WO2025063294A1 - Method for diagnosing colorectal cancer and method for evaluating effect of adjuvant chemotheraphy applied to colorectal cancer patient - Google Patents
Method for diagnosing colorectal cancer and method for evaluating effect of adjuvant chemotheraphy applied to colorectal cancer patient Download PDFInfo
- Publication number
- WO2025063294A1 WO2025063294A1 PCT/JP2024/033732 JP2024033732W WO2025063294A1 WO 2025063294 A1 WO2025063294 A1 WO 2025063294A1 JP 2024033732 W JP2024033732 W JP 2024033732W WO 2025063294 A1 WO2025063294 A1 WO 2025063294A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- gene
- subject
- colorectal cancer
- base sequence
- gpc6
- Prior art date
- Legal status (The legal status 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 status listed.)
- Pending
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING 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
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6844—Nucleic acid amplification reactions
- C12Q1/686—Polymerase chain reaction [PCR]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING 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
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
- C12Q1/6883—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
- C12Q1/6886—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
Definitions
- the present invention relates to a method for diagnosing colorectal cancer and a method for evaluating the effectiveness of adjuvant chemotherapy administered to colorectal cancer patients.
- NSCLC non-small cell lung cancer
- EGFR epidermal growth factor receptor
- Liquid biopsy is the most effective approach for MRD monitoring, diagnosing recurrence earlier than conventional diagnostic tools such as high-resolution computed tomography (CT) and magnetic resonance imaging (MRI).
- CT computed tomography
- MRI magnetic resonance imaging
- customized blood CGP tests are expected to play a central role in MRD monitoring in the future and facilitate the rapid selection of appropriate drugs.
- NGS next-generation sequencing
- Ci rculating exosomal microRNA-203 is associated with metastasis possibly via inducing tumor-associated macrophages in colorectal cancer. Oncotarget 2017;8(45): 78598-78613.
- the objective of the present invention is to provide a new technology that can predict the onset and recurrence of colorectal cancer more easily than conventional methods.
- the present invention is as follows.
- a method for detecting colon cancer in a subject comprising determining the methylation status of at least one of the FGD5 gene and the GPC6 gene in the subject.
- the method according to any one of [1] to [3], wherein the determination of the methylation status is carried out based on a liquid sample derived from the subject.
- the liquid sample is a blood sample.
- [6] The method according to any one of [1] to [5], wherein the methylation status is determined by a PCR method.
- a method for evaluating the effect of adjuvant chemotherapy being received by a subject comprising determining the methylation status of at least one of the FGD5 gene and the GPC6 gene in the subject receiving adjuvant chemotherapy for colorectal cancer.
- the method described in [7] further comprising a step of determining the methylation status of the MSC gene in the subject.
- a diagnostic kit for colon cancer including: (1) a PCR primer pair capable of amplifying a part or the whole of the nucleotide sequence of the FGD5 gene; and/or (2) A PCR primer pair capable of amplifying a part or the whole of the nucleotide sequence of the GPC6 gene.
- a PCR primer pair capable of amplifying a part or the whole of the base sequence of the FGD5 gene is a nucleic acid having a base sequence shown in SEQ ID NO: 4 and a nucleic acid having a base sequence shown in SEQ ID NO: 5;
- a PCR primer pair capable of amplifying a part or the whole of the base sequence of the GPC6 gene is a nucleic acid comprising the base sequence shown in SEQ ID NO: 6 and a nucleic acid comprising the base sequence shown in SEQ ID NO: 7.
- PCR primer pair capable of amplifying a part or all of the base sequence of the MSC gene is a nucleic acid having a base sequence shown in SEQ ID NO: 8 and a nucleic acid having a base sequence shown in SEQ ID NO: 9.
- the present invention makes it possible to determine whether a subject is suffering from colorectal cancer and whether colorectal cancer has recurred in a subject after radical surgery for colorectal cancer more quickly than with conventional methods. Furthermore, the present invention also makes it possible to evaluate whether adjuvant chemotherapy for colorectal cancer is effective in a subject receiving the chemotherapy.
- Figure 1 shows an example of the calculation of methylated ctDNA levels by the AMUSE (amplicon of methylated sites using a specific enzyme) assay based on digital PCR findings (hereinafter, the methylated ctDNA level quantified using the AMUSE assay is referred to as the AMUSE score).
- HapII is a methylation site-specific enzyme. Because RNase P contains genomic DNA sequences that cannot be methylated, RNase P cfDNA was used as a control group for HapII treatment. The cfDNA of the target gene and the cfDNA of the RNase P gene that was not treated with HapII were amplified, and the cfDNA (10)/RNase P (5) cfDNA ratio (2) was calculated.
- FIG. 2 is a diagram outlining the flow of the search for methylation biomarkers related to the prediction of colorectal cancer recurrence employed in the present invention.
- CRC colorectal cancer Verification of beta values of methylation sites of FDG5, GPC6, and MSC genes.
- Figure 4 shows the basic characteristics of the three biomarkers selected in the present invention.
- A) The methylation beta values of CpG islands of FGD5, GPC6, and MSC were investigated from 40 cases of colorectal cancer (stage I to IV) tumor/normal tissue pairs.
- FIG. 5-1 shows the confirmation of postoperative recurrence suspected on CT images using the AMUSE score.
- the boxes (AC) under the graphs indicate the duration of ACT (similar to Figures 5B-F and 6).
- the boxes in gray and black indicate the AMUSE score and serum CEA protein level, respectively.
- the date of surgery (P), the suspected recurrence on the images (Q), and the recurrence diagnosis (R) are shown. These six cases have a higher Q than the 28 recurrent cases.
- ACT adjuvant chemotherapy
- AMUSE amplicon of methylated sites using a specific enzyme
- CEA carcinoembryonic antigen
- Figure 5-2 is a continuation of Figure 5-1.
- Figure 6-1 shows the specificity of the AMUSE score in 19 cases without recurrence.
- the box (AC) under the graph indicates the period of ACT.
- Two patients (5G and 5I) showed false positive findings (arrows).
- Figure 6-2 is a continuation of Figure 6-1. 7 shows the clinical benefit of the AMUSE assay.
- D and E Consideration of recurrence cases (D) and all cases (E) by serum CEA.
- AMUSE amplicon of methylated sites using a specific enzyme
- CEA carcinoembryonic antigen
- Figure 8 shows the stratification of postoperative changes in tumor burden affected by adjuvant chemotherapy.
- ACT adjuvant chemotherapy
- MRD minimal residual disease.
- the present invention provides a method for detecting colorectal cancer in a subject (hereinafter, sometimes referred to as the "detection method of the present invention"), comprising a step of determining the methylation status of at least one of the FGD5 gene and the GPC6 gene in the subject.
- the methylation status of at least one of the FGD5 gene and the GPC6 gene is determined.
- the detection method of the present invention further determines the methylation status of the MSC gene.
- the detection method of the present invention determines the methylation status of the FGD5 gene, the GPC6 gene, and the MSC gene.
- the FGD5 gene (FYVE, RhoGEF and PH domain containing 5, Gene ID: 152273), also known as "ZFYVE23", is located at chromosomal position 8q13.3 in humans and is a gene with two exons.
- the FGD5 gene is known to be highly expressed in the spleen and lungs in normal human individuals.
- the base sequence of the human FGD5 gene is shown in SEQ ID NO:1.
- the GPC6 gene (glypican 6, Gene ID: 10082), also known as "OMIMD1", is located at chromosomal location 13q31.3-q32.1 in humans and is a gene with 12 exons.
- the GPC6 gene is known to be highly expressed in the gallbladder and urinary bladder in normal human individuals.
- the base sequence of the human GPC6 gene is shown in SEQ ID NO: 2.
- the MSC gene (musculin, Gene ID: 9242), also known as "ABF1,” “MYOR,” “ABF-1,” “bHLHa22,” etc., is located at chromosomal position 8q13.3 in humans, and the MSC gene, which is a gene with two exons, is known to be highly expressed in the placenta and gallbladder in normal humans.
- the base sequence of the human MSC gene is shown in SEQ ID NO: 3.
- the methylation state of the above-mentioned gene is determined, but the method of determining the methylation state of a gene is known to those skilled in the art, and any method may be used.
- "determining the methylation state of gene A” may also mean “determining the amount of methylation in gene A”.
- the methylation state of a gene can be detected, and the amount can be determined, for example, by methyl-DNA immunoprecipitation (MeDIP), methylation-sensitive restriction enzyme (MSRE) analysis, bisulfite sequencing, etc.
- MeDIP methyl-DNA immunoprecipitation
- MSRE methylation-sensitive restriction enzyme
- the methylation state of a gene can be determined by combining methylation-sensitive restriction enzyme analysis with a PCR method.
- the PCR method combined with methylation-sensitive restriction enzyme analysis is more preferably real-time PCR or digital PCR. Details of the determination of the methylation state by combining methylation-sensitive restriction enzyme analysis with a PCR method are described in the examples of this specification.
- determining the methylation status of gene A means qualitatively analyzing the presence or absence of a methylated region in the entire length or a part of gene A.
- the nucleotide length of the region for determining the methylation status is not particularly limited, but in one embodiment, it is preferable to set a length suitable for PCR amplification.
- the nucleotide length of the region for determining the methylation status may be, but is not limited to, usually 30 to 1000, preferably 50 to 300, more preferably 75 to 200, and particularly preferably 75 to 150 nucleotides. Note that when the region is amplified using a primer, this nucleotide length includes the primer binding site.
- the primer pair that can be used in the PCR reaction can be appropriately designed using a method known per se to amplify part or all of the regions that can be methylated in the FGD5 gene, the GPC6 gene, and the MSC gene.
- the following primer pairs can be used, but are not limited to these.
- Primer pair for FGD5 gene a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:4 and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:5.
- Primer pair for GPC6 gene a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:6, and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:7.
- Primer pair for MSC gene a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:8, and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:9.
- the present invention makes it possible to determine whether a subject is suffering from colorectal cancer, and whether colorectal cancer has recurred in a subject after radical surgery for colorectal cancer, more quickly than with conventional methods. Furthermore, the present invention also makes it possible to evaluate whether adjuvant chemotherapy for colorectal cancer is effective in a subject receiving the chemotherapy. Therefore, the present invention is extremely useful in the field of colorectal cancer treatment.
Landscapes
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Zoology (AREA)
- Genetics & Genomics (AREA)
- Wood Science & Technology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biotechnology (AREA)
- General Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Physics & Mathematics (AREA)
- General Health & Medical Sciences (AREA)
- Microbiology (AREA)
- Molecular Biology (AREA)
- Immunology (AREA)
- Biophysics (AREA)
- Analytical Chemistry (AREA)
- Biochemistry (AREA)
- Pathology (AREA)
- Biomedical Technology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Oncology (AREA)
- Hospice & Palliative Care (AREA)
- Plant Pathology (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Description
本発明は、大腸がんの診断方法および大腸がん患者に適用される補助化学療法の効果を評価する方法等に関する。 The present invention relates to a method for diagnosing colorectal cancer and a method for evaluating the effectiveness of adjuvant chemotherapy administered to colorectal cancer patients.
ステージIIIの大腸がん(CRC)の罹患率及び死亡率は、根治的手術後も全世界において依然として高止まりしている。補助化学療法(Adjuvant chemotherapy、ACT)は、ステージIIIのCRCにおける根治的手術後の臨床結果を改善するための標準的処置である。臨床結果を改善するためには、ACTの有効性を評価し、再発を検出するために、微小残存病変(minimal residual disease、MRD)の正確なモニタリングが必要となる。放射線学的手段、すなわち、コンピュータ断層撮影(CT)および磁気共鳴画像(MRI)による臨床的所見は、手術後の再発の確定診断として認定されている。しかし、癌胎児性抗原(carcinoembryonic antigen、CEA)および糖鎖抗原19-9(carbohydrate antigen、CA19-9)などの血清腫瘍マーカーを用いてMRDを追跡するための正確且つ容易に利用可能な方法が求められている。末梢血エクソソーム内のトランスクリプト(非特許文献1、2)やmicroRNA(miRNA)(非特許文献3~5)など、液体生検系における最も可能性がある適切なアプローチは現在も研究が続けられている。 Morbidity and mortality from stage III colorectal cancer (CRC) remain high worldwide even after curative surgery. Adjuvant chemotherapy (ACT) is the standard treatment to improve clinical outcomes after curative surgery in stage III CRC. To improve clinical outcomes, accurate monitoring of minimal residual disease (MRD) is required to evaluate the efficacy of ACT and detect recurrence. Clinical findings by radiological means, i.e., computed tomography (CT) and magnetic resonance imaging (MRI), are recognized as definitive diagnoses of recurrence after surgery. However, there is a need for accurate and easily accessible methods to track MRD using serum tumor markers such as carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9). The most promising and suitable approaches in liquid biopsy systems, such as transcripts in peripheral blood exosomes (Non-Patent Documents 1 and 2) and microRNAs (miRNAs) (Non-Patent Documents 3 to 5), are currently under investigation.
2017年の一般教書演説により、がんゲノム医学が立ち上げられた。腫瘍組織中の病原的標的に対処する適合性のある薬剤は、悪性腫瘍の臨床結果を改善できる可能性がある(非特許文献6~8)。特定のゲノム異常を示す患者に対する薬剤の適応を可能にする目的で、血漿検体を用いて循環腫瘍DNA(ctDNA)の遺伝的異常を検出するバイオマーカーとして液体生検が使用されたが、これにはコンパニオン診断(CDx)マーカーおよび包括的ゲノムプロファイリング(CGP)テスト(非特許文献9)が含まれる。非小細胞肺癌(NSCLC)患者は、CDxマーカーを用いて、表皮成長因子受容体(EGFR)のR858LまたはT790Mとの変異を検出するための液体生検を受け、この結果によって、GefitinibやOsimertinib(非特許文献10、11)など、使用されるべき最適な薬剤が決定される。CGP、Guardant360 CDx、およびFoundationOne(登録商標) Liquid CDxテストは、2020年8月に開始され、深刻に進行した症例の臨床治療において、現在の標準療法に続き適用された。 The 2017 State of the Union address launched cancer genomic medicine. Matched drugs addressing pathogenic targets in tumor tissues may improve clinical outcomes of malignant tumors (Non-Patent Documents 6-8). Liquid biopsies have been used as biomarkers to detect genetic abnormalities in circulating tumor DNA (ctDNA) using plasma samples, including companion diagnostic (CDx) markers and comprehensive genomic profiling (CGP) tests (Non-Patent Document 9), to enable drug adaptation to patients with specific genomic abnormalities. Patients with non-small cell lung cancer (NSCLC) undergo liquid biopsy to detect epidermal growth factor receptor (EGFR) mutations with R858L or T790M using CDx markers, and the results determine the optimal drug to be used, such as Gefitinib or Osimertinib (Non-Patent Documents 10, 11). The CGP, Guardant360 CDx, and FoundationOne® Liquid CDx tests were launched in August 2020 and were applied following the current standard of care in the clinical treatment of severely advanced cases.
リキッドバイオプシーは、高分解能コンピュータ断層撮影(CT)や磁気共鳴画像(MRI)などの従来の診断ツールよりも早く再発を診断する、MRDモニタリングにおいて最も効果的なアプローチである。再発の誤診を回避するためには、末梢血における、肉眼では発見できないが、がん特異的であって、且つ、ロバストな事象を正確に追跡する目的で、頻繁な術後血液検査が必要となる。オーダーメード化された血液CGPテストは、将来的にはMRDモニタリングの中心を担い、適切な薬物の迅速な選択を促すと考えられている。しかしながら、現状の次世代シーケンシング(NGS)に基づく頻繁なMRDモニタリングシステムはシーケンス費用が高額となる理由から現実的とは言えない。かかる背景から、大腸がんの根治手術後における再発の予測に関し、従来法と比較して安価且つ簡便にこの目的を達成できる新規技術の開発が強く求められている。 Liquid biopsy is the most effective approach for MRD monitoring, diagnosing recurrence earlier than conventional diagnostic tools such as high-resolution computed tomography (CT) and magnetic resonance imaging (MRI). To avoid misdiagnosis of recurrence, frequent postoperative blood tests are required to accurately track cancer-specific and robust events in peripheral blood that are not detectable by the naked eye. Customized blood CGP tests are expected to play a central role in MRD monitoring in the future and facilitate the rapid selection of appropriate drugs. However, the current frequent MRD monitoring system based on next-generation sequencing (NGS) is not realistic due to the high sequencing costs. In this context, there is a strong demand for the development of new technologies that can predict recurrence after curative surgery for colorectal cancer at a lower cost and more simply than conventional methods.
本発明は、従来法よりも簡便に大腸がんの発症や再発を予測することができる新規技術の提供を課題とする。 The objective of the present invention is to provide a new technology that can predict the onset and recurrence of colorectal cancer more easily than conventional methods.
本発明者らは、上記課題に対して鋭意検討した結果、被検体におけるFGD5遺伝子、GPC6遺伝子およびMSC遺伝子のメチル化状態が、大腸がん細胞の存在のマーカーとして極めて有用であることを見出し、かかる知見に基づいてさらに研究を進めることによって本発明を完成するに至った。
すなわち、本発明は以下の通りである。
As a result of intensive research into the above-mentioned problems, the inventors discovered that the methylation status of the FGD5 gene, GPC6 gene and MSC gene in a subject is extremely useful as a marker for the presence of colorectal cancer cells, and by conducting further research based on this finding, they have completed the present invention.
That is, the present invention is as follows.
[1]
被検体におけるFGD5遺伝子およびGPC6遺伝子の少なくとも1つのメチル化状態を決定する工程を含む、被検体における大腸がんを検出するための方法。
[2]
被検体におけるMSC遺伝子のメチル化状態をさらに決定する工程を含む、[1]記載の方法。
[3]
被検体が、大腸がんの根治手術を受けた被検体である、[1]または[2]記載の方法。
[4]
メチル化状態の決定が、被検体由来の液体試料に基づいて行われることを特徴とする、[1]~[3]のいずれか記載の方法。
[5]
液体試料が血液試料である、[4]記載の方法。
[6]
メチル化状態の決定をPCR法によって行なうことを特徴とする、[1]~[5]のいずれか記載の方法。
[7]
大腸がんの補助化学療法を受けている被検体におけるFGD5遺伝子およびGPC6遺伝子の少なくとも1つのメチル化状態を決定する工程を含む、被検体が受けている補助化学療法の効果を評価する方法。
[8]
被検体におけるMSC遺伝子のメチル化状態をさらに決定する工程を含む、[7]記載の方法。
[9]
被検体が、大腸がんの根治手術を受けた被検体である、[7]または[8]記載の方法。
[10]
メチル化状態の決定が、被検体由来の液体試料に基づいて行われることを特徴とする、[7]~[9]のいずれか記載の方法。
[11]
液体試料が血液試料である、[10]記載の方法。
[12]
メチル化状態の決定をPCR法によって行なうことを特徴とする、[7]~[10]のいずれか記載の方法。
[13]
以下を含む、大腸がんの診断用キット:
(1)FGD5遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対;および/または、
(2)GPC6遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対。
[14]
FGD5遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対が、SEQ ID NO:4に示される塩基配列を含む核酸と、SEQ ID NO:5に示される塩基配列を含む核酸であり、
GPC6遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対が、SEQ ID NO:6に示される塩基配列を含む核酸と、SEQ ID NO:7に示される塩基配列を含む核酸である、
[13]記載のキット。
[15]
(3)MSC遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対をさらに含む、[13]または[14]記載のキット。
[16]
MSC遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対が、SEQ ID NO:8に示される塩基配列を含む核酸と、SEQ ID NO:9に示される塩基配列を含む核酸である、[15]記載のキット。
[1]
A method for detecting colon cancer in a subject, comprising determining the methylation status of at least one of the FGD5 gene and the GPC6 gene in the subject.
[2]
The method described in [1], further comprising a step of determining the methylation status of the MSC gene in the subject.
[3]
The method according to [1] or [2], wherein the subject has undergone radical surgery for colorectal cancer.
[4]
The method according to any one of [1] to [3], wherein the determination of the methylation status is carried out based on a liquid sample derived from the subject.
[5]
The method according to [4], wherein the liquid sample is a blood sample.
[6]
The method according to any one of [1] to [5], wherein the methylation status is determined by a PCR method.
[7]
A method for evaluating the effect of adjuvant chemotherapy being received by a subject, comprising determining the methylation status of at least one of the FGD5 gene and the GPC6 gene in the subject receiving adjuvant chemotherapy for colorectal cancer.
[8]
The method described in [7], further comprising a step of determining the methylation status of the MSC gene in the subject.
[9]
The method according to [7] or [8], wherein the subject has undergone radical surgery for colorectal cancer.
[10]
The method according to any one of [7] to [9], wherein the determination of the methylation status is carried out based on a liquid sample derived from the subject.
[11]
The method according to [10], wherein the liquid sample is a blood sample.
[12]
The method according to any one of [7] to [10], wherein the methylation status is determined by a PCR method.
[13]
A diagnostic kit for colon cancer, including:
(1) a PCR primer pair capable of amplifying a part or the whole of the nucleotide sequence of the FGD5 gene; and/or
(2) A PCR primer pair capable of amplifying a part or the whole of the nucleotide sequence of the GPC6 gene.
[14]
A PCR primer pair capable of amplifying a part or the whole of the base sequence of the FGD5 gene is a nucleic acid having a base sequence shown in SEQ ID NO: 4 and a nucleic acid having a base sequence shown in SEQ ID NO: 5;
A PCR primer pair capable of amplifying a part or the whole of the base sequence of the GPC6 gene is a nucleic acid comprising the base sequence shown in SEQ ID NO: 6 and a nucleic acid comprising the base sequence shown in SEQ ID NO: 7.
[13] The kit described in.
[15]
(3) The kit according to [13] or [14], further comprising a PCR primer pair capable of amplifying a part or all of the base sequence of the MSC gene.
[16]
The kit according to [15], wherein the PCR primer pair capable of amplifying a part or all of the base sequence of the MSC gene is a nucleic acid having a base sequence shown in SEQ ID NO: 8 and a nucleic acid having a base sequence shown in SEQ ID NO: 9.
本発明によれば、被検体が大腸がんに罹患しているかどうかや、大腸がんの根治手術後の被検体において大腸がんが再発したかどうかを、従来の方法よりも早く決定することができる。さらに、本発明によれば、大腸がんの補助化学療法を受けている被検体において、当該補助化学療法が効果的であるかどうかを評価することもできる。 The present invention makes it possible to determine whether a subject is suffering from colorectal cancer and whether colorectal cancer has recurred in a subject after radical surgery for colorectal cancer more quickly than with conventional methods. Furthermore, the present invention also makes it possible to evaluate whether adjuvant chemotherapy for colorectal cancer is effective in a subject receiving the chemotherapy.
以下、本発明を詳細に説明する。 The present invention is explained in detail below.
1.被検体における大腸がんを検出するための方法
本発明は、被検体におけるFGD5遺伝子およびGPC6遺伝子の少なくとも1つのメチル化状態を決定する工程を含む、被検体における大腸がんを検出するための方法(以下、「本発明の検出方法」と称することがある)を提供する。
1. Method for detecting colorectal cancer in a subject The present invention provides a method for detecting colorectal cancer in a subject (hereinafter, sometimes referred to as the "detection method of the present invention"), comprising a step of determining the methylation status of at least one of the FGD5 gene and the GPC6 gene in the subject.
本発明の検出方法において、FGD5遺伝子およびGPC6遺伝子の少なくとも1つのメチル化状態が決定される。好ましい一態様において、本発明の検出方法においては、さらにMSC遺伝子のメチル化状態が決定される。特に好ましい一態様において、本発明の検出方法においては、FGD5遺伝子、GPC6遺伝子およびMSC遺伝子のメチル化状態が決定される。 In the detection method of the present invention, the methylation status of at least one of the FGD5 gene and the GPC6 gene is determined. In a preferred embodiment, the detection method of the present invention further determines the methylation status of the MSC gene. In a particularly preferred embodiment, the detection method of the present invention determines the methylation status of the FGD5 gene, the GPC6 gene, and the MSC gene.
FGD5遺伝子(FYVE, RhoGEF and PH domain containing 5、Gene ID:152273)は、「ZFYVE23」等とも称され、ヒトにおいては8q13.3の染色体位置に存在し、2個のエキソンを有する遺伝子である。FGD5遺伝子は、ヒト正常個体においては脾臓や肺において高発現することが知られている。ヒトにおけるFGD5遺伝子の塩基配列を配列番号1に示す。 The FGD5 gene (FYVE, RhoGEF and PH domain containing 5, Gene ID: 152273), also known as "ZFYVE23", is located at chromosomal position 8q13.3 in humans and is a gene with two exons. The FGD5 gene is known to be highly expressed in the spleen and lungs in normal human individuals. The base sequence of the human FGD5 gene is shown in SEQ ID NO:1.
GPC6遺伝子(glypican 6、Gene ID:10082)は、「OMIMD1」等とも称され、ヒトにおいては13q31.3-q32.1の染色体位置に存在し、12個のエキソンを有する遺伝子である。GPC6遺伝子は、ヒト正常個体においては胆嚢や膀胱において高発現することが知られている。ヒトにおけるGPC6遺伝子の塩基配列を配列番号2に示す。 The GPC6 gene (glypican 6, Gene ID: 10082), also known as "OMIMD1", is located at chromosomal location 13q31.3-q32.1 in humans and is a gene with 12 exons. The GPC6 gene is known to be highly expressed in the gallbladder and urinary bladder in normal human individuals. The base sequence of the human GPC6 gene is shown in SEQ ID NO: 2.
MSC遺伝子(musculin、Gene ID:9242)は、「ABF1」、「MYOR」、「ABF-1」、「bHLHa22」等とも称され、ヒトにおいては8q13.3の染色体位置に存在し、2個のエキソンを有する遺伝子であるMSC遺伝子は、ヒト正常個体においては胎盤や胆嚢において高発現することが知られている。ヒトにおけるMSC遺伝子の塩基配列を配列番号3に示す。 The MSC gene (musculin, Gene ID: 9242), also known as "ABF1," "MYOR," "ABF-1," "bHLHa22," etc., is located at chromosomal position 8q13.3 in humans, and the MSC gene, which is a gene with two exons, is known to be highly expressed in the placenta and gallbladder in normal humans. The base sequence of the human MSC gene is shown in SEQ ID NO: 3.
本発明の検出方法においては、上述した遺伝子のメチル化状態が決定されるが、遺伝子のメチル化状態を決定する方法は当業者にとって自体公知であり、いずれの方法を用いてもよい。尚、本発明の検出方法において、「遺伝子Aのメチル化状態を決定する」とは「遺伝子Aにおけるメチル化の量を決定すること」であってもよい。遺伝子のメチル化の状態は、例えば、メチルDNA免疫沈降(MeDIP)、メチル化感受性制限酵素(MSRE)分析、バイサルファイトシーケンシング等により検出することができ、また、その量を決定することができる。本発明の検出方法の好ましい一態様において、遺伝子のメチル化状態の決定は、メチル化感受性制限酵素分析とPCR法を組み合わせて用いることで実施され得る。尚、メチル化感受性制限酵素分析と組み合わせるPCR法は、リアルタイムPCRまたはデジタルPCRがより好ましい。尚、メチル化感受性制限酵素分析とPCR法とを組み合わせたメチル化状態の決定の詳細は、本明細書の実施例に記載されている。 In the detection method of the present invention, the methylation state of the above-mentioned gene is determined, but the method of determining the methylation state of a gene is known to those skilled in the art, and any method may be used. In the detection method of the present invention, "determining the methylation state of gene A" may also mean "determining the amount of methylation in gene A". The methylation state of a gene can be detected, and the amount can be determined, for example, by methyl-DNA immunoprecipitation (MeDIP), methylation-sensitive restriction enzyme (MSRE) analysis, bisulfite sequencing, etc. In a preferred embodiment of the detection method of the present invention, the methylation state of a gene can be determined by combining methylation-sensitive restriction enzyme analysis with a PCR method. The PCR method combined with methylation-sensitive restriction enzyme analysis is more preferably real-time PCR or digital PCR. Details of the determination of the methylation state by combining methylation-sensitive restriction enzyme analysis with a PCR method are described in the examples of this specification.
本明細書において「遺伝子Aのメチル化状態を決定する」という用語は、遺伝子Aの全長または一部においてメチル化された領域の有無を定性的に分析することを意味する。メチル化状態を決定する領域のヌクレオチド長は特に限定されないが、一態様において、PCR増幅に適した長さを設定することが好ましい。メチル化状態を決定する領域のヌクレオチド長としては、通常30~1000、好ましくは50~300、より好ましくは75~200、特に好ましくは75~150のヌクレオチド長であり得るが、これらに限定されない。尚、当該領域がプライマーを使用して増幅される場合、このヌクレオチド長はプライマー結合部位を含むものである。 In this specification, the term "determining the methylation status of gene A" means qualitatively analyzing the presence or absence of a methylated region in the entire length or a part of gene A. The nucleotide length of the region for determining the methylation status is not particularly limited, but in one embodiment, it is preferable to set a length suitable for PCR amplification. The nucleotide length of the region for determining the methylation status may be, but is not limited to, usually 30 to 1000, preferably 50 to 300, more preferably 75 to 200, and particularly preferably 75 to 150 nucleotides. Note that when the region is amplified using a primer, this nucleotide length includes the primer binding site.
本発明の検出方法において、メチル化状態の決定がデジタルPCR等のPCR法を伴う方法によって達成される場合、当該PCR反応において用い得るプライマー対は、FGD5遺伝子、GPC6遺伝子およびMSC遺伝子のメチル化され得る領域の一部または全部を増幅することができるプライマー対を自体公知の方法を用いて適宜設計すればよい。本発明の検出方法の好ましい一態様において、かかるプライマー対は、以下のものを用いることができるが、これらに限定されない。 In the detection method of the present invention, when the determination of the methylation state is achieved by a method involving a PCR method such as digital PCR, the primer pair that can be used in the PCR reaction can be appropriately designed using a method known per se to amplify part or all of the regions that can be methylated in the FGD5 gene, the GPC6 gene, and the MSC gene. In a preferred embodiment of the detection method of the present invention, the following primer pairs can be used, but are not limited to these.
FGD5遺伝子用のプライマー対:SEQ ID NO:4で示される塩基配列を含む、またはそれからなる核酸およびSEQ ID NO:5で示される塩基配列を含む、またはそれからなる核酸。
GPC6遺伝子用のプライマー対:SEQ ID NO:6で示される塩基配列を含む、またはそれからなる核酸、およびSEQ ID NO:7で示される塩基配列を含む、またはそれからなる核酸。
MSC遺伝子用のプライマー対:SEQ ID NO:8で示される塩基配列を含む、またはそれからなる核酸、およびSEQ ID NO:9で示される塩基配列を含む、またはそれからなる核酸。
Primer pair for FGD5 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:4 and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:5.
Primer pair for GPC6 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:6, and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:7.
Primer pair for MSC gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:8, and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:9.
また、一態様において、メチル化状態の決定のために、FGD5遺伝子、GPC6遺伝子、MSC遺伝子のターゲット領域の増幅を確認するためのプローブを合わせて用いることができる。かかる増幅を確認するためのプローブは、当業者であれば、増幅されるターゲット領域の核酸配列を考慮して適宜設計することができる。本発明の検出方法の一態様において、各遺伝子の増幅を確認するためのプローブは以下のものを用いることができるが、これらに限定されない。 In one embodiment, probes for confirming amplification of the target regions of the FGD5 gene, GPC6 gene, and MSC gene can be used in combination to determine the methylation status. A person skilled in the art can appropriately design such probes for confirming amplification, taking into consideration the nucleic acid sequence of the target region to be amplified. In one embodiment of the detection method of the present invention, the following probes can be used for confirming amplification of each gene, but are not limited to these.
FGD5遺伝子のターゲット領域の増幅の確認用のプローブ:SEQ ID NO:10で示される塩基配列を含む、またはそれからなる核酸。
GPC6遺伝子のターゲット領域の増幅の確認用のプローブ:SEQ ID NO:11で示される塩基配列を含む、またはそれからなる核酸。
MSC遺伝子のターゲット領域の増幅の確認用のプローブ:SEQ ID NO:12で示される塩基配列を含む、またはそれからなる核酸。
Probe for confirming amplification of a target region of the FGD5 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:10.
Probe for confirming amplification of a target region of the GPC6 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:11.
Probe for confirming amplification of a target region of the MSC gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:12.
被検体におけるFGD5遺伝子、GPC6遺伝子および/またはMSC遺伝子が正常対照と比較して高いメチル化状態を有する場合、被検体は大腸がんを有する可能性が高いと決定される。また、被検体が以前に大腸がんを罹患していた場合は、当該被検体におけるFGD5遺伝子、GPC6遺伝子および/またはMSC遺伝子が正常対照と比較して高いメチル化状態を有する場合、当該被検体において大腸がんが再発した可能性が高いと決定される。本発明の一実施形態において、被検体におけるFGD5遺伝子、GPC6遺伝子および/またはMSC遺伝子のメチル化状態をスコア化し、当該スコアを正常対象におけるメチル化状態のスコアと比較することもできる。尚、メチル化状態のスコア化の方法は自体公知の方法を用いればよい。被検体におけるFGD5遺伝子、GPC6遺伝子および/またはMSC遺伝子のメチル化状態のスコアが正常対照のそれよりも(統計学的に有意に)高い場合、被検体は大腸がんを有する可能性が高いと決定される。また、被検体が以前に大腸がんを罹患していた場合は、当該被検体におけるFGD5遺伝子、GPC6遺伝子および/またはMSC遺伝子のメチル化状態のスコアが正常対照のそれよりも(統計学的に有意に)高い場合、当該被検体において大腸がんが再発した可能性が高いと決定される。或いは、予めメチル化状態のスコアに関してのカットオフ値を設定し、当該カットオフ値よりも被検体のスコアが高い場合、被検体は大腸がんを有する可能性が高い、或いは当該被検体において大腸がんが再発した可能性が高いと決定することもできる。 If the FGD5 gene, GPC6 gene and/or MSC gene in the subject has a high methylation state compared to the normal control, the subject is determined to have a high possibility of having colorectal cancer. In addition, if the subject has previously had colorectal cancer, if the FGD5 gene, GPC6 gene and/or MSC gene in the subject has a high methylation state compared to the normal control, the subject is determined to have a high possibility of having recurrent colorectal cancer. In one embodiment of the present invention, the methylation state of the FGD5 gene, GPC6 gene and/or MSC gene in the subject can be scored and the score can be compared with the score of the methylation state in a normal subject. Note that the method for scoring the methylation state may be a method known per se. If the score of the methylation state of the FGD5 gene, GPC6 gene and/or MSC gene in the subject is (statistically significantly) higher than that of the normal control, the subject is determined to have a high possibility of having colorectal cancer. In addition, if the subject previously had colorectal cancer, if the methylation status score of the FGD5 gene, GPC6 gene, and/or MSC gene in the subject is (statistically significantly) higher than that of the normal control, it is determined that the subject is likely to have colorectal cancer recurrence. Alternatively, a cutoff value for the methylation status score can be set in advance, and if the subject's score is higher than the cutoff value, it can be determined that the subject is likely to have colorectal cancer or that the subject is likely to have colorectal cancer recurrence.
本発明の検出方法における「被検体」は、大腸がんを罹患する哺乳動物であれば特に限定されない。被検体には、例えば、家畜動物(ウシ、ヒツジ、ネコ、イヌおよびウマ等)、実験動物(ウサギ、マウスおよびラット等)、霊長類(ヒト及び非ヒト霊長類等)が含まれるが、これらに限定されない。好ましい一態様において、被検体はヒトである。尚、本明細書において「被検体」との用語は、「個体」、「患者」、「対象」等とも言い換えられる。 The "subject" in the detection method of the present invention is not particularly limited as long as it is a mammalian animal suffering from colorectal cancer. Examples of subjects include, but are not limited to, livestock animals (such as cows, sheep, cats, dogs, and horses), laboratory animals (such as rabbits, mice, and rats), and primates (such as humans and non-human primates). In a preferred embodiment, the subject is a human. In this specification, the term "subject" can also be referred to as "individual," "patient," "subject," etc.
本発明の検出方法の一態様において、被検体は、大腸がんの根治手術を受けた被検体であり得る。理論に拘束されることを望むものではないが、本発明において着目する遺伝子(即ち、FGD5遺伝子、GPC6遺伝子および/またはMSC遺伝子)のメチル化状態は、大腸がんの根治手術を受けた被検体におけるその再発を早期に検出することができると考えられる。 In one embodiment of the detection method of the present invention, the subject may be a subject who has undergone curative surgery for colorectal cancer. Without wishing to be bound by theory, it is believed that the methylation state of the genes of interest in the present invention (i.e., the FGD5 gene, the GPC6 gene, and/or the MSC gene) can enable early detection of recurrence of colorectal cancer in subjects who have undergone curative surgery.
本発明の検出方法において、「大腸がん」には、腺がん、扁平上皮がん、腺扁平上皮がんなどのあらゆる種類の大腸がんが含まれる。尚、本発明における「大腸がん」には、結腸がんや直腸がんも含まれ得る。 In the detection method of the present invention, "colon cancer" includes all types of colon cancer, such as adenocarcinoma, squamous cell carcinoma, and adenosquamous cell carcinoma. Note that "colon cancer" in the present invention can also include colon cancer and rectal cancer.
本発明の検出方法において、本発明において着目する遺伝子のメチル化状態の決定は、メチル化状態が決定できる限りどのような手段において行ってもよいが、侵襲性等の観点から、被検体由来の液体試料に基づいて行われることが好ましい。被検体由来の液体試料としては、その液体試料を分析することで本発明において着目する遺伝子のメチル化状態を決定し得るものであれば特に限定されない。被検体由来の液体試料としては、例えば、血液、リンパ液、組織液または体腔液などが挙げられるが、好ましくは血液であり、より好ましくは血漿である。血漿中に含まれるcfDNA(cell-free DNA)およびctDNA(circulating tumor DNA)を利用してFDG5遺伝子、GPC6遺伝子および/またはMSC遺伝子のメチル化状態を決定することにより、簡便、低コスト、低侵襲的に、被検体の大腸がんの早期診断が可能となる。 In the detection method of the present invention, the determination of the methylation state of the gene of interest in the present invention may be performed by any means as long as the methylation state can be determined, but from the viewpoint of invasiveness, it is preferable to perform the determination based on a liquid sample derived from a subject. The liquid sample derived from the subject is not particularly limited as long as the methylation state of the gene of interest in the present invention can be determined by analyzing the liquid sample. Examples of the liquid sample derived from the subject include blood, lymph, tissue fluid, and body cavity fluid, and preferably blood, and more preferably plasma. By determining the methylation state of the FDG5 gene, GPC6 gene, and/or MSC gene using cfDNA (cell-free DNA) and ctDNA (circulating tumor DNA) contained in plasma, early diagnosis of colon cancer in the subject can be made simply, at low cost, and in a minimally invasive manner.
2.補助化学療法の効果を評価する方法
本発明はまた、大腸がんの補助化学療法を受けている被検体におけるFDG5遺伝子およびGPC6遺伝子の少なくとも1つのメチル化状態を決定する工程を含む、被検体が受けている補助化学療法の効果を評価する方法(以下、「本発明の評価方法」と称することがある)を提供する。
2. Method for Evaluating the Effect of Adjuvant Chemotherapy The present invention also provides a method for evaluating the effect of adjuvant chemotherapy received by a subject, comprising a step of determining the methylation state of at least one of the FDG5 gene and the GPC6 gene in the subject receiving adjuvant chemotherapy for colorectal cancer (hereinafter, sometimes referred to as the "evaluation method of the present invention").
本発明の評価方法において、FGD5遺伝子およびGPC6遺伝子の少なくとも1つのメチル化状態が決定される。好ましい一態様において、本発明の評価方法においては、さらにMSC遺伝子のメチル化状態が決定される。特に好ましい一態様において、本発明の評価方法においては、FGD5遺伝子、GPC6遺伝子およびMSC遺伝子のメチル化状態が決定される。 In the evaluation method of the present invention, the methylation status of at least one of the FGD5 gene and the GPC6 gene is determined. In a preferred embodiment, the evaluation method of the present invention further determines the methylation status of the MSC gene. In a particularly preferred embodiment, the evaluation method of the present invention determines the methylation status of the FGD5 gene, the GPC6 gene, and the MSC gene.
本発明の評価方法における、「FDG5遺伝子」、「GPC6遺伝子」、「MSC遺伝子」、「被検体」、「大腸がん」、「遺伝子のメチル化状態の決定」、「被検体由来の試料」等は、上述した本発明の検出方法において説明したものと同様である。 In the evaluation method of the present invention, the "FDG5 gene," "GPC6 gene," "MSC gene," "subject," "colon cancer," "determination of gene methylation status," "sample derived from subject," etc. are the same as those described in the detection method of the present invention above.
本発明の評価方法において、大腸がんの補助化学療法を受けている被検体におけるFGD5遺伝子、GPC6遺伝子および/またはMSC遺伝子が正常対照と比較して高いメチル化状態を有する場合、被検体は大腸がんを有する可能性が高いと決定され、従って、当該被検体が現在受けている補助化学療法の効果が低いと評価される。反対に、大腸がんの補助化学療法を受けている被検体におけるFGD5遺伝子、GPC6遺伝子および/またはMSC遺伝子が、正常対照と比較して同等程度のメチル化状態を有する場合、当該被検体は大腸がんを有さない可能性が高いと決定され、従って、当該被検体が現在受けている補助化学療法の効果が高いと評価される。また、被検体が以前に大腸がんを罹患していた場合は、当該被検体におけるFGD5遺伝子、GPC6遺伝子および/またはMSC遺伝子が正常対照と比較して高いメチル化状態を有する場合、当該被検体は大腸がんを再発した可能性が高いと決定され、従って、当該被検体が現在受けている補助化学療法の効果が低いと評価される。反対に、当該被検体におけるFGD5遺伝子、GPC6遺伝子および/またはMSC遺伝子が、正常対照と比較して同等程度のメチル化状態を有する場合、当該被検体は大腸がんを再発していない可能性が高いと決定され、従って、当該被検体が現在受けている補助化学療法の効果が高いと評価される。また、上述の本発明の検出方法において説明した通り、メチル化状態をスコア化し、これを用いて評価を行うこともできる。 In the evaluation method of the present invention, if the FGD5 gene, GPC6 gene and/or MSC gene in a subject undergoing adjuvant chemotherapy for colorectal cancer have a high methylation state compared to a normal control, the subject is determined to have a high possibility of having colorectal cancer, and therefore, the effect of the adjuvant chemotherapy currently being received by the subject is evaluated to be low. Conversely, if the FGD5 gene, GPC6 gene and/or MSC gene in a subject undergoing adjuvant chemotherapy for colorectal cancer have a similar degree of methylation state compared to a normal control, the subject is determined to have a high possibility of not having colorectal cancer, and therefore, the effect of the adjuvant chemotherapy currently being received by the subject is evaluated to be high. In addition, if a subject has previously suffered from colorectal cancer, if the FGD5 gene, GPC6 gene and/or MSC gene in the subject have a high methylation state compared to a normal control, the subject is determined to have a high possibility of having recurrence of colorectal cancer, and therefore, the effect of the adjuvant chemotherapy currently being received by the subject is evaluated to be low. Conversely, if the FGD5 gene, GPC6 gene and/or MSC gene in the subject have a methylation state comparable to that of the normal control, it is determined that the subject is highly likely to have no recurrence of colorectal cancer, and therefore the adjuvant chemotherapy currently being received by the subject is evaluated as being highly effective. Also, as explained in the detection method of the present invention described above, the methylation state can be scored and used for evaluation.
3.大腸がんの診断用キット
本発明はまた、以下を含む、大腸がんの診断用キット:
(1)FDG5遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対;および/または、
(2)GPC6遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対(以下、「本発明のキット」と称することがある)を提供する。
3. Colon cancer diagnostic kit The present invention also relates to a colon cancer diagnostic kit comprising:
(1) a PCR primer pair capable of amplifying a part or the whole of the base sequence of the FDG5 gene; and/or
(2) To provide a PCR primer pair capable of amplifying a part or the whole of the base sequence of the GPC6 gene (hereinafter, sometimes referred to as the "kit of the present invention").
本発明のキットは、FGD5遺伝子およびGPC6遺伝子の少なくとも1つの塩基配列の一部または全部を増幅することができるPCRプライマー対を含む。好ましい一態様において、本発明のキットは、MSC遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対をさらに含む。特に好ましい一態様において、本発明のキットは、FGD5遺伝子、GPC6遺伝子およびMSC遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対を含む。 The kit of the present invention includes a PCR primer pair capable of amplifying a part or all of the base sequence of at least one of the FGD5 gene and the GPC6 gene. In a preferred embodiment, the kit of the present invention further includes a PCR primer pair capable of amplifying a part or all of the base sequence of the MSC gene. In a particularly preferred embodiment, the kit of the present invention includes a PCR primer pair capable of amplifying a part or all of the base sequence of the FGD5 gene, the GPC6 gene, and the MSC gene.
FDG5遺伝子、GPC6遺伝子およびMSC遺伝子を増幅するためのプライマー対の配列は、これらの遺伝子の一部または全部を増幅できるものであり、且つ、これらの遺伝子のメチル化状態を決定できる領域を増幅できるものである限り特に限定されない。また、各プライマーのヌクレオチド長についても特に限定はないが、通常16~40、好ましくは17~30、より好ましくは18~25であり得る。 The sequences of the primer pairs for amplifying the FDG5 gene, the GPC6 gene, and the MSC gene are not particularly limited, so long as they can amplify part or all of these genes and can amplify the regions in these genes that can determine the methylation status. There are also no particular limitations on the nucleotide length of each primer, but it can usually be 16 to 40, preferably 17 to 30, and more preferably 18 to 25.
本発明のキットの好ましい一態様において、かかるプライマー対は、以下のものを用いることができるが、これらに限定されない。 In a preferred embodiment of the kit of the present invention, the primer pair may be, but is not limited to, the following:
FGD5遺伝子用のプライマー対:SEQ ID NO:4で示される塩基配列を含む、またはそれからなる核酸およびSEQ ID NO:5で示される塩基配列を含む、またはそれからなる核酸。
GPC6遺伝子用のプライマー対:SEQ ID NO:6で示される塩基配列を含む、またはそれからなる核酸、およびSEQ ID NO:7で示される塩基配列を含む、またはそれからなる核酸。
MSC遺伝子用のプライマー対:SEQ ID NO:8で示される塩基配列を含む、またはそれからなる核酸、およびSEQ ID NO:9で示される塩基配列を含む、またはそれからなる核酸。
Primer pair for FGD5 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:4 and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:5.
Primer pair for GPC6 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:6, and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:7.
Primer pair for MSC gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:8, and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:9.
また、一態様において、本発明のキットは、プライマー対のみならず、FGD5遺伝子、GPC6遺伝子、MSC遺伝子のターゲット領域の増幅を確認するためのプローブを含めることもできる。かかる増幅を確認するためのプローブは、当業者であれば、増幅されるターゲット領域の核酸配列を考慮して適宜設計することができる。本発明のキットの一態様において、各遺伝子の増幅を確認するためのプローブとして以下のものをキットに含めることができるが、これらに限定されない。 In one embodiment, the kit of the present invention can include not only primer pairs but also probes for confirming amplification of the target regions of the FGD5 gene, the GPC6 gene, and the MSC gene. A person skilled in the art can appropriately design such probes for confirming amplification, taking into consideration the nucleic acid sequence of the target region to be amplified. In one embodiment of the kit of the present invention, the following probes for confirming amplification of each gene can be included in the kit, but are not limited to these.
FGD5遺伝子のターゲット領域の増幅の確認用のプローブ:SEQ ID NO:10で示される塩基配列を含む、またはそれからなる核酸。
GPC6遺伝子のターゲット領域の増幅の確認用のプローブ:SEQ ID NO:11で示される塩基配列を含む、またはそれからなる核酸。
MSC遺伝子のターゲット領域の増幅の確認用のプローブ:SEQ ID NO:12で示される塩基配列を含む、またはそれからなる核酸。
Probe for confirming amplification of a target region of the FGD5 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:10.
Probe for confirming amplification of a target region of the GPC6 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:11.
Probe for confirming amplification of a target region of the MSC gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:12.
本発明のキットには、上述したプライマー対やプローブのほかに試薬や陽性/陰性対照、説明書などを含めることができる。 The kit of the present invention may include, in addition to the primer pairs and probes described above, reagents, positive/negative controls, instructions, etc.
4.インビトロ/エクスビボでの大腸がんの診断または大腸がんの補助化学療法の効果の評価におけるFGD5遺伝子およびGPC6遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対の使用
本発明はまた、インビトロ/エクスビボでの大腸がんの診断または大腸がんの補助化学療法の効果の評価における、
(1)FDG5遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対;および/または、
(2)GPC6遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対
の使用(以下、「本発明の使用」と称することがある)を提供する。
4. Use of a PCR primer pair capable of amplifying a part or the whole of the base sequence of the FGD5 gene and the GPC6 gene in in vitro/ex vivo diagnosis of colon cancer or evaluation of the effect of adjuvant chemotherapy for colon cancer The present invention also relates to the use of a PCR primer pair capable of amplifying a part or the whole of the base sequence of the FGD5 gene and the GPC6 gene in in vitro/ex vivo diagnosis of colon cancer or evaluation of the effect of adjuvant chemotherapy for colon cancer.
(1) a PCR primer pair capable of amplifying a part or the whole of the base sequence of the FDG5 gene; and/or
(2) The present invention provides a use of a PCR primer pair capable of amplifying a part or the whole of the base sequence of the GPC6 gene (hereinafter, sometimes referred to as "use of the present invention").
本発明の使用においては、FGD5遺伝子およびGPC6遺伝子の少なくとも1つの塩基配列の一部または全部を増幅することができるPCRプライマー対が、インビトロ/エクスビボでの大腸がんの診断に用いられる。好ましい一態様において、本発明の使用においては、MSC遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対が更に用いられる。特に好ましい一態様において、本発明の使用においては、FGD5遺伝子、GPC6遺伝子およびMSC遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対が用いられる。 In the use of the present invention, a PCR primer pair capable of amplifying part or all of the base sequence of at least one of the FGD5 gene and the GPC6 gene is used for in vitro/ex vivo diagnosis of colorectal cancer. In a preferred embodiment, in the use of the present invention, a PCR primer pair capable of amplifying part or all of the base sequence of the MSC gene is further used. In a particularly preferred embodiment, in the use of the present invention, a PCR primer pair capable of amplifying part or all of the base sequence of the FGD5 gene, the GPC6 gene, and the MSC gene is used.
FDG5遺伝子、GPC6遺伝子およびMSC遺伝子を増幅するためのプライマー対の配列は、これらの遺伝子の一部または全部を増幅できるものであり、且つ、これらの遺伝子のメチル化状態を決定できる領域を増幅できるものである限り特に限定されない。また、各プライマーのヌクレオチド長についても特に限定はないが、通常16~40、好ましくは17~30、より好ましくは18~25であり得る。 The sequences of the primer pairs for amplifying the FDG5 gene, the GPC6 gene, and the MSC gene are not particularly limited, so long as they can amplify part or all of these genes and can amplify the regions in these genes that can determine the methylation status. There are also no particular limitations on the nucleotide length of each primer, but it can usually be 16 to 40, preferably 17 to 30, and more preferably 18 to 25.
本発明の使用の好ましい一態様において、かかるプライマー対は、以下のものを用いることができるが、これらに限定されない。 In a preferred embodiment of the present invention, the primer pair may be, but is not limited to, the following:
FGD5遺伝子用のプライマー対:SEQ ID NO:4で示される塩基配列を含む、またはそれからなる核酸およびSEQ ID NO:5で示される塩基配列を含む、またはそれからなる核酸。
GPC6遺伝子用のプライマー対:SEQ ID NO:6で示される塩基配列を含む、またはそれからなる核酸、およびSEQ ID NO:7で示される塩基配列を含む、またはそれからなる核酸。
MSC遺伝子用のプライマー対:SEQ ID NO:8で示される塩基配列を含む、またはそれからなる核酸、およびSEQ ID NO:9で示される塩基配列を含む、またはそれからなる核酸。
Primer pair for FGD5 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:4 and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:5.
Primer pair for GPC6 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:6, and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:7.
Primer pair for MSC gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:8, and a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:9.
また、本発明の使用においては、プライマー対のみならず、FGD5遺伝子、GPC6遺伝子、MSC遺伝子のターゲット領域の増幅を確認するためのプローブを更に用いることもできる。かかる増幅を確認するためのプローブは、当業者であれば、増幅されるターゲット領域の核酸配列を考慮して適宜設計することができる。一態様において、各遺伝子の増幅を確認するためのプローブとして以下のものを用いることができるが、これらに限定されない。 In addition, when using the present invention, not only primer pairs but also probes for confirming amplification of the target regions of the FGD5 gene, GPC6 gene, and MSC gene can be used. A person skilled in the art can appropriately design such probes for confirming amplification, taking into consideration the nucleic acid sequence of the target region to be amplified. In one embodiment, the following probes can be used for confirming amplification of each gene, but are not limited to these.
FGD5遺伝子のターゲット領域の増幅の確認用のプローブ:SEQ ID NO:10で示される塩基配列を含む、またはそれからなる核酸。
GPC6遺伝子のターゲット領域の増幅の確認用のプローブ:SEQ ID NO:11で示される塩基配列を含む、またはそれからなる核酸。
MSC遺伝子のターゲット領域の増幅の確認用のプローブ:SEQ ID NO:12で示される塩基配列を含む、またはそれからなる核酸。
Probe for confirming amplification of a target region of the FGD5 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:10.
Probe for confirming amplification of a target region of the GPC6 gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:11.
Probe for confirming amplification of a target region of the MSC gene: a nucleic acid comprising or consisting of the base sequence shown in SEQ ID NO:12.
本発明の使用において、被検体が大腸がんを有すると診断される基準や、大腸がんの補助化学療法の効果の評価に用いられる基準等は、「1.被検体における大腸がんを検出するための方法」や「2.補助化学療法の効果を評価する方法」で説明したものと同様である。 In using the present invention, the criteria for diagnosing a subject as having colorectal cancer and the criteria used to evaluate the effectiveness of adjuvant chemotherapy for colorectal cancer are the same as those explained in "1. Method for detecting colorectal cancer in a subject" and "2. Method for evaluating the effectiveness of adjuvant chemotherapy."
5.大腸がんの診断方法
本発明はまた、被検体におけるFGD5遺伝子およびGPC6遺伝子の少なくとも1つのメチル化状態を決定する工程を含む、被検体における大腸がんを診断するための方法(以下、「本発明の診断方法」と称することがある)を提供する。
5. Diagnosis Method for Colorectal Cancer The present invention also provides a method for diagnosing colorectal cancer in a subject, comprising a step of determining the methylation status of at least one of the FGD5 gene and the GPC6 gene in the subject (hereinafter, sometimes referred to as the "diagnostic method of the present invention").
本発明の診断方法における各遺伝子の配列やメチル化状態の決定方法等は、「1.被検体における大腸がんを検出するための方法」で説明したものと同様である。 The method for determining the sequence and methylation status of each gene in the diagnostic method of the present invention is the same as that described in "1. Method for detecting colorectal cancer in a subject."
一態様において、本発明の診断方法は、本発明の診断方法を用いることにより大腸がんを有すると診断された被検体に対して治療有効量の抗がん剤を投与する工程を含み得る。 In one embodiment, the diagnostic method of the present invention may include a step of administering a therapeutically effective amount of an anticancer agent to a subject diagnosed as having colorectal cancer by using the diagnostic method of the present invention.
被検体に対して投与される抗がん剤は特に限定されず、任意のものを用いることができる。抗がん剤としては、例えば、アルキル化剤(例、シクロホスファミド、シスプラチン等)、抗代謝剤(例、メトトレキサート、5-FU等)、微小管阻害剤(例、パクリタキセル、ドセタキセル等)、トポイソメラーゼ阻害剤(例、イリノテカン、エトポシド等)、分子標的剤(例、イマチニブ、トラスツズマブ等)、免疫チェックポイント阻害剤(例、ニボルマブ、ペムブロリズマブ等)、ホルモン療法薬(例、タモキシフェン、フルタミド等)、プロテアソーム阻害剤(例、ボルテゾミブ、カルフィルゾミブ等)、PARP阻害剤(例、オラパリブ、ルカパリブ等)が挙げられるが、これらに限定されない。 The anticancer drug administered to the subject is not particularly limited, and any drug can be used. Examples of anticancer drugs include, but are not limited to, alkylating agents (e.g., cyclophosphamide, cisplatin, etc.), antimetabolic agents (e.g., methotrexate, 5-FU, etc.), microtubule inhibitors (e.g., paclitaxel, docetaxel, etc.), topoisomerase inhibitors (e.g., irinotecan, etoposide, etc.), molecular targeted agents (e.g., imatinib, trastuzumab, etc.), immune checkpoint inhibitors (e.g., nivolumab, pembrolizumab, etc.), hormone therapy drugs (e.g., tamoxifen, flutamide, etc.), proteasome inhibitors (e.g., bortezomib, carfilzomib, etc.), and PARP inhibitors (e.g., olaparib, rucaparib, etc.).
以下の実施例において本発明を更に具体的に説明するが、本発明はこれらの例によってなんら限定されるものではない。 The present invention will be explained in more detail in the following examples, but the present invention is not limited to these examples.
本研究は、がん研究会有明病院の研究倫理審査委員会により承認された。IRB受領番号は2010-1058(2019年3月7日)。さらに、九州大学医系地区部局の臨床研究倫理審査委員会がプロジェクトを審査し、承認した(番号2020-321(2020年9月1日))。 This study was approved by the Research Ethics Committee of the Cancer Institute Ariake Hospital. The IRB receipt number is 2010-1058 (March 7, 2019). In addition, the Clinical Research Ethics Committee of the Kyushu University Medical Department reviewed and approved the project (number 2020-321 (September 1, 2020)).
[方法] [Method]
[臨床例]
全体で47名のステージIIIaまたはIIIbの大腸がん(n=30)および直腸がん(n=17)の患者が根治手術を受けた(表1)。全ての患者が、補助療法として標準的なカペシタビンとオキサリプラチン(6ヶ月で8コース)を受け、CAPOX治療を行った。28名の患者が術後170日から1070日の間に再発し、19名が、術後少なくとも1090日後の時点で非再発であった。再発した患者から、術前、術後、ACT中、ACT後、および再発時に合計180の血液サンプルを採取し、抽出した。非再発患者からは、合計で111のサンプルを分析のために採取した。
[Clinical cases]
A total of 47 patients with stage IIIa or IIIb colon cancer (n=30) and rectal cancer (n=17) underwent curative surgery (Table 1). All patients received standard capecitabine and oxaliplatin (8 courses over 6 months) as adjuvant therapy and CAPOX treatment. Twenty-eight patients relapsed between 170 and 1070 days after surgery, and 19 were non-relapsed at least 1090 days after surgery. A total of 180 blood samples were collected and extracted from relapsed patients before surgery, after surgery, during ACT, after ACT, and at the time of relapse. A total of 111 samples were collected from non-relapsed patients for analysis.
[サンプル保存とcfDNAの抽出]
患者由来の漏出性血漿サンプルは、4℃、2500×Gで10分間遠心分離した。上清液を2mLチューブに分注し、-80℃の冷凍庫で保存した。16,000×Gで10分間遠心分離後、上清液を分注した。King Fisher Duo Prime(Thermo Fisher Scientific)を使用し、2.5mLおよび30μLのMagmax細胞外DNA(cfDNA) Lysis/Binding溶液と磁性ビーズをプレートAの第1列に置き、すべての抽出サンプルを加えた。第2列は空で、第3列と第4列は1mLのMagmax cfDNA洗浄溶液で満たした。プレートBの第1列には50μLのMagmax cfDNA溶出溶液を入れ、第2列には500μLの80%エタノールを入れ、第3列には2mLの80%エタノールを入れた。cfDNA抽出プロトコルを用いて、30分後にcfDNAを抽出した。
[Sample storage and cfDNA extraction]
The patient-derived leaky plasma samples were centrifuged at 2500×G for 10 min at 4°C. The supernatant was dispensed into 2 mL tubes and stored in a −80°C freezer. After centrifugation at 16,000×G for 10 min, the supernatant was dispensed. Using a King Fisher Duo Prime (Thermo Fisher Scientific), 2.5 mL and 30 μL of Magmax extracellular DNA (cfDNA) Lysis/Binding solution and magnetic beads were placed in the first row of plate A and all the extracted samples were added. The second row was empty and the third and fourth rows were filled with 1 mL of Magmax cfDNA Wash Solution. Plate B contained 50 μL of Magmax cfDNA Elution Solution in row 1, 500 μL of 80% ethanol in row 2, and 2 mL of 80% ethanol in row 3. cfDNA was extracted after 30 minutes using the cfDNA extraction protocol.
[溶出されたcfDNAの制限酵素処理]
計15μLのcfDNAを対照として5μLの純水に溶解した。制限酵素処理のために、15μLのcfDNA、1μLのHapII、2μLの10×ローディングバッファー、および2μLの純水を加えた。対照および制限酵素処理液をサーマルサイクラーに配置し、37℃で1時間インキュベートし、制限酵素処理を完了した。
[Restriction enzyme treatment of eluted cfDNA]
A total of 15 μL of cfDNA was dissolved in 5 μL of pure water as a control. For restriction enzyme treatment, 15 μL of cfDNA, 1 μL of HapII, 2 μL of 10× loading buffer, and 2 μL of pure water were added. The control and restriction enzyme treatment solutions were placed in a thermal cycler and incubated at 37° C. for 1 hour to complete the restriction enzyme treatment.
[デジタルPCR用のサンプル調製]
二つの完成した溶液を用いてデジタルPCRのためのサンプルを調製した。まず、サンプルを混合するためのマスター溶液を調製した。FGD5、GPC6、およびMSCを対象とするプライマー(FおよびR)並びにプローブを用いた。合計6μLのRNase P、6μLのプローブ、6μLの各プライマー(F、R)、4μLの純水、および60μLのマスターミックスを混合した。完成した溶液の合計10.5μLを分注し、4μLの各サンプルの対照および制限酵素処理液を混合した。完成した溶液の14.5μLをデジタルPCRに供した。尚、プライマーの配列は以下の通りである:
[Sample preparation for digital PCR]
The two completed solutions were used to prepare samples for digital PCR. First, a master solution for mixing samples was prepared. Primers (F and R) and probes targeting FGD5, GPC6, and MSC were used. A total of 6 μL of RNase P, 6 μL of probe, 6 μL of each primer (F, R), 4 μL of pure water, and 60 μL of master mix were mixed. A total of 10.5 μL of the completed solution was dispensed, and 4 μL of each sample's control and restriction enzyme treatment solution were mixed. 14.5 μL of the completed solution was subjected to digital PCR. The primer sequences are as follows:
FGD5 primer f(SEQ ID NO:4)
5’-CCTTCTCAGCCTTGGCGAG
FGD5 primer r(SEQ ID NO:5)
5’-GCGTCTTTCTTCGTCGTGGAA
FGD5 primer f (SEQ ID NO: 4)
5'-CCTTCTCAGCCTTGGCGAG
FGD5 primer r (SEQ ID NO: 5)
5'-GCGTCTTTCTTCGTCGTGGAA
GPC6 primer f(SEQ ID NO:6)
5’-GCGGGCTTTCGGCTTGAG
GPC6 primer r(SEQ ID NO:7)
5’-CCAAGAGGGGAAGAATCACAGC
GPC6 primer f (SEQ ID NO: 6)
5'-GCGGGCTTTCGGCTTGAG
GPC6 primer r (SEQ ID NO: 7)
5'-CCAAGAGGGGAAGAATCACAGC
MSC primer f(SEQ ID NO:8)
5’-AGCAGTCGCAGCGGAACG
MSC primer r(SEQ ID NO:9)
5’-CAGGGCAGGCTGGTCTTGA
MSC primer f (SEQ ID NO:8)
5'-AGCAGTCGCAGCGGAACG
MSC primer r (SEQ ID NO:9)
5'-CAGGGCAGGCTGGTCTTGA
FGD5 probe(SEQ ID NO:10)
5’-CGCCAGTATCCCACTCGCACGGC
GPC6 probe(SEQ ID NO:11)
5’-CCGATCCAAGAAGGCATGGTGCAACATACA
MSC probe(SEQ ID NO:12)
5’-CCTGGAGAAGGCTTTGCTCAGCACGC
FGD5 probe (SEQ ID NO: 10)
5'-CGCCAGTATCCCACTCGCACGGC
GPC6 probe (SEQ ID NO: 11)
5'-CCGATCCAAGAAGGCATGGTGCAACATACA
MSC probe (SEQ ID NO: 12)
5'-CCTGGAGAAGGCTTTGCTCAGCACGC
[メチル化感受性制限酵素消化]
溶出したcfDNAを2つのチューブに分注した:一方を制限酵素消化用とし、他方を酵素無しの対照用とした。15μLのcfDNA、1μLのHapII(10units/μL)(Takara Bio)、1μLのLバッファー(10×)(Takara Bio)を含む反応混合物(20μL)を調製し、37℃で1時間インキュベートした。対照サンプルは15μLのcfDNAを水で20μLに希釈し、37℃で1時間インキュベートすることで調製した。
[Methylation-sensitive restriction enzyme digestion]
The eluted cfDNA was dispensed into two tubes: one for restriction enzyme digestion and the other for a no enzyme control. A reaction mixture (20 μL) containing 15 μL cfDNA, 1 μL HapII (10 units/μL) (Takara Bio), and 1 μL L buffer (10x) (Takara Bio) was prepared and incubated at 37° C. for 1 h. A control sample was prepared by diluting 15 μL cfDNA to 20 μL with water and incubating at 37° C. for 1 h.
[メチル化特異的デジタルPCR]
QuantStudio 3D Digital PCR System(Thermo Fisher Scientific)を用いてデジタルPCRを実施した。cfDNA 4μL、QuantStudio 3D Digital PCR Master Mix v2(ThermoFisher Scientific)7.25μL、フォワードプライマー(18μM)0.725μL、リバースプライマー(18μM)0.725μL、TaqMan probe(5μM)0.725μL、TaqMan Copy Number Reference Assay RNase P(ThermoFisher Scientific)0.725μLを含むデジタルPCR反応混合物(14.5μL)を調製した。デジタルPCR混合物は、QuantStudio 3D Digital PCR Chip Loaderを使用してQuantStudio 3D Digital PCR Chip v2に装填した。以下のプログラムを使用してProFlex 2xFlat PCR systemでPCRを実行した:96℃で10分、60℃で2分の39サイクル、98℃で30秒、60℃で2分、10℃。チップ画像はQuantStudio 3D Digital PCR Instrumentを使用してキャプチャし、QuantStudio 3D Analysis Suite Softwareを使用して解析した。
[Methylation-specific digital PCR]
Digital PCR was performed using the QuantStudio 3D Digital PCR System (Thermo Fisher Scientific). A digital PCR reaction mixture (14.5 μL) was prepared containing 4 μL cfDNA, 7.25 μL QuantStudio 3D Digital PCR Master Mix v2 (ThermoFisher Scientific), 0.725 μL forward primer (18 μM), 0.725 μL reverse primer (18 μM), 0.725 μL TaqMan probe (5 μM), and 0.725 μL TaqMan Copy Number Reference Assay RNase P (ThermoFisher Scientific). Digital PCR mixtures were loaded onto a QuantStudio 3D Digital PCR Chip v2 using the QuantStudio 3D Digital PCR Chip Loader. PCR was run on a ProFlex 2xFlat PCR system using the following program: 96°C for 10 min, 60°C for 2 min, 39 cycles of 98°C for 30 s, 60°C for 2 min, 10°C. Chip images were captured using the QuantStudio 3D Digital PCR Instrument and analyzed using the QuantStudio 3D Analysis Suite Software.
[メチル化比率の計算]
HapIIで消化したテストサンプルと未消化の対照サンプルを調製した。各サンプル中のターゲットおよびRNase Pのコピー数を測定し、メチル化比率を以下により計算した:
[Calculation of methylation ratio]
Test samples digested with HapII and undigested control samples were prepared. The copy numbers of target and RNase P in each sample were measured, and the methylation ratio was calculated as follows:
[メチル化感受性制限酵素に基づくメチル化定量のためのアッセイ設計]
メチル化感受性制限酵素を使用して、大腸がん特異的メチル化サイトのメチル化比率を測定するためのデジタルPCRアッセイを設計した。血漿からcfDNAを抽出し、メチル化感受性制限酵素であるHapIIで消化した。HapII認識サイトは、アンプリコンがメチル化されていない場合に切断される。したがって、cfDNAをHapIIで消化した後、認識サイトを挟むプライマーを使用してPCRを行うと、HapII認識サイトがメチル化されている場合にのみ増幅物が生ずる。cfDNAをHapIIで消化した後、認識サイトを挟むプライマーを使用してデジタルPCRを実行し、メチル化サイトのコピー数を測定した。同様の反応により、レファレンス遺伝子であるRNase Pのコピー数も測定した。全細胞がレファレンス遺伝子を含んでいることから、RNase Pのコピーは正常細胞と腫瘍細胞から放出される全cfDNAを表す。HapIIで消化したサンプル中の大腸がん特異的メチル化サイトのコピー数は、HapIIを含まないものと比較し、サイトのメチル化比率を計算した。ピペッティングバイアスがメチル化比率に大きな影響を及ぼす可能性があるため、各サンプルのターゲットサイトはRNase Pで除し、正確なメチル化比率を計算した(図1)。
[Assay design for methylation quantification based on methylation-sensitive restriction enzymes]
A digital PCR assay was designed to measure the methylation ratio of colon cancer-specific methylation sites using methylation-sensitive restriction enzymes. cfDNA was extracted from plasma and digested with HapII, a methylation-sensitive restriction enzyme. The HapII recognition site is cleaved when the amplicon is not methylated. Therefore, if cfDNA is digested with HapII and then PCR is performed using primers that flank the recognition site, amplification products will only occur if the HapII recognition site is methylated. After cfDNA is digested with HapII, digital PCR was performed using primers that flank the recognition site to measure the copy number of the methylation site. The copy number of the reference gene, RNase P, was also measured by a similar reaction. Since all cells contain the reference gene, the copies of RNase P represent the total cfDNA released from normal and tumor cells. The copy number of colon cancer-specific methylation sites in samples digested with HapII was compared to those without HapII, and the methylation ratio of the site was calculated. Because pipetting bias can have a significant effect on methylation ratios, the target site in each sample was divided by RNase P to calculate the exact methylation ratio (Figure 1).
[結果] [Results]
1.最適なメチル化サイトの同定
有用なメチル化サイトを選択するためにインシリコ分析を実施した。Illumina Infinium Human Methylation 450Kの473,864プローブを選択に使用し、大腸がん組織中のメチル化サイトの数(SN比>1.5)を100プローブに絞り込んだ。がんゲノムアトラス(TCGA)のデータを用いて、腫瘍組織と正常組織間のメチル化率の差が顕著であるが、正常組織と血液細胞でのメチル化率は低いものを選択することで、メチル化サイトの数を50プローブに絞り込んだ。尚、具体的な選択基準は次の通りである:
1. Identification of optimal methylation sites In silico analysis was performed to select useful methylation sites. 473,864 probes from Illumina Infinium Human Methylation 450K were used for selection, and the number of methylation sites (signal-to-noise ratio > 1.5) in colorectal cancer tissues was narrowed down to 100 probes. Using data from The Cancer Genome Atlas (TCGA), the number of methylation sites was narrowed down to 50 probes by selecting probes with a significant difference in methylation rate between tumor tissues and normal tissues, but low methylation rates in normal tissues and blood cells. The specific selection criteria are as follows:
1)ベータ値(腫瘍/正常組織)>0.4
2)ベータ値(正常血液)<0.153)ベータ値(正常組織)<0.2(図2参照)
1) Beta value (tumor/normal tissue) > 0.4
2) Beta value (normal blood) < 0.15 3) Beta value (normal tissue) < 0.2 (see Figure 2)
絞り込まれた50個のメチル化サイトのベータ値ローカスがユビキタスに高いことが確認され、また、ターゲットサイトのメチル化のベータ値は、mRNA発現と反比例していた。さらに、メチル化されたターゲットサイトのベータ値と臨床結果との逆相関を考慮し、選択肢を3つのプローブに絞り込んだ。FGD5のメチル化サイトのベータ値が増加した59人の患者は、FGD5のベータ値が減少した60人の患者よりも著しく予後が悪かった(p<0.05)。GPC6とMSCについても、同様に統計的に有意な結果が明らかにされた(図3)。結果として、FGD5、GPC6及びMSCの3つの遺伝子のターゲットメチル化サイトを選択し、末梢血中のcfDNAのさらなる分析に使用した。 The narrowed down 50 methylation site beta loci were confirmed to be ubiquitously high, and the methylation beta value of the target site was inversely proportional to mRNA expression. Furthermore, considering the inverse correlation between the beta value of the methylated target site and clinical outcome, the options were narrowed down to three probes. The 59 patients with increased beta value of the methylation site of FGD5 had a significantly worse prognosis than the 60 patients with decreased beta value of FGD5 (p<0.05). Statistically significant results were also revealed for GPC6 and MSC (Figure 3). As a result, the target methylation sites of the three genes FGD5, GPC6, and MSC were selected and used for further analysis of cfDNA in peripheral blood.
2.メチル化マーカーの感度と特異性
3つのターゲットサイトのメチル化状態を、大腸がん患者と大腸内視鏡検査で診断された患者を区別するために使用した。対照群は、2019年1月から2020年3月までの間に九州大学別府病院の外科で大腸内視鏡検査を受け、異常所見のない10人の患者からの血漿サンプルで構成した。腫瘍群は、がん研究会有明病院で収集された大腸がん(ステージ1~4;40人の患者、各群10人)の患者からの血漿を使用した。3つのマーカーのうち、FGD5とGPC6は腫瘍群でメチル化が有意に増加していた(それぞれp=0.028、p=0.0231;図4A)。MSCは、正常組織と比較して腫瘍組織でのメチル化値が高い傾向を示した(p=0.062)(図4A)。これらの結果は、本発明の3つのマーカーが、大腸がんの患者を識別できることを示しており、これらのマーカーががんのスクリーニングに適用し得るものであることを示唆している。これらのマーカーの受信者動作特性(ROC)曲線は、曲線下面積(AUC)が0.840と非常に高く(図4B)、感度65%、特異性100%と、現行の実用的な方法と比較して非常に高いことが示された。
2. Sensitivity and specificity of methylation markers The methylation status of the three target sites was used to distinguish patients with colorectal cancer from those diagnosed by colonoscopy. The control group consisted of plasma samples from 10 patients who underwent colonoscopy at the Department of Surgery, Kyushu University Beppu Hospital between January 2019 and March 2020 and had no abnormal findings. The tumor group used plasma from patients with colorectal cancer (stages 1-4; 40 patients, 10 in each group) collected at the Cancer Research Institute Ariake Hospital. Of the three markers, FGD5 and GPC6 had significantly increased methylation in the tumor group (p = 0.028, p = 0.0231, respectively; Figure 4A). MSCs showed a tendency to have higher methylation values in tumor tissues compared to normal tissues (p = 0.062) (Figure 4A). These results indicate that the three markers of the present invention can distinguish patients with colorectal cancer, suggesting that these markers may be applicable for cancer screening. The receiver operating characteristic (ROC) curve of these markers showed a very high area under the curve (AUC) of 0.840 (Figure 4B), with a sensitivity of 65% and a specificity of 100%, both of which are very high compared to current practical methods.
3.CTとAMUSEアッセイによるMRDモニタリングの感度の比較
病理学的ステージIII A/BおよびCurAの大腸がん(再発陽性28人、再発陰性19人)を持つ47人の患者が、2012年6月から2017年6月までの間にがん研究会有明病院で手術を受けた。患者は術後のACTにCAPOXが使用された。高分化-低分化腺癌の病理学的組織学的診断が本研究に含まれた。メチル化状態は、術前と術後、ACT後、再発時に採取された血漿サンプルを使用して測定した。再発がない患者については、術後平均1551日(1093-1897日)再発がない場合を再発がないケースと定義した。cfDNAの3つの遺伝子のメチル化値がカットオフ値を超える患者は、液体生検によって再発陽性と診断した。CT画像を使用した術後の再発の臨床診断前に、28人の患者のうち22人(78.5%)が再発と診断された(感度、78.5%)。
3. Comparison of the sensitivity of MRD monitoring by CT and AMUSE assay Forty-seven patients with pathological stage III A/B and CurA colon cancer (28 recurrence-positive, 19 recurrence-negative) underwent surgery at the Cancer Institute Ariake Hospital between June 2012 and June 2017. Patients were treated with CAPOX for postoperative ACT. Pathological histological diagnoses of well-differentiated-poorly differentiated adenocarcinoma were included in the study. Methylation status was measured using plasma samples collected preoperatively and postoperatively, after ACT, and at the time of recurrence. For patients without recurrence, cases without recurrence were defined as those without recurrence for a mean of 1551 days (1093-1897 days) after surgery. Patients with methylation values of the three genes in cfDNA above the cutoff value were diagnosed as positive for recurrence by liquid biopsy. Twenty-two of 28 patients (78.5%) were diagnosed with recurrence before clinical diagnosis of postoperative recurrence using CT images (sensitivity, 78.5%).
4.AMUSEアッセイを用いた術後再発の偽陰性ケースのレスキューの可能性
術後の再発の早期発見は、完全除去を可能にし、大腸がんの臨床結果を良好なものとする。しかし、血清腫瘍マーカーが正常範囲内でCTでも所見が曖昧である場合には再発の確定診断ができないことがある。直腸がんのケースにおいて、放射線療法がR時点での再発リンパ節(矢印)に対して行われた(図5A)。しかしながら、その部位は時点Rよりも140日早い時点(疑わしい;時点Q)で注目はされていた。一方で、本発明のAMUSEアッセイを用いた場合は、時点QにおけるAMUSEスコアに基づき、即時除去または根治的な放射線照射の判断を可能とした。また、図3Bは、右半結腸切除術後の昇行大腸がんに対してACTが施されたケースを示す。両肺に小さな再発領域(矢印)が時点Qにおいて見出された。本発明のAMUSEアッセイを用いた場合は、術後500日の時点で再発が診断できたが、従来の血清マーカー(CEA)のレベルは低く、従来の指標を採用した場合は積極的な治療が阻まれることとなった。
4. Possibility of Rescuing False-Negative Cases of Postoperative Recurrence Using the AMUSE Assay Early detection of postoperative recurrence allows complete removal and improves clinical outcomes of colorectal cancer. However, when serum tumor markers are within the normal range and CT findings are unclear, a definitive diagnosis of recurrence may not be possible. In a case of rectal cancer, radiotherapy was administered to the recurrent lymph node (arrow) at time point R (Figure 5A). However, the site was noted 140 days earlier than time point R (suspicious; time point Q). On the other hand, when the AMUSE assay of the present invention was used, the decision to immediately remove or to radically irradiate was made based on the AMUSE score at time point Q. Also, Figure 3B shows a case in which ACT was administered to ascending colorectal cancer after right hemicolectomy. Small recurrent areas (arrows) were found in both lungs at time point Q. When the AMUSE assay of the present invention was used, recurrence could be diagnosed 500 days after surgery, but the level of the conventional serum marker (CEA) was low, and aggressive treatment would be hindered if the conventional indicator was used.
補助的治療を延長したり、他の化学療法に切り替えたりすることなく、追跡調査を続行した。最終的に、時点Rで、疑われた領域の大きさの増加により肺の再発の確定診断が可能となった。根治的な治療を行う機会は失われ、全身化学療法のみが実施された。しかしながら、S状結腸がんの場合(図5C)、ACTの初日から再発を診断する日までのAMUSEアッセイのベータ値は連続的に陽性で、肝臓転移部位で手術を行う機会(時点Q1と時点Q2)が複数回逃された。このケースでは、全身化学療法が転移部位のコントロールのために施された。さらに、全大腸切除術を行った悪性腫瘍を伴う潰瘍性大腸炎の直腸がんの局所再発は誤診された(時点Q点)(図5D)。AMUSEアッセイのベータ値はACT直後に上昇し、再発の臨床診断が行われる時点Rまで陽性を保持した。従って、このケースでは、潜在的なMRD病変は化学療法に感受性があった。しかし、初期のプロトコルに従い、薬物は中止された。AMUSEアッセイがACTの有効性を評価するために適用されていれば、ACTの延長により患者の生存期間を延ばすことが可能であったと考えられる。直腸がんの根治的前方切除と術後ACTを行った患者に対してAMUSEアッセイが行われた(図5E)。右骨盤領域に再発リンパ節が時点Rで見つかった。しかし、それらは再発診断日よりも652日早い術後392日目(時点Q)にも検出されていた。 The follow-up was continued without extending the adjuvant treatment or switching to another chemotherapy. Finally, at time point R, the increase in the size of the suspicious area allowed a definitive diagnosis of lung recurrence. The opportunity to perform a definitive treatment was missed, and only systemic chemotherapy was administered. However, in the case of sigmoid colon cancer (Figure 5C), the beta value of the AMUSE assay was continuously positive from the first day of ACT to the day of diagnosing the recurrence, and the opportunity to perform surgery at the site of liver metastasis was missed multiple times (time points Q1 and Q2). In this case, systemic chemotherapy was administered to control the metastatic site. Furthermore, the local recurrence of rectal cancer in a patient with ulcerative colitis with malignant tumor who underwent total colectomy was misdiagnosed (time point Q) (Figure 5D). The beta value of the AMUSE assay increased immediately after ACT and remained positive until time point R, when the clinical diagnosis of recurrence was made. Thus, in this case, the underlying MRD lesion was sensitive to chemotherapy. However, according to the initial protocol, the drug was discontinued. If the AMUSE assay had been applied to evaluate the efficacy of ACT, it would have been possible to extend the patient's survival time by extending ACT. The AMUSE assay was performed on a patient who underwent radical anterior resection of rectal cancer and postoperative ACT (Figure 5E). Recurrent lymph nodes were found in the right pelvic region at time point R. However, they were also detected 392 days after surgery (time point Q), 652 days earlier than the date of recurrence diagnosis.
根治的手術を受けた直腸がんの患者の例を図5Fに示す。ACTの後、患者は肺に再発の疑いがあったが、画像所見と血清腫瘍マーカーのレベルに基づき治療されなかった(時点Q)。90日後、AMUSEアッセイとCTを用いて再発が確認された(時点R)。血清腫瘍マーカーであるCEAでは時点Rにおいて再発を検出できなかった。 An example of a patient with rectal cancer who underwent curative surgery is shown in Figure 5F. After ACT, the patient had suspected pulmonary recurrence but was not treated based on imaging findings and serum tumor marker levels (time point Q). Ninety days later, recurrence was confirmed using the AMUSE assay and CT (time point R). The serum tumor marker CEA failed to detect recurrence at time point R.
5.AMUSEアッセイの臨床的利点
大腸がん患者19名において、各根治的手術後1093~1897日間、追跡調査を行い、「非再発」と宣言された。AMUSEアッセイにおいて、19名の患者のうち17名(89.5%)が陰性であったが、2つのポイントが偽陽性のベータ値を示した(図6Gおよび6I、矢印)。
5. Clinical Benefits of the AMUSE Assay Nineteen patients with colorectal cancer were followed up for 1093-1897 days after each radical operation and declared “non-recurrence.” Seventeen of the 19 patients (89.5%) were negative in the AMUSE assay, but two points showed false-positive beta values (Figures 6G and 6I, arrows).
AMUSEアッセイによる診断は、従来の臨床的再発診断と比較して平均208日早く再発の診断が可能であった(図7A)。再発疑いの病変については、再発の診断に明確な所見が見られなかったため、追跡調査を行った。しかしながら、CTの利用により、再発の明確な診断が立証された。いくつかの事例は、AMUSEアッセイのアプローチが実施されていたなら、再発病変の治癒的治療が可能であったことを示唆している。 The AMUSE assay enabled diagnosis of recurrence 208 days earlier on average than conventional clinical diagnosis (Figure 7A). Suspected recurrent lesions were followed up because no clear findings were found for the diagnosis of recurrence. However, the use of CT confirmed the clear diagnosis of recurrence. Several cases suggest that curative treatment of recurrent lesions would have been possible if the AMUSE assay approach had been implemented.
28件の症例のうち、術後ACT開始時の2回目の採血ポイントとACT施行中の3回目の採血ポイントで再発が陽性であった23件と、2回目と3回目の採血ポイントの両方で陽性であった5件との間で、無病再発率が比較された(表2)。前者のグループの患者は、後者よりも有意に再発の可能性が高かった(p=0.013)(図7B)。また、両方のテストで陽性だった5人の患者と他の42人の患者との間で無病再発率を比較すると、前者のグループで有意な再発が見られた(p=1.8E-05)(図7C)。したがって、ACT期間中のAMUSEアッセイにより再発を予測できることが示された。 Among the 28 cases, the disease-free recurrence rate was compared between 23 cases in which recurrence was positive at the second blood sampling point at the start of postoperative ACT and at the third blood sampling point during ACT, and 5 cases in which recurrence was positive at both the second and third blood sampling points (Table 2). Patients in the former group were significantly more likely to recur than those in the latter group (p = 0.013) (Figure 7B). In addition, when the disease-free recurrence rate was compared between the 5 patients who were positive in both tests and the other 42 patients, a significant recurrence was observed in the former group (p = 1.8E-05) (Figure 7C). Therefore, it was demonstrated that the AMUSE assay during ACT can predict recurrence.
一方、ACT期間全体および2回目と3回目のポイントで同時にAMUSE陽性を示した進行CRC症例は、同じサンプリングポイントで陽性の血清CEAレベルを示した(図7E)。しかし、AMUSE陽性とAMUSE陰性の生存率の差は、CEA陽性とCEA陰性の生存率の差よりも大きかった。 On the other hand, advanced CRC cases that were AMUSE positive throughout the ACT period and at the second and third time points simultaneously showed positive serum CEA levels at the same sampling points (Figure 7E). However, the difference in survival rates between AMUSE positive and AMUSE negative patients was greater than the difference in survival rates between CEA positive and CEA negative patients.
術後ステージIIIのCRCに対する根治的な切除後のMRDについては、標準治療として会議によって承認されたACTが施行されているが、ACTのプロトコルは、各個人の腫瘍量を考慮せずに固定された投与量及び期間を規定している。47例のうち、7例の偽陰性と2例の偽陽性を除いた39例が層別化された(図8)。注目すべきは、適切なACT治療を受けたケースの割合が、グループAの39例中6例(16.9%)しかないことである。グループBの7例(17.9%)、グループCの15例(38.5%)に対してACTは効果がなく、また、グループDの11例(28.2%)に対しては、ACTは必要ではなかったと考えられる。 ACT, approved by the conference as the standard treatment for MRD after curative resection for postoperative stage III CRC, is performed, but the ACT protocol prescribes a fixed dose and duration without considering each individual's tumor burden. Of the 47 cases, 39 cases were stratified, excluding 7 false negatives and 2 false positives (Figure 8). It is noteworthy that the proportion of cases that received appropriate ACT treatment was only 6 out of 39 cases (16.9%) in group A. ACT was ineffective in 7 cases (17.9%) in group B and 15 cases (38.5%) in group C, and ACT was not considered necessary in 11 cases (28.2%) in group D.
本発明によれば、被検体が大腸がんに罹患しているかどうかや、大腸がんの根治手術後の被検体において大腸がんが再発したかどうかを、従来の方法よりも早く決定することができる。さらに、本発明によれば、大腸がんの補助化学療法を受けている被検体において、当該補助化学療法が効果的であるかどうかを評価することもできる。従って、本発明は、大腸がん治療の分野において極めて有用である。 The present invention makes it possible to determine whether a subject is suffering from colorectal cancer, and whether colorectal cancer has recurred in a subject after radical surgery for colorectal cancer, more quickly than with conventional methods. Furthermore, the present invention also makes it possible to evaluate whether adjuvant chemotherapy for colorectal cancer is effective in a subject receiving the chemotherapy. Therefore, the present invention is extremely useful in the field of colorectal cancer treatment.
本出願は、日本で出願された特願2023-158487(出願日:2023年9月22日)を基礎としており、その内容は本明細書に全て包含されるものである。 This application is based on patent application No. 2023-158487 filed in Japan (filing date: September 22, 2023), the contents of which are incorporated in their entirety into this specification.
Claims (16)
(1)FGD5遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対;および/または、
(2)GPC6遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対。 A diagnostic kit for colon cancer, including:
(1) a PCR primer pair capable of amplifying a part or the whole of the nucleotide sequence of the FGD5 gene; and/or
(2) A PCR primer pair capable of amplifying a part or the whole of the nucleotide sequence of the GPC6 gene.
GPC6遺伝子の塩基配列の一部または全部を増幅することができるPCRプライマー対が、SEQ ID NO:6に示される塩基配列を含む核酸と、SEQ ID NO:7に示される塩基配列を含む核酸である、
請求項13記載のキット。 A PCR primer pair capable of amplifying a part or the whole of the base sequence of the FGD5 gene is a nucleic acid having a base sequence shown in SEQ ID NO: 4 and a nucleic acid having a base sequence shown in SEQ ID NO: 5;
A PCR primer pair capable of amplifying a part or the whole of the base sequence of the GPC6 gene is a nucleic acid comprising the base sequence shown in SEQ ID NO: 6 and a nucleic acid comprising the base sequence shown in SEQ ID NO: 7.
The kit of claim 13.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2023-158487 | 2023-09-22 | ||
| JP2023158487 | 2023-09-22 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025063294A1 true WO2025063294A1 (en) | 2025-03-27 |
Family
ID=95071440
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2024/033732 Pending WO2025063294A1 (en) | 2023-09-22 | 2024-09-20 | Method for diagnosing colorectal cancer and method for evaluating effect of adjuvant chemotheraphy applied to colorectal cancer patient |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025063294A1 (en) |
Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2022238560A1 (en) * | 2021-05-14 | 2022-11-17 | Universal Diagnostics Sl | Methods for disease detection |
-
2024
- 2024-09-20 WO PCT/JP2024/033732 patent/WO2025063294A1/en active Pending
Patent Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2022238560A1 (en) * | 2021-05-14 | 2022-11-17 | Universal Diagnostics Sl | Methods for disease detection |
Non-Patent Citations (2)
| Title |
|---|
| BARAULT LUDOVIC, AMATU ALESSIO, SIRAVEGNA GIULIA, PONZETTI AGOSTINO, MORAN SEBASTIAN, CASSINGENA ANDREA, MUSSOLIN BENEDETTA, FALCO: "Discovery of methylated circulating DNA biomarkers for comprehensive non-invasive monitoring of treatment response in metastatic colorectal cancer", GUT MICROBIOTA, BRITISH MEDICAL ASSOCIATION , LONDON, UK, vol. 67, no. 11, 1 November 2018 (2018-11-01), UK , pages 1995 - 2005, XP093024614, ISSN: 0017-5749, DOI: 10.1136/gutjnl-2016-313372 * |
| DMITRIEV ALEXEY A., RUDENKO EVGENIYA E., KUDRYAVTSEVA ANNA V., KRASNOV GEORGE S., GORDIYUK VASILY V., MELNIKOVA NATALIYA V., STAKH: "Epigenetic Alterations of Chromosome 3 Revealed by NotI-Microarrays in Clear Cell Renal Cell Carcinoma", BIOMED RESEARCH INTERNATIONAL, HINDAWI PUBLISHING CORPORATION, vol. 2014, 1 January 2014 (2014-01-01), pages 1 - 9, XP093293891, ISSN: 2314-6133, DOI: 10.1155/2014/735292 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Obama et al. | Genome‐wide analysis of gene expression in human intrahepatic cholangiocarcinoma | |
| KR101437718B1 (en) | Markers for predicting gastric cancer prognostication and Method for predicting gastric cancer prognostication using the same | |
| JP6203209B2 (en) | Plasma microRNA for detection of early colorectal cancer | |
| US8198024B2 (en) | Gene expression markers for colorectal cancer prognosis | |
| Carr et al. | Differentiation of small bowel and pancreatic neuroendocrine tumors by gene-expression profiling | |
| US20100216131A1 (en) | Gene expression profiling of esophageal carcinomas | |
| JP6864089B2 (en) | Postoperative prognosis or antineoplastic compatibility prediction system for patients with advanced gastric cancer | |
| WO2018044979A1 (en) | Micrornas as biomarkers for endometriosis | |
| US20130302327A1 (en) | Marker for determination of sensitivity to triplet combination anti-cancer agent | |
| JP2024500872A (en) | Method of cancer detection using extraembryonic methylated CpG islands | |
| US10036070B2 (en) | Methods and means for molecular classification of colorectal cancers | |
| RS59161B1 (en) | Micro-rna biomarkers for identifying risk of and/or for diagnosing lung tumour | |
| US20180230545A1 (en) | Method for the prediction of progression of bladder cancer | |
| EP3129509B1 (en) | Methods and kits for identifying pre-cancerous colorectal polyps and colorectal cancer | |
| US20240068044A1 (en) | Marker composition for predicting prognosis of cancer, method for prognosis of cancer and method for providing information for determining strategy of cancer treatment | |
| Sun et al. | Circulating tumor DNA as a novel biomarker optimizing treatment for triple negative breast cancer | |
| WO2025063294A1 (en) | Method for diagnosing colorectal cancer and method for evaluating effect of adjuvant chemotheraphy applied to colorectal cancer patient | |
| US10351913B2 (en) | Compositions and methods for identification of relapse risk and treatment in patients with colorectal cancer | |
| ES2332557B1 (en) | GENOMIC FOOTPRINT FOR THE FORECAST OF THE EVOLUTION OF COLORECTAL ADENOCARCINOMA. | |
| EP4547872A1 (en) | Biomarkers in colorectal cancer | |
| JP7610265B2 (en) | Colon cancer diagnostic marker, method for assisting in the diagnosis of colon cancer, method for collecting data for the diagnosis of colon cancer, diagnostic kit for colon cancer, therapeutic agent for colon cancer, method for treating colon cancer, and method for diagnosing colon cancer | |
| WO2021191485A1 (en) | Biomarkers for predicting a patient's response to bcg therapy, methods and uses based thereon | |
| Yhim et al. | Prognostic implications of thymidylate synthase gene polymorphisms in patients with advanced small bowel adenocarcinoma treated with first-line fluoropyrimidine-based chemotherapy | |
| US20250129428A1 (en) | Methods and materials for predicting the progression of prostate cancer and treating same | |
| WO2017011440A1 (en) | Detection of tumor-derived dna in cerebrospinal fluid |
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: 24868370 Country of ref document: EP Kind code of ref document: A1 |