WO2023101457A1 - Composition pour la prévention ou le traitement du cancer des voies biliaires - Google Patents

Composition pour la prévention ou le traitement du cancer des voies biliaires Download PDF

Info

Publication number
WO2023101457A1
WO2023101457A1 PCT/KR2022/019329 KR2022019329W WO2023101457A1 WO 2023101457 A1 WO2023101457 A1 WO 2023101457A1 KR 2022019329 W KR2022019329 W KR 2022019329W WO 2023101457 A1 WO2023101457 A1 WO 2023101457A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
traf3
pum1
acid
present
Prior art date
Application number
PCT/KR2022/019329
Other languages
English (en)
Korean (ko)
Inventor
송시영
정다운
조중현
박수빈
김상우
서미경
Original Assignee
연세대학교 산학협력단
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 연세대학교 산학협력단 filed Critical 연세대학교 산학협력단
Publication of WO2023101457A1 publication Critical patent/WO2023101457A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/27Esters, e.g. nitroglycerine, selenocyanates of carbamic or thiocarbamic acids, meprobamate, carbachol, neostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • the present invention relates to a composition for preventing or treating bile duct cancer, and can provide a method for maximizing drug effects by simultaneously diagnosing a patient's genetic characteristics.
  • the bile duct is a tube that carries bile produced in the liver to the duodenum.
  • the sac that temporarily stores and concentrates bile is called the gallbladder, and the bile duct and the gallbladder are collectively called the biliary tract.
  • Biliary Tract Cancer (BTC) also called cholangiocarcinoma, is a malignant tumor arising from the epithelium of the bile duct.
  • Bile duct cancer is largely classified into extrahepatic cholangiocarcinoma, intrahepatic cholangiocarcinoma, and gallbladder cancer. It is one of the incurable cancers that are very difficult to treat due to its low survival rate. As described above, since cholangiocarcinoma is discovered after a significant progression of the cancer and has a poor prognosis and is difficult to treat, early diagnosis and treatment of cholangiocarcinoma are urgently needed.
  • the treatment method is selected in consideration of the size, location, stage of the cancer, age and health condition of the patient, etc., and one or several methods are used in combination.
  • biliary tract cancer is one of the most lethal malignancies with a 5-year survival rate of less than 20%.
  • gemcitabine is known to be most effective in biliary tract cancer. Combination therapy is being performed, but there is a limitation that it is not effective in all patients with cholangiocarcinoma.
  • the inventors of the present invention completed the present invention by discovering a new therapeutic target drug capable of precisely providing treatment according to the genetic characteristics of patients with bile duct cancer.
  • One object of the present invention is to provide a composition for determining the suitability of a therapeutic agent for cholangiocarcinoma.
  • Another object of the present invention is to provide a kit for determining the suitability of a therapeutic agent for bile duct cancer.
  • Another object of the present invention is to provide a method for providing information on the suitability of a therapeutic agent for cholangiocarcinoma.
  • Another object of the present invention is to provide a diagnostic device that provides information on the suitability of a therapeutic agent for cholangiocarcinoma.
  • Another object of the present invention is to provide a composition for preventing, improving or treating cholangiocarcinoma and a method for preventing, ameliorating or treating cholangiocarcinoma.
  • Another object of the present invention is to provide a method for accompanying diagnosis of a subject for application as a therapeutic agent for bile duct cancer.
  • composition for determining the suitability of a cancer treatment agent it relates to a composition for determining the suitability of a cancer treatment agent.
  • the composition for discrimination may include an agent for measuring the expression level of a pumilio RNA binding family member 1-TNF receptor associated factor 3 (PUM1-TRAF3) fusion protein or a gene encoding the same.
  • PUM1-TRAF3 pumilio RNA binding family member 1-TNF receptor associated factor 3
  • the fusion protein or the fusion gene encoding the same is caused by a kind of genetic mutation, and the fusion gene is a type of genetic mutation, which is generated when two genes existing at different positions are arranged side by side.
  • the fusion gene When the fusion gene occurs, it produces a chimeric protein that affects normal cell proliferation. That is, the fusion gene forms a chimera protein, and the fusion protein causes a modified activity different from that of the existing protein and acts as a principle to affect cell growth.
  • a fusion gene arranged side by side causes a problem in that the later gene is affected by the promoter of the preceding gene and is expressed abnormally.
  • CML Philadelphia chromosome
  • the PUM1-TRAF3 fusion protein of the present invention is a PUF (pumilio RNA binding family member 1) that plays a certain role in cell division, differentiation and development, PUM1 (pumilio RNA binding family member 1) and the TNF receptor (TNFR) superfamily It is a protein in which some sites of TRAF3 (TNF receptor associated factor 3), which is associated with members of and mediates signal transduction therefrom, exist in a fused form.
  • the PUM1-TRAF3 fusion protein of the present invention has a 3'-end truncated PUM1 (pumilio RNA binding family member 1) gene fragment sequence on the 5' side, and TRAF3 (TNF receptor associated factor 3) gene fragment sequence on the 3' side.
  • PUM1-TRAF3 fusion protein encoded by a PUM1-TRAF3 fusion gene having a fragment sequence on the 3' side, the 3'-end is truncated on the 5' side and exon 14 is deleted
  • PUM1 (pumilio RNA binding family member 1)
  • PUM1-TRAF3 fusion protein encoded by a PUM1-TRAF3 fusion gene having a fragment sequence of a gene and having a 3' fragment sequence of a TRAF3 (TNF receptor associated factor 3) gene on the 3' side.
  • TRAF3 TRAF3 receptor associated factor 3
  • the amino acid sequence of the PUM1-TRAF3 fusion protein may include at least one of the sequences represented by SEQ ID NO: 1 or SEQ ID NO: 2, more preferably the sequences represented by SEQ ID NO: 1 and SEQ ID NO: 2 It may include all, but is not limited thereto.
  • the nucleic acid base sequence encoding the PUM1-TRAF3 fusion protein may include at least one of the sequences represented by SEQ ID NO: 3 or SEQ ID NO: 4, more preferably SEQ ID NO: 3 and SEQ ID NO: 4. It may include all of the sequences shown, but is not limited thereto.
  • the agent for measuring the level of the PUM1-TRAF3 fusion protein of the present invention consists of an antibody, oligopeptide, ligand, PNA (peptide nucleic acid) and aptamer that specifically bind to the PUM1-TRAF3 fusion protein. It may include at least one selected from the group, but is not limited thereto.
  • the "antibody” of the present invention refers to a protein molecule capable of specifically binding to the PUM1-TRAF3 fusion protein, and the form of the antibody is not particularly limited, but polyclonal antibody, monoclonal antibody, or antigen binding. If it has, it may be included even if it is part of an antibody, and all types of immunoglobulin antibodies may be included. In addition, special antibodies such as humanized antibodies may be included, and the antibodies include functional fragments of antibody molecules as well as complete forms having two full-length light chains and two full-length heavy chains.
  • a functional fragment of an antibody molecule refers to a fragment having at least an antigen-binding function, and may include Fab, F(ab'), F(ab') 2, Fv, etc., but is not limited thereto.
  • the "oligopeptide” is a peptide composed of 2 to 20 amino acids and may include a dipeptide, tripeptide, tetrapeptide and pentapeptide, but is not limited thereto.
  • PNA Peptide Nucleic Acid
  • DNA has a phosphate-ribose backbone
  • PNA has a repeated N-(2-aminoethyl)-glycine backbone linked by peptide bonds, which greatly increases the binding force and stability to DNA or RNA, and is thus used in molecular biology. , diagnostic assays and antisense therapies.
  • PNA is described by Nielsen PE, Egholm M, Berg RH, Buchardt O (December 1991). "Sequence-selective recognition of DNA by strand displacement with a thymine-substituted polyamide". Science 254 (5037): 1497-1500.
  • the "aptamer" of the present invention refers to a single-stranded oligonucleotide, and refers to a nucleic acid molecule having binding activity to the PUM1-TRAF3 fusion protein.
  • the aptamer may have various three-dimensional structures according to its base sequence, and may have high affinity for a specific substance, such as an antigen-antibody reaction.
  • An aptamer can inhibit the activity of a given target molecule by binding to the given target molecule.
  • the aptamer may be RNA, DNA, modified nucleic acid, or a mixture thereof, and may be linear or cyclic in shape.
  • the agent for measuring the expression level of the gene encoding the PUM1-TRAF3 fusion protein of the present invention is one selected from the group consisting of primers, probes and antisense nucleotides that specifically bind to the gene encoding the PUM1-TRAF3 fusion protein It may include the above, but is not limited thereto.
  • the "primer” is a fragment that recognizes a target gene sequence, and includes a forward and reverse primer pair, preferably a primer pair that provides an analysis result having specificity and sensitivity. High specificity can be imparted when the nucleic acid sequence of the primer is a sequence that is inconsistent with the non-target sequence present in the sample, so that the primer amplifies only the target gene sequence containing a complementary primer binding site and does not cause non-specific amplification. .
  • the "probe” means a substance that can specifically bind to a target substance to be detected in a sample, and means a substance that can specifically confirm the presence of a target substance in a sample through the binding.
  • the type of probe is not limited as a material commonly used in the art, but preferably may be peptide nucleic acid (PNA), locked nucleic acid (LNA), peptide, polypeptide, protein, RNA or DNA, and most preferably Most likely it is PNA.
  • the probe is a biomaterial, including one derived from or similar to a living organism or manufactured in vitro, for example, enzymes, proteins, antibodies, microorganisms, animal and plant cells and organs, nerve cells, DNA, and It may be RNA, DNA includes cDNA, genomic DNA, and oligonucleotides, RNA includes genomic RNA, mRNA, and oligonucleotides, and examples of proteins may include antibodies, antigens, enzymes, peptides, and the like.
  • LNA Locked nucleic acids
  • LNA nucleosides contain the common nucleic acid bases of DNA and RNA, and can base pair according to the Watson-Crick base pairing rules. However, due to molecular 'locking' due to the methylene bridge, the LNA does not form an ideal shape in the Watson-Crick bond.
  • LNAs When LNAs are included in DNA or RNA oligonucleotides, they can more rapidly pair with complementary nucleotide chains to increase the stability of the double helix.
  • the "antisense” refers to a sequence of nucleotide bases in which an antisense oligomer is hybridized with a target sequence in RNA by Watson-Crick base pairing, allowing the formation of a heteroduplex of mRNA and RNA oligomers in the target sequence, and It refers to an oligomer having an inter-subunit backbone. Oligomers may have exact sequence complementarity or near complementarity to the target sequence.
  • composition for discrimination of the present invention compares the expression level of the PUM1-TRAF3 fusion protein or the gene encoding it measured in a biological sample isolated from a target object with a control group to determine whether the expression level is increased or decreased, thereby treating cancer suitability can be diagnosed.
  • the "determination" means to confirm the presence or characteristics of a pathological state, and more specifically, to determine the suitability of a therapeutic agent by confirming the existence or characteristics of a pathological state, in particular, accompanies for application to customized treatment
  • Companion Diagnostics refers to pre-selecting target patients for the purpose of diagnosis and determining whether or not there is a high possibility that a specific treatment will have a good treatment effect.
  • the companion diagnostics is a test that selects a subject for a target therapeutic agent in advance, and is performed by examining the genetic characteristics of the subject, more specifically, the level of expression of a gene or protein, the presence or absence of a specific gene, etc. It can be. This can provide essential information for effective use of the target drug, and can monitor the response during future treatment by predicting or treating the subject's drug reactivity, drug sensitivity, and the possibility of drug side effects through accompanying diagnosis. there is. Recently, in the field of anticancer drugs, companion diagnostics for personalized treatment are in the limelight.
  • the efficiency of cancer treatment can be increased or the side effects of the treatment can be reduced, and furthermore, the economic effect of preventing unnecessary medical expenses is also achieved. can be expected
  • the cancer therapeutic agent refers to a candidate drug that inhibits proliferation of cancer cells, invasion or metastasis of cancer cells, and prevents or treats cancer-related diseases even if it corresponds to drugs for preventing or treating diseases other than cancer. It may be included if it corresponds to a drug with a predicted effect, preferably a drug that inhibits nuclear factor kappa-light-chain-enhancer of activated B cells (NF- ⁇ B) or NF- ⁇ B-induced kinase (A drug that inhibits NF- ⁇ B-inducing kinase; NIK) may be an NF- ⁇ B inhibitor or a NIK inhibitor, but is not limited thereto as long as it is a drug that is expected to have a preventive or therapeutic effect on cancer-related diseases.
  • a drug that inhibits nuclear factor kappa-light-chain-enhancer of activated B cells (NF- ⁇ B) or NF- ⁇ B-induced kinase NF- ⁇ B-inducing kinas
  • the NF- ⁇ B inhibitor is QNZ or a pharmaceutically acceptable salt thereof; MG132 or a pharmaceutically acceptable salt thereof; At least one selected from the group consisting of pyrrolidinedithiocarbamate ammonium or a pharmaceutically acceptable salt thereof and caffeic acid phenethyl ester or a pharmaceutically acceptable salt thereof; It can be done, but is not limited thereto.
  • the QNZ is 4-N-[2-(4-phenoxyphenyl)ethyl]-1,2-dihydroquinazoline-4,6-diamine represented by Formula 1 (4-N-[2 -(4-Phenoxyphenyl)ethyl]-1,2-dihydroquinazoline-4,6-diamine).
  • the MG132 is benzyl[(2S)-4-methyl-1- ⁇ [(2S)-4-methyl-1- ⁇ [(2S)-4-methyl-1-oxopentane represented by Formula 2 -2-yl]amino ⁇ -1-oxopentan-2-yl]amino ⁇ -1-oxopentan-2-yl]carbamate (Benzyl [(2S)-4-methyl-1- ⁇ [(2S)- 4-methyl-1- ⁇ [(2S)-4-methyl-1-oxopentan-2-yl]amino ⁇ -1-oxopentan-2-yl]amino ⁇ -1-oxopentan-2-yl]carbamate) there is.
  • the pyrrolidinedithiocarbamate ammonium may be pyrrolidine-1-carbodithioate represented by Formula 3 below.
  • the caffeic acid phenethyl ester is 2-phenylethyl (E) -3- (3,4-dihydroxyphenyl) prop-2-inoate represented by the following formula (4) -3-(3,4-dihydroxyphenyl)prop-2-enoate).
  • the pharmaceutically acceptable salts may include acid or base addition salts, and stereochemical isomers thereof.
  • the compounds may be in the form of addition salts of organic or inorganic acids. Salts have desirable effects on patients when administered to patients, and include any salts that retain the activity of their parent compound, but may not be particularly limited thereto.
  • salts include inorganic and organic salts such as acetic acid, nitric acid, aspartic acid, sulfonic acid, sulfuric acid, maleic acid, glutamic acid, formic acid, succinic acid, phosphoric acid, phthalic acid, tannic acid, tartaric acid, hydrobromic acid, propionic acid, benzenesulfonic acid, Benzoic acid, stearic acid, lactic acid, bicarboxylic acid, bisulfuric acid, bitartaric acid, oxalic acid, butyric acid, calcium idete, carbonic acid, chlorobenzoic acid, citric acid, idetic acid, toluenesulfonic acid, fumaric acid, gluseptic acid, ecilin Acid, pamoic acid, gluconic acid, methyl nitric acid, malonic acid, hydrochloric acid, hydroiodoic acid, hydroxynaphtholic acid, isethionic acid, lactobi
  • Addition salts of bases include salts of alkali metals or alkaline earth metals, such as salts of ammonium, lithium, sodium, potassium, magnesium, calcium and the like; salts with organic bases such as benzathine, N-methyl-D-glucamine, hydrabamine and the like; and salts with amino acids such as arginine, lysine, and the like. Also, these salts can be converted to the free form by treatment with an appropriate base or acid.
  • the NIK inhibitor is B022 or a pharmaceutically acceptable salt thereof; XT2 or a pharmaceutically acceptable salt thereof; And NIK SMI1 or a pharmaceutically acceptable salt thereof; may include at least one selected from the group consisting of, but is not limited thereto.
  • B022 is 4-[1-(2-amino-5-chloropyrimidin-4-yl)-2,3-dihydroindol-6-yl]-2-(1 represented by Formula 5) ,3-thiazol-2-yl)but-3-yn-2-ol (4-[1-(2-amino-5-chloropyrimidin-4-yl)-2,3-dihydroindol-6-yl]- 2-(1,3-thiazol-2-yl)but-3-yn-2-ol).
  • the XT2 is (R) -4- (3- (2-amino-5H-pyrrolo [3,2-d] pyrimidin-7-yl) -4-fluorophenyl represented by the following formula (6) )-2-(thiazol-2-yl)but-3-yn-2-ol ((R)-4-(3-(2-Amino-5H-pyrrolo[3,2-d]pyrimidin-7- yl)-4-fluorophenyl)-2-(thiazol-2-yl)but-3-yn-2-ol).
  • the NIK SMI1 is 6-[3-[2-[(3R)-3-hydroxy-1-methyl-2-oxopyrrolidin-3-yl]ethynyl]phenyl represented by Formula 7 below ]-4-methoxypyridine-2-carboxamide (6-[3-[2-[(3R)-3-hydroxy-1-methyl-2-oxopyrrolidin-3-yl]ethynyl]phenyl]-4- methoxypyridine-2-carboxamide).
  • the pharmaceutically acceptable salts may include acid or base addition salts, and stereochemical isomers thereof.
  • the compounds may be in the form of addition salts of organic or inorganic acids. Salts have desirable effects on patients when administered to patients, and include any salts that retain the activity of their parent compound, but may not be particularly limited thereto.
  • salts include inorganic and organic salts such as acetic acid, nitric acid, aspartic acid, sulfonic acid, sulfuric acid, maleic acid, glutamic acid, formic acid, succinic acid, phosphoric acid, phthalic acid, tannic acid, tartaric acid, hydrobromic acid, propionic acid, benzenesulfonic acid, Benzoic acid, stearic acid, lactic acid, bicarboxylic acid, bisulfuric acid, bitartaric acid, oxalic acid, butyric acid, calcium idete, carbonic acid, chlorobenzoic acid, citric acid, idetic acid, toluenesulfonic acid, fumaric acid, gluseptic acid, ecilin Acid, pamoic acid, gluconic acid, methyl nitric acid, malonic acid, hydrochloric acid, hydroiodoic acid, hydroxynaphtholic acid, isethionic acid, lactobi
  • Addition salts of bases include salts of alkali metals or alkaline earth metals, such as salts of ammonium, lithium, sodium, potassium, magnesium, calcium and the like; salts with organic bases such as benzathine, N-methyl-D-glucamine, hydrabamine and the like; and salts with amino acids such as arginine, lysine, and the like. Also, these salts can be converted to the free form by treatment with an appropriate base or acid.
  • the "subject” is a mammal including humans, for example, humans, rats, mice, guinea pigs, hamsters, rabbits, monkeys, dogs, cats, cows, horses, pigs, sheep and goats. It may be selected from, but may be preferably a human, but is not limited thereto.
  • cancer refers to or refers to a physiological condition typically characterized by unregulated cell growth in mammals.
  • the cancer is biliary tract cancer, gastric cancer, thyroid cancer, parathyroid cancer, ovarian cancer, colon cancer, pancreatic cancer, liver cancer, breast cancer, cervical cancer, lung cancer, non-small cell lung cancer, prostate cancer, gallbladder cancer, biliary tract cancer, non-Hodgkin's lymphoma, Hodgkin's lymphoma, blood cancer, bladder cancer, kidney cancer, melanoma, colon cancer, bone cancer, skin cancer, head cancer, uterine cancer, rectal cancer, brain tumor, perianal cancer, fallopian tube carcinoma, endometrial carcinoma, vaginal cancer, vulvar carcinoma, esophageal cancer, small intestine cancer , endocrine adenocarcinoma, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, ureteral cancer, renal cell carcinoma, renal pelvic
  • the cholangiocarcinoma is also referred to as cholangiocarcinoma, and is a tumor arising from the epithelium of the bile duct, and is divided into intrahepatic cholangiocarcinoma and extrahepatic cholangiocarcinoma according to its location.
  • intrahepatic cholangiocarcinoma it is distinguished from hepatocellular carcinoma, which we commonly call liver cancer.
  • cholangiocarcinoma occurs frequently in the age group of 60 years or older, and is known to occur 1.3 times more often in men than in women. Since biliary obstruction progresses slowly, when cancer is clinically diagnosed, 70 to 80% of cancers are already quite advanced. .
  • the composition for determining the suitability of the cancer therapeutic agent is non-canonical induced by expression of a PUM1-TRAF3 fusion gene by using a PUM1-TRAF3 fusion marker as a new target marker related to cancer, particularly cholangiocarcinoma, preferably cholangiocarcinoma. It is effective in selecting subjects with genetic characteristics capable of maximizing the therapeutic effect of cholangiocarcinoma because it can determine the suitability of a therapeutic agent with a mechanism that suppresses the activation of NF- ⁇ B signaling (non-canonical NF- ⁇ B signaling).
  • the present invention relates to a kit for determining the suitability of the cancer therapeutic agent of the present invention.
  • the kit of the present invention can be predicted to be suitable for cancer treatment when the PUM1-TRAF3 fusion protein or the gene encoding it is present at a higher level in a subject using the composition of the present invention than a control group.
  • a control group nuclear factor kappa-light-chain-enhancer of activated B cells; NF- ⁇ B ) inhibitor or a candidate drug corresponding to an NF- ⁇ B-inducing kinase (NIK) inhibitor can be predicted to have therapeutic suitability.
  • control group is the expression level of the corresponding biomarker protein or the gene encoding the protein in a normal control group, or the average expression level of the corresponding marker protein or the gene encoding the same in a biological sample derived from a patient with a biliary tract disease.
  • value or median value or may be the average value or median value of the expression level of the corresponding marker protein or gene encoding it in biological samples derived from cancer patients, preferably from patients with other carcinomas other than cholangiocarcinoma, but are not limited thereto.
  • the kit of the present invention predicts that the target subject has suitability for cancer treatment when the expression level of the PUM1-TRAF3 fusion protein or the gene encoding it is higher than that of a normal control in the target subject using the composition of the present invention.
  • the description of the PUM1-TRAF3 fusion protein or the gene encoding it, cancer, cancer treatment, NF- ⁇ B inhibitor, NIK inhibitor, etc. is the same as that described in the composition for discrimination, so excessive omitted to avoid complexity.
  • the kit of the present invention may be an RT-PCR kit, a DNA chip kit, an ELISA kit, a protein chip kit, a Rapid kit, or a Multiple Reaction Monitoring (MRM) kit, but is not limited thereto.
  • MRM Multiple Reaction Monitoring
  • the kit of the present invention may further include one or more other component compositions, solutions or devices suitable for the assay method.
  • the kit may further include essential elements necessary for carrying out the reverse transcription polymerase reaction.
  • the reverse transcription polymerase reaction kit contains a pair of primers specific for a gene encoding a marker protein.
  • the primer is a nucleotide having a sequence specific to the nucleic acid sequence of the gene, and may have a length of about 7 bp to 50 bp, more preferably about 10 bp to 30 bp.
  • a primer specific for the nucleic acid sequence of the control gene may be included.
  • reverse transcription polymerase reaction kits contain a test tube or other suitable container, reaction buffer (with varying pH and magnesium concentration), deoxynucleotides (dNTPs), enzymes such as Taq-polymerase and reverse transcriptase, DNase and RNase inhibitor DEPC. -Water (DEPC-water), sterilized water, etc. may be included.
  • the kit for discrimination of the present invention may include essential elements required to perform DNA chip.
  • the DNA chip kit may include a substrate to which cDNA or oligonucleotides corresponding to genes or fragments thereof are attached, and reagents, reagents, enzymes, and the like for producing fluorescently labeled probes.
  • the substrate may include a cDNA or oligonucleotide corresponding to a control gene or a fragment thereof.
  • the kit for discrimination of the present invention may include essential elements necessary for performing ELISA.
  • ELISA kits contain antibodies specific for the protein.
  • An antibody is an antibody that has high specificity and affinity for a marker protein and little cross-reactivity to other proteins, and is a monoclonal antibody, polyclonal antibody, or recombinant antibody.
  • the ELISA kit may include antibodies specific for a control protein.
  • Other ELISA kits include reagents capable of detecting bound antibodies, such as labeled secondary antibodies, chromophores, enzymes (eg, conjugated with antibodies) and substrates thereof or those capable of binding the antibody. may contain other substances and the like.
  • the fixture for the antigen-antibody binding reaction includes a nitrocellulose film, a PVDF film, a well plate made of polyvinyl resin or polystyrene resin, and a glass slide. Glass or the like may be used, but is not limited thereto.
  • the marker of the secondary antibody in the kit for discrimination of the present invention is preferably a conventional coloring agent that reacts with color, and HRP (horseradish peroxidase), basic dephosphorylation enzyme (alkaline phosphatase), colloid gold, FITC (Poly L-lysine-fluorscein isothiocyanate), RITC (rhodamine-B-isothiocyanate), and other fluorescent substances (fluorescein) and dyes (dyes) may be used, but are not limited thereto no.
  • HRP horseradish peroxidase
  • basic dephosphorylation enzyme alkaline phosphatase
  • colloid gold FITC (Poly L-lysine-fluorscein isothiocyanate), RITC (rhodamine-B-isothiocyanate), and other fluorescent substances (fluorescein) and dyes (dyes)
  • fluorescein fluorescein
  • dyes dyes
  • a chromogenic substrate for inducing color development in the kit for discrimination of the present invention according to a label that produces a color reaction, such as TMB (3,3',5,5'-tetramethyl bezidine), ABTS [2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)], OPD (o-phenylenediamine), etc. can be used.
  • the chromogenic substrate is provided in a state dissolved in a buffer solution (0.1 M NaAc, pH 5.5).
  • a chromogenic substrate such as TMB is degraded by HRP used as a marker for the secondary antibody conjugate to generate chromogenic deposits, and the presence or absence of the marker proteins is detected by visually checking the degree of chromogenic deposits.
  • the washing solution preferably includes a phosphate buffer solution, NaCl and Tween 20, and a buffer solution (PBST) composed of 0.02 M phosphate buffer solution, 0.13 M NaCl, and 0.05% Tween 20 this is more preferable
  • PBST buffer solution
  • the washing solution is washed 3 to 6 times by reacting the secondary antibody with the antigen-antibody conjugate after the antigen-antibody binding reaction, and then adding an appropriate amount to the stationary body.
  • a sulfuric acid solution H 2 SO 4
  • H 2 SO 4 sulfuric acid solution
  • it relates to a method for providing information on suitability for cancer treatment.
  • the method of the present invention includes measuring the expression level of a PUM1-TRAF3 (pumilio RNA binding family member 1-TNF receptor associated factor 3) fusion protein or a gene encoding the same in a biological sample isolated from a subject of interest. .
  • PUM1-TRAF3 pumpmilio RNA binding family member 1-TNF receptor associated factor 3
  • the method of the present invention may be for screening whether a biological sample isolated from a subject of interest is suitable for a cancer treatment.
  • the "object of interest” refers to mammals including humans, such as humans, rats, mice, guinea pigs, hamsters, rabbits, monkeys, dogs, cats, cows, horses, pigs, sheep and goats. It may be selected from the group consisting of, and may preferably be a human, but is not limited thereto.
  • the "human” may refer to a person who has developed cancer or is suspected of having a high possibility of developing cancer, and may mean a patient who needs or is expected to receive appropriate treatment for cancer disease, preferably requires appropriate treatment for biliary tract cancer or It may be an expected patient, but is not limited thereto.
  • the "biological sample” of the present invention refers to any material, biological fluid, tissue, or cell obtained from or derived from a patient with a cancer disease or a subject suspected of having a cancer disease, for example, whole blood Whole blood, leukocytes, peripheral blood mononuclear cells, buffy coat, blood including plasma and serum, sputum, tears ), mucus, nasal washes, nasal aspirate, breath, urine, semen, saliva, peritoneal washings, pelvis Pelvic fluids, cystic fluid, meningeal fluid, amniotic fluid, glandular fluid, pancreatic fluid, lymph fluid, pleural fluid ), nipple aspirate, bronchial aspirate, synovial fluid, joint aspirate, organ secretions, cell, cell extract Or cerebrospinal fluid (cerebrospinal fluid) may be included, but is not limited thereto.
  • the step of measuring the level of the fusion protein of the present invention includes protein chip analysis, immunoassay, ligand binding assay, MALDI-TOF (Matrix Assisted Laser Desorption/Ionization Time of Flight Mass Spectrometry) using the composition of the present invention.
  • the step of measuring the expression level of the gene encoding the fusion protein of the present invention is reverse transcription polymerase reaction (RT-PCR), competitive reverse transcription polymerase reaction (Competitive RT-PCR), real-time reverse transcription using the composition of the present invention It may be performed by polymerase reaction (Real-time RT-PCR), RNase protection assay (RPA), Northern blotting, or DNA chip, but is not limited thereto.
  • the expression level of the PUM1-TRAF3 fusion protein or the gene encoding the same measured in the biological sample is higher than that of the normal control group, it can be predicted that the subject has suitability for cancer treatment. More preferably, when the expression level of the PUM1-TRAF3 fusion protein or the gene encoding it is higher than that of the normal control, nuclear factor kappa-light-chain-enhancer of activated B cells (NF- ⁇ B) inhibition It can be predicted that there is suitability for the treatment of bile duct cancer by drugs or drugs that inhibit NF- ⁇ B-inducing kinase (NIK).
  • NF- ⁇ B nuclear factor kappa-light-chain-enhancer of activated B cells
  • the cancer is biliary tract cancer, gastric cancer, thyroid cancer, parathyroid cancer, ovarian cancer, colon cancer, pancreatic cancer, liver cancer, breast cancer, cervical cancer, lung cancer, non-small cell lung cancer, prostate cancer, gallbladder cancer, biliary tract cancer, non-Hodgkin's lymphoma, Hodgkin's lymphoma, blood cancer, bladder cancer, kidney cancer, melanoma, colon cancer, bone cancer, skin cancer, head cancer, uterine cancer, rectal cancer, brain tumor, perianal cancer, fallopian tube carcinoma, endometrial carcinoma, vaginal cancer, vulvar carcinoma, esophageal cancer, small intestine cancer , endocrine adenocarcinoma, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, ureteral cancer, renal cell carcinoma, renal pelvic carcinoma, CNS central nervous system tumor, primary CNS lymphoma, spinal cord tumor, brainstem gliom
  • PUM1-TRAF3 fusion protein or a gene encoding it an agent for measuring expression level, cancer, cancer treatment, NF- ⁇ B inhibitor, NIK inhibitor, control, etc.
  • the description is the same as that described in the composition for discrimination, and is omitted to avoid excessive complexity of the present specification.
  • it relates to a diagnostic device for determining the suitability of a cancer treatment.
  • the measurement unit of the diagnostic device of the present invention can measure the expression level of a PUM1-TRAF3 (pumilio RNA binding family member 1-TNF receptor associated factor 3) fusion protein or a gene encoding it in a biological sample obtained from a subject of interest. .
  • PUM1-TRAF3 pumpmilio RNA binding family member 1-TNF receptor associated factor 3
  • the object of interest may be selected from the group consisting of humans, rats, mice, guinea pigs, hamsters, rabbits, monkeys, dogs, cats, cows, horses, pigs, sheep and goats, and is preferably a human. It may be, but is not limited thereto.
  • the biological sample is whole blood, leukocytes, peripheral blood mononuclear cells, leukocyte buffy coat, plasma, serum, sputum , tears, mucus, nasal washes, nasal aspirate, breath, urine, semen, saliva, peritoneal washings), ascites, cystic fluid, meningeal fluid, amniotic fluid, glandular fluid, pancreatic fluid, lymph fluid, pleural fluid ), nipple aspirate, bronchial aspirate, synovial fluid, joint aspirate, organ secretions, cell, cell extract And it may be at least one selected from the group consisting of cerebrospinal fluid, etc., but is not limited thereto.
  • the agent used in the measuring unit of the diagnostic device of the present invention may be an agent that measures the expression level of the PUM1-TRAF3 fusion protein or the gene encoding it. More specifically, it may include one or more selected from the group consisting of an antibody, an oligopeptide, a ligand, a peptide nucleic acid (PNA), and an aptamer that specifically binds to the fusion protein, and the fusion gene It may include at least one selected from the group consisting of a primer, a probe, and an antisense nucleotide that specifically binds.
  • PNA peptide nucleic acid
  • the suitability of a cancer therapeutic agent can be predicted by checking the expression level of the fusion protein or fusion gene using the agent.
  • the diagnostic device of the present invention adds a detection unit that predicts and outputs the suitability of the cancer treatment agent for the target individual, more preferably, the suitability for treatment of cholangiocarcinoma, from the expression level of the fusion protein or fusion gene obtained from the measurement unit. can be further included.
  • the detection unit generates and classifies information on the suitability of a cancer therapeutic agent according to the category of the expression level of the fusion protein or fusion gene obtained by the measurement unit, thereby discriminating suitability for treatment of cancer, more preferably, biliary tract cancer. can be diagnosed
  • the detection unit predicts that the target individual has high suitability for cancer treatment and outputs the result. can do. More preferably, when the expression level of the PUM1-TRAF3 fusion protein or the gene encoding it is higher than that of the normal control, nuclear factor kappa-light-chain-enhancer of activated B cells (NF- ⁇ B) inhibition It can be predicted that there is suitability for the treatment of bile duct cancer by drugs or drugs that inhibit NF- ⁇ B-inducing kinase (NIK).
  • NF- ⁇ B nuclear factor kappa-light-chain-enhancer of activated B cells
  • composition for preventing, improving or treating cancer relates to a composition for preventing, improving or treating cancer.
  • the composition of the present invention is effective as a nuclear factor kappa-light-chain-enhancer of activated B cells (NF- ⁇ B) inhibitor or an NF- ⁇ B-inducing kinase (NIK) inhibitor It may include as a component, the NF- ⁇ B inhibitor is QNZ or a pharmaceutically acceptable salt thereof; MG132 or a pharmaceutically acceptable salt thereof; At least one selected from the group consisting of pyrrolidinedithiocarbamate ammonium or a pharmaceutically acceptable salt thereof and caffeic acid phenethyl ester or a pharmaceutically acceptable salt thereof;
  • the NIK inhibitor may be B022 or a pharmaceutically acceptable salt thereof; XT2 or a pharmaceutically acceptable salt thereof; And NIK SMI1 or a pharmaceutically acceptable salt thereof; may include at least one selected from the group consisting of, but is not limited thereto.
  • the QNZ is 4-N-[2-(4-phenoxyphenyl)ethyl]-1,2-dihydroquinazoline-4,6-diamine (4-N-[2-( 4-Phenoxyphenyl)ethyl] -1,2-dihydroquinazoline-4,6-diamine) and may be a compound represented by Formula 1 below.
  • the MG132 is benzyl[(2S)-4-methyl-1- ⁇ [(2S)-4-methyl-1- ⁇ [(2S)-4-methyl-1-oxopentane-2 -yl]amino ⁇ -1-oxopentan-2-yl]amino ⁇ -1-oxopentan-2-yl]carbamate (Benzyl [(2S)-4-methyl-1- ⁇ [(2S)-4- methyl-1- ⁇ [(2S)-4-methyl-1-oxopentan-2-yl]amino ⁇ -1-oxopentan-2-yl]amino ⁇ -1-oxopentan-2-yl]carbamate) to Formula 2 It may be a compound represented by
  • the pyrrolidinedithiocarbamate ammonium is pyrrolidine-1-carbodithioate and may be a compound represented by Formula 3 below. .
  • the caffeic acid phenethyl ester is 2-phenylethyl (E) -3- (3,4-dihydroxyphenyl) prop-2-inoate (2-phenylethyl (E)- 3-(3,4-dihydroxyphenyl)prop-2-enoate) may be a compound represented by Formula 4 below.
  • the B022 is 4-[1-(2-amino-5-chloropyrimidin-4-yl)-2,3-dihydroindol-6-yl]-2-(1, 3-thiazol-2-yl)but-3-yn-2-ol (4-[1-(2-amino-5-chloropyrimidin-4-yl)-2,3-dihydroindol-6-yl]-2 -(1,3-thiazol-2-yl)but-3-yn-2-ol) may be a compound represented by Formula 5 below.
  • the XT2 is (R)-4-(3-(2-amino-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-4-fluorophenyl) -2-(Thiazol-2-yl)but-3-yn-2-ol ((R)-4-(3-(2-Amino-5H-pyrrolo[3,2-d]pyrimidin-7-yl )-4-fluorophenyl)-2-(thiazol-2-yl)but-3-yn-2-ol) and may be a compound represented by Formula 6 below.
  • the NIK SMI1 is 6-[3-[2-[(3R)-3-hydroxy-1-methyl-2-oxopyrrolidin-3-yl]ethynyl]phenyl] -4-methoxypyridine-2-carboxamide (6-[3-[2-[(3R)-3-hydroxy-1-methyl-2-oxopyrrolidin-3-yl]ethynyl]phenyl]-4-methoxypyridine -2-carboxamide) may be a compound represented by Formula 7 below.
  • the compounds represented by Chemical Formulas 1 to 7 or pharmaceutically acceptable salts thereof of the present invention can very effectively inhibit the growth, metastasis and invasion of cancer cells.
  • the composition of the present invention can be used very effectively for preventing, improving or treating cancer by inhibiting uncontrolled growth of cancer cells or reducing metastasis and invasiveness of cancer cells, and more preferably preventing or improving biliary tract cancer. Or it can be used very effectively for treatment.
  • the pharmaceutically acceptable salts of the present invention may include acid or base addition salts, and stereochemical isomers thereof.
  • the compounds may be in the form of addition salts of organic or inorganic acids. Salts, when administered to a patient, have desirable effects on the patient, and include, but are not particularly limited to, any salts that retain the activity of their parent compound.
  • salts include inorganic and organic salts such as acetic acid, nitric acid, aspartic acid, sulfonic acid, sulfuric acid, maleic acid, glutamic acid, formic acid, succinic acid, phosphoric acid, phthalic acid, tannic acid, tartaric acid, hydrobromic acid, propionic acid, benzenesulfonic acid, Benzoic acid, stearic acid, lactic acid, bicarboxylic acid, bisulfuric acid, bitartaric acid, oxalic acid, butyric acid, calcium idete, carbonic acid, chlorobenzoic acid, citric acid, idetic acid, toluenesulfonic acid, fumaric acid, gluseptic acid, ecilin Acid, pamoic acid, gluconic acid, methyl nitric acid, malonic acid, hydrochloric acid, hydroiodoic acid, hydroxynaphtholic acid, isethionic acid, lactobi
  • Addition salts of bases include salts of alkali metals or alkaline earth metals, such as salts of ammonium, lithium, sodium, potassium, magnesium, calcium and the like; salts with organic bases such as benzathine, N-methyl-D-glucamine, hydrabamine and the like; and salts with amino acids such as arginine, lysine, and the like. Also, these salts can be converted to the free form by treatment with an appropriate base or acid.
  • the disease to be prevented, improved or treated in the composition of the present invention may be a cancer that has occurred or is likely to develop in a subject of interest, preferably bile duct cancer, specifically a PUM1-TRAF3 fusion protein, It may be cholangiocarcinoma that has been or is likely to be developed in individuals whose genes encoding these are highly expressed.
  • the "object of interest” refers to mammals including humans, such as humans, rats, mice, guinea pigs, hamsters, rabbits, monkeys, dogs, cats, cows, horses, pigs, sheep and goats. It may be selected from the group consisting of, and may preferably be a human, but is not limited thereto.
  • the "human” may refer to a patient who has or is suspected of having cancer and requires or is expected to receive appropriate treatment for cancer, preferably bile duct cancer, but is not limited thereto.
  • the disease to be prevented or treated refers to biliary tract cancer, gastric cancer, thyroid cancer, parathyroid cancer, ovarian cancer, colon cancer, pancreatic cancer, liver cancer, breast cancer, cervical cancer, lung cancer, non-small cell lung cancer, Prostate cancer, gallbladder cancer, biliary tract cancer, non-Hodgkin's lymphoma, Hodgkin's lymphoma, blood cancer, bladder cancer, kidney cancer, melanoma, colon cancer, bone cancer, skin cancer, head cancer, uterine cancer, rectal cancer, brain tumor, perianal cancer, fallopian tube carcinoma, uterus Endometrial carcinoma, vaginal cancer, vulvar carcinoma, esophageal cancer, small intestine cancer, endocrine adenocarcinoma, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, ureteric cancer, renal cell carcinoma, renal pelvic carcinoma, CNS central nervous system tumor, primary It
  • the "prevention" of the present invention can be included without limitation as long as it is an act of blocking, suppressing or delaying symptoms caused by uncontrolled growth, metastasis, invasion, etc. of cancer cells using the composition of the present invention. there is.
  • the "improvement" of the present invention may include without limitation any action that improves or benefits symptoms caused by uncontrolled growth, metastasis, invasion, etc. of cancer cells by using the composition of the present invention.
  • treatment of the present invention can be included without limitation as long as symptoms caused by uncontrolled growth, metastasis, invasion, etc. of cancer cells are improved or beneficial by using the composition of the present invention.
  • composition for preventing, improving or treating cancer of the present invention may be used as a pharmaceutical composition or a food composition, but is not limited thereto.
  • the pharmaceutical composition may be in the form of capsules, tablets, granules, injections, ointments, powders or beverages, and the pharmaceutical composition may be intended for humans.
  • the pharmaceutical composition of the present invention is not limited to these, but is formulated according to conventional methods into oral formulations such as powders, granules, capsules, tablets, aqueous suspensions, external preparations, suppositories and sterile injection solutions.
  • the pharmaceutical composition of the present invention may include a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers may include binders, lubricants, disintegrants, excipients, solubilizers, dispersants, stabilizers, suspending agents, pigments, flavors, etc. for oral administration, and buffers, preservatives, and painless agents for injections.
  • the formulation of the pharmaceutical composition of the present invention may be variously prepared by mixing with the above-described pharmaceutically acceptable carrier.
  • a base, excipient, lubricant, preservative, etc. may be used.
  • the formulation of the pharmaceutical composition of the present invention may be variously prepared by mixing with the above-described pharmaceutically acceptable carrier.
  • for oral administration it can be prepared in the form of tablets, troches, capsules, elixirs, suspensions, syrups, wafers, etc., and in the case of injections, it can be prepared in unit dosage ampoules or multiple dosage forms. there is.
  • it may be formulated into solutions, suspensions, tablets, capsules, sustained-release preparations, and the like.
  • examples of carriers, excipients and diluents suitable for formulation include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, malditol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, Cellulose, methyl cellulose, microcrystalline cellulose, polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate or mineral oil and the like can be used.
  • fillers, anti-coagulants, lubricants, wetting agents, flavoring agents, emulsifiers, preservatives, and the like may be further included.
  • the route of administration of the pharmaceutical composition according to the present invention is, but is not limited to, oral, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, subcutaneous, intraperitoneal, intranasal, intestinal, topical , sublingual or rectal. Oral or parenteral administration is preferred.
  • parenteral includes subcutaneous, intradermal, intravenous, intramuscular, intraarticular, intracapsular, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • the pharmaceutical composition of the present invention may also be administered in the form of a suppository for rectal administration.
  • the pharmaceutical composition of the present invention depends on various factors including the activity of the specific compound used, age, body weight, general health, sex, diet, administration time, route of administration, excretion rate, drug combination and severity of the specific disease to be prevented or treated.
  • the dosage of the pharmaceutical composition may be variously varied, and the dosage of the pharmaceutical composition varies depending on the patient's condition, weight, disease severity, drug form, administration route and period, but may be appropriately selected by those skilled in the art, and is 0.0001 to 50 mg per day. /kg or 0.001 to 50 mg/kg. Administration may be administered once a day, or may be administered in several divided doses. The dosage is not intended to limit the scope of the present invention in any way.
  • the pharmaceutical composition according to the present invention may be formulated into a pill, dragee, capsule, liquid, gel, syrup, slurry, or suspension.
  • composition for preventing, improving or treating cancer in the present invention may be administered in combination with other anticancer agents.
  • other anticancer agents when used in combination with other anticancer agents, it is possible to exert a remarkable effect in preventing or treating cancer by more effectively suppressing cancer growth or metastasis.
  • the anticancer agent of the present invention is nitrogen mustard, imatinib, oxaliplatin, rituximab, erlotinib, neratinib, lapatinib, gefitinib, vandetanib, nirotinib, semasanib, bosutinib, axitinib, cedi ranib, lestaurtinib, trastuzumab, gefitinib, bortezomib, sunitinib, carboplatin, sorafenib, bevacizumab, cisplatin, cetuximab, viscum album, asparaginase, tretinoin, hydroxyl Carbamide, dasatinib, estramustine, gemtuzumab ozogamicin, ibritumomabtucetan, heptaplatin, methylaminolevulinic acid, amsacrine, ale
  • a nuclear factor kappa-light-chain-enhancer of activated B cells (NF- ⁇ B) inhibitor or NF- ⁇ B-induced kinase (NF- ⁇ B -inducing kinase relates to a method for preventing or treating cancer, comprising administering a pharmaceutically effective amount of a composition containing a NIK) inhibitor as an active ingredient.
  • the NF- ⁇ B inhibitor is QNZ or a pharmaceutically acceptable salt thereof; MG132 or a pharmaceutically acceptable salt thereof; At least one selected from the group consisting of pyrrolidinedithiocarbamate ammonium or a pharmaceutically acceptable salt thereof and caffeic acid phenethyl ester or a pharmaceutically acceptable salt thereof;
  • the NIK inhibitor may be B022 or a pharmaceutically acceptable salt thereof; XT2 or a pharmaceutically acceptable salt thereof; And NIK SMI1 or a pharmaceutically acceptable salt thereof; may include at least one selected from the group consisting of, but is not limited thereto.
  • the "administration" means providing a given compound of the present invention to a subject by any suitable method.
  • the "subject" in need of the administration may include both mammals and non-mammals.
  • the mammal include humans, non-human primates such as chimpanzees, other apes or monkey species; livestock animals such as cattle, horses, sheep, goats, pigs; domesticated animals such as rabbits, dogs or cats; Laboratory animals, such as rodents, such as rats, mice, or guinea pigs may be included, but are not limited thereto.
  • examples of the non-mammals in the present invention may include birds or fish, but are not limited thereto.
  • the formulation of the compound administered as described above is not particularly limited, and may be administered as a solid formulation, a liquid formulation, or an aerosol formulation for inhalation, and a liquid formulation for oral or parenteral administration immediately before use. It can be administered in solid form preparations intended to be converted into, for example, oral formulations such as powders, granules, capsules, tablets, aqueous suspensions, external preparations, suppositories and sterile injection solutions. However, it is not limited thereto.
  • a pharmaceutically acceptable carrier may be additionally administered together with the compound of the present invention during the administration.
  • the pharmaceutically acceptable carrier may be a binder, a lubricant, a disintegrant, an excipient, a solubilizer, a dispersant, a stabilizer, a suspending agent, a colorant, a flavoring agent, etc. for oral administration, and in the case of an injection, a buffer, Preservatives, analgesics, solubilizers, tonicity agents, stabilizers, etc. may be mixed and used, and in the case of topical administration, bases, excipients, lubricants, preservatives, etc. may be used.
  • Formulations of the compound of the present invention can be variously prepared by mixing with the pharmaceutically acceptable carriers described above.
  • it can be prepared in the form of tablets, troches, capsules, elixirs, suspensions, syrups, wafers, etc., and in the case of injections, it can be prepared in unit dosage ampoules or multiple dosage forms. there is.
  • it may be formulated into solutions, suspensions, tablets, capsules, sustained-release preparations, and the like.
  • examples of carriers, excipients and diluents suitable for formulation include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, malditol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, Cellulose, methyl cellulose, microcrystalline cellulose, polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate or mineral oil and the like can be used.
  • fillers, anti-coagulants, lubricants, wetting agents, flavoring agents, emulsifiers, preservatives, and the like may be further included.
  • Routes of administration of the compounds according to the present invention are, but are not limited to, oral, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual or work included. Oral or parenteral administration is preferred.
  • parenteral includes subcutaneous, intradermal, intravenous, intramuscular, intraarticular, intracapsular, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • the pharmaceutical composition of the present invention may also be administered in the form of a suppository for rectal administration.
  • “pharmaceutically effective amount” refers to a sufficient amount of an agent to provide a desired biological result. The result may be reduction and/or alleviation of the signs, symptoms or causes of a disease, or any other desirable change in a biological system.
  • an “effective amount” for therapeutic use is the amount of a compound disclosed herein required to provide a clinically significant reduction in disease.
  • An “effective” amount suitable in any individual case can be determined by one skilled in the art using routine experimentation.
  • the expression “effective amount” generally refers to an amount of an active substance that has a therapeutic effect.
  • the active substance is a growth inhibitor of cancer cells or cancer tumor cells, an NF- ⁇ B inhibitor, or a NIK inhibitor, and is a preventive, ameliorative, or therapeutic agent for cancer.
  • the compounds of the present invention vary depending on a number of factors, including the activity of the specific compound used, age, body weight, general health, sex, dosage, time of administration, route of administration, excretion rate, drug formulation, and severity of the specific disease to be prevented or treated.
  • the dosage of the compound may vary, depending on the patient's condition, body weight, disease severity, drug form, administration route and period, but may be appropriately selected by those skilled in the art, and is 0.0001 to 100 mg/kg or 0.001 to 0.001 mg/kg per day. It can be administered at 100 mg/kg. Administration may be administered once a day, or may be administered in several divided doses. The dosage is not intended to limit the scope of the present invention in any way.
  • the compound according to the present invention may be formulated as a pill, dragee, capsule, liquid, gel, syrup, slurry, or suspension.
  • the compounds of the present invention can be used alone or in combination with methods using surgery, radiation therapy, hormone therapy, chemotherapy and biological response modifiers.
  • the compound of the present invention may be further used in combination with other anticancer agents.
  • it relates to a method of accompanying diagnosis for a cancer treatment application.
  • the method may include measuring the expression level of a PUM1-TRAF3 (pumilio RNA binding family member 1-TNF receptor associated factor 3) fusion protein or a gene encoding the same in a biological sample isolated from a subject of interest. there is.
  • a PUM1-TRAF3 pumpmilio RNA binding family member 1-TNF receptor associated factor 3
  • the subject may be used interchangeably with a subject of interest, and the subject of interest may be a mammal including a human, for example, a human, rat, mouse, guinea pig, hamster, rabbit, monkey, dog, cat , may be selected from the group consisting of cows, horses, pigs, sheep and goats, preferably human, but is not limited thereto.
  • a human, rat, mouse, guinea pig, hamster, rabbit, monkey, dog, cat may be selected from the group consisting of cows, horses, pigs, sheep and goats, preferably human, but is not limited thereto.
  • the "human” may refer to a person who has developed cancer or is suspected of having a high possibility of developing cancer, and may mean a patient who needs or is expected to receive appropriate treatment for cancer disease, preferably requires appropriate treatment for biliary tract cancer or It may be an expected patient, but is not limited thereto.
  • the "biological sample” of the present invention refers to any material, biological fluid, tissue, or cell obtained from or derived from a patient with a cancer disease or a subject suspected of having a cancer disease, for example, whole blood Whole blood, leukocytes, peripheral blood mononuclear cells, buffy coat, blood including plasma and serum, sputum, tears ), mucus, nasal washes, nasal aspirate, breath, urine, semen, saliva, peritoneal washings, pelvis Pelvic fluids, cystic fluid, meningeal fluid, amniotic fluid, glandular fluid, pancreatic fluid, lymph fluid, pleural fluid ), nipple aspirate, bronchial aspirate, synovial fluid, joint aspirate, organ secretions, cell, cell extract Or cerebrospinal fluid (cerebrospinal fluid) may be included, but is not limited thereto.
  • the cancer therapeutic agent of the present invention is a nuclear factor kappa-light-chain-enhancer of activated B cells (NF- ⁇ B) inhibitor or an NF- ⁇ B-inducing kinase (NIK) inhibitor. It may include, but is not limited to, any drug that is expected to have a preventive or therapeutic effect on cancer-related diseases.
  • NF- ⁇ B nuclear factor kappa-light-chain-enhancer of activated B cells
  • NIK NF- ⁇ B-inducing kinase
  • the NF- ⁇ B inhibitor is QNZ or a pharmaceutically acceptable salt thereof; MG132 or a pharmaceutically acceptable salt thereof; At least one selected from the group consisting of pyrrolidinedithiocarbamate ammonium or a pharmaceutically acceptable salt thereof and caffeic acid phenethyl ester or a pharmaceutically acceptable salt thereof; It can be done, but is not limited thereto.
  • the NIK inhibitor is B022 or a pharmaceutically acceptable salt thereof; XT2 or a pharmaceutically acceptable salt thereof; And NIK SMI1 or a pharmaceutically acceptable salt thereof; may include at least one selected from the group consisting of, but is not limited thereto.
  • Companion Diagnostics using the biomarker of PUM1-TRAF3 can pre-select a subject predicted to have high drug reactivity when the above-mentioned cancer treatment is applied, so that the prevention or treatment efficiency of cancer-related diseases will be good. and measuring the expression level of the subject's PUM1-TRAF3 fusion protein or the gene encoding it can provide essential information for the effective use of the therapeutic agent.
  • the accompanying diagnosis method of the present invention may further include selecting an individual whose expression level of the PUM1-TRAF3 fusion protein measured above or a gene encoding the same is higher than that of a normal control group.
  • an individual whose expression level of the PUM1-TRAF3 fusion protein or the gene encoding it is higher than that of the normal control group activates the non-canonical NF- ⁇ B signaling induced by the expression of the PUM1-TRAF3 fusion gene (non-canonical NF- ⁇ B signaling ) Since the efficacy of therapeutic agents having an inhibitory effect is predicted to be high, cancer, preferably cholangiocarcinoma, most preferably by overexpression of the PUM1-TRAF3 fusion gene through administration of the composition for preventing or treating cancer of the present invention The resulting bile duct cancer can be more effectively prevented or treated.
  • the description of the PUM1-TRAF3 fusion protein or the gene encoding it, the cancer, the type of cancer drug, accompanying diagnosis, etc. is the same as that described in the composition for determining the suitability of a cancer drug, thereby avoiding the excessive complexity of the present specification. omitted to avoid
  • the companion diagnosis method of the present invention has the meaning of precisely presenting a patient's treatment method according to the patient's characteristics, and thus increases the efficiency of cancer treatment or reduces the side effects of the treatment by selecting the patient's drug reactivity before prescribing a cancer treatment. It can reduce, and furthermore, the economic effect of preventing unnecessary medical expenses is expected.
  • composition according to the present invention maximizes the therapeutic effect of cholangiocarcinoma by applying an NF- ⁇ B inhibitor or a NIK inhibitor to selected patients with high expression of the PUM1-TRAF3 fusion gene using a companion diagnostic biomarker called the PUM1-TRAF3 fusion gene.
  • FIG. 1 is a diagram showing a flow chart for fusion gene analysis from a biliary tract cancer (BTC) patient according to an embodiment of the present invention.
  • FIG. 2A is a diagram showing a PUM1-TRAF3 fusion gene including PUM1 (PUF domain, chromosome 1) and TRAF3 (MATH domain, chromosome 14) according to an embodiment of the present invention.
  • Figure 2b shows fusion signals (indicated by arrows) through fluorescence in situ hybridization (FISH) analysis using two probes for PUM1 (red) and TRAF3 (green) in P1 tissue slides according to an embodiment of the present invention. It is also
  • Figure 2c is a P1 patient tissue slide (left) in which the PUM1-TRAF3 fusion gene was detected using the PUM1 antibody and the TRAF3 antibody according to an embodiment of the present invention and a patient tissue slide (right) in which the PUM1-TRAF3 fusion gene was not detected. It is a diagram showing the results (DAPI: blue, PT (PUM1-TRAF3 fusion gene): red) of performing in situ proximal ligation assay (PLA) to visualize PUM1-TRAF3 protein expression in .
  • DAPI blue
  • PT PUM1-TRAF3 fusion gene
  • Figure 2d is a diagram showing the results of PCR analysis (NL: normal, CA: cancer) of P1 to P26 tissue samples according to an embodiment of the present invention.
  • Figure 3a shows the results of confirming the cell proliferation effect upon expression of PUM1-TRAF3 in SNU1196 cells or SNU308 cells according to an embodiment of the present invention (C: control vector transduced cells, PT: PUM1-TRAF3 fusion gene transduced cells) is the diagram shown.
  • Figure 3b is a result of confirming cell metastasis and cell invasiveness upon expression of PUM1-TRAF3 in SNU1196 cells or SNU308 cells according to an embodiment of the present invention (C: control vector transduced cells, PT: PUM1-TRAF3 fusion gene transduced cells) ) is shown.
  • Figures 4a and 4b show the results of measuring the tumor size at 7-day intervals from mice of the PUM1-TRAF3 transduced group and the control vector transduced group according to an embodiment of the present invention
  • C control vector transduced cells
  • PT It is a diagram showing PUM1-TRAF3 fusion gene-transduced cells).
  • Figure 5a shows the results of analyzing proteins differentially expressed in PUM1-TRAF3 transduced cells according to an embodiment of the present invention (C: control vector transduced cells, PT: PUM1-TRAF3 fusion gene transduced cells). It is also
  • Figure 5b shows the immunohistochemical staining results of NF- ⁇ B2, RelB and green fluorescent protein (GFP) from mouse tumor tissue according to an embodiment of the present invention
  • C control vector transduced cells
  • PT PUM1-TRAF3 fusion gene trait It is a diagram showing introduced cells).
  • 5C is a diagram showing immunohistochemical staining results of NF- ⁇ B and RelB in the nucleus from BTC patient tissues (P1 to P11) expressing the PUM1-TRAF3 fusion protein according to an embodiment of the present invention.
  • 6a to 6c are diagrams confirming target sites for gRNA design for generation of the TRAF3 knockout cell line of the present invention and protein expression of cells derived from cells transfected with CRISPR through Western blotting.
  • FIGS. 7a to 7c are diagrams confirming target sites for gRNA design for generation of NF- ⁇ B2 knockout cell lines according to an embodiment of the present invention and protein expression of cells derived from cells transfected with CRISPR through Western blotting; am.
  • Figure 8a is a control vector transduced SNU1196 cells and PUM1-TRAF3 fusion gene transduced cells or a TRAF3 knockout control vector transduced SNU1196 cells and PUM1-TRAF3 transduced SNU1196 cells according to an embodiment of the present invention in NF- ⁇ B2 and RelB It is a diagram showing the result of immunofluorescence staining for.
  • 8B is a diagram showing the results of immunofluorescence staining for NF- ⁇ B2 and RelB when NF- ⁇ B2 was knocked out in control vector-transduced SNU1196 cells or PUM1-TRAF3-transduced SNU1196 cells according to an embodiment of the present invention.
  • Figure 9a is a diagram showing the results of confirming cell proliferation changes after treatment with NF- ⁇ B inhibitor QNZ to SNU1196 and SNU308 cells transduced with PUM1-TRAF3 or a control vector according to an embodiment of the present invention.
  • Figure 9b shows changes in NF- ⁇ B2 and RelB compared to administration of DMSO, a control drug, after treatment with QNZ, an NF- ⁇ B inhibitor, in SNU1196 and SNU308 cells transduced with PUM1-TRAF3 or a control vector according to an embodiment of the present invention. It is a diagram showing the result of immunofluorescence staining for.
  • 10a and 10b show the expression levels of p52 and RelB in the nucleus or cytoplasm of SNU1196 cells transduced with PUM1-TRAF3 or a control vector according to an embodiment of the present invention, treated with NIK inhibitor B022, and then subjected to western blotting. It is a diagram showing the result of confirming the change in .
  • FIG. 11 is a diagram showing the results of immunofluorescence staining for NIK, NF- ⁇ B2 and RelB after treating SNU1196 cells transduced with PUM1-TRAF3 or a control vector according to an embodiment of the present invention with the NIK inhibitor B022 .
  • 12a and 12b are images obtained by immunohistochemistry analysis by dividing NIK-weak and NIK-strong groups in cholangiocarcinoma patients according to an embodiment of the present invention, and NIK according to PUM1-TRAF3 positive/negative groups. It is a diagram showing the result of confirming the relative frequency of NIK expression in the -weak and NIK-strong groups.
  • This diagram confirms the Kaplan-Meier analysis results for overall survival (OS).
  • 14a and 14b are diagrams illustrating the results of confirming DFS and OS through multivariate analysis using a Cox proportional hazards model according to an embodiment of the present invention.
  • composition for determining the suitability of a cancer treatment comprising an agent for measuring the expression level of a PUM1-TRAF3 (pumilio RNA binding family member 1-TNF receptor associated factor 3) fusion protein or a fusion gene encoding the same it's about
  • PUM1-TRAF3 pumpmilio RNA binding family member 1-TNF receptor associated factor 3
  • Another embodiment of the present invention relates to a kit for determining the suitability of a cancer therapeutic agent including the composition for determining the suitability of the cancer therapeutic agent.
  • Another embodiment of the present invention includes measuring the expression level of a PUM1-TRAF3 (pumilio RNA binding family member 1-TNF receptor associated factor 3) fusion protein or a gene encoding the same in a biological sample isolated from a subject of interest. It relates to a method for providing information on the suitability of a cancer treatment for cancer treatment.
  • PUM1-TRAF3 pumpmilio RNA binding family member 1-TNF receptor associated factor 3
  • PUM1-TRAF3 pumpmilio RNA binding family member 1-TNF receptor associated factor 3
  • nuclear factor kappa-light-chain-enhancer of activated B cells (NF- ⁇ B) inhibitor or NF- ⁇ B-inducing kinase (NIK) inhibitor It relates to a pharmaceutical composition for preventing or treating cancer comprising as an active ingredient.
  • a nuclear factor kappa-light-chain-enhancer of activated B cells (NF- ⁇ B) inhibitor or an NF- ⁇ B-inducing kinase is administered to a subject in need of administration.
  • kinase; NIK) inhibitor as an active ingredient relates to a method for preventing or treating cancer comprising administering a composition for preventing or treating cancer in a pharmaceutically effective amount.
  • the step of measuring the expression level of a PUM1-TRAF3 (pumilio RNA binding family member 1-TNF receptor associated factor 3) fusion protein or a fusion gene encoding the same in a biological sample isolated from a subject of interest It relates to a method for concurrently diagnosing a subject for application of a cancer treatment comprising the present invention.
  • the present inventors obtained written consent among patients who underwent surgery for the purpose of treatment for biliary tract cancer (BTC) at Severance Hospital, Yonsei University College of Medicine. Patient samples and clinical information were obtained from the recipient patients. The study protocol complied with the ethical standards of the institutional research committee and the 1964 Declaration of Helsinki and its subsequent amendments or similar ethical standards, and was approved by the ethics committee and institutional review committee of Yonsei University College of Medicine (IRB approval code: 4-2011-0625). ) was received and the experiment was performed. Tumors were staged according to the American Joint Committee on Cancer (AJCC) stage classification, and information on collected patient samples is shown in Table 1 below.
  • AJCC American Joint Committee on Cancer
  • Human BTC cell lines SNU245, SNU308, SNU478, SNU869, SNU1079 and SNU1196 were obtained from the Korean Cell Line Bank (KCLB). Each of the above cell lines was cultured in RPMI1640 (Invitrogen) medium containing 10% fetal bovine serum (FBS; Hyclone, Logan), 2mM L-glutamine, 100 U/ml penicillin, and 100 ⁇ g/ml streptomycin according to the guidelines of the Korean Cell Line Bank. cultured.
  • FBS fetal bovine serum
  • FBS fetal bovine serum
  • 2mM L-glutamine 100 U/ml penicillin
  • streptomycin 100 ⁇ g/ml streptomycin
  • Hucct-1, OZ and KKU 100 cell lines were obtained from the Japan Collection of Research Bioresources Cell Bank (JCRB) and cultured in Dulbecco's Modified Eagle Medium (DMEM; Invitrogen) supplemented with 10% FBS (Hyclone). did 293FT cells were purchased from Invitrogen and cultured in DMEM (Invitrogen) medium supplemented with 10% FBS (Hyclone). All cell lines were cultured in an incubator at 37 °C and 5% CO 2 , and were used for experiments after contamination testing.
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS FBS
  • All cell lines were cultured in an incubator at 37 °C and 5% CO 2 , and were used for experiments after contamination testing.
  • RNA-seq of 5 tissue samples from 201 Korean BTC patients was performed, including 5 tumors and 5 matched normal bile duct tissues for initial detection (see Table 2).
  • Fusion genes were discovered through step-by-step filtering through a custom bioinformatics pipeline using ChimeraScan (version 0.4.5), FusionCatcher (version 0.99.4d), and JAFFA (version 1.06) (see Figure 1).
  • the two fusion genes finally selected after filtering were identified with high confidence, and the PUM1-TRAF3 and ASH1L 204 DOCK7 fusion genes were identified in patients with intrahepatic cholangiocarcinoma (P1 or B01). Since ASH1L-DOCK7 was out of frame, it was excluded from further verification, and then additional verification experiments of PUM1-TRAF3 were performed.
  • PUM-TRAF3 an in-frame transcript, consists of 2,988 bp of PUM1 (located at chr1:p35.2 of 3558 bp) and 744 bp of TRAF3 (located at chr14:q32.32 of 1704 bp), as shown in FIG.
  • TRAF3 transcripts conserve the MATH domain of TRAF3 required for physical binding to NF- ⁇ B-inducing kinase (NIK), and the other two domains of TRAF3 required for ubiquitination and degradation of NIK (e.g. For example, it was confirmed that RING finger and zinc finger domains) were lost.
  • NIK NF- ⁇ B-inducing kinase
  • FISH analysis using PUM1 and TRAF3 probes at the gene or protein level was performed to confirm genomic translocation (see Fig. 2b), and in situ PLA was performed to confirm the presence of the endogenous PUM1-TRAF3 fusion protein in patient tissue to dissociate the protein complex. Detected and visualized, it is shown in Figure 2c.
  • the fusion protein could be visually observed in a speckle-like pattern of PLA signals on patient tissue slides under a fluorescence microscope. Taken together, it was confirmed that the PUM1-TRAF3 fusion event occurred by genomic translocation and was actively expressed to produce a fusion protein in BTC.
  • PCR analysis was performed on 26 pairs of frozen tissues (normal and cancer tissues) to be applied to additional BTC cases and validated in an expanded cohort, and the results are shown in Figure 2d. Reanalysis of 26 sample pairs by ddPCR confirmed the presence of the fusion gene in tissue and plasma samples of 7 patients expressing the fusion transcript, and it was confirmed that the PUM1-TRAF3 fusion transcript was not detected in the healthy control group.
  • BTC cholangiocarcinoma
  • PUM1-TRAF3 was stably transduced into BTC cell lines to generate a PUM1-TRAF3 expressing cell line (PT cell line).
  • PT cell line a PUM1-TRAF3 expressing cell line
  • the Lentiviral vector and Packaging vector were co-transfected into 293FT cells using Lipofectamine 2000 (Invitrogen), and the supernatant was collected and added to the BTC cell line to transduce the PT protein.
  • Green fluorescent protein cells were selected using flow cytometry and used in the experiments.
  • cells were dissociated and resuspended at 1.5 x 10 5 cells/mL in 1% FBS and seeded in 24-well transwell plates (Costar, Bethesda, MD) at a density of 3 x 10 4 cells/well.
  • the upper chamber was pre-coated with Matrigel and cells were seeded at 3 x 10 4 cells/well.
  • the bottom chamber was filled with culture medium containing 20% FBS, and after incubation at 37° C., the cells were fixed with 5% glutaraldehyde for 30 minutes and stained with 0.1% crystal violet.
  • mice were injected with SNU1196 cells expressing PT and control vectors or TRAF3 knockout SNU1196 cells (5 x 10 6 cells/site). All animal studies were performed using protocols approved by the Institutional Animal Care and Use Committee.
  • N 5/group
  • mice injected with PT cells injected subcutaneously with tumor cells of the PT cell line 2528.38 ⁇ 371.97 mm 3
  • mice injected with PT cells injected subcutaneously with tumor cells of the PT cell line 2528.38 ⁇ 371.97 mm 3
  • mice injected with PT cells injected subcutaneously with tumor cells of the PT cell line 2528.38 ⁇ 371.97 mm 3
  • mice injected with PT cells injected subcutaneously with tumor cells of the PT cell line 2528.38 ⁇ 371.97 mm 3
  • mice injected with PT cells injected subcutaneously with tumor cells of the PT cell line 2528.38 ⁇ 371.97 mm 3
  • FIGS. 4a and 4b see FIGS. 4a and
  • the noncanonical nuclear factor kappa-light chain enhancer of the activated B cell (NF- ⁇ B) pathway including NF- ⁇ B-induced kinase (NIK), phospho-IKK ⁇ (activated IKK ⁇ ), p52 and RelB (the non -canonical nuclear factor kappa-light-chain-enhancer) gene expression was significantly increased in PT cells (see FIG. 5a). Accordingly, the expression levels of p52 and RelB in tumors were analyzed using immunohistochemical analysis, and compared to control vector-transduced cells (1196C), p52 and RelB were analyzed in the nucleus of mouse tumor tissue (1196PT) expressing the PUM1-TRAF3 fusion protein.
  • NF- ⁇ B activated B cell pathway
  • PUM1-TRAF3 induces activation of non-canonical NF- ⁇ B signaling through NF- ⁇ B2 accumulation.
  • the SNU1196 cell line was used to obtain TRAF3 and NF- ⁇ B2 knockout cells using Macrogen (Macrogen Korea, Seoul Korea) CRISPR/CAS technology, and specific target locations are shown in FIGS. 6a and 7a.
  • Cells transfected with CRISPR were selected with puromycin, and protein expression of colonies derived from single cells was further analyzed by Western blotting (see Figs. 6b, 6c, 7b and 7c).
  • the PUM1-TRAF3 fusion protein affects cell proliferation through a mechanism that interferes with the NF- ⁇ B signaling pathway or inhibits NF- ⁇ B2 gene expression. This suggests that the tumor formation process can be efficiently suppressed through inhibition of the NF- ⁇ B signaling pathway in BTC patients carrying a fusion gene by PUM1-TRAF3.
  • PUM1-TRAF3 transduced PT and CV cell lines and control vector-transduced cell lines were treated with diluted QNZ (CAS number; 545380-34-5) among NF- ⁇ B inhibitors within the concentration range of 0 to 500 nM to obtain PUM1 -
  • concentration range was applied by approximating the IC70 value for the cell line.
  • NF- ⁇ B2 and RelB showed reduced expression in the nuclei of 1196PT cells treated with QNZ at 30 nM (approximately IC20) compared to cells treated with DMSO.
  • RNA sequencing to identify BTC-specific molecular markers and RNA sequencing of tissue samples from BTC patients revealed a novel pumilio RNA binding family member 1 (PUM1) arising from a 1:14 chromosomal translocation.
  • PUM1 pumilio RNA binding family member 1
  • TNF receptor-related factor 3 TNF receptor-related factor 3
  • tissue samples from cholangiocarcinoma patients were analyzed to verify that the fusion gene was specific to cholangiocarcinoma.
  • proliferative ability and motility and degree of invasion in xenograft tumors were analyzed, and PUM1 for non-canonical NF- ⁇ B signaling affecting cancer cell survival -The effect of the TRAF3 fusion gene was confirmed.
  • BTC cells expressing the fusion gene proliferation, metastasis and tumorigenicity of BTC cells were increased by the PUM1-TRAF3 fusion gene, and the fusion gene activates non-canonical NF- ⁇ B signaling It was confirmed that BTC tumor formation was induced by induction.
  • the PUM1-TRAF3 fusion gene was highly expressed. It is expected that the treatment effect of bile duct cancer patients can be maximized.
  • the expression and activity of markers related to PUM1 and TRAF3-related signal transduction pathways were measured by western blotting, NF- ⁇ B-induced kinase (NIK) , expression of phospho-IKK ⁇ (activated IKK ⁇ ), p52 and RelB, the non-canonical nuclear factor kappa-light-chain-enhancer gene of the activated B cell (NF- ⁇ B) pathway Since it was confirmed that the NF- ⁇ B activity induced by the PUM1-TRAF3 fusion gene was significantly increased in these cholangiocarcinoma (PT) cells, the relationship between the NF- ⁇ B activity and NIK carcinogenicity was further confirmed.
  • NIK NF- ⁇ B-induced kinase
  • NF- ⁇ B activated B cell
  • B022 belonging to the NIK inhibitor number; 1202764-53-1) was used.
  • NIK expression is related to the expression of the PUM1-TRAF3 fusion marker in BTC patients and also to a worse prognosis.
  • the PUM1-TRAF3 fusion gene is used as an index that can preemptively determine the suitability of treatment. It is expected to maximize the treatment effect for patients.
  • the composition of the present invention when using the composition of the present invention, by using a companion diagnostic biomarker called the PUM1-TRAF3 fusion gene, the therapeutic effect of biliary tract cancer by applying an NF- ⁇ B inhibitor or a NIK inhibitor to selected patients with high expression of the PUM1-TRAF3 fusion gene can maximize
  • SEQ ID NO: 1 PUM1 site amino acid sequence of PUM1-TRAF3 fusion protein
  • SEQ ID NO: 2 amino acid sequence of TRAF3 region of PUM1-TRAF3 fusion protein
  • SEQ ID NO: 3 PUM1 region gene sequence of PUM1-TRAF3 fusion gene
  • SEQ ID NO: 4 TRAF3 region gene sequence of PUM1-TRAF3 fusion gene

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Oncology (AREA)
  • Wood Science & Technology (AREA)
  • Hospice & Palliative Care (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Emergency Medicine (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)

Abstract

Une composition selon la présente invention peut maximiser un effet de traitement des voies biliaires, par sélection de patients présentant une surexpression de la protéine hybride PUM1-TRAF3 ou d'un gène la codant pour sélectionner des sujets présentant des caractéristiques génétiques appropriées pour un médicament inhibiteur NF-κB, plus particulièrement un médicament contenant du QNZ ou un sel pharmaceutiquement acceptable de celui-ci, et administration du médicament aux sujets sélectionnés.
PCT/KR2022/019329 2021-12-01 2022-12-01 Composition pour la prévention ou le traitement du cancer des voies biliaires WO2023101457A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2021-0170087 2021-12-01
KR20210170087 2021-12-01

Publications (1)

Publication Number Publication Date
WO2023101457A1 true WO2023101457A1 (fr) 2023-06-08

Family

ID=86612735

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2022/019329 WO2023101457A1 (fr) 2021-12-01 2022-12-01 Composition pour la prévention ou le traitement du cancer des voies biliaires

Country Status (2)

Country Link
KR (1) KR20230084059A (fr)
WO (1) WO2023101457A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20170124683A (ko) * 2016-05-02 2017-11-13 연세대학교 산학협력단 담도암 진단용 신규 바이오마커로서의 융합 유전자 및 융합 단백질
KR20190054801A (ko) * 2017-11-14 2019-05-22 연세대학교 원주산학협력단 카페인산 페네틸 에스테르 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 대장성 질환 예방 또는 치료용 약학적 조성물
WO2020082016A1 (fr) * 2018-10-18 2020-04-23 The General Hospital Corporation Ciblage de la voie nfkb non canonique dans l'immunothérapie anticancéreuse

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20170124683A (ko) * 2016-05-02 2017-11-13 연세대학교 산학협력단 담도암 진단용 신규 바이오마커로서의 융합 유전자 및 융합 단백질
KR20190054801A (ko) * 2017-11-14 2019-05-22 연세대학교 원주산학협력단 카페인산 페네틸 에스테르 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 대장성 질환 예방 또는 치료용 약학적 조성물
WO2020082016A1 (fr) * 2018-10-18 2020-04-23 The General Hospital Corporation Ciblage de la voie nfkb non canonique dans l'immunothérapie anticancéreuse

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
EASON A B; SIN S -H; DITTMER D PSZABO E; WELLER MPHILLIPS D J; DROSTE M; SHAMSHIEV A: "Antitumor activity of DLX1008, a single chain antibody fragment binding to VEGF-A, in in vivo preclinical models of Kaposi sarcoma and glioblastoma", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 78, no. 13, Supplement 1, 1 July 2018 (2018-07-01), US , XP009544068, ISSN: 1538-7445, DOI: 10.1158/1538-7445.AM2018-4 *
KIM KAHEE: "Identification of bile duct cancer specific fusion genes in patient tissues", THESIS YONSEI UNIVERSITY, 1 December 2017 (2017-12-01), pages 1 - 33, XP093070668 *
RASMI RAJAN RADHA; SAKTHIVEL KUNNATHUR MURUGESAN; GURUVAYOORAPPAN CHANDRASEKARAN: "NF-κB inhibitors in treatment and prevention of lung cancer", BIOMEDICINE & PHARMACOTHERAPY, ELSEVIER, FR, vol. 130, 1 August 2020 (2020-08-01), FR , XP086321001, ISSN: 0753-3322, DOI: 10.1016/j.biopha.2020.110569 *

Also Published As

Publication number Publication date
KR20230084059A (ko) 2023-06-12

Similar Documents

Publication Publication Date Title
WO2012081898A2 (fr) Marqueur destiné à établir un pronostic du cancer de l'estomac et procédé d'établissement d'un pronostic du cancer de l'estomac
WO2010047448A1 (fr) Kit diagnostique du cancer du côlon utilisant un marqueur associé au cancer du côlon et procédé diagnostique de celui-ci
WO2009113814A2 (fr) Marqueur protéique utilisé pour le diagnostic précoce du cancer du foie
US10175244B2 (en) Dominant negative HSP110 mutant and its use in prognosing and treating cancers
WO2016036172A1 (fr) Biomarqueur pour prédire la sensibilité à un inhibiteur de protéine kinase et utilisation de celui-ci
WO2021101339A1 (fr) Composition pour prédire une réponse à une chimioradiothérapie préopératoire standard et un pronostic après traitement, et procédé et composition pour prédire des patients susceptibles de présenter des pronostics très insatisfaisants après une thérapie standard
WO2016129890A1 (fr) Biomarqueur pour diagnostiquer des maladies vasculaires et utilisation de celui-ci
WO2021107644A1 (fr) UTILISATION D'UNE COMPOSITION POUR AMÉLIORER L'EFFET ANTICANCÉREUX, COMPRENANT UN INHIBITEUR DE ERRγ COMME PRINCIPE ACTIF
WO2017082655A1 (fr) Procédés pour déterminer la résistance à une thérapie anticancéreuse et composition utilisée à cet effet
WO2021177691A1 (fr) Composition pour le diagnostic ou le traitement de la résistance aux médicaments anticancéreux
WO2021182843A1 (fr) Composition pour le diagnostic ou le traitement de la résistance aux médicaments anticancéreux
WO2011081421A9 (fr) C9 du complément comme marqueur pour le diagnostic d'un cancer
WO2021201526A1 (fr) Biomarqueur pour l'immunothérapie contre le cancer et son utilisation
WO2014014157A1 (fr) Utilisation de l'adcy3 pour diagnostiquer et traiter le cancer de l'estomac
WO2015108328A1 (fr) Nouveau gène de fusion ntrk1 comme marqueur du cancer colorectal et application associée
WO2023101457A1 (fr) Composition pour la prévention ou le traitement du cancer des voies biliaires
EP2361317A1 (fr) Utilisation de eif3m pour le diagnostic et le traitement du cancer
WO2017164568A1 (fr) Procédé de prédiction du taux de survie et du pronostic de patient présentant un cancer de l'œsophage en mesurant le niveau d'expression de protéine de la sous-unité v-atpase v1e1
WO2019146841A1 (fr) Biomarqueur de diagnostic et de pronostic du cancer du foie et son utilisation
WO2022203314A2 (fr) Composition pour le diagnostic différentiel d'une tumeur maligne des gaines nerveuses périphériques
WO2023042944A1 (fr) Composition pour prévenir, améliorer ou traiter le cancer gastrique
WO2022216066A1 (fr) Biomarqueur pour l'estimation du pronostic chez des patients atteints de cancer et son utilisation
WO2021025412A1 (fr) Méthode de diagnostic de la maladie d'alzheimer au moyen du composant c8 gamma du complément
WO2009131366A2 (fr) Cdca5 en tant que marqueur diagnostique et agent thérapeutique pour le cancer gastrique ou le cancer colorectal
WO2021167177A1 (fr) Composition de biomarqueur de diagnostic compagnon et kit de diagnostic compagnon la contenant

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: 22901815

Country of ref document: EP

Kind code of ref document: A1