Abstract
Objectives
Osteosarcoma stands as a highly aggressive primary bone malignancy with a notable penchant for metastasis and a grim prognosis. The exploration of metabolic gene signatures, particularly those involved in glycolysis and cholesterol synthesis, has recently garnered attention for their potential to predict cancer progression and therapeutic outcomes. This study probes the prognostic value of a glycolysis and cholesterol synthesis-related gene signature (GCSRG) in osteosarcoma, along with its influence on the tumor immune microenvironment.
Methods
A comprehensive bioinformatics approach was applied to osteosarcoma samples from the TCGA database, incorporating unsupervised clustering to delineate patient subsets, differential gene expression analysis to identify key metabolic pathways, and survival analysis to ascertain prognostic validity.
Results
The investigation yielded a distinct GCSRG with significant prognostic capabilities. Notably, a high GCSRG score correlated with worse patient outcomes but revealed a marked enrichment in immune cell infiltration within the tumor milieu, suggesting a complex relationship between metabolism and immune surveillance in osteosarcoma.
Conclusion
The GCSRG emerges as a promising biomarker for osteosarcoma prognosis, offering new vistas for assessing patient suitability for immunotherapeutic interventions. The potential of the GCSRG to act as a guide for personalized treatment strategies is highlighted, underscoring the need for strategic therapeutic modulation based on metabolic and immune interactions to improve patient prognosis in osteosarcoma.
Introduction
Osteosarcoma represents the predominant form of primary malignant bone tumors in pediatric and adolescent populations, accounting for approximately 20–35 % of all malignant bone tumors [1, 2]. While localized osteosarcoma exhibits a 5-year survival rate ranging from 60–70 %, the prognosis becomes considerably poorer for patients with metastatic or relapsed disease, with survival rates falling between 15 and 30 % [3, 4]. This malignancy is characterized by its remarkable tendency for metastasis and an aggressive clinical trajectory. Despite advancements in surgical techniques, chemotherapy, and radiation, the overall survival rate of osteosarcoma patients has not significantly improved in recent years. To improve clinical outcomes for individuals with osteosarcoma, it is imperative to identify novel therapeutic targets and prognostic indicators that can guide treatment decisions and enhance patient care.
Emerging evidence highlights the critical role of cancer metabolism in the initiation and dissemination of tumors [5], [6], [7]. Dysregulated metabolic pathways, including enhanced glycolysis and cholesterol synthesis, have been observed to facilitate rapid proliferation, survival, and metastasis of tumor cells [8], [9], [10]. The glycolysis and cholesterol synthesis-related gene (GCSRG) signature, composed of genes associated with both cholesterol synthesis and glycolysis, has been implicated in various malignancies [11, 12]. However, the potential significance of the GCSRG signature in osteosarcoma remains largely unexplored.
The tumor microenvironment (TME) plays a crucial role in guiding the onset, progression, and therapeutic responses of tumors [13], [14], [15]. Within this milieu, immune cells are instrumental in determining cancer growth, metastasis, and the ability of cancer cells to dodge immune detection [16], [17], [18]. Impressively, immune checkpoint inhibitors have surfaced as revolutionary treatments, showing extraordinary effectiveness against various cancers, including melanoma and non-small cell lung cancer [19], [20], [21], [22]. Yet, even with these strides in immunotherapies, their benefits in osteosarcoma remain inconsistent, with the underlying determinants of their effectiveness still elusive. Delving into the relationship between the GCSRG signature and osteosarcoma’s immune landscape may shed light on ways tumors sidestep immune detection, opening doors to potential therapeutic innovations. A deeper understanding of these dynamics can pave the way for improved osteosarcoma treatment outcomes.
This study’s core aim was to discern the influence of the GCSRG signature on osteosarcoma’s prognosis and its immune microenvironment. Through our meticulous analysis, we aspire to unravel the multifaceted role of the GCSRG signature, both in its prognostic capacity and its impact on the immune backdrop of osteosarcoma. These revelations could spotlight groundbreaking therapeutic and prognostic markers, substantially elevating the care standards for osteosarcoma patients.
Methods
Data collection and processing
The osteosarcoma cohort (TARGET-OS) was acquired from The Cancer Genome Atlas (TCGA) database (https://portal.gdc.cancer.gov/), encompassing 88 tissue samples. The obtained data were in the form of Fragments Per Kilobase of exon model per Million mapped fragments (FPKM) values, which underwent logarithmic transformation for subsequent analysis.
To further enrich our investigation, we gathered a collection of 72 genes associated with “REACTOME_GLYCOLYSIS” and 27 genes associated with “REACTOME_ CHOLESTEROL _BIOSYNTHESIS” from the Gene Set Enrichment Analysis (GSEA) website available at http://www.gsea-msigdb.org. Upon identification of one undetected gene within the TCGA cohort, our final gene set consisted of 98 genes, forming the basis of the glycolysis and cholesterol biosynthesis-related gene set for subsequent analysis.
Unsupervised clustering analysis of TARGET-OS samples
To explore the heterogeneity within the 88 TARGET-OS samples, we performed an extensive unsupervised clustering analysis using the Consensus Clustering method. This approach significantly enhances the precision and reliability of deciphering complex patterns within the dataset [23, 24]. The assessment of the optimal number of subgroups was based on evaluating the Cumulative Distribution Function (CDF) values and carefully inspecting the clustering heatmap.
Following the identification of distinct subgroups, we conducted a comprehensive differential gene expression analysis by considering all genes associated with osteosarcoma. Additionally, we compared the overall survival rates between these subgroups. To specifically investigate the differential expression patterns of the 98 GCSRG genes within the identified subgroups, we employed the “limma” R package to perform a differential analysis.
Analysis of mutation frequency and tumor mutation burden in different subgroups
To gain a comprehensive understanding of the genetic characteristics underlying osteosarcoma and identify potential therapeutic targets, we conducted an in-depth analysis of mutation frequencies within the distinct GCSRG subgroups. By leveraging the rich genomic data available from the TCGA database, our objective was to uncover the divergent mutational profiles between these subgroups and their potential implications for clinical outcomes.
Furthermore, we conducted an extensive examination of potential variations in tumor mutation burden (TMB) between the two subgroups using the “maftools” R package. This comprehensive analysis allowed us to evaluate the significance of TMB in relation to osteosarcoma, shedding light on its potential as a prognostic biomarker and its influence on the immune microenvironment and response to treatment.
Tumor Immune Dysfunction and Exclusion (TIDE) score analysis
We scrutinized the immune therapy response between the high- and low-risk groups by analyzing the Tumor Immune Dysfunction and Exclusion (TIDE) (http://tide.dfci.harvard.edu/) scores, which could provide indications of immunotherapy responsiveness in osteosarcoma patients.
Assessment of immune microenvironment in different subtypes
In order to investigate the association between the two subtypes of TARGET-OS samples and immune cell subpopulations, we utilized the CIBERSORT algorithm. This approach enabled us to comprehensively evaluate the infiltration of 22 distinct immune cell types within the TARGET-OS samples. Furthermore, we conducted an in-depth analysis of the expression patterns of human leukocyte antigen (HLA)-related genes and immune checkpoint-related genes across the different subtypes, providing valuable insights into their immunological characteristics.
Regression model construction for GCSRG-related genes
Our regression model for the GCSRG-related genes was meticulously developed through a series of comprehensive steps. Initially, a univariate Cox regression analysis was performed to identify genes significantly associated with overall survival, utilizing a significance threshold of p-value <0.05. Subsequently, we refined the selection of survival-related genes by employing LASSO regression and multivariate Cox regression analyses. These rigorous techniques enabled us to construct a robust prognostic model that accurately estimates the prognosis of osteosarcoma patients based on the expression levels of genes [25]. This meticulous approach ensured the inclusion of only the most crucial genes, enhancing the reliability and clinical utility of our prognostic model.
GSEA enrichment analysis
To elucidate the distinct molecular processes that differentiate the high- and low-risk groups, we conducted Gene Set Enrichment Analysis (GSEA). This approach enabled us to identify enriched signaling pathways within each group, providing valuable insights into the underlying biological mechanisms. By employing GSEA, we sought to unravel the intricate molecular landscape and shed light on the key pathways driving the divergent risk profiles observed in our study.
Statistical processing
All statistical analyses were conducted using R software, version 4.1.0, developed by the R Foundation for Statistical Computing, Vienna, Austria. The Student’s t-test or Wilcoxon test was employed to compare the two groups. The Kruskal–Wallis test was used to analyze comparisons between several groups. The log-rank test and Kaplan–Meier plots, which display survival curves, were compared. If the p-value was less than 0.05, statistical findings were declared significant.
Results
Optimal subgroup identification
Based on the CDF values and clustering heatmap, it was determined that the optimal number of subgroups for our analysis was two. This partitioning maximized the dissimilarities between the groups while minimizing variations within each group (Figure 1A and B).

Identification of optimal subgroups based on gene expression profiles. (A) Clustering heatmap showing the gene expression pattern of the identified subgroups. (B) Cumulative distribution function (CDF) plot for determining the optimal number of subgroups. (C) Heatmap illustrating the differential gene expression between the two subgroups. (D) Kaplan–Meier survival curve comparing overall survival rates of the GCSRG low and high subgroups.
Through differential gene expression analysis, we identified substantial alterations in gene expression patterns between the two groups (Figure 1C). Additionally, when comparing the overall survival rates, we observed a significantly higher overall survival in the GCSRG low group (Figure 1D).
Mutation frequency analysis in different subgroups and TIDE score analysis
The analysis unveiled a notable disparity in mutation frequency between the GCSRG low subgroup (56 %) and the GCSRG high subgroup (80 %). TP53 and MUC16 emerged as the two most frequently altered genes across both subgroups, underscoring their potential significance (Figure 2A and B).

Mutation frequency analysis in the GCSRG low and high subgroups. (A) Mutation frequency plot for the GCSRG low subgroup. (B) Mutation frequency plot for the GCSRG high subgroup. (C) Box plot comparing the tumor mutation burden between the two subgroups. (D) Box plot comparing the TIDE scores between the two subgroups.
Contrary to our expectations, no statistically significant distinction in tumor mutation burden was observed when comparing the two groups (Figure 2C). However, intriguingly, the high-risk group exhibited lower TIDE scores, indicative of a higher likelihood of favorable response to immunotherapy (Figure 2D).
Immune microenvironment in different subtypes
The bar plot in Figure 3A effectively visualized the abundance and distribution of the 22 distinct immune cell types within the TARGET-OS samples. Subsequent correlation heatmap analysis unveiled predominantly negative correlations among the various immune cell populations (Figure 3B). Notably, employing the ESTIMATE algorithm, we observed elevated stromal scores, immune scores, and microenvironment scores in the GCSRG low subtype, indicating a more dynamic and active microenvironment in this subgroup (Figure 3C–F). Furthermore, our assessment of HLA-related genes revealed higher expression levels in the GCSRG low subtype compared to the GCSRG high subtype (Figure 3G).

Immune microenvironment analysis of the different subtypes. (A) Bar plot illustrating the infiltration of 22 immune cell types in the TARGET-OS samples. (B) Correlation heatmap of immune cell infiltration. (C–F) Box plots showing stromal scores, immune scores, and microenvironment scores determined using the ESTIMATE algorithm. (G) Box plot displaying the expression levels of HLA-related genes in the GCSRG low and high subtypes. (H) Box plot showing the expression levels of immune checkpoint-related genes in the two subtypes. *Indicates a p-value less than 0.05, **denotes a p-value less than 0.01, and ***signifies a p-value less than 0.001.
Consistent with our previous findings, the differential analysis of immune checkpoint-related genes demonstrated heightened expression levels in the GCSRG low subtype (Figure 3H). This observation further supports the notion that the GCSRG low subtype exhibits a heightened susceptibility to immune cell infiltration. Consequently, patients harboring this particular subtype may potentially benefit from enhanced effectiveness of immunotherapy interventions.
Regression model construction for GCSRG-related genes
Through the utilization of univariate Cox regression analysis, we successfully identified eight genes that displayed a significant association with overall survival (Figure 4A). To refine our selection of survival-related genes, we employed two filtering approaches. The first involved LASSO regression, where eight genes were retained based on the optimal lambda value, thus establishing a more precise gene subset (Figure 4B and C). Subsequently, employing multivariate Cox regression analysis, we established a prognostic model comprising eight genes: GNPDA2, NUP98, PFKFB2, PFKFB3, PKLR, PRKACB, SQLE, and TM7SF2.

Regression model construction for GCSRG-related genes. (A) Forest plot of univariate Cox regression analysis identifying overall survival-related genes. (B–C) LASSO regression plot for selecting survival-related genes. (D) Kaplan–Meier survival curve comparing overall survival rates between high- and low-risk groups based on the risk scores. (E) ROC curve for evaluating the sensitivity and accuracy of the risk scores. (F–G) Plots illustrating the distribution of risk scores and survival status. (H) Heatmap of the expression levels of the eight genes in the high- and low-risk groups.
Risk scores, derived from the expression levels of the aforementioned eight genes and their respective regression coefficients, were calculated to stratify patients into high- and low-risk groups. Notably, these risk groups exhibited substantial disparities in overall survival outcomes (Figure 4D). Furthermore, the ROC curve analysis demonstrated that the risk score displayed remarkable sensitivity and accuracy in distinguishing between high- and low-risk patients (Figure 4E).
By utilizing the risk scores, we implemented a ranking system for patients and observed a consistent pattern where higher risk scores were associated with decreased survival rates (Figure 4F and G).
This aligns with the expected characteristics of a risk model. Among the eight genes included in the model, TM7SF2, SQLE, PKLR, and PFKFB3 exhibited high expression levels in the high-risk group, while the remaining four genes showed lower expression levels in the high-risk group (Figure 4H).
Prognostic model construction
In our study, univariate Cox regression analysis revealed that metastasis and risk score achieved statistical significance with a p-value below 0.05 (Figure 5A). Subsequently, in the multivariate Cox regression analysis, both metastasis and risk score emerged as independent prognostic indicators, underscoring their importance in predicting patient outcomes (Figure 5B). Notably, when assessing the performance of the four components through ROC curve analysis, the risk score exhibited the highest AUC value of 0.85 (Figure 5C).

Prognostic model construction and TIDE score analysis. (A) Forest plot of univariate Cox regression analysis identifying factors with p-value <0.05. (B) Forest plot of multivariate Cox regression analysis determining independent prognostic factors. (C) ROC curve analysis of the four factors. (D) Construction of the prognostic prediction model based on the four factors. (E) Calibration curve of the prognostic prediction model. (F) Harrell’s C-index value calculation. (G) Box plot showing TIDE scores for the high- and low-risk groups, *** denotes a p-value less than 0.001.
Using the aforementioned four parameters, we constructed a comprehensive prognostic prediction model (Figure 5D). The calibration curve analysis further demonstrated excellent discrimination and calibration of the model (Figure 5E). Remarkably, when computing Harrell’s C-index, the risk score emerged as the parameter with the highest value, underscoring its superior predictive capability (Figure 5F).
GSEA enrichment analysis
The GSEA analysis revealed distinct enrichment patterns in signaling pathways between the low- and high-risk groups. Specifically, the low-risk group exhibited pronounced enrichment in signaling pathways such as the Jak–STAT signaling pathway, cytokine-cytokine receptor interaction, and neuroactive ligand–receptor interaction (Figure 6A). In contrast, the high-risk group demonstrated a prominent enrichment of pathways associated with Huntington’s disease, olfactory transduction, and other relevant pathways (Figure 6B).

GSEA enrichment analysis results. (A) Enrichment plot for signaling pathways enriched in the low-risk group. (B) Enrichment plot for signaling pathways enriched in the high-risk group.
Discussion
This study endeavored to illuminate the influence of the GCSRG signature on the immunological landscape and the prognosis of osteosarcoma. Through an exhaustive analysis, we revealed a pronounced link between the GCSRG signature and diverse clinical traits, patterns of immune cell infiltration, and the efficacy of immunotherapy in osteosarcoma patients. Significantly, within the GCSRG signature, we pinpointed certain genes that stand out as promising therapeutic and prognostic candidates for osteosarcoma management.
Our study revealed a significant association between the GCSRG signature and overall survival in patients with osteosarcoma. By evaluating the expression levels of GCSRG genes, we identified two distinct subtypes of osteosarcoma characterized by markedly different clinical outcomes. These findings align with previous research conducted in various cancer types, highlighting the predictive value of gene signatures related to glycolysis and cholesterol synthesis in estimating patient survival. For instance, Wang et al. demonstrated a significant association between a gene signature related to cholesterol synthesis and the prognosis of patients with hepatocellular carcinoma [26]. Similarly, Zhang et al. uncovered a glycolysis-related gene signature that exhibited predictive value for overall survival in individuals with lung adenocarcinoma [27]. Building upon these previous findings, our research highlights the potential of metabolic gene profiles as prognostic biomarkers not only in bone cancers but also specifically in the context of osteosarcoma.
In addition, we examined the correlation between the GCSRG signature and immune cell infiltration within the tumor microenvironment (TME) of osteosarcoma patients. Notably, distinct patterns of immune cell infiltration were observed between the high- and low-risk GCSRG groups, with the low-risk group exhibiting a more dynamic and active immunological microenvironment. These findings suggest that the GCSRG signature exerts a substantial influence on the immunological response to osteosarcoma, potentially influencing disease progression and patient prognosis. Our findings align with prior research that has elucidated the influence of metabolic pathways, namely glycolysis and cholesterol synthesis, on immune cell function and anti-tumor immune responses [28, 29]. For instance, Westerterp et al. demonstrated the role of cholesterol biosynthesis in tumor-associated macrophages in promoting tumor growth and metastasis by exerting inhibitory effects on anti-tumor immunity [30]. Similarly, Qin et al. revealed that tumor-derived lactate, a metabolic byproduct of glycolysis, can impede the activity of cytotoxic T cells and natural killer (NK) cells [31].
In our study, we also explored the therapeutic potential of GCSRG-related genes in the context of osteosarcoma. We identified eight genes that exhibited significant predictive value and have the potential to serve as promising targets for future therapeutic interventions. Notably, one of the genes we discovered, PFKFB3, encodes a crucial enzyme involved in regulating the rate of glycolysis. This gene has been implicated in the migration and proliferation of various cancer cells, including osteosarcoma. In preclinical models of osteosarcoma, targeting and inhibiting PFKFB3 has demonstrated the ability to impede tumor growth and metastasis, underscoring its potential as a promising therapeutic target. Similarly, SQLE, another gene within our prognostic signature, plays a pivotal role in cholesterol production and has been implicated in poor prognosis across various cancer types [32], [33], [34]. Encouragingly, reports have indicated that inhibiting SQLE activity could potentially slow tumor progression and sensitize cancer cells to chemotherapy and immunotherapy [35], [36], [37]. These compelling findings emphasize the therapeutic prospects associated with GCSRG-related genes in the context of osteosarcoma and warrant further exploration through both preclinical and clinical investigations.
Furthermore, we evaluated the response to immunotherapy in the high- and low-risk GCSRG groups through the analysis of TIDE scores. Interestingly, our findings revealed that the high-risk group displayed lower TIDE scores, implying a potentially enhanced responsiveness to immunotherapy interventions. This observation highlights the potential of the GCSRG signature as a predictive biomarker for immunotherapy response in osteosarcoma patients, offering valuable insights that can inform personalized treatment approaches. Immunotherapy has emerged as a promising therapeutic modality across various cancer types, including osteosarcoma. However, the efficacy of immunotherapy varies among patients, necessitating the identification of reliable biomarkers to select individuals likely to respond favorably to such treatments. Our findings propose that the GCSRG signature may serve as a valuable tool in this regard, facilitating the optimization of immunotherapeutic strategies for patients with osteosarcoma. Importantly, previous studies have also highlighted the predictive value of metabolic gene signatures in determining immunotherapy response in other malignancies. For instance, Choi et al. demonstrated the prognostic significance of a glycolysis-related gene signature in predicting the response to immune checkpoint inhibitors in patients with non-small cell lung cancer [38, 39].
Despite the promising findings of this research, there are several limitations to consider. First, our study primarily relied on data from the TCGA database, which might not encompass the vast heterogeneity of osteosarcoma cases globally. Relying on a single database could introduce selection bias, potentially affecting the generalizability of our results. Second, while we identified associations between the GCSRG signature and clinical characteristics, a causal relationship cannot be firmly established due to the retrospective nature of our analysis. Experimental studies are essential to validate these associations. Additionally, the functional roles of the identified genes within the GCSRG signature need further in-depth experimental validation in both in vitro and in vivo settings. Lastly, the study did not account for potential confounders such as patients’ overall health, previous treatments, or other environmental factors, which could influence immune cell infiltration and response to immunotherapy. Future studies incorporating a more diverse dataset and addressing these limitations are needed to reinforce and expand upon our findings.
Conclusions
In essence, our research highlights the GCSRG signature’s critical prognostic role in osteosarcoma, emphasizing its influence on the immune microenvironment and immunotherapy responsiveness. By pinpointing key GCSRG-related genes, we’ve spotlighted potential therapeutic avenues. Our results also underscore the GCSRG signature’s promise as a predictive biomarker for immunotherapy efficacy. By employing GCSRG-based stratification, we advocate for a tailored approach in osteosarcoma care, which can inform treatment choices and improve patient outcomes. Future studies should validate our findings and delve deeper into the intricate relationships between the GCSRG signature, immune dynamics, and therapeutic responses.
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: YC designed the study. JF and JZ performed data analysis. JF drafted the manuscript. YC and JZ revised the manuscript. All authors read and approved the final manuscript.
-
Competing interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
-
Research funding: None.
-
Data availability: The original contributions presented in the study are included in the article/supplementary material, further inquiries can be directed to the corresponding author.
References
1. Puri, A. Chondrosarcomas in children and adolescents. EFORT Open Rev 2020;5:90–5, https://doi.org/10.1302/2058-5241.5.190052.Search in Google Scholar PubMed PubMed Central
2. Lee, JA, Lim, J, Jin, HY, Park, M, Park, HJ, Park, JW, et al.. Osteosarcoma in adolescents and young adults. Cells 2021;10.2684, https://doi.org/10.3390/cells10102684,Search in Google Scholar PubMed PubMed Central
3. Sheng, G, Gao, Y, Yang, Y, Wu, H. Osteosarcoma and metastasis. Front Oncol 2021;11:780264, https://doi.org/10.3389/fonc.2021.780264.Search in Google Scholar PubMed PubMed Central
4. Yang, C, Tian, Y, Zhao, F, Chen, Z, Su, P, Li, Y, et al.. Bone microenvironment and osteosarcoma metastasis. Int J Mol Sci 2020;21:6985, https://doi.org/10.3390/ijms21196985,Search in Google Scholar PubMed PubMed Central
5. Avnet, S, Baldini, N, Brisson, L, De Milito, A, Otto, AM, Pastoreková, S, et al.. Annual Meeting of the International Society of cancer metabolism (ISCaM): cancer metabolism. Front Oncol 2018;8:329, https://doi.org/10.3389/fonc.2018.00329.Search in Google Scholar PubMed PubMed Central
6. Kim, SY. Cancer metabolism: a hope for curing cancer. Biomol Ther 2018;26:1–3, https://doi.org/10.4062/biomolther.2017.300.Search in Google Scholar PubMed PubMed Central
7. Basetti, M. Special issue: cancer metabolism. Metabolites 2017;7:41, https://doi.org/10.3390/metabo7030041,Search in Google Scholar PubMed PubMed Central
8. Chen, YJ, Guo, X, Liu, ML, Yu, YY, Cui, YH, Shen, XZ, et al.. Interaction between glycolysis‒cholesterol synthesis axis and tumor microenvironment reveal that gamma-glutamyl hydrolase suppresses glycolysis in colon cancer. Front Immunol 2022;13:979521, https://doi.org/10.3389/fimmu.2022.979521.Search in Google Scholar PubMed PubMed Central
9. Jiang, J, Zheng, Q, Zhu, W, Chen, X, Lu, H, Chen, D, et al.. Alterations in glycolytic/cholesterogenic gene expression in hepatocellular carcinoma. Aging 2020;12:10300–16, https://doi.org/10.18632/aging.103254.Search in Google Scholar PubMed PubMed Central
10. Zhang, E, Chen, Y, Bao, S, Hou, X, Hu, J, Mu, OYN, et al.. Identification of subgroups along the glycolysis-cholesterol synthesis axis and the development of an associated prognostic risk model. Hum Genomics 2021;15:53, https://doi.org/10.1186/s40246-021-00350-3.Search in Google Scholar PubMed PubMed Central
11. Pedersen, AF, Vedsted, P. Cancer beliefs in cancer survivors, cancer relatives and persons with no cancer experience. Scand J Public Health 2019;47:497–503, https://doi.org/10.1177/1403494817715380.Search in Google Scholar PubMed
12. Xu, F, Yan, J, Peng, Z, Liu, J, Li, Z. Comprehensive analysis of a glycolysis and cholesterol synthesis-related genes signature for predicting prognosis and immune landscape in osteosarcoma. Front Immunol 2022;13:1096009, https://doi.org/10.3389/fimmu.2022.1096009.Search in Google Scholar PubMed PubMed Central
13. Anderson, NM, Simon, MC. The tumor microenvironment. Curr Biol 2020;30:R921–r5, https://doi.org/10.1016/j.cub.2020.06.081.Search in Google Scholar PubMed PubMed Central
14. Butturini, E, Carcereri de Prati, A, Boriero, D, Mariotto, S. Tumor dormancy and interplay with hypoxic tumor microenvironment. Int J Mol Sci 2019;20:4305, https://doi.org/10.3390/ijms20174305,Search in Google Scholar PubMed PubMed Central
15. Laplane, L, Duluc, D, Bikfalvi, A, Larmonier, N, Pradeu, T. Beyond the tumour microenvironment. Int J Cancer 2019;145:2611–8, https://doi.org/10.1002/ijc.32343.Search in Google Scholar PubMed PubMed Central
16. Gao, Y, Pan, Z, Li, H, Wang, F. Antitumor therapy targeting the tumor microenvironment. J Oncol 2023;2023:6886135–16, https://doi.org/10.1155/2023/6886135.Search in Google Scholar PubMed PubMed Central
17. Dart, A. Tumour microenvironment: radical changes. Nat Rev Cancer 2018;18:65, https://doi.org/10.1038/nrc.2018.4.Search in Google Scholar PubMed
18. Ravi Kiran, A, Kusuma Kumari, G, Krishnamurthy, PT, Khaydarov, RR. Tumor microenvironment and nanotherapeutics: intruding the tumor fort. Biomater Sci 2021;9:7667–704, https://doi.org/10.1039/d1bm01127h.Search in Google Scholar PubMed
19. Carlino, MS, Larkin, J, Long, GV. Immune checkpoint inhibitors in melanoma. Lancet 2021;398:1002–14, https://doi.org/10.1016/s0140-6736(21)01206-x.Search in Google Scholar PubMed
20. Furue, M, Ito, T, Wada, N, Wada, M, Kadono, T, Uchi, H. Melanoma and immune checkpoint inhibitors. Curr Oncol Rep 2018;20:29, https://doi.org/10.1007/s11912-018-0676-z.Search in Google Scholar PubMed
21. Zhou, F, Qiao, M, Zhou, C. The cutting-edge progress of immune-checkpoint blockade in lung cancer. Cell Mol Immunol 2021;18:279–93, https://doi.org/10.1038/s41423-020-00577-5.Search in Google Scholar PubMed PubMed Central
22. Bianco, A, Malapelle, U, Rocco, D, Perrotta, F, Mazzarella, G. Targeting immune checkpoints in non small cell lung cancer. Curr Opin Pharmacol 2018;40:46–50, https://doi.org/10.1016/j.coph.2018.02.006.Search in Google Scholar PubMed
23. Huang, L, Xiong, W, Cheng, L, Li, H. Bioinformatics-based analysis of programmed cell death pathway and key prognostic genes in gastric cancer: implications for the development of therapeutics. J Gene Med 2023;2023:e3590, https://doi.org/10.1002/jgm.3590.Search in Google Scholar PubMed
24. Li, H, Zhang, X, Shang, J, Feng, X, Yu, L, Fan, J, et al.. Identification of NETs-related biomarkers and molecular clusters in systemic lupus erythematosus. Front Immunol 2023;14:1150828, https://doi.org/10.3389/fimmu.2023.1150828.Search in Google Scholar PubMed PubMed Central
25. Cheng, L, Xiong, W, Li, S, Wang, G, Zhou, J, Li, H. CRISPR-Cas9 screening identified lethal genes enriched in necroptosis pathway and of prognosis significance in osteosarcoma. J Gene Med 2023;2023:e3563, https://doi.org/10.1002/jgm.3563.Search in Google Scholar PubMed
26. Wang, Y, Wang, J, Li, X, Xiong, X, Wang, J, Zhou, Z, et al.. N(1)-methyladenosine methylation in tRNA drives liver tumourigenesis by regulating cholesterol metabolism. Nat Commun 2021;12:6314, https://doi.org/10.1038/s41467-021-26718-6.Search in Google Scholar PubMed PubMed Central
27. Zhang, L, Zhang, Z, Yu, Z. Identification of a novel glycolysis-related gene signature for predicting metastasis and survival in patients with lung adenocarcinoma. J Transl Med 2019;17:423, https://doi.org/10.1186/s12967-019-02173-2.Search in Google Scholar PubMed PubMed Central
28. Tatsuguchi, T, Uruno, T, Sugiura, Y, Oisaki, K, Takaya, D, Sakata, D, et al.. Pharmacological intervention of cholesterol sulfate-mediated T cell exclusion promotes antitumor immunity. Biochem Biophys Res Commun 2022;609:183–8, https://doi.org/10.1016/j.bbrc.2022.04.035.Search in Google Scholar PubMed
29. Tang, W, Zhou, J, Yang, W, Feng, Y, Wu, H, Mok, MTS, et al.. Aberrant cholesterol metabolic signaling impairs antitumor immunosurveillance through natural killer T cell dysfunction in obese liver. Cell Mol Immunol 2022;19:834–47, https://doi.org/10.1038/s41423-022-00872-3.Search in Google Scholar PubMed PubMed Central
30. Westerterp, M, Tall, AR. A new pathway of macrophage cholesterol efflux. Proc Natl Acad Sci U S A 2020;117:11853–5, https://doi.org/10.1073/pnas.2007836117.Search in Google Scholar PubMed PubMed Central
31. Qin, WH, Yang, ZS, Li, M, Chen, Y, Zhao, XF, Qin, YY, et al.. High serum levels of cholesterol increase antitumor functions of nature killer cells and reduce growth of liver tumors in Mice. Gastroenterology 2020;158:1713–27, https://doi.org/10.1053/j.gastro.2020.01.028.Search in Google Scholar PubMed
32. Shi, L, Pan, H, Liu, Z, Xie, J, Han, W. Roles of PFKFB3 in cancer. Signal Transduct Target Ther 2017;2:17044, https://doi.org/10.1038/sigtrans.2017.44.Search in Google Scholar PubMed PubMed Central
33. Thirusangu, P, Ray, U, Sarkar Bhattacharya, S, Oien, DB, Jin, L, Staub, J, et al.. PFKFB3 regulates cancer stemness through the hippo pathway in small cell lung carcinoma. Oncogene 2022;41:4003–17, https://doi.org/10.1038/s41388-022-02391-x.Search in Google Scholar PubMed PubMed Central
34. Yang, Q, Hou, P. Targeting PFKFB3 in the endothelium for cancer therapy. Trends Mol Med 2017;23:197–200, https://doi.org/10.1016/j.molmed.2017.01.008.Search in Google Scholar PubMed
35. Yang, F, Kou, J, Liu, Z, Li, W, Du, W. MYC enhances cholesterol biosynthesis and supports cell proliferation through SQLE. Front Cell Dev Biol 2021;9:655889, https://doi.org/10.3389/fcell.2021.655889.Search in Google Scholar PubMed PubMed Central
36. Li, J, Yang, T, Wang, Q, Li, Y, Wu, H, Zhang, M, et al.. Upregulation of SQLE contributes to poor survival in head and neck squamous cell carcinoma. Int J Biol Sci 2022;18:3576–91, https://doi.org/10.7150/ijbs.68216.Search in Google Scholar PubMed PubMed Central
37. Qin, Y, Zhang, Y, Tang, Q, Jin, L, Chen, Y. SQLE induces epithelial-to-mesenchymal transition by regulating of miR-133b in esophageal squamous cell carcinoma. Acta Biochim Biophys Sin 2017;49:138–48, https://doi.org/10.1093/abbs/gmw127.Search in Google Scholar PubMed
38. Yao, J, Li, R, Liu, X, Zhou, X, Li, J, Liu, T, et al.. Prognostic implication of glycolysis related gene signature in non-small cell lung cancer. J Cancer 2021;12:885–98, https://doi.org/10.7150/jca.50274.Search in Google Scholar PubMed PubMed Central
39. Choi, SH, Jin, CC, Do, SK, Lee, SY, Choi, JE, Kang, HG, et al.. Polymorphisms in glycolysis-related genes are associated with clinical outcomes of paclitaxel-cisplatin chemotherapy in non-small cell lung cancer. Oncology 2020;98:468–77, https://doi.org/10.1159/000504175.Search in Google Scholar PubMed
© 2023 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Review Article
- Mesenchymal stem cell exosomes: a promising delivery system for glioma therapy
- Research Articles
- A multi-cancer analysis unveils ITGBL1 as a cancer prognostic molecule and a novel immunotherapy target
- MicroRNA-302a enhances 5-fluorouracil sensitivity in HepG2 cells by increasing AKT/ULK1-dependent autophagy-mediated apoptosis
- Integrated analysis of TCGA data identifies endoplasmic reticulum stress-related lncRNA signature in stomach adenocarcinoma
- Glioblastoma with PRMT5 gene upregulation is a key target for tumor cell regression
- BRCA mutation in Vietnamese prostate cancer patients: a mixed cross-sectional study and case series
- Microglia increase CEMIP expression and promote brain metastasis in breast cancer through the JAK2/STAT3 signaling pathway
- Integrative machine learning algorithms for developing a consensus RNA modification-based signature for guiding clinical decision-making in bladder cancer
- Transcriptome analysis of tertiary lymphoid structures (TLSs)-related genes reveals prognostic value and immunotherapeutic potential in cancer
- Prognostic value of a glycolysis and cholesterol synthesis related gene signature in osteosarcoma: implications for immune microenvironment and personalized treatment strategies
- Identification of potential biomarkers in follicular thyroid carcinoma: bioinformatics and immunohistochemical analyses
- Rapid Communication
- Comparison of the Molecular International Prognostic Scoring System (IPSS-M) and Revised International Prognostic Scoring System (IPSS-R) in predicting the prognosis of patients with myelodysplastic neoplasms treated with decitabine
- Case Reports
- Intrapericardial nonfunctional paraganglioma: a case report and literature review
- Treatment of central nervous system relapse in PLZF::RARA-positive acute promyelocytic leukemia by venetoclax combined with arubicin, cytarabine and intrathecal therapy: a case report
Articles in the same Issue
- Frontmatter
- Review Article
- Mesenchymal stem cell exosomes: a promising delivery system for glioma therapy
- Research Articles
- A multi-cancer analysis unveils ITGBL1 as a cancer prognostic molecule and a novel immunotherapy target
- MicroRNA-302a enhances 5-fluorouracil sensitivity in HepG2 cells by increasing AKT/ULK1-dependent autophagy-mediated apoptosis
- Integrated analysis of TCGA data identifies endoplasmic reticulum stress-related lncRNA signature in stomach adenocarcinoma
- Glioblastoma with PRMT5 gene upregulation is a key target for tumor cell regression
- BRCA mutation in Vietnamese prostate cancer patients: a mixed cross-sectional study and case series
- Microglia increase CEMIP expression and promote brain metastasis in breast cancer through the JAK2/STAT3 signaling pathway
- Integrative machine learning algorithms for developing a consensus RNA modification-based signature for guiding clinical decision-making in bladder cancer
- Transcriptome analysis of tertiary lymphoid structures (TLSs)-related genes reveals prognostic value and immunotherapeutic potential in cancer
- Prognostic value of a glycolysis and cholesterol synthesis related gene signature in osteosarcoma: implications for immune microenvironment and personalized treatment strategies
- Identification of potential biomarkers in follicular thyroid carcinoma: bioinformatics and immunohistochemical analyses
- Rapid Communication
- Comparison of the Molecular International Prognostic Scoring System (IPSS-M) and Revised International Prognostic Scoring System (IPSS-R) in predicting the prognosis of patients with myelodysplastic neoplasms treated with decitabine
- Case Reports
- Intrapericardial nonfunctional paraganglioma: a case report and literature review
- Treatment of central nervous system relapse in PLZF::RARA-positive acute promyelocytic leukemia by venetoclax combined with arubicin, cytarabine and intrathecal therapy: a case report