[go: up one dir, main page]

WO2021225135A1 - Therapeutic agent for nakajo-nishimura syndrome - Google Patents

Therapeutic agent for nakajo-nishimura syndrome Download PDF

Info

Publication number
WO2021225135A1
WO2021225135A1 PCT/JP2021/017367 JP2021017367W WO2021225135A1 WO 2021225135 A1 WO2021225135 A1 WO 2021225135A1 JP 2021017367 W JP2021017367 W JP 2021017367W WO 2021225135 A1 WO2021225135 A1 WO 2021225135A1
Authority
WO
WIPO (PCT)
Prior art keywords
mcp
mls
production
nns
cudc
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/JP2021/017367
Other languages
French (fr)
Japanese (ja)
Inventor
潤 齋藤
直也 加瀬
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kyoto University NUC
Original Assignee
Kyoto University NUC
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 Kyoto University NUC filed Critical Kyoto University NUC
Priority to JP2022519962A priority Critical patent/JP7709752B2/en
Publication of WO2021225135A1 publication Critical patent/WO2021225135A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/4045Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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

Definitions

  • the present invention relates to a therapeutic agent for Nakajo-Nishimura syndrome.
  • This application claims priority based on US Patent Application No. 63 / 021,112, which was provisionally filed in the United States on May 7, 2020, the contents of which are incorporated herein by reference.
  • Nakajo-Nishimura syndrome is one of the proteasome-related autoinflammatory syndromes (PRAAS) and is caused by homozygous mutations in the proteasome subunit beta 8 (PSMB8) / LMP-7 gene. NNS begins with a chilblain-like rash in early infancy, develops regular high fever, and ultimately results in fat / muscle atrophy and joint contracture, primarily in the upper body.
  • PRAAS proteasome-related autoinflammatory syndromes
  • PSMB8 proteasome subunit beta 8
  • All NNS patients are homozygous for the PSMB8 gene. It has a 602G> T mutation (a guanine to thymine mutation at the 602nd nucleotide residue), which is a G201V amino acid mutation at the ⁇ 5i subunit (a glycine to valine mutation at the 201st amino acid residue). Mutation).
  • NNS patients are thought to cause immune proteasome dysfunction, which induces cell stress and thus inflammation.
  • MCP-1 monocyte chemotaxis protein-1
  • IP-10 interferon gamma-induced protein 10
  • Oral steroids are used as a treatment method for NNS, but they are not effective for fat atrophy and joint contracture, and have serious side effects especially for infants, so the development of new therapeutic agents is eagerly desired.
  • the inventors have previously prepared NNS disease model cells using human induced pluripotent stem cells (iPSCs) and human embryonic stem cells (ESCs) (see Non-Patent Document 1). Specifically, wild-type iPSCs (WT-iPS) were prepared by recovering mutations from mutant iPSCs (MT-iPS) established from NNS patients using the genome editing technology CRISPR / Cas9 system. In addition, a mutant type (MT-ES) in which mutations were introduced into the human embryonic stem cell line KhES-1 (WT-ES) was prepared in the same manner.
  • WT-iPS wild-type iPSCs
  • MT-iPS mutant iPSCs
  • KhES-1 human embryonic stem cell line
  • MLs monocyte phenotype monocytic cell lines
  • WT-iPS-MLs monocyte phenotype monocytic cell lines
  • MT-iPS-MLs monocyte phenotype monocytic cell lines
  • WT-ES-MLs WT-ES-MLs
  • MT-ES-MLs monocyte phenotype monocytic cell lines
  • Non-Patent Document 1 also describes antioxidants and Janus Kinase (JAK) inhibitors as candidates for therapeutic agents for NNS.
  • JK Janus Kinase
  • an object of the present invention is to provide a therapeutic agent for NNS.
  • a therapeutic agent for Nakajo-Nishimura syndrome which contains a histone deacetylase inhibitor.
  • the histone deacetylase inhibitor is at least one compound selected from the group consisting of CUDC-907, JNJ-26481585, LAQ824, romidepsin and tricostatin A, or a pharmacologically acceptable salt thereof or
  • the therapeutic agent for Nakajo-Nishimura syndrome according to [1] which is a solvate thereof.
  • FIG. 1 is a schematic diagram showing an experimental schedule of Experimental Example 1.
  • FIG. 2 is a graph showing the production inhibition rates of the compounds MCP-1 and IP-10 in the primary screening of Experimental Example 1.
  • FIG. 3 is a graph showing the production inhibition rates of the compounds MCP-1 and IP-10 in the secondary screening of Experimental Example 1.
  • FIG. 4 is a graph showing the production inhibition rates of the compounds MCP-1 and IP-10 in MT-ES-MLs of Experimental Example 1.
  • FIG. 5 is a graph showing the results of cytotoxicity evaluation in Experimental Example 1.
  • FIG. 6 is a schematic diagram showing the results of high-throughput screening in Experimental Example 1.
  • 7 (a) to 7 (c) are dose-response curves for the production of MCP-1 of each compound measured in Experimental Example 2.
  • FIG. 7 (d) to 7 (f) are dose-response curves for the production of IP-10 of each compound measured in Experimental Example 2.
  • FIG. 8A is a graph showing the results of measuring intracellular reduced nicotinamide adenine dinucleotide (NADH) in Experimental Example 2.
  • FIG. 8B is a graph showing the results of measuring extracellular lactate dehydrogenase (LDH) activity in Experimental Example 2.
  • FIG. 9 is a graph showing the expression level of MCP-1 by each cell measured in Experimental Example 3.
  • FIG. 10 is a graph showing the dose-dependent effect of inhibiting the production of MCP-1 of CUDC-907 in fibroblasts derived from NNS patients, which was measured in Experimental Example 3.
  • FIG. 11A is a graph showing the results of measuring intracellular NADH in Experimental Example 3.
  • FIG. 11B is a graph showing the results of measuring the extracellular LDH activity in Experimental Example 3.
  • FIG. 12A is a graph showing the results of comparing the production amount of MCP-1 in each cell in Experimental Example 4.
  • FIG. 12B is a graph showing the results of comparing the amount of IP-10 produced in each cell in Experimental Example 4.
  • FIG. 13 is a graph showing the amount of MCP-1 produced in a fibroblast cell line derived from a healthy subject and a fibroblast cell derived from an NNS patient, which was measured in Experimental Example 4.
  • FIG. 14A is a graph showing the results of measuring the expression level of the CCL2 gene in Experimental Example 5.
  • FIG. 14A is a graph showing the results of measuring the expression level of the CCL2 gene in Experimental Example 5.
  • FIG. 14B is a graph showing the results of measuring the expression level of the CXCL10 gene in Experimental Example 5.
  • FIG. 15 is an image showing the results of typical Western blotting in Experimental Example 5.
  • FIG. 16 (a) is a dose-response curve for the production of MCP-1 of each compound measured in Experimental Example 6.
  • FIG. 16 (b) is a dose-response curve for the production of IP-10 of each compound measured in Experimental Example 6.
  • the present invention provides a therapeutic agent for Nakajo-Nishimura syndrome, which comprises a histone deacetylase (HDAC) inhibitor.
  • HDAC histone deacetylase
  • HDAC inhibitors suppress the overproduction of MCP-1 and IP-10 in NNS disease model cells and reduce them to healthy levels. Therefore, HDAC inhibitors can be used as therapeutic agents for Nakajo-Nishimura syndrome.
  • histone deacetylase inhibitors examples include CUDC-907 (CAS number: 1339928-25-4), JNJ-26481585 (CAS number: 875320-29-9), LAQ824 (CAS number: 404951-53-7), and the like.
  • examples thereof include romidepsin (CAS number: 128517-07-7), tricostatin A (CAS number: 58880-19-6), pharmacologically acceptable salts thereof, and solvates thereof.
  • Trichostatin A The chemical formula of Trichostatin A is shown in the following formula (5).
  • pharmaceutically acceptable salts include, for example, inorganic acid salts, alkali metal salts, alkaline earth metal salts, metal salts, ammonium salts, organic amine addition salts, amino acid addition salts and the like.
  • inorganic acid salts such as hydrochlorides, sulfates, hydrobromates, nitrates, phosphates; acetates, mesylates, succinates, maleates, fumarates, etc.
  • Organic acid salts such as citrate and tartrate; alkali metal salts such as sodium salt and potassium salt; alkaline earth metal salts such as magnesium salt and calcium salt; metal salts such as aluminum salt and zinc salt; ammonium salt and tetra Ammonium salts such as methylammonium salt; organic amine addition salts such as morpholin and piperidine; amino acid addition salts such as glycine, phenylalanine, lysine, aspartic acid and glutamic acid can be mentioned.
  • Examples of pharmaceutically acceptable solvates include hydrates and organic solvates.
  • the therapeutic agent of the present embodiment is preferably formulated as a pharmaceutical composition containing the above-mentioned compound and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition can be administered orally in the form of, for example, liquids, powders, granules, tablets, capsules, etc., or parenterally in the form of injections, suppositories, external skin preparations, etc. .. More specific examples of the external skin preparation include dosage forms such as ointments and patches.
  • binders such as gelatin, cornstarch, tragant gum, and gum arabic
  • excipients such as starch and crystalline cellulose
  • swelling agents such as alginic acid
  • solvents for injections such as water, ethanol, and glycerin
  • adhesives such as rubber-based adhesives and silicone-based adhesives.
  • the pharmaceutical composition may contain additives.
  • Additives include lubricants such as calcium stearate and magnesium stearate; sweeteners such as sucrose, lactose, saccharin and martitol; flavors such as peppermint and red mono oil; stabilizers such as benzyl alcohol and phenol; phosphoric acid. Buffering agents such as salts and sodium acetate; solubilizers such as benzyl benzoate and benzyl alcohol; antioxidants such as ascorbic acid; preservatives such as paraoxybenzoic acid esters (paraben), benzalconium chloride, chlorobutanol, cresol, etc. Agents and the like can be mentioned.
  • the dose of the pharmaceutical composition varies depending on the subject's symptoms, body weight, age, gender, etc. and cannot be unconditionally determined, but in the case of oral administration, for example, 0.1 to 100 mg / kg body weight per administration unit form.
  • the active ingredient (the above-mentioned compound) may be administered once a day or in 2 to 4 divided doses. In the case of an injection, for example, 0.01 to 50 mg of the active ingredient may be administered per administration unit form.
  • the present invention provides a method for treating Nakajo-Nishimura syndrome, which comprises administering an effective amount of a histone deacetylase inhibitor to a subject in need of treatment.
  • the histone deacetylase inhibitor is the same as described above.
  • the "effective amount" of the compound refers to the amount of the compound required to bring about a therapeutic effect on the treated subject, and more specifically, at least one or more related to Nakajo-Nishimura syndrome. The amount may be sufficient to prevent, delay, or minimize the symptoms of.
  • the "effective amount" of a compound is the prevention, delay, or minimization of one or more symptoms associated with Nakajo-Nishimura syndrome, either alone or in combination with other compounds or other treatments. It may be sufficient to do so. Effective amounts of compounds may vary depending on the severity of symptoms, route of administration, use of excipients, and combination with other therapeutic treatments, as will be appreciated by those skilled in the art.
  • the present invention provides a histone deacetylase inhibitor for use in the treatment of Nakajo-Nishimura syndrome.
  • the histone deacetylase inhibitor is the same as described above.
  • the present invention provides the use of histone deacetylase inhibitors for the production of therapeutic agents for Nakajo-Nishimura syndrome.
  • the histone deacetylase inhibitor is the same as described above.
  • Cell culture Fibroblasts collected from 2 healthy subjects (healthy subjects # 1 and # 2) and 2 NNS patients (NNS # 1 and # 2) were used. As cells, 10% fetal bovine serum (catalog number "F0926", Sigma-Aldrich) was added to Dulbecco's modified Eagle's medium (catalog number "08459-64", Nacalai Tesque). Cells were dissociated and subcultured into single cells using 0.25% trypsin EDTA solution (Cat. No. "25200-072", Thermo Fisher Scientific).
  • the fibroblasts used in the experiment were 10 ng / mL TNF- ⁇ (catalog number "210-TA”, R & D Systems) and 10 ng / mL IFN- ⁇ (catalog number "285-IF") to induce immune proteasomes. R & D Systems) stimulated for 72 hours.
  • MT-iPS-MLs is a cell line obtained by inducing differentiation of iPS cells (iPSCs) established from fibroblasts (NNS # 1) into cells having a homozygous phenotype (MLs), and is a PSMB8 gene.
  • iPSCs iPS cells
  • NNS # 1 fibroblasts
  • MLs a homozygous phenotype
  • WT-iPS-MLs are wild-type cell lines that are genetically homogeneous with MT-iPS-MLs.
  • MT-iPS-MLs a homozygous mutation of the PSMB8 gene similar to MT-iPS-MLs was introduced into KhES1, which is a human embryonic stem cell (ESCs) strain, by genome editing, and this was introduced into a cell line having a monocyte phenotype. Differentiation was induced into MLs) to obtain MT-ES-MLs.
  • WT-ES-MLs are wild-type cell lines that are genetically homogeneous with MT-ES-MLs.
  • WT-iPS-MLs, MT-iPS-MLs, WT-ES-MLs, MT-ES-MLs were added to StemPro TM- 34 SFM medium (catalog number "10639-011", Thermofisher Scientific) at 2 mM L. -Glutamine (catalog number "25030-081", Thermofisher Scientific), 50 ng / mL macrophage Colony Stimulating Factor (M-CSF, catalog number "216-MC”, R & D Systems) and 50 ng / mL granulocyte-macrosis It was cultured in a medium supplemented with factoror (GM-CSF, catalog number "215-GM”, R & D Systems).
  • M-CSF macrophage Colony Stimulating Factor
  • High-throughput screening 5,821 compounds, which have already been reported to have bioactive effects, were screened by high-throughput screening (HTS). Screening was performed by chemokine measurement based on homogeneous time-resolved fluorescence (HTRF). Table 1 below shows the compound library used for screening.
  • MT-iPS-MLs were seeded in 384-well plates at 5 ⁇ 10 3 per well and compounds were added at 1 ⁇ M (primary screening) or 100 nM (secondary screening). After incubation for 3 hours, 50 ng / mL lipopolysaccharide (Lipopolysaccharide (LPS), catalog number "tlll-peklps", in vivogen) for induction of MCP-1 or 100 ng / for induction of IP-10. mL IFN- ⁇ was added.
  • LPS lipopolysaccharide
  • ELISA Enzyme-linked immunosorbent assay
  • MT-ES-MLs were 5 ⁇ 10 4 (cell viability) or 1 ⁇ 10 4 (cytotoxic), NNS # One fibroblast was seeded at a cell density of 5 ⁇ 10 3 (cell viability) or 2 ⁇ 10 3 (cytotoxic), and the compound was added at an arbitrary concentration. After incubation for 24 hours, a commercially available kit (product name "Cell Counting Kit-8", catalog number "CK04", Cytotoxicity Research Institute) was used to measure cell viability, and a commercially available kit (product) was used to measure cytotoxicity.
  • RNA extraction and quantitative RT-PCR Total RNA was extracted using the RNeasy Mini kit (catalog number “74106", Qiagen) and treated with RNase-free DNase (catalog number "79254", Qiagen). The purified RNA was reverse transcribed using a commercially available kit (product name "PrimeScript TM RT Master Mix”, catalog number "RR037A”, Takara). qPCR was performed using a commercially available kit (product name "StepOnePlus TM ", Applied Biosystems, product name "TB Green Premix Ex Taq II", catalog number "RR820A”, Takara). The base sequence of the primers used is shown in Table 2 below.
  • the total amount of protein in the cytolyte was measured using a commercially available kit (product name "DC TM Protein Assay", catalog number “500-0116JA”, Biorad) and EnVision Multilabel Plate Readers (PerkinElmer), and each was measured. The protein concentration during cytolysis was adjusted to be the same.
  • cytolytic solution was boiled for 5 minutes in 4 ⁇ Remley sample buffer (catalog number “161-0747”, Bio-Rad) containing 2-mercaptoethanol (catalog number “21418-42”, Nacalai Tesque). Subsequently, the protein was separated by SDS polyacrylamide gel electrophoresis and transferred to Immobilon-P membrane (catalog number "IPVH000010], Merck).
  • the membrane is Tris-buffered saline supplemented with 10% skim milk (catalog number "190-12865", Fujifilm Wako Pure Chemical Industries, Ltd.) and 0.1% Tween 20 (catalog number "9005-64-5", Sigma-Aldrich). It was blocked with water and reacted with the antibody.
  • Anti-GAPDH monoclonal antibody catalog number "2118", cell signaling technology
  • anti-MCP-1 polyclonal antibody catalog number "ab9669", abcam
  • anti-IP-10 polyclonal antibody catalog number "ab8098", abcam
  • HRP labeling Anti-rabbit IgG antibody catalog number "7074", cell signaling technology
  • HRP-labeled anti-mouse IgG antibody catalog number "7076", cell signaling technology
  • HTS histone deacetylase inhibitors as potential therapeutic agents for NNS
  • HTS identified potential therapeutic agents for NNS.
  • Compounds were evaluated by the rate of inhibition of production of MCP-1 and IP-10, both of which are pro-inflammatory chemokines that are particularly elevated in NNS patients.
  • IL-6 is also particularly elevated in NNS patients, but blocking the IL-6 receptor by administration of the anti-IL-6 receptor antibody tocilizumab has a limited therapeutic effect on NNS, and thus MCP- We focused on compounds that inhibit the production of both 1 and IP-10.
  • the compound library was composed of 5,821 compounds and contained approved agents, kinase inhibitors, and bioactive compounds.
  • FIG. 1 shows the experiment schedule. After treating the cells with the compound for 3 hours, LPS or IFN- ⁇ treatment induced the production of MCP-1 or MP-10. MT-iPS-MLs were used in the primary and secondary screenings.
  • FIG. 2 is a graph showing the production inhibition rates of MCP-1 and IP-10.
  • 642 compounds having an MCP-1 production inhibition rate of more than 60%, an IP-10 production inhibition rate of more than 50%, or both production inhibition rates of more than 30% are defined as hit compounds. bottom.
  • the compound existing in the region surrounded by the square is a hit compound. The reproducibility of the hit compounds was confirmed and those suspected of having strong cytotoxicity were removed, and 108 of the 642 compounds were evaluated by the secondary screening.
  • FIG. 3 is a graph showing the production inhibition rates of MCP-1 and IP-10. As shown in FIG. 3, 26 compounds in which the production inhibition rate of MCP-1 and IP-10 was more than 80% with respect to the compound concentration of 100 nM were designated as hit compounds. In FIG. 3, the compound existing in the region surrounded by the square is a hit compound.
  • FIG. 4 is a graph showing the production inhibition rates of MCP-1 and IP-10. As a result, as shown in FIG. 4, 13 compounds having a production inhibition rate of MCP-1 and IP-10 of more than 80% were designated as hit compounds. In FIG. 4, the compound existing in the region surrounded by the square is a hit compound.
  • FIG. 5 is a graph showing the results of cytotoxicity evaluation.
  • the vertical axis shows the amount of free lactate dehydrogenase (LDH).
  • the dotted line indicates the mean value + 1 standard deviation (SD).
  • “United” shows the result of the negative control in which DMSO was added instead of the compound
  • “Lysis” shows the result of the cytolytic solution used as the positive control.
  • the four compounds were considered non-cytotoxic because the amount of free LDH was less than the mean of the untreated control + 1 standard deviation (SD).
  • FIG. 6 is a schematic diagram showing the results of HTS. As shown in FIG. 6, since 3 of the 4 hit compounds were histone deacetylase (HDAC) inhibitors, a more detailed study was conducted on these 3 compounds.
  • HDAC histone deacetylase
  • Table 3 below shows the 50% inhibitory concentration (IC 50 ) of each compound calculated based on FIGS. 7 (a) to 7 (f).
  • CUDC-907 was found to exhibit the lowest an IC 50 value for the production of MCP-1 and IP-10.
  • HDAC inhibitors are known to arrest the cell cycle. Therefore, in order to confirm the safety of CUDC-907, the effect on cell viability and cytotoxicity on MT-iPS-MLs was examined.
  • CUDC-907 was treated at a maximum concentration of 100 nM, and the cell viability and cytotoxicity with respect to the DMSO-treated group were examined.
  • Cell viability was assessed by detecting intracellular reduced nicotinamide adenine dinucleotide (NADH).
  • Cytotoxicity was assessed by detecting extracellular lactate dehydrogenase (LDH) activity.
  • DMSO indicates the result of adding DMSO instead of the compound
  • Lysite indicates the result of cytolysis.
  • CUDC-907 showed almost the same cell viability and cytotoxicity as the DMSO-treated group at any concentration. From this result, it was clarified that CUDC-907 is a compound showing an effect of inhibiting the production of MCP-1 and IP-10 with extremely low toxicity to MT-iPS-MLs.
  • the expression level of immune proteasome in fibroblasts is lower than the expression level in immune cells. Therefore, the immunoproteasome was induced by treatment with TNF- ⁇ and IFN- ⁇ for a longer period of time (72 hours) to sufficiently show the phenotype of the NNS patient, and then the amount of cytokine produced was measured.
  • "UT” indicates the result of fibroblasts not treated with TNF- ⁇ and IFN- ⁇ .
  • "*”, "**”, and "***” have significant differences in p ⁇ 0.05, p ⁇ 0.01, and p ⁇ 0.005 as a result of Student's T-test, respectively. Show that.
  • FIG. 10 is a graph showing the dose-dependent effect of inhibiting the production of MCP-1 of CUDC-907 in NNS # 1, which is a fibroblast derived from an NNS patient.
  • IC 50 of the production inhibition of MCP-1 by CUDC-907 in fibroblasts was found to be comparable with the values in the MT-iPS-MLs.
  • DMSO indicates the result of adding DMSO instead of CUDC-907
  • Lysite indicates the result of cytolysis.
  • CUDC-907 shows almost no cytotoxicity at an effective concentration. From the above results, it is clear that CUDC-907 inhibits the production of MCP-1 by fibroblasts derived from NNS patients at the same concentration as that in MT-iPS-MLs without showing cytotoxicity. It became.
  • "**", "***”, and "*****” are the results of one-way analysis of variance (ANOVA) and Dunnett's multiple comparison, respectively, and p ⁇ 0. 0.01, p ⁇ 0.005, p ⁇ 0.001 indicate that there is a significant difference, and "NS" indicates that there is no significant difference.
  • “**” indicates that there is a significant difference at p ⁇ 0.01 as a result of Student's T-test, and “NS” indicates that there is no significant difference.
  • WT-iPS-MLs cells were treated with 10 nM CUDC-907 for 3 hours and then stimulated with 100 ng / mL TNF- ⁇ and 100 ng / mL IFN- ⁇ for 3, 9, 15 and 21 hours for quantitative RT-PCR. went.
  • the expression level of each gene was standardized by the expression level of the GAPDH gene.
  • FIG. 14A is a graph showing the results of measuring the expression level of the CCL2 gene. Further, FIG. 14 (b) is a graph showing the results of measuring the expression level of the CXCL10 gene. In FIGS. 14A and 14B, the vertical axis of the graph indicates the expression level (relative value).
  • CUDC-907 increases the expression level of these genes, unlike the result of ELISA. This result indicates that CUDC-907 inhibits the production of MCP-1 and IP-10 after transcription.
  • MCP-1 and IP-10 were examined at the protein level.
  • the extracellular release of these cytokines was inhibited by Brefeldin A treatment.
  • Breferdin A is known to inhibit protein transport from the endoplasmic reticulum to the Golgi apparatus.
  • WT-iPS-MLs cells were treated with 10 nM CUDC-907 for 30 minutes and then stimulated with 100 ng / mL TNF- ⁇ and 100 ng / mL IFN- ⁇ for 6 hours. Breferdin A was added 1 hour prior to cell recovery.
  • FIG. 15 is an image showing a typical Western blotting result. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was detected as a loading control. In FIG. 15, “-" indicates that it was not added, and “+” indicates that it was added.
  • Glyceraldehyde 3-phosphate dehydrogenase Glyceraldehyde 3-phosphate dehydrogenase
  • CUDC-907 reduced intracellular levels of MCP-1 and IP-10. This result indicates that CUDC-907 inhibits post-transcriptional production of MCP-1 and IP-10.
  • FIG. 16 (a) is a dose-response curve for the production of MCP-1 of each compound.
  • FIG. 16 (b) is a dose-response curve for IP-10 production of each compound.
  • Table 4 below shows the 50% inhibitory concentration (IC 50 ) of each compound calculated based on FIGS. 16 (a) and 16 (b).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This therapeutic agent for Nakajo-Nishimura syndrome comprises a histone deacetylase inhibitor, wherein the histone deacetylase inhibitor may be: at least one compound selected from the group consisting of CUDC-907, JNJ-26481585, LAQ824, romidepsin, and trichostatin A; a pharmaceutically acceptable salt thereof; or solvates thereof.

Description

中條・西村症候群の治療剤A therapeutic agent for Nakajo-Nishimura syndrome

 本発明は、中條・西村症候群の治療剤に関する。本願は、2020年5月7日に米国に仮出願された米国特許出願第63/021,112号明細書に基づき優先権を主張し、その内容をここに援用する。 The present invention relates to a therapeutic agent for Nakajo-Nishimura syndrome. This application claims priority based on US Patent Application No. 63 / 021,112, which was provisionally filed in the United States on May 7, 2020, the contents of which are incorporated herein by reference.

 中條・西村症候群(NNS)は、プロテアソーム関連自己炎症性症候群(PRAAS)の1つであり、proteasome subunit beta 8(PSMB8)/LMP-7遺伝子におけるホモ接合型の変異により生じる。NNSは、乳児期初期に、しもやけのような発疹を示すことから始まり、定期的な高熱を発症し、最終的には、主に上半身における脂肪・筋萎縮及び関節拘縮を生じる。 Nakajo-Nishimura syndrome (NNS) is one of the proteasome-related autoinflammatory syndromes (PRAAS) and is caused by homozygous mutations in the proteasome subunit beta 8 (PSMB8) / LMP-7 gene. NNS begins with a chilblain-like rash in early infancy, develops regular high fever, and ultimately results in fat / muscle atrophy and joint contracture, primarily in the upper body.

 全てのNNS患者は、PSMB8遺伝子に、ホモ接合型のc.602G>T変異(第602番目のヌクレオチド残基におけるグアニンからチミンへの変異)を有しており、この変異はβ5iサブユニットにおけるG201Vアミノ酸変異(第201番目のアミノ酸残基におけるグリシンからバリンへの変異)をもたらす。 All NNS patients are homozygous for the PSMB8 gene. It has a 602G> T mutation (a guanine to thymine mutation at the 602nd nucleotide residue), which is a G201V amino acid mutation at the β5i subunit (a glycine to valine mutation at the 201st amino acid residue). Mutation).

 NNS患者におけるPSMB8遺伝子の変異は、免疫プロテアソームの機能不全を引き起こし、これが細胞ストレスを誘導することで炎症を引き起こすと考えられている。しかしながら、免疫プロテアソームの機能不全により誘導される炎症の詳細なメカニズムは依然として不明な点が多い。NNS患者は炎症性ケモカインのmonocyte chemotactic protein-1(MCP-1)及びinterferon gamma-induced protein 10(IP-10)を過剰産生することが知られており、これが炎症症状に関連しているとされている。 Mutations in the PSMB8 gene in NNS patients are thought to cause immune proteasome dysfunction, which induces cell stress and thus inflammation. However, the detailed mechanism of inflammation induced by immune proteasome dysfunction remains unclear. NNS patients are known to overproduce the inflammatory chemokines monocyte chemotaxis protein-1 (MCP-1) and interferon gamma-induced protein 10 (IP-10), which have been implicated in inflammatory symptoms. ing.

 NNSの治療法としてはステロイド内服が行われているが、脂肪萎縮や関節拘縮には効果がなく、特に乳幼児に対する副作用も重篤なことから、新規治療薬の開発が切望されている。 Oral steroids are used as a treatment method for NNS, but they are not effective for fat atrophy and joint contracture, and have serious side effects especially for infants, so the development of new therapeutic agents is eagerly desired.

 発明者らは、以前に、ヒト人工多能性幹細胞(iPSCs)及びヒト胚性幹細胞(ESCs)を用いてNNS疾患モデル細胞を作製した(非特許文献1を参照。)。具体的には、NNSの患者より樹立した変異型iPSCs(MT-iPS)に対し、ゲノム編集技術CRISPR/Cas9システムを用いて変異を回復させた野生型iPSCs(WT-iPS)を作製した。また、ヒト胚性幹細胞株のKhES-1(WT-ES)に対しても同様の方法で変異を導入した変異型(MT-ES)を作製した。 The inventors have previously prepared NNS disease model cells using human induced pluripotent stem cells (iPSCs) and human embryonic stem cells (ESCs) (see Non-Patent Document 1). Specifically, wild-type iPSCs (WT-iPS) were prepared by recovering mutations from mutant iPSCs (MT-iPS) established from NNS patients using the genome editing technology CRISPR / Cas9 system. In addition, a mutant type (MT-ES) in which mutations were introduced into the human embryonic stem cell line KhES-1 (WT-ES) was prepared in the same manner.

 更に、炎症におけるNNSの疾患の再現を確認するため、これらの細胞を単球の表現型を持つmonocytic cell line(MLs)に分化及び株化させた(WT-iPS-MLs、MT-iPS-MLs、WT-ES-MLs、MT-ES-MLs)。 Furthermore, in order to confirm the reproduction of NNS disease in inflammation, these cells were differentiated and established into monocyte phenotype monocytic cell lines (MLs) (WT-iPS-MLs, MT-iPS-MLs). , WT-ES-MLs, MT-ES-MLs).

 続いて、作製したMLsに対し、TNF-α及びIFN-γにより誘導されるサイトカインやケモカインの産生量を比較したところ、MT-iPS-MLs、MT-ES-MLsにおいて、NNS特異的に過剰産生される、MCP-1、IP-10及びinterleukin-6(IL-6)の産生が増加していることが確認された。 Subsequently, when the production amounts of cytokines and chemokines induced by TNF-α and IFN-γ were compared with the produced MLs, overproduction was specifically overproduced in MT-iPS-MLs and MT-ES-MLs. It was confirmed that the production of MCP-1, IP-10 and interleukin-6 (IL-6) was increased.

 非特許文献1にはまた、NNSの治療薬の候補として、抗酸化剤やJanus Kinase(JAK)阻害剤が記載されている。 Non-Patent Document 1 also describes antioxidants and Janus Kinase (JAK) inhibitors as candidates for therapeutic agents for NNS.

Honda-Ozaki and Terashima et al., Pluripotent Stem Cell Model of Nakajo-Nishimura Syndrome Untangles Proinflammatory Pathways Mediated by Oxidative Stress, Stem Cell Reports, 10 (6), 1835-1850, 2018.Honda-Ozaki and Terashima et al., Pluripotent Stem Cell Model of Nakajo-Nishimura Syndrome Untangles Proinflammatory Pathways Mediated by Oxidative Stress, Stem Cell Reports, 10 (6), 1835-1850, 2018.

 しかしながら、抗酸化剤やJAK阻害剤は、NNS疾患モデル細胞における過剰なMCP-1及びIP-10の産生を健常レベルまで低下させるのに不十分である。そこで、本発明は、NNSの治療剤を提供することを目的とする。 However, antioxidants and JAK inhibitors are inadequate to reduce excess MCP-1 and IP-10 production in NNS disease model cells to healthy levels. Therefore, an object of the present invention is to provide a therapeutic agent for NNS.

 本発明は以下の態様を含む。
[1]ヒストン脱アセチル化酵素阻害剤を含む、中條・西村症候群の治療剤。
[2]前記ヒストン脱アセチル化酵素阻害剤が、CUDC-907、JNJ-26481585、LAQ824、ロミデプシン及びトリコスタチンAからなる群より選択される少なくとも1つの化合物又はその薬理学的に許容される塩若しくはそれらの溶媒和物である、[1]に記載の中條・西村症候群の治療剤。
The present invention includes the following aspects.
[1] A therapeutic agent for Nakajo-Nishimura syndrome, which contains a histone deacetylase inhibitor.
[2] The histone deacetylase inhibitor is at least one compound selected from the group consisting of CUDC-907, JNJ-26481585, LAQ824, romidepsin and tricostatin A, or a pharmacologically acceptable salt thereof or The therapeutic agent for Nakajo-Nishimura syndrome according to [1], which is a solvate thereof.

 本発明によれば、中條・西村症候群の治療剤を提供することができる。 According to the present invention, it is possible to provide a therapeutic agent for Nakajo-Nishimura syndrome.

図1は、実験例1の実験スケジュールを示す模式図である。FIG. 1 is a schematic diagram showing an experimental schedule of Experimental Example 1. 図2は、実験例1の1次スクリーニングにおける、化合物のMCP-1及びIP-10の産生阻害率を示すグラフである。FIG. 2 is a graph showing the production inhibition rates of the compounds MCP-1 and IP-10 in the primary screening of Experimental Example 1. 図3は、実験例1の2次スクリーニングにおける、化合物のMCP-1及びIP-10の産生阻害率を示すグラフである。FIG. 3 is a graph showing the production inhibition rates of the compounds MCP-1 and IP-10 in the secondary screening of Experimental Example 1. 図4は、実験例1のMT-ES-MLsにおける、化合物のMCP-1及びIP-10の産生阻害率を示すグラフである。FIG. 4 is a graph showing the production inhibition rates of the compounds MCP-1 and IP-10 in MT-ES-MLs of Experimental Example 1. 図5は、実験例1における細胞毒性評価の結果を示すグラフである。FIG. 5 is a graph showing the results of cytotoxicity evaluation in Experimental Example 1. 図6は、実験例1におけるハイスループットスクリーニングの結果を示す模式図である。FIG. 6 is a schematic diagram showing the results of high-throughput screening in Experimental Example 1. 図7(a)~(c)は、実験例2において測定した、各化合物のMCP-1の産生に対する用量反応曲線である。図7(d)~(f)は、実験例2において測定した、各化合物のIP-10の産生に対する用量反応曲線である。7 (a) to 7 (c) are dose-response curves for the production of MCP-1 of each compound measured in Experimental Example 2. 7 (d) to 7 (f) are dose-response curves for the production of IP-10 of each compound measured in Experimental Example 2. 図8(a)は、実験例2において、細胞内還元型ニコチンアミドアデニンジヌクレオチド(NADH)を測定した結果を示すグラフである。図8(b)は、実験例2において、細胞外乳酸脱水素酵素(LDH)活性を測定した結果を示すグラフである。FIG. 8A is a graph showing the results of measuring intracellular reduced nicotinamide adenine dinucleotide (NADH) in Experimental Example 2. FIG. 8B is a graph showing the results of measuring extracellular lactate dehydrogenase (LDH) activity in Experimental Example 2. 図9は、実験例3において測定した、各細胞によるMCP-1の発現量を示すグラフである。FIG. 9 is a graph showing the expression level of MCP-1 by each cell measured in Experimental Example 3. 図10は、実験例3において測定した、NNS患者由来の線維芽細胞における、CUDC-907の用量依存的なMCP-1の産生阻害効果を示すグラフである。FIG. 10 is a graph showing the dose-dependent effect of inhibiting the production of MCP-1 of CUDC-907 in fibroblasts derived from NNS patients, which was measured in Experimental Example 3. 図11(a)は、実験例3において、細胞内NADHを測定した結果を示すグラフである。図11(b)は、実験例3において細胞外LDH活性を測定した結果を示すグラフである。FIG. 11A is a graph showing the results of measuring intracellular NADH in Experimental Example 3. FIG. 11B is a graph showing the results of measuring the extracellular LDH activity in Experimental Example 3. 図12(a)は、実験例4において、各細胞におけるMCP-1の産生量を比較した結果を示すグラフである。図12(b)は、実験例4において、各細胞におけるIP-10の産生量を比較した結果を示すグラフである。FIG. 12A is a graph showing the results of comparing the production amount of MCP-1 in each cell in Experimental Example 4. FIG. 12B is a graph showing the results of comparing the amount of IP-10 produced in each cell in Experimental Example 4. 図13は、実験例4において測定した、健常者由来の線維芽細胞株及びNNS患者由来の線維芽細胞におけるMCP-1の産生量を示すグラフである。FIG. 13 is a graph showing the amount of MCP-1 produced in a fibroblast cell line derived from a healthy subject and a fibroblast cell derived from an NNS patient, which was measured in Experimental Example 4. 図14(a)は、実験例5において、CCL2遺伝子の発現量を測定した結果を示すグラフである。図14(b)は、実験例5において、CXCL10遺伝子の発現量を測定した結果を示すグラフである。FIG. 14A is a graph showing the results of measuring the expression level of the CCL2 gene in Experimental Example 5. FIG. 14B is a graph showing the results of measuring the expression level of the CXCL10 gene in Experimental Example 5. 図15は、実験例5における代表的なウエスタンブロッティングの結果を示す画像である。FIG. 15 is an image showing the results of typical Western blotting in Experimental Example 5. 図16(a)は、実験例6において測定した、各化合物のMCP-1の産生に対する用量反応曲線である。図16(b)は、実験例6において測定した、各化合物のIP-10の産生に対する用量反応曲線である。FIG. 16 (a) is a dose-response curve for the production of MCP-1 of each compound measured in Experimental Example 6. FIG. 16 (b) is a dose-response curve for the production of IP-10 of each compound measured in Experimental Example 6.

[中條・西村症候群の治療剤]
 1実施形態において、本発明は、ヒストン脱アセチル化酵素(HDAC)阻害剤を含む、中條・西村症候群の治療剤を提供する。
[Therapeutic agent for Nakajo-Nishimura syndrome]
In one embodiment, the present invention provides a therapeutic agent for Nakajo-Nishimura syndrome, which comprises a histone deacetylase (HDAC) inhibitor.

 実施例において後述するように、発明者らは、HDAC阻害剤がNNS疾患モデル細胞におけるMCP-1及びIP-10の過剰産生を抑制し、健常レベルまで低下させることを明らかにした。したがって、HDAC阻害剤を中條・西村症候群の治療剤として用いることができる。 As will be described later in the Examples, the inventors have shown that HDAC inhibitors suppress the overproduction of MCP-1 and IP-10 in NNS disease model cells and reduce them to healthy levels. Therefore, HDAC inhibitors can be used as therapeutic agents for Nakajo-Nishimura syndrome.

 ヒストン脱アセチル化酵素阻害剤としては、CUDC-907(CAS番号:1339928-25-4)、JNJ-26481585(CAS番号:875320-29-9)、LAQ824(CAS番号:404951-53-7)、ロミデプシン(CAS番号:128517-07-7)、トリコスタチンA(CAS番号:58880-19-6)、これらの薬理学的に許容される塩、これらの溶媒和物等が挙げられる。 Examples of histone deacetylase inhibitors include CUDC-907 (CAS number: 1339928-25-4), JNJ-26481585 (CAS number: 875320-29-9), LAQ824 (CAS number: 404951-53-7), and the like. Examples thereof include romidepsin (CAS number: 128517-07-7), tricostatin A (CAS number: 58880-19-6), pharmacologically acceptable salts thereof, and solvates thereof.

 CUDC-907の化学式を下記式(1)に示す。

Figure JPOXMLDOC01-appb-C000001
The chemical formula of CUDC-907 is shown in the following formula (1).
Figure JPOXMLDOC01-appb-C000001

 JNJ-26481585の化学式を下記式(2)に示す。

Figure JPOXMLDOC01-appb-C000002
The chemical formula of JNJ-26481585 is shown in the following formula (2).
Figure JPOXMLDOC01-appb-C000002

 LAQ824の化学式を下記式(3)に示す。

Figure JPOXMLDOC01-appb-C000003
The chemical formula of LAQ824 is shown in the following formula (3).
Figure JPOXMLDOC01-appb-C000003

 ロミデプシンの化学式を下記式(4)に示す。

Figure JPOXMLDOC01-appb-C000004
The chemical formula of romidepsin is shown in the following formula (4).
Figure JPOXMLDOC01-appb-C000004

 トリコスタチンAの化学式を下記式(5)に示す。

Figure JPOXMLDOC01-appb-C000005
The chemical formula of Trichostatin A is shown in the following formula (5).
Figure JPOXMLDOC01-appb-C000005

 本実施形態の治療剤において、薬学的に許容される塩としては、例えば、無機酸塩、アルカリ金属塩、アルカリ土類金属塩、金属塩、アンモニウム塩、有機アミン付加塩、アミノ酸付加塩等が挙げられる。より具体的には、例えば、塩酸塩、硫酸塩、臭化水素酸塩、硝酸塩、リン酸塩等の無機酸塩;酢酸塩、メシル酸塩、コハク酸塩、マレイン酸塩、フマル酸塩、クエン酸塩、酒石酸塩等の有機酸塩;ナトリウム塩、カリウム塩等のアルカリ金属塩;マグネシウム塩、カルシウム塩等のアルカリ土類金属塩;アルミニウム塩、亜鉛塩等の金属塩;アンモニウム塩、テトラメチルアンモニウム塩等のアンモニウム塩;モルホリン、ピペリジン等の有機アミン付加塩;グリシン、フェニルアラニン、リジン、アスパラギン酸、グルタミン酸等のアミノ酸付加塩等が挙げられる。また、薬学的に許容される溶媒和物としては、例えば、水和物、有機溶媒和物等が挙げられる。 In the therapeutic agent of the present embodiment, pharmaceutically acceptable salts include, for example, inorganic acid salts, alkali metal salts, alkaline earth metal salts, metal salts, ammonium salts, organic amine addition salts, amino acid addition salts and the like. Can be mentioned. More specifically, for example, inorganic acid salts such as hydrochlorides, sulfates, hydrobromates, nitrates, phosphates; acetates, mesylates, succinates, maleates, fumarates, etc. Organic acid salts such as citrate and tartrate; alkali metal salts such as sodium salt and potassium salt; alkaline earth metal salts such as magnesium salt and calcium salt; metal salts such as aluminum salt and zinc salt; ammonium salt and tetra Ammonium salts such as methylammonium salt; organic amine addition salts such as morpholin and piperidine; amino acid addition salts such as glycine, phenylalanine, lysine, aspartic acid and glutamic acid can be mentioned. Examples of pharmaceutically acceptable solvates include hydrates and organic solvates.

 本実施形態の治療剤は、上述した化合物と、薬学的に許容される担体とを含む医薬組成物として製剤化されていることが好ましい。医薬組成物は、例えば、液剤、散剤、顆粒剤、錠剤、カプセル剤等の形態で経口的に、あるいは、注射剤、坐剤、皮膚外用剤等の形態で非経口的に投与することができる。皮膚外用剤としては、より具体的には、軟膏剤、貼付剤等の剤型が挙げられる。 The therapeutic agent of the present embodiment is preferably formulated as a pharmaceutical composition containing the above-mentioned compound and a pharmaceutically acceptable carrier. The pharmaceutical composition can be administered orally in the form of, for example, liquids, powders, granules, tablets, capsules, etc., or parenterally in the form of injections, suppositories, external skin preparations, etc. .. More specific examples of the external skin preparation include dosage forms such as ointments and patches.

 薬学的に許容される担体としては、通常医薬組成物の製剤に用いられるものを特に制限なく用いることができる。より具体的には、例えば、ゼラチン、コーンスターチ、トラガントガム、アラビアゴム等の結合剤;デンプン、結晶性セルロース等の賦形剤;アルギン酸等の膨化剤;水、エタノール、グリセリン等の注射剤用溶剤;ゴム系粘着剤、シリコーン系粘着剤等の粘着剤等が挙げられる。 As the pharmaceutically acceptable carrier, those usually used for the preparation of pharmaceutical compositions can be used without particular limitation. More specifically, for example, binders such as gelatin, cornstarch, tragant gum, and gum arabic; excipients such as starch and crystalline cellulose; swelling agents such as alginic acid; solvents for injections such as water, ethanol, and glycerin; Examples thereof include adhesives such as rubber-based adhesives and silicone-based adhesives.

 医薬組成物は添加剤を含んでいてもよい。添加剤としては、ステアリン酸カルシウム、ステアリン酸マグネシウム等の潤滑剤;ショ糖、乳糖、サッカリン、マルチトール等の甘味剤;ペパーミント、アカモノ油等の香味剤;ベンジルアルコール、フェノール等の安定剤;リン酸塩、酢酸ナトリウム等の緩衝剤;安息香酸ベンジル、ベンジルアルコール等の溶解補助剤;アスコルビン酸等の酸化防止剤;パラオキシ安息香酸エステル類(パラベン)、塩化ベンザルコニウム、クロロブタノール、クレゾール等の防腐剤等が挙げられる。 The pharmaceutical composition may contain additives. Additives include lubricants such as calcium stearate and magnesium stearate; sweeteners such as sucrose, lactose, saccharin and martitol; flavors such as peppermint and red mono oil; stabilizers such as benzyl alcohol and phenol; phosphoric acid. Buffering agents such as salts and sodium acetate; solubilizers such as benzyl benzoate and benzyl alcohol; antioxidants such as ascorbic acid; preservatives such as paraoxybenzoic acid esters (paraben), benzalconium chloride, chlorobutanol, cresol, etc. Agents and the like can be mentioned.

 医薬組成物の投与量は、対象の症状、体重、年齢、性別等によって異なり、一概には決定できないが、経口投与の場合には、例えば、投与単位形態あたり0.1~100mg/kg体重の有効成分(上述した化合物)を1日1回又は2~4回程度に分けて投与すればよい。また、注射剤の場合には、例えば、投与単位形態あたり0.01~50mgの有効成分を投与すればよい。 The dose of the pharmaceutical composition varies depending on the subject's symptoms, body weight, age, gender, etc. and cannot be unconditionally determined, but in the case of oral administration, for example, 0.1 to 100 mg / kg body weight per administration unit form. The active ingredient (the above-mentioned compound) may be administered once a day or in 2 to 4 divided doses. In the case of an injection, for example, 0.01 to 50 mg of the active ingredient may be administered per administration unit form.

[その他の実施形態]
 1実施形態において、本発明は、ヒストン脱アセチル化酵素阻害剤の有効量を、治療を必要とする対象に投与することを含む、中條・西村症候群の治療方法を提供する。ヒストン脱アセチル化酵素阻害剤については上述したものと同様である。ここで、化合物の「有効量」とは、処置された対象に治療効果をもたらすのに必要とされる化合物の量を指し、より具体的には、中條・西村症候群に関連する少なくとも1つ以上の症状を阻止、遅延、又は最小限にするために十分な量であってよい。ある態様において、化合物の「有効量」とは、化合物単独で、又は他の化合物ないしは他の処置を組み合わせて、中條・西村症候群に関連する1つ以上の症状を阻止、遅延、又は最小限にするために十分な量であってよい。化合物の有効量は、当業者によって認識されるように、症状の程度、投与経路、賦形剤の使用、及び他の治療処置との併用に応じて変化し得る。
[Other Embodiments]
In one embodiment, the present invention provides a method for treating Nakajo-Nishimura syndrome, which comprises administering an effective amount of a histone deacetylase inhibitor to a subject in need of treatment. The histone deacetylase inhibitor is the same as described above. Here, the "effective amount" of the compound refers to the amount of the compound required to bring about a therapeutic effect on the treated subject, and more specifically, at least one or more related to Nakajo-Nishimura syndrome. The amount may be sufficient to prevent, delay, or minimize the symptoms of. In some embodiments, the "effective amount" of a compound is the prevention, delay, or minimization of one or more symptoms associated with Nakajo-Nishimura syndrome, either alone or in combination with other compounds or other treatments. It may be sufficient to do so. Effective amounts of compounds may vary depending on the severity of symptoms, route of administration, use of excipients, and combination with other therapeutic treatments, as will be appreciated by those skilled in the art.

 1実施形態において、本発明は、中條・西村症候群の治療における使用のための、ヒストン脱アセチル化酵素阻害剤を提供する。ヒストン脱アセチル化酵素阻害剤については上述したものと同様である。 In one embodiment, the present invention provides a histone deacetylase inhibitor for use in the treatment of Nakajo-Nishimura syndrome. The histone deacetylase inhibitor is the same as described above.

 1実施形態において、本発明は、中條・西村症候群の治療剤の製造のための、ヒストン脱アセチル化酵素阻害剤の使用を提供する。ヒストン脱アセチル化酵素阻害剤については上述したものと同様である。 In one embodiment, the present invention provides the use of histone deacetylase inhibitors for the production of therapeutic agents for Nakajo-Nishimura syndrome. The histone deacetylase inhibitor is the same as described above.

 次に実施例を示して本発明を更に詳細に説明するが、本発明は以下の実施例に限定されるものではない。 Next, the present invention will be described in more detail with reference to Examples, but the present invention is not limited to the following Examples.

[材料及び方法]
 以下の実験は、京都大学(R0091/G0259)と和歌山県立医科大学の倫理委員会によって承認された。ヘルシンキ宣言にしたがって、患者又はその保護者から書面によるインフォームドコンセントを得た。ヒト胚性幹細胞(ESCs)の使用は、文部科学省によって承認された。
[Materials and methods]
The following experiments were approved by the Institutional Review Board of Kyoto University (R0091 / G0259) and Wakayama Medical University. Written informed consent was obtained from the patient or his or her guardian in accordance with the Declaration of Helsinki. The use of human embryonic stem cells (ESCs) has been approved by the Ministry of Education, Culture, Sports, Science and Technology.

(細胞培養)
 線維芽細胞は、健常者2名(健常者#1及び#2)及びNNS患者2名(NNS#1及び#2)より採取したものを用いた。細胞は、ダルベッコ変法イーグル培地(カタログ番号「08459-64」、ナカライテスク)に10%ウシ胎児血清(カタログ番号「F0926」、シグマ-アルドリッチ)を加えたものを用いた。細胞は0.25%トリプシンEDTA溶液(カタログ番号「25200-072」、サーモフィッシャーサイエンティフィック)を用いて単一細胞に解離して継代した。実験に用いた線維芽細胞は、免疫プロテアソームを誘導するため、10ng/mL TNF-α(カタログ番号「210-TA」、R&Dシステムズ)及び10ng/mL IFN-γ(カタログ番号「285-IF」、R&Dシステムズ)で72時間刺激した。
(Cell culture)
Fibroblasts collected from 2 healthy subjects (healthy subjects # 1 and # 2) and 2 NNS patients (NNS # 1 and # 2) were used. As cells, 10% fetal bovine serum (catalog number "F0926", Sigma-Aldrich) was added to Dulbecco's modified Eagle's medium (catalog number "08459-64", Nacalai Tesque). Cells were dissociated and subcultured into single cells using 0.25% trypsin EDTA solution (Cat. No. "25200-072", Thermo Fisher Scientific). The fibroblasts used in the experiment were 10 ng / mL TNF-α (catalog number "210-TA", R & D Systems) and 10 ng / mL IFN-γ (catalog number "285-IF") to induce immune proteasomes. R & D Systems) stimulated for 72 hours.

 非特許文献1に記載された、WT-iPS-MLs、MT-iPS-MLs、WT-ES-MLs、MT-ES-MLs細胞を使用した。MT-iPS-MLsは、線維芽細胞(NNS#1)から樹立したiPS細胞(iPSCs)を、単球の表現型を持つ細胞(MLs)に分化誘導して得た細胞株であり、PSMB8遺伝子にホモ接合型の変異を有する。WT-iPS-MLsは、MT-iPS-MLsと遺伝的に同質の野生型細胞株である。 The WT-iPS-MLs, MT-iPS-MLs, WT-ES-MLs, and MT-ES-MLs cells described in Non-Patent Document 1 were used. MT-iPS-MLs is a cell line obtained by inducing differentiation of iPS cells (iPSCs) established from fibroblasts (NNS # 1) into cells having a homozygous phenotype (MLs), and is a PSMB8 gene. Has a homozygous mutation in. WT-iPS-MLs are wild-type cell lines that are genetically homogeneous with MT-iPS-MLs.

 また、ヒト胚性幹細胞(ESCs)株であるKhES1に、ゲノム編集によりMT-iPS-MLsと同様のPSMB8遺伝子のホモ接合型の変異を導入し、これを単球の表現型を持つ細胞株(MLs)に分化誘導してMT-ES-MLsを得た。WT-ES-MLsは、MT-ES-MLsと遺伝的に同質の野生型細胞株である。 In addition, a homozygous mutation of the PSMB8 gene similar to MT-iPS-MLs was introduced into KhES1, which is a human embryonic stem cell (ESCs) strain, by genome editing, and this was introduced into a cell line having a monocyte phenotype. Differentiation was induced into MLs) to obtain MT-ES-MLs. WT-ES-MLs are wild-type cell lines that are genetically homogeneous with MT-ES-MLs.

 WT-iPS-MLs、MT-iPS-MLs、WT-ES-MLs、MT-ES-MLsは、StemProTM-34 SFM培地(カタログ番号「10639-011」、サーモフィッシャーサイエンティフィック)に、2mM L-グルタミン(カタログ番号「25030-081」、サーモフィッシャーサイエンティフィック)、50ng/mL macrophage Colony Stimulating Factor(M-CSF、カタログ番号「216-MC」、R&Dシステムズ)及び50ng/mL granulocyte macrophage colony-stimulating factor(GM-CSF、カタログ番号「215-GM」、R&Dシステムズ)を添加した培地で培養した。 WT-iPS-MLs, MT-iPS-MLs, WT-ES-MLs, MT-ES-MLs were added to StemPro TM- 34 SFM medium (catalog number "10639-011", Thermofisher Scientific) at 2 mM L. -Glutamine (catalog number "25030-081", Thermofisher Scientific), 50 ng / mL macrophage Colony Stimulating Factor (M-CSF, catalog number "216-MC", R & D Systems) and 50 ng / mL granulocyte-macrosis It was cultured in a medium supplemented with factoror (GM-CSF, catalog number "215-GM", R & D Systems).

(ハイスループットスクリーニング)
 既に生理活性作用が報告されている5,821化合物をハイスループットスクリーニング(HTS)によりスクリーニングした。スクリーニングは、均一系時間分解蛍光(homogeneous time-resolved fluorescence、HTRF)に基づくケモカイン測定により行った。下記表1に、スクリーニングに使用した化合物ライブラリを示す。
(High-throughput screening)
5,821 compounds, which have already been reported to have bioactive effects, were screened by high-throughput screening (HTS). Screening was performed by chemokine measurement based on homogeneous time-resolved fluorescence (HTRF). Table 1 below shows the compound library used for screening.

Figure JPOXMLDOC01-appb-T000006
Figure JPOXMLDOC01-appb-T000006

 1次及び2次スクリーニングでは、384ウェルプレートにMT-iPS-MLsを1ウェルあたり5×10個で播種し、化合物を1μM(1次スクリーニング)又は100nM(2次スクリーニング)添加した。3時間インキュベートした後、MCP-1の誘導のために50ng/mLリポ多糖(Lipopolysaccharid(LPS)、カタログ番号「tlrl-peklps」、インビボジェン)を、又は、IP-10の誘導のために100ng/mL IFN-γを添加した。続いて、21時間インキュベートした後、上清を別の384ウェルプレートに移し、Human CCL2(MCP-1)Kit(カタログ番号「62HCCL2PEG」、シスバイオ)又はHuman CXCL10(IP-10)Kit(カタログ番号「62HCX10PEG、シスバイオ)を加え、PowerScan4(DSファーマバイオメディカル)によりMCP-1又はIP-10の濃度を測定した。続いて、MT-ES-MLsを用いて、2次スクリーニングと同様の手順によりヒット化合物の検証を行った。 In the primary and secondary screenings, MT-iPS-MLs were seeded in 384-well plates at 5 × 10 3 per well and compounds were added at 1 μM (primary screening) or 100 nM (secondary screening). After incubation for 3 hours, 50 ng / mL lipopolysaccharide (Lipopolysaccharide (LPS), catalog number "tlll-peklps", in vivogen) for induction of MCP-1 or 100 ng / for induction of IP-10. mL IFN-γ was added. Subsequently, after incubating for 21 hours, the supernatant was transferred to another 384-well plate and transferred to Human CCL2 (MCP-1) Kit (catalog number "62HCCL2PEG", cisbio) or Human CXCL10 (IP-10) Kit (catalog number "". 62HCX10PEG, cisbio) was added and the concentration of MCP-1 or IP-10 was measured by PowerScan4 (DS Pharma Biomedical). Subsequently, using MT-ES-MLs, the hit compound was subjected to the same procedure as in the secondary screening. Was verified.

(酵素結合免疫吸着検定法(ELISA))
 MLs又は線維芽細胞の培養上清中のMCP-1及びIP-10の濃度の測定は、市販のキット(製品名「LEGENDplexTM Human Adipokine Panel、バイオレジェンド)を用いて行なった。上清採取のために、細胞を96ウェルプレートに1ウェルあたり1×10個(MLs)又は5×10個(線維芽細胞)の細胞密度で播種し、ジメチルスルホキシド(DMSO、カタログ番号「D2650」、シグマアルドリッチ)に溶解した化合物を任意の濃度で添加した。
(Enzyme-linked immunosorbent assay (ELISA))
The concentrations of MCP-1 and IP-10 in the culture supernatant of MLs or fibroblasts were measured using a commercially available kit (product name "LEGENDplex TM Human Adipokine Panel, BioLegend"). To allow cells to be seeded in 96-well plates at a cell density of 1 x 10 4 (MLs) or 5 x 10 3 (fibroblasts) per well, dimethylsulfoxide (DMSO, catalog number "D2650", sigma). The compound dissolved in Aldrich) was added at an arbitrary concentration.

 続いて、3時間インキュベートした後、100ng/mL(MLs)又は10ng/mL(線維芽細胞)のTNF-α及びIFN-γを添加し刺激した。21時間インキュベートした後、上清を採取した。 Subsequently, after incubating for 3 hours, 100 ng / mL (MLs) or 10 ng / mL (fibroblasts) of TNF-α and IFN-γ were added and stimulated. After incubating for 21 hours, the supernatant was collected.

(細胞生存率及び細胞毒性の測定)
 細胞生存率及び細胞毒性評価のために、細胞を96ウェルプレートに1ウェルあたり、MT-ES-MLsは5×10個(細胞生存率)又は1×10個(細胞毒性)、NNS#1線維芽細胞は5×10個(細胞生存率)又は2×10個(細胞毒性)の細胞密度で播種し、化合物を任意の濃度で添加した。24時間インキュベートした後、細胞生存率の測定では、市販のキット(製品名「Cell Counting Kit-8」、カタログ番号「CK04」、同仁化学研究所)、細胞毒性の測定では、市販のキット(製品名「Cytotoxicity Detection KitPLUS」、カタログ番号「4744934001」、メルク)を加え、2104 EnVision Multilabel Plate Readers(パーキンエルマー)で測定した。細胞生存率・細胞毒性評価の陽性コントロール(100%細胞死)として、同数の細胞の溶解液を使用した。
(Measurement of cell viability and cytotoxicity)
For cell viability and cytotoxicity assessment, per well of cells in a 96-well plate, MT-ES-MLs were 5 × 10 4 (cell viability) or 1 × 10 4 (cytotoxic), NNS # One fibroblast was seeded at a cell density of 5 × 10 3 (cell viability) or 2 × 10 3 (cytotoxic), and the compound was added at an arbitrary concentration. After incubation for 24 hours, a commercially available kit (product name "Cell Counting Kit-8", catalog number "CK04", Cytotoxicity Research Institute) was used to measure cell viability, and a commercially available kit (product) was used to measure cytotoxicity. The name "Cytotoxicity Detection Kit PLUS ", catalog number "4744934001", Merck) was added, and the measurement was performed with 2104 EnVision Multilabel Plate Readers (PerkinElmer). As a positive control (100% cell death) for cell viability / cytotoxicity evaluation, the same number of cell lysates were used.

(RNA抽出及び定量的RT-PCR(qPCR))
 RNeasy Mini kit(カタログ番号「74106」、キアゲン)を用いて全RNAを抽出し、RNase-free DNase(カタログ番号「79254」、キアゲン)で処理した。精製したRNAは市販のキット(製品名「PrimeScriptTM RT Master Mix」、カタログ番号「RR037A」、タカラ)を用いて逆転写した。qPCRは市販のキット(製品名「StepOnePlusTM」、アプライドバイオシステムズ、及び、製品名「TB Green Premix Ex Taq II」、カタログ番号「RR820A」、タカラ)を用いて行った。使用したプライマーの塩基配列を下記表2に示す。
(RNA extraction and quantitative RT-PCR (qPCR))
Total RNA was extracted using the RNeasy Mini kit (catalog number "74106", Qiagen) and treated with RNase-free DNase (catalog number "79254", Qiagen). The purified RNA was reverse transcribed using a commercially available kit (product name "PrimeScript TM RT Master Mix", catalog number "RR037A", Takara). qPCR was performed using a commercially available kit (product name "StepOnePlus TM ", Applied Biosystems, product name "TB Green Premix Ex Taq II", catalog number "RR820A", Takara). The base sequence of the primers used is shown in Table 2 below.

Figure JPOXMLDOC01-appb-T000007
Figure JPOXMLDOC01-appb-T000007

(ウエスタンブロッティング)
 3×10個の細胞を、プロテアーゼ阻害剤カクテル(カタログ番号「04080-11」、ナカライテスク)を添加したRIPAバッファー(カタログ番号「188-02453」、富士フイルム和光純薬)中、氷上で30分静置して溶解した。続いて、150,000×g、4℃で5分間遠心した後、上清を回収した。
(Western blotting)
30 x 10 5 cells on ice in a RIPA buffer (catalog number "188-02453", Fujifilm Wako Pure Chemical Industries) supplemented with a protease inhibitor cocktail (catalog number "04080-11", Nacalai Tesque). It was allowed to stand for a minute and dissolved. Subsequently, after centrifuging at 150,000 × g at 4 ° C. for 5 minutes, the supernatant was collected.

 細胞溶解液中の全タンパク質量を、市販のキット(製品名「DCTM Protein Assay」、カタログ番号「500-0116JA」、バイオラッド)及びEnVision Multilabel Plate Readers(パーキンエルマー)を用いて測定し、各細胞溶解中のタンパク質濃度が同一になるよう調整した。 The total amount of protein in the cytolyte was measured using a commercially available kit (product name "DC TM Protein Assay", catalog number "500-0116JA", Biorad) and EnVision Multilabel Plate Readers (PerkinElmer), and each was measured. The protein concentration during cytolysis was adjusted to be the same.

 細胞溶解液を、2-メルカプトエタノール(カタログ番号「21418-42」、ナカライテスク)を含む4×レムリーサンプルバッファー(カタログ番号「161-0747」、バイオラッド)中で5分間煮沸した。続いて、タンパク質をSDSポリアクリルアミドゲル電気泳動により分離し、Immobilon-P membrane(カタログ番号「IPVH00010]、メルク)に転写した。 The cytolytic solution was boiled for 5 minutes in 4 × Remley sample buffer (catalog number “161-0747”, Bio-Rad) containing 2-mercaptoethanol (catalog number “21418-42”, Nacalai Tesque). Subsequently, the protein was separated by SDS polyacrylamide gel electrophoresis and transferred to Immobilon-P membrane (catalog number "IPVH000010], Merck).

 膜は、10%脱脂乳(カタログ番号「190-12865」、富士フイルム和光純薬)及び0.1%Tween20(カタログ番号「9005-64-5」、シグマ-アルドリッチ)を添加したトリス緩衝生理食塩水でブロッキングし、抗体と反応させた。 The membrane is Tris-buffered saline supplemented with 10% skim milk (catalog number "190-12865", Fujifilm Wako Pure Chemical Industries, Ltd.) and 0.1% Tween 20 (catalog number "9005-64-5", Sigma-Aldrich). It was blocked with water and reacted with the antibody.

 使用した抗体は以下の通りであった。抗GAPDHモノクローナル抗体(カタログ番号「2118」、セルシグナリングテクノロジー)、抗MCP-1ポリクローナル抗体(カタログ番号「ab9669」、アブカム)、抗IP-10ポリクローナル抗体(カタログ番号「ab8098」、アブカム)、HRP標識抗ウサギIgG抗体(カタログ番号「7074」、セルシグナリングテクノロジー)及びHRP標識抗マウスIgG抗体(カタログ番号「7076」、セルシグナリングテクノロジー)。 The antibodies used were as follows. Anti-GAPDH monoclonal antibody (catalog number "2118", cell signaling technology), anti-MCP-1 polyclonal antibody (catalog number "ab9669", abcam), anti-IP-10 polyclonal antibody (catalog number "ab8098", abcam), HRP labeling Anti-rabbit IgG antibody (catalog number "7074", cell signaling technology) and HRP-labeled anti-mouse IgG antibody (catalog number "7076", cell signaling technology).

 化学発光には市販のキット(製品名「SuperSignalTM West Femto Maximum Sensitivity Substrate」、カタログ番号「34095」、サーモフィッシャーサイエンティフィック)を使用した。イメージは ImageQuant LAS 4000(GEヘルスケア)を使用して取得した。 A commercially available kit (product name "SuperSignal TM West Femto Maximum Sensitivity Substrate", catalog number "34095", Thermo Fisher Scientific) was used for chemiluminescence. Images were acquired using ImageQuant LAS 4000 (GE Healthcare).

(統計解析)
 全ての統計解析及びIC50値の算出はGraphPad Prismソフトウエア(GraphPad Software)を使用して行った。
(Statistical analysis)
Calculated for all statistical analyzes and IC 50 values were performed using GraphPad Prism software (GraphPad Software).

[実験例1]
(HTSにより、ヒストン脱アセチル化酵素阻害剤がNNSの治療剤の候補であることが同定された)
 HTSにより、NNSの治療剤の候補を同定した。いずれもNNS患者で特に上昇する炎症誘発性ケモカインである、MCP-1及びIP-10の産生阻害率で化合物を評価した。IL-6もNNS患者で特に上昇するが、抗IL-6受容体抗体であるトシリズマブの投与によりIL-6受容体をブロックしても、NNSの治療効果が限定的であることから、MCP-1及びIP-10の双方の産生を阻害する化合物に着目した。
[Experimental Example 1]
(HTS identified histone deacetylase inhibitors as potential therapeutic agents for NNS)
HTS identified potential therapeutic agents for NNS. Compounds were evaluated by the rate of inhibition of production of MCP-1 and IP-10, both of which are pro-inflammatory chemokines that are particularly elevated in NNS patients. IL-6 is also particularly elevated in NNS patients, but blocking the IL-6 receptor by administration of the anti-IL-6 receptor antibody tocilizumab has a limited therapeutic effect on NNS, and thus MCP- We focused on compounds that inhibit the production of both 1 and IP-10.

 上記表1に示すように、化合物ライブラリは5,821化合物から構成され、承認薬、キナーゼ阻害薬、生理活性化合物を含んでいた。図1に実験スケジュールを示す。細胞を化合物で3時間処理した後、LPS又はIFN-γ処理によりMCP-1又はMP-10の産生を誘導した。1次及び2次スクリーニングでは、MT-iPS-MLsを使用した。 As shown in Table 1 above, the compound library was composed of 5,821 compounds and contained approved agents, kinase inhibitors, and bioactive compounds. FIG. 1 shows the experiment schedule. After treating the cells with the compound for 3 hours, LPS or IFN-γ treatment induced the production of MCP-1 or MP-10. MT-iPS-MLs were used in the primary and secondary screenings.

 まず、5,821化合物のMT-iPS-MLsに対する効果を1μMの濃度で評価した。図2は、MCP-1及びIP-10の産生阻害率を示すグラフである。図2に示すように、MCP-1の産生阻害率が60%超、IP-10の産生阻害率が50%超、又は、両者の産生阻害率が30%超である642化合物をヒット化合物とした。図2中、四角で囲んだ領域に存在する化合物がヒット化合物である。ヒット化合物に対し、再現性の確認及び強い細胞毒性が疑われるものの除去を行い、化合物642種のうち108種を2次スクリーニングで評価した。 First, the effect of 5,821 compounds on MT-iPS-MLs was evaluated at a concentration of 1 μM. FIG. 2 is a graph showing the production inhibition rates of MCP-1 and IP-10. As shown in FIG. 2, 642 compounds having an MCP-1 production inhibition rate of more than 60%, an IP-10 production inhibition rate of more than 50%, or both production inhibition rates of more than 30% are defined as hit compounds. bottom. In FIG. 2, the compound existing in the region surrounded by the square is a hit compound. The reproducibility of the hit compounds was confirmed and those suspected of having strong cytotoxicity were removed, and 108 of the 642 compounds were evaluated by the secondary screening.

 2次スクリーニングも同様にMCP-1及びIP-10の産生阻害率で評価を行った。図3は、MCP-1及びIP-10の産生阻害率を示すグラフである。図3に示すように、化合物の濃度100nMに対し、MCP-1及びIP-10の産生阻害率が80%超である26化合物をヒット化合物とした。図3中、四角で囲んだ領域に存在する化合物がヒット化合物である。 The secondary screening was also evaluated based on the production inhibition rate of MCP-1 and IP-10. FIG. 3 is a graph showing the production inhibition rates of MCP-1 and IP-10. As shown in FIG. 3, 26 compounds in which the production inhibition rate of MCP-1 and IP-10 was more than 80% with respect to the compound concentration of 100 nM were designated as hit compounds. In FIG. 3, the compound existing in the region surrounded by the square is a hit compound.

 次に、これらヒット化合物のMCP-1及びIP-10産生阻害効果がクローン特異的である可能性を排除するため、MT-ES-MLsを用い2次スクリーニングと同様の条件で26化合物を評価した。図4は、MCP-1及びIP-10の産生阻害率を示すグラフである。その結果、図4に示すように、MCP-1及びIP-10の産生阻害率が80%超である13化合物をヒット化合物とした。図4中、四角で囲んだ領域に存在する化合物がヒット化合物である。 Next, in order to eliminate the possibility that the MCP-1 and IP-10 production inhibitory effects of these hit compounds were clone-specific, 26 compounds were evaluated using MT-ES-MLs under the same conditions as in the secondary screening. .. FIG. 4 is a graph showing the production inhibition rates of MCP-1 and IP-10. As a result, as shown in FIG. 4, 13 compounds having a production inhibition rate of MCP-1 and IP-10 of more than 80% were designated as hit compounds. In FIG. 4, the compound existing in the region surrounded by the square is a hit compound.

 最後に、これらの13化合物の濃度100nMにおけるMT-ES-MLsに対する毒性を評価した。図5は、細胞毒性評価の結果を示すグラフである。縦軸は遊離した乳酸脱水素酵素(LDH)の量を示す。点線は、平均値+1標準偏差(SD)を示す。図5中、「Untreated」は化合物の代わりにDMSOを添加したネガティブコントロールの結果を示し、「Lysis」はポジティブコントロールとして用いた細胞溶解液の結果を示す。図5に示すように、4化合物は、遊離したLDHの量が未処理コントロールの平均値+1標準偏差(SD)未満であったことから、細胞毒性がないと考えられた。 Finally, the toxicity of these 13 compounds to MT-ES-MLs at a concentration of 100 nM was evaluated. FIG. 5 is a graph showing the results of cytotoxicity evaluation. The vertical axis shows the amount of free lactate dehydrogenase (LDH). The dotted line indicates the mean value + 1 standard deviation (SD). In FIG. 5, "United" shows the result of the negative control in which DMSO was added instead of the compound, and "Lysis" shows the result of the cytolytic solution used as the positive control. As shown in FIG. 5, the four compounds were considered non-cytotoxic because the amount of free LDH was less than the mean of the untreated control + 1 standard deviation (SD).

 図6は、HTSの結果を示す模式図である。図6に示すように、ヒットした4化合物のうち3化合物がヒストン脱アセチル化酵素(HDAC)阻害剤であったことから、これらの3化合物についてより詳細な検討を行った。 FIG. 6 is a schematic diagram showing the results of HTS. As shown in FIG. 6, since 3 of the 4 hit compounds were histone deacetylase (HDAC) inhibitors, a more detailed study was conducted on these 3 compounds.

[実験例2]
(CUDC-907は低濃度でMCP-1及びIP-10の産生を阻害した)
 実験例1で同定された3種のHDAC阻害剤である、CUDC-907、JNJ-26481585及びLAQ824について、MT-iPS-MLsによるMCP-1及びIP-10の産生阻害効果を検討した。
[Experimental Example 2]
(CUDC-907 inhibited the production of MCP-1 and IP-10 at low concentrations)
For the three HDAC inhibitors identified in Experimental Example 1, CUDC-907, JNJ-26481585 and LAQ824, the effect of MT-iPS-MLs on the production of MCP-1 and IP-10 was examined.

 図7(a)~(c)は各化合物のMCP-1の産生に対する用量反応曲線である(n=3)。図7(d)~(f)は各化合物のIP-10の産生に対する用量反応曲線である(n=3)。下記表3に、図7(a)~(f)に基づいて算出した各化合物の50%阻害濃度(IC50)を示す。 7 (a) to 7 (c) are dose-response curves for the production of MCP-1 of each compound (n = 3). 7 (d) to 7 (f) are dose-response curves for the production of IP-10 of each compound (n = 3). Table 3 below shows the 50% inhibitory concentration (IC 50 ) of each compound calculated based on FIGS. 7 (a) to 7 (f).

Figure JPOXMLDOC01-appb-T000008
Figure JPOXMLDOC01-appb-T000008

 その結果、3化合物のうち、CUDC-907がMCP-1及びIP-10の産生に対する最も低いIC50値を示すことが明らかとなった。 As a result, among the three compounds, CUDC-907 was found to exhibit the lowest an IC 50 value for the production of MCP-1 and IP-10.

 HDAC阻害剤は細胞周期を停止させることが知られている。そこで、CUDC-907の安全性を確認するため、MT-iPS-MLsに対する細胞生存率及び細胞毒性への影響を検討した。 HDAC inhibitors are known to arrest the cell cycle. Therefore, in order to confirm the safety of CUDC-907, the effect on cell viability and cytotoxicity on MT-iPS-MLs was examined.

 CUDC-907を最大100nMの濃度で処理し、DMSO処理群に対する細胞生存率及び細胞毒性を検討した。細胞生存率は、細胞内還元型ニコチンアミドアデニンジヌクレオチド(NADH)を検出することにより評価した。細胞毒性は、細胞外乳酸脱水素酵素(LDH)活性を検出することにより評価した。 CUDC-907 was treated at a maximum concentration of 100 nM, and the cell viability and cytotoxicity with respect to the DMSO-treated group were examined. Cell viability was assessed by detecting intracellular reduced nicotinamide adenine dinucleotide (NADH). Cytotoxicity was assessed by detecting extracellular lactate dehydrogenase (LDH) activity.

 図8(a)は、細胞内NADHを測定した結果を示すグラフである(n=3)。図8(b)は、細胞外LDH活性を測定した結果を示すグラフである(n=3)。図8(a)及び(b)中、「DMSO」は化合物の代わりにDMSOを添加した結果であることを示し、「Lysate」は細胞溶解液の結果であることを示す。 FIG. 8A is a graph showing the results of measuring intracellular NADH (n = 3). FIG. 8B is a graph showing the results of measuring extracellular LDH activity (n = 3). In FIGS. 8A and 8B, "DMSO" indicates the result of adding DMSO instead of the compound, and "Lysite" indicates the result of cytolysis.

 その結果、CUDC-907は、いずれの濃度においてもDMSO処理群とほぼ同等の細胞生存率及び細胞毒性を示した。この結果から、CUDC-907が、MT-iPS-MLsに対して、極めて低い毒性でMCP-1及びIP-10の産生阻害効果を示す化合物であることが明らかとなった。 As a result, CUDC-907 showed almost the same cell viability and cytotoxicity as the DMSO-treated group at any concentration. From this result, it was clarified that CUDC-907 is a compound showing an effect of inhibiting the production of MCP-1 and IP-10 with extremely low toxicity to MT-iPS-MLs.

[実験例3]
(CUDC-907はNNS患者由来の線維芽細胞におけるMCP-1及びIP-10の産生を効果的に阻害した)
 CUDC-907の効果がMT-iPS-MLs特異的でないことを確認するため、NNS患者由来線維芽細胞を用いてその効果を確認した。
[Experimental Example 3]
(CUDC-907 effectively inhibited the production of MCP-1 and IP-10 in fibroblasts from NNS patients)
In order to confirm that the effect of CUDC-907 is not specific to MT-iPS-MLs, the effect was confirmed using fibroblasts derived from NNS patients.

 線維芽細胞における免疫プロテアソームの発現量は免疫細胞における発現量よりも低い。そこで、TNF-α及びIFN-γで、より長時間(72時間)処理して免疫プロテアソームを誘導し、NNS患者の表現型を十分に示す状態にした後にサイトカインの産生量を測定した。 The expression level of immune proteasome in fibroblasts is lower than the expression level in immune cells. Therefore, the immunoproteasome was induced by treatment with TNF-α and IFN-γ for a longer period of time (72 hours) to sufficiently show the phenotype of the NNS patient, and then the amount of cytokine produced was measured.

 線維芽細胞からのIP-10の発現量は非常に少なかったため、健常者由来の線維芽細胞とNNS患者由来の線維芽細胞を比較することはできなかった。 Since the expression level of IP-10 from fibroblasts was very low, it was not possible to compare fibroblasts derived from healthy subjects with fibroblasts derived from NNS patients.

 図9は、健常者由来の線維芽細胞である、Healthy#1、Healthy#2、及び、NNS患者由来の線維芽細胞である、NNS#1、NNS#2におけるMCP-1の発現量を示すグラフである(n=3)。図9中、「UT」はTNF-α及びIFN-γで処理していない線維芽細胞の結果であることを示す。また、「*」、「**」、「***」は、それぞれ、スチューデントのT検定の結果、p<0.05、p<0.01、p<0.005で有意差が存在することを示す。 FIG. 9 shows the expression levels of MCP-1 in Health # 1, Health # 2, and NNS patient-derived fibroblasts, NNS # 1 and NNS # 2, which are fibroblasts derived from healthy subjects. It is a graph (n = 3). In FIG. 9, "UT" indicates the result of fibroblasts not treated with TNF-α and IFN-γ. In addition, "*", "**", and "***" have significant differences in p <0.05, p <0.01, and p <0.005 as a result of Student's T-test, respectively. Show that.

 図10は、NNS患者由来の線維芽細胞であるNNS#1における、CUDC-907の用量依存的なMCP-1の産生阻害効果を示すグラフである。その結果、線維芽細胞におけるCUDC-907によるMCP-1の産生阻害のIC50はMT-iPS-MLsにおける値と同程度であることが明らかとなった。 FIG. 10 is a graph showing the dose-dependent effect of inhibiting the production of MCP-1 of CUDC-907 in NNS # 1, which is a fibroblast derived from an NNS patient. As a result, IC 50 of the production inhibition of MCP-1 by CUDC-907 in fibroblasts was found to be comparable with the values in the MT-iPS-MLs.

 続いて、NNS患者由来線維芽細胞であるNNS#1に対するCUDC-907の細胞生存率及び細胞毒性への影響を検討した。図11(a)は、細胞内NADHを測定した結果を示すグラフである(n=3)。図11(b)は、細胞外LDH活性を測定した結果を示すグラフである(n=3)。図11(a)及び(b)中、「DMSO」はCUDC-907の代わりにDMSOを添加した結果であることを示し、「Lysate」は細胞溶解液の結果であることを示す。 Subsequently, the effects of CUDC-907 on cell viability and cytotoxicity on NNS # 1, which is a fibroblast derived from NNS patients, were examined. FIG. 11A is a graph showing the results of measuring intracellular NADH (n = 3). FIG. 11B is a graph showing the results of measuring extracellular LDH activity (n = 3). In FIGS. 11 (a) and 11 (b), "DMSO" indicates the result of adding DMSO instead of CUDC-907, and "Lysite" indicates the result of cytolysis.

 その結果、CUDC-907は、有効濃度においてほとんど細胞毒性を示さないことが明らかとなった。以上の結果から、CUDC-907は、NNS患者由来の線維芽細胞によるMCP-1の産生を、MT-iPS-MLsにおけるものと同程度の濃度で、細胞毒性を示さずに阻害することが明らかとなった。 As a result, it was clarified that CUDC-907 shows almost no cytotoxicity at an effective concentration. From the above results, it is clear that CUDC-907 inhibits the production of MCP-1 by fibroblasts derived from NNS patients at the same concentration as that in MT-iPS-MLs without showing cytotoxicity. It became.

[実験例4]
(CUDC-907によるMCP-1及びIP-10の産生阻害効果の検討)
 CUDC-907のNNS疾患モデルに対するMCP-1及びIP-10の産生阻害効果が、野生型コントロールの産生量と比較して十分であるかを確認した。
[Experimental Example 4]
(Examination of the effect of CUDC-907 on inhibiting the production of MCP-1 and IP-10)
It was confirmed whether the production inhibitory effect of MCP-1 and IP-10 on the NNS disease model of CUDC-907 was sufficient as compared with the production amount of the wild-type control.

 図12(a)は、CUDC-907の存在下及び非存在下において、WT-iPS-MLs及びMT-iPS-MLsのMCP-1の産生量を比較した結果を示すグラフである(n=3)。図12(b)は、CUDC-907の存在下及び非存在下において、WT-iPS-MLs及びMT-iPS-MLsのIP-10の産生量を比較した結果を示すグラフである(n=3)。図12(a)及び(b)中、「**」、「***」、「****」は、それぞれ、一元配置分散分析(ANOVA)及びダネットの多重比較の結果、p<0.01、p<0.005、p<0.001で有意差が存在することを示し、「N.S.」は有意差がないことを示す。 FIG. 12A is a graph showing the results of comparing the production amounts of MCP-1 of WT-iPS-MLs and MT-iPS-MLs in the presence and absence of CUDC-907 (n = 3). ). FIG. 12B is a graph showing the results of comparing the production amounts of IP-10 of WT-iPS-MLs and MT-iPS-MLs in the presence and absence of CUDC-907 (n = 3). ). In FIGS. 12A and 12B, "**", "***", and "*****" are the results of one-way analysis of variance (ANOVA) and Dunnett's multiple comparison, respectively, and p <0. 0.01, p <0.005, p <0.001 indicate that there is a significant difference, and "NS" indicates that there is no significant difference.

 その結果、10nMのCUDC-907をMT-iPS-MLsに処理したところ、MCP-1及びIP-10の産生量はWT-iPS-MLsと同等かそれ以下まで減少することが明らかとなった。 As a result, when 10 nM CUDC-907 was treated into MT-iPS-MLs, it was clarified that the production amounts of MCP-1 and IP-10 were reduced to the same level as or less than WT-iPS-MLs.

 また、図13は、健常者由来の線維芽細胞株である、Healthy#1、Healthy#2におけるMCP-1の産生量、及び、CUDC-907の存在下及び非存在下において、NNS患者由来の線維芽細胞である、NNS#1、NNS#2におけるMCP-1の産生量を示すグラフである(n=3)。図13中、「**」は、スチューデントのT検定の結果、p<0.01で有意差が存在することを示し、「N.S.」は有意差がないことを示す。 In addition, FIG. 13 shows the amount of MCP-1 produced in Health # 1 and Health # 2, which are fibroblast cell lines derived from healthy subjects, and derived from NNS patients in the presence and absence of CUDC-907. It is a graph which shows the production amount of MCP-1 in NNS # 1 and NNS # 2, which are fibroblasts (n = 3). In FIG. 13, “**” indicates that there is a significant difference at p <0.01 as a result of Student's T-test, and “NS” indicates that there is no significant difference.

 その結果、Healthy#1、Healthy#2に対しNNS#1、#2でMCP-1産生の増加が確認された。更に、30nMのCUDC-907をNNS#1、#2に処理したところ、MCP-1の産生量はHealthy#1、Healthy#2と同等かそれ以下まで減少した。 As a result, an increase in MCP-1 production was confirmed in NNS # 1 and # 2 with respect to Health # 1 and Health # 2. Furthermore, when 30 nM CUDC-907 was treated with NNS # 1 and # 2, the amount of MCP-1 produced decreased to the same level as or less than that of Health # 1 and Health # 2.

 以上の結果から、CUDC-907の効果は、NNSによる過剰なMCP-1及びIP-10の産生を健常レベルまで減少させるのに十分であることが明らかとなった。 From the above results, it was clarified that the effect of CUDC-907 is sufficient to reduce the excessive production of MCP-1 and IP-10 by NNS to a healthy level.

[実験例5]
(CUDC-907は転写後にMCP-1及びIP-10の産生を阻害した)
 CUDC-907によるMCP-1及びIP-10の産生阻害のメカニズムを明らかにするために、MCP-1をコードするCCL2遺伝子及びIP-10をコードするCXCL10遺伝子の発現レベルを定量した。
[Experimental Example 5]
(CUDC-907 inhibited the production of MCP-1 and IP-10 after transcription)
To elucidate the mechanism by which CUDC-907 inhibits the production of MCP-1 and IP-10, the expression levels of the CCL2 gene encoding MCP-1 and the CXCL10 gene encoding IP-10 were quantified.

 WT-iPS-MLs細胞を10nMのCUDC-907で3時間処理した後、100ng/mL TNF-α及び100ng/mL IFN-γで3、9、15、21時間刺激し、定量的RT-PCRを行った。各遺伝子の発現量はGAPDH遺伝子の発現量で標準化した。 WT-iPS-MLs cells were treated with 10 nM CUDC-907 for 3 hours and then stimulated with 100 ng / mL TNF-α and 100 ng / mL IFN-γ for 3, 9, 15 and 21 hours for quantitative RT-PCR. went. The expression level of each gene was standardized by the expression level of the GAPDH gene.

 図14(a)はCCL2遺伝子の発現量を測定した結果を示すグラフである。また、図14(b)はCXCL10遺伝子の発現量を測定した結果を示すグラフである。図14(a)及び(b)中、グラフの縦軸は、発現量(相対値)を示す。 FIG. 14A is a graph showing the results of measuring the expression level of the CCL2 gene. Further, FIG. 14 (b) is a graph showing the results of measuring the expression level of the CXCL10 gene. In FIGS. 14A and 14B, the vertical axis of the graph indicates the expression level (relative value).

 その結果、ELISAの結果と異なり、CUDC-907はこれらの遺伝子の発現レベルを上昇させることが明らかとなった。この結果は、CUDC-907が転写後にMCP-1及びIP-10の産生を阻害することを示す。 As a result, it was clarified that CUDC-907 increases the expression level of these genes, unlike the result of ELISA. This result indicates that CUDC-907 inhibits the production of MCP-1 and IP-10 after transcription.

 続いて、MCP-1及びIP-10の発現量をタンパク質レベルで検討した。ウエスタンブロッティングにより細胞内におけるMCP-1及びIP-10の発現量を検出するため、これらのサイトカインの細胞外への放出をブレフェルジンA(Brefeldin A)処理により阻害した。ブレフェルジンAは、小胞体からゴルジ体へのタンパク質輸送を阻害することが知られている。 Subsequently, the expression levels of MCP-1 and IP-10 were examined at the protein level. In order to detect the intracellular expression levels of MCP-1 and IP-10 by Western blotting, the extracellular release of these cytokines was inhibited by Brefeldin A treatment. Breferdin A is known to inhibit protein transport from the endoplasmic reticulum to the Golgi apparatus.

 WT-iPS-MLs細胞を10nMのCUDC-907で30分間処理した後、100ng/mL TNF-α及び100ng/mL IFN-γで6時間刺激した。ブレフェルジンAは細胞回収の1時間前に添加した。 WT-iPS-MLs cells were treated with 10 nM CUDC-907 for 30 minutes and then stimulated with 100 ng / mL TNF-α and 100 ng / mL IFN-γ for 6 hours. Breferdin A was added 1 hour prior to cell recovery.

 図15は代表的なウエスタンブロッティングの結果を示す画像である。グリセルアルデヒド3リン酸脱水素酵素(GAPDH)をローディングコントロールとして検出した。図15中、「-」は添加しなかったことを示し、「+」は添加したことを示す。 FIG. 15 is an image showing a typical Western blotting result. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was detected as a loading control. In FIG. 15, "-" indicates that it was not added, and "+" indicates that it was added.

 その結果、ブレフェルジンAの存在下では、MCP-1及びIP-10の細胞内レベルが上昇した。この結果は、ブレフェルジンA処理がMCP-1及びIP-10の細胞外への放出を阻害したことを示す。 As a result, in the presence of Breferdin A, the intracellular levels of MCP-1 and IP-10 increased. This result indicates that Breferdin A treatment inhibited the extracellular release of MCP-1 and IP-10.

 また、ブレフェルジンAの存在下において、CUDC-907は、MCP-1及びIP-10の細胞内レベルを減少させた。この結果は、CUDC-907が、MCP-1及びIP-10の産生を転写後に阻害することを示す。 Also, in the presence of Breferdin A, CUDC-907 reduced intracellular levels of MCP-1 and IP-10. This result indicates that CUDC-907 inhibits post-transcriptional production of MCP-1 and IP-10.

[実験例6]
(HDAC阻害剤によるMCP-1及びIP-10の産生阻害)
 HDAC阻害剤である、CUDC-907、JNJ-26481585、LAQ824、ロミデプシン及びトリコスタチンAについて、MT-iPS-MLsによるMCP-1及びIP-10の産生阻害効果を検討した。
[Experimental Example 6]
(Inhibition of MCP-1 and IP-10 production by HDAC inhibitors)
The effects of MT-iPS-MLs on the production of MCP-1 and IP-10 were investigated for the HDAC inhibitors CUDC-907, JNJ-26481585, LAQ824, romidepsin and tricostatin A.

 図16(a)は各化合物のMCP-1の産生に対する用量反応曲線である。図16(b)は各化合物のIP-10の産生に対する用量反応曲線である。下記表4に、図16(a)及び(b)に基づいて算出した各化合物の50%阻害濃度(IC50)を示す。 FIG. 16 (a) is a dose-response curve for the production of MCP-1 of each compound. FIG. 16 (b) is a dose-response curve for IP-10 production of each compound. Table 4 below shows the 50% inhibitory concentration (IC 50 ) of each compound calculated based on FIGS. 16 (a) and 16 (b).

Figure JPOXMLDOC01-appb-T000009
Figure JPOXMLDOC01-appb-T000009

 本発明によれば、中條・西村症候群の治療剤を提供することができる。 According to the present invention, it is possible to provide a therapeutic agent for Nakajo-Nishimura syndrome.

Claims (2)

 ヒストン脱アセチル化酵素阻害剤を含む、中條・西村症候群の治療剤。 A therapeutic agent for Nakajo-Nishimura syndrome, including a histone deacetylase inhibitor.  前記ヒストン脱アセチル化酵素阻害剤が、CUDC-907、JNJ-26481585、LAQ824、ロミデプシン及びトリコスタチンAからなる群より選択される少なくとも1つの化合物又はその薬理学的に許容される塩若しくはそれらの溶媒和物である、請求項1に記載の中條・西村症候群の治療剤。 The histone deacetylase inhibitor is at least one compound selected from the group consisting of CUDC-907, JNJ-26481585, LAQ824, romidepsin and tricostatin A, or a pharmacologically acceptable salt thereof or a solvent thereof. The therapeutic agent for Nakajo-Nishimura syndrome according to claim 1, which is a Japanese product.
PCT/JP2021/017367 2020-05-07 2021-05-06 Therapeutic agent for nakajo-nishimura syndrome Ceased WO2021225135A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2022519962A JP7709752B2 (en) 2020-05-07 2021-05-06 Treatment for Nakajo-Nishimura syndrome

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063021112P 2020-05-07 2020-05-07
US63/021,112 2020-05-07

Publications (1)

Publication Number Publication Date
WO2021225135A1 true WO2021225135A1 (en) 2021-11-11

Family

ID=78468000

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2021/017367 Ceased WO2021225135A1 (en) 2020-05-07 2021-05-06 Therapeutic agent for nakajo-nishimura syndrome

Country Status (2)

Country Link
JP (1) JP7709752B2 (en)
WO (1) WO2021225135A1 (en)

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BREHM, A. ET AL. ET AL.: "Additive loss-of-function proteasome subunit mutations in CANDLE/PRAAS patients promote type I IFN production", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 125, no. 11, 2015, pages 4196 - 4211, XP055873092 *
COWARD, W. ET AL. ET AL.: "Repression of IP-10 by Interactions between Histone Deacetylation and Hypermethylation in Idiopathic Pulmonary Fibrosis", MOLECULAR AND CELLULAR BIOLOGY, vol. 30, no. 12, 2010, pages 2874 - 2886, XP055873095 *
HONDA-OZAKI, F. ET AL. ET AL.: "Pluripotent Stem Cell Model of Nakajo-Nishimura Syndrome Untangles Proinflammatory Pathways Mediated by Oxidative Stress", STEM CELL REPORTS, vol. 10, 2018, pages 1835 - 1850, XP055873090 *
HULL, E. ET AL. ET AL.: "HDAC Inhibitors as Epigenetic Regulators of the Immune System: Impacts on Cancer Therapy and Inflammatory Diseases", BIOMED RESEARCH INTERNATIONAL, 2016, XP055717155, DOI: 10.1155/2016/8797206 *
KASE, N. ET AL . ET AL.: "Pluripotent stem cell -based screening identifies CUDC-907 as an effective compound for restoring the in vitro phenotype of Nakajo-Nishimura syndrome", STEM CELLS TRANSLATIONAL MEDICINE, vol. 10, no. 3, 14 October 2020 (2020-10-14), pages 455 - 464, XP055873099, DOI: 10.1002/sctm.20-0198 *

Also Published As

Publication number Publication date
JPWO2021225135A1 (en) 2021-11-11
JP7709752B2 (en) 2025-07-17

Similar Documents

Publication Publication Date Title
CN116947880B (en) Aryl receptor modulators and methods of making and using the same
JP5038905B2 (en) CXCR4 antagonist for the treatment of medical disorders
US20220098190A1 (en) Compounds and uses thereof
US9408845B2 (en) Formulations containing pyridazine compounds
RS65359B1 (en) Valbenazine dosage regimen for the treatment of hyperkinetic movement disorders
CA3083975A1 (en) Compounds for treatment of diseases related to dux4 expression
WO2002002090A2 (en) Inhibitors of copper-containing amine oxidases
AU2001282163A1 (en) Inhibitors of copper-containing amine oxidases
JP2008535779A (en) CXCR4 antagonist for the treatment of HIV infection
CN112996541A (en) Method for removing senescent cells and method for preparing senescent cells
EP3108883A1 (en) Therapeutic uses of non-peptide inhibitors of the calcineurin - nfat signalling pathway
WO2022226078A1 (en) Therapy for alcohol-related liver disease
WO2021158792A1 (en) Methods of treating autoimmune or inflammatory diseases or disorders
US20220048906A1 (en) Compounds and uses thereof
Shi et al. Compound Danshen Dripping Pill effectively alleviates cGAS-STING-triggered diseases by disrupting STING-TBK1 interaction
JP7709752B2 (en) Treatment for Nakajo-Nishimura syndrome
EP2123280A1 (en) Agent for prevention and/or treatment of systemic lupus erythematosus
Guemei et al. Do Desipramine [10, 11-Dihydro-5-[3-(methylamino) propyl]-5 H-dibenz [b, f] azepine monohydrochloride] and Fluoxetine [N-Methyl-3-phenyl-3-[4-(trifluoromethyl) phenoxy]-propan-1-amine] Ameliorate the Extent of Colonic Damage Induced by Acetic Acid in Rats?
US20190111016A1 (en) Methods of treating muscle and liver disorders
JP2024510949A (en) Compositions for the treatment of EBV-related diseases or conditions
CN114521142B (en) Compositions and methods for inhibiting and/or treating neurodegenerative diseases and/or clinical conditions thereof
US9650351B2 (en) Inhibition of IL-2 production
WO2022025709A1 (en) Preventive, relief or therapeutic use of 2,3,5-substituted thiophene compound against gastrointestinal stromal tumor
US20170165254A1 (en) Treating flavivirus infections with amodiaquine and derivatives thereof
KR20250145681A (en) How to Treat Dermatological Disorders

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: 21800457

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2022519962

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: 21800457

Country of ref document: EP

Kind code of ref document: A1