WO2019112242A1 - Composition comprising sirna for prevention or treatment of neurofibrosarcoma - Google Patents

Composition comprising sirna for prevention or treatment of neurofibrosarcoma Download PDF

Info

Publication number
WO2019112242A1
WO2019112242A1 PCT/KR2018/014954 KR2018014954W WO2019112242A1 WO 2019112242 A1 WO2019112242 A1 WO 2019112242A1 KR 2018014954 W KR2018014954 W KR 2018014954W WO 2019112242 A1 WO2019112242 A1 WO 2019112242A1
Authority
WO
WIPO (PCT)
Prior art keywords
sirna
tns3
neurofibromatosis
tumor
expression
Prior art date
Application number
PCT/KR2018/014954
Other languages
French (fr)
Korean (ko)
Inventor
정선용
박은국
Original Assignee
아주대학교산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 아주대학교산학협력단 filed Critical 아주대학교산학협력단
Publication of WO2019112242A1 publication Critical patent/WO2019112242A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to an siRNA that inhibits the expression of the TNS3 gene and a composition for treating neurofibromatosis malignancies comprising the same as an effective ingredient.
  • Neurofibromatosis Type 1 was first described by Fredrich von Recklin-ghausen in 1882 and is an autosomal dominant genetic disorder that forms tumors along the peripheral nerves. It occurs in about 1 in 3,500 patients. The tumor is called neurofibromatosis because it can occur in any part of the nervous body, and the number is more than one. The main symptoms are neurofibroma and cafe-au-lait spots, Lisch nodules, optic pathway glioma, scoliosis, Tibial dysplasia, plexiform neurofibroma, pontine glioma, etc. (see Fig. 1 of the present invention).
  • the most common type 1 neurofibromatosis patient is a neurofibroma, a benign tumor that occurs in the skin.
  • About 30-40% of patients with type 1 neurofibromatosis have gunshot nerve fibromatosis, a benign tumor that occurs under the skin or in the deep part of the body.
  • gunshot neurofibromas show a massive lump (lump) shape that grows sideways and spreads sideways in various body parts (see FIG. 2 of the present invention), and a large number of multiple gunshot neurofibromas develop in the deep part 3 of the present invention).
  • plexiform neurofibroma is a benign tumor, it develops very seriously in patients, and in about 10% of patients with type 1 neurofibromatosis, neurofibromatosis is a malignant neurofibroma neurofibrosarcoma, malignant peripheral nerve sheath tumor (MPNST) (see FIG. 4 of the present invention).
  • MPNST peripheral nerve sheath tumor
  • PN neurofibromatosis
  • Type 1 neurofibromatosis is caused by a mutation in the NF1 gene, and about 50% of the patients are caused by naturally occurring mutations that are not hereditary.
  • the NF1 gene is located on chromosome 17q11.2, and consists of 350 kb genomic DNA and 57 exons.
  • the mRNA is about 11 to 13 kb.
  • the tumor suppressor gene NF1 encodes a neurofibromin protein in the cytoplasm of ⁇ 320 kDa that encodes a GAP-related protein that negatively regulates the Ras signal at amino acid 1125-1537 Domain (GAP-related domain).
  • GTPase-activating protein modulates the activity of Ras by stimulating intrinsic GTPase of Ras and converting it into an inactive form of Ras-GDP in the activated form, Ras-GTP.
  • Ras protein which is encoded by Ras, is expressed in both neuronal and endothelial cells, and the signal is transmitted downstream (downstream).
  • Increased GTP-Ras induces signal transduction through a representative downstream pathway, Raf kinase, and Raf induces signaling through kinase kinase including Erk1 and Erk2 isoforms of MEK kinase and mitogen-activated protein (MAP) kinase (See FIG. 6 of the present invention) by activating the cell.
  • MAP mitogen-activated protein
  • Ras genes There are three types of Ras genes: K-Ras, H-Ras, and N-Ras. Mutations in the Ras gene are found in about 50% of human cancers. Mutation of the Ras gene and excessive activity of the Ras protein are involved in the carcinogenesis of the cell, and thus research on the cancer treatment through suppression of the intracellular expression of the Ras protein has been actively carried out. It is known that all three Ras proteins, K-Ras, H-Ras and N-Ras, are overactivated by the mutation of the NF1 gene.
  • Another object of the present invention is to provide a pharmaceutical composition for preventing or treating nerve fibrosis, which is a malignant tumor of type 1 neurofibromatosis.
  • the present invention provides short interfering RNA (siRNA) comprising any one of the nucleotide sequences of SEQ ID NOS: 1 to 4.
  • the present invention provides a neurofibrosarcoma (malignant) tumor comprising short interfering RNA (siRNA) comprising any one of the nucleotide sequences of SEQ ID NOS: 1 to 4 as an active ingredient.
  • peripheral nerve sheath tumor, MPNST peripheral nerve sheath tumor
  • the present invention relates to a composition comprising an siRNA which effectively inhibits TNS3 gene expression as an active ingredient.
  • siRNA In the treatment of siRNA, the survival rate of cancer cells in malignant tumors is remarkably decreased, and isopamide, Etoposide ) And carboplatin (Carboplatin).
  • the siRNA-containing composition can effectively prevent, ameliorate, or treat malignant tumors of type 1 neurofibromatosis, A pharmaceutical composition for prevention or treatment of sarcoma, a health functional food composition for preventing or ameliorating nerve fibrosis, and the like.
  • Figure 1 shows representative symptoms of type 1 neurofibromatosis (A) milk-cough-au-lait spots, (B) neurofibroma, (C) Lisch nodules, (D) optic pathway glioma, (E) scoliosis, (F) tibial dysplasia, (G) plexiform neurofibroma, (H) glioma).
  • A milk-cough-au-lait spots
  • B neurofibroma
  • C Lisch nodules
  • D optic pathway glioma
  • E scoliosis
  • F tibial dysplasia
  • G plexiform neurofibroma
  • H glioma
  • FIG. 1 shows gunshot neurofibroma occurring in various body parts.
  • FIG. 3 shows an example of a gunshot neurofibroma occurring in the deep part.
  • Fig. 4 shows an example of nerve fibrosarcoma developed malignantly in benign bullet neurofibroma.
  • Figure 5 shows the process of tumor formation and malignancy of type 1 neurofibromatosis.
  • Fig. 6 schematically shows the role of neurofibromin and the mechanism of Ras activation.
  • FIG. 7 shows the results of comparing the amount of Tensin 3 protein and TNS3 mRNA expression in normal Schwann cells (HSC) and NF1 malignant tumor Schwann cell lines (FIG. 7a: increased expression of TNS3 gene in malignant tumor cells (S462) , Fig. 7b: expression of TNS3 gene is increased in malignant tumor (MPNST) sNF02.2, sNF96.2, S462 cell line).
  • HSC normal Schwann cells
  • Fig. 7b expression of TNS3 gene is increased in malignant tumor (MPNST) sNF02.2, sNF96.2, S462 cell line).
  • FIG. 8 shows ERK1 / 2 activation (phospho-ERK1 / 2) and Bcl-2 overexpression (FIG. 8A) upon inhibition of TNS3 expression using siRNA # 1 and # 2 in type 1 neurofibromatosis malignant tumor Schwann cell line (S462) RAS activation (Fig. 8B).
  • FIG. 10 shows the in vivo efficacy of TNS3 gene inhibition inhibition.
  • FIG. 10 shows the effect of SNS2, a type 1 neurofibromatous malignant tumor cell, on the size, volume, and volume of malignant tumor tissue of a xenograft mouse, (Observation of the anticancer effect of ICE and TNS3 gene siRNA (# 2) alone and in combination treatment).
  • Fig. 11 shows the result of comparing the size of the tumor and the size of the tumor in vivo by inhibition of TNS3 gene expression.
  • Neurofibromatosis Type 1 (NF1) was first described by Fredrich von Recklin-ghausen in 1882 and is an autosomal dominant genetic disorder that forms tumors along the peripheral nerves. It occurs in about 1 in 3,500 cases. Malignant tumors present in about 8 to 13% of patients are unpredictable, have severe clinical symptoms and high mortality rates, but are difficult to treat due to the inability to perform surgery depending on the location of the tumor. The efficacy of anticancer drugs is low and recurrence rates are high.
  • the inventors of the present invention have found that the suppression of the overexpression of the TNS3 gene (tensin 3 protein) in malignant tumors of patients with type 1 neurofibromatosis inhibits the proliferation and treatment of malignant tumors. Thereby completing the invention.
  • the present invention provides a short interfering RNA (siRNA) comprising any one of the nucleotide sequences of SEQ ID NOS: 1 to 4.
  • siRNA short interfering RNA
  • the siRNA can inhibit TNS3 gene expression and thus can be used to prevent, ameliorate, or treat neurofibromatosis by inhibiting TNS3.
  • the present invention provides neurofibrosarcoma (MPNST) comprising short interfering RNA (siRNA) consisting of any one of the nucleotide sequences of SEQ ID NOS: 1 to 4, ) ≪ / RTI >
  • miRNA short interfering RNA
  • the neurofilament sarcoma may be caused by malignant transformation of the bulbar neurofibroma. That is, the present invention can treat neurofibrosarcoma corresponding to a malignant tumor. It is known that the neurofilament sarcoma is caused by malignant development in about 8-13% of patients with gunshot neurofibroma.
  • the siRNA can prevent or treat nerve fibrosis by inhibiting the expression of the TNS3 gene.
  • the siRNA can prevent or treat nerve fibrosis by inhibiting the activation of ERK1 / 2 and RAS or the expression of Bcl-2.
  • the composition may be administered in combination with, but not limited to, any one or more anticancer agents selected from the group consisting of isopamide, carboplatin, and etoposide. That is, the composition may further comprise at least one anticancer agent selected from the group consisting of isoparamide, carboplatin and etoposide. In this case, the composition exhibits the best synergy and increases the killing effect of malignant tumor cells .
  • composition of the present invention when it is a pharmaceutical composition, for administration, it may contain a pharmaceutically acceptable carrier, excipient or diluent in addition to the above-mentioned effective ingredient.
  • a pharmaceutically acceptable carrier examples include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methylcellulose, microcrystalline cellulose, Polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.
  • the pharmaceutical composition of the present invention may be formulated in the form of powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols or the like, oral preparations, suppositories or sterilized injection solutions according to conventional methods .
  • it when formulating, it can be prepared using diluents or excipients such as fillers, weights, binders, humectants, disintegrants, surfactants and the like which are usually used.
  • Solid form preparations for oral administration include, but are not limited to, tablets, pills, powders, granules, capsules and the like.
  • Such a solid preparation may be prepared by mixing at least one excipient such as starch, calcium carbonate, sucrose, lactose, gelatin and the like in addition to the active ingredient.
  • excipients such as starch, calcium carbonate, sucrose, lactose, gelatin and the like in addition to the active ingredient.
  • lubricants such as magnesium stearate and talc may also be used.
  • Liquid preparations for oral administration, liquid paraffin, and various excipients such as wetting agents, sweeteners, fragrances, preservatives and the like.
  • Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized preparations and tasks.
  • Non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, injectable esters such as ethyl oleate, and the like.
  • a base for suppositories it is possible to use witepsol, macrosole, tween 61, cacao paper, laurin, glycerogelatin and the like.
  • the appropriate dose of the pharmaceutical composition of the present invention varies depending on the condition and the weight of the patient, the degree of disease, the type of drug, and the time, but can be appropriately selected by the person skilled in the art. 0.001 mg / kg to 50 mg / kg, and may be administered once to several times per day as needed.
  • HSC Human cells
  • sNF96.2, sNF02.2, S462 American Type Culture Collection, USA
  • MPNST neurofibrosarcoma
  • FBS fetal bovine serum
  • penicillin 100 U / ml
  • streptomycin 100 ⁇ g / ml
  • the cultured cells were lysed with 50 ⁇ g / ml phenylmethylsulfonyl fluoride (PMSF) and lysis buffer (150 mM NaCl, 1% Nonidet P-40, pH 7.4) supplemented with protease inhibitor cocktail , 0.5% sodium deoxycholate, and 0.1% SDS). Proteins were quantitated using the Lowry protein assay reagent kit (Bio-Rad Laboratories, USA).
  • PMSF phenylmethylsulfonyl fluoride
  • lysis buffer 150 mM NaCl, 1% Nonidet P-40, pH 7.4
  • protease inhibitor cocktail 0.5% sodium deoxycholate
  • SDS 0.1% SDS
  • the same amount of protein was taken for each condition, mixed with SDS-PAGE sample loading buffer (5X), heated at 100 ° C for 10 minutes, loaded and loaded on SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) Respectively.
  • SDS-PAGE sodium dodecyl sulfate-polyacrylamide gel electrophoresis
  • the proteins separated by electrophoresis were immobilized on an immobilon-P transfer membrane (0.45) at 350 mA for 120 minutes using a transfer buffer (20% methanol, 25 mM Tris-HCl, 192 mM glycine) (PVDF transfer membrane, Millipore Corporation, USA).
  • Protein-transferred membranes were blocked with 3% non-fat dry milk (skim milk solution), reacted with primary antibody for 24 h at 4 ° C, and incubated with TBST (tris-buffered saline and tween 20 ) ≪ / RTI > Thereafter, Western blot analysis was performed with each specific antibody as follows.
  • the primary antibodies used were TNS3 (Tensin 3) 1: 3000 (Sigma Aldrich, USA), ERK, phospo-ERK, cleaved caspase 3, Bcl- , Neurofibromin, ⁇ -actin 1: 3000 (Santa Cruz Biotechnology, INC, USA), the second antibody was peroxidase-conjugated chlorine
  • the cells were reacted with a goat anti-rabbit antibody, a goat anti-mouse antibody 1: 5000 (Santa Cruz Biotechnology, INC, USA), followed by WEST-ZOL Plus (iNtRON Biotechnology , KOREA) were processed and developed on the film.
  • Ras activation assay kit (Upstate Biotech. Lake Placid, NY).
  • the cultured S462 cells were dissolved in lysis buffer (25 mM HEPES pH 7.5, 150 mM NaCl, 1% Igepal CA-630, 10 mM MgCl 2 , 1 mM EDTA and 2% glycerol) ) was reacted with agarose beads binding to Raf-1 RBD fusion protein at 4 ° C for 1 hour.
  • lysis buffer 25 mM HEPES pH 7.5, 150 mM NaCl, 1% Igepal CA-630, 10 mM MgCl 2 , 1 mM EDTA and 2% glycerol
  • the agarose beads were washed three times with 1 ml of dissolution buffer, mixed with a loading buffer (5X), heated at 100 ° C for 10 minutes, and subjected to 12-15% SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis).
  • the agarose beads were collected by centrifugation and then washed with a dissolution buffer. Finally, a sample buffer was added and heated at 95 ° C for 5 minutes. Then, western blotting was performed using Ras specific antibody to analyze the amount of GTP-Ras.
  • siRNA short interfering RNA
  • siRNAs complementary to the TNS3 gene mRNA were designed and synthesized (Cosmogenetech, Korea).
  • a complementary two-stranded RNA-DNA hybrid oligomer (Sense and Antisense) was prepared by synthesizing each of the preceding 19 sequences with RNA and the last 2 TTs with DNA, (Double strand RNA) was used as the siRNA.
  • the nucleotide sequences of the sense and antisense siRNAs represented by SEQ ID NOS: 1 and 2 (# 1) and SEQ ID NOS: 3 and 4 (# 2) prepared at this time are shown in Table 1 below.
  • Trizol TM
  • DNase I Invitrogen Co., USA
  • STBR Green (TaKaRa, Shiga, Japan), which binds to double-stranded DNA and exhibits fluorescence, was added to cDNA (150 ng) and the fluorescence intensity was measured using an ABI Prism 7000 Sequence Detection System (Applied Biosystems; Foster City, CA, U, S , A).
  • the primer sequences used were as shown in Table 2 below.
  • Cell viability was analyzed using the D-Plus CCK cell viability assay kit (Donglin LS, Korea). Cells were seeded at a cell density of 7 x 10 < 3 > cells per well in 96 well plates and cultured for 24 hours after siRNA (see Table 1 above) and / or anticancer treatment. Subsequently, the culture medium was replaced with 100 ⁇ l of a culture medium supplemented with 10 ⁇ l of CCK reagent and further cultured for 2 hours. Cell viability was determined by measuring the absorbance at 450 nm using an ELISA plate reader (Bio-Rad Model 680).
  • Example 7 Formation of type 1 neurofibromatosis malignant xenograft mouse model and administration of short interfering RNA (siRNA)
  • Nerve fiber sarcoma S462 cells were centrifuged at 1,000 rpm for 5 minutes to remove the supernatant, followed by 1 ⁇ 10 7 cells / ml in phosphate-buffered saline (PBS), followed by subcutaneous injection of 0.1 ml Respectively. Tumor volume was measured and individuals reaching about 80 to 120 mm 3 were selected and grouped to equalize on the basis of tumor volume and body weight.
  • PBS phosphate-buffered saline
  • SiTNS3 (# 2) prepared in Example 4 was mixed with siTNS3 (10 ⁇ g) and in vivo-JETPEI (Polyplus, France) in a 5% glucose solution for 15 minutes and then local injection was performed on the tumor tissue .
  • siTNS3 was directly administered to the tumor, and siTNS3 10 ⁇ g / 100 ⁇ l was administered three times weekly (Monday, Wednesday, and Friday) for 3 weeks.
  • ifosfamide, carboplatin, and etoposide were prepared by intraperitoneal injection with physiological saline according to the concentration of the drug, kg) and etoposide (3.3 mg / kg) were administered intraperitoneally for 5 days (Mon. through Fri) for the first week, and intraperitoneal administration of carboplatin (13.3 mg / Respectively.
  • the maximum length (L) and the perpendicular width (W) of the tumor were measured using a caliper twice a week twice during the observation period, and the tumor volume (TV) Respectively.
  • mice were euthanized with CO 2 gas, and tumors were harvested and weighed.
  • Tumor growth inhibition rate was calculated by substituting the extracted tumor into the following equation (2).
  • T Mean tumor weight in experimental and control groups
  • TNS3 tensin-like SH2 domain containing 1, and Tensin 3
  • HSC normal schwann cells
  • S462 NF1 malignant tumor Schwann cell line
  • FIG. 7A expression of TNS3 gene was increased in malignant tumors Respectively.
  • Tensin 3 has been shown to be involved in Rho GTPase signaling and cell adhesion regulation, and is known to be phosphorylated (activated) by Src and is involved in malignant tumors.
  • TNS3 gene was also increased in other types of malignant tumors (MPNST) sNF02.2 and sNF96.2 cells. Therefore, it was hypothesized that inhibition of TNS3 (Tensin 3) expression would have a beneficial effect in the treatment of type 1 neurofibromatosis malignancy.
  • siTNS3 # 1 and # 2 prepared in Example 4 were cultured in the presence of type 1 neurofibromatous malignant tumor Schwann cell line (S462) using RNAi MAX (Invitrogen) ) To inhibit TNS3 expression.
  • Western blot analysis revealed the presence of ERK1 / 2 activation (phospho-ERK1 / 2, p-ERK1 / 2) and Bcl-2 expression level.
  • GTP-RAS degree of RAS activation
  • siTNS3 # 2 thus prepared was treated to inhibit the expression of TNS3, and then the change of NF1 malignant tumor Schwann cell line (S462) was observed.
  • siTNS3 # 2 was treated alone or in combination with ICE (Ifosfamide 1.8 mM, Carboplatin 90 ⁇ M, and Etoposide 30 ⁇ M), which are typical anticancer drugs used, by 10 pmole / ml, respectively, and cell viability was analyzed.
  • ICE Ifosfamide 1.8 mM, Carboplatin 90 ⁇ M, and Etoposide 30 ⁇ M
  • TNS3 gene siRNA (siTNS3 # 2) in S462 malignant cells, and the cell survival rate was significantly decreased in combination with ICO (Ifosfamide, Carboplatin, Etoposide) It is confirmed that there is synergy effect in city. In HSC cells, TNS3 gene siRNA (siTNS3 # 2) treatment did not show a synergistic effect with the combination of ICE (Ifosfamide, Carboplatin, Etoposide) and cell viability.
  • ICO Ifosfamide, Carboplatin, Etoposide
  • NF1 malignant tumor Schwannoma cell line (S462) was inoculated into nude mice to induce carcinogenesis of type 1 neurofibromatosis malignant xenograft Mouse, siTNS3 # 2 alone, chemotherapeutic agent ICE alone, siTNS3 # 2 and anti-cancer agent ICE (3 times a week, local injection, anticancer agent) were used as complementary binding to TNS3 gene as shown in Example 7-2
  • the first dose was administered to the abdomen for 21 days, and the size of the tumor was observed for 43 days.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a composition comprising TNS3 gene expression-suppressing short interfering RNA (siRNA) as an effective ingredient for preventing, alleviating, or treating malignant tumors of neurofibromatosis. According to the present invention, the siRNA was found to have good effects on the proliferation suppression and treatment of malignant tumors by suppressing the expression of TNS3 gene, which is overexpressed in tumors of neurofibromatosis type 1 patients, as the TNS3 gene expression-suppressing siRNA exhibited good effects in cell and animal tests. In addition, the TNS3 siRNA composition particularly has an excellent effect on the proliferation suppression and treatment of malignant tumors of neurofibromatosis type 1 when used in combination with the anticancer agents ICE (ifosfamide, carboplatin, etoposide).

Description

siRNA를 포함하는 신경섬유육종 예방 또는 치료용 조성물composition for preventing or treating nerve fibrosis including siRNA
본 발명은 TNS3 유전자의 발현을 억제하는 siRNA 및 이를 유효성분으로 포함하는 신경섬유종증 악성종양 치료용 조성물에 관한 것이다.The present invention relates to an siRNA that inhibits the expression of the TNS3 gene and a composition for treating neurofibromatosis malignancies comprising the same as an effective ingredient.
제1형 신경섬유종증(Neurofibromatosis Type 1, NF1)은 1882년 Fredrich von Recklin-ghausen이 처음 기술하였으며, 말초신경을 따라 종양을 형성하는 상염색체 우성유전 질환으로 약 3,500명 중 1명꼴로 발생한다. 종양은 신경이 있는 신체 어느 부위에나 발생할 수 있고, 그 수가 한 개 이상으로 생기는 다발성이기 때문에 '신경섬유종증'이라고 한다. 주된 증상은 신경이 있는 곳이면 어디서나 발생하는 신경섬유종(neurofibroma)과 밀크 커피색 반점(cafe-au-lait spots), 홍채결절(Lisch nodules), 시신경교종(optic pathway glioma), 척추측만(scoliosis), 경골형성이상(tibial dysplasia), 총상신경섬유종(plexiform neurofibroma), 교뇌신경교종(pontine glioma) 등이 있다(본원발명의 도 1 참조).Neurofibromatosis Type 1 (NF1) was first described by Fredrich von Recklin-ghausen in 1882 and is an autosomal dominant genetic disorder that forms tumors along the peripheral nerves. It occurs in about 1 in 3,500 patients. The tumor is called neurofibromatosis because it can occur in any part of the nervous body, and the number is more than one. The main symptoms are neurofibroma and cafe-au-lait spots, Lisch nodules, optic pathway glioma, scoliosis, Tibial dysplasia, plexiform neurofibroma, pontine glioma, etc. (see Fig. 1 of the present invention).
제1형 신경섬유종증 환자에서 가장 많이 발생하는 것은 피부에 발생하는 양성종양인 신경섬유종이다. 제1형 신경섬유종증 환자의 약 30-40%에서는 피부 아래 또는 몸의 심부에서 생기는 양성종양인 총상신경섬유종이 있다. 특히 총상신경섬유종의 경우는 다양한 신체부위에 옆으로 확산되어 자라서 자루처럼 늘어진 거대한 덩어리(혹) 모양을 나타내며(본원발명의 도 2 참조), 심부에 많은 수의 다발성 총상신경섬유종이 발생하기도 한다(본원발명의 도 3 참조). 이러한 총상신경섬유종(plexiform neurofibroma; PN)은 비록 양성 종양이지만 끊임없이 자라기 때문에 환자들에게 매우 심각한 증상이 발생하며, 제1형 신경섬유종증 환자의 약 10%에서 총상신경섬유종이 악성종양인 신경섬유육종(neurofibrosarcoma; malignant peripheral nerve sheath tumor, MPNST)으로 발전한다(본원발명의 도 4 참조). 이러한 제1형 신경섬유종증 환자에서 양성종양의 악성전개(malignant progression)는 급속도로 전개되기 때문에 매우 높은 치사율을 나타낸다.The most common type 1 neurofibromatosis patient is a neurofibroma, a benign tumor that occurs in the skin. About 30-40% of patients with type 1 neurofibromatosis have gunshot nerve fibromatosis, a benign tumor that occurs under the skin or in the deep part of the body. In particular, gunshot neurofibromas show a massive lump (lump) shape that grows sideways and spreads sideways in various body parts (see FIG. 2 of the present invention), and a large number of multiple gunshot neurofibromas develop in the deep part 3 of the present invention). Although plexiform neurofibroma (PN) is a benign tumor, it develops very seriously in patients, and in about 10% of patients with type 1 neurofibromatosis, neurofibromatosis is a malignant neurofibroma neurofibrosarcoma, malignant peripheral nerve sheath tumor (MPNST) (see FIG. 4 of the present invention). In patients with type I neurofibromatosis, the malignant progression of benign tumors develops rapidly, resulting in a very high mortality rate.
총상신경섬유종(PN)은 뼈와 주위 조직의 성장을 자극하여 거대한 크기로 자라나 추형의 외모를 나타내기도 하며, 이러한 환자들의 약 8~13%는 신경섬유육종(MPNSTs)으로 악성화 된다(본원발명의 도 5 참조). MPNST는 고도로 악성화된 전이암으로 높은 사망률과 화학요법과 방사능에 낮은 반응을 보이는 것으로 알려져 있다.(PN) stimulates the growth of bone and surrounding tissues and grows to a large size, but it may also appear to be a skewed appearance, and about 8 to 13% of such patients are malignant with neurofibromatosis (MPNSTs) 5). MPNST is a highly malignant metastatic cancer that is known to have a high mortality rate and low response to chemotherapy and radiation.
제1형 신경섬유종증은 NF1 유전자의 돌연변이에 의해 발병하며, 환자의 약 50%는 유전이 아닌 자연발생적인 돌연변이에 의해 발생한다. NF1 유전자는 염색체 17q11.2에 위치하며, 350 kb의 지노믹 DNA(genomic DNA), 57개의 엑손으로 구성되어 있고, mRNA는 11 내지 13 Kb정도이다. 종양억제유전자(tumor suppress gene) NF1은 ~320 kDa의 세포질에 존재하는 뉴로파이브로민(neurofibromin) 단백질을 암호화하고 있는데, 아미노산 1125-1537부위에 Ras 신호를 음성적으로(negatively) 조절하는 GAP-관련 도메인(GAP-related domain ; GRD) 영역을 가지고있다. GTPase-활성화 단백질(GTPase-activating protein ; GAP)은 Ras의 내재성(intrinsic) GTPase를 자극하여 활성화 형태인 Ras-GTP에서 Ras-GDP 불활성화 형태로 전환시킴으로써 Ras의 활성을 조절한다. 종양 유전자(oncogene) Ras에 의해 코딩되는 Ras 단백질은 신경능선세포와 내피세포 모두에서 발현되고 그 신호는 여러 가지 다운스트림 경로(downstream pathway)로 전달된다. 증가된 GTP-Ras는 대표적 다운스트림 경로인 Raf 키나아제를 통해 신호전달을 유발하고, Raf는 MEK 키나아제와 마이토젠-활성화 단백질(mitogen-activated protein ; MAP) 키나아제의 Erk1과 Erk2 아이소폼을 포함한 키나아제 캐스캐이드를 활성화시킴으로써 세포의 증식, 분화, 사멸 등에 관여한다(본원발명의 도 6 참조). Type 1 neurofibromatosis is caused by a mutation in the NF1 gene, and about 50% of the patients are caused by naturally occurring mutations that are not hereditary. The NF1 gene is located on chromosome 17q11.2, and consists of 350 kb genomic DNA and 57 exons. The mRNA is about 11 to 13 kb. The tumor suppressor gene NF1 encodes a neurofibromin protein in the cytoplasm of ~ 320 kDa that encodes a GAP-related protein that negatively regulates the Ras signal at amino acid 1125-1537 Domain (GAP-related domain). GTPase-activating protein (GAP) modulates the activity of Ras by stimulating intrinsic GTPase of Ras and converting it into an inactive form of Ras-GDP in the activated form, Ras-GTP. Oncogene The Ras protein, which is encoded by Ras, is expressed in both neuronal and endothelial cells, and the signal is transmitted downstream (downstream). Increased GTP-Ras induces signal transduction through a representative downstream pathway, Raf kinase, and Raf induces signaling through kinase kinase including Erk1 and Erk2 isoforms of MEK kinase and mitogen-activated protein (MAP) kinase (See FIG. 6 of the present invention) by activating the cell.
Ras 유전자는 크게 K-Ras, H-Ras, N-Ras의 3종류가 있으며 사람에게 발생하는 약 50% 암에서 Ras 유전자의 돌연변이가 발견된다. Ras 유전자의 돌연변이와 Ras 단백질의 과잉 활성은 세포의 암화에 관여하기 때문에 Ras 단백질의 세포내 발현억제를 통한 암 치료에 대한 연구가 활발히 진행되고 있다. NF1 유전자의 돌연변이에 의해 K-Ras, H-Ras, N-Ras의 3종류 Ras 단백질 모두가 과잉 활성화된다고 알려져 왔으나, 최근의 연구에서 제1형 신경섬유종증 환자의 별아교세포(astrocytes)의 경우 H-Ras와 N-Ras의 활성화는 보이지 않고 K-Ras만이 특이적으로 활성화되고, 조혈세포의 경우는 H-Ras만이 활성화된다는 사실이 보고되었다.There are three types of Ras genes: K-Ras, H-Ras, and N-Ras. Mutations in the Ras gene are found in about 50% of human cancers. Mutation of the Ras gene and excessive activity of the Ras protein are involved in the carcinogenesis of the cell, and thus research on the cancer treatment through suppression of the intracellular expression of the Ras protein has been actively carried out. It is known that all three Ras proteins, K-Ras, H-Ras and N-Ras, are overactivated by the mutation of the NF1 gene. However, recent studies have shown that astrocytes in patients with type I neurofibromatosis have H- Ras and N-Ras are not activated, only K-Ras is specifically activated, and hematopoietic cells are activated only by H-Ras.
뉴로파이브로민(neurofibromin)의 손실(loss) 혹은 기능 장애로 인해 과잉 활성 및 축적된 RAS에 의해 발병하는 NF1 환자의 20%에서는 생명을 위협하는 심한 합병증이 야기되고, 환자의 약 8~13%에서 나타나는 악성화는 예측이 불가능하며 임상적 증상이 심각하지만 유전성이기 때문에 근본 치료는 불가능하다. 신경섬유육종으로 악성화가 된 경우, 종양의 절제 등의 대중요법만이 유일한 치료법이나, 종양의 위치에 따라 수술이 불가능한 경우가 많아 암의 진행시기에 관계없이 환자의 생명과 직결된다. 제1형 신경섬유종증의 임상적 표현형의 다양성과 악성화 과정에 대한 정확한 기전은 여전히 밝혀지지 않았다. 전통적인 치료방법으로 방사선 요법, 외과적 수술 절제, 세포 독성약이 있지만 장기간의 효과는 크게 없는 것으로 밝혀졌다.20% of NF1 patients with overactive and accumulating RAS due to neurofibromin loss or dysfunction cause severe life-threatening complications and about 8 to 13% And the clinical symptoms are severe, but inherited, so it is impossible to treat it. In the case of malignant neurofibrosarcoma, only mass therapy such as resection of the tumor is the only treatment, or surgery is impossible according to the location of the tumor, and it is directly related to the life of the patient regardless of the timing of the cancer. The exact mechanism of the variability and malignancy of the clinical phenotype of type 1 neurofibromatosis remains unclear. Traditional therapies include radiotherapy, surgical resection, and cytotoxic drugs, but no long-term effects were found.
본 발명의 목적은 TNS3 유전자 발현을 억제할 수 있는 짧은 간섭 RNA를 제공하는 데에 있다.It is an object of the present invention to provide short interfering RNA capable of inhibiting TNS3 gene expression.
본 발명의 다른 목적은 제1형 신경섬유종증의 악성종양인 신경섬유육종의 예방 또는 치료용 약학 조성물을 제공하는 데에 있다.Another object of the present invention is to provide a pharmaceutical composition for preventing or treating nerve fibrosis, which is a malignant tumor of type 1 neurofibromatosis.
상기 목적을 달성하기 위하여, 본 발명은 서열번호 1 내지 서열번호 4로 표시되는 염기 서열 중 어느 하나로 이루어지는 짧은 간섭 RNA(short interfering RNA; siRNA)를 제공한다.In order to achieve the above object, the present invention provides short interfering RNA (siRNA) comprising any one of the nucleotide sequences of SEQ ID NOS: 1 to 4.
상기 다른 목적을 달성하기 위하여, 본 발명은 서열번호 1 내지 서열번호 4로 표시되는 염기 서열 중 어느 하나로 이루어지는 짧은 간섭 RNA(short interfering RNA; siRNA)를 유효성분으로 함유하는 신경섬유육종(neurofibrosarcoma; malignant peripheral nerve sheath tumor, MPNST) 예방 또는 치료용 약학 조성물을 제공한다.In order to accomplish the above object, the present invention provides a neurofibrosarcoma (malignant) tumor comprising short interfering RNA (siRNA) comprising any one of the nucleotide sequences of SEQ ID NOS: 1 to 4 as an active ingredient. peripheral nerve sheath tumor, MPNST).
본 발명은 TNS3 유전자 발현을 효과적으로 억제하는 siRNA를 유효성분으로 포함하는 조성물에 관한 것으로, 상기 siRNA 처리 시에 악성종양의 암세포의 생존율이 현저히 감소하였으며, 이소파마이드(Isofamide), 에토포시드(Etoposide) 및 카보플라틴(Carboplatin) 등의 항암제와의 병용 처리 시에도 상승효과를 나타낸 바, 상기 siRNA를 포함하는 조성물은 제1형 신경섬유종증의 악성종양을 효과적으로 예방, 개선 내지 치료할 수 있으므로, 신경섬유육종 예방 또는 치료용 약학 조성물, 신경섬유육종 예방 또는 개선용 건강기능식품 조성물 등으로 유용하게 활용될 수 있다.The present invention relates to a composition comprising an siRNA which effectively inhibits TNS3 gene expression as an active ingredient. In the treatment of siRNA, the survival rate of cancer cells in malignant tumors is remarkably decreased, and isopamide, Etoposide ) And carboplatin (Carboplatin). The siRNA-containing composition can effectively prevent, ameliorate, or treat malignant tumors of type 1 neurofibromatosis, A pharmaceutical composition for prevention or treatment of sarcoma, a health functional food composition for preventing or ameliorating nerve fibrosis, and the like.
도 1은 제1형 신경섬유종증의 대표적인 증상을 나타낸 것이다((A) 밀크 커피색 반점(cafe-au-lait spots), (B) 피부신경섬유종(neurofibroma), (C) 홍채결절(Lisch nodules), (D) 시신경교종(optic pathway glioma), (E) 척추측만(scoliosis), (F) 경골형성이상(tibial dysplasia), (G) 총상신경섬유종(plexiform neurofibroma), (H) 교뇌신경교종(pontine glioma)).Figure 1 shows representative symptoms of type 1 neurofibromatosis (A) milk-cough-au-lait spots, (B) neurofibroma, (C) Lisch nodules, (D) optic pathway glioma, (E) scoliosis, (F) tibial dysplasia, (G) plexiform neurofibroma, (H) glioma).
도 2는 다양한 신체부위에 발생하는 총상신경섬유종을 나타낸 것이다.Figure 2 shows gunshot neurofibroma occurring in various body parts.
도 3은 심부에 발생한 총상신경섬유종의 예를 나타낸 것이다.FIG. 3 shows an example of a gunshot neurofibroma occurring in the deep part.
도 4는 양성 총상신경섬유종에서 악성으로 전개된 신경섬유육종의 예를 나타낸 것이다.Fig. 4 shows an example of nerve fibrosarcoma developed malignantly in benign bullet neurofibroma.
도 5는 제1형 신경섬유종증의 종양형성 및 악성화의 과정을 나타낸 것이다.Figure 5 shows the process of tumor formation and malignancy of type 1 neurofibromatosis.
도 6은 뉴로파이브로민(neurofibromin)의 역할과 Ras 활성화 기전을 모식적으로 나타낸 것이다.Fig. 6 schematically shows the role of neurofibromin and the mechanism of Ras activation.
도 7은 정상 슈반 세포(HSC)와 NF1 악성종양 슈반 세포주에서의 Tensin 3 단백질과 TNS3 mRNA 발현량을 비교한 결과를 나타낸 것이다(도 7a: 악성 종양 세포(S462)에서 TNS3 유전자의 발현이 증가되어 있음, 도 7b: 악성 종양(MPNST) sNF02.2, sNF96.2, S462 세포주에서 TNS3 유전자의 발현이 증가 되어 있음).FIG. 7 shows the results of comparing the amount of Tensin 3 protein and TNS3 mRNA expression in normal Schwann cells (HSC) and NF1 malignant tumor Schwann cell lines (FIG. 7a: increased expression of TNS3 gene in malignant tumor cells (S462) , Fig. 7b: expression of TNS3 gene is increased in malignant tumor (MPNST) sNF02.2, sNF96.2, S462 cell line).
도 8은 제1형 신경섬유종증 악성종양 슈반 세포주(S462)에 siRNA #1, #2를 이용한 TNS3 발현 저해 시의 ERK1/2 활성화(phospho-ERK1/2)와 Bcl-2 과발현(도 8A) 및 RAS 활성화(도 8B)에 미치는 영향을 확인한 결과이다.FIG. 8 shows ERK1 / 2 activation (phospho-ERK1 / 2) and Bcl-2 overexpression (FIG. 8A) upon inhibition of TNS3 expression using siRNA # 1 and # 2 in type 1 neurofibromatosis malignant tumor Schwann cell line (S462) RAS activation (Fig. 8B).
도 9는 TNS3 유전자에 대한 siRNA(#2) 단독 처리 또는 항암제와의 병용 처리 시의 세포 생존율을 확인한 결과이다.9 shows the results of confirming the cell survival rate of siRNA (# 2) alone or in combination with an anticancer drug against the TNS3 gene.
도 10은 TNS3 유전자 발현억제에 의한 in vivo 효능을 확인한 것으로, 제1형 신경섬유종증 악성종양 세포인 S462를 마우스에 이식하여 제작한 악성종양 모델 동물인 이종이식 마우스의 악성종양 조직의 크기, 부피, 무게의 변화를 관찰한 결과이다(항암제인 ICE와 TNS3 유전자 siRNA(#2) 단독 및 병용 처리 시의 현저한 항암 효과 확인).FIG. 10 shows the in vivo efficacy of TNS3 gene inhibition inhibition. FIG. 10 shows the effect of SNS2, a type 1 neurofibromatous malignant tumor cell, on the size, volume, and volume of malignant tumor tissue of a xenograft mouse, (Observation of the anticancer effect of ICE and TNS3 gene siRNA (# 2) alone and in combination treatment).
도 11은 TNS3 유전자 발현억제에 의한 in vivo 실험에서 종양의 외형과 적출한 종양의 크기를 비교한 결과를 나타낸 것이다.Fig. 11 shows the result of comparing the size of the tumor and the size of the tumor in vivo by inhibition of TNS3 gene expression.
제1형 신경섬유종증(Neurofibromatosis Type1, NF1)은 1882년 Fredrich von Recklin-ghausen이 처음 기술하였으며, 말초신경을 따라 종양을 형성하는 상염색체 우성유전 질환으로 약 3,500명 중 1명꼴로 발생한다. 환자의 약 8 내지 13%에서 나타나는 악성종양은 예측이 불가능하고 임상적 증상이 심각하며 치사율이 높지만, 종양의 위치에 따라 수술이 불가능한 경우가 많아 치료가 매우 힘든 질환이다. 항암제 효과가 낮고 재발율이 높아 아직까지 적절한 약물치료제가 없는 실정이다. Neurofibromatosis Type 1 (NF1) was first described by Fredrich von Recklin-ghausen in 1882 and is an autosomal dominant genetic disorder that forms tumors along the peripheral nerves. It occurs in about 1 in 3,500 cases. Malignant tumors present in about 8 to 13% of patients are unpredictable, have severe clinical symptoms and high mortality rates, but are difficult to treat due to the inability to perform surgery depending on the location of the tumor. The efficacy of anticancer drugs is low and recurrence rates are high.
이에, 본 발명의 발명자들은 제1형 신경섬유종증 환자의 악성종양에서 과발현(overexpression)되어 있는 TNS3 유전자(tensin 3 단백질)의 발현을 억제함으로써 악성종양의 증식 억제와 치료에 좋은 효과가 있음을 밝히고 본 발명을 완성하였다.Accordingly, the inventors of the present invention have found that the suppression of the overexpression of the TNS3 gene (tensin 3 protein) in malignant tumors of patients with type 1 neurofibromatosis inhibits the proliferation and treatment of malignant tumors. Thereby completing the invention.
따라서, 본 발명은 서열번호 1 내지 서열번호 4로 표시되는 염기 서열 중 어느 하나로 이루어지는 짧은 간섭 RNA(short interfering RNA; siRNA)를 제공한다.Accordingly, the present invention provides a short interfering RNA (siRNA) comprising any one of the nucleotide sequences of SEQ ID NOS: 1 to 4.
본 발명의 일 실시예에서, 상기 siRNA는 TNS3 유전자 발현을 억제할 수 있는 바, 따라서 TNS3을 억제하여 신경섬유종증을 예방, 개선 내지 치료하는 데에 사용될 수 있다.In one embodiment of the present invention, the siRNA can inhibit TNS3 gene expression and thus can be used to prevent, ameliorate, or treat neurofibromatosis by inhibiting TNS3.
이에, 본 발명은 서열번호 1 내지 서열번호 4로 표시되는 염기 서열 중 어느 하나로 이루어지는 짧은 간섭 RNA(short interfering RNA; siRNA)를 유효성분으로 함유하는 신경섬유육종(neurofibrosarcoma; malignant peripheral nerve sheath tumor, MPNST) 예방 또는 치료용 약학 조성물을 제공한다.Accordingly, the present invention provides neurofibrosarcoma (MPNST) comprising short interfering RNA (siRNA) consisting of any one of the nucleotide sequences of SEQ ID NOS: 1 to 4, ) ≪ / RTI >
상기 신경섬유육종은 총상신경섬유종의 악성화로 유발된 것일 수 있는 바, 즉, 본 발명은 악성 종양에 해당하는 신경섬유육종을 치료할 수 있다. 상기 신경섬유육종은 총상신경섬유종 환자의 약 8-13%에서 악성화가 전개되어 유발되는 것으로 알려져 있다.The neurofilament sarcoma may be caused by malignant transformation of the bulbar neurofibroma. That is, the present invention can treat neurofibrosarcoma corresponding to a malignant tumor. It is known that the neurofilament sarcoma is caused by malignant development in about 8-13% of patients with gunshot neurofibroma.
이때, 상기 siRNA는 TNS3 유전자의 발현을 억제함으로써 신경섬유육종을 예방 또는 치료할 수 있다.At this time, the siRNA can prevent or treat nerve fibrosis by inhibiting the expression of the TNS3 gene.
또한, 상기 siRNA는 ERK1/2 및 RAS의 활성화 또는 Bcl-2의 발현을 억제함으로써 신경섬유육종을 예방 또는 치료할 수 있다.In addition, the siRNA can prevent or treat nerve fibrosis by inhibiting the activation of ERK1 / 2 and RAS or the expression of Bcl-2.
상기 조성물은 이소파마이드(Isofamide), 카보플라틴(Carboplatin) 및 에토포시드(Etoposide)로 이루어진 군에서 선택된 어느 하나 이상의 항암제와 병용 투여할 수 있으나, 이에 제한되는 것은 아님을 명시한다. 즉, 상기 조성물은 이소파마이드, 카보플라틴 및 에토포시드로 이루어진 군에서 선택된 어느 하나 이상의 항암제를 더 포함할 수 있는 바, 이러한 경우, 가장 우수한 시너지 효과를 나타내어 악성종양 세포의 사멸 효과를 증가시킬 수 있다.The composition may be administered in combination with, but not limited to, any one or more anticancer agents selected from the group consisting of isopamide, carboplatin, and etoposide. That is, the composition may further comprise at least one anticancer agent selected from the group consisting of isoparamide, carboplatin and etoposide. In this case, the composition exhibits the best synergy and increases the killing effect of malignant tumor cells .
본 발명의 조성물이 약학 조성물인 경우, 투여를 위하여, 상기 기재한 유효성분 이외에 약학적으로 허용 가능한 담체, 부형제 또는 희석제를 포함할 수 있다. 상기 담체, 부형제 및 희석제로는 락토오스, 덱스트로오스, 수크로오스, 소르비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로오스, 메틸 셀룰로오스, 미정질 셀룰로오스, 폴리비닐피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유를 들 수 있다.When the composition of the present invention is a pharmaceutical composition, for administration, it may contain a pharmaceutically acceptable carrier, excipient or diluent in addition to the above-mentioned effective ingredient. Examples of the carrier, excipient and diluent include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia rubber, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methylcellulose, microcrystalline cellulose, Polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.
본 발명의 약학 조성물은 각각 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 현탁액, 에멀젼, 시럽, 에어로졸 등의 경구형 제형, 외용제, 좌제 또는 멸균 주사용액의 형태로 제형화하여 사용할 수 있다. 상세하게는 제형화할 경우 통상 사용하는 충진제, 중량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제될 수 있다. 경구투여를 위한 고형 제제로는 정제, 환제, 산제, 과립제, 캡슐제 등을 포함하나, 이에 한정되는 것은 아니다. 이러한 고형 제제는 상기 유효성분 외에 적어도 하나 이상의 부형제, 예를 들면, 전분, 칼슘 카보네이트, 수크로오스, 락토오스, 젤라틴 등을 섞어 조제될 수 있다. 또한, 단순한 부형제 이외에 마그네슘 스테아레이트, 탈크 같은 윤활제들도 사용될 수 있다. 경구를 위한 액상물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등을 첨가하여 조제될 수 있다. 비경구 투여를 위한 제제는 멸균된 수용액, 비수성 용제, 현탁제, 유제, 동결건조 제제 및 과제를 포함한다. 비수성 용제 및 현탁제로는 프로필렌글리콜, 폴리에틸렌글리콜, 올리브 오일과 같은 식물성 오일, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔, 마크로솔, 트윈 61, 카카오지, 라우린지, 글리세로젤라틴 등이 사용될 수 있다.The pharmaceutical composition of the present invention may be formulated in the form of powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols or the like, oral preparations, suppositories or sterilized injection solutions according to conventional methods . In detail, when formulating, it can be prepared using diluents or excipients such as fillers, weights, binders, humectants, disintegrants, surfactants and the like which are usually used. Solid form preparations for oral administration include, but are not limited to, tablets, pills, powders, granules, capsules and the like. Such a solid preparation may be prepared by mixing at least one excipient such as starch, calcium carbonate, sucrose, lactose, gelatin and the like in addition to the active ingredient. In addition to simple excipients, lubricants such as magnesium stearate and talc may also be used. Liquid preparations for oral administration, liquid paraffin, and various excipients such as wetting agents, sweeteners, fragrances, preservatives and the like. Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized preparations and tasks. Non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, injectable esters such as ethyl oleate, and the like. As a base for suppositories, it is possible to use witepsol, macrosole, tween 61, cacao paper, laurin, glycerogelatin and the like.
본 발명의 약학 조성물의 적합한 투여량은 환자의 상태 및 체중, 질병의 정도, 약물형태, 시간에 따라 다르지만, 당 업자에 의해 적절하게 선택될 수 있는 바, 상기 조성물의 일일 투여량은 바람직하게는 0.001 mg/kg 내지 50 mg/kg이며, 필요에 따라 일일 1회 내지 수회로 나누어 투여할 수 있다.The appropriate dose of the pharmaceutical composition of the present invention varies depending on the condition and the weight of the patient, the degree of disease, the type of drug, and the time, but can be appropriately selected by the person skilled in the art. 0.001 mg / kg to 50 mg / kg, and may be administered once to several times per day as needed.
이하, 본 발명의 이해를 돕기 위하여 실시예를 들어 상세하게 설명하기로 한다. 다만 하기의 실시예는 당업계에서 평균적인 지식을 가진 자에게 본 발명을 보다 완전하게 설명하기 위해 제공되는 것이며 본 발명의 내용을 예시하는 것일 뿐이므로 본 발명의 범위가 하기 실시예에 한정되는 것은 아니다.BEST MODE FOR CARRYING OUT THE INVENTION Hereinafter, the present invention will be described in detail with reference to the following examples. It is to be understood that both the foregoing description and the following detailed description are exemplary and explanatory and are intended to provide further explanation of the invention as claimed. no.
실시예 1: 세포 배양Example 1: Cell culture
정상 세포인 인간 세포(HSC)(ScienCell Research Laboratories)와 신경섬유육종(neurofibrosarcoma; malignant peripheral nerve sheath tumor, MPNST) 세포인 sNF96.2, sNF02.2, S462(American Type culture Collection, USA)는 10% 우태아혈청(fetal bovine serum; FBS, Hyclone Laboratories, Logan, USA), 페니실린(100 U/ml)과 스트렙토마이신(100 μg/ml)을 첨가한 둘베코의 수정 이글배지(Dulbecco's modified Eagle's medium ; Hyclone Laboratories, Logan, USA)를 이용하여 37℃, CO2 배양기에서 배양하였다. Human cells (HSC) (ScienCell Research Laboratories) and sNF96.2, sNF02.2, S462 (American Type Culture Collection, USA), which are neurofibrosarcoma (MPNST) Dulbecco's modified Eagle's medium supplemented with fetal bovine serum (FBS, Hyclone Laboratories, Logan, USA), penicillin (100 U / ml) and streptomycin (100 μg / ml) The cells were cultured in a CO 2 incubator at 37 ° C using a Bio-Rad Laboratories, Logan, USA.
실시예 2: 웨스턴 블롯 분석Example 2: Western blot analysis
배양한 세포를 50 μg/ml 페닐메탄설포닐 플루오라이드(phenylmethylsulfonyl fluoride, PMSF), 단백분해효소 억제제 칵테일(Protease inhibitor cocktail)이 첨가된 용해 버퍼(lysis buffer; 150 mM NaCl, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS)를 첨가하여 단백질을 추출한 후, 여기서 얻어진 시료를 Lowry protein assay reagent kit(Bio-rad laboratories, U.S.A.)를 이용하여 단백질을 정량하였다.The cultured cells were lysed with 50 μg / ml phenylmethylsulfonyl fluoride (PMSF) and lysis buffer (150 mM NaCl, 1% Nonidet P-40, pH 7.4) supplemented with protease inhibitor cocktail , 0.5% sodium deoxycholate, and 0.1% SDS). Proteins were quantitated using the Lowry protein assay reagent kit (Bio-Rad Laboratories, USA).
각 조건마다 동일한 양의 단백질을 취하여 SDS-PAGE 샘플 로딩 버퍼(5X)와 섞은 후, 100℃에서 10분간 가열하고, 이를 로딩하여 8-15% SDS-PAGE(sodium dodecyl sulfate-polyacrylamide gel electrophoresis)로 분리하였다. 전기영동으로 분리한 단백질은 트랜스퍼 버퍼(transfer buffer; 20% 메탄올, 25 mM Tris-HCl, 192 mM 글라이신)를 이용하여 350 mA에서 120분간 이모빌론-피-트랜스퍼 멤브레인(immobilon-P transfer membrane(0.45 ㎛); PVDF transfer membrane, Millipore Corporation, USA)으로 흡착 이동시켰다. 단백질이 이동된 멤브레인은 3% 탈지유(non-fat dry milk; skim milk solution)로 블로킹(blocking)시킨 후, 4℃에서 1차 항체로 24시간 동안 반응시키고, TBST(tris-buffered saline and tween 20)로 3회 세척하였다. 이후, 다음과 같이 각각의 특이 항체로 웨스턴 블롯을 시행하여 분석하였다. The same amount of protein was taken for each condition, mixed with SDS-PAGE sample loading buffer (5X), heated at 100 ° C for 10 minutes, loaded and loaded on SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) Respectively. The proteins separated by electrophoresis were immobilized on an immobilon-P transfer membrane (0.45) at 350 mA for 120 minutes using a transfer buffer (20% methanol, 25 mM Tris-HCl, 192 mM glycine) (PVDF transfer membrane, Millipore Corporation, USA). Protein-transferred membranes were blocked with 3% non-fat dry milk (skim milk solution), reacted with primary antibody for 24 h at 4 ° C, and incubated with TBST (tris-buffered saline and tween 20 ) ≪ / RTI > Thereafter, Western blot analysis was performed with each specific antibody as follows.
사용한 1차 항체는 TNS3(Tensin 3) 1:3000 (Sigma Aldrich, U.S.A.), ERK, phospo-ERK, 분열된 카스파아제 3(cleaved-Caspase 3), Bcl-2, 총 Ras 1:3000(Cell signaling, Thecnology, Inc. Beverly, MA., U.S.A.), 뉴로피브로민(Neurofibromin), β-액틴 1:3000(Santa Cruz Biotechnology, INC, U.S.A.), 2차 항체로는 퍼록시다아제가 컨쥬게이트된 염소 항-토끼 항체(goat anti-rabbit antibody), 염소 항-마우스 항체(goat anti-mouse antibody) 1:5000 (Santa cruz Biotechnology, INC, USA)를 이용하여 반응시킨 뒤, WEST-ZOL Plus (iNtRON Biotechnology, KOREA)를 처리하여 필름에 현상하였다.The primary antibodies used were TNS3 (Tensin 3) 1: 3000 (Sigma Aldrich, USA), ERK, phospo-ERK, cleaved caspase 3, Bcl- , Neurofibromin, β-actin 1: 3000 (Santa Cruz Biotechnology, INC, USA), the second antibody was peroxidase-conjugated chlorine The cells were reacted with a goat anti-rabbit antibody, a goat anti-mouse antibody 1: 5000 (Santa Cruz Biotechnology, INC, USA), followed by WEST-ZOL Plus (iNtRON Biotechnology , KOREA) were processed and developed on the film.
실시예 3: Ras 활성 분석Example 3: Ras activity assay
Ras 활성을 측정하기 위해, Ras activation assay kit(Upstate Biotech. Lake Placid, NY)를 사용하여 친화 침전(affinity precipitation) 방식으로 GTP-Ras의 양을 분석하였다. 배양한 S462 세포를 용해 버퍼(25 mM HEPES pH 7.5, 150 mM NaCl, 1% Igepal CA-630, 10 mM MgCl2, 1 mM EDTA, 및 2% 글리세롤)에 용해한 후, 용해물(500 μg의 단백질 함유)을 Raf-1 RBD 융합 단백질과 결합하고 있는 아가로즈 비드(agarose bead)와 함께 4℃에서 1시간동안 반응시켰다. 아가로즈 비드를 1 ml의 용해 버퍼로 3회 세척한 후, 샘플 로딩 버퍼(loading buffer, 5X)와 혼합한 뒤, 100℃에서 10분 동안 가열하고, 이를 12-15% SDS-PAGE(sodium dodecyl sulfate-polyacrylamide gel electrophoresis)에 로딩하여 전기영동하였다. 아가로오스 비드는 원심분리하여 모은 후 용해 버퍼로 세척하였다. 최종적으로 샘플 버퍼를 첨가하여 95℃에서 5분간 가열한 후, Ras 특이 항체를 사용하여 웨스턴 블롯을 수행하여 GTP-Ras의 양을 분석하였다.In order to measure Ras activity, the amount of GTP-Ras was analyzed by affinity precipitation method using Ras activation assay kit (Upstate Biotech. Lake Placid, NY). The cultured S462 cells were dissolved in lysis buffer (25 mM HEPES pH 7.5, 150 mM NaCl, 1% Igepal CA-630, 10 mM MgCl 2 , 1 mM EDTA and 2% glycerol) ) Was reacted with agarose beads binding to Raf-1 RBD fusion protein at 4 ° C for 1 hour. The agarose beads were washed three times with 1 ml of dissolution buffer, mixed with a loading buffer (5X), heated at 100 ° C for 10 minutes, and subjected to 12-15% SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis). The agarose beads were collected by centrifugation and then washed with a dissolution buffer. Finally, a sample buffer was added and heated at 95 ° C for 5 minutes. Then, western blotting was performed using Ras specific antibody to analyze the amount of GTP-Ras.
실시예 4: TNS3 유전자 발현 억제용 짧은 간섭 RNA(short interfering RNA; siRNA) 디자인 및 제작Example 4: Design and production of short interfering RNA (siRNA) for inhibiting TNS3 gene expression
TNS3 유전자의 발현억제를 위해, TNS3 유전자 mRNA에 상보적으로 결합 가능한 siRNA를 디자인 및 합성하였다(Cosmogenetech, Korea). 센스(Sense)와 안티센스(Antisense)의 상보적인 2 가닥의 RNA-DNA 하이브리드 올리고머(hybrid oligomer ; 앞 19개 배열은 RNA이고 마지막 2개 TT는 DNA)를 각각 합성하여, 2가닥을 상보적으로 결합시킨 2중 가닥 RNA(double strand RNA)를 siRNA로 사용하였다. 이때 제조한 서열번호 1 및 2(#1)및 서열번호 3 및 4(#2)로 표시되는 siRNA의 센스와 안티센스의 염기서열은 하기 <표 1>에 나타난 바와 같았다.To suppress the expression of the TNS3 gene, siRNAs complementary to the TNS3 gene mRNA were designed and synthesized (Cosmogenetech, Korea). A complementary two-stranded RNA-DNA hybrid oligomer (Sense and Antisense) was prepared by synthesizing each of the preceding 19 sequences with RNA and the last 2 TTs with DNA, (Double strand RNA) was used as the siRNA. The nucleotide sequences of the sense and antisense siRNAs represented by SEQ ID NOS: 1 and 2 (# 1) and SEQ ID NOS: 3 and 4 (# 2) prepared at this time are shown in Table 1 below.
siRNAsiRNA 종류Kinds 염기서열Base sequence 서열번호SEQ ID NO:
siTNS3 #1 siTNS3 # 1 SenseSense 5‘-CCC AGC AAA GCG TTC AAA CTT-3 5'-CCC AGC AAA GCG TTC AAA CTT-3 1One
AntisenseAntisense 5‘-GUU UGA ACG CUU UGC UGG GTT-3’5'-GUU UGA ACG CUU UGC UGG GTT-3 ' 22
siTNS3 #2 siTNS3 # 2 Sense Sense 5‘-GGU CCG AAC ACU UGU ACA ATT-3’’ 5'-GGU CCG AAC ACU UGU ACA ATT-3 '' 33
AntisenseAntisense 5‘-UUG UAC AAG UGU UCG GAC CTT-3’5'-UUG UAC AAG UGU UCG GAC CTT-3 ' 44
실시예 5: 총 RNA 추출, cDNA 합성 및 정량 PCRExample 5: Total RNA Extraction, cDNA Synthesis and Quantitative PCR
TNS3의 발현량을 확인하기 위하여, 상기 실시예 1에서 배양한 세포에서 Trizol™ 시약(Invitrogen Co., U.S.A.)을 이용하여 RNA를 추출하고, 그 중 RNA 3 μg을 이용하여 DNase I(Invitrogen Co., U.S.A.) 3U을 처리하여 남아있는 gDNA를 제거한 후 cDNA를 합성하였고, 합성한 cDNA를 이용하여 PCR로 증폭하여 확인하였다. 사용한 프라이머(표 2)는, RNA가 PCR의 주형이 될 수 없으며 인트론 영역은 gDNA에만 존재하는 특성을 이용하여 제작하였다. PCR 결과 gDNA가 완전히 제거된 것으로 확인이 된 RNA를 TransScriptII Reverse Transcriptase PCR Kit(Qiagen, U.S.A.)를 이용하여 cDNA를 합성하였다. 이종이식 마우스에서 적출한 종양의 경우, 균질화기(Homogenizer)를 사용하여 종양을 분쇄한 후 Trizol 시약(Invitrogen Co., U.S.A.)을 이용하여 RNA를 추출하여 위와 같은 방법으로 cDNA를 합성하였다.To confirm the expression level of TNS3, RNA was extracted from the cells cultured in Example 1 using Trizol (TM) reagent (Invitrogen Co., USA), and DNase I (Invitrogen Co., USA) was added thereto using 3 μg of RNA. , USA). After removing the remaining gDNA, cDNA was synthesized and amplified by PCR using the synthesized cDNA. Primers used (Table 2) were constructed using the property that the RNA can not be a template for PCR and the intron region exists only in gDNA. CDNA was synthesized using TransScript II Reverse Transcriptase PCR Kit (Qiagen, USA). For tumors isolated from xenotransplantation mice, tumors were pulverized using a homogenizer, and RNA was extracted using Trizol reagent (Invitrogen Co., USA), and cDNA was synthesized as described above.
이와 같이 배양한 세포에서 총 RNA를 추출하여, 합성한 cDNA를 주형으로 하여 유전자 특이 프라이머를 사용하여 실시간 정량적 실시간 중합효소연쇄반응(quantitative real-time PCR)으로 증폭하였다. 이때, 이중 가닥 DNA에 결합하여 형광을 나타내는 STBR Green(TaKaRa, Shiga, Japan)을 cDNA(150 ng)에 첨가하여 형광강도를 ABI Prism 7000 Sequence Detection System(Applied Biosystems; Foster City, CA, U,S,A)로 측정하였다. 사용한 프라이머 서열은 하기 <표 2>에 나타난 바와 같았다.Total RNA was extracted from the cultured cells and amplified by real-time quantitative real-time PCR using the synthesized cDNA as a template using a gene-specific primer. STBR Green (TaKaRa, Shiga, Japan), which binds to double-stranded DNA and exhibits fluorescence, was added to cDNA (150 ng) and the fluorescence intensity was measured using an ABI Prism 7000 Sequence Detection System (Applied Biosystems; Foster City, CA, U, S , A). The primer sequences used were as shown in Table 2 below.
유전자gene 종류Kinds 염기서열Base sequence 서열번호SEQ ID NO:
HumanHuman 정방향Forward 5'-CGT TCT TTG GGC TCA GTC TC-3'5'-CGT TCT TTG GGC TCA GTC TC-3 ' 55
역방향Reverse 5'-CTG AAG CCT TGG AAA AGT CG-3'5'-CTG AAG CCT TGG AAA AGT CG-3 ' 66
MouseMouse 정방향Forward 5'-GAG GGG TGG TAA AGG ACG C-3'5'-GAG GGG TGG TAA AGG ACG C-3 ' 77
역방향Reverse 5'-GGA GGG CTC CAT TAA TGC TGA A-3'5'-GGA GGG CTC CAT TAA TGC TGAA-3 ' 88
실시예 6: 세포 생존율(Cell viability) 측정Example 6: Measurement of cell viability
세포 생존율은 D-Plus™ CCK cell viability assay kit(DonglinLS, Korea)를 사용하여 분석하였다. 96 웰 플레이트에 세포를 웰 당 7×10³세포 농도로 접종하고 siRNA(상기 표 1 참조) 또는/및 항암제 처리 후 24시간 동안 배양하였다. 이후 배지를 CCK 시약(10 μl)이 첨가된 배지(100 μl)로 교체하고 2시간 추가 배양하였다. ELISA 플레이트 리더기(Bio-Rad Model 680)를 이용하여 450 nm에서 흡광도를 측정하여 세포 생존율을 분석하였다.Cell viability was analyzed using the D-Plus CCK cell viability assay kit (Donglin LS, Korea). Cells were seeded at a cell density of 7 x 10 &lt; 3 &gt; cells per well in 96 well plates and cultured for 24 hours after siRNA (see Table 1 above) and / or anticancer treatment. Subsequently, the culture medium was replaced with 100 μl of a culture medium supplemented with 10 μl of CCK reagent and further cultured for 2 hours. Cell viability was determined by measuring the absorbance at 450 nm using an ELISA plate reader (Bio-Rad Model 680).
실시예 7 : 제1형 신경섬유종증 악성화 이종이식 마우스 모델 제작 및 짧은 간섭 RNA(short interfering RNA; siRNA) 투여Example 7: Formation of type 1 neurofibromatosis malignant xenograft mouse model and administration of short interfering RNA (siRNA)
7-1. 제1형 신경섬유종증 악성화 이종이식 마우스 모델 제작7-1. Generation of type 1 neurofibromatosis malignant xenograft mouse model
신경섬유육종 S462 세포를 1,000 rpm으로 5분 원심분리하여 상층액을 제거한 후 인산완충식염수(PBS)에 1×107 세포/ml로 준비한 후, 8주령 마우스 우측 등 부위의 피하에 0.1 ml씩 투여하여 이식하였다. 종양의 부피를 측정하여 약 80~120 mm3에 도달한 개체를 선별하고, 종양의 부피 및 체중을 기초로 하여 균등하도록 군을 분리하였다. Nerve fiber sarcoma S462 cells were centrifuged at 1,000 rpm for 5 minutes to remove the supernatant, followed by 1 × 10 7 cells / ml in phosphate-buffered saline (PBS), followed by subcutaneous injection of 0.1 ml Respectively. Tumor volume was measured and individuals reaching about 80 to 120 mm 3 were selected and grouped to equalize on the basis of tumor volume and body weight.
7-2. 항암제 및/또는 siRNA 투여7-2. Administration of anticancer agents and / or siRNA
상기 실시예 4에서 제조한 siTNS3(#2)를 5% 포도당 용액에 siTNS3(10 μg)와 in vivo-JETPEI(Polyplus, France)를 섞고 15분간 배양한 후 종양조직에 국부주입(local injection)하였다. siTNS3를 직접 종양에 투여하였으며, siTNS3 10 μg/100 μl를 주 3회(월, 수, 금) 3주간 투여하였다.SiTNS3 (# 2) prepared in Example 4 was mixed with siTNS3 (10 μg) and in vivo-JETPEI (Polyplus, France) in a 5% glucose solution for 15 minutes and then local injection was performed on the tumor tissue . siTNS3 was directly administered to the tumor, and siTNS3 10 μg / 100 μl was administered three times weekly (Monday, Wednesday, and Friday) for 3 weeks.
또한, 항암제 이포스파마이드(Ifosfamide), 카보플라틴(Carboplatin), 에토포시드(Etoposide)는 투여 농도에 따라 생리식염수와 섞어 조제하여 복강 내 주사(intraperitoneal injection)하였으며, 이때, 이포스파마이드(40 mg/kg)와 에토포시드(3.3 mg/kg)는 첫 주 5일간(월~금) 복강 투여하였고, 카보플라틴(13.3 mg/kg)은 첫 주 2일 간 (월, 화) 복강투여 하였다.In addition, ifosfamide, carboplatin, and etoposide were prepared by intraperitoneal injection with physiological saline according to the concentration of the drug, kg) and etoposide (3.3 mg / kg) were administered intraperitoneally for 5 days (Mon. through Fri) for the first week, and intraperitoneal administration of carboplatin (13.3 mg / Respectively.
7-3. 종양 부피 측정과 종양 적출 및 중량 분석7-3. Tumor volume measurement, tumor excision and weight analysis
관찰기간 동안 주 2회, 캘리퍼스(caliper)를 사용하여 종양의 장축(maximum length, L)과 단축(perpendicular width, W)을 측정하고, 하기 계산식 1에 대입하여 종양의 부피(tumor volume, TV)를 계산하였다. The maximum length (L) and the perpendicular width (W) of the tumor were measured using a caliper twice a week twice during the observation period, and the tumor volume (TV) Respectively.
[계산식 1][Equation 1]
TV(mm3)=L(mm)×W2(mm2)×1/2TV (mm 3 ) = L (mm) x W 2 (mm 2 ) x 1/2
또한, 마우스를 CO2가스로 안락사 시킨 후, 종양을 적출하여 중량을 측정하였다. 적출된 종양은 하기 계산식 2에 대입하여 종양 성장 억제율(tumor growth inhibition rate, IR)을 계산하였다. In addition, the mice were euthanized with CO 2 gas, and tumors were harvested and weighed. Tumor growth inhibition rate (IR) was calculated by substituting the extracted tumor into the following equation (2).
[계산식 2][Equation 2]
IR(%) = (1-T/C)×100 IR (%) = (1-T / C) 100
T: 실험군 및 대조군의 평균 종양 무게T: Mean tumor weight in experimental and control groups
C: 음성 대조군의 종양 무게C: Tumor weight of negative control
실시예 8: 통계 분석Example 8: Statistical analysis
In vitro 그래프의 유의성 검증을 위해 Student's t-test를 사용하였다. In vivo 실험에서 얻어진 체중, 종양의 부피 및 종양의 중량은 SAS(Version 9.3, SAS Institute Inc., USA)를 사용하여 검정하였다. 체중, 종양의 부피 및 종양의 중량은 Bartlett test를 실시하여 등분산성을 검정하였다(유의수준: < 0.05). 등분산성인 경우 one-way analysis of variance(ANOVA)를 실시(유의수준: < 0.05)하여 유의성이 관찰되면 음성 대조군에 대한 각 시험군의 유의성을 확인하기 위해 Dunnett's t-test의 다중검정을 실시하였다(유의수준: 단측 < 0.05 및 < 0.01). 등분산성이 기각되면 Kruskal-Wallis test를 실시(유의수준: < 0.05)하여 유의성이 관찰되면 음성 대조군에 대한 각 시험군의 유의성을 확인하기 위해 Steel's test의 다중검정을 실시하였다.Student's t-test was used to verify the significance of the in vitro graph. Body weights, tumor volume, and tumor weights obtained in the in vivo experiment were assayed using SAS (Version 9.3, SAS Institute Inc., USA). Body weights, tumor volume and tumor weights were tested by Bartlett test (significance level: <0.05). In the case of equal distribution, one-way analysis of variance (ANOVA) was performed (significance level: <0.05). When significance was observed, a multiple test of Dunnett's t-test was performed to confirm the significance of each test group for negative control (Significance level: one side <0.05 and <0.01). When the equal distribution was rejected, the Kruskal-Wallis test was performed (significance level: <0.05). When significance was observed, a multiple test of the steel's test was performed to confirm the significance of each test group for the negative control group.
실험예 1 : TNS3의 과발현 여부 확인Experimental Example 1: Determination of overexpression of TNS3
TNS3의 발현량을 확인하기 위해, 웨스턴 블롯 및 정량적 실시간 중합효소연쇄반응(quantitative real-time PCR)을 수행한 결과, 제1형 신경섬유종증 환자 유래의 악성종양 세포주에서 TNS3(tensin-like SH2 domain containing 1, Tensin 3)의 과발현되어 있음을 발견하였다. 즉, 정상 슈반 세포(HSC)와 NF1 악성종양 슈반 세포주(S462)에서의 Tensin 3 단백질과 TNS3 mRNA 발현량을 비교한 결과, 도 7a에 나타난 바와 같이, 악성 종양에서 TNS3 유전자의 발현이 증가되어 있음을 확인하였다. Tensin 3는 Rho GTPase 신호와 세포 접착 조절에 관여한다고 밝혀져 있으며, Src에 의해 인산화(활성화)되어 종양의 악성화에 관여하는 것으로 알려져 있다.To confirm the expression level of TNS3, Western blotting and quantitative real-time PCR were performed. As a result, malignant tumor cell lines derived from patients with type 1 neurofibromatosis contained TNS3 (tensin-like SH2 domain containing 1, and Tensin 3) overexpression. That is, the expression levels of Tensin 3 protein and TNS3 mRNA in normal schwann cells (HSC) and NF1 malignant tumor Schwann cell line (S462) were increased, and as shown in FIG. 7A, expression of TNS3 gene was increased in malignant tumors Respectively. Tensin 3 has been shown to be involved in Rho GTPase signaling and cell adhesion regulation, and is known to be phosphorylated (activated) by Src and is involved in malignant tumors.
또한, 도 7b에 나타난 바와 같이, 다른 종류의 악성 종양(MPNST) sNF02.2, sNF96.2 세포주에서도 TNS3 유전자의 발현이 증가되어 있음을 확인하였다. 따라서, TNS3(Tensin 3)의 발현 억제가 제1형 신경섬유종증 악성종양 치료에 좋은 효과가 있을 것이라고 가설을 세웠다.In addition, as shown in FIG. 7B, it was confirmed that the expression of TNS3 gene was also increased in other types of malignant tumors (MPNST) sNF02.2 and sNF96.2 cells. Therefore, it was hypothesized that inhibition of TNS3 (Tensin 3) expression would have a beneficial effect in the treatment of type 1 neurofibromatosis malignancy.
실험예 2 : NF1 악성 종양 슈반 세포주에서 siRNA의 ERK1/2와 RAS 활성화 억제 및 Bcl-2 발현 억제 효과Experimental Example 2 Inhibition of ERK1 / 2 and RAS Activation and Bcl-2 Expression of siRNA in NF1 Malignant Tumor Model
상기 실험예 1에서 얻은 결과를 토대로, 상기 실시예 4에서 제조한 siTNS3 #1, #2를 10 pmole/ml 농도로 RNAi MAX(Invitrogen) 시약을 사용하여 제1형 신경섬유종증 악성종양 슈반 세포주(S462)에 처리하여 TNS3 발현을 저해시킨 후, 웨스턴 블롯을 통해 ERK1/2 활성화(phospho-ERK1/2, p-ERK1/2) 여부와 Bcl-2 발현량을 확인하였다. 또한, RAS 활성화(GTP-RAS) 정도를 조사하였다.Based on the results obtained in Experimental Example 1, siTNS3 # 1 and # 2 prepared in Example 4 were cultured in the presence of type 1 neurofibromatous malignant tumor Schwann cell line (S462) using RNAi MAX (Invitrogen) ) To inhibit TNS3 expression. Western blot analysis revealed the presence of ERK1 / 2 activation (phospho-ERK1 / 2, p-ERK1 / 2) and Bcl-2 expression level. In addition, the degree of RAS activation (GTP-RAS) was examined.
그 결과, 도 8에 나타난 바와 같이, 신경섬유육종 세포주(S462)에 siTNS3 #1, #2 처리 시에 상기 ERK1/2 활성화(p-ERK1/2)와 Bcl-2 발현량이 모두 현저하게 감소됨을 확인하였다. 또한, siTNS3 #2 처리 시에는 RAS 활성화(GTP-RAS)가 현저하게 감소됨을 확인하였다. 이러한 결과는 TNS3의 발현량이 제1형 신경섬유종증 악성화와 밀접한 관련이 있으며, 본 발명의 siTNS3 #1, #2에 의한 TNS3의 발현 저해가 신경섬유종증 악성종양 치료에 좋은 효과가 있을 것임을 시사한다.As a result, as shown in FIG. 8, the ERK1 / 2 activation (p-ERK1 / 2) and Bcl-2 expression levels were remarkably decreased in the neurofilament sarcoma cell line (S462) when siTNS3 # Respectively. In addition, it was confirmed that RTP activation (GTP-RAS) was significantly reduced during siTNS3 # 2 treatment. These results suggest that the expression level of TNS3 is closely related to the type 1 neurofibromatosis and that inhibition of TNS3 expression by siTNS3 # 1 and # 2 of the present invention will be effective for the treatment of neurofibromatosis malignancy.
실험예 3 : NF1 악성종양 슈반 세포에서 siRNA에 의한 종양세포 사멸 효과Experimental Example 3: Effect of siRNA on tumor cell death in NF1 malignant tumor Schwann cells
먼저, 상기 실험예 2에 개시된 바와 같이, 제조한 siTNS3 #2를 처리하여 TNS3의 발현을 저해시킨 후, NF1 악성 종양 슈반 세포주(S462)의 변화를 관찰하였다. 이때 siTNS3 #2를 단독으로 처리하거나, 기존에 사용되는 대표적 항암제인 ICE(Ifosfamide 1.8 mM, Carboplatin 90 μM, Etoposide 30 μM)와 각각 10 pmole/ml 만큼 병용처리 하였고, 세포 생존율을 분석하였다.First, as described in Experimental Example 2, the siTNS3 # 2 thus prepared was treated to inhibit the expression of TNS3, and then the change of NF1 malignant tumor Schwann cell line (S462) was observed. At this time, siTNS3 # 2 was treated alone or in combination with ICE (Ifosfamide 1.8 mM, Carboplatin 90 μM, and Etoposide 30 μM), which are typical anticancer drugs used, by 10 pmole / ml, respectively, and cell viability was analyzed.
그 결과, 도 9에 나타난 바와 같이, S462 악성세포에 TNS3 유전자 siRNA(siTNS3 #2) 처리 시에 유의하게 세포 생존율이 감소되는 것을 확인하였으며, 대표적인 항암제인 ICE(Ifosfamide, Carboplatin, Etoposide)와 병용 처리 시에 시너지 효과가 있음을 확인하였다. 정상세포인 HSC 세포에 TNS3 유전자 siRNA(siTNS3 #2) 처리 시에는 세포 생존율이 감소와 ICE(Ifosfamide, Carboplatin, Etoposide)와 병용 치의 시너지 효과가 나타나지 않았다.As a result, as shown in FIG. 9, it was confirmed that the cell survival rate was significantly decreased upon treatment with TNS3 gene siRNA (siTNS3 # 2) in S462 malignant cells, and the cell survival rate was significantly decreased in combination with ICO (Ifosfamide, Carboplatin, Etoposide) It is confirmed that there is synergy effect in city. In HSC cells, TNS3 gene siRNA (siTNS3 # 2) treatment did not show a synergistic effect with the combination of ICE (Ifosfamide, Carboplatin, Etoposide) and cell viability.
실험예 4: 제1형 신경섬유종증 악성화 이종이식 마우스 모델에서 siRNA의 악성종양 억제 효과Experimental Example 4: Malignant tumor suppression effect of siRNA in mouse model of type 1 neurofibromatosis malignant xenograft
in vivo에서의 TNS 유전자 발현 억제 효능을 확인하기 위해, 실시예 7-1에 나타난 바와 같이, NF1 악성종양 슈반 세포주(S462)를 누드마우스에 접종하여 발암을 유도한 제1형 신경섬유종증 악성화 이종이식 마우스를 제작하였고, 실시예 7-2에 나타난 바와 같이, TNS3 유전자에 상보적으로 결합하는 siTNS3 #2 단독, 항암제 ICE 단독, siTNS3 #2와 항암제 ICE 병용(siRNA는 주 3회 local injection, 항암제는 첫 1회 복강투여)으로 21일까지 투여하여 43일간 종양의 크기를 관찰하였다. In order to confirm the inhibitory effect of TNS gene expression in vivo, as shown in Example 7-1, NF1 malignant tumor Schwannoma cell line (S462) was inoculated into nude mice to induce carcinogenesis of type 1 neurofibromatosis malignant xenograft Mouse, siTNS3 # 2 alone, chemotherapeutic agent ICE alone, siTNS3 # 2 and anti-cancer agent ICE (3 times a week, local injection, anticancer agent) were used as complementary binding to TNS3 gene as shown in Example 7-2 The first dose was administered to the abdomen for 21 days, and the size of the tumor was observed for 43 days.
그 결과, 도 10에 나타난 바와 같이, TNS3 유전자 siTNS3 #2 단독으로도 매우 뛰어난 항암작용(종양 조직의 증식 억제)이 관찰되었으며 항암제인 ICE와 병용 처리 시에 더 좋은 시너지 효과가 나타났다. 또한, 43일째 각 군의 평균 종양 부피와 무게를 비교하였을 시에 TNS3 유전자 siTNS3 #2 ICE 병용 처리군에서 현저한 종양 억제 효과가 나타났다. 더욱이, 도 11a 및 11b에 나타난 바와 같이, 종양의 외형과 적출한 종양의 크기를 비교하였을 경우에도 siTNS3 #2 및 ICE 병용 처리군에서 뛰어난 악성종양 억제 효과가 있었다.As a result, as shown in Fig. 10, excellent anticancer activity (tumor cell proliferation inhibition) was observed even with the TNS3 gene siTNS3 # 2 alone, and synergistic effect was further improved in combination with ICE, which is an anticancer agent. In addition, when the mean tumor volume and weight of each group were compared at 43 days, a significant tumor suppression effect was observed in the TNS3 gene siTNS3 # 2 ICE combination treatment group. Furthermore, as shown in Figs. 11A and 11B, even when the size of the tumor and the size of the tumor removed were compared, there was an excellent malignant tumor suppression effect in siTNS3 # 2 and ICE combination treatment group.
이상으로 본 발명 내용의 특정한 부분을 상세히 기술하였는 바, 당업계의 통상의 지식을 가진 자에게 있어서, 이러한 구체적 기술은 단지 바람직한 실시양태일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백하다. 즉, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다.While the present invention has been particularly shown and described with reference to specific embodiments thereof, those skilled in the art will appreciate that such specific embodiments are merely preferred embodiments and that the scope of the present invention is not limited thereby. Do. That is, the practical scope of the present invention is defined by the appended claims and their equivalents.

Claims (5)

  1. 서열번호 1 내지 서열번호 4로 표시되는 염기 서열 중 어느 하나로 이루어지는 짧은 간섭 RNA(short interfering RNA; siRNA)를 유효성분으로 함유하는 신경섬유육종(neurofibrosarcoma; malignant peripheral nerve sheath tumor, MPNST) 예방 또는 치료용 약학 조성물.A prophylactic or therapeutic agent for neurofibrosarcoma (MPNST) containing short interfering RNA (siRNA) consisting of any one of the nucleotide sequences of SEQ ID NOS: 1 to 4 as an active ingredient A pharmaceutical composition.
  2. 제 1 항에 있어서, 상기 신경섬유육종은 총상신경섬유종의 악성화로 유발되는 것을 특징으로 하는 신경섬유육종 예방 또는 치료용 약학 조성물.The pharmaceutical composition according to claim 1, wherein the neurofilament sarcoma is caused by malignant transformation of neurofibromatosis.
  3. 제 1 항에 있어서, 상기 siRNA는 TNS3 유전자의 발현을 억제함으로써 신경섬유육종을 예방 또는 치료하는 것을 특징으로 하는 신경섬유육종 예방 또는 치료용 약학 조성물.The pharmaceutical composition according to claim 1, wherein the siRNA inhibits the expression of TNS3 gene to prevent or treat neurofilament sarcoma.
  4. 제 1 항에 있어서, 상기 siRNA는 ERK1/2 및 RAS의 활성화 또는 Bcl-2의 발현을 억제함으로써 신경섬유육종을 예방 또는 치료하는 것을 특징으로 하는 신경섬유육종 예방 또는 치료용 약학 조성물.The pharmaceutical composition for preventing or treating neurofibromatosis according to claim 1, wherein the siRNA inhibits or treats neurofilament sarcoma by inhibiting ERK1 / 2 and RAS activation or Bcl-2 expression.
  5. 제 1 항에 있어서, 상기 조성물은 이소파마이드(Isofamide), 카보플라틴(Carboplatin) 및 에토포시드(Etoposide)로 이루어진 군에서 선택된 어느 하나 이상의 항암제와 병용 투여하는 것을 특징으로 하는 신경섬유육종 예방 또는 치료용 약학 조성물.The method according to claim 1, wherein the composition is administered in combination with any one or more anticancer agents selected from the group consisting of isopamide, carboplatin, and etoposide. Or &lt; / RTI &gt;
PCT/KR2018/014954 2017-12-05 2018-11-29 Composition comprising sirna for prevention or treatment of neurofibrosarcoma WO2019112242A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20170165869 2017-12-05
KR10-2017-0165869 2017-12-05
KR10-2018-0031589 2018-03-19
KR1020180031589A KR101894769B1 (en) 2017-12-05 2018-03-19 Composition Comprising Short Interfering RNAs for Preventing or Treating Neurofibrosarcoma

Publications (1)

Publication Number Publication Date
WO2019112242A1 true WO2019112242A1 (en) 2019-06-13

Family

ID=63598277

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2018/014954 WO2019112242A1 (en) 2017-12-05 2018-11-29 Composition comprising sirna for prevention or treatment of neurofibrosarcoma

Country Status (2)

Country Link
KR (1) KR101894769B1 (en)
WO (1) WO2019112242A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101894769B1 (en) * 2017-12-05 2018-09-04 아주대학교산학협력단 Composition Comprising Short Interfering RNAs for Preventing or Treating Neurofibrosarcoma

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101894769B1 (en) * 2017-12-05 2018-09-04 아주대학교산학협력단 Composition Comprising Short Interfering RNAs for Preventing or Treating Neurofibrosarcoma

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA022611B1 (en) 2008-06-27 2016-02-29 Индиана Юниверсити Рисерч Энд Текнолоджи Корпорейшн Method for treating plexiform neurofibroma

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101894769B1 (en) * 2017-12-05 2018-09-04 아주대학교산학협력단 Composition Comprising Short Interfering RNAs for Preventing or Treating Neurofibrosarcoma

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CARTER, J. A. O: "CpG dinucleotide-specific hypermethylation of the TNS3 gene promoter in human renal cell carcinoma", EPIGENETICS, 1 July 2013 (2013-07-01), pages 739 - 747, XP055614831 *
KATZ, M.: "A reciprocal tensin-3-cten switch mediates EGF-driven mammary cell migration", NAT CELL BIOL., vol. 9, no. 8, August 2007 (2007-08-01), pages 961 - 969,1-5, XP002496878 *
MARTUSZEWSKA, D: "Tensin3 Is a Negative Regulator of Cell Migration and All Four Tensin Family Members Are Downregulated in Human Kidney Cancer", PLOS ONE, vol. 4, no. 2, e4350, 4 February 2009 (2009-02-04), pages 1 - 13, XP055614830 *
QIAN, X.: "The tensin-3 protein, including its SH2 domain, is phosphorylated by Src and contributes to tumorigenesis and metastasis", CANCER CELL, vol. 16, no. 3, 8 September 2009 (2009-09-08), pages 246 - 258, XP055614700 *

Also Published As

Publication number Publication date
KR101894769B1 (en) 2018-09-04

Similar Documents

Publication Publication Date Title
US10987317B2 (en) Inhibition of glutaminase C
Chang et al. Specificity protein 1-modulated superoxide dismutase 2 enhances temozolomide resistance in glioblastoma, which is independent of O6-methylguanine-DNA methyltransferase
Carén et al. The good, the bad and the ugly: epigenetic mechanisms in glioblastoma
KR100775549B1 (en) A therapeutic agent containing niacinamide or its derivatives as an effective ingredient for prevention and treatment of cancer
CN111073979A (en) Gastric cancer treatment method for blocking CCL28 chemotactic pathway
CN114796226B (en) Application of Olaparib in inducing nucleolus stress
Chiaradonna et al. RAS and PKA pathways in cancer: new insight from transcriptional analysis
WO2019112242A1 (en) Composition comprising sirna for prevention or treatment of neurofibrosarcoma
Kitazono et al. The histone deacetylase inhibitor FR901228 (depsipeptide) restores expression and function of pseudo-null p53
WO2019112243A1 (en) Composition containing interferon-gamma as active ingredient for prevention or treatment of neurofibroma
Zheng et al. The influence of the p53 gene on the in vitro chemosensitivity of colorectal cancer cells
EP2101747B1 (en) Cancer chemoprevention strategy based on loss of imprinting of igf2
CN115054605B (en) Application of G9a inhibitor in preparation of medicines for treating uveal melanoma
George et al. Loss of p21Waf1/Cip1/Sdi1 enhances intestinal stem cell survival following radiation injury
CN110585184A (en) Treatment of cancer
US11642362B2 (en) Methods of inhibiting cell proliferation and METTL8 activity
US20220145303A1 (en) Fragile x mental retardation protein interfering oligonucleotides and methods of using same
KR101771924B1 (en) Method for Screening a Composition for Preventing or Treating cancer using BRD4-E2F2 cell cycle regulation
CN115960018B (en) EGFR inhibitor, composition and application thereof
WO2020246717A1 (en) Pharmaceutical composition containing heres expression inhibitor for preventing or treating squamous cell carcinomamodulator
CN114085832B (en) SiRNA molecules for inhibiting PRR14 gene
WO2023020561A1 (en) Application of benzisoquinoline diketone compound in treatment of tumor
KR101902845B1 (en) Composition for preventing or treating of neurofibrosarcoma
KR101913537B1 (en) Anti-cancer agents for preventing or treating of neurofibrosarcoma
Peralta-Arrieta et al. Epigenetica en carcinomas pulmonares de células no pequeñas

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: 18885224

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: 18885224

Country of ref document: EP

Kind code of ref document: A1