WO2021112196A1 - Composition pour supprimer la perte de tissu musculaire - Google Patents

Composition pour supprimer la perte de tissu musculaire Download PDF

Info

Publication number
WO2021112196A1
WO2021112196A1 PCT/JP2020/045116 JP2020045116W WO2021112196A1 WO 2021112196 A1 WO2021112196 A1 WO 2021112196A1 JP 2020045116 W JP2020045116 W JP 2020045116W WO 2021112196 A1 WO2021112196 A1 WO 2021112196A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
receptor
composition according
composition
inhibits
Prior art date
Application number
PCT/JP2020/045116
Other languages
English (en)
Japanese (ja)
Inventor
浩徳 原田
嘉宏 林
Original Assignee
学校法人東京薬科大学
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 学校法人東京薬科大学 filed Critical 学校法人東京薬科大学
Priority to JP2021562730A priority Critical patent/JPWO2021112196A1/ja
Publication of WO2021112196A1 publication Critical patent/WO2021112196A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology

Definitions

  • the present invention relates to a composition for suppressing a decrease in muscle tissue.
  • Cachexia is a complex metabolic disorder syndrome that is associated with various diseases. Cachexia is particularly present in 50-80% of all cancer patients and is directly linked to reduced treatment tolerance and poor quality of life in patients. In cancer patients, weight loss, malnutrition, and exhaustion gradually progress as the condition progresses, and such a condition in cancer patients is particularly referred to as "cancer cachexia”. Called. Because patients with cancer cachexia often have anorexia, they are sometimes referred to as cancerous anorexia cachexia syndrome (CACS).
  • CACS cancerous anorexia cachexia syndrome
  • weight loss especially loss of muscle mass.
  • Weight loss is particularly expressed in lean body mass (LBM), where LBM is maintained in the case of normal starvation weight loss, whereas LBM is reduced in cachexia and cachexia. And hunger differ in this respect.
  • LBM lean body mass
  • Non-Patent Document 1 Although cachexia has been suggested to be indirectly involved in about 20% of cancer deaths (Non-Patent Document 1), its pathogenic mechanism has not been fully elucidated. For example, the relationship between systemic inflammation and metabolic disorders has been pointed out, but the detailed molecular mechanism of pathogenesis is still unknown.
  • Anamorelin is a ghrelin-like agent that promotes appetite. This method attempts to treat cancer cachexia mainly by increasing the appetite of the patient (Patent Document 1, Patent Document 2, Patent Document 3).
  • An object of the present invention is to provide a composition for suppressing a decrease in muscle tissue.
  • CMML Chronic myelomonocytic leukemia
  • CMML chronic myelomonocytic leukemia
  • the present inventors have succeeded in establishing a disease model mouse that faithfully reproduces the pathological condition of CMML using a novel NUP98-HBO1 fusion gene identified from a CMML patient (Hayashi, et al. Blood Advances. 2019).
  • NUP98-HBO1 CMML model mice Based on the idea that the increase in inflammatory monocytes specific to CMML may be involved in the development of malaise, which is also prominent in CMML, the present inventors have selected NUP98-HBO1 CMML model mice. Interestingly, the increased inflammatory monocytes in this model mouse are different from those in wild-type mice. It was revealed that they show different gene expression patterns (Figs. 1 to 3). Furthermore, a comparison between NUP98-HBO1 CMML model mice also revealed that there is a difference in the gene expression pattern of inflammatory monocytes depending on the presence or absence of cachexia (Fig. 5).
  • IL1F9 IL-36 ⁇
  • IL-36 ⁇ IL1F9 derived from monocyte lineage cells induces muscle atrophy.
  • the present invention is: [1] A composition for suppressing a decrease in muscle tissue in a subject having monocytes in which the expression of CD38 is enhanced.
  • a composition comprising a compound that inhibits the activity of the IL-36 receptor; [2] The composition according to [1], wherein the subject is a mammal; [3] The composition according to [1] or [2], wherein the mammal is selected from the group consisting of humans, monkeys, mice, rats, cows, horses, sheep, dogs and cats; [4] The composition according to [2] or [3], wherein the mammal is a human; [5] A subject having monocytes with enhanced expression of CD38 suffers from at least one selected from the group consisting of cancer, inflammatory diseases, autoimmune diseases and chronic infectious diseases [1].
  • the blood cancer is at least one selected from the group consisting of malignant lymphoma, multiple myeloma, leukemia and myeloproliferative neoplasm.
  • the composition according to [6], wherein the solid cancer is at least one selected from the group consisting of breast cancer, colon cancer, malignant melanoma, lung cancer, pancreatic cancer, gastric cancer, and lung cancer; [8]
  • the blood cancer is chronic myelomonocytic leukemia.
  • composition according to [6] or [7], wherein the solid cancer is breast cancer; [9] The composition according to any one of [1] to [8], wherein the subject has cancer-related symptoms; [10] The composition according to [9], wherein the cancer-related symptom is cancer cachexia; [11] The compound that inhibits the activity of the IL-36 receptor is ⁇ CD38 antagonist, The composition according to any one of [1] to [10], which is at least one selected from the group consisting of an IL-36 ⁇ antagonist and an IL-36 receptor antagonist; [12] The compound that inhibits the activity of the IL-36 receptor is ⁇ Anti-CD38 antibody, The composition according to any one of [1] to [11], which is at least one selected from the group consisting of an anti-IL-36 ⁇ antibody and an anti-IL-36 receptor antibody.
  • the IL-36 ⁇ was produced by monocytes expressing CD38.
  • the composition comprises a compound that inhibits the activity of the IL-36 receptor;
  • the IL-36 receptor activation pathway comprises a monocyte expressing CD38 and an IL-36 receptor.
  • the composition comprises a compound that inhibits the activity of the IL-36 receptor; [17] The composition according to [16], wherein the IL-36 receptor activation pathway comprises that monocytes expressing CD38 produce IL-36 ⁇ and IL-36 ⁇ binds to the IL-36 receptor. ; [18] The compound that inhibits the activity of the IL-36 receptor is ⁇ CD38 antagonist, The composition according to any one of [15] to [17], which is at least one selected from the group consisting of an IL-36 ⁇ antagonist and an IL-36 receptor antagonist.
  • FIG. 1 shows a novel fusion gene NUP98-HBO1 identified from a patient with chronic myelomonocytic leukemia (CMML).
  • FIG. 2A shows a characteristic increase in inflammatory monocytes in CMML model mice obtained by bone marrow transplantation (Bone Marlow Transplant: BMT) of cells into which the novel fusion gene NUP98-HBO1 has been introduced into wild-type mice. Show that.
  • FIG. 2B shows a marked increase in leukocytes (White Blood Cell: WBC) in CMML model mice obtained by bone marrow transplantation (Bone Marlow Transplant: BMT) of cells into which the novel fusion gene NUP98-HBO1 was introduced into wild-type mice.
  • WBC White Blood Cell
  • FIG. 2C shows that megakaryocyte dysplasia was remarkably observed in CMML model mice obtained by bone marrow transplantation (Bone Marlow Transplant: BMT) of cells into which the novel fusion gene NUP98-HBO1 was introduced into wild-type mice. Is shown.
  • FIG. 2D shows a CMML model mouse obtained by bone marrow transplantation (Bone Marlow Transplant: BMT) of cells into which a novel fusion gene NUP98-HBO1 was introduced into a wild-type mouse, and the increase in blasts in the bone marrow was less than 20%. Show that.
  • FIG. 2E shows that the gene group (NUP98-HBO1 inducing gene) whose expression was upregulated when NUP98-HBO1 was introduced into human cord blood hematopoietic stem cells / progenitor cells was significantly upregulated in the CMML patient group (NUP98-HBO1 inducible gene).
  • FIG. 3A shows that cachexia (progressive weight loss and skeletal muscle atrophy), which is a characteristic symptom of CMML, was observed in the CMML mice established by the present inventors.
  • FIG. 3B shows that cachexia (progressive weight loss and skeletal muscle atrophy), which is a characteristic symptom of CMML, was observed in the CMML mice established by the present inventors.
  • FIG. 3A shows that cachexia (progressive weight loss and skeletal muscle atrophy), which is a characteristic symptom of CMML, was observed in the CMML mice established by the present inventors.
  • FIG. 3B shows that cachexia (progressive weight loss and skeletal muscle atrophy
  • FIG. 4A shows that whole bone marrow cells of NUP98-HBO1 mice and control mice or Ly6C-positive inflammatory monocytes collected by a cell sorter were co-cultured with C2C12 cells differentiated into muscle fibrous cells.
  • FIG. 4B shows the measurement results of the width of C2C12 muscle fiber cells after inserting the culture insert and co-culturing for 72 hours under the condition that the cells do not come into direct contact with each other.
  • FIG. 5 shows a specific gene expression pattern of cachexia-onset CMML mouse monocytes.
  • FIG. 6A shows the results of RNA-Seq data analysis of gene expression in cachexia-onset CMML mouse monocytes.
  • FIG. 6B shows the results of flow cytometric analysis of the actual protein expression of some of the gene candidates with increased expression shown in FIG. 6A. It was revealed that the expression of CD38 was enhanced in the inflammatory monocytes of cachexia-onset CMML mice.
  • FIG. 7A shows the results of RNA-Seq data analysis of gene expression in cachexia-onset CMML mouse monocytes.
  • IL1F9 IL-36 ⁇ was focused on as a gene whose expression is upregulated only in CMML mice that have developed cachexia.
  • FIG. 7B shows NF ⁇ B signaling involving IL1F9 (IL-36 ⁇ ) and its receptors (IL-1Rrp2 and IL-1RAcP).
  • FIG. 8 shows the level of IL-36 ⁇ in the peripheral blood (plasma) of cachexia-onset CMML mice measured by the ELISA method.
  • FIG. 9 shows the verification and results of the induction of muscular atrophy of IL-36 ⁇ derived from monocyte cells.
  • FIG. 10A shows subcutaneous transplantation of 4T1 cells into wild-type BALB / c mice.
  • FIG. 10B shows the measurement results of the body weight of the 4T1 cell transplanted mouse.
  • FIG. 10C shows the results of measuring the proportion of the CD38-positive population in peripheral blood monocytes (CD115-positive) of 4T1 cell-transplanted mice using flow cytometry.
  • the first aspect of the present invention relates to a composition for suppressing the decrease in muscle tissue in a subject having monocytes in which the expression of CD38 is enhanced.
  • the composition contains a compound that inhibits the activity of the IL-36 receptor.
  • a subject having monocytes with enhanced expression of CD38 refers to a subject whose expression level of CD38 on the surface of monocytes is measured to be increased as compared with a healthy subject. .. Elevated expression levels of CD38 can be detected by methods known in the art, such as flow cytometry, immune cell staining, real-time RT-PCR and the like.
  • the expression level of CD38 on the surface of monocytes is, for example, more than 1.0 times, preferably 1.2 times or more, more preferably, as compared with a healthy subject.
  • the "target” is a vertebrate, preferably a mammal.
  • the term “mammal” is used to refer to any animal that is classified as a mammal, including, for example, humans, monkeys, mice, rats, cows, horses, sheep, dogs, and cats. However, it is not limited to these.
  • the preferred mammal is a human.
  • Muscle tissue loss means that at least one value of muscle tissue volume and weight is reduced, and “suppressing muscle tissue loss” is meant of muscle tissue volume and weight. Means to maintain or increase at least one value of. Muscle tissue volume and weight are measured according to methods known in the art. In the subject, evaluation of whether or not "loss of muscle tissue” is suppressed includes, for example, measuring the width of muscle tissue, measuring the weight of muscle tissue, measuring the volume of muscle tissue, and the like. It can be carried out by a method known in the art. For example, the width of muscle tissue is measured by taking an image while observing the cell morphology with an inverted microscope and measuring the width of muscle fiber cells in the photographed image.
  • the term "compound that inhibits the activity of the IL-36 receptor” is used in the broadest sense, and is used in the pathway that causes the activation of the IL-36 receptor (hereinafter, also referred to as "IL-36 receptor activation pathway”). It is intended to include compounds that inhibit the biological activity of any one or more of the involved compounds and cells.
  • the "IL-36 receptor activation pathway” comprises a monocyte expressing CD38 and an IL-36 receptor, preferably a monocyte expressing CD38, IL-36 ⁇ , and the like. It contains the IL-36 receptor. More specifically, the "IL-36 receptor activation pathway” includes a pathway in which monocytes expressing CD38 produce IL-36 ⁇ and the produced IL-36 ⁇ binds to the IL-36 receptor. To say.
  • the "IL-36 receptor” is IL-1Rrp2 (also referred to as “1L-1RL2" or “1L1RL2”).
  • the IL-36 receptor ligands (IL-36 ⁇ , IL-36 ⁇ and IL-36 ⁇ ) bind to the IL-36 receptor, and the ligand-bound IL-36 receptor is an IL-36 receptor accessory. It has been reported to initiate an intracellular signaling cascade by forming a heterodimer with a protein (IL-1RAcP) (see BLOOD, 7 APRIL 2011, VOLUME 117, NUMBER 14).
  • the present inventors have enhanced expression of CD38 on the surface of inflammatory monocytes in mice with cancer cachexia symptoms, and further, IL1F9 (IL) produced by such inflammatory monocytes.
  • IL1F9 IL
  • -36 ⁇ induces muscle atrophy, specifically, the binding of IL1F9 (IL-36 ⁇ ) to the transmembrane IL-36 receptor induces muscle atrophy via intracellular signal transduction.
  • the findings obtained by the present inventors are to target a pathway that causes activation of the IL-36 receptor (“IL-36 receptor activation pathway”) in the treatment of a subject who develops cachexia. It shows its usefulness. Therefore, the terms "compounds involved in the pathway that causes the activation of the IL-36 receptor” and “cells involved in the pathway that causes the activation of the IL-36 receptor” include, for example, expressing CD38 and CD38 on the surface. Monocytes, IL-36 receptor ligands (IL-36 ⁇ , IL-36 ⁇ and IL-36 ⁇ , especially IL-36 ⁇ ), IL-36 receptor, cells having IL-36 receptor and the like are included.
  • the "compound that inhibits the activation of the IL-36 receptor” includes the above-mentioned compound and a compound that inhibits the activity of at least one of the cells, and includes, for example, a CD38 antagonist, an IL-36 ⁇ antagonist, and an IL-36 receptor antagonist. Can be mentioned.
  • an “antagonist” is used in the broadest sense.
  • An “antagonist” is a molecule that partially or completely blocks, inhibits, neutralizes, defends and / or interferes with the biological activity of a target molecule, regardless of the underlying mechanism of action.
  • the "biological activity" of the target molecule "CD38” is, for example, the ability of CD38 to bind a ligand and IL-36 ⁇ in monocytes expressing CD38 on the surface. , IL-36 ⁇ and IL-36 ⁇ , in particular the ability to induce the production of IL-36 ⁇ .
  • the CD38 antagonist specifically recognizes and binds to CD38, for example, antibody-dependent cellular cytotoxicity (ADCC) activity and complement-dependent cellular cytotoxicity (ADCC) activity due to activation of immune system cells.
  • ADCC antibody-dependent cellular cytotoxicity
  • ADCC complement-dependent cellular cytotoxicity
  • CD38 antagonists for example, incubate monocytes expressing CD38 on the surface in the presence and absence of test compounds, further incubate with serum containing effector cells or complement, and in culture with cytotoxicity. It can be identified by measuring substances released in the clear, such as LDH. If the concentration of the substance released with cytotoxicity is higher in the presence of the test compound than in the absence of the test compound, the test compound is a CD38 antagonist.
  • CD38 antagonists can also, for example, incubate monocytes expressing CD38 on the surface in the presence and absence of test compounds and monitor IL-36 ⁇ levels in cell culture supernatants, for example by ELISA. Yes, if the IL-36 ⁇ level is lower in the presence of the test compound than in the absence of the test compound, then the test compound is a CD38 antagonist.
  • real-time RT-PCR can be used before and after treatment with the test compound to monitor the expression of IL-36 ⁇ mRNA in tissues expressing CD38. Decreased IL-36 ⁇ mRNA levels in the presence of the test compound indicates that the compound is a CD38 antagonist.
  • CD38 antagonists include neutralizing antibodies to whole or native CD38 polypeptide subunits, immunoadhesion factors containing CD38 subunit fused to immunoglobulin constant region sequences, small molecules, native CD38 polypeptides.
  • the amino acid sequence of the natural CD38 polypeptide can be obtained from a database known in the art.
  • the amino acid sequence of the natural CD38 polypeptide include SEQ ID NO: 1 (Human CD38, NCBI Reference Sequence: NP_001766.2) and SEQ ID NO: 2 (Musmusculus CD38, NCBI Reference Sequence: NP_0312) shown below. .. Therefore, the CD38 antagonist includes an antibody and a fragment thereof that specifically recognize a polypeptide having such a sequence, and a certain sequence identity (for example, 80% or more, 85% or more, or 90% or more) with the sequence. Antibodies that specifically recognize polypeptides having (sequence identity) and fragments thereof are included.
  • the nucleotide sequences of the genes encoding the natural CD38 polypeptide include, for example, SEQ ID NO: 3 (Human CD38; NCBI Reference Sequence: NM_001775.4 (Coding sequence, CDS)) and SEQ ID NO: 4 (Mus musculus CD38). NCBI Reference Sequence: NM_007646.5 (Coding sequence, CDS). Therefore, for CD38 antagonists, -A gene encoding a polypeptide having the above amino sequence, A gene encoding a polypeptide having a certain sequence identity (eg, 80% or more, 85% or more, or 90% or more sequence identity) with the amino sequence as described above.
  • -Genes with the above base sequence -Inhibiting at least one of translation and transcription of a gene having a base sequence having a certain sequence identity (for example, 80% or more, 85% or more, or 90% or more sequence identity) with the above-mentioned base sequence.
  • Nucleic acid molecules that can be produced are included.
  • the "CD38 antagonist” may include those known in the art.
  • Examples thereof include anti-CD38 antibodies and fragments thereof described in Japanese Patent Application Laid-Open No. 164788, and commercially available products and investigational agents having a function as anti-CD38 antibodies.
  • Examples of commercially available products and investigational agents include daratumumab, isatuximab, MOR-202, and Dalazarex (registered trademark) (Jansen Pharma Co., Ltd.), which is a human anti-CD38 monoclonal antibody.
  • the "biological activity" of the target molecule IL-36 ⁇ is, for example, the ability to bind to the IL-36 receptor.
  • IL-36 ⁇ antagonists are, for example, the ability to specifically recognize IL-36 ⁇ and neutralize IL-36 ⁇ , or the IL-36 receptor of IL-36 ⁇ by specifically recognizing and / or neutralizing IL-36 ⁇ . It may be identified based on its ability to inhibit or block binding to.
  • IL-36 ⁇ antagonists can be identified, for example, by adding a test compound to the culture supernatant of IL-36 ⁇ -producing cells and monitoring IL-36 ⁇ levels in the cell culture supernatant, eg, by ELISA. If the IL-36 ⁇ level is lower in the presence of the test compound than in the absence of the test compound, then the test compound is an IL-36 ⁇ antagonist.
  • IL-36 ⁇ antagonists include neutralizing antibodies to the entire native IL-36 ⁇ polypeptide or the native IL-36 ⁇ polypeptide subunit, an immunoadhesion factor comprising the IL-36 ⁇ subunit fused to the immunoglobulin constant region sequence. Inhibits at least one of the translation and transcription of a small molecule, an aptamer that binds to the whole or native IL-36 ⁇ polypeptide subunit, or a gene encoding the whole or subunit of the native IL-36 ⁇ polypeptide. Can include, but are not limited to, nucleic acid molecules such as siRNA, antisense subunits, decoys, ribozymes, and the like.
  • the sequence of the natural IL-36 ⁇ polypeptide can be obtained from a database known in the art.
  • Examples of the sequence of the natural IL-36 ⁇ polypeptide include, as shown below, SEQ ID NO: 5 (Human IL1F9; NCBI Reference Sequence: NP_062564.1) and SEQ ID NO: 6 (Musmusculus IL1F9; NCBI ReferenceSequen) Can be mentioned. Therefore, the IL-36 ⁇ antagonist includes an antibody and a fragment thereof that specifically recognize a polypeptide having such a sequence, and a certain sequence identity with the sequence (for example, 80% or more, 85% or more, or 90). % Or more sequence identity) includes antibodies that specifically recognize polypeptides and fragments thereof.
  • the nucleotide sequence of the gene encoding the natural IL-36 ⁇ polypeptide is, for example, SEQ ID NO: 7 (Human IL1F9; NCBI Reference Sequence: NM_019618.4 (Coding sequence, CDS)) and SEQ ID NO: 8 (Mus) shown below. Musculus IL1F9; NCBI Reference Sequence: NM_153511.3 (Coding sequence, CDS)). Therefore, for IL-36 ⁇ antagonists, -A gene encoding a polypeptide having the above amino sequence, A gene encoding a polypeptide having a certain sequence identity (eg, 80% or more, 85% or more, or 90% or more sequence identity) with the amino sequence as described above.
  • SEQ ID NO: 7 Human IL1F9; NCBI Reference Sequence: NM_019618.4 (Coding sequence, CDS)
  • SEQ ID NO: 8 Musculus IL1F9; NCBI Reference Sequence: NM_153511.3 (
  • -Genes with the above base sequence -Inhibiting at least one of translation and transcription of a gene having a base sequence having a certain sequence identity (for example, 80% or more, 85% or more, or 90% or more sequence identity) with the above-mentioned base sequence.
  • Nucleic acid molecules that can be produced are included.
  • IL-36 ⁇ IL-36G
  • IL36 ⁇ IL36G
  • IL1F9 IL-1F9
  • the "biological activity" of the target molecule IL-36 receptor is, for example, signal transduction mediated by an IL-36 ligand (particularly IL-36 ⁇ ). ..
  • the "IL-36 receptor antagonist” inhibits the binding of IL-36 receptor ligands ( ⁇ , ⁇ and ⁇ ) to the IL-36 receptor.
  • IL-36 receptor antagonist is based, for example, on its ability to act as a competitive inhibitor of IL-36 ⁇ that binds to the IL-36 receptor, and to inhibit or block the binding of IL-36 ⁇ to the IL-36 receptor. Can be identified. Also, for example, binding affinity for a particular IL-36 receptor epitope, inhibition of binding of the IL-36 receptor to its ligand, neutralization or inhibition of IL-36 receptor activity in vivo (eg, IC 50 ), and It can be identified on the basis of cross-reactivity (eg, cross-reactivity of the IL-36 receptor protein with homologs or orthologs, or with other proteins or tissues).
  • the IL-36 receptor antagonist of the invention exhibits one or more of the following biological activities: the IL-36 receptor and at least one of IL-36 ⁇ , IL-36 ⁇ and IL-36 ⁇ . Inhibition of interaction between the two, inhibition of intracellular signals mediated by the IL-36 receptor, and inhibition of cross-reactivity and activity of human and non-human primates with the IL-36 receptor.
  • receptor antagonists recognized in the art include, for example, avidity, selectivity, solubility, folding, immunotoxicity and expression. These properties or features can be observed, measured and / or evaluated using techniques known in the art.
  • the known techniques include, for example, ELISA, competitive ELISA, surface plasmon resonance assay (BIACORET M), KINEXAT M, in vitro or in vivo neutralization assay, receptor-ligand binding assay, cytokine or growth factor production and /.
  • ELISA ELISA
  • competitive ELISA surface plasmon resonance assay
  • KINEXAT M KINEXAT M
  • in vitro or in vivo neutralization assay in vitro or in vivo neutralization assay
  • receptor-ligand binding assay cytokine or growth factor production and /.
  • it includes, but is not limited to, secretory assays, as well as signaling and immunohistochemical assays.
  • the IL-36 receptor antagonist for example, incubates cells expressing the IL-36 receptor in the presence and absence of the test compound and intracellularly activates the IL-36 receptor signaling pathway, eg, NF- ⁇ B signal.
  • IL-36 receptor signaling pathway eg, NF- ⁇ B signal.
  • Expression of mRNAs of related genes can be identified, for example, by monitoring using real-time RT-PCR. If the mRNA expression of the NF- ⁇ B signal-related gene is lower in the presence of the test compound than in the absence of the test compound, the test compound is an IL-36 receptor antagonist.
  • IL-36 receptor antagonists include neutralizing antibodies against the entire native IL-36 receptor polypeptide or the native IL-36 receptor polypeptide subunit, the IL-36 receptor subunit fused with an immunoglobulin constant region sequence.
  • Immunoadhesion factors, small molecules, aptamers that bind to the entire native IL-36 receptor polypeptide or the native IL-36 receptor polypeptide subunit, translation of genes encoding the entire or subunit of the native IL-36 receptor polypeptide and Nucleic acid molecules capable of inhibiting at least one of transcriptions (eg, siRNA, antisense subunits, decoys, ribozymes) and the like are included, but are not limited to these.
  • the sequence of the native IL-36 receptor polypeptide can be obtained from a database known in the art.
  • Examples of the sequence of the natural IL-36 receptor polypeptide include SEQ ID NO: 9 (Human IL1RL2; NCBI Reference Sequence: NP_003845.2) and SEQ ID NO: 10 (Musmusculus IL1RL2; NCBIRefenceSequen) Can be mentioned. Therefore, the IL-36 receptor antagonist includes an antibody and a fragment thereof that specifically recognize a polypeptide having such a sequence, and a certain sequence identity (for example, 80% or more, 85% or more, or 90) with the sequence. % Or more sequence identity) includes antibodies that specifically recognize polypeptides and fragments thereof.
  • IL-36 receptor polypeptide for example, SEQ ID NO: 11 (Human IL1RL2; NCBI Reference Sequence: NM_003854.4 (Coding sequence, CDS)), SEQ ID NO: 12 (CDS) shown below. Mus musculus IL1RL2; NCBI Reference Sequence: NM_133193.4 (Coding sequence, CDS)). Therefore, for IL-36 ⁇ antagonists, -A gene encoding a polypeptide having the above amino sequence, A gene encoding a polypeptide having a certain sequence identity (eg, 80% or more, 85% or more, or 90% or more sequence identity) with the amino sequence as described above.
  • sequence identity eg. 80% or more, 85% or more, or 90% or more sequence identity
  • -Genes with the above base sequence -Inhibiting at least one of translation and transcription of a gene having a base sequence having a certain sequence identity (for example, 80% or more, 85% or more, or 90% or more sequence identity) with the above-mentioned base sequence.
  • a certain sequence identity for example, 80% or more, 85% or more, or 90% or more sequence identity
  • IL-1Rrp2 In the present specification, the terms “IL-1Rrp2”, “1L-1RL2” and “1L1RL2” are synonymous with respect to “IL-36 receptor” and are used interchangeably.
  • the "IL-36 receptor antagonist” may include those known in the art, and are described in, for example, Japanese Patent No. 6289375, Japanese Patent Application Laid-Open No. 2018-093885, Japanese Patent Application Laid-Open No. 2018-512157, and the like. Antibodies and fragments thereof, molecules described in JP-A-2017-114829, commercial products and investigational agents may be included. Examples of the investigational drug include BI655130 (Boehringer Ingelheim), which is an anti-IL-36 receptor monoclonal antibody.
  • the IL-36 receptor antagonist of the present invention is an anti-IL-36 receptor antibody.
  • the anti-IL-36 receptor antibody has high molecular / cell binding capacity.
  • IL-1F5 is known as an IL-36 receptor antagonist.
  • antibody is used in the broadest sense, in particular monoclonal antibodies (eg, neutralizing and working antibodies), polyclonal antibodies, multispecific antibodies (eg, bispecific antibodies) and antibody fragments. Is included. Monoclonal antibodies include chimeric antibodies and fragments of such antibodies as long as they exhibit the desired biological activity.
  • a chimeric antibody is one in which a portion of a heavy chain, a light chain, or both is identical or homologous to or homologous to an antibody of a particular species, or an antibody belonging to a particular antibody class or subclass, and the remaining strand.
  • Monoclonal antibodies further include "humanized" antibodies or fragments containing the smallest sequences from non-human immunoglobulins, which include, for example, Fv, Fab, Fab', F (ab') 2 , Other examples include, but are not limited to, antigen-binding partial sequences of antibodies.
  • a humanized antibody is a human immunity in which a residue derived from the recipient's CDR is replaced with a residue derived from the CDR of a non-human species (donor antibody) having the desired specificity, affinity, and ability. It may be globulin (receptor antibody). Examples of non-human species include mice or rats, rabbits and the like. In one example, the Fv, FR residues of human immunoglobulin have been replaced with the corresponding non-human residues.
  • the humanized antibody may contain residues not found in the CDR or framework sequence introduced into the recipient antibody. These modifications are made for the purpose of improving the performance of the antibody.
  • humanized antibodies contain substantially any one or more, typically two variable domains, where all or substantially all CDR regions correspond to the CDR regions of non-human immunoglobulins. And all or substantially all FR regions are FR regions of human immunoglobulin sequences.
  • Optimal humanized antibodies also include at least a portion of an immunoglobulin constant region (Fc), typically including a constant region of human immunoglobulin.
  • Fc immunoglobulin constant region
  • Humanized antibodies may include primated antibodies.
  • references to literature known in the art for example, Jones et al. , Nature, 321: 522-525 (1986) and Reichmann et al. , Nature, 332: 323-329 (1998) and the like.
  • antibody fragment includes a portion of a full-length antibody, generally its antigen-binding or variable region.
  • antibody fragments are Fab, Fab', F (ab') 2 and Fv fragments; single chain antibody molecules; diabodies; linear antibodies; multispecificity formed from single chain antibody molecules and antibody fragments. Contains antibodies.
  • the term "monoclonal antibody” refers to a substantially homogeneous antibody, i.e. an antibody obtained from a population in which each antibody, including the population, is equivalent except for naturally occurring mutations in which a small amount may be present. Point to. Monoclonal antibodies are highly specific and point to a single antigenic site. Moreover, each monoclonal antibody is directed to a single determinant on the antigen, as opposed to conventional (polyclonal) antibody preparations that typically contain different antibodies directed to different determinants (epitopes). There is.
  • antisense oligonucleotide is defined as a nucleic acid molecule capable of inhibiting transcription and / or translation of a target gene in a sequence-specific manner.
  • antisense refers to the nucleic acid being complementary to the coding ("sense") gene sequence of the target gene.
  • the antisense oligonucleotide hybridizes to the nascent mRNA in the antiparallel direction by Watson-Crick base binding. By binding to the target mRNA template, the antisense oligonucleotide inhibits the achievement of translation of the encoded protein.
  • ribozyme an antisense agent which is designed to induce catalytic cleavage of the target RNA by adding a sequence with spontaneous self-splicing activity (eg, W). See arzocha et al., Leuk. Lymphoma 24: 267-281 [1997]).
  • Another embodiment of the present invention relates to a composition that inhibits the production of IL-36 ⁇ , which comprises a CD38 antagonist.
  • a composition containing a CD38 antagonist can inhibit the production of IL-36 ⁇ .
  • Yet another embodiment of the invention relates to a composition that inhibits the activity of the IL-36 receptor, comprising at least one of a CD38 antagonist, an IL-36 ⁇ antagonist and an IL-36 receptor antagonist.
  • a composition comprising one or more of these can inhibit the activity of the IL-36 receptor.
  • Another embodiment of the present invention relates to a composition for suppressing muscle tissue loss, which comprises a compound that inhibits the activity of the IL-36 receptor.
  • a composition for suppressing muscle tissue loss which comprises a compound that inhibits the activity of the IL-36 receptor.
  • the present inventors have found that muscle atrophy is induced through intracellular signal transduction by activation of the IL-36 receptor (Fig. 7B). Therefore, by inhibiting the activity of the IL-36 receptor, intracellular signal transduction can be inhibited and muscle atrophy (that is, reduction of muscle tissue) can be suppressed.
  • the subject has “monocytes with enhanced expression of CD38” means that the subject has cancer and / or has cancer-related symptoms.
  • the “cancer-related symptom” includes cancer cachexia, weight loss, loss of appetite and muscle weakness, and the cancer-related symptom is preferably cancer cachexia.
  • the second aspect of the present invention is a pharmaceutical composition for treating cancer, and in particular, a pharmaceutical composition for treating cancer-related symptoms.
  • “Cancer” includes both blood cancer and solid cancer.
  • “Blood cancer” includes, but is not limited to, malignant lymphoma, multiple myeloma, leukemia (including chronic and acute), and myeloproliferative neoplasms.
  • Examples of “solid cancer” include breast cancer, colon cancer, malignant melanoma, bone tumor, soft tumor, brain tumor, tongue cancer, pharyngeal cancer, esophageal cancer, thyroid cancer, lung cancer, gastric cancer, and ovary.
  • Cancer, cervical cancer, uterine body cancer, renal cell cancer, prostate cancer, bladder cancer, skin cancer, liver cancer and pancreatic cancer include, but are not limited to.
  • the "blood cancer” is chronic myelomonocytic leukemia.
  • the "solid cancer” is breast cancer.
  • the subject may have monocytes expressing CD38 and have a decrease in muscle tissue, or a decrease in muscle tissue may be expected.
  • the subject has monocytes with enhanced expression of CD38 means that the subject suffers from an inflammatory disease.
  • inflammatory disease refers to a pathological condition that causes inflammation, which is typically caused by the chemotaxis of neutrophils.
  • Inflammatory disorders include, but are not limited to: inflammatory skin disorders (eg, psoriasis and dermatitis), generalized scleroderma, systemic sclerosis, inflammatory enteritis (eg, psoriasis and dermatitis).
  • IBD IBD
  • Crohn's disease inflammatory colitis, injury and myocardial infarction due to surgical tissue reperfusion, myocardial ischemia such as myocardial infarction, cardiac arrest, reperfusion after heart surgery, stenosis after percutaneous transluminal coronary angioplasty, deficiency Bloody reperfusion disease (eg, stroke and abdominal aneurysm), secondary stroke cerebral edema, cranial trauma, blood loss shock, asphyxia, adult respiratory distress syndrome acute lung injury, Bechet's disease, scleroderma, polymyositis, multiple Scleroderma (MS), dermatitis, meningitis, encephalitis, vegetation, osteoarthritis, lupus nephritis, joint inflammatory conditions (eg, rheumatoid arthritis (RA), rheumatic spondylitis, gouty arthritis), Antigen-antibody complex including Sjogren's syndrome, vasculitis, diseases with extravasation
  • the subject has “monocytes with enhanced expression of CD38” means that the subject suffers from an autoimmune disease.
  • autoimmune diseases include systemic lupus erythematosus, multiple sclerosis, rheumatoid arthritis, psoriasis, type I diabetes, hyperthyroidism, autoimmune adrenal dysfunction, autoimmune hemolytic anemia, and multiple sclerosis. , Psoriasis arthritis, Sjogren's syndrome, multiple sclerosis, dermatomyositis, severe myasthenia, idiopathic thrombocytopenic purpura, autoimmune optic neuropathy, scleroderma, etc., but not limited to these.
  • the "autoimmune disease” is preferably at least one selected from the group consisting of systemic lupus erythematosus, multiple sclerosis, rheumatoid arthritis and psoriasis.
  • the subject "has monocytes with enhanced expression of CD38” means that the subject suffers from a chronic infection.
  • chronic infectious disease include, but are not limited to, HIV infection, tuberculosis, malaria, leprosy, and syphilis.
  • a "chronic infection” is preferably at least one selected from the group consisting of HIV infection, tuberculosis and malaria.
  • treatment refers to both therapeutic treatments and preventative measures, the purpose of which is to prevent or delay (alleviate) unwanted physiological changes or disorders.
  • beneficial or desirable clinical outcomes include alleviation of symptoms and reduction of the scope of the disease, stabilization of the disease state (ie, not exacerbating), progression of the disease. Includes, but is not limited to, delay or deceleration, improvement or temporary relief of the disease state, and remission (including, but not limited to, partial and total relief).
  • Treatment can also mean an extension of survival compared to the expected survival without treatment. Subjects in need of treatment include those who already have the condition or disorder, as well as those who are vulnerable to the condition or disorder or whose condition or disorder should be prevented.
  • compositions according to the invention typically include pharmaceutically acceptable additives such as excipients, binders, lubricants, solvents, diluents, stabilizers known in the art. , Isotonicity agent and the like may be included.
  • Excipients include starch, lactose, methyl cellulose, crystalline cellulose, synthetic aluminum silicate, etc.
  • binders include, for example, hydroxypropyl cellulose and polyvinylpyrrolidone
  • lubricants include, for example, talc, magnesium stearate, calcium stearate.
  • sodium chloride, glucose, glycerol, mannitol and the like are used.
  • the pharmaceutical composition according to the present invention is formulated into, for example, tablets, powders, granules, capsules, injections, liquids, suppositories, etc., and is administered orally, intravenously, subcutaneously, intraperitoneally, intramuscularly, for example. can do.
  • the dose of the pharmaceutical composition according to the present invention varies depending on the age, body weight, medical condition, etc. of the subject.
  • composition according to the present invention may be used as a combination agent in combination with a further pharmaceutical composition.
  • each pharmaceutical composition ie, the first active ingredient and the second active ingredient
  • the ingredients in each pharmaceutical composition are, together, sequentially or individually, one combination unit dosage form or two individual units. It may be administered in a dosage form.
  • a therapeutically effective amount of each active ingredient of the combination according to the present invention may be administered simultaneously, individually or in any order, sequentially or sequentially.
  • the first active ingredient and the second active ingredient may be present in a plurality of units.
  • Combinations also include, for example, kits containing their respective active ingredients and instructions for use.
  • Plat-E packaging cells were added to Dulvecco's Modified Eagle's Medium (DMEM) (Fujifilm Wako Pure Chemical Industries), 10% fetal bovine serum (FBS), 1% penicillin, 1% penicillin. ) (Nacalai Tesque), 1 ng / ml Puromycin, 10 ng / ml Blastside was added to the culture medium, and the cells were cultured at 37 ° C. in the presence of 5% CO 2. A 0.25 w / v% trypsin 1 mmol / L EDTA ⁇ 4Na solution (containing phenol red) (Fujifilm Wako Pure Chemical Industries, Ltd.) was used for cell detachment at the time of passage.
  • DMEM Dulvecco's Modified Eagle's Medium
  • FBS fetal bovine serum
  • penicillin 1% penicillin.
  • penicillin. 1% penicillin.
  • 1 ng / ml Blastside was added to the culture medium,
  • C2C12 cells were cultured in DMEM medium containing 10% FBS and 1% P / S, and when differentiated into muscle fibers, they were replaced with DMEM medium containing 2% horse serum (Thermo Fisher Scientific) and 1% P / S. .. 4T1 cells and THP1 cells were cultured in RPMI1640 medium containing 10% FBS and 1% P / S.
  • DMEM medium containing 10% FBS and 1% P / S
  • Plat-E cells were donated by Dr. Toshio Kitamura, Institute of Medical Science, University of Tokyo.
  • C2C12 cells were purchased from RIKEN BRC and 4T1 cells from ATCC.
  • the PCR product was purified and subcloned into the pCRBluntIITOPO vector using ZeroBluntTOPOPCR CloningKit (Thermo Fisher Scientific). Using this as a template, the ORF sequence of Il1f9 was amplified by the PCR method, and a restriction enzyme (XhoI, NotI) recognition sequence was added. The primers used are shown below.
  • the PCR product was treated with a restriction enzyme and inserted into the Multi-cloning-site of the pMYs-IRES-GFP retroviral vector using Ligationhigh ver.2 (Toyobo).
  • the pMYs-NUP98-HBO1-IRES-GFP vector is described in Hayashi Y, et al. Blood Adv. 2019; 3 (7): 1047-1060.
  • a retroviral vector was introduced into Plat-E cells having a cell density of 70 to 80% by subculturing to a 10 cm dish the day before using FuGENE-HD (Promega) (transfection).
  • OPTI-MEM Thermo Fisher Scientific
  • the medium was changed 24 hours after transfection, and Plat-E culture supernatant containing virus particles was collected 48 hours after transfection.
  • the supernatant was passed through a PVDF filter (Merck) having a pore size of 0.45 ⁇ m, mixed with 4xPEG buffer (3: 1 ratio), mixed by inversion, and allowed to stand at 4 ° C. overnight. Then, it was centrifuged at 1800 g at 4 degrees for 45 minutes and resuspended in 2 ml of medium to prepare a concentrated virus solution.
  • mice [Mouse hematopoietic stem cell / progenitor cell collection and culture] Wild C57BL / 6 mice (7 weeks old, female) were intraperitoneally administered with 150 mg / kg of 5 fluorouracil (5FU) (Kyowa Hakko Kirin), and 96 hours later, sufficient inhalation with isoflurane (Fujifilm Wako Pure Chemical Industries, Ltd.). The mice were euthanized under anesthesia. All subsequent operations were performed on ice.
  • 5FU fluorouracil
  • isoflurane Flujifilm Wako Pure Chemical Industries, Ltd.
  • 2% FBS / Phosphate Buffered Salts the femur and tibia were collected and placed in a 50 ml centrifuge tube using a 21 G needle (Terumo) and a 1 ml syringe (Terumo). Flashed with. After centrifuging the bone marrow cell suspension (400 g, 4 degrees, 5 minutes), the supernatant was aspirated and resuspended in Pharma Lyse (BD Biosciences) to hemolyze and remove mature erythrocytes. After centrifugation (400 g, 4 degrees, 5 minutes), the supernatant was aspirated and resuspended in 2% FBS / PBS for washing.
  • PBS Phosphate Buffered Salts
  • Iscover's Modified Dulvecco's Medium (IMDM) (Fujifilm Wako Pure Drug) 20% FBS (Eagle's Bio), 1% penicillin streptomycin, 50 ng / ml mouse (SCF), 50 ng / ml bone marrow interleukin 6 (IL-6), 50 ng / ml bone marrow Thrombopoietin (TPO), 50 ng / ml bone marrow FMS-like medium kinase 3 (FLT3) I used the one.
  • IMDM Modified Dulvecco's Medium
  • FBS Fegle's Bio
  • IL-6 50 ng / ml bone marrow interleukin 6
  • TPO ng / ml bone marrow Thrombopoietin
  • FLT3 FMS-like medium kinase 3
  • mice bone marrow transplant The transgenic mouse bone marrow cells were collected, centrifuged (400 g, 4 ° C., 5 minutes), and the supernatant was aspirated and resuspended in PBS (200 ⁇ L PBS per recipient mouse). The cell suspension for transplantation was allowed to stand on ice until just before transplantation. Wild-type C57BL / 6 mice (7 weeks old, female) were irradiated with a sublethal dose (6-7 Gray) of radiation, and a cell suspension for transplantation was injected through the tail vein (bone marrow transplantation). A 27G insulin syringe (Terumo) was used for injection.
  • Peripheral blood smear A few ⁇ L of peripheral blood was placed on a slide glass to prepare a peripheral blood smear. After sufficiently drying with cold air, Diff-Quik staining (simple Bright-Giemsa staining) (Sysmex) was performed, and the cell morphology was observed with an upright microscope Nikon Eclipse Ni (Nikon).
  • Il1f9 (Il36g) levels in mouse plasma were measured using Mouse Interleukin-36 gumma ELISA Kit (Mybiosource).
  • mice were euthanized by cervical dislocation under sufficient inhalation anesthesia with isoflurane (Fujifilm Wako Pure Chemical Industries, Ltd.). All subsequent operations were performed on ice.
  • the femur and tibia were harvested and flushed in a 2% FBS / PBS in a 15 ml centrifuge tube or 5 ml FACS tube using a 21 G needle (Terumo) and a 1 ml syringe (Terumo).
  • the supernatant was aspirated and resuspended in 500 ⁇ L of Pharma Lyse (BD Biosciences) to hemolyze and remove mature erythrocytes. After centrifuging (400 g, 4 ° C., 5 minutes) with a total liquid volume of 5 ml with 2% FBS / PBS, the supernatant was aspirated and resuspended with an appropriate amount of 2% FBS / PBS, and used in the experiment.
  • Pharma Lyse BD Biosciences
  • C2C12 cells were seeded on a 6-well plate and differentiated into muscle fibers.
  • Cell culture inserts ThinCert (0.4 ⁇ m), Greiner) Insert the mouse whole bone marrow cells (2 ⁇ 10 6 cells) or mouse inflammatory monocytes (2-4 ⁇ 10 5 cells) of the Noto coculture went.
  • Cell surface antigens include anti-mouse Gr-1 (RB6-8C5), anti-mouse CD115 (AFS98), anti-mouse c-Kit (2B8), anti-mouse Ly6C (HK1.4), anti-mouse B220 (RA3-6B2), Staining with anti-mouse CD38 (90) antibody (Biolegend) on ice and in the dark for 30 minutes. After staining, the cells were washed with a sufficient amount of FACS buffer (centrifugal: 400 g, 4 ° C., 3 minutes) and then resuspended in 300 ⁇ L FACS buffer. The cell suspension was transferred to a FACS tube through a 40-100 ⁇ m filter immediately prior to analysis.
  • FACS buffer centrifugal: 400 g, 4 ° C., 3 minutes
  • Dead cells were labeled with 7-Aminoactinomycin D (7AAD) (Biolegend).
  • FACS Canto (Beckton Dickinson) was used for flow cytometry analysis, and FlowJo (Tree Star) was used for data analysis.
  • RNA-Seq Ly6C high CD115 + inflammatory monocytes in mouse bone marrow were collected using a cell sorter (SH800, Sony), and total RNA was extracted using RNeasy Mini Kit (Qiagen). RNA samples were submitted to GeneNex for RNA-Seqing (Novaseq, paired-end, 150bp, 6G / sample). Principal component analysis (PCA) and data analysis were performed using AltAnalyze software (v.2.1.0-Win64).
  • GSEA Gene Set Enrichment Analysis
  • a novel fusion gene NUP98-HBO1 (Fig. 1) identified from CMML patient cells of a CMML model mouse using a novel NUP98-HBO1 fusion gene was cloned, and a retroviral vector (pMYs-NUP98-HBO1-IRES-GFP) was used. Created. Mouse bone marrow hematopoietic stem cells / progenitor cells were infected with retrovirus, NUP98-HBO1 was introduced into the gene, and the cells were bone marrow transplanted (Bone Marrow Transplant: BMT) into wild-type mice.
  • BMT bone marrow Transplant
  • mice after BMT showed marked leukocyte (WBC) increase, macrocytic anemia (high MCV, Hb decrease), platelet (PLTS) decrease, and characteristic inflammatory monocyte increase (FIGS. 2A and 2B). ). Precursor cell increase in bone marrow was less than 20%, and megakaryocyte dysplasia was prominent (FIGS. 2C and 2D).
  • the gene group (NUP98-HBO1 inducible gene) whose expression was upregulated when NUP98-HBO1 was introduced into human cord blood hematopoietic stem cells / progenitor cells was significantly upregulated in the CMML patient group (Fig. 2E).
  • FIG. 4B show that the width of co-cultured C2C12 muscle fibrous cells is reduced in NUP98-HBO1 mice compared to control mice, which is characteristic inflammation in NUP98-HBO1 mice.
  • sex monocytes are involved in the induction of muscle tissue atrophy.
  • IL1F9 (IL36G) is a gene whose expression is upregulated only in CMML mice that have developed cachexia among the genes encoding humoral factors. ).
  • NF ⁇ B signaling involving IL1F9 (IL-36 ⁇ ) and its receptors (IL-1Rrp2 and IL-1RAcP) is shown in FIG. 7B.
  • IL1F9 may be involved in the development of cachexia in subjects with inflammatory monocytes that highly expressed CD38. I stood up.
  • IL36G level in peripheral blood (plasma) of bad fluid-onset CMML mice When the IL36G level in peripheral blood (plasma) of bad fluid-onset CMML mice was measured by the ELISA method, bad fluid was measured. It was confirmed that the level of IL36G in the peripheral blood (plasma) of pledge-onset CMML mice was elevated as compared with the control mice (Fig. 8).
  • CD38-positive monocytes were also induced in breast cancer, which is a solid cancer. From these results, the induction of cachexia-related CD38-positive monocytes is not limited to CMML, which is a blood cancer, and may appear in solid tumor cachexia model mice in accordance with the progression of the pathological condition. confirmed.
  • composition according to the present invention can be used to suppress the loss of muscle tissue in a subject having monocytes expressing CD38.
  • existing drugs containing as an active ingredient a compound involved in a pathway that causes activation of the IL-36 receptor and a compound that inhibits the biological activity of any one or more of cells suppress the loss of muscle tissue. It is also suggested that it can be used for.

Abstract

Le problème à résoudre par la présente invention est de fournir une composition pour supprimer la perte de tissu musculaire. La solution selon l'invention porte sur une composition destinée à supprimer la perte de tissu musculaire chez un sujet ayant des monocytes avec une expression accrue de CD38 et contient un composé pour inhiber l'activité du récepteur IL-36.
PCT/JP2020/045116 2019-12-05 2020-12-03 Composition pour supprimer la perte de tissu musculaire WO2021112196A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2021562730A JPWO2021112196A1 (fr) 2019-12-05 2020-12-03

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2019220024 2019-12-05
JP2019-220024 2019-12-05

Publications (1)

Publication Number Publication Date
WO2021112196A1 true WO2021112196A1 (fr) 2021-06-10

Family

ID=76222421

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2020/045116 WO2021112196A1 (fr) 2019-12-05 2020-12-03 Composition pour supprimer la perte de tissu musculaire

Country Status (2)

Country Link
JP (1) JPWO2021112196A1 (fr)
WO (1) WO2021112196A1 (fr)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100080784A1 (en) * 2008-09-12 2010-04-01 Torrey Pines Institute For Molecular Studies Methods for treating cachexia and lymphopenia
JP2013079238A (ja) * 2005-10-12 2013-05-02 Morphosys Ag ヒトCD38に特異的な完全ヒトHuCALGOLD由来の治療抗体の生成とプロファイリング
JP2015500633A (ja) * 2011-11-16 2015-01-08 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 抗il−36r抗体
JP2016537356A (ja) * 2013-11-20 2016-12-01 オハイオ・ステイト・イノベーション・ファウンデーション がん関連悪液質を抑制するためのhdac阻害剤
JP2018512157A (ja) * 2015-04-15 2018-05-17 アナプティスバイオ インコーポレイティッド インターロイキン36受容体(il−36r)に対する抗体
WO2020037154A1 (fr) * 2018-08-17 2020-02-20 23Andme, Inc. Anticorps anti-il1rap et leurs méthodes d'utilisation

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013079238A (ja) * 2005-10-12 2013-05-02 Morphosys Ag ヒトCD38に特異的な完全ヒトHuCALGOLD由来の治療抗体の生成とプロファイリング
US20100080784A1 (en) * 2008-09-12 2010-04-01 Torrey Pines Institute For Molecular Studies Methods for treating cachexia and lymphopenia
JP2015500633A (ja) * 2011-11-16 2015-01-08 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 抗il−36r抗体
JP2016537356A (ja) * 2013-11-20 2016-12-01 オハイオ・ステイト・イノベーション・ファウンデーション がん関連悪液質を抑制するためのhdac阻害剤
JP2018512157A (ja) * 2015-04-15 2018-05-17 アナプティスバイオ インコーポレイティッド インターロイキン36受容体(il−36r)に対する抗体
WO2020037154A1 (fr) * 2018-08-17 2020-02-20 23Andme, Inc. Anticorps anti-il1rap et leurs méthodes d'utilisation

Also Published As

Publication number Publication date
JPWO2021112196A1 (fr) 2021-06-10

Similar Documents

Publication Publication Date Title
JP6960507B2 (ja) Cd47によって媒介される食作用を操作するための方法
US20150174203A1 (en) Compositions And Methods For Modulating Pro-Inflammatory Immune Response
US9381210B2 (en) Induced pluripotent stem cell model of chronic myeloid leukemia revealed olfactomedin 4 as a novel therapeutic target in leukemia stem cells
KR101886890B1 (ko) 항인간 ccr7 항체, 하이브리도마, 핵산, 벡터, 세포, 의약 조성물 및 항체 고정화 담체
US20200017571A9 (en) Tnfrsf14 / hvem proteins and methods of use thereof
JP2016523930A (ja) 骨髄異形成症候群(mds)を治療するための可溶性cd33
JP2021505172A (ja) 造血幹細胞及び前駆細胞を動員させるための投薬レジメン
CN116948011A (zh) Mage-a1特异性t细胞受体及其用途
CN111432838A (zh) 使用双特异性抗体和il-15进行联合治疗
AU2014201010B2 (en) Methods for manipulating phagocytosis mediated by CD47
JP2004504816A (ja) 幹細胞を同定および/または単離し、白血病治療に対する応答性を予後判定する方法
WO2021112196A1 (fr) Composition pour supprimer la perte de tissu musculaire
JP4950885B2 (ja) 白血病における酵素阻害剤
JP6770522B2 (ja) 全身性エリテマトーデスを予防し、及び/又は、治療するためのptgdr−1及び/又はptgdr−2アンタゴニスト
CN116970066B (zh) Mage-a1特异性t细胞受体及其用途
TW202321287A (zh) 特異性靶向間皮素之經工程改造之免疫細胞及其用途
CN117624338A (zh) Mage-a1特异性t细胞受体及其用途
CN117624339A (zh) Mage-a1特异性t细胞受体及其用途
WO2023240291A2 (fr) Compositions et agents thérapeutiques pour moduler des réponses inflammatoires des cellules tumorales et immunitaires
CN117659165A (zh) Mage-a1特异性t细胞受体及其用途
CN117545489A (zh) 修饰的干细胞组合物及其使用方法
CN116496411A (zh) Afp特异性t细胞受体及其用途
AU2019253783A1 (en) Methods for manipulating phagocytosis mediated by CD47
CN117924461A (zh) Mage-a1特异性t细胞受体及其用途
CN117946248A (zh) Mage-a1特异性t细胞受体及其用途

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: 20895500

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021562730

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20895500

Country of ref document: EP

Kind code of ref document: A1