WO2023030422A1 - Gene combination for human tumor grading and use thereof - Google Patents

Gene combination for human tumor grading and use thereof Download PDF

Info

Publication number
WO2023030422A1
WO2023030422A1 PCT/CN2022/116390 CN2022116390W WO2023030422A1 WO 2023030422 A1 WO2023030422 A1 WO 2023030422A1 CN 2022116390 W CN2022116390 W CN 2022116390W WO 2023030422 A1 WO2023030422 A1 WO 2023030422A1
Authority
WO
WIPO (PCT)
Prior art keywords
gene
tumor
mutation
copy number
cancer
Prior art date
Application number
PCT/CN2022/116390
Other languages
French (fr)
Chinese (zh)
Inventor
熊耕砚
周利群
Original Assignee
北京大学第一医院
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 北京大学第一医院 filed Critical 北京大学第一医院
Publication of WO2023030422A1 publication Critical patent/WO2023030422A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA

Definitions

  • This application relates to the field of tumor grade detection, in particular to a gene combination for human tumor grade and its application.
  • Urothelial carcinoma is a common malignant tumor of the urinary system, mainly including bladder cancer, renal pelvis cancer, ureter cancer and urethral cancer.
  • bladder cancer mainly including bladder cancer, renal pelvis cancer, ureter cancer and urethral cancer.
  • urothelial carcinoma of the bladder which ranks fourth among all malignant tumors in men and ninth among all malignant tumors in women bit, and is rising at a rate of about 2.5% per year.
  • the most common symptom of urothelial carcinoma is hematuria, and the main treatment method is comprehensive treatment based on surgery.
  • Transurethral resection of bladder tumors can be used for bladder urothelial carcinoma with low malignancy and early stage, so as to preserve the bladder and improve the quality of life of patients; Urothelial carcinoma requires radical cystectomy + urinary diversion. Although the tumor is treated, the quality of life of the patient will be significantly reduced. Therefore, in the treatment of urothelial carcinoma, it is often necessary to grade the malignancy of the tumor tissue to assist doctors in the diagnosis of the disease, determine the treatment methods and plans, and evaluate the prognosis of patients such as tumor recurrence and survival.
  • the WHO urothelial carcinoma grading system is the most widely used urothelial carcinoma malignancy grading system.
  • the WHO urothelial carcinoma grading system was proposed in 1973, and the malignancy and risk of the tumor are graded and evaluated mainly based on the nuclei of the tissue.
  • urothelial carcinoma is divided into three levels: G1 (well differentiated), G2 (moderately differentiated), and G3 (poorly differentiated).
  • G1 well differentiated
  • G2 moderately differentiated
  • G3 poorly differentiated
  • the malignancy of urothelial carcinoma becomes more severe.
  • the risk of recurrence and metastasis after disease treatment also increases.
  • the WHO urothelial carcinoma grading system was updated, and urothelial carcinoma was divided into low-grade urothelial carcinoma and high-grade urothelial carcinoma.
  • the WHO urothelial carcinoma grading system is a purely pathological image classification system, which has the following defects: 1. It needs to be judged according to the personal experience of pathologists, which has a certain degree of subjectivity, and there are huge differences between different pathologists; 2. The actual In operation, the 1973 version of G2 grade (moderately differentiated) pathology should be finally judged as high-grade or low-grade urothelial carcinoma in the 2004 version. There are significant differences between different pathologists. The above defects are likely to lead to inaccurate classification of the malignancy of the disease, errors in the judgment of disease progression and prognosis, and affect the diagnosis and treatment of the disease.
  • next-generation sequencing technology With the maturity and promotion of next-generation sequencing technology, the method of using genetic testing to diagnose diseases has attracted widespread attention. For example, through whole exome sequencing, the mutation and copy number variation of marker genes can be detected to diagnose tumors or Assess tumor progression.
  • This method overcomes the shortcomings of subjectivity and difficulty in traditional tumor grading, and is of great significance for early diagnosis of tumors, selection of treatment methods, and judgment of prognosis.
  • the existing gene combinations and methods based on next-generation sequencing technology for grading the malignancy of urothelial carcinoma lack external validation, the malignancy grading is unreliable, and cannot be applied clinically. Therefore, it is urgent to find a method based on specific gene detection.
  • the new tumor malignancy and risk grading system grades the malignancy of urothelial carcinoma.
  • the technical problem to be solved in this application is to provide a gene combination for grading urothelial carcinoma and its application, which can grade the malignancy of urothelial carcinoma and can be used for prognosis prediction of urothelial carcinoma patients.
  • this application uses the whole exome sequencing technology to screen the data of patients with urothelial carcinoma in Peking University First Hospital who were specially screened and grouped, and finally obtain this gene combination for the detection of urothelial cancer.
  • the grade of malignancy and prognosis prediction provide clinicians and patients with more accurate malignancy information and disease prediction information of urothelial carcinoma.
  • the application provides a gene combination for human tumor grading, the gene combination is composed of gene set A, gene fragment set B and/or gene fragment set C;
  • the gene set A includes: ACVR1B, ATP4B, AZGP1, BRAF, BRCA1, BRCA2, CRYZL1, DCTN1, E2F3, EGFR, ERBB2, ERCC2, ESPL1, FGFR3, FKBP6, GZMM, H3-5, IGLL5, IRF7, METTL24, At least one of MORN5, MTOR, NPY1R, PET100, PIK3CA, PPARG, PTPN11, PTX4, RGPD8, SERPINA12, STAG2 and ZNF141;
  • the gene fragment set C includes: at least one of chr3: 12030001-12639999, chr17: 2293001-2306999 and chr17: 37830001-37979999; the position of the gene fragment in the gene fragment set C is annotated with GRCh37 as the standard, and in GRCh38 or In the new version of the human reference genome that will appear in the future, its number may change, but the pointed objective fragment position and the genes that can be used for detection will not change;
  • the detailed genes included in the gene fragment set B are as follows:
  • the detailed genes included in the gene fragment set C are as follows:
  • the gene set A includes: ACVR1B, ATP4B, AZGP1, BRAF, CRYZL1, DCTN1, FKBP6, GZMM, H3-5, IGLL5, IRF7, METTL24, MORN5, NPY1R, PET100, PTPN11, PTX4, RGPD8, SERPINA12 and at least one of ZNF141;
  • the gene fragment set C includes: chr17:2293001-2306999.
  • the tumor is a tumor of the urinary system or pan-cancer;
  • the tumor of the urinary system is a malignant tumor of the urinary system
  • the tumor of the urinary system is urothelial carcinoma
  • the pan-cancer is a cancer type in TCGA pan-cancer data.
  • the tumor grade refers to the judgment of tumor malignancy and the prediction of tumor prognosis
  • the tumor grades are divided into high-risk group and low-risk group.
  • the product includes primers, probes, reagents, kits, gene chips or detection systems for detecting the genotypes of the genes in the gene combination.
  • the product is for detection of exons and related intron regions of genes in gene set A, gene fragment set B and gene fragment set C.
  • the method for grading the tumor comprises the following steps:
  • Step S1 Evaluate the gene mutation of the genes contained in the gene set A in the cancer cell tissue, and evaluate the gene copy number variation of the gene fragment set B and the gene fragment set C in the cancer cell tissue;
  • Step S2 Based on the evaluation results of step S1, the degree of malignancy of the cancer is judged and the prognosis of the tumor is predicted.
  • the gene mutation includes base substitution mutation, deletion mutation, insertion mutation and/or fusion mutation
  • the gene copy number variation includes gene copy number increase and/or gene copy number decrease.
  • step S1 by comparing the sequencing data of the tumor tissue and the normal tissue, it is used to evaluate the gene mutation of the genes contained in the gene set A, and simultaneously evaluate the gene fragment set B and the gene fragment set Gene copy number variation in C.
  • the tumor is graded as a low-risk group; on the contrary, there is no gene mutation or copy number variation in gene set A, and at the same time, there is no gene copy number decrease in any fragment in gene fragment set B, and there is no gene fragment number in gene fragment set C Gene copy number gain in any segment was graded as a high-risk group.
  • the gene mutation status in gene set A can be used alone, without using the gene copy number status of gene fragment set B and gene fragment set C, to grade tumor malignancy and predict tumor prognosis.
  • the tumor is graded as a low-risk group; otherwise, that is, no gene in the gene set A has a gene mutation, and the tumor is graded as a high risk group risk group.
  • any gene fragments are selected from the gene combination and combined to form a new gene combination, and the same tumor grading method is used to grade tumor malignancy and predict tumor prognosis.
  • the present invention also provides a method for detecting human tumor grade using the above-mentioned gene combination.
  • the tumor is a tumor of the urinary system or pan-cancer;
  • the tumor of the urinary system is a malignant tumor of the urinary system
  • the tumor of the urinary system is urothelial carcinoma
  • the pan-cancer is a cancer type in TCGA pan-cancer data.
  • the tumor grade refers to the judgment of tumor malignancy and the prediction of tumor prognosis, which is used to guide clinical diagnosis and treatment;
  • the tumor grades are divided into high-risk group and low-risk group.
  • the method includes using primers, probes, reagents, kits, gene chips or detection systems for detecting the genotypes of the genes in the gene combination.
  • the method is to detect exons and related intron regions of genes in gene set A, gene fragment set B and gene fragment set C.
  • the method for grading the tumor comprises the following steps:
  • Step S1 Evaluate the gene mutation of the genes contained in the gene set A in the cancer cell tissue, and evaluate the gene copy number variation of the gene fragment set B and the gene fragment set C in the cancer cell tissue;
  • Step S2 Based on the evaluation results of step S1, the degree of malignancy of the cancer is judged and the prognosis of the tumor is predicted.
  • the gene mutation includes base substitution mutation, deletion mutation, insertion mutation and/or fusion mutation
  • the gene copy number variation includes gene copy number increase and/or gene copy number decrease.
  • step S1 by comparing the sequencing data of the tumor tissue and normal tissue, it is used to evaluate the gene mutation of the genes contained in the gene set A, and simultaneously evaluate the gene fragment set B and the gene fragment set C gene copy number variation.
  • the tumor is graded as a low-risk group; on the contrary, there is no gene mutation in gene set A, and at the same time, there is no gene copy number reduction in any fragment in gene fragment set B, and there is no gene in any fragment in gene fragment set C. Copy number gain, the tumor was graded as high risk group.
  • the tumor is graded as a low-risk group; otherwise, that is, no gene in the gene set A has a gene mutation, and the tumor is graded as a high risk group. risk group.
  • any gene fragments from the gene combination to form a new gene combination and use the same tumor grading method to grade tumor malignancy and predict tumor prognosis, so as to guide clinical diagnosis and treatment.
  • the detection gene combination in this application is obtained from the high-throughput sequencing data of actual urothelial cancer cases in Peking University First Hospital through specific paired cluster analysis, and the real data has higher The reliability and credibility of the system can accurately grade the degree of malignancy and predict the prognosis of urothelial carcinoma and pan-cancer.
  • the gene combination described in this application includes the diversity of the gene combination, from which a variety of gene combinations can be optimized for judging the malignancy of urothelial carcinoma and pan-cancer, and for different clinical situations.
  • this application conducts targeted sequencing analysis for specific genes and DNA fragments, which can significantly improve the sequencing depth and accuracy at the same cost. In this case, the cost can be significantly saved, and the universality is wide.
  • Fig. 1 is the Kaplan-Meier survival analysis graph with tumor-specific survival as the primary endpoint after the urothelial carcinoma malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 1 of the present application;
  • Fig. 2 is a Kaplan-Meier survival analysis chart with tumor progression-free survival as the primary endpoint after the urothelial carcinoma malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 1 of the present application;
  • Fig. 3 is a Kaplan-Meier survival analysis chart with tumor disease-free survival as the primary endpoint after the urothelial carcinoma malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 1 of the present application;
  • Fig. 4 is a Kaplan-Meier survival analysis chart with overall survival as the primary endpoint after the urothelial carcinoma malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 1 of the present application;
  • Fig. 5 is a graph of Kaplan-Meier survival analysis with tumor-specific survival as the primary end point after using the gene combination 1 of the present application to grade the malignancy of urothelial carcinoma in Experimental Example 2 of the present application.
  • Fig. 6 is a graph of Kaplan-Meier survival analysis with tumor disease-free survival as the primary endpoint after grading the malignancy of urothelial carcinoma using gene combination 1 of the present application in Experimental Example 2 of the present application.
  • Fig. 7 is a graph of Kaplan-Meier survival analysis with the overall survival as the primary endpoint after the urothelial carcinoma malignancy grading standard was graded using the gene combination 1 of the present application in Experimental Example 2 of the present application.
  • Fig. 8 is a graph of Kaplan-Meier survival analysis with tumor-specific survival as the primary end point after using the gene set A of the present application to grade the malignant degree of urothelial carcinoma in Experimental Example 3 of the present application.
  • Fig. 9 is a graph of Kaplan-Meier survival analysis with tumor progression-free survival as the primary endpoint after grading the malignancy of urothelial carcinoma using the gene set A of the present application in Experimental Example 3 of the present application.
  • Fig. 10 is a graph of Kaplan-Meier survival analysis with tumor disease-free survival as the primary endpoint after grading the malignancy of urothelial carcinoma using the gene set A of the present application in Experimental Example 3 of the present application.
  • Fig. 11 is a graph of Kaplan-Meier survival analysis with overall survival as the primary endpoint after grading the malignancy of urothelial carcinoma using the gene set A of the present application in Experimental Example 3 of the present application.
  • Fig. 12 is a Kaplan-Meier survival analysis graph with tumor disease-free survival as the primary endpoint after the pan-cancer malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 4 of the present application;
  • Fig. 13 is a Kaplan-Meier survival analysis chart with tumor progression-free survival as the primary endpoint after using the gene combination in Example 1 of the present application in Experimental Example 4 of the present application to perform pan-cancer malignancy grading standard grading;
  • Figure 14 is a Kaplan-Meier survival analysis chart with overall survival as the primary endpoint after the pan-cancer malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 4 of the present application;
  • Fig. 15 is a Kaplan-Meier survival analysis chart with tumor-specific survival as the primary endpoint after the pan-cancer malignancy grading standard is graded using the gene combination 1 of the present application in Experimental Example 5 of the present application;
  • Fig. 16 is a Kaplan-Meier survival analysis graph with tumor progression-free survival as the primary endpoint after the pan-cancer malignancy grading standard grading was performed using the gene combination 1 of the present application in Experimental Example 5 of the present application;
  • Fig. 17 is a Kaplan-Meier survival analysis graph with tumor disease-free survival as the primary endpoint after the pan-cancer malignancy grading standard is graded using the gene combination 1 of the present application in Experimental Example 5 of the present application;
  • Fig. 18 is a Kaplan-Meier survival analysis chart with overall survival as the primary endpoint after the pan-cancer malignancy grading standard is graded using the gene combination 1 of the present application in Experimental Example 5 of the present application;
  • Fig. 19 is a graph of Kaplan-Meier survival analysis with tumor-specific survival as the primary endpoint after using the gene set A of the present application to perform pan-cancer malignancy grading standard grading in Experimental Example 6 of the present application.
  • Fig. 20 is a graph of Kaplan-Meier survival analysis with tumor progression-free survival as the primary endpoint after using the gene set A of the present application to perform pan-cancer malignancy grading standard grading in Experimental Example 6 of the present application.
  • Fig. 21 is a graph of Kaplan-Meier survival analysis with tumor disease-free survival as the primary endpoint after using the gene set A of the present application to perform pan-cancer malignancy grading standard grading in Experimental Example 6 of the present application.
  • Fig. 22 is a graph of Kaplan-Meier survival analysis with overall survival as the primary endpoint after using the gene set A of the present application to perform pan-cancer malignancy grading standard grading in Experimental Example 6 of the present application.
  • Embodiment 1 is used for the gene combination (Panel) of human tumor classification
  • This application mainly uses Peking University First Hospital urothelial carcinoma exome sequencing high-throughput database for screening, and confirmed a gene combination (panel) for human tumor grading, the gene combination includes gene set A, gene fragment Set B and gene fragment set C;
  • the gene set A includes: ACVR1B, ATP4B, AZGP1, BRAF, BRCA1, BRCA2, CRYZL1, DCTN1, E2F3, EGFR, ERBB2, ERCC2, ESPL1, FGFR3, FKBP6, GZMM, H3-5, IGLL5, IRF7, METTL24, At least one of MORN5, MTOR, NPY1R, PET100, PIK3CA, PPARG, PTPN11, PTX4, RGPD8, SERPINA12, STAG2 and ZNF141;
  • the gene fragment set C includes: at least one of chr3: 12030001-12639999, chr17: 2293001-2306999 and chr17: 37830001-37979999; the position of the gene fragment in the gene fragment set C is annotated with GRCh37 as the standard, and in GRCh38 or In the new version of the human reference genome that will appear in the future, its number may change, but the pointed objective fragment position and the genes that can be used for detection will not change;
  • the detailed genes included in the gene fragment set B are as follows:
  • the detailed genes included in the gene fragment set C are as follows:
  • Example 2 A method for grading the degree of malignancy and predicting prognosis of human urothelial carcinoma
  • This embodiment provides a method for detecting the grade of human tumors, including using the gene panel in Embodiment 1 to grade the malignancy of human urothelial carcinoma and predict the prognosis, and the specific steps are as follows:
  • the urothelial carcinoma tissue specimens can be urothelial carcinoma cell lines, fresh urothelial carcinoma specimens, frozen urothelial carcinoma specimens or paraffin-embedded urine Urothelial carcinoma specimens; healthy control tissues can be tissues from known healthy people, or paracancerous tissues or peripheral blood from urothelial carcinoma patients themselves.
  • the paraffin-embedded urothelial carcinoma specimens were selected, and the healthy control tissue was normal paracancerous tissue, DNA was extracted by conventional methods, and a library was constructed by conventional methods, and finally the gene combination (panel ) to perform targeted high-throughput sequencing, compare the sequencing data of urothelial cancer tissue and healthy tissue, and obtain the gene mutation (Mutation) of each gene of gene set A in the gene combination of the urothelial cancer tissue, and the gene fragment Copy number variation (CNV) of each gene in set B and gene fragment set C.
  • the gene mutation includes base substitution mutation, deletion mutation, insertion mutation and fusion mutation
  • the gene copy number variation includes gene copy number increase and gene copy number decrease.
  • the urothelial carcinoma Patients belong to the low-risk group, with lower tumor malignancy and better tumor prognosis; on the contrary, no genes in gene set A have gene mutations, and at the same time, no gene fragments in gene set B have gene copy number reduction, and at the same time, gene set B has In C, there is no gene copy number increase in any segment, and the urothelial carcinoma patients are in the high-risk group, with higher tumor malignancy and poor tumor prognosis.
  • This application allows only the detection of gene mutations without the need for detection of copy number variations, that is, the detection of gene mutations in gene set A alone, and the same criteria are used to grade the malignancy of urothelial carcinoma and predict tumor prognosis: if there is gene set A Gene mutation of at least one gene in gene set A, the urothelial cancer patients are in the low-risk group, the tumor malignancy is lower, and the tumor prognosis is better; on the contrary, there is no gene mutation in gene set A, and the urinary tract cancer patients are in the low-risk group. Patients with roadside carcinoma belong to the high-risk group, with higher tumor malignancy and poor prognosis.
  • the genes in the gene combination (panel) in Example 1 are allowed to be selected and recombined to form a new gene combination.
  • the judging criteria are, from the gene set For the genes selected in A, the gene mutation of at least one of them indicates that the urothelial cancer patients are in the low-risk group; for the gene fragments selected from gene fragment set B, the gene copy number of at least one region decrease, indicating that the patients with urothelial carcinoma are in the low-risk group; for the gene fragments selected from the gene fragment set C, at least one region of the gene copy number increases, indicating that the patients with urothelial carcinoma are in the low-risk group
  • Example 4 A method for grading and predicting the degree of malignancy of human pancancer (Pancancer)
  • This embodiment provides a method for human tumor grading detection, including, using the gene combination (panel) in Example 1 to perform human pancancer (Pancancer) malignancy grading and prognosis prediction, and the specific steps are as follows:
  • pan-cancer is defined as all cancer types in TCGA pan-cancer data, including adrenal cancer, urothelial cancer, breast cancer, cervical cancer, bile duct cancer, colon cancer, lymphoma, esophageal cancer, gum Glioblastoma, head and neck squamous cell carcinoma, chromophobe renal cell carcinoma, clear cell renal cell carcinoma, renal papillary cell carcinoma, leukemia, glioma, hepatocellular carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma , ovarian serous cystadenocarcinoma, pancreatic cancer, pheochromocytoma and paraganglioma, prostate cancer, rectal cancer, sarcoma, skin melanoma, gastric cancer, testicular cancer, thyroid cancer, thymus cancer, endometrial cancer, uterine Sarcoma, uveal melanoma
  • Example 1 paraffin-embedded pan-cancer specimens were selected, and normal paracancerous tissues were used as healthy control tissues. DNA was extracted by conventional methods, and libraries were constructed by conventional methods. Finally, the gene combination (panel) in Example 1 was used to carry out Targeted high-throughput sequencing, compare the sequencing data of pan-cancer tissue and healthy tissue, and obtain the gene mutation (Mutation) of each gene in gene set A in the gene combination of the pan-cancer tissue, as well as gene fragment set B and gene fragment set Copy number variation (CNV) status of each fragment in C.
  • the gene mutation includes base substitution mutation, deletion mutation, insertion mutation and fusion mutation
  • the gene copy number variation includes gene copy number increase and gene copy number decrease.
  • the pan-cancer patient is The low-risk group has a better tumor prognosis; on the contrary, there is no gene mutation in gene set A, and there is no gene copy number reduction in any fragment in gene fragment set B, and there is no gene copy in any fragment in gene fragment set C.
  • the number of patients with pan-cancer increases, and the pan-cancer patients are a high-risk group with poor tumor prognosis.
  • This application allows the detection of gene mutations only, without the need to detect copy number variations, that is, the mutations of genes in gene set A are detected separately, and the same criteria are used to grade the degree of malignancy of pan-cancer and predict tumor prognosis: if there is at least If there is a gene mutation in one gene, the pan-cancer patients are in the low-risk group, the tumor malignancy is lower, and they have better tumor prognosis; on the contrary, if there is no gene mutation in gene set A, the pan-cancer patients are in the high-risk group. The risk group, with higher tumor malignancy, has poorer tumor prognosis.
  • the genes in the gene combination (panel) in Example 1 are allowed to be selected and recombined to form a new gene combination.
  • the judging criteria are: from the gene set For the genes selected in A, the gene mutation of at least one gene indicates that the pan-cancer patients are in the low-risk group; for the gene fragments selected from the gene fragment set B, the copy number of at least one region of the gene is reduced, It indicates that the pan-cancer patients belong to the low-risk group; if the gene fragments selected from the gene fragment set C have an increased gene copy number in at least one region, it indicates that the pan-cancer patients belong to the low-risk group.
  • pan-cancer patients are a high-risk group.
  • Experimental Example 1 Feasibility verification of the gene combination and detection method used for human tumor grading in evaluating the malignancy grading and prognosis prediction of human urothelial carcinoma
  • Urothelial carcinoma is the most common pathological type of urothelial tumors, accounting for more than 90% of all urothelial carcinomas.
  • TCGA PanCancer Atlas
  • Bladder Urothelial Cancer Database is a globally recognized bladder urothelial cancer database , the database was updated in 2020, and after merging the data of Memorial Sloan Kettering Cancer Center (MSKCC) bladder urothelial carcinoma, the MSK/TCGA2020 bladder urothelial carcinoma database was formed and released to the public.
  • MSK 2015 upper urinary tract urothelial cancer (renal pelvis cancer, ureteral cancer) database is a highly reliable upper urinary tract urothelial cancer database with complete follow-up data released to the public.
  • the combined new database is collectively referred to as the MSK/TCGA database in this application.
  • the MSK/TCGA database has a total of 972 patients with urothelial carcinoma, of which 928 patients have complete gene mutation and copy number variation data, which are suitable for the application conditions of this application.
  • 861 patients in this database have complete prognostic data of overall survival, which can be used for prognostic detection of overall survival;
  • 704 patients in this database have complete prognostic data of tumor progression-free survival, which can be used for tumor progression-free survival Prognosis detection;
  • 391 patients in this database have complete tumor-specific (Disease Specific) survival prognosis data, which can be used for tumor-specific survival prognosis detection;
  • 401 patients in this database have complete tumor-specific disease-free (Disease Free) survival data , which can be used for prognostic detection of tumor disease-free survival.
  • the tumor-specific survival ( Figure 1), tumor progression-free survival ( Figure 2), tumor disease-free survival (Figure 3) and overall survival ( Figure 4) in the low-risk group were all statistically different and in line with the grouping expectations of this application:
  • This application allows to select any gene fragments from the gene panel and combine them to form a new gene combination, and use the same criteria to grade the malignancy of urothelial carcinoma and predict the prognosis of the tumor.
  • the gene set A1 (Table 4) is selected from the gene set A of the gene combination (panel)
  • the gene fragment set B1 (Table 5) is selected from the gene fragment set B
  • the gene fragment is selected from the gene fragment set C Set C1 (Table 5) constitutes gene panel 1 (panel 1), which is used for grading the malignancy of urothelial carcinoma and predicting prognosis, and uses the MSK/TCGA database for feasibility analysis.
  • the judgment criteria are: if there is a gene mutation in at least one gene in gene set A1, or there is a gene copy number decrease in at least one region in gene fragment set B1, or there is an increase in gene copy number in at least one region in gene fragment set C1 , indicating that the urothelial carcinoma patients are in the low-risk group and have better tumor prognosis; on the contrary, no genes in the gene set A1 have gene mutations, and at the same time, there is no gene copy number reduction in any fragments in the gene fragment set B1, and at the same time If there is no gene copy number increase in any fragment in the gene fragment set C1, the patients with this type of urothelial carcinoma are in the high-risk group and have poor tumor prognosis.
  • the gene combination 1 panel 1 is optimized on the basis of the gene combination (panel). Since there are fewer genes to be detected, it has a lower cost.
  • This application allows the detection of gene mutations only, without the detection of copy number variations, and uses the same criteria to grade the malignancy of urothelial carcinoma and predict tumor prognosis.
  • the gene mutations of all genes in gene set A are used for grading the malignancy of urothelial carcinoma and predicting prognosis, and the MSK/TCGA database is used for feasibility analysis.
  • the judging criteria are: if there is a gene mutation in at least one gene in gene set A, it indicates that the urothelial carcinoma patients are in the low-risk group and have a better tumor prognosis; otherwise, there is no gene in gene set A If there is a gene mutation, the patients with this type of urothelial carcinoma belong to the high-risk group and have a poor tumor prognosis. It should be noted that in this experimental example, only gene mutations are detected, and copy number variations are not detected, so the cost is lower.
  • Example 2 According to the method described in Example 2, the above-mentioned 926 patients were graded for malignancy using gene combination A, and were successfully divided into high-risk group and low-risk group, of which the high-risk group accounted for 33.6%, and the low-risk group accounted for 66.4%.
  • gene mutation status of gene set A in this application is detected, and the gene copy number variation is not detected, and the malignant degree and prognosis prediction of urothelial carcinoma can still be carried out.
  • Gene mutation detection of other gene combinations can still grade the malignancy and predict the prognosis of patients with urothelial carcinoma.
  • the TCGA (PanCancer Atlas) pan-cancer database is a globally recognized pan-cancer database, which can be used to test the feasibility and credibility of this application for assessing pan-cancer malignancy grading and prognosis prediction.
  • TCGA PanCancer Atlas pan-cancer data has a total of 10,953 patients (10,967 cases) pan-cancer data, which can be used to test the feasibility and reliability of this application for assessing pan-cancer malignancy grading and prognosis prediction.
  • Experimental Example 5 Feasibility verification of the optimal gene combination and detection method in assessing human pan-cancer malignancy grading and prognosis prediction
  • This application allows to select any gene fragments from the gene panel and combine them to form a new gene combination, and use the same judgment standard to grade the degree of malignancy of pan-cancer and predict the prognosis of tumors.
  • select and use the gene combination 1 (panel 1) in Experimental Example 2 for pan-cancer malignancy grading and prognosis prediction and use the TCGA (PanCancer Atlas) pan-cancer database for feasibility analyze.
  • the judgment criteria are: if there is a gene mutation in at least one gene in gene set A1, or there is a gene copy number decrease in at least one region in gene fragment set B1, or there is an increase in gene copy number in at least one region in gene fragment set C1 , indicating that the pan-cancer patients are a low-risk group with better tumor prognosis; on the contrary, no genes in the gene set A1 have gene mutations, and at the same time, no gene fragments in the gene fragment set B1 have gene copy number reduction, and at the same time, the gene fragments If there is no gene copy number increase in any fragment in set C1, then this type of pan-cancer patients is a high-risk group with poor tumor prognosis. It should be noted that in this experimental example, the gene combination 1 (panel 1) is optimized on the basis of the gene combination (panel). Since there are fewer genes to be detected, it has a lower cost.
  • gene combination 1 (panel 1) carry out malignancy classification to 10967 routine patients in the above-mentioned pan-cancer database, successfully divide into high-risk group and low-risk group, wherein high-risk group It accounted for 81.5%, and the low-risk group accounted for 18.5%.
  • high-risk group and low-risk group have tumor-specific survival ( Figure 15), tumor progression-free survival ( Figure 16), and tumor-free survival.
  • This application allows the detection of gene mutations only, without the detection of copy number variations, and uses the same criteria to grade the malignancy of urothelial carcinoma and predict tumor prognosis.
  • the gene mutations of all genes in gene set A in Example 1 are used for pan-cancer malignancy grading and prognosis prediction, and the TCGA pan-cancer database is used for feasibility analysis.
  • the criterion for judging is: if there is a gene mutation in at least one gene in gene set A, it indicates that the pan-cancer patients are in the low-risk group and have a better tumor prognosis; otherwise, no gene in gene set A has a gene mutation mutation, the pan-cancer patients belong to the high-risk group with poor tumor prognosis. It should be noted that in this experimental example, only gene mutations are detected, and copy number variations are not detected, so the cost is lower.
  • Example 4 the above-mentioned 10,967 patients were graded for malignancy using gene combination A, and were successfully divided into high-risk group and low-risk group, of which the high-risk group accounted for 64.1 %, the low-risk group accounted for 35.9%.
  • the tumor-specific survival Fig. 19
  • tumor progression-free survival Fig. 20
  • tumor disease-free survival Fig. 21
  • overall survival Fig.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present application relates to the field of tumor grading detection, and in particular to a gene combination for human tumor grading and an application thereof. The gene combination used for human tumor grading comprises a gene set A, a gene fragment set B and a gene fragment set C.

Description

一种用于人肿瘤分级的基因组合及其用途A gene combination for human tumor grading and its use
相关申请的交叉引用Cross References to Related Applications
本申请要求在2021年9月2日提交中国专利局、申请号为202111028348.8、发明名称为“一种用于人肿瘤分级的基因组合及其用途”的中国专利申请的优先权,其全部内容通过引用的方式并入本文中。This application claims the priority of the Chinese patent application with the application number 202111028348.8 and the title of the invention "A Gene Combination for Human Tumor Grading and Its Use" submitted to the China Patent Office on September 2, 2021, the entire content of which is passed Incorporated herein by reference.
技术领域technical field
本申请涉及肿瘤分级检测领域,具体涉及一种用于人肿瘤分级的基因组合及其用途。This application relates to the field of tumor grade detection, in particular to a gene combination for human tumor grade and its application.
背景技术Background technique
尿路上皮癌是常见的泌尿系统恶性肿瘤,主要包括膀胱癌、肾盂癌、输尿管癌和尿道癌。近年来,尿路上皮癌的发病率逐年升高,其中最常见的为膀胱尿路上皮癌,其发病率在男性中位于所有恶性肿瘤的第四位,在女性中位于所有恶性肿瘤的第九位,且每年以2.5%左右的速度上升。尿路上皮癌最常见的症状为血尿,其主要治疗手段是以手术为基础的综合治疗。针对恶性程度较低、分期较早的膀胱尿路上皮癌,可以采用经尿道膀胱肿瘤电切术进行治疗,从而保留膀胱,患者生活质量较高;而针对恶性程度较高、分期较晚的膀胱尿路上皮癌则需要行根治性膀胱切除术+尿流改道术,虽然治疗了肿瘤,但患者的生活质量将明显下降。故针对尿路上皮癌,治疗中往往需要对肿瘤组织进行恶性程度分级以协助医生进行疾病诊断,确定治疗手段和方案,同时评估患者肿瘤复发和生存等预后情况。目前,WHO尿路上皮癌分级系统是应用最广泛的尿路上皮癌恶性程度分级系统。Urothelial carcinoma is a common malignant tumor of the urinary system, mainly including bladder cancer, renal pelvis cancer, ureter cancer and urethral cancer. In recent years, the incidence of urothelial carcinoma has been increasing year by year, the most common of which is urothelial carcinoma of the bladder, which ranks fourth among all malignant tumors in men and ninth among all malignant tumors in women bit, and is rising at a rate of about 2.5% per year. The most common symptom of urothelial carcinoma is hematuria, and the main treatment method is comprehensive treatment based on surgery. Transurethral resection of bladder tumors can be used for bladder urothelial carcinoma with low malignancy and early stage, so as to preserve the bladder and improve the quality of life of patients; Urothelial carcinoma requires radical cystectomy + urinary diversion. Although the tumor is treated, the quality of life of the patient will be significantly reduced. Therefore, in the treatment of urothelial carcinoma, it is often necessary to grade the malignancy of the tumor tissue to assist doctors in the diagnosis of the disease, determine the treatment methods and plans, and evaluate the prognosis of patients such as tumor recurrence and survival. Currently, the WHO urothelial carcinoma grading system is the most widely used urothelial carcinoma malignancy grading system.
WHO尿路上皮癌分级系统于1973年被提出,主要根据组织细胞核情况对该肿瘤的恶性程度及危险度进行分级评价。依据1973版WHO分级标准,将尿路上皮癌分为G1(高分化),G2(中分化),G3(低分化)三个层级,随着层级的增高,尿路上皮癌的恶性程度就越高,疾病治疗后出现复发、转移的风险也随之升高。2004年,WHO尿路上皮癌分级系统进行了更新,将尿路上皮癌分为低级别尿路上皮癌和高级别尿路上皮癌,其中,高级别尿路上皮癌具有更高的恶性程度,疾病治疗后出现复发、转移的风险也随之升高。然而,WHO尿路上皮癌分级系统是纯病理图像分型系统,存在如下缺陷:1、需要根据病理医生个人的经验进行判断,存在一定的主观性,不同病理医生之间差别巨大;2、实际操作中,1973版G2级(中分化)病理最终应判定为2004版中高级别或低级别尿路上皮癌在不同的病理医生间具有明显差别。以上的缺陷容易导致疾病的恶性程度分级不准确,对疾病进展和预后判断发生误差,影响疾病的诊疗。The WHO urothelial carcinoma grading system was proposed in 1973, and the malignancy and risk of the tumor are graded and evaluated mainly based on the nuclei of the tissue. According to the 1973 WHO grading standard, urothelial carcinoma is divided into three levels: G1 (well differentiated), G2 (moderately differentiated), and G3 (poorly differentiated). As the level increases, the malignancy of urothelial carcinoma becomes more severe. High, the risk of recurrence and metastasis after disease treatment also increases. In 2004, the WHO urothelial carcinoma grading system was updated, and urothelial carcinoma was divided into low-grade urothelial carcinoma and high-grade urothelial carcinoma. Among them, high-grade urothelial carcinoma has a higher degree of malignancy. The risk of recurrence and metastasis after disease treatment also increases. However, the WHO urothelial carcinoma grading system is a purely pathological image classification system, which has the following defects: 1. It needs to be judged according to the personal experience of pathologists, which has a certain degree of subjectivity, and there are huge differences between different pathologists; 2. The actual In operation, the 1973 version of G2 grade (moderately differentiated) pathology should be finally judged as high-grade or low-grade urothelial carcinoma in the 2004 version. There are significant differences between different pathologists. The above defects are likely to lead to inaccurate classification of the malignancy of the disease, errors in the judgment of disease progression and prognosis, and affect the diagnosis and treatment of the disease.
随着二代测序技术的成熟和推广,利用基因检测来诊断疾病的方法受到了广泛的瞩目,如通过全外显子测序,检测标记物基因的突变及拷贝数变异情况,以此诊断肿瘤或判断肿瘤的进展。此方法克服了传统的肿瘤分级中存在的主观性影响和分级难度大的缺陷,对于肿瘤的早期诊断、治疗方式选择和预后判断均具有重大的意义。然而,现存基于二代测序技术且用于尿路上皮癌恶性程度分级的基因组合和方法,均缺乏外部验证,恶性程度分级不可靠,临床无法应用,因此,亟需找到一种基于特定基因检测的新型的肿瘤恶性程度及危险度分级系统对尿路上皮癌进行恶性程度分级。With the maturity and promotion of next-generation sequencing technology, the method of using genetic testing to diagnose diseases has attracted widespread attention. For example, through whole exome sequencing, the mutation and copy number variation of marker genes can be detected to diagnose tumors or Assess tumor progression. This method overcomes the shortcomings of subjectivity and difficulty in traditional tumor grading, and is of great significance for early diagnosis of tumors, selection of treatment methods, and judgment of prognosis. However, the existing gene combinations and methods based on next-generation sequencing technology for grading the malignancy of urothelial carcinoma lack external validation, the malignancy grading is unreliable, and cannot be applied clinically. Therefore, it is urgent to find a method based on specific gene detection. The new tumor malignancy and risk grading system grades the malignancy of urothelial carcinoma.
发明内容Contents of the invention
因此,本申请要解决的技术问题在于提供一种用于尿路上皮癌分级的基因组合及其用途,它能够对尿路上皮癌恶性程度进行分级并可用于尿路上皮癌患者的预后预测,为了实现这一目的,本申请使用全外显子测序技术,针对特殊筛选和分组的北京大学第一医院的尿路上皮癌患者数据进行筛选,最终得到本基因组合,用于尿路上皮癌的恶性程度分级和预后预测,为临床医生和患者提供了更为准确的尿路上皮癌恶性程度信息和疾病预测信息。Therefore, the technical problem to be solved in this application is to provide a gene combination for grading urothelial carcinoma and its application, which can grade the malignancy of urothelial carcinoma and can be used for prognosis prediction of urothelial carcinoma patients, In order to achieve this goal, this application uses the whole exome sequencing technology to screen the data of patients with urothelial carcinoma in Peking University First Hospital who were specially screened and grouped, and finally obtain this gene combination for the detection of urothelial cancer. The grade of malignancy and prognosis prediction provide clinicians and patients with more accurate malignancy information and disease prediction information of urothelial carcinoma.
本申请提供了一种用于人肿瘤分级的基因组合,所述的基因组合由基因集A、基因片段集B和/或基因片段集C组成;The application provides a gene combination for human tumor grading, the gene combination is composed of gene set A, gene fragment set B and/or gene fragment set C;
所述的基因集A包括:ACVR1B、ATP4B、AZGP1、BRAF、BRCA1、BRCA2、CRYZL1、DCTN1、 E2F3、EGFR、ERBB2、ERCC2、ESPL1、FGFR3、FKBP6、GZMM、H3-5、IGLL5、IRF7、METTL24、MORN5、MTOR、NPY1R、PET100、PIK3CA、PPARG、PTPN11、PTX4、RGPD8、SERPINA12、STAG2和ZNF141中至少一个;The gene set A includes: ACVR1B, ATP4B, AZGP1, BRAF, BRCA1, BRCA2, CRYZL1, DCTN1, E2F3, EGFR, ERBB2, ERCC2, ESPL1, FGFR3, FKBP6, GZMM, H3-5, IGLL5, IRF7, METTL24, At least one of MORN5, MTOR, NPY1R, PET100, PIK3CA, PPARG, PTPN11, PTX4, RGPD8, SERPINA12, STAG2 and ZNF141;
所述的基因片段集B包括:chr1:155289001-155911999、chr1:226259001-226736999、chr8:30515001-30892999、chr8:42065001-43147999、chr9:134612001-135906999、chr11:48509001-55112999、chr14:24768001-105145999、chr16:1-73083999、chr18:1-319999和chr18:15325001-19749999中至少一个;所述基因片段集B中基因片段位置以GRCh37为标准进行注释,在GRCh38或未来出现的新版人类参考基因组中,其数字可能发生改变,但指向的客观片段位置和可用于检测的基因不会发生改变;所述的基因片段集B包括:chr1:155289001-155911999、chr1:226259001-226736999、chr8:30515001-30892999、chr8:42065001-43147999、chr9:134612001-135906999、chr11:48509001-55112999、chr14:24768001-105145999 , at least one of chr16:1-73083999, chr18:1-319999 and chr18:15325001-19749999; the position of the gene segment in the gene segment set B is annotated with GRCh37 as the standard, in GRCh38 or the new version of the human reference genome that will appear in the future , its number may change, but the targeted fragment position and the genes available for detection will not change;
所述的基因片段集C包括:chr3:12030001-12639999、chr17:2293001-2306999和chr17:37830001-37979999中至少一个;所述基因片段集C中基因片段位置以GRCh37为标准进行注释,在GRCh38或未来出现的新版人类参考基因组中,其数字可能发生改变,但指向的客观片段位置和可用于检测的基因不会发生改变;The gene fragment set C includes: at least one of chr3: 12030001-12639999, chr17: 2293001-2306999 and chr17: 37830001-37979999; the position of the gene fragment in the gene fragment set C is annotated with GRCh37 as the standard, and in GRCh38 or In the new version of the human reference genome that will appear in the future, its number may change, but the pointed objective fragment position and the genes that can be used for detection will not change;
可选的,所述的基因片段集B中包括的详细基因如下表:Optionally, the detailed genes included in the gene fragment set B are as follows:
表1基因片段集BTable 1 Gene Fragment Set B
Figure PCTCN2022116390-appb-000001
Figure PCTCN2022116390-appb-000001
Figure PCTCN2022116390-appb-000002
Figure PCTCN2022116390-appb-000002
Figure PCTCN2022116390-appb-000003
Figure PCTCN2022116390-appb-000003
Figure PCTCN2022116390-appb-000004
Figure PCTCN2022116390-appb-000004
Figure PCTCN2022116390-appb-000005
Figure PCTCN2022116390-appb-000005
Figure PCTCN2022116390-appb-000006
Figure PCTCN2022116390-appb-000006
可选的,所述的基因片段集C中包括的详细基因如下表:Optionally, the detailed genes included in the gene fragment set C are as follows:
表2基因片段集CTable 2 Gene fragment set C
基因片段位置gene fragment position 可用于检测的基因Genes Available for Testing
chr3:12030001-12639999Chr3: 12030001-12639999 TIMP4,SYN2,PPARG,TSEN2,MKRN2中至少一个At least one of TIMP4, SYN2, PPARG, TSEN2, MKRN2
chr17:2293001-2306999chr17:2293001-2306999 MNTMNT
chr17:37830001-37979999chr17:37830001-37979999 GRB7,ERBB2,IKZF3中至少一个At least one of GRB7, ERBB2, IKZF3
可选的,所述基因集A包括:ACVR1B、ATP4B、AZGP1、BRAF、CRYZL1、DCTN1、FKBP6、GZMM、H3-5、IGLL5、IRF7、METTL24、MORN5、NPY1R、PET100、PTPN11、PTX4、RGPD8、SERPINA12和ZNF141至少一个;Optionally, the gene set A includes: ACVR1B, ATP4B, AZGP1, BRAF, CRYZL1, DCTN1, FKBP6, GZMM, H3-5, IGLL5, IRF7, METTL24, MORN5, NPY1R, PET100, PTPN11, PTX4, RGPD8, SERPINA12 and at least one of ZNF141;
可选的,所述基因片段集B包括:chr1:155289001-155911999、chr1:226259001-226736999、chr8:30515001-30892999、chr8:42065001-43147999、chr9:134612001-135906999、chr14:24768001-105145999、chr16:1-73083999、chr18:1-319999和chr18:15325001-19749999至少一个;可选的,所述基因片段集B包括:chr1:155289001-155911999、chr1:226259001-226736999、chr8:30515001-30892999、chr8:42065001-43147999、chr9:134612001-135906999、chr14:24768001-105145999、chr16: At least one of 1-73083999, chr18:1-319999 and chr18:15325001-19749999;
可选的,所述基因片段集C包括:chr17:2293001-2306999。Optionally, the gene fragment set C includes: chr17:2293001-2306999.
所述的基因组合在制备用于人肿瘤分级检测的产品中的用途。Use of the gene combination in preparing products for human tumor grade detection.
可选的,所述的肿瘤为泌尿系统肿瘤或泛癌;Optionally, the tumor is a tumor of the urinary system or pan-cancer;
可选的,所述泌尿系统肿瘤为泌尿系统恶性肿瘤;Optionally, the tumor of the urinary system is a malignant tumor of the urinary system;
可选的,所述泌尿系统肿瘤为尿路上皮癌;Optionally, the tumor of the urinary system is urothelial carcinoma;
可选的,所述泛癌为TCGA泛癌数据中的癌种。Optionally, the pan-cancer is a cancer type in TCGA pan-cancer data.
可选的,所述的肿瘤分级是指肿瘤恶性程度判断和肿瘤预后的预测;Optionally, the tumor grade refers to the judgment of tumor malignancy and the prediction of tumor prognosis;
可选的,所述肿瘤分级分为高风险组和低风险组。Optionally, the tumor grades are divided into high-risk group and low-risk group.
可选的,所述产品包括用于检测所述基因组合中基因的基因类型的引物、探针、试剂、试剂盒、基因芯片或检测系统。Optionally, the product includes primers, probes, reagents, kits, gene chips or detection systems for detecting the genotypes of the genes in the gene combination.
可选的,所述产品为针对基因集A、基因片断集B和基因片段集C中基因的外显子和相关内含子区域进行检测。Optionally, the product is for detection of exons and related intron regions of genes in gene set A, gene fragment set B and gene fragment set C.
可选的,所述肿瘤分级的方法包括如下步骤:Optionally, the method for grading the tumor comprises the following steps:
步骤S1:评估所述癌细胞组织中的基因集A中所包含基因的基因突变,评估癌细胞组织中的基因片段集B和基因片段集C的基因拷贝数变异;Step S1: Evaluate the gene mutation of the genes contained in the gene set A in the cancer cell tissue, and evaluate the gene copy number variation of the gene fragment set B and the gene fragment set C in the cancer cell tissue;
步骤S2:基于步骤S1的评估结果,判断癌症恶性程度并进行肿瘤预后预测。Step S2: Based on the evaluation results of step S1, the degree of malignancy of the cancer is judged and the prognosis of the tumor is predicted.
可选的,所述的基因突变包括碱基置换突变、缺失突变、插入突变和/或融合突变,所述基因拷贝数变异包括基因拷贝数增加和/或基因拷贝数减少。Optionally, the gene mutation includes base substitution mutation, deletion mutation, insertion mutation and/or fusion mutation, and the gene copy number variation includes gene copy number increase and/or gene copy number decrease.
可选的,所述步骤S1中,通过比较所述肿瘤组织与正常组织的测序数据,用于评估所述基因集A中包含基因的基因突变,同时评估所述基因片段集B和基因片段集C的基因拷贝数变异。Optionally, in the step S1, by comparing the sequencing data of the tumor tissue and the normal tissue, it is used to evaluate the gene mutation of the genes contained in the gene set A, and simultaneously evaluate the gene fragment set B and the gene fragment set Gene copy number variation in C.
可选的,所述步骤S2中,如果基因集A中至少一个基因出现基因突变,或基因片段集B中至少一个片段出现基因拷贝数减少,或基因片段集C中至少一个片段出现基因拷贝数增加,所述肿瘤分级为低风险组;反之,即基因集A中没有基因出现基因突变或拷贝数变异,同时基因片段集B中没有任何片段出现基因拷贝数减少,同时基因片段集C中没有任何片段出现基因拷贝数增加,所述肿瘤分级为高风险组。Optionally, in the step S2, if at least one gene in the gene set A has a gene mutation, or at least one segment in the gene segment set B has a gene copy number reduction, or at least one segment in the gene segment set C has a gene copy number increase, the tumor is graded as a low-risk group; on the contrary, there is no gene mutation or copy number variation in gene set A, and at the same time, there is no gene copy number decrease in any fragment in gene fragment set B, and there is no gene fragment number in gene fragment set C Gene copy number gain in any segment was graded as a high-risk group.
可选的,可以单独使用基因集A中的基因突变情况,不使用基因片段集B和基因片段集C的基因拷贝数情况,对肿瘤恶性程度进行分级和肿瘤预后预测。Optionally, the gene mutation status in gene set A can be used alone, without using the gene copy number status of gene fragment set B and gene fragment set C, to grade tumor malignancy and predict tumor prognosis.
可选的,所述步骤S2中,如果基因集A中至少一个基因出现基因突变,所述肿瘤分级为低风险组;反之,即基因集A中没有基因出现基因突变,所述肿瘤分级为高风险组。Optionally, in the step S2, if there is a gene mutation in at least one gene in the gene set A, the tumor is graded as a low-risk group; otherwise, that is, no gene in the gene set A has a gene mutation, and the tumor is graded as a high risk group risk group.
可选的,从所述基因组合中选择任意基因片段进行组合,形成新的基因组合,使用相同的肿瘤分级的方法对肿瘤恶性程度进行分级和肿瘤预后预测。Optionally, any gene fragments are selected from the gene combination and combined to form a new gene combination, and the same tumor grading method is used to grade tumor malignancy and predict tumor prognosis.
本发明还提供了使用上述基因组合检测人肿瘤分级的方法。The present invention also provides a method for detecting human tumor grade using the above-mentioned gene combination.
可选地,所述的肿瘤为泌尿系统肿瘤或泛癌;Optionally, the tumor is a tumor of the urinary system or pan-cancer;
可选的,所述泌尿系统肿瘤为泌尿系统恶性肿瘤;Optionally, the tumor of the urinary system is a malignant tumor of the urinary system;
可选的,所述泌尿系统肿瘤为尿路上皮癌;Optionally, the tumor of the urinary system is urothelial carcinoma;
可选的,所述泛癌为TCGA泛癌数据中的癌种。Optionally, the pan-cancer is a cancer type in TCGA pan-cancer data.
可选地,所述的肿瘤分级是指肿瘤恶性程度判断和肿瘤预后的预测,用于指导临床诊疗;Optionally, the tumor grade refers to the judgment of tumor malignancy and the prediction of tumor prognosis, which is used to guide clinical diagnosis and treatment;
可选的,所述肿瘤分级分为高风险组和低风险组。Optionally, the tumor grades are divided into high-risk group and low-risk group.
可选地,所述方法包括使用检测所述基因组合中基因的基因类型的引物、探针、试剂、试剂盒、基因芯片或检测系统。Optionally, the method includes using primers, probes, reagents, kits, gene chips or detection systems for detecting the genotypes of the genes in the gene combination.
可选地,所述方法为针对基因集A、基因片段集B和基因片段集C中基因的外显子和相关内含子区域进行检测。Optionally, the method is to detect exons and related intron regions of genes in gene set A, gene fragment set B and gene fragment set C.
可选地,所述肿瘤分级的方法包括如下步骤:Optionally, the method for grading the tumor comprises the following steps:
步骤S1:评估所述癌细胞组织中的基因集A中所包含基因的基因突变,评估癌细胞组织中的基因片段集B和基因片段集C的基因拷贝数变异;Step S1: Evaluate the gene mutation of the genes contained in the gene set A in the cancer cell tissue, and evaluate the gene copy number variation of the gene fragment set B and the gene fragment set C in the cancer cell tissue;
步骤S2:基于步骤S1的评估结果,判断癌症恶性程度并进行肿瘤预后预测。Step S2: Based on the evaluation results of step S1, the degree of malignancy of the cancer is judged and the prognosis of the tumor is predicted.
可选地,所述的基因突变包括碱基置换突变、缺失突变、插入突变和/或融合突变,所述基因拷贝数变异包括基因拷贝数增加和/或基因拷贝数减少。Optionally, the gene mutation includes base substitution mutation, deletion mutation, insertion mutation and/or fusion mutation, and the gene copy number variation includes gene copy number increase and/or gene copy number decrease.
可选地,在步骤S1中,通过比较所述肿瘤组织与正常组织的测序数据,用于评估所述基因集A中包含基因的基因突变,同时评估所述基因片段集B和基因片段集C的基因拷贝数变异。Optionally, in step S1, by comparing the sequencing data of the tumor tissue and normal tissue, it is used to evaluate the gene mutation of the genes contained in the gene set A, and simultaneously evaluate the gene fragment set B and the gene fragment set C gene copy number variation.
可选地,所述步骤S2中,如果基因集A中至少一个基因出现基因突变,或基因片段集B中至少一个片段出现基因拷贝数减少,或基因片段集C中至少一个片段出现基因拷贝数增加,所述肿瘤分级为低风险组;反之,即基因集A中没有基因出现基因突变,同时基因片段集B中没有任何片段出现基因拷贝数减少,同时基因片段集C中没有任何片段出现基因拷贝数增加,所述肿瘤分级为高风险组。Optionally, in the step S2, if at least one gene in the gene set A has a gene mutation, or at least one segment in the gene segment set B has a gene copy number reduction, or at least one segment in the gene segment set C has a gene copy number increase, the tumor is graded as a low-risk group; on the contrary, there is no gene mutation in gene set A, and at the same time, there is no gene copy number reduction in any fragment in gene fragment set B, and there is no gene in any fragment in gene fragment set C. Copy number gain, the tumor was graded as high risk group.
可选地,所述步骤S2中,如果基因集A中至少一个基因出现基因突变,所述肿瘤分级为低风险组;反之,即基因集A中没有基因出现基因突变,所述肿瘤分级为高风险组。Optionally, in the step S2, if there is a gene mutation in at least one gene in the gene set A, the tumor is graded as a low-risk group; otherwise, that is, no gene in the gene set A has a gene mutation, and the tumor is graded as a high risk group. risk group.
可选地,从所述基因组合中选择任意基因片段进行组合,形成新的基因组合,使用相同的肿瘤分级的方法对肿瘤恶性程度进行分级和肿瘤预后预测,从而指导临床诊疗。Optionally, select any gene fragments from the gene combination to form a new gene combination, and use the same tumor grading method to grade tumor malignancy and predict tumor prognosis, so as to guide clinical diagnosis and treatment.
本申请技术方案,具有如下优点:The technical solution of the present application has the following advantages:
1.本申请的所述检测基因组合是从北京大学第一医院的实际尿路上皮癌病例的高通量测序数据中,通过特定的配对聚类分析得来,来源于真实的数据具有更高的可靠性和可信度,可以准确针对尿路上皮癌、泛癌进行恶性程度分级和预后预测。1. The detection gene combination in this application is obtained from the high-throughput sequencing data of actual urothelial cancer cases in Peking University First Hospital through specific paired cluster analysis, and the real data has higher The reliability and credibility of the system can accurately grade the degree of malignancy and predict the prognosis of urothelial carcinoma and pan-cancer.
2.本申请所述基因组合包括基因组合具有多元性,可以从中优选出多种基因组合用于尿路上皮癌、泛癌恶性程度的判断,用于不同的临床情况。2. The gene combination described in this application includes the diversity of the gene combination, from which a variety of gene combinations can be optimized for judging the malignancy of urothelial carcinoma and pan-cancer, and for different clinical situations.
3.相比于全外显子测序,本申请针对特定的基因和DNA片段进行靶向测序分析,在相同的成本前提下可以明显提高测序深度和精准度,在相同测序深度和精准度的前提下,可以明显节约成本,普适性广。3. Compared with whole exome sequencing, this application conducts targeted sequencing analysis for specific genes and DNA fragments, which can significantly improve the sequencing depth and accuracy at the same cost. In this case, the cost can be significantly saved, and the universality is wide.
4.相比于WHO尿路上皮癌病理分级系统,本申请完全不受病理医生的主观印象影响,具有极佳的客观性和可信度。4. Compared with the WHO urothelial carcinoma pathological grading system, this application is not affected by the subjective impression of pathologists at all, and has excellent objectivity and credibility.
附图说明Description of drawings
为了更清楚地说明本申请具体实施方式或现有技术中的技术方案,下面将对具体实施方式或现有技术描述中所需要使用的附图作简单地介绍,显而易见地,下面描述中的附图是本申请的一些实施方式,对于本领域普通技术人员来讲,在不付出创造性劳动的前提下,还可以根据这些附图获得其他的附图。In order to more clearly illustrate the specific embodiments of the present application or the technical solutions in the prior art, the following will briefly introduce the accompanying drawings that need to be used in the description of the specific embodiments or prior art. Obviously, the accompanying drawings in the following description The drawings are some implementations of the present application, and those skilled in the art can obtain other drawings based on these drawings without creative work.
图1是本申请实验例1中利用本申请实施例1中的基因组合进行尿路上皮癌恶性程度分级标准分级后,将肿瘤特异性生存作为主要终点的Kaplan-Meier生存分析图;Fig. 1 is the Kaplan-Meier survival analysis graph with tumor-specific survival as the primary endpoint after the urothelial carcinoma malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 1 of the present application;
图2是本申请实验例1中利用本申请实施例1中的基因组合进行尿路上皮癌恶性程度分级标准分级后,将肿瘤无进展生存作为主要终点的Kaplan-Meier生存分析图;Fig. 2 is a Kaplan-Meier survival analysis chart with tumor progression-free survival as the primary endpoint after the urothelial carcinoma malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 1 of the present application;
图3是本申请实验例1中利用本申请实施例1中的基因组合进行尿路上皮癌恶性程度分级标准分级后,将肿瘤无病生存作为主要终点的Kaplan-Meier生存分析图;Fig. 3 is a Kaplan-Meier survival analysis chart with tumor disease-free survival as the primary endpoint after the urothelial carcinoma malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 1 of the present application;
图4是本申请实验例1中利用本申请实施例1中的基因组合进行尿路上皮癌恶性程度分级标准分级后,将总生存作为主要终点的Kaplan-Meier生存分析图;Fig. 4 is a Kaplan-Meier survival analysis chart with overall survival as the primary endpoint after the urothelial carcinoma malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 1 of the present application;
图5是本申请实验例2中利用本申请基因组合1进行尿路上皮癌恶性程度分级标准分级后,将肿瘤特异性生存作为主要终点的Kaplan-Meier生存分析图。Fig. 5 is a graph of Kaplan-Meier survival analysis with tumor-specific survival as the primary end point after using the gene combination 1 of the present application to grade the malignancy of urothelial carcinoma in Experimental Example 2 of the present application.
图6是本申请实验例2中利用本申请基因组合1进行尿路上皮癌恶性程度分级标准分级后,将肿瘤无病生存作为主要终点的Kaplan-Meier生存分析图。Fig. 6 is a graph of Kaplan-Meier survival analysis with tumor disease-free survival as the primary endpoint after grading the malignancy of urothelial carcinoma using gene combination 1 of the present application in Experimental Example 2 of the present application.
图7是本申请实验例2中利用本申请基因组合1进行尿路上皮癌恶性程度分级标准分级后,将总生存作为主要终点的Kaplan-Meier生存分析图。Fig. 7 is a graph of Kaplan-Meier survival analysis with the overall survival as the primary endpoint after the urothelial carcinoma malignancy grading standard was graded using the gene combination 1 of the present application in Experimental Example 2 of the present application.
图8是本申请实验例3中利用本申请基因集A进行尿路上皮癌恶性程度分级标准分级后,将肿瘤特异性生存作为主要终点的Kaplan-Meier生存分析图。Fig. 8 is a graph of Kaplan-Meier survival analysis with tumor-specific survival as the primary end point after using the gene set A of the present application to grade the malignant degree of urothelial carcinoma in Experimental Example 3 of the present application.
图9是本申请实验例3中利用本申请基因集A进行尿路上皮癌恶性程度分级标准分级后,将肿瘤无进展生存作为主要终点的Kaplan-Meier生存分析图。Fig. 9 is a graph of Kaplan-Meier survival analysis with tumor progression-free survival as the primary endpoint after grading the malignancy of urothelial carcinoma using the gene set A of the present application in Experimental Example 3 of the present application.
图10是本申请实验例3中利用本申请基因集A进行尿路上皮癌恶性程度分级标准分级后,将肿瘤无病生存作为主要终点的Kaplan-Meier生存分析图。Fig. 10 is a graph of Kaplan-Meier survival analysis with tumor disease-free survival as the primary endpoint after grading the malignancy of urothelial carcinoma using the gene set A of the present application in Experimental Example 3 of the present application.
图11是本申请实验例3中利用本申请基因集A进行尿路上皮癌恶性程度分级标准分级后,将总生存作为主要终点的Kaplan-Meier生存分析图。Fig. 11 is a graph of Kaplan-Meier survival analysis with overall survival as the primary endpoint after grading the malignancy of urothelial carcinoma using the gene set A of the present application in Experimental Example 3 of the present application.
图12是本申请实验例4中利用本申请实施例1中的基因组合进行泛癌恶性程度分级标准分级后,将肿瘤无病生存作为主要终点的Kaplan-Meier生存分析图;Fig. 12 is a Kaplan-Meier survival analysis graph with tumor disease-free survival as the primary endpoint after the pan-cancer malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 4 of the present application;
图13是本申请实验例4中利用本申请实施例1中的基因组合进行泛癌恶性程度分级标准分级后,将肿瘤无进展生存作为主要终点的Kaplan-Meier生存分析图;Fig. 13 is a Kaplan-Meier survival analysis chart with tumor progression-free survival as the primary endpoint after using the gene combination in Example 1 of the present application in Experimental Example 4 of the present application to perform pan-cancer malignancy grading standard grading;
图14是本申请实验例4中利用本申请实施例1中的基因组合进行泛癌恶性程度分级标准分级后,将总生存作为主要终点的Kaplan-Meier生存分析图;Figure 14 is a Kaplan-Meier survival analysis chart with overall survival as the primary endpoint after the pan-cancer malignancy grading standard is graded using the gene combination in Example 1 of the present application in Experimental Example 4 of the present application;
图15是本申请实验例5中利用本申请基因组合1进行泛癌恶性程度分级标准分级后,将肿瘤特异性生存作为主要终点的Kaplan-Meier生存分析图;Fig. 15 is a Kaplan-Meier survival analysis chart with tumor-specific survival as the primary endpoint after the pan-cancer malignancy grading standard is graded using the gene combination 1 of the present application in Experimental Example 5 of the present application;
图16是本申请实验例5中利用本申请基因组合1进行泛癌恶性程度分级标准分级后,将肿瘤无进展生存作为主要终点的Kaplan-Meier生存分析图;Fig. 16 is a Kaplan-Meier survival analysis graph with tumor progression-free survival as the primary endpoint after the pan-cancer malignancy grading standard grading was performed using the gene combination 1 of the present application in Experimental Example 5 of the present application;
图17是本申请实验例5中利用本申请基因组合1进行泛癌恶性程度分级标准分级后,将肿瘤无病生存作为主要终点的Kaplan-Meier生存分析图;Fig. 17 is a Kaplan-Meier survival analysis graph with tumor disease-free survival as the primary endpoint after the pan-cancer malignancy grading standard is graded using the gene combination 1 of the present application in Experimental Example 5 of the present application;
图18是本申请实验例5中利用本申请基因组合1进行泛癌恶性程度分级标准分级后,将总生存作为主要终点的Kaplan-Meier生存分析图;Fig. 18 is a Kaplan-Meier survival analysis chart with overall survival as the primary endpoint after the pan-cancer malignancy grading standard is graded using the gene combination 1 of the present application in Experimental Example 5 of the present application;
图19是本申请实验例6中利用本申请基因集A进行泛癌恶性程度分级标准分级后,将肿瘤特异性生存作为主要终点的Kaplan-Meier生存分析图。Fig. 19 is a graph of Kaplan-Meier survival analysis with tumor-specific survival as the primary endpoint after using the gene set A of the present application to perform pan-cancer malignancy grading standard grading in Experimental Example 6 of the present application.
图20是本申请实验例6中利用本申请基因集A进行泛癌恶性程度分级标准分级后,将肿瘤无进展生存作为主要终点的Kaplan-Meier生存分析图。Fig. 20 is a graph of Kaplan-Meier survival analysis with tumor progression-free survival as the primary endpoint after using the gene set A of the present application to perform pan-cancer malignancy grading standard grading in Experimental Example 6 of the present application.
图21是本申请实验例6中利用本申请基因集A进行泛癌恶性程度分级标准分级后,将肿瘤无病生存作为主要终点的Kaplan-Meier生存分析图。Fig. 21 is a graph of Kaplan-Meier survival analysis with tumor disease-free survival as the primary endpoint after using the gene set A of the present application to perform pan-cancer malignancy grading standard grading in Experimental Example 6 of the present application.
图22是本申请实验例6中利用本申请基因集A进行泛癌恶性程度分级标准分级后,将总生存作为主要终点的Kaplan-Meier生存分析图。Fig. 22 is a graph of Kaplan-Meier survival analysis with overall survival as the primary endpoint after using the gene set A of the present application to perform pan-cancer malignancy grading standard grading in Experimental Example 6 of the present application.
具体实施方式Detailed ways
提供下述实施例是为了更好地进一步理解本申请,并不局限于所述最佳实施方式,不对本申请的内容和保护范围构成限制,任何人在本申请的启示下或是将本申请与其他现有技术的特征进行组合而得出的任何与本申请相同或相近似的产品,均落在本申请的保护范围之内。The following examples are provided in order to further understand the application better, and are not limited to the best implementation mode, and do not limit the content and protection scope of the application. Anyone under the inspiration of the application or the application Any product identical or similar to the present application obtained by combining features of other prior art falls within the protection scope of the present application.
实施例中未注明具体实验步骤或条件者,按照本领域内的文献所描述的常规实验步骤的操作或条件即可进行。所用试剂或仪器未注明生产厂商者,均为可以通过市购获得的常规试剂产品。If no specific experimental steps or conditions are indicated in the examples, it can be carried out according to the operation or conditions of the conventional experimental steps described in the literature in this field. The reagents or instruments used, whose manufacturers are not indicated, are all commercially available conventional reagent products.
实施例1用于人肿瘤分级的基因组合(Panel)Embodiment 1 is used for the gene combination (Panel) of human tumor classification
本申请主要利用北京大学第一医院尿路上皮癌外显子测序高通量数据库进行筛选,确认了一种用于人肿瘤分级的基因组合(panel),该基因组合包括基因集A、基因片段集B和基因片段集C;This application mainly uses Peking University First Hospital urothelial carcinoma exome sequencing high-throughput database for screening, and confirmed a gene combination (panel) for human tumor grading, the gene combination includes gene set A, gene fragment Set B and gene fragment set C;
所述的基因集A包括:ACVR1B、ATP4B、AZGP1、BRAF、BRCA1、BRCA2、CRYZL1、DCTN1、E2F3、EGFR、ERBB2、ERCC2、ESPL1、FGFR3、FKBP6、GZMM、H3-5、IGLL5、IRF7、METTL24、MORN5、MTOR、NPY1R、PET100、PIK3CA、PPARG、PTPN11、PTX4、RGPD8、SERPINA12、STAG2和ZNF141中至少一个;The gene set A includes: ACVR1B, ATP4B, AZGP1, BRAF, BRCA1, BRCA2, CRYZL1, DCTN1, E2F3, EGFR, ERBB2, ERCC2, ESPL1, FGFR3, FKBP6, GZMM, H3-5, IGLL5, IRF7, METTL24, At least one of MORN5, MTOR, NPY1R, PET100, PIK3CA, PPARG, PTPN11, PTX4, RGPD8, SERPINA12, STAG2 and ZNF141;
所述的基因片段集B包括:chr1:155289001-155911999、chr1:226259001-226736999、chr8:30515001-30892999、chr8:42065001-43147999、chr9:134612001-135906999、chr11:48509001-55112999、chr14:24768001-105145999、chr16:1-73083999、chr18:1-319999和chr18:15325001-19749999中至少一个;所述基因片段集B中基因片段位置以GRCh37为标准进行注释,在GRCh38或未来出现的新版人类参考基因组中,其数字可能发生改变,但指向的客观片段位置和可用于检测的基因不会发生改变;所述的基因片段集B包括:chr1:155289001-155911999、chr1:226259001-226736999、chr8:30515001-30892999、chr8:42065001-43147999、chr9:134612001-135906999、chr11:48509001-55112999、chr14:24768001-105145999 , at least one of chr16:1-73083999, chr18:1-319999 and chr18:15325001-19749999; the position of the gene segment in the gene segment set B is annotated with GRCh37 as the standard, in GRCh38 or the new version of the human reference genome that will appear in the future , its number may change, but the targeted fragment position and the genes available for detection will not change;
所述的基因片段集C包括:chr3:12030001-12639999、chr17:2293001-2306999和chr17:37830001-37979999中至少一个;所述基因片段集C中基因片段位置以GRCh37为标准进行注释,在GRCh38或未来出现的新版人类参考基因组中,其数字可能发生改变,但指向的客观片段位置和可用于检测的基因不会发生改变;The gene fragment set C includes: at least one of chr3: 12030001-12639999, chr17: 2293001-2306999 and chr17: 37830001-37979999; the position of the gene fragment in the gene fragment set C is annotated with GRCh37 as the standard, and in GRCh38 or In the new version of the human reference genome that will appear in the future, its number may change, but the pointed objective fragment position and the genes that can be used for detection will not change;
可选的,所述的基因片段集B中包括的详细基因如下表:Optionally, the detailed genes included in the gene fragment set B are as follows:
表1基因片段集BTable 1 Gene Fragment Set B
Figure PCTCN2022116390-appb-000007
Figure PCTCN2022116390-appb-000007
Figure PCTCN2022116390-appb-000008
Figure PCTCN2022116390-appb-000008
Figure PCTCN2022116390-appb-000009
Figure PCTCN2022116390-appb-000009
Figure PCTCN2022116390-appb-000010
Figure PCTCN2022116390-appb-000010
Figure PCTCN2022116390-appb-000011
Figure PCTCN2022116390-appb-000011
可选的,所述的基因片段集C中包括的详细基因如下表:Optionally, the detailed genes included in the gene fragment set C are as follows:
表2基因片段集CTable 2 Gene Fragment Set C
Figure PCTCN2022116390-appb-000012
Figure PCTCN2022116390-appb-000012
Figure PCTCN2022116390-appb-000013
Figure PCTCN2022116390-appb-000013
实施例2一种用于人尿路上皮癌恶性程度分级和预后预测的方法Example 2 A method for grading the degree of malignancy and predicting prognosis of human urothelial carcinoma
本实施例提供了一种用于人肿瘤分级检测的的方法,包括,利用实施例1中的基因组合(panel)进行人尿路上皮癌恶性程度分级和预后预测,具体步骤如下:This embodiment provides a method for detecting the grade of human tumors, including using the gene panel in Embodiment 1 to grade the malignancy of human urothelial carcinoma and predict the prognosis, and the specific steps are as follows:
(1)取尿路上皮癌组织和健康对照组织标本,所述尿路上皮癌组织标本可以是尿路上皮癌细胞系、新鲜尿路上皮癌标本、冰冻尿路上皮癌标本或石蜡包埋尿路上皮癌标本;健康对照组织可以是已知公认健康人的组织,也可以是尿路上皮癌患者本人的癌旁组织或外周血。在本实施例中选择石蜡包埋的尿路上皮癌标本,健康对照组织使用的是癌旁正常组织,通过常规方法提取DNA,通过常规方法构建文库,最终使用实施例1中的基因组合(panel)进行靶向高通量测序,比较尿路上皮癌组织与健康组织的测序数据,得到所述尿路上皮癌组织的基因组合中基因集A各个基因的基因突变(Mutation)情况,以及基因片段集B和基因片段集C中各个基因的拷贝数变异(CNV)情况。(1) Take urothelial carcinoma tissue and healthy control tissue specimens, the urothelial carcinoma tissue specimens can be urothelial carcinoma cell lines, fresh urothelial carcinoma specimens, frozen urothelial carcinoma specimens or paraffin-embedded urine Urothelial carcinoma specimens; healthy control tissues can be tissues from known healthy people, or paracancerous tissues or peripheral blood from urothelial carcinoma patients themselves. In this embodiment, the paraffin-embedded urothelial carcinoma specimens were selected, and the healthy control tissue was normal paracancerous tissue, DNA was extracted by conventional methods, and a library was constructed by conventional methods, and finally the gene combination (panel ) to perform targeted high-throughput sequencing, compare the sequencing data of urothelial cancer tissue and healthy tissue, and obtain the gene mutation (Mutation) of each gene of gene set A in the gene combination of the urothelial cancer tissue, and the gene fragment Copy number variation (CNV) of each gene in set B and gene fragment set C.
所述的基因突变包括碱基置换突变、缺失突变、插入突变和融合突变,所述基因拷贝数变异包括基因拷贝数增加和基因拷贝数减少。The gene mutation includes base substitution mutation, deletion mutation, insertion mutation and fusion mutation, and the gene copy number variation includes gene copy number increase and gene copy number decrease.
(2)基于步骤(1)中获得的尿路上皮癌组织的基因组合中各基因的突变和/或变异情况进行判断:(2) Judgment based on the mutation and/or variation of each gene in the gene combination of the urothelial carcinoma tissue obtained in step (1):
如果存在基因集A中至少一个基因的基因突变,或存在基因片段集B中至少一个区域基因拷贝数减少,或存在基因片段集C中至少一个区域基因拷贝数增加,所述的尿路上皮癌患者为低风险组,肿瘤恶性程度更低,具有更好的肿瘤预后;反之,基因集A中没有基因出现基因突变,同时基因片段集B中没有任何片段出现基因拷贝数减少,同时基因片段集C中没有任何片段出现基因拷贝数增加,所述的尿路上皮癌患者为高风险组,肿瘤恶性程度更高,具有较差的肿瘤预后。If there is a gene mutation in at least one gene in gene set A, or there is a gene copy number decrease in at least one region in gene fragment set B, or there is a gene copy number increase in at least one region in gene fragment set C, the urothelial carcinoma Patients belong to the low-risk group, with lower tumor malignancy and better tumor prognosis; on the contrary, no genes in gene set A have gene mutations, and at the same time, no gene fragments in gene set B have gene copy number reduction, and at the same time, gene set B has In C, there is no gene copy number increase in any segment, and the urothelial carcinoma patients are in the high-risk group, with higher tumor malignancy and poor tumor prognosis.
本申请允许仅检测基因突变,无需检测拷贝数变异,即单独检测基因集A中基因的突变情况,使用相同的判断标准对尿路上皮癌恶性程度进行分级和肿瘤预后预测:如果存在基因集A中至少一个基因的基因突变,所述的尿路上皮癌患者为低风险组,肿瘤恶性程度更低,具有更好的肿瘤预后;反之,基因集A中没有基因出现基因突变,所述的尿路上皮癌患者为高风险组,肿瘤恶性程度更高,具有较差的肿瘤预后。This application allows only the detection of gene mutations without the need for detection of copy number variations, that is, the detection of gene mutations in gene set A alone, and the same criteria are used to grade the malignancy of urothelial carcinoma and predict tumor prognosis: if there is gene set A Gene mutation of at least one gene in gene set A, the urothelial cancer patients are in the low-risk group, the tumor malignancy is lower, and the tumor prognosis is better; on the contrary, there is no gene mutation in gene set A, and the urinary tract cancer patients are in the low-risk group. Patients with roadside carcinoma belong to the high-risk group, with higher tumor malignancy and poor prognosis.
实施例3Example 3
作为实施例2的可替换的实施方式,在本申请中,允许对实施例1中的基因组合(panel)中的基因进行挑选并重新组合,形成新的基因组合,评判标准为,从基因集A中挑选出来的基因,则其中至少一个基因的基因突变,表明所述的尿路上皮癌患者为低风险组;从基因片段集B中挑选出来的基因片段,则其中至少一个区域基因拷贝数减少,表明所述的尿路上皮癌患者为低风险组;从基因片段集C中挑选出来的基因片段,则其中至少一个区域基因拷贝数增加,表明所述的尿路上皮癌患者为低风险组;反之,基因集A中挑选出的基因中没有出现基因突变或拷贝数变异,同时基因片段集B中挑选出来的片段中没有出现拷贝数减少,同时基因片断集C中挑选出来的片段中没有出现拷贝数增加,所述尿路上皮癌患者为高风险组。As an alternative implementation of Example 2, in this application, the genes in the gene combination (panel) in Example 1 are allowed to be selected and recombined to form a new gene combination. The judging criteria are, from the gene set For the genes selected in A, the gene mutation of at least one of them indicates that the urothelial cancer patients are in the low-risk group; for the gene fragments selected from gene fragment set B, the gene copy number of at least one region decrease, indicating that the patients with urothelial carcinoma are in the low-risk group; for the gene fragments selected from the gene fragment set C, at least one region of the gene copy number increases, indicating that the patients with urothelial carcinoma are in the low-risk group On the contrary, there is no gene mutation or copy number variation in the genes selected in gene set A, and at the same time, there is no copy number reduction in the fragments selected in gene fragment set B, and at the same time, there is no copy number reduction in the fragments selected in gene fragment set C No copy number gain occurred, and the urothelial carcinoma patient was a high-risk group.
实施例4一种用于人泛癌(Pancancer)恶性程度分级和预后预测的方法Example 4 A method for grading and predicting the degree of malignancy of human pancancer (Pancancer)
本实施例提供了一种用于人肿瘤分级检测的的方法,包括,利用实施例1中的基因组合(panel)进行人泛癌(Pancancer)恶性程度分级和预后预测,具体步骤如下:This embodiment provides a method for human tumor grading detection, including, using the gene combination (panel) in Example 1 to perform human pancancer (Pancancer) malignancy grading and prognosis prediction, and the specific steps are as follows:
(1)取泛癌(此处泛癌定义为TCGA泛癌数据中所有癌种,包括肾上腺癌,尿路上皮癌,乳腺癌,宫颈癌,胆管癌,结肠癌,淋巴瘤,食管癌,胶质母细胞瘤,头颈鳞状细胞癌,肾嫌色细胞癌,肾透明细胞癌,肾乳头状细胞癌,白血病,脑胶质瘤,肝细胞肝癌,肺腺癌,肺鳞癌,间皮瘤,卵巢浆液性囊腺癌,胰腺癌,嗜铬细胞瘤与副神经节瘤,前列腺癌,直肠癌,肉瘤,皮肤黑色素瘤,胃癌,睾丸癌,甲状腺癌,胸腺癌,子宫内膜癌,子宫肉瘤,葡萄膜黑色素瘤,本实施例中所提泛癌均定义为此,不再重复叙述)组织和健康对照组织标本,所述泛癌组织标本可以是泛癌细胞系、新鲜泛癌标本、冰冻泛癌标本或石蜡包埋泛癌标本;健康对照组织可以是已知公认健康人的组织,也可以是泛癌患者本人的癌旁组织或外周血。在本实施例中选择石蜡包埋的泛癌标本,健康对照组织使用的是癌旁正常组织,通过常规方法提取DNA,通过常规方法构建文库,最终使用实施例1中的基因组合(panel)进行靶向高通量测序,比较泛癌组织与健康组织的测序数据,得到所述泛癌组织的基因组合中基因集A各个基因的基因突变(Mutation)情况,以及基因片段集B和基因片段集C中各个片段的拷贝数变异(CNV)情况。(1) Take pan-cancer (here pan-cancer is defined as all cancer types in TCGA pan-cancer data, including adrenal cancer, urothelial cancer, breast cancer, cervical cancer, bile duct cancer, colon cancer, lymphoma, esophageal cancer, gum Glioblastoma, head and neck squamous cell carcinoma, chromophobe renal cell carcinoma, clear cell renal cell carcinoma, renal papillary cell carcinoma, leukemia, glioma, hepatocellular carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma , ovarian serous cystadenocarcinoma, pancreatic cancer, pheochromocytoma and paraganglioma, prostate cancer, rectal cancer, sarcoma, skin melanoma, gastric cancer, testicular cancer, thyroid cancer, thymus cancer, endometrial cancer, uterine Sarcoma, uveal melanoma, the pan-cancer mentioned in this embodiment are all defined as this, and will not be repeated) tissue and healthy control tissue specimens, the pan-cancer tissue specimens can be pan-cancer cell lines, fresh pan-cancer specimens, Frozen pan-cancer specimens or paraffin-embedded pan-cancer specimens; healthy control tissues can be tissues from known healthy people, or paracancerous tissues or peripheral blood of pan-cancer patients themselves. In this example, paraffin-embedded pan-cancer specimens were selected, and normal paracancerous tissues were used as healthy control tissues. DNA was extracted by conventional methods, and libraries were constructed by conventional methods. Finally, the gene combination (panel) in Example 1 was used to carry out Targeted high-throughput sequencing, compare the sequencing data of pan-cancer tissue and healthy tissue, and obtain the gene mutation (Mutation) of each gene in gene set A in the gene combination of the pan-cancer tissue, as well as gene fragment set B and gene fragment set Copy number variation (CNV) status of each fragment in C.
所述的基因突变包括碱基置换突变、缺失突变、插入突变和融合突变,所述基因拷贝数变异包括基因拷贝数增加和基因拷贝数减少。The gene mutation includes base substitution mutation, deletion mutation, insertion mutation and fusion mutation, and the gene copy number variation includes gene copy number increase and gene copy number decrease.
(2)基于步骤(1)中获得的泛癌组织的基因组合中各基因的突变和/或变异情况进行判断:(2) Judgment based on the mutation and/or variation of each gene in the gene combination of the pan-cancer tissue obtained in step (1):
如果存在基因集A中至少一个基因的基因突变,或存在基因片段集B中至少一个区域基因拷贝数减少,或存在基因片段集C中至少一个区域基因拷贝数增加,所述的泛癌患者为低风险组,具有更好的肿瘤预后;反之,基因集A中没有基因出现基因突变,同时基因片段集B中没有任何片段出现基因拷贝数减少,同时基因片段集C中没有任何片段出现基因拷贝数增加,所述的泛癌患者为高风险组,具有较差的肿瘤预后。If there is a gene mutation of at least one gene in gene set A, or there is a gene copy number decrease in at least one region in gene fragment set B, or there is an increase in gene copy number in at least one region in gene fragment set C, the pan-cancer patient is The low-risk group has a better tumor prognosis; on the contrary, there is no gene mutation in gene set A, and there is no gene copy number reduction in any fragment in gene fragment set B, and there is no gene copy in any fragment in gene fragment set C The number of patients with pan-cancer increases, and the pan-cancer patients are a high-risk group with poor tumor prognosis.
本申请允许仅检测基因突变,无需检测拷贝数变异,即单独检测基因集A中基因的突变情况,使用相同的判断标准对泛癌恶性程度进行分级和肿瘤预后预测:如果存在基因集A中至少一个基因的基因突变,所述的泛癌患者为低风险组,肿瘤恶性程度更低,具有更好的肿瘤预后;反之,基因集A中没有基因出现基因突变,所述的泛癌患者为高风险组,肿瘤恶性程度更高,具有较差的肿瘤预后。This application allows the detection of gene mutations only, without the need to detect copy number variations, that is, the mutations of genes in gene set A are detected separately, and the same criteria are used to grade the degree of malignancy of pan-cancer and predict tumor prognosis: if there is at least If there is a gene mutation in one gene, the pan-cancer patients are in the low-risk group, the tumor malignancy is lower, and they have better tumor prognosis; on the contrary, if there is no gene mutation in gene set A, the pan-cancer patients are in the high-risk group. The risk group, with higher tumor malignancy, has poorer tumor prognosis.
实施例5Example 5
作为实施例4的可替换的实施方式,在本申请中,允许对实施例1中的基因组合(panel)中的基因进行挑选并重新组合,形成新的基因组合,评判标准为,从基因集A中挑选出来的基因,则其中至少一个基因的基因突变,表明所述的泛癌患者为低风险组;从基因片段集B中挑选出来的基因片段,则其中至少一个区域基因拷贝数减少,表明所述的泛癌患者为低风险组;从基因片段集C中挑选出来的基因片段,则其中至少一个区域基因拷贝数增加,表明所述的泛癌患者为低风险组。反之,基因集A中挑选出的基因中没有出现基因突变,同时基因片段集B中挑选出来的片段中没有出现拷贝数减少,同时基因片段集C中挑选出来的片段中没有出现拷贝数增加,所述泛癌患者为高风险组。As an alternative implementation of Example 4, in this application, the genes in the gene combination (panel) in Example 1 are allowed to be selected and recombined to form a new gene combination. The judging criteria are: from the gene set For the genes selected in A, the gene mutation of at least one gene indicates that the pan-cancer patients are in the low-risk group; for the gene fragments selected from the gene fragment set B, the copy number of at least one region of the gene is reduced, It indicates that the pan-cancer patients belong to the low-risk group; if the gene fragments selected from the gene fragment set C have an increased gene copy number in at least one region, it indicates that the pan-cancer patients belong to the low-risk group. On the contrary, there is no gene mutation in the genes selected in gene set A, and at the same time, there is no copy number decrease in the fragments selected in gene fragment set B, and there is no copy number increase in the fragments selected in gene fragment set C, The pan-cancer patients are a high-risk group.
实验例1用于人肿瘤分级的基因组合和检测方法在评估人尿路上皮癌恶性程度分级和预后预测的可行性验证Experimental Example 1 Feasibility verification of the gene combination and detection method used for human tumor grading in evaluating the malignancy grading and prognosis prediction of human urothelial carcinoma
尿路上皮癌是尿路上皮肿瘤最常见的病理类型,占到全部尿路上皮癌的90%以上,TCGA(PanCancer Atlas)膀胱尿路上皮癌细胞癌数据库是全球公认的膀胱尿路上皮癌数据库,该数据库于2020年进行了更新,同时合并Memorial Sloan Kettering Cancer Center(MSKCC)膀胱尿路上皮癌数据后,形成MSK/TCGA2020膀胱尿路上皮癌数据库并向社会公开。MSK 2015上尿路尿路上皮癌(肾盂癌、输尿管癌)数据库是目前向社会公布的高度可信且具有完整随访数据的上尿路尿路上皮癌数据库。将上述数据库合并后,可用其检验本申请用于评估尿路上皮癌恶性程度分级和预后预测的可行性和可信度。为描述方便,合并后新数据库在本申请中统称为MSK/TCGA数据库。Urothelial carcinoma is the most common pathological type of urothelial tumors, accounting for more than 90% of all urothelial carcinomas. TCGA (PanCancer Atlas) Bladder Urothelial Cancer Database is a globally recognized bladder urothelial cancer database , the database was updated in 2020, and after merging the data of Memorial Sloan Kettering Cancer Center (MSKCC) bladder urothelial carcinoma, the MSK/TCGA2020 bladder urothelial carcinoma database was formed and released to the public. The MSK 2015 upper urinary tract urothelial cancer (renal pelvis cancer, ureteral cancer) database is a highly reliable upper urinary tract urothelial cancer database with complete follow-up data released to the public. After merging the above databases, it can be used to test the feasibility and reliability of the application for evaluating the grade of malignancy and predicting prognosis of urothelial carcinoma. For the convenience of description, the combined new database is collectively referred to as the MSK/TCGA database in this application.
MSK/TCGA数据库共有972名尿路上皮癌患者资料,其中928名患者具有完整的基因突变和拷贝数变异数据,适用于本申请的应用条件。该数据库中861名患者具有完整的总(Overall)生存预后数据,可用于总生存预后检测;该数据库中704名患者具有完整的肿瘤无进展(Progression Free)生存预后数据,可用于肿瘤无进展生存预后检测;该数据库中391名患者具有完整的肿瘤特异性(Disease Specific)生存预后数据,可用于肿瘤特异性生存预后检测;该数据库中401名患者具有完整的肿瘤无病(Disease Free)生存数据,可用于肿瘤无病生存预后检测。The MSK/TCGA database has a total of 972 patients with urothelial carcinoma, of which 928 patients have complete gene mutation and copy number variation data, which are suitable for the application conditions of this application. 861 patients in this database have complete prognostic data of overall survival, which can be used for prognostic detection of overall survival; 704 patients in this database have complete prognostic data of tumor progression-free survival, which can be used for tumor progression-free survival Prognosis detection; 391 patients in this database have complete tumor-specific (Disease Specific) survival prognosis data, which can be used for tumor-specific survival prognosis detection; 401 patients in this database have complete tumor-specific disease-free (Disease Free) survival data , which can be used for prognostic detection of tumor disease-free survival.
按照实施例2所述的方法实施,本实验例中选取实施例1中基因集A的全部基因、基因片段集B中的所有片段和基因片段集C中的所有片段进行实施,基因片段集B和基因片段集C中实际用于检测的基因如表3。将上述928例患者进行恶性程度分级,顺利分为高风险组和低风险组,其中高风险组占比29.4%,低风险组占比70.6%,通过Kaplan-Meier生存分析,可见高风险组和低风险组的肿瘤特异性生存(图1)、肿瘤无进展生存(图2)、肿瘤无病生存(图3)和总生存(图4)均具有统计学差异并符合本申请的分组预期:低风险组具有明显更好的肿瘤特异性生存(Log-rank p值=7.065e-6)、肿瘤无进展生存(Log-rank p值=5.47e-7)、肿瘤无病生存(Log-rank p值=5.853e-5)和总生存(Log-rank p值=7.60e-13)。故使用本申请的基因组合对尿路上皮癌患者进行恶性程度分级和预后预测准确可靠。Implementation according to the method described in Example 2, in this experimental example, select all the genes in the gene set A in Example 1, all the segments in the gene fragment set B and all the fragments in the gene fragment set C for implementation, and the gene fragment set B The genes actually used for detection in gene fragment set C are shown in Table 3. The above-mentioned 928 patients were classified into high-risk group and low-risk group, and the high-risk group accounted for 29.4%, and the low-risk group accounted for 70.6%. Through the Kaplan-Meier survival analysis, it can be seen that the high-risk group and the low-risk group The tumor-specific survival (Figure 1), tumor progression-free survival (Figure 2), tumor disease-free survival (Figure 3) and overall survival (Figure 4) in the low-risk group were all statistically different and in line with the grouping expectations of this application: The low-risk group had significantly better tumor-specific survival (Log-rank p value = 7.065e-6), tumor progression-free survival (Log-rank p value = 5.47e-7), tumor disease-free survival (Log-rank p-value=5.853e-5) and overall survival (Log-rank p-value=7.60e-13). Therefore, it is accurate and reliable to use the gene combination of the present application to grade the malignant degree and predict the prognosis of patients with urothelial carcinoma.
表3实验例1中针对基因片段集B和C进行实际检测的基因The genes actually detected for the gene fragment sets B and C in the experimental example 1 of Table 3
Figure PCTCN2022116390-appb-000014
Figure PCTCN2022116390-appb-000014
Figure PCTCN2022116390-appb-000015
Figure PCTCN2022116390-appb-000015
实验例2优选的基因组合和检测方法在评估人尿路上皮癌恶性程度分级和预后预测的可行性验证Feasibility verification of the optimal gene combination and detection method in Experimental Example 2 in assessing the malignant degree grading and prognosis prediction of human urothelial carcinoma
本申请允许从基因组合(panel)中挑选任意基因片段进行组合,形成新的基因组合,使用相同的判断标准对尿路上皮癌恶性程度进行分级和肿瘤预后预测。此处从基因组合(panel)的基因集A中挑选出基因集A1(表4),从基因片段集B中挑选出基因片段集B1(表5),从基因片段集C中挑选出基因片段集C1(表5),组成基因组合1(panel 1),用于尿路上皮癌恶性程度分级和预后预测,并使用MSK/TCGA数据库进行可行性分析。同理的,判断标准为:如果存在基因集A1中至少一个基因的基因突变,或存在基因片段集B1中至少一个区域基因拷贝数减少,或存在基因片段集C1中至少一个区域基因拷贝数增加,表明所述的尿路上皮癌患者为低风险组,具有更好的肿瘤预后;反之,基因集A1中没有基因出现基因突变,同时基因片段集B1中没有任何片段出现基因拷贝数减少,同时基因片段集C1中没有任何片段出现基因拷贝数增加,则该类尿路上皮癌患者为高风险组,具有较差的肿瘤预后。需要特殊说明的是,在本实验例中,基因组合1(panel 1)是在基因组合(panel)的基础上优选而来,由于检测的基因更少,故具有更低的成本。This application allows to select any gene fragments from the gene panel and combine them to form a new gene combination, and use the same criteria to grade the malignancy of urothelial carcinoma and predict the prognosis of the tumor. Here, the gene set A1 (Table 4) is selected from the gene set A of the gene combination (panel), the gene fragment set B1 (Table 5) is selected from the gene fragment set B, and the gene fragment is selected from the gene fragment set C Set C1 (Table 5) constitutes gene panel 1 (panel 1), which is used for grading the malignancy of urothelial carcinoma and predicting prognosis, and uses the MSK/TCGA database for feasibility analysis. Similarly, the judgment criteria are: if there is a gene mutation in at least one gene in gene set A1, or there is a gene copy number decrease in at least one region in gene fragment set B1, or there is an increase in gene copy number in at least one region in gene fragment set C1 , indicating that the urothelial carcinoma patients are in the low-risk group and have better tumor prognosis; on the contrary, no genes in the gene set A1 have gene mutations, and at the same time, there is no gene copy number reduction in any fragments in the gene fragment set B1, and at the same time If there is no gene copy number increase in any fragment in the gene fragment set C1, the patients with this type of urothelial carcinoma are in the high-risk group and have poor tumor prognosis. It should be noted that in this experimental example, the gene combination 1 (panel 1) is optimized on the basis of the gene combination (panel). Since there are fewer genes to be detected, it has a lower cost.
表4基因集A1Table 4 Gene set A1
Figure PCTCN2022116390-appb-000016
Figure PCTCN2022116390-appb-000016
表5基因片段集B1和C1Table 5 Gene fragment sets B1 and C1
Figure PCTCN2022116390-appb-000017
Figure PCTCN2022116390-appb-000017
Figure PCTCN2022116390-appb-000018
Figure PCTCN2022116390-appb-000018
按照实施例2所述的方法实施,基因组合1(panel 1)将上述926例患者进行恶性程度分级,顺利分为高风险组和低风险组,其中高风险组占比79.9%,低风险组占比20.1%,通过Kaplan-Meier生存分析,可见高风险组和低风险组的肿瘤特异性生存(图5)、肿瘤无病生存(图6)和总生存(图7)均具有统计学差异并符合本申请的分组预期:低风险组具有明显更好的肿瘤特异性生存(Log-rank p值=7.319e-3)、肿瘤无病生存(Log-rank p值=2.361e-3)和总生存(Log-rank p值=1.325e-3)。故使用本申请从基因组合(panel)中挑选出的其他基因组合仍然可以对尿路上皮癌患者进行恶性程度分级和预后预测。Carry out according to the method described in embodiment 2, gene group 1 (panel 1) carries out malignant degree classification to above-mentioned 926 patients, is successfully divided into high-risk group and low-risk group, wherein high-risk group accounts for 79.9%, low-risk group Accounting for 20.1%, through Kaplan-Meier survival analysis, it can be seen that there are statistical differences in tumor-specific survival (Figure 5), tumor-free survival (Figure 6) and overall survival (Figure 7) between the high-risk group and the low-risk group And meet the grouping expectations of this application: the low-risk group has significantly better tumor-specific survival (Log-rank p value = 7.319e-3), tumor disease-free survival (Log-rank p value = 2.361e-3) and Overall survival (Log-rank p-value=1.325e-3). Therefore, using other gene combinations selected from the gene combination (panel) in this application can still be used for grading the degree of malignancy and predicting the prognosis of patients with urothelial carcinoma.
实验例3优选的基因组合和检测方法(仅检测基因突变)在评估人尿路上皮癌恶性程度分级和预后预测的可行性验证Experimental Example 3 Feasibility verification of the optimal gene combination and detection method (only detection of gene mutations) in evaluating the grade of malignancy and prognosis of human urothelial carcinoma
本申请允许仅检测基因突变,无需检测拷贝数变异,使用相同的判断标准对尿路上皮癌恶性程度进行分级和肿瘤预后预测。此处使用基因集A中全部基因的基因突变情况,用于尿路上皮癌恶性程度分级和预后预测,并使用MSK/TCGA数据库进行可行性分析。同理的,判断标准为:如果存在基因集A中至少一个基因的基因突变,表明所述的尿路上皮癌患者为低风险组,具有更好的肿瘤预后;反之,基因集A中没有基因出现基因突变,则该类尿路上皮癌患者为高风险组,具有较差的肿瘤预后。需要特殊说明的是,在本实验例中,由于仅检测基因突变情况,不检测拷贝数变异情况,故具有更低的成本。This application allows the detection of gene mutations only, without the detection of copy number variations, and uses the same criteria to grade the malignancy of urothelial carcinoma and predict tumor prognosis. Here, the gene mutations of all genes in gene set A are used for grading the malignancy of urothelial carcinoma and predicting prognosis, and the MSK/TCGA database is used for feasibility analysis. Similarly, the judging criteria are: if there is a gene mutation in at least one gene in gene set A, it indicates that the urothelial carcinoma patients are in the low-risk group and have a better tumor prognosis; otherwise, there is no gene in gene set A If there is a gene mutation, the patients with this type of urothelial carcinoma belong to the high-risk group and have a poor tumor prognosis. It should be noted that in this experimental example, only gene mutations are detected, and copy number variations are not detected, so the cost is lower.
按照实施例2所述的方法实施,使用基因组合A将上述926例患者进行恶性程度分级,顺利分为高风险组和低风险组,其中高风险组占比33.6%,低风险组占比66.4%,通过Kaplan-Meier生存分析,可见高风险组和低风险组的肿瘤特异性生存(图8)、肿瘤无进展生存(图9)、肿瘤无病生存(图10)和总生存(图11)均具有统计学差异并符合本申请的分组预期:低风险组具有明显更好的肿瘤特异性生存(Log-rank p值=1.431e-4)、肿瘤无进展生存(Log-rank p值=1.297e-6)、肿瘤无病生存(Log-rank p值=3.228e-4)和总生存(Log-rank p值=5.79e-11)。故仅检测本申请中基因集A的基因突变状态,不检测基因拷贝数变异时,仍然可以对尿路上皮癌进行恶性程度分级和预后预测,可以预期的是,针对从基因集A中挑选出的其他基因组合进行基因突变检测仍然可以对尿路上皮癌患者进行恶性程度分级和预后预测。According to the method described in Example 2, the above-mentioned 926 patients were graded for malignancy using gene combination A, and were successfully divided into high-risk group and low-risk group, of which the high-risk group accounted for 33.6%, and the low-risk group accounted for 66.4%. %, through Kaplan-Meier survival analysis, we can see the tumor-specific survival (Figure 8), tumor progression-free survival (Figure 9), tumor disease-free survival (Figure 10) and overall survival (Figure 11) of high-risk group and low-risk group ) all have statistical differences and meet the grouping expectations of this application: the low-risk group has significantly better tumor-specific survival (Log-rank p value=1.431e-4), tumor progression-free survival (Log-rank p value=1.431e-4), tumor progression-free survival (Log-rank p value= 1.297e-6), tumor disease-free survival (Log-rank p value = 3.228e-4) and overall survival (Log-rank p value = 5.79e-11). Therefore, only the gene mutation status of gene set A in this application is detected, and the gene copy number variation is not detected, and the malignant degree and prognosis prediction of urothelial carcinoma can still be carried out. Gene mutation detection of other gene combinations can still grade the malignancy and predict the prognosis of patients with urothelial carcinoma.
实验例4用于人肿瘤分级的基因组合和检测方法在评估人泛癌(Pancancer)恶性程度分级和预后预测的可行性验证Experimental Example 4 Feasibility verification of the gene combination and detection method used for human tumor grading in evaluating the malignant degree grading and prognosis prediction of human pancancer (Pancancer)
TCGA(PanCancer Atlas)泛癌数据库是全球公认的泛癌数据库,可用其检验本申请用于评估泛癌恶性程度分级和预后预测的可行性和可信度。The TCGA (PanCancer Atlas) pan-cancer database is a globally recognized pan-cancer database, which can be used to test the feasibility and credibility of this application for assessing pan-cancer malignancy grading and prognosis prediction.
TCGA(PanCancer Atlas)泛癌数据共有10953名患者(10967例)泛癌资料,可用其检验本申请用于评估泛癌恶性程度分级和预后预测的可行性和可信度。TCGA (PanCancer Atlas) pan-cancer data has a total of 10,953 patients (10,967 cases) pan-cancer data, which can be used to test the feasibility and reliability of this application for assessing pan-cancer malignancy grading and prognosis prediction.
按照实施例4所述的方法实施,本实验例中选取实施例1中基因集A的全部基因、基因片段集B和基因片段集C中的所有片段进行实施,基因片段集B和基因片段集C中实际用于检测的基因同实验例1中表3。将上述10967例患者进行恶性程度分级,顺利分为高风险组和低风险组,其中高风险组占比59.5%,低风险组占比40.5%,通过Kaplan-Meier生存分析,可见高风险组和低风险组的肿瘤无病生存(图12)、肿瘤无进展生存(图13)和总生存(图14)均具有统计学差异并符合本申请的分组预期:低风险组具有明显更好的肿瘤无病生存(Log-rank p值=5.993e-6)、肿瘤无进展生存(Log-rank p值=8.726e-3)和总生存(Log-rank p值=0.0428)。故使用本申请的基因组合对泛癌患者进行恶性程度分级和预后预测准确可靠。According to the method described in Example 4, in this experimental example, all the genes in the gene set A, the gene fragment set B and all the fragments in the gene fragment set C are selected for implementation in this experimental example, the gene fragment set B and the gene fragment set The genes actually used for detection in C are the same as Table 3 in Experimental Example 1. The above-mentioned 10967 patients were graded for malignancy and successfully divided into high-risk group and low-risk group, of which the high-risk group accounted for 59.5%, and the low-risk group accounted for 40.5%. Through the Kaplan-Meier survival analysis, it can be seen that the high-risk group and the low-risk group accounted for 40.5%. The tumor disease-free survival (Figure 12), tumor progression-free survival (Figure 13) and overall survival (Figure 14) of the low-risk group were all statistically different and met the grouping expectations of this application: the low-risk group had significantly better tumor Disease-free survival (Log-rank p value=5.993e-6), tumor progression-free survival (Log-rank p value=8.726e-3) and overall survival (Log-rank p value=0.0428). Therefore, it is accurate and reliable to use the gene combination of the application to grade the malignant degree and predict the prognosis of pan-cancer patients.
实验例5优选的基因组合和检测方法在评估人泛癌恶性程度分级和预后预测的可行性验证Experimental Example 5: Feasibility verification of the optimal gene combination and detection method in assessing human pan-cancer malignancy grading and prognosis prediction
本申请允许从基因组合(panel)中挑选任意基因片段进行组合,形成新的基因组合,使用相同的判断标准对泛癌恶性程度进行分级和肿瘤预后预测。此处按实施例5所述方法,挑选并使用实验例2中的基因组合1(panel 1),用于泛癌恶性程度分级和预后预测,并使用TCGA(PanCancer Atlas)泛癌数据库进行可行性分析。同理的,判断标准为:如果存在基因集A1中至少一个基因的基因突变,或存在基因片段集B1中至少一个区域基因拷贝数减少,或存在基因片段集C1中至少一个区域基因拷贝数增加,表明所述的泛癌患者为低风险组,具有更好的肿瘤预后;反之,基因集A1中没有基因出现基因突变,同时基因片段集B1中没有任何片段出现基因拷贝数减少,同时基因片段集C1中没有任何片段出现基因拷贝数增加,则该类泛癌患者为高风险组,具有较差的肿瘤预后。需要特殊说明的是,在本实验例中,基 因组合1(panel 1)是在基因组合(panel)的基础上优选而来,由于检测的基因更少,故具有更低的成本。This application allows to select any gene fragments from the gene panel and combine them to form a new gene combination, and use the same judgment standard to grade the degree of malignancy of pan-cancer and predict the prognosis of tumors. Here, according to the method described in Example 5, select and use the gene combination 1 (panel 1) in Experimental Example 2 for pan-cancer malignancy grading and prognosis prediction, and use the TCGA (PanCancer Atlas) pan-cancer database for feasibility analyze. Similarly, the judgment criteria are: if there is a gene mutation in at least one gene in gene set A1, or there is a gene copy number decrease in at least one region in gene fragment set B1, or there is an increase in gene copy number in at least one region in gene fragment set C1 , indicating that the pan-cancer patients are a low-risk group with better tumor prognosis; on the contrary, no genes in the gene set A1 have gene mutations, and at the same time, no gene fragments in the gene fragment set B1 have gene copy number reduction, and at the same time, the gene fragments If there is no gene copy number increase in any fragment in set C1, then this type of pan-cancer patients is a high-risk group with poor tumor prognosis. It should be noted that in this experimental example, the gene combination 1 (panel 1) is optimized on the basis of the gene combination (panel). Since there are fewer genes to be detected, it has a lower cost.
按照实施例4和实施例5所述的方法实施,基因组合1(panel 1)将上述泛癌数据库中10967例患者进行恶性程度分级,顺利分为高风险组和低风险组,其中高风险组占比81.5%,低风险组占比18.5%,通过Kaplan-Meier生存分析,可见高风险组和低风险组的肿瘤特异性生存(图15)、肿瘤无进展生存(图16)、肿瘤无病生存(图17)和总生存(图18)均具有统计学差异并符合本申请的分组预期:低风险组具有明显更好的肿瘤特异性生存(Log-rank p值=7.74e-8)、肿瘤无进展生存(Log-rank p值=2.368e-3)、肿瘤无病生存(Log-rank p值=5.455e-3)和总生存(Log-rank p值=8.92e-9)。故使用本申请从基因组合(panel)中挑选出的其他基因组合仍然可以对泛癌患者进行恶性程度分级和预后预测。Carry out according to the method described in embodiment 4 and embodiment 5, gene combination 1 (panel 1) carry out malignancy classification to 10967 routine patients in the above-mentioned pan-cancer database, successfully divide into high-risk group and low-risk group, wherein high-risk group It accounted for 81.5%, and the low-risk group accounted for 18.5%. Through Kaplan-Meier survival analysis, it can be seen that the high-risk group and low-risk group have tumor-specific survival (Figure 15), tumor progression-free survival (Figure 16), and tumor-free survival. Survival (Figure 17) and overall survival (Figure 18) were statistically different and in line with the grouping expectations of this application: the low-risk group had significantly better tumor-specific survival (Log-rank p value = 7.74e-8), Tumor progression-free survival (Log-rank p value=2.368e-3), tumor disease-free survival (Log-rank p value=5.455e-3) and overall survival (Log-rank p value=8.92e-9). Therefore, using other gene combinations selected from the gene panels (panels) in this application can still be used to grade the degree of malignancy and predict the prognosis of pan-cancer patients.
实验例6优选的基因组合和检测方法(仅检测基因突变)在评估人泛癌恶性程度分级和预后预测的可行性验证Experimental Example 6: Feasibility verification of the optimal gene combination and detection method (only detection of gene mutation) in assessing human pan-cancer malignancy grading and prognosis prediction
本申请允许仅检测基因突变,无需检测拷贝数变异,使用相同的判断标准对尿路上皮癌恶性程度进行分级和肿瘤预后预测。此处使用实施例1中基因集A中全部基因的基因突变情况,用于泛癌恶性程度分级和预后预测,并使用TCGA泛癌数据库进行可行性分析。同理的,判断标准为:如果存在基因集A中至少一个基因的基因突变,表明所述的泛癌患者为低风险组,具有更好的肿瘤预后;反之,基因集A中没有基因出现基因突变,则该类泛癌患者为高风险组,具有较差的肿瘤预后。需要特殊说明的是,在本实验例中,由于仅检测基因突变情况,不检测拷贝数变异情况,故具有更低的成本。This application allows the detection of gene mutations only, without the detection of copy number variations, and uses the same criteria to grade the malignancy of urothelial carcinoma and predict tumor prognosis. Here, the gene mutations of all genes in gene set A in Example 1 are used for pan-cancer malignancy grading and prognosis prediction, and the TCGA pan-cancer database is used for feasibility analysis. Similarly, the criterion for judging is: if there is a gene mutation in at least one gene in gene set A, it indicates that the pan-cancer patients are in the low-risk group and have a better tumor prognosis; otherwise, no gene in gene set A has a gene mutation mutation, the pan-cancer patients belong to the high-risk group with poor tumor prognosis. It should be noted that in this experimental example, only gene mutations are detected, and copy number variations are not detected, so the cost is lower.
按照实施例2、实施例4和实施例5所述的方法实施,使用基因组合A将上述10967例患者进行恶性程度分级,顺利分为高风险组和低风险组,其中高风险组占比64.1%,低风险组占比35.9%,通过Kaplan-Meier生存分析,可见高风险组和低风险组的肿瘤特异性生存(图19)、肿瘤无进展生存(图20)、肿瘤无病生存(图21)和总生存(图22)均具有统计学差异并符合本申请的分组预期:低风险组具有明显更好的肿瘤特异性生存(Log-rank p值=0.0320)、肿瘤无进展生存(Log-rank p值=7.190e-3)、肿瘤无病生存(Log-rank p值=8.83e-8)和总生存(Log-rank p值=6.108e-3)。故仅检测本申请中基因集A的基因突变状态,不检测基因拷贝数变异时,仍然可以对泛癌进行恶性程度分级和预后预测,可以预期的是,针对从基因集A中挑选出的其他基因组合进行基因突变检测仍然可以对泛癌患者进行恶性程度分级和预后预测。According to the methods described in Example 2, Example 4, and Example 5, the above-mentioned 10,967 patients were graded for malignancy using gene combination A, and were successfully divided into high-risk group and low-risk group, of which the high-risk group accounted for 64.1 %, the low-risk group accounted for 35.9%. Through the Kaplan-Meier survival analysis, it can be seen that the tumor-specific survival (Fig. 19), tumor progression-free survival (Fig. 20), tumor disease-free survival (Fig. 21) and overall survival (Fig. 22) were statistically different and met the grouping expectations of this application: the low-risk group had significantly better tumor-specific survival (Log-rank p value = 0.0320), tumor progression-free survival (Log -rank p-value=7.190e-3), tumor disease-free survival (Log-rank p-value=8.83e-8) and overall survival (Log-rank p-value=6.108e-3). Therefore, only the gene mutation status of gene set A in this application is detected, and when gene copy number variation is not detected, the malignancy degree and prognosis prediction of pan-cancer can still be performed. It can be expected that for other genes selected from gene set A Gene mutation detection by gene combination can still grade the malignancy and predict the prognosis of pan-cancer patients.
显然,上述实施例仅仅是为清楚地说明所作的举例,而并非对实施方式的限定。对于所属领域的普通技术人员来说,在上述说明的基础上还可以做出其它不同形式的变化或变动。这里无需也无法对所有的实施方式予以穷举。而由此所引伸出的显而易见的变化或变动仍处于本发明创造的保护范围之中。Apparently, the above-mentioned embodiments are only examples for clear description, rather than limiting the implementation. For those of ordinary skill in the art, other changes or changes in different forms can be made on the basis of the above description. It is not necessary and impossible to exhaustively list all the implementation manners here. And the obvious changes or changes derived therefrom are still within the scope of protection of the present invention.

Claims (25)

  1. 一种用于人肿瘤分级的基因组合,其特征在于,所述的基因组合由基因集A、基因片段集B和/或基因片段集C组成;A gene combination for grading human tumors, characterized in that the gene combination consists of gene set A, gene fragment set B and/or gene fragment set C;
    所述的基因集A包括:ACVR1B、ATP4B、AZGP1、BRAF、BRCA1、BRCA2、CRYZL1、DCTN1、E2F3、EGFR、ERBB2、ERCC2、ESPL1、FGFR3、FKBP6、GZMM、H3-5、IGLL5、IRF7、METTL24、MORN5、MTOR、NPY1R、PET100、PIK3CA、PPARG、PTPN11、PTX4、RGPD8、SERPINA12、STAG2和ZNF141中至少一个;The gene set A includes: ACVR1B, ATP4B, AZGP1, BRAF, BRCA1, BRCA2, CRYZL1, DCTN1, E2F3, EGFR, ERBB2, ERCC2, ESPL1, FGFR3, FKBP6, GZMM, H3-5, IGLL5, IRF7, METTL24, At least one of MORN5, MTOR, NPY1R, PET100, PIK3CA, PPARG, PTPN11, PTX4, RGPD8, SERPINA12, STAG2 and ZNF141;
    所述的基因片段集B包括:chr1:155289001-155911999、chr1:226259001-226736999、chr8:30515001-30892999、chr8:42065001-43147999、chr9:134612001-135906999、chr11:48509001-55112999、chr14:24768001-105145999、chr16:1-73083999、chr18:1-319999和chr18:15325001-19749999中至少一个;所述的基因片段集B包括:chr1:155289001-155911999、chr1:226259001-226736999、chr8:30515001-30892999、chr8:42065001-43147999、chr9:134612001-135906999、chr11:48509001-55112999、chr14:24768001-105145999 , at least one of chr16:1-73083999, chr18:1-319999 and chr18:15325001-19749999;
    所述的基因片段集C包括:chr3:12030001-12639999、chr17:2293001-2306999和chr17:37830001-37979999中至少一个。The gene fragment set C includes: at least one of chr3:12030001-12639999, chr17:2293001-2306999 and chr17:37830001-37979999.
  2. 根据权利要求1所述的一种用于人肿瘤分级的基因组合,其特征在于,所述的基因片段集B中包括的详细基因如下表:A gene combination for human tumor grading according to claim 1, wherein the detailed genes included in the gene fragment set B are as follows:
    表1基因片段集BTable 1 Gene Fragment Set B
    Figure PCTCN2022116390-appb-100001
    Figure PCTCN2022116390-appb-100001
    Figure PCTCN2022116390-appb-100002
    Figure PCTCN2022116390-appb-100002
    Figure PCTCN2022116390-appb-100003
    Figure PCTCN2022116390-appb-100003
    Figure PCTCN2022116390-appb-100004
    Figure PCTCN2022116390-appb-100004
    Figure PCTCN2022116390-appb-100005
    Figure PCTCN2022116390-appb-100005
    Figure PCTCN2022116390-appb-100006
    Figure PCTCN2022116390-appb-100006
  3. 根据权利要求1所述的一种用于人肿瘤分级的基因组合,其特征在于,所述的基因片段集C中包括的详细基因如下表:A gene combination for human tumor grading according to claim 1, wherein the detailed genes included in the gene fragment set C are as follows:
    表2基因片段集CTable 2 Gene fragment set C
    基因片段位置gene fragment location 可用于检测的基因Genes Available for Testing chr3:12030001-12639999Chr3: 12030001-12639999 TIMP4,SYN2,PPARG,TSEN2,MKRN2中至少一个At least one of TIMP4, SYN2, PPARG, TSEN2, MKRN2 chr17:2293001-2306999chr17:2293001-2306999 MNTMNT chr17:37830001-37979999chr17:37830001-37979999 GRB7,ERBB2,IKZF3中至少一个At least one of GRB7, ERBB2, IKZF3
    可选的,所述基因集A包括:ACVR1B、ATP4B、AZGP1、BRAF、CRYZL1、DCTN1、FKBP6、GZMM、H3-5、IGLL5、IRF7、METTL24、MORN5、NPY1R、PET100、PTPN11、PTX4、RGPD8、SERPINA12和ZNF141中至少一个;Optionally, the gene set A includes: ACVR1B, ATP4B, AZGP1, BRAF, CRYZL1, DCTN1, FKBP6, GZMM, H3-5, IGLL5, IRF7, METTL24, MORN5, NPY1R, PET100, PTPN11, PTX4, RGPD8, SERPINA12 and at least one of ZNF141;
    所述基因片段集B包括:chr1:155289001-155911999、chr1:226259001-226736999、chr8:30515001-30892999、chr8:42065001-43147999、chr9:134612001-135906999、chr14:24768001-105145999、chr16:1-73083999、chr18:1-319999和chr18:15325001-19749999中至少一个;所述基因片段集B包括:chr1:155289001-155911999、chr1:226259001-226736999、chr8:30515001-30892999、chr8:42065001-43147999、chr9:134612001-135906999、chr14:24768001-105145999、chr16:1-73083999、 At least one of chr18:1-319999 and chr18:15325001-19749999;
    所述基因片段集C包括:chr17:2293001-2306999。The gene fragment set C includes: chr17:2293001-2306999.
  4. 权利要求1-3任一项所述的基因组合在制备用于人肿瘤分级检测的产品中的用途。Use of the gene combination described in any one of claims 1-3 in the preparation of products for human tumor grading detection.
  5. 根据权利要求4所述的用途,其特征在于,所述的肿瘤为泌尿系统肿瘤或泛癌;The use according to claim 4, wherein the tumor is a tumor of the urinary system or pan-cancer;
    可选的,所述泌尿系统肿瘤为泌尿系统恶性肿瘤;Optionally, the tumor of the urinary system is a malignant tumor of the urinary system;
    可选的,所述泌尿系统肿瘤为尿路上皮癌;Optionally, the tumor of the urinary system is urothelial carcinoma;
    可选的,所述泛癌为TCGA泛癌数据中的癌种。Optionally, the pan-cancer is a cancer type in TCGA pan-cancer data.
  6. 根据权利要求4或5所述的用途,其特征在于,所述的肿瘤分级是指肿瘤恶性程度判断和肿瘤预后的预测,用于指导临床诊疗;The use according to claim 4 or 5, characterized in that the tumor grade refers to the judgment of tumor malignancy and the prediction of tumor prognosis, which is used to guide clinical diagnosis and treatment;
    可选的,所述肿瘤分级分为高风险组和低风险组。Optionally, the tumor grades are divided into high-risk group and low-risk group.
  7. 根据权利要求4-6任一项所述的用途,其特征在于,所述产品包括用于检测所述基因组合中基因的基因类型的引物、探针、试剂、试剂盒、基因芯片或检测系统。The use according to any one of claims 4-6, wherein the product includes primers, probes, reagents, test kits, gene chips or detection systems for detecting the genotype of genes in the gene combination .
  8. 根据权利要求7所述的用途,其特征在于,所述产品为针对基因集A、基因片段集B和基因片段集C中基因的外显子和相关内含子区域进行检测。The use according to claim 7, characterized in that the product is for detection of exons and related intron regions of genes in gene set A, gene fragment set B and gene fragment set C.
  9. 根据权利要求4-8任一项所述的用途,其特征在于,所述肿瘤分级的方法包括如下步骤:The use according to any one of claims 4-8, wherein the method for grading tumors comprises the following steps:
    步骤S1:评估所述癌细胞组织中的基因集A中所包含基因的基因突变,评估癌细胞组织中的基因片段集B和基因片段集C的基因拷贝数变异;Step S1: Evaluate the gene mutation of the genes contained in the gene set A in the cancer cell tissue, and evaluate the gene copy number variation of the gene fragment set B and the gene fragment set C in the cancer cell tissue;
    步骤S2:基于步骤S1的评估结果,判断癌症恶性程度并进行肿瘤预后预测。Step S2: Based on the evaluation results of step S1, the degree of malignancy of the cancer is judged and the prognosis of the tumor is predicted.
  10. 根据权利要求9所述的用途,其特征在于,所述的基因突变包括碱基置换突变、缺失突变、插入突变和/或融合突变,所述基因拷贝数变异包括基因拷贝数增加和/或基因拷贝数减少。The use according to claim 9, wherein the gene mutation includes base substitution mutation, deletion mutation, insertion mutation and/or fusion mutation, and the gene copy number variation includes gene copy number increase and/or gene mutation Copy number reduction.
  11. 根据权利要求9或10所述的用途,其特征在于,在步骤S1中,通过比较所述肿瘤组织与正常组织的测序数据,用于评估所述基因集A中包含基因的基因突变,同时评估所述基因片段集B和基因片段集C的基因拷贝数变异。The use according to claim 9 or 10, characterized in that in step S1, by comparing the sequencing data of the tumor tissue and normal tissue, it is used to evaluate the gene mutation of the genes contained in the gene set A, and at the same time evaluate The gene copy number variation of the gene fragment set B and the gene fragment set C.
  12. 根据权利要求8或9或10或11所述的用途,其特征在于,所述步骤S2中,如果基因集A中至少一个基因出现基因突变,或基因片段集B中至少一个片段出现基因拷贝数减少,或基因片段集C中至少一个片段出现基因拷贝数增加,所述肿瘤分级为低风险组;反之,即基因集A中没有基因出现基因突变,同时基因片段集B中没有任何片段出现基因拷贝数减少,同时基因片段集C中没有任何片段出现基因拷贝数增加,所述肿瘤分级为高风险组。The use according to claim 8 or 9 or 10 or 11, characterized in that in step S2, if at least one gene in the gene set A has a gene mutation, or at least one gene fragment in the gene set B has a gene copy number Decrease, or at least one fragment in gene fragment set C has gene copy number increase, and the tumor is graded as a low-risk group; on the contrary, no gene in gene set A has gene mutation, and at the same time, there is no gene in any fragment in gene fragment set B If the copy number is reduced, and there is no gene copy number increase in any fragment in the gene fragment set C, the tumor is graded as a high-risk group.
  13. 根据权利要求8或9或10或11所述的用途,其特征在于,所述步骤S2中,如果基因集A中至少一个基因出现基因突变,所述肿瘤分级为低风险组;反之,即基因集A中没有基因出现基因突变,所述肿瘤分级为高风险组。The use according to claim 8 or 9 or 10 or 11, characterized in that in step S2, if at least one gene in the gene set A has a gene mutation, the tumor is graded as a low-risk group; In set A, no genes are mutated, and the tumors are graded as high-risk group.
  14. 根据权利要求4-13任一项所述的用途,其特征在于,从所述基因组合中选择任意基因片段进行组合,形成新的基因组合,使用相同的肿瘤分级的方法对肿瘤恶性程度进行分级和肿瘤预后预测,从而指导临床诊疗。The use according to any one of claims 4-13, characterized in that any gene fragments are selected from the gene combination for combination to form a new gene combination, and the tumor malignancy is graded using the same tumor grading method And tumor prognosis prediction, so as to guide clinical diagnosis and treatment.
  15. 使用权利要求1-3任一项所述的基因组合检测人肿瘤分级的方法。A method for detecting human tumor grades using the gene combination described in any one of claims 1-3.
  16. 根据权利要求15所述的方法,其特征在于,所述的肿瘤为泌尿系统肿瘤或泛癌;The method according to claim 15, wherein the tumor is a tumor of the urinary system or pan-cancer;
    可选的,所述泌尿系统肿瘤为泌尿系统恶性肿瘤;Optionally, the tumor of the urinary system is a malignant tumor of the urinary system;
    可选的,所述泌尿系统肿瘤为尿路上皮癌;Optionally, the tumor of the urinary system is urothelial carcinoma;
    可选的,所述泛癌为TCGA泛癌数据中的癌种。Optionally, the pan-cancer is a cancer type in TCGA pan-cancer data.
  17. 根据权利要求15或16所述的方法,其特征在于,所述的肿瘤分级是指肿瘤恶性程度判断和肿瘤预后的预测,用于指导临床诊疗;The method according to claim 15 or 16, characterized in that the tumor grade refers to the judgment of tumor malignancy and the prediction of tumor prognosis, which is used to guide clinical diagnosis and treatment;
    可选的,所述肿瘤分级分为高风险组和低风险组。Optionally, the tumor grades are divided into high-risk group and low-risk group.
  18. 根据权利要求15-17任一项所述的方法,其特征在于,所述方法包括使用检测所述基因组合中基因的基因类型的引物、探针、试剂、试剂盒、基因芯片或检测系统。The method according to any one of claims 15-17, characterized in that the method comprises using primers, probes, reagents, kits, gene chips or detection systems for detecting the genotypes of genes in the gene combination.
  19. 根据权利要求18所述的方法,其特征在于,所述方法为针对基因集A、基因片段集B和基因片段集C中基因的外显子和相关内含子区域进行检测。The method according to claim 18, characterized in that, the method is to detect exons and related intron regions of genes in gene set A, gene fragment set B and gene fragment set C.
  20. 根据权利要求15-19任一项所述的方法,其特征在于,所述肿瘤分级的方法包括如下步骤:The method according to any one of claims 15-19, wherein the method for grading the tumor comprises the following steps:
    步骤S1:评估所述癌细胞组织中的基因集A中所包含基因的基因突变,评估癌细胞组织中的基因片段集B和基因片段集C的基因拷贝数变异;Step S1: Evaluate the gene mutation of the genes contained in the gene set A in the cancer cell tissue, and evaluate the gene copy number variation of the gene fragment set B and the gene fragment set C in the cancer cell tissue;
    步骤S2:基于步骤S1的评估结果,判断癌症恶性程度并进行肿瘤预后预测。Step S2: Based on the evaluation results of step S1, the degree of malignancy of the cancer is judged and the prognosis of the tumor is predicted.
  21. 根据权利要求20所述的方法,其特征在于,所述的基因突变包括碱基置换突变、缺失突变、插入突变和/或融合突变,所述基因拷贝数变异包括基因拷贝数增加和/或基因拷贝数减少。The method according to claim 20, wherein the gene mutation includes base substitution mutation, deletion mutation, insertion mutation and/or fusion mutation, and the gene copy number variation includes gene copy number increase and/or gene mutation Copy number reduction.
  22. 根据权利要求20或21所述的方法,其特征在于,在步骤S1中,通过比较所述肿瘤组织与正常组织的测序数据,用于评估所述基因集A中包含基因的基因突变,同时评估所述基因片段集B和基因片段集C的基因拷贝数变异。The method according to claim 20 or 21, characterized in that, in step S1, by comparing the sequencing data of the tumor tissue and normal tissue, it is used to evaluate the gene mutation of the genes contained in the gene set A, and at the same time evaluate The gene copy number variation of the gene fragment set B and the gene fragment set C.
  23. 根据权利要求19或20或21或22所述的方法,其特征在于,所述步骤S2中,如果基因集A中至少一个基因出现基因突变,或基因片段集B中至少一个片段出现基因拷贝数减少,或基因片段集C中至少一个片段出现基因拷贝数增加,所述肿瘤分级为低风险组;反之,即基因集A中没有基因出现基因突变,同时基因片段集B中没有任何片段出现基因拷贝数减少,同时基因片段集C中没有任何片段出现基因拷贝数增加,所述肿瘤分级为高风险组。The method according to claim 19 or 20 or 21 or 22, characterized in that in step S2, if at least one gene in the gene set A has a gene mutation, or at least one gene fragment in the gene set B has a gene copy number Decrease, or at least one fragment in gene fragment set C has gene copy number increase, and the tumor is graded as a low-risk group; on the contrary, no gene in gene set A has gene mutation, and at the same time, there is no gene in any fragment in gene fragment set B If the copy number is reduced, and there is no gene copy number increase in any fragment in the gene fragment set C, the tumor is graded as a high-risk group.
  24. 根据权利要求19或20或21或22所述的方法,其特征在于,所述步骤S2中,如果基因集A中至少一个基因出现基因突变,所述肿瘤分级为低风险组;反之,即基因集A中没有基因出现基因突变,所述肿瘤分级为高风险组。The method according to claim 19 or 20 or 21 or 22, characterized in that in step S2, if at least one gene in the gene set A has a gene mutation, the tumor is graded as a low-risk group; In set A, no genes are mutated, and the tumors are graded as high-risk group.
  25. 根据权利要求15-24任一项所述的方法,其特征在于,从所述基因组合中选择任意基因片段进行组合,形成新的基因组合,使用相同的肿瘤分级的方法对肿瘤恶性程度进行分级和肿瘤预后预测,从而指导临床诊疗。The method according to any one of claims 15-24, characterized in that any gene fragments are selected from the gene combination for combination to form a new gene combination, and the tumor malignancy is graded using the same tumor grading method And tumor prognosis prediction, so as to guide clinical diagnosis and treatment.
PCT/CN2022/116390 2021-09-02 2022-09-01 Gene combination for human tumor grading and use thereof WO2023030422A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111028348.8A CN113604571A (en) 2021-09-02 2021-09-02 Gene combination for human tumor classification and application thereof
CN202111028348.8 2021-09-02

Publications (1)

Publication Number Publication Date
WO2023030422A1 true WO2023030422A1 (en) 2023-03-09

Family

ID=78310000

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/116390 WO2023030422A1 (en) 2021-09-02 2022-09-01 Gene combination for human tumor grading and use thereof

Country Status (2)

Country Link
CN (1) CN113604571A (en)
WO (1) WO2023030422A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113604571A (en) * 2021-09-02 2021-11-05 北京大学第一医院 Gene combination for human tumor classification and application thereof
CN114854861A (en) * 2022-05-20 2022-08-05 北京大学第一医院 Application of gene combination in preparation of human tumor homologous recombination defect, tumor mutation load and microsatellite instability grading detection product

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014071218A2 (en) * 2012-11-02 2014-05-08 University Of Utah Research Foundation Biomarkers for breast cancer and methods of using same
US20160024588A1 (en) * 2013-03-14 2016-01-28 The Broad Institute, Inc. Osteosarcoma-associated risk markers and uses thereof
EP3118324A1 (en) * 2015-07-13 2017-01-18 Cartagenia N.V. Method for analyzing copy number variation in the detection of cancer
US20170037480A1 (en) * 2014-04-11 2017-02-09 Whitehead Institute For Biomedical Research Hsf1 in tumor stroma
US20170322217A1 (en) * 2014-08-11 2017-11-09 Agency For Science, Technology And Research A method for prognosis of ovarian cancer, patient's stratification
US20180298448A1 (en) * 2015-01-21 2018-10-18 Bin Tean Teh Method and kit for pathologic grading of breast neoplasm
CN113025716A (en) * 2021-03-02 2021-06-25 北京大学第一医院 Gene combination for human tumor classification and application thereof
CN113604571A (en) * 2021-09-02 2021-11-05 北京大学第一医院 Gene combination for human tumor classification and application thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7955800B2 (en) * 2002-06-25 2011-06-07 Advpharma Inc. Metastasis-associated gene profiling for identification of tumor tissue, subtyping, and prediction of prognosis of patients
WO2017008165A1 (en) * 2015-07-14 2017-01-19 British Columbia Cancer Agency Branch Classification method and treatment for endometrial cancers
CN111833965A (en) * 2019-11-08 2020-10-27 中国科学院北京基因组研究所 Urinary sediment genomic DNA classification method, device and application
CN111951893B (en) * 2020-08-24 2022-11-15 中山大学附属第三医院 Method for constructing tumor mutation load TMB panel

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014071218A2 (en) * 2012-11-02 2014-05-08 University Of Utah Research Foundation Biomarkers for breast cancer and methods of using same
US20160024588A1 (en) * 2013-03-14 2016-01-28 The Broad Institute, Inc. Osteosarcoma-associated risk markers and uses thereof
US20170037480A1 (en) * 2014-04-11 2017-02-09 Whitehead Institute For Biomedical Research Hsf1 in tumor stroma
US20170322217A1 (en) * 2014-08-11 2017-11-09 Agency For Science, Technology And Research A method for prognosis of ovarian cancer, patient's stratification
US20180298448A1 (en) * 2015-01-21 2018-10-18 Bin Tean Teh Method and kit for pathologic grading of breast neoplasm
EP3118324A1 (en) * 2015-07-13 2017-01-18 Cartagenia N.V. Method for analyzing copy number variation in the detection of cancer
CN113025716A (en) * 2021-03-02 2021-06-25 北京大学第一医院 Gene combination for human tumor classification and application thereof
CN113604571A (en) * 2021-09-02 2021-11-05 北京大学第一医院 Gene combination for human tumor classification and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DEAN MATTHEW; DAVIS DAVID A.; BURDETTE JOANNA E.: "Activin A stimulates migration of the fallopian tube epithelium, an origin of high-grade serous ovarian cancer, through non-canonical signaling", CANCER LETTERS, NEW YORK, NY, US, vol. 391, 20 January 2017 (2017-01-20), US , pages 114 - 124, XP029926115, ISSN: 0304-3835, DOI: 10.1016/j.canlet.2017.01.011 *

Also Published As

Publication number Publication date
CN113604571A (en) 2021-11-05

Similar Documents

Publication Publication Date Title
WO2023030422A1 (en) Gene combination for human tumor grading and use thereof
WO2022184073A1 (en) Gene combination for human tumor grading, and use thereof
Campan et al. Genome-scale screen for DNA methylation-based detection markers for ovarian cancer
KR102170726B1 (en) Method for selecting biomarker and method for providing information for diagnosis of cancer using thereof
US20160326594A1 (en) Prostate cancer gene profiles and methods of using the same
Huang et al. Construction of a genome instability-derived lncRNA-based risk scoring system for the prognosis of hepatocellular carcinoma
Wang et al. Development of an autophagy-related gene prognostic model and nomogram for estimating renal clear cell carcinoma survival
Zhan et al. Development and validation of a prognostic gene signature in clear cell renal cell carcinoma
EP3950960A1 (en) Dna methylation marker for predicting recurrence of liver cancer, and use thereof
WO2023221865A1 (en) Use of gene combination in preparation of human tumor homologous recombination deficiency, tumor mutation burden and microsatellite instability grading detection products
Chen et al. Identification and validation of a novel genomic instability-associated long non-coding RNA prognostic signature in head and neck squamous cell carcinoma
CN110408706A (en) It is a kind of assess recurrent nasopharyngeal carcinoma biomarker and its application
Zheng et al. A prognostic predictive model constituted with gene mutations of APC, BRCA2, CDH1, SMO, and TSC2 in colorectal cancer
Cao et al. An epithelial-mesenchymal transition (EMT) preoperative nomogram for prediction of lymph node metastasis in bladder cancer (BLCA)
Ke et al. DPP6 and MFAP5 are associated with immune infiltration as diagnostic biomarkers in distinguishing uterine leiomyosarcoma from leiomyoma
CN113774135B (en) Group of markers for predicting prognosis of high-grade serous ovarian cancer and application thereof
US20200370122A1 (en) Immune index methods for predicting breast cancer outcome
Yu et al. Identification of a prognostic biomarker predicting biochemical recurrence and construction of a novel nomogram for prostate cancer
WO2021211057A1 (en) Method of predicting the responsiveness to a cancer therapy
CN108342483B (en) Group of genes for molecular typing of non-hyper-mutant colorectal cancer and application thereof
Zhong et al. Identification of prognostic marker genes in head and neck squamous cell carcinoma: A study based on The Cancer Genome Atlas database and experimental validation
CN111471773A (en) Diagnostic biomarker for predicting prognosis of gastric adenocarcinoma patient and determination method and application thereof
CN110564851A (en) Group of genes for molecular typing of non-hyper-mutant rectal cancer and application thereof
Wei et al. Comprehensive analysis of aneuploidy status and its effect on the efficacy of EGFR-TKIs in lung cancer
Meng et al. Identification and validation of a novel prognostic gene model for colorectal cancer

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: 22863557

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE