WO2022019583A1 - Therapeutic agent for exosome-mediated disease - Google Patents

Therapeutic agent for exosome-mediated disease Download PDF

Info

Publication number
WO2022019583A1
WO2022019583A1 PCT/KR2021/009224 KR2021009224W WO2022019583A1 WO 2022019583 A1 WO2022019583 A1 WO 2022019583A1 KR 2021009224 W KR2021009224 W KR 2021009224W WO 2022019583 A1 WO2022019583 A1 WO 2022019583A1
Authority
WO
WIPO (PCT)
Prior art keywords
botulinum toxin
cancer
composition
tumor
exosome
Prior art date
Application number
PCT/KR2021/009224
Other languages
French (fr)
Korean (ko)
Inventor
곽성성
김태경
김민주
이효진
강원호
양기혁
Original Assignee
(주)메디톡스
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
Priority claimed from KR1020210009344A external-priority patent/KR20220012790A/en
Application filed by (주)메디톡스 filed Critical (주)메디톡스
Publication of WO2022019583A1 publication Critical patent/WO2022019583A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Definitions

  • the present disclosure relates to a therapeutic agent for an exosome-mediated disease, and more particularly, to an exosome-mediated disease containing botulinum toxin as an active ingredient, for example, a therapeutic agent for cancer.
  • Exosomes are a type of extracellular vesicles, and have a size of approximately 100 nm in diameter. They are surrounded by a lipid bilayer and are secreted by most cells. Exosomes are generated as an intraluminal membrane vesicle (ILV) within the endocytic system and are secreted during the fusion of the cell membrane and the multivesicular body (MVB). It has been reported that exosomes contribute to maintaining tissue homeostasis by regulating cell-cell communication. Exosomes released from cancer cells are involved in cancer progression.
  • IMV intraluminal membrane vesicle
  • Proliferative cancer cells (1) expand cancer tissue using angiogenesis, (2) acquire the ability to migrate and invade, and (3) acquire the ability to evade attack from immune cells, and ultimately metastatic lesion indicates the formation of Research results to date show that exosomes are involved in each of these processes.
  • International Publication No. WO2010/056337 discloses a method for detecting and diagnosing various cancers by quantifying exosomes isolated from a biological sample such as blood from a subject.
  • Botulinum toxin (Botulinum neurotoxin, BoNT) is a polypeptide product of the anaerobic bacterium Clostridium botulinum, and is a toxic substance that specifically acts on nerve cells. Botulinum toxin is a toxic substance that intrinsically causes lethality, but it is associated with a number of conditions including: It is used to treat urologic disease, migraine, and the like.
  • International Publication No. WO2006/025976 discloses a method for treating atypical tissues such as hyperplastic tissues, cysts and neoplasms by topical administration of botulinum toxin to or near cancer.
  • International Publication No. WO2010/062955 discloses that a therapeutically effective amount of botulinum toxin is administered in combination with an anticancer drug or anticancer therapy to a non-neoplastic area near a neoplasm, but a therapeutically effective amount of botulinum toxin is A non-penetrate method for inhibiting the growth or metastasis of a neoplasm is disclosed.
  • a first object of the present disclosure is to provide a composition for treating an exosome-mediated disease, comprising a botulinum toxin as an active ingredient.
  • a second object of the present disclosure is to provide a method for treating an exosome-mediated disease by administering a therapeutically effective amount of a botulinum toxin to a subject in need thereof.
  • a third object of the present disclosure is to provide a botulinum toxin for use as a therapeutic agent for exosome-mediated diseases.
  • a fourth object of the present disclosure relates to the use of botulinum toxin as a therapeutic agent for exosome-mediated diseases.
  • a first aspect of the present disclosure relates to a composition for treating an exosome-mediated disease, comprising a botulinum toxin as an active ingredient.
  • a second aspect of the present disclosure relates to a method of treating an exosome-mediated disease by administering to a subject in need thereof a therapeutically effective amount of a botulinum toxin.
  • a third aspect of the present disclosure relates to a botulinum toxin for use as a therapeutic agent for exosome-mediated diseases.
  • a fourth aspect of the present disclosure relates to the use of botulinum toxin as a therapeutic agent for exosome-mediated diseases.
  • 'botulinum toxin' refers to a botulinum toxin protein molecule produced by bacteria or recombinantly, including all serotypes and variants or fusion proteins thereof.
  • the Clostridium botulinum strain produces immunologically distinct neurotoxins (types A-G).
  • the molecular masses of type A-G neurotoxin (NTX) are about 150 kDa (7S).
  • NTX neurotoxin
  • PTX progenitor toxins
  • 12S toxin consists of NTX and a nontoxic nonhemagglutinin (named NTNH) having no hemagglutinin activity
  • 19S and 16S toxin consists of NTX, NTNH and hemagglutinin.
  • Type A strains produce three types of toxins (19S, 16S and 12S).
  • Type B, C and D strains produce 16S and 12S toxins. Under alkaline conditions, PTX dissociates into NTX and non-toxic components, 19S and 16S toxins dissociate into NTX and NTNH and non-toxic components (complex) of hemagglutinin, and 12S toxins dissociate into NTX and NTNH.
  • the botulinum toxin may be of any of the above-described forms, for example, may have a molecular weight of 19S, 16S, 12S, or 7S.
  • Botulinum toxin includes botulinum toxin derivatives.
  • the botulinum toxin derivative may be a natural botulinum toxin having activity or a recombinant prototype botulinum toxin, or a derivative comprising one or more chemical or functional modifications on a partial chain.
  • the botulinum toxin derivative may be a modified toxin having one or more amino acids deleted, modified or substituted.
  • the botulinum toxin may be a recombinant peptide, a fusion protein, or a hybrid neurotoxin prepared, for example, from subunits or domains of different toxin serotypes.
  • the botulinum toxin may also be part of a whole molecule having toxin activity, and may be used in combination or as part of a conjugated molecule, eg, a fusion protein.
  • the botulinum toxin is a botulinum toxin that does not comprise a chemical or functional modification, e.g., a natural botulinum toxin or a recombinant proto-botulinum toxin or a portion thereof, e.g., a complexed botulinum toxin (e.g., 19S toxin) or a non- complexed botulinum toxin (eg, 7S toxin).
  • a complexed botulinum toxin e.g., 19S toxin
  • a non- complexed botulinum toxin e.g., 7S toxin
  • administering' or 'administering' refers to a step of providing a pharmaceutical composition or an active ingredient to a subject.
  • the pharmaceutical composition may be administered via a variety of suitable routes.
  • “Pharmaceutical composition” means a formulation in which the active ingredient may be botulinum toxin.
  • the term 'formulation' means that in addition to the active ingredient, at least one additional ingredient is present in the pharmaceutical composition, such as albumin (human serum albumin or recombinant human albumin) and/or sodium chloride.
  • a pharmaceutical composition is a formulation suitable for administration to a subject, such as a human patient.
  • the pharmaceutical composition may be in lyophilized form, for example a solution formed after reconstitution of the lyophilized pharmaceutical composition with saline or water, or in the form of a solution that does not require reconstitution.
  • Pharmaceutical compositions may be liquid or solid.
  • the pharmaceutical composition may be free of animal-derived proteins and/or albumin.
  • 'therapeutically effective amount means the level, amount or concentration of a pharmaceutical composition comprising an agent, e.g., botulinum toxin or botulinum toxin, required to treat a disease, disorder or condition without causing significant adverse or adverse side effects do.
  • an agent e.g., botulinum toxin or botulinum toxin
  • 'Treat', 'treating' or 'treatment' refers to, for example, healing of wounded or damaged tissue, or altering, altering, enhancing, ameliorating, ameliorating and/or modifying an existing or recognized disease, disorder or condition. Means alleviation or reduction (including partial reduction, significant reduction, near complete reduction and complete reduction), resolution or prevention (temporary or permanent) of a disease, disorder or condition, by beautification, so as to achieve a desired therapeutic outcome do.
  • 'treatment' is a concept including prevention. By 'prevention' is meant delaying the onset of a disease, disorder or condition. Prevention may be considered complete if the onset of the disease, disorder or condition is delayed for a predetermined period.
  • treatment refers to the treatment of a disease, disorder, or medical condition in a patient, such as a mammal (especially a human), comprising one or more of the following:
  • botulinum toxin or a composition comprising the same may be useful as a therapeutic agent for exosome-mediated diseases, such as cancer, for example, metastatic cancer, for example, solid cancer by inhibiting exosome secretion. it is based
  • the botulinum toxin can be used without limitation of serotype, type A (BoNT/A), B (BoNT/B), C (BoNT/C), D (BoNT/D), E (BoNT/E) ), F(BoNT/F), G(BoNT/G), H(BoNT/H), X(BoNT/X), Enterococcus sp.
  • botulinum toxin J eBoNT/J
  • mosaic botulinum toxin and/or variants thereof It may be selected from the group consisting of.
  • mosaictoxins include BoNT/DC, BoNT/CD and BoNT/FA.
  • type A can be used.
  • the botulinum toxin is selected from various BoNT/A subtypes, eg, A1, A2, A3, A4, A5, A6, A7, A9, A10; BoNT/B subtypes such as B1, B2, B3, B4, B5, B6, B7, B8, Bnp and Bbv; BoNT/C subtypes such as C and CD; BoNT/D subtypes such as D and DC; BoNT/E subtypes such as E1, E2, E3, E4, E5, E6, E7, E8, E9; BoNT/F subtypes such as F1, F2, F3, F4, F5, F6, F7; and from the BoNT/G subtype, eg, subtype G.
  • BoNT subtypes include chimeric BoNTs, such as BoNT/DC, BoNT/CD, BoNT/FA, and the like.
  • a botulinum toxin or a composition comprising the same may be for treating an exosome-mediated disease.
  • the exosome-mediated disease may be cancer, a viral infection, a neurological disease or a rheumatic disease, and in particular the exosome-mediated disease may be cancer.
  • the cancer may be metastatic cancer.
  • the cancer may be a solid tumor.
  • solid cancer include breast cancer, lung cancer, head or neck cancer, colorectal cancer, esophageal cancer, laryngeal cancer, stomach cancer, liver cancer, pancreatic cancer, bone cancer, skin cancer, skin or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, perianal cancer, Colon cancer, breast cancer, fallopian tube carcinoma, endometrial carcinoma, cervical carcinoma, vaginal carcinoma, vulvar carcinoma, Hodgkin's disease, small intestine cancer, endocrine adenocarcinoma, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, prostate cancer , chronic or acute leukemia, lymphocytic lymphoma, bladder cancer, kidney or ureter cancer, renal cell carcinoma, renal pelvic carcinoma, CNS tumor, primary CNS lymphoma, spinal cord tumor, brain tumor, such as glioma (
  • the botulinum toxin can be used without limitation of serotype, and type A, B, C, D, E, F, G, H, or a combination thereof, for example type A, can be used.
  • composition comprising botulinum toxin may further comprise one or more pharmaceutically acceptable excipients or additives.
  • compositions or additives include stabilizers, ionic compounds, surfactants, buffers, lyophilization protectants, or combinations thereof, such as amino acids (eg methionine), salts (eg NaCl). ), buffers, nonionic surfactants (e.g. polysorbate, e.g. polysorbate 20), sugars (e.g. disaccharides such as sucrose), sugar alcohols (e.g. sorbitol), or their It can be a combination.
  • amino acids eg methionine
  • salts eg NaCl
  • nonionic surfactants e.g. polysorbate, e.g. polysorbate 20
  • sugars e.g. disaccharides such as sucrose
  • sugar alcohols e.g. sorbitol
  • composition may not contain albumin or animal-derived components or polysaccharides.
  • compositions include, for example, those disclosed in WO2009-008595 A1 or WO2012-134240 A2, the contents of which are incorporated herein by reference in their entirety.
  • compositions include, without limitation, commercially available pharmaceutical compositions containing botulinum toxin, such as, for example, BOTOX® (botulinum toxin type A neurotoxin complex with human serum albumin and sodium chloride) (Allergan, Inc.) (Allergan, Inc: Irvine, CA, USA), DYSPORT® (a powder reconstituted with 0.9% sodium chloride prior to use, Clostridium botulinum type A toxin hemagglutinin with human serum albumin and lactose in the formulation complex) (Ipsen Limited (Berkshire, UK)), MYOBLOCTM (injection solution containing botulinum toxin type B, human serum albumin, sodium succinate, and sodium chloride (approximately pH 56)) (Solstice Neurosciences) , Inc.
  • BOTOX® botulinum toxin type A neurotoxin complex with human serum albumin and sodium chloride
  • DYSPORT® a powder reconstituted with 0.9%
  • the composition may be Medytoxin®, Innotox®, Coretox®, or one or more combinations thereof, but is not limited thereto.
  • the composition may be formulated in any form, such as a solid or liquid formulation, for example, a lyophilized powder, a liquid, or a pre-filled syringe formulation.
  • the composition may be a formulation for topical administration.
  • topical administration may be intratumoral administration or peritumoral administration, and typically may be accomplished by intratumoral injection or peritumoral injection of an aqueous botulinum toxin solution.
  • the formulation can be administered subcutaneously or into a soft tissue, eg, intramuscularly.
  • the composition can inhibit tumor growth or inhibit tumor development at a location away from the location by intratumoral or peripheral administration at a specific location, and the composition can exhibit systemic effects when administered locally.
  • a therapeutically effective amount of botulinum toxin is about 0.01 U/kg to about 100 U/kg, about 0.1 U/kg to about 100 U/kg, about 0.2 U/kg to about 100 U/kg, about 0.2 U/kg kg to about 50 U/kg, from about 0.2 U/kg to about 30 U/kg, from about 0.2 U/kg to about 10 U/kg, from about 0.2 U/kg to about 1 U/kg.
  • unit refers to the LD50 dose, defined as the amount of botulinum toxin that killed 50% of mice receiving a botulinum toxin injection. , are used interchangeably within a product.
  • the botulinum toxin may be administered in single or multiple treatment sessions. Dosages may be administered as single or divided injections at the site of injection. In multiple treatment sessions, botulinum toxin may be administered at intervals of up to 6 months. In the multiple treatment session, the administration interval of the botulinum toxin includes the first treatment and the second treatment, and the dose of the second treatment may be less than, more than, or the same as the dose of the first treatment.
  • the dose, administration time, administration method, administration period or interval, etc. of the botulinum toxin may be appropriately selected and used by a person skilled in the art according to the patient's weight, age, sex, health condition, severity of disease, and the like.
  • botulinum toxin or a composition comprising the same can be used in subjects with increased exosome secretion.
  • a method of treating an exosome-mediated disease may comprise the steps of:
  • the secreted exosomes can be separated and quantified from a biological sample of a subject, for example, blood according to a conventional method known in the art, and there is no particular limitation on the type of sample, separation and quantification method.
  • the subject to which the botulinum toxin or a composition comprising the same is administered may include, without limitation, a human or an animal, for example, a human, a pig, a dog, a cat, a cow, a horse, a rat, and the like.
  • botulinum toxin or a composition comprising the same may be administered in combination with one or more other drugs or therapies.
  • “combination administration” refers to any form of two or more different therapeutic agents that allow a second therapeutic agent to be administered while the previously administered therapeutic agent is still effective in the body.
  • two therapeutic agents are simultaneously effective in a subject, which may include a synergistic effect of the two therapeutic agents.
  • the different therapeutic agents may be administered simultaneously or sequentially in the same formulation or in separate formulations.
  • botulinum toxin or a composition comprising the same may be administered in combination with other therapeutic agents, for example, anticancer agents, antiviral agents, cytokines, or immune agents.
  • other therapeutic agents for example, anticancer agents, antiviral agents, cytokines, or immune agents.
  • the botulinum toxin or a composition comprising the same may be administered in combination with a chemotherapeutic agent.
  • chemotherapeutic agents include alkylating agents, nitrosourases, antimetabolites, anticancer drugs, plant-derived alkaloids, topoisomerase inhibitors, hormonal drugs, hormone antagonists, leukopenia (neutropenia) therapeutic drugs, thrombocytopenia.
  • Exemplary cytotoxic agents that may be administered in combination include anti-microtubule agents, topoisomerase inhibitors, antimetabolites, mitotic inhibitors, alkylating agents, anthracyclines, vinca alkaloids, intercalating agents, and may interfere with signal transduction pathways. agents that promote apoptosis, proteosome inhibitors, and radiation (local or systemic irradiation).
  • additional therapeutic agents include, but are not limited to, peptides, polypeptides, proteins, fusion proteins, nucleic acid molecules, small molecules, mimetic agents, synthetic drugs, inorganic molecules, and organic molecules.
  • the botulinum toxin or a composition comprising the same may be administered in combination with an immunotherapeutic agent.
  • the immunotherapeutic agent may use any substance or therapy known in the art, and may include cytokines, cancer vaccines, oncolytic viruses, monoclonal antibodies, non-cytokine adjuvants, immune cells (T cells, NK cells, dendritic cells, B cells, etc.), and immune checkpoint inhibitors.
  • the immunotherapeutic agent is an immune checkpoint inhibitor.
  • Immune checkpoint inhibitors include peptides, antibodies, nucleic acid molecules and small molecules.
  • an immune checkpoint inhibitor may be administered to enhance the proliferative, migratory, persistence and/or cytotoxic activity of CD8+ T cells in a subject, and particularly tumor invasiveness of CD8+ T cells in a subject.
  • checkpoint inhibitors are activated T lymphocytes, such as cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PD-1), or the killer cell immunoglobulin-like receptor (KIR) family.
  • CTL4 cytotoxic T lymphocyte-associated protein 4
  • PD-1 programmed cell death 1
  • KIR killer cell immunoglobulin-like receptor
  • antagonists that block immunosuppressive receptors expressed by NK cells such as various members of It is a blocking antagonist.
  • an immune checkpoint inhibitor is an antibody or antigen-binding fragment thereof.
  • the checkpoint inhibitor is an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-TIM-3 antibody, an anti-LAG3 antibody, an anti-IDO1 antibody.
  • immune checkpoint inhibitors include Ipilimumab (Yervoy®, BMS/Ono), Tremelimumab (AstraZeneca), atezolizumab (Tecentric®, Roche), and nivolumab (nivolumab) (Opdivo®, BMS/Ono), Pembrolizumab (Keytruda®, MSD), Avelumab (Bavencio®, Pfizer/Merck Germany), Durvalumab ( Imfinzi®, AstraZeneca/MedImmune), and antigen-binding fragments thereof may be at least one selected from, but not limited to.
  • the other therapeutic agent may include a CTLA-4 antagonist, a PD-1 antagonist, a PD-L1 antagonist, a PD-L2 antagonist, or an EGFR antagonist.
  • a botulinum toxin or a composition comprising the same may be useful as a therapeutic agent for an exosome-mediated disease, for example, a cancer, for example, a metastatic cancer, for example, a solid cancer by inhibiting exosome secretion.
  • 1 is a graph showing tumor growth over time after administration of botulinum toxin in a mouse malignant melanoma transplantation model.
  • FIG. 2 is a graph showing the number of lung metastatic lesions by dose after administration of botulinum toxin in a mouse malignant melanoma transplant model.
  • 3A is a graph showing CCL2 levels in blood for each dose administered after administration of botulinum toxin in a mouse malignant melanoma transplantation model.
  • Figure 3b is a graph showing the level of IL-6 in the blood for each dose administered after botulinum toxin administration in a mouse malignant melanoma transplantation model.
  • FIG. 3c is a graph showing TNF levels in blood for each dose administered after administration of botulinum toxin in a mouse malignant melanoma transplantation model.
  • FIG. 4 is a graph showing tumor growth over time after administration of botulinum toxin in a mouse mammary gland tumor transplantation model.
  • 5 is a graph showing the number of exosomes in blood for each dose administered after botulinum toxin administration in a mouse mammary gland tumor transplantation model.
  • FIG. 6A is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a mouse malignant melanoma cell line (B16-F10).
  • 6B is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a human lung cancer cell line (A549).
  • FIG. 6c is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a human pancreatic cancer cell line (SNU324).
  • 6D is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a human pancreatic cancer cell line (Capan-2).
  • FIG. 7 is a graph showing the effect of reducing exosomes by treatment with botulinum toxin in mouse melanoma.
  • FIG. 8 is a graph showing the effect of reducing exosomes by treatment with botulinum toxin in a human pancreatic cancer cell line.
  • 9A is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a human brain cancer cell line (U87MG).
  • 9B is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a human prostate cancer cell line (PC3).
  • Example 1 Anticancer efficacy test of botulinum toxin in mouse malignant melanoma transplantation model
  • mice Male 6-week-old C57BL/6 (place of purchase: Orient Bio) mice were purchased, acclimatized and quarantined for 1 week, and 7-week-old mice were used for the experiment.
  • feed sterilized solid feed for laboratory animals (R40-10, SAFE, France) was freely fed, and negative water was fed freely by autoclaving tap water at high temperature.
  • the mouse was set at a temperature of 23 ⁇ 3°C, relative humidity of 55 ⁇ 15%, lighting time of 12 hours (8am to 8pm), ventilation frequency of 15 times/hour, and illumination intensity of 150 ⁇ 300 Lux. Raised under specific-pathogen-free conditions. This experiment was conducted after review and approval (A-2020-004) by the Animal Experimental Ethics Committee of Medytox Co., Ltd.
  • a B16-F10 mouse malignant melanoma cell line (KCLB No: 80008) was obtained from the Korean Cell Line Bank (KCLB).
  • B16-F10 cells were cultured in DMEM medium (CAT No: 11965-092, Gibco) containing 10% FBS (CAT No: 10082-147, Gibco) and 1% penicillin-streptomycin (CAT No: 15140-122, Gibco). °C, 5% CO 2 Conditions were cultured in an incubator. 0.1 ml of 5 ⁇ 10 5 cells were transplanted into the right flank of anesthetized C57BL/6 mice using a syringe.
  • sterile physiological saline Lith No: M8T7AF3, Daehan Pharmaceutical Co., Ltd.
  • Coretox Lith No: NSA19007A, Medytox
  • Dosing frequency One 10 vehicle 1 ml/kg within the tumor Once (8 days after tumor transplantation) 2 10 botulinum toxin 1.5 units/kg within the tumor Once (8 days after tumor transplantation) 3 10 botulinum toxin 5 units/kg within the tumor Once (8 days after tumor transplantation) 4 8 * botulinum toxin 15 units/kg within the tumor Once (8 days after tumor transplantation) 5 10 botulinum toxin 50 units/kg within the tumor Once (8 days after tumor transplantation)
  • mice On the 8th day after transplantation of malignant melanoma in mice, the body weight and tumor size were measured, and mice were randomly assigned to each group at an average size of about 55 mm 3 without any difference between the test groups.
  • vehicle and botulinum toxin were administered intratumorally according to the composition of the test group based on body weight. Tumor size was measured twice a week after tumor transplantation. The tumor size was measured with a vernier caliper and the tumor volume was calculated using the following equation using the measurements.
  • Tumor volume (mm3) (W 2 ⁇ L)/2
  • W (mm) width (short axis of tumor)
  • L (mm) length of tumor (long axis of tumor)
  • Tumor growth inhibition (1-Mean tumor size test group / Mean tumor size vehicle ) ⁇ 100
  • the measurement results of the tumor growth inhibition rate are shown in Table 2 and FIG. 1 .
  • intratumoral administration of 1.5 units/kg botulinum toxin alone showed an average tumor size of 1150 mm 3 and a tumor growth inhibition rate of 33%, and 5 units
  • the average 1336 mm3, 22% in the 15 units/kg administration group, the average 1195 mm3, 30%, and 50 units/kg administration group showed an average 682 mm3 and 60% tumor growth inhibition rate.
  • FIG. 2 The results of measuring the number of lung metastatic lesions are shown in FIG. 2 . As shown in FIG. 2 , the number of lung metastatic lesions was significantly reduced in the 1.5, 15, and 50 units/kg administration groups compared to the vehicle administration group.
  • CCL2 was decreased in the 15 units/kg administration group compared to the vehicle administration group ( FIG. 3a ), and TNF was significantly reduced in the 5 and 50 units/kg administration group ( FIG. 3b ). It was confirmed that IL-6 in the blood was also reduced by administration of botulinum toxin ( FIG. 3C ).
  • Example 2 Anticancer efficacy test of botulinum toxin in mouse mammary tumor transplantation model
  • mice After purchasing a 6-week-old female BALB/C mouse (Orient Bio), acclimatization and quarantine for 1 week, 7-week-old mice were used for the experiment. Feed, drinking water, and breeding conditions are the same as in Example 1. This experiment was conducted after review and approval (A-2020-004) by the Animal Experimental Ethics Committee of Medytox Co., Ltd.
  • the 4T1 mouse mammary carcinoma cell line (ATCC® CRL-2539TM) was obtained from the American Type Culture Collection (ATCC). 4T1 cells were cultured in RPMI-1640 medium (CAT No: LM011-51, Welgene) containing 10% FBS (CAT No: 10082-147, Gibco) and 1% penicillin-streptomycin (CAT No: 15140-122, Gibco) 37 °C, 5% CO 2 Conditions were cultured in an incubator. Using a syringe, 0.05 ml of 5 ⁇ 10 5 cells were transplanted into the fat layer in the fourth right breast of anesthetized female BALB/C mice.
  • RPMI-1640 medium CAT No: LM011-51, Welgene
  • FBS CAT No: 10082-147, Gibco
  • penicillin-streptomycin CAT No: 15140-122, Gibco
  • sterile physiological saline Lit No: M8T7AF3, Daehan Pharmaceutical Co., Ltd.
  • Coretox Lit No: NSA19007A, Medytox
  • Dosing frequency One 10 vehicle 1 ml/kg within the tumor Once (5 days after tumor transplantation) 2 10 botulinum toxin 1.5 units/kg within the tumor Once (5 days after tumor transplantation) 3 10 botulinum toxin 5 units/kg within the tumor Once (5 days after tumor transplantation) 4 10 botulinum toxin 15 units/kg within the tumor Once (5 days after tumor transplantation) 5 10 botulinum toxin 50 units/kg within the tumor Once (5 days after tumor transplantation)
  • mice On the 5th day after transplantation of mouse mammary gland carcinoma, the body weight and tumor size were measured, and the mice were randomly assigned to each group at an average size of about 69 mm 3 without any difference between the test groups.
  • vehicle and botulinum toxin were administered intratumorally according to the composition of the test group based on body weight. Tumor size was measured twice a week after tumor transplantation. Tumor size was measured in the same manner as in Example 1. On day 21 after tumor transplantation, before the tumor size reached 1000 mm 3 , blood was collected from surviving individuals and all animals were euthanized.
  • Exosomes in blood were isolated using Exoquick exosome precipitation solution (Cat No: EXOQ20A-1, Lot No: 200107-001, System Biosciences), and ExoELISA-ULTRA Complete Kit (Cat No: EXELULTRA-CD63-1, Lot No: 200226-002, System Biosciences). Statistical processing was performed in the same manner as in Example 1.
  • the measurement results of the tumor growth inhibition rate are shown in Table 4 and FIG. 4 .
  • the results of measuring the number of exosomes in the blood are shown in FIG. 5 .
  • the number of exosomes in the blood was significantly reduced in the 15 and 50 units/kg botulinum toxin administration group compared to the vehicle group.
  • B16-F10 mouse malignant melanoma cell lines (KCLB No: 80008), A549 human lung cancer cell line (KCLB No: 10185), SNU324 (KCLB No: 00324), and Capan-2 (KCLB No: 30080) human pancreatic cancer cell lines were obtained from the Korea Cell Line Bank. (KCLB: Korean Cell Line Bank) was obtained.
  • B16-F10 cells were cultured in DMEM medium (CAT No: 11965-092, Gibco) containing 10% FBS (CAT No: 10082-147, Gibco) and 1% penicillin-streptomycin (CAT No: 15140-122, Gibco). °C, 5% CO 2 Conditions were cultured in an incubator.
  • This experiment using a human tumor cell line was performed after being exempted from deliberation (IRB-2020-002) by the Institutional Bioethics Committee of Medytox Co., Ltd.
  • Coretox (Lot No: NSA19007A or NSA19004, Medytox) was used as a botulinum toxin, and doxorubicin (LOT No: DROTK-GP, TCI) was used as a positive control material.
  • CCK8 proliferation assay Cell Counting Kit-8, CK04, Dojindo
  • cells were seeded at a rate of 5 ⁇ 10 3 cells/0.1 ml per well in a 96-well plate (Cat No: 167008, Thermo Fisher) and cultured in an incubator for 24 hours. After 24 hours, after removing the medium from each well, botulinum toxin was diluted in the medium used for tumor cell line culture at a concentration of 0.016-250 U/ml, and treated with 10 ⁇ g/ml doxorubicin.
  • test substance was re-treated into each well. It was cultured in an incubator for 24 hours from the time of initial botulinum toxin treatment. After incubation for a total of 24 hours, each well was treated with 10 ⁇ l of CCK-8 reagent, and after additional incubation for 3 hours, absorbance was measured at 450 nm using SpectraMax i3 (Model No: 10192-220, Molecular Devices). The proliferation rate was compared. The growth rate was calculated using the following equation.
  • Example 4 Exosome reduction effect test of botulinum toxin in mouse melanoma cell line
  • a B16-F10 mouse malignant melanoma cell line (KCLB No: 80008) was obtained from the Korean Cell Line Bank (KCLB).
  • B16-F10 cells were cultured in DMEM medium (CAT No: 11965-092) containing 10% exosome-depleted FBS (CAT No: A2720801, Gibco) and 1% penicillin-streptomycin (CAT No: 15140-122, Gibco). , Gibco) in 37 °C, 5% CO 2 Conditions were cultured in an incubator.
  • Coretox (Lot No: NSA19004, Medytox) was used as a botulinum toxin.
  • Example 5 Testing the effect of botulinum toxin on reducing exosomes in human pancreatic cancer cell lines
  • the SNU324 human pancreatic cancer cell line (KCLB No: 00324) was obtained from the Korean Cell Line Bank (KCLB). SNU324 cells were cultured in RPMI-1640 medium (CAT No: LM011-51) containing 10% exosome-free FBS (CAT No: A2720801, Gibco) and 10% FBS, 25 mM HEPES (CAT No: 15630-080, Gibco). , Gibco) in 37 °C, 5% CO 2 Conditions were cultured in an incubator. This experiment using a human tumor cell line was conducted after being exempted from deliberation by the Institutional Bioethics Committee of Medytox Co., Ltd. (IRB-2020-002).
  • 1.2 ⁇ 10 6 cells/2 ml per well were seeded in a total of 6 wells and cultured for 24 hours in an incubator. After 24 hours, the medium from each well was removed, and botulinum toxin was diluted in the medium used for tumor cell line culture at a concentration of 10 U/ml, and the same amount of medium was treated for the control group. After 6 hours from the initial treatment time, the medium from each well was removed, and the same test substance was re-treated into each well. It was cultured in an incubator for 24 hours from the time of initial botulinum toxin treatment. After culturing for 24 hours, exosomes were measured for each well of the medium.
  • exosomes in the medium were isolated using ExoQuick-TC exosome precipitation solution (Cat No: EXOTC50A-1, Lot No: 191231-002, System Biosciences), and ExoELISA-ULTRA Complete Kit (Cat No: EXEL-ULTRA-CD63-1, Lot No: 200226-002, System Biosciences).
  • the results are shown in FIG. 8 .
  • the measured number of exosomes was calculated using a two-sided t-test between the test group and the control group ( * p ⁇ 0.05) As shown in FIG. 8, the effect of reducing exosomes in human pancreatic cancer cell lines by 10 U/ml botulinum toxin was it was confirmed that there is
  • U87MG Human brain cancer cell lines U87MG (KCLB No: 30014) and PC3 (KCLB No: 21435) human prostate cancer cell lines were obtained from Korean Cell Line Bank (KCLB).
  • U87MG cell line is DMEM (CAT No: LM007-01, Welgene) and PC3 cell line is RPMI-1640 (CAT No: LM011-01, Welgene) in 10% FBS (CAT No: 10082-147, Gibco) and 1% penicillin- Streptomycin (CAT No: 15140-122, Gibco) in a medium containing 37 °C, 5% CO 2 Conditions were cultured in an incubator. This experiment using a human tumor cell line was performed after being exempted from deliberation by the Institutional Bioethics Committee (IRB-2020-015) of Medytox Co., Ltd.
  • IRS-2020-015 Institutional Bioethics Committee
  • botulinum toxin 100 units Coretox (Lot No: NSA19007A or NSA19004, Medytox) was used, and doxorubicin (LOT No: DROTKGP, TCI) was used as a positive control.
  • CCK8 proliferation assay (Cell Counting Kit-8, CK04, Dojindo) was used.
  • Cells were seeded in a 96-well plate (Cat No: 167008, Thermo Fisher) at an amount of 5 ⁇ 10 3 cells/0.1 ml per well, and cultured in an incubator for 24 hours.
  • botulinum toxin was diluted in the medium used for tumor cell line culture at a concentration of 0.016-50 U/ml, or treated with 10 ⁇ g/ml doxorubicin.
  • the medium was removed from each well, and the same test substance was re-treated in each well. It was cultured in an incubator for 24 hours from the time of initial botulinum toxin treatment.
  • the measured tumor growth rate was calculated using a two-tailed t -test between the test group and the control group (* p ⁇ 0.05, ** p ⁇ 0.01, *** p ⁇ 0.001) of human brain tumor and prostate cancer cells by botulinum toxin. It was found that growth was significantly inhibited. A significant growth inhibitory effect was observed in the human brain tumor cell line (U87MG) in the 0.016 ⁇ 50 U / ml treatment group (Fig. 9a), and in the human prostate cancer (PC3) cell line in the 0.4 ⁇ 50 U / ml treatment group (Fig. 9b). .

Abstract

Disclosed is a therapeutic agent for an exosome-mediated disease, for example a cancer such as metastatic cancer, e.g., a solid cancer.

Description

엑소좀-매개된 질환 치료제Therapeutic agents for exosome-mediated diseases
본 개시는 엑소좀-매개된 질환의 치료제에 관한 것으로서, 보다 상세하게는 유효성분으로 보툴리눔 독소를 포함하는 엑소좀-매개된 질환, 예를 들어 암 치료제에 관한 것이다.The present disclosure relates to a therapeutic agent for an exosome-mediated disease, and more particularly, to an exosome-mediated disease containing botulinum toxin as an active ingredient, for example, a therapeutic agent for cancer.
엑소좀(exosomes)은 세포외 소낭(extracellular vesicles)의 일종으로, 대략 직경 100 ㎚의 크기를 갖는다. 이들은 지질 이중막으로 둘러싸여 있으며 대부분의 세포로부터 분비된다. 엑소좀은 엔도사이틱 시스템(endocytic system) 내에서 ILV(intraluminal membrane vesicle)로 생성되며 세포막과 MVB(multivesicular body)의 융합 동안 분비된다. 엑소좀이 세포-세포 소통(cell-cell communication)을 조절하여 조직 항상성(homeostasis) 유지에 기여하는 것으로 보고된 바 있다. 암 세포로부터 방출된 엑소좀은 암 진행에 관여한다. 증식성 암 세포는 (1) 혈관신생을 이용한 암 조직의 확장, (2) 이동 및 침투 능력 획득, 및 (3) 면역 세포로부터의 공격을 회피하는 능력 획득, 및 궁극적으로 전이성 병변(metastatic lesion)의 형성을 나타낸다. 지금까지의 연구 결과는 엑소좀이 이들 과정의 각각에 관여하고 있음을 보여준다. 국제공개 WO2010/056337호는 대상으로부터 혈액 등과 같은 생물학적 시료 내에서 분리된 엑소좀을 정량하여 다양한 암을 검출 및 진단하는 방법을 개시하고 있다.Exosomes are a type of extracellular vesicles, and have a size of approximately 100 nm in diameter. They are surrounded by a lipid bilayer and are secreted by most cells. Exosomes are generated as an intraluminal membrane vesicle (ILV) within the endocytic system and are secreted during the fusion of the cell membrane and the multivesicular body (MVB). It has been reported that exosomes contribute to maintaining tissue homeostasis by regulating cell-cell communication. Exosomes released from cancer cells are involved in cancer progression. Proliferative cancer cells (1) expand cancer tissue using angiogenesis, (2) acquire the ability to migrate and invade, and (3) acquire the ability to evade attack from immune cells, and ultimately metastatic lesion indicates the formation of Research results to date show that exosomes are involved in each of these processes. International Publication No. WO2010/056337 discloses a method for detecting and diagnosing various cancers by quantifying exosomes isolated from a biological sample such as blood from a subject.
보툴리눔 독소(Botulinum neurotoxin, BoNT)는 혐기성 박테리아 클로스트리디움 보툴리눔(Clostridium botulinum)의 폴리펩티드 생성물로서, 신경세포에 특이적으로 작용하는 독성물질이다. 보툴리눔 독소는 본래 치사를 일으키는 독성 물질이지만, 연축성 사경(cervical dystonia: CD), 안검경련(blepharospasm), 다한증(hyperhidrosis), 사시(strabismus), 이완불능(achalasia), 신경성 방광(neurogenic bladder), 비뇨기 질환(urologic disease), 편두통(migraine) 등의 치료를 위해 사용되고 있다.Botulinum toxin (Botulinum neurotoxin, BoNT) is a polypeptide product of the anaerobic bacterium Clostridium botulinum, and is a toxic substance that specifically acts on nerve cells. Botulinum toxin is a toxic substance that intrinsically causes lethality, but it is associated with a number of conditions including: It is used to treat urologic disease, migraine, and the like.
국제공개 WO2006/025976호는 보툴리눔 독소를 암 또는 그 근처에 국소 투여하여 과형성(hyperplastic) 조직, 낭포(cysts) 및 신생물(neoplasms)과 같은 비정형 조직을 치료하는 방법을 개시하고 있다. 또한, 국제공개 WO2010/062955호는 신생물 근처의 비-신생성 부위(non-neoplastic area)에 치료 유효량의 보툴리눔 독소를 항암 약물이나 항암 요법과 병용 투여하되, 치료 유효량의 보툴리눔 독소가 신생물을 투과(penetrate)하지 않는, 신생물의 성장이나 전이를 저해하는 방법을 개시하고 있다.International Publication No. WO2006/025976 discloses a method for treating atypical tissues such as hyperplastic tissues, cysts and neoplasms by topical administration of botulinum toxin to or near cancer. In addition, International Publication No. WO2010/062955 discloses that a therapeutically effective amount of botulinum toxin is administered in combination with an anticancer drug or anticancer therapy to a non-neoplastic area near a neoplasm, but a therapeutically effective amount of botulinum toxin is A non-penetrate method for inhibiting the growth or metastasis of a neoplasm is disclosed.
보툴리눔 독소의 엑소좀 분비 억제나 엑소좀-매개된 질환의 치료 효과에 대해서는 알려진 바 없었다.It was not known about the suppression of exosome secretion of botulinum toxin or the therapeutic effect of exosome-mediated diseases.
본 개시의 제 1 목적은 유효성분으로 보툴리눔 독소를 포함하는, 엑소좀-매개된 질환을 치료하기 위한 조성물을 제공하는 것이다.A first object of the present disclosure is to provide a composition for treating an exosome-mediated disease, comprising a botulinum toxin as an active ingredient.
본 개시의 제 2 목적은 치료 유효량의 보툴리눔 독소를 이를 필요로 하는 대상에게 투여하여 엑소좀-매개된 질환을 치료하는 방법을 제공하는 것이다.A second object of the present disclosure is to provide a method for treating an exosome-mediated disease by administering a therapeutically effective amount of a botulinum toxin to a subject in need thereof.
본 개시의 제 3 목적은 엑소좀-매개된 질환 치료제로 사용하기 위한 보툴리눔 독소를 제공하는 것이다.A third object of the present disclosure is to provide a botulinum toxin for use as a therapeutic agent for exosome-mediated diseases.
본 개시의 제 4 목적은 보툴리눔 독소의 엑소좀-매개된 질환 치료제로서의 사용에 관한 것이다.A fourth object of the present disclosure relates to the use of botulinum toxin as a therapeutic agent for exosome-mediated diseases.
본 개시의 제 1 측면은 유효성분으로 보툴리눔 독소를 포함하는, 엑소좀-매개된 질환을 치료하기 위한 조성물에 관한 것이다.A first aspect of the present disclosure relates to a composition for treating an exosome-mediated disease, comprising a botulinum toxin as an active ingredient.
본 개시의 제 2 측면은 치료 유효량의 보툴리눔 독소를 이를 필요로 하는 대상에게 투여하여 엑소좀-매개된 질환을 치료하는 방법에 관한 것이다.A second aspect of the present disclosure relates to a method of treating an exosome-mediated disease by administering to a subject in need thereof a therapeutically effective amount of a botulinum toxin.
본 개시의 제 3 측면은 엑소좀-매개된 질환 치료제로 사용하기 위한 보툴리눔 독소에 관한 것이다.A third aspect of the present disclosure relates to a botulinum toxin for use as a therapeutic agent for exosome-mediated diseases.
본 개시의 제 4 측면은 보툴리눔 독소의 엑소좀-매개된 질환 치료제로서의 사용에 관한 것이다.A fourth aspect of the present disclosure relates to the use of botulinum toxin as a therapeutic agent for exosome-mediated diseases.
본 명세서에서 사용되는, 용어 '보툴리눔 독소(botulinum toxin)'는 모든 혈청형(serotype) 및 이것의 변이체 또는 융합 단백질을 포함하여 세균 또는 재조합에 의해 생성되는 보툴리눔 독소 단백질 분자를 의미한다.As used herein, the term 'botulinum toxin' refers to a botulinum toxin protein molecule produced by bacteria or recombinantly, including all serotypes and variants or fusion proteins thereof.
클로스트리디움 보툴리눔 균주는 면역학적으로 구별되는 신경독소(타입 A-G)를 생산한다. 타입 A-G 신경독소(NTX)의 분자량(molecular masses)은 약 150 kDa(7S)이다. 산성 조건을 갖는 배양 배지 및 식품에서, NTX는 비독성 성분과 결합하여 PTX(progenitor toxins)라고 불리는 큰 복합체를 형성한다. 900kDa(19S), 500 kDa(16S) 및 300 kDa(12S)의 분자량을 갖는 PTX가 발견된다. 12S 독소는 NTX와 헤마글루티닌 활성을 갖지 않는 비독성 성분(nontoxic nonhemagglutinin; NTNH로 명명)으로 이루어지며, 19S 및 16S 독소는 NTX, NTNH 및 헤마글루티닌으로 이루어진다. 타입 A 균주는 3가지 형태의 독소(19S, 16S 및 12S)를 생산한다. 타입 B, C 및 D 균주는 16S와 12S 독소를 생산한다. 알칼리 조건에서, PTX는 NTX와 비독성 성분으로 해리되고, 19S 및 16S 독소는 NTX와 NTNH 및 헤마글루티닌의 비독성 성분(복합체)으로 해리되며, 12S 독소는 NTX와 NTNH로 해리된다. 본 명세서에서, 보툴리눔 독소는 상기한 어떠한 형태의 것일 수도 있으며, 예를 들어 19S, 16S, 12S, 또는 7S의 분자량을 갖는 것일 수 있다.The Clostridium botulinum strain produces immunologically distinct neurotoxins (types A-G). The molecular masses of type A-G neurotoxin (NTX) are about 150 kDa (7S). In culture media and foods with acidic conditions, NTX combines with non-toxic components to form large complexes called progenitor toxins (PTX). PTX with molecular weights of 900 kDa (19S), 500 kDa (16S) and 300 kDa (12S) are found. 12S toxin consists of NTX and a nontoxic nonhemagglutinin (named NTNH) having no hemagglutinin activity, and 19S and 16S toxin consists of NTX, NTNH and hemagglutinin. Type A strains produce three types of toxins (19S, 16S and 12S). Type B, C and D strains produce 16S and 12S toxins. Under alkaline conditions, PTX dissociates into NTX and non-toxic components, 19S and 16S toxins dissociate into NTX and NTNH and non-toxic components (complex) of hemagglutinin, and 12S toxins dissociate into NTX and NTNH. In the present specification, the botulinum toxin may be of any of the above-described forms, for example, may have a molecular weight of 19S, 16S, 12S, or 7S.
보툴리눔 독소는 보툴리눔 독소 유도체를 포함한다. 보툴리눔 독소 유도체는 보툴리눔 독소 활성을 갖는 천연 보툴리눔 독소 또는 재조합 원형 보툴리눔 독소의 일부 또는 일부 사슬 상에 하나 이상의 화학적 또는 기능적 변형을 포함하는 유도체일 수 있다. 예를 들어, 보툴리눔 독소 유도체는 결실, 변형 또는 치환된 하나 이상의 아미노산을 갖는 변형된 독소일 수 있다. 보툴리눔 독소는 재조합 펩티드, 융합 단백질, 또는 예를 들어, 상이한 독소 혈청형의 서브유닛 또는 도메인으로부터 제조된 하이브리드 신경독소일 수 있다. 보툴리눔 독소는 또한 독소 활성을 갖는 전체 분자의 일부일 수 있으며, 이것의 조합 또는 컨쥬게이션된 분자, 예를 들어, 융합 단백질의 일부로서 이용될 수 있다.Botulinum toxin includes botulinum toxin derivatives. The botulinum toxin derivative may be a natural botulinum toxin having activity or a recombinant prototype botulinum toxin, or a derivative comprising one or more chemical or functional modifications on a partial chain. For example, the botulinum toxin derivative may be a modified toxin having one or more amino acids deleted, modified or substituted. The botulinum toxin may be a recombinant peptide, a fusion protein, or a hybrid neurotoxin prepared, for example, from subunits or domains of different toxin serotypes. The botulinum toxin may also be part of a whole molecule having toxin activity, and may be used in combination or as part of a conjugated molecule, eg, a fusion protein.
일 태양에서, 보툴리눔 독소는 화학적 또는 기능적 변형을 포함하지 않는 보툴리눔 독소, 예를 들어 천연 보툴리눔 독소 또는 재조합 원형 보툴리눔 독소 또는 그의 일부, 예를 들어 복합체화된 보툴리눔 독소(예: 19S 독소) 또는 비-복합체화된 보툴리눔 독소(예: 7S 독소)일 수 있다.In one aspect, the botulinum toxin is a botulinum toxin that does not comprise a chemical or functional modification, e.g., a natural botulinum toxin or a recombinant proto-botulinum toxin or a portion thereof, e.g., a complexed botulinum toxin (e.g., 19S toxin) or a non- complexed botulinum toxin (eg, 7S toxin).
'투여' 또는 '투여하는 것'은 제약 조성물 또는 유효성분을 대상에게 제공하는 단계를 의미한다. 제약 조성물은 다양한 적절한 경로를 통하여 투여될 수 있다.'Administering' or 'administering' refers to a step of providing a pharmaceutical composition or an active ingredient to a subject. The pharmaceutical composition may be administered via a variety of suitable routes.
'제약 조성물'은 유효성분이 보툴리눔 독소일 수 있는 제제를 의미한다. “Pharmaceutical composition” means a formulation in which the active ingredient may be botulinum toxin.
'제제'라는 용어는 보툴리눔 신경독소 유효성분 이외에 적어도 1종의 추가적인 성분, 예를 들면 알부민(인간 혈청 알부민 또는 재조합 인간 알부민) 및/또는 소듐 클로라이드가 제약 조성물 중에 존재한다는 것을 의미한다. 제약 조성물은 인간 환자와 같은 대상에의 투여에 적합한 제제이다. 제약 조성물은 동결건조된 형태, 예를 들면 식염수 또는 물을 사용한 동결건조된 제약 조성물의 재구성 후 형성되는 용액, 또는 재구성을 필요로 하지 않는 용액 형태일 수 있다. 제약 조성물은 액체 또는 고체일 수 있다. 제약 조성물에는 동물 유래 단백질 및/또는 알부민이 없을 수 있다.The term 'formulation' means that in addition to the active ingredient, at least one additional ingredient is present in the pharmaceutical composition, such as albumin (human serum albumin or recombinant human albumin) and/or sodium chloride. A pharmaceutical composition is a formulation suitable for administration to a subject, such as a human patient. The pharmaceutical composition may be in lyophilized form, for example a solution formed after reconstitution of the lyophilized pharmaceutical composition with saline or water, or in the form of a solution that does not require reconstitution. Pharmaceutical compositions may be liquid or solid. The pharmaceutical composition may be free of animal-derived proteins and/or albumin.
'치료 유효량'은 유의성 있는 부정적이거나 불리한 부작용을 야기하지 않으면서 질환, 장애 또는 이상을 치료하는 데에 요구되는 작용제, 예를 들어 보툴리눔 독소 또는 보툴리눔 독소를 포함하는 제약 조성물의 수준, 양 또는 농도를 의미한다.'therapeutically effective amount' means the level, amount or concentration of a pharmaceutical composition comprising an agent, e.g., botulinum toxin or botulinum toxin, required to treat a disease, disorder or condition without causing significant adverse or adverse side effects do.
'치료하다', '치료하는 것' 또는 '치료'는 예컨대 상처를 입거나 손상된 조직의 치유, 또는 기존의 것이거나 인지된 질환, 장애 또는 이상을 변경, 변화, 강화, 개선, 개량 및/또는 미화하는 것에 의해, 원하는 치료상의 결과를 달성하도록 하는, 질환, 장애 또는 이상의 경감 또는 감소(일부 감소, 상당한 감소, 거의 완전한 감소 및 완전한 감소 포함), 해결 또는 예방(일시적이거나 영구적인 것)을 의미한다. 본 명세서에서, '치료'는 예방을 포함하는 개념이다. '예방'은 질병, 장애 또는 질환의 발병의 지연을 의미한다. 질병, 장애 또는 질환의 발병이 예정된 기간 동안 지연된 경우 예방은 완전한 것으로 간주될 수 있다.'Treat', 'treating' or 'treatment' refers to, for example, healing of wounded or damaged tissue, or altering, altering, enhancing, ameliorating, ameliorating and/or modifying an existing or recognized disease, disorder or condition. Means alleviation or reduction (including partial reduction, significant reduction, near complete reduction and complete reduction), resolution or prevention (temporary or permanent) of a disease, disorder or condition, by beautification, so as to achieve a desired therapeutic outcome do. As used herein, 'treatment' is a concept including prevention. By 'prevention' is meant delaying the onset of a disease, disorder or condition. Prevention may be considered complete if the onset of the disease, disorder or condition is delayed for a predetermined period.
일 태양에서 '치료(treatment)'는, 하기 중 하나 이상을 포함하는, 포유동물 (특히, 인간)과 같은 환자에서, 질환, 장애, 또는 의학적 병태의 치료를 의미한다:In one aspect 'treatment' refers to the treatment of a disease, disorder, or medical condition in a patient, such as a mammal (especially a human), comprising one or more of the following:
(a) 상기 질환, 장애, 또는 의학적 병태의 발생 방지, 즉 상기 질환 또는 의학적 병태의 재발 방지 또는 상기 질환 또는 의학적 병태에 걸리기 쉬운(pre-disposed) 환자의 예방적 치료(prophylactic treatment);(a) preventing the occurrence of said disease, disorder, or medical condition, i.e., preventing recurrence of said disease or medical condition, or prophylactic treatment of a patient pre-disposed to said disease or medical condition;
(b) 다른 치료제의 효과에 길항 작용을 하는 것을 포함하여, 상기 질환, 장애, 또는 의학적 병태의 개선(ameliorating), 즉 환자에서 상기 질환, 장애, 또는 의학적 병태를 제거 또는 퇴행(regression);(b) ameliorating said disease, disorder, or medical condition, i.e., eliminating or regressing said disease, disorder, or medical condition in a patient, including antagonizing the effect of another therapeutic agent;
(c) 상기 질환, 장애, 또는 의학적 병태의 억제(suppressing), 즉 환자에서 상기 질환, 장애, 또는 의학적 병태의 발전을 둔화 또는 저지; 또는(c) suppressing the disease, disorder, or medical condition, ie, slowing or arresting the development of the disease, disorder, or medical condition in a patient; or
(d) 환자에서 상기 질환, 장애, 또는 의학적 병태의 증상 완화,(d) alleviating the symptoms of said disease, disorder, or medical condition in the patient;
본 개시는 보툴리눔 독소 또는 이를 포함하는 조성물이 엑소좀 분비를 억제함으로써 엑소좀-매개된 질환, 예를 들어 암, 예를 들어 전이성 암, 예를 들어 고형암의 치료제로 유용할 수 있음을 발견한 것에 기초한 것이다.The present disclosure responds to the discovery that botulinum toxin or a composition comprising the same may be useful as a therapeutic agent for exosome-mediated diseases, such as cancer, for example, metastatic cancer, for example, solid cancer by inhibiting exosome secretion. it is based
일 태양에서, 보툴리눔 독소는 혈청형에 제한 없이 사용 가능하며, 타입 A(BoNT/A), B(BoNT/B), C(BoNT/C), D(BoNT/D), E(BoNT/E), F(BoNT/F), G(BoNT/G), H(BoNT/H), X(BoNT/X), 엔테로코커스 종 보툴리눔 독소 J(eBoNT/J) 및 모자이크 보툴리눔 독소 및/또는 그의 변이체로 이루어진 군으로부터 선택될 수 있다. 모자이크 독소의 예는 BoNT/DC, BoNT/CD 및 BoNT/FA를 포함한다. 예를 들어 타입 A를 사용할 수 있다.In one embodiment, the botulinum toxin can be used without limitation of serotype, type A (BoNT/A), B (BoNT/B), C (BoNT/C), D (BoNT/D), E (BoNT/E) ), F(BoNT/F), G(BoNT/G), H(BoNT/H), X(BoNT/X), Enterococcus sp. botulinum toxin J (eBoNT/J) and mosaic botulinum toxin and/or variants thereof It may be selected from the group consisting of. Examples of mosaictoxins include BoNT/DC, BoNT/CD and BoNT/FA. For example, type A can be used.
일 태양에서, 보툴리눔 독소는 다양한 BoNT/A 서브타입, 예를 들어 A1, A2, A3, A4, A5, A6, A7, A9, A10; BoNT/B 서브타입, 예를 들어 B1, B2, B3, B4, B5, B6, B7, B8, Bnp 및 Bbv; BoNT/C 서브타입, 예를 들어 C 및 CD; BoNT/D 서브타입, 예를 들어 D 및 DC; BoNT/E 서브타입, 예를 들어 E1, E2, E3, E4, E5, E6, E7, E8, E9; BoNT/F 서브타입, 예를 들어 F1, F2, F3, F4, F5, F6, F7; 및 BoNT/G 서브타입, 예를 들어 서브타입 G로부터 유래된다. BoNT 서브타입은 키메라 BoNT, 예를 들어 BoNT/DC, BoNT/CD, BoNT/FA 등을 포함한다.In one aspect, the botulinum toxin is selected from various BoNT/A subtypes, eg, A1, A2, A3, A4, A5, A6, A7, A9, A10; BoNT/B subtypes such as B1, B2, B3, B4, B5, B6, B7, B8, Bnp and Bbv; BoNT/C subtypes such as C and CD; BoNT/D subtypes such as D and DC; BoNT/E subtypes such as E1, E2, E3, E4, E5, E6, E7, E8, E9; BoNT/F subtypes such as F1, F2, F3, F4, F5, F6, F7; and from the BoNT/G subtype, eg, subtype G. BoNT subtypes include chimeric BoNTs, such as BoNT/DC, BoNT/CD, BoNT/FA, and the like.
본 개시의 일 측면에 따르면, 보툴리눔 독소 또는 이를 포함하는 조성물은 엑소좀-매개된 질환을 치료하기 위한 것일 수 있다.According to one aspect of the present disclosure, a botulinum toxin or a composition comprising the same may be for treating an exosome-mediated disease.
구체예에서, 엑소좀-매개된 질환은 암, 바이러스 감염, 신경 질환 또는 류마티스성 질환일 수 있으며, 특히 엑소좀-매개된 질환은 암일 수 있다.In an embodiment, the exosome-mediated disease may be cancer, a viral infection, a neurological disease or a rheumatic disease, and in particular the exosome-mediated disease may be cancer.
구체예에서, 암은 전이성 암(metastatic cancer)일 수 있다.In an embodiment, the cancer may be metastatic cancer.
구체예에서, 암은 고형암(solid tumor)일 수 있다. 고형암의 비제한적인 예들은 유방암, 폐암, 두부 또는 경부암, 대장암, 식도암, 후두암, 위암, 간암, 췌장암, 골암, 피부암, 피부 또는 안구내 흑색종, 자궁암, 난소암, 직장암, 항문부근암, 결장암, 유방암, 나팔관암종, 자궁내막암종, 자궁경부암종, 질암종, 음문암종, 호지킨병, 소장암, 내분비선암, 갑상선암, 부갑상선암, 부신암, 연조직 육종, 요도암, 음경암, 전립선암, 만성 또는 급성 백혈병, 림프구 림프종, 방광암, 신장 또는 수뇨관 암, 신장세포 암종, 신장골반 암종, CNS 종양, 1차 CNS 림프종, 척수 종양, 뇌종양, 예를 들어 신경교종(예: 성상세포종(astrocytoma), 교모세포종 (glioblastoma), 핍지교종(oligodendroglioma) 상의세포종(ependymoma), 배아세포종, 수막종, 뇌간신경교종, 뇌하수체 선종, 신경초종, 선천성종양, 두개인두종, 전이성 뇌종양을 포함할 수 있다. 예를 들어, 고형암은 흑색종, 유방암, 폐암, 췌장암, 뇌종양 및 전립선암으로 이루어진 군으로부터 선택되는 하나 이상의 것일 수 있으나, 이들로 제한되는 것은 아니다.In an embodiment, the cancer may be a solid tumor. Non-limiting examples of solid cancer include breast cancer, lung cancer, head or neck cancer, colorectal cancer, esophageal cancer, laryngeal cancer, stomach cancer, liver cancer, pancreatic cancer, bone cancer, skin cancer, skin or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, perianal cancer, Colon cancer, breast cancer, fallopian tube carcinoma, endometrial carcinoma, cervical carcinoma, vaginal carcinoma, vulvar carcinoma, Hodgkin's disease, small intestine cancer, endocrine adenocarcinoma, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, prostate cancer , chronic or acute leukemia, lymphocytic lymphoma, bladder cancer, kidney or ureter cancer, renal cell carcinoma, renal pelvic carcinoma, CNS tumor, primary CNS lymphoma, spinal cord tumor, brain tumor, such as glioma (eg, astrocytoma) , glioblastoma, oligodendroglioma, ependymoma, germ cell tumor, meningioma, brainstem glioma, pituitary adenoma, schwannoma, congenital tumor, craniopharyngoma, metastatic brain tumor. For example, solid cancer may be one or more selected from the group consisting of melanoma, breast cancer, lung cancer, pancreatic cancer, brain tumor and prostate cancer, but is not limited thereto.
일 태양에서, 보툴리눔 독소는 혈청형에 제한 없이 사용 가능하며, 타입 A, B, C, D, E, F, G, H, 또는 이들의 조합, 예를 들어 타입 A를 사용할 수 있다.In one embodiment, the botulinum toxin can be used without limitation of serotype, and type A, B, C, D, E, F, G, H, or a combination thereof, for example type A, can be used.
일 태양에서, 보툴리눔 독소를 포함하는 조성물은 하나 이상의 약제학적으로 허용되는 부형제 또는 첨가제를 추가로 포함할 수 있다.In one aspect, the composition comprising botulinum toxin may further comprise one or more pharmaceutically acceptable excipients or additives.
약제학적으로 허용되는 부형제 또는 첨가제는 안정화제, 이온 화합물(ionic compound), 계면활성제, 완충제, 동결건조 보호제, 또는 이들의 조합, 예를 들어 아미노산(예를 들어 메티오닌), 염(예를 들어 NaCl), 완충액, 비이온성 계면활성제(예를 들어 폴리소르베이트, 예를 들어 폴리소르베이트 20), 당(예를 들어 백당 등과 같은 다이사카라이드), 당 알코올(예를 들어 소르비톨), 또는 이들의 조합일 수 있다.Pharmaceutically acceptable excipients or additives include stabilizers, ionic compounds, surfactants, buffers, lyophilization protectants, or combinations thereof, such as amino acids (eg methionine), salts (eg NaCl). ), buffers, nonionic surfactants (e.g. polysorbate, e.g. polysorbate 20), sugars (e.g. disaccharides such as sucrose), sugar alcohols (e.g. sorbitol), or their It can be a combination.
상기 조성물은 알부민 또는 동물 유래 성분이나 폴리사카라이드를 포함하지 않는 것일 수 있다.The composition may not contain albumin or animal-derived components or polysaccharides.
상기 조성물은 예를 들어 WO2009-008595 A1 또는 WO2012-134240 A2에 개시되어 있는 것들을 포함하며, 상기 특허의 내용은 그 전체로 본 명세서에서 참조로 인용된다.Said compositions include, for example, those disclosed in WO2009-008595 A1 or WO2012-134240 A2, the contents of which are incorporated herein by reference in their entirety.
상기 조성물은 상업적으로 입수가능한 보툴리눔 독소 함유 제약 조성물을 제한 없이 포함하며, 예를 들어 보톡스(BOTOX)®(인간 혈청 알부민 및 염화나트륨과의 보툴리눔 독소 타입 A 신경독소 복합체)(알러간, 인코포레이티드 (Allergan, Inc: 미국 캘리포니아주 얼바인)), 디스포트(DYSPORT)®(사용 전 0.9% 염화나트륨으로 재구성되는 분제로서, 제제 중 인간 혈청 알부민 및 락토스와의 클로스트리디움 보툴리눔 타입 A 독소 헤마글루티닌 복합체)(입센 리미티드 (Ipsen Limited: 영국 버크셔)), 미오블록(MYOBLOC)™(보툴리눔 독소 타입 B, 인간 혈청 알부민, 숙신산 나트륨, 및 염화나트륨을 포함하는 주사액(약 pH 56))(솔스티스 뉴로사이언시즈, 인크.(Solstice Neurosciences, Inc: 미국 캘리포니아주 사우스 샌프란시스코)), 메디톡신®(클로스트리디움 보툴리눔독소 A형, 사람혈청알부민 및 염화나트륨을 포함하는 동결건조 분말 제제)(㈜메디톡스, 한국), 이노톡스®(클로스트리디움 보툴리눔독소 A형, L-메티오닌, 폴리소르베이트20, 염화나트륨 및 주사용수를 포함하는 액상 제제)(㈜메디톡스, 한국), 코어톡스®(클로스트리디움 보툴리눔독소 A형(150kD), 폴리소르베이트20, 백당 및 염화나트륨을 포함하는 동결건조 분말 제제)(㈜메디톡스, 한국)를 포함할 수 있다. 구체예에서, 상기 조성물은 메디톡신®, 이노톡스®, 코어톡스® 또는 이들의 하나 이상의 조합일 수 있으나, 이들로 제한되는 것은 아니다.The compositions include, without limitation, commercially available pharmaceutical compositions containing botulinum toxin, such as, for example, BOTOX® (botulinum toxin type A neurotoxin complex with human serum albumin and sodium chloride) (Allergan, Inc.) (Allergan, Inc: Irvine, CA, USA), DYSPORT® (a powder reconstituted with 0.9% sodium chloride prior to use, Clostridium botulinum type A toxin hemagglutinin with human serum albumin and lactose in the formulation complex) (Ipsen Limited (Berkshire, UK)), MYOBLOC™ (injection solution containing botulinum toxin type B, human serum albumin, sodium succinate, and sodium chloride (approximately pH 56)) (Solstice Neurosciences) , Inc. (Solstice Neurosciences, Inc: South San Francisco, CA, USA)), Medytoxin® (a lyophilized powder formulation containing Clostridium botulinum toxin type A, human serum albumin and sodium chloride) (Medytox, Korea), Inno Tox® (Liquid formulation containing Clostridium botulinum toxin type A, L-methionine, polysorbate 20, sodium chloride and water for injection) (Medytox, Korea), Coretox® (Clostridium botulinum toxin type A (150kD) ), polysorbate 20, lyophilized powder formulation containing sucrose and sodium chloride) (Medytox, Korea). In an embodiment, the composition may be Medytoxin®, Innotox®, Coretox®, or one or more combinations thereof, but is not limited thereto.
일 태양에서, 상기 조성물은 고체 또는 액체 제제, 예를 들어 동결건조 분말, 액상, 또는 프리필드 시린지(pre-filled syringe) 제제 등 어떠한 형태로도 제제화될 수 있다.In one embodiment, the composition may be formulated in any form, such as a solid or liquid formulation, for example, a lyophilized powder, a liquid, or a pre-filled syringe formulation.
일 태양에서, 상기 조성물은 국소로 투여하기 위한 제제일 수 있다. 예를 들어, 국소 투여는 종양 내 투여 또는 종양 주변 투여일 수 있으며, 전형적으로 보툴리눔 독소 수용액의 종양 내 주사(intratumoral injection) 또는 종양 주변 주사(peritumoral injection)에 의해 이루어질 수 있다. 예를 들어, 제제는 피하 또는 연부조직, 예를 들어 근육 내로 투여될 수 있다. 상기 조성물은 특정 위치의 종양 내 또는 주변 투여에 의해 상기 위치로부터 떨어진 위치에서 종양 성장을 억제하거나 종양 발생을 저지할 수 있는바, 상기 조성물은 국소 투여로 전신적인 효과를 나타낼 수 있다.In one embodiment, the composition may be a formulation for topical administration. For example, topical administration may be intratumoral administration or peritumoral administration, and typically may be accomplished by intratumoral injection or peritumoral injection of an aqueous botulinum toxin solution. For example, the formulation can be administered subcutaneously or into a soft tissue, eg, intramuscularly. The composition can inhibit tumor growth or inhibit tumor development at a location away from the location by intratumoral or peripheral administration at a specific location, and the composition can exhibit systemic effects when administered locally.
일 태양에서, 보툴리눔 독소의 치료 유효량은 약 0.01 U/㎏ 내지 약 100 U/㎏, 약 0.1 U/㎏ 내지 약 100 U/㎏, 약 0.2 U/㎏ 내지 약 100 U/㎏, 약 0.2 U/㎏ 내지 약 50 U/㎏, 약 0.2 U/㎏ 내지 약 30 U/㎏, 약 0.2 U/㎏ 내지 약 10 U/㎏, 약 0.2 U/㎏ 내지 약 1 U/㎏이다. 다른 태양에서, 체중 60 ㎏ 성인 기준으로 약 1 U 내지 약 10,000 U, 약 1 U 내지 약 5,000 U, 약 1 U 내지 약 2,500 U, 약 1 U 내지 약 1,000 U, 약 1 U 내지 약 500 U, 약 1 U 내지 약 300 U, 약 1 U 내지 약 200 U, 약 10 U 내지 약 200 U, 약 10 U 내지 약 100 U, 약 10 U 내지 약 50 U일 수 있다.In one aspect, a therapeutically effective amount of botulinum toxin is about 0.01 U/kg to about 100 U/kg, about 0.1 U/kg to about 100 U/kg, about 0.2 U/kg to about 100 U/kg, about 0.2 U/kg kg to about 50 U/kg, from about 0.2 U/kg to about 30 U/kg, from about 0.2 U/kg to about 10 U/kg, from about 0.2 U/kg to about 1 U/kg. In another aspect, based on an adult weighing 60 kg, from about 1 U to about 10,000 U, from about 1 U to about 5,000 U, from about 1 U to about 2,500 U, from about 1 U to about 1,000 U, from about 1 U to about 500 U, about 1 U to about 300 U, about 1 U to about 200 U, about 10 U to about 200 U, about 10 U to about 100 U, about 10 U to about 50 U.
본 명세서에서 사용되는, "유닛", "unit(s)", 또는 "U"라는 용어는 보툴리눔 독소 주사를 맞은 마우스 중 50%를 사멸시킨 보툴리눔 독소의 양으로서 정의되는, LD50 용량을 지칭하는 것으로, 한 제품 내에서 호환 가능하게 사용된다.As used herein, the term "unit", "unit(s)", or "U" refers to the LD50 dose, defined as the amount of botulinum toxin that killed 50% of mice receiving a botulinum toxin injection. , are used interchangeably within a product.
일 태양에서, 보툴리눔 독소는 단회 또는 다회 치료 세션으로 투여될 수 있다. 투여량은 주사 부위에 단일 또는 분할 주입으로 투여될 수 있다. 다회 치료 세션에서 보툴리눔 독소는 6개월 이하의 간격으로 투여할 수 있다. 다회 치료 세션에서 보툴리눔 독소의 투여간격은 1차 치료와 2차 치료를 포함하며, 2차 치료의 투여량은 1차 치료의 투여량보다 적거나, 많거나 또는 동일할 수 있다.In one aspect, the botulinum toxin may be administered in single or multiple treatment sessions. Dosages may be administered as single or divided injections at the site of injection. In multiple treatment sessions, botulinum toxin may be administered at intervals of up to 6 months. In the multiple treatment session, the administration interval of the botulinum toxin includes the first treatment and the second treatment, and the dose of the second treatment may be less than, more than, or the same as the dose of the first treatment.
보툴리눔 독소의 투여 용량, 투여 시간, 투여 방법, 투여 기간 또는 간격 등은 환자의 체중, 연령, 성별, 건강상태, 질환의 중증도 등에 따라 통상의 기술자가 적절히 선택하여 사용할 수 있다.The dose, administration time, administration method, administration period or interval, etc. of the botulinum toxin may be appropriately selected and used by a person skilled in the art according to the patient's weight, age, sex, health condition, severity of disease, and the like.
일 태양에서, 보툴리눔 독소 또는 이를 포함하는 조성물은 엑소좀 분비가 증가된 대상에서 사용할 수 있다.In one embodiment, botulinum toxin or a composition comprising the same can be used in subjects with increased exosome secretion.
일 태양에서, 엑소좀-매개된 질환을 치료하는 방법은 하기 단계를 포함하는 것일 수 있다:In one aspect, a method of treating an exosome-mediated disease may comprise the steps of:
1) 대상으로부터 수득된 생물학적 시료에서 분비된 엑소좀을 정량하는 단계; 및,1) quantifying the exosomes secreted from the biological sample obtained from the subject; and,
2) 엑소좀 분비가 증가된 대상에게 치료 유효량의 보툴리눔 독소 또는 이를 포함하는 조성물을 투여하는 단계.2) administering a therapeutically effective amount of a botulinum toxin or a composition comprising the same to a subject with increased exosome secretion.
분비된 엑소좀은 당업계에 알려져 있는 통상적인 방법에 따라, 대상의 생물학적 시료, 예를 들어 혈액으로부터 분리하여 정량할 수 있으며, 시료의 종류, 분리 및 정량 방법에 특별한 제한이 있는 것은 아니다.The secreted exosomes can be separated and quantified from a biological sample of a subject, for example, blood according to a conventional method known in the art, and there is no particular limitation on the type of sample, separation and quantification method.
보툴리눔 독소 또는 이를 포함하는 조성물이 투여되는 대상은 인간이나 동물, 예를 들어 인간, 돼지, 개, 고양이, 소, 말, 쥐 등을 제한 없이 포함할 수 있다.The subject to which the botulinum toxin or a composition comprising the same is administered may include, without limitation, a human or an animal, for example, a human, a pig, a dog, a cat, a cow, a horse, a rat, and the like.
일 태양에서, 보툴리눔 독소 또는 이를 포함하는 조성물은 1종 이상의 다른 약물 또는 요법과 병용 투여될 수 있다. 본 명세서에서, "병용 투여"는 사전에 투여된 치료제가 체내에서 여전히 유효한 동안 제2 치료제가 투여되도록 하는 2종 이상의 상이한 치료제의 임의의 형태를 가리킨다. 예를 들어, 2종의 치료제는 대상에서 동시에 유효하며, 이는 2종의 치료제의 상승 효과를 포함할 수 있다. 상이한 치료제는 동일한 제제 또는 별개의 제제로 동시에 또는 순차적으로 투여될 수 있다.In one aspect, botulinum toxin or a composition comprising the same may be administered in combination with one or more other drugs or therapies. As used herein, "combination administration" refers to any form of two or more different therapeutic agents that allow a second therapeutic agent to be administered while the previously administered therapeutic agent is still effective in the body. For example, two therapeutic agents are simultaneously effective in a subject, which may include a synergistic effect of the two therapeutic agents. The different therapeutic agents may be administered simultaneously or sequentially in the same formulation or in separate formulations.
구체예에서, 보툴리눔 독소 또는 이를 포함하는 조성물은 다른 치료제, 예를 들어 항암제, 항바이러스제, 사이토카인 또는 면역 작용제와 병용 투여될 수 있다.In an embodiment, botulinum toxin or a composition comprising the same may be administered in combination with other therapeutic agents, for example, anticancer agents, antiviral agents, cytokines, or immune agents.
보툴리눔 독소 또는 이를 포함하는 조성물은 화학치료제와 병용하여 투여될 수 있다. 화학치료제의 비제한적인 예는 알킬화제, 나이트로소우레아제, 항대사물질, 항암 항생제, 식물-기원 알칼로이드, 토포아이소머라제 저해제, 호르몬 약물, 호르몬 길항제, 백혈구감소증(호중구감소증) 치료 약물, 혈소판감소증 치료 약물, 항구토제, 아로마타제 저해제, P-당단백질 저해제, 백금 착물 유도체, 다른 면역치료 약물 및 다른 항암 약물을 포함한다. 병용 투여될 수 있는 예시적인 세포독성제는 항미세소관 작용제, 토포아이소머라제 저해제, 항대사물질, 유사분열 저해제, 알킬화제, 안트라사이클린, 빈카 알카로이드, 인터칼레이팅제, 신호 전달 경로를 방해할 수 있는 작용제, 아포토시스를 촉진하는 작용제, 프로테오좀 저해제 및 방사선(국소 또는 전신 조사)을 포함한다. 추가적인 치료제의 비제한적인 예는 펩타이드, 폴리펩타이드, 단백질, 융합 단백질, 핵산 분자, 소분자, 모방 작용제, 합성 약물, 무기 분자 및 유기 분자를 포함하지만 이들로 제한되지 않는다.The botulinum toxin or a composition comprising the same may be administered in combination with a chemotherapeutic agent. Non-limiting examples of chemotherapeutic agents include alkylating agents, nitrosourases, antimetabolites, anticancer drugs, plant-derived alkaloids, topoisomerase inhibitors, hormonal drugs, hormone antagonists, leukopenia (neutropenia) therapeutic drugs, thrombocytopenia. therapeutic drugs, antiemetics, aromatase inhibitors, P-glycoprotein inhibitors, platinum complex derivatives, other immunotherapeutic drugs, and other anticancer drugs. Exemplary cytotoxic agents that may be administered in combination include anti-microtubule agents, topoisomerase inhibitors, antimetabolites, mitotic inhibitors, alkylating agents, anthracyclines, vinca alkaloids, intercalating agents, and may interfere with signal transduction pathways. agents that promote apoptosis, proteosome inhibitors, and radiation (local or systemic irradiation). Non-limiting examples of additional therapeutic agents include, but are not limited to, peptides, polypeptides, proteins, fusion proteins, nucleic acid molecules, small molecules, mimetic agents, synthetic drugs, inorganic molecules, and organic molecules.
보툴리눔 독소 또는 이를 포함하는 조성물은 면역요법제와 병용하여 투여될 수 있다. 일 태양에서, 면역요법제는 당업계에 알려져 있는 어떠한 물질이나 요법도 사용 가능하며, 사이토카인, 암 백신, 항암 바이러스(oncolytic virus), 단일클론 항체, 비사이토카인 보조제, 면역 세포(T 세포, NK 세포, 수지상 세포, B 세포 등), 면역관문 억제제를 포함할 수 있다. 구체예에서, 면역요법제는 면역관문 억제제이다. 면역관문 억제제는 펩티드, 항체, 핵산 분자 및 소형 분자를 포함한다. 예를 들어, 면역관문 억제제는 대상에서 CD8+ T 세포의 증식성, 이동성, 지속성 및/또는 세포독성 활성, 및 특히 대상의 CD8+ T 세포의 종양 침윤성을 증강시키기 위해 투여될 수 있다. 전형적으로, 면역관문 억제제는 활성화된 T 림프구, 예를 들어 세포독성 T 림프구-연관 단백질 4(CTLA4) 및 프로그램된 세포사멸 1(PD-1), 또는 살해 세포 면역글로불린-유사 수용체(KIR) 패밀리의 다양한 구성원과 같은, NK 세포에 의해 발현되는 면역 억제성 수용체를 차단하는 길항제이거나, 이들 수용체의 주요 리간드, 예를 들어 PD-1 리간드 CD274(PD-L1 또는 B7-H1로 가장 잘 알려짐)를 차단하는 길항제이다. 예를 들어, 면역관문 억제제는 항체 또는 이의 항원-결합 단편이다. 구체적으로, 면역관문 억제제는 항-PD-1 항체, 항-PD-L1 항체, 항-PD-L2 항체, 항-CTLA-4 항체, 항-TIM-3 항체, 항-LAG3 항체, 항-IDO1 항체, 항-TIGIT 항체, 항-B7H3 항체, 항-B7H4 항체, 항-BTLA 항체, 및 항-B7H6 항체, 및 이의 항원-결합 단편으로 이루어진 군으로부터 선택되는 하나 이상일 수 있다. 보다 구체적으로, 면역관문 억제제는 이필리무맙(Ipilimumab)(여보이®, BMS/오노), 트레멜리무맙(Tremelimumab)(아스트라제네카), 아테졸리주맙(atezolizumab)(티쎈트릭®, 로슈), 니볼루맙(nivolumab)(옵디보®, BMS/오노), 펨브롤리주맙(Pembrolizumab)(키트루다®, MSD), 아벨루맙(Avelumab)(바벤시오®, 화이자/독일머크), 더발루맙(Durvalumab)(임핀지®, 아스트라제네카/메드이뮨), 및 이의 항원-결합 단편 중에서 선택되는 하나 이상일 수 있으나, 이들로 제한되는 것은 아니다.The botulinum toxin or a composition comprising the same may be administered in combination with an immunotherapeutic agent. In one embodiment, the immunotherapeutic agent may use any substance or therapy known in the art, and may include cytokines, cancer vaccines, oncolytic viruses, monoclonal antibodies, non-cytokine adjuvants, immune cells (T cells, NK cells, dendritic cells, B cells, etc.), and immune checkpoint inhibitors. In an embodiment, the immunotherapeutic agent is an immune checkpoint inhibitor. Immune checkpoint inhibitors include peptides, antibodies, nucleic acid molecules and small molecules. For example, an immune checkpoint inhibitor may be administered to enhance the proliferative, migratory, persistence and/or cytotoxic activity of CD8+ T cells in a subject, and particularly tumor invasiveness of CD8+ T cells in a subject. Typically, checkpoint inhibitors are activated T lymphocytes, such as cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PD-1), or the killer cell immunoglobulin-like receptor (KIR) family. antagonists that block immunosuppressive receptors expressed by NK cells, such as various members of It is a blocking antagonist. For example, an immune checkpoint inhibitor is an antibody or antigen-binding fragment thereof. Specifically, the checkpoint inhibitor is an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-TIM-3 antibody, an anti-LAG3 antibody, an anti-IDO1 antibody. antibody, anti-TIGIT antibody, anti-B7H3 antibody, anti-B7H4 antibody, anti-BTLA antibody, and anti-B7H6 antibody, and antigen-binding fragment thereof. More specifically, immune checkpoint inhibitors include Ipilimumab (Yervoy®, BMS/Ono), Tremelimumab (AstraZeneca), atezolizumab (Tecentric®, Roche), and nivolumab (nivolumab) (Opdivo®, BMS/Ono), Pembrolizumab (Keytruda®, MSD), Avelumab (Bavencio®, Pfizer/Merck Germany), Durvalumab ( Imfinzi®, AstraZeneca/MedImmune), and antigen-binding fragments thereof may be at least one selected from, but not limited to.
다른 치료제는 CTLA-4 길항제, PD-1 길항제, PD-L1 길항제, PD-L2 길항제, 또는 EGFR 길항제를 포함할 수 있다.The other therapeutic agent may include a CTLA-4 antagonist, a PD-1 antagonist, a PD-L1 antagonist, a PD-L2 antagonist, or an EGFR antagonist.
본 개시에 따르면, 보툴리눔 독소 또는 이를 포함하는 조성물은 엑소좀 분비를 억제함으로써 엑소좀-매개된 질환, 예를 들어 암, 예를 들어 전이성 암, 예를 들어 고형암의 치료제로 유용할 수 있다.According to the present disclosure, a botulinum toxin or a composition comprising the same may be useful as a therapeutic agent for an exosome-mediated disease, for example, a cancer, for example, a metastatic cancer, for example, a solid cancer by inhibiting exosome secretion.
도 1은 마우스 악성 흑색종 이식 모델에서 보툴리눔 독소 투여 후 경시적인 종양 성장을 보여주는 그래프이다.1 is a graph showing tumor growth over time after administration of botulinum toxin in a mouse malignant melanoma transplantation model.
도 2는 마우스 악성 흑색종 이식 모델에서 보툴리눔 독소 투여 후 투여 용량별 폐 전이성 병소의 수를 보여주는 그래프이다.2 is a graph showing the number of lung metastatic lesions by dose after administration of botulinum toxin in a mouse malignant melanoma transplant model.
도 3a는 마우스 악성 흑색종 이식 모델에서 보툴리눔 독소 투여 후 투여 용량별 혈액 내 CCL2 수치를 나타낸 그래프이다.3A is a graph showing CCL2 levels in blood for each dose administered after administration of botulinum toxin in a mouse malignant melanoma transplantation model.
도 3b는 마우스 악성 흑색종 이식 모델에서 보툴리눔 독소 투여 후 투여 용량별 혈액 내 IL-6 수치를 나타낸 그래프이다.Figure 3b is a graph showing the level of IL-6 in the blood for each dose administered after botulinum toxin administration in a mouse malignant melanoma transplantation model.
도 3c는 마우스 악성 흑색종 이식 모델에서 보툴리눔 독소 투여 후 투여 용량별 혈액 내 TNF 수치를 나타낸 그래프이다.FIG. 3c is a graph showing TNF levels in blood for each dose administered after administration of botulinum toxin in a mouse malignant melanoma transplantation model.
도 4는 마우스 유선 종양 이식 모델에서 보툴리눔 독소 투여 후 경시적인 종양 성장을 보여주는 그래프이다.4 is a graph showing tumor growth over time after administration of botulinum toxin in a mouse mammary gland tumor transplantation model.
도 5는 마우스 유선 종양 이식 모델에서 보툴리눔 독소 투여 후 투여 용량별 혈액 내 엑소좀 수를 나타낸 그래프이다.5 is a graph showing the number of exosomes in blood for each dose administered after botulinum toxin administration in a mouse mammary gland tumor transplantation model.
도 6a는 마우스 악성 흑색종 세포주(B16-F10)에서 보툴리눔 독소의 농도별 처리에 의한 성장 저해 효과를 보여주는 그래프이다.FIG. 6A is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a mouse malignant melanoma cell line (B16-F10).
도 6b는 인간 폐암 세포주(A549)에서 보툴리눔 독소의 농도별 처리에 의한 성장 저해 효과를 보여주는 그래프이다.6B is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a human lung cancer cell line (A549).
도 6c는 인간 췌장암 세포주(SNU324)에서 보툴리눔 독소의 농도별 처리에 의한 성장 저해 효과를 보여주는 그래프이다.FIG. 6c is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a human pancreatic cancer cell line (SNU324).
도 6d는 인간 췌장암 세포주(Capan-2)에서 보툴리눔 독소의 농도별 처리에 의한 성장 저해 효과를 보여주는 그래프이다.6D is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a human pancreatic cancer cell line (Capan-2).
도 7은 마우스 흑색종에서 보툴리눔 독소의 처리에 의한 엑소좀 감소 효과를 보여주는 그래프이다.7 is a graph showing the effect of reducing exosomes by treatment with botulinum toxin in mouse melanoma.
도 8은 인간 췌장암 세포주에서 보툴리눔 독소의 처리에 의한 엑소좀 감소 효과를 보여주는 그래프이다.8 is a graph showing the effect of reducing exosomes by treatment with botulinum toxin in a human pancreatic cancer cell line.
도 9a는 인간 뇌암 세포주(U87MG)에서 보툴리눔 독소의 농도별 처리에 의한 성장 저해 효과를 보여주는 그래프이다.9A is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a human brain cancer cell line (U87MG).
도 9b는 인간 전립선암 세포주(PC3)에서 보툴리눔 독소의 농도별 처리에 의한 성장 저해 효과를 보여주는 그래프이다.9B is a graph showing the growth inhibitory effect of each concentration of botulinum toxin in a human prostate cancer cell line (PC3).
이하 본 개시를 실시예를 들어 보다 상세히 설명하나, 이는 본 개시를 설명하기 위한 것일 뿐 그 범위를 어떤 식으로든지 제한하고자 하는 것은 아니다.Hereinafter, the present disclosure will be described in more detail with reference to examples, but this is only for explaining the present disclosure and is not intended to limit the scope in any way.
실시예 1: 마우스 악성 흑색종 이식 모델에서 보툴리눔 독소의 항암 효능 시험Example 1: Anticancer efficacy test of botulinum toxin in mouse malignant melanoma transplantation model
수컷 6주령 C57BL/6(구입처: 오리엔트바이오) 마우스를 구입하여 1주간 순화 및 검역 후 7주령 마우스를 실험에 사용하였다. 사료는 멸균된 실험동물용 고형사료(R40-10, SAFE, France)를 자유 급이하였으며, 음수는 상수도수를 고온고압멸균(autoclave)하여 자유 급이하였다. 순화, 검역 및 실험기간 동안 마우스는 온도 23±3℃, 상대습도 55±15%, 조명시간 12시간(오전8시~오후8시), 환기횟수 15회/시간 및 조도 150~300 Lux로 설정된 특정-병원체-부재 조건하에서 사육되었다. 본 실험은 ㈜메디톡스 동물실험윤리위원회의 검토 및 승인(A-2020-004) 후 수행되었다.Male 6-week-old C57BL/6 (place of purchase: Orient Bio) mice were purchased, acclimatized and quarantined for 1 week, and 7-week-old mice were used for the experiment. For feed, sterilized solid feed for laboratory animals (R40-10, SAFE, France) was freely fed, and negative water was fed freely by autoclaving tap water at high temperature. During the acclimatization, quarantine, and experiment period, the mouse was set at a temperature of 23±3℃, relative humidity of 55±15%, lighting time of 12 hours (8am to 8pm), ventilation frequency of 15 times/hour, and illumination intensity of 150~300 Lux. Raised under specific-pathogen-free conditions. This experiment was conducted after review and approval (A-2020-004) by the Animal Experimental Ethics Committee of Medytox Co., Ltd.
B16-F10 마우스 악성 흑색종 세포주(KCLB No: 80008)를 한국세포주 은행(KCLB: Korean Cell Line Bank)으로부터 수득하였다. B16-F10 세포를 10% FBS(CAT No: 10082-147, Gibco) 및 1% 페니실린-스트렙토마이신(CAT No: 15140-122, Gibco) 함유 DMEM 배지(CAT No: 11965-092, Gibco)에서 37 ℃, 5% CO2 조건으로 인큐베이터에서 배양하였다. 마취된 C57BL/6 마우스 우측 옆구리에 주사기를 이용하여 5×105 세포를 0.1 ㎖ 이식하였다.A B16-F10 mouse malignant melanoma cell line (KCLB No: 80008) was obtained from the Korean Cell Line Bank (KCLB). B16-F10 cells were cultured in DMEM medium (CAT No: 11965-092, Gibco) containing 10% FBS (CAT No: 10082-147, Gibco) and 1% penicillin-streptomycin (CAT No: 15140-122, Gibco). ℃, 5% CO 2 Conditions were cultured in an incubator. 0.1 ml of 5×10 5 cells were transplanted into the right flank of anesthetized C57BL/6 mice using a syringe.
비히클로서 보툴리눔 독소 조제에 사용된 멸균 생리식염수(Lot No: M8T7AF3, 대한약품공업)를 사용하였으며, 보툴리눔 독소로는 100 units 코어톡스 (Lot No: NSA19007A, 메디톡스)를 사용하였다. 아래 표 1에 나타낸 바에 따라 각 시험물질을 조제하여 각 군에 투여하였다.As a vehicle, sterile physiological saline (Lot No: M8T7AF3, Daehan Pharmaceutical Co., Ltd.) used for the preparation of botulinum toxin was used, and as a botulinum toxin, 100 units Coretox (Lot No: NSA19007A, Medytox) was used. As shown in Table 1 below, each test substance was prepared and administered to each group.
army 동물수number of animals 투여물질substance to be administered 용량Volume 투여경로route of administration 투여빈도Dosing frequency
1One 1010 비히클vehicle 1 ㎖/㎏1 ml/kg 종양 내within the tumor 1회(종양 이식 후 8일째)Once (8 days after tumor transplantation)
22 1010 보툴리눔 독소botulinum toxin 1.5 units/㎏1.5 units/kg 종양 내within the tumor 1회(종양 이식 후 8일째)Once (8 days after tumor transplantation)
33 1010 보툴리눔 독소 botulinum toxin 5 units/㎏5 units/kg 종양 내within the tumor 1회(종양 이식 후 8일째)Once (8 days after tumor transplantation)
44 8* 8 * 보툴리눔 독소 botulinum toxin 15 units/㎏15 units/kg 종양 내within the tumor 1회(종양 이식 후 8일째)Once (8 days after tumor transplantation)
55 1010 보툴리눔 독소 botulinum toxin 50 units/㎏50 units/kg 종양 내within the tumor 1회(종양 이식 후 8일째)Once (8 days after tumor transplantation)
* 2마리 마우스 제외 (개체간 다툼으로 종양 주변 상처 발생) * Excluding 2 mice (injuries around tumors caused by fighting between individuals)
마우스 악성 흑색종 이식 후 8일째 체중 및 종양 크기를 측정하여 평균 약 55 ㎣ 크기에서 마우스를 시험군에 따라 각 그룹 당 10마리씩 시험군간 차이가 없게 무작위 배정하였다. 종양 이식 후 8일째 체중을 바탕으로 비히클 및 보툴리눔 독소를 시험군 구성에 따라 종양 내로 투여하였다. 종양 이식 후 주 2회 종양 크기를 측정하였다. 종양 크기는 버니어 캘리퍼로 측정하였고 측정치를 사용해 다음의 식을 이용하여 종양 부피를 계산하였다.On the 8th day after transplantation of malignant melanoma in mice, the body weight and tumor size were measured, and mice were randomly assigned to each group at an average size of about 55 mm 3 without any difference between the test groups. On the 8th day after tumor transplantation, vehicle and botulinum toxin were administered intratumorally according to the composition of the test group based on body weight. Tumor size was measured twice a week after tumor transplantation. The tumor size was measured with a vernier caliper and the tumor volume was calculated using the following equation using the measurements.
종양 부피(㎣)=(W2×L)/2Tumor volume (mm3)=(W 2 ×L)/2
W(㎜)=너비(종양의 단축), L(㎜)=종양의 길이(종양의 장축)W (mm) = width (short axis of tumor), L (mm) = length of tumor (long axis of tumor)
종양성장 억제율(TGI; tumor growth inhibition)(%)=(1-평균종양크기시험군/평균종양크기비히클)×100Tumor growth inhibition (TGI; tumor growth inhibition) (%) = (1-Mean tumor size test group / Mean tumor size vehicle )×100
종양 이식 후 19일째, 종양의 크기가 2000 ㎣에 도달하기 전 생존 개체의 혈액을 EDTA 튜브를 이용하여 채취하였고 모든 동물을 안락사하였다. 폐 전이 분석을 위해 폐 조직 적출 후 무게를 측정하고 폐 전이성 병소의 수를 측정하였다. 혈액 내 염증성 사이토카인 CCL2(C-C Motif Chemokine Ligand 2), IL-6(interleukin 6), TNF(tumor necrosis factor)을 BD CBA mouse inflammation kit(Cat No: 552364, Lot No: 0031162, BD)을 이용하여 측정하였다. Graphpad Prism(version 7.05, GraphPad Software Inc, CA, USA)을 그래프 제시에 사용하였으며, SPSS software(version 25.0, SPSS Inc., IL, USA)를 통계 분석에 사용하였다. 모수 데이터는 양측(two-tailed) t-검정을 수행하였고 비모수 데이터는 Mann-Whitney 검정 통계분석을 유의 수준 p = 0.05에서 수행하였다.On the 19th day after tumor transplantation, before the tumor size reached 2000 mm 3 , blood was collected from surviving individuals using an EDTA tube, and all animals were euthanized. For lung metastasis analysis, after lung tissue removal, the weight was measured and the number of lung metastatic lesions was measured. Blood inflammatory cytokines CCL2 (CC Motif Chemokine Ligand 2), IL-6 (interleukin 6), and TNF (tumor necrosis factor) were analyzed using the BD CBA mouse inflammation kit (Cat No: 552364, Lot No: 0031162, BD). measured. Graphpad Prism (version 7.05, GraphPad Software Inc, CA, USA) was used for graph presentation, and SPSS software (version 25.0, SPSS Inc., IL, USA) was used for statistical analysis. For parametric data, a two-tailed t -test was performed, and for non-parametric data, statistical analysis was performed by Mann-Whitney test at a significance level of p = 0.05.
종양 성장 억제율 측정 결과를 표 2 및 도 1에 나타내었다.The measurement results of the tumor growth inhibition rate are shown in Table 2 and FIG. 1 .
army 투여물질substance to be administered 용량Volume 평균 종양 크기1)
(평균 ㎣±SEM)
Average tumor size 1)
(average ㎣±SEM)
TGI(%)1) TGI (%) 1)
1One 비히클vehicle 1 ㎖/㎏1 ml/kg 1705±2691705±269 00
22 보툴리눔 독소botulinum toxin 1.5 units/㎏1.5 units/kg 1150±2661150±266 3333
33 보툴리눔 독소 botulinum toxin 5 units/㎏5 units/kg 1336±2081336±208 2222
44 보툴리눔 독소 botulinum toxin 15 units/㎏15 units/kg 1195±2551195±255 3030
55 보툴리눔 독소 botulinum toxin 50 units/㎏50 units/kg 682±135** 682±135 ** 6060
1) 종양 이식 후 18일째의 결과, ** p<0.01, vs. 비히클. 양측 t-검정. 1) Results on day 18 after tumor transplantation, ** p <0.01, vs. vehicle. Two -sided t -test.
표 2 및 도 1에 나타낸 바와 같이, 마우스 악성 흑색종 이식 후 18일째, 1.5 units/㎏ 보툴리눔 독소 단독 종양 내 투여에 의해 평균 1150 ㎣의 종양 크기와 33%의 종양 성장 억제율을 보였으며, 5 units/㎏ 투여군에서는 평균 1336 ㎣, 22%, 15 units/㎏ 투여군에서는 평균 1195 ㎣, 30%, 50 units/㎏ 투여군에서는 평균 682 ㎣, 60% 종양 성장 억제율을 보였다.As shown in Table 2 and FIG. 1 , on the 18th day after mouse malignant melanoma transplantation, intratumoral administration of 1.5 units/kg botulinum toxin alone showed an average tumor size of 1150 mm 3 and a tumor growth inhibition rate of 33%, and 5 units In the /kg administration group, the average 1336 mm3, 22%, in the 15 units/kg administration group, the average 1195 mm3, 30%, and 50 units/kg administration group showed an average 682 mm3 and 60% tumor growth inhibition rate.
폐 전이성 병소 수 측정 결과를 도 2에 나타내었다. 도 2에 나타낸 바와 같이, 1.5, 15, 50 units/㎏ 투여군에서 비히클 투여군 대비 폐 전이성 병소의 수가 유의적으로 감소되었다.The results of measuring the number of lung metastatic lesions are shown in FIG. 2 . As shown in FIG. 2 , the number of lung metastatic lesions was significantly reduced in the 1.5, 15, and 50 units/kg administration groups compared to the vehicle administration group.
혈액 내 염증성 사이토카인을 분석한 결과, CCL2는 15 units/㎏ 투여군에서 비히클 투여군 대비 감소되었으며(도 3a), TNF 는 5, 50 units/㎏ 투여군에서 유의적으로 감소되었다(도 3b). 혈액 내 IL-6 또한 보툴리눔 독소 투여에 의해 감소된 것이 확인되었다(도 3c).As a result of analyzing inflammatory cytokines in the blood, CCL2 was decreased in the 15 units/kg administration group compared to the vehicle administration group ( FIG. 3a ), and TNF was significantly reduced in the 5 and 50 units/kg administration group ( FIG. 3b ). It was confirmed that IL-6 in the blood was also reduced by administration of botulinum toxin ( FIG. 3C ).
실시예 2: 마우스 유선 종양 이식 모델에서 보툴리눔 독소의 항암 효능 시험Example 2: Anticancer efficacy test of botulinum toxin in mouse mammary tumor transplantation model
암컷 6주령 BALB/C 마우스(구입처: 오리엔트바이오)를 구입하여 1주간 순화 및 검역 후 7주령 마우스를 실험에 사용하였다. 사료, 음수, 사육조건은 실시예 1과 같다. 본 실험은 ㈜메디톡스 동물실험윤리위원회의 검토 및 승인(A-2020-004) 후 수행되었다.After purchasing a 6-week-old female BALB/C mouse (Orient Bio), acclimatization and quarantine for 1 week, 7-week-old mice were used for the experiment. Feed, drinking water, and breeding conditions are the same as in Example 1. This experiment was conducted after review and approval (A-2020-004) by the Animal Experimental Ethics Committee of Medytox Co., Ltd.
4T1 마우스 유선 암종 세포주(ATCC® CRL-2539™)를 아메리칸 타입 컬쳐 컬렉션(American Type Culture Collection: ATCC)으로부터 수득하였다. 4T1 세포를 10% FBS(CAT No: 10082-147, Gibco) 및 1% 페니실린-스트렙토마이신(CAT No: 15140-122, Gibco) 함유 RPMI-1640 배지(CAT No: LM011-51, Welgene)에서 37℃, 5% CO2 조건으로 인큐베이터에서 배양하였다. 마취된 암컷 BALB/C 마우스 4번째 우측 유방 내 지방층에 주사기를 이용하여 5×105 세포를 0.05 ㎖ 이식하였다.The 4T1 mouse mammary carcinoma cell line (ATCC® CRL-2539™) was obtained from the American Type Culture Collection (ATCC). 4T1 cells were cultured in RPMI-1640 medium (CAT No: LM011-51, Welgene) containing 10% FBS (CAT No: 10082-147, Gibco) and 1% penicillin-streptomycin (CAT No: 15140-122, Gibco) 37 ℃, 5% CO 2 Conditions were cultured in an incubator. Using a syringe, 0.05 ml of 5×10 5 cells were transplanted into the fat layer in the fourth right breast of anesthetized female BALB/C mice.
비히클로서 보툴리눔 독소 조제에 사용된 멸균 생리식염수(Lot No: M8T7AF3, 대한약품공업)를 사용하였으며, 보툴리눔 독소로서 100 units 코어톡스 (Lot No: NSA19007A, 메디톡스)를 사용하였다. 표 3에 나타낸 바에 따라 각 시험물질을 조제하여 각 군에 투여하였다.As a vehicle, sterile physiological saline (Lot No: M8T7AF3, Daehan Pharmaceutical Co., Ltd.) used for botulinum toxin preparation was used, and 100 units Coretox (Lot No: NSA19007A, Medytox) was used as the botulinum toxin. As shown in Table 3, each test substance was prepared and administered to each group.
army 동물수number of animals 투여물질substance to be administered 용량Volume 투여경로route of administration 투여빈도Dosing frequency
1One 1010 비히클vehicle 1 ㎖/㎏1 ml/kg 종양 내within the tumor 1회(종양 이식 후 5일째)Once (5 days after tumor transplantation)
22 1010 보툴리눔 독소botulinum toxin 1.5 units/㎏1.5 units/kg 종양 내within the tumor 1회(종양 이식 후 5일째)Once (5 days after tumor transplantation)
33 1010 보툴리눔 독소 botulinum toxin 5 units/㎏5 units/kg 종양 내within the tumor 1회(종양 이식 후 5일째)Once (5 days after tumor transplantation)
44 1010 보툴리눔 독소 botulinum toxin 15 units/㎏15 units/kg 종양 내within the tumor 1회(종양 이식 후 5일째)Once (5 days after tumor transplantation)
55 1010 보툴리눔 독소 botulinum toxin 50 units/㎏50 units/kg 종양 내within the tumor 1회(종양 이식 후 5일째)Once (5 days after tumor transplantation)
마우스 유선 암종 이식 후 5일째 체중 및 종양 크기를 측정하여 평균 약 69 ㎣ 크기에서 마우스를 시험군에 따라 각 그룹 당 10마리씩 시험군간 차이가 없게 무작위 배정하였다. 종양 이식 후 5일째 체중을 바탕으로 비히클 및 보툴리눔 독소를 시험군 구성에 따라 종양 내로 투여하였다. 종양 이식 후 주 2회 종양크기를 측정하였다. 종양 크기는 실시예 1에 기재된 방법과 동일하게 측정하였다. 종양 이식 후 21일째, 종양의 크기가 1000 ㎣에 도달하기 전에 생존 개체의 혈액을 채취하였고 모든 동물을 안락사하였다. 혈액 내 엑소좀을 Exoquick exosome precipitation solution(Cat No: EXOQ20A-1, Lot No: 200107-001, System Biosciences)을 이용하여 분리하였고 ExoELISA-ULTRA Complete Kit(Cat No: EXELULTRA-CD63-1, Lot No: 200226-002, System Biosciences)을 이용해 측정하였다. 실시예 1과 동일하게 통계처리를 수행하였다.On the 5th day after transplantation of mouse mammary gland carcinoma, the body weight and tumor size were measured, and the mice were randomly assigned to each group at an average size of about 69 mm 3 without any difference between the test groups. On the 5th day after tumor transplantation, vehicle and botulinum toxin were administered intratumorally according to the composition of the test group based on body weight. Tumor size was measured twice a week after tumor transplantation. Tumor size was measured in the same manner as in Example 1. On day 21 after tumor transplantation, before the tumor size reached 1000 mm 3 , blood was collected from surviving individuals and all animals were euthanized. Exosomes in blood were isolated using Exoquick exosome precipitation solution (Cat No: EXOQ20A-1, Lot No: 200107-001, System Biosciences), and ExoELISA-ULTRA Complete Kit (Cat No: EXELULTRA-CD63-1, Lot No: 200226-002, System Biosciences). Statistical processing was performed in the same manner as in Example 1.
종양 성장 억제율 측정 결과를 표 4 및 도 4에 나타내었다.The measurement results of the tumor growth inhibition rate are shown in Table 4 and FIG. 4 .
army 투여물질substance to be administered 용량Volume 평균 종양 크기1)
(평균 ㎣±SEM)
Average tumor size 1)
(average ㎣±SEM)
TGI(%)1)TGI (%)1)
1One 비히클vehicle 1 ㎖/㎏1 ml/kg 989±82989±82 00
22 보툴리눔 독소botulinum toxin 1.5 units/㎏1.5 units/kg 916±59916±59 77
33 보툴리눔 독소 botulinum toxin 5 units/㎏5 units/kg 773±89773±89 2222
44 보툴리눔 독소 botulinum toxin 15 units/㎏15 units/kg 776±37* 776±37 * 2222
55 보툴리눔 독소 botulinum toxin 50 units/㎏50 units/kg 833±57833±57 1616
1) 종양 이식 후 21일째의 결과, * p<0.05, vs. 비히클. 양측 t-검정. 1) Results on day 21 after tumor transplantation, * p <0.05, vs. vehicle. Two -sided t -test.
표 4 및 도 4에 나타낸 바와 같이, 마우스 유선 종양 이식 후 21일째, 1.5 units/㎏ 보툴리눔 독소 단독 종양 내 투여에 의해 평균 916 ㎣의 종양 크기와 7%의 종양 성장 억제율을 보이며, 5 units/㎏ 투여군에서는 평균 773 ㎣ 및 22%, 15 units/㎏ 투여군에서는 평균 776 ㎣ 및 22%, 50 units/㎏ 투여군에서는 평균 833 ㎣ 및 16% 종양 성장 억제율을 보였다.As shown in Table 4 and FIG. 4, on the 21st day after mouse mammary tumor transplantation, 1.5 units/kg botulinum toxin alone intratumoral administration showed an average tumor size of 916 mm 3 and a tumor growth inhibition rate of 7%, and 5 units/kg In the administration group, an average of 773 mm 3 and 22%, in the 15 units/kg administration group, an average of 776 mm 3 and 22%, and in the 50 units/kg administration group, an average 833 mm 3 and 16% tumor growth inhibition rate were shown.
혈액 내 엑소좀의 수 측정 결과를 도 5에 나타내었다. 도 5에 나타낸 바와 같이, 15, 50 units/㎏ 보툴리눔 독소 투여군에서 비히클 군 대비 혈액 내 엑소좀의 수가 유의적으로 감소되었다.The results of measuring the number of exosomes in the blood are shown in FIG. 5 . As shown in FIG. 5 , the number of exosomes in the blood was significantly reduced in the 15 and 50 units/kg botulinum toxin administration group compared to the vehicle group.
실시예 3: 보툴리눔 독소의 종양 세포주 증식 억제 효과 시험Example 3: Botulinum toxin tumor cell line proliferation inhibitory effect test
B16-F10 마우스 악성 흑색종 세포주(KCLB No: 80008), A549 인간 폐암세포주(KCLB No: 10185), SNU324(KCLB No: 00324), Capan-2(KCLB No: 30080) 인간 췌장암 세포주를 한국세포주은행(KCLB: Korean Cell Line Bank)으로부터 수득하였다. B16-F10 세포를 10% FBS(CAT No: 10082-147, Gibco) 및 1% 페니실린-스트렙토마이신(CAT No: 15140-122, Gibco) 함유 DMEM 배지(CAT No: 11965-092, Gibco)에서 37 ℃, 5% CO2 조건으로 인큐베이터에서 배양하였다. A549 세포주는 10%, 1% 페니실린-스트렙토마이신 함유 RPMI1640(CAT No: LM011-51, Welgene Inc) 배지에서 SNU324 및 Capan-2 세포주는 10% FBS, 25mM HEPES(CAT No: 15630-080, Gibco) 함유 RPMI1640 배지에서 37 ℃, 5% CO2 조건으로 인큐베이터에서 배양하였다. 인간 종양 세포주를 사용하는 본 실험은 ㈜메디톡스 기관생명윤리위원회의 심의면제(IRB-2020-002) 후 수행되었다.B16-F10 mouse malignant melanoma cell lines (KCLB No: 80008), A549 human lung cancer cell line (KCLB No: 10185), SNU324 (KCLB No: 00324), and Capan-2 (KCLB No: 30080) human pancreatic cancer cell lines were obtained from the Korea Cell Line Bank. (KCLB: Korean Cell Line Bank) was obtained. B16-F10 cells were cultured in DMEM medium (CAT No: 11965-092, Gibco) containing 10% FBS (CAT No: 10082-147, Gibco) and 1% penicillin-streptomycin (CAT No: 15140-122, Gibco). ℃, 5% CO 2 Conditions were cultured in an incubator. A549 cell line 10%, 1% penicillin-streptomycin containing RPMI1640 (CAT No: LM011-51, Welgene Inc) medium SNU324 and Capan-2 cell line 10% FBS, 25 mM HEPES (CAT No: 15630-080, Gibco) It was cultured in an incubator at 37 °C, 5% CO 2 conditions in RPMI1640 containing medium. This experiment using a human tumor cell line was performed after being exempted from deliberation (IRB-2020-002) by the Institutional Bioethics Committee of Medytox Co., Ltd.
보툴리눔 독소로서 100 units 코어톡스(Lot No: NSA19007A 또는 NSA19004, 메디톡스)가 사용되었으며, 양성 대조물질로 독소루비신(LOT No: DROTK-GP, TCI)이 사용되었다.100 units Coretox (Lot No: NSA19007A or NSA19004, Medytox) was used as a botulinum toxin, and doxorubicin (LOT No: DROTK-GP, TCI) was used as a positive control material.
보툴리눔 독소의 종양 세포 증식에 대한 효과를 알아보고자 CCK8 proliferation assay(Cell Counting Kit-8, CK04, Dojindo)를 사용하였다. 구체적으로, 96-웰 플레이트(Cat No: 167008, Thermo Fisher)에 1 웰 당 5×103 cells/0.1 ㎖ 씩 세포를 분주하고 인큐베이터에서 24시간 배양하였다. 24시간 뒤 각 웰의 배지를 제거한 뒤 보툴리눔 독소를 0.016~250 U/㎖ 농도로 종양 세포주 배양에 사용되는 배지에 희석하여 처리하였고, 10 ㎍/㎖ 독소루비신을 처리하였다. 최초 처리 시점을 기준으로 6시간 뒤 각 웰의 배지를 제거한 뒤 동일한 시험물질을 각 웰에 재처리하였다. 최초 보툴리눔 독소 처리 시점을 기준으로 24시간 동안 인큐베이터에서 배양하였다. 총 24시간 배양한 뒤 각 웰에 CCK-8 시약을 10 ㎕씩 처리하고 3시간 동안 추가 배양한 뒤, SpectraMax i3(Model No: 10192-220, Molecular Devices)를 이용하여 450 ㎚에서 흡광도를 측정하여 증식율(proliferation rate)을 비교하였다. 증식율은 아래 식을 이용하여 계산하였다.To investigate the effect of botulinum toxin on tumor cell proliferation, CCK8 proliferation assay (Cell Counting Kit-8, CK04, Dojindo) was used. Specifically, cells were seeded at a rate of 5×10 3 cells/0.1 ml per well in a 96-well plate (Cat No: 167008, Thermo Fisher) and cultured in an incubator for 24 hours. After 24 hours, after removing the medium from each well, botulinum toxin was diluted in the medium used for tumor cell line culture at a concentration of 0.016-250 U/ml, and treated with 10 μg/ml doxorubicin. After removing the medium from each well 6 hours after the initial treatment time, the same test substance was re-treated into each well. It was cultured in an incubator for 24 hours from the time of initial botulinum toxin treatment. After incubation for a total of 24 hours, each well was treated with 10 μl of CCK-8 reagent, and after additional incubation for 3 hours, absorbance was measured at 450 nm using SpectraMax i3 (Model No: 10192-220, Molecular Devices). The proliferation rate was compared. The growth rate was calculated using the following equation.
증식율(%) =
Figure PCTKR2021009224-appb-I000001
(Ab: blank, Ac: negative control)
Growth rate (%) =
Figure PCTKR2021009224-appb-I000001
(Ab: blank, Ac: negative control)
각 농도 당 96-웰 플레이트의 4 웰씩 사용하여 이에 대한 평균값을 결과로 나타내었으며, 총 3개 플레이트에 대해 반복적으로 실험을 수행하였다. 통계분석은 실시예 1과 동일하게 수행하였다.For each concentration, 4 wells of a 96-well plate were used, and the average value thereof was shown as a result, and the experiment was repeated for a total of 3 plates. Statistical analysis was performed in the same manner as in Example 1.
실험 결과, 보툴리눔 독소에 의해 종양 세포의 성장이 유의적으로 저해되는 것으로 나타났다. 마우스 흑색종 세포주(B16-F10)에서는 0.4, 2, 50 U/㎖ 보툴리눔 독소 처리군에서 유의적으로 성장이 저해되었으며(도 6a), 인간 폐암 세포주(A549)에서는 2, 50, 250 U/㎖ 보툴리눔 독소 처리군에서(도 6b), 인간 췌장암(SNU324) 세포주에서는 0.08~50 U/㎖ 처리군에서(도 6c), 인간 췌장암(Capan-2) 세포주에서는 0.4~50 U/㎖ 보툴리눔 독소 처리군에서(도 6d) 유의적인 성장 저해 효과가 관찰되었다.As a result of the experiment, it was found that the growth of tumor cells was significantly inhibited by botulinum toxin. In the mouse melanoma cell line (B16-F10), growth was significantly inhibited in the 0.4, 2, and 50 U/ml botulinum toxin treatment group (FIG. 6a), and in the human lung cancer cell line (A549), 2, 50, and 250 U/ml In the botulinum toxin treatment group (Fig. 6b), in the human pancreatic cancer (SNU324) cell line, in the 0.08-50 U/ml treatment group (Fig. 6c), in the human pancreatic cancer (Capan-2) cell line, in the 0.4-50 U/ml botulinum toxin treatment group In (Fig. 6d) a significant growth inhibitory effect was observed.
실시예 4: 보툴리눔 독소의 마우스 흑색종 세포주에서 엑소좀 감소 효과 시험Example 4: Exosome reduction effect test of botulinum toxin in mouse melanoma cell line
B16-F10 마우스 악성 흑색종 세포주(KCLB No: 80008)를 한국세포주 은행(KCLB: Korean Cell Line Bank)으로부터 수득하였다. B16-F10 세포를 10% 엑소좀이 제거된 FBS(CAT No: A2720801, Gibco) 및 1% 페니실린-스트렙토마이신(CAT No: 15140-122, Gibco)을 함유하는 DMEM 배지(CAT No: 11965-092, Gibco)에서 37 ℃, 5% CO2 조건으로 인큐베이터에서 배양하였다.A B16-F10 mouse malignant melanoma cell line (KCLB No: 80008) was obtained from the Korean Cell Line Bank (KCLB). B16-F10 cells were cultured in DMEM medium (CAT No: 11965-092) containing 10% exosome-depleted FBS (CAT No: A2720801, Gibco) and 1% penicillin-streptomycin (CAT No: 15140-122, Gibco). , Gibco) in 37 ℃, 5% CO 2 Conditions were cultured in an incubator.
보툴리눔 독소로서 100 units 코어톡스(Lot No: NSA19004, 메디톡스)가 사용되었다.100 units Coretox (Lot No: NSA19004, Medytox) was used as a botulinum toxin.
6-웰 플레이트(Cat No: 140675, Thermo Fisher)에 1 웰 당 8×105 cells/2 ㎖ 씩 총 6개 웰에 세포를 분주하고 인큐베이터에서 24시간 배양하였다. 24시간 뒤 각 웰의 배지를 제거한 뒤 보툴리눔 독소를 10 U/㎖ 농도로 종양 세포주 배양에 사용되는 배지에 희석하여 처리하였고, 대조군의 경우 동량의 배지를 처리하였다. 최초 처리 시점을 기준으로 6시간 뒤 각 웰의 배지를 제거한 뒤 동일한 시험물질을 각 웰에 재처리하였다. 최초 보툴리눔 독소 처리 시점을 기준으로 24시간 동안 인큐베이터에서 배양하였다. 24시간 배양 후 각 웰 당 배지에 대해 엑소좀을 측정하였다. 배지 내 엑소좀은 ExoQuick-TC exosome precipitation solution(Cat No: EXOTC50A-1, Lot No: 191231-002, System Biosciences)을 이용해 분리하였고In a 6-well plate (Cat No: 140675, Thermo Fisher), 8×10 5 cells/2 ㎖ per well were seeded into 6 wells in total and cultured for 24 hours in an incubator. After 24 hours, the medium from each well was removed, and botulinum toxin was diluted in the medium used for tumor cell line culture at a concentration of 10 U/ml, and the same amount of medium was treated for the control group. After 6 hours from the initial treatment time, the medium from each well was removed, and the same test substance was re-treated into each well. It was cultured in an incubator for 24 hours from the time of initial botulinum toxin treatment. After culturing for 24 hours, exosomes were measured for each well of the medium. The exosomes in the medium were isolated using ExoQuick-TC exosome precipitation solution (Cat No: EXOTC50A-1, Lot No: 191231-002, System Biosciences).
ExoELISA-ULTRA Complete Kit(Cat No: EXEL-ULTRA-CD63-1, Lot No: 200226-002, System Biosciences)을 이용해 측정하였다.Measurements were made using the ExoELISA-ULTRA Complete Kit (Cat No: EXEL-ULTRA-CD63-1, Lot No: 200226-002, System Biosciences).
그 결과를 도 7에 나타내었다. 도 7에서와 같이, 10 U/㎖ 보툴리눔 독소에 의해 마우스 흑색종에서 엑소좀 감소 효과가 있는 것으로 확인되었다.The results are shown in FIG. 7 . As shown in FIG. 7 , it was confirmed that 10 U/ml botulinum toxin had an effect of reducing exosomes in mouse melanoma.
실시예 5: 보툴리눔 독소의 인간 췌장암 세포주에서 엑소좀 감소 효과 시험Example 5: Testing the effect of botulinum toxin on reducing exosomes in human pancreatic cancer cell lines
SNU324 인간 췌장암 세포주(KCLB No: 00324)를 한국세포주은행 (KCLB: Korean Cell Line Bank)으로부터 수득하였다. SNU324 세포를 10% 엑소좀이 제거된 FBS(CAT No: A2720801, Gibco) 및 10% FBS, 25mM HEPES(CAT No: 15630-080, Gibco)를 함유하는 RPMI-1640 배지(CAT No: LM011-51, Gibco)에서 37 ℃, 5% CO2 조건으로 인큐베이터에서 배양하였다. 인간 종양 세포주를 사용하는 본 실험은 ㈜메디톡스 기관생명윤리위원회의 심의면제 (IRB-2020-002) 후 수행되었다.The SNU324 human pancreatic cancer cell line (KCLB No: 00324) was obtained from the Korean Cell Line Bank (KCLB). SNU324 cells were cultured in RPMI-1640 medium (CAT No: LM011-51) containing 10% exosome-free FBS (CAT No: A2720801, Gibco) and 10% FBS, 25 mM HEPES (CAT No: 15630-080, Gibco). , Gibco) in 37 ℃, 5% CO 2 Conditions were cultured in an incubator. This experiment using a human tumor cell line was conducted after being exempted from deliberation by the Institutional Bioethics Committee of Medytox Co., Ltd. (IRB-2020-002).
보툴리눔 독소로서는 100 units 코어톡스(Lot No: NSA19004, 메디톡스)가 사용되었다.As the botulinum toxin, 100 units Coretox (Lot No: NSA19004, Medytox) was used.
6-웰 플레이트(Cat No: 140675, Thermo Fisher)에 1 웰 당 1.2×106 cells/2 ㎖ 씩 총 6개 웰에 세포를 분주하고 인큐베이터에서 24시간 배양하였다. 24시간 뒤 각 웰의 배지를 제거한 뒤 보툴리눔 독소를 10 U/㎖ 농도로 종양 세포주 배양에 사용되는 배지에 희석하여 처리하였고, 대조군의 경우 동량의 배지를 처리하였다. 최초 처리 시점을 기준으로 6시간 뒤 각 웰의 배지를 제거한 뒤 동일한 시험물질을 각 웰에 재처리하였다. 최초 보툴리눔 독소 처리 시점을 기준으로 24시간 동안 인큐베이터에서 배양하였다. 24시간 배양 후 각 웰 당 배지에 대해 엑소좀을 측정하였다. 배지 내 엑소좀은 ExoQuick-TC exosome precipitation solution(Cat No: EXOTC50A-1, Lot No: 191231-002, System Biosciences)을 이용해 분리하였고 ExoELISA-ULTRA Complete Kit(Cat No: EXEL-ULTRA-CD63-1, Lot No: 200226-002, System Biosciences)을 이용해 측정하였다.In a 6-well plate (Cat No: 140675, Thermo Fisher), 1.2×10 6 cells/2 ㎖ per well were seeded in a total of 6 wells and cultured for 24 hours in an incubator. After 24 hours, the medium from each well was removed, and botulinum toxin was diluted in the medium used for tumor cell line culture at a concentration of 10 U/ml, and the same amount of medium was treated for the control group. After 6 hours from the initial treatment time, the medium from each well was removed, and the same test substance was re-treated into each well. It was cultured in an incubator for 24 hours from the time of initial botulinum toxin treatment. After culturing for 24 hours, exosomes were measured for each well of the medium. The exosomes in the medium were isolated using ExoQuick-TC exosome precipitation solution (Cat No: EXOTC50A-1, Lot No: 191231-002, System Biosciences), and ExoELISA-ULTRA Complete Kit (Cat No: EXEL-ULTRA-CD63-1, Lot No: 200226-002, System Biosciences).
그 결과를 도 8에 나타내었다. 측정된 엑소좀 수에 대해 시험군과 대조군간 양측 t-검정을 사용하여 계산하였다(*p<0.05) 도 8에서와 같이, 10 U/㎖ 보툴리눔 독소에 의해 인간 췌장암 세포주에서 엑소좀 감소 효과가 있는 것으로 확인되었다.The results are shown in FIG. 8 . The measured number of exosomes was calculated using a two-sided t-test between the test group and the control group ( * p<0.05) As shown in FIG. 8, the effect of reducing exosomes in human pancreatic cancer cell lines by 10 U/ml botulinum toxin was it was confirmed that there is
실시예 6: 보툴리눔 독소의 종양 세포주 증식 억제 효과 시험Example 6: Test of botulinum toxin tumor cell line proliferation inhibitory effect
인간 뇌암 세포주 U87MG(KCLB No: 30014) 및 PC3(KCLB No: 21435) 인간 전립선암 세포주를 한국세포주은행(KCLB: Korean Cell Line Bank)으로부터 수득하였다. U87MG 세포주는 DMEM(CAT No: LM007-01, Welgene) 및 PC3 세포주는 RPMI-1640(CAT No: LM011-01, Welgene)에 10% FBS(CAT No: 10082-147, Gibco) 및 1% 페니실린-스트렙토마이신(CAT No: 15140-122, Gibco)을 함유하는 배지에서 37℃, 5% CO2 조건으로 인큐베이터에서 배양하였다. 인간 종양 세포주를 사용하는 본 실험은 ㈜메디톡스 기관생명윤리위원회의 심의면제(IRB-2020-015) 후 수행되었다.Human brain cancer cell lines U87MG (KCLB No: 30014) and PC3 (KCLB No: 21435) human prostate cancer cell lines were obtained from Korean Cell Line Bank (KCLB). U87MG cell line is DMEM (CAT No: LM007-01, Welgene) and PC3 cell line is RPMI-1640 (CAT No: LM011-01, Welgene) in 10% FBS (CAT No: 10082-147, Gibco) and 1% penicillin- Streptomycin (CAT No: 15140-122, Gibco) in a medium containing 37 ℃, 5% CO 2 Conditions were cultured in an incubator. This experiment using a human tumor cell line was performed after being exempted from deliberation by the Institutional Bioethics Committee (IRB-2020-015) of Medytox Co., Ltd.
보툴리눔 독소로서는 100 units 코어톡스(Lot No: NSA19007A 또는 NSA19004, 메디톡스)가 사용되었으며 양성 대조물질로 독소루비신(LOT No: DROTKGP, TCI)이 사용되었다.As the botulinum toxin, 100 units Coretox (Lot No: NSA19007A or NSA19004, Medytox) was used, and doxorubicin (LOT No: DROTKGP, TCI) was used as a positive control.
보툴리눔 독소의 종양 세포 증식에 대한 효과를 알아보고자 CCK8 proliferation assay(Cell Counting Kit-8, CK04, Dojindo)를 사용하였다. 96-웰 플레이트(Cat No: 167008, Thermo Fisher)에 1 웰 당 5×103 cells/0.1 ㎖씩 세포를 분주하고 인큐베이터에서 24시간 배양하였다. 24시간 뒤 각 웰의 배지를 제거한 뒤 보툴리눔 독소를 0.016 ~ 50 U/㎖ 농도로 종양 세포주 배양에 사용되는 배지에 희석하여 처리하거나, 10 ㎍/㎖ 독소루비신을 처리하였다. 최초 처리 시점을 기준으로 6시간 뒤 각 웰의 배지를 제거한 뒤 동일한 시험물질을 각 웰에 재 처리하였다. 최초 보툴리눔 독소 처리 시점을 기준으로 24시간 동안 인큐베이터에서 배양하였다.To investigate the effect of botulinum toxin on tumor cell proliferation, CCK8 proliferation assay (Cell Counting Kit-8, CK04, Dojindo) was used. Cells were seeded in a 96-well plate (Cat No: 167008, Thermo Fisher) at an amount of 5×10 3 cells/0.1 ml per well, and cultured in an incubator for 24 hours. After 24 hours, after removing the medium from each well, botulinum toxin was diluted in the medium used for tumor cell line culture at a concentration of 0.016-50 U/ml, or treated with 10 μg/ml doxorubicin. After 6 hours from the initial treatment time, the medium was removed from each well, and the same test substance was re-treated in each well. It was cultured in an incubator for 24 hours from the time of initial botulinum toxin treatment.
총 24시간 배양한 뒤 각 웰에 CCK-8 시약을 10 ㎕씩 처리하고 3시간 동안 추가 배양한 뒤, SpectraMax i3(Model No: 10192-220, Molecular Devices)를 이용하여 450 ㎚에서 흡광도를 측정하여 증식율(proliferation rate)을 비교하였다. 증식율은 아래 식을 이용하여 계산하였다.After incubation for a total of 24 hours, each well was treated with 10 μl of CCK-8 reagent, and after additional incubation for 3 hours, absorbance was measured at 450 nm using SpectraMax i3 (Model No: 10192-220, Molecular Devices). The proliferation rates were compared. The growth rate was calculated using the following equation.
증식율(%) =
Figure PCTKR2021009224-appb-I000002
(Ab: blank, Ac: negative control)
Growth rate (%) =
Figure PCTKR2021009224-appb-I000002
(Ab: blank, Ac: negative control)
각 농도 당 96-웰 플레이트의 4 웰씩 사용하여 이에 대한 평균값을 결과로 나타내었으며, 총 3개 플레이트에 대해 반복적으로 실험을 수행하였다. Graphpad Prism(version 7.05, GraphPad Software Inc, CA, USA)은 그래프 제시에 사용되었으며 SPSS software(version 25.0, SPSS Inc, IL, USA) 또는 Excel(2013, MS, USA)에 의해 통계 분석에 사용되었다. 모수 데이터는 양측(two-tailed) t-검정을 수행하였고 비모수 데이터는 Mann-Whitney 검정 통계분석을 유의 수준 p = 0.05에서 수행되었다.For each concentration, 4 wells of a 96-well plate were used, and the average value thereof was shown as a result, and the experiment was repeated for a total of 3 plates. Graphpad Prism (version 7.05, GraphPad Software Inc, CA, USA) was used for graph presentation and was used for statistical analysis by SPSS software (version 25.0, SPSS Inc, IL, USA) or Excel (2013, MS, USA). For parametric data, a two-tailed t -test was performed, and for nonparametric data, statistical analysis was performed using Mann-Whitney test at a significance level of p = 0.05.
측정된 종양 성장율에 대해 시험군과 대조군간 양측 t-검정을 사용하여 계산하였다(* p<0.05, ** p<0.01, *** p<0.001) 보툴리눔 독소에 의해 인간 뇌종양, 전립선암 세포의 성장이 유의적으로 저해되는 것으로 나타났다. 인간 뇌종양 세포주(U87MG)에서는 0.016 ~ 50 U/㎖ 처리군에서(도 9a), 인간 전립선암(PC3) 세포주에서는 0.4 ~ 50 U/㎖ 처리군에서(도 9b) 유의적인 성장 저해 효과가 관찰되었다.The measured tumor growth rate was calculated using a two-tailed t -test between the test group and the control group (* p <0.05, ** p <0.01, *** p <0.001) of human brain tumor and prostate cancer cells by botulinum toxin. It was found that growth was significantly inhibited. A significant growth inhibitory effect was observed in the human brain tumor cell line (U87MG) in the 0.016 ~ 50 U / ml treatment group (Fig. 9a), and in the human prostate cancer (PC3) cell line in the 0.4 ~ 50 U / ml treatment group (Fig. 9b). .

Claims (15)

  1. 유효성분으로 보툴리눔 독소(botulinum neurotoxin)를 포함하는, 엑소좀-매개된 질환을 치료하기 위한 조성물.A composition for treating exosome-mediated diseases, comprising botulinum toxin as an active ingredient.
  2. 제1항에 있어서, 엑소좀-매개된 질환이 암인 것인 조성물.The composition of claim 1 , wherein the exosome-mediated disease is cancer.
  3. 제2항에 있어서, 암이 전이성 암(metastatic cancer)인 것인 조성물.The composition of claim 2, wherein the cancer is metastatic cancer.
  4. 제2항 또는 제3항에 있어서, 암이 고형암(solid tumor)인 것인 조성물.The composition according to claim 2 or 3, wherein the cancer is a solid tumor.
  5. 제4항에 있어서, 고형암이 흑색종, 유방암, 폐암, 췌장암, 뇌종양 및 전립선암으로 이루어진 군으로부터 선택되는 하나 이상의 것인 조성물.The composition according to claim 4, wherein the solid cancer is at least one selected from the group consisting of melanoma, breast cancer, lung cancer, pancreatic cancer, brain tumor and prostate cancer.
  6. 제1항 내지 제5항 중 어느 한 항에 있어서, 엑소좀 분비가 증가된 대상에서 사용하기 위한 조성물.A composition according to any one of claims 1 to 5 for use in subjects with increased exosome secretion.
  7. 제1항 내지 제6항 중 어느 한 항에 있어서, 보툴리눔 독소가 혈청형 A, B, C, D, E, F, G, H, 또는 이들의 조합인 것인 조성물.7. The composition of any one of claims 1-6, wherein the botulinum toxin is serotype A, B, C, D, E, F, G, H, or a combination thereof.
  8. 제1항 내지 제7항 중 어느 한 항에 있어서, 약제학적으로 허용되는 부형제 또는 첨가제를 추가로 포함하는 조성물.8. The composition according to any one of claims 1 to 7, further comprising a pharmaceutically acceptable excipient or additive.
  9. 제8항에 있어서, 약제학적으로 허용되는 부형제 또는 첨가제는 안정화제, 이온 화합물, 계면활성제, 완충제, 동결건조 보호제, 또는 이들의 조합인 것인 조성물.The composition of claim 8, wherein the pharmaceutically acceptable excipient or additive is a stabilizer, an ionic compound, a surfactant, a buffer, a lyophilization protectant, or a combination thereof.
  10. 제9항에 있어서, 약제학적으로 허용되는 부형제 또는 첨가제는 아미노산, 염, 완충액, 비이온성 계면활성제, 당, 당 알코올, 또는 이들의 조합인 것인 조성물.10. The composition of claim 9, wherein the pharmaceutically acceptable excipient or additive is an amino acid, a salt, a buffer, a nonionic surfactant, a sugar, a sugar alcohol, or a combination thereof.
  11. 제8항 내지 제10항 중 어느 한 항에 있어서, 알부민 또는 동물 유래 성분이나 폴리사카라이드는 포함하지 않는 조성물.11. The composition according to any one of claims 8 to 10, which does not contain albumin or animal-derived components or polysaccharides.
  12. 제8항 내지 제11항 중 어느 한 항에 있어서, 동결건조 분말, 액상, 또는 프리필드 시린지 제제 형태의 조성물.12. The composition according to any one of claims 8 to 11, in the form of a lyophilized powder, liquid, or prefilled syringe formulation.
  13. 제8항 내지 제12항 중 어느 한 항에 있어서, 국소 투여하기 위한 조성물.13. A composition according to any one of claims 8 to 12 for topical administration.
  14. 제13항에 있어서, 종양 내 또는 종양 주변으로 투여하기 위한 조성물.14. The composition of claim 13 for intratumoral or peritumoral administration.
  15. 제8항 내지 제14항 중 어느 한 항에 있어서, 보툴리눔 독소를 50 내지 200 units으로 포함하는 조성물.15. The composition according to any one of claims 8 to 14, comprising 50 to 200 units of botulinum toxin.
PCT/KR2021/009224 2020-07-23 2021-07-19 Therapeutic agent for exosome-mediated disease WO2022019583A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20200091701 2020-07-23
KR10-2020-0091701 2020-07-23
KR10-2021-0009344 2021-01-22
KR1020210009344A KR20220012790A (en) 2020-07-23 2021-01-22 Agents for treating exosome-mediated disorders

Publications (1)

Publication Number Publication Date
WO2022019583A1 true WO2022019583A1 (en) 2022-01-27

Family

ID=79728873

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/009224 WO2022019583A1 (en) 2020-07-23 2021-07-19 Therapeutic agent for exosome-mediated disease

Country Status (1)

Country Link
WO (1) WO2022019583A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002074327A2 (en) * 2001-03-15 2002-09-26 Allergan, Inc. Compositions and methods for treating gonadotrophin related illnesses
KR20070045292A (en) * 2004-08-27 2007-05-02 알러간, 인코포레이티드 Methods for treating cancer
WO2010056337A2 (en) * 2008-11-12 2010-05-20 Caris Mpi, Inc. Methods and systems of using exosomes for determining phenotypes

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002074327A2 (en) * 2001-03-15 2002-09-26 Allergan, Inc. Compositions and methods for treating gonadotrophin related illnesses
KR20070045292A (en) * 2004-08-27 2007-05-02 알러간, 인코포레이티드 Methods for treating cancer
WO2010056337A2 (en) * 2008-11-12 2010-05-20 Caris Mpi, Inc. Methods and systems of using exosomes for determining phenotypes

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KATHLEEN M. MCANDREWS, RAGHU KALLURI: "Mechanisms associated with biogenesis of exosomes in cancer", MOLECULAR CANCER, vol. 18, no. 1, 1 December 2019 (2019-12-01), XP055732518, DOI: 10.1186/s12943-019-0963-9 *
MITTAL SHIVAM O., JABBARI BAHMAN: "Botulinum Neurotoxins and Cancer—A Review of the Literature", TOXINS, vol. 12, no. 32, pages 1 - 14, XP055888972, DOI: 10.3390/toxins12010032 *

Similar Documents

Publication Publication Date Title
JP7431409B2 (en) Use of bacteria, bacterial products, and other immunomodulatory entities in combination with anti-CTLA-4 antibodies and/or anti-PD-1 antibodies to treat solid tumor malignancies
CN108883187A (en) SMC conjoint therapy use for cancer treatment
US9474783B2 (en) HIP/PAP protein and derivatives thereof for use in treating cancer
WO2017095191A1 (en) Pharmaceutical composition comprising immunoglobulin fc-fused interleukin-7 fusion protein for preventing or treating human papillomavirus-caused diseases
EP3713585A1 (en) Msc-expressed immunomodulators in combination with car-t for cancer therapy
WO2018117345A1 (en) Selective induction of cancer apoptosis through combined inhibition of glutathione, thioredoxin, and nrf2 antioxidant
US11919854B2 (en) P2RX7 modulators in therapy
WO2022019583A1 (en) Therapeutic agent for exosome-mediated disease
WO2020116810A1 (en) Pharmaceutical composition, comprising inhibitory peptide against fas signaling, for prevention or treatment of obesity, fatty liver, or steatohepatitis
WO2022019584A1 (en) Cancer therapeutic agent
KR20220012790A (en) Agents for treating exosome-mediated disorders
US20220339233A1 (en) Compositions and methods for preventing recurrence of cancer
KR20220012809A (en) Agents for treating cancers
KR20220012791A (en) Agents for treating cancers
CA3199417A1 (en) Pharmaceutical composition for preventing or treating cancer comprising recombinant stabilized galectin 9 protein
KR20210127137A (en) Neuronal Oxide Synthase Inhibitors for Immunotherapy
WO2022092784A1 (en) Composition comprising cationic substance, and use for same
WO2021141465A1 (en) Composition for preventing or treating anticancer agent-induced allodynia and therapeutic method using same
AU2018308312B2 (en) Composition for preventing or treating cancer comprising a vascular disrupting agent and immune checkpoint inhibitor
CN115154611B (en) Use of immune checkpoint inhibitor in combination with anti-aging drug in preparation of tumor treatment product
WO2022139490A1 (en) Novel modified reovirus and use thereof
US20230022045A1 (en) Combination therapies for the treatment of cancer
WO2022114409A1 (en) Exosomes expressing rsvf and use thereof
Wang et al. Inflammation, Immunity, Fibrosis, and Infection: Distinct roles of intracellular heat shock protein 70 in maintaining gastrointestinal homeostasis
Chang Distinct roles of intracellular heat shock protein 70 (Hsp70) in maintaining gastrointestinal 1 homeostasis 2

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: 21846531

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: 21846531

Country of ref document: EP

Kind code of ref document: A1