Skip to main content
Erschienen in: BMC Cancer 1/2024

Open Access 01.12.2024 | Research

G6PD and machine learning algorithms as prognostic and diagnostic indicators of liver hepatocellular carcinoma

verfasst von: Fei Li, Boshen Wang, Hao Li, Lu Kong, Baoli Zhu

Erschienen in: BMC Cancer | Ausgabe 1/2024

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

Liver Hepatocellular carcinoma (LIHC) exhibits a high incidence of liver cancer with escalating mortality rates over time. Despite this, the underlying pathogenic mechanism of LIHC remains poorly understood.

Materials & methods

To address this gap, we conducted a comprehensive investigation into the role of G6PD in LIHC using a combination of bioinformatics analysis with database data and rigorous cell experiments. LIHC samples were obtained from TCGA, ICGC and GEO databases, and the differences in G6PD expression in different tissues were investigated by differential expression analysis, followed by the establishment of Nomogram to determine the percentage of G6PD in causing LIHC by examining the relationship between G6PD and clinical features, and the subsequent validation of the effect of G6PD on the activity, migration, and invasive ability of hepatocellular carcinoma cells by using the low expression of LI-7 and SNU-449. Additionally, we employed machine learning to validate and compare the predictive capacity of four algorithms for LIHC patient prognosis.

Results

Our findings revealed significantly elevated G6PD expression levels in liver cancer tissues as compared to normal tissues. Meanwhile, Nomogram and Adaboost, Catboost, and Gbdt Regression analyses showed that G6PD accounted for 46%, 31%, and 49% of the multiple factors leading to LIHC. Furthermore, we observed that G6PD knockdown in hepatocellular carcinoma cells led to reduced proliferation, migration, and invasion abilities. Remarkably, the Decision Tree C5.0 decision tree algorithm demonstrated superior discriminatory performance among the machine learning methods assessed.

Conclusion

The potential diagnostic utility of G6PD and Decision Tree C5.0 for LIHC opens up a novel avenue for early detection and improved treatment strategies for hepatocellular carcinoma.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-024-11887-6.
Fei Li and Boshen Wang these authors have contributed equally to this work and share first authorship.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
LIHC
Liver Hepatocellular carcinoma
G6PD
Glucose-6-phosphate Dehydrogenase gene
PPP
The pentose phosphate pathway
NADPH
Nicotinamide adenine dinucleotide phosphate
TCGA
The Cancer Genome Atlas database
ICGC-LIRI-JP cohort
International Cancer Genome Consortium database
GEO
The Gene Expression Omnibus database
IHC
Immunohistochemistry
HPA
The protein atlas
OS
Overall survival
PFS
Progression-free survival
TMB
Tumor mutation load
ROC
The receiver operating characteristic curve
CDC20
Homo sapiens cell division cycle 20 gene
CEP55
Centrosomal protein 55kda gene
TRIP13
Homo sapiens thyroid hormone receptor interactor 13 gene
MYBL2
Homo sapiens v-myb avian myeloblastosis viral oncogene homolog-like 2
GO
The Gene Ontology
KEGG
Kyoto Encyclopedia of Genes and Genomes pathway analysis
GSEA
Gene-set enrichment analysis
IC50
Half-maximal inhibitory concentration
TCIA
The Cancer Immunoomics Atlas
HR
Hazard ratio
AUC
The area under the curve
ICB
Immune checkpoint blockade
PD1
Programmed cell death protein 1
CTLA4
Cytotoxic T-lymphocyte-associated protein 4
TME
Tumor microenvironment

Introduction

Liver Hepatocellular carcinoma (LIHC) has a very high incidence of liver cancer. Meanwhile, liver cancer ranks third among cancer deaths [1]. In 2012, there were more than 700,000 confirmed cases of Hepatocellular carcinoma in China, and the number of death cases also exceeded 700,000. The incidence and mortality of LIHC continue to rise worldwide, including in China [13]. Because the clinical symptoms of LIHC are not easily detected, 80% of patients are not well treated. Better treatment options are urgently needed for patients with LIHC [4].
G6PD is a very important biomarker in the pentose phosphate pathway (PPP), which is a precursor for nicotinamide adenine dinucleotide phosphate (NADPH) production in tumor cells [5]. Research has shown that high expression of G6PD is strongly correlated to the poor clinical prognosis of bladder cancer, lung cancer, and breast cancer [68]. Animal studies have shown a relationship between G6PD and precancerous lesions in rat liver. The high expression of G6PD increased the incidence of precancerous lesions, and the number and volume of LIHC cells were also higher than those in the low expression group of G6PD [9]. Nevertheless, the specific process of G6PD participating in LIHC needs to be further studied.
In recent years, clinicians have applied machine learning to cancer diagnosis and prognosis prediction, which has significantly improved the survival rate of cancer patients [10, 11]. Through machine learning, clinicians can use big data analysis to analyze a large amount of clinical data, accurately predict the prognosis of patients, and facilitate the finding of feasible treatment methods and symptomatic treatment [12]. Therefore, a large number of Machine learning algorithms have been developed, such as Bayesian Classifier, Neural network algorithm, Support vectors machine and Decision Tree C5.0 [1315]. However, the application performance of machine learning in LIHC has not been validated.
In this study, data analysis investigated the possible mechanism between G6PD and LIHC. We concluded that the expression level of G6PD could be regarded as a prognostic and diagnostic criterion for LIHC patients. Gene correlation studies, immunoassays, and drug sensitivity analysis have provided new ideas for treating LIHC. Finally, we compared the ability of four machine learning algorithms to distinguish between LIHC and para-cancerous tissues.

Materials and methods

Datasets acquisition

We obtained related clinical information and the RNA sequencing data of LIHC patients from the Cancer Genome Atlas (TCGA) database (https://​portal.​gdc.​cancer.​gov/​projects/​TCGA-LIHC, dbGaP Study Accession:phs000178, 26/7/2022, human), the International Cancer Genome Consortium (ICGC-LIRI-JP cohort, https://​dcc.​icgc.​org/​projects/​LIRI-JP, 28/8/2022, human) database and the Gene Expression Omnibus (GEO) database (http://​www.​ncbi.​nlm.​nih.​gov/​geo/​,28/​8/​2022, human). The TCGA database was elected as a training cohort and the databases from ICGC (LIRI-JP) and GEO (GSE14520, GSE20140, GSE62232, GSE84005) as validation cohorts. It was verified that the G6PD gene expression level data obeyed a normal distribution, subsequently, we matched the clinical data and gene expression of each patient in each database, and then classified those with G6PD expression above the average level as high expression group, and those with G6PD expression below the average level as low expression group. Figure 1 is the flow chart of this experimental study.

Differential expression analysis and verification of protein expressions of G6PD

The Bioconductor package edgeR was employed to identify the differential expression levels of G6PD in different cancer and normal tissues. Immunohistochemistry (IHC) is an approach to detecting the distribution and expression of relative proteins based on the specific binding of antigens to antibodies. We captured the prognosis-related protein expression profiles of the G6PD gene from the Protein Atlas (HPA, https://​www.​proteinatlas.​org/​) database for hepatocellular carcinoma tissues and normal tissues.

Correlation analysis between G6PD expression level and clinical characteristics

Cox regression analyses were fulfilled to value the connection between traditional clinical characteristics (including Age, Gender, Grade, TNM stage, and Tumor stage I-IV) and G6PD expression level, which provided the basis for the establishment of the Nomogram.

Nomogram construction and verification

A nomogram is a method to predict the clinical outcome of LIHC patients. R software was used to construct a Nomogram including Age, Gender, Grade, Stage, and G6PD. In addition, the receiver operating characteristic (ROC) curve and the calibration curves were drawn to score the prediction accuracy of the Nomogram.

Assessment of the importance of G6PD in LIHC

To further assess the value of G6PD as a clinical prognostic indicator of hepatocellular carcinoma, three machine learning models, Adaboost regression, Catboost Regression, and Gbdt Regression, were utilized to appraise the importance of G6PD in the prognosis of patients with LIHC.

Gene correlation analysis

The R software was utilized to probe the correlation between CDC20, CEP55, TRIP13, MYBL2 and G6PD. Gene correlation analysis was used to explore the valuable genes that may be related to G6PD, this could point to the possibility that these genes may have biologically similar functions, participate in the same pathways, or be similarly regulated.

GO, KEGG, and GSEA analysis

In order to understand the cellular component, molecular function, and biological processes of G6PD in humans, the Gene Ontology (GO) was performed by the R software. Meanwhile, the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene-set enrichment analysis (GSEA) were implemented to comprehend the related signaling pathways of G6PD.

Drug sensitivity assessment

Drug sensitivity data for LIHC patients is available in the Cancer Drug Sensitivity Genomics Database (https://​www.​cancerrxgene.​org/​). the drug response was presented by the half-maximal inhibitory concentration (IC50) which used the R software. The results have appeared in box plots.

Calculation of tumor mutational burden

Tumor mutation load (TMB) is an index used to reflect the ability and degree of the tumor to produce new antigens, which can indirectly predict the effect of immunotherapy on all kinds of tumors. A high TMB index indicates a better clinical immunotherapy effect. The TMB values were calculated using Perl scripts, and the above results were displayed in a scatter diagram.

Immunological analysis

To understand the relationship between Tumor Microenvironment (TME) and G6PD, we plotted the violin of Stromal score, the immune scores, and the estimate scores in two groups of LIHC patients with high and low G6PD expression. We used the CIBERSORT algorithm to calculate differences in 22 tumor-infiltrating immune cells in LIHC patients with high and low G6PD expression levels. CTLA4 and PD-1 Immune checkpoint is commonly used in immunotherapy. Finally, we obtained the immunophenotype (IPS) of LIHC from the LIHC project of Cancer Immunoomics Atlas (TCIA, https://​www.​tcia.​at/​home) to predict the response to immunotherapy in the group with high and low G6PD expression.

Survival analysis

Clinical data from TCGA database, GEO (GSE14520) database and ICGC database were analyzed for survival by Kaplan–Meier curve survival analysis using R software. Overall survival (OS) and progression-free survival (PFS) were analyzed for TCGA data, and OS was analyzed for GEO (GSE14520) database and ICGC database.

Feasibility of machine learning algorithms in clinical prognosis of LIHC patients

In order to accurately predict a patient's prognosis, many machine learning algorithms have been developed in recent years. We selected four machine learning algorithms for comparison: Bayesian classifier, Neural network algorithm, Support vectors machine and Decision Tree C5.0.

Cell culture and transfection

We purchased WRL68 liver normal cells from Shanghai Fuheng Biotechnology Co., LTD. (Fuheng, Shanghai, China), LI-7, SNU-398, SNU-449, SK-HEP-1 Liver cancer cells were acquired from Guangzhou Cellcook Biotechnology Co., LTD (Cellcook, Guangzhou, China). Dulbecco’s modified Eagle’s medium (DMEM; Gibco, Shanghai, China) was used to culture WRL68. RPMI 1640’s modified Eagle’s medium was used to culture SNU-398 (RPMI 1640 modified; Gibco, Shanghai, China). Meanwhile, we cultured LI-7, SNU-449, SK-HEP-1 cells using RPMI 1640 medium. siRNA-G6PD and non-targeting control siRNA (NC-siRNA) were purchased from Ribobio Biotechnology Co., LTD. (Ribobio, Guangzhou, China). The sequence of siRNA-G6PD was TCCTCTATGTGGAGAATGA. The stareffect II transfection reagent (GenStar, Beijing, China) was bound to siRNA-G6PD or NC-SIRRNA for 10 min and transfected into LI-7 or SNU-449 cells. The solution was changed after 6 h, and cells were collected after 48 h.

Quantitative real-time PCR (qRT-PCR)

Total RNA from WRL68, LI-7, SNU-398, SNU-449, SK-HEP-1 cells was extracted using RNA-easy Isolation Reagent (Vazyme, Nanjing, China). For qRT-PCR, the following primers were used: human G6PD, 5′-AAGAACGTGAAGCTCCCTGA-3′ (Forward) and 5′-AATATAGGGGATGGGCTTGG-3′ (Reverse); human β-actin, 5′-GGAAATCGTGCGTGACAT-3′ (Forward) and 5′-GGTGATGACCTGGCCGTT-3′ (Reverse). Relative expression of G6PD was analyzed using the 2 − ΔΔCT method.

Western blots

The RIPA buffer (GenStar, Beijing, China) was used to lyse the cells and obtain the protein, which was then measured by BCA and a protein sample was made. The proteins were transferred into the membrane by electrophoresis and membrane transfer steps. Then it was sealed with a sealing solution for 1h, incubated with primary antibody for 8h-12h, incubated with secondary antibody for 1h, and developed by chemiluminescence. Primary antibodies were: Recombinant Anti-Glucose 6 Phosphate Dehydrogenase antibody [EPR20668] (Abcam, ab210702), and Anti-beta Actin [mAbcam 8226] (Abcam, ab8226).

Cell viability

LI-7 and SNU-449 were inoculated into 96-well plates with 2000 cells/well and incubated for 24 h to attach to the wall. 48 h after transfection with siRNA-G6PD or NC-siRNA, fresh medium and 10 μL cell counting Kit 8 (CCK-8, Beyotime, China) were added to each well, and after incubation at 37°C for 1 h, absorbance at 450 nm was obtained by spectrophotometer and cell viability was analyzed.

Transwell invasion and migration assay

Cell chambers were placed in 24-well plates and invasion and migration experiments were performed with or without BD Matrigel TM (BD Bioscience, USA). Migration experiment: 0.3 mL serum-free medium containing 2 × 105 cells was added to the upper part of the cell chamber and 0.7ml medium containing 10% serum was added to the lower part of the cell chamber for 12 and 24 h. Invasion experiment: 0.3 mL serum-free medium containing 4 × 105 cells was added to the upper part of the cell chamber and 0.7ml medium containing 10% serum was added to the lower part of the cell chamber for 24 and 48 h. First, cells were fixed with 4% paraformaldehyde for 10 min, then stained with 0.5% crystal violet for 30 min, cleaned and wiped away the excess cells.

Statistical analysis

All statistical data were analyzed using R software (4.0.5). Univariate and multivariate Cox regression analyses were applied to appraise the association between G6PD and different clinical features. Log-rank test and Kaplan–Meier analysis were used to assess the effect of G6PD expression level on the survival status of LIHC patients. ROC curve was applied to evaluate the performance of the Nomogram and the machine learning algorithm. the statistical significance was installed at P < 0.05.

Results

The expression level of G6PD

First of all, we analyzed the discrepancy of G6PD expression levels between different cancer tissues and normal tissues, and there were significant differences in G6PD in 17 tissues. LIHC was selected as the research direction of this paper (Fig. 2A). Then, we contrasted the expression levels of G6PD between normal Hepatocellular tissue and LIHC tissue. The expression level of G6PD in LIHC tissue was high, and LIHC tissues of the same person tended towards higher G6PD expression levels than normal tissues (Fig. 2B-C). the results of the ICGC and GSE databases were consistent with those of the TCGA database (Fig. S1A-D). the HPA database showed that G6PD was highly expressed in LIHC tissues (Fig. S1E-F). Subsequently, we analyzed the G6PD gene expression in WRL68, LI-7, SNU-398, SNU-449, SK-HEP-1 cells, and showed that compared with normal liver cells, the G6PD expression level of liver cancer cells was significantly increased, and the expression level of LI-7 and SNU-449 was the highest (Fig. 2D). the results of Western blots were the same as those of PCR (Fig. 2E-F).

Effect of G6PD expression levels on prognosis

The Kaplan–Meier curve plotted had been described by the log-rank test, which showed that the high expression level of G6PD patients had poor OS compared with the low expression level of G6PD patients (p < 0.01, Fig. 3A). Meanwhile, In the PFS curve, the PFS of patients with a high expression level of G6PD was observably lower than that of patients with a low expression level (p < 0.01, Fig. 3B). The results of GEO database and ICGC database were consistent with the TCGA database (Fig. 3C-D). Overall Survival odds ratios (OR) for the TGCA, GEO, and ICGC databases, respectively 2.03 (1.46–2.83), 1.56 (1.01–2.41), 2.96 (1.50–5.86).

Establishment and evaluation of the prognostic nomogram

First, we explored the relationship between G6PD and Age, Gender, Grade, Tumor stage and TNM stage, and found that G6PD expression level was dramatically different in Grade, Tumor stage and T stage, respectively (Fig. S2A-G). the heatmap was described, which brings together a variety of factors (Fig. 4A). To establish a prognostic nomogram consisting of multiple clinical features as a model for predicting the prognosis of LIHC, G6PD, Age, Gender, Grade, Tumor stage and TNM stage was taken into account. At the same time, univariate and multivariate Cox regression was carried out. According to univariate Cox regression, G6PD (p < 0.001), Stage (p < 0.001) and the difference in the T stage part were significant and statistically significant (Fig. 4B). As shown by multivariate analysis, the G6PD (HR = 1.338, 95% CI = 1.177–1.521, p < 0.001) and Tumor stage (HR = 1.588, 95% CI = 1.279–1.970, p < 0.001) were considered as the critical prognostic factors that forecasted the OS for LIHC (Fig. 4C). Then, we built a fresh nomogram to predict the one-, three-, and five-year OS rates of LIHC patients (Fig. 4D). Each patient corresponds to a score through the nomogram, and Patients with high scores had poorer outcomes than those with low scores. Survival calibration curves including one-, three- and five-year survival have been established. A scatter converging to a 45° diagonal would indicate a better model fit. The calibration curves show that our model predicts well (Fig. 4E). Finally, we evaluated the performance of the nomogram, and the results showed that the area under the curve (AUC) values of one-, three-, and five-year survival were 0.730, 0.635, and 0.612, respectively (Fig. 4F).

Evaluation of the predictive power of Adaboost regression, Catboost regression and Gbdt regression

The results of Adaboost regression, Catboost Regression and Gbdt Regression showed that the prediction results fitted well with the actual value, and G6PD occupied a significant proportion of the clinical prognosis of LIHC. (Fig. 5A-C), The evaluation indexes of the three machine learning algorithms are shown in Table 1.
Table 1
Adaboost regression, Catboost regression, Gbdt regression statistical results
algorithms
indexes
Training set
Cross-validation set
Adaboost
MSE
0.036
5.323
RMSE
0.189
2.281
MAE
0.064
1.644
Catboost
MSE
0.329
4.361
RMSE
0.574
2.058
MAE
0.434
1.542
Gbdt
MSE
0.007
6.723
RMSE
0.086
2.571
MAE
0.041
1.963

G6PD knockout inhibited the proliferation, migration and invasion of hepatocellular carcinoma cells

To confirm the effect of G6PD on liver cancer cells, we transfected genOFF st-h-G6PD into LI-7 and SNU-449 liver cancer cells using transfection reagents. PCR experiments showed that G6PD gene was knocked down in both cells (Fig. 6A), and the results of western blots experiments were shown in Fig. (Fig. 6C). The cell proliferation experiment showed that the low expression of G6PD significantly inhibited the activity of SNU-449 and LI-7 cells, and the proliferation rate decreased significantly compared with normal and NC cells (Fig. 6B). Cell migration and invasion experiments showed that the migration and invasion ability of cells with low G6PD expression was weakened (Fig. S3A-B, Fig. S4A-B).

Gene correlation analysis

For investigating the correlation between G6PD and other genes, Correlation Analysis was performed by R package. G6PD was positively correlated with CDC20, CEP55, TRIP13, MYBL2, and the correlation was statistically significant, respectively, R = 0.62, 0.61, 0.6, 0.6 (Fig. 7A-E).

GO, KEGG and GSEA analysis

GO, KEGG and GSEA enrichment analysis were implemented to comprehend the biological processes, cellular components, molecular function and related signaling pathways of G6PD. The GO analysis results showed that the biological processes of G6PD primarily assembled in immunoglobulin and B cell-mediated immune response, humoral immune response, immunoglobulin complement activation. The cellular component of G6PD mainly focused on external side of plasma membrane, neuronal cell body, synaptic membrane, and plasma membrane signaling receptor. The molecular function of G6PD mainly enriched in channel activity, antigen binding, passive transmembrane transporter activity and ion channel activity (Fig. S5A, Fig. 8A). The KEGG analysis results indicated that the signal pathway of G6PD mainly focused on Neuroactive ligand − receptor interaction pathway, Cell adhesion molecules pathway, PI3K − Akt signaling pathway and Cytokine − cytokine receptor interaction pathway (Fig. S5B). GSEA enrichment analysis shown that the pathway of the low expression group of G6PD was mainly enriched in Complement and Coagulation Cascades, Fatty Acid Metabolism, Glycine Serine and Threonine Metabolism, Peroxisome, Primary Bile Acid Biosynthesis (Fig. 8B).

The connection between G6PD expression and immune system

To further research the connection between the expression level of G6PD in LIHC patients and immune status, the connection between G6PD and the TME score, immune cell, immune checkpoint was carried out. As shown, compared with the high expression level of G6PD, the TME score of immune score and the estimated score were lower in the low expression level of G6PD, and the difference was statistically significant, the difference in stromal cell score was not statistically significant (Fig. 9A). Analysis of immune cell subsets suggested that immune cell scores, including B cells naïve, Macrophages M0, Monocytes, T cells CD4 memory resting were markedly different between the low- and high-expression level of G6PD groups, respectively, R = -0.43, 0.43, -0.23, -0.35, P < 0.05 (Fig. 9B, Fig. S6A-E). Then, Fig. 9C showed that the relationship between G6PD and tumor mutation burden, R = 0.11, p < 0.05. this indicated that LIHC patients with high G6PD expression levels had high TMB scores and good immunotherapy effects. Fig. S6F showed the coefficient diagram of a relationship between G6PD and immune checkpoint. At the same time, we analyzed routine immune checkpoints including PD1 and CTLA4. Whether the expression level of G6PD was high or low, the mean IPS showed no significance. (Fig. 9D-G).

Drug sensitivity analysis

To explore the clinical significance of G6PD, the drug sensitivity of LIHC was predicted by R software. The consequences revealed that the high expression of the G6PD group was more sensitive to 2 kinds of drugs, including Phenformin and Erlotinib (Fig. 10A-B). In comparison, the low expression of the G6PD group was more sensitive to Sorafenib (Fig. 10C).

Prediction of clinical prognosis of LIHC patients by four machine learning methods

To predict the clinical prognosis of patients with LIHC, we selected four machine learning algorithms, including the Bayesian Classifier, Neural network algorithm, Support vectors machine, and Decision Tree C5.0. The research results showed that in the training group, the correct rates of the Bayesian Classifier, Neural network algorithm, Support vectors machine, and Decision Tree C5.0 were 83.04%,83.74%, 91.70%, and 93.08%, respectively (Fig. 11A). In the test group, the correct rates were 83.33% 84.06%, 88.41% and 87.68%, respectively. (Fig. 11B). At the same time, we evaluated the prediction performance of the four algorithms, and the evaluation outcomes revealed that in the training group, the AUC of Bayesian Classifier, Neural network algorithm, Support vectors machine and Decision Tree C5.0 were 0.845,0.773,0.941,0.987, respectively (Fig. 11C). In the test group, the AUC were 0.738,0.706,0.84 and 0.929, respectively (Fig. 11D).

Discussion

Baba et al. found that the expression levels of G6PD may be strongly associated with the growth of precancerous and neoplastic lesions [9]. Dore et al. found that G6PD deficiency significantly reduced the incidence of hepatocellular carcinoma in a case–control study [16]. However, the mechanism between G6PD expression and LIHC remains unclear. In this study, we explored the correlation between G6PD and LIHC, and the possible signaling pathways leading to LIHC by bioinformatics analysis. We found a statistically prominent difference in G6PD expression level between cancer tissues and para-carcinoma tissues of patients with LIHC. Immunohistochemical results obtained from the human protein atlas confirmed that G6PD expression levels were significantly increased in LIHC. We also confirmed this by PCR and WB experiments on one type of normal liver cells and four types of liver cancer cells.
Tumor mutation burden has long been used as a biomarker for tumor prediction [17]. Consequently, we investigated the tumor mutation burden of G6PD, and its prognostic impact on hepatocellular carcinoma and evaluated the plausibility of the impact. The expression levels of G6PD were positively related to the tumor mutation burden, indicating that the group with high G6PD expression levels had a poor immunotherapy outcome and a poor prognosis, which was the same as the research result of Cao [18]. We also found that elevated G6PD expression resulted in poor OS and PFS in LIHC. We studied the influence of G6PD on the proliferation, migration and invasion of LI-7 and SNU-449 by knockout of G6PD gene. The results showed that G6PD knockdown significantly reduced the proliferation rate, migration and invasion ability of LI-7 and SNU-449. Cao et al. showed that the up-regulation of G6PD promoted the survival, metastasis, and invasion of HepG2 cells [18]. Lu et al. showed that the high expression of G6PD was significantly connected to the metastasis and poor prognosis of LIHC, and the migration and invasion of LIHC cells were inhibited when the G6PD gene was knocked out [19]. Li et al. showed that the low expression of G6PD observably prolonged the orthotopic tumor model mice's survival time. When the high expression of G6PD resumed, the tumor growth, tumor size, volume, and weight were restored [20]. High expression of G6PD produces high levels of NADPH via the PPP pathway, as well as an increase in glutathione, which in turn counteracts oxidative stress and DNA damage, which promotes immune escape, tumor progression, and drug resistance [5]. Therefore, high G6PD expression is closely associated with poor LIHC prognosis, and high G6PD expression promotes the migration and invasive ability of LIHC cells through reorganization of the glucose metabolism pathway.
We found a positive connection between the expression level of G6PD and CDC20, CEP55, TRIP13, MYBL2 in LIHC patients. Several scientists pointed out that CDC20, CEP55, TRIP13, MYBL2 were overexpressed in hepatocellular carcinoma, compared with adjacent normal tissues [2124]. However, their interactions in promoting hepatocellular carcinoma are not investigated. GO, KEGG and GSEA analysis was executed to probe the possible involvement of G6PD in hepatocellular carcinoma formation. Our study showed that G6PD was mainly involved in the immune system and signaling pathway (e.g., PI3K-Akt signaling pathway, Cell adhesion molecules pathway) in promoting the occurrence and development of LIHC. Cheng et al. indicated that there was an interaction between the PI3K-Akt signaling pathway and G6PD, which promoted the development of cancer [25].
By immune analysis, we discovered that the expression level of G6PD was positively related to Macrophages M0, and negatively correlated with B cells naive, Monocytes, and T cells CD4 memory resting. Tekin et al. showed that macrophage M0 had anti-tumorigenic activity and impaired the growth of pancreatic cancer cells through TNF-α secretion [26]. Therefore, the increase of macrophage M0 may have an inhibitory effect on LIHC. Nevertheless, there is no research on the correlation between Macrophages M0, B cells naive, Monocytes, T cells CD4 memory resting and hepatocellular carcinoma. Immune checkpoint analysis showed that G6PD was positively associated with most immune checkpoints. Still, the results of the risk score values of immune checkpoint blockade (ICB) suggested that G6PD may not played a role in predicting the risk score model of PD1 and CTLA4 treatment response.
The drug sensitivity test found that the group with high G6PD expression level was sensitive to Phenformin and Erlotinib, and the group with low G6PD expression level was sensitive to Sorafenib. Phenformin is a diabetes treatment but causes a fatal lactic acidosis reaction [27]. Recently, a study has suggested that Phenformin has an anti-tumor effect and can inhibit the glucose metabolism of tumor cells [28]. Huang et al. found that the combination of Phenformin and Sorafenib showed a synergistic ability to restrain the proliferation and migration of LIHC [29]. Erlotinib is a treatment for non-small cell lung cancer [30]. Zheng et al. indicated that the efficacy of Erlotinib in the treatment of hepatocellular carcinoma was unclear, but they found that 2-methoxy estradiol enhanced the inhibitory effect of Erlotinib on hepatocellular carcinoma [31]. Phase 3 clinical trial showed that the combination of sorafenib and erlotinib had little effect on the survival rate of patients with advanced LIHC [32]. Sorafenib is the gold standard for the treatment of advanced hepatocellular carcinoma. However, due to individual heterogeneity, the resistance to Sorafenib has gradually attracted attention [33]. High glucose metabolism due to GLUT1/ALDOB/G6PD axis expression promotes drug resistance in pancreatic cancer, and inhibition of the GLUT1/ALDOB/G6PD axis may serve as a target for drug resistance therapy [34]. Thus, the high level of G6PD may be an important reason for Sorafenib resistance.
The Bayesian classifier is an artificial intelligence widely used in medical decision-making. Junath et al. applied it to the prognosis diagnosis of breast cancer and considered it feasible and effective [35]. Bo et al. believed that using support vector machines could effectively predict the time and location of cancer recurrence, and the effect was better than that of the neural network algorithm, which was consistent with our research results [36]. Noh et al. trained the Decision Tree C5.0 decision tree classifier by adding 7 histological features to predict the prognosis of patients with advanced gastric cancer and concluded that the model had a more accurate prediction effect [37]. In this study, TCGA database data was used to compare the performance of these four machine-learning algorithms. We believe that Decision Tree c5.0 can predict the prognosis of LIHC patients better than other algorithms.
Of course, there are still many shortcomings in this study. First, the data in this study come from a public database, and we don’t supervise the collection of data, so the reliability of the data is not clear. Second, the research results are only obtained through data analysis, and the credibility of the results needs to be demonstrated through experimental research.

Conclusion

In this investigation, we conducted a comprehensive assessment of G6PD's clinical value as a diagnostic and prognostic indicator for LIHC. Our study results demonstrated the potential of G6PD to enhance LIHC diagnosis, enabling early detection and prompt treatment for affected patients. Through meticulous cell experiments, we confirmed the substantial impact of low G6PD expression on the proliferative activity, migration, and invasion of liver cancer cells. Machine learning shows Decision Tree c5.0 has a better ability to predict the clinical prognosis of LIHC patients. In conclusion, G6PD, along with Decision Tree c5.0, holds promise as a valuable tool for predicting the prognosis of LIHC patients and offering diagnostic insights to clinicians.

Acknowledgements

We thank all the authors for their contributions to this study.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare no competing interests.
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.

Publisher’s Note

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

Supplementary Information

Literatur
1.
2.
Zurück zum Zitat Henley SJ, Ward EM, Scott S, et al. Annual report to the nation on the status of cancer, part I: national cancer statistics. Cancer. 2020;126(10):2225–49.CrossRefPubMed Henley SJ, Ward EM, Scott S, et al. Annual report to the nation on the status of cancer, part I: national cancer statistics. Cancer. 2020;126(10):2225–49.CrossRefPubMed
3.
Zurück zum Zitat Llovet JM, Castet F, Heikenwalder M, et al. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol. 2022;19(3):151–72.CrossRefPubMed Llovet JM, Castet F, Heikenwalder M, et al. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol. 2022;19(3):151–72.CrossRefPubMed
4.
Zurück zum Zitat Yin ZY, Li XW. Immunotherapy for hepatocellular carcinoma. Cancer Lett. 2020;470:8–17.CrossRef Yin ZY, Li XW. Immunotherapy for hepatocellular carcinoma. Cancer Lett. 2020;470:8–17.CrossRef
6.
Zurück zum Zitat Chen XY, Xu ZJ, Zhu ZJ, et al. Modulation of G6PD affects bladder cancer via ROS accumulation and the AKT pathway in vitro. Int J Oncol. 2018;53(4):1703–12.PubMed Chen XY, Xu ZJ, Zhu ZJ, et al. Modulation of G6PD affects bladder cancer via ROS accumulation and the AKT pathway in vitro. Int J Oncol. 2018;53(4):1703–12.PubMed
7.
Zurück zum Zitat Forteleoni G, Argiolas L, Farris A, et al. G6PD deficiency and breast-cancer. Tumori. 1988;74(6):665–7.CrossRefPubMed Forteleoni G, Argiolas L, Farris A, et al. G6PD deficiency and breast-cancer. Tumori. 1988;74(6):665–7.CrossRefPubMed
8.
Zurück zum Zitat Wang JX, Yuan WJ, Chen ZK, et al. Overexpression of G6PD is associated with poor clinical outcome in gastric cancer. Tumor Biol. 2012;33(1):95–101.CrossRef Wang JX, Yuan WJ, Chen ZK, et al. Overexpression of G6PD is associated with poor clinical outcome in gastric cancer. Tumor Biol. 2012;33(1):95–101.CrossRef
9.
Zurück zum Zitat Baba M, Yamamoto R, Iishi H, et al. Role of glucose-6-phosphate-dehydrogenase on enhanced proliferation of pre-neoplastic and neoplastic-cells in rat-liver induced by n-nitrosomorpholine. Int J Cancer. 1989;43(5):892–5.CrossRefPubMed Baba M, Yamamoto R, Iishi H, et al. Role of glucose-6-phosphate-dehydrogenase on enhanced proliferation of pre-neoplastic and neoplastic-cells in rat-liver induced by n-nitrosomorpholine. Int J Cancer. 1989;43(5):892–5.CrossRefPubMed
10.
Zurück zum Zitat Huang SG, Yang J, Fong S, et al. Artificial intelligence in cancer diagnosis and prognosis: opportunities and challenges. Cancer Lett. 2020;471:61–71.CrossRefPubMed Huang SG, Yang J, Fong S, et al. Artificial intelligence in cancer diagnosis and prognosis: opportunities and challenges. Cancer Lett. 2020;471:61–71.CrossRefPubMed
11.
Zurück zum Zitat Feng SJ, Wang JH, Wang LH, et al. Current status and analysis of machine learning in hepatocellular carcinoma. J Clin Translatl Hepatol. 2023;11(5):1184–91. Feng SJ, Wang JH, Wang LH, et al. Current status and analysis of machine learning in hepatocellular carcinoma. J Clin Translatl Hepatol. 2023;11(5):1184–91.
12.
Zurück zum Zitat Handelman GS, Kok HK, Chandra RV, et al. eDoctor: machine learning and the future of medicine. J Intern Med. 2018;284(6):603–19.CrossRefPubMed Handelman GS, Kok HK, Chandra RV, et al. eDoctor: machine learning and the future of medicine. J Intern Med. 2018;284(6):603–19.CrossRefPubMed
13.
Zurück zum Zitat Maglogiannis I, Zafiropoulos E, Anagnostopoulos I. An intelligent system for automated breast cancer diagnosis and prognosis using SVM based classifiers. Appl Intell. 2009;30(1):24–36.CrossRef Maglogiannis I, Zafiropoulos E, Anagnostopoulos I. An intelligent system for automated breast cancer diagnosis and prognosis using SVM based classifiers. Appl Intell. 2009;30(1):24–36.CrossRef
14.
Zurück zum Zitat Kuo RJ, Huang MH, Cheng WC, et al. Application of a two-stage fuzzy neural network to a prostate cancer prognosis system. Artif Intell Med. 2015;63(2):119–33.CrossRefPubMed Kuo RJ, Huang MH, Cheng WC, et al. Application of a two-stage fuzzy neural network to a prostate cancer prognosis system. Artif Intell Med. 2015;63(2):119–33.CrossRefPubMed
15.
Zurück zum Zitat Peng JF, Chen C, Zhou M, et al. A Machine-learning approach to forecast aggravation risk in patients with acute exacerbation of chronic obstructive pulmonary disease with clinical indicators. Sci Rep. 2020;10(1):3118.CrossRefPubMedPubMedCentral Peng JF, Chen C, Zhou M, et al. A Machine-learning approach to forecast aggravation risk in patients with acute exacerbation of chronic obstructive pulmonary disease with clinical indicators. Sci Rep. 2020;10(1):3118.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Dore MP, Vidili G, Marras G, et al. Inverse association between glucose-6-phosphate dehydrogenase deficiency and hepatocellular carcinoma. Asian Pacific J Cancer Prevent. 2018;19(4):1069–73. Dore MP, Vidili G, Marras G, et al. Inverse association between glucose-6-phosphate dehydrogenase deficiency and hepatocellular carcinoma. Asian Pacific J Cancer Prevent. 2018;19(4):1069–73.
17.
18.
Zurück zum Zitat Cao F, Luo AG, Yang CW. G6PD inhibits ferroptosis in hepatocellular carcinoma by targeting cytochrome P450 oxidoreductase. Cellu Signal. 2021;87:110098.CrossRef Cao F, Luo AG, Yang CW. G6PD inhibits ferroptosis in hepatocellular carcinoma by targeting cytochrome P450 oxidoreductase. Cellu Signal. 2021;87:110098.CrossRef
19.
Zurück zum Zitat Lu M, Lu L, Dong QZ, et al. Elevated G6PD expression contributes to migration and invasion of hepatocellular carcinoma cells by inducing epithelial-mesenchymal transition. Acta Biochim Biophys Sin. 2018;50(4):370–80.CrossRefPubMed Lu M, Lu L, Dong QZ, et al. Elevated G6PD expression contributes to migration and invasion of hepatocellular carcinoma cells by inducing epithelial-mesenchymal transition. Acta Biochim Biophys Sin. 2018;50(4):370–80.CrossRefPubMed
20.
Zurück zum Zitat Li M, He XX, Guo WX, et al. Aldolase B suppresses hepatocellular carcinogenesis by inhibiting G6PD and pentose phosphate pathways. Nat Cancer. 2020;1(7):735-+.CrossRefPubMed Li M, He XX, Guo WX, et al. Aldolase B suppresses hepatocellular carcinogenesis by inhibiting G6PD and pentose phosphate pathways. Nat Cancer. 2020;1(7):735-+.CrossRefPubMed
21.
Zurück zum Zitat Yang LP, He Y, Zhang ZF, et al. Upregulation of CEP55 predicts dismal prognosis in patients with liver cancer. Biomed Res Int. 2020;2020:4139320.PubMedPubMedCentral Yang LP, He Y, Zhang ZF, et al. Upregulation of CEP55 predicts dismal prognosis in patients with liver cancer. Biomed Res Int. 2020;2020:4139320.PubMedPubMedCentral
22.
Zurück zum Zitat Ju LL, Li XF, Shao JG, et al. Upregulation of thyroid hormone receptor interactor 13 is associated with human hepatocellular carcinoma. Oncol Rep. 2018;40(6):3794–802.PubMed Ju LL, Li XF, Shao JG, et al. Upregulation of thyroid hormone receptor interactor 13 is associated with human hepatocellular carcinoma. Oncol Rep. 2018;40(6):3794–802.PubMed
23.
Zurück zum Zitat Li J, Gao JZ, Du JL, et al. Increased CDC20 expression is associated with development and progression of hepatocellular carcinoma. Int J Oncol. 2014;45(4):1547–55.CrossRefPubMed Li J, Gao JZ, Du JL, et al. Increased CDC20 expression is associated with development and progression of hepatocellular carcinoma. Int J Oncol. 2014;45(4):1547–55.CrossRefPubMed
24.
Zurück zum Zitat Guan Z, Cheng W, Huang D, et al. High MYBL2 expression and transcription regulatory activity is associated with poor overall survival in patients with hepatocellular carcinoma. Curr Res Translatl Med. 2018;66(1):27–32.CrossRef Guan Z, Cheng W, Huang D, et al. High MYBL2 expression and transcription regulatory activity is associated with poor overall survival in patients with hepatocellular carcinoma. Curr Res Translatl Med. 2018;66(1):27–32.CrossRef
25.
Zurück zum Zitat Cheng J, Huang Y, Zhang XH, et al. TRIM21 and PHLDA3 negatively regulate the crosstalk between the PI3K/AKT pathway and PPP metabolism. Nat Commun. 2020;11(1):1880.CrossRefPubMedPubMedCentral Cheng J, Huang Y, Zhang XH, et al. TRIM21 and PHLDA3 negatively regulate the crosstalk between the PI3K/AKT pathway and PPP metabolism. Nat Commun. 2020;11(1):1880.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Tekin C, Aberson HL, Bijlsma MF, et al. Early macrophage infiltrates impair pancreatic cancer cell growth by TNF-alpha secretion. Bmc Cancer. 2020;20(1):1–9.CrossRef Tekin C, Aberson HL, Bijlsma MF, et al. Early macrophage infiltrates impair pancreatic cancer cell growth by TNF-alpha secretion. Bmc Cancer. 2020;20(1):1–9.CrossRef
27.
Zurück zum Zitat Lu HC, Parikh PP, Lorber DL. Phenformin-associated lactic acidosis due to imported phenformin. Diabetes Care. 1996;19(12):1449–50.CrossRefPubMed Lu HC, Parikh PP, Lorber DL. Phenformin-associated lactic acidosis due to imported phenformin. Diabetes Care. 1996;19(12):1449–50.CrossRefPubMed
28.
Zurück zum Zitat Rubino MEG, Carrillo E, Alcala GR, et al. Phenformin as an anticancer agent: challenges and prospects. Int J Mol Sci. 2019;20(13):3316.CrossRef Rubino MEG, Carrillo E, Alcala GR, et al. Phenformin as an anticancer agent: challenges and prospects. Int J Mol Sci. 2019;20(13):3316.CrossRef
29.
Zurück zum Zitat Huang LL, Xiao D, Wu TY, et al. Phenformin synergistically sensitizes liver cancer cells to sorafenib by downregulating CRAF/ERK and PI3K/AKT/mTOR pathways. American J Transll Res. 2021;13(7):7508–23. Huang LL, Xiao D, Wu TY, et al. Phenformin synergistically sensitizes liver cancer cells to sorafenib by downregulating CRAF/ERK and PI3K/AKT/mTOR pathways. American J Transll Res. 2021;13(7):7508–23.
30.
Zurück zum Zitat Li F, Zhang SH, Pang LM. Meta-analysis of efficacy and adverse events of erlotinib-based targeted therapies for advanced/metastatic non-small cell lung cancer. Oncotarget. 2017;8(49):86816–27.CrossRefPubMedPubMedCentral Li F, Zhang SH, Pang LM. Meta-analysis of efficacy and adverse events of erlotinib-based targeted therapies for advanced/metastatic non-small cell lung cancer. Oncotarget. 2017;8(49):86816–27.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Zheng SF, Ni JP, Li Y, et al. 2-Methoxyestradiol synergizes with Erlotinib to suppress hepatocellular carcinoma by disrupting the PLAGL2-EGFR-HIF-1/2 alpha signaling loop. Pharmacol Res. 2021;169:105685. Zheng SF, Ni JP, Li Y, et al. 2-Methoxyestradiol synergizes with Erlotinib to suppress hepatocellular carcinoma by disrupting the PLAGL2-EGFR-HIF-1/2 alpha signaling loop. Pharmacol Res. 2021;169:105685.
32.
Zurück zum Zitat Zhu AX, Rosmorduc O, Evans TRJ, et al. SEARCH: a phase III, randomized, double-blind, placebo-controlled trial of sorafenib plus erlotinib in patients with advanced hepatocellular carcinoma. J Clin Oncol. 2015;33(6):559–66.CrossRefPubMed Zhu AX, Rosmorduc O, Evans TRJ, et al. SEARCH: a phase III, randomized, double-blind, placebo-controlled trial of sorafenib plus erlotinib in patients with advanced hepatocellular carcinoma. J Clin Oncol. 2015;33(6):559–66.CrossRefPubMed
33.
Zurück zum Zitat Chen J, Jin RN, Zhao J, et al. Potential molecular, cellular and microenvironmental mechanism of sorafenib resistance in hepatocellular carcinoma. Cancer Lett. 2015;367(1):1–11.CrossRefPubMed Chen J, Jin RN, Zhao J, et al. Potential molecular, cellular and microenvironmental mechanism of sorafenib resistance in hepatocellular carcinoma. Cancer Lett. 2015;367(1):1–11.CrossRefPubMed
34.
Zurück zum Zitat Li YG, Tang SJ, Shi XH, et al. Metabolic classification suggests the GLUT1/ALDOB/G6PD axis as a therapeutic target in chemotherapy-resistant pancreatic cancer. Cell Rep Med. 2023;4(9):101162.CrossRefPubMedPubMedCentral Li YG, Tang SJ, Shi XH, et al. Metabolic classification suggests the GLUT1/ALDOB/G6PD axis as a therapeutic target in chemotherapy-resistant pancreatic cancer. Cell Rep Med. 2023;4(9):101162.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Junath N, Bharadwaj A, Tyagi S, et al. Prognostic diagnosis for breast cancer patients using probabilistic bayesian classification. Biomed Res Int. 2022;2022:1859222.CrossRefPubMedPubMedCentral Junath N, Bharadwaj A, Tyagi S, et al. Prognostic diagnosis for breast cancer patients using probabilistic bayesian classification. Biomed Res Int. 2022;2022:1859222.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Bo JZ, Li S, Ma PF, et al. Research on Early Warning mechanism and model of liver cancer rehabilitation based on CS-SVM. J Healthcare Eng. 2021;2021:6658776. Bo JZ, Li S, Ma PF, et al. Research on Early Warning mechanism and model of liver cancer rehabilitation based on CS-SVM. J Healthcare Eng. 2021;2021:6658776.
37.
Zurück zum Zitat Noh MG, Yoon Y, Kim G, et al. Practical prediction model of the clinical response to programmed death-ligand 1 inhibitors in advanced gastric cancer. Exp Mol Med. 2021;53(2):223–34.CrossRefPubMedPubMedCentral Noh MG, Yoon Y, Kim G, et al. Practical prediction model of the clinical response to programmed death-ligand 1 inhibitors in advanced gastric cancer. Exp Mol Med. 2021;53(2):223–34.CrossRefPubMedPubMedCentral
Metadaten
Titel
G6PD and machine learning algorithms as prognostic and diagnostic indicators of liver hepatocellular carcinoma
verfasst von
Fei Li
Boshen Wang
Hao Li
Lu Kong
Baoli Zhu
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2024
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-024-11887-6

Weitere Artikel der Ausgabe 1/2024

BMC Cancer 1/2024 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Alter verschlechtert Prognose bei Endometriumkarzinom

11.05.2024 Endometriumkarzinom Nachrichten

Ein höheres Alter bei der Diagnose eines Endometriumkarzinoms ist mit aggressiveren Tumorcharakteristika assoziiert, scheint aber auch unabhängig von bekannten Risikofaktoren die Prognose der Erkrankung zu verschlimmern.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.