WO2023053315A1 - Pd-1 expression inhibitor, pharmaceutical composition for immune checkpoint inhibition, and method for inhibiting pd-1 expression - Google Patents

Pd-1 expression inhibitor, pharmaceutical composition for immune checkpoint inhibition, and method for inhibiting pd-1 expression Download PDF

Info

Publication number
WO2023053315A1
WO2023053315A1 PCT/JP2021/036056 JP2021036056W WO2023053315A1 WO 2023053315 A1 WO2023053315 A1 WO 2023053315A1 JP 2021036056 W JP2021036056 W JP 2021036056W WO 2023053315 A1 WO2023053315 A1 WO 2023053315A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
expression
lymphocytes
polynucleotide
truncated
Prior art date
Application number
PCT/JP2021/036056
Other languages
French (fr)
Japanese (ja)
Inventor
宗夫 沼崎
Original Assignee
国立大学法人東北大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立大学法人東北大学 filed Critical 国立大学法人東北大学
Priority to PCT/JP2021/036056 priority Critical patent/WO2023053315A1/en
Publication of WO2023053315A1 publication Critical patent/WO2023053315A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to a PD-1 expression inhibitor, a pharmaceutical composition for immune checkpoint inhibition, and a method for inhibiting PD-1 expression.
  • PD-1 is a molecule expressed on the surface of activated lymphocytes such as cytotoxic T cells (CTL). It is known that when PD-1 binds to a cancer cell surface ligand (such as PD-L1), CTL activation is suppressed, and cytotoxic activity against cancer cells is suppressed. Immune checkpoint inhibitors targeting PD-1 inhibit the reduction of cytotoxic activity of CTL by inhibiting ligand binding to PD-1.
  • Interleukin 36 is a cytokine reported as a molecule with homology to IL-1 by DNA database analysis. IL-36, along with IL-1, IL-18, IL-33, IL-37, and IL-38, make up the IL-1 cytokine family. IL-36 is a secretory protein that is secreted from specific producing cells, but the IL-36 gene does not have a sequence corresponding to a signal sequence, and the secretion mechanism has not been elucidated.
  • the receptor for IL-36 consists of a heterodimer of IL-36 receptor (IL-36R) and IL-1 accessory protein (IL-1RAcP).
  • IL-36 has three isoforms, IL-36 ⁇ , IL-36 ⁇ and IL-36 ⁇ , as splice variants.
  • IL-36 ⁇ is mainly produced by epithelial cells, monocytes, B cells and the like. It has been reported that truncated IL-36 ⁇ , in which 30 amino acids at the N-terminus have been removed, has about 1000-fold stronger activity than native full-length IL-36 ⁇ (Non-Patent Document 1). . It has been reported that IL-36 ⁇ acts on dendritic cells to enhance the production of IL-6, IL-12, GM-CSF, etc., and enhances the expression of cell surface MHC class II, CD80, CD86, etc. (Non-Patent Document 2).
  • Interleukin-36 (IL-36) ligands require processing for full agonist (IL-36 ⁇ , IL-36 ⁇ , and IL-36 ⁇ ) or antagonist (IL-36Ra) activity. J Biol Chem. 2011 Dec 9; 286(49):42594-602. Vigne S, et al., IL-36R ligands are potent regulators of dendritic and T cells. Blood. 2011 Nov 24; 118(22):5813-23.
  • Monoclonal antibodies against immune checkpoint molecules such as PD-1 are used as immune checkpoint inhibitors.
  • PD-1 Monoclonal antibodies against immune checkpoint molecules
  • an object of the present invention is to provide a PD-1 expression suppressing agent, a pharmaceutical composition for immune checkpoint inhibition, and a method for suppressing PD-1 expression that can suppress the expression of PD-1 in lymphocytes.
  • a PD-1 expression inhibitor comprising at least one component selected from the group consisting of the following (a) to (d): (a) interleukin 36 or a variant thereof; (b) a polynucleotide having a nucleotide sequence encoding interleukin-36 or a variant thereof; (c) a vector comprising the polynucleotide of (b); and (d) a cell that secretes interleukin-36 or a variant thereof.
  • the PD-1 expression inhibitor according to [1] which suppresses PD-1 expression in CD8-positive T cells or CD4-positive T cells.
  • a pharmaceutical composition for immune checkpoint inhibition comprising the PD-1 expression inhibitor of [1] or [2] and a pharmaceutically acceptable carrier.
  • a method of suppressing PD-1 expression in lymphocytes comprising culturing lymphocytes in the presence of interleukin 36 or a variant thereof.
  • a PD-1 expression inhibitor a pharmaceutical composition for immune checkpoint inhibition, and a method for inhibiting PD-1 expression are provided, which are capable of suppressing PD-1 expression in lymphocytes.
  • FIG. 4 shows the results of analyzing the PD1 expression inhibitory effect of truncated mouse IL-36 ⁇ on CD8-positive T cells and CD4-positive T cells by flow cytometry.
  • FIG. 4 shows the results of analyzing the PD1 expression inhibitory effect of truncated mouse IL-36 ⁇ on CD8-positive T cells and CD4-positive T cells by flow cytometry.
  • FIG. 2 shows the mean fluorescence intensity (MFI) of PE (PD-1 positive) based on the flow cytometry analysis results of FIGS. 1 and 2.
  • FIG. FIG. 4 shows the results of analyzing the PD1 expression inhibitory effect of truncated mouse IL-36 ⁇ on CD8-positive T cells and CD4-positive T cells by flow cytometry.
  • FIG. 1 mean fluorescence intensity
  • FIG. 4 shows the results of analyzing the PD1 expression inhibitory effect of truncated mouse IL-36 ⁇ on CD8-positive T cells and CD4-positive T cells by flow cytometry.
  • FIG. 5 shows the mean fluorescence intensity (MFI) of PE (PD-1 positive) based on the flow cytometry analysis results of FIGS. 4 and 5.
  • FIG. Results for CD8-positive T cells are shown. Splenocytes of mice transplanted with B16-F10Neo cells, B16-F10-PTH-truncated IL-36 ⁇ cells, or B16-F10-GH-truncated IL-36 ⁇ cells were harvested, and PD-1 expression was analyzed by flow cytometry. Analyzed results are shown.
  • the term “comprise” means that it may include components other than the target components.
  • the term “consist of” means containing no elements other than the subject element.
  • the term “consistently of” means that it does not include constituent elements other than the subject constituent elements in a mode that exhibits a special function (such as a mode that completely loses the effects of the invention). means.
  • the term “comprise” includes aspects that "consist of” and aspects that "consist essentially of.”
  • Proteins, peptides, polynucleotides (DNA, RNA), vectors, and cells can be isolated.
  • isolated means separated from the natural state or other components.
  • isolated can be substantially free of other components.
  • substantially free of other components means that the content of other components contained in the isolated component is negligible.
  • the content of other components contained in the isolated component is, for example, 10% by mass or less, 5% by mass or less, 4% by mass or less, 3% by mass or less, 2% by mass or less, 1% by mass or less, 0.5% by mass or less. It may be 5% by mass or less, or 0.1% by mass or less.
  • proteins, peptides, polynucleotides DNA, RNA
  • vectors and cells described herein may be isolated proteins, isolated peptides, isolated polynucleotides (isolated DNA, isolated RNA), isolated vectors, and isolated cells.
  • Polynucleotide refers to a nucleotide polymer in which nucleotides are linked by phosphodiester bonds.
  • a “polynucleotide” may be DNA, RNA, or may be composed of a combination of DNA and RNA.
  • a “polynucleotide” may be a polymer of natural nucleotides, including natural nucleotides and non-natural nucleotides (analogs of natural nucleotides, nucleotides in which at least one of the base, sugar and phosphate moieties is modified). (for example, a phosphorothioate skeleton), etc.), or a polymer of non-natural nucleotides.
  • Nucleotide sequences of polynucleotides are written in the generally accepted single-letter code unless otherwise indicated. Unless otherwise indicated, nucleotide sequences are written 5' to 3'. Nucleotide residues constituting a polynucleotide may be simply described as adenine, thymine, cytosine, guanine, uracil, or the like, or their one-letter codes.
  • Gene refers to a polynucleotide containing at least one open reading frame that encodes a particular protein.
  • a gene may contain both exons and introns.
  • peptide polypeptide
  • protein protein
  • Peptides polypeptides and proteins
  • Amino acid sequences are written in the generally accepted one-letter or three-letter code unless otherwise specified. Unless otherwise specified, amino acid sequences are written from the N-terminal side to the C-terminal side.
  • operably linked when used in reference to polynucleotides means that a first nucleotide sequence is placed sufficiently close to a second nucleotide sequence such that the first nucleotide sequence is either the second nucleotide sequence or the second nucleotide sequence. means that it can affect areas under the control of For example, that a polynucleotide is operably linked to a promoter means that the polynucleotide is linked so that it is expressed under the control of the promoter.
  • expressible state refers to a state in which a gene of interest can be transcribed in a cell into which the gene of interest has been introduced.
  • expression vector refers to a vector containing a gene of interest and equipped with a system that renders the gene of interest expressible in cells into which the vector has been introduced.
  • a promoter can function means that a polynucleotide operably linked to the promoter can be expressed in the cells of the subject organism.
  • In-frame means that the reading frame of the codon is not shifted.
  • Sequence identity between amino acid sequences or nucleotide sequences is defined by aligning the two amino acid sequences or nucleotide sequences so that the corresponding amino acids or bases match the most, leaving gaps for insertions and deletions, It is determined as the ratio of matched amino acids or bases to the entire amino acid sequence or nucleotide sequence excluding gaps in the resulting alignment. Sequence identity between amino acid sequences or between nucleotide sequences can be determined using various homology search software known in the art. For example, the sequence identity value of an amino acid sequence can be obtained by calculation based on alignments obtained by the known homology search software BLASTP, and the sequence identity value of a nucleotide sequence can be obtained by a known homology search. It can be obtained by calculation based on alignments obtained by the software BLASTN. Sequence identity is also referred to as homology.
  • Stringent conditions means conditions under which two polynucleotides with high sequence identity can specifically hybridize. Two polynucleotides with high sequence identity, the sequence identity between the two polynucleotides, for example, 80% or more, 85% or more, 90% or more, 95% or more, 96% or more, 97% Above, 98% or more, or 99% or more. Stringent conditions include, for example, conditions described in Molecular Cloning-A Laboratory Manual Third Edition (Sambrook et al., Cold Spring Harbor Laboratory Press).
  • stringent conditions include, for example, 6 x SSC (composition of 20 x SSC: 3 M sodium chloride, 0.3 M citric acid solution, pH 7.0), 5 x Denhardt's solution (composition of 100 x Denhardt's solution: 42 in a hybridization buffer consisting of 2% by weight bovine serum albumin, 2% by weight Ficoll, 2% by weight polyvinylpyrrolidone), 0.5% by weight SDS, 0.1 mg/mL salmon sperm DNA, and 50% formamide.
  • Conditions include incubation at ⁇ 70°C for several hours to overnight. Washing buffers used for washing after incubation include, for example, 0.1% by mass SDS-containing 1 ⁇ SSC solution and 0.1% by mass SDS-containing 0.1 ⁇ SSC solution.
  • a first aspect of the present disclosure is a PD-1 expression inhibitor.
  • the PD-1 expression inhibitor contains at least one component selected from the group consisting of the following (a) to (d): (a) IL-36 or variant thereof; (b) a polynucleotide having a nucleotide sequence encoding IL-36 or a variant thereof; (c) a vector comprising the polynucleotide of (b); and (d) a cell secreting IL-36 or a variant thereof.
  • lymphocyte PD-1 expression is suppressed in lymphocytes cultured in the presence of IL-36 compared to lymphocytes cultured in the absence of IL-36. was confirmed.
  • lymphocytes from mice transplanted with tumor cells that produce IL-36 the expression of PD-1 is suppressed compared to lymphocytes from mice transplanted with tumor cells that do not produce IL-36. confirmed.
  • the PD-1 expression inhibitor of this embodiment can be added to a medium for culturing lymphocytes and used to prepare lymphocytes in which PD-1 expression is suppressed in vitro or ex vivo.
  • Lymphocytes targeted for suppression of PD-1 expression are not particularly limited as long as they express PD-1.
  • Lymphocytes targeted for suppression of PD-1 expression include, for example, T cells and natural killer cells. Examples of T cells include CD8-positive T cells (CTL and the like), CD4-positive T cells (helper T cells and the like), and the like.
  • Organisms from which lymphocytes are derived are not particularly limited, but include, for example, humans and mammals other than humans.
  • Lymphocytes may be genetically modified.
  • the type of genetic modification is not particularly limited.
  • Specific examples of genetically modified lymphocytes include lymphocytes modified to express chimeric antigen receptors (CAR-T cells, etc.), lymphocytes modified to express exogenous TCR (TCR- T cells, etc.) and the like.
  • the PD-1 expression inhibitor of the present embodiment may be administered to a living body and used in vivo to suppress PD-1 expression of lymphocytes present in the living body.
  • the organism to which the PD-1 inhibitor is administered is not particularly limited as long as it has lymphocytes expressing PD-1.
  • Organisms to be administered include the same organisms as those described above.
  • IL-36 is a cytokine belonging to the IL-1 cytokine family.
  • IL-36 has three isoforms, IL-36 ⁇ , IL-36 ⁇ and IL-36 ⁇ , as splice variants.
  • IL-36 may be any of IL-36 ⁇ , IL-36 ⁇ , and IL-36 ⁇ , with IL-36 ⁇ being preferred.
  • the organism from which IL-36 is derived is not particularly limited, it is preferably the same species as the organism from which the lymphocytes whose PD-1 expression is to be suppressed are derived.
  • human lymphocytes when human lymphocytes are targeted for suppression of PD-1 expression, it is preferable to use human IL-36.
  • Mouse IL-36 is preferably used when the target for suppression of PD-1 expression is mouse lymphocytes.
  • IL-36 may be naturally occurring or modified.
  • Native IL-36 is IL-36 produced in nature.
  • Native IL-36 is, for example, IL-36 produced in mammals as described above.
  • Specific examples of native IL-36 include human IL-36 and mouse IL-36.
  • Examples of the amino acid sequence of human IL-36 ⁇ include the amino acid sequence described in SEQ ID NO: 2 (nucleotide sequence: SEQ ID NO: 1), the amino acid sequence described in SEQ ID NO: 4 (nucleotide sequence: SEQ ID NO: 3) and the like.
  • the amino acid sequence of mouse IL-36 ⁇ (NCBI Gene ID: 69677) includes, for example, the amino acid sequence set forth in SEQ ID NO: 6 (nucleotide sequence: SEQ ID NO: 5).
  • Native IL-36 is not limited to those having the above amino acid sequences, and may be homologues (orthologs or paralogs) thereof.
  • the amino acid sequence of native IL-36 can be obtained from sequence databases such as GenBank.
  • Modified IL-36 (hereinafter also referred to as "variants of IL-36”) is a polypeptide obtained by modifying native IL-36, and is a polypeptide that maintains the function of native IL-36. .
  • a variant of IL-36 has an amino acid sequence in which one or more amino acids are altered (substitution, deletion, addition, insertion, or a combination thereof) as compared with the amino acid sequence of native IL-36.
  • the number of amino acids to be modified is not particularly limited. , 1 to 3, or 1 to 2, and the like.
  • Modified IL-36 may have an amino acid sequence with 80% or more sequence identity with the amino acid sequence of native IL-36. Sequence identity can be, for example, 85% or greater, 90% or greater, 95% or greater, 96% or greater, 97% or greater, 98% or greater, or 99% or greater.
  • the variant of IL-36 preferably has at least one activity of native IL-36 that is equal to or greater than that of native IL-36.
  • the activity of native IL-36 includes, for example, the activity of suppressing PD-1 expression in lymphocytes.
  • IL-36 activity includes binding activity to a receptor consisting of a heterodimer of IL-36R and IL-1RAcp, activity to cause cells having the receptor (e.g., lung fibroblasts) to produce IL-6, and It may be antitumor activity or the like.
  • a variant of IL-36 having an activity equal to or greater than that of native IL-36 ⁇ has a relative activity of, for example, 80 or more, 85 or more, 90 or more, or 95 or more when the activity exhibited by natural IL-36 ⁇ is taken as 100. It shows activity.
  • Mouse IL-36 ⁇ has been reported to exhibit approximately 1,000-fold stronger activity compared to native IL-36 ⁇ in a variant in which 30 amino acids at the N-terminus have been removed (Towne JE, et al. 2011 Dec 9; 286(49):42594-602.). In human IL-36 ⁇ , it has been reported that a variant in which 4 amino acids at the N-terminus have been removed exhibits stronger activity than wild-type IL-36 ⁇ (Towne JE, et al., J Biol. Chem. 2011 Dec 9; 286(49):42594-602.).
  • variants of IL-36 ⁇ include those from which amino acids in the N-terminal region of native IL-36 ⁇ have been removed (hereinafter also referred to as "truncated IL-36 ⁇ ").
  • the upper limit of the number of N-terminal amino acids to be removed is, for example, 40 or less, 35 or less, 30 or less, 25 or less, 20 or less, 15 or less, 10 or less, 5 or less, or 4 Below are listed.
  • the lower limit for the number of N-terminal amino acids to be removed is 1 or more. The above upper limit and lower limit can be arbitrarily combined.
  • the number of N-terminal amino acids removed includes, for example, 1-40, 5-35, or 10-30.
  • the number of N-terminal amino acids removed includes, for example, 1-40, 2-30, 3-20, or 4-10.
  • the amino acid sequence of truncated human IL-36 ⁇ includes, for example, the amino acid sequence set forth in SEQ ID NO: 8 (nucleotide sequence: SEQ ID NO: 7), the amino acid sequence set forth in SEQ ID NO: 10 (nucleotide sequence: SEQ ID NO: 9), and the like. .
  • the amino acid sequence of truncated mouse IL-36 ⁇ is, for example, the amino acid sequence set forth in SEQ ID NO: 12 (nucleotide sequence: SEQ ID NO: 11).
  • the polynucleotide of (b) has a nucleotide sequence encoding IL-36 or a variant thereof (hereinafter referred to as "IL-36 coding sequence").
  • An IL-36 coding sequence is the nucleotide sequence of the IL-36 gene.
  • the polynucleotide (b) can also be said to be a polynucleotide containing the IL-36 gene.
  • the IL-36 coding sequence may encode native IL-36 or variants of IL-36.
  • the polynucleotides of (b) include, for example, the following polynucleotides. (1) A polynucleotide encoding a polypeptide having the amino acid sequence of native IL-36 (eg, SEQ ID NO: 2, 4 or 6). (2) an amino acid sequence in which one or more amino acids are modified (deleted, substituted, added, inserted, or a combination thereof) in the amino acid sequence of native IL-36 (e.g., SEQ ID NO: 2, 4 or 6); and which has IL-36 activity.
  • a polypeptide consisting of an amino acid sequence having 80% or more sequence identity with the amino acid sequence of native IL-36 (e.g., SEQ ID NO: 2, 4 or 6) and having IL-36 activity A polynucleotide that encodes a peptide.
  • a polynucleotide having the nucleotide sequence of a native IL-36 gene eg, SEQ ID NO: 1, 3 or 5).
  • a polynucleotide having a nucleotide sequence having 80% or more sequence identity with the nucleotide sequence of a native IL-36 gene (e.g., SEQ ID NO: 1, 3 or 5), and having IL-36 ⁇ activity A polynucleotide that encodes a polypeptide.
  • a polynucleotide that encodes a peptide is a polynucleotide having the nucleotide sequence of a native IL-36 gene (e.g., SEQ ID NO: 1, 3 or 5) and has IL-36 activity.
  • the number of modified amino acids in (2) above and the value of sequence identity in (3) above are as exemplified in ⁇ (a) IL-36 ⁇ > above.
  • the number of modified nucleotides in (5) is, for example, 1 to 100, 1 to 80 or less, 1 to 60, 1 to 50, 1 to 40, 1 to 30, 1 to 20, 1 to 10, 1 to 5, 1 to 4, 1 to 3, or 1 to 2, and the like.
  • the sequence identity in (6) may be, for example, 85% or more, 90% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more.
  • the IL-36 coding sequence may encode truncated IL-36 ⁇ . Examples include the following polynucleotides. (8) A polynucleotide encoding a polypeptide having the amino acid sequence set forth in SEQ ID NO:8, 10 or 12. (9) A polypeptide having an amino acid sequence in which one or more amino acids are altered (deleted, substituted, added, inserted, or a combination thereof) in the amino acid sequence set forth in SEQ ID NO: 8, 10 or 12, and a polypeptide that has IL-36 ⁇ activity.
  • the number of modified amino acids in (9) is, for example, 1 to 50, 1 to 40, 1 to 30, 1 to 20, 1 to 10, or 1 to 5, 1 to 4, 1 to 3, or 1 to 2, and the like.
  • the sequence identity in (10) may be, for example, 85% or more, 90% or more, or 95% or more.
  • the number of modified bases in (12) is, for example, 1 to 100, 1 to 80 or less, 1 to 60, 1 to 50, 1 to 40, 1 to 30, 1 to 20, 1 to 10 or 1 to 5, and the like.
  • the sequence identity in (13) includes, for example, 5% or more, 90% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more.
  • the polynucleotide of (b) may contain other nucleotide sequences in addition to the IL-36 coding sequence.
  • Other nucleotide sequences include, for example, a nucleotide sequence encoding a signal peptide (hereinafter referred to as "signal peptide coding sequence"), a sequence controlling IL-36 gene expression (hereinafter referred to as "expression control sequence”), and the like. is mentioned.
  • the polynucleotide of (b) preferably further has a signal peptide coding sequence for a secretory protein in addition to the IL-36 coding sequence.
  • the signal peptide coding sequence is preferably directly or indirectly linked in-frame to the 5' end of the IL-36 coding sequence.
  • a signal peptide coding sequence is ligated in-frame to the 5′ end of the IL-36 coding sequence, so that when IL-36 is produced in cells into which the polynucleotide of (b) has been introduced, IL- 36 ⁇ can be secreted extracellularly.
  • the signal peptide coding sequence includes a sequence that encodes a signal peptide that can function as a secretory signal in cells into which the polynucleotide of (b) can be introduced (hereinafter also referred to as "introduction target cells").
  • the signal peptide coding sequence is preferably derived from the same species of organism as the target cell.
  • the cells to be introduced are human cells, it is preferable to use the signal peptide coding sequence of a human secretory protein.
  • the cells to be introduced are mouse cells, it is preferable to use the signal peptide coding sequence of a mouse secretory protein.
  • the secreted protein from which the signal peptide coding sequence is derived is not particularly limited.
  • secretory proteins include peptide hormones such as parathyroid hormone and growth hormone; cytokines such as IFN- ⁇ , GM-CSF and IL-6; immunoglobulin heavy or light chains; not.
  • the signal peptide coding sequence include a nucleotide sequence (eg, SEQ ID NO: 13) encoding the amino acid sequence set forth in SEQ ID NO: 14 (the signal peptide of human parathyroid hormone (hPTH)), and the amino acid sequence set forth in SEQ ID NO: 16. and a nucleotide sequence (eg, SEQ ID NO: 15) encoding (signal peptide of human growth hormone (hGH)).
  • the signal peptide coding sequence may be directly linked to the 5' end of the IL-36 coding sequence or indirectly via a linker.
  • the linker is not particularly limited as long as it does not impair IL-36 activity when the signal peptide coding sequence-linker sequence-IL-36 coding sequence is translated.
  • the length of the linker may be, for example, 3 nucleotides, 6 nucleotides, 9 nucleotides, or 12 nucleotides.
  • the signal peptide coding sequence is directly linked to the 5' end of the IL-36 coding sequence.
  • the polynucleotide of (b) preferably has an expression control sequence in addition to the IL-36 coding sequence.
  • expression control sequences include promoters, enhancers, poly-A addition signals, terminators and the like.
  • the promoter is not particularly limited as long as it can function in the cells to be introduced.
  • Promoters include, for example, CMV (cytomegalovirus) promoter, SR ⁇ promoter, SV40 early promoter, retroviral LTR, RSV (Rous sarcoma virus) promoter, HSV-TK (herpes simplex virus thymidine kinase) promoter, EF1 ⁇ promoter, metallothionein promoters, heat shock promoters, etc., but are not limited thereto.
  • the IL-36 coding sequence is preferably operably linked to a suitable promoter. If the polynucleotide of (b) has a signal peptide coding sequence, it is preferred that the signal peptide coding sequence and the IL-36 coding sequence are operably linked to a suitable promoter.
  • the polynucleotide of (b) may be DNA or RNA.
  • DNA it may be in the form of a vector described below.
  • RNA it may be in the form of mRNA.
  • the polynucleotide (b) may have a 5'Cap structure, poly-A-tail, or the like.
  • the vector of (c) contains the polynucleotide of (b).
  • the type of vector is not particularly limited, and an expression vector or the like generally used for gene expression can be used.
  • Vectors may be linear or circular. Examples of vectors include non-viral vectors such as plasmids, viral vectors, transposon vectors, episomal vectors, artificial chromosome vectors and the like.
  • plasmid vectors include animal cell expression plasmid vectors such as pA1-11, pXT1, pRc/CMV, pRc/RSV, and pcDNAI/Neo.
  • Viral vectors include, for example, Sendai virus vectors, retrovirus (including lentivirus) vectors, adenovirus vectors, adeno-associated virus vectors, herpes virus vectors, vaccinia virus vectors, pox virus vectors, polio virus vectors, silvis virus vectors , rhabdovirus vector, paramyxovirus vector, orthomyxovirus vector and the like.
  • An episomal vector is a vector that can replicate autonomously outside the chromosome.
  • Examples of episomal vectors include vectors containing, as vector elements, sequences necessary for autonomous replication derived from EBV, SV40, and the like.
  • Vector elements required for autonomous replication specifically include genes encoding replication origins and proteins that bind to replication origins to control replication.
  • EBV includes the replication origin oriP and the EBNA-1 gene
  • SV40 includes the replication origin ori and the SV40LT gene.
  • artificial chromosome vectors examples include YAC (Yeast artificial chromosome) vectors, BAC (Bacterial artificial chromosome) vectors, PAC (P1-derived artificial chromosome) vectors, and the like.
  • Vectors can be produced using commercially available products.
  • a vector containing the polynucleotide (b) can be produced by inserting the polynucleotide (b) into the multicloning site of a commercially available vector. Insertion of the polynucleotide (b) into a commercially available vector can be performed by, for example, a combination of restriction enzyme treatment, ligase treatment, and the like.
  • the vector (c) is a targeting vector that is integrated into the genome of the target cell by genome editing technology or the like, it preferably contains homology arms for homologous recombination.
  • a homology arm has a sequence homologous to any genomic region of the cell to be introduced.
  • the genomic region homologous to the homology arms is the region into which the polynucleotide of (b) is inserted.
  • the homology arms are located flanking the 5' and 3' sides of the polynucleotide of (b).
  • the sequence of homology arms can be appropriately designed according to the type of cell to be introduced and the target region. For example, when the cells to be introduced are tumor cells, an oncogene possessed by the tumor cells may be selected as a target region and homology arms may be designed.
  • the cells of (d) are cells that secrete IL-36 or variants thereof (hereinafter referred to as "IL-36-secreting cells").
  • the IL-36-secreting cells may be native IL-36-secreting cells or transgenic IL-36-secreting cells.
  • the IL-36-secreting cells are preferably cells of the organism to which they are administered.
  • Native IL-36 secreting cells are cells that endogenously produce and secrete IL-36.
  • Native IL-36-secreting cells include, for example, epithelial cells, monocytes, B cells, and the like.
  • Transgenic IL-36-secreting cells are cells into which the polynucleotide of (b) or the vector of (c) has been introduced.
  • Host cells are not particularly limited, but include, for example, cells collected from an administration subject, cells having the same MHC as that of the administration subject, and the like. Examples of cell types include, but are not limited to, tumor cells, immune cells (dendritic cells, T cells, natural killer cells, etc.), fibroblasts, and the like.
  • a host cell can be, for example, a T cell. Examples of T cells include CD8-positive T-cells (CTL etc.), CD4-positive T-cells (helper T-cells etc.) and the like.
  • Transgenic IL-36-secreting cells can be prepared using known gene transfer methods depending on the type of vector.
  • Gene introduction methods include, for example, the virus infection method, lipofection method, microinjection method, calcium phosphate method, DEAE-dextran method, electroporation method, method using transposon, particle gun method and the like.
  • Transgenic IL-36-secreting cells may be prepared using known gene editing techniques and the like.
  • Gene editing techniques include, for example, techniques using endonucleases such as zinc finger nucleases, TALENs (transcription activation-like effector nucleases), and CRISPR-Cas systems.
  • the vector (c) may contain a marker gene.
  • a marker gene can be used to select IL-36 ⁇ -transfected cells.
  • Commonly used marker genes eg, drug resistance genes, fluorescent dye genes
  • Examples of marker genes include, but are not limited to, neomycin resistance gene, ampicillin resistance gene, hygromycin resistance gene, tetracycline resistance gene, chloramphenicol resistance gene, and the like.
  • the marker gene may be linked to the IL-36 coding sequence (IL-36 gene) via a nucleotide sequence encoding a self-cleaving terminal peptide such as 2A peptide, or an IRES (internal ribozyme entry site) sequence or the like. . By interposing these sequences, the IL-36 gene and the marker gene can be expressed independently from one promoter.
  • IL-36 secretion from IL-36-secreting cells can be confirmed by a known method.
  • IL-36 secretion from IL-36-secreting cells is confirmed by collecting the culture supernatant of IL-36-secreting cells and measuring IL-36 by Western blotting or an immunological method such as ELISA. can do.
  • IL-36-secreting cells by allowing the culture supernatant of IL-36-secreting cells to act on IL-6-producing cells such as fibroblasts and measuring the amount of IL-6 secreted from the fibroblasts.
  • the PD-1 expression inhibitor of the present embodiment may contain any one of the components (a) to (d) alone, or may contain two or more in combination.
  • the PD-1 expression inhibitor can contain at least one component selected from the group consisting of (a) to (d) as an active ingredient for the PD-1 expression inhibitory effect.
  • the "PD-1 expression inhibitory effect" means that when the PD-1 expression inhibitor is exposed to the lymphocytes, compared with the case where the PD-1 expression inhibitor is not exposed to the lymphocytes, PD-1 of the lymphocytes. It refers to the effect of suppressing expression.
  • culturing lymphocytes in the presence of a PD-1 expression inhibitor suppresses PD-1 expression in lymphocytes compared to culturing in the absence of a PD-1 expression inhibitor.
  • PD-1 expression level is 100 when lymphocytes are cultured in the absence of a PD-1 expression inhibitor
  • PD- The relative expression level of 1 may be 90 or less, 80 or less, 70 or less, 60 or less, 50 or less, or 40 or less.
  • the PD-1 expression level of lymphocytes can be confirmed by a known method. Methods for confirming the PD-1 expression level include, for example, flow cytometry analysis, RT-qPCR and the like.
  • PD-1 expression in lymphocytes can be effectively suppressed by the PD-1 expression inhibitor of the present embodiment.
  • the PD-1 expression inhibitor of this embodiment can be used to prepare lymphocytes used for adoptive immunotherapy or CAR-T therapy for cancer.
  • lymphocytes with suppressed PD-1 expression can be prepared by culturing lymphocytes in the presence of the PD-1 expression inhibitor of the present embodiment. By administering such lymphocytes to cancer patients, they can bypass the immune checkpoint system and express cytotoxic activity against cancer cells.
  • component (a) or (d) it is preferable to use component (a) or (d).
  • the PD-1 expression inhibitor of the present embodiment may be administered to a living body in order to suppress PD-1 expression of lymphocytes in the living body.
  • the PD-1 expression inhibitor is the component (a)
  • IL-36 or a variant thereof contacts lymphocytes in vivo and PD-1 expression in the lymphocytes is inhibited.
  • the PD-1 expression inhibitor is the component (b) or (c)
  • cells incorporating the polynucleotide of (b) or the vector of (c) produce IL-36 or a variant thereof.
  • the produced IL-36 or its variant comes into contact with lymphocytes in vivo, and PD-1 expression in the lymphocytes is suppressed.
  • the PD-1 expression inhibitor is a component of (d)
  • IL-36 produced by the cells of (d) or a variant thereof contacts lymphocytes in vivo, and PD-1 expression of the lymphocytes is suppressed.
  • a second aspect of the present disclosure is a pharmaceutical composition for immune checkpoint inhibition.
  • the pharmaceutical composition for immune checkpoint inhibition contains the PD-1 expression inhibitor of the first aspect and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition of this embodiment is administered to a subject to suppress the immune checkpoint system.
  • the administered or produced IL-36 or its variant acts on lymphocytes in vivo. This suppresses PD-1 expression in lymphocytes and suppresses the PD-1-mediated immune checkpoint system.
  • the subject of administration of the pharmaceutical composition of the present embodiment is a subject requiring inhibition of the immune checkpoint system via PD-1.
  • Administration subjects include, for example, cancer patients.
  • the pharmaceutical composition of the present embodiment may be administered alone or in combination with an anticancer agent.
  • the pharmaceutical composition of this embodiment contains a pharmaceutically acceptable carrier in addition to the PD-1 expression inhibitor of the first aspect.
  • “Pharmaceutically acceptable carrier” means a carrier that does not inhibit the physiological activity of the active ingredient and does not exhibit substantial toxicity to the subject to which it is administered.
  • the phrase “substantially non-toxic” means that the component does not show toxicity to the subject at the dose normally used.
  • the pharmaceutically acceptable carrier does not inhibit the PD-1 expression inhibitory activity of the components (a) to (d), and is substantially It is a non-toxic carrier.
  • Pharmaceutically acceptable carriers include any known pharmaceutically acceptable ingredients that are typically considered non-active ingredients.
  • Pharmaceutically acceptable carriers include, but are not limited to, solvents, diluents, vehicles, excipients, glidants, binders, granulating agents, dispersing agents, suspending agents, wetting agents, Lubricants, disintegrants, solubilizers, stabilizers, emulsifiers, fillers, preservatives (e.g., antioxidants), chelating agents, flavoring agents, sweeteners, thickeners, buffering agents, coloring agents, etc. mentioned.
  • One type of pharmaceutically acceptable carrier may be used alone, or two or more types may be used in combination.
  • pharmaceutically acceptable carriers may be solvents, diluents, vehicles, buffers and the like.
  • the pharmaceutically acceptable carrier may be a cell culture medium, a buffer (physiological saline, PBS, citrate buffer, etc.), or the like.
  • the pharmaceutical composition of the present embodiment in addition to those listed as pharmaceutically acceptable carriers, additives commonly used in the pharmaceutical field can be used without particular limitation.
  • the pharmaceutical composition may contain a transfection promoter, a genome editing reagent (CRISPR/Cas9 system, etc.) and the like.
  • the dosage form of the pharmaceutical composition is not particularly limited, and may be a dosage form commonly used as a pharmaceutical preparation.
  • the pharmaceutical composition may be an oral formulation or a parenteral formulation.
  • oral preparations include tablets, coated tablets, pills, powders, granules, capsules, syrups, fine granules, liquids, drops, emulsions and the like.
  • parenteral formulations include injections, suppositories, ointments, sprays, nasal drops, and inhalants.
  • Pharmaceutical compositions in these dosage forms can be formulated according to standard methods (eg, methods described in the Japanese Pharmacopoeia).
  • the pharmaceutical composition of this embodiment is preferably a parenteral formulation, more preferably an injection.
  • the pharmaceutical composition of this embodiment may contain active ingredients other than the PD-1 expression inhibitor of the first aspect.
  • the active ingredient is not particularly limited. Animal/microorganism extract, antipruritic, anti-inflammatory analgesic, antifungal agent, antihistamine, sedative hypnotic, tranquilizer, antihypertensive, hypotensive diuretic, antibiotic, anesthetic, antibacterial, antiepileptic, coronary artery Examples include, but are not limited to, dilating agents, herbal medicines, antipruritic agents, keratin softening exfoliants, and the like.
  • the active ingredient may also be another anti-tumor agent. Other antitumor agents may be appropriately selected according to the type of cancer to be applied. Other active ingredients may be used alone or in combination of two or more.
  • the pharmaceutical composition of this embodiment can contain a therapeutically effective amount of the PD-1 expression inhibitor of the first aspect.
  • therapeutically effective amount is meant an amount of drug effective to achieve the purpose of the pharmaceutical composition.
  • a therapeutically effective amount of any of components (a)-(d) can be an amount capable of suppressing PD-1 expression in lymphocytes.
  • the therapeutically effective amount is, for example, 0.01 ⁇ g to 100 mg, 0.1 ⁇ g to 10 mg, 0.5 ⁇ g per dosage unit form. up to 5 mg, or 1 ⁇ g to 1 mg, and the like.
  • the therapeutically effective amount is, for example, 10 3 to 10 10 cells, 10 4 to 10 9 cells, or 10 5 to 10 5 cells per dosage unit form. 10 8 etc. are mentioned.
  • compositions can be administered to subjects in need thereof by known methods.
  • the administration method may be oral administration or parenteral administration, but parenteral administration is preferred.
  • a parenteral route may be any route of administration other than the oral route, such as intravenous administration, intramuscular administration, subcutaneous administration, intranasal administration, intradermal administration, intraocular administration, intracerebral administration, intrarectal administration. , vaginal administration, and intraperitoneal administration.
  • the administration method may be local administration or systemic administration. Preferred routes of administration include intravenous administration, subcutaneous administration, intramuscular administration, intratumoral administration, and the like.
  • the dosage and administration interval of the pharmaceutical composition can be appropriately selected depending on the age, sex, weight, etc. of the administration subject, the type, progression, symptoms, etc. of the disease, administration method, and the like.
  • the dosage can be a therapeutically effective amount of the PD-1 expression inhibitor of the first aspect.
  • the therapeutically effective amount is, for example, 0.01 ⁇ g to 100 mg, 0.1 ⁇ g to 10 mg, 0.5 ⁇ g per administration. up to 5 mg, or 1 ⁇ g to 1 mg, and the like.
  • the therapeutically effective amount is, for example, 10 3 to 10 10 cells, 10 4 to 10 9 cells, or 10 5 to 10 5 cells per administration. 10 8 etc. are mentioned.
  • the pharmaceutical composition may be a single dose or multiple doses.
  • the administration interval can be, for example, every day, every 2-3 days, every week, every 10-30 days, every month, every 3-6 months, or every year. .
  • a third aspect of the present disclosure is a method of inhibiting PD-1 expression in lymphocytes.
  • the method includes culturing lymphocytes in the presence of IL-36 or a variant thereof.
  • the method of this embodiment is a method performed in vitro or ex vivo. Lymphocytes with reduced PD-1 expression can be produced by the method of the present embodiment.
  • IL-36 or variant thereof As IL-36 or a variant thereof, the same ones as listed in [PD-1 expression inhibitor] can be used.
  • Lymphocytes are not particularly limited as long as they express PD-1.
  • lymphocytes include those listed in [PD-1 expression inhibitor].
  • Preferred lymphocytes include CD8-positive T cells (CTL, etc.), CD4-positive T cells (helper T cells, etc.), modified cells thereof (CAR-T, TCR-T, etc.), and the like.
  • Lymphocytes may be included in a cell population containing multiple types of cells.
  • Cell populations containing lymphocytes include, for example, peripheral blood mononuclear cells (PBMC), splenocytes, thymocytes, and the like.
  • PBMC peripheral blood mononuclear cells
  • ⁇ Culture> Culturing is performed in the presence of IL-36 or a variant thereof. More specifically, lymphocytes are cultured in a medium containing IL-36 or a variant thereof.
  • the medium includes, for example, a medium commonly used for culturing animal cells (hereinafter also referred to as "animal cell medium”) supplemented with IL-36 or a variant thereof.
  • Animal cell media include basal media for animal cell culture.
  • basal media include Doulbecco's modified Eagle's Medium (DMEM) medium, DMEM/F12 medium, Advanced DMEM/F12 medium, IMDM medium, Medium 199 medium, Eagle's Minimum Essential Medium (EMEM) medium, ⁇ MEM medium, Examples include Ham's F12 medium, RPMI1640 medium, Fischer's medium, and mixed medium thereof.
  • DMEM Doulbecco's modified Eagle's Medium
  • DMEM/F12 medium DMEM/F12 medium
  • Advanced DMEM/F12 medium Advanced DMEM/F12 medium
  • IMDM medium Medium 199 medium
  • EMEM Eagle's Minimum Essential Medium
  • ⁇ MEM medium Examples include Ham's F12 medium, RPMI1640 medium, Fischer's medium, and mixed medium thereof.
  • the animal cell medium may be a basal medium to which any component has been added. Additional components include, for example, serum (fetal bovine serum (FBS), etc.), serum substitutes, and the like. Serum replacements include, for example, albumin, transferrin, sodium selenite, ITS-X (Invitrogen), knockout serum replacement (KSR), N2 supplement (Invitrogen), B27 supplement (Invitrogen), fatty acids, Insulin, collagen precursors, trace elements, 2-mercaptoethanol, 3′-thiolglycerol, etc.
  • serum fetal bovine serum
  • Serum replacements include, for example, albumin, transferrin, sodium selenite, ITS-X (Invitrogen), knockout serum replacement (KSR), N2 supplement (Invitrogen), B27 supplement (Invitrogen), fatty acids, Insulin, collagen precursors, trace elements, 2-mercaptoethanol, 3′-thiolglycerol, etc.
  • Additional components include lipids, amino acids, L-glutamine, Glutamax, non-essential amino acids, vitamins, growth factors, Antibiotics (penicillin, streptomycin, etc.), antioxidants, pyruvic acid, buffers, inorganic salts, etc. These additive components may be used singly or in combination of two or more. good.
  • the concentration of IL-36 or its variant in the medium is not particularly limited as long as it is a concentration capable of suppressing PD-1 expression in lymphocytes.
  • Concentrations of IL-36 or variants thereof include, for example, 0.001 to 1000 ng/mL.
  • the lower limit of the concentration of IL-36 or its variant in the medium is, for example, 0.001 ng/mL or more, 0.003 ng/mL or more, 0.05 ng/mL or more, 0.07 ng/mL or more, or 0 .01 ng/mL or greater, 0.03 ng/mL or greater, 0.05 ng/mL or greater, 0.07 ng/mL or greater, 0.1 ng/mL or greater, 0.3 ng/mL or greater, or 0.5 ng/mL or greater. be done.
  • the upper limit of the concentration of IL-36 or its variant in the medium is, for example, 1000 ng/mL or less, 500 ng/mL or less, 300 ng/mL or less, 100 ng/mL or less, 70 ng/mL or less, 50 ng/mL or less, 40 ng/mL or less, or 30 ng/mL or less. These upper and lower limits can be combined arbitrarily.
  • the concentration range of IL-36 or its variants in the medium is, for example, 0.001 to 500 ng/mL, 0.005 to 300 ng/mL, 0.01 to 100 ng/mL, 0.01 to 50 ng/mL, 0.01 to 40 ng/mL, 0.01 to 30 ng/mL, 0.005 to 0.03 ng/mL, 0.3 to 40 ng/mL, or 0.5 to 30 ng/mL.
  • Culturing can be performed under culture conditions generally used for culturing lymphocytes.
  • Culture conditions include, for example, a culture temperature of 32-40° C. (preferably 35-38° C., typically 37° C.) and a CO 2 concentration of 2-5% (preferably 5%).
  • the culture period should be sufficient to suppress PD-1 expression.
  • Examples of the culture period include 1 to 150 hours, 5 to 100 hours, 10 to 80 hours, 15 to 70 hours, 20 to 60 hours, 25 to 50 hours, or 30 to 50 hours.
  • Lymphocyte activation treatment may be performed during the culture period or before the culture. Lymphocyte activation treatment can be performed by a known method. Methods of activation treatment of lymphocytes include methods of stimulating the TCR/CD3 complex and co-stimulatory molecules (such as CD28). Stimulation of the TCR/CD3 complex can be done using a CD3 binding agent. A co-stimulatory molecule can be stimulated using a co-stimulatory molecule-binding substance.
  • CD3 binding agents include anti-CD3 antibodies or antigen-binding fragments thereof.
  • Co-stimulatory molecule binding agents include anti-CD28 antibodies or antigen-binding fragments thereof.
  • An antigen-binding fragment is an antibody fragment that retains the antigen-binding properties of the original antibody.
  • Antigen-binding fragments include, for example, Fab, Fab', F(ab') 2 , Fv, scFv and the like.
  • Anti-CD3 antibody and anti-CD28 antibody Monoclonal antibodies are preferred.
  • Lymphocyte activation treatment can be performed by culturing lymphocytes in the presence of a CD3-binding substance and a CD28-binding substance.
  • the activation treatment of lymphocytes can be performed simultaneously with culturing in the presence of IL-36 or its variants.
  • a method of culturing lymphocytes in wells coated with a CD3-binding substance using a medium containing IL-36 or a variant thereof and a CD28-binding substance can be mentioned.
  • Examples of the medium, culture conditions, and culture period are the same as those described above.
  • Lymphocyte activation treatment may be performed prior to culturing in the presence of IL-36 or its variants.
  • a method of culturing lymphocytes in a well coated with a CD3 binding substance using a medium containing a CD28 binding substance can be mentioned.
  • the medium and culture conditions are the same as those described above.
  • the culture period is, for example, 15 to 100 hours, or 20 to 80 hours.
  • lymphocytes are collected.
  • the method of this embodiment can then be carried out by culturing the collected lymphocytes in the presence of IL-36 or a variant thereof.
  • the method of the present embodiment can suppress PD-1 expression in lymphocytes.
  • the PD-1 expression level in lymphocytes cultured in the absence of IL-36 or a variant thereof is set to 100
  • the relative expression level of PD-1 in lymphocytes obtained by the method of the present embodiment is, for example, It can be 90 or less, 80 or less, 70 or less, 60 or less, 50 or less, or 40 or less.
  • lymphocytes with a reduced PD-1 expression level can be suppressed to obtain lymphocytes with a reduced PD-1 expression level.
  • Lymphocytes with a reduced PD-1 expression level easily evade the immune checkpoint system when administered to a living body. Therefore, it can exhibit cytotoxic activity against target cells (for example, cancer cells) without being inhibited by the immune checkpoint system.
  • the lymphocytes obtained by the method of the present embodiment have a reduced PD-1 expression level, so they can be suitably used for cancer therapy and the like.
  • the method of the present embodiment can also be said to be a method for producing lymphocytes with reduced PD-1 expression level.
  • the present invention provides at least one component selected from the group consisting of (a) to (d), which is used for PD-1 expression suppression.
  • the present invention provides a method of suppressing PD-1 expression in lymphocytes, comprising administering to a subject at least one component selected from the group consisting of (a) to (d). offer.
  • the subject is, for example, a cancer patient.
  • the present invention provides at least one component selected from the group consisting of (a) to (d), which is used in the production of PD-1 expression inhibitors.
  • the present invention provides at least one component selected from the group consisting of (a) to (d), which is used for the production of a pharmaceutical composition for immune checkpoint inhibition.
  • the present invention provides in vitro use of at least one component selected from the group consisting of (a) to (d) for suppressing PD-1 expression in lymphocytes. do.
  • Example 1 (Preparation of splenocytes) Spleens were removed from 10-week-old female C57BL/6 mice and immediately immersed in 20 mL of RPMI1640 solution (RPMI1640 culture medium) without drying.
  • the RPMI1640 solution was RPMI1640 medium supplemented with 10% feat-inactivated fetal bovine serum (FBS), 0.1 mM non-essential amino acids, 100 IU/ml penicillin, and 100 ⁇ g/mL streptomycin.
  • FBS feat-inactivated fetal bovine serum
  • a spleen cell suspension was prepared by gently mashing the cells with a slide glass so as not to damage the cells.
  • the splenocyte suspension was aspirated with a 5 mL pipette and passed through a cell strainer with a pore size of 40 ⁇ m placed on the mouth of a 50 mL centrifuge tube to remove connective tissue debris.
  • a 50 mL centrifuge tube was centrifuged at 270 xg (about 1,200 rpm) for 5 minutes to collect the cells at the bottom of the centrifuge tube, and the supernatant was removed by aspiration with an aspirator.
  • PBS phosphate-buffered saline
  • 10 mL of Tris-buffered ammonium chloride for erythrocyte removal that had been kept at room temperature in advance was added.
  • the cell pellet was loosened by gentle pipetting with a pipette, and the cells were completely suspended and allowed to react at room temperature for 10 minutes. Cells were pelleted by centrifugation at 270 xg for 5 minutes and the supernatant was aspirated off.
  • a cover glass was placed on the hemocytometer, and a cell suspension mixed with trypan blue was gently put into the gap between the counting board and the cover glass using a micropipette. This was observed under a microscope and the number of viable cells was counted. Based on the calculated cell density, the concentration of the cell suspension was adjusted to 1.0 ⁇ 10 6 cells/mL. Cells were placed in a 15 mL centrifuge tube at 8.0 ⁇ 10 6 cells/centrifuge tube, centrifuged at 270 ⁇ g for 5 minutes to precipitate the cells, and the supernatant was removed by aspiration.
  • RPMI1640 solutions containing 0-50 ng/mL of truncated mouse IL-36 ⁇ (SEQ ID NO: 12) were added with a 5 mL pipette to suspend the splenocytes.
  • Concentrations of truncated mouse IL-36 ⁇ were 50 nm/mL, 30 ng/mL, 10 ng/mL, 5 ng/mL, 1 ng/mL, 0.5 ng/mL, 0.1 ng/mL, 0.05 ng/mL, 0.01 ng /mL, and 0 ng/mL.
  • rat anti-mouse CD3 monoclonal antibody mAb
  • 2 mL of cell suspension was added for each concentration group of mouse IL-36 ⁇ . rice field.
  • rat anti-mouse CD28 mAb was added to a concentration of 2 ⁇ g/mL. Then, it was cultured for 48 hours under conditions of 5% CO 2 and 37°C.
  • splenocytes were collected from 2 wells of each group, and the number of cells was counted using a hemocytometer. Then added to Eppendorf tubes at 1.0 ⁇ 10 6 cells/tube. Cells were collected at the bottom of an Eppendorf tube by centrifugation at 270 ⁇ g (approximately 1,200 rpm) for 5 minutes, and the supernatant was removed by aspiration with an aspirator. After washing twice with 0.1% sodium azide-containing PBS, the cells were suspended in 100 ⁇ L of PBS. To this, rat anti-mouse CD16/32 mAb was added to block the Fc receptor.
  • APC-conjugated rat anti-mouse CD8a mAb, FITC-conjugated anti-mouse CD4 mAb, and PE-conjugated anti-mouse CD279 (PD-1) mAb or PE-conjugated rat bat IgG2b ⁇ Isotype Acon The reaction was allowed to proceed for 30 minutes at room temperature. After washing twice with 1 mL of PBS, the cells were suspended in 500 ⁇ L of PBS, and PD-1 expression in CD8-positive T cells and CD4-positive T cells was measured using a flow cytometer.
  • FIG. 1 shows splenocytes cultured in the presence of 0 ng/mL, 50 ng/mL, or 30 ng/mL IL-36 ⁇ .
  • “no stimulation” means that stimulation with CD3 mAb and CD28 mAb was not performed, and IL-36 ⁇ was not added.
  • FIG. 2 shows splenocytes cultured in the presence of 10 ng/mL, 5 ng/mL, or 1 ng/mL IL-36 ⁇ .
  • FIG. 3 shows the mean fluorescence intensity (MFI) of PE (PD-1 positive) in CD8-positive T cells and CD4-positive T cells in FIGS.
  • MFI mean fluorescence intensity
  • FIG. 4 shows splenocytes cultured in the presence of 0 ng/mL, 10 ng/mL, 1 ng/mL, or 0.5 ng/mL IL-36 ⁇ .
  • FIG. 5 shows splenocytes cultured in the presence of 0.1 ng/mL, 0.05 ng/mL, or 0.01 ng/mL IL-36 ⁇ .
  • FIG. 6 shows the mean fluorescence intensity (MFI) of PE (PD-1 positive) in the CD8-positive T cells of FIGS.
  • MFI mean fluorescence intensity
  • truncated IL-36 ⁇ suppresses the expression of PD-1 in activated T cells (especially activated CD8-positive T cells). PD-1 expression was strongly suppressed even at relatively low concentrations (eg, 5 ng/ml, 1 ng/ml, 0.5 ng/ml) of truncated IL-36 ⁇ .
  • Example 2 (Preparation of IL-36 ⁇ -expressing melanoma cells) According to the method described in JP-A-2021-70683, three retroviral vectors (DFG-IRES-Neo, DFG-PTH-truncated IL-36 ⁇ -IRES-Neo, DFG-GH-truncated IL-36 ⁇ -IRES- Neo) was produced.
  • DFG-IRES-Neo is a retroviral vector that expresses only the neomycin phosphotransferase gene (Neo).
  • DFG-PTH-truncated IL-36 ⁇ -IRES-Neo is a retroviral vector expressing truncated IL-36 ⁇ (SEQ ID NOS: 17, 18) to which a human parathyroid hormone signal sequence (SEQ ID NOS: 13, 14) is added and Neo. is.
  • DFG-GH-truncated IL-36 ⁇ -IRES-Neo is a retroviral vector that expresses truncated IL-36 ⁇ (SEQ ID NOS: 19 and 20) to which a human growth hormone signal sequence (SEQ ID NOS: 15 and 16) is added and Neo. be.
  • DFG-IRES-Neo, DFG-PTH-truncated IL-36 ⁇ -IRES-Neo, and DFG-GH-truncated IL-36 ⁇ -IRES-Neo were introduced into B16-F10 melanoma cells, respectively, to obtain B16-F10 Neo cells, B16-F10-PTH-truncated IL-36 ⁇ cells and B16-F10-GH-truncated IL-36 ⁇ cells were established, respectively.
  • the inoculum amount was 3 ⁇ 10 5 cells/mouse. Twelve days after cell inoculation, the spleen of each mouse was harvested.
  • a cover glass was placed on the hemocytometer, and a cell suspension mixed with trypan blue was gently put into the gap between the counting board and the cover glass using a micropipette. This was observed under a microscope and the number of viable cells was counted. Based on the calculated cell density, the concentration of the cell suspension was adjusted to 1.0 ⁇ 10 6 cells/mL, and added to the Eppendorf tube so that 1.0 ⁇ 10 6 cells/tube. Cells were collected at the bottom of an Eppendorf tube by centrifugation at 270 ⁇ g (approximately 1,200 rpm) for 5 minutes, and the supernatant was aspirated off with an aspirator. After washing twice with 0.1% sodium azide-containing PBS, the cells were suspended in 100 ⁇ L of PBS.
  • rat anti-mouse CD16/32 mAb was added to block the Fc receptor. Then, PE-conjugated anti-mouse CD279 (PD-1) mAb or PE-conjugated rat IgG2b ⁇ Isotype control mAb was added and allowed to react at room temperature for 30 minutes. After washing twice with 1 mL of PBS, the cells were suspended in 500 ⁇ L of PBS, and PD-1 expression in splenocytes was measured using a flow cytometer.
  • PD-1 expression in splenocytes was measured using a flow cytometer.
  • Example 3 (Generation of IL-36 ⁇ -expressing tumor cells) DFG-IRES-Neo, DFG-PTH-truncated IL-36 ⁇ -IRES-Neo, and DFG-GH-truncated IL-36 ⁇ -IRES-Neo were each introduced into MCA205 cells (mouse fibrosarcoma cells) to obtain MCA205Neo cells. , MCA205-PTH-truncated IL-36 ⁇ cells, and MCA205-GH-truncated IL-36 ⁇ cells were established, respectively.
  • MCA205-PTH-truncated IL-36 ⁇ cells MCA205-GH-truncated IL-36 ⁇ cells were established, respectively.
  • DFG-IRES-Neo, DFG-PTH-truncated IL-36 ⁇ -IRES-Neo, and DFG-GH-truncated IL-36 ⁇ -IRES-Neo were each introduced into MC38 cells (mouse colon adenocarcinoma cells) to obtain MC38Neo. cells, MC38-PTH-truncated IL-36 ⁇ cells, and MC38-GH-truncated IL-36 ⁇ cells were established, respectively.
  • the inoculum amount was 1.5 ⁇ 10 5 cells/mouse.
  • the inoculum amount was 2 ⁇ 10 5 cells/mouse. Twelve days after inoculation, tumor tissue was harvested from each mouse.
  • the collected tumor tissue was immersed in a PBS-buffered formalin solution for 2 days and then embedded in paraffin. Sections were made from deparaffinized tissue for PD-1 staining. After treating this with normal goat serum, it was allowed to react with goat anti-mouse PD-1 antibody at 4° C. overnight. After washing with PBS, a biotinylated bovine anti-goat IgG antibody was reacted for 30 minutes. After washing with PBS, a solution containing peroxidase-conjugated streptavidin was applied. After washing with PBS, the color was developed with diaminobenzidine.
  • FIG. 8 shows the results of immunostaining.
  • PD-1 expression was significantly higher in tumor tissue collected from MCA205-PTH-truncated IL-36 ⁇ cell-inoculated mice or MCA205-GH-truncated IL-36 ⁇ cell-inoculated mice compared to tumor tissue collected from MCA205Neo cell-inoculated mice. It was clearly lowered (Fig. 8 upper).
  • PD-1 expression was significantly higher in tumor tissue collected from MC38-PTH-truncated IL-36 ⁇ cell-inoculated mice or MC38-GH-truncated IL-36 ⁇ cell-inoculated mice compared to tumor tissue collected from MC38Neo cell-inoculated mice.
  • a PD-1 expression inhibitor a pharmaceutical composition for immune checkpoint inhibition, and a method for inhibiting PD-1 expression are provided, which are capable of suppressing PD-1 expression in lymphocytes.

Abstract

This PD-1 expression inhibitor contains at least one component selected from the group consisting of the following (a) through (d): (a) interleukin 36 or a modified form thereof; (b) a polynucleotide having a nucleotide sequence encoding either interleukin 36 or a modified form thereof; (c) a vector containing the polynucleotide described in the abovementioned (b); and (d) cells that secrete either interleukin 36 or a modified form thereof.

Description

PD-1発現抑制剤、免疫チェックポイント阻害用医薬組成物、及びPD-1発現を抑制する方法PD-1 expression inhibitor, pharmaceutical composition for immune checkpoint inhibition, and method for inhibiting PD-1 expression
 本発明は、PD-1発現抑制剤、免疫チェックポイント阻害用医薬組成物、及びPD-1発現を抑制する方法に関する。 The present invention relates to a PD-1 expression inhibitor, a pharmaceutical composition for immune checkpoint inhibition, and a method for inhibiting PD-1 expression.
 がんは、日本人の死因の第一位であり、世界的にも患者数は多い。そのため、より良い治療薬の開発が望まれている。近年、がんの治療薬として、PD-1(Programmed cell death 1)等の免疫チェックポイント分子の働きを阻害する免疫チェックポイント阻害剤が導入されている(特許文献1)。
 PD-1は、細胞傷害性T細胞(CTL)等の活性化リンパ球の表面に発現する分子である。PD-1が、がん細胞表面のリガンド(PD-L1など)と結合すると、CTLの活性化が抑制され、がん細胞に対する細胞傷害活性が抑制されることが知られている。PD-1を標的とする免疫チェックポイント阻害剤は、PD-1に対するリガンドの結合を阻害することにより、CTLの細胞傷害活性の低下を阻害する。
Cancer is the number one cause of death in Japan, and there are many patients worldwide. Therefore, development of a better therapeutic agent is desired. In recent years, immune checkpoint inhibitors that inhibit the action of immune checkpoint molecules such as PD-1 (programmed cell death 1) have been introduced as therapeutic agents for cancer (Patent Document 1).
PD-1 is a molecule expressed on the surface of activated lymphocytes such as cytotoxic T cells (CTL). It is known that when PD-1 binds to a cancer cell surface ligand (such as PD-L1), CTL activation is suppressed, and cytotoxic activity against cancer cells is suppressed. Immune checkpoint inhibitors targeting PD-1 inhibit the reduction of cytotoxic activity of CTL by inhibiting ligand binding to PD-1.
 インターロイキン36(IL-36)は、DNAデータベースの解析により、IL-1に相同性を有する分子として報告されたサイトカインである。IL-36は、IL-1、IL-18、IL-33、IL-37、及びIL-38と共に、IL-1サイトカインファミリーを構成している。IL-36は、特定の産生細胞から分泌される分泌タンパク質であるが、IL-36遺伝子は、シグナル配列に該当する配列を有しておらず、分泌メカニズムは解明されていない。IL-36の受容体は、IL-36受容体(IL-36R)とIL-1アクセサリータンパク質(IL-1RAcP)とのヘテロ2量体からなる。 Interleukin 36 (IL-36) is a cytokine reported as a molecule with homology to IL-1 by DNA database analysis. IL-36, along with IL-1, IL-18, IL-33, IL-37, and IL-38, make up the IL-1 cytokine family. IL-36 is a secretory protein that is secreted from specific producing cells, but the IL-36 gene does not have a sequence corresponding to a signal sequence, and the secretion mechanism has not been elucidated. The receptor for IL-36 consists of a heterodimer of IL-36 receptor (IL-36R) and IL-1 accessory protein (IL-1RAcP).
 IL-36には、スプライスバリアントとして、IL-36α、IL-36β及びIL-36γの3つのアイソフォームがある。IL-36βは、主に、上皮細胞、単球及びB細胞等から産生される。N末端の30個のアミノ酸が除去されたtrundated型のIL-36βは、天然型の全長IL-36βと比較して、約1000倍強い活性を有することが報告されている(非特許文献1)。IL-36βは、樹状細胞に作用して、IL-6、IL-12及びGM-CSF等の産生を増強し、細胞表面のMHC class II、CD80及びCD86等の発現を増強することが報告されている(非特許文献2)。 IL-36 has three isoforms, IL-36α, IL-36β and IL-36γ, as splice variants. IL-36β is mainly produced by epithelial cells, monocytes, B cells and the like. It has been reported that truncated IL-36β, in which 30 amino acids at the N-terminus have been removed, has about 1000-fold stronger activity than native full-length IL-36β (Non-Patent Document 1). . It has been reported that IL-36β acts on dendritic cells to enhance the production of IL-6, IL-12, GM-CSF, etc., and enhances the expression of cell surface MHC class II, CD80, CD86, etc. (Non-Patent Document 2).
国際公開第2004/004771号WO2004/004771
 免疫チェックポイント阻害剤としては、PD-1等の免疫チェックポイント分子に対するモノクローナル抗体が用いられている。がん治療の選択肢を広げるためには、異なるメカニズムで、免疫チェックポイント分子の機能を抑制し得る薬剤の開発が求められる。 Monoclonal antibodies against immune checkpoint molecules such as PD-1 are used as immune checkpoint inhibitors. In order to expand the options for cancer treatment, it is necessary to develop drugs that can suppress the function of immune checkpoint molecules by different mechanisms.
 そこで、本発明は、リンパ球におけるPD-1の発現を抑制可能な、PD-1発現抑制剤、免疫チェックポイント阻害用医薬組成物、及びPD-1発現を抑制する方法を提供することを課題とする。 Therefore, an object of the present invention is to provide a PD-1 expression suppressing agent, a pharmaceutical composition for immune checkpoint inhibition, and a method for suppressing PD-1 expression that can suppress the expression of PD-1 in lymphocytes. and
 本開示は、以下の態様を含む。
[1]下記(a)~(d)からなる群より選択される少なくとも1種の成分を含む、PD-1発現抑制剤:(a)インターロイキン36又はその改変体;
 (b)インターロイキン36又はその改変体をコードするヌクレオチド配列を有するポリヌクレオチド;(c)前記(b)のポリヌクレオチドを含むベクター;及び(d)インターロイキン36又はその改変体を分泌する細胞。
[2]CD8陽性T細胞又はCD4陽性T細胞におけるPD-1の発現を抑制する、[1]に記載のPD-1発現抑制剤。
[3][1]又は[2]に記載のPD-1発現抑制剤と、薬学的に許容される担体とを含有する、免疫チェックポイント阻害用医薬組成物。
[4]リンパ球を、インターロイキン36又はその改変体の存在下で培養することを含む、リンパ球におけるPD-1発現を抑制する方法。
The present disclosure includes the following aspects.
[1] A PD-1 expression inhibitor comprising at least one component selected from the group consisting of the following (a) to (d): (a) interleukin 36 or a variant thereof;
(b) a polynucleotide having a nucleotide sequence encoding interleukin-36 or a variant thereof; (c) a vector comprising the polynucleotide of (b); and (d) a cell that secretes interleukin-36 or a variant thereof.
[2] The PD-1 expression inhibitor according to [1], which suppresses PD-1 expression in CD8-positive T cells or CD4-positive T cells.
[3] A pharmaceutical composition for immune checkpoint inhibition, comprising the PD-1 expression inhibitor of [1] or [2] and a pharmaceutically acceptable carrier.
[4] A method of suppressing PD-1 expression in lymphocytes, comprising culturing lymphocytes in the presence of interleukin 36 or a variant thereof.
 本発明によれば、リンパ球におけるPD-1の発現を抑制可能な、PD-1発現抑制剤、免疫チェックポイント阻害用医薬組成物、及びPD-1発現を抑制する方法が提供される。 According to the present invention, a PD-1 expression inhibitor, a pharmaceutical composition for immune checkpoint inhibition, and a method for inhibiting PD-1 expression are provided, which are capable of suppressing PD-1 expression in lymphocytes.
CD8陽性T細胞及びCD4陽性T細胞において、フローサイトメトリーにより、truncated マウスIL-36βによるPD1発現抑制効果を解析した結果を示す。FIG. 4 shows the results of analyzing the PD1 expression inhibitory effect of truncated mouse IL-36β on CD8-positive T cells and CD4-positive T cells by flow cytometry. CD8陽性T細胞及びCD4陽性T細胞において、フローサイトメトリーにより、truncated マウスIL-36βによるPD1発現抑制効果を解析した結果を示す。FIG. 4 shows the results of analyzing the PD1 expression inhibitory effect of truncated mouse IL-36β on CD8-positive T cells and CD4-positive T cells by flow cytometry. 図1及び図2のフローサイトメトリー解析結果に基づく、PE(PD-1陽性)の平均蛍光強度(Mean Fluorescence Intensity:MFI)を示す。FIG. 2 shows the mean fluorescence intensity (MFI) of PE (PD-1 positive) based on the flow cytometry analysis results of FIGS. 1 and 2. FIG. CD8陽性T細胞及びCD4陽性T細胞において、フローサイトメトリーにより、truncated マウスIL-36βによるPD1発現抑制効果を解析した結果を示す。FIG. 4 shows the results of analyzing the PD1 expression inhibitory effect of truncated mouse IL-36β on CD8-positive T cells and CD4-positive T cells by flow cytometry. CD8陽性T細胞及びCD4陽性T細胞において、フローサイトメトリーにより、truncated マウスIL-36βによるPD1発現抑制効果を解析した結果を示す。FIG. 4 shows the results of analyzing the PD1 expression inhibitory effect of truncated mouse IL-36β on CD8-positive T cells and CD4-positive T cells by flow cytometry. 図4及び図5のフローサイトメトリー解析結果に基づく、PE(PD-1陽性)の平均蛍光強度(Mean Fluorescence Intensity:MFI)を示す。CD8陽性T細胞における結果を示す。FIG. 5 shows the mean fluorescence intensity (MFI) of PE (PD-1 positive) based on the flow cytometry analysis results of FIGS. 4 and 5. FIG. Results for CD8-positive T cells are shown. B16-F10Neo細胞、B16-F10-PTH-truncated IL-36β細胞、又はB16-F10-GH-truncated IL-36β細胞を移植したマウスの脾細胞を採取し、フローサイトメトリーにより、PD-1発現を解析した結果を示す。Splenocytes of mice transplanted with B16-F10Neo cells, B16-F10-PTH-truncated IL-36β cells, or B16-F10-GH-truncated IL-36β cells were harvested, and PD-1 expression was analyzed by flow cytometry. Analyzed results are shown. MCA205Neo細胞、MCA205-PTH-truncated IL-36β細胞、MCA205-GH-truncated IL-36β細胞、MC38Neo細胞、MC38-PTH-truncated IL-36β細胞、又はMC38-GH-truncated IL-36β細胞を接種後12日目の腫瘍組織のPD-1分子の免疫染色の結果を示す。12 after inoculation of MCA205 Neo cells, MCA205-PTH-truncated IL-36β cells, MCA205-GH-truncated IL-36β cells, MC38Neo cells, MC38-PTH-truncated IL-36β cells, or MC38-GH-truncated IL-36β cells The results of PD-1 molecule immunostaining of day-old tumor tissues are shown.
 「を含む」(comprise)という用語は、対象となる構成要素以外の構成要素を含んでいてもよいことを意味する。「からなる」(consist of)という用語は、対象となる構成要素以外の構成要素を含まないことを意味する。「から本質的になる」(consist essentially of)という用語は、対象となる構成要素以外の構成要素を特別な機能を発揮する態様(発明の効果を完全に喪失させる態様など)では含まないことを意味する。本明細書において、「を含む」(comprise)と記載する場合、「からなる」(consist of)態様、及び「から本質的になる」(consist essentially of)態様を包含する。 The term "comprise" means that it may include components other than the target components. The term "consist of" means containing no elements other than the subject element. The term "consistently of" means that it does not include constituent elements other than the subject constituent elements in a mode that exhibits a special function (such as a mode that completely loses the effects of the invention). means. As used herein, the term "comprise" includes aspects that "consist of" and aspects that "consist essentially of."
 タンパク質、ペプチド、ポリヌクレオチド(DNA、RNA)、ベクター、及び細胞は、単離されたものであり得る。「単離された」とは、天然状態又は他の成分から分離された状態を意味する。「単離された」ものは、他の成分を実質的に含まないものであり得る。「他の成分を実質的に含まない」とは、単離された成分に含まれる他の成分の含有量が無視できる程度であることを意味する。単離された成分に含まれる他の成分の含有量は、例えば、10質量%以下、5質量%以下、4質量%以下、3質量%以下、2質量%以下、1質量%以下、0.5質量%以下、又は0.1質量%以下であり得る。本明細書に記載されるタンパク質、ペプチド、ポリヌクレオチド(DNA、RNA)、ベクター、及び細胞は、単離されたタンパク質、単離されたペプチド、単離されたポリヌクレオチド(単離されたDNA、単離されたRNA)、単離されたベクター、及び単離された細胞であり得る。 Proteins, peptides, polynucleotides (DNA, RNA), vectors, and cells can be isolated. "Isolated" means separated from the natural state or other components. "Isolated" can be substantially free of other components. "Substantially free of other components" means that the content of other components contained in the isolated component is negligible. The content of other components contained in the isolated component is, for example, 10% by mass or less, 5% by mass or less, 4% by mass or less, 3% by mass or less, 2% by mass or less, 1% by mass or less, 0.5% by mass or less. It may be 5% by mass or less, or 0.1% by mass or less. The proteins, peptides, polynucleotides (DNA, RNA), vectors and cells described herein may be isolated proteins, isolated peptides, isolated polynucleotides (isolated DNA, isolated RNA), isolated vectors, and isolated cells.
 「ポリヌクレオチド」は、ヌクレオチドがホスホジエステル結合によって結合したヌクレオチドポリマーを指す。「ポリヌクレオチド」は、DNAであってもよく、RNAであってもよく、DNAとRNAとの組み合わせから構成されてもよい。「ポリヌクレオチド」は、天然ヌクレオチドのポリマーであってもよく、天然ヌクレオチドと非天然ヌクレオチド(天然ヌクレオチドの類似体、塩基部分、糖部分及びリン酸部分のうち少なくとも一つの部分が修飾されているヌクレオチド(例えば、ホスホロチオエート骨格)等)とのポリマーであってもよく、非天然ヌクレオチドのポリマーであってもよい。
 ポリヌクレオチドのヌクレオチド配列は、特に明示しない限り、一般的に認められている1文字コードで記載される。特に明示しない限り、ヌクレオチド配列は、5’側から3’側に向かって記載する。ポリヌクレオチドを構成するヌクレオチド残基は、単に、アデニン、チミン、シトシン、グアニン、又はウラシル等、あるいはそれらの1文字コードで記載される場合がある。
"Polynucleotide" refers to a nucleotide polymer in which nucleotides are linked by phosphodiester bonds. A "polynucleotide" may be DNA, RNA, or may be composed of a combination of DNA and RNA. A “polynucleotide” may be a polymer of natural nucleotides, including natural nucleotides and non-natural nucleotides (analogs of natural nucleotides, nucleotides in which at least one of the base, sugar and phosphate moieties is modified). (for example, a phosphorothioate skeleton), etc.), or a polymer of non-natural nucleotides.
Nucleotide sequences of polynucleotides are written in the generally accepted single-letter code unless otherwise indicated. Unless otherwise indicated, nucleotide sequences are written 5' to 3'. Nucleotide residues constituting a polynucleotide may be simply described as adenine, thymine, cytosine, guanine, uracil, or the like, or their one-letter codes.
 「遺伝子」は、特定のタンパク質をコードする少なくとも1つのオープンリーディングフレームを含むポリヌクレオチドを指す。遺伝子は、エクソン及びイントロンの両方を含み得る。 "Gene" refers to a polynucleotide containing at least one open reading frame that encodes a particular protein. A gene may contain both exons and introns.
 「ペプチド」、「ポリペプチド」及び「タンパク質」という用語は、相互に互換的に使用され、アミド結合によって結合したアミノ酸のポリマーを指す。「ペプチド」、「ポリペプチド」及び「タンパク質」は、天然アミノ酸のポリマーであってもよく、天然アミノ酸と非天然アミノ酸(天然アミノ酸の化学的類似体、修飾誘導体等)とのポリマーであってもよく、非天然アミノ酸のポリマーであってもよい。
 アミノ酸配列は、特に明示しない限り、一般的に認められている1文字コード又は3文字コードで記載される。特に明示しない限り、アミノ酸配列は、N末端側からC末端側に向かって記載する。
The terms "peptide", "polypeptide" and "protein" are used interchangeably to refer to a polymer of amino acids linked by amide bonds. "Peptides", "polypeptides" and "proteins" may be polymers of naturally occurring amino acids or of naturally occurring and non-natural amino acids (chemical analogues, modified derivatives, etc. of naturally occurring amino acids). It may also be a polymer of unnatural amino acids.
Amino acid sequences are written in the generally accepted one-letter or three-letter code unless otherwise specified. Unless otherwise specified, amino acid sequences are written from the N-terminal side to the C-terminal side.
 ポリヌクレオチドに関して用いる「作動可能に連結」という用語は、第1のヌクレオチド配列が第2のヌクレオチド配列の十分に近くに配置され、第1のヌクレオチド配列が第2のヌクレオチド配列又は第2のヌクレオチド配列の制御下の領域に影響を及ぼしうることを意味する。例えば、あるポリヌクレオチドがプロモーターに作動可能に連結するとは、当該ポリヌクレオチドが、当該プロモーターの制御下で発現するように連結されていることを意味する。
 「発現可能な状態」という用語は、対象遺伝子が導入された細胞内で、該対象遺伝子が転写され得る状態にあることを指す。
 「発現ベクター」という用語は、対象遺伝子を含むベクターであって、該ベクターを導入した細胞内で、対象遺伝子を発現可能な状態にするシステムを備えたベクターを指す。
 「プロモーターが機能し得る」とは、対象生物の細胞内において、当該プロモーターに作動可能に連結されたポリヌクレオチドを発現させることができることを意味する。
 「インフレーム」とは、コドンの読み枠がずれないことを意味する。
The term "operably linked" when used in reference to polynucleotides means that a first nucleotide sequence is placed sufficiently close to a second nucleotide sequence such that the first nucleotide sequence is either the second nucleotide sequence or the second nucleotide sequence. means that it can affect areas under the control of For example, that a polynucleotide is operably linked to a promoter means that the polynucleotide is linked so that it is expressed under the control of the promoter.
The term "expressible state" refers to a state in which a gene of interest can be transcribed in a cell into which the gene of interest has been introduced.
The term "expression vector" refers to a vector containing a gene of interest and equipped with a system that renders the gene of interest expressible in cells into which the vector has been introduced.
“A promoter can function” means that a polynucleotide operably linked to the promoter can be expressed in the cells of the subject organism.
"In-frame" means that the reading frame of the codon is not shifted.
 アミノ酸配列又はヌクレオチド配列どうしの「配列同一性」は、2つのアミノ酸配列又はヌクレオチド配列を、対応するアミノ酸又は塩基が最も多く一致するように、挿入及び欠失に当たる部分にギャップを入れながら並置し、得られたアライメント中のギャップを除くアミノ酸配列全体又はヌクレオチド配列全体に対する一致したアミノ酸又は塩基の割合として求められる。アミノ酸配列同士又はヌクレオチド配列同士の配列同一性は、当該技術分野で公知の各種相同性検索ソフトウェアを用いて求めることができる。例えば、アミノ酸配列の配列同一性の値は、公知の相同性検索ソフトウェアBLASTPにより得られたアライメントを元にした計算によって得ることができ、ヌクレオチド配列の配列同一性の値は、公知の相同性検索ソフトウェアBLASTNにより得られたアライメントを元にした計算によって得ることができる。配列同一性は、相同性ともいう。 "Sequence identity" between amino acid sequences or nucleotide sequences is defined by aligning the two amino acid sequences or nucleotide sequences so that the corresponding amino acids or bases match the most, leaving gaps for insertions and deletions, It is determined as the ratio of matched amino acids or bases to the entire amino acid sequence or nucleotide sequence excluding gaps in the resulting alignment. Sequence identity between amino acid sequences or between nucleotide sequences can be determined using various homology search software known in the art. For example, the sequence identity value of an amino acid sequence can be obtained by calculation based on alignments obtained by the known homology search software BLASTP, and the sequence identity value of a nucleotide sequence can be obtained by a known homology search. It can be obtained by calculation based on alignments obtained by the software BLASTN. Sequence identity is also referred to as homology.
 「ストリンジェントな条件」とは、配列同一性が高い2個のポリヌクレオチドが、特異的にハイブリダイズ可能な条件を意味する。配列同一性が高い2個のポリヌクレオチドとは、前記2個のポリヌクレオチド間の配列同一性が、例えば、80%以上、85%以上、90%以上、95%以上、96%以上、97%以上、98%以上、又は99%以上であることをいう。ストリンジェントな条件は、例えば、Molecular Cloning-A LABORATORY MANUAL THIRD EDITION(Sambrook et al., Cold Spring Harbor Laboratory Press)に記載の条件等が挙げられる。ストリンジェントな条件の具体例としては、例えば、6×SSC(20×SSCの組成:3M塩化ナトリウム、0.3Mクエン酸溶液、pH7.0)、5×デンハルト溶液(100×デンハルト溶液の組成:2質量%ウシ血清アルブミン、2質量%フィコール、2質量%ポリビニルピロリドン)、0.5質量%のSDS、0.1mg/mLサケ精子DNA、及び50%フォルムアミドからなるハイブリダイゼーションバッファー中で、42~70℃で数時間から一晩インキュベーションを行う条件が挙げられる。インキュベーション後の洗浄に用いる洗浄バッファーとしては、例えば、0.1質量%SDS含有1×SSC溶液、及び0.1質量%SDS含有0.1×SSC溶液が挙げられる。 "Stringent conditions" means conditions under which two polynucleotides with high sequence identity can specifically hybridize. Two polynucleotides with high sequence identity, the sequence identity between the two polynucleotides, for example, 80% or more, 85% or more, 90% or more, 95% or more, 96% or more, 97% Above, 98% or more, or 99% or more. Stringent conditions include, for example, conditions described in Molecular Cloning-A Laboratory Manual Third Edition (Sambrook et al., Cold Spring Harbor Laboratory Press). Specific examples of stringent conditions include, for example, 6 x SSC (composition of 20 x SSC: 3 M sodium chloride, 0.3 M citric acid solution, pH 7.0), 5 x Denhardt's solution (composition of 100 x Denhardt's solution: 42 in a hybridization buffer consisting of 2% by weight bovine serum albumin, 2% by weight Ficoll, 2% by weight polyvinylpyrrolidone), 0.5% by weight SDS, 0.1 mg/mL salmon sperm DNA, and 50% formamide. Conditions include incubation at ~70°C for several hours to overnight. Washing buffers used for washing after incubation include, for example, 0.1% by mass SDS-containing 1×SSC solution and 0.1% by mass SDS-containing 0.1×SSC solution.
[PD-1発現抑制剤]
 本開示の第1の態様は、PD-1発現抑制剤である。前記PD-1発現抑制剤は、下記(a)~(d)からなる群より選択される少なくとも1種の成分を含む:
 (a)IL-36又はその改変体;
 (b)IL-36又はその改変体をコードするヌクレオチド配列を有するポリヌクレオチド;
 (c)前記(b)のポリヌクレオチドを含むベクター;及び
 (d)IL-36又はその改変体を分泌する細胞。
[PD-1 expression inhibitor]
A first aspect of the present disclosure is a PD-1 expression inhibitor. The PD-1 expression inhibitor contains at least one component selected from the group consisting of the following (a) to (d):
(a) IL-36 or variant thereof;
(b) a polynucleotide having a nucleotide sequence encoding IL-36 or a variant thereof;
(c) a vector comprising the polynucleotide of (b); and (d) a cell secreting IL-36 or a variant thereof.
 後述する実施例で示すように、IL-36の存在下で培養されたリンパ球は、IL-36の非存在下で培養されたリンパ球と比較して、PD-1の発現が抑制されることが確認された。また、IL-36を産生する腫瘍細胞を移植したマウスのリンパ球では、IL-36を産生しない腫瘍細胞を移植したマウスのリンパ球と比較して、PD-1の発現が抑制されることが確認された。これらの結果は、in vitro及びin vivoにおいて、リンパ球のPD-1発現が、IL-36により抑制されることを示す。したがって、前記(a)~(d)の各成分は、PD-1発現抑制剤として用いることができる。 As shown in the examples below, PD-1 expression is suppressed in lymphocytes cultured in the presence of IL-36 compared to lymphocytes cultured in the absence of IL-36. was confirmed. In lymphocytes from mice transplanted with tumor cells that produce IL-36, the expression of PD-1 is suppressed compared to lymphocytes from mice transplanted with tumor cells that do not produce IL-36. confirmed. These results demonstrate that lymphocyte PD-1 expression is suppressed by IL-36 in vitro and in vivo. Therefore, each of the above components (a) to (d) can be used as a PD-1 expression inhibitor.
 本実施形態のPD-1発現抑制剤は、リンパ球を培養する培地に添加して、PD-1発現が抑制されたリンパ球をin vitro又はex vivoで調製するために用いることができる。PD-1発現抑制の対象となるリンパ球は、PD-1を発現する限り、特に限定されない。PD-1発現抑制の対象となるリンパ球としては、例えば、T細胞、ナチュラルキラー細胞等が挙げられる。T細胞としては、CD8陽性T細胞(CTL等)、CD4陽性T細胞(ヘルパーT細胞等)等が挙げられる。リンパ球が由来する生物は、特に限定されないが、例えば、ヒト、及びヒト以外の哺乳類が挙げられる。ヒト以外の哺乳類としては、例えば、霊長類(サル、チンパンジー、ゴリラなど)、げっ歯類(マウス、ハムスター、ラットなど)、ウサギ、イヌ、ネコ、ウシ、ヤギ、ヒツジ、ウマ、ブタ等が挙げられるが、これらに限定されない。
 リンパ球は、遺伝子改変されたものでもよい。遺伝子改変の種類は、特に限定されない。遺伝子改変されたリンパ球の具体例としては、キメラ抗原受容体を発現するように改変されたリンパ球(CAR-T細胞等)、外因性TCRを発現するように改変されたリンパ球(TCR-T細胞等)等が挙げられる。
The PD-1 expression inhibitor of this embodiment can be added to a medium for culturing lymphocytes and used to prepare lymphocytes in which PD-1 expression is suppressed in vitro or ex vivo. Lymphocytes targeted for suppression of PD-1 expression are not particularly limited as long as they express PD-1. Lymphocytes targeted for suppression of PD-1 expression include, for example, T cells and natural killer cells. Examples of T cells include CD8-positive T cells (CTL and the like), CD4-positive T cells (helper T cells and the like), and the like. Organisms from which lymphocytes are derived are not particularly limited, but include, for example, humans and mammals other than humans. Mammals other than humans include, for example, primates (monkeys, chimpanzees, gorillas, etc.), rodents (mice, hamsters, rats, etc.), rabbits, dogs, cats, cows, goats, sheep, horses, pigs, and the like. include but are not limited to:
Lymphocytes may be genetically modified. The type of genetic modification is not particularly limited. Specific examples of genetically modified lymphocytes include lymphocytes modified to express chimeric antigen receptors (CAR-T cells, etc.), lymphocytes modified to express exogenous TCR (TCR- T cells, etc.) and the like.
 本実施形態のPD-1発現抑制剤は、生体に投与して、in vivoで、生体内に存在するリンパ球のPD-1発現を抑制するために用いてもよい。PD-1抑制剤の投与対象となる生物は、PD-1を発現するリンパ球を有する限り、特に限定されない。投与対象の生物としては、上記と同様の生物が挙げられる。 The PD-1 expression inhibitor of the present embodiment may be administered to a living body and used in vivo to suppress PD-1 expression of lymphocytes present in the living body. The organism to which the PD-1 inhibitor is administered is not particularly limited as long as it has lymphocytes expressing PD-1. Organisms to be administered include the same organisms as those described above.
<(a)IL-36>
 IL-36は、IL-1サイトカインファミリーに属するサイトカインである。IL-36には、スプライスバリアントとして、IL-36α、IL-36β及びIL-36γの3つのアイソフォームがある。IL-36は、IL-36α、IL-36β、及びIL-36γのいずれであってもよいが、IL-36βが好ましい。
<(a) IL-36>
IL-36 is a cytokine belonging to the IL-1 cytokine family. IL-36 has three isoforms, IL-36α, IL-36β and IL-36γ, as splice variants. IL-36 may be any of IL-36α, IL-36β, and IL-36γ, with IL-36β being preferred.
 IL-36が由来する生物は、特に限定されないが、PD-1発現の抑制対象であるリンパ球が由来する生物と同種であることが好ましい。例えば、PD-1発現の抑制対象がヒトリンパ球である場合、ヒトIL-36を用いることが好ましい。PD-1発現の抑制対象がマウスリンパ球である場合、マウスIL-36を用いることが好ましい。 Although the organism from which IL-36 is derived is not particularly limited, it is preferably the same species as the organism from which the lymphocytes whose PD-1 expression is to be suppressed are derived. For example, when human lymphocytes are targeted for suppression of PD-1 expression, it is preferable to use human IL-36. Mouse IL-36 is preferably used when the target for suppression of PD-1 expression is mouse lymphocytes.
 IL-36は、天然型であってもよく、改変型であってもよい。天然型IL-36は、自然界で産生されるIL-36である。天然型IL-36は、例えば、上記のような哺乳類の体内で産生されるIL-36である。天然型IL-36の具体例としては、ヒトIL-36、及びマウスIL-36が挙げられる。
 ヒトIL-36β(NCBI Gene ID:27177)のアミノ酸配列としては、例えば、配列番号2に記載のアミノ酸配列(ヌクレオチド配列:配列番号1)、配列番号4に記載のアミノ酸配列(ヌクレオチド配列:配列番号3)等が挙げられる。
 マウスIL-36β(NCBI Gene ID:69677)のアミノ酸配列としては、例えば、配列番号6に記載のアミノ酸配列(ヌクレオチド配列:配列番号5)等が挙げられる。天然型IL-36は、前記アミノ酸配列を有するものに限定されず、それらのホモログ(オーソログ又はパラログ)であってもよい。天然型IL-36のアミノ酸配列は、GenBank等の配列データベースから取得することができる。
IL-36 may be naturally occurring or modified. Native IL-36 is IL-36 produced in nature. Native IL-36 is, for example, IL-36 produced in mammals as described above. Specific examples of native IL-36 include human IL-36 and mouse IL-36.
Examples of the amino acid sequence of human IL-36β (NCBI Gene ID: 27177) include the amino acid sequence described in SEQ ID NO: 2 (nucleotide sequence: SEQ ID NO: 1), the amino acid sequence described in SEQ ID NO: 4 (nucleotide sequence: SEQ ID NO: 3) and the like.
The amino acid sequence of mouse IL-36β (NCBI Gene ID: 69677) includes, for example, the amino acid sequence set forth in SEQ ID NO: 6 (nucleotide sequence: SEQ ID NO: 5). Native IL-36 is not limited to those having the above amino acid sequences, and may be homologues (orthologs or paralogs) thereof. The amino acid sequence of native IL-36 can be obtained from sequence databases such as GenBank.
 改変型IL-36(以下、「IL-36の改変体」ともいう)は、天然型IL-36が改変されたポリペプチドであって、天然型IL-36の機能を維持するポリペプチドである。IL-36の改変体は、天然型IL-36のアミノ酸配列と比較して、1個以上のアミノ酸が改変(置換、欠失、付加、挿入、又はこれらの組合せ)されたアミノ酸配列を有する。改変されるアミノ酸の数は、特に限定されないが、例えば、1~50個、1~40個、1~30個、1~20個、1~10個、又は1~5個、1~4個、1~3個、又は1~2個等が挙げられる。改変型IL-36は、天然型IL-36のアミノ酸配列と80%以上の配列同一性を有するアミノ酸配列を有するものであってもよい。配列同一性は、例えば、85%以上、90%以上、95%以上、96%以上、97%以上、98%以上、又は99%以上であってもよい。 Modified IL-36 (hereinafter also referred to as "variants of IL-36") is a polypeptide obtained by modifying native IL-36, and is a polypeptide that maintains the function of native IL-36. . A variant of IL-36 has an amino acid sequence in which one or more amino acids are altered (substitution, deletion, addition, insertion, or a combination thereof) as compared with the amino acid sequence of native IL-36. The number of amino acids to be modified is not particularly limited. , 1 to 3, or 1 to 2, and the like. Modified IL-36 may have an amino acid sequence with 80% or more sequence identity with the amino acid sequence of native IL-36. Sequence identity can be, for example, 85% or greater, 90% or greater, 95% or greater, 96% or greater, 97% or greater, 98% or greater, or 99% or greater.
 IL-36の改変体は、天然型IL-36が有する少なくとも1つの活性について、天然型IL-36と同等以上の活性を有することが好ましい。天然型IL-36の活性(以下、「IL-36活性」ともいう)としては、例えば、リンパ球のPD-1発現を抑制する活性が挙げられる。IL-36活性は、IL-36RとIL-1RAcpのヘテロ2量体からなる受容体に対する結合活性、前記受容体を有する細胞(例えば、肺線維芽細胞)にIL-6を産生させる活性、及び抗腫瘍活性等であってもよい。天然型IL-36βと同等以上の活性を有するIL-36の改変体は、天然型IL-36βが示す活性を100としたとき、例えば、80以上、85以上、90以上、又は95以上の相対活性を示す。 The variant of IL-36 preferably has at least one activity of native IL-36 that is equal to or greater than that of native IL-36. The activity of native IL-36 (hereinafter also referred to as “IL-36 activity”) includes, for example, the activity of suppressing PD-1 expression in lymphocytes. IL-36 activity includes binding activity to a receptor consisting of a heterodimer of IL-36R and IL-1RAcp, activity to cause cells having the receptor (e.g., lung fibroblasts) to produce IL-6, and It may be antitumor activity or the like. A variant of IL-36 having an activity equal to or greater than that of native IL-36β has a relative activity of, for example, 80 or more, 85 or more, 90 or more, or 95 or more when the activity exhibited by natural IL-36β is taken as 100. It shows activity.
 マウスIL-36βでは、N末端の30個のアミノ酸を除去した改変体が、天然型IL-36βと比較して、約1000倍強い活性を示すことが報告されている(Towne JE, et al., J Biol Chem. 2011 Dec 9; 286(49):42594-602.)。ヒトIL-36βでは、N末端の4個のアミノ酸を除去した改変体が、天然型IL-36βと比較して、強い活性を示すことが報告されている(Towne JE, et al., J Biol Chem. 2011 Dec 9; 286(49):42594-602.)。したがって、IL-36βの改変体としては、天然型IL-36βのN末端領域のアミノ酸が除去されたもの(以下、「truncated IL-36β」ともいう)が挙げられる。除去されるN末端アミノ酸の数の上限値としては、例えば、40個以下、35個以下、30個以下、25個以下、20個以下、15個以下、10個以下、5個以下、又は4個以下が挙げられる。除去されるN末端アミノ酸の数の下限値は、1個以上である。前記上限値及び下限値は任意に組合せ可能である。マウスIL-36βの場合、除去されるN末端のアミノ酸の数としては、例えば、1~40個、5~35個、又は10~30個が挙げられる。ヒトIL-36βの場合、除去されるN末端のアミノ酸の数としては、例えば、1~40個、2~30個、3~20個、又は4~10個が挙げられる。truncated ヒトIL-36βのアミノ酸配列としては、例えば、配列番号8に記載のアミノ酸配列(ヌクレオチド配列:配列番号7)、配列番号10に記載のアミノ酸配列(ヌクレオチド配列:配列番号9)等が挙げられる。truncated マウスIL-36βのアミノ酸配列としては、例えば、配列番号12に記載のアミノ酸配列(ヌクレオチド配列:配列番号11)。 Mouse IL-36β has been reported to exhibit approximately 1,000-fold stronger activity compared to native IL-36β in a variant in which 30 amino acids at the N-terminus have been removed (Towne JE, et al. 2011 Dec 9; 286(49):42594-602.). In human IL-36β, it has been reported that a variant in which 4 amino acids at the N-terminus have been removed exhibits stronger activity than wild-type IL-36β (Towne JE, et al., J Biol. Chem. 2011 Dec 9; 286(49):42594-602.). Therefore, variants of IL-36β include those from which amino acids in the N-terminal region of native IL-36β have been removed (hereinafter also referred to as "truncated IL-36β"). The upper limit of the number of N-terminal amino acids to be removed is, for example, 40 or less, 35 or less, 30 or less, 25 or less, 20 or less, 15 or less, 10 or less, 5 or less, or 4 Below are listed. The lower limit for the number of N-terminal amino acids to be removed is 1 or more. The above upper limit and lower limit can be arbitrarily combined. For murine IL-36β, the number of N-terminal amino acids removed includes, for example, 1-40, 5-35, or 10-30. For human IL-36β, the number of N-terminal amino acids removed includes, for example, 1-40, 2-30, 3-20, or 4-10. The amino acid sequence of truncated human IL-36β includes, for example, the amino acid sequence set forth in SEQ ID NO: 8 (nucleotide sequence: SEQ ID NO: 7), the amino acid sequence set forth in SEQ ID NO: 10 (nucleotide sequence: SEQ ID NO: 9), and the like. . The amino acid sequence of truncated mouse IL-36β is, for example, the amino acid sequence set forth in SEQ ID NO: 12 (nucleotide sequence: SEQ ID NO: 11).
<(b)ポリヌクレオチド>
 (b)のポリヌクレオチドは、IL-36又はその改変体をコードするヌクレオチド配列(以下、「IL-36コード配列」という)を有する。IL-36コード配列は、IL-36遺伝子のヌクレオチド配列である。(b)のポリヌクレオチドは、IL-36遺伝子を含むポリヌクレオチドであるともいえる。
<(b) Polynucleotide>
The polynucleotide of (b) has a nucleotide sequence encoding IL-36 or a variant thereof (hereinafter referred to as "IL-36 coding sequence"). An IL-36 coding sequence is the nucleotide sequence of the IL-36 gene. The polynucleotide (b) can also be said to be a polynucleotide containing the IL-36 gene.
≪IL-36コード配列≫
 IL-36コード配列は、天然型IL-36をコードするものでもよく、IL-36の改変体をコードするものでもよい。(b)のポリヌクレオチドとしては、例えば、以下のポリヌクレオチドが挙げられる。
(1)天然型IL-36のアミノ酸配列(例、配列番号2、4又は6)を有するポリペプチドをコードするポリヌクレオチド。
(2)天然型IL-36のアミノ酸配列(例、配列番号2、4又は6)において1個以上のアミノ酸が改変(欠失、置換、付加、挿入、又はそれらの組合せ)されたアミノ酸配列を有するポリペプチドであって、且つIL-36活性を有するポリペプチド、をコードするポリヌクレオチド。
(3)天然型IL-36のアミノ酸配列(例、配列番号2、4又は6)と80%以上の配列同一性を有するアミノ酸配列からなるポリペプチドであって、且つIL-36活性を有するポリペプチド、をコードするポリヌクレオチド。
(4)天然型IL-36遺伝子のヌクレオチド配列(例、配列番号1、3又は5)を有するポリヌクレオチド。
(5)天然型IL-36遺伝子のヌクレオチド配列(例、配列番号1、3又は5)において1個以上のヌクレオチドが改変(欠失、置換、付加、挿入、又はそれらの組合せ)されたヌクレオチド配列を有するポリヌクレオチドであって、且つIL-36活性を有するポリペプチドをコードするポリヌクレオチド。
(6)天然型IL-36遺伝子のヌクレオチド配列(例、配列番号1、3又は5)と80%以上の配列同一性を有するヌクレオチド配列を有するポリヌクレオチドであって、且つIL-36β活性を有するポリペプチドをコードするポリヌクレオチド。
(7)天然型IL-36遺伝子のヌクレオチド配列(例、配列番号1、3又は5)を有するポリヌクレオチドとストリンジェントな条件でハイブリダイズするポリヌクレオチドであって、且つIL-36活性を有するポリペプチドをコードするポリヌクレオチド。
<<IL-36 coding sequence>>
The IL-36 coding sequence may encode native IL-36 or variants of IL-36. The polynucleotides of (b) include, for example, the following polynucleotides.
(1) A polynucleotide encoding a polypeptide having the amino acid sequence of native IL-36 (eg, SEQ ID NO: 2, 4 or 6).
(2) an amino acid sequence in which one or more amino acids are modified (deleted, substituted, added, inserted, or a combination thereof) in the amino acid sequence of native IL-36 (e.g., SEQ ID NO: 2, 4 or 6); and which has IL-36 activity.
(3) A polypeptide consisting of an amino acid sequence having 80% or more sequence identity with the amino acid sequence of native IL-36 (e.g., SEQ ID NO: 2, 4 or 6) and having IL-36 activity A polynucleotide that encodes a peptide.
(4) A polynucleotide having the nucleotide sequence of a native IL-36 gene (eg, SEQ ID NO: 1, 3 or 5).
(5) A nucleotide sequence in which one or more nucleotides are modified (deleted, substituted, added, inserted, or a combination thereof) in the nucleotide sequence of a native IL-36 gene (e.g., SEQ ID NO: 1, 3 or 5) and which encodes a polypeptide having IL-36 activity.
(6) A polynucleotide having a nucleotide sequence having 80% or more sequence identity with the nucleotide sequence of a native IL-36 gene (e.g., SEQ ID NO: 1, 3 or 5), and having IL-36β activity A polynucleotide that encodes a polypeptide.
(7) A polynucleotide that hybridizes under stringent conditions with a polynucleotide having the nucleotide sequence of a native IL-36 gene (e.g., SEQ ID NO: 1, 3 or 5) and has IL-36 activity A polynucleotide that encodes a peptide.
 前記(2)における改変アミノ酸の数、及びに前記(3)における配列同一性の値は、前記<(a)IL-36β>で例示したとおりである。
 前記(5)における改変ヌクレオチドの数としては、例えば、1~100個、1~80個以下、1~60個、1~50個、1~40個、1~30個、1~20個、1~10個、1~5個、1~4個、1~3個、又は1~2個等が挙げられる。
 前記(6)における配列同一性は、例えば、85%以上、90%以上、95%以上、96%以上、97%以上、98%以上、又は99%以上であってもよい。
The number of modified amino acids in (2) above and the value of sequence identity in (3) above are as exemplified in <(a) IL-36β> above.
The number of modified nucleotides in (5) is, for example, 1 to 100, 1 to 80 or less, 1 to 60, 1 to 50, 1 to 40, 1 to 30, 1 to 20, 1 to 10, 1 to 5, 1 to 4, 1 to 3, or 1 to 2, and the like.
The sequence identity in (6) may be, for example, 85% or more, 90% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more.
 IL-36コード配列は、truncated IL-36βをコードするものでもよい。例えば、以下のポリヌクレオチドが挙げられる。
(8)配列番号8、10又は12に記載のアミノ酸配列を有するポリペプチドをコードするポリヌクレオチド。
(9)配列番号8、10又は12に記載のアミノ酸配列において1個以上のアミノ酸が改変(欠失、置換、付加、挿入、又はそれらの組合せ)されたアミノ酸配列を有するポリペプチドであって、且つIL-36β活性を有するポリペプチド、をコードするポリヌクレオチド。
(10)配列番号8、10又は12に記載のアミノ酸配列と80%以上の配列同一性を有するアミノ酸配列からなるポリペプチドであって、且つIL-36β活性を有するポリペプチド、をコードするポリヌクレオチド。
(11)配列番号7、9又は11に記載のヌクレオチド配列を有するポリヌクレオチド。
(12)配列番号7、9又は11に記載のヌクレオチド配列において1個以上の塩基が改変(欠失、置換、付加、挿入、又はそれらの組合せ)されたヌクレオチド配列を有するポリヌクレオチドであって、且つIL-36β活性を有するポリペプチドをコードするポリヌクレオチド。
(13)配列番号7、9又は11に記載のヌクレオチド配列と80%以上の配列同一性を有するヌクレオチド配列を有するポリヌクレオチドであって、且つIL-36β活性を有するポリペプチドをコードするポリヌクレオチド。
(14)配列番号7、9又は11に示すヌクレオチド配列を有するポリヌクレオチドとストリンジェントな条件でハイブリダイズするポリヌクレオチドであって、且つIL-36β活性を有するポリペプチドをコードするポリヌクレオチド。
The IL-36 coding sequence may encode truncated IL-36β. Examples include the following polynucleotides.
(8) A polynucleotide encoding a polypeptide having the amino acid sequence set forth in SEQ ID NO:8, 10 or 12.
(9) A polypeptide having an amino acid sequence in which one or more amino acids are altered (deleted, substituted, added, inserted, or a combination thereof) in the amino acid sequence set forth in SEQ ID NO: 8, 10 or 12, and a polypeptide that has IL-36β activity.
(10) A polynucleotide encoding a polypeptide consisting of an amino acid sequence having 80% or more sequence identity with the amino acid sequence set forth in SEQ ID NO: 8, 10 or 12, and having IL-36β activity .
(11) A polynucleotide having the nucleotide sequence set forth in SEQ ID NO: 7, 9 or 11.
(12) A polynucleotide having a nucleotide sequence in which one or more bases are modified (deleted, substituted, added, inserted, or a combination thereof) in the nucleotide sequence set forth in SEQ ID NO: 7, 9 or 11, and a polynucleotide encoding a polypeptide having IL-36β activity.
(13) A polynucleotide having a nucleotide sequence having 80% or more sequence identity with the nucleotide sequence set forth in SEQ ID NO: 7, 9 or 11 and encoding a polypeptide having IL-36β activity.
(14) A polynucleotide that hybridizes under stringent conditions with a polynucleotide having the nucleotide sequence shown in SEQ ID NO: 7, 9 or 11 and that encodes a polypeptide having IL-36β activity.
 前記(9)における改変アミノ酸の数は、例えば、1~50個、1~40個、1~30個、1~20個、1~10個、又は1~5個、1~4個、1~3個、又は1~2個等が挙げられる。前記(10)における配列同一性は、例えば、85%以上、90%以上、又は95%以上であってもよい。前記(12)における改変塩基の数は、例えば、1~100個、1~80個以下、1~60個、1~50個、1~40個、1~30個、1~20個、1~10個又は1~5個等が挙げられる。前記(13)における配列同一性としては、例えば、5%以上、90%以上、95%以上、96%以上、97%以上、98%以上、又は99%以上が挙げられる。 The number of modified amino acids in (9) is, for example, 1 to 50, 1 to 40, 1 to 30, 1 to 20, 1 to 10, or 1 to 5, 1 to 4, 1 to 3, or 1 to 2, and the like. The sequence identity in (10) may be, for example, 85% or more, 90% or more, or 95% or more. The number of modified bases in (12) is, for example, 1 to 100, 1 to 80 or less, 1 to 60, 1 to 50, 1 to 40, 1 to 30, 1 to 20, 1 to 10 or 1 to 5, and the like. The sequence identity in (13) includes, for example, 5% or more, 90% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more.
≪他のヌクレオチド配列≫
 (b)のポリヌクレオチドは、IL-36コード配列に加えて、他のヌクレオチド配列を含んでいてもよい。他のヌクレオチド配列としては、例えば、シグナルペプチドをコードするヌクレオチド配列(以下、「シグナルペプチドコード配列」という)、及びIL-36遺伝子の発現を制御する配列(以下、「発現制御配列」という)等が挙げられる。
≪Other nucleotide sequences≫
The polynucleotide of (b) may contain other nucleotide sequences in addition to the IL-36 coding sequence. Other nucleotide sequences include, for example, a nucleotide sequence encoding a signal peptide (hereinafter referred to as "signal peptide coding sequence"), a sequence controlling IL-36 gene expression (hereinafter referred to as "expression control sequence"), and the like. is mentioned.
(シグナルペプチドコード配列)
 (b)のポリヌクレオチドは、IL-36コード配列に加えて、分泌タンパク質のシグナルペプチドコード配列をさらに有していることが好ましい。シグナルペプチドコード配列は、IL-36コード配列の5’末端に直接又は間接的にインフレームで連結されていることが好ましい。IL-36コード配列の5’末端にシグナルペプチドコード配列がインフレームで連結されることで、(b)のポリヌクレオチドが導入された細胞内で、IL-36が産生された場合に、IL-36βを細胞外に分泌させることができる。
(signal peptide coding sequence)
The polynucleotide of (b) preferably further has a signal peptide coding sequence for a secretory protein in addition to the IL-36 coding sequence. The signal peptide coding sequence is preferably directly or indirectly linked in-frame to the 5' end of the IL-36 coding sequence. A signal peptide coding sequence is ligated in-frame to the 5′ end of the IL-36 coding sequence, so that when IL-36 is produced in cells into which the polynucleotide of (b) has been introduced, IL- 36β can be secreted extracellularly.
 シグナルペプチドコード配列としては、(b)のポリヌクレオチドが導入され得る細胞(以下、「導入対象細胞」ともいう)において、分泌シグナルとして機能し得るシグナルペプチドをコードする配列が挙げられる。IL-36が安定的に細胞外に分泌され得るという観点から、シグナルペプチドコード配列は、導入対象細胞と同種の生物に由来することが好ましい。例えば、導入対象細胞がヒト細胞である場合、ヒトの分泌タンパク質のシグナルペプチドコード配列を用いることが好ましい。導入対象細胞がマウス細胞である場合、マウスの分泌タンパク質のシグナルペプチドコード配列を用いることが好ましい。 The signal peptide coding sequence includes a sequence that encodes a signal peptide that can function as a secretory signal in cells into which the polynucleotide of (b) can be introduced (hereinafter also referred to as "introduction target cells"). From the viewpoint that IL-36 can be stably secreted extracellularly, the signal peptide coding sequence is preferably derived from the same species of organism as the target cell. For example, when the cells to be introduced are human cells, it is preferable to use the signal peptide coding sequence of a human secretory protein. When the cells to be introduced are mouse cells, it is preferable to use the signal peptide coding sequence of a mouse secretory protein.
 シグナルペプチドコード配列が由来する分泌タンパク質は、特に限定されない。分泌タンパク質としては、例えば、副甲状腺ホルモン、成長ホルモン等のペプチドホルモン;IFN-γ、GM-CSF、IL-6等のサイトカイン;イムノグロブリンの重鎖又は軽鎖等が挙げられるが、これらに限定されない。
 シグナルペプチドコード配列としては、例えば、配列番号14に記載のアミノ酸配列(ヒト副甲状腺ホルモン(hPTH)のシグナルペプチド)をコードするヌクレオチド配列(例えば、配列番号13)、配列番号16に記載のアミノ酸配列(ヒト成長ホルモン(hGH)のシグナルペプチド)をコードするヌクレオチド配列(例えば、配列番号15)等が挙げられる。
The secreted protein from which the signal peptide coding sequence is derived is not particularly limited. Examples of secretory proteins include peptide hormones such as parathyroid hormone and growth hormone; cytokines such as IFN-γ, GM-CSF and IL-6; immunoglobulin heavy or light chains; not.
Examples of the signal peptide coding sequence include a nucleotide sequence (eg, SEQ ID NO: 13) encoding the amino acid sequence set forth in SEQ ID NO: 14 (the signal peptide of human parathyroid hormone (hPTH)), and the amino acid sequence set forth in SEQ ID NO: 16. and a nucleotide sequence (eg, SEQ ID NO: 15) encoding (signal peptide of human growth hormone (hGH)).
 シグナルペプチドコード配列は、IL-36コード配列の5’末端に、直接連結されてもよいし、リンカーを介して間接的に連結されてもよい。リンカーは、シグナルペプチドコード配列-リンカー配列-IL-36コード配列が翻訳された場合に、IL-36の活性を損なわないものであれば、特に限定されない。リンカーの長さとしては、例えば、3ヌクレオチド長、6ヌクレオチド長、9ヌクレオチド長、又は12ヌクレオチド長等が挙げられる。
 シグナルペプチドコード配列は、IL-36コード配列の5’末端に、直接連結されることが好ましい。
The signal peptide coding sequence may be directly linked to the 5' end of the IL-36 coding sequence or indirectly via a linker. The linker is not particularly limited as long as it does not impair IL-36 activity when the signal peptide coding sequence-linker sequence-IL-36 coding sequence is translated. The length of the linker may be, for example, 3 nucleotides, 6 nucleotides, 9 nucleotides, or 12 nucleotides.
Preferably, the signal peptide coding sequence is directly linked to the 5' end of the IL-36 coding sequence.
(発現制御配列)
 (b)のポリヌクレオチドは、IL-36コード配列に加えて、発現制御配列を有していることが好ましい。発現制御配列としては、例えば、プロモーター、エンハンサー、ポリA付加シグナル、及びターミネーター等が挙げられる。
 プロモーターは、導入対象細胞で機能し得るものであれば、特に限定されない。プロモーターとしては、例えば、CMV(サイトメガロウイルス)プロモーター、SRαプロモーター、SV40初期プロモーター、レトロウイルスのLTR、RSV(ラウス肉腫ウイルス)プロモーター、HSV-TK(単純ヘルペスウイルスチミジンキナーゼ)プロモーター、EF1αプロモーター、メタロチオネインプロモーター、ヒートショックプロモーター等が挙げられるが、これらに限定されない。
 IL-36コード配列は、適切なプロモーターに作動可能に連結されていることが好ましい。(b)のポリヌクレオチドがシグナルペプチドコード配列を有する場合、シグナルペプチドコード配列及びIL-36コード配列が、適切なプロモーターに作動可能に連結されていることが好ましい。
(expression control sequence)
The polynucleotide of (b) preferably has an expression control sequence in addition to the IL-36 coding sequence. Examples of expression control sequences include promoters, enhancers, poly-A addition signals, terminators and the like.
The promoter is not particularly limited as long as it can function in the cells to be introduced. Promoters include, for example, CMV (cytomegalovirus) promoter, SRα promoter, SV40 early promoter, retroviral LTR, RSV (Rous sarcoma virus) promoter, HSV-TK (herpes simplex virus thymidine kinase) promoter, EF1α promoter, metallothionein promoters, heat shock promoters, etc., but are not limited thereto.
The IL-36 coding sequence is preferably operably linked to a suitable promoter. If the polynucleotide of (b) has a signal peptide coding sequence, it is preferred that the signal peptide coding sequence and the IL-36 coding sequence are operably linked to a suitable promoter.
 (b)のポリヌクレオチドは、DNAであってもよく、RNAであってもよい。(b)のポリヌクレオチドがDNAである場合、後述のベクターの形態であってもよい。(b)のポリヌクレオチドがRNAである場合、mRNAの形態であってもよい。この場合、(b)のポリヌクレオチドは、5’Cap構造、poly-A-tail等を有していてもよい。 The polynucleotide of (b) may be DNA or RNA. When the polynucleotide of (b) is DNA, it may be in the form of a vector described below. When the polynucleotide of (b) is RNA, it may be in the form of mRNA. In this case, the polynucleotide (b) may have a 5'Cap structure, poly-A-tail, or the like.
<(c)ベクター>
 (c)のベクターは、前記(b)のポリヌクレオチドを含む。ベクターの種類は、特に限定されず、遺伝子発現に一般的に用いられる発現ベクター等を使用することができる。ベクターは、直鎖状であってもよく、環状であってもよい。ベクターとしては、例えば、プラスミドなどの非ウイルスベクター、ウイルスベクター、トランスポゾンベクター、エピソーマルベクター、人工染色体ベクター等が挙げられる。
<(c) Vector>
The vector of (c) contains the polynucleotide of (b). The type of vector is not particularly limited, and an expression vector or the like generally used for gene expression can be used. Vectors may be linear or circular. Examples of vectors include non-viral vectors such as plasmids, viral vectors, transposon vectors, episomal vectors, artificial chromosome vectors and the like.
 プラスミドベクターとしては、例えば、pA1-11、pXT1、pRc/CMV、pRc/RSV、pcDNAI/Neo等の、動物細胞発現用プラスミドベクターが挙げられる。 Examples of plasmid vectors include animal cell expression plasmid vectors such as pA1-11, pXT1, pRc/CMV, pRc/RSV, and pcDNAI/Neo.
 ウイルスベクターとしては、例えば、センダイウイルスベクター、レトロウイルス(レンチウイルスを含む)ベクター、アデノウイルスベクター、アデノ随伴ウイルスベクター、ヘルペスウイルスベクター、ワクシニアウイルスベクター、ポックスウイルスベクター、ポリオウイルスベクター、シルビスウイルスベクター、ラブドウイルスベクター、パラミクソウイルスベクター、オルソミクソウイルスベクター等が挙げられる。 Viral vectors include, for example, Sendai virus vectors, retrovirus (including lentivirus) vectors, adenovirus vectors, adeno-associated virus vectors, herpes virus vectors, vaccinia virus vectors, pox virus vectors, polio virus vectors, silvis virus vectors , rhabdovirus vector, paramyxovirus vector, orthomyxovirus vector and the like.
 エピソーマルベクターは、染色体外で自律複製可能なベクターである。エピソーマルベクターとしては、例えば、EBV、SV40等に由来する自律複製に必要な配列をベクター要素として含むベクターが挙げられる。自律複製に必要なベクター要素としては、具体的には、複製開始点と、複製開始点に結合して複製を制御するタンパク質をコードする遺伝子が挙げられる。例えば、EBVでは複製開始点oriPとEBNA-1遺伝子、SV40では複製開始点oriとSV40LT遺伝子が挙げられる。 An episomal vector is a vector that can replicate autonomously outside the chromosome. Examples of episomal vectors include vectors containing, as vector elements, sequences necessary for autonomous replication derived from EBV, SV40, and the like. Vector elements required for autonomous replication specifically include genes encoding replication origins and proteins that bind to replication origins to control replication. For example, EBV includes the replication origin oriP and the EBNA-1 gene, and SV40 includes the replication origin ori and the SV40LT gene.
 人工染色体ベクターとしては、例えば、YAC(Yeast artificial chromosome)ベクター、BAC(Bacterial artificial chromosome)ベクター、PAC(P1-derived artificial chromosome)ベクター等が挙げられる。 Examples of artificial chromosome vectors include YAC (Yeast artificial chromosome) vectors, BAC (Bacterial artificial chromosome) vectors, PAC (P1-derived artificial chromosome) vectors, and the like.
 ベクターは、市販のものを利用して作製することができる。例えば、市販のベクターのマルチクローニングサイトに、前記(b)のポリヌクレオチドを挿入することにより、前記(b)のポリヌクレオチドを含むベクターを作製することができる。市販のベクターへの前記(b)のポリヌクレオチドの挿入は、例えば、制限酵素処理、及びリガーゼ処理等を組み合わせて行うことができる。 Vectors can be produced using commercially available products. For example, a vector containing the polynucleotide (b) can be produced by inserting the polynucleotide (b) into the multicloning site of a commercially available vector. Insertion of the polynucleotide (b) into a commercially available vector can be performed by, for example, a combination of restriction enzyme treatment, ligase treatment, and the like.
 (c)のベクターが、ゲノム編集技術等により導入対象細胞のゲノムに組み込まれるターゲティングベクターである場合、相同組換えのためのホモロジーアームを含むことが好ましい。ホモロジーアームは、導入対象細胞の任意のゲノム領域に相同な配列を有する。ホモロジーアームに相同なゲノム領域は、(b)のポリヌクレオチドが挿入される領域である。ホモロジーアームは、(b)のポリヌクレオチドの5’側及び3’側に隣接して配置される。
 ホモロジーアームの配列は、導入対象細胞の種類及び標的領域に応じて、適宜、設計することができる。例えば、導入対象細胞が、腫瘍細胞である場合、当該腫瘍細胞が有する腫瘍遺伝子を標的領域として選択し、ホモロジーアームを設計してもよい。
When the vector (c) is a targeting vector that is integrated into the genome of the target cell by genome editing technology or the like, it preferably contains homology arms for homologous recombination. A homology arm has a sequence homologous to any genomic region of the cell to be introduced. The genomic region homologous to the homology arms is the region into which the polynucleotide of (b) is inserted. The homology arms are located flanking the 5' and 3' sides of the polynucleotide of (b).
The sequence of homology arms can be appropriately designed according to the type of cell to be introduced and the target region. For example, when the cells to be introduced are tumor cells, an oncogene possessed by the tumor cells may be selected as a target region and homology arms may be designed.
<(d)細胞>
 (d)の細胞は、IL-36又はその改変体を分泌する細胞(以下、「IL-36分泌細胞」という)である。IL-36分泌細胞は、天然型IL-36分泌細胞であってもよく、遺伝子導入型IL-36分泌細胞であってもよい。IL-36分泌細胞が生体に投与される場合、IL-36分泌細胞は、投与対象である生物の細胞であることが好ましい。
<(d) cells>
The cells of (d) are cells that secrete IL-36 or variants thereof (hereinafter referred to as "IL-36-secreting cells"). The IL-36-secreting cells may be native IL-36-secreting cells or transgenic IL-36-secreting cells. When IL-36-secreting cells are administered to a living organism, the IL-36-secreting cells are preferably cells of the organism to which they are administered.
 天然型IL-36分泌細胞は、内生的にIL-36を産生し、分泌する細胞である。天然型IL-36分泌細胞としては、例えば、上皮細胞、単球、及びB細胞等が挙げられる。
 遺伝子導入型IL-36分泌細胞は、(b)のポリヌクレオチド又は(c)のベクターが導入された細胞である。ホスト細胞は、特に限定されないが、例えば、投与対象から採取した細胞、投与対象と同じMHCを有する細胞等が挙げられる。細胞の種類としては、例えば、腫瘍細胞、免疫細胞(樹状細胞、T細胞及びナチュラルキラー細胞等)、線維芽細胞等が挙げられるが、これらに限定されない。ホスト細胞は、例えば、T細胞であってもよい。T細胞としては、例えば、CD8陽性T細胞(CTL等)、CD4陽性T細胞(ヘルパーT細胞等)等が挙げられる。
Native IL-36 secreting cells are cells that endogenously produce and secrete IL-36. Native IL-36-secreting cells include, for example, epithelial cells, monocytes, B cells, and the like.
Transgenic IL-36-secreting cells are cells into which the polynucleotide of (b) or the vector of (c) has been introduced. Host cells are not particularly limited, but include, for example, cells collected from an administration subject, cells having the same MHC as that of the administration subject, and the like. Examples of cell types include, but are not limited to, tumor cells, immune cells (dendritic cells, T cells, natural killer cells, etc.), fibroblasts, and the like. A host cell can be, for example, a T cell. Examples of T cells include CD8-positive T-cells (CTL etc.), CD4-positive T-cells (helper T-cells etc.) and the like.
 遺伝子導入型IL-36分泌細胞は、ベクターの種類に応じて、公知の遺伝子導入方法を用いて作製することができる。遺伝子導入方法としては、例えば、ウイルス感染法、リポフェクション法、マイクロインジェクション法、カルシウムリン酸法、DEAE-デキストラン法、エレクトロポーレーション法、トランスポゾンを用いる方法、パーティクルガン法等が挙げられる。
 遺伝子導入型IL-36分泌細胞は、公知の遺伝子編集技術等を用いて作製してもよい。遺伝子編集技術としては、例えば、ジンクフィンガーヌクレアーゼ、TALEN(転写活性化様エフェクターヌクレアーゼ)、CRISPR-Casシステム等のエンドヌクレアーゼを用いる技術が挙げられる。
Transgenic IL-36-secreting cells can be prepared using known gene transfer methods depending on the type of vector. Gene introduction methods include, for example, the virus infection method, lipofection method, microinjection method, calcium phosphate method, DEAE-dextran method, electroporation method, method using transposon, particle gun method and the like.
Transgenic IL-36-secreting cells may be prepared using known gene editing techniques and the like. Gene editing techniques include, for example, techniques using endonucleases such as zinc finger nucleases, TALENs (transcription activation-like effector nucleases), and CRISPR-Cas systems.
 遺伝子導入型IL-36分泌細胞を作製する場合、(c)のベクターは、マーカー遺伝子を含んでいてもよい。マーカー遺伝子は、IL-36β遺伝子導入細胞を選別するために用いることができる。マーカー遺伝子は、一般的に用いられるもの(例えば、薬剤耐性遺伝子、蛍光色素遺伝子)を特に制限なく用いることができる。マーカー遺伝子としては、例えば、ネオマイシン耐性遺伝子、アンピシリン耐性遺伝子、ハイグロマイシン耐性遺伝子、テトラサイクリン耐性遺伝子、及びクロラムフェニコール耐性遺伝子等が挙げられるが、これらに限定されない。
 マーカー遺伝子は、2Aペプチド等の自己切断端ペプチドをコードするヌクレオチド配列、又はIRES(internal ribozyme entry site)配列等を介在させて、IL-36コード配列(IL-36遺伝子)に連結してもよい。これらの配列を介在させることにより、1つのプロモーターから、IL-36遺伝子とマーカー遺伝子とを独立して発現させることができる。
When producing transgenic IL-36-secreting cells, the vector (c) may contain a marker gene. A marker gene can be used to select IL-36β-transfected cells. Commonly used marker genes (eg, drug resistance genes, fluorescent dye genes) can be used without particular limitations. Examples of marker genes include, but are not limited to, neomycin resistance gene, ampicillin resistance gene, hygromycin resistance gene, tetracycline resistance gene, chloramphenicol resistance gene, and the like.
The marker gene may be linked to the IL-36 coding sequence (IL-36 gene) via a nucleotide sequence encoding a self-cleaving terminal peptide such as 2A peptide, or an IRES (internal ribozyme entry site) sequence or the like. . By interposing these sequences, the IL-36 gene and the marker gene can be expressed independently from one promoter.
 IL-36分泌細胞からのIL-36の分泌は、公知の方法で確認することができる。例えば、IL-36分泌細胞の培養上清を採取し、ウエスタンブロッティング、又はELISA等の免疫学的方法によりIL-36を測定することで、IL-36分泌細胞からのIL-36の分泌を確認することができる。あるいは、線維芽細胞等のIL-6産生細胞に、IL-36分泌細胞の培養上清を作用させ、前記線維芽細胞からのIL-6の分泌量を測定することにより、IL-36分泌細胞からのIL-36の分泌を確認することができる。 Secretion of IL-36 from IL-36-secreting cells can be confirmed by a known method. For example, IL-36 secretion from IL-36-secreting cells is confirmed by collecting the culture supernatant of IL-36-secreting cells and measuring IL-36 by Western blotting or an immunological method such as ELISA. can do. Alternatively, IL-36-secreting cells by allowing the culture supernatant of IL-36-secreting cells to act on IL-6-producing cells such as fibroblasts and measuring the amount of IL-6 secreted from the fibroblasts. IL-36 secretion from .
 本実施形態のPD-1発現抑制剤は、前記(a)~(d)の成分のいずれか1種を単独で含んでもよく、2種以上を組み合わせて含んでもよい。
 PD-1発現抑制剤は、(a)~(d)からなる群より選択される少なくとも1種の成分を、PD-1発現抑制効果の有効成分として含むことができる。「PD-1発現抑制効果」とは、リンパ球にPD-1発現抑制剤を暴露した場合に、リンパ球にPD-1発現抑制剤を曝露しない場合と比較して、リンパ球のPD-1発現が抑制される効果をいう。例えば、PD-1発現抑制剤の存在下で、リンパ球を培養すると、PD-1発現抑制剤の非存在下で培養したときと比較して、リンパ球のPD-1発現が抑制される。例えば、PD-1発現抑制剤の非存在下でリンパ球を培養したときのPD-1発現量を100とした場合、PD-1発現抑制剤の存在下でリンパ球を培養したときのPD-1の相対発現量は、90以下、80以下、70以下、60以下、50以下、又は40以下であってもよい。リンパ球のPD-1発現量は、公知の方法で、確認することができる。PD-1発現量の確認方法としては、例えば、フローサイトメトリー解析、RT-qPCR等が挙げられる。
The PD-1 expression inhibitor of the present embodiment may contain any one of the components (a) to (d) alone, or may contain two or more in combination.
The PD-1 expression inhibitor can contain at least one component selected from the group consisting of (a) to (d) as an active ingredient for the PD-1 expression inhibitory effect. The "PD-1 expression inhibitory effect" means that when the PD-1 expression inhibitor is exposed to the lymphocytes, compared with the case where the PD-1 expression inhibitor is not exposed to the lymphocytes, PD-1 of the lymphocytes. It refers to the effect of suppressing expression. For example, culturing lymphocytes in the presence of a PD-1 expression inhibitor suppresses PD-1 expression in lymphocytes compared to culturing in the absence of a PD-1 expression inhibitor. For example, when the PD-1 expression level is 100 when lymphocytes are cultured in the absence of a PD-1 expression inhibitor, when the lymphocytes are cultured in the presence of a PD-1 expression inhibitor, PD- The relative expression level of 1 may be 90 or less, 80 or less, 70 or less, 60 or less, 50 or less, or 40 or less. The PD-1 expression level of lymphocytes can be confirmed by a known method. Methods for confirming the PD-1 expression level include, for example, flow cytometry analysis, RT-qPCR and the like.
 本実施形態のPD-1発現抑制剤によれば、リンパ球のPD-1発現を効果的に抑制することができる。
 本実施形態のPD-1発現抑制剤は、がんの養子免疫療法又はCAR-T療法等に使用するリンパ球を調製するために用いることができる。例えば、本実施形態のPD-1発現抑制剤の存在下で、リンパ球を培養することで、PD-1の発現が抑制されたリンパ球を調製することができる。このようなリンパ球をがん患者に投与することで、免疫チェックポイントシステムを回避して、がん細胞に対する細胞傷害活性を発現し得る。本実施形態のPD-1発現抑制剤を、in vitroで用いる場合、(a)又は(d)の成分を用いることが好ましい。
PD-1 expression in lymphocytes can be effectively suppressed by the PD-1 expression inhibitor of the present embodiment.
The PD-1 expression inhibitor of this embodiment can be used to prepare lymphocytes used for adoptive immunotherapy or CAR-T therapy for cancer. For example, lymphocytes with suppressed PD-1 expression can be prepared by culturing lymphocytes in the presence of the PD-1 expression inhibitor of the present embodiment. By administering such lymphocytes to cancer patients, they can bypass the immune checkpoint system and express cytotoxic activity against cancer cells. When the PD-1 expression inhibitor of the present embodiment is used in vitro, it is preferable to use component (a) or (d).
 本実施形態のPD-1発現抑制剤は、生体内のリンパ球のPD-1発現を抑制するために、生体に投与されてもよい。PD-1発現抑制剤が(a)の成分である場合、IL-36又はその改変体が生体内でリンパ球に接触し、当該リンパ球のPD-1発現が抑制される。PD-1発現抑制剤が(b)又は(c)の成分である場合、(b)のポリヌクレオチド又は(c)のベクターを取り込んだ細胞がIL-36又はその改変体を産生する。産生されたIL-36又はその改変体が生体内でリンパ球に接触し、当該リンパ球のPD-1発現が抑制される。PD-1発現抑制剤が(d)の成分である場合、(d)の細胞が産生したIL-36又はその改変体が、生体内でリンパ球に接触し、当該リンパ球のPD-1発現が抑制される。 The PD-1 expression inhibitor of the present embodiment may be administered to a living body in order to suppress PD-1 expression of lymphocytes in the living body. When the PD-1 expression inhibitor is the component (a), IL-36 or a variant thereof contacts lymphocytes in vivo and PD-1 expression in the lymphocytes is inhibited. When the PD-1 expression inhibitor is the component (b) or (c), cells incorporating the polynucleotide of (b) or the vector of (c) produce IL-36 or a variant thereof. The produced IL-36 or its variant comes into contact with lymphocytes in vivo, and PD-1 expression in the lymphocytes is suppressed. When the PD-1 expression inhibitor is a component of (d), IL-36 produced by the cells of (d) or a variant thereof contacts lymphocytes in vivo, and PD-1 expression of the lymphocytes is suppressed.
[医薬組成物]
 本開示の第2の態様は、免疫チェックポイント阻害用医薬組成物である。前記免疫チェックポイント阻害用医薬組成物は、第1の態様のPD-1発現抑制剤と、薬学的に許容される担体とを含有する。
[Pharmaceutical composition]
A second aspect of the present disclosure is a pharmaceutical composition for immune checkpoint inhibition. The pharmaceutical composition for immune checkpoint inhibition contains the PD-1 expression inhibitor of the first aspect and a pharmaceutically acceptable carrier.
 本実施形態の医薬組成物は、免疫チェックポイントシステムを抑制するために、対象に投与される。本実施形態の医薬組成物が投与されると、投与又は産生されたIL-36若しくはその改変体が、生体内のリンパ球に作用する。これにより、リンパ球のPD-1発現が抑制されて、PD-1を介した免疫チェックポイントシステムが抑制される。 The pharmaceutical composition of this embodiment is administered to a subject to suppress the immune checkpoint system. When the pharmaceutical composition of this embodiment is administered, the administered or produced IL-36 or its variant acts on lymphocytes in vivo. This suppresses PD-1 expression in lymphocytes and suppresses the PD-1-mediated immune checkpoint system.
 本実施形態の医薬組成物の投与対象は、PD-1を介した免疫チェックポイントシステムの抑制が必要な対象である。投与対象としては、例えば、がん患者が挙げられる。本実施形態の医薬組成物をがん患者に投与する場合、本実施形態の医薬組成物を単独で投与してもよく、抗がん剤と組み合わせて投与してもよい。 The subject of administration of the pharmaceutical composition of the present embodiment is a subject requiring inhibition of the immune checkpoint system via PD-1. Administration subjects include, for example, cancer patients. When administering the pharmaceutical composition of the present embodiment to a cancer patient, the pharmaceutical composition of the present embodiment may be administered alone or in combination with an anticancer agent.
 本実施形態の医薬組成物は、第1の態様のPD-1発現抑制剤に加えて、薬学的に許容される担体を含有する。「薬学的に許容される担体」とは、有効成分の生理活性を阻害せず、且つ、その投与対象に対して実質的な毒性を示さない担体を意味する。「実質的な毒性を示さない」とは、その成分が通常使用される投与量において、投与対象に対して毒性を示さないことを意味する。本実施形態の医薬組成物において、薬学的に許容される担体は、前記(a)~(d)の成分のPD-1発現抑制活性を阻害せず、且つその投与対象に対して実質的な毒性を示さない担体である。薬学的に許容される担体は、典型的には非活性成分とみなされる、公知のあらゆる薬学的に許容され得る成分を包含する。薬学的に許容される担体は、特に限定されないが、例えば、溶媒、希釈剤、ビヒクル、賦形剤、流動促進剤、結合剤、造粒剤、分散化剤、懸濁化剤、湿潤剤、滑沢剤、崩壊剤、可溶化剤、安定剤、乳化剤、充填剤、保存剤(例えば、酸化防止剤)、キレート剤、矯味矯臭剤、甘味剤、増粘剤、緩衝剤、着色剤等が挙げられる。薬学的に許容される担体は、1種を単独で用いてもよく、2種以上を併用してもよい。医薬組成物が前記(b)又は(c)を含有する場合、薬学的に許容される担体は、溶媒、希釈剤、ビヒクル、緩衝液等であってもよい。医薬組成物が前記(d)を含有する場合、薬学的に許容される担体は、細胞培養液、又は緩衝液(生理食塩水、PBS、クエン酸緩衝液など)等であってもよい。 The pharmaceutical composition of this embodiment contains a pharmaceutically acceptable carrier in addition to the PD-1 expression inhibitor of the first aspect. "Pharmaceutically acceptable carrier" means a carrier that does not inhibit the physiological activity of the active ingredient and does not exhibit substantial toxicity to the subject to which it is administered. The phrase "substantially non-toxic" means that the component does not show toxicity to the subject at the dose normally used. In the pharmaceutical composition of this embodiment, the pharmaceutically acceptable carrier does not inhibit the PD-1 expression inhibitory activity of the components (a) to (d), and is substantially It is a non-toxic carrier. Pharmaceutically acceptable carriers include any known pharmaceutically acceptable ingredients that are typically considered non-active ingredients. Pharmaceutically acceptable carriers include, but are not limited to, solvents, diluents, vehicles, excipients, glidants, binders, granulating agents, dispersing agents, suspending agents, wetting agents, Lubricants, disintegrants, solubilizers, stabilizers, emulsifiers, fillers, preservatives (e.g., antioxidants), chelating agents, flavoring agents, sweeteners, thickeners, buffering agents, coloring agents, etc. mentioned. One type of pharmaceutically acceptable carrier may be used alone, or two or more types may be used in combination. When the pharmaceutical composition contains (b) or (c) above, pharmaceutically acceptable carriers may be solvents, diluents, vehicles, buffers and the like. When the pharmaceutical composition contains (d) above, the pharmaceutically acceptable carrier may be a cell culture medium, a buffer (physiological saline, PBS, citrate buffer, etc.), or the like.
 本実施形態の医薬組成物は、薬学的に許容される担体として挙げたものに加えて、医薬分野において常用される添加剤等を特に制限なく使用することができる。医薬組成物が前記(b)又は(c)を含有する場合、前記医薬組成物は、トランスフェクション促進剤、ゲノム編集試薬(CRISPR/Cas9システムなど)等を含有してもよい。 In the pharmaceutical composition of the present embodiment, in addition to those listed as pharmaceutically acceptable carriers, additives commonly used in the pharmaceutical field can be used without particular limitation. When the pharmaceutical composition contains (b) or (c), the pharmaceutical composition may contain a transfection promoter, a genome editing reagent (CRISPR/Cas9 system, etc.) and the like.
 医薬組成物の剤型は、特に制限されず、医薬品製剤として一般的に用いられる剤型とすることができる。医薬組成物は、経口製剤であってもよく、非経口製剤であってもよい。経口製剤としては、例えば、錠剤、被覆錠剤、丸剤、散剤、顆粒剤、カプセル剤、シロップ剤、細粒剤、液剤、ドロップ愛、乳剤等が例示される。非経口製剤としては、例えば、注射剤、坐剤、軟膏、スプレー剤、点鼻剤、吸入剤等が例示される。これらの剤型の医薬組成物は、定法(例えば、日本薬局方記載の方法)に従って、製剤化することができる。
 本実施形態の医薬組成物は、非経口剤が好ましく、注射剤がより好ましい。
The dosage form of the pharmaceutical composition is not particularly limited, and may be a dosage form commonly used as a pharmaceutical preparation. The pharmaceutical composition may be an oral formulation or a parenteral formulation. Examples of oral preparations include tablets, coated tablets, pills, powders, granules, capsules, syrups, fine granules, liquids, drops, emulsions and the like. Examples of parenteral formulations include injections, suppositories, ointments, sprays, nasal drops, and inhalants. Pharmaceutical compositions in these dosage forms can be formulated according to standard methods (eg, methods described in the Japanese Pharmacopoeia).
The pharmaceutical composition of this embodiment is preferably a parenteral formulation, more preferably an injection.
 本実施形態の医薬組成物は、第1の態様のPD-1発現阻害剤以外の活性成分を含んでいてもよい。前記活性成分としては、特に限定されず、例えば、ビタミン類及びその誘導体類、消炎剤、抗炎症剤、血行促進剤、刺激剤、ホルモン類、刺激緩和剤、鎮痛剤、細胞賦活剤、植物・動物・微生物エキス、鎮痒剤、消炎鎮痛剤、抗真菌剤、抗ヒスタミン剤、催眠鎮静剤、精神安定剤、抗高血圧剤、降圧利尿剤、抗生物質、麻酔剤、抗菌性物質、抗てんかん剤、冠血管拡張剤、生薬、止痒剤、角質軟化剥離剤等が挙げられるが、これらに限定されない。活性成分は、他の抗腫瘍剤であってもよい。他の抗腫瘍剤は、適用するがんの種類に応じて適宜選択すればよい。他の活性成分は、1種を単独で用いてもよく、2種以上を併用してもよい。 The pharmaceutical composition of this embodiment may contain active ingredients other than the PD-1 expression inhibitor of the first aspect. The active ingredient is not particularly limited. Animal/microorganism extract, antipruritic, anti-inflammatory analgesic, antifungal agent, antihistamine, sedative hypnotic, tranquilizer, antihypertensive, hypotensive diuretic, antibiotic, anesthetic, antibacterial, antiepileptic, coronary artery Examples include, but are not limited to, dilating agents, herbal medicines, antipruritic agents, keratin softening exfoliants, and the like. The active ingredient may also be another anti-tumor agent. Other antitumor agents may be appropriately selected according to the type of cancer to be applied. Other active ingredients may be used alone or in combination of two or more.
 本実施形態の医薬組成物は、第1の態様のPD-1発現阻害剤を治療的有効量含むことができる。「治療的有効量」とは、医薬組成物の目的達成のために有効な薬剤の量を意味する。例えば、(a)~(d)のいずれかの成分の治療的有効量は、リンパ球のPD-1発現を抑制できる量であり得る。医薬組成物が(a)~(c)のいずれかの成分を含有する場合、治療的有効量としては、例えば、投与単位形態当たり、0.01μg~100mg、0.1μg~10mg、0.5μg~5mg、又は1μg~1mg等が挙げられる。医薬組成物が(d)の成分を含有する場合、治療的有効量としては、例えば、投与単位形態当たり、細胞の個数として10~1010個、10~10個、又は10~10個等が挙げられる。 The pharmaceutical composition of this embodiment can contain a therapeutically effective amount of the PD-1 expression inhibitor of the first aspect. By "therapeutically effective amount" is meant an amount of drug effective to achieve the purpose of the pharmaceutical composition. For example, a therapeutically effective amount of any of components (a)-(d) can be an amount capable of suppressing PD-1 expression in lymphocytes. When the pharmaceutical composition contains any one of components (a) to (c), the therapeutically effective amount is, for example, 0.01 μg to 100 mg, 0.1 μg to 10 mg, 0.5 μg per dosage unit form. up to 5 mg, or 1 μg to 1 mg, and the like. When the pharmaceutical composition contains component (d), the therapeutically effective amount is, for example, 10 3 to 10 10 cells, 10 4 to 10 9 cells, or 10 5 to 10 5 cells per dosage unit form. 10 8 etc. are mentioned.
<投与方法>
 医薬組成物は、公知の方法により、投与が必要とされる対象に投与することができる。投与方法は、経口投与であってもよく、非経口投与であってもよいが、非経口投与が好ましい。非経口経路は、経口以外のいずれの投与経路であってもよく、例えば、静脈内投与、筋肉内投与、皮下投与、鼻腔内投与、皮内投与、眼内投与、脳内投与、直腸内投与、腟内投与、及び腹腔内投与等が挙げられる。投与方法は、局所投与であってもよく、全身投与であってもよい。好ましい投与経路としては、静脈内投与、皮下投与、筋肉内投与、腫瘍内投与等が挙げられる。
<Administration method>
Pharmaceutical compositions can be administered to subjects in need thereof by known methods. The administration method may be oral administration or parenteral administration, but parenteral administration is preferred. A parenteral route may be any route of administration other than the oral route, such as intravenous administration, intramuscular administration, subcutaneous administration, intranasal administration, intradermal administration, intraocular administration, intracerebral administration, intrarectal administration. , vaginal administration, and intraperitoneal administration. The administration method may be local administration or systemic administration. Preferred routes of administration include intravenous administration, subcutaneous administration, intramuscular administration, intratumoral administration, and the like.
 医薬組成物の投与量及び投与間隔は、投与対象の年齢、性別及び体重等、疾患の種類、進行度及び症状等、並びに投与方法等により、適宜選択することができる。投与量は、第1の態様のPD-1発現抑制剤の治療的有効量とすることができる。医薬組成物が(a)~(c)のいずれかの成分を含有する場合、治療的有効量としては、例えば、1回投与当たり、0.01μg~100mg、0.1μg~10mg、0.5μg~5mg、又は1μg~1mg等が挙げられる。医薬組成物が(d)の成分を含有する場合、治療的有効量としては、例えば、1回投与当たり、細胞の個数として10~1010個、10~10個、又は10~10個等が挙げられる。 The dosage and administration interval of the pharmaceutical composition can be appropriately selected depending on the age, sex, weight, etc. of the administration subject, the type, progression, symptoms, etc. of the disease, administration method, and the like. The dosage can be a therapeutically effective amount of the PD-1 expression inhibitor of the first aspect. When the pharmaceutical composition contains any one of components (a) to (c), the therapeutically effective amount is, for example, 0.01 μg to 100 mg, 0.1 μg to 10 mg, 0.5 μg per administration. up to 5 mg, or 1 μg to 1 mg, and the like. When the pharmaceutical composition contains component (d), the therapeutically effective amount is, for example, 10 3 to 10 10 cells, 10 4 to 10 9 cells, or 10 5 to 10 5 cells per administration. 10 8 etc. are mentioned.
 医薬組成物は、単回投与であってもよく、複数回投与であってもよい。複数回投与である場合、投与間隔は、例えば、1日毎、2~3日毎、1週毎、10~30日毎、1月毎、3~6月毎、又は1年毎等とすることができる。 The pharmaceutical composition may be a single dose or multiple doses. In the case of multiple administrations, the administration interval can be, for example, every day, every 2-3 days, every week, every 10-30 days, every month, every 3-6 months, or every year. .
[PD-1発現を抑制する方法]
 本開示の第3の態様は、リンパ球におけるPD-1発現を抑制する方法である。前記方法は、リンパ球を、IL-36又はその改変体の存在下で培養することを含む。
[Method for Suppressing PD-1 Expression]
A third aspect of the present disclosure is a method of inhibiting PD-1 expression in lymphocytes. The method includes culturing lymphocytes in the presence of IL-36 or a variant thereof.
 本実施形態の方法は、in vitro又はex vivoで行われる方法である。本実施形態の方法により、PD-1発現が低減されたリンパ球を製造することができる。 The method of this embodiment is a method performed in vitro or ex vivo. Lymphocytes with reduced PD-1 expression can be produced by the method of the present embodiment.
<IL-36又はその改変体>
 IL-36又はその改変体は、[PD-1発現阻害剤]で挙げたものと同様のものを用いることができる。
<IL-36 or variant thereof>
As IL-36 or a variant thereof, the same ones as listed in [PD-1 expression inhibitor] can be used.
<リンパ球>
 リンパ球は、PD-1を発現していれば、特に限定されない。リンパ球としては、[PD-1発現阻害剤]で挙げたものと同様のものが挙げられる。好ましいリンパ球としては、CD8陽性T細胞(CTL等)、CD4陽性T細胞(ヘルパーT細胞等)、及びこれらの改変細胞(CAR-T、TCR-T等)等が挙げられる。
 リンパ球は、複数種の細胞を含む細胞集団に含まれるものであってもよい。リンパ球を含む細胞集団としては、例えば、末梢血単核細胞(PBMC)、脾細胞、胸腺細胞等が挙げられる。
<Lymphocytes>
Lymphocytes are not particularly limited as long as they express PD-1. Examples of lymphocytes include those listed in [PD-1 expression inhibitor]. Preferred lymphocytes include CD8-positive T cells (CTL, etc.), CD4-positive T cells (helper T cells, etc.), modified cells thereof (CAR-T, TCR-T, etc.), and the like.
Lymphocytes may be included in a cell population containing multiple types of cells. Cell populations containing lymphocytes include, for example, peripheral blood mononuclear cells (PBMC), splenocytes, thymocytes, and the like.
<培養>
 培養は、IL-36又はその改変体の存在下で行う。より具体的には、IL-36又はその改変体を含有する培地で、リンパ球を培養する。
<Culture>
Culturing is performed in the presence of IL-36 or a variant thereof. More specifically, lymphocytes are cultured in a medium containing IL-36 or a variant thereof.
(培地)
 培地としては、例えば、動物細胞の培養に一般的に用いられる培地(以下、「動物細胞用培地」ともいう)に、IL-36又はその改変体を添加した培地が挙げられる。
(Culture medium)
The medium includes, for example, a medium commonly used for culturing animal cells (hereinafter also referred to as "animal cell medium") supplemented with IL-36 or a variant thereof.
 動物細胞用培地としては、動物細胞培養用の基礎培地が挙げられる。基礎培地としては、例えば、Doulbecco’s modified Eagle’s Medium(DMEM)培地、DMEM/F12培地、Advanced DMEM/F12培地、IMDM培地、Medium199培地、Eagle’sMinimum Essential Medium(EMEM)培地、αMEM培地、Ham’s F12培地、RPMI1640培地、Fischer’s培地、及びこれらの混合培地等が挙げられる。 Animal cell media include basal media for animal cell culture. Examples of basal media include Doulbecco's modified Eagle's Medium (DMEM) medium, DMEM/F12 medium, Advanced DMEM/F12 medium, IMDM medium, Medium 199 medium, Eagle's Minimum Essential Medium (EMEM) medium, αMEM medium, Examples include Ham's F12 medium, RPMI1640 medium, Fischer's medium, and mixed medium thereof.
 動物細胞用培地は、基礎培地に、任意の成分を添加したものであってもよい。添加成分としては、例えば、血清(牛胎児血清(FBS)など)、血清代替物等が挙げられる。血清代替物としては、例えば、アルブミン、トランスフェリン、亜セレン酸ナトリウム、ITS-X(Invitrogen)、ノックアウト血清代替物(Knockout Serum Replacement(KSR)、N2サプリメント(Invitrogen)、B27サプリメント(Invitrogen)、脂肪酸、インスリン、コラーゲン前駆体、微量元素、2-メルカプトエタノール、3’-チオールグリセロール等が挙げられる。添加成分としては、さらに、脂質、アミノ酸、L-グルタミン、Glutamax、非必須アミノ酸、ビタミン、増殖因子、抗生物質(ペニシリン、ストレプトマイシン等)、抗酸化剤、ピルビン酸、緩衝剤、無機塩類等が挙げられる。これらの添加成分は、1種を単独で用いてもよく、2種以上を併用してもよい。 The animal cell medium may be a basal medium to which any component has been added. Additional components include, for example, serum (fetal bovine serum (FBS), etc.), serum substitutes, and the like. Serum replacements include, for example, albumin, transferrin, sodium selenite, ITS-X (Invitrogen), knockout serum replacement (KSR), N2 supplement (Invitrogen), B27 supplement (Invitrogen), fatty acids, Insulin, collagen precursors, trace elements, 2-mercaptoethanol, 3′-thiolglycerol, etc. Additional components include lipids, amino acids, L-glutamine, Glutamax, non-essential amino acids, vitamins, growth factors, Antibiotics (penicillin, streptomycin, etc.), antioxidants, pyruvic acid, buffers, inorganic salts, etc. These additive components may be used singly or in combination of two or more. good.
 培地中のIL-36又はその改変体の濃度は、リンパ球のPD-1発現を抑制できる濃度であれば、特に限定されない。IL-36又はその改変体の濃度としては、例えば、0.001~1000ng/mLが挙げられる。培地中のIL-36又はその改変体の濃度の下限値としては、例えば、0.001ng/mL以上、0.003ng/mL以上、0.05ng/mL以上、0.07ng/mL以上、又は0.01ng/mL以上、0.03ng/mL以上、0.05ng/mL以上、0.07ng/mL以上、0.1ng/mL以上、0.3ng/mL以上、又は0.5ng/mL以上が挙げられる。培地中のIL-36又はその改変体の濃度の上限値としては、例えば、1000ng/mL以下、500ng/mL以下、300ng/mL以下、100ng/mL以下、70ng/mL以下、50ng/mL以下、40ng/mL以下、又は30ng/mL以下が挙げられる。これらの上限値及び下限値は任意に組合せ可能である。
 培地中のIL-36又はその改変体の濃度範囲としては、例えば、0.001~500ng/mL、0.005~300ng/mL、0.01~100ng/mL、0.01~50ng/mL、0.01~40ng/mL、0.01~30ng/mL、0.005~0.03ng/mL、0.3~40ng/mL、又は0.5~30ng/mL等が挙げられる。
The concentration of IL-36 or its variant in the medium is not particularly limited as long as it is a concentration capable of suppressing PD-1 expression in lymphocytes. Concentrations of IL-36 or variants thereof include, for example, 0.001 to 1000 ng/mL. The lower limit of the concentration of IL-36 or its variant in the medium is, for example, 0.001 ng/mL or more, 0.003 ng/mL or more, 0.05 ng/mL or more, 0.07 ng/mL or more, or 0 .01 ng/mL or greater, 0.03 ng/mL or greater, 0.05 ng/mL or greater, 0.07 ng/mL or greater, 0.1 ng/mL or greater, 0.3 ng/mL or greater, or 0.5 ng/mL or greater. be done. The upper limit of the concentration of IL-36 or its variant in the medium is, for example, 1000 ng/mL or less, 500 ng/mL or less, 300 ng/mL or less, 100 ng/mL or less, 70 ng/mL or less, 50 ng/mL or less, 40 ng/mL or less, or 30 ng/mL or less. These upper and lower limits can be combined arbitrarily.
The concentration range of IL-36 or its variants in the medium is, for example, 0.001 to 500 ng/mL, 0.005 to 300 ng/mL, 0.01 to 100 ng/mL, 0.01 to 50 ng/mL, 0.01 to 40 ng/mL, 0.01 to 30 ng/mL, 0.005 to 0.03 ng/mL, 0.3 to 40 ng/mL, or 0.5 to 30 ng/mL.
(培養条件)
 培養は、リンパ球の培養に一般的に用いられる培養条件で行うことができる。培養条件としては、例えば、培養温度32~40℃(好ましくは35~38℃、典型的には37℃)、CO濃度2~5%(好ましくは5%)が挙げられる。
(Culture conditions)
Culturing can be performed under culture conditions generally used for culturing lymphocytes. Culture conditions include, for example, a culture temperature of 32-40° C. (preferably 35-38° C., typically 37° C.) and a CO 2 concentration of 2-5% (preferably 5%).
 培養期間は、PD-1発現が抑制されるのに十分な期間であればよい。培養期間としては、例えば、1~150時間、5~100時間、10~80時間、15~70時間、20~60時間、25~50時間、又は30~50時間等が挙げられる。 The culture period should be sufficient to suppress PD-1 expression. Examples of the culture period include 1 to 150 hours, 5 to 100 hours, 10 to 80 hours, 15 to 70 hours, 20 to 60 hours, 25 to 50 hours, or 30 to 50 hours.
 培養期間中又は培養前に、リンパ球の活性化処理を行ってもよい。リンパ球の活性化処理は、公知の方法で行うことができる。リンパ球の活性化処理の方法としては、TCR/CD3複合体及び共刺激分子(CD28等)を刺激する方法が挙げられる。TCR/CD3複合体の刺激は、CD3結合物質を用いて行うことができる。共刺激分子の刺激は、共刺激分子結合物質を用いて行うことができる。CD3結合物質としては、抗CD3抗体又はその抗原結合断片が挙げられる。共刺激分子結合物質としては、抗CD28抗体又はその抗原結合断片が挙げられる。抗原結合断片とは、元の抗体の抗原結合性を維持する抗体断片である。抗原結合断片としては、例えば、Fab、Fab’、F(ab’)、Fv、scFv等が挙げられる。抗CD3抗体、及び抗CD28抗体は。モノクローナル抗体が好ましい。
 CD3結合物質及びCD28結合物質の存在下で、リンパ球を培養することにより、リンパ球の活性化処理を行うことができる。
Lymphocyte activation treatment may be performed during the culture period or before the culture. Lymphocyte activation treatment can be performed by a known method. Methods of activation treatment of lymphocytes include methods of stimulating the TCR/CD3 complex and co-stimulatory molecules (such as CD28). Stimulation of the TCR/CD3 complex can be done using a CD3 binding agent. A co-stimulatory molecule can be stimulated using a co-stimulatory molecule-binding substance. CD3 binding agents include anti-CD3 antibodies or antigen-binding fragments thereof. Co-stimulatory molecule binding agents include anti-CD28 antibodies or antigen-binding fragments thereof. An antigen-binding fragment is an antibody fragment that retains the antigen-binding properties of the original antibody. Antigen-binding fragments include, for example, Fab, Fab', F(ab') 2 , Fv, scFv and the like. Anti-CD3 antibody and anti-CD28 antibody. Monoclonal antibodies are preferred.
Lymphocyte activation treatment can be performed by culturing lymphocytes in the presence of a CD3-binding substance and a CD28-binding substance.
 リンパ球の活性化処理は、IL-36又はその改変体の存在下での培養と同時に行うことができる。この場合、例えば、CD3結合物質でコーティングしたウェル内で、IL-36若しくはその改変体とCD28結合物質とを含む培地を用いて、リンパ球を培養する方法が挙げられる。培地、培養条件、及び培養期間としては、上記と同様のものが挙げられる。  The activation treatment of lymphocytes can be performed simultaneously with culturing in the presence of IL-36 or its variants. In this case, for example, a method of culturing lymphocytes in wells coated with a CD3-binding substance using a medium containing IL-36 or a variant thereof and a CD28-binding substance can be mentioned. Examples of the medium, culture conditions, and culture period are the same as those described above.
 リンパ球の活性化処理は、IL-36又はその改変体の存在下での培養の前に行ってもよい。この場合、例えば、CD3結合物質でコーティングしたウェル内で、CD28結合物質を含む培地を用いて、リンパ球を培養する方法が挙げられる。培地、及び培養条件としては、上記と同様のものが挙げられる。培養期間としては、例えば、15~100時間、又は20~80時間等が挙げられる。リンパ球の活性化処理の後、リンパ球を回収する。次いで、回収したリンパ球を、IL-36又はその改変体の存在下での培養することにより、本実施形態の方法を実施することができる。 Lymphocyte activation treatment may be performed prior to culturing in the presence of IL-36 or its variants. In this case, for example, a method of culturing lymphocytes in a well coated with a CD3 binding substance using a medium containing a CD28 binding substance can be mentioned. Examples of the medium and culture conditions are the same as those described above. The culture period is, for example, 15 to 100 hours, or 20 to 80 hours. After lymphocyte activation treatment, lymphocytes are collected. The method of this embodiment can then be carried out by culturing the collected lymphocytes in the presence of IL-36 or a variant thereof.
 本実施形態の方法により、リンパ球のPD-1発現を抑制することができる。IL-36又はその改変体の非存在下で培養したリンパ球におけるPD-1発現量を100とした場合、本実施形態の方法により得られるリンパ球におけるPD-1の相対発現量は、例えば、90以下、80以下、70以下、60以下、50以下、又は40以下であり得る。 The method of the present embodiment can suppress PD-1 expression in lymphocytes. When the PD-1 expression level in lymphocytes cultured in the absence of IL-36 or a variant thereof is set to 100, the relative expression level of PD-1 in lymphocytes obtained by the method of the present embodiment is, for example, It can be 90 or less, 80 or less, 70 or less, 60 or less, 50 or less, or 40 or less.
 本実施形態の方法によれば、リンパ球のPD-1発現を抑制し、PD-1発現量が低減されたリンパ球を得ることができる。PD-1発現量が低減されたリンパ球は、生体に投与した場合に、免疫チェックポイントシステムを回避しやすい。そのため、免疫チェックポイントシステムに阻害されることなく、標的細胞(例えば、がん細胞)に対する細胞傷害活性を発揮することができる。 According to the method of the present embodiment, PD-1 expression in lymphocytes can be suppressed to obtain lymphocytes with a reduced PD-1 expression level. Lymphocytes with a reduced PD-1 expression level easily evade the immune checkpoint system when administered to a living body. Therefore, it can exhibit cytotoxic activity against target cells (for example, cancer cells) without being inhibited by the immune checkpoint system.
 本実施形態の方法で得られたリンパ球は、PD-1発現量が低減されているため、がん治療等に好適に用いることができる。本実施形態の方法は、PD-1発現量が低減されたリンパ球の作製方法であるともいえる。 The lymphocytes obtained by the method of the present embodiment have a reduced PD-1 expression level, so they can be suitably used for cancer therapy and the like. The method of the present embodiment can also be said to be a method for producing lymphocytes with reduced PD-1 expression level.
 一実施形態において、本発明は、PD-1発現抑制のために用いられる、前記(a)~(d)からなる群より選択される少なくとも1種の成分を提供する。 In one embodiment, the present invention provides at least one component selected from the group consisting of (a) to (d), which is used for PD-1 expression suppression.
 一実施形態において、本発明は、前記(a)~(d)からなる群より選択される少なくとも1種の成分を対象に投与することを含む、リンパ球のPD-1発現を抑制する方法を提供する。前記対象は、例えば、がん患者である。
 一実施形態において、本発明は、PD-1発現抑制剤の製造に用いられる、前記(a)~(d)からなる群より選択される少なくとも1種の成分を提供する。
 一実施形態において、本発明は、免疫チェックポイント阻害用医薬組成物の製造に用いられる、前記(a)~(d)からなる群より選択される少なくとも1種の成分を提供する。
 一実施形態において、本発明は、リンパ球におけるPD-1発現を抑制のための、前記(a)~(d)からなる群より選択される少なくとも1種の成分のin vitroでの使用を提供する。
In one embodiment, the present invention provides a method of suppressing PD-1 expression in lymphocytes, comprising administering to a subject at least one component selected from the group consisting of (a) to (d). offer. The subject is, for example, a cancer patient.
In one embodiment, the present invention provides at least one component selected from the group consisting of (a) to (d), which is used in the production of PD-1 expression inhibitors.
In one embodiment, the present invention provides at least one component selected from the group consisting of (a) to (d), which is used for the production of a pharmaceutical composition for immune checkpoint inhibition.
In one embodiment, the present invention provides in vitro use of at least one component selected from the group consisting of (a) to (d) for suppressing PD-1 expression in lymphocytes. do.
 本発明を実施例に基づいて説明する。ただし、本発明の実施態様は、これら実施例の記載に限定されるものではない。 The present invention will be explained based on examples. However, embodiments of the present invention are not limited to the description of these examples.
[実施例1]
(脾細胞の調製)
 10週齢のメスC57BL/6マウスから脾臓を摘出し、乾かさないように直ちに20mLのRPMI1640溶液(RPMI1640培養液)の中に浸した。RPMI1640溶液は、RPMI1640培地に、10%のfeat-inactivated fetal bovine serum(FBS)、0.1mMの非必須アミノ酸、100 IU/mlのペニシリン、および100μg/mLのストレプトマイシンを添加したものを用いた。細胞が傷つかないように軽くスライドガラスですりつぶして脾細胞浮遊液を作製した。脾細胞浮遊液を5mLピペットで吸い取り、50mL遠心管口に載せたポアサイズ40μmのセルストレーナーに通して結合組織片を除去した。
[Example 1]
(Preparation of splenocytes)
Spleens were removed from 10-week-old female C57BL/6 mice and immediately immersed in 20 mL of RPMI1640 solution (RPMI1640 culture medium) without drying. The RPMI1640 solution was RPMI1640 medium supplemented with 10% feat-inactivated fetal bovine serum (FBS), 0.1 mM non-essential amino acids, 100 IU/ml penicillin, and 100 μg/mL streptomycin. A spleen cell suspension was prepared by gently mashing the cells with a slide glass so as not to damage the cells. The splenocyte suspension was aspirated with a 5 mL pipette and passed through a cell strainer with a pore size of 40 μm placed on the mouth of a 50 mL centrifuge tube to remove connective tissue debris.
 50mL遠心管を270×g(約1,200回転)で5分間遠心して細胞を遠心管の底に集め、上清をアスピレータで吸引して除去した。細胞を、リン酸緩衝液(phosphate-buffered saline;PBS)で1回洗浄した後、予め室温に保っておいた赤血球除去用塩化アンモニウム溶液(Tris-buffered ammonium chloride)10mLを加えた。ピペットで軽くピペッティングして細胞のペレットをほぐし、細胞を完全に浮遊させ、室温で10分間反応させた。270×gで5分間遠心して細胞を沈殿させ、上清を吸引除去した。RPMI1640培養液10mLを加え、ピペットで細胞のペレットを丁寧にほぐして細胞を浮遊させた。次いで、50mL遠心管口に載せたポアサイズ40μmのセルストレーナーに通して赤血球の残骸を除去した。RPMI1640培養液10mLを加えてピペットで細胞を丁寧にほぐして細胞を浮遊させ、270×gで5分間遠心して細胞を沈殿させ、上清を吸引除去した。この洗浄操作を2回繰り返して、塩化アンモニウムを細胞から完全に除いた。2回の洗浄後、細胞をRPMI1640培養液20mLに浮遊させて、細胞浮遊液を調製した。 A 50 mL centrifuge tube was centrifuged at 270 xg (about 1,200 rpm) for 5 minutes to collect the cells at the bottom of the centrifuge tube, and the supernatant was removed by aspiration with an aspirator. After the cells were washed once with phosphate-buffered saline (PBS), 10 mL of Tris-buffered ammonium chloride for erythrocyte removal that had been kept at room temperature in advance was added. The cell pellet was loosened by gentle pipetting with a pipette, and the cells were completely suspended and allowed to react at room temperature for 10 minutes. Cells were pelleted by centrifugation at 270 xg for 5 minutes and the supernatant was aspirated off. 10 mL of RPMI1640 culture medium was added, and the cell pellet was gently loosened with a pipette to suspend the cells. Next, erythrocyte debris was removed by passing it through a cell strainer with a pore size of 40 μm placed on a 50 mL centrifugal tube mouth. 10 mL of RPMI1640 culture medium was added, the cells were gently loosened with a pipette to suspend the cells, the cells were precipitated by centrifugation at 270×g for 5 minutes, and the supernatant was removed by aspiration. This washing operation was repeated twice to completely remove ammonium chloride from the cells. After washing twice, the cells were suspended in 20 mL of RPMI1640 culture medium to prepare a cell suspension.
(脾細胞の刺激)
 血球計算板にカバーグラスを載せ、計算盤とカバーグラスの隙間に、マイクロピペットでトリパンブルーと混ぜた細胞浮遊液を静かに入れた。これを顕微鏡で観察し、生細胞数を計数した。計算した細胞密度を元に、細胞浮遊液の濃度が1.0×10個/mLになるよう調整した。15mLの遠心管に、8.0×10細胞/遠心管となるように細胞を入れ、270×gで5分間遠心して細胞を沈殿させ、上清を吸引除去した。
 5mLピペットで、0~50ng/mLのtruncated マウスIL-36β(配列番号12)を含有するRPMI1640溶液をそれぞれ加えて脾細胞を浮遊させた。truncated マウスIL-36βの濃度は、50nm/mL、30ng/mL、10ng/mL、5ng/mL、1ng/mL、0.5ng/mL、0.1ng/mL、0.05ng/mL、0.01ng/mL、および0ng/mLとした。予め0.1μgのrat anti-mouse CD3 monoclonal antibody(mAb)をコーティングしておいた12ウェルculture plateの各ウェルに、マウスIL-36βの各濃度群2ウェルずつとして、2mLの細胞浮遊液を加えた。最後に、2μg/mLの濃度になるようにrat anti-mouse CD28 mAbを加えた。次いで、5% CO、37℃の条件下で、48時間培養した。
(stimulation of splenocytes)
A cover glass was placed on the hemocytometer, and a cell suspension mixed with trypan blue was gently put into the gap between the counting board and the cover glass using a micropipette. This was observed under a microscope and the number of viable cells was counted. Based on the calculated cell density, the concentration of the cell suspension was adjusted to 1.0×10 6 cells/mL. Cells were placed in a 15 mL centrifuge tube at 8.0×10 6 cells/centrifuge tube, centrifuged at 270×g for 5 minutes to precipitate the cells, and the supernatant was removed by aspiration.
RPMI1640 solutions containing 0-50 ng/mL of truncated mouse IL-36β (SEQ ID NO: 12) were added with a 5 mL pipette to suspend the splenocytes. Concentrations of truncated mouse IL-36β were 50 nm/mL, 30 ng/mL, 10 ng/mL, 5 ng/mL, 1 ng/mL, 0.5 ng/mL, 0.1 ng/mL, 0.05 ng/mL, 0.01 ng /mL, and 0 ng/mL. To each well of a 12-well culture plate precoated with 0.1 μg of rat anti-mouse CD3 monoclonal antibody (mAb), 2 mL of cell suspension was added for each concentration group of mouse IL-36β. rice field. Finally, rat anti-mouse CD28 mAb was added to a concentration of 2 μg/mL. Then, it was cultured for 48 hours under conditions of 5% CO 2 and 37°C.
(PD-1発現解析)
 48時間培養後に、各群2ウェルから増殖した脾細胞を回収し、血球計算板にて細胞数を計数した。次いで、エッペンドルフチューブに、1.0×10細胞/チューブになるように加えた。270×g(約1,200回転)で5分間遠心して細胞をエッペンドルフチューブの底に集め、上清をアスピレータで吸引して除去した。0.1%アジ化ナトリウム含有PBSで2回洗浄後、100μLのPBSに細胞を浮遊させた。これに、rat anti-mouse CD16/32 mAbを加えて、Fc receptorをブロッキングした。次いで、APC-conjugated rat anti-mouse CD8a mAb、FITC-conjugated anti-mouse CD4 mAb、及びPE-conjugated anti-mouse CD279(PD-1) mAb若しくはPE-conjugated rat IgG2b κ Isotype control mAbを添加して、室温で、30分間反応させた。1mLのPBSで2回洗浄後、500μLのPBSに細胞を浮遊させ、フローサイトメーターを用いて、CD8陽性T細胞及びCD4陽性T細胞におけるPD-1発現を測定した。
(PD-1 expression analysis)
After culturing for 48 hours, proliferated splenocytes were collected from 2 wells of each group, and the number of cells was counted using a hemocytometer. Then added to Eppendorf tubes at 1.0×10 6 cells/tube. Cells were collected at the bottom of an Eppendorf tube by centrifugation at 270×g (approximately 1,200 rpm) for 5 minutes, and the supernatant was removed by aspiration with an aspirator. After washing twice with 0.1% sodium azide-containing PBS, the cells were suspended in 100 μL of PBS. To this, rat anti-mouse CD16/32 mAb was added to block the Fc receptor. Then, add APC-conjugated rat anti-mouse CD8a mAb, FITC-conjugated anti-mouse CD4 mAb, and PE-conjugated anti-mouse CD279 (PD-1) mAb or PE-conjugated rat bat IgG2b κ Isotype Acon, The reaction was allowed to proceed for 30 minutes at room temperature. After washing twice with 1 mL of PBS, the cells were suspended in 500 μL of PBS, and PD-1 expression in CD8-positive T cells and CD4-positive T cells was measured using a flow cytometer.
(結果)
 結果を図1~6に示す。図1は、0ng/mL、50ng/mL、又は30ng/mLのIL-36βの存在下で、脾細胞を培養したものである。図1中、「no stimulation」は、CD3 mAb及びCD28 mAbによる刺激を行わず、IL-36βを添加していないものである。図2は、10ng/mL、5ng/mL、又は1ng/mLのIL-36βの存在下で、脾細胞を培養したものである。図3は、図1~2のCD8陽性T細胞及びCD4陽性T細胞におけるPE(PD-1陽性)の平均蛍光強度(maen fluorescence intensity:MFI)を示す。図4は、0ng/mL、10ng/mL、1ng/mL、又は0.5ng/mLのIL-36βの存在下で、脾細胞を培養したものである。図5は、0.1ng/mL、0.05ng/mL、又は0.01ng/mLのIL-36βの存在下で、脾細胞を培養したものである。図6は、図4~5のCD8陽性T細胞におけるPE(PD-1陽性)の平均蛍光強度(maen fluorescence intensity:MFI)を示す。
(result)
The results are shown in Figures 1-6. FIG. 1 shows splenocytes cultured in the presence of 0 ng/mL, 50 ng/mL, or 30 ng/mL IL-36β. In FIG. 1, "no stimulation" means that stimulation with CD3 mAb and CD28 mAb was not performed, and IL-36β was not added. FIG. 2 shows splenocytes cultured in the presence of 10 ng/mL, 5 ng/mL, or 1 ng/mL IL-36β. FIG. 3 shows the mean fluorescence intensity (MFI) of PE (PD-1 positive) in CD8-positive T cells and CD4-positive T cells in FIGS. FIG. 4 shows splenocytes cultured in the presence of 0 ng/mL, 10 ng/mL, 1 ng/mL, or 0.5 ng/mL IL-36β. FIG. 5 shows splenocytes cultured in the presence of 0.1 ng/mL, 0.05 ng/mL, or 0.01 ng/mL IL-36β. FIG. 6 shows the mean fluorescence intensity (MFI) of PE (PD-1 positive) in the CD8-positive T cells of FIGS.
 図1~6に示す結果から、truncated IL-36βは、活性化T細胞(特に活性化CD8陽性T細胞)におけるPD-1の発現を抑制することが確認された。PD-1の発現は、比較的低濃度(例えば、5ng/ml、1ng/ml、0.5ng/ml)のtruncated IL-36βでも、強力に抑制された。 From the results shown in Figures 1 to 6, it was confirmed that truncated IL-36β suppresses the expression of PD-1 in activated T cells (especially activated CD8-positive T cells). PD-1 expression was strongly suppressed even at relatively low concentrations (eg, 5 ng/ml, 1 ng/ml, 0.5 ng/ml) of truncated IL-36β.
[実施例2]
(IL-36β発現メラノーマ細胞の作製)
 特開2021-70683号公報に記載の方法に従い、3種のレトロウイルスベクター(DFG-IRES-Neo、DFG-PTH-truncated IL-36β-IRES-Neo、DFG-GH-truncated IL-36β-IRES-Neo)を作製した。DFG-IRES-Neoは、neomycin phosphotransferase gene(Neo)のみを発現するレトロウイルスベクターである。DFG-PTH-truncated IL-36β-IRES-Neoは、ヒト副甲状腺ホルモンシグナル配列(配列番号13、14)が付加されたtruncated IL-36β(配列番号17、18)及びNeoを発現するレトロウイルスベクターである。DFG-GH-truncated IL-36β-IRES-Neoは、ヒト成長ホルモンシグナル配列(配列番号15、16)が付加されたtruncated IL-36β(配列番号19、20)及びNeoを発現するレトロウイルスベクターである。
[Example 2]
(Preparation of IL-36β-expressing melanoma cells)
According to the method described in JP-A-2021-70683, three retroviral vectors (DFG-IRES-Neo, DFG-PTH-truncated IL-36β-IRES-Neo, DFG-GH-truncated IL-36β-IRES- Neo) was produced. DFG-IRES-Neo is a retroviral vector that expresses only the neomycin phosphotransferase gene (Neo). DFG-PTH-truncated IL-36β-IRES-Neo is a retroviral vector expressing truncated IL-36β (SEQ ID NOS: 17, 18) to which a human parathyroid hormone signal sequence (SEQ ID NOS: 13, 14) is added and Neo. is. DFG-GH-truncated IL-36β-IRES-Neo is a retroviral vector that expresses truncated IL-36β (SEQ ID NOS: 19 and 20) to which a human growth hormone signal sequence (SEQ ID NOS: 15 and 16) is added and Neo. be.
 B16-F10メラノーマ細胞に、DFG-IRES-Neo、DFG-PTH-truncated IL-36β-IRES-Neo、及びDFG-GH-truncated IL-36β-IRES-Neoをそれぞれ導入して、B16-F10Neo細胞、B16-F10-PTH-truncated IL-36β細胞、及びB16-F10-GH-truncated IL-36β細胞をそれぞれ樹立した。 DFG-IRES-Neo, DFG-PTH-truncated IL-36β-IRES-Neo, and DFG-GH-truncated IL-36β-IRES-Neo were introduced into B16-F10 melanoma cells, respectively, to obtain B16-F10 Neo cells, B16-F10-PTH-truncated IL-36β cells and B16-F10-GH-truncated IL-36β cells were established, respectively.
(マウスへの細胞移植及び脾細胞の採取)
 B16-F10Neo細胞、B16-F10-PTH-truncated IL-36β細胞、又はB16-F10-GH-truncated IL-36β細胞を、10週齢の雌マウスC57BL/6(n=3)の腹部に接種した。接種量は、3×10細胞/マウスとした。細胞の接種後12日目に、各マウスの脾臓を採取した。
(Cell transplantation into mice and collection of splenocytes)
B16-F10Neo cells, B16-F10-PTH-truncated IL-36β cells, or B16-F10-GH-truncated IL-36β cells were inoculated into the abdomen of 10-week-old female C57BL/6 mice (n=3). . The inoculum amount was 3×10 5 cells/mouse. Twelve days after cell inoculation, the spleen of each mouse was harvested.
(PD-1発現解析)
 採取した脾臓を、実施例1の(脾細胞の調製)で記載した方法と同様に処理し、脾細胞の細胞浮遊液を調製した。
(PD-1 expression analysis)
The collected spleen was treated in the same manner as described in Example 1 (Preparation of splenocytes) to prepare a cell suspension of splenocytes.
 血球計算板にカバーグラスを載せ、計算盤とカバーグラスの隙間に、マイクロピペットでトリパンブルーと混ぜた細胞浮遊液を静かに入れた。これを顕微鏡で観察し、生細胞数を計数した。計算した細胞密度を元に、細胞浮遊液の濃度が1.0×10細胞/mLになるよう調整し、エッペンドルフチューブに1.0×10細胞/チューブになるように加えた。270×g(約1,200回転)で5分間遠心してエッペンドルフチューブの底に細胞を集め、上清をアスピレータで吸引除去した。0.1%アジ化ナトリウム含有PBSで2回洗浄後、100μLのPBSに浮遊させた。これに、rat anti-mouse CD16/32 mAb を加えてFc receptorをブロッキングした。次いで、PE-conjugated anti-mouse CD279(PD-1)mAb又はPE-conjugated rat IgG2b κ Isotype control mAbを加えて、室温で、30分間反応させた。1mLのPBSで2回洗浄後、500μLのPBSに細胞を浮遊させ、フローサイトメーターを用いて、脾細胞のPD-1発現を測定した。 A cover glass was placed on the hemocytometer, and a cell suspension mixed with trypan blue was gently put into the gap between the counting board and the cover glass using a micropipette. This was observed under a microscope and the number of viable cells was counted. Based on the calculated cell density, the concentration of the cell suspension was adjusted to 1.0×10 6 cells/mL, and added to the Eppendorf tube so that 1.0×10 6 cells/tube. Cells were collected at the bottom of an Eppendorf tube by centrifugation at 270×g (approximately 1,200 rpm) for 5 minutes, and the supernatant was aspirated off with an aspirator. After washing twice with 0.1% sodium azide-containing PBS, the cells were suspended in 100 μL of PBS. To this, rat anti-mouse CD16/32 mAb was added to block the Fc receptor. Then, PE-conjugated anti-mouse CD279 (PD-1) mAb or PE-conjugated rat IgG2b κ Isotype control mAb was added and allowed to react at room temperature for 30 minutes. After washing twice with 1 mL of PBS, the cells were suspended in 500 μL of PBS, and PD-1 expression in splenocytes was measured using a flow cytometer.
(結果)
 結果を図7に示す。B16-F10-PTH-truncated IL-36β細胞接種マウス又はB16-F10-GH-truncated IL-36β細胞接種マウスから採取した脾細胞は、B16-F10Neo細胞接種マウスから採取した脾細胞と比較して、PD-1発現が著明に低下していた。これらの結果から、IL-36βは、生体内でも、リンパ球におけるPD-1発現を抑制することが確認された。
(result)
The results are shown in FIG. Splenocytes collected from B16-F10-PTH-truncated IL-36β cell-inoculated mice or B16-F10-GH-truncated IL-36β cell-inoculated mice were compared with splenocytes collected from B16-F10Neo cell-inoculated mice, PD-1 expression was markedly reduced. These results confirmed that IL-36β suppresses PD-1 expression in lymphocytes even in vivo.
[実施例3]
(IL-36β発現腫瘍細胞の作製)
 MCA205細胞(マウス線維肉腫細胞)に、DFG-IRES-Neo、DFG-PTH-truncated IL-36β-IRES-Neo、及びDFG-GH-truncated IL-36β-IRES-Neoをそれぞれ導入して、MCA205Neo細胞、MCA205-PTH-truncated IL-36β細胞、及びMCA205-GH-truncated IL-36β細胞をそれぞれ樹立した。MC38細胞(マウス結腸腺癌細胞)に、DFG-IRES-Neo、DFG-PTH-truncated IL-36β-IRES-Neo、及びDFG-GH-truncated IL-36β-IRES-Neoをそれぞれ導入して、MC38Neo細胞、MC38-PTH-truncated IL-36β細胞、及びMC38-GH-truncated IL-36β細胞をそれぞれ樹立した。
[Example 3]
(Generation of IL-36β-expressing tumor cells)
DFG-IRES-Neo, DFG-PTH-truncated IL-36β-IRES-Neo, and DFG-GH-truncated IL-36β-IRES-Neo were each introduced into MCA205 cells (mouse fibrosarcoma cells) to obtain MCA205Neo cells. , MCA205-PTH-truncated IL-36β cells, and MCA205-GH-truncated IL-36β cells were established, respectively. DFG-IRES-Neo, DFG-PTH-truncated IL-36β-IRES-Neo, and DFG-GH-truncated IL-36β-IRES-Neo were each introduced into MC38 cells (mouse colon adenocarcinoma cells) to obtain MC38Neo. cells, MC38-PTH-truncated IL-36β cells, and MC38-GH-truncated IL-36β cells were established, respectively.
(マウスへの細胞移植及び腫瘍組織の採取)
 MCA205Neo細胞、MCA205-PTH-truncated IL-36β細胞、又MCA205-GH-truncated IL-36β細胞を、10週齢の雌マウスC57BL/6(n=3)の腹部に接種した。接種量は、1.5×10細胞/マウスとした。MC38Neo細胞、MC38-PTH-truncated IL-36β細胞、又MC38-GH-truncated IL-36β細胞を、10週齢の雌マウスC57BL/6(n=3)の腹部に接種した。接種量は、2×10細胞/マウスとした。接種後12日目に、各マウスから腫瘍組織を採取した。
(Cell transplantation into mice and collection of tumor tissue)
MCA205Neo cells, MCA205-PTH-truncated IL-36β cells, or MCA205-GH-truncated IL-36β cells were inoculated into the abdomen of 10-week-old female C57BL/6 mice (n=3). The inoculum amount was 1.5×10 5 cells/mouse. MC38Neo cells, MC38-PTH-truncated IL-36β cells, or MC38-GH-truncated IL-36β cells were inoculated into the abdomen of 10-week-old female C57BL/6 mice (n=3). The inoculum amount was 2×10 5 cells/mouse. Twelve days after inoculation, tumor tissue was harvested from each mouse.
(免疫染色)
 採取した腫瘍組織を、PBS緩衝ホルマリン溶液に2日間浸した後、パラフィンに包埋した。PD-1染色のために、脱パラフィンした組織から切片を作製した。これを、正常ヤギ血清で処理した後、ヤギ抗マウスPD-1抗体を4℃で一晩反応させた。PBSで洗浄後、ビオチン化ウシ抗ヤギIgG抗体を30分間反応させた。PBSで洗浄後、ペルオキシダーゼ結合ストレプトアビジンを含む溶液を作用させた。PBSで洗浄後、ジアミノベンジジンにより発色させた。
(Immunostaining)
The collected tumor tissue was immersed in a PBS-buffered formalin solution for 2 days and then embedded in paraffin. Sections were made from deparaffinized tissue for PD-1 staining. After treating this with normal goat serum, it was allowed to react with goat anti-mouse PD-1 antibody at 4° C. overnight. After washing with PBS, a biotinylated bovine anti-goat IgG antibody was reacted for 30 minutes. After washing with PBS, a solution containing peroxidase-conjugated streptavidin was applied. After washing with PBS, the color was developed with diaminobenzidine.
(結果)
 免疫染色の結果を図8に示す。MCA205-PTH-truncated IL-36β細胞接種マウス又はMCA205-GH-truncated IL-36β細胞接種マウスから採取した腫瘍組織では、MCA205Neo細胞接種マウスから採取した腫瘍組織と比較して、PD-1発現が著明に低下していた(図8上段)。MC38-PTH-truncated IL-36β細胞接種マウス又はMC38-GH-truncated IL-36β細胞接種マウスから採取した腫瘍組織では、MC38Neo細胞接種マウスから採取した腫瘍組織と比較して、PD-1発現が著明に低下していた(図8下段)。これらの結果は、truncated IL-36βを発現する腫瘍細胞を移植したマウスの腫瘍組織では、腫瘍に浸潤しているリンパ球におけるPD-1発現が低下することを示す。これらの結果から、IL-36βは、生体内でも、リンパ球におけるPD-1発現を抑制することが確認された。
(result)
FIG. 8 shows the results of immunostaining. PD-1 expression was significantly higher in tumor tissue collected from MCA205-PTH-truncated IL-36β cell-inoculated mice or MCA205-GH-truncated IL-36β cell-inoculated mice compared to tumor tissue collected from MCA205Neo cell-inoculated mice. It was clearly lowered (Fig. 8 upper). PD-1 expression was significantly higher in tumor tissue collected from MC38-PTH-truncated IL-36β cell-inoculated mice or MC38-GH-truncated IL-36β cell-inoculated mice compared to tumor tissue collected from MC38Neo cell-inoculated mice. It was clearly lowered (Fig. 8, bottom). These results demonstrate that PD-1 expression is reduced in tumor-infiltrating lymphocytes in tumor tissue from mice implanted with tumor cells expressing truncated IL-36β. These results confirmed that IL-36β suppresses PD-1 expression in lymphocytes even in vivo.
 本発明によれば、リンパ球におけるPD-1の発現を抑制可能な、PD-1発現抑制剤、免疫チェックポイント阻害用医薬組成物、及びPD-1発現を抑制する方法が提供される。 According to the present invention, a PD-1 expression inhibitor, a pharmaceutical composition for immune checkpoint inhibition, and a method for inhibiting PD-1 expression are provided, which are capable of suppressing PD-1 expression in lymphocytes.
 以上、本発明の好ましい実施形態を説明および図示してきたが、これらは本発明を例示するものであり、限定的なものとみなされるべきではないことを理解すべきである。本発明の精神または範囲から逸脱することなく、追加、省略、置換、およびその他の変更を行うことができる。したがって、本発明は、前述の説明によって限定されるものとはみなされず、添付の請求項の範囲によってのみ限定される。 While the preferred embodiments of the invention have been described and illustrated above, it should be understood that they are intended to be illustrative of the invention and should not be taken as limiting. Additions, omissions, substitutions, and other changes can be made without departing from the spirit or scope of the invention. Accordingly, the present invention should not be viewed as limited by the foregoing description, but only by the scope of the appended claims.

Claims (4)

  1.  下記(a)~(d)からなる群より選択される少なくとも1種の成分を含む、PD-1発現抑制剤:
     (a)インターロイキン36又はその改変体;
     (b)インターロイキン36又はその改変体をコードするヌクレオチド配列を有するポリヌクレオチド;
     (c)前記(b)のポリヌクレオチドを含むベクター;及び
     (d)インターロイキン36又はその改変体を分泌する細胞。
    A PD-1 expression inhibitor comprising at least one component selected from the group consisting of the following (a) to (d):
    (a) interleukin 36 or a variant thereof;
    (b) a polynucleotide having a nucleotide sequence encoding interleukin 36 or a variant thereof;
    (c) a vector comprising the polynucleotide of (b); and (d) a cell secreting interleukin-36 or a variant thereof.
  2.  CD8陽性T細胞又はCD4陽性T細胞におけるPD-1の発現を抑制する、請求項1に記載のPD-1発現抑制剤。 The PD-1 expression inhibitor according to claim 1, which suppresses PD-1 expression in CD8-positive T cells or CD4-positive T cells.
  3.  請求項1又は2に記載のPD-1発現抑制剤と、薬学的に許容される担体とを含有する、免疫チェックポイント阻害用医薬組成物。 A pharmaceutical composition for immune checkpoint inhibition, comprising the PD-1 expression inhibitor according to claim 1 or 2 and a pharmaceutically acceptable carrier.
  4.  リンパ球を、インターロイキン36又はその改変体の存在下で培養することを含む、リンパ球におけるPD-1発現を抑制する方法。 A method for suppressing PD-1 expression in lymphocytes, comprising culturing lymphocytes in the presence of interleukin 36 or a variant thereof.
PCT/JP2021/036056 2021-09-30 2021-09-30 Pd-1 expression inhibitor, pharmaceutical composition for immune checkpoint inhibition, and method for inhibiting pd-1 expression WO2023053315A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/JP2021/036056 WO2023053315A1 (en) 2021-09-30 2021-09-30 Pd-1 expression inhibitor, pharmaceutical composition for immune checkpoint inhibition, and method for inhibiting pd-1 expression

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/JP2021/036056 WO2023053315A1 (en) 2021-09-30 2021-09-30 Pd-1 expression inhibitor, pharmaceutical composition for immune checkpoint inhibition, and method for inhibiting pd-1 expression

Publications (1)

Publication Number Publication Date
WO2023053315A1 true WO2023053315A1 (en) 2023-04-06

Family

ID=85781566

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2021/036056 WO2023053315A1 (en) 2021-09-30 2021-09-30 Pd-1 expression inhibitor, pharmaceutical composition for immune checkpoint inhibition, and method for inhibiting pd-1 expression

Country Status (1)

Country Link
WO (1) WO2023053315A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108524918A (en) * 2018-05-09 2018-09-14 苏州大学 A kind of anti-tumor compositions and its expression vector and application
CN110075275A (en) * 2019-05-15 2019-08-02 苏州云海生物技术有限公司 The application of IL-36 β
JP2021070683A (en) * 2019-10-30 2021-05-06 国立大学法人東北大学 Antitumor drug for fibrosarcoma, pharmaceutical composition for treating fibrosarcoma, antitumor effect potentiator, and pharmaceutical composition used for potentiating antitumor effect on fibrosarcoma

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108524918A (en) * 2018-05-09 2018-09-14 苏州大学 A kind of anti-tumor compositions and its expression vector and application
CN110075275A (en) * 2019-05-15 2019-08-02 苏州云海生物技术有限公司 The application of IL-36 β
JP2021070683A (en) * 2019-10-30 2021-05-06 国立大学法人東北大学 Antitumor drug for fibrosarcoma, pharmaceutical composition for treating fibrosarcoma, antitumor effect potentiator, and pharmaceutical composition used for potentiating antitumor effect on fibrosarcoma

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
DING GUOPING, SHEN TAO, YAN CHEN, ZHANG MINGJIE, WU ZHENGRONG, CAO LIPING: "IFN-γ down-regulates the PD-1 expression and assist nivolumab in PD-1-blockade effect on CD8+ T-lymphocytes in pancreatic cancer", BMC CANCER, vol. 19, no. 1, 1 December 2019 (2019-12-01), XP093054651, DOI: 10.1186/s12885-019-6145-8 *
QUEEN DAWN, EDIRIWEERA CHATHUMADAVI, LIU LIANG: "Function and Regulation of IL-36 Signaling in Inflammatory Diseases and Cancer Development", FRONTIERS IN CELL AND DEVELOPMENTAL BIOLOGY, vol. 7, XP093054646, DOI: 10.3389/fcell.2019.00317 *
WANG XUEFENG; ZHAO XIN; FENG CHAO; WEINSTEIN ALIYAH; XIA RUI; WEN WEN; LV QUANSHENG; ZUO SHUTING; TANG PEIJUN; YANG XI; CHEN XIAOJ: "IL-36y Transforms the Tumor Microenvironment and Promotes Type 1 Lymphocyte-Mediated Antitumor Immune Respo", CANCER CELL, vol. 28, no. 3, 27 August 2015 (2015-08-27), US , pages 296 - 306, XP029268946, ISSN: 1535-6108, DOI: 10.1016/j.ccell.2015.07.014 *
WEINSTEIN ALIYAH M., CHEN LU, BRZANA EMILY A., PATIL PRASHANTI R., TAYLOR JENNIFER L., FABIAN KELLSYE L., WALLACE CALLEN T., JONES: "Tbet and IL-36γ cooperate in therapeutic DC-mediated promotion of ectopic lymphoid organogenesis in the tumor microenvironment", ONCOIMMUNOLOGY, vol. 6, no. 6, 3 June 2017 (2017-06-03), XP093054638, DOI: 10.1080/2162402X.2017.1322238 *
WEINSTEIN ALIYAH M.; GIRALDO NICOLAS A.; PETITPREZ FLORENT; JULIE CATHERINE; LACROIX LAETITIA; PESCHAUD FRéDéRIQUE; EMIL: "Association of IL-36γ with tertiary lymphoid structures and inflammatory immune infiltrates in human colorectal cancer", CANCER IMMUNOLOGY IMMUNOTHERAPY, vol. 68, no. 1, 12 October 2018 (2018-10-12), Berlin/Heidelberg , pages 109 - 120, XP036671532, ISSN: 0340-7004, DOI: 10.1007/s00262-018-2259-0 *
ZHAO XIN, CHEN XIAOJUAN, SHEN XINGHUA, TANG PEIJUN, CHEN CHEN, ZHU QITAI, LI MUYAO, XIA RUI, YANG XI, FENG CHAO, ZHU XINGUO, ZHU Y: "IL-36β Promotes CD8+ T Cell Activation and Antitumor Immune Responses by Activating mTORC1", FRONTIERS IN IMMUNOLOGY, vol. 10, 1 January 2019 (2019-01-01), XP093054648, DOI: 10.3389/fimmu.2019.01803 *

Similar Documents

Publication Publication Date Title
JP6989634B2 (en) Chimeric antigen receptor targeting B cell maturation antigen
US20230355677A1 (en) Tethered interleukin-15 and interleukin-21
JP2021516996A (en) Biologically related orthogonal cytokine / receptor pair
CA3099039A1 (en) Interleukin 15 fusion proteins, and compositions and therapeutic methods thereof
CN109320615B (en) Chimeric antigen receptor targeting novel BCMA and uses thereof
WO2017159736A1 (en) Immunocompetent cell and expression vector expressing regulatory factors of immune function
CN111511911B (en) Immunocompetent cells expressing cell surface molecules, IL-7 and CCL19 that specifically recognize human mesothelin
JP6761113B2 (en) Chimeric antigen receptor
CN115151635A (en) Gene-modified NK cell lines transduced with genes encoding novel chimeric antigen receptors and uses thereof
CN115885038A (en) Immunocompetent cells expressing chimeric antigen receptors
CN115397973A (en) CAR-expressing NK cell ablation of BCMA-positive malignancies
JP2021512637A (en) Cyclin A1-specific T cell receptor and its use
US20210238258A1 (en) Chimeric orthogonal receptor proteins and methods of use
WO2021207274A2 (en) Human immune cells genomically modified to express orthogonal receptors
WO2023053315A1 (en) Pd-1 expression inhibitor, pharmaceutical composition for immune checkpoint inhibition, and method for inhibiting pd-1 expression
WO2019184886A1 (en) Method for promoting immune cell proliferation
CN114457117B (en) Dendritic cell tumor vaccine and use thereof
CN111378046A (en) Immune effector cell conversion receptor
KR20240013750A (en) T cell receptor targeting RAS mutations and uses thereof
JP7475088B2 (en) Immunocompetent cells expressing cell surface molecules that specifically recognize human mesothelin, IL-7, and CCL19
US11364267B1 (en) Bi-specific targeting human NKG2DL and CLDN18A2 chimeric antigen receptor cells, preparation method and application thereof
WO2023276395A1 (en) Chimeric cytokine receptor
US20240018213A1 (en) Modulators of notch signaling and methods of use thereof
WO2024015723A1 (en) Tunable cytokine receptor signaling domains
KR20240035506A (en) A chimeric antigen receptor, a cell expressing the receptor, a pharmaceutical composition containing the cell, a method for producing the cell, and a polynucleotide or vector containing a base sequence encoding the chimeric antigen receptor.

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: 21959357

Country of ref document: EP

Kind code of ref document: A1