WO2023068328A1 - Agent for inducing decomposition of target peptide - Google Patents
Agent for inducing decomposition of target peptide Download PDFInfo
- Publication number
- WO2023068328A1 WO2023068328A1 PCT/JP2022/039089 JP2022039089W WO2023068328A1 WO 2023068328 A1 WO2023068328 A1 WO 2023068328A1 JP 2022039089 W JP2022039089 W JP 2022039089W WO 2023068328 A1 WO2023068328 A1 WO 2023068328A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- target peptide
- peptide degradation
- binding
- ubiquitin
- degradation
- 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.)
- Ceased
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/02—Peptides of undefined number of amino acids; Derivatives thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
-
- 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
- A61K47/56—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 organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
-
- 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
- A61K47/62—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 a protein, peptide or polyamino acid
-
- 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
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
-
- 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
- A61P25/14—Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
-
- 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
- A61P25/14—Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
- A61P25/16—Anti-Parkinson drugs
-
- 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
- A61P25/28—Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- 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
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
-
- 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
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/115—Aptamers, i.e. nucleic acids binding a target molecule specifically and with high affinity without hybridising therewith ; Nucleic acids binding to non-nucleic acids, e.g. aptamers
-
- 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
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/62—DNA sequences coding for fusion proteins
-
- 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
Definitions
- the present invention relates to a target peptide degradation inducer composed of ubiquitin and a binding factor linked to its C-terminus that binds to the target peptide, and a disease suppressor using the same.
- Non-Patent Document 1 Transcription factors are DNA-binding proteins that regulate gene expression, and dysregulation of their activity causes various diseases including cancer (Non-Patent Document 1). Therefore, transcription factors can be important drug targets in the field of drug discovery. However, most transcription factors do not have active domains like enzymes or ligand binding pockets like receptors. Therefore, it was a target molecule that was difficult to develop (undruggable) with conventional low-molecular-weight compounds and antibody drugs that inhibit the activity of the target molecule or function as an antagonist (Non-Patent Document 2). Therefore, the establishment of a new drug discovery approach targeting transcription factors has been strongly desired from the viewpoint of cell function control and disease treatment.
- Non-Patent Document 3 utilizes the ubiquitin-proteasome system (Ub-PSM system) of the protein degradation mechanism present in all cells as shown in FIG. 11 to induce degradation of target proteins.
- a target protein degradation inducer is a compound consisting of a binder site that binds to a target protein and an E3 ligase, respectively, and a linker site that connects them.
- a target protein degradation inducer is a compound consisting of a binder site that binds to a target protein and an E3 ligase, respectively, and a linker site that connects them.
- the target protein whose ubiquitination is thereby promoted is degraded after being induced to the proteasome.
- PROTAC registered trademark
- target protein degradation inducers promote direct ubiquitination of target proteins by E3 ligases.
- target proteins such as transcription factors are difficult to ubiquitinate by E3 ligases. Therefore, such target proteins remained undruggable target molecules.
- the task of the present invention is to develop and provide a new ubiquitin drug discovery technology that can induce the Ub-PSM system to degrade even target proteins that are difficult to ubiquitinate, such as transcription factors.
- the object of the present invention is to develop and provide a target peptide degradation inducer capable of inducing the degradation of any target protein based on the above new ubiquitin drug discovery technology.
- An object of the present invention is to provide a disease-suppressing agent capable of treating, improving, preventing, etc., a disease by inducing the degradation of even a target protein that causes a disease and has been difficult to develop a drug with the prior art. is.
- the present inventors have conducted intensive research and succeeded in developing a completely new target protein degradation technology using an indirect ubiquitination method.
- the Ub-PSM system is used to degrade the target protein as in the conventional ubiquitin drug discovery technology.
- it does not require a direct ubiquitination process to the target protein via ubiquitin ligase E3, and instead uses a target peptide degradation inducer consisting of a binding factor pre-linked with ubiquitin as shown in FIG.
- a complex of the target protein and the target peptide degradation inducer is formed by binding the binding factor of the target peptide degradation inducer to the target protein.
- the target protein becomes indirectly ubiquitinated in this complex, resulting in recognition by the proteasome as if the target protein had been directly ubiquitinated, followed by degradation.
- the present invention is based on the newly developed technology for degrading target peptides by the indirect ubiquitination method, and provides the following.
- a target peptide degradation inducer linked to the C-terminus of one or more ubiquitins linked to a binding factor that binds to a target peptide (2) The target peptide degradation inducer according to (1), wherein the ubiquitin consists of any one of the amino acid sequences described in (a) to (c) below.
- the target peptide degradation inducer according to any one of (1) to (3), wherein the binding factor is a nucleic acid.
- the ubiquitin and the binding agent are linked via a thiol group introduced by substitution or addition to the C-terminus of ubiquitin, via an azide-alkyne cycloaddition reaction, or via an oxime/hydrazone bond;
- the target peptide degradation inducer according to any one of (1) to (8).
- the target peptide is a transcription factor, amyloid, tau protein, ⁇ -synuclein, caspase, modified huntingtin, pseudokinase, K-RAS mutant, Atg factor, mTOR complex 1, GAPR-1 and orphan receptor
- the target peptide degradation inducer according to any one of (1) to (11), which is selected from the group consisting of: (13)
- An expression vector comprising the polynucleotide of (14).
- a nucleic acid type target peptide degradation inducer comprising the polynucleotide of (14) or the expression vector of (15).
- a target peptide degradation-inducing composition comprising: (18) A Huntington's disease inhibitor containing the target peptide degradation inducer according to (12), wherein the target peptide is denatured huntingtin. (19) An autophagy inhibitor containing the target peptide degradation inducer according to (12), wherein the target peptide is an Atg factor.
- the target peptide degradation inducer of the present invention it is possible to induce the Ub-PSM system to degrade any undruggable target protein that was difficult to ubiquitinate directly in the prior art.
- any disease-causing protein can be degraded via the Ub-PSM system to treat, ameliorate, prevent, etc. the disease.
- FIG. 4 shows a gel shift assay by the binding of Ub-C77-Decoy, which is a target peptide degradation inducer of the present invention, and NF- ⁇ B p50, which is a target peptide.
- FIG. 10 shows the results of Ub-PSM system-mediated degradation of NF- ⁇ B p50 indirectly ubiquitinated by Ub-C77-Decoy.
- MG132 is a proteasome inhibitor
- Decoy DNA and Ub-C77 show only Ub-C77-Decoy binding factor and only ubiquitin, respectively.
- the lower band diagrams are western blotting diagrams showing the degradation results of each sample.
- FIG. 1 shows a gel shift assay by the binding of Ub-C77-Decoy, which is a target peptide degradation inducer of the present invention
- NF- ⁇ B p50 which is a target peptide.
- FIG. 10 shows the results of Ub-PSM system-mediated degradation of NF- ⁇
- FIG. 10 is a diagram showing the survival rate of the target peptide NF- ⁇ B p50 at each concentration of Ub-C77-Decoy.
- the values are shown relative to the amount of NF- ⁇ B p50 immediately after introduction of Ub-C77-Decoy into cells as 100% remaining amount.
- FIG. 3 shows the degradation results of the peptide NF- ⁇ B p50 via the Ub-PSM system.
- FIG. 10 is a diagram showing the survival rate of target peptide NF-kB p65 in cell extracts after addition of UbR-C77-decoy. The figure shows relative values when the amount of NF- ⁇ B p65 remaining when UbR-C77-decoy was not added (Control) was taken as 100%. Residual amounts were calculated as the mean of three experimental results, and error bars indicate standard deviation.
- FIG. 10 is a diagram showing the survival rate of target peptide NF-kB p65 in cell extracts after addition of UbR-C77-decoy. The figure shows relative values when the amount of NF- ⁇ B p65 remaining when UbR-C77-decoy was not added (Control) was taken as 100%. Residual amounts were calculated as the mean of three experimental results, and error bars indicate standard deviation.
- FIG. 10 is a diagram showing the survival rate of target peptide NF-kB p65 in cell extracts after addition of UbR-C77-decoy.
- FIG. 2 is a western blotting diagram showing the results of residual NF- ⁇ B p65 (upper) in Hela cells transfected with UbR-C77-decoy, which is a target peptide degradation inducer of the present invention, and in MCF-7 cells.
- the residual amount of NF- ⁇ B p65 was calculated as a ratio to the band intensity in the western blotting image of the housekeeping protein ⁇ -actin (bottom row).
- a and f are fluorescence images of FAM (fluorescein) showing Ub-C77-Decoy
- b and g are fluorescence images of MCF-7 cells stained with Hoechst 33258
- c and h are images of a and b and f and g, respectively.
- FIG. 4 is a diagram showing the cell viability, which suggests apoptosis induction by inhibition of NF- ⁇ B activity using Ub-C77-Decoy, which is the target peptide degradation inducer of the present invention, as a relative value when blank is 1.0.
- FIG. 2 shows the results of degradation of target peptide streptavidin by Ub-cys-biotin, a low-molecular compound-type target peptide degradation inducer of the present invention, via the Ub-PSM system.
- FIG. 2 is a Western blotting diagram showing the results of degradation of target peptide Mcl-1 by UbR 3 -B6 and UbR 4 -B6, which are peptide-type target peptide degradation inducers of the present invention, via the Ub-PSM system.
- FIG. 2 is a conceptual diagram showing the ubiquitin-proteasome system (Ub-PSM system) of the protein degradation mechanism.
- Ub represents ubiquitin
- E1 represents ubiquitin activating enzyme
- E2 represents ubiquitin conjugating enzyme
- E3 represents ubiquitin ligase.
- ubiquitinated target peptides that are monoubiquitinated and tetraubiquitinated are exemplified as ubiquitinated substrates, but the number of ubiquitins in the ubiquitinated target peptide does not matter.
- BRIEF DESCRIPTION OF THE DRAWINGS It is a conceptual diagram which shows the structure (A) of the target peptide degradation inducer of this invention, and the degradation mechanism (B) of the indirect ubiquitination method of this invention via a Ub-PSM system.
- the binding agent is the binding agent that binds to the target peptide.
- This figure exemplifies a target peptide degradation inducer in which one ubiquitin is linked to the binding factor. may be
- a first aspect of the present invention is a target peptide degradation inducer.
- the target peptide degradation inducer of the present invention has a structure in which a binding factor that binds to a target peptide is linked to the C-terminus of ubiquitin. According to the target peptide degradation inducer of the present invention, the target peptide can be guided to the proteasome by administration in vivo without requiring direct ubiquitination of the target peptide.
- peptide refers to an amino acid polymer in which multiple amino acids are linked by peptide bonds. Any number of amino acids can be linked. Therefore, when the term “peptide” is used herein, it means a generic term including oligopeptides, polypeptides and proteins.
- nucleic acid basically refers to a biopolymer composed of nucleotides linked by phosphodiester bonds.
- DNA linked with deoxyribonucleotides having any of adenine (A), guanine (G), cytosine (C) and thymine (T) bases, and/or adenine, guanine, cytosine and uracil (U) A natural nucleic acid formed by connecting natural nucleotides existing in nature such as RNA to which ribonucleotides having any base are linked corresponds.
- Ubiquitin (herein often abbreviated as “Ub”) is a highly conserved protein in eukaryotes, consisting of 76 amino acids. It is expressed in all cells and functions in various biological phenomena such as protein degradation, signal transduction, DNA repair, and selective autophagy through ubiquitination of target proteins. Proteolytic degradation by the ubiquitin-proteasome system is the primary function of ubiquitin, and in this specification also, without limitation, functions relating to proteolytic degradation are mentioned unless otherwise stated.
- Ubiquitination is also called “ubiquitin modification”, and refers to protein post-translational modification by addition of ubiquitin to a target protein.
- a state in which multiple ubiquitins are linked and added in a chain is particularly referred to as “polyubiquitination”.
- polyubiquitination In polyubiquitinated target proteins, the attached polyubiquitin chains function as degradation signals in the ubiquitin-proteasome system.
- Target protein refers to a protein that is ubiquitinated in the ubiquitin-proteasome system and targeted for degradation. Usually, proteins that are no longer needed in vivo and should be removed correspond.
- Ubiquitin-Proteasome system (herein often referred to as “Ub-PSM system”) is one of the ATP-dependent proteolytic systems that regulate various biological phenomena in vivo. is one.
- Ub-PSM system a series of reactions takes place, including activation of ubiquitin by three enzymes, ubiquitin modification to target proteins, and degradation of ubiquitinated target proteins by the proteasome.
- ubiquitin-activating enzyme (E1) uses ATP to thioester-bond with ubiquitin at its own cysteine (Cys) residue to form an E1-Ub complex.
- ubiquitin in E1-Ub is transferred to the Cys residue of ubiquitin conjugating enzyme E2 via a thioester bond, forming an E2-Ub complex and ubiquitin is activated.
- the activated ubiquitin in the E2-Ub complex is transferred to the target protein by the activity of the heterocomplex ubiquitin ligase E3, and the target protein is ubiquitin-modified.
- an isopeptide bond is formed between the C-terminus of ubiquitin and the lysine (Lys) residue of the target protein.
- target peptide refers to a peptide to be degraded that is induced to the proteasome by the target peptide degradation-inducing agent of the present invention. say. It is substantially synonymous with the target protein. Any type of target peptide can be used. For example, it may be a target protein that is difficult to ubiquitinate directly in a normal Ub-PSM system. Examples of target peptides include, but are not limited to, transcription factors, enzymes, ligands, receptors, constituent proteins, peptide hormones, cytokines, antibodies, antigenic proteins, and the like.
- target peptides include amyloid, tau protein, ⁇ -synuclein, caspase, modified huntingtin, pseudokinase, K-RAS mutant, Atg factor, mTOR complex 1, GAPR-1, and orphan receptor. A body etc. are mentioned.
- binding agent refers to a substance capable of binding to a target peptide. A detailed configuration of the binding factor will be described later.
- living organism refers to living cells, tissues, organs, or individuals.
- the target peptide degradation-inducing agent of the present invention contains a binding factor and ubiquitin as essential constituent factors, and a linker factor as an optional constituent factor. Each component will be specifically described below.
- binding agent refers to a molecule that can bind to a target peptide. Molecules that specifically bind the target peptide are preferred.
- the binding factor may be of any type as long as it has the property of binding to the target peptide. Examples include nucleic acids, peptides, or low-molecular-weight compounds. Each of these will be explained below.
- nucleic acid The binding factor may be composed of nucleic acid. Binding agents composed of nucleic acids are referred to herein as “nucleic acid-type binding agents”.
- nucleic acids correspond to natural nucleic acids, but in the present specification, non-natural nucleic acids can also be included. Accordingly, a nucleic acid-type binding agent can include naturally occurring nucleic acids and/or non-naturally occurring nucleic acids.
- non-natural nucleic acid refers to biopolymers, nucleic acid analogues, etc. containing non-natural nucleotides as structural units.
- non-natural nucleotide refers to any nucleotide other than natural nucleotides. For example, non-naturally occurring nucleotides that are artificially constructed or artificially chemically modified and that have similar properties and/or structures to natural nucleotides are applicable.
- nucleic acid analogue refers to an artificially constructed compound having a structure and/or properties similar to those of naturally occurring nucleic acids.
- crosslinked nucleic acid BNA / LNA: Bridged Nucleic Acid / Locked Nucleic Acid
- morpholino nucleic acid morpholino nucleic acid
- PNA Peptide Nucleic Acid
- methylphosphonate type DNA or RNA phosphorothioate It can also contain type DNA or RNA, phosphoramidate type DNA or RNA.
- nucleic acid-type binding factors include nucleic acid aptamers and nucleic acids that recognize nucleic acid-binding domains.
- nucleic acid aptamer is an aptamer composed of nucleic acid, and the secondary structure and tertiary structure of a single-stranded nucleic acid molecule through hydrogen bonding, etc.
- a ligand molecule that has the ability to bind tightly and specifically. Therefore, in the present specification, a nucleic acid aptamer that strongly and specifically binds to a target peptide is applicable.
- Nucleic acid aptamers are generally known as RNA aptamers and DNA aptamers, but the nucleic acids that constitute the nucleic acid aptamers in the present specification are not particularly limited. Any of DNA aptamers, RNA aptamers, and aptamers composed of a combination of DNA and RNA can be included.
- the nucleic acid aptamer used herein can be produced by a method known in the art with a target peptide as a binding target. Examples thereof include an in vitro selection method using the SELEX (systematic evolution of ligands by exponential enrichment) method.
- the SELEX method is a known method, and a specific method may be performed, for example, according to Pan et al. (Proc. Natl. Acad. Sci. 1995, U.S.A.92: 11509-11513).
- NBD Nucleic acid binding domain recognition nucleic acid
- NBD nucleic acid-Binding Domain
- An NBD can be either a DNA binding domain (DNB) or an RNA binding domain (RNB).
- NBDs examples include, but are not limited to, zinc fingers, leucine zippers, ⁇ -hairpins, helix-turn helices, helix-loop helices, wing-helices, HMG boxes, RRM domains, and TAL effectors. be done.
- nucleic acid binding domain recognition nucleic acid refers to a nucleic acid containing an NBD recognition sequence.
- An NBD-recognizing nucleic acid can bind to the NBD of a target peptide via the included NBD-recognition sequence.
- the NBD-recognizing nucleic acid may consist of any nucleic acid. Examples include naturally occurring nucleic acids consisting of DNA, RNA, or a combination thereof, as well as non-natural nucleic acids in which all or part of them are replaced with non-natural nucleic acids. Furthermore, the NBD-recognizing nucleic acid may consist of a single-stranded nucleic acid, a double-stranded nucleic acid, or a combination thereof, and may have a secondary structure such as a hairpin structure, or a tertiary structure.
- At least the included NBD recognition sequence should consist of a nucleic acid and a sequence that the NBD of the target peptide recognizes and binds to, and there are no particular restrictions on the specific configuration.
- the NBD of the target peptide is a double-stranded DNA binding domain (DNB) that recognizes a specific base sequence
- the NBD-recognizing nucleic acid is composed of double-stranded DNA containing the DNB recognition sequence as the target sequence. All you have to do is
- NBD-recognizing nucleic acids include transcription factor binding sequences contained in promoters.
- a more specific example is an NF- ⁇ B-binding hairpin DNA represented by SEQ ID NO: 2 or 3 and containing a nucleotide sequence to which the transcription factor NF- ⁇ B binds.
- a binding agent may be composed of a peptide. Binding agents composed of peptides are referred to herein as “peptide-type binding agents”.
- Peptide-type binding agents bind to target peptides through peptide-peptide (protein-protein) interactions. This binding is preferably a stable and specific binding.
- the inter-peptide interaction is not limited, but may be a dissociable transient interaction. Such peptide-peptide interactions are achieved, for example, by hydrophobic bonds, van der Waals forces, or salt bridges between the amino acids that make up the peptide.
- the type of the peptide-type binding factor is not limited as long as it is a peptide.
- the peptide-type binding agent may consist of its ligand or its receptor binding site.
- the peptide-type binding agent may consist of its receptor or ligand binding site.
- the peptide-type binding agent may consist of an antibody that specifically recognizes it or an active fragment thereof. Alternatively, it may be composed of commercially available target protein degradation-inducing molecules such as PROTAC (Registered Trademark) (Proteolysis Targeting Chimeric Molecules).
- HXR9 As a more specific example of the peptide-type binding factor, HXR9 (Morgan R. et al., 2007, Cancer Res. 67, 5806-5813).
- the binding agent may be composed of a low-molecular-weight compound.
- a binding agent composed of a low-molecular-weight compound is referred to herein as a “low-molecular-weight compound-type binding agent”.
- Low-molecular-weight compounds generally refer to natural compounds or chemically synthesized compounds with a molecular weight of about several hundred to several thousand.
- the low-molecular-weight compound-type binding agent herein is a low-molecular-weight compound capable of specifically interacting and binding to the target peptide based on the three-dimensional structure of the target peptide. , and types are not particularly limited.
- Specific examples of the low-molecular-weight compound-type binding factor constituting the target peptide degradation inducer of the present invention include methotrexate (MTX), which is an inhibitor against dihydrofolate reductase (DHFR), biotin, etc., which has the ability to specifically recognize avidin. is mentioned.
- MTX methotrexate
- DHFR dihydrofolate reductase
- biotin etc.
- Ubiquitin is a protein consisting of 26 amino acids highly conserved among eukaryotic species, as described above. Therefore, the biological species from which the ubiquitin constituting the target peptide degradation inducer of the present invention is derived is also not limited. It may be ubiquitin that any eukaryote has.
- ubiquitin is a human-derived ubiquitin consisting of the amino acid sequence shown in SEQ ID NO: 1 (MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGG), in which one or more or several amino acids are added, deleted, or substituted in the amino acid sequence shown in SEQ ID NO: 1.
- mutant ubiquitin examples include ubiquitin mutants containing mutations that confer resistance to degradation by deubiquitinating enzymes, although this is not conclusive.
- ubiquitin mutant in the human ubiquitin shown in SEQ ID NO: 1, arginine at position 72 (methionine encoded by the initiation codon is designated as position 1, hereinafter the same) and arginine at position 74 located on the C-terminal side.
- Ubiquitin mutants (R72A/R74T) containing amino acid substitutions in which residues are substituted with alanine and threonine residues, respectively, amino acid substitutions in which arginine residues at positions 72 and 74 are substituted with proline and threonine residues, respectively and a ubiquitin mutant containing an amino acid substitution in which the leucine residue at position 73 is replaced with a proline residue (L73P).
- a specific example of the ubiquitin mutant is a ubiquitin mutant consisting of the amino acid sequence shown in SEQ ID NO: 6 (MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLALTGG).
- a specific example of the ubiquitin mutant is a ubiquitin mutant consisting of the amino acid sequence shown in SEQ ID NO: 7 (MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLPLTGG).
- ubiquitin mutant is a ubiquitin mutant consisting of the amino acid sequence shown in SEQ ID NO: 8 (MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRPRGG).
- plural refers to, for example, 2 to 20, 2 to 15, 2 to 10, 2 to 7, 2 to 5, 2 to 4, or 2 to 3.
- severeal pieces means 2 to 3 pieces.
- substitution (of amino acids) means that among the 20 types of amino acids that make up natural proteins, within a group of conservative amino acids with similar properties such as charge, side chain, polarity, and aromaticity It means replacement.
- uncharged polar amino acids 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
- substitutions within the group of aromatic amino acids Phe, Tyr, Trp.
- Amino acid substitutions within these groups are preferred because they are known to be less likely to cause changes in peptide properties.
- amino acid identity refers to the amino acid sequences of two peptides to be compared, with appropriate gaps inserted in one or both of the amino acid sequences to maximize the number of matching amino acid residues. It refers to the ratio (%) of the number of matching amino acid residues to the number of all amino acid residues when aligned. Alignment of two amino acid sequences for calculating amino acid identity can be performed using known programs such as Blast, FASTA, ClustalW.
- the number of ubiquitins linked to the binding factor is not limited. For example, it may be one, or two or more (for example, 2, 3, 4, 5, 6, 7, 8, or 9) linked polyubiquitin good. When two or more are linked, each ubiquitin may be the same or different. For example, a ubiquitin having the amino acid sequence shown in SEQ ID NO: 1 and a mutant ubiquitin having one substitution in the ubiquitin amino acid sequence having the amino acid sequence shown in SEQ ID NO: 1 may be linked.
- linker factor refers to a molecule (crosslinker) that mediates the linkage between the binding factor and ubiquitin in the target peptide degradation inducer of the present invention.
- crosslinker a molecule that mediates the linkage between the binding factor and ubiquitin in the target peptide degradation inducer of the present invention.
- the binding factor and ubiquitin are indirectly linked via the linker factor.
- linker factor does not matter.
- nucleic acids, peptides, and/or low-molecular-weight compounds may be used.
- the basic configuration of these conforms to the configuration of nucleic acids, peptides, and/or low-molecular-weight compounds described in the aforementioned binding factor. Therefore, in the present specification, a linker factor composed of a nucleic acid is referred to as a "nucleic acid-type linker factor," a linker factor composed of a peptide is referred to as a "peptide-type linker factor,” and a linker factor composed of a low-molecular-weight compound is referred to as a "low-molecular-weight linker factor.” compound-type linker factor”.
- the linker factor may be composed of a combination of different types. Examples include a combination of a nucleic acid-type linker factor and a low-molecular-weight compound-type linker factor. In this case, each linker factor may be linked in tandem, and such linked linker factors are referred to herein as "nucleic acid/low molecular weight compound type linker factors".
- the linker factor may be of the same type as either the binding factor or ubiquitin, or both, or may be of a different type.
- the linker factor when the target peptide degradation-inducing agent of the present invention is composed of a nucleic acid-type binding factor, a linker factor, and ubiquitin, the linker factor may be composed of the same nucleic acid as the nucleic acid-type binding factor, or may be composed of the same peptide as ubiquitin. may be configured. Alternatively, it may be composed of a low-molecular-weight compound different from any of them.
- the linker factor when the linker factor is composed of nucleic acid, the nucleic acid-type binding factor and the linker factor may be synthesized as a single fused nucleic acid strand.
- linker factor that connects a binding factor and ubiquitin via a coordinate bond to a metal.
- a linker factor that connects a binding factor and ubiquitin via a coordinate bond to a metal.
- a linker factor consisting of polyhistidine (His-tag) and nickel-nitrilotriacetic acid (Ni-NTA) is one example.
- His-tag polyhistidine
- Ni-NTA nickel-nitrilotriacetic acid
- linker factors that connect binding factors and ubiquitin via host-guest interactions are included.
- a low-molecular-weight compound-type linker factor consisting of adamantane, a cage-like molecule having the same carbon configuration as diamond, and ⁇ -cyclodextrin, a cyclic oligosaccharide, is one example.
- the linker molecule can be bound or ligated to each within the linker molecule, for example, when direct ligation between the binding factor and ubiquitin is difficult when producing the target peptide degradation inducer of the present invention. By including the configuration, both can be easily connected.
- Binding between Constituent Factors Direct binding between constituent factors in the target peptide degradation-inducing agent of the present invention is not particularly limited.
- the complex formed by the binding of the target peptide degradation inducer of the present invention administered in vivo to the target peptide does not readily dissociate until it is incorporated into the proteasome. Any combination is acceptable.
- Binding between constituent elements means, for example, between a binding agent and ubiquitin, between a binding agent and a linker A connection between factors.
- component-to-component bonds include covalent bonds or chemical bonds such as ionic bonds.
- Others include high affinity non-covalent binding such as the binding between biotin and avidin.
- Bonding between constituent factors can be achieved using methods known in the art, depending on the type and composition of each constituent factor. Examples thereof include bonding via chemical reactions such as nucleophilic addition reactions, nucleophilic substitution reactions, or electrophilic substitution reactions between functional groups present in each factor.
- a functional group that contributes to such bonding is preferably, but not limited to, an active functional group having chemical activity that enables linkage with a partner molecule by covalent bonding or the like.
- a functional group that contributes to binding between constituent factors may be a functional group inherently included in each constituent factor, or may be introduced by addition or substitution as necessary.
- cysteine (C) is added to the C-terminus of ubiquitin, or glycine (G) at position 76, which is the C-terminal amino acid of ubiquitin, is replaced with cysteine to introduce a thiol group, which is an active functional group, to the C-terminus of ubiquitin.
- both functional groups forming a bond between constituent factors are not limited, but it is desirable to select a combination that can form a covalent bond between the functional groups.
- Examples include amino and aldehyde groups, thiol and maleimide groups, azido and ethynyl groups, hydrazine and ketone groups, and hydrazine and aldehyde groups.
- the type of bond between functional groups that form the bond between constituent factors is not limited.
- a bond via a thiol group a bond by an azide-alkyne cycloaddition reaction (Huisgen cycloaddition reaction), an oxime/hydrazone bond, or the like can be mentioned.
- Specific examples of bonding via a thiol group include bonding by Michael addition reaction, thiolene reaction, or thiolyne reaction, or disulfide bond.
- Constituent factors of the target peptide degradation-inducing agent of the present invention may be labeled as necessary.
- a labeling substance known in the art may be used depending on the type of each factor.
- the phosphate group, sugar, and/or base may be labeled.
- the constituent amino acid residues may be labeled.
- the included functional groups may be labeled.
- the type of constituent factor to be labeled is also not limited. However, labeling of the binding agent is convenient, and is preferred because it is direct and convenient when detecting binding to the target peptide. Also, multiple constituent factors may be labeled.
- the labeling position may be appropriately determined according to the characteristics and purpose of use of the labeling substance, and is not particularly limited.
- the 5'-end and/or 3'-end is preferably used as the labeling site.
- the side chain of each amino acid residue, the amino group of the N-terminal amino acid residue, or the carboxy group of the C-terminal amino acid residue is suitable as the labeling site.
- the functional groups it contains can be labeled.
- any substance known in the art can be used as the labeling substance.
- examples thereof include radioisotopes, fluorescent substances, quenchers, chemiluminescent substances, DIG, biotin, magnetic beads and the like.
- Radioisotope refers to an element that emits radiation among isotopes with different mass numbers. Examples include 32P , 3H , 14C .
- Fluorescent substance refers to a substance that has the property of being excited by absorbing excitation light of a specific wavelength and emitting fluorescence when returning to the original ground state.
- fluorescent substance refers to a substance that has the property of being excited by absorbing excitation light of a specific wavelength and emitting fluorescence when returning to the original ground state.
- FITC Rhodamine, Fluorescamine, Fluorescein, Texas Red®, Cy3, Cy5, Cy7, FAM, HEX, JOE, ROX, TET, Bodipy493, NBD, TAMRA, Quasar®, CAL Fluor® ) Red610, SYBR Green [registered trademark], Eva Green [registered trademark], SYTOX Green [registered trademark], and the like.
- Quencher refers to a substance that absorbs the excitation energy of the fluorescent substance and has the property of suppressing fluorescence. Examples include AMRA, DABCYL, BHQ-1, BHQ-2, or BHQ-3.
- a "chemiluminescent substance” is a substance that has the property of emitting the differential energy as light when returning to the ground state after being excited by a chemical reaction. For example, an acridinium ester etc. are mentioned.
- Each constituent factor can be labeled at multiple locations with two or more of the same or different labeling substances.
- a component labeled with a labeling substance can be a useful tool in detecting a target peptide degradation inducer.
- the target peptide can be indirectly ubiquitinated as a whole by binding to the target peptide to form a complex. As a result, it can be directed to the proteasome for degradation without direct ubiquitination of the target peptide.
- target peptides that have been undraggable due to the difficulty of direct ubiquitination can be used as drug discovery targets.
- the field of molecular-targeted medicine will expand dramatically, and it is expected that the options for drug development and design will greatly expand.
- any target peptide can be degraded by the Ub-PSM system by selecting a binding factor that specifically binds to the target peptide.
- the target peptide degradation inducer can be an apoptosis inhibitor.
- the target peptide degradation inducer can be an autophagy inhibitor.
- the target peptide degradation inducer can be an autophagy promoter.
- Nucleic acid type target peptide degradation inducer 2-1 is a nucleic acid-type target peptide degradation inducer.
- the nucleic acid-type target peptide degradation-inducing agent of the present invention is composed of a polynucleotide encoding the target peptide degradation-inducing agent according to the first aspect, which is composed only of peptides, or an expression vector containing the same.
- the nucleic acid-type target peptide degradation inducer of the present invention has relatively high stability, is easy to store, and can amplify an active target peptide degradation inducer after administration in vivo.
- Nucleic acid-type target peptide degradation inducer refers to a target peptide degradation inducer composed of a nucleic acid.
- the target peptide degradation-inducing agent according to the first aspect is composed only of a peptide
- the nucleic acid-type target peptide degradation-inducing agent is composed of a polynucleotide encoding the amino acid sequence, or an expression vector containing the polynucleotide.
- Polynucleotides here are, in principle, natural polynucleotides. Although composed of either DNA and/or RNA, polynucleotides composed solely of DNA are preferred.
- a target peptide degradation inducer composed only of a peptide corresponds to a peptide-type binding factor and ubiquitin, or a target peptide degradation inducer composed of a peptide-type binding factor, a peptide-type linker factor, and ubiquitin.
- the term "expression vector” refers to an expression unit that contains a gene or gene fragment (in the present invention, the polynucleotide) in an expressible state and can control the expression of the gene or the like.
- the term “expressible state” refers to a state in which the gene or the like is placed under the control of a promoter and the expression of the gene or the like can be induced by activation of the promoter.
- Any type of expression vector can be used. Any expression vector whose promoter can be activated in vivo in a subject to whom the nucleic acid-type target peptide degradation inducer of the present invention is administered can be used.
- viral vectors are included.
- Viral vectors include various vectors derived from retroviruses (including lentiviruses), adenoviruses, adeno-associated viruses, and the like.
- the functions and effects of the nucleic acid-type target peptide degradation inducer of this aspect conform to those of the target peptide degradation inducer described in the first aspect. However, due to the structural difference that they are composed of nucleic acids, their mechanisms of action are different. That is, the target peptide degradation-inducing agent according to the first aspect does not itself have the activity of inducing degradation of the target peptide, and acts directly on the target peptide after administration to the living body, whereas the nucleic acid-type target peptide degradation-inducing agent of this aspect has no activity to induce degradation of the target peptide.
- the target peptide degradation inducer encoded by the nucleic acid-type target peptide degradation inducer is expressed intracellularly, and the expressed target peptide degradation inducer acts on the target peptide. This makes it possible to amplify the target peptide degradation inducer in vivo.
- a third aspect of the present invention is a target peptide degradation-inducing composition.
- the composition of the present invention includes any two or more of the target peptide degradation inducer according to the first aspect and/or the nucleic acid-type target peptide degradation inducer according to the second aspect as essential constituents.
- a target peptide degradation-inducing agent or the like can be stably provided as a pharmaceutical composition in a form that reduces the burden and invasiveness upon administration to the living body and is easier to administer. can be done.
- the target peptide degradation-inducing composition of this embodiment (hereinbelow, often abbreviated as "this composition") comprises an active ingredient as an essential constituent factor, and a carrier and/or Contains solvent. Each component will be specifically described below.
- the present composition comprises the target peptide degradation inducer according to the first aspect and/or the nucleic acid-type target peptide degradation inducer according to the second aspect (in this specification, these are collectively referred to as "POI degradation (referred to as “inducing agent, etc.") as an essential active ingredient.
- POI degradation referred to as "inducing agent, etc.
- the present composition can contain two or more different POI degradation inducers and the like.
- other known target protein degradation inducers such as PROTAC (registered trademark) may be selectively included as active ingredients.
- the content of the POI decomposition inducer, etc. contained in the composition includes the type and/or effective amount of the POI decomposition inducer, etc., the dosage form of the composition, the type of carrier or additive described later, and Since it varies depending on the type of disease, it may be determined appropriately in consideration of each condition.
- the term "effective amount” refers to an amount necessary for the POI decomposition inducer, etc., as an active ingredient in the present composition, to exhibit its function, and to cause no harmful side effects to the living body to which it is applied. , refers to the amount that occurs little or not at all. This effective amount may vary depending on various conditions such as subject information, route of administration, and frequency of administration.
- the term "subject” refers to a living organism to which the POI degradation inducer or the like or the present composition is applied.
- humans livestock (cows, horses, sheep, goats, pigs, chickens, ostriches, etc.), racehorses, pet animals (dogs, cats, rabbits, etc.), laboratory animals (mouse, rats, guinea pigs, monkeys, etc.), etc. Applicable. Humans are preferred (in this case, they are particularly referred to as "subjects").
- subject information refers to various individual information of the living body to be applied.
- the effective amount of the POI degradation inducer, etc. in this composition and the applicable amount calculated based thereon are ultimately determined by doctors, dentists, veterinarians, etc., depending on the information of individual subjects. determined by
- composition can contain a pharmaceutically acceptable solvent. Solvents are optional constituents in the composition and may be added as needed. “Pharmaceutically acceptable” means harmless or low toxicity to living bodies, and can be used normally in the field of formulation technology, preferably in pharmaceutical compositions.
- Solvents include, for example, water or aqueous solutions, or organic solvents.
- Pharmaceutically acceptable aqueous solutions include, for example, physiological saline, isotonic solutions containing glucose and other adjuvants, phosphate buffers, sodium acetate buffers.
- adjuvants here include D-sorbitol, D-mannose, D-mannitol, sodium chloride, low-concentration nonionic surfactants, polyoxyethylene sorbitan fatty acid esters, and the like.
- Pharmaceutically acceptable organic solvents include, for example, ethanol, butanol, and the like.
- Carrier can contain a pharmaceutically acceptable carrier.
- a carrier is an optional component in the composition and may be added as needed.
- Carriers include, for example, suspending agents, diluents, solubilizers, dispersing agents, surfactants, emulsifying agents, soothing agents, stabilizers, preservatives, preservatives, antioxidants, buffers, and tonicity agents. agents and the like.
- excipients fillers, binders, disintegrants, absorption enhancers, bulking agents, humectants, humectants, humectants, adsorbents, and disintegration inhibitors commonly used in pharmaceuticals, if necessary Agents, coating agents, coloring agents and the like may also be included as appropriate.
- Such a carrier is mainly used to facilitate the formation of a dosage form, maintain the dosage form and drug effect, and make the POI degradation inducer, etc., which is an active ingredient, difficult to be decomposed in vivo. It should be used appropriately as needed.
- the dosage form of the present composition does not inactivate the POI degradation inducer and other known target protein degradation inducers, which are active ingredients, and can exhibit the pharmacological effect of the active ingredient in vivo after administration. There is no particular limitation as long as it is in a form. Moreover, the specific dosage form of the present composition may be appropriately selected according to the administration method and/or prescription conditions. In general, administration methods can be broadly classified into oral administration and parenteral administration, and the present composition may be formulated in dosage forms suitable for each administration method.
- the dosage forms include solids (tablets, pills, sublinguals, capsules, drops), granules, powders, powders, liquids (internal liquids, suspensions, emulsions, syrups). including agents) and the like.
- Solid formulations can optionally be in the form of coatings known in the art, such as sugar-coated tablets, gelatin-coated tablets, enteric-coated tablets, film-coated tablets, double tablets, and multi-layer tablets. .
- Parenteral administration can be divided into systemic administration and topical administration, and local administration can be further subdivided into tissue administration, transepidermal administration, transmucosal administration, and transrectal administration.
- the composition may also be formulated in dosage forms suitable for each administration method.
- dosage forms suitable for systemic administration or intra-tissue administration include liquid injections.
- dosage forms suitable for transepidermal or transmucosal administration include liquids (including ointments, eye drops, nasal drops, and inhalants), suspensions (including emulsions and creams), and powders (nasal drops). , inhalants), pastes, gels, ointments, plasters, and the like.
- Dosage forms suitable for rectal administration include suppositories and the like.
- the present composition can be applied for treatment or prevention of various diseases as described in the fourth aspect.
- the target site for application varies depending on the target disease.
- the target disease is a neurodegenerative disease, it will mainly be the cerebrospinal cord, which is the central nervous system. Therefore, the method of administration is not limited, but intratissue administration or systemic administration via the circulatory system can be used. Specific examples include intratissue administration by intracerebrospinal injection, or intracirculatory administration such as intravascular injection (including intravenous injection and intraarterial injection) or intralymphatic injection.
- the application target sites are mainly primary lesions and metastatic lesions. In this case, too, the method of administration is not limited, but intra-tissue administration to primary lesions and metastatic lesions or systemic administration via the circulatory system can be used.
- Injectables can be prepared by appropriately combining the aforementioned emulsifying agents, suspending agents, surfactants, stabilizers, pH adjusters, etc., and mixing them in a unit dose form generally required for pharmaceutical practice. They are often provided in unit dose ampoules or in multi-dose containers.
- each dosage form described above are not particularly limited as long as they are within the range of dosage forms known in the art for each dosage form.
- a fourth aspect of the present invention is a disease suppressing agent.
- the disease-suppressing agent of the present invention is one form of the target peptide degradation-inducing agent according to the first aspect, and has a specific disease-causing protein as a target peptide.
- the disease-suppressing agent of the present invention can treat or prevent various diseases by using proteins that cause various diseases as target peptides and inducing their degradation.
- nucleic acid-binding proteins that are considered undruggable in ubiquitin drug discovery, such as transcription factors, can be targeted for degradation.
- nucleic acid-binding proteins that are considered undruggable in ubiquitin drug discovery, such as transcription factors
- transcription factors such as transcription factors
- the options in the development and design of molecular-targeted drugs are expanded, and a wide range of drug discovery and drug discovery technologies for various diseases can be provided.
- disease suppressing agent refers to an agent that suppresses the onset of a specific disease, or alleviates or treats the aggravation or chronicity of a specific disease.
- the disease suppressor of the present invention contains a binding factor and ubiquitin as essential constituent factors, and a linker factor as an selective constituent factor. Its basic configuration conforms to the target peptide degradation inducer described in the first aspect. Therefore, the specific description of the common configuration is omitted, and the characteristic points of the disease suppressing agent of the present invention are described here.
- the disease-suppressing agent of the present invention is characterized in that the binding factor binds to the disease-causing peptide.
- the disease-causing peptide is the target peptide in the disease-suppressing agent of this embodiment.
- disease-causing peptide refers to a peptide (including protein) that causes the onset, exacerbation, or chronicity of a disease.
- disease-causing peptides include wild-type peptides as well as mutant peptides such as gain-of-function (activation), hyperfunction, and loss-of-function.
- target peptides include transcription factors, amyloid, tau protein, ⁇ -synuclein, caspases, denatured huntingtin, pseudokinases, K-RAS mutants, Atg factors, mTOR complex 1, GAPR-1 and orphans.
- Diseases caused by receptors and the like, including receptors, can all be targets of the disease-suppressing agent of the present invention.
- the target peptide degradation inducer induces degradation by the Ub-PSM system by indirectly ubiquitylating the target peptide via a binding factor. Therefore, by selecting a binding factor that binds to a disease-causing peptide, it can be used as a therapeutic or preventive drug for various diseases.
- the relationship between the target disease of the disease-suppressing agent of the present invention and the disease-causing peptide will be described below with some examples.
- the disease inhibitor of the present invention can be an anticancer agent.
- the disease-suppressing agent comprises a binding agent that binds to a cancer-causing disease-causing peptide.
- diseases-causing peptides include transcription factors such as NF- ⁇ B, pseudokinases such as HER3 (Xie T., et al., 2014, Nat. Chem. Biol., 10: 1006). -1011), K-RAS mutants (Bond MJ, et al., 2020, ACS Cent. Sci. 6: 1367-1375), or orphan receptors such as ERR- ⁇ , but are not limited to do not.
- the composition of an anticancer drug when NF- ⁇ B is used as a disease-causing peptide Inhibition of NF- ⁇ B activity is known to promote apoptosis induction by TNF- ⁇ (Tang F., et al., 2002, Mol. Cell. Biol., 22: 8571-8579). Therefore, by selecting a binding factor that specifically binds to NF- ⁇ B, the disease suppressor of the present invention can function as an apoptosis inducer. By applying this apoptosis inducer to cancer cells, it can function as an anticancer agent.
- the specific composition of the binding factor that binds to NF- ⁇ B is not limited. p50 and p52 belonging to class I of NF- ⁇ B and p65 (RelA), c-Rel and RelB belonging to class II may be configured to bind, or a configuration that specifically binds to any of them may be used.
- can be Examples thereof include nucleic acid-type binding agents consisting of a nucleic acid sequence to which NF- ⁇ B binds. More specifically, for example, the NF- ⁇ B-binding hairpin DNA represented by SEQ ID NO: 2 or 3.
- This NF- ⁇ B-binding hairpin DNA is NF- ⁇ B Decoy DNA containing a nucleotide sequence mimicking the target nucleotide sequence of NF- ⁇ B.
- the disease suppressor of the present invention can be an agent for preventing Alzheimer's disease (AD) or for suppressing its progress.
- the disease-suppressing agent comprises a binding agent that binds to a disease-causing peptide that causes Alzheimer's disease.
- Alzheimer's disease begins with the aggregation and accumulation of ⁇ -amyloid protein in neurons in the brain, followed by hyperphosphorylation of tau protein, a microtubule protein, resulting in fibrosis, followed by destruction of neurons and brain atrophy. . Therefore, ⁇ -amyloid, tau protein, ⁇ -synuclein, and the like can be applicable as disease-causing peptides for Alzheimer's disease. Therefore, by selecting a binding factor that specifically binds to ⁇ -amyloid, tau protein, ⁇ -synuclein, or the like, the disease inhibitor of the present invention can function as an Alzheimer's disease inhibitor.
- the specific composition of the binding factor that binds to ⁇ -amyloid and tau protein is not limited. Examples thereof include low-molecular-weight compounds capable of binding to fibrillar ⁇ -amyloid or tau protein. More specific examples include thioflavin T (ThT) as a binding factor for ⁇ -amyloid aggregates and pyridoindole as a binding factor for tau protein.
- ThT thioflavin T
- the disease suppressor of the present invention can be a prophylactic agent for Parkinson's disease (PD) or an agent for suppressing the progression thereof.
- the disease-suppressing agent comprises a binding agent that binds to a disease-causing peptide that causes Parkinson's disease.
- the onset of Parkinson's disease is believed to be related to neuronal cell death due to multimerization and accumulation of misfolded ⁇ -synuclein, which constitutes protein aggregates called Lewy bodies. Therefore, misfolded mutant ⁇ -synuclein and the like can be applicable as disease-causing peptides for Parkinson's disease. Therefore, by selecting a binding factor that specifically binds to mutant ⁇ -synuclein, the disease suppressing agent of the present invention can function as a Parkinson's disease suppressing agent.
- the specific configuration of the binding factor that binds to mutant ⁇ -synuclein is not limited. Examples thereof include short peptides such as partial sequences of ⁇ -synuclein that selectively bind to ⁇ -synuclein. More specifically, ⁇ syn36 consisting of the amino acid sequence (GVLYVGSKTR) shown in SEQ ID NO: 5 (Shaltiel-Karyo R., et al., 2010, PLoS One, 5, e13863.; Fun X., et al. ., 2014, Nat. Neurosci., 17, 471-480.).
- GVLYVGSKTR amino acid sequence
- the agent for suppressing disease of the present invention can be an agent for preventing Huntington's disease (HD) or suppressing its progression.
- the disease-suppressing agent comprises a binding agent that binds to the disease-causing peptide responsible for Huntington's disease.
- Huntington's disease is known to be involved in the onset of mutated huntingtin (mHTT), which is caused by the expression of the mutated huntingtin (mHTT) gene with an expanded number of CAG repeats and induces neuronal dysfunction and cell death. (Tomoshige S., et al., 2017, Angew. Chem. Int. Ed., 56, 11530-11533). Therefore, by selecting a binding factor that specifically binds to mutant huntingtin, the disease-suppressing agent of the present invention can function as a Huntington's disease-suppressing agent.
- the specific composition of the binding factor that binds to mutant huntingtin is not limited. Examples include low-molecular-weight compounds that bind to mutant huntingtin multimers and aggregates. More specifically, for example, benzothiazole aniline (BTA) or phenyldiazenyl benzothiazole (PDB) (Tomoshige S., et al., 2017, Angew. Chem. Int. Ed., 56, 11530- 11533), etc.
- BTA benzothiazole aniline
- PDB phenyldiazenyl benzothiazole
- target peptide degradation inducer of the present invention will be specifically described below with reference to examples. However, the configuration and the like described here are only one specific example of the target peptide degradation inducer of the present invention, and do not limit the scope thereof.
- Example 1 Preparation of nucleic acid type target peptide degradation inducer> (the purpose) A target peptide degradation inducer of the present invention is prepared, and it is verified that the target peptide is captured by the target peptide degradation inducer.
- NF- ⁇ B was selected as the target peptide
- two target peptide degradation inducers Ub-cys-Decoy and Ub-C77-Decoy
- ubiquitin-cysteamine Ub-cys
- cysteamine used for linkage with a binding factor was added to the C-terminus of ubiquitin.
- 250 ⁇ M ubiquitin, 2 ⁇ M E1, 2 mM ATP, 5 mM cysteamine, and 5 mM TCEP in 1 mL (final volume) of buffer containing 100 mM Tris-HCl (pH 7.5), 250 mM sucrose, 50 mM KCl, 3 mM MgCl at 4 °C. Incubated overnight.
- the reaction mixture was subsequently purified by Superdex TM 75 Increase 10/300GL size exclusion chromatography (Cytiva). After that, the desired Ub-cys was eluted with 1.5 column volumes of S buffer (20 mM NaPO 4 , pH 7.2, 150 mM NaCl).
- NF- ⁇ B decoy DNA 1 eq
- NF- ⁇ B decoy DNA two types of polynucleotides shown in SEQ ID NO: 2 or 3 below were used.
- NF- ⁇ B ODN1 (27mer) 5′(amino)-AGGGAAATCCCAAAATGGGATTTCCCT-3′(FAM): SEQ ID NO: 2
- NF- ⁇ B ODN2 (28mer) 5′(amino)-AGGGGATTCCCAAAATGGGAATTCCCCT-3′(FAM): SEQ ID NO: 3
- NF- ⁇ B Decoy DNAs have been shown to form a hairpin structure and be recognized and bound by NF- ⁇ B. In addition, both are labeled with FAM (fluorescein) at the 3'.
- FAM fluorescein
- Ub-cys Ub-cysteamine
- 10 mM TCEP Flujifilm Wako Pure Chemical Industries, Ltd.
- a target peptide degradation inducer of the present invention consisting of ubiquitin and NF- ⁇ B Decoy DNA.
- the target peptide degradation inducer of the present invention was prepared using a Ub-C77 mutant (UbR-C77) in which cysteine was added as the 77th amino acid to the C-terminus of the ubiquitin mutant shown in SEQ ID NO: 6. prepared.
- Ub-C77 and UbR-C77 with a cysteine added to the C-terminus were prepared by expression in E. coli as a host using a pET-26b(+) plasmid containing genes encoding Ub-C77 and UbR-C77. .
- NF- ⁇ B Decoy DNA-SMCC was prepared according to the method (2) above.
- Ub-C77-Decoy was purified by anion exchange chromatography HiTrap Q HP (GE Healthcare). The column was equilibrated with 5 column volumes of buffer A (50 mM ammonium acetate (pH 4.5), 0.1 mM EDTA) and the desired Ub-C77-Decoy was eluted with a stepwise gradient of NaCl (200 mM, 450 mM, 500 mM). bottom. The eluate was concentrated and dialyzed against Ub storage buffer (25 mM NaPO4 (pH 7.2), 50 mM NaCl). Yield was 14.4%.
- Example 2 Gel shift assay by binding of Ub-C77-Decoy and NF- ⁇ B p50> (the purpose) It is confirmed by gel shift assay that the target peptide degradation inducer Ub-C77-Decoy of the present invention prepared in Example 1 binds to the target peptide NF- ⁇ B p50.
- Example 3 Target peptide NF- ⁇ B p50 degradation assay (1)> (the purpose) It is confirmed that the target peptide is degraded by binding to the target peptide degradation inducer of the present invention prepared in Example 1 without direct ubiquitination in the presence of proteasome.
- NF- ⁇ B p50/Ub-C77-Decoy was added with 100 nM 26S proteasome (gift from Dr. Yasushi Saeki, Tokyo Metropolitan Institute of Medical Science), 2 mM ATP, and 5 mM MgCl 2 and incubated at 37° C. for 60 minutes. . In the meantime, a portion of the mixed solution at the start of incubation was taken.
- the mixed solution after reaction was mixed with SDS sample buffer (62.5mM Tris-HCl (pH6.8), 2% SDS, 10% Glycerol, 5% 2-mercaptoethanol, 0.002% Bromophenol blue) and After heating for 5 minutes, they were separated by SDS-PAGE. After Western blotting by a standard method, the resulting membrane was incubated with primary antibodies (Anti NFKB1, p105, p50-Specific: Proteintech) diluted 1:500 in blocking solution for 1.5 hours with agitation.
- SDS sample buffer 2.5mM Tris-HCl (pH6.8), 2% SDS, 10% Glycerol, 5% 2-mercaptoethanol, 0.002% Bromophenol blue
- the target peptide degradation inducer of the present invention indirectly ubiquitinates the target peptide by binding to the target peptide, and degrades the target peptide using the proteasome as in the conventional Ub-PSM system. proved to be possible. Although not shown, similar decomposition results were obtained when Ub-cys-Decoy was used.
- Example 4 Verification of NF- ⁇ B p50 decomposition rate> (the purpose) The target peptide degradation rate by the target peptide degradation inducer of the present invention is verified.
- target peptides are different, indirect ubiquitination of target peptides by binding of the target peptide degradation-inducing agent of the present invention can be used as a signal to mediate efficient degradation by the proteasome compared to conventional direct ubiquitination. It was clarified that the target peptide could be decomposed with a similar degree of decomposition rate.
- NF- ⁇ B p50 degradation assay (2)> (the purpose) We will verify the ability of UbR-C77-Decoy with mutated ubiquitin to induce target peptide degradation.
- NF- ⁇ B p65 degradation assay in cell extract > (the purpose) Degradation of the cell-derived target peptide NF- ⁇ B p65 contained in the cell extract by indirect ubiquitination of UbR-C77-Decoy is verified.
- the residual amount was obtained as a relative value when the band intensity of NF- ⁇ B p65 when UbR-C77-decoy was not added was taken as 100%. At that time, the band intensity of NF- ⁇ B p65 was normalized by the band intensity of ⁇ -actin.
- Example 7 Target peptide NF- ⁇ B p65 degradation assay in living cells> (the purpose) To verify the degradation induction of NF- ⁇ B p65 by UbR-C77-Decoy in living cells. (Method) MCF-7 cells (RIKEN BioResource Research Center) and HeLa cells (RIKEN BioResource Research Center) were plated at 3 ⁇ 10 4 cells/well in a 48-well plate and cultured at 37°C under 5% CO 2 for 24 hours. and grown to 80% confluency. Next, after replacing the medium with Opti-MEM, 1 ⁇ M UbR-C77-decoy was introduced into the cells by lipofection using Lipofectamine TM LTX (Thermo Fisher Scientific).
- the medium was replaced with DMEM (10% FBS, 0.5% PS), and TNF- ⁇ (final concentration 20ng/mL) and protein translation material cycloheximide (CHX)/0.5% DMSO (final concentration 50 ⁇ g/mL) were added. was added to the medium and cultured at 37°C under 5% CO 2 for 4 hours. After culturing, cells were lysed with RIPA buffer (Nacalai tesque) and analyzed for NF- ⁇ B p65 by Western blotting. At that time, the band intensity of NF- ⁇ B p65 was normalized by the band intensity of ⁇ -actin.
- Example 8 Apoptosis induction using Ub-C77-Decoy> (the purpose) Inhibition of NF- ⁇ B activity promotes apoptosis induction by TNF- ⁇ as described above. Therefore, it is verified that apoptosis is induced by degradation of NF- ⁇ B by Ub-C77-Decoy, which is the target peptide degradation inducer of the present invention.
- DMEM penicillin/streptomycin
- the transfected cells were incubated at 37°C under 5% CO 2 for 2 hours, stained with Hoechst33258, incubated again at 37°C under 5% CO 2 for 2 hours, and then subjected to fluorescence microscopy (IX83: OLYMPUS ) to observe nuclear-stained cells.
- Ub-C77-Decoy was introduced by lipofection from images (c and h) in which the fluorescence diagrams (b and g) derived from Hoechst33258 used for nuclear staining and the fluorescence (a and f) derived from FAM were superimposed. It was also suggested that some of the translocated cells translocated not only into the cell but also into the nucleus (c).
- Example 9 Preparation of low-molecular compound-type target peptide degradation inducer> (the purpose)
- the target peptide degradation inducer Ub-cys-biotin of the present invention is prepared using biotin, which is a low-molecular-weight compound, as a binding factor.
- Ub-cys-biotin conjugate using Ub-Cys
- cysteamine to Ub
- Ub-cys-biotin was prepared by Michael addition reaction between Ub-cys and carbonyl acryl biotin prepared in (1) above. Specifically, 187 ⁇ M Ub-Cys and 938 ⁇ M 3-carbonylacrylbiotin were mixed in 20 mM Tris-HCl (pH 8.0) and reacted at 25° C. for 3 hours. The desired product was then purified by Superdex TM 75 Increase 10/300GL size exclusion chromatography (Cytiva). The desired Ub-cys-biotin was eluted using 1.5 column volumes of Tris buffer (20 mM Tris-HCl, 150 mM NaCl, pH 8.0).
- Example 10 Target Peptide Degradation Assay Using Low-Molecular-Type Target Peptide Degradation Inducing Agent> (the purpose) The degradation induction of the target peptide (streptavidin) by the low-molecular compound-type target peptide degradation inducer (Ub-cys-biotin) prepared in Example 9 is verified.
- a target peptide degradation inducer of the present invention is prepared using a peptide ligand as a binding agent.
- the ligand peptide which is a binding factor, is a selective binding peptide to Mcl-1, which is an anti-apoptotic protein, and B6 peptide (Journal of Biological Chemistry, 2009, 284 , 31315-31326.) was used.
- UbR 3 -B6 or UbR 4 -B6 was prepared by introducing an expression vector in which a gene encoding each full-length peptide was introduced into a pET15b plasmid into Escherichia coli and expressing it.
- Example 12 Target Peptide Degradation Assay Using Peptide-Type Target Peptide Degradation Inducing Agent> (the purpose) Degradation induction of the target peptide (Mcl-1) by the peptide-type target peptide degradation inducers (UbR 3 -B6 and UbR 4 -B6) prepared in Example 11 is verified.
- the target peptide degradation-inducing agent of the present invention can induce degradation of the target peptide even when a peptide is used as the binding factor.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- General Health & Medical Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- Biomedical Technology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Molecular Biology (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- Epidemiology (AREA)
- General Engineering & Computer Science (AREA)
- Wood Science & Technology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Neurology (AREA)
- Neurosurgery (AREA)
- Microbiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Plant Pathology (AREA)
- Immunology (AREA)
- Physics & Mathematics (AREA)
- Gastroenterology & Hepatology (AREA)
- Toxicology (AREA)
- Psychology (AREA)
- Psychiatry (AREA)
- Hospice & Palliative Care (AREA)
- Mycology (AREA)
- Peptides Or Proteins (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Description
本発明はユビキチンとそのC末端に連結された、標的ペプチドに結合する結合因子で構成される標的ペプチド分解誘導剤、及びそれを用いた疾患抑制剤に関する。 The present invention relates to a target peptide degradation inducer composed of ubiquitin and a binding factor linked to its C-terminus that binds to the target peptide, and a disease suppressor using the same.
転写因子は遺伝子の発現を調節するDNA結合タンパク質であり、その活性制御異常は、癌をはじめとする様々な疾患を引き起こす(非特許文献1)。したがって、転写因子は、創薬分野において重要な創薬ターゲットとなり得る。ところが、大多数の転写因子は酵素のような活性ドメインやレセプターのようなリガンド結合ポケットが存在しない。そのため、標的分子の活性を阻害し、又はアンタゴニストとして機能する従来の低分子化合物や抗体医薬では創薬困難(アンドラッガブル)な標的分子であった(非特許文献2)。したがって、転写因子を標的とする新規創薬アプローチの確立が細胞の機能制御、及び疾病治療の観点から強く求められていた。 Transcription factors are DNA-binding proteins that regulate gene expression, and dysregulation of their activity causes various diseases including cancer (Non-Patent Document 1). Therefore, transcription factors can be important drug targets in the field of drug discovery. However, most transcription factors do not have active domains like enzymes or ligand binding pockets like receptors. Therefore, it was a target molecule that was difficult to develop (undruggable) with conventional low-molecular-weight compounds and antibody drugs that inhibit the activity of the target molecule or function as an antagonist (Non-Patent Document 2). Therefore, the establishment of a new drug discovery approach targeting transcription factors has been strongly desired from the viewpoint of cell function control and disease treatment.
近年、そのようなアンドラッガブルな分子に対する新しい創薬戦略として、標的タンパク質分解誘導剤を用いて、標的タンパク質をユビキチン化して分解するユビキチン創薬技術が注目されている(非特許文献3)。この技術は、全ての細胞に存在する図11に示すようなタンパク質分解機構のユビキチン-プロテアソーム系(Ub-PSM系)を利用して標的タンパク質の分解を誘導する。標的タンパク質分解誘導剤は、標的タンパク質とE3リガーゼのそれぞれに結合するバインダー部位と、それらを連結するリンカー部位からなる化合物であり、標的タンパク質とE3リガーゼに結合して、両タンパク質‐タンパク質相互作用を促進する。それによりユビキチン化が促進された標的タンパク質はプロテアソームに誘導された後、分解される。市販のPROTAC(登録商標)は、代表的な標的タンパク質分解誘導剤の一つである。 In recent years, as a new drug discovery strategy for such undruggable molecules, ubiquitin drug discovery technology that uses target protein degradation inducers to ubiquitinate and degrade target proteins has attracted attention (Non-Patent Document 3). This technique utilizes the ubiquitin-proteasome system (Ub-PSM system) of the protein degradation mechanism present in all cells as shown in FIG. 11 to induce degradation of target proteins. A target protein degradation inducer is a compound consisting of a binder site that binds to a target protein and an E3 ligase, respectively, and a linker site that connects them. Facilitate. The target protein whose ubiquitination is thereby promoted is degraded after being induced to the proteasome. Commercially available PROTAC (registered trademark) is one of representative target protein degradation inducers.
ところが、上記ユビキチン創薬技術にも大きな課題がある。前述のように標的タンパク質分解誘導剤は、E3リガーゼによる標的タンパク質の直接的なユビキチン化を促進する。しかし、転写因子のような標的タンパク質は、E3リガーゼによるユビキチン化自体が困難である。そのため、そのような標的タンパク質は、依然としてアンドラッガブルな標的分子のままであった。 However, the above ubiquitin drug discovery technology also faces major challenges. As described above, target protein degradation inducers promote direct ubiquitination of target proteins by E3 ligases. However, target proteins such as transcription factors are difficult to ubiquitinate by E3 ligases. Therefore, such target proteins remained undruggable target molecules.
本発明の課題は、転写因子のようなユビキチン化されにくい標的タンパク質であってもUb-PSM系に誘導し、分解できる新たなユビキチン創薬技術を開発し、提供することである。 The task of the present invention is to develop and provide a new ubiquitin drug discovery technology that can induce the Ub-PSM system to degrade even target proteins that are difficult to ubiquitinate, such as transcription factors.
本発明の課題は、上記新たなユビキチン創薬技術に基づき、任意の標的タンパク質の分解を誘導できる標的ペプチド分解誘導剤を開発し、提供することである。 The object of the present invention is to develop and provide a target peptide degradation inducer capable of inducing the degradation of any target protein based on the above new ubiquitin drug discovery technology.
本発明の課題は、疾患原因となり、かつ従来技術では創薬困難であった標的タンパク質であっても分解誘導でき、その疾患の治療、改善、予防等に機能し得る疾患抑制剤を提供することである。 An object of the present invention is to provide a disease-suppressing agent capable of treating, improving, preventing, etc., a disease by inducing the degradation of even a target protein that causes a disease and has been difficult to develop a drug with the prior art. is.
上記課題を解決するために、本発明者らは鋭意研究を重ねた結果、間接的ユビキチン化法による全く新しい標的タンパク質分解技術の開発に成功した。この方法では、従来のユビキチン創薬技術と同様にUb-PSM系を利用して標的タンパク質の分解を行う。しかし、ユビキチンリガーゼE3を介した標的タンパク質への直接的なユビキチン化プロセスを必要とせず、代わりに図12で示すように予めユビキチンが連結された結合因子からなる標的ペプチド分解誘導剤を用いる。この標的ペプチド分解誘導剤の結合因子が標的タンパク質に結合することで、標的タンパク質と標的ペプチド分解誘導剤による複合体が形成される。標的タンパク質は、この複合体において間接的にユビキチン化された状態となる結果、あたかも標的タンパク質が直接的にユビキチン化されたかのようにプロテアソームによって認識された後、分解される。 In order to solve the above problems, the present inventors have conducted intensive research and succeeded in developing a completely new target protein degradation technology using an indirect ubiquitination method. In this method, the Ub-PSM system is used to degrade the target protein as in the conventional ubiquitin drug discovery technology. However, it does not require a direct ubiquitination process to the target protein via ubiquitin ligase E3, and instead uses a target peptide degradation inducer consisting of a binding factor pre-linked with ubiquitin as shown in FIG. A complex of the target protein and the target peptide degradation inducer is formed by binding the binding factor of the target peptide degradation inducer to the target protein. The target protein becomes indirectly ubiquitinated in this complex, resulting in recognition by the proteasome as if the target protein had been directly ubiquitinated, followed by degradation.
この間接的ユビキチン化法であれば、標的タンパク質に結合する分子を結合因子として用いることで、直接的なユビキチン化が困難な標的タンパク質であっても、Ub-PSM系を利用した創薬ターゲットとすることができる。 With this indirect ubiquitination method, by using a molecule that binds to the target protein as a binding factor, even if the target protein is difficult to ubiquitinate directly, it can be used as a drug discovery target using the Ub-PSM system. can do.
本発明は、上記新たに開発された間接的ユビキチン化法による標的ペプチドの分解技術に基づくものであって、以下を提供する。 The present invention is based on the newly developed technology for degrading target peptides by the indirect ubiquitination method, and provides the following.
(1)標的ペプチドに結合する結合因子を一又は連結した二以上のユビキチンのC末端に連結した標的ペプチド分解誘導剤。
(2)前記ユビキチンが以下の(a)~(c)に記載のいずれか一のアミノ酸配列からなる、(1)に記載の標的ペプチド分解誘導剤。
(a)配列番号1で示すアミノ酸配列、
(b)配列番号1で示すアミノ酸配列において1又は複数個のアミノ酸が付加、欠失又は置換されたアミノ酸配列、又は
(c)配列番号1で示すアミノ酸配列と90%以上のアミノ酸同一性を有するアミノ酸配列
(3)前記ユビキチンが配列番号1で示すアミノ酸配列において、以下の(I)~(III)に記載のいずれか一のアミノ酸置換を含むアミノ酸配列からなる、(2)に記載の標的ペプチド分解誘導剤。
(I)72位及び74位のアルギニン残基がそれぞれアラニン残基及びスレオニン残基に置換されたアミノ酸配列、
(II)72位及び74位のアルギニン残基がそれぞれプロリン残基及びスレオニン残基に置換されたアミノ酸配列、又は
(III)73位のロイシン残基がプロリン残基に置換されたアミノ酸配列
(4)前記結合因子が核酸である、(1)~(3)のいずれかに記載の標的ペプチド分解誘導剤。
(5)前記核酸が核酸アプタマー又は核酸結合ドメイン認識核酸である、(4)に記載の標的ペプチド分解誘導剤。
(6)前記結合因子がペプチドである、(1)~(3)に記載の標的ペプチド分解誘導剤。
(7)前記ペプチドがリガンド、レセプター、そのリガンド結合部位、抗体、及びその活性断片からなる群から選択される、(6)に記載の標的ペプチド分解誘導剤。
(8)前記結合因子が低分子化合物である、(1)~(3)に記載の標的ペプチド分解誘導剤。
(9)前記ユビキチンと結合因子が、ユビキチンのC末端に置換又は付加によって導入されたチオール基を介した結合、アジド-アルキン環化付加反応による結合、又はオキシム/ヒドラゾン結合により連結されている、(1)~(8)のいずれかに記載の標的ペプチド分解誘導剤。
(10)前記チオール基を介した結合がマイケル付加反応、チオールエン反応又はチオールイン反応による結合である、(9)に記載の標的ペプチド分解誘導剤。
(11)前記結合因子と前記ユビキチンがリンカー因子を介して連結されている、(1)~(10)のいずれかに記載の標的ペプチド分解誘導剤。
(12)前記標的ペプチドが転写因子、アミロイド、タウタンパク質、α-シヌクレイン、カスパーゼ、変性ハンチンチン、偽キナーゼ、K-RAS変異体、Atg因子、mTOR複合体1、GAPR-1及びオーファン受容体からなる群から選択される、(1)~(11)のいずれかに記載の標的ペプチド分解誘導剤。
(13)前記転写因子がNF-κBである、(12)に記載の標的ペプチド分解誘導剤。
(14)(6)又は(7)に記載の標的ペプチド分解誘導剤をコードするポリヌクレオチド。
(15)(14)に記載のポリヌクレオチドを含む発現ベクター。
(16)(14)に記載のポリヌクレオチド又は(15)に記載の発現ベクターからなる核酸型標的ペプチド分解誘導剤。
(17)(1)~(13)のいずれかに記載の標的ペプチド分解誘導剤、及び/又は(16)に記載の核酸型標的ペプチド分解誘導剤からなる群から選択されるいずれか2種以上を包含する標的ペプチド分解誘導組成物。
(18)前記標的ペプチドが変性ハンチンチンである、(12)に記載の標的ペプチド分解誘導剤を含むハンチントン病抑制剤。
(19)前記標的ペプチドがAtg因子である、(12)に記載の標的ペプチド分解誘導剤を含むオートファジー抑制剤。
(20)前記標的ペプチドがmTOR複合体1又はGAPR-1である、(12)に記載の標的ペプチド分解誘導剤を含むオートファジー促進剤。
(21)(13)に記載の標的ペプチド分解誘導剤を含む抗癌剤。
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2021-172029号の開示内容を包含する。
(1) A target peptide degradation inducer linked to the C-terminus of one or more ubiquitins linked to a binding factor that binds to a target peptide.
(2) The target peptide degradation inducer according to (1), wherein the ubiquitin consists of any one of the amino acid sequences described in (a) to (c) below.
(a) the amino acid sequence shown in SEQ ID NO: 1,
(b) an amino acid sequence in which one or more amino acids are added, deleted or substituted in the amino acid sequence shown in SEQ ID NO: 1, or (c) having 90% or more amino acid identity with the amino acid sequence shown in SEQ ID NO: 1 Amino acid sequence (3) The target peptide according to (2), wherein the ubiquitin comprises an amino acid sequence containing any one of the amino acid substitutions described in (I) to (III) below in the amino acid sequence shown in SEQ ID NO: 1. Decomposition inducer.
(I) an amino acid sequence in which arginine residues at positions 72 and 74 are substituted with alanine residues and threonine residues, respectively;
(II) an amino acid sequence in which the arginine residues at positions 72 and 74 are substituted with proline and threonine residues, respectively, or (III) an amino acid sequence in which the leucine residue at position 73 is substituted with a proline residue (4 ) The target peptide degradation inducer according to any one of (1) to (3), wherein the binding factor is a nucleic acid.
(5) The target peptide degradation inducer according to (4), wherein the nucleic acid is a nucleic acid aptamer or nucleic acid-binding domain-recognizing nucleic acid.
(6) The target peptide degradation inducer according to (1) to (3), wherein the binding factor is a peptide.
(7) The target peptide degradation inducer according to (6), wherein the peptide is selected from the group consisting of ligands, receptors, ligand-binding sites thereof, antibodies, and active fragments thereof.
(8) The target peptide degradation inducer according to (1) to (3), wherein the binding factor is a low-molecular-weight compound.
(9) the ubiquitin and the binding agent are linked via a thiol group introduced by substitution or addition to the C-terminus of ubiquitin, via an azide-alkyne cycloaddition reaction, or via an oxime/hydrazone bond; The target peptide degradation inducer according to any one of (1) to (8).
(10) The target peptide degradation inducer according to (9), wherein the bond via the thiol group is a bond by Michael addition reaction, thiolene reaction or thiolyne reaction.
(11) The target peptide degradation inducer according to any one of (1) to (10), wherein the binding factor and the ubiquitin are linked via a linker factor.
(12) the target peptide is a transcription factor, amyloid, tau protein, α-synuclein, caspase, modified huntingtin, pseudokinase, K-RAS mutant, Atg factor,
(13) The target peptide degradation inducer according to (12), wherein the transcription factor is NF-κB.
(14) A polynucleotide encoding the target peptide degradation inducer of (6) or (7).
(15) An expression vector comprising the polynucleotide of (14).
(16) A nucleic acid type target peptide degradation inducer comprising the polynucleotide of (14) or the expression vector of (15).
(17) Any two or more selected from the group consisting of the target peptide degradation inducer according to any one of (1) to (13) and/or the nucleic acid-type target peptide degradation inducer according to (16) A target peptide degradation-inducing composition comprising:
(18) A Huntington's disease inhibitor containing the target peptide degradation inducer according to (12), wherein the target peptide is denatured huntingtin.
(19) An autophagy inhibitor containing the target peptide degradation inducer according to (12), wherein the target peptide is an Atg factor.
(20) An autophagy promoter comprising the target peptide degradation inducer according to (12), wherein the target peptide is
(21) An anticancer agent comprising the target peptide degradation inducer of (13).
This specification includes the disclosure content of Japanese Patent Application No. 2021-172029, which is the basis of priority of this application.
本発明の標的ペプチド分解誘導剤によれば、直接的なユビキチン化も困難であった、従来技術ではアンドラッガブルな任意の標的タンパク質を、Ub-PSM系に誘導し、分解することができる。 According to the target peptide degradation inducer of the present invention, it is possible to induce the Ub-PSM system to degrade any undruggable target protein that was difficult to ubiquitinate directly in the prior art.
本発明の疾患抑制剤によれば、任意の疾患原因タンパク質をUb-PSM系を介して分解することにより、その疾患の治療、改善、予防等が可能となる。 According to the disease inhibitor of the present invention, any disease-causing protein can be degraded via the Ub-PSM system to treat, ameliorate, prevent, etc. the disease.
1.標的ペプチド分解誘導剤
1-1.概要
本発明の第1の態様は、標的ペプチド分解誘導剤である。本発明の標的ペプチド分解誘導剤は、ユビキチンのC末端に標的ペプチドに結合する結合因子を連結した構成を有する。本発明の標的ペプチド分解誘導剤によれば、生体内に投与することで標的ペプチドの直接的なユビキチン化を必要とせずに、標的ペプチドをプロテアソームへ誘導することができる。
1. Target Peptide Degradation Inducing Agent 1-1. Overview A first aspect of the present invention is a target peptide degradation inducer. The target peptide degradation inducer of the present invention has a structure in which a binding factor that binds to a target peptide is linked to the C-terminus of ubiquitin. According to the target peptide degradation inducer of the present invention, the target peptide can be guided to the proteasome by administration in vivo without requiring direct ubiquitination of the target peptide.
1-2.定義
本明細書で頻用する以下の用語について定義する。
本明細書において「ペプチド」とは、複数のアミノ酸がペプチド結合によって連結されたアミノ酸重合体をいう。連結されるアミノ酸の数は問わない。したがって、本明細書で単にペプチドと表記する場合、オリゴペプチド及びポリペプチド、タンパク質を包含する包括的用語を意味する。
1-2. Definitions The following terms frequently used in this specification are defined.
As used herein, the term "peptide" refers to an amino acid polymer in which multiple amino acids are linked by peptide bonds. Any number of amino acids can be linked. Therefore, when the term "peptide" is used herein, it means a generic term including oligopeptides, polypeptides and proteins.
本明細書において、「核酸」とは、原則として、ヌクレオチドを構成単位とし、それらがホスホジエステル結合によって連結した生体高分子をいう。通常は、アデニン(A)、グアニン(G)、シトシン(C)及びチミン(T)のいずれかの塩基を有するデオキシリボヌクレオチドが連結したDNA、及び/又はアデニン、グアニン、シトシン及びウラシル(U)のいずれかの塩基を有するリボヌクレオチドが連結したRNA等の自然界に存在する天然型ヌクレオチドが連結してなる天然型核酸が該当する。 As used herein, the term "nucleic acid" basically refers to a biopolymer composed of nucleotides linked by phosphodiester bonds. Usually, DNA linked with deoxyribonucleotides having any of adenine (A), guanine (G), cytosine (C) and thymine (T) bases, and/or adenine, guanine, cytosine and uracil (U) A natural nucleic acid formed by connecting natural nucleotides existing in nature such as RNA to which ribonucleotides having any base are linked corresponds.
「ユビキチン」(本明細書では、しばしば「Ub」と表記する)とは、76個のアミノ酸で構成される、真核生物において高度に保存されたタンパク質である。あらゆる細胞で発現し、標的タンパク質のユビキチン化を介し、タンパク質分解、シグナル伝達、DNA修復、選択的オートファジー等の様々な生命現象において機能している。ユビキチン-プロテアソーム系によるタンパク質分解はユビキチンの主要な機能であり、本明細書においても、限定はしないが、特に断りのない限りタンパク質分解に関する機能について述べる。 "Ubiquitin" (herein often abbreviated as "Ub") is a highly conserved protein in eukaryotes, consisting of 76 amino acids. It is expressed in all cells and functions in various biological phenomena such as protein degradation, signal transduction, DNA repair, and selective autophagy through ubiquitination of target proteins. Proteolytic degradation by the ubiquitin-proteasome system is the primary function of ubiquitin, and in this specification also, without limitation, functions relating to proteolytic degradation are mentioned unless otherwise stated.
「ユビキチン化」とは、「ユビキチン修飾」とも称し、標的タンパク質へのユビキチンの付加によるタンパク質翻訳後修飾をいう。複数のユビキチンが鎖状に連結付加された状態は、特に「ポリユビキチン化」という。ポリユビキチン化された標的タンパク質において、付加されたポリユビキチン鎖はユビキチン-プロテアソーム系において分解シグナルとして機能する。 "Ubiquitination" is also called "ubiquitin modification", and refers to protein post-translational modification by addition of ubiquitin to a target protein. A state in which multiple ubiquitins are linked and added in a chain is particularly referred to as “polyubiquitination”. In polyubiquitinated target proteins, the attached polyubiquitin chains function as degradation signals in the ubiquitin-proteasome system.
「標的タンパク質」とは、ユビキチン-プロテアソーム系においてユビキチン化され、分解対象となるタンパク質をいう。通常は生体内において不要となった除去すべきタンパク質が該当する。 "Target protein" refers to a protein that is ubiquitinated in the ubiquitin-proteasome system and targeted for degradation. Usually, proteins that are no longer needed in vivo and should be removed correspond.
「ユビキチン-プロテアソーム系(Ubiquitin-Proteasome system)」(本明細書では、しばしば「Ub-PSM系」と表記する)とは、生体内において様々な生命現象を制御するATP依存性タンパク質分解系の一つである。Ub-PSM系は、3種の酵素によるユビキチンの活性化、及び標的タンパク質へのユビキチン修飾、並びにユビキチン化された標的タンパク質のプロテアソームによる分解までを担う一連の反応が行われる。具体的には、まずユビキチン活性化酵素(E1)がATPを利用して自身のシステイン(Cys)残基でユビキチンとチオエステル結合し、E1-Ub複合体が形成される。次に、E1-Ub中のユビキチンがユビキチン結合酵素E2のCys残基にチオエステル結合を介して受け渡され、E2-Ub複合体が形成されると共にユビキチンが活性化される。続いて、E2-Ub複合体中の活性化ユビキチンがヘテロ複合体であるユビキチンリガーゼE3の活性により標的タンパク質に受け渡され、標的タンパク質がユビキチン修飾される。このときユビキチンのC末端と標的タンパク質のリジン(Lys)残基間がイソペプチド結合により結合される。その後、新たなユビキチンのC末端と標的タンパク質に結合したユビキチン中の、主として48番目のLys残基間でE3を介した縮合反応が繰り返されることで、複数のユビキチンが連結したポリユビキチン鎖が形成される。このポリユビキチン鎖が標識となり、ポリユビキチン化タンパク質はプロテアソームによって認識され、ATP依存的に分解される。修飾に用いられたユビキチンは、標的タンパク質から切断された後、再利用される。 “Ubiquitin-Proteasome system” (herein often referred to as “Ub-PSM system”) is one of the ATP-dependent proteolytic systems that regulate various biological phenomena in vivo. is one. In the Ub-PSM system, a series of reactions takes place, including activation of ubiquitin by three enzymes, ubiquitin modification to target proteins, and degradation of ubiquitinated target proteins by the proteasome. Specifically, ubiquitin-activating enzyme (E1) uses ATP to thioester-bond with ubiquitin at its own cysteine (Cys) residue to form an E1-Ub complex. Next, ubiquitin in E1-Ub is transferred to the Cys residue of ubiquitin conjugating enzyme E2 via a thioester bond, forming an E2-Ub complex and ubiquitin is activated. Subsequently, the activated ubiquitin in the E2-Ub complex is transferred to the target protein by the activity of the heterocomplex ubiquitin ligase E3, and the target protein is ubiquitin-modified. At this time, an isopeptide bond is formed between the C-terminus of ubiquitin and the lysine (Lys) residue of the target protein. After that, E3-mediated condensation is repeated between the C-terminus of the new ubiquitin and the ubiquitin bound to the target protein, mainly at the 48th Lys residue, to form a polyubiquitin chain in which multiple ubiquitins are linked. be done. This polyubiquitin chain serves as a label, and the polyubiquitinated protein is recognized by the proteasome and degraded in an ATP-dependent manner. Ubiquitin used for modification is reused after being cleaved from the target protein.
本明細書において「標的ペプチド」(本明細書では、しばしば「POI」(peptide of interest)と表記する)とは、本発明の標的ペプチド分解誘導剤によってプロテアソームへと誘導される分解すべきペプチドをいう。前記標的タンパク質と実質的に同義である。標的ペプチドの種類は問わない。例えば、通常のUb-PSM系において、直接的なユビキチン化が困難な標的タンパク質等であってもよい。標的ペプチドの例として、限定はしないが、転写因子、酵素、リガンド、受容体、構成タンパク質、ペプチドホルモン、サイトカイン、抗体、及び抗原タンパク質等が挙げられる。標的ペプチドのより具体的な例として、アミロイド、タウタンパク質、α-シヌクレイン、カスパーゼ、変性ハンチンチン、偽キナーゼ、K-RAS変異体、Atg因子、mTOR複合体1、GAPR-1、及びオーファン受容体等が挙げられる。
As used herein, the term "target peptide" (herein often referred to as "POI" (peptide of interest)) refers to a peptide to be degraded that is induced to the proteasome by the target peptide degradation-inducing agent of the present invention. say. It is substantially synonymous with the target protein. Any type of target peptide can be used. For example, it may be a target protein that is difficult to ubiquitinate directly in a normal Ub-PSM system. Examples of target peptides include, but are not limited to, transcription factors, enzymes, ligands, receptors, constituent proteins, peptide hormones, cytokines, antibodies, antigenic proteins, and the like. More specific examples of target peptides include amyloid, tau protein, α-synuclein, caspase, modified huntingtin, pseudokinase, K-RAS mutant, Atg factor,
本明細書において「結合因子」とは、標的ペプチドと結合し得る物質をいう。結合因子の詳細な構成については後述する。
本明細書において「生体」とは、生きた細胞、組織、器官、又は個体をいう。
As used herein, the term "binding agent" refers to a substance capable of binding to a target peptide. A detailed configuration of the binding factor will be described later.
As used herein, the term "living organism" refers to living cells, tissues, organs, or individuals.
1-3.構成
1-3-1.構成因子
本発明の標的ペプチド分解誘導剤は、結合因子及びユビキチンを必須の構成因子として、またリンカー因子を選択的な構成因子として含む。以下、各構成因子について具体的に説明をする。
1-3. Configuration 1-3-1. Constituent Factors The target peptide degradation-inducing agent of the present invention contains a binding factor and ubiquitin as essential constituent factors, and a linker factor as an optional constituent factor. Each component will be specifically described below.
(1)結合因子
「結合因子」とは、標的ペプチドと結合し得る分子をいう。標的ペプチドと特異的に結合する分子が好ましい。
結合因子は、標的ペプチドと結合する性質を有する限り、その種類は問わない。例えば、核酸、ペプチド、又は低分子化合物が挙げられる。以下、それぞれについて、説明をする。
(1) Binding Agent A “binding agent” refers to a molecule that can bind to a target peptide. Molecules that specifically bind the target peptide are preferred.
The binding factor may be of any type as long as it has the property of binding to the target peptide. Examples include nucleic acids, peptides, or low-molecular-weight compounds. Each of these will be explained below.
(1-1)核酸
結合因子は核酸で構成されていてもよい。核酸で構成される結合因子を本明細書では「核酸型結合因子」と表記する。
(1-1) Nucleic acid The binding factor may be composed of nucleic acid. Binding agents composed of nucleic acids are referred to herein as “nucleic acid-type binding agents”.
前述のように、核酸は、原則として天然型核酸が該当するが、本明細書においては、非天然型核酸も含むことができる。したがって、核酸型結合因子は、天然型核酸及び/又は非天然型核酸を含み得る。 As mentioned above, in principle, nucleic acids correspond to natural nucleic acids, but in the present specification, non-natural nucleic acids can also be included. Accordingly, a nucleic acid-type binding agent can include naturally occurring nucleic acids and/or non-naturally occurring nucleic acids.
本明細書において「非天然型核酸」とは、非天然型ヌクレオチドを構成単位として含む生体高分子、及び核酸類似体等をいう。本明細書において「非天然型ヌクレオチド」とは、天然ヌクレオチド以外の任意のヌクレオチドをいう。例えば、人工的に構築された又は人工的に化学修飾された、天然型ヌクレオチドに類似の性質及び/又は構造を有する自然界には存在しないヌクレオチドが該当する。具体的には、例えば、2'-O-メチルリボース、2'-O(CH2)2OCH3リボース、2'-F(2'-フルオロ)リボース、3'-O-メチルリボース等を含むヌクレオチドが挙げられる。本明細書において「核酸類似体」とは、天然型核酸に類似の構造及び/又は性質を有する人工的に構築された化合物をいう。具体的には、例えば、架橋化核酸(BNA/LNA:Bridged Nucleic Acid/Locked Nucleic Acid)、モルホリノ核酸、ペプチド核酸(PNA:Peptide Nucleic Acid)等が挙げられる他、メチルホスホネート型DNA又はRNA、ホスホロチオエート型DNA又はRNA、ホスホルアミデート型DNA又はRNAを含むこともできる。
核酸型結合因子の具体的な例として、核酸アプタマーや核酸結合ドメイン認識核酸が挙げられる。
As used herein, the term “non-natural nucleic acid” refers to biopolymers, nucleic acid analogues, etc. containing non-natural nucleotides as structural units. As used herein, "non-natural nucleotide" refers to any nucleotide other than natural nucleotides. For example, non-naturally occurring nucleotides that are artificially constructed or artificially chemically modified and that have similar properties and/or structures to natural nucleotides are applicable. Specific examples include 2'-O- methyl ribose, 2'-O(CH2)2OCH3 ribose , 2'-F(2'-fluoro) ribose, 3'-O-methyl ribose and the like. Nucleotides. As used herein, the term "nucleic acid analogue" refers to an artificially constructed compound having a structure and/or properties similar to those of naturally occurring nucleic acids. Specifically, for example, crosslinked nucleic acid (BNA / LNA: Bridged Nucleic Acid / Locked Nucleic Acid), morpholino nucleic acid, peptide nucleic acid (PNA: Peptide Nucleic Acid), etc., methylphosphonate type DNA or RNA, phosphorothioate It can also contain type DNA or RNA, phosphoramidate type DNA or RNA.
Specific examples of nucleic acid-type binding factors include nucleic acid aptamers and nucleic acids that recognize nucleic acid-binding domains.
(核酸アプタマー)
「核酸アプタマー」とは、核酸で構成されたアプタマーであって、水素結合等を介した一本鎖核酸分子の二次構造、及び三次構造に基づいて形成される立体構造に基づいて標的物質と強固、かつ特異的に結合する能力を持つリガンド分子をいう。したがって、本明細書においては、標的ペプチドと強固、かつ特異的に結合する核酸アプタマーが該当する。
(nucleic acid aptamer)
"Nucleic acid aptamer" is an aptamer composed of nucleic acid, and the secondary structure and tertiary structure of a single-stranded nucleic acid molecule through hydrogen bonding, etc. A ligand molecule that has the ability to bind tightly and specifically. Therefore, in the present specification, a nucleic acid aptamer that strongly and specifically binds to a target peptide is applicable.
核酸アプタマーは、一般に、RNAアプタマーとDNAアプタマーが知られているが、本明細書における核酸アプタマーを構成する核酸は特に限定はしない。DNAアプタマー、RNAアプタマー、DNAとRNAの組み合わせで構成されるアプタマーのいずれも含み得る。 Nucleic acid aptamers are generally known as RNA aptamers and DNA aptamers, but the nucleic acids that constitute the nucleic acid aptamers in the present specification are not particularly limited. Any of DNA aptamers, RNA aptamers, and aptamers composed of a combination of DNA and RNA can be included.
本明細書で使用する核酸アプタマーは、標的ペプチドを結合対象として、当該分野で公知の方法により作製することができる。例えば、SELEX(systematic evolution of ligands by exponential enrichment)法を用いた試験管内選別法が挙げられる。SELEX法は、公知の方法であり、具体的な方法は、例えば、Panら(Proc. Natl. Acad. Sci. 1995, U.S.A.92: 11509-11513)に準じて行えばよい。 The nucleic acid aptamer used herein can be produced by a method known in the art with a target peptide as a binding target. Examples thereof include an in vitro selection method using the SELEX (systematic evolution of ligands by exponential enrichment) method. The SELEX method is a known method, and a specific method may be performed, for example, according to Pan et al. (Proc. Natl. Acad. Sci. 1995, U.S.A.92: 11509-11513).
(核酸結合ドメイン認識核酸)
本明細書において「核酸結合ドメイン(Nucleic acid-Binding Domain)」(本明細書では、しばしば「NBD」と略称する)とは、核酸結合性タンパク質中に包含され、核酸結合ドメイン認識配列(本明細書では、しばしば「NBD認識配列」と表記する)を認識し、それに結合するドメインをいう。NBDはDNA結合ドメイン(DNB)及びRNA結合ドメイン(RNB)のいずれであってもよい。NBDを構成する具体的な構造モチーフの例として、限定はしないが、ジンクフィンガー、ロイシンジッパー、βヘアピン、ヘリックスターンヘリックス、ヘリックスループヘリックス、ウィングヘリックス、HMGボックス、RRMドメイン、及びTALエフェクター等が挙げられる。
(Nucleic acid binding domain recognition nucleic acid)
As used herein, a "Nucleic acid-Binding Domain" (herein often abbreviated as "NBD") is included in a nucleic acid-binding protein and includes a nucleic acid-binding domain recognition sequence (herein In literature, it is often referred to as an "NBD recognition sequence") and refers to a domain that recognizes and binds to it. An NBD can be either a DNA binding domain (DNB) or an RNA binding domain (RNB). Examples of specific structural motifs that constitute NBDs include, but are not limited to, zinc fingers, leucine zippers, β-hairpins, helix-turn helices, helix-loop helices, wing-helices, HMG boxes, RRM domains, and TAL effectors. be done.
本明細書において「核酸結合ドメイン認識核酸」(本明細書では、しばしば「NBD認識核酸」と略称する)とは、NBD認識配列を含む核酸をいう。NBD認識核酸は、包含するNBD認識配列を介して標的ペプチドのNBDに結合できる。 As used herein, the term "nucleic acid binding domain recognition nucleic acid" (herein often abbreviated as "NBD recognition nucleic acid") refers to a nucleic acid containing an NBD recognition sequence. An NBD-recognizing nucleic acid can bind to the NBD of a target peptide via the included NBD-recognition sequence.
NBD認識核酸は、いずれの核酸で構成されていてもよい。例えば、DNA、RNA、又はそれらの組合せからなる天然型核酸の他、全部又は一部を非天然型核酸に置き換えた非天然型核酸が挙げられる。さらにNBD認識核酸は一本鎖核酸、二本鎖核酸、又はそれらの組合せで構成されていてもよく、ヘアピン構造のような二次構造、又は三次構造を有していてもよい。 The NBD-recognizing nucleic acid may consist of any nucleic acid. Examples include naturally occurring nucleic acids consisting of DNA, RNA, or a combination thereof, as well as non-natural nucleic acids in which all or part of them are replaced with non-natural nucleic acids. Furthermore, the NBD-recognizing nucleic acid may consist of a single-stranded nucleic acid, a double-stranded nucleic acid, or a combination thereof, and may have a secondary structure such as a hairpin structure, or a tertiary structure.
少なくとも包含するNBD認識配列が、標的ペプチドのNBDが認識し、結合する核酸及び配列で構成されていればよく、具体的な構成に関しては特に限定しない。例えば、標的ペプチドのNBDが特定の塩基配列を認識する二本鎖DNA結合ドメイン(DNB)である場合、NBD認識核酸は、そのDNBの認識配列を標的配列として含む二本鎖DNAで構成されていればよい。 At least the included NBD recognition sequence should consist of a nucleic acid and a sequence that the NBD of the target peptide recognizes and binds to, and there are no particular restrictions on the specific configuration. For example, if the NBD of the target peptide is a double-stranded DNA binding domain (DNB) that recognizes a specific base sequence, the NBD-recognizing nucleic acid is composed of double-stranded DNA containing the DNB recognition sequence as the target sequence. All you have to do is
NBD認識核酸の具体例として、プロモーター中に含まれる転写因子結合配列等が挙げられる。より具体的な例として、配列番号2又は3で示され、転写因子NF-κBが結合する塩基配列を含むNF-κB結合型ヘアピンDNAが挙げられる。 Specific examples of NBD-recognizing nucleic acids include transcription factor binding sequences contained in promoters. A more specific example is an NF-κB-binding hairpin DNA represented by SEQ ID NO: 2 or 3 and containing a nucleotide sequence to which the transcription factor NF-κB binds.
(1-2)ペプチド
結合因子はペプチドで構成されていてもよい。ペプチドで構成される結合因子を本明細書では「ペプチド型結合因子」と表記する。
(1-2) Peptide A binding agent may be composed of a peptide. Binding agents composed of peptides are referred to herein as “peptide-type binding agents”.
ペプチド型結合因子は、ペプチド間(タンパク質間)相互作用によって標的ペプチドと結合する。この結合は、安定的かつ特異的結合であることが好ましい。前記ペプチド間相互作用は、限定はしないが、解離可能な一過性相互作用であればよい。このようなペプチド間相互作用は、例えば、ペプチドを構成するアミノ酸間の疎水的結合、ファンデルワールス力、又は塩橋により達成される。 Peptide-type binding agents bind to target peptides through peptide-peptide (protein-protein) interactions. This binding is preferably a stable and specific binding. The inter-peptide interaction is not limited, but may be a dissociable transient interaction. Such peptide-peptide interactions are achieved, for example, by hydrophobic bonds, van der Waals forces, or salt bridges between the amino acids that make up the peptide.
ペプチド型結合因子は、ペプチドであれば、その種類は限定しない。例えば、標的ペプチドがレセプターであれば、ペプチド型結合因子は、そのリガンド又はそのレセプター結合部位で構成されていればよい。逆に標的ペプチドがリガンドであれば、ペプチド型結合因子は、そのレセプター又はリガンド結合部位で構成されていればよい。また標的ペプチドが抗原であれば、ペプチド型結合因子は、それを特異的に認識する抗体又はその活性断片で構成されていればよい。あるいは、PROTAC(登録商標)(Proteolysis Targeting Chimeric Molecules)のような市販の標的タンパク質分解誘導分子で構成されていてもよい。 The type of the peptide-type binding factor is not limited as long as it is a peptide. For example, if the target peptide is a receptor, the peptide-type binding agent may consist of its ligand or its receptor binding site. Conversely, if the target peptide is a ligand, the peptide-type binding agent may consist of its receptor or ligand binding site. Also, if the target peptide is an antigen, the peptide-type binding agent may consist of an antibody that specifically recognizes it or an active fragment thereof. Alternatively, it may be composed of commercially available target protein degradation-inducing molecules such as PROTAC (Registered Trademark) (Proteolysis Targeting Chimeric Molecules).
ペプチド型結合因子の、より具体的な例として配列番号4で示すアミノ酸配列(WYPWMKKHHRRRRRRRR)からなり、癌関連転写因子のPBXに結合するHXR9(Morgan R. et al., 2007, Cancer Res. 67, 5806-5813)が挙げられる。 As a more specific example of the peptide-type binding factor, HXR9 (Morgan R. et al., 2007, Cancer Res. 67, 5806-5813).
(1-3)低分子化合物
結合因子は低分子化合物で構成されていてもよい。低分子化合物で構成される結合因子を本明細書では「低分子化合物型結合因子」と表記する。
(1-3) Low-molecular-weight compound The binding agent may be composed of a low-molecular-weight compound. A binding agent composed of a low-molecular-weight compound is referred to herein as a “low-molecular-weight compound-type binding agent”.
「低分子化合物」とは、一般に、分子量約数百~数千の天然化合物又は化学合成物をいう。ただし、本明細書における低分子化合物型結合因子は、標的ペプチドの立体構造等に基づいて、標的ペプチドと、好ましくは特異的に相互作用し、結合し得る低分子化合物であれば、分子量、構造、及び種類は特に限定しない。本発明の標的ペプチド分解誘導剤を構成する低分子化合物型結合因子の具体例として、ジヒドロ葉酸還元酵素(DHFR)に対する阻害剤であるメトトレキサート(MTX)、又はアビジンに対する特異的認識能を有するビオチン等が挙げられる。 "Low-molecular-weight compounds" generally refer to natural compounds or chemically synthesized compounds with a molecular weight of about several hundred to several thousand. However, the low-molecular-weight compound-type binding agent herein is a low-molecular-weight compound capable of specifically interacting and binding to the target peptide based on the three-dimensional structure of the target peptide. , and types are not particularly limited. Specific examples of the low-molecular-weight compound-type binding factor constituting the target peptide degradation inducer of the present invention include methotrexate (MTX), which is an inhibitor against dihydrofolate reductase (DHFR), biotin, etc., which has the ability to specifically recognize avidin. is mentioned.
(2)ユビキチン
「ユビキチン」は、前述のように真核生物種間内で高度に保存された26アミノ酸からなるタンパク質である。したがって、本発明の標的ペプチド分解誘導剤を構成するユビキチンの由来する生物種も限定はしない。いずれの真核生物が有するユビキチンであってもよい。ユビキチンの具体例として、配列番号1で示すアミノ酸配列(MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGG)からなるヒト由来のユビキチン、配列番号1で示すアミノ酸配列において1又は複数個若しくは数個のアミノ酸が付加、欠失、又は置換されたアミノ酸配列からなる変異型ユビキチン若しくはユビキチンオルソログ、又は配列番号1で示すアミノ酸配列と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%以上のアミノ酸同一性を有するアミノ酸配列からなる変異型ユビキチン若しくはユビキチンオルソログが挙げられる。
(2) Ubiquitin “Ubiquitin” is a protein consisting of 26 amino acids highly conserved among eukaryotic species, as described above. Therefore, the biological species from which the ubiquitin constituting the target peptide degradation inducer of the present invention is derived is also not limited. It may be ubiquitin that any eukaryote has. A specific example of ubiquitin is a human-derived ubiquitin consisting of the amino acid sequence shown in SEQ ID NO: 1 (MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRLRGG), in which one or more or several amino acids are added, deleted, or substituted in the amino acid sequence shown in SEQ ID NO: 1. 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more of the amino acid sequence shown in SEQ ID NO: 1, or a mutant ubiquitin or ubiquitin ortholog consisting of an amino acid sequence Mutant ubiquitin or ubiquitin orthologues consisting of amino acid sequences having 98% or more amino acid identity or 99% or more amino acid identity are mentioned above.
変異型ユビキチンの例として、断定はしないが、脱ユビキチン化酵素に対する分解耐性を付与する変異を含むユビキチン変異体が挙げられる。そのようなユビキチン変異体の具体例として、配列番号1で示すヒトユビキチンにおいて、C末端側に位置する72位(開始コドンがコードするメチオニンを1位とする。以下同じ。)及び74位のアルギニン残基がそれぞれアラニン残基及びスレオニン残基に置換したアミノ酸置換を含むユビキチン変異体(R72A/R74T)、72位及び74位のアルギニン残基がそれぞれプロリン残基及びスレオニン残基に置換したアミノ酸置換を含むユビキチン変異体(R72P/R74T)、及び73位のロイシン残基がプロリン残基に置換したアミノ酸置換を含むユビキチン変異体(L73P)が挙げられる。ユビキチン変異体(R72A/R74T)の具体例としては、配列番号6で示すアミノ酸配列(MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLALTGG)からなるユビキチン変異体が挙げられる。ユビキチン変異体(R72P/R74T)の具体例としては、配列番号7で示すアミノ酸配列(MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLPLTGG)からなるユビキチン変異体が挙げられる。また、ユビキチン変異体(L73P)の具体例としては、配列番号8で示すアミノ酸配列(MQIFVKTLTGKTITLEVEPSDTIENVKAKIQDKEGIPPDQQRLIFAGKQLEDGRTLSDYNIQKESTLHLVLRPRGG)からなるユビキチン変異体が挙げられる。
Examples of mutant ubiquitin include ubiquitin mutants containing mutations that confer resistance to degradation by deubiquitinating enzymes, although this is not conclusive. As a specific example of such a ubiquitin mutant, in the human ubiquitin shown in SEQ ID NO: 1, arginine at position 72 (methionine encoded by the initiation codon is designated as
本明細書において「複数個」とは、例えば、2~20個、2~15個、2~10個、2~7個、2~5個、2~4個又は2~3個をいう。また、「数個」とは、2~3個をいう。 As used herein, "plurality" refers to, for example, 2 to 20, 2 to 15, 2 to 10, 2 to 7, 2 to 5, 2 to 4, or 2 to 3. In addition, "several pieces" means 2 to 3 pieces.
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種類のアミノ酸間において、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いう。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。これらの群内でのアミノ酸置換であれば、ペプチドの性質に変化を生じにくいことが知られているため好ましい。 As used herein, the term "substitution (of amino acids)" means that among the 20 types of amino acids that make up natural proteins, within a group of conservative amino acids with similar properties such as charge, side chain, polarity, and aromaticity It means replacement. For example, uncharged polar amino acids 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), substitutions within the group of aromatic amino acids (Phe, Tyr, Trp). Amino acid substitutions within these groups are preferred because they are known to be less likely to cause changes in peptide properties.
本明細書において「アミノ酸同一性」とは、比較する2つの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限となるように、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。アミノ酸同一性を算出するための2つのアミノ酸配列の整列化は、Blast、FASTA、ClustalW等の既知プログラムを用いて行うことができる。 As used herein, the term "amino acid identity" refers to the amino acid sequences of two peptides to be compared, with appropriate gaps inserted in one or both of the amino acid sequences to maximize the number of matching amino acid residues. It refers to the ratio (%) of the number of matching amino acid residues to the number of all amino acid residues when aligned. Alignment of two amino acid sequences for calculating amino acid identity can be performed using known programs such as Blast, FASTA, ClustalW.
本発明の標的ペプチド分解誘導剤において、結合因子に連結されるユビキチンの数は限定しない。例えば、1個であってもよいし、2個以上(例えば、2個、3個、4個、5個、6個、7個、8個、又は9個)連結したポリユビキチンであってもよい。2個以上が連結される場合、それぞれのユビキチンは同一であってもよいし、異なっていてもよい。例えば、配列番号1で示すアミノ酸配列からなるユビキチンと、配列番号1で示すアミノ酸配列からなるユビキチンのアミノ酸配列において1個が置換された変異ユビキチンが連結されていてもよい。 In the target peptide degradation inducer of the present invention, the number of ubiquitins linked to the binding factor is not limited. For example, it may be one, or two or more (for example, 2, 3, 4, 5, 6, 7, 8, or 9) linked polyubiquitin good. When two or more are linked, each ubiquitin may be the same or different. For example, a ubiquitin having the amino acid sequence shown in SEQ ID NO: 1 and a mutant ubiquitin having one substitution in the ubiquitin amino acid sequence having the amino acid sequence shown in SEQ ID NO: 1 may be linked.
(3)リンカー因子
「リンカー因子」とは、本発明の標的ペプチド分解誘導剤において結合因子とユビキチン間の連結を介在する分子(クロスリンカー)をいう。リンカー因子を含む標的ペプチド分解誘導剤では、結合因子とユビキチンがリンカー因子を介して間接的に連結される。
(3) Linker Factor The term "linker factor" refers to a molecule (crosslinker) that mediates the linkage between the binding factor and ubiquitin in the target peptide degradation inducer of the present invention. In the target peptide degradation inducer containing a linker factor, the binding factor and ubiquitin are indirectly linked via the linker factor.
リンカー因子の種類は問わない。例えば、核酸、ペプチド、及び/又は低分子化合物であればよい。これらの基本構成は、前述の結合因子に記載の核酸、ペプチド、及び/又は低分子化合物等の構成に準ずる。したがって、本明細書では核酸で構成されるリンカー因子を「核酸型リンカー因子」、ペプチドで構成されるリンカー因子を「ペプチド型リンカー因子」、そして低分子化合物で構成されるリンカー因子を「低分子化合物型リンカー因子」と表記する。また、リンカー因子は異なる複数種の組合せで構成されていてもよい。例えば、核酸型リンカー因子と低分子化合物型リンカー因子の組合せが挙げられる。この場合、各リンカー因子は、タンデムに連結されていればよく、そのような連結リンカー因子は、本明細書では「核酸/低分子化合物型リンカー因子」のように表記する。 The type of linker factor does not matter. For example, nucleic acids, peptides, and/or low-molecular-weight compounds may be used. The basic configuration of these conforms to the configuration of nucleic acids, peptides, and/or low-molecular-weight compounds described in the aforementioned binding factor. Therefore, in the present specification, a linker factor composed of a nucleic acid is referred to as a "nucleic acid-type linker factor," a linker factor composed of a peptide is referred to as a "peptide-type linker factor," and a linker factor composed of a low-molecular-weight compound is referred to as a "low-molecular-weight linker factor." compound-type linker factor". Moreover, the linker factor may be composed of a combination of different types. Examples include a combination of a nucleic acid-type linker factor and a low-molecular-weight compound-type linker factor. In this case, each linker factor may be linked in tandem, and such linked linker factors are referred to herein as "nucleic acid/low molecular weight compound type linker factors".
リンカー因子は、結合因子又はユビキチンのいずれか一方、又は両方と同じ種類であってもよいし、両方と異なる種類であってもよい。例えば、本発明の標的ペプチド分解誘導剤が核酸型結合因子、リンカー因子及びユビキチンで構成される場合、リンカー因子は核酸型結合因子と同じ核酸で構成されていてもよいし、ユビキチンと同じペプチドで構成されていてもよい。またはそれらのいずれとも異なる低分子化合物で構成されていてもよい。前例においてリンカー因子が核酸で構成される場合、核酸型結合因子とリンカー因子は融合された一本の核酸鎖として合成してもよい。 The linker factor may be of the same type as either the binding factor or ubiquitin, or both, or may be of a different type. For example, when the target peptide degradation-inducing agent of the present invention is composed of a nucleic acid-type binding factor, a linker factor, and ubiquitin, the linker factor may be composed of the same nucleic acid as the nucleic acid-type binding factor, or may be composed of the same peptide as ubiquitin. may be configured. Alternatively, it may be composed of a low-molecular-weight compound different from any of them. In the previous example, when the linker factor is composed of nucleic acid, the nucleic acid-type binding factor and the linker factor may be synthesized as a single fused nucleic acid strand.
リンカー因子のいくつかの具体例を以下で述べる。
まず、金属への配位結合を介して結合因子とユビキチンを連結するリンカー因子が挙げられる。例えば、ポリヒスチジン(His-tag)及びニッケル-ニトリロ三酢酸(Ni-NTA)からなるペプチド/低分子化合物型リンカー因子はその一例である。結合因子をNi-NTA修飾し、ユビキチンC末端にHis-tagを付加しておくことにより、両者の混合時にNi-NTAとHis-tagの結合を介して結合因子とユビキチンが連結される。
Some specific examples of linker elements are described below.
First, there is a linker factor that connects a binding factor and ubiquitin via a coordinate bond to a metal. For example, a peptide/low molecular compound type linker factor consisting of polyhistidine (His-tag) and nickel-nitrilotriacetic acid (Ni-NTA) is one example. By modifying the binding factor with Ni-NTA and attaching a His-tag to the C-terminus of ubiquitin, the binding factor and ubiquitin are linked via the binding of Ni-NTA and His-tag when both are mixed.
また、ホスト-ゲスト相互作用を介して結合因子とユビキチンを連結するリンカー因子が挙げられる。例えば、ダイヤモンドと同じ炭素配置構造を有する籠型分子のアダマンタンと環状オリゴ糖のβ-シクロデキストリンからなる低分子化合物型リンカー因子はその一例である。 In addition, linker factors that connect binding factors and ubiquitin via host-guest interactions are included. For example, a low-molecular-weight compound-type linker factor consisting of adamantane, a cage-like molecule having the same carbon configuration as diamond, and β-cyclodextrin, a cyclic oligosaccharide, is one example.
リンカー分子は、例えば、本発明の標的ペプチド分解誘導剤を製造する際に、結合因子とユビキチンとの直接的な連結が困難な場合等に、リンカー分子内に、それぞれと結合又は連結が可能な構成を包含させることにより、両者を容易に連結することができる。 The linker molecule can be bound or ligated to each within the linker molecule, for example, when direct ligation between the binding factor and ubiquitin is difficult when producing the target peptide degradation inducer of the present invention. By including the configuration, both can be easily connected.
1-3-2.構成因子間結合
本発明の標的ペプチド分解誘導剤における各構成因子間の直接的な結合は、特に限定はしない。生体内に投与された本発明の標的ペプチド分解誘導剤と標的ペプチドの結合により形成される複合体(標的ペプチド分解誘導剤-標的ペプチド複合体)がプロテアソームに取り込まれるまでの間、容易に解離しない結合であればよい。
1-3-2. Binding between Constituent Factors Direct binding between constituent factors in the target peptide degradation-inducing agent of the present invention is not particularly limited. The complex formed by the binding of the target peptide degradation inducer of the present invention administered in vivo to the target peptide (target peptide degradation inducer-target peptide complex) does not readily dissociate until it is incorporated into the proteasome. Any combination is acceptable.
「各構成因子間の直接的な結合」(本明細書では、しばしば「構成因子間結合」と表記する)とは、例えば、結合因子とユビキチン間、結合因子とリンカー因子間、及びユビキチンとリンカー因子間の結合をいう。そのような構成因子間結合の例として、共有結合、又はイオン結合のような化学結合が挙げられる。その他、ビオチンとアビジン間の結合のような高親和力による非共有結合も含まれる。 "Direct binding between constituent elements" (herein, often referred to as "binding between constituent elements") means, for example, between a binding agent and ubiquitin, between a binding agent and a linker A connection between factors. Examples of such component-to-component bonds include covalent bonds or chemical bonds such as ionic bonds. Others include high affinity non-covalent binding such as the binding between biotin and avidin.
構成因子間結合は、各構成因子の種類や構成に応じて、当該分野で公知の方法を用いて達成することができる。例えば、それぞれの因子に存在する官能基間の求核的付加反応、求核置換反応、又は求電子置換反応等の化学反応を介した結合が挙げられる。そのような結合に寄与する官能基は、限定はしないが相手分子との共有結合等による連結が可能な化学活性を有する活性官能基であることが好ましい。例えば、ヒドロキシ基(-OH)、ケトン基(-C(=O)-)、アルデヒド基(-CHO)、カルボキシ基(-COOH)、メトキシ基(-OCH3)、スルホ基(-SO3H)、アミノ基(-NH2)、オキシム(>C=N-OH)、カルボニル基(-C(=O)-)、チオール基(-SH)、シアノ基(-CN)、ニトロ基(-NO2)、又はアゾ基(-N=N-)を含む。構成因子間結合に寄与する官能基は、各構成因子が固有に包含する官能基であってもよいし、必要に応じて付加又は置換によって導入してもよい。例えば、ユビキチンのC末端にシステイン(C)を付加する、又はユビキチンのC末端アミノ酸である76位のグリシン(G)をシステインに置換して活性官能基であるチオール基をユビキチンのC末端に導入してもよい。 Bonding between constituent factors can be achieved using methods known in the art, depending on the type and composition of each constituent factor. Examples thereof include bonding via chemical reactions such as nucleophilic addition reactions, nucleophilic substitution reactions, or electrophilic substitution reactions between functional groups present in each factor. A functional group that contributes to such bonding is preferably, but not limited to, an active functional group having chemical activity that enables linkage with a partner molecule by covalent bonding or the like. For example, hydroxyl group (-OH), ketone group (-C(=O)-), aldehyde group (-CHO), carboxy group (-COOH), methoxy group ( -OCH3 ), sulfo group ( -SO3H ), amino group (-NH 2 ), oxime (>C=N-OH), carbonyl group (-C(=O)-), thiol group (-SH), cyano group (-CN), nitro group (- NO 2 ), or an azo group (-N=N-). A functional group that contributes to binding between constituent factors may be a functional group inherently included in each constituent factor, or may be introduced by addition or substitution as necessary. For example, cysteine (C) is added to the C-terminus of ubiquitin, or glycine (G) at position 76, which is the C-terminal amino acid of ubiquitin, is replaced with cysteine to introduce a thiol group, which is an active functional group, to the C-terminus of ubiquitin. You may
官能基間の結合の場合、構成因子間結合を形成する両官能基の種類は限定しないが、官能基間で共有結合を形成し得る組み合わせを選択することが望ましい。例えば、アミノ基とアルデヒド基、チオール基とマレイミド基、アジド基とエチニル基、ヒドラジン基とケトン基、及びヒドラジン基とアルデヒド基が含まれる。 In the case of bonding between functional groups, the types of both functional groups forming a bond between constituent factors are not limited, but it is desirable to select a combination that can form a covalent bond between the functional groups. Examples include amino and aldehyde groups, thiol and maleimide groups, azido and ethynyl groups, hydrazine and ketone groups, and hydrazine and aldehyde groups.
構成因子間結合を形成する官能基間の結合の種類は限定しない。例えば、チオール基を介した結合、アジド-アルキン環化付加反応(Huisgen環化付加反応)による結合、又はオキシム/ヒドラゾン結合等が挙げられる。チオール基を介した結合の具体例としては、マイケル付加反応、チオールエン反応、又はチオールイン反応による結合、もしくはジスルフィド結合が挙げられる。 The type of bond between functional groups that form the bond between constituent factors is not limited. For example, a bond via a thiol group, a bond by an azide-alkyne cycloaddition reaction (Huisgen cycloaddition reaction), an oxime/hydrazone bond, or the like can be mentioned. Specific examples of bonding via a thiol group include bonding by Michael addition reaction, thiolene reaction, or thiolyne reaction, or disulfide bond.
1-3-3.構成因子の標識
本発明の標的ペプチド分解誘導剤の構成因子は、必要に応じて標識されていてもよい。標識は各因子の種類に応じて、当該分野で公知の標識物質を利用すればよい。
1-3-3. Labeling of Constituent Factors Constituent factors of the target peptide degradation-inducing agent of the present invention may be labeled as necessary. For labeling, a labeling substance known in the art may be used depending on the type of each factor.
例えば、核酸型結合因子や核酸型リンカー因子のような核酸を標識する場合であれば、リン酸基、糖、及び/又は塩基を標識すればよい。また、ユビキチン、ペプチド型結合因子、又はペプチド型リンカー因子のようなペプチドを標識する場合であれば、構成アミノ酸残基を標識すればよい。さらに、低分子化合物型結合因子又は低分子化合物型リンカー因子のような低分子化合物であれば、包含する官能基を標識すればよい。標識する構成因子の種類も限定はしない。ただし、結合因子の標識が簡便であり、標的ペプチドとの結合を検出する際にも直接的で便利であることから好ましい。また、複数の構成因子を標識してもよい。 For example, when labeling a nucleic acid such as a nucleic acid-type binding factor or a nucleic acid-type linker factor, the phosphate group, sugar, and/or base may be labeled. In addition, when labeling a peptide such as ubiquitin, a peptide-type binding factor, or a peptide-type linker factor, the constituent amino acid residues may be labeled. Furthermore, in the case of a low-molecular-weight compound such as a low-molecular-weight compound-type binding factor or a low-molecular-weight compound-type linker factor, the included functional groups may be labeled. The type of constituent factor to be labeled is also not limited. However, labeling of the binding agent is convenient, and is preferred because it is direct and convenient when detecting binding to the target peptide. Also, multiple constituent factors may be labeled.
いずれの構成因子の場合であっても標識位置は、その標識物質の特性や使用目的に応じて適宜定めればよく、特に限定はしない。核酸型の因子であれば、5'末端部及び/又は3'末端部が標識部位として好適に利用される。また、ペプチド型の因子であれば、各アミノ酸残基の側鎖、又はN末端アミノ酸残基のアミノ基、若しくはC末端アミノ酸残基のカルボキシ基が標識部位として好適である。さらに、低分子化合物型の因子であれば、包含する官能基を標識することができる。 In the case of any constituent factor, the labeling position may be appropriately determined according to the characteristics and purpose of use of the labeling substance, and is not particularly limited. In the case of a nucleic acid-type factor, the 5'-end and/or 3'-end is preferably used as the labeling site. In the case of a peptide-type factor, the side chain of each amino acid residue, the amino group of the N-terminal amino acid residue, or the carboxy group of the C-terminal amino acid residue is suitable as the labeling site. Furthermore, if it is a low-molecular-weight compound-type factor, the functional groups it contains can be labeled.
標識物質は、当該分野で公知のあらゆる物質を利用することができる。例えば、放射性同位元素、蛍光物質、クエンチャー、化学発光物質、DIG、ビオチン、又は磁気ビーズ等が挙げられる。 Any substance known in the art can be used as the labeling substance. Examples thereof include radioisotopes, fluorescent substances, quenchers, chemiluminescent substances, DIG, biotin, magnetic beads and the like.
「放射性同位元素」とは、質量数が異なる同位元素のうち、放射線を放出する元素をいう。例えば、32P、3H、14Cが挙げられる。 “Radioisotope” refers to an element that emits radiation among isotopes with different mass numbers. Examples include 32P , 3H , 14C .
「蛍光物質」とは、特定波長の励起光を吸収することで励起状態となり、元の基底状態に戻る際に蛍光を発する性質を有する物質をいう。例えば、FITC、ローダミン、フルオレサミン、フルオレセイン、Texas Red[登録商標]、Cy3、Cy5、Cy7、FAM、HEX、JOE、ROX、TET、Bodipy493、NBD、TAMRA、Quasar[登録商標]、CAL Fluor(登録商標)Red610、SYBR Green[登録商標]、Eva Green[登録商標]、SYTOX Green[登録商標]等が挙げられる。 "Fluorescent substance" refers to a substance that has the property of being excited by absorbing excitation light of a specific wavelength and emitting fluorescence when returning to the original ground state. For example, FITC, Rhodamine, Fluorescamine, Fluorescein, Texas Red®, Cy3, Cy5, Cy7, FAM, HEX, JOE, ROX, TET, Bodipy493, NBD, TAMRA, Quasar®, CAL Fluor® ) Red610, SYBR Green [registered trademark], Eva Green [registered trademark], SYTOX Green [registered trademark], and the like.
「クエンチャー」とは、前記蛍光物質の励起エネルギーを吸収し、蛍光を抑制する性質を有する物質をいう。例えば、AMRA、DABCYL、BHQ-1、BHQ-2、又はBHQ-3等が挙げられる。 "Quencher" refers to a substance that absorbs the excitation energy of the fluorescent substance and has the property of suppressing fluorescence. Examples include AMRA, DABCYL, BHQ-1, BHQ-2, or BHQ-3.
「化学発光物質」とは、化学反応によって励起された後、基底状態に戻る際に、差分のエネルギーを光として放出する性質を有する物質をいう。例えば、アクリジニウムエステル等が挙げられる。 A "chemiluminescent substance" is a substance that has the property of emitting the differential energy as light when returning to the ground state after being excited by a chemical reaction. For example, an acridinium ester etc. are mentioned.
各構成因子への標識は、同一の、又は異なる二以上の標識物質で複数箇所を標識することができる。標識物質で標識された構成因子は、標的ペプチド分解誘導剤を検出する際に有用なツールとなり得る。 Each constituent factor can be labeled at multiple locations with two or more of the same or different labeling substances. A component labeled with a labeling substance can be a useful tool in detecting a target peptide degradation inducer.
1-4.効果
本発明の標的ペプチド分解誘導剤によれば、標的ペプチドに結合して複合体を形成することにより、複合体全体として標的ペプチドを間接的にユビキチン化することができる。その結果、標的ペプチドを直接的にユビキチン化することなく、プロテアソームに誘導し、分解することができる。これにより、低分子化合物や抗体医薬のみならず、直接的なユビキチン化が困難なためアンドラッガブルであった標的ペプチドであっても、創薬ターゲットとすることが可能になる。その結果、分子標的医薬分野が飛躍的に拡大し、医薬品開発設計の選択肢が大きく広がることが期待できる。
1-4. Effect According to the target peptide degradation inducer of the present invention, the target peptide can be indirectly ubiquitinated as a whole by binding to the target peptide to form a complex. As a result, it can be directed to the proteasome for degradation without direct ubiquitination of the target peptide. As a result, not only low-molecular-weight compounds and antibody drugs, but also target peptides that have been undraggable due to the difficulty of direct ubiquitination can be used as drug discovery targets. As a result, the field of molecular-targeted medicine will expand dramatically, and it is expected that the options for drug development and design will greatly expand.
本発明の標的ペプチド分解誘導剤によれば、標的ペプチドと特異的に結合する結合因子を選択することで、任意の標的ペプチドをUb-PSM系にて分解することが可能となる。これにより、従来ユビキチン化が困難であった標的ペプチドであってもUb-PSM系による選択的分解が実現できる。例えば、結合因子をカスパーゼに結合する分子にした場合、その標的ペプチド分解誘導剤はアポトーシスの抑制剤となり得る。また、結合因子をAtg因子に結合する分子にした場合、その標的ペプチド分解誘導剤はオートファジー抑制剤になり得、さらに結合因子をmTOR複合体1又はGAPR-1に結合する分子にした場合、その標的ペプチド分解誘導剤はオートファジー促進剤になり得る。 According to the target peptide degradation inducer of the present invention, any target peptide can be degraded by the Ub-PSM system by selecting a binding factor that specifically binds to the target peptide. This enables selective degradation by the Ub-PSM system even for target peptides that have been difficult to ubiquitinate in the past. For example, if the binding factor is a molecule that binds to caspase, the target peptide degradation inducer can be an apoptosis inhibitor. When the binding factor is a molecule that binds to Atg factor, the target peptide degradation inducer can be an autophagy inhibitor. The target peptide degradation inducer can be an autophagy promoter.
2.核酸型標的ペプチド分解誘導剤
2-1.概要
本発明の第2の態様は、核酸型標的ペプチド分解誘導剤である。本発明の核酸型標的ペプチド分解誘導剤は、ペプチドのみで構成される前記第1態様に記載の標的ペプチド分解誘導剤をコードするポリヌクレオチド、又はそれを含む発現ベクターで構成される。本発明の核酸型標的ペプチド分解誘導剤は、比較的安定性が高く、保存が容易な上、生体内に投与後、活性を有する標的ペプチド分解誘導剤を増幅することが可能となる。
2. Nucleic acid type target peptide degradation inducer 2-1. Overview A second aspect of the present invention is a nucleic acid-type target peptide degradation inducer. The nucleic acid-type target peptide degradation-inducing agent of the present invention is composed of a polynucleotide encoding the target peptide degradation-inducing agent according to the first aspect, which is composed only of peptides, or an expression vector containing the same. The nucleic acid-type target peptide degradation inducer of the present invention has relatively high stability, is easy to store, and can amplify an active target peptide degradation inducer after administration in vivo.
2-2.構成
「核酸型標的ペプチド分解誘導剤」とは、核酸で構成された標的ペプチド分解誘導剤をいう。
核酸型標的ペプチド分解誘導剤は、第1態様に記載の標的ペプチド分解誘導剤がペプチドのみで構成される場合、そのアミノ酸配列をコードするポリヌクレオチド、又はそのポリヌクレオチドを含む発現ベクターで構成される。ここでいうポリヌクレオチドは、原則として天然型ポリヌクレオチドである。DNA及び/又はRNAのいずれかで構成されるが、DNAのみで構成されるポリヌクレオチドが好適である。
2-2. Structure "Nucleic acid-type target peptide degradation inducer" refers to a target peptide degradation inducer composed of a nucleic acid.
When the target peptide degradation-inducing agent according to the first aspect is composed only of a peptide, the nucleic acid-type target peptide degradation-inducing agent is composed of a polynucleotide encoding the amino acid sequence, or an expression vector containing the polynucleotide. . Polynucleotides here are, in principle, natural polynucleotides. Although composed of either DNA and/or RNA, polynucleotides composed solely of DNA are preferred.
ペプチドのみで構成される標的ペプチド分解誘導剤とは、ペプチド型結合因子及びユビキチン、又はペプチド型結合因子、ペプチド型リンカー因子及びユビキチンで構成された標的ペプチド分解誘導剤が該当する。 A target peptide degradation inducer composed only of a peptide corresponds to a peptide-type binding factor and ubiquitin, or a target peptide degradation inducer composed of a peptide-type binding factor, a peptide-type linker factor, and ubiquitin.
本明細書において「発現ベクター」とは、遺伝子や遺伝子断片(本発明においては、前記ポリヌクレオチド)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いう。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前記遺伝子等が配置され、プロモーターの活性化により前記遺伝子等の発現を誘導できる状態をいう。発現ベクターの種類は問わない。本発明の核酸型標的ペプチド分解誘導剤を投与する被験体の生体内で、包含する前記プロモーターが活性化し得るあらゆる発現ベクターを利用することができる。例えば、被験体がヒトの場合には、ウイルスベクターが挙げられる。ウイルスベクターには、レトロウイルス(レンチウイルスを含む)、アデノウイルス、アデノ随伴ウイルス等に由来する種々のベクターが含まれる。 As used herein, the term "expression vector" refers to an expression unit that contains a gene or gene fragment (in the present invention, the polynucleotide) in an expressible state and can control the expression of the gene or the like. As used herein, the term "expressible state" refers to a state in which the gene or the like is placed under the control of a promoter and the expression of the gene or the like can be induced by activation of the promoter. Any type of expression vector can be used. Any expression vector whose promoter can be activated in vivo in a subject to whom the nucleic acid-type target peptide degradation inducer of the present invention is administered can be used. For example, when the subject is human, viral vectors are included. Viral vectors include various vectors derived from retroviruses (including lentiviruses), adenoviruses, adeno-associated viruses, and the like.
本態様の核酸型標的ペプチド分解誘導剤の機能及び効果は、第1態様に記載の標的ペプチド分解誘導剤に準ずる。しかし、核酸で構成されているという構造的な相違から、その作用機序が異なる。すなわち、第1態様に記載の標的ペプチド分解誘導剤はそれ自身には標的ペプチドを分解誘導する活性はなく、生体投与後に標的ペプチドに直接作用するのに対して、本態様の核酸型標的ペプチド分解誘導剤は、投与後に細胞内で核酸型標的ペプチド分解誘導剤にコードされた標的ペプチド分解誘導剤の発現を経て、その発現した標的ペプチド分解誘導剤が標的ペプチドに作用する。これにより、生体内で標的ペプチド分解誘導剤を増幅することが可能となる。 The functions and effects of the nucleic acid-type target peptide degradation inducer of this aspect conform to those of the target peptide degradation inducer described in the first aspect. However, due to the structural difference that they are composed of nucleic acids, their mechanisms of action are different. That is, the target peptide degradation-inducing agent according to the first aspect does not itself have the activity of inducing degradation of the target peptide, and acts directly on the target peptide after administration to the living body, whereas the nucleic acid-type target peptide degradation-inducing agent of this aspect has no activity to induce degradation of the target peptide. After administration of the inducer, the target peptide degradation inducer encoded by the nucleic acid-type target peptide degradation inducer is expressed intracellularly, and the expressed target peptide degradation inducer acts on the target peptide. This makes it possible to amplify the target peptide degradation inducer in vivo.
3.標的ペプチド分解誘導組成物
3-1.概要
本発明の第3の態様は、標的ペプチド分解誘導組成物である。本発明の組成物は、第1態様に記載の標的ペプチド分解誘導剤、及び/又は第2態様に記載の核酸型標的ペプチド分解誘導剤のいずれか2種以上を必須の構成因子として包含する。本発明のペプチド分解誘導組成物によれば、標的ペプチド分解誘導剤等を安定的に、また医薬組成物として、生体投与時の負荷や侵襲性を低減し、より投与しやすい形態で提供することができる。
3. Target Peptide Degradation Inducing Composition 3-1. Overview A third aspect of the present invention is a target peptide degradation-inducing composition. The composition of the present invention includes any two or more of the target peptide degradation inducer according to the first aspect and/or the nucleic acid-type target peptide degradation inducer according to the second aspect as essential constituents. According to the peptide degradation-inducing composition of the present invention, a target peptide degradation-inducing agent or the like can be stably provided as a pharmaceutical composition in a form that reduces the burden and invasiveness upon administration to the living body and is easier to administer. can be done.
3-2.構成
3-2-1.構成因子
本態様の標的ペプチド分解誘導組成物は(本明細書では、以下、しばしば「本組成物」と略称する)、必須の構成因子として有効成分を、また選択的構成因子として担体及び/又は溶媒を含む。以下、それぞれの構成因子について具体的に説明をする。
3-2. Configuration 3-2-1. Constituent Factors The target peptide degradation-inducing composition of this embodiment (hereinbelow, often abbreviated as "this composition") comprises an active ingredient as an essential constituent factor, and a carrier and/or Contains solvent. Each component will be specifically described below.
(1)有効成分
本組成物は、第1態様に記載の標的ペプチド分解誘導剤及び/又は第2態様に記載の核酸型標的ペプチド分解誘導剤(本明細書では、これらをまとめて「POI分解誘導剤等」と表記する)を必須の有効成分として含む。本組成物は、それぞれ異なる2種以上のPOI分解誘導剤等を含むことができる。また、POI分解誘導剤等以外で、PROTAC(登録商標)のような他の公知の標的タンパク質分解誘導剤を有効成分として選択的に含んでいてもよい。
(1) Active ingredient The present composition comprises the target peptide degradation inducer according to the first aspect and/or the nucleic acid-type target peptide degradation inducer according to the second aspect (in this specification, these are collectively referred to as "POI degradation (referred to as "inducing agent, etc.") as an essential active ingredient. The present composition can contain two or more different POI degradation inducers and the like. In addition to POI degradation inducers and the like, other known target protein degradation inducers such as PROTAC (registered trademark) may be selectively included as active ingredients.
本組成物に含まれるPOI分解誘導剤等の含有量は、包含されるPOI分解誘導剤等の種類及び/又はその有効量、本組成物の剤形、後述する担体又は添加物の種類、並びに疾患の種類によって異なるため、それぞれの条件を勘案して適宜定めればよい。 The content of the POI decomposition inducer, etc. contained in the composition includes the type and/or effective amount of the POI decomposition inducer, etc., the dosage form of the composition, the type of carrier or additive described later, and Since it varies depending on the type of disease, it may be determined appropriately in consideration of each condition.
本明細書において「有効量」とは、本組成物においてPOI分解誘導剤等が有効成分として、その機能を発揮する上で必要な量で、かつそれを適用する生体に対して有害な副作用を、ほとんど又は全く生じない量をいう。この有効量は、被験体の情報、投与経路、及び投与回数等の様々な条件によって変化し得る。 As used herein, the term "effective amount" refers to an amount necessary for the POI decomposition inducer, etc., as an active ingredient in the present composition, to exhibit its function, and to cause no harmful side effects to the living body to which it is applied. , refers to the amount that occurs little or not at all. This effective amount may vary depending on various conditions such as subject information, route of administration, and frequency of administration.
本明細書において「被験体」とは、POI分解誘導剤等又は本組成物の適用対象となる生体をいう。例えば、ヒト、家畜(ウシ、ウマ、ヒツジ、ヤギ、ブタ、ニワトリ、ダチョウ等)、競走馬、愛玩動物(イヌ、ネコ、ウサギ等)、実験動物(マウス、ラット、モルモット、サル等)等が該当する。好ましくはヒトである(この場合、特に「被験者」と称する)。また、「被験体の情報」とは、適用する生体の様々な個体情報であって、例えば、被験者の場合であれば、全身の健康状態、疾患・病害に罹患している場合にはその進行度や重症度、年齢、体重、性別、食生活、薬剤感受性、併用薬物の有無及び治療に対する耐性等を含む。本組成物におけるPOI分解誘導剤等の有効量、及びそれに基づいて算出される適用量は、個々の被験体の情報等に応じて、最終的には医師、歯科医師、又は獣医師等の判断によって決定される。 As used herein, the term "subject" refers to a living organism to which the POI degradation inducer or the like or the present composition is applied. For example, humans, livestock (cows, horses, sheep, goats, pigs, chickens, ostriches, etc.), racehorses, pet animals (dogs, cats, rabbits, etc.), laboratory animals (mouse, rats, guinea pigs, monkeys, etc.), etc. Applicable. Humans are preferred (in this case, they are particularly referred to as "subjects"). In addition, "subject information" refers to various individual information of the living body to be applied. Including degree and severity, age, body weight, sex, diet, drug sensitivity, presence or absence of concomitant drugs, tolerance to treatment, etc. The effective amount of the POI degradation inducer, etc. in this composition and the applicable amount calculated based thereon are ultimately determined by doctors, dentists, veterinarians, etc., depending on the information of individual subjects. determined by
(2)溶媒
本組成物は、薬学的に許容可能な溶媒を含むことができる。溶媒は、本組成物における選択的構成因子であり、必要に応じて添加すればよい。「薬学的に許容可能な」とは、生体に無害又は低毒性で、製剤技術分野において通常使用可能な、好ましくは医薬組成物に使用可能なことをいう。
(2) Solvent The composition can contain a pharmaceutically acceptable solvent. Solvents are optional constituents in the composition and may be added as needed. “Pharmaceutically acceptable” means harmless or low toxicity to living bodies, and can be used normally in the field of formulation technology, preferably in pharmaceutical compositions.
溶媒には、例えば、水若しくは水溶液、又は有機溶剤が挙げられる。薬学的に許容可能な水溶液には、例えば、生理食塩水、ブドウ糖やその他の補助剤を含む等張液、リン酸塩緩衝液、酢酸ナトリウム緩衝液が挙げられる。ここでいう補助剤には、例えば、D-ソルビトール、D-マンノース、D-マンニトール、塩化ナトリウム、その他にも低濃度の非イオン性界面活性剤、ポリオキシエチレンソルビタン脂肪酸エステル類等が挙げられる。薬学的に許容可能な有機溶媒には、例えば、エタノール、ブタノール等が挙げられる。 Solvents include, for example, water or aqueous solutions, or organic solvents. Pharmaceutically acceptable aqueous solutions include, for example, physiological saline, isotonic solutions containing glucose and other adjuvants, phosphate buffers, sodium acetate buffers. Examples of adjuvants here include D-sorbitol, D-mannose, D-mannitol, sodium chloride, low-concentration nonionic surfactants, polyoxyethylene sorbitan fatty acid esters, and the like. Pharmaceutically acceptable organic solvents include, for example, ethanol, butanol, and the like.
(3)担体
本組成物は、薬学的に許容可能な担体を含むことができる。担体は、本組成物における選択的構成因子であり、必要に応じて添加すればよい。
(3) Carrier The composition can contain a pharmaceutically acceptable carrier. A carrier is an optional component in the composition and may be added as needed.
担体には、例えば、懸濁剤、希釈剤、可溶化剤、分散剤、界面活性剤、乳化剤、無痛化剤、安定剤、保存剤、防腐剤、抗酸化剤、緩衝剤、及び等張化剤等が挙げられる。 Carriers include, for example, suspending agents, diluents, solubilizers, dispersing agents, surfactants, emulsifying agents, soothing agents, stabilizers, preservatives, preservatives, antioxidants, buffers, and tonicity agents. agents and the like.
上記の他にも、必要であれば医薬において通常用いられる賦形剤、充填剤、結合剤、崩壊剤、吸収促進剤、増量剤、付湿剤、保湿剤、湿潤剤、吸着剤、崩壊抑制剤、コーティング剤、着色剤等を適宜含むこともできる。 In addition to the above, excipients, fillers, binders, disintegrants, absorption enhancers, bulking agents, humectants, humectants, humectants, adsorbents, and disintegration inhibitors commonly used in pharmaceuticals, if necessary Agents, coating agents, coloring agents and the like may also be included as appropriate.
このような担体は、主として剤形形成を容易にし、また剤形及び薬剤効果を維持する他、有効成分であるPOI分解誘導剤等が生体内での分解を受け難くするために用いられるものであって、必要に応じて適宜使用すればよい。 Such a carrier is mainly used to facilitate the formation of a dosage form, maintain the dosage form and drug effect, and make the POI degradation inducer, etc., which is an active ingredient, difficult to be decomposed in vivo. It should be used appropriately as needed.
3-2-2.剤形
本組成物の剤形は、有効成分であるPOI分解誘導剤等及び他の公知の標的タンパク質分解誘導剤を不活化させず、投与後に生体内でその有効成分の薬理効果を発揮し得る形態であれば特に限定しない。また、本組成物の具体的な剤形は、投与法及び/又は処方条件に応じて適宜選択すればよい。一般に投与法は、経口投与と非経口投与に大別することができるが、本組成物はそれぞれの投与法に適した剤形にすればよい。
3-2-2. Dosage form The dosage form of the present composition does not inactivate the POI degradation inducer and other known target protein degradation inducers, which are active ingredients, and can exhibit the pharmacological effect of the active ingredient in vivo after administration. There is no particular limitation as long as it is in a form. Moreover, the specific dosage form of the present composition may be appropriately selected according to the administration method and/or prescription conditions. In general, administration methods can be broadly classified into oral administration and parenteral administration, and the present composition may be formulated in dosage forms suitable for each administration method.
経口投与の場合、剤形としては、固形剤(錠剤、丸剤、舌下剤、カプセル剤、ドロップ剤を含む)、顆粒剤、粉剤、散剤、液剤(内用水剤、懸濁剤、乳剤、シロップ剤を含む)等が挙げられる。固形剤は、必要に応じて当該技術分野で公知の剤皮を施した剤形、例えば、糖衣錠、ゼラチン被包錠、腸溶錠、フィルムコーティング錠、二重錠、多層錠にすることができる。 For oral administration, the dosage forms include solids (tablets, pills, sublinguals, capsules, drops), granules, powders, powders, liquids (internal liquids, suspensions, emulsions, syrups). including agents) and the like. Solid formulations can optionally be in the form of coatings known in the art, such as sugar-coated tablets, gelatin-coated tablets, enteric-coated tablets, film-coated tablets, double tablets, and multi-layer tablets. .
非経口投与の場合、全身投与及び局所投与に分けることができ、また局所投与は、さらに組織内投与、経表皮投与、経粘膜投与及び経直腸的投与にさらに細分することもできる。本組成物もそれぞれの投与法に適した剤形にすればよい。例えば、全身投与又は組織内投与に適した剤形としては、液剤である注射剤を挙げることができる。経表皮投与又は経粘膜投与に適した剤形としては、液剤(塗布剤、点眼剤、点鼻剤、吸引剤を含む)、懸濁剤(乳剤、クリーム剤を含む)、粉剤(点鼻剤、吸引剤を含む)、ペースト剤、ゲル剤、軟膏剤、硬膏剤等を挙げることができる。経直腸的投与に適した剤形としては、坐剤等を挙げることができる。 Parenteral administration can be divided into systemic administration and topical administration, and local administration can be further subdivided into tissue administration, transepidermal administration, transmucosal administration, and transrectal administration. The composition may also be formulated in dosage forms suitable for each administration method. For example, dosage forms suitable for systemic administration or intra-tissue administration include liquid injections. Dosage forms suitable for transepidermal or transmucosal administration include liquids (including ointments, eye drops, nasal drops, and inhalants), suspensions (including emulsions and creams), and powders (nasal drops). , inhalants), pastes, gels, ointments, plasters, and the like. Dosage forms suitable for rectal administration include suppositories and the like.
本組成物は、第4態様に記載のように、様々な疾患の治療用又は予防用として適用することができる。適用対象部位は、対象疾患に応じて変動する。例えば、対象疾患が神経変性疾患であれば、主に中枢神経系である脳脊髄となる。したがって、投与方法は限定されないが、組織内投与か循環系を介した全身投与が使用され得る。具体的には、例えば、脳脊髄内注射による組織内投与、又は血管内注射(静脈内注射、動脈内注射を含む)若しくはリンパ管内注射等の循環器内投与が挙げられる。また、対象疾患が癌であれば、適用対象部位は主に原発巣及び転移巣となる。この場合も投与方法は限定されないが、原発巣及び転移巣への組織内投与か循環系を介した全身投与が使用され得る。 The present composition can be applied for treatment or prevention of various diseases as described in the fourth aspect. The target site for application varies depending on the target disease. For example, if the target disease is a neurodegenerative disease, it will mainly be the cerebrospinal cord, which is the central nervous system. Therefore, the method of administration is not limited, but intratissue administration or systemic administration via the circulatory system can be used. Specific examples include intratissue administration by intracerebrospinal injection, or intracirculatory administration such as intravascular injection (including intravenous injection and intraarterial injection) or intralymphatic injection. In addition, if the target disease is cancer, the application target sites are mainly primary lesions and metastatic lesions. In this case, too, the method of administration is not limited, but intra-tissue administration to primary lesions and metastatic lesions or systemic administration via the circulatory system can be used.
注射剤には、前記乳化剤、懸濁剤、界面活性剤、安定剤、pH調節剤等と適宜組み合わせて、一般に認められた製薬実施で要求される単位用量形態で混和することによって製剤化すればよく、単位用量アンプル又は多用量容器の状態で提供される。 Injectables can be prepared by appropriately combining the aforementioned emulsifying agents, suspending agents, surfactants, stabilizers, pH adjusters, etc., and mixing them in a unit dose form generally required for pharmaceutical practice. They are often provided in unit dose ampoules or in multi-dose containers.
なお、上記各剤形の具体的な形状、大きさについては、いずれもそれぞれの剤形において当該分野で公知の剤形の範囲内にあればよく、特に限定はしない。 The specific shape and size of each dosage form described above are not particularly limited as long as they are within the range of dosage forms known in the art for each dosage form.
4.疾患抑制剤
4-1.概要
本発明の第4の態様は、疾患抑制剤である。本発明の疾患抑制剤は、前記第1態様に記載の標的ペプチド分解誘導剤の一形態であって、特定の疾患原因となるタンパク質を標的ペプチドとする。本発明の疾患抑制剤は、各種疾患の原因となるタンパク質を標的ペプチドとし、その分解を誘導することで、様々な疾患を治療又は予防することができる。
4. Disease Suppressant 4-1. Overview A fourth aspect of the present invention is a disease suppressing agent. The disease-suppressing agent of the present invention is one form of the target peptide degradation-inducing agent according to the first aspect, and has a specific disease-causing protein as a target peptide. The disease-suppressing agent of the present invention can treat or prevent various diseases by using proteins that cause various diseases as target peptides and inducing their degradation.
本発明の疾患抑制剤によれば、転写因子のように、ユビキチン創薬においてはアンドラッガブルとされている核酸結合性タンパク質であっても分解対象とすることができる。それによって、分子標的医薬の開発設計における選択肢が拡大し、様々な疾病に対する幅広い創薬及び創薬技術を提供することができる。 According to the disease suppressor of the present invention, even nucleic acid-binding proteins that are considered undruggable in ubiquitin drug discovery, such as transcription factors, can be targeted for degradation. As a result, the options in the development and design of molecular-targeted drugs are expanded, and a wide range of drug discovery and drug discovery technologies for various diseases can be provided.
4-2.構成
本明細書において「疾患抑制剤」とは、特定の疾患の発症を抑制し、又は悪化若しくは慢性化を軽減又は治療する薬剤をいう。
4-2. Configuration As used herein, the term "disease suppressing agent" refers to an agent that suppresses the onset of a specific disease, or alleviates or treats the aggravation or chronicity of a specific disease.
本発明の疾患抑制剤は、結合因子及びユビキチンを必須の構成因子として、またリンカー因子を選択的な構成因子として含む。その基本構成は、第1態様に記載の標的ペプチド分解誘導剤に準ずる。それ故に、共通する構成についての具体的な説明は省略し、ここでは本発明の疾患抑制剤に特徴的な点について説明をする。 The disease suppressor of the present invention contains a binding factor and ubiquitin as essential constituent factors, and a linker factor as an selective constituent factor. Its basic configuration conforms to the target peptide degradation inducer described in the first aspect. Therefore, the specific description of the common configuration is omitted, and the characteristic points of the disease suppressing agent of the present invention are described here.
本発明の疾患抑制剤は、結合因子が疾患原因ペプチドに結合することを特徴とする。つまり、本態様の疾患抑制剤では疾患原因ペプチドが標的ペプチドとなる。 The disease-suppressing agent of the present invention is characterized in that the binding factor binds to the disease-causing peptide. In other words, the disease-causing peptide is the target peptide in the disease-suppressing agent of this embodiment.
本明細書において「疾患原因ペプチド」とは、疾患の発症、悪化、又は慢性化等の原因となるペプチド(タンパク質を含む)をいう。そのような疾患原因ペプチドは、野生型ペプチドの他、機能獲得型(活性化型)、機能亢進型、又は機能喪失型等の変異型ペプチドを含む。 As used herein, the term "disease-causing peptide" refers to a peptide (including protein) that causes the onset, exacerbation, or chronicity of a disease. Such disease-causing peptides include wild-type peptides as well as mutant peptides such as gain-of-function (activation), hyperfunction, and loss-of-function.
本発明の疾患抑制剤の対象となる疾患の種類は、疾患原因がペプチドであれば限定しない。前述のように、標的ペプチドは、転写因子、アミロイド、タウタンパク質、α-シヌクレイン、カスパーゼ、変性ハンチンチン、偽キナーゼ、K-RAS変異体、Atg因子、mTOR複合体1、GAPR-1及びオーファン受容体等を含むが、それらが原因となる疾患は、いずれも本発明の疾患抑制剤の対象となり得る。
The type of disease targeted by the disease suppressing agent of the present invention is not limited as long as the cause of the disease is a peptide. As previously mentioned, target peptides include transcription factors, amyloid, tau protein, α-synuclein, caspases, denatured huntingtin, pseudokinases, K-RAS mutants, Atg factors,
第1態様に記載の標的ペプチド分解誘導剤は、結合因子を介して標的ペプチドを間接的にユビキチン化することによりUb-PSM系による分解を誘導する。したがって、疾患原因ペプチドに結合する結合因子を選択することで、様々な疾患治療薬又は予防薬となり得る。以下、本発明の疾患抑制剤の対象疾患とその疾患原因ペプチドとの関連について、いくつかの例を挙げて説明をする。 The target peptide degradation inducer according to the first aspect induces degradation by the Ub-PSM system by indirectly ubiquitylating the target peptide via a binding factor. Therefore, by selecting a binding factor that binds to a disease-causing peptide, it can be used as a therapeutic or preventive drug for various diseases. The relationship between the target disease of the disease-suppressing agent of the present invention and the disease-causing peptide will be described below with some examples.
(1)抗癌剤
本発明の疾患抑制剤は、抗癌剤となり得る。この場合、疾患抑制剤は癌の原因となる疾患原因ペプチドに結合する結合因子を含む。そのような疾患原因ペプチドの一例として、NF-κBのような転写因子、HER3のような偽キナーゼ群(pseudokinase)(Xie T., et al., 2014, Nat. Chem. Biol., 10: 1006-1011)、K-RAS変異体(Bond M.J., et al., 2020, ACS Cent. Sci. 6: 1367-1375)、又はERR-α等のオーファン受容体等が挙げられるが、それに限定はしない。これらの疾患原因ペプチドは、従来の低分子化合物からなる阻害剤では機能阻害が困難とされてきたが、本発明の疾患抑制剤ではそれぞれの疾患原因ペプチドに結合する結合因子を取得できれば、Ub-PSM系を介した疾患原因ペプチドの分解によるノックダウンにより癌の治療、改善又は進行抑制が可能となる。
(1) Anticancer agent The disease inhibitor of the present invention can be an anticancer agent. In this case, the disease-suppressing agent comprises a binding agent that binds to a cancer-causing disease-causing peptide. Examples of such disease-causing peptides include transcription factors such as NF-κB, pseudokinases such as HER3 (Xie T., et al., 2014, Nat. Chem. Biol., 10: 1006). -1011), K-RAS mutants (Bond MJ, et al., 2020, ACS Cent. Sci. 6: 1367-1375), or orphan receptors such as ERR-α, but are not limited to do not. It has been difficult to inhibit the function of these disease-causing peptides with inhibitors composed of conventional low-molecular-weight compounds. Knockdown by degradation of disease-causing peptides via the PSM system makes it possible to treat, ameliorate, or suppress progression of cancer.
ここでは一例として、NF-κBを疾患原因ペプチドとしたときの抗癌剤の構成について説明をする。NF-κBの活性阻害は、TNF-αによるアポトーシス誘導を促進することが知られている(Tang F., et al., 2002, Mol. Cell. Biol., 22: 8571-8579)。したがって、NF-κBに特異的に結合する結合因子を選択することで、本発明の疾患抑制剤はアポトーシス誘導剤として機能し得る。このアポトーシス誘導剤を癌細胞に適用することで、抗癌剤として機能し得る。 Here, as an example, we will explain the composition of an anticancer drug when NF-κB is used as a disease-causing peptide. Inhibition of NF-κB activity is known to promote apoptosis induction by TNF-α (Tang F., et al., 2002, Mol. Cell. Biol., 22: 8571-8579). Therefore, by selecting a binding factor that specifically binds to NF-κB, the disease suppressor of the present invention can function as an apoptosis inducer. By applying this apoptosis inducer to cancer cells, it can function as an anticancer agent.
NF-κBに結合する結合因子の具体的な構成については限定しない。NF-κBのクラスIに属するp50及びp52、及びクラスIIに属するp65(RelA)、c-Rel及びRelBのいずれにも結合できる構成であっても良いし、いずれかに特異的に結合する構成であっても良い。例えば、NF-κBが結合する核酸配列からなる核酸型結合因子が挙げられる。より具体的には、例えば、配列番号2又は3で示すNF-κB結合型ヘアピンDNAである。このNF-κB結合型ヘアピンDNAは、NF-κBの標的塩基配列を模した塩基配列を含むNF-κB Decoy DNAである。 The specific composition of the binding factor that binds to NF-κB is not limited. p50 and p52 belonging to class I of NF-κB and p65 (RelA), c-Rel and RelB belonging to class II may be configured to bind, or a configuration that specifically binds to any of them may be used. can be Examples thereof include nucleic acid-type binding agents consisting of a nucleic acid sequence to which NF-κB binds. More specifically, for example, the NF-κB-binding hairpin DNA represented by SEQ ID NO: 2 or 3. This NF-κB-binding hairpin DNA is NF-κB Decoy DNA containing a nucleotide sequence mimicking the target nucleotide sequence of NF-κB.
(2)アルツハイマー病抑制剤
本発明の疾患抑制剤は、アルツハイマー病(Alzheimer's disease:AD)の予防又はその進行抑制剤となり得る。この場合、疾患抑制剤はアルツハイマー病の原因となる疾患原因ペプチドに結合する結合因子を含む。アルツハイマー病は、βアミロイドタンパク質の脳内の神経細胞に凝集蓄積に始まり、微小管タンパク質であるタウタンパク質が過リン酸化されて線維化した後に、神経細胞が破壊され脳が萎縮することにより発症する。したがって、アルツハイマー病の疾患原因ペプチドとしてはβアミロイド、又はタウタンパク質の他、α-シヌクレイン等が該当し得る。したがって、βアミロイド、タウタンパク質又はα-シヌクレイン等に特異的に結合する結合因子を選択することで、本発明の疾患抑制剤はアルツハイマー病抑制剤として機能し得る。
(2) Alzheimer's Disease Suppressant The disease suppressor of the present invention can be an agent for preventing Alzheimer's disease (AD) or for suppressing its progress. In this case, the disease-suppressing agent comprises a binding agent that binds to a disease-causing peptide that causes Alzheimer's disease. Alzheimer's disease begins with the aggregation and accumulation of β-amyloid protein in neurons in the brain, followed by hyperphosphorylation of tau protein, a microtubule protein, resulting in fibrosis, followed by destruction of neurons and brain atrophy. . Therefore, β-amyloid, tau protein, α-synuclein, and the like can be applicable as disease-causing peptides for Alzheimer's disease. Therefore, by selecting a binding factor that specifically binds to β-amyloid, tau protein, α-synuclein, or the like, the disease inhibitor of the present invention can function as an Alzheimer's disease inhibitor.
βアミロイドやタウタンパク質に結合する結合因子の具体的な構成については限定しない。例えば、繊維化βアミロイド又はタウタンパク質へ結合能を有する低分子化合物等が挙げられる。より具体的な例として、βアミロイドの凝集体に対する結合因子としてはチオフラビンT(ThT)が、またタウタンパク質に対する結合因子としてはピリドインドールが該当する。 The specific composition of the binding factor that binds to β-amyloid and tau protein is not limited. Examples thereof include low-molecular-weight compounds capable of binding to fibrillar β-amyloid or tau protein. More specific examples include thioflavin T (ThT) as a binding factor for β-amyloid aggregates and pyridoindole as a binding factor for tau protein.
(3)パーキンソン病抑制剤
本発明の疾患抑制剤は、パーキンソン病(Parkinson's disease:PD)の予防又はその進行抑制剤となり得る。この場合、疾患抑制剤はパーキンソン病の原因となる疾患原因ペプチドに結合する結合因子を含む。パーキンソン病は、レビー小体(Lewy body)と呼ばれるタンパク質凝集体を構成するミスフォールドされたα-シヌクレイン(α-Synuclein)の多量化と蓄積による神経細胞死が発症に関与するとされている。したがって、パーキンソン病の疾患原因ペプチドとしてはミスフォールドした変異型α-シヌクレイン等が該当し得る。したがって、変異型α-シヌクレインに特異的に結合する結合因子を選択することで、本発明の疾患抑制剤はパーキンソン病抑制剤として機能し得る。
(3) Parkinson's Disease Suppressant The disease suppressor of the present invention can be a prophylactic agent for Parkinson's disease (PD) or an agent for suppressing the progression thereof. In this case, the disease-suppressing agent comprises a binding agent that binds to a disease-causing peptide that causes Parkinson's disease. The onset of Parkinson's disease is believed to be related to neuronal cell death due to multimerization and accumulation of misfolded α-synuclein, which constitutes protein aggregates called Lewy bodies. Therefore, misfolded mutant α-synuclein and the like can be applicable as disease-causing peptides for Parkinson's disease. Therefore, by selecting a binding factor that specifically binds to mutant α-synuclein, the disease suppressing agent of the present invention can function as a Parkinson's disease suppressing agent.
変異型α-シヌクレインに結合する結合因子の具体的な構成については限定しない。例えば、α-シヌクレインへ選択的に結合するβ-シヌクレインの部分配列のような短鎖ペプチド等が挙げられる。より具体的には、配列番号5で示されるアミノ酸配列(GVLYVGSKTR)からなるβsyn36が挙げられる(Shaltiel-Karyo R., et al., 2010, PLoS One, 5, e13863.; Fun X., et al., 2014, Nat. Neurosci., 17, 471-480.)。 The specific configuration of the binding factor that binds to mutant α-synuclein is not limited. Examples thereof include short peptides such as partial sequences of β-synuclein that selectively bind to α-synuclein. More specifically, βsyn36 consisting of the amino acid sequence (GVLYVGSKTR) shown in SEQ ID NO: 5 (Shaltiel-Karyo R., et al., 2010, PLoS One, 5, e13863.; Fun X., et al. ., 2014, Nat. Neurosci., 17, 471-480.).
(4)ハンチントン病抑制剤
本発明の疾患抑制剤は、ハンチントン病(Huntington's disease:HD)の予防又はその進行抑制剤となり得る。この場合、疾患抑制剤はハンチントン病の原因となる疾患原因ペプチドに結合する結合因子を含む。ハンチントン病は、CAGリピート数が伸長した変異ハンチンチン(mutated Huntingtin:mHTT)遺伝子の発現により生じる変異ハンチンチン(mHTT)が神経細胞の機能障害と細胞死を誘導し、発症に関与することが知られている(Tomoshige S., et al., 2017, Angew.Chem.Int. Ed., 56,11530-11533)。したがって、変異型ハンチンチンに特異的に結合する結合因子を選択することで、本発明の疾患抑制剤はハンチントン病抑制剤として機能し得る。
(4) Agent for suppressing Huntington's disease The agent for suppressing disease of the present invention can be an agent for preventing Huntington's disease (HD) or suppressing its progression. In this case, the disease-suppressing agent comprises a binding agent that binds to the disease-causing peptide responsible for Huntington's disease. Huntington's disease is known to be involved in the onset of mutated huntingtin (mHTT), which is caused by the expression of the mutated huntingtin (mHTT) gene with an expanded number of CAG repeats and induces neuronal dysfunction and cell death. (Tomoshige S., et al., 2017, Angew. Chem. Int. Ed., 56, 11530-11533). Therefore, by selecting a binding factor that specifically binds to mutant huntingtin, the disease-suppressing agent of the present invention can function as a Huntington's disease-suppressing agent.
変異型ハンチンチンに結合する結合因子の具体的な構成については限定しない。例えば、変異型ハンチンチンの多量体及び凝集体に結合する低分子化合物が挙げられる。より具体的には、例えば、ベンゾチアゾールアニリン(BTA)、又はフェニルジアゼニルベンゾチアゾール(phenyldiazenyl benzothiazole:PDB)(Tomoshige S., et al., 2017, Angew. Chem. Int. Ed., 56, 11530 -11533)等が該当する。 The specific composition of the binding factor that binds to mutant huntingtin is not limited. Examples include low-molecular-weight compounds that bind to mutant huntingtin multimers and aggregates. More specifically, for example, benzothiazole aniline (BTA) or phenyldiazenyl benzothiazole (PDB) (Tomoshige S., et al., 2017, Angew. Chem. Int. Ed., 56, 11530- 11533), etc.
本発明の標的ペプチド分解誘導剤について、以下で実施例を挙げて具体的に説明をする。ただし、ここで記載する構成等は、本発明の標的ペプチド分解誘導剤の具体例の一つに過ぎず、その範囲を限定するものではない。 The target peptide degradation inducer of the present invention will be specifically described below with reference to examples. However, the configuration and the like described here are only one specific example of the target peptide degradation inducer of the present invention, and do not limit the scope thereof.
<実施例1:核酸型標的ペプチド分解誘導剤の調製>
(目的)
本発明の標的ペプチド分解誘導剤を調製し、その標的ペプチド分解誘導剤によって標的ペプチドが捕捉されることを検証する。本実施例では、標的ペプチドとしてNF-κBを選択し、そのNF-κBと結合するNF-κB Decoy DNAを核酸型結合因子とする2種の標的ペプチド分解誘導剤(Ub-cys-Decoy、及びUb-C77-Decoy)を調製する。
<Example 1: Preparation of nucleic acid type target peptide degradation inducer>
(the purpose)
A target peptide degradation inducer of the present invention is prepared, and it is verified that the target peptide is captured by the target peptide degradation inducer. In this example, NF-κB was selected as the target peptide, and two target peptide degradation inducers (Ub-cys-Decoy and Ub-C77-Decoy) is prepared.
(方法)
(1)ユビキチン-システアミン(Ub-cys)の調製
本発明の標的ペプチド分解誘導剤を調製するために、まずユビキチンのC末端に、結合因子との連結に用いるシステアミンを付加した。250μMユビキチン、2μM E1、2mM ATP、5mMシステアミン、及び5mM TCEPを、100mM Tris-HCl(pH7.5)、250mMスクロース、50mM KCl、3mM MgCl2を含むバッファー1mL(最終容量)中で4℃にて一晩インキュベートした。続いて、反応混合物をSuperdexTM 75 Increase 10/300GLサイズ排除クロマトグラフィー(Cytiva社)で精製した。その後、目的のUb-cysを1.5カラム容量のSバッファー(20mM NaPO4、pH7.2、150mM NaCl)で溶出した。
(Method)
(1) Preparation of ubiquitin-cysteamine (Ub-cys) In order to prepare the target peptide degradation-inducing agent of the present invention, first, cysteamine used for linkage with a binding factor was added to the C-terminus of ubiquitin. 250 μM ubiquitin, 2 μM E1, 2 mM ATP, 5 mM cysteamine, and 5 mM TCEP in 1 mL (final volume) of buffer containing 100 mM Tris-HCl (pH 7.5), 250 mM sucrose, 50 mM KCl, 3 mM MgCl at 4 °C. Incubated overnight. The reaction mixture was subsequently purified by Superdex ™ 75 Increase 10/300GL size exclusion chromatography (Cytiva). After that, the desired Ub-cys was eluted with 1.5 column volumes of S buffer (20 mM NaPO 4 , pH 7.2, 150 mM NaCl).
(2)Ub-cysを用いたユビキチン-デコイ(Ub-cys-Decoy)コンジュゲートの調製
続いて、NF-κB結合因子であるNF-κB Decoy DNAの5'末端にSMCC(4-(N-マレイミドメチル)シクロヘキサンカルボン酸N-スクシンイミジル)を付加した後、前記(1)で調製したUb-システアミンと連結させて、本発明の標的ペプチド分解誘導剤であるUb-Decoyを調製した。
(2) Preparation of ubiquitin-decoy (Ub-cys-Decoy) conjugate using Ub-cys Subsequently, SMCC (4-(N- After addition of maleimidomethyl)cyclohexanecarboxylate (N-succinimidyl), it was linked to Ub-cysteamine prepared in (1) above to prepare Ub-Decoy, the target peptide degradation inducer of the present invention.
具体的には、まず、ジメチルホルムアミド(DMF)(100eq)中の3mM SMCC(Thermo Fisher Scientific社)を30μM NF-κBデコイDNA(1eq)を含むNaPO4バッファー(pH7.5)に添加した。使用したNF-κBデコイDNAは、以下の配列番号2又は3で示すポリヌクレオチド2種を用いた。
(i)NF-κB ODN1(27mer)
5'(amino)-AGGGAAATCCCAAAATGGGATTTCCCT-3'(FAM):配列番号2
(ii)NF-κB ODN2(28mer)
5'(amino)-AGGGGATTCCCAAAATGGGAATTCCCCT-3'(FAM):配列番号3
Specifically, first, 3 mM SMCC (Thermo Fisher Scientific) in dimethylformamide (DMF) (100 eq) was added to NaPO4 buffer (pH 7.5) containing 30 μM NF-κB decoy DNA (1 eq). As the NF-κB decoy DNA used, two types of polynucleotides shown in SEQ ID NO: 2 or 3 below were used.
(i) NF-κB ODN1 (27mer)
5′(amino)-AGGGAAATCCCAAAATGGGATTTCCCT-3′(FAM): SEQ ID NO: 2
(ii) NF-κB ODN2 (28mer)
5′(amino)-AGGGGATTCCCAAAATGGGAATTCCCCT-3′(FAM): SEQ ID NO: 3
上記NF-κB Decoy DNAは、いずれもヘアピン構造を形成し、NF-κBが認識、及び結合することが明らかとなっている。また、いずれも3'がFAM(Fluorescein)で標識されている。 All of the above NF-κB Decoy DNAs have been shown to form a hairpin structure and be recognized and bound by NF-κB. In addition, both are labeled with FAM (fluorescein) at the 3'.
前記反応混合物を室温で90分間インキュベートした後、残留SMCCをエタノール沈殿により除去し、生成物であるNF-κB Decoy DNA-SMCCを得た。 After incubating the reaction mixture at room temperature for 90 minutes, residual SMCC was removed by ethanol precipitation to obtain the product NF-κB Decoy DNA-SMCC.
続いて、前記(1)で調製した250μMのUb-システアミン(Ub-cys)(25eq)、10μMのNF-κB Decoy DNA-SMCC(1eq)、及び還元剤である10mM TCEP (富士フィルム和光純薬)を混合して、室温で一晩インキュベートして、ユビキチンとNF-κB Decoy DNAからなる本発明の標的ペプチド分解誘導剤であるUb-cys-Decoyを得た。 Subsequently, 250 μM Ub-cysteamine (Ub-cys) (25 eq) prepared in (1) above, 10 μM NF-κB Decoy DNA-SMCC (1 eq), and 10 mM TCEP (Fujifilm Wako Pure Chemical Industries, Ltd.) as a reducing agent ) and incubated overnight at room temperature to obtain Ub-cys-Decoy, a target peptide degradation inducer of the present invention consisting of ubiquitin and NF-κB Decoy DNA.
(3)Ub-C77を用いたユビキチン-デコイ(Ub-C77-Decoy)コンジュゲートの調製
ここでは、Ub-cysのようなユビキチンのC末端への官能基の付加に代えて、配列番号1で示すヒトユビキチンのC末端に77個目のアミノ酸としてシステインを付加したUb-C77、及び配列番号1で示すヒトユビキチンにおいて72位及び74位の2カ所のアルギニン残基がそれぞれアラニン残基及びスレオニン残基に置換した、配列番号6で示すユビキチン変異体のC末端に77個目のアミノ酸としてシステインを付加したUb-C77の変異体(UbR-C77)を用いて本発明の標的ペプチド分解誘導剤を調製した。
(3) Preparation of ubiquitin-decoy (Ub-C77-Decoy) conjugate using Ub-C77 Here, instead of adding a functional group to the C-terminus of ubiquitin such as Ub-cys, SEQ ID NO: 1 Ub-C77 in which cysteine is added as the 77th amino acid to the C-terminus of human ubiquitin shown, and two arginine residues at positions 72 and 74 in human ubiquitin shown in SEQ ID NO: 1 are alanine residues and threonine residues, respectively. The target peptide degradation inducer of the present invention was prepared using a Ub-C77 mutant (UbR-C77) in which cysteine was added as the 77th amino acid to the C-terminus of the ubiquitin mutant shown in SEQ ID NO: 6. prepared.
C末端にシステインを付加したUb-C77及びUbR-C77は、Ub-C77及びUbR-C77をコードする遺伝子を導入したpET-26b(+)プラスミドを用い、大腸菌を宿主として発現させることにより調製した。
NF-κB Decoy DNA-SMCCの調製は前記(2)の方法に準じた。
Ub-C77 and UbR-C77 with a cysteine added to the C-terminus were prepared by expression in E. coli as a host using a pET-26b(+) plasmid containing genes encoding Ub-C77 and UbR-C77. .
NF-κB Decoy DNA-SMCC was prepared according to the method (2) above.
2.12mM Ub-C77(50eq)、42.4μM NF-κB Decoy DNA-SMCC(1eq)、及び10mM TCEPを混合し、室温で一晩インキュベートした。次に、陰イオン交換クロマトグラフィーHiTrap Q HP(GE Healthcare社)によってUb-C77-Decoyを精製した。カラムは5カラム容量のバッファーA(50mM酢酸アンモニウム(pH 4.5)、0.1mM EDTA)で平衡化し、目的のUb-C77-Decoyは、NaClの段階的勾配(200 mM、450 mM、500mM)で溶出した。溶出液を濃縮し、Ub保存バッファー(25mM NaPO4(pH 7.2)、50mM NaCl)で透析した。収率は14.4%であった。 2.12 mM Ub-C77 (50 eq), 42.4 μM NF-κB Decoy DNA-SMCC (1 eq), and 10 mM TCEP were mixed and incubated overnight at room temperature. Next, Ub-C77-Decoy was purified by anion exchange chromatography HiTrap Q HP (GE Healthcare). The column was equilibrated with 5 column volumes of buffer A (50 mM ammonium acetate (pH 4.5), 0.1 mM EDTA) and the desired Ub-C77-Decoy was eluted with a stepwise gradient of NaCl (200 mM, 450 mM, 500 mM). bottom. The eluate was concentrated and dialyzed against Ub storage buffer (25 mM NaPO4 (pH 7.2), 50 mM NaCl). Yield was 14.4%.
145μM UbR-C77(20eq)、7.2μM NF-κB Decoy DNA-SMCC(1eq)を、20mM リン酸ナトリウム緩衝液(pH 7.0)中で混合し、室温で一晩インキュベートした。次に、SuperdexTM75 Increase 10/300GLサイズ排除クロマトグラフィーカラム(Cytiva)により精製した。その際、目的のUbR-C77-decoyは、1.5カラム容量のSバッファー(20mM リン酸ナトリウム、150 mM 塩化ナトリウム、pH 7.2)を用いて溶出した。 145 μM UbR-C77 (20 eq), 7.2 μM NF-κB Decoy DNA-SMCC (1 eq) were mixed in 20 mM sodium phosphate buffer (pH 7.0) and incubated overnight at room temperature. It was then purified by Superdex ™ 75 Increase 10/300GL size exclusion chromatography column (Cytiva). At that time, the target UbR-C77-decoy was eluted using 1.5 column volumes of S buffer (20 mM sodium phosphate, 150 mM sodium chloride, pH 7.2).
<実施例2: Ub-C77-DecoyとNF-κB p50の結合によるゲルシフトアッセイ>
(目的)
実施例1で調製した本発明の標的ペプチド分解誘導剤Ub-C77-Decoyが標的ペプチドNF-κB p50と結合することをゲルシフトアッセイにより確認する。
<Example 2: Gel shift assay by binding of Ub-C77-Decoy and NF-κB p50>
(the purpose)
It is confirmed by gel shift assay that the target peptide degradation inducer Ub-C77-Decoy of the present invention prepared in Example 1 binds to the target peptide NF-κB p50.
(方法)
500nM NF-κB p50(Cayman Chemical社)と25nM Ub-C77-Decoyを結合バッファー(10mM HEPES(pH7.9)、50mM KCl、0.1mM EDTA、2.5mM DTT、10%グリセロール、0.05%NP-40)中にて、室温で30分間インキュベートした。その後、サンプルを6%Native PAGEにアプライし、150V、0.5×TBE、4℃で25分間泳動した。対照用として、Ub-C77-Decoyのみを用いた。泳動後、NF-κBデコイDNAの3'末端に修飾したFAMの蛍光に基づいて、各サンプルの位置を確認した。
(Method)
500 nM NF-κB p50 (Cayman Chemical) and 25 nM Ub-C77-Decoy in binding buffer (10 mM HEPES (pH 7.9), 50 mM KCl, 0.1 mM EDTA, 2.5 mM DTT, 10% glycerol, 0.05% NP-40) Incubate at room temperature for 30 minutes. Samples were then applied to 6% Native PAGE and run at 150V, 0.5xTBE, 4°C for 25 minutes. For control, only Ub-C77-Decoy was used. After electrophoresis, the position of each sample was confirmed based on the fluorescence of FAM modified at the 3' end of NF-κB decoy DNA.
(結果)
結果を図1に示す。Ub-C77-Decoyのみのレーン1と比較して、Ub-C77-DecoyとNF-κB p50を混合したレーン2は、明らかにバンドシフトが起きており、Ub-C77-Decoyが標的ペプチドのNF-κB p50と結合することが確認できた。
なお、図示しないが、Ub-cys-Decoy、及びUbR-C77-decoyを用いた場合にも、同様の結合結果が得られた。
(result)
The results are shown in FIG. Compared to
Although not shown, similar binding results were obtained when Ub-cys-Decoy and UbR-C77-decoy were used.
<実施例3:標的ペプチドNF-κB p50分解アッセイ(1)>
(目的)
標的ペプチドが、プロテアソーム存在下で直接的なユビキチン化を介することなく、実施例1で調製した本発明の標的ペプチド分解誘導剤と結合することによって分解されることを確認する。
<Example 3: Target peptide NF-κB p50 degradation assay (1)>
(the purpose)
It is confirmed that the target peptide is degraded by binding to the target peptide degradation inducer of the present invention prepared in Example 1 without direct ubiquitination in the presence of proteasome.
(方法)
200nM NF-κB p50及び1μM Ub-C77-Decoyを、結合バッファー(25 mM HEPES (pH7.5)、100mM NaCl、1mM DTT、10%グリセロール、0.05% NP-40)中で25℃にて30分間インキュベートし、標的ペプチドであるNF-κB p50と本発明の標的ペプチド分解誘導剤であるUb-C77-Decoyが結合した複合体NF-κB p50/Ub-C77-Decoyを調製した。
(Method)
200 nM NF-κB p50 and 1 μM Ub-C77-Decoy in binding buffer (25 mM HEPES (pH 7.5), 100 mM NaCl, 1 mM DTT, 10% glycerol, 0.05% NP-40) for 30 minutes at 25°C. After incubation, a complex NF-κB p50/Ub-C77-Decoy in which the target peptide NF-κB p50 and the target peptide degradation inducer Ub-C77-Decoy of the present invention were bound was prepared.
次に、NF-κB p50/Ub-C77-Decoyに100nM 26Sプロテアソーム(東京都医学総合研究所 佐伯泰博士より恵贈)、2mM ATP、及び5mM MgCl2をに添加し、37℃で60分間インキュベートした。その間、インキュベート開始時の混合溶液を一部取り分けた。 Next, NF-κB p50/Ub-C77-Decoy was added with 100 nM 26S proteasome (gift from Dr. Yasushi Saeki, Tokyo Metropolitan Institute of Medical Science), 2 mM ATP, and 5 mM MgCl 2 and incubated at 37° C. for 60 minutes. . In the meantime, a portion of the mixed solution at the start of incubation was taken.
また、Ub-PSM系による分解であることを立証するために、プロテアソーム阻害剤である100μMのMG132(AdipoGen Life Sciences)を添加したサンプル、及び26Sプロテアソーム未添加のサンプルを調製した。さらに、対照用として、本発明の標的ペプチド分解誘導剤の構成因子であるNF-κB-Decoy DNAのみ、又はUb-C77のみを26Sプロテアソームに添加したサンプルも同時に調製した。 In addition, in order to prove that the degradation is due to the Ub-PSM system, a sample with 100 μM MG132 (AdipoGen Life Sciences), a proteasome inhibitor, and a sample without 26S proteasome were prepared. Furthermore, as controls, samples were also prepared in which only NF-κB-Decoy DNA or only Ub-C77, which is a component of the target peptide degradation inducer of the present invention, was added to the 26S proteasome.
続いて、反応後の混合溶液をSDSサンプルバッファー(62.5mM Tris-HCl(pH6.8), 2% SDS, 10% Glycerol, 5% 2-mercaptoethanol, 0.002% Bromophenol blue)と混合し、95℃で5分間加熱した後、SDS-PAGEで分離した。常法によりウェスタンブロッティングを行った後、得られたメンブレンを、ブロッキング溶液で1:500に希釈した一次抗体(Anti NFKB1, p105, p50-Specific:Proteintech社)と共に1.5時間撹拌しながらインキュベートした。1×TBST (20mM Tris-HCl pH7.6, 150mM NaCl, 0.2% Tween-20) で3回洗浄した後、ブロッキング溶液で1:1000に希釈した二次抗体(Anti-IgG, Mouse, Goat-Poly, HRP:R&D Systems社) と共に1時間緩やかに振盪しながらインキュベートし、1×TBSTで3回洗浄した。その後、メンブレンを基質/検出剤混合液(エンハンサー溶液:化学発光過酸化物試薬=1:1)と共に2分間インキュベートし、化学発光イメージングシステム(ChemiDoc XRS Plus:BIO-RAD社)で分析した。 Subsequently, the mixed solution after reaction was mixed with SDS sample buffer (62.5mM Tris-HCl (pH6.8), 2% SDS, 10% Glycerol, 5% 2-mercaptoethanol, 0.002% Bromophenol blue) and After heating for 5 minutes, they were separated by SDS-PAGE. After Western blotting by a standard method, the resulting membrane was incubated with primary antibodies (Anti NFKB1, p105, p50-Specific: Proteintech) diluted 1:500 in blocking solution for 1.5 hours with agitation. After washing three times with 1×TBST (20 mM Tris-HCl pH 7.6, 150 mM NaCl, 0.2% Tween-20), secondary antibodies (Anti-IgG, Mouse, Goat-Poly , HRP: R&D Systems) for 1 hour with gentle shaking, and washed 3 times with 1×TBST. The membrane was then incubated with a substrate/detector mixture (enhancer solution: chemiluminescent peroxide reagent = 1:1) for 2 minutes and analyzed with a chemiluminescent imaging system (ChemiDoc XRS Plus: BIO-RAD).
(結果)
結果を図2に示す。NF-κB p50/Ub-C77-Decoyと26Sプロテアソームとを混合した60分後のレーン2のみでNF-κB p50の顕著な減少が確認された。一方、NF-κB p50/Ub-C77-Decoyと26Sプロテアソームとの混合物に、さらにMG132を添加した場合には、NF-κB p50の顕著な減少はほとんど認められなかった(レーン3)。さらに、NF-κB p50/Ub-C77-Decoy単独の場合(レーン4)、Decoy-DNAと26Sプロテアソームとを混合した場合(レーン5)及びUb-C77と26Sプロテアソームとを混合した場合(レーン6)もNF-κB p50の顕著な減少はほとんど認められなかった。
(result)
The results are shown in FIG. A marked decrease in NF-κB p50 was confirmed only in
これらの結果から、本発明の標的ペプチド分解誘導剤は、標的ペプチドに結合することによって間接的に標的ペプチドをユビキチン修飾し、従来のUb-PSM系と同様にプロテアソームを利用して標的ペプチドを分解できることが立証された。
なお、図示しないが、Ub-cys-Decoyを用いた場合にも、同様の分解結果が得られた。
Based on these results, the target peptide degradation inducer of the present invention indirectly ubiquitinates the target peptide by binding to the target peptide, and degrades the target peptide using the proteasome as in the conventional Ub-PSM system. proved to be possible.
Although not shown, similar decomposition results were obtained when Ub-cys-Decoy was used.
<実施例4: NF-κBp50分解率の検証>
(目的)
本発明の標的ペプチド分解誘導剤による標的ペプチドの分解率を検証する。
<Example 4: Verification of NF-κB p50 decomposition rate>
(the purpose)
The target peptide degradation rate by the target peptide degradation inducer of the present invention is verified.
(方法)
基本操作は、実施例3の方法に準じた。本実施例では、標的ペプチド分解誘導剤であるUb-C77-Decoyの濃度を50nM、100nM、200nM、400nM、800nM、及び1000nM(1μM)で振り分け、反応後の発光強度に基づいて、各濃度におけるNF-κB p50の残存量を測定した。残存量は、Ub-C77-Decoy未添加(0nM)の時の発光強度を100%としたときの相対値、すなわちUb-C77-Decoyの残存率で算出した。
(Method)
The basic operation conformed to the method of Example 3. In this example, the concentration of Ub-C77-Decoy, which is a target peptide degradation inducer, was divided into 50 nM, 100 nM, 200 nM, 400 nM, 800 nM, and 1000 nM (1 μM), and based on the luminescence intensity after the reaction, at each concentration The residual amount of NF-κB p50 was measured. The residual amount was calculated as a relative value when the emission intensity when Ub-C77-Decoy was not added (0 nM) was taken as 100%, that is, the residual rate of Ub-C77-Decoy.
(結果)
結果を図3に示す。反応前の添加量に対する反応60分後のNF-κB p50残留率は、Ub-C77-Decoyの添加量依存的に減少し、1000nM Ub-C77-Decoyでは32%であった。つまり、標的ペプチドが本発明の標的ペプチド分解誘導剤によって約70%の分解率で分解されたことを示唆している。
(result)
The results are shown in FIG. The NF-κB p50 retention rate after 60 minutes of reaction relative to the amount added before reaction decreased depending on the amount of Ub-C77-Decoy added, and was 32% at 1000 nM Ub-C77-Decoy. In other words, it suggests that the target peptide was degraded by the target peptide degradation inducer of the present invention at a degradation rate of about 70%.
一方、標的タンパク質への直接的なユビキチン化を介したUb-PSM系による分解の場合、反応1時間後の標的ペプチドα-グロブリンの残留率は、Sunら(Sun H., et al., 2019, Proc. Natl. Acad. Sci. USA., 116: 7805-7812)の図6によれば47%である。つまり、直接的なユビキチン化を介したUb-PSM系では、標的ペプチドは約60%の分解率で分解されたことを示唆している。 On the other hand, in the case of degradation by the Ub-PSM system via direct ubiquitination to the target protein, the retention rate of the target peptide α-globulin after 1 hour of reaction was determined by Sun et al. (Sun H., et al., 2019 , Proc. Natl. Acad. Sci. USA., 116: 7805-7812), 47%. This suggests that the direct ubiquitination-mediated Ub-PSM system degraded the target peptide with a degradation rate of approximately 60%.
したがって、標的ペプチドの種類は異なるものの、本発明の標的ペプチド分解誘導剤の結合による標的ペプチドの間接的なユビキチン化は、プロテアソームによる効率的な分解を媒介するシグナルとして従来の直接的なユビキチン化と同程度の分解率で標的ペプチドを分解できることが明らかとなった。 Therefore, although the types of target peptides are different, indirect ubiquitination of target peptides by binding of the target peptide degradation-inducing agent of the present invention can be used as a signal to mediate efficient degradation by the proteasome compared to conventional direct ubiquitination. It was clarified that the target peptide could be decomposed with a similar degree of decomposition rate.
<実施例5:NF-κB p50の分解アッセイ(2)>
(目的)
ユビキチンに変異導入したUbR-C77-Decoyにおける標的ペプチド分解誘導能について検証する。
<Example 5: NF-κB p50 degradation assay (2)>
(the purpose)
We will verify the ability of UbR-C77-Decoy with mutated ubiquitin to induce target peptide degradation.
(方法)
基本操作は実施例3に準じた。本実施例では、UbR-C77-decoyを終濃度1μMでNF-κB p50と混合し、NF-κB p50の残存量をウェスタンブロッティング法により評価した。残存量は、Ub-C77-Decoy未添加(0nM)の時のバンド強度を100%としたときの相対値で算出した。ポジティブコントロールには、終濃度1μMのUb-C77-decoyを用いた。
(Method)
The basic operation conformed to Example 3. In this example, UbR-C77-decoy was mixed with NF-κB p50 at a final concentration of 1 μM, and the residual amount of NF-κB p50 was evaluated by Western blotting. The residual amount was calculated as a relative value when the band intensity when Ub-C77-Decoy was not added (0 nM) was taken as 100%. Ub-C77-decoy with a final concentration of 1 μM was used as a positive control.
(結果)
結果を図4に示す。ペプチド分解誘導剤未添加時のNF-κB p50の残存量(レーン1)に対するUbR-C77-decoy(レーン2)、及びUb-C77-decoy(レーン3)の添加時のNF-κB p50の残存量は、それぞれ34%、及び25%であった。一方、Decoy DNA(レーン4)、UbR-C77(レーン5)、Ub-C77(レーン6)をそれぞれ単独で混合した場合には、NF-κB p50残存量の顕著な低下は見られなかった。以上から、UbR-C77-decoyは、Ub-C77-decoyと同等の分解誘導能を有することが実証された。
(result)
The results are shown in FIG. Residual NF-κB p50 after addition of UbR-C77-decoy (lane 2) and Ub-C77-decoy (lane 3) relative to residual NF-κB p50 (lane 1) when peptide degradation inducer was not added The amounts were 34% and 25% respectively. On the other hand, when Decoy DNA (lane 4), UbR-C77 (lane 5), and Ub-C77 (lane 6) were individually mixed, no significant decrease in residual NF-κB p50 was observed. From the above, it was demonstrated that UbR-C77-decoy has a degradation-inducing ability equivalent to that of Ub-C77-decoy.
<実施例6:細胞抽出液中におけるNF-κB p65の分解アッセイ>
(目的)
細胞抽出液中に含まれる細胞由来の標的ペプチドNF-κB p65について、UbR-C77-Decoyの間接的ユビキチン化による分解を検証する。
<Example 6: NF-κB p65 degradation assay in cell extract>
(the purpose)
Degradation of the cell-derived target peptide NF-κB p65 contained in the cell extract by indirect ubiquitination of UbR-C77-Decoy is verified.
(方法)
最終タンパク質濃度を0.7mg/mLに調整したMDA-MB-231細胞(ATCC)の細胞抽出液、終濃度1μMのUbR-C77-decoy、終濃度0.1μMの26S酵母プロテアソーム(東京都医学総合研究所 佐伯泰博士より恵贈)を分解アッセイ用緩衝液(5mM HEPES、10mM NaCl、4mM TCEP、10% グリセロール、0.01% NP-40)中で混合し、室温で6時間静置した。各サンプルにおけるNF-κB p65の残存量は定法によりウェスタンブロッティング法により分析した。残存量はUbR-C77-decoy非添加時のNF-κB p65のバンド強度を100%とした時の相対値として求めた。その際、NF-κB p65のバンド強度は、β-actinのバンド強度によって正規化した。
(Method)
Cell extract of MDA-MB-231 cells (ATCC) adjusted to a final protein concentration of 0.7 mg/mL, UbR-C77-decoy at a final concentration of 1 μM, 26S yeast proteasome at a final concentration of 0.1 μM (Tokyo Metropolitan Institute of Medical Science) A gift from Dr. Yasushi Saeki) was mixed in a degradation assay buffer (5 mM HEPES, 10 mM NaCl, 4 mM TCEP, 10% glycerol, 0.01% NP-40) and allowed to stand at room temperature for 6 hours. The amount of residual NF-κB p65 in each sample was analyzed by Western blotting according to a standard method. The residual amount was obtained as a relative value when the band intensity of NF-κB p65 when UbR-C77-decoy was not added was taken as 100%. At that time, the band intensity of NF-κB p65 was normalized by the band intensity of β-actin.
(結果)
結果を図5に示す。UbR-C77-decoyの存在下で、NF-κB p65の残存量の顕著な減少が認められ、その分解率は59%に達していた。この結果から、本発明の標的ペプチド分解誘導剤を用いた間接的ユビキチン化による標的ペプチドの分解誘導は、細胞抽出液中でも、その効果を奏し得ることが実証された。
(result)
The results are shown in FIG. In the presence of UbR-C77-decoy, a marked decrease in the residual amount of NF-κB p65 was observed, and its degradation rate reached 59%. These results demonstrate that the induction of target peptide degradation by indirect ubiquitination using the target peptide degradation inducer of the present invention can be effective even in cell extracts.
<実施例7:生細胞中における標的ペプチドNF-κB p65の分解アッセイ>
(目的)
生細胞中でのUbR-C77-DecoyによるNF-κB p65の分解誘導を検証する。
(方法)
MCF-7細胞(理化学研究所バイオリソース研究センター)及びHeLa細胞(理化学研究所バイオリソース研究センター)を48wellプレートに3×104 cells/wellで播種し、5% CO2下、37℃で24時間培養して80%コンフルエンシーまで増殖させた。次に、培地をOpti-MEMに交換後、LipofectamineTM LTX(Thermo Fisher Scientific社)を用いたリポフェクションにより1μMのUbR-C77-decoyを細胞に導入した。その後、培地をDMEM(10% FBS, 0.5% PS)に交換し、TNF-α(終濃度 20ng/mL)及びタンパク質翻訳素材剤であるシクロヘキシミド(CHX)/0.5% DMSO(終濃度 50μg/mL)を培地に添加した後、5% CO2下、37℃で4時間培養した。培養後、細胞をRIPAバッファー(Nacalai tesque)で溶解し、ウェスタンブロッティングによりNF-κB p65の分析を行った。その際、NF-κB p65のバンド強度は、β-actinのバンド強度によって正規化した。
<Example 7: Target peptide NF-κB p65 degradation assay in living cells>
(the purpose)
To verify the degradation induction of NF-κB p65 by UbR-C77-Decoy in living cells.
(Method)
MCF-7 cells (RIKEN BioResource Research Center) and HeLa cells (RIKEN BioResource Research Center) were plated at 3×10 4 cells/well in a 48-well plate and cultured at 37°C under 5% CO 2 for 24 hours. and grown to 80% confluency. Next, after replacing the medium with Opti-MEM, 1 μM UbR-C77-decoy was introduced into the cells by lipofection using Lipofectamine ™ LTX (Thermo Fisher Scientific). After that, the medium was replaced with DMEM (10% FBS, 0.5% PS), and TNF-α (final concentration 20ng/mL) and protein translation material cycloheximide (CHX)/0.5% DMSO (final concentration 50μg/mL) were added. was added to the medium and cultured at 37°C under 5% CO 2 for 4 hours. After culturing, cells were lysed with RIPA buffer (Nacalai tesque) and analyzed for NF-κB p65 by Western blotting. At that time, the band intensity of NF-κB p65 was normalized by the band intensity of β-actin.
(結果)
結果を図6に示す。NF-κB p65のβ-actinに対する相対量は、UbR-C77-decoyを導入した場合、HeLa細胞、及びMCF-7細胞において、それぞれ34%、及び29%となり、いずれの細胞でも30%程度にまで減少することが明らかとなった。この結果から、本発明の標的ペプチド分解誘導剤は、生細胞中においても標的ペプチドをUb-PSM系を介した分解へ誘導し得ることが実証された。
(result)
The results are shown in FIG. When UbR-C77-decoy was introduced, the relative amount of NF-κB p65 to β-actin was 34% and 29% in HeLa cells and MCF-7 cells, respectively, and decreased to about 30% in both cells. was found to decrease to These results demonstrate that the target peptide degradation inducer of the present invention can induce the target peptide to be degraded via the Ub-PSM system even in living cells.
<実施例8: Ub-C77-Decoyを用いたアポトーシス誘導>
(目的)
NF-κBの活性阻害は、前述のようにTNF-αによるアポトーシス誘導を促進する。そこで、本発明の標的ペプチド分解誘導剤であるUb-C77-DecoyによるNF-κBの分解によりアポトーシスが誘導されることを検証する。
<Example 8: Apoptosis induction using Ub-C77-Decoy>
(the purpose)
Inhibition of NF-κB activity promotes apoptosis induction by TNF-α as described above. Therefore, it is verified that apoptosis is induced by degradation of NF-κB by Ub-C77-Decoy, which is the target peptide degradation inducer of the present invention.
(方法)
(1)細胞培養
ヒト乳癌細胞株MCF-7細胞を10%FBS及び0.5%ペニシリン/ストレプトマイシンを含むDMEM(以下「DMEM(FBS/PS)」と表記する)を入れた10cmディッシュで、5%CO2下、37℃で前培養した。
(Method)
(1) Cell culture Human breast cancer cell line MCF-7 cells were placed in DMEM containing 10% FBS and 0.5% penicillin/streptomycin (hereinafter referred to as "DMEM (FBS/PS)") in a 10 cm dish and incubated with 5% CO. It was pre-incubated at 37°C under 2 .
(2)Ub-C77-Decoyのリポフェクションによる細胞内導入の確認
前培養後のMCF-7細胞を35mmガラス底ディッシュにφ=12 mm(5×104cells/dish)(IWAKI社)で播種し、5%CO2下、37℃で24時間インキュベートした。その後、培地をOpti-MEMに交換した後、Lipofectamine(登録商標) LTX with Plus Reagent(Thermo Fisher Scientific社)を用いて、Ub-C77-DecoyをMCF-7細胞に一過的にトランスフェクトした。対照用にはNF-κB Decoy DNAを用いた。導入後の細胞を5%CO2下で、37℃にて2時間インキュベートした後、Hoechst33258で染色し、5%CO2下で37℃にて再度2時間インキュベートした後、蛍光顕微鏡(IX83:OLYMPUS)にて核染色した細胞を観察した。
(2) Confirmation of intracellular introduction of Ub-C77-Decoy by lipofection Pre-cultured MCF-7 cells were plated on a 35 mm glass bottom dish at φ = 12 mm (5 × 10 4 cells/dish) (IWAKI). , incubated for 24 h at 37 °
(3)細胞生存率アッセイ
基本的操作は上記(2)に記載の方法に準じた。前培養後のMCF-7細胞をDMEM(FBS/PS)で2.0×104 cells/200μLに調製した。次に、96ウェルプレートに200μL/wellで播種し、5%CO2下、37℃にて約80%のコンフルエンスまで24時間以内で培養した。続いて、培地をOpti-MEMに交換した後、Lipofectamine(登録商標)LTX with Plus Reagent(Thermo Fisher Scientific社)を用いて、200nM Ub-C77-DecoyをMCF-7細胞に一過的にトランスフェクトした。陰性対照用には200nM NF-κB Decoy DNAを用いた。導入後、細胞を5%CO2下で37℃にて1.5時間インキュベートした。その後、培地をDMEM(FBS/PS)に交換し、5ng/mLのTNF-αを細胞懸濁液に添加し、5%CO2下で37℃にて6時間又は24時間インキュベートした。PrestoBlueTM Cell Viability Reagent(Thermo Fisher Scientific社)を添加し、5%CO2下で37℃にて30分間インキュベートして染色し、プレートリーダー(Cytation5:BioTek)を用いて細胞生存率を測定した。
(3) Cell Viability Assay The basic procedure followed the method described in (2) above. MCF-7 cells after preculture were adjusted to 2.0×10 4 cells/200 μL with DMEM (FBS/PS). Next, the cells were seeded in 96-well plates at 200 μL/well and cultured at 37° C. under 5% CO 2 within 24 hours to approximately 80% confluence. Subsequently, after replacing the medium with Opti-MEM, MCF-7 cells were transiently transfected with 200 nM Ub-C77-Decoy using Lipofectamine® LTX with Plus Reagent (Thermo Fisher Scientific). bottom. 200 nM NF-κB Decoy DNA was used for negative control. After transduction, cells were incubated for 1.5 hours at 37° C. under 5% CO 2 . After that, the medium was changed to DMEM (FBS/PS) and 5 ng/mL TNF-α was added to the cell suspension and incubated at 37° C. under 5% CO 2 for 6 hours or 24 hours. PrestoBlue ™ Cell Viability Reagent (Thermo Fisher Scientific) was added, incubated for 30 minutes at 37°C under 5% CO 2 for staining, and cell viability was measured using a plate reader (Cytation5: BioTek).
(結果)
図7に顕微鏡画像を、また図8に細胞生存率を示す。
図7より、Ub-C77-Decoyをリポフェクションにより導入した細胞内からは、Ub-C77-Decoyに修飾したFAMに由来する蛍光が観察された(f)。また、f画像は、位相差顕微鏡画像(i)との重ね合わせから、リポフェクションでUb-C77-Decoyの導入を行った細胞群の大部分で蛍光が観察されることが示された(j)。一方、対照のUb-C77-Decoyを直接培地に添加した場合では、蛍光が観察された細胞はほとんどみられなかった(a、d、及びe)。以上の結果は、一般的なリポフェクションにより、Ub-C77-Decoyを高い効率で細胞内に導入可能であることを示唆している。
(result)
Microscopic images are shown in FIG. 7, and cell viability is shown in FIG.
From FIG. 7, fluorescence derived from FAM modified with Ub-C77-Decoy was observed from within the cells into which Ub-C77-Decoy was introduced by lipofection (f). In addition, the f image was superimposed on the phase-contrast microscope image (i), and showed that fluorescence was observed in most of the cell groups into which Ub-C77-Decoy was introduced by lipofection (j). . On the other hand, when the control Ub-C77-Decoy was directly added to the medium, few cells exhibited fluorescence (a, d, and e). The above results suggest that Ub-C77-Decoy can be introduced into cells with high efficiency by general lipofection.
なお、核染色に用いたHoechst33258に由来する蛍光図(b及びg)とFAMに由来する蛍光(a及びf)を重ね合わせた画像(c及びh)から、リポフェクションによりUb-C77-Decoyを導入した細胞では一部、細胞内だけでなく、核内へも移行していることも示唆された(c)。 Ub-C77-Decoy was introduced by lipofection from images (c and h) in which the fluorescence diagrams (b and g) derived from Hoechst33258 used for nuclear staining and the fluorescence (a and f) derived from FAM were superimposed. It was also suggested that some of the translocated cells translocated not only into the cell but also into the nucleus (c).
図8より、無添加のblankにおける細胞生存率を1としたときの相対生存率は、陰性対照のNF-κB Decoy DNA(図中のDecoy DNA)が0.81であるのに対して、Ub-C77-Decoyは0.64であった。これらの結果は、本発明の標的ペプチド分解誘導剤Ub-C77-Decoyが標的ペプチドであるNF-κBを分解した結果、TNF-α存在下でアポトーシスが誘導され、細胞生存率の低下したことを示唆している。 From FIG. 8, the relative survival rate when the cell survival rate in the unadded blank is 1 is 0.81 for the negative control NF-κB Decoy DNA (Decoy DNA in the figure), whereas Ub-C77 -Decoy was 0.64. These results indicate that the target peptide degradation inducer Ub-C77-Decoy of the present invention degraded the target peptide NF-κB, resulting in the induction of apoptosis in the presence of TNF-α and a decrease in cell viability. suggesting.
<実施例9:低分子化合物型標的ペプチド分解誘導剤の調製>
(目的)
低分子化合物であるビオチンを結合因子として用い、本発明の標的ペプチド分解誘導剤Ub-cys-biotinを調製する。
<Example 9: Preparation of low-molecular compound-type target peptide degradation inducer>
(the purpose)
The target peptide degradation inducer Ub-cys-biotin of the present invention is prepared using biotin, which is a low-molecular-weight compound, as a binding factor.
(方法)
(1)カルボニルアクリルビオチン(Carbonyl acrylic biotin)の調製
低分子化合物型結合因子として低分子リガンドであるビオチンを用いた。ユビキチンへのビオチンの付加は、Bernardimら(Nat. Commun. 2016, 7, 13128.)のマイケル付加反応に準じて行った。まず、ビオチン(富士フィルム和光純薬社)と3-ベンゾイルアクリル酸(Alfa Aesar社)を、1,2-エチレンジアミン(N-Boc ethylendiamine : Sigma-aldrich社)を介して縮合したカルボニルアクリルビオチンの合成を行った。N-ヒドロキシスクシンイミド(NHS)によって活性エステル化した3-ベンゾイルアクリル酸に、N-Boc ethylendiamineをジクロロメタン中ジイソプロピルエチルアミン(DIPEA)存在下で室温にて作用させて、縮合した。その後、Boc基を20(v/v)%トリフルオロ酢酸を含むジクロロメタン溶液中で脱保護した。続いて、得られた中間体を、NHSにより活性エステル化したビオチンとジクロロメタン中DIPEA存在下で室温にて反応させることで、目的のCarbonyl acrylic biotinを得た。
(Method)
(1) Preparation of carbonyl acrylic biotin A low-molecular-weight ligand, biotin, was used as a low-molecular-weight compound-type binding agent. Biotin was added to ubiquitin according to the Michael addition reaction of Bernardim et al. (Nat. Commun. 2016, 7, 13128.). First, biotin (Fujifilm Wako Pure Chemical Industries, Ltd.) and 3-benzoylacrylic acid (Alfa Aesar) were condensed via 1,2-ethylenediamine (N-Boc ethylendiamine: Sigma-Aldrich) to synthesize carbonylacrylic biotin. did 3-Benzoylacrylic acid activated by N-hydroxysuccinimide (NHS) was condensed with N-Boc ethylendiamine in the presence of diisopropylethylamine (DIPEA) in dichloromethane at room temperature. After that, the Boc group was deprotected in a dichloromethane solution containing 20(v/v)% trifluoroacetic acid. Subsequently, the obtained intermediate was reacted with NHS-activated biotin in the presence of DIPEA in dichloromethane at room temperature to obtain the desired Carbonyl acrylic biotin.
(2)Ub-Cysを用いたユビキチン-ビオチン(Ub-cys-biotin)コンジュゲートの調製
Ubへのシステアミンの付加は、実施例1(1)に記載の方法に準じた。Ub-cys-biotinは、Ub-cysと前記(1)で作製したカルボニルアクリルビオチン間のマイケル付加反応により調製した。具体的には、187μM Ub-Cysと938μM 3-カルボニルアクリルビオチンを20mM Tris-HCl(pH8.0)中で混合し、25℃で3時間反応させた。その後、目的物をSuperdexTM 75 Increase 10/300GLサイズ排除クロマトグラフィー(Cytiva社)により精製した。目的のUb-cys-biotinは、1.5カラム容量のTrisバッファー(20mM Tris-HCl、150mM NaCl、pH8.0)を用いて溶出した。
(2) Preparation of ubiquitin-biotin (Ub-cys-biotin) conjugate using Ub-Cys Addition of cysteamine to Ub followed the method described in Example 1(1). Ub-cys-biotin was prepared by Michael addition reaction between Ub-cys and carbonyl acryl biotin prepared in (1) above. Specifically, 187 μM Ub-Cys and 938 μM 3-carbonylacrylbiotin were mixed in 20 mM Tris-HCl (pH 8.0) and reacted at 25° C. for 3 hours. The desired product was then purified by Superdex ™ 75 Increase 10/300GL size exclusion chromatography (Cytiva). The desired Ub-cys-biotin was eluted using 1.5 column volumes of Tris buffer (20 mM Tris-HCl, 150 mM NaCl, pH 8.0).
<実施例10:低分子化合物型標的ペプチド分解誘導剤を用いた標的ペプチドの分解アッセイ>
(目的)
実施例9で調製した低分子化合物型標的ペプチド分解誘導剤(Ub-cys-biotin)による標的ペプチド(ストレプトアビジン)の分解誘導を検証する。
<Example 10: Target Peptide Degradation Assay Using Low-Molecular-Type Target Peptide Degradation Inducing Agent>
(the purpose)
The degradation induction of the target peptide (streptavidin) by the low-molecular compound-type target peptide degradation inducer (Ub-cys-biotin) prepared in Example 9 is verified.
(方法)
2μM ストレプトアビジン(富士フィルム和光純薬社)、2μMのUb-cys-biotinをTris-HCl緩衝液(50mM Tris-HCl(pH7.6)、100mM NaCl, 10mM MgCl2, 2mM ATP、5mM DTT、10% Glycerol)中で混合し、37℃で30分間インキュベートした。その後、0.1μMの26S酵母プロテアソームを添加し、37℃で1時間静置した。各サンプルはSDS-PAGEによりサイズ分画を行い、ストレプトアビジンの残存量はCBB染色により分析を行った。
(Method)
2 μM streptavidin (Fujifilm Wako Pure Chemical Industries, Ltd.), 2 μM Ub-cys-biotin in Tris-HCl buffer (50 mM Tris-HCl (pH 7.6), 100 mM NaCl, 10 mM MgCl 2 , 2 mM ATP, 5 mM DTT, 10 mM % Glycerol) and incubated at 37° C. for 30 minutes. After that, 0.1 µM of 26S yeast proteasome was added, and the mixture was allowed to stand at 37°C for 1 hour. Each sample was subjected to size fractionation by SDS-PAGE, and the residual amount of streptavidin was analyzed by CBB staining.
(結果)
結果を図9に示す。Ub-cys-biotin未添加の対照(Control)でのストレプトアビジン量を100%としたときのUb-cys-biotin添加時(Ub-cys-biotin)のストレプトアビジン量の相対残存量は61%であった。これは、ストレプトアビジンがUb-PSM系を介してUb-cys-biotinにより39%分解されたことを示唆する。
以上の結果から、結合因子として低分子化合物を用いた場合も、本願発明の標的ペプチド分解誘導剤は、標的ペプチドの分解を誘導できることが実証された。
(result)
The results are shown in FIG. The relative residual amount of streptavidin when Ub-cys-biotin was added (Ub-cys-biotin) was 61% when the amount of streptavidin in the control without Ub-cys-biotin was set to 100%. there were. This suggests that streptavidin was 39% degraded by Ub-cys-biotin via the Ub-PSM system.
From the above results, it was demonstrated that the target peptide degradation-inducing agent of the present invention can induce degradation of the target peptide even when a low-molecular-weight compound is used as the binding factor.
<実施例11:ペプチド型標的ペプチド分解誘導剤の調製>
(目的)
ペプチドリガンドを結合因子として用いた、本発明の標的ペプチド分解誘導剤を調製する。
<Example 11: Preparation of peptide-type target peptide degradation inducer>
(the purpose)
A target peptide degradation inducer of the present invention is prepared using a peptide ligand as a binding agent.
(方法)
(1)直鎖状マルチユビキチンとペプチドリガンドのコンジュゲートの調製
結合因子としてペプチド配列を用いた本発明の標的ペプチド分解誘導剤Ub3-B6及びUb4-B6の調製を行った。
直鎖状マルチユビキチンには、配列番号1で示すアミノ酸配列においてR72A及びR74Tの2ヶ所の変異を有する変異ユビキチン(UbR)が、それぞれ配列番号9で示す6アミノ酸残基(GSGGGG)からなるリンカーペプチドを介して直鎖状に3つ又は4つ連結したUbR3又はUbR4を用いた。
また、結合因子であるリガンドペプチドには、抗アポトーシスタンパク質であるMcl-1に対する選択的結合ペプチドであり、配列番号10で示すアミノ酸配列(SYDDLALMLRSIGDSL)からなるB6ペプチド(Journal of Biological Chemistry, 2009, 284, 31315-31326.)を用いた。
目的のペプチド型標的ペプチド分解誘導剤であるUbR3-B6又はUbR4-B6は、UbR3又はUbR4を前記ペプチドリガンドB6と前記リンカーペプチドを介して連結した構成を有する。UbR3-B6又はUbR4-B6の調製は、それぞれの全長ペプチドをコードした遺伝子をpET15bプラスミドに導入した発現ベクターを大腸菌に導入して発現させることによって調製した。
(Method)
(1) Preparation of Conjugates of Linear Multiubiquitin and Peptide Ligand Targeted peptide degradation inducers Ub 3 -B6 and Ub 4 -B6 of the present invention were prepared using peptide sequences as binding factors.
In the linear multi-ubiquitin, mutant ubiquitin (UbR) having two mutations of R72A and R74T in the amino acid sequence shown in SEQ ID NO: 1 has a linker peptide consisting of 6 amino acid residues (GSGGGG) shown in SEQ ID NO: 9. UbR 3 or UbR 4 in which 3 or 4 are linked in a straight chain via is used.
The ligand peptide, which is a binding factor, is a selective binding peptide to Mcl-1, which is an anti-apoptotic protein, and B6 peptide (Journal of Biological Chemistry, 2009, 284 , 31315-31326.) was used.
UbR 3 -B6 or UbR 4 -B6, which is the target peptide-type target peptide degradation inducer of interest, has a structure in which UbR 3 or UbR 4 is linked to the peptide ligand B6 via the linker peptide. UbR 3 -B6 or UbR 4 -B6 was prepared by introducing an expression vector in which a gene encoding each full-length peptide was introduced into a pET15b plasmid into Escherichia coli and expressing it.
<実施例12:ペプチド型標的ペプチド分解誘導剤を用いた標的ペプチドの分解アッセイ>
(目的)
実施例11で調製したペプチド型標的ペプチド分解誘導剤(UbR3-B6及びUbR4-B6)による標的ペプチド(Mcl-1)の分解誘導を検証する。
<Example 12: Target Peptide Degradation Assay Using Peptide-Type Target Peptide Degradation Inducing Agent>
(the purpose)
Degradation induction of the target peptide (Mcl-1) by the peptide-type target peptide degradation inducers (UbR 3 -B6 and UbR 4 -B6) prepared in Example 11 is verified.
(方法)
結合因子としてペプチド配列を用いた標的ペプチド分解誘導剤の開発と検証する。
2μMのUbR3-B6及びUbR4-B6を0.25μM 組換えMcl-1(Novus Biologicals社)とTris-HCl緩衝液(50mM Tris-HCl(pH 7.5), 100mM NaCl, 10mM MgCl2, 5mM DTT, 2mM ATP, 10% Glycerol)中で混合し、30分間インキュベートした。その後、終濃度0.1mg/mLの26S ヒトプロテアソーム(LifeSensors社)と混合し、2時間反応させた。各サンプルについてSDS-PAGEによりサイズ分画をしたのち、抗Mcl-1抗体(Cell Signaling Technology社)を1次抗体として、ウェスタンブロッティングにより分析を行った。
(Method)
Development and validation of targeted peptide degradation inducers using peptide sequences as binding factors.
2 μM UbR3-B6 and UbR4-B6, 0.25 μM recombinant Mcl-1 (Novus Biologicals) and Tris-HCl buffer (50 mM Tris-HCl (pH 7.5), 100 mM NaCl, 10 mM MgCl 2 , 5 mM DTT, 2 mM ATP , 10% Glycerol) and incubated for 30 minutes. Then, it was mixed with 26S human proteasome (LifeSensors) at a final concentration of 0.1 mg/mL and allowed to react for 2 hours. Each sample was subjected to size fractionation by SDS-PAGE, and then analyzed by Western blotting using an anti-Mcl-1 antibody (Cell Signaling Technology) as a primary antibody.
(結果)
結果を図10に示す。UbR3-B6及びUbR4-B6未添加の対照(レーン1)におけるMcl-1残存量を100%としたときに対するUbR3-B6を添加した場合(レーン4)又はUbR4-B6を添加した場合(レーン2)のMcl-1残存量は、それぞれ56%と9%であった。これは、UbR3-B6及びUbR4-B6がUb-PSM系依存的なMcl-1の分解誘導剤として機能することを示すと共に、特にUbR4が特に強力な分解誘導シグナルとして機能し得ることを示唆する。
以上の結果から、結合因子としてペプチドを用いた場合も、本願発明の標的ペプチド分解誘導剤は、標的ペプチドの分解を誘導できることが実証された。
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。
(result)
The results are shown in FIG. When UbR 3 -B6 and UbR 4 -B6 were not added to the control (lane 1), the remaining amount of Mcl-1 was assumed to be 100% when UbR 3 -B6 was added (lane 4) or when UbR 4 -B6 was added The residual Mcl-1 levels in case (lane 2) were 56% and 9%, respectively. This indicates that UbR 3 -B6 and UbR 4 -B6 function as Ub-PSM system-dependent Mcl-1 degradation inducers, and that UbR 4 in particular can function as a particularly strong degradation-inducing signal. Suggest.
From the above results, it was demonstrated that the target peptide degradation-inducing agent of the present invention can induce degradation of the target peptide even when a peptide is used as the binding factor.
All publications, patents and patent applications cited herein are hereby incorporated by reference in their entirety.
Claims (20)
(a)配列番号1で示すアミノ酸配列、
(b)配列番号1で示すアミノ酸配列において1又は複数個のアミノ酸が付加、欠失又は置換されたアミノ酸配列、又は
(c)配列番号1で示すアミノ酸配列と90%以上のアミノ酸同一性を有するアミノ酸配列 The target peptide degradation inducer according to claim 1, wherein said ubiquitin consists of any one of the amino acid sequences described in (a) to (c) below.
(a) the amino acid sequence shown in SEQ ID NO: 1,
(b) an amino acid sequence in which one or more amino acids are added, deleted or substituted in the amino acid sequence shown in SEQ ID NO: 1, or (c) having 90% or more amino acid identity with the amino acid sequence shown in SEQ ID NO: 1 amino acid sequence
(1)72位及び74位のアルギニン残基がそれぞれアラニン残基及びスレオニン残基に置換されたアミノ酸配列、
(2)72位及び74位のアルギニン残基がそれぞれプロリン残基及びスレオニン残基に置換されたアミノ酸配列、又は
(3)73位のロイシン残基がプロリン残基に置換されたアミノ酸配列 The target peptide degradation-inducing agent according to claim 2, wherein the ubiquitin comprises an amino acid sequence containing any one of the following amino acid substitutions described in (1) to (3) in the amino acid sequence shown in SEQ ID NO:1.
(1) an amino acid sequence in which arginine residues at positions 72 and 74 are substituted with alanine residues and threonine residues, respectively;
(2) an amino acid sequence in which the arginine residues at positions 72 and 74 are substituted with proline residues and threonine residues, respectively; or (3) an amino acid sequence in which the leucine residue at position 73 is substituted with a proline residue
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2023554735A JPWO2023068328A1 (en) | 2021-10-20 | 2022-10-20 |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2021-172029 | 2021-10-20 | ||
| JP2021172029 | 2021-10-20 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2023068328A1 true WO2023068328A1 (en) | 2023-04-27 |
Family
ID=86058325
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2022/039089 Ceased WO2023068328A1 (en) | 2021-10-20 | 2022-10-20 | Agent for inducing decomposition of target peptide |
Country Status (2)
| Country | Link |
|---|---|
| JP (1) | JPWO2023068328A1 (en) |
| WO (1) | WO2023068328A1 (en) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JP2006096663A (en) * | 2002-10-17 | 2006-04-13 | Sangaku Renkei Kiko Kyushu:Kk | Cancer gene vaccine |
| WO2007058235A1 (en) * | 2005-11-17 | 2007-05-24 | Medinet Co., Ltd. | Fusion protein and use thereof for pharmaceutical purposes |
| US20160076074A1 (en) * | 2014-09-17 | 2016-03-17 | Northwestern University | Probes and assays for measuring e3 ligase activity |
| US20200172954A1 (en) * | 2014-09-30 | 2020-06-04 | University Of Dundee | Cysteine Labelling |
-
2022
- 2022-10-20 JP JP2023554735A patent/JPWO2023068328A1/ja active Pending
- 2022-10-20 WO PCT/JP2022/039089 patent/WO2023068328A1/en not_active Ceased
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JP2006096663A (en) * | 2002-10-17 | 2006-04-13 | Sangaku Renkei Kiko Kyushu:Kk | Cancer gene vaccine |
| WO2007058235A1 (en) * | 2005-11-17 | 2007-05-24 | Medinet Co., Ltd. | Fusion protein and use thereof for pharmaceutical purposes |
| US20160076074A1 (en) * | 2014-09-17 | 2016-03-17 | Northwestern University | Probes and assays for measuring e3 ligase activity |
| US20200172954A1 (en) * | 2014-09-30 | 2020-06-04 | University Of Dundee | Cysteine Labelling |
Also Published As
| Publication number | Publication date |
|---|---|
| JPWO2023068328A1 (en) | 2023-04-27 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US8334101B2 (en) | Intracellular DNA receptor | |
| JP6077543B2 (en) | peptide | |
| JP2015519344A (en) | Translocation of non-natural chemical entities through the anthrax protective antigen pore | |
| US20190269761A1 (en) | Methods, compositions and screens for therapeutics for the treatment of synovial sarcoma | |
| US7399583B2 (en) | Method for the identification of inhibitors of the binding of ARE-containing mRNA and a HuR protein | |
| Wang et al. | A macropinocytosis-intensifying albumin domain-based scFv antibody and its conjugate directed against K-Ras mutant pancreatic cancer | |
| WO2011071280A2 (en) | Intracelluar targeting bipodal peptide binder | |
| JP2013535954A (en) | Peptides, structures and uses thereof | |
| Vickers et al. | Interaction of ASOs with PC4 is highly influenced by the cellular environment and ASO chemistry | |
| Orbán et al. | A new daunomycin–peptide conjugate: synthesis, characterization and the effect on the protein expression profile of HL-60 cells in vitro | |
| Luo et al. | The heparin‐binding domain of HB‐EGF as an efficient cell‐penetrating peptide for drug delivery | |
| CN105999227B (en) | Expression and application of FOXM1 protein nitrogen terminal (1-234 aa) | |
| Miyajima et al. | Identification of low-density lipoprotein receptor-related protein 1 as a CXCL14 receptor using chemically synthesized tetrafunctional probes | |
| US20200360529A1 (en) | Peptidic materials that traffic efficiently to the cell cytosol and nucleus | |
| Winkler et al. | Oligonucleotides conjugated to short lysine chains | |
| WO2023068328A1 (en) | Agent for inducing decomposition of target peptide | |
| Bergen et al. | Evaluation of an LC8-binding peptide for the attachment of artificial cargo to dynein | |
| Alcock et al. | Potent Cyclic Peptide Inhibitors Disrupt the FANCM–RMI Interaction | |
| He et al. | Targeted degradation of cell surface proteins through endocytosis triggered by cell-penetrating peptide-small molecule conjugates | |
| Tang et al. | Interactions of hepatitis B core antigen and peptide inhibitors | |
| Li et al. | Efficient Synthetic Approach to Linear Dasatinib–DNA Conjugates by Click Chemistry | |
| JP2016053026A (en) | Virus-like particle vector for delivery of pharmaceutical agents, process for manufacture thereof, uses thereof and pharmaceutical composition | |
| Lambrechts et al. | The BRD4-nucleosome interaction is enhanced modestly and non-selectively by histone acetylation | |
| Li et al. | Chemoenzymatic Synthesis of DNP-Functionalized FGFR1-Binding Peptides as Novel Peptidomimetic Immunotherapeutics for Treating Lung Cancer | |
| WO2012082069A1 (en) | Protein aptamers based on unstructured scaffold proteins |
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: 22883632 Country of ref document: EP Kind code of ref document: A1 |
|
| ENP | Entry into the national phase |
Ref document number: 2023554735 Country of ref document: JP Kind code of ref document: A |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 22883632 Country of ref document: EP Kind code of ref document: A1 |