WO2019151514A1 - Monocyte differentiation inducer containing albendazole - Google Patents

Monocyte differentiation inducer containing albendazole Download PDF

Info

Publication number
WO2019151514A1
WO2019151514A1 PCT/JP2019/003802 JP2019003802W WO2019151514A1 WO 2019151514 A1 WO2019151514 A1 WO 2019151514A1 JP 2019003802 W JP2019003802 W JP 2019003802W WO 2019151514 A1 WO2019151514 A1 WO 2019151514A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
differentiation
leukemia
albendazole
monocyte
Prior art date
Application number
PCT/JP2019/003802
Other languages
French (fr)
Japanese (ja)
Inventor
剣 森田
壯一 足立
正敏 萩原
Original Assignee
国立大学法人京都大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立大学法人京都大学 filed Critical 国立大学法人京都大学
Publication of WO2019151514A1 publication Critical patent/WO2019151514A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D235/30Nitrogen atoms not forming part of a nitro radical
    • C07D235/32Benzimidazole-2-carbamic acids, unsubstituted or substituted; Esters thereof; Thio-analogues thereof

Definitions

  • This application relates to a monocyte differentiation inducer containing albendazole as an active ingredient.
  • hematopoietic stem cells there are diseases caused by inhibition of the differentiation process of hematopoietic stem cells, and examples include leukemia, aplastic anemia, and Costman syndrome.
  • Leukemia is a disease thought to occur when hematopoietic stem cells (or slightly differentiated cells) become tumors.
  • Chemotherapy is the basis of definitive treatment in leukemia.
  • chemotherapy requires a sufficient general state and organ function to withstand organ toxicity and complications.
  • elderly patients with leukemia who are unable to apply therapy and have no cure.
  • Differentiation induction therapy is known as a treatment for leukemia with few side effects.
  • differentiation-inducing therapy leukemia cells that continue to proliferate while remaining in the state of progenitor cells are killed by the normal life of normal leukocytes by differentiation and maturation, so leukemia cells such as many anticancer drugs and chemotherapy There are fewer side effects compared to treatments that directly destroy.
  • ATRA all-trans retinoic acid
  • arsenic as a differentiation-inducing agent for neutrophils.
  • no effective differentiation induction therapy has been established.
  • Acute promyelocytic leukemia is ATRA-resistant.
  • a new differentiation induction therapy effective for a wide range of leukemias is strongly desired in the medical field.
  • Aplastic anemia is a disease in which the differentiation process is inhibited by some abnormality in hematopoietic stem cells, and Costman syndrome (also known as severe congenital neutropenia) is found to inhibit the maturation of myeloid granulocyte cells.
  • Costman syndrome also known as severe congenital neutropenia
  • neutrophils decrease.
  • neutrophil differentiation inducers such as granulocyte colony stimulating factor (G-CSF) are currently administered. There are cases where it does not increase.
  • G-CSF granulocyte colony stimulating factor
  • An object of the present application is to provide a novel blood cell differentiation inducer.
  • albendazole a parasite-controlling agent
  • albendazole unexpectedly strongly induces differentiation of leukemia cells into monocyte-like cells
  • albendazole induces differentiation of normal progenitor cells into monocytes. That is, the present application provides a monocyte differentiation inducer containing albendazole.
  • the invention of the present application will be described in detail below.
  • [5] The agent for inducing differentiation of monocytes according to any one of [2] to [4], wherein the leukemia or a related disease thereof is chronic myelogenous leukemia, acute myeloid leukemia or myelodysplastic syndrome.
  • [7] A method for inducing differentiation into monocytes, comprising administering an effective amount of albendazole to a subject in need thereof. [8] The method according to [7], wherein the method is used in the treatment of a disease involving inhibition of the differentiation process of hematopoietic stem cells.
  • Albendazole for use in inducing monocyte differentiation.
  • Albendazole has a high differentiation-inducing activity toward monocytes and can be used as a monocyte differentiation-inducing agent. Furthermore, albendazole is already on the market as a therapeutic agent for hydatid disease (echinococcosis), and since data on safety to humans is accumulated, a safe monocyte differentiation inducer can be provided.
  • THP-1 cells of Test Example 3-1 were collected by treatment with 3 ⁇ M doxycycline for 48 hours, and cell surface expression of CD11b and CD14 was measured by flow cytometry.
  • Data are mean ⁇ SEM values. * P ⁇ 0.05, *** P ⁇ 0.001, two-sided student t test.
  • THP-1 cells treated with albendazole were measured by flow cytometry. Representative microscopic images of THP-1 cells of Test Example 4-3. Growth curves in the presence of 1 nMPMA of THP-1 cells transduced with shRNA targeting KLF4 (sh_KLF4 # 1 and # 2), DPYSL2A (sh_DPYSL2A # 1 and # 2) or control luciferase (sh_Luc.).
  • This application provides a monocyte differentiation inducer containing albendazole as an active ingredient.
  • the monocyte differentiation inducer of the present application can be used, for example, to treat a disease accompanied by inhibition of the differentiation process of hematopoietic stem cells.
  • the monocyte differentiation inducer of the present application can be used for patients with reduced neutrophils or monocytes regardless of the presence or absence of a disease accompanied by inhibition of the differentiation process of hematopoietic stem cells. It can be used to prevent or ameliorate symptoms that can be caused by a decrease in spheres or monocytes (eg, bacterial infections).
  • Albendazole [Chemical name: (methyl 5- (propylthio) -2-benzimidazolecarbamate)] Is known as a benzimidazole-type anthelmintic agent and has already been marketed as a therapeutic agent for encaustic diseases (eg, echinococcosis) (for example, Escazole (registered trademark) tablets in Japan). The action is thought to be due to the inhibition of tubulin binding to the colchicine binding site and the formation of microtubules.
  • encaustic diseases eg, echinococcosis
  • Escazole registered trademark
  • JP-T-2004-525140, JP-T-2008-522984, and JP-T-2016-535014 disclose solid cancers such as liver cancer, colon cancer, pancreatic cancer, ovarian cancer and the like using albendazole known as an anthelmintic agent. However, it does not describe a specific effect on leukemia. Moreover, these literatures do not describe induction of blood cell differentiation by albendazole.
  • albendazole may be synthesized by a known method, or may be obtained as a commercial product (for example, Tokyo Chemical Industry Co., Ltd.). Albendazole may also be in the form of a pharmaceutically acceptable salt or hydrate.
  • monocyte differentiation induction means a hematopoietic stem cell (strictly not a hematopoietic stem cell but an immature cell group that has the ability to differentiate into both myeloid and lymphoid cells (eg, c-kit surface antigen positive cells) And / or induce differentiation of myeloid hematopoietic progenitor cells into monocytes.
  • Monocytes differentiated by the monocyte differentiation inducer of the present application are at least one of the morphological features commonly used to identify monocytes (eg, notched or partially depressed) With nuclei; with basophil cytoplasmic granules (azurole granules); with vacuoles in the cytoplasm) and / or with features based on surface antigen classification (eg CD11b positive and CD14 positive) and self-proliferating ability And cells that do not have the ability to immortalize.
  • the gene information of the monocytes induced to differentiate is not particularly limited, and for example, gene information derived from a disease can be retained.
  • myeloid hematopoietic progenitor cell means a progenitor cell found in the process of differentiation from a hematopoietic stem cell into a myeloid blood cell, such as a myeloid hematopoietic stem cell and a mature differentiated cell from a myeloid hematopoietic stem cell. It means a progenitor cell in the process of differentiation into (for example, mast cell, basophil, eosinophil, neutrophil, monocyte, megakaryocyte, or erythrocyte).
  • myeloid hematopoietic progenitor cells include: myeloid hematopoietic stem cells, granule / monocytic stem cells, erythroblast / megakaryocytic stem cells, myeloblasts, promyelocytes, myelospheres, postmyelocytes, rods Examples thereof include nuclear spheres, segmented nucleus nuclei, monoblasts, pre-monocytes, pre-erythroblasts, erythroblasts, and pre-megakaryocytes.
  • Hematopoietic stem cells which are not strictly hematopoietic stem cells but include immature cells that have the ability to differentiate into both myeloid and lymphoid
  • myeloid progenitor cells are normal even if they are normal cells. (For example, canceration).
  • the monocyte differentiation inducer of the present application includes hematopoietic stem cells (including not strictly hematopoietic stem cells but immature cells and cells capable of differentiating into both myeloid and lymphoid systems) and / or myeloid progenitors.
  • hematopoietic stem cells including not strictly hematopoietic stem cells but immature cells and cells capable of differentiating into both myeloid and lymphoid systems
  • myeloid progenitors can treat diseases involving inhibition of the differentiation process of hematopoietic stem cells (eg, aplastic anemia, Costman syndrome, or leukemia or related diseases (eg myelodysplastic syndrome)) .
  • Aplastic anemia is susceptible to severe bacterial infections such as pneumonia and sepsis because the differentiation process is inhibited by some abnormality in hematopoietic stem cells, and blood cells in blood, particularly neutrophils, decrease.
  • Costman syndrome also known as severe congenital neutropenia
  • severe congenital neutropenia is a group of diseases in which severe chronic neutropenia is observed due to genetic mutation, and bacterial infections occur repeatedly.
  • Diagnosis by the bone marrow image shows inhibition of maturation of bone marrow granulocyte cells.
  • the monocyte differentiation inducer of the present application is a symptom (for example, bacterial infection) that is accompanied by a decrease in blood cells (particularly neutrophils) in addition to their fundamental treatment, for example.
  • a symptom for example, bacterial infection
  • blood cells particularly neutrophils
  • the functions of macrophages matured from monocytes to bacterial infection are similar to those of neutrophils.
  • symptoms such as bacterial infections can be treated. Can be done.
  • Leukemia is a disease thought to occur when hematopoietic stem cells (or slightly differentiated cells) become tumors.
  • Tumorized cells (leukemia cells) grow abnormally and occupy the bone marrow, and normal hematopoietic function is significantly inhibited.
  • red blood cells, normal white blood cells, and platelets decrease, and as the disease progresses, enlargement of the spleen, liver, and lymph nodes due to infiltration of leukemia cells begins to occur.
  • leukemia cells which are progenitor cells that have been differentiated to a certain stage, have proliferated (acute leukemia) and have the ability to differentiate and mature (chronic leukemia). It is divided roughly into. They are also classified into myeloid leukemia and lymphocytic leukemia depending on the cancerous cell lineage.
  • FAB classification based on morphological classification is known.
  • acute myeloid leukemia and acute lymphoblastic leukemia are roughly classified by peroxidase staining, and acute myeloid leukemia is further classified into the following M0 to M7.
  • the “leukemia” that can be treated with the monocyte differentiation inducer of the present application is not particularly limited as long as it is determined as leukemia based on general criteria used by doctors or researchers. Since the monocyte differentiation inducer of the present application has the ability to induce differentiation from myeloid hematopoietic progenitor cells to monocytes, it is preferably used for the treatment of acute and / or chronic myeloid leukemia, particularly preferably acute myeloid Used to treat leukemia.
  • the monocyte differentiation inducer of the present application can induce differentiation of abnormally proliferating leukemia cells into monocytes in the treatment of leukemia.
  • leukemia cells can be induced to differentiate into monocytes lacking the ability of self-proliferation and immortalization, and can be killed gently with the original short life span of normal monocytes. That is, the monocyte differentiation inducer of the present application can be used as differentiation induction therapy.
  • the monocyte differentiation inducer of the present application can gently kill leukemia cells, side effects (eg, anorexia, nausea, vomiting, diarrhea, etc.) that may occur with conventional chemotherapeutic agents with cell killing action Symptoms, fever, general malaise, sepsis, hair loss, acute respiratory distress syndrome, interstitial pneumonia, liver dysfunction, jaundice, arrhythmia, heart failure, gastrointestinal dysfunction, central nervous system disorder, liver abscess, acute pancreatitis, lung edema, Painful erythema, CRP elevation, ALT (GPT) elevation, AST (GOT) elevation, etc.) can be suppressed.
  • side effects eg, anorexia, nausea, vomiting, diarrhea, etc.
  • MDS myelodysplastic syndrome
  • the monocyte differentiation inducer of the present application can induce the differentiation of immature abnormal cells and gently kill them in the treatment of myelodysplastic syndromes as well as leukemia.
  • the monocyte differentiation inducer of the present application can improve the level of monocytes in the treatment of leukemia or related diseases, and prevent various symptoms (for example, bacterial infections etc.) caused by neutrophil / monocyte depletion. It can also be used for improvement.
  • treatment refers to treating disease, preventing disease, alleviating disease symptoms, delaying disease progression, suppressing disease symptoms, ameliorating disease symptoms, And inducing amelioration of disease symptoms.
  • the “effective amount” is the amount of the active agent necessary to give the patient the benefit of inducing differentiation of monocytes.
  • albendazole can be administered in combination with other drugs simultaneously, separately or sequentially.
  • albendazole and the other drug may be included in the same formulation or in separate formulations. When included in separate formulations, they may be administered simultaneously via the same or different routes of administration.
  • albendazole and the other agent may be administered separately according to different dosing schedules and may be administered by different routes of administration.
  • any agent may be administered first.
  • the time between administration of one therapeutic agent and the other is preferably less than 8 hours. More preferably, it is less than 4 hours, and even more preferably, it is less than 1 hour.
  • drugs for example, carmustine, chlorambucil, lomustine, mechloretamine, azacitidine, acyclovir, L-asparaginase, Arsenite, alemtuzumab, idarubicin, ifosfamide, ibritumomab tiuxetan, imatinib, irinotecan, interferon alpha, etoposide, enocitabine, epirubicin, erythropoietin, ofatatumumab, all-trans retinoic acid, carbogetum zobin, Mycin, thalidomide, cyclosporine, cyclophosphamide, cisplatin, cytarabine, zidobucin, sobuzoxane, daunorubicin, daca
  • drugs for example, carmustine, chlorambucil, lomustine, mechloretamine, azacitidine
  • the albendazole-containing monocyte differentiation inducer in the present application is mixed with one or more pharmaceutically acceptable carriers, for example, tablets, capsules, granules, powders, troches, syrups, emulsions, suspensions, etc. Or a parenteral preparation such as an external preparation, a suppository, an injection, an eye drop, a nasal preparation, a pulmonary preparation and the like.
  • pharmaceutically acceptable carriers for example, tablets, capsules, granules, powders, troches, syrups, emulsions, suspensions, etc.
  • a parenteral preparation such as an external preparation, a suppository, an injection, an eye drop, a nasal preparation, a pulmonary preparation and the like.
  • Examples of preferable dosage forms of the monocyte differentiation inducer according to the present application include tablets and injections.
  • the carrier / additive contained in the oral preparation of the present application is not particularly limited as long as it is a pharmaceutically acceptable one that is usually used.
  • Excipients such as lactose, sucrose, D-mannitol, D-sorbitol, corn starch, dextrin, microcrystalline cellulose, crystalline cellulose, carmellose, carmellose calcium, sodium carboxymethyl starch, low substituted hydroxypropylcellulose, and arabic Rubber);
  • Disintegrants eg, carmellose, carmellose calcium, carmellose sodium, carboxymethyl starch sodium, croscarmellose sodium, crospovidone, low substituted hydroxypropylcellulose, hydroxypropylmethylcellulose, and crystalline cellulose
  • Binders eg, hydroxypropylcellulose, hydroxypropylmethylcellulose, povidone, crystalline cellulose, sucrose, dextrin, starch, gelatin, carmellose sodium, and gum arabic
  • Fluidizing agents eg, light anhydrous silicic acid
  • the carrier / additive contained in the liquid preparation (including injection) of the present application is not particularly limited as long as it is a pharmaceutically acceptable one that is usually used.
  • Solvent eg, water, ethanol, propylene glycol, macrogol, glycerin, etc.
  • Solubilizers eg, propylene glycol, benzyl benzoate, ethanol, triethanolamine, sodium carbonate, sodium citrate, trometamol (tris [hydroxymethyl] aminomethane), meglumine, etc.
  • Isotonic agents eg, glucose, glycerin, D-mannitol, D-sorbitol, sodium chloride, sucrose, potassium chloride, etc.
  • Buffers eg trisodium phosphate, sodium hydrogen phosphate, sodium dihydrogen phosphate, potassium dihydrogen phosphate, boric acid, citric acid, sodium citrate, tartaric acid, acetic acid, sodium acetate, epsilon-aminocaproic acid
  • the amount of albendazole contained in the monocyte differentiation inducer according to the present application is not particularly limited and is selected in a wide range.
  • it is about 0.05% to about 90% by weight based on the composition.
  • it is preferably about 0.01 wt% to about 50 wt%, more preferably about 0.05 wt% to about 20 wt%, more preferably about 0.2 wt% to about 10 wt%.
  • a diluting solution eg, physiological saline
  • administered eg, intravenous administration
  • the dose of albendazole of the present application is appropriately selected depending on the administration method, the subject, the age of the subject, the degree of the disease, symptoms, dosage form, administration route, etc., for example, orally 0.1 mg per day It can be administered at a dose of ⁇ 100 g, preferably 1 mg to 50 g, more preferably 10 mg to 20 g, more preferably 100 mg to 10 g.
  • Examples of the lower limit of the daily dose of albendazole include 0.1 mg, 1 mg, 10 mg, 100 mg, and 400 mg, and examples of the upper limit include 700 mg, 1 g, 3 g, 5 g, 10 g, 20 g, 50 g and 100 g can be mentioned, and a preferable range of the daily dose of albendazole can be indicated by a combination of the upper limit value and the lower limit value.
  • the amount of albendazole per day may be administered in one to several divided doses.
  • the subject of administration of the monocyte differentiation inducer of the present application is not particularly limited, but is preferably a mammal (human and non-human mammals (eg, cows, horses, pigs, dogs, cats, mice, rats, rabbits, monkeys)). More preferably, it is a human.
  • a mammal human and non-human mammals (eg, cows, horses, pigs, dogs, cats, mice, rats, rabbits, monkeys)). More preferably, it is a human.
  • Kruppel-like transcription factor 4 is one of the KLF family transcription factors and is known to be suppressed in hematopoietic tumors (Non-patent Document 1).
  • Test Examples 1 to 3 below the present inventors have clarified that the signal pathway by KLF4 and its downstream factor DPYSL2A contributes to the differentiation from hematopoietic progenitor cells to monocytes ( Presented at American Society of Hematology in 2016: Non-Patent Document 2).
  • the inventors of the present application show that albendazole (ABZ) increases the expression of KLF4 and is highly active in inducing differentiation into monocytes through stimulation of DPYSL2A. It was revealed for the first time that the present invention was achieved.
  • the present invention will be described with reference to test examples, but the present invention is not limited to these test examples.
  • Acute myeloid leukemia (AML) -derived THP-1 and KG-1a cells were purchased from RIKEN biological resource center (BRC) (Japan).
  • BRC RIKEN biological resource center
  • SKNO-1 cells and fetal kidney-derived HEK293T cells were purchased from Japanese Collection of Research Bioresources (JCRB) (Japan).
  • AML-derived OCI-AML3 and MOLM-13 cells were purchased from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) (Germany).
  • DSMZ Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH
  • AML-derived MV4-11 and KG-1a cells were purchased from American Type Culture Collection (ATCC) (USA).
  • NB4 and UF-1 cells derived from acute promyelocytic leukemia (APL) were provided by Dr. Y.
  • HEK293T cells were cultured in Dulbecco's Modified Eagle Medium (DMEM) (supplemented with 10% heat-inactivated fetal calf serum (FBS) and 1% penicillin-streptomycin (PS)) in a humidified environment of 5% CO 2 and 95% air, 37 Maintained in an incubator at 0 ° C. Other cells were cultured at 37 ° C. in 5% CO 2 and 95% air in Roswell Park Memorial Institute (RPMI) 1640 medium (containing 10% FBS and 1% PS).
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS heat-inactivated fetal calf serum
  • PS penicillin-streptomycin
  • MOLM-13 is a leukemia that developed from myelodysplastic syndrome (MDS).
  • MDS myelodysplastic syndrome
  • NB4 and UF-1 are all-trans retinoic acid (ATRA) resistant.
  • ⁇ IC 50 evaluation> For cell viability analysis, cells were seeded at a concentration of 1 ⁇ 10 5 cells / mL. A predetermined concentration of drug was added to the culture medium, and the cells were cultured for 48 hours. Followinged by WST assay Cell viability, Cell Count Reagent (nacalai tesque, Inc.) and Infinite was evaluated using as described in (TM) 200 PRO multimode reader the (TECAN) manufacturer. Depict percent inhibition curve, an IC 50 value of the drug median-effect method (Chou TC, Talalay P. Quantitative analysis of dose-effect relationships:.. The combined effects of multiple drugs or enzyme inhibitors Adv Enzyme Regul 1984; 22 : 27-55).
  • the primers used for qRT-PCR are shown in the table below.
  • CSIV-TRE-Ubc-KT Human KLF4, DPYSL2A and DPYSL2B cDNAs were amplified by PCR and then inserted into pENTR1A dual selection vector (Thermo Fisher Scientific), CSIV-TRE-Ubc-KT, and pLenti CMV Puro DEST (addgene) expression vectors.
  • CSIV-TRE-Ubc-KT was provided by Dr. H. Miyoshi (RIKEN, BRC, Japan). All PCR products were confirmed by DNA sequencing.
  • Lentivirus production is performed as described in Morita K, Suzuki K, Maeda S, et al. Genetic regulation of the RUNX transcription factor family has antitumor effects. J Clin Invest. 2017; 127 (7): 2815-2828. It was. Briefly, HEK293T cells were transiently co-transfected with polyethyleneimine (PEI, Sigma-Aldrich) with lentiviral vectors (eg psPAX2 and pMD2.G).
  • PEI polyethyleneimine
  • lentiviral vectors eg psPAX2 and pMD2.G
  • the membrane was treated with the following primary antibodies: anti-KLF4 (# 4038, Cell Signaling Technology), anti-GAPDH (FL-335, Santa Cruz Biotechnology, Inc.), anti-DPYSL2 (HPA002381, Sigma-Aldrich), anti-DPYSL antibody (HPA002381, Sigma) -Aldrich) and HRP-conjugated anti-rabbit IgG and anti-mouse IgG (Cell Signaling Technology) were used as secondary antibodies. Blots were visualized using Chemi-Lumi One Super (nacalai tesque, Inc.) and ChemiDoc TM XRS + Imager (Bio-Rad Laboratories, Inc.) according to the manufacturer's recommendations. Protein levels were quantified using Image Lab Software (Bio-Rad Laboratories, Inc.).
  • ⁇ Statistics> The statistical significance of the difference between the control group and the test group was evaluated by a two-sided unpaired student t test, and was considered significant when the p value was less than 0.05. Equal variance in the two populations was calculated by F test. Results were expressed as mean ⁇ SEM values obtained from three independent tests. In the transplantation experiment, the animals were randomly divided into test groups, and the treatment was performed in a blinded manner. The survival rate of each group was compared using a log-rank test.
  • NOD / Shi-scid, IL-2R ⁇ KO (NOG) mice were purchased from Central Institute for Experimental Animals (Japan). Litters were used as controls for all studies.
  • ⁇ Xenograft mouse model A xenograft mouse model of a human cancer cell line was created using NOG mice.
  • THP-1 cells (2 ⁇ 10 6 cells / mouse) were administered intravenously.
  • Peripheral blood (PB) was collected weekly and chimerization was checked with anti-human CD45 antibody (BD Biosciences) by flow cytometer.
  • PB Peripheral blood
  • BD Biosciences anti-human CD45 antibody
  • mice received albendazole (50 mg / kg body weight, 2 times / week, oral (po)), or an equivalent amount of dimethyl sulfoxide (DMSO).
  • albendazole 50 mg / kg body weight, 2 times / week, oral (po)
  • DMSO dimethyl sulfoxide
  • THP-1 cells transduced with KLF4 or control encoding lentivirus were treated with 3 ⁇ M doxycycline for 48 hours, then harvested and cytospun onto glass slides. Each slide was subjected to Diff-Quik staining (modified Giemsa staining).
  • Diff-Quik staining modified Giemsa staining.
  • FIG. 1 A representative microscopic image of THP-1 cells is shown in FIG. As shown in FIG. 1, when KLF4 was overexpressed in the AML cell line, abnormal cells were induced to differentiate into monocyte-like cells.
  • DPYSL2A is a direct downstream factor of KLF4.
  • THP-1 and MOLM-13 cells transformed with lentivirus encoding KLF4 or control (Ctrl) were treated with 3 ⁇ M doxycycline for 48 hours, lysed for protein extraction, and immunoblotting of KLF4, DPYSL2A, and GAPDH Went. The results are shown in FIG.
  • KLF4 up-regulated DPYSL2A gene expression.
  • DPYSL2A is an important mediator of KLF4-induced differentiation.
  • Test Example 3-1 THP-1 cells transduced with a lentivirus encoding shRNA targeting KLF4 and DPYSL2A (sh_DPYSL2A # 1 and # 2) or control luciferase (sh_Luc.) Were treated with 3 ⁇ M doxycycline for 48 hours And then lysed for protein extraction and immunoblotted for KLF4, DPYSL2A and GAPDH. The results are shown in Fig. 3-1.
  • Test Example 3-2 THP-1 cells of Test Example 3-1 were collected by treatment with 3 ⁇ M doxycycline for 48 hours, and cell surface expression of CD11b and CD14 was measured by flow cytometry. The results are shown in Fig. 3-2.
  • Test Example 3-4 A representative microscopic image of the THP-1 cell of Test Example 3-1 is shown in FIG. 3-4. Cells were collected by treatment with 3 ⁇ M doxycycline for 48 hours and cytospun onto glass slides. Each slide was subjected to Diff-Quik staining (modified Giemsa staining).
  • THP-1 cells expressing KLF4 and DPYSL2A differentiated into monocyte-like cells, and after 5 days of culture, the cells were significantly different from those in which DPYSL2A was knocked down. The number has decreased.
  • Test Example 4-2 Cell surface expression of CD11b and CD14 in THP-1 cells treated with albendazole was measured by flow cytometry. Cells were treated with albendazole (1 ⁇ M) for 24 hours and harvested for flow cytometric analysis. The results are shown in Fig. 4-2.
  • Test Example 4-3 Representative microscopic image of THP-1 cells of Test Example 4-2. Diff-Quik staining (modified Giemsa staining) was performed on each slide. (Enlargement 20 ⁇ , scale bar: 50 ⁇ m). The results are shown in Fig. 4-3.
  • Test Example 4-4 Presence of 1 nMPMA in THP-1 cells transduced with shRNA targeting KLF4 (sh_KLF4 # 1 and # 2), DPYSL2A (sh_DPYSL2A # 1 and # 2) or control luciferase (sh_Luc.) The growth curve below. shRNA expression was induced with 3 ⁇ M doxycycline. The results are shown in Fig. 4-4.
  • albendazole increased the expression of KLF4 and DPYSL2A and differentiated THP-1 cells into monocyte-like cells.
  • FIG. 4-4 since the antitumor effect of albendazole was suppressed in cells knocked down with KLF4 or DPYSL2A, the expression of KLF4 by albendazole shows that such an antitumor effect appears. It was done.
  • the table below shows the IC 50 of ATRA and / or albendazole for each cell.
  • Albendazole showed a very low IC 50 for each leukemia cell.
  • IC 50 evaluation-II AML cell lines were treated with various concentrations of albendazole for 48 hours and IC 50 calculated. The results are shown in the table below. Albendazole showed a very low IC 50 for a wide variety of AML cells.
  • the albendazole and DMSO groups were treated with albendazole (50 mg / kg body weight, 2 times / week, po) or an equivalent amount of DMSO (control).
  • overall survival of the albendazole group was significantly prolonged compared to the DMSO group.
  • Test Example 7-2 Mice treated as in Test Example 7-1 were appropriately anesthetized and sacrificed. Bone marrow tissue was collected and hematoxylin and eosin (H & E) staining and immunohistochemical staining with anti-human CD45 antibody were performed on each slide. A representative image of the spleen and liver of an AML (THP-1 cell) xenograft mouse (30 days after transplantation) is shown in FIG. 6-3. As shown in FIG. 6-2, the proliferation of THP-1 cells was suppressed. As shown in FIG. 6-3, albendazole suppressed the enlargement of the liver and spleen.
  • Test Example 8 From the bone marrow cells of wild-type C57BL / 6 mice, c-Kit surface antigen was stained and positive cells (this cell is considered to be an immature cell that has hardly differentiated, and to both the myeloid and lymphatic systems. Were collected using a flow cytometer. 5000 collected c-kit surface antigen positive cells were seeded on a methylcellulose semi-solid medium, and albendazole was administered thereto at 0 nM, 100 nM, and 500 nM, and cultured at 37 ° C. for 1 week. One week later, colony formation was observed with a microscope and counted. The results are shown in FIG. As apparent from FIG. 7, the number of monocytic colonies increased from that of granulocyte colonies.

Abstract

Provided is a monocyte differentiation inducer which contains albendazole as an active ingredient.

Description

アルベンダゾール含有単球分化誘導剤Albendazole-containing monocyte differentiation inducer
 本願は、アルベンダゾールを有効成分とする単球分化誘導剤に関する。 This application relates to a monocyte differentiation inducer containing albendazole as an active ingredient.
 血液疾患の中には、造血幹細胞の分化過程が阻害されることに起因する疾患が存在し、例えば白血病、再生不良性貧血、コストマン症候群挙げられる。 Among blood diseases, there are diseases caused by inhibition of the differentiation process of hematopoietic stem cells, and examples include leukemia, aplastic anemia, and Costman syndrome.
 白血病は、造血幹細胞(あるいはやや分化した細胞)が腫瘍化することによって発生すると考えられている疾患である。白血病における根治治療の基本は化学療法であるが、化学療法を行うには臓器毒性や合併症に耐えられる全身状態や臓器機能が充分に保たれている必要があるため、高齢の患者においては化学療法を適用できない場合も多く、何ら治癒の手立てがない高齢白血病患者が存在する。このような患者のためにも副作用の少ない新たな治療剤が必要とされている。 Leukemia is a disease thought to occur when hematopoietic stem cells (or slightly differentiated cells) become tumors. Chemotherapy is the basis of definitive treatment in leukemia. However, chemotherapy requires a sufficient general state and organ function to withstand organ toxicity and complications. There are many cases where elderly patients with leukemia who are unable to apply therapy and have no cure. There is a need for new therapeutic agents with few side effects for such patients.
 副作用の少ない白血病に対する治療法として分化誘導療法が知られている。分化誘導療法では、前駆細胞の状態に留まったまま増殖を続ける白血病細胞を分化・成熟させることによって正常白血球の本来の寿命で死滅させるため、多くの抗がん剤や化学療法のような白血病細胞を直接破壊する治療法と比較して副作用は少ない。しかしながら、急性前骨髄球性白血病ではオールトランス型レチノイン酸(ATRA)および/またはヒ素を好中球への分化誘導剤として使用する分化誘導療法の有効性が確立されているが、他の白血病には現在のところ有効な分化誘導療法は確立していない。また、急性前骨髄球性白血病にはATRA耐性のもの存在する。広範囲な白血病に有効な新たな分化誘導療法が医療現場において強く望まれている。 ”Differentiation induction therapy is known as a treatment for leukemia with few side effects. In differentiation-inducing therapy, leukemia cells that continue to proliferate while remaining in the state of progenitor cells are killed by the normal life of normal leukocytes by differentiation and maturation, so leukemia cells such as many anticancer drugs and chemotherapy There are fewer side effects compared to treatments that directly destroy However, in acute promyelocytic leukemia, the effectiveness of differentiation-inducing therapy using all-trans retinoic acid (ATRA) and / or arsenic as a differentiation-inducing agent for neutrophils has been established. Currently, no effective differentiation induction therapy has been established. Acute promyelocytic leukemia is ATRA-resistant. A new differentiation induction therapy effective for a wide range of leukemias is strongly desired in the medical field.
 再生不良性貧血は造血幹細胞における何らかの異常により分化過程が阻害される疾患であり、コストマン症候群(別名:重症先天性好中球減少症)は骨髄顆粒球系細胞の成熟阻害が認められ、好中球が減少する疾患群である。
 白血病、再生不良性貧血、コストマン症候群のような造血幹細胞の分化過程が阻害されることに起因する疾患では、造血機能が低下するため、好中球や単球が減少して細胞感染症が問題となる。これらの疾患における細菌感染症の対処としては、現在のところ、顆粒球コロニー刺激因子(G-CSF)等の好中球分化誘導剤が投与されるが、G-CSF投与後も好中球が増加しない場合も存在する。これらの疾患における症状を改善するために、造血幹細胞からの分化過程を促進する有効な血球系分化誘導剤が望まれている。
Aplastic anemia is a disease in which the differentiation process is inhibited by some abnormality in hematopoietic stem cells, and Costman syndrome (also known as severe congenital neutropenia) is found to inhibit the maturation of myeloid granulocyte cells. A group of diseases in which neutrophils decrease.
In diseases caused by inhibition of the differentiation process of hematopoietic stem cells, such as leukemia, aplastic anemia, and Costman syndrome, hematopoietic function is reduced, resulting in decreased neutrophils and monocytes and cell infection. It becomes a problem. As a countermeasure against bacterial infections in these diseases, neutrophil differentiation inducers such as granulocyte colony stimulating factor (G-CSF) are currently administered. There are cases where it does not increase. In order to improve the symptoms in these diseases, an effective hematopoietic differentiation inducer that promotes the differentiation process from hematopoietic stem cells is desired.
 本明細書において引用する先行技術文献の開示は全て、参照することにより、本明細書に組み込まれる。 All disclosures of prior art documents cited in this specification are incorporated herein by reference.
 本願の課題は、新規な血球系細胞の分化誘導剤を提供することである。 An object of the present application is to provide a novel blood cell differentiation inducer.
 本発明者等は、上記課題を解決すべく鋭意検討を行ったところ、寄生虫の駆除剤であるアルベンダゾールが、意外にも白血病細胞を単球様細胞に強力に分化誘導することを見出し、本願に至った。さらに意外なことにアルベンダゾールは正常血球系前駆細胞についてもこれを単球に分化誘導することを見出し本願に至った。すなわち、本願はアルベンダゾールを含む単球分化誘導剤を提供する。
 以下に本願の発明を詳説する。
As a result of diligent studies to solve the above-mentioned problems, the present inventors have found that albendazole, a parasite-controlling agent, unexpectedly strongly induces differentiation of leukemia cells into monocyte-like cells, It came to this application. Surprisingly, it has been found that albendazole induces differentiation of normal progenitor cells into monocytes. That is, the present application provides a monocyte differentiation inducer containing albendazole.
The invention of the present application will be described in detail below.
[1]アルベンダゾールを有効成分として含む、単球分化誘導剤。
[2]造血幹細胞の分化過程の阻害を伴う疾患を治療するための、[1]に記載の単球分化誘導剤。
[3]該造血幹細胞の分化過程の阻害を伴う疾患が、再生不良性貧血、コストマン症候群、または白血病もしくはその類縁疾患から選択される、[2]に記載の単球分化誘導剤。
[4]該造血幹細胞の分化過程の阻害を伴う疾患が、白血病またはその類縁疾患である、[2]または[3]に記載の単球分化誘導剤。
[5]該白血病またはその類縁疾患が、慢性骨髄性白血病、急性骨髄性白血病または骨髄異形成症候群である、[2]~[4]のいずれか1項に記載の単球分化誘導剤。
[6]分化誘導療法として使用される、[4]または[5]に記載の単球分化誘導剤。
[1] A monocyte differentiation inducer containing albendazole as an active ingredient.
[2] The agent for inducing differentiation of monocytes according to [1] for treating a disease accompanied by inhibition of a differentiation process of hematopoietic stem cells.
[3] The agent for inducing differentiation of monocytes according to [2], wherein the disease accompanied by inhibition of the differentiation process of hematopoietic stem cells is selected from aplastic anemia, Costman syndrome, or leukemia or related diseases.
[4] The monocyte differentiation inducer according to [2] or [3], wherein the disease accompanied by inhibition of the differentiation process of the hematopoietic stem cells is leukemia or a related disease thereof.
[5] The agent for inducing differentiation of monocytes according to any one of [2] to [4], wherein the leukemia or a related disease thereof is chronic myelogenous leukemia, acute myeloid leukemia or myelodysplastic syndrome.
[6] The monocyte differentiation inducer according to [4] or [5], which is used as a differentiation induction therapy.
[7]単球への分化を誘導する方法であって、その必要がある対象に有効量のアルベンダゾールを投与することを含む方法。
[8]該方法が造血幹細胞の分化過程の阻害を伴う疾患の治療において用いられる、[7]に記載の方法。
[7] A method for inducing differentiation into monocytes, comprising administering an effective amount of albendazole to a subject in need thereof.
[8] The method according to [7], wherein the method is used in the treatment of a disease involving inhibition of the differentiation process of hematopoietic stem cells.
[9]単球分化誘導剤を製造するための、アルベンダゾールの使用。
[10]該単球分化誘導剤が造血幹細胞の分化過程の阻害を伴う疾患を治療するために使用される、[9]に記載の使用。
[9] Use of albendazole for producing a monocyte differentiation inducer.
[10] The use according to [9], wherein the agent for inducing differentiation of monocytes is used for treating a disease accompanied by inhibition of a differentiation process of hematopoietic stem cells.
[11]単球分化誘導における使用のための、アルベンダゾール。
[12]造血幹細胞の分化過程の阻害を伴う疾患の治療における単球分化誘導における使用のための、[11]に記載のアルベンダゾール。
[11] Albendazole for use in inducing monocyte differentiation.
[12] The albendazole according to [11] for use in inducing monocyte differentiation in the treatment of a disease accompanied by inhibition of the differentiation process of hematopoietic stem cells.
 アルベンダゾールは単球への高い分化誘導活性を有し、単球分化誘導剤として使用できる。さらにアルベンダゾールは、包虫症(エキノコックス症)治療薬として既に市販されており、ヒトへの安全性についてもデータが蓄積されているため、安全な単球分化誘導剤を提供できる。 Albendazole has a high differentiation-inducing activity toward monocytes and can be used as a monocyte differentiation-inducing agent. Furthermore, albendazole is already on the market as a therapeutic agent for hydatid disease (echinococcosis), and since data on safety to humans is accumulated, a safe monocyte differentiation inducer can be provided.
KLF4またはコントロールをコードするレンチウイルスで形質導入されたTHP-1細胞の代表的な顕微鏡画像(Diff-Quik染色)(拡大20×)。Representative microscopic image of THP-1 cells transduced with KLF4 or control encoding lentivirus (Diff-Quik staining) (magnification 20 ×). KLF4またはコントロールをコードするレンチウイルスで形質転換されたTHP-1およびMOLM-13細胞中のKLF4、DPYSL2A(DPYSL2A)およびGAPDHの免疫ブロット。Immunoblots of KLF4, DPYSL2A (DPYSL2A) and GAPDH in THP-1 and MOLM-13 cells transformed with KLF4 or lentivirus encoding control. KLF4およびDPYSL2A(sh_DPYSL2A #1および#2)またはコントロールルシフェラーゼ(sh_Luc.)を標的にするshRNAをコードするレンチウイルスで形質導入されたTHP-1細胞におけるKLF4、DPYSL2AおよびGAPDHの免疫ブロット。Immunoblots of KLF4, DPYSL2A and GAPDH in THP-1 cells transduced with lentivirus encoding shRNA targeting KLF4 and DPYSL2A (sh_DPYSL2A # 1 and # 2) or control luciferase (sh_Luc.). 試験例3-1のTHP-1細胞を3μMドキシサイクリンで48時間処理して収集し、CD11bおよびCD14の細胞表面発現をフローサイトメトリーで測定した。THP-1 cells of Test Example 3-1 were collected by treatment with 3 μM doxycycline for 48 hours, and cell surface expression of CD11b and CD14 was measured by flow cytometry. 試験例3-1のTHP-1細胞の細胞増殖曲線。データは平均値±SEM値である。*P<0.05、***P<0.001、両側スチューデントt検定による。The cell growth curve of the THP-1 cell of Test Example 3-1. Data are mean ± SEM values. * P <0.05, *** P <0.001, two-sided student t test. 試験例3-1のTHP-1細胞の代表的な顕微鏡画像。(拡大20×、スケールバー:50μm)A representative microscopic image of THP-1 cells of Test Example 3-1. (Enlarged 20 ×, scale bar: 50 μm) アルベンダゾールで処理されたTHP-1細胞におけるKLF4およびDPYSL2Aの相対的な発現。左:処理前。右:処理後。データは平均値±SEM値である。**P<0.01、***P<0.001、両側スチューデントt検定による。Relative expression of KLF4 and DPYSL2A in THP-1 cells treated with albendazole. Left: Before processing. Right: After processing. Data are mean ± SEM values. ** P <0.01, *** P <0.001, two-sided Student t test. アルベンダゾールで処理されたTHP-1細胞におけるCD11bおよびCD14の細胞表面発現をフローサイトメトリーで測定した。Cell surface expression of CD11b and CD14 in THP-1 cells treated with albendazole was measured by flow cytometry. 試験例4-3のTHP-1細胞の代表的な顕微鏡画像。Representative microscopic images of THP-1 cells of Test Example 4-3. KLF4(sh_KLF4#1および#2)、DPYSL2A(sh_DPYSL2A#1および#2)またはコントロールルシフェラーゼ(sh_Luc.)を標的にするshRNAで形質導入されたTHP-1細胞の1nMPMAの存在下における成長曲線。Growth curves in the presence of 1 nMPMA of THP-1 cells transduced with shRNA targeting KLF4 (sh_KLF4 # 1 and # 2), DPYSL2A (sh_DPYSL2A # 1 and # 2) or control luciferase (sh_Luc.). THP-1およびMOLM-13細胞におけるアルベンダゾールの用量反応曲線。Albendazole dose response curves in THP-1 and MOLM-13 cells. ATRA耐性UF-1細胞におけるアルベンダゾールの用量反応曲線。Albendazole dose response curve in ATRA resistant UF-1 cells. ATRA耐性NB4細胞におけるアルベンダゾールの用量反応曲線。Albendazole dose response curve in ATRA resistant NB4 cells. THP-1細胞を移植し、アルベンダゾールまたはコントロールで処理したNOGマウスの全生存。P<0.001、log-rank(Mantel-Cox)検定。Overall survival of NOG mice transplanted with THP-1 cells and treated with albendazole or control. P <0.001, log-rank (Mantel-Cox) test. AML(THP-1細胞)異種移植マウスの骨髄の代表的な顕微鏡画像(移植後30日)。(拡大4×および20×、スケールバー100μm)。Representative microscopic image of bone marrow of AML (THP-1 cell) xenograft mice (30 days after transplantation). (Magnification 4 × and 20 ×, scale bar 100 μm). (D)(C)のAML(THP-1細胞)異種移植マウス(移植後30日)の脾臓および肝臓の代表的な画像。(D) Representative images of spleen and liver of AML (THP-1 cells) xenograft mice (30 days after transplant) in (C). 野生型C57BL/6マウスの骨髄から採取したc-kit表面抗原陽性細胞にアルベンダゾールを0nM、100nM、500nMで投与し、1週間後にコロニー形成を確認した。縦軸は個/ディッシュを示す。Albendazole was administered at 0 nM, 100 nM, and 500 nM to c-kit surface antigen positive cells collected from bone marrow of wild type C57BL / 6 mice, and colony formation was confirmed after 1 week. The vertical axis represents pieces / dish.
 本願はアルベンダゾールを有効成分として含む単球分化誘導剤を提供する。本願の単球分化誘導剤は、例えば、造血幹細胞の分化過程の阻害を伴う疾患を治療するために使用されうる。また、本願の単球分化誘導剤は、造血幹細胞の分化過程の阻害を伴う疾患の有無に関わらず、好中球または単球の減少が生じている患者に対しても使用され得、好中球または単球の減少により引き起こされうる症状(例えば細菌感染症)の予防または改善に使用されうる。 This application provides a monocyte differentiation inducer containing albendazole as an active ingredient. The monocyte differentiation inducer of the present application can be used, for example, to treat a disease accompanied by inhibition of the differentiation process of hematopoietic stem cells. In addition, the monocyte differentiation inducer of the present application can be used for patients with reduced neutrophils or monocytes regardless of the presence or absence of a disease accompanied by inhibition of the differentiation process of hematopoietic stem cells. It can be used to prevent or ameliorate symptoms that can be caused by a decrease in spheres or monocytes (eg, bacterial infections).
 アルベンダゾール[化学名:(methyl 5-(propylthio)-2-benzimidazolecarbamate)]
はベンズイミダゾール系の駆虫剤として知られ、包虫症(エキノコックス症)の治療薬(例えば日本ではエスカゾール(登録商標)錠)として既に上市されている。その作用はチューブリンのコルヒチン結合部位への結合を阻害し、微小管の形成を阻害することに起因すると考えられている。
Albendazole [Chemical name: (methyl 5- (propylthio) -2-benzimidazolecarbamate)]
Is known as a benzimidazole-type anthelmintic agent and has already been marketed as a therapeutic agent for encaustic diseases (eg, echinococcosis) (for example, Escazole (registered trademark) tablets in Japan). The action is thought to be due to the inhibition of tubulin binding to the colchicine binding site and the formation of microtubules.
 特表2004-525140号公報、特表2008-522984号公報および特表2016-535014号公報には、駆虫剤として知られるアルベンダゾールを用いた肝癌、結腸癌、膵臓癌、卵巣癌等の固形癌の治療方法について記載されているが、白血病に対しての具体的な効果については記載されていない。また、これらの文献では、アルベンダゾールによる血球系細胞の分化誘導については記載されていない。 JP-T-2004-525140, JP-T-2008-522984, and JP-T-2016-535014 disclose solid cancers such as liver cancer, colon cancer, pancreatic cancer, ovarian cancer and the like using albendazole known as an anthelmintic agent. However, it does not describe a specific effect on leukemia. Moreover, these literatures do not describe induction of blood cell differentiation by albendazole.
 本願においてアルベンダゾールは公知の方法により合成してもよく、市販品(例えば東京化成工業株式会社)として入手することもできる。また、アルベンダゾールは医薬的に許容される塩または水和物の形態であっても良い。 In the present application, albendazole may be synthesized by a known method, or may be obtained as a commercial product (for example, Tokyo Chemical Industry Co., Ltd.). Albendazole may also be in the form of a pharmaceutically acceptable salt or hydrate.
 本願において「単球分化誘導」とは、造血幹細胞(厳密には造血幹細胞ではないが未熟な細胞群で骨髄系・リンパ系両方への分化能を有する細胞(例えばc-kit表面抗原陽性細胞)を含む)、および/または骨髄系の血球系前駆細胞の単球への分化を誘導することを意味する。本願の単球分化誘導剤により分化誘導された単球は、単球を特定するために一般的に使用される形態学的特徴の少なくとも1つ(例えば、切れ込みがあったり部分的に陥凹した核を持つ;好塩基性の細胞質顆粒を持つ(アズール顆粒);細胞質に空胞を持つ)および/または表面抗原分類に基づく特徴(例えばCD11b陽性かつCD14陽性)を有し、かつ、自己増殖能および不死化能を有さない細胞を意味する。該分化誘導された単球の遺伝子情報は特に限定されず、例えば疾患由来の遺伝子情報を保有し得る。 In the present application, “monocyte differentiation induction” means a hematopoietic stem cell (strictly not a hematopoietic stem cell but an immature cell group that has the ability to differentiate into both myeloid and lymphoid cells (eg, c-kit surface antigen positive cells) And / or induce differentiation of myeloid hematopoietic progenitor cells into monocytes. Monocytes differentiated by the monocyte differentiation inducer of the present application are at least one of the morphological features commonly used to identify monocytes (eg, notched or partially depressed) With nuclei; with basophil cytoplasmic granules (azurole granules); with vacuoles in the cytoplasm) and / or with features based on surface antigen classification (eg CD11b positive and CD14 positive) and self-proliferating ability And cells that do not have the ability to immortalize. The gene information of the monocytes induced to differentiate is not particularly limited, and for example, gene information derived from a disease can be retained.
 本願において「骨髄系の血球系前駆細胞」とは、造血幹細胞から骨髄系の血球への分化の過程に見られる前駆細胞を意味し、例えば骨髄系造血幹細胞、および骨髄系造血幹細胞から成熟分化細胞(例えば、肥満細胞、好塩基球、好酸球、好中球、単球、巨核球、または赤血球)に分化する過程の前駆細胞を意味する。骨髄系の血球系前駆細胞の例として、骨髄系造血幹細胞、顆粒系/単球系幹細胞、赤芽球系/巨核球系幹細胞、骨髄芽球、前骨髄球、骨髄球、後骨髄球、桿状核球、分葉核球、単芽球、前単球、前赤芽球、赤芽球、前巨核球等が挙げられる。造血幹細胞(厳密には造血幹細胞ではないが未熟な細胞群で骨髄系・リンパ系両方への分化能を有する細胞を含む)および骨髄系の血球系前駆細胞は、正常細胞であっても何らかの異変(例えばガン化)を有していてもよい。 In the present application, “myeloid hematopoietic progenitor cell” means a progenitor cell found in the process of differentiation from a hematopoietic stem cell into a myeloid blood cell, such as a myeloid hematopoietic stem cell and a mature differentiated cell from a myeloid hematopoietic stem cell. It means a progenitor cell in the process of differentiation into (for example, mast cell, basophil, eosinophil, neutrophil, monocyte, megakaryocyte, or erythrocyte). Examples of myeloid hematopoietic progenitor cells include: myeloid hematopoietic stem cells, granule / monocytic stem cells, erythroblast / megakaryocytic stem cells, myeloblasts, promyelocytes, myelospheres, postmyelocytes, rods Examples thereof include nuclear spheres, segmented nucleus nuclei, monoblasts, pre-monocytes, pre-erythroblasts, erythroblasts, and pre-megakaryocytes. Hematopoietic stem cells (which are not strictly hematopoietic stem cells but include immature cells that have the ability to differentiate into both myeloid and lymphoid) and myeloid progenitor cells are normal even if they are normal cells. (For example, canceration).
 本願の単球分化誘導剤は、造血幹細胞(厳密には造血幹細胞ではないが未熟な細胞群で骨髄系・リンパ系両方への分化能を有する細胞を含む)および/または骨髄系の血球系前駆細胞を単球へ分化誘導することによって、造血幹細胞の分化過程の阻害を伴う疾患(例えば再生不良性貧血、コストマン症候群、または白血病もしくはその類縁疾患(例えば骨髄異形成症候群))を治療しうる。 The monocyte differentiation inducer of the present application includes hematopoietic stem cells (including not strictly hematopoietic stem cells but immature cells and cells capable of differentiating into both myeloid and lymphoid systems) and / or myeloid progenitors. Inducing differentiation of cells into monocytes can treat diseases involving inhibition of the differentiation process of hematopoietic stem cells (eg, aplastic anemia, Costman syndrome, or leukemia or related diseases (eg myelodysplastic syndrome)) .
 再生不良性貧血は造血幹細胞における何らかの異常により分化過程が阻害され、血液中の血球、特に好中球が減少するため、肺炎や敗血症のような重症の細菌感染症を引き起こし易い。 Aplastic anemia is susceptible to severe bacterial infections such as pneumonia and sepsis because the differentiation process is inhibited by some abnormality in hematopoietic stem cells, and blood cells in blood, particularly neutrophils, decrease.
 コストマン症候群(別名:重症先天性好中球減少症)は遺伝子変異によって重症慢性好中球減少が認められる疾患群であり、細菌感染症が反復して起こる。その骨髄像による診断では骨髄顆粒球系細胞の成熟阻害が認められる。 Costman syndrome (also known as severe congenital neutropenia) is a group of diseases in which severe chronic neutropenia is observed due to genetic mutation, and bacterial infections occur repeatedly. Diagnosis by the bone marrow image shows inhibition of maturation of bone marrow granulocyte cells.
 本願の単球分化誘導剤は、再生不良性貧血またはコストマン症候群の治療においては、例えば、それらの根本的治療に加え、血球(特に好中球)減少により併発する症状(例えば細菌感染症)を治療するために使用されうる。単球からさらに成熟したマクロファージの細菌感染に対する機能は好中球のそれと類似しており、本願の単球分化誘導剤により単球への分化を誘導することで、細菌感染症等の症状が治療されうる。 In the treatment of aplastic anemia or costman syndrome, the monocyte differentiation inducer of the present application is a symptom (for example, bacterial infection) that is accompanied by a decrease in blood cells (particularly neutrophils) in addition to their fundamental treatment, for example. Can be used to treat. The functions of macrophages matured from monocytes to bacterial infection are similar to those of neutrophils. By inducing differentiation into monocytes using the monocyte differentiation inducer of the present application, symptoms such as bacterial infections can be treated. Can be done.
 白血病は造血幹細胞(あるいはやや分化した細胞)が腫瘍化することによって発生すると考えられている疾患である。腫瘍化した細胞(白血病細胞)が異常増殖して骨髄を占拠し、正常な造血機能は著しく阻害される。その結果、赤血球、正常白血球および血小板が減少し、病状が進行すると白血病細胞が浸潤することによる脾臓、肝臓およびリンパ節の腫大がみられるようになる。 Leukemia is a disease thought to occur when hematopoietic stem cells (or slightly differentiated cells) become tumors. Tumorized cells (leukemia cells) grow abnormally and occupy the bone marrow, and normal hematopoietic function is significantly inhibited. As a result, red blood cells, normal white blood cells, and platelets decrease, and as the disease progresses, enlargement of the spleen, liver, and lymph nodes due to infiltration of leukemia cells begins to occur.
 白血病は、分化が一定の段階まで進んで停止した血球系前駆細胞の白血病細胞が増殖している場合(急性白血病)と、白血病細胞が分化・成熟する能力を保持している場合(慢性白血病)に大別される。また、がん化している細胞系列によって、骨髄性白血病とリンパ性白血病に分類される。 In leukemia, leukemia cells, which are progenitor cells that have been differentiated to a certain stage, have proliferated (acute leukemia) and have the ability to differentiate and mature (chronic leukemia). It is divided roughly into. They are also classified into myeloid leukemia and lymphocytic leukemia depending on the cancerous cell lineage.
 急性白血病の分類の1つとして形態分類法に基づくFAB分類が知られる。FAB分類ではペルオキシダーゼ染色により急性骨髄性白血病と急性リンパ性白血病に大別され、さらに急性骨髄性白血病は以下のM0~M7に分類される。
Figure JPOXMLDOC01-appb-T000001
As one of the classifications of acute leukemia, FAB classification based on morphological classification is known. In the FAB classification, acute myeloid leukemia and acute lymphoblastic leukemia are roughly classified by peroxidase staining, and acute myeloid leukemia is further classified into the following M0 to M7.
Figure JPOXMLDOC01-appb-T000001
 本願の単球分化誘導剤により治療されうる「白血病」は、医師または研究者に用いられる一般的な基準に基づいて白血病と判断されるものであれば、特に限定されない。本願の単球分化誘導剤が骨髄系の血球系前駆細胞からの単球へ分化誘導能を有することから、好ましくは急性および/または慢性骨髄性白血病の治療に使用され、特に好ましくは急性骨髄性白血病の治療に使用される。 The “leukemia” that can be treated with the monocyte differentiation inducer of the present application is not particularly limited as long as it is determined as leukemia based on general criteria used by doctors or researchers. Since the monocyte differentiation inducer of the present application has the ability to induce differentiation from myeloid hematopoietic progenitor cells to monocytes, it is preferably used for the treatment of acute and / or chronic myeloid leukemia, particularly preferably acute myeloid Used to treat leukemia.
 本願の単球分化誘導剤は、白血病の治療において、異常増殖する白血病細胞を単球に分化誘導し得る。本願の単球分化誘導剤により白血病細胞は自己増殖能および不死化能を欠いた単球に分化誘導され得、正常単球の本来の短い寿命で穏やかに死滅しうる。すなわち、本願の単球分化誘導剤は分化誘導療法として使用されうる。 The monocyte differentiation inducer of the present application can induce differentiation of abnormally proliferating leukemia cells into monocytes in the treatment of leukemia. With the monocyte differentiation inducer of the present application, leukemia cells can be induced to differentiate into monocytes lacking the ability of self-proliferation and immortalization, and can be killed gently with the original short life span of normal monocytes. That is, the monocyte differentiation inducer of the present application can be used as differentiation induction therapy.
 本願の単球分化誘導剤は穏やかに白血病細胞を死滅させうるため、従来使用されてきた殺細胞作用を伴う化学療法剤で生じ得る副作用(例えば、食欲不振、嘔気、嘔吐、下痢等の消化器症状、発熱、全身倦怠感、敗血症、脱毛、急性呼吸促迫症候群、間質性肺炎、肝機能障害、黄疸、不整脈、心不全、消化管障害、中枢神経系障害、肝膿瘍、急性膵炎、肺浮腫、有痛性紅斑、CRP上昇、ALT(GPT)上昇、AST(GOT)上昇等)は抑制されうる。 Since the monocyte differentiation inducer of the present application can gently kill leukemia cells, side effects (eg, anorexia, nausea, vomiting, diarrhea, etc.) that may occur with conventional chemotherapeutic agents with cell killing action Symptoms, fever, general malaise, sepsis, hair loss, acute respiratory distress syndrome, interstitial pneumonia, liver dysfunction, jaundice, arrhythmia, heart failure, gastrointestinal dysfunction, central nervous system disorder, liver abscess, acute pancreatitis, lung edema, Painful erythema, CRP elevation, ALT (GPT) elevation, AST (GOT) elevation, etc.) can be suppressed.
 白血病(具体的には急性骨髄性白血病)の類縁疾患の例として、骨髄異形成症候群(MDS)が挙げられる。これは造血幹細胞の異常により生じ、血液細胞が分化の過程で未熟な細胞のままで異常増殖する。進行すると急性骨髄性白血病に移行することがある。 An example of a related disease of leukemia (specifically acute myeloid leukemia) is myelodysplastic syndrome (MDS). This is caused by an abnormality in hematopoietic stem cells, and the blood cells proliferate abnormally while remaining immature in the process of differentiation. As it progresses, it may shift to acute myeloid leukemia.
 本願の単球分化誘導剤は、骨髄異形成症候群の治療においても、白血病と同様に、未熟な異常細胞の分化を誘導して穏やかに死滅させうる。 The monocyte differentiation inducer of the present application can induce the differentiation of immature abnormal cells and gently kill them in the treatment of myelodysplastic syndromes as well as leukemia.
 本願の単球分化誘導剤は、白血病またはその類縁疾患の治療において、単球のレベルを改善し得、好中球/単球の減少により引き起こされる様々な症状(例えば細菌感染症等)の予防や改善のためにも使用されうる。 The monocyte differentiation inducer of the present application can improve the level of monocytes in the treatment of leukemia or related diseases, and prevent various symptoms (for example, bacterial infections etc.) caused by neutrophil / monocyte depletion. It can also be used for improvement.
 本明細書において「治療」とは、疾患を治療することに加え、疾患を予防する、疾患の症状を緩和する、疾患の進行を遅らせる、疾患の症状を抑制する、疾患の症状を寛解する、および疾患の症状の改善を誘発することを含み得る。 As used herein, "treatment" refers to treating disease, preventing disease, alleviating disease symptoms, delaying disease progression, suppressing disease symptoms, ameliorating disease symptoms, And inducing amelioration of disease symptoms.
 本願において「有効量」は、単球分化誘導による利益を患者に与えるために必要な活性薬剤の量である。 In the present application, the “effective amount” is the amount of the active agent necessary to give the patient the benefit of inducing differentiation of monocytes.
 本願において、アルベンダゾールは他の薬剤と同時に、別々に、または連続的に組み合わせて投与されることができる。
 同時に投与される場合、アルベンダゾールと他の薬剤は同一の製剤中に含まれていても、別々の製剤中に含まれていてもよい。別々の製剤中に含まれる場合、同一または異なる投与経路を介して同時に投与されてもよい。
 別々に投与される場合、アルベンダゾールと他の薬剤は異なる投薬計画に従って別々に投与されてもよく、異なる投与経路によって投与されてもよい。
 連続的に投与される場合、いずれの薬剤が先に投与されてもよい。一方の治療薬の投与と、他方の投与との間の時間は、8時間未満が好ましい。より好ましくは、4時間未満、さらにより好ましくは、1時間未満である。
In the present application, albendazole can be administered in combination with other drugs simultaneously, separately or sequentially.
When administered simultaneously, albendazole and the other drug may be included in the same formulation or in separate formulations. When included in separate formulations, they may be administered simultaneously via the same or different routes of administration.
When administered separately, albendazole and the other agent may be administered separately according to different dosing schedules and may be administered by different routes of administration.
When administered sequentially, any agent may be administered first. The time between administration of one therapeutic agent and the other is preferably less than 8 hours. More preferably, it is less than 4 hours, and even more preferably, it is less than 1 hour.
 白血病またはその類縁疾患(例えば骨髄異形成症候群)の治療においては、当該他の薬剤の例として、従来使用されている薬剤(例えば、カルムスチン、クロラムブシル、ロムスチン、メクロレタミン、アザシチジン、アシクロビル、L-アスパラギナーゼ、亜ヒ酸、アレムツズマブ、イダルビシン、イホスファミド、イブリツモマブチウキセタン、イマチニブ、イリノテカン、インターフェロンα、エトポシド、エノシタビン、エピルビシン、エリスロポエチン、オファツムマブ、オールトランス型レチノイン酸、カルボプラチン、クラドリビン、ゲムシタビン、ゲムツヅマブオゾガマイシン、サリドマイド、シクロスポリン、シクロホスファミド、シスプラチン、シタラビン、ジドブシン、ソブゾキサン、ダウノルビシン、ダカルバジン、ダサチニブ、タミバロテン、チオグアニン、デキサメタゾン、ドキソルビシン、ニロチニブ、ネララビン、ヒドロキシウレア、ピラルビシン、ビンクリスチン、ビンデシン、ビンブラスチン、ブスルファン、フルダラビン、ブレオマイシン、プレドニゾロン、プロカルバジン、ベンダムスチン、ペントスタチン、ボルテゾミブ、ポマリドミド、ミトキサントロン、メチルプレドニゾロン、メトトレキサート、メルカルトプリン、メルファラン、モガムリズマブ、ラニムスチン、リツキシマブ、ルキソリチニブ、レナリドミド等)が挙げられる。 In the treatment of leukemia or related diseases (for example, myelodysplastic syndrome), as other examples of such drugs, conventionally used drugs (for example, carmustine, chlorambucil, lomustine, mechloretamine, azacitidine, acyclovir, L-asparaginase, Arsenite, alemtuzumab, idarubicin, ifosfamide, ibritumomab tiuxetan, imatinib, irinotecan, interferon alpha, etoposide, enocitabine, epirubicin, erythropoietin, ofatatumumab, all-trans retinoic acid, carbogetum zobin, Mycin, thalidomide, cyclosporine, cyclophosphamide, cisplatin, cytarabine, zidobucin, sobuzoxane, daunorubicin, dacarbazine, Dasatinib, tamibarotene, thioguanine, dexamethasone, doxorubicin, nilotinib, nelarabine, hydroxyurea, pirarubicin, vincristine, vindesine, vinblastine, busulfan, fludarabine, bleomycin, prednisolone, procarbazine, bendamustine, pentomistatone, bortezidotron , Methotrexate, mercatopurine, melphalan, mogamulizumab, ranimustine, rituximab, ruxolitinib, lenalidomide, etc.).
 本願におけるアルベンダゾール含有単球分化誘導剤は、1以上の医薬上許容される担体を混合して、例えば、錠剤、カプセル剤、顆粒剤、散剤、トローチ剤、シロップ剤、乳剤、懸濁剤等の経口剤、或いは外用剤、坐剤、注射剤、点眼剤、経鼻剤、経肺剤等の非経口剤の形態に、通常の方法で調製されうる。 The albendazole-containing monocyte differentiation inducer in the present application is mixed with one or more pharmaceutically acceptable carriers, for example, tablets, capsules, granules, powders, troches, syrups, emulsions, suspensions, etc. Or a parenteral preparation such as an external preparation, a suppository, an injection, an eye drop, a nasal preparation, a pulmonary preparation and the like.
 本願にかかる単球分化誘導剤の好ましい製剤形として、例えば、錠剤および注射剤が挙げられる。 Examples of preferable dosage forms of the monocyte differentiation inducer according to the present application include tablets and injections.
 本願の経口剤に含まれる担体/添加剤は、通常用いられる医薬的に許容されるものであれば特に限定されないが、例えば、
 賦形剤(例えば、乳糖、白糖、D-マンニトール、D-ソルビトール、トウモロコシデンプン、デキストリン、微結晶セルロース、結晶セルロース、カルメロース、カルメロースカルシウム、カルボキシメチルスターチナトリウム、低置換度ヒドロキシプロピルセルロース、およびアラビアゴム);
 崩壊剤(例えば、カルメロース、カルメロースカルシウム、カルメロースナトリウム、カルボキシメチルスターチナトリウム、クロスカルメロースナトリウム、クロスポビドン、低置換度ヒドロキシプロピルセルロース、ヒドロキシプロピルメチルセルロース、および結晶セルロース);
 結合剤(例えば、ヒドロキシプロピルセルロース、ヒドロキシプロピルメチルセルロース、ポビドン、結晶セルロース、白糖、デキストリン、デンプン、ゼラチン、カルメロースナトリウム、およびアラビアゴム);
 流動化剤(例えば、軽質無水ケイ酸、およびステアリン酸マグネシウム);
 滑沢剤(例えば、ステアリン酸マグネシウム、ステアリン酸カルシウム、およびタルク)などが使用され得る。
The carrier / additive contained in the oral preparation of the present application is not particularly limited as long as it is a pharmaceutically acceptable one that is usually used.
Excipients such as lactose, sucrose, D-mannitol, D-sorbitol, corn starch, dextrin, microcrystalline cellulose, crystalline cellulose, carmellose, carmellose calcium, sodium carboxymethyl starch, low substituted hydroxypropylcellulose, and arabic Rubber);
Disintegrants (eg, carmellose, carmellose calcium, carmellose sodium, carboxymethyl starch sodium, croscarmellose sodium, crospovidone, low substituted hydroxypropylcellulose, hydroxypropylmethylcellulose, and crystalline cellulose);
Binders (eg, hydroxypropylcellulose, hydroxypropylmethylcellulose, povidone, crystalline cellulose, sucrose, dextrin, starch, gelatin, carmellose sodium, and gum arabic);
Fluidizing agents (eg, light anhydrous silicic acid, and magnesium stearate);
Lubricants such as magnesium stearate, calcium stearate, and talc can be used.
 本願の液剤(注射剤を含む)に含まれる担体/添加剤は、通常用いられる医薬的に許容されるものであれば特に限定されないが、例えば、
 溶剤(例えば、水、エタノール、プロピレングリコール、マクロゴール、グリセリンなど);
 溶解補助剤(例えば、プロピレングリコール、安息香酸ベンジル、エタノール、トリエタノールアミン、炭酸ナトリウム、クエン酸ナトリウム、トロメタモール(トリス[ヒドロキシメチル]アミノメタン)、メグルミンなど);
 等張化剤(例えば、ブドウ糖、グリセリン、D-マンニトール、D-ソルビトール、塩化ナトリウム、ショ糖、塩化カリウムなど);
 緩衝剤(例えばリン酸三ナトリウム、リン酸水素ナトリウム、リン酸二水素ナトリウム、リン酸二水素カリウム、ホウ酸、クエン酸、クエン酸ナトリウム、酒石酸、酢酸、酢酸ナトリウム、イプシロン-アミノカプロン酸、グルタミン酸ナトリウムなど);
 pH調整剤(例えば、リン酸水素ナトリウム、酢酸ナトリウム、炭酸ナトリウム、クエン酸ナトリウム、水酸化ナトリウム、塩酸など);
 溶解剤(例えばメグルミン)などが使用され得る。
The carrier / additive contained in the liquid preparation (including injection) of the present application is not particularly limited as long as it is a pharmaceutically acceptable one that is usually used.
Solvent (eg, water, ethanol, propylene glycol, macrogol, glycerin, etc.);
Solubilizers (eg, propylene glycol, benzyl benzoate, ethanol, triethanolamine, sodium carbonate, sodium citrate, trometamol (tris [hydroxymethyl] aminomethane), meglumine, etc.);
Isotonic agents (eg, glucose, glycerin, D-mannitol, D-sorbitol, sodium chloride, sucrose, potassium chloride, etc.);
Buffers (eg trisodium phosphate, sodium hydrogen phosphate, sodium dihydrogen phosphate, potassium dihydrogen phosphate, boric acid, citric acid, sodium citrate, tartaric acid, acetic acid, sodium acetate, epsilon-aminocaproic acid, sodium glutamate Such);
pH adjusting agents (for example, sodium hydrogen phosphate, sodium acetate, sodium carbonate, sodium citrate, sodium hydroxide, hydrochloric acid, etc.);
Solubilizers (eg, meglumine) and the like can be used.
 本願にかかる単球分化誘導剤に含有されるアルベンダゾールの量はとくに限定されず広範囲に選択される。例えば、錠剤の場合、組成物に対して約0.05重量%~約90重量%である。注射剤の場合は、好ましくは約0.01重量%~約50重量%、さらに好ましくは約0.05重量%~約20重量%、さらに好ましくは約0.2重量%~約10重量%であり、使用時に希釈用液(例えば生理食塩液)で適宜希釈して投与(例えば静脈投与)されうる。 The amount of albendazole contained in the monocyte differentiation inducer according to the present application is not particularly limited and is selected in a wide range. For example, in the case of a tablet, it is about 0.05% to about 90% by weight based on the composition. In the case of an injection, it is preferably about 0.01 wt% to about 50 wt%, more preferably about 0.05 wt% to about 20 wt%, more preferably about 0.2 wt% to about 10 wt%. Yes, at the time of use, it can be appropriately diluted with a diluting solution (eg, physiological saline) and administered (eg, intravenous administration).
 本願のアルベンダゾールの投与量は、投与方法、対象、対象の年齢、疾患の程度、症状、剤形、投与ルート等により適宜選択されるが、例えば、経口的に、一日あたり、0.1mg~100g、好ましくは1mg~50g、さらに好ましくは10mg~20g、より好ましくは100mg~10gの用量で、投与されうる。アルベンダゾールの1日あたりの投与量の下限値の例として、0.1mg、1mg、10mg、100mg、および400mgが挙げられ、上限値の例として、700mg、1g、3g、5g、10g、20g、50g、100gが挙げられ、アルベンダゾールの1日あたりの投与量の好ましい範囲は該上限値と該下限値の組合せにより示されうる。1日あたりのアルベンダゾールの量を、1回~数回に分けて投与してもよい。 The dose of albendazole of the present application is appropriately selected depending on the administration method, the subject, the age of the subject, the degree of the disease, symptoms, dosage form, administration route, etc., for example, orally 0.1 mg per day It can be administered at a dose of ˜100 g, preferably 1 mg to 50 g, more preferably 10 mg to 20 g, more preferably 100 mg to 10 g. Examples of the lower limit of the daily dose of albendazole include 0.1 mg, 1 mg, 10 mg, 100 mg, and 400 mg, and examples of the upper limit include 700 mg, 1 g, 3 g, 5 g, 10 g, 20 g, 50 g and 100 g can be mentioned, and a preferable range of the daily dose of albendazole can be indicated by a combination of the upper limit value and the lower limit value. The amount of albendazole per day may be administered in one to several divided doses.
 本願の単球分化誘導剤の投与対象は特に限定されないが、好ましくは哺乳動物(ヒトおよびヒト以外の哺乳類(例えばウシ、ウマ、ブタ、イヌ、ネコ、マウス、ラット、ウサギ、サル))であり、さらに好ましくはヒトである。 The subject of administration of the monocyte differentiation inducer of the present application is not particularly limited, but is preferably a mammal (human and non-human mammals (eg, cows, horses, pigs, dogs, cats, mice, rats, rabbits, monkeys)). More preferably, it is a human.
 クルッペル様転写因子4(KLF4)はKLFファミリー転写因子の一つであり、造血器腫瘍で発現が抑制されていることが知られている(非特許文献1)。本願発明者らは下記の試験例1~3に示すように、KLF4とその下流因子であるDPYSL2Aによるシグナル経路が血球系前駆細胞から単球への分化に寄与していることを明らかにした(2016年American Society of Hematologyにて発表:非特許文献2)。
 上記の知見に基づき、下記の試験例1~7に示すとおり、本願発明者らは、アルベンダゾール(ABZ)がKLF4の発現を上昇させDPYSL2Aの刺激を介して単球への分化誘導に高い活性を示すことを初めて明らかにし、本願発明に至った。
 以下、試験例を挙げて、本願発明を説明するが、本願発明はこれらの試験例に限定されるものではない。
Kruppel-like transcription factor 4 (KLF4) is one of the KLF family transcription factors and is known to be suppressed in hematopoietic tumors (Non-patent Document 1). As shown in Test Examples 1 to 3 below, the present inventors have clarified that the signal pathway by KLF4 and its downstream factor DPYSL2A contributes to the differentiation from hematopoietic progenitor cells to monocytes ( Presented at American Society of Hematology in 2016: Non-Patent Document 2).
Based on the above findings, as shown in Test Examples 1 to 7 below, the inventors of the present application show that albendazole (ABZ) increases the expression of KLF4 and is highly active in inducing differentiation into monocytes through stimulation of DPYSL2A. It was revealed for the first time that the present invention was achieved.
Hereinafter, the present invention will be described with reference to test examples, but the present invention is not limited to these test examples.
方法
<細胞株>
 急性骨髄性白血病(AML)由来THP-1およびKG-1a細胞をRIKEN biological resource center(BRC)(Japan)より購入した。
 SKNO-1細胞および胎児腎由来HEK293T細胞をJapanese Collection of Research Bioresources(JCRB)(Japan)より購入した。
 AML由来OCI-AML3およびMOLM-13細胞をDeutsche Sammlung von Mikroorganismen und Zellkulturen GmbH(DSMZ)(Germany)より購入した。
 AML由来MV4-11およびKG-1a細胞をAmerican Type Culture Collection(ATCC)(USA)より購入した。
 急性前骨髄球性白血病(APL)由来NB4およびUF-1細胞はDr. Y. Ikeda(Keio University School of Medicine、Japan)から提供された。
 AML由来ME-1細胞はDr. PP Liu(National Human Genome Research Institute, National Institutes of Health、米国)から提供された。
 HEK293T細胞は、ダルベッコ変法イーグル培地(DMEM)(10%熱失活ウシ胎仔血清(FBS)および1%ペニシリン-ストレプトマイシン(PS)補充)中、5%COおよび95%空気の加湿環境、37℃のインキュベーター内で維持した。
 他の細胞はRoswell Park Memorial Institute(RPMI)1640培地(10%FBSおよび1%PS含有)中、5%COおよび95%空気下、37℃で培養した。
Method <Cell line>
Acute myeloid leukemia (AML) -derived THP-1 and KG-1a cells were purchased from RIKEN biological resource center (BRC) (Japan).
SKNO-1 cells and fetal kidney-derived HEK293T cells were purchased from Japanese Collection of Research Bioresources (JCRB) (Japan).
AML-derived OCI-AML3 and MOLM-13 cells were purchased from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) (Germany).
AML-derived MV4-11 and KG-1a cells were purchased from American Type Culture Collection (ATCC) (USA).
NB4 and UF-1 cells derived from acute promyelocytic leukemia (APL) were provided by Dr. Y. Ikeda (Keio University School of Medicine, Japan).
AML-derived ME-1 cells were provided by Dr. PP Liu (National Human Genome Research Institute, National Institutes of Health, USA).
HEK293T cells were cultured in Dulbecco's Modified Eagle Medium (DMEM) (supplemented with 10% heat-inactivated fetal calf serum (FBS) and 1% penicillin-streptomycin (PS)) in a humidified environment of 5% CO 2 and 95% air, 37 Maintained in an incubator at 0 ° C.
Other cells were cultured at 37 ° C. in 5% CO 2 and 95% air in Roswell Park Memorial Institute (RPMI) 1640 medium (containing 10% FBS and 1% PS).
 上記の白血病細胞株はFAB分類では下表のように分類される。
Figure JPOXMLDOC01-appb-T000002
*:MOLM-13は骨髄異形成症候群(MDS)から発症した白血病である。
**:NB4およびUF-1はオールトランス型レチノイン酸(ATRA)耐性である。
The above leukemia cell lines are classified as shown in the following table in the FAB classification.
Figure JPOXMLDOC01-appb-T000002
*: MOLM-13 is a leukemia that developed from myelodysplastic syndrome (MDS).
**: NB4 and UF-1 are all-trans retinoic acid (ATRA) resistant.
<IC50評価>
 細胞生存分析のために、細胞を1×10細胞/mLの濃度で播種した。所定濃度の薬物を培養培地に加え、細胞を48時間培養した。ついで細胞生存率をWSTアッセイにより、Cell Count Reagent(nacalai tesque, Inc.)およびInfinite(登録商標)200 PRO multimode reader(TECAN)を製造者の説明に従って使用して評価した。パーセント阻害曲線を描いて、当該薬物のIC50値をmedian-effect法(Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul. 1984;22:27-55)に基づいて算出した。
<IC 50 evaluation>
For cell viability analysis, cells were seeded at a concentration of 1 × 10 5 cells / mL. A predetermined concentration of drug was added to the culture medium, and the cells were cultured for 48 hours. Followed by WST assay Cell viability, Cell Count Reagent (nacalai tesque, Inc.) and Infinite was evaluated using as described in (TM) 200 PRO multimode reader the (TECAN) manufacturer. Depict percent inhibition curve, an IC 50 value of the drug median-effect method (Chou TC, Talalay P. Quantitative analysis of dose-effect relationships:.. The combined effects of multiple drugs or enzyme inhibitors Adv Enzyme Regul 1984; 22 : 27-55).
<リアルタイム定量的PCR(qRT-PCR)>
 全RNAをRNeasy mini kit(Qiagen)を用いて単離し、Reverse script kit(TOYOBO)を用いて逆転写し、cDNAを作成した。リアルタイム定量的ポリメラーゼ連鎖反応(PCR)を7500 Real-Time PCR System(Applied Biosystems)により、製造者の推奨に従って行った。結果をGAPDHレベルに対して標準化した。相対的な発現レベルを2-ΔΔCt法を用いて算出した。qRT-PCRのために使用したプライマーを下表に示す。
Figure JPOXMLDOC01-appb-T000003
<Real-time quantitative PCR (qRT-PCR)>
Total RNA was isolated using RNeasy mini kit (Qiagen) and reverse transcribed using Reverse script kit (TOYOBO) to prepare cDNA. Real-time quantitative polymerase chain reaction (PCR) was performed on a 7500 Real-Time PCR System (Applied Biosystems) according to manufacturer's recommendations. Results were normalized to GAPDH levels. The relative expression level was calculated using the 2-ΔΔCt method. The primers used for qRT-PCR are shown in the table below.
Figure JPOXMLDOC01-appb-T000003
<siRNA干渉>
 ヒトKLF4およびDPYSL2Aを標的にする特異的なshRNAを設計し、pENTR4-H1tetOx1およびCS-RfA-ETVベクターへサブクローニングした。これらのベクターはDr. H. Miyoshi(RIKEN, BRC, Japan)から提供された。非標的コントロールshRNAをルシフェラーゼ(sh_Luc)に対して設計した。標的配列を下表に示す。
Figure JPOXMLDOC01-appb-T000004
<SiRNA interference>
Specific shRNAs targeting human KLF4 and DPYSL2A were designed and subcloned into the pENTR4-H1tetOx1 and CS-RfA-ETV vectors. These vectors were provided by Dr. H. Miyoshi (RIKEN, BRC, Japan). A non-targeted control shRNA was designed against luciferase (sh_Luc). The target sequences are shown in the table below.
Figure JPOXMLDOC01-appb-T000004
<発現プラスミド>
 ヒトKLF4、DPYSL2AおよびDPYSL2Bの各cDNAをPCRで増幅し、ついでpENTR1Aデュアルセレクションベクター(Thermo Fisher Scientific)、CSIV-TRE-Ubc-KT、およびpLenti CMV Puro DEST(addgene)発現ベクターに挿入した。CSIV-TRE-Ubc-KTはDr. H. Miyoshi(RIKEN, BRC, Japan)より提供を受けた。PCR産物は全てDNAシーケンシングにより確認した。
<Expression plasmid>
Human KLF4, DPYSL2A and DPYSL2B cDNAs were amplified by PCR and then inserted into pENTR1A dual selection vector (Thermo Fisher Scientific), CSIV-TRE-Ubc-KT, and pLenti CMV Puro DEST (addgene) expression vectors. CSIV-TRE-Ubc-KT was provided by Dr. H. Miyoshi (RIKEN, BRC, Japan). All PCR products were confirmed by DNA sequencing.
<レンチウイルスの生産および形質導入>
 レンチウイルスの生産をMorita K, Suzuki K, Maeda S, et al. Genetic regulation of the RUNX transcription factor family has antitumor effects. J Clin Invest. 2017;127(7):2815-2828.に記載のように行った。簡潔に言えば、HEK293T細胞をレンチウイルスベクター(例えばpsPAX2およびpMD2.G)でポリエチレンイミン(PEI, Sigma-Aldrich)によって一過性に同時導入した。トランスフェクションの48時間後、ウイルスの上清を集め、直ちに感染に使用し、ついで成功裏に形質導入された細胞をフローサイトメーターAria III(BD Biosciences)で免疫蛍光(Kusabira-OrangeまたはVenus)に基づいて選別した。
<Lentivirus production and transduction>
Lentivirus production is performed as described in Morita K, Suzuki K, Maeda S, et al. Genetic regulation of the RUNX transcription factor family has antitumor effects. J Clin Invest. 2017; 127 (7): 2815-2828. It was. Briefly, HEK293T cells were transiently co-transfected with polyethyleneimine (PEI, Sigma-Aldrich) with lentiviral vectors (eg psPAX2 and pMD2.G). Forty-eight hours after transfection, viral supernatants are collected and used immediately for infection, and then successfully transduced cells are immunofluorescent (Kusabira-Orange or Venus) with a flow cytometer Aria III (BD Biosciences). Sorted based on.
<免疫ブロット>
 免疫ブロットを、Morita K, Masamoto Y, Kataoka K, et al. BAALC potentiates oncogenic ERK pathway through interactions with MEKK1 and KLF4. Leukemia. 2015;29(11):2248-2256.に記載のように行った。メンブランを以下の一次抗体:抗KLF4(#4038, Cell Signaling Technology)、抗GAPDH(FL-335, Santa Cruz Biotechnology, Inc.)、抗DPYSL2(HPA002381, Sigma-Aldrich)、抗DPYSL抗体(HPA002381、Sigma-Aldrich)で反応させ、HRPコンジュゲート抗ウサギIgGおよび抗マウスIgG(Cell Signaling Technology)を2次抗体として使用した。Chemi-Lumi One Super(nacalai tesque, Inc.)およびChemiDoc(商標) XRS+ Imager(Bio-Rad Laboratories, Inc.)を製造者の推奨に従って使用して、ブロットを可視化した。タンパク質レベルをImage Lab Software(Bio-Rad Laboratories, Inc.)を用いて定量した。
<Immunoblotting>
Immunoblots were performed as described by Morita K, Masamoto Y, Kataoka K, et al. BAALC potentiates oncogenic ERK pathway through interactions with MEKK1 and KLF4. Leukemia. 2015; 29 (11): 2248-2256. The membrane was treated with the following primary antibodies: anti-KLF4 (# 4038, Cell Signaling Technology), anti-GAPDH (FL-335, Santa Cruz Biotechnology, Inc.), anti-DPYSL2 (HPA002381, Sigma-Aldrich), anti-DPYSL antibody (HPA002381, Sigma) -Aldrich) and HRP-conjugated anti-rabbit IgG and anti-mouse IgG (Cell Signaling Technology) were used as secondary antibodies. Blots were visualized using Chemi-Lumi One Super (nacalai tesque, Inc.) and ChemiDoc XRS + Imager (Bio-Rad Laboratories, Inc.) according to the manufacturer's recommendations. Protein levels were quantified using Image Lab Software (Bio-Rad Laboratories, Inc.).
<統計>
 コントロール群と試験群間の差違の統計的有意性は両側unpairedスチューデントt検定により評価し、p値が0.05より小さい時に有意とした。2つの母集団における等分散性はF検定で算出した。結果は、3つの独立した試験から得られた平均±SEM値として表した。移植実験においては、動物をランダムに各試験群に分け、処理は盲検法により行った。各群の生存率をlog-rank検定を用いて比較した。
<Statistics>
The statistical significance of the difference between the control group and the test group was evaluated by a two-sided unpaired student t test, and was considered significant when the p value was less than 0.05. Equal variance in the two populations was calculated by F test. Results were expressed as mean ± SEM values obtained from three independent tests. In the transplantation experiment, the animals were randomly divided into test groups, and the treatment was performed in a blinded manner. The survival rate of each group was compared using a log-rank test.
<マウス>
 NOD/Shi-scid、IL-2RγKO(NOG)マウスをCentral Institute for Experimental Animals(Japan)から購入した。同腹仔をコントロールとして全ての試験に使用した。
<Mouse>
NOD / Shi-scid, IL-2RγKO (NOG) mice were purchased from Central Institute for Experimental Animals (Japan). Litters were used as controls for all studies.
<異種移植マウスモデル>
 ヒトがん細胞株の異種移植マウスモデルをNOGマウスを用いて作成した。AMLマウスモデルのために、THP-1細胞(2×10細胞/マウス)を静脈内投与した。末梢血(PB)を毎週採取し、キメラ化をフローサイトメーターにより抗ヒトCD45抗体(BD Biosciences)を用いてチェックした。注射1週間後、マウスにアルベンダゾール(50mg/kg体重、2回/週、経口(p.o.))、または等量のジメチルスルホキシド(DMSO)を投与した。
<Xenograft mouse model>
A xenograft mouse model of a human cancer cell line was created using NOG mice. For the AML mouse model, THP-1 cells (2 × 10 6 cells / mouse) were administered intravenously. Peripheral blood (PB) was collected weekly and chimerization was checked with anti-human CD45 antibody (BD Biosciences) by flow cytometer. One week after injection, mice received albendazole (50 mg / kg body weight, 2 times / week, oral (po)), or an equivalent amount of dimethyl sulfoxide (DMSO).
<試験例1>:KLF4の過剰発現はAML細胞を分化誘導する。
 KLF4またはコントロールをコードするレンチウイルスで形質導入されたTHP-1細胞を3μMドキシサイクリンで48時間処理した後、収集し、スライドガラス上にサイトスピンした。各スライドについてDiff-Quik染色(改変Giemsa染色)を行った。THP-1細胞の代表的な顕微鏡画像を図1に示す。
 図1に示すとおり、KLF4をAML細胞株で過剰発現させると、異常細胞が単球様細胞に分化誘導された。
<Test Example 1>: Overexpression of KLF4 induces differentiation of AML cells.
THP-1 cells transduced with KLF4 or control encoding lentivirus were treated with 3 μM doxycycline for 48 hours, then harvested and cytospun onto glass slides. Each slide was subjected to Diff-Quik staining (modified Giemsa staining). A representative microscopic image of THP-1 cells is shown in FIG.
As shown in FIG. 1, when KLF4 was overexpressed in the AML cell line, abnormal cells were induced to differentiate into monocyte-like cells.
<試験例2>:DPYSL2AはKLF4の直接的な下流因子である。 <Test Example 2>: DPYSL2A is a direct downstream factor of KLF4.
KLF4またはコントロール(Ctrl)をコードするレンチウイルスで形質転換されたTHP-1およびMOLM-13細胞を3μMドキシサイクリンで48時間処理し、タンパク質抽出のために溶解し、KLF4、DPYSL2A、およびGAPDHの免疫ブロットを行った。結果を図2に示す。 THP-1 and MOLM-13 cells transformed with lentivirus encoding KLF4 or control (Ctrl) were treated with 3 μM doxycycline for 48 hours, lysed for protein extraction, and immunoblotting of KLF4, DPYSL2A, and GAPDH Went. The results are shown in FIG.
 図2に示されるとおり、KLF4はDPYSL2Aの遺伝子発現をアップレギュレートした。 As shown in FIG. 2, KLF4 up-regulated DPYSL2A gene expression.
<試験例3>:DPYSL2AはKLF4誘導分化の重要なメディエーターである。 <Test Example 3>: DPYSL2A is an important mediator of KLF4-induced differentiation.
試験例3-1:KLF4およびDPYSL2A(sh_DPYSL2A #1および#2)またはコントロールルシフェラーゼ(sh_Luc.)を標的にするshRNAをコードするレンチウイルスで形質導入されたTHP-1細胞を3μMドキシサイクリンで48時間処理し、ついでタンパク質抽出のために溶解して、KLF4、DPYSL2AおよびGAPDHについて免疫ブロットを行った。結果を図3-1に示す。 Test Example 3-1 THP-1 cells transduced with a lentivirus encoding shRNA targeting KLF4 and DPYSL2A (sh_DPYSL2A # 1 and # 2) or control luciferase (sh_Luc.) Were treated with 3 μM doxycycline for 48 hours And then lysed for protein extraction and immunoblotted for KLF4, DPYSL2A and GAPDH. The results are shown in Fig. 3-1.
試験例3-2:試験例3-1のTHP-1細胞を3μMドキシサイクリンで48時間処理して収集し、CD11bおよびCD14の細胞表面発現をフローサイトメトリーで測定した。結果を図3-2に示す。 Test Example 3-2: THP-1 cells of Test Example 3-1 were collected by treatment with 3 μM doxycycline for 48 hours, and cell surface expression of CD11b and CD14 was measured by flow cytometry. The results are shown in Fig. 3-2.
試験例3-3:試験例3-1のTHP-1細胞の細胞増殖曲線を図3-3に示す。
 細胞を3μMドキシサイクリンの存在下に培養した(n=3)。
Test Example 3-3: The cell growth curve of the THP-1 cell of Test Example 3-1 is shown in FIG. 3-3.
Cells were cultured in the presence of 3 μM doxycycline (n = 3).
試験例3-4:試験例3-1のTHP-1細胞の代表的な顕微鏡画像を図3-4に示す。
 細胞を3μMドキシサイクリンで48時間処理して収集し、スライドガラス上にサイトスピンした。各スライドについてDiff-Quik染色(改変Giemsa染色)を行った。
Test Example 3-4: A representative microscopic image of the THP-1 cell of Test Example 3-1 is shown in FIG. 3-4.
Cells were collected by treatment with 3 μM doxycycline for 48 hours and cytospun onto glass slides. Each slide was subjected to Diff-Quik staining (modified Giemsa staining).
 図3-1~4に示されるとおり、KLF4およびDPYSL2Aが発現したTHP-1細胞は単球様細胞に分化し、5日間の培養後ではDPYSL2Aがノックダウンされた細胞と比較して有意に細胞数は減少した。 As shown in FIGS. 3-1 to 4, THP-1 cells expressing KLF4 and DPYSL2A differentiated into monocyte-like cells, and after 5 days of culture, the cells were significantly different from those in which DPYSL2A was knocked down. The number has decreased.
<試験例4>:アルベンダゾールはKLF4-DPYSL2A系を刺激することによってAML細胞を強力に分化する。 <Test Example 4>: Albendazole strongly differentiates AML cells by stimulating the KLF4-DPYSL2A system.
試験例4-1:アルベンダゾールで処理されたTHP-1細胞におけるKLF4およびDPYSL2Aの相対的な発現。
 細胞をアルベンダゾール(1μM)で24時間処理し、ついで全RNAを調製して、リアルタイムRT-PCRで分析した。値はGAPDHで各サンプルごとに標準化されたのち、コントロールベクター形質導入細胞に対して標準化した(n=3)。結果を図4-1に示す。
Test Example 4-1: Relative expression of KLF4 and DPYSL2A in THP-1 cells treated with albendazole.
Cells were treated with albendazole (1 μM) for 24 hours, then total RNA was prepared and analyzed by real-time RT-PCR. Values were normalized for each sample with GAPDH and then normalized to control vector transduced cells (n = 3). The results are shown in Fig. 4-1.
試験例4-2:アルベンダゾールで処理されたTHP-1細胞におけるCD11bおよびCD14の細胞表面発現をフローサイトメトリーで測定した。
 細胞をアルベンダゾール(1μM)で24時間処理し、フローサイトメトリー分析のために収集した。結果を図4-2に示す。
Test Example 4-2: Cell surface expression of CD11b and CD14 in THP-1 cells treated with albendazole was measured by flow cytometry.
Cells were treated with albendazole (1 μM) for 24 hours and harvested for flow cytometric analysis. The results are shown in Fig. 4-2.
試験例4-3:試験例4-2のTHP-1細胞の代表的な顕微鏡画像。
 Diff-Quik染色(改変Giemsa染色)を各スライドについて行った。(拡大20×、スケールバー:50μm)。結果を図4-3に示す。
Test Example 4-3: Representative microscopic image of THP-1 cells of Test Example 4-2.
Diff-Quik staining (modified Giemsa staining) was performed on each slide. (Enlargement 20 ×, scale bar: 50 μm). The results are shown in Fig. 4-3.
試験例4-4:KLF4(sh_KLF4#1および#2)、DPYSL2A(sh_DPYSL2A#1および#2)またはコントロールルシフェラーゼ(sh_Luc.)を標的にするshRNAで形質導入されたTHP-1細胞の1nMPMAの存在下における成長曲線。
 shRNA発現を3μMドキシサイクリンで誘導した。結果を図4-4に示す。
Test Example 4-4: Presence of 1 nMPMA in THP-1 cells transduced with shRNA targeting KLF4 (sh_KLF4 # 1 and # 2), DPYSL2A (sh_DPYSL2A # 1 and # 2) or control luciferase (sh_Luc.) The growth curve below.
shRNA expression was induced with 3 μM doxycycline. The results are shown in Fig. 4-4.
 図4-1~3に示されるとおり、アルベンダゾールはKLF4およびDPYSL2Aの発現を上昇させ、THP-1細胞を単球様細胞に分化させた。図4-4に示されるとおり、KLF4またはDPYSL2Aをノックダウンした細胞ではアルベンダゾールの抗腫瘍効果が抑制されたことから、アルベンダゾールによるKLF4の発現によってこのような抗腫瘍効果が現れるということが示された。 As shown in FIGS. 4-1 to 3, albendazole increased the expression of KLF4 and DPYSL2A and differentiated THP-1 cells into monocyte-like cells. As shown in FIG. 4-4, since the antitumor effect of albendazole was suppressed in cells knocked down with KLF4 or DPYSL2A, the expression of KLF4 by albendazole shows that such an antitumor effect appears. It was done.
<試験例5>:IC50評価-I <Test Example 5>: IC 50 evaluation-I
試験例5-1:THP-1細胞およびMOLM-13細胞におけるアルベンダゾールの用量反応曲線。
 細胞を所定濃度のアルベンダゾールで48時間処理した(n=3)。結果を図5-1に示す。
Test Example 5-1: Dose-response curve of albendazole in THP-1 cells and MOLM-13 cells.
Cells were treated with a predetermined concentration of albendazole for 48 hours (n = 3). The results are shown in FIG.
試験例5-2:ATRA耐性NB4およびUF-1細胞におけるアルベンダゾールおよびATRAの用量反応曲線。
 細胞を所定濃度のアルベンダゾールまたはATRAで48時間処理した(n=3)。結果を図5-2および図5-3に示す。データは平均値±SEM値である。
Test Example 5-2: Albendazole and ATRA dose response curves in ATRA-resistant NB4 and UF-1 cells.
Cells were treated with a predetermined concentration of albendazole or ATRA for 48 hours (n = 3). The results are shown in FIGS. 5-2 and 5-3. Data are mean ± SEM values.
 下表に各細胞に対する、ATRAおよび/またはアルベンダゾールのIC50を示す。アルベンダゾールは各白血病細胞に対して非常に低いIC50を示した。
Figure JPOXMLDOC01-appb-T000005
The table below shows the IC 50 of ATRA and / or albendazole for each cell. Albendazole showed a very low IC 50 for each leukemia cell.
Figure JPOXMLDOC01-appb-T000005
<試験例6>:IC50評価-II
 AML細胞株を種々の濃度のアルベンダゾールで48時間処理し、IC50を算出した。結果を下表に示す。
 アルベンダゾールは幅広い種類のAML細胞に対して非常に低いIC50を示した。
Figure JPOXMLDOC01-appb-T000006
<Test Example 6>: IC 50 evaluation-II
AML cell lines were treated with various concentrations of albendazole for 48 hours and IC 50 calculated. The results are shown in the table below.
Albendazole showed a very low IC 50 for a wide variety of AML cells.
Figure JPOXMLDOC01-appb-T000006
<試験例7>インビボ試験:異種移植マウスモデル <Test Example 7> In vivo test: xenograft mouse model
 試験例7-1
 NOGマウスにTHP-1細胞を移植し、移植7日後、マウスをランダムに2群(アルベンダゾール群、DMSO群)に分けた(n=各7)。アルベンダゾール群およびDMSO群をアルベンダゾール(50mg/kg体重、2回/週、p.o.)または等量のDMSO(コントロール)で処理した。図6-1に示すとおり、DMSO群と比較してアルベンダゾール群の全生存は有意に延長した。
Test Example 7-1
NOG mice were transplanted with THP-1 cells, and 7 days after transplantation, the mice were randomly divided into 2 groups (albendazole group and DMSO group) (n = 7 each). The albendazole and DMSO groups were treated with albendazole (50 mg / kg body weight, 2 times / week, po) or an equivalent amount of DMSO (control). As shown in FIG. 6-1, overall survival of the albendazole group was significantly prolonged compared to the DMSO group.
 試験例7-2
 試験例7-1のとおり処理されたマウスを、適切に麻酔し、屠殺した。骨髄組織を採取し、ヘマトキシリン・エオシン(H&E)染色および抗ヒトCD45抗体による免疫組織化学的染色を各スライドについて行った。AML(THP-1細胞)異種移植マウス(移植後30日)の脾臓および肝臓の代表的な画像を図6-3に示す。図6-2に示すとおり、THP-1細胞の増殖は抑制された。図6-3に示すとおり、アルベンダゾールでは肝臓および脾臓の腫大は抑制された。
Test Example 7-2
Mice treated as in Test Example 7-1 were appropriately anesthetized and sacrificed. Bone marrow tissue was collected and hematoxylin and eosin (H & E) staining and immunohistochemical staining with anti-human CD45 antibody were performed on each slide. A representative image of the spleen and liver of an AML (THP-1 cell) xenograft mouse (30 days after transplantation) is shown in FIG. 6-3. As shown in FIG. 6-2, the proliferation of THP-1 cells was suppressed. As shown in FIG. 6-3, albendazole suppressed the enlargement of the liver and spleen.
 試験例8
 野生型C57BL/6マウスの骨髄細胞の中から、c-Kit表面抗原を染色し、陽性の細胞(この細胞はほとんど分化していない未熟な細胞と考えられており、骨髄系・リンパ系両方への分化能を有する)をフローサイトメーターを使って回収した。メチルセルロース半固形培地に、採取したc-kit表面抗原陽性細胞を5000個播種し、これにアルベンダゾールを0nM、100nM、500nMで投与して、1週間37℃で培養した。1週間後にコロニー形成を顕微鏡で観察し、カウントした。結果を図7に示す。図7から明かなとおり、単球系コロニーが顆粒球系コロニーよりも増えた。
Test Example 8
From the bone marrow cells of wild-type C57BL / 6 mice, c-Kit surface antigen was stained and positive cells (this cell is considered to be an immature cell that has hardly differentiated, and to both the myeloid and lymphatic systems. Were collected using a flow cytometer. 5000 collected c-kit surface antigen positive cells were seeded on a methylcellulose semi-solid medium, and albendazole was administered thereto at 0 nM, 100 nM, and 500 nM, and cultured at 37 ° C. for 1 week. One week later, colony formation was observed with a microscope and counted. The results are shown in FIG. As apparent from FIG. 7, the number of monocytic colonies increased from that of granulocyte colonies.

Claims (6)

  1.  アルベンダゾールを有効成分として含む、単球分化誘導剤。 A monocyte differentiation inducer containing albendazole as an active ingredient.
  2.  造血幹細胞の分化過程の阻害を伴う疾患を治療するための、請求項1に記載の単球分化誘導剤。 The agent for inducing differentiation of monocytes according to claim 1, for treating a disease accompanied by inhibition of the differentiation process of hematopoietic stem cells.
  3.  該造血幹細胞の分化過程の阻害を伴う疾患が、再生不良性貧血、コストマン症候群、または白血病もしくはその類縁疾患から選択される、請求項2に記載の単球分化誘導剤。 The monocyte differentiation inducer according to claim 2, wherein the disease accompanied by inhibition of the differentiation process of the hematopoietic stem cells is selected from aplastic anemia, Costman syndrome, leukemia or related diseases.
  4.  該造血幹細胞の分化過程の阻害を伴う疾患が、白血病またはその類縁疾患である、請求項2または3に記載の単球分化誘導剤。 The monocyte differentiation inducer according to claim 2 or 3, wherein the disease accompanied by inhibition of the differentiation process of hematopoietic stem cells is leukemia or a related disease thereof.
  5.  該白血病またはその類縁疾患が、慢性骨髄性白血病、急性骨髄性白血病または骨髄異形成症候群である、請求項2~4のいずれか1項に記載の単球分化誘導剤。 The monocyte differentiation inducer according to any one of claims 2 to 4, wherein the leukemia or a related disease thereof is chronic myelogenous leukemia, acute myeloid leukemia or myelodysplastic syndrome.
  6.  分化誘導療法として使用される、請求項4または5に記載の単球分化誘導剤。 The monocyte differentiation inducer according to claim 4 or 5, which is used as differentiation induction therapy.
PCT/JP2019/003802 2018-02-05 2019-02-04 Monocyte differentiation inducer containing albendazole WO2019151514A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2018018622A JP2021059495A (en) 2018-02-05 2018-02-05 Albendazole-containing monocyte differentiation inducing agent
JP2018-018622 2018-02-05

Publications (1)

Publication Number Publication Date
WO2019151514A1 true WO2019151514A1 (en) 2019-08-08

Family

ID=67479688

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2019/003802 WO2019151514A1 (en) 2018-02-05 2019-02-04 Monocyte differentiation inducer containing albendazole

Country Status (2)

Country Link
JP (1) JP2021059495A (en)
WO (1) WO2019151514A1 (en)

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
MATCHETT, K. B. ET AL.: "High-throughput screen identification of albendazole as a novel repurposed drug in acute myeloid leukaemia", BLOOD, vol. 130, no. 1, 7 December 2017 (2017-12-07), pages 5062, ISSN: 1528-0020, Retrieved from the Internet <URL:http://www.bloodjournal.org/content/130/Suppl_1/5062> [retrieved on 20190222] *
NAKAJIMA, 0. ET AL.: "Increase in the chemically-induced differentiation of human leukemia cell lines by tubulin disruptors", BIOLOGICAL AND PHARMACEUTICAL BULLETIN, vol. 17, no. 5, 15 May 1994 (1994-05-15), pages 742 - 744, XP001149147, ISSN: 0918-6158 *
ZHAO, Y. ET AL.: "Solubilisation and cytotoxicity of albendazole encapsulated in cucurbit[n]uril", ORGANIC & BIOMOLECULAR, vol. 6, no. 24, 21 December 2008 (2008-12-21), pages 4509 - 4515, XP055628491, ISSN: 1477-0520 *

Also Published As

Publication number Publication date
JP2021059495A (en) 2021-04-15

Similar Documents

Publication Publication Date Title
JP7432624B2 (en) Use of NAD+ and/or NAD+ inhibitors and/or NAD+ agonists and combinations thereof
US20180200301A1 (en) Low-Oxygen-Treated Mesenchymal Stem Cell and Use Thereof
Meshkibaf et al. Protective role of G-CSF in dextran sulfate sodium-induced acute colitis through generating gut-homing macrophages
JPWO2015178426A1 (en) Cancer stem cell growth inhibitor
Guo et al. Genetically modified" obligate" anaerobic Salmonella typhimurium as a therapeutic strategy for neuroblastoma
JP2023053338A (en) Modulating inflammasome activation of myeloid-derived suppressor cells for treating gvhd or tumor
Shi et al. Combination of quercetin and Adriamycin effectively suppresses the growth of refractory acute leukemia
US20150272992A1 (en) Treatment of Tumors with Activated Mesenchymal Stem Cells
US20210128639A1 (en) Cell preparation for treating brain tumor
Fowler et al. Targeting the canonical nuclear factor-κB pathway with a high-potency IKK2 inhibitor improves outcomes in a mouse model of idiopathic pneumonia syndrome
KR20220026959A (en) Pharmaceutical composition for treating or preventing lung cancer or inhibiting cancer metastasis, comprising exosomes as an active ingredient
Hossain et al. N-Myc knockdown and apigenin treatment controlled growth of malignant neuroblastoma cells having N-Myc amplification
US11746327B2 (en) Composition for culturing NK cells and method for culturing NK cells using same
WO2019151514A1 (en) Monocyte differentiation inducer containing albendazole
Gu et al. Enhancing fatty acid catabolism of macrophages within aberrant breast cancer tumor microenvironment can re-establish antitumor function
WO2021073610A1 (en) Methods and compositions for improving bone marrow hematopoietic functions
US20230059785A1 (en) Methods for enhancing t cells using venetoclax
Scolaro et al. Short-Term Caloric Restriction in Mice Promotes Resolution of Atherosclerosis, While Weight Regain Accelerates Its Progression
Wei et al. Genkwanin attenuates lung cancer development by repressing proliferation and invasion via phosphatidylinositol 3-kinase/protein kinase B pathway
Hu et al. Dysregulation of NK and CD8+ T cells by the microbiota promotes the progression of lung cancer
JP2019131505A (en) Anti-cancer agent
US20230365973A1 (en) Fats as a target for treating tumors and uses thereof
CN109913413B (en) PD-1 antibody loaded T cell in-vitro culture method, cell preparation and application thereof
JP2019514418A (en) Methods of treating diseases associated with ILC3 cells
Ngwa Targeting Vascular Endothelial Glutaminase in Triple Negative Breast Cancer

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: 19747601

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19747601

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP