CN111733253B - Marker for immune-related adverse reaction and application thereof - Google Patents

Marker for immune-related adverse reaction and application thereof Download PDF

Info

Publication number
CN111733253B
CN111733253B CN202010846330.8A CN202010846330A CN111733253B CN 111733253 B CN111733253 B CN 111733253B CN 202010846330 A CN202010846330 A CN 202010846330A CN 111733253 B CN111733253 B CN 111733253B
Authority
CN
China
Prior art keywords
immune
related adverse
lcp1
adverse reaction
adpgk
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
CN202010846330.8A
Other languages
Chinese (zh)
Other versions
CN111733253A (en
Inventor
薛新颖
景莹
庄光磊
刘锦
臧学磊
高杰
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 Xinnuo Weikang Technology Co ltd
Original Assignee
Beijing Xinnuo Weikang Technology 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 Xinnuo Weikang Technology Co ltd filed Critical Beijing Xinnuo Weikang Technology Co ltd
Priority to CN202011352976.7A priority Critical patent/CN112557668B/en
Priority to CN202010846330.8A priority patent/CN111733253B/en
Publication of CN111733253A publication Critical patent/CN111733253A/en
Application granted granted Critical
Publication of CN111733253B publication Critical patent/CN111733253B/en
Priority to PCT/CN2021/112310 priority patent/WO2022037474A1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4712Muscle proteins, e.g. myosin, actin, protein

Abstract

The present invention relates to immune-related adverse reaction biomarkers and uses thereof, comprising LCP1 and/or ADPGK. By adopting the Immune-related adverse reactions (IRAEs) biomarker and the application thereof, the invention provides the kit and the method which are helpful for early prediction and active intervention of Immune-related adverse reactions caused by anti-PD-1/PD-L1 antibody treatment, thereby improving the prognosis effect of patients.

Description

Marker for immune-related adverse reaction and application thereof
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to a marker for immune-related adverse reactions and application thereof.
Background
Anti-programmed death receptor 1 (PD-1)/anti-programmed death ligand 1 (PD-L1) antibody therapy activates the body's Immune response and breaks Immune homeostasis, and Immune-related adverse reactions (iraEs) triggered thereby affect the organs of the body and, in severe cases, can lead to death. Pneumonia is the most common fatal injury of irAEs, leading to death in 10% of patients, accounting for 35% of the mortality associated with anti-PD-1/PD-L1 antibody treatment. Myocarditis is the most important lethal injury of irAEs with mortality rates of up to 50%. Thus, the discovery of irAEs biomarkers is determinative of the prediction of the ratio of therapeutic benefit to risk for patients receiving anti-PD-1/PD-L1 antibody treatment. T Cell Receptor (TCR) diversity, CD8+ T cell clonal expansion, tumor burden mutation (TMB) have been reported to have predictive irAEs potential, but these conclusions are either established in a one-way analysis or limited to a limited number of cases. Therefore, comprehensive analytical studies on the prediction of irAEs biomarkers are needed.
Disclosure of Invention
Based on the above-mentioned state of the art, the inventors tried to provide an Immune-related adverse effect (irAEs) biomarker, thereby providing a kit for early prediction and active intervention of Immune-related adverse effects caused by anti-PD-1/PD-L1 antibody therapy, and applications thereof. In order to realize the purpose of the invention, the following technical scheme is adopted:
one aspect of the invention relates to immune-related adverse reaction biomarkers comprising LCP1 and/or ADPGK. LCP1 belongs to actin-plastin family, and under the co-stimulation of CD3/CD2 or CD3/CD8, LCP promotes the activation of T cells by accelerating the transport of CD69 and CD25 to the surfaces of the T cells; ADPGK mediates metabolic switching in T cell activation, accelerates glycolysis, reduces mitochondrial respiration, and enhances T cell glucose uptake. The invention unexpectedly finds that LCP1 and ADPGK are biomarkers for predicting the occurrence of iraE, and particularly the AUC value of the combination of LCP1 and ADPGK reaches over 0.8.
Another aspect of the invention relates to a pre-test kit for immune-related adverse reactions comprising reagents for detecting the expression level of LCP1 and/or ADPGK. Preferably, the agent is an immunochemical agent.
The invention also relates to application of the marker or the pre-test kit in preparing a kit for predicting immune-related adverse reactions.
In a preferred embodiment of the invention, the immune-related side effects are immune-related side effects caused by anti-PD-1/PD-L1 antibody treatment. Such immune-related adverse reactions include, but are not limited to, pneumonia, myocarditis, colitis, pancreatitis, hypothyroidism, hyperthyroidism, thyroiditis, hypophysitis, type i diabetes, adrenal insufficiency, sarcoidosis, vitiligo, severe adverse skin reactions, thrombocytopenia, hepatitis, gastrointestinal toxicity, nervous system disorders, nephritis, uveitis. Particularly preferred immune-related adverse reactions include enzootic pneumonia.
In one aspect of the invention, the anti-PD-1 antibody includes, but is not limited to, nivolumab, pembrolizumab, cimicimab, and the anti-PD-L1 includes, but is not limited to, atelizumab, avizumab, and dulvacizumab.
In a preferred embodiment of the invention, the subject treated with the anti-PD-1/PD-L1 antibody includes, but is not limited to, lung adenocarcinoma; SKCM, cutaneous melanoma; PRAD, prostate cancer; BLCA, bladder urothelial cancer; MESO, mesothelioma; BRCA, breast invasive carcinoma; CESC, cervical squamous carcinoma and cervical adenocarcinoma; PAAD, pancreatic cancer; OV, ovarian serous cystadenocarcinoma; HNSC, head and neck squamous cell carcinoma; STAD, gastric adenocarcinoma; THCA, thyroid cancer; CHOL, cholangiocarcinoma; ACC, adrenocortical carcinoma; READ, rectal adenocarcinoma; COAD, colon cancer; LIHC, hepatocellular carcinoma; LGG, brain low-grade glioma; GBM, glioblastoma multiforme; UVM, uveal melanoma; UCS, uterine carcinosarcoma.
Advantageous effects
By adopting the Immune-related adverse reactions (IRAEs) biomarker and the application thereof, the invention provides the kit and the method which are helpful for early prediction and active intervention of Immune-related adverse reactions caused by anti-PD-1/PD-L1 antibody treatment, thereby improving the prognosis effect of patients.
Drawings
Fig. 1 is a graph showing calculation of irAE Report Odds Ratio (ROR) for 26 cancer types, and evaluation of correlation between immunotherapy response-related factors and ROR. a. Histological location of the different carcinogenesis (left) and the respective irAE ROR (right); b. spearman correlation test was performed for irAE ROR and 36 immunotherapy response-related factors, with the red bar representing positive correlation and the blue bar representing negative correlation. Indicates significant correlation (FDR)<0.05); the common relevant factor for irAE/immunotherapy is indicated by orange. c. And establishing a two-variable model to evaluate the irAE prediction effect of the combination of the irAE significance related factors of the top 6 ranks. Spearman R (Rs) calculated the correlation between prediction and observed irAE ROR. Boxes are light in color indicating Rs, size indicating significance of log-likelihood ratio test d.tcr diversity and combined predictive effect of CD8+ T cell bivariate model (Spearman correction, Rs =0.75, FDR =8.24 × 10-4). Two-variable model equation: 0.31 × TCR diversity + 8.87 × 13 CD8+T Cells + 0.27. LUAD, lung adenocarcinoma; SKCM, cutaneous melanoma; PRAD, prostate cancer; BLCA, bladder urothelial cancer; MESO, mesothelioma; BRCA, breast invasive carcinoma; CESC, squamous carcinoma of the cervix and cervical glandCancer; PAAD, pancreatic cancer; OV, ovarian serous cystadenocarcinoma; HNSC, head and neck squamous cell carcinoma; STAD, gastric adenocarcinoma; THCA, thyroid cancer; CHOL, cholangiocarcinoma; ACC, adrenocortical carcinoma; READ, rectal adenocarcinoma; COAD, colon cancer; LIHC, hepatocellular carcinoma; LGG, brain low-grade glioma; GBM, glioblastoma multiforme; UVM, uveal melanoma; UCS, uterine carcinosarcoma;
fig. 2 is a diagram showing a comprehensive discrimination irAE potential predictor. a. Pathway enrichment analysis was performed on the top 10 genes most significantly associated with irAE ROR of multiple cancer types. b. The correlation line Spearman test for LCP1 and irAE ROR. c. The first 10 genes that were significantly related to irAE ROR were analyzed by a bivariate predictive model. Spearman correlation (Rs) tests were performed on the predicted and observed irAE ROR. The color shade of the squares indicates the magnitude of Rs, which indicates the significance difference of the log-likelihood test. Lcp1 and ADPGK were combined for two-factor model evaluation (Spearman correlation, Rs =0.91, FDR =7.94 × 10)-9). Two-variable regression model formula: 0.37 XLCP 1+ 0.70 XADPGK-9.10.
Figure 3 is a graph showing the predictive ability to validate LCP1 and ADPGK in an independent patient cohort. a. Immunohistochemical staining pictures of ADPGK and LCP1 in patients with and without irAE. The picture size is as follows: 200X 200 μm2. b. LCP and ADPGK immunohistochemical staining signals were quantified. Unpaired two-tailed student's t test was performed between immunohistochemical staining signals from patients who developed irAE and those who did not. c. The geometric mean of the LCP1 and ADPGK staining signals was calculated. The unpaired two-tailed student's t test was performed on patients who developed irAE and on patients who did not develop irAE. d. ROC curves for LCP1, ADPGK, LCP1+ ADPGK are shown for this patient cohort (n = 28).
FIG. 4 is a graph showing the Spearman correlation test for iriAE ROR and objective remission rate in 21 cancer types.
Fig. 5 is a graph showing irAE ROR and a cytolytic activity, B. interferon γ signature, c.pd-1 expression, d.tcr diversity, e. estimated M1 macrophage abundance, f. estimated CD8+ T cell abundance, g. estimated naive B cell abundance, in patients treated with anti-PD-1/PD-L1 in each cancer type. The color of the dots represents different tumor types.
Fig. 6 is a graph showing the predicted effect of a bivariate model calculating all combinations of 7 most significant correlation factors. The results only show combinations of significant p-values for the log-likelihood test.
Fig. 7 is a graph showing Variance Inflectionfactor (VIF) showing 7 most significant correlation factors with irAE ROR.
FIG. 8 is a graph showing a Spearman correlation test for iraE ROR and the top 10 significantly related genes (except LCP 1). The X-axis represents the expression level of the gene transformed with log 2.
Fig. 9 is a graph showing the predicted effect of two-by-two combinations of the first 10 genes associated with the bivariate model evaluation and irAE ROR. The results only show combinations of significant p-values for the log-likelihood test.
Fig. 10 is a graph showing Variance Inflation Factor (VIF) demonstrating the top 10 significantly related genes.
Fig. 11 is a diagram showing the testing of the predictive model by the independent patient cohort. a. LCP1, ADPGK, LCP1+ ADPGK gene expression in brain regions with and without irAE. b. LCP1, ADPGK, LCP1+ ADPGK gene expression in the presence of irAE heart muscle and in the absence of irAE smooth muscle. The calculation of LCP1+ ADPGK model scores was built on the LCP1+ ADPGK two-variable model (fig. 2 d). TPM: number of transcripts per million kilobases.
FIG. 12 is a graph showing two-variable model detection of the combination of the first 7 factors and the first 10 genes of iraE. Spearman R (Rs) calculated the correlation between prediction and observed irAE ROR. The squares are light and dark in color to indicate Rs and large in size to indicate the significance of the log-likelihood ratio test. b. The two-factor model calculated the combined effect of CD8+ T cells and LCP1 (Spearman corporation, Rs =0.87, FDR =2.84 × 10-72).
FIG. 13 is a graph showing the predictive effect of the bivariate model evaluating the first 7 factors and the first 10 related genes in combinations of two. The results only show combinations of significant p-values for the log-likelihood test.
FIG. 14 is a graph showing the objective remission rate against PD-1/PD-L1 and LCP1, ADPGK expression for a Spearman correlation test in 21 tumor types.
Figure 15 is a graph showing the predictive ability to test LCP1 and ADPGK in a lung tumor patient cohort. a. Lung cancer patients were quantitatively analyzed for ADPGK, LCP1 immunohistochemical staining signals. Unpaired two-tailed t-tests were performed between the groups with and without irAE. b. The geometric mean of the staining signals for ADPGK, LCP1 was calculated. Unpaired two-tailed t-tests were performed between the groups with and without irAE. c. LCP1, ADPGK, LCP1+ ADPGK ROC curve (n = 26) of this patient cohort.
Detailed Description
In order to further understand the present invention, the technical solutions in the embodiments of the present invention will be clearly and completely described below with reference to 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 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.
Unless otherwise specified, the reagents involved in the examples of the present invention are all commercially available products, and all of them are commercially available.
Example 1
1 method of experiment
(1.1) data analysis of FAERS personal safety reports
The invention relates to FAERS (https:// www.fda.gov/drugs/queries-and-answers-fdas-overture-event-
reporting-system-faces/fda-added-event-reporting-system-faces-public-dashboard) obtains personal security reports from 7/1/2014 to 6/30/2019. Adverse reaction reports were collected only for patients receiving anti-PD-1 antibodies (nivolumitumumab, pembrolizumab, cimetizumab) and anti-PD-L1 (alemtuzumab, avizumab, doxumab) and excluded patients (leprimumab, tremelimumab) who had received anti-CTLA-4 antibody treatment at the same time. The irAE was defined using the peer approved irAE administration manual as a standard. And (4) adopting an imbalance analysis, and calculating ROR of the risk occurrence of iraE by using the whole database as a comparison sample. Patients were classified into the irAE group if any of the irAE types were produced.
(1.2) analysis of TCGA database and independent database
Molecular data including mRNA expression, miRNA expression, protein expression, somatic mutations were obtained for 26 cancer types from the TCGA database (https:// portal.gdc.candr.gov /). TCR diversity, neoantigen load, predicted immune cell abundance, and intra-tumor heterogeneity data were obtained from GDC PanImmune data (https:// GDC. cancer. gov/aboutdata/publications-/panimmun). TMB counts the number of non-silent somatic mutations per sample. The "GSVA" R package was used to calculate T-cell inflammatory Gene Expression (GEP) for each sample, with the T-cell inflammatory GEP signature and interferon gamma signature defined by the eyers et al. The cytolytic activity is defined as the geometric mean of the expression levels of two cytolytic marker genes (GZMA, PRF 1). RNA-seq data was obtained from Johnson et al for brain areas affected/unaffected by iraE, and for heart muscles affected by iraE and unaffected smooth muscles.
(1.3) identification of biomarkers combining multiple sets of mathematical data and real world data
In the analysis of the present invention, the number of cancer types was much lower than the number of variables (26 cancer types)>The 50,000 variables, including 20,000 mRNA expression, 12,000 non-coding RNA expression, 18,000 gene mutation, 200 protein expression, and 2,400miRNA expression) may lead to the first class of errors, causing more false positives when using other advanced algorithms, such as Lasso, Elastic net, Ridge. Therefore, the invention optimizes the calculation result by adding variables. Median values for each factor in each cancer type were calculated. The human anatomy diagrams were plotted using the "gganatogram" R-package and the irAE ROR was predicted in two-and three-variable linear regression models using the Cross-Validation test (Leave-One-Out Cross Validation) method provided by the "caret" R-package (see fig. 8). Using Spearman rank correlation coefficient (Rs) and unexplained coefficient of variation (1-Rs)2) Evaluating the prediction. The goodness of fit of the models was compared using the log-likelihood ratio test in the R package "lmtest". And carrying out log likelihood ratio test on the two-factor model and the single factor with higher Rs in the two factors to obtain data between the fitness of the two-variable model. For the examination of the three-factor fitness, a row log-likelihood ratio test between a two-factor model and a three-factor model is adopted. The multiple collinearity was evaluated using the 'vif' function in the R package 'car' to calculate the coefficient of variance expansion. The pathway enrichment was calculated using the R package "clusterprofiler". The area under the ROC curve was calculated using R package 'pROC'. Significant difference was defined as two-tailed P<0.05 and/or FDR<0.05。
(1.4) immunohistochemistry
The study of the invention conforms to the ethical guidelines of the protective general guidelines of the U.S. subjects and is approved by the ethical committee of the general hospital of the chinese liberation force. All patients signed informed consent. All patients received treatment at the general hospital of the chinese liberation force and were analyzed retrospectively for clinical information and tissue samples. Formalin-fixed paraffin embedding (FFPE) was obtained in pathological examination. FFPE tissue sections were 5 μm thick and immunohistochemical examination was performed. The primary antibody was either LCP1 (1: 200, Cell Signaling Technology #3588) or ADPGK (1:900, Novus Biologicals # NBP 1-91653). After washing the primary antibody, horseradish peroxidase-labeled secondary antibodies were incubated, followed by color development using a DAB horseradish peroxidase color development kit (Dako). The section is counterstained by hematoxylin, dehydrated and loaded with broken sections and sealed. The whole piece was scanned using Aperio ScanScope system (Leica Biosystems) and the staining results were quantified using the positive pixel count v9 (PPCv9) algorithm provided by Aperio ImageScope software v 14.3. Necrotic areas or image defects are ignored. 7 random fields of view of 20 times were selected for each section to count the average color signal of the section.
2. Results and discussion
(2.1) analysis of the role of known factors in predicting irAEs
To identify potential biomarkers for anti-PD-1/PD-L1 antibody treatment of iraEs, the present invention was derived from the U.S. Food and Drug Administration (FDA) adverse event reporting systemThe system (FAERS) obtained information on 18,706 patients receiving anti-PD-1/PD-L1 antibody therapy covering 26 cancer types, 52,282 adverse reaction (AEs) events. Of these, 3,706 (19.8%) patients developed at least one irAEs. The present invention calculates the proportion of irAEs reported by anti-PD-1/PD-L1 antibody treatment and the proportion of irAEs reported by other drugs in the database, and calculates the Reporting Odds Ratio (ROR) of irAEs caused by anti-PD-1/PD-L1 antibody treatment. IrAE ROR varies between different cancer types and has the highest value for lung adenocarcinoma (LUAD) (3.29, 95% confidence interval [ CL)]2.97-3.65), uterine sarcoma (UCS) was the lowest value (0.65, 95% CL, 0.02-4.18) (fig. 1 a). The present invention analyzes 6 irAE-related factors including TMB, T cell receptor diversity, interferon gamma expression, tumor necrosis factor alpha expression, eosinophils and neutrophils. Surprisingly, these factors show a positive correlation between irAE incidence and benefit rate in patients receiving immune checkpoint inhibitors and are therefore also biomarkers of immunotherapeutic response. It was also observed that there was only a slight significant correlation between irAE ROR and Objective Remission Rate (ORR) for anti-PD-1/PD-L1 antibody treatment (Rs =0.44; P =0.049; fig. 4). Next, 36 factors associated with immunotherapy response were examined, including TMB, cytolytic activity, neoantigen loading. Molecular data for these factors were obtained from The tumor Genome project (TCGA) database, irAE risk reports were obtained from FAERS, and correlations between molecular data for each factor and irAE risk index were calculated. 7 potential predictors were found, including cytolytic activity (Spearman R, Rs =0.64; False discovery rate [ FDR ]]= 0.01), interferon gamma signature (Rs =0.61, FDR = 0.01), PD-1 expression (Rs =0.60, FDR = 0.01), TCR diversity (Rs =0.59, FDR = 0.01), M1 type macrophages (Rs =0.55, FDR = 0.03), CD8+ T cell abundance (Rs =0.50, FDR = 0.05), naive B cells (Rs =0.49, FDR = 0.05) (fig. 1B; fig. 5). To obtain a more accurate prediction model, the Spearman correlation coefficient and log-likelihood ratio fitness test value (log-likelihood ratio test) of the 7 factors were subjected to a two-variable model evaluationAnd (6) estimating. The use of CD8+ T cell abundance in combination with TCR diversity or naive B cell factors significantly improved the goodness of fit of the model compared to the use of a single factor (fig. 1c; fig. 5 and 6). Especially when CD8+ T cell abundance and TCR diversity are considered in combination, the best predictive level can be reached ((Rs =0.75, FDR =8.24 × 10)-4) (FIG. 1 d). The correlation coefficient (Rs, 0.75) accounts for 56% of the observed iraE ROR (Rs)20.56) can be interpreted using the two-variable regression model. Multiple collinearity of these 7 factors was assessed using Variance Inflation Factor (VIF), with no multiple collinearity observed in TCR diversity and CD8+ T cells (fig. 7). Meanwhile, no significant correlation was found between TCR diversity and CD8+ T cell abundance (P = 0.26), indicating that the two are independent in predicting irAE. The prediction effect of other factors combined with a TCR diversity-CD 8+ T cell abundance bivariate model is further evaluated, and no trivariate model is found to be helpful for improving the correlation coefficient or increasing the accuracy.
(2.2) comprehensive identification of potential biomarkers for irAE
mRNA, miRNA, lncRNA, protein expression, non-silent gene mutations of 26 cancer types were screened in full in an attempt to find novel predictors of irAE. The top-ranked predictors are mostly genes whose expression changes and are highly enriched in the course of immune responses, including T cell activation and cell killing (fig. 2 a). This result further confirms that T cells are a key regulatory factor for irAEs. Surprisingly, the correlation coefficient level of lymphocyte plasma-encapsulating protein 1 (LCP 1) involved in T cell activation was highest (Rs =0.82, FDR = 156.69 × 10)-3Fig. 2 b). Further two-variable model evaluation of the irAE related genes ranked in the top ten shows that LCP1 can achieve better prediction effect in combination with most other irAE related genes (FIG. 2c and FIG. 9). Linear regression analysis of all bivariate models was performed and when adenosine diphosphate dependent glucokinase (ADPGK), which mediates metabolic switching in T cell activation, was used in combination with LCP1, it was possible to achieveBest accuracy (Rs =0.91, FDR =7.94 × 10-9, fig. 2 d). VIF was also used to evaluate the multiple collinearity of the top 10 genes, with no multiple collinearity observed in LCP1 and ADPGK (fig. 10). The increased 3 rd gene did not improve the predictive value of the LPC1-ADPGK two-variable model. Significant increases in LCP1 and ADPGK expression were observed in the area/tissue where irAE occurred compared to the area/tissue where irAE did not occur (fig. 11), which validated the accuracy of the model. Further evaluation of the combined effect of all the significant factors and genes is expected to find a more powerful combined evaluation mode, but no better performing model is found (fig. 12 and 13). Given the straightforward and unequivocal assessment of LCP1 and ADPGK, this result may be more readily translated to clinical use. There are no reports showing that LCP1 and/or ADPGK are associated with immunotherapeutic responses. Further correlation tests of LCP1/ADPGK and objective remission rates revealed no significant correlation (figure 14), indicating that there was only a limited confounding effect between changes in LCP1 and ADPGK expression and efficacy.
(2.3) verification that LCP1 and ADPGK are irAE biomarkers
To examine the predictive power of LCP1 and ADPGK, a validated cohort of 28 patients receiving anti-PD-1/PD-L1 inhibitor treatment with high quality Formaldehyde Fixed Paraffin Embedded (FFPE) tissue sections and clinical pathology information was collected. The median age of the patients was 56 years (range, 37 to 82 years), of which 22 (78.6%) were male patients and 6 (21.4%) were female patients. Lung cancer was confirmed in 26/28 (92.9%) patients. Immunohistochemistry was used to examine the levels of LCP1 and ADPGK expression in patients in the validation cohort. LCP1 and ADPGK stained significantly more in the irAE group (fig. 3 a). Immunostaining signals for LCP1 and ADPGK were quantified using the Positive Pixel Count v9 (PPCv9) algorithm of Aperio ImageScope software (v 14.3). Results as expected, the expression of LCP1 ((p-value = 0.008) and ADPGK (p-value = 0.010) in patients with irAE was significantly higher than that in patients without irAE found (fig. 3b), the geometric mean of LCP1 and ADPGK expression was also significantly higher in patients with irAE ((p-value = 0.005, fig. 3c), the area under the curve (AUC) of the receiver operating characteristic curve (rock) of LCP1 and ADPGK predicting irAE was 0.78 and 0.78, respectively, and the combination of LCP1 and ADPGK resulted in a better AUC area (0.8, fig. 3 d), furthermore, the success of LCP1, ADPGK, 1+ ADPGK predicted localized pneumonia in 26 patients with lung cancer, the AUC was 0.74, 0.76, and 0.77, respectively, and the combined LCP 6777 and ADPGK predicted the occurrence of a certain type of anti-pgk inhibitor in patients was able to verify that the combined ADPGK was able to predict the occurrence of a certain type of irAE in patients with a certain type of ADPGK 6778, LCP 6777, and/3 d, and the combined LCP 3 g Whether irAE will occur.
(2.4) conclusion
In the study of the present invention, real world and molecular data from patients receiving anti-PD-1/PD-L1 therapy were integrated in 26 tumor types and systematically analyzed for potential predictors of risk of developing irAE. 7 potential predictors were identified, the CD8+ T cell and TCR diversity combinations achieved the best prediction accuracy for iraE, with an unexplained coefficient of variation from 0.59 (1-0.64)2) Down to 0.44 (1-0.75)2). Considering that the unexplained coefficient of variation is still at 0.44, a large-scale comprehensive screen was performed to obtain better irAE ROR predictors. New possible irAE predictors were found to be enriched during T cell activation. The combination of the two genes related to T cell activation, LCP1 and ADPGK, evaluated in a linear regression model, gave an unexplained coefficient of variation from 0.44 ((1-0.75)2) Reduced to 0.17 (1-0.91)2). Furthermore, AUC values for LCP1 and ADPGK reached 0.8 in the patient level validation cohort, indicating that LCP1 and ADPGK in combination are biomarkers predictive of irAE occurrence.
The foregoing describes preferred embodiments of the present invention, but is not intended to limit the invention thereto. Modifications and variations of the embodiments disclosed herein may be made by those skilled in the art without departing from the scope and spirit of the invention.

Claims (6)

1. The application of the biomarker of the immune-related adverse reaction containing LCP1 in the preparation of a kit for predicting the immune-related adverse reaction; the immune-related adverse reaction is the immune-related adverse reaction caused by the anti-PD-1/PD-L1 antibody treatment.
2. The application of the immune-related adverse reaction biomarker comprising LCP1 and ADPGK in the preparation of a kit for predicting immune-related adverse reactions; the immune-related adverse reaction is the immune-related adverse reaction caused by the anti-PD-1/PD-L1 antibody treatment.
3. The application of a pre-kit for immune-related adverse reactions of a reagent for detecting the expression level of LCP1 in the preparation of a kit for predicting the immune-related adverse reactions; the immune-related adverse reaction is the immune-related adverse reaction caused by the anti-PD-1/PD-L1 antibody treatment.
4. The application of a pre-kit for immune-related adverse reactions of reagents for detecting the expression levels of LCP1 and ADPGK in the preparation of a kit for predicting immune-related adverse reactions; the immune-related adverse reaction is the immune-related adverse reaction caused by the anti-PD-1/PD-L1 antibody treatment.
5. The use according to any one of claims 1 to 4, wherein the anti-PD-1 antibody is selected from the group consisting of one or more of nivolumab, pembrolizumab, and cimetizumab; the anti-PD-L1 is selected from one or more of atelizumab, avizumab and Duvacizumab.
6. The use according to claim 5, wherein the subject treated with the anti-PD-1/PD-L1 antibody is lung adenocarcinoma; cutaneous melanoma; prostate cancer; bladder urothelial cancer; mesothelioma; invasive carcinoma of the breast; squamous carcinoma of the cervix and adenocarcinoma of the cervix; pancreatic cancer; ovarian serous cystadenocarcinoma; squamous cell carcinoma of the head and neck; gastric adenocarcinoma; thyroid cancer; bile duct cancer; adrenocortical carcinoma; rectal adenocarcinoma; colon cancer; hepatocellular carcinoma; brain low-grade glioma; glioblastoma multiforme; patients with uveal melanoma or uterine carcinosarcoma.
CN202010846330.8A 2020-08-21 2020-08-21 Marker for immune-related adverse reaction and application thereof Active CN111733253B (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN202011352976.7A CN112557668B (en) 2020-08-21 2020-08-21 Marker for immune-related adverse reaction and application thereof
CN202010846330.8A CN111733253B (en) 2020-08-21 2020-08-21 Marker for immune-related adverse reaction and application thereof
PCT/CN2021/112310 WO2022037474A1 (en) 2020-08-21 2021-08-12 Immune-related adverse event biomarker and prediction kit

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010846330.8A CN111733253B (en) 2020-08-21 2020-08-21 Marker for immune-related adverse reaction and application thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN202011352976.7A Division CN112557668B (en) 2020-08-21 2020-08-21 Marker for immune-related adverse reaction and application thereof

Publications (2)

Publication Number Publication Date
CN111733253A CN111733253A (en) 2020-10-02
CN111733253B true CN111733253B (en) 2020-12-22

Family

ID=72658651

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202011352976.7A Active CN112557668B (en) 2020-08-21 2020-08-21 Marker for immune-related adverse reaction and application thereof
CN202010846330.8A Active CN111733253B (en) 2020-08-21 2020-08-21 Marker for immune-related adverse reaction and application thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202011352976.7A Active CN112557668B (en) 2020-08-21 2020-08-21 Marker for immune-related adverse reaction and application thereof

Country Status (2)

Country Link
CN (2) CN112557668B (en)
WO (1) WO2022037474A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112557668B (en) * 2020-08-21 2022-06-10 北京信诺卫康科技有限公司 Marker for immune-related adverse reaction and application thereof
CN115831216B (en) * 2022-08-26 2023-08-25 中山大学孙逸仙纪念医院 Tumor immune treatment efficacy prediction model based on tumor immune microenvironment and construction method thereof
CN115807084B (en) * 2022-09-13 2023-11-03 南方医科大学南方医院 Biomarker for predicting adverse events of blocking treatment of tumor immune checkpoints and application of biomarker
CN117129678A (en) * 2023-08-18 2023-11-28 深圳大学 Use of biomarkers in connection with assessment of tuberculous pleural effusion

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103237901B (en) * 2010-03-01 2016-08-03 卡里斯生命科学瑞士控股有限责任公司 For treating the biomarker of diagnosis
CN103038635B (en) * 2010-05-11 2016-12-28 威拉赛特公司 For diagnosing the method and composition of condition of illness
WO2013169388A1 (en) * 2012-05-08 2013-11-14 H. Lee Moffitt Cancer Center And Research Institute, Inc. Predictive biomarkers for ctla-4 blockade therapy and for pd-1 blockade therapy
WO2017069958A2 (en) * 2015-10-09 2017-04-27 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
CN109715802A (en) * 2016-03-18 2019-05-03 卡里斯科学公司 Oligonucleotide probe and application thereof
WO2019117132A1 (en) * 2017-12-12 2019-06-20 地方独立行政法人神奈川県立病院機構 Biomarker for prognostic prediction of cancer immunotherapy
CN110063970A (en) * 2019-04-30 2019-07-30 上海心脉途医疗科技有限公司 The treatment and prevention method of intestinal flora relevant to irAE and irAE
CN112557668B (en) * 2020-08-21 2022-06-10 北京信诺卫康科技有限公司 Marker for immune-related adverse reaction and application thereof

Also Published As

Publication number Publication date
CN112557668B (en) 2022-06-10
WO2022037474A1 (en) 2022-02-24
CN111733253A (en) 2020-10-02
CN112557668A (en) 2021-03-26

Similar Documents

Publication Publication Date Title
CN111733253B (en) Marker for immune-related adverse reaction and application thereof
Fernandez-Garcia et al. Plasma cell-free DNA (cfDNA) as a predictive and prognostic marker in patients with metastatic breast cancer
Ambatipudi et al. DNA methylome analysis identifies accelerated epigenetic ageing associated with postmenopausal breast cancer susceptibility
Wolf et al. Posttraumatic psychopathology and the pace of the epigenetic clock: a longitudinal investigation
Telesco et al. Gene expression signature for prediction of golimumab response in a phase 2a open-label trial of patients with ulcerative colitis
Dai et al. Breast cancer risk assessment with five independent genetic variants and two risk factors in Chinese women
Sharma et al. Validation of the DNA damage immune response signature in patients with triple-negative breast cancer from the SWOG 9313c trial
Sharma et al. Predicting pancreatic cancer in the UK biobank cohort using polygenic risk scores and diabetes mellitus
Kresovich et al. Epigenetic mortality predictors and incidence of breast cancer
JP2020522690A (en) Method and system for identifying or monitoring lung disease
US20190249228A1 (en) Biomarkers for prospective determination of risk for development of active tuberculosis
Graf et al. Predictive genomic biomarkers of hormonal therapy versus chemotherapy benefit in metastatic castration-resistant prostate cancer
Chen et al. A novel radiogenomics biomarker for predicting treatment response and pneumotoxicity from programmed cell death protein or ligand-1 inhibition immunotherapy in NSCLC
Bozack et al. DNA methylation age at birth and childhood: performance of epigenetic clocks and characteristics associated with epigenetic age acceleration in the Project Viva cohort
Dean et al. The association between telomere length and mortality in Bangladesh
Kamer et al. Predicting brain metastasis in early stage non-small cell lung cancer patients by gene expression profiling
Punnoose et al. BCL2 expression in first-line diffuse large B-cell lymphoma identifies a patient population with poor prognosis
Capoluongo et al. Alternative academic approaches for testing homologous recombination deficiency in ovarian cancer in the MITO16A/MaNGO-OV2 trial
Baio et al. Association between obesity and frequency of high‑grade prostate cancer on biopsy in men: A single‑center retrospective study
Chin et al. Germline genetic variation and predicting immune checkpoint inhibitor induced toxicity
Visvanathan et al. Evaluation of a liquid biopsy-breast cancer methylation (LBx-BCM) cartridge assay for predicting early disease progression and survival: TBCRC 005 prospective trial
Karim et al. Gene expression study of breast cancer using Welch Satterthwaite t-test, Kaplan-Meier estimator plot and Huber loss robust regression model
US11427873B2 (en) Methods and systems for assessing proliferative potential and resistance to immune checkpoint blockade
Somasegar et al. Germline mutations in Black patients with ovarian, fallopian tube and primary peritoneal carcinomas
Cirillo et al. Gestational biomarkers of daughter’s breast cancer in the Child Health and Development Studies

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