカイン-HSA融合蛋白質に含まれるものとする。また,ヒトサイトカイン-HSA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するヒトサイトカイン-HSA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒトサイトカインとヒト以外の動物種のSAとの融合蛋白質(ヒトサイトカイン-SA)についても同様のことがいえる。 In other words, a human cytokine-HSA fusion protein modified with a sugar chain is included in the human cytokine-HSA fusion protein having the original amino acid sequence. A human cytokine-HSA fusion protein modified with phosphate is included in the human cytokine-HSA fusion protein having the original amino acid sequence. A human cytokine-HSA fusion protein modified with something other than a sugar chain or phosphate is also included in the human cytokine-HSA fusion protein having the original amino acid sequence. A human cytokine-HSA fusion protein in which the side chains of the amino acids constituting the human cytokine-HSA fusion protein have been changed by a substitution reaction or the like is also included in the human cytokine-HSA fusion protein having the original amino acid sequence. Such a change includes, but is not limited to, the change of a cysteine residue to formylglycine. The same can be said about a fusion protein of a human cytokine and SA of an animal species other than human (human cytokine-SA).
 また,ヒトサイトカイン-HSA融合蛋白質であって,これを構成するヒトサイトカインが,ヒトサイトカインの前駆体であるものもヒトサイトカイン-HSA融合蛋白質である。ヒトサイトカインとヒト以外の動物種のSAとの融合蛋白質(ヒトサイトカイン-SA)についても同様のことがいえる。  Furthermore, a human cytokine-HSA fusion protein in which the constituent human cytokine is a precursor of a human cytokine is also a human cytokine-HSA fusion protein. The same can be said for a fusion protein of a human cytokine and SA of an animal species other than human (human cytokine-SA).
 本発明の一実施形態において,「hIL-10-SA融合蛋白質」又は「hIL-10-SA」の語は,hIL-10のアミノ酸配列のC末端に,直接又はリンカーを介して,SAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hIL-10としての機能を有するものを示す。hIL-10-SA融合蛋白質において,hIL-10がhIL-10としての機能を有するというときは,hIL-10が,通常の野生型のhIL-10の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでhIL-10-SA融合蛋白質におけるhIL-10の比活性は,当該融合蛋白質の単位質量当たりのhIL-10の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhIL-10に相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "hIL-10-SA fusion protein" or "hIL-10-SA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hIL-10, and which has the function of hIL-10. When hIL-10 in the hIL-10-SA fusion protein is said to have the function of hIL-10, it means that hIL-10 retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hIL-10 is taken as 100%. Here, the specific activity of hIL-10 in the hIL-10-SA fusion protein is calculated by multiplying the physiological activity of hIL-10 per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hIL-10).
 本発明の一実施形態において,「hIL-10-HSA融合蛋白質」又は「hIL-10-HSA」の語は,hIL-10のアミノ酸配列のC末端に,直接又はリンカーを介して,HSAのアミノ酸配列のN末端が結合されている,アミノ酸配列を有する融合蛋白質であって,hIL-10としての機能を有するものを示す。ここでhIL-10-HSA融合蛋白質がhIL-10としての機能を有するというときは,上記のhIL-10-SAにおける定義を適用することができる。また,これら融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, the term "hIL-10-HSA fusion protein" or "hIL-10-HSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hIL-10, and which has the function of hIL-10. When it is said that the hIL-10-HSA fusion protein has the function of hIL-10, the definition of hIL-10-SA above can be applied. In addition, in these fusion proteins, it is preferable that the SA has the function of SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but this is not limited to this.
 本発明の一実施形態において好ましいhIL-10-HSA融合蛋白質は,例えば配列番号52で示されるアミノ酸配列を有するものである。配列番号52で示されるhIL-10-HSA融合蛋白質は,野生型hIL-10のC末端に直接野生型HSAが結合したものである。配列番号52で示されるhIL-10-HSA融合蛋白質は,例えば,配列番号53で示される塩基配列を有する遺伝子にコードされる。また,配列番号52で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hIL-10としての機能を有するものである限りhIL-10-HSA融合蛋白質に含まれる。hIL-10-HSA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred hIL-10-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 52. The hIL-10-HSA fusion protein shown in SEQ ID NO: 52 is a fusion protein in which wild-type HSA is directly bound to the C-terminus of wild-type hIL-10. The hIL-10-HSA fusion protein shown in SEQ ID NO: 52 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 53. In addition, the hIL-10-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 52 are replaced with other amino acid residues, deleted, or mutated by addition, as long as it has the function of hIL-10. The hIL-10-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 配列番号52で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号52で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:52 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:52.
 野生型hIL-10と野生型HSAとの融合蛋白質であるhIL-10-HSA融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhIL-10部分には変異を加えないことも,HSA部分には変異を加えずにhIL-10部分にのみ変異を加えることも,また,HSA部分とhIL-10部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hIL-10部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hIL-10に変異が加えられたhIL-10のアミノ酸配列となる。HSA部分とhIL-10部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hIL-10部分のアミノ酸配列は上述した野生型hIL-10に変異が加えられたhIL-10のアミノ酸配列となる。配列番号52で示されるアミノ酸配列を有するhIL-10-HSA融合蛋白質についても同様である。野生型hIL-10とヒト以外の動物種の野生型SA(hIL-10-SA)との融合蛋白質についても同様のことがいえる。 When mutations are introduced into the hIL-10-HSA fusion protein, which is a fusion protein of wild-type hIL-10 and wild-type HSA, mutations can be introduced only in the HSA portion and not in the hIL-10 portion, mutations can be introduced only in the hIL-10 portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hIL-10 portion. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are introduced only in the hIL-10 portion, the amino acid sequence of the portion is the amino acid sequence of hIL-10 obtained by adding a mutation to the wild-type hIL-10 described above. When mutations are introduced in both the HSA portion and the hIL-10 portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hIL-10 portion is the amino acid sequence of hIL-10 obtained by adding a mutation to the wild-type hIL-10 described above. The same is true for the hIL-10-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 52. The same is true for the fusion protein of wild-type hIL-10 and wild-type SA (hIL-10-SA) of an animal species other than human.
 配列番号52で示されるアミノ酸配列を有するhIL-10-HSA融合蛋白質に変異を加える場合について,以下例示する。配列番号52で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号52で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。hIL-10-HSA融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hIL-10部分にのみ加えても,又は両方の部分に加えてもよい。 The following is an example of a case where a mutation is made to a hIL-10-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:52. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:52 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:52 or to the N-terminus or C-terminus of the amino acid sequence. The hIL-10-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hIL-10 portion, or both portions.
 配列番号52で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号52で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:52 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:52.
 hIL-10-HSA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもhIL-10-HSA融合蛋白質である。また,hIL-10-HSA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもhIL-10-HSA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもhIL-10-HSA融合蛋白質である。また,hIL-10-HSA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもhIL-10-HSA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhIL-10との融合蛋白質(SA-hIL-10)についても同様のことがいえる。  hIL-10-HSA fusion proteins in which the amino acids constituting the fusion protein are modified with sugar chains are also hIL-10-HSA fusion proteins. In addition, hIL-10-HSA fusion proteins in which the amino acids constituting the fusion protein are modified with phosphate are also hIL-10-HSA fusion proteins. In addition, hIL-10-HSA fusion proteins modified with anything other than sugar chains and phosphate are also hIL-10-HSA fusion proteins. In addition, hIL-10-HSA fusion proteins in which the side chains of the amino acids constituting the fusion protein are changed by a substitution reaction or the like are also hIL-10-HSA fusion proteins. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of SA and hIL-10 of animal species other than humans (SA-hIL-10).
 つまり,糖鎖により修飾されたhIL-10-HSA融合蛋白質は,元のアミノ酸配列を有するhIL-10-HSA融合蛋白質に含まれるものとする。また,リン酸により修飾されたhIL-10-HSA融合蛋白質は,元のアミノ酸配列を有するhIL-10-HSA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するhIL-10-HSA融合蛋白質に含まれるものとする。また,hIL-10-HSA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するhIL-10-HSA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhIL-10との融合蛋白質(SA-hIL-10)についても同様のことがいえる。 In other words, hIL-10-HSA fusion proteins modified with sugar chains are included in the hIL-10-HSA fusion proteins having the original amino acid sequence. Also, hIL-10-HSA fusion proteins modified with phosphate are included in the hIL-10-HSA fusion proteins having the original amino acid sequence. Also, those modified with something other than sugar chains and phosphate are also included in the hIL-10-HSA fusion proteins having the original amino acid sequence. Also, those in which the side chains of the amino acids constituting the hIL-10-HSA fusion protein have been changed by a substitution reaction or the like are also included in the hIL-10-HSA fusion proteins having the original amino acid sequence. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hIL-10 of animal species other than humans (SA-hIL-10).
 また,hIL-10-HSA融合蛋白質であって,これを構成するhIL-10が,hIL-10の前駆体であるものもhIL-10-HSA融合蛋白質である。ヒト以外の動物種のSAとhIL-10との融合蛋白質(SA-hIL-10)についても同様のことがいえる。ヒト以外の動物種のSAとhIL-10との融合蛋白質(SA-hIL-10),及びヒト以外の動物種のSAとヒト以外の動物種のIL-10との融合蛋白質,例えばMSA-マウスIL-10との融合蛋白質についても同様のことがいえる。  Also, a hIL-10-HSA fusion protein in which the constituent hIL-10 is a precursor of hIL-10 is also a hIL-10-HSA fusion protein. The same can be said for a fusion protein of SA and hIL-10 of a non-human animal species (SA-hIL-10). The same can be said for a fusion protein of SA and hIL-10 of a non-human animal species (SA-hIL-10), and a fusion protein of SA and IL-10 of a non-human animal species, such as a fusion protein of MSA-mouse IL-10.
 本発明の一実施形態において,「HSAとヒトサイトカインとの融合蛋白質」,「ヒトサイトカインとHSAとの融合蛋白質」,又は「ヒト血清アルブミンとヒトサイトカインとの融合蛋白質」,というときは,上記の「HSA-ヒトサイトカイン融合蛋白質」及び「ヒトサイトカインHSA融合蛋白質」のいずれをも含むものとする。また,「HSAとhIL-10との融合蛋白質」,「hIL-10とHSAとの融合蛋白質」,又は「ヒト血清アルブミンとヒトサイトカイン10との融合蛋白質」,というときは,上記の「HSA-hIL-10融合蛋白質」及び「hIL-10-HSA融合蛋白質」のいずれをも含むものとする。 In one embodiment of the present invention, the terms "HSA and human cytokine fusion protein", "human cytokine and HSA fusion protein", or "human serum albumin and human cytokine fusion protein" include both the above-mentioned "HSA-human cytokine fusion protein" and "human cytokine HSA fusion protein". In addition, the terms "HSA and hIL-10 fusion protein", "hIL-10 and HSA fusion protein", or "human serum albumin and human cytokine 10 fusion protein" include both the above-mentioned "HSA-hIL-10 fusion protein" and "hIL-10-HSA fusion protein".
 以下,HSAとhIL-10との融合蛋白質について製造法等につき詳述するが,これらの記載事項は,サイトカインがIL-10以外のものである場合,SAがヒト以外の動物種である場合,サイトカインがヒト以外の動物種である場合にも適用され得る。例えば,ヒト以外の動物種のSAとhIL-10との融合蛋白質,ヒト以外の動物種のSAとヒト以外の動物種のIL-10との融合蛋白質についても適用される。  The manufacturing method and other details of the fusion protein of HSA and hIL-10 are described below, but these descriptions can also be applied when the cytokine is other than IL-10, when the SA is from an animal species other than human, or when the cytokine is from an animal species other than human. For example, they also apply to a fusion protein of SA of an animal species other than human and hIL-10, and a fusion protein of SA of an animal species other than human and IL-10 of an animal species other than human.
 本発明の一実施形態の融合蛋白質において,SAとサイトカインとは,直接又はリンカーを介して結合される。ここで,「リンカー」というときは,SAのアミノ酸配列とサイトカインのアミノ酸配列のいずれにも属さない部分のことをいう。つまり,リンカーはSAとサイトカインとの間に介在するペプチド鎖のことである。リンカーは,種々の機能を有する。その機能には,SAとサイトカインとの間にあってSAとサイトカインとを結合する機能の他,SA及びサイトカインの,融合蛋白質分子内での距離を離すことにより相互の干渉を低減させる機能,SAとサイトカインとの間にあってSAとサイトカインとを連結するヒンジとなって,融合蛋白質の立体構造に柔軟性を与える機能等が含まれる。融合蛋白質の分子内において,リンカーは,これら機能の少なくとも一つを発揮する。このことは,例えばSAがHSAであっても,また例えばサイトカインがヒトサイトカイン,例えばhIL-10であっても,同様である。 In one embodiment of the fusion protein, the SA and the cytokine are linked directly or via a linker. Here, the term "linker" refers to a portion that does not belong to either the amino acid sequence of the SA or the amino acid sequence of the cytokine. In other words, the linker is a peptide chain that is interposed between the SA and the cytokine. The linker has various functions. These functions include a function between the SA and the cytokine to link the SA and the cytokine, a function to reduce mutual interference between the SA and the cytokine by increasing the distance between them within the fusion protein molecule, and a function between the SA and the cytokine to act as a hinge that connects the SA and the cytokine and gives flexibility to the three-dimensional structure of the fusion protein. Within the fusion protein molecule, the linker exerts at least one of these functions. This is the same, for example, whether the SA is HSA or whether the cytokine is a human cytokine, such as hIL-10.
 SAとサイトカインとの融合蛋白質において,ペプチドリンカーのアミノ酸配列は,融合蛋白質分子の中にあって,リンカーとしての機能が発揮されるものである限り特に限定はない。また,ペプチドリンカーの長さにも,融合蛋白質分子の中にあって,リンカーとしての機能が発揮されるものである限り特に限定はない。ペプチドリンカーは一個又は複数個のアミノ酸から構成されるものである。ペプチドリンカーが複数個のアミノ酸から構成される場合,そのアミノ酸の個数は,好ましくは2~50個であり,より好ましくは5~30個であり,更に好ましくは10~25個である。ペプチドリンカーの好適な例として,Gly-Ser,Gly-Gly-Ser,又は配列番号9~11で示されるアミノ酸配列(これらをあわせて基本配列という)からなるもの,及びこれらを含むものが挙げられる。例えば,ペプチドリンカーは,基本配列が2~10回反復したアミノ酸配列を含むものであり,基本配列が2~6回反復したアミノ酸配列を含むものであり,基本配列が3~5回反復したアミノ酸配列を含むものである。これらのアミノ酸配列中の1個又は複数個のアミノ酸が,欠失,他のアミノ酸へ置換,付加等されたものであってもよい。アミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1又は2個である。アミノ酸を他のアミノ酸へ置換させる場合,置換させるアミノ酸の個数は,好ましくは1又は2個である。アミノ酸を付加する場合,付加されるアミノ酸の個数は,好ましくは1又は2個である。これらアミノ酸の欠失,置換,及び付加を組み合わせて,所望のリンカー部のアミノ酸配列とすることもできる。ペプチドリンカーは一つのアミノ酸からなるものであってもよく,リンカーを構成するアミノ酸は,例えばグリシン,セリンである。 In the fusion protein of SA and a cytokine, the amino acid sequence of the peptide linker is not particularly limited as long as it functions as a linker in the fusion protein molecule. The length of the peptide linker is also not particularly limited as long as it functions as a linker in the fusion protein molecule. The peptide linker is composed of one or more amino acids. When the peptide linker is composed of multiple amino acids, the number of amino acids is preferably 2 to 50, more preferably 5 to 30, and even more preferably 10 to 25. Suitable examples of peptide linkers include those consisting of Gly-Ser, Gly-Gly-Ser, or amino acid sequences shown in SEQ ID NOs: 9 to 11 (collectively referred to as basic sequences), and those containing these. For example, the peptide linker is one that contains an amino acid sequence in which the basic sequence is repeated 2 to 10 times, one that contains an amino acid sequence in which the basic sequence is repeated 2 to 6 times, or one that contains an amino acid sequence in which the basic sequence is repeated 3 to 5 times. One or more amino acids in these amino acid sequences may be deleted, replaced with other amino acids, added, etc. When deleting an amino acid, the number of amino acids to be deleted is preferably 1 or 2. When replacing an amino acid with another amino acid, the number of amino acids to be replaced is preferably 1 or 2. When adding an amino acid, the number of amino acids to be added is preferably 1 or 2. The amino acid sequence of the desired linker portion can be obtained by combining these deletions, substitutions, and additions of amino acids. The peptide linker may be composed of one amino acid, and the amino acid that constitutes the linker is, for example, glycine or serine.
 HSAとhIL-10との融合蛋白質は,HSAをコードする遺伝子の下流又は上流に,インフレームでhIL-10をコードする遺伝子を結合させたDNA断片を組み込んだ発現ベクターを作製し,この発現ベクターを導入して形質転換させた宿主細胞を培養することにより,組換え蛋白質として作製することができる。このようにして組換え蛋白質として作製される融合蛋白質は,一本鎖ポリペプチドからなる。 The fusion protein of HSA and hIL-10 can be produced as a recombinant protein by creating an expression vector incorporating a DNA fragment in which a gene encoding hIL-10 is linked in-frame to the downstream or upstream of a gene encoding HSA, and then culturing a host cell transformed with this expression vector. The fusion protein produced as a recombinant protein in this way consists of a single polypeptide chain.
 組換え融合蛋白質として融合蛋白質を作製する場合,HSAをコードする遺伝子の下流にインフレームでhIL-10をコードする遺伝子を結合させることにより,HSAのアミノ酸配列のC末端にhIL-10のアミノ酸配列を有するHSA-hIL-10融合蛋白質を得ることができる。逆に,HSAをコードする遺伝子の上流にインフレームでhIL-10をコードする遺伝子を結合させることにより,HSAのアミノ酸配列のN末端にhIL-10のアミノ酸配列を有するhIL-10-HSA融合蛋白質が得られる。いずれの場合においても,組換え融合蛋白質として作製される融合蛋白質は,一本鎖ポリペプチドである。 When producing a fusion protein as a recombinant fusion protein, a gene encoding hIL-10 can be linked in-frame downstream of a gene encoding HSA to obtain an HSA-hIL-10 fusion protein having the amino acid sequence of hIL-10 at the C-terminus of the amino acid sequence of HSA. Conversely, a gene encoding hIL-10 can be linked in-frame upstream of a gene encoding HSA to obtain an hIL-10-HSA fusion protein having the amino acid sequence of hIL-10 at the N-terminus of the amino acid sequence of HSA. In either case, the fusion protein produced as a recombinant fusion protein is a single-chain polypeptide.
 融合蛋白質の一本鎖ポリペプチド内で,HSAのアミノ酸配列がhIL-10のアミノ酸配列のN末端側に位置する場合,HSAのC末端とhIL-10のN末端が,ペプチド結合により直接,又はリンカーを介して結合される。図5にN末端側からHSA,リンカー及びhIL-10を順に有する一本鎖ポリペプチドのHSA-hIL-10融合蛋白質を模式的に示す。該HSA-hIL-10融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とhIL-10のN末端がペプチド結合により結合したものである。 When the amino acid sequence of HSA is located on the N-terminal side of the amino acid sequence of hIL-10 within the single-chain polypeptide of the fusion protein, the C-terminus of HSA and the N-terminus of hIL-10 are bound directly by a peptide bond or via a linker. Figure 5 shows a schematic diagram of an HSA-hIL-10 fusion protein, a single-chain polypeptide having HSA, a linker, and hIL-10 in that order from the N-terminus. In the HSA-hIL-10 fusion protein, the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hIL-10 by a peptide bond.
 また,融合蛋白質の一本鎖ポリペプチド内で,hIL-10のアミノ酸配列がHSAのアミノ酸配列のN末端に位置する場合,hIL-10のC末端とHSAのN末端が,ペプチド結合により直接,又はリンカーを介して結合される。図6にN末端側からhIL-10,リンカー及びHSAを順に有する一本鎖ポリペプチドのhIL-10-HSA融合蛋白質を模式的に示す。該hIL-10-HSA融合蛋白質はhIL-10のC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。 In addition, when the amino acid sequence of hIL-10 is located at the N-terminus of the amino acid sequence of HSA within the single-chain polypeptide of the fusion protein, the C-terminus of hIL-10 and the N-terminus of HSA are bound directly by a peptide bond or via a linker. Figure 6 shows a schematic diagram of a hIL-10-HSA fusion protein, which is a single-chain polypeptide having hIL-10, a linker, and HSA in that order from the N-terminus. In the hIL-10-HSA fusion protein, the C-terminus of hIL-10 is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of HSA by a peptide bond.
 本発明の一実施形態において,HSAとhIL-10との融合蛋白質というときは,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhIL-10を組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhIL-10としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,3~4倍等となることを特徴とする融合蛋白質である。ここで同一条件下とは,発現ベクター,宿主細胞,培養条件等が同一であることをいう。このとき用いられる好ましい宿主細胞は,CHO細胞,NS/0細胞等の哺乳動物細胞であるが,特にCHO細胞である。 In one embodiment of the present invention, the fusion protein of HSA and hIL-10 refers to a fusion protein characterized in that when expressed in a host cell as a recombinant protein, particularly when expressed so that the recombinant protein is secreted from the cells and accumulated in the culture medium, the amount of expression of hIL-10 in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hIL-10 is expressed in a host cell as a recombinant protein under the same conditions. Here, "under the same conditions" means that the expression vector, host cell, culture conditions, etc. are the same. The preferred host cells used in this case are mammalian cells such as CHO cells and NS/0 cells, but particularly CHO cells.
 かかるHSAとhIL-10との融合蛋白質の好ましい実施形態として,以下の(1)~(4)が挙げられる:
(1)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号24で示される野生型のhIL-10のアミノ酸配列のN末端が直接結合したものである,配列番号50で示されるアミノ酸配列を有するもの,
(2)配列番号24で示される野生型のhIL-10のアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端が直接結合したものである,配列番号52で示されるアミノ酸配列を有するもの。
(3)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号24で示される野生型のhIL-10のアミノ酸配列のN末端が配列番号9で示されるリンカーを介して結合したものである,配列番号54で示されるアミノ酸配列を有するもの,
(4)配列番号24で示される野生型のhIL-10のアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端が配列番号9で示されるリンカーを介して結合したものである,配列番号55で示されるアミノ酸配列を有するもの。
Preferred embodiments of the fusion protein of HSA and hIL-10 include the following (1) to (4):
(1) A substance having an amino acid sequence shown in SEQ ID NO:50, in which the N-terminus of the amino acid sequence of wild-type hIL-10 shown in SEQ ID NO:24 is directly linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO:3;
(2) Having the amino acid sequence shown in SEQ ID NO:52, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO:3 is directly linked to the C-terminus of the amino acid sequence of wild-type hIL-10 shown in SEQ ID NO:24.
(3) A polypeptide having an amino acid sequence represented by SEQ ID NO:54, in which the N-terminus of the amino acid sequence of wild-type hIL-10 represented by SEQ ID NO:24 is linked to the C-terminus of the amino acid sequence of wild-type HSA represented by SEQ ID NO:3 via a linker represented by SEQ ID NO:9;
(4) Having the amino acid sequence shown in SEQ ID NO:55, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO:3 is linked to the C-terminus of the amino acid sequence of wild-type hIL-10 shown in SEQ ID NO:24 via a linker shown in SEQ ID NO:9.
 また,かかるHSAとhIL-10との融合蛋白質の好ましい実施形態として,配列番号3で示される野生型のHSAのアミノ酸配列のN末端から320番目のアミノ酸残基であるアラニンをトレオニンで置換させた,配列番号13で示される585個のアミノ酸からなるヒト血清アルブミン(HSA-A320T)を用いたものである,以下の(5)~(8)がさらに挙げられる:
(5)配列番号13で示されるヒト血清アルブミン(HSA-A320T)のアミノ酸配列のC末端に,配列番号24で示される野生型のhIL-10のアミノ酸配列のN末端が直接結合したものである,配列番号56で示されるアミノ酸配列を有するもの,
(6)配列番号24で示される野生型のhIL-10のアミノ酸配列のC末端に,配列番号13で示されるヒト血清アルブミン(HSA-A320T)のアミノ酸配列のN末端が直接結合したものである,全体として配列番号57で示されるアミノ酸配列を有するもの。
(7)配列番号13で示されるヒト血清アルブミン(HSA-A320T)のアミノ酸配列のC末端に,配列番号24で示される野生型のhIL-10のアミノ酸配列のN末端が配列番号9で示されるリンカーを介して結合したものである,配列番号58で示されるアミノ酸配列を有するもの,
(8)配列番号24で示される野生型のhIL-10のアミノ酸配列のC末端に,配列番号13で示されるヒト血清アルブミン(HSA-A320T)のアミノ酸配列のN末端が配列番号9で示されるリンカーを介して結合したものである,配列番号59で示されるアミノ酸配列を有するもの。
Furthermore, preferred embodiments of the fusion protein of HSA and hIL-10 include human serum albumin (HSA-A320T) consisting of 585 amino acids as shown in SEQ ID NO:13, in which the 320th amino acid residue from the N-terminus of the amino acid sequence of wild-type HSA as shown in SEQ ID NO:3 is replaced with threonine, and further include the following (5) to (8):
(5) A peptide having an amino acid sequence shown in SEQ ID NO:56, in which the N-terminus of the amino acid sequence of wild-type hIL-10 shown in SEQ ID NO:24 is directly linked to the C-terminus of the amino acid sequence of human serum albumin (HSA-A320T) shown in SEQ ID NO:13;
(6) The C-terminus of the amino acid sequence of wild-type hIL-10 shown in SEQ ID NO:24 is directly linked to the N-terminus of the amino acid sequence of human serum albumin (HSA-A320T) shown in SEQ ID NO:13, and the overall amino acid sequence is shown in SEQ ID NO:57.
(7) A peptide having an amino acid sequence represented by SEQ ID NO:58, in which the N-terminus of the amino acid sequence of wild-type hIL-10 represented by SEQ ID NO:24 is linked to the C-terminus of the amino acid sequence of human serum albumin (HSA-A320T) represented by SEQ ID NO:13 via a linker represented by SEQ ID NO:9;
(8) Having the amino acid sequence shown in SEQ ID NO:59, in which the N-terminus of the amino acid sequence of human serum albumin (HSA-A320T) shown in SEQ ID NO:13 is linked to the C-terminus of the amino acid sequence of wild-type hIL-10 shown in SEQ ID NO:24 via a linker shown in SEQ ID NO:9.
 本発明の一実施形態におけるHSAとhIL-10との融合蛋白質は,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件下で野生型のhIL-10を組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhIL-10としての発現量が,濃度あるいは生理活性換算で増加することを特徴とするものである。従って,HSAとhIL-10との融合蛋白質は,組換え蛋白質として製造したときに,野生型のhIL-10と比較して生産効率を上昇させることができるので,生産コストを低減することができる。なお,組換え蛋白質を有効成分として含有する医薬品は,非常に高価であることが知られている。従って,同一条件で製造したときに得られる組換え蛋白質の量を数パーセント,例えば3~9%上昇させることにも,多大な経済的効果がある。同様のことは,サイトカイン,例えばhIL-10についてもいえる。 In one embodiment of the present invention, the fusion protein of HSA and hIL-10 is characterized in that when expressed in a host cell as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hIL-10 expressed in the culture supernatant is increased in terms of concentration or physiological activity, compared to when wild-type hIL-10 is expressed in a host cell as a recombinant protein under the same conditions. Therefore, when produced as a recombinant protein, the fusion protein of HSA and hIL-10 can increase the production efficiency compared to wild-type hIL-10, and therefore can reduce production costs. It is known that pharmaceuticals containing recombinant proteins as active ingredients are very expensive. Therefore, even increasing the amount of recombinant protein obtained when produced under the same conditions by a few percent, for example 3 to 9%, has a significant economic effect. The same can be said for cytokines, such as hIL-10.
 なお,本明細書において,組換え蛋白質として宿主細胞で発現させたHSAとhIL-10との融合蛋白質と,同一条件下で組換え蛋白質として宿主細胞で発現させた野生型のhIL-10の,発現量を比較するときは,発現した蛋白質の質量で比較するのではなく,発現した蛋白質の分子数またはhIL-10生理活性で比較するものとする。ヒト以外の動物種のSAとhIL-10との融合蛋白質,SAとその他のサイトカインとの融合蛋白質についても同様とする。 In this specification, when comparing the expression levels of a fusion protein of HSA and hIL-10 expressed in a host cell as a recombinant protein with wild-type hIL-10 expressed in a host cell under the same conditions as a recombinant protein, the comparison is not based on the mass of the expressed protein, but on the number of molecules of the expressed protein or the hIL-10 physiological activity. The same applies to fusion proteins of SA and hIL-10 from animal species other than human, and fusion proteins of SA and other cytokines.
 HSAとhIL-10との融合蛋白質は,上述したHSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質を製造するために用いることのできる,発現ベクター,宿主細胞,培地等を用いて製造することができる。なお,これに限らず,SAとサイトカインとの融合蛋白質全般についても同様のことがいえる。
が好まし
The fusion protein of HSA and hIL-10 can be produced using an expression vector, host cell, medium, etc. that can be used to produce the above-mentioned fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA. The same can be said for fusion proteins of SA and cytokines in general.
is preferred
 HSAとhIL-10との融合蛋白質は,これをコードする遺伝子を組み込んだ発現ベクターを導入した宿主細胞を上記の無血清培地で培養して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhIL-10を組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhIL-10としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,3~4倍等となることを特徴とする。ここで同一条件下とは,発現ベクター,宿主細胞,培養条件等が同一であることをいう。このとき用いられる宿主細胞は,好ましくは哺乳動物細胞,特に,CHO細胞,NS/0細胞等の組換え蛋白質の製造に用いられる通常の細胞である。 The fusion protein of HSA and hIL-10 is characterized in that when host cells into which an expression vector incorporating a gene encoding this protein has been introduced are cultured in the above-mentioned serum-free medium to be expressed as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of expression of hIL-10 in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hIL-10 is expressed as a recombinant protein in host cells under the same conditions. Here, "under the same conditions" means that the expression vector, host cells, culture conditions, etc. are the same. The host cells used in this case are preferably mammalian cells, particularly ordinary cells used for producing recombinant proteins such as CHO cells and NS/0 cells.
 野生型のhIL-10は,これをコードする遺伝子を組込んだ発現ベクターで形質転換させた宿主細胞を用いて組換え体として発現させたときの発現量が低い傾向にあり,効率よく組換え体として大量に製造することが困難である。しかし,hIL-10は,hIL-10をHSAとの融合蛋白質として発現させることにより,組換え体として比較的発現量を増加させることができる。つまり,HSAとhIL-10との融合蛋白質は,これをコードする遺伝子を組み込んだ発現ベクターを導入した宿主を無血清培地で培養して組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型hIL-10を同一条件で組換え蛋白質として発現させたときと比較して,効率よく発現させることができるので,大量生産に適している。ヒト以外の動物種のSAとhIL-10の融合蛋白質についても同様である。なお,当該融合蛋白質の発現に用いられる宿主細胞は,好ましくは哺乳動物細胞,特に,CHO細胞,NS/0細胞等の組換え蛋白質の製造に用いられる通常の細胞である。つまり,本発明の一実施形態は,SAとIL-10との組換え融合蛋白質,特にHSAとhIL-10との組換え融合蛋白質である。  Wild-type hIL-10 tends to have a low expression level when expressed as a recombinant using host cells transformed with an expression vector incorporating a gene encoding it, making it difficult to efficiently mass-produce it as a recombinant. However, by expressing hIL-10 as a fusion protein with HSA, the expression level of the recombinant can be increased. In other words, when a host into which an expression vector incorporating a gene encoding it has been introduced is cultured in a serum-free medium and expressed as a recombinant protein, especially when the recombinant protein is expressed so that it is secreted from the cells and accumulated in the culture medium, the fusion protein of HSA and hIL-10 can be expressed more efficiently than when wild-type hIL-10 is expressed as a recombinant protein under the same conditions, making it suitable for mass production. The same applies to fusion proteins of SA and hIL-10 of animal species other than humans. The host cells used to express the fusion protein are preferably mammalian cells, especially ordinary cells used for producing recombinant proteins such as CHO cells and NS/0 cells. That is, one embodiment of the present invention is a recombinant fusion protein of SA and IL-10, particularly a recombinant fusion protein of HSA and hIL-10.
 HSAとhIL-10との融合蛋白質をコードする宿主細胞を培養することにより,細胞内又は培地中に発現させることができる。HSAとhIL-10との融合蛋白質は,カラムクロマトグラフィー等の方法により不純物から分離し,精製することができる。精製されたHSAとhIL-10との融合蛋白質は,医薬組成物として使用することができる。特に,HSAとhIL-10との融合蛋白質は,炎症性疾患又は癌を対象疾患とする医薬組成物として使用することができる。 By culturing host cells encoding the fusion protein of HSA and hIL-10, it can be expressed in cells or in the medium. The fusion protein of HSA and hIL-10 can be purified by separating it from impurities using methods such as column chromatography. The purified fusion protein of HSA and hIL-10 can be used as a pharmaceutical composition. In particular, the fusion protein of HSA and hIL-10 can be used as a pharmaceutical composition targeting inflammatory diseases or cancer.
 HSAとhIL-10との融合蛋白質を有効成分として含有してなる医薬組成物は,注射剤として静脈内,筋肉内,腹腔内,皮下又は脳室内に投与することができる。それらの注射剤は,凍結乾燥製剤又は水性液剤として供給することができる。水性液剤とする場合,バイアルに充填した形態としてもよく,注射器に予め充填したものであるプレフィルド型の製剤として供給することもできる。凍結乾燥製剤の場合,使用前に水性媒質に溶解し復元して使用する。  A pharmaceutical composition containing a fusion protein of HSA and hIL-10 as an active ingredient can be administered intravenously, intramuscularly, intraperitoneally, subcutaneously, or intracerebroventricularly as an injection. These injections can be supplied as lyophilized preparations or aqueous liquid preparations. In the case of an aqueous liquid preparation, it may be in the form of a vial filled with the drug, or it can be supplied as a prefilled preparation in a syringe. In the case of a lyophilized preparation, it is dissolved in an aqueous medium and reconstituted before use.
 HSAとhIL-10との融合蛋白質は,更に,抗体又はリガンドとの結合体とすることができる。例えば,本発明の一実施形態におけるHSAとhIL-10との融合蛋白質は,脳血管内皮細胞上の受容体と特異的に結合することのできる抗体又はリガンドとの結合体とすることができる。HSAとhIL-10との融合蛋白質又はHSAとhGBAとの融合蛋白質は,脳血管内皮細胞上の受容体と特異的に結合することのできる抗体又はリガンドとの結合体とすることにより,脳血管内皮細胞上の受容体に結合できるようになる。脳血管内皮細胞上の受容体に結合した融合蛋白質は,血液脳関門(BBB)を通過して,中枢神経系(CNS)の組織に到達できる。従って,HSAとhIL-10との融合蛋白質は,かかる抗体又はリガンドとの結合体とすることで,血液脳関門(BBB)を通過させて,中枢神経系(CNS)において機能を発揮させるようにすることができる。 The fusion protein of HSA and hIL-10 can further be conjugated with an antibody or a ligand. For example, in one embodiment of the present invention, the fusion protein of HSA and hIL-10 can be conjugated with an antibody or a ligand capable of specifically binding to a receptor on cerebrovascular endothelial cells. The fusion protein of HSA and hIL-10 or the fusion protein of HSA and hGBA can bind to a receptor on cerebrovascular endothelial cells by being conjugated with an antibody or a ligand capable of specifically binding to a receptor on cerebrovascular endothelial cells. The fusion protein bound to the receptor on cerebrovascular endothelial cells can pass through the blood-brain barrier (BBB) and reach the tissues of the central nervous system (CNS). Therefore, the fusion protein of HSA and hIL-10 can be conjugated with such an antibody or ligand to pass through the blood-brain barrier (BBB) and exert its function in the central nervous system (CNS).
 HSAとhIL-10との融合蛋白質と抗体との結合体において,hIL-10がhIL-10としての機能を有するというときは,hIL-10が,通常の野生型のhIL-10の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性,例えば90%以上,95%以上の比活性を有することをいう。なお,ここで当該結合体におけるhIL-10の比活性は,当該結合体の単位質量当たりのhIL-10の生理活性に(当該結合体の分子量/当該結合体中のhIL-10に相当する部分の分子量)を乗じて算出される。 When hIL-10 in a conjugate of an antibody and a fusion protein of HSA and hIL-10 is said to have the function of hIL-10, it means that, when the specific activity of normal wild-type hIL-10 is taken as 100%, hIL-10 has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more. Here, the specific activity of hIL-10 in the conjugate is calculated by multiplying the physiological activity of hIL-10 per unit mass of the conjugate by (the molecular weight of the conjugate/the molecular weight of the portion of the conjugate corresponding to hIL-10).
 本発明の一実施形態において,HSAとhIL-10との融合蛋白質と抗体との結合体というときは,当該結合体を組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhIL-10を組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhIL-10としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,3~4倍等となることを特徴とするものである。 In one embodiment of the present invention, a conjugate of an antibody and a fusion protein of HSA and hIL-10 refers to a conjugate characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of expression of hIL-10 in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hIL-10 is expressed as a recombinant protein in a host cell under the same conditions.
  神経栄養因子の中には,野生型のものをコードする遺伝子を組込んだ発現ベクターを導入した宿主細胞を用いて組換え蛋白質として発現させた場合に,発現量が限定的であるため大量に製造することが比較的困難なものがある。そのような神経栄養因子には,例えば,BDNF(脳由来神経栄養因子),NGF(神経成長因子),NT-3(ニューロトロフィン-3),NT-4(ニューロトロフィン-4),及びNT-5(ニューロトロフィン-5)がある。NT-4とNT-5の構造の違いは種間変異であり,これらは同一の因子であると考えられているため,NT-4/5等と表記される場合があるが,本明細書においてはNT-4と表記するものとする本発明の一実施形態は,このような組換え蛋白質として製造が比較的困難な神経栄養因子を,SAとの融合蛋白質とした組換え蛋白質である。かかる組換え蛋白質は,これをコードする遺伝子を組込んだ発現ベクターを導入した宿主細胞を用いて,大量に製造することが,対応する神経栄養因子と比較して,容易になる。ここで,SAと結合させる神経栄養因子の動物種に特に制限はないが,好ましくはヒト神経栄養因子である。また,神経栄養因子と結合させるSAの動物種に特に制限はないが,好ましくはHSAである。 Some neurotrophic factors are relatively difficult to produce in large quantities when expressed as recombinant proteins using host cells introduced with an expression vector incorporating a gene encoding the wild-type neurotrophic factor, due to limited expression levels. Examples of such neurotrophic factors include BDNF (brain-derived neurotrophic factor), NGF (nerve growth factor), NT-3 (neurotrophin-3), NT-4 (neurotrophin-4), and NT-5 (neurotrophin-5). The difference in structure between NT-4 and NT-5 is an interspecies variation, and since these are considered to be the same factor, they may be referred to as NT-4/5, etc., but in this specification, they will be referred to as NT-4. One embodiment of the present invention is a recombinant protein in which such a neurotrophic factor, which is relatively difficult to produce as a recombinant protein, is fused with SA. Such a recombinant protein can be produced in large quantities more easily using host cells introduced with an expression vector incorporating a gene encoding the recombinant protein, compared to the corresponding neurotrophic factor. Here, there is no particular restriction on the animal species of the neurotrophic factor to be bound to SA, but human neurotrophic factor is preferable. In addition, there are no particular limitations on the animal species of SA to be bound to the neurotrophic factor, but HSA is preferred.
 本明細書において,単に「ヒト神経栄養因子」というときは,通常の野生型のヒト神経栄養因子に加え,当該ヒト神経栄養因子の種類に対応する生理活性を有する等のヒト神経栄養因子としての機能を有するものである限り,野生型のヒト神経栄養因子のアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するヒト神経栄養因子の変異体も特に区別することなく包含する。ヒト以外の動物種の神経栄養因子についても同様である。 In this specification, when the term "human neurotrophic factor" is used, it includes not only normal wild-type human neurotrophic factor, but also mutant human neurotrophic factors in which one or more amino acid residues have been substituted, deleted, and/or added (in this specification, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence of the wild-type human neurotrophic factor, as long as the mutant has the function of a human neurotrophic factor, such as having physiological activity corresponding to the type of human neurotrophic factor. The same applies to neurotrophic factors of animal species other than humans.
 なお,ここで,ヒト神経栄養因子がヒト神経栄養因子としての機能を有するというときは,ヒト神経栄養因子が,通常の野生型のヒト神経栄養因子の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。ここで比活性とは蛋白質の質量当たりの生理活性のことをいう。なお,ヒト神経栄養因子とその他の蛋白質との融合蛋白質の比活性は,融合蛋白質の中でヒト神経栄養因子に相当する部分の質量当たりの生理活性として求められる。なお,ここで融合蛋白質におけるヒト神経栄養因子の比活性は,当該融合蛋白質の単位質量当たりのヒト神経栄養因子の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のヒト神経栄養因子に相当する部分の分子量)を乗じて算出される。ヒト以外の動物種の神経栄養因子についても同様である。 Here, when it is said that a human neurotrophic factor has a function as a human neurotrophic factor, it means that the human neurotrophic factor has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of a normal wild-type human neurotrophic factor is taken as 100%. Here, specific activity refers to the physiological activity per mass of the protein. The specific activity of a fusion protein of a human neurotrophic factor and another protein is calculated as the physiological activity per mass of the portion of the fusion protein that corresponds to the human neurotrophic factor. Here, the specific activity of the human neurotrophic factor in the fusion protein is calculated by multiplying the physiological activity of the human neurotrophic factor per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein that corresponds to the human neurotrophic factor). The same applies to neurotrophic factors of animal species other than humans.
 野生型のヒト神経栄養因子のアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のヒト神経栄養因子のアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のヒト神経栄養因子のN末端又はC末端のアミノ酸残基を1個欠失させたアミノ酸配列からなるヒト神経栄養因子の変異体,野生型のヒト神経栄養因子のN末端又はC末端のアミノ酸残基を2個欠失させたアミノ酸配列からなるヒト神経栄養因子の変異体等も,ヒト神経栄養因子である。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型のヒト神経栄養因子のアミノ酸配列に加えることもできる。ヒト以外の動物種の神経栄養因子についても同様である。 When amino acid residues in the amino acid sequence of wild-type human neurotrophic factor are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of wild-type human neurotrophic factor are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. A human neurotrophic factor mutant consisting of an amino acid sequence in which one amino acid residue is deleted from the N-terminus or C-terminus of wild-type human neurotrophic factor, a human neurotrophic factor mutant consisting of an amino acid sequence in which two amino acid residues are deleted from the N-terminus or C-terminus of wild-type human neurotrophic factor, etc. are also human neurotrophic factors. In addition, a mutation that combines the substitution and deletion of these amino acid residues can be added to the amino acid sequence of wild-type human neurotrophic factor. The same applies to neurotrophic factors of animal species other than humans.
 野生型のヒト神経栄養因子のアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はヒト神経栄養因子のアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型のヒト神経栄養因子のアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型のヒト神経栄養因子のアミノ酸配列に加えることもできる。ヒト以外の動物種の神経栄養因子についても同様である。 When adding amino acid residues to the amino acid sequence of wild-type human neurotrophic factor, one or more amino acid residues are added into the amino acid sequence of human neurotrophic factor or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type human neurotrophic factor, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type human neurotrophic factor. The same applies to neurotrophic factors of animal species other than humans.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型のヒト神経栄養因子のアミノ酸配列に加えることもできる。例えば,野生型のヒト神経栄養因子のアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもヒト神経栄養因子であり,野生型のヒト神経栄養因子のアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもヒト神経栄養因子であり,野生型のヒト神経栄養因子のアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもヒト神経栄養因子であり,野生型のヒト神経栄養因子のアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもヒト神経栄養因子であり,野生型のヒト神経栄養因子のアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもヒト神経栄養因子である。ヒト以外の動物種の神経栄養因子についても同様である。 Furthermore, a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of wild-type human neurotrophic factor. For example, a human neurotrophic factor obtained by deleting 1 to 10 amino acid residues from the amino acid sequence of wild-type human neurotrophic factor, substituting 1 to 10 amino acid residues with other amino acid residues, and adding 1 to 10 amino acid residues is also a human neurotrophic factor, a human neurotrophic factor obtained by deleting 1 to 5 amino acid residues from the amino acid sequence of wild-type human neurotrophic factor, substituting 1 to 5 amino acid residues with other amino acid residues, and adding 1 to 5 amino acid residues is also a human neurotrophic factor, and a human neurotrophic factor obtained by deleting 1 to 3 amino acid residues from the amino acid sequence of wild-type human neurotrophic factor, and adding 1 to 3 amino acid residues is also a human neurotrophic factor. Human neurotrophic factors are those in which an amino acid residue has been replaced with another amino acid residue and one to three amino acid residues have been added. Human neurotrophic factors are also those in which one or two amino acid residues have been deleted from the amino acid sequence of wild-type human neurotrophic factors, one or two amino acid residues have been replaced with another amino acid residue, and one or two amino acid residues have been added. Human neurotrophic factors are also those in which one amino acid residue has been deleted from the amino acid sequence of wild-type human neurotrophic factors, one amino acid residue has been replaced with another amino acid residue, and one amino acid residue has been added. The same applies to neurotrophic factors of animal species other than humans.
 ヒト神経栄養因子変異体における通常の野生型ヒト神経栄養因子と比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両ヒト神経栄養因子のアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種の神経栄養因子についても同様である。 The location and type (deletion, substitution, and addition) of each mutation in a human neurotrophic factor mutant compared to normal wild-type human neurotrophic factor can be easily confirmed by aligning the amino acid sequences of both human neurotrophic factors. The same applies to neurotrophic factors of animal species other than humans.
 ヒト神経栄養因子変異体のアミノ酸配列は,通常の野生型ヒト神経栄養因子のアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。ヒト以外の動物種の神経栄養因子についても同様である。 The amino acid sequence of the human neurotrophic factor mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of a normal wild-type human neurotrophic factor. The same applies to neurotrophic factors of animal species other than humans.
 野生型ヒト神経栄養因子のアミノ酸配列中のアミノ酸の他のアミノ酸への置換換は,好ましくは保存的アミノ酸置換である。ヒト以外の動物種の神経栄養因子についても同様である。 The substitution of an amino acid in the amino acid sequence of wild-type human neurotrophic factor with another amino acid is preferably a conservative amino acid substitution. The same applies to neurotrophic factors of animal species other than humans.
 上記の野生型又は変異型のヒト神経栄養因子であって,これを構成するアミノ酸が糖鎖により修飾されたものもヒト神経栄養因子である。また,上記の野生型又は変異型のヒト神経栄養因子であって,これを構成するアミノ酸がリン酸により修飾されたものもヒト神経栄養因子である。また,糖鎖及びリン酸以外のものにより修飾されたものもヒト神経栄養因子である。また,上記の野生型又は変異型のヒト神経栄養因子であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもヒト神経栄養因子である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種の神経栄養因子についても同様である。  The above wild-type or mutant human neurotrophic factors in which the constituent amino acids are modified with sugar chains are also human neurotrophic factors. The above wild-type or mutant human neurotrophic factors in which the constituent amino acids are modified with phosphate are also human neurotrophic factors. In addition, human neurotrophic factors modified with something other than sugar chains and phosphate are also human neurotrophic factors. In addition, the above wild-type or mutant human neurotrophic factors in which the side chains of the constituent amino acids have been converted by a substitution reaction or the like are also human neurotrophic factors. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same applies to neurotrophic factors of animal species other than humans.
 つまり,糖鎖により修飾されたヒト神経栄養因子は,元のアミノ酸配列を有するヒト神経栄養因子に含まれるものとする。また,リン酸により修飾されたヒト神経栄養因子は,元のアミノ酸配列を有するヒト神経栄養因子に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するヒト神経栄養因子に含まれるものとする。また,ヒト神経栄養因子を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するヒト神経栄養因子に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種の神経栄養因子についても同様である。 In other words, human neurotrophic factors modified with sugar chains are included in human neurotrophic factors with the original amino acid sequence. Human neurotrophic factors modified with phosphate are included in human neurotrophic factors with the original amino acid sequence. Human neurotrophic factors modified with things other than sugar chains and phosphate are also included in human neurotrophic factors with the original amino acid sequence. Human neurotrophic factors in which the side chains of the amino acids that make up human neurotrophic factors have been converted by substitution reactions or the like are also included in human neurotrophic factors with the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same applies to neurotrophic factors of animal species other than humans.
 本明細書において,単に「ヒト脳由来神経栄養因子」,「ヒトBDNF」,又は「hBDNF」というときは,配列番号60で示される119個のアミノ酸残基からなる通常の野生型のhBDNFに加え,標的細胞表面上にある特異的受容体TrkBに結合し,神経細胞の発生,成長,維持,及び再生を促進させる神経栄養因子としての生理活性を有する等のhBDNFとしての機能を有するものである限り,配列番号60で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するhBDNFの変異体も特に区別することなく包含する。野生型のhBDNFは,例えば,配列番号61で示される塩基配列を有する遺伝子にコードされる。ヒト以外の動物種のBDNFについても同様である。 In this specification, when simply referring to "human brain-derived neurotrophic factor," "human BDNF," or "hBDNF," it includes not only the normal wild-type hBDNF consisting of 119 amino acid residues as shown in SEQ ID NO: 60, but also includes, without any particular distinction, mutants of hBDNF in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence as shown in SEQ ID NO: 60, as long as they have the functions of hBDNF, such as binding to the specific receptor TrkB on the surface of target cells and having physiological activity as a neurotrophic factor that promotes the development, growth, maintenance, and regeneration of nerve cells. Wild-type hBDNF is, for example, encoded by a gene having the base sequence as shown in SEQ ID NO: 61. The same applies to BDNF of animal species other than humans.
 なお,ここで,hBDNFがhBDNFとしての機能を有するというときは,hBDNFが,通常の野生型のhBDNFの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。ここで比活性とは蛋白質の質量当たりの生理活性のことをいう。なお,hBDNFとその他の蛋白質との融合蛋白質の比活性は,融合蛋白質の中でhBDNFに相当する部分の質量当たりの生理活性として求められる。なお,ここで融合蛋白質におけるhBDNFの比活性は,当該融合蛋白質の単位質量当たりのhBDNFの生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhBDNFに相当する部分の分子量)を乗じて算出される。ヒト以外の動物種のBDNFについても同様である。 When hBDNF is said to have the function of hBDNF, it means that hBDNF preferably retains a specific activity of 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, of the specific activity of normal wild-type hBDNF taken as 100%. Here, specific activity refers to the physiological activity per mass of protein. The specific activity of a fusion protein of hBDNF and another protein is determined as the physiological activity per mass of the portion of the fusion protein that corresponds to hBDNF. The specific activity of hBDNF in the fusion protein is calculated by multiplying the physiological activity of hBDNF per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hBDNF). The same applies to BDNF of animal species other than humans.
 野生型のhBDNFのアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhBDNFのアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhBDNFのN末端又はC末端のアミノ酸残基を1個欠失させた118個のアミノ酸残基からなるhBDNFの変異体,野生型のhBDNFのN末端又はC末端のアミノ酸残基を2個欠失させた117個のアミノ酸残基からなるhBDNFの変異体等も,hBDNFである。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型のhBDNFのアミノ酸配列に加えることもできる。ヒト以外の動物種のBDNFについても同様である。 When amino acid residues in the amino acid sequence of wild-type hBDNF are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of wild-type hBDNF are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. hBDNF mutants consisting of 118 amino acid residues obtained by deleting one amino acid residue at the N-terminus or C-terminus of wild-type hBDNF, and hBDNF mutants consisting of 117 amino acid residues obtained by deleting two amino acid residues at the N-terminus or C-terminus of wild-type hBDNF are also hBDNF. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hBDNF. The same applies to BDNF of animal species other than humans.
 野生型のhBDNFのアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はhBDNFのアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型のhBDNFのアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型のhBDNFのアミノ酸配列に加えることもできる。ヒト以外の動物種のBDNFについても同様である。 When amino acid residues are added to the amino acid sequence of wild-type hBDNF, one or more amino acid residues are added into the amino acid sequence of hBDNF or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hBDNF, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hBDNF. The same applies to BDNF of animal species other than humans.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型のhBDNFのアミノ酸配列に加えることもできる。例えば,配列番号60で示される野生型のhBDNFのアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもhBDNFであり,配列番号60で示される野生型のhBDNFのアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもhBDNFであり,配列番号60で示される野生型のhBDNFのアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもhBDNFであり,配列番号60で示される野生型のhBDNFのアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもhBDNFであり,配列番号60で示される野生型のhBDNFのアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもhBDNFである。ヒト以外の動物種のBDNFについても同様である。 Furthermore, a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of wild-type hBDNF. For example, a wild-type hBDNF shown in SEQ ID NO:60 in which 1 to 10 amino acid residues have been deleted, 1 to 10 amino acid residues have been replaced with other amino acid residues, and 1 to 10 amino acid residues have been added is also hBDNF, a wild-type hBDNF shown in SEQ ID NO:60 in which 1 to 5 amino acid residues have been deleted, 1 to 5 amino acid residues have been replaced with other amino acid residues, and 1 to 5 amino acid residues have been added is also hBDNF, and a wild-type hBDNF shown in SEQ ID NO:60 in which 1 to 3 amino acid residues have been deleted, hBDNF is also obtained by deleting one or two amino acid residues from the amino acid sequence of wild-type hBDNF shown in SEQ ID NO:60, substituting one or two amino acid residues with other amino acid residues, and adding one or two amino acid residues. hBDNF is also obtained by deleting one amino acid residue from the amino acid sequence of wild-type hBDNF shown in SEQ ID NO:60, substituting one amino acid residue with other amino acid residues, and adding one amino acid residue. The same applies to BDNF of animal species other than humans.
 hBDNF変異体における通常の野生型hBDNFと比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両hBDNFのアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種のBDNFについても同様である。 The location and type (deletion, substitution, and addition) of each mutation in the hBDNF mutant compared to normal wild-type hBDNF can be easily confirmed by aligning the amino acid sequences of both hBDNFs. The same is true for BDNFs of animal species other than humans.
 hBDNF変異体のアミノ酸配列は,配列番号60で示される通常の野生型hBDNFのアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The amino acid sequence of the hBDNF mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type hBDNF shown in SEQ ID NO:60.
 本明細書において,単に「ヒト神経成長因子」,「ヒトNGF」,又は「hNGF」というときは,配列番号62で示される120個のアミノ酸残基からなる通常の野生型のhNGFに加え,神経軸策の伸長及び神経伝達物質の合成促進作用,神経細胞の維持作用,細胞損傷時の修復作用,及び脳神経の機能回復を促し老化を防止する作用等の神経栄養因子としての生理活性を有する等のhNGFとしての機能を有するものである限り,配列番号62で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するhNGFの変異体も特に区別することなく包含する。野生型のhNGFは,例えば,配列番号63で示される塩基配列を有する遺伝子にコードされる。ヒト以外の動物種のNGFについても同様である。 In this specification, when simply referring to "human nerve growth factor", "human NGF" or "hNGF", it includes, without distinction, not only the usual wild-type hNGF consisting of the 120 amino acid residues shown in SEQ ID NO: 62, but also mutants of hNGF in which one or more amino acid residues have been substituted, deleted and/or added (in this specification, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 62, as long as they have the functions of hNGF, such as the physiological activity as a neurotrophic factor, such as the action of promoting the extension of nerve axons and the synthesis of neurotransmitters, the action of maintaining nerve cells, the action of repairing damaged cells, and the action of promoting the recovery of brain nerve functions and preventing aging. Wild-type hNGF is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 63. The same applies to NGFs of animal species other than humans.
 なお,ここで,hNGFがhNGFとしての機能を有するというときは,hNGFが,通常の野生型のhNGFの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。ここで比活性とは蛋白質の質量当たりの生理活性のことをいう。なお,hNGFとその他の蛋白質との融合蛋白質の比活性は,融合蛋白質の中でhNGFに相当する部分の質量当たりの生理活性として求められる。なお,ここで融合蛋白質におけるhNGFの比活性は,当該融合蛋白質の単位質量当たりのhNGFの生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhNGFに相当する部分の分子量)を乗じて算出される。ヒト以外の動物種のNGFについても同様である。 When hNGF is said to have the function of hNGF, it means that hNGF preferably retains a specific activity of 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, of the specific activity of normal wild-type hNGF taken as 100%. Here, specific activity refers to the physiological activity per mass of protein. The specific activity of a fusion protein of hNGF and another protein is determined as the physiological activity per mass of the portion of the fusion protein that corresponds to hNGF. The specific activity of hNGF in the fusion protein is calculated by multiplying the physiological activity of hNGF per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hNGF). The same applies to NGFs of animal species other than humans.
 野生型のhNGFのアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhNGFのアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhNGFのN末端又はC末端のアミノ酸残基を1個欠失させた119個のアミノ酸残基からなるhNGFの変異体,野生型のhNGFのN末端又はC末端のアミノ酸残基を2個欠失させた118個のアミノ酸残基からなるhNGFの変異体等も,hNGFである。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型のhNGFのアミノ酸配列に加えることもできる。ヒト以外の動物種のNGFについても同様である。 When amino acid residues in the amino acid sequence of wild-type hNGF are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of wild-type hNGF are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. hNGF mutants consisting of 119 amino acid residues obtained by deleting one amino acid residue at the N-terminus or C-terminus of wild-type hNGF, and hNGF mutants consisting of 118 amino acid residues obtained by deleting two amino acid residues at the N-terminus or C-terminus of wild-type hNGF, are also hNGF. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hNGF. The same applies to NGFs of animal species other than humans.
 野生型のhNGFのアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はhNGFのアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型のhNGFのアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型のhNGFのアミノ酸配列に加えることもできる。ヒト以外の動物種のNGFについても同様である。 When amino acid residues are added to the amino acid sequence of wild-type hNGF, one or more amino acid residues are added into the amino acid sequence of hNGF or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hNGF, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hNGF. The same applies to NGFs of animal species other than humans.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型のhNGFのアミノ酸配列に加えることもできる。例えば,配列番号62で示される野生型のhNGFのアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもhNGFであり,配列番号62で示される野生型のhNGFのアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもhNGFであり,配列番号62で示される野生型のhNGFのアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもhNGFであり,配列番号62で示される野生型のhNGFのアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもhNGFであり,配列番号62で示される野生型のhNGFのアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもhNGFである。ヒト以外の動物種のNGFについても同様である。 Furthermore, a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of wild-type hNGF. For example, a wild-type hNGF shown in SEQ ID NO:62 in which 1 to 10 amino acid residues have been deleted, 1 to 10 amino acid residues have been replaced with other amino acid residues, and 1 to 10 amino acid residues have been added is also an hNGF; a wild-type hNGF shown in SEQ ID NO:62 in which 1 to 5 amino acid residues have been deleted, 1 to 5 amino acid residues have been replaced with other amino acid residues, and 1 to 5 amino acid residues have been added is also an hNGF; a wild-type hNGF shown in SEQ ID NO:62 in which 1 to 3 amino acid residues have been deleted, hNGF is also obtained by deleting one or two amino acid residues from the amino acid sequence of wild-type hNGF shown in SEQ ID NO:62, substituting one or two amino acid residues with other amino acid residues, and adding one or two amino acid residues. hNGF is also obtained by deleting one amino acid residue from the amino acid sequence of wild-type hNGF shown in SEQ ID NO:62, substituting one amino acid residue with other amino acid residues, and adding one amino acid residue. The same applies to NGFs of animal species other than humans.
 hNGF変異体における通常の野生型hNGFと比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両hNGFのアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種のNGFについても同様である。 The location and type (deletion, substitution, and addition) of each mutation in the hNGF mutant compared to normal wild-type hNGF can be easily confirmed by aligning the amino acid sequences of both hNGFs. The same is true for NGFs of animal species other than humans.
 hNGF変異体のアミノ酸配列は,配列番号62で示される通常の野生型hNGFのアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The amino acid sequence of the hNGF mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type hNGF shown in SEQ ID NO:62.
 本明細書において,単に「ヒトニューロトロフィン-3」,「ヒトNT-3」,又は「hNT-3」というときは,配列番号64で示される119個のアミノ酸残基からなる通常の野生型のhNT-3に加え,TrkCを介して作用し神経細胞の生存維持ならびに分化促進に必要な神経成長因子としての生理活性を有する等のhNT-3としての機能を有するものである限り,配列番号64で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するhNT-3の変異体も特に区別することなく包含する。野生型のhNT-3は,例えば,配列番号65で示される塩基配列を有する遺伝子にコードされる。ヒト以外の動物種のhNT-3についても同様である。 In this specification, when simply referring to "human neurotrophin-3", "human NT-3", or "hNT-3", it includes, without distinction, not only the usual wild-type hNT-3 consisting of the 119 amino acid residues shown in SEQ ID NO: 64, but also mutants of hNT-3 in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 64, as long as they have the functions of hNT-3, such as having physiological activity as a nerve growth factor that acts via TrkC and is necessary for maintaining the survival and promoting differentiation of nerve cells. Wild-type hNT-3 is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 65. The same applies to hNT-3 of animal species other than humans.
 なお,ここで,hNT-3がhNT-3としての機能を有するというときは,hNT-3が,通常の野生型のhNT-3の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。ここで比活性とは蛋白質の質量当たりの生理活性のことをいう。なお,hNT-3とその他の蛋白質との融合蛋白質の比活性は,融合蛋白質の中でhNT-3に相当する部分の質量当たりの生理活性として求められる。なお,ここで融合蛋白質におけるhNT-3の比活性は,当該融合蛋白質の単位質量当たりのhNT-3の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhNT-3に相当する部分の分子量)を乗じて算出される。ヒト以外の動物種のhNT-3についても同様である。 When hNT-3 is said to have the function of hNT-3, it means that hNT-3 preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of normal wild-type hNT-3 is taken as 100%. Here, specific activity refers to the physiological activity per mass of the protein. The specific activity of a fusion protein of hNT-3 and another protein is determined as the physiological activity per mass of the portion of the fusion protein that corresponds to hNT-3. Here, the specific activity of hNT-3 in the fusion protein is calculated by multiplying the physiological activity of hNT-3 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hNT-3). The same applies to hNT-3 from animal species other than humans.
 野生型のhNT-3のアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhNT-3のアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhNT-3のN末端又はC末端のアミノ酸残基を1個欠失させた118個のアミノ酸残基からなるhNT-3の変異体,野生型のhNT-3のN末端又はC末端のアミノ酸残基を2個欠失させた117個のアミノ酸残基からなるhNT-3の変異体等も,hNT-3である。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型のhNT-3のアミノ酸配列に加えることもできる。ヒト以外の動物種のhNT-3についても同様である。 When amino acid residues in the amino acid sequence of wild-type hNT-3 are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of wild-type hNT-3 are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. hNT-3 mutants consisting of 118 amino acid residues obtained by deleting one amino acid residue at the N-terminus or C-terminus of wild-type hNT-3, and hNT-3 mutants consisting of 117 amino acid residues obtained by deleting two amino acid residues at the N-terminus or C-terminus of wild-type hNT-3, etc. are also hNT-3. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hNT-3. The same applies to hNT-3 of animal species other than humans.
 野生型のhNT-3のアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はhNT-3のアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型のhNT-3のアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型のhNT-3のアミノ酸配列に加えることもできる。ヒト以外の動物種のhNT-3についても同様である。 When amino acid residues are added to the amino acid sequence of wild-type hNT-3, one or more amino acid residues are added into the amino acid sequence of hNT-3 or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hNT-3, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hNT-3. The same applies to hNT-3 from animal species other than humans.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型のhNT-3のアミノ酸配列に加えることもできる。例えば,配列番号64で示される野生型のhNT-3のアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもhNT-3であり,配列番号64で示される野生型のhNT-3のアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもhNT-3であり,配列番号64で示される野生型のhNT-3のアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもhNT-3であり,配列番号64で示される野生型のhNT-3のアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもhNT-3であり,配列番号64で示される野生型のhNT-3のアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもhNT-3である。ヒト以外の動物種のhNT-3についても同様である。 Furthermore, a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of wild-type hNT-3. For example, a wild-type hNT-3 shown in SEQ ID NO:64 has 1 to 10 amino acid residues deleted, 1 to 10 amino acid residues replaced with other amino acid residues, and 1 to 10 amino acid residues added to it, which is also an hNT-3; a wild-type hNT-3 shown in SEQ ID NO:64 has 1 to 5 amino acid residues deleted, 1 to 5 amino acid residues replaced with other amino acid residues, and 1 to 5 amino acid residues added to it, which is also an hNT-3; a wild-type hNT-3 shown in SEQ ID NO:64 has 1 to 3 amino acid residues deleted, 1 to 10 amino acid residues replaced with other amino acid residues, and 1 to 10 amino acid residues added to it, which is also an hNT-3. hNT-3 also includes a wild-type hNT-3 amino acid sequence shown in SEQ ID NO:64 in which one or two amino acid residues have been deleted, one or two amino acid residues have been replaced with other amino acid residues, and one or two amino acid residues have been added; and hNT-3 also includes a wild-type hNT-3 amino acid sequence shown in SEQ ID NO:64 in which one amino acid residue has been deleted, one amino acid residue has been replaced with other amino acid residue, and one amino acid residue has been added. The same applies to hNT-3 from animal species other than humans.
 hNT-3変異体における通常の野生型hNT-3と比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両hNT-3のアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種のhNT-3についても同様である。 The location and type (deletion, substitution, and addition) of each mutation in the hNT-3 mutant compared to normal wild-type hNT-3 can be easily confirmed by aligning the amino acid sequences of both hNT-3. The same is true for hNT-3 from animal species other than humans.
 hNT-3変異体のアミノ酸配列は,配列番号64で示される通常の野生型hNT-3のアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The amino acid sequence of the hNT-3 mutant preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of the normal wild-type hNT-3 shown in SEQ ID NO:64.
 本明細書において,単に「ヒトニューロトロフィン-4」,「ヒトNT-4」,又は「hNT-4」というときは,配列番号66で示される130個のアミノ酸残基からなる通常の野生型のhNT-4に加え,TrkBを介して作用し神経細胞の生存維持ならびに分化促進に必要な神経成長因子としての生理活性を有する等のhNT-4としての機能を有するものである限り,配列番号66で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,置換,欠失,及び/又は付加(本明細書において,アミノ酸残基の「付加」は,配列の末端又は内部に残基を追加することを意味する。)されたものに相当するhNT-4の変異体も特に区別することなく包含する。野生型のhNT-4は,例えば,配列番号67で示される塩基配列を有する遺伝子にコードされる。ヒト以外の動物種のhNT-4についても同様である。 In this specification, when simply referring to "human neurotrophin-4," "human NT-4," or "hNT-4," it includes, without distinction, not only the usual wild-type hNT-4 consisting of the 130 amino acid residues shown in SEQ ID NO: 66, but also mutants of hNT-4 in which one or more amino acid residues have been substituted, deleted, and/or added (as used herein, "addition" of an amino acid residue means adding a residue to the end or inside of the sequence) to the amino acid sequence shown in SEQ ID NO: 66, as long as they have the functions of hNT-4, such as having physiological activity as a nerve growth factor that acts via TrkB and is necessary for maintaining the survival and promoting differentiation of nerve cells. Wild-type hNT-4 is, for example, encoded by a gene having the base sequence shown in SEQ ID NO: 67. The same applies to hNT-4 of animal species other than humans.
 なお,ここで,hNT-4がhNT-4としての機能を有するというときは,hNT-4が,通常の野生型のhNT-4の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。ここで比活性とは蛋白質の質量当たりの生理活性のことをいう。なお,hNT-4とその他の蛋白質との融合蛋白質の比活性は,融合蛋白質の中でhNT-4に相当する部分の質量当たりの生理活性として求められる。なお,ここで融合蛋白質におけるhNT-4の比活性は,当該融合蛋白質の単位質量当たりのhNT-4の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhNT-4に相当する部分の分子量)を乗じて算出される。ヒト以外の動物種のhNT-4についても同様である。 When hNT-4 is said to have the function of hNT-4, it means that hNT-4 preferably retains a specific activity of 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, of the specific activity of normal wild-type hNT-4 taken as 100%. Here, specific activity refers to the physiological activity per mass of protein. The specific activity of a fusion protein of hNT-4 with another protein is determined as the physiological activity per mass of the portion of the fusion protein that corresponds to hNT-4. The specific activity of hNT-4 in the fusion protein is calculated by multiplying the physiological activity of hNT-4 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to hNT-4). The same applies to hNT-4 from animal species other than humans.
 野生型のhNT-4のアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhNT-4のアミノ酸配列中のアミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。野生型のhNT-4のN末端又はC末端のアミノ酸残基を1個欠失させた129個のアミノ酸残基からなるhNT-4の変異体,野生型のhNT-4のN末端又はC末端のアミノ酸残基を2個欠失させた128個のアミノ酸残基からなるhNT-4の変異体等も,hNT-4である。また,これらアミノ酸残基の置換と欠失を組み合わせた変異を野生型のhNT-4のアミノ酸配列に加えることもできる。ヒト以外の動物種のhNT-4についても同様である。 When amino acid residues in the amino acid sequence of wild-type hNT-4 are replaced with other amino acid residues, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When amino acid residues in the amino acid sequence of wild-type hNT-4 are deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. hNT-4 mutants consisting of 129 amino acid residues obtained by deleting one amino acid residue at the N-terminus or C-terminus of wild-type hNT-4, and hNT-4 mutants consisting of 128 amino acid residues obtained by deleting two amino acid residues at the N-terminus or C-terminus of wild-type hNT-4, etc. are also hNT-4. Mutations that combine the substitution and deletion of these amino acid residues can also be added to the amino acid sequence of wild-type hNT-4. The same applies to hNT-4 of animal species other than humans.
 野生型のhNT-4のアミノ酸配列にアミノ酸残基を付加する場合,アミノ酸残基はhNT-4のアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。このとき付加されるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。また,これらアミノ酸残基の付加と上記の置換を組み合わせた変異を野生型のhNT-4のアミノ酸配列に加えることもでき,これらアミノ酸残基の付加と上記の欠失を組み合わせた変異を野生型のhNT-4のアミノ酸配列に加えることもできる。ヒト以外の動物種のhNT-4についても同様である。 When amino acid residues are added to the amino acid sequence of wild-type hNT-4, one or more amino acid residues are added into the amino acid sequence of hNT-4 or to the N-terminus or C-terminus of the amino acid sequence. The number of amino acid residues added in this case is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. Mutations that combine the addition of these amino acid residues with the above-mentioned substitutions can also be added to the amino acid sequence of wild-type hNT-4, and mutations that combine the addition of these amino acid residues with the above-mentioned deletions can also be added to the amino acid sequence of wild-type hNT-4. The same applies to hNT-4 from animal species other than humans.
 更に,上記のアミノ酸残基の置換及び欠失,及び付加の3種類を組み合わせた変異を野生型のhNT-4のアミノ酸配列に加えることもできる。例えば,配列番号66で示される野生型のhNT-4のアミノ酸配列に対し,1~10個のアミノ酸残基を欠失させ,1~10個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~10個のアミノ酸残基を付加させたものもhNT-4であり,配列番号66で示される野生型のhNT-4のアミノ酸配列に対し,1~5個のアミノ酸残基を欠失させ,1~5個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~5個のアミノ酸残基を付加させたものもhNT-4であり,配列番号66で示される野生型のhNT-4のアミノ酸配列に対し,1~3個のアミノ酸残基を欠失させ,1~3個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1~3個のアミノ酸残基を付加させたものもhNT-4であり,配列番号66で示される野生型のhNT-4のアミノ酸配列に対し,1又は2個のアミノ酸残基を欠失させ,1又は2個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1又は2個のアミノ酸残基を付加させたものもhNT-4であり,配列番号66で示される野生型のhNT-4のアミノ酸配列に対し,1個のアミノ酸残基を欠失させ,1個のアミノ酸残基を他のアミノ酸残基へ置換させ,及び1個のアミノ酸残基を付加させたものもhNT-4である。ヒト以外の動物種のhNT-4についても同様である。 Furthermore, a combination of the above three types of mutations, substitution, deletion, and addition of amino acid residues, can be added to the amino acid sequence of wild-type hNT-4. For example, a wild-type hNT-4 amino acid sequence shown in SEQ ID NO:66 in which 1 to 10 amino acid residues have been deleted, 1 to 10 amino acid residues have been replaced with other amino acid residues, and 1 to 10 amino acid residues have been added is also an hNT-4; a wild-type hNT-4 amino acid sequence shown in SEQ ID NO:66 in which 1 to 5 amino acid residues have been deleted, 1 to 5 amino acid residues have been replaced with other amino acid residues, and 1 to 5 amino acid residues have been added is also an hNT-4; a wild-type hNT-4 amino acid sequence shown in SEQ ID NO:66 in which 1 to 3 amino acid residues have been deleted, hNT-4 also includes a wild-type hNT-4 amino acid sequence shown in SEQ ID NO:66 in which one or two amino acid residues have been deleted, one or two amino acid residues have been replaced with other amino acid residues, and one or two amino acid residues have been added; and hNT-4 also includes a wild-type hNT-4 amino acid sequence shown in SEQ ID NO:66 in which one amino acid residue has been deleted, one amino acid residue has been replaced with other amino acid residue, and one amino acid residue has been added. The same applies to hNT-4 from animal species other than humans.
 hNT-4変異体における通常の野生型hNT-4と比較したときの各変異の位置及びその形式(欠失,置換,及び付加)は,両hNT-4のアミノ酸配列のアラインメントにより,容易に確認することができる。ヒト以外の動物種のhNT-4についても同様である。 The location and type (deletion, substitution, and addition) of each mutation in the hNT-4 mutant compared to normal wild-type hNT-4 can be easily confirmed by aligning the amino acid sequences of both hNT-4. The same is true for hNT-4 from animal species other than humans.
 hNT-4変異体のアミノ酸配列は,配列番号66で示される通常の野生型hNT-4のアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The amino acid sequence of the hNT-4 mutant preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, to the amino acid sequence of normal wild-type hNT-4 shown in SEQ ID NO:66.
 野生型hBDNF又のアミノ酸配列中のアミノ酸の他のアミノ酸への置換,野生型hNGFのアミノ酸配列中のアミノ酸の他のアミノ酸への置換,野生型hNT-3のアミノ酸配列中のアミノ酸の他のアミノ酸への置換,野生型hNT-4のアミノ酸配列中のアミノ酸の他のアミノ酸への置換は,例えば,保存的アミノ酸置換である。ヒト以外の動物種のこれらの神経栄養因子についても同様のことがいえる。  Substitutions of amino acids in the amino acid sequence of wild-type hBDNF or wild-type hNGF, wild-type hNT-3, and wild-type hNT-4 are, for example, conservative amino acid substitutions. The same is true for these neurotrophic factors of animal species other than humans.
 上記の野生型又は変異型のhBDNF,hNGF,hNT-3,又はhNT-4であって,これを構成するアミノ酸が糖鎖により修飾されたものも,それぞれhBDNF,hNGF,hNT-3,又はhNT-4である。また,これらの野生型又は変異型のヒト神経栄養因子であって,これを構成するアミノ酸がリン酸により修飾されたものについても同様である。また,糖鎖及びリン酸以外のものにより修飾されたものについても同様である。また,これらの野生型又は変異型のヒト神経栄養因子であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものについても同様である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。hBDNF,hNGF,hNT-3,及びhNT-4についても同様のことがいえる。また,ヒト以外の動物種のこれらの神経栄養因子についても同様のことがいえる。  The above wild-type or mutant hBDNF, hNGF, hNT-3, or hNT-4, in which the constituent amino acids are modified with sugar chains, are also hBDNF, hNGF, hNT-3, or hNT-4, respectively. The same applies to these wild-type or mutant human neurotrophic factors in which the constituent amino acids are modified with phosphate. The same applies to those modified with something other than sugar chains and phosphate. The same applies to these wild-type or mutant human neurotrophic factors in which the side chains of the constituent amino acids have been changed by substitution reactions, etc. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same applies to hBDNF, hNGF, hNT-3, and hNT-4. The same applies to these neurotrophic factors of animal species other than humans.
 つまり,糖鎖により修飾されたhBDNF,hNGF,hNT-3,又はhNT-4は,元のアミノ酸配列を有するhBDNF,hNGF,hNT-3,又はhNT-4に,それぞれ含まれるものとする。また,リン酸により修飾されたこれらのヒト神経栄養因子についても同様である。また,糖鎖及びリン酸以外のものにより修飾されたものについても同様である。また,これらのヒト神経栄養因子を構成するアミノ酸の側鎖が,置換反応等により変換したものについても同様である。かかる変換としては,システイン残基のホルミルグリシンへの変換がある。また,ヒト以外の動物種の神経栄養因子についても同様のことがいえる。 In other words, hBDNF, hNGF, hNT-3, or hNT-4 modified with sugar chains are included in hBDNF, hNGF, hNT-3, or hNT-4 having the original amino acid sequence, respectively. The same applies to these human neurotrophic factors modified with phosphate. The same applies to those modified with things other than sugar chains and phosphate. The same also applies to those in which the side chains of the amino acids that make up these human neurotrophic factors have been changed by substitution reactions, etc. One such conversion is the conversion of a cysteine residue to formylglycine. The same also applies to neurotrophic factors of animal species other than humans.
 本発明の一実施形態は,野生型の神経栄養因子をHSAと融合させた組換え蛋白質である。かかる融合蛋白質は,CHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型ヒト神経栄養因子を同様の手法により組換え蛋白質として発現させたときと比較して,培養上清中のヒト神経栄養因子としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となるようにすることができる。 One embodiment of the present invention is a recombinant protein in which a wild-type neurotrophic factor is fused with HSA. When such a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of expression of human neurotrophic factor in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc., compared to when wild-type human neurotrophic factor is expressed as a recombinant protein by a similar method.
 また,本発明の一実施形態は,野生型hBDNFをHSAと融合させた組換え蛋白質である。かかる融合蛋白質は,CHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型hBDNFを同様の手法により組換え蛋白質として発現させたときと比較して,培養上清中のhBDNFとしての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となるようにすることができる。 Another embodiment of the present invention is a recombinant protein in which wild-type hBDNF is fused with HSA. When such a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hBDNF expressed in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc., compared to when wild-type hBDNF is expressed as a recombinant protein by a similar method.
 また,本発明の一実施形態は,野生型hNGFをHSAと融合させた組換え蛋白質である。かかる融合蛋白質は,CHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型hNGFを同様の手法により組換え蛋白質として発現させたときと比較して,培養上清中のhNGFとしての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となるようにすることができる。 Another embodiment of the present invention is a recombinant protein in which wild-type hNGF is fused with HSA. When such a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hNGF expressed in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc., compared to when wild-type hNGF is expressed as a recombinant protein by a similar method.
 また,本発明の一実施形態は,野生型hNT-3をHSAと融合させた組換え蛋白質である。かかる融合蛋白質は,CHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型hNT-3を同様の手法により組換え蛋白質として発現させたときと比較して,培養上清中のhNT-3としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となるようにすることができる。 Another embodiment of the present invention is a recombinant protein in which wild-type hNT-3 is fused with HSA. When such a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of expression of hNT-3 in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc., compared to when wild-type hNT-3 is expressed as a recombinant protein by a similar method.
 また,本発明の一実施形態は,野生型hNT-4をHSAと融合させた組換え蛋白質である。かかる融合蛋白質は,CHO等の宿主細胞を用いて組換え蛋白質として発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,野生型hNT-4を同様の手法により組換え蛋白質として発現させたときと比較して,培養上清中のhNT-4としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となるようにすることができる。 Another embodiment of the present invention is a recombinant protein in which wild-type hNT-4 is fused with HSA. When such a fusion protein is expressed as a recombinant protein using a host cell such as CHO, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hNT-4 expressed in the culture supernatant can be at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, for example, 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc., compared to when wild-type hNT-4 is expressed as a recombinant protein by a similar method.
 本発明の一実施形態は,ヒト神経栄養因子のアミノ酸配列を含むポリペプチドと,SAのアミノ酸配列を含むポリペプチドとを結合させた,融合蛋白質に関する。ここで,「ポリペプチドを結合させる」というときは,直接又はリンカーを介して間接的に,共有結合により異なるポリペプチドを結合させることをいう。ここで,神経栄養因子は好ましくはヒト神経栄養因子である。また,SAは好ましくはHSAである。また,ヒト神経栄養因子は,例えばhBDNF,hNGF,hNT-3,又はhNT-4である。ヒト以外の動物種の神経栄養因子についても同様である。 One embodiment of the present invention relates to a fusion protein in which a polypeptide containing the amino acid sequence of a human neurotrophic factor is bound to a polypeptide containing the amino acid sequence of SA. Here, the term "binding polypeptides" refers to binding different polypeptides by covalent bonding, either directly or indirectly via a linker. Here, the neurotrophic factor is preferably human neurotrophic factor. Also, SA is preferably HSA. Also, the human neurotrophic factor is, for example, hBDNF, hNGF, hNT-3, or hNT-4. The same applies to neurotrophic factors of animal species other than humans.
 2つの異なるポリペプチドを結合させる方法としては,例えば,一方のポリペプチドをコードする遺伝子の下流に,インフレームで他方のポリペプチドをコードする遺伝子を結合させたDNA断片を組み込んだ発現ベクターを作製し,この発現ベクターを用いて形質転換させた宿主細胞を培養することにより,組換え蛋白質として発現させる方法が一般的である。得られた組換え蛋白質は,2つのポリペプチドが,直接又は別のアミノ酸配列を介してペプチド結合した,一本鎖ポリペプチドである。 A common method for linking two different polypeptides is, for example, to create an expression vector incorporating a DNA fragment in which a gene encoding one polypeptide is linked in-frame downstream of a gene encoding the other polypeptide, and then to express the polypeptide as a recombinant protein by culturing a host cell transformed with this expression vector. The resulting recombinant protein is a single-chain polypeptide in which the two polypeptides are peptide-linked, either directly or via different amino acid sequences.
 本発明の一実施形態において,「SA-ヒト神経栄養因子融合蛋白質」又は「SA-ヒト神経栄養因子」の語は,SAのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト神経栄養因子のアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒト神経栄養因子としての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。SA-ヒト神経栄養因子融合蛋白質において,ヒト神経栄養因子がヒト神経栄養因子としての機能を有するというときは,ヒト神経栄養因子が,通常の野生型のヒト神経栄養因子の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでSA-ヒト神経栄養因子融合蛋白質におけるヒト神経栄養因子の比活性は,当該融合蛋白質の単位質量当たりのヒト神経栄養因子の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のヒト神経栄養因子に相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "SA-human neurotrophic factor fusion protein" or "SA-human neurotrophic factor" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of human neurotrophic factor is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and which has the function of a human neurotrophic factor. Here, the animal species of SA is not particularly limited, but preferably it is a mammalian SA, more preferably it is a primate SA, and even more preferably it is HSA. When it is said that the human neurotrophic factor in the SA-human neurotrophic factor fusion protein has the function of a human neurotrophic factor, it means that the human neurotrophic factor preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of a normal wild-type human neurotrophic factor is taken as 100%. Here, the specific activity of human neurotrophic factor in the SA-human neurotrophic factor fusion protein is calculated by multiplying the physiological activity of human neurotrophic factor per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein that corresponds to human neurotrophic factor).
 本発明の一実施形態において,「HSA-ヒト神経栄養因子融合蛋白質」又は「HSA-ヒト神経栄養因子」の語は,HSAのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト神経栄養因子のアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒト神経栄養因子としての機能を有するものを示す。ここでHSA-ヒト神経栄養因子融合蛋白質がヒト神経栄養因子としての機能を有するというときは,上記のSA-神経栄養因子融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "HSA-human neurotrophic factor fusion protein" or "HSA-human neurotrophic factor" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of human neurotrophic factor is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of a human neurotrophic factor. When it is said here that the HSA-human neurotrophic factor fusion protein has the function of a human neurotrophic factor, the definition of the SA-neurotrophic factor fusion protein described above can be applied.
 SA-ヒト神経栄養因子融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。HSA-ヒト神経栄養因子融合蛋白質においても同様である。HSA-hBDNF融合蛋白質,HSA-hNGF融合蛋白質,HSA-hNT-3融合蛋白質,及びHSA-hNT-4融合蛋白質についても同様である。 In the SA-human neurotrophic factor fusion protein, it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this. The same is true for the HSA-human neurotrophic factor fusion protein. The same is true for the HSA-hBDNF fusion protein, HSA-hNGF fusion protein, HSA-hNT-3 fusion protein, and HSA-hNT-4 fusion protein.
 野生型HSAと野生型ヒト神経栄養因子との融合蛋白質であるHSA-ヒト神経栄養因子融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えヒト神経栄養因子部分には変異を加えないことも,HSA部分には変異を加えずにヒト神経栄養因子部分にのみ変異を加えることも,また,HSA部分とヒト神経栄養因子部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。ヒト神経栄養因子部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型ヒト神経栄養因子に変異が加えられたヒト神経栄養因子のアミノ酸配列となる。HSA部分とヒト神経栄養因子部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,ヒト神経栄養因子部分のアミノ酸配列は上述した野生型ヒト神経栄養因子に変異が加えられたヒト神経栄養因子のアミノ酸配列となる。ヒト以外の動物種の野生型SAと野生型ヒト神経栄養因子との融合蛋白質についても同様のことがいえる。 When mutations are added to an HSA-human neurotrophic factor fusion protein, which is a fusion protein of wild-type HSA and wild-type human neurotrophic factor, mutations can be added only to the HSA portion without mutations in the human neurotrophic factor portion, mutations can be added only to the human neurotrophic factor portion without mutations in the HSA portion, or mutations can be added to both the HSA portion and the human neurotrophic factor portion. When mutations are added only to the HSA portion, the amino acid sequence of the portion is the amino acid sequence of the wild-type HSA with a mutation added. When mutations are added only to the human neurotrophic factor portion, the amino acid sequence of the portion is the amino acid sequence of the wild-type human neurotrophic factor with a mutation added. When mutations are added to both the HSA portion and the human neurotrophic factor portion, the amino acid sequence of the HSA portion is the amino acid sequence of the wild-type HSA with a mutation added, and the amino acid sequence of the human neurotrophic factor portion is the amino acid sequence of the wild-type human neurotrophic factor with a mutation added. The same can be said about fusion proteins between wild-type SA of non-human animal species and wild-type human neurotrophic factor.
 HSA-ヒト神経栄養因子融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもHSA-ヒト神経栄養因子融合蛋白質である。また,HSA-ヒト神経栄養因子融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもHSA-ヒト神経栄養因子融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもHSA-ヒト神経栄養因子融合蛋白質である。また,HSA-ヒト神経栄養因子融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSA-ヒト神経栄養因子融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとヒト神経栄養因子との融合蛋白質(SA-ヒト神経栄養因子)についても同様のことがいえる。  HSA-human neurotrophic factor fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-human neurotrophic factor fusion proteins. HSA-human neurotrophic factor fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-human neurotrophic factor fusion proteins. HSA-human neurotrophic factor fusion proteins modified with something other than sugar chains and phosphate are also HSA-human neurotrophic factor fusion proteins. HSA-human neurotrophic factor fusion proteins in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like are also HSA-human neurotrophic factor fusion proteins. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of SA of animal species other than humans and human neurotrophic factor (SA-human neurotrophic factor).
 つまり,糖鎖により修飾されたHSA-ヒト神経栄養因子融合蛋白質は,元のアミノ酸配列を有するHSA-ヒト神経栄養因子融合蛋白質に含まれるものとする。また,リン酸により修飾されたHSA-ヒト神経栄養因子融合蛋白質は,元のアミノ酸配列を有するHSA-ヒト神経栄養因子融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSA-ヒト神経栄養因子融合蛋白質に含まれるものとする。また,HSA-ヒト神経栄養因子融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSA-ヒト神経栄養因子融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとヒト神経栄養因子との融合蛋白質(SA-ヒト神経栄養因子)についても同様のことがいえる。 In other words, HSA-human neurotrophic factor fusion proteins modified with sugar chains are included in HSA-human neurotrophic factor fusion proteins having the original amino acid sequence. Also, HSA-human neurotrophic factor fusion proteins modified with phosphate are included in HSA-human neurotrophic factor fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are included in HSA-human neurotrophic factor fusion proteins having the original amino acid sequence. Also, HSA-human neurotrophic factor fusion proteins in which the side chains of the amino acids constituting the HSA-human neurotrophic factor fusion proteins have been converted by substitution reactions or the like are included in HSA-human neurotrophic factor fusion proteins having the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and human neurotrophic factor of animal species other than humans (SA-human neurotrophic factor).
 また,HSA-ヒト神経栄養因子融合蛋白質であって,これを構成するヒト神経栄養因子が,ヒト神経栄養因子の前駆体であるものもHSA-ヒト神経栄養因子融合蛋白質である。ここで前駆体というときは,HSA-ヒト神経栄養因子融合蛋白質として生合成されたものであって,当該生合成後に,ヒト神経栄養因子として機能する部分が,製造過程において又は生体に投与したときに当該生体内において,当該融合蛋白質から切り離されて,単独でもヒト神経栄養因子としての機能を発揮することができるタイプのものをいう。この場合,HSA-ヒト神経栄養因子融合蛋白質は生合成された後に,加水分解酵素等により,特定の箇所で切断され,HSAを含む部分と成熟型ヒト神経栄養因子を含む部分とが分離することがある。この場合,得られるヒト神経栄養因子はHSAとの融合蛋白質ではないが,当該神経栄養因子が製造される過程で,一旦,HSA-ヒト神経栄養因子融合蛋白質が合成されることになる。従って,かかる方法によりヒト神経栄養因子を製造する場合,当該製造方法は,HSA-ヒト神経栄養因子の製造方法に含まれる。ヒト以外の動物種のSAとヒト神経栄養因子との融合蛋白質(SA-ヒト神経栄養因子)についても同様のことがいえる。  An HSA-human neurotrophic factor fusion protein in which the human neurotrophic factor constituting the protein is a precursor of human neurotrophic factor is also an HSA-human neurotrophic factor fusion protein. The term "precursor" used here refers to a type of protein that is biosynthesized as an HSA-human neurotrophic factor fusion protein, and in which the portion that functions as a human neurotrophic factor after the biosynthesis is cleaved from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as a human neurotrophic factor by itself. In this case, the HSA-human neurotrophic factor fusion protein may be cleaved at a specific site after biosynthesis by a hydrolase or the like, and the portion containing HSA and the portion containing mature human neurotrophic factor may be separated. In this case, the human neurotrophic factor obtained is not a fusion protein with HSA, but the HSA-human neurotrophic factor fusion protein is synthesized once during the process of manufacturing the neurotrophic factor. Therefore, when human neurotrophic factor is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-human neurotrophic factor. The same can be said about fusion proteins of SA from non-human animal species and human neurotrophic factor (SA-human neurotrophic factor).
 本発明の一実施形態において,「SA-hBDNF融合蛋白質」又は「SA-hBDNF」の語は,SAのアミノ酸配列のC末端に,直接又はリンカーを介して,hBDNFのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hBDNFとしての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。SA-hBDNF融合蛋白質において,hBDNFがhBDNFとしての機能を有するというときは,hBDNFが,通常の野生型のhBDNFの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでSA-hBDNF融合蛋白質におけるhBDNFの比活性は,当該融合蛋白質の単位質量当たりのhBDNFの生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhBDNFに相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "SA-hBDNF fusion protein" or "SA-hBDNF" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hBDNF is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and which has the function of hBDNF. Here, the animal species of SA is not particularly limited, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA. When hBDNF in an SA-hBDNF fusion protein is said to have the function of hBDNF, it means that hBDNF retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hBDNF is taken as 100%. Here, the specific activity of hBDNF in the SA-hBDNF fusion protein is calculated by multiplying the physiological activity of hBDNF per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hBDNF).
 本発明の一実施形態において,「HSA-hBDNF融合蛋白質」又は「HSA-hBDNF」の語は,HSAのアミノ酸配列のC末端に,直接又はリンカーを介して,hBDNFのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hBDNFとしての機能を有するものを示す。ここでHSA-hBDNF融合蛋白質がhBDNFとしての機能を有するというときは,上記のSA-hBDNF融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "HSA-hBDNF fusion protein" or "HSA-hBDNF" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hBDNF is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hBDNF. When it is said here that the HSA-hBDNF fusion protein has the function of hBDNF, the definition of the SA-hBDNF fusion protein described above can be applied.
 本発明の一実施形態において好ましいHSA-hBDNF融合蛋白質は,例えば配列番号68で示されるアミノ酸配列を有するものである。配列番号68で示されるHSA-hBDNF融合蛋白質は,野生型HSAのC末端にリンカー配列Gly-Serを介して野生型hBDNFが結合したものである。配列番号68で示されるHSA-hBDNF融合蛋白質は,例えば,配列番号69で示される塩基配列を有する遺伝子にコードされる。また,配列番号68で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hBDNFとしての機能を有するものである限りHSA-hBDNF融合蛋白質に含まれる。HSA-hBDNF融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred HSA-hBDNF fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 68. The HSA-hBDNF fusion protein shown in SEQ ID NO: 68 is obtained by linking wild-type hBDNF to the C-terminus of wild-type HSA via the linker sequence Gly-Ser. The HSA-hBDNF fusion protein shown in SEQ ID NO: 68 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 69. In addition, HSA-hBDNF fusion proteins include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 68 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hBDNF. It is preferable that the HSA-hBDNF fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 野生型HSAと野生型hBDNFとの融合蛋白質であるHSA-hBDNF融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhBDNF部分には変異を加えないことも,HSA部分には変異を加えずにhBDNF部分にのみ変異を加えることも,また,HSA部分とhBDNF部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hBDNF部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hBDNFに変異が加えられたhBDNFのアミノ酸配列となる。HSA部分とhBDNF部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hBDNF部分のアミノ酸配列は上述した野生型hBDNFに変異が加えられたhBDNFのアミノ酸配列となる。配列番号68で示されるアミノ酸配列を有するHSA-hBDNF融合蛋白質についても同様である。ヒト以外の動物種の野生型SAと野生型hBDNFとの融合蛋白質についても同様のことがいえる。 When a mutation is added to an HSA-hBDNF fusion protein, which is a fusion protein of wild-type HSA and wild-type hBDNF, a mutation may be added only to the HSA portion and not to the hBDNF portion, or a mutation may be added only to the hBDNF portion without any mutation to the HSA portion, or a mutation may be added to both the HSA portion and the hBDNF portion. When a mutation is added only to the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When a mutation is added only to the hBDNF portion, the amino acid sequence of the portion is the amino acid sequence of hBDNF obtained by adding a mutation to the wild-type hBDNF described above. When a mutation is added to both the HSA portion and the hBDNF portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hBDNF portion is the amino acid sequence of hBDNF obtained by adding a mutation to the wild-type hBDNF described above. The same applies to the HSA-hBDNF fusion protein having the amino acid sequence shown in SEQ ID NO: 68. The same can be said about fusion proteins of wild-type SA and wild-type hBDNF from non-human animal species.
 配列番号68で示されるアミノ酸配列を有するHSA-hBDNF融合蛋白質に変異を加える場合について,以下例示する。配列番号68で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号68で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。HSA-hBDNF融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hIL-10部分にのみ加えても,又は両方の部分に加えてもよい。 The following is an example of a case where a mutation is made to an HSA-hBDNF fusion protein having the amino acid sequence shown in SEQ ID NO:68. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:68 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:68 or to the N-terminus or C-terminus of the amino acid sequence. The HSA-hBDNF fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA part, only to the hIL-10 part, or to both parts.
 配列番号68で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号68で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:68 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:68.
 HSA-hBDNF融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもHSA-hBDNF融合蛋白質である。また,HSA-hBDNF融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもHSA-hBDNF融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもHSA-hBDNF融合蛋白質である。また,HSA-hBDNF融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSA-hBDNF融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhBDNFとの融合蛋白質(SA-hBDNF)についても同様のことがいえる。  HSA-hBDNF fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hBDNF fusion proteins. HSA-hBDNF fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hBDNF fusion proteins. HSA-hBDNF fusion proteins modified with something other than sugar chains and phosphate are also HSA-hBDNF fusion proteins. HSA-hBDNF fusion proteins in which the side chains of the amino acids constituting the protein have been changed by a substitution reaction or the like are also HSA-hBDNF fusion proteins. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of SA and hBDNF of animal species other than humans (SA-hBDNF).
 つまり,糖鎖により修飾されたHSA-hBDNF融合蛋白質は,元のアミノ酸配列を有するHSA-hBDNF融合蛋白質に含まれるものとする。また,リン酸により修飾されたHSA-hBDNF融合蛋白質は,元のアミノ酸配列を有するHSA-hBDNF融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSA-hBDNF融合蛋白質に含まれるものとする。また,HSA-hBDNF融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSA-hBDNF融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhBDNFとの融合蛋白質(SA-hBDNF)についても同様のことがいえる。 In other words, HSA-hBDNF fusion proteins modified with sugar chains are included in HSA-hBDNF fusion proteins with the original amino acid sequence. Also, HSA-hBDNF fusion proteins modified with phosphate are included in HSA-hBDNF fusion proteins with the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are also included in HSA-hBDNF fusion proteins with the original amino acid sequence. Also, those in which the side chains of the amino acids constituting the HSA-hBDNF fusion protein have been converted by substitution reactions or the like are also included in HSA-hBDNF fusion proteins with the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hBDNF of animal species other than humans (SA-hBDNF).
 また,HSA-hBDNF融合蛋白質であって,これを構成するhBDNFが,hBDNFの前駆体であるものもHSA-hBDNF融合蛋白質である。ここで前駆体というときは,HSA-hBDNF融合蛋白質として生合成されたものであって,当該生合成後に,hBDNFとして機能する部分が,製造過程において又は生体に投与したときに当該生体内において,当該融合蛋白質から切り離されて,単独でもhBDNFとしての機能を発揮することができるタイプのものをいう。この場合,HSA-hBDNF融合蛋白質は生合成された後,加水分解酵素等により,特定の箇所で切断され,HSAを含む部分と成熟型のhBDNFを含む部分とが分離することがある。得られるhBDNFはHSAとの融合蛋白質ではないが,hBDNFが製造される過程で,一旦,HSA-hBDNF融合蛋白質が合成されることになる。従って,かかる方法によりhBDNFを製造する場合,当該製造方法は,HSA-hBDNF融合蛋白質の製造方法に含まれる。ヒト以外の動物種のSAとhBDNFとの融合蛋白質(SA-hBDNF)についても同様のことがいえる。  Also, HSA-hBDNF fusion proteins in which the constituent hBDNF is a precursor of hBDNF are also HSA-hBDNF fusion proteins. Here, the term "precursor" refers to a type that is biosynthesized as an HSA-hBDNF fusion protein, and after the biosynthesis, the part that functions as hBDNF is separated from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hBDNF by itself. In this case, after the HSA-hBDNF fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the part containing HSA and the part containing mature hBDNF may be separated. The obtained hBDNF is not a fusion protein with HSA, but the HSA-hBDNF fusion protein is synthesized once during the process of manufacturing hBDNF. Therefore, when hBDNF is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-hBDNF fusion protein. The same can be said for a fusion protein of SA and hBDNF of an animal species other than human (SA-hBDNF).
 本発明の一実施形態において,「SA-hNGF融合蛋白質」又は「SA-hNGF」の語は,SAのアミノ酸配列のC末端に,直接又はリンカーを介して,hNGFのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hNGFとしての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。SA-hNGF融合蛋白質において,hNGFがhNGFとしての機能を有するというときは,hNGFが,通常の野生型のhNGFの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでSA-hNGF融合蛋白質におけるhNGFの比活性は,当該融合蛋白質の単位質量当たりのhNGFの生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhNGFに相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "SA-hNGF fusion protein" or "SA-hNGF" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNGF is linked to the C-terminus of the amino acid sequence of SA, either directly or via a linker, and having the function of hNGF. Here, the animal species of SA is not particularly limited, but preferably it is a mammalian SA, more preferably it is a primate SA, and even more preferably it is HSA. When it is said that hNGF in an SA-hNGF fusion protein has the function of hNGF, it means that hNGF retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hNGF is taken as 100%. Here, the specific activity of hNGF in the SA-hNGF fusion protein is calculated by multiplying the physiological activity of hNGF per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hNGF).
 本発明の一実施形態において,「HSA-hNGF融合蛋白質」又は「HSA-hNGF」の語は,HSAのアミノ酸配列のC末端に,直接又はリンカーを介して,hNGFのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hNGFとしての機能を有するものを示す。ここでHSA-hNGF融合蛋白質がhNGFとしての機能を有するというときは,上記のSA-hNGF融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "HSA-hNGF fusion protein" or "HSA-hNGF" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNGF is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hNGF. When it is said here that the HSA-hNGF fusion protein has the function of hNGF, the definition of the SA-hNGF fusion protein described above can be applied.
 本発明の一実施形態において好ましいHSA-hNGF融合蛋白質は,例えば配列番号70で示されるアミノ酸配列を有するものである。配列番号70で示されるHSA-hNGF融合蛋白質は,野生型HSAのC末端にリンカー配列Gly-Serを介して野生型hNGFが結合したものである。配列番号70で示されるHSA-hNGF融合蛋白質は,例えば,配列番号71で示される塩基配列を有する遺伝子にコードされる。また,配列番号70で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hNGFとしての機能を有するものである限りHSA-hNGF融合蛋白質に含まれる。HSA-hNGF融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred HSA-hNGF fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 70. The HSA-hNGF fusion protein shown in SEQ ID NO: 70 is obtained by linking wild-type hNGF to the C-terminus of wild-type HSA via the linker sequence Gly-Ser. The HSA-hNGF fusion protein shown in SEQ ID NO: 70 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 71. In addition, HSA-hNGF fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 70 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hNGF. It is preferable that the HSA-hNGF fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 野生型HSAと野生型hNGFとの融合蛋白質であるHSA-hNGF融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhNGF部分には変異を加えないことも,HSA部分には変異を加えずにhNGF部分にのみ変異を加えることも,また,HSA部分とhNGF部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hNGF部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hNGFに変異が加えられたhNGFのアミノ酸配列となる。HSA部分とhNGF部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hNGF部分のアミノ酸配列は上述した野生型hNGFに変異が加えられたhNGFのアミノ酸配列となる。配列番号70で示されるアミノ酸配列を有するHSA-hNGF融合蛋白質についても同様である。ヒト以外の動物種の野生型SAと野生型hNGFとの融合蛋白質についても同様のことがいえる。 When mutations are introduced into an HSA-hNGF fusion protein, which is a fusion protein of wild-type HSA and wild-type hNGF, mutations can be introduced only in the HSA portion and not in the hNGF portion, mutations can be introduced only in the hNGF portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNGF portion. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are introduced only in the hNGF portion, the amino acid sequence of the portion is the amino acid sequence of hNGF obtained by adding a mutation to the wild-type hNGF described above. When mutations are introduced in both the HSA portion and the hNGF portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hNGF portion is the amino acid sequence of hNGF obtained by adding a mutation to the wild-type hNGF described above. The same applies to the HSA-hNGF fusion protein having the amino acid sequence shown in SEQ ID NO: 70. The same can be said about fusion proteins of wild-type SA and wild-type hNGF from animal species other than humans.
 配列番号70で示されるアミノ酸配列を有するHSA-hNGF融合蛋白質に変異を加える場合について,以下例示する。配列番号70で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号70で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。HSA-hNGF融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hNGF部分にのみ加えても,又は両方の部分に加えてもよい。ヒト以外の動物種のSAとhNGFとの融合蛋白質(SA-hNGF)についても同様のことがいえる。 The following is an example of a case where a mutation is made to an HSA-hNGF fusion protein having the amino acid sequence shown in SEQ ID NO:70. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:70 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:70 or to the N-terminus or C-terminus of the amino acid sequence. The HSA-hNGF fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hNGF portion, or to both portions. The same applies to a fusion protein of SA and hNGF of an animal species other than human (SA-hNGF).
 配列番号70で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号70で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:70 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:70.
 HSA-hNGF融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもHSA-hNGF融合蛋白質である。また,HSA-hNGF融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもHSA-hNGF融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもHSA-hNGF融合蛋白質である。また,HSA-hNGF融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSA-hNGF融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。変異は,HSA部分にのみ加えても,hNGF部分にのみ加えても,又は両方の部分に加えてもよい。ヒト以外の動物種のSAとhNGFとの融合蛋白質(SA-hNGF)についても同様のことがいえる。  HSA-hNGF fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hNGF fusion proteins. HSA-hNGF fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hNGF fusion proteins. HSA-hNGF fusion proteins modified with anything other than sugar chains and phosphate are also HSA-hNGF fusion proteins. HSA-hNGF fusion proteins in which the side chains of the amino acids constituting the protein are modified by substitution reactions, etc., are also HSA-hNGF fusion proteins. Such modifications include, but are not limited to, the conversion of cysteine residues to formylglycine. Mutations may be made only to the HSA portion, only to the hNGF portion, or to both portions. The same can be said about fusion proteins of SA and hNGF of animal species other than humans (SA-hNGF).
 つまり,糖鎖により修飾されたHSA-hNGF融合蛋白質は,元のアミノ酸配列を有するHSA-hNGF融合蛋白質に含まれるものとする。また,リン酸により修飾されたHSA-hNGF融合蛋白質は,元のアミノ酸配列を有するHSA-hNGF融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSA-hNGF融合蛋白質に含まれるものとする。また,HSA-hNGF融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSA-hNGF融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。変異は,HSA部分にのみ加えても,hNGF部分にのみ加えても,又は両方の部分に加えてもよい。ヒト以外の動物種のSAとhNGFとの融合蛋白質(SA-hNGF)についても同様のことがいえる。  In other words, HSA-hNGF fusion proteins modified with sugar chains are included in HSA-hNGF fusion proteins having the original amino acid sequence. Also, HSA-hNGF fusion proteins modified with phosphate are included in HSA-hNGF fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are included in HSA-hNGF fusion proteins having the original amino acid sequence. Also, HSA-hNGF fusion proteins in which the side chains of the amino acids constituting the HSA-hNGF fusion protein have been changed by substitution reactions or the like are included in HSA-hNGF fusion proteins having the original amino acid sequence. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. Mutations may be made only to the HSA portion, only to the hNGF portion, or to both portions. The same can be said for fusion proteins of SA and hNGF of animal species other than humans (SA-hNGF).
 また,HSA-hNGF融合蛋白質であって,これを構成するhNGFが,hNGFの前駆体であるものもHSA-hNGF融合蛋白質である。ここで前駆体というときは,HSA-hNGF融合蛋白質として生合成されたものであって,当該生合成後に,hNGFとして機能する部分が,製造過程において又は生体に投与したときに当該生体内において,当該融合蛋白質から切り離されて,単独でもhNGFとしての機能を発揮することができるタイプのものをいう。この場合,HSA-hNGF融合蛋白質は生合成された後に,加水分解酵素等により,特定の箇所で切断され,HSAを含む部分と成熟型のhNGFを含む部分とが分離することがある。得られるhNGFはHSAとの融合蛋白質ではないが,hNGFが製造される過程で,一旦,HSA-hNGF融合蛋白質が合成されることになる。従って,かかる方法によりhNGFを製造する場合,当該製造方法は,HSA-hNGF融合蛋白質の製造方法に含まれる。変異は,HSA部分にのみ加えても,hNGF部分にのみ加えても,又は両方の部分に加えてもよい。ヒト以外の動物種のSAとhNGFとの融合蛋白質(SA-hNGF)についても同様のことがいえる。  Also, HSA-hNGF fusion proteins in which the hNGF constituting the protein is a precursor of hNGF are also HSA-hNGF fusion proteins. The term "precursor" here refers to a type of protein that is biosynthesized as an HSA-hNGF fusion protein, and after the biosynthesis, the portion that functions as hNGF is separated from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hNGF by itself. In this case, after the HSA-hNGF fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the portion containing HSA and the portion containing mature hNGF may be separated. The resulting hNGF is not a fusion protein with HSA, but the HSA-hNGF fusion protein is synthesized once during the process of manufacturing hNGF. Therefore, when hNGF is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-hNGF fusion protein. The mutation may be added only to the HSA portion, only to the hNGF portion, or both portions. The same can be said about fusion proteins of SA and hNGF from non-human animal species (SA-hNGF).
 本発明の一実施形態において,「SA-hNT-3融合蛋白質」又は「SA-hNT-3」の語は,SAのアミノ酸配列のC末端に,直接又はリンカーを介して,hNT-3のアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hNT-3としての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。SA-hNT-3融合蛋白質において,hNT-3がhNT-3としての機能を有するというときは,hNT-3が,通常の野生型のhNT-3の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでSA-hNT-3融合蛋白質におけるhNT-3の比活性は,当該融合蛋白質の単位質量当たりのhNT-3の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhNT-3に相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "SA-hNT-3 fusion protein" or "SA-hNT-3" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNT-3 is linked, directly or via a linker, to the C-terminus of the amino acid sequence of SA, and which has the function of hNT-3. Here, there is no particular limitation on the animal species of SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA. When it is said that hNT-3 in the SA-hNT-3 fusion protein has the function of hNT-3, it means that hNT-3 preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of normal wild-type hNT-3 is taken as 100%. Here, the specific activity of hNT-3 in the SA-hNT-3 fusion protein is calculated by multiplying the physiological activity of hNT-3 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hNT-3).
 本発明の一実施形態において,「HSA-hNT-3融合蛋白質」又は「HSA-hNT-3」の語は,HSAのアミノ酸配列のC末端に,直接又はリンカーを介して,hNT-3のアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hNT-3としての機能を有するものを示す。ここでHSA-hNT-3融合蛋白質がhNT-3としての機能を有するというときは,上記のSA-hNT-3融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "HSA-hNT-3 fusion protein" or "HSA-hNT-3" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNT-3 is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hNT-3. When it is said here that the HSA-hNT-3 fusion protein has the function of hNT-3, the definition of the SA-hNT-3 fusion protein described above can be applied.
 本発明の一実施形態において好ましいHSA-hNT-3融合蛋白質は,例えば配列番号72で示されるアミノ酸配列を有するものである。配列番号72で示されるHSA-hNT-3融合蛋白質は,野生型HSAのC末端にリンカー配列Gly-Serを介して野生型hNT-3が結合したものである。配列番号72で示されるHSA-hNT-3融合蛋白質は,例えば,配列番号73で示される塩基配列を有する遺伝子にコードされる。また,配列番号72で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hNT-3としての機能を有するものである限りHSA-hNT-3融合蛋白質に含まれる。HSA-hNT-3融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred HSA-hNT-3 fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 72. The HSA-hNT-3 fusion protein shown in SEQ ID NO: 72 is obtained by linking wild-type hNT-3 to the C-terminus of wild-type HSA via the linker sequence Gly-Ser. The HSA-hNT-3 fusion protein shown in SEQ ID NO: 72 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 73. In addition, HSA-hNT-3 fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 72 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hNT-3. It is preferable that the HSA-hNT-3 fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 野生型HSAと野生型hNT-3との融合蛋白質であるHSA-hNT-3融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhNT-3部分には変異を加えないことも,HSA部分には変異を加えずにhNT-3部分にのみ変異を加えることも,また,HSA部分とhNT-3部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hNT-3部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hNT-3に変異が加えられたhNT-3のアミノ酸配列となる。HSA部分とhNT-3部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hNT-3部分のアミノ酸配列は上述した野生型hNT-3に変異が加えられたhNT-3のアミノ酸配列となる。配列番号72で示されるアミノ酸配列を有するHSA-hNT-3融合蛋白質についても同様である。ヒト以外の動物種の野生型SAと野生型hNT-3との融合蛋白質についても同様のことがいえる。 When mutations are introduced into the HSA-hNT-3 fusion protein, which is a fusion protein of wild-type HSA and wild-type hNT-3, mutations can be introduced only in the HSA portion and not in the hNT-3 portion, mutations can be introduced only in the hNT-3 portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNT-3 portion. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are introduced only in the hNT-3 portion, the amino acid sequence of the portion is the amino acid sequence of hNT-3 obtained by adding a mutation to the wild-type hNT-3 described above. When mutations are introduced in both the HSA portion and the hNT-3 portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hNT-3 portion is the amino acid sequence of hNT-3 obtained by adding a mutation to the wild-type hNT-3 described above. The same is true for the HSA-hNT-3 fusion protein having the amino acid sequence shown in SEQ ID NO: 72. The same is true for a fusion protein of wild-type SA and wild-type hNT-3 of an animal species other than human.
 配列番号72で示されるアミノ酸配列を有するHSA-hNT-3融合蛋白質に変異を加える場合について,以下例示する。配列番号72で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号72で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。HSA-hNT-3融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hGALC部分にのみ加えても,又は両方の部分に加えてもよい。 The following is an example of a case where a mutation is made to an HSA-hNT-3 fusion protein having the amino acid sequence shown in SEQ ID NO:72. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:72 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:72 or to the N-terminus or C-terminus of the amino acid sequence. The HSA-hNT-3 fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hGALC portion, or to both portions.
 配列番号72で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号72で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:72 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:72.
 HSA-hNT-3融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもHSA-hNT-3融合蛋白質である。また,HSA-hNT-3融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもHSA-hNT-3融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもHSA-hNT-3融合蛋白質である。また,HSA-hNT-3融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSA-hNT-3融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhNT-3との融合蛋白質(SA-hNT-3)についても同様のことがいえる。  HSA-hNT-3 fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hNT-3 fusion proteins. HSA-hNT-3 fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hNT-3 fusion proteins. HSA-hNT-3 fusion proteins modified with something other than sugar chains and phosphate are also HSA-hNT-3 fusion proteins. HSA-hNT-3 fusion proteins in which the side chains of the amino acids constituting the protein have been changed by a substitution reaction or the like are also HSA-hNT-3 fusion proteins. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of SA and hNT-3 of animal species other than humans (SA-hNT-3).
 つまり,糖鎖により修飾されたHSA-hNT-3融合蛋白質は,元のアミノ酸配列を有するHSA-hNT-3融合蛋白質に含まれるものとする。また,リン酸により修飾されたHSA-hNT-3融合蛋白質は,元のアミノ酸配列を有するHSA-hNT-3融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSA-hNT-3融合蛋白質に含まれるものとする。また,HSA-hNT-3融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSA-hNT-3融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhNT-3との融合蛋白質(SA-hNT-3)についても同様のことがいえる。 In other words, HSA-hNT-3 fusion proteins modified with sugar chains are included in HSA-hNT-3 fusion proteins having the original amino acid sequence. Also, HSA-hNT-3 fusion proteins modified with phosphate are included in HSA-hNT-3 fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are also included in HSA-hNT-3 fusion proteins having the original amino acid sequence. Also, HSA-hNT-3 fusion proteins in which the side chains of the amino acids constituting the HSA-hNT-3 fusion proteins have been converted by substitution reactions or the like are also included in HSA-hNT-3 fusion proteins having the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hNT-3 of animal species other than humans (SA-hNT-3).
 また,HSA-hNT-3融合蛋白質であって,これを構成するhNT-3が,hNT-3の前駆体であるものもHSA-hNT-3融合蛋白質である。ここで前駆体というときは,HSA-hNT-3融合蛋白質として生合成されたものであって,当該生合成後に,hNT-3として機能する部分が,製造過程において又は生体に投与したときに当該生体内において,当該融合蛋白質から切り離されて,単独でもhNT-3としての機能を発揮することができるタイプのものをいう。この場合,HSA-hNT-3融合蛋白質は生合成された後に,加水分解酵素等により,特定の箇所で切断され,HSAを含む部分と成熟型のhNT-3を含む部分とが分離することがある。得られるhNT-3はHSAとの融合蛋白質ではないが,当該hNT-3が合成される過程で,一旦,HSA-hNT-3融合蛋白質が合成されることになる。従って,かかる方法によりhNT-3を製造する場合,当該製造方法は,HSA-hNT-3融合蛋白質の製造方法に含まれる。ヒト以外の動物種のSAとhNT-3との融合蛋白質(SA-hNT-3)についても同様のことがいえる。  Also, HSA-hNT-3 fusion proteins in which the hNT-3 constituting the protein is a precursor of hNT-3 are also HSA-hNT-3 fusion proteins. The term "precursor" here refers to a type of protein that is biosynthesized as an HSA-hNT-3 fusion protein, and in which the portion that functions as hNT-3 after the biosynthesis is cleaved from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hNT-3 by itself. In this case, after the HSA-hNT-3 fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the portion containing HSA and the portion containing mature hNT-3 may be separated. The resulting hNT-3 is not a fusion protein with HSA, but the HSA-hNT-3 fusion protein is synthesized once during the process of synthesizing the hNT-3. Therefore, when hNT-3 is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-hNT-3 fusion protein. The same can be said about the fusion protein of SA and hNT-3 from non-human animal species (SA-hNT-3).
 本発明の一実施形態において,「SA-hNT-4融合蛋白質」又は「SA-hNT-4」の語は,SAのアミノ酸配列のC末端に,直接又はリンカーを介して,hNT-4のアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hNT-4としての機能を有するものを示す。ここで,SAの動物種に特に限定はないが,好ましくは哺乳動物のSAであり,より好ましくは霊長類のSAであり,更に好ましくはHSAである。SA-hNT-4融合蛋白質において,hNT-4がhNT-4としての機能を有するというときは,hNT-4が,通常の野生型のhNT-4の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでSA-hNT-4融合蛋白質におけるhNT-4の比活性は,当該融合蛋白質の単位質量当たりのhNT-4の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhNT-4に相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "SA-hNT-4 fusion protein" or "SA-hNT-4" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNT-4 is linked, directly or via a linker, to the C-terminus of the amino acid sequence of SA, and which has the function of hNT-4. Here, there is no particular limitation on the animal species of SA, but it is preferably a mammalian SA, more preferably a primate SA, and even more preferably HSA. When it is said that hNT-4 in the SA-hNT-4 fusion protein has the function of hNT-4, it means that hNT-4 preferably retains a specific activity of 10% or more, more preferably a specific activity of 20% or more, even more preferably a specific activity of 50% or more, and even more preferably a specific activity of 80% or more, when the specific activity of normal wild-type hNT-4 is taken as 100%. Here, the specific activity of hNT-4 in the SA-hNT-4 fusion protein is calculated by multiplying the physiological activity of hNT-4 per unit mass of the fusion protein by (molecular weight of the fusion protein/molecular weight of the portion of the fusion protein corresponding to hNT-4).
 本発明の一実施形態において,「HSA-hNT-4融合蛋白質」又は「HSA-hNT-4」の語は,HSAのアミノ酸配列のC末端に,直接又はリンカーを介して,hNT-4のアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hNT-4としての機能を有するものを示す。ここでHSA-hNT-4融合蛋白質がhNT-4としての機能を有するというときは,上記のSA-hNT-4融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "HSA-hNT-4 fusion protein" or "HSA-hNT-4" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of hNT-4 is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of HSA, and which has the function of hNT-4. When it is said here that the HSA-hNT-4 fusion protein has the function of hNT-4, the definition of the SA-hNT-4 fusion protein above can be applied.
 本発明の一実施形態において好ましいHSA-hNT-4融合蛋白質は,例えば配列番号74で示されるアミノ酸配列を有するものである。配列番号74で示されるHSA-hNT-4融合蛋白質は,野生型HSAのC末端にリンカー配列Gly-Serを介して野生型hNT-4が結合したものである。配列番号74で示されるHSA-hNT-4融合蛋白質は,例えば,配列番号75で示される塩基配列を有する遺伝子にコードされる。また,配列番号74で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hNT-4としての機能を有するものである限りHSA-hNT-4融合蛋白質に含まれる。HSA-hNT-4融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred HSA-hNT-4 fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 74. The HSA-hNT-4 fusion protein shown in SEQ ID NO: 74 is obtained by linking wild-type hNT-4 to the C-terminus of wild-type HSA via the linker sequence Gly-Ser. The HSA-hNT-4 fusion protein shown in SEQ ID NO: 74 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 75. In addition, HSA-hNT-4 fusion proteins also include those having an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 74 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as they have the function of hNT-4. It is preferable that the HSA-hNT-4 fusion protein has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 野生型HSAと野生型hNT-4との融合蛋白質であるHSA-hNT-4融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhNT-4部分には変異を加えないことも,HSA部分には変異を加えずにhNT-4部分にのみ変異を加えることも,また,HSA部分とhNT-4部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hNT-4部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hNT-4に変異が加えられたhNT-4のアミノ酸配列となる。HSA部分とhNT-4部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hNT-4部分のアミノ酸配列は上述した野生型hNT-4に変異が加えられたhNT-4のアミノ酸配列となる。配列番号74で示されるアミノ酸配列を有するHSA-hNT-4融合蛋白質についても同様である。ヒト以外の動物種の野生型SAと野生型hNT-4との融合蛋白質についても同様のことがいえる。 When mutations are introduced into the HSA-hNT-4 fusion protein, which is a fusion protein of wild-type HSA and wild-type hNT-4, mutations can be introduced only in the HSA portion and not in the hNT-4 portion, mutations can be introduced only in the hNT-4 portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNT-4 portion. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are introduced only in the hNT-4 portion, the amino acid sequence of the portion is the amino acid sequence of hNT-4 obtained by adding a mutation to the wild-type hNT-4 described above. When mutations are introduced in both the HSA portion and the hNT-4 portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hNT-4 portion is the amino acid sequence of hNT-4 obtained by adding a mutation to the wild-type hNT-4 described above. The same is true for the HSA-hNT-4 fusion protein having the amino acid sequence shown in SEQ ID NO: 74. The same is true for the fusion protein of wild-type SA and wild-type hNT-4 of an animal species other than human.
 配列番号74で示されるアミノ酸配列を有するHSA-hNT-4融合蛋白質に変異を加える場合について,以下例示する。配列番号74で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号74で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。HSA-hNT-4融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hNT-4部分にのみ加えても,又は両方の部分に加えてもよい。ヒト以外の動物種のSAとhNT-3との融合蛋白質(SA-hNT-4)についても同様のことがいえる。 The following is an example of a case where a mutation is made to an HSA-hNT-4 fusion protein having the amino acid sequence shown in SEQ ID NO:74. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:74 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:74 or to the N-terminus or C-terminus of the amino acid sequence. The HSA-hNT-4 fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hNT-4 portion, or to both portions. The same can be said about the fusion protein (SA-hNT-4) of SA and hNT-3 from non-human animal species.
 配列番号74で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号74で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:74 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably shows 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:74.
 HSA-hNT-4融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもHSA-hNT-4融合蛋白質である。また,HSA-hNT-4融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもHSA-hNT-4融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもHSA-hNT-4融合蛋白質である。また,HSA-hNT-4融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもHSA-hNT-4融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhNT-3との融合蛋白質(SA-hNT-4)についても同様のことがいえる。  HSA-hNT-4 fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also HSA-hNT-4 fusion proteins. HSA-hNT-4 fusion proteins in which the amino acids constituting the protein are modified with phosphate are also HSA-hNT-4 fusion proteins. HSA-hNT-4 fusion proteins modified with something other than sugar chains and phosphate are also HSA-hNT-4 fusion proteins. HSA-hNT-4 fusion proteins in which the side chains of the amino acids constituting the protein have been changed by a substitution reaction or the like are also HSA-hNT-4 fusion proteins. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins (SA-hNT-4) of SA and hNT-3 of animal species other than humans.
 つまり,糖鎖により修飾されたHSA-hNT-4融合蛋白質は,元のアミノ酸配列を有するHSA-hNT-4融合蛋白質に含まれるものとする。また,リン酸により修飾されたHSA-hNT-4融合蛋白質は,元のアミノ酸配列を有するHSA-hNT-4融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するHSA-hNT-4融合蛋白質に含まれるものとする。また,HSA-hNT-4融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するHSA-hNT-4融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のSAとhNT-3との融合蛋白質(SA-hNT-4)についても同様のことがいえる。 In other words, HSA-hNT-4 fusion proteins modified with sugar chains are included in HSA-hNT-4 fusion proteins having the original amino acid sequence. Also, HSA-hNT-4 fusion proteins modified with phosphate are included in HSA-hNT-4 fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are also included in HSA-hNT-4 fusion proteins having the original amino acid sequence. Also, HSA-hNT-4 fusion proteins in which the side chains of the amino acids constituting the HSA-hNT-4 fusion protein have been converted by substitution reactions or the like are also included in HSA-hNT-4 fusion proteins having the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of SA and hNT-3 of animal species other than humans (SA-hNT-4).
 また,HSA-hNT-4融合蛋白質であって,これを構成するhNT-4が,hNT-4の前駆体であるものもHSA-hNT-4融合蛋白質である。ここで前駆体というときは,HSA-hNT-4融合蛋白質として生合成されたものであって,当該生合成後に,hNT-4として機能する部分が,製造過程において又は生体に投与したときに当該生体内において,当該融合蛋白質から切り離されて,単独でもhNT-4としての機能を発揮することができるタイプのものをいう。この場合,HSA-hNT-4融合蛋白質は生合成された後に,加水分解酵素等により,特定の箇所で切断され,HSAを含む部分とhNT-4を含む部分とが分離することがある。得られるhNT-4はHSAとの融合蛋白質ではないが,当該hNT-4が合成される過程で,一旦,HSA-hNT-4融合蛋白質が合成されることになる。従って,かかる方法によりhNT-4を製造する場合,当該製造方法は,HSA-hNT-4融合蛋白質の製造方法に含まれる。ヒト以外の動物種のSAとhNT-4との融合蛋白質(SA-hNT-4)についても同様のことがいえる。  Also, HSA-hNT-4 fusion proteins in which the constituent hNT-4 is a precursor of hNT-4 are also HSA-hNT-4 fusion proteins. Here, the term "precursor" refers to a type of protein that is biosynthesized as an HSA-hNT-4 fusion protein, and in which the portion that functions as hNT-4 after the biosynthesis is cleaved from the fusion protein during the manufacturing process or in the living body when administered to the living body, and can function as hNT-4 by itself. In this case, after the HSA-hNT-4 fusion protein is biosynthesized, it may be cleaved at a specific site by a hydrolase or the like, and the portion containing HSA and the portion containing hNT-4 may be separated. The resulting hNT-4 is not a fusion protein with HSA, but the HSA-hNT-4 fusion protein is synthesized once during the process of synthesizing the hNT-4. Therefore, when hNT-4 is manufactured by such a method, the manufacturing method is included in the manufacturing method of HSA-hNT-4 fusion protein. The same can be said about the fusion protein of SA and hNT-4 from non-human animal species (SA-hNT-4).
 本発明の一実施形態において,「ヒト神経栄養因子-SA融合蛋白質」又は「ヒト神経栄養因子-SA」の語は,ヒト神経栄養因子のアミノ酸配列のC末端に,直接又はリンカーを介して,SAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,ヒト神経栄養因子としての機能を有するものを示す。ヒト神経栄養因子-SA融合蛋白質において,ヒト神経栄養因子がヒト神経栄養因子としての機能を有するというときは,ヒト神経栄養因子が,通常の野生型のヒト神経栄養因子の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでヒト神経栄養因子-SA融合蛋白質におけるヒト神経栄養因子の比活性は,当該融合蛋白質の単位質量当たりのヒト神経栄養因子の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のヒト神経栄養因子に相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "human neurotrophic factor-SA fusion protein" or "human neurotrophic factor-SA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked directly or via a linker to the C-terminus of the amino acid sequence of human neurotrophic factor, and which has the function of human neurotrophic factor. When the human neurotrophic factor in the human neurotrophic factor-SA fusion protein is said to have the function of human neurotrophic factor, it means that the human neurotrophic factor retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of a normal wild-type human neurotrophic factor is taken as 100%. Here, the specific activity of human neurotrophic factor in the human neurotrophic factor-SA fusion protein is calculated by multiplying the physiological activity of human neurotrophic factor per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to human neurotrophic factor).
 本発明の一実施形態において,「ヒト神経栄養因子-HSA融合蛋白質」又は「ヒト神経栄養因子-HSA」の語は,ヒト神経栄養因子のアミノ酸配列のC末端に,直接又はリンカーを介して,HSAのアミノ酸配列のN末端が結合されている,アミノ酸配列を有する融合蛋白質であって,ヒト神経栄養因子としての機能を有するものを示す。ここでヒト神経栄養因子-HSA融合蛋白質がヒト神経栄養因子としての機能を有するというときは,上記の神経栄養因子-HSA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "human neurotrophic factor-HSA fusion protein" or "human neurotrophic factor-HSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of human neurotrophic factor, and which has the function of a human neurotrophic factor. When it is said that the human neurotrophic factor-HSA fusion protein has the function of a human neurotrophic factor, the definition of the neurotrophic factor-HSA fusion protein described above can be applied.
 ヒト神経栄養因子-SA融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。ヒト神経栄養因子-HSA融合蛋白質においても同様である。hBDNF-HSA融合蛋白質,hNGF-HSA融合蛋白質,hNT-3-HSA融合蛋白質,hNT-4-HSA融合蛋白質についても同様である。 In human neurotrophic factor-SA fusion protein, it is preferable that the SA has a function as an SA, such as the function of binding and transporting endogenous substances in the blood and exogenous substances such as drugs, but is not limited to this. The same is true for human neurotrophic factor-HSA fusion protein. The same is true for hBDNF-HSA fusion protein, hNGF-HSA fusion protein, hNT-3-HSA fusion protein, and hNT-4-HSA fusion protein.
 本発明の一実施形態において,ヒト以外の動物種の神経栄養因子のアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のSAのN末端が結合されているアミノ酸配列を有する融合蛋白質であって,神経栄養因子としての機能を有するものは,「(ヒト以外の動物種)神経栄養因子-(ヒト以外の動物種)SA融合蛋白質」と表記される。例えば,マウス神経栄養因子とマウスSAの融合蛋白質は,「マウス神経栄養因子-MSA融合蛋白質」又は「マウス神経栄養因子-MSA」と表記される。これら融合蛋白質についても,神経栄養因子としての機能を有するというときは,上記のヒト神経栄養因子-SA融合蛋白質における定義を適用することができる。また,これら融合蛋白質において,SAは血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のSAとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of a neurotrophic factor of a non-human animal species, and which has a function as a neurotrophic factor, is referred to as a "(non-human animal species) neurotrophic factor-(non-human animal species) SA fusion protein." For example, a fusion protein of mouse neurotrophic factor and mouse SA is referred to as a "mouse neurotrophic factor-MSA fusion protein" or "mouse neurotrophic factor-MSA." When these fusion proteins are said to have a function as a neurotrophic factor, the definition of the human neurotrophic factor-SA fusion protein described above can be applied. In addition, in these fusion proteins, it is preferable that the SA has a function as an SA, such as a function to bind and transport endogenous substances in the blood and exogenous substances such as drugs, but this is not limited thereto.
 野生型ヒト神経栄養因子と野生型HSAとの融合蛋白質であるヒト神経栄養因子-HSA融合蛋白質に変異を加える場合,ヒト神経栄養因子部分にのみ変異を加えHSA部分には変異を加えないことも,ヒト神経栄養因子部分には変異を加えずにHSA部分にのみ変異を加えることも,また,ヒト神経栄養因子部分とHSA部分の何れにも変異を加えることもできる。ヒト神経栄養因子部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型ヒト神経栄養因子に変異が加えられたヒト神経栄養因子のアミノ酸配列となる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。ヒト神経栄養因子部分とHSA部分の何れにも変異を加える場合にあっては,ヒト神経栄養因子部分のアミノ酸配列は上述した野生型ヒト神経栄養因子に変異が加えられたヒト神経栄養因子のアミノ酸配列となり,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。野生型ヒト神経栄養因子とヒト以外の動物種の野生型SAとの融合蛋白質についても同様のことがいえる。 When a mutation is added to a human neurotrophic factor-HSA fusion protein, which is a fusion protein of wild-type human neurotrophic factor and wild-type HSA, a mutation can be added only to the human neurotrophic factor portion and not to the HSA portion, a mutation can be added only to the HSA portion without adding a mutation to the human neurotrophic factor portion, or a mutation can be added to both the human neurotrophic factor portion and the HSA portion. When a mutation is added only to the human neurotrophic factor portion, the amino acid sequence of that portion is the amino acid sequence of the human neurotrophic factor obtained by adding a mutation to the wild-type human neurotrophic factor described above. When a mutation is added only to the HSA portion, the amino acid sequence of that portion is the amino acid sequence of the HSA obtained by adding a mutation to the wild-type HSA described above. When a mutation is added to both the human neurotrophic factor portion and the HSA portion, the amino acid sequence of the human neurotrophic factor portion is the amino acid sequence of the human neurotrophic factor obtained by adding a mutation to the wild-type human neurotrophic factor described above, and the amino acid sequence of the HSA portion is the amino acid sequence of the HSA obtained by adding a mutation to the wild-type HSA described above. The same can be said about fusion proteins between wild-type human neurotrophic factor and wild-type SA of non-human animal species.
 ヒト神経栄養因子-HSA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもヒト神経栄養因子-HSA融合蛋白質である。また,ヒト神経栄養因子-HSA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもヒト神経栄養因子-HSA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもヒト神経栄養因子-HSA融合蛋白質である。また,ヒト神経栄養因子-HSA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもヒト神経栄養因子-HSA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト神経栄養因子とヒト以外の動物種のSAとの融合蛋白質(ヒト神経栄養因子-SA)についても同様のことがいえる。 Human neurotrophic factor-HSA fusion proteins in which the amino acids constituting the protein are modified with sugar chains are also human neurotrophic factor-HSA fusion proteins. Human neurotrophic factor-HSA fusion proteins in which the amino acids constituting the protein are modified with phosphate are also human neurotrophic factor-HSA fusion proteins. Human neurotrophic factor-HSA fusion proteins modified with something other than sugar chains and phosphate are also human neurotrophic factor-HSA fusion proteins. Human neurotrophic factor-HSA fusion proteins in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like are also human neurotrophic factor-HSA fusion proteins. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of human neurotrophic factor and SA of animal species other than human (human neurotrophic factor-SA).
 つまり,糖鎖により修飾されたヒト神経栄養因子-HSA融合蛋白質は,元のアミノ酸配列を有するヒト神経栄養因子-HSA融合蛋白質に含まれるものとする。また,リン酸により修飾されたヒト神経栄養因子-HSA融合蛋白質は,元のアミノ酸配列を有するヒト神経栄養因子-HSA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するヒト神経栄養因子-HSA融合蛋白質に含まれるものとする。また,ヒト神経栄養因子-HSA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するヒト神経栄養因子-HSA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト神経栄養因子とヒト以外の動物種のSAとの融合蛋白質(ヒト神経栄養因子-SA)についても同様のことがいえる。 In other words, a human neurotrophic factor-HSA fusion protein modified by a sugar chain is included in the human neurotrophic factor-HSA fusion protein having the original amino acid sequence. A human neurotrophic factor-HSA fusion protein modified by phosphate is included in the human neurotrophic factor-HSA fusion protein having the original amino acid sequence. A human neurotrophic factor-HSA fusion protein modified by something other than a sugar chain or phosphate is also included in the human neurotrophic factor-HSA fusion protein having the original amino acid sequence. A human neurotrophic factor-HSA fusion protein in which the side chains of the amino acids constituting the human neurotrophic factor-HSA fusion protein have been converted by a substitution reaction or the like is also included in the human neurotrophic factor-HSA fusion protein having the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about a fusion protein of human neurotrophic factor and SA of an animal species other than human (human neurotrophic factor-SA).
 また,ヒト神経栄養因子-HSA融合蛋白質であって,これを構成するヒト神経栄養因子が,ヒト神経栄養因子の前駆体であるものもヒト神経栄養因子-HSA融合蛋白質である。但し,ヒト神経栄養因子の前駆体が,ヒト神経栄養因子としての機能を示さない場合には,これがインビボ又はインビトロでプロセシングされてヒト神経栄養因子としての機能を示すものに転換されるものであることが必要である。  In addition, a human neurotrophic factor-HSA fusion protein in which the human neurotrophic factor that constitutes it is a precursor of human neurotrophic factor is also a human neurotrophic factor-HSA fusion protein. However, if the precursor of human neurotrophic factor does not exhibit the function of human neurotrophic factor, it is necessary that it is processed in vivo or in vitro and converted into one that exhibits the function of human neurotrophic factor.
 本発明の一実施形態において,「hBDNF-SA融合蛋白質」又は「hBDNF-SA」の語は,hBDNFのアミノ酸配列のC末端に,直接又はリンカーを介して,SAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hBDNFとしての機能を有するものを示す。hBDNF-SA融合蛋白質において,hBDNFがhBDNFとしての機能を有するというときは,hBDNFが,通常の野生型のhBDNFの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでhBDNF-SA融合蛋白質におけるhBDNFの比活性は,当該融合蛋白質の単位質量当たりのhBDNFの生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhBDNFに相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "hBDNF-SA fusion protein" or "hBDNF-SA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hBDNF, and which has the function of hBDNF. When hBDNF in an hBDNF-SA fusion protein is said to have the function of hBDNF, it means that hBDNF retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hBDNF is taken as 100%. Here, the specific activity of hBDNF in an hBDNF-SA fusion protein is calculated by multiplying the physiological activity of hBDNF per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hBDNF).
 本発明の一実施形態において,「hBDNF-HSA融合蛋白質」又は「hBDNF-HSA」の語は,hBDNFのアミノ酸配列のC末端に,直接又はリンカーを介して,HSAのアミノ酸配列のN末端が結合されている,アミノ酸配列を有する融合蛋白質であって,hBDNFとしての機能を有するものを示す。ここでhBDNF-HSA融合蛋白質がhBDNFとしての機能を有するというときは,上記のhBDNF-SA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "hBDNF-HSA fusion protein" or "hBDNF-HSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hBDNF, and which has the function of hBDNF. When it is said here that the hBDNF-HSA fusion protein has the function of hBDNF, the definition of the hBDNF-SA fusion protein described above can be applied.
 本発明において,ヒト以外の動物種のBDNFのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のSAのN末端が結合されているアミノ酸配列を有する融合蛋白質であって,BDNFとしての機能を有するものは,「(ヒト以外の動物種)BDNF-(ヒト以外の動物種)SA融合蛋白質」と表記される。例えば,マウスBDNFとマウスSAの融合蛋白質は,「マウスBDNF-マウスSA融合蛋白質」又は「マウスBDNF-MSA」と表記される。これら融合蛋白質についても,BDNFとしての機能を有するというときは,上記のhBDNF-SA融合蛋白質における定義を適用することができる。 In the present invention, a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of BDNF of a non-human animal species, and which has the function of BDNF, is referred to as a "(non-human animal species) BDNF-(non-human animal species) SA fusion protein." For example, a fusion protein of mouse BDNF and mouse SA is referred to as a "mouse BDNF-mouse SA fusion protein" or "mouse BDNF-MSA." When these fusion proteins are said to have the function of BDNF, the definition of hBDNF-SA fusion protein described above can be applied.
 本発明の一実施形態において好ましいhBDNF-HSA融合蛋白質は,例えば配列番号76で示されるアミノ酸配列を有するものである。配列番号76で示されるhBDNF-HSA融合蛋白質は,野生型hBDNFのC末端にリンカー配列Gly-Serを介して野生型HSAが結合したものである。配列番号76で示されるhBDNF-HSA融合蛋白質は,例えば,配列番号77で示される塩基配列を有する遺伝子にコードされる。また,配列番号76で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hBDNFとしての機能を有するものである限りhBDNF-HSA融合蛋白質に含まれる。hBDNF-HSA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred hBDNF-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 76. The hBDNF-HSA fusion protein shown in SEQ ID NO: 76 is obtained by linking wild-type HSA to the C-terminus of wild-type hBDNF via the linker sequence Gly-Ser. The hBDNF-HSA fusion protein shown in SEQ ID NO: 76 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 77. In addition, the hBDNF-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 76 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as it has the function of hBDNF. The hBDNF-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 配列番号76で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号76で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:76 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:76.
 野生型hBDNFと野生型HSAとの融合蛋白質であるhBDNF-HSA融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhBDNF部分には変異を加えないことも,HSA部分には変異を加えずにhBDNF部分にのみ変異を加えることも,また,HSA部分とhBDNF部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hBDNF部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hBDNFに変異が加えられたhBDNFのアミノ酸配列となる。HSA部分とhBDNF部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hBDNF部分のアミノ酸配列は上述した野生型hBDNFに変異が加えられたhBDNFのアミノ酸配列となる。配列番号76で示されるアミノ酸配列を有するhBDNF-HSA融合蛋白質についても同様である。野生型hBDNFとヒト以外の動物種の野生型SA(hBDNF-SA)との融合蛋白質についても同様のことがいえる。 When mutations are added to an hBDNF-HSA fusion protein, which is a fusion protein of wild-type hBDNF and wild-type HSA, mutations can be added only to the HSA portion and not to the hBDNF portion, mutations can be added only to the hBDNF portion without mutations to the HSA portion, or mutations can be added to both the HSA portion and the hBDNF portion. When mutations are added only to the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are added only to the hBDNF portion, the amino acid sequence of the portion is the amino acid sequence of hBDNF obtained by adding a mutation to the wild-type hBDNF described above. When mutations are added to both the HSA portion and the hBDNF portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hBDNF portion is the amino acid sequence of hBDNF obtained by adding a mutation to the wild-type hBDNF described above. The same applies to the hBDNF-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 76. The same can be said about fusion proteins of wild-type hBDNF and wild-type SA of non-human animal species (hBDNF-SA).
 配列番号76で示されるアミノ酸配列を有するhBDNF-HSA融合蛋白質に変異を加える場合について,以下例示する。配列番号76で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号76で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。hBDNF-HSA融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hBDNF部分にのみ加えても,又は両方の部分に加えてもよい。hBDNFとヒト以外の動物種のSAとの融合蛋白質(hBDNF-SA)についても同様のことがいえる。 The following is an example of a case where a mutation is made to an hBDNF-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:76. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:76 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:76 or to the N-terminus or C-terminus of the amino acid sequence. The hBDNF-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hBDNF portion, or to both portions. The same applies to a fusion protein of hBDNF and SA of an animal species other than human (hBDNF-SA).
 hBDNF-HSA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもhBDNF-HSA融合蛋白質である。また,hBDNF-HSA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもhBDNF-HSA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもhBDNF-HSA融合蛋白質である。また,hBDNF-HSA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもhBDNF-HSA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。hBDNFとヒト以外の動物種のSAとの融合蛋白質(hBDNF-SA)についても同様のことがいえる。  An hBDNF-HSA fusion protein in which the constituent amino acids are modified with sugar chains is also an hBDNF-HSA fusion protein. An hBDNF-HSA fusion protein in which the constituent amino acids are modified with phosphate is also an hBDNF-HSA fusion protein. An hBDNF-HSA fusion protein modified with something other than sugar chains and phosphate is also an hBDNF-HSA fusion protein. An hBDNF-HSA fusion protein in which the side chains of the constituent amino acids have been converted by a substitution reaction or the like is also an hBDNF-HSA fusion protein. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about a fusion protein of hBDNF and SA of an animal species other than human (hBDNF-SA).
 つまり,糖鎖により修飾されたhBDNF-HSA融合蛋白質は,元のアミノ酸配列を有するhBDNF-HSA融合蛋白質に含まれるものとする。また,リン酸により修飾されたhBDNF-HSA融合蛋白質は,元のアミノ酸配列を有するhBDNF-HSA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するhBDNF-HSA融合蛋白質に含まれるものとする。また,hBDNF-HSA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するhBDNF-HSA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。hBDNFとヒト以外の動物種のSAとの融合蛋白質(hBDNF-SA)についても同様のことがいえる。 In other words, hBDNF-HSA fusion proteins modified with sugar chains are included in hBDNF-HSA fusion proteins having the original amino acid sequence. Also, hBDNF-HSA fusion proteins modified with phosphate are included in hBDNF-HSA fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are also included in hBDNF-HSA fusion proteins having the original amino acid sequence. Also, hBDNF-HSA fusion proteins in which the side chains of the amino acids constituting the hBDNF-HSA fusion protein have been changed by substitution reactions or the like are also included in hBDNF-HSA fusion proteins having the original amino acid sequence. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of hBDNF and SA of animal species other than human (hBDNF-SA).
 また,hBDNF-HSA融合蛋白質であって,これを構成するhBDNFが,hBDNFの前駆体であるものもhBDNF-HSA融合蛋白質である。但し,hBDNFが前駆体であるhBDNF-HSA融合蛋白質が,hBDNFとしての機能を示さない場合には,これがインビボ又はインビトロでプロセシングされてhBDNFとしての機能を示すものに転換されるものであることが必要である。  An hBDNF-HSA fusion protein in which the hBDNF that constitutes it is a precursor of hBDNF is also an hBDNF-HSA fusion protein. However, if the hBDNF-HSA fusion protein in which hBDNF is a precursor does not exhibit the functions of hBDNF, it is necessary that it be processed in vivo or in vitro and converted into one that exhibits the functions of hBDNF.
 本発明の一実施形態において,「hNGF-SA融合蛋白質」又は「hNGF-SA」の語は,hNGFのアミノ酸配列のC末端に,直接又はリンカーを介して,SAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hNGFとしての機能を有するものを示す。hNGF-SA融合蛋白質において,hNGFがhNGFとしての機能を有するというときは,hNGFが,通常の野生型のhNGFの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでhNGF-SA融合蛋白質におけるhNGFの比活性は,当該融合蛋白質の単位質量当たりのhNGFの生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhNGFに相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "hNGF-SA fusion protein" or "hNGF-SA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked to the C-terminus of the amino acid sequence of hNGF, either directly or via a linker, and which has the function of hNGF. When hNGF in an hNGF-SA fusion protein is said to have the function of hNGF, it means that hNGF retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hNGF is taken as 100%. Here, the specific activity of hNGF in an hNGF-SA fusion protein is calculated by multiplying the physiological activity of hNGF per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hNGF).
 本発明の一実施形態において,「hNGF-HSA融合蛋白質」又は「hNGF-HSA」の語は,hNGFのアミノ酸配列のC末端に,直接又はリンカーを介して,HSAのアミノ酸配列のN末端が結合されている,アミノ酸配列を有する融合蛋白質であって,hNGFとしての機能を有するものを示す。ここでhNGF-HSA融合蛋白質がhNGFとしての機能を有するというときは,上記のhNGF-SA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "hNGF-HSA fusion protein" or "hNGF-HSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hNGF, and which has the function of hNGF. When it is said here that the hNGF-HSA fusion protein has the function of hNGF, the definition of the hNGF-SA fusion protein described above can be applied.
 本発明の一実施形態において,ヒト以外の動物種のNGFのアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のSAのN末端が結合されているアミノ酸配列を有する融合蛋白質であって,NGFとしての機能を有するものは,「(ヒト以外の動物種)NGF-(ヒト以外の動物種)SA融合蛋白質」と表記される。例えば,マウスNGFとマウスSAの融合蛋白質は,「マウスNGF-マウスSA融合蛋白質」又は「マウスNGF-MSA」と表記される。これら融合蛋白質についても,NGFとしての機能を有するというときは,上記のhNGF-SA融合蛋白質における定義を適用することができる。また, In one embodiment of the present invention, a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, directly or via a linker, to the C-terminus of the amino acid sequence of NGF of a non-human animal species, and which has the function of NGF, is referred to as a "(non-human animal species) NGF-(non-human animal species) SA fusion protein." For example, a fusion protein of mouse NGF and mouse SA is referred to as a "mouse NGF-mouse SA fusion protein" or "mouse NGF-MSA." When these fusion proteins are said to have the function of NGF, the definition of the hNGF-SA fusion protein described above can be applied. In addition,
 本発明の一実施形態において好ましいhNGF-HSA融合蛋白質は,例えば配列番号78で示されるアミノ酸配列を有するものである。配列番号78で示されるhNGF-HSA融合蛋白質は,野生型hNGFのC末端にリンカー配列Gly-Serを介して野生型HSAが結合したものである。配列番号78で示されるhNGF-HSA融合蛋白質は,例えば,配列番号79で示される塩基配列を有する遺伝子にコードされる。また,配列番号78で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hNGFとしての機能を有するものである限りhNGF-HSA融合蛋白質に含まれる。hNGF-HSA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred hNGF-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 78. The hNGF-HSA fusion protein shown in SEQ ID NO: 78 is obtained by linking wild-type HSA to the C-terminus of wild-type hNGF via the linker sequence Gly-Ser. The hNGF-HSA fusion protein shown in SEQ ID NO: 78 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 79. In addition, the hNGF-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 78 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as it has the function of hNGF. The hNGF-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 配列番号78で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号78で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:78 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:78.
 野生型hNGFと野生型HSAとの融合蛋白質であるhNGF-HSA融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhNGF部分には変異を加えないことも,HSA部分には変異を加えずにhNGF部分にのみ変異を加えることも,また,HSA部分とhNGF部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hNGF部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hNGFに変異が加えられたhNGFのアミノ酸配列となる。HSA部分とhNGF部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hNGF部分のアミノ酸配列は上述した野生型hNGFに変異が加えられたhNGFのアミノ酸配列となる。配列番号78で示されるアミノ酸配列を有するhNGF-HSA融合蛋白質についても同様である。野生型hNGFとヒト以外の動物種の野生型SA(hNGF-SA)との融合蛋白質についても同様のことがいえる。 When mutations are introduced into an hNGF-HSA fusion protein, which is a fusion protein of wild-type hNGF and wild-type HSA, mutations can be introduced only in the HSA portion and not in the hNGF portion, mutations can be introduced only in the hNGF portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNGF portion. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are introduced only in the hNGF portion, the amino acid sequence of the portion is the amino acid sequence of hNGF obtained by adding a mutation to the wild-type hNGF described above. When mutations are introduced in both the HSA portion and the hNGF portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hNGF portion is the amino acid sequence of hNGF obtained by adding a mutation to the wild-type hNGF described above. The same applies to the hNGF-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 78. The same can be said about fusion proteins between wild-type hNGF and wild-type SA (hNGF-SA) of non-human animal species.
 配列番号78で示されるアミノ酸配列を有するhNGF-HSA融合蛋白質に変異を加える場合について,以下例示する。配列番号78で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号78で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。hNGF-HSA融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hNGF部分にのみ加えても,又は両方の部分に加えてもよい。ヒト以外の動物種のNGFとHSAとの融合蛋白質(NGF-SA),及びヒト以外の動物種のNGFとヒト以外の動物種のSAとの融合蛋白質についても同様のことがいえる。 The following is an example of a case where a mutation is made to an hNGF-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:78. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:78 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:78 or to the N-terminus or C-terminus of the amino acid sequence. The hNGF-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hNGF portion, or to both portions. The same can be said for fusion proteins of non-human animal species' NGF and HSA (NGF-SA), and fusion proteins of non-human animal species' NGF and non-human animal species' SA.
 hNGF-HSA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもhNGF-HSA融合蛋白質である。また,hNGF-HSA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもhNGF-HSA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもhNGF-HSA融合蛋白質である。また,hNGF-HSA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもhNGF-HSA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。hNGFとヒト以外の動物種のSAとの融合蛋白質(hNGF-SA)についても同様のことがいえる。  An hNGF-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also an hNGF-HSA fusion protein. An hNGF-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also an hNGF-HSA fusion protein. An hNGF-HSA fusion protein modified with something other than sugar chains and phosphate is also an hNGF-HSA fusion protein. An hNGF-HSA fusion protein in which the side chains of the amino acids constituting the protein have been changed by a substitution reaction or the like is also an hNGF-HSA fusion protein. Such a change includes, but is not limited to, the change of a cysteine residue to formylglycine. The same can be said about a fusion protein (hNGF-SA) between hNGF and SA of an animal species other than human.
 つまり,糖鎖により修飾されたhNGF-HSA融合蛋白質は,元のアミノ酸配列を有するhNGF-HSA融合蛋白質に含まれるものとする。また,リン酸により修飾されたhNGF-HSA融合蛋白質は,元のアミノ酸配列を有するhNGF-HSA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するhNGF-HSA融合蛋白質に含まれるものとする。また,hNGF-HSA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するhNGF-HSA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。hNGFとヒト以外の動物種のSAとの融合蛋白質(hNGF-SA)についても同様のことがいえる。 In other words, hNGF-HSA fusion proteins modified with sugar chains are included in the hNGF-HSA fusion proteins having the original amino acid sequence. Also, hNGF-HSA fusion proteins modified with phosphate are included in the hNGF-HSA fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are included in the hNGF-HSA fusion proteins having the original amino acid sequence. Also, hNGF-HSA fusion proteins in which the side chains of the amino acids constituting the hNGF-HSA fusion proteins have been changed by substitution reactions or the like are included in the hNGF-HSA fusion proteins having the original amino acid sequence. Such changes include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about fusion proteins of hNGF and SA of animals other than humans (hNGF-SA).
 また,hNGF-HSA融合蛋白質であって,これを構成するhNGFが,hNGFの前駆体であるものもhNGF-HSA融合蛋白質である。但し,hNGFが前駆体であるhNGF-HSA融合蛋白質が,hNGFとしての機能を示さない場合には,これがインビボ又はインビトロでプロセシングされてhNGFとしての機能を示すものに転換されるものであることが必要である。hNGFとヒト以外の動物種のSAとの融合蛋白質(hNGF-SA)についても同様のことがいえる。  An hNGF-HSA fusion protein in which the hNGF that constitutes it is a precursor of hNGF is also an hNGF-HSA fusion protein. However, if the hNGF-HSA fusion protein, in which hNGF is a precursor, does not function as hNGF, it is necessary that it be processed in vivo or in vitro and converted into one that functions as hNGF. The same can be said for a fusion protein of hNGF and SA of an animal species other than human (hNGF-SA).
 本発明の一実施形態において,「hNT-3-SA融合蛋白質」又は「hNT-3-SA」の語は,hNT-3のアミノ酸配列のC末端に,直接又はリンカーを介して,SAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hNT-3としての機能を有するものを示す。hNT-3-SA融合蛋白質において,hNT-3がhNT-3としての機能を有するというときは,hNT-3が,通常の野生型のhNT-3の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでhNT-3-SA融合蛋白質におけるhNT-3の比活性は,当該融合蛋白質の単位質量当たりのhNT-3の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhNT-3に相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "hNT-3-SA fusion protein" or "hNT-3-SA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hNT-3, and which has the function of hNT-3. When hNT-3 in an hNT-3-SA fusion protein is said to have the function of hNT-3, it means that hNT-3 retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hNT-3 is taken as 100%. Here, the specific activity of hNT-3 in the hNT-3-SA fusion protein is calculated by multiplying the physiological activity of hNT-3 per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hNT-3).
 本発明の一実施形態において,「hNT-3-HSA融合蛋白質」又は「hNT-3-HSA」の語は,hNT-3のアミノ酸配列のC末端に,直接又はリンカーを介して,HSAのアミノ酸配列のN末端が結合されている,アミノ酸配列を有する融合蛋白質であって,hNT-3としての機能を有するものを示す。ここでhNT-3-HSA融合蛋白質がhNT-3としての機能を有するというときは,上記のhNT-3-SA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "hNT-3-HSA fusion protein" or "hNT-3-HSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hNT-3, and which has the function of hNT-3. When it is said here that the hNT-3-HSA fusion protein has the function of hNT-3, the definition of the hNT-3-SA fusion protein described above can be applied.
 本発明の一実施形態において,ヒト以外の動物種のNT-3のアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のSAのN末端が結合されているアミノ酸配列を有する融合蛋白質であって,NT-3としての機能を有するものは,「(ヒト以外の動物種)NT-3-(ヒト以外の動物種)SA融合蛋白質」と表記される。例えば,マウスNT-3とマウスSAの融合蛋白質は,「マウスNT-3-マウスSA融合蛋白質」又は「マウスNT-3-MSA」と表記される。これら融合蛋白質についても,NT-3としての機能を有するというときは,上記のhNT-3-SA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of NT-3 of a non-human animal species, and which has the function of NT-3, is referred to as a "(non-human animal species) NT-3-(non-human animal species) SA fusion protein." For example, a fusion protein of mouse NT-3 and mouse SA is referred to as a "mouse NT-3-mouse SA fusion protein" or "mouse NT-3-MSA." When these fusion proteins are said to have the function of NT-3, the definition of hNT-3-SA fusion protein described above can be applied.
 本発明の一実施形態において好ましいhNT-3-HSA融合蛋白質は,例えば配列番号80で示されるアミノ酸配列を有するものである。配列番号80で示されるhNT-3-HSA融合蛋白質は,野生型hNT-3のC末端にリンカー配列Gly-Serを介して野生型HSAが結合したものである。配列番号80で示されるhNT-3-HSA融合蛋白質は,例えば,配列番号81で示される塩基配列を有する遺伝子にコードされる。また,配列番号80で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hNT-3としての機能を有するものである限りhNT-3-HSA融合蛋白質に含まれる。hNT-3-HSA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred hNT-3-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 80. The hNT-3-HSA fusion protein shown in SEQ ID NO: 80 is obtained by linking wild-type HSA to the C-terminus of wild-type hNT-3 via the linker sequence Gly-Ser. The hNT-3-HSA fusion protein shown in SEQ ID NO: 80 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 81. In addition, the hNT-3-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 80 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as it has the function of hNT-3. The hNT-3-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 配列番号80で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号80で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:80 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably exhibits 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:80.
 野生型hNT-3と野生型HSAとの融合蛋白質であるhNT-3-HSA融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhNT-3部分には変異を加えないことも,HSA部分には変異を加えずにhNT-3部分にのみ変異を加えることも,また,HSA部分とhNT-3部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hNT-3部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hNT-3に変異が加えられたhNT-3のアミノ酸配列となる。HSA部分とhNT-3部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hNT-3部分のアミノ酸配列は上述した野生型hNT-3に変異が加えられたhNT-3のアミノ酸配列となる。配列番号80で示されるアミノ酸配列を有するhNT-3-HSA融合蛋白質についても同様である。野生型hNT-3とヒト以外の動物種の野生型SA(hNT-3-SA)との融合蛋白質についても同様のことがいえる。 When mutations are introduced into the hNT-3-HSA fusion protein, which is a fusion protein of wild-type hNT-3 and wild-type HSA, mutations can be introduced only in the HSA portion and not in the hNT-3 portion, mutations can be introduced only in the hNT-3 portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNT-3 portion. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are introduced only in the hNT-3 portion, the amino acid sequence of the portion is the amino acid sequence of hNT-3 obtained by adding a mutation to the wild-type hNT-3 described above. When mutations are introduced both in the HSA portion and the hNT-3 portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hNT-3 portion is the amino acid sequence of hNT-3 obtained by adding a mutation to the wild-type hNT-3 described above. The same is true for the hNT-3-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 80. The same is true for the fusion protein of wild-type hNT-3 and wild-type SA (hNT-3-SA) of an animal species other than human.
 配列番号80で示されるアミノ酸配列を有するhNT-3-HSA融合蛋白質に変異を加える場合について,以下例示する。配列番号80で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号80で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。hNT-3-HSA融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hNT-3部分にのみ加えても,又は両方の部分に加えてもよい。hNT-3とヒト以外の動物種のSAとの融合蛋白質(hNT-3-SA)についても同様のことがいえる。 The following is an example of a case where a mutation is made to an hNT-3-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:80. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:80 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:80 or to the N-terminus or C-terminus of the amino acid sequence. The hNT-3-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hNT-3 portion, or to both portions. The same can be said about the fusion protein (hNT-3-SA) between hNT-3 and SA of a non-human animal species.
 hNT-3-HSA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもhNT-3-HSA融合蛋白質である。また,hNT-3-HSA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもhNT-3-HSA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもhNT-3-HSA融合蛋白質である。また,hNT-3-HSA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもhNT-3-HSA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。hNT-3とヒト以外の動物種のSAとの融合蛋白質(hNT-3-SA)についても同様のことがいえる。  An hNT-3-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also an hNT-3-HSA fusion protein. An hNT-3-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also an hNT-3-HSA fusion protein. An hNT-3-HSA fusion protein modified with something other than sugar chains and phosphate is also an hNT-3-HSA fusion protein. An hNT-3-HSA fusion protein in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like is also an hNT-3-HSA fusion protein. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said about a fusion protein (hNT-3-SA) of hNT-3 and SA of an animal species other than human.
 つまり,糖鎖により修飾されたhNT-3-HSA融合蛋白質は,元のアミノ酸配列を有するhNT-3-HSA融合蛋白質に含まれるものとする。また,リン酸により修飾されたhNT-3-HSA融合蛋白質は,元のアミノ酸配列を有するhNT-3-HSA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するhNT-3-HSA融合蛋白質に含まれるものとする。また,hNT-3-HSA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するhNT-3-HSA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。hNT-3とヒト以外の動物種のSAとの融合蛋白質(hNT-3-SA)についても同様のことがいえる。 In other words, hNT-3-HSA fusion proteins modified with sugar chains are included in the hNT-3-HSA fusion proteins having the original amino acid sequence. Also, hNT-3-HSA fusion proteins modified with phosphate are included in the hNT-3-HSA fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are included in the hNT-3-HSA fusion proteins having the original amino acid sequence. Also, hNT-3-HSA fusion proteins in which the side chains of the amino acids constituting the hNT-3-HSA fusion proteins have been converted by substitution reactions or the like are included in the hNT-3-HSA fusion proteins having the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for fusion proteins of hNT-3 and SA of animal species other than human (hNT-3-SA).
 また,hNT-3-HSA融合蛋白質であって,これを構成するhNT-3が,hNT-3の前駆体であるものもhNT-3-HSA融合蛋白質である。但し,hNT-3が前駆体であるhNT-3-HSA融合蛋白質が,hNT-3としての機能を示さない場合には,これがインビボ又はインビトロでプロセシングされてhNT-3としての機能を示すものに転換されるものであることが必要である。hNT-3とヒト以外の動物種のSAとの融合蛋白質(hNT-3-SA)についても同様のことがいえる。  An hNT-3-HSA fusion protein in which the hNT-3 that constitutes it is a precursor of hNT-3 is also an hNT-3-HSA fusion protein. However, if the hNT-3-HSA fusion protein, of which hNT-3 is a precursor, does not function as hNT-3, it is necessary that it be processed in vivo or in vitro and converted into one that functions as hNT-3. The same can be said for a fusion protein of hNT-3 and SA of an animal species other than human (hNT-3-SA).
 本発明の一実施形態において,「hNT-4-SA融合蛋白質」又は「hNT-4-SA」の語は,hNT-4のアミノ酸配列のC末端に,直接又はリンカーを介して,SAのアミノ酸配列のN末端が結合されているアミノ酸配列を有する融合蛋白質であって,hNT-4としての機能を有するものを示す。hNT-4-SA融合蛋白質において,hNT-4がhNT-4としての機能を有するというときは,hNT-4が,通常の野生型のhNT-4の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性を保持することをいう。なお,ここでhNT-4-SA融合蛋白質におけるhNT-4の比活性は,当該融合蛋白質の単位質量当たりのhNT-4の生理活性に(当該融合蛋白質の分子量/当該融合蛋白質中のhNT-4に相当する部分の分子量)を乗じて算出される。 In one embodiment of the present invention, the term "hNT-4-SA fusion protein" or "hNT-4-SA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of SA is linked, directly or via a linker, to the C-terminus of the amino acid sequence of hNT-4, and which has the function of hNT-4. When hNT-4 in an hNT-4-SA fusion protein is said to have the function of hNT-4, it means that hNT-4 retains a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, when the specific activity of normal wild-type hNT-4 is taken as 100%. Here, the specific activity of hNT-4 in the hNT-4-SA fusion protein is calculated by multiplying the physiological activity of hNT-4 per unit mass of the fusion protein by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hNT-4).
 本発明の一実施形態において,「hNT-4-HSA融合蛋白質」又は「hNT-4-HSA」の語は,hNT-4のアミノ酸配列のC末端に,直接又はリンカーを介して,HSAのアミノ酸配列のN末端が結合されている,アミノ酸配列を有する融合蛋白質であって,hNT-4としての機能を有するものを示す。ここでhNT-4-HSA融合蛋白質がhNT-4としての機能を有するというときは,上記のhNT-4-SA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, the term "hNT-4-HSA fusion protein" or "hNT-4-HSA" refers to a fusion protein having an amino acid sequence in which the N-terminus of the amino acid sequence of HSA is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of hNT-4, and which has the function of hNT-4. When it is said here that the hNT-4-HSA fusion protein has the function of hNT-4, the definition of the hNT-4-SA fusion protein described above can be applied.
 本発明の一実施形態において,ヒト以外の動物種のNT-4のアミノ酸配列のC末端に,直接又はリンカーを介して,ヒト以外の動物種のSAのN末端が結合されているアミノ酸配列を有する融合蛋白質であって,NT-4としての機能を有するものは,「(ヒト以外の動物種)NT-4-(ヒト以外の動物種)SA融合蛋白質」と表記される。例えば,マウスNT-4とマウスSAの融合蛋白質は,「マウスNT-4-マウスSA融合蛋白質」又は「マウスNT-4-MSA」と表記される。これら融合蛋白質についても,NT-4としての機能を有するというときは,上記のhNT-4-SA融合蛋白質における定義を適用することができる。 In one embodiment of the present invention, a fusion protein having an amino acid sequence in which the N-terminus of SA of a non-human animal species is linked, either directly or via a linker, to the C-terminus of the amino acid sequence of NT-4 of a non-human animal species, and which has the function of NT-4, is referred to as a "(non-human animal species) NT-4-(non-human animal species) SA fusion protein." For example, a fusion protein of mouse NT-4 and mouse SA is referred to as a "mouse NT-4-mouse SA fusion protein" or "mouse NT-4-MSA." When these fusion proteins are said to have the function of NT-4, the definition of hNT-4-SA fusion protein described above can be applied.
 本発明の一実施形態において好ましいhNT-4-HSA融合蛋白質は,例えば配列番号82で示されるアミノ酸配列を有するものである。配列番号82で示されるhNT-4-HSA融合蛋白質は,野生型hNT-4のC末端にリンカー配列Gly-Serを介して野生型HSAが結合したものである。配列番号82で示されるhNT-4-HSA融合蛋白質は,例えば,配列番号83で示される塩基配列を有する遺伝子にコードされる。また,配列番号82で示されるアミノ酸配列に対し,1個又は複数個のアミノ酸残基が,他のアミノ酸残基へ置換され,欠失し,或いは付加等の変異が加えられたアミノ酸配列を有するものも,hNT-4としての機能を有するものである限りhNT-4-HSA融合蛋白質に含まれる。hNT-4-HSA融合蛋白質は,血中の内因性の物質及び薬剤等の外因性の物質を結合して運搬する機能等のヒト血清アルブミンとしての機能を有するものであることが好ましいが,これに限定されない。 In one embodiment of the present invention, a preferred hNT-4-HSA fusion protein has, for example, the amino acid sequence shown in SEQ ID NO: 82. The hNT-4-HSA fusion protein shown in SEQ ID NO: 82 is obtained by linking wild-type HSA to the C-terminus of wild-type hNT-4 via the linker sequence Gly-Ser. The hNT-4-HSA fusion protein shown in SEQ ID NO: 82 is encoded by a gene having, for example, the base sequence shown in SEQ ID NO: 83. In addition, the hNT-4-HSA fusion protein also includes an amino acid sequence in which one or more amino acid residues in the amino acid sequence shown in SEQ ID NO: 82 have been replaced with other amino acid residues, deleted, or mutated by addition, as long as it has the function of hNT-4. The hNT-4-HSA fusion protein is preferably one that has the function of human serum albumin, such as the function of binding and transporting endogenous substances in blood and exogenous substances such as drugs, but is not limited thereto.
 配列番号82で示されるアミノ酸配列に変異を加えたときの各変異の位置及びその形式(欠失,置換,及び付加)は,変異前後のアミノ酸配列のアラインメントにより,容易に確認することができる。変異を加えたアミノ酸配列は,配列番号82で示されるアミノ酸配列と,好ましくは80%以上の同一性を示し,85%以上の同一性を示し,90%以上の同一性を示し,又は,95%以上の同一性を示し,例えば,98%以上,又は99%以上の同一性を示すものである。 The position and type (deletion, substitution, and addition) of each mutation when the amino acid sequence shown in SEQ ID NO:82 is mutated can be easily confirmed by alignment of the amino acid sequence before and after the mutation. The mutated amino acid sequence preferably has 80% or more identity, 85% or more identity, 90% or more identity, or 95% or more identity, for example, 98% or more, or 99% or more identity, with the amino acid sequence shown in SEQ ID NO:82.
 野生型hNT-4と野生型HSAとの融合蛋白質であるhNT-4-HSA融合蛋白質に変異を加える場合,HSA部分にのみ変異を加えhNT-4部分には変異を加えないことも,HSA部分には変異を加えずにhNT-4部分にのみ変異を加えることも,また,HSA部分とhNT-4部分の何れにも変異を加えることもできる。HSA部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となる。hNT-4部分にのみ変異を加える場合にあっては,当該部分のアミノ酸配列は上述した野生型hNT-4に変異が加えられたhNT-4のアミノ酸配列となる。HSA部分とhNT-4部分の何れにも変異を加える場合にあっては,HSA部分のアミノ酸配列は上述した野生型HSAに変異が加えられたHSAのアミノ酸配列となり,hNT-4部分のアミノ酸配列は上述した野生型hNT-4に変異が加えられたhNT-4のアミノ酸配列となる。配列番号82で示されるアミノ酸配列を有するhNT-4-HSA融合蛋白質についても同様である。野生型hNT-4とヒト以外の動物種の野生型SA(hNT-4-SA)との融合蛋白質についても同様のことがいえる。 When mutations are introduced into the hNT-4-HSA fusion protein, which is a fusion protein of wild-type hNT-4 and wild-type HSA, mutations can be introduced only in the HSA portion and not in the hNT-4 portion, mutations can be introduced only in the hNT-4 portion without mutations in the HSA portion, or mutations can be introduced in both the HSA portion and the hNT-4 portion. When mutations are introduced only in the HSA portion, the amino acid sequence of the portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above. When mutations are introduced only in the hNT-4 portion, the amino acid sequence of the portion is the amino acid sequence of hNT-4 obtained by adding a mutation to the wild-type hNT-4 described above. When mutations are introduced in both the HSA portion and the hNT-4 portion, the amino acid sequence of the HSA portion is the amino acid sequence of HSA obtained by adding a mutation to the wild-type HSA described above, and the amino acid sequence of the hNT-4 portion is the amino acid sequence of hNT-4 obtained by adding a mutation to the wild-type hNT-4 described above. The same is true for the hNT-4-HSA fusion protein having the amino acid sequence shown in SEQ ID NO: 82. The same is true for the fusion protein of wild-type hNT-4 and wild-type SA (hNT-4-SA) of an animal species other than human.
 配列番号82で示されるアミノ酸配列を有するhNT-4-HSA融合蛋白質に変異を加える場合について,以下例示する。配列番号82で示されるアミノ酸配列中のアミノ酸残基を他のアミノ酸残基で置換する場合,置換するアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を欠失させる場合,欠失させるアミノ酸残基の個数は,1~10個であり,1~5個であり,又は1~3個であり,例えば,1個又は2個である。アミノ酸残基を付加する場合,アミノ酸残基は配列番号82で示されるアミノ酸配列中に又は当該アミノ酸配列のN末端若しくはC末端に,1個又は複数個のアミノ酸残基が付加される。hNT-4-HSA融合蛋白質は,これらアミノ酸残基の置換,欠失,及び付加を組み合わせたものであってもよい。変異は,HSA部分にのみ加えても,hNT-4部分にのみ加えても,又は両方の部分に加えてもよい。hNT-4とヒト以外の動物種のSAとの融合蛋白質(hNT-4-SA)についても同様のことがいえる。 The following is an example of a case where a mutation is made to an hNT-4-HSA fusion protein having the amino acid sequence shown in SEQ ID NO:82. When an amino acid residue in the amino acid sequence shown in SEQ ID NO:82 is replaced with another amino acid residue, the number of amino acid residues to be replaced is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is deleted, the number of amino acid residues to be deleted is 1 to 10, 1 to 5, or 1 to 3, for example, 1 or 2. When an amino acid residue is added, one or more amino acid residues are added to the amino acid sequence shown in SEQ ID NO:82 or to the N-terminus or C-terminus of the amino acid sequence. The hNT-4-HSA fusion protein may be a combination of these substitutions, deletions, and additions of amino acid residues. The mutation may be made only to the HSA portion, only to the hNT-4 portion, or to both portions. The same can be said about the fusion protein of hNT-4 and SA of a non-human animal species (hNT-4-SA).
 hNT-4-HSA融合蛋白質であって,これを構成するアミノ酸が糖鎖により修飾されたものもhNT-4-HSA融合蛋白質である。また,hNT-4-HSA融合蛋白質であって,これを構成するアミノ酸がリン酸により修飾されたものもhNT-4-HSA融合蛋白質である。また,糖鎖及びリン酸以外のものにより修飾されたものもhNT-4-HSA融合蛋白質である。また,hNT-4-HSA融合蛋白質であって,これを構成するアミノ酸の側鎖が,置換反応等により変換したものもhNT-4-HSA融合蛋白質である。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のNT-4とHSAとの融合蛋白質(NT-4-SA),及びヒト以外の動物種のNT-4とヒト以外の動物種のSAとの融合蛋白質についても同様のことがいえる。  An hNT-4-HSA fusion protein in which the amino acids constituting the protein are modified with sugar chains is also an hNT-4-HSA fusion protein. An hNT-4-HSA fusion protein in which the amino acids constituting the protein are modified with phosphate is also an hNT-4-HSA fusion protein. An hNT-4-HSA fusion protein in which the amino acids constituting the protein are modified with something other than sugar chains and phosphate is also an hNT-4-HSA fusion protein. An hNT-4-HSA fusion protein in which the side chains of the amino acids constituting the protein have been converted by a substitution reaction or the like is also an hNT-4-HSA fusion protein. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for a fusion protein of NT-4 of a non-human animal species and HSA (NT-4-SA), and a fusion protein of NT-4 of a non-human animal species and SA of a non-human animal species.
 つまり,糖鎖により修飾されたhNT-4-HSA融合蛋白質は,元のアミノ酸配列を有するhNT-4-HSA融合蛋白質に含まれるものとする。また,リン酸により修飾されたhNT-4-HSA融合蛋白質は,元のアミノ酸配列を有するhNT-4-HSA融合蛋白質に含まれるものとする。また,糖鎖及びリン酸以外のものにより修飾されたものも,元のアミノ酸配列を有するhNT-4-HSA融合蛋白質に含まれるものとする。また,hNT-4-HSA融合蛋白質を構成するアミノ酸の側鎖が,置換反応等により変換したものも,元のアミノ酸配列を有するhNT-4-HSA融合蛋白質に含まれるものとする。かかる変換としては,システイン残基のホルミルグリシンへの変換があるが,これに限られるものではない。ヒト以外の動物種のNT-4とHSAとの融合蛋白質(NT-4-SA),及びヒト以外の動物種のNT-4とヒト以外の動物種のSAとの融合蛋白質についても同様のことがいえる。 In other words, hNT-4-HSA fusion proteins modified with sugar chains are included in the hNT-4-HSA fusion proteins having the original amino acid sequence. Also, hNT-4-HSA fusion proteins modified with phosphate are included in the hNT-4-HSA fusion proteins having the original amino acid sequence. Also, those modified with things other than sugar chains and phosphate are included in the hNT-4-HSA fusion proteins having the original amino acid sequence. Also, those in which the side chains of the amino acids constituting the hNT-4-HSA fusion protein have been converted by a substitution reaction or the like are included in the hNT-4-HSA fusion protein having the original amino acid sequence. Such conversions include, but are not limited to, the conversion of cysteine residues to formylglycine. The same can be said for the fusion protein of NT-4 of a non-human animal species and HSA (NT-4-SA), and the fusion protein of NT-4 of a non-human animal species and SA of a non-human animal species.
 また,hNT-4-HSA融合蛋白質であって,これを構成するhNT-4が,hNT-4の前駆体であるものもhNT-4-HSA融合蛋白質である。但し,hNT-4が前駆体であるhNT-4-HSA融合蛋白質が,hNT-4としての機能を示さない場合には,これがインビボ又はインビトロでプロセシングされてhNT-4としての機能を示すものに転換されるものであることが必要である。  An hNT-4-HSA fusion protein in which the hNT-4 that constitutes it is a precursor of hNT-4 is also an hNT-4-HSA fusion protein. However, if the hNT-4-HSA fusion protein, of which hNT-4 is a precursor, does not exhibit the function of hNT-4, it is necessary that it be processed in vivo or in vitro and converted into one that exhibits the function of hNT-4.
 本発明の一実施形態において,「HSAとヒト神経栄養因子との融合蛋白質」,「ヒト神経栄養因子とHSAとの融合蛋白質」,又は「ヒト血清アルブミンとヒト神経栄養因子との融合蛋白質」,というときは,上記の「HSA-ヒト神経栄養因子融合蛋白質」及び「ヒト神経栄養因子-HSA融合蛋白質」のいずれをも含む。これらのことは,SAがヒト以外の動物種である場合にも,又は,神経成長因子がヒト以外の動物種である場合にも適用される。 In one embodiment of the present invention, the terms "fusion protein of HSA and human neurotrophic factor", "fusion protein of human neurotrophic factor and HSA", or "fusion protein of human serum albumin and human neurotrophic factor" include both the above-mentioned "HSA-human neurotrophic factor fusion protein" and "human neurotrophic factor-HSA fusion protein". These also apply when SA is from an animal species other than human, or when nerve growth factor is from an animal species other than human.
 また,「HSAとhBDNFとの融合蛋白質」,「hBDNFとHSAとの融合蛋白質」,又は「ヒト血清アルブミンとヒト脳神経由来栄養因子との融合蛋白質」,というときは,上記の「HSA-hBDNF融合蛋白質」及び「hBDNF-HSA融合蛋白質」のいずれをも含む。これらのことは,SAがヒト以外の動物種である場合にも,又は,BDNFがヒト以外の動物種である場合にも適用される。  In addition, when we say "a fusion protein of HSA and hBDNF," "a fusion protein of hBDNF and HSA," or "a fusion protein of human serum albumin and human brain-derived trophic factor," we include both the above-mentioned "HSA-hBDNF fusion protein" and "hBDNF-HSA fusion protein." These also apply when SA is from an animal species other than human, or when BDNF is from an animal species other than human.
 また,「HSAとhNGFとの融合蛋白質」,「hNGFとHSAとの融合蛋白質」,又は「ヒト血清アルブミンとヒト神経成長因子との融合蛋白質」,というときは,上記の「HSA-hNGF融合蛋白質」及び「hNGF-HSA融合蛋白質」のいずれをも含む。これらのことは,SAがヒト以外の動物種である場合にも,又は,NGFがヒト以外の動物種である場合にも適用される。  In addition, when we say "a fusion protein of HSA and hNGF," "a fusion protein of hNGF and HSA," or "a fusion protein of human serum albumin and human nerve growth factor," we include both the above-mentioned "HSA-hNGF fusion protein" and "hNGF-HSA fusion protein." These also apply when SA is from an animal species other than human, or when NGF is from an animal species other than human.
 また,「HSAとhNT-3との融合蛋白質」,「hNT-3とHSAとの融合蛋白質」,又は「ヒト血清アルブミンとヒトニューロトロフィン-3との融合蛋白質」,というときは,上記の「HSA-hNT-3融合蛋白質」及び「hNT-3-HSA融合蛋白質」のいずれをも含む。これらのことは,SAがヒト以外の動物種である場合にも,又は,NT-3がヒト以外の動物種である場合にも適用される。 Furthermore, when we say "a fusion protein of HSA and hNT-3," "a fusion protein of hNT-3 and HSA," or "a fusion protein of human serum albumin and human neurotrophin-3," we include both the above-mentioned "HSA-hNT-3 fusion protein" and "hNT-3-HSA fusion protein." These also apply when SA is of an animal species other than human, or when NT-3 is of an animal species other than human.
 また,「HSAとhNT-4との融合蛋白質」,「hNT-4とHSAとの融合蛋白質」,又は「ヒト血清アルブミンとヒトニューロトロフィン-4との融合蛋白質」,というときは,上記の「HSA-hNT-4融合蛋白質」及び「hNT-4-HSA融合蛋白質」のいずれをも含む。これらのことは,SAがヒト以外の動物種である場合にも,又は,NT-4がヒト以外の動物種である場合にも適用される。 Furthermore, when we say "a fusion protein of HSA and hNT-4," "a fusion protein of hNT-4 and HSA," or "a fusion protein of human serum albumin and human neurotrophin-4," we include both the above-mentioned "HSA-hNT-4 fusion protein" and "hNT-4-HSA fusion protein." These also apply when SA is from an animal species other than human, or when NT-4 is from an animal species other than human.
 本発明の一実施形態のSAと神経栄養因子との融合蛋白質において,SAと神経栄養因子とは,直接又はリンカーを介して結合される。ここで,「リンカー」というときは,SAのアミノ酸配列と神経栄養因子のアミノ酸配列のいずれにも属さない部分のことをいう。つまり,リンカーはSAと神経栄養因子との間に介在するペプチド鎖のことである。リンカーは,種々の機能を有する。その機能には,SAと神経栄養因子との間にあってSAと神経栄養因子とを結合する機能の他,SA及び神経栄養因子の,融合蛋白質分子内での距離を離すことにより相互の干渉を低減させる機能,SAと神経栄養因子との間にあってSAと神経栄養因子とを連結するヒンジとなって,融合蛋白質の立体構造に柔軟性を与える機能等が含まれる。融合蛋白質の分子内において,リンカーは,これら機能の少なくとも一つを発揮する。これらのことは,SAの動物種,及び神経成長因子の動物種にかかわらず適用される。神経栄養因子がBDNF,NGF,NT-3又はNT-4である場合に限らず,他の神経栄養因子であっても適用される。 In one embodiment of the present invention, in the fusion protein of SA and a neurotrophic factor, the SA and the neurotrophic factor are linked directly or via a linker. Here, the term "linker" refers to a portion that does not belong to either the amino acid sequence of SA or the amino acid sequence of a neurotrophic factor. In other words, the linker is a peptide chain that is interposed between the SA and the neurotrophic factor. The linker has various functions. These functions include a function between the SA and the neurotrophic factor to bind the SA and the neurotrophic factor, a function to reduce mutual interference between the SA and the neurotrophic factor by increasing the distance between them in the fusion protein molecule, and a function between the SA and the neurotrophic factor to act as a hinge that connects the SA and the neurotrophic factor and gives flexibility to the three-dimensional structure of the fusion protein. In the molecule of the fusion protein, the linker exerts at least one of these functions. These are applicable regardless of the animal species of the SA and the animal species of the nerve growth factor. They are not limited to cases where the neurotrophic factor is BDNF, NGF, NT-3, or NT-4, but also apply to other neurotrophic factors.
 以下,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,及びHSAとhNT-4との融合蛋白質を例にとり,SAと神経栄養因子との融合蛋白質の製造法等について詳述する。ここで詳述される事項は,SAがヒト以外の動物種である場合にも,又は,BDNF,NGF,NT-3又は,NT-4がヒト以外の動物種である場合にも,適用することができる。また,BDNF,NGF,NT-3又は,NT-4以外の神経栄養因子にも適用することができる。  Below, the methods for producing fusion proteins of SA and neurotrophic factors are described in detail, taking as examples a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, and a fusion protein of HSA and hNT-4. The matters described in detail here can be applied when SA is from an animal species other than human, or when BDNF, NGF, NT-3, or NT-4 is from an animal species other than human. They can also be applied to neurotrophic factors other than BDNF, NGF, NT-3, or NT-4.
 HSAとhBDNFとの融合蛋白質は,HSAをコードする遺伝子の下流又は上流に,インフレームでhBDNFをコードする遺伝子を結合させたDNA断片を組み込んだ発現ベクターを作製し,この発現ベクターを導入して形質転換させた宿主細胞を培養することにより,組換え蛋白質として作製することができる。また,HSAとhNGFとの融合蛋白質は,HSAをコードする遺伝子の下流又は上流に,インフレームでhNGFをコードする遺伝子を結合させたDNA断片を組み込んだ発現ベクターを作製し,この発現ベクターを導入して形質転換させた宿主細胞を培養することにより,組換え蛋白質として作製することができる。また,HSAとhNT-3との融合蛋白質は,HSAをコードする遺伝子の下流又は上流に,インフレームでhNT-3をコードする遺伝子を結合させたDNA断片を組み込んだ発現ベクターを作製し,この発現ベクターを導入して形質転換させた宿主細胞を培養することにより,組換え蛋白質として作製することができる。また,HSAとhNT-4との融合蛋白質は,HSAをコードする遺伝子の下流又は上流に,インフレームでhNT-4をコードする遺伝子を結合させたDNA断片を組み込んだ発現ベクターを作製し,この発現ベクターを導入して形質転換させた宿主細胞を培養することにより,組換え蛋白質として作製することができる。このようにして組換え蛋白質として作製される融合蛋白質は,一本鎖ポリペプチドからなる。 A fusion protein of HSA and hBDNF can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hBDNF is linked in-frame downstream or upstream of a gene encoding HSA, and culturing a host cell transformed with the expression vector. A fusion protein of HSA and hNGF can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hNGF is linked in-frame downstream or upstream of a gene encoding HSA, and culturing a host cell transformed with the expression vector. A fusion protein of HSA and hNT-3 can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hNT-3 is linked in-frame downstream or upstream of a gene encoding HSA, and culturing a host cell transformed with the expression vector. In addition, a fusion protein of HSA and hNT-4 can be produced as a recombinant protein by preparing an expression vector incorporating a DNA fragment in which a gene encoding hNT-4 is linked in-frame to the downstream or upstream of a gene encoding HSA, and culturing a host cell transformed with the expression vector. The fusion protein produced as a recombinant protein in this way consists of a single polypeptide chain.
 組換え融合蛋白質として融合蛋白質を作製する場合,HSAをコードする遺伝子の下流にインフレームでhBDNFをコードする遺伝子を結合させることにより,HSAのアミノ酸配列のC末端にhBDNFのアミノ酸配列を有する融合蛋白質を得ることができる。逆に,HSAをコードする遺伝子の上流にインフレームでhBDNFをコードする遺伝子を結合させることにより,HSAのアミノ酸配列のN末端にhBDNFのアミノ酸配列を有する融合蛋白質が得られる。また,組換え融合蛋白質として融合蛋白質を作製する場合,HSAをコードする遺伝子の下流にインフレームでhNGFをコードする遺伝子を結合させることにより,HSAのアミノ酸配列のC末端にhNGFのアミノ酸配列を有する融合蛋白質を得ることができる。逆に,HSAをコードする遺伝子の上流にインフレームでhNGFをコードする遺伝子を結合させることにより,HSAのアミノ酸配列のN末端にhNGFのアミノ酸配列を有する融合蛋白質が得られる。また,組換え融合蛋白質として融合蛋白質を作製する場合,HSAをコードする遺伝子の下流にインフレームでhNT-3をコードする遺伝子を結合させることにより,HSAのアミノ酸配列のC末端にhNT-3のアミノ酸配列を有する融合蛋白質を得ることができる。逆に,HSAをコードする遺伝子の上流にインフレームでhNT-3をコードする遺伝子を結合させることにより,HSAのアミノ酸配列のN末端にhNT-3のアミノ酸配列を有する融合蛋白質が得られる。また,組換え融合蛋白質として融合蛋白質を作製する場合,HSAをコードする遺伝子の下流にインフレームでhNT-4をコードする遺伝子を結合させることにより,HSAのアミノ酸配列のC末端にhNT-4のアミノ酸配列を有する融合蛋白質を得ることができる。逆に,HSAをコードする遺伝子の上流にインフレームでhNT-4をコードする遺伝子を結合させることにより,HSAのアミノ酸配列のN末端にhNT-4のアミノ酸配列を有する融合蛋白質が得られる。いずれの場合においても,組換え融合蛋白質として作製される融合蛋白質は,一本鎖ポリペプチドである。 When preparing a fusion protein as a recombinant fusion protein, a gene encoding hBDNF can be linked in-frame downstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hBDNF at the C-terminus of the amino acid sequence of HSA. Conversely, a gene encoding hBDNF can be linked in-frame upstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hBDNF at the N-terminus of the amino acid sequence of HSA. When preparing a fusion protein as a recombinant fusion protein, a gene encoding hNGF can be linked in-frame downstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hNGF at the C-terminus of the amino acid sequence of HSA. Conversely, a gene encoding hNGF can be linked in-frame upstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hNGF at the N-terminus of the amino acid sequence of HSA. When preparing a fusion protein as a recombinant fusion protein, a gene encoding hNT-3 can be linked in-frame downstream of a gene encoding HSA to obtain a fusion protein having the amino acid sequence of hNT-3 at the C-terminus of the amino acid sequence of HSA. Conversely, by linking a gene encoding hNT-3 in frame upstream of the gene encoding HSA, a fusion protein having the amino acid sequence of hNT-3 at the N-terminus of the amino acid sequence of HSA can be obtained. In addition, when producing a fusion protein as a recombinant fusion protein, by linking a gene encoding hNT-4 in frame downstream of the gene encoding HSA, a fusion protein having the amino acid sequence of hNT-4 at the C-terminus of the amino acid sequence of HSA can be obtained. Conversely, by linking a gene encoding hNT-4 in frame upstream of the gene encoding HSA, a fusion protein having the amino acid sequence of hNT-4 at the N-terminus of the amino acid sequence of HSA can be obtained. In either case, the fusion protein produced as a recombinant fusion protein is a single-chain polypeptide.
 融合蛋白質の一本鎖ポリペプチド内で,HSAのアミノ酸配列がhBDNF,hNGF,hNT-3又はhNT-4のアミノ酸配列のN末端側に位置する場合,HSAのC末端とhBDNF,hNGF,hNT-3又はhNT-4のN末端が,ペプチド結合により直接,又はリンカーを介して結合される。ここで「リンカー」とは,当該一本鎖ポリペプチド内において,HSAのC末端とhBDNF,hNGF,hNT-3又はhNT-4のN末端との間に存在する,HSA及びhBDNF,hNGF,hNT-3又はhNT-4のいずれにも属さないアミノ酸配列を有する部分のことをいう。図7にN末端側からHSA,リンカー及びhBDNFを順に有する一本鎖ポリペプチドのHSA-hBDNF融合蛋白質を模式的に示す。該HSA-hBDNF融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とhBDNFのN末端がペプチド結合により結合したものである。また,図8にN末端側からHSA,リンカー及びhNGFを順に有する一本鎖ポリペプチドのHSA-hNGF融合蛋白質を模式的に示す。該HSA-hNGF融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とhNGFのN末端がペプチド結合により結合したものである。また,図9にN末端側からHSA,リンカー及びhNT-3を順に有する一本鎖ポリペプチドのHSA-hNT-3融合蛋白質を模式的に示す。該HSA-hNT-3融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とhNT-3のN末端がペプチド結合により結合したものである。また,図10にN末端側からHSA,リンカー及びhNT-4を順に有する一本鎖ポリペプチドのHSA-hNT-4融合蛋白質を模式的に示す。該HSA-hNT-4融合蛋白質はHSAのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とhNT-4のN末端がペプチド結合により結合したものである。 In the single-chain polypeptide of the fusion protein, when the amino acid sequence of HSA is located on the N-terminal side of the amino acid sequence of hBDNF, hNGF, hNT-3 or hNT-4, the C-terminus of HSA and the N-terminus of hBDNF, hNGF, hNT-3 or hNT-4 are linked directly by a peptide bond or via a linker. Here, the "linker" refers to a portion of the single-chain polypeptide that is present between the C-terminus of HSA and the N-terminus of hBDNF, hNGF, hNT-3 or hNT-4 and has an amino acid sequence that does not belong to HSA, hBDNF, hNGF, hNT-3 or hNT-4. Figure 7 shows a schematic diagram of a single-chain polypeptide HSA-hBDNF fusion protein that has HSA, a linker and hBDNF in this order from the N-terminus. In the HSA-hBDNF fusion protein, the C-terminus of HSA and the N-terminus of the linker are linked by a peptide bond, and the C-terminus of the linker and the N-terminus of hBDNF are linked by a peptide bond. FIG. 8 shows a schematic diagram of a single-chain polypeptide HSA-hNGF fusion protein having HSA, a linker, and hNGF in this order from the N-terminus. In the HSA-hNGF fusion protein, the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hNGF by a peptide bond. FIG. 9 shows a schematic diagram of a single-chain polypeptide HSA-hNT-3 fusion protein having HSA, a linker, and hNT-3 in this order from the N-terminus. In the HSA-hNT-3 fusion protein, the C-terminus of HSA is bound to the N-terminus of the linker by a peptide bond, and the C-terminus of the linker is bound to the N-terminus of hNT-3 by a peptide bond ... FIG. 10 shows a schematic diagram of a single-chain polypeptide HSA-hNT-4 fusion protein having HSA, a linker, and hNT-4 in this order from the N-terminus. The HSA-hNT-4 fusion protein is composed of the C-terminus of HSA and the N-terminus of the linker bound by a peptide bond, and the C-terminus of the linker and the N-terminus of hNT-4 bound by a peptide bond.
 また,融合蛋白質の一本鎖ポリペプチド内で,hBDNF,hNGF,hNT-3又はhNT-4のアミノ酸配列がHSAのアミノ酸配列のN末端に位置する場合,hBDNF,hNGF,hNT-3又はhNT-4のC末端とHSAのN末端が,ペプチド結合により直接,又はリンカーを介して結合される。ここで「リンカー」とは,当該一本鎖ポリペプチド内において,hBDNF,hNGF,hNT-3又はhNT-4のC末端及びHSAのN末端との間に存在する,HSA及びhBDNF,hNGF,hNT-3又はhNT-4のいずれにも属さないアミノ酸配列を有する部分のことをいう。図11にN末端側からhBDNF,リンカー及びHSAを順に有する一本鎖ポリペプチドのhBDNF-HSA融合蛋白質を模式的に示す。該hBDNF-HSA融合蛋白質はhBDNFのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。また,図12にN末端側からhNGF,リンカー及びHSAを順に有する一本鎖ポリペプチドのhNGF-HSA融合蛋白質を模式的に示す。該hNGF-HSA融合蛋白質はhNGFのC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。また,図13にN末端側からhNT-3,リンカー及びHSAを順に有する一本鎖ポリペプチドのhNT-3-HSA融合蛋白質を模式的に示す。該hNT-3-HSA融合蛋白質はhNT-3のC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。また,図14にN末端側からhNT-4,リンカー及びHSAを順に有する一本鎖ポリペプチドのhNT-4-HSA融合蛋白質を模式的に示す。該hNT-4-HSA融合蛋白質はhNT-4のC末端と該リンカーのN末端がペプチド結合により結合し,該リンカーのC末端とHSAのN末端がペプチド結合により結合したものである。 In addition, when the amino acid sequence of hBDNF, hNGF, hNT-3 or hNT-4 is located at the N-terminus of the amino acid sequence of HSA in the single-chain polypeptide of the fusion protein, the C-terminus of hBDNF, hNGF, hNT-3 or hNT-4 and the N-terminus of HSA are linked directly by a peptide bond or via a linker. Here, the "linker" refers to a portion of the single-chain polypeptide that is present between the C-terminus of hBDNF, hNGF, hNT-3 or hNT-4 and the N-terminus of HSA and has an amino acid sequence that does not belong to HSA, hBDNF, hNGF, hNT-3 or hNT-4. Figure 11 shows a schematic diagram of a single-chain polypeptide hBDNF-HSA fusion protein having hBDNF, a linker and HSA in that order from the N-terminus. In the hBDNF-HSA fusion protein, the C-terminus of hBDNF and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond. FIG. 12 shows a schematic representation of a single-chain polypeptide hNGF-HSA fusion protein having hNGF, a linker, and HSA in this order from the N-terminus. In the hNGF-HSA fusion protein, the C-terminus of hNGF and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond ... FIG. 13 shows a schematic representation of a single-chain polypeptide hNT-3-HSA fusion protein having hNT-3, a linker, and HSA in this order from the N-terminus. In the hNT-3-HSA fusion protein, the C-terminus of hNT-3 and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond. In addition, Figure 14 shows a schematic diagram of the hNT-4-HSA fusion protein, which is a single-chain polypeptide having, in order from the N-terminus, hNT-4, a linker, and HSA. In the hNT-4-HSA fusion protein, the C-terminus of hNT-4 and the N-terminus of the linker are bound by a peptide bond, and the C-terminus of the linker and the N-terminus of HSA are bound by a peptide bond.
 ペプチドリンカーのアミノ酸配列は,融合蛋白質分子の中にあって,リンカーとしての機能が発揮されるものである限り特に限定はない。また,ペプチドリンカーの長さにも,融合蛋白質分子の中にあって,リンカーとしての機能が発揮されるものである限り特に限定はない。ペプチドリンカーは一個又は複数個のアミノ酸から構成されるものである。ペプチドリンカーが複数個のアミノ酸から構成される場合,そのアミノ酸の個数は,好ましくは2~50個であり,より好ましくは5~30個であり,更に好ましくは10~25個である。ペプチドリンカーの好適な例として,Gly-Ser,Gly-Gly-Ser,又は配列番号9~11で示されるアミノ酸配列(これらをあわせて基本配列という)からなるもの,及びこれらを含むものが挙げられる。例えば,ペプチドリンカーは,基本配列が2~10回反復したアミノ酸配列を含むものであり,基本配列が2~6回反復したアミノ酸配列を含むものであり,基本配列が3~5回反復したアミノ酸配列を含むものである。これらのアミノ酸配列中の1個又は複数個のアミノ酸が,欠失,他のアミノ酸へ置換,付加等されたものであってもよい。アミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1又は2個である。アミノ酸を他のアミノ酸へ置換させる場合,置換させるアミノ酸の個数は,好ましくは1又は2個である。アミノ酸を付加する場合,付加されるアミノ酸の個数は,好ましくは1又は2個である。これらアミノ酸の欠失,置換,及び付加を組み合わせて,所望のリンカー部のアミノ酸配列とすることもできる。ペプチドリンカーは一つのアミノ酸からなるものであってもよく,リンカーを構成するアミノ酸は,例えばグリシン,セリンである。 There is no particular limitation on the amino acid sequence of the peptide linker, so long as it functions as a linker in the fusion protein molecule. There is also no particular limitation on the length of the peptide linker, so long as it functions as a linker in the fusion protein molecule. The peptide linker is composed of one or more amino acids. When the peptide linker is composed of multiple amino acids, the number of amino acids is preferably 2 to 50, more preferably 5 to 30, and even more preferably 10 to 25. Suitable examples of peptide linkers include those consisting of Gly-Ser, Gly-Gly-Ser, or amino acid sequences shown in SEQ ID NOs: 9 to 11 (collectively referred to as basic sequences), and those containing these. For example, the peptide linker is one that contains an amino acid sequence in which the basic sequence is repeated 2 to 10 times, one that contains an amino acid sequence in which the basic sequence is repeated 2 to 6 times, or one that contains an amino acid sequence in which the basic sequence is repeated 3 to 5 times. One or more amino acids in these amino acid sequences may be deleted, replaced with other amino acids, added, etc. When deleting an amino acid, the number of amino acids to be deleted is preferably 1 or 2. When replacing an amino acid with another amino acid, the number of amino acids to be replaced is preferably 1 or 2. When adding an amino acid, the number of amino acids to be added is preferably 1 or 2. The amino acid sequence of the desired linker portion can be obtained by combining these deletions, substitutions, and additions of amino acids. The peptide linker may be composed of one amino acid, and the amino acid that constitutes the linker is, for example, glycine or serine.
 本発明の一実施形態において,
 (1)HSAとhBDNFとの融合蛋白質というときは,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件下で野生型のhBDNFを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhBDNFとしての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となることを特徴とする融合蛋白質であり,
 (2)本発明の一実施形態において,HSAとhNGFとの融合蛋白質というときは,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件下で野生型のhNGFを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhNGFとしての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となることを特徴とする融合蛋白質であり,
 (3)本発明の一実施形態において,HSAとhNT-3との融合蛋白質というときは,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件下で野生型のhNT-3を組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhNT-3としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となることを特徴とする融合蛋白質であり,
 (4)本発明の一実施形態において,HSAとhNT-4との融合蛋白質というときは,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件下で野生型のhNT-4を組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhNT-4としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となることを特徴とする融合蛋白質である。
In one embodiment of the present invention,
(1) A fusion protein of HSA and hBDNF refers to a fusion protein characterized in that when expressed in a host cell as a recombinant protein, particularly when expressed so that the recombinant protein is secreted from the cells and accumulated in the culture medium, the amount of hBDNF expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hBDNF is expressed in a host cell as a recombinant protein under the same conditions, such as 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc.
(2) In one embodiment of the present invention, the term "fusion protein of HSA and hNGF" refers to a fusion protein characterized in that, when expressed in a host cell as a recombinant protein, in particular when expressed so that the recombinant protein is secreted from the cells and accumulated in the culture medium, the amount of hNGF expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hNGF is expressed in a host cell as a recombinant protein under the same conditions, such as 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc.
(3) In one embodiment of the present invention, a fusion protein of HSA and hNT-3 refers to a fusion protein characterized in that, when expressed in a host cell as a recombinant protein, particularly when expressed so that the recombinant protein is secreted from the cells and accumulated in the culture medium, the expression amount of hNT-3 in the culture supernatant is at least 1.1 times or more, 1.2 times or more, 1.5 times or more, 2 times or more, 2.5 times or more, or 3.5 times or more, in terms of concentration or physiological activity, compared to when wild-type hNT-3 is expressed in a host cell as a recombinant protein under the same conditions,
(4) In one embodiment of the present invention, a fusion protein of HSA and hNT-4 refers to a fusion protein characterized in that, when expressed in host cells as a recombinant protein, in particular when expressed so that the recombinant protein is secreted from the cells and accumulated in the culture medium, the expression amount of hNT-4 in the culture supernatant is at least 1.1 times or more, 1.2 times or more, 1.5 times or more, 2 times or more, 2.5 times or more, or 3.5 times or more, in terms of concentration or physiological activity, compared to when wild-type hNT-4 is expressed in host cells as a recombinant protein under the same conditions.
 上記(1)~(4)の実施形態において,同一条件下とは,発現ベクター,宿主細胞,培養条件等が同一であることをいう。このとき用いられる好ましい宿主細胞は,CHO細胞,NS/0細胞等の哺乳動物細胞であるが,特にCHO細胞である。 In the above embodiments (1) to (4), "under the same conditions" means that the expression vector, host cells, culture conditions, etc. are the same. The preferred host cells used in this case are mammalian cells such as CHO cells and NS/0 cells, and particularly CHO cells.
 かかるHSAとhBDNFとの融合蛋白質の好ましい実施形態として,以下の(1)~(4)が挙げられる:
(1)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号60で示される野生型のhBDNFのアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号68で示されるアミノ酸配列を有するもの,
(2)配列番号60で示される野生型のhBDNFのアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号76で示されるアミノ酸配列を有するもの。
(3)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号84で示される野生型のhBDNF(プロ体)のアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号85で示されるアミノ酸配列を有するもの,
(4)配列番号84で示される野生型のhBDNF(プロ体)のアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号86で示されるアミノ酸配列を有するもの。
Preferred embodiments of the fusion protein of HSA and hBDNF include the following (1) to (4):
(1) A peptide having an amino acid sequence shown in SEQ ID NO:68, in which the N-terminus of the amino acid sequence of wild-type hBDNF shown in SEQ ID NO:60 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO:3 via the linker sequence Gly-Ser;
(2) Having the amino acid sequence shown in SEQ ID NO: 76, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hBDNF shown in SEQ ID NO: 60 via the linker sequence Gly-Ser.
(3) A peptide having an amino acid sequence shown in SEQ ID NO: 85, in which the N-terminus of the amino acid sequence of wild-type hBDNF (pro-form) shown in SEQ ID NO: 84 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via the linker sequence Gly-Ser;
(4) Having the amino acid sequence shown in SEQ ID NO: 86, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hBDNF (pro-form) shown in SEQ ID NO: 84 via the linker sequence Gly-Ser.
 また,かかるHSAとhNGFとの融合蛋白質の好ましい実施形態として,以下の(1)~(4)が挙げられる:
(1)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号62で示される野生型のhNGFのアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号70で示されるアミノ酸配列を有するもの,
(2)配列番号62で示される野生型のhNGFのアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号78で示されるアミノ酸配列を有するもの。
(3)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号87で示される野生型のhNGF(プロ体)のアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号88で示されるアミノ酸配列を有するもの,
(4)配列番号87で示される野生型のhNGF(プロ体)のアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号89で示されるアミノ酸配列を有するもの。
Preferred embodiments of the fusion protein of HSA and hNGF include the following (1) to (4):
(1) A peptide having an amino acid sequence shown in SEQ ID NO: 70, in which the N-terminus of the amino acid sequence of wild-type hNGF shown in SEQ ID NO: 62 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via the linker sequence Gly-Ser;
(2) Having the amino acid sequence shown in SEQ ID NO: 78, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hNGF shown in SEQ ID NO: 62 via the linker sequence Gly-Ser.
(3) A peptide having an amino acid sequence shown in SEQ ID NO: 88, in which the N-terminus of the amino acid sequence of wild-type hNGF (pro-form) shown in SEQ ID NO: 87 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via the linker sequence Gly-Ser;
(4) Having the amino acid sequence shown in SEQ ID NO: 89, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hNGF (pro-form) shown in SEQ ID NO: 87 via the linker sequence Gly-Ser.
 また,かかるHSAとhNT-3との融合蛋白質の好ましい実施形態として,以下の(1)~(4)が挙げられる:
(1)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号64で示される野生型のhNT-3のアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号72で示されるアミノ酸配列を有するもの,
(2)配列番号64で示される野生型のhNT-3のアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号80で示されるアミノ酸配列を有するもの。
(3)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号90で示される野生型のhNT-3(プロ体)のアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号91で示されるアミノ酸配列を有するもの,
(4)配列番号90で示される野生型のhNT-3(プロ体)のアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号92で示されるアミノ酸配列を有するもの。
Furthermore, preferred embodiments of the fusion protein of HSA and hNT-3 include the following (1) to (4):
(1) A peptide having an amino acid sequence shown in SEQ ID NO: 72, in which the N-terminus of the amino acid sequence of wild-type hNT-3 shown in SEQ ID NO: 64 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via the linker sequence Gly-Ser;
(2) Having the amino acid sequence shown in SEQ ID NO: 80, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hNT-3 shown in SEQ ID NO: 64 via the linker sequence Gly-Ser.
(3) A peptide having an amino acid sequence shown in SEQ ID NO: 91, in which the N-terminus of the amino acid sequence of wild-type hNT-3 (pro-form) shown in SEQ ID NO: 90 is linked via a linker sequence Gly-Ser to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3;
(4) Having the amino acid sequence shown in SEQ ID NO: 92, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hNT-3 (pro-form) shown in SEQ ID NO: 90 via the linker sequence Gly-Ser.
 また,かかるHSAとhNT-4との融合蛋白質の好ましい実施形態として,以下の(1)~(4)が挙げられる:
(1)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号66で示される野生型のhNT-4のアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号74で示されるアミノ酸配列を有するもの,
(2)配列番号66で示される野生型のhNT-4のアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号82で示されるアミノ酸配列を有するもの。
(3)配列番号3で示される野生型のHSAのアミノ酸配列のC末端に,配列番号93で示される野生型のhNT-4(プロ体)のアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号94で示されるアミノ酸配列を有するもの,
(4)配列番号93で示される野生型のhNT-4(プロ体)のアミノ酸配列のC末端に,配列番号3で示される野生型のHSAのアミノ酸配列のN末端がリンカー配列Gly-Serを介して結合したものである,配列番号95で示されるアミノ酸配列を有するもの。
Preferred embodiments of the fusion protein of HSA and hNT-4 include the following (1) to (4):
(1) A peptide having an amino acid sequence shown in SEQ ID NO: 74, in which the N-terminus of the amino acid sequence of wild-type hNT-4 shown in SEQ ID NO: 66 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via the linker sequence Gly-Ser;
(2) Having the amino acid sequence shown in SEQ ID NO: 82, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 is linked to the C-terminus of the amino acid sequence of wild-type hNT-4 shown in SEQ ID NO: 66 via the linker sequence Gly-Ser.
(3) A peptide having an amino acid sequence shown in SEQ ID NO: 94, in which the N-terminus of the amino acid sequence of wild-type hNT-4 (pro-form) shown in SEQ ID NO: 93 is linked to the C-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO: 3 via the linker sequence Gly-Ser;
(4) Having the amino acid sequence shown in SEQ ID NO:95, in which the N-terminus of the amino acid sequence of wild-type HSA shown in SEQ ID NO:3 is linked to the C-terminus of the amino acid sequence of wild-type hNT-4 (pro-form) shown in SEQ ID NO:93 via the linker sequence Gly-Ser.
 本発明の一実施形態におけるHSAとhBDNFとの融合蛋白質は,組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件下で野生型のhBDNFを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhBDNFとしての発現量が,濃度あるいは生理活性換算で増加することを特徴とするものである。従って,HSAとhBDNFとの融合蛋白質は,組換え蛋白質として製造したときに,野生型のhBDNFと比較して生産効率を上昇させることができるので,生産コストを低減することができる。なお,組換え蛋白質を有効成分として含有する医薬品は,非常に高価であることが知られている。従って,同一条件で製造したときに得られる組換え蛋白質の量を数パーセント,例えば3~9%上昇させることにも,多大な経済的効果がある。同様のことは,神経栄養因子,例えば,hBDNF,hNGF,hNT-3,及びhNT-4についてもいえる。 In one embodiment of the present invention, the fusion protein of HSA and hBDNF is characterized in that when expressed in a host cell as a recombinant protein, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hBDNF expressed in the culture supernatant is increased in terms of concentration or physiological activity compared to when wild-type hBDNF is expressed in a host cell as a recombinant protein under the same conditions. Therefore, when produced as a recombinant protein, the fusion protein of HSA and hBDNF can increase production efficiency compared to wild-type hBDNF, thereby reducing production costs. It is known that pharmaceuticals containing recombinant proteins as active ingredients are very expensive. Therefore, even increasing the amount of recombinant protein obtained when produced under the same conditions by a few percent, for example 3 to 9%, has a significant economic effect. The same can be said for neurotrophic factors such as hBDNF, hNGF, hNT-3, and hNT-4.
 なお,本明細書において,組換え蛋白質として宿主細胞で発現させたHSAとhBDNFとの融合蛋白質と,同一条件下で組換え蛋白質として宿主細胞で発現させた野生型のhBDNFの,発現量を比較するときは,発現した蛋白質の質量で比較するのではなく,発現した蛋白質の分子数またはhBDNF生理活性で比較するものとする。このルールは,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,HSAとhNT-4との融合蛋白質についても適用される。また,このルールは,融合蛋白質中を構成するSAの動物種,及び神経成長因子の動物種にかかわらず適用される。 In this specification, when comparing the expression levels of a fusion protein of HSA and hBDNF expressed in a host cell as a recombinant protein with wild-type hBDNF expressed in a host cell under the same conditions, the comparison is not based on the mass of the expressed protein, but on the number of molecules of the expressed protein or on the hBDNF biological activity. This rule also applies to fusion proteins of HSA and hNGF, fusion proteins of HSA and hNT-3, and fusion proteins of HSA and hNT-4. This rule also applies regardless of the animal species of the SA and nerve growth factor that make up the fusion protein.
 HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,及びHSAとhNT-4との融合蛋白質は,当該融合蛋白質をコードする遺伝子を組み込んだ発現ベクターを用いて形質転換させた宿主細胞を培養することにより,組換え蛋白質として製造することができる。このとき,上述したHSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質を製造するために用いることのできる,発現ベクター,宿主細胞,培地等を用いて製造することができる。 HSA and hBDNF fusion protein, HSA and hNGF fusion protein, HSA and hNT-3 fusion protein, and HSA and hNT-4 fusion protein can be produced as recombinant proteins by culturing host cells transformed with an expression vector incorporating a gene encoding the fusion protein. In this case, they can be produced using an expression vector, host cells, culture medium, etc. that can be used to produce the above-mentioned HSA and hGALC fusion protein or HSA and hGBA fusion protein.
 HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質をコードする宿主細胞を培養することにより,細胞内又は培地中に発現させることができる。これらの融合蛋白質は,カラムクロマトグラフィー等の方法により不純物から分離し,精製することができる。 By culturing host cells encoding a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, the protein can be expressed in cells or in the medium. These fusion proteins can be separated from impurities and purified by methods such as column chromatography.
 精製されたHSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質は,医薬組成物として使用することができる。特に,HSAとhBDNFとの融合蛋白質は,アルツハイマー病,パーキンソン病,ハンチントン病などの神経変性疾患,筋萎縮性側策硬化症などの脊髄変性疾患,この他,糖尿病性神経障害,脳虚血性疾患,Rett症候群などの発達障害,統合失調症,うつ病およびRett症候群等を対象疾患とする医薬組成物として使用することができる。また特に,HSAとhNGFとの融合蛋白質は,アルツハイマー病,パーキンソン病,ハンチントン病などの神経変性疾患等を対象疾患とする医薬組成物として使用することができる。また特に,HSAとhNT-3との融合蛋白質及びHSAとhNT-4との融合蛋白質は,神経変性疾患等を対象疾患とする医薬組成物として使用することができる。  The purified fusion protein of HSA and hBDNF, the fusion protein of HSA and hNGF, the fusion protein of HSA and hNT-3, or the fusion protein of HSA and hNT-4 can be used as a pharmaceutical composition. In particular, the fusion protein of HSA and hBDNF can be used as a pharmaceutical composition for treating neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, spinal degenerative diseases such as amyotrophic lateral sclerosis, and other diseases such as diabetic neuropathy, cerebral ischemic disease, developmental disorders such as Rett syndrome, schizophrenia, depression, and Rett syndrome. In particular, the fusion protein of HSA and hNGF can be used as a pharmaceutical composition for treating neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. In particular, the fusion protein of HSA and hNT-3 and the fusion protein of HSA and hNT-4 can be used as a pharmaceutical composition for treating neurodegenerative diseases.
 HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質を有効成分として含有してなる医薬組成物は,注射剤として静脈内,筋肉内,腹腔内,皮下又は脳室内に投与することができる。それらの注射剤は,凍結乾燥製剤又は水性液剤として供給することができる。水性液剤とする場合,バイアルに充填した形態としてもよく,注射器に予め充填したものであるプレフィルド型の製剤として供給することもできる。凍結乾燥製剤の場合,使用前に水性媒質に溶解し復元して使用する。  A pharmaceutical composition containing as an active ingredient a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4 can be administered intravenously, intramuscularly, intraperitoneally, subcutaneously, or intracerebroventricularly as an injection. These injections can be supplied as lyophilized preparations or aqueous liquid preparations. In the case of an aqueous liquid preparation, it may be in the form of a vial filled with the preparation, or it can be supplied as a prefilled preparation in which the preparation is filled in a syringe beforehand. In the case of a lyophilized preparation, it is dissolved in an aqueous medium and reconstituted before use.
 HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質は,これらを更に抗体又はリガンドと結合させた結合体とすることができる。例えば,HSAとhBDNF又はリガンドとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,及びHSAとhNT-4との融合蛋白質は,それぞれ脳血管内皮細胞上の受容体と特異的に結合することのできる抗体又はリガンドとの結合体とすることができる。HSAとhBDNF,HSAとhNGF,HSAとhNT-3,及びHSAとhNT-4との融合蛋白質は,脳血管内皮細胞上の受容体と特異的に結合することのできる抗体又はリガンドとの結合体とすることにより,脳血管内皮細胞上の受容体に結合できるようになる。脳血管内皮細胞上の受容体に結合した融合蛋白質は,血液脳関門(BBB)を通過して,中枢神経系(CNS)の組織に到達できる。従って,これら融合蛋白質は,かかる抗体又はリガンドとの結合体とすることで,血液脳関門(BBB)を通過させて,中枢神経系(CNS)において機能を発揮させるようにすることができる。SAがヒト以外の動物種のSAである場合,BDNF,NGF,hNT-3,及びhNT-4が,ヒト以外の動物種のものである場合にも同様である。また,神経栄養因子が,BDNF,NGF,hNT-3,及びhNT-4以外の神経栄養因子である場合でも同様である。 The fusion protein of HSA and hBDNF, the fusion protein of HSA and hNGF, the fusion protein of HSA and hNT-3, or the fusion protein of HSA and hNT-4 can be further bound to an antibody or a ligand to form a conjugate. For example, the fusion protein of HSA and hBDNF or a ligand, the fusion protein of HSA and hNGF, the fusion protein of HSA and hNT-3, and the fusion protein of HSA and hNT-4 can each be bound to an antibody or a ligand that can specifically bind to a receptor on cerebrovascular endothelial cells. The fusion proteins of HSA and hBDNF, HSA and hNGF, HSA and hNT-3, and HSA and hNT-4 can bind to the receptor on cerebrovascular endothelial cells by being bound to an antibody or a ligand that can specifically bind to the receptor on cerebrovascular endothelial cells. The fusion proteins bound to the receptor on cerebrovascular endothelial cells can pass through the blood-brain barrier (BBB) and reach the tissues of the central nervous system (CNS). Therefore, these fusion proteins can be conjugated with such antibodies or ligands to pass through the blood-brain barrier (BBB) and exert their functions in the central nervous system (CNS). The same applies when the SA is from an animal species other than human, and when BDNF, NGF, hNT-3, and hNT-4 are from an animal species other than human. The same applies when the neurotrophic factors are neurotrophic factors other than BDNF, NGF, hNT-3, and hNT-4.
 HSAとhBDNFとの融合蛋白質と抗体又はリガンドとの結合体において,hBDNFがhBDNFとしての機能を有するというときは,hBDNFが,通常の野生型のhBDNFの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性,例えば90%以上,95%以上の比活性を有することをいう。なお,ここで当該結合体におけるhBDNFの比活性は,当該結合体の単位質量当たりのhBDNFの生理活性に(当該結合体の分子量/当該結合体中のhBDNFに相当する部分の分子量)を乗じて算出される。これらは,SAがヒト以外の動物種に由来するものである場合にも,BDNFがヒト以外の動物種に由来するものである場合にも適用される。 When hBDNF is said to have the function of hBDNF in a conjugate of a fusion protein of HSA and hBDNF with an antibody or ligand, it means that hBDNF has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more, when the specific activity of normal wild-type hBDNF is taken as 100%. Here, the specific activity of hBDNF in the conjugate is calculated by multiplying the physiological activity of hBDNF per unit mass of the conjugate by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hBDNF). These apply both when SA is derived from an animal species other than human and when BDNF is derived from an animal species other than human.
 また,HSAとhNGFとの融合蛋白質と抗体又はリガンドとの結合体において,hNGFがhNGFとしての機能を有するというときは,hNGFが,通常の野生型のhNGFの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性,例えば90%以上,95%以上の比活性を有することをいう。なお,ここで当該結合体におけるhNGFの比活性は,当該結合体の単位質量当たりのhNGFの生理活性に(当該結合体の分子量/当該結合体中のhNGFに相当する部分の分子量)を乗じて算出される。これらは,SAがヒト以外の動物種に由来するものである場合にも,NGFがヒト以外の動物種に由来するものである場合にも適用される。 In addition, when hNGF is said to have the function of hNGF in a conjugate of a fusion protein of HSA and hNGF with an antibody or ligand, it means that hNGF has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more, when the specific activity of normal wild-type hNGF is taken as 100%. Here, the specific activity of hNGF in the conjugate is calculated by multiplying the physiological activity of hNGF per unit mass of the conjugate by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hNGF). These apply whether SA is derived from an animal species other than human or NGF is derived from an animal species other than human.
 また,HSAとhNT-3との融合蛋白質と抗体又はリガンドとの結合体において,hNT-3がhNT-3としての機能を有するというときは,hNT-3が,通常の野生型のhNT-3の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性,例えば90%以上,95%以上の比活性を有することをいう。なお,ここで当該結合体におけるhNT-3の比活性は,当該結合体の単位質量当たりのhNT-3の生理活性に(当該結合体の分子量/当該結合体中のhNT-3に相当する部分の分子量)を乗じて算出される。これらは,SAがヒト以外の動物種に由来するものである場合にも,NT-3がヒト以外の動物種に由来するものである場合にも適用される。 In addition, when hNT-3 in a conjugate of a fusion protein of HSA and hNT-3 with an antibody or ligand is said to have the function of hNT-3, it means that hNT-3 has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more, when the specific activity of normal wild-type hNT-3 is taken as 100%. Here, the specific activity of hNT-3 in the conjugate is calculated by multiplying the physiological activity of hNT-3 per unit mass of the conjugate by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hNT-3). These apply both when SA is derived from an animal species other than human and when NT-3 is derived from an animal species other than human.
 また,HSAとhNT-4との融合蛋白質と抗体又はリガンドとの結合体において,hNT-4がhNT-4としての機能を有するというときは,hNT-4が,通常の野生型のhNT-4の比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性,例えば90%以上,95%以上の比活性を有することをいう。なお,ここで当該結合体におけるhNT-4の比活性は,当該結合体の単位質量当たりのhNT-4の生理活性に(当該結合体の分子量/当該結合体中のhNT-4に相当する部分の分子量)を乗じて算出される。これらは,SAがヒト以外の動物種に由来するものである場合にも,NT-4がヒト以外の動物種に由来するものである場合にも適用される。 In addition, when hNT-4 is said to have the function of hNT-4 in a conjugate of a fusion protein of HSA and hNT-4 with an antibody or ligand, it means that hNT-4 has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more, 95% or more, when the specific activity of normal wild-type hNT-4 is taken as 100%. Here, the specific activity of hNT-4 in the conjugate is calculated by multiplying the physiological activity of hNT-4 per unit mass of the conjugate by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hNT-4). These apply both when SA is derived from an animal species other than human and when NT-4 is derived from an animal species other than human.
 本発明の一実施形態において,HSAとhBDNFとの融合蛋白質と抗体又はリガンドとの結合体というときは,当該結合体を組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhBDNFを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhBDNFとしての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となることを特徴とするものである。 In one embodiment of the present invention, a conjugate between a fusion protein of HSA and hBDNF and an antibody or ligand is characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hBDNF expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hBDNF is expressed as a recombinant protein in a host cell under the same conditions, such as 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc.
 また,本発明の一実施形態において,HSAとhNGFとの融合蛋白質と抗体又はリガンドとの結合体というときは,当該結合体を組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhNGFを組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhNGFとしての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となることを特徴とするものである。 In one embodiment of the present invention, a conjugate between a fusion protein of HSA and hNGF and an antibody or ligand is characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the amount of hNGF expressed in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hNGF is expressed as a recombinant protein in a host cell under the same conditions.
 また,本発明の一実施形態において,HSAとhNT-3との融合蛋白質と抗体又はリガンドとの結合体というときは,当該結合体を組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhNT-3を組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhNT-3としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となることを特徴とするものである。 In one embodiment of the present invention, a conjugate between a fusion protein of HSA and hNT-3 and an antibody or ligand is characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hNT-3 in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hNT-3 is expressed as a recombinant protein in a host cell under the same conditions.
 また,本発明の一実施形態において,HSAとhNT-4との融合蛋白質と抗体又はリガンドとの結合体というときは,当該結合体を組換え蛋白質として宿主細胞で発現させたとき,特に組換え蛋白質を細胞から分泌して培養液中に蓄積するように発現させたときに,同一条件で野生型のhNT-4を組換え蛋白質として宿主細胞で発現させたときと比較して,培養上清中のhNT-4としての発現量が,濃度あるいは生理活性換算で,少なくとも1.1倍以上,1.2倍以上,1.5倍以上,2倍以上,2.5倍以上,又は3.5倍以上,例えば,1.1~4倍,1.5~3.6倍,2~3.6倍,10~20倍,20~30倍等となることを特徴とするものである。 In one embodiment of the present invention, a conjugate between a fusion protein of HSA and hNT-4 and an antibody or ligand is characterized in that when the conjugate is expressed as a recombinant protein in a host cell, particularly when the recombinant protein is expressed so as to be secreted from the cells and accumulated in the culture medium, the expression amount of hNT-4 in the culture supernatant is at least 1.1 times, 1.2 times, 1.5 times, 2 times, 2.5 times, or 3.5 times, in terms of concentration or physiological activity, compared to when wild-type hNT-4 is expressed as a recombinant protein in a host cell under the same conditions, and is 1.1 to 4 times, 1.5 to 3.6 times, 2 to 3.6 times, 10 to 20 times, 20 to 30 times, etc.
 神経栄養因子の中には,野生型のものをコードする遺伝子を組込んだ発現ベクターを導入した宿主細胞を用いて組換え蛋白質として発現させた場合に,発現量が限定的であるため大量に製造することが困難なものがある。かかる神経栄養因子としてBDNF,NGF,NT-3,及びNT-4が挙げられる。本発明の一実施形態は,このような組換え蛋白質として製造が困難な神経栄養因子を,SAとの融合蛋白質とした組換え蛋白質である。かかる組換え蛋白質は,これをコードする遺伝子を組込んだ発現ベクターを導入した宿主細胞を用いて,大量に製造することが,対応する野生型神経栄養因子と比較して,容易になる。ここで,SAと結合させる神経栄養因子の動物種に特に制限はないが,好ましくはヒト神経栄養因子である。また,神経栄養因子と結合させるSAの動物種に特に制限はないが,好ましくはHSAである。 Some neurotrophic factors are difficult to produce in large quantities when expressed as recombinant proteins using host cells introduced with an expression vector incorporating a gene encoding the wild-type neurotrophic factor, because the expression level is limited. Examples of such neurotrophic factors include BDNF, NGF, NT-3, and NT-4. One embodiment of the present invention is a recombinant protein in which such a neurotrophic factor that is difficult to produce as a recombinant protein is fused with SA. Such a recombinant protein can be produced in large quantities more easily using host cells introduced with an expression vector incorporating a gene encoding the recombinant protein, compared to the corresponding wild-type neurotrophic factor. Here, there is no particular restriction on the animal species of the neurotrophic factor to be bound to SA, but human neurotrophic factor is preferred. There is also no particular restriction on the animal species of the SA to be bound to the neurotrophic factor, but HSA is preferred.
 本発明において,「抗体」の語は,主としてヒト抗体,マウス抗体,ヒト化抗体,ラクダ科動物(アルパカを含む)由来の抗体,ヒト抗体と他の哺乳動物の抗体とのキメラ抗体,及びマウス抗体と他の哺乳動物の抗体とのキメラ抗体のことをいうが,特定の抗原に特異的に結合する性質を有するものである限り,これらに限定されるものではなく,また,抗体の動物種にも特に制限はない。 In the present invention, the term "antibody" primarily refers to human antibodies, mouse antibodies, humanized antibodies, antibodies derived from camelids (including alpacas), chimeric antibodies between human antibodies and antibodies from other mammals, and chimeric antibodies between mouse antibodies and antibodies from other mammals, but is not limited to these as long as it has the property of specifically binding to a specific antigen, and there is also no particular restriction on the animal species of the antibody.
 本発明において,「ヒト抗体」の語は,その蛋白質全体がヒト由来の遺伝子にコードされている抗体をいう。但し,遺伝子の発現効率を上昇させる等の目的で,元のヒトの遺伝子に,元のアミノ酸配列に変化を与えることなく変異を加えた遺伝子にコードされる抗体も,「ヒト抗体」に含まれる。また,ヒト抗体をコードする2つ以上の遺伝子を組み合わせて,あるヒト抗体の一部を他のヒト抗体の一部に置き換えて作製した抗体も,「ヒト抗体」である。ヒト抗体は,軽鎖の3箇所の相補性決定領域(CDR)と重鎖の3箇所の相補性決定領域(CDR)を有する。軽鎖の3箇所のCDRは,N末端側にあるものから順にCDR1,CDR2及びCDR3という。重鎖の3箇所のCDRも,N末端側にあるものから順にCDR1,CDR2及びCDR3という。あるヒト抗体のCDRを,その他のヒト抗体のCDRに置き換えることにより,ヒト抗体の抗原特異性,親和性等を改変した抗体も,ヒト抗体に含まれる。 In the present invention, the term "human antibody" refers to an antibody whose entire protein is encoded by a gene of human origin. However, "human antibodies" also include antibodies encoded by genes in which mutations have been added to the original human gene without changing the original amino acid sequence for the purpose of increasing gene expression efficiency, etc. In addition, antibodies produced by combining two or more genes encoding human antibodies and replacing a part of a human antibody with a part of another human antibody are also "human antibodies". Human antibodies have three complementarity determining regions (CDRs) in the light chain and three complementarity determining regions (CDRs) in the heavy chain. The three CDRs in the light chain are called CDR1, CDR2, and CDR3, starting from the N-terminus. The three CDRs in the heavy chain are also called CDR1, CDR2, and CDR3, starting from the N-terminus. Antibodies in which the antigen specificity, affinity, etc. of a human antibody have been modified by replacing the CDR of a certain human antibody with the CDR of another human antibody are also included in human antibodies.
 本発明において,元のヒト抗体の遺伝子を改変することにより,元の抗体のアミノ酸配列に置換,欠失,付加等の変異を加えた抗体も,「ヒト抗体」に含まれる。元の抗体のアミノ酸配列中のアミノ酸を他のアミノ酸で置換する場合,置換するアミノ酸の個数は,好ましくは1~20個,より好ましくは1~5個,更に好ましくは1~3個である。元の抗体のアミノ酸配列中のアミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1~20個,より好ましくは1~5個,更に好ましくは1~3個である。また,これらアミノ酸の置換と欠失を組み合わせた変異を加えた抗体も,ヒト抗体である。アミノ酸を付加する場合,元の抗体のアミノ酸配列中若しくはN末端又はC末端に,好ましくは1~20個,より好ましくは1~5個,更に好ましくは1~3個のアミノ酸を付加する。これらアミノ酸の付加,置換及び欠失を組み合わせた変異を加えた抗体も,ヒト抗体である。変異を加えた抗体のアミノ酸配列は,元の抗体のアミノ酸配列と,好ましくは80%以上の同一性を示し,より好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更により好ましくは98%以上の同一性を示す。ヒト抗体に上記の変異を加える場合,当該変異は抗体の可変領域に加えることができる。抗体の可変領域に上記の変異を加える場合,当該変異は可変領域のCDRとフレームワーク領域の何れに加えてもよいが,特にフレームワーク領域に加えられる。即ち,「ヒト由来の遺伝子」というときは,ヒト由来の元の遺伝子に加えて,これに改変を加えることにより得られる遺伝子も含まれる。 In the present invention, the term "human antibody" also includes antibodies in which mutations such as substitution, deletion, and addition have been added to the amino acid sequence of the original antibody by modifying the gene of the original human antibody. When an amino acid in the amino acid sequence of the original antibody is replaced with another amino acid, the number of amino acids to be replaced is preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3. When an amino acid in the amino acid sequence of the original antibody is deleted, the number of amino acids to be deleted is preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3. Furthermore, antibodies in which mutations that combine these amino acid substitutions and deletions have been added are also human antibodies. When amino acids are added, preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3 amino acids are added to the amino acid sequence or the N-terminus or C-terminus of the original antibody. Antibodies in which mutations that combine these amino acid additions, substitutions, and deletions have been added are also human antibodies. The amino acid sequence of the mutated antibody preferably exhibits 80% or more identity to the amino acid sequence of the original antibody, more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity. When the above mutations are added to a human antibody, the mutations can be added to the variable region of the antibody. When the above mutations are added to the variable region of the antibody, the mutations can be added to either the CDR or framework region of the variable region, but are particularly added to the framework region. In other words, the term "human-derived gene" includes not only the original human-derived gene, but also genes obtained by modifying it.
 本発明において,「ヒト化抗体」の語は,可変領域の一部(例えば,特にCDRの全部又は一部)のアミノ酸配列がヒト以外の哺乳動物由来であり,それ以外の領域がヒト由来である抗体のことをいう。例えば,ヒト化抗体として,ヒト抗体を構成する軽鎖の3箇所の相補性決定領域(CDR)と重鎖の3箇所の相補性決定領域(CDR)を,他の哺乳動物のCDRに置き換えることによって作製された抗体が挙げられる。ヒト抗体の適切な位置に移植されるCDRの由来となる他の哺乳動物の生物種は,ヒト以外の哺乳動物である限り特に限定はないが,好ましくは,マウス,ラット,ウサギ,ウマ,又はヒト以外の霊長類であり,より好ましくはマウス及びラットであり,更に好ましくはマウスである。また,元のヒト化抗体のアミノ酸配列に上記のヒト抗体に加えることのできる変異と同様の変異を加えた抗体も,「ヒト化抗体」に含まれる。 In the present invention, the term "humanized antibody" refers to an antibody in which the amino acid sequence of a part of the variable region (e.g., all or part of the CDR in particular) is derived from a mammal other than human, and the other regions are derived from humans. For example, a humanized antibody can be an antibody produced by replacing the three complementarity determining regions (CDRs) of the light chain and the three complementarity determining regions (CDRs) of the heavy chain that constitute a human antibody with the CDRs of another mammal. The species of the other mammal from which the CDRs are grafted to the appropriate positions of the human antibody are derived is not particularly limited as long as it is a mammal other than human, but is preferably a mouse, rat, rabbit, horse, or a non-human primate, more preferably a mouse or rat, and even more preferably a mouse. In addition, an antibody in which the amino acid sequence of the original humanized antibody is modified with the same mutations as those that can be added to the above-mentioned human antibody is also included in the "humanized antibody".
 本発明において,「キメラ抗体」の語は,2つ以上の異なる種に由来する,2つ以上の異なる抗体の断片が連結されてなる抗体のことをいう。 In the present invention, the term "chimeric antibody" refers to an antibody that is formed by linking fragments of two or more different antibodies derived from two or more different species.
 ヒト抗体と他の哺乳動物の抗体とのキメラ抗体とは,ヒト抗体の一部がヒト以外の哺乳動物の抗体の一部によって置き換えられた抗体である。抗体は,以下に説明するFc領域,Fab領域及びヒンジ部とからなる。このようなキメラ抗体の具体例として,Fc領域がヒト抗体に由来する一方でFab領域が他の哺乳動物の抗体に由来するキメラ抗体が挙げられる。ヒンジ部は,ヒト抗体又は他の哺乳動物の抗体のいずれかに由来する。逆に,Fc領域が他の哺乳動物に由来する一方でFab領域がヒト抗体に由来するキメラ抗体が挙げられる。ヒンジ部は,ヒト抗体又は他の哺乳動物の抗体のいずれかに由来する。 A chimeric antibody between a human antibody and an antibody of another mammal is an antibody in which a part of a human antibody is replaced by a part of an antibody of a mammal other than human. The antibody consists of an Fc region, a Fab region, and a hinge region, which are explained below. A specific example of such a chimeric antibody is a chimeric antibody in which the Fc region is derived from a human antibody while the Fab region is derived from an antibody of another mammal. The hinge region is derived from either a human antibody or an antibody of another mammal. Conversely, a chimeric antibody in which the Fc region is derived from another mammal while the Fab region is derived from a human antibody is included. The hinge region is derived from either a human antibody or an antibody of another mammal.
 また,抗体は,可変領域と定常領域とからなるということもできる。キメラ抗体の他の具体例として,重鎖の定常領域(C)と軽鎖の定常領域(C)がヒト抗体に由来する一方で,重鎖の可変領域(V)及び軽鎖の可変領域(V)が他の哺乳動物の抗体に由来するもの,逆に,重鎖の定常領域(C)と軽鎖の定常領域(C)が他の哺乳動物の抗体に由来する一方で,重鎖の可変領域(V)及び軽鎖の可変領域(V)がヒト抗体に由来するものも挙げられる。ここで,他の哺乳動物の生物種は,ヒト以外の哺乳動物である限り特に限定はないが,好ましくは,マウス,ラット,ウサギ,ウマ,又はヒト以外の霊長類であり,例えばマウスである。 An antibody can also be said to be composed of a variable region and a constant region. Other specific examples of chimeric antibodies include those in which the heavy chain constant region ( CH ) and the light chain constant region ( CL ) are derived from a human antibody, while the heavy chain variable region ( VH ) and the light chain variable region ( VL ) are derived from an antibody of another mammal, and conversely, those in which the heavy chain constant region ( CH ) and the light chain constant region ( CL ) are derived from an antibody of another mammal, while the heavy chain variable region ( VH ) and the light chain variable region ( VL ) are derived from a human antibody. Here, the species of the other mammal is not particularly limited as long as it is a mammal other than human, but is preferably a mouse, rat, rabbit, horse, or a non-human primate, such as a mouse.
 本発明の一実施形態における抗体は,2本の免疫グロブリン軽鎖(又は単に「軽鎖」)と2本の免疫グロブリン重鎖(又は単に「重鎖」)の計4本のポリペプチド鎖からなる基本構造を有する。但し,「抗体」というときは,この基本構造を有するものに加え,
 (1)1本の軽鎖と1本の重鎖の計2本のポリペプチド鎖からなるもの,
 (2)本来の意味での抗体の基本構造からFc領域が欠失したものであるFab領域からなるもの及びFab領域とヒンジ部の全部若しくは一部とからなるもの(Fab,F(ab’)及びF(ab’)を含む),
 (3)軽鎖のC末端側にリンカーを,そして更にそのC末端側に重鎖を結合させてなるものである一本鎖抗体,
 (4)重鎖のC末端側にリンカーを,そして更にそのC末端側に軽鎖を結合させてなるものである一本鎖抗体,
 (5)本来の意味での抗体の基本構造からFab領域が欠失したものであるFc領域からなるものであって,且つ当該Fc領域のアミノ酸配列が特定の抗原に特異的に結合する性質を有するように改変されたもの(Fc抗体),
 (6)後述する単一ドメイン抗体も,「抗体」に含まれる。
In one embodiment of the present invention, an antibody has a basic structure consisting of a total of four polypeptide chains: two immunoglobulin light chains (or simply "light chains") and two immunoglobulin heavy chains (or simply "heavy chains"). However, when referring to an "antibody", in addition to those having this basic structure,
(1) Consists of two polypeptide chains, one light chain and one heavy chain,
(2) Those consisting of a Fab region in which the Fc region has been deleted from the basic structure of an antibody in the original sense, and those consisting of a Fab region and all or part of the hinge region (including Fab, F(ab') and F(ab') 2 ),
(3) A single-chain antibody, which is composed of a light chain with a linker at its C-terminus and a heavy chain at its C-terminus.
(4) A single-chain antibody, which is composed of a heavy chain with a linker at its C-terminus and a light chain at its C-terminus.
(5) An antibody that is composed of an Fc region in which the Fab region has been deleted from the basic structure of the original antibody, and the amino acid sequence of the Fc region has been modified so as to have the property of specifically binding to a specific antigen (Fc antibody).
(6) Single domain antibodies, described below, are also included in the term "antibody."
 本発明の一実施形態における抗体は,ラクダ科動物(アルパカを含む)由来の抗体である。ラクダ科動物の抗体には,ジスルフィド結合により連結された2本の重鎖からなるものがある。この2本の重鎖からなる抗体を重鎖抗体という。VHHは,重鎖抗体を構成する重鎖の可変領域を含む1本の重鎖からなる抗体,又は重鎖抗体を構成する定常領域(CH)を欠く1本の重鎖からなる抗体である。VHHも本発明の実施形態における抗体の一つである。その他,ジスルフィド結合により連結された2本の軽鎖からなる抗体も本発明の実施形態における抗体の一つである。この2本の軽鎖からなる抗体を軽鎖抗体という。ラクダ科動物由来の抗体(VHHを含む)をヒトに投与したときの抗原性を低減させるために,ラクダ科動物の抗体のアミノ酸配列に変異を加えたものも,本発明の一実施形態における抗体である。ラクダ科動物の抗体のアミノ酸に変異を加える場合,本明細書に記載の抗体に加えることのできる変異と同様の変異を加えることができる。 The antibody in one embodiment of the present invention is an antibody derived from a camelid (including an alpaca). Some camelid antibodies are composed of two heavy chains linked by a disulfide bond. An antibody composed of two heavy chains is called a heavy chain antibody. A VHH is an antibody composed of a single heavy chain including the variable region of the heavy chain constituting a heavy chain antibody, or an antibody composed of a single heavy chain lacking the constant region (CH) constituting a heavy chain antibody. A VHH is also one of the antibodies in an embodiment of the present invention. Another antibody in an embodiment of the present invention is an antibody composed of two light chains linked by a disulfide bond. An antibody composed of two light chains is called a light chain antibody. In order to reduce the antigenicity of an antibody derived from a camelid (including a VHH) when administered to a human, an antibody in one embodiment of the present invention is also an antibody in which a mutation has been added to the amino acid sequence of an antibody of a camelid. When a mutation is added to the amino acid of an antibody of a camelid, the same mutation as that which can be added to an antibody described in this specification can be added.
 本発明の一実施形態における抗体は,サメ由来の抗体である。サメの抗体は,ジスルフィド結合により連結された2本の重鎖からなる。この2本の重鎖からなる抗体を重鎖抗体という。VNARは,重鎖抗体を構成する重鎖の可変領域を含む1本の重鎖からなる抗体,又は重鎖抗体を構成する定常領域(CH)を欠く1本の重鎖からなる抗体である。VNARも本発明の実施形態における抗体の一つである。サメ由来の抗体(VNARを含む)をヒトに投与したときの抗原性を低減させるために,サメの抗体のアミノ酸配列に変異を加えたものも,本発明の一実施形態における抗体である。サメの抗体のアミノ酸に変異を加える場合,本明細書に記載の抗体に加えることのできる変異と同様の変異加えることができる。サメの抗体をヒト化したものも本発明の実施形態における抗体の一つである。 The antibody in one embodiment of the present invention is a shark-derived antibody. A shark antibody consists of two heavy chains linked by a disulfide bond. An antibody consisting of these two heavy chains is called a heavy chain antibody. A VNAR is an antibody consisting of a single heavy chain that includes the variable region of the heavy chain that constitutes a heavy chain antibody, or an antibody consisting of a single heavy chain that lacks the constant region (CH) that constitutes a heavy chain antibody. VNAR is also one of the antibodies in an embodiment of the present invention. In order to reduce the antigenicity of a shark-derived antibody (including VNAR) when administered to a human, an antibody in one embodiment of the present invention is also an antibody in one embodiment of the present invention in which mutations have been added to the amino acid sequence of the shark antibody. When mutations are added to the amino acids of a shark antibody, the same mutations that can be added to the antibodies described in this specification can be added. A humanized shark antibody is also one of the antibodies in one embodiment of the present invention.
 2本の軽鎖と2本の重鎖の計4本のポリペプチド鎖からなる基本構造を有する抗体は,軽鎖の可変領域(V)に3箇所の相補性決定領域(CDR)と重鎖の可変領域(V)に3箇所の相補性決定領域(CDR)を有する。軽鎖の3箇所のCDRは,N末端側にあるものから順にCDR1,CDR2及びCDR3という。重鎖の3箇所のCDRも,N末端側にあるものから順にCDR1,CDR2及びCDR3という。ただし,これらCDRの一部又は全部が不完全であるか,又は存在しないものであっても,特定の抗原に特異的に結合する性質を有するものである限り,抗体に含まれる。軽鎖及び重鎖の可変領域(V及びV)のCDR以外の領域は,フレームワーク領域(FR)という。FRは,N末端側にあるものから順にFR1,FR2,FR3及びFR4という。通常,CDRとFRはN末端側から順に,FR1,CDR1,FR2,CDR2,FR3,CDR3,FR4の順で存在する。 An antibody has a basic structure consisting of four polypeptide chains, two light chains and two heavy chains, and has three complementarity determining regions (CDRs) in the variable region of the light chain (V L ) and three complementarity determining regions (CDRs) in the variable region of the heavy chain (V H ). The three CDRs in the light chain are called CDR1, CDR2, and CDR3, starting from the N-terminus. The three CDRs in the heavy chain are also called CDR1, CDR2, and CDR3, starting from the N-terminus. However, even if some or all of these CDRs are incomplete or absent, they are included in the antibody as long as they have the property of specifically binding to a specific antigen. The regions other than the CDRs in the variable regions of the light and heavy chains (V L and V H ) are called framework regions (FR). The FRs are called FR1, FR2, FR3, and FR4, starting from the N-terminus. Typically, CDRs and FRs are present in the following order from the N-terminus: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
 本発明の一実施形態において,元の抗体のアミノ酸配列に置換,欠失,付加等の変異を加えた抗体も,抗体に含まれる。元の抗体のアミノ酸配列中のアミノ酸を他のアミノ酸で置換する場合,置換するアミノ酸の個数は,好ましくは1~20個,より好ましくは1~5個,更に好ましくは1~3個である。元の抗体のアミノ酸配列中のアミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1~20個,より好ましくは1~5個,更に好ましくは1~3個である。また,これらアミノ酸の置換と欠失を組み合わせた変異を加えたものも,抗体である。アミノ酸を付加する場合,元の抗体のアミノ酸配列中若しくはN末端又はC末端に,好ましくは1~20個,より好ましくは1~5個,更に好ましくは1~3個のアミノ酸を付加する。これらアミノ酸の付加,置換及び欠失を組み合わせた変異を加えたものも,抗体である。変異を加えた抗体のアミノ酸配列は,元の抗体のアミノ酸配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは,90%以上,95%以上,又は98%以上の同一性を示す。 In one embodiment of the present invention, the antibody also includes an antibody in which a mutation such as substitution, deletion, or addition has been added to the amino acid sequence of the original antibody. When an amino acid in the amino acid sequence of the original antibody is replaced with another amino acid, the number of amino acids to be replaced is preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3. When an amino acid in the amino acid sequence of the original antibody is deleted, the number of amino acids to be deleted is preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3. In addition, an antibody in which a mutation combining these amino acid substitutions and deletions has been added is also an antibody. When an amino acid is added, preferably 1 to 20, more preferably 1 to 5, and even more preferably 1 to 3 amino acids are added to the amino acid sequence or the N-terminus or C-terminus of the original antibody. An antibody in which a mutation has been added combining these amino acid additions, substitutions, and deletions is also an antibody. The amino acid sequence of the mutated antibody preferably exhibits 80% or more identity with the amino acid sequence of the original antibody, more preferably 85% or more identity, and even more preferably 90% or more, 95% or more, or 98% or more identity.
 本発明の一実施形態において,元の抗体の可変領域のアミノ酸配列に置換,欠失,付加等の変異を加えた抗体も,抗体に含まれる。元の抗体のアミノ酸配列中のアミノ酸を他のアミノ酸で置換する場合,置換するアミノ酸の個数は,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個である。元の抗体のアミノ酸配列中のアミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個である。また,これらアミノ酸の置換と欠失を組み合わせた変異を加えたものも,抗体である。アミノ酸を付加する場合,元の抗体のアミノ酸配列中若しくはN末端又はC末端に,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個のアミノ酸を付加する。これらアミノ酸の付加,置換及び欠失を組み合わせた変異を加えたものも,抗体である。変異を加えた抗体のアミノ酸配列は,元の抗体のアミノ酸配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは,90%以上,95%以上,又は98%以上の同一性を示す。抗体の可変領域に変異を加える場合,当該変異は可変領域のCDRとフレームワーク領域の何れに加えてもよいが,特にフレームワーク領域に加えられる。 In one embodiment of the present invention, the antibody also includes an antibody in which a mutation such as substitution, deletion, or addition has been added to the amino acid sequence of the variable region of the original antibody. When an amino acid in the amino acid sequence of the original antibody is replaced with another amino acid, the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3. When an amino acid in the amino acid sequence of the original antibody is deleted, the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3. In addition, an antibody in which a mutation that combines the substitution and deletion of these amino acids has been added is also an antibody. When an amino acid is added, preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3 amino acids are added to the amino acid sequence or the N-terminus or C-terminus of the original antibody. An antibody in which a mutation has been added is also an antibody in which a mutation that combines the addition, substitution, and deletion of these amino acids has been added is also an antibody. The amino acid sequence of the antibody in which the mutation has been added is preferably 80% or more identical to the amino acid sequence of the original antibody, more preferably 85% or more identical, and even more preferably 90% or more, 95% or more, or 98% or more identical. When mutations are made to the variable region of an antibody, the mutations may be made in either the CDR or framework region of the variable region, but are particularly made in the framework region.
 本発明の一実施形態において,元の抗体の可変領域のフレームワーク領域にアミノ酸配列に置換,欠失,付加等の変異を加えた抗体も,抗体に含まれる。元の抗体のアミノ酸配列中のアミノ酸を他のアミノ酸で置換する場合,置換するアミノ酸の個数は,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個である。元の抗体のアミノ酸配列中のアミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個である。また,これらアミノ酸の置換と欠失を組み合わせた変異を加えたものも,抗体である。アミノ酸を付加する場合,元の抗体のアミノ酸配列中若しくはN末端又はC末端に,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個のアミノ酸を付加する。これらアミノ酸の付加,置換及び欠失を組み合わせた変異を加えたものも,抗体である。変異を加えた抗体のアミノ酸配列は,元の抗体のアミノ酸配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは,90%以上,95%以上,又は98%以上の同一性を示す。 In one embodiment of the present invention, the antibody also includes an antibody in which a mutation such as substitution, deletion, or addition has been added to the amino acid sequence of the framework region of the variable region of the original antibody. When an amino acid in the amino acid sequence of the original antibody is replaced with another amino acid, the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3. When an amino acid in the amino acid sequence of the original antibody is deleted, the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3. In addition, an antibody in which a mutation that combines these amino acid substitutions and deletions has been added is also an antibody. When an amino acid is added, preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3 amino acids are added to the amino acid sequence or the N-terminus or C-terminus of the original antibody. An antibody in which a mutation that combines these amino acid additions, substitutions, and deletions has been added is also an antibody. The amino acid sequence of the mutated antibody preferably exhibits 80% or more identity to the amino acid sequence of the original antibody, more preferably 85% or more identity, and even more preferably 90% or more, 95% or more, or 98% or more identity.
 本発明の一実施形態において,元の抗体の可変領域のCDR領域にアミノ酸配列に置換,欠失,付加等の変異を加えた抗体も,抗体に含まれる。元の抗体のアミノ酸配列中のアミノ酸を他のアミノ酸で置換する場合,置換するアミノ酸の個数は,好ましくは1~5個,より好ましくは1~3個,更に好ましくは1又は2個である。元の抗体のアミノ酸配列中のアミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1~5個,より好ましくは1~3個,更に好ましくは1又は2個である。また,これらアミノ酸の置換と欠失を組み合わせた変異を加えたものも,抗体である。アミノ酸を付加する場合,元の抗体のアミノ酸配列中若しくはN末端又はC末端に,好ましくは1~5個,よりに好ましくは1~3個,更に好ましくは1又は2個のアミノ酸を付加する。これらアミノ酸の付加,置換及び欠失を組み合わせた変異を加えたものも,抗体である。変異を加えた抗体のアミノ酸配列は,元の抗体のアミノ酸配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは,90%以上,95%以上,又は98%以上の同一性を示す。 In one embodiment of the present invention, the antibody also includes an antibody in which a mutation such as substitution, deletion, or addition has been added to the amino acid sequence in the CDR region of the variable region of the original antibody. When an amino acid in the amino acid sequence of the original antibody is replaced with another amino acid, the number of amino acids to be replaced is preferably 1 to 5, more preferably 1 to 3, and even more preferably 1 or 2. When an amino acid in the amino acid sequence of the original antibody is deleted, the number of amino acids to be deleted is preferably 1 to 5, more preferably 1 to 3, and even more preferably 1 or 2. In addition, an antibody in which a mutation combining these amino acid substitutions and deletions has been added is also an antibody. When an amino acid is added, preferably 1 to 5, more preferably 1 to 3, and even more preferably 1 or 2 amino acids are added to the amino acid sequence or the N-terminus or C-terminus of the original antibody. An antibody in which a mutation combining these amino acid additions, substitutions, and deletions has been added is also an antibody. The amino acid sequence of the mutated antibody preferably exhibits 80% or more identity to the amino acid sequence of the original antibody, more preferably 85% or more identity, and even more preferably 90% or more, 95% or more, or 98% or more identity.
 本発明の一実施形態において,Fabとは,可変領域とC領域(軽鎖の定常領域)を含む1本の軽鎖と,可変領域とC1領域(重鎖の定常領域の部分1)を含む1本の重鎖が,それぞれに存在するシステイン残基同士でジスルフィド結合により結合した分子のことをいう。Fabにおいて,重鎖は,可変領域とC1領域(重鎖の定常領域の部分1)に加えて,更にヒンジ部の一部を含んでもよいが,この場合のヒンジ部は,ヒンジ部に存在して抗体の重鎖どうしを結合するシステイン残基を欠くものである。Fabにおいて,軽鎖と重鎖とは,軽鎖の定常領域(C領域)に存在するシステイン残基と,重鎖の定常領域(C1領域)又はヒンジ部に存在するシステイン残基との間で形成されるジスルフィド結合により結合する。Fabを形成する重鎖のことをFab重鎖という。Fabは,ヒンジ部に存在して抗体の重鎖どうしを結合するシステイン残基を欠いているので,1本の軽鎖と1本の重鎖とからなる。Fabを構成する軽鎖は,可変領域とC領域を含む。Fabを構成する重鎖は,可変領域とC1領域からなるものであってもよく,可変領域,C1領域に加えてヒンジ部の一部を含むものであってもよい。但しこの場合,ヒンジ部で2本の重鎖の間でジスルフィド結合が形成されないように,ヒンジ部は重鎖間を結合するシステイン残基を含まないように選択される。F(ab’)においては,その重鎖は可変領域とC1領域に加えて,重鎖どうしを結合するシステイン残基を含むヒンジ部の全部又は一部を含む。F(ab’)2は2つのF(ab’)が互いのヒンジ部に存在するシステイン残基どうしでジスルフィド結合により結合した分子のことをいう。F(ab’)又はF(ab’)2を形成する重鎖のことをFab’重鎖という。また,複数の抗体が直接又はリンカーを介して結合してなるニ量体,三量体等の重合体も,抗体である。更に,これらに限らず,抗体分子の一部を含み,且つ,抗原に特異的に結合する性質を有するものは何れも,本発明でいう「抗体」に含まれる。即ち,軽鎖というときは,軽鎖に由来し,その可変領域の全て又は一部のアミノ酸配列を有するものが含まれる。また,重鎖というときは,重鎖に由来し,その可変領域の全て又は一部のアミノ酸配列を有するものが含まれる。従って,可変領域の全て又は一部のアミノ酸配列を有する限り,例えば,Fc領域が欠失したものも,重鎖である。 In one embodiment of the present invention, Fab refers to a molecule in which one light chain including a variable region and a CL region (light chain constant region) and one heavy chain including a variable region and a CH1 region (part 1 of the heavy chain constant region) are bound by disulfide bonds between cysteine residues present in each. In Fab, the heavy chain may further include a part of the hinge region in addition to the variable region and the CH1 region (part 1 of the heavy chain constant region), but in this case, the hinge region lacks cysteine residues present in the hinge region that bind the heavy chains of the antibody. In Fab, the light chain and the heavy chain are bound by disulfide bonds formed between cysteine residues present in the light chain constant region ( CL region) and cysteine residues present in the heavy chain constant region ( CH1 region) or hinge region. The heavy chain that forms Fab is called a Fab heavy chain. Fab lacks the cysteine residues present in the hinge region that bind the heavy chains of an antibody, and therefore consists of one light chain and one heavy chain. The light chain that constitutes Fab contains a variable region and a CL region. The heavy chain that constitutes Fab may be composed of a variable region and a CH1 region, or may contain a part of the hinge region in addition to the variable region and the CH1 region. In this case, however, the hinge region is selected so as not to contain a cysteine residue that binds the heavy chains, so that a disulfide bond is not formed between the two heavy chains at the hinge region. In F(ab'), the heavy chain contains the whole or part of the hinge region that contains the cysteine residue that binds the heavy chains, in addition to the variable region and the CH1 region. F(ab') 2 refers to a molecule in which two F(ab') are bound by disulfide bonds between the cysteine residues present in the hinge regions of each other. The heavy chain that forms F(ab') or F(ab') 2 is called a Fab' heavy chain. In addition, polymers such as dimers and trimers formed by binding multiple antibodies directly or via a linker are also antibodies. Furthermore, not limited to these, any one that contains a part of an antibody molecule and has the property of specifically binding to an antigen is included in the "antibody" as used in the present invention. That is, the term "light chain" includes one that is derived from a light chain and has all or part of the amino acid sequence of its variable region. In addition, the term "heavy chain" includes one that is derived from a heavy chain and has all or part of the amino acid sequence of its variable region. Therefore, as long as it has all or part of the amino acid sequence of the variable region, for example, one that has deleted the Fc region is also a heavy chain.
 また,ここでFc又はFc領域とは,抗体分子中の,C2領域(重鎖の定常領域の部分2),及びC3領域(重鎖の定常領域の部分3)からなる断片を含む領域のことをいう。 Furthermore, Fc or Fc region herein refers to a region in an antibody molecule that includes a fragment consisting of the CH2 region (part 2 of the heavy chain constant region) and the CH3 region (part 3 of the heavy chain constant region).
 更には,本発明の一実施形態における抗体は,
 (7)上記(2)で示したFab,F(ab’)又はF(ab’)2を構成する軽鎖と重鎖を,リンカーを介して結合させて,それぞれ一本鎖抗体としたscFab,scF(ab’),及びscF(ab’)2も含まれる。ここで,scFab,scF(ab’),及びscF(ab’)2にあっては,軽鎖のC末端側にリンカーを,そして更にそのC末端側に重鎖を結合させてなるものでもよく,また,重鎖のC末端側にリンカーを,そして更にそのC末端側に軽鎖を結合させてなるものでもよい。更には,軽鎖の可変領域と重鎖の可変領域をリンカーを介して結合させて一本鎖抗体としたscFvも,抗体に含まれる。scFvにあっては,軽鎖の可変領域のC末端側にリンカーを,そして更にそのC末端側に重鎖の可変領域を結合させてなるものでもよく,また,重鎖の可変領域のC末端側にリンカーを,そして更にそのC末端側に軽鎖の可変領域を結合させてなるものでもよい。
Furthermore, the antibody in one embodiment of the present invention comprises:
(7) Also included are scFab, scF(ab'), and scF(ab') 2 , which are single-chain antibodies formed by linking the light chain and the heavy chain constituting Fab, F(ab'), or F(ab') 2 shown in (2) above via a linker. Here, scFab, scF(ab'), and scF(ab') 2 may be formed by linking a linker to the C-terminus of the light chain and further linking a heavy chain to the C-terminus, or may be formed by linking a linker to the C-terminus of the heavy chain and further linking a light chain to the C-terminus. Furthermore, antibodies also include scFv, which is a single-chain antibody formed by linking a light chain variable region and a heavy chain variable region via a linker. For scFv, it may be formed by linking a linker to the C-terminus of the light chain variable region and further linking a heavy chain variable region to the C-terminus, or may be formed by linking a linker to the C-terminus of the heavy chain variable region and further linking a light chain variable region to the C-terminus.
 更には,本明細書でいう「抗体」には,完全長抗体,上記(1)~(7)に示されるものに加えて,(1)~(7)を含むより広い概念である,完全長抗体の一部が欠損したものである抗原結合性断片(抗体フラグメント)のいずれの形態も含まれる。抗原結合性断片には,重鎖抗体,軽鎖抗体,VHH,VNAR,及びこれらの一部が欠損したものも含まれる。 Furthermore, the term "antibody" as used herein includes full-length antibodies, as well as those listed in (1) to (7) above, and also includes antigen-binding fragments (antibody fragments) in which a portion of a full-length antibody is deleted, which is a broader concept including (1) to (7). Antigen-binding fragments include heavy chain antibodies, light chain antibodies, VHHs, VNARs, and those in which a portion of these antibodies is deleted.
 「抗原結合性断片」の語は,抗原との特異的結合活性の少なくとも一部を保持している抗体の断片のことをいう。結合性断片の例としては,Fab,Fab’,F(ab’)2,可変領域(Fv),重鎖可変領域(V)と軽鎖可変領域(V)とを適当なリンカーで連結させた一本鎖抗体(scFv),重鎖可変領域(V)と軽鎖可変領域(V)を含むポリペプチドの二量体であるダイアボディ,scFvの重鎖(H鎖)に定常領域の一部(C3)が結合したものの二量体であるミニボディ,その他の低分子化抗体等を包含する。但し,抗原との結合能を有している限りこれらの分子に限定されない。 The term "antigen-binding fragment" refers to a fragment of an antibody that retains at least a part of the specific binding activity with an antigen. Examples of binding fragments include Fab, Fab', F(ab') 2 , variable region (Fv), single-chain antibody (scFv) in which the heavy chain variable region ( VH ) and the light chain variable region ( VL ) are linked with an appropriate linker, diabody, which is a dimer of a polypeptide containing a heavy chain variable region ( VH ) and a light chain variable region ( VL ), minibody, which is a dimer of a scFv heavy chain (H chain) bound to a part of the constant region ( CH3 ), other low molecular weight antibodies, etc. However, it is not limited to these molecules as long as it has the ability to bind to an antigen.
 本発明の一実施形態において,「一本鎖抗体」というときは,軽鎖の可変領域の全て又は一部を含むアミノ酸配列のC末端側にリンカーが結合し,更にそのC末端側に重鎖の可変領域の全て又は一部を含むアミノ酸配列が結合してなり,特定の抗原に特異的に結合することのできる蛋白質をいう。また,重鎖の可変領域の全て又は一部を含むアミノ酸配列のC末端側にリンカーが結合し,更にそのC末端側に軽鎖の可変領域の全て又は一部を含むアミノ酸配列が結合してなり,特定の抗原に特異的に結合することのできる蛋白質も,「一本鎖抗体」である。重鎖のC末端側にリンカーを介して軽鎖が結合した一本鎖抗体にあっては,通常,重鎖は,Fc領域が欠失している。軽鎖の可変領域は,抗体の抗原特異性に関与する相補性決定領域(CDR)を3つ有している。同様に,重鎖の可変領域も,CDRを3つ有している。これらのCDRは,抗体の抗原特異性を決定する主たる領域である。従って,一本鎖抗体には,重鎖の3つのCDRが全てと,軽鎖の3つのCDRの全てとが含まれることが好ましい。但し,抗体の抗原特異的な親和性が維持される限り,CDRの1個又は複数個を欠失させた一本鎖抗体とすることもできる。 In one embodiment of the present invention, the term "single-chain antibody" refers to a protein that can specifically bind to a specific antigen, which is composed of an amino acid sequence containing all or part of the variable region of a light chain, to which a linker is attached at the C-terminus, and to which an amino acid sequence containing all or part of the variable region of a heavy chain is further attached at the C-terminus. Also, a protein that can specifically bind to a specific antigen, which is composed of an amino acid sequence containing all or part of the variable region of a heavy chain, to which a linker is attached at the C-terminus, and to which an amino acid sequence containing all or part of the variable region of a light chain is further attached at the C-terminus, is also a "single-chain antibody". In a single-chain antibody in which a light chain is attached to the C-terminus of a heavy chain via a linker, the heavy chain usually lacks an Fc region. The variable region of the light chain has three complementarity determining regions (CDRs) that are involved in the antigen specificity of the antibody. Similarly, the variable region of the heavy chain also has three CDRs. These CDRs are the main regions that determine the antigen specificity of the antibody. Therefore, it is preferred that a single chain antibody contains all three CDRs of the heavy chain and all three CDRs of the light chain. However, a single chain antibody can also be one in which one or more CDRs are deleted, so long as the antigen-specific affinity of the antibody is maintained.
 一本鎖抗体において,抗体の軽鎖と重鎖の間に配置されるリンカーは,好ましくは2~50個,より好ましくは8~50個,更に好ましくは10~30個,更により好ましくは12~18個又は15~25個,例えば15個若しくは25個のアミノ酸残基から構成されるペプチド鎖である。そのようなリンカーは,これにより両鎖が連結されてなる抗hTfR抗体がhTfRに対する親和性を保持する限り,そのアミノ酸配列に限定はないが,好ましくは,グリシンのみ又はグリシンとセリンから構成されるものであり,例えば,アミノ酸配列Gly-Ser,アミノ酸配列Gly-Gly-Ser,アミノ酸配列Gly-Gly-Gly,アミノ酸配列Gly-Gly-Gly-Gly-Ser(配列番号9),アミノ酸配列Gly-Gly-Gly-Gly-Gly-Ser(配列番号10),アミノ酸配列Ser-Gly-Gly-Gly-Gly(配列番号11),又はこれらのアミノ酸配列が2~10回,あるいは2~5回繰り返された配列を含むものである。例えば,重鎖の可変領域の全領域からなるアミノ酸配列のC末端側に,リンカーを介して軽鎖の可変領域を結合させる場合,アミノ酸配列Gly-Gly-Gly-Gly-Ser(配列番号9)の3個が連続したものに相当する計15個のアミノ酸を含むリンカーが好適である。 In a single-chain antibody, the linker disposed between the light and heavy chains of the antibody is preferably a peptide chain composed of 2 to 50, more preferably 8 to 50, even more preferably 10 to 30, even more preferably 12 to 18 or 15 to 25, for example 15 or 25 amino acid residues. There are no limitations on the amino acid sequence of such a linker, so long as the anti-hTfR antibody formed by linking both chains by the linker retains affinity for hTfR, but it is preferably composed of only glycine or glycine and serine, and includes, for example, the amino acid sequence Gly-Ser, the amino acid sequence Gly-Gly-Ser, the amino acid sequence Gly-Gly-Gly, the amino acid sequence Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 9), the amino acid sequence Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 10), the amino acid sequence Ser-Gly-Gly-Gly-Gly (SEQ ID NO: 11), or a sequence in which these amino acid sequences are repeated 2 to 10 times or 2 to 5 times. For example, when linking a light chain variable region via a linker to the C-terminus of an amino acid sequence consisting of the entire region of the heavy chain variable region, a linker containing a total of 15 amino acids corresponding to three consecutive amino acid sequences Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 9) is preferred.
 本発明の一実施形態において,単一ドメイン抗体とは,単一の可変領域で抗原に特異的に結合する性質を有する抗体のことをいう。単一ドメイン抗体には,可変領域が重鎖の可変領域のみからなる抗体(重鎖単一ドメイン抗体),可変領域が軽鎖の可変領域のみからなる抗体(軽鎖単一ドメイン抗体)が含まれる。VHH,VNARは単一ドメイン抗体の一種である。 In one embodiment of the present invention, a single domain antibody refers to an antibody that has the property of specifically binding to an antigen through a single variable region. Single domain antibodies include antibodies whose variable region consists only of the variable region of a heavy chain (heavy chain single domain antibodies) and antibodies whose variable region consists only of the variable region of a light chain (light chain single domain antibodies). VHH and VNAR are types of single domain antibodies.
 本発明の一実施形態において,「ヒトトランスフェリン受容体」の語は,配列番号22に示されるアミノ酸配列を有する膜蛋白質をいう。本発明の抗体は,その一実施態様において,配列番号22で示されるアミノ酸配列中N末端側から89番目のシステイン残基からC末端のフェニルアラニンまでの部分(トランスフェリン受容体の細胞外領域)に対して特異的に結合するものであるが,これに限定されない。 In one embodiment of the present invention, the term "human transferrin receptor" refers to a membrane protein having the amino acid sequence shown in SEQ ID NO: 22. In one embodiment, the antibody of the present invention specifically binds to the portion of the amino acid sequence shown in SEQ ID NO: 22 from the 89th cysteine residue from the N-terminus to the phenylalanine at the C-terminus (the extracellular domain of the transferrin receptor), but is not limited thereto.
 所望の蛋白質に対する抗体の作製方法としては,当該蛋白質をコードする遺伝子を組み込んだ発現ベクターを導入した細胞を用いて,組換え蛋白質を作製し,この組換え蛋白質を用いてマウス等の動物を免疫して得る方法が一般的である。免疫後の動物から組換え蛋白質に対する抗体産生細胞を取り出し,これとミエローマ細胞とを融合させることにより,組換え蛋白質に対する抗体の産生能を有するハイブリドーマ細胞を作製することができる。 The most common method for producing antibodies against a desired protein is to produce a recombinant protein using cells that have been introduced with an expression vector incorporating a gene that codes for the protein, and then immunize animals such as mice with this recombinant protein. After immunization, antibody-producing cells against the recombinant protein are extracted from the animal, and these are then fused with myeloma cells to produce hybridoma cells capable of producing antibodies against the recombinant protein.
 また,マウス等の動物より得た免疫系細胞を体外免疫法により所望の蛋白質で免疫することによっても,当該蛋白質に対する抗体を産生する細胞を取得できる。体外免疫法を用いる場合,その免疫系細胞が由来する動物種に特に限定はないが,好ましくは,マウス,ラット,ウサギ,モルモット,イヌ,ネコ,ウマ,及びヒトを含む霊長類であり,より好ましくは,マウス,ラット及びヒトであり,更に好ましくはマウス及びヒトである。マウスの免疫系細胞としては,例えば,マウスの脾臓から調製した脾細胞を用いることができる。ヒトの免疫系細胞としては,ヒトの末梢血,骨髄,脾臓等から調製した細胞を用いることができる。ヒトの免疫系細胞を体外免疫法により免疫した場合,組換え蛋白質に対するヒト抗体を得ることができる。  Also, by immunizing immune system cells obtained from an animal such as a mouse with a desired protein by in vitro immunization, cells that produce antibodies against the protein can be obtained. When using in vitro immunization, there is no particular limitation on the animal species from which the immune system cells are derived, but preferred are mice, rats, rabbits, guinea pigs, dogs, cats, horses, and primates including humans, more preferably mice, rats, and humans, and even more preferably mice and humans. For example, splenocytes prepared from mouse spleens can be used as mouse immune system cells. For human immune system cells, cells prepared from human peripheral blood, bone marrow, spleen, etc. can be used. When human immune system cells are immunized by in vitro immunization, human antibodies against the recombinant protein can be obtained.
 体外免疫法により免疫系細胞を免疫した後,細胞をミエローマ細胞と融合させることにより,抗体産生能を有するハイブリドーマ細胞を作製することができる。また,免疫後の細胞からmRNAを抽出してcDNAを合成し,このcDNAを鋳型としてPCR反応により免疫グロブリンの軽鎖及び重鎖をコードする遺伝子を含むDNA断片を増幅し,これらを用いて人工的に抗体遺伝子を再構築することもできる。  Immune system cells can be immunized by in vitro immunization, and then fused with myeloma cells to produce hybridoma cells capable of producing antibodies. It is also possible to extract mRNA from the immunized cells, synthesize cDNA, and use this cDNA as a template to amplify DNA fragments containing genes encoding the light and heavy chains of immunoglobulins through PCR reactions, which can then be used to artificially reconstruct antibody genes.
 上記方法により得られたままのハイブリドーマ細胞には,目的外の蛋白質を抗原として認識する抗体を産生する細胞も含まれる。また,所望の蛋白質に対する抗体を産生するハイブリドーマ細胞の全てが,当該蛋白質に対して高親和性を有する等の所望の特性を示す抗体を産生するとも限らない。 Hybridoma cells obtained as is by the above method also include cells that produce antibodies that recognize non-target proteins as antigens. Furthermore, not all hybridoma cells that produce antibodies against a desired protein necessarily produce antibodies that exhibit the desired properties, such as having high affinity for that protein.
 同様に,人工的に再構築した抗体遺伝子には,目的外の蛋白質を抗原として認識する抗体をコードする遺伝子も含まれる。また,所望の蛋白質に対する抗体をコードする遺伝子の全てが,当該蛋白質に対して高親和性を有する等の所望の特性を示す抗体をコードするものとも限らない。 Similarly, artificially reconstructed antibody genes also include genes that code for antibodies that recognize untargeted proteins as antigens. Furthermore, not all genes that code for antibodies against a desired protein necessarily code for antibodies that exhibit the desired properties, such as having high affinity for that protein.
 従って,上記で得られたままのハイブリドーマ細胞から,所望の特性を有する抗体を産生するハイブリドーマ細胞を選択するステップが必要となる。また,人工的に再構築した抗体遺伝子にあっては,当該抗体遺伝子から,所望の特性を有する抗体をコードする遺伝子を選択するステップが必要となる。例えば,所望の蛋白質に対して高親和性を示す抗体(高親和性抗体)を産生するハイブリドーマ細胞,又は高親和性抗体をコードする遺伝子を選択する方法として,以下に詳述する方法が有効である。 Therefore, a step is required to select hybridoma cells that produce antibodies with the desired characteristics from the hybridoma cells obtained as described above. Furthermore, in the case of artificially reconstructed antibody genes, a step is required to select genes that code for antibodies with the desired characteristics from the antibody genes. For example, the method described in detail below is effective as a method for selecting hybridoma cells that produce antibodies that show high affinity for a desired protein (high affinity antibodies), or genes that code for high affinity antibodies.
 例えば,所望の蛋白質に対して高親和性の抗体を産生するハイブリドーマ細胞を選択する場合,当該蛋白質をプレートに添加してこれに保持させた後,ハイブリドーマ細胞の培養上清を添加し,次いで当該蛋白質と結合していない抗体をプレートから除去し,プレートに保持された抗体の量を測定する方法が用いられる。この方法によれば,プレートに添加したハイブリドーマ細胞の培養上清に含まれる抗体の当該蛋白質に対する親和性が高いほど,プレートに保持される抗体の量が多くなる。従って,プレートに保持された抗体の量を測定し,より多くの抗体が保持されたプレートに対応するハイブリドーマ細胞を,当該蛋白質に対して相対的に高い親和性を有する抗体を産生する細胞株として選択することができる。この様にして選択された細胞株から,mRNAを抽出してcDNAを合成し,このcDNAを鋳型として,当該蛋白質に対する抗体をコードする遺伝子を含むDNA断片をPCR法を用いて増幅することにより,高親和性抗体をコードする遺伝子を単離することもできる。 For example, when selecting hybridoma cells that produce antibodies with high affinity to a desired protein, the protein is added to a plate and retained thereon, then the culture supernatant of the hybridoma cells is added, and antibodies that are not bound to the protein are removed from the plate, and the amount of antibody retained on the plate is measured. According to this method, the higher the affinity of the antibody contained in the culture supernatant of the hybridoma cells added to the plate for the protein, the greater the amount of antibody retained on the plate. Therefore, the amount of antibody retained on the plate can be measured, and the hybridoma cells corresponding to the plate that retain more antibodies can be selected as cell lines that produce antibodies with relatively high affinity for the protein. From the cell line selected in this way, mRNA can be extracted to synthesize cDNA, and the cDNA can be used as a template to amplify a DNA fragment containing a gene encoding an antibody against the protein using PCR, thereby isolating a gene encoding a high-affinity antibody.
 上記の人工的に再構築した抗体遺伝子から,高親和性を有する目的の蛋白質に対する抗体をコードする遺伝子を選択する場合は,一旦,人工的に再構築した抗体遺伝子を発現ベクターに組み込み,この発現ベクターをホスト細胞に導入する。このとき,ホスト細胞として用いる細胞としては,人工的に再構築した抗体遺伝子を組み込んだ発現ベクターを導入することにより抗体遺伝子を発現させることのできる細胞であれば原核細胞,真核細胞を問わず特に限定はないが,ヒト,マウス,チャイニーズハムスター等の哺乳動物由来の細胞が好ましく,特にチャイニーズハムスター卵巣由来のCHO細胞,又はマウス骨髄腫に由来するNS/0細胞が好ましい。また,抗体遺伝子をコードする遺伝子を組み込んで発現させるために用いる発現ベクターは,哺乳動物細胞内に導入させたときに,該遺伝子を発現させるものであれば特に限定なく用いることができる。発現ベクターに組み込まれた該遺伝子は,哺乳動物細胞内で遺伝子の転写の頻度を調節することができるDNA配列(遺伝子発現制御部位)の下流に配置される。本発明において用いることのできる遺伝子発現制御部位としては,例えば,サイトメガロウイルス由来のプロモーター,SV40初期プロモーター,ヒト伸長因子-1アルファ(EF-1α)プロモーター,ヒトユビキチンCプロモーター等が挙げられる。 When selecting a gene encoding an antibody against a target protein having high affinity from the above artificially reconstructed antibody genes, the artificially reconstructed antibody gene is first incorporated into an expression vector, and this expression vector is introduced into a host cell. In this case, the cells used as the host cell are not particularly limited, regardless of whether they are prokaryotic or eukaryotic, as long as they can express the antibody gene by introducing an expression vector incorporating the artificially reconstructed antibody gene, but cells derived from mammals such as humans, mice, and Chinese hamsters are preferred, and CHO cells derived from Chinese hamster ovaries or NS/0 cells derived from mouse myeloma are particularly preferred. In addition, the expression vector used to incorporate and express a gene encoding an antibody gene can be used without any particular limitation as long as it expresses the gene when introduced into a mammalian cell. The gene incorporated into the expression vector is located downstream of a DNA sequence (gene expression control site) that can regulate the frequency of gene transcription in a mammalian cell. Examples of gene expression control sites that can be used in the present invention include a promoter derived from a cytomegalovirus, an SV40 early promoter, a human elongation factor-1 alpha (EF-1α) promoter, and a human ubiquitin C promoter.
 このような発現ベクターが導入された哺乳動物細胞は,発現ベクターに組み込まれた上述の人工的に再構築した抗体を発現するようになる。このようにして得た,人工的に再構築した抗体を発現する細胞から,所望の蛋白質に対する抗体に対して高親和性を有する抗体を産生する細胞を選択する場合,当該蛋白質をプレートに添加してこれに保持させた後,当該蛋白質に細胞の培養上清を接触させ,次いで,当該蛋白質と結合していない抗体をプレートから除去し,プレートに保持された抗体の量を測定する方法が用いられる。この方法によれば,細胞の培養上清に含まれる抗体の当該蛋白質に対する親和性が高いほど,プレートに保持された抗体の量が多くなる。従って,プレートに保持された抗体の量を測定し,より多くの抗体が保持されたプレートに対応する細胞を,当該蛋白質に対して相対的に高い親和性を有する抗体を産生する細胞株として選択することができ,ひいては,当該蛋白質に対して高親和性を有する抗体をコードする遺伝子を選択できる。このようにして選択された細胞株から,当該蛋白質に対する抗体をコードする遺伝子を含むDNA断片を,PCR法を用いて増幅することにより,高親和性抗体をコードする遺伝子を単離することもできる。  Mammalian cells into which such an expression vector has been introduced will express the above-mentioned artificially reconstructed antibody incorporated in the expression vector. When selecting cells that produce antibodies with high affinity for antibodies against a desired protein from the cells thus obtained that express the artificially reconstructed antibodies, a method is used in which the protein is added to a plate and retained thereon, the cell culture supernatant is brought into contact with the protein, and then antibodies that are not bound to the protein are removed from the plate, and the amount of antibody retained on the plate is measured. According to this method, the higher the affinity of the antibody contained in the cell culture supernatant for the protein, the greater the amount of antibody retained on the plate. Therefore, by measuring the amount of antibody retained on the plate, cells corresponding to the plate that retain a greater number of antibodies can be selected as cell lines that produce antibodies with relatively high affinity for the protein, and thus a gene encoding an antibody with high affinity for the protein can be selected. From the cell line thus selected, a DNA fragment containing a gene encoding an antibody against the protein can be amplified using PCR to isolate a gene encoding a high affinity antibody.
 本発明の一実施形態におけるSAとリソソーム酵素との融合蛋白質,SAとサイトカインとの融合蛋白質,又はSAと神経栄養因子との融合蛋白質を,抗体と結合させた結合体を製造する方法としては,融合蛋白質と抗体とを非ペプチドリンカー又はペプチドリンカーを介して結合させる方法がある。非ペプチドリンカーとしては,ポリエチレングリコール,ポリプロピレングリコール,エチレングリコールとプロピレングリコールとの共重合体,ポリオキシエチル化ポリオール,ポリビニルアルコール,多糖類,デキストラン,ポリビニルエーテル,生分解性高分子,脂質重合体,キチン類,及びヒアルロン酸,又はこれらの誘導体,若しくはこれらを組み合わせたものを用いることができる。ペプチドリンカーは,ペプチド結合した1~50個のアミノ酸から構成されるペプチド鎖若しくはその誘導体であって,そのN末端とC末端が,それぞれ融合蛋白質又は抗体のいずれかと共有結合を形成することにより,融合蛋白質と抗体とを結合させるものである。ここで,SA,リソソーム酵素,サイトカイン,神経栄養因子は,それぞれヒト由来のものでもよく,ヒト以外の動物種由来のものであってもよい。 In one embodiment of the present invention, a method for producing a conjugate in which a fusion protein of SA and a lysosomal enzyme, a fusion protein of SA and a cytokine, or a fusion protein of SA and a neurotrophic factor is bound to an antibody is a method in which the fusion protein and the antibody are bound via a non-peptide linker or a peptide linker. As the non-peptide linker, polyethylene glycol, polypropylene glycol, a copolymer of ethylene glycol and propylene glycol, polyoxyethylated polyol, polyvinyl alcohol, polysaccharides, dextran, polyvinyl ether, biodegradable polymers, lipid polymers, chitins, and hyaluronic acid, or derivatives thereof, or combinations thereof, can be used. The peptide linker is a peptide chain or a derivative thereof consisting of 1 to 50 amino acids bound by peptide bonds, and its N-terminus and C-terminus form covalent bonds with either the fusion protein or the antibody, respectively, thereby binding the fusion protein and the antibody. Here, the SA, lysosomal enzyme, cytokine, and neurotrophic factor may each be derived from a human or a non-human animal.
 融合蛋白質と抗体の結合体は,融合蛋白質と抗体とを別々に製造し,次いで,これらを非ペプチドリンカー又はペプチドリンカーと反応させて,リンカーの一端に融合蛋白質結合させ,他の一端に抗体を結合させることにより製造される。融合蛋白質と抗体は,それぞれ組換え蛋白質として製造したものを用いることができる。この製造方法によれば,例えば,HSAとヒトリソソーム酵素との融合蛋白質と抗体が非ペプチドリンカー又はペプチドリンカーを介して結合した結合体,HSAとヒトサイトカインとの融合蛋白質と抗体が非ペプチドリンカー又はペプチドリンカーを介して結合した結合体,HSAとヒト神経栄養因子との融合蛋白質と抗体が非ペプチドリンカー又はペプチドリンカーを介して結合した結合体が製造される。 A conjugate of a fusion protein and an antibody is produced by producing the fusion protein and the antibody separately, then reacting them with a non-peptide linker or a peptide linker to bind the fusion protein to one end of the linker and the antibody to the other end. The fusion protein and the antibody can each be produced as recombinant proteins. According to this production method, for example, a conjugate in which a fusion protein of HSA and a human lysosomal enzyme is bound to an antibody via a non-peptide linker or a peptide linker, a conjugate in which a fusion protein of HSA and a human cytokine is bound to an antibody via a non-peptide linker or a peptide linker, or a conjugate in which a fusion protein of HSA and a human neurotrophic factor is bound to an antibody via a non-peptide linker or a peptide linker can be produced.
 かかる製造方法により,例えば,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質が,抗体又はリガンドと,非ペプチドリンカー又はペプチドリンカーを介して抗体と結合した結合体を取得することができる。 By using this manufacturing method, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4 can be obtained in which the fusion protein is bound to an antibody or a ligand via a non-peptide linker or a peptide linker.
 本発明の一実施形態におけるSAとリソソーム酵素との融合蛋白質,SAとサイトカインとの融合蛋白質,又はSAと神経栄養因子との融合蛋白質を,抗体又はリガンドと結合させた結合体を製造する方法としては,これら融合蛋白質のC末端又はN末端を,リンカーを介して又は直接,抗体又はリガンドのN末端又はC末端にペプチド結合により結合させたものとして製造する方法がある。かかる結合体は,以下に例示する製造方法により,組換え蛋白質として製造することができる。 In one embodiment of the present invention, a conjugate formed by binding a fusion protein of SA and a lysosomal enzyme, a fusion protein of SA and a cytokine, or a fusion protein of SA and a neurotrophic factor to an antibody or a ligand can be produced by binding the C-terminus or N-terminus of these fusion proteins to the N-terminus or C-terminus of an antibody or ligand via a peptide bond, either directly or via a linker. Such conjugates can be produced as recombinant proteins by the production methods exemplified below.
 例えば,抗体と,HSAとhGALCとの融合蛋白質とは,抗体の重鎖又は軽鎖のC末端側又はN末端側に,リンカーを介して又は直接に,それぞれ融合蛋白質のN末端又はC末端をペプチド結合により結合させたものとして製造できる。このように抗体と,HSAとhGALCとの融合蛋白質とを結合させてなる結合体は,抗体の重鎖又は軽鎖をコードするcDNAの3’末端側又は5’末端側に,直接又はリンカーをコードするDNA断片を挟んで,HSAとhGALCとの融合蛋白質をコードするcDNAがインフレームで配置されたDNA断片を,哺乳動物細胞用の発現ベクターに組み込み,この発現ベクターを導入した哺乳動物細胞を培養することにより,結合体である蛋白質として得ることができる。この哺乳動物細胞には,HSAとhGALCとの融合蛋白質をコードするDNA断片を重鎖と結合させる場合にあっては,抗体の軽鎖をコードするcDNA断片を組み込んだ哺乳動物細胞用の発現ベクターも,同じホスト細胞に導入されることができ,また,HSAとhGALCとの融合蛋白質をコードするDNA断片を軽鎖と結合させる場合にあっては,抗体の重鎖をコードするcDNA断片を組み込んだ哺乳動物細胞用の発現ベクターも,同じホスト細胞に導入されることができる。抗体が一本鎖抗体又はVHHである場合,抗体とHSAとhGALCとの融合蛋白質とを結合させた結合体は,HSAとhGALCとの融合蛋白質をコードするcDNAの5’末端側又は3’末端側に,直接,又はリンカーをコードするDNA断片を挟んで,一本鎖抗体又はVHHをコードするcDNAを連結したDNA断片を,(哺乳動物細胞,酵母等の真核生物又は大腸菌等の原核生物細胞用の)発現ベクターに組み込み,この発現ベクターを導入したこれらの細胞中で発現させることにより,組換え蛋白質として得ることができる。HSAとhGBAとの融合蛋白質と抗体との結合体,HSAとhIL-10との融合蛋白質と抗体との結合体,HSAとhBDNFとの融合蛋白質と抗体との結合体,HSAとhNGFとの融合蛋白質と抗体との結合体,HSAとhNT-3との融合蛋白質と抗体との結合体,及びHSAとhNT-4との融合蛋白質と抗体との結合体も,HSAとhGALCとの融合蛋白質と抗体の結合体と同様にして組換え蛋白質として得ることができる。 For example, an antibody and a fusion protein of HSA and hGALC can be produced by binding the N-terminus or C-terminus of the fusion protein to the C-terminus or N-terminus of the heavy or light chain of the antibody via a peptide bond, either directly or via a linker. In this way, a conjugate formed by binding an antibody to a fusion protein of HSA and hGALC can be obtained as a conjugate protein by inserting a DNA fragment in which the cDNA encoding the fusion protein of HSA and hGALC is placed in frame at the 3'-terminus or 5'-terminus of the cDNA encoding the heavy or light chain of the antibody, either directly or via a DNA fragment encoding a linker, into an expression vector for mammalian cells, and culturing mammalian cells into which this expression vector has been introduced. In the case where a DNA fragment encoding a fusion protein of HSA and hGALC is bound to a heavy chain, an expression vector for mammalian cells incorporating a cDNA fragment encoding the light chain of an antibody can also be introduced into the same host cell, and in the case where a DNA fragment encoding a fusion protein of HSA and hGALC is bound to a light chain, an expression vector for mammalian cells incorporating a cDNA fragment encoding the heavy chain of an antibody can also be introduced into the same host cell. In the case where the antibody is a single-chain antibody or VHH, the conjugate of the antibody and the fusion protein of HSA and hGALC can be obtained as a recombinant protein by incorporating a DNA fragment encoding a single-chain antibody or VHH linked to the 5'-end or 3'-end of the cDNA encoding the fusion protein of HSA and hGALC directly or via a DNA fragment encoding a linker into an expression vector (for mammalian cells, eukaryotic cells such as yeast, or prokaryotic cells such as E. coli) and expressing the DNA fragment in the cells into which the expression vector has been introduced. Conjugates of the fusion protein of HSA and hGBA and an antibody, conjugates of the fusion protein of HSA and hIL-10 and an antibody, conjugates of the fusion protein of HSA and hBDNF and an antibody, conjugates of the fusion protein of HSA and hNGF and an antibody, conjugates of the fusion protein of HSA and hNT-3 and an antibody, and conjugates of the fusion protein of HSA and hNT-4 and an antibody can also be obtained as recombinant proteins in the same manner as the conjugates of the fusion protein of HSA and hGALC and an antibody.
 抗体,HSA及びhGALCは,HSAとhGALCとの間に抗体を介在させる形態の結合体とすることもできる。この場合,HSAのC末端に,リンカーを介して又は直接に,抗体の重鎖又は軽鎖のN末端を結合させ,更にそのC末端に,リンカーを介して又は直接に,hGALCを,それぞれペプチド結合により結合させたものとして製造できる。又は,hGALCのC末端に,リンカーを介して又は直接に,抗体の重鎖又は軽鎖のN末端を結合させ,更にそのC末端に,リンカーを介して又は直接に,HSAを,それぞれペプチド結合により結合させたものとして製造できる。抗体が一本鎖抗体又はVHHであっても,抗体,HSA及びhGALCは,HSAとhGALCとの間に抗体を介在させる形態の融合蛋白質とすることができる。この場合,HSAのC末端に,リンカーを介して又は直接に,一本鎖抗体又はVHHのN末端を結合させ,更にそのC末端に,リンカーを介して又は直接にhGALCを,それぞれペプチド結合により結合させたものとして製造できる。又は,hGALCのC末端にリンカーを介して又は直接に,一本鎖抗体又はVHHのN末端を,更にそのC末端にリンカーを介して又は直接にHSAを,それぞれペプチド結合により結合させたものとして製造できる。同様の結合体は,hGALC以外のリソソーム酵素,例えばhGBAについても製造することができ,また,サイトカイン,例えば,hIL-10についても製造することができ,また,神経栄養因子,例えば,hBDNF,hNGF,hNT-3,hNT-4についても製造することができる。このように,抗体を介在させる形態の結合体も,便宜上,抗体と他のヒトリソソーム酵素とHSAとの融合蛋白質,抗体と他のヒトサイトカインとHSAとの融合蛋白質,及び抗体と他のヒト神経栄養因子とHSAとの融合蛋白質という。 Antibody, HSA and hGALC can also be conjugated in a form in which an antibody is interposed between HSA and hGALC. In this case, the N-terminus of the heavy or light chain of the antibody can be bound to the C-terminus of HSA via a linker or directly, and hGALC can be bound to the C-terminus via a peptide bond either via a linker or directly. Alternatively, the N-terminus of the heavy or light chain of the antibody can be bound to the C-terminus of hGALC via a linker or directly, and HSA can be bound to the C-terminus via a linker or directly, respectively. Even if the antibody is a single-chain antibody or VHH, the antibody, HSA and hGALC can be made into a fusion protein in a form in which an antibody is interposed between HSA and hGALC. In this case, the N-terminus of the single-chain antibody or VHH can be bound to the C-terminus of HSA via a linker or directly, and hGALC can be bound to the C-terminus via a peptide bond either via a linker or directly. Alternatively, the N-terminus of a single chain antibody or VHH can be bound to the C-terminus of hGALC via a linker or directly, and HSA can be bound to the C-terminus of the N-terminus of the single chain antibody or VHH via a linker or directly, respectively, by peptide bonds. Similar conjugates can also be produced for lysosomal enzymes other than hGALC, such as hGBA, cytokines, such as hIL-10, and neurotrophic factors, such as hBDNF, hNGF, hNT-3, and hNT-4. For the sake of convenience, conjugates in this manner that involve an antibody are also referred to as fusion proteins of an antibody, another human lysosomal enzyme, and HSA, fusion proteins of an antibody, another human cytokine, and HSA, and fusion proteins of an antibody, another human neurotrophic factor, and HSA.
 上記の手法は,他のヒトリソソーム酵素とHSAとの融合蛋白質,他のヒトサイトカインとHSAとの融合蛋白質,又は他のヒト神経栄養因子とHSAとの融合蛋白質と,抗体との結合体の製造にも応用できる。また,SAがヒト以外の動物種であるものである結合体,リソソーム酵素,サイトカイン,及び神経栄養因子がヒト以外の動物種である結合体の製造にも応用できる。 The above method can also be applied to the production of conjugates of fusion proteins of other human lysosomal enzymes and HSA, fusion proteins of other human cytokines and HSA, or fusion proteins of other human neurotrophic factors and HSA, and antibodies. It can also be applied to the production of conjugates in which the SA is from an animal species other than human, and conjugates in which the lysosomal enzymes, cytokines, and neurotrophic factors are from an animal species other than human.
 上述したHSAとhGALCとの融合蛋白質又はHSAとhGBAとの融合蛋白質を製造するために用いることのできる,発現ベクター,宿主細胞,培地等は,上記の融合蛋白質と抗体との結合体の製造においても用いることができる。 The expression vectors, host cells, media, etc. that can be used to produce the above-mentioned fusion protein of HSA and hGALC or the fusion protein of HSA and hGBA can also be used to produce a conjugate of the above-mentioned fusion protein and antibody.
 抗体,HSA及びhGALCとの結合体の好ましい実施形態として,以下の(1)~(4)が挙げられる。すなわち:
 (1)抗体の重鎖のC末端に直接又はリンカーを介してHSAとhGALCとの融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (2)抗体の重鎖のN末端に直接又はリンカーを介してHSAとhGALCとの融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (3)抗体の軽鎖のC末端に直接又はリンカーを介してHSAとhGALCとの融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (4)抗体の軽鎖のN末端に直接又はリンカーを介してHSAとhGALCとの融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (5)HSAのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にhGALCが結合した結合体;
 (6)hGALCのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にHSAが結合した結合体。
 (1)の好適な一例として,配列番号25のアミノ酸配列を有するFab重鎖のC末端に,配列番号9が3回繰り返されるアミノ酸配列を有するリンカー,更にそのC末端に配列番号3のアミノ酸配列を有するHSA,更にそのC末端に配列番号9のアミノ酸配列を有するリンカー,更にそのC末端に配列番号1のアミノ酸配列を有するhGALCが結合した結合体Aと,配列番号23の軽鎖とを含むものが挙げられる。ここで結合体Aは配列番号28のアミノ酸配列を有する。
 (2)の好適な一例として,配列番号3のアミノ酸配列を有するHSAのC末端に,配列番号9のアミノ酸配列を有するリンカー,更にそのC末端に配列番号1のアミノ酸配列を有するhGALC,更にそのC末端に配列番号9が3回繰り返されるアミノ酸配列を有するリンカー,更にそのC末端に配列番号25のアミノ酸配列を有するFab重鎖が結合した結合体Bと,配列番号23の軽鎖とを含むものが挙げられる。ここで当該結合体Bは配列番号30のアミノ酸配列を有する。
 (3)の好適な一例として,配列番号25のアミノ酸配列を有するFab重鎖のC末端に,配列番号9が3回繰り返されるアミノ酸配列を有するリンカー,更にそのC末端に配列番号1のアミノ酸配列を有するhGALC,更にそのC末端に配列番号9のアミノ酸配列を有するリンカー,更にそのC末端に配列番号3のアミノ酸配列を有するHSAが結合した結合体Cと,配列番号23の軽鎖とを含むものが挙げられる。ここで当該結合体Cは配列番号32のアミノ酸配列を有する。
 (4)の好適な一例として,配列番号1のアミノ酸配列を有するhGALCのC末端に,配列番号9のアミノ酸配列を有するリンカー,更にそのC末端に配列番号3のアミノ酸配列を有するHSA,更にそのC末端に配列番号9が3回繰り返されるアミノ酸配列を有するリンカー,更にそのC末端に配列番号25のアミノ酸配列を有するFab重鎖が結合した結合体Dと,配列番号23の軽鎖とを含むものが挙げられる。ここで当該結合体Cは配列番号34のアミノ酸配列を有する。  
Preferred embodiments of the conjugates of antibody, HSA and hGALC include the following (1) to (4):
(1) A conjugate in which a fusion protein of HSA and hGALC is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(2) A conjugate in which a fusion protein of HSA and hGALC is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(3) A conjugate in which a fusion protein of HSA and hGALC is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(4) A conjugate in which a fusion protein of HSA and hGALC is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hGALC is further bound to the C-terminus of the single-chain antibody or VHH;
(6) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of hGALC directly or via a linker, and HSA is further bound to the C-terminus of the antibody or VHH.
A preferred example of (1) is a conjugate A in which a Fab heavy chain having the amino acid sequence of SEQ ID NO:25 is connected to its C-terminus with a linker having an amino acid sequence in which SEQ ID NO:9 is repeated three times, and further connected to its C-terminus with HSA having the amino acid sequence of SEQ ID NO:3, and further connected to its C-terminus with a linker having the amino acid sequence of SEQ ID NO:9, and further connected to its C-terminus with hGALC having the amino acid sequence of SEQ ID NO:1, and a light chain of SEQ ID NO:23. Here, conjugate A has the amino acid sequence of SEQ ID NO:28.
A suitable example of (2) is a conjugate B in which a linker having the amino acid sequence of SEQ ID NO: 9 is attached to the C-terminus of HSA having the amino acid sequence of SEQ ID NO: 3, hGALC having the amino acid sequence of SEQ ID NO: 1 at its C-terminus, a linker having an amino acid sequence in which SEQ ID NO: 9 is repeated three times at its C-terminus, and a Fab heavy chain having the amino acid sequence of SEQ ID NO: 25 at its C-terminus is attached to the light chain of SEQ ID NO: 23. Here, conjugate B has the amino acid sequence of SEQ ID NO: 30.
A preferred example of (3) is a conjugate C in which a linker having an amino acid sequence of SEQ ID NO: 25, an amino acid sequence in which SEQ ID NO: 9 is repeated three times, and hGALC having the amino acid sequence of SEQ ID NO: 1 at its C-terminus, a linker having the amino acid sequence of SEQ ID NO: 9 at its C-terminus, and HSA having the amino acid sequence of SEQ ID NO: 3 at its C-terminus are bound to the light chain of SEQ ID NO: 23. Here, the conjugate C has the amino acid sequence of SEQ ID NO: 32.
A preferred example of (4) is a conjugate D in which hGALC having the amino acid sequence of SEQ ID NO:1 is connected to its C-terminus with a linker having the amino acid sequence of SEQ ID NO:9, and further connected to its C-terminus with HSA having the amino acid sequence of SEQ ID NO:3, and further connected to its C-terminus with a linker having an amino acid sequence in which SEQ ID NO:9 is repeated three times, and further connected to its C-terminus with a Fab heavy chain having the amino acid sequence of SEQ ID NO:25, and a light chain of SEQ ID NO:23. Here, conjugate C has the amino acid sequence of SEQ ID NO:34.
 抗体,HSA及びhGBAとの結合体の好ましい実施形態として,以下の(1)~(4)が挙げられる。すなわち:
 (1)抗体の重鎖のC末端に直接又はリンカーを介してHSAとhGBAとの融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (2)抗体の重鎖のN末端に直接又はリンカーを介してHSAとhGBAとの融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (3)抗体の軽鎖のC末端に直接又はリンカーを介してHSAとhGBAとの融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (4)抗体の軽鎖のN末端に直接又はリンカーを介してHSAとhGBAとの融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (5)HSAのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にhGBAが結合した結合体;
 (6)hGBAのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にHSAが結合した結合体。
Preferred embodiments of the conjugates of antibody, HSA and hGBA include the following (1) to (4):
(1) A conjugate in which a fusion protein of HSA and hGBA is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(2) A conjugate in which a fusion protein of HSA and hGBA is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(3) A conjugate in which a fusion protein of HSA and hGBA is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(4) A conjugate in which a fusion protein of HSA and hGBA is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hGBA is further bound to the C-terminus of the single-chain antibody or VHH;
(6) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of hGBA directly or via a linker, and HSA is further bound to the C-terminus of the antibody or VHH.
 抗体,HSA及びhIL-10との結合体の好ましい実施形態として,以下の(1)~(4)が挙げられる。すなわち:
 (1)抗体の重鎖のC末端に直接又はリンカーを介してHSAとhIL-10との融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (2)抗体の重鎖のN末端に直接又はリンカーを介してHSAとhIL-10との融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (3)抗体の軽鎖のC末端に直接又はリンカーを介してHSAとhIL-10との融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (4)抗体の軽鎖のN末端に直接又はリンカーを介してHSAとhIL-10との融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (5)HSAのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にhIL-10が結合した結合体;
 (6)hIL-10のC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にHSAが結合した結合体。
Preferred embodiments of the conjugate of an antibody, HSA and hIL-10 include the following (1) to (4):
(1) A conjugate in which a fusion protein of HSA and hIL-10 is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(2) A conjugate in which a fusion protein of HSA and hIL-10 is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(3) A conjugate in which a fusion protein of HSA and hIL-10 is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(4) A conjugate in which a fusion protein of HSA and hIL-10 is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hIL-10 is further bound to the C-terminus of the single-chain antibody or VHH;
(6) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of hIL-10 directly or via a linker, and HSA is further bound to the C-terminus of the antibody or VHH.
 抗体,HSA及びhBDNFとの結合体の好ましい実施形態として,以下の(1)~(4)が挙げられる。すなわち:
 (1)抗体の重鎖のC末端に直接又はリンカーを介してHSAとhBDNFとの融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (2)抗体の重鎖のN末端に直接又はリンカーを介してHSAとhBDNFとの融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (3)抗体の軽鎖のC末端に直接又はリンカーを介してHSAとhBDNFとの融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (4)抗体の軽鎖のN末端に直接又はリンカーを介してHSAとhBDNFとの融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (5)HSAのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にhBDNFが結合した結合体;
 (6)hBDNFのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にHSAが結合した結合体。
Preferred embodiments of the conjugate of an antibody, HSA and hBDNF include the following (1) to (4):
(1) A conjugate in which a fusion protein of HSA and hBDNF is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(2) A conjugate in which a fusion protein of HSA and hBDNF is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(3) A conjugate in which a fusion protein of HSA and hBDNF is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(4) A conjugate in which a fusion protein of HSA and hBDNF is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hBDNF is further bound to the C-terminus of the single-chain antibody or VHH;
(6) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of hBDNF directly or via a linker, and HSA is further bound to the C-terminus of the antibody or VHH.
 抗体,HSA及びhNGFの結合体の好ましい実施形態として,以下の(1)~(4)が挙げられる。すなわち:
 (1)抗体の重鎖のC末端に直接又はリンカーを介してHSAとhNGFとの融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (2)抗体の重鎖のN末端に直接又はリンカーを介してHSAとhNGFとの融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (3)抗体の軽鎖のC末端に直接又はリンカーを介してHSAとhNGFとの融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (4)抗体の軽鎖のN末端に直接又はリンカーを介してHSAとhNGFとの融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (5)HSAのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にhNGFが結合した結合体;
 (6)hNGFのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にHSAが結合した結合体。
Preferred embodiments of the conjugate of antibody, HSA and hNGF include the following (1) to (4):
(1) A conjugate in which a fusion protein of HSA and hNGF is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(2) A conjugate in which a fusion protein of HSA and hNGF is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(3) A conjugate in which a fusion protein of HSA and hNGF is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(4) A conjugate in which a fusion protein of HSA and hNGF is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hNGF is further bound to the C-terminus of the single-chain antibody or VHH;
(6) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of hNGF directly or via a linker, and HSA is further bound to the C-terminus of the antibody or VHH.
 抗体,HSA及びhNT-3の結合体の好ましい実施形態として,以下の(1)~(4)が挙げられる。すなわち:
 (1)抗体の重鎖のC末端に直接又はリンカーを介してHSAとhNT-3との融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (2)抗体の重鎖のN末端に直接又はリンカーを介してHSAとhNT-3との融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (3)抗体の軽鎖のC末端に直接又はリンカーを介してHSAとhNT-3との融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (4)抗体の軽鎖のN末端に直接又はリンカーを介してHSAとhNT-3との融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (5)HSAのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にhNT-3が結合した結合体;
 (6)hNT-3のC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にHSAが結合した結合体。
Preferred embodiments of the conjugate of an antibody, HSA and hNT-3 include the following (1) to (4):
(1) A conjugate in which a fusion protein of HSA and hNT-3 is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(2) A conjugate in which a fusion protein of HSA and hNT-3 is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(3) A conjugate in which a fusion protein of HSA and hNT-3 is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(4) A conjugate in which a fusion protein of HSA and hNT-3 is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hNT-3 is further bound to the C-terminus of the single-chain antibody or VHH;
(6) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of hNT-3 directly or via a linker, and HSA is further bound to the C-terminus of the antibody or VHH.
 抗体,HSA及びhNT-4の結合体の好ましい実施形態として,以下の(1)~(4)が挙げられる。すなわち:
 (1)抗体の重鎖のC末端に直接又はリンカーを介してHSAとhNT-4との融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (2)抗体の重鎖のN末端に直接又はリンカーを介してHSAとhNT-4との融合蛋白質が結合した結合体と,抗体の軽鎖とを含むもの;
 (3)抗体の軽鎖のC末端に直接又はリンカーを介してHSAとhNT-4との融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (4)抗体の軽鎖のN末端に直接又はリンカーを介してHSAとhNT-4との融合蛋白質が結合した結合体と,抗体の重鎖とを含むもの;
 (5)HSAのC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にhNT-4が結合した結合体;
 (6)hNT-4のC末端に直接又はリンカーを介して一本鎖抗体又はVHHが結合し,更にそのC末端にHSAが結合した結合体。
Preferred embodiments of the conjugate of an antibody, HSA and hNT-4 include the following (1) to (4):
(1) A conjugate in which a fusion protein of HSA and hNT-4 is bound to the C-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(2) A conjugate in which a fusion protein of HSA and hNT-4 is bound to the N-terminus of an antibody heavy chain directly or via a linker, and an antibody light chain;
(3) A conjugate in which a fusion protein of HSA and hNT-4 is bound to the C-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(4) A conjugate in which a fusion protein of HSA and hNT-4 is bound to the N-terminus of an antibody light chain directly or via a linker, and an antibody heavy chain;
(5) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of HSA directly or via a linker, and hNT-4 is further bound to the C-terminus of the single-chain antibody or VHH;
(6) A conjugate in which a single-chain antibody or VHH is bound to the C-terminus of hNT-4 directly or via a linker, and HSA is further bound to the C-terminus of the antibody or VHH.
 SAとリソソーム酵素との融合蛋白質,SAとサイトカインとの融合蛋白質,又は,SAと神経栄養因子との融合蛋白質と,抗体とを,リンカーを介して結合させる場合のリンカーについて,以下に詳述する。これらのリンカーは,特に,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質と,抗体とを,結合させる場合のリンカーとしても好適である。  Linkers used when binding antibodies to a fusion protein of SA and a lysosomal enzyme, a fusion protein of SA and a cytokine, or a fusion protein of SA and a neurotrophic factor are described in detail below. These linkers are particularly suitable as linkers when binding antibodies to a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4.
 抗体又はリガンドと融合蛋白質との間に配置されるリンカーは,好ましくは1~60個又は1~50個,より好ましくは1~17個,更に好ましくは1~10個,更により好ましくは1~5個のアミノ酸から構成されるペプチド鎖であるが,リンカーを構成するアミノ酸の個数は,1個,2個,3個,1~17個,1~10個,10~40個,20~34個,23~31個,25~29個,27個等と適宜調整できる。そのようなリンカーは,これにより連結された抗体が脳血管内皮細胞上の受容体との親和性を保持し,且つ当該リンカーにより連結された融合蛋白質が,生理的条件下で当該融合蛋白質の生理活性を発揮できる限り,そのアミノ酸配列に限定はないが,好ましくは,グリシンとセリンから構成されるものである。例えば,グリシン又はセリンのいずれか1個のアミノ酸からなるもの,アミノ酸配列Gly-Ser,アミノ酸配列Ser-Ser,アミノ酸配列Gly-Gly-Ser,アミノ酸配列Gly-Gly-Gly-Gly-Ser(配列番号9),アミノ酸配列Gly-Gly-Gly-Gly-Gly-Ser(配列番号10),アミノ酸配列Ser-Gly-Gly-Gly-Gly(配列番号11),又はこれらのアミノ酸配列が1~10個,あるいは2~5個連続してなる配列を含むものが挙げられる。1~50個のアミノ酸からなる配列,2~17個,2~10個,10~40個,20~34個,23~31個,25~29個,又は27個のアミノ酸からなる配列等を有するものである。例えば,アミノ酸配列Gly-Serを含むものはリンカーとして好適に用いることができる。また,アミノ酸配列Gly-Serに続いてアミノ酸配列Gly-Gly-Gly-Gly-Ser(配列番号9)が5個連続してなる計27個のアミノ酸配列を含むものはリンカーとして好適に用いることができる。更には,アミノ酸配列Gly-Gly-Gly-Gly-Ser(配列番号9)が5個連続してなる計25個のアミノ酸配列を含むものもリンカーとして好適に用いることができる。
 なお,本発明において,1つのペプチド鎖に複数のリンカーが含まれる場合,便宜上,各リンカーはN末端側から順に,第1のリンカー,第2のリンカーというように命名する。
The linker disposed between the antibody or ligand and the fusion protein is preferably a peptide chain composed of 1 to 60 or 1 to 50, more preferably 1 to 17, even more preferably 1 to 10, and even more preferably 1 to 5 amino acids, but the number of amino acids constituting the linker can be appropriately adjusted to 1, 2, 3, 1 to 17, 1 to 10, 10 to 40, 20 to 34, 23 to 31, 25 to 29, 27, etc. Such a linker is not limited in its amino acid sequence as long as the antibody linked thereto retains affinity for a receptor on cerebrovascular endothelial cells and the fusion protein linked by the linker can exert the physiological activity of the fusion protein under physiological conditions, but is preferably composed of glycine and serine. For example, those consisting of one amino acid of either glycine or serine, the amino acid sequence Gly-Ser, the amino acid sequence Ser-Ser, the amino acid sequence Gly-Gly-Ser, the amino acid sequence Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 9), the amino acid sequence Gly-Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 10), the amino acid sequence Ser-Gly-Gly-Gly-Gly (SEQ ID NO: 11), or those containing a sequence consisting of 1 to 10 or 2 to 5 consecutive amino acids of these amino acid sequences. Those having a sequence consisting of 1 to 50 amino acids, 2 to 17, 2 to 10, 10 to 40, 20 to 34, 23 to 31, 25 to 29, or 27 amino acids, etc. For example, those containing the amino acid sequence Gly-Ser can be suitably used as a linker. In addition, a linker containing a total of 27 amino acids consisting of the amino acid sequence Gly-Ser followed by five consecutive amino acid sequences of Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 9) can be preferably used as the linker. Furthermore, a linker containing a total of 25 amino acids consisting of five consecutive amino acid sequences of Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 9) can also be preferably used as the linker.
In the present invention, when one peptide chain contains multiple linkers, for convenience, each linker is named, starting from the N-terminus, as the first linker, the second linker, and so on.
 SAとリソソーム酵素との融合蛋白質,SAとサイトカインとの融合蛋白質,又は,SAと神経栄養因子との融合蛋白質と結合させるべき抗体の具体的な例として,以下の抗ヒトトランスフェリン受容体抗体(抗hTfR抗体)が挙げられる。例えば,
 (1)軽鎖の可変領域が,CDR1として配列番号96のアミノ酸配列を,CDR2として配列番号97のアミノ酸配列を,及びCDR3として配列番号98のアミノ酸配列を,それぞれ含んでなり,かつ重鎖の可変領域が,CDR1として配列番号99のアミノ酸配列を,CDR2として配列番号100のアミノ酸配列を,及びCDR3として配列番号101のアミノ酸配列を,それぞれ含んでなるもの,及び
 (2)軽鎖の可変領域が,CDR1として配列番号96のアミノ酸配列を,CDR2として配列番号97のアミノ酸配列を,及びCDR3として配列番号98のアミノ酸配列を,それぞれ含んでなり,かつ重鎖の可変領域が,CDR1として配列番号102のアミノ酸配列を,CDR2として配列番号103のアミノ酸配列を,及びCDR3として配列番号104のアミノ酸配列を,それぞれ含んでなるもの,
 である。ただし,これらに限られず,上記アミノ酸配列に対しては適宜,置換,欠失,付加等の変異を加えることができる。なお,上記(1)及び(2)の抗hTfR抗体はヒト化抗hTfR抗体である。また,上記(1)及び(2)の抗hTfR抗体はFabであっても良い。
Specific examples of antibodies to be bound to the fusion protein of SA and a lysosomal enzyme, the fusion protein of SA and a cytokine, or the fusion protein of SA and a neurotrophic factor include the following anti-human transferrin receptor antibodies (anti-hTfR antibodies).
(1) a light chain variable region comprising the amino acid sequence of SEQ ID NO:96 as CDR1, the amino acid sequence of SEQ ID NO:97 as CDR2, and the amino acid sequence of SEQ ID NO:98 as CDR3, and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO:99 as CDR1, the amino acid sequence of SEQ ID NO:100 as CDR2, and the amino acid sequence of SEQ ID NO:101 as CDR3, respectively; and (2) a light chain variable region comprising the amino acid sequence of SEQ ID NO:96 as CDR1, the amino acid sequence of SEQ ID NO:97 as CDR2, and the amino acid sequence of SEQ ID NO:98 as CDR3, respectively, and a heavy chain variable region comprising the amino acid sequence of SEQ ID NO:102 as CDR1, the amino acid sequence of SEQ ID NO:103 as CDR2, and the amino acid sequence of SEQ ID NO:104 as CDR3,
However, the present invention is not limited to these, and the above amino acid sequences may be appropriately mutated by substitution, deletion, addition, etc. The above anti-hTfR antibodies (1) and (2) are humanized anti-hTfR antibodies. The above anti-hTfR antibodies (1) and (2) may be Fab.
 上記(1)及び(2)の抗hTfR抗体は,特に,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質と結合させるべき抗体として好適なものである。 The anti-hTfR antibodies (1) and (2) above are particularly suitable as antibodies to be bound to a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4.
 上記(1)及び(2)の抗hTfR抗体に置換,付加,欠失等を加えたものも,ヒトトランスフェリン受容体への親和性を保持するものである限り,結合体を構成する抗体とすることができる。 The anti-hTfR antibodies (1) and (2) above with substitutions, additions, deletions, etc. can also be used as antibodies constituting the conjugate, so long as they retain their affinity for the human transferrin receptor.
 上記(1)及び(2)の抗hTfR抗体の軽鎖のアミノ酸配列のアミノ酸を他のアミノ酸へ置換させる場合,置換させるアミノ酸の個数は,好ましくは1~10個であり,より好ましくは1~5個であり,更に好ましくは1~3個,更により好ましくは1又は2個である。軽鎖のアミノ酸配列中のアミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1~10個であり,より好ましくは1~5個であり,更に好ましくは1~3個であり,更により好ましくは1又は2個である。また,これらアミノ酸の置換と欠失を組み合わせた変異を加えることもできる。 When replacing amino acids in the amino acid sequence of the light chain of the anti-hTfR antibody of (1) and (2) above with other amino acids, the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2. When deleting amino acids in the amino acid sequence of the light chain, the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2. Mutations that combine these amino acid replacements and deletions can also be added.
 上記(1)及び(2)の抗hTfR抗体の軽鎖のアミノ酸配列にアミノ酸を付加する場合,軽鎖のアミノ酸配列中若しくはN末端側又はC末端側に,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個,更により好ましくは1又は2個のアミノ酸が付加される。これらアミノ酸の付加,置換及び欠失を組み合わせた変異を加えることもできる。変異を加えた軽鎖のアミノ酸配列は,元の軽鎖のアミノ酸配列と,好ましくは80%以上の同一性を有し,より好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示す。 When amino acids are added to the amino acid sequence of the light chain of the anti-hTfR antibody of (1) and (2) above, preferably 1 to 10 amino acids, more preferably 1 to 5 amino acids, even more preferably 1 to 3 amino acids, and even more preferably 1 or 2 amino acids are added to the amino acid sequence of the light chain or to the N-terminus or C-terminus. Mutations that combine the addition, substitution, and deletion of these amino acids can also be added. The amino acid sequence of the mutated light chain preferably has an identity of 80% or more, more preferably an identity of 90% or more, and even more preferably an identity of 95% or more, with the amino acid sequence of the original light chain.
 上記(1)及び(2)の抗hTfR抗体の重鎖のアミノ酸配列のアミノ酸を他のアミノ酸へ置換させる場合,置換させるアミノ酸の個数は,好ましくは1~10個であり,より好ましくは1~5個であり,更に好ましくは1~3個,更により好ましくは1又は2個である。重鎖のアミノ酸配列中のアミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1~10個であり,より好ましくは1~5個であり,更に好ましくは1~3個であり,更により好ましくは1又は2個である。また,これらアミノ酸の置換と欠失を組み合わせた変異を加えることもできる。 When amino acids in the amino acid sequence of the heavy chain of the anti-hTfR antibody of (1) and (2) above are replaced with other amino acids, the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2. When amino acids in the amino acid sequence of the heavy chain are deleted, the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2. Mutations that combine these amino acid substitutions and deletions can also be added.
 上記(1)及び(2)の抗hTfR抗体の重鎖のアミノ酸配列にアミノ酸を付加する場合,重鎖のアミノ酸配列中若しくはN末端側又はC末端側に,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個,更により好ましくは1又は2個のアミノ酸が付加される。これらアミノ酸の付加,置換及び欠失を組み合わせた変異を加えることもできる。変異を加えた重鎖のアミノ酸配列は,元の重鎖のアミノ酸配列と,好ましくは80%以上の同一性を有し,より好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示す。 When amino acids are added to the amino acid sequence of the heavy chain of the anti-hTfR antibody of (1) and (2) above, preferably 1 to 10 amino acids, more preferably 1 to 5 amino acids, even more preferably 1 to 3 amino acids, and even more preferably 1 or 2 amino acids are added to the amino acid sequence of the heavy chain or to the N-terminus or C-terminus. Mutations that combine the addition, substitution, and deletion of these amino acids can also be added. The amino acid sequence of the mutated heavy chain preferably has an identity of 80% or more, more preferably an identity of 90% or more, and even more preferably an identity of 95% or more, with the amino acid sequence of the original heavy chain.
 また,SAとリソソーム酵素との融合蛋白質,SAとサイトカインとの融合蛋白質,又は,SAと神経栄養因子との融合蛋白質と結合させるべき抗体の具体的な例として,以下の抗hTfR重鎖抗体可変領域(VHH)ペプチド(hTfR親和性ペプチド)が挙げられる。すなわち,
 (3)hTfR親和性ペプチドであって,CDR1として配列番号105又は配列番号106のアミノ酸配列を,CDR2として配列番号107又は配列番号108のアミノ酸配列を,及びCDR3として配列番号109又は配列番号110のアミノ酸配列を,それぞれ含んでなるもの,及び
 (4)hTfR親和性ペプチドであって,CDR1として配列番号105又は配列番号106のアミノ酸配列を,CDR2として配列番号111又は配列番号112のアミノ酸配列を,及びCDR3として配列番号109又は配列番号110のアミノ酸配列を,それぞれ含んでなるもの。
 である。ただし,これらに限られず,上記アミノ酸配列に対しては適宜,置換,欠失,付加等の変異を加えることができる。なお,上記(3)及び(4)のhTfR親和性ペプチドは本明細書でいう抗体に含まれる。
Specific examples of antibodies to be bound to the fusion protein of SA and a lysosomal enzyme, the fusion protein of SA and a cytokine, or the fusion protein of SA and a neurotrophic factor include the following anti-hTfR heavy chain antibody variable region (VHH) peptides (hTfR affinity peptides):
(3) An hTfR affinity peptide comprising the amino acid sequence of SEQ ID NO: 105 or SEQ ID NO: 106 as CDR1, the amino acid sequence of SEQ ID NO: 107 or SEQ ID NO: 108 as CDR2, and the amino acid sequence of SEQ ID NO: 109 or SEQ ID NO: 110 as CDR3, and (4) an hTfR affinity peptide comprising the amino acid sequence of SEQ ID NO: 105 or SEQ ID NO: 106 as CDR1, the amino acid sequence of SEQ ID NO: 111 or SEQ ID NO: 112 as CDR2, and the amino acid sequence of SEQ ID NO: 109 or SEQ ID NO: 110 as CDR3,
However, the present invention is not limited to these, and the above amino acid sequences may be appropriately mutated by substitution, deletion, addition, etc. The above hTfR affinity peptides (3) and (4) are included in the antibody referred to in this specification.
 上記(3)及び(4)のhTfR親和性ペプチドは,特に,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質と結合させるべき抗体として好適なものである。 The hTfR affinity peptides (3) and (4) above are particularly suitable as antibodies to be bound to a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4.
 上記(3)及び(4)のhTfR親和性ペプチドに置換,付加,欠失等を加えたものも,ヒトトランスフェリン受容体への親和性を保持するものである限り,結合体を構成する抗体とすることができる。 The above hTfR affinity peptides (3) and (4) can also be used as antibodies that contain substitutions, additions, deletions, etc., as long as they retain their affinity for the human transferrin receptor.
 上記(3)及び(4)のhTfR親和性ペプチドのアミノ酸配列のアミノ酸を他のアミノ酸へ置換させる場合,置換させるアミノ酸の個数は,好ましくは1~10個であり,より好ましくは1~5個であり,更に好ましくは1~3個,更により好ましくは1又は2個である。hTfR親和性ペプチドのアミノ酸配列中のアミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1~10個であり,より好ましくは1~5個であり,更に好ましくは1~3個であり,更により好ましくは1又は2個である。また,これらアミノ酸の置換と欠失を組み合わせた変異を加えることもできる。 When amino acids in the amino acid sequence of the hTfR affinity peptide in (3) and (4) above are replaced with other amino acids, the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2. When amino acids in the amino acid sequence of the hTfR affinity peptide are deleted, the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, even more preferably 1 to 3, and even more preferably 1 or 2. Mutations that combine these amino acid substitutions and deletions can also be added.
 上記(3)及び(4)のhTfR親和性ペプチドのアミノ酸配列にアミノ酸を付加する場合,軽鎖のアミノ酸配列中若しくはN末端側又はC末端側に,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個,更により好ましくは1又は2個のアミノ酸が付加される。これらアミノ酸の付加,置換及び欠失を組み合わせた変異を加えることもできる。変異を加えた軽鎖のアミノ酸配列は,元のhTfR親和性ペプチドのアミノ酸配列と,好ましくは80%以上の同一性を有し,より好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示す。 When amino acids are added to the amino acid sequence of the hTfR affinity peptide in (3) and (4) above, preferably 1 to 10 amino acids are added to the amino acid sequence of the light chain or to the N-terminus or C-terminus, more preferably 1 to 5 amino acids, even more preferably 1 to 3 amino acids, and even more preferably 1 or 2 amino acids. Mutations that combine the addition, substitution, and deletion of these amino acids can also be added. The amino acid sequence of the mutated light chain preferably has an identity of 80% or more, more preferably 90% or more, and even more preferably 95% or more with the amino acid sequence of the original hTfR affinity peptide.
 HSAとhGALCとの融合蛋白質は,脳血管内皮細胞上の受容体に対する抗体又はリガンドと結合させた結合体とすることにより,血液脳関門を通過させて脳内で機能を発揮させることができる。従って,当該結合体は,GALCの欠損を原因とする中枢神経系の疾患,例えばクラッベ病の治療のための血中投与用薬剤の製造のために使用することができる。また,当該結合体は,GALCの欠損を原因とする中枢神経系の疾患,例えばクラッベ病の治療上有効量を,該当する患者に血中投与(点滴静注等の静脈注射を含む)することを含む,治療方法において使用することができる。血中投与された当該結合体は,脳内に到達する他,GALCを発現する他の臓器や器官にも到達することができる。また,当該薬剤は,当該疾患の発症を予防するために用いることもできる。 The fusion protein of HSA and hGALC can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration into the blood to treat central nervous system diseases caused by GALC deficiency, such as Krabbe disease. The conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the conjugate into the blood (including intravenous injection such as intravenous drip) to a patient for central nervous system diseases caused by GALC deficiency, such as Krabbe disease. The conjugate administered into the blood can reach not only the brain, but also other organs and tissues that express GALC. The drug can also be used to prevent the onset of the disease.
 HSAとhGBAとの融合蛋白質は,脳血管内皮細胞上の受容体に対する抗体又はリガンドと結合させた結合体とすることにより,血液脳関門を通過させて脳内で機能を発揮させることができる。従って,当該結合体は,GBAの欠損を原因とする中枢神経系の疾患,例えばゴーシェ病の治療のための血中投与用薬剤の製造のために使用することができる。また,当該結合体は,GBAの欠損を原因とする中枢神経系の疾患,例えばゴーシェ病の治療上有効量を,該当する患者に血中投与(点滴静注等の静脈注射を含む)することを含む,治療方法において使用することができる。血中投与された当該結合体は,は,脳内に到達する他,他の臓器や器官にも到達することができる。また,当該薬剤は,当該疾患の発症を予防するために用いることもできる。 The fusion protein of HSA and hGBA can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration into the blood to treat central nervous system diseases caused by GBA deficiency, such as Gaucher disease. The conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the conjugate into the blood (including intravenous injection such as intravenous drip) to a patient for central nervous system diseases caused by GBA deficiency, such as Gaucher disease. The conjugate administered into the blood can reach not only the brain but also other organs and tissues. The drug can also be used to prevent the onset of the disease.
 HSAとhIL-10との融合蛋白質は,脳血管内皮細胞上の受容体に対する抗体又はリガンドと結合させた結合体とすることにより,血液脳関門を通過させて脳内で機能を発揮させることができる。従って,当該結合体は,中枢神経系の疾患の治療のための血中投与用薬剤の製造のために使用することができる。また,当該結合体は,中枢神経系の疾患の治療上有効量を,該当する患者に血中投与(点滴静注等の静脈注射を含む)することを含む,治療方法において使用することができる。血中投与された当該結合体は,は,脳内に到達する他,IL-10によって治療効果を発揮することのできる他の臓器や器官にも到達することができる。また,当該薬剤は,当該疾患の発症を予防するために用いることもできる。 The fusion protein of HSA and hIL-10 can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and exert its function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration to the blood for the treatment of a disease of the central nervous system. The conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the disease of the central nervous system to a patient in the blood (including intravenous injection such as intravenous drip). The conjugate administered to the blood can reach not only the brain, but also other organs and tissues in which a therapeutic effect can be exerted by IL-10. The drug can also be used to prevent the onset of the disease.
 HSAとhBDNFとの融合蛋白質は,脳血管内皮細胞上の受容体に対する抗体又はリガンドと結合させた結合体とすることにより,血液脳関門を通過させて脳内で機能を発揮させることができる。従って,当該結合体は,中枢神経系の疾患の治療のための血中投与用薬剤の製造のために使用することができる。また,当該結合体は,中枢神経系の疾患の治療上有効量を,該当する患者に血中投与(点滴静注等の静脈注射を含む)することを含む,治療方法において使用することができる。血中投与された当該結合体は,は,脳内に到達する他,BDNFによって治療効果を発揮することのできる他の臓器や器官にも到達することができる。また,当該薬剤は,当該疾患の発症を予防するために用いることもできる。 The fusion protein of HSA and hBDNF can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and exert its function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration to the blood for the treatment of a disease of the central nervous system. The conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the disease of the central nervous system to a patient in the blood (including intravenous injection such as intravenous drip). The conjugate administered to the blood can reach not only the brain, but also other organs and tissues in which BDNF can exert a therapeutic effect. The drug can also be used to prevent the onset of the disease.
 HSAとhNGFとの融合蛋白質は,脳血管内皮細胞上の受容体に対する抗体又はリガンドと結合させた結合体とすることにより,血液脳関門を通過させて脳内で機能を発揮させることができる。従って,当該結合体は,中枢神経系の疾患の治療のための血中投与用薬剤の製造のために使用することができる。また,当該結合体は,中枢神経系の疾患の治療上有効量を,該当する患者に血中投与(点滴静注等の静脈注射を含む)することを含む,治療方法において使用することができる。血中投与された当該結合体は,脳内に到達する他,NGFによって治療効果を発揮することのできる他の臓器や器官にも到達することができる。また,当該薬剤は,当該疾患の発症を予防するために用いることもできる。 The fusion protein of HSA and hNGF can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and exert its function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration to the blood for the treatment of diseases of the central nervous system. The conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the conjugate to the blood of a patient (including intravenous injection such as intravenous drip infusion). The conjugate administered to the blood can reach not only the brain, but also other organs and tissues in which NGF can exert a therapeutic effect. The drug can also be used to prevent the onset of the disease.
 HSAとhNT-3との融合蛋白質は,脳血管内皮細胞上の受容体に対する抗体又はリガンドと結合させた結合体とすることにより,血液脳関門を通過させて脳内で機能を発揮させることができる。従って,当該結合体は,中枢神経系の疾患の治療のための血中投与用薬剤の製造のために使用することができる。また,当該結合体は,は,中枢神経系の疾患の治療上有効量を,該当する患者に血中投与(点滴静注等の静脈注射を含む)することを含む,治療方法において使用することができる。血中投与された当該結合体は,脳内に到達する他,NT-3によって治療効果を発揮することのできる他の臓器や器官にも到達することができる。また,当該薬剤は,当該疾患の発症を予防するために用いることもできる。 The fusion protein of HSA and hNT-3 can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and exert its function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration to the blood for the treatment of diseases of the central nervous system. The conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the drug to a patient in the blood (including intravenous injection such as intravenous drip infusion). The conjugate administered to the blood can reach not only the brain, but also other organs and tissues in which NT-3 can exert a therapeutic effect. The drug can also be used to prevent the onset of the disease.
 HSAとhNT-4との融合蛋白質は,脳血管内皮細胞上の受容体に対する抗体又はリガンドと結合させた結合体とすることにより,血液脳関門を通過させて脳内で機能を発揮させることができる。従って,当該結合体は,中枢神経系の疾患の治療のための血中投与用薬剤の製造のために使用することができる。また,当該結合体は,中枢神経系の疾患の治療上有効量を,該当する患者に血中投与(点滴静注等の静脈注射を含む)することを含む,治療方法において使用することができる。血中投与された当該結合体は,脳内に到達する他,NT-4によって治療効果を発揮することのできる他の臓器や器官にも到達することができる。また,当該薬剤は,当該疾患の発症を予防するために用いることもできる。 The fusion protein of HSA and hNT-4 can be bound to an antibody or ligand for a receptor on cerebrovascular endothelial cells to form a conjugate that can pass through the blood-brain barrier and exert its function in the brain. Therefore, the conjugate can be used to manufacture a drug for administration to the blood for the treatment of a disease of the central nervous system. The conjugate can also be used in a treatment method that includes administering a therapeutically effective amount of the disease of the central nervous system to a patient in the blood (including intravenous injection such as intravenous drip). The conjugate administered to the blood can reach not only the brain, but also other organs and tissues in which NT-4 can exert a therapeutic effect. The drug can also be used to prevent the onset of the disease.
 HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,及びHSAとhNT-4との融合蛋白質は,それぞれ脳血管内皮細胞上の受容体に対する抗体又はリガンドと結合させることにより,血中に投与して中枢神経系(CNS)において薬効を発揮させるべき薬剤として使用することができる。かかる薬剤は,一般に点滴静脈注射等による静脈注射,皮下注射,筋肉注射により患者に投与されるが,投与経路には特に限定はない。 HSA-hGALC fusion protein, HSA-hGBA fusion protein, HSA-hIL-10 fusion protein, HSA-hBDNF fusion protein, HSA-hNGF fusion protein, HSA-hNT-3 fusion protein, and HSA-hNT-4 fusion protein can be used as drugs that are administered into the blood and exert their medicinal effects in the central nervous system (CNS) by binding to antibodies or ligands for receptors on cerebrovascular endothelial cells. Such drugs are generally administered to patients by intravenous injection, subcutaneous injection, or intramuscular injection, but there are no particular limitations on the administration route.
 HSAとhGALCとの融合蛋白質と抗体との結合体において,hGALCがhGALCとしての機能を有するというときは,hGALCが,通常の野生型のhGALCの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性,例えば90%以上,95%以上の比活性を有することをいう。また,HSAとhGALCとの融合蛋白質と抗体以外の蛋白質との結合体において,hGALCがhGALCとしての機能を有するというときは,hGALCが,通常の野生型のhGALCの比活性を100%としたときに,好ましくは10%以上の比活性を,より好ましくは20%以上の比活性を,更に好ましくは50%以上の比活性を,より更に好ましくは80%以上の比活性,例えば90%以上,95%以上の比活性を有することをいう。なお,ここで当該結合体におけるhGALCの比活性は,当該結合体の単位質量当たりのhGALCの酵素活性(μM/時間/mg蛋白質)に(当該結合体の分子量/当該結合体中のhGALCに相当する部分の分子量)を乗じて算出される。HSAとhGBAとの融合蛋白質と抗体との結合体についても同様のことがいえる。また,HSAとhIL-10との融合蛋白質と抗体との結合体,HSAとhBDNFとの融合蛋白質と抗体との結合体,HSAとhNGFとの融合蛋白質と抗体との結合体,HSAとhNT-3との融合蛋白質と抗体との結合体,及びHSAとhNT-4との融合蛋白質と抗体との結合体についても同様のことがいえるが,もっとも,hIL-10,hBDNF,hNGF,hNT-3,及びhNT-4は酵素ではないため,その比活性とは酵素活性ではなく,hIL-10,hBDNF,hNGF,hNT-3,又はhNT-4それぞれの蛋白質の生理活性に基づくものである。 When it is said that hGALC has the function of hGALC in a conjugate of an antibody and a fusion protein of HSA and hGALC, it means that hGALC has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more or 95% or more, when the specific activity of normal wild-type hGALC is taken as 100%. Also, when it is said that hGALC has the function of hGALC in a conjugate of an antibody and a fusion protein of HSA and hGALC, it means that hGALC has a specific activity of preferably 10% or more, more preferably 20% or more, even more preferably 50% or more, and even more preferably 80% or more, for example 90% or more or 95% or more, when the specific activity of normal wild-type hGALC is taken as 100%. The specific activity of hGALC in the conjugate is calculated by multiplying the enzyme activity of hGALC per unit mass of the conjugate (μM/hour/mg protein) by (molecular weight of the conjugate/molecular weight of the portion of the conjugate corresponding to hGALC). The same can be said for the conjugate of the fusion protein of HSA and hGBA and an antibody. The same can be said for the conjugate of the fusion protein of HSA and hIL-10 and an antibody, the conjugate of the fusion protein of HSA and hBDNF and an antibody, the conjugate of the fusion protein of HSA and hNGF and an antibody, the conjugate of the fusion protein of HSA and hNT-3 and an antibody, and the conjugate of the fusion protein of HSA and hNT-4 and an antibody, although hIL-10, hBDNF, hNGF, hNT-3, and hNT-4 are not enzymes, so the specific activity is based on the physiological activity of each of the proteins hIL-10, hBDNF, hNGF, hNT-3, and hNT-4, rather than on the enzyme activity.
 本発明の一実施形態におけるSAとリソソーム酵素との融合蛋白質,SAとサイトカインとの融合蛋白質,又はSAと神経栄養因子との融合蛋白質は,リガンドとの結合体とすることもできる。ここで,SA,リソソーム酵素,サイトカイン,及び神経栄養因子は,何れもヒト由来のものであっても,ヒト以外の動物種由来のものであってもよい。例えば,SAとリソソーム酵素との融合蛋白質としては,HSAとhGALCとの融合蛋白質及びHSAとhGBAとの融合蛋白質,SAとサイトカインとの融合蛋白質としてはHSAとhIL-10との融合蛋白質,及びSAと神経栄養因子との融合蛋白質としては,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,並びにHSAとhNT-4との融合蛋白質があげられる。 In one embodiment of the present invention, the fusion protein of SA and a lysosomal enzyme, the fusion protein of SA and a cytokine, or the fusion protein of SA and a neurotrophic factor may be a conjugate with a ligand. Here, the SA, the lysosomal enzyme, the cytokine, and the neurotrophic factor may all be derived from humans or from animals other than humans. For example, examples of the fusion protein of SA and a lysosomal enzyme include a fusion protein of HSA and hGALC and a fusion protein of HSA and hGBA, examples of the fusion protein of SA and a cytokine include a fusion protein of HSA and hIL-10, and examples of the fusion protein of SA and a neurotrophic factor include a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, and a fusion protein of HSA and hNT-4.
 ここで,リガンドは,例えば,脳血管内皮細胞上の受容体と特異的に結合することのできるものであり,例えばインスリン受容体,トランスフェリン受容体,レプチン受容体,リポ蛋白質受容体,及びIGF受容体である。インスリン受容体,トランスフェリン受容体,レプチン受容体,リポ蛋白質受容体,及びIGF受容体のそれぞれに対応するリガンドは,インスリン,レプチン,リポ蛋白質及びIGF(IGF-1,IGF-2)である。リガンドは野生型のものであっても,ターゲットとなる受容体に特異的に結合できるものである限り,野生型に変異を加えたものであってもよい。また,ターゲットとなる受容体に特異的に結合できるものである限り,リガンドの断片であってもよい。これらのリガンドは,特に,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質と結合させるリガンドとして好適である。 Here, the ligand is, for example, one that can specifically bind to a receptor on cerebrovascular endothelial cells, such as the insulin receptor, transferrin receptor, leptin receptor, lipoprotein receptor, and IGF receptor. The corresponding ligands for the insulin receptor, transferrin receptor, leptin receptor, lipoprotein receptor, and IGF receptor are insulin, leptin, lipoprotein, and IGF (IGF-1, IGF-2), respectively. The ligand may be a wild type or a mutant wild type, so long as it can specifically bind to the target receptor. Also, it may be a fragment of the ligand, so long as it can specifically bind to the target receptor. These ligands are particularly suitable as ligands to be bound to a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4.
 例えば,リガンドがトランスフェリン(Tf)である場合,トランスフェリンはヒトTfであってもヒト以外の動物種のTfであってもよいが,好ましくはヒトTfである。また,トランスフェリンは野生型であってもよく,トランスフェリン受容体(TfR)への親和性を有するものである限り,変異を導入したものであってもよい。変異は,例えば1~2個のアミノ酸の置換,付加,又は欠失である。また,トランスフェリンは全長であってもよく,トランスフェリン受容体(TfR)への親和性を有するものである限り,その断片であってもよい。インスリン,レプチン,リポ蛋白質及びIGF(IGF-1,IGF-2)についても同様のことがいえる。 For example, when the ligand is transferrin (Tf), the transferrin may be human Tf or Tf of an animal species other than human, but is preferably human Tf. The transferrin may be wild-type, or may have a mutation introduced therein, so long as it has affinity for the transferrin receptor (TfR). The mutation may be, for example, a substitution, addition, or deletion of one or two amino acids. The transferrin may be full-length, or may be a fragment thereof, so long as it has affinity for the transferrin receptor (TfR). The same can be said for insulin, leptin, lipoproteins, and IGF (IGF-1, IGF-2).
 本発明の一実施例におけるSAとリソソーム酵素との融合蛋白質,SAとサイトカインとの融合蛋白質,又はSAと神経栄養因子との融合蛋白質と,リガンドとの結合体は,以下の(1)又は(2)の方法により製造することができる:
(1)融合蛋白質とリガンドとを非ペプチドリンカー又はペプチドリンカーを介して結合体とする;
(2)融合蛋白質とリガンドとを,リンカー配列を介して又は直接,ペプチド結合により結合させた組換え蛋白質として製造する。
In one embodiment of the present invention, a conjugate of a fusion protein of SA and a lysosomal enzyme, a fusion protein of SA and a cytokine, or a fusion protein of SA and a neurotrophic factor, and a ligand can be produced by the following method (1) or (2):
(1) A fusion protein and a ligand are conjugated via a non-peptide linker or a peptide linker;
(2) The fusion protein and the ligand are produced as a recombinant protein bound via a linker sequence or directly via a peptide bond.
 上記(1)の場合,非ペプチドリンカーとしては,ポリエチレングリコール,ポリプロピレングリコール,エチレングリコールとプロピレングリコールとの共重合体,ポリオキシエチル化ポリオール,ポリビニルアルコール,多糖類,デキストラン,ポリビニルエーテル,生分解性高分子,脂質重合体,キチン類,及びヒアルロン酸,又はこれらの誘導体,若しくはこれらを組み合わせたものを用いることができる。ペプチドリンカーは,ペプチド結合した1~50個のアミノ酸から構成されるペプチド鎖若しくはその誘導体であって,そのN末端とC末端が,それぞれ融合蛋白質又はリガンドのいずれかと共有結合を形成することにより,融合蛋白質とリガンドとを結合させるものである。融合蛋白質とリガンドは,それぞれ組換え蛋白質として製造される。 In the case of (1) above, examples of the non-peptide linker that can be used include polyethylene glycol, polypropylene glycol, copolymers of ethylene glycol and propylene glycol, polyoxyethylated polyol, polyvinyl alcohol, polysaccharides, dextran, polyvinyl ether, biodegradable polymers, lipid polymers, chitins, and hyaluronic acid, or derivatives or combinations of these. The peptide linker is a peptide chain or derivatives consisting of 1 to 50 peptide-bonded amino acids, whose N-terminus and C-terminus form covalent bonds with either the fusion protein or the ligand, respectively, thereby binding the fusion protein and the ligand. The fusion protein and the ligand are each produced as recombinant proteins.
 上記(2)の場合,融合蛋白質とリガンドは,融合蛋白質のC末端に,リンカー配列を介して又は直接,リガンドのN末端が結合した組換え蛋白質として,又はリガンドのC末端に,リンカー配列を介して又は直接,融合蛋白質のN末端が結合した組換え蛋白質として製造される。かかる組換え蛋白質を製造するために用いられる発現ベクター,宿主細胞,培地は,上記のHSAとhGALCとの融合蛋白質の製造に用いられるものを応用することができる。 In the case of (2) above, the fusion protein and the ligand are produced as a recombinant protein in which the N-terminus of the ligand is bound to the C-terminus of the fusion protein via a linker sequence or directly, or the N-terminus of the fusion protein is bound to the C-terminus of the ligand via a linker sequence or directly. The expression vectors, host cells, and media used to produce such recombinant proteins can be those used to produce the above-mentioned fusion protein of HSA and hGALC.
 本発明の一実施形態におけるSAとリソソーム酵素との融合蛋白質,SAとサイトカインとの融合蛋白質,又はSAと神経栄養因子との融合蛋白質は,抗体及びリガンドに限らず,その他の蛋白質(A)との結合体とすることもできる。ここで,SA,リソソーム酵素,サイトカイン,及び神経栄養因子は,何れもヒト由来のものであっても,ヒト以外の動物種由来のものであってもよい。例えば,SAとリソソーム酵素との融合蛋白質としては,HSAとhGALCとの融合蛋白質及びHSAとhGBAとの融合蛋白質,SAとサイトカインとの融合蛋白質としてはHSAとhIL-10との融合蛋白質,及びSAと神経栄養因子との融合蛋白質としては,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,並びにHSAとhNT-4との融合蛋白質があげられる。 In one embodiment of the present invention, the fusion protein of SA and a lysosomal enzyme, the fusion protein of SA and a cytokine, or the fusion protein of SA and a neurotrophic factor can be a conjugate with another protein (A) as well as an antibody and a ligand. Here, the SA, the lysosomal enzyme, the cytokine, and the neurotrophic factor may be derived from humans or animals other than humans. For example, examples of the fusion protein of SA and a lysosomal enzyme include a fusion protein of HSA and hGALC and a fusion protein of HSA and hGBA, examples of the fusion protein of SA and a cytokine include a fusion protein of HSA and hIL-10, and examples of the fusion protein of SA and a neurotrophic factor include a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, and a fusion protein of HSA and hNT-4.
 本発明の一実施例におけるSAとリソソーム酵素との融合蛋白質,SAとサイトカインとの融合蛋白質,又はSAと神経栄養因子との融合蛋白質と,蛋白質(A)との結合体は,以下の(1)又は(2)の方法により製造することができる:
(1)融合蛋白質と蛋白質(A)とを非ペプチドリンカー又はペプチドリンカーを介して結合体とする;
(2)融合蛋白質と蛋白質(A)とを,リンカー配列を介して又は直接,ペプチド結合により結合させた組換え蛋白質として製造する。
In one embodiment of the present invention, a conjugate of a fusion protein of SA and a lysosomal enzyme, a fusion protein of SA and a cytokine, or a fusion protein of SA and a neurotrophic factor, and a protein (A) can be produced by the following method (1) or (2):
(1) The fusion protein and protein (A) are conjugated via a non-peptide linker or a peptide linker;
(2) The fusion protein and protein (A) are produced as a recombinant protein by binding them via a linker sequence or directly via a peptide bond.
 上記(1)の場合,非ペプチドリンカーとしては,ポリエチレングリコール,ポリプロピレングリコール,エチレングリコールとプロピレングリコールとの共重合体,ポリオキシエチル化ポリオール,ポリビニルアルコール,多糖類,デキストラン,ポリビニルエーテル,生分解性高分子,脂質重合体,キチン類,及びヒアルロン酸,又はこれらの誘導体,若しくはこれらを組み合わせたものを用いることができる。ペプチドリンカーは,ペプチド結合した1~50個のアミノ酸から構成されるペプチド鎖若しくはその誘導体であって,そのN末端とC末端が,それぞれ融合蛋白質又は蛋白質(A)のいずれかと共有結合を形成することにより,融合蛋白質と蛋白質(A)とを結合させるものである。融合蛋白質と蛋白質(A)は,それぞれ組換え蛋白質として製造される。 In the case of (1) above, examples of the non-peptide linker that can be used include polyethylene glycol, polypropylene glycol, copolymers of ethylene glycol and propylene glycol, polyoxyethylated polyol, polyvinyl alcohol, polysaccharides, dextran, polyvinyl ether, biodegradable polymers, lipid polymers, chitins, and hyaluronic acid, or derivatives or combinations of these. The peptide linker is a peptide chain or derivatives thereof consisting of 1 to 50 peptide-bonded amino acids, and its N-terminus and C-terminus form covalent bonds with either the fusion protein or protein (A), respectively, thereby binding the fusion protein and protein (A). The fusion protein and protein (A) are each produced as recombinant proteins.
 上記(2)の場合,融合蛋白質とリガンドは,融合蛋白質のC末端に,リンカー配列を介して又は直接,リガンドのN末端が結合した組換え蛋白質として,又はリガンドのC末端に,リンカー配列を介して又は直接,融合蛋白質のN末端が結合した組換え蛋白質として製造される。かかる組換え蛋白質を製造するために用いられる発現ベクター,宿主細胞,培地は,HSAとhGALCとの融合蛋白質の製造に用いられるものを応用することができる。 In the case of (2) above, the fusion protein and the ligand are produced as a recombinant protein in which the N-terminus of the ligand is bound to the C-terminus of the fusion protein via a linker sequence or directly, or the N-terminus of the fusion protein is bound to the C-terminus of the ligand via a linker sequence or directly. The expression vectors, host cells, and media used to produce such recombinant proteins can be those used to produce a fusion protein of HSA and hGALC.
 HSAとhGALCとの融合蛋白質と抗体の結合体,及びHSAとhGALCとの融合蛋白質とリガンド又は蛋白質(A)との結合体は,何れもクラッベ病(ガラクトシルセラミドリピドーシス,又はグロボイド細胞白質ジストロフィー)を対象疾患とする医薬組成物として使用することができる。 The conjugate of the fusion protein of HSA and hGALC with an antibody, and the conjugate of the fusion protein of HSA and hGALC with a ligand or protein (A) can both be used as a pharmaceutical composition for the treatment of Krabbe disease (galactosylceramide lipidosis, or globoid cell leukodystrophy).
 HSAとhGBAとの融合蛋白質と抗体の結合体,及びHSAとhGBAとの融合蛋白質とリガンド又は蛋白質(A)との結合体は,何れもゴーシェ病を対象疾患とする医薬組成物として使用することができる。 The conjugate of the fusion protein of HSA and hGBA with an antibody, and the conjugate of the fusion protein of HSA and hGBA with a ligand or protein (A) can both be used as a pharmaceutical composition for the treatment of Gaucher disease.
 HSAとhIL-10との融合蛋白質と抗体の結合体,及びHSAとhIL-10との融合蛋白質とリガンド又は蛋白質(A)との結合体は,何れも炎症性疾患,神経傷害性疼痛,多発性硬化症,脊髄傷害,ALS,神経炎症,関節炎および関節の他の疾患に関連する症状,及び自己免疫疾患等を対象疾患とする医薬組成物として使用することができる。 The conjugate of the fusion protein of HSA and hIL-10 with an antibody, and the conjugate of the fusion protein of HSA and hIL-10 with a ligand or protein (A) can both be used as pharmaceutical compositions for treating inflammatory diseases, neuropathic pain, multiple sclerosis, spinal cord injury, ALS, neuroinflammation, symptoms associated with arthritis and other joint diseases, and autoimmune diseases, etc.
 HSAとhBDNFとの融合蛋白質と抗体の結合体,及びHSAとhBDNFとの融合蛋白質とリガンド又は蛋白質(A)との結合体は,何れもアルツハイマー病,パーキンソン病,ハンチントン病などの神経変性疾患,筋萎縮性側策硬化症などの脊髄変性疾患,糖尿病性神経障害,脳虚血性疾患,Rett症候群などの発達障害,統合失調症,うつ病およびRett症候群等を対象疾患とする医薬組成物として使用することができる。 The conjugate of the fusion protein of HSA and hBDNF with an antibody, and the conjugate of the fusion protein of HSA and hBDNF with a ligand or protein (A) can both be used as pharmaceutical compositions for treating neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, spinal degenerative diseases such as amyotrophic lateral sclerosis, diabetic neuropathy, cerebral ischemic disease, developmental disorders such as Rett syndrome, schizophrenia, depression, and Rett syndrome, etc.
 HSAとhNGFとの融合蛋白質と抗体の結合体,及びHSAとhNGFとの融合蛋白質とリガンド又は蛋白質(A)との結合体は,何れもアルツハイマー病,パーキンソン病,ハンチントン病などの神経変性疾患等を対象疾患とする医薬組成物として使用することができる。 The conjugate of the fusion protein of HSA and hNGF with an antibody, and the conjugate of the fusion protein of HSA and hNGF with a ligand or protein (A) can both be used as pharmaceutical compositions for treating neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease.
 HSAとhNT-3との融合蛋白質と抗体の結合体,及びHSAとhNT-3との融合蛋白質とリガンド又は蛋白質(A)との結合体は,何れも神経変性疾患等を対象疾患とする医薬組成物として使用することができる。 The conjugate of the fusion protein of HSA and hNT-3 with an antibody, and the conjugate of the fusion protein of HSA and hNT-3 with a ligand or protein (A) can both be used as a pharmaceutical composition for treating diseases such as neurodegenerative disorders.
 HSAとhNT-4との融合蛋白質と抗体の結合体,及びHSAとhNT-4との融合蛋白質とリガンド又は蛋白質(A)との結合体は,何れも神経変性疾患等を対象疾患とする医薬組成物として使用することができる。 The conjugate of the fusion protein of HSA and hNT-4 with an antibody, and the conjugate of the fusion protein of HSA and hNT-4 with a ligand or protein (A) can both be used as a pharmaceutical composition for treating diseases such as neurodegenerative disorders.
 HSAとhGALCとの融合蛋白質と抗体の結合体,又はHSAとhGALCとの融合蛋白質とリガンド又は蛋白質(A)との結合体を有効成分として含有してなる医薬組成物は,注射剤として静脈内,筋肉内,腹腔内,皮下又は脳室内に投与することができる。それらの注射剤は,凍結乾燥製剤又は水性液剤として供給することができる。水性液剤とする場合,バイアルに充填した形態としてもよく,注射器に予め充填したものであるプレフィルド型の製剤として供給することもできる。凍結乾燥製剤の場合,使用前に水性媒質に溶解し復元して使用する。水性液剤中に含まれるHSAとhGALCとの融合蛋白質と抗体の結合体における,総蛋白質に占める単量体の比率(単量体の質量/総蛋白質の質量×100(%))は,好ましくは70%以上,より好ましくは80%以上,更に好ましくは90%以上,例えば95%以上であり95%以上である。凍結乾燥製剤を水性媒質に溶解し復元した溶液についても同じことがいえる。以上のことは,HSAとhGBAとの融合蛋白質と抗体の結合体,又はHSAとhGBAとの融合蛋白質とリガンド又は蛋白質(A)との結合体を有効成分として含有してなる医薬組成物,HSAとhIL-10との融合蛋白質と抗体の結合体,又はHSAとhIL-10との融合蛋白質とリガンド又は蛋白質(A)との結合体を有効成分として含有してなる医薬組成物,HSAとhBDNFとの融合蛋白質と抗体の結合体,又はHSAとhBDNFとの融合蛋白質とリガンド又は蛋白質(A)との結合体を有効成分として含有してなる医薬組成物,HSAとhNGFとの融合蛋白質と抗体の結合体,又はHSAとhNGFとの融合蛋白質とリガンド又は蛋白質(A)との結合体を有効成分として含有してなる医薬組成物,HSAとNT-3との融合蛋白質と抗体の結合体,又はHSAとhNT-3との融合蛋白質とリガンド又は蛋白質(A)との結合体を有効成分として含有してなる医薬組成物,及びHSAとNT-4との融合蛋白質と抗体の結合体,又はHSAとhNT-4との融合蛋白質とリガンド又は蛋白質(A)との結合体を有効成分として含有してなる医薬組成物,についても適用し得る。 A pharmaceutical composition containing as an active ingredient a conjugate of an antibody and a fusion protein of HSA and hGALC, or a conjugate of a fusion protein of HSA and hGALC and a ligand or protein (A), can be administered intravenously, intramuscularly, intraperitoneally, subcutaneously, or intracerebroventricularly as an injection. These injections can be supplied as freeze-dried preparations or aqueous liquid preparations. In the case of an aqueous liquid preparation, it may be in the form of a vial filled with the fusion protein, or it can be supplied as a prefilled preparation in a syringe. In the case of a freeze-dried preparation, it is dissolved in an aqueous medium before use and reconstituted. In the conjugate of an antibody and a fusion protein of HSA and hGALC contained in an aqueous liquid preparation, the ratio of monomer to total protein (mass of monomer/mass of total protein x 100 (%)) is preferably 70% or more, more preferably 80% or more, even more preferably 90% or more, for example 95% or more. The same can be said for a solution obtained by dissolving a freeze-dried preparation in an aqueous medium and reconstituting it. The above-mentioned facts are not intended to limit the scope of the present invention, but are intended to provide a pharmaceutical composition comprising, as an active ingredient, a conjugate of a fusion protein of HSA and hGBA and an antibody, or a conjugate of a fusion protein of HSA and hGBA and a ligand or protein (A), a pharmaceutical composition comprising, as an active ingredient, a conjugate of a fusion protein of HSA and hIL-10 and an antibody, or a conjugate of a fusion protein of HSA and hIL-10 and a ligand or protein (A), a pharmaceutical composition comprising, as an active ingredient, a conjugate of a fusion protein of HSA and hBDNF and an antibody, or a conjugate of a fusion protein of HSA and hBDNF and a ligand or protein (A), It may also be applied to pharmaceutical compositions containing as active ingredients a conjugate of a fusion protein of SA and hNGF and an antibody, or a conjugate of a fusion protein of HSA and hNGF and a ligand or protein (A), a conjugate of a fusion protein of HSA and NT-3 and an antibody, or a conjugate of a fusion protein of HSA and hNT-3 and a ligand or protein (A), and a conjugate of a fusion protein of HSA and NT-4 and an antibody, or a conjugate of a fusion protein of HSA and hNT-4 and a ligand or protein (A).
 野生型hGALCは,組換え体蛋白質として発現させたとき,2量体,4量体等の多量体として取得される。これに対して,HSAとhGALCとの融合蛋白質は,組換え体蛋白質として発現させたとき,そのほとんどが単量体として取得される。組換え体蛋白質として製造されたHSAとhGALCとの融合蛋白質における,総蛋白質に占める単量体の比率(単量体の質量/総蛋白質の質量×100(%))は,好ましくは70%以上,より好ましくは80%以上,更に好ましくは90%以上,例えば95%以上であり95%以上である。 When wild-type hGALC is expressed as a recombinant protein, it is obtained as a dimer, tetramer, or other polymer. In contrast, when a fusion protein of HSA and hGALC is expressed as a recombinant protein, most of it is obtained as a monomer. In the fusion protein of HSA and hGALC produced as a recombinant protein, the ratio of monomers to the total protein (monomer mass/total protein mass x 100(%)) is preferably 70% or more, more preferably 80% or more, even more preferably 90% or more, for example 95% or more.
 HSAとhGALCとの融合蛋白質,例えばHSA-hGALC及びhGALC-HSAは,組換え蛋白質として発現させたとき,単量体として均質なものを取得することができるので,製造管理の観点からも,hGALCをHSAとの融合蛋白質として製造するhGALCの製造方法は,hGALCの製造方法として優れたものということができる。 HSA-hGALC fusion proteins, such as HSA-hGALC and hGALC-HSA, can be expressed as recombinant proteins to obtain homogeneous monomers. From the standpoint of production control, the method of producing hGALC as a fusion protein with HSA can be said to be an excellent method of producing hGALC.
 また,HSAとhGALCとの融合蛋白質と抗体の結合体は,組換え体蛋白質として発現させたとき,そのほとんどが単量体として取得される。組換え体蛋白質として製造された当該結合体における,総蛋白質に占める単量体の比率(単量体の質量/総蛋白質の質量×100(%))は,好ましくは70%以上,より好ましくは80%以上,更に好ましくは90%以上,例えば95%以上であり95%以上である。 In addition, when the conjugate of the antibody and the fusion protein of HSA and hGALC is expressed as a recombinant protein, most of it is obtained as a monomer. In the conjugate produced as a recombinant protein, the ratio of monomer to total protein (monomer mass/total protein mass x 100(%)) is preferably 70% or more, more preferably 80% or more, and even more preferably 90% or more, for example 95% or more.
 HSAとhGALCとの融合蛋白質と抗体の結合体は,組換え蛋白質として発現させたとき,単量体として均質なものを取得することができるので,製造管理の観点からも,hGALCをHSA及び抗体との融合蛋白質として製造する方法は,hGALCの製造方法として優れたものということができる。 When the conjugate of the fusion protein of HSA and hGALC with an antibody is expressed as a recombinant protein, a homogeneous monomer can be obtained. Therefore, from the standpoint of production control, the method of producing hGALC as a fusion protein of HSA and an antibody can be said to be an excellent method of producing hGALC.
 SAとリソソーム酵素との融合蛋白質,SAとサイトカインとの融合蛋白質,又はSAと神経栄養因子との融合蛋白質は,当該結合体をコードする遺伝子を含む核酸分子は,遺伝子治療に用いることができる。また,これら融合蛋白質の何れかと抗体との結合体をコードする遺伝子を含む核酸分子は,これら融合蛋白質の何れかとリガンドとの結合体をコードする遺伝子を含む核酸分子,これら融合蛋白質の何れかと蛋白質(A)との結合体をコードする遺伝子を含む核酸分子も,遺伝子治療に用いることができる。  A fusion protein between SA and a lysosomal enzyme, a fusion protein between SA and a cytokine, or a fusion protein between SA and a neurotrophic factor, as well as a nucleic acid molecule containing a gene encoding the conjugate, can be used in gene therapy. In addition, a nucleic acid molecule containing a gene encoding a conjugate between any of these fusion proteins and an antibody, a nucleic acid molecule containing a gene encoding a conjugate between any of these fusion proteins and a ligand, and a nucleic acid molecule containing a gene encoding a conjugate between any of these fusion proteins and protein (A) can also be used in gene therapy.
 ここで,融合蛋白質は,例えば,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質である。また,抗体は,例えば抗トランスフェリン受容体抗体である。また,リガンドは,例えばトランスフェリン又はその断片である。 Here, the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4. The antibody is, for example, an anti-transferrin receptor antibody. The ligand is, for example, transferrin or a fragment thereof.
 本発明の一実施形態における,遺伝子治療に用いることのできる核酸分子としては,例えば以下に示す塩基配列を含むものが挙げられる:
(1)第一の逆方向末端反復(ITR)又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質をコードする塩基配列,更に下流に第二の逆方向末端反復(ITR)又はその機能的等価物を含む塩基配列;
(2)第一の長い末端反復(LTR)又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質をコードする塩基配列,更に下流に第二の長い末端反復(LTR)又はその機能的等価物を含む塩基配列;
(3)リーダー又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質をコードする塩基配列,更に下流にトレイラー又はその機能的等価物を含む塩基配列;
(4)第一の逆方向末端反復(ITR)又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質と抗体との結合体をコードする塩基配列,更に下流に第二の逆方向末端反復(ITR)又はその機能的等価物を含む塩基配列;
 (5)第一の長い末端反復(LTR)又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質と抗体との結合体をコードする塩基配列,更に下流に第二の長い末端反復(LTR)又はその機能的等価物を含む塩基配列;
 (6)リーダー又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質と抗体との結合体をコードする塩基配列,更に下流にトレイラー又はその機能的等価物を含む塩基配列;
(7)第一の逆方向末端反復(ITR)又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質とリガンドとの結合体をコードする塩基配列,更に下流に第二の逆方向末端反復(ITR)又はその機能的等価物を含む塩基配列;
(8)第一の長い末端反復(LTR)又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質とリガンドとの結合体をコードする塩基配列,更に下流に第二の長い末端反復(LTR)又はその機能的等価物を含む塩基配列;
(9)リーダー又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質とリガンドとの結合体をコードする塩基配列,更に下流にトレイラー又はその機能的等価物を含む塩基配列;
(10)第一の逆方向末端反復(ITR)又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質と蛋白質(A)との結合体をコードする塩基配列,更に下流に第二の逆方向末端反復(ITR)又はその機能的等価物を含む塩基配列;
(11)第一の長い末端反復(LTR)又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質と蛋白質(A)との結合体をコードする塩基配列,更に下流に第二の長い末端反復(LTR)又はその機能的等価物を含む塩基配列;及び
(12)リーダー又はその機能的等価物を含む塩基配列,その下流に遺伝子発現制御部位を含む塩基配列,更に下流に融合蛋白質と蛋白質(A)との結合体をコードする塩基配列,更に下流にトレイラー又はその機能的等価物を含む塩基配列。
In one embodiment of the present invention, a nucleic acid molecule that can be used for gene therapy includes, for example, one having the following base sequence:
(1) A base sequence including a first inverted terminal repeat (ITR) or a functional equivalent thereof, a base sequence including a gene expression control site downstream of the first inverted terminal repeat (ITR), a base sequence encoding a fusion protein further downstream, and a base sequence including a second inverted terminal repeat (ITR) or a functional equivalent thereof further downstream;
(2) a nucleotide sequence containing a first long terminal repeat (LTR) or a functional equivalent thereof, a nucleotide sequence downstream thereof containing a gene expression control site, a nucleotide sequence further downstream thereof encoding a fusion protein, and a nucleotide sequence further downstream thereof containing a second long terminal repeat (LTR) or a functional equivalent thereof;
(3) a base sequence containing a leader or a functional equivalent thereof, a base sequence downstream thereof containing a gene expression control site, a base sequence further downstream thereof encoding a fusion protein, and a base sequence further downstream thereof containing a trailer or a functional equivalent thereof;
(4) a nucleotide sequence containing a first inverted terminal repeat (ITR) or a functional equivalent thereof, a nucleotide sequence containing a gene expression control site downstream of the first inverted terminal repeat (ITR), a nucleotide sequence encoding a conjugate of a fusion protein and an antibody further downstream, and a nucleotide sequence containing a second inverted terminal repeat (ITR) or a functional equivalent thereof further downstream;
(5) a nucleotide sequence containing a first long terminal repeat (LTR) or a functional equivalent thereof, a nucleotide sequence downstream thereof containing a gene expression control site, a nucleotide sequence further downstream thereof encoding a conjugate of a fusion protein and an antibody, and a nucleotide sequence further downstream thereof containing a second long terminal repeat (LTR) or a functional equivalent thereof;
(6) a nucleotide sequence containing a leader or a functional equivalent thereof, followed downstream by a nucleotide sequence containing a gene expression control site, followed further downstream by a nucleotide sequence encoding a conjugate of a fusion protein and an antibody, followed further downstream by a nucleotide sequence containing a trailer or a functional equivalent thereof;
(7) A base sequence including a first inverted terminal repeat (ITR) or a functional equivalent thereof, a base sequence including a gene expression control site downstream of the first inverted terminal repeat (ITR), a base sequence encoding a conjugate of a fusion protein and a ligand further downstream, and a base sequence including a second inverted terminal repeat (ITR) or a functional equivalent thereof further downstream;
(8) A base sequence including a first long terminal repeat (LTR) or a functional equivalent thereof, a base sequence including a gene expression control site downstream of the first long terminal repeat (LTR), a base sequence further downstream encoding a conjugate of a fusion protein and a ligand, and a base sequence including a second long terminal repeat (LTR) or a functional equivalent thereof downstream of the first long terminal repeat (LTR);
(9) A base sequence containing a leader or a functional equivalent thereof, a base sequence downstream thereof containing a gene expression control site, a base sequence further downstream thereof encoding a conjugate of a fusion protein and a ligand, and a base sequence further downstream thereof containing a trailer or a functional equivalent thereof;
(10) a base sequence including a first inverted terminal repeat (ITR) or a functional equivalent thereof, a base sequence including a gene expression control site downstream of the first inverted terminal repeat (ITR), a base sequence encoding a conjugate of a fusion protein and protein (A) further downstream, and a base sequence including a second inverted terminal repeat (ITR) or a functional equivalent thereof further downstream;
(11) a base sequence comprising a first long terminal repeat (LTR) or a functional equivalent thereof, a base sequence comprising a gene expression control site downstream thereof, a base sequence encoding a conjugate of a fusion protein and protein (A) further downstream, and a base sequence comprising a second long terminal repeat (LTR) or a functional equivalent thereof; and (12) a base sequence comprising a leader or a functional equivalent thereof, a base sequence comprising a gene expression control site downstream thereof, a base sequence encoding a conjugate of a fusion protein and protein (A) further downstream, and a trailer or a functional equivalent thereof further downstream.
 本発明において「核酸分子」というときは,主に,デオキシリボヌクレオチドがホスホジエステル結合により重合したものであるDNA,リボヌクレオチドがホスホジエステル結合により重合したものであるRNAの何れかのことをいう。 In the present invention, the term "nucleic acid molecule" refers primarily to either DNA, which is a polymer of deoxyribonucleotides formed by phosphodiester bonds, or RNA, which is a polymer of ribonucleotides formed by phosphodiester bonds.
 「核酸分子」がDNAである場合のDNAは一重鎖(一本鎖)でもよく,又,相補鎖を伴う二重鎖であってもよい。DNAが一重鎖である場合のDNAは,(+)鎖であっても(-)鎖であってもよい。DNAを構成する個々のデオキシリボヌクレオチドは,DNAに含まれる蛋白質をコードする遺伝子が哺乳動物(特にヒト)の細胞内でmRNAに翻訳されることができる限り,天然に存在する型のものであってもよく,天然型に修飾を加えたものであってもよい。本発明の一実施形態において,DNAを構成する個々のデオキシリボヌクレオチドは,哺乳動物(特にヒト)の細胞内で,DNAに含まれる蛋白質をコードする遺伝子がmRNAに翻訳され,且つ,DNAの全部又は一部が複製されることができるものである限り,天然に存在する型のものであってもよく,天然型に修飾を加えたものであってもよい。 When the "nucleic acid molecule" is DNA, the DNA may be single-stranded (single-stranded) or double-stranded with a complementary strand. When the DNA is single-stranded, the DNA may be a (+) strand or a (-) strand. The individual deoxyribonucleotides constituting the DNA may be of a naturally occurring type or may be modified from the natural type, so long as the gene encoding the protein contained in the DNA can be translated into mRNA in the cells of a mammal (particularly a human). In one embodiment of the present invention, the individual deoxyribonucleotides constituting the DNA may be of a naturally occurring type or may be modified from the natural type, so long as the gene encoding the protein contained in the DNA can be translated into mRNA in the cells of a mammal (particularly a human) and the whole or part of the DNA can be replicated.
 また,「核酸分子」がRNAである場合のRNAは一重鎖(一本鎖)でもよく,相補鎖を伴う二重鎖であってもよい。RNAが一本鎖である場合のRNAは,(+)鎖であっても(-)鎖であってもよい。本発明の一実施形態において,RNAを構成する個々のリボヌクレオチドは,RNAに含まれる蛋白質をコードする遺伝子が哺乳動物(特にヒト)の細胞内でDNAに逆転写されることができる限り,天然に存在する型のものであってもよく,修飾されたものであってもよい。また,本発明の一実施形態において,RNAを構成する個々のリボヌクレオチドは,RNAに含まれる蛋白質をコードする遺伝子が哺乳動物(特にヒト)の細胞内で蛋白質に翻訳されることができる限り,天然に存在する型のものであってもよく,修飾されたものであってもよい。リボヌクレオチドの修飾は,例えば,RNAのRNaseによる分解を抑制し,RNAの細胞内における安定性を高めるために行われる。 In addition, when the "nucleic acid molecule" is RNA, the RNA may be single-stranded (single-stranded) or double-stranded with a complementary strand. When the RNA is single-stranded, the RNA may be a (+) strand or a (-) strand. In one embodiment of the present invention, the individual ribonucleotides constituting the RNA may be of a naturally occurring type or may be modified, so long as the gene encoding the protein contained in the RNA can be reverse transcribed into DNA in a mammalian (particularly human) cell. In one embodiment of the present invention, the individual ribonucleotides constituting the RNA may be of a naturally occurring type or may be modified, so long as the gene encoding the protein contained in the RNA can be translated into a protein in a mammalian (particularly human) cell. Modification of ribonucleotides is performed, for example, to suppress degradation of RNA by RNase and to increase the stability of RNA in cells.
 本発明の一実施形態において,逆方向末端反復(ITR)というときは,ウイルスゲノムの末端に存在する,同じ配列が反復して存在する塩基配列のことをいう。ITRとして,アデノ随伴ウイルスに由来するもの,及びアデノウイルスに由来するものは好適に用いることができる。例えば,アデノ随伴ウイルスのITRは,おおよそ145塩基の鎖長の領域であり,複製開始点等として機能する。本発明の一実施形態において,核酸分子中には,逆方向末端反復(ITR)が2つ存在し,それぞれ第一の逆方向末端反復(ITR),第二の逆方向末端反復(ITR)という。ここで,2つのITRの間に,融合蛋白質をコードする遺伝子を配置したときに,その5’側に位置するITRを第一の逆方向末端反復(ITR)といい,3’側に位置するITRを第二の逆方向末端反復(ITR)という。逆方向末端反復(ITR)は,複製開始点としての機能,宿主細胞への遺伝子挿入等の,本来のITRの機能の少なくも一つを有するものである限り,何れのウイルス由来のものであってもよく,アデノ随伴ウイルスのITRは好適なものの一つである。 In one embodiment of the present invention, the term "inverted terminal repeat (ITR)" refers to a base sequence that exists at the end of the viral genome and in which the same sequence is repeated. As ITR, those derived from adeno-associated virus and those derived from adenovirus can be preferably used. For example, the ITR of the adeno-associated virus is a region of approximately 145 bases in length and functions as a replication origin, etc. In one embodiment of the present invention, there are two inverted terminal repeats (ITR) in the nucleic acid molecule, which are called the first inverted terminal repeat (ITR) and the second inverted terminal repeat (ITR), respectively. Here, when a gene encoding a fusion protein is placed between two ITRs, the ITR located on the 5' side is called the first inverted terminal repeat (ITR), and the ITR located on the 3' side is called the second inverted terminal repeat (ITR). The inverted terminal repeat (ITR) may be derived from any virus as long as it has at least one of the functions of an original ITR, such as functioning as an origin of replication or inserting a gene into a host cell, and the ITR of the adeno-associated virus is one suitable one.
 ITRがアデノ随伴ウイルスである場合の,AAVの血清型に特に限定はなく,血清型1,2,3,4,5,6,7,8,9,10又は11の何れのものであってもよい。例えば,血清型2のAAVの逆方向末端反復(ITR)は,第一のITRが配列番号113で示される塩基配列を含み,第二のITRが配列番号114で示される塩基配列を含む。 When the ITR is an adeno-associated virus, the serotype of AAV is not particularly limited and may be any of serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11. For example, the inverted terminal repeat (ITR) of AAV serotype 2 has a first ITR that contains the base sequence shown in SEQ ID NO: 113 and a second ITR that contains the base sequence shown in SEQ ID NO: 114.
 また,ITRは,野生型のITRに限らず,野生型のITRの塩基配列に置換,欠失,付加等の改変を加えたものであってもよい。野生型のITRの塩基配列の塩基を他の塩基で置換する場合,置換する塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個である。野生型のITRの塩基配列を欠失させる場合,欠失させる塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個である。また,これら塩基の置換と欠失を組み合わせた変異を加えることもできる。野生型のITRに塩基を付加する場合,ITRの塩基配列中若しくは5’末端又は3’末端に,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個の塩基が付加される。これら塩基の付加,置換及び欠失を組み合わせた変異を加えることもできる。変異を加えたITRの塩基配列は,野生型のITRの塩基配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更に好ましくは98%以上の同一性を示す。 In addition, the ITR is not limited to the wild-type ITR, and may be modified by substitution, deletion, addition, or the like, in the base sequence of the wild-type ITR. When a base in the base sequence of the wild-type ITR is replaced with another base, the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. When a base sequence of the wild-type ITR is deleted, the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. In addition, a mutation combining these base substitutions and deletions can also be added. When a base is added to the wild-type ITR, preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3 bases are added in the base sequence of the ITR or to the 5' end or 3' end. A mutation combining these base additions, substitutions, and deletions can also be added. The nucleotide sequence of the mutated ITR preferably exhibits 80% or more identity to the nucleotide sequence of the wild-type ITR, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
 ITRの機能的等価物とは,機能的にITRに代えて用いることができるものをいう。また, ITRに基づいて人工的に構築されたITRも,ITRを代替することができるものである限り, ITRの機能的等価物である。 A functional equivalent of an ITR is something that can be used functionally in place of an ITR. In addition, an ITR artificially constructed based on an ITR is also a functional equivalent of an ITR as long as it can replace an ITR.
 AAVのITRの機能的等価物とは,機能的にAAVのITRに代えて用いることができるものをいう。また,AAVのITRに基づいて人工的に構築されたITRも,AAVのITRを代替することができるものである限り,AAVのITRの機能的等価物である。  A functional equivalent of an AAV ITR is one that can be used functionally in place of an AAV ITR. In addition, an ITR artificially constructed based on an AAV ITR is also a functional equivalent of an AAV ITR as long as it can replace an AAV ITR.
 人工的に構築された第1のAAVの逆方向末端反復(第一のAAV-ITR)の機能的等価物として,配列番号115で示される塩基配列を有するものが挙げられる(第一のAAV-ITRの機能的等価物)。この配列番号115で示される塩基配列に,置換,欠失,変異を加えたものも,機能的に第一のAAVのITRに代えて用いることができるものである限り,第一のAAV-ITRの機能的等価物に含まれる。配列番号115で示される塩基配列中の塩基を他の塩基で置換する場合,置換する塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個である。配列番号115で示される塩基配列中の塩基を欠失させる場合,欠失させる塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個である。また,これら塩基の置換と欠失を組み合わせた変異を加えたITRも,第一のAAV-ITRの機能的等価物である。配列番号115で示される塩基配列に塩基を付加する場合,当該塩基配列中若しくは5’末端又は3’末端に,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個の塩基を付加する。これら塩基の付加,置換及び欠失を組み合わせた変異を加えたITRも,第一のAAV-ITRの機能的等価物に含まれる。変異を加えたITRの塩基配列は,配列番号115で示される塩基配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更により好ましくは98%以上の同一性を示す。 An example of a functional equivalent of the artificially constructed first AAV inverted terminal repeat (first AAV-ITR) is one having the base sequence shown in SEQ ID NO: 115 (functional equivalent of the first AAV-ITR). This base sequence shown in SEQ ID NO: 115 to which substitutions, deletions, or mutations have been added is also included in the functional equivalent of the first AAV-ITR, so long as it can be used functionally in place of the ITR of the first AAV. When bases in the base sequence shown in SEQ ID NO: 115 are replaced with other bases, the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3. When bases in the base sequence shown in SEQ ID NO: 115 are deleted, the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3. In addition, an ITR to which mutations that combine the substitution and deletion of these bases have been added is also a functional equivalent of the first AAV-ITR. When bases are added to the base sequence shown in SEQ ID NO:115, preferably 1 to 20 bases, more preferably 1 to 10 bases, even more preferably 1 to 5 bases, and even more preferably 1 to 3 bases are added to the base sequence or to the 5' end or 3' end. ITRs with mutations that combine the addition, substitution, and deletion of these bases are also included in the functional equivalent of the first AAV-ITR. The base sequence of the mutated ITR preferably shows 80% or more identity to the base sequence shown in SEQ ID NO:115, more preferably shows 85% or more identity, even more preferably shows 90% or more identity, even more preferably shows 95% or more identity, and even more preferably shows 98% or more identity.
 また,人工的に構築された第2の逆方向末端反復(第二のAAV-ITR)の機能的等価物として,配列番号116で示される塩基配列を有するものが挙げられる(第二のAAV-ITRの機能的等価物)。この配列番号116で示される塩基配列に,置換,欠失,変異を加えたものも,機能的に第二のAAVのITRに代えて用いることができるものである限り,第二のAAVのITRの機能的等価物に含まれる。配列番号116で示される塩基配列中の塩基を他の塩基で置換する場合,置換する塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個である。配列番号116で示される塩基配列中の塩基を欠失させる場合,欠失させる塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個である。また,これら塩基の置換と欠失を組み合わせた変異を加えたITRも,第二のAAVのITRの機能的等価物である。配列番号116で示される塩基配列に塩基を付加する場合,当該塩基配列中若しくは5’末端又は3’末端に,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個の塩基を付加する。これら塩基の付加,置換及び欠失を組み合わせた変異を加えたITRも,第二のAAVのITRの機能的等価物に含まれる。変異を加えたITRの塩基配列は,配列番号116で示される塩基配列と,好ましくは85%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更により好ましくは98%以上の同一性を示す。 Furthermore, an example of a functional equivalent of an artificially constructed second inverted terminal repeat (second AAV-ITR) is one having the base sequence shown in SEQ ID NO: 116 (functional equivalent of the second AAV-ITR). This base sequence shown in SEQ ID NO: 116 to which substitutions, deletions, or mutations have been added is also included in the functional equivalent of the ITR of the second AAV, so long as it can be used functionally in place of the ITR of the second AAV. When bases in the base sequence shown in SEQ ID NO: 116 are replaced with other bases, the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3. When bases in the base sequence shown in SEQ ID NO: 116 are deleted, the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3. In addition, an ITR to which mutations have been added that combine the substitution and deletion of these bases is also a functional equivalent of the ITR of the second AAV. When bases are added to the base sequence shown in SEQ ID NO:116, preferably 1 to 20 bases, more preferably 1 to 10 bases, even more preferably 1 to 5 bases, and even more preferably 1 to 3 bases are added to the base sequence or to the 5' end or 3' end. ITRs with mutations that combine the addition, substitution, and deletion of these bases are also included in the functional equivalent of the ITR of the second AAV. The base sequence of the mutated ITR preferably shows 85% or more identity with the base sequence shown in SEQ ID NO:116, more preferably shows 85% or more identity, even more preferably shows 90% or more identity, even more preferably shows 95% or more identity, and even more preferably shows 98% or more identity.
 本発明の一実施形態において,長い末端反復(LTR)というときは,例えば,真核生物のレトロトランスポゾン,又はレトロウイルスゲノム,レンチウイルスゲノム等の末端に存在する,同じ配列が数百から数千回反復して存在する塩基配列のことをいう。本発明の一実施形態において,核酸分子中には,長い末端反復(LTR)が2つ存在し,それぞれ第一の長い末端反復(LTR),第二の長い末端反復(LTR)という。ここで,2つのLTRの間に,融合蛋白質をコードする遺伝子を配置したときに,5’側に位置するLTRを第一の長い末端反復(LTR)といい,3’側に位置するLTRを第二の長い末端反復(LTR)という。長い末端反復(LTR)は,複製開始点としての機能,宿主細胞への遺伝子挿入等の,本来のLTRの機能の少なくも一つを有するものである限り,何れのウイルス由来のものであってもよく,好適なものとしてはレトロウイルスゲノム及びレンチウイルスが挙げられる。またLTRは野生型のLTRに限らず,野生型のLTRの塩基配列に置換,欠失,付加等の改変を加えたものであってもよい。 In one embodiment of the present invention, the term "long terminal repeat (LTR)" refers to a base sequence in which the same sequence is repeated hundreds to thousands of times, for example, at the end of a retrotransposon of a eukaryote, or a retrovirus genome, lentivirus genome, etc. In one embodiment of the present invention, there are two long terminal repeats (LTRs) in a nucleic acid molecule, which are called the first long terminal repeat (LTR) and the second long terminal repeat (LTR). Here, when a gene encoding a fusion protein is placed between two LTRs, the LTR located on the 5' side is called the first long terminal repeat (LTR), and the LTR located on the 3' side is called the second long terminal repeat (LTR). The long terminal repeat (LTR) may be derived from any virus as long as it has at least one of the functions of an original LTR, such as a function as a replication origin or gene insertion into a host cell, and preferred examples include retrovirus genomes and lentiviruses. In addition, the LTR is not limited to a wild-type LTR, and may be a wild-type LTR with a modified base sequence, such as a substitution, deletion, or addition.
 野生型のLTRの塩基配列の塩基を他の塩基で置換する場合,置換する塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個である。野生型のLTRの塩基配列を欠失させる場合,欠失させる塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個である。また,これら塩基の置換と欠失を組み合わせた変異を加えることもできる。野生型のLTRに塩基を付加する場合,LTRの塩基配列中若しくは5’末端又は3’末端に,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個の塩基が付加される。これら塩基の付加,置換及び欠失を組み合わせた変異を加えることもできる。変異を加えたLTRの塩基配列は,野生型のLTRの塩基配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更により好ましくは98%以上の同一性を示す。 When bases in the wild-type LTR base sequence are replaced with other bases, the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. When the wild-type LTR base sequence is deleted, the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. Mutations combining these base substitutions and deletions can also be added. When bases are added to the wild-type LTR, preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3 bases are added in the LTR base sequence or at the 5' end or 3' end. Mutations combining these base additions, substitutions, and deletions can also be added. The mutated LTR base sequence preferably exhibits 80% or more identity with the wild-type LTR base sequence, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
 LTRの機能的等価物とは,機能的にLTRに代えて用いることができるものをいう。また,LTRに基づいて人工的に構築されたLTRも,LTRを代替することができるものである限り, LTRの機能的等価物である。  A functional equivalent of an LTR is something that can be used functionally in place of an LTR. In addition, an artificially constructed LTR based on an LTR is also a functional equivalent of an LTR as long as it can replace an LTR.
 レンチウイルスのLTRの機能的等価物とは,機能的にレンチウイルスのLTRに代えて用いることができるものをいう。また,レンチウイルスのLTRに基づいて人工的に構築されたLTRも,レンチウイルスのLTRを代替することができるものである限り,レンチウイルスのLTRの機能的等価物である。  A functional equivalent of a lentiviral LTR is one that can be used functionally in place of the lentiviral LTR. In addition, an artificially constructed LTR based on a lentiviral LTR is also a functional equivalent of the lentiviral LTR as long as it can replace the lentiviral LTR.
 レトロウイルスのLTRの機能的等価物とは,機能的にレトロウイルスのLTRに代えて用いることができるものをいう。また,レトロウイルスのLTRに基づいて人工的に構築されたLTRも,レトロウイルスのLTRを代替することができるものである限り,レトロウイルスのLTRの機能的等価物である。  A functional equivalent of a retroviral LTR is one that can be used functionally in place of a retroviral LTR. In addition, an artificially constructed LTR based on a retroviral LTR is also a functional equivalent of a retroviral LTR as long as it can replace the retroviral LTR.
 第一のLTRの機能的等価物として,配列番号117で示される塩基配列を有するものが挙げられる。この配列番号117で示される塩基配列に,置換,欠失,変異を加えたものも,機能的に第一のLTRとして用いることができるものである限り,第一のLTRの機能的等価物に含まれる。配列番号117で示される塩基配列中の塩基を他の塩基で置換する場合,置換する塩基の個数は,好ましくは1~20個,より好ましくは1~20個,更に好ましくは1~5個,更に好ましくは1~3個である。配列番号117で示される塩基配列中の塩基を欠失させる場合,欠失させる塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個である。また,これら塩基の置換と欠失を組み合わせた変異を加えたLTRも,第一のLTRの機能的等価物である。配列番号117で示される塩基配列に塩基を付加する場合,当該塩基配列中若しくは5’末端又は3’末端に,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個の塩基を付加する。これら塩基の付加,置換及び欠失を組み合わせた変異を加えたLTRも,第一のLTRの機能的等価物に含まれる。変異を加えたLTRの塩基配列は,配列番号117で示される塩基配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは95%以上の同一性を示し,更により好ましくは98%以上の同一性を示す。 An example of a functional equivalent of the first LTR is one having the base sequence shown in SEQ ID NO: 117. This base sequence shown in SEQ ID NO: 117 with substitutions, deletions, or mutations is also included in the functional equivalent of the first LTR, so long as it can be functionally used as the first LTR. When bases in the base sequence shown in SEQ ID NO: 117 are replaced with other bases, the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 20, even more preferably 1 to 5, and even more preferably 1 to 3. When bases in the base sequence shown in SEQ ID NO: 117 are deleted, the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3. In addition, an LTR with a mutation that combines the substitution and deletion of these bases is also a functional equivalent of the first LTR. When bases are added to the base sequence shown in SEQ ID NO:117, preferably 1 to 20 bases, more preferably 1 to 10 bases, even more preferably 1 to 5 bases, and even more preferably 1 to 3 bases are added to the base sequence or to the 5' end or 3' end. LTRs with mutations that combine the addition, substitution, and deletion of these bases are also included in the functional equivalent of the first LTR. The base sequence of the mutated LTR preferably shows 80% or more identity with the base sequence shown in SEQ ID NO:117, more preferably shows 85% or more identity, even more preferably shows 90% or more identity, even more preferably shows 95% or more identity, and even more preferably shows 98% or more identity.
 また,第二のLTRの機能的等価物として,配列番号118で示される塩基配列を有するものが挙げられる。この配列番号118で示される塩基配列に,置換,欠失,変異を加えたものも,機能的に第二のLTRとして用いることができるものである限り,第二のLTRの機能的等価物に含まれる。配列番号118で示される塩基配列中の塩基を他の塩基で置換する場合,置換する塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個である。配列番号118で示される塩基配列中の塩基を欠失させる場合,欠失させる塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個である。また,これら塩基の置換と欠失を組み合わせた変異を加えたLTRも,第二のLTRの機能的等価物である。配列番号118で示される塩基配列に塩基を付加する場合,当該塩基配列中若しくは5’末端又は3’末端に,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~5個,更に好ましくは1~3個の塩基を付加する。これら塩基の付加,置換及び欠失を組み合わせた変異を加えたLTRも,第二のLTRの機能的等価物に含まれる。変異を加えたLTRの塩基配列は,配列番号118で示される塩基配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更により好ましくは98%以上の同一性を示す。 Furthermore, an example of a functional equivalent of the second LTR is one having the base sequence shown in SEQ ID NO: 118. This base sequence shown in SEQ ID NO: 118 with substitutions, deletions, or mutations is also included in the functional equivalent of the second LTR as long as it can be functionally used as the second LTR. When bases in the base sequence shown in SEQ ID NO: 118 are replaced with other bases, the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3. When bases in the base sequence shown in SEQ ID NO: 118 are deleted, the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, even more preferably 1 to 5, and even more preferably 1 to 3. Furthermore, an LTR with a mutation that combines the substitution and deletion of these bases is also a functional equivalent of the second LTR. When bases are added to the base sequence shown in SEQ ID NO:118, preferably 1 to 20 bases, more preferably 1 to 10 bases, even more preferably 1 to 5 bases, and even more preferably 1 to 3 bases are added to the base sequence or to the 5' or 3' end. LTRs with mutations that combine the addition, substitution, and deletion of these bases are also included in the functional equivalent of the second LTR. The base sequence of the mutated LTR preferably shows 80% or more identity with the base sequence shown in SEQ ID NO:118, more preferably shows 85% or more identity, even more preferably shows 90% or more identity, even more preferably shows 95% or more identity, and even more preferably shows 98% or more identity.
 本発明の一実施形態において,リーダー及びトレイラーというときは,ウイルスゲノムの末端に存在する,互いに部分的に相補な塩基配列のことをいう。リーダー及びトレイラーとして,センダイウイルスに由来するものは好適に用いることができる。センダイウイルスのリーダー及びトレイラーは,何れも約50塩基の鎖長の領域である。通常,5’側にリーダー,3’側にトレイラーが位置する。 In one embodiment of the present invention, the terms leader and trailer refer to partially complementary base sequences present at the ends of the viral genome. Those derived from Sendai virus can be preferably used as leaders and trailers. Both the leader and trailer of Sendai virus are regions with a chain length of approximately 50 bases. Usually, the leader is located on the 5' side and the trailer is located on the 3' side.
 本発明の一実施形態において好適に用いられるものとして,配列番号119で示される塩基配列を有するセンダイウイスル由来のリーダーと,配列番号120で示される塩基配列を有するセンダイウイスル由来のトレイラーが挙げられる。 Examples that are preferably used in one embodiment of the present invention include a leader derived from Sendai virus having the base sequence shown in SEQ ID NO: 119 and a trailer derived from Sendai virus having the base sequence shown in SEQ ID NO: 120.
 野生型のセンダイウイスル由来のリーダー及びトレイラーに変異を加えたものも,本発明において好適に使用できる。塩基配列の塩基を他の塩基で置換する場合,置換する塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個である。野生型のリーダー又は/及びトレイラーの塩基配列を欠失させる場合,欠失させる塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個である。また,これら塩基の置換と欠失を組み合わせた変異を加えることもできる。塩基を付加する場合,リーダー又は/及びトレイラーの塩基配列中若しくは5’末端又は3’末端に,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個の塩基が付加される。これら塩基の付加,置換及び欠失を組み合わせた変異を加えることもできる。変異を加えた塩基配列は,野生型の塩基配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更により好ましくは98%以上の同一性を示す。 Mutations in the leader and trailer derived from wild-type Sendai virus can also be suitably used in the present invention. When bases in the base sequence are replaced with other bases, the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. When base sequences of the wild-type leader and/or trailer are deleted, the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. Mutations combining these base substitutions and deletions can also be added. When bases are added, preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3 bases are added in the base sequence of the leader and/or trailer or to the 5' end or 3' end. Mutations combining these base additions, substitutions, and deletions can also be added. The mutated base sequence preferably exhibits 80% or more identity to the wild-type base sequence, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
 本発明の一実施形態において,融合蛋白質の遺伝子の発現を制御する遺伝子発現制御部位を含む塩基配列として用いることのできる塩基配列に,それが融合蛋白質をコードする遺伝子が導入される先の哺乳動物(特にヒト)の細胞,組織又は生体内で,この融合蛋白質を発現させることができるものである限り,特に制限はないが,サイトメガロウイルス由来のプロモーター(任意選択でエンハンサーを含む),SV40初期プロモーター,ヒト伸長因子-1α(EF-1α)プロモーター,ヒトユビキチンCプロモーター,レトロウイルスのラウス肉腫ウイルスLTRプロモーター,ジヒドロ葉酸還元酵素プロモーター,及びβ-アクチンプロモーター,ホスホグリセリン酸キナーゼ(PGK)プロモーター,マウスアルブミンプロモーター,ヒトアルブミンプロモーター,及びヒトα-1アンチトリプシンプロモーターを含むものが好適である。例えば,マウスαフェトプロテインエンハンサーの下流にマウスアルブミンプロモーターを含む配列番号121で示される塩基配列を有する合成プロモーター(マウスαフェトプロテインエンハンサー/マウスアルブミンプロモーター)は遺伝子発現制御部位として好適に利用できる。マウスαフェトプロテインエンハンサー/マウスアルブミンプロモーターの下流に配列番号122で示される塩基配列を有するニワトリβアクチン/MVMキメライントロンをその下流に配置させてもよい。かかるイントロンを配置することにより,遺伝子発現制御部位で制御される蛋白質の発現量を増加させることができる。なお,配列番号121で示される塩基配列において,1~219番目の塩基配列がマウスαフェトプロテインエンハンサーであり,241~549番目の塩基配列がマウスアルブミンプロモーターである。 In one embodiment of the present invention, the base sequence that can be used as the base sequence containing the gene expression control site that controls the expression of the gene of the fusion protein is not particularly limited as long as it can express the fusion protein in the cells, tissues, or living body of a mammal (especially a human) into which the gene encoding the fusion protein is introduced. However, preferred are those containing a cytomegalovirus-derived promoter (optionally containing an enhancer), SV40 early promoter, human elongation factor-1α (EF-1α) promoter, human ubiquitin C promoter, retroviral Rous sarcoma virus LTR promoter, dihydrofolate reductase promoter, and β-actin promoter, phosphoglycerate kinase (PGK) promoter, mouse albumin promoter, human albumin promoter, and human α-1 antitrypsin promoter. For example, a synthetic promoter having the base sequence shown in SEQ ID NO: 121 containing a mouse albumin promoter downstream of a mouse α-fetoprotein enhancer (mouse α-fetoprotein enhancer/mouse albumin promoter) can be preferably used as the gene expression control site. A chicken β-actin/MVM chimeric intron having the base sequence shown in SEQ ID NO: 122 may be placed downstream of the mouse α-fetoprotein enhancer/mouse albumin promoter. By placing such an intron, the expression level of the protein controlled by the gene expression control site can be increased. In the base sequence shown in SEQ ID NO: 121, the base sequence from 1 to 219 is the mouse α-fetoprotein enhancer, and the base sequence from 241 to 549 is the mouse albumin promoter.
 遺伝子制御部位は,臓器特異的又は細胞種特異的に発現する遺伝子のプロモーターであってもよい。臓器特異的発現プロモーター又は細胞種特異的発現プロモーターを用いることにより,核酸分子に組込んだ融合蛋白質をコードする遺伝子を,所望の臓器又は細胞で特異的に発現させることができる。 The gene regulatory site may be a promoter of a gene that is expressed in an organ-specific or cell type-specific manner. By using an organ-specific expression promoter or a cell type-specific expression promoter, the gene encoding the fusion protein incorporated into the nucleic acid molecule can be expressed specifically in a desired organ or cell.
 本発明の一実施形態における核酸分子は,第一の逆方向末端反復(ITR)と第二の逆方向末端反復(ITR)の間に,融合蛋白質と抗体との結合体をコードする遺伝子を含むものである。ここで,融合蛋白質は例えば,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質であり,抗体は例えば,抗トランスフェリン受容体抗体である。以下の(1)~(4)は当該核酸分子の例示である:
(1)第一の逆方向末端反復(ITR)と第二の逆方向末端反復(ITR)の間に,融合蛋白質と抗体との結合体をコードする遺伝子を含むものであって,当該遺伝子が,抗体の軽鎖をコードする遺伝子の下流に内部リボソーム結合部位をコードする塩基配列,その下流に抗体の重鎖のC末端側又はN末端側に融合蛋白質を直接又はリンカーを介して結合させた結合体をコードする塩基配列を含むもの;
(2)第一の逆方向末端反復(ITR)と第二の逆方向末端反復(ITR)の間に,融合蛋白質と抗体との結合体をコードする遺伝子を含むものであって,当該遺伝子が,抗体の重鎖のC末端側又はN末端側に融合蛋白質を直接又はリンカーを介して結合させた結合体をコードする遺伝子の下流に内部リボソーム結合部位をコードする塩基配列,その下流に抗体の軽鎖をコードする塩基配列を含むもの;
(3)第一の逆方向末端反復(ITR)と第二の逆方向末端反復(ITR)の間に,融合蛋白質と抗体との結合体をコードする遺伝子を含むものであって,当該遺伝子が,抗体の重鎖をコードする遺伝子の下流に内部リボソーム結合部位をコードする塩基配列,その下流に抗体の軽鎖のC末端側又はN末端側に融合蛋白質を直接又はリンカーを介して結合させた結合体をコードする塩基配列を含むもの;及び
(4)第一の逆方向末端反復(ITR)と第二の逆方向末端反復(ITR)の間に,融合蛋白質と抗体との結合体をコードする遺伝子を含むものであって,当該遺伝子が,抗体の軽鎖のC末端側又はN末端側に融合蛋白質を直接又はリンカーを介して結合させた結合体をコードする遺伝子の下流に内部リボソーム結合部位をコードする塩基配列,その下流に抗体の重鎖をコードする塩基配列を含むもの。
In one embodiment of the present invention, the nucleic acid molecule contains a gene encoding a conjugate of a fusion protein and an antibody between a first inverted terminal repeat (ITR) and a second inverted terminal repeat (ITR). Here, the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody. The following (1) to (4) are examples of the nucleic acid molecule:
(1) A gene encoding a conjugate between a fusion protein and an antibody between a first inverted terminal repeat (ITR) and a second inverted terminal repeat (ITR), the gene including a base sequence encoding an internal ribosome binding site downstream of a gene encoding an antibody light chain, and a base sequence encoding a conjugate in which a fusion protein is bound directly or via a linker to the C-terminus or N-terminus of an antibody heavy chain downstream of the internal ribosome binding site;
(2) A gene encoding a conjugate between a first inverted terminal repeat (ITR) and a second inverted terminal repeat (ITR), the gene including a base sequence encoding an internal ribosome binding site downstream of the gene encoding a conjugate in which the fusion protein is bound directly or via a linker to the C-terminus or N-terminus of the antibody heavy chain, and a base sequence encoding an antibody light chain downstream of the base sequence encoding an internal ribosome binding site;
(3) A gene encoding a conjugate between a fusion protein and an antibody between the first inverted terminal repeat (ITR) and the second inverted terminal repeat (ITR), which contains a base sequence encoding an internal ribosome binding site downstream of a gene encoding the heavy chain of an antibody, and a base sequence encoding a conjugate in which a fusion protein is bound directly or via a linker to the C-terminal or N-terminal side of the light chain of the antibody, downstream of that base sequence; and (4) A gene encoding a conjugate between a fusion protein and an antibody between the first inverted terminal repeat (ITR) and the second inverted terminal repeat (ITR), which contains a gene encoding a conjugate between a fusion protein and an antibody between the first inverted terminal repeat (ITR) and the second inverted terminal repeat (ITR), which contains a base sequence encoding an internal ribosome binding site downstream of a gene encoding a conjugate in which a fusion protein is bound directly or via a linker to the C-terminal or N-terminal side of the light chain of the antibody, and a base sequence encoding the heavy chain of the antibody, downstream of that base sequence.
 上記(1)~(4)において,核酸分子は,第一の逆方向末端反復(ITR)と融合蛋白質と抗体との結合体をコードする遺伝子の間に,遺伝子発現制御部位を含む塩基配列をさらに含むものであってもよい。また,上記(1)~(4)において,内部リボソーム結合部位をコードする塩基配列を,遺伝子発現制御部位を含む塩基配列に代えてもよい。なお,これに限らず,核酸分子が2つの遺伝子発現制御部位を有する場合,便宜上,第一の逆方向末端反復(ITR)側から順に,第一の遺伝子発現制御部位及び第一の遺伝子発現制御部位とする。また,上記(1)~(4)において,内部リボソーム結合部位をコードする塩基配列を,2A自己切断ペプチドをコードする塩基配列に代えてもよい。豚テッショウウイルス由来2Aペプチドは,2A自己切断ペプチドの好適な一例である。 In the above (1) to (4), the nucleic acid molecule may further include a base sequence including a gene expression control site between the first inverted terminal repeat (ITR) and the gene encoding the conjugate of the fusion protein and the antibody. In the above (1) to (4), the base sequence encoding the internal ribosome binding site may be replaced with a base sequence including a gene expression control site. In addition, when the nucleic acid molecule has two gene expression control sites, for convenience, the first gene expression control site and the second gene expression control site are referred to in order from the first inverted terminal repeat (ITR). In the above (1) to (4), the base sequence encoding the internal ribosome binding site may be replaced with a base sequence encoding a 2A self-cleaving peptide. The 2A peptide derived from the porcine teschovirus is a suitable example of a 2A self-cleaving peptide.
 本発明の一実施形態における核酸分子は,第一の長い末端反復(LTR)と第二の長い末端反復(LTR)の間に,融合蛋白質と抗体との結合体をコードする遺伝子を含むものである。ここで,融合蛋白質は例えば,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質であり,抗体は例えば,抗トランスフェリン受容体抗体である。以下の(1)~(4)は当該核酸分子の例示である:
(1)第一の長い末端反復(LTR)と第二の長い末端反復(LTR)の間に,融合蛋白質と抗体との結合体をコードする遺伝子を含むものであって,当該遺伝子が,抗体の軽鎖をコードする遺伝子の下流に内部リボソーム結合部位をコードする塩基配列,その下流に抗体の重鎖のC末端側又はN末端側に融合蛋白質を直接又はリンカーを介して結合させた結合体をコードする塩基配列を含むもの;
(2)第一の長い末端反復(LTR)と第二の長い末端反復(LTR)の間に,融合蛋白質と抗体との結合体をコードする遺伝子を含むものであって,当該遺伝子が,抗体の重鎖のC末端側又はN末端側に融合蛋白質を直接又はリンカーを介して結合させた結合体をコードする遺伝子の下流に内部リボソーム結合部位をコードする塩基配列,その下流に抗体の軽鎖をコードする塩基配列を含むもの;
(3)第一の長い末端反復(LTR)と第二の長い末端反復(LTR)の間に,融合蛋白質と抗体との結合体をコードする遺伝子を含むものであって,当該遺伝子が,抗体の重鎖をコードする遺伝子の下流に内部リボソーム結合部位をコードする塩基配列,その下流に抗体の軽鎖のC末端側又はN末端側に融合蛋白質を直接又はリンカーを介して結合させた結合体をコードする塩基配列を含むもの;
(4)第一の長い末端反復(LTR)と第二の長い末端反復(LTR)の間に,融合蛋白質と抗体との結合体をコードする遺伝子を含むものであって,当該遺伝子が,抗体の軽鎖のC末端側又はN末端側に融合蛋白質を直接又はリンカーを介して結合させた結合体をコードする遺伝子の下流に内部リボソーム結合部位をコードする塩基配列,その下流に抗体の重鎖をコードする塩基配列を含むもの。
 上記(1)~(4)において,核酸分子は,第一の長い末端反復(LTR)と融合蛋白質と抗体との結合体をコードする遺伝子の間に,遺伝子発現制御部位を含む塩基配列をさらに含むものであってもよい。また,上記(1)~(4)において,内部リボソーム結合部位をコードする塩基配列を,遺伝子発現制御部位を含む塩基配列に代えてもよい。なお,これに限らず,核酸分子が2つの遺伝子発現制御部位を有する場合,便宜上,第一の逆方向末端反復(ITR)側から順に,第一の遺伝子発現制御部位及び第一の遺伝子発現制御部位とする。また,上記(1)~(4)において,内部リボソーム結合部位をコードする塩基配列を,2A自己切断ペプチドをコードする塩基配列に代えてもよい。豚テッショウウイルス由来2Aペプチドは,2A自己切断ペプチドの好適な一例である。
In one embodiment of the present invention, the nucleic acid molecule contains a gene encoding a conjugate of a fusion protein and an antibody between a first long terminal repeat (LTR) and a second long terminal repeat (LTR). Here, the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody. The following (1) to (4) are examples of the nucleic acid molecule:
(1) A gene encoding a conjugate of a fusion protein and an antibody is contained between a first long terminal repeat (LTR) and a second long terminal repeat (LTR), and the gene contains a base sequence encoding an internal ribosome binding site downstream of a gene encoding the light chain of the antibody, and a base sequence encoding a conjugate in which the fusion protein is bound directly or via a linker to the C-terminus or N-terminus of the heavy chain of the antibody downstream of the internal ribosome binding site;
(2) A gene encoding a conjugate of a fusion protein and an antibody between the first long terminal repeat (LTR) and the second long terminal repeat (LTR), the gene including a base sequence encoding an internal ribosome binding site downstream of the gene encoding a conjugate in which the fusion protein is bound directly or via a linker to the C-terminus or N-terminus of the antibody heavy chain, and a base sequence encoding an antibody light chain downstream of the base sequence encoding an internal ribosome binding site;
(3) A gene encoding a conjugate between a first long terminal repeat (LTR) and a second long terminal repeat (LTR), the gene including a base sequence encoding an internal ribosome binding site downstream of a gene encoding the heavy chain of an antibody, and a base sequence encoding a conjugate in which the fusion protein is bound directly or via a linker to the C-terminus or N-terminus of the light chain of the antibody downstream of the internal ribosome binding site;
(4) A gene encoding a conjugate between a first long terminal repeat (LTR) and a second long terminal repeat (LTR), the gene including a base sequence encoding an internal ribosome binding site downstream of the gene encoding a conjugate in which the fusion protein is bound directly or via a linker to the C-terminus or N-terminus of the antibody light chain, and a base sequence encoding an antibody heavy chain downstream of that.
In the above (1) to (4), the nucleic acid molecule may further include a base sequence including a gene expression control site between the first long terminal repeat (LTR) and the gene encoding the conjugate of the fusion protein and the antibody. In the above (1) to (4), the base sequence encoding the internal ribosome binding site may be replaced with a base sequence including a gene expression control site. In addition, when the nucleic acid molecule has two gene expression control sites, for convenience, the first gene expression control site and the second gene expression control site are referred to in order from the first inverted terminal repeat (ITR). In the above (1) to (4), the base sequence encoding the internal ribosome binding site may be replaced with a base sequence encoding a 2A self-cleaving peptide. The 2A peptide derived from the porcine teschovirus is a suitable example of the 2A self-cleaving peptide.
 上記内部リボソーム結合部位をコードする塩基配列を含む核酸分子にあっては,遺伝子発現制御部位から融合蛋白質を構成する一方のペプチド鎖の,内部リボソーム結合部位をコードする塩基配列から他方のペプチド鎖の発現が制御されることになる。両ペプチド鎖は,細胞内で対をなして融合蛋白質と抗体との結合体が形成される。 In the case of a nucleic acid molecule containing a base sequence encoding the above-mentioned internal ribosome binding site, the expression of one peptide chain constituting the fusion protein is controlled by the gene expression control site, while the expression of the other peptide chain is controlled by the base sequence encoding the internal ribosome binding site. The two peptide chains pair up within the cell to form a conjugate between the fusion protein and the antibody.
 本発明の一実施形態における核酸分子は,AAVベクター系において用いることができる。野生型のAAVが宿主細胞のヒト細胞に単独で感染すると,ウイルスゲノムは,ウイルスゲノムの両端に存在する逆方向末端反復(ITR)を介して,第19番染色体に部位特異的に組み込まれる。宿主細胞のゲノム中に取り込まれたウイルスゲノムの遺伝子はほとんど発現しないが,細胞がヘルパーウイルスに感染するとAAVは宿主ゲノムから切り出され,感染性ウイルスの複製が開始される。ヘルパーウイルスがアデノウイルスの場合,ヘルパー作用を担う遺伝子はE1A,E1B,E2A,VA1,及びE4である。ここで,宿主細胞が,アデノウイルスのE1A,E1Bでトランスフォームしたヒト胎児腎組織由来細胞であるHEK293細胞の場合にあっては,E1A及びE1B遺伝子は,宿主細胞において元々発現している。 The nucleic acid molecule in one embodiment of the present invention can be used in an AAV vector system. When wild-type AAV infects a human host cell alone, the viral genome is site-specifically integrated into chromosome 19 via the inverted terminal repeats (ITRs) present at both ends of the viral genome. The genes of the viral genome integrated into the genome of the host cell are hardly expressed, but when the cell is infected with a helper virus, AAV is excised from the host genome and replication of the infectious virus begins. When the helper virus is an adenovirus, the genes responsible for the helper function are E1A, E1B, E2A, VA1, and E4. Here, when the host cell is a HEK293 cell, which is a human fetal kidney tissue-derived cell transformed with adenovirus E1A and E1B, the E1A and E1B genes are originally expressed in the host cell.
 野生型AAVゲノムは2つの遺伝子rep及びcapを含む。rep遺伝子から産生されるrep蛋白質(rep78,rep68,rep52及びrep40)は,カプシド形成に必須であるとともに,ウイルスゲノムの染色体への組み込みに介在する。cap遺伝子は3つのカプシド蛋白質(VP1,VP2及びVP3)の産生を担う。 The wild-type AAV genome contains two genes, rep and cap. The rep proteins (rep78, rep68, rep52, and rep40) produced by the rep gene are essential for capsid formation and mediate the integration of the viral genome into the chromosome. The cap gene is responsible for the production of three capsid proteins (VP1, VP2, and VP3).
 野生型AAVのゲノムでは,両端にITRが存在し,ITRの間にrep遺伝子及びcap遺伝子が存在する。このゲノムがカプシドに内包されてAAVビリオンが形成される。組換えAAVビリオン(rAAVビリオン)は,rep遺伝子及びcap遺伝子を含む領域を,外来の蛋白質をコードする遺伝子で置換したものが,カプシドに内包されたものである。  In the wild-type AAV genome, there are ITRs at both ends, with the rep gene and the cap gene between the ITRs. This genome is packaged in a capsid to form an AAV virion. A recombinant AAV virion (rAAV virion) is a virion in which the region containing the rep gene and the cap gene has been replaced with a gene that codes for a foreign protein, and then packaged in a capsid.
 rAAVビリオンは,外来の遺伝子を細胞に送達するためのベクターとして治療目的に用いられる。 rAAV virions are used as vectors to deliver foreign genes to cells for therapeutic purposes.
 外来の遺伝子を細胞,組織,又は生体内に導入するために用いられる組換えAAVビリオンの生産には,通常,(1)AAV等のウイルスに由来する第一の逆方向末端反復(ITR)を含む塩基配列と第二の逆方向末端反復(ITR)を含む塩基配列,及びこれら2つのITRの間に配置された所望の蛋白質をコードする遺伝子を含む構造を有するプラスミド(プラスミド1),(2)前記ITR配列に挟まれた領域(ITR配列を含む)の塩基配列を宿主細胞のゲノム中に組み込むために必要な機能を有するAAV Rep遺伝子と,AAVのカプシド蛋白質をコードする遺伝子とを含むプラスミド(プラスミド2),及び(3)アデノウイルスのE2A領域,E4領域,及びVA1 RNA領域を含むプラスミド(プラスミド3)の3種類のプラスミドが用いられる。但し,これに限らず,プラスミド1~3のうちの2つを連結させて一つのプラスミドとしたものと,残りの一つのプラスミドの2種類のプラスミドを用いることもできる。更に,プラスミド1~3を連結させて一つのプラスミドとしたものを用いることもできる。本発明の一実施形態における所望の蛋白質は,融合蛋白質と抗体との結合体である。ここで,融合蛋白質は例えば,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質であり,抗体は例えば,抗トランスフェリン受容体抗体である。 To produce recombinant AAV virions used to introduce foreign genes into cells, tissues, or living organisms, three types of plasmids are usually used: (1) a plasmid (plasmid 1) having a structure containing a base sequence including a first inverted terminal repeat (ITR) and a base sequence including a second inverted terminal repeat (ITR) derived from a virus such as AAV, and a gene encoding a desired protein arranged between these two ITRs; (2) a plasmid (plasmid 2) containing an AAV Rep gene having the function required to integrate the base sequence of the region (including the ITR sequence) sandwiched between the ITR sequences into the genome of a host cell, and a gene encoding an AAV capsid protein; and (3) a plasmid (plasmid 3) containing the E2A region, E4 region, and VA1 RNA region of adenovirus. However, this is not limited to this, and two types of plasmids, one formed by linking two of plasmids 1 to 3 and the remaining one, can also be used. Furthermore, one formed by linking plasmids 1 to 3 can also be used. In one embodiment of the present invention, the desired protein is a conjugate of a fusion protein and an antibody. Here, the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody.
 一般に,組換えAAVビリオンの生産には,まず,これら3種類のプラスミドが,アデノウイルスのE1a遺伝子とE1b遺伝子がゲノム中に組み込まれたHEK293細胞等の宿主細胞に一般的なトランスフェクション手法により導入される。そうすると第一の逆方向末端反復(ITR)を含む塩基配列と第二の逆方向末端反復(ITR)を含む塩基配列,及びこれら2つのITRの間に配置された所望の蛋白質をコードする遺伝子を含む領域が宿主細胞で複製され,生じた一重鎖DNAがAAVのカプシド蛋白質にパッケージングされて,組換えAAVビリオンが形成される。この組換えAAVビリオンは,感染力を有するので,外来の遺伝子を細胞,組織,又は生体内に導入するために用いることができる。本発明の一実施形態における外来の遺伝子は,融合蛋白質と抗体との結合体をコードする遺伝子である。導入先の細胞等ではこの遺伝子から該結合体が発現するようになる。ここで,融合蛋白質は例えば,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質であり,抗体は例えば,抗トランスフェリン受容体抗体である。 In general, to produce recombinant AAV virions, these three types of plasmids are first introduced into host cells, such as HEK293 cells, in which the adenovirus E1a and E1b genes have been incorporated into the genome, by a general transfection method. Then, a region containing a base sequence containing a first inverted terminal repeat (ITR), a base sequence containing a second inverted terminal repeat (ITR), and a gene encoding a desired protein arranged between these two ITRs is replicated in the host cell, and the resulting single-stranded DNA is packaged into the capsid protein of AAV to form a recombinant AAV virion. This recombinant AAV virion has infectivity and can be used to introduce a foreign gene into a cell, tissue, or living body. In one embodiment of the present invention, the foreign gene is a gene encoding a conjugate of a fusion protein and an antibody. In the cell or the like to which it is introduced, the conjugate is expressed from this gene. Here, the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody.
 アデノ随伴ウイルス(AAV)のRep蛋白質は,AAVのrep遺伝子にコードされる。Rep蛋白質は,例えばAAVゲノムを,当該ゲノム中に存在するITRを介して,宿主細胞のゲノム中に組み込むために必要な機能を有する。Rep蛋白質は複数のサブタイプが存在するが,AAVゲノムの宿主細胞のゲノムへの組み込みには,Rep68とRep78の2種類が必要とされる。Rep68とRep78は,同一の遺伝子から選択的スプライシングにより転写される2種類のmRNAの翻訳産物である。AAVのRep蛋白質というときは,少なくともRep68とRep78の2種類の蛋白質を含むものである。 The Rep protein of adeno-associated virus (AAV) is encoded by the AAV rep gene. The Rep protein has the necessary function of integrating, for example, the AAV genome into the genome of a host cell via the ITRs present in the genome. There are multiple subtypes of Rep proteins, but two types, Rep68 and Rep78, are required for the integration of the AAV genome into the genome of a host cell. Rep68 and Rep78 are the translation products of two types of mRNA transcribed by alternative splicing from the same gene. When we say AAV Rep protein, we mean one that includes at least the two types of proteins, Rep68 and Rep78.
 本発明の一実施形態において,アデノ随伴ウイルスのRep蛋白質をコードする塩基配列(Rep領域の塩基配列)というときは,少なくともRep68とRep78をコードする塩基配列,又はこれに変異を加えた塩基配列のことをいう。Rep蛋白質は,好ましくは血清型2のAAVのものであることが好ましいが,これに限定されることはなく,血清型1,3,4,5,6,7,8,9,10又は11の何れのものであってもよい。野生型の血清型2のAAVのRep蛋白質をコードする塩基配列の好ましい一例として配列番号123で示される塩基配列を有するものが挙げられる。 In one embodiment of the present invention, the base sequence encoding the Rep protein of an adeno-associated virus (the base sequence of the Rep region) refers to a base sequence encoding at least Rep68 and Rep78, or a base sequence with a mutation added thereto. The Rep protein is preferably that of an AAV of serotype 2, but is not limited thereto, and may be any of serotypes 1, 3, 4, 5, 6, 7, 8, 9, 10, or 11. A preferred example of a base sequence encoding the Rep protein of a wild-type AAV of serotype 2 is one having the base sequence shown in SEQ ID NO: 123.
 また,Rep68は,その機能を発揮する限り,血清型1,2,3,4,5,6,7,8,9,10又は11の何れかのAAVの野生型のRep68のアミノ酸配列に置換,欠失,付加等の改変がなされた変異体であってもよい。これら変異を加えたRep68も,Rep68に含まれる。  In addition, Rep68 may be a mutant in which the amino acid sequence of wild-type Rep68 of any of AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 has been modified by substitution, deletion, addition, or other means, so long as it exerts its function. Rep68 with these mutations is also included in Rep68.
 野生型のRep68のアミノ酸配列中のアミノ酸を他のアミノ酸で置換する場合,置換するアミノ酸の個数は,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個である。野生型のRep68のアミノ酸配列中のアミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個である。また,これらアミノ酸の置換と欠失を組み合わせた変異を加えたRep68も,Rep68である。アミノ酸を付加する場合,野生型のRep68のアミノ酸配列中若しくはN末端又はC末端に,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個のアミノ酸を付加する。これらアミノ酸の付加,置換及び欠失を組み合わせた変異を加えたRep68も,Rep68に含まれる。変異を加えたRep68のアミノ酸配列は,野生型のRep68のアミノ酸配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更に好ましくは98%以上の同一性を示す。 When amino acids in the amino acid sequence of wild-type Rep68 are replaced with other amino acids, the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3. When amino acids in the amino acid sequence of wild-type Rep68 are deleted, the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3. Rep68 with mutations that combine these amino acid substitutions and deletions is also Rep68. When amino acids are added, preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3 amino acids are added to the amino acid sequence or to the N-terminus or C-terminus of wild-type Rep68. Rep68 with mutations that combine these amino acid additions, substitutions, and deletions is also included in Rep68. The amino acid sequence of the mutated Rep68 preferably exhibits 80% or more identity to the amino acid sequence of wild-type Rep68, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
 また,Rep78は,その機能を発揮する限り,血清型1,2,3,4,5,6,7,8,9,10又は11の何れかのAAVの野生型のRep78のアミノ酸配列に置換,欠失,付加等の改変がなされた変異体であってもよい。これら変異を加えたRep78も,Rep78に含まれる。  In addition, Rep78 may be a mutant in which the amino acid sequence of wild-type Rep78 of any of AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 has been modified by substitution, deletion, addition, or other means, so long as it exerts its function. Rep78 with these mutations is also included in Rep78.
 野生型のRep78のアミノ酸配列中のアミノ酸を他のアミノ酸で置換する場合,置換するアミノ酸の個数は,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個である。野生型のRep78のアミノ酸配列中のアミノ酸を欠失させる場合,欠失させるアミノ酸の個数は,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個である。また,これらアミノ酸の置換と欠失を組み合わせた変異を加えたRep78も,Rep78である。アミノ酸を付加する場合,野生型のRep78のアミノ酸配列中若しくはN末端又はC末端に,好ましくは1~10個,より好ましくは1~5個,更に好ましくは1~3個のアミノ酸を付加する。これらアミノ酸の付加,置換及び欠失を組み合わせた変異を加えたRep78も,Rep78に含まれる。変異を加えたRep78のアミノ酸配列は,野生型のRep78のアミノ酸配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更に好ましくは98%以上の同一性を示す。 When amino acids in the amino acid sequence of wild-type Rep78 are replaced with other amino acids, the number of amino acids to be replaced is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3. When amino acids in the amino acid sequence of wild-type Rep78 are deleted, the number of amino acids to be deleted is preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3. Rep78 with mutations that combine these amino acid substitutions and deletions is also Rep78. When amino acids are added, preferably 1 to 10, more preferably 1 to 5, and even more preferably 1 to 3 amino acids are added to the amino acid sequence or to the N-terminus or C-terminus of wild-type Rep78. Rep78 with mutations that combine these amino acid additions, substitutions, and deletions is also included in Rep78. The amino acid sequence of the mutated Rep78 preferably exhibits 80% or more identity to the amino acid sequence of wild-type Rep78, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
 AAVのRep蛋白質の機能的等価物とは,Rep68にあっては,機能的にRep68に代えて用いることができるものをいい,Rep78にあっては,機能的にRep78に代えて用いることができるものをいう。Rep蛋白質の機能的等価物は,野生型のRep蛋白質の変異体であってもよい。 The functional equivalent of the AAV Rep protein refers to a substance that can be used functionally in place of Rep68, and to a substance that can be used functionally in place of Rep78. The functional equivalent of the Rep protein may be a mutant of the wild-type Rep protein.
 配列番号123で示される塩基配列は,機能的なRep68及びRep78をコードするものである限り,置換,欠失,付加等の改変を加えることができる。配列番号123で示される塩基配列中の塩基を他の塩基で置換する場合,置換する塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個である。配列番号123で示される塩基配列中の塩基を欠失させる場合,欠失させる塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個である。また,これら塩基の置換と欠失を組み合わせた変異を加えたものも,Rep蛋白質をコードする塩基配列として使用できる。塩基を付加する場合,配列番号123で示される塩基配列中若しくは5’末端又は3’末端に,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個の塩基を付加する。これら塩基の付加,置換及び欠失を組み合わせた変異を加えたものも,Rep蛋白質をコードする核酸分子として使用できる。変異を加えた塩基配列は,配列番号123で示される塩基配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更に好ましくは98%以上の同一性を示す。但し,これら変異を加える場合において,Rep蛋白質のRep68とRep78とをコードする遺伝子の開始コドンをACGとすることが好ましい。 The base sequence shown in SEQ ID NO:123 can be modified by substitution, deletion, addition, etc., so long as it encodes functional Rep68 and Rep78. When a base in the base sequence shown in SEQ ID NO:123 is replaced with another base, the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. When a base in the base sequence shown in SEQ ID NO:123 is deleted, the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. Furthermore, a mutation that combines the substitution and deletion of these bases can also be used as a base sequence encoding a Rep protein. When a base is added, preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3 bases are added to the base sequence shown in SEQ ID NO:123 or to the 5' end or 3' end. A mutation that combines the addition, substitution, and deletion of these bases can also be used as a nucleic acid molecule encoding a Rep protein. The mutated base sequence preferably exhibits 80% or more identity, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity to the base sequence shown in SEQ ID NO: 123. However, when these mutations are added, it is preferable that the start codon of the genes encoding the Rep proteins Rep68 and Rep78 is ACG.
 本発明の一実施形態においてアデノ随伴ウイルスのCap蛋白質をコードする塩基配列(Cap領域の塩基配列)というときは,少なくともAAVのカプシドを構成する蛋白質の一種であるVP1をコードする塩基配列,又はこれに変異を加えた塩基配列を含む塩基配列のことをいう。VP1は,好ましくは血清型8のAAVのものであることが好ましいが,これに限定されることはなく,血清型1,2,3,4,5,6,7,8,9,10又は11の何れのものであってもよい。血清型8のCap領域の塩基配列の好適例として,配列番号124で示される塩基配列を含むものが挙げられる。 In one embodiment of the present invention, the base sequence encoding the Cap protein of an adeno-associated virus (base sequence of the Cap region) refers to a base sequence that encodes at least VP1, a protein that constitutes the capsid of AAV, or a base sequence that includes a mutated base sequence thereof. VP1 is preferably that of AAV serotype 8, but is not limited thereto, and may be any of serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11. A suitable example of a base sequence of the Cap region of serotype 8 is one that includes the base sequence shown in SEQ ID NO: 124.
 また,VP1は,その機能を発揮する限り,血清型1,2,3,4,5,6,7,8,9,10又は11の何れかのAAVの野生型のVP1のアミノ酸配列に置換,欠失,付加等の改変がなされたものであってもよい。本発明の一実施形態において,これら変異を加えたVP1も,VP1に含まれる。 In addition, VP1 may be modified by substitution, deletion, addition, or other alteration to the amino acid sequence of wild-type VP1 of any of AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11, so long as it exerts its function. In one embodiment of the present invention, VP1 with these mutations is also included in VP1.
 配列番号124で示される塩基配列中の塩基を他の塩基で置換する場合,置換する塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個である。配列番号124で示される塩基配列中の塩基を欠失させる場合,欠失させる塩基の個数は,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個である。また,これら塩基の置換と欠失を組み合わせた変異を加えたものも,Cap蛋白質をコードする塩基配列として使用できる。塩基を付加する場合,配列番号124で示される塩基配列中若しくは5’末端又は3’末端に,好ましくは1~20個,より好ましくは1~10個,更に好ましくは1~3個の塩基を付加する。これら塩基の付加,置換及び欠失を組み合わせた変異を加えたものも,Cap蛋白質をコードする核酸分子として使用できる。変異を加えた塩基配列は,配列番号124で示される塩基配列と,好ましくは80%以上の同一性を示し,より好ましくは85%以上の同一性を示し,更に好ましくは90%以上の同一性を示し,更に好ましくは,95%以上の同一性を示し,更により好ましくは98%以上の同一性を示す。 When bases in the base sequence shown in SEQ ID NO: 124 are replaced with other bases, the number of bases to be replaced is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. When bases in the base sequence shown in SEQ ID NO: 124 are deleted, the number of bases to be deleted is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3. Furthermore, a mutation that combines the substitution and deletion of these bases can also be used as a base sequence encoding a Cap protein. When bases are added, preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 3 bases are added to the base sequence shown in SEQ ID NO: 124 or to the 5' end or 3' end. A mutation that combines the addition, substitution, and deletion of these bases can also be used as a nucleic acid molecule encoding a Cap protein. The mutated base sequence preferably exhibits 80% or more identity to the base sequence shown in SEQ ID NO:124, more preferably 85% or more identity, even more preferably 90% or more identity, even more preferably 95% or more identity, and even more preferably 98% or more identity.
 AAVベクターにより,第一のITRと第二のITRとの間に外来の遺伝子を含む核酸分子がパッケージングされた組換えAAVビリオンを得ることができる。組換えAAVビリオンは,感染力を有するので,外来の遺伝子を細胞,組織,又は生体内に導入するために用いることができる。本発明の一実施形態における外来の遺伝子は,融合蛋白質と抗体との結合体をコードする遺伝子である。ここで,融合蛋白質は例えば,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質であり,抗体は例えば,抗トランスフェリン受容体抗体である。当該遺伝子が導入された細胞等ではこの遺伝子から融合蛋白質が発現するようになる。第一のITRと第二のITRとの間に外来の遺伝子を含む核酸分子がパッケージングされた組換えビリオンであって,細胞等に遺伝子を導入するために用いることができるものとして,AAVベクター系以外に,アデノウイルスベクター系がある。アデノウイルスベクター系においては,第一のITRと第二のITRとの間に外来の遺伝子を含む核酸分子がパッケージングされた組換えアデノウイルスビリオンが得られる。組換えアデノウイルスビリオンは,感染力を有するので,外来の遺伝子を細胞,組織,又は生体内に導入するために用いることができる。 The AAV vector can produce a recombinant AAV virion in which a nucleic acid molecule containing a foreign gene is packaged between the first and second ITRs. The recombinant AAV virion has infectivity and can be used to introduce a foreign gene into cells, tissues, or living organisms. In one embodiment of the present invention, the foreign gene is a gene encoding a conjugate of a fusion protein and an antibody. Here, the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody. In cells, etc., into which the gene has been introduced, the fusion protein is expressed from this gene. In addition to the AAV vector system, there is an adenovirus vector system, which is a recombinant virion in which a nucleic acid molecule containing a foreign gene is packaged between the first and second ITRs and can be used to introduce a gene into cells, etc. In the adenovirus vector system, a recombinant adenovirus virion is obtained in which a nucleic acid molecule containing a foreign gene is packaged between the first and second ITRs. The recombinant adenovirus virion has infectivity and can be used to introduce a foreign gene into a cell, tissue, or living organism.
 レンチウイルスはレトロウイルス科の亜科の一つであるオルトレトロウイルス亜科のうちレンチウイルス属に属するウイルスであり,一本鎖の(+)鎖RNAゲノム(ssRNA)を有する。レンチウイルスのゲノムは必須遺伝子としてgag(カプシド蛋白質を含む構造蛋白質をコードする領域),pol(逆転写酵素を含む酵素群をコードする領域),env(宿主細胞への結合に必要なエンベロープ蛋白質をコードする領域)を含み,これらが2つのLTR(5’LTR及び3’LTR)に挟まれている。またこれらに加え,レンチウイルスのゲノムは,補助遺伝子として,Rev(ウイルスRNA内に存在するRRE(rev responsive element)に結合してウイルスRNAを核から細胞質に輸送する機能を有する蛋白質をコードする領域),tat(5’LTR内にあるTARに結合してLTRのプロモーター活性を上昇させる機能を有する蛋白質をコードする領域),その他vif,vpr,vpu,nef等を含む。 Lentiviruses are viruses that belong to the Lentivirus genus in the Orthoretrovirinae subfamily of the Retroviridae family, and have a single-stranded (+) strand RNA genome (ssRNA). The genome of lentiviruses contains essential genes, gag (a region that codes for structural proteins including capsid proteins), pol (a region that codes for a group of enzymes including reverse transcriptase), and env (a region that codes for envelope proteins required for binding to host cells), which are sandwiched between two LTRs (5'LTR and 3'LTR). In addition to these, the genome of lentiviruses contains auxiliary genes, such as Rev (a region that codes for a protein that binds to the RRE (rev responsive element) present in the viral RNA and transports the viral RNA from the nucleus to the cytoplasm), tat (a region that codes for a protein that binds to the TAR in the 5'LTR and increases the promoter activity of the LTR), and others, such as vif, vpr, vpu, and nef.
 レンチウイルスはエンベロープウイルスであり,エンベロープが細胞膜と融合することで細胞に感染する。また,レンチウイルスはRNAウイルスであり,ビリオン中には逆転写酵素が存在する。レンチウイルスの感染後,逆転写酵素により,(+)鎖RNAゲノムから一本鎖のプラス鎖DNAが複製され,更に二重鎖DNAが合成される。この二重鎖DNAからビリオンの構成成分である蛋白質が発現し,これに(+)鎖RNAゲノムがパッケージングされることによりビリオンが増殖する。本発明の一実施形態において,レンチウイルスベクターは,レンチウイルスの一種であるHIV-1のゲノムに基づき開発されたものがあるが,これに限られるものではない。  Lentiviruses are enveloped viruses that infect cells by fusing the envelope with the cell membrane. Lentiviruses are also RNA viruses, and reverse transcriptase is present in virions. After lentivirus infection, single-stranded plus-strand DNA is replicated from the (+) strand RNA genome by reverse transcriptase, and double-stranded DNA is then synthesized. Proteins that are components of virions are expressed from this double-stranded DNA, and the (+) strand RNA genome is packaged into this to cause virions to proliferate. In one embodiment of the present invention, the lentivirus vector has been developed based on the genome of HIV-1, a type of lentivirus, but is not limited to this.
 第一世代レンチウイルスベクターは,パッケージングプラスミド,Envプラスミド,及び導入プラスミドの3種のプラスミドからなる。パッケージングプラスミドはCMVプロモーター等の制御下にgag及びpolの各遺伝子を有する。EnvプラスミドはCMVプロモーター制御下にenv遺伝子を有する。導入プラスミドは,5’LTR,RRE,CMVプロモーター制御下に所望の蛋白質をコードする遺伝子,及び3’LTRを有する。これらを宿主細胞に一般的なトランスフェクション手法により導入することで,第一のLTRと第二のLTRとの間に外来の遺伝子を含む核酸分子がカプシド蛋白質中にパッケージングされた組換えビリオンが得られる。第一世代レンチウイルスベクターのパッケージングプラスミドには,ウイルス由来のrev,tat,vif,vpr,vpu,及びnefの各補助遺伝子も含まれる。ここにおいて本発明の一実施形態における所望の蛋白質は,融合蛋白質と抗体との結合体である。ここで,融合蛋白質は例えば,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質であり,抗体は例えば,抗トランスフェリン受容体抗体である。なお,所望の蛋白質をコードする遺伝子を制御するプロモーターをCMVプロモーター以外のプロモーター,例えばSV40初期プロモーター,ヒト伸長因子-1α(EF-1α)プロモーター,ヒトユビキチンCプロモーター,レトロウイルスのラウス肉腫ウイルスLTRプロモーター,ジヒドロ葉酸還元酵素プロモーター,β-アクチンプロモーター,ホスホグリセリン酸キナーゼ(PGK)プロモーター,マウスアルブミンプロモーター,ヒトアルブミンプロモーター,及びヒトα-1アンチトリプシンプロモーターを含むものが好適である。例えば,マウスαフェトプロテインエンハンサーの下流にマウスアルブミンプロモーターを含む配列番号121で示される塩基配列を有する合成プロモーター(マウスαフェトプロテインエンハンサー/マウスアルブミンプロモーター)等とすることもできる。 A first-generation lentiviral vector consists of three types of plasmids: a packaging plasmid, an Env plasmid, and a transfer plasmid. The packaging plasmid has the gag and pol genes under the control of a CMV promoter, etc. The Env plasmid has the env gene under the control of a CMV promoter. The transfer plasmid has a 5'LTR, an RRE, a gene encoding a desired protein under the control of a CMV promoter, and a 3'LTR. By introducing these into a host cell by a general transfection method, a recombinant virion is obtained in which a nucleic acid molecule containing a foreign gene between the first LTR and the second LTR is packaged in the capsid protein. The packaging plasmid of the first-generation lentiviral vector also contains the auxiliary genes rev, tat, vif, vpr, vpu, and nef derived from the virus. Here, the desired protein in one embodiment of the present invention is a conjugate of a fusion protein and an antibody. Here, the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody. Note that the promoter controlling the gene encoding the desired protein is preferably a promoter other than the CMV promoter, such as an SV40 early promoter, a human elongation factor-1α (EF-1α) promoter, a human ubiquitin C promoter, a Rous sarcoma virus LTR promoter of a retrovirus, a dihydrofolate reductase promoter, a β-actin promoter, a phosphoglycerate kinase (PGK) promoter, a mouse albumin promoter, a human albumin promoter, and a human α-1 antitrypsin promoter. For example, it can be a synthetic promoter having the base sequence shown in SEQ ID NO: 121, which includes a mouse albumin promoter downstream of a mouse alpha-fetoprotein enhancer (mouse alpha-fetoprotein enhancer/mouse albumin promoter), etc.
 第二世代レンチウイルスベクターも,第一世代と同じくパッケージングプラスミド,Envプラスミド(エンベローププラスミド),及び導入プラスミドの3種のプラスミドからなる。但し,パッケージングプラスミドから,必須遺伝子でないvif,vpr,vpu,及びnefの各補助遺伝子は削除されている。 Second-generation lentiviral vectors, like the first-generation, consist of three plasmids: a packaging plasmid, an Env plasmid (envelope plasmid), and a transfer plasmid. However, the non-essential auxiliary genes vif, vpr, vpu, and nef have been deleted from the packaging plasmid.
 第三世代レンチウイルスベクターは,パッケージングプラスミド,Envプラスミド(エンベローププラスミド),Revプラスミド,及び導入プラスミドの4種のプラスミドからなる。第三世代では,第二世代でパッケージングプラスミドにあったrevを独立させてRevプラスミドとしている。また,パッケージングプラスミドからtatが削除されている。更に,導入プラスミドの5’LTR内のTARがCMVプロモーターに置き換えられている。 Third-generation lentiviral vectors consist of four types of plasmids: packaging plasmid, Env plasmid (envelope plasmid), Rev plasmid, and transfer plasmid. In the third generation, the rev that was in the packaging plasmid in the second generation has been separated to form the Rev plasmid. In addition, tat has been deleted from the packaging plasmid. Furthermore, the TAR in the 5'LTR of the transfer plasmid has been replaced with a CMV promoter.
 組換えビリオンは,感染力を有するので,外来の遺伝子を細胞,組織,又は生体内に導入するために用いることができる。本発明の一実施形態における外来の遺伝子は,融合蛋白質と抗体との結合体をコードする遺伝子である。当該遺伝子が導入された細胞等ではこの遺伝子から結合体が発現するようになる。ここで,融合蛋白質は例えば,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質であり,抗体は例えば,抗トランスフェリン受容体抗体である。 Since recombinant virions have infectivity, they can be used to introduce foreign genes into cells, tissues, or living organisms. In one embodiment of the present invention, the foreign gene is a gene encoding a conjugate of a fusion protein and an antibody. In cells, etc., into which the gene is introduced, the conjugate is expressed from this gene. Here, the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody.
 レトロウイルスは,一本鎖の(+)鎖RNAゲノム(ssRNA)を有する。ウイルスゲノムには,gag(カプシド蛋白質を含む構造蛋白質をコード),pol(逆転写酵素を含む酵素群をコード),env(宿主細胞への結合に必要なエンベロープ蛋白質をコード)及びパッケージングシグナル(Ψ)の各遺伝子がコードされており,これらが2つの長い末端反復(LTR,5’LTR及び3’LTR)に挟まれている。レトロウイルスは宿主細胞に感染すると,(+)鎖RNA及び逆転写酵素が細胞内に移行し,2本鎖DNAへと逆転写される。 Retroviruses have a single-stranded (+) strand RNA genome (ssRNA). The viral genome encodes the gag (encoding structural proteins including capsid protein), pol (encoding a group of enzymes including reverse transcriptase), env (encoding envelope proteins required for binding to host cells) and packaging signal (Ψ) genes, which are flanked by two long terminal repeats (LTR, 5'LTR and 3'LTR). When a retrovirus infects a host cell, the (+) strand RNA and reverse transcriptase are transferred into the cell and reverse transcribed into double-stranded DNA.
 レトロウイルスベクターは主にマウス白血病ウイルスに基づき開発され,病原性を失わせ安全性を高めることを目的として,その感染力を保ちつつ自己複製能を欠損させるためにウイルスゲノムが分割されている。第1世代レトロウイルスベクターは,パッケージングプラスミド(パッケージングシグナルを除いたウイルスゲノム)と導入プラスミド(パッケージングシグナル,gagの一部,外来遺伝子の両端を5’LTR及び3’LTRで挟んだもの)からなる。したがって,パッケージングプラスミドと導入プラスミドに共通して存在するgag配列部分で相同組み換えが起こると自己複製可能なレトロウイルス,すなわちRC(Replication competent)ウイルスが出現する。第2世代レトロウイルスベクターでは,第1世代のパッケージングプラスミドの3'側のLTRをポリA付加シグナルに置換している。これにより,RCウイルスが発生するためにはgag配列及びポリA付加シグナル上流の2か所で同時に相同組み換えが起こる必要があり,その発生確率は非常に低く,安全性が高められている。第3世代は3つのプラスミドから構成されており,第2世代のパッケージングプラスミドがさらにgag/polをコードするプラスミドとenvをコードするプラスミドに分割されている。これにより,RCウイルスが発生するためには3か所で同時に相同組み換えが生じる必要があり,その発生確率は極めて低く,さらに安全性が高められている。 Retroviral vectors are mainly developed based on mouse leukemia viruses, and the viral genome is divided to lose the ability to self-replicate while maintaining its infectivity in order to eliminate pathogenicity and increase safety. The first generation retroviral vector consists of a packaging plasmid (the viral genome excluding the packaging signal) and an introduction plasmid (the packaging signal, a part of gag, and a foreign gene flanked by 5'LTR and 3'LTR at both ends). Therefore, when homologous recombination occurs in the gag sequence portion that is common to the packaging plasmid and the introduction plasmid, a self-replicating retrovirus, i.e., an RC (replication competent) virus, appears. In the second generation retroviral vector, the 3' LTR of the first generation packaging plasmid is replaced with a polyA addition signal. As a result, for an RC virus to be generated, homologous recombination must occur simultaneously at two locations, the gag sequence and the upstream of the polyA addition signal, and the probability of this occurring is extremely low, increasing safety. The third generation is composed of three plasmids, and the second generation packaging plasmid is further divided into a plasmid encoding gag/pol and a plasmid encoding env. This means that for an RC virus to emerge, homologous recombination must occur simultaneously at three sites, making the probability of this occurring extremely low, further increasing safety.
 一般に,組換えレトロウイルスビリオンの生産には,まず,これらパッケージングプラスミドと導入プラスミドが,宿主細胞に一般的なトランスフェクション手法により導入される。そうすると5’LTR及び3’LTR,及びこれら2つのLTRの間に配置された所望の蛋白質をコードする遺伝子を含む領域が宿主細胞で複製され,生じた一本鎖の(+)鎖RNAがレトロウイルスのカプシド蛋白質にパッケージングされて,組換えレトロウイルスビリオンが形成される。この組換えレトロウイルスビリオンは,感染力を有するので,外来の遺伝子を細胞,組織,又は生体内に導入するために用いることができる。本発明の一実施形態における外来の遺伝子は,融合蛋白質と抗体との結合体をコードする遺伝子である。当該遺伝子が導入された細胞等ではこの遺伝子から結合体が発現するようになる。ここで,融合蛋白質は例えば,HSAとhGALCとの融合蛋白質,HSAとhGBAとの融合蛋白質,HSAとhIL-10との融合蛋白質,HSAとhBDNFとの融合蛋白質,HSAとhNGFとの融合蛋白質,HSAとhNT-3との融合蛋白質,又はHSAとhNT-4との融合蛋白質であり,抗体は例えば,抗トランスフェリン受容体抗体である。 In general, to produce recombinant retroviral virions, these packaging plasmids and transfer plasmids are first transferred into host cells by a general transfection method. Then, the region containing the 5'LTR and 3'LTR and the gene encoding the desired protein located between these two LTRs is replicated in the host cell, and the resulting single-stranded (+) strand RNA is packaged in the retroviral capsid protein to form a recombinant retroviral virion. This recombinant retroviral virion has infectivity and can be used to transfer a foreign gene into a cell, tissue, or living body. In one embodiment of the present invention, the foreign gene is a gene encoding a conjugate of a fusion protein and an antibody. In cells, etc., into which the gene has been introduced, the conjugate is expressed from this gene. Here, the fusion protein is, for example, a fusion protein of HSA and hGALC, a fusion protein of HSA and hGBA, a fusion protein of HSA and hIL-10, a fusion protein of HSA and hBDNF, a fusion protein of HSA and hNGF, a fusion protein of HSA and hNT-3, or a fusion protein of HSA and hNT-4, and the antibody is, for example, an anti-transferrin receptor antibody.
 本発明の一実施形態において,核酸分子は,これをリポソーム,脂質ナノ粒子(Lipid Nanoparticle;LNP)等に内包させた形態とすることもできる。リポソームとは,脂質二重層を持つ球状の小胞をいい,主にリン脂質,特にホスファチジルコリンから構成される。但しこれに限らず,脂質二重層を形成する限り,リポソームは卵黄ホスファチジルエタノールアミン等の他の脂質を加えられたものであっても良い。細胞膜は主にリン脂質二重膜から構成されているため,リポソームは生体適合性に優れるという利点がある。脂質ナノ粒子は,直径10 nm~1000 nm,典型的には約200 nm未満の,脂質を主成分とする粒子をいい,疎水性(親油性)分子を内包させることができ,トリグリセリド,ジグリセリド,モノグリセリド,脂肪酸,ステロイド等の生体適合性のある脂質を主な構成成分とする。リポソーム又は脂質ナノ粒子に内包させた遺伝子を生体内に投与すると,細胞の細胞膜と直接融合,又はエンドサイトーシス等により細胞に取り込まれ,その後核に移行して遺伝子が細胞に導入されると考えられている。リポソーム又は脂質ナノ粒子による遺伝子導入方法は,ウイルスベクターを用いた遺伝子導入と比較して,導入する遺伝子の大きさに制限が無い点や,安全性が高い点において優れている。本発明の核酸分子を内包させたリポソーム,脂質ナノ粒子等は,融合蛋白質と抗体との結合体の遺伝子を細胞,組織,又は生体内に導入するために用いることができる。当該遺伝子が導入された細胞等ではこの遺伝子から融合蛋白質が発現するようになる。本明細において「リポソーム,脂質ナノ粒子等」というときは,上述のリポソーム及び脂質ナノ粒子に加え,ポリマーナノ粒子,ミセル,エマルジョン,ナノエマルジョン,マイクロスフェア,ナノスフェア,マイクロカプセル,ナノカプセル,デンドリマー,ナノゲル,金属ナノ粒子,その他薬物送達システム(DDS)として用いられることのできるあらゆるナノ・マイクロ粒子を含むものとする。 In one embodiment of the present invention, the nucleic acid molecule may be encapsulated in a liposome, lipid nanoparticle (LNP), or the like. A liposome is a spherical vesicle with a lipid bilayer, and is composed mainly of phospholipids, particularly phosphatidylcholine. However, the present invention is not limited to this, and liposomes may contain other lipids, such as egg yolk phosphatidylethanolamine, as long as they form a lipid bilayer. Since cell membranes are mainly composed of phospholipid bilayers, liposomes have the advantage of being highly biocompatible. A lipid nanoparticle is a particle with a diameter of 10 nm to 1000 nm, typically less than about 200 nm, that is mainly composed of lipids, and can encapsulate hydrophobic (lipophilic) molecules and is mainly composed of biocompatible lipids, such as triglycerides, diglycerides, monoglycerides, fatty acids, and steroids. It is believed that when a gene encapsulated in liposomes or lipid nanoparticles is administered to a living body, it fuses directly with the cell membrane of the cell or is taken up by endocytosis, etc., and then migrates to the nucleus and is introduced into the cell. The gene introduction method using liposomes or lipid nanoparticles is superior to gene introduction using a viral vector in that there is no limit to the size of the gene to be introduced and that it is highly safe. The liposomes, lipid nanoparticles, etc. encapsulating the nucleic acid molecule of the present invention can be used to introduce a gene of a conjugate of a fusion protein and an antibody into a cell, tissue, or living body. In the cell, etc. into which the gene is introduced, the fusion protein is expressed from the gene. In this specification, the term "liposomes, lipid nanoparticles, etc." includes not only the above-mentioned liposomes and lipid nanoparticles, but also polymer nanoparticles, micelles, emulsions, nanoemulsions, microspheres, nanospheres, microcapsules, nanocapsules, dendrimers, nanogels, metal nanoparticles, and any other nano-microparticles that can be used as a drug delivery system (DDS).
 本発明の一実施形態において,プラスミドの形態,組換ウイルスビリオンに内包された形態,又はリポソーム,脂質ナノ粒子等に内包された形態で細胞,組織,又は生体内に導入された核酸分子の挙動について,以下に(1)~(6)に例示する。但し,核酸分子の挙動はこれらに限られるものではない。
(1)一実施形態において,核酸分子は一本鎖の(+)鎖RNAであり,細胞内に導入されると,核酸分子に含まれる融合蛋白質と抗体との結合体をコードする遺伝子が翻訳されて該結合体が発現する。
(2)一実施形態において,核酸分子は一本鎖の(+)鎖RNAであり,細胞内に導入されると,該核酸分子が逆転写されて一本鎖の(+)鎖DNAとなり,次いでこのDNAが転写,翻訳されて該結合体が発現する。
(3)一実施形態において,核酸分子は一本鎖の(+)鎖RNA又は(-)鎖RNAであり,細胞内に導入されると,該核酸分子が逆転写されて二重鎖DNAとなり,次いでこのDNAが転写,翻訳されて該結合体が発現する。
(4)一実施形態において,核酸分子は一本鎖の(+)鎖又は(-)RNAであり,細胞内に導入されると,該核酸分子が逆転写されて二重鎖DNAとなり,次いでこのDNAが宿主細胞のゲノムとランダム組換え又は相同組換えを起こしてゲノム内に組込まれ,組み込まれたDNAが転写,翻訳されて該結合体が発現する。
(5)一実施形態において,核酸分子は一本鎖の(+)鎖DNAであり,細胞内に導入されると,該核酸分子が転写,翻訳されて該結合体が発現する。
(6)一実施形態において,核酸分子は二重鎖DNAであり,細胞内に導入されると,該核酸分子が転写,翻訳されて該結合体が発現する。
In one embodiment of the present invention, the behavior of a nucleic acid molecule introduced into a cell, tissue, or living body in the form of a plasmid, encapsulated in a recombinant virus virion, or encapsulated in a liposome, lipid nanoparticle, or the like is exemplified below as (1) to (6). However, the behavior of the nucleic acid molecule is not limited to these.
(1) In one embodiment, the nucleic acid molecule is a single-stranded (+) strand RNA, and when introduced into a cell, a gene encoding a conjugate between a fusion protein and an antibody contained in the nucleic acid molecule is translated, thereby expressing the conjugate.
(2) In one embodiment, the nucleic acid molecule is a single-stranded (+) strand RNA, which, when introduced into a cell, is reverse transcribed to form a single-stranded (+) strand DNA, which is then transcribed and translated to express the conjugate.
(3) In one embodiment, the nucleic acid molecule is a single-stranded (+) strand RNA or (-) strand RNA, and when introduced into a cell, the nucleic acid molecule is reverse transcribed to form double-stranded DNA, which is then transcribed and translated to express the conjugate.
(4) In one embodiment, the nucleic acid molecule is a single-stranded (+) or (-) RNA, and when introduced into a cell, the nucleic acid molecule is reverse transcribed to form double-stranded DNA, which then undergoes random or homologous recombination with the genome of the host cell and is integrated into the genome, and the integrated DNA is transcribed and translated to express the conjugate.
(5) In one embodiment, the nucleic acid molecule is a single-stranded (+) strand DNA, and when introduced into a cell, the nucleic acid molecule is transcribed and translated to express the conjugate.
(6) In one embodiment, the nucleic acid molecule is double-stranded DNA, and when introduced into a cell, the nucleic acid molecule is transcribed and translated to express the conjugate.
 プラスミドの形態,組換ウイルスビリオンに内包された形態,又はリポソーム,脂質ナノ粒子等に内包された形態の核酸分子は,細胞,組織,又は生体内に導入することができる。 Nucleic acid molecules in the form of plasmids, encapsulated in recombinant viral virions, or encapsulated in liposomes, lipid nanoparticles, etc. can be introduced into cells, tissues, or organisms.
 核酸分子の導入先が生体内である場合,核酸分子は,プラスミドの形態,組換ウイルスビリオンに内包された形態,又はリポソーム,脂質ナノ粒子等に内包された形態で,皮下注射,筋肉内注射,静脈内注射等の非経口的手段により投与される。 When the nucleic acid molecule is to be introduced into a living body, the nucleic acid molecule is administered parenterally, such as subcutaneous injection, intramuscular injection, or intravenous injection, in the form of a plasmid, encapsulated in a recombinant virus virion, or encapsulated in a liposome or lipid nanoparticle.
 核酸分子の導入先が細胞である場合,細胞の種類に特に限定はないが,例えば,間葉系幹細胞,歯髄由来幹細胞,造血幹細胞,胚性幹細胞,内皮幹細胞,乳腺幹細胞,腸幹細胞,肝幹細胞,膵幹細胞,神経幹細胞,及びiPS細胞である。 When the nucleic acid molecule is introduced into a cell, there is no particular limitation on the type of cell, but examples include mesenchymal stem cells, dental pulp-derived stem cells, hematopoietic stem cells, embryonic stem cells, endothelial stem cells, mammary stem cells, intestinal stem cells, hepatic stem cells, pancreatic stem cells, neural stem cells, and iPS cells.
 核酸分子が導入された細胞は,融合蛋白質と抗体との結合体を発現するようになるので,これを治療目的で患者に移植することができる。 Cells that have been transfected with the nucleic acid molecule will express the fusion protein-antibody conjugate, which can then be transplanted into a patient for therapeutic purposes.
 以下,実施例を参照して本発明を更に詳細に説明するが,本発明が実施例に限定されることは意図しない。 The present invention will be explained in more detail below with reference to examples, but it is not intended that the present invention be limited to these examples.
 以下,実施例1~13はHSAとhGALCとの融合蛋白質に関する実験結果である。 The following Examples 1 to 13 show the experimental results of a fusion protein of HSA and hGALC.
〔実施例1〕野生型hGALC発現プラスミド,HSA-hGALC発現プラスミド及びhGALC-HSA発現プラスミドの作成
 配列番号1で示されるアミノ酸配列を有する野生型hGALCをコードする遺伝子を含む,配列番号2で示される塩基配列を有するDNA断片を合成した。これをMluI及びNotI(タカラバイオ社)で制限酵素処理し,アガロースゲル電気泳動にて分離した。EtBr染色を行った後,UV照射下で目的のDNA断片を含むバンドを切り出し,QIAEX II Gel Extraction Kit(QIAGEN社)を用いてゲルからDNAを抽出した。同様に,pCI-neoベクター(プロメガ社)をMluI及びNotIで制限酵素処理し,ゲル抽出,精製を行った。制限酵素で処理した各ベクターに,制限酵素で処理した各DNA断片をそれぞれ混合し,Ligation high Ver.2(東洋紡社)を用いて16℃で30~60分間ライゲーション反応を行い,DNA断片をpCIベクターに組込んだ。野生型HSA(配列番号3)のC末端と野生型hGALC(配列番号1)のN末端とをリンカー配列Gly-Serを介して結合させた融合蛋白質である,配列番号5で示されるHSA-hGALCをコードする遺伝子を含む,配列番号6で示される塩基配列を含むDNA断片,及び,野生型hGALC(配列番号1)のC末端と野生型HSA(配列番号3)のN末端とをリンカー配列Gly-Serを介して結合させた融合蛋白質である,配列番号7で示されるhGALC-HSAをコードする遺伝子を含む,配列番号8で示される塩基配列を含むDNA断片についても,それぞれ同様に,制限酵素処理,抽出,精製した後にライゲーション反応を行い,各DNA断片をpCIベクターに組込んだ。
Example 1: Preparation of wild-type hGALC expression plasmid, HSA-hGALC expression plasmid, and hGALC-HSA expression plasmid A DNA fragment having the base sequence shown in SEQ ID NO: 2, including a gene encoding wild-type hGALC having the amino acid sequence shown in SEQ ID NO: 1, was synthesized. This was treated with restriction enzymes MluI and NotI (Takara Bio Inc.) and separated by agarose gel electrophoresis. After EtBr staining, a band containing the target DNA fragment was excised under UV irradiation, and DNA was extracted from the gel using a QIAEX II Gel Extraction Kit (QIAGEN). Similarly, pCI-neo vector (Promega Corp.) was treated with restriction enzymes MluI and NotI, and gel extraction and purification were performed. Each DNA fragment treated with a restriction enzyme was mixed with each vector treated with a restriction enzyme, and a ligation reaction was performed at 16°C for 30 to 60 minutes using Ligation high Ver.2 (Toyobo Co., Ltd.), and the DNA fragment was inserted into the pCI vector. A DNA fragment containing the base sequence shown in SEQ ID NO: 6 and including a gene encoding HSA-hGALC shown in SEQ ID NO: 5, which is a fusion protein in which the C-terminus of wild-type HSA (SEQ ID NO: 3) and the N-terminus of wild-type hGALC (SEQ ID NO: 1) are linked via the linker sequence Gly-Ser, and a DNA fragment containing the base sequence shown in SEQ ID NO: 8 and including a gene encoding hGALC-HSA shown in SEQ ID NO: 7, which is a fusion protein in which the C-terminus of wild-type hGALC (SEQ ID NO: 1) and the N-terminus of wild-type HSA (SEQ ID NO: 3) are linked via the linker sequence Gly-Ser, were similarly treated with restriction enzymes, extracted, purified, and then ligated, and each DNA fragment was inserted into the pCI vector.
 次いで,各ライゲーション反応液を用いて大腸菌(E. coli DH5α Competent Cells,タカラバイオ社)をそれぞれ形質転換した。得られた形質転換体が目的のプラスミドDNAを保持しているか確認するため,シングルコロニーをLB液体培地(LB Broth,Sigma-Aldrich社)で一晩培養し,FastGene Plasmid Mini Kit(日本ジェネティクス社)を用いて菌体からプラスミドDNAを精製した。精製したプラスミドDNAをMluI及びNotIで制限酵素処理し,アガロースゲル電気泳動にて分離することで,目的のインサートDNAが挿入されていることを確認した。野生型hGALC,HSA-hGALC,及びhGALC-HSAが組込まれていることを確認した各プラスミドを常法によりそれぞれ精製した。 Escherichia coli (E. coli DH5α Competent Cells, Takara Bio) was then transformed using each ligation reaction mixture. To confirm whether the obtained transformants retained the desired plasmid DNA, single colonies were cultured overnight in LB liquid medium (LB Broth, Sigma-Aldrich), and plasmid DNA was purified from the cells using FastGene Plasmid Mini Kit (Nihon Genetics). The purified plasmid DNA was subjected to restriction enzyme treatment with MluI and NotI, and separated by agarose gel electrophoresis to confirm that the desired insert DNA had been inserted. Plasmids that were confirmed to contain wild-type hGALC, HSA-hGALC, and hGALC-HSA were purified using standard methods.
〔実施例2〕野生型hGALC,HSA-hGALC,及びhGALC-HSAの一過性発現
 野生型hGALC,HSA-hGALC,及びhGALC-HSAの一過性発現には,実施例1で得た精製したpCI-neoベクターに野生型hGALC,HSA-hGALC,及びhGALC-HSAをそれぞれコードする遺伝子を組み込んだプラスミドを用いた。
[Example 2] Transient expression of wild-type hGALC, HSA-hGALC, and hGALC-HSA For transient expression of wild-type hGALC, HSA-hGALC, and hGALC-HSA, plasmids were used in which the genes encoding wild-type hGALC, HSA-hGALC, and hGALC-HSA, respectively, were incorporated into the purified pCI-neo vector obtained in Example 1.
 ExpiCHOTM Expression System(Thermo Fisher Scientific社)のHigh titerプロトコールに従い,野生型hGALC,HSA-hGALC,及びhGALC-HSAをそれぞれコードする遺伝子を組み込んだプラスミドを用いて,ExpiCHO細胞を形質転換させた。形質転換後,細胞を8日間培養し,野生型hGALC,HSA-hGALC,及びhGALC-HSAをそれぞれ培養上清中に発現させた。培養開始から6,7,及び8日後の培養液をそれぞれ遠心分離して培養上清を回収した。 According to the High titer protocol of the ExpiCHO TM Expression System (Thermo Fisher Scientific), ExpiCHO cells were transformed with plasmids incorporating genes encoding wild-type hGALC, HSA-hGALC, and hGALC-HSA, respectively. After transformation, the cells were cultured for 8 days, and wild-type hGALC, HSA-hGALC, and hGALC-HSA were expressed in the culture supernatant, respectively. The culture solutions 6, 7, and 8 days after the start of culture were centrifuged to collect the culture supernatant.
〔実施例3〕一過性発現による野生型hGALC,HSA-hGALC,及びhGALC-HSAの発現量の確認(SDS page電気泳動)
 実施例2で得られた培養上清それぞれ10 μLを,8 μLの2×Sample Buffer(バイオラッド社)及び2 μLの2-Mercaptoethanolと混合し,100℃で3分間加熱することにより還元条件下で熱変性させた。熱変性後の溶液を,0.1% SDSを含む50 mM Tris緩衝液/380 mM グリシン緩衝液(pH 8.3)内に設置した5-20%ポリアクリルアミドゲルのウェルに5 μLずつ付し,25 mA定電流にて電気泳動した。電気泳動後のゲルをOriole Fluorescent Gel Stain(バイオラッド社)に浸し,室温で90分間振とうした。ゲルを純水で洗浄した後,ルミノイメージアナライザー(Amersham Imager 600RGB,サイティバ社)を用いて各蛋白質に相当するバンドが可視化された電気泳動画像を得た。
[Example 3] Confirmation of the expression levels of wild-type hGALC, HSA-hGALC, and hGALC-HSA by transient expression (SDS page electrophoresis)
10 μL of each culture supernatant obtained in Example 2 was mixed with 8 μL of 2×Sample Buffer (Bio-Rad) and 2 μL of 2-Mercaptoethanol, and heated at 100° C. for 3 minutes to be heat-denatured under reducing conditions. 5 μL of the heat-denatured solution was added to each well of a 5-20% polyacrylamide gel placed in 50 mM Tris buffer/380 mM glycine buffer (pH 8.3) containing 0.1% SDS, and electrophoresis was performed at a constant current of 25 mA. The gel after electrophoresis was immersed in Oriole Fluorescent Gel Stain (Bio-Rad) and shaken at room temperature for 90 minutes. After washing the gel with pure water, an electrophoretic image was obtained in which the bands corresponding to each protein were visualized using a lumino image analyzer (Amersham Imager 600RGB, Cytiva).
〔実施例4〕一過性発現による野生型hGALC,HSA-hGALC及びhGALC-HSAの発現量の確認(ウエスタンブロッティング)
 実施例3に記載した方法と同様に電気泳動を行い,ニトロセルロース膜と電気泳動後のゲルを,20%メタノールを含む25 mM Tris緩衝液/192 mM グリシン緩衝液に浸したブロッティングペーパーで挟み,ブロッティング装置で1.0 A,25 Vで10分間通電することで,蛋白質をニトロセルロース膜に転写した。転写後のニトロセルロース膜を5%スキムミルクを含むPBSTに浸して1時間振とうした後,0.4 μg/mLに希釈したAnti-GALC 抗体(Rabbit polyclonal to GALC,abcam社)に浸して1時間振とうした。PBSTで膜を洗浄後,0.4 μg/mLに希釈したAnti-mouse IgG (H+L), HRP Conjugate(プロメガ社)溶液に浸して30分間振とうし,再度PBSTで洗浄した。膜の転写面にHRP検出試薬(バイオラッド社)を滴下して5分間反応させ,ルミノイメージアナライザー(Amersham Imager 600RGB,サイティバ社)を用いて各蛋白質に相当するバンドが可視化されたウエスタンブロッティング画像を得た。
[Example 4] Confirmation of expression levels of wild-type hGALC, HSA-hGALC, and hGALC-HSA by transient expression (Western blotting)
Electrophoresis was performed in the same manner as described in Example 3, and the nitrocellulose membrane and the gel after electrophoresis were sandwiched between blotting papers soaked in 25 mM Tris buffer/192 mM glycine buffer containing 20% methanol, and the proteins were transferred to the nitrocellulose membrane by applying electricity at 1.0 A and 25 V for 10 minutes in a blotting device. The nitrocellulose membrane after transfer was immersed in PBST containing 5% skim milk and shaken for 1 hour, and then immersed in Anti-GALC antibody (Rabbit polyclonal to GALC, Abcam) diluted to 0.4 μg/mL and shaken for 1 hour. After washing the membrane with PBST, it was immersed in Anti-mouse IgG (H+L), HRP Conjugate (Promega) solution diluted to 0.4 μg/mL and shaken for 30 minutes, and washed again with PBST. An HRP detection reagent (Bio-Rad) was dropped onto the transfer surface of the membrane and reacted for 5 minutes, and a Western blotting image was obtained using a luminometer image analyzer (Amersham Imager 600RGB, Cytiva) in which bands corresponding to each protein were visualized.
〔実施例5〕一過性発現による野生型hGALC,HSA-hGALC,及びhGALC-HSAの発現量の確認(酵素活性測定)
 試料溶液として,実施例2で得られた培養上清を0.5% Triton X-100を含むクエン酸緩衝液(pH4.5)で100倍希釈したものを調製した。試料溶液は,必要に応じてさらに適宜希釈して測定に供した。標準溶液として,4-MU(4-Methylumbelliferone,シグマアルドリッチ社)を0.1% BSAを含む100 mM クエン酸緩衝液(pH4.2)で75~4.39 μMに段階希釈したものを調製した。基質溶液として,hGALCの人工基質である4-Methylumbelliferyl-β-D-galactopyranoside(シグマアルドリッチ社)を0.5% Triton X-100を含むクエン酸緩衝液(pH4.5)で1 mmol/Lに希釈したものを調製した。マイクロプレートに試料溶液又は標準溶液を25 μL/well添加し,更に各ウェルに25 μLの基質溶液を添加してプレートシェイカーで撹拌し混合した。プレートを37℃で1時間静置した後,各ウェルに150 μLの200 mmol/L グリシン-NaOH緩衝液(pH10.6)を添加して反応を停止させた。次いで,蛍光プレートリーダー(Gemini XPS,モレキュラーデバイス社)を用いて各ウェルの蛍光強度を測定した(励起波長365 nm,蛍光波長460 nm)。この蛍光強度は,各ウェル中の溶液に含まれる4-MU(4-Methylumbelliferone)の濃度に比例する。標準溶液の測定結果に基づき検量線を作成し,これに各試料溶液の測定値を内挿して,酵素活性量(μM/時間)を求めた。なお,この酵素活性量の単位は,1時間当たりに分解される基質の量を示すものである。
[Example 5] Confirmation of expression levels of wild-type hGALC, HSA-hGALC, and hGALC-HSA by transient expression (enzyme activity measurement)
The culture supernatant obtained in Example 2 was diluted 100-fold with citrate buffer (pH 4.5) containing 0.5% Triton X-100 to prepare a sample solution. The sample solution was further diluted appropriately as necessary and used for measurement. A standard solution was prepared by serially diluting 4-MU (4-Methylumbelliferone, Sigma-Aldrich) from 75 to 4.39 μM with 100 mM citrate buffer (pH 4.2) containing 0.1% BSA. A substrate solution was prepared by diluting 4-Methylumbelliferyl-β-D-galactopyranoside (Sigma-Aldrich), an artificial substrate for hGALC, to 1 mmol/L with citrate buffer (pH 4.5) containing 0.5% Triton X-100. 25 μL/well of the sample solution or standard solution was added to a microplate, and 25 μL of substrate solution was added to each well and mixed by stirring with a plate shaker. After the plate was left at 37°C for 1 hour, 150 μL of 200 mmol/L glycine-NaOH buffer (pH 10.6) was added to each well to stop the reaction. The fluorescence intensity of each well was then measured using a fluorescence plate reader (Gemini XPS, Molecular Devices, Inc.) (excitation wavelength 365 nm, fluorescence wavelength 460 nm). This fluorescence intensity is proportional to the concentration of 4-MU (4-Methylumbelliferone) contained in the solution in each well. A calibration curve was created based on the measurement results of the standard solution, and the measured values of each sample solution were interpolated to determine the enzyme activity (μM/hour). The unit of enzyme activity indicates the amount of substrate decomposed per hour.
〔実施例6〕一過性発現による野生型hGALC,HSA-hGALC,及びhGALC-HSAの発現量の確認(結果)
 図15(a)に,実施例5で測定した一過性発現による野生型hGALC,HSA-hGALC,及びhGALC-HSAの酵素活性測定の結果を示す。また,酵素活性測定の結果に基づく野生型hGALC,HSA-hGALC,及びhGALC-HSAの相対量(野生型hGALCの各測定日における発現量を1.0としたときの相対量)を表1に示す。
[Example 6] Confirmation of expression levels of wild-type hGALC, HSA-hGALC, and hGALC-HSA by transient expression (Results)
15(a) shows the results of enzyme activity measurement of wild-type hGALC, HSA-hGALC, and hGALC-HSA by transient expression measured in Example 5. In addition, the relative amounts of wild-type hGALC, HSA-hGALC, and hGALC-HSA based on the results of enzyme activity measurement (relative amounts when the expression amount of wild-type hGALC on each measurement day is set to 1.0) are shown in Table 1.
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000001
 これらの結果から,培養開始後6日後の発現量(酵素活性量換算)をみると,HSA-hGALC及びhGALC-HSAの発現量は,野生型hGALCのそれと比較して,それぞれ3.2倍及び1.5倍の値を示し,野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なhGALCを,HSA-hGALC又はhGALC-HSAの形態で発現させることにより,より多くのhGALCを組換え蛋白質として効率よく発現させることができることがわかった。培養開始後7日後及び8日後においても同様のことがいえる。 These results show that, when looking at the expression levels (converted into enzyme activity) 6 days after the start of culture, the expression levels of HSA-hGALC and hGALC-HSA were 3.2 and 1.5 times higher, respectively, than that of wild-type hGALC. This shows that hGALC, which is difficult to mass-produce due to its low expression level when expressed as a recombinant in its wild form, can be efficiently expressed in greater amounts as a recombinant protein by expressing it in the form of HSA-hGALC or hGALC-HSA. The same can be said for 7 and 8 days after the start of culture.
 実施例3で得た電気泳動画像とウエスタンブロッティング画像を図15(b)及び(c)にそれぞれ示す。ウエスタンブロッティング画像から,電気泳動画像上の野生型hGALC,HSA-hGALC,及びhGALC-HSAに相当するバンドを特定した。ウエスタンブロッティング画像のバンドの濃さから概算される野生型hGALC,HSA-hGALCのhGALCに相当する部分,及びhGALC-HSAのhGALCに相当する部分の発現量(分子数換算)は,図15(a)及び表1で示される発現量(酵素活性量換算)とほぼ相関するものであった。つまり,表1に発現量(酵素活性量換算)として示される発現量の相対値は,このまま,野生型hGALC,HSA-hGALCのhGALCに相当する部分,及びhGALC-HSAのhGALCに相当する部分の発現量(分子数換算)ということができる。すなわち,これらの結果は,野生型hGALCをHSAとの融合蛋白質とすることにより,野生型のままでは組換え体として大量に製造することが困難なhGALCを,融合蛋白質の形態とすることにより,比活性を低下させることなく,より多くの分子数発現させることができることを示すものである。なお,ここで融合蛋白質におけるhGALCの比活性は,当該融合蛋白質の単位質量当たりのhGALCの酵素活性(μM/時間/mg蛋白質)に(当該融合蛋白質の分子量/当該融合蛋白質中のhGALCに相当する部分の分子量)を乗じて算出される。 Electrophoretic images and Western blotting images obtained in Example 3 are shown in Figures 15(b) and (c), respectively. From the Western blotting images, bands corresponding to wild-type hGALC, HSA-hGALC, and hGALC-HSA on the electrophoretic images were identified. The expression levels (in terms of molecular number) of the parts of wild-type hGALC, HSA-hGALC corresponding to hGALC, and the parts of hGALC-HSA corresponding to hGALC, estimated from the density of the bands in the Western blotting images, were approximately correlated with the expression levels (in terms of enzyme activity) shown in Figure 15(a) and Table 1. In other words, the relative values of the expression levels shown as the expression levels (in terms of enzyme activity) in Table 1 can be said to be the expression levels (in terms of molecular number) of the parts of wild-type hGALC, HSA-hGALC corresponding to hGALC, and hGALC-HSA corresponding to hGALC. In other words, these results show that by making wild-type hGALC into a fusion protein with HSA, it is possible to express a larger number of molecules of hGALC, which is difficult to mass-produce as a recombinant in its wild form, without reducing its specific activity by making it into a fusion protein. Note that the specific activity of hGALC in the fusion protein is calculated by multiplying the enzyme activity of hGALC per unit mass of the fusion protein (μM/hour/mg protein) by (the molecular weight of the fusion protein/the molecular weight of the portion of the fusion protein corresponding to hGALC).
〔実施例7〕野生型hGALC,HSA-hGALC,及びhGALC-HSAの一過性発現における細胞の生存率等
 実施例2で実施した野生型hGALC,HSA-hGALC,及びhGALC-HSAの一過性発現において,形質転換後のExpiCHO細胞の培養開始から6,7,及び8日後における培養液中の生細胞密度及び細胞生存率を,EVE Automated Cell Counter(Nano EnTek社)を用いて測定した。野生型hGALC,HSA-hGALC,及びhGALC-HSAを発現するそれぞれの細胞について,培養開始から6,7,及び8日後における培養液中の生細胞密度及び細胞生存率の結果を表2に示す。
[Example 7] Cell viability, etc. in transient expression of wild-type hGALC, HSA-hGALC, and hGALC-HSA In the transient expression of wild-type hGALC, HSA-hGALC, and hGALC-HSA performed in Example 2, the viable cell density and cell viability in the culture medium 6, 7, and 8 days after the start of culture of transformed ExpiCHO cells were measured using an EVE Automated Cell Counter (Nano EnTek). Table 2 shows the viable cell density and cell viability in the culture medium 6, 7, and 8 days after the start of culture for each of the cells expressing wild-type hGALC, HSA-hGALC, and hGALC-HSA.
Figure JPOXMLDOC01-appb-T000002
Figure JPOXMLDOC01-appb-T000002
 生細胞密度については,野生型hGALC,HSA-hGALC,及びhGALC-HSAそれぞれを発現する細胞の間で大きな差はみられなかった。しかし,細胞の生存率については,HSA-hGALCを発現する細胞では培養開始6日後から8日後にかけて80%以上の高い生存率を維持し,hGALC-HSAを発現する細胞では培養開始6日後から8日後にかけて徐々に低下しながら70%以上の細胞生存率を維持した一方で,野生型hGALCを発現する細胞では培養開始6日後から8日後にかけて細胞の生存率が大きく下落し,培養開始8日後では60%まで低下した。各測定日における野生型hGALC発現細胞の生存率を1.0としたときの,生存率の相対比は,培養開始6日後ではHSA-hGALC及びhGALC-HSA発現細胞でそれぞれ1.1及び1.1,培養開始7日後ではHSA-hGALC及びhGALC-HSA発現細胞でそれぞれ1.3及び1.2,培養開始8日後ではHSA-hGALC及びhGALC-HSA発現細胞でそれぞれ1.5及び1.2であった。 There was no significant difference in viable cell density between cells expressing wild-type hGALC, HSA-hGALC, and hGALC-HSA. However, in terms of cell viability, cells expressing HSA-hGALC maintained a high viability of over 80% from 6 to 8 days after the start of culture, while cells expressing hGALC-HSA maintained a cell viability of over 70% from 6 to 8 days after the start of culture, although the cell viability gradually decreased from 6 to 8 days after the start of culture. On the other hand, the cell viability of cells expressing wild-type hGALC dropped significantly from 6 to 8 days after the start of culture, dropping to 60% at 8 days after the start of culture. When the viability of wild-type hGALC expressing cells on each measurement day was set to 1.0, the relative viability ratios were 1.1 and 1.1 for HSA-hGALC and hGALC-HSA expressing cells, respectively, 6 days after the start of culture, 1.3 and 1.2 for HSA-hGALC and hGALC-HSA expressing cells, respectively, 7 days after the start of culture, and 1.5 and 1.2 for HSA-hGALC and hGALC-HSA expressing cells, respectively, 8 days after the start of culture.
 これらの結果は,野生型hGALCは,これを組換え蛋白質として発現させたときに,宿主細胞にストレスを与えてその生存率を低下させること,及び,野生型hGALCにHSAを結合させた融合蛋白質として発現させることにより,野生型hGALCが宿主細胞に与えるストレスを低減させて,生存率の低下を抑制できることを示すものと考えられる。つまり,実施例6で示された,野生型hGALCと比較したときの,HSA-hGALC又はhGALC-HSAの組換え蛋白質として発現させたときの発現量の増加の一要因として,野生型hGALCを発現させたときに宿主細胞に与えられるストレスが低減されて細胞の生存率の低下が抑制されたことが考えられる。 These results suggest that when wild-type hGALC is expressed as a recombinant protein, it causes stress to the host cells, reducing their viability, and that when wild-type hGALC is expressed as a fusion protein in which HSA is bound to the wild-type hGALC, the stress that wild-type hGALC causes to the host cells can be reduced, suppressing the reduction in viability. In other words, one factor behind the increase in expression level when expressed as a recombinant protein of HSA-hGALC or hGALC-HSA compared to wild-type hGALC, as shown in Example 6, is thought to be the reduction in the stress caused to the host cells when wild-type hGALC is expressed, thereby suppressing the reduction in cell viability.
〔実施例8〕まとめ
 以上の結果は,hGALCを組換え蛋白質として製造する場合に,hGALCを,HSA-hGALC又はhGALC-HSAの融合蛋白質の形態で製造することにより,hGALCの比活性を低下させることなく,約1.5~3.6倍の分子数のhGALCを得ることができることを示す。つまり,hGALCをHSA-hGALC又はhGALC-HSAの融合蛋白質の形態で発現させる組換えhGALCの製造方法は,大量の組換えhGALCを製造する方法として極めて有効な手段といえる。また,死細胞からは,その内容物が流出し,それらが夾雑物となるので,培養中に死細胞が増加することは,発現した融合蛋白質の精製を困難にする可能性がある。従って,hGALCをHSA-hGALC又はhGALC-HSAの融合蛋白質の形態で発現させることにより,培養中の細胞死を抑制できるので,発現した融合蛋白質の精製を容易にすることができる。これにより精製時における精製効率を高めることもできる。
[Example 8] Summary The above results show that when producing hGALC as a recombinant protein, producing hGALC in the form of a fusion protein of HSA-hGALC or hGALC-HSA makes it possible to obtain about 1.5 to 3.6 times the number of molecules of hGALC without decreasing the specific activity of hGALC. In other words, the method of producing recombinant hGALC by expressing hGALC in the form of a fusion protein of HSA-hGALC or hGALC-HSA can be said to be an extremely effective means for producing a large amount of recombinant hGALC. In addition, since contents of dead cells leak out and become impurities, an increase in dead cells during culture may make it difficult to purify the expressed fusion protein. Therefore, by expressing hGALC in the form of a fusion protein of HSA-hGALC or hGALC-HSA, cell death during culture can be suppressed, making it easier to purify the expressed fusion protein. This also increases the purification efficiency during purification.
〔実施例9〕一過性発現による野生型hGALC,HSA-hGALC,及びhGALC-HSAのSE-HPLC分析
 一過性発現により得られた野生型hGALC,HSA-hGALC,及びhGALC-HSAの物性を評価するため,各蛋白質の培養上清を精製工程に供し,その後SE-HPLC分析を行った。
[Example 9] SE-HPLC analysis of wild-type hGALC, HSA-hGALC, and hGALC-HSA obtained by transient expression In order to evaluate the physical properties of wild-type hGALC, HSA-hGALC, and hGALC-HSA obtained by transient expression, the culture supernatant of each protein was subjected to a purification process and then subjected to SE-HPLC analysis.
〔精製工程〕
 実施例2で得られた,一過性発現による野生型hGALC,HSA-hGALC,及びhGALC-HSAをそれぞれ含む8日目の培養上清に,等倍容の50 mM NaClを含有する25 mM MES緩衝液(pH 6.5)を添加した。この溶液を,50 mM NaClを含有する25 mM MES緩衝液(pH 6.5)で平衡化した,強陰イオン交換カラムであるHitrap Qカラム(cytiva社)に,一定流速で負荷させた。次いで,カラム体積の5倍容の同緩衝液を同流速で供給してカラムを洗浄した。次いで,500 mM NaClを含有する25 mM MES緩衝液(pH 6.5)を用いて,塩濃度勾配で強陰イオン交換カラムに吸着した野生型hGALC,HSA-hGALC及びhGALC-HSAを溶出させた。
[Refining process]
An equal volume of 25 mM MES buffer (pH 6.5) containing 50 mM NaCl was added to the culture supernatant on day 8 containing wild-type hGALC, HSA-hGALC, and hGALC-HSA obtained by transient expression in Example 2. This solution was loaded at a constant flow rate onto a strong anion exchange column, Hitrap Q column (Cytiva), equilibrated with 25 mM MES buffer (pH 6.5) containing 50 mM NaCl. The column was then washed by supplying 5 times the column volume of the same buffer at the same flow rate. The wild-type hGALC, HSA-hGALC, and hGALC-HSA adsorbed on the strong anion exchange column were then eluted with a salt concentration gradient using 25 mM MES buffer (pH 6.5) containing 500 mM NaCl.
〔SE-HPLC工程〕
 島津HPLCシステムLC-20A(島津製作所社)に,サイズ排除カラムクロマトグラフィーカラムであるTSKgel G3000SWXL 5 μmカラム(7.8 mm径×30 cm長,TOSOH社)をセットした。また,カラムの下流に吸光光度計を設置し,カラムからの流出液の吸光度(測定波長215 nm)を連続して測定できるようにした。10%のエタノールを含む0.2 M リン酸ナトリウム緩衝水溶液でカラムを平衡化させた後,上記精製工程で得られた野生型hGALC,HSA-hGALC,hGALC-HSAを含有する各溶出液をそれぞれカラムに負荷し,更に10%のエタノールを含む0.2 Mリン酸ナトリウム緩衝水溶液を0.5 mL/分の流速で流した。この間,カラムからの流出液の吸光度(測定波長215 nm)を測定することにより,溶出プロフィールを得た。
[SE-HPLC process]
A size-exclusion column, TSKgel G3000SWXL 5 μm column (7.8 mm diameter × 30 cm length, TOSOH) was set on a Shimadzu HPLC system LC-20A (Shimadzu Corporation). A spectrophotometer was also installed downstream of the column to continuously measure the absorbance (measurement wavelength 215 nm) of the effluent from the column. After equilibrating the column with 0.2 M sodium phosphate buffer solution containing 10% ethanol, each eluate containing wild-type hGALC, HSA-hGALC, and hGALC-HSA obtained in the above purification process was loaded onto the column, and then 0.2 M sodium phosphate buffer solution containing 10% ethanol was passed through the column at a flow rate of 0.5 mL/min. During this time, the absorbance (measurement wavelength 215 nm) of the effluent from the column was measured to obtain an elution profile.
〔物性評価〕
 溶出プロフィールを図16に示す。野生型hGALCでは,メインピークが2つあり,それぞれのピークは溶出プロフィール上の左側から順にhGALCの四量体,及びhGALCの二量体に対応すると考えられる。一方,HSA-hGALC及びhGALC-HSAでは,ともにメインピークが1つであり,それぞれHSA-hGALC又はhGALC-HSAの単量体のピークであると考えられる。このことから,細胞に野生型hGALCを発現させた場合と比較して,HSAとhGALCとの融合蛋白質を発現させた場合には,hGALC活性を有する分子が単量体として安定的に得られていることがわかる。なお,HSA-hGALC及びhGALC-HSAのそれぞれにおいて,全発現量に占める単量体の質量比率は,90%以上である。組換え蛋白質として発現させたとき,HSA-hGALC及びhGALC-HSAは単量体として均質なものを取得することができるので,製造管理の観点からも,hGALCをHSAとの融合蛋白質として製造するhGALCの製造方法は優れたものということができる。
〔Evaluation of the physical properties〕
The elution profile is shown in Figure 16. Wild-type hGALC has two main peaks, which are considered to correspond to the hGALC tetramer and the hGALC dimer, respectively, from the left side of the elution profile. On the other hand, HSA-hGALC and hGALC-HSA both have one main peak, which is considered to be the peak of the HSA-hGALC or hGALC-HSA monomer, respectively. This shows that, compared to the case where wild-type hGALC is expressed in cells, when the fusion protein of HSA and hGALC is expressed, a molecule having hGALC activity is stably obtained as a monomer. In addition, the mass ratio of the monomer to the total expression amount is 90% or more in each of HSA-hGALC and hGALC-HSA. When expressed as recombinant proteins, HSA-hGALC and hGALC-HSA can be obtained as homogeneous monomers, so from the viewpoint of production control, the method of producing hGALC as a fusion protein of hGALC with HSA can be said to be superior.
〔実施例10〕抗hTfR抗体と,HSAとhGALCとの融合蛋白質との結合体を発現するプラスミドの作製
 配列番号25で示されるアミノ酸配列を有する重鎖と,配列番号23で示されるアミノ酸配列を有する軽鎖とからなる抗hTfR抗体(Fab)と,HSAとhGALCとの融合蛋白質との結合体として,以下に示す4種類の蛋白質を発現するプラスミドをそれぞれ作製した。
(1)抗hTfR抗体(Fab)の重鎖のC末端にリンカーを介してHSA-hGALCのN末端を結合させた融合蛋白質と,抗hTfR抗体(Fab)の軽鎖とからなる結合体(Fab-HSA-hGALCとする);
(2)抗hTfR抗体(Fab)の重鎖のN末端にリンカーを介してHSA-hGALCのC末端を結合させた融合蛋白質と,抗hTfR抗体(Fab)の軽鎖とからなる結合体(HSA-hGALC-Fabとする);
(3)抗hTfR抗体(Fab)の重鎖のC末端にリンカーを介してhGALC-HSAのN末端を結合させた融合蛋白質と,抗hTfR抗体(Fab)の軽鎖とからなる結合体(Fab-hGALC-HSAとする);及び
(4)抗hTfR抗体(Fab)の重鎖のN末端にリンカーを介してhGALC-HSAのC末端を結合させた融合蛋白質と,抗hTfR抗体(Fab)の軽鎖とからなる結合体(hGALC-HSA-Fabとする)。
Example 10: Preparation of plasmids expressing conjugates of anti-hTfR antibody and fusion protein of HSA and hGALC Plasmids were prepared that express the four types of proteins shown below as conjugates of an anti-hTfR antibody (Fab) consisting of a heavy chain having the amino acid sequence shown in SEQ ID NO:25 and a light chain having the amino acid sequence shown in SEQ ID NO:23, and a fusion protein of HSA and hGALC.
(1) A conjugate consisting of a fusion protein in which the N-terminus of HSA-hGALC is linked via a linker to the C-terminus of the heavy chain of an anti-hTfR antibody (Fab) and the light chain of an anti-hTfR antibody (Fab) (referred to as Fab-HSA-hGALC);
(2) A conjugate consisting of a fusion protein in which the C-terminus of HSA-hGALC is linked via a linker to the N-terminus of the heavy chain of an anti-hTfR antibody (Fab) and the light chain of an anti-hTfR antibody (Fab) (HSA-hGALC-Fab);
(3) A conjugate consisting of a fusion protein in which the N-terminus of hGALC-HSA is linked via a linker to the C-terminus of the heavy chain of an anti-hTfR antibody (Fab) and a light chain of an anti-hTfR antibody (Fab) (referred to as Fab-hGALC-HSA); and (4) a conjugate consisting of a fusion protein in which the C-terminus of hGALC-HSA is linked via a linker to the N-terminus of the heavy chain of an anti-hTfR antibody (Fab) and a light chain of an anti-hTfR antibody (Fab) (referred to as hGALC-HSA-Fab).
〔Dual(+)pCI-neoベクターの作製〕
 5’側より,NheIサイト,AscIサイト,NruIサイト,AfeIサイト,及びNotI無効化配列を挿入したNotIサイトを含む配列番号26で示される塩基配列を有するプライマー(pCI Insert F3プライマー)を合成した。また,これと相補的な配列を含む配列番号27で示される塩基配列を有するプライマー(pCI Insert R3プライマー)を合成した。これら2つのプライマーをアニーリングさせ,両端にNheI及びNotI消化したDNAと,相補的な突出末端を有するDNA断片を得た。
[Construction of Dual(+)pCI-neo vector]
A primer (pCI Insert F3 primer) having the base sequence shown in SEQ ID NO: 26, which includes an NheI site, an AscI site, an NruI site, an AfeI site, and a NotI site with a NotI-disabling sequence inserted, was synthesized from the 5' side. A primer (pCI Insert R3 primer) having the base sequence shown in SEQ ID NO: 27, which includes a complementary sequence to the primer, was also synthesized. These two primers were annealed to obtain DNA digested with NheI and NotI at both ends and a DNA fragment having complementary protruding ends.
 pCIベクター(プロメガ社)をNheIとNotI(タカラバイオ社)で消化し,これに上記DNA断片をライゲーション反応により挿入した。得られたプラスミドをpCI MCS-modifiedベクターとした(図17)。pCI MCS-modifiedベクターをBamHIとBglII(タカラバイオ社)で消化し,CMV immediate-earlyエンハンサー/プロモーター領域,マルチクローニングサイト,polyA配列を含むDNA断片を切り出した。pCI-neoベクター(プロメガ社)をBamHI消化及びCIAP(タカラバイオ社)処理し,切り出したDNA断片をライゲーション反応により挿入した。得られたプラスミドをDual(+)pCI-neoベクターとした(図18)。 The pCI vector (Promega) was digested with NheI and NotI (Takara Bio), and the above DNA fragment was inserted into it by ligation. The resulting plasmid was named pCI MCS-modified vector (Figure 17). The pCI MCS-modified vector was digested with BamHI and BglII (Takara Bio), and a DNA fragment containing the CMV immediate-early enhancer/promoter region, multicloning site, and polyA sequence was excised. The pCI-neo vector (Promega) was digested with BamHI and treated with CIAP (Takara Bio), and the excised DNA fragment was inserted into it by ligation. The resulting plasmid was named Dual(+)pCI-neo vector (Figure 18).
〔各結合体発現プラスミドの作製〕
 配列番号28で示されるアミノ酸配列を有する,抗hTfR抗体(Fab)の重鎖のC末端にリンカーを介してHSA-hGALCのN末端を結合させた結合体(抗hTfR抗体Fab重鎖-HSA-hGALC)をコードする遺伝子を含む,配列番号29で示される塩基配列を含むDNA断片を制限酵素MluI及びNotI(タカラバイオ社)で消化し,Dual(+)pCI-neoベクターのMluI及びNotIサイト間に挿入した。更に,抗hTfR抗体Fab重鎖-HSA-hGALCをコードする遺伝子を挿入したDual(+)pCI-neoベクターを制限酵素AscI及びAfeI(New England BioLabs社)で消化し,これに配列番号23で示されるアミノ酸配列を有する,抗hTfR抗体(Fab)の軽鎖をコードする遺伝子を含む,配列番号36で示される塩基配列を含むDNA断片をAscI及びAfeIで消化したものを挿入した。得られたプラスミドを,Fab-HSA-hGALC発現プラスミドとした。
[Preparation of expression plasmids for each conjugate]
A DNA fragment containing the base sequence shown in SEQ ID NO:29, including a gene encoding a conjugate (anti-hTfR antibody Fab heavy chain-HSA-hGALC) in which the N-terminus of HSA-hGALC is linked to the C-terminus of the heavy chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO:28 via a linker, was digested with restriction enzymes MluI and NotI (Takara Bio Inc.) and inserted between the MluI and NotI sites of the Dual(+)pCI-neo vector. Furthermore, the Dual(+)pCI-neo vector into which the gene encoding the anti-hTfR antibody Fab heavy chain-HSA-hGALC was inserted was digested with restriction enzymes AscI and AfeI (New England BioLabs), and a DNA fragment containing the base sequence shown in SEQ ID NO:36, including a gene encoding the light chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO:23, digested with AscI and AfeI, was inserted into the vector. The resulting plasmid was used as a Fab-HSA-hGALC expression plasmid.
 同様にして,配列番号30で示されるアミノ酸配列を有する,抗hTfR抗体(Fab)の重鎖のN末端にリンカーを介してHSA-hGALCのC末端を結合させた結合体をコードする遺伝子を含む,配列番号31で示される塩基配列を含むDNA断片,及び配列番号23で示されるアミノ酸配列を有する,抗hTfR抗体(Fab)の軽鎖をコードする遺伝子を含む,配列番号36で示される塩基配列を含むDNA断片を用いて,HSA-hGALC-Fab発現プラスミドを作製した。 In a similar manner, an HSA-hGALC-Fab expression plasmid was prepared using a DNA fragment containing the base sequence shown in SEQ ID NO: 31, which contains a gene encoding a conjugate in which the C-terminus of HSA-hGALC is linked via a linker to the N-terminus of the heavy chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 30, and a DNA fragment containing the base sequence shown in SEQ ID NO: 36, which contains a gene encoding the light chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 23.
 同様にして,配列番号32で示されるアミノ酸配列を有する,抗hTfR抗体(Fab)の重鎖のC末端にリンカーを介してhGALC-HSAのN末端を結合させた結合体をコードする遺伝子を含む,配列番号33で示される塩基配列を含むDNA断片,及び配列番号23で示されるアミノ酸配列を有する,抗hTfR抗体(Fab)の軽鎖をコードする遺伝子を含む,配列番号36で示される塩基配列を含むDNA断片を用いて,Fab-hGALC-HSA発現プラスミドを作製した。 In a similar manner, a Fab-hGALC-HSA expression plasmid was prepared using a DNA fragment containing the base sequence shown in SEQ ID NO: 33, which contains a gene encoding a conjugate in which the N-terminus of hGALC-HSA is linked via a linker to the C-terminus of the heavy chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 32, and a DNA fragment containing the base sequence shown in SEQ ID NO: 36, which contains a gene encoding the light chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 23.
 同様にして,配列番号34で示されるアミノ酸配列を有する,抗hTfR抗体(Fab)の重鎖のN末端にリンカーを介してhGALC-HSAのC末端を結合させた結合体をコードする遺伝子を含む,配列番号35で示される塩基配列を含むDNA断片,及び配列番号23で示されるアミノ酸配列を有する,抗hTfR抗体(Fab)の軽鎖をコードする遺伝子を含む,配列番号36で示される塩基配列を含むDNA断片を用いて,hGALC-HSA-Fab発現プラスミドを作製した。 In a similar manner, an hGALC-HSA-Fab expression plasmid was prepared using a DNA fragment containing the base sequence shown in SEQ ID NO: 35, which contains a gene encoding a conjugate in which the C-terminus of hGALC-HSA is linked via a linker to the N-terminus of the heavy chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 34, and a DNA fragment containing the base sequence shown in SEQ ID NO: 36, which contains a gene encoding the light chain of an anti-hTfR antibody (Fab) having the amino acid sequence shown in SEQ ID NO: 23.
〔実施例11〕各結合体の一過性発現
 ExpiCHOTM Expression System(Thermo Fisher Scientific社)のHigh titerプロトコールに従い,実施例10で作製した各結合体発現プラスミドを用いて,ExpiCHO細胞をそれぞれ形質転換させた。形質転換後,細胞を8日間培養し,Fab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabをそれぞれ培養上清中に発現させた。培養開始から8日後の培養液をそれぞれ遠心分離して培養上清を回収した。
[Example 11] Transient expression of each conjugate According to the High titer protocol of the ExpiCHO Expression System (Thermo Fisher Scientific), each conjugate expression plasmid prepared in Example 10 was used to transform ExpiCHO cells. After transformation, the cells were cultured for 8 days, and Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab were expressed in the culture supernatant. After 8 days from the start of culture, the culture solutions were centrifuged to collect the culture supernatants.
〔実施例12〕一過性発現によるFab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabの発現量の確認(酵素活性測定)
 試料溶液として実施例11で得られた培養上清を用い,実施例5に記載の方法と同様にして一過性発現によるFab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabの発現量を酵素活性測定により確認した。
[Example 12] Confirmation of expression levels of Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab by transient expression (enzyme activity measurement)
Using the culture supernatant obtained in Example 11 as a sample solution, the expression levels of Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab by transient expression were confirmed by enzyme activity measurement in the same manner as described in Example 5.
 図19に,野生型hGALC,Fab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabの酵素活性測定の結果を示す。なお,野生型hGALCの酵素活性測定の結果の値は,実施例2で得られた,野生型hGALCを発現する細胞の培養開始から8日後の培養上清を試料溶液として用いて得られた実施例に記載の測定値を使用した。また,酵素活性測定の結果に基づく野生型hGALC,Fab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabの相対量(野生型hGALCの,培養開始から8日後における発現量を1.0としたときの相対量)を表3に示す。 FIG. 19 shows the results of enzyme activity measurement of wild-type hGALC, Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab. The values obtained in Example 2 using the culture supernatant 8 days after the start of culturing cells expressing wild-type hGALC as the sample solution were used for the enzyme activity measurement results. The relative amounts of wild-type hGALC, Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab based on the results of enzyme activity measurement (relative amounts when the expression amount of wild-type hGALC 8 days after the start of culturing is set to 1.0) are shown in Table 3.
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000003
 これらの結果から,培養開始後8日後の発現量(酵素活性量換算)をみると,Fab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabの発現量は,野生型hGALCのそれと比較して,それぞれ約3.8倍,約2.3倍,約3.0倍,及び約1.7倍の値を示し,野生型hGALCをHSA-hGALC又はhGALC-HSAとすることにより,これらをさらに抗体と結合させた結合体の形態であっても,より多くのhGALCを組換え蛋白質として発現させることができることがわかった。かかる複雑な構造を有し,野生型hGALCと比較して巨大な分子量を有する,抗hTfR抗体とHSAとhGALCとの融合蛋白質との結合体である,Fab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabの何れもが,組換え蛋白質として発現させたときに野生型hGALCと比較して発現量が減少すると予想されるところ,上記の結果は予想外のものであった。 These results show that, when the expression levels (converted into enzyme activity) 8 days after the start of culture were examined, the expression levels of Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab were approximately 3.8-fold, 2.3-fold, 3.0-fold, and 1.7-fold, respectively, compared to that of wild-type hGALC. This indicates that by converting wild-type hGALC to HSA-hGALC or hGALC-HSA, it is possible to express more hGALC as recombinant protein, even in the form of a conjugate further bound to an antibody. The above results were unexpected because all of the conjugates of anti-hTfR antibody, HSA, and hGALC fusion proteins, Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab, which have such a complex structure and a larger molecular weight than wild-type hGALC, are expected to show reduced expression levels compared to wild-type hGALC when expressed as recombinant proteins.
〔実施例13〕一過性発現によるFab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-FabのSE-HPLC分析
 一過性発現により得られたFab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabの物性を評価するため,各蛋白質の培養上清を精製工程に供し,その後SE-HPLC分析を行った。
[Example 13] SE-HPLC analysis of Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab obtained by transient expression In order to evaluate the physical properties of Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab obtained by transient expression, the culture supernatant of each protein was subjected to a purification process, followed by SE-HPLC analysis.
〔精製工程〕
 実施例11で得られた,一過性発現によるFab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabをそれぞれ含む8日目の培養上清に,0.1倍容の2 M アルギニン溶液(pH 6.5)を添加した。この溶液を,カラム体積の5倍容の200 mM アルギニンを含有する25 mM MES緩衝液(pH6.5)で平衡化した,Capture SelectTM CH1-XLカラム(Thermo Fisher SCINTIFIC社)に負荷し,各結合体をカラムに吸着させた。Capture SelectTM CH1-XLカラムは,IgG抗体のCH1ドメインと特異的に結合する性質を有するリガンドが担体に固定化されたアフィニティーカラムである。次いで,カラム体積の5倍容の同緩衝液を供給してカラムを洗浄した。次いでカラム体積の3倍容の25 mM MES緩衝液(pH 6.5)を供給してカラムを更に洗浄した。次いで,カラム体積の5倍容の20 mM 酢酸緩衝液(pH 4.0)で,カラムに吸着した各結合体を溶出させた。溶出液は,予め250 mM MES緩衝液(pH 6.0)と2 M NaCl溶液を入れた容器に受けて,直ちに中和した。
[Refining process]
0.1 volume of 2 M arginine solution (pH 6.5) was added to the culture supernatant on the 8th day containing Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab by transient expression obtained in Example 11. This solution was loaded onto a Capture Select CH1-XL column (Thermo Fisher SCINTIFIC) equilibrated with 25 mM MES buffer (pH 6.5) containing 200 mM arginine in a volume 5 times the column volume, and each conjugate was adsorbed onto the column. The Capture Select CH1-XL column is an affinity column in which a ligand having the property of specifically binding to the CH1 domain of an IgG antibody is immobilized on a carrier. The column was then washed by supplying the same buffer in a volume 5 times the column volume. The column was then further washed by supplying 25 mM MES buffer (pH 6.5) in a volume 3 times the column volume. The bound proteins were then eluted with 5 column volumes of 20 mM acetate buffer (pH 4.0) and immediately neutralized in a container that had previously contained 250 mM MES buffer (pH 6.0) and 2 M NaCl solution.
〔SE-HPLC工程〕
 島津HPLCシステムLC-20A(島津製作所社)に,サイズ排除カラムクロマトグラフィーカラムであるTSKgel G3000SWXL 5 μmカラム(7.8 mm径×30 cm長,TOSOH社)をセットした。また,カラムの下流に吸光光度計を設置し,カラムからの流出液の吸光度(測定波長215 nm)を連続して測定できるようにした。10%のエタノールを含む0.2 M リン酸ナトリウム緩衝水溶液でカラムを平衡化させた後,上記精製工程で得られたFab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabを含有する各溶出液をそれぞれカラムに負荷し,更に10%のエタノールを含む0.2 Mリン酸ナトリウム緩衝水溶液を0.5 mL/分の流速で流した。この間,カラムからの流出液の吸光度(測定波長215 nm)を測定することにより,溶出プロフィールを得た。
[SE-HPLC process]
A size-exclusion column, TSKgel G3000SWXL 5 μm column (7.8 mm diameter × 30 cm length, TOSOH) was set on a Shimadzu HPLC system LC-20A (Shimadzu Corporation). A spectrophotometer was also installed downstream of the column to continuously measure the absorbance (measurement wavelength 215 nm) of the effluent from the column. After equilibrating the column with 0.2 M sodium phosphate buffer solution containing 10% ethanol, each eluate containing Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab obtained in the above purification process was loaded onto the column, and 0.2 M sodium phosphate buffer solution containing 10% ethanol was further passed through the column at a flow rate of 0.5 mL/min. During this time, the absorbance (measurement wavelength 215 nm) of the effluent from the column was measured to obtain an elution profile.
〔物性評価〕
 溶出プロフィールを図20に示す。Fab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,及びhGALC-HSA-Fabの全てにおいて,メインピークが1つであり,それぞれFab-HSA-hGALC,HSA-hGALC-Fab,Fab-hGALC-HSA,又はhGALC-HSA-Fabの単量体のピークであると考えられる。実施例9において得られた,野生型hGALCの溶出プロファイル(図16)上で,メインピークが2種類であったことと比較すると,細胞に野生型hGALCを発現させた場合と比較して,HSAとhGALCとの融合蛋白質と抗体との結合体を発現させた場合にも,hGALC活性を有する分子が単量体として安定的に得られていることがわかる。すなわち,HSAとhGALCとの融合蛋白質を,さらに抗体との結合体とした場合にあっても,hGALCにHSAを融合させることが,hGALCの安定化に寄与していることが示された。なお,結合体のそれぞれにおいて,全発現量に占める単量体の質量比率は,90%以上である。組換え蛋白質として発現させたとき,これらの結合体は単量体として均質なものを取得することができるので,製造管理の観点からも,hGALCをHSAと抗体(ここにおいてFab)の結合体として製造するhGALCの製造方法は優れたものということができる。
〔Evaluation of the physical properties〕
The elution profile is shown in Figure 20. All of Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, and hGALC-HSA-Fab had one main peak, which is considered to be the peak of the monomer of Fab-HSA-hGALC, HSA-hGALC-Fab, Fab-hGALC-HSA, or hGALC-HSA-Fab, respectively. Compared with the fact that there were two main peaks on the elution profile of wild-type hGALC obtained in Example 9 (Figure 16), it can be seen that molecules having hGALC activity were stably obtained as monomers even when the conjugate of the fusion protein of HSA and hGALC and the antibody was expressed, compared to the case where wild-type hGALC was expressed in cells. In other words, it was shown that fusing HSA to hGALC contributes to the stabilization of hGALC, even when the fusion protein of HSA and hGALC was further conjugated with an antibody. In each of the conjugates, the mass ratio of the monomer to the total expression amount is 90% or more. When expressed as recombinant proteins, these conjugates can be obtained as homogeneous monomers, so from the viewpoint of production control, the method of producing hGALC as a conjugate of HSA and an antibody (here, Fab) can be said to be excellent.
 以下,実施例14~20はHSAとhGBAとの融合蛋白質に関する実験結果である。 The following Examples 14 to 20 show the experimental results of a fusion protein of HSA and hGBA.
〔実施例14〕野生型hGBA発現プラスミドの作製
 pE-neo7ベクター及びpCAGIPuroベクター(Miyahara M. et.al., J. Biol. Chem. 275, 613-618(2000))をそれぞれBamHI及びNotI(タカラバイオ社)で制限酵素処理し,アガロースゲル電気泳動にて分離した。なお,pE-neo7ベクターは国際公開公報WO2012/101998に記載の方法で作成した。EtBr染色を行った後,UV照射下で目的のDNA断片を含むバンドを切り出し,QIAEX II Gel Extraction Kit(QIAGEN社)を用いてゲルからDNAを抽出した。Ligation high Ver.2(東洋紡社)を用いて16℃で60分間ライゲーション反応を行いpEI-puroベクターを完成させた。
 野生型hGBA(配列番号37)をコードする遺伝子を含む,配列番号38で示される塩基配列を有するDNA断片を合成した。これをMluI及びNotI(タカラバイオ社)で制限酵素処理し,アガロースゲル電気泳動にて分離した。EtBr染色を行った後,UV照射下で目的のDNA断片を含むバンドを切り出し,QIAEX II Gel Extraction Kit(QIAGEN社)を用いてゲルからDNAを抽出した。
 当該DNA断片及びMluIとNotIで制限酵素処理したpEI-puroベクターを混合し,Ligation high Ver.2を用いて16℃で60分間ライゲーション反応を行い,pEI-puro-hGBAベクターを完成させた。
 pEI-puro-hGBAベクターとpE-mIRES-GSベクターをMluIとNotIで制限酵素処理し,ゲル抽出,精製を行った。なお,pE-mIRES-GSベクターは国際公開公報WO2013/161958に記載の方法で作成した。制限酵素で処理した各ベクターを混合し,Ligation high Ver.2を用いて16℃で60分間ライゲーション反応を行い,pEmIGS-hGBAベクターを完成させた(図21
)。
 pEmIGS-hGBAベクターとpCIneoベクター(プロメガ社)をMluIとNotIで制限酵素処理し,ゲル抽出,精製を行った。そして,制限酵素で処理した各ベクターを混合し,Ligation high Ver.2を用いて16℃で60分間ライゲーション反応を行い,野生型hGBA発現プラスミドであるpCIneo-hGBAベクターを完成させた(図22)。
Example 14: Preparation of wild-type hGBA expression plasmid pE-neo7 vector and pCAGIPuro vector (Miyahara M. et.al., J. Biol. Chem. 275, 613-618(2000)) were treated with restriction enzymes BamHI and NotI (Takara Bio), respectively, and separated by agarose gel electrophoresis. The pE-neo7 vector was prepared by the method described in International Publication WO2012/101998. After EtBr staining, the band containing the target DNA fragment was excised under UV irradiation, and DNA was extracted from the gel using QIAEX II Gel Extraction Kit (QIAGEN). Ligation reaction was performed at 16°C for 60 minutes using Ligation high Ver.2 (Toyobo Co., Ltd.) to complete the pEI-puro vector.
A DNA fragment having the base sequence shown in SEQ ID NO: 38, which contains a gene encoding wild-type hGBA (SEQ ID NO: 37), was synthesized. This was treated with restriction enzymes MluI and NotI (Takara Bio Inc.) and separated by agarose gel electrophoresis. After EtBr staining, the band containing the target DNA fragment was excised under UV irradiation, and DNA was extracted from the gel using a QIAEX II Gel Extraction Kit (QIAGEN).
The DNA fragment was mixed with the pEI-puro vector that had been treated with the restriction enzymes MluI and NotI, and a ligation reaction was carried out at 16°C for 60 minutes using Ligation high Ver.2 to complete the pEI-puro-hGBA vector.
The pEI-puro-hGBA vector and the pE-mIRES-GS vector were treated with restriction enzymes MluI and NotI, and then gel extracted and purified. The pE-mIRES-GS vector was prepared by the method described in International Publication WO2013/161958. The vectors treated with the restriction enzymes were mixed and ligated for 60 minutes at 16°C using Ligation high Ver.2 to complete the pEmIGS-hGBA vector (Figure 21).
).
The pEmIGS-hGBA vector and the pCIneo vector (Promega) were treated with restriction enzymes MluI and NotI, and gel extracted and purified. Then, the vectors treated with the restriction enzymes were mixed and ligated at 16°C for 60 minutes using Ligation high Ver.2 to complete the wild-type hGBA expression plasmid, the pCIneo-hGBA vector (Figure 22).
〔実施例15〕HSA-hGBA発現プラスミド及びhGBA-HSA発現プラスミドの作製
 野生型HSA(配列番号3)のC末端と野生型hGBA(配列番号37)のN末端とを配列番号9で示されるリンカー配列を介して結合させた融合蛋白質である,配列番号39で示されるHSA-hGBAをコードする遺伝子を含む,配列番号40で示される塩基配列を有するDNA断片を合成した。これをMluI及びNotIで制限酵素処理し,アガロースゲル電気泳動にて分離した。EtBr染色を行った後,UV照射下で目的のDNA断片を含むバンドを切り出し,QIAEX II Gel Extraction Kitを用いてゲルからDNAを抽出した。同様に,pCI-neoベクター(プロメガ社)もMluI及びNotIで制限酵素処理し,ゲル抽出,精製を行った。制限酵素で処理したベクター及びDNA断片を混合し,Ligation high Ver.2を用いて16℃で60分間ライゲーション反応を行い,HSA-hGBA発現プラスミドであるpCIneo-HSA-hGBAベクターを完成させた(図23)。野生型hGBA(配列番号37)のC末端と野生型HSA(配列番号3)のN末端とを配列番号9で示されるリンカー配列を介して結合させた融合蛋白質である,配列番号41で示されるhGBA-HSAをコードする遺伝子を含む,配列番号42で示される塩基配列を有するDNA断片についても,同様にライゲーション反応を行い,hGBA-HSA発現プラスミドであるpCIneo-hGBA-HSAベクターを完成させた。
Example 15: Preparation of HSA-hGBA expression plasmid and hGBA-HSA expression plasmid A DNA fragment having a base sequence shown in SEQ ID NO: 40 was synthesized, including a gene encoding HSA-hGBA shown in SEQ ID NO: 39, which is a fusion protein in which the C-terminus of wild-type HSA (SEQ ID NO: 3) and the N-terminus of wild-type hGBA (SEQ ID NO: 37) are linked via a linker sequence shown in SEQ ID NO: 9. This was treated with restriction enzymes MluI and NotI and separated by agarose gel electrophoresis. After EtBr staining, a band containing the target DNA fragment was excised under UV irradiation, and DNA was extracted from the gel using a QIAEX II Gel Extraction Kit. Similarly, pCI-neo vector (Promega) was also treated with restriction enzymes MluI and NotI, and gel extracted and purified. The vector and DNA fragment treated with the restriction enzyme were mixed and ligated for 60 minutes at 16°C using Ligation high Ver.2 to complete the pCIneo-HSA-hGBA vector, an HSA-hGBA expression plasmid (Figure 23). A ligation reaction was also carried out in the same manner with a DNA fragment having the base sequence shown in SEQ ID NO: 42, which contains a gene encoding hGBA-HSA shown in SEQ ID NO: 41, a fusion protein in which the C-terminus of wild-type hGBA (SEQ ID NO: 37) and the N-terminus of wild-type HSA (SEQ ID NO: 3) are linked via a linker sequence shown in SEQ ID NO: 9, to complete the pCIneo-hGBA-HSA vector, an hGBA-HSA expression plasmid.
〔実施例16〕各プラスミドの確認及び精製
 実施例14及び実施例15で得られたpCIneo-hGBAベクター,pCIneo-HSA-hGBAベクター,及びpCIneo-hGBA-HSAベクターをそれぞれ含む各ライゲーション反応液を用いて大腸菌(E. coli DH5α Competent Cells,タカラバイオ社)をそれぞれ形質転換した。得られた形質転換体から,実施例1に記載と同様の手法により,野生型hGBA,HSA-hGBA,及びhGBA-HSAが組込まれている各プラスミドをそれぞれ精製した。
[Example 16] Confirmation and purification of each plasmid E. coli (E. coli DH5α Competent Cells, Takara Bio Inc.) was transformed with each ligation reaction solution containing the pCIneo-hGBA vector, pCIneo-HSA-hGBA vector, and pCIneo-hGBA-HSA vector obtained in Examples 14 and 15. From the obtained transformants, each of the plasmids incorporating wild-type hGBA, HSA-hGBA, and hGBA-HSA was purified by the same method as described in Example 1.
〔実施例17〕野生型hGBA,HSA-hGBA,及びhGBA-HSAの一過性発現
 野生型hGBA,HSA-hGBA,及びhGBA-HSAの一過性発現には,実施例16で得た精製したpCIneo-hGBAベクター,pCIneo-HSA-hGBAベクター,及びpCIneo-hGBA-HSAベクターを用いた。
[Example 17] Transient expression of wild-type hGBA, HSA-hGBA, and hGBA-HSA For transient expression of wild-type hGBA, HSA-hGBA, and hGBA-HSA, the purified pCIneo-hGBA vector, pCIneo-HSA-hGBA vector, and pCIneo-hGBA-HSA vector obtained in Example 16 were used.
 ExpiCHOTM Expression System(Thermo Fisher Scientific社)のHigh titerプロトコールに従い,pCIneo-hGBAベクター,pCIneo-HSA-hGBAベクター,及びpCIneo-hGBA-HSAベクターを用いて,ExpiCHO細胞を形質転換させた。形質転換後,細胞を8日間培養し,野生型hGBA,HSA-hGBA,及びhGBA-HSAをそれぞれ培養上清中に発現させた。8日後の培養液をそれぞれ遠心分離して培養上清を回収した。 ExpiCHO cells were transformed with the pCIneo-hGBA vector, pCIneo-HSA-hGBA vector, and pCIneo-hGBA-HSA vector according to the High titer protocol of the ExpiCHO TM Expression System (Thermo Fisher Scientific). After transformation, the cells were cultured for 8 days, and wild-type hGBA, HSA-hGBA, and hGBA-HSA were expressed in the culture supernatant. After 8 days, the culture medium was centrifuged to collect the culture supernatant.
〔実施例18〕一過性発現による野生型hGBA,HSA-hGBA,及びhGBA-HSAの発現量の確認(酵素活性測定)
 試料溶液として,実施例17で得られた培養上清を0.4 M KH2PO4/ 0.4 M K2HPO4/ 0.125% Na-taurocholate/ 0.15% TrionX-100/ 0.1% BSA溶液で適宜希釈して測定に供した。標準溶液として,4-MU(4-Methylumbelliferone,シグマアルドリッチ社)を0.4 M KH2PO4/ 0.4 M K2HPO4/ 0.125% Na-taurocholate/ 0.15% TrionX-100/ 0.1% BSA溶液で200~3.13 μMに段階希釈したものを調製した。基質溶液として,hGBAの人工基質である4-Methylumbelliferyl-β-D-glucopyranoside(シグマアルドリッチ社)を0.4 M KH2PO4/ 0.4 M K2HPO4/ 0.125% Na-taurocholate/ 0.15% TrionX-100/ 0.1% BSA溶液で4 mmol/Lに希釈したものを調製した。マイクロプレートに試料溶液又は標準溶液を10 μL/well添加し,更に各ウェルに70 μLの基質溶液を添加してプレートシェイカーで撹拌し混合した。プレートを37℃で1時間静置した後,各ウェルに200 μLの200 mM グリシン/ 50 mM Na2CO3緩衝液(pH 10.7)を添加して反応を停止させた。次いで,蛍光プレートリーダー(Gemini XPS,モレキュラーデバイス社)を用いて各ウェルの蛍光強度を測定した(励起波長365 nm,蛍光波長460 nm)。この蛍光強度は,各ウェル中の溶液に含まれる4-MU(4-Methylumbelliferone)の濃度に比例する。標準溶液の測定結果に基づき検量線を作成し,これに各試料溶液の測定値を内挿して,酵素活性量(μM/時間)を求めた。なお,この酵素活性量の単位は,1時間当たりに分解される基質の量を示すものである。
[Example 18] Confirmation of expression levels of wild-type hGBA, HSA-hGBA, and hGBA-HSA by transient expression (enzyme activity measurement)
The culture supernatant obtained in Example 17 was appropriately diluted with 0.4 M KH2PO4 / 0.4 M K2HPO4 /0.125% Na-taurocholate/0.15% TrionX-100/0.1% BSA solution as a sample solution and used for measurement. As a standard solution, 4-MU (4-Methylumbelliferone, Sigma - Aldrich) was serially diluted from 200 to 3.13 μM with 0.4 M KH2PO4 / 0.4 M K2HPO4/0.125% Na-taurocholate/0.15% TrionX-100/0.1% BSA solution to prepare a standard solution. Substrate solution was prepared by diluting 4-Methylumbelliferyl-β-D-glucopyranoside (Sigma-Aldrich), an artificial substrate for hGBA, to 4 mmol/L with 0.4 M KH 2 PO 4 / 0.4 M K 2 HPO 4 / 0.125% Na-taurocholate / 0.15% TrionX-100 / 0.1% BSA solution. 10 μL/well of sample solution or standard solution was added to a microplate, and 70 μL of substrate solution was added to each well and mixed by shaking on a plate shaker. After leaving the plate at 37°C for 1 hour, 200 μL of 200 mM glycine / 50 mM Na2CO3 buffer (pH 10.7) was added to each well to stop the reaction. Next, the fluorescence intensity of each well was measured using a fluorescence plate reader (Gemini XPS, Molecular Devices) (excitation wavelength 365 nm, fluorescence wavelength 460 nm). This fluorescence intensity is proportional to the concentration of 4-MU (4-Methylumbelliferone) contained in the solution in each well. A calibration curve was created based on the measurement results of the standard solution, and the measured values of each sample solution were interpolated to determine the enzyme activity (μM/hour). The unit of enzyme activity indicates the amount of substrate decomposed per hour.
〔実施例19〕一過性発現による野生型hGBA,HSA-hGBA,及びhGBA-HSAの発現量の確認(SDS page電気泳動)
 実施例17で得られた培養上清それぞれ10 μLを,最終濃度が0.4 MとなるようDTTを添加した10 μLの2×Sample Buffer(バイオラッド社)と混合し,100℃で5分間加熱することにより還元条件下で熱変性させた。熱変性後の溶液を,0.1% SDSを含む50 mM Tris緩衝液/380 mM グリシン緩衝液(pH 8.3)内に設置した5-20%ポリアクリルアミドゲルのウェルに15 μLずつ供し,25 mA定電流にて電気泳動した。電気泳動後のゲルをOriole Fluorescent Gel Stain(バイオラッド社)に浸し,室温で60分間振とうした。ゲルを純水で洗浄した後,ルミノイメージアナライザー(Amersham Imager 600RGB,サイティバ社)を用いて各蛋白質に相当するバンドが可視化された電気泳動画像を得た。
[Example 19] Confirmation of the expression levels of wild-type hGBA, HSA-hGBA, and hGBA-HSA by transient expression (SDS page electrophoresis)
10 μL of each culture supernatant obtained in Example 17 was mixed with 10 μL of 2×Sample Buffer (Bio-Rad) to which DTT had been added so that the final concentration was 0.4 M, and the mixture was heat-denatured under reducing conditions by heating at 100° C. for 5 minutes. 15 μL of the heat-denatured solution was applied to wells of a 5-20% polyacrylamide gel placed in 50 mM Tris buffer/380 mM glycine buffer (pH 8.3) containing 0.1% SDS, and electrophoresis was performed at a constant current of 25 mA. The gel after electrophoresis was immersed in Oriole Fluorescent Gel Stain (Bio-Rad) and shaken at room temperature for 60 minutes. After washing the gel with pure water, an electrophoretic image was obtained in which the bands corresponding to each protein were visualized using a luminoimage analyzer (Amersham Imager 600RGB, Cytiva).
〔実施例20〕一過性発現による野生型hGBA,HSA-hGBA,及びhGBA-HSAの発現量の確認(結果)
 図24に,実施例18で測定した一過性発現による野生型hGBA,HSA-hGBA,及びhGBA-HSAの酵素活性測定の結果を示す。また,酵素活性測定の結果に基づく野生型hGBA,HSA-hGBA,及びhGBA-HSAの相対量を表4に示す。
[Example 20] Confirmation of expression levels of wild-type hGBA, HSA-hGBA, and hGBA-HSA by transient expression (Results)
24 shows the results of enzyme activity measurement of wild-type hGBA, HSA-hGBA, and hGBA-HSA by transient expression measured in Example 18. In addition, the relative amounts of wild-type hGBA, HSA-hGBA, and hGBA-HSA based on the results of enzyme activity measurement are shown in Table 4.
Figure JPOXMLDOC01-appb-T000004
Figure JPOXMLDOC01-appb-T000004
 上記の結果から,培養開始後8日後の発現量(酵素活性量換算)をみると,HSA-hGBA及びhGBA-HSAの発現量は,野生型hGBAのそれと比較して,それぞれ22倍及び18倍の値を示し,野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なhGBAを,HSA-hGBA又はhGBA-HSAの形態で発現させることにより,より多くのhGBAを組換え蛋白質として効率よく発現させることができることがわかった。 The above results show that, when looking at the expression levels (converted into enzyme activity) 8 days after the start of culture, the expression levels of HSA-hGBA and hGBA-HSA were 22 and 18 times higher, respectively, than that of wild-type hGBA. This shows that hGBA is difficult to mass-produce because of its low expression level when expressed as a recombinant in its wild-type form, but by expressing it in the form of HSA-hGBA or hGBA-HSA, it is possible to efficiently express more hGBA as a recombinant protein.
 次に,実施例19で得たSDS-PAGEの泳動画像を図25に示す。SDS-PAGEの結果は,バンド強度の比較からHSA-hGBA及びhGBA-HSAは野生型hGBAより10倍~20倍以上発現量が高く,図24及び表4で示される発現量(酵素活性量換算)とほぼ相関するものであった。すなわち,野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なhGBAを,HSA-hGBA又はhGBA-HSAの形態で発現させることにより,より多くのhGBAを組換え蛋白質として効率よく発現させることができることがわかった。 Next, the SDS-PAGE electrophoresis images obtained in Example 19 are shown in Figure 25. The results of SDS-PAGE show that HSA-hGBA and hGBA-HSA have 10 to 20 times higher expression levels than wild-type hGBA, based on a comparison of band intensities, and are roughly correlated with the expression levels (converted into enzyme activity) shown in Figure 24 and Table 4. In other words, it was found that hGBA, which is difficult to mass-produce due to its low expression level when expressed as a recombinant in its wild form, can be efficiently expressed in greater amounts as a recombinant protein by expressing it in the form of HSA-hGBA or hGBA-HSA.
 以上の結果は,hGBAを組換え蛋白質として製造する場合に,hGBAを,HSA-hGBA又はhGBA-HSAの融合蛋白質の形態で製造することにより,約20倍の分子数のhGBAを得ることができることを示す。つまり,hGBAをHSA-hGBA又はhGBA-HSAの融合蛋白質の形態で発現させる組換えhGBAの製造方法は,大量の組換えhGBAを製造する方法として極めて有効な手段といえる。 The above results indicate that when producing hGBA as a recombinant protein, it is possible to obtain approximately 20 times the number of hGBA molecules by producing hGBA in the form of an HSA-hGBA or hGBA-HSA fusion protein. In other words, the method of producing recombinant hGBA in which hGBA is expressed in the form of an HSA-hGBA or hGBA-HSA fusion protein can be said to be an extremely effective means of producing large quantities of recombinant hGBA.
 以下,実施例21~26はマウス血清アルブミン(MSA)とマウスIL-10(mIL-10)との融合蛋白質に関する。なお,実施例21~26において,MSAは全て配列番号16で示されるアミノ酸配列を有する野生型MSAの,320番目のアミノ酸であるアラニンをトレオニンへと置換した変異型MSA(MSA-A320T,配列番号125)を用いた。 The following Examples 21 to 26 relate to a fusion protein of mouse serum albumin (MSA) and mouse IL-10 (mIL-10). In Examples 21 to 26, the MSA used was a mutant MSA (MSA-A320T, SEQ ID NO: 125) in which the 320th amino acid, alanine, of the wild-type MSA having the amino acid sequence shown in SEQ ID NO: 16 was replaced with threonine.
〔実施例21〕野生型mIL-10発現プラスミド,mIL-10-MSA発現プラスミド及びMSA-mIL-10発現プラスミドの作製
 野生型mIL-10(配列番号126)をコードする遺伝子を含む配列番号127で示される塩基配列を有するDNA断片,野生型mIL-10(配列番号126)のC末端と変異型MSA(配列番号125)のN末端とを直接結合させた融合蛋白質である,配列番号128で示されるmIL-10-MSAをコードする遺伝子を含む,配列番号129で示される塩基配列を有するDNA断片,及び変異型MSA(配列番号125)のC末端と野生型mIL-10(配列番号126)のN末端とを直接結合させた融合蛋白質である,配列番号130で示されるMSA-mIL-10をコードする遺伝子を含む,配列番号131で示される塩基配列を有するDNA断片を合成した。これらをMluIおよびNotI(タカラバイオ社)で制限酵素処理し,アガロースゲル電気泳動にて分離した。EtBr染色を行った後,UV照射下で目的のDNA断片を含むバンドを切り出し,QIAEX II Gel Extraction Kit(QIAGEN社)を用いてゲルからDNAを抽出した。同様に,pCI-neoベクター(プロメガ社)をMluIおよびNotIで制限酵素処理し,ゲル抽出,精製を行った。制限酵素で処理したベクターに,制限酵素で処理した各DNA断片をそれぞれ混合し,DNA Ligation Kit Mighty Mix(タカラバイオ社)を用いて16℃で60分間ライゲーション反応を行った。
Example 21: Preparation of wild-type mIL-10 expression plasmid, mIL-10-MSA expression plasmid, and MSA-mIL-10 expression plasmid A DNA fragment having the nucleotide sequence shown in SEQ ID NO: 127 and including a gene encoding wild-type mIL-10 (SEQ ID NO: 126), a DNA fragment having the nucleotide sequence shown in SEQ ID NO: 129 and including a gene encoding mIL-10-MSA (SEQ ID NO: 128), which is a fusion protein in which the C-terminus of wild-type mIL-10 (SEQ ID NO: 126) and the N-terminus of mutant MSA (SEQ ID NO: 125) are directly linked, and a DNA fragment having the nucleotide sequence shown in SEQ ID NO: 131 and including a gene encoding MSA-mIL-10 (SEQ ID NO: 130), which is a fusion protein in which the C-terminus of mutant MSA (SEQ ID NO: 125) and the N-terminus of wild-type mIL-10 (SEQ ID NO: 126) are directly linked were synthesized. These were treated with restriction enzymes MluI and NotI (Takara Bio Inc.) and separated by agarose gel electrophoresis. After EtBr staining, the band containing the target DNA fragment was excised under UV irradiation, and DNA was extracted from the gel using a QIAEX II Gel Extraction Kit (QIAGEN). Similarly, pCI-neo vector (Promega) was digested with restriction enzymes MluI and NotI, and gel extracted and purified. The restriction enzyme-treated vector was mixed with each DNA fragment, and a ligation reaction was performed at 16°C for 60 minutes using a DNA Ligation Kit Mighty Mix (Takara Bio).
 次いで,各ライゲーション反応液を用いて大腸菌(One Shot MAX Efficiency DH10B T1 Phage-Resistant Cells,Thermo Fisher Scientific社)をそれぞれ形質転換した。得られた形質転換体から,実施例1に記載と同様の手法により,目的遺伝子が組み込まれている各プラスミドをそれぞれ精製した。 Then, each ligation reaction solution was used to transform Escherichia coli (One Shot MAX Efficiency DH10B T1 Phage-Resistant Cells, Thermo Fisher Scientific). Plasmids containing the target genes were purified from the resulting transformants using the same method as described in Example 1.
〔実施例22〕野生型mIL-10,mIL-10-MSA及びMSA-mIL-10の一過性発現
 ExpiCHO Expression System(Thermo Fisher Scientific社)のHigh Titerプロトコールに従い,野生型mIL-10,mIL-10-MSA及びMSA-mIL-10をそれぞれコードする遺伝子を組み込んだプラスミドを用いて,ExpiCHO細胞を形質転換させた。形質転換後,細胞を8日間培養し,野生型mIL-10,mIL-10-MSA及びMSA-mIL-10をそれぞれ培養上清中に発現させた。培養後,培養液を遠心分離して培養上清を回収した。
[Example 22] Transient expression of wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 According to the High Titer protocol of the ExpiCHO Expression System (Thermo Fisher Scientific), ExpiCHO cells were transformed with plasmids incorporating genes encoding wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10, respectively. After transformation, the cells were cultured for 8 days to express wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 in the culture supernatant, respectively. After culturing, the culture medium was centrifuged to collect the culture supernatant.
〔実施例23〕一過性発現による野生型mIL-10,mIL-10-MSA及びMSA-mIL-10の発現量確認(SDS-page電気泳動)
 実施例22で得られた培養上清6 μLを,(±)-ジチオトレイトール(富士フィルム和光純薬社)を40 mg/mLとなるように添加した4×Sample Buffer(バイオラッド社)2 μLと混合し,100℃で3分間加熱することにより還元条件下で熱変性させた。熱変性後の試料を,0.1% SDSを含む50 mM トリス緩衝液/380 mM グリシン緩衝液(pH8.3)内に設置した5-20%ポリアクリルアミドゲル(富士フィルム和光純薬社)のウェルに5 μLずつアプライし,25 mA定電流にて電気泳動した。電気泳動後のゲルをEZFluor UV 1-step Fluorescent Protein Gel Stain(メルク社)に浸し,室温で60分間振とうした。ゲルを純水で洗浄した後,ルミノイメージアナライザー(Amersham ImageQuan 800,Cytiva社)で蛋白質のバンドを検出した。
[Example 23] Confirmation of expression levels of wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 by transient expression (SDS-page electrophoresis)
6 μL of the culture supernatant obtained in Example 22 was mixed with 2 μL of 4×Sample Buffer (Bio-Rad) to which (±)-dithiothreitol (Fujifilm Wako Pure Chemical Industries, Ltd.) had been added to a concentration of 40 mg/mL, and the mixture was heated at 100°C for 3 minutes to be thermally denatured under reducing conditions. 5 μL of the thermally denatured sample was applied to wells of a 5-20% polyacrylamide gel (Fujifilm Wako Pure Chemical Industries, Ltd.) placed in 50 mM Tris buffer/380 mM glycine buffer (pH 8.3) containing 0.1% SDS, and electrophoresis was performed at a constant current of 25 mA. The gel after electrophoresis was immersed in EZFluor UV 1-step Fluorescent Protein Gel Stain (Merck) and shaken at room temperature for 60 minutes. After washing the gel with pure water, protein bands were detected using a luminometer image analyzer (Amersham ImageQuan 800, Cytiva).
〔実施例24〕一過性発現による野生型mIL-10,mIL-10-MSA及びMSA-mIL-10の発現量確認(ELISA)
 Mouse IL-10 ELISA Kit(プロテインテック社)を用いて,ELISAを行った。実施例22で得られた培養上清をキット付属のSample Diluentで50000倍希釈し,試料溶液とした。野生型mIL-10についてはキット付属のStandardをSample Diluentで希釈したものを標準溶液とし,mIL-10-MSA及びMSA-mIL-10については下記の実施例25に示す方法で得られたそれぞれの精製品をSample Diluentで希釈したものを標準溶液とした。抗マウスIL-10抗体が固相化された96 wellプレートの各ウェルに試料溶液または標準溶液を100 μLを添加し,37℃で100分間保温した。次いでキット付属のWash Buffer 300 μLで各ウェルを4回洗浄後,mouse IL-10 detection antibodyを100 μLずつ各ウェルに添加し,37℃で60分間保温した。Wash Buffer 300 μLで各ウェルを4回洗浄後,Streptavidin-HRPを100 μLずつ各ウェルに添加し,37℃で40分間保温した。Wash Buffer 300 μLで各ウェルを4回洗浄後,TMB substrateを100 μLずつ各ウェルに添加し,37℃で発色反応させた。充分な発色が得られた後,TMB stop solutionを100 μLずつ各ウェルに添加して反応を停止させた。プレートリーダー(SpectraMax iD3,Molecular Device社)で波長450 nmにおける吸光度を測定し,解析ソフトウェア(SoftMax Pro 7.1,Molecular Device社)を用いて検量線を作成し,これに各試料溶液の測定値を内挿して,各培養上清中の濃度(物質量に換算)を算出した。
[Example 24] Confirmation of expression levels of wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 by transient expression (ELISA)
ELISA was performed using Mouse IL-10 ELISA Kit (Proteintech). The culture supernatant obtained in Example 22 was diluted 50,000 times with Sample Diluent included in the kit to prepare a sample solution. For wild-type mIL-10, the Standard included in the kit was diluted with Sample Diluent to prepare a standard solution, and for mIL-10-MSA and MSA-mIL-10, the purified products obtained by the method shown in Example 25 below were diluted with Sample Diluent to prepare standard solutions. 100 μL of the sample solution or standard solution was added to each well of a 96-well plate on which anti-mouse IL-10 antibody was immobilized, and the plate was incubated at 37° C. for 100 minutes. Next, each well was washed four times with 300 μL of Wash Buffer included in the kit, and 100 μL of mouse IL-10 detection antibody was added to each well, and the plate was incubated at 37° C. for 60 minutes. After washing each well four times with 300 μL of wash buffer, 100 μL of streptavidin-HRP was added to each well and incubated at 37°C for 40 minutes. After washing each well four times with 300 μL of wash buffer, 100 μL of TMB substrate was added to each well and the color reaction was allowed to proceed at 37°C. After sufficient color development was achieved, 100 μL of TMB stop solution was added to each well to stop the reaction. The absorbance at a wavelength of 450 nm was measured using a plate reader (SpectraMax iD3, Molecular Devices), and a calibration curve was created using analysis software (SoftMax Pro 7.1, Molecular Devices), and the measured values of each sample solution were inserted into this to calculate the concentration (converted to the amount of substance) in each culture supernatant.
〔実施例25〕mIL-10-MSA及びMSA-mIL-10の精製品取得
 実施例24で標準溶液調製のために用いたmIL-10-MSA及びMSA-mIL-10の精製品は,下記の方法に従って得た。
[Example 25] Obtaining purified products of mIL-10-MSA and MSA-mIL-10 The purified products of mIL-10-MSA and MSA-mIL-10 used for preparing the standard solution in Example 24 were obtained according to the following method.
〔mIL-10-MSA発現プラスミド及びMSA-mIL-10発現プラスミドの作製〕
 実施例21で作製したmIL-10-MSA発現プラスミド及びMSA-mIL-10発現プラスミドをMlu IおよびNot I(タカラバイオ社)で制限酵素処理し,mIL-10-MSA遺伝子またはMSA-mIL-10遺伝子の塩基配列を含むDNA断片をアガロースゲル電気泳動にて分離した。EtBr染色を行った後,UV照射下で目的のDNA断片を含むバンドを切り出し,FastGene Gel/PCR Extraction Kit(日本ジェネティクス社)を用いてゲルからDNAを抽出した。同様に,pE-mIRES-GS-puroベクターをMluIおよびNotIで制限酵素処理し,ゲル抽出,精製を行った。なお,mIRES-GS-puroベクターはWO2013/161958に記載の方法により構築した。制限酵素で処理したベクターに,制限酵素で処理した各DNA断片をそれぞれ混合し,Ligation high Ver.2(東洋紡社)を用いて16℃で30~60分間ライゲーション反応を行った。
[Construction of mIL-10-MSA expression plasmid and MSA-mIL-10 expression plasmid]
The mIL-10-MSA expression plasmid and MSA-mIL-10 expression plasmid prepared in Example 21 were treated with restriction enzymes Mlu I and Not I (Takara Bio), and DNA fragments containing the base sequence of the mIL-10-MSA gene or MSA-mIL-10 gene were separated by agarose gel electrophoresis. After EtBr staining, the band containing the target DNA fragment was excised under UV irradiation, and DNA was extracted from the gel using FastGene Gel/PCR Extraction Kit (Nihon Genetics). Similarly, the pE-mIRES-GS-puro vector was treated with restriction enzymes Mlu I and Not I, and gel extraction and purification were performed. The mIRES-GS-puro vector was constructed by the method described in WO2013/161958. The vector treated with the restriction enzymes and each DNA fragment treated with the restriction enzymes were mixed, and ligation reaction was performed at 16°C for 30 to 60 minutes using Ligation high Ver.2 (Toyobo Co., Ltd.).
 次いで,各ライゲーション反応液を用いて大腸菌(E. coli DH5α Competent Cells,タカラバイオ社)をそれぞれ形質転換した。得られた形質転換体から,実施例1に記載と同様の手法により,目的遺伝子が組み込まれている各プラスミドをそれぞれ精製した。 Then, each ligation reaction solution was used to transform Escherichia coli (E. coli DH5α Competent Cells, Takara Bio Inc.). From the resulting transformants, plasmids incorporating the target genes were purified using the same method as described in Example 1.
〔mIL-10-MSA発現バルク細胞及びMSA-mIL-10発現バルク細胞の作製〕
 遺伝子導入装置(スーパーエレクトロポレーターNEPA21,ネッパジーン社)を用いて,CHO-K1細胞の無血清馴化株に,上記で得られたmIL-10-MSA又はMSA-mIL-10をコードする遺伝子を組み込んだ発現プラスミドをそれぞれ導入し,メチオニンスルホキシミン(SIGMA社)及びPuromycin(Thermo Fisher Scientific社)を含むCD OptiCHO培地(Thermo Fisher Scientific社)にて選択培養を行った。選択培養の際には,メチオニンスルホキシミン及びPuromycinの濃度を段階的に上昇させて,最終的にメチオニンスルホキシミンの濃度を300 μM,Puromycinの濃度を10 μg/mLとした。3~4日おきに培地交換を繰り返しながら順次培養液量を拡大し,培養中の細胞の生存率が90%を超えた時点で細胞を回収し,これをmIL-10-MSA発現バルク細胞及びMSA-mIL-10発現バルク細胞とした。
[Preparation of mIL-10-MSA expressing bulk cells and MSA-mIL-10 expressing bulk cells]
Using a gene transfer device (Super Electroporator NEPA21, Nepa Gene), expression plasmids incorporating genes encoding mIL-10-MSA or MSA-mIL-10 obtained above were introduced into serum-free adapted CHO-K1 cells, and selective culture was performed in CD OptiCHO medium (Thermo Fisher Scientific) containing methionine sulfoximine (SIGMA) and Puromycin (Thermo Fisher Scientific). During selective culture, the concentrations of methionine sulfoximine and Puromycin were gradually increased to a final concentration of 300 μM for methionine sulfoximine and 10 μg/mL for Puromycin. The culture medium was replaced every 3 to 4 days, and the amount of culture medium was gradually increased. When the survival rate of the cells during culture exceeded 90%, the cells were harvested and used as mIL-10-MSA-expressing bulk cells and MSA-mIL-10-expressing bulk cells.
〔mIL-10-MSA発現バルク細胞及びMSA-mIL-10発現バルク細胞の培養〕
 300 μMメチオニンスルホキシミン及び10 μg/mL Puromycinを含むCD OptiCHO培地に,上記で得られたmIL-10-MSA発現バルク細胞及びMSA-mIL-10発現バルク細胞を,それぞれ2×105 cells/mLの細胞密度で播種し,37℃,5% CO2存在下で振とう培養した。MSA-mIL-10発現バルク細胞は培養開始から3日目以降は32℃条件下で培養した。培養開始から10日後に培養上清を遠心分離して培養上清を回収した。
[Cultivation of mIL-10-MSA expressing bulk cells and MSA-mIL-10 expressing bulk cells]
The mIL-10-MSA-expressing bulk cells and MSA-mIL-10-expressing bulk cells obtained above were seeded at a cell density of 2 x 105 cells/mL in CD OptiCHO medium containing 300 μM methionine sulfoximine and 10 μg/mL puromycin, and cultured with shaking at 37°C in the presence of 5% CO2 . The MSA-mIL-10-expressing bulk cells were cultured at 32°C from the third day onwards. The culture supernatant was collected by centrifugation 10 days after the start of culture.
〔mIL-10-MSA及びMSA-mIL-10の精製〕
 上記で回収したmIL-10-MSA発現バルク細胞の培養上清をRapid-Flow PESメンブレンフィルターユニット(孔径0.2 μm,Thermo Fisher Scientific社)によりろ過し,100 mM NaClを含有する25 mM HEPES緩衝液(pH7.4)で平衡化したHiTrap Q HPカラム(Cytiva社)に通してmIL-10-MSAを樹脂に吸着させた。続いて100 mM NaClを含有する25 mM HEPES緩衝液(pH7.4)でカラムを洗浄し,350 mM NaClを含有する25 mM HEPES緩衝液(pH7.4)でmIL-10-MSAを樹脂から溶出させた。
Purification of mIL-10-MSA and MSA-mIL-10
The culture supernatant of the mIL-10-MSA-expressing bulk cells collected above was filtered through a Rapid-Flow PES membrane filter unit (pore size 0.2 μm, Thermo Fisher Scientific) and passed through a HiTrap Q HP column (Cytiva) equilibrated with 25 mM HEPES buffer (pH 7.4) containing 100 mM NaCl to adsorb mIL-10-MSA to the resin. The column was then washed with 25 mM HEPES buffer (pH 7.4) containing 100 mM NaCl, and mIL-10-MSA was eluted from the resin with 25 mM HEPES buffer (pH 7.4) containing 350 mM NaCl.
 mIL-10-MSAを含むHiTrap Q HPカラムからの溶出液を5 mMリン酸を含む25 mM MES緩衝液(pH6.8)で6倍に希釈し,これを5 mMリン酸を含む25 mM MES緩衝液(pH6.8)で平衡化したCHT TypeIカラム(バイオラッド社)に通してmIL-10-MSAを樹脂に吸着させた。続いて5 mMリン酸を含む25 mM MES緩衝液(pH6.8)でカラムを洗浄し,200 mMリン酸を含む25 mM MES緩衝液(pH6.8)でmIL-10-MSAを樹脂から溶出させ,mIL-10-MSA精製品とした。同様にして,MSA-mIL-10を精製し,MSA-mIL-10精製品とした。 The eluate from the HiTrap Q HP column containing mIL-10-MSA was diluted 6-fold with 25 mM MES buffer (pH 6.8) containing 5 mM phosphate, and passed through a CHT Type I column (Bio-Rad) equilibrated with 25 mM MES buffer (pH 6.8) containing 5 mM phosphate to adsorb mIL-10-MSA to the resin. The column was then washed with 25 mM MES buffer (pH 6.8) containing 5 mM phosphate, and mIL-10-MSA was eluted from the resin with 25 mM MES buffer (pH 6.8) containing 200 mM phosphate to obtain the mIL-10-MSA purified product. MSA-mIL-10 was purified in the same manner to obtain the MSA-mIL-10 purified product.
〔実施例26〕一過性発現による野生型mIL-10,mIL-10-MSA及びMSA-mIL-10の発現量の確認(結果)
 図26に,実施例24で測定した一過性発現による野生型mIL-10,mIL-10-MSA及びMSA-mIL-10のELISA濃度測定の結果を示す。また,ELISA濃度測定の結果に基づく野生型mIL-10,mIL-10-MSA及びMSA-mIL-10の相対量を表5に示す。
[Example 26] Confirmation of expression levels of wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 by transient expression (Results)
26 shows the results of ELISA concentration measurement of wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 by transient expression measured in Example 24. In addition, the relative amounts of wild-type mIL-10, mIL-10-MSA, and MSA-mIL-10 based on the results of ELISA concentration measurement are shown in Table 5.
Figure JPOXMLDOC01-appb-T000005
Figure JPOXMLDOC01-appb-T000005
 上記の結果から,培養開始後8日後の発現量(濃度)をみると,mIL-10-MSA及びMSA-mIL-10の発現量は,野生型mIL-10のそれと比較して,それぞれ4.5倍及び4.2倍の値を示し,野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なIL-10を,IL-10-SA又はSA-IL-10の形態で発現させることにより,より多くのIL-10を組換え蛋白質として効率よく発現させることができることがわかった。 The above results show that, looking at the expression levels (concentrations) 8 days after the start of culture, the expression levels of mIL-10-MSA and MSA-mIL-10 were 4.5 and 4.2 times higher, respectively, than that of wild-type mIL-10. This shows that wild-type IL-10 is difficult to mass-produce due to its low expression level when expressed as a recombinant form, but by expressing it in the form of IL-10-SA or SA-IL-10, it is possible to efficiently express more IL-10 as a recombinant protein.
 次に,実施例23で得たSDS-PAGEの泳動画像を図27に示す。SDS-PAGEの結果は,バンド強度の比較からmIL-10-MSA及びMSA-mIL-10は野生型mIL-10より発現量が高く,図26及び表5で示されるELISAによる測定値に対応するものであった。野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なIL-10を,IL-10-SA又はSA-IL-10の形態で発現させることにより,より多くのIL-10を組換え蛋白質として効率よく発現させることができることがわかった。 Next, the electrophoretic image of SDS-PAGE obtained in Example 23 is shown in Figure 27. The results of SDS-PAGE showed that mIL-10-MSA and MSA-mIL-10 had higher expression levels than wild-type mIL-10, based on a comparison of band intensities, and corresponded to the measured values by ELISA shown in Figure 26 and Table 5. IL-10, which is expressed in low amounts when expressed as a recombinant in its wild form and is difficult to mass-produce, was found to be able to be efficiently expressed in greater amounts as a recombinant protein by expressing it in the form of IL-10-SA or SA-IL-10.
 以上の結果は,mIL-10を組換え蛋白質として製造する場合に,mIL-10を,mIL-10-MSA又はMSA-mIL-10の融合蛋白質の形態で製造することにより,約4倍の分子数のmIL-10を得ることができることを示すものであり,さらにhIL-10を組換え蛋白質として製造する場合に,hIL-10をhIL-10-HSA又はHSA-hIL-10の融合蛋白質の形態で製造することにより,より多くの分子数のhIL-10を得ることができることを示すものである。つまり,IL-10をIL-10-SA又はSA-IL-10の融合蛋白質の形態で発現させる組換えIL-10の製造方法は,大量の組換えIL-10を製造する方法として極めて有効な手段といえる。 The above results indicate that when mIL-10 is produced as a recombinant protein, approximately four times the number of molecules of mIL-10 can be obtained by producing mIL-10 in the form of a mIL-10-MSA or MSA-mIL-10 fusion protein, and further indicate that when hIL-10 is produced as a recombinant protein, a greater number of molecules of hIL-10 can be obtained by producing hIL-10 in the form of a hIL-10-HSA or HSA-hIL-10 fusion protein. In other words, the method of producing recombinant IL-10 in which IL-10 is expressed in the form of an IL-10-SA or SA-IL-10 fusion protein can be said to be an extremely effective means of producing large amounts of recombinant IL-10.
 以下,実施例27~33はHSAとヒト神経栄養因子(hBDNF,hNGF,hNT-3,又はhNT-4)との融合蛋白質に関する。 Below, Examples 27 to 33 relate to fusion proteins of HSA and human neurotrophic factors (hBDNF, hNGF, hNT-3, or hNT-4).
〔実施例27〕野生型ヒト神経栄養因子発現プラスミドの作製
 以下の(1)~(4)のDNA断片を合成した:
(1)C末端にGly-Serで示されるアミノ酸配列(以下GSという)及び6個のヒスチジンからなる配列番号195のアミノ酸配列を有するヒスチジンタグ(以下His6という)をこの順に付加した野生型hBDNF(配列番号132)をコードする遺伝子を含む配列番号133で示される塩基配列を有するDNA断片,
(2)C末端にHis6を付加した野生型hNGF(配列番号134)をコードする遺伝子を含む配列番号135で示される塩基配列を有するDNA断片,
(3)C末端にHis6を付加した野生型hNT-4(配列番号136)をコードする遺伝子を含む配列番号137で示される塩基配列を有するDNA断片,及び
(4)C末端にHis6を付加した野生型hNT-3(配列番号138)をコードする遺伝子を含む配列番号139で示される塩基配列を有するDNA断片。
[Example 27] Preparation of wild-type human neurotrophic factor expression plasmid The following DNA fragments (1) to (4) were synthesized:
(1) A DNA fragment having a base sequence shown in SEQ ID NO: 133, which contains a gene encoding wild-type hBDNF (SEQ ID NO: 132) to which an amino acid sequence shown by Gly-Ser (hereinafter referred to as GS) and a histidine tag having the amino acid sequence of SEQ ID NO: 195 consisting of six histidines (hereinafter referred to as His6) have been added in this order at the C-terminus,
(2) a DNA fragment having the base sequence shown in SEQ ID NO: 135, which contains a gene encoding wild-type hNGF (SEQ ID NO: 134) with His6 added to the C-terminus;
(3) A DNA fragment having the base sequence shown in SEQ ID NO: 137, which contains a gene encoding wild-type hNT-4 (SEQ ID NO: 136) with His6 added to the C-terminus, and (4) a DNA fragment having the base sequence shown in SEQ ID NO: 139, which contains a gene encoding wild-type hNT-3 (SEQ ID NO: 138) with His6 added to the C-terminus.
 上記(1)~(4)のDNA断片をMluIおよびNotI(タカラバイオ社)で制限酵素処理し,アガロースゲル電気泳動にて分離した。EtBr染色を行った後,UV照射下で目的のDNA断片を含むバンドを切り出し,QIAEX II Gel Extraction Kit(QIAGEN社)を用いてゲルからDNAを抽出した。同様に,pCI-neoベクター(プロメガ社)をMluI及びNotIで制限酵素処理し,ゲル抽出,精製を行った。制限酵素で処理したベクターに,制限酵素で処理した各DNA断片をそれぞれ混合し,Ligation Mix(タカラバイオ社)を用いて16℃で30~60分間ライゲーション反応を行った。 The DNA fragments (1) to (4) above were treated with restriction enzymes MluI and NotI (Takara Bio) and separated by agarose gel electrophoresis. After EtBr staining, the band containing the target DNA fragment was excised under UV irradiation, and the DNA was extracted from the gel using a QIAEX II Gel Extraction Kit (QIAGEN). Similarly, pCI-neo vector (Promega) was treated with restriction enzymes MluI and NotI, and gel extracted and purified. The restriction enzyme-treated vector was mixed with each of the DNA fragments, and a ligation reaction was carried out at 16°C for 30 to 60 minutes using Ligation Mix (Takara Bio).
 次いで,各ライゲーション反応液を用いて大腸菌(ECOSTM X Competent E. coli DH5α,ニッポンジーン社)をそれぞれ形質転換した。得られた形質転換体から,実施例1に記載と同様の手法により,目的遺伝子が組み込まれている各プラスミドをそれぞれ精製した。精製した各ベクターをそれぞれ野生型hBDNF発現ベクター,野生型hNGF発現ベクター,野生型hNT-4発現ベクター,野生型hNT-3発現ベクターとして以下の実験で用いた。 Next, each ligation reaction solution was used to transform Escherichia coli (ECOS X Competent E. coli DH5α, Nippon Gene Co., Ltd.). From the obtained transformants, each plasmid incorporating the target gene was purified by the same method as described in Example 1. Each purified vector was used in the following experiments as a wild-type hBDNF expression vector, a wild-type hNGF expression vector, a wild-type hNT-4 expression vector, and a wild-type hNT-3 expression vector.
〔実施例28〕HSAとヒト神経栄養因子との融合蛋白質を発現するプラスミドの作製
 実施例27で合成した(1)~(4)のDNA断片をMluI及びBamHI(タカラバイオ社)で制限酵素処理し,アガロースゲル電気泳動にて分離した

EtBr染色を行った後,UV照射下で目的のDNA断片を含むバンドを切り出し,QIAEX II Gel Extraction Kit(QIAGEN社)を用いてゲルからDNA断片を抽出して精製した。精製したDNA断片をインサートDNA1とした。
[Example 28] Preparation of a plasmid expressing a fusion protein of HSA and human neurotrophic factor The DNA fragments (1) to (4) synthesized in Example 27 were treated with restriction enzymes MluI and BamHI (Takara Bio Inc.) and separated by agarose gel electrophoresis.
After EtBr staining, the band containing the target DNA fragment was excised under UV irradiation, and the DNA fragment was extracted from the gel and purified using a QIAEX II Gel Extraction Kit (QIAGEN). The purified DNA fragment was designated insert DNA 1.
 以下の(1)~(5)のDNA断片を合成した:
(1)C末端にHis6を付加した野生型HSA(配列番号140)をコードする遺伝子を含む配列番号141で示される塩基配列を有するDNA断片,
(2)C末端にHis6を付加した野生型hBDNF(配列番号132)をコードする遺伝子を含む配列番号133で示される塩基配列を有するDNA断片,
(3)C末端にHis6を付加した野生型hNGF(配列番号134)をコードする遺伝子を含む配列番号135で示される塩基配列を有するDNA断片,
(4)C末端にHis6を付加した野生型hNT-4(配列番号136)をコードする遺伝子を含む配列番号137で示される塩基配列を有するDNA断片,及び
(5)C末端にHis6を付加した野生型hNT-3(配列番号138)をコードする遺伝子を含む配列番号139で示される塩基配列を有するDNA断片。
 上記(1)~(5)のDNA断片を鋳型として,表6に示すフォワードプライマー及びリバースプライマーを用いて,No. 1~5のPCR反応を行った。なお,便宜上,C末端にHis6を付加した野生型HSAも,以下野生型HSAと記載する。野生型hBDNF,野生型hNGF,野生型hNT-4,及び野生型hNT-3についても同様とする。
The following DNA fragments (1) to (5) were synthesized:
(1) A DNA fragment having the base sequence shown in SEQ ID NO: 141, which contains a gene encoding wild-type HSA (SEQ ID NO: 140) with His6 added to the C-terminus,
(2) a DNA fragment having the base sequence shown in SEQ ID NO: 133, which contains a gene encoding wild-type hBDNF (SEQ ID NO: 132) with His6 added to the C-terminus;
(3) A DNA fragment having the base sequence shown in SEQ ID NO: 135, which contains a gene encoding wild-type hNGF (SEQ ID NO: 134) with His6 added to the C-terminus.
(4) A DNA fragment having the base sequence shown in SEQ ID NO: 137, which contains a gene encoding wild-type hNT-4 (SEQ ID NO: 136) with His6 added to the C-terminus, and (5) a DNA fragment having the base sequence shown in SEQ ID NO: 139, which contains a gene encoding wild-type hNT-3 (SEQ ID NO: 138) with His6 added to the C-terminus.
Using the DNA fragments (1) to (5) above as templates, PCR reactions No. 1 to 5 were carried out using the forward and reverse primers shown in Table 6. For convenience, wild-type HSA with His6 added to the C-terminus will hereinafter be referred to as wild-type HSA. The same applies to wild-type hBDNF, wild-type hNGF, wild-type hNT-4, and wild-type hNT-3.
Figure JPOXMLDOC01-appb-T000006
Figure JPOXMLDOC01-appb-T000006
 上記No. 1~5のPCRで得られたDNA断片をBglIIおよびNotI(タカラバイオ社)で制限酵素処理し,ゲル抽出,精製を行った。精製したDNA断片をインサートDNA2とした。 The DNA fragments obtained by PCR No. 1 to 5 above were treated with restriction enzymes BglII and NotI (Takara Bio), gel extracted, and purified. The purified DNA fragment was designated insert DNA 2.
 pCI-neoベクター(プロメガ社)をMluI及びNotIで制限酵素処理し,ゲル抽出,精製を行った。制限酵素で処理したベクターに,上記のインサートDNA1及びインサートDNA2を表7に示す組み合わせでそれぞれ混合し,Ligation Mix(タカラバイオ社)を用いて16℃で30~60分間ライゲーション反応を行った。この反応により,pCI-neoベクターに,インサートDNA1の3’末端にインサートDNA2がインフレームで連結したDNA断片が挿入される。  pCI-neo vector (Promega) was treated with restriction enzymes MluI and NotI, and gel extracted and purified. The above insert DNA 1 and insert DNA 2 were mixed with the restriction enzyme-treated vector in the combinations shown in Table 7, and a ligation reaction was carried out at 16°C for 30 to 60 minutes using Ligation Mix (Takara Bio). As a result of this reaction, a DNA fragment in which insert DNA 2 is linked in frame to the 3' end of insert DNA 1 is inserted into the pCI-neo vector.
 なお,表7にはインサートDNA1及びインサートDNA2の各組み合わせについて,それぞれ得られる遺伝子がコードする目的の融合蛋白質を示す。本実施例における融合蛋白質の名称とその構造は下記(1)~(8)に示す通りである:
(1)hBDNF-HSA:野生型hBDNF(配列番号60)のC末端に,アミノ酸配列Gly-Serで示されるリンカーを介して野生型HSA(配列番号3)が結合し,そのさらにC末端にHis6が結合したものである,配列番号148で示されるアミノ酸配列を有する融合蛋白質;
(2)hNGF-HSA:野生型hNGF(配列番号62)のC末端に,アミノ酸配列Gly-Serで示されるリンカーを介して野生型HSA(配列番号3)が結合し,そのさらにC末端にHis6が結合したものである,配列番号149で示されるアミノ酸配列を有する融合蛋白質;
(3)hNT-4-HSA:野生型hNT-4(配列番号66)のC末端に,アミノ酸配列Gly-Serで示されるリンカーを介して野生型HSA(配列番号3)が結合し,そのさらにC末端にHis6が結合したものである,配列番号150で示されるアミノ酸配列を有する融合蛋白質;
(4)hNT-3-HSA:野生型hNT-3(配列番号64)のC末端に,アミノ酸配列Gly-Serで示されるリンカーを介して野生型HSA(配列番号3)が結合し,そのさらにC末端にHis6が結合したものである,配列番号151で示されるアミノ酸配列を有する融合蛋白質;
(5)HSA-hBDNF:野生型HSA(配列番号3)のC末端に,アミノ酸配列Gly-Serで示されるリンカーを介して野生型hBDNFのプロ体(配列番号84)が結合し,そのさらにC末端にHis6が結合したものである,配列番号152で示されるアミノ酸配列を有する融合蛋白質;
(6)HSA-hNGF:野生型HSA(配列番号3)のC末端に,アミノ酸配列Gly-Serで示されるリンカーを介して野生型hNGFのプロ体(配列番号87)が結合し,そのさらにC末端にHis6が結合したものである,配列番号153で示されるアミノ酸配列を有する融合蛋白質;
(7)HSA-hNT-4:野生型HSA(配列番号3)のC末端に,アミノ酸配列Gly-Serで示されるリンカーを介して野生型hNT-4のプロ体(配列番号93)が結合し,そのさらにC末端にHis6が結合したものである,配列番号154で示されるアミノ酸配列を有する融合蛋白質;及び
(8)HSA-hNT-3:野生型HSA(配列番号3)のC末端に,アミノ酸配列Gly-Serで示されるリンカーを介して野生型hNT-3のプロ体(配列番号90)が結合し,そのさらにC末端にHis6が結合したものである,配列番号155で示されるアミノ酸配列を有する融合蛋白質。
 ここで,HSA-hBDNF,HSA-hNGF,HSA-hNT-4及びHSA-hNT-3については,HSAのC末端側に融合させるヒト神経栄養因子(hBDNF,hNGF,hNT-4又はhNT-3)として全てプロ体のものを用いているため,これらを組み換え蛋白質として発現させた場合,融合蛋白質の一部又は全ての分子が切断され最終的に単体の野生型ヒト神経栄養因子(hBDNF,hNGF,hNT-4又はhNT-3)として得られる可能性があるが,実施例30及び実施例31で得られるSDS-PAGE及びウエスタンブロッティングの結果を観察する場合を除き,全て融合蛋白質として発現しているものとして取り扱う。
Table 7 shows the target fusion proteins encoded by the genes obtained for each combination of insert DNA 1 and insert DNA 2. The names and structures of the fusion proteins in this example are as shown below in (1) to (8):
(1) hBDNF-HSA: A fusion protein having the amino acid sequence shown in SEQ ID NO: 148, in which wild-type HSA (SEQ ID NO: 3) is linked to the C-terminus of wild-type hBDNF (SEQ ID NO: 60) via a linker shown in the amino acid sequence Gly-Ser, and His6 is further linked to the C-terminus of the linker;
(2) hNGF-HSA: a fusion protein having the amino acid sequence shown in SEQ ID NO: 149, in which wild-type HSA (SEQ ID NO: 3) is linked to the C-terminus of wild-type hNGF (SEQ ID NO: 62) via a linker shown in the amino acid sequence Gly-Ser, and His6 is further linked to the C-terminus of the linker;
(3) hNT-4-HSA: a fusion protein having the amino acid sequence shown in SEQ ID NO: 150, in which wild-type HSA (SEQ ID NO: 3) is linked to the C-terminus of wild-type hNT-4 (SEQ ID NO: 66) via a linker shown in the amino acid sequence Gly-Ser, and His6 is further linked to the C-terminus of the linker;
(4) hNT-3-HSA: a fusion protein having the amino acid sequence shown in SEQ ID NO: 151, in which wild-type HSA (SEQ ID NO: 3) is linked to the C-terminus of wild-type hNT-3 (SEQ ID NO: 64) via a linker shown in the amino acid sequence Gly-Ser, and His6 is further linked to the C-terminus of the linker;
(5) HSA-hBDNF: a fusion protein having the amino acid sequence shown in SEQ ID NO: 152, in which the pro-form of wild-type hBDNF (SEQ ID NO: 84) is bound to the C-terminus of wild-type HSA (SEQ ID NO: 3) via a linker shown in the amino acid sequence Gly-Ser, and His6 is further bound to the C-terminus of the linker;
(6) HSA-hNGF: a fusion protein having the amino acid sequence shown in SEQ ID NO: 153, in which the pro-form of wild-type hNGF (SEQ ID NO: 87) is bound to the C-terminus of wild-type HSA (SEQ ID NO: 3) via a linker shown in the amino acid sequence Gly-Ser, and His6 is further bound to the C-terminus of the linker;
(7) HSA-hNT-4: a fusion protein having the amino acid sequence shown in SEQ ID NO: 154, in which the pro-form of wild-type hNT-4 (SEQ ID NO: 93) is bound to the C-terminus of wild-type HSA (SEQ ID NO: 3) via a linker shown in the amino acid sequence Gly-Ser, and His6 is further bound to the C-terminus of the wild-type hNT-4; and (8) HSA-hNT-3: a fusion protein having the amino acid sequence shown in SEQ ID NO: 155, in which the pro-form of wild-type hNT-3 (SEQ ID NO: 90) is bound to the C-terminus of wild-type HSA (SEQ ID NO: 3) via a linker shown in the amino acid sequence Gly-Ser, and His6 is further bound to the C-terminus of the wild-type hNT-3.
Here, for HSA-hBDNF, HSA-hNGF, HSA-hNT-4 and HSA-hNT-3, the human neurotrophic factors (hBDNF, hNGF, hNT-4 or hNT-3) used to be fused to the C-terminus of HSA are all in the pro-form. Therefore, when these are expressed as recombinant proteins, some or all of the molecules of the fusion protein may be cleaved, and ultimately, a single wild-type human neurotrophic factor (hBDNF, hNGF, hNT-4 or hNT-3) may be obtained. However, except for the observation of the results of SDS-PAGE and Western blotting obtained in Examples 30 and 31, all are treated as being expressed as fusion proteins.
Figure JPOXMLDOC01-appb-T000007
Figure JPOXMLDOC01-appb-T000007
 次いで,各ライゲーション反応液を用いて大腸菌(ECOSTM X Competent E. coli DH5α,ニッポンジーン社)を上記(1)~(8)の融合蛋白質をコードする発現ベクターを用いてそれぞれ形質転換した。得られた形質転換体から,実施例1に記載と同様の手法により,目的遺伝子が組み込まれている各プラスミドをそれぞれ精製した。 Then, each ligation reaction solution was used to transform Escherichia coli (ECOSTM X Competent E. coli DH5α, Nippon Gene Co., Ltd.) with the expression vectors encoding the fusion proteins (1) to (8) above. From the resulting transformants, each plasmid incorporating the target gene was purified using the same method as described in Example 1.
〔実施例29〕野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質の一過性発現
 ExpiCHO Expression System(Thermo Fisher Scientific社)のHigh Titerプロトコールに従い,実施例27及び実施例28で得られた野生型ヒト神経栄養因子又はHSAとヒト神経栄養因子との融合蛋白質をそれぞれコードする遺伝子を組み込んだプラスミドを用いて,ExpiCHOTM細胞を形質転換させた。形質転換後,細胞を8日間培養し,野生型ヒト神経栄養因子又はHSAとヒト神経栄養因子との融合蛋白質をそれぞれ培養上清中に発現させた。培養後,培養液を遠心分離して培養上清を回収した。
[Example 29] Transient expression of wild-type human neurotrophic factor and fusion protein of HSA and human neurotrophic factor According to the High Titer protocol of ExpiCHO Expression System (Thermo Fisher Scientific), ExpiCHO TM cells were transformed with the plasmids obtained in Examples 27 and 28 incorporating genes encoding wild-type human neurotrophic factor or fusion protein of HSA and human neurotrophic factor, respectively. After transformation, the cells were cultured for 8 days to express wild-type human neurotrophic factor or fusion protein of HSA and human neurotrophic factor in the culture supernatant, respectively. After culturing, the culture medium was centrifuged to collect the culture supernatant.
〔実施例30〕一過性発現による野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質の発現量確認(SDS-PAGE)
 実施例29で得られた培養上清10 μLを,8 μLの2×Sample Buffer(バイオラッド社)および2 μLの2-メルカプトエタノール(バイオラッド社)と混合し,100℃で3分間加熱することにより還元条件下で熱変性させた。熱変性後の試料を,0.1% SDSを含む50 mM トリス緩衝液/380 mM グリシン緩衝液(pH8.3)内に設置した10-20%ポリアクリルアミドゲル(富士フィルム和光純薬社)のウェルに5 μLずつアプライし,25 mA定電流にて電気泳動した。電気泳動後のゲルをOriole Fluorescent Gel Stain(バイオラッド社)に浸し,室温で90分間振とうした。ゲルを純粋で洗浄した後,ルミノイメージアナライザー(Amersham Imager 600RGB,Cytiva社)で蛋白質のバンドを検出した。
[Example 30] Confirmation of expression levels of wild-type human neurotrophic factor and fusion protein of HSA and human neurotrophic factor by transient expression (SDS-PAGE)
10 μL of the culture supernatant obtained in Example 29 was mixed with 8 μL of 2×Sample Buffer (Bio-Rad) and 2 μL of 2-mercaptoethanol (Bio-Rad), and heat-denatured under reducing conditions by heating at 100°C for 3 minutes. 5 μL of the heat-denatured sample was applied to wells of a 10-20% polyacrylamide gel (Fujifilm Wako Pure Chemical Industries, Ltd.) placed in 50 mM Tris buffer/380 mM glycine buffer (pH 8.3) containing 0.1% SDS, and electrophoresis was performed at a constant current of 25 mA. The gel after electrophoresis was immersed in Oriole Fluorescent Gel Stain (Bio-Rad) and shaken at room temperature for 90 minutes. After washing the gel with pure water, protein bands were detected with a luminometer image analyzer (Amersham Imager 600RGB, Cytiva).
〔実施例31〕一過性発現による野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質の発現量確認(ウエスタンブロッティング)
 実施例30に記載した方法と同様に電気泳動を行い,ニトロセルロース膜と電気泳動後のゲルを,20%メタノールを含む25 mM トリス緩衝液/192 mM グリシン緩衝液に浸したブロッティングペーパーで挟み,ブロッティング装置(Trans-Blot Turbo Transfer System,バイオラッド社)で1.0 A,25 Vで10分間通電することで蛋白質をニトロセルロース膜に転写した。転写後のニトロセルロース膜を,5%スキムミルクを含むTBSTに浸して1時間振とう後,0.4 μg/mLに希釈したMouse anti-His tag mAb(医学生物学研究所)溶液に浸して1時間振とうした。TBSTで膜を洗浄後,0.2 μg/mLに希釈したAnti-mouse IgG (H+L), HRP Conjugate(プロメガ社)溶液に浸して60分振とうし,再度TBSTで洗浄した。膜の転写面にHRP検出試薬(バイオラッド社)を滴下して5分間反応させ,ルミノイメージアナライザーで検出した。
[Example 31] Confirmation of expression levels of wild-type human neurotrophic factor and fusion protein of HSA and human neurotrophic factor by transient expression (Western blotting)
Electrophoresis was performed in the same manner as described in Example 30, and the nitrocellulose membrane and the gel after electrophoresis were sandwiched between blotting papers soaked in 25 mM Tris buffer/192 mM glycine buffer containing 20% methanol, and the proteins were transferred to the nitrocellulose membrane by applying electricity at 1.0 A and 25 V for 10 minutes using a blotting device (Trans-Blot Turbo Transfer System, Bio-Rad). The nitrocellulose membrane after transfer was immersed in TBST containing 5% skim milk and shaken for 1 hour, and then immersed in a solution of Mouse anti-His tag mAb (Medical and Biological Laboratories) diluted to 0.4 μg/mL and shaken for 1 hour. After washing the membrane with TBST, it was immersed in a solution of Anti-mouse IgG (H+L), HRP Conjugate (Promega) diluted to 0.2 μg/mL, shaken for 60 minutes, and washed again with TBST. HRP detection reagent (Bio-Rad) was dropped on the transfer surface of the membrane, reacted for 5 minutes, and detected with a lumino image analyzer.
〔実施例32〕一過性発現による野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質の発現量確認(ELISA)
 一過性発現による野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質の発現量を比較するため,Human Multi-Neurotrophin Rapid Screening ELISA Kit(Biosensis社)を用いて,ELISA測定を行った。実施例29で得られた培養上清を,キット付属のAssay diluent Aで適宜希釈したものを試料溶液として,培養上清中の野生型ヒト神経栄養因子又はHSAとヒト神経栄養因子との融合蛋白質の濃度を算出した。単体の野生型ヒト神経栄養因子又はHSAと融合させたヒト神経栄養因子の種類ごとに,下記詳述する。
[Example 32] Confirmation of expression levels of wild-type human neurotrophic factor and fusion protein of HSA and human neurotrophic factor by transient expression (ELISA)
In order to compare the expression levels of wild-type human neurotrophic factor and the fusion protein of HSA and human neurotrophic factor by transient expression, ELISA measurement was performed using the Human Multi-Neurotrophin Rapid Screening ELISA Kit (Biosensis). The culture supernatant obtained in Example 29 was appropriately diluted with Assay diluent A included in the kit to prepare a sample solution, and the concentration of wild-type human neurotrophic factor or the fusion protein of HSA and human neurotrophic factor in the culture supernatant was calculated. Details are given below for each type of wild-type human neurotrophic factor alone or human neurotrophic factor fused with HSA.
〔野生型hBDNF,及びHSAとhBDNFとの融合蛋白質の測定〕
 試料溶液として,実施例29で得られた野生型hBDNF,hBDNF-HSA,又はHSA-hBDNFを発現させたExpiCHO培養上清をAssay diluent Aで10万倍希釈したものを調製した。標準溶液として,キット付属の1000 pg/mL BDNF溶液をAssay diluent Aで段階希釈し,500,250,125,63,31,16,及び8 pg/mLとしたものをそれぞれ調製した。抗BDNF抗体が固相化された96ウェルプレートの各ウェルに試料溶液または標準溶液を100 μLずつ添加し,プレートシェーカーで室温にて90分間振とうした。次いで,200 μLのWash bufferで各ウェルを5回洗浄後,BDNF detection antibody(ビオチン標識抗BDNF抗体)を100 μLずつ各ウェルに添加し,プレートシェーカーで室温にて30分間振とうした。次いで,200 μLのWash bufferで各ウェルを5回洗浄後,Streptavidin-HRPを100 μLずつ各ウェルに添加し,プレートシェーカーで室温にて30分間振とうした。200 μLのWash bufferで各ウェルを5回洗浄後,TMB substrateを100 μLずつ各ウェルに添加し,室温で静置して発色反応させた。充分な発色が得られた後,TMB stop solutionを100 μLずつ各ウェルに添加して反応を停止させた。マイクロプレートリーダー(Spectramax 190 EXT,Molecular Device社)で波長450 nmにおける吸光度を測定し,解析ソフトウェア(SoftMax Pro,Molecular Device社)を用いて検量線を作成し,これに各試料溶液の測定値を内挿して,各培養上清中の濃度(物質量に換算)を算出した。
[Measurement of wild-type hBDNF and fusion protein of HSA and hBDNF]
As sample solutions, the culture supernatant of ExpiCHO expressing wild-type hBDNF, hBDNF-HSA, or HSA-hBDNF obtained in Example 29 was diluted 100,000 times with Assay diluent A to prepare a standard solution. The 1000 pg/mL BDNF solution included in the kit was serially diluted with Assay diluent A to prepare 500, 250, 125, 63, 31, 16, and 8 pg/mL solutions, respectively. 100 μL of the sample solution or standard solution was added to each well of a 96-well plate on which anti-BDNF antibody was immobilized, and the plate was shaken at room temperature for 90 minutes on a plate shaker. Next, each well was washed five times with 200 μL of Wash buffer, and then 100 μL of BDNF detection antibody (biotin-labeled anti-BDNF antibody) was added to each well, and the plate was shaken at room temperature for 30 minutes on a plate shaker. Next, each well was washed five times with 200 μL of wash buffer, and 100 μL of streptavidin-HRP was added to each well and shaken on a plate shaker at room temperature for 30 minutes. After washing each well five times with 200 μL of wash buffer, 100 μL of TMB substrate was added to each well and left to stand at room temperature for color reaction. After sufficient color development was obtained, 100 μL of TMB stop solution was added to each well to stop the reaction. The absorbance at a wavelength of 450 nm was measured using a microplate reader (Spectramax 190 EXT, Molecular Device), and a calibration curve was created using analysis software (SoftMax Pro, Molecular Device), and the measured values of each sample solution were inserted into this to calculate the concentration (converted to the amount of substance) in each culture supernatant.
〔野生型hNGF,及びHSAとhNGFとの融合蛋白質の測定〕
 試料溶液として,実施例29で得られた野生型hNGF,hNGF-HSA,又はHSA-hNGFを発現させたExpiCHO培養上清をAssay diluent Aで100000倍希釈したものを調製した。標準溶液として,キット付属の500 pg/mL NGF溶液をAssay diluent Aで段階希釈し,250,125,63,31,16,8,及び4 pg/mLとしたものをそれぞれ調製した。抗NGF抗体が固相化された96ウェルプレートの各ウェルに試料溶液または標準溶液を100 μLずつ添加し,プレートシェーカーで室温にて90分間振とうした。次いで,200 μLのWash bufferで各ウェルを5回洗浄後,NGF detection antibody(ビオチン標識抗NGF抗体)を100 μLずつ各ウェルに添加し,プレートシェーカーで室温にて30分間振とうした。次いで,200 μLのWash bufferで各ウェルを5回洗浄後,Streptavidin-HRPを100 μLずつ各ウェルに添加し,プレートシェーカーで室温にて30分間振とうした。200 μLのWash bufferで各ウェルを5回洗浄後,TMB substrateを100 μLずつ各ウェルに添加し,室温で静置して発色反応させた。充分な発色が得られた後,TMB stop solutionを100 μLずつ各ウェルに添加して反応を停止させた。マイクロプレートリーダー(Spectramax 190 EXT,Molecular Device社)で波長450 nmにおける吸光度を測定し,解析ソフトウェア(SoftMax Pro,Molecular Device社)を用いて検量線を作成し,これに各試料溶液の測定値を内挿して,各培養上清中の濃度(物質量に換算)を算出した。
[Measurement of wild-type hNGF and fusion protein of HSA and hNGF]
As sample solutions, the culture supernatant of ExpiCHO expressing wild-type hNGF, hNGF-HSA, or HSA-hNGF obtained in Example 29 was diluted 100,000 times with Assay diluent A to prepare the standard solutions. The 500 pg/mL NGF solution included in the kit was serially diluted with Assay diluent A to prepare 250, 125, 63, 31, 16, 8, and 4 pg/mL solutions, respectively. 100 μL of the sample solution or standard solution was added to each well of a 96-well plate on which anti-NGF antibody was immobilized, and the plate was shaken at room temperature for 90 minutes on a plate shaker. Next, each well was washed five times with 200 μL of Wash buffer, and then 100 μL of NGF detection antibody (biotin-labeled anti-NGF antibody) was added to each well, and the plate was shaken at room temperature for 30 minutes on a plate shaker. Next, each well was washed five times with 200 μL of wash buffer, and 100 μL of streptavidin-HRP was added to each well and shaken on a plate shaker at room temperature for 30 minutes. After washing each well five times with 200 μL of wash buffer, 100 μL of TMB substrate was added to each well and left to stand at room temperature for color reaction. After sufficient color development was obtained, 100 μL of TMB stop solution was added to each well to stop the reaction. The absorbance at a wavelength of 450 nm was measured using a microplate reader (Spectramax 190 EXT, Molecular Device), and a calibration curve was created using analysis software (SoftMax Pro, Molecular Device), and the measured values of each sample solution were inserted into this to calculate the concentration (converted to the amount of substance) in each culture supernatant.
〔野生型hNT-4,及びHSAとhNT-4との融合蛋白質の測定〕
 試料溶液として,実施例29で得られた野生型hNT-4,hNT-4-HSA,及びHSA-hNT-4を発現させたExpiCHO培養上清をAssay diluent Aで100000倍希釈したものを調製した。標準溶液として,キット付属の2000 pg/mL NT-4溶液をAssay diluent Aで段階希釈し,1000,500,250,125,63,31,及び16 pg/mLとしたものをそれぞれ調製した。抗NT-4抗体が固相化された96ウェルプレートの各ウェルに試料溶液または標準溶液を100 μLずつ添加し,プレートシェーカーで室温にて90分間振とうした。次いで,200 μLのWash bufferで各ウェルを5回洗浄後,NT-4 detection antibody(ビオチン標識抗NT-4抗体)を100 μLずつ各ウェルに添加し,プレートシェーカーで室温にて30分間振とうした。次いで,200 μLのWash bufferで各ウェルを5回洗浄後,Streptavidin-HRPを100 μLずつ各ウェルに添加し,プレートシェーカーで室温にて30分間振とうした。200 μLのWash bufferで各ウェルを5回洗浄後,TMB substrateを100 μLずつ各ウェルに添加し,室温で静置して発色反応させた。充分な発色が得られた後,TMB stop solutionを100 μLずつ各ウェルに添加して反応を停止させた。マイクロプレートリーダー(Spectramax 190 EXT,Molecular Device社)で波長450 nmにおける吸光度を測定し,解析ソフトウェア(SoftMax Pro,Molecular Device社)を用いて検量線を作成し,これに各試料溶液の測定値を内挿して,各培養上清中の濃度(モノマーを1分子として物質量に換算)を算出した。
[Measurement of wild-type hNT-4 and fusion protein of HSA and hNT-4]
As sample solutions, the culture supernatants of ExpiCHO expressing wild-type hNT-4, hNT-4-HSA, and HSA-hNT-4 obtained in Example 29 were diluted 100,000 times with Assay diluent A to prepare them. As standard solutions, the 2000 pg/mL NT-4 solution included in the kit was serially diluted with Assay diluent A to prepare 1000, 500, 250, 125, 63, 31, and 16 pg/mL solutions, respectively. 100 μL of the sample solution or standard solution was added to each well of a 96-well plate on which anti-NT-4 antibody was immobilized, and the plate was shaken at room temperature for 90 minutes on a plate shaker. Next, each well was washed five times with 200 μL of Wash buffer, and then 100 μL of NT-4 detection antibody (biotin-labeled anti-NT-4 antibody) was added to each well, and the plate was shaken at room temperature for 30 minutes on a plate shaker. Next, each well was washed five times with 200 μL of Wash buffer, and 100 μL of Streptavidin-HRP was added to each well and shaken on a plate shaker at room temperature for 30 minutes. After washing each well five times with 200 μL of Wash buffer, 100 μL of TMB substrate was added to each well and left to stand at room temperature for color reaction. After sufficient color development was obtained, 100 μL of TMB stop solution was added to each well to stop the reaction. The absorbance at a wavelength of 450 nm was measured using a microplate reader (Spectramax 190 EXT, Molecular Device), and a calibration curve was created using analysis software (SoftMax Pro, Molecular Device), and the measured values of each sample solution were inserted into this to calculate the concentration in each culture supernatant (converted to the amount of substance with one monomer as one molecule).
〔野生型hNT-3,及びHSAとhNT-3との融合蛋白質の測定〕
 試料溶液として,実施例29で得られた野生型hNT-3,hNT-3-HSA,及びHSA-hNT-3を発現させたExpiCHO培養上清をAssay diluent Aで100000倍希釈したものを調製した。標準溶液として,キット付属の2000 pg/mL NT-3溶液をAssay diluent Aで段階希釈し,1000,500,250,125,63,31,及び16 pg/mLとしたものをそれぞれ調製した。抗NT-3抗体が固相化された96ウェルプレートの各ウェルに試料溶液または標準溶液を100 μLずつ添加し,プレートシェーカーで室温にて90分間振とうした。次いで,200 μLのWash bufferで各ウェルを5回洗浄後,NT-3 detection antibody(ビオチン標識抗NT-3抗体)を100 μLずつ各ウェルに添加し,プレートシェーカーで室温にて30分間振とうした。次いで,200 μLのWash bufferで各ウェルを5回洗浄後,Streptavidin-HRPを100 μLずつ各ウェルに添加し,プレートシェーカーで室温にて30分間振とうした。200 μLのWash bufferで各ウェルを5回洗浄後,TMB substrateを100 μLずつ各ウェルに添加し,室温で静置して発色反応させた。充分な発色が得られた後,TMB stop solutionを100 μLずつ各ウェルに添加して反応を停止させた。マイクロプレートリーダー(Spectramax 190 EXT,Molecular Device社)で波長450 nmにおける吸光度を測定し,解析ソフトウェア(SoftMax Pro,Molecular Device社)を用いて検量線を作成し,これに各試料溶液の測定値を内挿して,各培養上清中の濃度(モノマーを1分子として物質量に換算)を算出した。
[Measurement of wild-type hNT-3 and fusion protein of HSA and hNT-3]
As sample solutions, the culture supernatants of ExpiCHO expressing wild-type hNT-3, hNT-3-HSA, and HSA-hNT-3 obtained in Example 29 were diluted 100,000 times with Assay diluent A to prepare them. As standard solutions, the 2000 pg/mL NT-3 solution included in the kit was serially diluted with Assay diluent A to prepare 1000, 500, 250, 125, 63, 31, and 16 pg/mL solutions, respectively. 100 μL of the sample solution or standard solution was added to each well of a 96-well plate on which anti-NT-3 antibody was immobilized, and the plate was shaken at room temperature for 90 minutes on a plate shaker. Next, each well was washed five times with 200 μL of Wash buffer, and then 100 μL of NT-3 detection antibody (biotin-labeled anti-NT-3 antibody) was added to each well, and the plate was shaken at room temperature for 30 minutes on a plate shaker. Next, each well was washed five times with 200 μL of Wash buffer, and 100 μL of Streptavidin-HRP was added to each well and shaken on a plate shaker at room temperature for 30 minutes. After washing each well five times with 200 μL of Wash buffer, 100 μL of TMB substrate was added to each well and left to stand at room temperature for color reaction. After sufficient color development was obtained, 100 μL of TMB stop solution was added to each well to stop the reaction. The absorbance at a wavelength of 450 nm was measured using a microplate reader (Spectramax 190 EXT, Molecular Device), and a calibration curve was created using analysis software (SoftMax Pro, Molecular Device), and the measured values of each sample solution were inserted into this to calculate the concentration in each culture supernatant (converted to the amount of substance with one monomer as one molecule).
〔実施例33〕一過性発現による野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質の発現量の確認(結果)
 図28に,実施例32で測定した一過性発現による野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質のELISA濃度測定の結果を示す。また,ELISA濃度測定の結果に基づく野生型ヒト神経栄養因子及びHSAとヒト神経栄養因子との融合蛋白質の相対量を表8に示す。
[Example 33] Confirmation of the expression levels of wild-type human neurotrophic factor and fusion protein of HSA and human neurotrophic factor by transient expression (Results)
28 shows the results of ELISA concentration measurement of the wild-type human neurotrophic factor and the fusion protein of HSA and human neurotrophic factor by transient expression measured in Example 32. In addition, the relative amounts of the wild-type human neurotrophic factor and the fusion protein of HSA and human neurotrophic factor based on the results of ELISA concentration measurement are shown in Table 8.
Figure JPOXMLDOC01-appb-T000008
Figure JPOXMLDOC01-appb-T000008
 上記の結果から,培養開始後8日後の発現量(濃度)をみると,hBDNF-HSA及びHSA-hBDNFの発現量は,野生型hBDNFのそれと比較して,それぞれ29倍及び3.0倍の値を示し,野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なhBDNFを,hBDNFとHSAとの融合蛋白質の形態で発現させることにより,より多くのhBDNFを組換え蛋白質として効率よく発現させることができることがわかった。 The above results show that, when the expression levels (concentrations) were examined 8 days after the start of culture, the expression levels of hBDNF-HSA and HSA-hBDNF were 29 and 3.0 times higher, respectively, than that of wild-type hBDNF. This shows that hBDNF, which is difficult to mass-produce due to its low expression level when expressed as a recombinant in its wild form, can be efficiently expressed in greater amounts as a recombinant protein by expressing it in the form of a fusion protein of hBDNF and HSA.
 また,hNGF-HSA及びHSA-hNGFの発現量は,野生型hNGFのそれと比較して,それぞれ1.6倍及び1.3倍の値を示し,野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なhNGFを,hNGFとHSAとの融合蛋白質の形態で発現させることにより,より多くのhNGFを組換え蛋白質として効率よく発現させることができることがわかった。 In addition, the expression levels of hNGF-HSA and HSA-hNGF were 1.6 and 1.3 times higher than that of wild-type hNGF, respectively. This indicates that hNGF is difficult to mass-produce because of its low expression level when expressed as a recombinant form in its wild-type form, but by expressing it in the form of a fusion protein of hNGF and HSA, it is possible to efficiently express more hNGF as a recombinant protein.
 また,hNT-4-HSA及びHSA-hNT-4の発現量は,野生型hNT-4のそれと比較して,それぞれ4.1倍及び1.8倍の値を示し,野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なhNT-4を,hNT-4とHSAとの融合蛋白質の形態で発現させることにより,より多くのhNT-4を組換え蛋白質として効率よく発現させることができることがわかった。 In addition, the expression levels of hNT-4-HSA and HSA-hNT-4 were 4.1 and 1.8 times higher than that of wild-type hNT-4, respectively. This indicates that hNT-4, which is difficult to mass-produce due to its low expression level when expressed as a recombinant in its wild form, can be efficiently expressed in larger amounts as a recombinant protein by expressing it in the form of a fusion protein of hNT-4 and HSA.
 また,hNT-3-HSAの発現量は,野生型hNT-3のそれと比較して,12倍の値を示し,野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なhNT-3を,hNT-3とHSAとの融合蛋白質の形態で発現させることにより,より多くのhNT-3を組換え蛋白質として効率よく発現させることができることがわかった。 In addition, the expression level of hNT-3-HSA was 12 times higher than that of wild-type hNT-3. Wild-type hNT-3 is difficult to mass-produce due to its low expression level when expressed as a recombinant form, but by expressing it in the form of a fusion protein of hNT-3 and HSA, it was found that more hNT-3 could be efficiently expressed as a recombinant protein.
 次に,実施例30で得たSDS-PAGEの泳動画像を図29に示す。SDS-PAGEの結果は,バンド強度の比較からhBDNF-HSA及びHSA-hBDNFは野生型hBDNFより発現量が高く,hNGF-HSA及びHSA-hNGFは野生型hNGFより発現量が高く,hNT-4-HSA及びHSA-hNT-4は野生型hNT-4より発現量が高く,hNT-3-HSA及びHSA-hNT-3は野生型hNT-3より発現量が高く,図28及び表8で示される発現量(濃度)と対応するものであった。野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なヒト神経栄養因子を,HSAとヒト神経栄養因子との融合蛋白質の形態で発現させることにより,より多くのヒト神経栄養因子を組換え蛋白質として効率よく発現させることができることがわかった。 Next, the electrophoretic image of SDS-PAGE obtained in Example 30 is shown in Figure 29. The results of SDS-PAGE showed that hBDNF-HSA and HSA-hBDNF were expressed at higher levels than wild-type hBDNF, hNGF-HSA and HSA-hNGF were expressed at higher levels than wild-type hNGF, hNT-4-HSA and HSA-hNT-4 were expressed at higher levels than wild-type hNT-4, and hNT-3-HSA and HSA-hNT-3 were expressed at higher levels than wild-type hNT-3, which correspond to the expression levels (concentrations) shown in Figure 28 and Table 8. Human neurotrophic factor, which is expressed in low levels when expressed as a recombinant in its wild form and is difficult to mass-produce, was found to be able to be efficiently expressed in greater amounts as a recombinant protein by expressing it in the form of a fusion protein of HSA and human neurotrophic factor.
 次に,実施例31で得たウエスタンブロッティングの泳動画像を図30に示す。ウエスタンブロッティングの結果は,バンド強度の比較からhBDNF-HSA及びHSA-hBDNFは野生型hBDNFより発現量が高く,hNGF-HSA及びHSA-hNGFは野生型hNGFより発現量が高く,hNT-4-HSA及びHSA-hNT-4は野生型hNT-4より発現量が高く,hNT-3-HSA及びHSA-hNT-3は野生型hNT-3より発現量が高く,図28及び表8で示される発現量(濃度)と対応するものであった。ただし,HSA-hBDNF,HSA-hNGF,HSA-hNT-4,及びHSA-hNT-3は,途中で切断され単体の成熟体ヒト神経栄養因子として検出されているバンドもあわせて観察した。野生型のままでは組換え体として発現させたときに発現量が少なく大量に製造することが困難なヒト神経栄養因子を,HSAとヒト神経栄養因子との融合蛋白質の形態で発現させることにより,より多くのヒト神経栄養因子を組換え蛋白質として効率よく発現させることができることがわかった。 Next, the electrophoretic images of the Western blotting obtained in Example 31 are shown in Figure 30. The results of the Western blotting showed that hBDNF-HSA and HSA-hBDNF were expressed at higher levels than wild-type hBDNF, hNGF-HSA and HSA-hNGF were expressed at higher levels than wild-type hNGF, hNT-4-HSA and HSA-hNT-4 were expressed at higher levels than wild-type hNT-4, and hNT-3-HSA and HSA-hNT-3 were expressed at higher levels than wild-type hNT-3, which corresponded to the expression levels (concentrations) shown in Figure 28 and Table 8. However, for HSA-hBDNF, HSA-hNGF, HSA-hNT-4, and HSA-hNT-3, bands that were cleaved midway and detected as single mature human neurotrophic factors were also observed. Human neurotrophic factor is difficult to mass-produce because the expression level is low when expressed as a recombinant wild type. However, by expressing it in the form of a fusion protein of HSA and human neurotrophic factor, it was found that a larger amount of human neurotrophic factor can be efficiently expressed as a recombinant protein.
 以上の結果は,ヒト神経栄養因子を組換え蛋白質として製造する場合に,ヒト神経栄養因子を,ヒト神経栄養因子とHSAとの融合蛋白質の形態で製造することにより,より多くの分子数のヒト神経栄養因子を得ることができることを示すものである。つまり,ヒト神経栄養因子をヒト神経栄養因子とHSAとの融合蛋白質の形態で発現させる組換えヒト神経栄養因子の製造方法は,大量の組換えヒト神経栄養因子を製造する方法として極めて有効な手段といえる。 The above results indicate that when producing human neurotrophic factor as a recombinant protein, a larger number of molecules of human neurotrophic factor can be obtained by producing the human neurotrophic factor in the form of a fusion protein of human neurotrophic factor and HSA. In other words, a method for producing recombinant human neurotrophic factor in which human neurotrophic factor is expressed in the form of a fusion protein of human neurotrophic factor and HSA can be said to be an extremely effective means for producing large quantities of recombinant human neurotrophic factor.
 本発明によれば,例えば,組換え蛋白質として製造することが困難な蛋白質を,SAとの融合蛋白質の形態で組換え蛋白質として発現させることにより,効率よく工業的に生産できる。 According to the present invention, for example, proteins that are difficult to produce as recombinant proteins can be efficiently and industrially produced by expressing them as recombinant proteins in the form of fusion proteins with SA.
配列番号1:野生型hGALCのアミノ酸配列の一例
配列番号2:野生型hGALCをコードする塩基配列の一例,合成配列
配列番号3:野生型HSAのアミノ酸配列の一例
配列番号4:野生型HSAをコードする塩基配列の一例,合成配列
配列番号5:HSA-hGALCのアミノ酸配列の一例
配列番号6:HSA-hGALCをコードする塩基配列の一例,合成配列
配列番号7:hGALC-HSAのアミノ酸配列の一例
配列番号8:hGALC-HSAをコードする塩基配列の一例,合成配列
配列番号9:リンカーのアミノ酸配列の例1
配列番号10:リンカーのアミノ酸配列の例2
配列番号11:リンカーのアミノ酸配列の例3
配列番号12:ヒト血清アルブミンRedhillのアミノ酸配列
配列番号13:変異型ヒト血清アルブミン(HSA-A320T)のアミノ酸配列
配列番号14:野生型mGALCのアミノ酸配列の一例
配列番号15:野生型mGALCをコードする塩基配列の一例,合成配列
配列番号16:野生型MSAのアミノ酸配列の一例
配列番号17:野生型MSAをコードする塩基配列の一例,合成配列
配列番号18:MSA-mGALCのアミノ酸配列の一例
配列番号19:MSA-mGALCをコードする塩基配列の一例,合成配列
配列番号20:mGALC-MSAのアミノ酸配列の一例
配列番号21:mGALC-MSAをコードする塩基配列の一例,合成配列
配列番号22:ヒトトランスフェリン受容体のアミノ酸配列
配列番号23:抗hTfR抗体の軽鎖のアミノ酸配列
配列番号24:野生型hIL-10のアミノ酸配列の一例
配列番号25:抗hTfR抗体のFab重鎖のアミノ酸配列
配列番号26:pCI Insert F3プライマー,合成配列
配列番号27:pCI Insert R3プライマー,合成配列
配列番号28:抗hTfR抗体Fab重鎖-HSA-hGALCのアミノ酸配列
配列番号29:抗hTfR抗体Fab重鎖-HSA-hGALCをコードする塩基配列,合成配列
配列番号30:HSA-hGALC-抗hTfR抗体Fab重鎖のアミノ酸配列
配列番号31:HSA-hGALC-抗hTfR抗体Fab重鎖をコードする塩基配列,合成配列
配列番号32:抗hTfR抗体Fab重鎖-hGALC-HSAのアミノ酸配列
配列番号33:抗hTfR抗体Fab重鎖-hGALC-HSAをコードする塩基配列,合成配列
配列番号34:hGALC-HSA-抗hTfR抗体Fab重鎖のアミノ酸配列
配列番号35:hGALC-HSA-抗hTfR抗体Fab重鎖をコードする塩基配列,合成配列
配列番号36:抗hTfR抗体の軽鎖をコードする塩基配列,合成配列
配列番号37:野生型hGBAのアミノ酸配列の一例
配列番号38:野生型hGBAをコードする塩基配列の一例
配列番号39:HSA-hGBAのアミノ酸配列の一例
配列番号40:HSA-hGBAをコードする塩基配列の一例,合成配列
配列番号41:hGBA-HSAのアミノ酸配列の一例
配列番号42:hGBA-HSAをコードする塩基配列の一例,合成配列
配列番号43:野生型mGBAのアミノ酸配列の一例
配列番号44:野生型mGBAをコードする塩基配列の一例
配列番号45:MSA-mGBAのアミノ酸配列の一例
配列番号46:MSA-mGBAをコードする塩基配列の一例,合成配列
配列番号47:mGBA-MSAのアミノ酸配列の一例
配列番号48:mGBA-MSAをコードする塩基配列の一例,合成配列
配列番号49:野生型hIL-10をコードする塩基配列の一例,合成配列
配列番号50:HSA-hIL-10のアミノ酸配列の一例
配列番号51:HSA-hIL-10をコードする塩基配列の一例,合成配列
配列番号52:hIL-10-HSAのアミノ酸配列の一例
配列番号53:hIL-10-HSAをコードする塩基配列の一例,合成配列
配列番号54:HSA-hIL-10のアミノ酸配列の一例2
配列番号55:hIL-10-HSAのアミノ酸配列の一例2
配列番号56:HSA-hIL-10のアミノ酸配列の一例3
配列番号57:hIL-10-HSAのアミノ酸配列の一例3
配列番号58:HSA-hIL-10のアミノ酸配列の一例4
配列番号59:hIL-10-HSAのアミノ酸配列の一例4
配列番号60:野生型hBDNFのアミノ酸配列の一例
配列番号61:野生型hBDNFをコードする塩基配列の一例,合成配列
配列番号62:野生型hNGFのアミノ酸配列の一例
配列番号63:野生型hNGFをコードする塩基配列の一例,合成配列
配列番号64:野生型hNT-3のアミノ酸配列の一例
配列番号65:野生型hNT-3をコードする塩基配列の一例,合成配列
配列番号66:野生型hNT-4のアミノ酸配列の一例
配列番号67:野生型hNT-4をコードする塩基配列の一例,合成配列
配列番号68:HSA-hBDNFのアミノ酸配列の一例
配列番号69:HSA-hBDNFをコードする塩基配列の一例,合成配列
配列番号70:HSA-hNGFのアミノ酸配列の一例
配列番号71:HSA-hNGFをコードする塩基配列の一例,合成配列
配列番号72:HSA-hNT-3のアミノ酸配列の一例
配列番号73:HSA-hNT-3をコードする塩基配列の一例,合成配列
配列番号74:HSA-hNT-4のアミノ酸配列の一例
配列番号75:HSA-hNT-4をコードする塩基配列の一例,合成配列
配列番号76:hBDNF-HSAのアミノ酸配列の一例
配列番号77:hBDNF-HSAをコードする塩基配列の一例,合成配列
配列番号78:hNGF-HSAのアミノ酸配列の一例
配列番号79:hNGF-HSAをコードする塩基配列の一例,合成配列
配列番号80:hNT-3-HSAのアミノ酸配列の一例
配列番号81:hNT-3-HSAをコードする塩基配列の一例,合成配列
配列番号82:hNT-4-HSAのアミノ酸配列の一例
配列番号83:hNT-4-HSAをコードする塩基配列の一例,合成配列
配列番号84:野生型hBDNF(プロ体)のアミノ酸配列の一例
配列番号85:HSA-hBDNFのアミノ酸配列の一例2
配列番号86:hBDNF-HSAのアミノ酸配列の一例2
配列番号87:野生型hNGF(プロ体)のアミノ酸配列の一例
配列番号88:HSA-hNGFのアミノ酸配列の一例2
配列番号89:hNGF-HSAのアミノ酸配列の一例2
配列番号90:野生型hNT-3(プロ体)のアミノ酸配列の一例
配列番号91:HSA-hNT-3のアミノ酸配列の一例2
配列番号92:hNT-3-HSAのアミノ酸配列の一例2
配列番号93:野生型hNT-4(プロ体)のアミノ酸配列の一例
配列番号94:HSA-hNT-4のアミノ酸配列の一例2
配列番号95:hNT-4-HSAのアミノ酸配列の一例2
配列番号96:抗hTfR抗体の軽鎖CDR1のアミノ酸配列
配列番号97:抗hTfR抗体の軽鎖CDR2のアミノ酸配列
配列番号98:抗hTfR抗体の軽鎖CDR3のアミノ酸配列
配列番号99:抗hTfR抗体の重鎖CDR1のアミノ酸配列1
配列番号100:抗hTfR抗体の重鎖CDR2のアミノ酸配列1
配列番号101:抗hTfR抗体の重鎖CDR3のアミノ酸配列1
配列番号102:抗hTfR抗体の重鎖CDR1のアミノ酸配列2
配列番号103:抗hTfR抗体の重鎖CDR2のアミノ酸配列2
配列番号104:抗hTfR抗体の重鎖CDR3のアミノ酸配列2
配列番号105:hTfR親和性ペプチドのCDR1のアミノ酸配列1
配列番号106:hTfR親和性ペプチドのCDR1のアミノ酸配列2
配列番号107:hTfR親和性ペプチドのCDR2のアミノ酸配列1
配列番号108:hTfR親和性ペプチドのCDR2のアミノ酸配列2
配列番号109:hTfR親和性ペプチドのCDR3のアミノ酸配列1
配列番号110:hTfR親和性ペプチドのCDR3のアミノ酸配列2
配列番号111:hTfR親和性ペプチドのCDR2のアミノ酸配列3
配列番号112:hTfR親和性ペプチドのCDR2のアミノ酸配列4
配列番号113:血清型2のAAVの第一の逆方向末端反復(第一のITR)の塩基配列,野生型
配列番号114:血清型2のAAVの第一の逆方向末端反復(第二のITR)の塩基配列,野生型
配列番号115:第一の逆方向末端反復(第一のITR)の塩基配列の好適な一例,合成配列
配列番号116:第二の逆方向末端反復(第二のITR)の塩基配列の好適な一例,合成配列
配列番号117:第一の長い末端反復(第一のLTR)の塩基配列の好適な一例,合成配列
配列番号118:第二の長い末端反復(第二のLTR)の塩基配列の好適な一例,合成配列
配列番号119:センダイウイルスゲノムのリーダーの塩基配列
配列番号120:センダイウイルスゲノムのトレイラーの塩基配列
配列番号121:マウスαフェトプロテインエンハンサー/マウスアルブミンプロモーターの塩基配列,合成配列
配列番号122:ニワトリβアクチン/MVMキメライントロンの塩基配列,合成配列
配列番号123:AAV2のRep領域を含む塩基配列,合成配列
配列番号124:AAV8のCap領域の塩基配列
配列番号125:変異型マウス血清アルブミン(MSA-A320T)のアミノ酸配列
配列番号126:野生型mIL-10のアミノ酸配列の一例
配列番号127:野生型mIL-10をコードする塩基配列の一例,合成配列
配列番号128:mIL-10-MSAのアミノ酸配列の一例
配列番号129:mIL-10-MSAをコードする塩基配列の一例,合成配列
配列番号130:MSA-mIL-10のアミノ酸配列の一例
配列番号131:MSA-mIL-10をコードする塩基配列の一例,合成配列
配列番号132:His6を付加した野生型hBDNFのアミノ酸配列
配列番号133:His6を付加した野生型hBDNFをコードする塩基配列の一例,合成配列
配列番号134:His6を付加した野生型hNGFのアミノ酸配列
配列番号135:His6を付加した野生型hNGFをコードする塩基配列の一例,合成配列
配列番号136:His6を付加した野生型hNT-4のアミノ酸配列
配列番号137:His6を付加した野生型hNT-4をコードする塩基配列の一例,合成配列
配列番号138:His6を付加した野生型hNT-3のアミノ酸配列
配列番号139:His6を付加した野生型hNT-3をコードする塩基配列の一例,合成配列
配列番号140:His6を付加した野生型HSAのアミノ酸配列
配列番号141:His6を付加した野生型HSAをコードする塩基配列の一例,合成配列
配列番号142:フォワードプライマー1,合成配列
配列番号143:フォワードプライマー2,合成配列
配列番号144:フォワードプライマー3,合成配列
配列番号145:フォワードプライマー4,合成配列
配列番号146:フォワードプライマー5,合成配列
配列番号147:リバースプライマー,合成配列
配列番号148:hBDNF-HSAのアミノ酸配列
配列番号149:hNGF-HSAのアミノ酸配列
配列番号150:hNT-4-HSAのアミノ酸配列
配列番号151:hNT-3-HSAのアミノ酸配列
配列番号152:HSA-hBDNFのアミノ酸配列
配列番号153:HSA-hNGFのアミノ酸配列
配列番号154:HSA-hNT-4のアミノ酸配列
配列番号155:HSA-hNT-3のアミノ酸配列
SEQ ID NO: 1: An example of the amino acid sequence of wild-type hGALC SEQ ID NO: 2: An example of a base sequence encoding wild-type hGALC, synthetic sequence SEQ ID NO: 3: An example of an amino acid sequence of wild-type HSA SEQ ID NO: 4: An example of a base sequence encoding wild-type HSA, synthetic sequence SEQ ID NO: 5: An example of an amino acid sequence of HSA-hGALC SEQ ID NO: 6: An example of a base sequence encoding HSA-hGALC, synthetic sequence SEQ ID NO: 7: An example of an amino acid sequence of hGALC-HSA SEQ ID NO: 8: An example of a base sequence encoding hGALC-HSA, synthetic sequence SEQ ID NO: 9: Example of an amino acid sequence of a linker 1
SEQ ID NO: 10: Example 2 of the amino acid sequence of the linker
SEQ ID NO: 11: Example 3 of the amino acid sequence of the linker
SEQ ID NO:12: Amino acid sequence of human serum albumin Redhill SEQ ID NO:13: Amino acid sequence of mutant human serum albumin (HSA-A320T) SEQ ID NO:14: An example of an amino acid sequence of wild-type mGALC SEQ ID NO:15: An example of a base sequence encoding wild-type mGALC, synthetic sequence SEQ ID NO:16: An example of an amino acid sequence of wild-type MSA SEQ ID NO:17: An example of a base sequence encoding wild-type MSA, synthetic sequence SEQ ID NO:18: An example of an amino acid sequence of MSA-mGALC SEQ ID NO:19: An example of a base sequence encoding MSA-mGALC, synthetic sequence SEQ ID NO:20: An example of an amino acid sequence of mGALC-MSA SEQ ID NO:21: An example of a base sequence encoding mGALC-MSA, synthetic sequence SEQ ID NO:22: Amino acid sequence of human transferrin receptor SEQ ID NO:23: Amino acid sequence of light chain of anti-hTfR antibody SEQ ID NO:24: An example of amino acid sequence of wild-type hIL-10 SEQ ID NO:25: Amino acid sequence of Fab heavy chain of anti-hTfR antibody SEQ ID NO:26: pCI Insert F3 primer, synthetic sequence SEQ ID NO:27: pCI Insert R3 primer, synthetic sequence SEQ ID NO:28: Amino acid sequence of anti-hTfR antibody Fab heavy chain-HSA-hGALC SEQ ID NO:29: Nucleotide sequence encoding anti-hTfR antibody Fab heavy chain-HSA-hGALC, synthetic sequence SEQ ID NO:30: Amino acid sequence of HSA-hGALC-anti-hTfR antibody Fab heavy chain SEQ ID NO:31: Nucleotide sequence encoding HSA-hGALC-anti-hTfR antibody Fab heavy chain, synthetic sequence SEQ ID NO:32: Amino acid sequence of anti-hTfR antibody Fab heavy chain-hGALC-HSA SEQ ID NO:33: Anti-hTfR antibody SEQ ID NO:34: Amino acid sequence of hGALC-HSA-anti-hTfR antibody Fab heavy chain SEQ ID NO:35: Base sequence encoding hGALC-HSA-anti-hTfR antibody Fab heavy chain, synthetic sequence SEQ ID NO:36: Base sequence encoding anti-hTfR antibody light chain, synthetic sequence SEQ ID NO:37: An example of the amino acid sequence of wild-type hGBA SEQ ID NO:38: An example of the base sequence encoding wild-type hGBA SEQ ID NO:39: An example of the amino acid sequence of HSA-hGBA No. 40: An example of a base sequence encoding HSA-hGBA, synthetic sequence No. 41: An example of an amino acid sequence of hGBA-HSA No. 42: An example of a base sequence encoding hGBA-HSA, synthetic sequence No. 43: An example of an amino acid sequence of wild-type mGBA No. 44: An example of a base sequence encoding wild-type mGBA No. 45: An example of an amino acid sequence of MSA-mGBA No. 46: An example of a base sequence encoding MSA-mGBA, synthetic sequence No. 47: An example of an amino acid sequence of mGBA-MSA No. 48: An example of a base sequence encoding mGBA-MSA, synthetic sequence No. 49: An example of a base sequence encoding wild-type hIL-10, synthetic sequence No. 50: An example of an amino acid sequence of HSA-hIL-10 No. 51: An example of a base sequence encoding HSA-hIL-10, synthetic sequence No. 52: An example of an amino acid sequence of hIL-10-HSA No. 53: An example of a base sequence encoding hIL-10-HSA, synthetic sequence No. 54: An example of an amino acid sequence of HSA-hIL-10 2
SEQ ID NO: 55: Example 2 of the amino acid sequence of hIL-10-HSA
SEQ ID NO: 56: An example of the amino acid sequence of HSA-hIL-10 3
SEQ ID NO: 57: An example of the amino acid sequence of hIL-10-HSA 3
SEQ ID NO: 58: An example of the amino acid sequence of HSA-hIL-10 4
SEQ ID NO: 59: An example of the amino acid sequence of hIL-10-HSA 4
SEQ ID NO:60: An example of an amino acid sequence of wild-type hBDNF SEQ ID NO:61: An example of a base sequence encoding wild-type hBDNF, synthetic sequence SEQ ID NO:62: An example of an amino acid sequence of wild-type hNGF SEQ ID NO:63: An example of a base sequence encoding wild-type hNGF, synthetic sequence SEQ ID NO:64: An example of an amino acid sequence of wild-type hNT-3 SEQ ID NO:65: An example of a base sequence encoding wild-type hNT-3, synthetic sequence SEQ ID NO:66: An example of an amino acid sequence of wild-type hNT-4 SEQ ID NO:67: An example of a base sequence encoding wild-type hNT-4, synthetic sequence SEQ ID NO:68: An example of an amino acid sequence of HSA-hBDNF SEQ ID NO:69: An example of a base sequence encoding HSA-hBDNF, synthetic sequence SEQ ID NO:70: An example of an amino acid sequence of HSA-hNGF SEQ ID NO:71: An example of a base sequence encoding HSA-hNGF, synthetic sequence SEQ ID NO:72: An example of an amino acid sequence of HSA-hNT-3 SEQ ID NO:73: An example of a base sequence encoding HSA-hNT-3, synthetic sequence SEQ ID NO: 74: An example of an amino acid sequence of HSA-hNT-4 SEQ ID NO: 75: An example of a base sequence encoding HSA-hNT-4, synthetic sequence SEQ ID NO: 76: An example of an amino acid sequence of hBDNF-HSA SEQ ID NO: 77: An example of a base sequence encoding hBDNF-HSA, synthetic sequence SEQ ID NO: 78: An example of an amino acid sequence of hNGF-HSA SEQ ID NO: 79: An example of a base sequence encoding hNGF-HSA, synthetic sequence SEQ ID NO: 80: An example of an amino acid sequence of hNT-3-HSA SEQ ID NO: 81: An example of a base sequence encoding hNT-3-HSA, synthetic sequence SEQ ID NO: 82: An example of an amino acid sequence of hNT-4-HSA SEQ ID NO: 83: An example of a base sequence encoding hNT-4-HSA, synthetic sequence SEQ ID NO: 84: An example of an amino acid sequence of wild-type hBDNF (pro-form) SEQ ID NO: 85: An example of an amino acid sequence of HSA-hBDNF 2
SEQ ID NO: 86: Example 2 of the amino acid sequence of hBDNF-HSA
SEQ ID NO: 87: An example of the amino acid sequence of wild-type hNGF (pro-form) SEQ ID NO: 88: An example of the amino acid sequence of HSA-hNGF 2
SEQ ID NO: 89: Example 2 of the amino acid sequence of hNGF-HSA
SEQ ID NO: 90: An example of the amino acid sequence of wild-type hNT-3 (pro-form) SEQ ID NO: 91: An example of the amino acid sequence of HSA-hNT-3 2
SEQ ID NO: 92: Example 2 of the amino acid sequence of hNT-3-HSA
SEQ ID NO: 93: An example of the amino acid sequence of wild-type hNT-4 (pro-form) SEQ ID NO: 94: An example of the amino acid sequence of HSA-hNT-4 2
SEQ ID NO: 95: Example 2 of the amino acid sequence of hNT-4-HSA
SEQ ID NO: 96: Amino acid sequence of the light chain CDR1 of the anti-hTfR antibody SEQ ID NO: 97: Amino acid sequence of the light chain CDR2 of the anti-hTfR antibody SEQ ID NO: 98: Amino acid sequence of the light chain CDR3 of the anti-hTfR antibody SEQ ID NO: 99: Amino acid sequence of the heavy chain CDR1 of the anti-hTfR antibody
SEQ ID NO: 100: Amino acid sequence of the heavy chain CDR2 of anti-hTfR antibody 1
SEQ ID NO: 101: Amino acid sequence of the heavy chain CDR3 of anti-hTfR antibody 1
SEQ ID NO: 102: Amino acid sequence 2 of heavy chain CDR1 of anti-hTfR antibody
SEQ ID NO: 103: Amino acid sequence 2 of the heavy chain CDR2 of anti-hTfR antibody
SEQ ID NO: 104: Amino acid sequence 2 of the heavy chain CDR3 of anti-hTfR antibody
SEQ ID NO: 105: Amino acid sequence of CDR1 of hTfR affinity peptide 1
SEQ ID NO: 106: Amino acid sequence 2 of CDR1 of hTfR affinity peptide
SEQ ID NO: 107: Amino acid sequence of CDR2 of hTfR affinity peptide 1
SEQ ID NO: 108: Amino acid sequence 2 of CDR2 of hTfR affinity peptide
SEQ ID NO: 109: Amino acid sequence of CDR3 of hTfR affinity peptide 1
SEQ ID NO: 110: Amino acid sequence 2 of CDR3 of hTfR affinity peptide
SEQ ID NO: 111: Amino acid sequence 3 of CDR2 of hTfR affinity peptide
SEQ ID NO: 112: Amino acid sequence 4 of CDR2 of hTfR affinity peptide
SEQ ID NO:113: Nucleotide sequence of the first inverted terminal repeat (first ITR) of AAV of serotype 2, wild type SEQ ID NO:114: Nucleotide sequence of the first inverted terminal repeat (second ITR) of AAV of serotype 2, wild type SEQ ID NO:115: A preferred example of the nucleotide sequence of the first inverted terminal repeat (first ITR), synthetic sequence SEQ ID NO:116: A preferred example of the nucleotide sequence of the second inverted terminal repeat (second ITR), synthetic sequence SEQ ID NO:117: A preferred example of the nucleotide sequence of the first long terminal repeat (first LTR), synthetic sequence SEQ ID NO:118: A preferred example of the nucleotide sequence of the second long terminal repeat (second LTR), synthetic sequence SEQ ID NO:119: Nucleotide sequence of the leader of the Sendai virus genome SEQ ID NO:120: Nucleotide sequence of the trailer of the Sendai virus genome SEQ ID NO:121: Mouse alpha-fetoprotein enhancer/mouse albumin enhancer SEQ ID NO:122: Nucleotide sequence of chicken β-actin/MVM chimeric intron, synthetic sequence SEQ ID NO:123: Nucleotide sequence including the Rep region of AAV2, synthetic sequence SEQ ID NO:124: Nucleotide sequence of the Cap region of AAV8 SEQ ID NO:125: Amino acid sequence of mutant mouse serum albumin (MSA-A320T) SEQ ID NO:126: An example of the amino acid sequence of wild-type mIL-10 SEQ ID NO:127: An example of the base sequence encoding wild-type mIL-10, synthetic sequence SEQ ID NO:128: An example of the amino acid sequence of mIL-10-MSA SEQ ID NO:129: An example of the base sequence encoding mIL-10-MSA, synthetic sequence SEQ ID NO:130: An example of the amino acid sequence of MSA-mIL-10 SEQ ID NO:131: An example of the base sequence encoding MSA-mIL-10, synthetic sequence SEQ ID NO:132: Amino acid sequence of wild-type hBDNF with His6 added SEQ ID NO:133: An example of a base sequence encoding wild-type hBDNF with His6 added, synthetic sequence SEQ ID NO:134: Amino acid sequence of wild-type hNGF with His6 added SEQ ID NO:135: An example of a base sequence encoding wild-type hNGF with His6 added, synthetic sequence SEQ ID NO:136: Amino acid sequence of wild-type hNT-4 with His6 added SEQ ID NO:137: An example of a base sequence encoding wild-type hNT-4 with His6 added, synthetic sequence SEQ ID NO:138: Amino acid sequence of wild-type hNT-3 with His6 added SEQ ID NO:139: An example of a base sequence encoding wild-type hNT-3 with His6 added, synthetic sequence SEQ ID NO:140: Amino acid sequence of wild-type HSA with His6 added SEQ ID NO:141: An example of a base sequence encoding wild-type HSA with His6 added An example of a base sequence to be downloaded, synthetic sequence SEQ ID NO:142: forward primer 1, synthetic sequence SEQ ID NO:143: forward primer 2, synthetic sequence SEQ ID NO:144: forward primer 3, synthetic sequence SEQ ID NO:145: forward primer 4, synthetic sequence SEQ ID NO:146: forward primer 5, synthetic sequence SEQ ID NO:147: reverse primer, synthetic sequence SEQ ID NO:148: amino acid sequence of hBDNF-HSA SEQ ID NO:149: amino acid sequence of hNGF-HSA SEQ ID NO:150: amino acid sequence of hNT-4-HSA SEQ ID NO:151: amino acid sequence of hNT-3-HSA SEQ ID NO:152: amino acid sequence of HSA-hBDNF SEQ ID NO:153: amino acid sequence of HSA-hNGF SEQ ID NO:154: amino acid sequence of HSA-hNT-4 SEQ ID NO:155: amino acid sequence of HSA-hNT-3

Claims (88)

  1.  リソソーム酵素と,血清アルブミン(SA)を含んでなる,融合蛋白質。 A fusion protein comprising a lysosomal enzyme and serum albumin (SA).
  2.  該リソソーム酵素が,ヒトリソソーム酵素である,請求項1に記載の融合蛋白質。 The fusion protein of claim 1, wherein the lysosomal enzyme is a human lysosomal enzyme.
  3.  該SAが,ヒト血清アルブミン(HSA)である,請求項1又は2に記載の融合蛋白質。 The fusion protein according to claim 1 or 2, wherein the SA is human serum albumin (HSA).
  4.  該リソソーム酵素が,配列番号1で示されるアミノ酸配列を有する野生型ヒトガラクトシルセラミダーゼに対して80%以上の同一性を有するヒトガラクトシルセラミダーゼ(hGALC)であって,該SAが,配列番号3で示されるアミノ酸配列を有する野生型ヒト血清アルブミンに対して80%以上の同一性を有するヒト血清アルブミン(HSA)である,請求項1乃至3の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 1 to 3, wherein the lysosomal enzyme is human galactosylceramidase (hGALC) having 80% or more identity to wild-type human galactosylceramidase having the amino acid sequence shown in SEQ ID NO:1, and the SA is human serum albumin (HSA) having 80% or more identity to wild-type human serum albumin having the amino acid sequence shown in SEQ ID NO:3.
  5.  該hGALCが配列番号1で示されるアミノ酸配列を有する野生型hGALCに対して90%以上の同一性を有するものであり,該HSAが配列番号3で示されるアミノ酸配列を有する野生型HSAに対して90%以上の同一性を有するものである,請求項4に記載の融合蛋白質。 The fusion protein according to claim 4, wherein the hGALC has 90% or more identity to wild-type hGALC having the amino acid sequence shown in SEQ ID NO:1, and the HSA has 90% or more identity to wild-type HSA having the amino acid sequence shown in SEQ ID NO:3.
  6.  該hGALCが,配列番号1で示される野生型hGALCのアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項4に記載の融合蛋白質。 The fusion protein according to claim 4, wherein the hGALC comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1.
  7.  該hGALCが,配列番号1で示される野生型hGALCのアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項4に記載の融合蛋白質。 The fusion protein according to claim 4, wherein the hGALC comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1.
  8.  該hGALCが,配列番号1で示される野生型hGALCのアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項4に記載の融合蛋白質。 The fusion protein according to claim 4, wherein the hGALC comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1.
  9.  該hGALCが,配列番号1で示される野生型hGALCのアミノ酸配列に対して1個のアミノ酸が置換されたアミノ酸配列を含んでなるものである,請求項4に記載の融合蛋白質。 The fusion protein according to claim 4, wherein the hGALC comprises an amino acid sequence in which one amino acid has been substituted with respect to the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1.
  10.  該アミノ酸の置換が,側鎖が水酸化反応され得るアミノ酸であるアミノ酸ファミリー内での置換である,請求項9に記載の融合蛋白質。 The fusion protein according to claim 9, wherein the amino acid substitution is within an amino acid family in which the side chain is an amino acid that can be hydroxylated.
  11.  該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項4乃至10の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 4 to 10, wherein the HSA comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
  12.  該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項4乃至10の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 4 to 10, wherein the HSA comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
  13.  該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項4乃至10の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 4 to 10, wherein the HSA comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
  14.  該hGALCが配列番号1で示される野生型hGALCのアミノ酸配列を含んでなるものであり,該HSAが配列番号3で示される野生型ヒト血清アルブミンのアミノ酸配列を含んでなるものである,請求項4に記載の融合蛋白質。 The fusion protein according to claim 4, wherein the hGALC comprises the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1, and the HSA comprises the amino acid sequence of wild-type human serum albumin shown in SEQ ID NO:3.
  15.  該hGALCが配列番号1で示される野生型hGALCのアミノ酸配列を含んでなるものであり,該HSAが配列番号12で示される野生型ヒト血清アルブミンのアミノ酸配列を含んでなるものである,請求項4に記載の融合蛋白質。 The fusion protein according to claim 4, wherein the hGALC comprises the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1, and the HSA comprises the amino acid sequence of wild-type human serum albumin shown in SEQ ID NO:12.
  16.  該hGALCが配列番号1で示される野生型hGALCのアミノ酸配列を含んでなるものであり,該HSAが配列番号13で示される野生型ヒト血清アルブミンのアミノ酸配列を含んでなるものである,請求項4に記載の融合蛋白質。 The fusion protein according to claim 4, wherein the hGALC comprises the amino acid sequence of wild-type hGALC shown in SEQ ID NO:1, and the HSA comprises the amino acid sequence of wild-type human serum albumin shown in SEQ ID NO:13.
  17.  該hGALCのC末端に,直接又はリンカーを介して,該HSAが結合したものである,請求項4乃至16の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 4 to 16, in which the HSA is bound to the C-terminus of the hGALC directly or via a linker.
  18.  該HSAのC末端に,直接又はリンカーを介して,該hGALCが結合したものである,請求項4乃至16の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 4 to 16, in which the hGALC is bound to the C-terminus of the HSA directly or via a linker.
  19.  該リンカーが,1~150個のアミノ酸からなるペプチド鎖である,請求項17又は18に記載の融合蛋白質。 The fusion protein according to claim 17 or 18, wherein the linker is a peptide chain consisting of 1 to 150 amino acids.
  20.  該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列からなるものである,請求項19に記載の融合蛋白質:
     (a)Gly;
     (b)Ser;
     (c)Gly Ser;
     (d)Gly Gly Ser;
     (e)配列番号9で示されるアミノ酸配列;
     (f)配列番号10で示されるアミノ酸配列;及び,
     (g)配列番号11で示されるアミノ酸配列。
    The fusion protein according to claim 19, wherein the linker comprises an amino acid sequence selected from the group consisting of the following (a) to (g):
    (a) Gly;
    (b) Ser;
    (c) GlySer;
    (d) GlyGlySer;
    (e) the amino acid sequence set forth in SEQ ID NO:9;
    (f) the amino acid sequence set forth in SEQ ID NO: 10; and
    (g) The amino acid sequence shown in SEQ ID NO:11.
  21.  該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が2~10回反復したアミノ酸配列からなるものである,請求項19に記載の融合蛋白質:
     (a)Gly;
     (b)Ser;
     (c)Gly Ser;
     (d)Gly Gly Ser;
     (e)配列番号9で示されるアミノ酸配列;
     (f)配列番号10で示されるアミノ酸配列;及び,
     (g)配列番号11で示されるアミノ酸配列。
    The fusion protein according to claim 19, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 10 times:
    (a) Gly;
    (b) Ser;
    (c) GlySer;
    (d) GlyGlySer;
    (e) the amino acid sequence set forth in SEQ ID NO:9;
    (f) the amino acid sequence set forth in SEQ ID NO: 10; and
    (g) The amino acid sequence shown in SEQ ID NO:11.
  22.  該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が2~6回反復したアミノ酸配列からなるものである,請求項19に記載の融合蛋白質:
     (a)Gly;
     (b)Ser;
     (c)Gly Ser;
     (d)Gly Gly Ser;
     (e)配列番号9で示されるアミノ酸配列;
     (f)配列番号10で示されるアミノ酸配列;及び,
     (g)配列番号11で示されるアミノ酸配列。
    The fusion protein according to claim 19, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 6 times:
    (a) Gly;
    (b) Ser;
    (c) GlySer;
    (d) GlyGlySer;
    (e) the amino acid sequence set forth in SEQ ID NO:9;
    (f) the amino acid sequence set forth in SEQ ID NO: 10; and
    (g) The amino acid sequence shown in SEQ ID NO:11.
  23.  該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が3~5回反復したアミノ酸配列からなるものである,請求項19に記載の融合蛋白質:
     (a)Gly;
     (b)Ser;
     (c)Gly Ser;
     (d)Gly Gly Ser;
     (e)配列番号9で示されるアミノ酸配列;
     (f)配列番号10で示されるアミノ酸配列;及び,
     (g)配列番号11で示されるアミノ酸配列。
    The fusion protein according to claim 19, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 3 to 5 times:
    (a) Gly;
    (b) Ser;
    (c) GlySer;
    (d) GlyGlySer;
    (e) the amino acid sequence set forth in SEQ ID NO:9;
    (f) the amino acid sequence set forth in SEQ ID NO: 10; and
    (g) The amino acid sequence shown in SEQ ID NO:11.
  24.  該リンカーが,Gly Serで示されるアミノ酸配列からなるものである,請求項19に記載の融合蛋白質。 The fusion protein according to claim 19, wherein the linker consists of an amino acid sequence represented by Gly Ser.
  25.  配列番号5で示されるアミノ酸配列に対して80%以上の同一性を有するアミノ酸配列を含んでなる,請求項18に記載の融合蛋白質。 The fusion protein according to claim 18, comprising an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:5.
  26.  配列番号5で示されるアミノ酸配列に対して90%以上の同一性を有するアミノ酸配列を含んでなる,請求項18に記載の融合蛋白質。 The fusion protein according to claim 18, comprising an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:5.
  27.  該融合蛋白質が,配列番号5で示されるアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項26に記載の融合蛋白質。 The fusion protein according to claim 26, which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:5.
  28.  該融合蛋白質が,配列番号5で示されるアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項26に記載の融合蛋白質。 The fusion protein according to claim 26, which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:5.
  29.  該融合蛋白質が,配列番号5で示されるアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項26に記載の融合蛋白質。 The fusion protein according to claim 26, which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:5.
  30.  配列番号5で示されるアミノ酸配列を含んでなる,請求項18に記載の融合蛋白質。 The fusion protein according to claim 18, comprising the amino acid sequence shown in SEQ ID NO:5.
  31.  配列番号7で示されるアミノ酸配列に対して80%以上の同一性を有するアミノ酸配列を含んでなる,請求項17に記載の融合蛋白質。 The fusion protein according to claim 17, comprising an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:7.
  32.  配列番号7で示されるアミノ酸配列に対して90%以上の同一性を有するアミノ酸配列を含んでなる,請求項17に記載の融合蛋白質。 The fusion protein according to claim 17, comprising an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:7.
  33.  該融合蛋白質が,配列番号7で示されるアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項32に記載の融合蛋白質。 The fusion protein according to claim 32, which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:7.
  34.  該融合蛋白質が,配列番号7で示されるアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項32に記載の融合蛋白質。 The fusion protein according to claim 32, which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:7.
  35.  該融合蛋白質が,配列番号7で示されるアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項32に記載の融合蛋白質。 The fusion protein according to claim 32, which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:7.
  36.  配列番号7で示されるアミノ酸配列を含んでなる,請求項17に記載の融合蛋白質。 The fusion protein according to claim 17, comprising the amino acid sequence shown in SEQ ID NO:7.
  37.  通常の野生型のhGALCの比活性と比較して,10%以上の比活性を有するものである,請求項4乃至36の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 4 to 36, which has a specific activity of 10% or more compared to the specific activity of normal wild-type hGALC.
  38.  該リソソーム酵素が,配列番号37で示されるアミノ酸配列を有する野生型ヒトグルコセレブロシダーゼに対して80%以上の同一性を有するヒトグルコセレブロシダーゼ(hGBA)であって,該SAが,配列番号3で示されるアミノ酸配列を有する野生型ヒト血清アルブミンに対して80%以上の同一性を有するヒト血清アルブミン(HSA)である,請求項1乃至3の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 1 to 3, wherein the lysosomal enzyme is human glucocerebrosidase (hGBA) having 80% or more identity to wild-type human glucocerebrosidase having the amino acid sequence shown in SEQ ID NO: 37, and the SA is human serum albumin (HSA) having 80% or more identity to wild-type human serum albumin having the amino acid sequence shown in SEQ ID NO: 3.
  39.  該hGBAが配列番号37で示されるアミノ酸配列を有する野生型hGBAに対して90%以上の同一性を有するものであり,該HSAが配列番号3で示されるアミノ酸配列を有する野生型HSAに対して90%以上の同一性を有するものである,請求項38に記載の融合蛋白質。 The fusion protein according to claim 38, wherein the hGBA has 90% or more identity to wild-type hGBA having the amino acid sequence shown in SEQ ID NO:37, and the HSA has 90% or more identity to wild-type HSA having the amino acid sequence shown in SEQ ID NO:3.
  40.  該hGBAが,配列番号37で示される野生型hGBAのアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項38に記載の融合蛋白質。 The fusion protein according to claim 38, wherein the hGBA comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37.
  41.  該hGBAが,配列番号37で示される野生型hGBAのアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項38に記載の融合蛋白質。 The fusion protein according to claim 38, wherein the hGBA comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37.
  42.  該hGBAが,配列番号37で示される野生型hGBAのアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項38に記載の融合蛋白質。 The fusion protein according to claim 38, wherein the hGBA comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37.
  43.  該hGBAが,配列番号37で示される野生型hGBAのアミノ酸配列に対して1個のアミノ酸が置換されたアミノ酸配列を含んでなるものである,請求項38に記載の融合蛋白質。 The fusion protein according to claim 38, wherein the hGBA comprises an amino acid sequence in which one amino acid has been substituted with respect to the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37.
  44.  該アミノ酸の置換が,側鎖が水酸化反応され得るアミノ酸であるアミノ酸ファミリー内での置換である,請求項43に記載の融合蛋白質。 The fusion protein of claim 43, wherein the amino acid substitution is within an amino acid family whose side chains can be hydroxylated.
  45.  該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項38乃至44の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 38 to 44, wherein the HSA comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted, or/and added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
  46.  該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項38乃至44の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 38 to 44, wherein the HSA comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
  47.  該HSAが,配列番号3で示される野生型HSAのアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項38乃至44の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 38 to 44, wherein the HSA comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
  48.  該hGBAが配列番号37で示される野生型hGBAのアミノ酸配列を含んでなるものであり,該HSAが配列番号3で示される野生型HSAのアミノ酸配列を含んでなるものである,請求項38に記載の融合蛋白質。 The fusion protein of claim 38, wherein the hGBA comprises the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, and the HSA comprises the amino acid sequence of wild-type HSA shown in SEQ ID NO:3.
  49.  該hGBAが配列番号37で示される野生型hGBAのアミノ酸配列を含んでなるものであり,該HSAが配列番号12で示される野生型HSAのアミノ酸配列を含んでなるものである,請求項38に記載の融合蛋白質。 The fusion protein of claim 38, wherein the hGBA comprises the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, and the HSA comprises the amino acid sequence of wild-type HSA shown in SEQ ID NO:12.
  50.  該hGBAが配列番号37で示される野生型hGBAのアミノ酸配列を含んでなるものであり,該HSAが配列番号13で示される野生型HSAのアミノ酸配列を含んでなるものである,請求項38に記載の融合蛋白質。 The fusion protein of claim 38, wherein the hGBA comprises the amino acid sequence of wild-type hGBA shown in SEQ ID NO:37, and the HSA comprises the amino acid sequence of wild-type HSA shown in SEQ ID NO:13.
  51.  該hGBAのC末端に,直接又はリンカーを介して,該HSAが結合したものである,請求項38乃至50の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 38 to 50, in which the HSA is bound to the C-terminus of the hGBA directly or via a linker.
  52.  該HSAのC末端に,直接又はリンカーを介して,該hGBAが結合したものである,請求項38乃至50の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 38 to 50, in which the hGBA is bound to the C-terminus of the HSA directly or via a linker.
  53.  該リンカーが,1~150個のアミノ酸からなるペプチド鎖である,請求項51又は52に記載の融合蛋白質。 The fusion protein according to claim 51 or 52, wherein the linker is a peptide chain consisting of 1 to 150 amino acids.
  54.  該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列からなるものである,請求項53に記載の融合蛋白質:
     (a)Gly;
     (b)Ser;
     (c)Gly Ser;
     (d)Gly Gly Ser;
     (e)配列番号9で示されるアミノ酸配列;
     (f)配列番号10で示されるアミノ酸配列;及び,
     (g)配列番号11で示されるアミノ酸配列。
    The fusion protein of claim 53, wherein the linker comprises an amino acid sequence selected from the group consisting of:
    (a) Gly;
    (b) Ser;
    (c) GlySer;
    (d) GlyGlySer;
    (e) the amino acid sequence set forth in SEQ ID NO:9;
    (f) the amino acid sequence set forth in SEQ ID NO: 10; and
    (g) The amino acid sequence shown in SEQ ID NO:11.
  55.  該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が2~10回反復したアミノ酸配列からなるものである,請求項53に記載の融合蛋白質:
     (a)Gly;
     (b)Ser;
     (c)Gly Ser;
     (d)Gly Gly Ser;
     (e)配列番号9で示されるアミノ酸配列;
     (f)配列番号10で示されるアミノ酸配列;及び,
     (g)配列番号11で示されるアミノ酸配列。
    The fusion protein according to claim 53, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 10 times:
    (a) Gly;
    (b) Ser;
    (c) GlySer;
    (d) GlyGlySer;
    (e) the amino acid sequence set forth in SEQ ID NO:9;
    (f) the amino acid sequence set forth in SEQ ID NO: 10; and
    (g) The amino acid sequence shown in SEQ ID NO:11.
  56.  該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が2~6回反復したアミノ酸配列からなるものである,請求項53に記載の融合蛋白質:
     (a)Gly;
     (b)Ser;
     (c)Gly Ser;
     (d)Gly Gly Ser;
     (e)配列番号9で示されるアミノ酸配列;
     (f)配列番号10で示されるアミノ酸配列;及び,
     (g)配列番号11で示されるアミノ酸配列。
    The fusion protein according to claim 53, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 2 to 6 times:
    (a) Gly;
    (b) Ser;
    (c) GlySer;
    (d) GlyGlySer;
    (e) the amino acid sequence set forth in SEQ ID NO:9;
    (f) the amino acid sequence set forth in SEQ ID NO: 10; and
    (g) The amino acid sequence shown in SEQ ID NO:11.
  57.  該リンカーが,以下の(a)~(g)からなる群から選択されるアミノ酸配列が3~5回反復したアミノ酸配列からなるものである,請求項53に記載の融合蛋白質:
     (a)Gly;
     (b)Ser;
     (c)Gly Ser;
     (d)Gly Gly Ser;
     (e)配列番号9で示されるアミノ酸配列;
     (f)配列番号10で示されるアミノ酸配列;及び,
     (g)配列番号11で示されるアミノ酸配列。
    The fusion protein according to claim 53, wherein the linker comprises an amino acid sequence in which an amino acid sequence selected from the group consisting of the following (a) to (g) is repeated 3 to 5 times:
    (a) Gly;
    (b) Ser;
    (c) GlySer;
    (d) GlyGlySer;
    (e) the amino acid sequence set forth in SEQ ID NO:9;
    (f) the amino acid sequence set forth in SEQ ID NO: 10; and
    (g) The amino acid sequence shown in SEQ ID NO:11.
  58.  該リンカーが,Gly Serで示されるアミノ酸配列からなるものである,請求項53に記載の融合蛋白質。 The fusion protein according to claim 53, wherein the linker consists of an amino acid sequence represented by Gly Ser.
  59.  配列番号39で示されるアミノ酸配列に対して80%以上の同一性を有するアミノ酸配列を含んでなる,請求項52に記載の融合蛋白質。 The fusion protein according to claim 52, comprising an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:39.
  60.  配列番号39で示されるアミノ酸配列に対して90%以上の同一性を有するアミノ酸配列を含んでなる,請求項52に記載の融合蛋白質。 The fusion protein according to claim 52, comprising an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:39.
  61.  該融合蛋白質が,配列番号39で示されるアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項60に記載の融合蛋白質。 The fusion protein according to claim 60, which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:39.
  62.  該融合蛋白質が,配列番号39で示されるアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項60に記載の融合蛋白質。 The fusion protein according to claim 60, which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:39.
  63.  該融合蛋白質が,配列番号39で示されるアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項60に記載の融合蛋白質。 The fusion protein according to claim 60, which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:39.
  64.  配列番号41で示されるアミノ酸配列を含んでなる,請求項51に記載の融合蛋白質。 The fusion protein of claim 51, comprising the amino acid sequence shown in SEQ ID NO:41.
  65.  配列番号41で示されるアミノ酸配列に対して80%以上の同一性を有するアミノ酸配列を含んでなる,請求項51に記載の融合蛋白質。 The fusion protein according to claim 51, comprising an amino acid sequence having 80% or more identity to the amino acid sequence shown in SEQ ID NO:41.
  66.  配列番号41で示されるアミノ酸配列に対して90%以上の同一性を有するアミノ酸配列を含んでなる,請求項51に記載の融合蛋白質。 The fusion protein according to claim 51, comprising an amino acid sequence having 90% or more identity to the amino acid sequence shown in SEQ ID NO:41.
  67.  該融合蛋白質が,配列番号41で示されるアミノ酸配列に対して1~10個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項66に記載の融合蛋白質。 The fusion protein according to claim 66, which comprises an amino acid sequence in which 1 to 10 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:41.
  68.  該融合蛋白質が,配列番号41で示されるアミノ酸配列に対して1~5個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項66に記載の融合蛋白質。 The fusion protein according to claim 66, which comprises an amino acid sequence in which 1 to 5 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:41.
  69.  該融合蛋白質が,配列番号41で示されるアミノ酸配列に対して1~3個のアミノ酸が置換,欠失又は/及び付加されたアミノ酸配列を含んでなるものである,請求項66に記載の融合蛋白質。 The fusion protein according to claim 66, which comprises an amino acid sequence in which 1 to 3 amino acids have been substituted, deleted and/or added to the amino acid sequence shown in SEQ ID NO:41.
  70.  配列番号41で示されるアミノ酸配列を含んでなる,請求項51に記載の融合蛋白質。 The fusion protein of claim 51, comprising the amino acid sequence shown in SEQ ID NO:41.
  71.  通常の野生型のhGBAの比活性と比較して,10%以上の比活性を有するものである,請求項38乃至70の何れかに記載の融合蛋白質。 The fusion protein according to any one of claims 38 to 70, which has a specific activity of 10% or more compared to the specific activity of normal wild-type hGBA.
  72.  請求項1乃至71の何れかに記載の融合蛋白質をコードする遺伝子を含んでなるDNA。 DNA comprising a gene encoding the fusion protein according to any one of claims 1 to 71.
  73.  請求項72に記載のDNAを含んでなる発現ベクター。 An expression vector comprising the DNA of claim 72.
  74.  請求項73に記載の発現ベクターで形質転換された哺乳動物細胞。 A mammalian cell transformed with the expression vector of claim 73.
  75.  請求項74に記載の哺乳動物細胞を無血清培地で培養するステップを含む,融合蛋白質の製造方法。 A method for producing a fusion protein, comprising the step of culturing the mammalian cell according to claim 74 in a serum-free medium.
  76.  請求項1乃至71の何れかに記載の融合蛋白質と,抗体との結合体であって,該抗体が脳血管内皮細胞上の受容体と結合することにより,該融合蛋白質が血液脳関門(BBB)を通過することのできるものである,結合体。 A conjugate of the fusion protein according to any one of claims 1 to 71 and an antibody, in which the antibody binds to a receptor on cerebrovascular endothelial cells, thereby enabling the fusion protein to pass through the blood-brain barrier (BBB).
  77.  該脳血管内皮細胞上の受容体が,インスリン受容体,トランスフェリン受容体,レプチン受容体,リポタンパク質受容体,及びIGF受容体からなる群から選択されるものである,請求項76に記載の結合体。 The conjugate of claim 76, wherein the receptor on the cerebrovascular endothelial cell is selected from the group consisting of an insulin receptor, a transferrin receptor, a leptin receptor, a lipoprotein receptor, and an IGF receptor.
  78.  該脳血管内皮細胞上の受容体が,トランスフェリン受容体である,請求項76に記載の結合体。 The conjugate of claim 76, wherein the receptor on cerebrovascular endothelial cells is a transferrin receptor.
  79.  該抗体が,Fab抗体,F(ab’)抗体,F(ab’)抗体,単一ドメイン抗体,一本鎖抗体,又はFc抗体の何れかである,請求項76乃至78の何れかに記載の結合体。 79. The conjugate of any of claims 76 to 78, wherein the antibody is a Fab antibody, an F(ab') 2 antibody, an F(ab') antibody, a single domain antibody, a single chain antibody, or an Fc antibody.
  80.  該融合蛋白質が,該抗体の軽鎖のC末端側又はN末端側の何れかに結合しているものである,請求項76乃至79の何れかに記載の結合体。 The conjugate according to any one of claims 76 to 79, wherein the fusion protein is bound to either the C-terminus or the N-terminus of the light chain of the antibody.
  81.  該融合蛋白質が,該抗体の重鎖のC末端側又はN末端側の何れかに結合しているものである,請求項76乃至79の何れかに記載の結合体。 The conjugate according to any one of claims 76 to 79, wherein the fusion protein is bound to either the C-terminus or the N-terminus of the heavy chain of the antibody.
  82.  該融合蛋白質が,該抗体の軽鎖のC末端側若しくはN末端側の何れか,又は重鎖のC末端側若しくはN末端側の何れかに,リンカー配列を介して結合しているものである,請求項76乃至81の何れかに記載の結合体。 The conjugate according to any one of claims 76 to 81, wherein the fusion protein is bound to either the C-terminal or N-terminal side of the light chain of the antibody, or to either the C-terminal or N-terminal side of the heavy chain, via a linker sequence.
  83.  該リンカー配列が,1~50個のアミノ酸残基からなるものである,請求項82に記載の結合体。 The conjugate according to claim 82, wherein the linker sequence consists of 1 to 50 amino acid residues.
  84.  該リンカー配列が,1個のグリシン,1個のセリン,アミノ酸配列Gly-Ser,アミノ酸配列Ser-Ser,アミノ酸配列Gly-Gly-Ser,配列番号9のアミノ酸配列,配列番号10のアミノ酸配列,配列番号11のアミノ酸配列,及びこれらのアミノ酸配列が1~10個連続してなるアミノ酸配列からなる群より選ばれるアミノ酸配列を含んでなるものである,請求項83に記載の結合体。 The conjugate according to claim 83, wherein the linker sequence comprises an amino acid sequence selected from the group consisting of one glycine, one serine, the amino acid sequence Gly-Ser, the amino acid sequence Ser-Ser, the amino acid sequence Gly-Gly-Ser, the amino acid sequence of SEQ ID NO: 9, the amino acid sequence of SEQ ID NO: 10, the amino acid sequence of SEQ ID NO: 11, and an amino acid sequence consisting of 1 to 10 consecutive amino acids of these amino acid sequences.
  85.  請求項76乃至84の何れかに記載の結合体をコードする遺伝子を含んでなるDNA。 DNA comprising a gene encoding the conjugate of any one of claims 76 to 84.
  86.  請求項85に記載のDNAを含んでなる発現ベクター。 An expression vector comprising the DNA of claim 85.
  87.  請求項86に記載の発現ベクターで形質転換された哺乳動物細胞。 A mammalian cell transformed with the expression vector of claim 86.
  88.  請求項87に記載の哺乳動物細胞を無血清培地で培養するステップを含む,生理活性を有する蛋白質とSAとの融合蛋白質と抗体との結合体の製造方法。 A method for producing a conjugate of an antibody and a fusion protein of a physiologically active protein and SA, comprising the step of culturing the mammalian cell according to claim 87 in a serum-free medium.
PCT/JP2023/036884 2022-10-11 2023-10-11 Fusion protein of serum albumin and physiologically active protein WO2024080304A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2022163603 2022-10-11
JP2022-163603 2022-10-11

Publications (1)

Publication Number Publication Date
WO2024080304A1 true WO2024080304A1 (en) 2024-04-18

Family

ID=90669587

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2023/036884 WO2024080304A1 (en) 2022-10-11 2023-10-11 Fusion protein of serum albumin and physiologically active protein

Country Status (2)

Country Link
JP (1) JP2024056665A (en)
WO (1) WO2024080304A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007522806A (en) * 2004-02-09 2007-08-16 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド Albumin fusion protein
WO2018124121A1 (en) * 2016-12-26 2018-07-05 Jcrファーマ株式会社 Novel anti-human transferrin receptor antibody capable of penetrating blood-brain barrier
WO2023090409A1 (en) * 2021-11-19 2023-05-25 Jcrファーマ株式会社 Peptide having affinity for human transferrin receptor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007522806A (en) * 2004-02-09 2007-08-16 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド Albumin fusion protein
WO2018124121A1 (en) * 2016-12-26 2018-07-05 Jcrファーマ株式会社 Novel anti-human transferrin receptor antibody capable of penetrating blood-brain barrier
WO2023090409A1 (en) * 2021-11-19 2023-05-25 Jcrファーマ株式会社 Peptide having affinity for human transferrin receptor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MATSUDA, JUNKO: "Significance of sphingolipid activator proteins-saposins and pro-saposin-in health and disease", JOURNAL OF JAPANESE BIOCHEMICAL SOCIETY, NIPPON SEIKAGAKKAI, TOKYO, JP, vol. 89, no. 6, 25 December 2017 (2017-12-25), JP , pages 808 - 819, XP009554110, ISSN: 0037-1017, DOI: 10.14952/SEIKAGAKU.2017.890808 *

Also Published As

Publication number Publication date
JP2024056665A (en) 2024-04-23

Similar Documents

Publication Publication Date Title
TWI769982B (en) Anti-human transferrin receptor antibody that passes through blood-brain barrier
JP6616189B2 (en) A novel anti-transferrin receptor antibody that crosses the blood-brain barrier
US20200384061A1 (en) Fusion Protein Including BDNF
WO2022153957A1 (en) Nucleic acid molecule containing incorporated gene encoding fused protein of ligand and protein having physiological activity
EP4424325A1 (en) Peptide having affinity for human transferrin receptor
TW202128740A (en) Variant igf2 constructs
WO2022045273A1 (en) MUTANT OF α-N-ACETYLGLUCOSAMINIDASE
US20180009904A1 (en) Lysosomal targeting and uses thereof
WO2024080304A1 (en) Fusion protein of serum albumin and physiologically active protein
WO2024080305A1 (en) Fusion protein of serum albumin and bioactive protein
WO2024080306A1 (en) Fusion protein of serum albumin and physiologically-active protein
US20240309076A1 (en) Tau-specific antibody gene therapy compositions, methods and uses thereof
TW202246326A (en) Method for producing antibody-lysosomal enzyme fusion protein
WO2023127879A1 (en) Fusion protein of anti-transferrin receptor antibody and bioactive protein for safe gene therapy
WO2024010067A1 (en) Nuecleic acid molecule, vector, recombinant cells, and drug for treating central nervous system diseases
WO2024166950A1 (en) Human transferrin receptor-avid peptide
WO2023204202A1 (en) Glycoprotein having reduced number of mannose-6-phosphates
WO2023187728A1 (en) Gene therapy for diseases with cns manifestations
WO2024026494A1 (en) Viral particles retargeted to transferrin receptor 1

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: 23877307

Country of ref document: EP

Kind code of ref document: A1