CN109448790B - Storage medium, equipment and device for determining tumor immunotherapy marker - Google Patents

Storage medium, equipment and device for determining tumor immunotherapy marker Download PDF

Info

Publication number
CN109448790B
CN109448790B CN201811376543.8A CN201811376543A CN109448790B CN 109448790 B CN109448790 B CN 109448790B CN 201811376543 A CN201811376543 A CN 201811376543A CN 109448790 B CN109448790 B CN 109448790B
Authority
CN
China
Prior art keywords
aps
tumor
tigs
cancer
individual
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.)
Active
Application number
CN201811376543.8A
Other languages
Chinese (zh)
Other versions
CN109448790A (en
Inventor
刘雪松
王诗翔
何早柯
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.)
ShanghaiTech University
Original Assignee
ShanghaiTech University
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 ShanghaiTech University filed Critical ShanghaiTech University
Priority to CN201811376543.8A priority Critical patent/CN109448790B/en
Publication of CN109448790A publication Critical patent/CN109448790A/en
Application granted granted Critical
Publication of CN109448790B publication Critical patent/CN109448790B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

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

Landscapes

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

Abstract

The invention relates to the field of biology, in particular to a marker for tumor immunotherapy. The present invention provides a computer readable storage medium storing a computer program which when executed performs method steps comprising: obtaining mRNA expression data of an individual; obtaining an antigen presentation index (APS) based on a GSVA algorithm according to mRNA expression data of an individual; calculating to obtain APS according to formula Inormalized(ii) a Tumor immunogenicity index (TIGS) was calculated according to formula II. The TIGS provided by the invention can predict the objective response rate of pan-cancer Immune Checkpoint Inhibitor (ICI) and the clinical effect of cancer patients after ICI treatment.

Description

Storage medium, equipment and device for determining tumor immunotherapy marker
Technical Field
The invention relates to the field of biology, in particular to a marker for tumor immunotherapy.
Background
The development of immunotherapy, represented by Immune Checkpoint Inhibitors (ICI), including anti-PD-1 antibodies, anti-PD-L1 antibodies, anti-CTLA-4 antibodies or combinations thereof, is a revolutionary breakthrough in the area of cancer therapy. Conventional cancer treatment methods (such as radiotherapy and chemotherapy) are often ineligible for the advanced metastatic cancers, and immunotherapy can exert very significant treatment effects on partial advanced metastatic cancers. However, most unselected patients do not respond to ICI. The response rate to PD- (L)1 inhibition was less than 40% for most tumor types, and the response rate reported for each tumor type was highly correlated with Tumor Mutational Burden (TMB) for each tumor type. Various factors have been reported to affect ICI effectiveness, including: PD-L1 expression, tumor mutation burden, DNA mismatch repair deficiency, degree of cytotoxic T cell infiltration, mutation profile, antigen presentation deficiency, interferon signaling, tumor aneuploidy, and T cell gene expression profile. However, none of these factors is sufficient to achieve an accurate prediction of the effectiveness of immunotherapy.
Disclosure of Invention
In view of the above-mentioned drawbacks of the prior art, it is an object of the present invention to provide a marker for tumor immunotherapy, which solves the problems of the prior art.
To achieve the above and other related objects, an aspect of the present invention provides a computer-readable storage medium storing a computer program which, when executed, can implement method steps, the method steps comprising:
acquiring APS based on GSVA algorithm according to individual mRNA expression data;
calculating to obtain APS according to formula Inormalized
Figure BDA0001870909870000011
Calculating to obtain TIGS according to formula II
TIGS=APSnormalizedXln (TMB +1) (formula II).
In some embodiments of the invention, the individual is selected from an animal receiving tumor immunotherapy.
In some embodiments of the invention, the subject is selected from a human.
In some embodiments of the invention, the tumor immunotherapy comprises immune checkpoint inhibitor therapy.
In some embodiments of the invention, the tumor immunotherapy comprises a method of treatment of an individual by administering one or more of an anti-PD-1 antibody, a PD-L1 antibody, a CTLA-4 antibody.
In some embodiments of the invention, the mRNA expression data and/or TMB of the individual is derived from a tumor sample.
In some embodiments of the invention, the tumor is selected from melanoma, lung cancer, urothelial cancer, head and neck cancer, kidney cancer, lymphoma, prostate cancer, breast cancer, glioma, cervical cancer, endometrial cancer, esophageal cancer, liver cancer, mesothelioma, ovarian cancer, pancreatic cancer, paraganglioma, colon cancer, sarcoma, gastric cancer, testicular germ cell tumor, thyroid cancer, or thymoma.
In some embodiments of the invention, the genes used to calculate APS include at least 14 genes of PSMB5, PSMB6, PSMB7, PSMB8, PSMB9, PSMB10, TAP1, TAP2, ERAP1, ERAP2, CANX, CALR, PDIA3, TAPBP, B2M, HLA-A, HLA-B, and HLA-C.
In some embodiments of the invention, APSpancan_min=-1~-0.86。
In some embodiments of the invention, APSpancan_max=0.88~1。
In another aspect, the invention provides an apparatus comprising: a processor and a memory;
the memory for storing a computer program, the processor for executing the computer program stored by the memory to cause the terminal to perform method steps comprising:
obtaining mRNA expression data of an individual;
acquiring APS based on GSVA algorithm according to individual mRNA expression data;
calculating to obtain APS according to formula Inormalized
Figure BDA0001870909870000021
Calculating to obtain TIGS according to formula II
TIGS=APSnormalizedXln (TMB +1) (formula II).
Another aspect of the present invention provides an apparatus, which may include:
the mRNA expression data acquisition module is used for acquiring mRNA expression data of individuals;
the APS processing module is used for acquiring APS based on a GSVA algorithm according to individual mRNA expression data;
APSnormalizeda processing module for calculating APS according to formula Inormalized
Figure BDA0001870909870000022
And the TIGS processing module is used for calculating and obtaining TIGS according to the formula II.
TIGS=APSnormalizedXln (TMB +1) (formula II).
Drawings
Figure 1 shows tumor immunogenicity index (TIGS) analysis for 32 cancer types.
FIG. 2 shows the Objective Response Rate (ORR) of TIGS in predicting PD-1 inhibition for different types of tumors.
FIG. 3 shows a comparison of the performance of TIGS and TMB in predicting the clinical effect of ICI in tumor patients.
Detailed Description
The inventor finds that the basic determinants of the tumor immunogenicity comprise tumor antigenicity and antigen processing and presenting efficiency, the antigen processing and presenting efficiency is calculated by applying a GSVA method, and the calculation result is expressed as: the present invention has been completed based on the fact that an antigen presentation index (APM Score, APS) and then a tumor immunogenicity index (TIGS) were calculated by combining APM Score and TMB, TIGS predicts the objective response rate of pan-cancer ICI and ICI clinical response is consistently superior to TMB, indicating that TIGS is a novel and effective tumor intrinsic biomarker for predicting ICI response.
In a first aspect, the present invention provides a diagnostic method comprising:
obtaining mRNA expression data of an individual;
acquiring APS (Antigen presentation score) based on GSVA algorithm according to mRNA expression data of an individual;
calculating to obtain APS according to formula Inormalized(standardized APS)
Figure BDA0001870909870000031
Calculating according to formula II to obtain TIGS (tumor immunogenicity index)
TIGS=APSnormalizedXln (TMB +1) (formula II).
In the diagnostic method provided by the present invention, the subject is usually an animal (including human) that can be treated with tumor immunotherapy, and the subject usually includes human, non-human primate, and can be mammal, dog, cat, horse, sheep, pig, cow, etc. The mRNA expression data can generally be genome-wide mRNA expression data, and is typically derived from a tumor sample of the individual, and methods for obtaining mRNA expression data for the individual will be known to those skilled in the art, e.g., the sample can be examined by means including, but not limited to, gene chips, RNA-seq, etc., to obtain genome-wide mRNA expression data for the individual.
In the diagnostic method provided by the present invention, the method of acquiring the mRNA expression data of the individual may be inputting the mRNA expression data of the individual, or the like. Genes used for calculating APS comprise at least 14 or more genes of PSMB5, PSMB6, PSMB7, PSMB8, PSMB9, PSMB10, TAP1, TAP2, ERAP1, ERAP2, CANX, CALR, PDIA3, TAPBP, B2M, HLA-A, HLA-B and HLA-C, the number of the included genes can be 14, 15, 16, 17 or 18, and when APS is actually calculated, expression data of no more than 4 genes are randomly deleted from 18 genes, so that the calculation result is not greatly influenced. GSVA algorithms should be known to the skilled person, for example, reference may be made to the literature (Hanzelmann S, Castelo R, Guinney J.GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 2013; 14: 7).
In the diagnostic method provided by the present invention, APSpancan_minGenerally refers to the lowest value of APS in a population of tumor patients corresponding to an individual. In one embodiment of the present invention, APSpancan_minStatistics of the minimum of the original APS of the 9613 TCGA 32 tumor samples from the public database of tumor genomes, the APS minimum is usually-0.86, APSpancan_minThe range may be-1 to-0.86. In one embodiment of the present invention, APSpancan_minThe value is typically-0.86, and in rare cases, if the original APS value of a sample is less than-0.86, the value can be taken to replace APSpancan_minFinal APS of the samplenormalized=0。
In the diagnostic method provided by the present invention, APSpancan_maxGenerally refers to the maximum value of APS in a population of tumor patients corresponding to an individual. In one embodiment of the present invention, APSpancan_maxStatistics of the maximum value of the original APS of 9613 TCGA 32 tumors from the public database of tumor genomes, the APS maximum value is usually 0.88, APSpancan_maxThe value range may be 0.88-1. In one embodiment of the present invention, APSpancan_maxThe value is generally 0.88, and in a rare case, if the original APS value of a sample is greater than 0.88, the value can be taken to replace APSpancan_maxFinal APS of the samplenormalized=1。
In the diagnostic method provided by the present invention, the total number of somatic gene coding errors, base substitutions, gene insertions or deletion errors detected in TMB (Tumor mutation load) usually per million bases, i.e. the number of nonsynonymous mutations per MB (10^6 base pairs), the corresponding test sample usually comes from the Tumor sample of the individual, and the method for obtaining the TMB of the individual should be known to those skilled in the art, for example, the sample can be detected by means including but not limited to WES, targeted sequencing, etc. to obtain the TMB of the individual.
In the diagnosis method provided by the present invention, the prognosis of the tumor immunotherapy effect of an individual can be determined by the TIGS obtained by calculation. For example, individuals with higher TIGS scores typically have a better prognosis (e.g., survival time, etc.) for tumor immunotherapy, and individuals with lower TIGS scores typically have a poorer prognosis for tumor immunotherapy. As another example, individuals with higher TIGS scores are generally considered to have a higher probability of responding well to, or to be better suited for, tumor immunotherapy (e.g., the rate of objective response of a tumor to ICI, ORR, etc.), and individuals with lower TIGS scores are generally considered to have a lower probability of responding well to, or to be less suited for, tumor immunotherapy.
In the diagnosis method provided by the present invention, whether an individual is susceptible to a tumor suitable for tumor immunotherapy can be determined by the TIGS obtained by calculation. For example, individuals with higher TIGS scores typically have a better prognosis (e.g., survival time, etc.) for tumor immunotherapy when developing a tumor, and individuals with lower TIGS scores typically have a poorer prognosis for tumor immunotherapy when developing a tumor. For another example, individuals with higher TIGS scores are generally considered to have a higher probability of responding well to, or to be better suited for, tumor immunotherapy (e.g., the objective response rate ORR of tumors to ICI, etc.) when developing tumors, and individuals with lower TIGS scores are generally considered to have a lower probability of responding well, or to be less suitable for, tumor immunotherapy when developing tumors.
In the diagnostic method provided by the present invention, whether or not a tumor suffered by an individual is suitable for tumor immunotherapy can be diagnosed by using the TIGS obtained by calculation. For example, individuals with tumors with higher TIGS scores are generally considered to have a better prognosis (e.g., survival time, etc.) for tumor immunotherapy, and individuals with tumors with lower TIGS scores are generally considered to have a poorer prognosis for tumor immunotherapy. For another example, individuals with tumors with higher TIGS scores are generally considered to have a higher probability of responding well to tumor immunotherapy or to be more suitable for tumor immunotherapy (e.g., the objective response rate ORR of tumors to ICI, etc.), and individuals with tumors with lower TIGS scores are generally considered to have a lower probability of responding well to tumor immunotherapy or to be less suitable for tumor immunotherapy.
In the diagnosis method provided by the present invention, the tumor to which the diagnosis method is applied may include, but is not limited to, melanoma, lung cancer, urothelial cancer, head and neck cancer, kidney cancer, lymphoma, prostate cancer, breast cancer, glioma, cervical cancer, endometrial cancer, esophageal cancer, liver cancer, mesothelioma, ovarian cancer, pancreatic cancer, paraganglioma, colon cancer, sarcoma, gastric cancer, testicular germ cell tumor, thyroid cancer, thymoma, etc.
In the diagnostic method provided by the present invention, the tumor immunotherapy effect can be any immunotherapy method suitable for tumor patients in the field, and specifically can be a therapy method including but not limited to immune checkpoint inhibitor therapy, and the like, and more specifically can be a therapy method including but not limited to administering one or more of anti-PD-1 antibody, PD-L1 antibody, CTLA-4 antibody, and the like to an individual, and these therapy methods can be administered alone or in combination with other therapies.
A second aspect of the invention provides a computer-readable storage medium storing a computer program which, when executed, may implement method steps including those provided by the diagnostic method as described above.
The third aspect of the present invention provides an apparatus which can be used for screening whether an individual is susceptible to develop a tumor suitable for tumor immunotherapy, diagnosing whether an individual has a tumor suitable for tumor immunotherapy, making a prognosis on the effect of tumor immunotherapy on the individual, and the like, comprising: a processor and a memory;
the memory is used for storing computer programs, and the processor is used for executing the computer programs stored by the memory so as to enable the terminal to execute the diagnosis method.
In a fourth aspect, the present invention provides a device, which can be used for screening whether an individual is susceptible to a tumor suitable for tumor immunotherapy, diagnosing whether an individual has a tumor suitable for tumor immunotherapy, performing prognosis on the effect of tumor immunotherapy on the individual, and the like, and the device can include:
an mRNA expression data acquisition module for acquiring mRNA expression data of an individual, for example, mRNA expression data of an input individual, or the like;
the APS processing module is used for acquiring APS based on a GSVA algorithm according to individual mRNA expression data;
APSnormalizeda processing module for calculating APS according to formula Inormalized
And the TIGS processing module is used for calculating and obtaining TIGS according to the formula II.
In the present invention, the operation principle of each module in the above apparatus may refer to the diagnosis method described above, and is not described herein again.
ICI helps the patient's immune system to recognize and attack cancer cells, whose immunogenicity is the fundamental determinant of the ICI response. In theory, tumors with very low or no immunogenicity do not respond to therapeutic strategies that enhance the immune response, whereas ICI is generally applicable to tumors with sufficient immunogenicity. In addition, enhancing tumor immunogenicity may potentially convert immunotherapy non-responsive tumors to immunotherapy responsive tumors. Therefore, the actual immunogenicity of a tumor is not easily measurable, theoretically the tumor immunogenicity is determined by the tumor cells themselves, and is also influenced by the tumor microenvironment etc., e.g. the function of professional antigen presenting cells such as dendritic cells. The inventor provides a novel molecular marker for predicting whether the immunotherapy is effective, namely tumor immunogenicity index (TIGS). TIGS can efficiently distinguish immunotherapy non-responsive patients from immunotherapy responsive patients, comparing the predictive power of TIGS with TMB in the prediction of Objective Response Rate (ORR) for pan-cancerous ICI and prediction of clinical response to ICI in individual patients. The result shows that TIGS predicts the objective response rate of pan-cancer ICI, the ICI clinical response is always superior to TMB, the TIGS is a novel and effective tumor inherent biomarker for predicting the ICI response, and the prediction effect of the marker is better than that of the marker 'tumor mutation load' in the current clinical application, so that the marker has important clinical application value of immunotherapy.
The embodiments of the present invention are described below with reference to specific embodiments, and other advantages and effects of the present invention will be easily understood by those skilled in the art from the disclosure of the present specification. The invention is capable of other and different embodiments and of being practiced or of being carried out in various ways, and its several details are capable of modification in various respects, all without departing from the spirit and scope of the present invention.
Before the present embodiments are further described, it is to be understood that the scope of the invention is not limited to the particular embodiments described below; it is also to be understood that the terminology used in the examples is for the purpose of describing particular embodiments, and is not intended to limit the scope of the present invention; in the description and claims of the present application, the singular forms "a", "an" and "the" include plural referents unless the context clearly dictates otherwise.
When numerical ranges are given in the examples, it is understood that both endpoints of each of the numerical ranges and any value therebetween can be selected unless the invention otherwise indicated. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In addition to the specific methods, devices, and materials used in the examples, any methods, devices, and materials similar or equivalent to those described in the examples may be used in the practice of the invention in addition to the specific methods, devices, and materials used in the examples, in keeping with the knowledge of one skilled in the art and with the description of the invention.
Unless otherwise indicated, the experimental methods, detection methods, and preparation methods disclosed herein all employ techniques conventional in the art of molecular biology, biochemistry, chromatin structure and analysis, analytical chemistry, cell culture, recombinant DNA technology, and related arts. These techniques are well described in the literature, and may be found in particular in the study of the MOLECULAR CLONING, Sambrook et al: a LABORATORY MANUAL, Second edition, Cold Spring Harbor LABORATORY Press, 1989and Third edition, 2001; ausubel et al, Current PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, 1987and periodic updates; the series METHODS IN ENZYMOLOGY, Academic Press, San Diego; wolffe, CHROMATIN STRUCTURE AND FUNCTION, Third edition, Academic Press, San Diego, 1998; (iii) METHODS IN ENZYMOLOGY, Vol.304, Chromatin (P.M.Wassarman and A.P.Wolffe, eds.), Academic Press, San Diego, 1999; and METHODS IN MOLECULAR BIOLOGY, Vol.119, chromatography Protocols (P.B.Becker, ed.) Humana Press, Totowa, 1999, etc.
Example 1
Based on the GSVA algorithm, the Antigen Presentation Score (APS) of the tumor sample was calculated. And acquiring the gene mRNA expression data of the tumor samples by using a gene chip or RNA-seq mode, and calculating the APS of each sample by using a GSVA algorithm on the basis. Genes used for calculating APS include (PSMB5, PSMB6, PSMB7, PSMB8, PSMB9, PSMB10, TAP1, TAP2, ERAP1, ERAP2, CANX, CALR, PDIA3, TAPBP, B2M, HLA-A, HLA-B, HLA-C). The values of the original APS range from-1 to 1. To multiply with tumor antigenicity, a standardized APS was used, ranging from 0 to 1, as an indication of "antigen processing and presentation capacity".
Figure BDA0001870909870000071
Wherein, APSpancan_minAnd APSpancan_maxThe values of (a) are-0.86 and 0.88, respectively.
Tumor immunogenicity index (TIGS) was calculated using the following formula:
TIGS=APSnormalized×In(TMB+1)。
the distribution of TIGS in different types of tumors is shown in fig. 1, where TIGS analysis of a, 32 cancer types. B, results of Cox proportional hazards regression analysis using TIGS on all solid cancers. Forest plots show loge risk ratios (95% confidence intervals). Cancer types with high TIGS include: cutaneous Melanoma (SKCM), diffuse large B-cell lymphoma (DLBC), colon adenocarcinoma (COAD), head and neck squamous cell carcinoma (HNSC). Univariate Cox regression analysis showed that TIGS scores correlated with survival rates of cancer patients, some were significant
Example 2
The predictive effect of TIGS on Objective Response Rate (ORR) of different tumor types against immune checkpoint inhibitors was further examined. Data were determined from literature searches for ORR of 26 tumor types or subtypes corresponding to immune checkpoint inhibitors. For each tumor type, response data from the largest published study that evaluated ORR was summarized (see table 1, table 1 showing median TMB, APS and clinical Objective Response Rate (ORR) to ICI for 26 tumors). Only anti-PD-1 or anti-PD-L1 monotherapy studies were included in which at least 10 patients not selected for PD-L1 tumor expression were enrolled. The median Tumor Mutational Burden (TMB) for each tumor type is derived from that provided by the literature (Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. analysis of 100,000human cancer genes novales the landscapes of tumor tissue, genome Med 2017; 9: 34). Based on the public database TCGA for tumor genome and the public database GEO for gene expression, we calculated APS information for 26 tumor types, from which TIGS values for each tumor type were calculated, with the calculation method referred to example 1.
Objective Response Rate (ORR) for TIGS in predicting different types of tumors to PD-1 inhibition is shown in figure 2, where the correlation between aps (a), tmb (b), TIGS (c) and objective response rate to anti-PD-1 or anti-PD-L1 treatment is shown for 26 cancer types. Median normalized APS, log-scale median TMB median (non-synonymous mutations/MB) and TIGS among 26 tumor types or subtypes receiving PD-1 or PD-L1 inhibitors are shown. The number of patients evaluating the objective response rate was shown for each tumor type (size of circle), and the number of tumor samples analyzed to calculate the tumor mutation load (degree of shading of circle). Significant correlations were observed between APS, TMB, TIGS and ORR, with correlation coefficients of 0.529 and 0.667 between APS and ORR, TMB and ORR, respectively, indicating that 28% and 44% of the ORR differences for the cancer types were provided by the variation in APS and TMB, respectively. The correlation coefficient between TIGS and ORR is 0.814, suggesting that 66% of ORR difference can be provided by TIGS. This pan-cancerous ORR analysis indicates that TIGS performs significantly better than the known molecular marker TMB in predicting the response rate of immunotherapy.
TABLE 1
Cancer_Type Pooled_ORR Median_TMB Median_APS
Adrenocortical Carcinoma 6 2.7 0.4436434
Breast Carcinoma 6.18 3.6 0.3211102
Cervical Cancer 18.28 4.7 0.7340055
Cutaneous Squamous Cell Carcinoma 47.3 45.2 0.5061751
Endometrial Cancer 13 3.9 0.6345594
Esophagogastric Carcinoma 10.69 5 0.3533898
Germ Cell Tumors 0 2.7 0.2472031
Glioblastoma 9.43 2.7 0.2699075
Head and Neck 15.39 5 0.7336607
Hepatocellular Carcinoma 18.07 3.6 0.7651938
Melanoma 36.62 13.3 0.6719016
Merkel Cell Carcinoma 43.12 4.3 0.5347724
Mesothelioma 13.08 1.8 0.7745981
NSCLC 22.41 6.8 0.5265
NSCLC-Nonsquamous 17.66 6.4 0.6018597
NSCLC-Squamous 19.77 9 0.4493587
Ovarian Cancer 9.29 3 0.3160356
Pancreatic Cancer 0 2 0.488062
Prostate Cancer 6.82 2.8 0.3467583
Renal Cell Carcinoma 23.69 2.7 0.6606
Sarcoma 10.91 2.3 0.4649772
Small-Cell Lung Cancer 14.51 9.9 0.4545876
Urothelial Carcinoma 19.03 6.8 0.5882142
Uveal Melanoma 36 13 04498145
Lymphoma 69 10 08156333
Thymic Carcinoma 21.9 2.5 0.5681527
Example 3
It is now known that TMB can be used to predict the effect of a tumor patient on treatment with an immune checkpoint inhibitor. The performance of TIGS in predicting ICI clinical response in individual patients was further evaluated in this example. To assess the predictive power of TIGS in ICI clinical response, an ICI dataset with both TMB and transcriptome data of individual patients was selected. A total of two melanoma and one urothelial cancer data sets were available for this analysis, the data sources being specifically as follows: the Van Allen 2015 dataset was downloaded from a supplement to the literature reference (Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, et al, Genomic reactors of stress to CTLA-4block in metallic media. science 2015; 350: 207-11). This data set investigated the clinical effect of CTLA-4 antibodies in metastatic melanoma. The Hugo 2016 dataset was downloaded from the reference (Hugo W, Zarretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, et al, genomic and Transcriptomic Features of Response to Anti-PD-1Therapy in Metastatic Melanoma. cell 2016; 165: 35-44). This data set investigated the therapeutic effect of PD-1 antibody in metastatic melanoma. The Snyder 2017 dataset (Snyder A, Nathanson T, Funt SA, Ahuja A, Buros Novik J, Hellmann MD, et al. content of system and systematic factors to clinical response and resistance to PD-L1blockade in a clinical analyzer. plos Med 2017; 14: e1002309) was downloaded from https:// githu. com/hammerlab/multi-organic-nuclear-polar-interpol 1. This data set investigated the therapeutic effect of PD-L1 antibody in urothelial cancer.
To assess the predictive performance of ICI clinical responses, Receiver Operating Characteristic (ROC) curves were used to measure false and true positive rates at various thresholds of TIGS or TMB values, which consistently better performance was obtained in all three ICI datasets compared to the widely used ICI response biomarker TMB, with specific results as shown in fig. 3, where a, ROC curves show the performance of TIGS and TMB in predicting anti-CTLA 4 immunotherapy responses in 35 melanoma patients (Van Allen 2015 dataset); b, ROC curves for TIGS and TMB predicting the performance of anti-PD-1 immunotherapy responses in 27 melanoma patients (Hugo 2016 dataset); c, ROC curves show the performance of TIGS and TMB in predicting anti-PD-L1 immunotherapy responses in 22 patients with urothelial cancer (Snyder 2017 dataset); d, grouping the patients according to the TMB or TIGS state, comparing the TMB-High with the TMB-Low (left panel, 100 patients) or the Kaplan-Meier (KM) total survival curve between the TIGS-High and TIGS-Low (right panel, 35 patients), Van Ellen 2015 data set; e, grouping patients according to TMB or TIGS state, comparing KM overall survival curve between TMB-High and TMB-Low (left panel, 37 patients) or TIGS-High and TIGS-Low (right panel, 26 patients), Hugo 2016 dataset; f, grouping patients according to TMB or TIGS state, comparing KM overall survival curve between TMB-High and TMB-Low (left panel, 22 patients) or TIGS-High and TIGS-Low (right panel, 22 patients), and Snyder 2017 data set. In all three available data sets, the Kaplan-Meier overall survival curves were further compared in patients with high/low TIGS or TMB levels. Patients with TIGS above the median were defined as "TIGS-high" and the rest as "TIGS-low". Similar definitions of "TMB-high" and "TMB-low". TMB-high patients showed better survival curves than TMB-low in all three ICI data sets, even though the difference did not reach significance in all three data sets, probably due to sample size. However, TIGS-high patients showed significantly better survival curves compared to TIGS-low in all three ICI data sets (FIG. 3). These analyses indicate that, in all three available data sets, TIGS consistently outperforms TMB in prediction of ICI clinical response.
In conclusion, the present invention effectively overcomes various disadvantages of the prior art and has high industrial utilization value.
The foregoing embodiments are merely illustrative of the principles and utilities of the present invention and are not intended to limit the invention. Any person skilled in the art can modify or change the above-mentioned embodiments without departing from the spirit and scope of the present invention. Accordingly, it is intended that all equivalent modifications or changes which can be made by those skilled in the art without departing from the spirit and technical spirit of the present invention be covered by the claims of the present invention.

Claims (12)

1. A computer-readable storage medium storing a computer program that when executed performs method steps comprising:
obtaining an antigen presentation index based on a GSVA algorithm according to mRNA expression data of an individual;
calculating to obtain APS according to formula Inormalized
Figure FDA0003248116790000011
In formula I:
APSnormalizeda normalized antigen presentation index;
APS refers to antigen presentation index;
APSpancan_minrefers to the lowest value of APS in the population of tumor patients corresponding to the individual;
APSpancan_maxrefers to the maximum value of APS in a population of tumor patients corresponding to the individual;
calculating to obtain tumor immunogenicity index according to formula II
TIGS=APSnormalizedX ln (TMB +1) (formula II);
in formula II:
TIGS refers to tumor immunogenicity index;
TMB refers to the tumor mutation burden, i.e., the total number of somatic gene coding errors, base substitutions, gene insertion or deletion errors detected per million bases.
2. The computer-readable storage medium of claim 1, wherein the individual is selected from an animal receiving tumor immunotherapy.
3. The computer-readable storage medium of claim 2, wherein the animal receiving tumor immunotherapy is a human.
4. The computer-readable storage medium of claim 2, wherein the tumor immunotherapy comprises an immune checkpoint inhibitor therapy.
5. The computer-readable storage medium of claim 2, wherein the tumor immunotherapy comprises a method of treatment of an individual by administering one or more of an anti-PD-1 antibody, a PD-L1 antibody, a CTLA-4 antibody.
6. The computer-readable storage medium of claim 1, wherein the mRNA expression data and/or TMB of the individual is derived from a tumor sample.
7. The computer-readable storage medium of claim 4, wherein the tumor is selected from melanoma, lung cancer, urothelial cancer, head and neck cancer, kidney cancer, lymphoma, prostate cancer, breast cancer, glioma, cervical cancer, endometrial cancer, esophageal cancer, liver cancer, mesothelioma, ovarian cancer, pancreatic cancer, paraganglioma, colon cancer, sarcoma, gastric cancer, testicular germ cell tumor, thyroid cancer, or thymoma.
8. The computer-readable storage medium of claim 1, wherein the genes used to calculate APS comprise at least 14 genes of PSMB5, PSMB6, PSMB7, PSMB8, PSMB9, PSMB10, TAP1, TAP2, ERAP1, ERAP2, CANX, CALR, PDIA3, TAPBP, B2M, HLA-A, HLA-B, and HLA-C.
9. The computer-readable storage medium of claim 1, wherein APSpancan_min=-1~-0.86。
10. The computer-readable storage medium of claim 1, wherein APSpancan_max=0.88~1。
11. An apparatus, comprising: a processor and a memory;
the memory for storing a computer program, the processor for executing the computer program stored by the memory to cause the terminal to perform method steps comprising:
obtaining mRNA expression data of an individual;
acquiring APS based on GSVA algorithm according to individual mRNA expression data;
calculating to obtain APS according to formula Inormalized
Figure FDA0003248116790000021
In formula I:
APSnormalizeda normalized antigen presentation index;
APS refers to antigen presentation index;
APSpancan_minrefers to the lowest value of APS in the population of tumor patients corresponding to the individual;
APSpancan_maxrefers to the maximum value of APS in a population of tumor patients corresponding to the individual;
calculating to obtain TIGS according to formula II
TIGS=APSnormalizedX ln (TMB +1) (formula II);
in formula II:
TIGS refers to tumor immunogenicity index;
TMB refers to the tumor mutation burden, i.e., the total number of somatic gene coding errors, base substitutions, gene insertion or deletion errors detected per million bases.
12. An apparatus, the apparatus comprising:
the mRNA expression data acquisition module is used for acquiring mRNA expression data of individuals;
the APS processing module is used for acquiring APS based on a GSVA algorithm according to individual mRNA expression data;
APSnormalizeda processing module for calculating APS according to formula Inormalized
Figure FDA0003248116790000031
In formula I:
APSnormalizeda normalized antigen presentation index;
APS refers to antigen presentation index;
APSpancan_minrefers to the lowest value of APS in the population of tumor patients corresponding to the individual;
APSpancan_maxrefers to the maximum value of APS in a population of tumor patients corresponding to the individual;
TIGS processing module for obtaining TIGS according to formula II calculation
TIGS=APSnormalizedX ln (TMB +1) (formula II);
in formula II:
TIGS refers to tumor immunogenicity index;
TMB refers to the tumor mutation burden, i.e., the total number of somatic gene coding errors, base substitutions, gene insertion or deletion errors detected per million bases.
CN201811376543.8A 2018-11-19 2018-11-19 Storage medium, equipment and device for determining tumor immunotherapy marker Active CN109448790B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811376543.8A CN109448790B (en) 2018-11-19 2018-11-19 Storage medium, equipment and device for determining tumor immunotherapy marker

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811376543.8A CN109448790B (en) 2018-11-19 2018-11-19 Storage medium, equipment and device for determining tumor immunotherapy marker

Publications (2)

Publication Number Publication Date
CN109448790A CN109448790A (en) 2019-03-08
CN109448790B true CN109448790B (en) 2021-11-09

Family

ID=65554570

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811376543.8A Active CN109448790B (en) 2018-11-19 2018-11-19 Storage medium, equipment and device for determining tumor immunotherapy marker

Country Status (1)

Country Link
CN (1) CN109448790B (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110223729B (en) * 2019-06-11 2023-04-07 上海科技大学 Simple and efficient tumor immunotherapy biomarker and application thereof
CN112133365B (en) * 2020-09-03 2022-05-10 南方医科大学南方医院 Gene set for evaluating tumor microenvironment, scoring model and application of gene set
CN112992266B (en) * 2021-02-05 2021-09-21 深圳裕康医学检验实验室 Method, device and storage medium for evaluating tumor immune exhaustion state
CN114974420A (en) * 2021-10-29 2022-08-30 无锡臻和生物科技有限公司 Marker for predicting curative effect of gastrointestinal tumor immune checkpoint inhibitor and application thereof
CN114231628A (en) * 2021-12-03 2022-03-25 无锡臻和生物科技有限公司 Marker combination for predicting curative effect of gastrointestinal tumor immune checkpoint inhibitor and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1731619A2 (en) * 1998-05-11 2006-12-13 The University Of Nottingham Tumour markers
CN108107213A (en) * 2017-11-10 2018-06-01 上海科技大学 A kind of tumour immunity biomarker and application thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112016000853A2 (en) * 2013-07-16 2017-12-12 Genentech Inc methods for treating or delaying, reducing or inhibiting cancer relapse or progression and the progression of an immune-related disease in an individual, to increase, improve or stimulate an immune response or function in an individual and kit.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1731619A2 (en) * 1998-05-11 2006-12-13 The University Of Nottingham Tumour markers
CN108107213A (en) * 2017-11-10 2018-06-01 上海科技大学 A kind of tumour immunity biomarker and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Cytolytic Activity Score to Assess Anticancer Immunity in Colorectal Cancer;Sumana Narayanan等;《Annals of Surgical Oncology》;20180516;全文 *
肿瘤相关抗原及抗体作为肿瘤标志物的研究及应用现状;管星;《中国保健营养》;20130831;全文 *

Also Published As

Publication number Publication date
CN109448790A (en) 2019-03-08

Similar Documents

Publication Publication Date Title
CN109448790B (en) Storage medium, equipment and device for determining tumor immunotherapy marker
Wang et al. Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction
Zare et al. An evaluation of copy number variation detection tools for cancer using whole exome sequencing data
Finotello et al. Molecular and pharmacological modulators of the tumor immune contexture revealed by deconvolution of RNA-seq data
Cheng et al. Comprehensive detection of germline variants by MSK-IMPACT, a clinical diagnostic platform for solid tumor molecular oncology and concurrent cancer predisposition testing
McNamara et al. Impact of high tumor mutational burden in solid tumors and challenges for biomarker application
AU2019228512B2 (en) Systems and methods for detection of residual disease
Amarasinghe et al. CoNVEX: copy number variation estimation in exome sequencing data using HMM
Richard et al. Exome analysis reveals genomic markers associated with better efficacy of nivolumab in lung cancer patients
CN106676178B (en) Method and system for evaluating tumor heterogeneity
JP6975350B2 (en) Surrogate markers and methods for measuring tumor mutation levels
Zhao et al. A leukocyte infiltration score defined by a gene signature predicts melanoma patient prognosis
Chibuk et al. Horizons in veterinary precision oncology: fundamentals of cancer genomics and applications of liquid biopsy for the detection, characterization, and management of cancer in dogs
CN110997943A (en) Methods of assessing the suitability of cancer immunotherapy
Varn et al. AB cell-derived gene expression signature associates with an immunologically active tumor microenvironment and response to immune checkpoint blockade therapy
Golkaram et al. Spatiotemporal evolution of the clear cell renal cell carcinoma microenvironment links intra-tumoral heterogeneity to immune escape
Bortone et al. Improved T-cell receptor diversity estimates associate with survival and response to anti–PD-1 therapy
Shui et al. Baseline and post-treatment biomarkers of resistance to anti-PD-1 therapy in acral and mucosal melanoma: an observational study
Jiang et al. Tertiary lymphoid structure patterns predicted anti-PD1 therapeutic responses in gastric cancer
CA2889276A1 (en) Method for identifying a target molecular profile associated with a target cell population
Lo et al. Indication-specific tumor evolution and its impact on neoantigen targeting and biomarkers for individualized cancer immunotherapies
CN110223729B (en) Simple and efficient tumor immunotherapy biomarker and application thereof
CN116179701A (en) Marker gene for predicting curative effect of esophageal squamous carcinoma immunotherapy and application thereof
Liu et al. Predicting patient outcomes after treatment with immune checkpoint blockade: A review of biomarkers derived from diverse data modalities
Wood et al. Burden of tumor mutations, neoepitopes, and other variants are cautionary predictors of cancer immunotherapy response and overall survival

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
GR01 Patent grant
GR01 Patent grant