Open Access

Immune system‑associated genes increase malignant progression and can be used to predict clinical outcome in patients with hepatocellular carcinoma

  • Authors:
    • Rongfu Huang
    • Zheng Chen
    • Wenli Li
    • Chunmei Fan
    • Jun Liu
  • View Affiliations

  • Published online on: February 24, 2020     https://doi.org/10.3892/ijo.2020.4998
  • Pages: 1199-1211
  • Copyright: © Huang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is one of the most malignant types of cancer, and is associated with high recurrence rates and a poor response to chemotherapy. Immune signatures in the microenvironment of HCC have not been well explored systematically. The aim of the present study was to identify prognostic immune signatures and build a nomogram for use in clinical evaluation. Using bioinformatics analysis, RNA‑seq data and overall survival (OS) information on 370 HCC cases from TCGA and 232 HCC cases from ICGC were analyzed. The differential expression of select immune genes, based on previously published studies, between HCC and adjacent tissue were analyzed using the limma package in R. Enrichment of pathways and gene ontology analysis was performed using clusterProfiler. Subsequently, univariate Cox regression analysis, Lasso penalty linear regression and multivariate Cox regression models were used to construct a model for immune risk score (IRS). The R packages, survival and survivalROC, were used to plot survival and the associated receiver operating characteristic curves. Infiltration of immune cells was calculated using Tumor IMmune Estimation Resource, with significance examined using a Pearson's correlation test. P<0.05 was considered significant. Based on the analysis, expression of 200 immune genes were upregulated and 47 immune genes were downregulated immune genes. In the multivariate Cox model, 5 genes (enhancer of zest homology 2, ferritin light chain, complement factor H related 3, isthmin 2, cyclin dependent kinase 5) were used to generate the IRS. By stratifying according to the median IRS, it was shown that patients with a high IRS had poor OS rates after 1, 2, 3 and 5 years, and this result was consistent across the testing, training and independent validation cohorts. Additionally, the IRS was correlated with the abundance of infiltrating immune cells. The nomogram built using IRS and clinical characteristics, was able to predict 1, 3 and 5 year OS with area under the curve values of >0.8. These results suggest that the model developed to calculate the IRS may be used to monitor the effectiveness of treatment strategies and for prognostic prediction.

Introduction

Hepatocellular carcinoma (HCC) has become the second leading cause of cancer-associated death, which affects patients worldwide, and is associated with early recurrence and a poor response to chemotherapy (1,2). As our understanding of the roles of immune checkpoints in tumor cells and the surrounding non-tumor cells in the cancer microenvironment has advanced, novel technologies, such as Chimeric antigen receptor T-cell therapy and PD-1/PD-L1 checkpoint inhibition therapy, have been developed to target the immune environment of HCC to improve the prognosis of patients following HCC resection (3-6). However, the overall responses rates of patients treated with specific checkpoint blockers in HCC, such as those targeting PD-1 or CTLA-4, are not favorable, possibly due to unknown changes to the immune microenvironment (7).

The emergence of high-throughput nucleotide sequencing analysis provides new perspectives to understand the genomic changes in tumors, revealing the differentially expressed genetic signatures between tumor tissues and normal tissues (8,9). Several studies in different types of cancer, including breast cancer, thyroid cancer, non-squamous non-small cell lung cancer and colorectal carcinoma, have examined differences in the patterns of immune signatures to improve our understanding of the immune environment and the mechanisms underlying tumor development and progression (10-14). However, the specific immune genetic changes in HCC have not been extensively studied, although one study found that the levels of immune cells infiltration, specifically by T-cells, cytotoxic cells, Th2 cells and macrophages, in HCC were associated with improved survival in patients based on n silico analysis, suggesting that the type of immune cells present in HCC tissues were different from the immune cell profile of the normal liver (15). As the liver is now considered to a 'immune associated organ', the presence of immune cells in HCC should be taken into consideration as a leading factor for predicting prognosis following resection, and should not be restricted to specific types of immune cells (16-18).

In the present study, the changes in expression of immune related genes in HCC tissues were compared with the adjacent healthy matching tissues, using bioinformatics analysis. The immune-associated genes identified was derived from a comprehensive study of the immune landscape of 20 solid tumors, which allowed for evaluation of relevant immune functions and the immune status of solid tumors in a simplified manner (19,20). The aim of the present study was to identify the immune-related genetic changes between HCC tissues and normal liver tissues, to understand the effects of immune regulation of HCC, and the effect on progression of HCC. Additionally, an immune evaluating model for prognostic evaluation in HCC patients was constructed, with the aim of differentiating patients into sub-populations for more personalized clinical treatment to maximize the efficacy of therapies used, particularly for treatment with immune checkpoint inhibition.

Materials and methods

Datasets

Data on patients with HCC were obtained from The Cancer Genome Atlas (TCGA; cancer.gov/tcga) and ICGC (icgc.org), which are publicly available databases (21,22). The databases contained information on 370 (TCGA) and 232 cases (ICGC) of HCC, which included RNA sequencing information and the clinical characteristics (Table I). In the data obtained from TCGA, there were 249 men and 121 women with a median age of 61 (range, 16-85). In the ICGC dataset, there were 171 men and 61 women with a median age of 69 (range, 31-89). The list of immune-related genes for analysis was obtained from previous studies (19,20) which contained a total of 821 immune related genes.

Table I

Clinicopathological characteristics of patients in TCGA and ICGC.

Table I

Clinicopathological characteristics of patients in TCGA and ICGC.

TCGA. n=370
Clinical characteristicsn%
Survival status
 Survived2446555
 Died12634.05
Age
 ≤65 years23262.70
 >65 years13837.30
Sex
 Male24967.30
 Female12132.70
Histological grade
 G15514.86
 G217747.84
 G312132.70
 G4123.24
Stage
 I17146.22
 II8522.57
 III8522.57
 IV51.35
T classification
 T11814852
 T29325.14
 T38021.62
 T4133.51
 TX10.27
M classification
 M026671.89
 Ml41.08
 MX10027.03
N classification
 N025268.11
 NI41.08
 NX11330.54
ICGC, n=232
Clinical characteristicsn%
Survival status
 Survived18981.47
 Died4318.53
Age
 ≤65 years9038.79
 >65 years14261.21
Sex
 Male17173.71
 Female6126.29
Clinical characteristicsn%
Stage
 I3615.52
 II10645.69
 III7130.60
 IV198.19
Prior malignancy
 No20287.07
 Yes3012.93

[i] TCGA, The Cancer Genome Atlas; ICGC, International Cancer Gene Consortium.

Differential expression analysis

DEIGs between adjacent and HCC tissues were analyzed using the limma package on the cohort from TCGA (23). The raw data were normalized and log2(x+1) transformed. Genes with a fold change >1 and an adjusted P-value <0.05 were considered significant (based on false discovery rates using the Benjamini-Hochberg approach) (24). A heatmap of significantly up or downregu-lated immune-associated genes was plotted using the heatmap package version 1.8.0 (git.bioconductor.org/packages/heat-maps), and these genes were used for further prognostic analysis.

Gene ontology annotation and pathway enrichment

Immune genes determined to be significantly differentially expressed were functionally annotated using the clusterProfiler package (25), which stratified pathways according to one of the following categories: Cellular compartment, biological process or molecular function, and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis was performed to enrich the pathways associated with the identified genes (26).

Construction of the immune risk score (IRS) model

The entire cohort of patients with HCC from TCGA were randomly divided into a training set and a testing set to construct and assess the prognostic model. The DEIGs were evaluated using a univariate Cox model for individual risk factors affecting OS of the training set (P<0.05), and the associated genes were analyzed together using a Lasso penalty linear regression model, which were subsequently used to construct a multivariate Cox model. In Lasso regression, the patients were subsampled 1,000 times, and the genes with an occurrence >900 instances were selected. In multivariate Cox regression analysis, a stepwise method is used, where all combinations of the identified genes are assessed to construct the best combination of the immune associated gene set. The prognostic value of the linear IRS model was validated in the testing set, the entire TCGA cohort and the independent cohort from the ICGC database separately, with patients divided into high- and low- risk sub-populations according to the median IRS. Kaplan-Meier survival curves and time-dependent receiver operating characteristic curves (ROC) were used to demonstrate the prognostic value of the 5-gene IRS model, using the R packages of survival (rdocumentation.org/packages/survival) and survivalROC (27).

Independent prognostic value of IRS and the construction of a nomogram

The independent prognostic value of IRS was further examined through univariate and multivariate Cox regression analysis in combination with the clinical characteristics, such as age, sex, tumor grade and tumor stages. Following evaluation of the risk effect of clinical characteristics and IRS, a nomogram model was constructed for prognostic prediction, which included the IRS and tumor stage. The predictive value of the nomogram was further confirmed using ROC curves for prediction of the 1, 3 and 5 year OS rates, in which the predictive value of the single risk factors were also assessed independently. The C-index of the nomogram was calculated with a bootstrap of 1,000 resamples, and the results ranged between 0.5-1.0, where 0.5 indicated a random chance and 1.0 indicated perfect separation of the outcomes. Calibration curves were also plotted to demonstrate the precision of the nomogram, contrasting the predictive probability with the actual incidence.

Tumor infiltrating immune cells and their correlation with IRS

The calculation of tumor infiltrating immune cells in patients with HCC from TCGA was performed using Tumor IMmune Estimation Resource (TIMER), an online tool which contains the reanalyzed genomic expression data across 32 types of cancer, including over 10,897 samples from TCGA (28,29). The online portal calculates the abundance of 6 types of infiltrating immune cells; B cells, CD4+ T cells, CD8+ T cells, neutrophils, dendritic cells and macrophages. The abundance of infiltrating immune cells was correlated with IRS, and significance was examined using a Pearson's correlation test. P<0.05 was considered to indicate a statistically significant difference. The degree of correlation between immune cell abundance and IRS was defined as follows: Very low, 0.0-0.2; low, 0.2-0.4; medium, 0.4-0.6; high, 0.6-0.8; and very high, 0.8-1.0.

Statistical analysis

Statistical analysis was performed in R (version 3.6.1), using R studio (version 1.2.1335) (30,31). DEIGs between adjacent and HCC tissues were analyzed using a Wilcoxon Signed-rank test. Univariate Cox regression, Lasso regression and multivariate Cox regression analysis were performed to construct the IRS model. The infiltration levels of different immune cells between HCC and para-tumor tissues were compared using Pearson's correlation coefficients. P<0.05 was considered to indicate a statistically significant difference.

Results

Analysis strategy and overview of the DEIGs

Analysis of the TCGA dataset identified 247 significantly differently expressed genes, of which 200 were upregulated and 47 were downregulated (Fig. 1A and B). Gene ontology of the DEIGs were primarily associated with immune cell activation, adhesion or responses to stimuli (Fig. 1C and E). The enriched pathways for those DEIGs were primarily enriched in cytokine and cytokine receptor interactions between cells, in which the cytokine-cytokine receptor interaction pathways had the highest counts of associated genes and significance. Additionally, the z-score of the cytokine-cytokine receptor interaction pathways was the highest ranked amongst all enriched pathways (Fig. 1D and F).

Relative risk effect of the immune associated genes and construction of the IRS based

Patients from TCGA were divided into two sets; a training set and an internal test set. In the training set, significant DEIGs (P<0.05) associated with OS in the Cox model (Fig. 2A), were further analyzed using a Lasso penalty linear regression model (Fig. 2B). The final multivariate Cox model was constructed using 5 genes [Enhancer of zest homology 2 (EZH2), ferritin light chain (FTL), complement factor H related 3 (CFHR3), isthmin 2 (ISM2), cyclin dependent kinase 5 (CDK5)], of which EZH2 and ISM2 still significant and had high risk effect following adjustment (Fig. 2C).

The final model was then used to calculate the IRS of patients for prognostic evaluation in four separate cohorts: The training cohort from TCGA (Fig. 3A-C), the test cohort from TCGA (Fig. 3D-F), the entire TCGA cohort (Fig. 4A-C) and the independent cohort from ICGC (Fig. 4D-F). The median IRSs of the four cohorts were used to stratify patients into high- and low-score groups.

Survival validation of the IRS model

The IRS model, based on the 5 significantly changed immune associated genes, was able to divide the patients with HCC into high- and low-risk sub-groups based on the corresponding score levels. Patients with higher scores had a worse prognosis in all four cohorts (Fig. 5A-D). The area under the curve (AUC) values of the model for 0.5, 1, 2, 3 and 5 year survival in all four cohorts were all ~0.7, with a lowest value of 0.643 for 5 year survival in the test cohort (Fig. 5E-H) Further analysis of patients with different tumor stages and grades, showed that a high IRS predicted worse survival in both datasets from TCGA and ICGC (P<0.05), demonstrating the independent prognostic value of IRS for clinical use. In patients with stage I & II, III & IV and grade 1 & 2 cancer in TCGA, the curves for high- and low-IRS showed notable differences in the 6 year survival, whereas after 6 years of follow-up, the curves nearly overlapped (Fig. 6A-C). In patients with grade 3 & 4 cancer from TCGA, and stage I & II, and III & IV patients from ICGC, the curves of patients with high- and low-IRS diverted with no overlap (Fig. 6D-F). Regarding disease free survival, patients with a high IRS also exhibited worse outcomes compared with patients with a low IRS, and this difference was significant in the entire TCGA cohort, in patients with stage I & II, and grade 1 & 2 cancer from TCGA (Fig. S1).

Nomogram of IRS and other associated clinical factors

To assess the clinical relevance and significance of the IRS model, the IRS model was combined with the clinical characteristics for prognostic prediction of the data obtained from TCGA and ICGC. In univariate and multivariate analysis of patients from TCGA, cancer stage and IRS were significantly associated with survival, with or without adjustment (Fig. 7A and B). In the ICGC dataset, in addition to cancer stage and IRS, the presence of a previous malignancy was also significantly associated with survival following adjustment (Fig. 7C and D). The correlations between the 5-gene model and clinical characteristics are presented in Fig. 7E and F.

For clinical use, a nomogram was constructed for all the significant factors, including clinical tumor stage and IRS, using the entire TCGA cohort. (Fig. 8A) The AUCs for 1, 3 and 5 year survival were higher in the nomogram compared with IRS or tumor stage (Fig. 8C) The C-index for the nomogram-predicted OS was 0.749, with 1,000 cycles of bootstrapping (Table II). Calibration graphs were drawn to evaluate the corresponding performance of the nomogram for predicting 1, 3 and 5 year OS, and the lines almost overlapped, suggesting its accuracy (Fig. 8B). These results all show the value of the nomogram for predicting OS in patients following resection, and was shown to be more accurate than tumor stage or IRS, for both short- and long-term.

Table II

C-index analysis of models.

Table II

C-index analysis of models.

ModelC-index
Stage model0.654
Prognostic model0.746
Nomogram model0.749
Potential roles of immune infiltrating cells in prognostic prediction

Using the evaluation scores of six common types of immune infiltrating cells from TIMER, the abundance of immune-cells in HCC tissue was estimated. Patients in TCGA were used to calculate the abundance of infiltration, and a score was generated by the tool. The correlation between the scores of B cells, CD8+ T cells, dendritic cells, macrophages, neutrophils and IRS were significant and positive, suggesting an association between increased infiltration and IRS (Fig. 9). However, five types of infiltrating immune cells had very low correlation coefficients with IRS, and macrophages had a low correlation, suggesting that the IRS based on the five-gene model was primarily dependent on change sin expression of immune-associated genes in HCC tissues, as opposed to infiltration of immune cells.

Discussion

Malignant HCC is associated with poor outcomes and with high recurrence rates following resection (1,2). In addition, a growing body of evidence highlights the vital value of immune regulation in HCC, and poor responses to treatment with chemotherapy highlight the need for drugs with greater specificity for immune targets (3-6). The development of immune checkpoint blockers, such as nivolumab and pembrolizumab have not yielded optimistic results for patients with HCC, and this may be associated with the immune microenvironment of HCC tissues, as patients with a high degree of immune cell infiltration often exhibit more favorable outcomes (3). Understanding the immune environment and immune status of HCC may result in improved strategies for treatment of patients, resulting in improved prognosis.

In the present study, the differentially expressed immune associated genes between HCC and adjacent tissues were identified, and following Lasso regression and multivariate Cox analysis, 5 significantly differentially expressed OS-related immune associated genes (EZH2, FTL, CFHR3, ISM2, CDK5) were used to build a prognostic nomogram in combination with clinical characteristics. The prognostic values of the nomogram for 1, 3 and 5 year survival achieved was >0.8, and performed better compared with the individual clinical risk factors, and may thus be used to stratify patients with HCC in clinical practice, preventing early recurrence.

EZH2 is expression is low in the normal liver, and is associated with methylation of histone H3K27 and recruitment of methyltransferases, which are involved in DNA replication for cancer progression, and stem cell maintenance and differentiation of other cell lineages, such as immune cells (32,33). Several studies have confirmed its prognostic value and importance in various types of cancer, including lymphoma, glioma, head and neck carcinoma, and cervical neoplasia (34-38). Novel therapeutic methods have been developed to target EZH2, although high expression of EZH2 is not always correlated with the malignancy of a cancer, such as in colorectal cancer (39-41). In the present study, it was shown that high expression of EZH2 was significantly associated with poor OS in patients in the univariate analysis, including after adjustment, highlighting the prognostic value of high EZH2 expression in patients with HCC. EZH2 may promote the development and proliferation of HCC, and may thus result in recurrence following tumor resection.

Ferritin light chain (FTL) is the light subunit of the ferritin protein, which is involved in iron release and uptake in tissues. Several allergic diseases, inflammation status and oxidative damage are associated with the roles of FTL, including systemic lupus erythematosus, cataracts, hepatitis E virus infection and Alzheimer's disease (42-47). FTL also serves a role in cancer, where the dysfunction of iron metabolism is a hallmark of various types of cancer, and is involved drug resistance and malignant progression (48-50). Although the risk effect of FTL was small following adjustment in the multivariate Cox model, the potential role of changes to iron metabolism in the progression of HCC should not be ignored.

CFHR3 was demonstrated to exert a protective effect in patients with HCC, and physiologically, CFHR3 is exclusively expressed in normal liver (51). CFHR3 is associated with compliment factor H, which can bind to the C3d region of C3b, regulating the function of compliment system (52). The loss of CFHR3 results in age-related macular degeneration, and high expression levels of CFHR3 may result in atypical hemolytic-uremic syndrome (53-54). There are no published articles regarding the expression of CFHR3 in cancer, and the protective role of CFHR3 observed in the present study may highlight a potential change in the expression profile that may be used for improving the prognosis of patients with HCC.

In both univariate and multivariate analysis, ISM2 was significantly associated with poor outcomes and was considered a high risk factor in patients with HCC. ISM2 is a component of thrombospondin (THBS), which promotes the activity of mesenchymal and stromal cells through TGF-β, and regulates secretion of inflammatory cytokines through the NF-κB signaling pathway (55,56). THBS promotes epithelial-to-mesenchymal transition in melanoma, and exacerbates the progression of prostate cancer to more advanced stages (57,58). Additionally, THBS may serve a role in gastric carcinogenesis, and invasiveness of breast cancer and nodal metastasis (59-62). The role of ISM2 or THBS in HCC has not been explored to the best of our knowledge, and further analysis is required to understand their potential roles and effects.

CDK5 has been extensively studied as an important factor in tumor development and metastasis (63-65). Although CDK5 shares homologous structure with other CDKs, it is not cyclin-dependent and does not need to be phosphorylated in the T-loop for activation (66). CDK5 expression is upregulated in several types of cancer, and inhibition induces cancer cell death through a FOXO1-Bim pathway or mitochondrial dysfunction (67-73). Ehrlich et al (74) showed that expression of CDK5 was increased in HCC tissues, and was correlated with malignant phenotypes. Additionally, CDK5 was most active in the G2/M phase of cancer cells in the nucleus, and regulated DNA damage response through phosphorylation of ataxia telangiectasia mutated kinase, validating the prognostic role of CDK5 in the present study.

Infiltration of immune cells in HCC tissues was assessed, and 5 of the 6 common types of immune cells were significantly associated with IRS; however the correlation coefficients for all 6 types of cells were either low or very low. Thus, although patients with a high degree of immune cell infiltration may have a high IRS, the IRS based on the five immune associated genes primarily accounted for the functional status of the microenvironment in HCC tissues. Recent studies have focused on the roles of infiltrating immune cells in the tumor microenvironment, to explain the mechanisms underlying immune evasion and to predict drug response or prognosis (75-77). Further analysis of the sub-types of immune cells in HCC may improve our understanding of immune function in tumor, and with advances in technologies, changing the types of immune cells present may be considered as a potential treatment strategy, emphasizing the importance of restoring immune function in HCC (78-80).

The IRS model and the nomogram developed in the present study may exhibit value in clinical practice for prognostic prediction. Based on the IRS model and nomogram, it may be possible to tailor therapeutic regimens to each specific patient, or they may be useful for predicting/detecting early recurrence, and to evaluate immune function in HCC to optimize the benefits of monoclonal targeting therapies.

The present study has some limitations. First, the cohorts from TCGA and ICGA are primarily from several local populations, and thus may not be applicable to all ethnicities. Second, the DEIGs in this analysis may not reveal the holistic changes in the immune microenvironment in HCC. Third, the present study focused primarily on overall survival of patients following tumor resection, and disease-free survival was not assessed as this information was not contained in the data-sets. Furthermore, experimental validation of the prognostic signatures in HCC cell lines and human tissues is required to validate their relevance and improve our understanding of their respective roles, and will be performed in future experiments.

In conclusion, a 5-gene model was constructed from differentially expressed immune associated genes to evaluate the IRS of patients for independent prognostic prediction. By combining IRS with clinical tumor stage, a nomogram was constructed with efficient predictive value for 1, 3 and 5 year OS for patients with HCC. This nomogram may be used clinically for monitoring early recurrence and prognostic prediction.

Supplementary Data

Funding

The present study was supported by the Education and Scientific Research Project of Middle and Young Teachers in Fujian Province (grant no. JAT170245).

Availability of data and materials

The datasets analyzed during the present study are available from The Cancer Genome Atlas (portal.gdc.cancer.gov/) and International Cancer Genome Consortium (icgc.org/) repository.

Authors' contributions

WL and CF designed the study and participated in data collection. RH, ZC and JL analyzed and interpreted the data, and wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests

Acknowledgements

Not applicable.

References

1 

Singal AG, Lampertico P and Nahon P: Epidemiology and surveillance for hepatocellular carcinoma: New trends. J Hepatol. 72:250–261. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer sta-tistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA-Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar

3 

Chen L and Han X: Anti-PD-1/PD-L1 therapy of human cancer: Past, present, and future. J Clin Invest. 125:3384–3391. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Sun C, Mezzadra R and Schumacher TN: Regulation and function of the PD-L1 checkpoint. Immunity. 48:434–452. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Liu G, Rui W, Zheng H, Huang D, Yu F, Zhang Y, Dong J, Zhao X and Lin X: CXCR2-modified CAR-T cells have enhanced trafficking ability that improves treatment of hepatocellular carcinoma. Eur J Immunol. Jan 24–2020.Epub ahead of print. View Article : Google Scholar

6 

Batra SA, Rathi P, Guo L, Courtney AN, Fleurence J, Balzeau J, Shaik RS, Nguyen TP, Wu MF, Bulsara S, et al: Glypican-3-specific CAR T cells co-expressing IL15 and IL21 have superior expansion and antitumor activity against hepato-cellular carcinoma. Cancer Immunol Res. Jan 17–2020.Epub ahead of print. View Article : Google Scholar

7 

Jindal A, Thadi A and Shailubhai K: Hepatocellular carcinoma: Etiology and current and future drugs. J Clin Exp Hepatol. 9:221–232. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Zucman-Rossi J, Villanueva A, Nault JC and Llovet JM: Genetic landscape and biomarkers of hepatocellular carcinoma. Gastroenterology. 149:1226–1239.e4. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Cancer Genome Atlas Research Network: Electronic address simplewheeler@bcm.edu; Cancer Genome Atlas Research Network: Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell. 169:1327–1341.e23. 2017. View Article : Google Scholar

10 

Li B, Cui Y, Diehn M and Li R: Development and validation of an individualized immune prognostic signature in early-stage nonsquamous non-small cell lung cancer. JAMA Oncol. 3:1529–1537. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Chen CH, Lu YS, Cheng AL, Huang CS, Kuo WH, Wang MY, Chao M, Chen IC, Kuo CW, Lu TP and Lin CH: Disparity in tumor immune microenvironment of breast cancer and prognostic impact: Asian versus Western populations. Oncologist. 25:e16–e23. 2020. View Article : Google Scholar :

12 

Ge P, Wang W, Li L, Zhang G, Gao Z, Tang Z, Dang X and Wu Y: Profiles of immune cell infiltration and immune-related genes in the tumor microenvironment of colorectal cancer. Biomed Pharmacother. 118:1092282019. View Article : Google Scholar : PubMed/NCBI

13 

Lin P, Guo YN, Shi L, Li XJ, Yang H, He Y, Li Q, Dang YW, Wei KL and Chen G: Development of a prognostic index based on an immunogenomic landscape analysis of papillary thyroid cancer. Aging (Albany NY). 11:480–500. 2019. View Article : Google Scholar

14 

Wang J, Li Y, Fu W, Zhang Y, Jiang J, Zhang Y and Qi X: Prognostic nomogram based on immune scores for breast cancer patients. Cancer Med. 8:5214–5222. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Foerster F, Hess M, Gerhold-Ay A, Marquardt JU, Becker D, Galle PR, Schuppan D, Binder H and Bockamp E: The immune contexture of hepatocellular carcinoma predicts clinical outcome. Sci Rep. 8:53512018. View Article : Google Scholar : PubMed/NCBI

16 

Zheng M and Tian Z: Liver-mediated adaptive immune tolerance. Front Immunol. 10:25252019. View Article : Google Scholar : PubMed/NCBI

17 

Lu LC, Hsu C, Shao YY, Chao Y, Yen CJ, Shih IL, Hung YP, Chang CJ, Shen YC, Guo JC, et al: Differential differential organ-specific tumor response to immune checkpoint inhibitors in hepatocellular carcinoma. J Liver Cancer. 8:480–490. 2019. View Article : Google Scholar

18 

Keenan BP, Fong L and Kelley RK: Immunotherapy in hepatocel-lular carcinoma: The complex interface between inflammation, fibrosis, and the immune response. J Immunother Cancer. 7:2672019. View Article : Google Scholar

19 

Charoentong P, Finotello F, Angelova M, Mayer C, Efremova M, Rieder D, Hackl H and Trajanoski Z: Pan-cancer immunoge-nomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 18:248–262. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Rooney MS, Shukla SA, Wu CJ, Getz G and Hacohen N: Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 160:48–61. 2015. View Article : Google Scholar : PubMed/NCBI

21 

International Cancer Genome Consortium; Hudson TJ, Anderson W, Aretz A, Barker AD, Bell C, Bernabé RR, Bhan MK, Calvo F, Eerola I, et al: International network of cancer genome projects. Nature. 464:993–998. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Tomczak K, Czerwinska P and Wiznerowicz M: The cancer genome atlas (TCGA): An immeasurable source of knowledge. Contemp Oncol (Pozn). 19:A68–A77. 2015.

23 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

24 

Hu J, Koh H, He L, Liu M, Blaser MJ and Li H: A two-stage microbial association map-ping framework with advanced FDR control. Microbiome. 6:1312018. View Article : Google Scholar

25 

Yu G, Wang LG, Han Y and He QY: clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Kanehisa M: Post-genome Informatics. Oxford University Press; 2000

27 

Heagerty PJ: Compute time-dependent ROC curve from censored survival data using Kaplan-Meier (KM) or Nearest Neighbor Estimation (NNE) method of Heagerty, Lumley & Pepe. Biometrics. 56:337–344. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Li B, Severson E, Pignon JC, Zhao H, Li T, Novak J, Jiang P, Shen H, Aster JC, Rodig S, et al: Comprehensive analyses of tumor immunity: Implications for cancer immunotherapy. Genome Biol. 17:1742016. View Article : Google Scholar : PubMed/NCBI

29 

Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, Li B and Liu XS: TIMER: A web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res. 77:e108–e110. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Team RC: R: A language and environment for statistical computing. R Foundation for Statistical Computing; Vienna, Austria: 2012

31 

Team RS: RStudio: Integrated Development for R. RStudio, Inc; Boston, MA: 2015

32 

A P, Xu X, Wang C, Yang J, Wang S, Dai J and Ye L: EZH2 promotes DNA replication by stabilizing interaction of POLδ and PCNA via methylation-mediated PCNA trimerization. Epigenetics Chromatin. 11:442018. View Article : Google Scholar

33 

Batool A, Jin C and Liu YX: Role of EZH2 in cell lineage determination and relative signaling pathways. Front Biosci (Landmark Ed). 24:947–960. 2019. View Article : Google Scholar

34 

Cheng T and Xu Y: Effects of enhancer of zeste homolog 2 (EZH2) expression on brain glioma cell proliferation and tumori-genesis. Med Sci Monit. 24:7249–7255. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Mochizuki D, Misawa Y, Kawasaki H, Imai A, Endo S, Mima M, Yamada S, Nakagawa T, Kanazawa T and Misawa K: Aberrant epigenetic regulation in head and neck cancer due to distinct EZH2 overexpression and DNA hypermethylation. Int J Mol Sci. 19:pii: E3707. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Karlowee V, Amatya VJ, Takayasu T, Takano M, Yonezawa U, Takeshima Y, Sugiyama K, Kurisu K and Yamasaki F: Immunostaining of increased expression of enhancer of zeste homolog 2 (EZH2) in diffuse midline glioma H3K27M-mutant patients with poor survival. Pathobiology. 86:152–161. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Makk E, Bálint L, Cifra J, Tornóczky T, Oszter A, Tóth A, Kálmán E and Kovács K: Robust expression of EZH2 in endocervical neoplastic lesions. Virchows Arch. 475:95–104. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Romanchikova N and Trapencieris P: Wedelolactone targets EZH2-mediated Histone H3K27 methylation in mantle cell lymphoma. Anticancer Res. 39:4179–4184. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Zhou L, Wei E, Zhou B, Bi G, Gao L, Zhang T, Huang J, Wei Y and Ge B: Anti-proliferative benefit of curcumol on human bladder cancer cells via inactivating EZH2 effector. Biomed Pharmacother. 104:798–805. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Böhm J, Muenzner JK, Caliskan A, Ndreshkjana B, Erlenbach-Wünsch K, Merkel S, Croner R, Rau TT, Geppert CI, Hartmann A, et al: Loss of enhancer of zeste homologue 2 (EZH2) at tumor invasion front is correlated with higher aggressiveness in colorectal cancer cells. J Cancer Res Clin Oncol. 145:2227–2240. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Natsumeda M, Liu Y, Nakata S, Miyahara H, Hanaford A, Ahsan S, Stearns D, Skuli N, Kahlert UD, Raabe EH, et al: Inhibition of enhancer of zest homologue 2 is a potential therapeutic target for high-MYC medulloblastoma. Neuropathology. 39:71–77. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Cozzi A, Rovelli E, Frizzale G, Campanella A, Amendola M, Arosio P and Levi S: Oxidative stress and cell death in cells expressing L-ferritin variants causing neuroferritinopathy. Neurobiol Dis. 37:77–85. 2010. View Article : Google Scholar

43 

Vanarsa K, Ye Y, Han J, Xie C, Mohan C and Wu T: Inflammation associated anemia and ferritin as disease markers in SLE. Arthritis Res Ther. 14:R1822012. View Article : Google Scholar : PubMed/NCBI

44 

Kwiatek-Majkusiak J, Dickson DW, Tacik P, Aoki N, Tomasiuk R, Koziorowski D and Friedman A: Relationships between typical histopathological hallmarks and the ferritin in the hippocampus from patients with Alzheimer's disease. Acta Neurobiol Exp (Wars). 75:391–398. 2015.

45 

Döring M, Cabanillas Stanchi KM, Feucht J, Queudeville M, Teltschik HM, Lang P, Feuchtinger T, Handgretinger R and Müller I: Ferritin as an early marker of graft rejection after allogeneic hematopoietic stem cell transplantation in pediatric patients. Ann Hematol. 95:311–323. 2016. View Article : Google Scholar

46 

Ojha NK and Lole KS: Hepatitis E virus ORF1 encoded macro domain protein interacts with light chain subunit of human ferritin and inhibits its secretion. Mol Cell Biochem. 417:75–85. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Yazar S, Franchina M, Craig JE, Burdon KP and Mackey DA: Ferritin light chain gene mutation in a large Australian family with hereditary hyperferritinemia-cataract syndrome. Ophthalmic Genet. 38:171–174. 2017. View Article : Google Scholar

48 

Chekhun VF, Lukyanova NY, Burlaka CA, Bezdenezhnykh NA, Shpyleva SI, Tryndyak VP, Beland FA and Pogribny IP: Iron metabolism disturbances in the MCF-7 human breast cancer cells with acquired resistance to doxorubicin and cisplatin. Int J Oncol. 43:1481–1486. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Wu T, Li Y, Liu B, Zhang S, Wu L, Zhu X and Chen Q: Expression of ferritin light chain (FTL) is elevated in glioblastoma, and FTL silencing inhibits glioblastoma cell proliferation via the GADD45/JNK pathway. PLoS One. 11:e01493612016. View Article : Google Scholar : PubMed/NCBI

50 

Khanna V, Karjodkar F, Robbins S, Behl M, Arya S and Tripathi A: Estimation of serum ferritin level in potentially malignant disorders, oral squamous cell carcinoma, and treated cases of oral squamous cell carcinoma. J Cancer Res Ther. 13:550–555. 2017.PubMed/NCBI

51 

Liu J, Li W and Zhao H: CFHR3 is a potential novel biomarker for hepatocellular carcinoma. J Cell Biochem. Nov 10–2019.Epub ahead of print.

52 

Hellwage J, Jokiranta TS, Koistinen V, Vaarala O, Meri S and Zipfel PF: Functional properties of complement factor H-related proteins FHR-3 and FHR-4: Binding to the C3d region of C3b and differential regulation by heparin. FEBS Lett. 462:345–352. 1999. View Article : Google Scholar

53 

Spencer KL, Hauser MA, Olson LM, Schmidt S, Scott WK, Gallins P, Agarwal A, Postel EA, Pericak-Vance MA and Haines JL: Deletion of CFHR3 and CFHR1 genes in age-related macular degeneration. Hum Mol Genet. 17:971–977. 2008. View Article : Google Scholar

54 

Pouw RB, Gómez Delgado I, López Lera A, Rodríguez de Córdoba S, Wouters D, Kuijpers TW and Sánchez-Corral P: High complement factor H-related (FHR)-3 levels are associated with the atypical hemolytic-uremic syndrome-risk allele CFHR3*B. Front Immunol. 9:8482018. View Article : Google Scholar

55 

Belotti D, Capelli C, Resovi A, Introna M and Taraboletti G: Thrombospondin-1 promotes mesenchymal stromal cell functions via TGFβ and in cooperation with PDGF. Matrix Biol. 55:106–116. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Xing T, Wang Y, Ding WJ, Li YL, Hu XD, Wang C, Ding A and Shen JL: Thrombospondin-1 production regulates the inflammatory cytokine secretion in THP-1 cells through NF-κB signaling pathway. Inflammation. 40:1606–1621. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Firlej V, Mathieu JR, Gilbert C, Lemonnier L, Nakhlé J, Gallou-Kabani C, Guarmit B, Morin A, Prevarskaya N, Delongchamps NB and Cabon F: Thrombospondin-1 triggers cell migration and development of advanced prostate tumors. Cancer Res. 71:7649–7658. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Jayachandran A, Anaka M, Prithviraj P, Hudson C, McKeown SJ, Lo PH, Vella LJ, Goding CR, Cebon J and Behren A: Thrombospondin 1 promotes an aggressive phenotype through epithelial-to-mesenchymal transition in human melanoma. Oncotarget. 5:5782–5797. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Ioachim E, Damala K, Tsanou E, Briasoulis E, Papadiotis E, Mitselou A, Charhanti A, Doukas M, Lampri L and Arvanitis DL: Thrombospondin-1 expression in breast cancer: Prognostic significance and association with p53 alterations, tumour angiogenesis and extracellular matrix components. Histol Histopathol. 27:209–216. 2012.

60 

Lin XD, Chen SQ, Qi YL, Zhu JW, Tang Y and Lin JY: Overexpression of thrombospondin-1 in stromal myofibroblasts is associated with tumor growth and nodal metastasis in gastric carcinoma. J Surg Oncol. 106:94–100. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Horiguchi H, Yamagata S, Rong Qian Z, Kagawa S and Sakashita N: Thrombospondin-1 is highly expressed in desmo-plastic components of invasive ductal carcinoma of the breast and associated with lymph node metastasis. J Med Invest. 60:91–96. 2013. View Article : Google Scholar

62 

Kashihara H, Shimada M, Yoshikawa K, Higashijima J, Tokunaga T, Nishi M, Takasu C and Ishikawa D: Correlation between thrombospondin-1 expression in non-cancer tissue and gastric carcinogenesis. Anticancer Res. 37:3547–3552. 2017.PubMed/NCBI

63 

Pozo K and Bibb JA: The emerging role of Cdk5 in cancer. Trends Cancer. 2:606–618. 2016. View Article : Google Scholar : PubMed/NCBI

64 

Lopes JP and Agostinho P: Cdk5: Multitasking between physiological and pathological conditions. Prog Neurobiol. 94:49–63. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Liebl J, Weitensteiner SB, Vereb G, Takács L, Fürst R, Vollmar AM and Zahler S: Cyclin-dependent kinase 5 regulates endothelial cell migration and angiogenesis. J Biol Chem. 285:35932–35943. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Shupp A, Casimiro MC and Pestell RG: Biological functions of CDK5 and potential CDK5 targeted clinical treatments. Oncotarget. 8:17373–17382. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Liang Q, Li L, Zhang J, Lei Y, Wang L, Liu DX, Feng J, Hou P, Yao R, Zhang Y, et al: CDK5 is essential for TGF-β1-induced epithelial-mesenchymal transition and breast cancer progression. Sci Rep. 3:29322013. View Article : Google Scholar

68 

Yushan R, Wenjie C, Suning H, Yiwu D, Tengfei Z, Madushi WM, Feifei L, Changwen Z, Xin W, Roodrajeetsing G, et al: Insights into the clinical value of cyclin-dependent kinase 5 in glioma: A retrospective study. World J Surg Oncol. 13:2232015. View Article : Google Scholar : PubMed/NCBI

69 

Zhang X, Zhong T, Dang Y, Li Z, Li P and Chen G: Aberrant expression of CDK5 infers poor outcomes for nasopharyngeal carcinoma patients. Int J Clin Exp Pathol. 8:8066–8074. 2015.PubMed/NCBI

70 

Pan DH, Zhu ML, Lin XM, Lin XG, He RQ, Ling YX, Su ST, Wickramaarachchi MM, Dang YW, Wei KL and Chen G: Evaluation and clinical significance of cyclin-dependent kinase5 expression in cervical lesions: A clinical research study in Guangxi, China. Eur J Med Res. 21:282016. View Article : Google Scholar : PubMed/NCBI

71 

Wei K, Ye Z, Li Z, Dang Y, Chen X, Huang N, Bao C, Gan T, Yang L and Chen G: An immunohistochemical study of cyclin-dependent kinase 5 (CDK5) expression in non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC): A possible prognostic biomarker. World J Surg Oncol. 14:342016. View Article : Google Scholar : PubMed/NCBI

72 

Mandl MM, Zhang S, Ulrich M, Schmoeckel E, Mayr D, Vollmar AM and Liebl J: Inhibition of Cdk5 induces cell death of tumor-initiating cells. Br J Cancer. 116:912–922. 2017. View Article : Google Scholar : PubMed/NCBI

73 

NavaneethaKrishnan S, Rosales JL and Lee KY: Loss of Cdk5 in breast cancer cells promotes ROS-mediated cell death through dysregulation of the mitochondrial permeability transition pore. Oncogene. 37:1788–1804. 2018. View Article : Google Scholar : PubMed/NCBI

74 

Ehrlich SM, Liebl J, Ardelt MA, Lehr T, De Toni EN, Mayr D, Brandl L, Kirchner T, Zahler S, Gerbes AL and Vollmar AM: Targeting cyclin dependent kinase 5 in hepatocellular carcinoma-A novel therapeutic approach. J Hepatol. 63:102–113. 2015. View Article : Google Scholar : PubMed/NCBI

75 

Chen QF, Li W, Wu PH, Shen LJ and Huang ZL: Significance of tumor-infiltrating immunocytes for predicting prognosis of hepatitis B virus-related hepatocellular carcinoma. World J Gastroenterol. 25:5266–5282. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Lu J, Xu Y, Wu Y, Huang XY, Xie JW, Wang JB, Lin JX, Li P, Zheng CH, Huang AM and Huang CM: Tumor-infiltrating CD8+ T cells combined with tumor-associated CD68+ macrophages predict postoperative prognosis and adjuvant chemotherapy benefit in resected gastric cancer. BMC Cancer. 19:9202019. View Article : Google Scholar : PubMed/NCBI

77 

Wang J, Li Z, Gao A, Wen Q and Sun Y: The prognostic landscape of tumor-infiltrating immune cells in cervical cancer. Biomed Pharmacother. 120:1094442019. View Article : Google Scholar : PubMed/NCBI

78 

Kobayashi N, Hiraoka N, Yamagami W, Ojima H, Kanai Y, Kosuge T, Nakajima A and Hirohashi S: FOXP3+ regulatory T cells affect the development and progression of hepatocarcino-genesis. Clin Cancer Res. 13:902–911. 2007. View Article : Google Scholar : PubMed/NCBI

79 

Mathai AM, Kapadia MJ, Alexander J, Kernochan LE, Swanson PE and Yeh MM: Role of Foxp3-positive tumor-infiltrating lymphocytes in the histologic features and clinical outcomes of hepatocellular carcinoma. Am J Surg Pathol. 36:980–986. 2012. View Article : Google Scholar : PubMed/NCBI

80 

Brunner SM, Rubner C, Kesselring R, Martin M, Griesshammer E, Ruemmele P, Stempfl T, Teufel A, Schlitt HJ and Fichtner-Feigl S: Tumor-infiltrating, interleukin-33-producing effector-memory CD8(+) T cells in resected hepatocellular carcinoma prolong patient survival. Hepatology. 61:1957–1967. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2020
Volume 56 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang R, Chen Z, Li W, Fan C and Liu J: Immune system‑associated genes increase malignant progression and can be used to predict clinical outcome in patients with hepatocellular carcinoma. Int J Oncol 56: 1199-1211, 2020
APA
Huang, R., Chen, Z., Li, W., Fan, C., & Liu, J. (2020). Immune system‑associated genes increase malignant progression and can be used to predict clinical outcome in patients with hepatocellular carcinoma. International Journal of Oncology, 56, 1199-1211. https://doi.org/10.3892/ijo.2020.4998
MLA
Huang, R., Chen, Z., Li, W., Fan, C., Liu, J."Immune system‑associated genes increase malignant progression and can be used to predict clinical outcome in patients with hepatocellular carcinoma". International Journal of Oncology 56.5 (2020): 1199-1211.
Chicago
Huang, R., Chen, Z., Li, W., Fan, C., Liu, J."Immune system‑associated genes increase malignant progression and can be used to predict clinical outcome in patients with hepatocellular carcinoma". International Journal of Oncology 56, no. 5 (2020): 1199-1211. https://doi.org/10.3892/ijo.2020.4998