WO2016152352A1 - Biomarqueur spécifique du mélanome et son utilisation - Google Patents

Biomarqueur spécifique du mélanome et son utilisation Download PDF

Info

Publication number
WO2016152352A1
WO2016152352A1 PCT/JP2016/054968 JP2016054968W WO2016152352A1 WO 2016152352 A1 WO2016152352 A1 WO 2016152352A1 JP 2016054968 W JP2016054968 W JP 2016054968W WO 2016152352 A1 WO2016152352 A1 WO 2016152352A1
Authority
WO
WIPO (PCT)
Prior art keywords
melanoma
gene
protein
expression
brp44l
Prior art date
Application number
PCT/JP2016/054968
Other languages
English (en)
Japanese (ja)
Inventor
昌志 加藤
伊知朗 矢嶋
湖州恵 武田
友二 後藤
Original Assignee
国立大学法人名古屋大学
学校法人中部大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立大学法人名古屋大学, 学校法人中部大学 filed Critical 国立大学法人名古屋大学
Priority to JP2017507616A priority Critical patent/JPWO2016152352A1/ja
Publication of WO2016152352A1 publication Critical patent/WO2016152352A1/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
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • 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
    • 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
    • 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

Definitions

  • the present invention relates to a melanoma-specific biomarker. Specifically, the present invention relates to a novel biomarker specific to melanoma and a test method using the same.
  • This application claims priority based on Japanese Patent Application No. 2015-058692 filed on Mar. 20, 2015, the entire contents of which are incorporated by reference.
  • Non-Patent Document 1 S100, melanomainhibiting activity (MIA), 5-S-cysteinyldopa (5-S-CD), 6-hydroxy-5-methoxyindole-2-carboxylic acid (6-H-5) are used as tumor markers for the diagnosis of melanoma. -MI-2CA) is used (Non-Patent Document 1).
  • dacarbazine has been used for a long time as a molecular targeted therapeutic drug for melanoma, but recently, new molecular targeted drugs targeting MAP kinase pathway related molecules (BRAF, MEK) have been developed and put into clinical practice. It has appeared (Non-Patent Document 2).
  • Melanoma has the highest rate of increase among all carcinomas (world statistics). Moreover, melanoma with advanced stage has a poor prognosis, and early detection is important. On the other hand, conventional biomarkers for melanoma are effective only for advanced stage melanoma and are less useful for early diagnosis. If a gene or protein that is highly related to malignant melanoma can be used as a new diagnostic marker, a more reliable diagnosis can be made earlier. Moreover, pathological diagnosis of melanoma is not easy, and if a biomarker highly specific to melanoma can be provided, it can contribute to improvement of pathological diagnosis accuracy.
  • This invention responds to the said request, and makes it a subject to contribute to development and improvement of the medical treatment (diagnosis method, treatment method) with respect to melanoma.
  • PDZRN3, DTX3L, CSS3, and BRP44L significantly increased expression levels in both mouse melanoma without Braf mutation (RET-mouse and B16 strain) and human melanoma with BRAF mutation (PDZRN3, DTX3L). , CSS3) or decrease (BRP44L), it can be expected to be an effective target in the treatment of melanoma that is resistant to BRAF inhibitors.
  • a melanoma-specific biomarker comprising a biomolecule selected from the group consisting of PDZRN3 gene, PDZRN3 protein, DTX3L gene, DTX3L protein, CSS3 gene, CSS3 protein, BRP44L gene and BRP44L protein.
  • the PDZRN3 gene PDZRN3 protein, DTX3L gene, DTX3L protein, CSS3 gene and CSS3 protein, according to the criteria that the detection value is high and the malignancy is high, or that the detection possibility is high
  • the melanocyte according to [3] wherein the determination of step (3) is performed according to the criterion that the malignancy is high if the detection value is low, or the criterion that the malignancy is high if it cannot be detected. Tumor examination method.
  • a melanocyte tumor test reagent comprising a substance exhibiting specific binding property to the melanoma-specific biomarker according to [1].
  • a melanocyte-based tumor test kit comprising the melanocyte-based tumor test reagent according to [8] or [9].
  • a melanoma therapeutic agent comprising a compound that suppresses the expression of a target gene selected from the group consisting of a PDZRN3 gene, a DTX3L gene, and a CSS3 gene as an active ingredient.
  • the compound is a compound selected from the group consisting of the following (a) to (e): (a) siRNA targeting the target gene; (b) a nucleic acid construct that generates siRNA targeting the target gene in the cell; (c) a single-stranded RNA having an expression suppressing sequence that suppresses expression of the target gene and a complementary sequence that anneals to the sequence; (d) an antisense nucleic acid that targets the transcript of the target gene; (e) Ribozymes that target transcripts of target genes.
  • a therapeutic agent for melanoma comprising the following (A) or (B) as an active ingredient and used in combination with an anticancer agent or a drug having an antitumor action: (A) BRP44L protein or a part thereof (however, it has an effective effect on the anticancer drug sensitivity of melanoma cells); (B) An expression vector carrying the BRP44L gene or a part thereof (however, it encodes a protein having an action effective for anticancer drug sensitivity of melanoma cells). [14] The melanoma therapeutic agent according to any one of [11] to [13], which is applied to melanoma exhibiting resistance to a BRAF inhibitor. [15] A melanoma treatment method comprising the step of administering the melanoma therapeutic agent according to any one of [11] to [14] to a patient with melanoma.
  • PDZRN3 gene expression level in melanoma model mouse tumors PDZRN3 gene expression levels in benign tumors and malignant melanomas that spontaneously developed in melanoma model mice (RET-Tg) were measured using real-time PCR. PDZRN3 gene expression level is significantly increased in malignant melanoma compared to benign tumors. PDZRN3 gene expression level in human and mouse melanoma cell lines. a) Expression of PDZRN3 protein in human normal skin melanocytes (NHEM) and human melanoma cell lines (6 types). In all melanoma cell lines, the expression level of PDZRN3 protein is increased compared to NHEM.
  • PDZRN3 protein expression level in human melanoma tissue (benign tumor: Benign, primary melanoma: Malignant, metastatic melanoma: Metastasis). After immunostaining with anti-PDZRN3 antibody, the expression level was measured by quantifying the staining level.
  • the expression levels were categorized into three (negative / low, medium and high), and the ratio of each expression level was displayed as a band graph (bottom). Expression levels in primary and metastatic melanoma are significantly higher compared to benign tumors. Expression level of DTX3L gene in melanoma model mouse tumor. Real-time PCR (A), Western blotting (B) and immunohistochemical staining (DTX3L gene (A) and protein (B, C) expression levels in benign tumors and malignant melanomas spontaneously developed in melanoma model mice (RET-Tg) ( Measured using C). The expression levels of DTX3L gene and protein are greatly increased in malignant melanoma compared to benign tumors.
  • Expression level of DTX3L protein in mouse and human melanoma cell lines A) Expression level of Dtx3l protein in mouse melanoma cell lines (4 types). The B16 cell line is known to have lower invasive ability (metastatic activity) than other cell lines.
  • the expression level was measured by quantifying the staining level.
  • the expression levels were categorized into three (negative / low, medium and high), and the ratio of each expression level was displayed as a band graph (bottom).
  • Expression levels in primary and metastatic melanoma are significantly higher compared to benign tumors.
  • CSS1 and CSS2 genes are both benign and malignant and their expression levels are low, but CSS3 gene expression is significantly increased in malignant melanoma.
  • Expression level of CSS3 protein in human melanoma tissue (benign tumor: Benign, primary melanoma: Malignant, metastatic melanoma: Metastatic melanoma). After immunostaining with anti-CSS3 antibody, the expression level was measured by quantifying the staining level. The expression levels were categorized into three (negative / low, medium and high), and the ratio of each expression level was displayed as a band graph (bottom). Expression levels in primary and metastatic melanoma are significantly higher compared to benign tumors. Expression levels of BRP44L gene and protein in melanoma model mouse tumors.
  • BRP44L gene (A) and protein (B) expression levels in benign tumors and malignant melanoma spontaneously developed in melanoma model mice (RET-Tg) were measured using real-time PCR (A) and Western blotting (B).
  • BRP44L mRNA and protein are down-regulated in melanoma compared to benign tumors.
  • Expression level of BRP44L gene in human melanoma tissue The expression level of BRP44L mRNA in human melanoma tissue (benign tumor: nevus, melanoma: melanoma) was measured using real-time PCR.
  • the expression level in human melanoma tissue is greatly reduced compared to benign tumors.
  • a human melanoma cell line (A375P) was exposed to the existing melanoma molecular target drug PLX4720 for a long period of time to create a resistant strain.
  • DTX3L was reduced by siRNA introduction (A)
  • the effect on invasive ability was measured by an invasion assay (B).
  • Suppressing DTX3L expression is also effective against PLX-resistant melanoma cells, and can solve the problems of existing molecular targeting drugs.
  • Change in invasion ability (metastasis ability) in vivo by CSS3 forced expression A stable strain in which CSS3 was forcibly expressed in a mouse melanoma cell line (B16F10) was established, and the invasion ability of each stable strain was measured by an invasion assay.
  • the first aspect of the present invention relates to a melanoma-specific biomarker.
  • the “melanoma-specific biomarker” refers to a biomolecule that serves as an indicator of melanoma.
  • the melanoma-specific biomarker is useful for determining whether or not it is a melanoma and for determining the malignancy of melanoma.
  • the biomarker of the present invention is applied to, for example, a patient suffering from or possibly suffering from melanoma, or a sample / analyte derived from the patient.
  • biomolecule refers to a molecule (compound) found in the living body.
  • a specific biomolecule is used as a biomarker.
  • a biomolecule in a sample / specimen separated from a living body is used. Become.
  • the biomarker of the present invention comprises a biomolecule that has been correlated with the malignancy of melanocyte tumors (in other words, an increase or decrease in melanoma-specific expression level), and is used for evaluating the malignancy of melanocyte tumors. It is a useful indicator.
  • the biomarker of the present invention consists of PDZRN3 gene, PDZRN3 protein, DTX3L gene, DTX3L protein, CSS3 gene, CSS3 protein, BRP44L gene or BRP44L protein.
  • genes and proteins are comprehensively expressed using gene symbols (for example, “PDZRN3” represents both the PDZRN3 gene and the PDZRN3 protein collectively).
  • PDZRN3 SEQ ID NO: 1 (gene sequence, NCBI nucleotide database Accession No .: NM_015009), SEQ ID NO: 2 (amino acid sequence, NCBI protein database Accession No .: NP_055824)
  • DTX3L SEQ ID NO: 3 (gene sequence, NCBI nucleotide database Accession No .: NM_138287), SEQ ID NO: 4 (amino acid sequence, NCBI protein database Accession No .: NP_612144)
  • CSS3 SEQ ID NO: 5 (gene sequence, NCBI nucleotide database Accession No .: NM_175856), SEQ ID NO: 6 (amino acid sequence, NCBI protein database Accession No .: NP_787052)
  • BRP44L SEQ ID NO: SEQ ID NO: 5 (gene sequence, NCBI nucleotide database Accession No .: NM_175856), SEQ ID NO: 6 (amino
  • the second aspect of the present invention relates to the use of the biomarker of the present invention, and provides a test method for melanocyte tumor (hereinafter also referred to as “test method of the present invention”).
  • the test method of the present invention is useful as a means for determining the malignancy of a melanocyte tumor.
  • Malignant melanoline tumors that is, melanoma accounts for only about 5% of skin cancer, but it is highly metastatic and accounts for 80% of skin cancer deaths. For this reason, melanoma is said to be one of the most malignant cancers among skin cancers.
  • the test method of the present invention provides useful information for diagnosing melanocyte tumors and contributes to the prevention and treatment of melanoma. According to the test method of the present invention, it is possible to easily and objectively determine the malignancy of a melanocyte tumor.
  • the level of the biomarker of the present invention in a specimen derived from a subject is used as an index.
  • level typically means “amount” or “concentration”.
  • level is also used to indicate whether or not a molecule to be detected can be detected (ie, the presence or absence of an apparent presence) in accordance with common practice and common technical knowledge.
  • biomarkers PDZRN3, DTX3L, CSS3, BRP44L
  • PDZRN3, DTX3L, CSS3, BRP44L four types of biomarkers
  • Different types of melanocyte tumors can be handled by using different biomarkers. That is, the present invention has an advantage that a wide range of cases can be dealt with.
  • BRP44L in combination with other biomarkers (one or more of PDZRN3, DTX3L, and CSS3) can be expected to have an effect of improving the accuracy of pathological diagnosis in a pathological specimen of a tumor.
  • biomarkers one or more of PDZRN3, DTX3L, and CSS3
  • the number and type of biomarkers to be employed may be determined in consideration of the required accuracy and the ease of inspection.
  • the inspection method of the present invention performs the following steps. (1) A step of preparing a specimen derived from a subject (2) A step of detecting one or more biomolecules in the specimen (that is, one or more biomarkers employed as an index) (3) A step of determining the malignancy of the melanocyte tumor based on the detection result
  • a specimen derived from the subject is prepared.
  • skin tissue, blood, plasma, serum, urine, saliva, sweat, tumor tissue or the like of the subject can be used.
  • the subject is not particularly limited. That is, the present invention can be widely applied to those who need to determine the malignancy of melanocyte tumors.
  • “A person who needs to determine the malignancy of a melanocyte tumor” is typically a person who has developed a melanocyte tumor, but a subject who is suspected of developing a melanocyte tumor (potential patient) It is good.
  • the malignancy of a melanocyte tumor can be determined based on an objective index such as the level of a biomarker (biomolecule).
  • the determination result is useful information for diagnosing melanocyte tumors, and is useful for determining a more appropriate treatment policy (selecting an effective treatment method, etc.). Therefore, the present invention contributes to the improvement of the therapeutic effect and the improvement of the patient's QOL (Quality of Life). In pathological diagnosis of melanocyte tumors, even a skilled laboratory technician or doctor may be at a loss.
  • the highly objective data provided by the present invention provides information that is auxiliary but highly useful, especially in such cases.
  • step (2) biomarkers in the specimen are detected. It is not essential to strictly quantify the level of the biomarker. That is, the level of the biomarker may be detected to the extent that the malignancy of the melanocyte tumor can be determined in the subsequent step (3). For example, detection can be performed so that it can be determined whether or not the level of the biomarker in the sample exceeds a predetermined reference value.
  • Biomarker detection method is not particularly limited.
  • the biomarker adopted is a specific gene, it can be detected by quantifying the mRNA of the gene by a method using a nucleic acid amplification reaction such as RT-PCR.
  • Nucleic acid amplification reactions include PCR (Polymerase reaction) method or its modified method, and LAMP (Loop-Mediated Isothermal Amplification) method (Tsugunori Notomi et al. Nucleic Acids Research, Vol.28, No.12, e63, 2000) ; Kentaro Nagamine, Keiko Watanabe et al.
  • the biomarker employed is a specific protein
  • the biomarker is preferably detected using an immunological technique.
  • immunological techniques rapid and sensitive detection is possible.
  • the operation is simple.
  • a substance having specific binding property to the biomarker to be used is used.
  • an antibody is usually used, but any substance can be used as long as it has a specific binding property to the biomarker and can measure the binding amount.
  • the antibody newly prepared using the immunological method, the phage display method, the ribosome display method etc. may be used.
  • Examples of measurement methods include immunohistochemistry, fluorescence immunoassay (FIA method), enzyme immunoassay (EIA method), radioimmunoassay (RIA method), and Western blot method.
  • the detection target can be detected quickly and with high sensitivity. Also, the operation is simple. Therefore, the burden on the subject (patient) accompanying the detection is reduced.
  • immunohistochemical staining of living tissues generally, (1) fixation / paraffin embedding (or frozen embedding), (2) deparaffinization (not required for free embedding), (3) primary antibody reaction, ( 4) Add the labeling reagent, (5) color reaction, and (6) dehydration, penetration, and encapsulation.
  • antigen activation treatment endogenous peroxidase removal treatment (when peroxidase is used as a labeling substance), nonspecific reaction inhibition (treatment with bovine serum albumin solution, etc.) are performed. Is called.
  • nuclear staining for example, Mayer's hematoxylin can be used
  • immunohistochemical staining of biological tissues for example, “Enzyme Antibody Method, Revised 3rd Edition”, edited by Keiichi Watanabe and Kazuho Nakane, interdisciplinary project).
  • step (3) the malignancy of the melanocyte tumor is determined based on the detection result.
  • control for example, the biomarker level of healthy human skin tissue or the biomarker level of nevi (benign) can be used.
  • the determination of malignancy may be either qualitative or quantitative. It should be noted that the determination here can be automatically / mechanically performed without depending on the determination of a person having specialized knowledge such as a doctor or a laboratory technician, as is apparent from the determination criteria.
  • the biomarker of the present invention includes a biomarker (PDZRN3, DTX3L and CSS3) in which an increase in expression level correlates with malignancy of melanocyte tumor, and a biomarker (BRP44L) in which a decrease in expression level correlates with malignancy of melanocyte tumor )).
  • a biomarker PDZRN3, DTX3L and CSS3
  • BRP44L biomarker
  • the former is referred to as a first group of biomarkers
  • the latter is referred to as a second group of biomarkers.
  • the following criteria are employed for each biomarker. Needless to say, the criterion “high detection value is high malignancy” is synonymous with the criterion “low detection value is low malignancy” (the same applies to other criteria).
  • Biomarkers that adopt the criteria of“ High detection value means high malignancy ”or“ Detection is high malignancy ”: Biomarkers of the first group (PDZRN3, DTX3L, CSS3)
  • Biomarker of the second group (BRP44L)
  • Second group biomarker determination example 1 When the detected value of the biomarker (the level in the sample) is higher than the reference value, it is judged as “malignant” or “high possibility of malignancy”, and the detected value of the biomarker (the level in sample) is higher than the reference value Is low, it is judged as “benign” or “high possibility of benign”.
  • a reference value 1 for distinguishing benign and primary melanoma and a reference value 2 for distinguishing primary melanoma and metastatic melanoma are set (reference value 1 ⁇ reference value 2), and the determination is made as follows. It should be noted that a specific probability (for example, a determination of “80% or more probability”) may be shown instead of the evaluation “high possibility”.
  • a plurality of reference values that divide the malignancy level are set and determined as follows. In this example, four reference values (a ⁇ b ⁇ c ⁇ d) are set, but the number of reference values and the number of malignancy levels associated therewith are not limited to this. If the detected value of the biomarker ⁇ a: Grade 1 When a ⁇ biomarker detection value ⁇ b: Grade 2 When b ⁇ detection value of biomarker ⁇ c: Grade 3 When c ⁇ biomarker detection value ⁇ d: Grade 4 When d ⁇ biomarker detection value: Grade 5
  • a reference value 1 for distinguishing benign and primary melanoma and a reference value 2 for distinguishing primary melanoma and metastatic melanoma are set (reference value 1 ⁇ reference value 2), and the determination is made as follows. It should be noted that a specific probability (for example, a determination of “80% or more probability”) may be shown instead of the evaluation “high possibility”.
  • a plurality of reference values that divide the malignancy level are set and determined as follows. In this example, four reference values (a ⁇ b ⁇ c ⁇ d) are set, but the number of reference values and the number of malignancy levels associated therewith are not limited to this.
  • Biomarker detection value ⁇ a Grade 5 When a ⁇ biomarker detection value ⁇ b: Grade 4 When b ⁇ detection value of biomarker ⁇ c: Grade 3 When c ⁇ biomarker detection value ⁇ d: Grade 2 If d ⁇ biomarker detection value: Grade 1
  • each of the biomarkers 1 and 2 is the first group
  • a determination result may be obtained.
  • a determination result can be obtained according to the following examples.
  • determination may be made as in the following (1) or (2).
  • (1) “malignant” or “malignant” when positive for all of the biomarkers included in the combination greater than or equal to the cutoff value in the case of the first group biomarker and less than or equal to the cutoff value in the case of the second group biomarker) It is determined that “the possibility of malignancy is high”, and other cases are determined as “benign” or “high possibility of benign”.
  • diagnosis (detection) sensitivity and diagnosis (detection) specificity vary depending on the type and number of biomarkers to be combined. Therefore, an optimal combination of biomarkers may be selected according to the purpose. For example, combinations with high diagnostic sensitivity are suitable for screening tests. In contrast, a combination with a high diagnostic specificity is suitable for a test that requires a more reliable determination (for example, a secondary test or a tertiary test). By combining determination methods with different balances of diagnostic sensitivity and diagnostic specificity, it is possible to improve efficiency and improve accuracy or reliability.
  • a final determination is made using a combination of biomarkers giving high diagnostic specificity (secondary test). Not only such a two-step determination but also a three-step or higher determination can be performed.
  • the number of judgment categories, the level of the biomarker associated with each judgment category, and the judgment results can be arbitrarily set through preliminary experiments or the like without being bound by the above example.
  • the “reference value” when judging the grade of malignancy with a predetermined reference value (threshold) as the boundary, and the “biomarker level range” associated with the classification related to the grade of malignancy use a large number of specimens. Can be determined by statistical analysis.
  • the determination result is used to estimate the prognosis of melanoma.
  • the prognosis of melanoma is estimated based on the determination result of step (3).
  • the prognostic level is set in advance corresponding to the malignancy level (for example, 1: very good, 2: good, 3: relatively good, 4: relatively bad, 5: bad, 6: very good. It is also possible to determine which level corresponds to a plurality of levels.
  • the level of a biomarker measured at a certain time point is compared with the level of a biomarker measured in the past, Or examine the degree of increase or decrease.
  • the data regarding the change in the expression level of the biomarker obtained as a result is useful information for monitoring the malignancy, grasping the therapeutic effect, or estimating the prognosis. Specifically, for example, it can be determined that the grade of malignancy has increased or decreased or has not changed between the previous test and the current test based on the change in the biomarker level.
  • the present invention further provides reagents and kits for testing the malignancy of melanocyte tumors.
  • the reagent of the present invention comprises a substance (hereinafter referred to as “binding molecule”) that exhibits specific binding to the biomarker of the present invention.
  • binding molecules include antibodies, nucleic acid aptamers and peptide aptamers that specifically recognize biomarkers.
  • the type and origin of the binding molecule are not particularly limited as long as it has specific binding properties for the biomarker employed. In the case of antibodies, any of polyclonal antibodies, oligoclonal antibodies (mixtures of several to several tens of antibodies), and monoclonal antibodies may be used.
  • an anti-serum-derived IgG fraction obtained by animal immunization, or an affinity-purified antibody using an antigen can be used.
  • Antibody fragments such as Fab, Fab ′, F (ab ′) 2 , scFv, and dsFv antibodies may also be used.
  • the binding molecule may be prepared by a conventional method. If a commercial item is available, the commercial item may be used.
  • antibodies can be prepared using immunological techniques, phage display methods, ribosome display methods, and the like. Preparation of a polyclonal antibody by an immunological technique can be performed by the following procedure. An antigen (biomarker or a part thereof) is prepared, and an animal such as a mouse, rat or rabbit is immunized using the antigen. An antigen can be obtained by purifying a biological sample. A recombinant antigen can also be used.
  • a recombinant antigen can be obtained by, for example, introducing a gene encoding a biomarker (which may be a part of a gene) into a suitable host using a vector and expressing the gene in a recombinant cell obtained. Can be prepared.
  • an antigen to which a carrier protein is bound may be used.
  • the carrier protein KLH (KeyholeHLimpet) Hemocyanin), BSA (Bovine Serum Albumin), OVA (Ovalbumin) and the like are used.
  • the carbodiimide method, the glutaraldehyde method, the diazo condensation method, the MBS (maleimidobenzoyloxysuccinimide) method, etc. can be used for the coupling
  • an antigen in which a biomarker (or part thereof) is expressed as a fusion protein with GST, ⁇ -galactosidase, maltose-binding protein, histidine (His) tag or the like can also be used.
  • a fusion protein can be easily purified by a general method.
  • Immunization is repeated as necessary, and blood is collected when the antibody titer has sufficiently increased, and serum is obtained by centrifugation or the like. The obtained antiserum is affinity purified to obtain a polyclonal antibody.
  • monoclonal antibodies can be prepared by the following procedure. First, an immunization operation is performed in the same procedure as described above. Immunization is repeated as necessary, and antibody-producing cells are removed from the immunized animal when the antibody titer sufficiently increases. Next, the obtained antibody-producing cells and myeloma cells are fused to obtain a hybridoma. Subsequently, after this hybridoma is monoclonalized, a clone that produces an antibody having high specificity for the target protein is selected. The target antibody can be obtained by purifying the culture medium of the selected clone.
  • the desired antibody can be obtained by growing the hybridoma to a desired number or more, then transplanting it into the abdominal cavity of an animal (for example, a mouse), growing it in ascites, and purifying the ascites.
  • affinity chromatography using protein G, protein A or the like is preferably used.
  • affinity chromatography in which an antigen is immobilized may be used.
  • methods such as ion exchange chromatography, gel filtration chromatography, ammonium sulfate fractionation, and centrifugation can also be used. These methods can be used alone or in any combination.
  • a labeled antibody is used as the specific binding molecule, it is possible to directly detect the amount of the bound antibody using the labeled amount as an index. Therefore, a simpler inspection method can be constructed.
  • an indirect detection method such as a method using a secondary antibody to which a labeling substance is bound or a method using a polymer to which a secondary antibody and a labeling substance are bound.
  • the secondary antibody here is an antibody having a specific binding property to an antibody specific to the biomarker employed.
  • an antibody specific for a biomarker when an antibody specific for a biomarker is prepared as a rabbit antibody, an anti-rabbit IgG antibody can be used as a secondary antibody.
  • Labeled secondary antibodies that can be used against various types of antibodies such as rabbits, goats, and mice are commercially available (for example, Funakoshi Co., Ltd., Cosmo Bio Co., Ltd., etc.) and are appropriate for the reagents of the present invention. Can be appropriately selected and used.
  • labeling substances include peroxidase, microperoxidase, horseradish peroxidase (HRP), alkaline phosphatase, ⁇ -D-galactosidase, enzymes such as glucose oxidase and glucose-6-phosphate dehydrogenase, fluorescein isothiocyanate (FITC), Fluorescent materials such as tetramethylrhodamine isothiocyanate (TRITC) and europium, chemiluminescent materials such as luminol, isoluminol and acridinium derivatives, coenzymes such as NAD, biotin, and radioactive materials such as 131 I and 125 I.
  • HRP horseradish peroxidase
  • alkaline phosphatase ⁇ -D-galactosidase
  • enzymes such as glucose oxidase and glucose-6-phosphate dehydrogenase
  • FITC fluorescein isothiocyanate
  • TRITC
  • the reagent of the present invention is solid-phased according to its use.
  • the insoluble support used for the solid phase is not particularly limited.
  • a resin such as polystyrene resin, polycarbonate resin, silicon resin, nylon resin, or an insoluble support made of a water-insoluble substance such as glass can be used.
  • the antibody can be supported on the insoluble support by physical adsorption or chemical adsorption.
  • the kit of the present invention contains the reagent of the present invention as a main component.
  • Other reagents buffers, blocking reagents, enzyme substrates, coloring reagents, etc.
  • devices or instruments used in performing the test method may be included in the kit.
  • biomarker molecule of the present invention or a fragment thereof as a standard sample in a kit.
  • an instruction manual is attached to the kit of the present invention.
  • the present invention provides a therapeutic agent for melanoma (hereinafter referred to as the medicament of the present invention).
  • the medicament of the present invention comprises a compound that suppresses the expression of a target gene as an active ingredient.
  • the target gene here is a PDZRN3 gene, a DTX3L gene, or a CSS3 gene.
  • the compound that suppresses the expression of the target gene is a compound that suppresses the expression process of the target gene (including transcription, post-transcriptional regulation, translation, and post-translational regulation). Examples of the compound are as follows.
  • “suppression of expression” may be either transient suppression or permanent suppression.
  • siRNA targeting the target gene (a) siRNA targeting the target gene (b) Nucleic acid construct that generates siRNA targeting the target gene in the cell (c) Single-stranded RNA having an expression suppressing sequence that suppresses the expression of the target gene and a complementary sequence that anneals to the sequence (d) Antisense nucleic acid targeting the target gene transcript (e) Ribozyme targeting the target gene transcript
  • RNAi RNA interference
  • the expression of the target gene can be suppressed by RNAi.
  • RNAi is a sequence-specific post-transcriptional gene repression process that can occur in eukaryotic cells.
  • siRNA short double-stranded RNA
  • siRNAs are 21-23 base pairs.
  • dsRNA double-stranded RNA
  • sequence-specific pathways relatively long dsRNAs are split into short interfering RNAs (ie siRNAs).
  • ie siRNAs short interfering RNAs
  • a sequence non-specific pathway is considered to be caused by an arbitrary dsRNA regardless of the sequence as long as it has a predetermined length or more.
  • dsRNA is activated by two enzymes, namely PKR, which stops all protein synthesis by phosphorylating the translation initiation factor eIF2, and 2 ', 5' oligoadenyl, which is involved in the synthesis of RNAase L activation molecules.
  • RNA double stranded RNA
  • RNAi RNA composed of a sense RNA homologous to a part of the mRNA sequence of the target gene and an antisense RNA complementary thereto is introduced into the cell or expressed in the cell.
  • a siRNA composed of a sense RNA homologous to a part of the mRNA sequence of the target gene and an antisense RNA complementary thereto is introduced into the cell or expressed in the cell.
  • the above (a) is a compound corresponding to the former method
  • the above (b) is a compound corresponding to the latter method.
  • the siRNA targeting the target gene (in the present invention, the target gene) usually hybridizes with a sense RNA consisting of a sequence homologous to a continuous region in the mRNA sequence of the gene and an antisense RNA consisting of its complementary sequence. Double stranded RNA.
  • the length of the “continuous region” here is usually 15 to 30 bases, preferably 18 to 23 bases, more preferably 19 to 21 bases.
  • the length of the base that forms the overhang is not particularly limited, but is preferably 2 bases (for example, TT, UU).
  • SiRNA consisting of modified RNA may be used.
  • modifications herein include phosphorothioation and the use of modified bases (eg, fluorescently labeled bases).
  • SiRNA can be designed and prepared by conventional methods. In designing siRNA, a sequence unique to a target sequence (continuous sequence) is usually used. Note that programs and algorithms for selecting an appropriate target sequence have been developed.
  • nucleic acid construct that generates siRNA in a cell refers to a nucleic acid molecule that, when introduced into a cell, produces a desired siRNA (siRNA that causes RNAi against a target gene) by a process in the cell.
  • siRNA siRNA that causes RNAi against a target gene
  • shRNA short hairpin RNA
  • shRNA has a structure (hairpin structure) in which a sense RNA and an antisense RNA are linked via a loop structure part, and the loop structure part is cleaved in a cell to form a double-stranded siRNA, resulting in an RNAi effect.
  • the length of the loop structure is not particularly limited, but is usually 3 to 23 bases.
  • nucleic acid construct is a vector that can express a desired siRNA.
  • Such vectors include those that express shRNA that is converted to siRNA by a later process (inserted with a sequence encoding shRNA) (referred to as stem loop type or short hairpin type), sense RNA and antisense RNA.
  • stem loop type or short hairpin type a sequence encoding shRNA
  • sense RNA a sequence encoding shRNA
  • antisense RNA are vectors that are expressed separately (referred to as tandem type).
  • tandem type Those skilled in the art can prepare these vectors according to conventional methods (Brummelkamp TR et al. (2002) Science 296: 550-553; Lee NS et al. (2001) Nature Biotechnology 19: 500-505; Miyagishi M & Taira K (2002) Nature Biotechnology 19: 497-500; Paddison PJ et al. (2002) Proc. Natl.
  • RNAi vectors are available. You may decide to construct
  • a desired RNA for example, shRNA
  • the origin and structure of the vector are not limited as long as it has a function of generating siRNA that exerts RNAi action on the target gene in the cell. Accordingly, various viral vectors (adenovirus vectors, adeno-associated virus vectors, retrovirus vectors, lentivirus vectors, herpes virus vectors, Sendai virus vectors, etc.), non-viral vectors (liposomes, positively charged liposomes, etc.) and the like can be used. it can.
  • promoters that can be used in the vector are U6 promoter, H1 promoter, and tRNA promoter. These promoters are RNA polymerase III type promoters, and high expression efficiency can be expected.
  • single-stranded RNA having a predetermined structure is useful for suppressing the expression of a target gene (for example, WO2012 / 005368, JP2013-55913A, JP2013-138881A, JP2013-13A). No. 153736). Therefore, in one embodiment of the present invention, single-stranded RNA (above (c)) is used to suppress the expression of a target gene by the same mechanism as the suppression of expression by siRNA (that is, RNA interference).
  • the single-stranded RNA of the present invention has an expression suppressing sequence corresponding to the target gene and a complementary sequence that can be annealed to the sequence. The order in which the expression suppressing sequence and the complementary sequence are linked is not particularly limited.
  • the expression suppressing sequence and the complementary sequence may be directly linked or may be linked via a linker region.
  • the linker region can be composed of nucleotide residues or non-nucleotide residues (for example, comprising a structure such as polyalkylene glycol, pyrrolidine skeleton, piperidine skeleton, etc.).
  • a molecule (Example 2) that forms two double-stranded structures (stem structures) by intramolecular annealing of the side regions separately can be given.
  • the expression-suppressing sequence is a sequence showing an activity of suppressing the expression of the target gene when the single-stranded RNA of the present invention is introduced into the cell.
  • a sequence that causes expression suppression (ie, RNA interference) by siRNA is used as the expression suppression sequence.
  • the sequence of RNA constituting the above-described siRNA can be used as an expression suppressing sequence.
  • the length of the expression suppressing sequence is not particularly limited, but is, for example, 18 to 32 bases long, preferably 19 to 30 bases long, and more preferably 19 to 21 bases long.
  • the length of the single-stranded RNA of the present invention is not particularly limited.
  • the total number of bases constituting the single-stranded RNA has a lower limit of, for example, 38 bases, preferably 42 bases, more preferably 50 bases, still more preferably 51 bases, particularly preferably 52 bases.
  • the upper limit is, for example, 300 bases, preferably 200 bases, more preferably 150 bases, still more preferably 100 bases, particularly preferably 80 bases.
  • the single-stranded RNA of the present invention includes a linker region
  • the total number of bases excluding the linker region is, for example, 38 bases, preferably 42 bases, more preferably 50 bases, more preferably 51 bases, particularly preferably the lower limit.
  • Is 52 bases, and the upper limit is, for example, 300 bases, preferably 200 bases, more preferably 150 bases, still more preferably 100 bases, particularly preferably 80 bases.
  • the above (d) is a compound used for expression suppression by the antisense method.
  • the expression of the target gene can be suppressed by the antisense method.
  • an antisense construct that generates RNA complementary to a unique part of mRNA encoding a target gene when transcribed in the target cell is used.
  • Such an antisense construct is introduced into a target cell, for example, in the form of an expression plasmid.
  • an oligonucleotide probe that hybridizes with an mRNA / or genomic DNA sequence encoding a target gene and inhibits its expression when introduced into the target cell can also be employed.
  • an oligonucleotide probe one that is resistant to endogenous nucleases such as exonuclease and / or endonuclease is preferably used.
  • oligodeoxyribonucleotide derived from a region containing a translation initiation site (for example, a region of -10 to +10) of mRNA encoding the target gene is preferable.
  • the complementarity between the antisense nucleic acid and the target nucleic acid is exact, but some mismatch may exist.
  • the ability of an antisense nucleic acid to hybridize to a target nucleic acid generally depends on both the degree of complementarity and the length of both nucleic acids. Usually, the longer the antisense nucleic acid used, the more stable duplexes (or triplexes) can be formed with the target nucleic acid, even if the number of mismatches is large.
  • One skilled in the art can ascertain the degree of acceptable mismatch using standard techniques.
  • the antisense nucleic acid may be DNA, RNA, a chimeric mixture thereof, or a derivative or modified type thereof. Moreover, it may be single-stranded or double-stranded. By modifying the base moiety, sugar moiety, or phosphate skeleton moiety, the stability, hybridization ability, etc. of the antisense nucleic acid can be improved.
  • antisense nucleic acids can be used to promote cell membrane transport (for example, Letsingeretset al., 1989, Proc. Natl. Acad. Sci. USA 86: 6553-6556; 556Lemaitre et al., 1987, Proc. Natl. Acad. Sci. 84: 648-652; see PCT Publication No. W088 / 09810, published Dec 15, 15, 1988), or substances that enhance affinity for specific cells.
  • the antisense nucleic acid can be synthesized by a conventional method, for example, using a commercially available automatic DNA synthesizer (for example, Applied Biosystems). For example, Stein et al. (1988), Nucl. Acids Res. 16: 3209 and Sarin et al., (1988), Proc. Natl. Acad. Sci. USA 85: 7448- 7451 etc. can be referred to.
  • Strong promoters such as pol II and pol III can be used to enhance the action of antisense nucleic acids in target cells. That is, when a construct containing an antisense nucleic acid arranged under the control of such a promoter is introduced into a target cell, a sufficient amount of the antisense nucleic acid can be transcribed by the action of the promoter.
  • the expression of the antisense nucleic acid can be performed by any promoter (inducible promoter or constitutive promoter) known to function in mammalian cells (preferably human cells).
  • a promoter inducible promoter or constitutive promoter
  • mammalian cells preferably human cells
  • the SV40 early promoter region (Bernoist and Chambon, 1981, Nature 290: 304-310)
  • the promoter derived from the 3 ′ end region of Rous sarcoma virus Yamamoto et al., 1980, Cell 22: 787-797
  • herpes zoster thymidine A promoter such as a kinase promoter (Wagner et al., 1981, Proc. Natl. Acad. Sci. USA 78: 1441-1445) can be used.
  • expression suppression by ribozyme is used (in the case of the compound (e) above).
  • the target mRNA can be destroyed using a ribozyme that cleaves the mRNA with a site-specific recognition sequence, but preferably a hammerhead ribozyme is used.
  • a ribozyme that cleaves the mRNA with a site-specific recognition sequence but preferably a hammerhead ribozyme is used.
  • a hammerhead ribozyme for example, Haseloff and Gerlach, 1988, Nature, 334: 585-591 can be referred to.
  • a ribozyme may be constructed using a modified oligonucleotide.
  • a nucleic acid construct in which DNA encoding the ribozyme is placed under the control of a strong promoter (for example, pol II or pol III) can be used. preferable.
  • the pharmaceutical of this embodiment contains the following (A) (BRP44L protein) or (B) (expression vector holding the BRP44L gene) as an active ingredient, and is used in combination with an anticancer drug or a drug having an antitumor action.
  • BRP44L protein is an active ingredient.
  • a part (fragment) of the BPP44L protein can also be used as an active ingredient as long as it exhibits an effective action on the anticancer drug sensitivity of melanoma cells.
  • the amino acid sequence of human BRP44L protein is registered in a public database (SEQ ID NO: 8: NCBI protein database Accession No .: NP_057182).
  • a polypeptide containing an amino acid sequence equivalent to the amino acid sequence can also be used as the BRP44L protein.
  • the “equivalent amino acid sequence” here is partially different from the reference amino acid sequence (for example, the amino acid sequence of SEQ ID NO: 8), but the difference is related to the function of the protein (anticancer drug sensitivity of melanoma cells). It means an amino acid sequence that has no substantial effect on the effective action. Therefore, substantial functional identity is recognized between the reference amino acid sequence and the equivalent amino acid sequence. In order to determine the presence or absence of substantial functional identity, for example, using the experimental system (animal model or cell evaluation) described in the examples below, the effect on the anticancer drug sensitivity of melanoma cells What is necessary is just to confirm that there is no substantial difference between two amino acid sequences in terms of effect.
  • “Different in part of amino acid sequence” typically means deletion or substitution of 1 to several amino acids (upper limit is 3, 5, 7, 10) constituting the amino acid sequence. Alternatively, it means that a mutation (change) has occurred in the amino acid sequence due to addition, insertion, or a combination of 1 to several amino acids (the upper limit is, for example, 3, 5, 7, 10). Differences in amino acid sequences here are allowed as long as there is no significant decrease in the above functions. As long as this condition is satisfied, the positions where the amino acid sequences are different are not particularly limited, and differences may occur at a plurality of positions.
  • plural refers to, for example, a number corresponding to less than about 30% of all amino acids, preferably a number corresponding to less than about 20%, and more preferably a number corresponding to less than about 10%.
  • the number is preferably less than about 5%, and most preferably less than about 1%. That is, the equivalent amino acid sequence is, for example, about 70% or more, preferably about 80% or more, more preferably about 90% or more, still more preferably about 95% or more, and most preferably about 99% or more with the reference amino acid sequence.
  • conservative amino acid substitution refers to substitution of a certain amino acid residue with an amino acid residue having a side chain having the same properties.
  • a basic side chain eg lysine, arginine, histidine
  • an acidic side chain eg aspartic acid, glutamic acid
  • an uncharged polar side chain eg glycine, asparagine, glutamine, serine, threonine, tyrosine
  • Cysteine eg alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • ⁇ -branched side chains eg threonine, valine, isoleucine
  • aromatic side chains eg tyrosine, phenylalanine, Like tryptophan and histidine.
  • a conservative amino acid substitution is preferably a substitution between amino acid residues within the same family.
  • sequence identity % of two amino acid sequences or two nucleic acid sequences (hereinafter, “two sequences” is used as a term including them) can be determined, for example, by the following procedure.
  • two sequences are aligned for optimal comparison (eg, a gap may be introduced into the first sequence to optimize alignment with the second sequence).
  • a molecule amino acid residue or nucleotide
  • Gapped BLAST described in Altschul et al. (1997) Amino Acids Research 25 (17): 3389-3402 can be used.
  • BRP44L can be easily prepared by using standard genetic engineering techniques, molecular biological techniques, biochemical techniques, etc. with reference to the sequence information disclosed in this specification or the attached sequence listing. it can. For example, it can be prepared by transforming a suitable host cell (for example, E. coli, yeast) with DNA encoding BRP44L and recovering the protein expressed in the transformant. The recovered protein is appropriately purified according to the purpose. Thus, various modifications are possible if BRP44L is obtained as a recombinant protein.
  • suitable host cell for example, E. coli, yeast
  • BRP44L consisting of a recombinant protein linked to any peptide or protein
  • modification may be performed so that addition of sugar chain and / or lipid, or processing of N-terminal or C-terminal may occur.
  • BRP44L by genetic engineering techniques.
  • the method for preparing BRP44L is not limited to the genetic engineering method.
  • BRP44L can also be prepared from natural materials by standard techniques (crushing, extraction, purification, etc.).
  • an expression vector holding the BRP44L gene is used as an active ingredient. As long as it encodes a protein that exhibits an effective action on the anticancer drug sensitivity of melanoma cells, a part thereof may be used instead of the BRP44L gene.
  • “Expression vector” refers to a vector capable of introducing a nucleic acid inserted therein into a target cell (host cell) and allowing expression in the cell. In the expression vector according to the present invention, the BRP44L gene is retained so that it can be expressed. The type of vector is not particularly limited as long as the BRP44L gene can be introduced into the target cell and expressed in the target cell.
  • the “vector” herein includes viral vectors and non-viral vectors.
  • the gene transfer method using a virus vector skillfully utilizes the phenomenon that a virus infects cells, and high gene transfer efficiency can be obtained.
  • Adenovirus vectors, adeno-associated virus vectors, retrovirus vectors, lentivirus vectors, herpes virus vectors, Sendai virus vectors and the like have been developed as virus vectors.
  • Non-viral vectors include liposomes, positively charged liposomes (Felgner, PL, Gadek, TR, Holm, M. et al., Proc. Natl. Acad. Sci., 84: 7413-7417, 1987), HVJ (Hemagglutinating virus of Japan) -Liposome (Dzau, VJ, Mann, M., Morishita, R. et al., Proc. Natl. Acad. Sci., 93: 11421-11425, 1996, Kaneda, Y., Saeki, Y. & Morishita , R., Molecular Med. Today, 5: 298-303, 1999).
  • the expression vector in the present invention may be constructed as such a non-viral vector. Further, a YAC vector, a BAC vector or the like may be used.
  • Retroviral vectors are not suitable for gene transfer into non-dividing cells because cell division is required for integration of the viral genome into the host chromosome.
  • lentivirus vectors and adeno-associated virus vectors cause integration of foreign genes into the host chromosome after infection even in non-dividing cells. Therefore, these vectors are effective for stably and long-term expressing foreign genes in non-dividing cells.
  • Each virus vector can be prepared according to a previously reported method or using a commercially available dedicated kit.
  • an adenovirus vector can be prepared by the COS-TPC method or full-length DNA introduction method.
  • the COS-TPC method is a homologous recombination that occurs in 293 cells by co-transfecting a recombinant cosmid incorporating the target cDNA or expression cassette and a parent virus DNA-terminal protein complex (DNA-TPC) into 293 cells.
  • DNA-TPC parent virus DNA-terminal protein complex
  • the full-length DNA introduction method is a method for producing a recombinant adenovirus by subjecting a recombinant cosmid inserted with a target gene to restriction digestion and then transfecting 293 cells (Miho Terashima, Koki Kondo). Hiromi Kanegae, Izumi Saito (2003) Experimental Medicine 21 (7) 931.).
  • the COS-TPC method can be performed using Adenovirus® Expression® Vector® Kit® (Dual® Version) (Takara Bio Inc.) and Adenovirus® genome® DNA-TPC (Takara Bio Inc.).
  • the full-length DNA introduction method can be performed using Adenovirus® Expression® Vector® Kit® (Dual® Version) (Takara Bio Inc.).
  • retroviral vectors can be prepared by the following procedure. First, the virus genome (gag, pol, env gene) other than the packaging signal sequence between the LTRs (Long Terminal Repeat) existing at both ends of the virus genome is removed, and the target gene is inserted therein. The viral DNA thus constructed is introduced into a packaging cell that constitutively expresses the gag, pol, and env genes. Thereby, only the vector RNA having the packaging signal sequence is incorporated into the viral particle, and a retroviral vector is produced.
  • the virus genome gag, pol, env gene
  • LTRs Long Terminal Repeat
  • the expression vector of the present invention may be constructed as such a viral vector.
  • the BRP44L gene inserted into the expression vector is preferably composed of a base sequence described in SEQ ID NO: 7 (NCBI nucleotide database Accession No .: NM_016098.
  • a DNA having a base sequence equivalent to the base sequence hereinafter “equivalent” (Referred to as “DNA”) can also be used as the BRP44L gene.
  • the “equivalent base sequence” here is partly different from the reference base sequence (SEQ ID NO: 7), but it is encoded by the difference.
  • a base sequence in which the function of the protein (effective effect on the sensitivity of melanoma cells to anticancer drugs) is not substantially affected.
  • the specific example of equivalent DNA is complementary to the reference base sequence.
  • stringent conditions form a so-called specific hybrid that is non-specific.
  • stringent conditions are known to those skilled in the art, such as Molecular ⁇ ⁇ ⁇ Cloning (Third Edition, Cold Spring Harbor Laboratory Press, New York) and Current protocols in molecular biology (edited by Frederick M Ausubel et al., 1987)
  • hybridization solution 50% formamide, 10 ⁇ SSC (0.15M NaCl, 15 mM sodium citrate, pH 7.0), 5 ⁇ Denhardt solution, 1% SDS, 10% dextran sulfate, 10 ⁇ g / ml denatured salmon sperm DNA, 50 mM phosphate buffer (pH 7.5)
  • hybridization solution 50% formamide, 10 ⁇ SSC (0.15M NaCl, 15 mM sodium citrate, pH 7.0
  • Denhardt solution 1% SDS
  • 10% dextran sulfate 10 ⁇ g / ml denatured salmon sperm DNA, 50 mM phosphate buffer
  • More preferred stringent conditions include, for example, 50 hybridization solutions. Formamide, 5 ⁇ SSC (0.15M NaCl, 15 mM sodium citrate, pH 7.0), 1 ⁇ Denhardt solution, 1% SDS, 10% dextran sulfate, 10 ⁇ g / ml denatured salmon sperm DNA, 50 mM phosphate buffer (pH 7.5) ) Can be mentioned.
  • equivalent DNA it consists of a base sequence that includes substitution, deletion, insertion, addition, or inversion of one or more bases relative to a reference base sequence, and is effective in anticancer drug sensitivity of melanoma cells.
  • Examples thereof include DNA encoding a protein that exhibits an effective action on the protein.
  • Base substitution or deletion may occur at a plurality of sites.
  • the term “plurality” as used herein refers to, for example, 2 to 40 bases, preferably 2 to 20 bases, more preferably 2 to 10 bases, although it depends on the position and type of amino acid residues in the three-dimensional structure of the protein encoded by the DNA It is.
  • Such equivalent DNAs include, for example, restriction enzyme treatment, treatment with exonuclease and DNA ligase, position-directed mutagenesis (MolecularMCloning, Third Edition, Chapter 13, Cold Spring Harbor Laboratory Press, New York) Includes substitutions, deletions, insertions, additions, and / or inversions of bases using mutation introduction methods (Molecular Cloning, ingThird Edition, Chapterhap13, Cold Spring Harbor Laboratory Press, New York) Thus, it can obtain by modifying DNA which has a standard base sequence.
  • the equivalent DNA can also be obtained by other methods such as ultraviolet irradiation.
  • DNA in which a base difference as described above is recognized due to a polymorphism represented by SNP (single nucleotide polymorphism).
  • the BRP44L gene can be prepared by using standard genetic engineering techniques, molecular biological techniques, biochemical techniques, etc. with reference to the sequence information disclosed in this specification or the attached sequence listing.
  • the BRP44L gene can be isolated (and amplified) from a human cDNA library by appropriately using an oligonucleotide probe / primer that can specifically hybridize to the BRP44L gene.
  • the oligonucleotide probe primer for example, DNA complementary to the base sequence shown in SEQ ID NO: 7 or a continuous part thereof is used. Oligonucleotide probes and primers can be easily synthesized using a commercially available automated DNA synthesizer.
  • Molecular® Cloning • Third • Edition, • Cold®Spring®Harbor®Laboratory®Press, and “New York” are helpful.
  • An equivalent DNA can be prepared by using a cDNA library derived from a non-human mammalian cell (for example, monkey, mouse, rat, pig, bovine) instead of the human cDNA library.
  • a cDNA library derived from a non-human mammalian cell (for example, monkey, mouse, rat, pig, bovine) instead of the human cDNA library.
  • a non-human mammalian cell for example, monkey, mouse, rat, pig, bovine
  • the sequence of the mouse-derived Brp44l gene is shown in SEQ ID NO: 15.
  • PDZRN3, DTX3L, CSS3 and BRP44L significantly increased (PDZRN3, DTX3L, CSS3) or decreased the expression level in both mouse melanoma without Braf mutation and human melanoma with BRAF mutation. (BRP44L) was observed (Examples described later). Therefore, it can be said that PDZRN3, DTX3L, CSS3 and BRP44L are useful as target molecules in the treatment of melanoma regardless of the presence or absence of BRAF mutation.
  • the medicament of the present invention is applied to melanomas that are resistant to BRAF inhibitors.
  • the medicament of this embodiment is intended for treatment of melanoma patients who have acquired resistance to a BRAF inhibitor as a result of treatment with the BRAF inhibitor.
  • BRAF inhibitors are PLX4032 and PLX4720.
  • DTX3L is a target molecule. Therefore, as a preferred embodiment, a medicament comprising a compound that suppresses the expression of the DTX3L gene as an active ingredient is provided.
  • the pharmaceutical preparation of the present invention can be prepared according to a conventional method.
  • other pharmaceutically acceptable ingredients for example, carriers, excipients, disintegrants, buffers, emulsifiers, suspending agents, soothing agents, stabilizers, preservatives, preservatives, physiological Saline solution and the like.
  • excipient lactose, starch, sorbitol, D-mannitol, sucrose and the like can be used.
  • As the disintegrant starch, carboxymethylcellulose, calcium carbonate and the like can be used. Phosphate, citrate, acetate, etc. can be used as the buffer.
  • emulsifier gum arabic, sodium alginate, tragacanth and the like can be used.
  • suspending agent glyceryl monostearate, aluminum monostearate, methyl cellulose, carboxymethyl cellulose, hydroxymethyl cellulose, sodium lauryl sulfate and the like can be used.
  • soothing agent benzyl alcohol, chlorobutanol, sorbitol and the like can be used.
  • stabilizer propylene glycol, diethylin sulfite, ascorbic acid or the like can be used.
  • preservatives phenol, benzalkonium chloride, benzyl alcohol, chlorobutanol, methylparaben, and the like can be used.
  • preservatives benzalkonium chloride, paraoxybenzoic acid, chlorobutanol and the like can be used.
  • the dosage form for formulation is not particularly limited. Examples of dosage forms are tablets, powders, fine granules, granules, capsules, syrups, injections, external preparations, and suppositories.
  • the medicament of the present invention contains an active ingredient in an amount necessary for obtaining an expected therapeutic effect (or preventive effect) (ie, a therapeutically effective amount).
  • the amount of the active ingredient in the medicament of the present invention generally varies depending on the dosage form, but the amount of the active ingredient is set, for example, within the range of about 0.1 wt% to about 95 wt% so as to achieve a desired dose.
  • the medicament of the present invention is administered to the subject by oral administration or parenteral administration (intravenous, intraarterial, subcutaneous, intradermal, intramuscular or intraperitoneal injection, transdermal, nasal, transmucosal, etc.) depending on the dosage form.
  • oral administration or parenteral administration intravenous, intraarterial, subcutaneous, intradermal, intramuscular or intraperitoneal injection, transdermal, nasal, transmucosal, etc.
  • parenteral administration intravenous, intraarterial, subcutaneous, intradermal, intramuscular or intraperitoneal injection, transdermal, nasal, transmucosal, etc.
  • these administration routes are not mutually exclusive, and two or more arbitrarily selected can be used in combination (for example, intravenous injection or the like is performed simultaneously with oral administration or after a predetermined time has elapsed).
  • a nucleic acid construct is an active ingredient (for example, an embodiment using RNAi)
  • ex vivo administration can be employed
  • the “subject” to which the medicament of the present invention is administered is not particularly limited, and includes humans and non-human mammals (pet animals, domestic animals, laboratory animals. Specifically, for example, mice, rats, guinea pigs, hamsters, monkeys, cows) Pigs, goats, sheep, dogs, cats, chickens, quails, etc.).
  • the medicament of the present invention is applied to humans.
  • the dosage varies depending on the patient's symptoms, age, sex, weight, etc., those skilled in the art can appropriately set an appropriate dosage. In setting the administration schedule, it is possible to consider the patient's symptoms and the duration of the effect of the medicine.
  • the present application also provides a method for treating melanoma, which comprises administering a therapeutically effective amount of the medicament of the present invention to a melanoma patient.
  • RET-mice 304 / B6 mice (Kato, M. et al., J Invest Dermatol. 111: 640-4 .; Iwamoto, T. et al., EMBO J. 10: 3167-75 .; Kato, M et al., Oncogene. 17: 1885-8, etc.) was used to collect melanocyte benign tumors and melanoma.
  • the pCMV-c-Fa-Puro3 vector (Invitrogen) was used for the production of the gene forced expression cell line. After introducing the cDNA of the protein coding region into the pCMV-c-Fa-Puro3 vector, the cultured cells were transfected, and the cells were selected with 1 mg / ml puromycin (Wako Pure Chemical Industries, Ltd.). The pRNAT-U6-1-Neo vector (Invitrogen) was used for establishment of the knockdown cell line. After introducing the shRNA sequence into the pRNAT-U6-1-Neo vector, the cultured cells were transfected, and cells were selected with 1 mg / ml neomycin (Wako Pure Chemical Industries, Ltd.).
  • Human PDZRN3 gene SEQ ID NO: 1 (NCBI nucleotide database Accession No .: NM_015009)
  • Human PDZRN3 protein SEQ ID NO: 2 (NCBI protein database Accession No .: NP_055824)
  • Human DTX3L gene SEQ ID NO: 3 (NCBI nucleotide database Accession No .: NM_138287)
  • Human DTX3L protein SEQ ID NO: 4 (NCBI protein database Accession No .: NP_612144)
  • Human CSS3 gene SEQ ID NO: 5 (NCBI nucleotide database Accession No .: NM_175856)
  • Human CSS3 protein SEQ ID NO: 6 (NCBI protein database Accession No .: NP_787052)
  • Human BRP44L gene SEQ ID NO: 7 (NCBI nucleotide database Accession No .: NM_016098) Human
  • Mouse Pdzrn3 gene SEQ ID NO: 9 (NCBI nucleotide database Accession No .: NM_018884) Mouse Pdzrn3 protein: SEQ ID NO: 10 (NCBI protein database Accession No .: NP_061372) Mouse Dtx3l gene: SEQ ID NO: 11 (NCBI nucleotide database Accession No .: NM_001013371) Mouse Dtx3l protein: SEQ ID NO: 12 (NCBI protein database Accession No .: NP_001013389) Mouse Css3 gene: SEQ ID NO: 13 (NCBI nucleotide database Accession No .: NM_001081328) Mouse Css3 protein: SEQ ID NO: 14 (NCBI protein database Accession No .: NP_001074797) Mouse Brp44l gene: SEQ ID NO: 15 (NCBI nucleotide database Accession No .: NM_018819) Mouse Brp44l protein: SEQ ID NO: 16 (NC
  • mRNA was purified from spontaneously occurring melanocyte benign tumors and melanomas in melanoma model mice (RET-Tg), and gene expression analysis was performed using DNA microarrays. Genes with stronger or weaker expression in melanoma compared to benign tumors Thus, PDZ domain containing ring finger 3 (PDZRN3), Deltex-3-like (DTX3L), Chondroitin sulfate synthase 3 (CSS3), and BRP44L gene were identified as melanoma-related genes.
  • PDZ domain containing ring finger 3 PDZRN3
  • DTX3L Deltex-3-like
  • CCS3 Chondroitin sulfate synthase 3
  • BRP44L BRP44L gene were identified as melanoma-related genes.
  • the B16 cell line is known to have lower invasive ability (metastatic activity) than other cell lines. Compared with B16 with low invasive ability, the expression level in other cell lines with high invasive ability is greatly increased (FIG. 2b), suggesting that Pdzrn3 may be associated with invasive ability.
  • the expression level of PDZRN3 protein in human melanoma tissues was measured by quantifying the staining level after immunostaining with anti-PDZRN3 antibody.
  • the expression levels were categorized into 3 (negative / low, moderate, high) and the ratio of each expression level was compared.
  • the expression levels in primary and metastatic melanoma were significantly higher than those in benign tumors. It became clear (Fig. 3).
  • DTX3L DTX3L gene and protein expression levels in benign tumors and malignant melanoma spontaneously developed in RET-Tg mice were measured using real-time PCR (FIG. 4A), Western blotting (FIG. 4B) and immunohistochemical staining (FIG. 4C).
  • the expression level of DTX3L gene and protein was significantly increased in malignant melanoma compared to the expression level in benign tumors.
  • the expression level of Dtx3l protein in mouse melanoma cell lines (4 types) was measured using Western blotting (FIG. 5A).
  • the B16 cell line is known to have lower invasive ability (metastatic activity) than other cell lines.
  • CSS3 CSS3 has the same family genes, CSS1 and CSS2. As a result of measuring the expression levels of these three genes in benign tumors and malignant melanomas spontaneously developed in melanoma model mice (RET-Tg) using real-time PCR, the expression levels of CSS1 and CSS2 genes are both benign and malignant and low. However, the expression level of CSS3 gene was significantly increased in malignant melanoma (FIG. 7). The expression level of CSS3 protein in human melanoma tissue (benign tumor: Benign, primary melanoma: Malignant, metastatic melanoma: Meta) was measured by quantifying the staining level after immunostaining with anti-CSS3 antibody (FIG. 8). ). As a result of categorizing the expression level into 3 (negative / low, moderate, high) and comparing the ratio of each expression level, the expression level in primary melanoma and metastatic melanoma is significantly higher than in benign tumors It became clear.
  • BRP44L BRP44L gene and protein expression levels in benign tumors and malignant melanomas spontaneously developed in melanoma model mice were measured using real-time PCR (FIG. 9A) and Western blotting (FIG. 9B).
  • BRP44L mRNA and protein had decreased expression in melanoma compared to benign tumors.
  • the expression level of BRP44L mRNA in human melanoma tissue was measured using real-time PCR. As a result, the expression level in human melanoma tissue was significantly reduced compared to that in benign tumor. (Fig. 10).
  • FIG. 12a A stable strain with reduced expression of PDZRN3 was injected into the tail vein of nude mice, and the subsequent metastatic activity to the lung was investigated (FIG. 12a).
  • the cell line (shPdzrn3) in which the expression level of PDZRN3 was reduced was greatly inhibited from infiltrating into the lung (metastasis) compared to the control line (shControl).
  • the activation (phosphorylation) level of signal molecule (FAK) related to invasion ability in each stable strain was investigated, the phosphorylation (activity) level of FAK decreased in all PDZRN3 expression-suppressed clones (P1 to P3).
  • FIG. 12b it was suggested that PDZRN3 may control the activity of invasive ability-related signal molecule (FAK).
  • the stable strain was injected into the tail vein of nude mice, and the subsequent metastatic activity to the lung was investigated. Since the injected cells express GFP, lung metastasized cells can be visualized (FIG. 15A). The decrease in the expression level of DTX3L significantly inhibited infiltration (metastasis) of melanoma cells into the lung (FIG. 15).
  • Recently developed molecular therapeutics for BRAF, PLX4032 and PLX4720 are one of the new therapeutic candidates. However, long-term treatment with melanoma produces resistant cells and recurs frequently.
  • the human melanoma cell line (A375P) was exposed to the melanoma molecule targeted drug PLX4720 for a long period of time to create a resistant strain, and the effect on the invasive potential when DTX3L expression was reduced by siRNA introduction (FIG. 16A). Measured by invasion assay ( Figure 16B). Suppressing the expression of DTX3L is also effective against PLX-resistant melanoma cells, and a therapeutic effect on drug-resistant melanoma produced by existing molecular target reagents can be expected.
  • FIG. 17 A stable strain in which CSS3 was forcibly expressed in a mouse melanoma cell line (B16F10) was established, and the invasive ability of each stable strain was measured by an invasion assay (FIG. 17). It was revealed that forced expression of CSS3 further activated the invasive ability inherent in melanoma cells. Using a shRNA forced expression vector, a stable strain with reduced CSS3 expression was established in a mouse melanoma cell line (B16F10 FLAG-CSS3) in which CSS3 was forcibly expressed, and the invasion ability of each stable strain was measured by an invasion assay ( FIG. 18a). Decrease in the expression level of CSS3 inhibited the invasive ability of melanoma cells.
  • B16F10 control cells, CSS3 forced expression strain (+ CSS3), and expression-suppressed stable strain (CSS3 sh) were injected into the tail vein of nude mice, and then examined for metastatic activity to the lung. It was revealed that the number of metastases increased and the number of metastases decreased by suppressing the expression (FIG. 18b).
  • BRP44L Cell death by treatment of human melanoma cell line SKMel28 (SK-GFP # 6) and stable strains forcibly expressing BRP44L (SK-B # 7, SK-B # 8) with one kind of etoposide anticancer agent And cell viability was measured (FIG. 19). Under the high expression of BRP44L, cell death induction by etoposide was more strongly induced, suggesting that forced expression of BRP44L may increase the sensitivity to anticancer drugs.
  • the four genes and proteins analyzed this time can inhibit melanoma activity very sensitively by suppressing or enhancing their expression (see FIGS. 11 to 19).
  • PDZRN3, DTX3L, and CSS3 are mainly involved in invasion and metastasis, and BRP44L is involved in anticancer drug-induced cell death sensitivity.
  • BRP44L is involved in anticancer drug-induced cell death sensitivity.
  • the newly identified melanoma-related molecule (a candidate molecule for molecular target therapy for melanoma) can be said to be effective for the problems of melanoma therapeutic agents such as PLX4032 and PLX4720.
  • New melanoma-related molecules are deeply involved in melanoma metastasis and sensitivity to anticancer drugs.
  • the melanoma-related molecule has a particularly poor prognosis among various carcinomas, and can be expected to make a great contribution to the diagnosis and treatment of melanoma, which requires early detection for effective treatment.
  • the biomarker of the present invention is useful for early detection of melanoma, determination of malignancy of melanoma, and the like.
  • biomolecules constituting biomarkers are deeply involved in melanoma invasion, metastasis, sensitivity to anticancer agents, and the like, and are useful as therapeutic targets. Therefore, not only the diagnosis of melanoma but also the application or application of the present invention to the treatment of melanoma is expected.

Abstract

La présente invention résoud le problème consistant à obtenir une molécule de marquage permettant le diagnostic hautement efficace du mélanome, et concerne une nouvelle molécule sible thérapeutique pouvant être utilisée dans une méthode thérapeutique efficace. PDZRN3, DTX3L, CSS3 et BRP44L sont utilisés comme marqueurs spécifiques du mélanome. L'invention concerne également une méthode de test, une méthode thérapeutique et d'autres méthodes utilisant chacune ces molécules biologiques.
PCT/JP2016/054968 2015-03-20 2016-02-20 Biomarqueur spécifique du mélanome et son utilisation WO2016152352A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2017507616A JPWO2016152352A1 (ja) 2015-03-20 2016-02-20 メラノーマ特異的バイオマーカー及びその利用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2015058692 2015-03-20
JP2015-058692 2015-03-20

Publications (1)

Publication Number Publication Date
WO2016152352A1 true WO2016152352A1 (fr) 2016-09-29

Family

ID=56977316

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2016/054968 WO2016152352A1 (fr) 2015-03-20 2016-02-20 Biomarqueur spécifique du mélanome et son utilisation

Country Status (2)

Country Link
JP (1) JPWO2016152352A1 (fr)
WO (1) WO2016152352A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018143475A1 (fr) * 2017-02-06 2018-08-09 日産化学工業株式会社 Oligonucléotide monobrin
CN108884462A (zh) * 2016-01-26 2018-11-23 日产化学株式会社 单链寡核苷酸
CN109985244A (zh) * 2017-12-29 2019-07-09 广州威溶特医药科技有限公司 E3连接酶抑制剂和溶瘤病毒在制备抗肿瘤药物的应用
CN110241219A (zh) * 2019-07-18 2019-09-17 北京泱深生物信息技术有限公司 Myom3在黑色素瘤转移中的应用
CN110273002A (zh) * 2019-07-18 2019-09-24 北京泱深生物信息技术有限公司 生物标志物在黑色素瘤转移诊断中的应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010537629A (ja) * 2007-08-30 2010-12-09 アカデミシュ ジーケンハウス ビイ デ ユニヴェアズィテート ファン アムステルダム 動脈形成に関与する経路及びその使用
JP2011520451A (ja) * 2008-05-14 2011-07-21 ダームテック インターナショナル 核酸解析による黒色腫および日光黒子の診断法
WO2013148845A1 (fr) * 2012-03-30 2013-10-03 The Procter & Gamble Company Système d'identification de liens entre des perturbagènes et des gènes associés à une hyperpigmentation cutanée
WO2013192616A1 (fr) * 2012-06-22 2013-12-27 Htg Molecular Diagnostics, Inc. Malignité moléculaire dans des lésions mélanocytiques

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010537629A (ja) * 2007-08-30 2010-12-09 アカデミシュ ジーケンハウス ビイ デ ユニヴェアズィテート ファン アムステルダム 動脈形成に関与する経路及びその使用
JP2011520451A (ja) * 2008-05-14 2011-07-21 ダームテック インターナショナル 核酸解析による黒色腫および日光黒子の診断法
WO2013148845A1 (fr) * 2012-03-30 2013-10-03 The Procter & Gamble Company Système d'identification de liens entre des perturbagènes et des gènes associés à une hyperpigmentation cutanée
WO2013192616A1 (fr) * 2012-06-22 2013-12-27 Htg Molecular Diagnostics, Inc. Malignité moléculaire dans des lésions mélanocytiques

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
DANTE L. ET AL.: "Cloning and developmental expression of zebrafish pdzrn3.", INT J DEV BIOL., vol. 55, no. 10-12, 2011, pages 989 - 993, XP055317704 *
DATABASE EMBL 24 May 2008 (2008-05-24), ISOGAI T ET AL.: "Homo sapiens brain protein 44-like (BRP44L), mRNA.", XP055317729, Database accession no. AK312201 *
THANG ND. ET AL.: "Deltex-3-like (DTX3L) stimulates metastasis of melanoma through FAK/PI3K/AKT but not MEK/ERK pathway.", ONCOTARGET, vol. 6, no. 16, 10 June 2015 (2015-06-10), pages 14290 - 14299, XP055317738 *
YADA T. ET AL.: "Chondroitin sulfate synthase-3. Molecular cloning and characterization.", J BIOL CHEM., vol. 278, no. 41, 2003, pages 39711 - 39725, XP002976568 *
YAJIMA I ET AL.: "Functional analysis of GNG2 in human malignant melanoma cells.", J DERMATOL SCI., vol. 68, no. 3, 2012, pages 172 - 178, XP055317739 *
YAJIMA I. ET AL.: "Reduced GNG2 expression levels in mouse malignant melanomas and human melanoma cell lines.", AM J CANCER RES., vol. 2, no. 3, 2012, pages 322 - 329, XP055317709 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108884462A (zh) * 2016-01-26 2018-11-23 日产化学株式会社 单链寡核苷酸
US11530409B2 (en) 2016-01-26 2022-12-20 Nissan Chemical Corporation Single-stranded oligonucleotide
WO2018143475A1 (fr) * 2017-02-06 2018-08-09 日産化学工業株式会社 Oligonucléotide monobrin
JPWO2018143475A1 (ja) * 2017-02-06 2019-12-12 日産化学株式会社 一本鎖オリゴヌクレオチド
JP7188087B2 (ja) 2017-02-06 2022-12-13 日産化学株式会社 一本鎖オリゴヌクレオチド
US11572558B2 (en) 2017-02-06 2023-02-07 Nissan Chemical Corporation Single-stranded oligonucleotide
JP2023025156A (ja) * 2017-02-06 2023-02-21 日産化学株式会社 一本鎖オリゴヌクレオチド
CN109985244A (zh) * 2017-12-29 2019-07-09 广州威溶特医药科技有限公司 E3连接酶抑制剂和溶瘤病毒在制备抗肿瘤药物的应用
CN110241219A (zh) * 2019-07-18 2019-09-17 北京泱深生物信息技术有限公司 Myom3在黑色素瘤转移中的应用
CN110273002A (zh) * 2019-07-18 2019-09-24 北京泱深生物信息技术有限公司 生物标志物在黑色素瘤转移诊断中的应用
CN110241219B (zh) * 2019-07-18 2024-04-16 北京泱深生物信息技术有限公司 Myom3在黑色素瘤转移中的应用

Also Published As

Publication number Publication date
JPWO2016152352A1 (ja) 2018-01-11

Similar Documents

Publication Publication Date Title
Guo et al. RNA demethylase ALKBH5 prevents pancreatic cancer progression by posttranscriptional activation of PER1 in an m6A-YTHDF2-dependent manner
Liu et al. PRMT5-dependent transcriptional repression of c-Myc target genes promotes gastric cancer progression
EP2999797B1 (fr) Isoformes spécifiques de gata6 et nkx2-1 en tant que nouveaux marqueurs pour les approches de diagnostic et de thérapie du cancer et en tant que cibles pour la thérapie anticancéreuse
WO2016152352A1 (fr) Biomarqueur spécifique du mélanome et son utilisation
TWI816712B (zh) 癌促進因子表現抑制劑的有效成分之篩選用試藥及其篩選方法、癌之預防或治療劑的有效成分之篩選用試藥及其篩選方法、癌促進因子表現抑制劑及癌之預防或治療劑
WO2010011642A2 (fr) Procédés et compositions employant des protéines de régulation de l'épissage impliquées dans la suppression de tumeurs
Chen et al. SOX6 represses tumor growth of clear cell renal cell carcinoma by HMG domain‐dependent regulation of Wnt/β‐catenin signaling
JP2017006117A (ja) 肺癌および結腸直腸癌におけるbard1アイソフォームおよびその使用
Wang et al. Estrogen receptor beta increases clear cell renal cell carcinoma stem cell phenotype via altering the circPHACTR4/miR‐34b‐5p/c‐Myc signaling
WO2015093557A1 (fr) Nouveau gène de fusion en tant que gène responsable du cancer de l'estomac
US20130149320A1 (en) Asf1b as a Prognosis Marker and Therapeutic Target in Human Cancer
US11510911B2 (en) Method for prediction of susceptibility to sorafenib treatment by using SULF2 gene, and composition for treatment of cancer comprising SULF2 inhibitor
WO2011129427A1 (fr) Agent de diagnostic et agent thérapeutique pour les cancers
WO2019031637A1 (fr) Gènes marqueurs du cancer pour le cancer non mutationnel p53, et procédé de dépistage d'agent thérapeutique
US10865415B2 (en) Prevention, diagnosis and treatment of cancer overexpressing GPR160
CN104630338B (zh) Rrm2b基因或其蛋白在肝癌转移中的应用
CN112899275B (zh) circRHBDD1在制备治疗肝细胞癌的药物中应用
JP2005073548A (ja) 卵巣明細胞腺癌の検査、治療、および治療薬のスクリーニングのための、HNF−1βの利用
US20210164982A1 (en) Pharmaceutical use of actinin-4 involved in induction of cervical cancer
US20230272478A1 (en) Biomarker specific for liver cancer, and use thereof
US20190264292A1 (en) Use of h2a.z.1 as a hepatocellular carcinoma biomarker
KR102042332B1 (ko) 간암의 재발 및 예후 예측용 tcirg1 마커 및 이의 용도
KR101793174B1 (ko) Golgb1 또는 sf3b3을 이용한 재발암의 진단방법 및 golgb1 또는 sf3b3의 억제제를 함유하는 재발암 치료용 조성물
Yu et al. Increase of ASH1L Expression Promotes Hepatocellular Carcinoma Development and Affects the Prognosis of Liver Transplantation Patients
Xiong et al. METTL14 Promotes Lipid Metabolism Reprogramming and Sustains Nasopharyngeal Carcinoma Progression via Enhancing m6A Modification of ANKRD22 mRNA

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: 16768255

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017507616

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16768255

Country of ref document: EP

Kind code of ref document: A1