WO2025089345A1 - Anti-synaptogyrin 3 antibody - Google Patents
Anti-synaptogyrin 3 antibody Download PDFInfo
- Publication number
- WO2025089345A1 WO2025089345A1 PCT/JP2024/037921 JP2024037921W WO2025089345A1 WO 2025089345 A1 WO2025089345 A1 WO 2025089345A1 JP 2024037921 W JP2024037921 W JP 2024037921W WO 2025089345 A1 WO2025089345 A1 WO 2025089345A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- antibody
- present
- amino acid
- seq
- acid sequence
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/04—Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
- A61K38/07—Tetrapeptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K49/00—Preparations for testing in vivo
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K51/00—Preparations containing radioactive substances for use in therapy or testing in vivo
- A61K51/02—Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
- A61K51/04—Organic compounds
- A61K51/08—Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
- A61K51/10—Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K19/00—Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/74—Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/80—Vectors or expression systems specially adapted for eukaryotic hosts for fungi
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/80—Vectors or expression systems specially adapted for eukaryotic hosts for fungi
- C12N15/81—Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/10—Cells modified by introduction of foreign genetic material
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/02—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
Definitions
- the present invention relates to a monoclonal antibody that binds to the intravesicular domain of synaptogyrin 3.
- the antibody of the present invention can be used as a targeting agent for delivering a physiologically active substance and/or a labeled substance to a motor neuron, and a pharmaceutical composition containing the same.
- the nervous system is made up of the central nervous system and peripheral nervous system, and regulates various emotions and the functions of muscles, internal organs, etc.
- Such nervous function can be impaired by nerve damage, disease, aging, etc., which can impair physical and mental health and have a significant impact on social life. Therefore, maintaining and improving nervous function is an extremely important issue, as it is directly linked to maintaining and improving quality of life (QOL).
- QOL quality of life
- the central and peripheral nerves are each composed of nerve cells, and these cells exchange signals with each other through synapses.
- a synapse is a junction that includes a gap formed between the axon terminal of a nerve cell (presynapse) and the dendrite of another nerve cell or a cell of a skeletal muscle or organ (postsynapse), and signals are transmitted when chemicals released from the presynapse bind to receptors present in the postsynapse. Synapse formation is triggered by the interaction of specific membrane proteins expressed in the presynapse and postsynapse.
- Patent Document 1 describes that a specific peptide has the effect of promoting dendritic outgrowth and synapse formation in primary cultured cortical neuron cells (PCN), and that such peptides are used to treat mild cognitive impairment or early dementia.
- Patent Document 2 describes that C-terminal fragment ⁇ (CTF ⁇ ), which is generated by cleavage of amyloid precursor protein (APP) by ⁇ -secretase, promotes synapse formation, and that CTF ⁇ is used to treat neurodegenerative diseases, etc.
- CTF ⁇ C-terminal fragment ⁇
- Patent Document 3 describes a method for culturing motor neurons with presynapses using microbeads with LRRTM molecules or fusion proteins containing said molecules immobilized on their surface.
- the present inventors have developed a method that enables the controlled delivery of drugs to target motor neurons, using antibodies capable of binding to membrane proteins present in the synaptic vesicles of motor neurons (PCT/JP2023/016125). According to this method, it has been confirmed that it is possible to obtain the pharmacological effects of a physiologically active substance linked to an antibody in a target motor neuron, using an antibody capable of binding to the intravesicular domain of several types of vesicular membrane proteins, including synaptotagmin 2. However, because such a method of use had not been anticipated until now, no excellent antibodies capable of binding to membrane proteins that would exert a good effect using this method were known.
- the objective of the present invention is therefore to provide an excellent monoclonal antibody capable of binding to a membrane protein present in the synaptic vesicles of motor neurons, and to provide a means of targeting a substance to a motor neuron and a means of visualizing the targeting site using the same.
- An antibody capable of binding to the intravesicular domain of synaptogyrin 3, comprising a heavy chain variable region including an HCDR1 having the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 having the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 having the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region including an LCDR1 having the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 having the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 having the amino acid sequence shown in SEQ ID NO: 8.
- [3] A conjugate of the antibody according to [1] or [2] with a labeling substance and/or a physiologically active substance.
- [4] A nucleic acid molecule having a base sequence encoding the antibody according to [1] or [2].
- [5] A cell comprising the nucleic acid molecule according to [4].
- [6] A targeting agent for motor neurons, comprising the antibody according to [1] or [2].
- the targeting agent according to [6] further comprising a labeling substance and/or a physiologically active substance.
- the targeting agent according to [7] comprising a conjugate of the antibody with the labeling substance and/or the physiologically active substance.
- the targeting agent according to any one of [6] to [11] which is taken up into a cell by endocytosis.
- a pharmaceutical composition comprising the targeting agent according to any one of [7] to [12].
- a method for targeting a labeled substance and/or a physiologically active substance to a motor neuron comprising the steps of contacting a motor neuron with the targeting agent according to any one of [7] to [12] and/or the pharmaceutical composition according to [13], and delivering the targeting agent and/or the pharmaceutical composition to a synapse of the motor neuron.
- a method for visualizing motor neurons comprising the steps of contacting a targeting agent, which is the motor neuron visualization agent described in [10], with a motor neuron, delivering the targeting agent to a synapse of the motor neuron, and detecting a signal of the labeling substance.
- [16] A method for preventing or treating a condition or disease, comprising the steps of contacting a motor neuron with the targeting agent according to any one of [7] to [12], and delivering the targeting agent to the motor neuron synapse.
- the targeting agent comprises a conjugate of the antibody and the physiologically active substance.
- an antibody capable of binding to the intravesicular domain of synaptogyrin 3 in the manufacture of a pharmaceutical comprising the antibody and a physiologically active substance wherein the antibody comprises a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
- a composition for targeting motor neurons comprising an antibody capable of binding to the intravesicular domain of synaptogyrin 3, wherein the antibody comprises a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
- An antibody capable of binding to the intravesicular domain of synaptogyrin 3 for use in a method for preventing or treating a condition or disease comprising a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
- An antibody capable of binding to the intravesicular domain of synaptogyrin 3 for use in a method for targeting a labeling substance and/or a physiologically active substance to a motor neuron comprising a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
- This specification includes the disclosure of Japanese Patent Application No. 2023-182983, which is the priority basis of this application.
- the present invention provides a monoclonal antibody capable of targeting a desired substance to motor neurons.
- the substance is a physiologically active substance or a therapeutic agent, conditions and/or diseases caused by abnormalities in motor neurons can be treated.
- the substance is a labeling substance, motor neurons can be visualized.
- Figure 1 is a schematic diagram showing the experimental procedure for induction of presynaptic sites and delivery of antibodies using LRRTM2 beads.
- nerve axons extend from the neurospheres.
- LRRTM2 beads seeding LRRTM2 beads on the neurospheres, presynaptic sites are induced from the extended nerve axons on the surface of the LRRTM2 beads.
- FIG. 2 shows fluorescence images of the control antibody group, which was a conjugate of normal rabbit antibody and monomethyl auristatin E (MMAE).
- the fluorescence signal shown in the figure is a signal based on ⁇ III tubulin (Tuj1).
- the black dashed circle indicates the position of the neurosphere.
- Figures B and C show enlarged images of the area in the white frame in Figure A.
- the scale bar indicates 1 mm, and in Figures B and C, the scale bar indicates 100 ⁇ m.
- Figure 3 shows fluorescence images of the SYT2 antibody group in which anti-synaptotagmin 2 intravesicular domain (N-terminus) polyclonal antibody (polyclonal anti-SYT2 N-terminus antibody) was conjugated with MMAE.
- the fluorescent signal shown in the figure is a signal based on Tuj1.
- Figures B and C show enlarged images of the area enclosed in the white frame in Figure A.
- the scale bar indicates 1 mm, and in Figures B and C, the scale bar indicates 100 ⁇ m.
- Figure 4 shows a graph quantifying the pharmacological effect based on MMAE.
- the relative axonal mass is a standardized value, with the result when a control normal rabbit antibody and MMAE conjugate was used ("Cont. IgG-MMAE" in the figure) taken as 100%.
- the dashed line indicates the position where the relative axonal mass is 100%
- MMAE shows the result for the single-infusion group in which MMAE was introduced alone
- ⁇ -SYT2 IgG-MMAE shows the result for the SYT2 antibody group in which a polyclonal anti-SYT2 N-terminal antibody and MMAE conjugate were introduced.
- the error bars indicate the standard error, "*” indicates p ⁇ 0.05, and "***" indicates p ⁇ 0.001.
- Figure 5 shows a graph quantifying the pharmacological effect based on MMAE.
- the relative axonal volume is a standardized value, with the result when a control normal rabbit antibody and MMAE conjugate was used (in the figure, "Cont. IgG-MMAE") set as 100%.
- the dashed line indicates the position where the relative axonal volume is 100%, and " ⁇ -SYNGR3 IgG-MMAE” shows the result of the SYNGR3 antibody group introduced with a polyclonal anti-SYNGR3 intravesicular domain antibody and MMAE conjugate.
- the error bars indicate the standard error.
- Figure 6 shows a graph quantifying the pharmacological effect based on MMAE.
- the relative axonal mass is a standardized value, with the result when a polyclonal anti-SYNGR3 intravesicular domain antibody and MMAE conjugate was used (in the figure, "Polyclonal Ab") taken as 100%.
- the dashed line indicates the position where the relative axonal mass is 100%, and "Monoclonal Ab” shows the results for the SYNGR3 antibody group to which a conjugate of the monoclonal antibody of the present invention and MMAE was introduced.
- the error bars indicate the standard error, and "***" indicates p ⁇ 0.001.
- Figure 7 is a graph showing the quantification by ELISA of the binding affinity to synaptogyrin 3 when 100 ng/mL anti-SYNGR3 antibody was used.
- the relative antibody amount is a standardized value assuming the result when polyclonal anti-SYNGR3 antibody was used ("Polyclonal Ab" in the figure) as 1.0.
- the dashed line indicates the position where the relative antibody amount is 1.0, and "Monoclonal Ab” shows the result when the monoclonal antibody of the present invention was used.
- the error bars indicate the standard deviation, and "***" indicates p ⁇ 0.0001.
- Figure 8 is a graph showing the binding affinity to synaptogyrin 3 quantified by ELISA when 500 ng/mL anti-SYNGR3 antibody was used.
- the relative antibody amount is a standardized value assuming the result when polyclonal anti-SYNGR3 antibody was used ("Polyclonal Ab" in the figure) as 1.0.
- the dashed line indicates the position where the relative antibody amount is 1.0, and "Monoclonal Ab” shows the result when the monoclonal antibody of the present invention was used.
- the error bars indicate the standard deviation, and "***" indicates p ⁇ 0.0001.
- the present invention provides a monoclonal antibody (referred to as the "antibody of the present invention") that can bind to the intravesicular domain of synaptogyrin 3 and can be used to target a substance of interest to a motor neuron.
- antibody of the present invention a monoclonal antibody that can bind to the intravesicular domain of synaptogyrin 3 and can be used to target a substance of interest to a motor neuron.
- SYNGR3 synaptogyrin 3
- SYNGR3 synaptogyrin 3
- Synaptogyrin 3 has three cytoplasmic domains and two intravesicular domains separated by four transmembrane domains.
- the vesicular domain of synaptogyrin 3 is exposed to the lumen of synaptic vesicles.
- synaptic vesicles fuse with the plasma membrane as the intracellular calcium ion concentration increases, the lumen of the synaptic vesicle connects with the extracellular space, and the vesicular domain of synaptogyrin 3 is temporarily exposed to the outside of the cell.
- the plasma membrane portion containing synaptogyrin 3 is then retrieved into the cell as a synaptic vesicle membrane by endocytosis and reused as synaptic vesicles.
- the vesicular domain of synaptogyrin 3 is again exposed to the lumen of the synaptic vesicle.
- an exemplary human synaptogyrin 3 is a protein consisting of 229 amino acids whose amino acid sequence is represented by SEQ ID NO: 9.
- the positions of each domain are as follows: the first cytoplasmic domain is the region represented by the amino acid sequence of positions 1 to 29, the first transmembrane domain is the region represented by the amino acid sequence of positions 30 to 50, the first intravesicular domain is the region represented by the amino acid sequence of positions 51 to 69, the second transmembrane domain is the region represented by the amino acid sequence of positions 70 to 90, the second cytoplasmic domain is the region represented by the amino acid sequence of positions 91 to 104, the third transmembrane domain is the region represented by the amino acid sequence of positions 105 to 125, the second intravesicular domain is the region represented by the amino acid sequence of positions 126 to 147, the fourth transmembrane domain is the region represented by the amino acid sequence of positions 148 to 168, and the third cytoplasmic domain
- Sequence information for synaptogyrin 3 from mice and other organisms can be easily obtained from publicly known databases such as the NCBI database.
- the term "synaptogyrin 3 vesicular domain” refers to all or part of either of the two vesicular domains of synaptogyrin 3.
- the entire intravesicular domain includes, for example, the first intravesicular domain represented by the amino acid sequence of positions 51 to 69 in SEQ ID NO: 9 (SEQ ID NO: 10) and/or the second intravesicular domain represented by the amino acid sequence of positions 126 to 147 in SEQ ID NO: 11.
- the second intravesicular domain represented by SEQ ID NO: 11 is preferred.
- a portion of the intravesicular domain includes, for example, a region represented by a partial sequence of any length in SEQ ID NO: 10 and/or 11.
- a preferred portion of the intravesicular domain includes, for example, the amino acid sequence represented by the amino acid sequence of positions 127 to 144 in SEQ ID NO: 9 (SEQ ID NO: 12).
- antigen peptide used to obtain the antibody a peptide containing additional amino acids in the amino acid sequence of the intravesicular domain or a part of it can be used, so long as it is possible to obtain an antibody that can bind to the intravesicular domain.
- antigen peptides that can be used in this way include peptides having the amino acid sequence shown in SEQ ID NO: 13 or 14.
- the antibody of the present invention is a monoclonal antibody (referred to as “the antibody of the present invention") that can specifically recognize and bind to the intravesicular domain of synaptogyrin 3.
- the antibody of the present invention binds to the intravesicular domain of synaptogyrin 3, but preferably does not bind to the intravesicular domain of other membrane proteins belonging to the synaptogyrin family (e.g., synaptogyrin 1 and synaptogyrin 4).
- the antibody of the present invention binds to the intravesicular domain of human synaptogyrin 3, but may or may not be able to bind to the intravesicular domain of synaptogyrin 3 derived from other animals (e.g., mouse).
- binding and “specific binding” are not particularly limited, but may mean that the binding between the antigen and the antibody has a binding affinity with a KD value of 10 -8 M or less.
- the antibody of the present invention has the above-mentioned binding affinity to the intravesicular domain of synaptogyrin 3. It is preferable that the antibody of the present invention does not have the above-mentioned binding affinity to other antigens including other proteins (for example, intravesicular domains of other membrane proteins belonging to the synaptotagmin family such as synaptotagmin 1, etc.). Alternatively, the binding affinity may be determined by a large binding rate constant.
- the specific value of the binding rate constant in this case is not particularly limited, but may be, for example, 10 3 /Ms or more, 10 4 /Ms or more, 10 5 /Ms or more (10 ⁇ 10 5 /Ms or more, 15 ⁇ 10 5 /Ms or more, 16 ⁇ 10 5 /Ms or more, etc.).
- Methods for measuring the binding affinity and binding rate constant include, for example, surface plasmon resonance and biolayer interference, and for example, biolayer interference can be preferably used.
- telomere binding to a target substance can mean that the binding to a target substance, for example, the intravesicular domain of synaptogyrin 3, is at least two-fold, three-fold, or four-fold stronger than the binding to a substance other than the target substance, for example, when detected by a general binding assay using an excess amount of the target substance. It can also mean that, when binding is detected by fluorescent labeling as described above, the S/N ratio is 2 or more, 3 or more, or 4 or more.
- the antibody of the present invention may be any of non-human antibodies of mouse, rabbit, goat, etc., chimeric antibodies, humanized antibodies, and human antibodies, but when used to treat conditions and diseases in humans, it is preferable, although not limited thereto, that it be a humanized antibody or a human antibody. Therefore, an antibody that can be suitably used in the present invention may be an antibody that contains a framework region (FR) of a human antibody and a complementarity determining region (CDR) that can provide high binding affinity to the intravesicular domain of synaptogyrin 3.
- FR framework region
- CDR complementarity determining region
- the antibody of the present invention may be an IgG antibody molecule, or an antigen-binding fragment or derivative thereof.
- the antibody may be a complete antibody, a Fab, Fab', or F(ab') 2 fragment, or a single-chain antibody (scFv) fragment in which the heavy chain variable region (VH) and the light chain variable region (VL) are linked via a linker, such as scFv-Fc, sc(Fv) 2 , Fv, or a diabody.
- scFv, scFv-Fc, and sc(Fv) 2 are synthetic polypeptides in which variable regions are linked by a linker.
- the linker may be any linker commonly used in the art, and is not particularly limited.
- a peptide linker consisting of 5 to 25, preferably 10 to 20, amino acid residues, such as a GS linker, can be suitably used.
- the antibodies of the present invention further include derivatives that would be understood by those skilled in the art to the extent that they do not affect antigen-binding ability, such as derivatives modified to facilitate antibody purification or to increase stability, and conjugates bound to drugs such as anticancer drugs.
- derivatives modified to facilitate antibody purification or to increase stability
- conjugates bound to drugs such as anticancer drugs.
- fragments and derivatives that retain the ability to bind to the intravesicular domain of synaptogyrin 3 are intended to be included in the term "antibody” for the sake of convenience, unless the context is inconsistent.
- the antibody of the present invention can also be synthesized as a multimer, such as a dimer, trimer, or tetramer.
- the antibody of the present invention can be a bispecific antibody having a first specificity that binds to the intravesicular domain of synaptogyrin 3 and a second specificity that binds to another antigen.
- the second specificity can be, for example, for another antigen that can be expressed by the target cell, or for a physiologically active substance and/or a labeling substance to be delivered, and can be appropriately selected depending on the purpose.
- the second specificity is for another antigen that can be expressed by the target cell, i.e., the motor neuron
- the use of the bispecific antibody provides more specific delivery to the target cell.
- the second specificity is for a physiologically active substance and/or a labeling substance
- the substance can be delivered to the target site as a complex by administering it together with the bispecific antibody.
- antibodies that bind to the intravesicular domain of synaptogyrin 3 of the present invention include the following antibodies.
- An antibody comprising a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
- amino acid sequences of other regions of the present invention are not particularly limited.
- the binding characteristics of an antibody are basically determined by the CDRs of the heavy and light chains. Therefore, the amino acid sequence of the antibody of the present invention may be modified, but it is preferable that the modification is within the framework region or constant region from the viewpoint of having little effect on the binding characteristics. In addition, it is preferable that the modification does not significantly change the three-dimensional structure of the antibody.
- sequence identity refers to the percentage of matching amino acid residues out of the total number of amino acid residues in the amino acid sequences of two polypeptides being compared, when the sequences are aligned by inserting appropriate gaps into one or both sequences as necessary to maximize the number of matching amino acid residues. Sequence identity can be calculated using methods and software well known in the art.
- sequence identity of the amino acid sequence in the antibody of the present invention is not particularly limited as long as it is 90% or more.
- the amino acid sequence of the heavy chain variable region consists of an amino acid sequence that has a sequence identity of 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more to SEQ ID NO: 1.
- amino acid sequence of the light chain variable region of the antibody of the present invention consists of an amino acid sequence that has a sequence identity of 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more to SEQ ID NO: 2.
- “several” can be 5 or less, for example 2 or more, 3 or more, 4 or more, or 5.
- the antibody of the present invention can have 5 or less (e.g., 2 to 3) amino acid modifications in the heavy chain variable region and/or the light chain variable region of the antibody (b) above.
- Antibodies that are functionally equivalent to a certain antibody can be prepared by techniques well known in the art, such as site-directed mutagenesis.
- amino acid substitution refers to substitutions between the 20 types of amino acids that make up natural proteins.
- the amino acid substitutions can be, but are not limited to, conservative substitutions.
- Conservative substitutions refer to substitutions within a conservative amino acid group that has similar properties such as charge, side chain, polarity, and aromaticity.
- substitutions include substitutions within the uncharged polar amino acid group with low polarity side chains (Gly, Asn, Gln, Ser, Thr, Cys, Tyr), branched-chain amino acids (Leu, Val, Ile), neutral amino acids (Gly, Ile, Val, Leu, Ala, Met, Pro), neutral amino acids with hydrophilic side chains (Asn, Gln, Thr, Ser, Tyr, Cys), acidic amino acids (Asp, Glu), basic amino acids (Arg, Lys, His), and aromatic amino acids (Phe, Tyr, Trp).
- examples of the antibodies of the present invention that bind to the intravesicular domain of synaptogyrin 3 include the following antibodies: (c) An antibody having a heavy chain variable region consisting of the amino acid sequence shown in SEQ ID NO: 1 and a light chain variable region consisting of the amino acid sequence shown in SEQ ID NO: 2.
- the method for obtaining the antibody of the present invention is not particularly limited.
- a non-human mammal can be immunized with the intravesicular domain of synaptogyrin 3 identified as an antigen, and a monoclonal antibody can be obtained by a known method.
- the antibody of the present invention can also be obtained synthetically using genetic engineering techniques or chemical synthesis means based on the amino acid sequence information of the antibody of the present invention whose activity has been demonstrated, or the base sequence information of the polynucleotide encoding the antibody.
- polynucleotides encoding the heavy and light chains can be introduced into an appropriate host cell and expressed to obtain the antibody as a recombinant protein.
- the polynucleotide may be either DNA or RNA, and any method used in the art can be used as appropriate for introducing the polynucleotide into the host cell.
- a vector for introducing the polynucleotide into the host cell a virus vector, a plasmid vector, a phage vector, etc. can be used as appropriate.
- bacteria such as E. coli, yeast, insect cells, animal cells, etc. can be used.
- the polynucleotides encoding the heavy and light chains can be introduced into separate vectors, or can be linked to the same vector and introduced.
- the targeting effect of the antibody of the present invention can be confirmed by comparing the amount of axons when a conjugate of MMAE with a polyclonal goat anti-SYNGR3 intravesicular domain antibody (Synaptogyrin 3 Rabbit anti-Human Polyclonal Antibody; catalog number LS-C55548-100; antibody of LS Bio, etc.) is used with the amount of axons when a conjugate of the antibody of the present invention is used.
- the antibody of the present invention for example, the amount of axons is reduced compared to when a polyclonal antibody is used. For example, a statistically significant reduction would be sufficient; specifically, the average axon volume could be 95% or less, or 90% or less, of the amount observed when a polyclonal antibody was used.
- the present invention also provides a nucleic acid molecule having a base sequence encoding the antibody of the present invention.
- the nucleic acid molecule of the present invention may be a nucleic acid molecule encoding any one or more of the antibodies of the present invention.
- the base sequence of such a nucleic acid molecule is not particularly limited. Examples include a codon-optimized base sequence and a base sequence with an initiation codon (ATG) added to the 5'-end.
- the nucleic acid molecule of the present invention may be DNA, RNA, or a combination thereof, and may contain natural nucleotides as well as modified nucleotides, non-natural nucleotides, etc., as appropriate.
- the nucleic acid molecule of the present invention may contain any other components in addition to the nucleic acid encoding the antibody of the present invention.
- it may be a gene expression vector that further contains a promoter and is capable of expressing the antibody of the present invention in a cell.
- the term "gene expression vector” refers to a vector that contains genes or gene fragments (hereinafter referred to as "genes, etc.") in an expressible state and includes an expression unit that can control the expression of the genes, etc.
- the gene expression vector may be a plasmid vector, a virus vector, or a phage vector.
- it may be a plasmid vector that is easy to manipulate for genetic recombination, or a virus vector that can easily introduce genes into immune cells.
- the vector in the present invention may additionally contain a marker gene (selection marker), an enhancer, a terminator, a replication origin, a polyA signal, etc., as necessary.
- expressible state refers to the placement of a gene to be expressed downstream of a promoter under the control of the promoter.
- the plasmid vector may be a commercially available expression vector for mammalian cells, such as Promega's pCI vector, pSI vector, or pcDNA3 vector, or a shuttle vector capable of replicating between mammalian cells and bacteria such as E. coli.
- a retroviral vector including oncoretroviral vectors, lentiviral vectors, and pseudotype vectors
- an adenoviral vector including oncoretroviral vectors, lentiviral vectors, and pseudotype vectors
- an adenoviral vector including oncoretroviral vectors, lentiviral vectors, and pseudotype vectors
- an adenoviral vector including oncoretroviral vectors, lentiviral vectors, and pseudotype vectors
- AAV adeno-associated virus
- simian virus vector a simian virus vector
- a vaccinia virus vector a Sendai virus vector
- an Epstein-Barr virus (EBV) vector Epstein-Barr virus
- HSV vector an HSV vector
- a viral vector lacking replication ability so as not to self-replicate within an infected cell may also be used.
- a "promoter” refers to a gene expression regulatory region that can control the expression of a gene, etc., located downstream (3' end side) in a cell into which a gene expression vector has been introduced. Promoters can be classified into ubiquitous promoters (systemic promoters) and site-specific promoters based on the location where the gene, etc. under their expression control is expressed.
- a ubiquitous promoter is a promoter that controls the expression of a target gene, etc. (target gene, etc.) in all cells, i.e., the entire host individual.
- a site-specific promoter is a promoter that controls the expression of a target gene, etc. only in a specific cell or tissue.
- the promoter contained in the gene expression vector of the present invention may be either a ubiquitous promoter or a site-specific promoter, but it is preferable that it can induce expression in a host cell.
- Promoters are also classified into constitutively active promoters, expression-inducible promoters, and time-specifically active promoters based on the time of expression.
- Constitutively active promoters can constitutively express a target gene, etc. in a cell.
- Expression-inducible promoters can induce the expression of a target gene, etc. in a cell at any time.
- Time-specifically active promoters can induce the expression of a target gene, etc. in a cell only at a specific time during the developmental stage. Any of these promoters can be considered to be overexpression promoters, since they can cause excessive expression of a target gene in a host cell.
- the promoter contained in the gene expression vector of the present invention is preferably a constitutively active promoter, which allows for long-term persistence of the therapeutic effect.
- the promoter in the gene expression vector of this embodiment is a promoter that can induce the expression of a nucleic acid encoding an antibody of the present invention in a host cell.
- a promoter that can induce the expression of a nucleic acid encoding an antibody of the present invention in a host cell.
- Specific examples include a CMV promoter (CMV-IE promoter), an SV40 early promoter, an RSV promoter, an EF1 ⁇ promoter, an Ub promoter, and a 5' LTR promoter.
- CMV-IE promoter CMV promoter
- the nucleic acid encoding an antibody of the present invention can be placed downstream of the 5' LTR promoter to induce its gene expression.
- the present invention provides a cell capable of expressing the antibody of the present invention.
- the type of the host cell of the present invention is not particularly limited.
- the organism from which the cell originates is not particularly limited, but for example, cells derived from vertebrates described later in the targeting agent can be used as the cell of the present invention.
- the type of the host cell is not particularly limited.
- it may be an ectodermal cell, a mesodermal cell, an endodermal cell, or a combination thereof.
- it may be an epithelial tissue, a connective tissue, a cartilage tissue, a bone tissue, a blood tissue (including lymphatic tissue), a muscle tissue, a nerve tissue, or a combination thereof.
- the cell of the present invention may be a cell that does not have differentiation potential or a cell that has differentiation potential (for example, a multipotent cell, a pluripotent cell, etc.).
- a cell that has differentiation potential for example, a multipotent cell, a pluripotent cell, etc.
- Specific examples of cells that have differentiation potential include mesenchymal stem cells, hematopoietic stem cells, various cancer cell lines, neural stem cells, iPS cells, and ES cells.
- pluripotent stem cells used in the present invention refers to cells that have the ability to self-replicate, can be cultured in vitro, and have the multipotency to differentiate into cells that make up an individual.
- Specific examples include embryonic stem cells (ES cells), pluripotent stem cells (GS cells) derived from fetal primordial germ cells, and induced pluripotent stem cells (iPS cells) derived from somatic cells, but the cells preferably used in this method are human-derived iPS cells or ES cells.
- ES cells are often obtained from fertilized eggs, but they can also be obtained from sources other than fertilized eggs, such as adipose tissue, placenta, and testicular cells, and all ES cells are within the scope of the present invention.
- sources other than fertilized eggs such as adipose tissue, placenta, and testicular cells, and all ES cells are within the scope of the present invention.
- Methods for producing ES cells from sources other than fertilized eggs have been reported (e.g., WO2003/046141), and these reports can be used with appropriate reference.
- iPS cells are artificial stem cells derived from somatic cells, and can be produced by introducing specific reprogramming factors into somatic cells in the form of nucleic acids or proteins. They exhibit properties similar to those of ES cells (e.g., pluripotency and proliferation ability based on self-renewal).
- genes contained in the reprogramming factors include Oct3/4, Sox2, Sox1, Sox3, Sox15, Sox17, Klf4, Klf2, c-Myc, N-Myc, L-Myc, Nanog, Lin28, Fbx15, ERas, ECAT15-2, Tcl1, beta-catenin, Lin28b, Sall1, Sall4, Esrrb, Nr5a2, Tbx3, Glis1, or combinations thereof.
- the iPS cells that can be used in this method are preferably human-derived iPS cells, for example, human fibroblast-derived iPS cells.
- the cell of the present invention is capable of expressing the antibody of the present invention.
- the cell of the present invention comprises a nucleic acid molecule of the present invention.
- the cell of the present invention can comprise multiple copies of the nucleic acid molecule of the present invention.
- the present invention also relates to a targeting agent for motor neurons (referred to as the "targeting agent of the present invention"), which comprises an antibody of the present invention capable of binding to the intravesicular domain of synaptogyrin 3.
- the term "targeting agent” refers to an agent for delivering a specific substance to a target.
- the targeting agent is used to transport a labeled substance and/or a physiologically active substance (referred to as a "desired substance") to a motor neuron, particularly to a motor neuron synapse.
- the substance is taken up into the cell, particularly into synaptic vesicles, following the retrieval of synaptic vesicles by endocytosis, and is delivered to the cell body.
- the targeting agent of the present invention allows the transported substance to exert its physiological activity in the cytoplasm.
- the targeting agent of the present invention may be taken up into synaptic vesicles, and the transported desired substance may permeate the synaptic vesicle membrane and migrate into the cytoplasm.
- the targeting agent may migrate into the cytoplasm and act on a biological substance in the nucleus, a desired biological substance in the cytoplasm, or a biological substance on the cytoplasmic membrane.
- the biological substance on which the physiologically active substance acts is not particularly limited as long as it is a substance that can be present on a cell membrane or inside a cell, and may be, for example, any of polymeric compounds such as proteins and nucleic acids, low molecular compounds such as lipids, sugars, amino acids, and nucleotides, ions such as metal ions, or atoms, etc.
- biological substances in the nucleus include DNA, RNA (mRNA, siRNA, miRNA, etc.), transcription factors, and nuclear receptors
- biological substances in the cytoplasm include the above-mentioned nucleic acids as well as the cytoskeleton, enzymes, and metal ions.
- biological substances on the cell membrane include lipids in the cell membrane, receptors on the cell membrane, enzymes conjugated with receptors, and cell adhesion factors.
- motor neurons refers to a group of nerve cells that transmit stimuli from the central nervous system to skeletal muscles, which are effector organs.
- Motor neurons generally include primary motor neurons, which are central nerve cells, and secondary motor neurons, which are peripheral nerve cells, but the motor neurons in this specification are secondary motor neurons.
- the term "secondary motor neuron” refers to a motor neuron that has its cell body in the anterior horn of the spinal cord or the brain stem and extends its axon to the junction with skeletal muscle.
- the motor neuron includes, for example, alpha motor neuron, beta motor neuron, and gamma motor neuron.
- some cranial nerves such as the oculomotor nerve, trochlear nerve, abducens nerve, facial nerve, and hypoglossal nerve are also included.
- the motor neuron usually secretes acetylcholine as a neurotransmitter and is classified as a cholinergic neuron. However, it may be a motor neuron that secretes a neurotransmitter other than acetylcholine.
- the muscle cell to which the motor neuron projects is a skeletal muscle cell.
- Skeletal muscle cells refers to cells that constitute striated muscles that move the skeleton or cells that have the same phenotype. Skeletal muscle cells in this specification broadly include muscle cells attached to bones and other muscle cells contained in skeletal muscles, such as muscle spindles.
- skeletal muscle There is no particular limitation on the type of skeletal muscle, but examples include the diaphragm, vastus lateralis, vastus medialis, rectus femoris, vastus intermedius, biceps brachii, tibialis anterior, tibialis posterior, gastrocnemius, soleus, deltoid, latissimus dorsi, sternocleidomastoid, intercostal muscles, eye muscles, facial muscles, tongue muscles, and stapedius muscles.
- Skeletal muscle cells in this specification also include cultured skeletal muscle cells, such as cells differentiated in vitro from artificial stem cells (such as iPS cells and ES cells) and/or natural stem cells (such as mesenchymal stem cells and skeletal muscle stem cells).
- artificial stem cells such as iPS cells and ES cells
- natural stem cells such as mesenchymal stem cells and skeletal muscle stem cells
- the cells may be cells derived from vertebrates. Vertebrates include fish, reptiles, amphibians, birds, and mammals. Specific examples of mammals include primates (e.g., humans).
- the cells may also be cells derived from livestock (chickens, horses, cows, sheep, goats, pigs, etc.), pets (tropical fish, lizards, dogs, cats, rabbits, etc.), and laboratory animals (frogs, mice, rats, monkeys, etc.).
- the cells do not have to be derived from one type of tissue, individual, or animal species, but may be a mixture of multiple types of cells. Furthermore, there are no particular limitations on the health of the tissue and individual from which the cells are derived.
- the targeting agent of the present invention allows a desired substance to be targeted to a motor neuron (e.g., the axon terminal, axon, axon hillock, cell body, dendrites, etc. of a motor neuron) via the motor neuron synapse.
- a motor neuron e.g., the axon terminal, axon, axon hillock, cell body, dendrites, etc. of a motor neuron
- syne refers to a junction that includes a gap, which is formed between the axon terminal of a nerve cell and the dendrite of another nerve cell (in the case of the central nervous system) or the cells of skeletal muscles, organs, etc. (in the case of the peripheral nervous system).
- a synapse may be a chemical synapse, such as an excitatory synapse, an inhibitory synapse, etc.
- a synapse may be a synapse formed between a nerve cell and another nerve cell (e.g., a synapse formed between an axon of a nerve cell and a dendrite of another nerve cell), or a synapse formed between a nerve cell and a cell of another type (such as a muscle cell), but is preferably a synapse formed by a presynapse on a nerve cell and a postsynapse on a skeletal muscle cell (also called a "neuromuscular junction").
- presynapse refers to the enlarged portion formed at the axon terminal of a nerve cell in a synapse
- postsynapse refers to the portion facing the presynapse in a dendrite of another nerve cell or in another cell such as a skeletal muscle or organ.
- synaptic cleft refers to the space between the presynapse and the postsynapse. In a synapse, neurotransmitters accumulated in synaptic vesicles present in the presynapse are released into the synaptic cleft and bind to receptors present in the postsynapse, thereby transmitting a signal.
- synaptic vesicles refer to secretory vesicles present in the cytoplasm of presynaptic neurons.
- synaptic vesicles include not only vesicles that contain neurotransmitters and fuse with the cell membrane in response to a stimulus to release the neurotransmitter into the synaptic cleft, but also vesicles that are retrieved into the neuron by endocytosis (including bulk endocytosis) after the release of the neurotransmitter.
- the targeting agent of the present invention binds to the intravesicular domain of synaptogyrin 3 exposed on the cell membrane in the synaptic cleft, is taken up into synaptic vesicles by endocytosis, and can be delivered to the cell body of a motor neuron, etc. Therefore, the targeting agent of the present invention can target a desired substance (a labeled substance and/or a physiologically active substance) to the inside of a motor neuron, particularly to the cell body of the motor neuron.
- a desired substance a labeled substance and/or a physiologically active substance
- the targeting agent of the present invention when administered into cerebrospinal fluid, the desired substance (a labeled substance and/or a physiologically active substance) can be targeted to the inside of a neuron expressing synaptogyrin 3 on the cell membrane in the central nervous system, particularly to the cell body.
- the targeting agent of the present invention may further contain a desired substance (a labeling substance and/or a biologically active substance) in addition to the antibody of the present invention capable of binding to the intravesicular domain of synaptogyrin 3.
- a desired substance a labeling substance and/or a biologically active substance
- labeling substance refers to a substance that emits a signal that can detect its presence.
- labeling substances include luminescent labeling substances that emit light under specific conditions, such as fluorescent molecules and chemiluminescent substances, sound-emitting labeling substances that emit sound waves, such as photoacoustic effect probes, and radioactive labeling substances.
- fluorescent molecules include, but are not limited to, fluorescent proteins, fluorescein and its derivatives, pyrene and its derivatives, and quantum dots.
- chemiluminescent substances include enzymes such as peroxidase (HRP) and alkaline phosphatase (ALP).
- radioactive labeling substances examples include reagents containing 14 C, 3 H, 125 I, and the like.
- the photoacoustic effect refers to a phenomenon in which adiabatic expansion accompanying light absorption generates a thermoelastic wave, and this thermoelastic wave can be detected as an acoustic wave.
- photoacoustic effect probes include indocyanine green or its derivatives, curcumin derivatives, and choline derivatives.
- biologically active substance refers to a substance that can directly or indirectly exert a physiological effect on a living organism or a cell.
- examples include low molecular weight compounds that can exert a physiological effect on a target motor neuron, functional medium molecules such as peptides and aptamers, and polymeric compounds including biopolymers such as proteins such as antibodies and enzymes, and nucleic acids such as DNA and RNA.
- drugs or prodrugs such as synapse formation promoters, synapse maintenance agents, muscle strengthening agents, or nerve cell function modifiers can be used as bioactive substances.
- physiological effect refers to an effect that brings about quantitative and/or qualitative changes in biological molecules such as proteins, DNA, and RNA.
- a physiological effect for example, the functions and properties of living organisms, organs, tissues, cells, etc. may change. For example, effects such as promotion or inhibition of synapse formation, improvement or prevention of decline in neuronal function, or improvement or prevention of hyperactivity of nerve cells may be obtained.
- the targeting agent of the present invention contains a desired substance (a labeling substance and/or a physiologically active substance)
- the antibody of the present invention and the desired substance are not covalently linked (e.g., noncovalently linked) or are covalently linked.
- the antibody of the present invention and the desired substance are covalently linked, the antibody of the present invention capable of binding to the intravesicular domain of synaptogyrin 3 and the desired substance form a conjugate (referred to as the "conjugate of the present invention").
- conjugate refers to a substance in which two or more molecules are covalently linked.
- the conjugate of the present invention is a substance in which the antibody of the present invention is linked to a desired substance (a labeling substance and/or a biologically active substance).
- the covalent and non-covalent bonds between the antibody and the desired substance in the targeting agent of the present invention are not particularly limited, so long as the antibody of the present invention and the desired substance can reach the vicinity of the motor neuron in a linked state.
- synapse formation promoter refers to a drug that has the effect of promoting the formation of the presynapse and/or postsynapse.
- Promotion of synapse formation includes, for example, enhancing the strength of synapse connections, for example, (i) increasing the surface area and/or volume of the presynapse and/or postsynapse, (ii) increasing and/or qualitatively changing the amount, density, accumulation rate, accumulation frequency, etc. of a protein specifically expressed in the presynapse (e.g., synapsin 1 or synapsin 2, etc.), and (iii) increasing and/or qualitatively changing the amount, density, accumulation rate, accumulation frequency, etc. of a protein specifically expressed in the postsynapse (e.g., LRRTM family proteins).
- synapse maintenance agent refers to a drug that has the effect of suppressing or assisting in the degeneration of the presynapse and/or postsynapse.
- Synapse maintenance includes, for example, suppressing or assisting in the weakening of synaptic junction strength, for example, (i) suppressing or assisting in the reduction of the surface area and/or volume of the presynapse and/or postsynapse, (ii) suppressing or assisting in the reduction and/or qualitative change of the amount, density, accumulation rate, accumulation frequency, etc.
- a protein specifically expressed in the presynapse e.g., synapsin 1 or synapsin 2, etc.
- suppressing or assisting in the reduction and/or qualitative change of the amount, density, accumulation rate, accumulation frequency, etc. of a protein specifically expressed in the postsynapse e.g., LRRTM family proteins.
- muscle-enhancing agent refers to a drug that has the effect of enhancing muscle or inhibiting weakening, or the effect of promoting such enhancement.
- the type of muscle enhancement is not particularly limited as long as the muscle function is enhanced, but examples include an increase in muscle surface area and/or volume, an increase and/or qualitative change in the number, density, etc. of each element that constitutes muscle, such as muscle bundles, muscle fibers, myofibrils, sarcomeres, muscle cells, and/or an effect of causing a change in the expression level of a specific protein in the cells that constitute muscle, an effect of increasing muscle mass and/or muscle strength (e.g., skeletal muscle mass or strength), or an effect of inhibiting muscle weakening through these effects.
- muscle-enhancing agent refers to a drug that has the effect of enhancing muscle or inhibiting weakening, or the effect of promoting such enhancement.
- the type of muscle enhancement is not particularly limited as long as the muscle function is enhanced, but examples include an increase in muscle surface area and/or volume,
- synapse formation promoters include, but are not limited to, compounds discovered by the present inventors and disclosed in JP 2022-053535 (e.g., thiamine and its derivatives), and compounds disclosed in JP 2023-028848 (e.g., atropine, busulfan, chromocarb, procainamide, udenafil, propyphenazone, and their derivatives).
- neuron function modifying agent refers to an agent that has the effect of changing or promoting the function exerted by a neuron.
- the modification of a neuron's function is not particularly limited as long as it changes the degree and/or nature of the neuron's function, but includes, for example, changes in the electrophysiological properties of the neuron (such as the properties of conduction and transmission of stimuli), changes in gene expression patterns, and changes in morphological properties (such as the extension, retraction, and branching of neurites, and the formation and retraction of synapses).
- function modifying agents include, but are not limited to, cytoskeleton modifying agents such as AP-1 inhibitors (e.g., compounds disclosed in WO2020/196725 discovered by the present inventors), FUS inhibitors, SOD1 inhibitors, TDP-43 inhibitors (e.g., anacardic acid compounds), KIF1A inhibitors, and other microtubule polymerization inhibitors (including auristatin drugs such as monomethylauristatin E (MMAE), monomethylauristatin F, and auristatin PE).
- AP-1 inhibitors e.g., compounds disclosed in WO2020/196725 discovered by the present inventors
- FUS inhibitors e.g., compounds disclosed in WO2020/196725 discovered by the present inventors
- SOD1 inhibitors e.g., SOD1 inhibitors
- TDP-43 inhibitors e.g., anacardic acid compounds
- KIF1A inhibitors e.g., anacardic acid compounds
- cytoskeleton modifying agent refers to an agent that inhibits and/or promotes one or more selected from the group consisting of the formation, maintenance, degradation, branching, running, and localization of the cytoskeleton.
- the cytoskeleton modifying agent used herein also includes agents that modify the formation, etc. of the cytoskeleton by acting on molecules other than the cytoskeleton.
- the cytoskeleton includes microtubules, intermediate filaments, and actin filaments.
- an agent that inhibits the formation and maintenance of the cytoskeleton specifically, for example, a microtubule polymerization inhibitor, etc., can be used as a cytoskeleton modifying agent.
- the antibody of the present invention and the desired substance may be directly linked by a covalent bond, or they may be indirectly linked via a linker or the like.
- the desired substance preferably has a portion capable of binding to the antibody of the present invention.
- a desired substance that is covalently bound to another antibody capable of binding to the antibody of the present invention can be used.
- the specific configuration of the "another antibody capable of binding to the antibody of the present invention” is as described in the " ⁇ Antibody of the present invention>” section, except that the antigen is the antibody of the present invention and may be a polyclonal antibody.
- the binding between the moiety capable of binding to the antibody of the present invention and the desired substance is similar to the binding in the targeting agent or conjugate of the present invention. Therefore, the moiety capable of binding to the antibody of the present invention and the desired substance may be linked by a non-covalent bond, may be directly linked by a covalent bond, or may be indirectly linked via a linker or the like.
- the targeting agent of the present invention contains a desired substance
- the desired substance can be included in the targeting agent of the present invention in the form of a peptide complex in which the desired substance is non-covalently linked to the antibody of the present invention.
- the site at which the desired substance binds to the antibody of the present invention is not particularly limited, so long as it does not impair the binding of the antibody of the present invention to the intravesicular domain of SYNGR3.
- the desired substance can be bound to a site other than the hypervariable region (HVR) or to the constant region.
- the targeting agent of the present invention may contain multiple desired substances, so long as the functions of each substance are not impaired.
- the targeting agent may contain multiple substances of the same substance, or may contain one or multiple substances that are different from each other.
- Linkers that are suitable for use in the present invention may be any linkers that are suitable for use in the field.
- the structure and chain length of the linker may be appropriately selected within a range that does not impair the function of the resulting conjugate.
- the linker may be configured, for example, so that it can be cleaved after transport to the synapse.
- the linker may also be configured, for example, so that it cannot be cleaved after transport to the synapse.
- the linker may be any linker commonly used in the art, and is not particularly limited.
- a peptide linker consisting of 5 to 25, preferably 10 to 20, amino acid residues, such as a GS linker may be preferably used.
- a cleavable linker such as an acid-labile linker, a photolabile linker, a peptidase-sensitive linker, a dimethyl linker, or a disulfide-containing linker, may also be used.
- the antibody of the present invention contained in the targeting agent of the present invention binds to the intravesicular domain of synaptogyrin 3 that is temporarily exposed on the cell surface due to fusion of synaptic vesicles with the cell membrane, and can be delivered into the cell together with synaptogyrin 3 along with endocytosis of synaptic vesicles. Therefore, it is preferable that the targeting agent or conjugate is designed so that the desired substance is delivered to the synaptic vesicles of the target synapse.
- the characteristics of compounds that can be delivered to synaptic vesicles are well known in the art.
- the average (or median) particle size of the targeting agent or conjugate of the present invention can be, for example, 160 nm or less, 150 nm or less, 140 nm or less, 130 nm or less, 120 nm or less, 110 nm or less, 100 nm or less, or 90 nm or less.
- the average particle diameter (or median) of the conjugate of the present invention can be, for example, 60 nm or less, 55 nm or less, 50 nm or less, 45 nm or less, 40 nm or less, 35 nm or less, 30 nm or less, 25 nm or less, 23 nm or less, 20 nm or less, 18 nm or less, 15 nm or less, 14 nm or less, 13 nm or less, or 12 nm or less.
- the overall particle diameter when the desired substance is bound to the targeting agent can be in the above-mentioned range.
- the particle diameter may be designed to be large for the purpose of inhibiting endocytosis of synaptic vesicles or for the purpose of delivering a substance to the synapse surface or synaptic cleft.
- the particle diameter before separation may be designed to be large.
- the conjugates or targeting agents of the present invention may be configured to pass through the blood-brain barrier, but are not necessarily configured in this way. Typically, the conjugates or targeting agents of the present invention do not pass through the blood-brain barrier, and therefore do not act on the central nervous system, but only on synapses present in the periphery.
- the targeting effect on motor neurons via synapses can be determined, for example, by administering a conjugate or targeting agent of the present invention containing a physiologically active substance to a subject such as a vertebrate (e.g., a non-human mammal, a human, or another vertebrate) and evaluating the physiological effect of the conjugate or targeting agent of the present invention on the subject's neurons.
- the physiological effect can be evaluated, for example, by comparing the degree of physiological effect between a group administered with the conjugate or targeting agent of the present invention and a group not administered with the conjugate or targeting agent of the present invention, and/or by comparing the degree of physiological effect between a group administered with the conjugate or targeting agent of the present invention and a group administered with the physiologically active substance alone.
- presynaptic formation can be induced by co-culturing nerve cells with microbeads having LRRTM molecules (such as the extracellular domain of LRRTM2) immobilized on their surfaces (WO2021/006075).
- test substance has a targeting effect on motor neurons containing synapses can also be determined by examining whether or not the test substance is localized at the presynapse induced by co-culturing neurons with microbeads.
- LRRTM leucine-rich repeat transmembrane neuronal protein family protein
- the LRRTM family is a family of synaptic organizer proteins on the postsynaptic side, and has the activity of inducing the formation of presynapses. In mammals, including humans, four types of LRRTM family proteins have been reported: LRRTM1, LRRTM2, LRRTM3, and LRRTM4. The LRRTM family protein used in the microbeads may be any of these.
- the present invention relates to a conjugate of a desired substance (a labeling substance and/or a physiologically active substance) with the antibody of the present invention capable of binding to the intravesicular domain of synaptogyrin 3.
- the conjugate of the present invention is, for example, taken up into synaptic vesicles of a motor neuron and delivered to the motor neuron.
- the present invention provides a targeting agent (hereinafter referred to as the "visualizing agent of the present invention") that is an agent for visualizing motor neurons or their synapses.
- Visualizing a motor neuron refers to making the entire or a part of a motor neuron detectable.
- the part that is visualized may be random or a predetermined part.
- a synapse can be visualized as the predetermined part.
- the visualization agent of the present invention can be used as a synapse visualization agent.
- “Visualizing a synapse” refers to making a presynapse and/or a postsynapse detectable. Therefore, the visualization agent of the present invention can use any detectable labeling substance in addition to a labeling substance that can be detected directly by visual inspection.
- the visualization agent of the present invention can visualize the axon terminal, axon, axon hillock, cell body, dendrites, etc. of a motor neuron.
- the visualization agent of the present invention may be used in vivo or in vitro. Detection of the signal of the labeling substance may be performed while the motor neurons are alive or after the motor neurons are fixed. Labeling substances suitable for detection of live motor neurons are known in the art. Examples include fluorescent substances known in the field of in vivo imaging, luminescent substances such as chemo- or bioluminescent substances, sound-emitting substances such as photoacoustic probes, radioactive substances such as radioisotopes, or contrast agents.
- the visualization agent of the present invention may be used for any application, for example, to visualize the number, size or location of motor neurons or synapses (including neuromuscular junctions) or synaptic vesicles, or to visualize tissue in surgery or diagnosis.
- the visualization agent of the present invention may be provided in the form of a kit together with other reagents, such as reagents necessary for detecting the labeling substance contained in the visualization agent of the present invention.
- the labeling substance is an enzyme
- its substrate can be provided together with the visualization agent of the present invention.
- compositions or pharmaceutical composition ⁇ Composition or pharmaceutical composition>
- the present invention further relates to a composition (referred to as “the composition of the present invention") or a pharmaceutical composition (referred to as “the pharmaceutical composition of the present invention") comprising a conjugate or targeting agent of the present invention.
- composition or pharmaceutical composition of the present invention includes the targeting agent of the present invention, which contains a physiologically active substance.
- the composition or pharmaceutical composition of the present invention may also include additives (e.g., carriers (solid or liquid carriers, etc.), excipients, surfactants, binders, disintegrants, lubricants, solubilizing agents, suspending agents, coating agents, colorants, preservatives, buffers, pH adjusters), etc., as necessary.
- the additives can be appropriately selected depending on the dosage form of the composition or pharmaceutical composition.
- composition or pharmaceutical composition of the present invention may be prepared in any dosage form, including, but not limited to, a solid formulation, a liquid formulation, a gel formulation, an aerosol formulation, etc.
- a liquid formulation When the composition or pharmaceutical composition is used as a liquid formulation, it can also be prepared as a dry product intended to be reconstituted with, for example, physiological saline immediately before use.
- excipients include lactose, crystalline cellulose, and starch.
- binders include starch paste, gum arabic paste, and hydroxypropyl cellulose.
- disintegrants include starch, celluloses, and carbonates.
- lubricants include wax and talc.
- composition or pharmaceutical composition of the present invention contains a synapse formation promoter, a synapse maintenance agent, a muscle-enhancing agent, or a nerve cell function modifier as a physiologically active substance, it can improve or prevent a decline in nerve function by promoting synapse formation. Therefore, the composition or pharmaceutical composition of the present invention can be used to improve or prevent a decline in nerve function, such as a decline in nerve function due to nerve damage, a decline in nerve function due to aging, or a decline in nerve function due to disease, or to improve nerve function.
- nerve damage refers to damage at any point on a nerve, and includes damage physically inflicted from outside the body, as well as damage caused by internal factors such as cancer or tumors.
- aging refers to various functional declines, morphological changes, changes in appearance, etc. that occur in individual organisms over time, and the processes involved.
- Frailty and sarcopenia are known as conditions caused by aging.
- Frailty refers to a state in which physical and mental vitality (motor function, cognitive function, etc.) declines with age, and daily life functions are impaired and the body and mind become fragile, influenced by the coexistence of multiple chronic diseases.
- Examples of declines in physical and mental vitality include cognitive impairment, dizziness, eating disorders, swallowing disorders, visual impairment, depression, anemia, hearing loss, delirium, susceptibility to infection, weight loss, and muscle loss.
- chronic diseases include hypertension, heart disease, cerebrovascular disease, diabetes, respiratory disease, and malignant tumors.
- sarcopenia refers to a state in which skeletal muscle mass and muscle strength decrease due to aging or disease.
- Morphological changes such as synaptic detachment and partial or complete axonal detachment from the postsynapse have been observed in the neuromuscular junction of old mice, and it is believed that morphological changes in the neuromuscular junction associated with aging are involved in the decrease in skeletal muscle mass in sarcopenia.
- composition or pharmaceutical composition of the present invention can be used to improve or prevent the decline in neurological function due to aging, particularly in subjects who have or are at high risk of having frailty or sarcopenia.
- diseases include, for example, neurological diseases and neuromuscular diseases.
- the term "neurological disease” refers to a disease caused by a disorder of nerves such as the central nervous system or peripheral nerves, and refers to, for example, one or more diseases selected from the group consisting of Alzheimer's disease, Parkinson's disease, dementia with Lewy bodies, frontotemporal lobar degeneration, progressive supranuclear palsy, corticobasal degeneration, Huntington's disease, dystonia, prion disease, acanthocytic chorea, adrenoleukodystrophy, multiple system atrophy, spinocerebellar degeneration, amyotrophic lateral sclerosis, primary lateral sclerosis, spinal-bulbar muscular atrophy, spinal muscular atrophy, spastic paraplegia, syringomyelia, Charcot-Marie-Tooth disease, frontotemporal dementia, epilepsy, schizophrenia, autism, autism spectrum disorder, and the like.
- the term "neuromuscular disease” refers to a disease caused by a disorder of either the motor nerves, the neuromuscular junction, or muscle cells, and refers to, for example, one or more diseases selected from the group consisting of muscular dystrophy, myopathy, congenital myasthenic syndrome, hereditary periodic paralysis, myasthenia gravis, Lambert-Eaton syndrome, etc.
- Amyotrophic lateral sclerosis is a disease in which primary and secondary motor nerves are selectively and progressively degenerated and lost, and it is known that the initial pathology is detachment of motor nerves from skeletal muscles at the neuromuscular junction. Therefore, the composition or pharmaceutical composition of the present invention, which contains a synapse formation promoter or synapse maintenance agent that can promote the formation of synapses between skeletal muscles and motor nerves or inhibit degeneration, can be used particularly to improve or prevent the decline in neurological function caused by amyotrophic lateral sclerosis.
- the pharmaceutical composition of the present invention can be a pharmaceutical composition for treating amyotrophic lateral sclerosis, a pharmaceutical composition for treating spinal muscular atrophy, etc., which contains the conjugate or targeting agent of the present invention.
- composition or pharmaceutical composition of the present invention containing a muscle-enhancing agent can improve or prevent muscle weakness by strengthening muscles. Therefore, the composition or pharmaceutical composition of the present invention containing a muscle-enhancing agent can be used to improve or prevent muscle weakness, such as weakness after trauma or surgery, weakness due to aging, or weakness due to disease, or to improve muscle function.
- Trauma refers to damage to tissues or organs caused by external factors, and includes, for example, wounds, fractures, sprains, ruptured internal organs, burns, frostbite, etc.
- composition or pharmaceutical composition of the present invention containing a function-modifying agent can improve or prevent the decline or enhancement of nerve cells by modifying the function of motor nerve cells. Therefore, the composition or pharmaceutical composition of the present invention containing a function-modifying agent can also be used to improve or prevent nerve cell hyperactivity, for example, nerve cell hyperactivity due to a disease or condition, or to improve or prevent muscle tension, for example, tremors due to aging, or muscle tension due to trauma or disease.
- nerve cell hyperactivity for example, nerve cell hyperactivity due to a disease or condition
- muscle tension for example, tremors due to aging, or muscle tension due to trauma or disease.
- abnormal involuntary movements e.g., abnormal head movements, tremors, (painful) convulsions, muscle fasciculations, etc.
- abnormalities in walking and mobility e.g., ataxic gait, difficulty walking, etc.
- other coordination disorders e.g., ataxia, etc.
- other conditions related to the nervous system and musculoskeletal system e.g., tetany, abnormal reflexes, posture abnormalities, spasticity, hypertonia, myotonia, hyperactive deep tendon reflexes, dysphagia, etc.
- disease refers to a pathological condition that can be classified by identifiable symptoms or causes in a subject individual, and includes illnesses and disorders.
- condition refers to a pathological condition that includes identifiable symptoms in a subject individual and does not fall under the category of a disease.
- the composition or pharmaceutical composition of the present invention may be configured to exert a pharmacological effect in any cell other than motor neurons in which synaptogyrin 3 can be expressed.
- the specific target disease and condition vary depending on the target cell and are not particularly limited.
- the target may include any of the above-mentioned diseases and conditions, or may include other diseases and/or conditions.
- other diseases and/or conditions include, but are not limited to, infectious diseases (viral, bacterial, etc.), circulatory diseases, blood diseases, digestive system diseases, immune system diseases (inflammatory diseases, autoimmune diseases, etc.), cancer, lifestyle-related diseases, autonomic nervous system disorders, etc.
- composition or pharmaceutical composition of the present invention may contain a plurality of targeting agents of the present invention, and may further contain other active ingredients.
- the other active ingredients are not particularly limited as long as they do not impair the function of the targeting agent contained in the composition or pharmaceutical composition.
- a targeting agent capable of binding to a membrane protein other than synaptogyrin 3 may be contained.
- antibodies include anti-synaptotagmin 2 antibodies (anti-synaptotagmin 2 intravesicular domain (N-terminus) antibodies, etc.).
- a targeting agent containing an antibody capable of binding to a different site e.g., a different epitope or a different intravesicular domain
- the labeling substances and/or physiologically active substances contained in these targeting agents may be different or the same.
- nucleic acid molecules and/or cells of the present invention can be included in the composition or pharmaceutical composition of the present invention.
- the present invention relates to a method for targeting a motor neuron or a synapse.
- the antibody of the present invention that binds to the intravesicular domain of synaptogyrin 3 is brought into contact with a motor neuron, thereby allowing the antibody to be incorporated into the motor neuron (particularly into the synaptic vesicles of the motor neuron) and a desired substance (a labeling substance and/or a physiologically active substance) to be targeted to the motor neuron (e.g., the axon terminal, axon, axon hillock, cell body, dendrites, etc. of the motor neuron).
- a desired substance a labeling substance and/or a physiologically active substance
- the antibody that binds to the intravesicular domain of synaptogyrin 3 is targeted to the motor neuron or synapse.
- the desired substance can be delivered into the motor neuron (e.g., into the cell body or synaptic vesicles of the motor neuron) by directly or indirectly linking the antibody to the conjugate of the present invention via a linker and bringing the conjugate into contact with the motor neuron, or by separately bringing the antibody and a desired substance that can bind to the antibody into contact with the motor neuron.
- a method of targeting a motor neuron or synapse comprising contacting a cell, such as a motor neuron, with an antibody (optionally linked to a substance of interest).
- the present invention relates to a method for targeting a desired substance (a labeled substance and/or a biologically active substance), comprising the steps of contacting a motor neuron with a conjugate or targeting agent of the present invention and delivering the targeting agent to the motor neuron synapse.
- the subject of contact in the present invention is not particularly limited as long as it contains motor neurons.
- contact may be performed only on motor neurons, or on tissue containing cells other than motor neurons.
- the targeting agent of the present invention primarily targets the anterior part of the motor neuron synapse at the neuromuscular junction, it may be targeted at tissue that further contains skeletal muscle cells to which motor neurons project.
- the motor neurons in this method may include motor neurons of a vertebrate (non-human mammals, humans, other vertebrates).
- the motor neurons in this method preferably include human motor neurons.
- human motor neurons will be described using human motor neurons as an example, but the method is not limited to human motor neurons.
- Human motor neurons can be used without any restrictions, regardless of their origin. Examples include, but are not limited to, primary cultures of cells isolated from humans, cells isolated from humans and established as cell lines, and human motor neurons induced to differentiate from human-derived pluripotent stem cells.
- the pluripotent stem cells from which human motor neurons are derived are preferably human-derived pluripotent stem cells.
- the method is an in vitro method. In another embodiment, the method is an ex vivo method. In another embodiment, the method is an in vivo method. When the method is an in vivo method, the subject may be a mammal other than a human.
- the method may further include a step of inducing synapse formation.
- inducing synapse formation refers to causing the formation of a presynapse in the axon of a neuron and/or causing the formation of a postsynapse in the dendrite of another neuron or in cells of skeletal muscle, organs, etc.
- Synapse formation can also be induced by co-culturing a cell that is to form a presynapse with a cell that is to form a postsynapse.
- Synapse formation can also be induced by other methods, for example, by co-culturing a motor neuron with a bead coated with the extracellular domain of LRRTM2.
- the step of inducing synapse formation can be performed simultaneously with or before the step of contacting the motor neuron with the targeting agent of the present invention.
- the step of contacting the targeting agent of the present invention with motor neurons is carried out by contacting the targeting agent of the present invention with a sample containing motor neurons.
- the method of contact is not particularly limited as long as the motor neurons in the sample and the targeting agent can come into contact with each other.
- the targeting agent can be applied by directly sprinkling, spraying, dripping, or applying the targeting agent to the sample, by immersing the sample in the targeting agent, or by a combination thereof.
- the targeting agent may be applied by sprinkling, spraying, dripping, or applying to the carrier.
- the amount to be applied is not particularly limited, and can be set appropriately taking into consideration the number of motor neurons and other conditions.
- the concentration of IgG antibody that can be applied is 0.01 ⁇ g/mL or more, 0.1 ⁇ g/mL or more, 0.2 ⁇ g/mL or more, 0.5 ⁇ g/mL or more, 0.7 ⁇ g/mL or more, 0.9 ⁇ g/mL or more, 1 ⁇ g/mL or more, 2 ⁇ g/mL or more, 5 ⁇ g/mL or more, 7 ⁇ g/mL or more, 9 ⁇ g/mL or more, or 10 ⁇ g/mL or more.
- the step of contacting the targeting agent of the present invention with the test sample is carried out by administering the targeting agent of the present invention to a subject.
- the administration method is not particularly limited, but examples include local administration, enteral administration, and parenteral administration, specifically administration onto the skin, inhalation administration, enema administration, eye drops, ear drops, nasal administration, intravaginal administration, administration by tube feeding, intravenous administration, intraarterial administration, intramuscular administration, intracardiac administration, subcutaneous administration, intraosseous administration, intradermal administration, subarachnoid (cavity) administration, intraperitoneal administration, intravesical administration, transdermal administration, transmucosal administration, epidural administration, intravitreal administration, etc.
- the dosage is not particularly limited and can be set appropriately taking into consideration the target animal species and other conditions.
- the dosage per kg of body weight can be 0.1 mg/kg or more, 0.5 mg/kg or more, 1 mg/kg or more, 2 mg/kg or more, 4 mg/kg or more, or 5 mg/kg or more.
- the targeting agent used in this step does not have to be of one type.
- multiple types of targeting agents can be used together or separately.
- a targeting agent containing a conjugate and a targeting agent not containing a desired substance labeling substance and/or biologically active substance
- the desired substance used does not have to be of one type either.
- multiple types of labeling substances and/or biologically active substances can be used together or separately.
- a labeling substance and a biologically active substance may be used in combination.
- the step of contacting the targeting agent of the present invention with motor neurons can be carried out multiple times.
- the types of targeting agent and cells used, as well as the application method and administration method may be the same or different each time.
- the targeting agent of the present invention When the conjugate or targeting agent of the present invention is brought into contact with a motor neuron, the targeting agent of the present invention is taken up into the motor neuron via synaptic vesicles.
- the motor neuron When the targeting agent of the present invention is brought into contact with a motor neuron, the motor neuron can be activated or its activity can be promoted. By activating the motor neuron or promoting its activity, the efficiency of the uptake of the conjugate or targeting agent of the present invention into the motor neuron can be improved.
- the conjugate or targeting agent of the present invention is taken up into a synaptic vesicle, it is delivered directly to the cell body via the axon by retrograde transport.
- the method for activating motor neurons is not particularly limited, but examples include a method of spontaneously activating motor neurons for a sufficient period of time, and a method of activating motor neurons by artificial stimulation or promoting endocytosis of synaptic vesicles.
- Methods for spontaneous activation include, for example, placing motor neurons in an environment in which they can be activated for a sufficient period of time. Environments in which motor neurons can be activated are well known in the art.
- the time for activating motor neurons is not particularly limited, but for example, when the method is an in vitro method, it can be 1 hour or more, 3 hours or more, 6 hours or more, 12 hours or more, 18 hours or more, or 24 hours or more. Also, when the method is an in vivo method, it can be, for example, 1 hour or more, 3 hours or more, 6 hours or more, 12 hours or more, 18 hours or more, 24 hours or more, 36 hours or more, 48 hours or more, 60 hours or more, 72 hours or more, 100 hours or more, 120 hours or more, 150 hours or more, 168 hours or more, 200 hours or more, or 240 hours or more.
- Methods of activating or promoting motor neuron activity through artificial stimulation include, for example, placing motor neurons in an environment in which they can be actively active for a sufficient period of time.
- the method of activating or promoting motor neuron activity can include providing chemical and/or physical stimulation to the motor neuron.
- Stimuli for activating motor neuron activity are well known in the art.
- Compounds used for chemical stimulation include, for example, potassium ion channel inhibitors such as amiodarone, tetraethylammonium, 4-aminopyridine, barium, dendrotoxin, sodium channel agonists such as batrachotoxin, calcium channel agonists such as Bay K8644, high concentrations of potassium ions or neurotransmitters, or combinations thereof.
- Physical stimuli include, for example, temperature changes.
- the amount of compound added is not particularly limited.
- the compound can be added at a concentration of 1 ⁇ M or more, 10 ⁇ M or more, 50 ⁇ M or more, or 100 ⁇ M or more.
- the duration of the stimulation is not particularly limited, and can be set appropriately taking into consideration the type and intensity of the stimulation and other conditions.
- the stimulation can be applied for 2 minutes or more, 3 minutes or more, 4 minutes or more, 5 minutes or more, 8 minutes or more, 9 minutes or more, 10 minutes or more, 20 minutes or more, 25 minutes or more, 30 minutes or more, or for 1 hour or more.
- the method of activating or promoting the activity of motor neurons can be performed by a method of activating a subject (for example, a method of making a subject exercise or a method of activating brain activity, etc.) or a method of promoting the activity of motor neurons with a chemical substance, etc.
- Compounds that promote the activity of motor neurons include compounds used in the above-mentioned chemical stimulation, and can be administered to the subject.
- Compounds used in the above-mentioned chemical stimulation can be administered at a pharmaceutically acceptable concentration or method.
- the above-mentioned compounds can be administered to the subject so that the compound stimulation does not exert biotoxicity, but if biotoxicity is manifested, it is not necessary to administer the above-mentioned compounds to the subject.
- the activity of motor neurons is promoted, the same degree of effect can usually be expected in a short time compared to the method of causing spontaneous combustion.
- the desired substance and the targeting agent can be contacted with the motor neuron together or separately.
- the timing is not particularly limited as long as the desired substance can bind to the targeting agent.
- the desired substance can be contacted with the motor neuron before or after the targeting agent is contacted with the motor neuron.
- the method of each contact can be selected in accordance with the contact methods described above. For example, the same method or different methods can be used for each contact.
- the method may further include a step of confirming whether or not targeting has been successful.
- the targeting agent used in the method includes a physiologically active substance, it can be determined that targeting has been successful when a physiological effect is observed, for example, as described above in the section on targeting agents.
- the targeting agent used in the method includes a labeling substance, it can be determined that targeting has been successful when a signal is detected, similar to the step of detecting the signal of the labeling substance in the visualization method described below.
- the method can be used as a method for preventing or treating a condition or disease.
- the method may further include a step of allowing the physiological effect to be fully exerted in the subject.
- the physiologically active substance used is a substance that can exert an effect by itself
- the effect can be exerted by placing the subject in an environment where the subject is provided with sufficient nutrition for a period of time sufficient for the physiological effect to be exerted.
- another substance is necessary for the physiologically active substance used to exert an effect, that substance can be administered in addition.
- the present invention relates to a method for preventing or treating a condition or disease, comprising the steps of contacting a targeting agent containing the antibody of the present invention and a physiologically active substance with a motor neuron, and delivering the targeting agent to the motor neuron synapse.
- a targeting agent containing the antibody of the present invention and a physiologically active substance with a motor neuron e.g., a motor neuron
- the contacting step of this method preferably includes administering the targeting agent and/or the pharmaceutical composition to a subject.
- the method of the present invention is a method for improving or preventing a decline in nerve function, such as a decline in nerve function due to nerve damage, a decline in nerve function due to aging, or a decline in nerve function due to a disease, or for improving nerve function.
- the condition or disease is a condition or disease exhibiting a decline in nerve function.
- the condition or disease is a neurological disease or a neuromuscular disease.
- the targeting agent comprises a conjugate of the antibody and the physiologically active substance.
- the duration of this step can be appropriately determined depending on the condition of the subject, the type of physiologically active substance, the dosage, etc. For example, it may be determined based on the period it takes for a physiological effect to be exerted when a physiologically active substance is generally administered, or the physiological effect may be confirmed once or multiple times and continued until the physiological effect is fully exerted.
- the present invention relates to a method for visualizing motor neurons or synapses, the method comprising the steps of contacting a motor neuron with a visualization agent of the present invention, delivering the visualization agent to a motor neuron synapse, and detecting a signal of the labeling substance.
- the motor neurons used for contact are as described above in the method for targeting a labeling substance and/or a biologically active substance.
- the method is an in vitro method. In another embodiment, the method is an ex vivo method. In another embodiment, the method is an in vivo method. When the method is an in vivo method, the subject may be a human or a non-human mammal.
- the step of contacting a motor neuron with the visualization agent of the present invention and the step of delivering the visualization agent to the motor neuron synapse are the same as the step of contacting a motor neuron with the targeting agent of the present invention and the step of delivering the targeting agent to the motor neuron synapse described in the above-mentioned method for targeting a labeling substance and/or a physiologically active substance, except that the visualization agent is used as a targeting agent.
- This method may further include a step of generating a signal from the labeling substance, if necessary.
- the method of generating a signal is not particularly limited. The method of generating a signal and whether or not it is necessary can be determined depending on the type of labeling substance used, etc.
- a signal can be generated in the target by waiting a sufficient time for the visualization agent to be delivered to the target motor neuron synapse, that is, a sufficient time for the visualization agent to reach the target motor neuron synapse and for sufficient endocytosis of synaptic vesicles to occur at the motor neuron synapse.
- This time can be appropriately selected according to the time of the step of delivering the visualization agent to the motor neuron synapse.
- the labeling substance is a chemiluminescent substance
- this can be achieved by waiting a sufficient time for the visualization agent to be delivered to the target motor neuron synapse, as well as adding a substance such as its substrate that is used to generate a signal. This step can be carried out simultaneously with or prior to the step of detecting a signal, which will be described later.
- This method further includes a step of detecting the signal of the detection substance.
- the method used for detection is not particularly limited, and can be appropriately selected depending on conditions such as the type of labeling substance used.
- the labeled substance is a fluorescent substance
- excitation light containing light of the excitation wavelength of the labeled substance can be irradiated onto the motor neuron, and the fluorescent wavelength of the labeled substance can be detected using a detector capable of detecting the fluorescent wavelength of the labeled substance.
- the labeled substance is a chemiluminescent substance, for example, it can be detected using a detector capable of detecting the emission wavelength of the labeled substance.
- the labeled substance is a radioactive labeled substance, it can be detected using a detector capable of detecting the radiation emitted by the labeled substance.
- Detecting the signal of the labeled substance includes detecting the presence, location, or quantity of motor neurons (e.g., synapses, cell bodies, etc.) in a sample that contains motor neurons.
- motor neurons e.g., synapses, cell bodies, etc.
- the method of the present invention may further include a step of determining the presence, location, or amount of the signal of the labeling substance detected in the sample by comparing it with a signal in a standard sample containing the labeling substance, or with a previously prepared reference value.
- a standard sample there are no particular limitations on the standard sample, so long as it is a biological sample that serves as a reference for determining whether or not there is a specific condition or disease. Specific examples include those obtained from healthy individuals, those obtained from the same individual as the sample at a different collection time, and those obtained from individuals known to have a specific condition or disease.
- the standard sample may be, for example, a biological sample derived from the same biological species, individual, tissue, or cell as the sample, or may be a biological sample derived from another biological species, individual, tissue, or cell.
- the reference value there are no particular limitations on the reference value, so long as it is a value that serves as a reference for determining whether or not there is a target condition.
- the reference value can be set, for example, based on the intensity or number of signals generally detected in the standard sample.
- the method of comparison is not particularly limited.
- it can be done by visual inspection, by the magnitude of the numerical values, or by statistical methods.
- the present invention provides a method for administering a desired substance to a subject.
- the substance is in the form of a conjugate between the antibody of the present invention and the desired substance. This allows the desired substance to be delivered to a cell of the subject, for example, a motor neuron.
- the desired substance is a physiologically active substance
- the physiologically active substance can be delivered to a cell such as a motor neuron.
- the method can be used to observe the delivery site of the labeling substance (for example, a motor neuron or its synapse).
- the present invention also provides a conjugate of the antibody and the desired substance for use in this method, or a composition comprising the conjugate.
- the present invention relates to a composition for targeting a motor neuron or its synapse, comprising the antibody of the present invention or a conjugate of the antibody and a desired substance (a labeling substance and/or a physiologically active substance).
- the present invention provides a method for visualizing motor neurons in a subject, the method comprising administering to the subject an effective amount of a conjugate of the antibody of the present invention and a labeling substance.
- the present invention also provides a conjugate of the antibody and the labeling substance for use in the method, or a composition containing the conjugate.
- the present invention provides a method for delivering a physiologically active substance to a motor neuron of a subject, the method comprising administering to the subject an effective amount of a conjugate of the antibody of the present invention and the physiologically active substance.
- the present invention also provides a conjugate of the antibody and the physiologically active substance for use in the method, or a composition containing the conjugate.
- the present invention provides the antibody of the present invention or the conjugate of the present invention for use in any of the above methods.
- the present invention relates to the antibody of the present invention or the conjugate of the present invention for use in a method for preventing or treating a condition or disease.
- the present invention relates to the antibody of the present invention or the conjugate of the present invention for use in a method for targeting a labeling substance and/or a physiologically active substance to a motor neuron or its synapse.
- the present invention provides the above antibody or a conjugate of the above antibody and the above substance for use in the manufacture of a medicament for use in any of the above methods.
- the present invention also provides use of the antibody of the present invention or the conjugate of the present invention in the manufacture of a medicine comprising the antibody and a physiologically active substance.
- Example 1 Preparation of monoclonal antibodies The production of monoclonal antibodies and screening using the B cell cloning method were outsourced to Medical and Biological Laboratories Co., Ltd. The outline of the production and screening procedures is as follows.
- the immunization antigen was bound to a carrier protein to induce an immune response using Ellman's reagent.
- Keyhole limpet hemocyanin (KLH) was used as the carrier protein.
- mice The KLH-conjugated immunization antigen was prepared as a 1.0 mg/mL antigen solution, and four mice were immunized once a week for a total of four times. Serum for antibody titer confirmation and lymph node cells for B cell isolation were collected from the immunized mice.
- the antibody titer of the obtained serum was evaluated by ELISA using the screening antigen peptide synthesized in "1. Synthesis of antigen peptide" as an antigen. As a result, it was confirmed that antibodies capable of binding to the intravesicular domain of human synaptogyrin 3 were present in the collected mouse serum.
- the cells were harvested and stained with anti-B cell marker antibodies and streptavidin (which can bind to the biotin in the screening peptide).
- the stained cells were analyzed using FACS Melody (BD Japan), and cells that were positive for anti-B cell markers and biotin were isolated one by one as B cells that reacted to the antigen.
- variable region sequences were amplified using PCR from the cDNA synthesized from each cell, and agarose electrophoresis was used to confirm that genes of the desired size (VH, VL) had been amplified.
- TMB-US (catalog number: #TMB-US, Moss) was added as a color-developing substrate to the plate to develop the label, and the color reaction was stopped using H 2 PO 4 (catalog number: 167-02166, Fujifilm Wako Pure Chemical Industries, Ltd.).
- HEK293T cells were seeded in a 6-well plate (catalog number: #3820, IWAKI) at a seeding density of 5.0 ⁇ 10 5 cells/well, and an antigen expression vector was introduced.
- the antigen expression vector used was a plasmid vector in which a DNA fragment (SEQ ID NO: 15) having a gene sequence (SEQ ID NO: 16) encoding human SYNGR3 (amino acid sequence represented by SEQ ID NO: 9) was integrated into the multicloning site of pCMV6-AC-Myc-DDK-IRES-GFP-Puro Mammalian Expression Vector plasmid (ORIGENE).
- the gene introduction reagent and medium used were the same as those used in "6. Preparation of antibody-containing supernatant".
- Cell labeling was performed as follows. The prepared antibody-containing supernatant was added to each screening cell. Anti-Mouse IgG (H+L) pAb-PE (catalog number: #IM0855, Beckman Coulter) was used at a 200-fold dilution as the secondary antibody. As a negative control, negative control supernatant was added as the primary antibody, and the same secondary antibody as that used for the antibody-containing supernatant was added.
- amino acid sequences of the variable regions of the monoclonal antibodies of the present invention are shown in Table 2.
- CDRs complementarity determining regions
- 1x Dulbecco's phosphate buffered saline catalog number: D1408, Sigma Aldrich
- Example 2 Evaluation of monoclonal antibodies The antigen-binding properties of the obtained monoclonal antibodies were evaluated.
- the binding characteristics of the antibody were measured using sc-hSYNGR3 as an antigen using the Octet RED96e System (SARTORIUS).
- Biosensor Streptavidin biosensor (catalog number: #18-5019; SARTORIUS); Antibody concentration: 10nM/5nM/2.5nM/1.25nM/0.63nM/0.31nM/0.17nM/0nM; Buffer: PBS containing 0.02% Tween-20 and 0.01% BSA; Pre-immobilization equilibration: Incubate with buffer only for 60 seconds; Immobilization: Incubate with a buffer containing antigen peptide for 300 seconds; Equilibration before antibody addition: Incubate with buffer only for 300 seconds; Antibody binding reaction: Incubate with buffer containing antibody for 300 seconds; Antibody dissociation reaction: Incubate with buffer only for 900 seconds; Both were incubated at 30° C. with an agitation speed of 1000 rpm.
- a polyclonal goat anti-SYNGR3 intravesicular domain antibody was used as a control.
- the antigen was used at a concentration of 0.02 ⁇ g/mL.
- Tables 4 and 5 show the results for the polyclonal goat anti-SYNGR3 intravesicular domain antibody
- Table 5 shows the results for the monoclonal antibody of the present invention.
- the binding rate constant is a value that indicates how easily the binding reaction proceeds, and the higher the affinity, the larger the value.
- Example 3 Verification of pharmacological effects of drug delivery using intravesicular domain antibody Using synaptotagmin 2 (SYT2) as an example of a vesicular membrane protein, we investigated whether drug-based pharmacological effects would be observed when drugs were delivered into motor neurons using an antibody that binds to its intravesicular domain (N-terminus).
- SYT2 synaptotagmin 2
- Cell culture Human iPS-derived motor neurons (40HU-005-2M; iXCells Biotechnologies) were thawed using a Dead Cell Removal Kit (Veritas) according to the kit's protocol. The thawed cells were seeded in a 96-well plate (V-bottom) at a density of 2 ⁇ 104 cells/well and cultured in motor neuron maintenance medium (iXCells Biotechnologies) for 1 week to produce neurospheres. The produced neurospheres were selected based on size and circularity, and those that met these criteria were used in the following experiments.
- a 96-well EZVIEW® culture plate LB (AGC Technoglass Co., Ltd.) was coated with poly-D-lysine and Geltrex® Matrix (Thermo Fisher Scientific Co., Ltd.), and the selected neurospheres were seeded on the plate and cultured for 20 days.
- the above-mentioned motor neuron culture medium was used as the culture medium, and the medium was replaced with the same medium on the second day of culture. Thereafter, the medium was replaced three times a week with neuron medium (Neurobasal plus medium (B27 plus supplement (Thermo Fisher Scientific Co., Ltd.), 20 ng/mL BDNF, 20 ng/mL GDNF, and penicillin-streptomycin added)). All medium changes were performed at 50 ⁇ L/well.
- streptavidin-coated microspheres (Bangs Laboratories, Inc.; polystyrene, average diameter 9.94 ⁇ m) were washed twice with washing buffer (phosphate-buffered saline (PBS), 0.01% bovine serum albumin (BSA), 0.05% TritonX-100) and reacted with biotinylated anti-human IgG (Fc-specific) antibody (Sigma Aldrich; mouse monoclonal) in binding buffer (PBS, 0.01% BSA) to immobilize the biotinylated anti-human IgG (Fc-specific) antibody on the streptavidin-coated microbeads. The resulting beads were washed three times with washing buffer (anti-human IgG Fc antibody beads).
- washing buffer phosphate-buffered saline (PBS), 0.01% bovine serum albumin (BSA), 0.05% TritonX-100
- the anti-human IgGFc antibody beads were suspended in binding buffer, and a fusion protein of the extracellular domain of human LRRTM2 and the Fc portion of human IgG (LRRTM2-Fc; R&D Systems) was added thereto, and LRRTM2-Fc was immobilized on the anti-human IgGFc antibody beads.
- LRRTM2-Fc fusion protein of the extracellular domain of human LRRTM2 and the Fc portion of human IgG
- Presynaptic induction ( Figure 1) After 20 days of culture, LRRTM2 beads were seeded at 0.1 ⁇ g/well on the plate and cultured at 37° C. for 48 hours to induce the formation of presynapses.
- MMAE monomethyl auristatin E
- MMAE MedChemExpress
- Microtubule polymerization inhibitor a microtubule polymerization inhibitor.
- Conjugates of MMAE and antibodies were prepared using the MagicLink TM kit (BroadPharm). The conjugates were prepared according to the manufacturer's protocol.
- the antibodies used were polyclonal rabbit anti-SYT2 N-terminal antibody (Polyclonal rabbit purified antibody SYT2 lumenal domain; catalog number 105 223; Synaptic Systems), or, as a control, normal rabbit IgG antibody (Normal Rabbit IgG; catalog number AB-105-C; R&D Systems).
- conjugate solution After the neuronal medium was warmed at 37°C for 30 minutes, 4-aminopyridine (Sigma Aldrich) was added to the medium to a final concentration of 100 ⁇ M and mixed. Furthermore, the conjugate was added to a final concentration of 1 ⁇ g/mL and mixed. This crude conjugate solution was centrifuged at 200 g for 3 minutes at room temperature, and the supernatant was collected as the conjugate solution.
- 4-aminopyridine Sigma Aldrich
- the solution was collected and washed, and the medium was replaced with neuronal medium that did not contain the conjugate or MMAE. Then, the cells were cultured for another 24 hours to allow the axons to grow.
- Immunocytochemical staining was performed using the added antibody as the primary antibody. After permeabilization and blocking of the cell membrane of the fixed cells with a detergent, primary antibody reaction was performed using mouse anti- ⁇ III tubulin (Tuj1) antibody (catalog number 801202; Biolegend) as an additional primary antibody. Then, secondary antibody reaction was performed using Alexa 555-labeled anti-mouse antibody (catalog number A32727; Thermo Fisher Scientific), and fluorescent images were obtained. Blocking was performed using blocking buffer (PBS + 2% normal goat serum + 1% BSA + 1% fetal bovine serum + 0.02% TritonX-100). The number of samples was as follows: MMAE alone-injected group, 13 samples; control antibody group, 17 samples; polyclonal SYT2 antibody group, 27 samples.
- Fluorescence images were acquired using an inverted live cell (DMi8) microscope fluorescence microscope (Leica) equipped with LAS X software (Leica) at the following excitation wavelength, detection wavelength, exposure time, and detection threshold. A gamma correction value of 1 was used for all images.
- Excitation and detection wavelengths Alexa 555 maximum excitation 555 nm; maximum detection 580 nm; actual detection 595 nm.
- Exposure time and detection threshold Alexa 555 exposure time 50ms; detection threshold 100-3500.
- the result when the control normal rabbit antibody and MMAE conjugate was used was set as 100%, and the normalized value was calculated as the relative axonal volume.
- Brightness was acquired using LAS X software (Leica).
- Figures 2 to 4 are immunocytochemical staining images showing the appearance of axons in the control antibody group, which used a conjugate of a control normal rabbit antibody and MMAE ( Figure 2), and the SYT2 antibody group, which used a conjugate of a polyclonal anti-SYT2 N-terminal antibody ( Figure 3).
- Figure 4 is a graph that quantitatively shows the results.
- MMAE inhibits the polymerization of microtubules, a cytoskeleton important for the growth and maintenance of axons. Therefore, the stronger the effect of MMAE, the more axon growth and maintenance is inhibited, and the more axon mass is expected to decrease.
- Example 4 Verification of pharmacological effects of drug delivery using SYNGR3 antibody
- the pharmacological effects of drugs were examined when drugs were delivered into motor neurons in the same manner as for SYT2.
- the experiment was performed in the same manner as in Example 3, except that a polyclonal goat anti-SYNGR3 intravesicular domain antibody (Synaptogyrin 3 Rabbit anti-Human Polyclonal Antibody; catalog number LS-C55548-100; LS Bio) was used as the anti-SYNGR3 intravesicular domain antibody.
- the experiment was performed with 10 samples per group.
- Figure 5 is a graph quantitatively showing the results when polyclonal goat anti-SYNGR3 intravesicular domain antibody was used.
- MMAE inhibits the polymerization of microtubules, a cytoskeleton important for the growth and maintenance of axons. Therefore, the stronger the effect of MMAE, the more axon growth and maintenance is inhibited, and the more axon mass is expected to decrease.
- Example 5 Verification of pharmacological effects of drug delivery using the monoclonal antibody of the present invention When a drug is delivered into a motor neuron using the monoclonal antibody of the present invention obtained in Example 1, the influence on the pharmacological effect of the drug was examined.
- Example 4 The experiment was conducted in the same manner as in Example 4, except that the monoclonal antibody of the present invention obtained in Example 1 was used as the anti-SYNGR3 intravesicular domain antibody, and a polyclonal goat anti-SYNGR3 intravesicular domain antibody was used instead of the control antibody. The experiment was conducted with six samples per group.
- Figure 6 is a graph quantitatively showing the results when the monoclonal antibody of the present invention was used.
- Example 6 Confirmation of affinity of the monoclonal antibody of the present invention by ELISA method The binding affinity of the monoclonal antibody of the present invention obtained in Example 1 to an antigen was examined by ELISA using cultured cells expressing SYNGR3 on the cell membrane.
- the antigen expression vector used in "7-2. Screening by flow cytometry" in Example 1 was used as the SYNGR3 expression vector.
- HEK293T cells (American Type Culture Collection) were used for transfection as prepared as follows:
- HEK293 cells Purchased HEK293 cells were thawed and resuspended in maintenance medium (DMEM medium (Thermo Fisher Scientific) supplemented with 1% Penicillin-Streptomycin (Thermo Fisher Scientific), 10% Fetal Bovine Serum (Cytiva), and 1% GlutaMAXTM I (Thermo Fisher Scientific)).
- the cell suspension was centrifuged at 130 ⁇ g for 5 minutes at room temperature to remove the supernatant, and 10 mL to 20 mL of maintenance medium was added to suspend the cells. The number of cells was counted and the cells were seeded at a cell density of 2.0 ⁇ 106 cells/dish in a 10 cm dish (Greiner Bio-One).
- the medium was removed from the 10 cm dish and washed with PBS. After washing, 2 mL of 0.1% trypsin (Nacalai Tesque) was added and incubated at room temperature for 1.5 to 2 minutes to detach the cells, which were then collected in a 50 mL tube. The collected cells were dispersed, and the resulting cell suspension was centrifuged at 130 ⁇ g at room temperature for 5 minutes to remove the supernatant, and 10 mL to 20 mL of maintenance medium was added to suspend the cells. The number of cells was counted, and for transfection, the cells were seeded in a 96-well plate (Azone) at a cell density of 1.0 ⁇ 103 cells/well and cultured in maintenance medium until 80 to 90% confluent.
- Azone 96-well plate
- the medium was removed and replaced with 1 mL of medium for transfection. After medium replacement, 100 ⁇ L of the transfection solution was added and incubated at 37° C. in the presence of 5% CO 2 for 24 hours.
- a transfection solution was prepared by mixing 0.5 ⁇ L of a vector solution containing a plasmid vector at a concentration of 1 ⁇ g/ ⁇ L with 100 ⁇ L of Opti-MEM TM I Reduced Serum Medium (Thermo Fisher Scientific).
- the transfection medium used was Opti-MEM TM I Reduced Serum Medium (Thermo Fisher Scientific) supplemented with 1 ⁇ L of Lipofectamine TM 2000 Transfection Reagent (Thermo Fisher Scientific).
- the cells were washed with PBS, and blocking buffer (PBS + 2% normal goat serum + 1% BSA + 1% fetal bovine serum + 0.02% TritonX-100) was added to each well, and blocking was performed at room temperature for 1 hour.
- blocking buffer PBS + 2% normal goat serum + 1% BSA + 1% fetal bovine serum + 0.02% TritonX-100
- Primary antibody solutions were prepared in 1.5 mL tubes so that the concentrations of the primary antibody were 1 ⁇ g/mL, 500 ng/mL, and 100 ng/mL. Measurement medium was used as the solvent, and the primary antibody was a control antibody, a polyclonal goat anti-SYNGR3 intravesicular domain antibody, or a monoclonal antibody of the present invention.
- the medium was removed from the 96-well plate, and 100 ⁇ L/well of a primary antibody solution was added to each well, followed by incubation at 37° C. in the presence of 5% CO 2 for 1 hour.
- the antibody solution was removed and washed with PBS, and the secondary antibody solution was added and incubated at room temperature for 1 hour.
- the secondary antibody solution was prepared using blocking buffer as a solvent.
- Donkey anti-Goat 555 (catalog number #A32814; Thermo Fisher Scientific) was used as the secondary antibody against the polyclonal goat anti-SYNGR3 intravesicular domain antibody, and Goat anti-Mouse 555 (catalog number #A32723; Thermo Fisher Scientific) was used as the secondary antibody against the monoclonal antibody of the present invention.
- DAPI catalog number D1306; Thermo Fisher Scientific was also added to the secondary antibody solution for each staining. After the secondary antibody reaction, the antibody solution was removed and the sections were washed with PBS containing 0.02% TritonX-100.
- Detection and Measurement of Fluorescent Signals Fluorescent signals were measured using Promega GloMax (Promega Corporation) at the following excitation and detection wavelengths.
- DAPI Excitation wavelength 365nm, Emission wavelength 415nm-445nm, Detection wavelength 475nm Alexa 555: Excitation wavelength 525nm, Emission wavelength 580nm-640nm, Detection wavelength 520nm
- the fluorescence intensity of the detected Alexa 488 signal was divided by the fluorescence intensity of DAPI to obtain the relative fluorescence intensity of the anti-SYNGR3 antibody. Furthermore, the relative fluorescence intensity when the control antibody was used was standardized to 1.0 to obtain the relative antibody amount.
- Figure 7 is a graph showing the results when 100 ng/mL of anti-SYNGR3 antibody was used
- Figure 8 is a graph showing the results when 500 ng/mL of anti-SYNGR3 antibody was used.
- the antibody of the present invention had significantly higher binding ability to SYNGR3 than the control antibody, even when used at a low concentration of 100 ng/mL.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- General Health & Medical Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Medicinal Chemistry (AREA)
- Biomedical Technology (AREA)
- Zoology (AREA)
- Biochemistry (AREA)
- Biotechnology (AREA)
- Wood Science & Technology (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- General Engineering & Computer Science (AREA)
- Animal Behavior & Ethology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Biophysics (AREA)
- Microbiology (AREA)
- Epidemiology (AREA)
- Physics & Mathematics (AREA)
- Mycology (AREA)
- Plant Pathology (AREA)
- Hematology (AREA)
- Analytical Chemistry (AREA)
- Neurology (AREA)
- Cell Biology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Urology & Nephrology (AREA)
- General Physics & Mathematics (AREA)
- Food Science & Technology (AREA)
- Neurosurgery (AREA)
- Orthopedic Medicine & Surgery (AREA)
Abstract
Description
本発明は、シナプトジャイリン3の小胞内ドメインに結合するモノクローナル抗体に関する。本発明の抗体は、運動神経細胞への生理活性物質及び/又は標識物質送達のための標的化剤、及びそれを含む医薬組成物として使用することができる。 The present invention relates to a monoclonal antibody that binds to the intravesicular domain of synaptogyrin 3. The antibody of the present invention can be used as a targeting agent for delivering a physiologically active substance and/or a labeled substance to a motor neuron, and a pharmaceutical composition containing the same.
神経は、中枢神経及び末梢神経から構成され、種々の情動、筋肉及び内臓等諸臓器の機能等を調節する。このような神経機能は、神経の損傷、疾患、及び老化等によって低下する場合があり、かかる場合、心身の健康が害され、社会生活を営む上で甚大な影響を受けることとなり得る。したがって、神経機能を維持及び改善することは、クオリティ・オブ・ライフ(QOL)の維持及び改善に直結するため、極めて重大な課題といえる。 The nervous system is made up of the central nervous system and peripheral nervous system, and regulates various emotions and the functions of muscles, internal organs, etc. Such nervous function can be impaired by nerve damage, disease, aging, etc., which can impair physical and mental health and have a significant impact on social life. Therefore, maintaining and improving nervous function is an extremely important issue, as it is directly linked to maintaining and improving quality of life (QOL).
中枢神経及び末梢神経は、それぞれ神経細胞から構成され、これらの細胞はシナプスを介して相互にシグナルを交換する。シナプスは、神経細胞の軸索終末(シナプス前部)と、別の神経細胞の樹状突起又は骨格筋や臓器等の細胞(シナプス後部)との間に形成される間隙を含む接合部であり、シナプス前部から放出される化学物質がシナプス後部に存在する受容体に結合することでシグナルを伝達する。シナプス形成は、シナプス前部及び後部に発現する特異的な膜タンパク質の相互作用が引き金となって起こる。 The central and peripheral nerves are each composed of nerve cells, and these cells exchange signals with each other through synapses. A synapse is a junction that includes a gap formed between the axon terminal of a nerve cell (presynapse) and the dendrite of another nerve cell or a cell of a skeletal muscle or organ (postsynapse), and signals are transmitted when chemicals released from the presynapse bind to receptors present in the postsynapse. Synapse formation is triggered by the interaction of specific membrane proteins expressed in the presynapse and postsynapse.
近年、シナプス形成を促進することにより神経機能を維持及び改善することが可能な化合物やペプチドが開発されている。特許文献1には、特定のペプチドが、皮質ニューロン初代培養細胞(PCN)において樹状突起伸長促進作用及びシナプス形成促進作用を有すること、そのようなペプチドを軽度認知障害又は初期認知症の治療に用いることが記載されている。そして、特許文献2には、アミロイド前駆体タンパク(APP)がβセクレターゼによって切断されることにより生成するC末端フラグメントβ(CTFβ)が、シナプス形成を促進すること、CTFβを神経変性疾患の治療等に用いることが記載されている。 In recent years, compounds and peptides have been developed that can maintain and improve neural function by promoting synapse formation. Patent Document 1 describes that a specific peptide has the effect of promoting dendritic outgrowth and synapse formation in primary cultured cortical neuron cells (PCN), and that such peptides are used to treat mild cognitive impairment or early dementia. Patent Document 2 describes that C-terminal fragment β (CTFβ), which is generated by cleavage of amyloid precursor protein (APP) by β-secretase, promotes synapse formation, and that CTFβ is used to treat neurodegenerative diseases, etc.
さらに、特許文献3には、LRRTM分子又はその分子を含む融合タンパク質をその表面に固定化させたマイクロビーズを用いた、シナプス前部を有する運動神経細胞の培養方法が記載されている。このように、薬剤の運動神経細胞の機能への影響をin vitroにおいて簡便にスクリーニング可能な技術が開発されていることにより、今後、運動神経細胞に作用可能な化合物や薬剤候補が数多く提供されることが予想される。 Furthermore, Patent Document 3 describes a method for culturing motor neurons with presynapses using microbeads with LRRTM molecules or fusion proteins containing said molecules immobilized on their surface. In this way, with the development of a technology that allows for easy in vitro screening of the effects of drugs on motor neuron function, it is expected that in the future a large number of compounds and drug candidates that can act on motor neurons will be provided.
本発明者らは、運動神経細胞のシナプス小胞に存在する膜タンパク質に結合可能な抗体を利用した、標的の運動神経細胞への薬剤の制御された輸送を可能にする方法を開発した(PCT/JP2023/016125)。この方法によれば、シナプトタグミン2をはじめとする数種類の小胞膜タンパク質の小胞内ドメインに結合可能な抗体を用いて、標的の運動神経細胞において、抗体に連結した生理活性物質の薬理効果を得られることが確認されている。しかしながら、これまでこのような利用方法が想定されていなかったことから、この方法にて良好な効果を発揮する、膜タンパク質に結合可能な優れた抗体は知られていなかった。 The present inventors have developed a method that enables the controlled delivery of drugs to target motor neurons, using antibodies capable of binding to membrane proteins present in the synaptic vesicles of motor neurons (PCT/JP2023/016125). According to this method, it has been confirmed that it is possible to obtain the pharmacological effects of a physiologically active substance linked to an antibody in a target motor neuron, using an antibody capable of binding to the intravesicular domain of several types of vesicular membrane proteins, including synaptotagmin 2. However, because such a method of use had not been anticipated until now, no excellent antibodies capable of binding to membrane proteins that would exert a good effect using this method were known.
したがって、本発明は、運動神経細胞のシナプス小胞に存在する膜タンパク質に結合可能な優れたモノクローナル抗体の提供、及びそれを用いた運動神経細胞への物質の標的化手段及び標的化部位の可視化手段を提供することを課題とする。 The objective of the present invention is therefore to provide an excellent monoclonal antibody capable of binding to a membrane protein present in the synaptic vesicles of motor neurons, and to provide a means of targeting a substance to a motor neuron and a means of visualizing the targeting site using the same.
本発明者らは、上記課題を解決するため鋭意検討を重ねた結果、シナプトジャイリン3の小胞内ドメインに結合する優れたモノクローナル抗体を取得することに成功し、この抗体を用いることにより運動神経細胞シナプスに所望の物質を効率的に標的化可能であることを見出し、本発明を完成するに至った。 As a result of extensive research to solve the above problems, the inventors succeeded in obtaining an excellent monoclonal antibody that binds to the intravesicular domain of synaptogyrin 3, and discovered that this antibody can be used to efficiently target desired substances to motor neuron synapses, thus completing the present invention.
本発明は、これらの知見に基づいてなされたものであり、以下を提供する。
[1]シナプトジャイリン3の小胞内ドメインに結合可能な抗体であって、配列番号3に示されるアミノ酸配列からなるHCDR1、配列番号4に示されるアミノ酸配列からなるHCDR2、及び配列番号5に示されるアミノ酸配列からなるHCDR3を含む重鎖可変領域、及び配列番号6に示されるアミノ酸配列からなるLCDR1、配列番号7に示されるアミノ酸配列からなるLCDR2、及び配列番号8に示されるアミノ酸配列からなるLCDR3を含む軽鎖可変領域を含む、前記抗体。
[2]以下の(a)~(c)のいずれかから選択される、[1]に記載の抗体:(a)前記重鎖可変領域が、配列番号1に対して90%以上の配列同一性を有するアミノ酸配列からなり、前記軽鎖可変領域が、配列番号2に対して90%以上の配列同一性を有するアミノ酸配列からなる抗体;(b)前記重鎖可変領域が、配列番号1において1個若しくは数個のアミノ酸が置換、欠失、及び/又は付加されたアミノ酸配列からなり、前記軽鎖可変領域が配列番号2において1個若しくは数個のアミノ酸が置換、欠失、及び/又は付加されたアミノ酸配列からなる抗体;又は(c)前記重鎖可変領域が配列番号1に示されるアミノ酸配列からなり、前記軽鎖可変領域が配列番号2に示されるアミノ酸配列からなる抗体。
[3][1]又は[2]に記載の抗体と、標識物質及び/又は生理活性物質とのコンジュゲート。
[4][1]又は[2]に記載の抗体をコードする塩基配列を有する核酸分子。
[5][4]に記載の核酸分子を含む細胞。
[6][1]又は[2]に記載の抗体を含む、運動神経細胞への標的化剤。
[7]標識物質及び/又は生理活性物質をさらに含む、[6]に記載の標的化剤。
[8]前記抗体と、前記標識物質及び/又は前記生理活性物質とのコンジュゲートを含む、[7]に記載の標的化剤。
[9]前記標識物質が蛍光分子である、[7]又は[8]に記載の標的化剤。
[10]運動神経細胞可視化剤である、前記標識物質を含む[7]~[9]のいずれかに記載の標的化剤。
[11]前記生理活性物質が、シナプス形成促進剤、シナプス維持剤、筋肉増強剤及び神経細胞機能改変剤からなる群から選択される一以上である、[7]~[10]のいずれかに記載の標的化剤。
[12]エンドサイトーシスによって細胞内に取り込まれる、[6]~[11]のいずれかに記載の標的化剤。
[13][7]~[12]のいずれかに記載の標的化剤を含む医薬組成物。
[14][7]~[12]のいずれかに記載の標的化剤及び/又は[13]に記載の医薬組成物を運動神経細胞に接触させる工程、及び前記標的化剤及び/又は前記医薬組成物を前記運動神経細胞のシナプスに送達する工程を含む、運動神経細胞への前記標識物質及び/又は前記生理活性物質の標的化方法。
[15][10]に記載の運動神経細胞可視化剤である標的化剤を運動神経細胞に接触させる工程、前記標的化剤を前記運動神経細胞のシナプスに送達する工程、及び前記標識物質のシグナルを検出する工程を含む、運動神経細胞可視化方法。
[16][7]~[12]のいずれかに記載の標的化剤を運動神経細胞に接触させる工程、及び前記標的化剤を前記運動神経細胞シナプスに送達する工程を含む、状態又は疾患の予防又は治療方法。
[17]前記状態又は疾患が神経機能の低下を呈する状態若しくは疾患である、[16]に記載の予防又は治療方法。
[18]前記状態又は疾患が神経疾患及び神経筋疾患である、[16]又は[17]に記載の予防又は治療方法。
[19]前記標的化剤が、前記抗体と前記生理活性物質とのコンジュゲートを含む、[16]~[18]のいずれかに記載の予防又は治療方法。
[20]シナプトジャイリン3の小胞内ドメインに結合可能な抗体の、前記抗体及び生理活性物質を含む医薬の製造における使用であって、前記抗体が、配列番号3に示されるアミノ酸配列からなるHCDR1、配列番号4に示されるアミノ酸配列からなるHCDR2、及び配列番号5に示されるアミノ酸配列からなるHCDR3を含む重鎖可変領域、及び配列番号6に示されるアミノ酸配列からなるLCDR1、配列番号7に示されるアミノ酸配列からなるLCDR2、及び配列番号8に示されるアミノ酸配列からなるLCDR3を含む軽鎖可変領域を含む、前記使用。
[21]シナプトジャイリン3の小胞内ドメインに結合可能な抗体を含む、運動神経細胞への標的化用組成物であって、前記抗体が、配列番号3に示されるアミノ酸配列からなるHCDR1、配列番号4に示されるアミノ酸配列からなるHCDR2、及び配列番号5に示されるアミノ酸配列からなるHCDR3を含む重鎖可変領域、及び配列番号6に示されるアミノ酸配列からなるLCDR1、配列番号7に示されるアミノ酸配列からなるLCDR2、及び配列番号8に示されるアミノ酸配列からなるLCDR3を含む軽鎖可変領域を含む、前記組成物。
[22]状態又は疾患の予防又は治療方法において使用するための、シナプトジャイリン3の小胞内ドメインに結合可能な抗体であって、配列番号3に示されるアミノ酸配列からなるHCDR1、配列番号4に示されるアミノ酸配列からなるHCDR2、及び配列番号5に示されるアミノ酸配列からなるHCDR3を含む重鎖可変領域、及び配列番号6に示されるアミノ酸配列からなるLCDR1、配列番号7に示されるアミノ酸配列からなるLCDR2、及び配列番号8に示されるアミノ酸配列からなるLCDR3を含む軽鎖可変領域を含む、前記抗体。
[23]運動神経細胞への標識物質及び/又は生理活性物質の標的化方法において使用するための、シナプトジャイリン3の小胞内ドメインに結合可能な抗体であって、配列番号3に示されるアミノ酸配列からなるHCDR1、配列番号4に示されるアミノ酸配列からなるHCDR2、及び配列番号5に示されるアミノ酸配列からなるHCDR3を含む重鎖可変領域、及び配列番号6に示されるアミノ酸配列からなるLCDR1、配列番号7に示されるアミノ酸配列からなるLCDR2、及び配列番号8に示されるアミノ酸配列からなるLCDR3を含む軽鎖可変領域を含む、前記抗体。
本明細書は本願の優先権の基礎となる日本国特許出願番号2023-182983号の開示内容を包含する。
The present invention has been made based on these findings, and provides the following.
[1] An antibody capable of binding to the intravesicular domain of synaptogyrin 3, comprising a heavy chain variable region including an HCDR1 having the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 having the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 having the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region including an LCDR1 having the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 having the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 having the amino acid sequence shown in SEQ ID NO: 8.
[2] The antibody according to [1], selected from any of the following (a) to (c): (a) an antibody in which the heavy chain variable region consists of an amino acid sequence having 90% or more sequence identity to SEQ ID NO: 1, and the light chain variable region consists of an amino acid sequence having 90% or more sequence identity to SEQ ID NO: 2; (b) an antibody in which the heavy chain variable region consists of an amino acid sequence in SEQ ID NO: 1 with one or more amino acid substitutions, deletions, and/or additions, and the light chain variable region consists of an amino acid sequence in SEQ ID NO: 2 with one or more amino acid substitutions, deletions, and/or additions; or (c) an antibody in which the heavy chain variable region consists of the amino acid sequence shown in SEQ ID NO: 1, and the light chain variable region consists of the amino acid sequence shown in SEQ ID NO: 2.
[3] A conjugate of the antibody according to [1] or [2] with a labeling substance and/or a physiologically active substance.
[4] A nucleic acid molecule having a base sequence encoding the antibody according to [1] or [2].
[5] A cell comprising the nucleic acid molecule according to [4].
[6] A targeting agent for motor neurons, comprising the antibody according to [1] or [2].
[7] The targeting agent according to [6], further comprising a labeling substance and/or a physiologically active substance.
[8] The targeting agent according to [7], comprising a conjugate of the antibody with the labeling substance and/or the physiologically active substance.
[9] The targeting agent according to [7] or [8], wherein the labeling substance is a fluorescent molecule.
[10] The targeting agent according to any one of [7] to [9], which is a motor neuron visualization agent and contains the labeling substance.
[11] The targeting agent according to any one of [7] to [10], wherein the physiologically active substance is one or more selected from the group consisting of synapse formation promoters, synapse maintenance agents, muscle strengthening agents, and nerve cell function modifying agents.
[12] The targeting agent according to any one of [6] to [11], which is taken up into a cell by endocytosis.
[13] A pharmaceutical composition comprising the targeting agent according to any one of [7] to [12].
[14] A method for targeting a labeled substance and/or a physiologically active substance to a motor neuron, comprising the steps of contacting a motor neuron with the targeting agent according to any one of [7] to [12] and/or the pharmaceutical composition according to [13], and delivering the targeting agent and/or the pharmaceutical composition to a synapse of the motor neuron.
[15] A method for visualizing motor neurons, comprising the steps of contacting a targeting agent, which is the motor neuron visualization agent described in [10], with a motor neuron, delivering the targeting agent to a synapse of the motor neuron, and detecting a signal of the labeling substance.
[16] A method for preventing or treating a condition or disease, comprising the steps of contacting a motor neuron with the targeting agent according to any one of [7] to [12], and delivering the targeting agent to the motor neuron synapse.
[17] The method for prevention or treatment described in [16], wherein the condition or disease is a condition or disease exhibiting decreased neurological function.
[18] The method for prevention or treatment according to [16] or [17], wherein the condition or disease is a neurological disease or a neuromuscular disease.
[19] The method for prevention or treatment according to any one of [16] to [18], wherein the targeting agent comprises a conjugate of the antibody and the physiologically active substance.
[20] Use of an antibody capable of binding to the intravesicular domain of synaptogyrin 3 in the manufacture of a pharmaceutical comprising the antibody and a physiologically active substance, wherein the antibody comprises a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
[21] A composition for targeting motor neurons, comprising an antibody capable of binding to the intravesicular domain of synaptogyrin 3, wherein the antibody comprises a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
[22] An antibody capable of binding to the intravesicular domain of synaptogyrin 3 for use in a method for preventing or treating a condition or disease, comprising a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
[23] An antibody capable of binding to the intravesicular domain of synaptogyrin 3 for use in a method for targeting a labeling substance and/or a physiologically active substance to a motor neuron, comprising a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
This specification includes the disclosure of Japanese Patent Application No. 2023-182983, which is the priority basis of this application.
本発明により、所望の物質を運動神経細胞へと標的化することができるモノクローナル抗体が提供される。物質が生理活性物質又は治療剤である場合、運動神経細胞の異常による状態及び/又は疾患を処置することができる。また、物質が標識物質である場合、運動神経細胞を可視化することができる。 The present invention provides a monoclonal antibody capable of targeting a desired substance to motor neurons. When the substance is a physiologically active substance or a therapeutic agent, conditions and/or diseases caused by abnormalities in motor neurons can be treated. When the substance is a labeling substance, motor neurons can be visualized.
本発明は、シナプトジャイリン3の小胞内ドメインに結合可能であり、目的の物質の運動神経細胞への標的化に使用可能なモノクローナル抗体(「本発明の抗体」と称する)を提供する。 The present invention provides a monoclonal antibody (referred to as the "antibody of the present invention") that can bind to the intravesicular domain of synaptogyrin 3 and can be used to target a substance of interest to a motor neuron.
<シナプトジャイリン3>
本明細書において、「シナプトジャイリン3(Synaptogyrin 3:SYNGR3)」とは、シナプトジャイリンファミリーに属する4回膜貫通型タンパク質の1つを指す。このタンパク質は、運動神経細胞等のシナプス前小胞に存在し、制御性エキソサイトーシス、ドーパミンのリサイクリング等に関与していると考えられている。シナプトジャイリン3は、4つの膜貫通ドメインに隔てられた3つの細胞質ドメイン及び2つの小胞内ドメインを有する。
<Synaptogyrin 3>
As used herein, "synaptogyrin 3 (SYNGR3)" refers to one of the four-transmembrane proteins belonging to the synaptogyrin family. This protein is present in presynaptic vesicles of motor neurons and is thought to be involved in regulated exocytosis, dopamine recycling, and the like. Synaptogyrin 3 has three cytoplasmic domains and two intravesicular domains separated by four transmembrane domains.
シナプトジャイリン3の小胞内ドメインは、シナプス小胞の内腔に露出している。細胞内のカルシウムイオン濃度の上昇に伴ってシナプス小胞と細胞膜とが融合すると、シナプス小胞の内腔が細胞外空間とつながる結果、シナプトジャイリン3の小胞内ドメインが細胞外に一時的に露出する。その後、シナプトジャイリン3を含む細胞膜部分はシナプス小胞膜としてエンドサイトーシスにより細胞内に回収され、シナプス小胞として再利用される。この際にシナプトジャイリン3の小胞内ドメインは、再びシナプス小胞内腔に露出する。 The vesicular domain of synaptogyrin 3 is exposed to the lumen of synaptic vesicles. When synaptic vesicles fuse with the plasma membrane as the intracellular calcium ion concentration increases, the lumen of the synaptic vesicle connects with the extracellular space, and the vesicular domain of synaptogyrin 3 is temporarily exposed to the outside of the cell. The plasma membrane portion containing synaptogyrin 3 is then retrieved into the cell as a synaptic vesicle membrane by endocytosis and reused as synaptic vesicles. At this time, the vesicular domain of synaptogyrin 3 is again exposed to the lumen of the synaptic vesicle.
具体的には、例示的なヒトシナプトジャイリン3は、アミノ酸配列が配列番号9で表される229アミノ酸からなるタンパク質である。各ドメインの位置は、例えば、配列番号9においては、第1の細胞質ドメインは1位~29位のアミノ酸配列で示される領域、第1の膜貫通ドメインは30位~50位のアミノ酸配列で示される領域、第1の小胞内ドメインは51位~69位のアミノ酸配列で示される領域、第2の膜貫通ドメインは70位~90位のアミノ酸配列で示される領域、第2の細胞質ドメインは91位~104位のアミノ酸配列で示される領域、第3の膜貫通ドメインは105位~125位のアミノ酸配列で示される領域、第2の小胞内ドメインは126位~147位のアミノ酸配列で示される領域、第4の膜貫通ドメインは148位~168位のアミノ酸配列で示される領域、及び第3の細胞質ドメインは169位~229位のアミノ酸配列で示される領域である。
Specifically, an exemplary human synaptogyrin 3 is a protein consisting of 229 amino acids whose amino acid sequence is represented by SEQ ID NO: 9. For example, in SEQ ID NO: 9, the positions of each domain are as follows: the first cytoplasmic domain is the region represented by the amino acid sequence of positions 1 to 29, the first transmembrane domain is the region represented by the amino acid sequence of positions 30 to 50, the first intravesicular domain is the region represented by the amino acid sequence of positions 51 to 69, the second transmembrane domain is the region represented by the amino acid sequence of
マウスをはじめとする他の生物のシナプトジャイリン3の配列情報は、NCBIデータベース等の公知のデータベースより容易に取得することができる。 Sequence information for synaptogyrin 3 from mice and other organisms can be easily obtained from publicly known databases such as the NCBI database.
本明細書において、「シナプトジャイリン3の小胞内ドメイン」とは、シナプトジャイリン3の2つの小胞内ドメインのうちいずれかの全部又は一部を指す。小胞内ドメインの全部としては、例えば、配列番号9における51位~69位のアミノ酸配列(配列番号10)で示される第1の小胞内ドメイン及び/又は126位~147位のアミノ酸配列(配列番号11)で示される第2の小胞内ドメインが含まれる。好ましくは、配列番号11で示される第2の小胞内ドメインである。また、小胞内ドメインの一部としては、例えば、配列番号10及び/又は11における任意の長さの部分配列で示される領域が含まれる。好ましい小胞内ドメインの一部としては、例えば、配列番号9における127位~144位のアミノ酸配列(配列番号12)で示されるアミノ酸配列が挙げられる。 In this specification, the term "synaptogyrin 3 vesicular domain" refers to all or part of either of the two vesicular domains of synaptogyrin 3. The entire intravesicular domain includes, for example, the first intravesicular domain represented by the amino acid sequence of positions 51 to 69 in SEQ ID NO: 9 (SEQ ID NO: 10) and/or the second intravesicular domain represented by the amino acid sequence of positions 126 to 147 in SEQ ID NO: 11. The second intravesicular domain represented by SEQ ID NO: 11 is preferred. A portion of the intravesicular domain includes, for example, a region represented by a partial sequence of any length in SEQ ID NO: 10 and/or 11. A preferred portion of the intravesicular domain includes, for example, the amino acid sequence represented by the amino acid sequence of positions 127 to 144 in SEQ ID NO: 9 (SEQ ID NO: 12).
なお、抗体の取得の際に使用する抗原ペプチドとしては、最終的に小胞内ドメインに結合可能な抗体を取得可能であれば、小胞内ドメイン又はその一部のアミノ酸配列に追加で他のアミノ酸を含むペプチドを使用することができる。そのように使用可能な抗原ペプチドとしては、例えば、配列番号13又は14で示されるアミノ酸配列を有するペプチド等が挙げられる。 As for the antigen peptide used to obtain the antibody, a peptide containing additional amino acids in the amino acid sequence of the intravesicular domain or a part of it can be used, so long as it is possible to obtain an antibody that can bind to the intravesicular domain. Examples of antigen peptides that can be used in this way include peptides having the amino acid sequence shown in SEQ ID NO: 13 or 14.
<本発明の抗体>
本発明の抗体は、シナプトジャイリン3の小胞内ドメインを特異的に認識して結合し得るモノクローナル抗体(「本発明の抗体」と称する)である。本発明の抗体は、シナプトジャイリン3の小胞内ドメインには結合するが、好ましくは、シナプトジャイリンファミリーに属する他の膜タンパク質(例えば、シナプトジャイリン1やシナプトジャイリン4)の小胞内ドメインには結合しない。また、本発明の抗体は、ヒトシナプトジャイリン3の小胞内ドメインに結合するが、例えば、その他の動物(マウス等)由来のシナプトジャイリン3の小胞内ドメインには、結合可能であってもよく、結合可能でなくてもよい。
<Antibody of the Invention>
The antibody of the present invention is a monoclonal antibody (referred to as "the antibody of the present invention") that can specifically recognize and bind to the intravesicular domain of synaptogyrin 3. The antibody of the present invention binds to the intravesicular domain of synaptogyrin 3, but preferably does not bind to the intravesicular domain of other membrane proteins belonging to the synaptogyrin family (e.g., synaptogyrin 1 and synaptogyrin 4). In addition, the antibody of the present invention binds to the intravesicular domain of human synaptogyrin 3, but may or may not be able to bind to the intravesicular domain of synaptogyrin 3 derived from other animals (e.g., mouse).
なお、本明細書において、「結合する」及び「特異的に結合する」とは、特に限定するものではないが、抗原と抗体との結合が10-8M以下のKD値の結合親和性を有するものであることを意味し得る。本発明の抗体は、シナプトジャイリン3の小胞内ドメインに対して上記の結合親和性を有する。本発明の抗体は、他のタンパク質を含む他の抗原(例えば、シナプトタグミン1等のシナプトタグミンファミリーに属する他の膜タンパク質の小胞内ドメイン等)等に対しては上記の結合親和性を有さないことが好ましい。または、結合親和性は、結合速度定数が大きいことによって判断してもよい。この場合の結合速度定数の具体的な値は特に限定しないが、例えば、103/Ms以上、104/Ms以上、105/Ms以上(10×105/Ms以上、15×105/Ms以上、16×105/Ms以上等)であればよい。結合親和性及び結合速度定数の測定方法としては、例えば、表面プラズモン共鳴法の他、バイオレイヤー干渉法等が挙げられ、例えば、バイオレイヤー干渉法を好適に使用することができる。 In this specification, "binding" and "specific binding" are not particularly limited, but may mean that the binding between the antigen and the antibody has a binding affinity with a KD value of 10 -8 M or less. The antibody of the present invention has the above-mentioned binding affinity to the intravesicular domain of synaptogyrin 3. It is preferable that the antibody of the present invention does not have the above-mentioned binding affinity to other antigens including other proteins (for example, intravesicular domains of other membrane proteins belonging to the synaptotagmin family such as synaptotagmin 1, etc.). Alternatively, the binding affinity may be determined by a large binding rate constant. The specific value of the binding rate constant in this case is not particularly limited, but may be, for example, 10 3 /Ms or more, 10 4 /Ms or more, 10 5 /Ms or more (10 × 10 5 /Ms or more, 15 × 10 5 /Ms or more, 16 × 10 5 /Ms or more, etc.). Methods for measuring the binding affinity and binding rate constant include, for example, surface plasmon resonance and biolayer interference, and for example, biolayer interference can be preferably used.
あるいはまた、本明細書において「標的物質に特異的に結合する」とは、標的物質、例えばシナプトジャイリン3の小胞内ドメインに対する結合が、例えば過剰量の標的物質を使用し、一般的な結合アッセイで検出して、標的物質以外の物質に対する結合と比較して2倍以上、3倍以上、4倍以上である場合を意味し得る。また、上記したように蛍光標識によって結合を検出する場合に、S/N比が2以上、3以上、4以上である場合を意味し得る。 Alternatively, in this specification, "specifically binds to a target substance" can mean that the binding to a target substance, for example, the intravesicular domain of synaptogyrin 3, is at least two-fold, three-fold, or four-fold stronger than the binding to a substance other than the target substance, for example, when detected by a general binding assay using an excess amount of the target substance. It can also mean that, when binding is detected by fluorescent labeling as described above, the S/N ratio is 2 or more, 3 or more, or 4 or more.
本発明の抗体は、マウス、ウサギ、ヤギ等の非ヒト抗体、キメラ抗体、ヒト化抗体、ヒト抗体のいずれであっても良いが、ヒトにおける状態・疾患の治療に使用する場合、特に限定するものではないが、ヒト化抗体又はヒト抗体であることが好ましい。したがって、本発明において好適に使用することができる抗体は、ヒト抗体のフレームワーク領域(FR)と、シナプトジャイリン3の小胞内ドメインに対して高い結合親和性をもたらし得る相補性決定領域(CDR)とを含む抗体であり得る。 The antibody of the present invention may be any of non-human antibodies of mouse, rabbit, goat, etc., chimeric antibodies, humanized antibodies, and human antibodies, but when used to treat conditions and diseases in humans, it is preferable, although not limited thereto, that it be a humanized antibody or a human antibody. Therefore, an antibody that can be suitably used in the present invention may be an antibody that contains a framework region (FR) of a human antibody and a complementarity determining region (CDR) that can provide high binding affinity to the intravesicular domain of synaptogyrin 3.
本発明の抗体は、IgG抗体分子、又はその抗原結合性断片及び抗原結合性誘導体であり得る。例えば、抗体は、完全抗体、Fab、Fab'、F(ab')2断片、また重鎖可変領域(VH)及び軽鎖可変領域(VL)をリンカーで連結した一本鎖抗体(scFv)断片、scFv-Fc、sc(Fv)2、Fv、ダイアボディー等であり得る。 The antibody of the present invention may be an IgG antibody molecule, or an antigen-binding fragment or derivative thereof. For example, the antibody may be a complete antibody, a Fab, Fab', or F(ab') 2 fragment, or a single-chain antibody (scFv) fragment in which the heavy chain variable region (VH) and the light chain variable region (VL) are linked via a linker, such as scFv-Fc, sc(Fv) 2 , Fv, or a diabody.
scFv、scFv-Fc、及びsc(Fv)2はリンカーで可変領域を連結した合成ポリペプチドである。リンカーとしては、当分野で通常使用されるものであればいずれでも良く、特に限定するものではないが、例えば5~25個、好ましくは10~20個のアミノ酸残基からなるペプチドリンカー、例えばGSリンカー等を好適に使用することができる。 scFv, scFv-Fc, and sc(Fv) 2 are synthetic polypeptides in which variable regions are linked by a linker. The linker may be any linker commonly used in the art, and is not particularly limited. For example, a peptide linker consisting of 5 to 25, preferably 10 to 20, amino acid residues, such as a GS linker, can be suitably used.
本発明の抗体にはさらに、抗原結合性に影響しない範囲で当業者に理解され得る誘導体、例えば抗体精製を容易にしたり安定性を高めたりするための修飾が施された誘導体、及び抗がん剤等の薬物と結合させた複合体も含まれる。本明細書においては、シナプトジャイリン3の小胞内ドメインとの結合性を保持する断片及び誘導体を、文脈に矛盾のない限り、便宜的に「抗体」に含めることが意図される。 The antibodies of the present invention further include derivatives that would be understood by those skilled in the art to the extent that they do not affect antigen-binding ability, such as derivatives modified to facilitate antibody purification or to increase stability, and conjugates bound to drugs such as anticancer drugs. In this specification, fragments and derivatives that retain the ability to bind to the intravesicular domain of synaptogyrin 3 are intended to be included in the term "antibody" for the sake of convenience, unless the context is inconsistent.
本発明の抗体はまた、二量体、三量体、四量体等の多量体として合成することもできる。さらに、本発明の抗体は、シナプトジャイリン3の小胞内ドメインに結合する第1の特異性と、他の抗原に対して結合する第2の特異性とを有する二重特異性抗体であっても良い。第2の特異性は、例えば標的細胞が発現し得る別の抗原に対するものであっても、送達目的の生理活性物質及び/又は標識物質に対するものであっても良く、目的に応じて適宜選択することができる。第2の特異性が、標的細胞、つまり運動神経細胞が発現し得る別の抗原に対するものである場合、二重特異性抗体の使用によって、標的細胞への送達がより特異的にもたらされる。また、第2の特異性が、生理活性物質及び/又は標識物質に対するものである場合、それらの物質を、二重特異性抗体と共に投与することにより、複合体として標的部位に送達することができる。 The antibody of the present invention can also be synthesized as a multimer, such as a dimer, trimer, or tetramer. Furthermore, the antibody of the present invention can be a bispecific antibody having a first specificity that binds to the intravesicular domain of synaptogyrin 3 and a second specificity that binds to another antigen. The second specificity can be, for example, for another antigen that can be expressed by the target cell, or for a physiologically active substance and/or a labeling substance to be delivered, and can be appropriately selected depending on the purpose. When the second specificity is for another antigen that can be expressed by the target cell, i.e., the motor neuron, the use of the bispecific antibody provides more specific delivery to the target cell. Furthermore, when the second specificity is for a physiologically active substance and/or a labeling substance, the substance can be delivered to the target site as a complex by administering it together with the bispecific antibody.
当業者であれば、本明細書の記載、及び当分野における技術常識に基づいて、本発明の抗体を用途に応じた適切な形態のものとして取得することができる。 Those skilled in the art can obtain the antibodies of the present invention in an appropriate form according to the intended use based on the description in this specification and common technical knowledge in the field.
特に限定するものではないが、本発明のシナプトジャイリン3の小胞内ドメインに結合する抗体の例として、以下の抗体を挙げることができる。 Although not particularly limited, examples of antibodies that bind to the intravesicular domain of synaptogyrin 3 of the present invention include the following antibodies.
配列番号3に示されるアミノ酸配列からなるHCDR1、配列番号4に示されるアミノ酸配列からなるHCDR2、及び配列番号5に示されるアミノ酸配列からなるHCDR3を含む重鎖可変領域、及び
配列番号6に示されるアミノ酸配列からなるLCDR1、配列番号7に示されるアミノ酸配列からなるLCDR2、及び配列番号8に示されるアミノ酸配列からなるLCDR3を含む軽鎖可変領域
を含む、抗体。
An antibody comprising a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
この場合、本発明の他の領域のアミノ酸配列(例えば、FRのアミノ酸配列、定常領域のアミノ酸配列)は特に限定しない。抗体の結合特性は、基本的には重鎖及び軽鎖のCDRによって決定される。したがって、本発明の抗体においてアミノ酸配列が改変されてもよいが、その改変は、結合特性に与える影響が少ないという観点から、フレームワーク領域又は定常領域内にあることが好ましい。また、改変は、抗体の立体構造を大きく変更しないものであることが好ましい。 In this case, the amino acid sequences of other regions of the present invention (e.g., amino acid sequences of FR, amino acid sequences of constant regions) are not particularly limited. The binding characteristics of an antibody are basically determined by the CDRs of the heavy and light chains. Therefore, the amino acid sequence of the antibody of the present invention may be modified, but it is preferable that the modification is within the framework region or constant region from the viewpoint of having little effect on the binding characteristics. In addition, it is preferable that the modification does not significantly change the three-dimensional structure of the antibody.
また、本発明のシナプトジャイリン3の小胞内ドメインに結合する抗体として、以下の抗体が挙げられる:
(a)上述のHCDR1~3及びLCDR1~3を有し、重鎖可変領域が、配列番号1に対して90%以上の配列同一性を有するアミノ酸配列からなり、軽鎖可変領域が、配列番号2に対して90%以上の配列同一性を有するアミノ酸配列からなる抗体。
Furthermore, examples of the antibodies of the present invention that bind to the intravesicular domain of synaptogyrin 3 include the following antibodies:
(a) An antibody having the above-mentioned HCDR1 to 3 and LCDR1 to 3, wherein the heavy chain variable region consists of an amino acid sequence having 90% or more sequence identity to SEQ ID NO:1, and the light chain variable region consists of an amino acid sequence having 90% or more sequence identity to SEQ ID NO:2.
本明細書において(アミノ酸配列の)「配列同一性」とは、比較する2つのポリペプチドのアミノ酸配列において、アミノ酸残基の一致数が最大となるように、必要に応じて一方又は双方に適宜ギャップを挿入して整列化(アラインメント)したときに、全アミノ酸残基数における一致アミノ酸残基数の割合(%)をいう。配列同一性は当分野で周知の手法及びソフトウェアを用いて算出することができる。 As used herein, "sequence identity" (of amino acid sequences) refers to the percentage of matching amino acid residues out of the total number of amino acid residues in the amino acid sequences of two polypeptides being compared, when the sequences are aligned by inserting appropriate gaps into one or both sequences as necessary to maximize the number of matching amino acid residues. Sequence identity can be calculated using methods and software well known in the art.
ここで、本発明の抗体におけるアミノ酸配列の配列同一性は90%以上であれば特に限定しない。例えば、重鎖可変領域のアミノ酸配列は、配列番号1に対して90%以上、91%以上、92%以上、93%以上、94%以上、95%以上、96%以上、97%以上、98%以上、又は99%以上の配列同一性を有するアミノ酸配列からなる。また、本発明の抗体の軽鎖可変領域のアミノ酸配列は、例えば、配列番号2に対して90%以上、91%以上、92%以上、93%以上、94%以上、95%以上、96%以上、97%以上、98%以上、又は99%以上の配列同一性を有するアミノ酸配列からなる。 Here, the sequence identity of the amino acid sequence in the antibody of the present invention is not particularly limited as long as it is 90% or more. For example, the amino acid sequence of the heavy chain variable region consists of an amino acid sequence that has a sequence identity of 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more to SEQ ID NO: 1. Furthermore, the amino acid sequence of the light chain variable region of the antibody of the present invention consists of an amino acid sequence that has a sequence identity of 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more to SEQ ID NO: 2.
また、本発明のシナプトジャイリン3の小胞内ドメインに結合する抗体として、以下の抗体が挙げられる:
(b)上述のHCDR1~3及びLCDR1~3を有し、重鎖可変領域が、配列番号1において1個若しくは数個のアミノ酸が置換、欠失、及び/又は付加されたアミノ酸配列からなり、軽鎖可変領域が配列番号2において1個若しくは数個のアミノ酸が置換、欠失、及び/又は付加されたアミノ酸配列からなる抗体。
Furthermore, examples of the antibodies of the present invention that bind to the intravesicular domain of synaptogyrin 3 include the following antibodies:
(b) An antibody having the above-mentioned HCDR1 to 3 and LCDR1 to 3, wherein the heavy chain variable region consists of the amino acid sequence of SEQ ID NO: 1 in which one or several amino acids have been substituted, deleted, and/or added, and the light chain variable region consists of the amino acid sequence of SEQ ID NO: 2 in which one or several amino acids have been substituted, deleted, and/or added.
ここで、「数個」とは、5個以下、例えば2個以上、3個以上、4個以上又は5個であり得る。したがって、本発明の抗体は、上記(b)の抗体における重鎖可変領域及び/若しくは軽鎖可変領域でそれぞれ5個以下(例えば、2~3個)のアミノ酸の改変を有し得る。ある抗体と機能的に同等な抗体の調製は、例えば部位特異的変異誘発法等の当分野で周知の手法によって行うことができる。 Here, "several" can be 5 or less, for example 2 or more, 3 or more, 4 or more, or 5. Thus, the antibody of the present invention can have 5 or less (e.g., 2 to 3) amino acid modifications in the heavy chain variable region and/or the light chain variable region of the antibody (b) above. Antibodies that are functionally equivalent to a certain antibody can be prepared by techniques well known in the art, such as site-directed mutagenesis.
本明細書において「アミノ酸の置換」とは、天然のタンパク質を構成する20種類のアミノ酸間の置換をいう。アミノ酸の置換は、特に限定するものではないが、保存的置換であり得る。保存的置換とは、電荷、側鎖、極性、芳香族性等の性質の類似する保存的アミノ酸群内での置換を指す。例えば、低極性側鎖を有する無電荷極性アミノ酸群(Gly, Asn, Gln, Ser, Thr, Cys, Tyr)、分枝鎖アミノ酸群(Leu, Val, Ile)、中性アミノ酸群(Gly, Ile, Val, Leu, Ala, Met, Pro)、親水性側鎖を有する中性アミノ酸群(Asn, Gln, Thr, Ser, Tyr, Cys)、酸性アミノ酸群(Asp, Glu)、塩基性アミノ酸群(Arg, Lys, His)、芳香族アミノ酸群(Phe, Tyr, Trp)内での置換が挙げられる。 As used herein, "amino acid substitution" refers to substitutions between the 20 types of amino acids that make up natural proteins. The amino acid substitutions can be, but are not limited to, conservative substitutions. Conservative substitutions refer to substitutions within a conservative amino acid group that has similar properties such as charge, side chain, polarity, and aromaticity. Examples of such substitutions include substitutions within the uncharged polar amino acid group with low polarity side chains (Gly, Asn, Gln, Ser, Thr, Cys, Tyr), branched-chain amino acids (Leu, Val, Ile), neutral amino acids (Gly, Ile, Val, Leu, Ala, Met, Pro), neutral amino acids with hydrophilic side chains (Asn, Gln, Thr, Ser, Tyr, Cys), acidic amino acids (Asp, Glu), basic amino acids (Arg, Lys, His), and aromatic amino acids (Phe, Tyr, Trp).
また、本発明のシナプトジャイリン3の小胞内ドメインに結合する抗体として、以下の抗体が挙げられる:
(c)重鎖可変領域が配列番号1に示されるアミノ酸配列からなり、軽鎖可変領域が配列番号2に示されるアミノ酸配列からなる抗体。
Furthermore, examples of the antibodies of the present invention that bind to the intravesicular domain of synaptogyrin 3 include the following antibodies:
(c) An antibody having a heavy chain variable region consisting of the amino acid sequence shown in SEQ ID NO: 1 and a light chain variable region consisting of the amino acid sequence shown in SEQ ID NO: 2.
生体内において、ある抗原に対する抗体が生じた後、「親和性成熟(affinity maturation)」と呼ばれるプロセスにより、初代の抗体よりも抗原に対する親和性が向上した抗体が生じることが知られている。したがって、上記の具体的に開示した抗体のアミノ酸配列に基づいて、開示した抗体と同等、又は開示した抗体よりも向上した活性を有する抗体、具体的にはシナプトジャイリン3の小胞内ドメインに対する結合親和性、物質の送達のための標的化剤としての機能性が向上した抗体を取得することが可能である。これらの改変は、フレームワーク領域及び定常領域内である場合に加えて、CDR領域内においても生じ得る。 It is known that after an antibody against a certain antigen is produced in the body, a process called "affinity maturation" produces an antibody with improved affinity for the antigen compared to the original antibody. Therefore, based on the amino acid sequences of the specifically disclosed antibodies above, it is possible to obtain antibodies with activity equivalent to or improved over the disclosed antibodies, specifically antibodies with improved binding affinity to the intravesicular domain of synaptogyrin 3 and functionality as a targeting agent for substance delivery. These modifications can occur within the CDR regions, in addition to within the framework and constant regions.
本発明の抗体を取得する方法は、特に限定しない。例えば、抗原として特定されたシナプトジャイリン3の小胞内ドメインを非ヒト哺乳動物に免疫して、公知の手法によってモノクローナル抗体として取得することができる。本発明の抗体はまた、活性が実証された本発明の抗体のアミノ酸配列情報又は該抗体をコードするポリヌクレオチドの塩基配列情報に基づいて、遺伝子工学的手法を用い、あるいは化学合成手段を用いて、合成によって取得することもできる。 The method for obtaining the antibody of the present invention is not particularly limited. For example, a non-human mammal can be immunized with the intravesicular domain of synaptogyrin 3 identified as an antigen, and a monoclonal antibody can be obtained by a known method. The antibody of the present invention can also be obtained synthetically using genetic engineering techniques or chemical synthesis means based on the amino acid sequence information of the antibody of the present invention whose activity has been demonstrated, or the base sequence information of the polynucleotide encoding the antibody.
遺伝子工学的手法によって抗体を作製する場合、重鎖及び軽鎖をコードするポリヌクレオチドを適切な宿主細胞に導入して発現させ、組換えタンパク質として抗体を取得することができる。この場合、ポリヌクレオチドはDNAであってもRNAであっても良く、また宿主細胞への導入手段は当分野で使用されているものを適宜利用することができる。ポリヌクレオチドを宿主細胞に導入するためのベクターとして、ウイルスベクター、プラスミドベクター、ファージベクター等を適宜使用することができる。宿主細胞としては、例えば大腸菌等の細菌、酵母、昆虫細胞、動物細胞等を利用することができる。ここで、重鎖及び軽鎖をコードするポリヌクレオチドは、別個のベクターに導入しても、同一のベクターに連結して導入しても良い。 When producing an antibody by genetic engineering techniques, polynucleotides encoding the heavy and light chains can be introduced into an appropriate host cell and expressed to obtain the antibody as a recombinant protein. In this case, the polynucleotide may be either DNA or RNA, and any method used in the art can be used as appropriate for introducing the polynucleotide into the host cell. As a vector for introducing the polynucleotide into the host cell, a virus vector, a plasmid vector, a phage vector, etc. can be used as appropriate. As the host cell, for example, bacteria such as E. coli, yeast, insect cells, animal cells, etc. can be used. Here, the polynucleotides encoding the heavy and light chains can be introduced into separate vectors, or can be linked to the same vector and introduced.
例えば、本発明の抗体によれば、生理活性物質とのコンジュゲートの形態で、運動神経細胞において優れた生理活性を示すことができる。本発明の抗体の使用に基づく生理活性の改善は、例えば、事前にLRRTM分子が表面に固定されたマイクロビーズと共に共培養し、軸索の伸長及びシナプス前部の形成が誘導された運動神経細胞に対してモノメチルアウリスタチンE(MMAE)と本発明のコンジュゲートを投与することにより得られた結果を指標として確認することができる。この際、例えば、ポリクローナルヤギ抗SYNGR3小胞内ドメイン抗体(Synaptogyrin 3 Rabbit anti-Human Polyclonal Antibody;カタログ番号LS-C55548-100;LS Bio社の抗体等)とMMAEのコンジュゲートを使用した場合の軸索量と本発明の抗体とのコンジュゲートを使用した場合の軸索量を比較することにより、本発明の抗体による標的化効果を確認することができる。本発明の抗体を使用した場合、例えば、軸索量がポリクローナル抗体を使用した場合より減少する。例えば、統計学上有意に減少すればよいが、具体的には、平均で軸索量がポリクローナル抗体を使用した場合の95%以下、90%以下となり得る。 For example, the antibody of the present invention can exhibit excellent physiological activity in motor neurons in the form of a conjugate with a physiologically active substance. The improvement of physiological activity based on the use of the antibody of the present invention can be confirmed, for example, by using as an indicator the results obtained by administering monomethyl auristatin E (MMAE) and the conjugate of the present invention to motor neurons in which axonal elongation and presynaptic formation have been induced by co-culture with microbeads having LRRTM molecules immobilized on the surface in advance. In this case, the targeting effect of the antibody of the present invention can be confirmed by comparing the amount of axons when a conjugate of MMAE with a polyclonal goat anti-SYNGR3 intravesicular domain antibody (Synaptogyrin 3 Rabbit anti-Human Polyclonal Antibody; catalog number LS-C55548-100; antibody of LS Bio, etc.) is used with the amount of axons when a conjugate of the antibody of the present invention is used. When the antibody of the present invention is used, for example, the amount of axons is reduced compared to when a polyclonal antibody is used. For example, a statistically significant reduction would be sufficient; specifically, the average axon volume could be 95% or less, or 90% or less, of the amount observed when a polyclonal antibody was used.
<核酸分子>
本発明は、本発明の抗体をコードする塩基配列を有する核酸分子も提供する。本発明の核酸分子は、本発明の抗体のいずれか一以上をコードする核酸分子であればよい。そのような核酸分子の塩基配列は特に限定しない。例えば、コドン最適化した塩基配列や、5'末端側に開始コドン(ATG)が付加された塩基配列等が挙げられる。本発明の核酸分子はDNA、RNA又はそれらの組合せであってもよく、天然ヌクレオチドの他、修飾ヌクレオチド、非天然ヌクレオチド等を適宜含めることができる。
<Nucleic acid molecule>
The present invention also provides a nucleic acid molecule having a base sequence encoding the antibody of the present invention. The nucleic acid molecule of the present invention may be a nucleic acid molecule encoding any one or more of the antibodies of the present invention. The base sequence of such a nucleic acid molecule is not particularly limited. Examples include a codon-optimized base sequence and a base sequence with an initiation codon (ATG) added to the 5'-end. The nucleic acid molecule of the present invention may be DNA, RNA, or a combination thereof, and may contain natural nucleotides as well as modified nucleotides, non-natural nucleotides, etc., as appropriate.
本発明の核酸分子は、本発明の抗体をコードする核酸に加えて他の任意の構成要素を含むことができる。例えば、プロモーターをさらに含み、細胞内で本発明の抗体を発現可能な遺伝子発現ベクターとしてもよい。 The nucleic acid molecule of the present invention may contain any other components in addition to the nucleic acid encoding the antibody of the present invention. For example, it may be a gene expression vector that further contains a promoter and is capable of expressing the antibody of the present invention in a cell.
本明細書において「遺伝子発現ベクター」とは、遺伝子や遺伝子断片(以下「遺伝子等」と表記する)を発現可能な状態で含み、その遺伝子等の発現を制御できる発現単位を包含するベクターをいう。遺伝子発現ベクターは、プラスミドベクター、ウイルスベクター、ファージベクターであってもよい。例えば、遺伝子組換え操作の容易なプラスミドベクター、又は免疫細胞に容易に遺伝子を導入することできるウイルスベクターとすることができる。本発明におけるベクターには、必要に応じて、標識遺伝子(選択マーカー)、エンハンサー、ターミネーター、複製起点、及びポリAシグナル等を追加で含んでもよい。 In this specification, the term "gene expression vector" refers to a vector that contains genes or gene fragments (hereinafter referred to as "genes, etc.") in an expressible state and includes an expression unit that can control the expression of the genes, etc. The gene expression vector may be a plasmid vector, a virus vector, or a phage vector. For example, it may be a plasmid vector that is easy to manipulate for genetic recombination, or a virus vector that can easily introduce genes into immune cells. The vector in the present invention may additionally contain a marker gene (selection marker), an enhancer, a terminator, a replication origin, a polyA signal, etc., as necessary.
本明細書において「発現可能な状態」とは、プロモーターの制御下にあるプロモーター下流域に、発現すべき遺伝子等を配置していることをいう。 In this specification, "expressible state" refers to the placement of a gene to be expressed downstream of a promoter under the control of the promoter.
具体的なベクターの種類は特に限定しない。例えば、プラスミドベクターは、例えばPromega社のpCIベクター、pSIベクター、pcDNA3ベクター等の市販の哺乳動物細胞用発現ベクターや、哺乳動物細胞と大腸菌等の細菌間とで複製可能なシャトルベクターであってもよい。 The specific type of vector is not particularly limited. For example, the plasmid vector may be a commercially available expression vector for mammalian cells, such as Promega's pCI vector, pSI vector, or pcDNA3 vector, or a shuttle vector capable of replicating between mammalian cells and bacteria such as E. coli.
ウイルスベクターは、例えばレトロウイルスベクター(オンコレトロウイルスベクター、レンチウイルスベクター、及び偽型ベクターを含む)、アデノウイルスベクター、アデノ随伴ウイルス(AAV)ベクター、シミアンウイルスベクター、ワクシニアウイルスベクター、センダイウイルスベクター、エプスタイン-バーウイルス(EBV)ベクター、及びHSVベクター等のウイルスベクターが使用可能である。感染細胞内で自己複製しないように複製能を欠くウイルスベクターを使用してもよい。 As the viral vector, for example, a retroviral vector (including oncoretroviral vectors, lentiviral vectors, and pseudotype vectors), an adenoviral vector, an adeno-associated virus (AAV) vector, a simian virus vector, a vaccinia virus vector, a Sendai virus vector, an Epstein-Barr virus (EBV) vector, and an HSV vector can be used. A viral vector lacking replication ability so as not to self-replicate within an infected cell may also be used.
本明細書において「プロモーター」とは、遺伝子発現ベクターを導入した細胞において、下流(3'末端側)に配置された遺伝子等の発現を制御することのできる遺伝子発現調節領域である。プロモーターは、発現制御下にある遺伝子等を発現させる場所に基づいて、ユビキタスプロモーター(全身性プロモーター)と部位特異的プロモーターに分類することができる。ユビキタスプロモーターは、全細胞、すなわち宿主個体全体で対象とする遺伝子等(対象遺伝子等)の発現を制御するプロモーターである。また、部位特異的プロモーターは、特定の細胞又は組織でのみ対象遺伝子等の発現を制御するプロモーターである。本発明の遺伝子発現ベクターに含まれるプロモーターは、ユビキタスプロモーター又は部位特異的プロモーターのいずれであってもよいが、宿主細胞において発現を誘導できることが好ましい。 In this specification, a "promoter" refers to a gene expression regulatory region that can control the expression of a gene, etc., located downstream (3' end side) in a cell into which a gene expression vector has been introduced. Promoters can be classified into ubiquitous promoters (systemic promoters) and site-specific promoters based on the location where the gene, etc. under their expression control is expressed. A ubiquitous promoter is a promoter that controls the expression of a target gene, etc. (target gene, etc.) in all cells, i.e., the entire host individual. A site-specific promoter is a promoter that controls the expression of a target gene, etc. only in a specific cell or tissue. The promoter contained in the gene expression vector of the present invention may be either a ubiquitous promoter or a site-specific promoter, but it is preferable that it can induce expression in a host cell.
また、プロモーターは、発現の時期に基づいて構成的活性型プロモーター、発現誘導型プロモーター又は時期特異的活性型プロモーターに分類される。構成的活性型プロモーターは、細胞内で対象遺伝子等を恒常的に発現させることができる。発現誘導型プロモーターは、細胞内で対象遺伝子等の発現を任意の時期に誘導することができる。また、時期特異的活性型プロモーターは、細胞内で対象遺伝子等を発生段階の特定の時期にのみ発現誘導することができる。いずれのプロモーターも、宿主細胞内で対象遺伝子の過剰な発現をもたらし得ることから過剰発現型プロモーターと解することができる。本発明の遺伝子発現ベクターに含まれるプロモーターは、治療効果の長期持続性を可能とする構成的活性型プロモーターであることが好ましい。 Promoters are also classified into constitutively active promoters, expression-inducible promoters, and time-specifically active promoters based on the time of expression. Constitutively active promoters can constitutively express a target gene, etc. in a cell. Expression-inducible promoters can induce the expression of a target gene, etc. in a cell at any time. Time-specifically active promoters can induce the expression of a target gene, etc. in a cell only at a specific time during the developmental stage. Any of these promoters can be considered to be overexpression promoters, since they can cause excessive expression of a target gene in a host cell. The promoter contained in the gene expression vector of the present invention is preferably a constitutively active promoter, which allows for long-term persistence of the therapeutic effect.
本態様の遺伝子発現ベクターにおけるプロモーターは、本発明の抗体をコードする核酸を宿主細胞内で発現誘導できるプロモーターである。具体的には、例えば、CMVプロモーター(CMV-IEプロモーター)、SV40初期プロモーター、RSVプロモーター、EF1αプロモーター、Ubプロモーター、5' LTRプロモーター等が挙げられる。レトロウイルスベクターの場合、本発明の抗体をコードする核酸を5' LTRプロモーター下流に配置して、その遺伝子発現を誘導することができる。 The promoter in the gene expression vector of this embodiment is a promoter that can induce the expression of a nucleic acid encoding an antibody of the present invention in a host cell. Specific examples include a CMV promoter (CMV-IE promoter), an SV40 early promoter, an RSV promoter, an EF1α promoter, an Ub promoter, and a 5' LTR promoter. In the case of a retroviral vector, the nucleic acid encoding an antibody of the present invention can be placed downstream of the 5' LTR promoter to induce its gene expression.
<細胞>
さらに、本発明は、本発明の抗体を発現可能な細胞を提供する。
本発明の宿主細胞の種類は特に限定しない。例えば、細胞が由来する生物は特に限定しないが、例えば、標的化剤において後述する脊椎動物に由来する細胞を本発明の細胞として使用することができる。また、宿主細胞の細胞種は特に限定しない。例えば、外肺葉性の細胞、中胚葉性の細胞、内胚葉性の細胞、及びこれらの組合せであってもよい。例えば、上皮組織、結合組織、軟骨組織、骨組織、血液組織(リンパ組織を含む)、筋組織、神経組織、又はこれらの組合せの細胞であってもよい。また、本発明の細胞は、分化能を有さない細胞であっても、分化能を有する細胞(例えば、複能性細胞、多能性細胞等)であってもよい。分化能を有する細胞としては、具体的には、例えば、間葉系幹細胞、造血幹細胞、種々のがん細胞株、神経幹細胞、iPS細胞及びES細胞が挙げられる。
<Cells>
Furthermore, the present invention provides a cell capable of expressing the antibody of the present invention.
The type of the host cell of the present invention is not particularly limited. For example, the organism from which the cell originates is not particularly limited, but for example, cells derived from vertebrates described later in the targeting agent can be used as the cell of the present invention. In addition, the type of the host cell is not particularly limited. For example, it may be an ectodermal cell, a mesodermal cell, an endodermal cell, or a combination thereof. For example, it may be an epithelial tissue, a connective tissue, a cartilage tissue, a bone tissue, a blood tissue (including lymphatic tissue), a muscle tissue, a nerve tissue, or a combination thereof. In addition, the cell of the present invention may be a cell that does not have differentiation potential or a cell that has differentiation potential (for example, a multipotent cell, a pluripotent cell, etc.). Specific examples of cells that have differentiation potential include mesenchymal stem cells, hematopoietic stem cells, various cancer cell lines, neural stem cells, iPS cells, and ES cells.
本発明で用いる「多能性幹細胞」とは、自己複製能を有しin vitroにおいて培養することが可能で、かつ、個体を構成する細胞に分化しうる多分化能を有する細胞をいう。具体的には、胚性幹細胞(ES細胞)、胎児の始原生殖細胞由来の多能性幹細胞(GS細胞)、体細胞由来の人工多能性幹細胞(iPS細胞)等を挙げることができるが、本方法で好ましく用いられるのはヒト由来のiPS細胞又はES細胞である。 The term "pluripotent stem cells" used in the present invention refers to cells that have the ability to self-replicate, can be cultured in vitro, and have the multipotency to differentiate into cells that make up an individual. Specific examples include embryonic stem cells (ES cells), pluripotent stem cells (GS cells) derived from fetal primordial germ cells, and induced pluripotent stem cells (iPS cells) derived from somatic cells, but the cells preferably used in this method are human-derived iPS cells or ES cells.
ES細胞は、受精卵から取得することが多いが、受精卵以外、例えば、脂肪組織、胎盤、精巣細胞から取得することもでき、いずれのES細胞も本発明の対象である。受精卵以外からES細胞を作製する方法は報告されており(例えば、WO2003/046141)、それらの報告を適宜参照して用いることができる。 ES cells are often obtained from fertilized eggs, but they can also be obtained from sources other than fertilized eggs, such as adipose tissue, placenta, and testicular cells, and all ES cells are within the scope of the present invention. Methods for producing ES cells from sources other than fertilized eggs have been reported (e.g., WO2003/046141), and these reports can be used with appropriate reference.
iPS細胞は、体細胞に由来する人工的な幹細胞であって、特異的な再プログラム化因子を核酸又はタンパク質の形態で体細胞に導入することにより製造することができ、ES細胞とほぼ同等の特性(例えば、分化多能性及び自己複製に基づく増殖能)を示す。再プログラム化因子に含まれる遺伝子の例としては、Oct3/4、Sox2、Sox1、Sox3、Sox15、Sox17、Klf4、Klf2、c-Myc、N-Myc、L-Myc、Nanog、Lin28、Fbx15、ERas、ECAT15-2、Tcl1、beta-catenin、Lin28b、Sall1、Sall4、Esrrb、Nr5a2、Tbx3、Glis1、又はその組合せ等が挙げられる。iPS細胞を製造する方法については、多くの報告がなされており、これらの報告を参照し、それらを適宜変更して用いることができる。本方法で用いることができるiPS細胞は、好ましくはヒト由来のiPS細胞であり、例えば、ヒトの線維芽細胞由来のiPS細胞である。 iPS cells are artificial stem cells derived from somatic cells, and can be produced by introducing specific reprogramming factors into somatic cells in the form of nucleic acids or proteins. They exhibit properties similar to those of ES cells (e.g., pluripotency and proliferation ability based on self-renewal). Examples of genes contained in the reprogramming factors include Oct3/4, Sox2, Sox1, Sox3, Sox15, Sox17, Klf4, Klf2, c-Myc, N-Myc, L-Myc, Nanog, Lin28, Fbx15, ERas, ECAT15-2, Tcl1, beta-catenin, Lin28b, Sall1, Sall4, Esrrb, Nr5a2, Tbx3, Glis1, or combinations thereof. There have been many reports on methods for producing iPS cells, and these reports can be referred to and modified as appropriate for use. The iPS cells that can be used in this method are preferably human-derived iPS cells, for example, human fibroblast-derived iPS cells.
ヒトiPS細胞を末梢神経細胞へと分化誘導する方法は、多くの報告がなされており(例えば、Chambers, Stuart M., et al., Nature biotechnology 27.3 (2009): 275.)、これらの報告を参照し、適宜変更して用いることができる。あるいは、ヒトiPS細胞から分化誘導した様々な神経細胞が市販されており、例えば、FUJIFILM Cellular Dynamics, Inc.又はリプロセル(株)等から購入することができる。 Many methods for inducing differentiation of human iPS cells into peripheral nerve cells have been reported (e.g., Chambers, Stuart M., et al., Nature biotechnology 27.3 (2009): 275.), and these reports can be referred to and modified as appropriate for use. Alternatively, various nerve cells induced to differentiate from human iPS cells are commercially available and can be purchased, for example, from FUJIFILM Cellular Dynamics, Inc. or ReproCell Co., Ltd.
本発明の細胞は、本発明の抗体を発現可能である。好ましくは、本発明の細胞は本発明の核酸分子を含む。本発明の細胞は、本発明の核酸分子を複数コピー含むことができる。 The cell of the present invention is capable of expressing the antibody of the present invention. Preferably, the cell of the present invention comprises a nucleic acid molecule of the present invention. The cell of the present invention can comprise multiple copies of the nucleic acid molecule of the present invention.
<標的化剤>
本発明はまた、シナプトジャイリン3の小胞内ドメインに結合可能な本発明の抗体を含む、運動神経細胞への標的化剤(「本発明の標的化剤」と称する)に関する。
Targeting Agents
The present invention also relates to a targeting agent for motor neurons (referred to as the "targeting agent of the present invention"), which comprises an antibody of the present invention capable of binding to the intravesicular domain of synaptogyrin 3.
本明細書において、「標的化剤」とは、特定の物質を標的へと送達するための薬剤を指す。本明細書においては、標的化剤を用いることにより、標識物質及び/又は生理活性物質(「所望の物質」と称する)が運動神経細胞、特に運動神経細胞シナプスに輸送される。また、シナプス及びシナプス小胞への標的化においては、エンドサイトーシスによるシナプス小胞の回収に伴って細胞内、特にシナプス小胞内に取り込まれ、細胞体へと送達される。さらに、本発明の標的化剤により、輸送された物質が細胞質中で生理活性を発揮し得る。 As used herein, the term "targeting agent" refers to an agent for delivering a specific substance to a target. As used herein, the targeting agent is used to transport a labeled substance and/or a physiologically active substance (referred to as a "desired substance") to a motor neuron, particularly to a motor neuron synapse. In addition, in targeting to synapses and synaptic vesicles, the substance is taken up into the cell, particularly into synaptic vesicles, following the retrieval of synaptic vesicles by endocytosis, and is delivered to the cell body. Furthermore, the targeting agent of the present invention allows the transported substance to exert its physiological activity in the cytoplasm.
例えば、本発明の標的化剤がシナプス小胞に取り込まれると共に、輸送された所望の物質がシナプス小胞膜を透過し細胞質中に移行し得る。具体的には、例えば、生理活性物質が使用された場合、標的化剤中の少なくとも1つの生理活性物質が細胞質に移行し、核内の生体物質、細胞質中の所望の生体物質又は細胞質膜上の生体物質に作用することができる。生理活性物質が作用する生体物質は、細胞膜上又は細胞内に存在し得る物質であれば特に限定されず、例えば、タンパク質、核酸等の高分子化合物、脂質、糖、アミノ酸、ヌクレオチド等の低分子化合物、金属イオン等のイオン又は原子等のいずれでもよい。 For example, the targeting agent of the present invention may be taken up into synaptic vesicles, and the transported desired substance may permeate the synaptic vesicle membrane and migrate into the cytoplasm. Specifically, for example, when a physiologically active substance is used, at least one physiologically active substance in the targeting agent may migrate into the cytoplasm and act on a biological substance in the nucleus, a desired biological substance in the cytoplasm, or a biological substance on the cytoplasmic membrane. The biological substance on which the physiologically active substance acts is not particularly limited as long as it is a substance that can be present on a cell membrane or inside a cell, and may be, for example, any of polymeric compounds such as proteins and nucleic acids, low molecular compounds such as lipids, sugars, amino acids, and nucleotides, ions such as metal ions, or atoms, etc.
具体的には、例えば、核内の生体物質としては、DNA、RNA(mRNA、siRNA、miRNA等)、転写因子、核内受容体等が挙げられ、細胞質中の生体物質としては、上記核酸の他、細胞骨格、酵素、金属イオン等が挙げられる。細胞膜上の生体物質としては、例えば、細胞膜の脂質、細胞膜上受容体、受容体と共役している酵素及び細胞接着因子等が挙げられる。 Specific examples of biological substances in the nucleus include DNA, RNA (mRNA, siRNA, miRNA, etc.), transcription factors, and nuclear receptors, while biological substances in the cytoplasm include the above-mentioned nucleic acids as well as the cytoskeleton, enzymes, and metal ions. Examples of biological substances on the cell membrane include lipids in the cell membrane, receptors on the cell membrane, enzymes conjugated with receptors, and cell adhesion factors.
本明細書において、「運動神経細胞」とは、中枢からの刺激を効果器である骨格筋へと伝える神経細胞群を指す。運動神経細胞は、一般には、中枢神経細胞である一次運動神経細胞及び末梢神経細胞である二次運動神経細胞を含むが、本明細書における運動神経細胞は二次運動神経細胞である。 In this specification, "motor neurons" refers to a group of nerve cells that transmit stimuli from the central nervous system to skeletal muscles, which are effector organs. Motor neurons generally include primary motor neurons, which are central nerve cells, and secondary motor neurons, which are peripheral nerve cells, but the motor neurons in this specification are secondary motor neurons.
本明細書において、「二次運動神経細胞」とは、細胞体を脊髄前角や脳幹に有し、骨格筋との接合部まで軸索を伸ばす運動神経細胞を指す。本明細書における運動神経細胞としては、例えば、α運動神経細胞、β運動神経細胞、及びγ運動神経細胞等が含まれる。また、脊髄の前核に細胞体を有する脊髄神経細胞の他、動眼神経、滑車神経、外転神経、顔面神経、舌下神経等の一部の脳神経も含まれる。本明細書における運動神経細胞は、通常、アセチルコリンを神経伝達物質として分泌し、コリン作動性神経細胞に分類される。しかし、アセチルコリン以外の神経伝達物質を分泌する運動神経細胞であってもよい。本明細書における運動神経細胞が投射する筋細胞は骨格筋細胞である。 In this specification, the term "secondary motor neuron" refers to a motor neuron that has its cell body in the anterior horn of the spinal cord or the brain stem and extends its axon to the junction with skeletal muscle. In this specification, the motor neuron includes, for example, alpha motor neuron, beta motor neuron, and gamma motor neuron. In addition to spinal nerve cells that have their cell body in the anterior nucleus of the spinal cord, some cranial nerves such as the oculomotor nerve, trochlear nerve, abducens nerve, facial nerve, and hypoglossal nerve are also included. In this specification, the motor neuron usually secretes acetylcholine as a neurotransmitter and is classified as a cholinergic neuron. However, it may be a motor neuron that secretes a neurotransmitter other than acetylcholine. In this specification, the muscle cell to which the motor neuron projects is a skeletal muscle cell.
本明細書において、「骨格筋細胞」とは、骨格を動かす横紋筋を構成する細胞又はその表現型を有する細胞を指す。本明細書における骨格筋細胞は、骨に付着する筋肉細胞及び筋紡錘等の骨格筋に含まれるその他の筋肉細胞を広く含む。骨格筋の種類は特に限定しないが、例えば、横隔膜、外側広筋、内側広筋、大腿直筋、中間広筋、上腕二頭筋、前脛骨筋、後脛骨筋、腓腹筋、ヒラメ筋、三角筋、広背筋、胸鎖乳突筋、肋間筋、眼筋、顔面筋、舌筋、あぶみ骨筋等が挙げられる。また、本明細書における骨格筋細胞は、人工の幹細胞(iPS細胞及びES細胞等)及び/又は天然の幹細胞(間葉系幹細胞及び骨格筋幹細胞等)からin vitroで分化させた細胞等の培養骨格筋細胞も含む。 As used herein, "skeletal muscle cells" refers to cells that constitute striated muscles that move the skeleton or cells that have the same phenotype. Skeletal muscle cells in this specification broadly include muscle cells attached to bones and other muscle cells contained in skeletal muscles, such as muscle spindles. There is no particular limitation on the type of skeletal muscle, but examples include the diaphragm, vastus lateralis, vastus medialis, rectus femoris, vastus intermedius, biceps brachii, tibialis anterior, tibialis posterior, gastrocnemius, soleus, deltoid, latissimus dorsi, sternocleidomastoid, intercostal muscles, eye muscles, facial muscles, tongue muscles, and stapedius muscles. Skeletal muscle cells in this specification also include cultured skeletal muscle cells, such as cells differentiated in vitro from artificial stem cells (such as iPS cells and ES cells) and/or natural stem cells (such as mesenchymal stem cells and skeletal muscle stem cells).
本明細書において、細胞は、脊椎動物に由来する細胞であり得る。脊椎動物には、魚類、爬虫類、両生類、鳥類及び哺乳類が含まれる。具体的な哺乳類としては、例えば、霊長類(例えば、ヒト)が挙げられる。また細胞は、家畜(ニワトリ、ウマ、ウシ、ヒツジ、ヤギ、ブタ等)、愛玩動物(熱帯魚、トカゲ、イヌ、ネコ、ウサギ等)、実験動物(カエル、マウス、ラット、サル等)に由来する細胞であり得る。細胞は1種類の組織、個体、及び動物種に由来する必要はなく、複数種類の細胞の混合物であってもよい。また、細胞が由来する組織及び個体の健康状態は特に限定しない。 In this specification, the cells may be cells derived from vertebrates. Vertebrates include fish, reptiles, amphibians, birds, and mammals. Specific examples of mammals include primates (e.g., humans). The cells may also be cells derived from livestock (chickens, horses, cows, sheep, goats, pigs, etc.), pets (tropical fish, lizards, dogs, cats, rabbits, etc.), and laboratory animals (frogs, mice, rats, monkeys, etc.). The cells do not have to be derived from one type of tissue, individual, or animal species, but may be a mixture of multiple types of cells. Furthermore, there are no particular limitations on the health of the tissue and individual from which the cells are derived.
本発明の標的化剤によれば、所望の物質を運動神経細胞シナプスを介して運動神経細胞(例えば、運動神経細胞の軸索終末、軸索、軸索小丘、細胞体、樹状突起等)に標的化することができる。 The targeting agent of the present invention allows a desired substance to be targeted to a motor neuron (e.g., the axon terminal, axon, axon hillock, cell body, dendrites, etc. of a motor neuron) via the motor neuron synapse.
本明細書において、「シナプス」とは、神経細胞の軸索終末と、別の神経細胞の樹状突起(中枢神経系の場合)又は骨格筋、臓器等の細胞(末梢神経系の場合)との間に形成される、間隙を含む接合部を指す。 In this specification, "synapse" refers to a junction that includes a gap, which is formed between the axon terminal of a nerve cell and the dendrite of another nerve cell (in the case of the central nervous system) or the cells of skeletal muscles, organs, etc. (in the case of the peripheral nervous system).
本明細書において、シナプスは、化学的シナプス、例えば、興奮性シナプス、抑制性シナプス等であり得る。本明細書において、シナプスは、神経細胞と別の神経細胞との間に形成されるシナプス(例えば、神経細胞の軸索と別の神経細胞の樹状突起との間に形成されるシナプス)、又は神経細胞と他種細胞(筋細胞等)との間に形成されるシナプスであってもよいが、好ましくは、神経細胞のシナプス前部と骨格筋細胞上のシナプス後部により形成されるシナプス(「神経筋接合部」とも称される)である。 In this specification, a synapse may be a chemical synapse, such as an excitatory synapse, an inhibitory synapse, etc. In this specification, a synapse may be a synapse formed between a nerve cell and another nerve cell (e.g., a synapse formed between an axon of a nerve cell and a dendrite of another nerve cell), or a synapse formed between a nerve cell and a cell of another type (such as a muscle cell), but is preferably a synapse formed by a presynapse on a nerve cell and a postsynapse on a skeletal muscle cell (also called a "neuromuscular junction").
本明細書において、「シナプス前部」とは、シナプスにおいて神経細胞の軸索終末に形成される膨大した部分を指し、「シナプス後部」とは、別の神経細胞の樹状突起又は骨格筋、臓器等の他の細胞におけるシナプス前部に面する部分を指す。また、「シナプス間隙」とは、シナプス前部とシナプス後部の間の空間を指す。シナプスでは、シナプス前部中に存在するシナプス小胞内に蓄積された神経伝達物質がシナプス間隙に放出され、シナプス後部に存在する受容体に結合することでシグナルが伝達される。 In this specification, the term "presynapse" refers to the enlarged portion formed at the axon terminal of a nerve cell in a synapse, and the term "postsynapse" refers to the portion facing the presynapse in a dendrite of another nerve cell or in another cell such as a skeletal muscle or organ. Additionally, the term "synaptic cleft" refers to the space between the presynapse and the postsynapse. In a synapse, neurotransmitters accumulated in synaptic vesicles present in the presynapse are released into the synaptic cleft and bind to receptors present in the postsynapse, thereby transmitting a signal.
本明細書において、「シナプス小胞」とは、シナプス前部の神経細胞の細胞質中に存在する分泌小胞を指す。本明細書におけるシナプス小胞には、神経伝達物質をその内部に含み、刺激に応じて細胞膜と融合して神経伝達物質をシナプス間隙に放出する小胞のみならず、神経伝達物質の放出後にエンドサイトーシス(バルクエンドサイトーシスを含む)によって神経細胞中に回収された小胞も含まれる。 In this specification, "synaptic vesicles" refer to secretory vesicles present in the cytoplasm of presynaptic neurons. In this specification, synaptic vesicles include not only vesicles that contain neurotransmitters and fuse with the cell membrane in response to a stimulus to release the neurotransmitter into the synaptic cleft, but also vesicles that are retrieved into the neuron by endocytosis (including bulk endocytosis) after the release of the neurotransmitter.
本発明の標的化剤はシナプス間隙において、細胞膜上に露出したシナプトジャイリン3の小胞内ドメインに結合し、エンドサイトーシスによってシナプス小胞に取り込まれ、それにより運動神経細胞の細胞体等まで送達することができる。そのため、本発明の標的化剤によれば、所望の物質(標識物質及び/又は生理活性物質)を運動神経細胞の内部、特に運動神経細胞の細胞体へと標的化することができる。また、例えば、同様に本発明の標的化剤を脳脊髄液中に投与する場合は、中枢神経系におけるシナプトジャイリン3を細胞膜上に発現する神経細胞の内部、特にその細胞体へと所望の物質(標識物質及び/又は生理活性物質)を標的化することができる。 The targeting agent of the present invention binds to the intravesicular domain of synaptogyrin 3 exposed on the cell membrane in the synaptic cleft, is taken up into synaptic vesicles by endocytosis, and can be delivered to the cell body of a motor neuron, etc. Therefore, the targeting agent of the present invention can target a desired substance (a labeled substance and/or a physiologically active substance) to the inside of a motor neuron, particularly to the cell body of the motor neuron. Similarly, for example, when the targeting agent of the present invention is administered into cerebrospinal fluid, the desired substance (a labeled substance and/or a physiologically active substance) can be targeted to the inside of a neuron expressing synaptogyrin 3 on the cell membrane in the central nervous system, particularly to the cell body.
本発明の標的化剤は、シナプトジャイリン3の小胞内ドメインに結合可能な本発明の抗体に加え、所望の物質(標識物質及び/又は生理活性物質)をさらに含み得る。 The targeting agent of the present invention may further contain a desired substance (a labeling substance and/or a biologically active substance) in addition to the antibody of the present invention capable of binding to the intravesicular domain of synaptogyrin 3.
本明細書において、「標識物質」とは、その存在を検出可能なシグナルを発する物質を指す。標識物質としては、例えば、蛍光分子、化学発光物質等の特定の条件において光を発する発光性標識物質、光音響効果プローブ等の音波を発する発音性標識物質、又は放射性標識物質等が挙げられる。蛍光分子の例としては、特に限定するものではないが、蛍光タンパク質、フルオレセイン及びその誘導体、ピレン及びその誘導体、量子ドット等の蛍光分子が挙げられる。化学発光物質としては、例えばペルオキシダーゼ(HRP)、アルカリフォスファターゼ(ALP)等の酵素等が挙げられる。放射性標識物質としては、例えば14C、3H、125I等を含む試薬が挙げられる。光音響効果とは、光吸収に伴う断熱膨張によって熱弾性波が生じる現象をいい、この熱弾性波は音響波として検出することができる。光音響効果プローブとしては、例えば、インドシアニングリーン又はその誘導体、クルクミン誘導体、又はコリン誘導体等が挙げられる。使用する抗体、標識物質及び生理活性物質の吸光特性がわかっている場合には、必ずしも光音響効果用の標識物質を使用する必要はなく、例えば、発光性標識物質を光音響効果に基づいて検出してもよい。 In this specification, the term "labeling substance" refers to a substance that emits a signal that can detect its presence. Examples of labeling substances include luminescent labeling substances that emit light under specific conditions, such as fluorescent molecules and chemiluminescent substances, sound-emitting labeling substances that emit sound waves, such as photoacoustic effect probes, and radioactive labeling substances. Examples of fluorescent molecules include, but are not limited to, fluorescent proteins, fluorescein and its derivatives, pyrene and its derivatives, and quantum dots. Examples of chemiluminescent substances include enzymes such as peroxidase (HRP) and alkaline phosphatase (ALP). Examples of radioactive labeling substances include reagents containing 14 C, 3 H, 125 I, and the like. The photoacoustic effect refers to a phenomenon in which adiabatic expansion accompanying light absorption generates a thermoelastic wave, and this thermoelastic wave can be detected as an acoustic wave. Examples of photoacoustic effect probes include indocyanine green or its derivatives, curcumin derivatives, and choline derivatives. When the absorption characteristics of the antibody, labeling substance, and physiologically active substance used are known, it is not necessarily necessary to use a labeling substance for the photoacoustic effect. For example, a luminescent labeling substance may be detected based on the photoacoustic effect.
本明細書において、「生理活性物質」とは、生体又は細胞に対して直接的又は間接的に生理的効果を発揮し得る物質を指す。例えば、標的の運動神経細胞に生理的効果を発揮し得る低分子化合物、ペプチドやアプタマー等の機能性中分子、並びに抗体、酵素等のタンパク質及びDNA、RNA等の核酸等の生体高分子を含む高分子化合物等が挙げられる。例えば、シナプス形成促進剤、シナプス維持剤、筋肉増強剤又は神経細胞機能改変剤等の薬剤又はプロドラッグを生理活性物質として使用することができる。 In this specification, the term "biologically active substance" refers to a substance that can directly or indirectly exert a physiological effect on a living organism or a cell. Examples include low molecular weight compounds that can exert a physiological effect on a target motor neuron, functional medium molecules such as peptides and aptamers, and polymeric compounds including biopolymers such as proteins such as antibodies and enzymes, and nucleic acids such as DNA and RNA. For example, drugs or prodrugs such as synapse formation promoters, synapse maintenance agents, muscle strengthening agents, or nerve cell function modifiers can be used as bioactive substances.
「生理的効果」とは、タンパク質、DNA及びRNA等の生体分子における量的及び/又は質的な変化をもたらす効果を指す。生理的効果の結果として、例えば、生体、器官、組織、細胞等の機能や性質等が変化し得る。例えば、シナプス形成の促進若しくは抑制、神経機能の低下の改善若しくは予防、又は神経細胞の過活動の改善若しくは予防等の効果が得られる。 "Physiological effect" refers to an effect that brings about quantitative and/or qualitative changes in biological molecules such as proteins, DNA, and RNA. As a result of a physiological effect, for example, the functions and properties of living organisms, organs, tissues, cells, etc. may change. For example, effects such as promotion or inhibition of synapse formation, improvement or prevention of decline in neuronal function, or improvement or prevention of hyperactivity of nerve cells may be obtained.
本発明の標的化剤が所望の物質(標識物質及び/又は生理活性物質)を含む場合、本発明の抗体と所望の物質とが共有結合により連結していない状態で(例えば、非共有結合的に連結した状態で)、又は共有結合により連結した状態で含まれる。共有結合により連結した状態で含まれる場合、シナプトジャイリン3の小胞内ドメインに結合可能な本発明の抗体と所望の物質とはコンジュゲート(「本発明のコンジュゲート」と称する)を形成する。 When the targeting agent of the present invention contains a desired substance (a labeling substance and/or a physiologically active substance), the antibody of the present invention and the desired substance are not covalently linked (e.g., noncovalently linked) or are covalently linked. When the antibody of the present invention and the desired substance are covalently linked, the antibody of the present invention capable of binding to the intravesicular domain of synaptogyrin 3 and the desired substance form a conjugate (referred to as the "conjugate of the present invention").
本明細書において、「コンジュゲート」とは、2以上の分子が共有結合により連結された物質を指す。特に、本発明のコンジュゲートは、本発明の抗体と所望の物質(標識物質及び/又は生理活性物質)とが連結している。 As used herein, the term "conjugate" refers to a substance in which two or more molecules are covalently linked. In particular, the conjugate of the present invention is a substance in which the antibody of the present invention is linked to a desired substance (a labeling substance and/or a biologically active substance).
本発明の標的化剤における抗体と所望の物質との間の共有結合及び非共有結合は、本発明の抗体と所望の物質とが連結した状態で運動神経細胞の近傍まで到達可能な結合であれば特に限定しない。 The covalent and non-covalent bonds between the antibody and the desired substance in the targeting agent of the present invention are not particularly limited, so long as the antibody of the present invention and the desired substance can reach the vicinity of the motor neuron in a linked state.
本明細書において、「シナプス形成促進剤」とは、シナプス前部及び/又はシナプス後部の形成を促進する作用を有する薬剤を指す。シナプス形成促進には、例えば、シナプスの接合力を増強すること、例えば、(i)シナプス前部及び/若しくは後部の表面積及び/若しくは体積の増加、(ii)シナプス前部に特異的に発現するタンパク質(例えば、シナプシン1又はシナプシン2等)の量、密度、集積速度、集積頻度等の増加及び/若しくは定性的な変化、(iii)シナプス後部に特異的に発現するタンパク質(例えば、LRRTMファミリータンパク質)の量、密度、集積速度、集積頻度等の増加及び/若しくは定性的な変化をもたらすことが含まれる。 In this specification, the term "synapse formation promoter" refers to a drug that has the effect of promoting the formation of the presynapse and/or postsynapse. Promotion of synapse formation includes, for example, enhancing the strength of synapse connections, for example, (i) increasing the surface area and/or volume of the presynapse and/or postsynapse, (ii) increasing and/or qualitatively changing the amount, density, accumulation rate, accumulation frequency, etc. of a protein specifically expressed in the presynapse (e.g., synapsin 1 or synapsin 2, etc.), and (iii) increasing and/or qualitatively changing the amount, density, accumulation rate, accumulation frequency, etc. of a protein specifically expressed in the postsynapse (e.g., LRRTM family proteins).
本明細書において、「シナプス維持剤」とは、シナプス前部及び/又はシナプス後部の退縮を抑制する又はそれを補助する作用を有する薬剤を指す。シナプス維持には、例えば、シナプスの接合力の減弱を抑制する又はそれを補助すること、例えば、(i)シナプス前部及び/若しくは後部の表面積及び/若しくは体積の減少の抑制又はその補助、(ii)シナプス前部に特異的に発現するタンパク質(例えば、シナプシン1又はシナプシン2等)の量、密度、集積速度、集積頻度等の減少及び/若しくは定性的な変化の抑制又はその補助、(iii)シナプス後部に特異的に発現するタンパク質(例えば、LRRTMファミリータンパク質)の量、密度、集積速度、集積頻度等の減少及び/若しくは定性的な変化の抑制又はその補助をもたらすことが含まれる。 In this specification, the term "synapse maintenance agent" refers to a drug that has the effect of suppressing or assisting in the degeneration of the presynapse and/or postsynapse. Synapse maintenance includes, for example, suppressing or assisting in the weakening of synaptic junction strength, for example, (i) suppressing or assisting in the reduction of the surface area and/or volume of the presynapse and/or postsynapse, (ii) suppressing or assisting in the reduction and/or qualitative change of the amount, density, accumulation rate, accumulation frequency, etc. of a protein specifically expressed in the presynapse (e.g., synapsin 1 or synapsin 2, etc.), and (iii) suppressing or assisting in the reduction and/or qualitative change of the amount, density, accumulation rate, accumulation frequency, etc. of a protein specifically expressed in the postsynapse (e.g., LRRTM family proteins).
本明細書において、「筋肉増強剤」とは、筋肉の増強若しくは減弱の抑制をもたらす作用又はそれを促進する作用を有する薬剤を指す。筋肉の増強の種類は、筋肉の機能が強化される限り特に限定しないが、例えば、筋肉の表面積及び/若しくは体積の増加、筋束、筋線維、筋原線維、筋節、筋細胞等の筋肉を構成する各要素の数、密度等の増加及び/若しくは定性的な変化、並びに/又は筋肉を構成する細胞中の特定のタンパク質の発現量の変化をもたらす作用、筋量及び/若しくは筋力(例えば骨格筋の筋量若しくは筋力)を増加させる作用、又はこれらの作用により筋肉の減弱の抑制をもたらす作用等が挙げられる。 In this specification, the term "muscle-enhancing agent" refers to a drug that has the effect of enhancing muscle or inhibiting weakening, or the effect of promoting such enhancement. The type of muscle enhancement is not particularly limited as long as the muscle function is enhanced, but examples include an increase in muscle surface area and/or volume, an increase and/or qualitative change in the number, density, etc. of each element that constitutes muscle, such as muscle bundles, muscle fibers, myofibrils, sarcomeres, muscle cells, and/or an effect of causing a change in the expression level of a specific protein in the cells that constitute muscle, an effect of increasing muscle mass and/or muscle strength (e.g., skeletal muscle mass or strength), or an effect of inhibiting muscle weakening through these effects.
具体的なシナプス形成促進剤、シナプス維持剤及び筋肉増強剤としては、限定するものではないが、例えば、本発明者らが見出した、特開2022-053535において開示される化合物(例えば、チアミン及びその誘導体)、特開2023-028848において開示される化合物(例えば、アトロピン、ブスルファン、クロモカルブ、プロカインアミド、ウデナフィル、プロピフェナゾン及びそれらの誘導体)等が挙げられる。 Specific examples of synapse formation promoters, synapse maintenance agents, and muscle strengthening agents include, but are not limited to, compounds discovered by the present inventors and disclosed in JP 2022-053535 (e.g., thiamine and its derivatives), and compounds disclosed in JP 2023-028848 (e.g., atropine, busulfan, chromocarb, procainamide, udenafil, propyphenazone, and their derivatives).
本明細書において、「神経細胞機能改変剤」とは、神経細胞が発揮する機能を変化させる作用又は促進する作用を有する薬剤を指す。神経細胞の機能改変は、神経細胞の機能の程度及び/又は性質が変化する限り特に限定しないが、例えば、神経細胞の電気生理学的特性(刺激の伝導及び伝達の特性等)の変化、遺伝子発現様式の変化、並びに形態学的な特性の変化(神経突起の伸長、退縮及び分岐、並びにシナプスの形成及び退縮等)等が含まれる。 In this specification, the term "neuron function modifying agent" refers to an agent that has the effect of changing or promoting the function exerted by a neuron. The modification of a neuron's function is not particularly limited as long as it changes the degree and/or nature of the neuron's function, but includes, for example, changes in the electrophysiological properties of the neuron (such as the properties of conduction and transmission of stimuli), changes in gene expression patterns, and changes in morphological properties (such as the extension, retraction, and branching of neurites, and the formation and retraction of synapses).
具体的な機能改変剤としては、限定するものではないが、例えば、AP-1阻害剤(例えば、本発明者らが見出したWO2020/196725に開示される化合物等)、FUS阻害剤、SOD1阻害剤、TDP-43阻害剤(例えば、アナカルジン酸の化合物等)、KIF1A阻害剤、その他の微小管重合阻害剤(モノメチルアウリスタチンE(MMAE)、モノメチルアウリスタチンF、アウリスタチンPE等のアウリスタチン系薬剤等を含む)等の細胞骨格改変剤等が挙げられる。 Specific examples of function modifying agents include, but are not limited to, cytoskeleton modifying agents such as AP-1 inhibitors (e.g., compounds disclosed in WO2020/196725 discovered by the present inventors), FUS inhibitors, SOD1 inhibitors, TDP-43 inhibitors (e.g., anacardic acid compounds), KIF1A inhibitors, and other microtubule polymerization inhibitors (including auristatin drugs such as monomethylauristatin E (MMAE), monomethylauristatin F, and auristatin PE).
本明細書において、「細胞骨格改変剤」とは、細胞骨格の形成、維持、分解、分岐、走行及び局在からなる群から選択される一以上を抑制及び/又は促進する薬剤を指す。本明細書における細胞骨格改変剤には、細胞骨格以外の分子に作用することにより細胞骨格の形成等を改変する薬剤も含まれる。細胞骨格には、微小管、中間径フィラメント及びアクチンフィラメントのいずれも含まれる。例えば、細胞骨格改変剤として、細胞骨格の形成、維持を抑制する薬剤、具体的には、例えば、微小管の重合阻害剤等を使用することができる。 As used herein, the term "cytoskeleton modifying agent" refers to an agent that inhibits and/or promotes one or more selected from the group consisting of the formation, maintenance, degradation, branching, running, and localization of the cytoskeleton. The cytoskeleton modifying agent used herein also includes agents that modify the formation, etc. of the cytoskeleton by acting on molecules other than the cytoskeleton. The cytoskeleton includes microtubules, intermediate filaments, and actin filaments. For example, an agent that inhibits the formation and maintenance of the cytoskeleton, specifically, for example, a microtubule polymerization inhibitor, etc., can be used as a cytoskeleton modifying agent.
これらの薬剤は、神経細胞に対する作用を有していればよく、運動神経細胞特異的な作用を有する必要はない。 These drugs only need to have an effect on nerve cells, and do not need to have an effect specific to motor neurons.
本発明のコンジュゲートにおいては、本発明の抗体と所望の物質(標識物質及び/又は生理活性物質)とが直接的に共有結合で連結していてもよく、それらがリンカー等を介して間接的に連結していてもよい。 In the conjugate of the present invention, the antibody of the present invention and the desired substance (the labeling substance and/or the physiologically active substance) may be directly linked by a covalent bond, or they may be indirectly linked via a linker or the like.
本発明の標的化剤がコンジュゲートを含まない場合、所望の物質は、本発明の抗体と結合可能な部分を有することが好ましい。具体的には、例えば、本発明の抗体に結合可能な別の抗体と共有結合により結合している所望の物質を使用することができる。この場合の「本発明の抗体に結合可能な別の抗体」の具体的な構成は、抗原が本発明の抗体であり、ポリクローナル抗体であってもよい以外は「<本発明の抗体>」の項における記載に準ずる。 When the targeting agent of the present invention does not include a conjugate, the desired substance preferably has a portion capable of binding to the antibody of the present invention. Specifically, for example, a desired substance that is covalently bound to another antibody capable of binding to the antibody of the present invention can be used. In this case, the specific configuration of the "another antibody capable of binding to the antibody of the present invention" is as described in the "<Antibody of the present invention>" section, except that the antigen is the antibody of the present invention and may be a polyclonal antibody.
本発明の抗体に結合可能な部分と所望の物質との結合は、本発明の標的化剤又はコンジュゲートにおける結合に準ずる。したがって、本発明の抗体に結合可能な部分と所望の物質とは非共有結合により連結していてもよく、直接的に共有結合で連結していてもよく、リンカー等を介して間接的に連結していてもよい。 The binding between the moiety capable of binding to the antibody of the present invention and the desired substance is similar to the binding in the targeting agent or conjugate of the present invention. Therefore, the moiety capable of binding to the antibody of the present invention and the desired substance may be linked by a non-covalent bond, may be directly linked by a covalent bond, or may be indirectly linked via a linker or the like.
本発明の標的化剤が所望の物質を含む場合、所望の物質と本発明の抗体とが非共有結合的に連結しているペプチド複合体の形で、所望の物質を本発明の標的化剤に含めることができる。 When the targeting agent of the present invention contains a desired substance, the desired substance can be included in the targeting agent of the present invention in the form of a peptide complex in which the desired substance is non-covalently linked to the antibody of the present invention.
所望の物質が本発明の抗体に結合する部位は、本発明の抗体のSYNGR3の小胞内ドメインへの結合を損なわない限り特に限定しない。具体的には、例えば、高頻度可変領域(HVR)以外の部位又は定常領域に所望の物質を結合することができる。 The site at which the desired substance binds to the antibody of the present invention is not particularly limited, so long as it does not impair the binding of the antibody of the present invention to the intravesicular domain of SYNGR3. Specifically, for example, the desired substance can be bound to a site other than the hypervariable region (HVR) or to the constant region.
また、本発明の標的化剤は、互いの機能を損なわない限り、所望の物質を複数含んでもよい。この場合、例えば、同じ物質を複数含んでもよく、互いに異なる物質を1つずつ又は複数含んでもよい。 The targeting agent of the present invention may contain multiple desired substances, so long as the functions of each substance are not impaired. In this case, for example, the targeting agent may contain multiple substances of the same substance, or may contain one or multiple substances that are different from each other.
本発明において利用可能なリンカーとしては、当分野において好適に使用されるリンカーを適宜使用することができる。この場合、リンカーの構成及び鎖長は、得られるコンジュゲートの機能を損なわない範囲で適切なものを選択することができる。リンカーは、例えば、シナプスへの輸送後に切断可能なように構成されてもよい。また、リンカーは、例えば、シナプスへの輸送後に切断されないように構成されてもよい。 Linkers that are suitable for use in the present invention may be any linkers that are suitable for use in the field. In this case, the structure and chain length of the linker may be appropriately selected within a range that does not impair the function of the resulting conjugate. The linker may be configured, for example, so that it can be cleaved after transport to the synapse. The linker may also be configured, for example, so that it cannot be cleaved after transport to the synapse.
リンカーは、当分野で通常使用されるものであればいずれでも良く、特に限定するものではないが、例えば5~25個、好ましくは10~20個のアミノ酸残基からなるペプチドリンカー、例えばGSリンカー等を好適に使用することができる。また、例えば、酸不安定性リンカー、光不安定性リンカー、ペプチダーゼ感受性リンカー、ジメチルリンカー又はジスルフィド含有リンカー等の切断可能なリンカーを使用してもよい。 The linker may be any linker commonly used in the art, and is not particularly limited. For example, a peptide linker consisting of 5 to 25, preferably 10 to 20, amino acid residues, such as a GS linker, may be preferably used. In addition, a cleavable linker, such as an acid-labile linker, a photolabile linker, a peptidase-sensitive linker, a dimethyl linker, or a disulfide-containing linker, may also be used.
本発明の標的化剤に含まれる本発明の抗体は、シナプス小胞と細胞膜の融合により一時的に細胞表面に露出したシナプトジャイリン3の小胞内ドメインに結合し、シナプス小胞のエンドサイトーシスに伴って、シナプトジャイリン3と一緒に細胞内に送達され得る。したがって、標的化剤又はコンジュゲートは、標的となるシナプスのシナプス小胞に所望の物質が送達されるように設計されることが好ましい。シナプス小胞に送達され得る化合物の特徴は、当技術分野において周知である。本発明の標的化剤又はコンジュゲートの粒子径は、バルクエンドサイトーシスのエンドソームの直径が90nm~160nmであることから、その平均値(又は中央値)を、例えば、160nm以下、150nm以下、140nm以下、130nm以下、120nm以下、110nm以下、100nm以下、90nm以下とすることができる。また、シナプス小胞の直径が40nm~60nmであることから、本発明のコンジュゲートの粒子径は、その平均値(又は中央値)を、例えば、60nm以下、55nm以下、50nm以下、45nm以下、40nm以下、35nm以下、30nm以下、25nm以下、23nm以下、20nm以下、18nm以下、15nm以下、14nm以下、13nm以下、12nm以下とすることができる。例えば、標的化剤が所望の物質を含まない場合は、標的化剤に所望の物質が結合した際の全体の粒子径を上述の範囲とすることができる。ただし、例えば、シナプス小胞のエンドサイトーシスを阻害する目的で、又はシナプス表面若しくはシナプス間隙に物質を送達する目的で粒子径を大きく設計してもよい。また、例えば、シナプス小胞への送達の際には既に所望の物質が分離されているように設計する場合には、分離前の粒子径を大きく設計してもよい。 The antibody of the present invention contained in the targeting agent of the present invention binds to the intravesicular domain of synaptogyrin 3 that is temporarily exposed on the cell surface due to fusion of synaptic vesicles with the cell membrane, and can be delivered into the cell together with synaptogyrin 3 along with endocytosis of synaptic vesicles. Therefore, it is preferable that the targeting agent or conjugate is designed so that the desired substance is delivered to the synaptic vesicles of the target synapse. The characteristics of compounds that can be delivered to synaptic vesicles are well known in the art. Since the diameter of endosomes in bulk endocytosis is 90 nm to 160 nm, the average (or median) particle size of the targeting agent or conjugate of the present invention can be, for example, 160 nm or less, 150 nm or less, 140 nm or less, 130 nm or less, 120 nm or less, 110 nm or less, 100 nm or less, or 90 nm or less. In addition, since the diameter of synaptic vesicles is 40 nm to 60 nm, the average particle diameter (or median) of the conjugate of the present invention can be, for example, 60 nm or less, 55 nm or less, 50 nm or less, 45 nm or less, 40 nm or less, 35 nm or less, 30 nm or less, 25 nm or less, 23 nm or less, 20 nm or less, 18 nm or less, 15 nm or less, 14 nm or less, 13 nm or less, or 12 nm or less. For example, when the targeting agent does not contain the desired substance, the overall particle diameter when the desired substance is bound to the targeting agent can be in the above-mentioned range. However, for example, the particle diameter may be designed to be large for the purpose of inhibiting endocytosis of synaptic vesicles or for the purpose of delivering a substance to the synapse surface or synaptic cleft. In addition, for example, when designed so that the desired substance is already separated at the time of delivery to the synaptic vesicles, the particle diameter before separation may be designed to be large.
本発明のコンジュゲート又は標的化剤は、血液脳関門を通過するように構成されてもよいが、必ずしもそのような構成である必要は無い。通常は、本発明のコンジュゲート又は標的化剤は、血液脳関門を通過せず、そのため、中枢神経系には作用せず、末梢に存在するシナプスにおいてのみ作用し得る。 The conjugates or targeting agents of the present invention may be configured to pass through the blood-brain barrier, but are not necessarily configured in this way. Typically, the conjugates or targeting agents of the present invention do not pass through the blood-brain barrier, and therefore do not act on the central nervous system, but only on synapses present in the periphery.
シナプスを介した運動神経細胞への標的化作用は、例えば、生理活性物質を含む本発明のコンジュゲート又は標的化剤を脊椎動物等の対象(例えば、非ヒト哺乳動物、ヒト、その他脊椎動物)に投与し、本発明のコンジュゲート又は標的化剤による、対象の神経細胞における生理的効果を評価することにより判定することができる。生理的効果の評価は、例えば、本発明のコンジュゲート若しくは標的化剤を投与した群と投与していない群との生理的効果の程度を比較することにより、及び/又は本発明のコンジュゲート若しくは標的化剤を投与した群と生理活性物質を単体で投与した群の生理的効果の程度を比較することにより、行うことができる。 The targeting effect on motor neurons via synapses can be determined, for example, by administering a conjugate or targeting agent of the present invention containing a physiologically active substance to a subject such as a vertebrate (e.g., a non-human mammal, a human, or another vertebrate) and evaluating the physiological effect of the conjugate or targeting agent of the present invention on the subject's neurons. The physiological effect can be evaluated, for example, by comparing the degree of physiological effect between a group administered with the conjugate or targeting agent of the present invention and a group not administered with the conjugate or targeting agent of the present invention, and/or by comparing the degree of physiological effect between a group administered with the conjugate or targeting agent of the present invention and a group administered with the physiologically active substance alone.
本発明者等は、先に、神経細胞を、LRRTM分子(LRRTM2の細胞外ドメイン等)が表面に固定されたマイクロビーズと共に共培養することにより、シナプス前部の形成を誘導することができることを見出している(WO2021/006075)。 The inventors have previously found that presynaptic formation can be induced by co-culturing nerve cells with microbeads having LRRTM molecules (such as the extracellular domain of LRRTM2) immobilized on their surfaces (WO2021/006075).
したがって、被験物質がシナプスを含む運動神経細胞への標的化作用を有するか否かはまた、神経細胞とマイクロビーズとの共培養により誘導されたシナプス前部に被験物質が局在するか否かを調べることにより判定することもできる。 Therefore, whether or not a test substance has a targeting effect on motor neurons containing synapses can also be determined by examining whether or not the test substance is localized at the presynapse induced by co-culturing neurons with microbeads.
「LRRTM(ロイシンリッチリピート膜貫通型神経タンパク質、leucine-rich repeat transmembrane neuronal protein)ファミリータンパク質」とは、LRRTMファミリーに属するタンパク質を指す。LRRTMファミリーは、シナプス後部側のシナプスオーガナイザータンパク質ファミリーの1つであり、シナプス前部の形成を誘導する活性を有する。ヒトを含む哺乳類では、LRRTMファミリータンパク質として、LRRTM1、LRRTM2、LRRTM3、及びLRRTM4の4種類が報告されている。マイクロビーズに使用されるLRRTMファミリータンパク質はそのいずれであってもよい。 "LRRTM (leucine-rich repeat transmembrane neuronal protein) family protein" refers to proteins belonging to the LRRTM family. The LRRTM family is a family of synaptic organizer proteins on the postsynaptic side, and has the activity of inducing the formation of presynapses. In mammals, including humans, four types of LRRTM family proteins have been reported: LRRTM1, LRRTM2, LRRTM3, and LRRTM4. The LRRTM family protein used in the microbeads may be any of these.
<コンジュゲート>
本発明は、シナプトジャイリン3の小胞内ドメインに結合可能な本発明の抗体と、所望の物質(標識物質及び/又は生理活性物質)とのコンジュゲートに関する。本発明のコンジュゲートは、例えば、運動神経細胞のシナプス小胞内に取り込まれ、運動神経細胞に送達される。
<Conjugate>
The present invention relates to a conjugate of a desired substance (a labeling substance and/or a physiologically active substance) with the antibody of the present invention capable of binding to the intravesicular domain of synaptogyrin 3. The conjugate of the present invention is, for example, taken up into synaptic vesicles of a motor neuron and delivered to the motor neuron.
<運動神経細胞又はそのシナプスの可視化剤>
本発明は、運動神経細胞又はそのシナプスの可視化剤である標的化剤(「本発明の可視化剤」と称する)を提供する。
<Visualization agent for motor neurons or their synapses>
The present invention provides a targeting agent (hereinafter referred to as the "visualizing agent of the present invention") that is an agent for visualizing motor neurons or their synapses.
本発明の可視化剤は、運動神経細胞又はそのシナプスを可視化する用途に使用するための、標識物質を含む標的化剤である。 The visualization agent of the present invention is a targeting agent containing a labeling substance for use in visualizing motor neurons or their synapses.
「運動神経細胞を可視化する」とは、運動神経細胞の全体又は一部分を検出可能な状態にすることを指す。運動神経細胞の一部分を可視化する場合、可視化される部分は無作為であっても、所定の部分であってもよい。例えば、所定の部分としてシナプスを可視化することができる。その場合、本発明の可視化剤をシナプス可視化剤として利用することができる。「シナプスを可視化する」とは、シナプス前部及び/又はシナプス後部を検出可能な状態にすることを指す。そのため、本発明の可視化剤には、直接目視により検出可能な標識物質の他、検出可能な任意の標識物質を使用することができる。同様に、本発明の可視化剤は、運動神経細胞の軸索終末、軸索、軸索小丘、細胞体、樹状突起等を可視化することができる。 "Visualizing a motor neuron" refers to making the entire or a part of a motor neuron detectable. When visualizing a part of a motor neuron, the part that is visualized may be random or a predetermined part. For example, a synapse can be visualized as the predetermined part. In that case, the visualization agent of the present invention can be used as a synapse visualization agent. "Visualizing a synapse" refers to making a presynapse and/or a postsynapse detectable. Therefore, the visualization agent of the present invention can use any detectable labeling substance in addition to a labeling substance that can be detected directly by visual inspection. Similarly, the visualization agent of the present invention can visualize the axon terminal, axon, axon hillock, cell body, dendrites, etc. of a motor neuron.
本発明の可視化剤は、in vivo又はin vitroにおいて使用され得る。標識物質のシグナルの検出は、運動神経細胞が生きている間に、又は運動神経細胞を固定した後に行うことができる。運動神経細胞が生きた状態での検出に適した標識物質は当技術分野において公知である。例えば、in vivoイメージングの分野において公知の蛍光物質、化学若しくは生物発光物質等の発光性物質、光音響効果プローブ等の発音性物質、放射性同位体等の放射性物質又は造影剤等が挙げられる。 The visualization agent of the present invention may be used in vivo or in vitro. Detection of the signal of the labeling substance may be performed while the motor neurons are alive or after the motor neurons are fixed. Labeling substances suitable for detection of live motor neurons are known in the art. Examples include fluorescent substances known in the field of in vivo imaging, luminescent substances such as chemo- or bioluminescent substances, sound-emitting substances such as photoacoustic probes, radioactive substances such as radioisotopes, or contrast agents.
本発明の可視化剤は、任意の用途、例えば、運動神経細胞又はシナプス(神経筋接合部を含む)若しくはシナプス小胞の数、サイズ若しくは位置を可視化するため、又は外科手術若しくは診断において組織を可視化するために使用され得る。 The visualization agent of the present invention may be used for any application, for example, to visualize the number, size or location of motor neurons or synapses (including neuromuscular junctions) or synaptic vesicles, or to visualize tissue in surgery or diagnosis.
本発明の可視化剤は、他の試薬等、例えば、本発明の可視化剤に含まれる標識物質の検出のために必要な試薬等と共にキットの形態で提供されてもよい。 The visualization agent of the present invention may be provided in the form of a kit together with other reagents, such as reagents necessary for detecting the labeling substance contained in the visualization agent of the present invention.
特に、例えば、標識物質が酵素等である場合、その基質を本発明の可視化剤と共に提供することができる。 In particular, for example, when the labeling substance is an enzyme, its substrate can be provided together with the visualization agent of the present invention.
<組成物又は医薬組成物>
本発明はさらに、本発明のコンジュゲート又は標的化剤を含む、組成物(「本発明の組成物」と称する)又は医薬組成物(「本発明の医薬組成物」と称する)に関する。
<Composition or pharmaceutical composition>
The present invention further relates to a composition (referred to as "the composition of the present invention") or a pharmaceutical composition (referred to as "the pharmaceutical composition of the present invention") comprising a conjugate or targeting agent of the present invention.
本発明の組成物又は医薬組成物は、生理活性物質を含む本発明の標的化剤を含む。本発明の組成物又は医薬組成物は、標的化剤の他に、必要に応じて、添加剤(例えば、担体(固体や液体担体等)、賦形剤、界面活性剤、結合剤、崩壊剤、滑沢剤、溶解補助剤、懸濁化剤、コーティング剤、着色剤、保存剤、緩衝剤、pH調整剤)等を含んでもよい。このとき、添加剤は、組成物又は医薬組成物の剤形に応じて適宜選択することができる。 The composition or pharmaceutical composition of the present invention includes the targeting agent of the present invention, which contains a physiologically active substance. In addition to the targeting agent, the composition or pharmaceutical composition of the present invention may also include additives (e.g., carriers (solid or liquid carriers, etc.), excipients, surfactants, binders, disintegrants, lubricants, solubilizing agents, suspending agents, coating agents, colorants, preservatives, buffers, pH adjusters), etc., as necessary. In this case, the additives can be appropriately selected depending on the dosage form of the composition or pharmaceutical composition.
本発明の組成物又は医薬組成物は、限定するものではないが、例えば固形製剤、液体製剤、ジェル剤、エアロゾル剤等の任意の剤形に調製されたものであってよい。なお、組成物又は医薬組成物を液体製剤として用いる場合には、それを使用する直前に、例えば、生理食塩水で再構成することを意図した乾燥物として調製することもできる。 The composition or pharmaceutical composition of the present invention may be prepared in any dosage form, including, but not limited to, a solid formulation, a liquid formulation, a gel formulation, an aerosol formulation, etc. When the composition or pharmaceutical composition is used as a liquid formulation, it can also be prepared as a dry product intended to be reconstituted with, for example, physiological saline immediately before use.
賦形剤としては、例えば、乳糖、結晶セルロース、デンプン等が挙げられる。結合剤としては、例えば、デンプン糊、アラビアゴム糊、ヒドロキシプロピルセルロース等が挙げられる。崩壊剤としては、例えば、デンプン、セルロース類、炭酸塩等が挙げられる。滑沢剤としては、例えば、ワックス、タルク等である。 Examples of excipients include lactose, crystalline cellulose, and starch. Examples of binders include starch paste, gum arabic paste, and hydroxypropyl cellulose. Examples of disintegrants include starch, celluloses, and carbonates. Examples of lubricants include wax and talc.
本発明の組成物又は医薬組成物は、シナプス形成促進剤、シナプス維持剤、筋肉増強剤又は神経細胞機能改変剤を生理活性物質として含む場合、シナプス形成を促進することにより神経機能の低下を改善又は予防することができる。したがって、本発明の組成物又は医薬組成物は、神経機能の低下、例えば、神経の損傷による神経機能の低下、老化による神経機能の低下、若しくは疾患による神経機能の低下等を改善若しくは予防するため、又は神経機能の向上のために使用することができる。 When the composition or pharmaceutical composition of the present invention contains a synapse formation promoter, a synapse maintenance agent, a muscle-enhancing agent, or a nerve cell function modifier as a physiologically active substance, it can improve or prevent a decline in nerve function by promoting synapse formation. Therefore, the composition or pharmaceutical composition of the present invention can be used to improve or prevent a decline in nerve function, such as a decline in nerve function due to nerve damage, a decline in nerve function due to aging, or a decline in nerve function due to disease, or to improve nerve function.
本明細書において、「神経の損傷」とは、神経の任意の箇所での損傷を指し、体外から物理的に与えられた損傷、及び、がん、腫瘍等の体内の要因に起因する損傷を含む。 In this specification, "nerve damage" refers to damage at any point on a nerve, and includes damage physically inflicted from outside the body, as well as damage caused by internal factors such as cancer or tumors.
本明細書において、「老化」とは、時間の経過と共に生物の個体に起こる種々の機能低下、形態変化、外観変化等及びその過程を指す。 In this specification, "aging" refers to various functional declines, morphological changes, changes in appearance, etc. that occur in individual organisms over time, and the processes involved.
老化により生じる状態としてフレイル及びサルコペニア等が知られている。フレイルとは、加齢と共に、心身の活力(運動機能、認知機能等)が低下し、複数の慢性疾患の併存等の影響も受けつつ、生活機能に障害が生じ、心身が脆弱化した状態を意味する。心身の活力の低下としては、例えば、認知機能障害、めまい、摂食障害、嚥下障害、視力障害、うつ、貧血、難聴、せん妄、易感染性、体重減少、筋量低下等が挙げられる。慢性疾患としては、高血圧、心疾患、脳血管疾患、糖尿病、呼吸器疾患、悪性腫瘍等が挙げられる。一方、サルコペニアとは、加齢又は疾患等により骨格筋量が低下し筋力が低下した状態を意味する。老齢マウスの神経筋接合部では、シナプス解離(synaptic detachment)、シナプス後部からの部分的又は完全な軸索の離脱等の形態の変化が見られることから、加齢に伴う神経筋接合部の形態の変化がサルコペニアにおける骨格筋量の低下等に関与していると考えられている。 Frailty and sarcopenia are known as conditions caused by aging. Frailty refers to a state in which physical and mental vitality (motor function, cognitive function, etc.) declines with age, and daily life functions are impaired and the body and mind become fragile, influenced by the coexistence of multiple chronic diseases. Examples of declines in physical and mental vitality include cognitive impairment, dizziness, eating disorders, swallowing disorders, visual impairment, depression, anemia, hearing loss, delirium, susceptibility to infection, weight loss, and muscle loss. Examples of chronic diseases include hypertension, heart disease, cerebrovascular disease, diabetes, respiratory disease, and malignant tumors. On the other hand, sarcopenia refers to a state in which skeletal muscle mass and muscle strength decrease due to aging or disease. Morphological changes such as synaptic detachment and partial or complete axonal detachment from the postsynapse have been observed in the neuromuscular junction of old mice, and it is believed that morphological changes in the neuromuscular junction associated with aging are involved in the decrease in skeletal muscle mass in sarcopenia.
本発明の組成物又は医薬組成物は、特に、フレイル又はサルコペニアを有するか、又はそれを有するリスクが高い対象において、老化による神経機能の低下を改善又は予防するために使用することができる。 The composition or pharmaceutical composition of the present invention can be used to improve or prevent the decline in neurological function due to aging, particularly in subjects who have or are at high risk of having frailty or sarcopenia.
本発明において、疾患としては、例えば、神経疾患及び神経筋疾患が挙げられる。
本明細書において、「神経疾患」とは、中枢神経又は末梢神経等の神経の障害により生じた疾患を指し、例えば、以下からなる群から選択される一以上の疾患を指す:アルツハイマー病、パーキンソン病、レビー小体型認知症、前頭側頭葉変性症、進行性核上性麻痺、大脳皮質基底核変性症、ハンチントン病、ジストニア、プリオン病、有棘赤血球舞踏病、副腎白質ジストロフィー、多系統萎縮症、脊髄小脳変性症、筋萎縮性側索硬化症、原発性側索硬化症、球脊髄性筋萎縮症、脊髄性筋萎縮症、痙性対麻痺、脊髄空洞症、シャルコー・マリー・トゥース病、前頭側頭型認知症、てんかん、統合失調症、自閉症、自閉症スペクトラム障害等。
In the present invention, diseases include, for example, neurological diseases and neuromuscular diseases.
As used herein, the term "neurological disease" refers to a disease caused by a disorder of nerves such as the central nervous system or peripheral nerves, and refers to, for example, one or more diseases selected from the group consisting of Alzheimer's disease, Parkinson's disease, dementia with Lewy bodies, frontotemporal lobar degeneration, progressive supranuclear palsy, corticobasal degeneration, Huntington's disease, dystonia, prion disease, acanthocytic chorea, adrenoleukodystrophy, multiple system atrophy, spinocerebellar degeneration, amyotrophic lateral sclerosis, primary lateral sclerosis, spinal-bulbar muscular atrophy, spinal muscular atrophy, spastic paraplegia, syringomyelia, Charcot-Marie-Tooth disease, frontotemporal dementia, epilepsy, schizophrenia, autism, autism spectrum disorder, and the like.
本明細書において、「神経筋疾患」とは、運動神経、神経筋接合部、又は筋肉細胞のいずれかの障害により生じた疾患を指し、例えば、以下からなる群から選択される一以上の疾患を指す:筋ジストロフィー、ミオパチー、先天性筋無力症候群、遺伝性周期性四肢麻痺、重症筋無力症、ランバート・イートン症候群等。 In this specification, the term "neuromuscular disease" refers to a disease caused by a disorder of either the motor nerves, the neuromuscular junction, or muscle cells, and refers to, for example, one or more diseases selected from the group consisting of muscular dystrophy, myopathy, congenital myasthenic syndrome, hereditary periodic paralysis, myasthenia gravis, Lambert-Eaton syndrome, etc.
筋萎縮性側索硬化症(ALS)は、一次運動神経及び二次運動神経が選択的にかつ進行性に変性・消失する疾患であり、初期病態として神経筋接合部において骨格筋から運動神経が脱離することが知られている。したがって、骨格筋と運動神経との間のシナプスの形成を促進又は退縮の抑制をすることができるシナプス形成促進剤又はシナプス維持剤を含む本発明の組成物又は医薬組成物は、特に、筋萎縮性側索硬化症による神経機能の低下を改善又は予防するために使用することができる。 Amyotrophic lateral sclerosis (ALS) is a disease in which primary and secondary motor nerves are selectively and progressively degenerated and lost, and it is known that the initial pathology is detachment of motor nerves from skeletal muscles at the neuromuscular junction. Therefore, the composition or pharmaceutical composition of the present invention, which contains a synapse formation promoter or synapse maintenance agent that can promote the formation of synapses between skeletal muscles and motor nerves or inhibit degeneration, can be used particularly to improve or prevent the decline in neurological function caused by amyotrophic lateral sclerosis.
例えば、本発明の医薬組成物は、本発明のコンジュゲート又は標的化剤を含む筋萎縮性側索硬化症治療用医薬組成物、脊髄性筋委縮症治療用医薬組成物等とすることができる。 For example, the pharmaceutical composition of the present invention can be a pharmaceutical composition for treating amyotrophic lateral sclerosis, a pharmaceutical composition for treating spinal muscular atrophy, etc., which contains the conjugate or targeting agent of the present invention.
筋肉増強剤を含む本発明の組成物又は医薬組成物は、筋肉を増強することにより、筋力の低下を改善又は予防することができる。したがって、筋肉増強剤を含む本発明の組成物又は医薬組成物は、筋力の低下、例えば、外傷若しくは外科手術後の筋力の低下、老化による筋力の低下、疾患による筋力の低下を改善若しくは予防するため、又は筋肉機能の向上のために使用することができる。 The composition or pharmaceutical composition of the present invention containing a muscle-enhancing agent can improve or prevent muscle weakness by strengthening muscles. Therefore, the composition or pharmaceutical composition of the present invention containing a muscle-enhancing agent can be used to improve or prevent muscle weakness, such as weakness after trauma or surgery, weakness due to aging, or weakness due to disease, or to improve muscle function.
本明細書において、「外傷」とは、外的要因による組織又は臓器の損傷を指し、例えば、創傷、骨折、捻挫、内臓破裂、熱傷、凍傷等を含む。 In this specification, "trauma" refers to damage to tissues or organs caused by external factors, and includes, for example, wounds, fractures, sprains, ruptured internal organs, burns, frostbite, etc.
機能改変剤を含む本発明の組成物又は医薬組成物は、運動神経細胞の機能を改変することにより、神経細胞の低下又は亢進を改善又は予防することができる。したがって、機能改変剤を含む本発明の組成物又は医薬組成物は、神経細胞の過活動、例えば、疾患若しくは状態による神経細胞の過活動を改善若しくは予防するため、又は筋肉の緊張、例えば、老化による震え、外傷若しくは疾患による筋肉の緊張を改善若しくは予防するためにも使用することができる。 The composition or pharmaceutical composition of the present invention containing a function-modifying agent can improve or prevent the decline or enhancement of nerve cells by modifying the function of motor nerve cells. Therefore, the composition or pharmaceutical composition of the present invention containing a function-modifying agent can also be used to improve or prevent nerve cell hyperactivity, for example, nerve cell hyperactivity due to a disease or condition, or to improve or prevent muscle tension, for example, tremors due to aging, or muscle tension due to trauma or disease.
具体的には、上述の各種疾患及び状態の他、例えば、異常不随意運動(ジスキネジア)(例えば、異常頭部運動、振戦、(有痛性)痙攣、筋線維束性攣縮等)、歩行及び移動の異常(例えば、失調性歩行、歩行困難等)、その他の協調運動障害(例えば、運動失調症等)、神経系及び筋骨格系に関するその他の状態(例えば、テタニー、異常反射、姿勢異常、痙縮、筋緊張亢進、筋緊張症、深部腱反射亢進、嚥下障害等)等を改善又は予防するために使用することができる。 Specifically, in addition to the various diseases and conditions mentioned above, it can be used to improve or prevent, for example, abnormal involuntary movements (dyskinesia) (e.g., abnormal head movements, tremors, (painful) convulsions, muscle fasciculations, etc.), abnormalities in walking and mobility (e.g., ataxic gait, difficulty walking, etc.), other coordination disorders (e.g., ataxia, etc.), and other conditions related to the nervous system and musculoskeletal system (e.g., tetany, abnormal reflexes, posture abnormalities, spasticity, hypertonia, myotonia, hyperactive deep tendon reflexes, dysphagia, etc.).
本明細書において「疾患」は、対象個体における識別可能な症状や原因によって分類可能な病的状態を指し、疾病及び障害を包含する。本発明において「状態」とは、対象個体における識別可能な症状を含む病的状態のうち、疾患に該当しないものを指す。 In this specification, "disease" refers to a pathological condition that can be classified by identifiable symptoms or causes in a subject individual, and includes illnesses and disorders. In the present invention, "condition" refers to a pathological condition that includes identifiable symptoms in a subject individual and does not fall under the category of a disease.
本発明の組成物又は医薬組成物は、運動神経細胞以外のシナプトジャイリン3が発現し得る任意の細胞において薬理効果を発揮し得るように構成されていてもよい。この場合の具体的な対象疾患及び状態は標的の細胞によって異なり、特に限定しない。例えば、上述の疾患及び状態のいずれかを対象として含んでもよく、他の疾患及び/又は状態を含んでもよい。他の疾患及び/又は状態としては、例えば、感染症(ウイルス性、細菌性等)、循環器疾患、血液疾患、消化器系疾患、免疫系疾患(炎症性疾患、自己免疫疾患等)、がん、生活習慣病、自律神経障害等が挙げられるが、特に限定しない。 The composition or pharmaceutical composition of the present invention may be configured to exert a pharmacological effect in any cell other than motor neurons in which synaptogyrin 3 can be expressed. In this case, the specific target disease and condition vary depending on the target cell and are not particularly limited. For example, the target may include any of the above-mentioned diseases and conditions, or may include other diseases and/or conditions. Examples of other diseases and/or conditions include, but are not limited to, infectious diseases (viral, bacterial, etc.), circulatory diseases, blood diseases, digestive system diseases, immune system diseases (inflammatory diseases, autoimmune diseases, etc.), cancer, lifestyle-related diseases, autonomic nervous system disorders, etc.
本発明の組成物又は医薬組成物は、本発明の標的化剤を複数含んでもよく、その他の有効成分をさらに含んでもよい。その他の有効成分は、組成物又は医薬組成物に含まれる標的化剤の機能を損なわない限り、特に限定しない。標的化剤を複数種類含む場合、シナプトジャイリン3以外の膜タンパク質に結合可能な標的化剤を含んでもよい。このような抗体としては、例えば、抗シナプトタグミン2抗体(抗シナプトタグミン2 小胞内ドメイン(N末端)抗体等)等が挙げられる。また、例えば、シナプトジャイリン3であっても、異なる部位(例えば、異なるエピトープ又は異なる小胞内ドメイン)に結合可能な抗体を含む標的化剤を含むことができる。これらの標的化剤に含まれる標識物質及び/又は生理活性物質は互いに異なっても同じであってもよい。 The composition or pharmaceutical composition of the present invention may contain a plurality of targeting agents of the present invention, and may further contain other active ingredients. The other active ingredients are not particularly limited as long as they do not impair the function of the targeting agent contained in the composition or pharmaceutical composition. When a plurality of targeting agents are contained, a targeting agent capable of binding to a membrane protein other than synaptogyrin 3 may be contained. Examples of such antibodies include anti-synaptotagmin 2 antibodies (anti-synaptotagmin 2 intravesicular domain (N-terminus) antibodies, etc.). In addition, for example, even if it is synaptogyrin 3, a targeting agent containing an antibody capable of binding to a different site (e.g., a different epitope or a different intravesicular domain) may be contained. The labeling substances and/or physiologically active substances contained in these targeting agents may be different or the same.
本発明の核酸分子及び/又は細胞を本発明の組成物又は医薬組成物に含めることができる。 The nucleic acid molecules and/or cells of the present invention can be included in the composition or pharmaceutical composition of the present invention.
<運動神経細胞又はシナプスへの標的化方法>
本発明は、運動神経細胞又はシナプスへの標的化方法に関する。本発明によれば、シナプトジャイリン3の小胞内ドメインに結合する本発明の抗体を運動神経細胞に接触させることにより、当該抗体を運動神経細胞内(特に運動神経細胞のシナプス小胞内)に取り込ませ、所望の物質(標識物質及び/若しくは生理活性物質)を運動神経細胞(例えば、運動神経細胞の軸索終末、軸索、軸索小丘、細胞体、樹状突起等)に標的化することができる。このようにして、シナプトジャイリン3の小胞内ドメインに結合する抗体が運動神経細胞又はシナプスに標的化される。当該抗体に所望の物質を直接的に又はリンカーを介して間接的に連結して本発明のコンジュゲートを製造し、これを運動神経細胞に接触させることにより、又は当該抗体と、当該抗体に結合可能な所望の物質を別々に運動神経細胞に接触させることにより、当該所望の物質を運動神経細胞内(例えば、運動神経細胞の細胞体内又はシナプス小胞内)に送達することができる。したがって、本発明によれば、運動神経細胞又はシナプスの標的化方法が提供され、本方法は、運動神経細胞等の細胞に抗体(所望の物質と連結していてもよい)を接触させることを含む。
Methods for targeting motor neurons or synapses
The present invention relates to a method for targeting a motor neuron or a synapse. According to the present invention, the antibody of the present invention that binds to the intravesicular domain of synaptogyrin 3 is brought into contact with a motor neuron, thereby allowing the antibody to be incorporated into the motor neuron (particularly into the synaptic vesicles of the motor neuron) and a desired substance (a labeling substance and/or a physiologically active substance) to be targeted to the motor neuron (e.g., the axon terminal, axon, axon hillock, cell body, dendrites, etc. of the motor neuron). In this way, the antibody that binds to the intravesicular domain of synaptogyrin 3 is targeted to the motor neuron or synapse. The desired substance can be delivered into the motor neuron (e.g., into the cell body or synaptic vesicles of the motor neuron) by directly or indirectly linking the antibody to the conjugate of the present invention via a linker and bringing the conjugate into contact with the motor neuron, or by separately bringing the antibody and a desired substance that can bind to the antibody into contact with the motor neuron. Thus, in accordance with the present invention there is provided a method of targeting a motor neuron or synapse, the method comprising contacting a cell, such as a motor neuron, with an antibody (optionally linked to a substance of interest).
<標識物質及び/又は生理活性物質の標的化方法>
本発明は、所望の物質(標識物質及び/又は生理活性物質)の標的化方法であって、本発明のコンジュゲート又は標的化剤を運動神経細胞に接触させる工程、及び標的化剤を運動神経細胞シナプスに送達する工程を含む方法に関する。
Method for targeting labeling substances and/or biologically active substances
The present invention relates to a method for targeting a desired substance (a labeled substance and/or a biologically active substance), comprising the steps of contacting a motor neuron with a conjugate or targeting agent of the present invention and delivering the targeting agent to the motor neuron synapse.
本発明における接触の対象は、運動神経細胞を含んでいれば特に限定されない。接触は、例えば、運動神経細胞のみを対象として行ってもよいし、運動神経細胞以外の細胞を含む組織を対象として行ってもよい。例えば、本発明の標的化剤は神経筋接合部における運動神経細胞シナプスの前部を主な標的とするため、運動神経細胞が投射する骨格筋細胞をさらに含む組織を対象としてもよい。 The subject of contact in the present invention is not particularly limited as long as it contains motor neurons. For example, contact may be performed only on motor neurons, or on tissue containing cells other than motor neurons. For example, since the targeting agent of the present invention primarily targets the anterior part of the motor neuron synapse at the neuromuscular junction, it may be targeted at tissue that further contains skeletal muscle cells to which motor neurons project.
本方法における運動神経細胞は、脊椎動物(非ヒト哺乳動物、ヒト、その他脊椎動物)の運動神経細胞を含み得る。本方法における運動神経細胞は、好ましくはヒト運動神経細胞を含む。以下、ヒト運動神経細胞を例として本発明を説明するが、本方法はヒト運動神経細胞に限定されるものではない。 The motor neurons in this method may include motor neurons of a vertebrate (non-human mammals, humans, other vertebrates). The motor neurons in this method preferably include human motor neurons. Below, the present invention will be described using human motor neurons as an example, but the method is not limited to human motor neurons.
ヒト運動神経細胞は、その由来に限らず制限なく用いることができる。例えば、これに限定されないが、ヒトから単離した細胞の初代培養、ヒトから単離され細胞株として樹立された細胞、ヒト由来の多能性幹細胞から分化誘導したヒト運動神経細胞を挙げることができる。ヒト運動神経細胞が由来する多能性幹細胞は、好ましくは、ヒト由来の多能性幹細胞である。 Human motor neurons can be used without any restrictions, regardless of their origin. Examples include, but are not limited to, primary cultures of cells isolated from humans, cells isolated from humans and established as cell lines, and human motor neurons induced to differentiate from human-derived pluripotent stem cells. The pluripotent stem cells from which human motor neurons are derived are preferably human-derived pluripotent stem cells.
一実施形態において、本方法は、in vitroの方法である。別の実施形態において、本方法は、ex vivoの方法である。別の実施形態において、本方法は、in vivoの方法である。本方法がin vivoの方法である場合、対象はヒトを除く哺乳動物であり得る。 In one embodiment, the method is an in vitro method. In another embodiment, the method is an ex vivo method. In another embodiment, the method is an in vivo method. When the method is an in vivo method, the subject may be a mammal other than a human.
本方法がin vitroの方法である場合、本方法は、シナプス形成を誘導する工程をさらに含んでもよい。本明細書において、「シナプス形成を誘導する」とは、神経細胞の軸索においてシナプス前部の形成を引き起こすこと、及び/又は別の神経細胞の樹状突起若しくは骨格筋、臓器等の細胞においてシナプス後部の形成を引き起こすことを指す。シナプス形成の誘導は、シナプス前部を形成しようとする細胞とシナプス後部を形成しようとする細胞とを共培養することにより行うこともできる。また、シナプス形成の誘導はその他の方法、例えば、運動神経細胞とLRRTM2の細胞外ドメインで被覆されたビーズとを共培養することにより行うことができる。シナプス形成を誘導する工程は、本発明の標的化剤を運動神経細胞に接触させる工程と同時又はその前に行われ得る。 When the method is an in vitro method, the method may further include a step of inducing synapse formation. As used herein, "inducing synapse formation" refers to causing the formation of a presynapse in the axon of a neuron and/or causing the formation of a postsynapse in the dendrite of another neuron or in cells of skeletal muscle, organs, etc. Synapse formation can also be induced by co-culturing a cell that is to form a presynapse with a cell that is to form a postsynapse. Synapse formation can also be induced by other methods, for example, by co-culturing a motor neuron with a bead coated with the extracellular domain of LRRTM2. The step of inducing synapse formation can be performed simultaneously with or before the step of contacting the motor neuron with the targeting agent of the present invention.
本方法がin vitroの方法である場合、本発明の標的化剤を運動神経細胞に接触させる工程は、本発明の標的化剤を、運動神経細胞を含む試料に接触させることにより行われる。接触の方法は、試料中の運動神経細胞と標的化剤が互いに接触可能であれば特に限定しない。例えば、標的化剤を試料に直接散布、噴霧、滴下、塗布することによって、試料を標的化剤に浸漬することによって、又はその組合せによって適用することができる。また、試料が他の担体(例えば、培養培地等)中に存在する場合、標的化剤をその担体に散布、噴霧、滴下、塗布することにより適用してもよい。 When the method is an in vitro method, the step of contacting the targeting agent of the present invention with motor neurons is carried out by contacting the targeting agent of the present invention with a sample containing motor neurons. The method of contact is not particularly limited as long as the motor neurons in the sample and the targeting agent can come into contact with each other. For example, the targeting agent can be applied by directly sprinkling, spraying, dripping, or applying the targeting agent to the sample, by immersing the sample in the targeting agent, or by a combination thereof. In addition, when the sample is present in another carrier (e.g., culture medium, etc.), the targeting agent may be applied by sprinkling, spraying, dripping, or applying to the carrier.
適用量は特に限定せず、運動神経細胞の数やその他の条件を勘案して適宜設定することができる。例えば、IgG抗体の濃度で、0.01μg/mL以上、0.1μg/mL以上、0.2μg/mL以上、0.5μg/mL以上、0.7μg/mL以上、0.9μg/mL以上、1μg/mL以上、2μg/mL以上、5μg/mL以上、7μg/mL以上、9μg/mL以上、又は10μg/mL以上を適用することができる。 The amount to be applied is not particularly limited, and can be set appropriately taking into consideration the number of motor neurons and other conditions. For example, the concentration of IgG antibody that can be applied is 0.01 μg/mL or more, 0.1 μg/mL or more, 0.2 μg/mL or more, 0.5 μg/mL or more, 0.7 μg/mL or more, 0.9 μg/mL or more, 1 μg/mL or more, 2 μg/mL or more, 5 μg/mL or more, 7 μg/mL or more, 9 μg/mL or more, or 10 μg/mL or more.
本方法がin vivoの方法である場合、本発明の標的化剤を被験試料に接触させる工程は、本発明の標的化剤を対象に投与することにより行われる。 When the method is an in vivo method, the step of contacting the targeting agent of the present invention with the test sample is carried out by administering the targeting agent of the present invention to a subject.
投与方法は特に限定しないが、例えば、局所投与、経腸投与、及び非経口投与、具体的には、皮膚上投与、吸入投与、注腸投与、点眼、点耳、経鼻投与、膣内投与、経管栄養による投与、経静脈投与、経動脈投与、筋肉内投与、心臓内投与、皮下投与、骨内投与、皮内投与、くも膜下(腔)投与、腹腔内投与、膀胱内投与、経皮投与、経粘膜投与、硬膜外投与、硝子体内投与等が挙げられる。 The administration method is not particularly limited, but examples include local administration, enteral administration, and parenteral administration, specifically administration onto the skin, inhalation administration, enema administration, eye drops, ear drops, nasal administration, intravaginal administration, administration by tube feeding, intravenous administration, intraarterial administration, intramuscular administration, intracardiac administration, subcutaneous administration, intraosseous administration, intradermal administration, subarachnoid (cavity) administration, intraperitoneal administration, intravesical administration, transdermal administration, transmucosal administration, epidural administration, intravitreal administration, etc.
投与量は特に限定せず、対象の動物種やその他の条件を勘案して適宜設定することができる。例えば、IgG抗体をマウス投与する場合、体重1kgあたりの用量が0.1mg/kg以上、0.5mg/kg以上、1mg/kg以上、2mg/kg以上、4mg/kg以上、又は5mg/kg以上を適用することができる。 The dosage is not particularly limited and can be set appropriately taking into consideration the target animal species and other conditions. For example, when administering an IgG antibody to a mouse, the dosage per kg of body weight can be 0.1 mg/kg or more, 0.5 mg/kg or more, 1 mg/kg or more, 2 mg/kg or more, 4 mg/kg or more, or 5 mg/kg or more.
本工程において使用される標的化剤は1種類である必要はない。例えば、複数種類の標的化剤を一緒に又は別々に使用することができる。具体的には、例えば、コンジュゲートを含む標的化剤と、所望の物質(標識物質及び/又は生理活性物質)を含まない標的化剤を使用することができる。また、使用する所望の物質も1種類である必要はなく、例えば、複数種類の標識物質及び/又は生理活性物質を一緒に又は別々に使用することができる。具体的には、例えば、標識物質及び生理活性物質を組み合わせて使用してもよい。 The targeting agent used in this step does not have to be of one type. For example, multiple types of targeting agents can be used together or separately. Specifically, for example, a targeting agent containing a conjugate and a targeting agent not containing a desired substance (labeling substance and/or biologically active substance) can be used. In addition, the desired substance used does not have to be of one type either. For example, multiple types of labeling substances and/or biologically active substances can be used together or separately. Specifically, for example, a labeling substance and a biologically active substance may be used in combination.
本発明の標的化剤を運動神経細胞に接触させる工程は複数回行うことができる。本工程を複数回行う場合、それぞれにおいて使用される標的化剤及び細胞の種類、並びに適用方法及び投与方法は、毎回同じでも異なってもよい。 The step of contacting the targeting agent of the present invention with motor neurons can be carried out multiple times. When this step is carried out multiple times, the types of targeting agent and cells used, as well as the application method and administration method, may be the same or different each time.
運動神経細胞に本発明のコンジュゲート又は標的化剤を接触させると、本発明の標的化剤は、シナプス小胞を介して運動神経に取り込まれる。本発明の標的化剤を運動神経細胞に接触させる際には、運動神経細胞を活動させる又はその活動を促進させることができる。運動神経細胞を活動させる又はその活動を促進させることにより、本発明のコンジュゲート又は標的化剤の運動神経細胞内への取り込み効率を向上させることができる。通常、本発明のコンジュゲート又は標的化剤がシナプス小胞に取り込まれると、そのまま逆行輸送により軸索を通じて細胞体へと送達される。 When the conjugate or targeting agent of the present invention is brought into contact with a motor neuron, the targeting agent of the present invention is taken up into the motor neuron via synaptic vesicles. When the targeting agent of the present invention is brought into contact with a motor neuron, the motor neuron can be activated or its activity can be promoted. By activating the motor neuron or promoting its activity, the efficiency of the uptake of the conjugate or targeting agent of the present invention into the motor neuron can be improved. Usually, when the conjugate or targeting agent of the present invention is taken up into a synaptic vesicle, it is delivered directly to the cell body via the axon by retrograde transport.
運動神経細胞を活動させる方法は特に限定しないが、例えば、十分な時間にわたり、運動神経細胞を自発的に活動させる方法、及び人工的な刺激によって運動神経細胞を活動させる又はシナプス小胞のエンドサイトーシスを促進する方法等が挙げられる。 The method for activating motor neurons is not particularly limited, but examples include a method of spontaneously activating motor neurons for a sufficient period of time, and a method of activating motor neurons by artificial stimulation or promoting endocytosis of synaptic vesicles.
自発的に活動させる方法としては、例えば、十分な時間にわたり、活動可能な環境に運動神経細胞を置くことが含まれる。運動神経細胞が活動可能な環境は、当技術分野において周知である。 Methods for spontaneous activation include, for example, placing motor neurons in an environment in which they can be activated for a sufficient period of time. Environments in which motor neurons can be activated are well known in the art.
自発的に活動させる方法において運動神経細胞を活動させる時間(例えば、本発明のコンジュゲート又は標的化剤との接触時間等)は特に限定しないが、例えば、本方法がin vitroの方法である場合、1時間以上、3時間以上、6時間以上、12時間以上、18時間以上、又は24時間以上とすることができる。また、本方法がin vivoの方法である場合、例えば、1時間以上、3時間以上、6時間以上、12時間以上、18時間以上、24時間以上、36時間以上、48時間以上、60時間以上、72時間以上、100時間以上、120時間以上、150時間以上、168時間以上、200時間以上又は240時間以上とすることができる。 In the method for spontaneous activation, the time for activating motor neurons (e.g., contact time with the conjugate or targeting agent of the present invention) is not particularly limited, but for example, when the method is an in vitro method, it can be 1 hour or more, 3 hours or more, 6 hours or more, 12 hours or more, 18 hours or more, or 24 hours or more. Also, when the method is an in vivo method, it can be, for example, 1 hour or more, 3 hours or more, 6 hours or more, 12 hours or more, 18 hours or more, 24 hours or more, 36 hours or more, 48 hours or more, 60 hours or more, 72 hours or more, 100 hours or more, 120 hours or more, 150 hours or more, 168 hours or more, 200 hours or more, or 240 hours or more.
人工的な刺激によって運動神経細胞を活動させる又はその活動を促進させる方法としては、例えば、十分な時間にわたり、運動神経細胞が活発に活動し得る環境に運動神経細胞を置くことが含まれる。 Methods of activating or promoting motor neuron activity through artificial stimulation include, for example, placing motor neurons in an environment in which they can be actively active for a sufficient period of time.
本方法がin vitroの方法である場合、運動神経細胞を活動させる又はその活動を促進させる方法は、運動神経細胞に、化学的刺激及び/又は物理的刺激を与えることができる。運動神経細胞を活発に活動させるための刺激は、当技術分野においては周知である。化学的刺激に使用される化合物としては、例えば、アミオダロン、テトラエチルアンモニウム、4-アミノピリジン、バリウム、デンドロトキシン等のカリウムイオンチャネル阻害剤、バトラコトキシン等のナトリウムチャネルアゴニスト、Bay K8644等のカルシウムチャネルアゴニスト、高濃度のカリウムイオン若しくは神経伝達物質、又はこれらの組合せ等が挙げられる。物理的刺激としては、例えば、温度変化等が挙げられる。 When the method is an in vitro method, the method of activating or promoting motor neuron activity can include providing chemical and/or physical stimulation to the motor neuron. Stimuli for activating motor neuron activity are well known in the art. Compounds used for chemical stimulation include, for example, potassium ion channel inhibitors such as amiodarone, tetraethylammonium, 4-aminopyridine, barium, dendrotoxin, sodium channel agonists such as batrachotoxin, calcium channel agonists such as Bay K8644, high concentrations of potassium ions or neurotransmitters, or combinations thereof. Physical stimuli include, for example, temperature changes.
化学的刺激を行う場合、化合物の添加量は特に限定しない。例えば、1μM以上、10μM以上、50μM以上、又は100μM以上の濃度で添加することができる。 When chemical stimulation is performed, the amount of compound added is not particularly limited. For example, the compound can be added at a concentration of 1 μM or more, 10 μM or more, 50 μM or more, or 100 μM or more.
刺激の時間は特に限定せず、刺激の種類及び強度等の条件を勘案して適宜設定することができる。例えば、2分以上、3分以上、4分以上、5分以上、8分以上、9分以上、10分以上、20分以上、25分以上、30分以上、又は1時間以上にわたり刺激を与えることができる。 The duration of the stimulation is not particularly limited, and can be set appropriately taking into consideration the type and intensity of the stimulation and other conditions. For example, the stimulation can be applied for 2 minutes or more, 3 minutes or more, 4 minutes or more, 5 minutes or more, 8 minutes or more, 9 minutes or more, 10 minutes or more, 20 minutes or more, 25 minutes or more, 30 minutes or more, or for 1 hour or more.
本方法がin vivoの方法である場合、運動神経細胞を活動させる又はその活動を促進させる方法は、対象に活発に活動させる方法(例えば、対象に運動をさせる方法や脳活動を活発化させる方法等)や、化学物質等により運動神経細胞の活動を促進する方法等により行うことができる。運動神経の活動を促進する化合物としては、上記化学的刺激に使用される化合物が挙げられ、対象に投与することができる。上記化学的刺激に使用される化合物は、薬学的に許容される濃度又は方法で投与され得る。化合物刺激が生体毒性を発揮することがないように上記化合物を対象に投与することができるが、生体毒性が発現する場合には対象への上記化合物の投与をしなくてもよい。運動神経細胞の活動を促進した場合、通常、自然発火させる方法と比較して短時間で同程度の効果が見込まれる。 When the method is an in vivo method, the method of activating or promoting the activity of motor neurons can be performed by a method of activating a subject (for example, a method of making a subject exercise or a method of activating brain activity, etc.) or a method of promoting the activity of motor neurons with a chemical substance, etc. Compounds that promote the activity of motor neurons include compounds used in the above-mentioned chemical stimulation, and can be administered to the subject. Compounds used in the above-mentioned chemical stimulation can be administered at a pharmaceutically acceptable concentration or method. The above-mentioned compounds can be administered to the subject so that the compound stimulation does not exert biotoxicity, but if biotoxicity is manifested, it is not necessary to administer the above-mentioned compounds to the subject. When the activity of motor neurons is promoted, the same degree of effect can usually be expected in a short time compared to the method of causing spontaneous combustion.
所望の物質を含まない標的化剤を使用する場合、所望の物質と標的化剤とを一緒に又は別々に運動神経細胞に接触させることができる。その所望の物質と標的化剤とを別々に接触させる場合、所望の物質が標的化剤と結合可能であれば、そのタイミングは特に限定しない。例えば、所望の物質の運動神経細胞との接触は、標的化剤と運動神経細胞との接触の前から、又はその後に行うことができる。 When a targeting agent that does not contain the desired substance is used, the desired substance and the targeting agent can be contacted with the motor neuron together or separately. When the desired substance and the targeting agent are contacted separately, the timing is not particularly limited as long as the desired substance can bind to the targeting agent. For example, the desired substance can be contacted with the motor neuron before or after the targeting agent is contacted with the motor neuron.
各接触の方法は、上述の接触方法に準じて選択することができる。例えば、各接触について、同じ方法又は互いに異なる方法を使用することができる。 The method of each contact can be selected in accordance with the contact methods described above. For example, the same method or different methods can be used for each contact.
本方法は、必要に応じて、標的化の成否を確認する工程をさらに含んでもよい。本方法で使用した標的化剤が生理活性物質を含む場合には、例えば、標的化剤の項において上述したように、生理的効果が見られた場合に標的化が成功したものと判断することができる。また、本方法で使用した標的化剤が標識物質を含む場合には、後述の可視化方法の標識物質のシグナルを検出する工程に準じて、シグナルが検出された場合に標的化が成功したものと判断することができる。 If necessary, the method may further include a step of confirming whether or not targeting has been successful. When the targeting agent used in the method includes a physiologically active substance, it can be determined that targeting has been successful when a physiological effect is observed, for example, as described above in the section on targeting agents. Furthermore, when the targeting agent used in the method includes a labeling substance, it can be determined that targeting has been successful when a signal is detected, similar to the step of detecting the signal of the labeling substance in the visualization method described below.
本方法において使用する標的化剤として本発明の医薬組成物を使用する場合、本方法を状態若しくは疾患の予防若しくは治療方法として使用することができる。 When the pharmaceutical composition of the present invention is used as a targeting agent in this method, the method can be used as a method for preventing or treating a condition or disease.
この場合、対象において生理的効果を十分に発揮させる工程をさらに含むことができる。例えば、使用される生理活性物質が単独で効果を発揮することができる物質である場合には、生理的効果が発揮されるのに十分な時間だけ対象を栄養が十分に与えられた環境下に置くことにより効果を発揮させることができる。また、例えば、使用される生理活性物質が効果を発揮するために他の物質が必要である場合、その物質を追加で投与することができる。 In this case, the method may further include a step of allowing the physiological effect to be fully exerted in the subject. For example, if the physiologically active substance used is a substance that can exert an effect by itself, the effect can be exerted by placing the subject in an environment where the subject is provided with sufficient nutrition for a period of time sufficient for the physiological effect to be exerted. Also, for example, if another substance is necessary for the physiologically active substance used to exert an effect, that substance can be administered in addition.
この場合、本発明は、本発明の抗体及び生理活性物質を含む標的化剤を運動神経細胞に接触させる工程、及び前記標的化剤を前記運動神経細胞シナプスに送達する工程を含む、状態又は疾患の予防又は治療方法に関する。本方法によれば、医薬組成物に関して例示した各種状態又は疾患を予防又は治療することができる。本方法の接触させる工程は、好ましくは対象に標的化剤及び/又は医薬組成物を投与することを含む。 In this case, the present invention relates to a method for preventing or treating a condition or disease, comprising the steps of contacting a targeting agent containing the antibody of the present invention and a physiologically active substance with a motor neuron, and delivering the targeting agent to the motor neuron synapse. According to this method, it is possible to prevent or treat various conditions or diseases exemplified in relation to the pharmaceutical composition. The contacting step of this method preferably includes administering the targeting agent and/or the pharmaceutical composition to a subject.
したがって、例えば、本発明の方法は、神経機能の低下、例えば、神経の損傷による神経機能の低下、老化による神経機能の低下、若しくは疾患による神経機能の低下等を改善若しくは予防するため、又は神経機能の向上のための方法である。一実施形態において、状態又は疾患は、神経機能の低下を呈する状態若しくは疾患である。一実施形態において、状態又は疾患は、神経疾患及び神経筋疾患である。一実施形態において、標的化剤は、前記抗体と前記生理活性物質とのコンジュゲートを含む。 Thus, for example, the method of the present invention is a method for improving or preventing a decline in nerve function, such as a decline in nerve function due to nerve damage, a decline in nerve function due to aging, or a decline in nerve function due to a disease, or for improving nerve function. In one embodiment, the condition or disease is a condition or disease exhibiting a decline in nerve function. In one embodiment, the condition or disease is a neurological disease or a neuromuscular disease. In one embodiment, the targeting agent comprises a conjugate of the antibody and the physiologically active substance.
この工程の時間は、対象の状態、生理活性物質の種類、投与量等によって適宜決定することができる。例えば、一般に生理活性物質を投与した場合に生理的効果が発揮されるまでの期間を基準に判断してもよく、生理的効果の確認を1回又は複数回行い、生理的効果が十分に発揮されるまで継続してもよい。 The duration of this step can be appropriately determined depending on the condition of the subject, the type of physiologically active substance, the dosage, etc. For example, it may be determined based on the period it takes for a physiological effect to be exerted when a physiologically active substance is generally administered, or the physiological effect may be confirmed once or multiple times and continued until the physiological effect is fully exerted.
<運動神経細胞又はそのシナプスの可視化方法>
本発明は、運動神経細胞又はシナプスの可視化方法であって、本発明の可視化剤を運動神経細胞に接触させる工程、前記可視化剤を運動神経細胞シナプスに送達する工程、及び前記標識物質のシグナルを検出する工程を含む方法に関する。
<Method for visualizing motor neurons or their synapses>
The present invention relates to a method for visualizing motor neurons or synapses, the method comprising the steps of contacting a motor neuron with a visualization agent of the present invention, delivering the visualization agent to a motor neuron synapse, and detecting a signal of the labeling substance.
本方法において、接触に用いる運動神経細胞は、上記の標識物質及び/又は生理活性物質の標的化方法について記載した通りのものである。 In this method, the motor neurons used for contact are as described above in the method for targeting a labeling substance and/or a biologically active substance.
一実施形態において、本方法は、in vitroの方法である。別の実施形態において、本方法は、ex vivoの方法である。別の実施形態において、本方法は、in vivoの方法である。本方法がin vivoの方法である場合、対象はヒトであるか、又はヒトを除く哺乳動物であり得る。 In one embodiment, the method is an in vitro method. In another embodiment, the method is an ex vivo method. In another embodiment, the method is an in vivo method. When the method is an in vivo method, the subject may be a human or a non-human mammal.
本方法において、本発明の可視化剤を運動神経細胞に接触させる工程及び可視化剤を運動神経細胞シナプスに送達する工程は、標的化剤として可視化剤を使用すること以外は、上記の標識物質及び/又は生理活性物質の標的化方法について記載した本発明の標的化剤を運動神経細胞に接触させる工程及び標的化剤を運動神経細胞シナプスに送達する工程の通りである。 In this method, the step of contacting a motor neuron with the visualization agent of the present invention and the step of delivering the visualization agent to the motor neuron synapse are the same as the step of contacting a motor neuron with the targeting agent of the present invention and the step of delivering the targeting agent to the motor neuron synapse described in the above-mentioned method for targeting a labeling substance and/or a physiologically active substance, except that the visualization agent is used as a targeting agent.
本方法は、必要に応じて、標識物質からシグナルを発生させる工程をさらに含んでもよい。シグナルを発生させる方法は特に限定しない。シグナルを発生させる方法及びその要否は、使用する標識物質の種類等によって判断することができる。 This method may further include a step of generating a signal from the labeling substance, if necessary. The method of generating a signal is not particularly limited. The method of generating a signal and whether or not it is necessary can be determined depending on the type of labeling substance used, etc.
例えば、標識物質が蛍光分子又は放射性標識物質の場合には、可視化剤が標的の運動神経細胞シナプスに送達するのに十分な時間、つまり、可視化剤が標的の運動神経細胞シナプスに到達し、その運動神経細胞シナプスにおいて十分にシナプス小胞のエンドサイトーシスが起こるのに十分な時間待つことにより、標的においてシグナルを発生させることができる。この時間は、可視化剤を運動神経細胞シナプスに送達する工程の時間に準じて適宜選択することができる。 For example, when the labeling substance is a fluorescent molecule or a radioactive labeling substance, a signal can be generated in the target by waiting a sufficient time for the visualization agent to be delivered to the target motor neuron synapse, that is, a sufficient time for the visualization agent to reach the target motor neuron synapse and for sufficient endocytosis of synaptic vesicles to occur at the motor neuron synapse. This time can be appropriately selected according to the time of the step of delivering the visualization agent to the motor neuron synapse.
例えば、標識物質が化学発光物質の場合には、可視化剤が標的の運動神経細胞シナプスに送達されるのに十分な時間待つことに加え、シグナルの発生に使用されるその基質等の物質を添加することにより行うことができる。
本工程は、後述のシグナルを検出する工程と同時又はその前に行うことができる。
For example, when the labeling substance is a chemiluminescent substance, this can be achieved by waiting a sufficient time for the visualization agent to be delivered to the target motor neuron synapse, as well as adding a substance such as its substrate that is used to generate a signal.
This step can be carried out simultaneously with or prior to the step of detecting a signal, which will be described later.
本方法は、検出用物質のシグナルを検出する工程をさらに含む。検出に使用する方法は特に限定せず、使用する標識物質の種類等の条件に応じて適宜選択することができる。 This method further includes a step of detecting the signal of the detection substance. The method used for detection is not particularly limited, and can be appropriately selected depending on conditions such as the type of labeling substance used.
標識物質が蛍光物質である場合、例えば、標識物質の励起波長の光を含む励起光を運動神経細胞に照射し、標識物質の蛍光波長を検出可能な検出器を用いて検出することができる。また、標識物質が化学発光物質である場合、例えば、標識物質の発光波長を検出可能な検出器を用いて検出することができる。標識物質が放射性標識物質である場合には、標識物質が発する放射線を検出可能な検出器を用いて検出することができる。 If the labeled substance is a fluorescent substance, for example, excitation light containing light of the excitation wavelength of the labeled substance can be irradiated onto the motor neuron, and the fluorescent wavelength of the labeled substance can be detected using a detector capable of detecting the fluorescent wavelength of the labeled substance. If the labeled substance is a chemiluminescent substance, for example, it can be detected using a detector capable of detecting the emission wavelength of the labeled substance. If the labeled substance is a radioactive labeled substance, it can be detected using a detector capable of detecting the radiation emitted by the labeled substance.
標識物質のシグナルの検出は、運動神経細胞を含む試料における運動神経細胞(例えば、シナプス、細胞体等)の存在、位置若しくは量を検出することを含む。 Detecting the signal of the labeled substance includes detecting the presence, location, or quantity of motor neurons (e.g., synapses, cell bodies, etc.) in a sample that contains motor neurons.
本発明の方法は、試料中の標識物質のシグナルを検出する工程に加えて、試料で検出された標識物質のシグナルを、標識物質を含む標準試料におけるシグナル、又は予め作成した基準値と比較して存在、位置又は量を判定する工程をさらに含み得る。標準試料は、特定の状態又は疾患か否かを判断するための基準となる生物試料であれば特に限定しない。具体的には、例えば、健常個体から得られたもの、試料と同じ個体から異なる回収時期に得られたもの、又は特定の状態若しくは疾患であることがわかっている個体から得られたもの等が挙げられる。標準試料は、例えば、試料と同じ生物種、個体、組織又は細胞に由来する生物試料であってもよく、他の生物種、個体、組織又は細胞に由来する生物試料であってもよい。また、基準値は、目的の状態か否かを判断するための基準となる値であれば特に限定しない。基準値は、例えば、標準試料で一般に検出されるシグナルの強度又は数等に基づいて設定することができる。 In addition to the step of detecting the signal of the labeling substance in the sample, the method of the present invention may further include a step of determining the presence, location, or amount of the signal of the labeling substance detected in the sample by comparing it with a signal in a standard sample containing the labeling substance, or with a previously prepared reference value. There are no particular limitations on the standard sample, so long as it is a biological sample that serves as a reference for determining whether or not there is a specific condition or disease. Specific examples include those obtained from healthy individuals, those obtained from the same individual as the sample at a different collection time, and those obtained from individuals known to have a specific condition or disease. The standard sample may be, for example, a biological sample derived from the same biological species, individual, tissue, or cell as the sample, or may be a biological sample derived from another biological species, individual, tissue, or cell. In addition, there are no particular limitations on the reference value, so long as it is a value that serves as a reference for determining whether or not there is a target condition. The reference value can be set, for example, based on the intensity or number of signals generally detected in the standard sample.
いずれの場合においても、比較の方法は特に限定しない。例えば、目視により、数値の大きさにより又は統計的手法により行うことができる。 In either case, the method of comparison is not particularly limited. For example, it can be done by visual inspection, by the magnitude of the numerical values, or by statistical methods.
<その他の発明>
本発明は、対象に所望の物質を投与する方法を提供する。物質は、本発明の抗体と当該所望の物質とのコンジュゲートの形態である。これにより、対象の細胞、例えば運動神経細胞に前記所望の物質を送達することができる。前記所望の物質が生理活性物質である場合には、当該生理活性物質を運動神経細胞等の細胞に送達することができる。また、前記所望の物質が標識物質である場合には、当該標識物質の送達部位(例えば運動神経細胞又はそのシナプス)を観察することに用いることができる。本発明はまた、この方法に用いるための当該抗体と当該所望の物質とのコンジュゲート、又は当該コンジュゲートを含む組成物を提供する。この場合、本発明は、本発明の抗体、又は上記抗体と所望の物質(標識物質及び/若しくは生理活性物質)とのコンジュゲートを含む、運動神経細胞又はそのシナプスへの標的化用組成物に関する。
<Other inventions>
The present invention provides a method for administering a desired substance to a subject. The substance is in the form of a conjugate between the antibody of the present invention and the desired substance. This allows the desired substance to be delivered to a cell of the subject, for example, a motor neuron. When the desired substance is a physiologically active substance, the physiologically active substance can be delivered to a cell such as a motor neuron. When the desired substance is a labeling substance, the method can be used to observe the delivery site of the labeling substance (for example, a motor neuron or its synapse). The present invention also provides a conjugate of the antibody and the desired substance for use in this method, or a composition comprising the conjugate. In this case, the present invention relates to a composition for targeting a motor neuron or its synapse, comprising the antibody of the present invention or a conjugate of the antibody and a desired substance (a labeling substance and/or a physiologically active substance).
本発明は、対象の運動神経細胞を可視化する方法であって、当該対象に本発明の抗体と標識物質とのコンジュゲートの有効量を投与することを含む方法を提供する。本発明はまた、この方法に用いるための当該抗体と当該標識物質とのコンジュゲート、又は当該コンジュゲートを含む組成物を提供する。 The present invention provides a method for visualizing motor neurons in a subject, the method comprising administering to the subject an effective amount of a conjugate of the antibody of the present invention and a labeling substance. The present invention also provides a conjugate of the antibody and the labeling substance for use in the method, or a composition containing the conjugate.
本発明は、対象の運動神経細胞に生理活性物質を送達する方法であって、当該対象に本発明の抗体と当該生理活性物質とのコンジュゲートの有効量を投与することを含む方法を提供する。本発明はまた、この方法に用いるための当該抗体と当該生理活性物質とのコンジュゲート、又は当該コンジュゲートを含む組成物を提供する。 The present invention provides a method for delivering a physiologically active substance to a motor neuron of a subject, the method comprising administering to the subject an effective amount of a conjugate of the antibody of the present invention and the physiologically active substance. The present invention also provides a conjugate of the antibody and the physiologically active substance for use in the method, or a composition containing the conjugate.
本発明は、上記方法のいずれかにおいて使用するための、上記本発明の抗体又は上記本発明のコンジュゲートを提供する。例えば、本発明は、状態又は疾患の予防又は治療方法において使用するための、上記本発明の抗体又は上記本発明のコンジュゲートに関する。また、例えば、本発明は、運動神経細胞又はそのシナプスへの標識物質及び/又は生理活性物質の標的化方法において使用するための、上記本発明の抗体又は上記本発明のコンジュゲートに関する。 The present invention provides the antibody of the present invention or the conjugate of the present invention for use in any of the above methods. For example, the present invention relates to the antibody of the present invention or the conjugate of the present invention for use in a method for preventing or treating a condition or disease. Also, for example, the present invention relates to the antibody of the present invention or the conjugate of the present invention for use in a method for targeting a labeling substance and/or a physiologically active substance to a motor neuron or its synapse.
本発明は、上記方法のいずれかに用いるための医薬の製造において使用するための、上記抗体又は上記抗体と上記物質とのコンジュゲートを提供する。 The present invention provides the above antibody or a conjugate of the above antibody and the above substance for use in the manufacture of a medicament for use in any of the above methods.
また、本発明は、上記本発明の抗体又は上記本発明のコンジュゲートの、上記抗体及び生理活性物質を含む医薬の製造における使用を提供する。 The present invention also provides use of the antibody of the present invention or the conjugate of the present invention in the manufacture of a medicine comprising the antibody and a physiologically active substance.
以下、実施例を用いて本発明をさらに具体的に説明する。ただし、本発明の技術的範囲はこれら実施例に限定されるものではない。 The present invention will be explained in more detail below using examples. However, the technical scope of the present invention is not limited to these examples.
<実施例1:モノクローナル抗体の作製>
モノクローナル抗体は、株式会社医学生物学研究所に委託して、作製及びB細胞クローニング法によるスクリーニングを行った。作製及びスクリーニングの手順の概要は以下の通りである。
Example 1: Preparation of monoclonal antibodies
The production of monoclonal antibodies and screening using the B cell cloning method were outsourced to Medical and Biological Laboratories Co., Ltd. The outline of the production and screening procedures is as follows.
1.抗原ペプチドの合成
ユーロフィンジェノミクス株式会社に委託して、免疫用抗原及びスクリーニング用抗原として下記のペプチド(第2の小胞内ドメインの部分配列を含む)を合成した。
1. Synthesis of Antigen Peptides The synthesis of the following peptides (containing partial sequences of the second vesicle domain) was entrusted to Eurofins Genomics K.K. as immunization antigens and screening antigens.
免疫用抗原をエルマン試薬を用いて、免疫応答を誘導するためのキャリアタンパク質に結合させた。キャリアタンパク質としてはキーホールリンペットヘモシアニン(KLH)を用いた。 The immunization antigen was bound to a carrier protein to induce an immune response using Ellman's reagent. Keyhole limpet hemocyanin (KLH) was used as the carrier protein.
2.マウスの免疫
KLH結合免疫用抗原を1.0mg/mLの抗原溶液として調製し、マウス4匹に毎週1回、計4回免疫した。免疫の完了したマウスから抗体力価確認用の血清、及びB細胞単離用にリンパ節細胞を回収した。
2. Immunization of mice The KLH-conjugated immunization antigen was prepared as a 1.0 mg/mL antigen solution, and four mice were immunized once a week for a total of four times. Serum for antibody titer confirmation and lymph node cells for B cell isolation were collected from the immunized mice.
「1.抗原ペプチドの合成」で合成されたスクリーニング用抗原ペプチドを抗原として用いたELISA法により、得られた血清の抗体力価を評価した。その結果、ヒトシナプトジャイリン3の小胞内ドメインに結合可能な抗体が、回収されたマウス血清中に存在することが確認された。 The antibody titer of the obtained serum was evaluated by ELISA using the screening antigen peptide synthesized in "1. Synthesis of antigen peptide" as an antigen. As a result, it was confirmed that antibodies capable of binding to the intravesicular domain of human synaptogyrin 3 were present in the collected mouse serum.
3.B細胞の単離
-80℃において保存したB細胞単離用に回収したマウスリンパ節細胞を起眠し、37℃、5%CO2の条件で1時間インキュベーションした。
3. Isolation of B cells Mouse lymph node cells collected for B cell isolation and stored at -80°C were awake and incubated at 37°C, 5% CO2 for 1 hour.
細胞を回収し、抗B細胞マーカー抗体及びストレプトアビジン(スクリーニング用ペプチドのビオチンに結合可能)で染色した。 The cells were harvested and stained with anti-B cell marker antibodies and streptavidin (which can bind to the biotin in the screening peptide).
染色細胞をFACS Melody(日本BD社)で解析し、抗B細胞マーカー及びビオチンについて陽性の細胞を抗原に反応するB細胞として1細胞ずつ単離した。 The stained cells were analyzed using FACS Melody (BD Japan), and cells that were positive for anti-B cell markers and biotin were isolated one by one as B cells that reacted to the antigen.
4.cDNA合成及び可変領域配列の単離
単離された各B細胞からmRNAを抽出し、5' RACE法にてcDNAを合成した。
4. cDNA synthesis and isolation of variable region sequences mRNA was extracted from each isolated B cell, and cDNA was synthesized by the 5' RACE method.
さらに、各細胞から合成されたcDNAから、PCR法にて可変領域の配列を含む核酸断片を増幅し、アガロース電気泳動にて目的サイズの遺伝子(VH、VL)が増幅されていることを確認した。 Furthermore, nucleic acid fragments containing the variable region sequences were amplified using PCR from the cDNA synthesized from each cell, and agarose electrophoresis was used to confirm that genes of the desired size (VH, VL) had been amplified.
5.抗体発現ベクターの構築
設計したマウスIgG1定常領域(CH)及びマウスIgκ定常領域(CL)の遺伝子配列、及び「4.cDNA合成及び可変領域配列の単離」にて増幅された可変領域遺伝子(VH、VL)に基づいて人工抗体遺伝子を作製した。この人工抗体遺伝子の作製はIntegrated DNA Technologies株式会社に委託して行った。増幅されたVHをヒトIgG1定常領域(hCH)と共にpcDNA3.4プラスミドに挿入し(重鎖発現ベクター)、同様に、VLをヒトIgκ定常領域(hCL)と共にpcDNA3.4プラスミドに挿入することで(軽鎖発現ベクター)、抗体発現ベクターを構築した。
5. Construction of antibody expression vector An artificial antibody gene was constructed based on the designed gene sequences of mouse IgG1 constant region (CH) and mouse Igκ constant region (CL) and the variable region genes (VH, VL) amplified in "4. cDNA synthesis and isolation of variable region sequences". The construction of this artificial antibody gene was outsourced to Integrated DNA Technologies Co., Ltd. The amplified VH was inserted into the pcDNA3.4 plasmid together with the human IgG1 constant region (hCH) (heavy chain expression vector), and similarly, VL was inserted into the pcDNA3.4 plasmid together with the human Igκ constant region (hCL) (light chain expression vector) to construct an antibody expression vector.
6.抗体含有上清の調製
重鎖発現ベクター及び軽鎖発現ベクターをHEK293T細胞へ導入し、導入後3日目の培養上清を抗体含有上清として回収した。ベクターの導入にはLipofectamineTM2000 Transfection Reagent(カタログ番号:#11668027、Thermo Fisher Scientific社)を用い、導入用培地としてOpti-MEMTMReduced Serum Medium(カタログ番号:#31985070、Thermo Fisher Scientific社)を用いた。なお、陰性対照上清として遺伝子導入を行わなかったHEK293T細胞の培養上清を回収した。
6. Preparation of antibody-containing supernatant Heavy chain expression vector and light chain expression vector were introduced into HEK293T cells, and the culture supernatant 3 days after introduction was collected as antibody-containing supernatant. Lipofectamine TM 2000 Transfection Reagent (catalog number: #11668027, Thermo Fisher Scientific) was used for vector introduction, and Opti-MEM TM Reduced Serum Medium (catalog number: #31985070, Thermo Fisher Scientific) was used as the introduction medium. Note that the culture supernatant of HEK293T cells that had not been transfected was collected as a negative control supernatant.
7.抗体のスクリーニング
ELISA法及びフローサイトメトリーにより、得られた抗体のスクリーニングを行った。各スクリーニングの手順の概要は以下の通りである。
7. Antibody Screening The obtained antibodies were screened by ELISA and flow cytometry. The outline of each screening procedure is as follows.
7-1.ELISA法によるスクリーニング
ストレプトアビジンが固相化されたImmobilizer Streptavidinプレート(カタログ番号:#436020、Thermo Fisher Scientific社)に「1.抗原ペプチドの合成」にて合成したスクリーニング用ペプチドを添加して抗原結合ELISAプレートを調製した。さらに、この抗原結合ELISAプレートに一次抗体として抗体含有上清を添加してインキュベートした。ここで、陰性対照としては一次抗体として抗体含有上清の代わりに陰性対照上清を添加したものを使用した。インキュベート後に、二次抗体として抗マウス抗体であるAnti-IgG(H+L)pAb-PE(カタログ番号:#330、MBL社)を添加してインキュベートした。
7-1. Screening by ELISA The screening peptide synthesized in "1. Synthesis of antigen peptide" was added to an Immobilizer Streptavidin plate (catalog number: #436020, Thermo Fisher Scientific) on which streptavidin was immobilized to prepare an antigen-binding ELISA plate. Furthermore, antibody-containing supernatant was added as a primary antibody to this antigen-binding ELISA plate and incubated. Here, as a negative control, a negative control supernatant was added instead of the antibody-containing supernatant as the primary antibody. After incubation, an anti-mouse antibody, Anti-IgG (H+L) pAb-PE (catalog number: #330, MBL), was added as a secondary antibody and incubated.
二次抗体反応後のプレートに発色用基質としてTMB-US(カタログ番号:#TMB-US、Moss社)を添加して標識を発色させ、H2PO4(カタログ番号:167-02166、富士フイルム和光純薬)を用いて発色反応を停止させた。 After the secondary antibody reaction, TMB-US (catalog number: #TMB-US, Moss) was added as a color-developing substrate to the plate to develop the label, and the color reaction was stopped using H 2 PO 4 (catalog number: 167-02166, Fujifilm Wako Pure Chemical Industries, Ltd.).
7-2.フローサイトメトリーによるスクリーニング
まず、ヒトSYNGR3の小胞内ドメインを細胞表面に発現するスクリーニング用細胞を調製した。HEK293T細胞を6 well Plate(カタログ番号:#3820、IWAKI社)に5.0×105 cells/wellの播種密度で播種し、抗原発現ベクターを導入した。抗原発現ベクターとしては、pCMV6-AC-Myc-DDK-IRES-GFP-Puro Mammalian Expression Vectorプラスミド(ORIGENE社製)にヒトSYNGR3(配列番号9で表されるアミノ酸配列)をコードする遺伝子配列(配列番号16)を有するDNA断片(配列番号15)をマルチクローニングサイトに組み込んだプラスミドベクターを用いた。遺伝子導入試薬及び培地としては「6.抗体含有上清の調製」にて使用したものと同じものを用いた。
7-2. Screening by flow cytometry First, cells expressing the intravesicular domain of human SYNGR3 on the cell surface were prepared for screening. HEK293T cells were seeded in a 6-well plate (catalog number: #3820, IWAKI) at a seeding density of 5.0×10 5 cells/well, and an antigen expression vector was introduced. The antigen expression vector used was a plasmid vector in which a DNA fragment (SEQ ID NO: 15) having a gene sequence (SEQ ID NO: 16) encoding human SYNGR3 (amino acid sequence represented by SEQ ID NO: 9) was integrated into the multicloning site of pCMV6-AC-Myc-DDK-IRES-GFP-Puro Mammalian Expression Vector plasmid (ORIGENE). The gene introduction reagent and medium used were the same as those used in "6. Preparation of antibody-containing supernatant".
遺伝子導入後3日目の細胞をスクリーニング用細胞として回収し、フローサイトメトリーに供した。フローサイトメトリーにはCytoFLEX(Beckman coulter社)を用いた。 Cells were collected three days after gene transfer as screening cells and subjected to flow cytometry. CytoFLEX (Beckman Coulter) was used for flow cytometry.
細胞の標識は以下の通り行った。各スクリーニング用細胞に、調製した抗体含有上清を添加した。二次抗体としては、Anti-Mouse IgG(H+L)pAb-PE(カタログ番号:#IM0855、Beckman coulter社)を200倍希釈で使用した。陰性対照としては、一次抗体として陰性対照上清を添加し、二次抗体として抗体含有上清に対するものと同じものを添加した。 Cell labeling was performed as follows. The prepared antibody-containing supernatant was added to each screening cell. Anti-Mouse IgG (H+L) pAb-PE (catalog number: #IM0855, Beckman Coulter) was used at a 200-fold dilution as the secondary antibody. As a negative control, negative control supernatant was added as the primary antibody, and the same secondary antibody as that used for the antibody-containing supernatant was added.
8.抗体遺伝子配列の決定
ELISA法及びフローサイトメトリーによるスクリーニングの両方で反応が見られた1クローンの抗体を本発明のモノクローナル抗体として取得し、その抗体の発現のために使用した重鎖発現ベクター及び軽鎖発現ベクターを一組の本発明の抗体発現用ベクターとして取得した。本発明の抗体の配列を同定するために、本発明の抗体発現用ベクターの塩基配列を決定した。配列決定はアゼンタ株式会社に委託して、サンガーシーケンス法により行った。
8. Determination of antibody gene sequence One clone of antibody that showed a reaction in both ELISA and flow cytometry screening was obtained as the monoclonal antibody of the present invention, and the heavy chain expression vector and light chain expression vector used for expressing the antibody were obtained as a set of antibody expression vectors of the present invention. In order to identify the sequence of the antibody of the present invention, the nucleotide sequence of the antibody expression vector of the present invention was determined. The sequence determination was entrusted to Azenta Co., Ltd. and was performed by the Sanger sequencing method.
本発明のモノクローナル抗体の可変領域のアミノ酸配列を表2に示す。 The amino acid sequences of the variable regions of the monoclonal antibodies of the present invention are shown in Table 2.
9.相補性決定領域(CDR)の配列の同定
本発明のモノクローナル抗体のCDRのアミノ酸配列はKabatの方法に基づいて同定した。 同定された各CDRのアミノ酸配列を表3に示す。
9. Identification of the sequences of the complementarity determining regions (CDRs) The amino acid sequences of the CDRs of the monoclonal antibodies of the present invention were identified based on the Kabat method. The amino acid sequences of the identified CDRs are shown in Table 3.
10.精製抗体液の調製
一組の本発明の抗体発現用ベクターをExpi CHOTM細胞にトランスフェクションし、培養上清を回収した。
10. Preparation of purified antibody solution A set of the antibody expression vectors of the present invention was transfected into Expi CHO TM cells, and the culture supernatant was collected.
得られた培養上清からPierceTM Disposable Columns, 10mL(カタログ番号:#29924;Thermo Scientific社)を用いて抗体を取得した。回収される抗体濃度が0.1mg/mL以下になるまで0.1M Citrate buffer(pH=4.0)を用いた溶出を繰り返し、抗体濃度が0.1mg/mL以上のフラクションを抗体溶液として取得した。抗体溶液を、1×ダルベッコリン酸緩衝生理食塩水(カタログ番号:D1408、Sigma Aldrich社)で透析し、孔径0.22μmのフィルターで濾過して精製抗体液を調製した。 Antibodies were isolated from the culture supernatant using Pierce TM Disposable Columns, 10 mL (catalog number: #29924; Thermo Scientific). Elution with 0.1 M citrate buffer (pH = 4.0) was repeated until the antibody concentration was 0.1 mg/mL or less, and fractions with antibody concentrations of 0.1 mg/mL or more were isolated as antibody solutions. The antibody solution was dialyzed against 1x Dulbecco's phosphate buffered saline (catalog number: D1408, Sigma Aldrich) and filtered through a filter with a pore size of 0.22 μm to prepare purified antibody solutions.
<実施例2:モノクローナル抗体の評価>
得られたモノクローナル抗体の抗原への結合特性を評価した。
Example 2: Evaluation of monoclonal antibodies
The antigen-binding properties of the obtained monoclonal antibodies were evaluated.
sc-hSYNGR3を抗原として用い、Octet RED96e System(SARTORIUS社)を使用して抗体の結合特性を測定した。測定には以下の条件を用いた:
バイオセンサー:Streptavidinバイオセンサー(カタログ番号:#18-5019;SARTORIUS社);
抗体濃度:10nM/5nM/2.5nM/1.25nM/0.63nM/0.31nM/0.17nM/0nM;
バッファー:0.02%Tween-20及び0.01% BSAを含むPBS;
固相化前平衡化:バッファーのみで60秒インキュベート;
固相化:抗原ペプチドを含むバッファーで300秒インキュベート;
抗体添加前平衡化:バッファーのみで300秒インキュベート;
抗体結合反応:抗体を含むバッファーで300秒インキュベート;
抗体解離反応:バッファーのみで900秒インキュベート;
いずれも30℃、1000rpmの撹拌速度でインキュベートした。
The binding characteristics of the antibody were measured using sc-hSYNGR3 as an antigen using the Octet RED96e System (SARTORIUS). The following conditions were used for the measurement:
Biosensor: Streptavidin biosensor (catalog number: #18-5019; SARTORIUS);
Antibody concentration: 10nM/5nM/2.5nM/1.25nM/0.63nM/0.31nM/0.17nM/0nM;
Buffer: PBS containing 0.02% Tween-20 and 0.01% BSA;
Pre-immobilization equilibration: Incubate with buffer only for 60 seconds;
Immobilization: Incubate with a buffer containing antigen peptide for 300 seconds;
Equilibration before antibody addition: Incubate with buffer only for 300 seconds;
Antibody binding reaction: Incubate with buffer containing antibody for 300 seconds;
Antibody dissociation reaction: Incubate with buffer only for 900 seconds;
Both were incubated at 30° C. with an agitation speed of 1000 rpm.
対照としてはポリクローナルヤギ抗SYNGR3小胞内ドメイン抗体を用いた。また、抗原は0.02μg/mLの濃度で使用した。 A polyclonal goat anti-SYNGR3 intravesicular domain antibody was used as a control. The antigen was used at a concentration of 0.02 μg/mL.
測定結果を表4及び5に示す。表4はポリクローナルヤギ抗SYNGR3小胞内ドメイン抗体の結果を示し、表5は本発明のモノクローナル抗体の結果を示す。 The measurement results are shown in Tables 4 and 5. Table 4 shows the results for the polyclonal goat anti-SYNGR3 intravesicular domain antibody, and Table 5 shows the results for the monoclonal antibody of the present invention.
結合速度定数は結合反応の進みやすさを示す値であり、親和性が高い程値は大きくなる。 The binding rate constant is a value that indicates how easily the binding reaction proceeds, and the higher the affinity, the larger the value.
表4及び5に示す結果から、対照ポリクローナル抗体にて測定された結果と比較して、本発明のモノクローナル抗体は結合速度定数が高く、抗原との親和性が高いことがわかった。 The results shown in Tables 4 and 5 indicate that the monoclonal antibodies of the present invention have a higher binding rate constant and higher affinity with the antigen than the results measured with the control polyclonal antibody.
<実施例3:小胞内ドメイン抗体を用いた薬剤の送達による薬理効果の検証>
小胞膜タンパク質の一例としてシナプトタグミン2(SYT2)を用いて、その小胞内ドメイン(N末端)に結合する抗体により運動神経細胞内に薬剤を送達した場合、薬剤に基づく薬理効果が観察されるかどうか調べた。
Example 3: Verification of pharmacological effects of drug delivery using intravesicular domain antibody
Using synaptotagmin 2 (SYT2) as an example of a vesicular membrane protein, we investigated whether drug-based pharmacological effects would be observed when drugs were delivered into motor neurons using an antibody that binds to its intravesicular domain (N-terminus).
1.細胞の培養
Dead Cell Removalキット(Veritas社)を用い、同キットのプロトコルに従ってヒトiPS誘導運動神経細胞(40HU-005-2M;iXCells Biotechnologies社)を解凍した。解凍後の細胞を96ウェルプレート(V底)に2×104細胞/ウェルの密度で播種し、運動神経細胞培養培地(Motor Neuron Maintenance Medium;iXCells Biotechnologies社)中で1週間培養し、ニューロスフィアを作製した。作製されたニューロスフィアは、大きさと真円度を基準に選別し、この基準を満たしたものを以下の実験に用いた。
1. Cell culture Human iPS-derived motor neurons (40HU-005-2M; iXCells Biotechnologies) were thawed using a Dead Cell Removal Kit (Veritas) according to the kit's protocol. The thawed cells were seeded in a 96-well plate (V-bottom) at a density of 2 × 104 cells/well and cultured in motor neuron maintenance medium (iXCells Biotechnologies) for 1 week to produce neurospheres. The produced neurospheres were selected based on size and circularity, and those that met these criteria were used in the following experiments.
96ウェルEZVIEW(登録商標)カルチャープレートLB(AGCテクノグラス株式会社)をポリ-D-リシン及びGeltrex(登録商標)Matrix(Thermo Fisher Scientific社)でコーティングした後、選別されたニューロスフィアをプレートに播種し、20日間培養した。培地としては、まず、上述の運動神経細胞培養培地を使用し、培養2日目に同培地での培地交換を行った。その後は、ニューロン培地(Neurobasal plus medium(B27 plus supplement(Thermo Fisher Scientific社)、20ng/mL BDNF、20ng/mL GDNF及びペニシリン・ストレプトマイシンを添加))を用いて、週3回培地交換を行った。いずれの培地交換も50μL/ウェルで行った。 A 96-well EZVIEW® culture plate LB (AGC Technoglass Co., Ltd.) was coated with poly-D-lysine and Geltrex® Matrix (Thermo Fisher Scientific Co., Ltd.), and the selected neurospheres were seeded on the plate and cultured for 20 days. The above-mentioned motor neuron culture medium was used as the culture medium, and the medium was replaced with the same medium on the second day of culture. Thereafter, the medium was replaced three times a week with neuron medium (Neurobasal plus medium (B27 plus supplement (Thermo Fisher Scientific Co., Ltd.), 20 ng/mL BDNF, 20 ng/mL GDNF, and penicillin-streptomycin added)). All medium changes were performed at 50 μL/well.
2.LRRTM2ビーズの調製
LRRTM2の細胞外ドメインで被覆されたマイクロビーズ(活性ビーズ)を、WO2021/006075に開示されるようにして調製した。
2. Preparation of LRRTM2 beads Microbeads coated with the extracellular domain of LRRTM2 (active beads) were prepared as disclosed in WO2021/006075.
具体的には、ストレプトアビジン被覆マイクロビーズ(Streptavidin Coated Microspheres; Bangs Laboratories, Inc;ポリスチレン製、平均直径9.94μm)を洗浄緩衝液(リン酸緩衝食塩水(PBS)、0.01% ウシ血清アルブミン(BSA)、0.05% TritonX-100)で2回洗浄し、ビオチン化抗ヒトIgG(Fc特異的)抗体(Sigma Aldrich社;マウスモノクローナル)と結合緩衝液(PBS、0.01% BSA)中で反応させ、ストレプトアビジン被覆マイクロビーズにビオチン化抗ヒトIgG(Fc特異的)抗体を固相化した。得られたビーズを洗浄緩衝液で3回洗浄した(抗ヒトIgGFc抗体ビーズ)。 Specifically, streptavidin-coated microspheres (Bangs Laboratories, Inc.; polystyrene, average diameter 9.94 μm) were washed twice with washing buffer (phosphate-buffered saline (PBS), 0.01% bovine serum albumin (BSA), 0.05% TritonX-100) and reacted with biotinylated anti-human IgG (Fc-specific) antibody (Sigma Aldrich; mouse monoclonal) in binding buffer (PBS, 0.01% BSA) to immobilize the biotinylated anti-human IgG (Fc-specific) antibody on the streptavidin-coated microbeads. The resulting beads were washed three times with washing buffer (anti-human IgG Fc antibody beads).
次に抗ヒトIgGFc抗体ビーズを結合緩衝液に懸濁し、そこにヒトLRRTM2の細胞外ドメインとヒトIgGのFc部分との融合タンパク質(LRRTM2-Fc;R&D Systems社)を添加し、LRRTM2-Fcを抗ヒトIgGFc抗体ビーズに固相化した。得られたビーズを洗浄緩衝液で洗浄し、結合緩衝液に懸濁した(LRRTM2ビーズ懸濁液)。 Then, the anti-human IgGFc antibody beads were suspended in binding buffer, and a fusion protein of the extracellular domain of human LRRTM2 and the Fc portion of human IgG (LRRTM2-Fc; R&D Systems) was added thereto, and LRRTM2-Fc was immobilized on the anti-human IgGFc antibody beads. The resulting beads were washed with washing buffer and suspended in binding buffer (LRRTM2 bead suspension).
3.シナプス前部の誘導(図1)
20日間の培養を経たプレートに0.1μg/ウェルになるようにLRRTM2ビーズを播種し、48時間、37℃で培養し、シナプス前部の形成を誘導した。
3. Presynaptic induction (Figure 1)
After 20 days of culture, LRRTM2 beads were seeded at 0.1 μg/well on the plate and cultured at 37° C. for 48 hours to induce the formation of presynapses.
4.コンジュゲートの作製
薬剤としては微小管重合阻害剤であるモノメチルアウリスタチンE(MMAE:MedChemExpress社)を用いた。MMAEと抗体のコンジュゲートをMagicLinkTM kit(BroadPharm社)を用いて作製した。コンジュゲートの作製はメーカーのプロトコルに従って行った。
4. Preparation of conjugates The drug used was monomethyl auristatin E (MMAE: MedChemExpress), a microtubule polymerization inhibitor. Conjugates of MMAE and antibodies were prepared using the MagicLink TM kit (BroadPharm). The conjugates were prepared according to the manufacturer's protocol.
抗体としては、ポリクローナルウサギ抗SYT2 N末端抗体(Polyclonal rabbit purified antibody SYT2 lumenal domain;カタログ番号105 223;Synaptic Systems社)、又は対照として、正常ウサギIgG抗体(Normal Rabbit IgG;カタログ番号AB-105-C;R&D Systems社)を用いた。
The antibodies used were polyclonal rabbit anti-SYT2 N-terminal antibody (Polyclonal rabbit purified antibody SYT2 lumenal domain;
5.コンジュゲート溶液の調製
ニューロン培地を37℃で30分間温めた後、培地に、終濃度が100μMとなるように4-アミノピリジン(Sigma Aldrich社)を添加して混和した。さらに、コンジュゲートを終濃度が1μg/mLとなるように添加して混和した。この粗製コンジュゲート溶液を200gで3分間、室温で遠心し、その上清をコンジュゲート溶液として回収した。
5. Preparation of conjugate solution After the neuronal medium was warmed at 37°C for 30 minutes, 4-aminopyridine (Sigma Aldrich) was added to the medium to a final concentration of 100 μM and mixed. Furthermore, the conjugate was added to a final concentration of 1 μg/mL and mixed. This crude conjugate solution was centrifuged at 200 g for 3 minutes at room temperature, and the supernatant was collected as the conjugate solution.
また、さらなる対照群として、一部のサンプルではコンジュゲートでなくMMAE単体を導入した(サンプル数:13)。この場合、コンジュゲートの代わりに同濃度のMMAEを用いて上述の通りMMAE溶液を調製した。 As an additional control, some samples were loaded with MMAE alone rather than the conjugate (number of samples: 13). In these cases, the MMAE solution was prepared as described above, using the same concentration of MMAE instead of the conjugate.
6.コンジュゲートの導入
シナプス前部を形成させたプレートにて、100μLのコンジュゲート溶液又はMMAE溶液を用いて培地交換を行い、37℃で30分培養することにより、コンジュゲートを導入した。
6. Introduction of Conjugates In the plate in which the presynapse had been formed, the medium was replaced with 100 μL of the conjugate solution or MMAE solution, and the plate was cultured at 37° C. for 30 minutes to introduce the conjugate.
7.軸索の伸長反応
導入の後に溶液を回収して洗浄し、コンジュゲート及びMMAEを含まないニューロン培地に培地交換した。その後、さらに24時間培養することにより、軸索を伸長させた。
After the transfection, the solution was collected and washed, and the medium was replaced with neuronal medium that did not contain the conjugate or MMAE. Then, the cells were cultured for another 24 hours to allow the axons to grow.
8.細胞の固定
培養の後、ニューロン培地を用いて洗浄した後、2%パラホルムアルデヒド(PFA)溶液で固定した。
8. Fixation of cells After culturing, the cells were washed with neuronal medium and then fixed with 2% paraformaldehyde (PFA) solution.
9.免疫細胞化学染色
添加した抗体を一次抗体として、免疫細胞化学染色による染色を行った。固定後の細胞において、界面活性剤による細胞膜の透過処理及びブロッキングをした後、追加の一次抗体としてマウス抗βIIIチューブリン(Tuj1)抗体(カタログ番号801202;Biolegend社)を用いた一次抗体反応を行った。その後、Alexa 555標識抗マウス抗体(カタログ番号A32727;Thermo Fisher Scientific社)を用いた二次抗体反応を行い、蛍光画像を取得した。ブロッキングは、ブロッキング緩衝液(PBS+2%正常ヤギ血清+1% BSA+1%ウシ胎児血清+0.02% TritonX-100)を用いて行った。サンプル数は以下の通り:MMAE単独導入群、13サンプル;対照抗体群、17サンプル;ポリクローナルSYT2抗体群、27サンプル。
9. Immunocytochemical staining Immunocytochemical staining was performed using the added antibody as the primary antibody. After permeabilization and blocking of the cell membrane of the fixed cells with a detergent, primary antibody reaction was performed using mouse anti-βIII tubulin (Tuj1) antibody (catalog number 801202; Biolegend) as an additional primary antibody. Then, secondary antibody reaction was performed using Alexa 555-labeled anti-mouse antibody (catalog number A32727; Thermo Fisher Scientific), and fluorescent images were obtained. Blocking was performed using blocking buffer (PBS + 2% normal goat serum + 1% BSA + 1% fetal bovine serum + 0.02% TritonX-100). The number of samples was as follows: MMAE alone-injected group, 13 samples; control antibody group, 17 samples; polyclonal SYT2 antibody group, 27 samples.
蛍光画像の取得は、LAS Xソフトウェア(Leica社)を搭載した倒立ライブセル(DMi8)顕微鏡蛍光顕微鏡(Leica社)を用いて、以下の励起波長、検出波長、露光時間及び検出閾値で行った。ガンマ補正値はいずれも1を用いた。 Fluorescence images were acquired using an inverted live cell (DMi8) microscope fluorescence microscope (Leica) equipped with LAS X software (Leica) at the following excitation wavelength, detection wavelength, exposure time, and detection threshold. A gamma correction value of 1 was used for all images.
励起波長及び検出波長:Alexa 555 最大励起 555nm;最大検出 580nm;実際の検出 595nm。
露光時間及び検出閾値:Alexa 555 露光時間 50ms;検出閾値 100~3500。
Excitation and detection wavelengths: Alexa 555 maximum excitation 555 nm; maximum detection 580 nm; actual detection 595 nm.
Exposure time and detection threshold: Alexa 555 exposure time 50ms; detection threshold 100-3500.
10.軸索量の解析
軸索量は、取得した蛍光画像におけるTuj1シグナルの輝度の合計値(=(ピクセルあたりの蛍光強度)×(ピクセル数))として取得した。対照の正常ウサギ抗体とMMAEのコンジュゲートを使用した場合の結果を100%として標準化した値を相対軸索量として算出した。輝度の取得はLAS Xソフトウェア(Leica社)を用いて行った。
10. Analysis of axonal volume The axonal volume was calculated as the total brightness of the Tuj1 signal in the acquired fluorescent images (= (fluorescence intensity per pixel) × (number of pixels)). The result when the control normal rabbit antibody and MMAE conjugate was used was set as 100%, and the normalized value was calculated as the relative axonal volume. Brightness was acquired using LAS X software (Leica).
11.統計解析
統計解析は、GraphPad Prism9(GraphPad Software社)を使用してt検定により行った。有意水準としてはp<0.05を用いた。
11. Statistical analysis Statistical analysis was performed by t-test using GraphPad Prism9 (GraphPad Software). The significance level was p<0.05.
結果を図2~4に示す。図2及び3は、対照の正常ウサギ抗体とMMAEのコンジュゲートを使用した対照抗体群(図2)、及びポリクローナル抗SYT2 N末端抗体とのコンジュゲートを使用したSYT2抗体群(図3)の軸索の様子を示す免疫細胞化学染色像である。また、図4はその結果を定量的に示したグラフである。 The results are shown in Figures 2 to 4. Figures 2 and 3 are immunocytochemical staining images showing the appearance of axons in the control antibody group, which used a conjugate of a control normal rabbit antibody and MMAE (Figure 2), and the SYT2 antibody group, which used a conjugate of a polyclonal anti-SYT2 N-terminal antibody (Figure 3). Figure 4 is a graph that quantitatively shows the results.
いずれの実験条件においても、ニューロスフィアから軸索が伸長し、シナプス前部も正常に誘導されていた(図2A及び3A)。 Under all experimental conditions, axons extended from the neurospheres, and presynaptic sites were induced normally (Figs. 2A and 3A).
MMAEは、軸索の伸長及び維持に重要な細胞骨格である微小管の重合を阻害する。そのため、MMAEの効果が強い程、軸索の伸長及び維持が阻害され、軸索量が減少すると予想される。 MMAE inhibits the polymerization of microtubules, a cytoskeleton important for the growth and maintenance of axons. Therefore, the stronger the effect of MMAE, the more axon growth and maintenance is inhibited, and the more axon mass is expected to decrease.
対照抗体群では、ニューロスフィアから遠い遠位部でも多くの太い軸索と、膨らみとして観察されるシナプス前部が見られた(図2B)。また、ニューロスフィアに比較的近い近位部では、軸索が互いに平行に近い状態で整列して走行している様子が観察された(図2C)。 In the control antibody group, many thick axons and presynaptic regions observed as bulges were observed even in the distal area far from the neurosphere (Fig. 2B). In addition, in the proximal area relatively close to the neurosphere, axons were observed running in an almost parallel alignment (Fig. 2C).
一方、SYT2抗体群では、ニューロスフィアから遠い遠位部には少数の細い軸索のみが見られ、シナプス前部の形成も乏しかった(図3B)。また、ニューロスフィアに比較的近い近位部では、軸索の走行が、対照抗体群ほどは整列しておらず、軸索の走行が乱れている様子が観察された(図3C)。 On the other hand, in the SYT2 antibody group, only a few thin axons were observed in the distal area far from the neurosphere, and presynaptic formation was also poor (Figure 3B). Furthermore, in the proximal area relatively close to the neurosphere, the axonal trajectories were not as well aligned as in the control antibody group, and the axonal trajectories appeared to be disorganized (Figure 3C).
定量した結果からも観察結果が裏付けられた。対照抗体群(図4中、「Cont. IgG-MMAE」)と比較して、ポリクローナルSYT2抗体群(図4中、「α-SYT2 IgG-MMAE」)では軸索量は有意に減少し、相対軸索量は約87.15%となった。また、SYT2抗体群の結果は、MMAEを単独で導入した単独導入群(図4中、「MMAE」)と比較しても有意に減少し、軸索量は単独導入群の約82.7%であった。なお、単独導入群と対照抗体群では有意な差は見られなかった。 The quantitative results also supported the observations. Compared to the control antibody group ("Cont. IgG-MMAE" in Figure 4), the axonal volume was significantly reduced in the polyclonal SYT2 antibody group ("α-SYT2 IgG-MMAE" in Figure 4), with the relative axonal volume being approximately 87.15%. The results for the SYT2 antibody group were also significantly reduced compared to the single-infusion group in which MMAE was introduced alone ("MMAE" in Figure 4), with the axonal volume being approximately 82.7% of that in the single-infusion group. No significant differences were observed between the single-infusion group and the control antibody group.
以上の結果から、小胞膜タンパク質の小胞内ドメインに結合可能な抗体を使用することにより、その抗体に結合した生理活性物質等の分子を運動神経細胞に標的化することができ、生理活性物質の場合には、運動神経細胞においてその薬理効果が得られることが示唆された。 These results suggest that by using an antibody capable of binding to the intravesicular domain of a vesicular membrane protein, molecules such as physiologically active substances bound to the antibody can be targeted to motor neurons, and in the case of physiologically active substances, their pharmacological effects can be obtained in motor neurons.
<実施例4:SYNGR3抗体を用いた薬剤の送達による薬理効果の検証>
SYNGR3の小胞内ドメインに結合可能な抗体を用いた場合における、SYT2についてと同様に運動神経細胞内に薬剤を送達した場合の、薬剤に基づく薬理効果を調べた。
Example 4: Verification of pharmacological effects of drug delivery using SYNGR3 antibody
In the case of using an antibody capable of binding to the intravesicular domain of SYNGR3, the pharmacological effects of drugs were examined when drugs were delivered into motor neurons in the same manner as for SYT2.
抗SYNGR3の小胞内ドメイン抗体としてポリクローナルヤギ抗SYNGR3小胞内ドメイン抗体(Synaptogyrin 3 Rabbit anti-Human Polyclonal Antibody;カタログ番号LS-C55548-100;LS Bio社)を使用した以外は実施例3と同様に実験を行った。各群10サンプルずつで実験を行った。 The experiment was performed in the same manner as in Example 3, except that a polyclonal goat anti-SYNGR3 intravesicular domain antibody (Synaptogyrin 3 Rabbit anti-Human Polyclonal Antibody; catalog number LS-C55548-100; LS Bio) was used as the anti-SYNGR3 intravesicular domain antibody. The experiment was performed with 10 samples per group.
結果を図5に示す。図5はポリクローナルヤギ抗SYNGR3小胞内ドメイン抗体を使用した場合の結果を定量的に示したグラフである。 The results are shown in Figure 5. Figure 5 is a graph quantitatively showing the results when polyclonal goat anti-SYNGR3 intravesicular domain antibody was used.
MMAEは、軸索の伸長及び維持に重要な細胞骨格である微小管の重合を阻害する。そのため、MMAEの効果が強い程、軸索の伸長及び維持が阻害され、軸索量が減少すると予想される。 MMAE inhibits the polymerization of microtubules, a cytoskeleton important for the growth and maintenance of axons. Therefore, the stronger the effect of MMAE, the more axon growth and maintenance is inhibited, and the more axon mass is expected to decrease.
有意差は見られなかったものの、対照抗体を用いた場合(図5中、「Cont. IgG-MMAE」)と比較して、ポリクローナルヤギ抗SYNGR3小胞内ドメイン抗体群(図5中、「α-SYNGR3 IgG-MMAE」)では軸索量は減少傾向にあった。 Although no significant difference was observed, the axonal volume tended to decrease when using the polyclonal goat anti-SYNGR3 intravesicular domain antibody group ("α-SYNGR3 IgG-MMAE" in Figure 5) compared to when the control antibody was used ("Cont. IgG-MMAE" in Figure 5).
このことから、SYNGR3の小胞内ドメインに結合可能な抗体を使用することによっても生理活性物質等を運動神経細胞へ送達可能であることが示唆された。 This suggests that it may be possible to deliver physiologically active substances to motor neurons by using antibodies that can bind to the intravesicular domain of SYNGR3.
<実施例5:本発明のモノクローナル抗体を用いた薬剤の送達による薬理効果の検証>
実施例1において取得した本発明のモノクローナル抗体を用いて運動神経細胞内に薬剤を送達した場合の、薬剤に基づく薬理効果に与える影響を調べた。
Example 5: Verification of pharmacological effects of drug delivery using the monoclonal antibody of the present invention
When a drug is delivered into a motor neuron using the monoclonal antibody of the present invention obtained in Example 1, the influence on the pharmacological effect of the drug was examined.
実施例1で取得した本発明のモノクローナル抗体を抗SYNGR3小胞内ドメイン抗体として使用し、対照抗体の代わりにポリクローナルヤギ抗SYNGR3小胞内ドメイン抗体を使用した以外は実施例4と同様に実験を行った。各群6サンプルずつで実験を行った。 The experiment was conducted in the same manner as in Example 4, except that the monoclonal antibody of the present invention obtained in Example 1 was used as the anti-SYNGR3 intravesicular domain antibody, and a polyclonal goat anti-SYNGR3 intravesicular domain antibody was used instead of the control antibody. The experiment was conducted with six samples per group.
結果を図6に示す。図6は本発明のモノクローナル抗体を使用した場合の結果を定量的に示したグラフである。 The results are shown in Figure 6. Figure 6 is a graph quantitatively showing the results when the monoclonal antibody of the present invention was used.
ポリクローナル抗体群(図6中、「Polyclonal Ab」)と比較して、モノクローナルSYNGR3抗体群(図6中、「Monoclonal Ab」)では軸索量は有意に減少した。 Compared to the polyclonal antibody group ("Polyclonal Ab" in Figure 6), the axon mass was significantly reduced in the monoclonal SYNGR3 antibody group ("Monoclonal Ab" in Figure 6).
このことから、本発明のモノクローナル抗体を使用した場合は、有意に効率的に生理活性物質等を運動神経細胞へ送達可能であることが示唆された。 This suggests that when the monoclonal antibody of the present invention is used, it is possible to deliver physiologically active substances, etc. to motor neurons significantly and efficiently.
<実施例6:本発明のモノクローナル抗体のELISA法による親和性の確認>
実施例1において取得した本発明のモノクローナル抗体の抗原への結合親和性を、SYNGR3を細胞膜上に発現する培養細胞を用いたELISA法を用いて調べた。
Example 6: Confirmation of affinity of the monoclonal antibody of the present invention by ELISA method
The binding affinity of the monoclonal antibody of the present invention obtained in Example 1 to an antigen was examined by ELISA using cultured cells expressing SYNGR3 on the cell membrane.
1.細胞の調製
SYNGR3発現細胞は、以下の様に調製した。調製手順の概要は以下の通りである。
1. Preparation of cells SYNGR3-expressing cells were prepared as follows. The outline of the preparation procedure is as follows.
SYNGR3の発現ベクターとしては、実施例1の「7-2.フローサイトメトリーによるスクリーニング」において使用した抗原発現ベクターを用いた。 The antigen expression vector used in "7-2. Screening by flow cytometry" in Example 1 was used as the SYNGR3 expression vector.
トランスフェクション用細胞として、HEK293T細胞(American Type Culture Collection社製)を以下の様に調製したものを使用した。 HEK293T cells (American Type Culture Collection) were used for transfection as prepared as follows:
購入したHEK293細胞を解凍し維持用培地(1% Penicillin-Streptomycin(Thermo Fisher Scientific社製)と10% Fetal Bovine Serum(Cytiva社製)と1% GlutaMAXTM I(Thermo Fisher Scientific社製)を添加したDMEM培地(Thermo Fisher Scientific社製))に再懸濁した。細胞懸濁液を130×g、室温で5分間遠心して上清を除去し、10mL~20mLの維持用培地を添加して細胞を懸濁した。細胞数をカウントし、10cm dish(Greiner Bio-One社)2.0×106細胞/dishの細胞密度で播種した。 Purchased HEK293 cells were thawed and resuspended in maintenance medium (DMEM medium (Thermo Fisher Scientific) supplemented with 1% Penicillin-Streptomycin (Thermo Fisher Scientific), 10% Fetal Bovine Serum (Cytiva), and 1% GlutaMAXTM I (Thermo Fisher Scientific)). The cell suspension was centrifuged at 130 × g for 5 minutes at room temperature to remove the supernatant, and 10 mL to 20 mL of maintenance medium was added to suspend the cells. The number of cells was counted and the cells were seeded at a cell density of 2.0 × 106 cells/dish in a 10 cm dish (Greiner Bio-One).
3日間細胞を培養した後、10cm dishから培地を除去し、PBSで洗浄した。洗浄後に0.1%トリプシン(ナカライテスク社製)を2mL添加し、1.5分~2分間、室温でインキュベートして細胞を剥離して50mLチューブに回収した。回収した細胞を分散させて得られた細胞懸濁液を130×g、室温で5分間遠心して上清を除去し、10mL~20mLの維持用培地を添加して細胞を懸濁した。細胞数をカウントし、トランスフェクションのために1.0×103細胞/wellの細胞密度で96 wellプレート(Azone社製)に播種し、維持用培地中で80~90%コンフルエントになるまで培養した。 After culturing the cells for 3 days, the medium was removed from the 10 cm dish and washed with PBS. After washing, 2 mL of 0.1% trypsin (Nacalai Tesque) was added and incubated at room temperature for 1.5 to 2 minutes to detach the cells, which were then collected in a 50 mL tube. The collected cells were dispersed, and the resulting cell suspension was centrifuged at 130 × g at room temperature for 5 minutes to remove the supernatant, and 10 mL to 20 mL of maintenance medium was added to suspend the cells. The number of cells was counted, and for transfection, the cells were seeded in a 96-well plate (Azone) at a cell density of 1.0 × 103 cells/well and cultured in maintenance medium until 80 to 90% confluent.
トランスフェクション用細胞の培養の後、培地を除去し、トランスフェクション用培地1mLで培地交換を行った。培地交換の後、トランスフェクション用溶液100μLを添加して37℃、5%CO2の存在下にて24時間インキュベートした。 After culturing the cells for transfection, the medium was removed and replaced with 1 mL of medium for transfection. After medium replacement, 100 μL of the transfection solution was added and incubated at 37° C. in the presence of 5% CO 2 for 24 hours.
トランスフェクション用溶液としては、1μg/μLの濃度でプラスミドベクターを含むベクター溶液0.5μLとOpti-MEMTM I Reduced Serum Medium(Thermo Fisher Scientific社製)100μLを混合することにより調製した。 A transfection solution was prepared by mixing 0.5 μL of a vector solution containing a plasmid vector at a concentration of 1 μg/μL with 100 μL of Opti-MEM ™ I Reduced Serum Medium (Thermo Fisher Scientific).
トランスフェクション用培地としては、1μLのLipofectamineTM 2000 Transfection Reagent(Thermo Fisher Scientific社製)を添加したOpti-MEMTMI Reduced Serum Medium培地(Thermo Fisher Scientific社製)を用いた。 The transfection medium used was Opti-MEM ™ I Reduced Serum Medium (Thermo Fisher Scientific) supplemented with 1 μL of Lipofectamine ™ 2000 Transfection Reagent (Thermo Fisher Scientific).
2.ELISA試験
トランスフェクション後の96wellプレートから培地を除去して2%PFA溶液を添加し、室温で15分間かけて固定した後、0.25%のTritonX-100含有PBSを用いた細胞膜の透過処理を行った。
2. ELISA Test After transfection, the medium was removed from the 96-well plate, and 2% PFA solution was added to fix the cells at room temperature for 15 minutes. Then, the cell membrane was permeabilized with PBS containing 0.25% TritonX-100.
透過処理後の細胞をPBSで洗浄し、ブロッキング緩衝液(PBS+2%正常ヤギ血清+1% BSA+1%ウシ胎児血清+0.02% TritonX-100)を各ウェルに添加して、室温で1時間ブロッキングを行った。 After permeabilization, the cells were washed with PBS, and blocking buffer (PBS + 2% normal goat serum + 1% BSA + 1% fetal bovine serum + 0.02% TritonX-100) was added to each well, and blocking was performed at room temperature for 1 hour.
1.5mLチューブに一次抗体の濃度が1μg/mL、500ng/mL、100ng/mLになるように一次抗体液を調製した。溶媒としては測定用培地を用い、一次抗体としては、対照抗体であるポリクローナルヤギ抗SYNGR3小胞内ドメイン抗体又は本発明のモノクローナル抗体を用いた。 Primary antibody solutions were prepared in 1.5 mL tubes so that the concentrations of the primary antibody were 1 μg/mL, 500 ng/mL, and 100 ng/mL. Measurement medium was used as the solvent, and the primary antibody was a control antibody, a polyclonal goat anti-SYNGR3 intravesicular domain antibody, or a monoclonal antibody of the present invention.
ブロッキング後の96wellプレートから培地を除去し、100μL/wellの一次抗体液を各ウェルに添加し、37℃、5%CO2の存在下にて1時間インキュベートした。 After blocking, the medium was removed from the 96-well plate, and 100 μL/well of a primary antibody solution was added to each well, followed by incubation at 37° C. in the presence of 5% CO 2 for 1 hour.
一次抗体反応の後、抗体液を除去してPBSで洗浄し、二次抗体液を添加して室温で1時間インキュベートした。二次抗体液はブロッキング緩衝液を溶媒として用いて調製した。ポリクローナルヤギ抗SYNGR3の小胞内ドメイン抗体に対する二次抗体としてはDonkey anti-Goat 555(カタログ番号#A32814;Thermo Fisher Scientific社)を用い、本発明のモノクローナル抗体に対する二次抗体としてはGoat anti-Mouse 555(カタログ番号#A32723;Thermo Fisher Scientific社)を用いた。また各染色用に、二次抗体液にはDAPI(カタログ番号D1306;Thermo Fisher Scientific社)を添加した。
二次抗体反応の後、抗体液を除去して0.02%のTritonX-100含有PBSを用いて洗浄した。
After the primary antibody reaction, the antibody solution was removed and washed with PBS, and the secondary antibody solution was added and incubated at room temperature for 1 hour. The secondary antibody solution was prepared using blocking buffer as a solvent. Donkey anti-Goat 555 (catalog number #A32814; Thermo Fisher Scientific) was used as the secondary antibody against the polyclonal goat anti-SYNGR3 intravesicular domain antibody, and Goat anti-Mouse 555 (catalog number #A32723; Thermo Fisher Scientific) was used as the secondary antibody against the monoclonal antibody of the present invention. DAPI (catalog number D1306; Thermo Fisher Scientific) was also added to the secondary antibody solution for each staining.
After the secondary antibody reaction, the antibody solution was removed and the sections were washed with PBS containing 0.02% TritonX-100.
3.蛍光シグナルの検出及び測定
蛍光シグナルの測定にはPromega GloMax(Promega Corporation社製)を用いて、以下の励起波長及び検出波長で行った。
DAPI:励起波長 365nm、蛍光波長 415nm~445nm、検出波長 475nm
Alexa 555:励起波長 525nm 蛍光波長 580nm~640nm 検出波長 520nm
3. Detection and Measurement of Fluorescent Signals Fluorescent signals were measured using Promega GloMax (Promega Corporation) at the following excitation and detection wavelengths.
DAPI: Excitation wavelength 365nm, Emission wavelength 415nm-445nm, Detection wavelength 475nm
Alexa 555: Excitation wavelength 525nm, Emission wavelength 580nm-640nm, Detection wavelength 520nm
検出されたAlexa 488シグナルの蛍光強度をDAPIの蛍光強度で除算し、抗SYNGR3抗体の相対蛍光強度とした。さらに、対照抗体を使用した場合の相対蛍光強度を1.0として標準化した値を相対抗体量とした。 The fluorescence intensity of the detected Alexa 488 signal was divided by the fluorescence intensity of DAPI to obtain the relative fluorescence intensity of the anti-SYNGR3 antibody. Furthermore, the relative fluorescence intensity when the control antibody was used was standardized to 1.0 to obtain the relative antibody amount.
結果を図7及び8に示す。図7は100ng/mLの抗SYNGR3抗体を使用した場合の結果を示すグラフであり、図8は500ng/mLの抗SYNGR3抗体を使用した場合の結果を示すグラフである。 The results are shown in Figures 7 and 8. Figure 7 is a graph showing the results when 100 ng/mL of anti-SYNGR3 antibody was used, and Figure 8 is a graph showing the results when 500 ng/mL of anti-SYNGR3 antibody was used.
本発明の抗体は、100ng/mLという低濃度で使用した場合であっても、対照抗体と比較して有意にSYNGR3への結合能が高かった。 The antibody of the present invention had significantly higher binding ability to SYNGR3 than the control antibody, even when used at a low concentration of 100 ng/mL.
この結果から、実施例2にて行った抗体の親和性の評価の結果と一致して、本発明の抗体が優れた抗体であることが確認された。
本明細書で引用した全ての刊行物、特許及び特許出願はそのまま引用により本明細書に組み入れられるものとする。
These results were consistent with the results of the evaluation of antibody affinity performed in Example 2, and confirmed that the antibody of the present invention is an excellent antibody.
All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety.
Claims (15)
配列番号3に示されるアミノ酸配列からなるHCDR1、配列番号4に示されるアミノ酸配列からなるHCDR2、及び配列番号5に示されるアミノ酸配列からなるHCDR3を含む重鎖可変領域、及び
配列番号6に示されるアミノ酸配列からなるLCDR1、配列番号7に示されるアミノ酸配列からなるLCDR2、及び配列番号8に示されるアミノ酸配列からなるLCDR3を含む軽鎖可変領域
を含む、前記抗体。 An antibody capable of binding to the intravesicular domain of synaptogyrin 3,
The antibody comprising a heavy chain variable region comprising an HCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 3, an HCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 4, and an HCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 5, and a light chain variable region comprising an LCDR1 consisting of the amino acid sequence shown in SEQ ID NO: 6, an LCDR2 consisting of the amino acid sequence shown in SEQ ID NO: 7, and an LCDR3 consisting of the amino acid sequence shown in SEQ ID NO: 8.
(a)前記重鎖可変領域が、配列番号1に対して90%以上の配列同一性を有するアミノ酸配列からなり、前記軽鎖可変領域が、配列番号2に対して90%以上の配列同一性を有するアミノ酸配列からなる抗体;
(b)前記重鎖可変領域が、配列番号1において1個若しくは数個のアミノ酸が置換、欠失、及び/又は付加されたアミノ酸配列からなり、前記軽鎖可変領域が配列番号2において1個若しくは数個のアミノ酸が置換、欠失、及び/又は付加されたアミノ酸配列からなる抗体;又は
(c)前記重鎖可変領域が配列番号1に示されるアミノ酸配列からなり、前記軽鎖可変領域が配列番号2に示されるアミノ酸配列からなる抗体。 The antibody of claim 1, which is selected from any one of the following (a) to (c):
(a) an antibody in which the heavy chain variable region consists of an amino acid sequence having 90% or more sequence identity to SEQ ID NO:1, and the light chain variable region consists of an amino acid sequence having 90% or more sequence identity to SEQ ID NO:2;
(b) an antibody in which the heavy chain variable region consists of the amino acid sequence of SEQ ID NO: 1 in which one or more amino acids have been substituted, deleted, and/or added, and the light chain variable region consists of the amino acid sequence of SEQ ID NO: 2 in which one or more amino acids have been substituted, deleted, and/or added; or (c) an antibody in which the heavy chain variable region consists of the amino acid sequence shown in SEQ ID NO: 1 and the light chain variable region consists of the amino acid sequence shown in SEQ ID NO: 2.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2023182983 | 2023-10-25 | ||
| JP2023-182983 | 2023-10-25 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025089345A1 true WO2025089345A1 (en) | 2025-05-01 |
Family
ID=95515752
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2024/037921 Pending WO2025089345A1 (en) | 2023-10-25 | 2024-10-24 | Anti-synaptogyrin 3 antibody |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025089345A1 (en) |
Citations (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2019016123A1 (en) * | 2017-07-17 | 2019-01-24 | Vib Vzw | Targeting synaptogyrin-3 in tauopathy treatment |
| WO2023210585A1 (en) * | 2022-04-25 | 2023-11-02 | 株式会社Jiksak Bioengineering | Targeting agent |
-
2024
- 2024-10-24 WO PCT/JP2024/037921 patent/WO2025089345A1/en active Pending
Patent Citations (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2019016123A1 (en) * | 2017-07-17 | 2019-01-24 | Vib Vzw | Targeting synaptogyrin-3 in tauopathy treatment |
| WO2023210585A1 (en) * | 2022-04-25 | 2023-11-02 | 株式会社Jiksak Bioengineering | Targeting agent |
Non-Patent Citations (1)
| Title |
|---|
| BELIZAIRE ROGER, KOMANDURI CHERYL, WOOTEN KERRY, CHEN MING, THALLER CHRISTINA, JANZ ROGER: "Characterization of synaptogyrin 3 as a new synaptic vesicle protein", THE JOURNAL OF COMPARATIVE NEUROLOGY, vol. 470, no. 3, 8 March 2004 (2004-03-08), US , pages 266 - 281, XP093308929, ISSN: 0021-9967, DOI: 10.1002/cne.20008 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20250277028A1 (en) | Targeting agent | |
| Byun et al. | Stress induces behavioral abnormalities by increasing expression of phagocytic receptor MERTK in astrocytes to promote synapse phagocytosis | |
| CN104781278B (en) | Antibodies against TAU | |
| Pankratova et al. | The S100A4 protein signals through the ErbB4 receptor to promote neuronal survival | |
| EP3723732A1 (en) | Arc protein extracellular vesicle nucleic acid delivery platform | |
| Jiang et al. | Single-domain antibody-based protein degrader for synucleinopathies | |
| Liu et al. | Microglial TMEM119 binds to amyloid-β to promote its clearance in an Aβ-depositing mouse model of Alzheimer’s disease | |
| CN102834107A (en) | Nerve Damage Treatment | |
| WO2025089345A1 (en) | Anti-synaptogyrin 3 antibody | |
| WO2025089344A1 (en) | Anti-synaptic vesicle glycoprotein 2c antibody | |
| WO2025089349A1 (en) | Anti-synaptophysin antibody | |
| WO2025089348A1 (en) | Anti-synaptophysin antibody | |
| WO2025089341A1 (en) | Anti-synaptic vesicle glycoprotein 2c antibody | |
| WO2025089343A1 (en) | Anti-synaptic vesicle glycoprotein 2c antibody | |
| WO2025089342A1 (en) | Anti-synaptic vesicle glycoprotein 2c antibody | |
| WO2025089346A1 (en) | Anti-synaptophysin antibody | |
| WO2025089347A1 (en) | Anti-synaptophysin antibody | |
| WO2025084393A1 (en) | Anti-synaptotagmin 2 antibody | |
| WO2025084396A1 (en) | Anti-synaptotagmin-2 antibody | |
| WO2025084394A1 (en) | Anti-synaptotagmin-2 antibody | |
| WO2024248063A1 (en) | Anti-synaptotagmin 2 antibody | |
| WO2025084397A1 (en) | Anti-synaptotagmin 2 antibody | |
| WO2024248062A1 (en) | Anti-synaptotagmin 2 antibody | |
| WO2025084395A1 (en) | Anti-synaptotagmin 2 antibody | |
| US20240173356A1 (en) | Extracellular Vesicles From Human Induced Pluroptent Stem Cells |
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: 24882455 Country of ref document: EP Kind code of ref document: A1 |