CN111100909A - Method for calculating genetic heterogeneity in tumor - Google Patents

Method for calculating genetic heterogeneity in tumor Download PDF

Info

Publication number
CN111100909A
CN111100909A CN202010028311.4A CN202010028311A CN111100909A CN 111100909 A CN111100909 A CN 111100909A CN 202010028311 A CN202010028311 A CN 202010028311A CN 111100909 A CN111100909 A CN 111100909A
Authority
CN
China
Prior art keywords
tumor
sample
cancer
vaf
combination index
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
CN202010028311.4A
Other languages
Chinese (zh)
Inventor
鲁靖
黄莉莉
王鲁泉
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.)
Xinhua Bio Pharmaceutical Guangzhou Co Ltd
Original Assignee
Xinhua Bio Pharmaceutical Guangzhou 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 Xinhua Bio Pharmaceutical Guangzhou Co Ltd filed Critical Xinhua Bio Pharmaceutical Guangzhou Co Ltd
Priority to CN202010028311.4A priority Critical patent/CN111100909A/en
Publication of CN111100909A publication Critical patent/CN111100909A/en
Pending legal-status Critical Current

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
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/6858Allele-specific amplification
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/20Allele or variant detection, e.g. single nucleotide polymorphism [SNP] detection

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physics & Mathematics (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Evolutionary Biology (AREA)
  • Medical Informatics (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Theoretical Computer Science (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention discloses a method for calculating genetic heterogeneity in tumors, which belongs to the field of cancers and comprises the following steps: collecting tumor tissue samples to generate mutation group data of each tissue sample; combinatorial indices of genetic heterogeneity within the tumor were calculated from the mutant panel data. The invention calculates the combination index based on the mutation of the sample data of each tissue sample and the variation allele frequency corresponding to the mutation, and provides a quantitative tool for the hierarchical analysis of tumor patients as an index of genetic heterogeneity in tumors.

Description

Method for calculating genetic heterogeneity in tumor
Technical Field
The invention relates to the technical field of cancers, in particular to a method for calculating genetic heterogeneity in tumors.
Background
Cancer is an unsolved public health big problem in the medical field today. Intratumoral Genetic Heterogeneity (IGH) is a major cause of the problematic cancer problem. Immunotherapy (including personalized immunotherapy), such as immune checkpoint inhibitors and personalized tumor vaccines, is becoming a promising solution to meet this medical need. To facilitate effective immunotherapy, knowledge and metering of the IGH system is required.
In the microenvironment of a tumor, the immune system can often recognize and eradicate tumor cells. However, tumor cells can take a number of different strategies to suppress or "hide" the immune system of the human body and thus survive various stages of anti-tumor immune surveillance and response. This function is called immune escape of the tumor cells. Daniel Chen and Ira Mellman proposed the concept of cancer immune cycle, which includes the following seven steps: 1. release of cancer cell antigens (cancer cell death); 2. cancer antigen presentation; 3. priming and activation (APC and T cells); 4. trafficking T cells to tumors (CTLs); infiltration of T cells (CTL and endothelial cells); recognition of cancer cells by T cells (CTL and cancer cells); 7. killing cancer cells (immune cells and cancer cells). The tumor can inhibit the effective recognition and killing of the immune system to tumor cells through the abnormality of different links, thereby generating immune tolerance and even promoting the occurrence and development of the tumor.
Tumor immunotherapy is a therapeutic approach to control and eliminate tumors by restarting and maintaining the cancer immune cycle and restoring normal anti-tumor immune responses in humans, and includes monoclonal antibody-type immune checkpoint inhibitors, therapeutic antibodies, cancer vaccines, cell therapies, and small molecule therapy inhibitors. Many drugs have been designed for specific immunotherapeutic targets, such as checkpoint inhibitors (PD-1/PD-L1, CTLA-4), but not all patients benefit from these universal therapies and inhibit the development of cancer due to complex mutational events and multiple immune escapes. Therefore, there has been increasing interest in selecting and designing immunotherapies for individual differences in specific patients. Such as patients who find an effect on immune checkpoint inhibitors, to allow a more targeted choice of whether to use this class of drugs. Another strategy is to design a personalized vaccine that targets neoantigen-derived mutations specific to a particular patient's cancer cells. This completely personalized vaccine strategy can activate the immune process, killing tumor cells with high specificity, and the non-specific binding induced side effects are also small. Finding suitable targets in many patient-specific somatic mutations has become a major challenge in the design of new antigen-based vaccines. One of the important reasons behind this target discovery challenge is Intratumoral Genetic Heterogeneity (IGH).
Microsatellite instability (MSI) is an important indicator reflecting genomic instability, associated with many genetic diseases and the level of tumor mutational burden/IGH. This unique molecular change and highly variable phenotype is caused by a defective DNA mismatch repair (MMR) system. MSI can be defined as the absence of certain repetitive DNA sequences in the corresponding germline DNA. Samples are generally classified as high microsatellite instability (MSI-H), low microsatellite instability (MSI-L) and microsatellite stability (MSS). Determination of MSI status has prognostic and therapeutic guidance.
One method of measuring the IGH of a particular mutation is Variant Allele Frequency (VAF). To assess the overall statistical distribution of IGH in Tumor samples, Mutant Allele Tumor Heterogeneity (MATH) scores were developed by analyzing VAF for somatic mutations sampled in tumors. There is a meaningful correlation between a higher MATH score and the risk of tumor metastasis. However, the MATH score only reflects the dispersion of the VAF distribution of the tumor sample (the tighter the distribution, the lower the MATH score). The MATH score measures the VAF distribution normalized by the median value of VAF, and therefore does not reflect the absolute level of somatic mutant VAF that is critical for vaccine and immunotherapeutic target selection. The MATH score also did not include a normal control in its mutation calling method, only including mutations with high levels of mRNA expression.
Disclosure of Invention
The invention provides a method for calculating genetic heterogeneity in tumors, and solves the problem that the absolute level of somatic mutation VAF which is crucial to vaccine and immunotherapy target selection is not reflected in the prior art. The technical scheme of the invention is realized as follows:
a method for calculating genetic heterogeneity in tumor comprises the following steps:
s1, collecting a tumor tissue sample;
s2, generating mutation group data of the sample of each tissue sample;
s3, calculating a combined index of genetic heterogeneity within the tumor from the mutant set data.
As a preferred embodiment of the present invention, the combination index specifically refers to:
combination index 1:
Figure BDA0002363283560000031
VAFiis the variant allele frequency of each sample, and n is the sample mutation number of the tissue sample.
As a preferred embodiment of the present invention, the combination index specifically refers to:
combination index 2:
Figure BDA0002363283560000032
VAFiis the variant allele frequency of each sample, and n is the sample mutation number of the tissue sample.
As a preferred embodiment of the present invention, the combination index is used for stratification analysis of cancer patients.
As a preferred embodiment of the present invention, the use of a combination index for the stratification of cancer patients refers in particular to the identification of subpopulations that benefit from cancer treatment drugs.
As a preferred embodiment of the present invention, the cancer treatment drug specifically refers to a cancer immunotherapy drug.
As a preferred embodiment of the present invention, the cancer immunotherapy drug specifically refers to a neoantigen-based cancer therapy drug.
As a preferred embodiment of the present invention, the cancer immunotherapy drug specifically refers to an immune checkpoint inhibitor.
As a preferred embodiment of the invention, the immunodetection point inhibitor specifically refers to a PD-1/PD-L1 inhibitor.
As a preferred embodiment of the present invention, the sub-population benefiting from cancer treatment drugs refers in particular to the MSS patient sub-population benefiting from PD-1/PD-L1 inhibitors.
The invention has the beneficial effects that: and calculating a combination index based on the mutation of the sample data of each tissue sample and a corresponding Variant Allele (VAF), wherein the combination index is used as a quantitative index of genetic heterogeneity in the tumor, and a quantitative tool is provided for the hierarchical analysis of tumor patients.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below, and it is obvious that the drawings in the following description are only some embodiments of the present invention, and for those skilled in the art, other drawings can be obtained according to these drawings without creative efforts.
FIG. 1 is a flow chart of one embodiment of a method of calculating intratumoral genetic heterogeneity of the present invention;
FIG. 2 is one of the VAF profiles of tumor samples;
FIG. 3 is a second VAF profile of the tumor sample.
Detailed Description
The technical solutions in the embodiments of the present invention will be clearly and completely described below with reference to the drawings in the embodiments of the present invention, and it is obvious that the described embodiments are only a part of the embodiments of the present invention, and not all of the embodiments. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
As shown in fig. 1-3, the present invention provides a method for calculating genetic heterogeneity in tumor, which specifically comprises the following steps:
s1, collecting a tumor tissue sample;
the ethical committee of the first hospital in quanzhou city approved the study plan and procedure for patient tumor sample collection (QYL 2019-108). Three tumor tissue samples and one normal/tumor adjacent tissue sample were obtained from each of three patients receiving stage II or stage III colon cancer surgery.
S2, generating mutation group data of the sample of each tissue sample;
total RNA was extracted from tissues using Trizol (Invitrogen, Carlsbad, CA, USA). Approximately 60mg of tissue was ground to powder with liquid nitrogen in a 2mL tube, then homogenized for 2 minutes and placed horizontally for 5 minutes. The mixture was centrifuged at 12,000 Xg for 5 minutes at 4 ℃ and the supernatant was transferred to a fresh EP tube containing 0.3mL of chloroform/isoamyl alcohol (24: 1). The mixture was stirred vigorously for 15s and then centrifuged at 12,000 Xg for 10 min at 4 ℃. After centrifugation, the upper aqueous phase, which retained the RNA, was transferred to a new tube containing an equal volume of isopropanol supernatant and then centrifuged at 13, 600rpm for 20 minutes at 4 ℃. After discarding the supernatant, the RNA precipitate was washed twice with 1mL of 75% ethanol, and then the mixture was centrifuged at 13 ℃ rpm at 4 ℃ for 3 minutes to collect the residual ethanol, and then the precipitate was air-dried for 5 to 10 minutes. A biological safety cabinet. Finally, 25. mu.L to 100. mu.L of LDEPC-treated water was added to dissolve the RNA. Subsequently, total RNA was qualitatively and quantitatively determined using a Nano Drop and Agilent 2100 bioanalyzer (ThermoFisher Scientific, MA, USA).
mRNA was purified using oligodeoxynucleotide (OligodT) attached magnetic beads. At the appropriate temperature, the purified mRNA fragments are fragmented into small fragments using a fragment buffer. Random hexamer initiated reverse transcription was then used to generate first strand cDNA, followed by synthesis of second strand cDNA. Thereafter, end repair was performed by incubation with the addition of A-Tailing Mix and RNA Index Adapters. The cDNA fragment obtained from the previous step was amplified by PCR and the product was purified by Ampure XP Beads, which was then dissolved in EB solution. The product has been quality control verified on an Agilent Technologies 2100 bioanalyzer. The double-stranded PCR product from the previous step was heat denatured and circularized by splint oligonucleotide sequences to obtain the final library. Single-stranded loop dna (sscir dna) was formatted into the final library. The final library was amplified with phi29 to make DNA Nanospheres (DNBs) with 300 copies of one molecule, the DNBs were loaded into patterned nanoarrays, and reads of the terminal 100 bases were generated on the BGIseq500 platform (BGI-shenzhen, china). Samples from patients with stage II colon cancer generated 20GB of data per sample and were sampled as 10GB subsamples. The other samples per patient yielded only 10GB of data.
RNA-seq sequencing data were analyzed using the GATK procedure (v4.0.12.0). Somatic mutations were calculated from GATK4Mutect2, strelska (v2.9.2), SomaticSniper (v1.0.5.0) and Varscan2(v2.4.3), respectively, and all mutations calculated by the software were integrated using Somaticseq (v3.1.1) with reference to normal tissue samples. Somatseq is a machine learning-based somatic mutation integration software that integrates multiple VCF files of a tumor sample into one VCF file. SNVs and indels of quality "PASS" (PASS) were matched to the cosmc database. Variant Allele Frequency (VAF) was calculated as the ratio of alternative allele observations to read depth for each position. Statistical analysis was performed using Python. The MATH score is implemented by python and the MSI score is determined by MSISensor (v 0.6). Samples with scores > 3.5% are considered MSIs, otherwise MSSs. The MSI score was determined using the MSIsensor program (table 1). Samples with scores > 3.5% are considered MSIs, otherwise MSSs. In these three samples, patient 801 displayed the MSI, while patients 716 and 725 were considered MSS.
S3, calculating a combined index of genetic heterogeneity within the tumor from the mutant set data.
The calculation of the combined index of genetic heterogeneity within the tumor refers in particular to:
combination index 1:
Figure BDA0002363283560000051
or
Combination index 2:
Figure BDA0002363283560000061
VAF is variant allele frequency (Varianaleleffequency), and n is a mutation number. The part of the tumor not covered by the mutation is indicated as "1-VAF". Log values are used to reflect the geometric relationships between individual mutations (rather than additive relationships)
Figure BDA0002363283560000062
Cancer patient stratification example COMIC is the world's largest, most comprehensive resource for somatic mutations in human cancers. Using COSMIC as a reference database, mutations that overlap with the COSMIC database can reasonably be expected to be rich in high quality mutations, and functional and driver mutations are relatively rich. Similar to literature reports, only about 1-2% of the identified mutations in this study were observed to be present in the COSMIC database. However, this overlapping set should have relatively abundant mutations that can drive the development and proliferation of cancer. To provide an overall summary of the state of IGH, the average vaf (mean vaf) of all mutations in the sample and the average mutation vaf (mean COSMIC vaf) in the COSMIC database were measured.
In the sample from patient 716 (stage II), variants were enriched (ABC) over a VAF interval of 30% to 58% and the mean VAF (total or cosinc) was about 40%. The VAF distribution pattern and the mean VAF remained unchanged between the three different samples from patient 716. The variation distribution pattern was also very similar between samples with 20G and 10G sequencing depth (ABC vs DEF and table 2), demonstrating that the robustness of the method is independent of the sequencing depth. In the sample from patient 725 (stage III), variants were enriched (GHI) over the VAF interval of 2% to 26% and the mean VAF (total or cosinc) was about 20% (table 2). The VAF distribution pattern is also well conserved among the three different samples from patient 716 (G, H and I). In the sample from patient 801 (stage III), the variation was relatively smoothly distributed between 2% and 58%, and the mean VAF (total or cosinc) was about 30% for two out of three samples. One sample (801_ B) from patient 801 showed a very different distribution pattern (L, table 2), which may be due to heterogeneity between the three samples or contamination of normal tissue samples. Overall, significant differences were observed in the three VAF distribution patterns and mean values of the three patients (40%, 20%, 30%). The difference in VAF distribution between the two MSS patients (716, 725) was also evident (mean VAF 20% and 40%, respectively).
For all these patient samples, the MATH score, which is an indicator of tumor heterogeneity, was very high (>16 was considered to be highly heterogeneous) (table 1), indicating that there are limitations in using the MATH score to measure IGH gradients in tumor mutation landscape analysis, failing to reveal inter-tumor patient sample variability.
In addition to the VAF distribution pattern, the absolute number of mutations in the tumor sample is also important for immunotherapy design. The number of mutations (total or COSMIC) varied very among patients (Table 2), with all mutations ranging from hundreds to thousands (571-. The numerical value of the combinatorial index is used to reflect the geometric relationship (rather than the additive relationship) between individual mutations. The higher the value of the combination index, the better the coverage of the mutation. In this analysis, the combination index 1 ranged from 0.41 to 24.57 and the combination index 2 ranged from 0.42 to 27.86.
The complexity of personalizing immunotherapy for CRC patients with different IGH backgrounds and the personalized nature of the mutation profile are revealed. Considering the key impact of target coverage and target number on the final immunotherapy outcome, combinatorial indices are designed as tools to guide patient stratification and neoantigen analysis in personalized immunotherapy. For patients with a higher combination index, this may translate into better immunotherapy efficacy, and vice versa.
In various types of cancer, including CRC, the status of MSI-H is associated with a high response rate to treatment with PD-1/PD-L1 checkpoint inhibitors. One of the basic conditions for effective treatment of PD-1/PD-L1 inhibitors is the availability of neoantigens and their tumor coverage, which are systematically reflected in the combinatorial index. Patients similar to 716 were MSS, but had a large number of mutations compared to other MSS patients (e.g., patient 725), and the Vibrant Index values differed by two orders of magnitude. The combined index can be used to identify a subpopulation of MSS patients likely to benefit from treatment with a PD-1/PD-L1 inhibitor.
TABLE 1 patient sample information
Figure BDA0002363283560000071
Figure BDA0002363283560000081
QC30 is the RNA-seq sequencing quality score (above 0.9 is considered high quality). The MSI (microsatellite instability) score was determined by MSISensor and more than 3.5% was considered MSS. The MATH score measures heterogeneity of mutant allelic tumors, with greater than 16 being considered highly heterogeneous.
Table 2.vafs. distribution
Figure BDA0002363283560000082
The mean VAF is the mean VAF of all mutations, whereas STDEVVAF is the standard deviation of all mutated VAFs. The mean VAF (COSMIC) is the mean VAF for mutations in the COSMIC database, and stdevvaf (COSMIC) is the standard deviation of the VAFs for mutations in the COSMIC database. Mutations are the total number of mutations identified for a sample. Mutations (COSMICs) are the total number of mutations in the COSMIC in a sample.
For the bar graphs in fig. 2 and 3, the x-axis represents VAF intervals and the y-axis represents the proportion (%) of mutations in each VAF interval. For the line graph, the y-axis represents the proportion (%) of mutations above the VAF threshold.
The present invention calculates a combination index as an index for genetic heterogeneity in a tumor based on a mutation in sample data of each tissue sample and a VAF distribution pattern corresponding to the mutation. This combined index can be used to identify subpopulations that may benefit from cancer treatment drugs, particularly in cancer immunotherapy drugs, such as neoantigen-based immunotherapy or immune checkpoint inhibitors. The present combination parameters can be used to find a subset of MSS patients who may benefit from treatment with a PD-1/PD-L1 inhibitor.
The above description is only for the preferred embodiment of the present invention, and is not intended to limit the present invention, and any modifications, equivalents, improvements, etc. made by selecting mutation reference database other than COSMIC may be included in the scope of the present invention, as mutation group data may be obtained by various means, and mutation discovery tools may be widely selected within the spirit and principle of the present invention.

Claims (10)

1. A method for calculating genetic heterogeneity in a tumor, comprising the steps of:
s1, collecting a tumor tissue sample;
s2, generating mutation group data of the sample of each tissue sample;
s3, calculating a combined index of genetic heterogeneity within the tumor from the mutant set data.
2. The method of claim 1, wherein the combination index is a combination index of 1:
Figure FDA0002363283550000011
VAFiis the variant allele frequency of each sample, and n is the sample mutation number of the tissue sample.
3. The method of claim 1, wherein the combination index specifically refers to:
combination index 2:
Figure FDA0002363283550000012
VAFiis the variant allele frequency of each sample, and n is the sample mutation number of the tissue sample.
4. The method of claim 2 or 3, wherein the combination index is used for stratification of cancer patients.
5. The method of claim 4, wherein the combination index is used for the stratification of cancer patients, particularly for identifying subpopulations that benefit from cancer treatment drugs.
6. The method of claim 5, wherein the cancer treatment drug is cancer immunotherapy drug.
7. The method of claim 6, wherein the cancer immunotherapy drug is a neoantigen-based cancer therapy drug.
8. The method of claim 6, wherein the cancer immunotherapy drug is an immune checkpoint inhibitor.
9. The method of claim 8, wherein the immunodetection point inhibitor is PD-1/PD-L1 inhibitor.
10. The method of claim 9, wherein the subpopulation that benefits from cancer treatment drugs is specifically the MSS patient subpopulation that benefits from treatment with PD-1/PD-L1 inhibitor.
CN202010028311.4A 2020-01-10 2020-01-10 Method for calculating genetic heterogeneity in tumor Pending CN111100909A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010028311.4A CN111100909A (en) 2020-01-10 2020-01-10 Method for calculating genetic heterogeneity in tumor

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010028311.4A CN111100909A (en) 2020-01-10 2020-01-10 Method for calculating genetic heterogeneity in tumor

Publications (1)

Publication Number Publication Date
CN111100909A true CN111100909A (en) 2020-05-05

Family

ID=70426221

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010028311.4A Pending CN111100909A (en) 2020-01-10 2020-01-10 Method for calculating genetic heterogeneity in tumor

Country Status (1)

Country Link
CN (1) CN111100909A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023284260A1 (en) * 2021-07-12 2023-01-19 广州燃石医学检验所有限公司 Method for evaluating intra-tumor heterogeneity on basis of blood sequencing, and application thereof to prediction of response to immunotherapy

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109196359A (en) * 2016-02-29 2019-01-11 基础医疗股份有限公司 For assessing the method and system of Tumor mutations load
TW201921276A (en) * 2017-09-13 2019-06-01 日商三菱太空軟體股份有限公司 Copy number measurement device, copy number measurement program,copy number measurement method and gene panel
US20190172582A1 (en) * 2017-12-01 2019-06-06 Illumina, Inc. Methods and systems for determining somatic mutation clonality
WO2019155050A1 (en) * 2018-02-12 2019-08-15 F. Hoffmann-La Roche Ag Method of predicting response to therapy by assessing tumor genetic heterogeneity
CN110603593A (en) * 2017-02-09 2019-12-20 社会福祉法人三星生命公益财团 Target gene identification method for tumor treatment

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109196359A (en) * 2016-02-29 2019-01-11 基础医疗股份有限公司 For assessing the method and system of Tumor mutations load
CN110603593A (en) * 2017-02-09 2019-12-20 社会福祉法人三星生命公益财团 Target gene identification method for tumor treatment
TW201921276A (en) * 2017-09-13 2019-06-01 日商三菱太空軟體股份有限公司 Copy number measurement device, copy number measurement program,copy number measurement method and gene panel
US20190172582A1 (en) * 2017-12-01 2019-06-06 Illumina, Inc. Methods and systems for determining somatic mutation clonality
WO2019155050A1 (en) * 2018-02-12 2019-08-15 F. Hoffmann-La Roche Ag Method of predicting response to therapy by assessing tumor genetic heterogeneity

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
DAVID E LARSON等: "SomaticSniper: identification of somatic point mutations in whole genome sequencing data", 《BIOINFORMATICS》 *
JAVAD NOORBAKHSH等: "Distribution-based measures of tumor heterogeneity are sensitive to mutation calling and lack strong clinical predictive power", 《SCI REP》 *
LAYLA OESPER等: "THetA: inferring intra-tumor heterogeneity from high-throughput DNA sequencing data", 《GENOME BIOL》 *
LUC G T MORRIS等: "Pan-cancer analysis of intratumor heterogeneity as a prognostic determinant of survival", 《ONCOTARGET》 *
NIU BF等: "MSIsensor: microsatellite instability detection using paired tumor-normal sequence data", 《BIOINFORMATICS》 *
OH BY等: "Intratumor heterogeneity inferred from targeted deep sequencing as a prognostic indicator", 《SCIENTIFIC REPORTS》 *
TOMOKO SAITO等: "A temporal shift of the evolutionary principle shaping intratumor heterogeneity in colorectal cancer", 《NAT COMMUN》 *
YU FAN等: "MuSE: accounting for tumor heterogeneity using a sample-specific error model improves sensitivity and specificity in mutation calling from sequencing data", 《GENOME BIOL》 *
李小波: "数学建模及数据挖掘方法在结直肠癌转移机制研究中的应用", 《中国博士学位论文全文数据库 医药卫生科技辑》 *
樊代明: "《肿瘤研究前沿》", 30 November 2017, 西安:西安交通大学出版社 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023284260A1 (en) * 2021-07-12 2023-01-19 广州燃石医学检验所有限公司 Method for evaluating intra-tumor heterogeneity on basis of blood sequencing, and application thereof to prediction of response to immunotherapy

Similar Documents

Publication Publication Date Title
Litchfield et al. Meta-analysis of tumor-and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition
US20220213562A1 (en) Detection and treatment of disease exhibiting disease cell heterogeneity and systems and methods for communicating test results
JP7487163B2 (en) Detection and diagnosis of cancer evolution
Wang et al. Insights into beta cell regeneration for diabetes via integration of molecular landscapes in human insulinomas
CA2983833C (en) Diagnostic methods
US11193175B2 (en) Normalizing tumor mutation burden
US20190065670A1 (en) Predicting disease burden from genome variants
Parry et al. Evolutionary history of transformation from chronic lymphocytic leukemia to Richter syndrome
US20220119881A1 (en) Systems and methods for sample preparation, sample sequencing, and sequencing data bias correction and quality control
KR20210100650A (en) Ancestor-specific genetic risk score
JP2024028940A (en) Methods and systems for analyzing nucleic acid molecules
Alvarez-Breckenridge et al. Microenvironmental landscape of human melanoma brain metastases in response to immune checkpoint inhibition
CN114512184A (en) Method for predicting cancer curative effect and prognosis, device and application thereof
CN111100909A (en) Method for calculating genetic heterogeneity in tumor
Hersh et al. High-throughput sequencing in respiratory, critical care, and sleep medicine research. An Official American Thoracic Society Workshop Report
Li et al. Identification of inflammation-related genes and exploration of regulatory mechanisms in patients with osteonecrosis of the femoral head
CN115873936A (en) Biomarker CXCL8 and application thereof
US20220301654A1 (en) Systems and methods for predicting and monitoring treatment response from cell-free nucleic acids
Kamboj et al. Ub-ISAP: a streamlined UNIX pipeline for mining unique viral vector integration sites from next generation sequencing data
CN109402131B (en) Nucleic acid for encoding KANSL1 gene mutant and application thereof
CN113160895A (en) Colorectal cancer risk assessment model and system
CN111919257B (en) Method and system for reducing noise in sequencing data, and implementation and application thereof
Wang et al. Construction of ceRNA networks with different types of IDH1 mutation status in low-grade glioma patients
Brewer et al. 58. Improving the molecular diagnosis of SBDS-related disorders by identifying rare gene conversion events.
Zhang et al. Blood-based Genomic and Cellular Determinants of Response to Neoadjuvant PD-1 Blockade in Patients with Non-Small-Cell Lung Cancer

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: 20200505