中文English
ISSN 1001-5256 (Print)
ISSN 2097-3497 (Online)
CN 22-1108/R
Volume 40 Issue 12
Dec.  2024
Turn off MathJax
Article Contents

Establishment of a risk model based on immunogenic cell death-related genes and its value in predicting the prognosis and tumor microenvironment characteristics of hepatocellular carcinoma

DOI: 10.12449/JCH241218
Research funding:

Key Projects of Jiangxi Provincial Department of Education (GJJ210135);

Jiangxi Provincial Department of Education (GJJ200147);

Jiangxi Traditional Chinese Medicine Science and Technology Plan Project (2022B966)

More Information
  • Corresponding author: LI Huiming, hmingl2321@163.com (ORCID: 0000-0001-8297-2724)
  • Received Date: 2024-04-23
  • Accepted Date: 2024-07-10
  • Published Date: 2024-12-25
  •   Objective  To identify immunogenic cell death (ICD)-related genes in hepatocellular carcinoma (HCC), and to establish a scoring model based on these genes for predicting the prognosis and tumor microenvironment characteristics of HCC.  Methods  The Cancer Genome Atlas database was used to obtain HCC datasets, and heatmaps were used to display the expression of 57 ICD-related genes in HCC. A cluster analysis was conducted based on the expression of ICD-related genes, and two ICD subtypes (low and high ICD expression groups) were analyzed in terms of gene ontology enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, somatic mutation, and immune cell infiltration. The LASSO Cox regression risk model was constructed to evaluate its clinical application value, and a nomogram model was established to predict the 1-, 3-, and 5-year survival rates of patients. In addition, qRT-PCR was used to validate the expression levels of key genes in the model. The independent-samples t test was used for comparison between two groups, and the univariate and multivariate Cox regression analyses were used to determine prognostic factors among clinicopathological features. The Kaplan-Meier survival curve was used for prognostic analysis, and the Spearman rank correlation test was used for correlation analysis.  Results  The low ICD expression group had a poorer prognosis, while the high ICD expression group had relatively favorable clinical outcomes (P=0.004). Further analysis showed that the high ICD expression group was associated with an immune-active microenvironment, and the genes were mainly enriched in immune-related pathways such as immunoglobulin receptor binding, hematopoietic cell lineage, and B cell receptor. The results of somatic mutation analysis showed that the high ICD expression group had higher expression levels of CD274, CTLA4, HAVCR2, TIGIT, PDCD1, and PDCD1LG2 (all P<0.05). A risk prediction model was established using 8 ICD-related genes, i.e., HSP90AA1, ATG5, BAX, PPIA, HSPA4, TLR2, TREM1, and LY96, and this model showed a good predictive value across different clinical characteristics. The univariate and multivariate Cox regression analyses showed that age and risk score were independent prognostic factors for overall survival in the training set (both P<0.05). The results of qRT-PCR showed that the relative expression levels of HSPA4 and REM1 in HCC tumor samples were significantly higher than those in adjacent tissue samples (both P<0.001). For the patients with an increase in ICD risk score, the ICD risk score was negatively correlated with γδT cells (r=-0.29, P<0.05), plasma cells (r=-0.3, P<0.05), and CD8+T lymphocytes (r=-0.37, P<0.05) and was positively correlated with memory B cells (r=0.38, P<0.05), resting dendritic cells (r=0.47, P<0.05), and M0 macrophages (r=0.49, P<0.05).  Conclusion  This study identifies the ICD-related genes that are associated with the prognosis of HCC, which provides insights into the immune characteristics of different ICD expression profiles. The risk model and the nomogram model established in this study have a significant value for predicting the prognosis of HCC patients and guiding immunotherapy for HCC patients.

     

  • loading
  • [1]
    VOGEL A, MEYER T, SAPISOCHIN G, et al. Hepatocellular carcinoma[J]. Lancet, 2022, 400: 1345- 1362. DOI: 10.1016/S0140-6736(22)01200-4.
    [2]
    GANESAN P, KULIK LM. Hepatocellular carcinoma: New developments[J]. Clin Liver Dis, 2023, 27( 1): 85- 102. DOI: 10.1016/j.cld.2022.08.004.
    [3]
    LLOVET JM, KELLEY RK, VILLANUEVA A, et al. Hepatocellular carcinoma[J]. Nat Rev Dis Primers, 2021, 7( 1): 6. DOI: 10.1038/s41572-020-00240-3.
    [4]
    ZHANG YY, ZHANG ZM. The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications[J]. Cell Mol Immunol, 2020, 17( 8): 807- 821. DOI: 10.1038/s41423-020-0488-6.
    [5]
    NAIMI A, MOHAMMED RN, RAJI A, et al. Tumor immunotherapies by immune checkpoint inhibitors(ICIs); the pros and cons[J]. Cell Commun Signal, 2022, 20( 1): 44. DOI: 10.1186/s12964-022-00854-y.
    [6]
    KENNEDY LB, SALAMA AKS. A review of cancer immunotherapy toxicity[J]. CA Cancer J Clin, 2020, 70( 2): 86- 104. DOI: 10.3322/caac.21596.
    [7]
    LLOVET JM, CASTET F, HEIKENWALDER M, et al. Immunotherapies for hepatocellular carcinoma[J]. Nat Rev Clin Oncol, 2022, 19( 3): 151- 172. DOI: 10.1038/s41571-021-00573-2.
    [8]
    SANGRO B, SAROBE P, HERVÁS-STUBBS S, et al. Advances in immunotherapy for hepatocellular carcinoma[J]. Nat Rev Gastroenterol Hepatol, 2021, 18( 8): 525- 543. DOI: 10.1038/s41575-021-00438-0.
    [9]
    NIU X, CHEN LJ, LI Y, et al. Ferroptosis, necroptosis, and pyroptosis in the tumor microenvironment: Perspectives for immunotherapy of SCLC[J]. Semin Cancer Biol, 2022, 86( Pt 3): 273- 285. DOI: 10.1016/j.semcancer.2022.03.009.
    [10]
    CAI JY, HU YY, YE Z, et al. Immunogenic cell death-related risk signature predicts prognosis and characterizes the tumour microenvironment in lower-grade glioma[J]. Front Immunol, 2022, 13: 1011757. DOI: 10.3389/fimmu.2022.1011757.
    [11]
    KROEMER G, GALASSI C, ZITVOGEL L, et al. Immunogenic cell stress and death[J]. Nat Immunol, 2022, 23( 4): 487- 500. DOI: 10.1038/s41590-022-01132-2.
    [12]
    AHMED A, TAIT SWG. Targeting immunogenic cell death in cancer[J]. Mol Oncol, 2020, 14( 12): 2994- 3006. DOI: 10.1002/1878-0261.12851.
    [13]
    FUCIKOVA J, KEPP O, KASIKOVA L, et al. Detection of immunogenic cell death and its relevance for cancer therapy[J]. Cell Death Dis, 2020, 11( 11): 1013. DOI: 10.1038/s41419-020-03221-2.
    [14]
    ZHOU JY, WANG GY, CHEN YZ, et al. Immunogenic cell death in cancer therapy: Present and emerging inducers[J]. J Cell Mol Med, 2019, 23( 8): 4854- 4865. DOI: 10.1111/jcmm.14356.
    [15]
    CHIARAVALLI M, SPRING A, AGOSTINI A, et al. Immunogenic cell death: An emerging target in gastrointestinal cancers[J]. Cells, 2022, 11( 19): 3033. DOI: 10.3390/cells11193033.
    [16]
    GALLUZZI L, KEPP O, HETT E, et al. Immunogenic cell death in cancer: Concept and therapeutic implications[J]. J Transl Med, 2023, 21( 1): 162. DOI: 10.1186/s12967-023-04017-6.
    [17]
    WANG XW, WU SW, LIU F, et al. An immunogenic cell death-related classification predicts prognosis and response to immunotherapy in head and neck squamous cell carcinoma[J]. Front Immunol, 2021, 12: 781466. DOI: 10.3389/fimmu.2021.781466.
    [18]
    GARG AD, DE RUYSSCHER D, AGOSTINIS P. Immunological metagene signatures derived from immunogenic cancer cell death associate with improved survival of patients with lung, breast or ovarian malignancies: A large-scale meta-analysis[J]. Oncoimmunology, 2015, 5( 2): e1069938. DOI: 10.1080/2162402X.2015.1069938.
    [19]
    BROWN ZJ, TSILIMIGRAS DI, RUFF SM, et al. Management of hepatocellular carcinoma: A review[J]. JAMA Surg, 2023, 158( 4): 410- 420. DOI: 10.1001/jamasurg.2022.7989.
    [20]
    TRICOLI L, NITURE S, CHIMEH U, et al. Role of microRNAs in the development of hepatocellular carcinoma and drug resistance[J]. Front Biosci(Landmark Ed), 2019, 24( 2): 382- 391. DOI: 10.2741/4724.
    [21]
    CEBALLOS MP, QUIROGA AD, PALMA NF. Role of sirtuins in hepatocellular carcinoma progression and multidrug resistance: Mechanistical and pharmacological perspectives[J]. Biochem Pharmacol, 2023, 212: 115573. DOI: 10.1016/j.bcp.2023.115573.
    [22]
    DEVAN AR, KUMAR AR, NAIR B, et al. Insights into an immunotherapeutic approach to combat multidrug resistance in hepatocellular carcinoma[J]. Pharmaceuticals(Basel), 2021, 14( 7): 656. DOI: 10.3390/ph14070656.
    [23]
    LI ZL, LAI XQ, FU SQ, et al. Immunogenic cell death activates the tumor immune microenvironment to boost the immunotherapy efficiency[J]. Adv Sci(Weinh), 2022, 9( 22): e2201734. DOI: 10.1002/advs.202201734.
    [24]
    ZHANG SW, WANG J, KONG ZQ, et al. Emerging photodynamic nanotherapeutics for inducing immunogenic cell death and potentiating cancer immunotherapy[J]. Biomaterials, 2022, 282: 121433. DOI: 10.1016/j.biomaterials.2022.121433.
    [25]
    LIU GH, QIU YM, ZHANG P, et al. Immunogenic cell death enhances immunotherapy of diffuse intrinsic pontine glioma: From preclinical to clinical studies[J]. Pharmaceutics, 2022, 14( 9): 1762. DOI: 10.3390/pharmaceutics14091762.
    [26]
    SANSONE C, BRUNO A, PISCITELLI C, et al. Natural compounds of marine origin as inducers of immunogenic cell death(ICD): Potential role for cancer interception and therapy[J]. Cells, 2021, 10( 2): 231. DOI: 10.3390/cells10020231.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(8)  / Tables(2)

    Article Metrics

    Article views (81) PDF downloads(13) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return