CN114480660A - Gene Panel for detecting pan-cancer species, probe and application - Google Patents

Gene Panel for detecting pan-cancer species, probe and application Download PDF

Info

Publication number
CN114480660A
CN114480660A CN202210340759.9A CN202210340759A CN114480660A CN 114480660 A CN114480660 A CN 114480660A CN 202210340759 A CN202210340759 A CN 202210340759A CN 114480660 A CN114480660 A CN 114480660A
Authority
CN
China
Prior art keywords
tumor
genes
gene
forch
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202210340759.9A
Other languages
Chinese (zh)
Inventor
季序我
邓晨旭
董宇
韦宝耶
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Beijing Pukang Ruiren Medical Laboratory Co ltd
Predatum Biomedicine Suzhou Co ltd
Precision Scientific Technology Beijing Co ltd
Original Assignee
Beijing Pukang Ruiren Medical Laboratory Co ltd
Predatum Biomedicine Suzhou Co ltd
Precision Scientific Technology Beijing Co ltd
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 Beijing Pukang Ruiren Medical Laboratory Co ltd, Predatum Biomedicine Suzhou Co ltd, Precision Scientific Technology Beijing Co ltd filed Critical Beijing Pukang Ruiren Medical Laboratory Co ltd
Priority to CN202210340759.9A priority Critical patent/CN114480660A/en
Publication of CN114480660A publication Critical patent/CN114480660A/en
Pending legal-status Critical Current

Links

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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals

Abstract

The invention discloses a gene Panel for detecting pan-cancer species, a probe and application. The gene Panel comprises tumor pathway related genes, tumor genetic susceptibility genes, tumor high-frequency mutation genes, tumor targeting drug related genes, tumor driving genes, immunity curative effect related genes, key DDR pathway related genes and other genes playing important roles in the occurrence and development of cancers. The gene Panel can be used for detecting all biomarkers corresponding to solid tumor targeted drugs approved to be on the market by the national drug administration (NMPA)/American Food and Drug Administration (FDA) at one time, so that accurate treatment of tumors can be guided fully. The prognosis of the examined person can be evaluated, the relapse risk of the examined person is layered, and postoperative adjuvant therapy is guided; in addition, the method can also be used for evaluating the hereditary tumor of the examined person to prompt the family genetic risk, so as to realize early discovery, early diagnosis and early treatment.

Description

Gene Panel for detecting pan-cancer species, probe and application
Technical Field
The invention relates to the technical field of tumor polygene detection, in particular to a gene Panel, a probe and application for detecting pan-cancer species.
Background
According to statistics of national cancer centers, the number of new cancer cases in 2015 in China is 186.39/10 ten thousand, and the death rate is 105.84/10 ten thousand. Clinical studies have classified tumors as a gene disorder, mainly due to activation of proto-oncogenes or inactivation of suppressor genes. With the great progress of high-throughput sequencing (NGS) and biotechnology, a plurality of driver genes related to the occurrence and development of cancer and biomarkers for guiding tumor treatment are discovered, and a plurality of drugs are developed aiming at the driver genes and the biomarkers, so that precise treatment can be performed according to the detected driver gene mutation or biomarker in clinical treatment, and the treatment mode of tumors is changed from traditional treatment to precise treatment.
Currently, genetic testing techniques for precision therapy include Sanger sequencing (first-generation sequencing), RT-PCR (polymerase chain reaction), IHC (immunohistochemistry), FISH (fluorescence in situ hybridization), and high throughput sequencing (NGS) techniques, among others. Wherein, Sanger sequencing is a gold standard for sequencing, but one reaction of the sequencing technology can only obtain one sequence, so the sequencing flux is low; although individual reactions are inexpensive, the cost of obtaining large quantities of sequencing is high; the detection sensitivity is low, and generally, only mutations with mutation abundance of more than 20% can be detected, and the possibility of missing detection exists for some low-frequency mutations. RT-PCR can only detect known sites, and can not find unknown sites. IHC is mainly used for detecting protein expression, and cannot detect point mutation (SNV) of a gene, small fragment insertion deletion (Indel), and the like, and therefore, detection requiring medication guidance according to the gene SNV/Indel cannot be achieved by IHC. The FISH assay is a gold standard for identifying gene Fusion (Fusion) and amplification (CNV), but the SNV/Indel assay is not applicable. The high throughput sequencing (NGS) technique has the following advantages: high flux (several genes to hundreds of genes and even whole exome are detected at one time), high sensitivity, lower detection limit and stronger exploration capacity, can find unknown mutation, can detect multiple mutation types at one time (SNV/Indel/Fusion/CNV) and can also detect genome biomarkers (tumor mutation load/microsatellite instability). Comprehensive guidance on precise treatment can be achieved by only one test.
Disclosure of Invention
In order to solve the problems in Sanger sequencing (first generation sequencing), RT-PCR (polymerase chain reaction), IHC (immunohistochemistry) and FISH (fluorescence in situ hybridization) technologies, the invention selects a high throughput sequencing (NGS) technology. The invention provides the following technical scheme.
The invention provides a gene Panel for detecting pan-cancer species, which comprises tumor pathway related genes, tumor genetic susceptibility genes, tumor high-frequency mutation genes, tumor targeting drug related genes, tumor driving genes, immunity curative effect related genes, key DDR pathway related genes and other genes playing important roles in the occurrence and development of cancers; the key DDR pathway related genes comprise: ATM, ATR, BARD1, BLM, BRCA1, BRCA2, BRIP1, CHEK1, CHEK2, ERCC2, ERCC3, ERCC4, ERCC5, FAM175A, FANCA, FANCC, FANCD2, FANCL, MDC1, MLH1, MRE11A, MSH2, MSH3, MSH6, NBN, PALB2, PARP1, PMS1, PMS2, POLE, PRKDC, PTEN, RAD50, RAD51, RAD51B, RAD51C, RAD51D, RAD52, RAD54L, 53, TP53BP1, XRCC 2.
Preferably, the tumor pathway-associated genes include: AKT1, AKT2, AKT3, ALK, APC, AR, ARAF, ASXL1, ATM, ATR, ATRX, AURKA, AURKB, AXIN1, BCL2, BRAF, BRCA1, BRCA2, BRIP 2, BTK, CASP 2, CBL, CCND2, CCNE 2, CD79, CDK 2, KN 12, KN 22, MDM2, CDKN 22, CHEK2, CRKL 2, CRLF2, CSCSF 12, CTNNA 2, CTNNB 2, MDM2, 685MT 3, DOT 2, 685 2, 685 2, 685 2, 685, 2, 685 2, 685, 2, 685 2, 685 2, 685, 685 2, 685 2, 685 2, 685 2, 685 2, 685 2, 685, 2, 685 2, 685, 2, 685 2, 685 2, 685, 685, 685, 685 2, 685, 685, 685 2, 685, 685, 2, 685 2, 685, 685, 685, 685, 685, 685, 685, 685, 685, 685, 685, 685, PDGFRA, PDGFRB, PGR, PIK3CA, PIK3CG, PIK3R1, PIK3R2, PTCH1, PTEN, PTPN11, RAD50, RAD51, RB1, RET, RICTOR, RNF43, ROS1, RPTOR, SETD2, SMAD2, SMAD3, SMAD4, SMO, SOCS1, SRC, STAG2, STAT3, STAT4, STAT5A, STAT5B, SUFU, TET2, TGFBR1, TGFBR2, TNFAIP3, TP53, TSC1, TSC2, TSHR, WISP3, WT 1.
Preferably, the tumor genetic susceptibility gene comprises: ALK, APC, ATM, AXIN, BMPR1, BRCA, BRIP, CDH, CDKN2, CHEK, FH, FLCN, GREM, KIT, MEN, MET, MLH, MSH, MUTYH, NBN, NF, PALB, PMS, POLD, POLE, PTEN, RAD51, RB, SMAD, STK, TP, TSC, VHL, SDHD, TSC, PDGFRA, BARD, CDK, RET, SMARCA, SMARCB, SDHA, PRKAR1, HOXB, BRAF, PTCH, SDHAF.
Preferably, the tumor high-frequency mutant gene comprises: ACVR1B, AKT1, AMER1, APC, ARID 11, ARID1, ASXL1, ATM, ATRX, AXIN1, BAP1, BCL2L1, BCOR, BRA 1, BRCA1, BTG1, CASP 1, CBFB, CCND1, CCNE1, CD274, CDH1, CDK1, KN 11, MDM 11, CDKN K2 1, CHD 1, CREBB 1, CTNNA1, CTB 1, EGFR, ELF 1, EP300, ERBB4, ERBB 1, ERCC 1, ERBG 1, ERBBD 1, KM 1, 1, STK11, TBX3, TCF7L2, TERC, TERT, TGFBR2, TMPRSS2, TNFAIP3, TP53, TSC1, TSC2, U2AF1, VEGFA, VHL, WHSC1L 1.
Preferably, the tumor-targeted drug-associated gene comprises: ABL1, ABL2, ACVR1, AKT1, AKT2, AKT3, ALK, AR, ARAF, ARID1A, ATM, ATR, ATRX, AXL, BAP1, BARD1, BCL1, BLM, BRAF, BRCA1, BRD 1, BRIP1, BTK, C11orf 1, CCND1, CCNE1, CDK1, MDM 1, CDKN 11, CDKN 21, 685 1, 685K 1, KM K, 1, 685K 1, 685K 1, 685D, 1, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D 685, NOTCH2, NPM1, NRAS, NTRK1, NTRK2, NTRK3, PALB2, PDGFRA, PDGFRB, PGR, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PIK3R1, PMS2, POLD1, POLE, PTCH1, PTEN, RAB35, RAD50, RAD51, RAD52, RAF1, RARA, RB1, RET, RHEB, RICTOR, ROS1, SLX4, SMARCA4, SMARC 1, SMO, SRC, STK11, SYK, TMPRSS2, TOP2A, 53, TSC1, TSC2, VHL, XRCC 2.
Preferably, the tumor driver genes include: ABL1, ACVR1, ACVR1B, AKT1, ALK, AMER1, APC, AR, ARAF, ARID 11, ARID 51, ASXL1, ATM, ATRX, AXIN1, B2 1, BAP1, BCL2L1, BCOR, BRAF, BRCA1, BTG1, CARD1, CASP 1, CBFB, CCND1, CD79 1, CDH1, CDK1, KMK 1, KM K1, KM 1, 1, NCOR, NF, NFE2L, NOTCH, NPM, NRAS, NSD, NUP, PAX, PBRM, PDGFRA, PGR, PIK3R, PIM, PMS, POLE, PPM1, PPP2R1, PPP6, PRKAR1, PTCH, PTEN, PTPN, PTPRD, RAC, RAD, RAF, RARA, RASA, RB, RBM, RET, RHEB, RHOA, RIT, RNF, RRAS, RUNX, RXRRA, SETD, SF3B, SMAD, SMARCA, SOS, SOX, SPOP, SPTA, SF, STAG, SRK, TAF, TBEB, TCEB, TCF7L, TET, TGTGTGTP, VHTP, TSC, AIP, TSC, HAF, TSC, TMAF, TMAS, PTN, PTPN, PTPR, RTR, TSC, and TSC.
Preferably, the immunotherapy effect-related gene includes: TSC2, ATM, B2M, BRCA1, BRCA2, CCND1, DNMT3A, EGFR, FGF19, FGF3, FGF4, JAK1, JAK2, KRAS, MDM2, MDM4, MLH1, MSH2, MYC, PBRM1, PMS2, POLD1, POLE, PTEN, STK11, VHL, NF1, TP 53.
Preferably, the other genes that play an important role in the development of cancer include: DUSP, ETV, FOXP, GID, GRM, HIST1H3, HIST2H3, ICOSLG, IL, IRS, KNSTRN, MAGI, MAPKAP, MSI, NT5C, PAK, PMAIP, PPP4R, RFWD, RRAGC, SOX, TNFRSF, ZNF703, ANKRD, BABAM, E2F, EPCAM, ETV, FGF, FRS, HIST1H3, HIST3H, ID, IRS, MALT, MAX, MSI, NTHL, PAK, RAC, RANBP, RRAS, SGK, GSTM, TOP, TYRO, WWTR, BCORL, BTG, EED, HIST1H3, INHA, DNMST, MYOD, NKX-1, PRDM, PTP4A, SDHB, SH2B, SOX, TEST, SPEAP, SPE, SHCK 4, SHCK, SACK, SHCK 1H, SHCK, SACK, SHCK, SACK, SA, CD22, CENPA, EIF4A2, EPHB4, EZR, HIST1H3H, LTK, MAP3K13, NEGR1, NOTCH4, PARP3, PHOX2B, PIK3R3, PPARG, PRKCI, PTPRS, RECQL, SESN2, SLC34A2, SMYD3, SPRED1, STK19, TCF3, TIPARP, TRAF2, VTCN1, YES1, BCL2L1, CD74, CHD2, CSF3R, DNMT3B, PRRY F4E, HIST1H3I, INSR, MAP3K14, MEF2B, MYB, NUTM1, PIK3C2B, PRKD 685K 1, PRK 1, 6855, 1, 6855, 1, 6855, 1, 6855, 1, 6855, 1, 6855, 1, 6855, 1, 6855, 1, 6855, SACK 2K, 1, 6855, 1, 6855, 1, 6855, 1, SACK 2K, 1, 685.
The second aspect of the invention provides a probe Panel for detecting pan-cancer species, wherein the probe Panel is a detection probe for the tumor pathway related genes, the tumor genetic susceptibility genes, the tumor high-frequency mutation genes, the tumor targeting drug related genes, the tumor driving genes, the immune curative effect related genes, the key DDR pathway related genes and other genes playing an important role in the occurrence and development of cancer.
The third aspect of the invention provides the application of the gene Panel of the first aspect or the probe Panel of the second aspect in preparing a pan-cancer species detection device.
The invention has the beneficial effects that: the invention relates to Panel containing 561 gene, which can detect all biomarkers corresponding to solid tumor targeting drugs approved to be on the market by the national drug administration (NMPA)/American Food and Drug Administration (FDA) at one time. The biomarker for guiding accurate treatment is found by collecting tumor tissue samples and whole blood samples of the examined person, and performing tumor content evaluation, DNA extraction, library establishment, capture, sequencing, bioinformatics analysis and medical interpretation. Secondly, the prognosis of the patient can be predicted, and the relapse risk of the patient is layered by combining the clinical and pathological characteristics of the patient, so as to guide the adjuvant therapy. Finally, the genetic tumor of the detected person can be evaluated for prompting the family genetic risk, and early discovery, early diagnosis and early treatment can be realized.
Detailed Description
In order to better understand the above technical solutions, the above technical solutions are described in detail by specific embodiments below.
The invention aims to provide a detection Panel for pan-cancer species targeting, immunization, chemotherapy medication, molecular typing and genetic risk assessment. The invention utilizes the target region capture next-generation sequencing technology to analyze 561 all exon regions of genes related to tumor personalized medicine and genetic risk, hot spot regions of partial genes and chemotherapy drug related SNP sites. Therefore, biomarkers such as gene point mutation (SNV), small fragment insertion deletion (Indel), Copy Number Variation (CNV), gene Fusion (Fusion), tumor mutation load (TMB), microsatellite instability (MSI) and the like which are related to pan-cancer species (including but not limited to lung cancer, stomach cancer, intestinal cancer, endometrial cancer, ovarian cancer, breast cancer, prostate cancer, pancreatic cancer and the like) and have definite clinical significance are detected, and accurate treatment of the tumor is further guided. Meanwhile, the gene mutation detected by a tumor patient can be used for assisting the grouping of corresponding clinical tests. Provides guiding significance for clinical treatment, diagnosis and discovery of new targets.
The gene Panel provided by the embodiment of the invention mainly comprises the following genes:
genes related to the therapeutic effect of targeted drugs for solid tumors, which have been approved or approved by the National Cancer Complex Network (NCCN) guidelines and are to be approved or under investigation by the national drug administration (NMPA) or the U.S. Food and Drug Administration (FDA);
immunodetection point inhibitor therapeutic effect related genes, including genes positively correlated with therapeutic effect and genes correlated with drug resistance and risk;
core genes of DNA Damage Repair (DDR) pathways selected according to the National Cancer Complex Network (NCCN) guidelines, the american Society for Gynecological Oncology (SGO), etc.;
core genes of other important pathways during the development of cancer;
important driver genes for solid tumors;
frequently mutated genes of cancer species.
The gene Panel provided by the invention is a Panel product containing 561 genes, and comprises all biomarkers corresponding to solid tumor targeting drugs and immune drugs which are on the market at present. The gene Panel of the invention can be used for detecting all biomarkers corresponding to solid tumor targeted drugs which are approved to be on the market by the national drug administration (NMPA)/American Food and Drug Administration (FDA) at one time, thereby fully guiding the accurate treatment of tumors. The prognosis of the examined person can be evaluated, the relapse risk of the examined person is layered, and the postoperative adjuvant therapy is guided.
The gene Panel provided by the invention also comprises a gene related to pan-cancer hereditary tumor, so that the risk of hereditary tumor can be fully evaluated. When the subject has pathogenicity or possible pathogenicity heredity susceptibility gene mutation, healthy people who do not suffer from the disease in the family are recommended to carry out genetic evaluation, and the risk of the corresponding tumor is known in advance.
In addition, the gene Panel provided by the invention also comprises genes related to pan-carcinogenesis and development mechanisms, and the practicability is higher than that of whole exon sequencing. Valuable information can be provided both in clinical medication guidance and in subsequent biomarker detection.
In the specific application process, the biomarker for guiding accurate treatment can be found by collecting tumor tissue samples and whole blood samples of the detected persons, and performing tumor content evaluation, DNA extraction, library building, capturing, sequencing, bioinformatics analysis and medical interpretation. Secondly, the prognosis of the patient can be predicted, and the relapse risk of the patient is layered by combining the clinical and pathological characteristics of the patient, so as to guide the adjuvant therapy. Finally, the genetic tumor of the detected person can be evaluated for prompting the family genetic risk, and early discovery, early diagnosis and early treatment can be realized.
In the embodiment of the present invention, the following method may be specifically adopted for detection:
extracting DNA of a detection sample;
constructing a detection sample DNA library;
constructing a detection probe of the gene Panel as described above;
hybridizing the detection probe with the DNA library for capturing, eluting, purifying and sequencing;
performing bioinformatics analysis on the sequencing result to obtain mutation information of the detection sample;
and judging the pathogenicity of the mutation information.
Wherein the sample DNA comprises blood sample DNA and tumor tissue sample DNA. The following steps are mainly adopted for constructing the DNA library: DNA breaking, end repairing, joint connection, library amplification and purification.
In addition, in the embodiment of the present invention, some parameters that may affect the detection result are verified and evaluated, which specifically include:
1. initial volume assessment of reservoir building
The effect of different starting amounts on the test results was evaluated. 3 samples were taken and 30ng, 50ng and 100ng of starting pools were used, respectively, with three replicates per sample. And (4) comparing the influence of the initial amount on the detection result. Thereby evaluating whether the quality control of the sample passes and whether the target variation is detected under different initial quantities. The results showed that the sample quality control passed under the condition of 30ng and all the objective mutations were detected, so that the minimum input amount was 30 ng.
2. Positive match rate assessment
22 samples were tested, the types of mutations included Single Nucleotide Variation (SNV), small fragment insertion deletion (Indel) and Copy Number Variation (CNV), and the samples were obtained from clinical specimens. All mutations were verified by digital PCR or first-generation sequencing, QPCR, etc. The results showed that all the target variations were detected in total, i.e., the positive site coincidence rate was 100%.
3. Negative match rate evaluation
And (3) detecting 20 negative samples, wherein the sample source is a clinical sample, and evaluating whether a target positive variation is detected. The results showed that all the target variants were detected, i.e., the negative match rate was 100%.
4. Minimum detection limit evaluation
4 samples were selected, the types of variation including Single Nucleotide Variation (SNV), small fragment insertion deletion (Indel), Copy Number Variation (CNV) and Fusion (Fusion), and the sample sources were standard and clinical samples. Each mutation frequency was performed in 3 replicates. Setting mutation frequency: the expected frequencies for SNV and Indel settings were 1%, 2%, 5%, CNV settings were 2.5, 3, 3.5 copies, and Fusion settings were 1%, 2%, 5%. Finally, the lowest expected frequency of all the mutation sites is used as the lowest detection limit.
The final results show that:
1) the SNV mutation site frequency is detected in all sites of 2% or more, so the minimum detection limit of SNV is 2%.
2) The Indel mutation sites are detected in all sites with a frequency of 5% or more, and thus the minimum detection limit of Indel is 5%.
3) The CNV variant copy number was all detected at3 or more, and therefore the CNV minimum detection limit was 3.
4) Fusion mutation sites were detected at a frequency of 5% or more, and thus the minimum detection limit of Fusion was 5%.
5. Repeatability verification
6 clinical specimens were selected and tested for 3 intra-and 3 inter-batch replicates. The results show that the repeatability measurements within and between batches of the same sample are consistent.
6. Interferent analysis
6 samples were taken and the interfering substances bilirubin (concentration 342. mu.M/L), triglyceride (concentration 37 mM/L), hemoglobin (concentration 2 g/L) and 80% ethanol were added, 3 replicates per sample per treatment. The results show that: bilirubin (concentration 342. mu.M/L), triglyceride (concentration 342 mM/L), hemoglobin (concentration 2 g/L) and 80% ethanol all had no effect on the assay results.
7. DNA quality assessment
19 samples of different quality grades were selected and 100ng of DNA was used for the construction of the library. Thereby evaluating the effect of samples of different masses on mutation detection. The mutation sites of the samples with DNA main bands more than 500bp can be detected.
8. Tumor cell proportion evaluation
The verified tissue samples have 20% -90% of tumor content, and all positive sites are detected. Namely, the Panel can detect samples with the tumor content of more than or equal to 20 percent.
9. Sample stability assessment
The tissue samples used in the verification are collected from 3 months to 2021 months in 2019, and all positive sites are detected. Namely, the Panel can detect tissue samples with the storage time less than 2 years.
DETAILED DESCRIPTION OF EMBODIMENT (S) OF INVENTION
The invention is exemplified by the enriched 561 gene segment used for mutation detection of cancer patients based on next generation sequencing technology to guide the patients to be treated accurately.
The detection process mainly comprises the following steps: preparing a DNA sample library, constructing the DNA library, hybridizing a detection probe with the DNA library, detecting based on a next generation sequencing technology, analyzing and identifying mutation, interpreting the mutation and giving a report according to the interpretation result.
The method comprises the following steps: and extracting detection sample DNA (the sample DNA comprises blood sample DNA and tumor tissue sample DNA).
Step two: and (5) constructing a DNA library of the test sample. The method mainly comprises the following steps: DNA breaking, end repairing, joint connection, library amplification and purification. The method can be implemented according to the following steps:
1. sample preparation
For paraffin-embedded Tissue samples, QIAamp DNA FFPE Tissue Kit from QIAGEN was used, and for Blood samples, QIAamp DNA Blood Mini Kit was used, and the procedures were strictly followed. The DNA needs to be quantitatively detected, the extracted DNA is detected by using the Qubit dsDNA HS Assay Kit and a matched instrument, and the total extraction amount is not less than 50 ng.
2. DNA library construction
2.1 sample interruption
1) A breaking system as shown in table 1 was used:
Figure 523052DEST_PATH_IMAGE001
2) FFPE DNA was interrupted using Covaris and blood cell genomic DNA was interrupted using Covaris or Bioruptor.
The Covaris interruption operation is as follows:
a. preparing a Covaris broken tube, marking the serial number of the DNA sample on the tube cover
b. Adding TE Buffer pH 8.0 and sample into the interrupt tube
c. Samples were interrupted and the interruption instrument interruption parameters are shown in table 2:
Figure 423881DEST_PATH_IMAGE002
② Bioruptor interruption operation as follows:
a. prepare Bioruptor-cut tube, label DNA sample number on tube cap
b. TE Buffer pH 8.0 and sample were added to the disrupted tube
c. Samples were interrupted and the interruption instrument interruption parameters are shown in table 3:
Figure 672460DEST_PATH_IMAGE003
d. the broken DNA is subjected to quality inspection by 2% agarose gel electrophoresis, 50bp DNA Ladder and D2000 DNA Ladder are used, the voltage is 150V, and the electrophoresis band is qualified within 200bp-300 bp.
2.2 end repair
1) End Repair Mix was prepared as per table 4.
Figure 739773DEST_PATH_IMAGE004
2) The End Repair reaction program was set with reference to table 5, and the hot lid temperature was adjusted to 40 ℃.
Figure 272254DEST_PATH_IMAGE005
3) The sample was placed in a PCR instrument and the End Repair reaction program was run.
4) The used reagents were returned to the original kit and stored at-20 ℃.
2.3 preparation of Ligation1 Mix
Ligation1 Mix was prepared as in table 6.
Figure 829138DEST_PATH_IMAGE006
2.4 purification after repair of the termini
1) The AMPure XP magnetic beads are taken out of a refrigerator at4 ℃, and the temperature is balanced for 30 min.
2) 147.5. mu.L (2.5X) of AMPure XP magnetic beads were added to the end-repair sample and mixed well.
3) Standing at room temperature for 10min, transferring to magnetic rack, standing for 5min until the liquid is completely clarified, discarding the supernatant, and paying attention to avoid sucking magnetic beads.
4) 160 μ L of 80% ethanol was added slowly along the tube side walls, left to stand for 30s, and the supernatant was removed using a pipette.
5) Repeating the step 4) once.
6) The residual ethanol was removed by pipetting with a 10. mu.L pipette, and the mixture was left to dry at room temperature for 3 min.
7) Add 30. mu.L Ligation1 Mix to the sample tube, shake and Mix, and centrifuge instantaneously.
2.5、Ligation 1
1) The Ligation1 reaction program was set up with reference to Table 7, and the hot lid temperature was adjusted to 70 ℃.
Figure 768275DEST_PATH_IMAGE007
Note: after the end of the process, the sample can be stored at4 ℃ for no more than 2 h!
2) The Ligation1 reaction procedure was run.
2.6、Ligation 2
1) Ligation 2 Mix was prepared according to table 8.
Figure 956723DEST_PATH_IMAGE008
2) The PCR reaction tube of Ligation1 was removed from the PCR apparatus, subjected to instantaneous centrifugation, and placed on ice. Each reaction tube is subpackaged with 10 mu L of Ligation 2 Mix, evenly mixed by oscillation and instantaneously centrifuged.
3) The Ligation 2 reaction program was set up with reference to table 9, and the hot lid temperature was adjusted to 70 ℃.
Figure 297705DEST_PATH_IMAGE009
4) The sample was placed in a PCR instrument and the Ligation 2 reaction program was run.
2.7 purification after Adaptor ligation
1) Add 100. mu.L (2.5X) PEG/NaCl to the reacted sample after Ligation 2 and mix well.
2) Standing at room temperature for 10min, transferring to magnetic rack, standing for 5min until the liquid is completely clarified, discarding the supernatant, and paying attention to avoid sucking magnetic beads.
3) 160 μ L of 80% ethanol was added slowly along the tube side walls, left to stand for 30s, and the supernatant was removed using a pipette.
4) Repeating the step 3) once.
5) The residual ethanol was removed by pipetting with a 10. mu.L pipette, and the mixture was left to dry at room temperature for 3 min.
6) Add 20. mu.L of nucleic-Free Water to the sample tube, mix well with shaking, and centrifuge instantaneously.
2.8 PCR amplification
1) PCR reaction systems were prepared in PCR tubes according to the system in Table 10.
Figure 759911DEST_PATH_IMAGE010
2) Shaking and mixing evenly, and carrying out instantaneous centrifugation to ensure that all reaction liquid is placed at the bottom of the PCR tube.
PCR amplification procedure is as in table 11:
Figure 497928DEST_PATH_IMAGE011
2.9 library purification and quantification
1) Taking out AMPure XP magnetic beads from a refrigerator at4 ℃, and balancing for 30min at room temperature.
2) The PCR product was removed from the PCR instrument, centrifuged instantaneously, placed on a magnetic stand and allowed to stand for 5min, and the supernatant was transferred to a new tube.
3) Add 65. mu.L (1.3X) of AMPure XP magnetic beads to the PCR product, pipette (range 65. mu.L) down to 20, mix well.
4) Standing at room temperature for 10min, transferring to magnetic rack, standing for 5min until the liquid is completely clarified, discarding the supernatant, and paying attention to avoid sucking magnetic beads.
5) 160 μ L of 80% ethanol was added slowly along the tube side walls, left to stand for 30s, and the supernatant was removed using a pipette.
Note: if the magnetic beads are absorbed when the waste liquid is discarded, standing for 2min, and discarding the supernatant after the magnetic beads are absorbed.
6) Repeating the step 5) once.
7) The residual ethanol was removed by pipetting with a 10. mu.L pipette, and the mixture was left to dry at room temperature for 3 min.
8) Add 32. mu.L TE Buffer PH 8.0 to the sample tube, mix well with shaking, centrifuge instantaneously, incubate for 8min at room temperature.
9) The sample tube was placed on a magnetic rack for 5min until the liquid was completely clear, and 30 μ Ι _ of supernatant was carefully transferred to a new 1.5ml centrifuge tube using a pipettor.
10) The library, Qubit, was quantified using Qubit 4.0.
2.10 library quality testing, the results are shown in Table 12.
Figure 438203DEST_PATH_IMAGE012
2.11, storing the library, and storing the library in a refrigerator at-20 ℃ in an amplification area.
Step three: constructing a detection probe for detecting Panel, hybridizing with a DNA library, capturing, eluting, purifying and sequencing. Specifically, in the embodiment of the present invention, 30465 IDT probes were designed and manufactured by using the Target Capture Probe Design & Ordering Tool of the website of Integrated DNA Technologies IDT corporation.
Specifically, the method can be carried out as follows.
Library hybridization procedures were as follows:
1. shaking and uniformly mixing the Human Cot DNA and xGen Universal blocks-TS Mix, and instantaneously centrifuging.
2. Hybridization reaction Mix was prepared as shown in Table 13.
Figure 430429DEST_PATH_IMAGE013
3. Add 7. mu.L Mix and 500ng-1ug DNA library to the centrifuge tube, Mix well with shaking, and centrifuge instantaneously. And drying in a vacuum concentrator for later use.
4. XGen according to Table 14 ®2x Hybridization Buffer and xGen ®And (3) oscillating and uniformly mixing the 2x Hyb Buffer Enhancer, and performing instantaneous centrifugation to obtain a hybridization reaction solution.
Figure 594694DEST_PATH_IMAGE014
5. And adding the hybridization reaction solution and 4 mu L of probe into the evaporated sample, and incubating for 10min at 25 ℃ on a constant-temperature mixing machine.
6. And (5) putting the sample into a PCR instrument, clicking the program and starting to operate. PCR procedure as in table 15:
Figure 757691DEST_PATH_IMAGE015
the library elution procedure was as follows:
1. streptavidin magnetic bead cleaning
1) Will xGen®2X Hybridization Buffer and xGen 2X Hyb Buffer Enhancer are shaken, mixed evenly and centrifuged instantly.
2) The magnetic bead suspension Mix was prepared as in table 16.
Figure 970498DEST_PATH_IMAGE016
3) Dynabeads M-270 Streptavidin was shaken and mixed well. The amount of Dynabeads M-270 Streptavidin used per library was 50. mu.L.
4) The using amount of each library 1XBead Wash Buffer (nuclear-Free Water and xGen 2X Bead Wash Buffer prepared according to the proportion of 1: 1) is 100 muL, 1X Bead Wash Buffer with the corresponding volume is added into a tube, the mixture is oscillated and mixed evenly (n X100 muL), the mixture is subjected to instantaneous centrifugation and placed in a magnetic frame for 1min, and after the liquid is completely clarified, the supernatant is sucked and discarded by using a pipette.
5) Repeating the step 4) twice.
6) The residual liquid was aspirated with a pipette.
7) The amount of each library bead suspension was 17. mu.L, and a corresponding volume of bead suspension was added to the tube for suspension.
8) A corresponding number of PCR tubes were prepared, and 17. mu.L of a mixture of magnetic beads and a suspension was dispensed into each of the PCR tubes.
9) The PCR tube containing the magnetic bead suspension was placed in a PCR instrument and incubated for 5 min.
2. Streptavidin magnetic bead capture
1) After 4-16h of hybridization reaction, the resuspended streptavidin magnetic beads are added into the hybridization system twice after being mixed evenly. Transfer the sample tube to the running PCR instrument and click next.
2) Incubate at 65 ℃ for 45min, shake and mix once every 15min, ensure the magnetic beads are completely resuspended.
3. Thermal elution
1) Turn on PCR Instrument setup program according to Table 17
Figure 597657DEST_PATH_IMAGE017
2) The filled containers will contain 1X WashBuffer I (nucleic-Free Water and xGen)®10 × Wash Buffer I prepared in a ratio of 1: 9) and 1X Stringent Wash Buffer (Nuclear-Free Water and xGen)®10X Stringent Wash Buffer in a 1:9 ratio) was placed in a PCR apparatus and preheated.
3) After incubation at 65 ℃ for 45min, transferring the hybridization sample captured by the streptavidin magnetic beads into a PCR tube filled with 1X Wash Buffer I and mixing uniformly. The PCR tube was placed on a magnetic stand for 1min, and after the liquid was completely clarified, the supernatant was removed thoroughly with a pipette.
4) Transferring the 1X Stringent Wash Buffer into a PCR tube filled with sample magnetic beads, mixing uniformly, covering the tube cap tightly, and putting the tube cap into a PCR instrument to incubate for 5min at 65 ℃. After the time, the PCR tube with the sample is placed on a magnetic rack until clarified, and the supernatant is removed thoroughly with a pipette.
5) Repeating the step 4) once.
4. Eluting at normal temperature
1) The PCR tube containing the sample was removed from the magnetic stand, and 150ul of 1 XWash Buffer II (nucleic-Free Water and xGen) was pipetted from the dispensing tube®10x Wash Buffer II in a ratio of 1: 9) into a PCR tube filled with magnetic beads, incubating at room temperature for 2min, uniformly mixing for 30s in a constant-temperature mixer, standing for 30s, and alternately mixing to ensure sufficient mixing. The PCR tube was centrifuged instantaneously and then allowed to stand on a magnetic stand for 1min, and the supernatant was removed thoroughly with a pipette.
2) The PCR tube containing the sample was removed from the magnetic rack, subjected to instantaneous centrifugation, and then placed on the magnetic rack, and a small amount of residual Buffer was removed by a pipette.
3) Add 21. mu.L of Nuclear-Free Water to the tube, suck and mix.
4) KAPA HiFi HotStart ReadyMix, primers FCF (10. mu.M) and FCR (10. mu.M) were thawed 15-20min in advance at4 ℃.
5) The reaction system was prepared in PCR tubes or centrifuge tubes placed on ice-boxes as per Table 18.
Figure 401665DEST_PATH_IMAGE018
6) And (3) subpackaging the configured MIX into a PCR tube with a labeled sample serial number.
7) Transfer 20. mu.L of the product with magnetic beads to the corresponding PCR tube with a pipette, and mix well.
8) The procedure shown in table 19 was started on a PCR instrument:
Figure 802691DEST_PATH_IMAGE019
library purification steps were as follows:
1) and (3) oscillating and uniformly mixing the AMPure XP magnetic beads, sucking 60 mu L of the mixture and adding the mixture into a labeled PCR tube.
2) After PCR amplification, the PCR tube was taken out and placed in a magnetic rack for 2min, and after the liquid was completely clarified, the supernatant was transferred to the PCR tube in the magnetic rack.
3) Shaking and mixing, and incubating at room temperature for 10 min.
4) The PCR tube was centrifuged instantaneously and placed on a magnetic stand for 5min, after the liquid was completely clarified, the supernatant was removed (5-10. mu.L of liquid was left).
5) 200 μ L of 80% ethanol was added slowly along the side walls of the PCR tube, left to stand for 30s, and the supernatant was removed using a pipette.
6) Repeating the step 5) once.
7) The PCR tube was removed from the magnetic rack, placed on the magnetic rack after transient centrifugation, and a small amount of residual ethanol was removed using a pipette, taking care not to attract the magnetic beads.
8) And opening a PCR tube cover, and placing the magnetic beads at room temperature for drying, wherein the surfaces of the magnetic beads have no liquid to reflect light, and the magnetic beads cannot be dried excessively.
9) Add 31. mu.L of TE Buffer pH 8.0 to the PCR tube, mix well with shaking, incubate for 5min at room temperature.
10) The PCR tube was centrifuged briefly and placed on a magnetic rack for 1-2min until the liquid was clear, and 30. mu.L of the supernatant was carefully transferred to a new 1.5mL centrifuge tube with a pipette, taking care not to attract the beads.
11) Performing library quality inspection, and performing machine sequencing after the library quality inspection is qualified.
Step four: and (4) performing bioinformatics analysis on the sequencing result to obtain a mutation result of the detection sample. The method can be specifically implemented by the following steps:
and removing the adaptor sequence and the low-quality base sequence introduced in the experiment and sequencing links by using fastp (v0.19.4) software to obtain high-quality sequencing data. Wherein, the required data quality meets Q30 more than or equal to 80%, otherwise, the quality control is judged not to pass.
The above-described data satisfying the requirements were aligned to hg19 (GRCh 37) reference genome using bwa (0.7.17) sequence alignment software, generating BAM format files recording the alignment results. BAM files were then sorted, de-duplicated, and base quality corrected using Samtools (v 1.9) and genoanalysis tk (v 4.1.0) to obtain the final BAM file, and subsequent mutation analysis was performed based on this file.
Wherein, the comparison rate of the reference genome is more than or equal to 90 percent, the average sequencing depth of the target area of the control sample is more than or equal to 100 times, the average sequencing depth of the target area of the tumor sample is more than or equal to 500 times, the proportion of sites with the depth in the target area larger than the average depth multiplied by 0.2 is more than or equal to 90 percent, and the matching consistency of the tumor and the control sample is more than or equal to 90 percent. If any of the conditions is not met, the quality control is judged to fail, and the experiment needs to be carried out again.
Analyzing point mutation and insertion deletion mutation in a sample by using a variation identification module of genomeAnalystk (v4.1.0); analyzing the gene fusion event in the sample by using a fusion analysis module; copy number variation analysis modules (including the cnvkit (v 0.9.6) software and filter modules) were used to analyze copy number variation in samples. Resulting in mutations that can eventually enter the annotation process.
The resulting mutations were annotated as follows: the point mutation, small fragment insertion deletion, gene fusion and copy number variation are annotated by using a variation annotation module built based on Annovar (v2018.04.16), and databases used for annotation comprise clinvar, cosmic, 1000 genes and the like.
In addition, TMB (tumor mutation burden) values were calculated and samples were analyzed for microsatellite instability (MSI) using a microsatellite instability analysis module.
Step five: and (5) reading mutation results, and judging the pathogenicity of the mutation. Based on the annotated results of the variation in step four, the variation was classified for detected non-benign/likely-benign variations according to the classification criteria of variation as specified in ACMG genetic variation classification criteria and Guidelines and the Cancer variation Interpretation Guidelines (Standards and Guidelines for the Interpretation and Reporting of Sequence Variants in Cancer) jointly issued by the american association of pathology (AMP)/american clinical oncology society of oncology (ACSO)/american society of pathologists (CAP). As an example, such as: in the following colorectal cancer samples, KRAS c.35g > ap.g12d variation was detected in exon 2 of KRAS gene, and patients with this variation could not be treated with cetuximab and panitumumab according to the national cancer complex Network for Colorectal Cancer (NCCN) guidelines and the chinese clinical oncology society of cancer (CSCO) guidelines for colorectal cancer diagnosis and treatment. Finally classifying the variation as a type I variation according to the Cancer variation Interpretation guideline (Standards and Guidelines for the Interpretation and Reporting of Sequence Variants in Cancer) jointly issued by the American Association of Pathology Association (AMP)/American society of clinical Oncology (ACSO)/American society of pathologists (CAP), thereby providing a report and suggesting that the patient is resistant to cetuximab and panitumumab.
In practical application, the above steps are adopted to detect a sample of a patient with endometrial cancer, and the result is: microsatellite instability was high (MSI-H) and tumor mutational load (TMB) was 298.520 mut/Mb, which is attributed to high TMB. Solid tumor patients with high TMB or high microsatellite instability may benefit from curitant (pappaluzumab) according to approval by the united states Food and Drug Administration (FDA). The patients were treated by selecting kreota in 2018 at3 months, and the injection was performed once every 21 days, as the results of which the CA125 level decreased to within the normal range after two treatment courses and remained within the normal range thereafter. 2018-3-30, 2018-5-9 and 2019-3-11, and the results of abdominal ultrasound contrast in three stages show that the lesion is clearly seen to shrink earlier and then disappear.
The above steps are adopted to detect samples of patients with ovarian cancer, and the result is as follows: the tumor mutational load (TMB) was 15.162mut/Mb, which is high in TMB. TMB high solid tumor patients may benefit from curitant (pappalizumab) according to united states Food and Drug Administration (FDA) approval. Patients were treated with kreota as suggested by the test results, and CA125 returned to normal levels (from 179.2U/ml to 18.7U/ml) during treatment, with a significant reduction in metastatic foci (by more than 30%) and Partial Remission (PR).
The sample of the colorectal cancer patient is detected by adopting the steps, and the result is as follows: the variation KRAS c.35G > A p.G12D is detected, and the variation abundance is 17.92%. According to the National Cancer Complex Network (NCCN) guidelines for colorectal cancer and the chinese clinical oncology institute of technology (CSCO) guidelines for colorectal cancer diagnosis and treatment, patients with this variation cannot be treated with cetuximab and panitumumab. The patient may be treated in consideration of the remaining modalities, such as conventional chemotherapy.
Therefore, the technical scheme provided by the invention can be used for detecting pan-cancer species, and the detection result has a positive guiding effect on accurate treatment.
While preferred embodiments of the present invention have been described, additional variations and modifications in those embodiments may occur to those skilled in the art once they learn of the basic inventive concepts. Therefore, it is intended that the appended claims be interpreted as including preferred embodiments and all such alterations and modifications as fall within the scope of the invention. It will be apparent to those skilled in the art that various changes and modifications may be made in the present invention without departing from the spirit and scope of the invention. Thus, if such modifications and variations of the present invention fall within the scope of the claims of the present invention and their equivalents, the present invention is also intended to include such modifications and variations.

Claims (9)

1. A gene Panel for detecting pan-cancer species is characterized by comprising tumor pathway related genes, tumor genetic susceptibility genes, tumor high-frequency mutation genes, tumor targeting drug related genes, tumor driving genes, immunity curative effect related genes, key DDR pathway related genes and other genes playing important roles in the occurrence and development of cancer; the key DDR pathway related genes comprise: ATM, ATR, BARD1, BLM, BRCA1, BRCA2, BRIP1, CHEK1, CHEK2, ERCC2, ERCC3, ERCC4, ERCC5, FAM175A, FANCA, FANCC, FANCD2, FANCL, MDC1, MLH1, MRE11A, MSH2, MSH3, MSH6, NBN, PALB2, PARP1, PMS1, PMS2, POLE, PRKDC, PTEN, RAD50, RAD51, RAD51B, RAD51C, RAD51D, RAD52, RAD54L, 53, TP53BP1, XRCC 2; such other genes that play an important role in the development of cancer include: DUSP, ETV, FOXP, GID, GRM, HIST1H3, HIST2H3, ICOSLG, IL, IRS, KNSTRN, MAGI, MAPKAP, MSI, NT5C, PAK, PMAIP, PPP4R, RFWD, RRAGC, SOX, TNFRSF, ZNF703, ANKRD, BABAM, E2F, EPCAM, ETV, FGF, FRS, HIST1H3, HIST3H, ID, IRS, MALT, MAX, MSI, NTHL, PAK, RAC, RANBP, RRAS, SGK, GSTM, TOP, TYRO, WWTR, BCORL, BTG, EED, HIST1H3, INHA, MST, MYOD, NKX-1, PRDM, PTP4A, SDHB, SH2B, SOX, TET, SPEAP, SPEP, SHCK 1H, SHCK 1H3, SHCK, SACK, SADDS, SA, CENPA, EIF4A, EPHB, EZR, HIST1H3, LTK, MAP3K, NEGR, NOTCH, PARP, PHOX2, PIK3R, PPARG, PRKCI, PTPRS, RECQL, SESN, SLC34A, SMYD, SPRED, STK, TCF, TIPARP, TRAF, VTCN, YES, BCL2L, CD, CHD, CSF3, DNMT3, EIF4, HIST1H3, INSR, GRIN MAP3K, MEF2, MYB, NUTM, PIK3C2, PRKD, PTPRT, RECQL, RPS6KA, SESN, SLIT, CAIP, STK, TMEM127, TRAIFF, ZBTB, FOKML, HIST1H2, HIST1H3, P2, PIRY 3C2, PRXB 3, SDCK 6, SUBCK 2, PRXO, SLC 2, FORCH 2, PRXB, PRXC, PRXK, SLC 6, SLC 2, FORCH 2, FORCK, FORCH 3, FORCH 2, FORCK, FORCH 2, FORCK, FORCH, FORCK, FORCH 2, FORCH 3, FORCK, FORCH 2, FORCK, FORCH 2, FORCH, FORCK, FORCH 2, FORCH 2, FORCH 2, FORCH, FORC.
2. The gene Panel for detecting pan-cancer species of claim 1 wherein the tumor pathway associated gene comprises: AKT1, AKT2, AKT3, ALK, APC, AR, ARAF, ASXL1, ATM, ATR, ATRX, AURKA, AURKB, AXIN1, BCL2, BRAF, BRCA1, BRCA2, BRIP 2, BTK, CASP 2, CBL, CCND2, CCNE 2, CD79, CDK 2, KN 12, KN 22, MDM2, CDKN 22, CHEK2, CRKL 2, CRLF2, CSCSF 12, CTNNA 2, CTNNB 2, MDM2, 685MT 3, DOT 2, 685 2, 685 2, 685 2, 685, 2, 685 2, 685, 2, 685 2, 685 2, 685, 685 2, 685 2, 685 2, 685 2, 685 2, 685 2, 685, 2, 685 2, 685, 2, 685 2, 685 2, 685, 685, 685, 685 2, 685, 685, 685 2, 685, 685, 2, 685 2, 685, 685, 685, 685, 685, 685, 685, 685, 685, 685, 685, 685, PDGFRA, PDGFRB, PGR, PIK3CA, PIK3CG, PIK3R1, PIK3R2, PTCH1, PTEN, PTPN11, RAD50, RAD51, RB1, RET, RICTOR, RNF43, ROS1, RPTOR, SETD2, SMAD2, SMAD3, SMAD4, SMO, SOCS1, SRC, STAG2, STAT3, STAT4, STAT5A, STAT5B, SUFU, TET2, TGFBR1, TGFBR2, TNFAIP3, TP53, TSC1, TSC2, TSHR, WISP3, WT 1.
3. The gene Panel for detecting pan-cancer species of claim 1, wherein said tumor genetic susceptibility gene comprises: ALK, APC, ATM, AXIN, BMPR1, BRCA, BRIP, CDH, CDKN2, CHEK, FH, FLCN, GREM, KIT, MEN, MET, MLH, MSH, MUTYH, NBN, NF, PALB, PMS, POLD, POLE, PTEN, RAD51, RB, SMAD, STK, TP, TSC, VHL, SDHD, TSC, PDGFRA, BARD, CDK, RET, SMARCA, SMARCB, SDHA, PRKAR1, HOXB, BRAF, PTCH, SDHAF.
4. The gene Panel for detecting pan-cancer species according to claim 1, wherein said tumor high-frequency mutant gene comprises: ACVR1B, AKT1, AMER1, APC, ARID 11, ARID1, ASXL1, ATM, ATRX, AXIN1, BAP1, BCL2L1, BCOR, BRA 1, BRCA1, BTG1, CASP 1, CBFB, CCND1, CCNE1, CD274, CDH1, CDK1, KN 11, MDM 11, CDKN K2 1, CHD 1, CREBB 1, CTNNA1, CTB 1, EGFR, ELF 1, EP300, ERBB4, ERBB 1, ERCC 1, ERBG 1, ERBBD 1, KM 1, 1, STK11, TBX3, TCF7L2, TERC, TERT, TGFBR2, TMPRSS2, TNFAIP3, TP53, TSC1, TSC2, U2AF1, VEGFA, VHL, WHSC1L 1.
5. The gene Panel for detecting pan-cancer species of claim 1 wherein the tumor-targeting drug-associated gene comprises: ABL1, ABL2, ACVR1, AKT1, AKT2, AKT3, ALK, AR, ARAF, ARID1A, ATM, ATR, ATRX, AXL, BAP1, BARD1, BCL1, BLM, BRAF, BRCA1, BRD 1, BRIP1, BTK, C11orf 1, CCND1, CCNE1, CDK1, MDM 1, CDKN 11, CDKN 21, 685 1, 685K 1, KM K, 1, 685K 1, 685K 1, 685D, 1, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D, 685D 685, NOTCH2, NPM1, NRAS, NTRK1, NTRK2, NTRK3, PALB2, PDGFRA, PDGFRB, PGR, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PIK3R1, PMS2, POLD1, POLE, PTCH1, PTEN, RAB35, RAD50, RAD51, RAD52, RAF1, RARA, RB1, RET, RHEB, RICTOR, ROS1, SLX4, SMARCA4, SMARC 1, SMO, SRC, STK11, SYK, TMPRSS2, TOP2A, 53, TSC1, TSC2, VHL, XRCC 2.
6. The gene Panel for detecting pan-cancer species of claim 1, wherein the tumor driver gene comprises: ABL1, ACVR1, ACVR1B, AKT1, ALK, AMER1, APC, AR, ARAF, ARID 11, ARID 51, ASXL1, ATM, ATRX, AXIN1, B2 1, BAP1, BCL2L1, BCOR, BRAF, BRCA1, BTG1, CARD1, CASP 1, CBFB, CCND1, CD79 1, CDH1, CDK1, KMK 1, KM K1, KM 1, 1, NCOR, NF, NFE2L, NOTCH, NPM, NRAS, NSD, NUP, PAX, PBRM, PDGFRA, PGR, PIK3R, PIM, PMS, POLE, PPM1, PPP2R1, PPP6, PRKAR1, PTCH, PTEN, PTPN, PTPRD, RAC, RAD, RAF, RARA, RASA, RB, RBM, RET, RHEB, RHOA, RIT, RNF, RRAS, RUNX, RXRRA, SETD, SF3B, SMAD, SMARCA, SOS, SOX, SPOP, SPTA, SF, STAG, SRK, TAF, TBEB, TCEB, TCF7L, TET, TGTGTGTP, VHTP, TSC, AIP, TSC, HAF, TSC, TMAF, TMAS, PTN, PTPN, PTPR, RTR, TSC, and TSC.
7. The gene Panel for detecting pan-cancer species according to claim 1, wherein said immunotherapy efficacy-related gene comprises: TSC2, ATM, B2M, BRCA1, BRCA2, CCND1, DNMT3A, EGFR, FGF19, FGF3, FGF4, JAK1, JAK2, KRAS, MDM2, MDM4, MLH1, MSH2, MYC, PBRM1, PMS2, POLD1, POLE, PTEN, STK11, VHL, NF1, TP 53.
8. A probe Panel for detecting pan-cancer species, wherein the probe Panel is a detection probe for the tumor pathway-associated gene, the tumor genetic susceptibility gene, the tumor high-frequency mutation gene, the tumor-targeted drug-associated gene, the tumor driver gene, the immune efficacy-associated gene, the key DDR pathway-associated gene, and other genes playing important roles in the development of cancer according to any one of claims 1 to 7.
9. Use of the gene Panel according to any one of claims 1 to 7 or the probe Panel according to claim 8 for the preparation of a pan-cancer species detection device.
CN202210340759.9A 2022-04-02 2022-04-02 Gene Panel for detecting pan-cancer species, probe and application Pending CN114480660A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210340759.9A CN114480660A (en) 2022-04-02 2022-04-02 Gene Panel for detecting pan-cancer species, probe and application

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210340759.9A CN114480660A (en) 2022-04-02 2022-04-02 Gene Panel for detecting pan-cancer species, probe and application

Publications (1)

Publication Number Publication Date
CN114480660A true CN114480660A (en) 2022-05-13

Family

ID=81487673

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210340759.9A Pending CN114480660A (en) 2022-04-02 2022-04-02 Gene Panel for detecting pan-cancer species, probe and application

Country Status (1)

Country Link
CN (1) CN114480660A (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115449555A (en) * 2022-10-26 2022-12-09 山东大学 Application of ADGRA2 as breast cancer chemotherapy curative effect and prognosis evaluation biomarker
CN116287266A (en) * 2023-03-07 2023-06-23 江苏先声医学诊断有限公司 Application of DNA replication advanced region in diagnosis of flood cancer
CN117230203A (en) * 2023-11-14 2023-12-15 首都医科大学附属北京儿童医院 Primer probe set for detecting MYOD1 gene mutation and kit thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105986031A (en) * 2016-02-04 2016-10-05 北京肿瘤医院 Tumor susceptibility 62 genes and application thereof
US20180089373A1 (en) * 2016-09-23 2018-03-29 Driver, Inc. Integrated systems and methods for automated processing and analysis of biological samples, clinical information processing and clinical trial matching
CN109609647A (en) * 2019-01-25 2019-04-12 臻悦生物科技江苏有限公司 Detection Panel, detection kit and its application for the targeting of general cancer kind, chemotherapy and immune medication based on the sequencing of two generations
US20190256924A1 (en) * 2017-08-07 2019-08-22 The Johns Hopkins University Methods and materials for assessing and treating cancer
US20200010468A1 (en) * 2018-07-06 2020-01-09 Kymera Therapeutics, Inc. Tricyclic crbn ligands and uses thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105986031A (en) * 2016-02-04 2016-10-05 北京肿瘤医院 Tumor susceptibility 62 genes and application thereof
US20180089373A1 (en) * 2016-09-23 2018-03-29 Driver, Inc. Integrated systems and methods for automated processing and analysis of biological samples, clinical information processing and clinical trial matching
US20190256924A1 (en) * 2017-08-07 2019-08-22 The Johns Hopkins University Methods and materials for assessing and treating cancer
US20200010468A1 (en) * 2018-07-06 2020-01-09 Kymera Therapeutics, Inc. Tricyclic crbn ligands and uses thereof
CN109609647A (en) * 2019-01-25 2019-04-12 臻悦生物科技江苏有限公司 Detection Panel, detection kit and its application for the targeting of general cancer kind, chemotherapy and immune medication based on the sequencing of two generations

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
WING CHAN等: "development and validation of next generation sequencing based 35-gene hereditary cancer panel", 《HEREDITARY CANCER IN CLINICAL PRACTICE》 *
刘得光3P6N6ZQQ: "未来已来,大有可为-DDR通路基因面面观", 《360DOC.COM个人图书馆》 *
周进学等: "肿瘤基因高通量捕获测序技术检测肝癌细胞株体细胞突变", 《中华实验外科杂志》 *
陈慧娟等: "基于高通量靶向测序的常见实体肿瘤基因突变图谱研究", 《检验医学》 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115449555A (en) * 2022-10-26 2022-12-09 山东大学 Application of ADGRA2 as breast cancer chemotherapy curative effect and prognosis evaluation biomarker
CN115449555B (en) * 2022-10-26 2023-10-13 山东大学 Application of ADGRA2 as biomarker for breast cancer chemotherapy efficacy and prognosis evaluation
CN116287266A (en) * 2023-03-07 2023-06-23 江苏先声医学诊断有限公司 Application of DNA replication advanced region in diagnosis of flood cancer
CN117230203A (en) * 2023-11-14 2023-12-15 首都医科大学附属北京儿童医院 Primer probe set for detecting MYOD1 gene mutation and kit thereof
CN117230203B (en) * 2023-11-14 2024-02-06 首都医科大学附属北京儿童医院 Primer probe set for detecting MYOD1 gene mutation and kit thereof

Similar Documents

Publication Publication Date Title
US11001837B2 (en) Low-frequency mutations enrichment sequencing method for free target DNA in plasma
CN109609647B (en) Detection Panel for pan-cancer-species targeting, chemotherapy and immune drugs based on next-generation sequencing, detection kit and application thereof
CN109880910B (en) Detection site combination, detection method, detection kit and system for tumor mutation load
CN109427412B (en) Sequence combination for detecting tumor mutation load and design method thereof
CN108753967A (en) A kind of gene set and its panel detection design methods for liver cancer detection
JP2020530290A (en) Methods and Substances for Assessing and Treating Cancer
CN114480660A (en) Gene Panel for detecting pan-cancer species, probe and application
CA3083787A1 (en) Process for microsatellite instability detection
US11959142B2 (en) Detection of cancer
CN107475375A (en) A kind of DNA probe storehouse, detection method and kit hybridized for microsatellite locus related to microsatellite instability
CN110592213A (en) Gene panel for prediction of neoantigen load and detection of genomic mutations
CN111575380B (en) Probe library for multigene detection, hybridization kit and multigene detection method
CN111826447B (en) Method for detecting tumor mutation load and prediction model
WO2018133773A1 (en) Method for efficiently detecting ctdna in sample
CN106498082B (en) Method for constructing ovarian cancer susceptibility gene variation library
US20190234955A1 (en) Exosome-guided treatment of cancer
CN114512184A (en) Method for predicting cancer curative effect and prognosis, device and application thereof
CN113736878A (en) Gene panel for detecting nervous system tumor, kit and application thereof
CN114438218B (en) Gene Panel for detecting various tumors, kit and application
CN107858426B (en) Hybridization capture kit for hot spot detection of 16 genes for tumor individualized medication and method thereof
CN114525344A (en) Kit for detecting or assisting in detecting tumor-related gene variation and application thereof
CN113817822B (en) Tumor diagnosis kit based on methylation detection and application thereof
CN114908163A (en) Marker for predicting curative effect of lung cancer immune checkpoint inhibitor and application thereof
JP2023505031A (en) Methods and compositions for cancer analysis
CN115354081B (en) Gene detection combination for pan-solid tumor accurate medication and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
RJ01 Rejection of invention patent application after publication
RJ01 Rejection of invention patent application after publication

Application publication date: 20220513