Skip to main content

A novel natural killer-related signature to effectively predict prognosis in hepatocellular carcinoma

Abstract

Background

Hepatocellular carcinoma (HCC) is a prevalent tumor that poses a significant threat to human health, with 80% of cases being primary HCC. At present, Early diagnosis and predict prognosis of HCC is challenging and the it is characterized by a high degree of invasiveness, both of which negatively impact patient prognosis. Natural killer cells (NK) play an important role in the development, diagnosis and prognosis of malignant tumors. The potential of NK cell-related genes for evaluating the prognosis of patients with hepatocellular carcinoma remains unexplored. This study aims to address this gap by investigating the association between NK cell-related genes and the prognosis of HCC patients, with the goal of developing a reliable model that can provide novel insights into evaluating the immunotherapy response and prognosis of these patients. This work has the potential to significantly advance our understanding of the complex interplay between immune cells and tumors, and may ultimately lead to improved clinical outcomes for HCC patients.

Methods

For this study, we employed transcriptome expression data from the hepatocellular carcinoma cancer genome map (TCGA-LIHC) to develop a model consisting of NK cell-related genes. To construct the NK cell-related signature (NKRLSig), we utilized a combination of univariate COX regression, Area Under Curve (AUC) LASSO COX regression, and multivariate COX regression. To validate the model, we conducted external validation using the GSE14520 cohort.

Results

We developed a prognostic model based on 5-NKRLSig (IL18RAP, CHP1, VAMP2, PIC3R1, PRKCD), which divided patients into high- and low-risk groups based on their risk score. The high-risk group was associated with a poor prognosis, and the risk score had good predictive ability across all clinical subgroups. The risk score and stage were found to be independent prognostic indicators for HCC patients when clinical factors were taken into account. We further created a nomogram incorporating the 5-NKRLSig and clinicopathological characteristics, which revealed that patients in the low-risk group had a better prognosis. Moreover, our analysis of immunotherapy and chemotherapy response indicated that patients in the low-risk group were more responsive to immunotherapy.

Conclusion

The model that we developed not only sheds light on the regulatory mechanism of NK cell-related genes in HCC, but also has the potential to advance our understanding of immunotherapy for HCC. With its strong predictive capacity, our model may prove useful in evaluating the prognosis of patients and guiding clinical decision-making for HCC patients.

Peer Review reports

Background

Hepatocellular carcinoma represents the most prevalent form of malignant liver tumors in adults. Recent global cancer statistics indicate that primary liver cancer ranks as the sixth most frequently diagnosed cancerous condition, while simultaneously holding the second highest mortality rate [1]. At present, surgical resection and liver transplantation are considered the primary treatment options for liver cancer. However, the stealthy onset and rapid progression of liver cancer often result in late-stage diagnoses and metastases, making it challenging to treat [2]. Later stage diagnosis and metastasis often accompany liver cancer in many patients [3]. Despite the promising progress and widespread use of targeted therapy [4], the overall survival rate of patients with HCC has remained largely unchanged due to drug resistance and adverse effects associated with this treatment modality. Therefore, there is an urgent need to study new treatment targets and biomarkers to better improve the prognosis of patients. NK cells are important immune cells in the body [5]. Compared to B cells and T cells, NK cells have the unique ability to kill tumor cells in a nonspecific manner, without requiring prior sensitization [6]. NK cells play a crucial role in the immune response against tumors by effectively eliminating cancer cells [7]. Furthermore, NK cells can also play an important indirect immune-mediated role by cooperating with other innate immune cells to regulate the immune status and function of the body. They secrete various chemokines and cytokines that enhance immune defense and help to maintain the immune balance of the body [8]. Therefore, it is considered a potential target for cancer immunotherapy [9]. In recent years, immunotherapy, as the fourth treatment after surgery, radiotherapy, and chemotherapy, has been gradually applied to clinical practice [10]. In addition to the focus on chimeric antigen receptor T-cell (CRT-T) therapy, NK cell-based immunotherapy has emerged as a novel approach for treating both solid and hematologic tumors [11]. It has been reported that NK cell immunotherapy has greatly improved the adverse reactions and survival time of patients with liver cancer, which can effectively improve the quality of life of patients [12]. Currently, the significance of NK cell-related genes in the prognosis of HCC patients is not well understood. Thus, this study aims to conduct a comprehensive analysis of the relationship between the expression of genes associated with NK cells and the prognosis of HCC patients.

Materials and methods

Dataset and sample extraction

RNA sequencing data (RNA-seq), clinical features, and mutation data of TCGA-LIHC cohort dataset were obtained from The Cancer Genome Atlas (TCGA, https://portal.gdc.cancer.gov/) [13]. To begin with, data from 424 HCC patients were initially collected from the TCGA database. Patients with incomplete follow-up data or a survival time of less than 30 days, as well as those lacking complete clinical data, were excluded from the follow-up analysis. Ultimately, a total of 319 cancer patients and 50 healthy controls were enrolled. To account for the same genes with different probes, the average was taken. The data was then standardized using log2 (TPM + 1) transformation, and was normalized by “normalizeBetweenArrays” function of "limma" R package. To obtain the necessary data for this study, we downloaded the GSE14520 dataset from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/) [14]. We carefully excluded patients who had a survival time of less than 30 days, lacked clinical data, or had incomplete follow-up data. In total, 177 cases of HCC patients were included in the study. And standardize the data with the "limma" R package. GSE14520 is considered an externally validated dataset. To investigate the relationship between NK-cell-related genes and the prognosis of HCC patients, a total of 273 NK-cell-related genes were collected. These genes were identified by gathering 134 NK-cell-related genes from the ImmPort Portal website (Additional file 1. Table S1) and 31 gene sets obtained from the MSigDB database (Additional file 2. Table S2). Finally, we removed the duplicate genes, resulting in a final list of 273 NK-cell-related genes. This study is based on the Helsinki Declaration, which was revised in 2013.

Identification of NKRLSig and formulation of modal for prediction of prognosis of HCC patients

To investigate the differential expression of NK cell-related genes between normal and tumor tissues, we employed the "limma" R package, a widely used tool for analyzing microarray and RNA-seq data. The screening criterion we used was a threshold of |log2FC|> 0.585 and an adjusted p-value of less than 0.05 [15]. To construct the 5-NKRLSig, we intersected the differentially expressed genes obtained from the analysis of normal and tumor tissues with known NK cell-related genes. This approach allowed us to identify a list of genes that were differentially expressed and also functionally related to NK cells. Through univariate COX regression with p < 0.05 [16]. We identified the NK regulatory genes related to survival. Then we used "survivalROC" R package to calculate AUC [17] and "glmnet" package to run LASSO COX regression analysis [18] in the training set. To further screen genes, we utilized a five-fold cross-validation method to determine the penalty regularization parameters. Finally, to determine the most significant genes for the 5-NKRLSig model, we used multivariate COX regression analysis to identify the central gene and its coefficients. Based on the central gene and coefficients, we constructed the 5-NKRLSig model and calculated the risk score using the regression coefficient obtained from the multivariate COX regression analysis. The risk scoring formula is established as follows: Risk score = ExpressionmRNA1 × CoefmRNA1 + ExpressionmRNA2 × CoefmRNA2 + ExpressionmRNA3 × CoefmRNA3 + ExpressionmRNA4 × CoefmRNA4 + ExpressionmRNA5 × CoefmRNA5. According to the risk score, the cut-off point was calculated using the ‘surv.cutpoint’ function of "survminer" R package based on the minimum P-value. We divided all patients in the TCGA-LIHC cohort into high- and low-risk groups based on the cut-off value and plotted their respective Kaplan–Meier survival curves [19]. Finally, the accuracy of the model was evaluated using Time-ROC analysis [20].

Functional enrichment analysis

Gene Ontology (GO) is a structured standard biological model that covers three aspects: cell composition (CC), molecular function (MF), and biological process (BP) [21]. Kyoto Encyclopedia of Genes and Genomes (KEGG) is a database that provides a systematic and comprehensive mapping of genes and expression information to pathways, which aids in the understanding of gene metabolic processes in organisms. We utilized the "clusterProfiler" R package to perform GO and KEGG enrichment analysis of 115 NK-related genes, with a significance threshold of p < 0.05 [22]. The observed differences were found to be statistically significant, and to facilitate visualization of the results, we used the "circlize" R package. In addition, Gene set enrichment analysis (GSEA) was conducted to identify distinct biological processes and signaling pathways that differentiate high-risk and low-risk groups in HCC.

Construction of the nomogram

We integrated the established NKRLSig model with clinical information to evaluate the independent prognostic value of the risk score, using univariate COX regression, LASSO COX regression and multivariate COX regression. To improve clinical applicability, we further developed a nomogram by integrating the genetic model and clinical information, and created it using the "rms" R package. The nomogram was used to predict the 1-, 3-, and 5-year survival status of patients in the TCGA-LIHC cohort [23].

Analysis of somatic mutation data and TMB

TMB, which stands for tumor mutational burden, represents the sum of base substitution, insertion, and deletion mutations per trillion bases in the tumor exon coding region [24]. To visually analyze the number of copy number variations of somatic non-synonymous mutations in each sample, we utilized the "maftools" R package [25]. Additionally, we compared the TMB of the high- and low-risk groups and generated abrupt OncoPrint diagrams using the "ComplexHeatmap" R package [26].

Tumor microenvironment analysis

The "Estimate" R package was utilized to estimate the composition of the immune matrix in the HCC tumor microenvironment (TME) [27]. Subsequently, the immune score, matrix score, and ESTIMATE score were compared between the low-risk and high-risk groups.

Evaluation of immunotherapy, chemotherapy and target therapy based on risk score

To further evaluate the clinical utility of the risk score, we utilized the TIDE algorithm [28] to predict the immunotherapy sensitivity of the high-risk and low-risk groups. Additionally, we assessed the immune predictive score (IPS) of the two groups to predict the response to immunotherapy [29]. We obtained the patient immunotherapy data from the Cancer Immunotherapy Response Prediction and Outcome (CRIPO) group database and analyzed the response of patients in the high-risk and low-risk groups to different chemotherapeutic drugs using the "pRRophetic" R package to predict the semi-inhibitory concentration (IC50) [30].

Statistical analysis

The data were analyzed using R software version 4.2.1. Univariate COX regression analysis, AUC, LASSO-COX regression analysis, and multivariate COX regression analysis were employed to construct the 5-NKRLSig model. The Kaplan–Meier (KM) survival curve was used to compare the overall survival (OS) of the high-risk and low-risk groups. The performance of the model was evaluated using a time-dependent receiver operating characteristic (ROC) curve. The Wilcoxon rank-sum test was utilized to compare the proportion of tumor infiltrating immune cells, immune checkpoints, and immune function between the two groups. P value of less than 0.05 was considered statistically significant.

Results and discussion

Screening of NK cell-related genes and construction of the NKRLSig model

Figure 1 outlines the main design of this study. We obtained the intersection of NK cell genes, and a total of 85 up-regulated genes and 30 down-regulated genes were obtained (Fig. 2A). Next, we utilized the "pheatmap" R package to perform a comprehensive visual analysis, generating both a heat map (Fig. 2B) and a volcano map (Fig. 2C) to represent the differential expression of genes related to NK cells. Furthermore, the results of univariate COX regression analysis revealed that a total of 35 NK cell-related genes were significantly associated with the prognosis of HCC patients. The corresponding p-values and hazard ratios (HR) were also presented (Additional file 3. Table S3). To improve the model's accuracy, we performed single-factor screening and calculated the AUC for each gene [17]. Genes with an AUC value greater than 0. 6 were used as our screening condition. The ROC curve is significant because an AUC value greater than 0.6 indicates high precision in predicting patient survival using the gene. Figure 2D shows the P and HR values of variables filtered by univariate COX regression and AUC (Fig. 2D). To improve variable screening, we employed LASSO COX regression and conducted fivefold cross-validation to obtain the λ value, which was determined as the lowest point on the cross-validation plot. Subsequently, we screened the variables and generated a LASSO regression curve (Fig. 2E) and a cross-validation plot (Fig. 2F). Seven genes selected by LASSO regression (IL18RAP, CHP1, MCM10, PRKCD, PIK3R1, VAMP2, FYN) were included in multivariate cox regression and the coefficient was calculated.

Fig. 1
figure 1

Working flow chart, TCGA-LIHC: The Cancer Genome Atlas-Live Hepatocellular Carcinoma, ROC: Receiver operating characteristic

Fig. 2
figure 2

Construction of the NKRLSig model based on the target genes A A Venn diagram depicts 115 overlapping mRNAs considered NK cell-related, including 85 down-regulated genes and 30 up-regulated genes. B Heat map plot differentially expressed NKRLSig. C Volcano plot differentially expressed NKRLSig. D Forest plot showing 17 mRNAs with hazard ratios (95%confidence intervals) and P values based on the result of univariate Cox regression analysis. E–F mRNAs screened by the LASSO-Cox regression model

According to the results of the multivariate COX regression (Fig. 3A), we established the risk score formula: risk score = (- 1.08665 × IL18RAPexpression) + (- 0.43843 × CHP1expression) + (- 0.33298 × VAMP2Gexpression) + (- 0.19419 × PIK3R1expression) + (- 0.24772 × PRKCDexpression). To provide a more visual representation of the relationship between the target gene and the risk score, we constructed a correlation circus plot for the risk scores (Fig. 3B). According to the risk score, the cut-off point value was calculated to be -4.3882 by using "survminer" R package “sur.cutpoint” function, and all patients in the TCGA-LIHC cohort were divided into high-risk and low-risk groups. Finally, the study cohort consisted of 319 HCC patients, with 45 individuals in the high-risk training group and 274 in the low-risk training group. In the high-risk testing group, there were 56 patients, while in the low-risk testing group, there were 121 patients. Survival analysis was conducted separately on the training set and validation set, and the Kaplan–Meier analysis results (Fig. 3C-D) demonstrated that patients with low risk had a better prognosis compared to those with high risk. Furthermore, we generated a risk score distribution map (Fig. 3E) to visualize the association between risk scores and patient mortality, revealing that risk scores were significantly elevated in patients who had experienced more deaths. Additionally, differential expression analysis revealed distinct expression profiles of the five mRNAs between the high and low-risk groups, underscoring the close relationship between these mRNAs and patient prognosis in our study.

Fig. 3
figure 3

Evaluation of the predictive efficacy of the prognostic model. A The multivariate Cox relapse coefficient. B Circus plot show the correlation between risk scores and target genes. Kaplan–Meier survival curves in the high- and low-risk groups stratified by risk scores for overall survival in the training set C and test set D. E The risk score distribution and patient status for the TCGA-LIHC cohort

Evaluation of the precision of the 5-NKRLSig model

To further evaluate the accuracy of the 5-NKRLSig model we constructed, we drew time-dependent ROC curves for the training and testing sets (Fig. 4A-B). The AUC results further demonstrated the model's accuracy. Specifically, the AUC for the first, third, and fifth year in the training set were 0.793 (0.720–0.867), 0.761 (0.689–0.833), and 0.701 (0.599–0.803), respectively. In the external testing set (GSE14520), the AUC for the 1st, 3rd, and 5th years were 0.658 (0.530–0.787), 0.705 (0.617–0.792), and 0.637 (0.527–0.746), respectively. Overall, our model demonstrated good prediction ability for the accuracy of 1, 3, and 5 years in both the training and testing sets.

Fig. 4
figure 4

Verified the accuracy of the prognostic model. Time-dependent ROC curves analysis in the train set A and test set B. C GO and KEGG functional enrichment analysis of NKRLSig

Functional enrichment analysis

We conducted a comprehensive analysis of the differential expression of genes related to NK cells using GO and KEGG enrichment analysis. The selection criteria for significantly enriched items were a threshold of FDR < 0.05 and P < 0.05. GO enrichment analysis categorized the differential genes into molecular biological function, biological process, and cellular components to gain a more comprehensive understanding of gene function. The KEGG pathway enrichment score was used to functionally annotate differentially expressed genes to understand the related functions and pathways of these genes. In addition, we also used GSEA enrichment analysis between high- and low-risk groups.

Our analysis of 115 differential genes related to NK cells revealed that the biological processes with higher abundance were leukocyte-mediated immunity, regulation of immune effector processes, and cell killing. The most abundant cellular components were the external side of the plasma membrane, membrane raft, and membrane microdomain. The most abundant biological functions were cytokine receptor binding, immune receptor activity, and signaling receptor activator activity. In terms of KEGG pathway enrichment, the pathways with higher abundance were natural killer cell-mediated cytotoxicity, human cytomegalovirus infection, and Kaposi sarcoma-associated herpesvirus infection (Fig. 4C).

In this study, we performed Gene Set Enrichment Analysis (GSEA) to compare the high-risk and low-risk groups in the training set. The results revealed distinct enrichment patterns in each group. Specifically, the high-risk group showed significant enrichment in pathways associated with cell cycle, DNA replication, pathogenic Escherichia coli infection, ribosome, and spliceosome (Fig. 5A). Conversely, the low-risk group exhibited enrichment in pathways related to complement and coagulation cascades, drug metabolism cytochrome P450, fatty acid metabolism, retinol metabolism, and steroid hormone biosynthesis (Fig. 5B). These findings provide valuable insights into the underlying molecular mechanisms that contribute to the differential prognostic outcomes observed between the high-risk and low-risk groups in our study.

Fig. 5
figure 5

GSEA enrichment analysis identifies KEGG pathways associated with high-risk A and low-risk groups B in the training set

Identifying the role of risk level in clinical subgroups

To further investigate whether the prognosis of high-risk and low-risk patients in different clinical subgroups differs, we performed survival analysis on patients in the high and low-risk groups within different clinical subgroups, based on gender (male, female), age (< 65, ≥ 65), stage (stage I/II, stage III/IV), and grade (grade I/II, grade III/IV). The Kaplan–Meier analysis (Fig. 6A-P) indicated that the survival time of low-risk patients was significantly longer than that of high-risk patients. Moreover, time-ROC analysis (Fig. 6A-P) revealed that the constructed 5-NKRLSig model can reliably predict the prognosis of HCC across different clinical subgroups.

Fig. 6
figure 6

Clinical application of the 5-NKRLSig model in HCC. The difference in risk score by Female A-B, Male C-D, Age < 65 E–F, Age ≥ 65 G-H, Stage I/II I-J, Stage III/IV K-L, Grade I/II M–N, Grade III/IV O-P of HCC

Establishment of nomograms in combination with clinical characteristics

To enhance the clinical utility of the 5-NKRLSig model developed in this study, we also constructed a nomogram to predict the OS of HCC patients. We conducted univariate and multivariate COX regression analyses on both clinical information and genetic models. According to the results of the multivariate COX regression analysis (Fig. 7A), Stage and risk score were identified as independent influencing factors for HCC. We developed the nomogram (Fig. 7B) based on the patient's stage and risk score, which can more accurately predict the patient's survival rates at 1, 3, and 5 years. The C-index and 95% confidence interval (CI) of the nomogram were 0.743 (0.718–0.769), indicating that the nomogram had good predictive ability for the patient's survival rates at 1, 3, and 5 years (Fig. 7D). Moreover, we drew calibration curves for the training set and testing set respectively (Fig. 7C) to evaluate the consistency between the predicted and actual risks. The calibration curves for both sets demonstrated that the OS probability predicted by the nomogram was in good agreement with the actual OS probability. Furthermore, we compared the performance of our nomogram with AJCC staging using decision curve analysis (Fig. 7E), which revealed that our nomogram was not inferior to AJCC staging in predicting OS of HCC patients.

Fig. 7
figure 7

Construction of the nomogram. A Univariate and multivariate Cox regression analysis in TCGA-LIHC. B Nomogram integrating the Risk score and Stage. C Calibration curves for predicting 1, 3and 5 years OS in the train set and test set. D Concordance index curves depicting risk scores and other clinical parameters relevant to predicting HCC patient prognosis. E DCA of the nomogram and AJCC stage

Analysis the relationship between 5-NKRLSig modal risk score and somatic mutation and TMB

In this study, we aimed to comprehensively analyze the relationship between the expression of genes related to NK cells and the prognosis of HCC patients. Specifically, we investigated the relationship between the risk score level and somatic mutation and TMB cell mutation. Our findings (Fig. 8A-B) demonstrated that the rate of TP53 somatic mutation was significantly higher in the high-risk group as compared to the low-risk group (62% vs 22%). However, we observed no significant difference in somatic mutation rates of CTNNB1 and TTN between low-risk and high-risk patients. Furthermore, we compared the TMB scores of the high-risk and low-risk groups, and the results (Fig. 8C) showed that high-risk patients had a higher TMB, which was statistically significant.

Fig. 8
figure 8

Genetic alterations and tumor microenvironment. Top 10 gene mutations in high-risk A and low-risk B groups. C The analysis of TMB scores of high-risk and high-risk groups. D Comparison of the immune score, matrix score and ESTIMATE score in high- and low-risk groups

Analysis of the tumor microenvironment between high-risk and low-risk groups

The TME constitutes the milieu in which tumor cells thrive and is mainly composed of immune cells and stromal cells. The Estimate algorithm has been developed to estimate the abundance of immune and stromal components in tumors, which can be quantified as an immune score. There have been multiple studies showing that immune and stromal cells play an important role in tumor prognosis. In this study, we employed the "Estimate" R package to calculate the immune score, matrix score, and ESTIMATE score in the high-risk group. Our results revealed that the low-risk group displayed significantly higher immune score, matrix score, and ESTIMATE score compared to the high-risk group (Fig. 8D).

Evaluation of immunotherapy, chemotherapy and target therapy based on risk score

Immunotherapy has emerged as a promising treatment option for HCC patients.To further investigate the role of risk score in immunotherapy, we utilized the TIDE algorithm to analyze the response to immunotherapy in two groups. Our results (Fig. 9A) indicate that the low-risk group has a lower TIDE score than the high-risk group, suggesting that the low-risk group is more sensitive to immunotherapy.

Fig. 9
figure 9

Application of risk score in immunotherapy, Chemotherapy and target therapy. A Prediction of immunotherapy response based on the TIDE algorithm. IPS score for immunotherapy. B CTLA4- PD1 − . C CTLA4 + PD1 − . D CTLA4 − PD1 + . E CTLA4 + PD1 + . Risk score predicts chemotherapy sensitivity. Cetuximab F, Erlotinib G, Trametinib H, XAV939 I, Docetaxel J, MLN4924 K, OSU-03012 L, YM155 M, Salubrinal N, Vorinostat O

Next, we obtained IPS data of HCC patients from the Cancer Immunome Atlas (TCIA) database and analyzed the role of risk score in IPS (Fig. 9B-E). Our results show that the median of the low-risk group was higher than that of the high-risk group, supporting our previous finding that patients in the low-risk group are more sensitive to immunotherapy.

Furthermore, we compared the efficacy of 10 immunotherapeutic drugs in the high-risk and low-risk groups. Our results show that the IC50 of Cetuximab, Erlotinib, Trametinib and XAV939 in the low-risk group was relatively higher (F ig. 9F-I), whereas the IC50 of Docetaxel, MLN4924, OSU-03012, YM155, Salubrinal and Vorinostat in the low-risk group was lower (Fig. 9J-O) compared to the high-risk group. Based on the risk score, we can better understand the immunotherapy of HCC patients and improve the accuracy of drug treatment.

Discussion

Hepatocellular carcinoma is a highly heterogeneous disease with varying tumor growth patterns and response to treatment. While treatment options such as radiotherapy, chemotherapy, and immunotherapy have provided hope for HCC patients [31], the 5-year survival rate for HCC patients remains less than 20% [32]. One of the main reasons for the poor prognosis of HCC patients is that the disease is often diagnosed at an advanced stage. The conventional risk stratification methods based on tumor size, stage and lymph node metastasis have limited predictive value for the prognosis of patients. Therefore, there is an urgent need for a novel prognostic model. NK cells, with their various cytotoxic mechanisms and immune-regulatory functions through cytokine secretion, have emerged as important players in cancer immunity [33]. However, the regulatory role of NK-related genes in hepatocellular carcinoma has not been reported. Therefore, we established a biomarker prognostic model based on NK-related genes based on TCGA and GEO database. we constructed a 5-NKRLSig and calculated a risk score. We first used the ‘sur.cutpoint’ function to calculate the cutoff point and divide all HCC patients into two prognostic subgroups: high-risk and low-risk groups. Our comprehensive analysis demonstrated that our model has a superior predictive ability when compared with traditional clinical indicators such as age, sex, histological staging, and tumor staging. Importantly, our model also showed good predictive capacity in different clinical subgroups. These findings provide a robust theoretical basis for the clinical application of our model and can significantly improve patient prognosis.

To ensure the accuracy of our model, we employed the method of external testing set (GSE14520) for verification. Despite potential differences in chip specifications, our model's ability to establish a relationship between the risk score and patient prognosis was consistent with results from the training set, suggesting the wide applicability of our approach.

The 5-NKRLSig model constructed in this study comprises IL18RAP, CHP1, VAMP2, PIK3R1, and PRKCD. Previous studies have demonstrated the close association of these five genes with inflammation and tumor occurrence and development. For instance, Wang et al. reported that miR-493-5p overexpression induces apoptosis and inhibits proliferation and migration of hepatocellular carcinoma cells by negatively regulating VAMP expression [34]. Similarly, Ai et al. found that PIK3R1 overexpression activates the PI3K/Akt/mTOR signaling pathway in hepatocellular carcinoma cells, promoting HCC initiation and progression [35]. In the context of natural killer/T-cell lymphoma cells, Lin et al. highlighted the role of IL18RAP in cell growth, where knockout of IL18RAP inhibited NKTCL cell proliferation through cell cycle arrest [36]. CHP1 has been shown to regulate plasma membrane NA( +)/H( +) exchange and play a crucial role in cellular pH regulation [37]. Li et al. reported that lactic acid and low pH can suppress the immune activity of NK cells [38], leading us to speculate that CHP1 may regulate NK cell activity by modulating pH. PRKCD has been identified as an active regulator of mitochondrial autophagy and has significant implications for B cell proliferation and NK cell activation [39]. Ke et al. [40] and Wen J et al. [41] found that PRKCD plays an essential role in the occurrence and development of cancers such as cervical cancer and esophageal squamous cell carcinoma, as well as influencing immunotherapy response. Exploring the application of PRKCD in HCC patients will be a key direction for our future research. These findings underscore the importance of the 5-NKRLSig model and its potential implications in understanding HCC pathogenesis and guiding future therapeutic strategies.

Based on our bioinformatics analysis, the 115 differential genes related to NK cells are primarily enriched in immune regulation and cell killing. We also investigated whether the model gene can be used as a biological marker of prognosis of HCC. Interestingly, the ROC curve analysis demonstrated that the expression of the model gene can accurately predict the survival status of patients, providing theoretical evidence for clinical practice. This study is the first to combine prognostic and diagnostic markers in hepatocellular carcinoma, emphasizing the continuous and inseparable relationship between cancer incidence and prognosis.

Despite the clinical significance of our study in the prognosis evaluation and treatment selection of HCC patients, there are still some limitations that need to be addressed. Our study is retrospective and relies on data from TCGA and GEO databases. Therefore, the applicability of this model as a diagnostic and treatment indicator needs to be further validated in future prospective studies.

Conclusions

In summary, we have constructed a prognostic model of HCC based on NK-related genes and have verified the effectiveness of the model in an external verification set. This provides new evidence for the evaluation of prognosis and immunotherapy of HCC patients. The study highlights the potential importance of NK cells and their related genes in the prognosis of HCC and may lead to the development of new therapeutic strategies. However, further validation in larger cohorts and prospective studies is needed to confirm the clinical utility of the model.

Availability of data and materials

The RNA-seq datasets and clinical datasets are publicly available in the following database: The TCGA-LIHC dataset was downloaded from the GDC portal (https://portal.gdc.cancer.gov/), while the GSE14520 dataset was downloaded from the GEO database (https://www.ncbi.nlm.nih.gov/geo). For further enquiries, please contact the author (Chunyang Li; 13775989791@126.com).

References

  1. Llovet JM, Castet F, Heikenwalder M, et al. Immunotherapies for hepatocellular carcinoma[J]. Nat Rev Clin Oncol. 2022;19(3):151–72.

    CAS  PubMed  Google Scholar 

  2. Vogel A, Meyer T, Sapisochin G, et al. Hepatocellular carcinoma[J]. Lancet. 2022;400(10360):1345–62.

    CAS  PubMed  Google Scholar 

  3. Hartke J, Johnson M, Ghabril M. The diagnosis and treatment of hepatocellular carcinoma[J]. Semin Diagn Pathol. 2017;34(2):153–9.

    PubMed  Google Scholar 

  4. Yang JD, Heimbach JK. New advances in the diagnosis and management of hepatocellular carcinoma[J]. BMJ. 2020;371:m3544.

    PubMed  Google Scholar 

  5. Tang J, Zhu Q, Li Z, et al. Natural killer cell-targeted immunotherapy for cancer[J]. Curr Stem Cell Res Ther. 2022;17(6):513–26.

    CAS  PubMed  Google Scholar 

  6. Kay NE. Natural killer cells[J]. Crit Rev Clin Lab Sci. 1986;22(4):343–59.

    CAS  PubMed  Google Scholar 

  7. Kumar V. Natural killer cells in sepsis: Underprivileged innate immune cells[J]. Eur J Cell Biol. 2019;98(2–4):81–93.

    CAS  PubMed  Google Scholar 

  8. Laskowski TJ, Biederstädt A, Rezvani K. Natural killer cells in antitumour adoptive cell immunotherapy[J]. Nat Rev Cancer. 2022;22(10):557–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Chu J, Gao F, Yan M, et al. Natural killer cells: a promising immunotherapy for cancer[J]. J Transl Med. 2022;20(1):240.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Riley RS, June CH, Langer R, et al. Delivery technologies for cancer immunotherapy[J]. Nat Rev Drug Discov. 2019;18(3):175–96.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Xie G, Dong H, Liang Y, et al. CAR-NK cells: a promising cellular immunotherapy for cancer[J]. EBioMedicine. 2020;59:102975.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Taketomi A, Shimada M, Shirabe K, et al. Natural killer cell activity in patients with hepatocellular carcinoma[J]. Cancer. 1998;83:58.

    CAS  PubMed  Google Scholar 

  13. Tomczak K, Czerwińska P, Wiznerowicz M. The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge[J]. Contemp Oncol (Pozn). 2015;19(1A):A68–77.

    PubMed  Google Scholar 

  14. Barrett T, Wilhite SE, Ledoux P, et al. NCBI GEO: archive for functional genomics data sets–update[J]. Nucleic Acids Res. 2013;41(Database issue):D991–5.

    CAS  PubMed  Google Scholar 

  15. Smyth GK. Linear models and empirical bayes methods for assessing differential expression in microarray experiments[J]. Stat Appl Genet Mol Biol. 2004;3:e3.

    Google Scholar 

  16. Simon N, Friedman J, Hastie T, et al. Regularization paths for Cox’s proportional hazards model via coordinate descent[J]. J Stat Softw. 2011;39(5):1–13.

    PubMed  PubMed Central  Google Scholar 

  17. Wang S, Tian S, Li Y, et al. Development and validation of a novel scoring system developed from a nomogram to identify malignant pleural effusion[J]. EBioMedicine. 2020;58:102924.

    PubMed  PubMed Central  Google Scholar 

  18. Alhamzawi R, Ali H. The Bayesian adaptive lasso regression[J]. Math Biosci. 2018;303:75–82.

    PubMed  Google Scholar 

  19. George B, Seals S, Aban I. Survival analysis and regression models[J]. J Nucl Cardiol. 2014;21(4):686–94.

    PubMed  PubMed Central  Google Scholar 

  20. Robin X, Turck N, Hainard A, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves[J]. BMC Bioinformatics. 2011;12:77.

    PubMed  PubMed Central  Google Scholar 

  21. Denny P, Feuermann M, Hill DP, et al. Exploring autophagy with Gene Ontology[J]. Autophagy. 2018;14(3):419–36.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Yu G, Wang LG, Han Y, et al. clusterProfiler: an R package for comparing biological themes among gene clusters[J]. OMICS. 2012;16(5):284–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. He Q, Yang J, Jin Y. Development and validation of TACE refractoriness-related diagnostic and prognostic scores and characterization of tumor microenvironment infiltration in hepatocellular carcinoma[J]. Front Immunol. 2022;13:869993.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Addeo A, Friedlaender A, Banna GL, et al. TMB or not TMB as a biomarker: that is the question[J]. Crit Rev Oncol Hematol. 2021;163:103374.

    PubMed  Google Scholar 

  25. Mayakonda A, Lin DC, Assenov Y, et al. Maftools: efficient and comprehensive analysis of somatic variants in cancer[J]. Genome Res. 2018;28(11):1747–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data[J]. Bioinformatics. 2016;32(18):2847–9.

    CAS  PubMed  Google Scholar 

  27. Chakraborty H, Hossain A. R package to estimate intracluster correlation coefficient with confidence interval for binary data[J]. Comput Methods Programs Biomed. 2018;155:85–92.

    PubMed  Google Scholar 

  28. Fu J, Li K, Zhang W, et al. Large-scale public data reuse to model immunotherapy response and resistance[J]. Genome Med. 2020;12(1):21.

    PubMed  PubMed Central  Google Scholar 

  29. Van Allen EM, Miao D, Schilling B, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma[J]. Science. 2015;350(6257):207–11.

    PubMed  PubMed Central  Google Scholar 

  30. Geeleher P, Cox N, Huang RS. pRRophetic: an R package for prediction of clinical chemotherapeutic response from tumor gene expression levels[J]. PLoS ONE. 2014;9(9):e107468.

    PubMed  PubMed Central  Google Scholar 

  31. Falanga A, Schieppati F, Russo L. Pathophysiology 1. Mechanisms of thrombosis in cancer patients[J]. Cancer Treat Res. 2019;179:11–36.

    CAS  PubMed  Google Scholar 

  32. Chidambaranathan-Reghupaty S, Fisher PB, Sarkar D. Hepatocellular carcinoma (HCC): epidemiology, etiology and molecular classification[J]. Adv Cancer Res. 2021;149:1–61.

    CAS  PubMed  Google Scholar 

  33. Huntington ND, Cursons J, Rautela J. The cancer-natural killer cell immunity cycle[J]. Nat Rev Cancer. 2020;20(8):437–54.

    CAS  PubMed  Google Scholar 

  34. Wang G, Fang X, Han M, et al. MicroRNA-493-5p promotes apoptosis and suppresses proliferation and invasion in liver cancer cells by targeting VAMP2[J]. Int J Mol Med. 2018;41(3):1740–8.

    CAS  PubMed  Google Scholar 

  35. Ai X, Xiang L, Huang Z, et al. Overexpression of PIK3R1 promotes hepatocellular carcinoma progression[J]. Biol Res. 2018;51(1):52.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Lin GW, Xu C, Chen K, et al. Genetic risk of extranodal natural killer T-cell lymphoma: a genome-wide association study in multiple populations[J]. Lancet Oncol. 2020;21(2):306–16.

    CAS  PubMed  Google Scholar 

  37. Kanazawa H, Mitusi K, Matsushita M. Na+/H+ antiporters from bacteria to human: ion transport mechanism and reguratory factors [J. Seikagaku. 2007;79(6):569–78.

    CAS  PubMed  Google Scholar 

  38. Li Q, Shi Z, Ou M, et al. pH-labile artificial natural killer cells for overcoming tumor drug resistance[J]. J Control Release. 2022;352:450–8.

    CAS  PubMed  Google Scholar 

  39. Sharifinejad N, Azizi G, Behniafard N, et al. Protein kinase C-Delta defect in autoimmune lymphoproliferative syndrome-like disease: first case from the national iranian registry and review of the literature[J]. Immunol Invest. 2022;51(2):331–42.

    CAS  PubMed  Google Scholar 

  40. Ke G, Liang L, Yang JM, et al. MiR-181a confers resistance of cervical cancer to radiation therapy through targeting the pro-apoptotic PRKCD gene[J]. Oncogene. 2013;32(25):3019–27.

    CAS  PubMed  Google Scholar 

  41. Wen J, Hu Y, Liu Q, et al. miR-424 coordinates multilayered regulation of cell cycle progression to promote esophageal squamous cell carcinoma cell proliferation[J]. EBioMedicine. 2018;37:110–24.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors gratefully acknowledge contributions from the TCGA and GEO databases.

Funding

This research was funded by Prevention and Treatment of Major Infectious Diseases Such as AIDS and Viral Hepatitis (2018ZX10302206-003–010) and Establishment of Predictive Indexes and Models for Severe SARSCoV-2 Infection(20KJB320012).

Author information

Authors and Affiliations

Authors

Contributions

Deyang Xi was primarily responsible for the study design, data analysis, and manuscript writing, while Jialu Wang and Yinshuang Yang oversaw data collection and manuscript revisions. Xuebing Yan, Fang Ji, and Chunyang Li provided guidance in writing the article and contributed equally to finalizing the manuscript.

Corresponding authors

Correspondence to Chunyang Li or Xuebing Yan.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1.

NK-related genes from MSigDB database.

Additional file 2: Table S2.

NK-related genes from ImmPort Portal website.

Additional file 3:  Table S3.

Univariate COX regression results and AUC.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xi, D., Wang, J., Yang, Y. et al. A novel natural killer-related signature to effectively predict prognosis in hepatocellular carcinoma. BMC Med Genomics 16, 211 (2023). https://doi.org/10.1186/s12920-023-01638-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12920-023-01638-0

Keywords