Identification of Cuproptosis-Related Subtypes, Establishment of a Prognostic Signature and Characterization of the Tumor Microenvironment in Gastric Cancer

Purpose Cuproptosis is a newly identified form of programmed cell death. We aimed to comprehensively discuss the correlation of cuproptosis with gastric cancer (GC) using bioinformatic methods. Patients and Methods This study selected GC bulk and single-cell RNA sequencing profiles from public databases. Based on the enrichment pattern of cuproptosis-related gene sets (CRGSs), GC patients were classified into different cuproptosis subtypes. A series of systematic analyses was performed to investigate the correlation of cuproptosis subtype with biological function and immune cell infiltration. In addition, we established a CRGS risk score signature to quantify GC patients’ risk level, and analyzed the signature’s relationship with clinical features, tumor microenvironment (TME) and treatment responses. Genes used for the construction of the risk score model were also detected in GC tumor and normal tissues by real-time quantitative polymerase chain reaction (RT-qPCR). Results First, analysis of scRNA-seq data revealed the alterations in CRGS enrichment scores for patients with GC and precancerous diseases. Then, based on large GC patient cohorts, two cuproptosis subtypes with significant differences in survival, biological function and TME were identified. Furthermore, we established a CRGS risk score signature. High-risk patients on the CRGS risk score signature with worse overall survival were characterized by higher immune and stromal contents in the TME, more advanced clinicopathological features, and better sensitivity to a wider range of anti-tumor drugs. Low-risk patients were correlated with higher tumor purity, and demonstrated more favorable clinical outcomes and higher sensitivity to immunotherapy. Conclusion The current work elucidated that cuproptosis plays an important role in the regulation of TME landscapes in GC. Two cuproptosis subtypes with distinct TME characteristics were identified. In addition, the establishment of a CRGS risk score signature could provide novel insights into accurate prediction and personalized treatment for GC patients.

[1]  G. Zheng,et al.  Cuproptosis-Mediated Patterns Characterized by Distinct Tumor Microenvironment and Predicted the Immunotherapy Response for Gastric Cancer , 2023, ACS omega.

[2]  Mengna Zhang,et al.  Comprehensive analysis of cuproptosis-related genes in prognosis, tumor microenvironment infiltration, and immunotherapy response in gastric cancer , 2022, Journal of Cancer Research and Clinical Oncology.

[3]  S. Zhong,et al.  Identification of cuproptosis-related subtypes, construction of a prognosis model, and tumor microenvironment landscape in gastric cancer , 2022, Frontiers in Immunology.

[4]  Ronghua Yang,et al.  Construction and Validation of a Novel Pyroptosis-Related Four-lncRNA Prognostic Signature Related to Gastric Cancer and Immune Infiltration , 2022, Frontiers in immunology.

[5]  T. Golub,et al.  Copper induces cell death by targeting lipoylated TCA cycle proteins , 2022, Science.

[6]  Zhengrong Li,et al.  A Necroptosis-Related lncRNA-Based Signature to Predict Prognosis and Probe Molecular Characteristics of Stomach Adenocarcinoma , 2022, Frontiers in Genetics.

[7]  Junyu Huo,et al.  A Five-Gene Signature Associated With DNA Damage Repair Molecular Subtype Predict Overall Survival for Hepatocellular Carcinoma , 2022, Frontiers in Genetics.

[8]  Tong Liu,et al.  A novel ferroptosis-related lncRNA signature for prognosis prediction in gastric cancer , 2021, BMC Cancer.

[9]  Wei Song,et al.  Identification of stem cell-related subtypes and risk scoring for gastric cancer based on stem genomic profiling , 2021, Stem cell research & therapy.

[10]  Jun Ren,et al.  Pyroptosis Patterns Characterized by Distinct Tumor Microenvironment Infiltration Landscapes in Gastric Cancer , 2021, Genes.

[11]  X. Fang,et al.  Construction on of a Ferroptosis-Related lncRNA-Based Model to Improve the Prognostic Evaluation of Gastric Cancer Patients Based on Bioinformatics , 2021, Frontiers in Genetics.

[12]  A. Jemal,et al.  Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries , 2021, CA: a cancer journal for clinicians.

[13]  V. Seshan,et al.  The association between tumor mutational burden and prognosis is dependent on treatment context , 2020, Nature genetics.

[14]  Ashton C. Berger,et al.  Interplay of somatic alterations and immune infiltration modulates response to PD-1 blockade in advanced clear cell renal cell carcinoma , 2020, Nature Medicine.

[15]  D. Vaux,et al.  Cell Death in the Origin and Treatment of Cancer. , 2020, Molecular cell.

[16]  J. Califano,et al.  B Cells Improve Overall Survival in HPV-Associated Squamous Cell Carcinomas and Are Activated by Radiation and PD-1 Blockade , 2020, Clinical Cancer Research.

[17]  Mengyu Sun,et al.  Identification and validation of an individualized autophagy-clinical prognostic index in gastric cancer patients , 2020, Cancer Cell International.

[18]  Jeffrey E. Lee,et al.  B cells and tertiary lymphoid structures promote immunotherapy response , 2020, Nature.

[19]  D. Cao,et al.  High tumor mutation burden predicts better efficacy of immunotherapy: a pooled analysis of 103078 cancer patients , 2019, Oncoimmunology.

[20]  Peng Zhang,et al.  Dissecting the Single-Cell Transcriptome Network Underlying Gastric Premalignant Lesions and Early Gastric Cancer. , 2019, Cell reports.

[21]  R. DePinho,et al.  KRAS-IRF2 Axis Drives Immune Suppression and Immune Therapy Resistance in Colorectal Cancer. , 2019, Cancer cell.

[22]  C. Brennan,et al.  Tumor mutational load predicts survival after immunotherapy across multiple cancer types , 2019, Nature Genetics.

[23]  N. Chaput,et al.  Immunotherapy in advanced gastric cancer, is it the future? , 2019, Critical reviews in oncology/hematology.

[24]  J. Utikal,et al.  Circulating and Tumor Myeloid‐derived Suppressor Cells in Resectable Non‐Small Cell Lung Cancer , 2018, American journal of respiratory and critical care medicine.

[25]  Xia Li,et al.  TIP: A Web Server for Resolving Tumor Immunophenotype Profiling. , 2018, Cancer research.

[26]  X. Liu,et al.  Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response , 2018, Nature Medicine.

[27]  Isabelle Salmon,et al.  Methods of measurement for tumor mutational burden in tumor tissue. , 2018, Translational lung cancer research.

[28]  R. Bourgon,et al.  TGF-β attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells , 2018, Nature.

[29]  F. Kühnel,et al.  CD4 and CD8 T lymphocyte interplay in controlling tumor growth , 2017, Cellular and Molecular Life Sciences.

[30]  C. Slingluff,et al.  Vaccines targeting helper T cells for cancer immunotherapy. , 2017, Current opinion in immunology.

[31]  J. Wargo,et al.  Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy , 2017, Cell.

[32]  Pornpimol Charoentong,et al.  Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade , 2016, bioRxiv.

[33]  H. Stunnenberg,et al.  Transcriptional Landscape of Human Tissue Lymphocytes Unveils Uniqueness of Tumor-Infiltrating T Regulatory Cells , 2016, Immunity.

[34]  G. Gordon,et al.  Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: a multicentre, open-label, phase 2 study. , 2016, The Lancet. Oncology.

[35]  L. Zitvogel,et al.  Targeting the tumor microenvironment: removing obstruction to anticancer immune responses and immunotherapy. , 2016, Annals of oncology : official journal of the European Society for Medical Oncology.

[36]  T. Kipps,et al.  Targeting BCL2 with Venetoclax in Relapsed Chronic Lymphocytic Leukemia. , 2016, The New England journal of medicine.

[37]  Jason G. Jin,et al.  Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes , 2015, Nature Medicine.

[38]  Steven J. M. Jones,et al.  Comprehensive molecular characterization of gastric adenocarcinoma , 2014, Nature.

[39]  G. Getz,et al.  Inferring tumour purity and stromal and immune cell admixture from expression data , 2013, Nature Communications.

[40]  Bin Tean Teh,et al.  Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil. , 2013, Gastroenterology.

[41]  Hao Xiong,et al.  Substantial susceptibility of chronic lymphocytic leukemia to BCL2 inhibition: results of a phase I study of navitoclax in patients with relapsed or refractory disease. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[42]  D. Hanahan,et al.  Hallmarks of Cancer: The Next Generation , 2011, Cell.

[43]  W. Wilson,et al.  Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. , 2010, The Lancet. Oncology.

[44]  Stephen B Fox,et al.  Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[45]  E. Elkin,et al.  Decision Curve Analysis: A Novel Method for Evaluating Prediction Models , 2006, Medical decision making : an international journal of the Society for Medical Decision Making.

[46]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[47]  A. Wyllie,et al.  Apoptosis: A Basic Biological Phenomenon with Wide-ranging Implications in Tissue Kinetics , 1972, British Journal of Cancer.