Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Immunother Cancer ; 12(7)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39089739

ABSTRACT

BACKGROUND: Immune-related adverse events (irAEs), characterized by targeted inflammation, occur in up to 60% of patients with melanoma treated with immune checkpoint inhibitors (ICIs). Evidence proved that the baseline peripheral blood profiles of patients at risk for severe irAEs development paralleled clinical autoimmunity. Interleukin (IL)-23 blockade with risankizumab is recommended for cases that are suffering from autoimmune disease, such as autoimmune colitis. However, currently, the role of IL-23 in irAEs onset and severity remains poorly understood. METHODS: The pro-inflammatory cytokines most associated with severe irAEs onset were identified by retrospective analysis based on GSE186143 data set. To investigate the efficacy of prophylactic IL-23 blockade administration to prevent irAEs, refer to a previous study, we constructed two irAEs murine models, including dextran sulfate sodium salt (DSS)-induced colitis murine model and a combined-ICIs-induced irAEs murine model. To further explore the applicability of our findings, murine models with graft-versus-host disease were established, in which Rag2-/-Il2rg-/- mice were transferred with human peripheral blood mononuclear cells and received combined cytotoxic T-lymphocyte associated antigen 4 (CTLA-4) and programmed cell death protein-1 (PD-1) treatment. Human melanoma cells were xenografted into these mice concomitantly. RESULTS: Here we show that IL-23 was upregulated in the serum of patients suffering from irAEs after dual anti-CTLA-4 and anti-PD-1 treatment, and increased as a function of irAEs severity. Additionally, Augmented CD4+ Tems may preferentially underlie irAEs onset. Treating mice with anti-mouse IL-23 antibody concomitantly with combined CTLA-4 and PD-1 immunotherapy ameliorates colitis and, in addition, preserves antitumor efficacy. Moreover, in xenografted murine models with irAEs, prophylactic blockade of human IL-23 using clinically available IL-23 inhibitor (risankizumab) ameliorated colitis, hepatitis and lung inflammation, and moreover, immunotherapeutic control of tumors was retained. Finally, we also provided a novel machine learning-based computational framework based on two blood-based features-IL-23 and CD4+ Tems-that may have predictive potential for severe irAEs and ICIs response. CONCLUSIONS: Our study not only provides clinically feasible strategies to dissociate efficacy and toxicity in the use of combined ICIs for cancer immunotherapy, but also develops a blood-based biomarker that makes it possible to achieve a straightforward and non-invasive, detection assay for early prediction of irAEs onset.


Subject(s)
CTLA-4 Antigen , Interleukin-23 , Animals , Mice , Humans , CTLA-4 Antigen/antagonists & inhibitors , Interleukin-23/antagonists & inhibitors , Interleukin-23/metabolism , Female , Immunotherapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Disease Models, Animal , Melanoma/drug therapy , Colitis/chemically induced , Colitis/drug therapy , Male , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Retrospective Studies
2.
FASEB J ; 37(11): e23236, 2023 11.
Article in English | MEDLINE | ID: mdl-37846808

ABSTRACT

Immune checkpoint inhibitors have effectively transformed the treatment of many cancers, particularly those highly devastating malignancies. With their widespread popularity, the drawbacks of immune checkpoint inhibitors are also recognized, such as drug resistance and immune-related systematic side effects. Thus, it never stops investigating novel immune checkpoint inhibitors. Lymphocyte Activation Gene-3 (LAG-3) is a well-established co-inhibitory receptor that performs negative regulation on immune responses. Recently, a novel FDA-approved LAG-3 blocking agent, together with nivolumab as a new combinational immunotherapy for metastatic melanoma, brought LAG-3 back into focus. Clinical data suggests that anti-LAG-3 agents can amplify the therapeutic response of other immune checkpoint inhibitors with manageable side effects. In this review, we elucidate the intercellular and intracellular mechanisms of LAG-3, clarify the current understanding of LAG-3 in the tumor microenvironment, identify present LAG-3-associated therapeutic agents, discuss current LAG-3-involving clinical trials, and eventually address future prospects for LAG-3 inhibitors.


Subject(s)
Immune Checkpoint Inhibitors , Melanoma , Humans , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/pathology , Nivolumab/therapeutic use , Immunotherapy , Programmed Cell Death 1 Receptor , Tumor Microenvironment
3.
Eur J Pharmacol ; 954: 175867, 2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37369297

ABSTRACT

The eukaryotic translation initiation factor 4 (EIF4) family is a major contributor to the recruitment of mRNAs to ribosomes during the initial translation stage in eukaryotes, whose dysregulation either allows for cancer transformation or prevents disordered cancerous cell growth. Circular RNAs (circRNAs), which exhibit distinctive structures and are widely expressed in eukaryotes, are anticipated to be clinical diagnostic biomarkers for cancer therapy. There is considerable evidence that EIF4s can influence the biogenesis, transport, and function of circRNAs and, in turn, circRNAs can control the expressions of EIF4s through certain molecular pathways. Herein, we primarily review the emerging studies of the EIF4 family and pinpoint the roles of dysregulated EIF4s in cancer. We also evaluate the patterns of intricate interactions between circRNAs and EIF4s and discuss the potential utility of circRNA-based therapeutics targeting EIF4s in clinical cancer research.


Subject(s)
Neoplasms , RNA, Circular , Humans , RNA, Circular/genetics , RNA/metabolism , RNA, Messenger , Neoplasms/genetics , Neoplasms/therapy , Peptide Initiation Factors , Biomarkers
4.
Front Immunol ; 13: 807840, 2022.
Article in English | MEDLINE | ID: mdl-35812443

ABSTRACT

Prostate cancer, recognized as a "cold" tumor, has an immunosuppressive microenvironment in which regulatory T cells (Tregs) usually play a major role. Therefore, identifying a prognostic signature of Tregs has promising benefits of improving survival of prostate cancer patients. However, the traditional methods of Treg quantification usually suffer from bias and variability. Transcriptional characteristics have recently been found to have a predictive power for the infiltration of Tregs. Thus, a novel machine learning-based computational framework has been presented using Tregs and 19 other immune cell types using 42 purified immune cell datasets from GEO to identify Treg-specific mRNAs, and a prognostic signature of Tregs (named "TILTregSig") consisting of five mRNAs (SOCS2, EGR1, RRM2, TPP1, and C11orf54) was developed and validated to monitor the prognosis of prostate cancer using the TCGA and ICGC datasets. The TILTregSig showed a stronger predictive power for tumor immunity compared with tumor mutation burden and glycolytic activity, which have been reported as immune predictors. Further analyses indicate that the TILTregSig might influence tumor immunity mainly by mediating tumor-infiltrating Tregs and could be a powerful predictor for Tregs in prostate cancer. Moreover, the TILTregSig showed a promising potential for predicting cancer immunotherapy (CIT) response in five CIT response datasets and therapeutic resistance in the GSCALite dataset in multiple cancers. Our TILTregSig derived from PBMCs makes it possible to achieve a straightforward, noninvasive, and inexpensive detection assay for prostate cancer compared with the current histopathological examination that requires invasive tissue puncture, which lays the foundation for the future development of a panel of different molecules in peripheral blood comprising a biomarker of prostate cancer.


Subject(s)
Prostatic Neoplasms , T-Lymphocytes, Regulatory , Drug Resistance, Neoplasm/genetics , Humans , Immunologic Factors/metabolism , Immunotherapy/methods , Male , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Tumor Microenvironment
5.
Mol Cancer ; 21(1): 87, 2022 03 26.
Article in English | MEDLINE | ID: mdl-35346215

ABSTRACT

FBXW7 (F-Box and WD Repeat Domain Containing 7) (also referred to as FBW7 or hCDC4) is a component of the Skp1-Cdc53 / Cullin-F-box-protein complex (SCF/ß-TrCP). As a member of the F-box protein family, FBXW7 serves a role in phosphorylation-dependent ubiquitination and proteasome degradation of oncoproteins that play critical role(s) in oncogenesis. FBXW7 affects many regulatory functions involved in cell survival, cell proliferation, tumor invasion, DNA damage repair, genomic instability and telomere biology. This thorough review of current literature details how FBXW7 expression and functions are regulated through multiple mechanisms and how that ultimately drives tumorigenesis in a wide array of cell types. The clinical significance of FBXW7 is highlighted by the fact that FBXW7 is frequently inactivated in human lung, colon, and hematopoietic cancers. The loss of FBXW7 can serve as an independent prognostic marker and is significantly correlated with the resistance of tumor cells to chemotherapeutic agents and poorer disease outcomes. Recent evidence shows that genetic mutation of FBXW7 differentially affects the degradation of specific cellular targets resulting in a distinct and specific pattern of activation/inactivation of cell signaling pathways. The clinical significance of FBXW7 mutations in the context of tumor development, progression, and resistance to therapies as well as opportunities for targeted therapies is discussed.


Subject(s)
F-Box-WD Repeat-Containing Protein 7 , Neoplasms , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Humans , Loss of Function Mutation , Neoplasms/genetics , Neoplasms/metabolism , Ubiquitination
6.
Onco Targets Ther ; 14: 5065-5083, 2021.
Article in English | MEDLINE | ID: mdl-34707365

ABSTRACT

OBJECTIVE: Although many curative treatments are being applied in the clinic, a significant number of patients with liver hepatocellular carcinoma (LIHC) suffer from drug resistance. The tumour microenvironment (TME) has been found to be closely associated with resistance, suggesting that identification of predictive biomarkers related to the TME for resistance in LIHC will be very rewarding. However, there has been no study dedicated to identifying a TME-related biomarker that has the potential to predict resistance in LIHC. METHODS: An integrated analysis was conducted based on data of patients with LIHC suffering from drug resistance from the TCGA database and four GEO datasets. Subsequently, we also validated the expression levels of the identified genes in paraffin-embedded LIHC samples by immunohistochemistry. RESULTS: In this study, we developed a robust and acute TME-related signature consisted of five immune-related genes (FABP6, CD4, PRF1, EREG and COLEC10) that could independently predict both the RFS and OS of LIHC patients. Moreover, the TME-related signature was significantly associated with the immune score, immune cytolytic activity (CYT), HLA, interferon (IFN) response and tumour-infiltrating lymphocytes (TILs), and it might influence tumour immunity mainly by affecting B cells, CD8+ T cells and dendritic cells. Furthermore, our analysis also indicated that the TME-related signature was correlated with the immunotherapy response and had an enormous potential to predict sorafenib resistance in LIHC. CONCLUSION: Our findings demonstrated a TME-related signature which can independently predict both the RFS and OS of LIHC patients, highlighting the predictive potential of the signature for immunotherapy response and sorafenib resistance, potentially enabling more precise and personalized sorafenib treatment in LIHC in the future.

7.
Pathol Oncol Res ; 27: 596899, 2021.
Article in English | MEDLINE | ID: mdl-34257547

ABSTRACT

Esophageal cancer (ESCA) is a leading cause of cancer-related mortality, with poor prognosis worldwide. DNA damage repair is one of the hallmarks of cancer. Loss of genomic integrity owing to inactivation of DNA repair genes can increase the risk of cancer progression and lead to poor prognosis. We aimed to identify a novel gene signature related to DNA repair to predict the prognosis of ESCA patients. Based on gene expression profiles of ESCA patients from The Cancer Genome Atlas and gene set enrichment analysis, 102 genes related to DNA repair were identified as candidates. After stepwise Cox regression analysis, we established a five-gene prognostic model comprising DGCR8, POM121, TAF9, UPF3B, and BCAP31. Kaplan-Meier survival analysis confirmed a strong correlation between the prognostic model and survival. Moreover, we verified the clinical value of the prognostic signature under the influence of different clinical parameters. We found that small-molecule drugs (trametinib, selumetinib, and refametinib) could help to improve patient survival. In summary, our study provides a novel and promising prognostic signature based on DNA-repair-related genes to predict survival of patients with ESCA. Systematic data mining provides a theoretical basis for further exploring the molecular pathogenesis of ESCA and identifying therapeutic targets.


Subject(s)
Biomarkers, Tumor/genetics , Computational Biology/methods , DNA Repair Enzymes/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic , Transcriptome , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Survival Rate
8.
Pathol Oncol Res ; 27: 600727, 2021.
Article in English | MEDLINE | ID: mdl-34257557

ABSTRACT

Background: Complex antigen processing and presentation processes are involved in the development and progression of breast cancer (BC). A single biomarker is unlikely to adequately reflect the complex interplay between immune cells and cancer; however, there have been few attempts to find a robust antigen processing and presentation-related signature to predict the survival outcome of BC patients with respect to tumor immunology. Therefore, we aimed to develop an accurate gene signature based on immune-related genes for prognosis prediction of BC. Methods: Information on BC patients was obtained from The Cancer Genome Atlas. Gene set enrichment analysis was used to confirm the gene set related to antigen processing and presentation that contributed to BC. Cox proportional regression, multivariate Cox regression, and stratified analysis were used to identify the prognostic power of the gene signature. Differentially expressed mRNAs between high- and low-risk groups were determined by KEGG analysis. Results: A three-gene signature comprising HSPA5 (heat shock protein family A member 5), PSME2 (proteasome activator subunit 2), and HLA-F (major histocompatibility complex, class I, F) was significantly associated with OS. HSPA5 and PSME2 were protective (hazard ratio (HR) < 1), and HLA-F was risky (HR > 1). Risk score, estrogen receptor (ER), progesterone receptor (PR) and PD-L1 were independent prognostic indicators. KIT and ACACB may have important roles in the mechanism by which the gene signature regulates prognosis of BC. Conclusion: The proposed three-gene signature is a promising biomarker for estimating survival outcomes in BC patients.


Subject(s)
Antigen Presentation , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Endoplasmic Reticulum Chaperone BiP/metabolism , Histocompatibility Antigens Class I/metabolism , Proteasome Endopeptidase Complex/metabolism , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Endoplasmic Reticulum Chaperone BiP/genetics , Female , Follow-Up Studies , Histocompatibility Antigens Class I/genetics , Humans , Middle Aged , Prognosis , Proteasome Endopeptidase Complex/genetics , Survival Rate , Transcriptome
9.
Front Cell Dev Biol ; 9: 622018, 2021.
Article in English | MEDLINE | ID: mdl-34150744

ABSTRACT

BACKGROUND: Lung adenocarcinoma (LUAD) is the most common diagnostic histologic subtype of non-small cell lung cancer, but the role of receptor-type tyrosine-protein phosphatase-like N (PTPRN) in LUAD has not been studied. METHODS: We conducted a bioinformatic analysis to identify the expression of PTPRN on LUAD data from the Cancer Genome Atlas (TCGA) and the relationship between PTPRN and overall survival of LUAD patients. The effects of PTPRN on the migration ability of LUAD cells and the underlying mechanisms were investigated by in vitro and in vivo assays (i.e., wound healing assay, transwell assay, western blotting, xenograft model, and immunohistochemistry). Gene-set enrichment analysis and computational resource were used to analyze the correlation between PTPRN and different tumor-infiltrating immune cells (TIICs). Lactate dehydrogenase assay and Enzyme-linked immunosorbent assay were conducted to examine natural killer (NK) cell cytotoxicity. RESULTS: In our study, we found that PTPRN was up-regulated in LUAD and related to metastasis of LUAD patients. Besides, PTPRN was correlated with poor prognosis in the TCGA-LUAD dataset. PTPRN overexpression promoted LUAD cell migration and the expression of EMT markers by influencing MEK/ERK and PI3K/AKT signaling. Moreover, PTPRN expression was significantly associated with TIICs, especially NK cells. A549 and H1299 cells overexpressed PTPRN inhibited NK cell cytotoxicity. CONCLUSION: Taken together, these findings demonstrated that PTPRN might be a potential and novel therapeutic target modulating antitumor immune response in treatment of LUAD.

10.
BMC Cancer ; 21(1): 645, 2021 May 31.
Article in English | MEDLINE | ID: mdl-34059009

ABSTRACT

BACKGROUND: Breast cancer (BRCA) is a malignant tumor with high morbidity and mortality, which is a threat to women's health worldwide. Ferroptosis is closely related to the occurrence and development of breast cancer. Here, we aimed to establish a ferroptosis-related prognostic gene signature for predicting patients' survival. METHODS: Gene expression profile and corresponding clinical information of patients from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database. The Least absolute shrinkage and selection operator (LASSO)-penalized Cox regression analysis model was utilized to construct a multigene signature. The Kaplan-Meier (K-M) and Receiver Operating Characteristic (ROC) curves were plotted to validate the predictive effect of the prognostic signature. Gene Ontology (GO) and Kyoto Encyclopedia of Genes, Genomes (KEGG) pathway and single-sample gene set enrichment analysis (ssGSEA) were performed for patients between the high-risk and low-risk groups divided by the median value of risk score. RESULTS: We constructed a prognostic signature consisted of nine ferroptosis-related genes (ALOX15, CISD1, CS, GCLC, GPX4, SLC7A11, EMC2, G6PD and ACSF2). The Kaplan-Meier curves validated the fine predictive accuracy of the prognostic signature (p < 0.001). The area under the curve (AUC) of the ROC curves manifested that the ferroptosis-related signature had moderate predictive power. GO and KEGG functional analysis revealed that immune-related responses were largely enriched, and immune cells, including activated dendritic cells (aDCs), dendritic cells (DCs), T-helper 1 (Th1), were higher in high-risk groups (p < 0.001). Oppositely, type I IFN response and type II IFN response were lower in high-risk groups (p < 0.001). CONCLUSION: Our study indicated that the ferroptosis-related prognostic signature gene could serve as a novel biomarker for predicting breast cancer patients' prognosis. Furthermore, we found that immunotherapy might play a vital role in therapeutic schedule based on the level and difference of immune-related cells and pathways in different risk groups for breast cancer patients.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/genetics , Breast Neoplasms/mortality , Ferroptosis/genetics , Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Breast/immunology , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Datasets as Topic , Female , Ferroptosis/drug effects , Ferroptosis/immunology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Kaplan-Meier Estimate , Middle Aged , Predictive Value of Tests , Prognosis , Protein Interaction Maps/drug effects , Protein Interaction Maps/genetics , ROC Curve , Risk Assessment/methods
11.
Front Oncol ; 11: 598017, 2021.
Article in English | MEDLINE | ID: mdl-33796449

ABSTRACT

Accumulating evidence has proven that N6-methyladenosine (m6A) RNA methylation plays an essential role in tumorigenesis. However, the significance of m6A RNA methylation modulators in the malignant progression of papillary renal cell carcinoma (PRCC) and their impact on prognosis has not been fully analyzed. The present research set out to explore the roles of 17 m6A RNA methylation regulators in tumor microenvironment (TME) of PRCC and identify the prognostic values of m6A RNA methylation regulators in patients afflicted by PRCC. We investigated the different expression patterns of the m6A RNA methylation regulators between PRCC tumor samples and normal tissues, and systematically explored the association of the expression patterns of these genes with TME cell-infiltrating characteristics. Additionally, we used LASSO regression to construct a risk signature based upon the m6A RNA methylation modulators. Two-gene prognostic risk model including IGF2BP3 and HNRNPC was constructed and could predict overall survival (OS) of PRCC patients from the Cancer Genome Atlas (TCGA) dataset. The prognostic signature-based risk score was identified as an independent prognostic indicator in Cox regression analysis. Moreover, we predicted the three most significant small molecule drugs that potentially inhibit PRCC. Taken together, our study revealed that m6A RNA methylation regulators might play a significant role in the initiation and progression of PRCC. The results might provide novel insight into exploration of m6A RNA modification in PRCC and provide essential guidance for therapeutic strategies.

12.
Brief Bioinform ; 22(4)2021 07 20.
Article in English | MEDLINE | ID: mdl-33212483

ABSTRACT

NLRP3 inflammasome was introduced as a double-edged sword in tumorigenesis and influenced immunotherapy response by modulating host immunity. However, a systematic assessment of the NLRP3-inflammasome-related genes across human cancers is lacking, and the predictive role of NLRP3 inflammasome in cancer immunotherapy (CIT) response remains unexplored. Thus, in this study, we performed a pan-cancer analysis of NLRP3-inflammasome-related genes across 24 human cancers. Out of these 24 cancers, 15 cancers had significantly different expression of NLRP3-inflammasome-related genes between normal and tumor samples. Meanwhile, Cox regression analysis showed that the NLRP3 inflammasome score could be served as an independent prognostic factor in skin cutaneous melanoma. Further analysis indicated that NLRP3 inflammasome may influence tumor immunity mainly by mediating tumor-infiltrating lymphocytes and macrophages, and the effect of NLRP3 inflammasome on immunity is diverse across tumor types in tumor microenvironment. We also found that the NLRP3 inflammasome score could be a stronger predictor for immune signatures compared with tumor mutation burden (TMB) and glycolytic activity, which have been reported as immune predictors. Furthermore, analysis of the association between NLRP3 inflammasome and CIT response using six CIT response datasets revealed the predictive value of NLRP3 inflammasome for immunotherapy response of patients in diverse cancers. Our study illustrates the characterization of NLRP3 inflammasome in multiple cancer types and highlights its potential value as a predictive biomarker of CIT response, which can pave the way for further investigation of the prognostic and therapeutic potentials of NLRP3 inflammasome.


Subject(s)
Databases, Factual , Immunotherapy , Inflammasomes/immunology , Melanoma , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neoplasm Proteins/immunology , Skin Neoplasms , Tumor Microenvironment/immunology , Disease-Free Survival , Humans , Melanoma/immunology , Melanoma/mortality , Melanoma/therapy , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/therapy , Survival Rate , Melanoma, Cutaneous Malignant
13.
Int Immunopharmacol ; 89(Pt A): 107162, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33168410

ABSTRACT

Skin cutaneous melanoma (SKCM) is the most invasive form of skin cancer with poor prognosis. Growing evidence has demonstrated that tumor immune microenvironment plays a key contributing role in tumorigenesis and predicting clinical outcomes. The aim of this study was to recognize immune classification and a reliable prognostic signature for patients with SKCM. By using single-sample gene set enrichment (ssGSEA) and hierarchical clustering analyses, we evaluated the immune infiltration landscape of SKCM afflicted patients from The Cancer Genome Atlas (TCGA) dataset and named two SKCM subtypes: Immunity-high and Immunity-low. The Immunity-high subgroup was characterized by up-regulation of immune response and favorable survival probability. Seven candidate small molecule drugs which potentially reverse SKCM immune status were identified by using Connectivity map (CMap) database. A prognostic five-immune-associated gene (IAG) signature consisting IFITM1, TNFSF13B, APOBEC3G, CCL8 and KLRK1 with high predictive value was established using the LASSO Cox regression analysis in training set, and validated in testing set as well as Gene Expression Omnibus (GEO) external validation cohort (P < 0.05). Lower tumor purity and active immune-related signaling pathways were obviously presented in low-risk group. Furthermore, a novel composite nomogram based upon the five-IAG signature and other clinical parameters was built with excellent calibration curves. Collectively, comprehensively characterizing the SKCM subtypes based upon immune microenvironment landscape and development of a survival-related IAG signature may provide a promising avenue for improving individualized treatment design and prognosis prediction for patients with SKCM.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/immunology , Melanoma/metabolism , Skin Neoplasms/metabolism , Antineoplastic Agents/chemistry , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Melanoma/drug therapy , Melanoma/immunology , Middle Aged , Principal Component Analysis , Prognosis , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Small Molecule Libraries , Tumor Microenvironment/immunology , Melanoma, Cutaneous Malignant
14.
Cancer Med ; 9(21): 8186-8201, 2020 11.
Article in English | MEDLINE | ID: mdl-32902917

ABSTRACT

BACKGROUND: The tumor microenvironment (TME) plays a critical role in tumorigenesis, development, and therapeutic efficacy. Major advances have been achieved in the treatment of various cancers through immunotherapy. Nevertheless, only a minority of patients have positive responses to immunotherapy, which is partly due to conditions of the immunosuppressive microenvironment. Therefore, it is essential to identify prognostic biomarkers that reflect heterogeneous landscapes of the TME. METHODS AND MATERIALS: Based upon the ESTIMATE algorithm, we evaluated the infiltrating levels of immune and stromal components derived from patients afflicted by various types of cancer from The Cancer Genome Atlas database (TCGA). According to respective patient immune and stromal scores, we categorized cases into high- and low-scoring subgroups for each cancer type to explore associations between TME and patient prognosis. Gene Set Enrichment Analyses (GSEA) were conducted and genes enriched in IFNγ response signaling pathway were selected to facilitate establishment of a risk model for predicting overall survival (OS). Furthermore, we investigated the associations between the prognostic signature and tumor immune infiltration landscape by using CIBERSORT algorithm and TIMER database. RESULTS: Among the cancers assessed, the immune scores for skin cutaneous melanoma (SKCM) were the most significantly correlated with patients' survival time (P < .0001). We identified and validated a five-IFNγ response-related gene signature (UBE2L6, PARP14, IFIH1, IRF2, and GBP4), which was closely correlated with the prognosis for SKCM afflicted patients. Multivariate Cox regression analysis indicated that this risk model was an independent prognostic factor for SKCM. Tumor-infiltrating lymphocytes and specific immune checkpoint molecules had notably differential levels of expression in high- compared to low-risk samples. CONCLUSION: In this study, we established a novel five-IFNγ response-related gene signature that provided a better and increasingly comprehensive understanding of tumor immune landscape, and which demonstrated good performance in predicting outcomes for patients afflicted by SKCM.


Subject(s)
Interferon-gamma/metabolism , Melanoma/genetics , Melanoma/immunology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Tumor Microenvironment/immunology , Algorithms , Antigens, CD/metabolism , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/metabolism , CTLA-4 Antigen/metabolism , Databases, Genetic , Female , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Immune Checkpoint Inhibitors/therapeutic use , Interferon Regulatory Factor-2/genetics , Interferon-Induced Helicase, IFIH1/genetics , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating , Male , Melanoma/drug therapy , Melanoma/mortality , Middle Aged , Poly(ADP-ribose) Polymerases/genetics , Prognosis , Proportional Hazards Models , Risk Assessment/methods , Signal Transduction/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/mortality , Survival Rate , Transcriptome , Ubiquitin-Conjugating Enzymes/genetics , Lymphocyte Activation Gene 3 Protein
15.
J Cell Mol Med ; 24(11): 6283-6297, 2020 06.
Article in English | MEDLINE | ID: mdl-32306508

ABSTRACT

High mortality of patients with cervical cancer (CC) stresses the imperative of prognostic biomarkers for CC patients. Additionally, the vital status of post-translational modifications (PTMs) in the progression of cancers has been reported by numerous researches. Therefore, the purpose of this research was to dig a prognostic signature correlated with PTMs for CC. We built a five-mRNA (GALNTL6, ARSE, DPAGT1, GANAB and FURIN) prognostic signature associated with PTMs to predict both disease-free survival (DFS) (hazard ratio [HR] = 3.967, 95% CI = 1.985-7.927; P < .001) and overall survival (HR = 2.092, 95% CI = 1.138-3.847; P = .018) for CC using data from The Cancer Genome Atlas database. Then, the robustness of the signature was validated using GSE44001 and the Human Protein Atlas (HPA) database. CIBERSORT algorithm analysis displayed that activated CD4 memory T cell was also an independent indicator for DFS (HR = 0.426, 95% CI = 0.186-0.978; P = .044) which could add additional prognostic value to the signature. Collectively, the PTM-related signature and activated CD4 memory T cell can provide new avenues for the prognostic predication of CC. These findings give further insights into effective treatment strategies for CC, providing opportunities for further experimental and clinical validations.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neoplasm Recurrence, Local/genetics , Protein Processing, Post-Translational/genetics , Uterine Cervical Neoplasms/genetics , CD4-Positive T-Lymphocytes/immunology , Databases, Genetic , Female , Humans , Immunologic Memory , Kaplan-Meier Estimate , Lymphocyte Activation/immunology , Middle Aged , Molecular Sequence Annotation , Prognosis , Progression-Free Survival , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology
16.
ScientificWorldJournal ; 2014: 746260, 2014.
Article in English | MEDLINE | ID: mdl-24971389

ABSTRACT

A hybrid evolutionary algorithm using scalable encoding method for path planning is proposed in this paper. The scalable representation is based on binary tree structure encoding. To solve the problem of hybrid genetic algorithm and particle swarm optimization, the "dummy node" is added into the binary trees to deal with the different lengths of representations. The experimental results show that the proposed hybrid method demonstrates using fewer turning points than traditional evolutionary algorithms to generate shorter collision-free paths for mobile robot navigation.


Subject(s)
Algorithms , Robotics/methods
SELECTION OF CITATIONS
SEARCH DETAIL